• No results found

Limitations of current antiretroviral therapy in HIV-1 infection: the search for new strategies - CHAPTER 9 P-glycoprotein in human immunodeficiency virus type 1 infection and therapy

N/A
N/A
Protected

Academic year: 2021

Share "Limitations of current antiretroviral therapy in HIV-1 infection: the search for new strategies - CHAPTER 9 P-glycoprotein in human immunodeficiency virus type 1 infection and therapy"

Copied!
31
0
0

Bezig met laden.... (Bekijk nu de volledige tekst)

Hele tekst

(1)

UvA-DARE is a service provided by the library of the University of Amsterdam (https://dare.uva.nl)

Limitations of current antiretroviral therapy in HIV-1 infection: the search for new

strategies

Sankatsing, S.U.C.

Publication date

2004

Link to publication

Citation for published version (APA):

Sankatsing, S. U. C. (2004). Limitations of current antiretroviral therapy in HIV-1 infection: the

search for new strategies.

General rights

It is not permitted to download or to forward/distribute the text or part of it without the consent of the author(s)

and/or copyright holder(s), other than for strictly personal, individual use, unless the work is under an open

content license (like Creative Commons).

Disclaimer/Complaints regulations

If you believe that digital publication of certain material infringes any of your rights or (privacy) interests, please

let the Library know, stating your reasons. In case of a legitimate complaint, the Library will make the material

inaccessible and/or remove it from the website. Please Ask the Library: https://uba.uva.nl/en/contact, or a letter

to: Library of the University of Amsterdam, Secretariat, Singel 425, 1012 WP Amsterdam, The Netherlands. You

will be contacted as soon as possible.

(2)

P-glycoproteinn in human immunodeficiency virus

typee 1 infection and therapy

S.U.C.. Sankatsing, J.H. Beijnen, A.H. Schinkel, J.MA Lange,

J.M.. Prins

(3)
(4)

Withh the advent and widespread use of potent antiretroviral therapy in the

mid-1990s,, the clinical course of human immunodefiency virus (HIV) type 1

(HIV-1)) infection has changed dramatically in a substantial proportion of HIV-1

infectedd individuals. This has led to a significant decline in the incidence of

AIDSS and AIDS-related morbidity and mortality in the developed world

1

~*.

Proteasee inhibitors in combination with inhibitors of HIV-1 reverse

transcriptasee cause a dramatic reduction in plasma viremia, with the plasma

HIV-11 RNA load being below the limit of detection in many patients

5 6

.

However,, with the currently available drugs, complete eradication of HIV-1

fromm an infected person is not achieved because of the persistence of latently

infected,, resting CD4

+

T cells harbouring replication-competent HIV-1, and

becausee of ongoing low-level viral replication

7

"

14

. One cause of ongoing viral

replicationn can be suboptimal penetration of drugs into anatomical sanctuary

sitess like the central nervous system. It has been suggested that drug

transporterss like P glycoprotein (P-gp) may contribute to this suboptimal

penetration.. Such drug transporters might also lower intracellular drug levels,

therebyy limiting the therapeutic efficacy of antiviral drugs in peripheral blood

mononuclearr cells (PBMCs)

15

, including CD4

+

T cells

16

.

P-gp,, a plasma membrane protein encoded by the multi drug resistance

(MDR)) gene, was discovered in 1976

17

and functions as an ATP-dependent

drugg efflux pump

18

. It is a transporter of a wide range of compounds,

includingg hydrophobic amphiphatic drugs, calcium channel blockers,

antihistamines,, peptides and steroids. The function of P-gp is thoroughly

investigatedd in the oncology field, because of its ability to induce resistance to

antii cancer therapy by pumping the drugs out of tumor cells

1920

. In this

minirevieww we summarize the possible roles of P-gp in HIV-1 infection and

therapy. .

Tissuee distribution of P-gp

P-gpp expression on cell membranes can be detected with monoclonal

antibodiess like MRK16 and UIC2

21

~

23

. P-gp function can be studied with the

fluorescentt dye rhodamine 123 (Rh 123). Cells expressing functional P-gp

havee less intracellular accumulation of rhodamine, because of increased efflux

(5)

off the dye by P-gp

2 1 2 2 2 4 2 5

. The level of intracellular Rh 123 accumulation can

bee measured by flow cytometry.

P-gpp is expressed at high levels in tissues like the gastrointestinal tract, liver

andd kidney and on capillary endothelial cells of the brain and on PBMCs

21,23,24,26-300

p

_

g p w a s f o u n d o n fresh

|

y

j

S 0

|

a t

ed CD4

+

and CD8

+

T cells. P-gp

expressionn increased on CD4

+

and CD8

+

T cells after stimulation with

phytohemagglutininn (PHA), with almost 100% of CD8

+

T cells expressing P-gp

afterr stimulation, suggesting that the level of P-gp expression on these cells

increasedd upon immune activation

31

. An overview of P-gp tissue distribution

iss given in table 1.

Tablee 1 Localization and function of P-gp

Tissuee and cells s Jejunum, , ileum,, colon Liver r Kidney y Pancreas s Lung g Heart t Adrenall gland Brain n Testes s Uterus s Bonee marow Immune e System m Site e Mucosall surface Zó"i'

Biliaryy canalicular surfaces of hepatocytess and apical surfacess of cells lining the smalll biliary ductules 3 Apicall surfaces of epithelial cellss of the proximal tubulus 23 Apicall surfaces of epithelial cellss lining the small ductules

23 3

Capillaryy endothelium ** Endotheliall luminal membraness of cardiac arterioless and capillaries 34,35 Surfacess of cells in the cortex andd medulla 23

Capillaryy endothelium M Capillariess A

Placentaa if

Hematopoieticc stem cells u

Naturall killer cells a

BB lymfocytes iV

CD4++ T cells 31 CD8++ T cells 31

(Possible)) function Preventionn of uptake and facilitation of excretionn across the mucosa 32 Excretionn of xenobiotics in bile JJ

Excretionn of xenobiotics in urine

Unknown n

Unknown n

ProtectionProtection against cardiac toxicity of certainn drugs 35

Secretionn of Cortisol and aldosterone in thee cortex36

ProtectionProtection against toxic substances M ProtectionProtection against toxic substances ** ProtectionProtection of the fetus against toxic substancess 37"39

ProtectionProtection against toxic substances ""; a rolee in differentiation and proliferation of stemm cells by influencing regulatory substancess 2; secretion of certain growth factorss and cytokines28

NKK cell mediated toxicity *, ,*w Unknown n

Secretionn of cytokines a

(6)

P-gpp function

Itt is not clear whether P-gp expression is necessary for a normal life. P-gp

knockoutt mice do not express P-gp but still have normal viability and fertility

andd a normal life span

29

-

33

'

42

"*

5

. it has been suggested that in vivo P-gp

protectss cells against toxic substances by efflux of these compounds. In the

gastrointestinall tract (duodenum, jejunum, ileum, cecum and colon), high

levelss of P-gp are located only on the mucosal surfaces of these tissues. This

suggestss that the role of P-gp is to prevent the uptake of toxic substrates and

perhapss to facilitate excretion across the mucosa of the gastrointestinal tract

23,32,466

T n e p

_gp

e x

p

r e s s e (

j j

n

| j

v e r

and kidney could be responsible for the

secretionn of xenobiotics into the bile and the urine

23,33

. The role of P-gp in the

pancreass and lungs is not known so far.

P-gpp in the blood-brain barrier could prevent the uptake of toxic substances

intoo the brain

33

-

47

-

50

and the absence of P-gp in mice resulted in increased

levelss of accumulation of many drugs in the brain

33

-

47

-

4

^

51

. Therefore, the

absencee of P-gp or the inhibition of P-gp might increase the central nervous

systemm toxicities of some drugs. This was indeed demonstrated for ivermectin,

domperidone,, loperamide and ritonavir in mice

33

-

49

-

51

. p-gp expressed in the

placentaa plays an important role in the protection of the developing fetus

againstt toxic substances

38,39

.

P-gpp expression on bone marrow stem cells might protect them from toxic

substancess and could also be responsible for the transport of certain growth

factorss and cytokines produced by stem cells

28

. It is unclear whether P-gp

alsoo has this function in other cell types.

Itt has been suggested that NK-cell cytotoxicity requires P-gp function for the

effluxx of lytic products. Inhibition of P-gp resulted in a decreased cytotoxicity of

NKK cells

40,41

. The decrease in the lytic activities of NK cells in the presence of

R-verapamil,, an inhibitor of P-gp, was dose dependent

52

. However, no

differencee in cytotoxic T-cell function was found between wild-type mice and

P-gpp knockout mice

53

. It is possible that P-gp knockout mice develop some

compensationn mechanism for the lack of P-gp.

