Josephson 2010

You might also like

You are on page 1of 10

Review | doi: 10.1111/j.1365-2796.2010.02301.

Drug–drug interactions in the treatment of HIV infection: focus


on pharmacokinetic enhancement through CYP3A inhibition
F. Josephson
Medical Products Agency, Uppsala, Sweden

Abstract. Josephson F. (Medical Products Agency, Upp- vir boosting’ – will be reviewed. The emphasis here will
sala, Sweden). Drug–drug interactions in the treat- be on the treatment of important co-morbidities com-
ment of HIV infection: focus on pharmacokinetic mon in patients with HIV, including dyslipidaemia,
enhancement through CYP3A inhibition (Review- hypertension, tuberculosis and opiate dependence,
Symposium). J Intern Med 2010; 268: 530–539. as well as on the potentially life-threatening interac-
tion between ritonavir and inhalational steroids, and
The aim of this review is to discuss the effect of phar- on the effect of acid-reducing agents on some antiret-
macokinetic drug–drug interactions (DDIs) in the an- roviral drugs. Finally, further developments with re-
tiretroviral treatment of HIV infection. In particular, gard to the use of CYP3A-blocking agents to augment
but not exclusively, DDIs due to the cytochrome P450 the efficacy of antiviral therapy will be discussed.
3A (CYP3A) inhibitor ritonavir, which is used to in-
crease antiretroviral drug exposure – a technique Keywords: cobicistat, CYP3A, drug interaction, HIV,
known as pharmacokinetic enhancement or ‘ritona- ritonavir.

inactive metabolites [4–6]. To a varying extent, they


Introduction
are themselves inhibitors of CYP3A, and some are
Given our understanding of the disease and the avail- also inducers of hepatic drug-metabolizing enzymes.
able therapies, infection with human immunodefi- Furthermore, and of crucial importance, all first-line
ciency virus (HIV) currently requires lifelong therapy. recommended PI regimens include co-administra-
Combinations of three active drugs from at least two tion of the CYP3A inhibitor ritonavir to increase the
classes are recommended in most cases. The most pharmacokinetic exposure to the active PI through
commonly used combination regimens include two increased bioavailability and decreased clearance
nucleoside analogue inhibitors of reverse transcrip- (lopinavir is co-formulated with ritonavir). This prac-
tase (NRTIs) in combination with either a non-nucleo- tice of pharmacokinetic enhancement is termed ‘ri-
side inhibitor of reverse transcriptase (NNRTI) or an tonavir boosting’ and has been shown to increase the
HIV protease inhibitor (PI) (Table 1). The potential for potency and greatly augment the barrier to resistance
clinically relevant drug–drug interactions (DDIs) with of PI-based antiretroviral therapy [7].
the NRTIs is limited and mainly restricted to interac-
tions between the NRTIs themselves; though, there CYP3A, however, is not only responsible for the
are some exceptions to this rule. With regard to the metabolism of PIs, but is also the predominant en-
NNRTIs, all three drugs currently approved in the zyme for drug metabolism in general. Its substrates
European Union (efavirenz, nevirapine and etravi- include many important drugs used in a wide range
rine) are inducers of cytochrome P450 (CYP450) en- of therapeutic fields. Thus, ritonavir boosting creates
zymes and may in some cases cause clinically rele- a plethora of putative, clinically relevant DDIs, as
vant decreases in exposure to co-treating agents that illustrated in Table 2.
undergo CYP450-dependent metabolism [1–3]. Addi-
tionally, they are metabolized by several different In addition to NRTIs, NNRTIs and PIs, other currently
CYP450 enzymes, and co-treatment with potent he- available drugs include the entry inhibitors enfuvir-
patic enzyme inducers, such as rifampicin, may in tide and maraviroc, and the integrase inhibitor ralte-
turn cause lowered NNRTI exposure. The generally gravir. The former drugs have relatively limited use in
recommended PIs, including atazanavir, lopinavir Europe at present. Raltegravir, however, is a very po-
and darunavir, are almost exclusively metabolized by tent drug with a wide therapeutic index and is
CYP450 3A (CYP3A) and thereby biotransformed into increasingly used not only in treatment-experienced

530 ª 2010 The Association for the Publication of the Journal of Internal Medicine
F. Josephson
| Review-Symposium: Drug–drug interactions in the treatment of HIV infection

Table 1 Examples of recommended and alternative first-line antiretroviral treatment regimens (e.g. see European AIDS Clinical
Society guidelines, available at http://www.europeanaidsclinicalsociety.org/guidelines.asp)

NNRTI-based regimens Major pharmacokinetic DDI potential


NRTI NRTI NNRTI
abacavir + lamivudine + efavirenz Reduced exposure to co-treating agents because of induction of
tenofovir + emtricitabine + efavirenz drug-metabolizing enzymes. Reduced exposure to NNRTI because of
enzyme induction by co-treating agent.
PI-based regimens
NRTI NRTI PI ⁄ ritonavir (r)
abacavir + lamivudine + atazanavir ⁄ r Increased exposure to co-treating agents because of CYP3A inhibition.
abacavir + lamivudine + darunavir ⁄ r Reduced exposure to co-treating agents because of induction of
tenofovir + emtricitabine + atazanavir ⁄ r drug-metabolizing enzymes.
tenofovir + emtricitabine + darunavir ⁄ r
Integrase inhibitor (II)-based regimens
NRTI NRTI II
tenofovir + emtricitabine + raltegravir

