ArticlePDF Available

Oral co-administration of elacridar and ritonavir enhances plasma levels of oral paclitaxel and docetaxel without affecting relative brain accumulation

Authors:
  • Duajonessenhuis

Abstract and Figures

Background: The intestinal uptake of the taxanes paclitaxel and docetaxel is seriously hampered by drug efflux through P-glycoprotein (P-gp) and drug metabolism via cytochrome P450 (CYP) 3A. The resulting low oral bioavailability can be boosted by co-administration of P-gp or CYP3A4 inhibitors. Methods: Paclitaxel or docetaxel (10 mg/kg) was administered to CYP3A4-humanised mice after administration of the P-gp inhibitor elacridar (25 mg kg−1) and the CYP3A inhibitor ritonavir (12.5 mg kg−1). Plasma and brain concentrations of the taxanes were measured. Results: Oral co-administration of the taxanes with elacridar increased plasma concentrations of paclitaxel (10.7-fold, P<0.001) and docetaxel (four-fold, P<0.001). Co-administration with ritonavir resulted in 2.5-fold (paclitaxel, P<0.001) and 7.3-fold (docetaxel, P<0.001) increases in plasma concentrations. Co-administration with both inhibitors simultaneously resulted in further increased plasma concentrations of paclitaxel (31.9-fold, P<0.001) and docetaxel (37.4-fold, P<0.001). Although boosting of orally applied taxanes with elacridar and ritonavir potentially increases brain accumulation of taxanes, we found that only brain concentrations, but not brain-to-plasma ratios, were increased after co-administration with both inhibitors. Conclusions: The oral availability of taxanes can be enhanced by co-administration with oral elacridar and ritonavir, without increasing the brain penetration of the taxanes.
Content may be subject to copyright.
Oral co-administration of elacridar and
ritonavir enhances plasma levels of oral
paclitaxel and docetaxel without affecting
relative brain accumulation
J J M A Hendrikx*
,1,2
, J S Lagas
1
, E Wagenaar
2
, H Rosing
1
, J H M Schellens
3,4
, J H Beijnen
1,4
and A H Schinkel
2
1
Department of Pharmacy and Pharmacology, Slotervaart Hospital, PO 90440, 1006 BK Amsterdam, The Netherlands;
2
Division of
Molecular Oncology, The Netherlands Cancer Institute, PO 90203, 1006 BE Amsterdam, The Netherlands;
3
Department of Clinical
Pharmacology, The Netherlands Cancer Institute, PO 90203, 1006 BE Amsterdam, The Netherlands and
4
Department of
Pharmaceutical Sciences, Utrecht University, PO 80082, 3508 TB Utrecht, The Netherlands
Background: The intestinal uptake of the taxanes paclitaxel and docetaxel is seriously hampered by drug efflux through
P-glycoprotein (P-gp) and drug metabolism via cytochrome P450 (CYP) 3A. The resulting low oral bioavailability can be boosted by
co-administration of P-gp or CYP3A4 inhibitors.
Methods: Paclitaxel or docetaxel (10 mg/kg) was administered to CYP3A4-humanised mice after administration of the P-gp
inhibitor elacridar (25 mg kg
1
) and the CYP3A inhibitor ritonavir (12.5 mg kg
1
). Plasma and brain concentrations of the taxanes
were measured.
Results: Oral co-administration of the taxanes with elacridar increased plasma concentrations of paclitaxel (10.7-fold, Po0.001)
and docetaxel (four-fold, Po0.001). Co-administration with ritonavir resulted in 2.5-fold (paclitaxel, Po0.001) and 7.3-fold
(docetaxel, Po0.001) increases in plasma concentrations. Co-administration with both inhibitors simultaneously resulted in further
increased plasma concentrations of paclitaxel (31.9-fold, Po0.001) and docetaxel (37.4-fold, Po0.001). Although boosting of orally
applied taxanes with elacridar and ritonavir potentially increases brain accumulation of taxanes, we found that only brain
concentrations, but not brain-to-plasma ratios, were increased after co-administration with both inhibitors.
Conclusions: The oral availability of taxanes can be enhanced by co-administration with oral elacridar and ritonavir, without
increasing the brain penetration of the taxanes.
The taxane anticancer agents paclitaxel (Taxol) and docetaxel
(Taxotere) share the baccatin core ring structure (Vaishampayan
et al, 1999). Currently, paclitaxel and docetaxel are routinely
applied intravenously (i.v.) to treat several types of cancer, such as
non-small cell lung cancer, ovarian, breast, gastric, prostate and
head-and-neck cancer (Gligorov and Lotz, 2004; Koolen et al, 2010a).
The development of oral formulations of paclitaxel and
docetaxel is the focus of preclinical and clinical research in our
groups because oral administration has many advantages over i.v.
administration (Schellens et al, 2000; Koolen et al, 2010a). Oral
administration is more practical and convenient for patients as oral
medication can be taken by the patient at home, whereas
i.v. administration requires hospitalisation during infusion. Oral
administration in the home situation also reduces treatment cost.
Moreover, oral administration enables other dosing schedules such
as metronomic therapy (e.g., continuous or frequent treatment
*Correspondence: Dr JJMA Hendrikx; E-mail: Jeroen.Hendrikx@slz.nl
Revised 19 March 2014; accepted 27 March 2014; published online 29 April 2014
&2014 Cancer Research UK. All rights reserved 0007 0920/14
FULL PAPER
Keywords: P-glycoprotein (P-gp/MDR1); cytochrome P450 3A (CYP3A4); paclitaxel; docetaxel; oral bioavailability
British Journal of Cancer (2014) 110, 2669–2676 | doi: 10.1038/bjc.2014.222
www.bjcancer.com | DOI:10.1038/bjc.2014.222 2669
with low doses of anticancer drugs), which can increase efficacy of
taxane treatment and reduce adverse effects caused by high plasma
concentrations of docetaxel or paclitaxel (Jiang et al, 2010; Wu
et al, 2011).
A major limitation in the concept of oral administration of
taxanes is, however, the low oral availability of paclitaxel and
docetaxel (Schellens et al, 2000; Koolen et al, 2010a). Paclitaxel and
docetaxel have poor aqueous solubility and upon oral administra-
tion, intestinal uptake is seriously hampered by drug efflux through
P-glycoprotein (P-gp/MDR1/ABCB1), and systemic exposure is
further limited by drug metabolism via cytochrome P450 (CYP) 3A
(Sparreboom et al, 1997; van Asperen et al, 1997; Meerum Terwogt
et al, 1998; van Asperen et al, 1998; Bardelmeijer et al, 2004; Lagas
et al, 2006; van Waterschoot et al, 2009; Hendrikx et al, 2013).
P-glycoprotein is a member of the ATP-binding cassette efflux
transporter family and is expressed in multiple tissues such as
intestine, liver and kidney, but also at the blood–brain barrier
(BBB) (Gottesman and Ambudkar, 2001). P-glycoprotein-
mediated transport limits drug absorption across intestinal cells
and brain penetration across the BBB. In enterocytes, P-gp pumps
back absorbed taxanes into the intestinal lumen, whereas at the
BBB, taxanes are pumped back into the systemic circulation. In
liver and kidney, P-gp increases drug excretion by active efflux
transport into the bile and urine (Glaeser and Fromm, 2008).
CYP3A is a member of the CYP superfamily and CYP enzymes
are responsible for most phase-I drug metabolism (Thelen and
Dressman, 2009). CYP enzymes are mainly expressed in the liver,
but some CYP members are also expressed in enterocytes. CYP3A
is the most abundant CYP enzyme in liver and intestine,
representing 40% and 80% of the total CYP enzymes expressed
in each tissue, respectively (Paine et al, 2006). Docetaxel is
primarily metabolised by enzymes of the CYP3A subfamily,
whereas paclitaxel is metabolised by both CYP3A4 and CYP2C8
(Vaishampayan et al, 1999). In contrast to CYP3A, CYP2C8 is only
expressed in liver cells (Paine et al, 2006).
Although docetaxel is a good P-gp substrate, transport of
paclitaxel by P-gp is even more efficient. In addition, paclitaxel
metabolism is not solely CYP3A dependent. Therefore, it was
assumed that oral bioavailability of paclitaxel was primarily limited
by P-gp, and that of docetaxel primarily by CYP3A. However, in
mice, complete vs single knockout of Mdr1a/b and/or Cyp3a genes
resulted in further increased plasma exposure of paclitaxel and
docetaxel alike after oral administration, suggesting that both
systems are important for oral availability of both taxanes
(van Waterschoot et al, 2009; Hendrikx et al, 2013). The importance
of CYP3A4 for paclitaxel metabolism was further supported by our
finding that human CYP3A4 metabolises paclitaxel far more
efficiently than the mouse Cyp3a enzymes (Hendrikx et al, 2013).
Therefore, a promising strategy to boost the oral availability of
these taxanes is combining oral formulations of paclitaxel or
docetaxel with inhibitors of both P-gp and CYP3A4. In (pre)
clinical studies, it has already been demonstrated that the area
under the plasma concentration–time curves (AUC) after oral
administration of paclitaxel and docetaxel can be strongly
enhanced in both mice and humans by co-administration of the
potent CYP3A4 inhibitor ritonavir (Bardelmeijer et al, 2002;
Oostendorp et al, 2009; Koolen, 2011; Hendrikx et al, 2013). In
addition, co-administration of the potent P-gp inhibitor elacridar
results in increased oral plasma AUC of paclitaxel in mice and
humans (Bardelmeijer et al, 2000; Malingre et al, 2001).
There are potential risks involved when the oral bioavailability
of paclitaxel and docetaxel is increased by inhibition of metabolising
enzymes and drug transporters. For instance, co-administration of
oral elacridar in mice resulted in increased brain penetration of i.v.
administered paclitaxel by inhibition of P-gp at the BBB (Kemper
et al, 2003). Therefore, boosting oral uptake of taxanes using an
oral P-gp inhibitor might increase the relative risk of CNS toxicity.
Furthermore, oral administration of docetaxel or paclitaxel to mice
lacking both P-gp and Cyp3a resulted in substantially higher
plasma levels than administration of the taxanes to mice lacking
either P-gp or Cyp3a alone. Simultaneous inhibition of P-gp and
CYP3A by drugs that are co-administered with orally administered
taxanes may lead to further increased plasma levels of the taxanes
and therefore an increased risk of side effects and toxicity in
clinical practice.