(7)

Polymorphismm of P-gp

Thee MDR-1 (or ABCB1) gene encodes P-gp. So far, 28 single nucleotide

polymorphismss have been found at 27 positions

54,55

, and these are

sometimess linked to each other

56

. A C/T polymorphism in exon 26 (C3435T)

correlatess in a significant manner with P-gp expression and activity in the

duodenumm

55

. The polymorphism in exon 26 appears to be linked to single

nucleotidee polymorphisms in exon 12 (C1236T) and exon 21 (G2677T)

56

.

Individualss with the homozygous T genotype at position 3435 showed a P-gp

functionn higher than that of individuals with the homozygous C genotype

56

,

butt this is in contrast to the findings of another study, which demonstrated the

contraryy

55

. It has been suggested that the discrepancy between these studies

mightt reflect linkage to an as yet undefined promotor or enhancer region

polymorphism(s)) or nucleotide sequences that are important for messenger

RNAA processing

56

.

Thesee genetic differences might explain the differences in P-gp expression

betweenn individuals and, thereby, the differences in individual drug response

inn patients

56

"

58

. There is an ethnic difference in allele frequencies. Africans or

peoplee of African descent have a lower frequency of the T/T genotype and a

higherr frequency of the C/C genotype than Caucasians

5 6 , 5 9 , 6

.

Mechanismm of action

Thee most prevalent theory of the mechanism of action of P-gp favors a direct

pumpp mechanism

61

"

65

. The P-gp pump recognizes substrates through a

complexx substrate recognition region and directly pumps drugs out of the cell

byy using molecular mechanisms that are not yet well understood. ATP

hydrolysiss by P-gp provides the energy for this active process. The question

is,, how are the drugs actually pumped out of the cell? One proposed

mechanismm is that P-gp detects drugs and ejects them before they reach the

cytoplasmm by removing the drugs directly from the plasma membrane

20

'

66-68

.

Anotherr possibility is that P-gp acts as a flippase, carrying its substrate from

thee inner leaflet of the lipid bilayer to the outer leaflet

69

.

(8)

P-gpp inhibition

P-gpp inhibitors, also called reversal agents or P-gp modulators, inhibit the

effluxx of P-gp transported drugs in vitro and in mice

70

"

72

.

P-gpp inhibitors modulate P-gp function by competing with the binding and

transportt of the drug or through a non-competitive binding by binding either to

thee drug interaction site or to another modulator binding site, which leads to

allostericc changes

7374

. This is consistent with the finding that P-gp has at

leastt two independent drug-binding sites

74

.

Theree are three groups of P-gp inhibitors or modulators (table 2). The first

groupp of inhibitors are therapeutic agents. In vivo they function as a P-gp

inhibitorr only at concentrations higher than those required for therapeutic

activity.. Therefore, these agents cannot be used as P-gp inhibitors in vivo

becausee of their potential toxic effects. The second group of P-gp modulators

aree analogues of the first group of modulators. They are more potent and less

toxic.. For example emopamil, gallopamil and Ro11-2933 are analogues of

verapamil;; and PSC 833 is a non-immunosuppressive cyclosporin analogue.

Thee third group of modulators are developed and targeted against specific

MDRR mechanisms

19

. Clinical trials have demonstrated the beneficial effect of

P-gpp inhibitors for the treatment of cancer. One trial with breast cancer

patientss and one trial with lung cancer patients demonstrated a survival

benefitt with the addition of the P-gp inhibitor verapamil to chemotherapy

75,76

.

Inn another study, the combination of the P-gp inhibitor cyclosporine with

chemotherapyy statistically improved the relapse-free survival rate and the

overalll survival rate

77

. The proposed mechanism for this beneficial effect was

reversall of the P-gp induced drug efflux out of tumor cells. P-gp inhibitors can

causee side effects. The first group of P-gp inhibitors must be used at high

dosess to accomplish the inhibition of P-gp and can therefore cause side

effects.. In addition, side effects may be caused by the accumulation of

substratess of P-gp. The second and third groups of P-gp inhibitors are used at

muchh lower doses to accomplish the desired effect, and the side effects are

probablyy mainly caused by the accumulation of P-gp substrates

78

.

(9)

Tablee 2 Example of P-gp modulators

Firstt group Secondd group Thirdd group dexverapamil l Emopamil81 1 Gallopamil8186 6 PSCC 833 (valspodar) R011-293392 2 amiodaronee '9,8U bepridil83 3 caroverinee 85 clomipramine85 5 cyclosporinn A 90,91 diltiazemm ** felodipinee 95 isradipinee 95 nicardipinee M quinidine96 6 97 quinine e trifluoperazinee 85,98 II 85,99 verapamil l #

firstt generation modulators: therapeutic agents with a P-gp inhibitor effect at higher concentrationss than those required for therapeutic activity; &second generation modulators:: analogues of first generation modulators; +third generation modulators aree developed and targeted against specific MDR mechanisms.

GFF 120918c LYY 335979 6 OC144-093' ' VX-71089 9 XR90511 93

Relationn of P-gp and CYP3A4

Cytochromee P450 (CYP) mixed function oxidases account for the majority of oxidativee biotransformations of xenobiotics and endogenous compounds. Moree than 30 different human CYP enzymes have been identified, of which CYP3A44 appears to be one of the most important, as it contributes to the biotransformationn of approximately 60% of therapeutic drugs. Most of the activityy of CYP3A4 is located in the liver and in the small intestine, and CYP3A44 is responsible for the first-pass drug metabolism 1 0 . Some drugs, for examplee vinblastine, are substrates for both CYP3A4 and P-gp. Other drugs, forr example, verapamil are substrates for CYP3A4 and inhibitors of P-gp, Yet otherr drugs, for example ketoconazole, inhibit both CYP3A4 and P-gp 101/l02. Stt John's Wort contains compounds that are substrates for both P-gp and CYP3A44 and, in addition, induce intestinal P-gp and intestinal and hepatic CYP3A44 in humans 103'104.

P-gpp might enhance the metabolic effect of CYP3A4 in the small intestine in threee ways. First, P-gp can limit the uptake of substrates in the small intestine

(10)

therebyy limiting the amount of substrate that needs to be transformed by

CYP3A4.. Second, P-gp can increase the extent of metabolism by CYP3A4

throughh repeated cycles of intracellular uptake and efflux, thereby increasing

thee level of exposure of a drug to CYP3A4 before absorption in the systemic

circulation.. Finally, P-gp might preferentially remove drug metabolites

catalyzedd by CYP3A4, thereby limiting competitive inhibition

105

.

P-gpp expression and function during HIV-1 infection

Thee level of P-gp expression on T-cell and monocytic cell lines increases

uponn infection with HIV-1

106

. By using monoclonal antibody MRK-16, an

increasedd level of P-gp expression was established on CD4

+

T cells from

HIV-11 infected patients

28,107

. The level of P-gp expression on CD4

+

T cells

increasedd with the progression of HIV-1 infection

28,107

. Later studies did not

confirmm these findings, however

108-110

. in cells with increased levels of P-gp

expression,, one would expect a lower level of accumulation of Rh 123.

However,, CD4

+

, CD8

+

, CD16

+

and CD19

+

cells from HIV-1 infected patients

accumulatedd more Rh 123 than cells from healthy controls

107108

. in the

presencee of cyclosporin, a P-gp inhibitor, the level of intracellular

accumulationn of Rh 123 increased in CD4

+

and CD8

+

T cells from healthy

controls,, but in HIV-1 infected cells, the level of intracellular accumulation of

Rhh 123 did not increase further

107

. This suggests that even when the level of

P-gpp expression is increased in HIV-1-infected patients, the pump function of

P-gpp is decreased.

AA significantly reduced P-gp function was found in CD16

+

NK cells from HIV-1

infectedd patients compared with that found in cells from healthy individuals.

Thiss reduced P-gp function became more prominent with the progression of

thee HIV infection and was significantly correlated with a decreased NK-cell

cytotoxicc function

108

-

111

. This is consistent with the decreased NK-cell

cytotoxicityy in HIV-1 infected patients

112

. In vitro the reduced level of P-gp

expressionn and function of NK cells of HIV-1 infected individuals could be

recoveredd by stimulating them with IL-15

113

.

(11)

P-gpp expression and anti retroviral agents

Theree is clear evidence for interactions between P-gp activity and

antiretrovirall agents.

