• No results found

Limitations of current antiretroviral therapy in HIV-1 infection: the search for new strategies - CHAPTER 7 Effect of mycophenolate mofetil on the pharmacokinetics of antiretroviral drugs and the intracellular nucleoside

N/A
N/A
Protected

Academic year: 2021

Share "Limitations of current antiretroviral therapy in HIV-1 infection: the search for new strategies - CHAPTER 7 Effect of mycophenolate mofetil on the pharmacokinetics of antiretroviral drugs and the intracellular nucleoside "

Copied!
17
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)

Effectt of mycophenolate mofetil on the

pharmacokineticss of antiretroviral drugs and the

intracellularr nucleoside triphosphate pools

S.U.C.. Sankatsing, P.G. Hoggard, A.D.R. Huitema, R.W.. Sparidans, S. Kewn, K.M.L. Crommentuyn, J.M.A.. Lange, J.H. Beijnen, D.J. Back, J.M. Prins

(3)

Abstract t

Objective:Objective: To study the effect of mycophenolate mofetil (MMF) therapy on the

pharmacokineticc parameters of a number of antiretroviral drugs, and on the intracellularr deoxycytidine triphosphate (dCTP) and deoxyguanosine triphosphatee (dGTP) pools and triphosphate concentrations of lamivudine (3TCTP)) and abacavir (CBVTP).

Design:Design: Randomised pharmacokinetic study.

Participants:: 19HIV-1 infected patients.

Methods:Methods: Antiretroviral naive men starting treatment with didanosine 400 mg

QD,, lamivudine 150 mg BID, abacavir 300 mg BID, indinavir 800 mg BID, ritonavirr 100 mg BID and nevirapine 200 mg BID were randomized to a group withh or without MMF 500 mg BID. After 8 weeks of therapy the plasma pharmacokineticc profiles of mycophenolic acid (MPA, the active metabolite of MMF),, abacavir, indinavir and nevirapine, and the triphosphate concentrations (dCTP,, dGTP and 3TCTP) in peripheral blood mononuclear cells (PBMCs) weree determined.

Results:Results: Nine of the 19 patients received MMF. There was no difference in

plasmaa clearance of indinavir or abacavir between the 2 groups. The clearancee of nevirapine was higher in patients using MMF (p=0.04). In 12 patients,, of whom 5 also received MMF, intracellular triphosphates were measured.. There was no significant difference in intracellular dCTP, dGTP or 3TCTPP concentrations between the 2 groups. Due to assay interference CBVTPP could not be determined.

Conclusion:Conclusion: In this small cohort of patients MMF therapy reduced the plasma

concentrationn of nevirapine but had no effect on plasma indinavir and abacavir concentrations.. In contrast to our hypothesis, there were no consistent effects off MPA on the intracellular concentrations of dCTP, dGTP or 3TCTP.

(4)

Introduction n

Generallyy accepted therapy for HIV-1 consists of at least 3 different antiretrovirall drugs, usually from two different classes. We have demonstrated previouslyy that although a triple class 5-drug regimen resulted in a better suppressionn of the HIV-1 replication than a 3-drug regimen 1*2, HIV-1 replicationn was nevertheless not completely suppressed. Therefore new drugs orr new approaches with current drugs are necessary to further suppress this soo called residual replication 3. HIV-1 can only replicate in activated CD4+ T cellss 4, so strategies to limit the number of target cells might be an option. The usee of hydroxyurea, which reduces the availability of suitable target cells for HIVV by killing the dividing CD4+ T cells 5 and in vitro lowers endogenous deoxynucleosidee triphosphates 6, is such an approach. However, hydroxyurea cann result in serious toxicity and blunts the CD4+ T cell replenishment resultingg from effective antiretroviral therapy, and its effectiveness in HIV therapyy is disappointing 7.

Anotherr approach is the use of mycophenolate mofetil (MMF) in combination withh highly active antiretroviral therapy (HAART) 8 9. MMF is used to prevent organn rejection and to treat acute rejection after organ transplantation 10. It is aa prodrug that is rapidly converted into its active metabolite mycophenolic acid (MPA),, which selectively inhibits inosine 5'-monophosphate dehydrogenase (IMPDH)) conversion of inosine monophosphate (IMP) to xanthosine 5'-monophosphatee (XMP) in the de novo synthetic pathway of the purine guaninee 11,12. Blocking this enzyme in lymphocytes depletes guanosine triphosphatee (GTP) and deoxyguanosine triphosphate (dGTP) pools. This depletionn of dGTP pools inhibits the activation of T lymphocytes, thereby limitingg the availability of target cells for HIV. MPA also has a direct antiviral effectt on HIV itself, as dGTP is necessary for HIV replication 8'9.

