You are on page 1of 15

Clin Pharmacokinet 2008; 47 (2): 75-89

REVIEW ARTICLE 0312-5963/08/0002-0075/$48.00/0

© 2008 Adis Data Information BV. All rights reserved.

Drug Interactions between HIV Protease


Inhibitors and Acid-Reducing Agents
Ronald W. Falcon1 and Thomas N. Kakuda2
1 Tibotec Therapeutics, Bridgewater, New Jersey, USA
2 Tibotec Inc., Yardley, Pennsylvania, USA

Contents
Abstract . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 75
1. Gastric Acidity and Drug Absorption . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 76
2. Interaction of Protease Inhibitors and Acid-Reducing Agents . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 77
2.1 Atazanavir . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 77
2.1.1 In Combination with Proton Pump Inhibitors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 77
2.1.2 In Combination with Histamine H2-Receptor Antagonists . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 82
2.2 Darunavir . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 83
2.3 Fosamprenavir and Amprenavir . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 83
2.4 Indinavir . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 84
2.5 Lopinavir . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 84
2.6 Nelfinavir . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 85
2.7 Ritonavir . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 85
2.8 Saquinavir . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 86
2.9 Tipranavir . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 86
3. Discussion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 86
4. Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 87

Abstract Maximal efficacy of protease inhibitor-based antiretroviral therapy for HIV/AIDS requires adequate drug
absorption and bioavailability. However, the use of acid-reducing agents in patients treated with highly active
antiretroviral therapy is common and may induce clinically significant drug-drug interactions that alter plasma
protease inhibitor concentrations, which may lead to inadequate viral suppression or adverse events. As plasma
antiretroviral concentrations are not routinely monitored in patients, it is important to understand the risk of these
interactions and counsel patients appropriately, thereby maximizing antiviral potential and preventing the
development of antiretroviral resistance. We therefore reviewed the literature to assess the current understanding
of the effect of various acid-reducing agents on the pharmacokinetics of protease inhibitors.
The bioavailability of fosamprenavir, indinavir, lopinavir, nelfinavir and tipranavir has been reported to be
negatively affected to varying degrees by certain acid-reducing agents. The negative effect on atazanavir
concentrations observed in healthy subjects was more attenuated in HIV-infected patients and warrants further
investigation. While the plasma concentration of saquinavir increased, short-term studies in healthy subjects did
not indicate an increased risk of adverse events. No clinically relevant changes in plasma concentrations of
darunavir occurred when combined with acid-reducing agents. The individual effect of acid-reducing agents on
protease inhibitor concentrations is difficult to predict because of the large interpatient variability of plasma
concentrations with some protease inhibitors. Studies suggest that some of the interactions between protease
inhibitors and acid-reducing agents may be mitigated by temporal separation of dose administration. Educating
patients about the importance of reporting the use of any acid-reducing agents, whether prescription or over-the-
counter, is essential to optimizing the treatment of HIV disease, as is the need for care providers and patients to
76 Falcon & Kakuda

agree upon strategies for managing gastric symptoms and HIV disease simultaneously. Clinicians should be
aware of the potential drug-drug interactions between some protease inhibitors and acid-reducing agents.

Highly active antiretroviral therapy (HAART) has become the drug dissolution and absorption is gastric acidity.[5] Additional
mainstay of treatment for HIV disease because of the significant physiological factors influencing drug absorption include the rate
declines in morbidity and mortality demonstrated with its use.[1,2] of gastric emptying, gastric blood flow and the presence or ab-
The success of treatment with HAART may be limited by regi- sence of food in the stomach. Drug-specific characteristics that
men-related adverse effects, drug-drug interactions and develop- influence absorption include product solubility, ionisability and
ment of drug resistance.[3] A significant impediment to effective lipophilicity.[5] Gastric acidity is modulated by commonly used
therapy is the potential for negative drug-drug interactions, partic- acid-reducing agents, including histamine H2-receptor antagon-
ularly those caused by acid-reducing agents, which may reduce the ists, proton pump inhibitors (PPIs), antacids and buffered medica-
bioavailability of certain protease inhibitors. While some treat- tions (table I).
ment guidelines suggest minimum target concentrations for partic-
Patients undergoing treatment with HAART for HIV infection
ular antiretrovirals in patients with wild-type virus,[3,4] antiretrovi-
frequently use ancillary medications to reduce gastric acidity for
ral blood concentrations are not routinely monitored in the US.
symptomatic relief and treatment of heartburn, gastroesophageal
With limited pharmacokinetic data available in antiretroviral-
reflux disease (GERD) and gastrointestinal tract ulceration. Pa-
experienced patients, minimum protease inhibitor trough concen-
tients also often use over-the-counter (OTC) agents that affect
tration targets are not as clearly defined but are expected to be
gastric acid, such as calcium carbonate, to treat HAART-related
higher. Understanding the likelihood of drug-drug interactions,
diarrhoea.[6] Gastrointestinal adverse events may also prompt pa-
exploring regimens that minimize that likelihood, and educating
tients to interrupt or discontinue antiretroviral therapy. Nausea,
patients about how best to manage their gastric symptoms and HIV
vomiting and diarrhoea were the most common reasons for
disease simultaneously are all essential to optimize the treatment
HAART discontinuation in a retrospective cohort study of
for HIV/AIDS. This review surveys studies of protease inhibitors
antiretroviral-naive patients.[7] Luber et al.[8] evaluated sympto-
and their bioavailability in the presence of acid-reducing agents.
Literature reviews were conducted from November 2005 to June matic treatment of gastrointestinal adverse events in an Internet
2007 to assess the current understanding of the effect of various survey of 200 patients undergoing treatment with HAART. In this
acid-reducing agents on the pharmacokinetics of frequently pre- survey, the incidence of self-reported heartburn, GERD and gas-
scribed protease inhibitors. Published peer-reviewed pharmaco- trointestinal ulceration was 62.0%, 29.5% and 13.0%, respective-
kinetic studies with HIV-infected patients and/or healthy subjects ly. Over half (56.2%) of the patients used only OTC medications
were identified for review through PubMed, mainly by searching for management of gastrointestinal adverse events, while 38.8% of
each protease inhibitor for the term ‘interaction’. Other data
Table I. Commonly prescribed agents that suppress gastric acidity
sources included major HIV/AIDS scientific conferences
Proton pump inhibitors
(2002–7) and current product prescribing information. The main
outcome measures were the area under the plasma concentration- Esomeprazole

time curve (AUC), maximum plasma concentration (Cmax) and Lansoprazole

minimum plasma concentration (Cmin) during the dosing interval Omeprazole

(either 12 or 24 hours) of protease inhibitors coadministered with Pantoprazole


acid-reducing agents in HIV-infected patients and healthy sub- Rabeprazole
jects. In studies with HIV-infected patients, the virological res- Histamine H2-receptor antagonists
ponse, clinical outcome and adverse events/safety were examined, Cimetidine
if reported. Famotidine
Nizatidine
1. Gastric Acidity and Drug Absorption Ranitidine

The efficacy of protease inhibitor therapy requires adequate Other agents

and sustained drug concentrations in the blood and ultimately in Antacids, e.g. aluminum hydroxide/carbonate, calcium hydroxide/
carbonate, bismuth subsalicylate
virally infected cells, which are contingent upon adequate drug
Buffered medications, e.g. didanosine
solubility, absorption and bioavailability. A major determinant of

© 2008 Adis Data Information BV. All rights reserved. Clin Pharmacokinet 2008; 47 (2)
HIV Protease Inhibitor Interactions 77

