1-s2.0-S0378517313003359-main

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 15

International Journal of Pharmaceutics 457 (2013) 446–460

Contents lists available at ScienceDirect

International Journal of Pharmaceutics


journal homepage: www.elsevier.com/locate/ijpharm

Review

Food, physiology and drug delivery


F.J.O. Varum 1 , G.B. Hatton, A.W. Basit ∗
Department of Pharmaceutics, UCL School of Pharmacy, University College London, WC1N 1AX, London, UK

a r t i c l e i n f o a b s t r a c t

Article history: Gastrointestinal physiology is dynamic and complex at the best of times, and a multitude of known vari-
Received 17 December 2012 ables can affect the overall bioavailability of drugs delivered via the oral route. Yet while the influences
Received in revised form 8 April 2013 of food and beverage intake as just two of these variables on oral drug delivery have been extensively
Accepted 12 April 2013
documented in the wider literature, specific information on their effects remains sporadic, and is not so
Available online 21 April 2013
much contextually reviewed. Food co-ingestion with oral dosage forms can mediate several changes to
drug bioavailability, yet the precise mechanisms underlying this have yet to be fully elucidated. Like-
Keywords:
wise, the often detrimental effects of alcohol (ethanol) on dosage form performance have been widely
Pharmacokinetics
Dose dumping
observed experimentally, but knowledge of which has only moderately impacted on clinical practice.
Controlled release Here, we attempt to piece together the available subject matter relating to the influences of both solid
Colonic drug delivery and liquid foodstuffs on the gastrointestinal milieu and the implications for oral drug delivery, with par-
Enteric coatings ticular emphasis on the behaviour of modified-release dosage forms, formulation robustness and drug
Biorelevant dynamic dissolution testing absorption. Providing better insight into these influences, and exemplifying cases where formulations
have been developed or modified to circumvent their associated problems, can help to appropriately
direct the design of future in vitro digestive modelling systems as well as oral dosage forms resilient to
these effects. Moreover, this will help to better our understanding of the impact of food and alcohol intake
on normal gut behaviour and function.
© 2013 Elsevier B.V. All rights reserved.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 447
2. The fate of food and beverages in the gastrointestinal tract . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 447
3. Effects of food on gastrointestinal physiology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 448
3.1. Gastrointestinal luminal changes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 448
3.2. Gastrointestinal motility and transit time . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 448
3.3. Effects of alcoholic beverages on gastrointestinal physiology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 449
4. Trends on the biopharmaceutical effects of food and alcohol on modified-release dosage forms . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 450
4.1. Delayed release dosage forms . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 450
4.1.1. Food effects . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 450
4.1.2. Alcohol effects . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 451
4.2. Sustained release dosage forms (film coated and matrix formulations) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 451
4.2.1. Food effects . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 451
4.2.2. Alcohol effects . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 453
5. Formulation approaches to circumvent food and alcohol effects . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 454
6. In vitro simulation of food and alcohol effects on drug release from MR formulations . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 456
6.1. Simulation of fed-state fluids . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 456
6.2. Dynamic simulation of the gastrointestinal tract . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 456
7. Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 457
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 457

∗ Corresponding author. Tel.: +44 20 7753 5865; fax: +44 20 7753 5865.
E-mail addresses: abdul.basit@pharmacy.ac.uk, a.basit@ucl.ac.uk (A.W. Basit).
1
Present address: Tillotts Pharma AG, Baslerstrasse 15, CH-4310 Rheinfelden, Switzerland.

0378-5173/$ – see front matter © 2013 Elsevier B.V. All rights reserved.
http://dx.doi.org/10.1016/j.ijpharm.2013.04.034
F.J.O. Varum et al. / International Journal of Pharmaceutics 457 (2013) 446–460 447

1. Introduction of cases of positive, negative and no food effects, respectively


(Gu et al., 2007).
Peroral administration largely remains the most convenient The very nature of both food and alcohol effects on oral drug
and popular means of providing both immediate- and modified- bioavailability is not, however, exclusive to the drug per se, and such
release drug delivery. More often than not, however, this method effects may also compromise the in vivo performance of modified-
of administration occurs under conditions of food intake, and release dosage forms. Modified-release dosage forms are predomi-
usually several times a day. To this end, knowledge of the effects of nantly designed to facilitate once- or twice- daily dosing regimens,
foods and fluids on the performance of oral formulations, includ- and to achieve sustained therapeutic plasmatic drug levels over a
ing modified-release systems, and on drug pharmacokinetics is prolonged period of time: In the latter case, they are referred to as
crucial to our continued understanding of factors influencing oral ’sustained release’ dosage forms, whereas ’delayed’ drug release
bioavailability, and is of particular relevance to the frequently systems by contrast deliver a drug to a specific site in the gut.
poly-medicated geriatric population. There also exists the broader potential for use of modified-release
Indeed, the concomitant administration of oral dosage forms multiparticulates to improve pharmacokinetic reproducibility for
with food has the potential to markedly affect oral drug bioavail- a particular drug candidate over their single-unit counterparts
ability, supplementary to parameters including lag time, rate and (Varum et al., 2010). Especially relevant to these possible effects
extent of absorption (Fleisher et al., 1999; Yasuji et al., 2012). It is the fed gut may have on modified-release dosage forms - namely,
also an issue which has gained increased attention in recent years, sustained release formulations - is the concept of ’dose dumping’,
posing several challenges to the design of drugs from developmen- which may result in higher drug bioavailability and possible toxic-
tal, clinical and regulatory perspectives: Often regarded as physical ity (Schug et al., 2002b; Yasuji et al., 2012). This is also an issue when
barriers inasmuch as physicochemical ones, the complexity of alcoholic beverages are consumed simultaneously with sustained
food effects on oral drug bioavailability results from the dynamic release formulations, as highlighted recently (Lennerns̈, 2009). The
interplay between the changing physiological environment of the most notable reported case of an alcohol-dosage form interaction in
fed gut, the physicochemical properties of the drug, drug dosing, pharmacokinetic studies, however, is that of the product Palladone
and the characteristics of the formulations. XL® a once-daily, sustained release dosage form of hydromorphone
Food-mediated effects on oral drug bioavailability are already (Walden et al., 2007). An alcohol concentration-dependent effect
well described and reviewed in the literature (Fleisher et al., 1999; was noted as the Cmax increased 1.1-, 1.9- and 5.5-fold when alcohol
Schmidt and Dalhoff, 2002; Singh and Malhotra, 2004; Yasuji et al., was ingested concomitantly with Palladone XL® at concentrations
2012), and it is not our intention to revisit these issues in great of 4, 20 and 40%, respectively. Strikingly, a 16-fold increase in the
detail. Briefly, a food-drug interaction can lead to five main different Cmax for one subject was observed following ingestion of 240 ml of
outcomes (Welling, 1996): 40% alcohol (Walden et al., 2007). These results ultimately lead to
the voluntary suspension of sales and marketing activities by the
manufacturer after a FDA request was made over concerns of safety
No effect on oral bioavailability;
and lack of robustness of the formulation in the presence of alco-
Positive food effect and consequent increase in bioavailability,
hol. The incidenti has since driven the FDA and other regulatory
potentially compromising safety;
agencies to propose new requirements for the testing of modified-
Negative food effect and consequent decrease in bioavailability,
release products which need to demonstrate robustness in media
potentially compromising efficacy;
containing up to 40% ethanol during in vitro testing.
Delayed absorption;
The aim of this review is to provide deeper insights into the
Accelerated absorption.
role of food and alcohol intake on the in vivo performance of
modified-release systems, and the implications of their interac-
Particularly relevant to these potential effects are the tions. A background of physiological changes occurring during food
physicochemical characteristics of drugs, for which purpose intake will be elucidated, and selected case studies will be pre-
both the Biopharmaceutical Classification System (BCS) and the sented to highlight this phenomenon.
Biopharmaceutical Drug Disposition Classification System (BDDCS)
- categorizing drugs according to their solubility, permeability and
metabolism - have contributed to the creation of a framework 2. The fate of food and beverages in the gastrointestinal
used in food effect prediction (Amidon et al., 1995; Wu and Benet, tract
2005; Custodio et al., 2008). Overall, no food effect (high-fat meal)
in terms of extent of absorption would be expected for BDDCS Solid food is first masticated in the mouth and passed by peri-
class I drugs (high solubility/high permeability/high metabolism), staltic contraction through the oesophagus to the stomach as a
though due to the delayed gastric emptying occurring after food bolus. The proximal stomach itself functions as a reservoir for undi-
intake, a delayed Cmax is likely to occur (Fleisher et al., 1999; gested food material and the emptying of liquids, with an expansion
Custodio et al., 2008). As for BDDCS class II drugs (low solubil- capacity of around 4 l in volume; the distal stomach by contrast is
ity/high permeability/high metabolism), higher drug exposure more critically involved in the processes of mechanical mixing and
can often be predicted due to the higher solubilizing capacity gastric emptying of solid contents into the duodenum, per se con-
of intestinal fluid following a meal. With BDDCS class III drugs trolled by contractions of the pylorus. Various acidic and enzymatic
(high solubility/low permeability/low metabolism), on the other gastric, hepatic and pancreatic secretions released during the ear-
hand, a negative food effect is expected due to the interaction of lier phases of digestion serve to homogenize food products into
food components with intestinal transporters, and largely in the their component parts, which are then readily absorbed along the
case of co-ingested high-fat meals: A general trend has not yet length of the small intestine. Any remaining waste is then propelled
been identified for BDDCS class IV drugs (Custodio et al., 2008). into the colon prior to excretion.
The accuracy of this framework to predict food effects has been The presence of insoluble food products such as fibre and guar
recently estimated as 70% (Benet, 2013), whereas an additional gum adding bulk to a peristaltic mass in the gut also contribute to
statistical model taking into consideration drug physicochemical the mechanical influences on oral drug delivery through distension
properties such as solubility, permeability and dose has also been of the stomach, duodenal and small intestinal spaces, and through
proposed to correctly predict food effects in 83%, 73% and 83% delaying gastric emptying (Schwartz et al., 1982; Sandhu et al.,
448 F.J.O. Varum et al. / International Journal of Pharmaceutics 457 (2013) 446–460

1987). Digestion time is dependent upon intra-individual variables the pH to rise between 4.0 and 5.0 thanks to buffering and dilution
such as age and gender, though as will be discussed in the remit of effects (Evans et al., 1988; Fallingborg et al., 1989). No significant
this review, food intake is a prominent influence on physiological changes in the small and large intestine pH have been reported after
factors such as gastric emptying. Given that the majority of food and meal consumption; however, this may be masked by inter- and
drug absorption takes place at the level of the stomach and small intra-individual variability, and slight changes occuring in differ-
intestine, however, most in vitro models used for the simulation of ent stages of meal digestion (Bratten and Jones, 2009). Postprandial
digestion often overlook the digestive phases of the colon. duodenal pH has been reported to decrease, for instance, on emp-
Contrary to previous assumptions, only 10-20% of ingested tying of acidic contents from the stomach (Kalantzi et al., 2006).
ethanol from alcoholic beverages is absorbed through the gastric Similarly, Diakidou and co-workers have shown that the pH (mea-
mucosa: The main site of alcohol absorption is instead that of sured ex vivo) of colonic fluids decreases in the fed state (from pH
the small intestine (Cori et al., 1930; Elmslie and Harvey, 1968; 7.8 to 6.0) due to the fermentation of carbohydrates by bacteria and
Persson, 1991). Furthermore, it has been demonstrated that fac- production of short-chain fatty acids. However, it should be noted
tors known to delay gastric emptying - such as dietary fat and that in both studies the pH was measured in aspirated fluids and
certain drugs - may slow the rate of alcohol absorption (McFarlane not with radiotelemetric capsules, which otherwise better reflect
et al., 1986; Levitt, 2002), and potentially lead to deleterious effects the real in vivo pH dynamics (Diakidou et al., 2009b). Gastrointesti-
on modified-release dosage form performance. Though alcohol is nal fluid also plays a key role in drug dissolution, and particularly
primarily metabolized in the liver by the action of alcohol dehy- for poorly soluble molecules. The colon has been shown to hold
drogenases and aldehyde dehydrogenase enzymes, a small fraction between 1 and 100ml of free fluid which may affect the behaviour
of this metabolism is also carried out by the gastric mucosa (Levitt oral modified-release formulations, though free intestinal water
et al., 1997): If we take into account this reduced alcohol gastric only represents a small proportion of the total water content, and
metabolism, and in combination with a limited gastric absorption, is not spread homogeneously throughout the gut lumen: Instead,
the volume and concentration of alcohol in the stomach can be con- this water is formed as pockets of different volumes, and so it is
siderable, and may compromise drug release from modified-release unlikely for oral dosage forms to be in continuous contact with fluid
dosage forms sensitive to ethanol. Additionally, the apparent solu- (Schiller et al., 2005). Moreover, the composition of the gastroin-
bility of lipophilic drugs can be transiently increased, contributing testinal fluids determines the dissolution rate of ionizable drugs
to increased bioavailability and possible toxicity (Fagerberg et al., and enteric coatings, and is affected by both the buffer capac-
2012). ity and buffer species (Mooney et al., 1981; Aunins et al., 1985;
Ozturk et al., 1988). Additional other fluid components such as
3. Effects of food on gastrointestinal physiology bile salts, lipids and lipolysis products, which luminally increase
in response to a meal, can also influence bioavailability by affecting
Food intake induces dynamic changes in the composition and drug dissolution and solubilization, and particularly for BCS class II
volumes of luminal fluids as well as in the patterns of gastrointesti- molecules (Charman et al., 1997; Dressman et al., 1998; Camilleri,
nal motility, ultimately affecting the transit of dosage forms and 2006; Clarysse et al., 2009a; Diakidou et al., 2009a).
the mechanical stress applied during passage along the gastroin-
testinal tract. The main changes induced by food and beverages 3.2. Gastrointestinal motility and transit time
consumption are represented in Fig. 1.
The mechanics of gastrointestinal transit are highly complex and
3.1. Gastrointestinal luminal changes influenced by a multitude of factors, ranging from inter- and intra-
individual variability in gastrointestinal physiology (McConnell
The pH in the stomach in the fasted state ranges from 0.8 and et al., 2008; Freire et al., 2011) to the type and size of a formu-
2.0 due to hydrogen ion secretion, though ingestion of food causes lation (single units or multiple units), as well as the presence or