Thee HIV reverse transcriptase inhibitor abacavir (ABC) is an analogue of guanosinee 13,14. Abacavir is phosphorylated to its active form carbovir triphosphatee (CBVTP), which competes with endogenous dGTP for incorporationn into the growing viral DNA chain. The MPA mediated depletion off endogenous dGTP may result in an altered competition between CBVTP andd dGTP, thereby increasing the antiretroviral efficacy of ABC 8. It has

(5)

recentlyy been demonstrated that MPA also synergistically enhances the activityy of didanosine and tenofovir15.

Iff indeed MMF is to have a role in the treatment of HIV, a greater degree of understandingg is required about interactions between MPA and antiretroviral drugs,, as no such data are available at this time. Knowledge of such drug interactionss is also important in case of organ transplantation in HIV patients onn antiretroviral therapy, which is no longer a novelty 16. We therefore studied thee pharmacokinetic interactions between MPA and HAART and studied the effectt of MPA on the intracellular dGTP pools in peripheral blood mononuclear cellss (PBMC) in vivo.

Methods s

Patients Patients

Nineteenn antiretroviral naive patients were recruited in this study. All patients startedd with a triple class antiretroviral drug regimen consisting of didanosine entericc coated 400 mg QD, ABC 300 mg BID, lamivudine (3TC) 150 mg BID, indinavirr (IDV) 800 mg BID, ritonavir 100 mg BID to boost IDV plasma concentrationss and nevirapine (NVP) 200 mg BID (first two weeks 200 mg QD).. The patients were stratified for type of HIV infection (acute or chronic infection)) and randomized to a group with MMF or another without MMF. The firstt three patients randomized to MMF started with 500 mg BID, and after 4 weekss the dose was increased to 1000 mg BID. Because the plasma MPA concentrationss were well above the minimum desired concentration of 0.14 ug/mll 17, the dose was decreased to 500 mg BID after 12-24 weeks of treatmentt and all further patients randomized to MMF in the study used 500 mgg BID.

Thee local Medical Ethics Committee approved the study and written informed consentt was obtained from all patients.

T-cellT-cell proliferative capacity

T-celll proliferative capacity was determined in a whole-blood lymphocyte culturee assay as described previously in detail 18. Briefly, cells in 10 uJ of

(6)

heparinizedd blood were stimulated with mAb to CD3 in the presence of CD28 mAb.. Proliferative responses were measured after 4 days of culture by means off incorporation of [3H]-thymidine, added 24 hours before harvest. All cultures weree done in triplicate. Reactivity was calculated back to counts per minute (c.p.m.)) in 103 CD3+ T cells and results are expressed as percentages of the mediann responses in healthy controls investigated simultaneously.

PharmacokineticPharmacokinetic and intracellular triphosphate profiles

Afterr 8 weeks of therapy plasma pharmacokinetic profiles of MPA (the active metabolitee of MMF), ABC, IDV and NVP were determined during a dosing intervall in all patients. Heparinized blood was obtained at T=0,1, 2, 4, 6, 8,10 andd 12h after dosing. After centrifugation at 1500 g for 10 minutes, plasma wass collected and frozen until further analysis. On the same day at T=0, 2, 6 andd 12h after dosing, blood samples were taken in cell preparation tubes of 8 mll with sodium citrate (Beckton Dickinson's Vacutainer® CPT™, Beckton Dickinsonn and Company, Franklin Lakes, N.J., U.S.A.) and PBMCs were isolatedd for the measurement of the intracellular deoxycytidine triphosphate (dCTP)) and dGTP pools and triphosphate concentrations of 3TC (3TCTP) and ABCC (CBVTP). The CPT tubes were immediately centrifuged for 30 minutes at 15000 g and 21 C to obtain leukocyte rich plasma. Leukocyte rich plasma was centrifugedd for 10 minutes at 600 g and C to isolate the leukocytes. After discardingg the supernatant, the cell pellet was resuspended with 2.50 mL phosphatee buffered saline and (after removing 0.25 mL for a cell count) again centrifugedd for 10 minutes at 600 g and . After discarding the supernatant, thee remaining cell pellet was resuspended in 0.96 mL 50mM Tris-HCL, 1.44 mLL 100% methanol was added and the solution was stored overnight at . Thee next day the samples were centrifuged for 10 minutes at 600 g and C to gett rid of debris and the supernatant was dried by rotary evaporation and storedd at C until further analyses.