patients used both OTC and prescription medications, and 5% of the concurrent use of atazanavir and antacids, an interaction would
patients reported using only prescription acid-reducing agents. be expected given the finding that buffered didanosine significant-
Specifically, 51% reported using OTC PPIs or H2-receptor ant- ly reduces atazanavir exposure (see section 2.1.1).
agonists, 39% used prescription PPIs and 77% used antacids. The
2.1.1 In Combination with Proton Pump Inhibitors
investigators found that use of acid-reducing agents by patients
taking a protease inhibitor was similar to that of the entire study Healthy Subjects
population (46% OTC PPIs or H2-receptor antagonists, 42% pre- Pharmacokinetic analyses in healthy subjects have shown that
scription PPIs and 73% antacids). In this study, 88% of patients concurrent use of the PPI omeprazole with ritonavir-boosted or
taking PPIs continued acid-reduction therapy for a period of unboosted doses of atazanavir significantly reduces plasma
6 weeks or more.[8] Similar results have been reported from other atazanavir concentrations. In one study,[24] healthy subjects
clinics in Germany,[9] Italy,[10] Spain,[11] Switzerland[12] and the (n = 48) received atazanavir/ritonavir 300 mg/100 mg once daily,
US.[13] Optimal absorption of some protease inhibitors requires the dose indicated for use in antiretroviral-experienced patients,
increased gastric acidity (a low gastric pH), e.g. atazanavir,[14] for 10 days followed by atazanavir/ritonavir 300 mg/100 mg plus
fosamprenavir[15] and indinavir. [16] However, the acid-reducing omeprazole 40 mg once daily coadministered with (n = 16) or
agents widely used by HIV patients effectively decrease gastroin- without (n = 16) a carbonated beverage (8 oz cola), or atazanavir/
testinal adverse events by reducing gastric acidity and increasing ritonavir 400 mg/100 mg once daily plus omeprazole 40 mg once
the gastric pH. Decreased gastric acidity may impair protease daily (n = 16) for 10 days. Omeprazole was administered in the
inhibitor absorption, leading to decreased drug concentrations, fasted state 2 hours before atazanavir/ritonavir was taken with
which can reduce antiviral activity and increase the likelihood of food. There were significant reductions in the mean AUC from
viral resistance. 0 to 24 hours (AUC24) [76%], Cmax (72%) and Cmin (79%) for
atazanavir when using the standard atazanavir/ritonavir dose in
2. Interaction of Protease Inhibitors and combination with omeprazole (table II). Neither coadministration
Acid-Reducing Agents with a carbonated beverage nor the use of a higher dose of
atazanavir (400 mg) helped to mitigate the negative effect of this
Reduced protease inhibitor bioavailability attributed to nega-
drug-drug interaction.
tive drug-drug interactions with acid-reducing agents has been
In a subsequent study,[25] the atazanavir 400 mg once-daily dose
documented with atazanavir,[14] fosamprenavir,[17] indinavir,[18]
indicated for use in antiretroviral-naive patients was administered
nelfinavir,[19] tipranavir[20] and, possibly, lopinavir.[21] Table II
to healthy subjects (n = 48) for 6 days, followed by atazanavir
summarizes the results of formal drug-drug interaction studies
400 mg plus omeprazole 40 mg once daily coadministered with
between protease inhibitors and acid-reducing agents. It should be
(n = 16) or without (n = 16) a carbonated beverage (8 oz cola) on
noted that formal drug-drug interaction studies of protease medi-
days 7–12, or atazanavir/ritonavir 300 mg/100 mg once daily
cations are almost invariably conducted on healthy subjects, so as
(n = 16) on days 7–16. Omeprazole was administered in the fasted
not to unnecessarily jeopardize the complex medication regimens
state 2 hours before atazanavir was taken with food. Omeprazole
used by people with HIV and also to eliminate the confounding
coadministration, with or without a carbonated beverage, signifi-
factors resulting from their obligatory multidrug regimens.
cantly reduced the mean atazanavir AUC24, Cmax and Cmin by
2.1 Atazanavir >93% (table II). In subjects receiving atazanavir/ritonavir, reduced
mean AUC24 (50%) and Cmax (73%) values were observed for
The aqueous solubility of atazanavir decreases with an increas- atazanavir, although the mean Cmin increased by 23%. Increased
ing pH.[14] Consequently, the concomitant use of PPIs, H2-receptor mean AUC24 (45%) and Cmax values (24%) for omeprazole were
antagonists, antacids or buffered medications might be expected to also reported during coadministration with atazanavir. Given that
reduce the absorption and bioavailability of atazanavir. Pharmaco- omeprazole is metabolized by cytochrome P450 (CYP) 2C19 and
kinetic analyses conducted in healthy subjects and HIV-infected CYP3A,[39,40] the increases in omeprazole concentrations suggest
patients have demonstrated that plasma atazanavir concentrations that atazanavir may be an inhibitor of CYP2C19 and/or CYP3A.
can be significantly altered when acid-reducing agents are Tomilo et al.[26] conducted a single-dose study on the inter-
coadministered. Concerning PPIs, conflicting data among studies action between atazanavir 400 mg and lansoprazole 600 mg in
in healthy subjects and HIV-infected patients have led to consider- nine healthy subjects (table II). Coadministration of lansoprazole
able confusion regarding the clinical implications of this drug- significantly (p < 0.01) decreased the mean atazanavir AUC24
drug interaction. While there appear to be no published reports on (98%) and Cmax (94%). Taken together, the results for similar

© 2008 Adis Data Information BV. All rights reserved. Clin Pharmacokinet 2008; 47 (2)
78
Table II. Bioavailability of commonly used protease inhibitors in the presence of acid-reducing agentsa

Drug and dosage Acid-reducing agent Mean changeb Comments


Cmax (%) Cmin (%) AUC (%)
ATVc
ATV/RTV 300 mg/100 mg od FAM 40 mg bid (n = 16) ↓ 14 ↓ 28 ↓ 18 Addition of 8 oz carbonated beverage did not
compensate for interaction[22]
ATV 400 mg od FAM 40 mg bid (n = 16) ↓ 47 ↓ 42 ↓ 41 Addition of 8 oz carbonated beverage did not
compensate for interaction
Administer ATV 2 h prior to evening dose of FAM or
use ATV/RTV 300–400 mg/100 mg od[22]

© 2008 Adis Data Information BV. All rights reserved.


ATV/RTV 300 mg/100 mg + TDF 300 mg od FAM 40 mg bid (n = 20) ↓ 11 ↓ 33 ↓ 12 Temporal separation does not compensate for
2 h after ATV/RTV + TDF decreased ATV Cmin[23]
ATV/RTV 300 mg/100 mg + TDF 300 mg od FAM 40 mg od (n = 20) ↓ 26 ↓ 23 ↓ 22 Temporal separation does not compensate for
12 h after ATV/RTV + TDF decreased ATV Cmin[23]
ATV/RTV 300 mg/100 mg + TDF 300 mg od FAM 20 mg bid (n = 20) ↓9 ↓ 19 ↓ 10 FAM decreases ATV Cmin during concurrent
concurrent with ATV/RTV + TDF administration with ATV/RTV and TDF[23]
ATV/RTV 300 mg/100 mg + TDF 300 mg od FAM 20 mg bid 2 h after ↓4 ↓ 18 ↓4 Temporal separation does not compensate for
ATV/RTV + TDF (n = 20) decreased ATV Cmin[23]
ATV/RTV 300 mg/100 mg od OME 40 mg od (n = 16) ↓ 72 ↓ 79 ↓ 76 Addition of 8 oz carbonated beverage or increasing
ATV dose to 400 mg did not compensate for
interaction; contraindicated[24]
ATV 400 mg od OME 40 mg od (n = 16) ↓ 96 ↓ 95 ↓ 94 Addition of 8 oz carbonated beverage did not
compensate for interaction; contraindicated[25]
ATV 400 mg LAN 60 mg (n = 9) ↓ 94 ND ↓ 98 Single-dose study[26]

DRV
DRV/RTV 400 mg/100 mg bid RAN 150 mg bid (n = 18) ↓3 ↓6 ↓4 No significant interaction[27]
DRV/RTV 400 mg/100 mg bid OME 20 mg od (n = 18) ↑1 ↑7 ↑4 No significant interaction[27]

FPVd,e
FPV 1400 mg Maalox® TC 30 mL (n = 26) ↓ 35 ↑ 14 ↓ 18 Single-dose study[17]
FPV 1400 mg RAN 300 mg (n = 26) ↓ 51 ↓1 ↓ 30 Single-dose study; use RAN with caution[17]
FPV 1400 mg bid ESO 20 mg od (n = 25) ↓3 ↑3 ↓2 No significant interaction (but possible at higher
ESO doses); ESO AUC increased 55%[28]
FPV/RTV 700 mg/100 mg bid ESO 20 mg od (n = 23) ↓5 ↓7 ↓9 No significant interaction (but possible at higher
ESO doses); ESO AUC unchanged[28]

Continued next page

Clin Pharmacokinet 2008; 47 (2)


Falcon & Kakuda
Table II. Contd

Drug and dosage Acid-reducing agent Mean changeb Comments


Cmax (%) Cmin (%) AUC (%)
IDVf
IDV 400 mg CIM 600 mg bid (n = 12) ↑7 ↓ 18 ↓2 Single-dose study[18]
IDV 800 mg OME 40 mg od (n = 8) ND ND ↓ 10 Single-dose study[29]
IDV 800 mg OME 40 mg od (n = 14) ND ↓ 55 ↓ 47 Single-dose study; compensated with the addition of
RTV 200 mg[30]
HIV Protease Inhibitor Interactions

LPVg
LPV/RTV SGC 400 mg/100 mg bid Antacid, PPI or H2RA (n = 4) ND ↑ 18h ND HIV-infected patients. No apparent effect on LPV

© 2008 Adis Data Information BV. All rights reserved.


trough concentrations. Not a formal interaction
study[31]
LPV/RTV SGC 800 mg/200 mg od Antacid, PPI or H2RA (n = 10) ND ↑ 73h ND HIV-infected patients. Increased LPV trough
concentration. Not a formal interaction study[31]
LPV/RTV tablet 400 mg/100 mg bid RAN 150 mg (n = 12) ↓2 ↓3 ↓2 Single dose of RAN. No significant interaction[21]
LPV/RTV tablet 800 mg/200 mg od RAN 150 mg (n = 11) ↓2 ↓ 20 ↓4 Single dose of RAN. Significant (p < 0.05) decrease
in Cmin[21]
LPV/RTV tablet 400 mg/100 mg bid OME 40 mg od (n = 11) ↑8 ↓1 ↑7 No significant interaction[21]
LPV/RTV tablet 800 mg/200 mg od OME 40 mg od (n = 12) ↑ 11 ↓ 19 ↑7 Significant (p < 0.05) decrease in Cmin[21]

NFV
NFV 1250 mg bid OME 40 mg od (n = 20) ↓ 37 ↓ 39 ↓ 36 Significant interaction.[19] Coadministration not
recommended[32]