but also in treatment-naive patients. Of interest, sig- The DDI potential of ritonavir-boosted PI drug regi-
nificant DDIs are not commonly associated with ral- mens is complicated by the fact that ritonavir is not
tegravir, and it may be a valuable option when DDIs only a strong net inhibitor of CYP3A but is also a po-
are complicating the choice of antiretroviral agents. tent pregnane X receptor activator. It is therefore an
inducer of a number of clinically important drug-
metabolizing enzymes, including CYP1A2, CYP2C9,
Ritonavir
CYP2C19, CYP3A and glucuronyl transferases [8,
Ritonavir is itself a PI and was one of the first drugs 11]. Furthermore, it is an inducer as well as an inhibi-
in this class to be approved for the treatment of HIV. tor of transmembrane drug transporter p-glycopro-
At therapeutic doses (400–600 mg twice daily), it is, tein [16, 17]. At therapeutic but not boosting doses, it
however, arguably the least well-tolerated PI, with is also a significant inhibitor of CYP2D6 [8]. Owing to
side effects including severe gastrointestinal symp- the capacity of ritonavir (as well as some of the thera-
toms, as well as increased plasma levels of choles- peutic PIs) to induce hepatic enzymes, the spectrum
terol and triglycerides [8, 9]. As mentioned previ- of clinically relevant potential DDIs with ritonavir-
ously, ritonavir is also a very strong inhibitor of boosted PI regimens is not restricted to the risk of
CYP3A. At considerably more tolerable, though by no substantially increased drug exposure and toxicity
means side effect–free, doses of 100 mg once or twice because of CYP3A inhibition, but also in some cases
daily, which are used for ritonavir boosting, hepatic includes the possibility of decreased exposure and
CYP3A activity is blocked by >80% and presystemic therapeutic efficacy because of the induction of other
CYP3A even more so, using midazolam or alfentanil hepatic enzymes.
as a probe [10, 11]. Near-maximal CYP3A blockade
is seen at doses as low as 50 mg [12]. Ritonavir
PI and statin therapy
boosting increases the area under the concentra-
tion–time curve (AUC) 5- to 80-fold, depending on Co-treatment with lipid-lowering therapy is common
which ‘therapeutic’ PI is boosted. The highest values amongst patients treated for HIV infection. Dyslipida-
are seen for substances such as darunavir and lopin- emia is frequent in the general population and may be
avir. The bioavailability of these agents in the ab- more so amongst individuals with HIV, regardless of
sence of ritonavir is very low as a result of presystem- treatment [18]. Also, several antiretroviral agents are
ic metabolism, and therapeutic exposure cannot be associated with alterations in plasma lipids. Of
reached in the absence of profound CYP3A blockade importance, most PIs, including ritonavir at boosting
[4, 5, 13–15]. doses, may cause increased total cholesterol,

ª 2010 The Association for the Publication of the Journal of Internal Medicine Journal of Internal Medicine 268; 530–539 531
F. Josephson
| Review-Symposium: Drug–drug interactions in the treatment of HIV infection

Table 2 Examples of drugs that undergo CYP3A-dependent Table 2 (Continued)


metabolism, which are contraindicated or should be used
Drugs undergoing
with caution when co-treating with a ritonavir-boosted PI (list
based on the European SPC for Norvir [8], but note that the CYP3A-dependent
list is not exhaustive) metabolism Comments
Amlodipine
Drugs undergoing
Felodipine
CYP3A-dependent
Nifedipine
metabolism Comments
Diltiazem
Alpha-receptor Details of the metabolism of other
Verapamil
blockers drugs in this class, such as
Gastrointestinal See above, anti-arrhythmics
terazosin and doxazocin, have
motility agents
not been reported; they may also
Cisapride
be CYP3A substrates
Statins See text. Appropriate statin depends
Alfuzosin
on which boosted PI is used
Opiates CYP3A is not involved in morphine
Simvastatin
metabolism
Lovastatin
Pethidine
Atorvastatin
Fentanyl
Immunosuppressants Considerable increase in
Anti-arrhythmics The risks of co-administration of
immunosuppressant exposure.
QT-prolonging agents that are
Co-treatment is possible, however,
CYP3A substrates, together
with careful titration and use of
with strong CYP3A inhibitors such
therapeutic drug monitoring
as ritonavir, may be quite considerable
Cyclosporin
Amiodarone
Tacrolimus
Bepridil
Everolimus
Quinidine
PDE-5 inhibitors Co-treat with caution, using
Anticancer Note that a number of other anticancer
reduced doses of PDE5 inhibitors
agents agents are also CYP3A substrates
Sildenafil
Vincristine
Tadalafil
Vinblastine
Vardenafil
Anticonvulsants CYP3A is not primarily involved in
Benzodiazepines In contrast to the listed benzodiaze-
the metabolism of valproic acid
pines, oxazepam does not undergo
and lamotrigine
CYP3A-dependent metabolism
Carbamazepine
Diazepam
Antihistamines Cetirizine, which has considerable
Flurazepam
renal elimination, may be a
Midazolam
preferred alternative
Triazolam
Astemizole
Flunitrazepam
Terfenadine
Inhaled Reported that beclometasone does
Calcium channel See text. The magnitude of
corticosteroids not undergo CYP3A-dependent
antagonists increased exposure probably
metabolism
varies between agents. The risks of
Fluticasone
co-administration with ritonavir may
Mometasone
be higher with agents that affect
Budesonide
cardiac conduction at therapeutic doses

532 ª 2010 The Association for the Publication of the Journal of Internal Medicine Journal of Internal Medicine 268; 530–539
F. Josephson
| Review-Symposium: Drug–drug interactions in the treatment of HIV infection