Assays Assays

Plasmaa concentrations of ABC and MPA were simultaneously measured with aa liquid chromatographic assay using dual spectrophotometric detection as

(7)

describedd earlier 19 which has been validated in the 0.05-2 pg/ml range for ABCC and in the 0.01-10 pg/ml range for MPA for 200 (Jl plasma samples. The lowerr limits of quantification (LLQ) are 0.05 and 0.01 ug/ml for ABC and MPA, respectivelyy with an intra- and interday variation of 6 and 1 % for ABC and 6 andd 5% for MPA.

Plasmaa concentrations of IDV were measured using liquid chromatography coupledd with electrospray tandem mass spectrometry (LC-MS/MS) 20 which hass been validated over a concentration of 0.01 to 10 ug/mL The intra- and interdayy variation is less than 10%, the (LLQ) is 0.01 ug/ml.

Plasmaa NVP concentrations were measured using high-performance liquid chromatographyy (HPLC) with ultraviolet detection. This assay is validated over thee range of 0.05-10 ug/ml using 250 uL of plasma, with an intra- and inter-dayy variation of less than 4.5% and a LLQ of 0.05 ug/ml21.

Analysiss of the intracellular endogenous and drug triphosphates were performedd using enzymatic assays as described earlier22. Intra- and inter-day variabilityy of the 3TC assay was 14 and 15% respectively. The LLQ was 0.05 pmoll for 3TCTP. Intra- and inter-assay variability of the endogenous deoxynucleosidee triphosphate assays were less than 10%. The LLQ of endogenouss triphosphates is 0.01 pmol.

StatisticalStatistical analysis

Dataa were analyzed using SPSS 11.0.1, (SPSS Inc. Chicago, Illinois). Pharmacokineticc parameters of MPA, ABC, IDV and NVP were calculated usingg non-compartmental analyses (WinNonLin, version 3, Pharsight, Mountainn View, U.S.A.). Baseline patient characteristics, the change in [3 H]-thymidinee incorporation and the influence of MPA on the pharmacokinetic parameterss of ABC, IDV and NVP and the intracellular triphosphate concentrationss were compared using the Mann-Whitney U-test. In this analysiss both dosages of MMF were taken together. Intracellular triphosphate concentrationss before and during HAART were compared using the Wilcoxon two-samplee test.

(8)

Results s

PatientPatient characteristics

Alll patients were males. Baseline characteristics are shown in table 1. After 8 weekss of therapy, the pharmacokinetic profiles of MPA, IDV, ABC, and NVP weree assessed in all patients. After lowering the dose of MMF, the pharmacokineticc profiles were repeated in patient 02, 03 and 04 after 32, 24 andd 24 weeks of therapy, respectively. In patients 01, 02, 03 and 04 intracellularr triphosphates were measured 48, 32, 24 and 24 weeks after startingg therapy. In all other patients this was done after 8 weeks of therapy. Theree were no differences between the patient groups with and without MMF withh respect to adherence or co-medication. There was also no difference in virologicall response between the two groups 23.

Tablee 1 Patient characteristics

Numberr of patients Agee (years) [median (IQR)] Baselinee CD4+

T cells <x108

/l) [median (IQR)] Baselinee CD8+ T cells (x106/l) [median(IQR)] Baselinee plasma HIV-1 RNA (log copies/ml)

[mediann (IQR)] M M F --10 0 344 (28-40) 480(415-610) ) 1130(875-1240)* * 4.700 (4.26-5.79) M M FF + 9 9 355 (32-49) 4300 (320-500) 1410(905-2025) ) 4.63(3.12-5.46) ) P* * 0.50 0 0.26 6 0.14 4 0.55 5 #

Mann-Whitneyy test; 'Baseline CD4+

and CD8* T cells were not available for 1 patient. All patientss were male.