RTV
LPV/RTV 400/100 mg bid Antacid, PPI or H2RA (n = 4) ND ↑ 33h ND HIV-infected patients. Increase not statistically
significant. Not a formal interaction study[31]
LPV/RTV 800 mg/200 mg od Antacid, PPI or H2RA (n = 10) ND ↑ 64h ND HIV-infected patients treated. Increase not
statistically significant. Not a formal interaction
study[31]
LPV/RTV 400 mg/100 mg bid RAN 150 mg (n = 12) ↔ ↔ ↔ Single dose of RAN. No significant interaction for
RTV[21]
LPV/RTV 800 mg/200 mg od RAN 150 mg (n = 11) ↔ ↔ ↔ Single dose of RAN. No significant interaction for
RTV[21]
LPV/RTV 400 mg/100 mg bid OME 40 mg od (n = 11) ↔ ↔ ↔ No significant interaction for RTV[21]
LPV/RTV 800 mg/200 mg od OME 40 mg od (n = 12) ↔ ↔ ↔ No significant interaction for RTV[21]

Continued next page


79

Clin Pharmacokinet 2008; 47 (2)


Table II. Contd 80

Drug and dosage Acid-reducing agent Mean changeb Comments


Cmax (%) Cmin (%) AUC (%)
TPV/RTV 500 mg/200 mg OME 40 mg od (n = 15) ↓ 19 ND ↓ 17 Single-dose study.[33] Significant interaction for RTV

SQV

SQV-HGC 600 mg RAN 150 mg (n = 12) ↑ 114 ND ↑ 85 Single-dose study. No correlation between maximum
pH achieved and SQV AUC[34]

SQV-SGC 1200 mg bid CIM 400 mg bid (n = 12) ↑ 179 ↑ 32 ↑ 120 SQV 1200 mg tid for 13 days then 1200 mg bid with
CIM for 12 days[35]

© 2008 Adis Data Information BV. All rights reserved.


SQV-FCT/RTV 1000 mg/100 mg bid OME 40 mg od (n = 22) ↑ 75 ↑ 106 ↑ 82 SQV exposure increased significantly[36]

SQV/RTV bid OME 40 mg od (n = 12) ↑ 55 ↑ 73 ↑ 54 HIV-infected patients. SQV exposure increased


concurrent with SQV/RTV significantly.[37] RTV pharmacokinetics not
significantly changed

SQV/RTV bid OME 40 mg od (n = 12) ↑ 65 ↑ 97 ↑ 67 HIV-infected patients. SQV exposure increased


2 h prior to SQV/RTV significantly.[37] RTV pharmacokinetics not
significantly changed

TPV

TPV/RTV 500 mg/200 mg Maalox® plus 20 mL (n = 23) ↓ 25 ↓ 29 ↓ 27 Single-dose study.[38] Consider separating
administration[20]

TPV/RTV 500 mg/200 mg OME 40 mg (n = 15) ↑5 ND 0 Single-dose study.[33] No significant interaction for
TPV

a All studies were performed in healthy subjects and at steady state unless mentioned otherwise.

b Based on least squares geometric means ratio.

c Exhibits pH-dependent absorption.[14]

d Pharmacokinetics measure amprenavir.

e Exhibits pH-dependent absorption.[15]

f Exhibits pH-dependent absorption.[16]

g Does not exhibit pH-dependent absorption.[21]

h Comparing ‘users’ and ‘non-users’ of acid-suppression agents at week 48.

ATV = atazanavir; AUC = area under the plasma concentration versus time curve; bid = twice daily; Cmax = maximum concentration; Cmin = minimum concentration;
CIM = cimetidine; DRV = darunavir; ESO = esomeprazole; FAM = famotidine; FCT = film-coated tablet; FPV = fosamprenavir; H2RA = histamine H2-receptor antagonist;
HGC = hard-gel capsule; IDV = indinavir; LAN = lansoprazole; LPV = lopinavir; ND = no data; NFV = nelfinavir; od = once daily; OME = omeprazole; oz = ounce(s); PPI = proton
pump inhibitor; RAN = ranitidine; RTV = ritonavir; SGC = soft-gel capsule; SQV = saquinavir; TDF = tenofovir disoproxil fumarate; tid = three times daily; TPV = tipranavir;
↑ indicates increase; ↓ indicates decrease; ↔ indicates no change reported (without value given).

Clin Pharmacokinet 2008; 47 (2)


Falcon & Kakuda
HIV Protease Inhibitor Interactions 81

interaction of lansoprazole and esomeprazole with atazanavir sug- tient,[44] while another showed no effect on atazanavir exposure
gest that this drug-drug interaction may be a PPI class effect. Thus, with lansoprazole coadministration.[45] The value of these reports
the manufacturer of atazanavir recommends that omeprazole and is limited.
other PPIs not be coadministered with atazanavir, even when used Despite the documented negative pharmacokinetic interaction
with low-dose ritonavir.[14] between atazanavir and PPIs and H2-receptor antagonists, data
from small studies suggest that the impact on virological outcomes
HIV-Infected Patients may not be clinically significant. Although plasma atazanavir
Discordant results from studies of atazanavir in healthy volun- concentrations were not measured, no negative effect on the viral
teers and in HIV-infected patients have led to additional investiga- load was observed in a retrospective case review of 14 atazanavir-
tion into this interaction. Across ten HIV clinics in Los Angeles treated, antiretroviral-experienced patients using acid-reducing
(CA, USA), a review of electronic medical records identified 50 agents, PPI (n = 10), H2-receptor antagonists (n = 3) or didan-
patients receiving atazanavir, with or without ritonavir, who were osine-buffered tablets (n = 1).[46] Patients used either atazanavir
concurrently using PPIs or H2-receptor antagonists.[41] Atazanavir with low-dose ritonavir (n = 8) or atazanavir without ritonavir
Cmin values, measured in a central clinical laboratory, were re- (n = 6) doses for a median of 24 weeks (range 8–76 weeks).
corded for 15 patients using PPIs and 19 patients using H2- In a retrospective review of pharmacy and medical records,
receptor antagonists. Based on a reference Cmin value of 0.27 mg/ virological outcomes of patients who received an antiretroviral
L, six of 15 patients (40%) using PPIs had below-reference regimen that included atazanavir/ritonavir with (n = 10) or without
atazanavir Cmin values, five of whom were receiving atazanavir/ (n = 66) concomitant PPI use for at least 6 weeks were compared
ritonavir. Four of 19 patients (21%) using an H2-receptor ant- to assess the clinical relevance of this drug-drug interaction.[47]
agonist, two of whom were receiving atazanavir/ritonavir, also had PPI-treated patients used rabeprazole 20 mg (n = 8) or 40 mg
below-reference atazanavir Cmin values. The mean atazanavir (n = 1), or omeprazole 20 mg once daily (n = 1) for a mean
Cmin was considerably lower in the PPI-treated patients (0.65 mg/ duration of 21.6 weeks (range 6–52 weeks). Virological responses
L; range <0.05–1.85 mg/L) than in those who received H2-recep- reported for PPI-treated and non-PPI-treated patients were com-
tor antagonists (1.12 mg/L; range <0.05–3.17 mg/L). In agreement parable: HIV RNA <50 copies/mL (70% vs 63%) and <500
with the manufacturer’s warning, the investigators concluded that copies/mL (90% vs 83%). While both virological outcome assess-
PPIs should be avoided and H2-receptor antagonists should be ments suggested a lack of effect of this potential drug-drug inter-
administered separately in patients treated with atazanavir. It action, the small numbers of patients evaluated, the wide range of
should be noted that the currently recommended Cmin for follow-up periods and the lack of pharmacokinetic parameters
atazanavir in HIV-infected patients is 0.15 mg/L,[3] and the mini- limit the interpretation of these clinical observations. In another
mum effective concentration for 90% activity (MEC90) in the retrospective review,[48] virological and immunological responses
wild-type virus is ~6–11 ng/mL (9–15 nmol/L) in vitro.[42] were determined in HIV-positive patients who were receiving
A prospective study conducted in two HIV clinics in Paris atazanavir (with or without ritonavir) at recommended doses con-
evaluated and compared atazanavir Cmin values in atazanavir/ currently with either omeprazole 20 mg once daily (n = 15) or
ritonavir-treated patients with or without concurrent use of a pantoprazole 40 mg once daily (n = 17) for a mean duration of 17
PPI.[43] Among 92 patients, 13 reported PPI use (omeprazole and 4 months, respectively. Three patients in each PPI group had a
20 mg [n = 9] or 40 mg [n = 1] once daily, and rabeprazole 30 mg negative therapeutic outcome defined as a 0.5 log10 increase in the
once daily [n = 3]). The median atazanavir Cmin for PPI-treated viral load and/or a CD4 count reduction of >75 cells/mL. Plasma
patients (0.55 mg/L; range 0.20–1.98 mg/L) was comparable to atazanavir trough concentrations measured in four patients were
that reported in non-PPI-treated patients (0.47 mg/L; range above the expected therapeutic minimum concentration. A third
0.65–1.94 mg/L). In three patients, Cmin values obtained during retrospective review consisting of 12 HIV-infected patients indi-
concurrent PPI use and after PPI discontinuation were comparable. cated no clinically relevant effect of PPI and atazanavir coadmin-
Although the investigators concluded that PPI use had no effect on istration.[49] Five of the 12 subjects had a viral load of <400 copies/
the atazanavir Cmin in atazanavir/ritonavir-treated patients, the mL at initiation of atazanavir, with or without ritonavir, which was
Cmin ranges reported indicated that some patients may have had maintained during concurrent atazanavir and PPI therapy. Four
inadequate atazanavir concentrations, although individual patient additional subjects initiated atazanavir, with or without ritonavir,
concentration data were not shown. In published case reports of with a PPI and achieved a viral load of <400 copies/mL. The
individual patients receiving atazanavir/ritonavir, concurrent es- duration of concurrent therapy was 4–23 months in these nine
omeprazole reduced the atazanavir AUC and Cmin in one pa- subjects. In the three remaining patients who did not achieve a