decreased HDL cholesterol and increased triglyceride but an effect of darunavir on the transmembrane
levels [19]. transport of pravastatin has been suggested [22]. An-
other unexpected finding in the complex field of PI–
The liability to interact with ritonavir-boosted PIs var- statin DDIs was that ritonavir-boosted lopinavir dou-
ies between statins, and interestingly, also between bled the exposure to rosuvastatin, and this is not
PIs. With regard to elimination pathways, simvasta- readily explained on the basis of inhibition of metabo-
tin, lovastatin and atorvastatin are metabolized by lism [4]. However, it is considered that all statins are
CYP3A. Thus, one would expect clinically relevant substrates of the OATP1B1 transmembrane trans-
DDIs between these substances and ritonavir- porter, which impacts their intracellular availability
boosted PI regimens. Indeed, when ritonavir and sa- [21], and lopinavir was recently shown to inhibit this
quinavir were co-administered with simvastatin, the transporter at clinically relevant concentrations [23].
exposure to simvastatin acid increased 30-fold [20]. In summary, extrapolation of DDI data between sta-
Thus, the combination of simvastatin and PIs is con- tins and PIs has turned out to be more difficult than
traindicated because of the risk of rhabdomyolysis, initially expected, as multiple mechanisms other
as is that of lovastatin and PIs by argument of anal- than CYP3A inhibition are involved, and these appear
ogy. Exposure to atorvastatin when co-administered to differ between drugs in the class.
with a ritonavir-boosted PI only increases by three to
sixfold, and exposure to the active moiety even less so
PI and calcium channel antagonists
[4, 5, 20]. Although the use of atorvastatin together
with ritonavir-boosted PIs is not recommended in all There are numerous drugs in this important class,
cases, titration from a low starting dose under careful which are used for several common conditions,
monitoring is considered an acceptable strategy in including hypertension, angina pectoris and cardiac
cases of co-administration with, for example, ritona- arrhythmias. Most, if not all, calcium channel antag-
vir-boosted lopinavir or darunavir. The difference be- onists are substrates for CYP3A; some are also sub-
tween the magnitudes of the increased exposure be- strates for p-glycoprotein [24]. Thus, increases in cal-
tween simvastatin and atorvastatin may result from cium channel antagonist exposure may be expected
the fact that the former is administered as a lactone when co-treating with ritonavir. However, the phar-
prodrug and has a lower bioavailability because of macokinetic impact of ritonavir co-administration
presystemic CYP3A-mediated metabolism in the ab- appears to have been studied for relatively few cal-
sence of a strong CYP3A inhibitor [20, 21]. This illus- cium channel blockers. Available data support the
trates an important principle regarding DDIs due to theoretical assumption of an interaction potential;
ritonavir – that the greatest-fold increase in exposure though, the magnitude and clinical relevance of the
may be seen for agents that normally have restricted effect may vary between agents, e.g. because of differ-
bioavailability owing to presystemic metabolism. ing degrees of presystemic CYP3A-dependent metab-
This principle will be further illustrated below, with olism and ⁄ or the presence of alternative metabolic
regard to the interaction between inhalational corti- pathways. Co-administration of diltiazem or amlodi-
costeroids and ritonavir. pine together with indinavir in combination with ri-
tonavir at a dose of 800 ⁄ 100 mg twice daily led to an
Fluvastatin is metabolized by CYP2C9, whereas pra- increase in amlodipine exposure by a median of
vastatin and rosuvastatin do not undergo significant approximately 90%, whereas the increase in dil-
CYP450-dependent metabolism [21]. Fluvastatin tiazem exposure was more moderate [24]. In general,
and pravastatin are considered first-line possibilities if co-administered with ritonavir, dose titration of the
when a statin is needed in patients receiving ritona- calcium channel antagonist and careful monitoring
vir-boosted PI therapy. However, pravastatin expo- of the response seem prudent. A useful strategy may
sure was decreased by 50% when co-administered be to chose alternative agents to calcium channel
with saquinavir and ritonavir, presumably because blockers with effects on cardiac conduction at thera-
of induction of metabolism [20], and recommended peutic doses, if possible.
doses may often not be sufficiently efficacious. Also,
contrary to expectation, when the interaction be-
Co-treatment with ritonavir and inhaled corticosteroids
tween pravastatin and ritonavir-boosted darunavir
was studied, an 81% increase in pravastatin expo- In the late 1990s, two case reports of patients treated
sure was seen, and up to fivefold increases were noted with ritonavir developing Cushing’s syndrome, pre-
in a subset of subjects [5]. To our knowledge, the sumably because of an interaction with nasally in-
mechanism of this interaction has not been clarified, haled fluticasone, were published [25, 26]. To date,

ª 2010 The Association for the Publication of the Journal of Internal Medicine Journal of Internal Medicine 268; 530–539 533
F. Josephson
| Review-Symposium: Drug–drug interactions in the treatment of HIV infection