Fig. 1. Summary of gastrointestinal physiology changes upon food intake. Further details are provided in the manuscript.
F.J.O. Varum et al. / International Journal of Pharmaceutics 457 (2013) 446–460 449

absence of food. These issues have been extensively reviewed in outcome of a later study, revealing that meals with 70% fat, 70%
recent years (Newton, 2010; Varum et al., 2010; Wilson, 2010; protein or 70% carbohydrate content had no influence on the stag-
Yuen, 2010). Gastric retention and gastric emptying in the fasted nation of dosage forms or their movement to distal regions (Price
state are under the control of the migrating myoelectric complex et al., 1993). Another crucial aspect to be considered is the passage
(MMC), whereas in the presence of food, the MMC is disrupted and of dosage forms from the ileum to the caecum, as the sphincter in
is replaced by fed state contractions. It should be noted that some the ileocaecal junction applies a mechanical stress that, depending
MMC cycles may bypass the stomach and originate in the small on the size and type of formulation, can result in premature disrup-
intestine, thereby contributing to the prolonged gastric retention tion of the modified-release mechanism (Wilson, 2010). The basal
of modified-release dosage forms, and mainly monolithics (Kellow pressure (measured by intraluminal manometry) at the ileocaecal
et al., 1986). Aside from mixing and milling the gastric contents, junction increases to a value of 9.7 mmHg in the fasted state, and
these contraction cycles also exert mechanical pressure on dosage up to 11.8 in the first 30 min in response to a meal (Dinning et al.,
forms, namely through the pyloric sphincter. Using a wireless 1999).
motility capsule (SmartPill(r) ), wave pressures above 100 mbar and
occasionally close to 300 mbar could be recorded during passage
residence in the distal antrum and passage through the pyloric 3.3. Effects of alcoholic beverages on gastrointestinal physiology
sphincter (Cassilly et al., 2008). However, during such experimental
proceedings, the size of the SmartPill needs to be taken into account It has been shown that pure ethanol and alcoholic beverages
(13 mm x 26 mm), as smaller monolithic or multiparticulates may can inhibit gastric emptying (Franke et al., 2005). Lennernas and
not encounter such high motility pressures. A maximum gastric co-workers have since postulated that the inhibitory effect of alco-
emptying rate of 200 Kcal/h has been reported (Hunt, 1983), and the hol on gastric emptying may be related to the caloric content of
complexity involved in the control of gastric emptying has already the beverage, though somewhat interestingly these effects have
been highlighted in the apt statement by Olsson and Holmgren been shown to be independent of the caloric content of a meal
that älmost everything seems to affect gastric emptying(Olsson ¨ consumed at the same time (Lennerns̈, 2009). It has been shown
and Holmgren, 2001). This is another important issue where the that both fat and alcohol are major dietary contributors to the
controlled and reproducible behaviour of modified-release dosage slowing of gastric emptying, along with reports that consump-
forms is required in practice: Standardized fed state clinical stud- tion of fermented drinks which include high relative carbohydrate
ies are designed on the assumption that there is no further food contents - such as beer, red and white wine - leads to prolongation
intake until 4 h after breakfast, though this situation is difficult to of gastric emptying as compared to other ethanol-containing bev-
replicate on a daily basis, and is further complicated by the changes erages (Franke et al., 2005). It is moreover likely that the inhibitory
in meal composition. For instance, it has been shown that 10 mm effect of alcohol on gastric emptying is reduced in the fully fed
enteric coated tablets taken during breakfast, lunch and dinner did state, characterised by distension of the stomach and dilution of
not empty from the stomach for up to 12 h after the first tablet was its contents.
taken (unpublished data), whilst Katsuma and co-workers reported Additionally, it has been shown that the ingestion of alcohol
that in one subject a 300 mg enteric coated tablet (CODES(tm) sys- may induce gastric acid secretion (Lenz et al., 1983), though such
tem) given in the fasted state took 21 h to empty from the stomach results are conflicting, and may be again specific to the bever-
(Katsuma et al., 2004). Other small snacks consumed during the age consumed (Singer et al., 1983; Singer et al., 1991; Manabe
day additionally contribute to delays in gastric emptying, and sim- et al., 2003). Indeed, Lennerns̈ and co-workers have suggested
ilarly unpublished data brought to attention in a review article by that it is more likely to be the non-alcoholic constituents of these
Newton revealed that the emptying of 1.0-1.4 mm pellets was fur- beverages stimulating gastric acid secretions (Lennerns̈, 2009).
ther delayed with subsequent meals (Newton, 2010). In this study, Corroborating this observation of non-alcoholic constituents influ-
two out of six volunteers were shown to have all pellets still resid- encing gastrin release are studies which have shown there to be a
ing in the stomach after 4 h (second meal given) and 8 h (third negligible difference between a saline control and pure ethanol on
meal given), delaying gastric emptying for at least 12 h (Newton, the stimulation of gastric acid secretion administered by intragas-
2010). Equally, these influences should be considered in the case of tric infusion (Singer et al., 1983; Peterson et al., 1986; Chari et al.,
gastro-retentive dosage forms: An efficient gastro-retentive system 1993), though problems related to the lack of standardized con-
should be able to remain in the stomach for a prolonged period of trols have been reported. Nevertheless, as with alcohol-mediated
time when compared to standard formulations, and independent of effects on gastric emptying, such phenomena may contribute to
food consumption. However, significant advantages in dosage form affectation of oral modified-release dosage form performance at the
performance with these systems over single units given with a meal level of the stomach, potentially leading to dose dumping following
have yet to be witnessed (Waterman, 2007). Small intestinal transit rapid dosage form disintegration along with drug dissolution and
time is frequently quoted at 3-4 h, and is reported to be similar for absorption.
tablets, pellets and liquid formulations (Davis et al., 1986), though The study of the effects of moderate and chronic alcohol con-
this figure hides the considerable inter- and intra-subject variabil- sumption on gastrointestinal tissue functionality is beyond the
ity for small intestinal transit times of multiple- and single-unit scope of this review, though it has been addressed in some detail
systems, with values typically ranging from between 1 and 9.5 h by various other researchers (Huppe et al., 1988; Bujanda, 2000;
(Davis et al., 1986; Sugito et al., 1990; Coupe et al., 1991). Most oral Stermer, 2002; Taylor et al., 2005). For instance, it has been shown
dosage forms also spend the majority of the total transit time at that the permeability of the intestinal mucosa increases when
rest in the small intestine (McConnell et al., 2008; Weitschies et al., exposed to alcohol (Lavö et al., 1992). Therefore, higher drug dis-
2010). Feeding increases the motility in the terminal ileum/caecum, solution in the stomach in combination with a higher permeability
triggering what is known as the gastro-colonic reflex, and therein can result in a higher Cmax , which can ultimately trigger patient
helping the luminal contents to move distally along the gastroin- safety problems. It has also been shown in vitro that alcohol has
testinal tract whilst simultaneously accommodating food contents a direct dose-dependent effect on the activity of gastric and pan-
emptying from the stomach (Price et al., 1993; Haggar et al., 2003; creatic lipases by decreasing their capacity to degrade the fat
Wilson, 2010). Interestingly, the stagnation of dosage forms in the components from food (Sternby et al., 1996), though this effect is
caecum was found to be affected by the type of ingested food as generally only observed in individuals in whom pancreatic function
well as the meal size (Khosla et al., 1989). This contrasts with the is already compromised.
450 F.J.O. Varum et al. / International Journal of Pharmaceutics 457 (2013) 446–460

(Coupe et al., 1991) and, surprisingly, even 7 mm tablets have been


shown to readily empty from the stomach (Khosla et al., 1989). It
should also be noted that differences observed in drug absorption
from enteric coated delayed-release formulations may be due to
variability in gastrointestinal pH, rather than variability in tran-
sit itself (Fallingborg et al., 1989): Duodenal pH alone has been
shown to range between 5.17 and 6.10 during meals, and between
4.55 and 6.31 during the day time, increasing in the distal small
intestine to about pH 7.5 in the ileum (Bratten and Jones, 2009).
Furthermore, Kenyon and co-workers have shown that a delay
in gastric emptying arising from food intake did not compromise
the enteric properties of coated starch capsules, though disinte-
gration in the small intestine occurred earlier than for capsules
which emptied from the stomach quicker (Kenyon et al., 1994).
This may have been due to the elevated pH of the fed stomach,
which can contribute to softening of the enteric coating (Evans et al.,
1988). Also, a bioequivalence study (repeated dose) involving two
enteric coated formulations of omeprazole has shown that dose
administration after a high-fat meal reduced the rate and extent
of omeprazole bioavailability (Vaz-da-Silva et al., 2005). This may
again be related to the higher pH in the stomach and attributable to
the combined effect of repeated proton pump inhibitor administra-
tion and the high-fat meal, leading to premature drug release, drug
instability and decreasing bioavailability. Kamba and co-workers
proposed, through indirect measurements, that the fed stomach
also promotes a higher mechanical stress on dosage forms due to
the continuous milling of the food contents (Kamba et al., 2000),
though mechanical susceptibility in the small intestine did not
alter with food intake (Kamba et al., 2002). It is worth noting that
the mechanical strength of many oral solid dosage forms does,
however, drastically decrease on exposure to gastrointestinal fluid,
being more susceptible to damage by motility contractions during
passage along the gut (Sako et al., 1996).
Besides the traditional administration of oral dosage forms in
the fasted and fed states in standard clinical trial settings, other
Fig. 2. Mean plasma concentration of salicylic acid after administration of acetyl- options exist which allow for the demonstration of food effects on
salicylic acid as enteric coated tablets (top) or enteric coated granules (bottom) in oral drug delivery. For instance, Digenis and co-workers showed
the fasted (open circles) and fed state (closed circles). that the administration of a multiple-unit formulation 30 min
Source: Reprinted from Bogentoft et al. (1978) with permission. before food (pre-feed regimen) resulted in faster gastric emptying
when compared to the fasted state (Digenis et al., 1990). This sug-
gests that food intake leads to an increase in gastric motility, and
4. Trends on the biopharmaceutical effects of food and consequently faster gastric emptying. Furthermore, small intestine
alcohol on modified-release dosage forms transit was accelerated, leading to a lower overall erythromycin
bioavailability - erythromycin being preferably absorbed in the
4.1. Delayed release dosage forms small intestine. Peristaltic activity and intestinal flow have also
been shown to increase after food intake, which may have con-
4.1.1. Food effects tributed to the results obtained (Matuchansky et al., 1972; Kerlin
The susceptibility of a modified-release formulation to a food- et al., 1982). These findings were recently corroborated as a sim-
mediated interaction is frequently reduced for multiparticulates ilar acceleration of small intestinal transit, - in this case, with
(Varum et al., 2010). For instance, a lower variability and higher tablets - was observed under the pre-feed regimen (Fig. 3). Here,
bioavailability was shown for erythromycin enteric coated pellets tablets spent a significantly shorter time in the small intestine
in comparison to enteric coated tablets after food intake (Graffner (100 min) than that observed for the standard fasted (204 min) and
et al., 1986). Similarly, the oral bioavailability of acetylsalicylic acid, fed regimens (210 min) (Fadda et al., 2009). Equally, it has also been
given as enteric coated granules, was shown to be higher, less vari- reported that a meal can induce a gastro-ileocaecal reflex, thereby
able and not affected by the presence of food (Fig. 2), in contrast initiating the transport of fluids and solids from the distal small
to enteric coated tablets for which a significant delay in the onset intestine into the caecum, and providing room for further transport
of absorption was noted (Bogentoft et al., 1978). Delayed gastric along the small intestine (Schiller et al., 2005).
emptying of the tablet formulation was also likely to have occurred Liquids inasmuch as foodstuffs can also affect oral drug
due to the presence of food and subsequent meals (lunch and snack) bioavailability. For instance, the bioavailability of lansoprazole
further hindering gastric emptying. Generally speaking, the greater enteric-coated granules following intake of different juices (orange
the size of the formulation, the more prolonged the period of gas- juice, tomato juice, or a small amount of strained pears) was sim-
tric emptying, and mainly in the fed state (Davis et al., 1984; Sugito ilar to that of the encapsulated material administered with water.
et al., 1990; Abrahamsson et al., 1996). However, this may also However, the onset of drug absorption was significantly delayed
be an oversimplification: Faster emptying of multiple-unit dosage with consumption of orange juice as compared to the other juices
forms from the fed stomach was not shown with 1 mm pellets, and a control, resulting in a later tmax (3 h vs. 1.7 h in control) (Chun
for instance, when compared to 11 mm single-unit dosage forms et al., 2002). This may be related to differences in gastric emptying
F.J.O. Varum et al. / International Journal of Pharmaceutics 457 (2013) 446–460 451