© 2008 Adis Data Information BV. All rights reserved. Clin Pharmacokinet 2008; 47 (2)
82 Falcon & Kakuda

viral load of <400 copies/mL, poor adherence may have impaired least 2 hours before or 1 hour after taking the enteric-coated
the response. A recent single case report[50] suggested no loss of capsule or buffered tablet formulations of didanosine.
the virological or immunological response in a patient switched to
atazanavir (150 mg twice daily) in combination with omeprazole 2.1.2 In Combination with Histamine H2-Receptor Antagonists
(20 mg once daily) for 12 weeks, contrary to recommended Use of H2-receptor antagonists has also been shown to alter
atazanavir administration and coadministration with a PPI. atazanavir pharmacokinetics in formal drug-drug interaction stud-
Several variables may contribute to the inconsistent results and ies in healthy subjects (table II). Reductions in plasma atazanavir
conclusions of these pharmacokinetic and clinical studies of con- concentrations were observed with concurrent use of the
current atazanavir and PPI use. First, plasma atazanavir concentra- H2-receptor antagonist famotidine in healthy subjects. Relative to
tions measured in healthy subjects are approximately twice those concentrations achieved with atazanavir 400 mg once daily, coad-
measured in HIV-infected patients.[14] Therefore, pharmacokinetic ministration of famotidine 40 mg twice daily reduced the mean
assessments of atazanavir conducted in healthy subjects may not atazanavir AUC (41%) and Cmin (42%). However, temporal sepa-
be clinically applicable to HIV-infected populations. Moreover, ration of atazanavir and famotidine doses or use of ritonavir-
the broad range of Cmin values reported in the HIV patient co- boosted atazanavir (atazanavir/ritonavir 300 mg/100 mg once
horts[41,43] demonstrate a high degree of interpatient variability, an daily) with famotidine resulted in atazanavir exposure comparable
observation also reported by other investigators.[51] Hence, until to that achieved with atazanavir 400 mg alone.[22] In an evaluation
strong correlations between plasma atazanavir concentrations and of atazanavir/ritonavir 300 mg/100 mg once daily, coadministra-
the virological or clinical outcome have been established, these tion of famotidine 40 mg twice daily reduced the mean atazanavir
and other protease inhibitor drug concentration data should be AUC (18%) and Cmin (28%).[22] When coadministered with a
interpreted with caution. The particular PPIs used, the dose and the higher atazanavir dose (atazanavir/ritonavir 400 mg/100 mg once
timing of administration relative to atazanavir may also have daily), atazanavir exposure was comparable with that observed
with the standard atazanavir/ritonavir dosage (300 mg/100 mg
varying effects on plasma atazanavir concentrations. Formal stud-
once daily). The investigators also reported that the atazanavir
ies in healthy subjects used high-dose omeprazole (40 mg once
AUC and Cmax values were highly correlated with the average
daily) exclusively, whereas the studies in HIV-infected patients
6-hour gastric pH (r = –0.7).[22] A recently reported study by the
used mostly low doses of omeprazole and various other PPIs. Both
same group[23] indicated that famotidine (20 mg twice daily and
the timing of drug doses relative to blood collection and the timing
40 mg once or twice daily) significantly reduced the mean
of atazanavir administration relative to PPI administration were
atazanavir Cmin (18–33% depending on the schedule) regardless of
standardized and well controlled in the healthy subject studies,
temporal separation of its dosing (concurrent, 2 hours after and
whereas the studies of HIV-infected patients used variable meth-
12 hours after) in relation to coadministration of atazanavir/ritona-
ods with less controlled drug administration.
vir 300 mg/100 mg plus tenofovir 300 mg once daily in healthy
This uncertainty regarding the effect of concurrent PPI use on subjects. When the dosage of famotidine could be compared
plasma atazanavir concentrations is further complicated by drug- (20 mg vs 40 mg twice daily) using the same temporal dosing
drug interactions between atazanavir and other antiretrovirals such schedule (2 hours after coadministration of atazanavir/ritonavir
as tenofovir and didanosine. Because of the negative effect of with tenofovir), the degree of the reduction in atazanavir exposure
tenofovir on plasma atazanavir concentrations,[52] it is recommen- appeared to be related to the famotidine dose (mean Cmax [4% vs
ded that ritonavir-boosted atazanavir is used when combined 11%], Cmin [18% vs 33%], and AUC [4% vs 12%]), although it
with tenofovir.[14] Although data are limited, the concurrent should be noted that this comparison involved separate popula-
use of atazanavir/ritonavir with tenofovir and a PPI in HIV- tions of healthy subjects.
infected patients has not been shown to negatively affect the Based on these data in healthy subjects, different atazanavir
atazanavir Cmin or virological outcomes.[43,47] When combined dose recommendations are indicated according to patient type. For
with atazanavir, the buffered tablet formulation of didanosine has antiretroviral-naive patients, atazanavir 400 mg once daily may be
been shown to reduce the mean atazanavir AUC (87%), Cmax taken with food at least 2 hours before or at least 10 hours after
(89%) and Cmin (84%) in healthy subjects.[53] The more commonly taking an H2-receptor antagonist, or ritonavir-boosted atazanavir
used enteric-coated capsule formulation of didanosine requires (atazanavir/ritonavir 300 mg/100 mg once daily) may be taken
fasted conditions for proper absorption, while atazanavir should be with an H2-receptor antagonist with no regard to the timing of
taken with food for proper absorption. Therefore, dosing recom- doses. For antiretroviral-experienced patients, it is recommended
mendations indicate that atazanavir should be taken with food at that the atazanavir/ritonavir 300 mg/100 mg once-daily dose is

© 2008 Adis Data Information BV. All rights reserved. Clin Pharmacokinet 2008; 47 (2)
HIV Protease Inhibitor Interactions 83

taken with food either at least 2 hours before or at least 10 hours same subjects received ranitidine 300 mg 1 hour before taking
after an H2-receptor antagonist. Although pharmacokinetic data in fosamprenavir 1400 mg. While antacid use reduced the mean
healthy subjects showed that a higher atazanavir 400 mg dose amprenavir AUC24 by 18% and Cmax by 35% and increased the
boosted with ritonavir achieved atazanavir exposure comparable concentration at 12 hours [C12] by 14%, these changes were not
with standard atazanavir/ritonavir 300 mg/100 mg once-daily dos- considered clinically significant by the investigators. Concurrent
ing, long-term use of the higher atazanavir dose in antiretroviral- ranitidine administration reduced the mean amprenavir AUC24
experienced patients may contribute to increased adverse events or and Cmax to a greater degree (by 30% and 51%, respectively), and
other pharmacokinetic concerns. yet the C12 remained unchanged.[17] Based on these findings, it is
suggested that ranitidine and other H2-receptor antagonists be used
2.2 Darunavir with caution when coadministered with fosamprenavir.
An evaluation of fosamprenavir, with and without ritonavir-
In a randomized, three-way, crossover study,[27] coadministra-
boosting, when coadministered with esomeprazole showed no
tion of boosted darunavir with either omeprazole or ranitidine
effect on steady-state amprenavir pharmacokinetics. Healthy sub-
showed no effect on steady-state plasma darunavir pharmaco-
jects (n = 56) received esomeprazole 20 mg once daily for 7 days
kinetics. Healthy subjects (n = 18) each received the following:
and then added either fosamprenavir 1400 mg twice daily or
darunavir/ritonavir 400 mg/100 mg twice daily alone or in combi-
fosamprenavir/ritonavir 700 mg/100 mg twice daily for 14 days.
nation with omeprazole 20 mg once daily or ranitidine 150 mg
Following a 21- to 28-day washout period, subjects then received
twice daily for 4 days, followed by one morning dose on day 5.
fosamprenavir 1400 mg twice daily or fosamprenavir/ritonavir
Each treatment phase was separated by a washout period of at least
700 mg/100 mg twice daily without esomeprazole for another
7 days. Relative to darunavir/ritonavir alone, concurrent use of
14 days. With concurrent esomeprazole, the mean amprenavir
omeprazole or ranitidine had no significant effect (p > 0.05) on the
AUC12, Cmax and C12 remained unchanged in both fosamprenavir
mean AUC from 0 to 12 hours (AUC12), Cmax and Cmin of
treatment arms, suggesting that these agents may be safely
darunavir after multiple dosing. While data in HIV-infected pa-
coadministered.[28] The only significant effect was an increase
tients are not available, these results in healthy subjects suggest
(55%) in the esomeprazole AUC12 after coadministration of es-
that darunavir/ritonavir may be coadministered with either
omeprazole 20 mg once daily with fosamprenavir 1400 mg twice
omeprazole or ranitidine without the need for any dose adjustment.
daily. It should be noted that fosamprenavir and esomeprazole
There appear to be no published studies on the potential for
were simultaneously administered in a fasted state. Therefore, the
interaction between darunavir and antacids.
effects of staggered esomeprazole dosing or simultaneous dosing
of esomeprazole and fosamprenavir with food are unknown.
2.3 Fosamprenavir and Amprenavir
Data from HIV-infected patients are limited to one case report
The solubility of fosamprenavir, a prodrug of amprenavir, has that showed no drug-drug interaction with concurrent use of
been shown to decrease as the pH exceeds 3.3.[15] This pH- fosamprenavir/ritonavir and esomeprazole.[44] It is not possible to
dependant solubility presents a risk of negative drug-drug interac- draw any conclusions from this study. Given that steady-state
tions if fosamprenavir is coadministered with acid-reducing amprenavir pharmacokinetics are comparable between HIV-in-
agents. Also, fosamprenavir contains a phosphate group capable of fected patients and healthy subjects,[28] the results of the previous-
binding to the metal cations present in antacids, which may further ly described acid-reducing agent studies in healthy subjects are
affect the solubility of fosamprenavir or its conversion to the likely to be relevant to HIV-infected populations.
active form, amprenavir.[17] The conflicting observations regarding the impact of the H2-
Pharmacokinetic studies in healthy subjects have shown vary- receptor antagonist ranitidine and the PPI esomeprazole on plasma
ing effects of antacids, H2-receptor antagonists and PPIs when amprenavir concentrations are surprising, since both medications
coadministered with fosamprenavir (measured as plasma am- are effective acid-reducing agents. Moreover, these divergent find-
prenavir concentrations) [table II]. In a single-dose, cross- ings cannot be explained by inferring enzyme inhibition, because
over study,[17] healthy subjects (n = 26) received fosamprenavir ranitidine is not an inhibitor of CYP,[54] unlike esomeprazole,
1400 mg simultaneously with 30 mL of an oral antacid (magnesi- which inhibits CYP2C19.[55] Other differences in the study design,
um hydroxide 1800 mg/aluminum hydroxide dried gel 3600 mg, e.g. the dose or potency of the acid-reducing treatment, may have
Maalox® 1 TC). Following a 4- to 7-day washout period, these contributed to these conflicting results. One plausible explanation