more than 30 cases of Cushing’s syndrome and ⁄ or omeprazole reduces the AUC of ritonavir-boosted at-
secondary adrenal insufficiency because of bronchi- azanavir by 75% [33]. Even when the dose of ritona-
ally or nasally administered fluticasone in combina- vir-boosted atazanavir is increased from the standard
tion with ritonavir-boosted PI therapy have been re- 300 mg once daily to 400 mg, a 20-mg dose of omep-
ported. Some difficulties in diagnosis are noted in razole reduces the AUC of atazanavir by about 30%,
these case reports. According to one, the use of nasal and a 40-mg dose by about 60%, relative to the stan-
fluticasone was not reported by the patient as he did dard dose under normal conditions. Consequently,
not consider this over-the-counter drug to be ‘a medi- concomitant use of atazanavir and proton pump
cine’ [26]. In other reports, the differential diagnosis inhibitors is considered contraindicated, at least in
of antiretroviral therapy–associated lipodystrophy treatment-experienced patients who might require
was initially considered [27, 28]. higher exposure to atazanavir [6, 33]. Pharmacoki-
netic interactions with other acid-reducing agents
Like several other drugs in its class, such as budeso- are also significant and may require temporal separa-
nide and mometasone, the inhalational steroid fluti- tion of doses [6, 33]. By contrast, exposure to the in-
casone is a CYP3A substrate [29]. Under normal cir- tegrase inhibitor raltegravir is increased more than
cumstances, systemic exposure to fluticasone is twofold by proton pump inhibitors [32]. However, at
limited by a very high presystemic and systemic present, this is not thought to be clinically relevant.
CYP3A-mediated clearance. However, in the presence
of ritonavir 100 mg twice daily, systemic exposure to
Antiretroviral therapy and opiate substitution
nasally administered fluticasone increases more than
350-fold, with concomitant suppression of endoge- As intravenous drug use is a major transmission
nous cortisol synthesis [8]. Although similar effects route for HIV, many patients treated with antiretrovi-
might be expected from other drugs in this class, as ral therapy receive concomitant opiate substitution
mentioned previously, quantitative data are lacking with methadone or buprenorphine. Methadone is a
concerning the interaction between ritonavir and long-acting l-opioid receptor agonist. On the basis of
other agents. It is notable that the great majority of in vitro studies, the elimination of methadone was
published cases have involved fluticasone, and only previously at least partly attributed to CYP3A. How-
recently was a case of adrenal suppression and Cush- ever, as a recent study showed no relation between
ing’s syndrome presumed because of ritonavir and bu- CYP3A activity when inhibited by ritonavir and the
desonide reported [30]. It has been speculated that the impact on methadone pharmacokinetic parameters,
higher potency and lipophilicity, with consequent po- this has been questioned [34]. In addition to metabo-
tential for accumulation, might make the interaction lism, presumably involving several different drug-
between fluticasone and ritonavir more significant metabolizing enzymes, methadone is also cleared by
than between budesonide and ritonavir [29]. Also, if renal elimination [34]. Buprenorphine, which is
the systemic bioavailability of fluticasone under nor- administered sublingually because of extensive first-
mal circumstances is more restricted by CYP3A-medi- pass metabolism, is a partial agonist of the l-opioid
ated metabolism, the quantitative effect of ritonavir receptor. It is converted to an active metabolite, nor-
may be higher on fluticasone than on budesonide. Be- buprenorphine, via CYP3A and CYP2C8, and further
cause of this uncertainty concerning the DDI potential metabolized through glucuronidation [35]. Thus,
of other inhalational steroids that are metabolized by both of these drugs have the potential to interact with
CYP3A, beclometasone, which is not considered a PIs and NNRTIs.
substrate for this enzyme, is recommended when pa-
tients treated with a ritonavir-boosted PI are in need of Amongst the NNRTIs, efavirenz, which is an inducer
therapy with an inhalational steroid [8, 29]. of hepatic drug-metabolizing enzymes, approxi-
mately halved exposure to methadone and precipi-
tated withdrawal symptoms [1]. Clinically, the evalu-
Antiretrovirals and acid-reducing agents
ation of such symptoms may be complicated by the
The bioavailability of some antiretrovirals is affected fact that efavirenz, particularly during the first weeks
by gastric pH. Consequently, various classes of acid- of therapy, may cause CNS symptoms that can be dif-
reducing agents have been reported to affect the ficult to distinguish from symptoms of opiate with-
pharmacokinetics of several different antiretrovirals, drawal. Although the interaction between efavirenz
with increased or decreased exposure as a conse- and methadone may be managed by methadone dose
quence [6, 31, 32]. The bioavailability of atazanavir adjustments, many clinicians avoid the use of efavi-
decreases with increasing gastric pH. A 40-mg dose of renz in such situations. The effects of nevirapine on

534 ª 2010 The Association for the Publication of the Journal of Internal Medicine Journal of Internal Medicine 268; 530–539
F. Josephson
| Review-Symposium: Drug–drug interactions in the treatment of HIV infection

methadone are similar to those of efavirenz, whereas concentration during the dosing interval, prior to the
the effects of etravirine are negligible [2, 3]. When ef- next scheduled dose, and assumed to be the most
avirenz was co-administered with buprenorphine, important pharmacokinetic parameter for these anti-
the magnitude of the interaction in terms of pharma- retrovirals) are reduced by 26% and 32%, respec-
cokinetic effect was similar to that with methadone; tively, for efavirenz, and by 58% and 68%, respec-
buprenorphine AUC was decreased by 52% and nor- tively, for nevirapine [1, 2]. This has led to
buprenorphine AUC even more so. However, no pa- considerable controversy concerning the need for
tients displayed withdrawal symptoms [1]. Thus, the dose adjustments of these agents when co-treating
pharmacokinetic ⁄ pharmacodynamic relationship of with rifampicin. This issue has been complicated by
buprenorphine, a partial agonist, may be different the fact that efavirenz and to some extent nevirapine
from that of methadone in the clinically relevant are substrates not only for CYP3A but also for the
concentration intervals. Data on the interactions genetically polymorphic CYP2B6 enzyme [1, 41].
between the other NNRTIs that are licensed in the Thus, exposure to efavirenz varies greatly in a popu-
European Union and buprenorphine appear to be lation, with patients who are homozygous for inacti-
lacking. vating mutations in CYP2B6 exhibiting several-fold
higher exposures at the recommended dose and hav-
Ritonavir at a boosting dose (100 mg twice daily) did ing an increased risk of exposure-dependent central
not significantly affect methadone exposure [36], nervous system side effects [42]. The frequency of
whereas ritonavir at a therapeutic dose (500 mg twice inactive CYP2B6 alleles may be as high as 50% in Afri-
daily) decreased methadone exposure by 36% [8], can populations [43, 44], and increasing the dose in
illustrating the dose dependence of an interaction such patients may increase side effects [45]. Whereas
presumably due to hepatic enzyme induction. In an increased dose of efavirenz when co-treating with
addition, the net effect of different boosted PI regi- rifampicin is suggested (though not actually recom-
mens may depend on the potency of the individual PIs mended) according to European labelling [1], there is
as enzyme inducers. Thus, lopinavir in combination emerging evidence to suggest that this is not generally
with ritonavir at the boosting dose decreased metha- necessary to maintain antiretroviral efficacy [46–48].
done AUC by an average of 28%, which was associ- With nevirapine, evidence for maintained efficacy
ated with symptoms of opiate withdrawal [36]. with labelled doses when co-treating with rifampicin
is less strong [49, 50]; however, an increased dose of
nevirapine may be associated with an increased risk
DDIs and the co-treatment of HIV and tuberculosis
of hypersensitivity reactions, including rash and hep-
At least one-third of the people living with HIV are also atitis [49]. Of note, rash is an important side effect of
infected with Mycobacterium tuberculosis and are 20– isoniazid, and both isoniazid and pyrazinamide may
30 times more likely to develop clinical tuberculosis cause drug-induced hepatitis, complicating the dif-
(TB) than people without HIV [37]. In many cases, TB ferential diagnosis in case of such symptoms. There-
and HIV are diagnosed at the same time. In such fore, an efavirenz-based antiretroviral regimen has
cases, initiating TB treatment is the first priority, but emerged as a favoured option in cases of concomitant
an increasing body of evidence indicates that delay- TB.
ing HIV treatment until after completion of TB ther-
apy (usually 6 months) increases mortality, and par- In contrast to ritonavir-boosted PIs, efavirenz and ne-
ticularly so in patients with substantial immune virapine are both drugs with low barriers to resis-
deficiency [38]. Standard TB regimens include rifam- tance. This means that following virological failure on
picin, isoniazid and pyrazinamide with ⁄ without eth- a first-line regimen based on either of these agents,
ambutol [39]. The use of rifampicin, a crucial agent in full resistance will often have developed. Thus, partic-
the TB regimen, may pose formidable problems with ularly in the geographical regions where HIV–TB co-
regard to DDIs when co-treating HIV and TB. Rifam- infection is common, an effective second-line antiret-
picin may be the most potent inducer of hepatic drug- roviral regimen will require a ritonavir-boosted PI. As
metabolizing enzymes of all therapeutic agents in mentioned earlier, PI exposure is decreased by rifam-
current use [40]. As rifampicin induces CYP3A, it re- picin, and for pharmacokinetic reasons, unboosted
duces exposure to both PIs and NNRTIs. For example, PIs are not considered to be options for co-treatment
the NNRTIs efavirenz and nevirapine are the first-line [40]. Boosted PI regimens in combination with rifam-
antiretroviral drugs predominantly used in the geo- picin, however, are not well tolerated with a high fre-
graphical areas where HIV–TB co-infection is most quency of hepatotoxicity [51, 52]. Thus, such combi-
prevalent; the AUC and trough concentration (lowest nations should be avoided. The recommended