and co-workers reported a reduced water content for dissolution,


pH and surface tension in the fed state, though in contrast the buffer
capacity, osmolality, short-chain fatty acids, and bile salts (cholate
and chenodeoxycholate) were all higher in the fed state, and may
have contributed to the accelerated dissolution of modified-release
dosage forms arriving in the large intestine (Diakidou et al., 2009b).

4.1.2. Alcohol effects


The effects of alcohol on delayed-release dosage forms have
been scarcely explored. In these circumstances, premature drug
release due to an alcohol-mediated disruption of the delayed
release mechanism can significantly compromise oral drug
bioavailability. Many enteric coated formulations are produced
using polymers which are soluble in organic solvents, such as
Fig. 3. Relationship between gastric emptying time and small intestinal transit time ethanol, and therefore prolonged exposure to high alcohol con-
in a pre-feed regimen. Open symbols: small intestinal transit of tablets which were centration in the stomach can trigger premature dissolution of the
in the upper small intestine when a meal was given at 45 min after dosing. The enteric coating and compromise overall drug bioavailability. In the
tablets had a rapid and statistically significant acceleration in small intestinal tran-
case of enteric coated dosage forms, a higher pH in the fed state
sit compared to the tablets which were in the stomach when food arrived (closed
symbols). associated with intake of alcoholic beverages (even considering
Source: Reprinted from Fadda et al. (2009) with permission. possible dilution) can have deleterious effects on the enteric coat-
ing where gastric emptying is significantly delayed (Franke et al.,
2005). For instance, Fadda and co-workers evaluated the effect of
of the liquids, as orange juice is known to have a slower rate of emp- ethanol present in the dissolution medium on different mesalazine
tying than that of tomato juice, and is possibly attributable to the modified-release formulations, observing that drug release was
lower pH of orange juice itself (Haggard and Greenberg, 1941). Low generally accelerated for the different products but to differing
pH liquids are also known to have a slower rate of gastric clearance degrees depending on the formulation characteristics and the time
(Rasmussen et al., 1999; Chaw et al., 2001), which in turn hinders of exposure to alcohol (Fadda et al., 2008).
the rapid dissolution of enteric coatings.
It has been reported that the bioavailability of mesalazine 4.2. Sustained release dosage forms (film coated and matrix
from delayed release MMX® systems (Lialda/Mezavant) increases formulations)
(91% higher Cmax and 16% higher AUC) when administered
with a high-fat meal (Yang and McCormack, 2011). This indi- 4.2.1. Food effects
cates an earlier drug release taking place in the small intestine, As alluded to, the effects that food or alcohol may have on the
with a reduced amount of drug being available for the topi- oral drug bioavailability of sustained release products can be com-
cal action in the colon. Similarly, the dosing instructions for a promising to their safety, especially given that these systems often
new Eudragit S® coated mesalazine delayed release capsule prod- carry high drug loads to facilitate continuous drug release and allow
uct (http://www.delzicol.com/index.jsp) indicate that it should be for once-daily dosing. It is therefore a requirement by regulatory
taken 1 h prior to food or at least 2 h after a meal, hinting at a food authorities that generic versions of a modified-release product for
interaction. A gamma-scintigraphy study has also shown that in single dose administration must be tested in the fasted and fed
three out of eight volunteers Eudragit S® coated tablets failed to state to allow for direct comparison with the reference product.
disintegrate when administered in the fed state or 30 min before Often, the food effects on oral drug bioavailability from sustained
food, whereas in the fasted state only one subject was affected release dosage forms have been associated with the formulation
(Ibekwe et al., 2008b). However, factors other than pH and tran- type and the engineered mechanism of release, as in the case of
sit time may have contributed to the overall outcome: Different sustained release tablets showing much higher inter-subject vari-
food effects were, for instance, observed in the case of two budes- ability than sustained release multiparticulates in the fed state
onide colonic delivery formulations. Budesonide modified-release (Delhotal-Landes et al., 1988; Cnota et al., 2005). This phenomenon
pellets (Entocort®) produced a significantly higher Cmax and a later has been also linked to higher susceptibility to mechanical stress
tmax when administered after food intake (Edsbc̈ker et al., 2003), during transit along the gut, particularly through the bottle-necked
though a dissimilar effect on budesonide absorption was observed pylorus and ileocaecal junction (Garbacz et al., 2008). It would seem
when a high-fat and high caloric breakfast was taken before admin- logical that single units would be more sensitive to mechanical
istration of a new colonic drug delivery system (tablet) using the pressures along the gut, though it has been reported that sustained
MMX® technology: In this case, food intake resulted in a lower release multiparticulates systems, with a reduced hardness after
rate and extent of drug absorption. A food-drug interaction or soaking, can also suffer from premature drug release following
increased pre-systemic metabolism may, on the other hand, have mechanical damage induced by food (Katori et al., 1996).
contributed to the lower bioavailability in the fed state (Brunner In one study, food intake resulted in a shorter tmax and
et al., 2005). Other explanations can be related to activation of higher Cmax of verapamil when delivered as sustained release
the gastro-colonic reflex, which prompts a fast discharge of mate- ethylcellulose-coated pellets, with similar overall bioavailability
rial located at the ileo-caecal junction and further movement of to that achieved in the fasted state. Arguably, this was due to the
contents along the colon, therefore decreasing the time available increased residence time of pellets in the upper gastrointestinal
for drug release and dissolution in the colonic fluids. Hodges and co- tract and consequent exposure to the fluid available (Marvola et al.,
workers have shown that ingestion of food triggers the movement 1989). The fluid volume of the stomach in the fed state is, some-
of colonic release dosage forms distally along the colon in subjects what inevitably, significantly higher when compared to the fasted
where arrival in the ascending colon occurred prior to or simulta- state; however, this has been only reported as an increase in the
neously with food administration (Fig. 4), though a late arrival in stomach volume (Schiller et al., 2005). In the small intestine, the
the colon was also observed when the formulation was still residing free fluid available is reduced in the fed state and with no signif-
in the small intestine during feeding (Hodges et al., 2009). Diakidou icant differences in fluid volume in the large intestine, thought to
452 F.J.O. Varum et al. / International Journal of Pharmaceutics 457 (2013) 446–460

Fig. 4. Effects of dosage form localization when feeding takes palce on the moevement along the colon.
Source: Reprinted with permission from Hodges et al. (2009).

be predominantly due to the continuous absorption of water. The state to 3.1-22.4 mg/ml in the fat-enriched regimen, whereas the
changing environment of the stomach and proximal small intestine bile salts varied between 2.0 and 9.0 in the fasted and 4.4 and
in response to a meal - and particularly for fat-enriched meals - by 30.3 mM in the fat-enriched fed state (Clarysse et al., 2009b).
induction of pancreatic and biliary secretion and lipolysis has also Further to this, a pharmacoscintigraphic study demonstrated
been investigated by gastric (Diakidou et al., 2009a) and duodenal that a sustained release minitablets of diltiazem (Geomatrix) given
aspirates (Persson et al., 2006; Clarysse et al., 2009a; Clarysse et al., in the fasted state disintegrated completely in the large bowel,
2009b). The solubilizing capacity of the fed intestinal fluid of poorly providing sustained drug release and uniform absorption along
soluble drugs is extensively affected by the rate of bile acid secre- the entire gastrointestinal tract. In contrast, administration after
tion, the rate of lipolysis, and consequent generation of other lipidic a high-fat breakfast resulted in prolongation of gastric retention
entities such as phospholipids, fatty acids and mono- and diglicery- and tablet disintegration in the stomach in six out of eight volun-
des (Fig. 5) (Persson et al., 2005; Persson et al., 2006). Notably, a teers - another important example of how food may compromise
large inter-individual variability was observed in the composition drug formulation properties and result in dose dumping (Wilding
of the intestinal fluids after food intake: Lipid digestion products et al., 1995). In the fed state, the peristaltic activity in the distal
after 30 min of food intake ranged from 0.0-5.5 mg/ml in the fasted stomach promotes mixing and milling of solids, and controls the
F.J.O. Varum et al. / International Journal of Pharmaceutics 457 (2013) 446–460 453

Fig. 5. Concentrations (mM) of (a) bile acids and (b) phospholipids from intestinal secretions and NuTRIflex in intestinal fluids after the starting the perfusion of a nutri-
tional drink. TCA, taurocholic acid; GCA, glycocholic acid; TCDCA, taurochenodeoxycholic acid; TDCA, taurodeoxycholic acid; GCDCA, glycochenodeoxycholic acid; GDCA,
glycodeoxycholic acid; PC, phosphatidylcholine; LPC, lysophoshatidylcholine.
Source: Reprinted with permission from Persson et al. (2006).

