You are on page 1of 31

Review

Effects of medicines used to treat gastrointestinal diseases


on the pharmacokinetics of coadministered drugs: a PEARRL
Review
Chara Litoua,† , Angela Effingerb,†, Edmund S. Kostewicza, Karl J. Boxc, Nikoletta Fotakib and
Jennifer B. Dressmana
a
Institute of Pharmaceutical Technology, Goethe University, Frankfurt am Main, Germany, bDepartment of Pharmacy and Pharmacology, Faculty of
Science, University of Bath, Bath, UK and cPion Inc. (UK) Ltd., Forest Row, East Sussex, UK

Keywords Abstract
drug-drug interaction; GI pH; GI motility;
permeability; metabolism Objectives Drugs used to treat gastrointestinal diseases (GI drugs) are widely
used either as prescription or over-the-counter (OTC) medications and belong
Correspondence to both the 10 most prescribed and 10 most sold OTC medications worldwide.
Jennifer B. Dressman, Biocenter, Institute of
The objective of this review article is to discuss the most frequent interactions
Pharmaceutical Technology, Johann
Wolfgang Goethe University, Max-von-Laue-
between GI and other drugs, including identification of the mechanisms behind
Str. 9, Frankfurt am Main 60438, Germany. these interactions, where possible.
E-mail: dressman@em.uni-frankfurt.de Key findings Current clinical practice shows that in many cases, these drugs are
administered concomitantly with other drug products. Due to their metabolic
Received March 23, 2018 properties and mechanisms of action, the drugs used to treat gastrointestinal dis-
Accepted June 27, 2018 eases can change the pharmacokinetics of some coadministered drugs. In certain
cases, these interactions can lead to failure of treatment or to the occurrence of
doi: 10.1111/jphp.12983
serious adverse events. The mechanism of interaction depends highly on drug
properties and differs among therapeutic categories. Understanding these interac-

Chara Litou and Angela Effinger contributed tions is essential to providing recommendations for optimal drug therapy.
equally to this work. Summary Interactions with GI drugs are numerous and can be highly significant
clinically in some cases. While alterations in bioavailability due to changes in sol-
ubility, dissolution rate, GI transit and metabolic interactions can be (for the
most part) easily identified, interactions that are mediated through other mecha-
nisms, such as permeability or microbiota, are less well-understood. Future work
should focus on characterising these aspects.

and stomach discomfort. The treatment of functional dys-


Introduction
pepsia can involve various drug classes depending on the
It is estimated that 60–70 million US-Americans suffer symptoms as well as the possible causative factors.[4–6]
annually from various types of gastrointestinal (GI) dis- Crohn’s disease, by contrast, is a chronic inflammatory dis-
eases, with GI diseases being the underlying cause of order that can affect any part of the GI tract from the
approximately 10% of all deaths in the USA.[1,2] In fact, sta- mouth to the anus. Although as of yet there is no cure for
tistical data on global sales of prescription medication from Crohn’s disease, there are several treatment options which
2014 indicate that sales of drug products for the treatment can relieve the symptoms and prevent relapse.[7] As illus-
of GI diseases rank 12th with regard to sales of prescription trated by these two examples, it is evident that a diversity of
medication worldwide.[3] drugs with different mechanisms of action is required to
The term gastrointestinal diseases covers a wide range of address the various targets across the spectrum of GI dis-
disorders, which can be either acute or chronic. Non-ulcer eases.
or functional dyspepsia, for example, is usually an acute Frequently, patients are prescribed several drugs con-
condition that affects the upper GI tract and is expressed by comitantly. Drug–drug interactions (DDIs) are a common
symptoms such as nausea, vomiting, heartburn, bloating problem during drug treatment and can sometimes lead to

© 2018 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 71 (2019), pp. 643–673 643
Pharmacokinetic interactions with GI drugs Chara Litou et al.

failure of treatment, or can cause serious or even fatal interactions with over-the-counter medication (OTC).
adverse events.[8] FDA publishes information leaflets for consumers about
Medications used for the treatment of GI diseases can the most typical drug interactions that occur with speci-
alter the GI physiology and thus interact with the absorp- fic OTC medications. It is interesting to note that four
tion of concomitant medications, but they can also alter the out of the 12 drugs discussed by FDA in these leaflets
metabolism and/or elimination of coadministered drugs, involve drugs used to treat gastrointestinal diseases.[11]
potentially resulting, on the one hand, in a lack of efficacy European statistics indicate that there may be similar
of the coadministered drug or, on the other hand, in issues with concomitant use of OTC medication in the
adverse drug reactions. From a regulatory perspective, European Union, since 20–70% of those surveyed
studies of potential DDIs which lead to changes in absorp- reported using OTC medicines.[12]
tion are required for the marketing authorisation of medic- Keeping in mind these statistics, as well as the fact that
inal products in the European Union and United States.[8,9] medications used to treat GI diseases count among the 10
In particular, these studies are designed to evaluate the most prescribed medicines – and also fall within the top 10
effect of increased GI pH, the possibility of complexation in terms of sales of OTC medications – worldwide,[3,13] it is
and alterations in GI transit time.[8] Understanding the evident that there is a high potential for DDIs with these
effect of GI drugs on the physiology of the GI tract and medications.
achieving a mechanistic understanding of the interaction(s) The objective of this review was first to present and
involved are key to successfully managing concomitant discuss the effects of drugs used to treat GI diseases,
drug therapy. both prescription and OTC, on the pharmacokinetics
In clinical trials, drug performance is determined under and bioavailability of coadministered drugs and second,
controlled conditions (e.g. with strict inclusion/exclusion to identify the mechanisms behind these interactions
criteria, under absence of, or controlled comedication and insofar as possible. The review is organised according to
with monitoring of compliance). But, in clinical practice, the therapeutic indication of the drug (see Figure 1 for
where a much wider variety of patient characteristics, dis- an overview) and covers drugs used to prevent/treat all
ease states and multimorbidity is usual, the potential for major GI diseases. Although several reviews concerning
DDIs is much greater. In fact, statistics show that one in a DDIs of specific GI drug classes, for example proton-
hundred hospital admissions occurs as a result of a drug– pump inhibitors (PPIs), are available in the literature, to
drug interaction.[10] The number of unreported/less severe the best of these authors’ knowledge, this is the first to
interactions is probably far greater. provide an overview of interactions that are likely to
In addition to potential interactions with prescription occur across the range of drugs used to treat GI dis-
drugs, one must also consider the possibility of eases.

Figure 1 Gastrointestinal drugs discussed in this review. [Colour figure can be viewed at wileyonlinelibrary.com]

644 © 2018 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 71 (2019), pp. 643–673
Chara Litou et al. Pharmacokinetic interactions with GI drugs

Medicines used to treat


gastrointestinal diseases and their
effect on coadministered drugs

Agents affecting gastrointestinal motility


Various neurotransmitters have an effect on GI motility
and its coordination. Dopamine, for example, is present in
significant amounts in the GI wall and has an inhibitory
effect on motility.[14,15] Dopamine receptor antagonists are
currently being used for motor disorders of the upper GI
tract, gastroesophageal reflux disease, chronic dyspepsia
and gastroparesis and have also been investigated for ther-
apy of motility disorders of the lower GI tract.[16,17] Acetyl-
choline, by contrast, stimulates GI motility through
increased contractile activity by the smooth muscle.[18,19]
Serotonin, which is mainly present in the enterochromaffin
cells in the enteric epithelium and colon, has a wide range
of effects on the GI tract. The diversity of effects can be
explained by the presence of multiple subtypes of 5-HT
receptors, located on different types of cells. Both agonists
and antagonists of 5-HT receptors are used for the treat-
ment of GI diseases.[20,21]

Prokinetic agents
Prokinetic agents promote gut wall contractions and
increase their coordination, thus enhancing GI motility.
However, they do not disrupt the normal physiological pat-
tern of motility.[16,17]
Metoclopramide. Metoclopramide is a first-generation proki-
netic agent with antidopaminergic properties (D1 and D2
receptor antagonist). In addition, metoclopramide is a 5-HT3 Figure 2 Gastric emptying results in 12 gastroesophageal reflux
receptor antagonist and a 5-HT4 receptor agonist. Metoclo- patients with delayed basal emptying rates (a) and in 14 gastroe-
pramide promotes the response to acetylcholine in the upper sophageal reflux patients with normal basal emptying rates (b), in a
two-way crossover design consisting of a control phase and a phase
GI tract and therefore accelerates gastric emptying and
in which 10 mg metoclopramide was ingested orally. The data are
increases the tone of the lower oesophageal sphincter.[22] The
expressed as the mean percent (1 SEM) isotope remaining in the
effect is observed in both healthy volunteers and those with stomach for a period of 90 min after ingestion of an isotope-labelled
GI diseases.[23–25] For example, Fink et al.[25] demonstrated test meal.[25] Figure reprinted from Fink et al. with permission from
that metoclopramide accelerates gastric emptying in patients Springer Nature.
with gastroesophageal reflux disease independent of their gas-
tric emptying status (Figure 2a and 2b). Metoclopramide is
used for the symptomatic treatment of postoperative or
tolfenamic acid in patients diagnosed with migraine.
chemotherapy-induced nausea and vomiting, gastroe-
According to the protocol, the volunteers took part in the
sophageal reflux disease and gastroparesis.[23] A summary of
absorption studies twice in the absence of migraine and
the effects of concomitant use of metoclopramide on the
twice as soon as possible after the beginning of a migraine
absorption of several APIs is presented in Table 1, and mech-
attack. After rectal administration of metoclopramide, the
anistic explanations for the observed effects are presented in
absorption of the tolfenamic acid was accelerated compared
the following text.
to control (rectal administration of placebo) in all subjects.
It is known that migraine attacks are often accompanied However, the total bioavailability of tolfenamic acid did
by delayed gastric emptying.[26] Tokola et al.,[27] investi- not change significantly.[27] A similar study had been con-
gated the effect of metoclopramide on the absorption of ducted in 1975 by Volans, in which the effect of

© 2018 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 71 (2019), pp. 643–673 645
Pharmacokinetic interactions with GI drugs Chara Litou et al.

Table 1 Reported pharmacokinetic interactions with metoclopramide

Effect

Interaction with Rate of absorption Cmax Tmax AUC References

Drug–drug Acetaminophen ↑ ↑ ↓ Nimmo et al. (1973)[30]


interactions with Cimetidine ↓ ↓ Gugler et al. (1981)[36]
metoclopramide ↓ Lee et al. (2000)[341]
Cyclosporine ↑ ↓ ↑ Wadhwa et al. (1986)[42]
Digoxin ↓ ↓ (only for tablet) Johnson et al. (1984)[41]
↓ Manninen et al. (1973)[40]
Droxicam – ↓ – S
anchez et al. (1989)[33]
Levodopa ↑ ↑ ↓ Morris et al. (1976)[35]
Lithium ↓ Crammer et al. (1974)[32]
Methotrexate ↓ (paediatrics) Mahony et al. (1984)[37]
Mexiletine ↑ – Wing et al. (1980)[31]
Morphine – ↓ – Manara et al. (1988)[34]
Salicylic acid ↑ plasma levels (in patients Volans et al. (1975)[28]
with migraine attacks)
Tetracycline ↓ – Gothoni et al. (1972)[29]
Tolfenamic acid ↑ – Tokola et al. (1984)[27]

metoclopramide on the absorption of aspirin during dosing of mexiletine, the antiarrhythmic effect is unlikely
migraine attacks was investigated.[28] In that study, the to change after concomitant use of metoclopramide.[31] In
delayed gastric emptying during a migraine attack was con- a further study by Crammer et al.,[32] it was shown that
firmed. In addition, it was shown that the plasma levels of metoclopramide reduced the tmax of coadministered
salicylate achieved during a migraine attack, after intramus- lithium by 2 h. Sanchez et al.,[33] investigated the effect of
cular administration of metoclopramide, were higher in intravenous metoclopramide on the absorption of droxi-
comparison with those achieved without metoclopramide cam (a piroxicam prodrug) and Manara et al.,[34] investi-
pretreatment. gated the effect of oral metoclopramide after concomitant
Gothoni et al.[29] reported an earlier time to achieve administration of an oral controlled release formulation of
maximum plasma concentration (tmax) and elevated serum morphine. In both cases, a significant reduction in tmax was
tetracycline concentrations in six healthy volunteers after observed, but other pharmacokinetic parameters were not
coadministration of tetracycline with intramuscular meto- significantly different.[33,34] Thus, in most studies, it has
clopramide. Nonetheless, the total area under the curve been demonstrated that although concomitant administra-
(AUC) remained unaltered. In the same study, an increase tion of metoclopramide increases absorption rate, there is
in the rate of absorption of oral pivampicillin was reported little or no effect on AUC, or clinical efficacy.
when administered along with metoclopramide.[29] In a study by Morris et al.,[35] it was likewise observed
Concomitant administration of metoclopramide has also that the coadministration of metoclopramide resulted in an
been shown to increase the absorption rate of acetamino- increased rate of absorption of levodopa and higher peak
phen, mexiletine, lithium, droxicam and morphine. plasma concentrations, consistent with the earlier tmax. In
Nimmo et al.,[30] studied the absorption of acetaminophen this case, though, the authors emphasised the fact that
with and without coadministration of metoclopramide in higher peak concentrations of levodopa may result in dysk-
five healthy volunteers. The mean tmax was reduced from inetic movements, and therefore, this should be taken into
120 to 48 min while the mean maximum plasma concen- consideration when metoclopramide is coadministered
tration (Cmax) increased from 125 to 205 lg/ml. The uri- with levodopa.
nary excretion of acetaminophen was not influenced. Given Considering the properties of metoclopramide and the fact
the fact that tmax is a function of both absorption and elimi- that besides promoting gastric emptying, it also increases the
nation rates, the shortened tmax after pretreatment with upper small intestinal motility, administration of metoclo-
metoclopramide indicates an enhanced absorption rate.[30] pramide could also decrease the time available for absorption
Similar results were obtained in the study of Wing et al.,[31] in the small intestine and thus lead to a reduction in total
in which the authors demonstrated an increased absorption bioavailability. Gugler et al.,[36] explored this hypothesis by
rate of mexiletine after coadministration of metoclo- studying the absorption of cimetidine when given concomi-
pramide. Here too, it was observed that the bioavailability tantly with antacids or metoclopramide. The study was con-
of mexiletine was unaltered, indicating that during chronic ducted in eight healthy volunteers and showed that there was a

646 © 2018 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 71 (2019), pp. 643–673
Chara Litou et al. Pharmacokinetic interactions with GI drugs

tendency to a shorter time to reach maximum plasma reported examples are limited, it appears that after coad-
concentrations when metoclopramide was coadministered. ministration of metoclopramide small intestinal transit
Additionally, a decrease in AUC of approximately 22% was may be too fast for poorly permeable (e.g. cimetidine) or
observed, although in neither case did the difference reach sta- poorly dissolving (e.g. digoxin) drugs to be adequately
tistical significance.[36] On the other hand, Mahony et al.,[37] absorbed. Thus, in this case, BCS classification may be
conducted a clinical study with children with leukaemia and helpful in identifying potential problems in bioavailability
reported that concomitant administration of methotrexate when metoclopramide is coadministered.
tablets with oral metoclopramide led to significantly lower
AUC. Consistent with these findings, Pearson et al.,[38]
Anticholinergic agents
demonstrated that a very fast or slow small intestinal transit in
children with leukaemia reduces the Cmax of methotrex- Propantheline is an anticholinergic agent which reduces
ate.[37,38] gastrointestinal motility and prolongs gastric emptying
In the studies conducted by Manninen et al.,[39,40] coad- rate. It is usually used in combination with other medicines
ministration of metoclopramide with digoxin in eight to treat stomach ulcers. As for metoclopramide, propanthe-
healthy adults or in 11 patients on digoxin therapy resulted line has been investigated with respect to its potential effect
in reduced serum digoxin concentrations. The lower on the absorption of concomitant medications. As one
bioavailability of digoxin was attributed to its dissolution would anticipate, propantheline decreased the absorption
rate-limited absorption, as the changes were only observed rate of acetaminophen and lithium when given concur-
when digoxin was given as a tablet and not when it was rently.[30,32] Coadministration of propantheline with a
given as a solution. For this reason, authors suggested that rapidly and a slowly dissolving tablet of digoxin resulted in
fast dissolving tablets of digoxin would be less affected by increased serum digoxin concentrations only for the slowly
coadministration of drugs which alter the GI motility. Sup- dissolving formulation.[39,40]
porting this hypothesis, Johnson et al.,[41] demonstrated
that digoxin was absorbed completely and more quickly
Laxatives
when it was given as soft-gelatine capsules rather as a tablet.
Oral metoclopramide reduced the tmax for both formula- Laxatives promote defecation and are often used OTC for
tions, but only reduced the AUC of the tablet formula- the treatment of constipation. They can be grouped in
tion.[41] From these two studies, it is apparent that osmotic, stimulant and bulk laxatives (Table 2).[43] An
coadministration of metoclopramide may result in overview of the effects of laxatives and antidiarrheal agents
impaired drug absorption and decreased bioavailability in on gastrointestinal physiology is given in Table 3. Osmotic
cases when a poorly soluble API exhibits dissolution rate- laxatives (indigestible disaccharides, sugar alcohols, syn-
limited absorption. thetic macromolecules, saline laxatives) attract and retain
In contrast to the results discussed above, Wadhwa water in the intestinal lumen by increasing the luminal
et al.,[42] conducted a clinical study in 14 kidney transplant osmotic pressure. Stimulant laxatives (such as bisacodyl,
patients with the aim of increasing the bioavailability of senna and sodium picosulfate) act locally by increasing
cyclosporine. Cyclosporine is incompletely absorbed in the colonic motility and decreasing water absorption in the
small intestine with a dose-dependent rate and extent of large intestine.[44] Bulk laxatives such as bran, isphagula
absorption. The authors reasoned the concomitant admin- and sterculia adsorb and retain luminal fluids and increase
istration of cyclosporine with metoclopramide would the faecal mass. For constipation linked with specific dis-
increase the absorption rate and possibly the bioavailability eases, additional treatment options are available: lina-
of this immunosuppressive. Due to accelerated gastric emp- clotide, an agonist of guanylate cyclase-C, stimulates fluid
tying, there was a very significant increase in the Cmax of secretion, accelerates intestinal transit and is used for con-
cyclosporine, as well as a decrease in tmax. Furthermore, an stipation-predominant irritable bowel syndrome.[45]
average increase of 29% in the AUC was observed In general, laxatives shorten GI transit time, but depend-
(P = 0.003). However, the authors concluded that further ing on the type of laxative, the extent of the effect on transit
studies would be required to determine whether metoclo- time through specific GI compartments may vary (Fig-
pramide can reproducibly increase the absorption of ure 3). Studies have been conducted with a variety of meth-
cyclosporine on a long-term basis.[42] ods including radiopaque markers,[46–48] following transit
Overall, it appears that coadministration of metoclo- of a single metal sphere (diameter 6 m, density 1.4 g/ml)
pramide leads to a decreased tmax of the coadministered using a metal detector,[49] [13C]-octanoate and lactose-
drugs, indicating a faster rate of absorption. However, the [13C] ureide breath tests[50] and scintigraphy.[45,51–54]
effect of concomitant use of metoclopramide on the AUC For healthy subjects, the following observations have
of the coadministered drug is variable. Although the been reported: the total GI transit time was reduced in 13

© 2018 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 71 (2019), pp. 643–673 647
Pharmacokinetic interactions with GI drugs Chara Litou et al.