SideSide effects

Inn the group with MMF, one patient (11) discontinued MMF at week 16 at his ownn request. In the group without MMF, one patient (05) stopped ABC in weekk 7 because of a hypersensitivity reaction 24, two patients (07 and 09) switchedd from NVP to efavirenz because of a rash and elevated liver enzymes andd two patients (12 and 17) temporarily stopped therapy, after respectively 199 weeks and 4 days of therapy. These two patients restarted all medication exceptt NVP after 6 weeks (patient 12) and 4 days (patient 17). Overall, no moree side effects were seen in patients using MMF.

(9)

TT cell proliferative capacity

Thee median T cell proliferative capacity measured with [3H]-thymidine incorporationn was 8.0 % (interquartile range (IQR) 4.8 - 22.3) in the group of patientss using MMF compared with 64.0 % (IQR 28.8 - 76.0) in the group of patientss without MMF ( p=0.001, Mann-Whitney test). The T cell proliferative capacityy did not change after lowering the MMF dosage from 1000 mg BID to 5000 mg BID.

PharmacokineticsPharmacokinetics of MP A

Inn patients 02, 03 and 04, who were using MMF 1000 mg twice daily at that time,, the median MPA Cmin concentration was 1.20 ug/ml (IQR 0.44-1.45

ug/ml),, which is well above the minimum required concentration for lymphocytee inhibition 17. The median Cmaxwas 15.20 ug/ml (IQR 11.70-15.50

ug/ml)) and the median AUC was 46.71 ug.h/ml (IQR 25.29-55.34). To prevent sidee effects, we therefore lowered the dose to 500 mg BID after respectively 24,, 16 and 12 weeks of treatment. The pharmacokinetic profile was repeated forr these patients after using the lower dosage for at least 8 weeks. The mediann MPA Cmin concentration measured in all patients using 500 mg BID

(n=9)) was 0.39 ug/ml (IQR 0.37-0.66 ug/ml), which is above the minimum requiredd concentration 17(table 2, figure 1).

PharmacokineticsPharmacokinetics of antiretroviral drugs

Thee AUC and the clearance of ABC were comparable in the groups with and withoutt MMF, p=0.45 and p=0.95 respectively (Mann-Whitney test)(table 2, figuree 1). Likewise, the AUC and the clearance of IDV were not affected by MPA,, p=0.61 and p=0.09 respectively. The AUC of NVP was lower and the clearancee higher in the MMF group: p=0.07 and p=0.04 respectively (Mann-Whitneyy test) (table 2, figure 1).

IntracellularIntracellular triphosphates

Inn the first twelve patients, of which five also received MMF 500 mg BID, intracellularr triphosphates during therapy were measured. Unfortunately, CBVTPP could not be quantified in the cell extracts, due to assay interference.

(10)

Tablee 2 Pharmacokinetic parameters

MMFF 500 mg BID n=9 9 Cmaxx MPA (Mg/ml) [median (IQR)] 6.7 (4.90-12.85) Cmjnn MPA (Mg/ml) [median (IQR)] 0.39 (0.37-0.66)

AUGG MPA (ug.h/ml) [median (IQR)f 19.14 (16.33-33.51) Cl/FF MPA (Uh) [median (IQR)] 26.13 (15.80-30.64)

ABCC n=9 ABC n-10 (MMFF 500 mg BID) (No MMF) C ^^ ABC (Mg/ml) [median (IQR)] 1.98 (1.56-2.84) 1.86 (1.48-2.35) CminABCC (Mg/ml) [median (IQR)] <0.05* < 0.05+

AUCC ABC (ug.h/ml) [median (IQR)f 3.67 (3.10-6.13)) 3.25 (2.39-4.86) Cl/FF ABC (L/h) [median (tQR)] 96.23(78.17-148.17) 110.37(72.41-139.54)

IDVV n=8 IDVN=11 (MMFF 500 mg BID ) (No MMF)

Cmaxx IDV (ug/ml) [median (IQR)] 7.74(4.98-11.87) 9.32 (7.93-10.23) CminIDVV (Mg/ml) [median (IQR)] 0.17 (0.12-0.38) 0.34 (0.20-0.45)