Onlyy a few studies have addressed the effect of P-gp on the behavior of

nucleosidee reverse transcriptase inhibitoirs (NRTIs). Early in vitro studies

demonstratedd a decreased level of accumulation of the NRTI zidovudine in

HIV-11 infected cells compared to that in uninfected cells, which was correlated

withh an increased level of P-gp expression on HIV-1 infected cells

106

.

Zidovudinee was less effective in inhibiting HIV-1 replication in cells expressing

P-gpp

1 U

. It may be possible that this effect was mediated by another drug

transporter,, such as MRP4, which was discovered later

115

. Zidovudine,

didanosinee and zalcitabine do not appear to induce P-gp expression or

functionn in the cells analyzed

107108

.

Thee non-nucleoside reverse transcriptase inhibitors (NNRTIs) nevirapine,

efavirenzz and delavirdine are not substrates for transport by P-gp in Caco-2

celll lines. All are able to induce P-gp expression and function, resulting in

increasess in the levels of P-gp expression of 3.5, 1.75 and 2.35 fold,

respectivelyy

116t

and reduced levels of accumulation of Rh123 by 72%, 8 1 %

andd 85%, respectively

116

. Delavirdine is not only an inducer of P-gp but also

ann inhibitor of P-gp

116

.

Alll presently available HIV-1 protease inhibitors are substrates for P-gp

51,70,71,117-1233

a n c

j j

n

t

e r a c

t

W

jth p.gp with an affinity in the order ritonavir >

nelfinavirr > amprenavir > indinavir > saquinavir

15,124

. CEM cells expressing

P-gpp have reduced intracellular concentrations of the protease inhibitors

ritonavir,, indinavir, saquinavir and nelfinavir compared with those in CEM cells

nott expressing P-gp

16,125

. The intracellular accumulation of saquinavir and

otherr protease inhibitors increased in the presence of the P-gp inhibitors

verapamill and GF 120918

16

. In patients, the concentrations of protease

inhibitorss in PBMCs was inversely correlated with the amount of MDR-1

mRNAA expression

15

. This explains why the protease inhibitors ritonavir,

indinavirr and saquinavir are less effective against HIV-1 replication in cells

(12)

expressingg P-gp

121

, although this effect of P-gp was refuted by other

investigatorss

124

. The different cell lines used in those studies might explain

thiss difference.

Furtherr in vitro studies demonstrated that ritonavir, indinavir, saquinavir and

nelfinavirr have weak inhibitory effects on P-gp, with ritonavir being the most

potentt one 24,51,70,118,122-124,126

0 n e s t u d y r e p o r t e c

|

t n a t r

j

t0

navir is a sixfold

moree potent P-gp inhibitor than the cyclosporine analog SDZ PSC 833, which

iss assumed to be one of the most potent inhibitors

12?

. However, that study

usedd cultured pig brain endothelial cells, in which the transporter(s) affected

wass not unambiguously identified. On the other hand, the simultaneous

administrationn of more than one protease inhibitor did not result in a

decreasedd P-gp efflux function in contrast to that after exposure to

LY-335979,, a more potent P-gp inhibitor

71

. This suggests that ritonavir is only a

moderatee P-gp inhibitor

51

. Nelfinavir and, to a lesser extent, its metabolite M8

aree also inhibitors of P-gp function on CD4

+

and CD8

+

T cells

128

. Protease

inhibitorss are also able to increase the level of P-gp expression. Lopinavir, for

instance,, is both an inhibitor and inducer of P-gp, but the overall effect of

lopinavirr seemed to be induction

129

.

Givenn the tissue distribution of P-gp (Table 1), it might lower the bioavailability

off protease inhibitors and could be responsible for the existence of sanctuary

sites,, such as the brain and the testes, by limiting the levels of accumulation

off protease inhibitors in these tissues. In P-gp knockout mice, the levels of

indinavir,, saquinavir and nelfinavir in plasma were 2- to 5-fold higher after oral

administrationn compared with the levels in wildtype mice, and the

concentrationss in the brain were 7- to 36-fold higher after intravenous

administrationn and 10-fold higher after oral administration compared with the

levelss in wildtype mice

120123

. in wiidtype mice intravenous administration of

thee potent P-gp inhibitor LY-335979 resulted in a dose dependent increase in

thee

14

C-labeled nelfinavir, amprenavir, indinavir and saquinavir concentrations

inn the brain and an increase in the

14

C-labeled nelfinavir in the testes

70

.

Likewise,, in the same model, an increase in plasma saquinavir concentrations

(13)

andd improved penetration into the brain and testes were seen after

co-exposuree to the potent P-gp inhibitor GF120918

130

. The penetration of

amprenavirr into the brains of rats increased in the presence of GF120918

131

.

Thesee data support the idea that P-gp expression in the gastrointestinal tract

cann limit the bioavailabilities of these drugs and that P-gp can contribute to the

decreasedd concentrations of these drugs in sanctuary sites like the brain and

thee male genital tract

123

-

132

.

Inn vivo, the addition of low-dose ritonavir increased indinavir plasma trough

concentrationss and indinavir concentrations in seminal plasma and

cerebrospinall fluid

132

. Inhibition of CYP3A4 by ritonavir decreases the

metabolismm of indinavir, but this could not fully explain the increased indinavir

concentrationss in seminal plasma and cerebrospinal fluid

132

. Therefore,

ritonavirr might also influence the blood-brain and blood-testis barrier by its

P-gpp inhibitory function

118

even though ritonavir being only a moderate P-gp

inhibitor

51,133

. .

Effectt of P-gp on HIV-1 replication

Finally,, P-gp can influence infectivity and replication of HIV-1.

Inn vitro, P-gp expression on T cells inhibited HIV-1 fusion with the plasma

membranee and also inhibited virus replication at a later step in the viral life

cycle.. This reduction in the level of HIV-1 replication correlated with the level

off P-gp expression

134135

. Mutations of P-gp at the ATP utilization site, which

therebyy inactivated ATP hydrolysis and resulted in an inactive P-gp pump

function,, still resulted in decreased HIV-1 infectivity. This suggests that the

P-gpp function is not necessary to block the infectivity of HIV-1

134

. On the other

hand,, when CD4

+

T cells were incubated with quinidine or PSC 833 to inhibit

thee P-gp function but not P-gp expression, the levels of HIV-1 production in

thesee cells increased. In summary, P-gp expression and function can inhibit

HIV-11 infectivity and replication capacity.

(14)

P-gpp polymorphisms in HIV-1

Polymorphismss in MDR1 alleles might be of clinical importance in HIV

treatment,, although controversy remains about the effects of the different

polymorphismss on P-gp expression and function. As discussed earlier, it is not

clearr whether a C or a T allele at position 3435 in exon 26 is associated with a

higherr level of P-gp function. Patients with a homozygous T genotype at exon

266 had, on average, a lower concentration of nelfinavir in plasma compared

withh that in the plasma of patients with the homozygous C genotype

136

. This

findingss suggests that the T allele is associated with a higher level of P-gp

function,, but this was difficult to reconcile with the fact that the level of P-gp

mRNAA transcription in PBMCs was lower in these patients. In that study

plasmaa efavirenz levels were also lower in patients with the homozygous T

genotype,, although efavirenz is not a substrate for P-gp. The patients with the

homozygouss T genotype had a greater rise in CD4

+

T-cell count

136

. This was

explainedd by the protective function of P-gp on pluripotent stem cells

22

.

P-gpp and cytochrome P450 3A4 during HIV treatment

Antii retroviral drugs can not only influence and be influenced by P-gp, but they

mayy also be substrates for and influenced by CYP3A4 activity. The resulting

effectt is not always predictable.

Thee NNRTI delavirdine, which is an inducer and inhibitor of P-gp, is also an

inhibitorr of CYP3A4

116

. Likewise, nevirapine and efavirenz are inducers not

onlyy of P-gp but also of CYP3A4

137

'

138

.

Nott only are the protease inhibitors substrates for P-gp, but all of them are

alsoo substrates for CYP3A4 in the liver and small intestine, where they are

metabolisedd by at least 80 to 95%

139

. In vitro studies revealed, in addition,

CYP3A44 inhibitory capacities of the protease inhibitors, with ritonavir being the

mostt potent inhibitor, followed by indinavir, nelfinavir, amprenavir and

saquinavir

140

"

145

. .

Inn vitro, P-gp facilitated the removal of the metabolites of indinavir, thereby

preventingg competition for CYP3A4 by the metabolites and indinavir itself

146

.