In the present preclinical study, we examined whether we could
substantially increase the oral availability of taxanes by simulta-
neous inhibition of P-gp and CYP3A using oral co-administration
of elacridar and ritonavir, and to what extent this would affect P-gp
transport at the BBB.
MATERIALS AND METHODS
Drugs and chemicals. Paclitaxel, docetaxel, elacridar HCl and
ritonavir were purchased from Sequoia Research Products (Oxford,
UK). Drug-free lithium-heparinised human plasma was obtained
from Bioreclamation LLC (New York, NY, USA). All other
chemicals were of analytical grade and obtained from commercial
sources.
Animals. In compliance with Dutch legislation, mice were housed
and handled according to institutional guidelines, and approval of
the local (NKI) animal care and use committee was obtained before
the start of experiments. Mice were kept in a temperature-
controlled environment with a 12-h light/12-h dark cycle and
received a standard diet (AM-II, Hope Farms, Woerden, The
Netherlands) and acidified water ad libitum. In this study, Cyp3a
knockout mice with specific expression of human CYP3A4 in liver
and intestine (Cyp3a
/
Tg-3A4
Hep/Int
) were used (van Herwaarden
et al, 2007). The strain had a 499% FVB genetic background.
Cyp3a
/
Tg-3A4
Hep/Int
mice were used as there is a species
difference for paclitaxel in CYP3A substrate specificity or enzyme
activity between endogenous murine Cyp3a and human CYP3A4
(Hendrikx et al, 2013). Experiments comparing paclitaxel PK in
wild-type mice may therefore underestimate the impact of human
CYP3A on paclitaxel pharmacokinetics in patients. The difference
between species is minimised using Cyp3a
/
Tg-3A4
Hep/Int
mice.
A basic difference in docetaxel metabolite formation was not observed
between human, wild-type mice and Cyp3a
/
Tg-3A4
Hep/Int
mice
(Hendrikx et al, 2013). In all experiments, male mice of 9–14 weeks
of age were used.
In vivo analysis of plasma pharmacokinetics. Before the experi-
ments, stock solutions containing 6 mg ml
1
paclitaxel, 6 mg ml
1
docetaxel, 15 mg ml
1
elacridar HCl, 7.5 mg ml
1
ritonavir or
15 mg ml
1
elacridar HCl and 7.5 mg ml
1
ritonavir in ethanol:-
polysorbate 80 (1 : 1, v/v) were made and stored at 20 1C. On the
day of the experiments, stock solutions were diluted with water
(1 : 5, v/v) to obtain solutions for administration. Animals were
fasted 2 h before oral drug administration to minimise variation in
absorption. Paclitaxel and docetaxel were administered orally at a
dose of 10 mg kg
1
of bodyweight, ritonavir was administered
orally at a dose of 12.5 mg kg
1
of bodyweight, and elacridar was
administered orally at a dose of 25 mg kg
1
of bodyweight. Oral
administration was performed by gavage into the stomach using a
blunt-ended needle. In case of co-administration with ritonavir,
elacridar or ritonavir and elacridar, the booster(s) were orally
administered 15 min before oral taxane administration.
Sample collection. For determining plasma pharmacokinetics,
multiple blood samples (B50 ml) were collected from the tail vein
at 15 min and at 1, 2, 4, 8 and 24h using heparinised capillary tubes
(Oxford Labware, St Louis, MO, USA). All time point samples were
derived from the same mouse. At the last time point of sequential
BRITISH JOURNAL OF CANCER Elacridar and ritonavir boost oral taxanes
2670 www.bjcancer.com | DOI:10.1038/bjc.2014.222
sampling (48 h), blood was taken by cardiac puncture. Blood
samples were centrifuged at ambient temperature at 8000 gfor
5 min and subsequently plasma was collected. All samples were
stored at 20 1C until analysis.
For brain accumulation studies, blood samples at 2 h were taken
by cardiac puncture and brain tissue was isolated. Blood samples
were centrifuged at ambient temperature at 8000 gfor 5 min and
subsequently plasma was collected. Brain tissue was homogenised
in 1% bovine serum albumin. All samples were stored at 20 1C
until analysis. Brain-to-plasma ratios at t¼2 h were calculated per
mouse by dividing the brain concentration by the corresponding
plasma concentration.
Bioanalytical analysis. Previously developed liquid chromato-
graphy assays coupled with tandem mass spectrometry detection
(LC-MS/MS) were used to quantify paclitaxel (Stokvis et al, 2004)
and docetaxel (Kuppens et al, 2005). Labelled structure analogues
were used as internal standards. In summary, mouse plasma
samples of 20 ml were diluted with 180 ml of human plasma.
Human plasma was used for dilution of the samples as the
concentrations in the undiluted mouse plasma were outside the
calibration range, and also to mimic the calibration standards that
were in human plasma. Brain samples were not diluted as
concentrations were too low to quantify after dilution in some
samples. To 200 ml of diluted plasma sample or homogenised brain
sample, 25 ml of internal standard working solution was added.
Subsequently, the samples were mixed briefly, tertiary-butyl methyl
ether was added and the samples were shaken for 10 min at 1250
r.p.m. The samples were centrifuged at 23 000 g, snap-frozen and
the organic layer was collected. After evaporation of the organic
layer, the samples were reconstituted with reconstitution solvent
and an aliquot was injected into the LC-MS/MS system.
Calibration standards in human plasma in a range of 0.25–
1000 ng ml
1
or 0.25–500 ng ml
1
were used for quantification of
paclitaxel and docetaxel, respectively.
Pharmacokinetic calculations and statistical analysis. Pharmaco-
kinetic parameters, including the AUCs, were calculated using
the software package PK Solutions 2.0.2 (SUMMIT, Research
Services, Ashland, OH, USA). The AUC
0–last time point
was
calculated by trapezoid calculation using observed data points.
The total AUC extrapolated to infinity (AUC
0–inf
) was computed
by combining AUC
0–last time point
with an extrapolated value. One-
way ANOVA was used when multiple groups were compared and
the Bonferroni post hoc correction was used to accommodate
multiple testing. The two-sided unpaired Student’s t-test was used
when treatments or differences between two groups were
compared. Data that did not show normal distribution were log-
transformed to normalise the distribution of the data sets and
enable statistical comparison. The Kolmogorov–Smirnov test was
used to test for normal distribution. During all statistical analyses,
differences in group sizes were considered in the calculations.
Differences were considered statistically significant when Po0.05.
All data are presented as geometric mean±s.d.
Addition of previously reported data. Previously, we published
AUCs of paclitaxel after oral administration of 10 mg kg
1
paclitaxel with and without 12.5 mg kg
1
ritonavir to Cyp3a
/
Tg-3A4
Hep/Int
mice (five and seven animals were used, respectively)
(Hendrikx et al, 2013). These data were compared with
plasma concentrations after oral administration of 10 mg kg
1
paclitaxel with and without 12.5 mg kg
1
ritonavir obtained in
this study (six and four animals were used, respectively).
Previously obtained results were not statistically different from
the results in the present study (data not shown). Therefore,
these results were also used to decrease the number of animals
neededforthisstudy.
Comparison with previously reported data in knockout mice.
To estimate the extent of P-gp inhibition by elacridar and Cyp3a
inhibition by ritonavir, plasma exposure after chemical inhibition
was compared with plasma exposure after complete knockout of
P-gp or Cyp3A. Previously reported plasma AUCs
0–inf
after oral
administration of 10 mg kg
1
paclitaxel (Hendrikx et al, 2013) or
10 mg kg
1
docetaxel (van Waterschoot et al, 2009) to mice
lacking P-gp, Cyp3a or both were compared with AUCs
0–inf
after
chemical inhibition as obtained in this study. All plasma AUCs
were normalised for their matching control group, and these
relative plasma AUCs were used for comparison.
RESULTS
Paclitaxel exposure after oral co-administration with elacridar
and/or ritonavir. To study the effect of the P-gp inhibitor
elacridar and the CYP3A inhibitor ritonavir on oral bioavailability
of paclitaxel, we orally administered 10 mg kg
1
paclitaxel to the
CYP3A4-humanised Cyp3a
/
Tg-3A4
Hep/Int
mice and combined
paclitaxel administration with 25 mg kg
1
elacridar and/or
12.5 mg kg
1
ritonavir.
Oral co-administration of paclitaxel and elacridar or paclitaxel
and ritonavir resulted in increased plasma concentrations of
paclitaxel (Figure 1). The area under the plasma concentration–
time curve from 0 extrapolated to infinity (AUC
0–inf
) was 10.7-fold
higher after co-administration with elacridar than after single
paclitaxel administration (Po0.001). These results in humanised
mice are in line with the previously observed 6.6-fold increase in
paclitaxel AUC after oral co-administration of paclitaxel and
elacridar at the same dose to wild-type mice (Bardelmeijer et al,
2000). Co-administration of paclitaxel and ritonavir resulted in an
increase in the AUC
0–inf
of 2.5-fold (Po0.001). However, this
boosting effect with ritonavir was clearly less than that of elacridar
co-administration. Co-administration of paclitaxel with both
elacridar and ritonavir together resulted in further increased
plasma concentrations of paclitaxel (31.9-fold higher than single
paclitaxel administration; Po0.001). The increases in oral
AUC
0–inf
of paclitaxel after chemical inhibition with elacridar or
ritonavir, alone or in combination, were comparable to the
increases in oral AUC
0–inf
after complete genetic knockout of
P-gp or Cyp3a, alone or in combination (Table 1). These data
suggest that virtually complete inhibition of both P-gp and
CYP3A4 (intestinal and hepatic) was achieved with the combina-
tion elacridar and ritonavir.