AUCC IDV (|jg.h/ml) [median (IQR)f 33.63 (21.10-41.40) 34.63 (30.13-47.20) Cl/FF IDV (Uh) [median (IQR)] 24.04 (19.33-38.23) 23.11 (16.95-26.56)

NVPn=99 NVP N=8 (MMFF 500 mg BID) (No MMF) Cmaxx NVP (Mg/ml) [median (IQR)] 5.87(5.41-6.78) 7.49 (6.51-8.07) Cminn NVP (Mg/ml) [median (IQR)] 4.25 (3.67-4.74) 5.08 (3.76-5.98) AUCC NVP (|jg.h/ml) [median (IQR)f 61.63 (54.16-69.53) 70.90 (60.11-82.44) Cl/FF NVP (Uh) [median (IQR)] 3.25 (2.88-3.70) 2.83 (2.43-3.34)

"AUCC during a dosing interval (0-12 h); +ln all patients below the LLQ of 0.05 ug/ml; *ln one

patientt 0.06 ug/ml in the other patients below the LLQ of 0.05 ug/ml.

Theirr was no significant difference in dCTP, dGTP or 3TCTP pools or the 3TCTP/dCTPP ratio between the two groups at T=0 (p=0.15, 0.20, 0.88 and 0.20),, T=2 (p=0.54, 0.54, 0.66 and 0.43), T=6 (p=0.43, 0.34, 0.20 and 1.00) andd T=12h (p=0.27, 1.00, 0.64 and 0.27)(Mann-Whitney test)(figure 2). In 6 patientss without MMF, dCTP and dGTP concentrations were also assessed at thee day of starting antiretroviral therapy. There was no difference between

(11)

intracellularr dCTP or dGTP concentrations before and during HAART (p=0.12 andd p=0.46 respectively; Wilcoxon two-sample test). A baseline sample was onlyy available from one patient taking MMF. There was no difference between dCTPP or dGTP concentrations between baseline and week 8 in this patient.

Figuree 1 Pharmacokinetic profiles

r r

- —— MMF 500 mg BIO (n=9) MMFF 1000 mg BID (n=3) Minimumm desired concentrationn (0 14 ug/ml) —— No MMF {n=8) -- - MMF 500 mg BID (n=9) — -- MMF 1000 mg BID (n=3) 99 f s s -- No MMF (n=10) -- MMF 500 (n=9) - -- MMF 1000 (n=3) —— NoMMF(n=11) -- MMF 500 mg BID (n=8) — -- MMF 1000 mg BID (n=3)

Mediann plasma concentrations during a dosing interval; bars represent inter-quartile ranges, a)) concentrations of mycophenolic acid (MPA); (minimum desired concentration of MPA: 0.14 Ug/ml).. b) concentrations of nevirapine (NVP) with or without MMF; c) abacavir (ABC) ; d) indinavirr (IDV).

Discussion n

Thee aim of our study was to investigate the effect of MMF therapy on the pharmacokineticss of a number of antiretroviral drugs, which is important to knoww in case MMF is used in HIV treatment or in case of an organ transplantationn in an HIV patient using antiretroviral drugs. We also wanted to assesss if MPA indeed lowers the intracellular dGTP pool in PBMCs, as in vitro dataa have suggested 12.

(12)

Figuree 2 Intracellular concentrations of dCTP, dGTP, 3TCTP and the ratio 3TCTP/dCT T MMFF -MMFF < baselinee 0 2 Time e 3TCTP P Ü Ü s s 1 1 s s « « b£ £ seline e -*-"*" " 0 0 „.. — * * 2 2 » f f 6 6 . . ** + I 2 h h jurs s M M F --MMFF + ff i. 7.5-- 5.0-- 2.5--00 0-b s s — seline e dGTP P MMF--M MMF--M F H H 3TCTP/dCTP P MMF--MMFF *

Horizontall line represents median triphosphate concentration or median ratio at baseline and duringg a twelve hour dosing interval after at least 8 weeks of therapy.

Thee first 3 patients (patient 02, 03 and 04) using MMF started with 1000 mg BID.. Because these first three patients had high MPA plasma concentration andd because the lymphocyte proliferation was completely blocked, the dose wass lowered to 500 mg BID in these and all new patients to prevent side effects.. It has been demonstrated that with a dose of 500 mg BID the inhibitionn of the lymphocyte proliferation is only slightly lower compared with a 10000 mg BID regimen 25, and when the lymphocyte proliferation is fully inhibitedd a further increase of the MMF dosage does not have any beneficial effectt 17. MPA has antiretroviral activity at lower concentrations of MPA than necessaryy for inhibition of lymphocyte proliferation 815. After dose reduction thee lymphocyte proliferation remained completely blocked.