transfer of chyme into the duodenum. It is this activity which plays different study where an osmotic pump formulation was compared
a role in gastric emptying, responding to the osmotic pressure and to a erodible enteric coated tablet formulation (Slofedipine XL), a
energy content of digestion products of food. Indeed, the greater significant delay (15 h in 15 out of 24 subjects) in drug absorp-
the caloric content of a meal, the lower the volume transferred to tion was seen for the latter formulation following consumption of a
the duodenum (Hunt and Stubbs, 1975; Hunt et al., 1978). It has high-fat breakfast (Schug et al., 2002a) (Fig. 8). A number of generic
also been shown that dietary sugars and salt, available in high con- nifedipine sustained release products in turn demonstrated sus-
tent in western diets (including soft drinks, sport drinks, fruit juices, ceptibility to mechanical effects (Garbacz et al., 2009) simulated
confectionary and many others), can accelerate drug release from using a novel dissolution apparatus, which may relate to the food
HPMC matrices, with the most popular being sustained release plat- effects and lack of robustness observed in several prior bioequiv-
forms (Williams et al., 2009; Williams et al., 2010). Mechanistically, alence studies (Schug et al., 2002a; Schug et al., 2002b). From a
the polymer hydration and gel layer formation is compromised in different angle, Weitschies and co-workers highlighted the impact
the presence of high concentrations of dietary sugars, leading to a of the intragastric location of these sustained release preparations
higher mechanical susceptibility in the fed stomach (Williams et al., and related food effects. They proposed that the so-called dose-
2009). In contrast, a delayed drug release from HPMC matrices was dumping effects observed for some sustained release nifedipine
seen in vitro in the presence of lipid emulsions - a key component and felodipine products may be due to poor mixing in the proximal
of high-fat diets and biorelevant media (Williams et al., 2011). In a part of the stomach, leading to a delayed absorption as well as high
different study, an 800 mg ibuprofen sustained release formulation plasmatic levels after gastric emptying (Weitschies et al., 2005).
took 15 h to empty from the stomach in 2 out of 11 subjects after a
heavy breakfast, with a mean emptying time of 8.8 h. In the fasted 4.2.2. Alcohol effects
state, a double peak in the pharmacokinetic profile was obtained Observations of alcohol-induced effects on the oral bioavailabil-
(Fig. 6), which was correlated with higher disintegration rate of ity of Palladone XL® and its consequential marketing withdrawal
the formulation and likely due to the passage of the formulation (Walden et al., 2007) highlighted the major risks associated with
through the gastric pylorus and the ileocaecal junction (Garbacz co-administration of certain medications and alcoholic beverages.
et al., 2008). However, a light breakfast intake abolished the double However, the influence of alcohol on the in vivo pharmacokinetic
peak profile, leading to a drug plateau between 4 and 12 h. profile of hydromorphone was much more modest when formu-
The requirements for fed-state bioequivalence studies for new lated as an osmotic pump system (OROS) (Sathyan et al., 2008).
sustained release generic formulations have emerged from these Studies conducted by Johnson and co-workers, on the other hand,
observations that food intake can impact on drug integrity and revealed that sustained release formulations of morphine sulphate
performance, despite bioequivalence often being observed in the were not affected by simultaneous alcohol ingestion in both
fasted state. For instance, administration of a generic nifedipine the fasted and fed state, thereby demonstrating the potential of
60 mg erodible tablet (Coral(r) ) after a high-fat American breakfast certain formulation designs to overcome such problems (Johnson
resulted in dose dumping and a consequent increase in nifedip- et al., 2008). Equally, Henderson and co-workers have shown
ine plasma levels of 3-4-fold, though an osmotic driven system that the pharmacokinetic parameters after administration of a
(Adalat(r) , Oros) showed only a slight increase in bioavailability sustained release capsule of carvedilol were not affected by alcohol
when compared to the fasted state, revealing a clear lack of bio- intake (240 ml of 20% alcohol beverage) despite the dissolution
equivalence between both formulations in the fed state (Schug in vitro being clearly accelerated, and overestimating an alcohol
et al., 2002b). Similarly, a 30 mg nifedipine generic formulation interaction (Henderson et al., 2007). These conflicting results
(Nifedipine Merck, matrix monolayer tablet) has shown differences were subsequently explained on the basis of fed state behaviour
in in vitro dissolution (pH-independent vs. pH-dependent assays in the in vivo study: Here, the higher volume of fluid in the fed
compared to the reference formulation (Adalat OROS, osmotic push stomach promotes a dilution effect, reducing a potential alcohol-
pull pump)). When administered with a high-fat meal, the generic induced effect on modified-release formulations. In addition to
formulation led to a higher extent of absorption and correspond- the various food-mediated effects on gastrointestinal physiology,
ing dose-dumping, as seen in Fig. 7 (Wonnemann et al., 2008). In a the ingestion of alcohol is also known to influence gastrointestinal
454 F.J.O. Varum et al. / International Journal of Pharmaceutics 457 (2013) 446–460

motor activity and fluid composition (Lenz et al., 1983; Franke


et al., 2005). Both Walden and co-workers and Lennernas and
co-workers have shown that the dissolution and drug release from
several sustained release dosage forms is markedly increased with
co-ingestion of alcohol (Walden et al., 2007; Lennerns̈, 2009), and
which can dramatically influence drug pharmacokinetics following
an increase in drug absorption rate from the gastrointestinal tract
if the drug solubility and/or permeability are positively affected
by alcohol intake. Furthermore, some formulation excipients
may be prone to exhibit higher solubility in aqueous solutions
with an increasing ethanol content, and so facilitate faster drug
release.
Several reports have also addressed the question of pos-
sible alcohol-induced changes influencing drug release from
modified-release dosage forms through in vitro dissolution stud-
ies. Leuner and co-workers, for instance, showed that marketed
sustained release dosage forms containing the opioids oxycodone
hydrochloride, hydromorphone hydrochloride and morphine sul-
phate differed in their susceptibility to alcohol in dissolution media
(Leuner et al., 2007). In this study, monolithic matrix systems
were not affected by alcohol, though multiparticulate formula-
tions coated with ethylcellulose exhibited a faster release profile
in the presence of low ethanol concentrations (5-10%), therein
highlighting the importance of dosage form design. Levina and
co-workers have shown that hydrophilic matrices (HPMC) incor-
porating felodipine, gliclazide or metformin hydrochloride were
insensitive in vitro on exposure to ethanol concentrations of
up to 40%, irrespective of HPMC viscosity grade (Levina et al.,
2007), though in a different study, aspirin hypromellose matri-
ces showed susceptibility to faster release in vitro, linked to
the slower hydration of the polymer in the presence of ethanol
(Roberts et al., 2007). Films of ethylcellulose blended with HPMC,
on the other hand, show greater potential for dosage form fail-
ure in the presence of ethanol (Larsson et al., 2010). In a more
extended study, 27 modified-release formulations (tablets or
beads/pellets in a capsule) were tested in dissolution media con-
taining different concentrations of ethanol. An accelerated release
was noted for 9 out of 10 capsule and 2 out of 17 tablet sus-
tained release preparations in the presence of 40% ethanol (Smith
et al., 2010). Therefore, during their early development it would
be advisable to test such dosage forms in dissolution media of
varying ethanol concentrations in order to assess robustness,
improve formulation design, and mitigate the likelihood of dose
dumping.

5. Formulation approaches to circumvent food and alcohol


effects

The formulation approach chosen to deliver a drug by the oral


route can significantly contribute to the enhancement or reduction
of food and alcohol effects on oral drug bioavailability, as discussed
in the case studies presented here. For instance, it has been shown
that targeting trientine to the middle or lower part of the small
intestine by means of an enteric coating abolished the negative
food effects observed when given as an oral solution in the fed state
(Tanno et al., 2008). This has been attributed to the delayed gastric
emptying of the enteric coated formulation and distal release of
the drug avoiding physicochemical drug-food interactions. Simi-
larly, the bioavailability of DX-9065 administered in a capsule was
Fig. 6. Interplay between ibuprofen plasma concentration and rate of matrix disin- shown to be limited in the fasted state and further reduced in the
tegration in the fasted state and after a light or heavy breakfast.
fed state. The formulation of an enteric coated tablet, however,
Source: Reprinted with permission from Wilson et al. (1989).
increased oral drug bioavailability 5-fold and reduced food effects
by limiting interaction with bile salts (Fujii et al., 2011), which expo-
nentially increase in the duodenal fluid in the fed state (Schmidt and
Dalhoff, 2002). Recently, an enteric coated risedronate product has
F.J.O. Varum et al. / International Journal of Pharmaceutics 457 (2013) 446–460 455

Fig. 7. Mean (SD) plasma concentration–time curves of nifedipine 30 mg after oral, single-dose administration of test* and reference† formulations in 12 healthy white male
volunteers following a high-fat breakfast.
Source: Reprinted from Wonnemann et al. (2008) with permission.

been launched which offers the possibility of being safely admin- fat-rich meal led to an increase in bioavailability, thus rendering the
istered with food (http://www.atelvia.com/index.jsp) - a marked differences between both IR and ER formulation profiles negligible.
improvement on the original uncoated product, which needed to One possible explanation may be the modulation of the metabolic
be taken on an empty stomach to prevent the from drug chelating activity of intestinal CYP3A4 and inhibition of efflux transporters
with food components. The tablet core contains drug and EDTA, by secreted bile salts and fatty components of the meal. Other com-
and the product is coated with a layer of enteric polymer, releasing ponents of food may also be causative of such effects - grapefruit
its contents in the small intestine. At this level, the EDTA acts as a juice, for instance, is a well-established inhibitor of intestinal CYP
scavenger for food components (calcium ions), and so the drug is enzymes as well as efflux transporters (Bailey et al., 2013). The list
free to be absorbed. This is an example of a coating overcoming a of drugs administered by the oral route which act as substrates
food effect - although the main mechanism involves the EDTA, the for the CYP3A4 enzymes in the gut lining is extensive (McConnell
coating is still crucial to ensuring that both the drug and the EDTA et al., 2009), and the interaction with grapefruit juice can also lead
are released simultaneously at the same location in the intestine. to serious adverse effects due to a dramatic increase in bioavailabil-
Propiverine undergoes extensive first pass metabolism by ity (Ducharme et al., 1993; Lown et al., 1997; Kantola et al., 1998;
intestinal CYP3A4 and efflux transporters. The lower metabolism Lilja et al., 2000; Bailey et al., 2013).
in the distal small intestine and colon contributes to a higher Novel delayed release formulations designed to target the ileo-
oral bioavailability of propiverine when vehiculated through an colonic region and exploit gastrointestinal bacteria to trigger drug
extended release formulation, as compared to the immediate release have been shown to be robust and effective independent of
release dosage form which predisposes to a higher metabolism in the feeding regimen. In a study conducted by Basit and co-workers,
the proximal gut in both the fasted and fed state (Siegmund et al., theophylline pellets coated with a mixture of ethylcellulose and
2012). Interestingly, no food effect was observed for the extended amylose exhibited similar bioavailability in the fasted and fed state
release formulation, as the drug itself could be absorbed more dis- in a cross-over design despite the longer gastric retention in the fed
tally. However, in the case of the IR formulation, ingestion of a state, and also on higher exposure to pancreatic enzymes through-
out transit along the small intestine (Basit et al., 2009). Similarly,
a pH- and bacteria-combined triggered coating approach applied
on 8 mm tablets has been shown to produce a robust ileo-colonic
release platform, with accurate targeting of the colonic region in
the fasted, fed and pre-feed state (Ibekwe et al., 2008a). This shows
that the dynamic changes in the gastrointestinal physiology driven
by food intake did not influence these types of delayed release tech-
nologies, instead relying at least partially on the action of bacterial
enzymes to initiate drug release.
Roth and co-workers have shown that an innovative vera-
pamil hot-melt extrusion sustained release formulation (Verapamil
Meltrex) was not impaired in the presence of 40% ethanol in dis-
solution medium in vitro (Roth et al., 2009). In contrast, when
other marketed sustained release verapamil products were tested
in medium with 20-40% ethanol, a dose-dumping effect occurred,
and with 100% release achieved in 2 h. Recently, a new coating
material, Aquacoat® ARC (FMC Biopolymer), has been launched
with the purported benefit of providing robust sustained release in
Fig. 8. Geometric mean plasma concentration vs. time curves (n = 24) of nifedip- the presence of ethanol. This product combines a polymer insoluble
ine determined after oral administration of Adalat OROS and Slofedipine XL under in ethanol (guar gum) and ethylcellulose as a polymer insoluble in
fasting conditions and after a high-fat breakfast ( Adalat OROS, fasted administra-
ethanol-free media, with the combined properties of both poly-
tion; *Adalat OROS, fed administration; 䊉Slofedipine1 XL, fasted administration; 
Slofedipine1 XL, fed administration). mers contributing to overcome alcohol-induced dose-dumping
Source: Reprinted with permission from Shug et al. (2002). effects.
456 F.J.O. Varum et al. / International Journal of Pharmaceutics 457 (2013) 446–460

6. In vitro simulation of food and alcohol effects on drug lower bioavailability in the fed state, as the window of absorption
release from MR formulations is limited to the upper small intestine (Radwan et al., 2012).
Equally, the luminal composition of the stomach and small
The clinical significance of food effects on oral drug bioavailabil- intestine following intake of alcohol beverages is not well under-
ity is difficult to predict from traditional in vitro drug dissolution stood, and while it has been proposed that in vitro dissolution
studies. In vitro simulation of the environment in the fed gut (par- testing of modified-release systems in the presence of 40% ethanol
ticularly in the stomach and small intestine) is limiting, as the for 2 h would provide a surrogate for alcohol-induced dose-duping
composition of a meal in everyday daily life is so widely variable effects (Lennerns̈, 2009), gastric residence time may otherwise be
that a standard formula will never be able to accurately reflect the prolonged in the presence of alcohol. In this way, the exposure of
in vivo situation. Equally, successful prediction of a food-induced modified-release products would also be increased (Fraser et al.,
change in oral drug bioavailability is strongly dependent on the 1995; Higaki et al., 2008).
characteristics of the drug and the design of the modified-release
system (Lentz, 2008; Yasuji et al., 2012). 6.2. Dynamic simulation of the gastrointestinal tract