Docetaxel exposure after oral co-administration with elacridar
and/or ritonavir. Parallel to the paclitaxel experiments, we
studied the effect of elacridar and/or ritonavir co-administration
on the oral bioavailability of docetaxel. In the CYP3A4-humanised
mouse model, we observed a 7.3-fold increase in AUC
0–inf
after
oral administration of docetaxel and ritonavir when compared with
AUC
0–inf
after single docetaxel administration (Po0.001;
Figure 2). Oral co-administration of docetaxel and elacridar
resulted in a four-fold increase compared with single docetaxel
administration (Po0.001). The AUC
0–inf
of docetaxel after
boosting with elacridar was not significantly different from the
AUC
0–inf
after boosting with ritonavir (P40.05). As observed for
paclitaxel, co-administration of docetaxel with both elacridar and
ritonavir resulted in a further increase in AUC
0–inf
(37.4-fold
higher than single docetaxel administration; Po0.001). The
increase in oral AUC
0–inf
of docetaxel after chemical inhibition
with elacridar was comparable to the increase in oral AUC
0–inf
after complete genetic knockout of P-gp. However, the increase
after chemical inhibition with ritonavir was modestly, but
significantly (Po0.01), lower than after complete genetic knockout
of Cyp3a, and the same was true for combined CYP3A4 and P-gp
Elacridar and ritonavir boost oral taxanes BRITISH JOURNAL OF CANCER
www.bjcancer.com | DOI:10.1038/bjc.2014.222 2671
inhibition compared with full Cyp3a and P-gp knockout
(Po0.001; Table 1). These data suggest that for docetaxel in the
transgenic mice, the inhibition of intestinal and hepatic CYP3A4
by ritonavir was not entirely complete.
Brain concentrations of taxanes after oral co-administration
with elacridar and/or ritonavir. As brain accumulation could
potentially be increased after boosting of oral taxanes with a P-gp
inhibitor, we measured brain concentrations 2 h after oral
administration of paclitaxel or docetaxel, that is, roughly around
the plasma t
max
. Two effects might occur: first, increased taxane
brain concentrations simply as a consequence of the higher plasma
levels of the taxanes; and second, a further, disproportionate
increase in brain concentration relative to the plasma levels
because of inhibition of P-gp at the BBB, and/or possibly saturation
of P-gp activity at the BBB because of the much higher plasma
taxane levels. The second effects could result in poorly predictable
alterations in CNS toxicity of the taxanes. As these effects are most
likely to occur when plasma levels of both taxanes and inhibitors
are high, we chose the 2-h time point for sampling. Maximum
plasma concentrations of docetaxel and paclitaxel are reached at
2–4 h after oral administration. We did not measure plasma
concentrations of the inhibitors in this experiment, but maximum
plasma concentrations are reached in wild-type mice around 2 h
after oral administration of elacridar (Bardelmeijer et al, 2000) or
ritonavir (Supplementary Figure 1).
Brain concentrations of paclitaxel were significantly increased
after co-administration with elacridar (Po0.01 vs single paclitaxel
administration), but not after co-administration with ritonavir
(P40.05; Figure 3B). Co-administration of paclitaxel with both
Table 1. Area under the plasma concentration–time curve of paclitaxel and docetaxel after oral administration of 10 mgkg
1
paclitaxel or 10 mg kg
1
docetaxel in Cyp3a
/
Tg-3A4
Hep/Int
mice
Control group
a
P-gp inhib./KO CYP3A inhib./KO CYP3A and P-gp inhib./KO
Oral paclitaxel
AUC
0–inf
(ng hml
1
) after inhibition 314±74 3373±725 780±412 10002±2652
Fold vs control 1 10.7 2.5 31.9
Number of animals 11 8 11 7
AUC
0–inf
(ng hml
1
) in KO mice 320±224 3954±825 471±174 8830±1999
Fold vs control 1 12.4 1.5 27.6
Number of animals 10 5 9 5
Oral docetaxel
AUC
0–inf
(ng hml
1
) after inhibition 157±67 626±182 1146±281 5869±2520
Fold vs control 1 4.0 7.3 37.4
Number of animals 5 5 5 5
AUC
0–inf
(ng hml
1
) in KO mice 228±130 645±272 2627±1011 16466±2020
Fold vs control 1 2.8 11.5 72.2
Number of animals 6 6 6 7
Abbreviations: AUC
0–inf
¼area under the plasma concentration–time curve from 0 extrapolated to infinity; CYP3A ¼cytochrome P450 3A; Cyp3a
/
Tg-3A4
Hep/Int
¼Cyp3a KO mice with specific
expression of human CYP3A4 in liver and intestine; inhib.¼inhibition; KO¼knockout; P-gp¼P-glycoprotein (MDR1;ABCB1). Values represent the mean±s.d. Animals (5–11) per group were
used. Both drugs were administered as a single dose or co-administered with an oral dose of the CYP3A4 inhibitor ritonavir (12.5 mgkg
1
), the P-gp inhibitor elacridar (25mg kg
1
) or both.
Data are compared with previously reported data after oral administration of paclitaxel (Hendrikx et al, 2013) or docetaxel (van Waterschoot et al, 2009) to wild-type mice, P-gp KO
(Mdr1a/b
/
), Cyp3a KO (Cyp3a
/
) and combined P-gp and Cyp3a KO mice (Cyp3a/Mdr1a/b
/
).
a
When murine P-gp and human CYP3A are inhibited, the control group reflects single drug administration in Cyp3a
/
Tg-3A4
Hep/Int
mice. When murine P-gp and murine Cyp3a are knocked
out, the control group reflects single drug administration in wild-type mice.
10 000
AB
15 000
10 000
5000
AUC0–inf (ng×h ml–1)
0
PAC
PAC+ELC
PAC+RTV
PAC+ELC+RTV
PAC
PAC+ELC
PAC+RTV
PAC+ELC+RTV
1000
Paclitaxel (ng ml–1)
100
10
1
0 10203040
Time (h)
Figure 1. Panel Ashows plasma concentration–time curves in Cyp3a
/
mice expressing human CYP3A4 in liver and intestine (Cyp3a
/
Tg-3A4
Hep/Int
) after oral administration of 10 mg kg
1
paclitaxel. Paclitaxel was administered alone or co-administered with 25mgkg
1
oral
elacridar, 12.5 mg kg
1
oral ritonavir or both elacridar and ritonavir. Panel Bshows the area under the plasma concentration–time curves from 0
extrapolated to infinity (AUC
0–inf
). Data are presented as individual data points and lines represent the mean. Differences in AUC
0–inf
between all
groups were statistically significantly different (Po0.001), unless stated otherwise (NS, P40.05). Values represent the means±s.d. In all, 8–11
animals per group were used. Abbreviations: ELC ¼elacridar; PAC¼paclitaxel; RTV ¼ritonavir.
BRITISH JOURNAL OF CANCER Elacridar and ritonavir boost oral taxanes
2672 www.bjcancer.com | DOI:10.1038/bjc.2014.222
elacridar and ritonavir resulted in a similar increase in brain
concentrations as after co-administration of paclitaxel and
elacridar (P40.05 vs paclitaxel and elacridar administration;
Po0.001 vs single paclitaxel administration). However, correcting
for the increased plasma levels after boosting (Figure 3A), brain-
to-plasma ratios were not statistically different between the groups
(P40.05 for all comparisons; Figure 3C). These data suggest that
the relative brain accumulation of paclitaxel was not altered by
elacridar and ritonavir co-administration, despite the substantially
increased plasma levels of paclitaxel and the circulating elacridar
levels.
Co-administration with elacridar also increased docetaxel brain
concentrations (Po0.01 vs single docetaxel administration;
Figure 4B). In contrast to paclitaxel brain concentrations, docetaxel
brain concentrations were substantially increased after co-admin-
istration with ritonavir to comparable levels as seen after co-
administration with elacridar (P40.05 vs docetaxel and elacridar
administration; Po0.001 vs single docetaxel administration), thus
more or less following the pattern of effects of the inhibitors on
docetaxel plasma concentrations. Brain concentrations of docetaxel
were further increased after co-administration with both ritonavir
and elacridar (Po0.001 vs single docetaxel administration).
However, the increase in docetaxel brain concentrations was
primarily caused by the increased plasma concentrations after
boosting (Figure 4A), as brain-to-plasma ratios were not
statistically significantly different between any of the treatment
groups (P40.05 for all comparisons; Figure 4C).
DISCUSSION
Our data with CYP3A4-humanised mice show that it is possible to
markedly enhance the plasma AUC of oral paclitaxel and docetaxel
(30- to 40-fold) by orally co-administering elacridar and ritonavir.
Each inhibitor contributed substantially to the overall AUC
increase, although the contribution of elacridar was stronger for
paclitaxel and that of ritonavir for docetaxel. Yet, at the same time,
the relative brain accumulation of the taxanes (corrected for the
increased plasma levels) was not increased. This indicates that
neither the circulating elacridar levels, nor the increased plasma
taxane levels were sufficient to substantially inhibit or saturate the
1500
ABC
80 0.4
0.3
0.2
0.1
0.0
Brain/plasma ratio
NS
**
NS
NS NS
NS
60
40
20
0
Paclitaxel plasma
concentration (ng ml–1)
Paclitaxel brain
concentration (ng g–1)
1000
500
0
PAC
PAC+ELC
PAC+RTV
PAC+ELC+RTV
PAC
PAC+ELC
PAC + R T V
PAC +ELC+RTV
PAC
PAC+ELC
PAC + R T V
PAC +ELC+RTV
Figure 3. Plasma and brain concentrations of paclitaxel in Cyp3a
/
mice expressing human CYP3A4 in liver and intestine 2 h after oral
administration of 10 mg kg
1
paclitaxel. Paclitaxel was administered alone or co-administered with 25 mg kg
1
oral elacridar, 12.5 mg kg
1
oral
ritonavir or both elacridar and ritonavir. Panels reflect plasma concentrations (panel A), brain concentrations (panel B) or brain-to-plasma ratios
(panel C). Data are presented as individual data points and lines represent the mean. Differences in plasma or brain concentrations between
groups were statistically significantly different (Po0.001), unless stated otherwise (NS: not significant, P40.05 or **Po0.01). Differences in brain-
to-plasma ratios between all groups were not statistically significant. Values represent the means±s.d. Five animals per group were used.
Abbreviations: ELC ¼elacridar; PAC¼paclitaxel; RTV ¼ritonavir.