Table 2 Classification of laxatives and antidiarrheal agents[43–45]

Class Subgroup Examples

Laxatives Osmotic laxatives Indigestible disaccharides Lactulose


Sugar alcohols Sorbitol
Synthetic macromolecules Polyethylene glycol 4000
Saline laxatives Sodium sulphate
Magnesium sulphate
Stimulant laxatives Bisacodyl
Senna
Phenolphthalein
Casanthranol
Sodium picosulfate
Bulk laxatives Wheat bran
Isphagula
Sterculia
Others Linaclotide
Antidiarrheal agents Opioids Loperamide
Diphenoxylate
Codeine phosphate
Adsorbents/Bulking agents Kaolin
Isphagula
Methylcellulose
Miscellaneous Racecadotril

Table 3 Effects of laxatives and antidiarrheal agents on gastrointestinal conditions[45,46,49,51–54,58–60,65,342,343]

Drug category Effect on GI physiology

Laxatives ↓ Gastrointestinal transit time Small intestinal transit time (bisacodyl)


Colonic transit time (bisacodyl, linaclotide, lactulose, polyethylene glycol)
Whole gastrointestinal transit time (wheat bran, senna, bisacodyl)
pH in the colon ↓ pH (lactulose, senna, wheat bran, sodium sulphate)
↑ pH (magnesium sulphate)
Faecal short chain fatty acids ↑ (bisacodyl, senna, wheat bran)
Differences in gut microbiota ↑ Anaerobes, Bifidobacteria (lactulose)
↓ Bifidobacteria (polyethylene glycol-4000)
Haustra (small pouches in the colon) ↓ (chronic use of stimulant laxatives)
Antidiarrheal agents ↑ Gastrointestinal transit time ↑ intestinal transit time (loperamide)
Faecal short chain fatty acids ↑ (loperamide)

subjects after treatment for 9 days with either the bulk laxa- Laboratories Ltd., West End, Southampton, UK) three
tive wheat bran (39.0 h vs 69.0 h) or the stimulant laxative times daily for 5 days resulted in a significant decrease of
senna (41.0 h vs. 69.0 h) compared to the baseline the mean proximal colon transit time from 12.9  3.7 h to
value.[46] Small intestinal transit time was reduced by bisa- 7.0  2.5 h in 11 subjects.[53] The total colonic transit time
codyl (dose 10 mg) from approximately 2.5 to 1.5 h in 10 was reduced to a greater extent after administration of
subjects,[49] while the osmotic laxatives polyethylene glycol 10 mg bisacodyl (from 31  14 h to 7  8 h) than by
and lactulose had a minimum effect (if any) on the small treatment with 30 g lactulose (from 34  12 h to
intestinal transit time after being administered at a dose of 30  19 h) in 10 subjects.[49]
10 g twice daily for 5 days.[51] Administration of an isos- In patient populations, the following observations have
motic solution containing 40 g polyethylene glycol 3350 been reported: in 12 subjects with constipation-predomi-
resulted in a significant decrease in oro-caecal transit time nant irritable bowel syndrome, treatment with linaclotide
from 423.8  28.1 min to 313.8  17.2 min in 12 sub- (dose 100 lg or 1000 lg) did not affect the gastric or small
jects.[50] In another study, administration of 5 mg bisaco- intestinal transit time.[45] However, the ascending colon
dyl in 25 subjects significantly accelerated the transit transit time was decreased by 54% at a high dose of
through the ascending colon (median 6.5 h vs 11.0 h).[54] 1000 lg of linaclotide. At a lower dose of 100 lg, there was
Similarly, 10–20 ml of lactulose (Duphalac; Duphar a decrease of 33%, although this was not statistically

648 © 2018 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 71 (2019), pp. 643–673
Chara Litou et al. Pharmacokinetic interactions with GI drugs

Figure 3 Impact of laxatives on colonic transit times of (a) healthy subjects and (b) patients, measured by scintigraphy (1), metal detector (2) or
radiopaque markers (3); patterned bars represent controls.[45,47–49,53,54] [Colour figure can be viewed at wileyonlinelibrary.com]

significant. In line with these observations, the total colonic supraventricular extrasystoles.[55] Similarly, polyethylene
transit time was only significantly accelerated by the higher glycol 4000 reduced the absorption of digoxin by 30%
dose.[45] In nine subjects with chronic non-organic consti- when coadministered with digoxin tablets (dose 0.5 mg) in
pation, treatment with an isosmotic electrolyte solution 18 healthy subjects.[56] However, it is not clear whether the
containing polyethylene glycol 4000 (14.6 g) for 8 weeks same effect would be observed in cardiac patients or what
did not significantly alter the transit time through the prox- the clinical ramifications would be. Further, a trend
imal colon, while the transit through the left colon and rec- (although not statistically significant) to decreased AUC of
tum was significantly accelerated (46  29 h vs 62  20 h oestradiol glucuronide (dose 1.5 mg) was observed when
and 37  42 vs 78  21 h, respectively).[48] The results in coadministered for 10 days with the maximum tolerated
eight patients with slow transit constipation were similar dose of wheat bran ( 13%) and senna ( 10%) in 20
after administration of 60 g polyethylene glycol 4000 daily healthy postmenopausal women.[57]
for 6 weeks; the right colon transit time was not signifi- Many laxatives have been shown to alter the production
cantly different compared to placebo, while the transit time of short-chain fatty acids (SCFA). SCFA are usually associ-
through the left colon was significantly accelerated (13 h vs ated with a decrease in luminal pH. After treatment with
45 h) resulting in a reduction in total colonic transit time senna or wheat bran, faecal SCFA concentrations were
from 91 h to 43 h.[47] In summary, laxatives decrease tran- increased in healthy subjects (n = 13) by 82% and 19%,
sit times in healthy subjects throughout the GI tract, while respectively.[46] After administration of senna, the pH in
in constipated patients, the effects are mainly limited to the the middle and distal colon was decreased (6.39 vs 6.85,
colon. 6.66 vs 7.14).[46] Lactulose significantly acidified the con-
Changes in GI transit times induced by laxatives can lead tents in the lower small intestine as well as in the right
to changes in bioavailability. For example, coadministra- colon.[58–60] Sodium sulfate also decreased the pH, with
tion of senna (20 ml of Liquidepur; Fa. Nattermann, the greatest effect in the left colon.[58] By contrast, wheat
Cologne, Germany) with a sustained-release quinidine for- bran reduced the pH in the distal colon of 13 healthy sub-
mulation (0.5 g every 12 h) reduced quinidine plasma jects only slightly (6.88 vs 7.08).[46] But mechanisms other
levels by 25% in nine patients with cardiac arrhythmia on than via SCFA can also be at play. For example, the
long-term treatment, resulting in reoccurrence of increase in the pH in the lower small intestine, colon and

© 2018 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 71 (2019), pp. 643–673 649
Pharmacokinetic interactions with GI drugs Chara Litou et al.

rectum observed after administration of magnesium sul- by increasing fluid absorption and/or by reducing intestinal
phate is postulated to be the result of gastric conversion to secretions.[67] They can be classified according to their
magnesium chloride and subsequent reconversion to mechanism of action (Table 2). Opioids (such as lop-
insoluble magnesium carbonate in the colon prompted by eramide, diphenoxylate and codeine phosphate) inhibit
increased colonic bicarbonate secretion.[58] The possible intestinal transit by activating l-opioid receptors. Adsor-
pH changes observed with laxatives are not clearly associ- bents and bulking agents (kaolin, isphagula, methylcellu-
ated with changes in drug product performance. For lose) adsorb water and increase the faecal mass, while the
example, mesalazine release from a delayed-release, antisecretory action of racecadotril, an enkephalinase inhi-
pH-dependent formulation of mesalazine (Asacolâ, bitor, is linked to reducing chloride and fluid flux into the
SmithKline Beecham, Brentford, UK) was not affected by GI lumen.
the coadministration of ispaghula husk or lactulose despite Differences in the GI transit time have been observed
their known pH-lowering effect in the colon.[61,62] after oral loperamide administration (Figure 4). The total
Nonetheless, the UK manufacturers of delayed-release GI transit time was increased after loperamide administra-
mesalazine formulations (Asacolâ, Allergan Ltd, Bucks, tion in healthy subjects (74.0 h vs 50.3 h, n = 11), as mea-
UK and Salofalkâ granules, Dr. Falk Pharma UK Ltd, sured by radiopaque marker pellets, presumably due to
Bourne End, UK) suggest that drug release might be reduced, irregular motor activity and therefore prolonged
impaired by preparations with pH-lowering effect.[63,64] transit time in the jejunum.[46,68,69] Gastric emptying time
With respect to the gut microbiota, the faecal microbiota was not significantly different in 24 healthy subjects treated
of patients with chronic idiopathic constipation (n = 65) with 4 mg loperamide compared to placebo as measured
treated with lactulose over 28 days was increased in Anaer- with a radio-labelled meal.[70] However, gastric residence
obes by 3% and Bifidobacteria by 8%, while treatment with time measured with a radiotelemetry capsule was increased
polyethylene glycol 4000 resulted in a reduced faecal twofold in five healthy subjects treated with 8 mg lop-
amount of Bifidobacteria ( 14%).[65] Lactulose adminis- eramide (four doses, every 6 h).[71] Small intestinal transit
tration in patients taking coumarins (acenocoumarol, time, as measured with the hydrogen breath test, was
phenprocoumon) increased their risk of over-anticoagula- increased by 80–130% in healthy subjects receiving 4–8 mg
tion, as assessed in a population-based cohort study, of loperamide.[70–72]
because of changes in the vitamin K production of the colo- With respect to the composition of GI fluids, loperamide
nic bacterial flora. By contrast, concomitant intake of isph- has been shown to decrease prostaglandin-E2 induced
agula with coumarins did not alter the risk of over- water and electrolyte secretion in the jejunum of healthy
anticoagulation.[66] volunteers and reduce postprandial secretion of trypsin and
The importance of the gut microbiota on oral pharma- bilirubin by more than 50% in patients with short bowel
cotherapy is discussed further in the section on ‘Antibiotics’. syndrome.[69,73,74] Similarly, basal and amino acid stimu-
lated gallbladder motility was decreased by loperamide
(dose 8 mg) in eight healthy subjects as measured by ultra-
Antidiarrheal agents
sonography and bilirubin output in the duodenum.[75]
Antidiarrheal agents provide symptomatic relief of diar- After loperamide administration, faecal SCFA concentra-
rhoea by decreasing fluid loss, by slowing down the passage tions were decreased in healthy subjects (82.0 lmol/g wet
of the gastrointestinal contents through the digestive tract, weight vs 152.0 lmol/g wet weight; n = 13).[46]

Figure 4 Effect of loperamide on gastrointestinal transit time after oral administration in healthy subjects.[46,70–72] [Colour figure can be viewed
at wileyonlinelibrary.com]

650 © 2018 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 71 (2019), pp. 643–673
Chara Litou et al. Pharmacokinetic interactions with GI drugs

In terms of DDIs, administration of 4 mg loperamide disorders, has increased over the last years. However, there
24, 12 and 1 h before desmopressin administration are few studies that have investigated the impact of con-
increased the bioavailability of desmopressin in 18 healthy comitant use of dietary fibres with other drugs. From the
subjects (AUC 3.1-fold, Cmax 2.3-fold) and prolonged the studies available, it seems that the effect of the concomitant
time to reach the maximum plasma concentration (2 h vs use of dietary fibres depends on the type of fibre used.
1.3 h) without affecting the elimination half-life.[76] These The interaction of levothyroxine with dietary fibres is
effects could be explained by the decrease in GI motility. well-established. Concomitant use of dietary fibres, such as
Desmopressin is highly soluble but poorly permeable oat bran, soy fibre and ispaghula husk, result in decreased
(bioavailability approximately 0.1%), so longer transit bioavailability of levothyroxine, due to adsorption of the
times are expected to lead to a longer contact time of the drug to the fibres in the GI tract.[88] The authors com-
drug with the absorptive mucosa.[77] Coadministration of mented that the adsorption of levothyroxine to soluble
loperamide at the maximum tolerated dose over 10– fibres and the consequent reduction in bioavailability might
12 days also increased the AUC of estradiol glucuronide be greater than its adsorption to insoluble fibres. The inter-
(dose 1.5 mg) by 15% in 20 healthy postmenopausal action with levothyroxine is also noted by FDA in a con-
women, although the difference did not reach statistical sig- sumers’ information leaflet regarding drug interactions
nificance.[57] with food.[89]
On the other hand, a single dose of loperamide (16 mg) In a case study reported by Perlman, the blood levels of
decreased the bioavailability of the poorly soluble drug lithium were decreased by 48%, when a patient was treated
saquinavir (dose 600 mg) by 54% in 12 healthy subjects simultaneously with lithium and ispaghula husk.[90] There
when administered concomitantly. This could be explained is also some evidence that fibres interact with some tricyclic
by the decreased motility and/or a reduction in electrolyte antidepressants. The clinical effectiveness of tricyclic
and fluid secretion both of which could hinder dissolu- antidepressants appears usually after an administration per-
tion.[78] Additionally, it is possible that a decreased secre- iod of 2–6 weeks. During this period, due to anticholiner-
tion of bile salts secondary to reduced gallbladder gic effects of the drugs, constipation is a common side
motility[75] impeded the solubilisation of saquinavir. effect. Therefore, patients receiving antidepressant medica-
On the other hand, loperamide coadministration (8 mg tion often ingest dietary fibres. Already in 1992, Stewart
every 6 h) in 12 healthy male subjects decreased the absorp- observed a decrease in plasma concentrations of three tri-
tion rate of theophylline from a sustained-release 600 mg cyclic antidepressants (amitriptyline, doxepin and imipra-
formulation (Cmax 3.2 mg/l vs 4.6 mg/l, tmax 20 h vs 11 h), mine) in three patients, who concurrently ingested a diet
which could be explained by impeded release from the for- rich in fibres.[91]
mulation due to a decrease in hydrodynamics (decreased There are conflicting inputs in the literature about the
motility) or perhaps a prolonged gastric residence time of interaction of dietary fibres and digoxin. Brown et al.,[92]
the formulation/released drug. However, the AUC was not reported a significant decrease in the bioavailability of
affected.[72] digoxin when given concomitantly to 12 healthy volunteers
Last but not least, the surface of bulk laxatives and bulk- ingesting a regular or high fibre diet, as opposed to admin-
ing agents offers a site for drug adsorption. Concomitant istering digoxin alone in the fasted state. Albert et al.,[84]
administration of kaolin-pectin decreased the absorption of reported that when kaolin-pectin suspension was given
tetracycline (20%), aspirin (5–10%), procainamide (30%), simultaneously with digoxin, the total amount of digoxin
quinidine (58%), trimethoprim (12–20%), lincomycin absorbed was decreased by 62%. However, no significant
(90%), chloroquine (29%) and digoxin (15–62%), which is interactions were observed when digoxin was given 2 h
most likely the result of adsorption of the drugs onto before the administration of the fibre suspension.[84] How-
kaolin.[79–87] Drug adsorption is also observed onto dietary ever, studies by Lembcke et al., and Kasper et al., found no
fibres, and therefore, similar DDIs to those observed effect on the bioavailability of digoxin when it was adminis-
with dietary fibres are further considered in the section on tered together with guar gum or other fibres.[93,94] In a later
‘Dietary Fibres.’ study Huupponen et al.,[95] investigated the effect of guar
An overview of the effects of antidiarrheal agents on gas- gum on the absorption of digoxin in 10 healthy volunteers.
trointestinal physiology is given in Table 3. It was demonstrated that coadministration of guar gum
with digoxin resulted in reduced plasma concentrations of
digoxin and a decrease of 15% of the AUC for the first 6 h
Dietary fibres
(P < 0.05).[95]
The use of dietary fibres in the treatment of various dis- Holt et al.,[96] investigated the effect of coadministration
eases, such as diabetes, hypercholesterolemia, obesity, of the soluble fibres guar gum and pectin on the absorption
chronic constipation and gastrointestinal motility of acetaminophen. Concomitant administration with these

© 2018 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 71 (2019), pp. 643–673 651
Pharmacokinetic interactions with GI drugs Chara Litou et al.

fibres resulted in delayed absorption and decreased Cmax. Aprepitant is metabolised primarily by CYP3A4 and secon-
However, the total absorption of acetaminophen was not darily by CYP1A2 and CYP2C19. It also acts as a moderate
significantly reduced. The authors attributed their results to inhibitor of CYP1A2, CYP2C9, CYP2C19, CYP2E1 and as a
delayed gastric emptying. Moreover, they argued that weak inducer of CYP2C.[101,102] Caution is therefore neces-
because guar gum, when hydrated, forms a viscous colloidal sary, especially when administered concomitantly with
suspension, the high viscosity of this suspension could be a chemotherapy agents that are metabolised primarily by
possible reason for the observed delay in gastric empty- CYP3A4, as inhibition by aprepitant may lead to higher
ing.[96] The results from this study correlate well with the plasma levels and toxic side effects. According to the Public
study conducted by Reppas et al.,[97] in mongrel dogs, in Assessment Report, EMENDâ capsules (which contain
which the effect of elevated luminal viscosity on the absorp- aprepitant as API) should not be concomitantly adminis-
tion of acetaminophen, hydrochlorothiazide, cimetidine tered with ergot alkaloid derivatives, pimozide, terfenadine,
and mefenamic acid was investigated. Elevated luminal vis- astemizole or cisapride, as the competitive inhibition of the
cosity was achieved by administering saline solutions of the CYP3A4 by aprepitant results in elevated plasma concentra-
water-soluble guar gum. When given concurrently with the tions, leading to adverse effects.[102] Further pharmacoki-
guar gum solutions, the Cmax and AUC of the highly sol- netic interactions that have been reported for aprepitant in
uble acetaminophen and hydrochlorothiazide were signifi- the literature are those with midazolam, warfarin, dexam-
cantly decreased, suggesting that the decreased rate of ethasone and methylprednisolone.[22,103]
dissolution, due to the higher luminal viscosity, led to Majumdar et al.,[104] investigated the effect of aprepi-
lower concentrations at the absorption sites. In the case of tant on the pharmacokinetics of single dose midazolam on
cimetidine, concurrent administration of the guar gum day 1 and on day 5 during daily administration of aprepi-
solution led only to a decrease in Cmax and not AUC. For tant for 5 days. In this study, two dose regimens of aprepi-
the poorly soluble but highly permeable mefenamic acid, tant were used: 125/80 and 40/25 mg. It was concluded
neither the Cmax nor the AUC were significantly affected by that coadministration of midazolam with the 125/80 mg
the concomitant administration of the guar gum in regimen (125 mg on day 1 and 80 mg on days 2–5)
dogs.[97] Huupponen et al.,[95] reported a decrease in Cmax resulted in a 2.3-fold increase in midazolam AUC on day
and AUC of penicillin when given together with guar gum. 1 and a 3.3-fold increase on day 5. The plasma concentra-
Finally, Astarloa et al.,[98] investigated the effect of a diet tions achieved 1 h after dosing (C1h) and the half-life (t1/
rich in insoluble fibre on the pharmacokinetics of levodopa. 2) were also increased due to the inhibition of first pass
Consumption of 2 months of the dietary supplement with and systemic metabolism and subsequent reduction in
the usual dose of levodopa led to elevated plasma levels of clearance. Although coadministration of midazolam with
levodopa especially at 30 and 60 min after oral administra- the 40/25 mg dose regimen did not result in any signifi-
tion.[98,99] cant change in the pharmacokinetics of midazolam, this
It is evident from these studies that it is currently not lower dose is not used in clinical practice.[104] Majumdar
possible to make any generalisations about DDIs with diet- et al.,[105] later investigated the effect of aprepitant on
ary fibres although it seems that there is a tendency for intravenously administered midazolam and the findings
decreased maximum plasma concentrations of the coad- were consistent with the first study, but with an increase
ministered drug. These events are likely attributable to in AUC of 1.47-fold. The authors suggested that the lower
slower gastric emptying, higher viscosity and, perhaps in increase in AUC observed after intravenous administration
some cases, adsorption phenomena.[100] It also seems that of midazolam might be due to circumvention of the pre-
the type of interaction, if any, is highly dependent on the systemic metabolic interaction when midazolam is given
type of dietary fibre used. It remains to be investigated intravenously.[105]
whether these interactions, such as they exist, lead to clini- In an analogous study by McCrea et al.,[106] the effect of
cally significant differences. a 5-day administration of 125/80 mg aprepitant regimen
on the pharmacokinetics of orally administered methyl-
prednisolone and dexamethasone was evaluated. Due to the
Antiemetics
inhibition of CYP3A4 by aprepitant, the Cmax of methyl-
Antiemetics are classified according to their mechanism of prednisolone was increased 1.5-fold while the AUC
action. There are five receptors that play a key role in the increased 2.5-fold. An increase of 2.2-fold in AUC was
vomiting reflex; muscarinic, dopaminergic, histaminic, observed for dexamethasone.[106] Clinically, unnecessary
serotoninergic and substance P/neurokinin receptors. high exposure to corticosteroids should be avoided due to
Aprepitant is a very potent neurokinin-1 receptor antag- the potential risk of adverse effects such as hyperglycaemia
onist used for the prevention of acute and delayed and increased susceptibility to infections. For these reasons,
chemotherapy-induced nausea and vomiting.[101,102] it is suggested that the oral doses of dexamethasone and