ª 2010 The Association for the Publication of the Journal of Internal Medicine Journal of Internal Medicine 268; 530–539 535
F. Josephson
| Review-Symposium: Drug–drug interactions in the treatment of HIV infection

approach when treating TB in patients receiving a ri- boost the pharmacokinetics of another PI, as drugs in
tonavir-boosted PI regimen is to substitute rifabutin this class exhibit considerable similarity in terms of
for rifampicin. Rifabutin induces CYP3A to a consid- resistance profile and, when ritonavir-boosted, have
erably lower extent than rifampicin, and it does not a very high barrier to resistance. However, if ritonavir
affect exposure to ritonavir-boosted PIs in a clinically is used at boosting doses with an agent that is not a
relevant manner. Rifabutin is considered equipotent PI, resistance to PIs could be selected in cases of regi-
to rifampicin as an anti-TB agent [53]. It induces men failure, because of the presence of sub-thera-
CYP3A considerably less than rifampicin and does peutic ritonavir concentrations.
not affect exposure to ritonavir-boosted PIs in a clini-
cally meaningful way. Unfortunately, however, it is Several CYP3A inhibitors without intrinsic antiretro-
prohibitively expensive in most resource-poor set- viral activity are under development; furthest along
tings. In addition, although there is no relevant effect the path is cobicistat (GS-9350). According to preli-
of rifabutin on the pharmacokinetics of ritonavir- minary data, cobicistat is a weak inhibitor of
boosted PIs, the latter profoundly affect the disposi- CYP2D6, does not inhibit CYP1A2, CYP2C9 or
tion of the former. Rifabutin is metabolized, mainly CYP2C19 and has a low tendency towards the induc-
by non-CYP450 mechanisms, into the active metabo- tion of hepatic drug-metabolizing enzymes [56]. With
lite 25-O-desacetylrifabutin, which in turn is inacti- regard to CYP3A inhibition, cobicistat at a dose of
vated by CYP3A. Under normal circumstances, this 200 mg once daily in a multiple dose regimen pro-
comprises a minor part of the active moiety. However, duced a similar inhibition of oral midazolam clear-
when co-treating with a ritonavir-boosted PI, expo- ance as ritonavir 100 mg once daily [56]. Cobicistat is
sure to rifabutin increases moderately, whereas entering phase III trials, and it remains to be seen
exposure to the active metabolite increases more whether advantages over ritonavir as a booster will be
than 10-fold [5, 6]. Thus, the metabolite becomes a demonstrated in terms of side effect profile. Also of
considerable portion of the active moiety. As rifabutin interest is the putative effect of cobicistat on trans-
has exposure-dependent adverse effects, including membrane drug transporters; however, no data ap-
leucopenia and uveitis, a dose reduction is man- pear to have been published to date.
dated. When used without significant interacting
drugs, the usual dose of rifabutin is 150 or 300 mg Another development in the field of pharmacoki-
once daily. When co-treating with a ritonavir-boosted netic enhancement of antiviral therapy through
PI, the commonly recommended dose regimen is CYP3A inhibition is the boosting of inhibitors of the
150 mg thrice weekly or every other day, which yields hepatitis C virus (HCV) NS3 ⁄ 4A protease. Applica-
an exposure to the active moiety that is still somewhat tions for marketing approval of the first-generation,
greater than 300 mg once daily (and considerably directly acting antivirals against HCV, boceprevir
higher than with150 mg once daily) in the absence of and telaprevir are expected by the end of 2010. In
interacting drugs [5, 6]. phase III studies, both of these drugs have been
administered thrice daily; though, twice daily
administration of telaprevir is under investigation.
Future perspectives on CYP3A inhibition in antiviral therapy
Of interest, several second-generation HCV protease
As discussed previously, ritonavir was originally inhibitors are being investigated in ritonavir-
developed to be used as an antiretroviral agent, acting boosted dosing regimens [57, 58]. Whilst it remains
through inhibition of the HIV protease. In fact, the ini- to be seen whether this will increase therapeutic
tial studies of the combination of ritonavir and an- efficacy, it does provide the pharmacokinetic prere-
other PI were performed under the assumption that quisite for once-daily dosing.
there would be additive pharmacodynamic effects,
rather than to investigate the antiviral consequences
Conclusions
of pharmacokinetic boosting [54, 55]. It should be evi-
dent from the above that ritonavir is in fact not an The aim of this review has not been to provide a com-
‘ideal’ CYP3A inhibitor, for a number of reasons. First, plete overview of pharmacokinetic DDIs in antiretro-
even at boosting doses, ritonavir has gastrointestinal viral therapy, but rather to give an outline of the scope
and metabolic side effects. Secondly, the use of riton- of the problem and the complexity of the mechanisms
avir as a booster is complicated by its effects on multi- involved. Whereas knowledge of DDIs relating to anti-
ple other drug-metabolizing enzymes and transport retroviral agents is substantial amongst physicians
proteins. Thirdly, the fact that ritonavir has intrinsic who regularly treat patients with HIV, awareness of
antiretroviral activity is not a problem when used to the problems because of pharmacoenhancement by