Itt has been suggested that this might increase the level of metabolism by

CYP3A44

105

but that result could not be confirmed

146

. That study

146

(15)

supportedd the suggestion that the mechanism of increased drug metabolism

byy CYP3A4 is through repeated cycles of intracellular uptake and efflux by

P-gp.. In vivo, low-dose ritonavir increased the bioavailability of indinavir by

inhibitingg metabolism by CYP3A4 and probably by inhibiting drug transport by

P-gp.. This combination proved to be clinically relevant

132147

.

Otherr drug transporters relevant for HIV therapy

Memberss of the MDR protein (MRP) family, which belongs to the group of

ATP-bindingg cassette drug transporters, have also been recognized as

transporterss of nucleoside-based antiretroviral drugs

115148

and protease

inhibitorss

244571

-

115

-

149

. For example, MRP4 overexpression impairs the

antivirall efficacies of adefovir and zidovudine in vitro

115

, and MRP 5

overexpressionn in vitro results in the efflux of adefovir from cells

148

. In vitro

studiess demonstrated that cells expressing MRP1 have a reduced intracellular

concentrationn of saquinavir and ritonavir compared with those in cells not

expressingg MRP1, but the intracellular concentrations of nelfinavir and

indinavirr were not influenced by MRP1 expression

125149

. Also, saquinavir,

ritonavirr and indinavir are effectively transported in vitro by MRP2

150

but not

byy MRP1, MRP3 or MRP5. The conflictingg data regarding the effect of MRP1

onn saquinavir, ritonavir and indinavir might be explained by the different cell

liness used in these studies. MRP drug transporters seem to play a minor role

inn the transport of ritonavir and indinavir across the blood-brain barrier

71

. In

vivo,, expression of MRP 1 on lymphocytes was correlated with lower levels of

accumulationn of saquinavir and ritonavir in these lymphocytes

151

.

Furthermore,, saquinavir and ritonavir are also inhibitors of MRP1 and MRP2

mediatedd drug transport

24118

'

124152

. it seems likely that there is a combined

effectt of P-gp and the MRP-drug transporter family on the concentrations of

antiretrovirall drugs that results in the decreased bioavailabilities of these

drugs. .

MRP11 also directly influences HIV-1 replication. Overexpression of MRP1 on

CEMM cells resulted in an increase in the level of HIV-1 replication by a factor

1600 compared to that in control CEM cells

135

. The mechanism is unknown so

far. .

(16)

Breastt Cancer Resistance Protein (BCRP) is another drug transporter with the

abilityy to influence intestinal (re)uptake and hepatobiliary excretion of

transportedd drugs

153

. Preliminary data showed that BCRP is not an efficient

transporterr of the protease inhibitors saquinavir, ritonavir and indinavir

150

but

cann transport zidovudine

154

.

Summaryy and conclusions

Manyy of the data to date is generated from in vitro studies with different cell

lines.. The use of different cell lines might explain in part why some of the data

conflict,, and the observed effects might be different for different tissues in

vivo.. Nevertheless, some aspects of P-gp relevant for HIV-infection and

therapyy have become clear. The potential effects of P-gp activity relevant for

HIV-11 treatment include decreasing the uptake of protease inhibitors together

withh CYP3A4 in the small intestine, decreasing the entry of protease inhibitors

intoo the central nervous system, decreasing the entry of protease inhibitors

intoo the testis, and decreasing the intracellular accumulation of protease

inhibitorss and protecting against HIV-1 infectivity and replication in CD4

+

T

cells.. P-gp activity is also potentially necessary for the function of natural

killerr cells.

Byy limiting the penetration of protease inhibitors into anatomical sites like the

centrall nervous system, P-gp contributes to the maintenance of sanctuary

sites.. P-gp function also reduces intracellular protease inhibitor concentrations

andd could therefore result in intracellular concentrations too low to block HIV-1

replicationn completely. These sub-optimal concentrations in sanctuary sites

andd in cells could contribute to ongoing low level HIV-1 replication. Moreover,

P-gpp can be an important limiting factor in the oral bioavailability of protease

inhibitors. .

Itt must be kept in mind that P-gp is not the only drug transporter of protease

inhibitors.. Other drug transporters, especially transporters of the MRP drug

transportt family, have been identified. The relative contribution of each of the

transporterss to the overall effect needs to be clarified.

(17)

Furthermore,, the additional effect of CYP3A4 on protease inhibitor

concentrationss is also important, although the resulting effect is not always

predictable. .

Thee clinical relevance of P-gp during treatment with NNRTIs is less clear.

However,, it is conceivable that in a regimen consisting of protease inhibitors

andd NNRTIs the induction of P-gp expression and function by the NNRTIs

mightt contribute to lower concentrations of the protease inhibitors in plasma

andd cells.

Theree are variations in P-gp expression and function among patients. These

variationss are explained at least in part by polymorphisms in the MDR-1 gene

encodingg P-gp, and this could contribute to the interpatient variabilities in

plasmaa protease inhibitor concentrations. There is still controversy about the

exactt effects of the different polymorphisms on P-gp function. Insight into the

significancee of these polymorphisms might be of clinical use. If the MDR-1

genotypee in patients is known before the start of treatment with protease

inhibitors,, one could predict which patients are at risk for having low plasma

proteasee inhibitor levels. This could justify a dose adjustment at the start of

treatment. .

Thee bioavailability and the intracellular concentrations of protease inhibitors

cann be increased in the presence of potent P-gp inhibitors. Therefore a logical

stepp in order to increase the concentrations of protease inhibitors in plasma,

cellscells and sanctuary sites is to inhibit P-gp function, which can be done with

availablee potent P-gp inhibitors. Inhibition of P-gp function might, however,

leadd to increased toxicities of other drugs, for example, loperamide and

domperidone.. Moreover, P-gp expression appears to decrease HIV-1

infectivityy and replication in T cells, and P-gp inhibition might therefore be

counterproductive. .

Beforee manipulation of P-gp function is considered in anti-HIV-1 therapy, the

questionn of which effect of P-gp is clinically more important must be

answered:: P-gp as a drug efflux pump or P-gp as a protector against HIV

infectivityy and replication.

(18)

References s

1.. Detels R, Munoz A, McFariane G et al. Effectiveness of potent antiretroviral therapyy on time to AIDS and death in men with known HIV infection duration.. Multicenter AIDS Cohort Study Investigators. JAMA. 1998;280:1497-1503. .

2.. Lee LM, Karon JM, Selik R et al. Survival After AIDS Diagnosis in Adolescentss and Adults During the Treatment Era, United States, 1984-1997.. JAMA. 2001;285:1308-1315.

3.. Mocroft A, Vella S, Benfield TL et al. Changing patterns of mortality across Europee in patients infected with HIV-1. EuroSIDA Study Group. Lancet. 1998;352:1725-1730. .

4.. Palella FJ, Delaney KM, Moorman AC et al. Declining morbidity and mortalityy among patients with advanced human immunodeficiency virus infection.. HIV Outpatient Study Investigators. N Engl J Med. 1998;338:853-860. .

5.. Gulick RM, Mellors JW, Havlir D et al. Treatment with indinavir, zidovudine, andd lamivudine in adults with human immunodeficiency virus infection and priorr antiretroviral therapy. N Engl J Med. 1997;337:734-739.

6.. Hammer SM, Squires KE, Hughes MD et al. A controlled trial of two nucleosidee analogues plus indinavir in persons with human immunodeficiencyy virus infection and CD4 cell counts of 200 per cubic millimeterr or less. AIDS Clinical Trials Group 320 Study Team. N Engl J

Med.Med. 1997;337:725-733.

7.. Furtado MR, Callaway DS, Phair JP et al. Persistence of HIV-1 transcription inn peripheral-blood mononuclear cells in patients receiving potent antiretrovirall therapy. N Engl J Med. 1999;340:1614-1622.

8.. Sharkey ME, Teo I, Greenough T et al. Persistence of episomal HIV-1 infectionn intermediates in patients on highly active anti-retroviral therapy.

NatNat Med. 2000;6:76-81.

9.. Wong JK, Hezareh M, Gunthard HF et al. Recovery of replication-competentt HIV despite prolonged suppression of plasma viremia. Science. 1997;278:1291-1295. .

10.. Finzi D, Hermankova M, Pierson T et al. Identification of a reservoir for HIV-11 in patients on highly active antiretroviral therapy. Science. 1997;278:1295-1300. .

(19)

11.. Chun TW, Stuyver L, Mizell SB et al. Presence of an inducible HIV-1 latent reservoirr during highly active antiretroviral therapy. Proc Natl Acad Sci U S

A.A. 1997;94:13193-13197.