The in vitro simulation of gastrointestinal compartments has


6.1. Simulation of fed-state fluids been historically limited to the use of USP dissolution apparatus
(I-IV). However, attempts to recreate events taking place in the
As previously described in this review, the changing gut envi- gut by these approaches have failed insofar as fluid composition
ronment after meal consumption can affect the behaviour of and hydrodynamic or mechanical changes in the gut lumen are
modified-release dosage forms and the solubility of the carried concerned (McCallister, 2010). The situation becomes even more
drug. Therefore, close mimicry of the fed gut fluid is of critical complicated if a prediction of food effects is warranted. For this rea-
importance (Lentz, 2008; Klein, 2010). To this end, the use of milk or son, the Dynamic Gastric Model (IFR, Norwich) was developed to
milk-based products (Ensure Plus) has been proposed to simulate simulate the gastric digestion process, with gastric secretions and
the fed stomach fluid composition in vitro (Macheras et al., 1986; patterns of mixing and motility intended to closely resemble the
Ashby et al., 1989; Galia et al., 1998; Klein et al., 2004; Diakidou in vivo situation (Wickham et al., 2009). The disintegration of cap-
et al., 2009a). Ensure Plus was shown to have similar physico- sules under fasted conditions in this equipment generally matched
chemical properties to a homogenized FDA standard breakfast used the in vivo observations. In the fed state, however, a delayed cap-
in bioavailability and bioequivalence studies (Klein et al., 2004), sule disintegration was observed (Vardakou et al., 2011) that was
whereas milk has been shown to improve solubility of lipophilic significantly longer than for previous in vivo studies using gamma
drugs (Macheras et al., 1989), but also delays the disintegration of scintigraphy (Cole et al., 2004; Tuleu et al., 2007; Jones et al., 2012).
formulations (Abrahamsson et al., 2004; Anwar et al., 2005). Dress- This was attributed to the lower shear in the fundus as compared to
man and co-workers introduced a Fed State Simulated Intestinal the antral regions, where most of the mixing and mechanical stress
fluid (FeSSIF) which includes bile salts and lecithin in the buffer on the wet contents occurs. The increasingly broad application of
system to reflect bile secretions into the duodenum during a diges- such technology across the pharmaceutical field therefore offers
tion process (Dressman et al., 1998). However, these simulated the possibility of assessing food effects on modified-release formu-
fluids per se are only able to provide a static resemblance of the lations, and particularly for those more susceptible to mechanical
fed stomach and small intestine, and do not simulate the active stress during gastric residence, enabling better prediction of poten-
food digestion process characterised by physiological luminal com- tial dose-dumping effects.
position changes due to the formation of lipid end-products (fatty Similarly, the gastrointestinal model TIM-1 is a dynamic mul-
acid, monoglycerides) as well as variations in the rate of gastric and ticompartimental system developed by TNO Nutrition and Food
intestinal secretions. Recently, “snap-shot” dissolution media were Research that simulates both stomach and small intestinal phys-
proposed to simulate the dynamic changes in the fluid composi- iological processes, incorporating secretions, enzymatic reactions,
tion during digestion (Jantratid et al., 2008), and which correspond mixing and motility, pH changes, and the continuous removal of
to changes in the composition and physicochemical proper- digested compounds and small molecules through dialysis mem-
ties of gastrointestinal fluid in different phases of the digestive branes (Blanquet et al., 2004). The TIM-2 in turn is complementary
process. The main changes observed were in pH, osmolality and the model which simulates the fermentation process and peristaltic
milk/buffer ratio in simulated gastric fluid, and in the case of the mixing of the large intestine (Minekus et al., 1999). These sys-
simulated intestinal fluid, bile salt, lecithin, monoglycerides, fatty tems provide a more accurate representation of the exposure that
acid and pancreatin (facultative) concentrations were manipulated modified-release dosage forms experience during transit through
to resemble the lipid digestion process in the duodenum (Porter the gastrointestinal tract, and also allow for the assessment of
et al., 2007). Despite providing a better representation of the diges- drug metabolism by colonic bacteria (Sousa et al., 2008). However,
tion process, however, variability associated with food composition the data available in the public domain is still scarce. Souliman
in routine daily life and the timing of food ingestion and dosage and co-workers have demonstrated a better in vitro/in vivo cor-
form administration cannot be effectively replicated. Another fac- relation (IVIVC) in the fasted state when theophylline sustained
tor often neglected is the viscosity of the luminal contents in the release tablets (hydrophilic matrix) were tested in TIM-1 as com-
fed stomach, proposed to be in the range 200-2000 mPa s after pared to the USP II and IV apparatuses (Souliman et al., 2007).
food ingestion (Marciani et al., 2001). Despite an initial increase Sustained release paracetamol tablets additionally showed a pro-
in the apparent viscosity of the gastric contents in response to vis- longed absorption in the TIM-1 model, closely replicating in vivo
cous foodstuffs, it is subsequently reduced by dilution effects and data in the fasted state (Blanquet et al., 2004). In the fed state, a good
motility. The viscosity of a homogenized standard meal (as recom- mimicry of the in vivo performance of immediate release paraceta-
mended by the FDA) was replicated in vitro using HPMC as viscosity mol tablets was also established, replicating the lower absorption
enhancing agent in standard buffers, and a good in vitro-in vivo cor- previously observed in the presence of food (Souliman et al., 2006).
relation was found for the negative food effect observed with the Intake of a high-fat nutritional drink concomitantly with an imme-
BCS class III drug trospium chloride. The prolonged in vitro disin- diate release tablet of fosemprenavir led to a delay in disintegration
tegration time of commercial trospium chloride tablets and lower and a consequent shift of 2 h in the tmax (Brouwers et al., 2007).
dissolution rates as a result of reduced fluid ingress may explain its The gastric and duodenal concentrations of fosemprenavir in the
F.J.O. Varum et al. / International Journal of Pharmaceutics 457 (2013) 446–460 457

fed state could thus be closely predicted using the TIM-1 system, led to the development of a novel dissolution apparatus which
in contrast to static dissolution which was shown to overestimate combines new features such as irregular mechanical pressure,
the food effect (Brouwers et al., 2011). The impact of food effects shear force and periods without exposure to fluid during tran-
and gut physiology factors was also assessed in a delayed release sit through the gut (Garbacz et al., 2008). Using this system and
5-ASA product (Lialda, Shire) designed to target the colon. Through predetermined applied pressures, it was shown that changing the
coupling the TIM-1 and TIM-2 systems, it was demonstrated that applied pressure resulted in a different drug release profile from
in both the fasted and fed state (high-fat breakfast), less than 1% a diclofenac extended-release tablet formulation (Voltaren 100 mg
of drug was release after 6 h in TIM-1. After transition to the TIM- Retard). In vivo in the fasted state, multiple peaks were noted in
2, complete drug release was observed over 20 h (Tenjarla et al., the pharmacokinetic profile for individual subjects which was cor-
2007). However, in this study the gastric residence time was set related with sensitivity to mechanical stress during transit through
to 2 h, in both fasted and fed states, which may not resemble the the gastrointestinal tract, and namely during gastric emptying and
likelihood of prolonged gastric residence in the fed state, and in passage through the ileocaecal junction (Fig. 9). However, a pel-
particular for bigger dosage forms such as 1200 mg mesalamine let extended-release formulation did not exhibit multiple peaks,
tablets. Indeed, it has been shown that gastric emptying can take suggesting that multiple units are less affected by the mechanical
up to 12 h in the fed state, and can compromise the performance of pressure applied from the gut wall (Garbacz et al., 2008). Generic
modified-release formulations (Newton, 2010). Overall, The TIM- versions of a diclofenac sustained release preparation also showed
1 and TIM-2 systems provide a more dynamic and close mimicry variable susceptibility to mechanical stress, potentially leading to
of the luminal environment and motility when compared to other variable oral drug bioavailability (Garbacz and Weitschies, 2010).
currently available gut modelling systems, though their associated Abrahamsson and co-workers have otherwise proposed a new
complexity and slow rate of data generation limit widespread use. approach to predict the shear stress applied on extended release
The magnitude of mechanical stress applied to modified-release matrix formulations by the fed stomach which combines computer
formulations during transit through the gut depends on the gas- simulations and in vitro dissolution able to provide a controlled
trointestinal location and prandial status (Garbacz and Klein, 2012). hydrodynamic and shear stress on a tablet surface (Abrahamsson
Based on data collected from in vivo measurements of gastrointesti- et al., 2005).
nal pressures, residence times, fluid volumes and fluid distribution

7. Conclusions

We know gastrointestinal functionality to be both highly com-


plex and variable, and with particular implications for peroral drug
delivery and bioavailability. However, it is insufficient to merely
consider the in vivo pharmacokinetic and pharmacodynamic effects
on oral dosage forms without taking into account the consequences
of food and alcohol co-ingestion. Food intake and the specific com-
ponents of consumable solid and liquids, such as fats, can influence
gastric emptying and transit times, and in doing so affect the
rate and extent of oral drug absorption. In the case of modified-
release dosage forms, this can lead to premature drug release or
’dumping’ with failure of the dosage form: ethanol, for instance,
has been repeatedly shown to affect parameters such as fluid pH
and composition, and can influence the disintegration and dis-
solution of various solid oral modified-release formulations both
in vitro and in vivo. Some dosage forms or drugs may also be more
sensitive or susceptible to the effects of ethanol than others, as
evidenced by the aforementioned market withdrawal of an opioid
formulation. The precise mechanisms through which both food and
alcohol interfere with the performance of oral dosage forms still
remain not completely understood. Some gaps in our knowledge
are limiting the effective design of in silico and in vitro meth-
ods able to precisely predict food and alcohol-induced changes.
Therefore, better in vivo data collected from well designed and
controlled studies need to be gathered for a more insightful under-
standing of the interplay between food-dosage form-drug and its
prediction, ultimately contributing to the appropriate design and
development of robust dosage forms resistant to food and alcohol
effects.

References

Abrahamsson, B., Alpsten, M., Jonsson, U.E., Lundberg, P.J., Sandberg, A., Sundgren,
M., Svenheden, A., Tolli, J., 1996. Gastro-intestinal transit of a multiple-unit
formulation (metoprolol CR/ZOK) and a non-disintegrating tablet with the
emphasis on colon. Int. J. Pharm. 140, 229–235.
Fig. 9. Individual diclofenac plasma concentration profiles obtained after the
Abrahamsson, B., Albery, T., Eriksson, A., Gustafsson, I., Sjöberg, M., 2004. Food
administration of ER formulations in the fasted state (n = 24). In the insets the means effects on tablet disintegration. Eur. J. Pharm. Sci. 22, 165–172.
and standard deviations are shown. (A) Voltaren retard ER tablets; (B) Diclofenac- Abrahamsson, B., Pal, A., Sjöberg, M., Carlsson, M., Laurell, E., Brasseur, J., 2005.
ratiopharm ER pellets. A novel in vitro and numerical analysis of shear-induced drug release from
Source: Reprinted with permission from Garbacz et al. (2008). extended-release tablets in the fed stomach. Pharm. Res. 22, 1215–1226.
458 F.J.O. Varum et al. / International Journal of Pharmaceutics 457 (2013) 446–460