652 © 2018 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 71 (2019), pp. 643–673
Chara Litou et al. Pharmacokinetic interactions with GI drugs

methylprednisolone should be reduced by half when used Indeed, esomeprazole, a PPI, ranks among the top five
for the management of chemotherapy-induced nausea and most prescribed medications worldwide.[114] Of particular
vomiting concurrently with aprepitant.[106] The interaction concern for these drugs is their increasing OTC use. Despite
of aprepitant with warfarin is less clear.[107] In a study by the fact that gastric antisecretory agents or antacids are tol-
Takaki et al., a decrease in warfarin plasma levels was erated well, with a low overall frequency of adverse reac-
observed, but no significant interaction between warfarin tions,[115] their concurrent use with other medications can
and aprepitant was established. One possible reason for the have a great effect on drug absorption. If prescribed, identi-
lack of interaction could be the fact that the volunteers who fication of potential interactions by the prescribing physi-
took part in this clinical study were also receiving several cian and/or dispensing pharmacist is possible, but this
other chemotherapeutic agents. In any case, careful moni- control mechanism is largely lost if the drugs are obtained
toring of patients on chronic warfarin therapy is OTC or via e-pharmacies.
required.[103,108]
Serotonin plays an important role in various body func-
Proton pump inhibitors
tions. Most serotonin is synthesised in the GI tract, and it
affects various aspects of intestinal physiology. Multiple Proton-pump inhibitors are a group of substituted benz-
subtypes of 5-HT receptors exist on various types of cells, imidazole sulfoxide drugs with strong inhibitory effects on
such as smooth muscle and enterocytes, and agonists or gastric acid secretion from the parietal cells in the stom-
antagonists of 5-HT receptors are used in the treatment of ach. At present, six PPIs (dexlansoprazole, esomeprazole,
different gastrointestinal disorders.[21] 5-HT3 receptor lansoprazole, omeprazole, pantoprazole and rabeprazole)
antagonists, for example ondansetron and granisetron, have are available on the market.[116] PPIs are used in the treat-
been successfully used in the treatment of chemotherapy- ment of acid-related disorders and for the prevention of
induced nausea and vomiting. Recommendations, pub- gastrointestinal bleeding in patients receiving dual antipla-
lished by the American Society of Clinical Oncology telet therapy of clopidogrel and aspirin. Furthermore, they
(ASCO) for the use of the 5-HT3 receptor antagonists, do are used as a component of combination therapy for the
not distinguish among them with regard to their safety and eradication of H. pylori, because their properties enhance
efficacy. Nonetheless, these compounds differ signifi- the anti-H. pylori activity of the coadministered antibacte-
cantly in their pharmacokinetic properties and especially rials (clarithromycin and amoxicillin).[117] PPIs can affect
with respect to their potential to interact with CYP the absorption of the coadministered drugs to a great
enzymes.[109,110] Granisetron, for example, does not inhibit extent, mainly due to the increase in gastric pH. In a
any of the CYP enzymes which are commonly involved in recent study, the effect of 40 mg of pantoprazole adminis-
drug metabolism, whereas ondansetron inhibits both tered orally once per day for 4 days and 20 mg of the
CYP1A2 and CYP2D6 and can thus interact with various H2RA famotidine administered orally twice within 12 h,
concurrently used drugs. on the GI physiology of eight healthy male volunteers was
However, the interactions reported in the literature are investigated.[118] In both cases, the gastric pH differed sig-
not solely attributed to their enzyme inhibitory properties. nificantly in comparison to the control group (Figure 5).
Concomitant use of ondansetron with cyclophosphamide However, PPIs can also affect the pharmacokinetics of
resulted in reduced systemic exposure, probably due to coadministered drugs through other mechanisms,[119] and
increased systemic clearance.[111,112] In any case, there is a several excellent reviews have been written regarding the
need for more studies to increase knowledge about drug DDIs of PPIs.[120–122]
interactions of chemotherapeutic agents with commonly As already mentioned, gastric pH is an important param-
used antiemetics, as even a slight change in the pharma- eter that can affect absorption of drugs, especially these
cokinetic parameters or pharmacodynamics of the anti- which are poorly soluble weak bases. For example, Jaru-
cancer medication could jeopardise the effectiveness of ratanasirikul et al.,[123] investigated the effect of 40 mg oral
chemotherapy.[111] omeprazole on the pharmacokinetics of a single 200 mg
capsule of itraconazole in 11 healthy volunteers. Concomi-
tant use of omeprazole resulted in reduction of the mean
Gastric acid reducing agents and antacids
AUC and Cmax of itraconazole by 64% and 66%, respec-
Proton-pump inhibitors, H2-receptor antagonists (H2RAs) tively. No interaction due to omeprazole’s inhibition of
and antacids are widely used in the treatment of various CYP3A4 was reported.[123] On the other hand, Johnson
gastric acid-related disorders, such as peptic ulcers and gas- et al.,[124] investigated the effect of concomitant use of
troesophageal reflux disease. In fact, PPIs and H2RAs are 40 mg oral omeprazole with a 40 mg dose oral solution of
classified among the three most prescribed drug classes for itraconazole in 20 volunteers. It was reported that there was
the years 2011–2014 and the situation is similar today.[113] no statistically significant difference on the AUC, tmax and

© 2018 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 71 (2019), pp. 643–673 653
Pharmacokinetic interactions with GI drugs Chara Litou et al.

decreased by 41% and 47%, respectively.[130] Coadminis-


tration of omeprazole resulted in reduction to the systemic
exposure to both nelfinavir and its metabolite. In particu-
lar, the AUC of nelfinavir was decreased by 36%.[131]
Tomilo et al.,[132] reported a 94% and 91% decrease in
AUC and Cmax, respectively, of 400 mg oral atazanavir,
when coadministered with 60 mg lansoprazole in 10
healthy volunteers. The results were similar when omepra-
zole was coadministered.[133] However, the clinical impact
of this drug–drug interaction on the clinical effect of ataza-
navir is not clear.[134,135] It seems that coadministration of
PPIs with an atazanavir/ritonavir regimen does not affect
the ability of atazanavir to achieve the minimum plasma
concentration necessary for the virologic response, that is
the concomitant use of atazanavir/ritonavir regimen and
PPIs was not associated with higher virologic failure
Figure 5 pH in the stomach of fasted healthy adults as a function
of time, after administration of 240 ml table water into the antrum of
rate.[134] Nonetheless, further studies, in which both the
the stomach. Key: (From left to right boxes) White boxes, Phase 1 pharmacokinetic parameters and the clinical response rates
(control phase); Light pink boxes, Phase 2 (pantoprazole phase); Dark are simultaneously investigated, are needed to understand
blue boxes, Phase 3 (famotidine phase). Each box was constructed the interaction and its consequences more fully.
using 7–8 individual values.[118] [Colour figure can be viewed at wile In contrast to the results mentioned so far, in the study
yonlinelibrary.com] of Winston et al.,[136] coadministration of 40 mg oral
omeprazole with 1000 mg saquinavir (given orally as
Cmax with the coadministration of omeprazole.[124] The 1000 mg saquinavir/100 mg ritonavir combination)
results of these two clinical studies (one with a solid dosage resulted in an 82% increase in the mean AUC of saquinavir
form, one with itraconazole in solution) suggest that coad- in 18 healthy volunteers. The increase did not result in an
ministration of omeprazole and elevation of gastric pH increase in adverse effects. The authors commented that
affects the dissolution of itraconazole capsules rather than further work is necessary in order to understand the mech-
the permeability of itraconazole. The results regarding keto- anism of this DDI and to address whether the effects of
conazole are similar. In 1995, Chin et al., conducted a clini- omeprazole on saquinavir’s pharmacokinetics would be the
cal study with nine healthy volunteers, in which the effects same even in the absence of ritonavir. The authors also
of 60 mg oral omeprazole or an acidic beverage on the phar- raised the possibility of whether the increase could be the
macokinetics of orally administered 200 mg ketoconazole result of inhibition of transmembrane-transporters, such as
were investigated. Pretreatment with omeprazole resulted in P-gp or MRP by omeprazole.[136]
significantly lower AUC and Cmax and a prolongation of As for most of the antifungal and antiviral drugs, the
tmax.[125] Ketoconazole and itraconazole are both practically absorption of mycophenolate mofetil is impaired by con-
insoluble at pH > 4. Coadministration of PPIs with poorly comitant administration of PPIs. Kofler et al.,[137] mea-
soluble imidazole antifungal agents when given as capsules sured the levels of mycophenolic acid (active metabolite) in
or tablets is therefore not recommended.[126] Interestingly, 33 patients concurrently receiving 40 mg oral pantoprazole.
the elevated gastric pH does not affect the bioavailability of Cmax and AUC of mycophenolic acid were significantly
fluconazole tablets.[127] This lack of interaction is explained lower when patients were pretreated with pantoprazole.[137]
by the high solubility of fluconazole over the whole pH As anticipated, coadministration of pantoprazole with an
range of the GI tract. Thus, stomach acidity does not limit enteric coated formulation of mycophenolic acid had no
the dissolution rate of fluconazole or its absorption.[128,129] significant effect on its pharmacokinetics.[138]
The increase in the gastric pH caused by PPIs can also Apart from lowering the solubility of APIs in the stom-
greatly affect the bioavailability and effectiveness of ach, an increase in the gastric pH can jeopardise the
antiretroviral agents, depending on their pH/solubility pro- bioavailability of formulations with pH-dependent release.
files. Tappouni et al.,[130] conducted a clinical study with The effect of concomitant administration of esomeprazole
16 patients, in which the effect of omeprazole on indinavir on the bioavailability of risedronate sodium DR was evalu-
was evaluated. With pre-treatment and coadministration of ated in a clinical study involving 87 postmenopausal
20 mg oral omeprazole, the Cmax of indinavir decreased by women. The results showed that esomeprazole administra-
29% and the AUC by 34%, whereas at a higher dose of tion 1 h before dinner or 1 h before breakfast resulted in
40 mg omeprazole, the Cmax and AUC of indinavir 32% and 48% reduction in the bioavailability of risedronate

654 © 2018 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 71 (2019), pp. 643–673
Chara Litou et al. Pharmacokinetic interactions with GI drugs

sodium DR, respectively. In the report, it was suggested Last, but not least, there has been an increasing interest
that an increase in the gastric pH may compromise the in investigating the mechanism of drug interactions of PPIs
enteric coating of risedronate delayed release formulation, with clopidogrel. Clopidogrel is a prodrug that requires
thus resulting in release of risedronate sodium in the stom- activation via cytochrome P450 isozymes (CYP2C19,
ach, where it could convert to the less soluble free acid.[139] CYP3A4, CYP3A5) in order to transform to its pharmaco-
However, as it has been shown that PPIs (pantoprazole) logically active form. Therefore, inhibition of the cyto-
decrease buffer capacity as well as increase gastric pH,[118] a chrome isoenzymes, which are involved in the metabolic
premature release due to enteric coating failure appears pathway of clopidogrel, may reduce its antiplatelet activity
unlikely. and potentially increase the risk of thrombosis. In fact, in
A review of all the available clinical data from literature 2009 FDA published a warning note on the drug label of
describing the effect of the administration of various gas- Plavixâ (clopidogrel; Sanofi Clir SNC, Paris, France) and
tric acid reducing agents on the absorption and bioavail- continues to warn the public against concomitant use of
ability of coadministered weakly basic anticancer drugs clopidogrel and omeprazole. It should be noted that,
was published by Budha et al.[140] The authors attempted although studies have demonstrated that concomitant use
to correlate the physicochemical properties and pH-solu- of clopidogrel and PPIs, especially omeprazole, reduces the
bility profiles of the different anticancer drugs with the antiplatelet effect of clopidogrel, the mechanism behind
observed effect on the absorption caused by the elevation this interaction and the clinical importance (cardiovascular
of the gastric pH after the administration of the acid risk) has not yet been clearly established.[149–154]
reducing agents (PPIs, H2RAs and antacids). It was con-
cluded that the impact of the elevation of gastric pH is
H2 receptor antagonists
more prominent for the anticancer drugs which exhibit an
exponentially decreasing solubility in the pH range 1–4 The H2RAs are another drug class used to treat gastric acid-
and for which the maximum dose strength is not soluble related disorders. These compounds bind to histamine H2
in 250 ml of water. Elevation of gastric pH is expected to receptors on parietal cells and antagonise the action of his-
substantially decrease the dissolution rate of these drug tamine, which is the major transmitter for stimulation of
products, thus leading to incomplete dissolution of the acid secretion.[155] As with the PPIs, there are DDIs with
dose and impaired absorption. different classes of drugs and these are mainly attributed to
In 2013, Mitra and Kesisoglou described strategies to the elevation of the gastric pH (see Figure 5). For example,
minimise or avoid reduced absorption of weakly basic ketoconazole and itraconazole demonstrate impaired drug
drugs resulting from elevated gastric pH.[141] absorption when they are concomitantly used with H2RAs
The observed DDIs with PPIs occur not only because of as well as with PPIs. Piscitelli et al.,[156] investigated the
their elevation of gastric pH, but can also arise from other effect of 150 mg orally administered ranitidine on 400 mg
properties. It has been shown that concurrent administra- oral ketoconazole in six healthy volunteers. The decreased
tion of 10 mg of nifedipine with 20 mg of omeprazole for Cmax and AUC and bioavailability of ketoconazole in this
8 days (short-term treatment) resulted in an AUC increase study was attributed to the elevated gastric pH, which
of 26%, whereas no increase was observed after coadminis- resulted in a decreased and incomplete ketoconazole disso-
tration of a single 20 mg dose of omeprazole.[142] The lution.[156] The results were similar when the effect of cime-
authors hypothesise that the higher levels might be due to tidine on the absorption and pharmacokinetics of
inhibition of CYP3A4, but they note that this increase is ketoconazole was investigated.[121] Lim et al.,[157] investi-
not likely to have major clinical relevance, especially when gated the effect of famotidine on the absorption of flucona-
taking into account the intra- and inter-individual variabil- zole and itraconazole. Twenty healthy volunteers received
ity observed for nifedipine.[142] In contrast, in the study by orally 40 mg famotidine with 200 mg itraconazole or
Bliesath et al.,[143] coadministration of 20 mg of nifedipine 100 mg fluconazole. Coadministration of famotidine
with 40 mg of pantoprazole for 10 days had no effect on resulted in a 52.9% decrease in Cmax and a 51.1% decrease
the pharmacokinetics of nifedipine. This apparent discrep- in the AUC of itraconazole, but no difference was observed
ancy in DDI tendency might be due to the different CYP- in the pharmacokinetics of fluconazole.[157] This different
isoenzymes inhibitory properties of the two PPIs. It is behaviour of fluconazole had previously been observed by
believed that among all PPIs, omeprazole is the one which Blum et al.,[158] and can be explained by its much higher
has the greatest potential for drug interactions, as it has a solubility (see the section on ‘Proton pump inhibitors’).
high affinity for CYP2C19 and CYP3A4.[144–147] The situation is similar with antiretroviral medica-
Another example of a non-pH-related DDI with PPIs is tions.[159] Analogous to the PPIs/saquinavir interaction,
the delayed elimination of plasma methotrexate, indepen- coadministration of cimetidine resulted in increased expo-
dent of renal function.[148] sure to saquinavir.[136,160]

© 2018 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 71 (2019), pp. 643–673 655
Pharmacokinetic interactions with GI drugs Chara Litou et al.

Russell et al., investigated the effect of a single dose of treatment (administration of cimetidine started on the
40 mg of famotidine on the pharmacokinetics of the weak eighth day).[169] It was concluded that coadministration of
base dipyridamole in 11 elderly adults with normal gastric cimetidine resulted in decreased clearance of propranolol
acid secretion. After coadministration of famotidine, the and thus increased propranolol plasma levels at steady
Cmax and absorption constant (ka) of dipyridamole state. In a similar study, Reimann et al.[170] investigated
decreased significantly. The total AUC decreased by 37%, the effect of cimetidine (1000 mg daily, 1 day oral pre-
but this decrease was not found to be statistically signifi- treatment) and ranitidine (300 mg daily oral dose, 1 and
cant. The authors attributed the observed differences to 6 days pretreatment) on the steady-state propranolol
slower dissolution rate of dipyridamole tablets at elevated plasma levels (160 mg sustained-release capsule, once
gastric pH.[161] In other studies, coadministration of rani- daily) of five healthy volunteers. It was shown that 1-day
tidine with two weak bases, enoxacin and cefpodoxime, pretreatment with cimetidine resulted in elevated propra-
resulted in decreased bioavailability, which was again nolol plasma levels at steady state, while ranitidine pre-
attributed to decreased solubility in the gastric environ- treatment for 1 or 6 days did not affect significantly the
ment at elevated pH.[162,163] propranolol plasma levels at steady state. However, the
As with the PPIs, DDIs with H2RAs can occur not only authors stated that the elevated plasma levels of propra-
because of their elevation of gastric pH, but can also arise nolol observed after pretreatment with cimetidine did not
from their other properties. In particular, it has been shown lead to a clinically significant effect.[170] Again, the study
that, among the various H2RAs, cimetidine is the most was conducted in healthy volunteers and the clinical
potent inhibitor of the CYP450 enzymes. The inhibition is effects on patient populations could differ. Nonetheless, it
attributable to the imidazole ring in its structure and results should be noted that the companies are required by the
in changes in the metabolism of various coadministered regulatory authorities to inform the patients that there is a
drugs.[164] In cases where a clinical significant interaction is potentially clinically significant DDI of cimetidine and
suspected, other H2RAs (e.g. ranitidine, famotidine) are propranolol in the patient information leaflets.[171]
preferred over cimetidine.[165,166] Among the various meta- It is obvious that there are many interactions of PPIs and
bolic interactions that have been reported after coadminis- H2RAs with other concomitantly used drugs, especially
tration of cimetidine,[164] the metabolic interactions poorly soluble weak bases, and that their use should be
observed with warfarin and propranolol have been most monitored, particularly in cases where the DDI is well-
intensively studied and the clinical significance of these established. Besides the elevation of gastric pH and the
interactions has also been evaluated. Toon et al., investi- interactions with metabolic pathways, it should be noted
gated the effect of a 9-day short treatment of cimetidine that PPIs and H2RAs can also affect other aspects of the
and ranitidine (800 mg oral dose daily and 300 mg oral physiology in the GI tract. Recent data in the literature sug-
dose daily respectively) on the pharmacokinetics of 25 mg gest that administration of PPIs or H2RAs can be accompa-
of racemic warfarin, administered orally starting on the nied by reduced buffer capacity, chloride ion
fourth day of cimetidine treatment and continuing for the concentration, osmolality and surface tension in stomach
next 5 days, in nine healthy volunteers.[167] The prothrom- and an increase in the pH of the upper small intestine of up
bin time and Factor VII clotting time were also evaluated. to 0.7 units, an increase that would be especially relevant
While ranitidine had no effect on the pharmacokinetics of for compounds (basic or acidic) with pKas between 6 and
either of the two enantiomers of warfarin, cimetidine sig- 7.[118] Carefully designed DDI studies, in terms of dosing
nificantly increased the elimination half-life and decreased and duration of treatment, are needed in order to accu-
the clearance of the (R)-enantiomer of warfarin. In con- rately determine the effect of H2RAs or PPIs on the phar-
trast, the pharmacokinetics of the (S)-enantiomer of war- macokinetics of coadministered drugs and investigate the
farin were not affected by coadministration of cimetidine. clinical consequences of these interactions.
Nonetheless, coadministration of either ranitidine or cime-
tidine did not result in a clinically significant difference in
Antacids
terms of the anticoagulation effect of warfarin.[167] These
results were further confirmed by a later study from Niopas The term ‘antacids’ describe a category of salts, formulated
et al.[168] It should be noted, however, that both studies as the combination of polyvalent cations such as calcium,
were conducted in healthy volunteers, and therefore, the aluminium or magnesium with a base, such as hydroxide,
clinical effects on patient populations could differ. trisilicate or carbonate. Aluminium hydroxide alone, or in
The effect of a daily oral dose of 1000 mg cimetidine on combination with magnesium hydroxide, is the main ingre-
the steady-state plasma levels of propranolol, administered dient of many antacid products. Owing to the introduction
as a 160-mg sustained-release formulation daily, was eval- of PPIs and H2RAs, which are more potent drugs and can
uated in seven healthy volunteers during a 13-day be used for a wide variety of gastrointestinal disorders,