1 The use of trade names is for product identification purposes only and does not imply endorsement.

© 2008 Adis Data Information BV. All rights reserved. Clin Pharmacokinet 2008; 47 (2)
84 Falcon & Kakuda

may be the timing of administration and the amount of time those of an average population curve of 15 other patients also
necessary for esomeprazole activity to occur. If esomeprazole and treated with indinavir 800 mg three times daily and were found to
fosamprenavir are administered simultaneously, gastric acidity be below, above and within the 95% CI of the population curve in
may remain high long enough for fosamprenavir to be converted to four, one and four of the nine patients, respectively. While these
amprenavir and absorbed effectively. Similarly, acid-reducing results suggested a potential drug-drug interaction between in-
agents that have a shorter duration of effect might also be sched- dinavir and omeprazole, interpatient variability of indinavir
uled in a way that does not interfere with fosamprenavir absorp- pharmacokinetics may partially explain some of the differences in
tion. Separation of dosing and/or ritonavir-boosting is suggested, plasma indinavir concentrations. The small number of patients and
as well as careful monitoring of the antiretroviral treatment res- uncontrolled study design also limit the clinical interpretation of
ponse.[3] these data.
There is little published information on the potential interaction There appear to be no published studies on the potential for
between amprenavir and acid-reducing agents. It is advised that interaction between indinavir and antacids, although simultaneous
amprenavir is administered at least 1 hour apart from antacids or administration would be expected to decrease indinavir exposure
the buffered formulation of didanosine, as these medications were given the effects of buffered didanosine. Simultaneous single-dose
stated to decrease plasma amprenavir concentrations when admin- administration of buffered didanosine 200 mg and indinavir
istered concurrently, while no information is given concerning 800 mg in 16 subjects has been reported to significantly decrease
coadministration with H2-receptor antagonists or PPIs.[56] How- the mean indinavir AUC (84%) and Cmax (82%), whereas there
ever, the mean amprenavir AUC12 and C12 were not significantly was little effect when buffered didosine was administered 1 hour
affected by concurrent administration in the fasting state of a after the indinavir dose (the currently recommended temporal
single dose of buffered or enteric-coated didanosine (400 mg) with scheduling), as the mean indinavir AUC and Cmax were decreased
amprenavir 600 mg twice daily for 4 days in a study of 16 healthy by 11% and 4%, respectively.[59] Administration of buffered
subjects.[57] didanosine 400 mg 1 hour before indinavir 800 mg had no statisti-
cally significant effect on the mean indinavir AUC and Cmax
2.4 Indinavir (decreases of 5% and 15%, respectively) in a single-dose study of
12 HIV-infected patients.[60]
Animal studies have indicated that the aqueous solubility of
indinavir is highly dependent on the pH.[16] The results of formal 2.5 Lopinavir
drug-drug interaction studies are summarized in table II. Single-
dose studies with unboosted indinavir (which is currently not used Lopinavir solubility does not appear to be pH dependent.[21]
clinically) in small numbers of healthy subjects indicated no Therefore, the gastric pH and the effects of acid-reducing agents
significant interaction with cimetidine[18] or omeprazole.[29] In a would not be expected to influence plasma lopinavir concentra-
randomized, placebo-controlled, crossover study in 14 healthy tions. In a retrospective analysis of data from a clinical trial
subjects, Rublein et al.[30] determined the effect of administration comparing the soft-gel capsule formulation of lopinavir/ritonavir
of omeprazole 40 mg once daily for 7 days on the pharmaco- 800 mg/200 mg once daily and 400 mg/100 mg twice daily for
kinetics of a single dose of indinavir 800 mg or indinavir/ritonavir 48 weeks in HIV-infected patients (n = 190),[31] patients were
800 mg/200 mg. Coadministration of omeprazole 40 mg with identified as acid-reducing agent ‘users’ (PPIs, H2-receptor ant-
unboosted indinavir decreased the mean indinavir AUC24 and C12 agonists, or antacids) or ‘non-users’. The Cmin values for lopinavir
by 47% (p = 0.001) and 55% (p > 0.05), respectively. When rito- and ritonavir were available for 8–10 and 4–7 ‘users’ versus 76–95
navir plus indinavir was combined with coadministration of and 45–48 ‘non-users’ in the once-daily and twice-daily groups,
omeprazole, the mean indinavir AUC24 increased by 55%, which respectively, at weeks 4, 8, 16, 24 and 48. The patient numbers
was similar to the value attained without omeprazole coadmin- varied dependent on how many patients were assessed and had
istration, and suggests that ritonavir boosting counteracts the ef- evaluable data at each timepoint. The mean lopinavir Cmin was
fect of omeprazole on indinavir exposure. 50% (p = 0.01) and 71% (p = 0.06) higher in ‘users’ than in ‘non-
Data regarding the combined use of indinavir and PPIs have users’ at weeks 24 and 48, respectively, in the group receiving
been summarized in a therapeutic drug monitoring database for lopinavir/ritonavir 800 mg/200 mg once daily, whereas there was
nine HIV-infected patients who received indinavir 800 mg three no difference in the group receiving lopinavir/ritonavir 400 mg/
times daily and omeprazole 20–40 mg once daily.[58] Plasma 100 mg twice daily (table II). The Cmax and AUC values were not
indinavir concentrations in these patients were compared with significantly changed. The same group[61] has recently re-analysed

© 2008 Adis Data Information BV. All rights reserved. Clin Pharmacokinet 2008; 47 (2)
HIV Protease Inhibitor Interactions 85