12.. Chun TW, Carruth L, Finzi D et al. Quantification of latent tissue reservoirs andd total body viral load in HIV-1 infection. Nature. 1997;387:183-188. 13.. Yerly S, Kaiser L, Perneger TV et al. Time of initiation of antiretroviral

therapy:: impact on HIV-1 viraemia. The Swiss HIV Cohort Study. AIDS. 2000;14:243-249. .

14.. Dornadula G, Zhang H, VanUitert B et al. Residual HIV-1 RNA in blood plasmaa of patients taking suppressive highly active antiretroviral therapy.

JAMA.JAMA. 1999;282:1627-1632.

15.. Chaillou S, Durant J, Garraffo R et al. Intracellular concentration of protease inhibitorss in HIV-1-infected patients: correlation with MDR-1 gene expressionn and low dose of ritonavir. HIV Clin Trials. 2002;3:493-501. 16.. Jones K, Bray PG, Khoo SH et al. P-glycoprotein and transporter MRP1

reducee HIV protease inhibitor uptake in CD4 cells: potential for accelerated virall drug resistance? AIDS. 2001;15:1353-1358.

17.. Juliano RL, Ling V. A surface glycoprotein modulating drug permeability in Chinesee hamster ovary cell mutants. Biochim Biophys Acta. 1976;455:152-162. 1976;455:152-162.

18.. Sharom FJ, Yu X, Doige CA. Functional reconstitution of drug transport and ATPasee activity in proteoliposomes containing partially purified P-glycoprotein.. J Biol Chem. 1993;268:24197-24202.

19.. Krishna R, Mayer LD. Multidrug resistance (MDR) in cancer. Mechanisms, reversall using modulators of MDR and the role of MDR modulators in influencingg the pharmacokinetics of anticancer drugs. Eur J Pharm Sci. 2000;11:265-283. .

20.. Gottesman MM, Pastan I. Biochemistry of multidrug resistance mediated by thee multidrug transporter. Annu Rev Biochem. 1993;62:385-427.

21.. Chaudhary PM, Mechetner EB, Roninson IB. Expression and activity of the multidrugg resistance P-glycoprotein in human peripheral blood lymphocytes.

Blood.Blood. 1992;80:2735-2739.

22.. Chaudhary PM, Roninson IB. Expression and activity of P-glycoprotein, a multidrugg efflux pump, in human hematopoietic stem cells. Cell. 1991;66:85-94. .

(20)

23.. Thiebaut F, Tsuruo T, Hamada H et al. Cellular localization of the multidrug-resistancee gene product P-glycoprotein in normal human tissues. Proc Natl

AcadAcad Sci USA. 1987;84:7735-7738.

24.. Miller DS, Nobmann SN, Gutmann H et al. Xenobiotic transport across isolatedd brain microvessels studied by confocal microscopy. Mol Pharmacol. 2000;58:1357-1367. .

25.. Neyfakh AA. Use of fluorescent dyes as molecular probes for the study of multidrugg resistance. Exp Cell Res. 1988;174:168-176.

26.. Cordon-Cardo C, O'Brien JP, Casals D et al. Multidrug-resistance gene (P-glycoprotein)) is expressed by endothelial cells at blood-brain barrier sites.

ProcProc Natl Acad Sci USA. 1989;86:695-698.

27.. Fojo AT, Ueda K, Slamon DJ et al. Expression of a multidrug-resistance genee in human tumors and tissues. Proc Natl Acad Sci U SA. 1987;84:265-269. .

28.. Gupta S, Gollapudi S. P-glycoprotein (MDR 1 gene product) in cells of the immunee system: its possible physiologic role and alteration in aging and humann immunodeficiency virus-1 (HIV-1) infection. J Clin Immunol. 1993;13:289-301. .

29.. Holash JA, Stewart PA. The relationship of astrocyte-like cells to the vesselss that contribute to the blood-ocular barriers. Brain Res. 1993;629:218-224. .

30.. Thiebaut F, Tsuruo T, Hamada H et al. Immunohistochemical localization in normall tissues of different epitopes in the multidrug transport protein P170: evidencee for localization in brain capillaries and crossreactivity of one antibodyy with a muscle protein. J Histochem Cytochem. 1989;37:159-164. 31.. Gupta S, Kim CH, Tsuruo T et al. Preferential expression and activity of

multidrugg resistance gene 1 product (P-glycoprotein), a functionally active effluxx pump, in human CD8+ T cells: a role in cytotoxic effector function. J

ClinClin Immunol. 1992;12:451-458.

32.. Sparreboom A, van Asperen J, Mayer U et al. Limited oral bioavailability andd active epithelial excretion of paclitaxel (Taxol) caused by P-glycoprotein inn the intestine. Proc Natl Acad Sci USA. 1997;94:2031-2035.

33.. Schinkel AH, Smit JJ, van Tellingen O et al. Disruption of the mouse mdrla P-glycoproteinn gene leads to a deficiency in the blood-brain barrier and to increasedd sensitivity to drugs. Cell. 1994;77:491-502.

(21)

34.. Beaulieu E, Demeule M, Ghitescu L et al. P-glycoprotein is strongly expressedd in the luminal membranes of the endothelium of blood vessels in thee brain. Biochem J. 1997;326 ( Pt 2):539-544.

35.. Meissner K, Sperker B, Karsten C et al. Expression and localization of P-glycoproteinn in human heart: effects of cardiomyopathy. J Histochem

Cytochem.Cytochem. 2002;50:1351-1356.

36.. Ueda K, Okamura N, Hirai M et al. Human P-glycoprotein transports Cortisol,, aldosterone, and dexamethasone, but not progesterone. J Biol

Chem.Chem. 1992;267:24248-24252.

37.. Nakamura Y, Ikeda S, Furukawa T et al. Function of P-glycoprotein expressedd in placenta and mole. Biochem Biophys Res Commun. 1997;235:849-853. .

38.. Lankas GR, Wise LD, Cartwright ME et al. Placental P-glycoprotein deficiencyy enhances susceptibility to chemically induced birth defects in mice.. Reprod Toxicol. 1998;12:457-463.

39.. Smit JW, Huisman MT, van Tellingen O et al. Absence or pharmacological blockingg of placental P-glycoprotein profoundly increases fetal drug exposure.. J Clin Invest. Invest. 1999;104:1441-1447.

40.. Yamashiro T, Watanabe N, Yokoyama KK et al. Requirement of expression off P-glycoprotein on human natural killer leukemia cells for cell-mediated cytotoxicity.. Biochem Pharmacol. 1998;55:1385-1390.

41.. Chong AS, Markham PN, Gebel HM et al. Diverse multidrug-resistance-modificationn agents inhibit cytolytic activity of natural killer cells. Cancer

ImmunolImmunol Immunother. 1993;36:133-139.

42.. Borst P, Schinkel AH. What have we learnt thus far from mice with disrupted P-glycoproteinn genes? Eur J Cancer. 1996;32A:985-990.

43.. Schinkel AH, Mayer U, Wagenaar E et al. Normal viability and altered pharmacokineticss in mice lacking mdrl-type (drug-transporting) P-glycoproteins.. Proc Natl Acad Sci USA. 1997;94:4028-4033.

44.. Roepe PD. The role of the MDR protein in altered drug translocation across tumorr cell membranes. Biochim Biophys Acta. 1995;1241:385-405.

45.. Sawchuk RJ, Yang Z. Investigation of distribution, transport and uptake of anti-HIVV drugs to the central nervous system. Adv Drug Deliv Rev. 1999;39:5-31. .

46.. van Asperen J, van Tellingen O, Beijnen JH. The pharmacological role of P-glycoproteinn in the intestinal epithelium. Pharmacol Res. 1998;37:429-435.

(22)

47.. Borst P, Schinkel AH, Smit JJ et al. Classical and novel forms of multidrug resistancee and the physiological functions of P-glycoproteins in mammals.

PharmacolPharmacol Ther. 1993;60:289-299.

48.. van Asperen J, Mayer U, van Tellingen O et al. The functional role of P-glycoproteinn in the blood-brain barrier. J Pharm Sci. 1997;86:881-884. 49.. Schinkel AH, Wagenaar E, Mol CA et al. P-glycoprotein in the blood-brain

banierr of mice influences the brain penetration and pharmacological activity off many drugs. J Ciin Invest. 1996;97:2517-2524.

50.. Schinkel AH, Wagenaar E, Van Deemter L et al. Absence of the mdrla P-Glycoproteinn in mice affects tissue distribution and pharmacokinetics of dexamethasone,, digoxin, and cyclosporin A. J Clin Invest. 1995;96:1698-1705. .