656 © 2018 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 71 (2019), pp. 643–673
Chara Litou et al. Pharmacokinetic interactions with GI drugs

antacids are now mainly marketed as OTC medications. antacids with NSAIDs are not clearly established and no
However, the concomitant use of antacids with other drugs general recommendations can be made for this drug cate-
can significantly affect their absorption or even their thera- gory. However, there are studies indicating that coadminis-
peutic effect. Considering the fact that the use of OTC anta- tration with antacids containing magnesium hydroxide or
cids is widespread, there is a particular need for sodium bicarbonate could enhance the rate and possibly
appropriate information for patients, doctors and pharma- the extent of absorption of some NSAIDs, for example
cists. Besides interactions associated with increased pH, the ibuprofen, tolfenamic and mefenamic acid, diflunisal and
major DDIs with antacids involve chelation reactions. Vari- naproxen.[186–189] This has been attributed to the fact that
ous categories of drugs, such as quercetin, catechol deriva- magnesium hydroxide, in addition to increasing gastric pH,
tives and tetracyclines, are known to form drug/metal also accelerates gastric emptying. Such effects have not been
chelates.[172–174] Fluoroquinolones also interact with multi- observed for aluminium hydroxide, which in contrast to
valent cations and this interaction can lead to reduced magnesium hydroxide prolongs gastric emptying[190]
antimicrobial activity.[175] There have been many further studies investigating the
Deppermann et al.,[176] and Garty et al.,[177] investi- interactions of antacids with APIs from various drug
gated the effect of H2RAs or antacids (mixture of alu- classes, including corticosteroids, cardiovascular agents and
minium hydroxide and magnesium hydroxide) on the oral antidiabetic agents. However, it has not been possible to
absorption of various tetracycline antibiotics. The antacids make any generalisations about the observed interactions.
resulted in reduction of the oral bioavailability of tetracy- Furthermore, in some cases, there is no evidence that dif-
clines by 80% or more, whereas coadministration of the ferences in pharmacokinetic parameters translate into clini-
H2RAs did not affect the pharmacokinetic parameters of cally significant differences.[190]
tetracyclines.[176,177] For this reason, it was concluded that
chelation rather than elevation of gastric pH is the proba-
Probiotics
ble mechanism of this DDI. The complexes that are
formed by chelation are insoluble and therefore they pre- It is well-known that the intestinal microflora plays a key
cipitate, preventing absorption. The results are similar role in physiological, metabolic, immunological and nutri-
with coadministration of antacids and fluoroquinolones. tional processes in the human body. For this reason, there
Aluminium ions form a stable and insoluble complex with is currently great interest in influencing the composition of
quinolones, thus preventing their intestinal absorption the microflora and its activity using probiotics for both the
and reducing their bioavailability.[178,179] By contrast, con- prevention and treatment of various diseases.[191] Accord-
comitant administration of an H2RA did not have a signif- ing to WHO, probiotics are ‘live microorganisms which,
icant effect on the AUC of ciprofloxacin.[176] As the when administered in adequate amounts, confer a health
formation of the chelate complex is the limiting factor to benefit on the host’.[192] There are several clinical studies
absorption of quinolone antibiotics, many studies have that have illustrated their beneficial effects on gastrointesti-
been conducted in order to establish an optimal interval nal disorders such as diarrhoea and irritable bowel syn-
of antacid dosing before or after the administration of the drome. The gram-negative bacterium Escherichia coli Nissle
antimicrobial agents. With regard to fluoroquinolones, it 1917, for example, has been used since 1920 for the treat-
has been concluded that administration of antacids 4 h ment or prevention of irritable bowel syndrome, chronic
earlier or 2 h later than the administration of the antibi- constipation, non-ulcer dyspepsia and other gastrointesti-
otic would circumvent the interaction.[162,180–183] nal disorders.[193] The mechanism of action of the probi-
As with the PPIs and H2RAs, the elevation of gastric pH otics is not yet fully understood. It seems that they may
that is observed after administration of antacids could also modulate the intestinal epithelial barrier and transport
impact the dissolution or oral solid formulations and across it, noting that in inflammatory bowel diseases
change their pharmacokinetics. Indeed, coadministration (IBDs), for example ulcerative colitis and Crohn’s disease,
of itraconazole with antacids resulted in decreased the barrier properties of the epithelium are compromised
AUC.[184] However, in a pilot study by Brass et al.[185] due to secreted cytokines and/or medication.[194]
(n = 4), the absorption of ketoconazole was not signifi- Despite the wealth of evidence regarding their advanta-
cantly decreased. geous and well-tolerated use, the literature on interactions
The interaction of antacids and NSAIDs is also an inter- between concomitantly administered probiotics and drugs
esting case. NSAIDs are among the most popular OTC and with respect to drug pharmacokinetics is mainly limited to
frequently prescribed medications for acute or short-term animal experiments. In the study of Mikov et al.,[195] the
pain and chronic inflammatory diseases. As NSAIDs cause effect of coadministration of probiotics (oral 2 g dose of
dyspepsia and damage in the upper gastrointestinal freeze dried powder of a mixture of the strains Lactobacil-
mucosa, they are often given with antacids. Interactions of lus acidophilus L10, Bifidobacterium lactis B94 and

© 2018 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 71 (2019), pp. 643–673 657
Pharmacokinetic interactions with GI drugs Chara Litou et al.

Streptococcus salivarius K12 every 12 h for 3 days) on sul- Sullivan et al.[202] reviewed the effect of various antibi-
fasalazine metabolism (sulfasalazine administered as an oral otics on the abundance of bacterial types and species. Dif-
dose of 100 mg/kg dissolved in saline via gavage 6 h after ferences in the composition of the microbiota could alter
completing the 3 day treatment with probiotics) in the rat the composition of colonic fluids and permeability of the
gut lumen was investigated. The authors showed that gut wall as well as the abundance of bacterial enzymes.
administration of probiotics significantly increased the con- Colonic bacteria are involved in the cleavage of dietary
version of sulfasalazine to sulfapyridine and 5-aminosa- fibres to oligosaccharides and monosaccharides and their
licylic acid by increasing azoreductase activity. This could further fermentation to SCFAs such as acetate, propionate
possibly enhance sulfasalazine therapy, which would be and butyrate.[205] Patients treated with antibiotics showed a
important in patients with reduced gut microflora, subse- decreased colonic carbohydrate fermentation and conse-
quent to antibiotic therapy, or in severe diarrhoea.[195] Lee quently lower faecal concentrations of SCFAs.[206–210] In
et al.,[196] confirmed an increase in azoreductase activity in other studies, it was shown that SCFAs stimulate ileal and
ex vivo colon rat fluids. However, no differences were found colonic motility.[211–213] The inhibition of gastric emptying
in the pharmacokinetic parameters of sulfasalazine and sul- by nutrients that reach the ileo-colonic junction, the so-
fapyridine.[196] Kunes et al.,[197] investigated the effect of called ileocolonic brake, is also associated with SCFAs.[214]
E. coli Nissle 1917 probiotic medication on the absorption But GI transit times can also be affected by certain antibi-
kinetics of 5-aminosalicylic acid in rats. The results showed otics through other mechanisms: for example, erythromycin
that there was no difference in the pharmacokinetics of 5- accelerates gastric emptying ( 25% to 77%) by acting as a
aminosalicylic acid and that E. coli Nissle 1917 medication motilin agonist, while prolonging small intestinal transit
did not affect the absorption of 5-aminosalicylic acid.[197] time (+20% to +45%) for liquids and solids in healthy vol-
Al Salami et al.,[198] investigated the effect of a mixture of unteers and patients.[215–220] For example, when ery-
three probiotics in diabetic rats on gliclazide pharmacoki- thromycin was coadministered with a controlled-release
netics. They observed that gliclazide’s absorption and formulation of pregabalin, designed to remain for a pro-
bioavailability were reduced in healthy rats. The authors longed time in the stomach, in 18 healthy subjects there was
attributed this change to several possible causes, most of a reduction of AUC and Cmax by 17% and 13%, respectively,
which had to do with intestinal efflux drug trans- due to erythromycin’s prokinetic action.[221] As the prega-
porters.[198] Saksena et al.,[199] reported that Lactobacilli or balin exposure was still in the range calculated for patients
their soluble factors significantly enhanced P-gp expression receiving an immediate release formulation of pregabalin,
and function under normal and inflammatory conditions the interaction was deemed not to be clinically relevant.
in mice. Finally, Matuskova et al.,[200] investigated the If bacterial enzymes are involved in the biotransforma-
effect of administration of E. coli Nissle 1917 on amio- tion of a drug, the intake of antibiotics can affect its meta-
darone absorption in rats. This resulted in 43% increase in bolism by changing the composition of the microbiota and
the AUC of amiodarone. Interestingly, this effect was not thus altering the bacterial enzyme activity.[222,223] At least
observed when E. coli Nissle 1917 was replaced by a refer- 30 commercially available drugs have been reported to be
ence non-probiotic E. coli strain, suggesting that the metabolised by bacterial enzymes in the gastrointestinal
increase in AUC of amiodarone was due to the administra- tract.[222] The serum concentrations of digoxin, which is
tion of the probiotic.[200] partly metabolised by gut microbiota, increased twofold
Clearly, studies in humans are needed in order to investi- after administration of erythromycin or tetracycline for
gate whether these results can be extrapolated well to 5 days in four healthy volunteers.[224] In another report,
patients with altered intestinal microflora. toxic digoxin plasma levels were observed in a patient after
cotreatment with erythromycin, possibly due to the inhibi-
tion of Eubacterium lentum which converts digoxin to its
Antibiotics used for gastrointestinal
reduced derivatives.[225] Incubation of flucytosine with fae-
infections
cal specimens of neutropenic patients before and after
Antibiotics aim to attack targets specific to bacterial organ- treatment with antibiotics (ciprofloxacin, penicillin, cotri-
isms such as bacterial cell walls, bacterial cell membranes, moxazole) and antimycotics (amphotericin B, fluconazole,
bacterial metabolism or replication, in order to avoid dam- nystatin) indicated that the transformation of flucytosine
age to human cells. However, antibiotics are not 100% to its active metabolite, fluorouracil, was reduced.[226] Sim-
selective for bacteria that are pathogenic for the host organ- ilarly, concomitant administration with ampicillin (250 mg
ism. As a result, the GI microbiota is frequently disturbed four times daily for 5 days) with sulfasalazine (single dose
after treatment with antibiotics.[201,202] In fact, depending 2 g) led to a decrease in the AUC of sulfapyridine by 35%
on the antibiotic, 5–25% of patients treated experience in five healthy subjects suggesting a decrease in azoreduc-
diarrhoea.[203,204] tase activity and prodrug activation.[227]

658 © 2018 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 71 (2019), pp. 643–673
Chara Litou et al. Pharmacokinetic interactions with GI drugs

An altered colonic microflora could also adversely affect observed.[237] Concomitant oral administration of enoxacin
the drug release from colon-targeting formulations coated (400 mg twice daily for 6 days) with theophylline (250 mg
with water-insoluble polysaccharides.[228] As polysaccha- twice daily for 11 days) resulted in a reduction in total clear-
rides such as guar gum, pectin and chitosan are degraded ance of theophylline by 74% in six healthy subjects,[238] while
by bacterial enzymes in the colon, release of the drug relies ciprofloxacin (500 mg twice daily for two and a half days)
on the abundance and activity of the polysaccharide-speci- reduced theophylline’s total clearance by 19% after a single
fic bacterial enzymes. Samples (faecal slurries) from volun- oral dose of theophylline syrup (3.4 mg/kg) in nine healthy
teers treated with antibiotics within the last 3 months subjects.[239] In contrast, concomitant administration of nor-
should be excluded from the evaluation of such formula- floxacin (400 mg twice daily for 4 days) with theophylline
tions in in-vitro dissolution tests.[228] (200 mg three times daily for 4 days) had no significant
The microbiota is also involved in the modification of effect on theophylline’s total clearance in 10 healthy sub-
primary bile acids to secondary bile acids, such as deoxy- jects.[240] For more detailed information, the reader is
cholic acid and lithocholic acid, via microbial 7a-dehy- referred to several review articles.[233,237,241]
droxylase and in the deconjugation of conjugated bile
acids.[229] Unconjugated bile acids are less likely to be reab-
Anti-inflammatory drugs for inflammatory
sorbed in the terminal ileum and therefore, bacterial action
bowel disease
promotes the excretion of bile acids.[230] Thus, antibiotic
treatment may cause changes in the bile acid pool. Indeed, Anti-inflammatory agents, such as aminosalicylates and
treatment with oral vancomycin decreased faecal levels of corticosteroids, are the most commonly used drugs in IBD.
secondary bile acids and increased faecal levels of primary Treatment with aminosalicylates includes a range of pro-
bile acids in healthy volunteers (n = 10). By contrast, treat- drugs (sulfasalazine, olsalazine, balsalazine) or modified
ment with oral amoxicillin showed no such effect.[231] It release formulations to deliver aminosalicylates to their tar-
has also been hypothesised that antibiotic-induced differ- get site in the intestine. If remission cannot be achieved
ences in the bile acid composition could affect the solubili- with aminosalicylates, the next treatment option consists of
sation of lipophilic drugs. However, a recent study different corticosteroids ranging from locally acting drugs
evaluating the differences in the solubilisation capacity of (budesonide) to systemic acting ones (hydrocortisone,
primary and secondary bile acids for nine poorly water- prednisolone, dexamethasone).
soluble drugs revealed at most minor differences between Aminosalicylates have shown to alter the GI physiology.
conjugated and unconjugated bile acids. Only dehydroxyla- In terms of GI transit time, olsalazine accelerated transit,
tion at C-7 improved drug solubilisation significantly for with a mean gastric emptying time of 45.3  24.2 min vs
the compounds investigated.[232] 67.3  33.1 min, a mouth to caecum transit time of
With regard to DDIs at the level of metabolism, the effect 242  41 min vs 325  33 min and whole gut transit time
of antibiotics on metabolic enzymes is often specific to the of 37.8  17.8 h vs 60.5  26 h in six patients with ulcera-
antibiotic agent. Macrolide antibiotics interact with sub- tive colitis whereas intake of sulfasalazine had no effect in
strates metabolised by CYP3A4 (i.e. carbamazepine, terfe- six healthy subjects (measured by scintigraphy of a solid
nadine, cyclosporine) depending on the macrolide’s radio-labelled meal or hydrogen breath test).[242–244] The
specific affinity for CYP3A4. The interaction potential can authors commented that this may be the result of a direct
be high (troleandomycin, erythromycin), moderate (clar- action of olsalazine on contractile activity in the small
ithromycin, roxithromycin) or low (azithromycin).[233] For intestine, inducing hypersecretion or decreasing fluid
example, concomitant administration of erythromycin absorption.[243]
(500 mg three times daily for 7 days) with midazolam (sin- With respect to luminal pH, treatment with sulfasalazine
gle dose 15 mg) resulted in a fourfold increase of the AUC in patients with ulcerative colitis in remission resulted in a
of midazolam in 15 healthy subjects.[234] Similarly, when decrease in colonic pH to 4.90  1.3 compared to treat-
administered with clarithromycin (500 mg twice daily for ment with Asacolâ (mesalazine) with a colonic pH of
7 days), the bioavailability of midazolam (single dose 5.52  1.13 or Dipentumâ (olsalazine) with a pH of
4 mg) was increased 2.4-fold in 16 healthy subjects.[235] 5.51  0.37.[245] Nugent et al.[246] postulated that reduced
But, after pretreatment with azathioprine (500 mg daily for colonic pH may impair drug release from delayed-release
3 days), no significant effect on the pharmacokinetics of formulations targeting the terminal ileum/colon (trigger
midazolam (single dose 15 mg) was observed in 12 healthy pH for release is >6–7) or alter bacterial enzyme activity.
subjects.[236] Regarding permeability, jejunal perfusion studies showed
For the fluoroquinolones, depending on the fluoro- a decreased absorption of water, sodium, potassium and
quinolone’s specific affinity for CYP1A2, interactions with chloride in the presence of olsalazine or sulfasalazine.[247]
CYP1A2 substrates (i.e. clozapine, theophylline) have been In ileal perfusion studies, reduced absorption of water and

© 2018 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 71 (2019), pp. 643–673 659
Pharmacokinetic interactions with GI drugs Chara Litou et al.