these data with the addition of 38 HIV-infected patients from that, when coadministered with omeprazole, the systemic expo-
another clinical trial that compared the same formulation of lopi- sure of nelfinavir and its active metabolite are reduced significant-
navir/ritonavir 800 mg/200 mg once daily and 400 mg/100 mg ly, which may result in loss of the virological response and
twice daily for 48 weeks. There was still no significant reduction development of resistance. The major oxidative metabolite of
in the Cmax, Cmin or AUC values for lopinavir in ‘users’ of acid- nelfinavir has in vitro anti-HIV activity comparable with that of
reducing agents, although the Cmin values remained higher in the parent drug, although circulating levels of the active metabolite
patients receiving lopinavir/ritonavir once daily. are substantially lower than those of the parent drug.[62] It is
After the introduction of the new lopinavir/ritonavir tablet therefore unlikely that the reduction in the active metabolite with
formulation, a more formal assessment of concurrent omeprazole omeprazole coadministration is clinically significant. However,
or ranitidine administration was undertaken in healthy subjects.[21] coadministration of omeprazole and nelfinavir is not recommen-
In this parallel-group study, healthy subjects (n = 11–12 per treat- ded.[32] There appear to be no published studies on the potential for
ment) received either lopinavir/ritonavir 800 mg/200 mg once interaction between nelfinavir and antacids or H2-receptor ant-
daily or 400 mg/100 mg twice daily as the new tablet formulation agonists. On the basis of a single-dose study in ten HIV-infected
for 15 days, with the addition of either omeprazole 40 mg once patients,[59] the administration of buffered didanosine 200 mg 1
daily for days 11–15 or a single dose of ranitidine 150 mg on day hour after nelfinavir 750 mg (the currently recommended temporal
11. Relative to either dose of lopinavir/ritonavir alone, coadmin- separation for these agents) increased the mean nelfinavir AUC by
istration with either omeprazole or ranitidine did not significant- 12% and had no effect (or affected it by <10%) on the mean
ly alter the mean lopinavir AUC and Cmax values (table II), nelfinavir Cmax.
although the mean lopinavir Cmin values were significantly re-
duced (p < 0.05) during coadministration with lopinavir/ritonavir 2.7 Ritonavir
800 mg/200 mg once daily (19–20%) but not with lopinavir/
ritonavir 400 mg/100 mg twice daily (1–3%). There do not appear to be any published studies on the inter-
The results of the pharmacokinetic observations reported in action between ritonavir administered as a single agent with
HIV-infected clinical trial participants and in formal drug-drug antacids, H2-receptor antagonists or PPIs. It has been reported that
interaction studies in healthy volunteers suggest that exposure to no clinically significant interaction was observed with respect to
lopinavir is not altered to a clinically relevant degree when lopina- the mean ritonavir AUC and Cmax during coadministration of
vir/ritonavir twice daily is used in combination with acid-reducing buffered didanosine 200 mg twice daily and ritonavir 600 mg
agents. The decrease in the lopinavir Cmin observed when the new twice daily for 4 days in 12 subjects.[59] In studies where ritonavir
tablet formulation of lopinavir/ritonavir 800 mg/200 mg was ad- was administered in combination with lopinavir (see section 2.5
ministered once daily with ranitidine or omeprazole warrants for details), there has been no evidence that coadministration of
further investigation and is difficult to reconcile with earlier obser- acid-reducing agents decreases the bioavailability of ritonavir
vations that lopinavir Cmin values were increased in HIV patients (table II). However, one study[31] reported increases in the mean
receiving long-term lopinavir/ritonavir 800 mg/200 mg once daily ritonavir Cmin values of 63% and 33%, respectively, after receiv-
as the older soft-gel formulation when comparing ‘users’ and ing lopinavir/ritonavir 800 mg/200 mg once daily or 400 mg/
‘non-users’ of acid-reducing agents. 100 mg twice daily as the old soft-gel capsule formulation for 48
There appear to be no published studies on the potential for weeks in HIV patients who were using concurrent acid-reducing
interaction between lopinavir and antacids. agents compared with those who were not; these increases, al-
though substantial, were not statistically significant, and the num-
2.6 Nelfinavir bers of patients analysed as ‘users’ on each dosage (ten and four,
respectively, were low). In a recent study,[33] omeprazole 40 mg
In a two-period, fixed-sequence study,[19] 20 healthy subjects once daily for 5 days decreased the mean ritonavir AUC12 (19%)
received nelfinavir 1250 mg twice daily alone followed by coad- and Cmax (17%) after oral administration of a single dose of
ministration with omeprazole 40 mg once daily, each for 4 days, tipranavir/ritonavir 500 mg/200 mg in 15 healthy subjects. It
separated by a 1-week washout period. The mean AUC, Cmax, and should be noted that the Cmin values were not reported in this
Cmin of nelfinavir were significantly reduced (36–39%) during single-dose study of tipranavir/ritonavir, and nor was the timing of
coadministration with omeprazole (table II). Furthermore, the omeprazole administration in relation to that of tipranavir/ritona-
mean AUC, Cmax and Cmin of its active metabolite were decreased vir. There are no warnings or contraindications against use of
by 92%, 88% and 75%, respectively. The investigators concluded ritonavir with acid-reducing agents.[63]

© 2008 Adis Data Information BV. All rights reserved. Clin Pharmacokinet 2008; 47 (2)
86 Falcon & Kakuda

2.8 Saquinavir macokinetic evaluation in 23 healthy subjects showed that antacid


use significantly reduced tipranavir plasma concentrations. The
Saquinavir has low bioavailability, with 30% of an oral dose volunteers received the recommended dose of ritonavir-boosted
being absorbed and only 4% reaching the systemic circulation.[64] tipranavir (tipranavir/ritonavir 500 mg/200 mg twice daily) for
The absorption of saquinavir is dependent on intestinal 8 days, and then a single dose of tipranavir/ritonavir 500 mg/
CYP3A4.[65] It is therefore now recommended that saquinavir is 200 mg with 20 mL of antacid (magnesium hydroxide 100 mg/
administered in combination with ritonavir to ensure adequate aluminium hydroxide 3600 mg) was coadministered. Concurrent
absorption. Studies using older, unboosted formulations of sa- antacid use reduced the mean tipranavir AUC12, Cmax and C12 by
quinavir showed that coadministration of ranitidine[34] or cime- 25–29% (p < 0.01).[38] Accordingly, it is recommended that
tidine[35] resulted in increased plasma saquinavir concentrations in tipranavir/ritonavir be administered separately from antacids to
healthy subjects, an effect that appeared to be unrelated to the reduce the risk of inadequate tipranavir absorption in patients
effects of these H2-receptor antagonists on gastric acidity (table treated with tipranavir/ritonavir who use antacids.[20] In a recent
II). study,[33] omeprazole 40 mg once daily for 5 days had no statisti-
In a two-sequence study,[36] 18 healthy subjects received the cally significant effect on the tipranavir AUC12 (0%) and Cmax
currently recommended dose for saquinavir film-coated tablets (5% increase) after oral administration of a single dose of
boosted with ritonavir (saquinavir/ritonavir 1000 mg/100 mg tipranavir/ritonavir 500 mg/200 mg in 15 healthy subjects. It
twice daily) for 15 days, with coadministration of omeprazole should be noted that Cmin values were not reported in this single-
40 mg once daily on days 11–15 (table II). Relative to saquinavir/ dose study of tipranavir/ritonavir, nor was the timing of
ritonavir alone, concurrent administration of omeprazole substan- omeprazole administration in relation to that of tipranavir/ritona-
tially increased the mean saquinavir AUC12 (82%), Cmax (75%) vir. There appear to be no published studies on the potential for
and Cmin (106%). The interaction between omeprazole and sa- interaction between tipranavir and H2-receptor antagonists.
quinavir has recently been reported in a crossover study of
12 HIV-infected patients receiving saquinavir/ritonavir twice
3. Discussion
daily.[37] Omeprazole 40 mg once daily for 6 days administered
simultaneously or 2 hours before protease inhibitors increased the
The pH dependency of the aqueous solubility of some protease
saquinavir AUC12 (54% and 67%, respectively), Cmin (73% and
inhibitors (e.g. atazanavir, fosamprenavir and indinavir) is well
97%, respectively) and Cmax (55% and 65%, respectively), where-
documented. Consequently, the gastric pH can have a marked
as ritonavir pharmacokinetics were not significantly changed.
effect on the absorption and bioavailability of these agents. How-
Safety data in both studies indicated that the increased saquinavir
ever, this is not a class effect that is applicable to all protease
exposure did not result in an increased number of adverse events.
inhibitors. Coadministration of acid-reducing agents such as
Of note, these studies were limited in duration and in healthy
antacids, H2-receptor antagonists or PPIs has a profound effect on
subjects. Among 56 HIV-infected patients treated with saquinavir/
the gastric pH and would therefore be expected to affect the
ritonavir 1000 mg/100 mg twice daily for 48 weeks, high plasma
bioavailability of protease inhibitors that have pH-dependent solu-
concentrations of saquinavir were significantly associated with
bility. There is also the potential for specific drug-drug interaction
constitutional adverse effects.[66] Whether long-term coadministra-
between specific protease inhibitors and specific acid-reducing
tion of saquinavir and acid-suppressing agents leads to further
agents by other mechanisms.
adverse effects has not been formally evaluated.
Most patients receiving antiretroviral therapy experience gas-
The potential may therefore exist for increased saquinavir
trointestinal adverse effects at some time and often have to use
exposure when it is coadministered with H2-receptor antagonists
acid-reducing therapy to control symptoms. Initially, they may
or PPIs, probably via mechanisms unrelated to their acid-neutraliz-
turn to intermittent use of OTC medications and eventually to
ing properties. There are no warnings or contraindications against
prescription medication. Regardless, it is important to know
saquinavir/ritonavir use in combination with acid-reducing
whether there are any negative drug-drug interactions between
agents.[67] There do not appear to be any published studies on the
protease inhibitors and acid-reducing agents. During the develop-
potential for interaction between saquinavir and antacids.
ment of protease inhibitors, formal drug-drug interaction studies
2.9 Tipranavir are an essential component of research, and such studies are
usually performed in healthy subjects. However, published infor-
Studies have shown differing effects of acid-reducing agents on mation on potential interactions between each approved protease
the pharmacokinetics of tipranavir (table II). A single-dose phar- inhibitor and representative agents of each of the three main