10 000
AB
10 000
8000
AUC0–inf (ng×h ml–1)
6000
4000
2000
0
DOC
DOC+ELC
DOC+RTV
DOC+ELC+RTV
DOC
DOC+ELC
DOC+RTV
DOC+ELC+RTV
1000
100
Docetaxel (ng ml–1)
10
1
0102030
Time (h)
40
Figure 2. Panel Ashows plasma concentration–time curves in Cyp3a
/
mice expressing human CYP3A4 in liver and intestine (Cyp3a
/
Tg-3A4
Hep/Int
) after oral administration of 10 mg kg
1
docetaxel. Docetaxel was administered alone or co-administered with 25mg kg
1
oral
elacridar, 12.5 mg kg
1
oral ritonavir or both elacridar and ritonavir. Panel Bshows the area under the plasma concentration–time curves from 0
extrapolated to infinity (AUC
0–inf
). Data are presented as individual data points and lines represent the mean. Differences in AUC
0–inf
between all
groups were statistically significantly different (Po0.001), unless stated otherwise (NS, P40.05). Values represent the means±s.d. Five animals per
group were used. Abbreviations: ELC ¼elacridar; PAC ¼paclitaxel; RTV ¼ritonavir.
Elacridar and ritonavir boost oral taxanes BRITISH JOURNAL OF CANCER
www.bjcancer.com | DOI:10.1038/bjc.2014.222 2673
taxane export activity at the BBB. These data suggest that it may be
possible to greatly enhance the oral availability of taxanes in
patients by co-administration with oral elacridar and ritonavir,
without invoking the risk of increased CNS toxicity of the
taxanes.
To estimate the extent of P-gp inhibition by elacridar and
CYP3A4 inhibition by ritonavir, plasma exposures after chemical
inhibition were compared with plasma exposures after complete
knockout of P-gp and/or Cyp3a (Table 1). Plasma AUCs
0–inf
of
paclitaxel and docetaxel were comparable after complete knockout
of P-gp and after chemical inhibition of P-gp by elacridar. This
suggests that the intestinal and hepatic inhibition of P-gp was
complete at the used dose of elacridar.
The plasma AUCs
0–inf
of paclitaxel were similar after complete
gene knockout of Cyp3a and inhibition of CYP3A4 by ritonavir,
but the plasma AUC
0–inf
of docetaxel was slightly lower after
ritonavir inhibition than upon Cyp3a knockout. The difference is
not substantial (Table 1), but it can probably be attributed to
incomplete inhibition of human CYP3A4 in the CYP3A4-
humanised mice at later time points, as the ritonavir concentra-
tions likely drop considerably after a few hours. Although not
tested in these mice, ritonavir levels in plasma of wild-type mice
receiving 12.5 mg kg
1
oral ritonavir drop substantially after 2 h
(Supplementary Figure 1), which may result in incomplete
CYP3A4 inhibition at later time points. The effect may be more
obvious for docetaxel than for paclitaxel because docetaxel
metabolism is more strongly CYP3A dependent (van Waterschoot
et al, 2009).
Previously reported data showed that complete knockout of
both P-gp and Cyp3a resulted in higher plasma concentrations of
orally applied taxanes than single knockout of P-gp or Cyp3a (van
Waterschoot et al, 2009; Hendrikx et al, 2013). We show here that
chemical inhibition of P-gp and CYP3A4 with both elacridar and
ritonavir likewise further increased plasma concentrations of orally
applied taxanes. In humans, oral formulations of paclitaxel and
docetaxel were thus far tested with only one of these boosters. In
patients, oral availability of paclitaxel is boosted by elacridar (Malingre
et al, 2001) or ritonavir (Koolen, 2011) and oral availability of
docetaxel is boosted by ritonavir (Oostendorp et al, 2009; Marchetti
et al, 2012). Our results suggest that boosting of orally
applied taxanes by both elacridar and ritonavir might
further increase plasma exposure of taxanes in patients.
Moreover, combined inhibition of P-gp and CYP3A4 may
result in decreased interpatient and intrapatient variability in
oral taxane pharmacokinetics. Xenobiotics, similar to other drugs,
herbal derivatives or environmental pollutants can cause clinically
relevant P-gp and CYP3A4 induction via Pregnane X receptor
regulation (Wojnowski, 2004; Xu et al, 2005; di Masi et al, 2009).
Complete inhibition of both P-gp and CYP3A4 in intestine and
liver by elacridar and ritonavir can eliminate the effects of
xenobiotic-related induction of these detoxifying proteins, and
thereby decrease variability in taxane pharmacokinetics after oral
administration. Polymorphisms of genes encoding for P-gp and
CYP3A4 are currently not known to be related to variability in
pharmacokinetics of i.v. administered taxanes (Bosch et al, 2006;
Jabir et al, 2012), but after oral administration, polymorphisms of
these genes can become more important. Interpatient variability in
oral bioavailability of docetaxel is decreased when docetaxel is
co-administered with inhibitors of CYP3A or P-gp (Stuurman et al,
2013). Not only polymorphisms, but also incomplete CYP3A4
inhibition by various other co-administered drugs might
contribute to variable oral taxane plasma exposure (Koolen et al,
2010b; Quinney et al, 2013). Complete inhibition of CYP3A by
ritonavir might eliminate this risk. It should be noted that, as P-gp
and CYP3A4 act at both the intestinal and hepatic level, the risk of
interpatient and intrapatient variability after oral administration of
taxanes is likely higher than after i.v. administration of taxanes, as
two potentially variable barriers need to be passed instead of one.
This underscores the importance of reducing potential sources of
variability by using effective CYP3A and P-gp inhibitors in oral
taxane regimens.
Boosting orally applied taxanes with elacridar and ritonavir
potentially increases the relative brain penetration of taxanes and
would thereby increase the risk of brain toxicity, by either
substantial inhibition of BBB P-gp by the circulating elacridar or
saturation of BBB P-gp activity because of the highly increased
plasma taxane levels, or a combination of both. We found here that
brain concentrations were increased after co-administration of the
taxanes with elacridar and ritonavir, but brain-to-plasma ratios
were not. This indicates that the increased brain concentrations
after oral co-administration of the taxanes with elacridar or
ritonavir were merely a consequence of the increased plasma
concentrations. Kemper et al (2003) showed a three-fold increase
in brain-to-plasma ratios in wild-type mice at 1 h after adminis-
tration of 10 mg kg
1
intravenously administered paclitaxel due to
25 mg kg
1
orally administered elacridar (brain-to-plasma ratios
after administration of paclitaxel with and without elacridar were
0.08 and 0.22, respectively). This three-fold increase was
3000
ABC
2000
NS 100
80
60
40
20
0
NS 0.15
0.10
0.05
0.00
NS
**
** **
**
1000
Docetaxel plasma
concentration (ng ml–1)
Docetaxel brain
concentration (ng g–1)
Brain/plasma ratio
0
DOC
DOC+ELC
DOC+RTV
DOC+ELC+RTV
DOC
DOC+ELC
DOC+RTV
DOC+ELC+RTV
DOC
DOC+ELC
DOC+RTV
DOC+ELC+RTV
Figure 4. Plasma and brain concentrations of docetaxel in Cyp3a
/
mice expressing human CYP3A4 in liver and intestine 2 h after oral
administration of 10 mg kg
1
docetaxel. Docetaxel was administered alone or co-administered with 25 mg kg
1
oral elacridar, 12.5 mg kg
1
oral
ritonavir or both elacridar and ritonavir. Panels reflect plasma concentrations (panel A), brain concentrations (panel B) or brain-to-plasma ratios
(panel C). Data are presented as individual data points and lines represent the mean. Differences in plasma or brain concentrations between
groups were statistically significantly different (Po0.001), unless stated otherwise (NS: not significant, P40.05 or **Po0.01). Differences in brain-
to-plasma ratios between all groups were not statistically significant. Values represent the means±s.d. Five to six animals per group were used.
Abbreviations: DOC ¼docetaxel; ELC ¼elacridar; RTV ¼ritonavir.
BRITISH JOURNAL OF CANCER Elacridar and ritonavir boost oral taxanes
2674 www.bjcancer.com | DOI:10.1038/bjc.2014.222
comparable with the increase in brain-to-plasma ratios as observed
after i.v. administration of paclitaxel to P-gp knockout mice. Brain
concentrations were not further increased when the elacridar
dose was increased to 100 mg kg
1
. Both findings suggest that
25 mg kg
1
oral elacridar can largely, if not completely, inhibit
BBB P-gp activity. However, in our experiments, we observed no
increase in brain-to-plasma ratios after oral co-administration of
the same dose of paclitaxel and elacridar. This can most likely be
explained by the initially far higher plasma levels of paclitaxel after
i.v. administration compared with those after oral administration.
When operating close to saturation, P-gp at the BBB will be more
sensitive to partial inhibition (Kalvass et al, 2013). The absence of
increased brain-to-plasma ratios in the experiments by Kemper
et al (2003) at 4 h after administration of i.v. paclitaxel and oral
elacridar (when plasma concentrations of paclitaxel are much
lower) further supports this interpretation (brain-to-plasma ratios
in wild-type mice after administration of paclitaxel with and
without elacridar were 0.9 and 0.8, respectively, whereas brain-to-
plasma ratios in knockout mice were 2.7 after i.v. administration of
paclitaxel at this time point). Collectively, our data suggest that at
modest plasma concentrations of paclitaxel (and presumably also
docetaxel), P-gp in the BBB has little or no effect on the relative
brain accumulation of taxanes.
CONCLUSIONS
Comparison of the results in our study with previously reported
data obtained from oral administration of taxanes to knockout
mice showed that orally administered elacridar and ritonavir at
comparatively low doses can completely (for paclitaxel), or almost
completely (for docetaxel) inhibit intestinal and hepatic P-gp and
CYP3A4 activity.
We also demonstrated that co-administration of the taxanes
with elacridar and ritonavir simultaneously resulted in a further
increase in plasma levels of the taxanes. In contrast, relative brain
accumulation of the taxanes was not affected after boosting with
oral elacridar. Even at the highly increased plasma concentrations
of taxanes after boosting with both elacridar and ritonavir, relative
brain accumulation was still similar as seen after boosting with
elacridar, or even in otherwise untreated CYP3A4-humanised
animals.
We therefore believe that it will be worthwhile testing
whether simultaneous inhibition of P-gp and CYP3A may
provide a relatively safe strategy to boost plasma exposure of
orally applied taxanes in patients, as relative brain exposure is
unlikely to be higher than that in the currently used i.v.
schedules.