Amidon, G., Lennernäs, H., Shah, V., Crison, J., 1995. A theoretical basis for a biophar- form of sustained release tablet and microgranules. Biopharm. Drug Dispos. 9,
maceutic drug classification: the correlation of in vitro drug product dissolution 19–29.
and in vivo bioavailability. Pharm. Res. 12, 413–420. Diakidou, A., Vertzoni, M., Dressman, J.B., Reppas, C., 2009a. Estimation of intragas-
Anwar, S., Fell, J.T., Dickinson, P.A., 2005. An investigation of the disintegration of tric drug solubility in the fed state: comparison of various media with data in
tablets in biorelevant media. Int. J. Pharm. 290, 121–127. aspirates. Biopharm. Drug Dispos. 30, 318–325.
Ashby, L.J., Beezer, A.E., Buckton, G., 1989. In vitro dissolution testing of oral con- Diakidou, A., Vertzoni, M., Goumas, K., Söderlind, E., Abrahamsson, B., Dressman, J.B.,
trolled release preparations in the presence of artificial foodstuffs. I. Exploration Reppas, C., 2009b. Characterization of the contents of ascending colon to which
of alternative methodology: microcalorimetry. Int. J. Pharm. 51, 245–251. drugs are exposed after oral administration to healthy adults. Pharm. Res. 26,
Aunins, J.G., Southard, M.Z., Myers, R.A., Himmelstein, K.J., 1985. Dissolution 2141–2151.
of carboxylic acids. III: The effect of polyionizable buffers. J. Pharm. Sci. 74, Digenis, G.A., Sandefer, E.P., Parr, A.F., Beihn, R., McClain, C., Scheinthal, B.M., Ghebre-
1305–1316. Sellassie, I., Nesbitt, R.U., Randinitis, E., 1990. Gastrointestinal behaviour of
Bailey, D.G., Dresser, G., Arnold, J.M.O., 2013. Grapefruit-medication interactions: orally administered radiolabelled erythromycin pellets in man as determined
Forbidden fruit or avoidable consequences? Can. Med. Assoc. J. 185, 309–316. by gamma scintigraphy. J. Clin. Pharmacol. 30, 621–631.
Basit, A.W., Short, M.D., Mcconnell, E.L., 2009. Microbiota-triggered colonic delivery: Dinning, P.G., Bampton, P.A., Kennedy, M.L., Kajimoto, T., Lubowski, D.Z., de Carle,
robustness of the polysaccharide approach in the fed state in man. J. Drug Target. D.J., Cook, I.J., 1999. Basal pressure patterns and reflexive motor responses in
17, 64–71. the human ileocolonic junction. Am. J. Physiol. Gastrointest. Liver Physiol. 276,
Benet, L.Z., 2013. The role of BCS (Biopharmaceutics Classification System) and G331–G340.
BDDCS (Biopharmaceutics Drug Disposition Classification System) in drug Dressman, J.B., Amidon, G.L., Reppas, C., Shah, V.P., 1998. Dissolution testing as
development. J. Pharm. Sci. 102, 34–42. a prognostic tool for oral drug absorption: immediate release dosage forms.
Blanquet, S., Zeijdner, E., Beyssac, E., Meunier, J.-P., Denis, S., Havenaar, R., Alric, M., Pharm. Res. 15, 11–22.
2004. A dynamic artificial gastrointestinal system for studying the behaviour of Ducharme, M.P., Provenzano, R., Dehoorne-Smith, M., Edwards, D.J., 1993. Trough
orally administered drug dosage forms under various physiological conditions. concentrations of cyclosporine in blood following administration with grape-
Pharm. Res. 12, 585–591. fruit juice. Br. J. Clin. Pharmacol. 36, 457–459.
Bogentoft, C., Carlsson, I., Ekenved, G., Magnusson, A., 1978. Influence of food on the Edsbäcker, S., Bengtsson, B., Larsson, P., Lundin, P., Nilsson, Å., Ulmius, J.,
absorption of acetylsalicylic acid from enteric-coated dosage forms. Eur. J. Clin. Wollmer, P., 2003. A pharmacoscintigraphic evaluation of oral budesonide
Pharmacol. 14, 351–355. given as controlled-release (Entocort) capsules. Aliment. Pharmacol. Ther. 17,
Bratten, J., Jones, M.P., 2009. Prolonged recording of duodenal acid exposure in 525–536.
patients with functional dyspepsia and controls using a radiotelemetry pH mon- Elmslie, R.G., Harvey, N., 1968. Alcohol absorption rate from different parts of gut.
itoring system. J. Clin. Gastroenterol. 43, 527–533. Q. J. Stud. Alcohol. 29, 308–312.
Brouwers, J., Tack, J., Augustijns, P., 2007. Parallel monitoring of plasma and intra- Evans, D.F., Pye, G., Bramley, R., Clark, A.G., Dyson, T.J., Hardcastle, J.D., 1988. Mea-
luminal drug concentrations in man after oral administration of fosamprenavir surement of gastrointestinal pH profiles in normal ambulant human subjects.
in the fasted and fed state. Pharm. Res. 24, 1862–1869. Gut 29, 1035–1041.
Brouwers, J., Anneveld, B., Goudappel, G.-J., Duchateau, G., Annaert, P., Augustijns, Fadda, H.M., Mohamed, M.A.M., Basit, A.W., 2008. Impairment of the in vitro drug
P., Zeijdner, E., 2011. Food-dependent disintegration of immediate release fos- release behaviour of oral modified release preparations in the presence of alco-
amprenavir tablets: in vitro evaluation using magnetic resonance imaging and hol. Int. J. Pharm. 360, 171–176.
a dynamic gastrointestinal system. Eur. J. Pharm. Biopharm. 77, 313–319. Fadda, H.M., McConnell, E.L., Short, M.D., Basit, A.W., 2009. Meal-induced accel-
Brunner, M., Ziegler, S., Di Stefano, A.F.D., Dehghanyar, P., Kletter, K., Tschurlovits, eration of tablet transit through the human small intestine. Pharm. Res. 26,
M., Villa, R., Bozzella, R., Celasco, G., Moro, L., Rusca, A., Dudczak, R., Müller, M., 356–360.
2005. Gastrointestinal transit, release and plasma pharmacokinetics of a new Fagerberg, J.H., Al-Tikriti, Y., Ragnarsson, G., Bergstrom, C.A.S., 2012. Ethanol effects
oral budesonide formulation. Br. J. Clin. Pharmacol. 61, 31–38. on apparent solubility of poorly soluble drugs in simulated intestinal fluid. Mol.
Bujanda, L., 2000. The effects of alcohol consumption upon the gastrointestinal tract. Pharm. 9, 1942–1952.
Am. J. Gastroenterol. 95, 3374–3382. Fallingborg, J., Christensen, L.A., Ingeman-Nielsen, M., Jacobsen, B.A., Abildgaard, K.,
Camilleri, M., 2006. Integrated upper gastrointestinal response to food intake. Gas- Rasmussen, H.H., 1989. pH profile and regional transit times of the normal gut
troenterology 131, 640–658. measured by radiotelemetry device. Aliment. Pharmacol. Ther. 3, 605–613.
Cassilly, D., Kantor, S., Knight, L.C., Maurer, A.H., Fisher, R.S., Semler, J., Parkman, H.P., Fleisher, D., Li, C., Zhou, Y., Pao, L., Karim, A., 1999. Drug, meal and formula-
2008. Gastric emptying of a non-digestible solid: assessment with simultaneous tion interactions influencing drug absorption after oral administration: clinical
SmartPill pH and pressure capsule, antroduodenal manometry, gastric emptying implications. Clin. Pharmacokinet. 36, 233–254.
scintigraphy. Neurogastroenterol. Motil. 20, 311–319. Franke, A., Nakchbandi, I.A., Schneider, A., Harder, H., Singer, M.V., 2005. The effect of
Chari, S., Teyssen, S., Singer, M.V., 1993. Alcohol and gastric acid secretion in humans. ethanol and alcoholic beverages on gastric emptying of solid meals in humans.
Gut 34, 843–847. Alcohol Alcoholism. 40, 187–193.
Charman, W.N., Porter, C.J.H., Mithani, S., Dressman, J.B., 1997. Physicochemical and Fraser, A.G., Rosalki, S.B., Gamble, G.D., Pounder, R.E., 1995. Interindividual and
physiological mechanisms for the effects of food on drug absorption: the role of intraindividual variability of ethanol concentration-time profiles – comparison
lipids and pH. J. Pharm. Sci. 86, 269–282. of ethanol ingestion before or after an evening meal. Br. J. Clin. Pharmacol. 40,
Chaw, C.S., Yazaki, E., Evans, D.F., 2001. The effect of pH change on the gastric emp- 387–392.
tying of liquids measured by electrical impedance tomography and pH-sensitive Freire, A.C., Basit, A.W., Choudhary, R., Piong, C.W., Merchant, H.A., 2011. Does
radiotelemetry capsule. Int. J. Pharm. 227, 167–175. sex matter? The influence of gender on gastrointestinal physiology and drug
Chun, A.H., Erdman, K., Chiu, Y.L., Pilmer, B.L., Achari, R., Cavanaugh, J.H., 2002. delivery. Int. J. Pharm. 415, 15–28.
Bioavailability of lansoprazole granules administered in juice or soft food com- Fujii, Y., Kanamaru, T., Kikuchi, H., Yamashita, S., Sakuma, S., 2011. Enteric-coated
pared with the intact capsule formulation. Clin. Ther. 24, 1322–1331. tablets improve oral bioavailability of DX-9065, a novel anticoagulant. Eur. J.
Clarysse, S., Psachoulias, D., Brouwers, J., Tack, J., Annaert, P., Duchateau, G., Reppas, Pharm. Sci. 42, 392–399.
C., Augustijns, P., 2009a. Postprandial changes in solubilizing capacity of human Galia, E., Nicolaides, E., Hörter, D., Löbenberg, R., Reppas, C., Dressman, J.B., 1998.
intestinal fluids for BCS class II drugs. Pharm. Res. 26, 1456–1466. Evaluation of various dissolution media for predicting in vivo performance of
Clarysse, S., Tack, J., Lammert, F., Duchateau, G., Reppas, C., Augustijns, P., 2009b. Class I and II drugs. Pharm. Res. 15, 698–705.
Pharmacokinetics, pharmacodynamics and drug metabolism: Postprandial evo- Garbacz, G., Klein, S., 2012. Dissolution testing of oral modified-release dosage
lution in composition and characteristics of human duodenal fluids in different forms. J. Pharm. Pharmacol. 64, 944–968.
nutritional states. J. Pharm. Sci. 98, 1177–1192. Garbacz, G., Weitschies, W., 2010. Investigation of dissolution behavior of diclofenac
Cnota, P.J., Nowak, H., Tagarro, I., Erb, K., Schürer, M., Schulz, H.-U., Maus, J., 2005. sodium extended release formulations under standard and biorelevant test con-
Tramadol SR formulations: pharmacokinetic comparison of a multiple-units ditions. Drug Dev. Ind. Pharm. 36, 518–530.
dose (capsule) versus a single-unit dose (tablet). Clin. Drug Invest. 25, 435–443. Garbacz, G., Wedemeyer, R.S., Nagel, S., Giessmann, T., Monnikes, H., Wilson, C.G.,
Cole, E.T., Scott, R.A., Cade, D., Connor, A.L., Wilding, I.R., 2004. In vitro and in vivo Siegmund, W., Weitschies, W., 2008. Irregular absorption profiles observed
pharmacoscintigraphic evaluation of ibuprofen hypromellose and gelatin cap- from diclofenac extended release tablets can be predicted using a dissolution
sules. Pharm. Res. 21, 793–798. test apparatus that mimics in vivo physical stress. Eur. J. Pharm. Biopharm. 70,
Cori, C.F., Villiaume, E.L., Cori, G.T., 1930. Studies on intestinal absorption. 2. The 421–428.
absorption of ethyl alcohol. J. Biol. Chem. 87, 19–26. Garbacz, G., Golke, B., Wedemeyer, R.-S., Axell, M., Söderlind, E., Abrahamsson,
Coupe, A.J., Davis, S.S., Wilding, I.R., 1991. Variation in gastrointestinal transit of B., Weitschies, W., 2009. Comparison of dissolution profiles obtained from
pharmaceutical dosage forms in healthy subjects. Pharm. Res. 8, 360–364. nifedipine extended release once a day products using different dissolution test
Custodio, J.M., Wu, C.-Y., Benet, L.Z., 2008. Predicting drug disposition, absorp- apparatuses. Eur. J. Pharm. Sci. 38, 147–155.
tion/elimination/transporter interplay and the role of food on drug absorption. Graffner, C., Josefsson, K., Stockman, O., 1986. Intra- and intersubject variation
Adv. Drug Deliv. Rev. 60, 717–733. of erythromycin absorption from single-unit and multiple-unit enteric-coated
Davis, S.S., Hardy, J.G., Taylor, M.J., Whalley, D.R., Wilson, C.G., 1984. The effect of products. Biopharm. Drug Dispos. 7, 163–171.
food on the gastrointestinal transit of pellets and an osmotic device (Osmet). Gu, C.-H., Li, H., Levons, J., Lentz, K., Gandhi, R., Raghavan, K., Smith, R., 2007. Pre-
Int. J. Pharm. 21, 331–340. dicting effect of food on extent of drug absorption based on physicochemical
Davis, S.S., Hardy, J.G., Fara, J.W., 1986. Transit of pharmaceutical dosage forms properties. Pharm. Res. 24, 1118–1130.
through the small intestine. Gut 27, 886–892. Haggar, R., Kumar, D., Benson, M., Grundy, A., 2003. Colonic motor activity in slow-
Delhotal-Landes, B., Flouvat, B., Boutin, M.-S., Karpouzas, I., Prinseau, J., 1988. transit idiopathic constipation as identified by 24-h pancolonic ambulatory
Influence of food on the absorption of theophylline administered in the manumetry. Neurogastroenterol. Motil. 15, 515–522.
F.J.O. Varum et al. / International Journal of Pharmaceutics 457 (2013) 446–460 459