51.. Huisman MT, Smit JW, Wiltshire HR et al. P-glycoprotein limits oral availability,, brain, and fetal penetration of saquinavir even with high doses off ritonavir. Mol Pharmacol. 2001;59:806-813.

52.. Ludescher C, Pall G, Irschick EU et al. Differential activity of P-glycoprotein inn normal blood lymphocyte subsets. Br J Haematol. 1998;101:722-727. 53.. Eisenbraun MD, Miller RA. mdrla-encoded P-glycoprotein is not required

forr peripheral T cell proliferation, cytokine release, or cytotoxic effector functionn in mice. J Immunol. 1999;163:2621-2627.

54.. Sakaeda T, Nakamura T, Okumura K. MDR1 genotype-related pharmacokineticss and pharmacodynamics. Biol Pharm Bull. 2002:25:1391-1400. .

55.. Hoffmeyer S, Burk O, von Richter O et al. Functional polymorphisms of the humann multidrug-resistance gene: multiple sequence variations and correlationn of one allele with P-glycoprotein expression and activity in vivo.

ProcProc Natl Acad Sci USA. 2000;97:3473-3478.

56.. Kim RB, Leake BF, Choo EF et al. Identification of functionally variant MDR11 alleles among European Americans and African Americans. Clin

PharmacolPharmacol Ther. 2001;70:189-199.

57.. Brinkmann U, Eichelbaum M. Polymorphisms in the ABC drug transporter genee MDR1. Pharmacogenomics J. 2001;1:59-64.

58.. Kurata Y, leiri I, Kimura M et al. Role of human MDR1 gene polymorphism inn bioavailability and interaction of digoxin, a substrate of P-glycoprotein.

(23)

59.. Ameyaw MM, Regateiro F, Li T et al. MDR1 pharmacogenetics: frequency off the C3435T mutation in exon 26 is significantly influenced by ethnicity.

Pharmacogenetics.Pharmacogenetics. 2001; 11:217-221.

60.. Schaeffeler E, Eichelbaum M, Brinkmann U et al. Frequency of C3435T

polymorphismpolymorphism of MDR1 gene in African people. Lancet. 2001;358:383-384. 61.. Callaghan R, Berridge G, Ferry DR et al. The functional purification of

P-glycoproteinn is dependent on maintenance of a lipid-protein interface.

BiochimBiochim Biophys Acta. 1997;1328:109-124.

62.. Ramachandra M, Ambudkar SV, Chen D et al. Human P-glycoprotein exhibitss reduced affinity for substrates during a catalytic transition state.

Biochemistry.Biochemistry. 1998;37:5010-5019.

63.. Bruggemann EP, Germann UA, Gottesman MM et al. Two different regions off P-glycoprotein [corrected] are photoaffinity-labeled by azidopine. J Biol

Chem.Chem. 1989;264:15483-15488.

64.. Cornwell MM, Gottesman MM, Pastan IH. Increased vinblastine binding to membranee vesicles from multidrug-resistant KB cells. J Biol Chem. 1986;261:7921-7928. .

65.. Greenberger LM, Lisanti CJ, Silva JT et al. Domain mapping of the photoaffinityy drug-binding sites in P-glycoprotein encoded by mouse mdrl b.

JJ Biol Chem. 1991 ;266:20744-20751.

66.. Homolya L, Hollo Z, Germann UA et al. Fluorescent cellular indicators are extrudedd by the multidrug resistance protein. J Biol Chem. 1993;268:21493-21496. .

67.. Raviv Y, Pollard HB, Bruggemann EP et al. Photosensitized labeling of a functionall multidrug transporter in living drug-resistant tumor cells. J Biol

Chem.Chem. 1990;265:3975-3980.

68.. Stein WD, Cardarelli C, Pastan I et al. Kinetic evidence suggesting that the multidrugg transporter differentially handles influx and efflux of its substrates.

MolMol Pharmacol. 1994;45:763-772.

69.. Higgins CF, Gottesman MM. Is the multidrug transporter a flippase? Trends

BiochemBiochem Sci. 1992;17:18-21.

70.. Choo EF, Leake B, Wandel C et al. Pharmacological inhibition of P-glycoproteinn transport enhances the distribution of HIV-1 protease inhibitors intoo brain and testes. Drug Metab Dispos. 2000;28:655-660.

(24)

71.. van der Sandt CJ, Vos CM, Nabulsi L et al. Assessment of active transport off HIV protease inhibitors in various cell lines and the in vitro blood-brain barrier.. AIDS. 2001;15:483-491.

72.. Mayer U, Wagenaar E, Dorobek B et al. Full blockade of intestinal P-glycoproteinn and extensive inhibition of blood-brain barrier P-glycoprotein by orall treatment of mice with PSC833. J Clin Invest. 1997;100:2430-2436. 73.. Garrigos M, Mir LM, Orlowski S. Competitive and non-competitive inhibition

off the multidrug-resistance-associated P-glycoprotein ATPase-further experimentall evidence for a multisite model. Eur J Biochem. 1997;244:664-673. .

74.. Dey S, Ramachandra M, Pastan I et al. Evidence for two nonidentical drug-interactionn sites in the human P-glycoprotein. Proc Natl Acad Sci USA. 1997;94:10594-10599. .

75.. Belpomme D, Gauthier S, Pujade-Lauraine E et al. Verapamil increases the survivall of patients with anthracycline-resistant metastatic breast carcinoma.

AnnAnn Oncol. 2000;11:1471-1476.

76.. Millward MJ, Cantwell BM, Munro NC et al. Oral verapamil with chemotherapyy for advanced non-small cell lung cancer: a randomised study.. Br J Cancer. 1993;67:1031-1035.

77.. List AF, Kopecky KJ, Willman CL et al. Benefit of cyclosporine modulation of drugg resistance in patients with poor-risk acute myeloid leukemia: a Southwestt Oncology Group study. Blood. 2001;98:3212-3220.

78.. Tan B, Piwnica-Worms D, Ratner L. Multidrug resistance transporters and modulation.. CurrOpin Oncol. 2000;12:450-458.

79.. Chauffert B, Rey D, Coudert B et al. Amiodarone is more efficient than verapamill in reversing resistance to anthracyclines in tumour cells. Br J

Cancer.Cancer. 1987;56:119-122.

80.. Chauffert B, Martin M, Hammann A et al. Amiodarone-induced enhancementt of doxorubicin and 4'-deoxydoxorubicin cytotoxicity to rat colonn cancer cells in vitro and in vivo. Cancer Res. 1986;46:825-830. 81.. Pirker R, Keilhauer G, Raschack M et al. Reversal of multi-drug resistance

inn human KB cell lines by structural analogs of verapamil. Int J Cancer. 1990;45:916-919. .

82.. Hyafil F, Vergely C, Du VP et al. In vitro and in vivo reversal of multidrug resistancee by GF120918, an acridonecarboxamide derivative. Cancer Res. 1993;53:4595-4602. .

(25)

83.. Schuurhuis GJ, Broxterman HJ, van der Hoeven J J et al. Potentiation of doxorubicinn cytotoxicity by the calcium antagonist bepridil in anthracycline-resistantt and -sensitive cell lines. A comparison with verapamil. Cancer

ChemotherChemother Pharmacol. 1987;20:285-290.

84.. Dantzig AH, Shepard RL, Cao J et al. Reversal of P-glycoprotein-mediated multidrugg resistance by a potent cyclopropyldibenzosuberane modulator, LY335979.. CancerRes. 1996;56:4171-4179.

85.. Tsuruo T, lida H, Tsukagoshi S et al. Increased accumulation of vincristine andd adriamycin in drug-resistant P388 tumor cells following incubation with calciumm antagonists and calmodulin inhibitors. Cancer Res. 1982;42:4730-4733. .

86.. Toffoli G, Simone F, Corona G et al. Structure-activity relationship of verapamill analogs and reversal of multidrug resistance. Biochem

Pharmacol.Pharmacol. 1995;50:1245-1255.

87.. Newman MJ, Rodarte JC, Benbatoul KD et al. Discovery and characterizationn of OC144-093, a novel inhibitor of P-glycoprotein-mediated multidrugg resistance. CancerRes. 2000;60:2964-2972.

88.. Boesch D, Muller K, Pourtier-Manzanedo A et al. Restoration of daunomycinn retention in multidrug-resistant P388 cells by submicromolar concentrationss of SDZ PSC 833, a nonimmunosuppressive cyclosporin derivative.. Exp Cell Res. Res. 1991;196:26-32.