Theree were no differences in the pharmacokinetics of ABC and IDV between thee 2 groups. In the group using MMF the NVP clearance was slightly but significantlyy higher than in the group without MMF. The primary routes of NVP biotransformationn and elimination include CYP metabolism by CYP3A4 and

(13)

CYP2B66 isoenzymes, glucuronide conjugation and urinary excretion of the glucuronidee metabolite 26 . Following oral administration MMF is rapidly and completelyy absorbed and de-esterified to MPA. MPA is almost completely glucuronidatedd to its stable phenolic glucuronide metabolite (MPAG) which is excretedd in urine 27. Both NVP28 and MPA27 undergo enterohepatic recycling. Itt is possible that there is interference of MPA with the enterohepatic recycling off NVP. This could be due to competition for transporters necessary for excretionn in bile. Another possibility might be that MPA has a local immunosuppressivee effect in the gut resulting in a change in normal gut flora, whichh could result in a change of the enterohepatic recycling of NVP. If indeed NVPP concentrations are lowered because of alterations of the enterohepatic recycling,, this could mean that the use of antibiotics could also influence NVP concentrationss by affecting the normal gut flora necessary for the enterohepaticc recycling. Because MPA undergoes enterohepatic recycling a secondd peak in plasma MPA concentration would have been expected. The absencee of this peak in our patients might be caused by interference of the antiretrovirall drugs with the excretion of MPA into bile. This effect on MPA has beenn reported for cyclosporine A 29.

Inn the first 12 patients intracellular dCTP, dGTP and 3TCTP pools were measured.. In 6 of these patients, who were all without MMF, samples before thee start of treatment were available. Both dCTP and dGTP concentrations did nott change during treatment, which can be explained by the fact that both dCTPP and dGTP concentrations are not influenced by 3TC and ABC 30. We hadd a sample before treatment of only one patient using MMF. During therapyy the intracellular concentrations of dCTP did not differ between patientss with and without MMF, as was expected because the dCTP synthesis iss not directly affected by MPA. As a consequence the ratio 3TCTP/dCTP was comparablee in both groups. The relative high concentration of 3TCTP leads to aa preference for 3TCTP incorporation during HIV replication, thereby inhibiting replication.. Because MPA inhibits dGTP production, we expected a lower dGTPP concentration in patients treated with MMF. Although lymphocyte proliferationn was completely blocked in patients using MMF, dGTP pools were

(14)

nott significantly different in patients with or without MMF. This is substantially inn accordance with a recent small study, in which a consistent decrease of dGTPP was not demonstrated in each of the 5 patients receiving MMF 31. Also inn patients using hydroxyurea in combination with antiretroviral therapy, the expectedd decrease of endogenous triphosphates, particularly dATP, predicted fromm in vitro studies 32 was not seen in vivo 33. One possible explanation could bee the variability of triphosphorylation in vivo. It is also possible that proliferationn is more sensitive than dGTP measurement. Another explanation mightt be that indeed dGTP is lowered by MPA after a first dosing, but that cellss in vivo develop a compensation mechanism probably by the salvage pathway.. However this would not explain the decreased lymphocyte activity thatt was observed throughout the study. CBVTP and dGTP have similar affinityy for HIV reverse transcriptase 34, so it is unlikely that these findings are causedd by an accumulation of dGTP in the presence of CBVTP. Assessment off dGTP pools shortly after the start of MMF, as compared to prolonged use of MMF,, might elucidate this issue further.

Althoughh this was a small study, we demonstrated that MPA alters the pharmacokineticss of NVP but has no effect on ABC and IDV. Furthermore, despitee effectively blocking lymphocyte proliferation, MPA did not decrease intracellularr dGTP pools at the time points tested in this study. Clearly, these resultss warrant further investigations on the supposed working mechanism of MPA. .

Acknowledgement t

WeWe thank Marijke Roos, Sanquin Research and Landsteiner Laboratory, Universityy of Amsterdam, Amsterdam, the Netherlands for measuring lymphocytee proliferation. This study was financially supported by a private foundationn that wishes not to be named.