Haggard, H.W., Greenberg, L.A., 1941. The influence of certain fruit juices on gastric Levina, M., Vuong, H., Rajabi-Siahboomi, A.R., 2007. The influence of hydro-alcoholic
function. Am. J. Dig. Dis. 8, 163–170. media on hypromellose matrix systems. Drug Dev. Ind. Pharm. 33, 1125–1134.
Henderson, L.S., Tenero, D.M., Campanile, A.M., Baidoo, C.A., Danoff, T.M., 2007. Levitt, D.G., 2002. PKQuest: measurement of intestinal absorption and first
Ethanol does not alter the pharmacokinetic profile of the controlled-release pass metabolism–application to human ethanol pharmacokinetics. BMC Clin.
formulation of carvedilol. J. Clin. Pharmacol. 47, 1358–1365. Pharmacol. 2, 1–12.
Higaki, K., Choe, S.Y., Löbenberg, R., Welage, L.S., Amidon, G.L., 2008. Mechanis- Levitt, M.D., Li, R., Demaster, E.G., Elson, M., Furne, J., Levitt, D.G., 1997. Use of
tic understanding of time-dependent oral absorption based on gastric motor measurements of ethanol absorption from stomach and intestine to assess
activity in humans. Eur. J. Pharm. Biopharm. 70, 313–325. human ethanol metabolism. Am. J. Physiol. Gastrointest. Liver Physiol. 273,
Hodges, L.A., Connollya, S.M., Banda, J., O’Mahony, B., Ugurlub, T., Turkoglub, M., G951–G957.
Wilson, C.G., Stevens, H.N.E., 2009. Scintigraphic evaluation of colon targeting Lilja, J.J., Kivisto, K.T., Neuvonen, P.J., 2000. Duration of effect of grapefruit juice
pectin–HPMC tablets in healthy volunteers. Int. J. Pharm. 370, 144–150. on the pharmacokinetics of the CYP3A4 substrate simvastatin. Clin. Pharmacol.
Hunt, J.N., 1983. Mechanisms and disorders of gastric emptying. Ann. Rev. Med. 34, Ther. 68, 384–390.
219–229. Lown, K.S., Bailey, D.G., Fontana, R.J., Janardan, S.K., Adair, C.H., Fortlage, L.A., Brown,
Hunt, J.N., Stubbs, D.F., 1975. The volume and energy content of meals as determi- M.B., Guo, W., Watkins, P.B., 1997. Grapefruit juice increases felodipine oral
nants of gastric emptying. J. Physiol. 245, 209–225. availability in humans by decreasing intestinal CYP3A protein expression. J. Clin.
Hunt, J.N., Cash, R., Newland, P., 1978. Energy density of food, gastric emptying, and Invest. 99, 2545–2553.
obesity. Am. J. Clin. Nutr. 31, S259–S260. Macheras, P., Koupparis, M., Tsaprounis, C., 1986. Drug dissolution studies in milk
Huppe, D., Tonissen, R., Kuntz, H.D., May, B., 1988. Chronic alcohol consumption using the automated flow injection serial dynamic dialysis technique. Int. J.
and gastrointestinal motility. Zeitschrift Fur Gastroenterologie, 26. Pharm. 33, 125–136.
Ibekwe, V.C., Khela, M.K., Evans, D.F., Basit, A.W., 2008a. A new concept in colonic Macheras, P.E., Koupparis, M.A., Antimisiaris, S.G., 1989. Effect of temperature and
drug targeting: a combined pH-responsive and bacterially-triggered drug deliv- fat content on the solubility of hydrochlorothiazide and chlorothiazide in milk.
ery technology. Aliment. Pharmacol. Ther. 28, 911–916. J. Pharm. Sci. 78, 933–936.
Ibekwe, V.C., McConnell, E.L., Fadda, H.M., Khela, M.K., Evans, D.F., Basit, A.W., 2008b. Manabe, T., Sawai, H., Okada, Y., Funahashi, H., Yamamoto, M., Sato, M., Hayakawa,
Interplay between intestinal pH, transit time and feed status on the in vivo T., Yamaki, K., 2003. Effects of whisky on plasma gastrin and cholecystokinin in
performance of pH responsive ileo-colonic release systems. Pharm. Res. 25, young adult men. J. Int. Med. Res. 31, 210–214.
1828–1835. Marciani, L., Gowland, P.A., Spiller, R.C., Manoj, P., Moore, R.J., Young, P., Fillery-
Jantratid, E., Janssen, N., Reppas, C., Dressman, J.B., 2008. Dissolution media simulat- Travis, A.J., 2001. Effect of meal viscosity and nutrients on satiety, intragastric
ing conditions in the proximal human gastrointestinal tract: an update. Pharm. dilution, and emptying assessed by MRI. Am. J. Physiol. Gastrointest. Liver
Res. 25, 1663–1676. Physiol. 280, G1227–G1233.
Johnson, F., Wagner, G., Sun, S., Stauffer, J., 2008. Effect of concomitant ingestion of Marvola, M., Kannikoski, A., Aito, H., Nykänen, S., 1989. The effect of food on gas-
alcohol on the in vivo pharmacokinetics of Kadian (Morphine sulfate extended- trointestinal transit and drug absorption of a multiparticular sustained-release
release) capsules. J. Pain. 9, 330–336. verapamil formulation. Int. J. Pharm. 53, 145–155.
Jones, B.E., Basit, A.W., Tuleu, C., 2012. The disintegration behaviour of capsules in Matuchansky, C., Huet, P.M., Mary, J.Y., Rambaud, J.C., Bernier, J.-J., 1972. Effects
fed subjects: a comparison of hypromellose (carrageenan) capsules and standard of cholecystokinin and metoclopramide on jejunal movements of water and
gelatin capsules. Int. J. Pharm. 424, 40–43. electrolytes on transit time of luminal fluid in man. Eur. J. Clin. Invest. 2, 169.
Kalantzi, L., Goumas, K., Kalioras, V., Abrahamsson, B., Dressman, J.B., Reppas, C., McCallister, M., 2010. Dynamic dissolution: a step closer to predictive dissolution
2006. Characterization of the human upper gastrointestinal contents under testing? Mol. Pharm. 7, 1374–1387.
conditions simulating bioavailability/bioequivalence studies. Pharm. Res. 23, McConnell, E.L., Fadda, H.M., Basit, A.W., 2008. Gut instincts: Explorations in intesti-
165–176. nal physiology and drug delivery. Int. J. Pharm. 364, 213–226.
Kamba, M., Seta, Y., Kusai, A., Ikeda, M., Nishimura, K., 2000. A unique dosage form McConnell, E.L., Liu, F., Basit, A.W., 2009. Colonic treatments and targets: issues and
to evaluate the mechanical destructive force in the gastrointestinal tract. Int. J. opportunities. J. Drug Target. 17, 335–363.
Pharm. 208, 61–70. McFarlane, A., Pooley, L., Welch, I.M., Rumsey, R.D., Read, N.W., 1986. How does
Kamba, M., Seta, Y., Kusai, A., Nishimura, K., 2002. Comparison of the mechanical dietary lipid lower blood alcohol concentrations? Gut 27, 15–18.
destructive force in the small intestine of dog and human. Int. J. Pharm. 237, Minekus, M., Smeets-Peeters, M., Bernalier, A., Marol-Bonnin, S., Havenaar, R.,
139–149. Marteau, P., Alric, M., Fonty, G., Huis in’t Veld, J.H.J., 1999. A computer-controlled
Kantola, T., Kivisto, K.T., Neuvonen, P.J., 1998. Grapefruit juice greatly increases system to simulate conditions of the large intestine with peristaltic mixing,
serum concentrations of lovastatin and lovastatin acid. Clin. Pharmacol. Ther. water absorption and absorption of fermentation products. Appl. Microbiol.
63, 397–402. Biotechnol. 53, 108–114.
Katori, N., Ma M.-c. Aoyagi, N., Kojima, S., 1996. Effect of destruction force on drug Mooney, K.G., Mintun, M.A., Himmelstein, K.J., Stella, V.J., 1981. Dissolution kinetics
release from multiple unit controlled release dosage forms in humans. Pharm. of carboxylic acids II: effect of buffers. J. Pharm. Sci. 70, 22–32.
Res. 13, 1541–1546. Newton, J.M., 2010. Gastric emptying of multi-particulate dosage forms. Int. J.
Katsuma, M., Watanabe, S., Takemura, S., Sako, K., Sawada, T., Masuda, Y., Nakamura, Pharm. 395, 2–8.
K., Fukui, M., Connor, A.L., Wilding, I.R., 2004. Scintigraphic evaluation of a novel Olsson, C., Holmgren, S., 2001. The control of gut motility. Comp. Biochem. Physiol.
colon-targeted delivery system (CODES) in healthy volunteers. J. Pharm. Sci. 93, A Mol. Integr. Physiol. 128, 481–503.
1287–1299. Ozturk, S.S., Palsson, B.O., Dressman, J.B., 1988. Dissolution of ionizable drugs in
Kellow, J.E., Borody, T.J., Phillips, S.F., Tucker, R.L., Haddad, A.C., 1986. Human buffered and unbuffered solutions. Pharm. Res. 5, 272–282.
interdigestive motility: variations in patterns from esophagus to colon. Gas- Persson, J., 1991. Alcohol and the small intestine. Scand. J. Gastroenterol. 26, 3–15.
troenterology 91, 386–395. Persson, E., Gustafsson, A.-S., Carlsson, A., Nilsson, R., Knutson, L., Forsell, P., Hanisch,
Kenyon, C.J., Cole, E.T., Wilding, I.R., 1994. The effect of food on the in vivo behaviour G., Lennernäs, H., Abrahamsson, B., 2005. The effects of food on the dissolution
of enteric coated starch capsules. Int. J. Pharm. 112, 207–213. of poorly soluble drugs in human and in model small intestinal fluids. Pharm.
Kerlin, P., Zinsmeister, A.R., Phillips, S., 1982. Relationship of motility to flow of Res. 22, 2141–2151.
contents in the human small intestine. Gastroenterology 82, 701–706. Persson, E.M., Nilsson, R.G., Hansson, G.I., Lofgren, L.J., Liback, F., Knutson, L., Abra-
Khosla, R., Feely, L.C., Davis, S.S., 1989. Gastrointestinal transit of non-disintegrating hamsson, B., Lennernas, H., 2006. A clinical single-pass perfusion investigation
tablets in fed subjects. Int. J. Pharm. 53, 107–117. of the dynamic in vivo secretory response to a dietary meal in human proximal
Klein, S., 2010. The use of biorelevant dissolution media to forecast the in vivo small intestine. Pharm. Res. 23, 742–751.
performance of a drug. AAPS J. 12, 397–406. Peterson, W.L., Barnett, C., Walsh, J.H., 1986. Effect of intragastric infusions of ethanol
Klein, S., Butler, J., Hempenstall, J.M., Reppas, C., Dressman, J.B., 2004. Media to sim- and wine on serum gastrin concentration and gastric acid secretion. Gastroen-
ulate the postprandial stomach I. Matching the physicochemical characteristics terology 91, 1390–1395.
of standard breakfasts. J. Pharm. Pharmacol. 56, 605–610. Porter, C.J.H., Trevaskis, N.L., Charman, W.N., 2007. Lipids and lipid-based formula-
Larsson, M., Hjärtstam, J., Berndtsson, J., Stading, M., Larsson, A., 2010. Effect of tions: optimizing the oral delivery of lipophilic drugs. Nat. Rev. Drug Discov. 6,
ethanol on the water permeability of controlled release films composed of ethyl 231–248.
cellulose and hydroxypropyl cellulose. Eur. J. Pharm. Biopharm. 76, 428–432. Price, J.M.C., Davis, S.S., Sparrow, R.A., Wilding, I.R., 1993. The effect of meal compo-
Lavö, B., Colombel, J.F., Knutsson, L., Hällgren, R., 1992. Acute exposure of small sition on the gastrocolonic response: implications for drug delivery to the colon.
intestine to ethanol induces mucosal leakage and prostaglandin E2 synthesis. Pharm. Res. 10, 722–726.
Gastroenterology 102, 468–473. Radwan, A., Amidon, G.L., Langguth, P., 2012. Mechanistic investigation of food effect
Lennernäs, H., 2009. Ethanol-Drug absorption interaction: potential for a significant on disintegration and dissolution of BCS class III compound solid formulations:
effect on the plasma pharmacokinetics of ethanol vulnerable formulations. Mol. the importance of viscosity. Biopharm. Drug Dispos. 33, 403–416.
Pharm. 6, 1429–1440. Rasmussen, L., Øster-JØrgensen, E., Qvist, N., Pedersen, S.A., 1999. The effects of
Lentz, K.A., 2008. Current methods for predicting human food effect. AAPS J. 10, omeprazole on intragastric pH, intestinal motility, and gastric emptying rate.
282–288. Scand. J. Gastroenterol. 34, 671–675.
Lenz, H.J., Ferrari-Taylor, J., Isenberg, J.I., 1983. Wine and five percent ethanol are Roberts, M., Cespi, M., Ford, J.L., Dyas, A.M., Downing, J., Martini, L.G., Crowley, P.J.,
potent stimulants of gastric acid secretion in humans. Gastroenterology 85, 2007. Influence of ethanol on aspirin release from hypromellose matrices. Int.
1082–1087. J. Pharm. 332, 31–37.
Leuner, C., Prater, D., Scherer, S., Topham, T., Walden, M., 2007. Predictability of Roth, W., Setnik, B., Zietsch, M., Burst, A., Breitenbach, J., Sellers, E., Brennan, D., 2009.
the potential for ethanol related dose dumping in oral prolonged release opioid Ethanol effects on drug release from Verapamil Meltrex® , an innovative melt
formulations. AAPS J., 9. extruded formulation. Int. J. Pharm. 368, 72–75.
460 F.J.O. Varum et al. / International Journal of Pharmaceutics 457 (2013) 446–460