CONFLICT OF INTEREST
The research group of AHS receives revenue from commercial
distribution of some of the mouse strains used in this study. JHMS
and JHB are inventors on patents on the application of oral taxane
formulations. The remaining authors declare no conflict of interest.
REFERENCES
Bardelmeijer HA, Beijnen JH, Brouwer KR, Rosing H, Nooijen WJ,
Schellens JH, van Tellingen O (2000) Increased oral bioavailability of
paclitaxel by GF120918 in mice through selective modulation of
P-glycoprotein. Clin Cancer Res 6: 4416–4421.
Bardelmeijer HA, Ouwehand M, Beijnen JH, Schellens JH, van Tellingen O
(2004) Efficacy of novel P-glycoprotein inhibitors to increase the oral
uptake of paclitaxel in mice. Invest New Drugs 22: 219–229.
Bardelmeijer HA, Ouwehand M, Buckle T, Huisman MT, Schellens JH,
Beijnen JH, van Tellingen O (2002) Low systemic exposure of oral
docetaxel in mice resulting from extensive first-pass metabolism is boosted
by ritonavir. Cancer Res 62: 6158–6164.
Bosch TM, Meijerman I, Beijnen JH, Schellens JH (2006) Genetic polymorphisms
of drug-metabolising enzymes and drug transporters in the chemotherapeutic
treatment of cancer. Clin Pharmacokinet 45: 253–285.
di Masi A, De Marinis E, Ascenzi P, Marino M (2009) Nuclear receptors CAR
and PXR: Molecular, functional, and biomedical aspects. Mol Aspects Med
30: 297–343.
Glaeser H, Fromm MF (2008) Animal models and intestinal drug transport.
Exp Opin Drug Metab Toxicol 4: 347–361.
Gligorov J, Lotz JP (2004) Preclinical pharmacology of the taxanes:
implications of the differences. Oncologist 9(Suppl 2): 3–8.
Gottesman MM, Ambudkar SV (2001) Overview: ABC transporters and
human disease. J Bioenerg Biomembr 33: 453–458.
Hendrikx JJ, Lagas JS, Rosing H, Schellens JH, Beijnen JH, Schinkel AH (2013)
P-glycoprotein and cytochrome P450 3A act together in restricting the
oral bioavailability of paclitaxel. Int J Cancer 132: 2439–2447.
Jabir RS, Naidu R, Annuar MA, Ho GF, Munisamy M, Stanslas J (2012)
Pharmacogenetics of taxanes: impact of gene polymorphisms of drug
transporters on pharmacokinetics and toxicity. Pharmacogenomics 13:
1979–1988.
Jiang H, Tao W, Zhang M, Pan S, Kanwar JR, Sun X (2010) Low-dose
metronomic paclitaxel chemotherapy suppresses breast tumors and
metastases in mice. Cancer Invest 28: 74–84.
Kalvass JC, Polli JW, Bourdet DL, Feng B, Huang SM, Liu X, Smith QR,
Zhang LK, Zamek-Gliszczynski MJ (2013) Why clinical modulation of
efflux transport at the human blood-brain barrier is unlikely: the ITC
evidence-based position. Clin Pharmacol Ther 94: 80–94.
Kemper EM, van Zandbergen AE, Cleypool C, Mos HA, Boogerd W, Beijnen JH,
van Tellingen O (2003) Increased penetration of paclitaxel into the brain
by inhibition of P-Glycoprotein. Clin Cancer Res 9: 2849–2855.
Koolen SL. Intravenous-to-oral switch in anticancer chemotherapy.
Focus on taxanes and gemcitabine. Dissertation, Utrecht University:
Utrecht, The Netherlands, 2011.
Koolen SL, Beijnen JH, Schellens JHM (2010a) Intravenous-to-oral switch
in anticancer chemotherapy: a focus on docetaxel and paclitaxel. Clin
Pharmacol Ther 87: 126–129.
Koolen SL, Oostendorp RL, Beijnen JH, Schellens JHM, Huitema AD (2010b)
Population pharmacokinetics of intravenously and orally administered
docetaxel with or without co-administration of ritonavir in patients with
advanced cancer. Br J Clin Pharmacol 69: 465–474.
Kuppens IE, van Maanen MJ, Rosing H, Schellens JHM, Beijnen JH (2005)
Quantitative analysis of docetaxel in human plasma using liquid
chromatography coupled with tandem mass spectrometry. Biomed
Chromatogr 19: 355–361.
Lagas JS, Vlaming ML, van Tellingen O, Wagenaar E, Jansen RS, Rosing H,
Beijnen JH, Schinkel AH (2006) Multidrug resistance protein 2 is an
important determinant of paclitaxel pharmacokinetics. Clin Cancer Res 12:
6125–6132.
Malingre MM, Beijnen JH, Rosing H, Koopman FJ, Jewell RC, Paul EM,
ten Bokkel Huinink WW, Schellens JH (2001) Co-administration of
GF120918 significantly increases the systemic exposure to oral paclitaxel
in cancer patients. Br J Cancer 84: 42–47.
Marchetti S, Stuurman FE, Koolen SL, Moes JJ, Hendrikx JJ, Thijssen B,
Huitema AD, Nuijen B, Rosing H, Keessen M, Voest EE, Mergui-Roelvink M,
Beijnen JH, Schellens JH (2012) Phase I study of weekly oral docetaxel
(ModraDoc001) plus ritonavir in patients with advanced solid tumors.
ASCO Meeting Abstracts 30: 2550.
Meerum Terwogt JM, Beijnen JH, ten Bokkel Huinink WW, Rosing H,
Schellens JH (1998) Co-administration of cyclosporin enables oral therapy
with paclitaxel. Lancet 352: 285.
Oostendorp RL, Huitema A, Rosing H, Jansen RS, Ter Heine R, Keessen M,
Beijnen JH, Schellens JHM (2009) Coadministration of ritonavir strongly
enhances the apparent oral bioavailability of docetaxel in patients with
solid tumors. Clin Cancer Res 15: 4228–4233.
Paine MF, Hart HL, Ludington SS, Haining RL, Rettie AE, Zeldin DC (2006)
The human intestinal cytochrome P450 ‘pie’. Drug Metab Dispos 34:
880–886.
Quinney SK, Malireddy SR, Vuppalanchi R, Hamman MA, Chalasani N,
Gorski JC, Hall SD (2013) Rate of onset of inhibition of gut-wall and
hepatic CYP3A by clarithromycin. Eur J Clin Pharmacol 69: 439–448.
Elacridar and ritonavir boost oral taxanes BRITISH JOURNAL OF CANCER
www.bjcancer.com | DOI:10.1038/bjc.2014.222 2675
Schellens JHM, Malingre MM, Kruijtzer CM, Bardelmeijer HA, van Tellingen O,
Schinkel AH, Beijnen JH (2000) Modulation of oral bioavailability of
anticancer drugs: from mouse to man. Eur J Pharm Sci 12: 103–110.
Sparreboom A, van Asperen J, Mayer U, Schinkel AH, Smit JW, Meijer DK,
Borst P, Nooijen WJ, Beijnen JH, van Tellingen O (1997) Limited oral
bioavailability and active epithelial excretion of paclitaxel (Taxol) caused
by P-glycoprotein in the intestine. Proc Natl Acad Sci USA 94: 2031–2035.
Stokvis E, Ouwehand M, Nan LG, Kemper EM, van Tellingen O, Rosing H,
Beijnen JH (2004) A simple and sensitive assay for the quantitative
analysis of paclitaxel in human and mouse plasma and brain tumor tissue
using coupled liquid chromatography and tandem mass spectrometry.
J Mass Spectrom 39: 1506–1512.
Stuurman FE, Nuijen B, Beijnen JH, Schellens JH (2013) Oral anticancer
drugs: mechanisms of low bioavailability and strategies for improvement.
Clin Pharmacokinet 52: 399–414.
Thelen K, Dressman JB (2009) Cytochrome P450-mediated metabolism in the
human gut wall. J Pharm Pharmacol 61: 541–558.
Vaishampayan U, Parchment RE, Jasti BR, Hussain M (1999) Taxanes: an
overview of the pharmacokinetics and pharmacodynamics. Urology 54:
22–29.
van Asperen J, van Tellingen O, Sparreboom A, Schinkel AH, Borst P,
Nooijen WJ, Beijnen JH (1997) Enhanced oral bioavailability of paclitaxel
in mice treated with the P-glycoprotein blocker SDZ PSC 833. Br J Cancer
76: 1181–1183.
van Asperen J, van Tellingen O, van der Valk MA, Rozenhart M,
Beijnen JH (1998) Enhanced oral absorption and decreased elimination
of paclitaxel in mice cotreated with cyclosporin A. Clin Cancer Res 4:
2293–2297.
van Herwaarden AE, Wagenaar E, van der Kruijssen CM, van Waterschoot RA,
Smit JW, Song JY, van der Valk MA, van Tellingen O, van der Hoorn JW,
Rosing H, Beijnen JH, Schinkel AH (2007) Knockout of cytochrome P450
3A yields new mouse models for understanding xenobiotic metabolism.
J Clin Invest 117: 3583–3592.
van Waterschoot RA, Lagas JS, Wagenaar E, van der Kruijssen CM,
van Herwaarden AE, Song JY, Rooswinkel RW, van Tellingen O, Rosing H,
Beijnen JH, Schinkel AH (2009) Absence of both cytochrome P450 3A and
P-glycoprotein dramatically increases docetaxel oral bioavailability and
risk of intestinal toxicity. Cancer Res 69: 8996–9002.
Wojnowski L (2004) Genetics of the variable expression of CYP3A in humans.
Ther Drug Monit 26: 192–199.
Wu H, Xin Y, Zhao J, Sun D, Li W, Hu Y, Wang S (2011) Metronomic
docetaxel chemotherapy inhibits angiogenesis and tumor growth in a
gastric cancer model. Cancer Chemother Pharmacol 68: 879–887.
Xu C, Li CY, Kong AN (2005) Induction of phase I, II and III drug
metabolism/transport by xenobiotics. Arch Pharm Res 28: 249–268.
This work is published under the standard license to publish agree-
ment. After 12 months the work will become freely available and
the license terms will switch to a Creative Commons Attribution-
NonCommercial-Share Alike 3.0 Unported License.