glucose was observed, when olsalazine was present, which After treatment with corticosteroids, the phospholipid
in turn could explain the higher volume of ileostomy fluid mucus layer can be fluidised, resulting in a thinner mucus
observed after oral administration of this drug.[247,248] By barrier.[262] Impairment of membrane integrity can cause
contrast, no changes in absorption or volume of fluids was side effects such as gastrointestinal bleeding and bowel perfo-
observed in ileal perfusion studies in the presence of sul- ration.[263] The corticosteroids can also affect active trans-
fasalazine.[247] With regard to specific uptake mechanisms, port mechanisms such as bile salt reuptake and
sulfasalazine reduced the uptake of folic acid and exo-transport. Treatment with budesonide results in upregu-
methotrexate by folate transporters in biopsy specimens lation of the apical sodium-dependent bile acid transporter
taken from the duodenojejunal region while olsalazine only in the terminal ileum, which enhances bile acid absorption
decreased folic acid uptake.[249] In an intervention study, in both healthy controls and patients with Crohn’s dis-
sulfasalazine treatment was discontinued in rheumatoid ease.[264,265] Consequently, lower luminal bile salt concentra-
arthritis patients who had previously received a combina- tions may impede solubilisation and absorption of lipophilic
tion of sulfasalazine and methotrexate. The discontinuation poorly soluble drugs from the ileum.[266] In terms of trans-
of sulfasalazine resulted in a more than twofold increase of porters, budesonide and prednisone are substrates of the
methotrexate serum concentrations, in line with the ability efflux transporter P-glycoprotein.[267] However, it is unclear
of sulfasalazine to compete with methotrexate for the folic whether these alterations result in clinically significant DDIs.
acid transporter.[250] The main elimination pathway of corticosteroids is the
After treatment with sulfasalazine the faecal microbiota metabolism by intestinal and hepatic CYP3A4 which is
of patients with rheumatoid arthritis was richer in Bacillus, especially important for high-clearance corticosteroids such
whereas decreased numbers of aerobic bacteria, E. coli, as budesonide and prednisone.[268] Coadministration of
Clostridium perfringens and Bacteroides were prednisone with metronidazole in six patients with Crohn’s
observed.[251–253] Treatment with mesalazine resulted in a disease reduced the bioavailability of metronidazole by
decreased diversity of the intestinal microbiota and also 31%, most likely attributed to the induction of liver
reduced the quantity of faecal bacteria in patients with diar- enzymes responsible for metabolising metronidazole.[269]
rhoea-predominant irritable bowel syndrome.[254,255] These Likewise, cotreatment with prednisone resulted in
changes in colonic bacteria may have ramifications for decreased serum concentrations of salicylates in a 11-year-
drugs like digoxin, which are partly metabolised by bacte- old child with juvenile rheumatoid arthritis due to the
rial enzymes (see the section on ‘Antibiotics’).[256–258] induction of salicylate clearance by prednisone.[270] On the
With regard to DDIs, pretreatment with sulfasalazine other hand, drugs inhibiting CYP3A4 in the intestinal wall
(500 mg for 6 days) in 10 healthy subjects decreased the and liver such as ketoconazole, itraconazole, clarithromycin
AUC of digoxin by 25% after being administered as oral and HIV-protease inhibitors reduce the metabolism of cor-
solution (dose 0.5 mg).[256] The mechanism of the interac- ticosteroids and increase their bioavailability.[271–274]
tion is not yet understood. Differences in bioavailability
could possibly be attributed to a direct action of sul-
Immunosuppressive agents for
fasalazine on the intestinal mucosa or induced differences
inflammatory bowel disease
in the gut microbiota enhancing digoxin metabolism. For a
patient on concomitant treatment with cyclosporin Immunosuppressive agents are frequently used in gastroen-
(480 mg daily) and sulfasalazine (1.5 g daily), increased terology for the treatment of IBD, autoimmune hepatitis,
plasma concentrations of cyclosporine were observed autoimmune pancreatitis, sclerosing cholangitis and in the
5 days after the treatment of sulfasalazine was stopped, post-transplantation setting.[275] In particular, in IBD, ther-
making it necessary to reduce the dose of cyclosporine by apy with immunosuppressive agents has gained in impor-
60%.[259] While the interaction is not yet understood, an tance over the last few years.[276] Immunosuppressive
induction of metabolic enzymes is plausible considering the agents can be classified in immunomodulators (e.g. thiop-
time course of the observation. For 6-mercaptopurine (50– urines (6-mercaptopurine, azathioprine), methotrexate,
75 mg), a metabolic interaction was observed with con- tacrolimus, sirolimus, everolimus, cyclosporine A) and bio-
comitantly administered olsalazine (1000–1750 mg) in a logics (e.g. monoclonal antibodies: infliximab, adali-
patient with Crohn’s disease, resulting in bone marrow mumab, vedolizumab, golimumab).[276] Depending on the
suppression and requiring dose reduction of 6-mercapto- specific immunosuppressive agent, gastrointestinal transit
purine.[260] This interaction may be caused by the inhibi- time, bile flow and/or permeability can be altered, which
tion of thiopurine methyltransferase, which is responsible could further affect drug product performance of coadmin-
for 6-mercaptopurine metabolism; inhibition of this istered drugs.
enzyme by aminosalicylates has been demonstrated in in- Regarding transit time, gastric emptying time (as mea-
vitro enzyme kinetic studies.[261] sured with magnetic markers after a standardised meal

660 © 2018 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 71 (2019), pp. 643–673
Chara Litou et al. Pharmacokinetic interactions with GI drugs

using Alternating Current Biosusceptometry) was with statins or ezetimibe, and in the treatment of gastroin-
decreased in patients treated with tacrolimus after kidney testinal diseases.[302] Cholestyramine is indicated for diar-
transplant (47  34 min) compared to healthy subjects rhoea associated with Crohn’s disease, ileal resection,
(176  42 min) or patients treated with cyclosporine A vagotomy, diabetes, diabetic vagal neuropathy and radia-
(195  42 min).[277] tion.[303] While colesevelam is not licensed for the treat-
In terms of drug absorption, immunosuppressants can ment of bile acid malabsorption, several clinical trials have
result in increased permeability on the one hand, but demonstrated positive outcomes which has provoked its
decreased surface area on the other hand. Intestinal perme- off-label use in this indication.[304–306]
ability was increased (75% of median value; indicated by an Bile acid sequestrants are positively charged ion-
increased lactulose/L-rhamnose excretion ratio) in liver exchange resins which bind bile acids in the intestine to
graft recipients treated with tacrolimus (n = 12) compared form insoluble complexes and as a consequence reduce the
to healthy subjects (n = 9) and by 48% compared to bile acid pool.[303] As a result of decreased luminal bile acid
untreated liver transplant patients (n = 5).[278] Only the concentrations, BAS are expected to interfere with the
permeability via the transcellular pathway seems to be bioavailability of lipophilic, low-soluble compounds by
increased by tacrolimus, as indicated by an increased lactu- impeding their solubilisation. For several drugs, such as
lose/L-rhamnose ratio (+160%) and unchanged excretion rifaximin[307] and troglitazone,[308] the presence of bile
of lactulose in treated orthotopic liver transplantation acids was shown to increase drug solubility, and therefore,
patients.[278,279] their absorption may be impeded by cotherapy with BAS.
Another side effect of immunosuppressive therapy, espe- The positive charge of BAS leads to a high affinity for
cially with methotrexate (including low-dose therapy) is GI deprotonated acidic drugs in the intestine. Binding of these
mucositis resulting in the loss of villi in the duodenum, anions increases the excretion and impedes the absorption
crypts in the colon and enterocytes.[280–284] Oral mucositis of acidic coadministered drugs. Drugs that are known to be
is a side effect of azathioprine therapy.[285] In patients with affected by this mechanism are furosemide,[309] war-
oral mucositis, bupivacaine absorption from lozenges was farin,[310] phenprocoumon,[311,312] sulindac,[313] cerivas-
increased and a trend to higher fentanyl absorption admin- tatin,[314] levothyroxine,[315] glipizide,[316] mycophenolic
istered with a sublingual spray was observed but did not acid,[317] folic acid[318] and valproate.[319] The binding
reach statistical significance.[286,287] The effect may be due affinity for coadministered drugs can vary among the dif-
to impairment of the barrier function of the mucosa. ferent BAS, for example, cholestyramine, which has a high
In terms of transporter systems and metabolism, affinity for hydrophobic compounds,[302,320] decreased
immunosuppressants (cyclosporine A, tacrolimus, everoli- ibuprofen and diclofenac absorption to a higher extent than
mus and sirolimus) are substrates of P-glycoprotein and colestipol; while colesevelam has a favourable DDI-profile
CYP3A4.[288–290] As a result, various drug interactions with compared to other BAS.[321–323]
P-gp substrates such as aliskiren and anthracyclines have High-molecular lipophilic drugs are typical substrates for
been reported for cyclosporine A.[291–293] Additionally, enterohepatic recirculation.[324] By binding drugs or drug
concomitant administration of inhibitors (e.g. azole anti- metabolites that undergo enterohepatic recirculation, BAS
fungal drugs, macrolide antibiotics) and inducers (e.g. anti- can enhance drug elimination of the victim drug even if the
convulsants, rifampicin) of CYP3A4 can modify administration was not concomitant. Drugs affected by this
therapeutic response and toxicity of the above-mentioned mechanism include oral anticoagulants,[310–312] cardiac gly-
immunosuppressants.[294–296] Methotrexate intramuscular cosides[325] and mycophenolate mofetil.[317] It is difficult to
or subcutaneous cotreatment in patients with Crohn’s dis- predict which drugs that undergo enterohepatic recircula-
ease or oral cotreatment in patients with rheumatoid tion will be affected by BAS, as various factors such as
arthritis resulted in increased infliximab concentrations, polarity, ionisation properties and metabolism by liver and
most likely due to a decrease in the development of inflix- microbiota all influence biliary excretion.[326] Prolonging
imab antibodies.[297,298] Coadministration of azathioprine the interval between administration of BAS and comedica-
in patients treated with warfarin resulted in higher warfarin tion often reduces the potential for drug interactions and
doses needed to reach therapeutic anticoagulant effects but must be adapted for extended-release formulations.
the mechanism of the interaction is unclear.[299–301] Bile acid sequestrants can also affect GI transit time:
cholestyramine prolonged the transit time in the transverse
colon by up to 8 h in 13 patients with idiopathic bile acid
Bile acid sequestrants
diarrhoea (as measured with radiopaque markers), while
Bile acid sequestrants (BAS) such as cholestyramine, cole- total colonic transit was not altered.[327] After concomitant
sevelam and colestipol are used for the treatment of pri- administration of a sustained-release formulation of vera-
mary hyperlipidaemia, as monotherapy or in combination pamil (dose 240 mg) with colesevelam (dose 4.5 g), a

© 2018 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 71 (2019), pp. 643–673 661
Pharmacokinetic interactions with GI drugs Chara Litou et al.

reduction in AUC of 11% and decreased plasma levels of influence of DDIs on them will be necessary to tease out
verapamil were observed in 31 healthy subjects.[328] This the clinical implications of DDIs.
interaction was deemed not to be clinically relevant.[328] However, the number of studies that can be conducted
An overview of DDIs of bile acid sequestrants and their to test for potentially clinically relevant DDIs is limited,
mechanism is given in Table 4. due to both ethical and cost-related issues. So there is a
need for innovative evaluation methods to address knowl-
edge gaps and provide key information on safe and effective
Conclusions and future perspectives
drug use.[329] In the last 10 years, there has been an increas-
Gastrointestinal events and conditions play a key role in the ing use of Physiologically Based Pharmacokinetic (PBPK)
bioavailability of an orally administered drug and its thera- modelling and simulation at different stages of drug devel-
peutic action. Concomitant use of various medications can opment.[330] To date, PBPK modelling and simulation has
affect the absorption and the pharmacokinetics of the been mostly used for predicting enzyme interactions which,
administered drugs and therefore their performance. As as mentioned in this article, can also occur with concomi-
presented in this review article, various interactions tant administration of GI drugs.[331–336] PBPK modelling is
between drugs used to treat gastrointestinal diseases and gaining acceptance at the various regulatory agencies as a
coadministered drugs have been identified. These interac- tool to qualitatively and quantitatively predict DDIs and, in
tions are of particular concern, as GI drugs are commonly some cases, the simulation results may even be used to sup-
prescribed and many of them are also available OTC. Pre- port labelling, depending on the clinical importance of the
scribing physicians and pharmacists need to be aware of interaction.[8]
and monitor these potential interactions. Furthermore, One of the advantages of PBPK modelling is that it is
information involving interactions with GI drugs should be able to account for both formulation characteristics and
made available not only to clinical practitioners, but also to physiological parameters. As such, it can be used to help
patients, in order to prevent the appearance of adverse define a ‘safe space’ by identifying the range of dosing con-
effects, on the one hand, and failure of treatment on the ditions under which the pharmacokinetic parameters will
other hand. not be significantly affected by changes in the release prop-
It should be noted, however, that despite the large num- erties of the dosage form. This approach, which is some-
ber of DDI studies with GI drugs reported in literature, times referred to as ‘virtual bioequivalence’, has already
most studies have only investigated the effects of short- been used to explore whether bioequivalence decisions
term treatment and little is known about the ramifications based on clinical trials in healthy adults can be extrapolated
of long-term administration on DDIs. Furthermore, most to special populations, such as the hypochlorhydric or
DDI studies have been conducted in healthy volunteers and achlorhydric population, in whom the gastrointestinal
may not necessarily reflect the degree of interaction in physiology differs from that of healthy adults.[337–339]
patients. As most of the DDIs have been based on changes The same approach could be extended to predict preab-
in pharmacokinetics, it is also not clear in all cases whether sorptive DDIs with GI drugs, as these are intended to mod-
the DDI has any ramifications for the therapeutic effect. ify gastrointestinal physiology. First attempts have already
Indeed, some studies have suggested that even quite signifi- been made for acid reducing agents, with results from in-
cant changes in pharmacokinetics do not always lead to a vitro dissolution experiments, which are tailored to mimic
change in the clinical response. More work on pharmacoki- the changes in the upper gastrointestinal tract after the
netics/pharmacodynamics (PK/PD) relationships and the administration of these drugs, combined with PBPK

Table 4 Drug–drug interactions with concomitant administration of bile acid sequestrants

Implications for comedication Associated risk for comedication Reported interactions

Binding of weakly acidic drugs ↓ Bioavailability of coadministered Furosemide[309] warfarin,[310] phenprocoumon,[311,312] sulindac,[313]
drug cerivastatin,[314] levothyroxine,[315] glipizide,[316] mycophenolic acid,[317]
folic acid,[318] valproate[319]
Disruption of enterohepatic ↑ Excretion of coadministered Anticoagulants,[310–312] cardiac glycosides,[325] mycophenolate mofetil[317]
recirculation of drugs drug
Possible impact on ↓↑ Time available at Sustained-release formulation of verapamil[328]a
gastrointestinal transit time gastrointestinal absorption site,
effect on tmax
Reduced concentrations of bile ↓ Absorption of low-soluble
acids for drug solubilisation compounds
a
Not clinically significant due to high variability in the pharmacokinetics of verapamil.

662 © 2018 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 71 (2019), pp. 643–673
Chara Litou et al. Pharmacokinetic interactions with GI drugs

models for healthy adults.[337,338,340] This approach should whether these are likely to be clinically significant, in a wide
be broadened to encompass other classes of GI drugs. Pos- variety of populations and dosing conditions.
sible future steps include tailoring dissolution tests and
PBPK models to the physiological conditions observed in
Declaration
special populations, thus allowing for predictions of the in
vivo performance of drug products in special populations
Acknowledgement
(paediatrics, geriatrics, ethnic groups, the obese, hepatically
impaired, etc.) who concomitantly receive GI drugs. This This work was supported by the European Union’s Hori-
approach will provide the way forward to predicting phar- zon 2020 Research and Innovation Programme under grant
macokinetic differences resulting from these combinations agreement No 674909 (PEARRL).
and, especially when coupled with PK/PD relationships,

References gulatoryInformation/Guidances/defa 16. Longo WE, Vernava AM. Prokinetic


ult.htm. Accessed January 10, 2018. agents for lower gastrointestinal
1. Everhart JE, Ruhl CE. Burden of 10. Dechanont S et al. Hospital admis- motility disorders. Dis Colon Rectum
digestive diseases in the United sions/visits associated with drug-drug 1993; 36: 696–708.
States part I: overall and upper gas- interactions: a systematic review and 17. Tonini M. Recent advances in the
trointestinal diseases. Gastroenterol- meta-analysis. Pharmacoepidemiol pharmacology of gastrointestinal
ogy 2009; 136: 376–386. Drug Saf 2014; 23: 489–497. prokinetics. Pharmacol Res 1996; 33:
2. Peery A et al. Burden of gastroin- 11. Center for Drug Evaluation and 217–226.
testinal disease in the United States: Research. Resources for you – drug 18. Ehrlein HJ, Schemann M. Gastroin-
2012 update. Gastroenterology 2012; interactions: what you should know. testinal motility. Available at: http://
143: 1179–1187. Available at: https://www.fda.gov/d humanbiology.wzw.tum.de/motvid01/tu
3. Lindsley CW. 2014 global prescription rugs/resourcesforyou/ucm163354.htm. torial.pdf. Accessed January 15, 2018.
medication statistics: strong growth Accessed October 25, 2017. 19. Mandl P, Kiss JP. Role of presynaptic
and CNS well represented. ACS Chem 12. Eurostat. File:Self-reported use of nicotinic acetylcholine receptors in the
Neurosci 2015; 6: 505–506. non-prescribed medicines by sex, regulation of gastrointestinal motility.
4. Quigley EMM. Prokinetics in the 2014 (%).png – statistics explained. Brain Res Bull 2007; 72: 194–200.
management of functional gastroin- Available at: http://ec.europa.eu/eu 20. Gershon MD. Review article: sero-
testinal disorders. Curr Gastroenterol rostat/statistics-explained/index.php/ tonin receptors and transporters –
Rep 2017; 19: 53. File:Self-reported_use_of_non- roles in normal and abnormal
5. Enck P et al. Functional dyspepsia. prescribed_medicines_by_sex,_2014_ gastrointestinal motility. Aliment
Nat Rev Dis Primers 2017; 3: 17081. (%25).png. Accessed October 25, Pharmacol Ther 2004; 20: 3–14.
6. Pinto-Sanchez MI et al. Proton 2017. 21. Halpert A, Drossman D. 5-HT mod-
pump inhibitors for functional dys- 13. Connelly D. Sales of over-the-coun- ulators and other antidiarrheal agents
pepsia. Cochrane Database Syst Rev ter medicines in 2015 by clinical area and cathartics. In: Wolfe MM, Lowe
2017; (3): CD011194. and top 50 selling brands. Pharm J RC, eds. Pocket Guide to Gastroin-
7. Ford AC et al. Efficacy of 5-aminosa- 2016. https://doi.org/10.1211/PJ.2016. testinal Drugs. Chichester, UK: John
licylates in Crohn’s disease: system- 20200923. Wiley & Sons, Ltd, 2014: 57–81.
atic review and meta-analysis. Am J 14. Holzbauer M, Sharman DF. The distri- 22. Kale H, Fass R. Prokinetic agents and
Gastroenterol 2011; 106: 617–629. bution of catecholamines in verte- antiemetics. In: Wolfe MM, Lowe
8. EMA. Guideline on the investigation brates. In: Blaschko H, Muscholl E, eds. RC, eds. Pocket Guide to Gastroin-
of drug interactions. Guid Doc 2012; Catecholamines. Berlin, Heidelberg: testinal Drugs. Chichester, UK: John
44(June): 59. https://doi.org/10.1093/ Springer Berlin Heidelberg, 1972: 110– Wiley & Sons, Ltd, 2014: 1–14.
deafed/ens058 185. https://doi.org/10.1007/978-3-642- 23. Lee A, Kuo B. Metoclopramide in
9. Huang S-M. Clinical drug interac- 65249-3_5 the treatment of diabetic gastropare-
tion studies—study design, data 15. Orloff LA et al. Dopamine and nore- sis. Expert Rev Endocrinol Metab
analysis, and clinical implications pinephrine in the alimentary tract 2010; 5: 653–662.
guidance for industry. FDA Guid changes after chemical sympathec- 24. McCallum RW et al. Effects of meto-
2009. Available at: http://www.fda. tomy and surgical vagotomy. Life Sci clopramide and bethanechol on
gov/Drugs/GuidanceComplianceRe 1985; 36: 1625–1631. delayed gastric emptying present in

© 2018 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 71 (2019), pp. 643–673 663
Pharmacokinetic interactions with GI drugs Chara Litou et al.