536 ª 2010 The Association for the Publication of the Journal of Internal Medicine Journal of Internal Medicine 268; 530–539
F. Josephson
| Review-Symposium: Drug–drug interactions in the treatment of HIV infection

CYP3A inhibition, as well as of DDIs because of other will no doubt necessitate an increased awareness of
mechanisms, is lower outside the infectious disease the problem of DDIs caused by this strategy, also out-
speciality, where physicians may only rarely see pa- side the specialities prescribing CYP3A-inhibiting
tients treated with antiretroviral therapy. Yet, as dis- pharmacoenhancers.
cussed, several commonly used medications, and
some drugs used to treat co-morbidities that are
Conflict of interest
highly prevalent, may need to be avoided in patients
with certain antiretroviral drug regimens or should No conflict of interest was declared.
prompt dose adjustment of either of the co-treating
agents. Several valuable internet-based DDI data-
References
bases are available (e.g. http://www.hiv-druginter-
actions.org, http://www.janusinfo.se/In-English/), 1 European SPC Sustiva. Bristol-Myers Squibb 2010. Available
some of which are available free of charge. Also, the at: http://www.ema.europa.eu/humandocs/Humans/EPAR/
sustiva/sustiva.htm.
European summaries of product characteristics for
2 European SPC Viramune. Boehringer Ingelheim 2009. Available
many of the antiretroviral drugs, available at the at: http://www.ema.europa.eu/humandocs/Humans/EPAR/
European Medicines Agency home page (http:// viramune/viramune.htm.
www.ema.europa.eu), provide valuable guidance. 3 European SPC Intelence. Janssen-Cilag 2010. Available
at: http://www.ema.europa.eu/humandocs/Humans/EPAR/
In addition to a general awareness of the risk of DDIs, intelence/intelence.htm.
4 European SPC Kaletra. Abbott Laboratories 2010. Available
some very simple general principles may be of use
at: http://www.ema.europa.eu/humandocs/Humans/EPAR/
when facing the problem of a potentially clinically sig- kaletra/kaletra.htm.
nificant interaction in patients receiving antiretrovi- 5 European SPC Prezista. Janssen-Cilag 2010. Available
ral therapy. One is to consider the fact that quite of- at: http://www.ema.europa.eu/humandocs/Humans/EPAR/
ten drugs within the same pharmacodynamic class prezista/prezista.htm.
may have different pharmacokinetics and metabolic 6 European SPC Reyataz. Bristol-Myers Squibb 2010. Available at:
DDI potential (e.g. statins, rifamycins and corticos- http://www.ema.europa.eu/humandocs/Humans/EPAR/rey-
ataz/reyataz.htm.
teroids, as discussed earlier). In such cases, it may
7 Walmsley S, Bernstein B, King M et al. Lopinavir-ritonavir versus
be more rational to look for an equivalent drug that nelfinavir for the initial treatment of HIV infection. N Engl J Med
might not interact, rather than to make a – perhaps 2002; 346: 2039–46.
empirical – dose adjustment, or risk toxicities or lack 8 European SPC Norvir. Abbott Laboratories 2010. Available at:
of efficacy by ignoring the possibility of a DDI. When a http://www.ema.europa.eu/humandocs/Humans/EPAR/nor-
co-treatment is crucial and there are no equally good, vir/norvir.htm.
9 Moyle GJ, Back D. Principles and practice of HIV-protease inhibi-
noninteracting alternatives, consideration may also
tor pharmacoenhancement. HIV Med 2001; 2: 105–13.
be given to altering the antiretroviral regimen, unless 10 Kharasch ED, Hoffer C, Whittington D, Walker A, Bedynek PS.
there is a well-established dose adjustment de- Methadone pharmacokinetics are independent of cytochrome
scribed for use in the particular situation. With an P4503A (CYP3A) activity and gastrointestinal drug transport: in-
increasing number of antiretroviral agents, there sights from methadone interactions with ritonavir ⁄ indinavir.
may be alternatives for many patients; for instance, Anesthesiology 2009; 110: 660–72.
11 Yeh RF, Gaver VE, Patterson KB et al. Lopinavir ⁄ ritonavir in-
using regimens based on raltegravir, which has a
duces the hepatic activity of cytochrome P450 enzymes CYP2C9,
considerably lower tendency to cause DDIs than PIs CYP2C19, and CYP1A2 but inhibits the hepatic and intestinal
and NNRTIs, may sometimes be a way of avoiding activity of CYP3A as measured by a phenotyping drug cocktail in
potentially significant interactions. Furthermore, healthy volunteers. J Acquir Immune Defic Syndr 2006; 42:
when considering co-medications in patients treated 52–60.
with antiretroviral therapy or when facing symptoms 12 Mathias AA, German P, Murray BP et al. Dose-response of ritona-
and signs that may be because of DDIs, it should be vir on hepatic CYP3A activity and elvitegravir oral exposure. Clin
Pharmacol Ther 2009; 85: 64–70.
remembered that several over-the-counter medi-
13 Hsu A, Granneman GR, Cao G et al. Pharmacokinetic interaction
cines, and some natural remedies, may interact with between ritonavir and indinavir in healthy volunteers. Antimicrob
antiretroviral drugs, resulting in toxicities or lack of Agents Chemother, 1998; 42: 2784–91.
efficacy. 14 Merry C, Barry MG, Mulcahy F et al. Saquinavir pharmacokinet-
ics alone and in combination with ritonavir in HIV-infected pa-
Finally, it appears that the concept of pharmacoki- tients. AIDS 1997; 11: F29–33.
15 Sham HL, Kempf DJ, Molla A et al. ABT-378, a highly potent
netic boosting by CYP3A blockade is here to stay, with
inhibitor of the human immunodeficiency virus protease. Anti-
new agents being developed and its use in fields out- microb Agents Chemother 1998; 42: 3218–24.
side antiretroviral therapy under investigation. This