(15)

References s

1.. van Praag RM, Wit FW, Jurriaans S et al. Improved long-term suppression of HIV-11 replication with a triple-class multidrug regimen compared with standardd of care antiretroviral therapy. AIDS. 2002;16:719-725.

2.. Weveriing GJ, Lange JM, Jurriaans S et al. Alternative multidrug regimen providess improved suppression of HIV-1 replication over triple therapy. AIDS. 1998;12:F117-F122. .

3.. Zhang L, Ramratnam B, Tenner-Racz K et al. Quantifying residual HIV-1 replicationn in patients receiving combination antiretroviral therapy. N Engl J

Med.Med. 1999;340:1605-1613.

4.. Zack JA, Arrigo SJ, Weitsman SR et al. HIV-1 entry into quiescent primary lymphocytes:: molecular analysis reveals a labile, latent viral structure. Cell. 1990;61:213-222. .

5.. De Boer RJ, Boucher CA, Perelson AS. Target cell availability and the successfull suppression of HIV by hydroxyurea and didanosine. AIDS. 1998;12:1567-1570. .

6.. Lori F, Malykh A, Cara A et al. Hydroxyurea as an inhibitor of human immunodeficiencyy virus-type 1 replication. Science. 1994;266:801-805. 7.. Havlir DV, Gilbert PB, Bennett K et al. Effects of treatment intensification with

hydroxyureahydroxyurea in HIV-infected patients with virologie suppression. AIDS. 2001;15:1379-1388. .

8.. Margolis D, Heredia A, Gaywee J et al. Abacavir and mycophenolic acid, an inhibitorr of inosine monophosphate dehydrogenase, have profound and synergisticc anti-HIV activity. J Acquir Immune Defic Syndr. 1999;21:362-370. 9.. Chapuis AG, Paolo RG, D'Agostino C et al. Effects of mycophenolic acid on

humann immunodeficiency virus infection in vitro and in vivo. Nat Med. 2000;6:762-768. .

10.. Becker BN. Mycophenolate mofetil. Transplant Proc. 1999;31:2777-2778. 11.. Allison AC, Eugui EM. Mycophenolate mofetil and its mechanisms of action.

Immunopharmacology.Immunopharmacology. 2000;47:85-118.

12.. Allison AC, Eugui EM. Immunosuppressive and other effects of mycophenolic acidd and an ester prodrug, mycophenolate mofetil. Immunol Rev. 1993; 136:5-28. .

13.. Faletto MB, Miller WH, Garvey EP et al. Unique intracellular activation of the potentt anti-human immunodeficiency virus agent 1592U89. Antimicrob

(16)

14.. Stein DS, Moore KH. Phosphorylation of nucleoside analog antiretrovirals: a revieww for clinicians. Pharmacotherapy. 2001;21:11-34.

15.. Hossain MM, Coull JJ, Drusano GL et al. Dose proportional inhibition of HIV-1 replicationn by mycophenolic acid and synergistic inhibition in combination with abacavir,, didanosine, and tenofovir. Antiviral Res. 2002;55:41-52.

16.. Halpem SD, Ubel PA, Caplan AL. Solid-organ transplantation in HIV-infected patients.. NEnglJMed. 2002;347:284-287.

17.. Gummert JF, Barten MJ, Sherwood SW et al. Pharmacodynamics of immunosuppressionn by mycophenolic acid: inhibition of both lymphocyte proliferationn and activation correlates with pharmacokinetics. J Pharmacol

ExpExp Ther. 1999;291:1100-1112.

18.. Roos MT, Prins M, Koot M et al. Low T-cell responses to CD3 plus CD28 monoclonall antibodies are predictive of development of AIDS. AIDS. 1998;12:1745-1751. .

19.. Sparidans RW, Hoetelmans RM, Beijnen JH. Liquid chromatographic assay forr simultaneous determination of abacavir and mycophenolic acid in human plasmaa using dual spectrophotometric detection. J Chromatogr B Biomed Sci

Appl.Appl. 2001;750:155-161.

20.. Crommentuyn KM, Rosing H, Nan-Offeringa LG et al. Rapid quantification of HIVV protease inhibitors in human plasma by high-performance liquid chromatographyy coupled with electrospray ionization tandem mass spectrometry.. J Mass Spectrom. 2003;38:157-166.