gastroesophageal reflux patients. Gas- children with leukemia. Cancer Che- 50. Coremans G et al. Small doses of the
troenterology 1983; 84: 1573–1577. mother Pharmacol 1984; 12: 131–133. unabsorbable substance polyethylene
25. Fink SM et al. Effect of metoclo- 38. Pearson ADJ et al. Small intestinal glycol 3350 accelerate oro-caecal
pramide on normal and delayed gas- transit time affects methotrexate transit, but slow gastric emptying in
tric emptying in gastroesophageal absorption in children with acute healthy subjects. Dig Liver Dis 2005;
reflux patients. Dig Dis Sci 1983; 28: lymphoblastic leukemia. Cancer Che- 37: 97–101.
1057–1061. mother Pharmacol 1985; 14: 211–215. 51. Jou€et P et al. Effects of therapeutic
26. Parkman HP. Migraine and gastro- 39. Manninen V et al. Altered absorption doses of lactulose vs. polyethylene
paresis from a gastroenterologist’s of digoxin in patients given propan- glycol on isotopic colonic transit.
perspective. Headache 2013; 53: 4–10. theline and metoclopramide. Lancet Aliment Pharmacol Ther 2008; 27:
27. Tokola R, Neuvonen P. Effects of 1973; 1: 398–400. 988–993.
migraine attack and metoclopramide 40. Manninen V et al. Effect of propan- 52. Fritz E et al. Effects of lactulose and
on the absorption of tolfenamic acid. theline and metoclopramide on polyethylene glycol on colonic tran-
Br J Clin Pharmacol 1984; 17: 67–75. absorption of digoxin. Lancet 1973; sit. Aliment Pharmacol Ther 2005; 21:
28. Volans GN. The effect of metoclo- 1: 1118–1119. 259–268.
pramide on the absorption of effer- 41. Johnson BF et al. Effect of metoclo- 53. Barrow L et al. Scintigraphic demon-
vescent aspirin in migraine. Br J Clin pramide on digoxin absorption from stration of lactulose-induced acceler-
Pharmacol 1975; 2: 57–63. tablets and capsules. Clin Pharmacol ated proximal colon transit.
29. Gothoni G et al. Absorption of Ther 1984; 36: 724–730. Gastroenterology 1992; 103: 1167–1173.
antibiotics: influence of metoclo- 42. Wadhwa NK et al. The effect of oral 54. Manabe N et al. Effects of bisacodyl
pramide and atropine on serum metoclopramide on the absorption of on ascending colon emptying and
levels of pivampicillin and tetracy- cyclosporine. Transplantation 1987; overall colonic transit in healthy vol-
cline. Ann Clin Res 1972; 4: 228–232. 43: 211–213. unteers. Aliment Pharmacol Ther
30. Nimmo J et al. Pharmacological 43. Cash BD, Lacy BE. Systematic review: 2009; 30: 930–936.
modification of gastric emptying: FDA-approved prescription medica- 55. Guckenbiehl W et al. Effect of laxa-
effects of propantheline and metoclo- tions for adults with constipation. tives and metoclopramide on plasma
promide on paracetamol absorption. Gastroenterol Hepatol (N Y) 2006; 2: quinidine concentration during pro-
Br Med J 1973; 1: 587–589. 736–749. longed administration in patients
31. Wing LM et al. The effect of meto- 44. Tack J et al. Diagnosis and treatment with heart rhythm disorders. Med
clopramide and atropine on the of chronic constipation – a European Welt 1976; 26: 1273–1276. [in Ger-
absorption of orally administered perspective. Neurogastroenterol Motil man]
mexiletine. Br J Clin Pharmacol 1980; 2011; 23: 697–710. 56. Ragueneau I et al. Pharmacokinetic
9: 505–509. 45. Andresen V et al. Effect of 5 days and pharmacodynamic drug interac-
32. Crammer JL et al. Blood levels and linaclotide on transit and bowel tions between digoxin and macrogol
management of lithium treatment. Br function in females with constipa- 4000, a laxative polymer, in healthy
Med J 1974; 3: 650–654. tion-predominant irritable bowel volunteers. Br J Clin Pharmacol 1999;
33. Sanchez J et al. The influence of gas- syndrome. Gastroenterology 2007; 48: 453–456.
tric emptying on droxicam pharma- 133: 761–768. 57. Lewis SJ et al. Intestinal absorption
cokinetics. J Clin Pharmacol 1989; 29: 46. Lewis SJ, Heaton KW. Increasing of oestrogen: the effect of altering
739–745. butyrate concentration in the distal transit-time. Eur J Gastroenterol
34. Manara AR et al. The effect of meto- colon by accelerating intestinal tran- Hepatol 1998; 10: 33–39.
clopramide on the absorption of oral sit. Gut 1997; 41: 245–251. 58. Bown RL et al. Effects of lactulose
controlled release morphine. Br J 47. Klauser AG et al. Polyethylene glycol and other laxatives on ileal and colo-
Clin Pharmacol 1988; 25: 518–521. 4000 for slow transit constipation. Z nic pH as measured by a
35. Morris JGL et al. Plasma dopa con- Gastroenterol 1995; 33: 5–8. radiotelemetry device. Gut 1974; 15:
centrations after different prepara- 48. Corazziari E et al. Small volume isos- 999–1004.
tions of levodopa in normal subjects. motic polyethylene glycol electrolyte 59. Agostini L et al. Faecal ammonia and
Br J Clin Pharmacol 1976; 3: 983– balanced solution (PMF-100) in pH during lactulose administration
990. treatment of chronic nonorganic in man: comparison with other
36. Gugler R et al. Impaired cimetidine constipation. Dig Dis Sci 1996; 41: cathartics. Gut 1972; 13: 859–866.
absorption due to antacids and 1636–1642. 60. Mann NS et al. Effect of lactulose,
metoclopramide. Eur J Clin Pharma- 49. Ewe K et al. Effect of lactose, lactu- neomycin and antacid on colonic pH
col 1981; 20: 225–228. lose and bisacodyl on gastrointestinal recorded continuously with an
37. Mahony MJ et al. Modification of transit studied by metal detector. Ali- implanted electrode. Am J Gastroen-
oral methotrexate absorption in ment Pharmacol Ther 1995; 9: 69–73. terol 1979; 72: 141–145.

664 © 2018 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 71 (2019), pp. 643–673
Chara Litou et al. Pharmacokinetic interactions with GI drugs

61. Hussain FN et al. Mesalazine release 71. Kirby MG et al. Effect of metoclo- bioavailability. J Pharm Sci 1978; 67:
from a pH dependent formulation: pramide, bethanechol, and lop- 1582–1586.
effects of omeprazole and lactulose eramide on gastric residence time, 84. Albert KS et al. Pharmacokinetic eval-
co-administration. Br J Clin Pharma- gastric emptying, and mouth-to- uation of a drug interaction between
col 1998; 46: 173–175. cecum transit time. Pharmacotherapy kaolin–pectin and clindamycin. J
62. Riley SA et al. Mesalazine release 1989; 9: 226–231. Pharm Sci 1978; 67: 1579–1582.
from coated tablets: effect of dietary 72. Bryson JC et al. Effect of altering 85. Albert KS et al. Influence of kaolin-
fibre. Br J Clin Pharmacol 1991; 32: small bowel transit time on sustained pectin suspension on steady-state
248–250. release theophylline absorption. J plasma digoxin levels. J Clin Pharma-
63. Medicines.org.uk. Asacol 400 mg MR Clin Pharmacol 1989; 29: 733–738. col 1981; 21: 449–455.
tablets- summary of product charac- 73. Hughes S et al. Loperamide has antise- 86. Brown DD et al. Decreased bioavail-
teristics (SPC) – (eMC). 2018. Avail- cretory activity in the human jejunum ability of digoxin due to antacids and
able at: https://www.medicines.org. in vivo. Gut 1984; 25: 931–935. kaolin-pectin. N Engl J Med 1976;
uk/emc/product/2217/smpc. Accessed 74. Remington M et al. Inhibition of 295: 1034–1037.
June 3, 2018. postprandial pancreatic and biliary 87. Moustafa MA et al. Decreased
64. Medicines.org.uk. Salofalk 1000 mg secretion by loperamide in patients bioavailability of quinidine sulphate
gastro-resistant prolonged-release with short bowel syndrome*. Gut due to interactions with adsorbent
granules- summary of product charac- 1982; 23: 98–101. antacids and antidiarrhoeal mixtures.
teristics (SPC) – (eMC). 2018. Avail- 75. Thimister PWL et al. Inhibition of Int J Pharm 1987; 34: 207–211.
able at: https://www.medicines.org.uk/ pancreaticobiliary secretion by lop- 88. Liel Y et al. Evidence for a clinically
emc/product/140/smpc. Accessed June eramide in humans. Hepatology 1997; important adverse effect of fiber-
3, 2018. 26: 256–261. enriched diet on the bioavailability of
65. Bouhnik Y et al. Prospective, ran- 76. Callreus T et al. Changes in gastroin- levothyroxine in adult hypothyroid
domized, parallel-group trial to eval- testinal motility influence the absorp- patients. J Clin Endocrinol Metab
uate the effects of lactulose and tion of desmopressin. Eur J Clin 1996; 81: 857–859.
polyethylene glycol-4000 on colonic Pharmacol 1999; 55: 305–309. 89. FDA. Avoid food and drug interac-
flora in chronic idiopathic constipa- 77. Fredholt K et al. alpha-Chymotryp- tions. Available at: https://www.fda.
tion. Aliment Pharmacol Ther 2004; sin-catalyzed degradation of desmo- gov/downloads/drugs/resourcesforyou/c
19: 889–899. pressin (dDAVP): influence of pH, onsumers/buyingusingmedicinesafely/
66. Visser LE et al. Overanticoagulation concentration and various cyclodex- ensuringsafeuseofmedicine/generaluseof
associated with combined use of lac- trins. Int J Pharm 1999; 178: 223–229. medicine/ucm229033.pdf. Accessed
tulose and acenocoumarol or phen- 78. Mikus G et al. Reduction of saquina- September 6, 2017.
procoumon. Br J Clin Pharmacol vir exposure by coadministration of 90. Perlman B. Interaction between
2004; 57: 522–524. loperamide. Clin Pharmacokinet lithium salts and ispaghula husk.
67. Ippoliti C. Antidiarrheal agents for 2004; 43: 1015–1024. Lancet 1990; 335: 416.
the management of treatment-related 79. Wafik Gouda M. Effect of an 91. Stewart DE. High-fiber diet and
diarrhea in cancer patients. Am J antidiarrhoeal mixture on the serum tricyclic antidepressant levels.
Health Syst Pharm 1998; 55: 1573– bioavailability of tetracycline. Int J J Clin Psychopharmacol 1992; 12:
1580. Pharm 1993; 89: 75–77. 438–440.
68. Kachel G et al. Human intestinal 80. Juhl RP. Comparison of kaolin-pec- 92. Brown DD et al. Decreased bioavail-
motor activity and transport: effects tin and activated charcoal for inhibi- ability of digoxin due to hypocholes-
of a synthetic opiate. Gastroenterology tion of aspirin absorption. Am J terolemic interventions. Circulation
1986; 90: 85–93. Hosp Pharm 1979; 36: 1097–1098. 1978; 58: 164–172.
69. Press AG et al. Effect of loperamide 81. Al-Shora HI et al. Interactions of 93. Lembcke B et al. Plasma digoxin
on jejunal electrolyte and water procainamide, verapamil, guanethi- concentrations during administration
transport, prostaglandin E 2-induced dine and hydralazine with adsorbent of dietary fibre (guar gum) in man.
secretion and intestinal transit time antacids and antidiarrhoeal mixtures. Z Gastroenterol 1982; 20: 164–167.
in man. Eur J Clin Pharmacol 1991; Int J Pharm 1988; 47: 209–213. 94. Kasper H et al. The effect of dietary
41: 239–243. 82. Gupta KC et al. Effect of pectin and fiber on postprandial serum digoxin
70. Sninsky CA et al. Effect of lidami- kaolin on bioavailability of co-tri- concentration in man. Am J Clin
dine hydrochloride and loperamide moxazole suspension. Int J Clin Nutr 1979; 32: 2436–2438.
on gastric emptying and transit of Pharmacol Ther Toxicol 1987; 25: 95. Huupponen R et al. Effect of guar
the small intestine: a double-blind 320–321. gum, a fibre preparation, on digoxin
study. Gastroenterology 1986; 90: 68– 83. Albert KS et al. Influence of kaolin– and penicillin absorption in man.
73. pectin suspension on digoxin

© 2018 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 71 (2019), pp. 643–673 665
Pharmacokinetic interactions with GI drugs Chara Litou et al.

Eur J Clin Pharmacol 1984; 26: 279– chemotherapy. Biol Pharm Bull 2016; contents in the fasted state under
281. 39: 863–868. hypo- and A-chlorhydric gastric con-
96. Holt S et al. Effect of gel fibre on 108. EMENDâ. Clinical pharmacology ditions under conditions of typical
gastric emptying and absorption of and biopharmaceutics review. Avail- drug – drug interaction studies.
glucose and paracetamol. Lancet able at: https://www.accessdata.fda. Pharm Res 2016; 33: 1399–1412.
1979; 1: 636–639. gov/drugsatfda_docs/nda/2003/21- 119. Meyer UA. Interaction of proton
97. Reppas C et al. Effect of elevated vis- 549_Emend_biopharmr.pdf. Accessed pump inhibitors with cytochromes
cosity in the upper gastrointestinal August 31, 2017. P450: consequences for drug interac-
tract on drug absorption in dogs. 109. Blower PR. Granisetron: relating tions. Yale J Biol Med 1996; 69: 203–
Eur J Pharm Sci 1998; 6: 131–139. pharmacology to clinical efficacy. 209.
98. Astarloa R et al. Clinical and pharma- Support Care Cancer 2003; 11: 93– 120. Ogawa R, Echizen H. Drug-drug
cokinetic effects of a diet rich in insol- 100. interaction profiles of proton pump
uble fiber on Parkinson disease. Clin 110. Gralla RJ et al. Recommendations inhibitors. Clin Pharmacokinet 2010;
Neuropharmacol 1992; 15: 375–380. for the use of antiemetics: evidence- 49: 509–533.
99. Gonzalez Canga A et al. Dietary fiber based, clinical practice guidelines. J 121. Lahner E et al. Systematic review:
and its interaction with drugs. Nutr Clin Oncol 1999; 17: 2971–2994. impaired drug absorption related to
Hosp 2010; 25: 535–539. 111. Cagnoni PJ et al. Modification of the the co-administration of antisecre-
100. Reppas C et al. Effect of hydrox- pharmacokinetics of high-dose tory therapy. Aliment Pharmacol Ther
ypropylmethylcellulose on gastroin- cyclophosphamide and cisplatin by 2009; 29: 1219–1229.
testinal transit and luminal viscosity antiemetics. Bone Marrow Transplant 122. Wedemeyer R-S, Blume H. Pharma-
in dogs. Gastroenterology 1991; 100: 1999; 24: 1–4. cokinetic drug interaction profiles of
1217–1223. 112. Gilbert CJ et al. Pharmacokinetic proton pump inhibitors: an update.
101. FDA-emend capsules pharmacology interaction between ondansetron and Drug Saf 2014; 37: 201–211.
review part 1.pdf. Available at: https:// cyclophosphamide during high-dose 123. Jaruratanasirikul S, Sriwiriyajan S.
www.accessdata.fda.gov/drugsatfda_d chemotherapy for breast cancer. Effect of omeprazole on the pharma-
ocs/nda/2003/21-549_Emend.cfm. Cancer Chemother Pharmacol 1998; cokinetics of itraconazole. Eur J Clin
102. EMA. Emend: summary of product 42: 497–503. Pharmacol 1998; 54: 159–161.
characteristics. Available at: http:// 113. Speaks M. Health United States 124. Johnson MD et al. A randomized
www.ema.europa.eu/docs/en_GB/docu report 2016. 2016. Available at: comparative study to determine the
ment_library/EPAR_-_Product_Inf https://www.cdc.gov/nchs/data/hus/ effect of omeprazole on the peak
ormation/human/000527/WC50002 hus16.pdf#080. Accessed August 28, serum concentration of itraconazole
6537.pdf. Accessed October 25, 2017. 2017. oral solution. J Antimicrob Che-
103. Blower P et al. Drug-drug interac- 114. 100 best-selling, most prescribed mother 2003; 51: 453–457.
tions in oncology: why are they branded drugs through March. Avail- 125. Chin TW et al. Effects of an acidic
important and can they be mini- able at: http://www.medscape.com/vie beverage (Coca-Cola) on absorption
mized? Crit Rev Oncol Hematol 2005; warticle/844317#vp_1. Accessed of ketoconazole. Antimicrob Agents
55: 117–142. August 28, 2017. Chemother 1995; 39: 1671–1675.
104. Majumdar AK et al. Effects of aprepi- 115. Arnold R. Safety of proton pump 126. Nexiumâ. Clinical pharmacology
tant on cytochrome P450 3A4 activity inhibitors–an overview. Aliment Phar- and biopharmaceutics review. Avail-
using midazolam as a probe. Clin macol Ther 1994; 8(Suppl 1): 65–70. able at: https://www.accessdata.fda.
Pharmacol Ther 2003; 74: 150–156. 116. Blanton WP, Wolfe MM. Proton gov/drugsatfda_docs/nda/2001/21154_
105. Majumdar AK et al. Effect of aprepi- pump inhibitors. In: Wolfe MM, Nexium_biopharmr_P1.pdf. Accessed
tant on the pharmacokinetics of Lowe RC, eds. Pocket Guide to Gas- August 29, 2017.
intravenous midazolam. J Clin Phar- trointestinal Drugs. Chichester, UK: 127. DIFLUCANâ (Fluconazole tablets)
macol 2007; 47: 744–750. John Wiley & Sons, Ltd, 2014: 15– (fluconazole for oral suspension).
106. McCrea JB et al. Effects of the neu- 30. Available at: https://www.accessda
rokinin1 receptor antagonist aprepi- 117. Sugimoto M et al. Treatment strat- ta.fda.gov/drugsatfda_docs/label/2014/
tant on the pharmacokinetics of egy to eradicate Helicobacter pylori 019949s060,020090s044lbl.pdf. Acces-
dexamethasone and methylpred- infection: impact of pharmacoge- sed August 29, 2017.
nisolone. Clin Pharmacol Ther 2003; nomics-based acid inhibition regi- 128. H€orter D, Dressman J. Influence of
74: 17–24. men and alternative antibiotics. physicochemical properties on disso-
107. Takaki J et al. Assessment of drug- Expert Opin Pharmacother 2007; 8: lution of drugs in the gastrointestinal
drug interaction between warfarin 2701–2717. tract. Adv Drug Deliv Rev 2001; 46:
and aprepitant and its effects on PT- 118. Litou C et al. Characteristics of the 75–87.
INR of patients receiving anticancer human upper gastrointestinal

666 © 2018 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 71 (2019), pp. 643–673
Chara Litou et al. Pharmacokinetic interactions with GI drugs