ª 2010 The Association for the Publication of the Journal of Internal Medicine Journal of Internal Medicine 268; 530–539 537
F. Josephson
| Review-Symposium: Drug–drug interactions in the treatment of HIV infection

16 Drewe J, Gutmann H, Fricker G, Török M, Beglinger C, Huwyler 33 Reyataz US prescribing information. Bristol-Myers Squibb 2010.
J. HIV protease inhibitor ritonavir: a more potent inhibitor of Available at: http://www.packageinserts.bms.com/pi/pi_reyat-
P-glycoprotein than the cyclosporine analog SDZ PSC 833. az.pdf.
Biochem Pharmacol 1999; 57: 1147–52. 34 Kharasch ED, Bedynek PS, Park S, Whittington D, Walker A,
17 Perloff MD, Von Moltke LL, Marchand JE, Greenblatt DJ. Hoffer C. Mechanism of ritonavir changes in methadone pharma-
Ritonavir induces P-glycoprotein expression, multidrug cokinetics and pharmacodynamics: I. Evidence against CYP3A
resistance-associated protein (MRP1) expression, and drug mediation of methadone clearance. Clin Pharmacol Ther 2008;
transporter-mediated activity in a human intestinal cell line. 84: 497–505.
J Pharm Sci 2001; 90: 1829–37. 35 Gruber VA, McCance-Katz EF. Methadone, buprenorphine, and
18 Aberg JA. Cardiovascular complications in HIV management: street drug interactions with antiretroviral medications. Curr HI-
past, present, and future. J Acquir Immune Defic Syndr 2009; 50: V ⁄ AIDS Rep 2010; 7: 152–60.
54–64. 36 McCance-Katz EF, Rainey PM, Friedland G, Jatlow P. The prote-
19 Aberg JA. Lipid management in patients who have HIV and are ase inhibitor lopinavir-ritonavir may produce opiate withdrawal
receiving HIV therapy. Endocrinol Metab Clin North Am 2009; 38: in methadone-maintained patients. Clin Infect Dis 2003; 37:
207–22. 476–82.
20 Fichtenbaum CJ, Gerber JG, Rosenkranz SL et al. Pharmacoki- 37 World Health Organisation. HIV ⁄ TB factsheet 2009. Available
netic interactions between protease inhibitors and statins in HIV at: http://www.who.int/tb/challenges/hiv/en/index.html#Recent
seronegative volunteers: ACTG Study A5047. AIDS 2002; 16: %20WHO%20TB/HIV%20publications.
569–77. 38 Cohen K, Meintjes G. Management of individuals requiring anti-
21 Neuvonen PJ, Niemi M, Backman JT. Drug interactions with retroviral therapy and TB treatment. Curr Opin HIV AIDS 2010; 5:
lipid-lowering drugs: mechanisms and clinical relevance. Clin 61–9.
Pharmacol Ther, 2006; 80: 565–81. 39 Treatment of Tuberculosis, Guidelines. World Health Organisa-
22 Sekar VJ, Spinosa-Guzman S, Marien K et al. Pharmacokinetic tion 2010. Available at: http://www.who.int/tb/publications/
drug-drug interaction between the new HIV protease inhibitor cds_tb_2003_313/en/.
darunavir (TMC114) and the lipid-lowering agent pravastatin. 40 Niemi M, Backman JT, Fromm MF, Neuvonen PJ, Kivistö KT
Abstract 55. 8th International Workshop on Pharmacology of Pharmacokinetic interactions with rifampicin : clinical rele-
HIV Therapy. Budapest, Hungary, 2007. vance. Clin Pharmacokinet 2003; 42: 819–50.
23 Annaert P, Ye ZW, Stieger B, Augustijns P. Interaction of HIV pro- 41 Penzak SR, Kabuye G, Mugyenyi P et al. Cytochrome P450 2B6
tease inhibitors with OATP1B1, 1B3, and 2B1. Xenobiotica 2010; (CYP2B6) G516T influences nevirapine plasma concentrations
40: 163–76. in HIV-infected patients in Uganda. HIV Med 2007; 8: 86–91.
24 Glesby MJ, Aberg JA, Kendall MA et al. Pharmacokinetic interac- 42 Haas DW, Ribaudo HJ, Kim RB et al. Pharmacogenetics of efavi-
tions between indinavir plus ritonavir and calcium channel renz and central nervous system side effects: an Adult AIDS Clini-
blockers. Clin Pharmacol Ther 2005; 78: 143–53. cal Trials Group study. AIDS 2004; 18: 2391–400.
25 Chen F, Kearney T, Robinson S, Daley-Yates PT, Waldron S, 43 Klein K, Lang T, Saussele T et al. Genetic variability of CYP2B6 in
Churchill DR. Cushing’s syndrome and severe adrenal suppres- populations of African and Asian origin: allele frequencies, novel
sion in patients treated with ritonavir and inhaled nasal flutica- functional variants, and possible implications for anti-HIV
sone. Sex Transm Infect 1999; 75: 274. therapy with efavirenz. Pharmacogenet Genomics 2005; 15: 861–
26 Hillebrand-Haverkort ME, Prummel MF, ten Veen JH. Ritonavir- 73.
induced Cushing’s syndrome in a patient treated with nasal fluti- 44 Rotger M, Colombo S, Furrer H et al. Influence of CYP2B6 poly-
casone. AIDS 1999. 13: 1803. morphism on plasma and intracellular concentrations and toxic-
27 Gupta SK, Dube MP. Exogenous cushing syndrome mimicking ity of efavirenz and nevirapine in HIV-infected patients. Pharma-
human immunodeficiency virus lipodystrophy. Clin Infect Dis cogenet Genomics 2005; 15: 1–5.
2002; 35: E69–71. 45 Brennan-Benson P, Lyus R, Harrison T, Pakianathan M, Macal-
28 Samaras K, Pett S, Gowers A, McMurchie M, Cooper DA. Iatro- lan D. Pharmacokinetic interactions between efavirenz and rif-
genic Cushing’s syndrome with osteoporosis and secondary ampicin in the treatment of HIV and tuberculosis: one size does
adrenal failure in human immunodeficiency virus-infected not fit all. AIDS 2005; 19: 1541–3.
patients receiving inhaled corticosteroids and ritonavir-boosted 46 Friedland G, Khoo S, Jack C, Lalloo U. Administration of efavi-
protease inhibitors: six cases. J Clin Endocrinol Metab 2005; 90: renz (600 mg ⁄ day) with rifampicin results in highly variable lev-
4394–8. els but excellent clinical outcomes in patients treated for tuber-
29 Foisy MM, Yakiwchuk EM, Chiu I, Singh AE. Adrenal suppres- culosis and HIV. J Antimicrob Chemother 2006; 58: 1299–302.
sion and Cushing’s syndrome secondary to an interaction be- 47 Manosuthi W, Kiertiburanakul S, Sungkanuparph S, Ruxrung-
tween ritonavir and fluticasone: a review of the literature. HIV tham K, Vibhagool A, Rattanasiri S, Thakkinstian A. Efavirenz
Med 2008; 9: 389–96. 600 mg ⁄ day versus efavirenz 800 mg ⁄ day in HIV-infected
30 Gray D, Roux P, Carrihill M, Klein M. Adrenal suppression and patients with tuberculosis receiving rifampicin: 48 weeks
Cushing’s syndrome secondary to ritonavir and budesonide. S results. AIDS 2006; 20: 131–2.
Afr Med J 2010; 100: 296–7. 48 Manosuthi W, Sungkanuparph S, Tantanathip P et al. A random-
31 European SPC Invirase. Roche 2010. Available at: http:// ized trial comparing plasma drug concentrations and efficacies
www.ema.europa.eu/humandocs/Humans/EPAR/invirase/ between 2 nonnucleoside reverse-transcriptase inhibitor-based
invirase.htm. regimens in HIV-infected patients receiving rifampicin: the N2R
32 European SPC Isentress. Merck Sharpe and Dohme 2010. Avail- Study. Clin Infect Dis 2009; 48: 1752–9.
able at: http://www.ema.europa.eu/humandocs/Humans/ 49 Avihingsanon A, Manosuthi W, Kantipong P et al. Pharmacoki-
EPAR/isentress/isentress.htm. netics and 48-week efficacy of nevirapine: 400 mg versus