Supplementary Information accompanies this paper on British Journal of Cancer website (http://www.nature.com/bjc)
BRITISH JOURNAL OF CANCER Elacridar and ritonavir boost oral taxanes
2676 www.bjcancer.com | DOI:10.1038/bjc.2014.222
... are interested in the development of oral taxane formulations, because these are more patient-friendly and less costly [2,3]. Furthermore, it more easily allows metronomic therapy and patients are not subjected to intravenous exposure to pharmaceutical vehicles, which may trigger hypersensitivity reactions [4]. ...
... Furthermore, it more easily allows metronomic therapy and patients are not subjected to intravenous exposure to pharmaceutical vehicles, which may trigger hypersensitivity reactions [4]. However, the oral bioavailability of taxanes is seriously hampered by first-pass metabolism by Cytochrome P450 3A4/5 (CYP3A/5) enzymes in the liver and intestine [2,3]. Therefore, the systemic exposure of oral docetaxel is boosted by using the CYP3A inhibitor ritonavir (ModraDoc006/r), which highly increases the oral bioavailability as shown in mice and clinical studies [2,[5][6][7]. ...
... However, the oral bioavailability of taxanes is seriously hampered by first-pass metabolism by Cytochrome P450 3A4/5 (CYP3A/5) enzymes in the liver and intestine [2,3]. Therefore, the systemic exposure of oral docetaxel is boosted by using the CYP3A inhibitor ritonavir (ModraDoc006/r), which highly increases the oral bioavailability as shown in mice and clinical studies [2,[5][6][7]. ...
Article
Full-text available
Purpose An oral docetaxel formulation boosted by the Cytochrome P450 (CYP) 3 A inhibitor ritonavir, ModraDoc006/r, is currently under clinical investigation. Based on clinical data, the incidence of grade 1–2 diarrhea is increased with this oral docetaxel formulation compared to the conventional intravenous administration. Loperamide, a frequently used diarrhea inhibitor, could be added to the regimen as symptomatic treatment. However, loperamide is also a substrate of the CYP3A enzyme, which could result in competition between ritonavir and loperamide for this protein. Therefore, we were interested in the impact of coadministered loperamide on the pharmacokinetics of ritonavir-boosted oral docetaxel. Methods We administered loperamide simultaneously or with an 8-hour delay to humanized CYP3A4 mice (with expression in liver and intestine) receiving oral ritonavir and docetaxel. Concentrations of docetaxel, ritonavir, loperamide and two of its active metabolites were measured. Results The plasma exposure (AUC and Cmax) of docetaxel was not altered during loperamide treatment, nor were the ritonavir plasma pharmacokinetics. However, the hepatic and intestinal dispositions of ritonavir were somewhat changed in the simultaneous, but not 8-hour loperamide treatment groups, possibly due to loperamide-induced delayed drug absorption. The pharmacokinetics of loperamide itself did not seem to be influenced by ritonavir. Conclusion These results suggest that delayed loperamide administration can be added to ritonavir-boosted oral docetaxel treatment, without affecting the overall systemic exposure of docetaxel.
... After the oral administration of PTX, the drug is actively pumped out by P-gp and metabolized by the CYP2C8 and CYP3A4 enzymes located in the intestine, eventually causing its low intestinal uptake and systemic exposure [30]. Various approaches have been employed to enhance oral bioavailability of drugs including the use of P-gp inhibitors (e.g., cyclosporine A, valspodar SDZPSC833 or HM30181) [38][39][40] to increase oral absorption at the GI tract; the use of nanotechnology/nanoemulsion/liposome to enhance penetration [41]; or the use of CYP3A4 inhibitor (e.g., ritonavir) to reduce the drug metabolism [42]. ...
... The combined treatment of oral PTX with P-gp inhibitor cyclosporine analog SDZPSC833 resulted in an approximately 10-fold increase in the AUC of PTX [39]. In addition, oral co-administration with the P-gp inhibitor elacridar or CYP3A inhibitor ritonavir have also been reported to strongly enhance the AUC of PTX by 10.7-fold and 2.5-fold as compared to a single oral dose of PTX [42]. In agreement with the P-gp KO or CYP3A KO mice, the AUC improvement was relatively lower when co-administered with the CYP3A inhibitor than the P-gp inhibitor. ...
... Combined administration of elacridar and ritonavir was effective and gave 31.7-fold enhancement in AUC [42]. Comparatively, FM04 (AUC fold change = 30 to 66, Figure 6A) is more efficient and superior to PDSZPSC833, elacridar or ritonabvir alone or combined elacridar and ritonabvir to be applied in oral PTX formulation. ...
Article
Full-text available
Biotransformation of flavonoid dimer FD18 resulted in an active metabolite FM04. It was more druggable because of its improved physicochemical properties. FM04 (EC50 = 83 nM) was 1.8-fold more potent than FD18 in reversing P-glycoprotein (P-gp)-mediated paclitaxel (PTX) resistance in vitro. Similar to FD18, FM04 chemosensitized LCC6MDR cells towards multiple anticancer drugs by inhibiting the transport activity of P-gp and restoring intracellular drug levels. It stimulated the P-gp ATPase by 3.3-fold at 100 μM. Different from FD18, FM04 itself was not a transport substrate of P-gp and presumably, it cannot work as a competitive inhibitor. In the human melanoma MDA435/LCC6MDR xenograft, the co-administration of FM04 (28 mg/kg, I.P.) with PTX (12 mg/kg, I.V.) directly modulated P-gp-mediated PTX resistance and caused a 56% (*, p < 0.05) reduction in tumor volume without toxicity or animal death. When FM04 was administered orally at 45 mg/kg as a dual inhibitor of P-gp/CYP2C8 or 3A4 enzymes in the intestine, it increased the intestinal absorption of PTX from 0.2% to 14% in mice and caused about 57- to 66-fold improvement of AUC as compared to a single oral dose of PTX. Oral co-administration of FM04 (45 mg/kg) with PTX (40, 60 or 70 mg/kg) suppressed the human melanoma MDA435/LCC6 tumor growth with at least a 73% (***, p < 0.001) reduction in tumor volume without serious toxicity. Therefore, FM04 can be developed into a novel combination chemotherapy to treat cancer by directly targeting the P-gp overexpressed tumors or potentiating the oral bioavailability of P-gp substrate drugs.
... Taxanes have a low oral availability due to high first-pass metabolism by CYP3A4/5 in the liver and intestine. Therefore, coadministration of ritonavir with these drugs could enhance the oral availability of orally administered taxanes [20][21][22]. This is one example of a drug that can be boosted by ritonavir, but in the future, this may also be possible for other drugs. ...
... This is one example of a drug that can be boosted by ritonavir, but in the future, this may also be possible for other drugs. Additionally, for some currently running research programs, lower doses of ritonavir compared to the typically used boosting doses could also be of value, especially when combining ritonavir with (cyto-) toxic drugs [20,21,23]. These classes of drugs have a higher risk of (severe) side effects and this initial risk will increase when such drugs are boosted by ritonavir due to higher plasma exposure. ...
Article
Full-text available
Ritonavir, originally developed as HIV protease inhibitor, is widely used as a booster in several HIV pharmacotherapy regimens and more recently in Covid-19 treatment (e.g., Paxlovid). Its boosting capacity is due to the highly potent irreversible inhibition of the cytochrome P450 (CYP) 3 A enzyme, thereby enhancing the plasma exposure to coadministered drugs metabolized by CYP3A. Typically used booster doses of ritonavir are 100-200 mg once or twice daily. This review aims to address several aspects of this booster drug, including the possibility to use lower ritonavir doses, 20 mg for instance, resulting in partial CYP3A inactivation in patients. If complete CYP3A inhibition is not needed, lower ritonavir doses could be used, thereby reducing unwanted side effects. In this context, there are contradictory reports on the actual recovery time of CYP3A activity after ritonavir discontinuation, but probably this will take at least one day. In addition to ritonavir's CYP3A inhibitory effect, it can also induce and/or inhibit other CYP enzymes and drug transporters, albeit to a lesser extent. Although ritonavir thus exhibits gene induction capacities, with respect to CYP3A activity the inhibition capacity clearly predominates. Another potent CYP3A inhibitor, the ritonavir analog cobicistat, has been reported to lack the ability to induce enzyme and transporter genes. This might result in a more favorable drug-drug interaction profile compared to ritonavir, although the actual benefit appears to be limited. Indeed, ritonavir is still the clinically most used pharmacokinetic enhancer, indicating that its side effects are well manageable, even in chronic administration regimens.
... Coadministration with ritonavir resulted in a 7.3-and 2.5-fold increase in plasma concentration, while coadministration with elacridar/ ritonavir resulted in a 31.9-and 37.4-fold increment in paclitaxel and docetaxel plasma concentration, respectively (Hendrikx et al. 2014). Research has also shown that the bioavailability of the taxane is enhanced when co-administered with a P-gp inhibitor. ...
Article
Full-text available
Background Cancer remains one of the leading causes of death worldwide. Docetaxel, first marketed as ‘Taxotere’, has been approved for use as a chemotherapeutic for management of gastric, breast, neck, head, non-small cell lung cancer and prostate cancer. Main body Taxotere, is a docetaxel formulation solubilized in tween 80 and 13% ethanol solution. It is effective as a chemotherapeutic agent but has numerous toxic effects due to the ethanol and polysorbate. Aside from this, challenges with administration during mixing of the docetaxel with the diluent exist. Poor mixing results in gel formation while addition of the drug mix to an infusion may result in micelle formation. These challenges have necessitated remodeling of the currently available docetaxel formulation, but none has made it to clinical setting as an alternative. Efforts have also been made to develop oral docetaxel formulation to ease administration of the drug. Attempts have also been made to develop other dosage forms, notably transdermal formulations and implants to target cancer cells while avoiding systemic side effects. Formulation methods such as nanoformulation, drug coupling with other active moieties, 3D fabrication as well as electrospinning have been employed. Conclusions Development of novel formulations of docetaxel for different chemotherapeutic needs appears promising with some formulations currently in clinical trials. Exploring other drug formulation techniques such as the use of 2D LDH may produce novel anticancer formulations in the future.