129. Thorpe JE et al. Effect of oral antacid ta.fda.gov/drugsatfda_docs/nda/98/ www.fda.gov/Drugs/DrugSafety/uc


administration on the pharmacoki- 20835_Actonel_biopharmr.pdf. m231161.htm. Accessed August 29,
netics of oral fluconazole. Antimicrob Accessed August 29, 2017. 2017.
Agents Chemother 1990; 34: 2032– 140. Budha NR et al. Drug absorption 150. Stockl KM et al. Risk of rehospital-
2033. interactions between oral targeted ization for patients using clopidogrel
130. Tappouni HL et al. Effect of anticancer agents and ppis: is pH- with a proton pump inhibitor. Arch
omeprazole on the plasma concen- dependent solubility the Achilles heel Intern Med 2010; 170: 704.
trations of indinavir when adminis- of targeted therapy? Clin Pharmacol 151. Evanchan J et al. Recurrence of acute
tered alone and in combination with Ther 2012; 92: 203–213. myocardial infarction in patients dis-
ritonavir. Am J Health Syst Pharm 141. Mitra A, Kesisoglou F. Impaired charged on clopidogrel and a proton
2008; 65: 422–428. drug absorption due to high stomach pump inhibitor after stent placement
131. Fang AF et al. Significant decrease in pH: a review of strategies for mitiga- for acute myocardial infarction. Clin
nelfinavir systemic exposure after tion of such effect to enable pharma- Cardiol 2010; 33: 168–171.
omeprazole coadministration in ceutical product development. Mol 152. Gaglia MA et al. Relation of proton
healthy subjects. Pharmacotherapy Pharm 2013; 10: 3970–3979. pump inhibitor use after percuta-
2008; 28: 42–50. 142. Soons PA et al. Influence of single- neous coronary intervention with
132. Tomilo DL et al. Inhibition of ataza- and multiple-dose omeprazole treat- drug-eluting stents to outcomes. Am
navir oral absorption by lansoprazole ment on nifedipine pharmacokinetics J Cardiol 2010; 105: 833–838.
gastric acid suppression in healthy and effects in healthy subjects. Eur J 153. Chua D et al. Clopidogrel and pro-
volunteers. Pharmacotherapy 2006; Clin Pharmacol 1992; 42: 319–324. ton pump inhibitors: a new drug
26: 341–346. 143. Bliesath H et al. Pantoprazole does interaction? Can J Hosp Pharm 2010;
133. Klein CE et al. Effects of acid-redu- not interact with nifedipine in man 63: 47–50.
cing agents on the pharmacokinetics under steady-state conditions. Int J 154. Bundhun PK et al. Is the concomi-
of lopinavir/ritonavir and ritonavir- Clin Pharmacol Ther 1996; 34: 51–55. tant use of clopidogrel and Proton
boosted atazanavir. J Clin Pharmacol 144. Zvyaga T et al. Evaluation of six pro- Pump Inhibitors still associated with
2008; 48: 553–562. ton pump inhibitors as inhibitors of increased adverse cardiovascular out-
134. Furtek KJ et al. Proton pump inhibitor various human cytochromes P450: comes following coronary angio-
therapy in atazanavir-treated patients: focus on cytochrome P450 2C19. plasty?: a systematic review and
contraindicated? J Acquir Immune Drug Metab Dispos 2012; 40: 1698– meta-analysis of recently published
Defic Syndr 2006; 41: 394–396. 1711. studies (2012–2016). BMC Cardio-
135. Sahloff EG, Duggan JM. Clinical out- 145. Li X-Q et al. Comparison of vasc Disord 2017; 17: 3.
comes associated with concomitant inhibitory effects of the proton 155. Sugano K. Histamine H2-receptor
use of atazanavir and proton pump pump-inhibiting drugs omeprazole, antagonists. In: Wolf MM, Lowe RC,
inhibitors. Ann Pharmacother 2006; esomeprazole, lansoprazole, panto- eds. Pocket Guide to Gastrointestinal
40: 1731–1736. prazole, and rabeprazole on human Drugs. Chichester, UK: John Wiley &
136. Winston A et al. Effect of omepra- cytochrome P450 activities. Drug Sons, Ltd, 2014: 31–43.
zole on the pharmacokinetics of Metab Dispos 2004; 32: 821–827. 156. Piscitelli SC et al. Effects of rani-
saquinavir-500 mg formulation with 146. Ko JW et al. Evaluation of omepra- tidine and sucralfate on ketoconazole
ritonavir in healthy male and female zole and lansoprazole as inhibitors of bioavailability. Antimicrob Agents
volunteers. AIDS 2006; 20: 1401– cytochrome P450 isoforms. Drug Chemother 1991; 35: 1765–1771.
1406. Metab Dispos 1997; 25: 853–862. 157. Lim SG et al. Short report: the
137. Kofler S et al. Proton pump inhibi- 147. Blume H et al. Pharmacokinetic drug absorption of fluconazole and itra-
tor co-medication reduces mycophe- interaction profiles of proton pump conazole under conditions of low
nolate acid drug exposure in heart inhibitors. Drug Saf 2006; 29: 769– intragastric acidity. Aliment Pharma-
transplant recipients. J Hear Lung 784. col Ther 2007; 7: 317–321.
Transplant 2009; 28: 605–611. 148. Suzuki K et al. Co-administration of 158. Blum RA et al. Increased gastric pH
138. Rupprecht K et al. Bioavailability of proton pump inhibitors delays elimi- and the bioavailability of fluconazole
mycophenolate mofetil and enteric- nation of plasma methotrexate in and ketoconazole. Ann Intern Med
coated mycophenolate sodium is dif- high-dose methotrexate therapy. Br J 1991; 114: 755–757.
ferentially affected by pantoprazole Clin Pharmacol 2009; 67: 44–49. 159. Ford SL et al. Effect of antacids and
in healthy volunteers. J Clin Pharma- 149. Drug safety and availability – FDA ranitidine on the single-dose phar-
col 2009; 49: 1196–1201. reminder to avoid concomitant use macokinetics of fosamprenavir.
139. Actonelâ. Clinical pharmacology of Plavix (clopidogrel) and omepra- Antimicrob Agents Chemother 2005;
and biopharmaceutics review. Avail- zole. Available at: https:// 49: 467–469.
able at: https://www.accessda

© 2018 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 71 (2019), pp. 643–673 667
Pharmacokinetic interactions with GI drugs Chara Litou et al.

160. Boffito M et al. Pharmacokinetics of quercetin with Al(III). J Inorg Bio- gemifloxacin in healthy volunteers.
saquinavir co-administered with chem 2002; 92: 19–27. Chemotherapy 1999; 45: 504–511.
cimetidine. J Antimicrob Chemother 173. T€urkel N et al. Potentiometric and 184. Lohitnavy M et al. Reduced oral itra-
2002; 50: 1081–1084. spectroscopic studies on aluminium conazole bioavailability by antacid
161. Russell TL et al. pH-Related changes (III) complexes of some catechol suspension. J Clin Pharm Ther 2005;
in the absorption of dipyridamole in derivatives. Chem Pharm Bull 30: 201–206.
the elderly. Pharm Res 1994; 11: 136– (Tokyo) 2004; 52: 929–934. 185. Brass C et al. Disposition of keto-
143. 174. Khan MA et al. Differential binding conazole, an oral antifungal, in
162. Grasela TH et al. Inhibition of enox- of tetracyclines with serum albumin humans. Antimicrob Agents Che-
acin absorption by antacids or rani- and induced structural alterations in mother 1982; 21: 151–158.
tidine. Antimicrob Agents Chemother drug-bound protein. Int J Biol 186. Neuvonen PJ. The effect of magne-
1989; 33: 615–617. Macromol 2002; 30: 243–249. sium hydroxide on the oral absorp-
163. Hughes GS et al. The effects of gas- 175. Cordoba-Dıaz M et al. Modification tion of ibuprofen, ketoprofen and
tric pH and food on the pharma- of fluorescent properties of nor- diclofenac. Br J Clin Pharmacol 1991;
cokinetics of a new oral floxacin in the presence of certain 31: 263–266.
cephalosporin, cefpodoxime proxetil. antacids. J Pharm Biomed Anal 1998; 187. Tobert JA et al. Effect of antacids on
Clin Pharmacol Ther 1989; 46: 674– 18: 565–571. the bioavailability of diflunisal in the
685. 176. Deppermann KM et al. Influence of fasting and postprandial states. Clin
164. Gerber MC et al. Drug interactions ranitidine, pirenzepine, and alu- Pharmacol Ther 1981; 30: 385–389.
with cimetidine: an update. Pharma- minum magnesium hydroxide on the 188. Neuvonen PJ, Kivist€ o KT. Effect of
col Ther 1985; 27: 353–370. bioavailability of various antibiotics, magnesium hydroxide on the
165. Berardi RR et al. Comparison of including amoxicillin, cephalexin, absorption of tolfenamic and mefe-
famotidine with cimetidine and rani- doxycycline, and amoxicillin-clavula- namic acids. Eur J Clin Pharmacol
tidine. Clin Pharm 1988; 7: 271–284. nic acid. Antimicrob Agents Che- 1988; 35: 495–501.
166. O’Reilly RA. Comparative interaction mother 1989; 33: 1901–1907. 189. Segre EJ et al. Transport of organic
of cimetidine and ranitidine with 177. Garty M, Hurwitz A. Effect of cime- acids across cell membrane. N Engl J
racemic warfarin in man. Arch Intern tidine and antacids on gastrointesti- Med 1974; 291: 582.
Med 1984; 144: 989–991. nal absorption of tetracycline. Clin 190. Ogawa R, Echizen H. Clinically sig-
167. Toon S et al. Comparative effects of Pharmacol Ther 1980; 28: 203–207. nificant drug interactions with anta-
ranitidine and cimetidine on the 178. Timmers K, Sternglanz R. Ionization cids: an update. Drugs 2011; 71:
pharmacokinetics and pharmacody- and divalent cation dissociation con- 1839–1864.
namics of warfarin in man. Eur J stants of nalidixic and oxolinic acids. 191. Gareau MG et al. Probiotics and the
Clin Pharmacol 1987; 32: 165–172. Bioinorg Chem 1978; 9: 145–155. gut microbiota in intestinal health
168. Niopas I et al. The effect of cime- 179. Radandt JM et al. Interactions of flu- and disease. Nat Rev Gastroenterol
tidine on the steady-state pharma- oroquinolones with other drugs: Hepatol 2010; 7: 503–514.
cokinetics and pharmacodynamics of mechanisms, variability, clinical sig- 192. Guidelines for the evaluation of pro-
warfarin in humans. Eur J Clin Phar- nificance, and management. Clin biotics in food report. Joint FAO/
macol 1999; 55: 399–404. Infect Dis 1992; 14: 272–284. WHO Working Group report on
169. Reimann IW et al. Cimetidine 180. Nix DE et al. Effects of aluminum drafting guidelines for the evaluation
increases steady state plasma levels of and magnesium antacids and rani- of probiotics in food. 2002. Available
propranolol. Br J Clin Pharmacol tidine on the absorption of cipro- at: http://www.who.int/foodsafety/
1981; 12: 785–790. floxacin. Clin Pharmacol Ther 1989; fs_management/en/probiotic_guide
170. Reimann IW et al. Effects of cime- 46: 700–705. lines.pdf. Accessed September 5, 2017.
tidine and ranitidine on steady-state 181. Krishna G et al. Effect of an alu- 193. Westendorf AM et al. Intestinal
propranolol kinetics and dynamics. minum- and magnesium-containing immunity of Escherichia coli NISSLE
Clin Pharmacol Ther 1982; 32: 749– antacid on the bioavailability of gare- 1917: a safe carrier for therapeutic
757. noxacin in healthy volunteers. Phar- molecules. FEMS Immunol Med
171. Medicines.org.uk. Propranolol film- macotherapy 2007; 27: 963–969. Microbiol 2005; 43: 373–384.
coated tablets- Patient Information 182. Lober S et al. Pharmacokinetics of 194. Resta-Lenert SC, Barrett KE. Modu-
Leaflet (PIL) – (eMC). 2018. Avail- gatifloxacin and interaction with an lation of intestinal barrier properties
able at: https://www.medicines.org. antacid containing aluminum and by probiotics: role in reversing coli-
uk/emc/files/pil.2904.pdf. Accessed magnesium. Antimicrob Agents Che- tis. Ann N Y Acad Sci 2009; 1165:
June 3, 2018. mother 1999; 43: 1067–1071. 175–182.
172. Cornard J, Merlin J. Spectroscopic 183. Allen A et al. Effect of Maalox on 195. Mikov M et al. The influence of pro-
and structural study of complexes of the bioavailability of oral biotic treatment on sulfasalazine

668 © 2018 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 71 (2019), pp. 643–673
Chara Litou et al. Pharmacokinetic interactions with GI drugs

metabolism in rat gut contents. decreased in antibiotic-associated diet in healthy volunteers. Eur J Gas-
Asian J Pharmacodyn Pharmacokinet diarrhea. Gastroenterology 1991; 101: troenterol Hepatol 1995; 7: 797–802.
2006; 6: 337–342. 1497–1504. 218. Caron F et al. Effects of two oral ery-
196. Lee HJ et al. The influence of probi- 207. Edwards CA et al. Effect of clin- thromycin ethylsuccinate formula-
otic treatment on sulfasalazine meta- damycin on the ability of a continu- tions on the motility of the small
bolism in rat. Xenobiotica 2012; 42: ous culture of colonic bacteria to intestine in human beings. Antimi-
791–797. ferment carbohydrate. Gut 1986; 27: crob Agents Chemother 1996; 40:
197. Kunes M et al. Absorption kinetics 411–417. 1796–1800.
of 5-aminosalicylic acid in rat: influ- 208. Gustafsson A et al. Faecal short- 219. Annese V et al. Erythromycin accel-
ence of indomethacin-induced gas- chain fatty acids in patients with erates gastric emptying by inducing
trointestinal lesions and Escherichia antibiotic-associated diarrhoea, antral contractions and improved
coli Nissle 1917 medication. Neuro before and after faecal enema treat- gastroduodenal coordination. Gas-
Endocrinol Lett 2011; 32(Suppl 1): ment. Scand J Gastroenterol 1998; 33: troenterology 1992; 102: 823–828.
46–52. 721–727. 220. Leung WK et al. Effect of oral ery-
198. Al-Salami H et al. Probiotic treat- 209. Mellon AF et al. Effect of oral antibi- thromycin on gastric and small
ment reduces blood glucose levels otics on intestinal production of pro- bowel transit time of capsule endo-
and increases systemic absorption of pionic acid. Arch Dis Child 2000; 82: scopy. World J Gastroenterol 2005;
gliclazide in diabetic rats. Eur J Drug 169–172. 11: 4865–4868.
Metab Pharmacokinet 2008; 33: 101– 210. Høverstad T et al. Influence of oral 221. Chew ML et al. Effect of the gas-
106. intake of seven different antibiotics trointestinal prokinetic agent ery-
199. Saksena S et al. Upregulation of P- on faecal short-chain fatty acid thromycin on the pharmacokinetics
glycoprotein by probiotics in intesti- excretion in healthy subjects. Scand J of pregabalin controlled-release in
nal epithelial cells and in the dextran Gastroenterol 1986; 21: 997–1003. healthy individuals: a phase I, ran-
sulfate sodium model of colitis in 211. Kamath PS et al. Short-chain fatty domized crossover trial. Clin Drug
mice. Am J Physiol Gastrointest Liver acids stimulate ileal motility in Investig 2015; 35: 299–305.
Physiol 2011; 300: G1115–G1123. humans. Gastroenterology 1988; 95: 222. Sousa T et al. The gastrointestinal
200. Matuskova Z et al. Administration of 1496–1502. microbiota as a site for the biotrans-
a probiotic can change drug pharma- 212. Fich A et al. Stimulation of ileal formation of drugs. Int J Pharm
cokinetics: effect of E. coli Nissle emptying by short-chain fatty acids. 2008; 363: 1–25.
1917 on amidarone absorption in Dig Dis Sci 1989; 34: 1516–1520. 223. Saad R et al. Gut pharmacomicro-
rats. PLoS One 2014; 9: 3–7. 213. Aguilera M et al. Antibiotic-induced biomics: the tip of an iceberg of
201. Fr€ohlich EE et al. Cognitive impair- dysbiosis alters host-bacterial interac- complex interactions between drugs
ment by antibiotic-induced gut dys- tions and leads to colonic sensory and gut-associated microbes. Gut
biosis: analysis of gut microbiota- and motor changes in mice. Gut Pathog 2012; 4: 16.
brain communication. Brain Behav Microbes 2015; 6: 10–23. 224. Lindenbaum J et al. Inactivation of
Immun 2016; 56: 140–155. 214. Cherbut C et al. Effects of short- digoxin by the gut flora: reversal by
202. Sullivan A et al. Effect of antimicro- chain fatty acids on gastrointestinal antibiotic therapy. N Engl J Med
bial agents on the ecological balance motility. Scand J Gastroenterol 1997; 1981; 305: 789–794.
of human microflora. Lancet Infect 32: 58–61. 225. Morton MR, Cooper JW. Ery-
Dis 2001; 1: 101–114. 215. Edelbroek MA et al. Effects of ery- thromycin-induced digoxin toxicity.
203. Edlund C, Nord CE. Effect on the thromycin on gastric emptying, alco- DICP 1989; 23: 668–670.
human normal microflora of oral hol absorption and small intestinal 226. Vermes A et al. An in vitro study on
antibiotics for treatment of urinary transit in normal subjects. J Nucl the active conversion of flucytosine
tract infections. J Antimicrob Che- Med 1993; 34: 582–588. to fluorouracil by microorganisms in
mother 2000; 46(Suppl 1): 41–48; 216. Mantides A et al. The effect of ery- the human intestinal microflora.
discussion 63–5. thromycin in gastric emptying of Chemotherapy 2003; 49: 17–23.
204. Beaugerie L, Petit J-C. Antibiotic- solids and hypertonic liquids in 227. Houston JB et al. Azo reduction of
associated diarrhoea. Best Pract Res healthy subjects. Am J Gastroenterol sulphasalazine in healthy volunteers.
Clin Gastroenterol 2004; 18: 337–352. 1993; 88: 198–202. Br J Clin Pharmacol 1982; 14: 395–398.
205. Tremaroli V, B€ackhed F. Functional 217. Landry C et al. Effects of ery- 228. Singh SK et al. A novel dissolution
interactions between the gut micro- thromycin on gastric emptying, duo- method for evaluation of polysaccha-
biota and host metabolism. Nature deno-caecal transit time, gastric and ride based colon specific delivery sys-
2012; 489: 242–249. biliopancreatic secretion during con- tems: a suitable alternative to animal
206. Clausen MR et al. Colonic fermenta- tinuous gastric infusion of a liquid sacrifice. Eur J Pharm Sci 2015; 73:
tion to short-chain fatty acids is 72–80.

© 2018 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 71 (2019), pp. 643–673 669
Pharmacokinetic interactions with GI drugs Chara Litou et al.