538 ª 2010 The Association for the Publication of the Journal of Internal Medicine Journal of Internal Medicine 268; 530–539
F. Josephson
| Review-Symposium: Drug–drug interactions in the treatment of HIV infection

600 mg per day in HIV-tuberculosis coinfection receiving rifam- patients with virologic evidence of indinavir or ritonavir failure.
picin. Antivir Ther 2008; 13: 529–36. AIDS 1998; 12: F97–102.
50 Boulle A, Van Cutsem G, Cohen K et al. Outcomes of nevirapine- 56 Mathias AA, German P, Murray BP et al. Pharmacokinetics and
and efavirenz-based antiretroviral therapy when coadministered pharmacodynamics of GS-9350: a novel pharmacokinetic en-
with rifampicin-based antitubercular therapy. JAMA 2008; 300: hancer without anti-HIV activity. Clin Pharmacol Ther 2010; 87:
530–9. 322–9.
51 Nijland HM, L’homme RF, Rongen GA et al. High incidence of ad- 57 Gane E, Rouzier R, Stedman C et al. Ritonavir boosting of low
verse events in healthy volunteers receiving rifampicin and ad- dose RG7227 ⁄ ITMN-191, HCV NS3 ⁄ 4A protease inhibitor,
justed doses of lopinavir ⁄ ritonavir tablets. AIDS 2008; 22: 931– results in robust reduction in HCV RNA at lower exposures than
5. provided by unboosted regimens. Abstract 38. 45th Annual
52 Schmitt C, Riek M, Winters K, Schutz M, Grange S. Unexpected Meeting of the European Association for the Study of the Liver
Hepatotoxicity of Rifampin and Saquinavir ⁄ Ritonavir in Healthy (EASL). 2010: Vienna, Austria.
Male Volunteers. Arch Drug Inf 2009; 2: 8–16. 58 Vierling JM, Poordad F, Lawitz E et al. Once daily narlaprevir
53 Gonzalez-Montaner LJ, Natal S, Yongchaiyud P, Olliaro P. Rifab- (SCH 900518) in combination with PegIntron (peginterferon alfa-
utin for the treatment of newly-diagnosed pulmonary tuberculo- 2b) ⁄ ribavirin for treatment-naive subjects with genotype-1 CHC:
sis: a multinational, randomized, comparative study versus Rif- interim results from NEXT-1, a phase 2a study. Abstract LB4.
ampicin. Rifabutin Study Group. Tuber Lung Dis 1994; 75: 341– 60th Annual Meeting of the American Association for the Study of
7. Liver Diseases (AASLD 2009). 2009: Boston, USA.
54 Cameron DW, Japour AJ, Xu Y et al. Ritonavir and saquinavir
combination therapy for the treatment of HIV infection. AIDS Correspondence: Filip Josephson MD, PhD, Medical Products
1999; 13: 213–24. Agency, P.O. Box 26, SE-751 03, Uppsala, Sweden.
55 Deeks SG, Grant RM, Beatty GW, Horton C, Detmer J, Eastman (fax: +46(0) 18-548566; e-mail: filip.josephson@mpa.se).
S. Activity of a ritonavir plus saquinavir-containing regimen in

ª 2010 The Association for the Publication of the Journal of Internal Medicine Journal of Internal Medicine 268; 530–539 539

You might also like