21.. van Heeswijk RP, Hoetelmans RM, Meenhorst PL et al. Rapid determination off nevirapine in human plasma by ion-pair reversed-phase high-performance liquidd chromatography with ultraviolet detection. J Chromatogr B Biomed Sci

Appl.Appl. 1998;713:395-399.

22.. Kewn S, Hoggard PG, Sales SD et al. Development of enzymatic assays for quantificationn of intracellular lamivudine and carbovir triphosphate levels in peripherall blood mononuclear cells from human immunodeficiency virus-infectedd patients. Antimicrob Agents Chemother. 2002;46:135-143.

23.. Sankatsing SUC, Jurriaans S, Schuitemaker H et al. Mycophenolate Mofetil doess not have an additional effect on plasma HIV-RNA decay and on the pool off latently infected resting CD4* T lymphocytes in therapy naive HIV-1 patientss starting HAART. 10th Conference on Retroviruses and Opportunistic Infections,, Boston, USA, February 2003, Abstract 658a.

(17)

24.. Sankatsing SU, Prins JM. Agranulocytosis and fever seven weeks after startingg abacavir. AIDS. 2001;15:2464-2465.

25.. Brunet M, Martorell J, Oppenheimer F et al. Pharmacokinetics and pharmacodynamicss of mycophenolic acid in stable renal transplant recipients treatedd with low doses of mycophenolate mofetil. Transpl Int. 2000; 13 Suppl 1:S301-S305. .

26.. Boehringer Ingelheim Pharmaceuticals I. Product Information Viramune®,nevirapine.. 1999. Ridgefield U.S.A.

27.. Bullingham RE, Nicholls AJ, Kamm BR. Clinical pharmacokinetics of mycophenolatee mofetil. Clin Pharmacokinet. 1998;34:429-455.

28.. Cheeseman SH, Hattox SE, McLaughlin MM et al. Pharmacokinetics of nevirapine:: initial single-rising-dose study in humans. Antimicrob Agents

Chemother.Chemother. 1993;37:178-182.

29.. van Gelder T, Klupp J, Barten MJ et al. Comparison of the effects of tacrolimuss and cyclosporine on the pharmacokinetics of mycophenolic acid.

TherTher Drug Monit. 2001 ;23:119-128.

30.. Daluge SM, Good SS, Faletto MB et al. 1592U89, a novel carbocyclic nucleosidee analog with potent, selective anti-human immunodeficiency virus activity.. Antimicrob Agents Chemother. 1997;41:1082-1093.

31.. Margolis DM, Kewn S, Coull JJ et al. The addition of mycophenolate mofetil to antiretrovirall therapy including abacavir is associated with depletion of intracellularr deoxyguanosine triphosphate and a decrease in plasma HIV-1 RNA.. J Acquir Immune Defic Syndr. 2002;31:45-49.

32.. Gao WY, Johns DG, Chokekuchai S et al. Disparate actions of hydroxyurea in potentiationn of purine and pyrimidine 2',3'-dideoxynucleoside activities against replicationn of human immunodeficiency virus. Proc Natl Acad Sci USA. 1995;92:8333-8337. .

33.. Hoggard PG, Kewn S, Maherbe A et al. Time-dependent changes in HIV nucleosidee analogue phosphorylation and the effect of hydroxyurea. AIDS. 2002;16:2439-2446. .

34.. Miller WH, Daluge SM, Garvey EP et al. Phosphorylation of carbovir enantiomerss by cellular enzymes determines the stereoselectivity of antiviral activity.. J Biol Chem. 1992;267:21220-21224.

Referenties

GERELATEERDE DOCUMENTEN

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

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

The analysis in the 2002 KPMG Survey of Corporate Responsibility Reporting (2002, p.18) suggests that the increased adoption of sustainability assurance arises from “…the demand

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

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

UvA-DARE is a service provided by the library of the University of Amsterdam (http s ://dare.uva.nl) UvA-DARE (Digital Academic Repository).. Cultuur, koningen

DeDe Koninklijke Bibliotheek 53 HetHet Koninklijk Museum 54 HetHet Koninklijk Instituut 55 HetHet Algemeen Rijksarchief 55 GeenGeen academie en geen paleis 56.