89.. Germann UA, Shlyakhter D, Mason VS et al. Cellular and biochemical characterizationn of VX-710 as a chemosensitizer: reversal of P-glycoprotein-mediatedmediated multidrug resistance in vitro. Anticancer Drugs. 1997;8:125-140. 90.. Twentyman PR, Fox NE, White DJ. Cyclosporin A and its analogues as

modifierss of adriamycin and vincristine resistance in a multi-drug resistant humann lung cancer cell line. Br J Cancer. 1987;56:55-57.

91.. Nooter K, Oostrum R, Jonker R et al. Effect of cyclosporin A on daunorubicinn accumulation in multidrug-resistant P388 leukemia cells measuredd by real-time flow cytometry. Cancer Chemother Pharmacol. 1989;23:296-300. .

92.. Alaoui-Jamali MA, Schecter RL, Rustum YM et al. In vivo reversal of doxorubicinn resistance by a new tiapamil analog Ro11-2933. J Pharmacol

(26)

93.. Dale IL, Tuffley W, Callaghan R et al. Reversal of P-glycoprotein-mediated multidrugg resistance by XR9051, a novel diketopiperazine derivative. Br J

Cancer.Cancer. 1998;78:885-892.

94.. Tsuruo T, lida H, Nojiri M et al. Circumvention of vincristine and Adriamycin resistancee in vitro and in vivo by calcium influx blockers. Cancer Res. 1983;43:2905-2910. .

95.. Hollt V, Kouba M, Dietel M et al. Stereoisomers of calcium antagonists whichh differ markedly in their potencies as calcium blockers are equally effectivee in modulating drug transport by P-glycoprotein. Biochem

Pharmacol.Pharmacol. 1992;43:2601-2608.

96.. Tsuruo T, lida H, Kitatani Y et al. Effects of quinidine and related compoundss on cytotoxicity and cellular accumulation of vincristine and adriamycinn in drug-resistant tumor cells. Cancer Res. 1984;44:4303-4307. 97.. Solary E, Velay I, Chauffert B et al. Sufficient levels of quinine in the serum

circumventt the multidrug resistance of the human leukemic cell line K562/ADM.. Cancer. 1991;68:1714-1719.

98.. Ganapathi R, Grabowski D. Enhancement of sensitivity to adriamycin in resistantt P388 leukemia by the calmodulin inhibitor trifluoperazine. Cancer

Res.Res. 1983;43:3696-3699.

99.. Tsuruo T, lida H, Tsukagoshi S et al. Overcoming of vincristine resistance in P3888 leukemia in vivo and in vitro through enhanced cytotoxicity of vincristinee and vinblastine by verapamil. Cancer Res. 1981;41:1967-1972. 100.. Dresser GK, Spence JD, Bailey DG. Pharmacokinetic-pharmacodynamic

consequencess and clinical relevance of cytochrome P450 3A4 inhibition.

Clin Pharmacokinet.Clin Pharmacokinet. 2000;38:41-57.

101.. Wacher VJ, Wu CY, Benet LZ. Overlapping substrate specificities and tissuee distribution of cytochrome P450 3A and P-glycoprotein: implications forr drug delivery and activity in cancer chemotherapy. Mol Carcinog. 1995;13:129-134. .

102.. Zhang Y, Benet LZ. The gut as a barrier to drug absorption: combined role off cytochrome P450 3A and P-glycoprotein. Clin Pharmacokinet. 2001;40:159-168. .

103.. Durr D, Stieger B, Kullak-Ublick GA et al. St John's Wort induces intestinal P-glycoprotein/MDR11 and intestinal and hepatic CYP3A4. Clin Pharmacol

(27)

104.. Hennessy M, Kelleher D, Spiers JP et at. St Johns wort increases expressionn of P-glycoprotein: implications for drug interactions. Br J Clin

Pharmacol.Pharmacol. 2002;53:75-82.

105.. Watkins PB. The barrier function of CYP3A4 and P-glycoprotein in the small bowel.. Adv Drug Deliv Rev. 1997;27:161-170.

106.. Gollapudi S, Gupta S. Human immunodeficiency virus l-induced expression off P-glycoprotein. Biochem Biophys Res Commun. 1990;171:1002-1007. 107.. Andreana A, Aggarwal S, Gollapudi S et al. Abnormal expression of a

170-kilodaltonn P-glycoprotein encoded by MDR1 gene, a metabolically active effluxx pump, in CD4+ and CD8+ T cells from patients with human immunodeficiencyy virus type 1 infection. AIDS Res Hum Retroviruses. 1996;12:1457-1462. .

108.. Lucia MB, Cauda R, Landay AL et al. Transmembrane P-glycoprotein (P-gp/P-170)) in HIV infection: analysis of lymphocyte surface expression and drug-unrelatedd function. AIDS Res Hum Retroviruses. 1995; 11:893-901. 109.. Lucia MB, Rutella S, Leone G et al. In vitro and in vivo modulation of

MDR1/P-glycoproteinn in HIV-infected patients administered highly active antiretrovirall therapy and liposomal doxorubicin. J Acquir Immune Defic

Syndr.Syndr. 2002;30:369-378.

110.. Meaden ER, Hoggard PG, Maher B et al. Expression of P-glycoprotein and multidrugg resistance-associated protein in healthy volunteers and HIV-infectedd patients. AIDS Res Hum Retroviruses. 2001;17:1329-1332. 111.. Lucia MB, Cauda R, Malorni W et al. P-170 glycoprotein (P-170) is involved

inn the impairment of natural killer cell-mediated cytotoxicity in HIV+ patients.

ImmunolImmunol Lett. 1995;47:223-226.

112.. Brenner BG, Dascal A, Margolese RG et al. Natural killer cell function in patientss with acquired immunodeficiency syndrome and related diseases. J

LeukocBiol.LeukocBiol. 1989;46:75-83.

113.. Chang KH, Kim JM, Yoo NC et al. Restoration of P-glycoprotein function is involvedd in the increase of natural killer activity with exogenous interleukin-155 in human immunodeficiency virus-infected individuals. Yonsei Med J. 2000;41:600-606. .

114.. Antonelli G, Turriziani O, Cianfriglia M et al. Resistance of HIV-1 to AZT mightt also involve the cellular expression of multidrug resistance P-glycoprotein.. AIDS Res Hum Retroviruses. 1992;8:1839-1844.

(28)

115.. Schuetz JD, Connelly MC, Sun D et al. MRP4: A previously unidentified factorr in resistance to nucleoside-based antiviral drugs. Nat Med.

1999;5:1048-1051. .

116.. Stormer E, von Moltke LL, Perloff MD et al. Differential modulation of P-glycoproteinn expression and activity by non-nucleoside HIV-1 reverse transcriptasee inhibitors in cell culture. Pharm Res. 2002;19:1038-1045. 117.. Clayette P, Jorajuria S, Dormont D. Significance of P-glycoprotein for the

pharmacologyy and clinical use of HIV protease inhibitors. AIDS. 2000;14:235-236. .

118.. Gutmann H, Fricker G, Drewe J et al. Interactions of HIV protease inhibitors withh ATP-dependent drug export proteins. Mol Pharmacol. 1999;56:383-389. .

119.. Kim AE, Dintaman JM, Waddell DS et al. Saquinavir, an HIV protease inhibitor,, is transported by P-glycoprotein. J Pharmacol Exp Ther. 1998;286:1439-1445. .

120.. Kim RB, Fromm MF, Wandel C et al. The drug transporter P-glycoprotein limitss oral absorption and brain entry of HIV-1 protease inhibitors. J Clin

Invest.Invest. 1998;101:289-294.

121.. Lee CG, Gottesman MM, Cardarelli CO et al. HIV-1 protease inhibitors are substratess for the MDR1 multidrug transporter. Biochemistry. 1998;37:3594-3601. .

122.. Profit L, Eagling VA, Back DJ. Modulation of P-glycoprotein function in humann lymphocytes and Caco-2 cell monolayers by HIV-1 protease inhibitors.. AIDS. 1999;13:1623-1627.

123.. Washington CB, Wiltshire HR, Man M et al. The disposition of saquinavir in normall and P-glycoprotein deficient mice, rats, and in cultured cells. Drug

MetabMetab Dispos. 2000;28:1058-1062.

124.. Srinivas RV, Middlemas D, Flynn P et al. Human immunodeficiency virus proteasee inhibitors serve as substrates for multidrug transporter proteins MDR11 and MRP1 but retain antiviral efficacy in cell lines expressing these transporters.. Antimicrob Agents Chemother. 1998;42:3157-3162.