© 2008 Adis Data Information BV. All rights reserved. Clin Pharmacokinet 2008; 47 (2)
HIV Protease Inhibitor Interactions 87

classes of acid-reducing therapies (antacids, H2-receptor antagon- some protease inhibitors and acid-reducing agents. Some
ists and PPIs) has not been systematically recorded. We reviewed clinicians and investigators consider that the clinical significance
the published literature on these interactions and included infor- of the potential drug-drug interaction between certain protease
mation on the buffered tablet formulation of didanosine. inhibitors and acid-reducing agents in HIV-infected patients may
Given the frequent use of acid-reducing agents by patients with be minimal, and that with careful monitoring, concurrent use may
HIV, special attention must be paid to concomitant conditions and not be prohibitive in all patients. However, there may well be a
medications when selecting a protease inhibitor for antiretroviral clinically significant impact over the long term for two reasons:
therapy. It is possible that patients with HIV who are seeking firstly, because adherence may well slip as the timing of drugs
gastrointestinal symptom control may unwittingly sabotage the within a regimen become more complex (with or without food, 2
efficacy of their antiretroviral therapy by using acid-reducing hours after other medications); and secondly, because of the
agents. Moreover, these negative drug-drug interactions may not genetic barrier to resistance. Ritonavir-boosted protease inhibitors
be identified by the usual safety net, because patients do not are used specifically to obtain higher drug concentrations and
consistently report the use of OTC medications to their healthcare prevent development of resistance. With lower concentrations, the
provider. Initial and ongoing patient counselling must include likelihood of resistance development may possibly be more simi-
information on the interactions of OTC and prescription acid- lar to that of an unboosted protease inhibitor, and patients might
reducing agents with protease inhibitors and should include ac- fail with protease inhibitor mutations, unlike the general trend
ceptable options for controlling gastrointestinal symptoms that showing no primary protease inhibitor mutations when failing a
may be experienced as an adverse effect of HAART. It is critical ritonavir-boosted protease inhibitor. Further study is needed to
for patients to understand these interactions and the associated better clarify the potential for clinically significant interaction
risks, because other providers involved in the care of persons with between individual protease inhibitors and the major classes of
HIV (e.g. in the contexts of primary care, the emergency room or acid-reducing therapy, particularly with respect to the mainten-
urgent care) may not be familiar with the interactions and might ance of clinically effective minimum protease inhibitor concentra-
administer these medications without knowing their potential im- tions in practice for HIV-infected individuals and the potential risk
pact. Although this may increase counselling time with patients, it of resistance development.
would help to ensure the maximal antiviral effect by maintaining
optimal drug exposure while maintaining a high barrier to the
development of drug resistance. Identifying potential negative 4. Conclusion
drug-drug interactions affecting protease inhibitor bioavailability
can help to optimize antiretroviral therapy for patients with HIV Negative drug-drug interactions occur with fosamprenavir
disease, but only if these interactions are generally known. when it is taken concurrently with H2-receptor antagonists or
Clinicians are beginning to use electronic medical records to antacids, although contradictory data suggest a minimal effect of
assess the potential extent of this problem. low-dose esomeprazole on plasma fosamprenavir concentrations.
The individual effect of acid-reducing agents on protease inhib- Atazanavir bioavailability is also significantly decreased by con-
itor concentrations is difficult to predict because of the large current use of acid-reducing therapy with H2-receptor antagonists
interpatient variability of plasma concentrations of some protease and PPIs but appears compensated by ritonavir to some extent;
inhibitors. Furthermore, we feel that the ability to extrapolate however, it is usually recommended that atazanavir administration
within a class of acid-reducing agents is limited by differences in is separated from that of acid-reducing therapy with H2-receptor
individual drug effects on the gastric pH, specific metabolic path- antagonists, and concurrent use of atazanavir and PPIs is not
ways and dosing schedules, and any generalizations might be recommended. Plasma concentrations of indinavir, lopinavir and
potentially misleading. Studies have suggested that some of the tipranavir may be decreased by negative interactions with some
interactions between protease inhibitors and acid-reducing agents products that reduce gastric acidity. Plasma concentrations of
may be mitigated by temporal separation of dose administration. nelfinavir and its active metabolite are significantly decreased by
Education of patients about the importance of reporting the use of coadministration with omeprazole, and so coadministration of
any acid-reducing agents, whether OTC or prescription, is essen- nelfinavir and PPIs is contraindicated. In contrast to all other
tial to optimizing the treatment of HIV, as is the need for care protease inhibitors, exposure to saquinavir has been shown to
providers and patients to agree upon strategies for managing increase when the gastric pH is raised by acid-reducing agents.
gastric symptoms and HIV disease simultaneously. Clinicians No clinically meaningful interaction appears to occur between
should be aware of the potential drug-drug interactions between darunavir or ritonavir with acid-reducing therapy.

© 2008 Adis Data Information BV. All rights reserved. Clin Pharmacokinet 2008; 47 (2)
88 Falcon & Kakuda

16. Lin JH, Chen IW, Vastag KJ, et al. pH-dependent oral absorption of L-735,524, a
Acknowledgements
potent HIV protease inhibitor, in rats and dogs. Drug Metab Dispos 1995; 23:
730-5
Ronald Falcon is the Senior Medical Director at Tibotec Therapeutics and 17. Ford SL, Wire MB, Lou Y, et al. Effect of antacids and ranitidine on the single-
a former Medical Director of HIV at Roche Labs, and holds stock in Johnson dose pharmacokinetics of fosamprenavir. Antimicrob Agents Chemother 2005;
& Johnson. Thomas Kakuda is the Director of Clinical Pharmacology at 49: 467-9
Tibotec Inc. and a former Medical Science Liaison at Roche Labs, and holds 18. Crixivan® (indinavir sulfate) capsules [prescribing information]. Whitehouse Sta-
stock in Johnson & Johnson. The authors thank Peter A. Todd, PhD, for tion (NJ): Merck and Co., Inc., 2006 Aug
his assistance in manuscript editing and preparation, and acknowledge 19. Fang A, Damle B, Labadie R, et al. Omeprazole (OME) significantly decreases
Lori DeLaitsch (Clinical Affairs, Tibotec Therapeutics) for her valuable nelfinavir (NFV) systemic exposure in healthy subjects [abstract no. A-384].
46th Interscience Conference on Antimicrobial Agents and Chemotherapy;
contribution. Financial support for the preparation of this review was provided 2006 Sep 27-30; Chicago (IL)
by Tibotec Therapeutics. The authors have no other conflicts of interest that 20. Aptivus® (tipranavir) capsules, 250 mg [prescribing information]. Ridgefield (CT):
are directly relevant to the content of this review. Boehringer Ingelheim Pharmaceuticals Inc., 2006 Jun
21. Klein CE, Chiu Y-L, Cai Y, et al. Lack of effect of acid reducing agents on the
pharmacokinetics (PK) of lopinavir/ritonavir (LPV/r) tablet formulation [ab-
References stract no L-1003]. 13th Conference on Retroviruses and Opportunistic Infec-
1. Palella Jr FJ, Delaney KM, Moorman AC, et al. Declining morbidity and mortality tions; 2006 Feb 5-9; Denver (CO)
among patients with advanced human immunodeficiency virus infection. HIV 22. Agarwala S, Eley T, Villegas C, et al. Pharmacokinetic effect of famotidine on
Outpatient Study Investigators. N Engl J Med 1998; 338: 853-60 atazanavir with and without ritonavir in healthy subjects [abstract no. 11]. 6th
2. Palella Jr FJ, Baker RK, Moorman AC, et al. Mortality in the highly active International Workshop on Clinical Pharmacology of HIV Therapy; 2005 Apr
antiretroviral therapy era: changing causes of death and disease in the HIV 28-30; Quebec City (QC)
Outpatient Study. J Acquir Immune Defic Syndr 2006; 43: 27-34 23. Agarwala S, Persson A, Eley M, et al. Effect of famotidine 20 and 40mg dosing
3. DHHS Panel on Antiretroviral Guidelines for Adults and Adolescents – A Working regimens on the bioavailability of atazanavir with ritonavir in combination with
Group of the Office of AIDS Research Advisory Council (OARAC). Guide- tenofovir in healthy subjects [abstract no. 568]. 14th Conference on Retrovirus
lines for the use of antiretroviral agents in HIV-1-infected adults and adoles- and Opportunistic Infections; 2007 Feb 25-28; Los Angeles (CA)
cents, Dec 1, 2007 [online]. Available from URL: http://aidsinfo.nih.gov/ 24. Agarwala S, Gray K, Wang Y, et al. Pharmacokinetic effect of omeprazole on
ContentFiles/AdultandAdolescentGL.pdf [Accessed 2007 Dec 1] atazanavir co-administered with ritonavir in healthy subjects [abstract no. 658].
4. Gazzard B, Bernard AJ, Boffito M, et al. British HIV Association (BHIVA) 12th Conference on Retroviruses and Opportunistic Infections; 2005 Feb 22-25;
guidelines for the treatment of HIV-infected adults with antiretroviral therapy Boston (MA)
(2006). HIV Med 2006; 7: 487-503 25. Agarwala S, Gray K, Elay T, et al. Pharmacokinetic interaction between atazanavir
5. Lin JH, Chiba M, Baillie TA. Is the role of the small intestine in first-pass and omeprazole in healthy subjects [abstract no. WePe3.3C08]. 3rd Internation-
metabolism overemphasized? Pharmacol Rev 1999; 51: 135-58 al AIDS Society Conference on HIV Pathogenesis and Treatment; 2005 Jul
6. Turner MJ, Angel JB, Woodend K, et al. The efficacy of calcium carbonate in the 24-27; Rio de Janeiro
treatment of protease inhibitor-induced persistent diarrhea in HIV-infected 26. Tomilo DL, Smith PF, Ogundele AB, et al. Inhibition of atazanavir oral absorption
patients. HIV Clin Trials 2004; 5: 19-24 by lansoprazole gastric acid suppression in healthy volunteers. Pharmacother-
7. O’Brien ME, Clark RA, Besch CL, et al. Patterns and correlates of discontinuation apy 2006; 26: 341-6
of the initial HAART regimen in an urban outpatient cohort. J Acquir Immune
27. Sekar VJ, Lefebvre E, De Paepe E, et al. Pharmacokinetic interaction between
Defic Syndr 2003; 34: 407-14
darunavir boosted with ritonavir and omeprazole or ranitidine in human
8. Luber A, Garg V, Gharakhanian S. Survey of medication used by HIV-infected
immunodeficiency virus-negative healthy volunteers. Antimicrob Agents
patients that affect gastrointestinal (GI) acidity and potential for negative drug
Chemother 2007; 51: 958-61
interactions with HAART [abstract no. P294]. 7th International Congress on
28. Shelton MJ, Ford SL, Borland J, et al. Coadministration of esomeprazole with
Drug Therapy in HIV infection; 2004 Nov 14-18; Glasgow
fosamprenavir has no impact on steady-state plasma amprenavir pharmaco-
9. van Lunzen J, Liess H, Arastéh K, et al. Concomitant use of gastric acid-reducing
kinetics. J Acquir Immmune Defic Syndr 2006; 42: 61-7
agents is frequent among HIV-1-infected patients receiving protease inhibitor-
based highly active antiretroviral therapy. HIV Med 2007; 8: 220-5 29. Hugen PWH, Burger DM, ter Hofstede HJM, et al. Concomitant use of indinavir
and omeprazole: risk of antiretroviral subtherapy [abstract]. AIDS 1998; 12
10. Toma E, Ackad N, Auger S, et al. Upper gastrointestinal complaints and use of acid
Suppl. 4: S29
reducing agents are frequent in patients treated with antiretrovirals [abstract no.
P102]. 8th International Congress on Drug Therapy in HIV infection; 2004 Nov 30. Rublein JC, Donovan BJ, Hollowell SB, et al. Effect of omeprazole (OMP) on the
14-18; Glasgow plasma concentrations of indinavir (IDV) in HIV-negative subjects [abstract no.
11. Burgos A, Garrido J. Use of acid reducing agents among HIV-infected patients A-1621]. 43rd Interscience Conference on Antimicrobial Agents and Chemo-
receiving antiretrovirals in Spain [abstract no. PE4.3/5]. 10th European AIDS therapy; 2003 Sep 14-17; Chicago (IL)
Conference; 2005 Nov 17-20; Dublin 31. Bertz RJ, Chiu YL, Naylor C, et al. Lack of effect of gastric acid reducing agents on
12. Lajaunias F, Beron J. Use of gastric acid-reducing agents in HIV-infected patients lopinavir/ritonavir plasma concentrations in HIV-infected patients [abstract no.
treated with highly active antiretroviral therapy: a Swiss patient survey [abstract 201]. 7th International Congress on Drug Therapy in HIV Infection; 2004 Nov
no. P304]. 8th International Congress on Drug Therapy in HIV infection; 2004 14-18; Glasgow
Nov 14-18; Glasgow 32. Viracept® (nelfinavir mesylate) tablets and oral powder [prescribing information].
13. Fung H, Doan T, Bandres JC. Correlation between the use of acid reducing agents La Jolla (CA): Agouron Pharmaceuticals, Inc., 2006 Apr
and antiretrovirals in HIV+ individuals in the outpatient and inpatient settings 33. la Porte CJL, Cameron DW, Sabo JP, et al. The effect of omeprazole, food and
[abstract no. PE4.3/11]. 10th European AIDS Conference; 2005 Nov 17-20; formulation on the pharmacokinetics of tipranavir coadministered with ritona-
Dublin vir (TPV/r) [abstract no. 59]. 9th International Workshop on Clinical Pharma-
14. Reyataz® (atazanavir sulfate) capsules [prescribing information]. Princeton (NJ): cology of HIV Therapy; 2007 Apr 16-18; Budapest
Bristol-Myers Squibb Co., 2006 Oct 34. Kakuda TN, Falcon RW. Effect of food and ranitidine on saquinavir pharmaco-
15. Furfine ES, Baker CT, Hale MR, et al. Preclinical pharmacology and pharmaco- kinetics and gastric pH in healthy volunteers. Pharmacotherapy 2006; 26:
kinetics of GW433908, a water-soluble prodrug of the human immuno- 1060-8
deficiency virus protease inhibitor amprenavir. Antimicrob Agents Chemother 35. Boffito M, Carriero P, Trentini L, et al. Pharmacokinetics of saquinavir co-
2004; 48: 791-8 administered with cimetidine. J Antimicrob Chemother 2002; 50: 1081-4