Article
There is currently great interest in developing oral taxanes due to their lower costs and greater patient friendliness. We here wanted to test whether oral ritonavir, a cytochrome P450 3A (CYP3A) inhibitor, could boost the pharmacokinetics and tissue distribution of orally administered cabazitaxel (10 mg/kg) in male wild-type, Cyp3a-/-, and Cyp3aXAV (transgenic overexpression of human CYP3A4 in liver and intestine) mice. Ritonavir was initially administered at a dose of 25 mg/kg, but lower dosages of 10 and 1 mg/kg were also studied to assess the remaining amount of boosting, aiming to minimize possible side effects. Compared to the respective vehicle groups, plasma exposure of cabazitaxel (AUC0-24h) was enhanced 2.9-, 10.9-, and 13.9-fold in wild-type mice and 1.4-, 10.1-, and 34.3-fold in Cyp3aXAV mice by treatment with 1, 10, and 25 mg/kg ritonavir, respectively. Upon treatment with 1, 10, and 25 mg/kg of ritonavir, the peak plasma concentration (Cmax) was increased by 1.4-, 2.3-, and 2.8-fold in wild-type mice, while it increased by 1.7-, 4.2-, and 8.0-fold in Cyp3aXAV mice, respectively. AUC0-24h and Cmax remained unchanged in Cyp3a-/-. Biotransformation of cabazitaxel to its active metabolites still took place when coadministered with ritonavir, but this process was delayed due to the Cyp3a/CYP3A4 inhibition. These data indicate that CYP3A is the primary limiting factor in the plasma exposure to cabazitaxel and that cabazitaxel oral bioavailability could be dramatically enhanced by coadministration of an effective CYP3A inhibitor such as ritonavir. These findings could be a starting point for the setup of a clinical study, which would be needed to verify the boosting of cabazitaxel by ritonavir in humans.
Article
Full-text available
The drug efflux transporters ABCB1 and ABCG2 at the blood-brain barrier limit the delivery of drugs into the brain. Strategies to overcome ABCB1/ABCG2 have been largely unsuccessful, which poses a tremendous clinical problem to successfully treat CNS diseases. Understanding basic transporter biology, including intracellular regulation mechanisms that control these transporters, is critical to solve this clinical problem. In this comprehensive review, we summarize current knowledge on signaling pathways that regulate ABCB1/ABCG2 at the blood-brain barrier. In part I, we give a historical overview on blood-brain barrier research and introduce the role ABCB1 and ABCG2 play in this context. In part II, we summarize the most important strategies that have been tested to overcome the ABCB1/ABCG2 efflux system at the blood-brain barrier. In part III, the main component of this review, we provide detailed information on the signaling pathways that have been identified to control ABCB1/ABCG2 at the blood-brain barrier and their potential clinical relevance. This is followed by part IV, where we explain the clinical implications of ABCB1/ABCG2 regulation in the context of CNS disease. Lastly, in part V, we conclude by highlighting examples of how transporter regulation could be targeted for therapeutic purposes in the clinic. Significance Statement The ABCB1/ABCG2 drug efflux system at the blood-brain barrier poses a significant problem to successful drug delivery to the brain. Here, we review signaling pathways that regulate blood-brain barrier ABCB1/ABCG2 and could potentially be targeted for therapeutic purposes.
Article
Taxanes are the backbone cytotoxic agents that have been prescribed in most solid malignant treatments for decades. Taxane-induced peripheral neuropathy (PN) and drug resistance are inevitable. Novel taxanes include semisynthetic taxanes, taxane analogs, taxanes with a different drug-delivery method, taxanes without lipid carries, oral taxanes, taxanes with resistance to permeability glycoprotein (P-gp), and taxanes with easy penetration of the blood-brain barrier (BBB). Some taxanes have already shown promising activity in the clinic and have shown abilities to overcome the drug resistance commonly occurring with traditional taxane treatment. Predictive b for response to taxanes are being explored in order to offer precision medicine in cancer therapy. This review will focus on the history, current, and future directions of taxane development in breast cancer, especially that of novel taxanes such as oral taxanes. Clinical trials on novel taxanes, including the mature phase III trials of oral taxane and the recent setback, as well as the future direction of taxane research, will be discussed.
Article
Full-text available
Learning Objectives After completing this course, the reader will be able to: Identify the differences in molecular pharmacology of the taxanes. Describe the impact of the pharmacokinetic and pharmacodynamic profiles of the taxanes on their administration and toxicity. Outline the toxicity profiles of paclitaxel and docetaxel. Access and take the CME test online and receive 1 hour of AMA PRA category 1 credit at CME.TheOncologist.com Taxanes are one of the most powerful classes of compounds among all chemotherapeutic drugs. Only 30 years separate the isolation of the first taxane from the results of direct clinical comparisons in metastatic breast, ovarian, and lung cancer between the two taxanes available in routine clinical practice. These results suggest a more favorable benefit-to-risk ratio for docetaxel compared to paclitaxel when these drugs are used as single agents or in combination with other chemotherapeutic agents in an every-3-week dosing regimen. Pharmacological data support the difference between the taxanes, likely explaining the clinical results. Considering the molecular pharmacology of the two drugs, docetaxel appears to bind to β-tubulin with greater affinity and has a wider cell cycle activity than paclitaxel. Docetaxel also appears to have direct antitumoral activity via an apoptotic effect mediated by bcl-2 phosphorylation. In addition, docetaxel has a longer retention time in tumor cells than paclitaxel because of greater uptake and slower efflux. Pharmacokinetics and pharmacodynamics of the taxanes show both agents to be extensively metabolized in the liver, and paclitaxel has a nonlinear pharmacokinetic behavior while docetaxel has linear pharmacokinetics. These differences explain the more simple treatment schedule and favorable results for docetaxel as a single agent and in combination therapy. Last, but not least, there is a pharmacokinetic interaction between paclitaxel and the anthracyclines, an active class of compounds commonly used in the treatment of breast cancer. This pharmacokinetic interaction is associated with greater cardio- and myelotoxicities, which are sequence dependent. These pharmacological data likely explain the different clinical development strategies for the two molecules as well as the different clinical results from individual trials and direct comparisons.
Article
Full-text available
The use of oral anticancer drugs has increased during the last decade, because of patient preference, lower costs, proven efficacy, lack of infusion-related inconveniences, and the opportunity to develop chronic treatment regimens. Oral administration of anticancer drugs is, however, often hampered by limited bioavailability of the drug, which is associated with a wide variability. Since most anticancer drugs have a narrow therapeutic window and are dosed at or close to the maximum tolerated dose, a wide variability in the bioavailability can have a negative impact on treatment outcome. This review discusses mechanisms of low bioavailability of oral anticancer drugs and strategies for improvement. The extent of oral bioavailability depends on many factors, including release of the drug from the pharmaceutical dosage form, a drug's stability in the gastrointestinal tract, factors affecting dissolution, the rate of passage through the gut wall, and the pre-systemic metabolism in the gut wall and liver. These factors are divided into pharmaceutical limitations, physiological endogenous limitations, and patient-specific limitations. There are several strategies to reduce or overcome these limitations. First, pharmaceutical adjustment of the formulation or the physicochemical characteristics of the drug can improve the dissolution rate and absorption. Second, pharmacological interventions by combining the drug with inhibitors of transporter proteins and/or pre-systemic metabolizing enzymes can overcome the physiological endogenous limitations. Third, chemical modification of a drug by synthesis of a derivative, salt form, or prodrug could enhance the bioavailability by improving the absorption and bypassing physiological endogenous limitations. Although the bioavailability can be enhanced by various strategies, the development of novel oral products with low solubility or cell membrane permeability remains cumbersome and is often unsuccessful. The main reasons are unacceptable variation in the bioavailability and high investment costs. Furthermore, novel oral anticancer drugs are frequently associated with toxic effects including unacceptable gastrointestinal adverse effects. Therefore, compliance is often suboptimal, which may negatively influence treatment outcome.
Article
2550 Background: ModraDoc001 is a novel oral formulation containing docetaxel as a solid dispersion. Oral administration of docetaxel is feasible in combination with the CYP3A4 inhibitor ritonavir. Objectives were to determine the safety, maximum tolerated dose (MTD) and pharmacokinetics (PK) of weekly oral docetaxel (as ModraDoc001) in combination with ritonavir. Methods: Patients with advanced solid tumors, WHO PS ≤ 2, no concomitant use of MDR or CYP3A modulating drugs, adequate bone marrow (ANC ≥ 1.5x10 ⁹ /L), liver (bilirubin ≤ 1.5xULN, ALAT/ASAT ≤ 2.5xULN) and renal function (creatinine ≤ 1.5xULN or clearance ≥ 50 ml/min) were eligible. Docetaxel (ModraDoc001 10mg capsule) and ritonavir (Norvir 100mg) were simultaneously given once weekly in a ‘3+3 cohort’ dose escalation design. MTD was defined as the highest dose resulting in <1/6 probability of causing a dose limiting toxicity in the first 4 weeks of treatment. This cohort was expanded with 6 patients. PK was determined on days 1 and 8. Results: 40 patients (25 male, 35 evaluable for safety) were enrolled in 6 dose levels (30/100, 40/100, 60/100, 80/100, 60/200 and 80/200 mg docetaxel/ritonavir). Common treatment related adverse events in the 4 highest dose levels were diarrhea (68%), nausea (62%) and fatigue (62%), mostly CTC grade 1-2 (80%, 95% and 73% respectively). Five patients experienced a DLT (grade 3 diarrhea (4), elevated ASAT/ALAT (1), grade 4 dehydration and grade 3 mucositis (1), grade 3 fatigue (2)). The MTD was 60 mg/200 mg docetaxel/ritonavir. Partial remission was seen in 4 patients (CUP, NSCLC, gastric and mamma ca) and sustained stable disease in 15 patients (6x NSCLC). Both drugs were rapidly absorbed after oral administration. Mean Tmax was 2.0 hours (CV=73%) for docetaxel. Cmax and AUC of docetaxel increased less than proportionally with dose to 162 ng/ml (CV= 67%) and 1615 ng/ml*hr (CV= 81%), respectively, in 15 patients at the MTD. Conclusions: At the MTD (once weekly 60 mg docetaxel and 200 mg ritonavir) ModraDoc001 is safe, well tolerated and shows encouraging antitumor activity. The exposure of docetaxel with this regimen is comparable to weekly intravenous administration of 35 mg/m ² docetaxel. Phase II studies in solid tumors are planned.