Sako, K., Mizumoto, T., Kajiyama, A., Ohmura, T., 1996. Influence of physical factors in Tuleu, C., Khela, M.K., Evans, D.F., Jones, B.E., Nagata, S., Basit, A.W., 2007. A
gastrointestinal tract on acetaminophen release from controlled-release tablets scintigraphic investigation of the disintegration behaviour of capsules in fas-
in fasted dogs. Int. J. Pharm. 137, 225–232. ting subjects: A comparison of hypromellose capsules containing carrageenan
Sandhu, K.S., Elsamahi, M.M., Mena, I., Dooley, C.P., Valenzuela, J.E., 1987. Effect as a gelling agent and standard gelatin capsules. Eur. J. Pharm. Sci. 30,
of pectin on gastric emptying and gastroduodenal motility in normal subjects. 251–255.
Gastroenterology 92, 486–492. Vardakou, M., Mercuri, A., Naylor, T.A., Rizzo, D., Butler, J.M., Connolly, P.C., Wickham,
Sathyan, G., Sivakumar, K., Thipphawong, J., 2008. Pharmacokinetic profile of a 24- M.S.J., Faulks, R.M., 2011. Predicting the human in vivo performance of different
hour controlled-release OROS formulation of hydromorphone in the presence oral capsule shell types using a novel in vitro dynamic gastric model. Int. J. Pharm.
of alcohol. Curr. Med. Res. Opin. 24, 297–305. 419, 192–199.
Schiller, C., Frohlich, C.P., Giessmann, T., Siegmund, W., Mönnikes, H., Hosten, N., Varum, F.J.O., Merchant, H.A., Basit, A.W., 2010. Oral modified-release formulations
Weitschies, W., 2005. Intestinal fluid volumes and transit of dosage forms in motion: the relationship between gastrointestinal transit and drug absorp-
as assessed by magnetic resonance imaging. Aliment. Pharmacol. Ther. 22, tion. Int. J. Pharm. 395, 26–36.
971–979. Vaz-da-Silva, M., Loureiro, A., Nunes, T., Maia, J., Tavares, S., Falcão, A., Silveira, P.,
Schmidt, L.E., Dalhoff, K., 2002. Food–drug interactions. Drugs 62, 1481–1502. Almeida, L., Soares-da-Silva, P., 2005. Bioavailability and bioequivalence of two
Schug, B.S., Brendel, E., Chantraine, E., Wolf, D., Martin, W., Schall, R., Blume, H.H., enteric-coated formulations of omeprazole in fasting and fed conditions. Clin.
2002a. The effect of food on the pharmacokinetics of nifedipine in two slow Drug Invest. 25, 391–399.
release formulations: pronounced lag-time after a high fat breakfast. Br. J. Clin. Walden, M., Nicholls, F.A., Smith, K.J., Tucker, G.T., 2007. The effect of ethanol on
Pharmacol. 53, 582–588. the release of opioids from oral prolonged-release preparations. Drug Dev. Ind.
Schug, B.S., Brendel, E., Wolf, D., Wonnemann, M., Wargenau, M., Blume, H.H., 2002b. Pharm. 33, 1101–1111.
Formulation-dependent food effects demonstrated for nifedipine modified- Waterman, K.C., 2007. A critical review of gastric retentive controlled drug delivery.
release preparations marketed in the European Union. Eur. J. Pharm. Sci. 15, Pharm. Dev. Technol. 121, 1–10.
279–285. Weitschies, W., Wedemeyer, R.-S., Kosch, O., Fach, K., Nagel, S., Söderlind, E.,
Schwartz, S.E., Levine, R.A., Singh, A., Scheidecker, J.R., Track, N.S., 1982. Sustained Trahms, L., Abrahamsson, B., Mönnikes, H., 2005. Impact of the intragastric
pectin ingestion delays gastric emptying. Gastroenterology 83, 812–817. location of extended release tablets on food interactions. J. Control. Release 108,
Siegmund, W., Siegert, J., Richter, K., Schnabel, F., Feustel, C., Kirch, W., 2012. 375–385.
Influence of a fat-rich meal on bioavailability of extended-release and Weitschies, W., Blume, H., Mönnikes, H., 2010. Magnetic marker monitoring: high
immediate-release propiverine. J. Clin. Pharmacol. 52, 681–690. resolution real time tracking of oral solid dosage forms in the gastrointestinal
Singer, M.V., Eysselein, V., Goebell, H., 1983. Beer and wine but not whisky and pure tract. Eur. J. Pharm. Biopharm. 74, 93–101.
ethanol do stimulate release of gastrin in humans. Digestion 26, 73–79. Welling, P.G., 1996. Effects of food on drug absorption. Ann. Rev. Nutr. 16,
Singer, M.V., Teyssen, S., Eysselein, V.E., 1991. Action of beer and its ingredients on 383–415.
gastric acid secretion and release of gastrin in humans. Gastroenterology 101, Wickham, M., Faulks, R., Mills, C., 2009. In vitro digestion methods for assessing
935–942. the effect of food structure on allergen breakdown. Mol. Nutr. Food Res. 53,
Singh, B.N., Malhotra, B.K., 2004. Effects of food on the clinical pharmacokine- 952–958.
tics of anticancer agents: Underlying mechanisms and implications for oral Wilding, I.R., Davis, S.S., Sparrow, R.A., Ziemniak, J.A., Heald, D.L., 1995. Phar-
chemotherapy. Clin. Pharmacokinet. 43, 1127–1156. macoscintigraphic evaluation of a modified release (Geomatrix® ) diltiazem
Smith, A.P., Moore, T.W., Westenberger, B.J., Doub, W.H., 2010. In vitro dissolution formulation. J Control. Release 33, 89–97.
of oral modified-release tablets and capsules in ethanolic media. Int. J. Pharm. Williams, H.D., Ward, R., Hardy, I.J., Melia, C.D., 2009. The extended release proper-
398, 93–96. ties of HPMC matrices in the presence of dietary sugars. J. Control. Release 138,
Souliman, S., Blanquet, S., Beyssac, E., Cardot, J.-M., 2006. A level A in vitro/in vivo 251–259.
correlation in fasted and fed states using different methods: applied to solid Williams, H.D., Ward, R., Hardy, I.J., Melia, C.D., 2010. The effect of sucrose and salts
immediate release oral dosage form. Eur. J. Pharm. Sci. 27, 72–79. in combination on the drug release behaviour of an HPMC matrix. Eur. J. Pharm.
Souliman, S., Beyssac, E., Cardot, J.-M., Denis, S., Alric, M., 2007. Investigation of Biopharm. 76, 433–436.
the biopharmaceutical behaviour of theophylline hydrophilic matrix tablets Williams, H.D., Nott, K.P., Barrett, D.A., Ward, R., Hardy, I.J., Melia, C.D., 2011. Drug
using USP methods and an artificial digestive system. Drug Dev. Ind. Pharm. release from HPMC matrices in milk and fat-rich emulsions. J. Pharm. Sci. 100,
33, 475–483. 4823–4835.
Sousa, T., Paterson, R., Vanessa, M., Carlsson, A., Abrahamsson, B., Basit, A.W., 2008. Wilson, C.J., 2010. The transit of dosage forms through the colon. Int. J. Pharm. 395,
The gastrointestinal microbiota as a site for the biotransformation of drugs. Int. 17–25.
J. Pharm. 363, 1–25. Wilson, C.G., Washington, N., Greaves, J.L., Kamali, F., Rees, J.A., Sempik, A.K., Lam-
Stermer, E., 2002. Alcohol consumption and the gastrointestinal tract. Isr Med. Assoc. pard, J.F., 1989. Bimodal release of ibuprofen in a sustained-release formulation:
J. 4, 200–202. scintigraphic and pharmacokinetic open study in healthy volunteers under dif-
Sternby, B., Barros, H., Nilsson, Å., 1996. In vitro effects of ethanol on human gastric ferent conditions of food intake. Int. J. Pharm. 50, 155–161.
and pancreatic lipolytic activities/enzymes. Scand. J. Gastroenterol. 31, 146–153. Wonnemann, M., Schug, B., Anschütz, M., Brendel, E., Nucci, G.D., Blume, H., 2008.
Sugito, K., Hiroyasu, O., Goto, H., Noguchi, M., Kogure, T., Takano, M., Maruyama, Y., Comparison of two marketed nifedipine modified-release formulations: an
Sasaki, Y., 1990. Gastrointestinal transit of non-disintegrating solid formulations exploratory clinical food interaction study. Clin. Ther. 30, 48–58.
in humans. Int. J. Pharm. 60, 89–97. Wu, C.-Y., Benet, L.Z., 2005. Predicting drug disposition via application of BCS: trans-
Tanno, F.K., Sakuma, S., Masaoka, Y., Kataoka, M., Kozaki, T., Kamaguchi, R., Ikeda, port/absorption/elimination interplay and development of a biopharmaceutics
Y., Kokubo, H., Yamashita, S., 2008. Site-specific drug delivery to the middle- drug disposition classification system. Pharm. Res. 22, 11–23.
to-lower region of the small intestine reduces food–drug interactions that are Yang, L.P.H., McCormack, P.L., 2011. MMX Mesalazine: a review of its use in the
responsible for low drug absorption in the fed state. J. Pharm. Sci. 97, 5341–5353. management of mild to moderate ulcerative colitis. Drugs 71, 221–235.
Taylor, B., Rehm, J., Gmel, G., 2005. Moderate alcohol consumption and the gastroin- Yasuji, T., Kondo, H., Sako, K., 2012. The effect of food on the oral bioavailability of
testinal tract. Dig. Dis. 23, 170–176. drugs: a review of current developments and pharmaceutical technologies for
Tenjarla, S., Romasanta, V., Zeijdner, E., Villa, R., Moro, L., 2007. Release of 5- pharmacokinetic control. Ther. Dev. 3, 81–90.
aminosalicylate from an MMX mesalamine tablet during transit through a Yuen, K.-H., 2010. The transit of dosage forms through the small intestine. Int. J.
simulated gastrointestinal tract system. Adv. Ther. 24, 826–840. Pharm. 395, 9–16.

You might also like