125.. Jones K, Hoggard PG, Sales SD et al. Differences in the intracellular accumulationn of HIV protease inhibitors in vitro and the effect of active transport.. AIDS. 2001;15:675-681.

(29)

126.. Shiraki N, Hamada A, Yasuda K et al. Inhibitory effect of human immunodeficiencyy virus protease inhibitors on multidrug resistance transporterr P-glycoproteins. BiolPharm Bull. 2000;23:1528-1531.

127.. Drewe J, Gutmann H, Fricker G et al. HIV protease inhibitor ritonavir: a moree potent inhibitor of P-glycoprotein than the cyclosporine analog SDZ PSCC 833. Biochem Pharmacol. 1999;57:1147-1152.

128.. Donahue JP, Dowdy D, Ratnam KK et al. Effects of nelfinavir and its M8 metabolitee on lymphocyte P-glycoprotein activity during antiretroviral therapy.. Clin Pharmacol Ther. 2003;73:78-86.

129.. Vishnuvardhan D, Moltke LL, Richert C et al. Lopinavir: acute exposure inhibitss P-glycoprotein; extended exposure induces P-glycoprotein. AIDS. 2003;17:1092-1094. .

130.. Huisman MT, Smit JW, Wiltshire HR et al. Assessing safety and efficacy of directedd P-glycoprotein inhibition to improve the pharmacokinetic properties off saquinavir coadministered with ritonavir. J Pharmacol Exp Ther. 2003;304:596-602. .

131.. Edwards JE, Brouwer KR, McNamara PJ. GF120918, a P-glycoprotein modulator,, increases the concentration of unbound amprenavir in the centrall nervous system in rats. Antimicrob Agents Chemother. 2002;46:2284-2286. .

132.. van Praag RM, Weverling GJ, Portegies P et al. Enhanced penetration of indinavirr in cerebrospinal fluid and semen after the addition of low-dose ritonavir.. AIDS. 2000;14:1187-1194.

133.. Katoh M, Nakajima M, Yamazaki H et al. Inhibitory effects of CYP3A4 substratess and their metabolites on P-glycoprotein-mediated transport. Eur

JPharmJPharm Sci. 2001;12:505-513.

134.. Lee CG, Ramachandra M, Jeang KT et al. Effect of ABC transporters on HIV-11 infection: inhibition of virus production by the MDR1 transporter.

FASEBFASEB J. 2000;14:516-522.

135.. Speck RR, Yu XF, Hildreth J et al. Differential effects of p-glycoprotein and multidrugg resistance protein-1 on productive human immunodeficiency virus infection.. J Infect Dis. 2002;186:332-340.

136.. Fellay J, Marzolini C, Meaden ER et al. Response to antiretroviral treatment inn HIV-1-infected individuals with allelic variants of the multidrug resistance transporterr 1: a pharmacogenetics study. Lancet. 2002;359:30-36.

(30)

137.. Erickson DA, Mather G, Trager WF et al. Characterization of the in vitro biotransformationn of the HIV-1 reverse transcriptase inhibitor nevirapine by humann hepatic cytochromes P-450. Drug Metab Dispos. 1999;27:1488-1495. .

138.. von Moltke LL, Greenblatt DJ, Granda BW et al. Inhibition of human cytochromee P450 isoforms by nonnucleoside reverse transcriptase inhibitors.. J Clin Pharmacol. 2001,41:85-91.

139.. Barry M, Mulcahy F, Merry C et al. Pharmacokinetics and potential interactionss amongst antiretroviral agents used to treat patients with HIV infection.. Clin Pharmacokinet. Pharmacokinet. 1999;36:289-304.

140.. Fitzsimmons ME, Collins JM. Selective biotransformation of the human immunodeficiencyy virus protease inhibitor saquinavir by human small-intestinall cytochrome P4503A4: potential contribution to high first-pass metabolism.. Drug Metab Dispos. 1997;25:256-266.

141.. Koudriakova T, latsimirskaia E, Utkin I et al. Metabolism of the human immunodeficiencyy virus protease inhibitors indinavir and ritonavir by human intestinall microsomes and expressed cytochrome P4503A4/3A5: mechanism-basedd inactivation of cytochrome P4503A by ritonavir. Drug

MetabMetab Dispos. 1998;26:552-561.

142.. Kumar GN, Dykstra J, Roberts EM et al. Potent inhibition of the cytochrome P-4500 3A-mediated human liver microsomal metabolism of a novel HIV proteasee inhibitor by ritonavir: A positive drug-drug interaction. Drug Metab

Dispos.Dispos. 1999;27:902-908.

143.. Eagling VA, Back DJ, Barry MG. Differential inhibition of cytochrome P450 isoformss by the protease inhibitors, ritonavir, saquinavir and indinavir. Br J

ClinClin Pharmacol. 1997;44:190-194.

144.. Yamaji H, Matsumura Y, Yoshikawa Y et al. Pharmacokinetic interactions betweenn HIV-protease inhibitors in rats. Biopharm Drug Dispos. 1999;20:241-247. .

145.. Kempf DJ, Marsh KC, Kumar G et al. Pharmacokinetic enhancement of inhibitorss of the human immunodeficiency virus protease by coadministrationn with ritonavir. Antimicrob Agents Chemother. 1997 ;41:654-660. .

146.. Hochman JH, Chiba M, Nishime J et al. Influence of P-glycoprotein on the transportt and metabolism of indinavir in Caco-2 cells expressing cytochromee P-450 3A4. J Pharmacol Exp Ther. 2000;292:310-318.

(31)

147.. van Heeswijk RP, Veldkamp Al, Hoetelmans RM et al. The steady-state plasmaa pharmacokinetics of indinavir alone and in combination with a low dosee of ritonavir in twice daily dosing regimens in HIV-1 -infected individuals.

AIDS.AIDS. 1999;13:F95-F99.

148.. Wijnholds J, Mol CA, Van Deemter L et al. Multidrug-resistance protein 5 is aa multispecific organic anion transporter able to transport nucleotide analogs.. Proc Natl Acad Sci USA. 2000;97:7476-7481.

149.. Williams GC, Liu A, Knipp G et al. Direct evidence that saquinavir is transportedd by multidrug resistance-associated protein (MRP1) and canalicularr multispecific organic anion transporter (MRP2). Antimicrob

AgentsAgents Chemother. 2002;46:3456-3462.

150.. Huisman MT, Smit JW, Crommentuyn KM et al. Multidrug resistance protein 22 (MRP2) transports HIV protease inhibitors, and transport can be enhancedd by other drugs. AIDS. 2002;16:2295-2301.

151.. Meaden ER, Hoggard PG, Newton P et al. P-glycoprotein and MRP1 expressionn and reduced ritonavir and saquinavir accumulation in HIV-infectedd individuals. J Antimicrob Chemother. 2002;50:583-588.

152.. Olson DP, Scadden DT, D'Aquila RT et al. The protease inhibitor ritonavir inhibitss the functional activity of the multidrug resistance related-protein 1 (MRP-1).. AIDS. 2002;16:1743-1747.

153.. Jonker JW, Smit JW, Brinkhuis RF et al. Role of breast cancer resistance proteinn in the bioavailability and fetal penetration of topotecan. J Natl

CancerCancer Inst. 2000;92:1651-1656.

154.. Wang X, Furukawa T, Nitanda T et al. Breast cancer resistance protein (BCRP/ABCG2)) induces cellular resistance to HIV-1 nucleoside reverse transcriptasee inhibitors. Mol Pharmacol. 2003;63:65-72.

Referenties

GERELATEERDE DOCUMENTEN

We test these theoretical predictions using a laboratory experiments that in a between- subjects design varies the polarization of party positions and whether voting is mandatory

party is centrist. “Voluntary” is a dummy variable that is 1 if voting is voluntary. Standard errors given in brackets are clustered at the matching group level. For the

therefore evaluate it at most left-wing entry position possible since this leads to the most favorable outcome for the entrant. Therefore no extreme candidate will enter to

Comparing entry rates across electoral rules I find that the higher entry rates under proportional representation compared to plurality rule when the costs of entry are low are

Furthermore, player 3 shows a lower probability of accepting the middle option when payoffs are asymmetric but this effect is not significant (p-value: 0.23 when probabilities

Der Mechanismus, welcher diesem Effekt zu Grunde liegt ist, dass, wenn es eine Möglichkeit gibt der Wahl fern zu bleiben, die Unterstützer extremer Parteien seltener aus

Smithson and Foucault are writing, of course, from different positions and with different purposes; nonetheless this coincidence of terminology offers an opportunity to consider to