Article
Drug interactions due to efflux transporter inhibition at the blood-brain barrier have been receiving increasing scrutiny due to the theoretical possibility of adverse CNS effects based on findings from preclinical studies. In this commentary, evidence from pharmacokinetic, pharmacodynamic, imaging, pharmacogenetic, and pharmacovigilance studies, along with drug safety reports, is presented supporting a low probability of inhibiting efflux transporters at the human blood-brain barrier by currently marketed drugs.Clinical Pharmacology & Therapeutics (2013); accepted article preview 14 February 2013 doi:10.1038/clpt.2013.34.
Article
Interindividual variability in drug response and the emergence of adverse drug effects are the main causes of treatment failure in cancer therapy. Functional membrane drug transporters play important roles in altering pharmacokinetic profile, resistance to treatment, toxicity and patient survival. Pharmacogenetic studies of these transporters are expected to provide new approaches for optimizing therapy. Taxanes are approved for the treatment of various cancers. Circulating taxanes are taken up by SLCO1B3 into hepatocytes. The CYP450 enzymes CYP3A4, CYP3A5 and CYP2C8 are responsible for the conversion of taxanes into their metabolites. Ultimately, ABCB1 and ABCC2 will dispose the metabolites into bile canaliculi. Polymorphisms of genes encoding for proteins involved in the transport and clearance of taxanes reduce excretion of the drugs, leading to development of toxicity in patients. This review addresses current knowledge on genetic variations of transporters affecting taxanes pharmacokinetics and toxicity, and provides insights into future direction for personalized medicine.
Article
Paclitaxel is avidly transported by P-glycoprotein (P-gp/MDR1/ABCB1/rpar;. This results in low oral bioavailability, which can be boosted by co-administration of P-gp inhibitors. Unlike paclitaxel, docetaxel is extensively metabolized by CYP3A4 and its oral bioavailability can be enhanced in mice and humans by co-administration of the potent CYP3A inhibitor ritonavir. Unexpectedly, ritonavir also enhances the oral bioavailability of paclitaxel in humans. We aimed to resolve the mechanism underlying this enhancement. Using mice lacking Cyp3a and/or P-gp we investigated the combined and separate restricting roles of Cyp3a and P-gp in the oral bioavailability of paclitaxel, and the boosting effect of ritonavir. CYP3A4-humanized mice were used for translation to the human situation. P-gp had a dominant effect (11.6-fold, P < 0.001) over Cyp3a (<1.5-fold, n.s.) in limiting plasma concentrations of oral paclitaxel. However, in the absence of P-gp, Cyp3a decreased paclitaxel plasma concentrations 2-fold (P < 0.001). Co-administered ritonavir inhibited Cyp3a-mediated metabolism, but not P-gp-mediated transport of paclitaxel. Due to the dominant effect of P-gp, ritonavir only enhanced paclitaxel plasma concentrations in P-gp-deficient mice. Mouse liver microsomes metabolized paclitaxel far less efficiently than human or CYP3A4-transgenic liver microsomes, revealing much lower efficiency of paclitaxel metabolism by mouse than by human CYP3As. Accordingly, ritonavir could enhance the oral bioavailability of paclitaxel in CYP3A4-humanized mice, despite the fact that these mice are P-gp-proficient. Our results show that CYP3A4 inhibition most likely underlies the boosting effect of ritonavir on oral paclitaxel bioavailability in humans. Furthermore, CYP3A4-humanized mice allow improved understanding of CYP3A4-mediated paclitaxel metabolism in humans © 2012 Wiley Periodicals, Inc.
Article
Aims: To determine the extent and time-course of hepatic and intestinal cytochrome P450 3A (CYP3A) inactivation due to the mechanism-based inhibitor clarithromycin. Methods: Intestinal and hepatic CYP3A inhibition was examined in 12 healthy volunteers following the administration of single and multiple doses of oral clarithromycin (500 mg). Intestinal biopsies were obtained under intravenous midazolam sedation at baseline and after the first dose, on days 2-4, and on days 6-8 of the clarithromycin treatment. The formation of 1'-hydroxymidazolam in biopsy tissue and the serum 1'-hydroxymidazolam:midazolam ratio were indicators of intestinal and hepatic CYP3A activity, respectively. Results: Intestinal CYP3A activity decreased by 64 % (p = 0.0029) following the first dose of clarithromycin, but hepatic CYP3A activity did not significantly decrease. Repeated dosing of clarithromycin caused a significant decrease in hepatic CYP3A activity (p = 0.005), while intestinal activity showed little further decline. The CYP3A5 or CYP3A4*1B genotype were unable to account for inter-individual variability in CYP3A activity. Conclusions: Following the administration of clarithromycin, the onset of hepatic CYP3A inactivation is delayed compared to that of intestinal CYP3A. The time-course of drug-drug interactions due to clarithromycin will vary with the relative contribution of intestinal and hepatic CYP3A to the clearance and bioavailability of a victim substrate.
Article
Nuclear receptors (NRs) are ligand-activated transcription factors sharing a common evolutionary history and having similar sequence features at the protein level. Selective ligand(s) for some NRs is not known, therefore these NRs have been named “orphan receptors”. Whenever ligands have been recognized for any of the orphan receptor, it has been categorized and grouped as “adopted” orphan receptor. This group includes the constitutive androstane receptor (CAR) and the pregnane X receptor (PXR). They function as sensors of toxic byproducts derived from endogenous metabolites and of exogenous chemicals, in order to enhance their elimination. This unique function of CAR and PXR sets them apart from the steroid hormone receptors. The broad response profile has established that CAR and PXR are xenobiotic sensors that coordinately regulate xenobiotic clearance in the liver and intestine via induction of genes involved in drug and xenobiotic metabolism. In the past few years, research has revealed new and mostly unsuspected roles for CAR and PXR in modulating hormone, lipid, and energy homeostasis as well as cancer and liver steatosis. The purpose of this review is to highlight the structural and molecular bases of CAR and PXR impact on human health, providing information on mechanisms through which diet, chemical exposure, and environment ultimately impact health and disease.
Article
Low-dose metronomic (LDM) chemotherapy represents a new strategy to treat solid tumors by stronger antiangiogenic activity and less side effects. The aim of the study is to rationally develop a docetaxel metronomic regimen in preclinical settings of gastric cancer. In vitro cell proliferation, apoptosis, and thrombospondin-1/vascular endothelial growth factor (TSP-1/VEGF) expression analyses were performed on human umbilical vein endothelial cells (HUVECs) and gastric cancer (BGC-823) cells exposed for 144 h to metronomic concentrations of docetaxel. BGC-823 human gastric cancer xenograft model was used, and tumor growth and side effects were closely monitored. Quantitative real-time PCR was used to determine TSP-1/VEGF mRNA levels in tumor samples. Expression of VEGF and CD31 was observed by immunohistochemistry. Our results indicated that LDM docetaxel preferentially inhibited HUVEC cell proliferation and induced HUVEC apoptosis. Docetaxel significantly increased TSP-1 expression and secretion in HUVEC cells whereas the expression and secretion of VEGF significantly decreased in BGC-823 cells. LDM docetaxel significantly inhibited BGC-823 tumor growth in the absence of toxicity, which was accompanied by decreases in microvessel density (MVD) and VEGF and increases in TSP-1 gene expression in tumor tissues. In vitro results show the antiangiogenic properties of LDM docetaxel. In vivo, LDM docetaxel treatment is effective against gastric tumor and microvessel growth without toxic effect on nude mice.
Article
Most anticancer chemotherapeutic drugs are given intravenously. However, there is a growing interest in developing anticancer drugs for oral application. Different classes of anticancer drugs are already orally available and widely applied such as the tyrosine kinase inhibitors (imatinib, erlotinib, gefitinib, sorafenib and sunitinib), alkylating agents (temozolamide and cyclophosphamide) and the 5FU prodrug, capecitabine, a drug for which the intravenous-to-oral switch has already been successfully implemented in clinical practice. One of the aims of this thesis was to investigate the oral application of docetaxel. Docetaxel has a low bioavailability due to affinity for drug transporters, especially ABCB1 (P-glycoprotein, P-gp), extensive first pass metabolism by cytochrome P450 3A4 (CYP3A4) and poor drug solubility. In previous studies it has been demonstrated that the low systemic exposure to docetaxel is primarily determined by CYP3A4 in gut and liver. Inhibition of CYP3A4 in mice using low-dose ritonavir was found to increase the systemic exposure in mice by 50-fold. In patients the apparent bioavailability of docetaxel increased to more than 100% after co-administration of ritonavir. The studies described in this thesis investigated the pharmacology of orally administered docetaxel more thoroughly in order to determine the optimal boosted oral docetaxel regimen. We demonstrated that the concept of boosting docetaxel is possible with any strong CYP3A4 inhibitor. We selected low-dose ritonavir for the further development of boosted orally administered docetaxel, since ritonavir resulted in high systemic exposure to docetaxel, had a good safety profile and since ritonavir is used as booster as standard practice in multiple anti-HIV regimens. Most studies described in this thesis were conducted with a novel solid docetaxel formulation, ModraDoc001 capsules. To improve the poor aqueous solubility, a solid dispersion formulation of docetaxel has been designed by the pharmacy of the Slotervaart Hospital. This formulation was investigated in 55 patients and resulted in high systemic exposure to docetaxel. The variability was modest and of the same order as after intravenous administration of docetaxel. This is an important finding since high variability may lead to unexpected severe toxicity or under-dosing. The safety of weekly oral docetaxel in combination with ritonavir was determined in a dose escalation study. The most observed adverse event was diarrhea with an overall incidence of 66%. Major treatment limiting adverse effects observed after intravenous therapy with docetaxel, e.g. fluid retention, hematological toxicity and infusion reactions were not observed. Consequently, high doses of dexamethason to prevent fluid retention and allergic reactions were not required. Furthermore, the anti-tumor activity observed at the two highest dose-levels strengthens the concept that oral administration of docetaxel is feasible and potentially active. Concluding, the oral application of the taxanes docetaxel and paclitaxel was found to be feasible. The presented pharmacokinetic data demonstrated that the systemic exposure to the taxanes was high and signs of antitumor activity were observed after treatment with ritonavir boosted ModraDoc001 capsules. Plans for phase II clinical testing of ritonavir boosted ModraDoc001 are made for maintenance or second line treatment of patients with non-small-cell-lung-cancer