229. Hofmann AF, Hagey LR. Bile acids: gastrointestinal transit influence of 253. Bradley SM et al. Sequential study of
chemistry, pathochemistry, biology, olsalazine and sulphasalazine on gas- bacterial antibody levels and faecal
pathobiology, and therapeutics. Cell trointestinal transit. Scand J Gas- flora in rheumatoid arthritis patients
Mol Life Sci 2008; 65: 2461–2483. troenterol 1988; 23: 148–196. taking sulphasalazine. Br J Rheumatol
230. Brestoff JR, Artis D. Commensal bac- 243. Rao SS et al. Influence of olsalazine 1993; 32: 683–688.
teria at the interface of host metabo- on gastrointestinal transit in ulcera- 254. Xue L et al. The possible effects of
lism and the immune system. Nat tive colitis. Gut 1987; 28: 1474–1477. mesalazine on the intestinal micro-
Immunol 2013; 14: 676–684. 244. Staniforth DH. Comparison of oro- biota. Aliment Pharmacol Ther 2012;
231. Vrieze A et al. Impact of oral van- caecal transit times assessed by the 36: 813–814.
comycin on gut microbiota, bile acid lactulose/breath hydrogen and the 255. Andrews CN et al. Mesalazine (5-ami-
metabolism, and insulin sensitivity. J sulphasalazine/sulphapyridine meth- nosalicylic acid) alters faecal bacterial
Hepatol 2014; 60: 824–831. ods. Gut 1989; 30: 978–982. profiles, but not mucosal proteolytic
232. S€oderlind E et al. Simulating fasted 245. Raimundo A et al. Gastrointestinal activity in diarrhoea-predominant irri-
human intestinal fluids: understand- pH profiles in ulcerative colitis. Gas- table bowel syndrome. Aliment Phar-
ing the roles of lecithin and bile troenterology 1992; 4: A681. macol Ther 2011; 34: 374–383.
acids. Mol Pharm 2010; 7: 1498– 246. Nugent SG et al. Intestinal luminal 256. Juhl RP et al. Effect of sulfasalazine
1507. pH in inflammatory bowel disease: on digoxin bioavailability. Clin Phar-
233. von Rosensteil NA, Adam D. Macro- possible determinants and implica- macol Ther 1976; 20: 387–394.
lide antibacterials. Drug interactions tions for therapy with aminosalicy- 257. Marcus FI. Pharmacokinetic interac-
of clinical significance. Drug Saf lates and other drugs. Gut 2001; 48: tions between digoxin and other
1995; 13: 105–122. 571–577. drugs. J Am Coll Cardiol 1985; 5(5
234. Olkkola KT et al. A potentially haz- 247. Raimundo AH et al. Effects of olsala- Suppl A): 82A–90A.
ardous interaction between ery- zine and sulphasalazine on jejunal 258. Haiser HJ et al. Mechanistic insight
thromycin and midazolam. Clin and ileal water and electrolyte into digoxin inactivation by
Pharmacol Ther 1993; 53: 298–305. absorption in normal human sub- Eggerthella lenta augments our
235. Gorski JC et al. The contribution of jects. Gut 1991; 32: 270–274. understanding of its pharmacokinet-
intestinal and hepatic CYP3A to the 248. Sandberg-Gertzen H et al. Azodisal ics. Gut Microbes 2014; 5: 233–238.
interaction between midazolam and sodium in the treatment of ulcerative 259. Du Cheyron D et al. Effect of sul-
clarithromycin. Clin Pharmacol Ther colitis. A study of tolerance and fasalazine on cyclosporin blood con-
1998; 64: 133–143. relapse-prevention properties. Gas- centration. Eur J Clin Pharmacol
236. Yeates RA et al. Interaction between troenterology 1986; 90: 1024–1030. 1999; 55: 227–228.
midazolam and clarithromycin: com- 249. Zimmerman J. Drug interactions in 260. Lewis LD et al. Olsalazine and 6-
parison with azithromycin. Int J Clin intestinal transport of folic acid and mercaptopurine-related bone marrow
Pharmacol Ther 1996; 34: 400–405. methotrexate. Further evidence for suppression: a possible drug-drug
237. Douros A et al. Safety issues and the heterogeneity of folate transport interaction. Clin Pharmacol Ther
drug–drug interactions with com- in the human small intestine. Bio- 1997; 62: 464–475.
monly used quinolones. Expert Opin chem Pharmacol 1992; 44: 1839– 261. Lowry PW et al. Balsalazide and aza-
Drug Metab Toxicol 2014; 11: 1–15. 1842. thiprine or 6-mercaptopurine: evi-
238. Beckmann J et al. Enoxacin–a potent 250. Okada M et al. Drug interaction dence for a potentially serious drug
inhibitor of theophylline metabolism. between methotrexate and salazosul- interaction. Gastroenterology 1999;
Eur J Clin Pharmacol 1987; 33: 227– fapyridine in Japanese patients with 116: 1505–1506.
230. rheumatoid arthritis. J Pharm Health 262. Bengmark S, Jeppsson B. Gastroin-
239. Batty KT et al. The effect of cipro- Care Sci 2017; 3: 7. testinal surface protection and
floxacin on theophylline pharma- 251. Kanerud L et al. Effect of sul- mucosa reconditioning. JPEN J Par-
cokinetics in healthy subjects. Br J phasalazine on gastrointestinal enter Enteral Nutr 1995; 19: 410–415.
Clin Pharmacol 1995; 39: 305–311. microflora and on mucosal heat 263. Narum S et al. Corticosteroids and
240. Bowles SK et al. Effect of norfloxacin shock protein expression in patients risk of gastrointestinal bleeding: a
on theophylline pharmacokinetics at with rheumatoid arthritis. Br J systematic review and meta-analysis.
steady state. Antimicrob Agents Che- Rheumatol 1994; 33: 1039–1048. BMJ Open 2014; 4: e004587.
mother 1988; 32: 510–512. 252. Neumann VC et al. Effects of sul- 264. Jung D et al. Human ileal bile acid
241. Pai MP et al. Antibiotic drug inter- phasalazine on faecal flora in patients transporter gene ASBT (SLC10A2) is
actions. Med Clin North Am 2006; with rheumatoid arthritis: a compar- transactivated by the glucocorticoid
90: 1223–1255. ison with penicillamine. Br J receptor. Gut 2004; 53: 78–84.
242. Rao SS et al. Influence of olsalazine Rheumatol 1987; 26: 334–337. 265. Bajor A et al. Budesonide treatment
and sulphasalazine on is associated with increased bile acid

670 © 2018 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 71 (2019), pp. 643–673
Chara Litou et al. Pharmacokinetic interactions with GI drugs

absorption in collagenous colitis. Ali- 277. Teixeira Mdo CB et al. Influence of of CYP3A4 and CYP3A5 in vitro.
ment Pharmacol Ther 2006; 24: post-transplant immunosuppressive Drug Metab Dispos 2012; 40: 655–
1643–1649. therapy on gastrointestinal transit 661.
266. Fleisher D et al. Drug, meal and for- using biomagnetic method: a pilot 289. Moes DJAR et al. Sirolimus and
mulation interactions influencing study. Dig Dis Sci 2015; 60: 174–180. everolimus in kidney transplantation.
drug absorption after oral adminis- 278. Gabe SM et al. The effect of tacroli- Drug Discov Today 2015; 20: 1243–
tration. Clin Pharmacokinet 1999; 36: mus (FK506) on intestinal barrier 1249.
233–254. function and cellular energy produc- 290. Finch A, Pillans P. P-glycoprotein
267. Dilger K et al. Identification of tion in humans. Gastroenterology and its role in drug-drug interac-
budesonide and prednisone as sub- 1998; 115: 67–74. tions. Aust Prescr 2014; 37: 137–139.
strates of the intestinal drug efflux 279. Parrilli G et al. Effect of chronic 291. Rebello S et al. Effect of cyclosporine
pump P-glycoprotein. Inflamm Bowel administration of tacrolimus and on the pharmacokinetics of aliskiren
Dis 2004; 10: 578–583. cyclosporine on human gastrointesti- in healthy subjects. J Clin Pharmacol
268. Schwab M, Klotz U. Pharmacokinetic nal permeability. Liver Transpl 2003; 2011; 51: 1549–1560.
considerations in the treatment of 9: 484–488. 292. Rushing DA et al. The effects of
inflammatory bowel disease. Clin 280. Helderman JH, Goral S. Gastroin- cyclosporine on the pharmacokinet-
Pharmacokinet 2001; 40: 723–751. testinal complications of transplant ics of doxorubicin in patients with
269. Eradiri O et al. Interaction of immunosuppression. J Am Soc small cell lung cancer. Cancer 1994;
metronidazole with phenobarbital, Nephrol 2002; 13: 277–287. 74: 834–841.
cimetidine, prednisone, and sul- 281. Deeming GMJ et al. Methotrexate 293. Eising EG et al. Does the multidrug-
fasalazine in Crohn’s disease. Bio- and oral ulceration. Br Dent J 2005; resistance modulator cyclosporin A
pharm Drug Dispos 1988; 9: 219–227. 198: 83–85. increase the cardiotoxicity of high-
270. Koren G et al. Corticosteroids-salicy- 282. Kalantzis A et al. Oral effects of low- dose anthracycline chemotherapy?
late interaction in a case of juvenile dose methotrexate treatment. Oral Acta Oncol 1997; 36: 735–740.
rheumatoid arthritis. Ther Drug Surg Oral Med Oral Pathol Oral 294. Galetin A et al. Maximal inhibition
Monit 1987; 9: 177–179. Radiol Endod 2005; 100: 52–62. of intestinal first-pass metabolism as
271. Seidegard J. Reduction of the inhibi- 283. Troeltzsch M et al. Oral mucositis in a pragmatic indicator of intestinal
tory effect of ketoconazole on budes- patients receiving low-dose contribution to the drug-drug inter-
onide pharmacokinetics by methotrexate therapy for rheumatoid actions for CYP3A4 cleared drugs.
separation of their time of adminis- arthritis: report of 2 cases and litera- Curr Drug Metab 2007; 8: 685–693.
tration. Clin Pharmacol Ther 2000; ture review. Oral Surg Oral Med Oral 295. Yee GC, McGuire TR. Pharmacoki-
68: 13–17. Pathol Oral Radiol 2013; 115: e28– netic drug interactions with cyclos-
272. Raaska K et al. Plasma concentra- e33. porin (Part II). Clin Pharmacokinet
tions of inhaled budesonide and its 284. Fijlstra M et al. Reduced absorption 1990; 19: 400–415.
effects on plasma cortisol are of long-chain fatty acids during 296. Yee GC, McGuire TR. Pharmacoki-
increased by the cytochrome methotrexate-induced gastrointestinal netic drug interactions with cyclos-
P4503A4 inhibitor itraconazole. Clin mucositis in the rat. Clin Nutr 2013; porin (Part I). Clin Pharmacokinet
Pharmacol Ther 2002; 72: 362–369. 32: 452–459. 1990; 19: 319–332.
273. De Wachter E et al. Inhaled budes- 285. Chun JY et al. Adverse events associ- 297. Vermeire S et al. Effectiveness of con-
onide induced Cushing’s syndrome ated with azathioprine treatment in comitant immunosuppressive therapy
in cystic fibrosis patients, due to Korean pediatric inflammatory bowel in suppressing the formation of anti-
drug inhibition of cytochrome P450. disease patients. Pediatr Gastroenterol bodies to infliximab in Crohn’s dis-
J Cyst Fibros 2003; 2: 72–75. Hepatol Nutr 2013; 16: 171. ease. Gut 2007; 56: 1226–1231.
274. Gray D et al. Adrenal suppression 286. Mogensen S et al. Absorption of 298. Maini RN et al. Therapeutic efficacy
and Cushing’s syndrome secondary bupivacaine after administration of a of multiple intravenous infusions of
to ritonavir and budesonide. S Afr lozenge as topical treatment for pain anti-tumor necrosis factor? Mono-
Med J 2010; 100: 296. from oral mucositis. Basic Clin Phar- clonal antibody combined with low-
275. Orlicka K et al. Prevention of infec- macol Toxicol 2017; 120: 71–78. dose weekly methotrexate in rheuma-
tion caused by immunosuppressive 287. Parikh N et al. A single-dose phar- toid arthritis. Arthritis Rheum 1998;
drugs in gastroenterology. Ther Adv macokinetic study of fentanyl sublin- 41: 1552–1563.
Chronic Dis 2013; 4: 167–185. gual spray in cancer patients with 299. Havrda DE et al. A case report of
276. Zenlea T, Peppercorn MA. Immuno- and without oral mucositis. J Pain warfarin resistance due to azathio-
suppressive therapies for inflamma- 2013; 14: S73. prine and review of the literature.
tory bowel disease. World J 288. Amundsen R et al. Cyclosporine A- Pharmacotherapy 2001; 21: 355–357.
Gastroenterol 2014; 20: 3146–3152. and tacrolimus-mediated inhibition

© 2018 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 71 (2019), pp. 643–673 671
Pharmacokinetic interactions with GI drugs Chara Litou et al.

300. Joo Ng H, Crowther MA. Azathio- with cholestyramine. Br J Clin Phar- 323. al-Balla SR et al. The effects of
prine and inhibition of the anticoag- macol 1978; 5: 437–440. cholestyramine and colestipol on the
ulant effect of warfarin: evidence 311. Meinertz T et al. Interruption of absorption of diclofenac in man. Int
from a case report and a literature the enterohepatic circulation of J Clin Pharmacol Ther 1994; 32: 441–
review. Am J Geriatr Pharmacother phenprocoumon by cholestyramine. 445.
2006; 4: 75–77. Clin Pharmacol Ther 1977; 21: 731– 324. Weaver R, Jochemsen R. Nonclinical
301. Vazquez SR et al. Azathioprine- 735. pharmacokinetics and toxicokinetics.
induced warfarin resistance. Ann 312. Balmelli N et al. Fatal drug interac- In: Cartwright AC, Matthews BR,
Pharmacother 2008; 42: 1118–1123. tion between cholestyramine and eds. International Pharmaceutical
302. Scaldaferri F et al. Use and indica- phenprocoumon. Eur J Intern Med Product Registration, 2nd edn. Boca
tions of cholestyramine and bile acid 2002; 13: 210–211. Raton, FL: CRC Press, 2009: 336–
sequestrants. Intern Emerg Med 2013; 313. Malloy MJ et al. Influence of 376.
8: 205–210. cholestyramine resin administration 325. Caldwell JH, Greenberger NJ. Inter-
303. Joint Formulary Committee. on single dose sulindac pharmacoki- ruption of the enterohepatic circula-
Colestyramine. In: Joint Formulary netics. Int J Clin Pharmacol Ther tion of digitoxin by cholestyramine. J
Committee. British National Formu- 1994; 32: 286–289. Clin Invest 1971; 50: 2626–2637.
lary London: BMJ Group and Phar- 314. M€ uck W et al. Influence of 326. Malik MY et al. Role of enterohep-
maceutical Press [online] 2017. cholestyramine on the pharmacoki- atic recirculation in drug disposition:
Available at: https://bnf.nice.org.uk/d netics of cerivastatin. Int J Clin Phar- cooperation and complications. Drug
rug/colestyramine.html. Accessed macol Ther 1997; 35: 250–254. Metab Rev 2016; 48: 281–327.
June 26, 2017. 315. Kaykhaei MA et al. Low doses of 327. Stotzer P-O et al. Effect of
304. Bile acid malabsorption: colesevelam cholestyramine in the treatment of cholestyramine on gastrointestinal
| Guidance and guidelines | NICE. hyperthyroidism. Endocrine 2008; 34: transit in patients with idiopathic
Available at: https://www.nice.org.uk/ 52–55. bile acid diarrhea: a prospective,
advice/esuom22/chapter/Key-points- 316. Kivist€
o KT, Neuvonen PJ. The effect open-label study. Neuroenterology
from-the-evidence. Accessed Septem- of cholestyramine and activated char- 2013; 2: 5.
ber 28, 2017. coal on glipizide absorption. Br J 328. Donovan JM et al. Drug interactions
305. Wedlake L et al. Effectiveness and Clin Pharmacol 1990; 30: 733–736. with colesevelam hydrochloride, a
tolerability of colesevelam hydrochlo- 317. Bullingham RES et al. Clinical phar- novel, potent lipid-lowering agent.
ride for bile-acid malabsorption in macokinetics of mycophenolate Cardiovasc Drugs Ther 2000; 14: 681–
patients with cancer: a retrospective mofetil. Clin Pharmacokinet 1998; 34: 690.
chart review and patient question- 429–455. 329. Sinha V et al. Physiologically based
naire. Clin Ther 2009; 31: 2549–2558. 318. West RJ, Lloyd JK. The effect of pharmacokinetic modeling: from reg-
306. Odunsi-Shiyanbade ST et al. Effects cholestyramine on intestinal absorp- ulatory science to regulator policy.
of chenodeoxycholate and a bile acid tion. Gut 1975; 16: 93–98. Clin Pharmacol Ther 2014; 95: 478–
sequestrant, colesevelam, on intesti- 319. Malloy MJ et al. Effect of cholestyra- 480.
nal transit and bowel function. Clin mine resin on single dose valproate 330. Kesisoglou F et al. Physiologically
Gastroenterol Hepatol 2010; 8: 159– pharmacokinetics. Int J Clin Pharma- based absorption modeling to impact
165.e5. col Ther 1996; 34: 208–211. biopharmaceutics and formulation
307. Darkoh C et al. Bile acids improve 320. Zhu XX et al. Bile salt anion sorp- strategies in drug development—in-
the antimicrobial effect of rifaximin. tion by polymeric resins: comparison dustry case studies. J Pharm Sci
Antimicrob Agents Chemother 2010; of a functionalized polyacrylamide 2016; 105: 2723–2734.
54: 3618–3624. resin with cholestyramine. J Colloid 331. Duan P et al. Physiologically based
308. Young MA et al. Concomitant admin- Interface Sci 2000; 232: 282–288. pharmacokinetic (PBPK) modeling
istration of cholestyramine influences 321. He L et al. Lack of effect of coleseve- of pitavastatin and atorvastatin to
the absorption of troglitazone. Br J lam HCl on the single-dose pharma- predict drug-drug interactions
Clin Pharmacol 1998; 45: 37–40. cokinetics of aspirin, atenolol, (DDIs). Eur J Drug Metab Pharma-
309. Neuvonen PJ et al. Effects of resins enalapril, phenytoin, rosiglitazone, cokinet 2017; 42: 689–705.
and activated charcoal on the and sitagliptin. Diabetes Res Clin 332. Chen Y et al. Development of a
absorption of digoxin, carba- Pract 2014; 104: 401–409. physiologically based pharmacoki-
mazepine and frusemide. Br J Clin 322. al-Meshal MA et al. The effect of netic model for itraconazole pharma-
Pharmacol 1988; 25: 229–233. colestipol and cholestyramine on cokinetics and drug-drug interaction
310. J€ahnchen E et al. Enhanced elimina- ibuprofen bioavailability in man. Bio- prediction. Clin Pharmacokinet 2016;
tion of warfarin during treatment pharm Drug Dispos 1994; 15: 463–471. 55: 735–749.

672 © 2018 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 71 (2019), pp. 643–673
Chara Litou et al. Pharmacokinetic interactions with GI drugs

333. Min JS et al. Application of physio- 337. Cristofoletti R et al. Assessment of of salts of weak bases in the fasted
logically based pharmacokinetic bioequivalence of weak base formula- upper gastrointestinal lumen: data in
modeling in predicting drug-drug tions under various dosing condi- biorelevant media and in human
interactions for sarpogrelate tions using physiologically based aspirates. Eur J Pharm Biopharm
hydrochloride in humans. Drug Des pharmacokinetic simulations in vir- 2017; 115: 94–101.
Devel Ther 2016; 10: 2959–2972. tual populations. Case examples: 341. Lee HT et al. Effect of prokinetic
334. Grillo JA et al. Utility of a physiolog- ketoconazole and posaconazole. J agents, cisapride and metoclo-
ically-based pharmacokinetic (PBPK) Pharm Sci 2017; 106: 560–569. pramide, on the bioavailability in
modeling approach to quantitatively 338. Doki K et al. Virtual bioequivalence humans and intestinal permeability
predict a complex drug-drug-disease for achlorhydric subjects: the use of in rats of ranitidine, and intestinal
interaction scenario for rivaroxaban PBPK modelling to assess the formu- charcoal transit in rats. Res Commun
during the drug review process: lation-dependent effect of achlorhy- Mol Pathol Pharmacol 2000; 108:
implications for clinical practice. Bio- dria. Eur J Pharm Sci 2017; 109: 311–323.
pharm Drug Dispos 2012; 33: 99–110. 111–120. 342. Bustos D et al. Effect of loperamide
335. Mitra A et al. Using absorption simu- 339. Establishing bioequivalence in virtual and bisacodyl on intestinal transit
lation and gastric pH modulated dog space: are we really there? | AAPS time, fecal weight and short chain
model for formulation development Blog. Available at: https://aapsblog.aa fatty acid excretion in the rat. Acta
to overcome achlorhydria effect. Mol ps.org/2016/09/29/establishing-bioe Gastroenterol Latinoam 1991; 21: 3–9.
Pharm 2011; 8: 2216–2223. quivalence-in-virtual-space-are-we- 343. Joo JS et al. Alterations in colonic
336. Qi F et al. Influence of different pro- really-there/. Accessed January 25, anatomy induced by chronic stimu-
ton pump inhibitors on the pharma- 2018. lant laxatives: the cathartic colon
cokinetics of voriconazole. Int J 340. Litou C et al. The impact of reduced revisited. J Clin Gastroenterol 1998;
Antimicrob Agents 2017; 49: 403–409. gastric acid secretion on dissolution 26: 283–286.

© 2018 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 71 (2019), pp. 643–673 673

You might also like