© 2008 Adis Data Information BV. All rights reserved. Clin Pharmacokinet 2008; 47 (2)
HIV Protease Inhibitor Interactions 89

36. Winston A, Back D, Fletcher C, et al. Effect of omeprazole on the pharmaco- 52. Taburet AM, Piketty C, Chazallon C, et al. Interactions between atazanavir-
kinetics of saquinavir 500mg formulation with ritonavir in healthy male and ritonavir and tenofovir in heavily pretreated human immunodeficiency virus-
female volunteers. AIDS 2006; 20: 1401-6 infected patients. Antimicrob Agents Chemother 2004; 48: 2091-6
37. Singh K, Dickinson L, Back D, et al. Saquinavir plasma pharmacokinetics during 53. Reyataz® (atazanavir sulfate) capsules [prescribing information]. Princeton (NJ):
co-administration with omeprazole in HIV infected subject [abstract no. 62]. Bristol-Myers Squibb Company, 2006
8th International Workshop on Clinical Pharmacology; 2007 Apr 16-18; Buda-
54. Zantac® (ranitidine hydrochloride) [prescribing information]. Research Triangle
pest
Park (NC): GlaxoSmithKline, 2007 Jun
38. Van Heeswijk R, Sabo JP, Cooper C, et al. The pharmacokinetic interactions
between tipranavir/ritonavir 500/200mg bid (TPV/r) and atorvastatin, antacid, 55. Blume H, Donath F, Warnke A, et al. Pharmacokinetic drug interaction profiles of
and CYP3A4 in healthy adult volunteers [abstract no. 35]. 5th International proton pump inhibitors. Drug Saf 2006; 29: 769-84
Workshop on Clinical Pharmacology of HIV Therapy; 2004 April 1-3; Rome 56. Agenerase® (amprenavir) capsules [prescribing information]. Research Triangle
39. Abelo A, Andersson TB, Antonsson M, et al. Stereoselective metabolism of Park (NC): GlaxoSmithKline, 2005 May
omeprazole by human cytochrome P450 enzymes. Drug Metab Dispos 2000; 57. Shelton MJ, Giovanniello AA, Cloen D, et al. Effects of didanosine formulations
28: 966-72 on the pharmacokinetics of amprenavir. Pharmacotherapy 2003; 23: 835-42
40. Yamazaki H, Inoue K, Shaw PM, et al. Different contributions of cytochrome P450 58. Burger DM, Hugen PW, Kroon FP, et al. Pharmacokinetic interaction between the
2C19 and 3A4 in the oxidation of omeprazole by human liver microsomes: proton pump inhibitor omeprazole and the HIV protease inhibitor indinavir.
effects of contents of these two forms in individual human samples. AIDS 1998; 12: 2080-2
J Pharmacol Exp Ther 1997; 283: 434-42
59. Videx® EC (didanosine) [prescribing information]. Princeton (NJ): Bristol-Myers
41. Khanlou H, Farthing C. Co-administration of atazanavir with proton-pump inhibi- Squibb, 2003
tors and H. J Acquir Immune Defic Syndr 2005; 39 (4): 503
60. Shelton MJ, Mei H, Hewitt RG, et al. If taken 1 hour before indinavir (IDV),
42. Robinson BS, Riccardi KA, Gong Y-F, et al. BMS-232632, a highly potent human
didanosine does not affect IDV exposure, despite persistent buffering effects.
immunodeficiency virus protease inhibitor that can be used in combination with
Antimicrob Agents Chemother 2001; 45: 298-300
other available antiretroviral agents. Antimicrob Agents Chemother 2000; 44:
2093-9 61. Chiu YL, Klein CE, Woodward WC, et al. Lack of effect of gastric acid-reducing
agents on the pharmacokinetics of lopinavir/ritonavir in HIV-infected patients.
43. Guiard-Schmid JB, Poirier JM, Bonnard P, et al. Proton pump inhibitors do not
AIDS Patient Care STDs 2007; 21: 247-51
reduce atazanavir concentrations in HIV-infected patients treated with ritona-
vir-boosted atazanavir [letter]. AIDS 2005; 19: 1937-8 62. Zhang KE, Wu E, Patrick AK, et al. Circulating metabolites of the human
44. Kiser JJ, Lichtenstein KA, Anderson PL, et al. Effects of esomeprazole on the immunodeficiency virus protease inhibitor nelfinavir in humans: structural
pharmacokinetics of atazanavir and fosamprenavir in a patient with human identification, levels in plasma, and antiviral activities. Antimicrob Agents
immunodeficiency virus infection. Pharmacotherapy 2006; 26: 511-4 Chemother 2001; 45: 1086-93

45. Kosel BW, Storey SS, Collier AC. Lack of interaction between atazanavir and 63. Norvir® (ritonavir capsules) soft gelatin [prescribing information]. North Chicago
lansoprazole. AIDS 2005; 19: 637-8 (IL): Abbott Laboratories, 2006 Jan
46. Antoniou T, Yoong D, Beique L, et al. Impact of acid-suppressive therapy on 64. Williams P, Sampson A, Green C. Disposition and bioavailability of the HIV-
virologic response to atazanavir-based regimens in antiretroviral-experienced proteinase inhibitor, Ro 31-8959, after single doses in healthy volunteers.
patients: a case series. J Acquir Immune Defic Syndr 2005; 39: 126-8 Br J Clin Pharmacol 1992; 34: 155-6
47. Furtek KJ, Crum NF, Olsen PE, et al. Proton pump inhibitor therapy in atazanavir- 65. Fitzsimmons ME, Collins JM. Selective biotransformation of the human immuno-
treated patients: contraindicated? J Acquir Immune Defic Syndr 2006; 41: 394- deficiency virus protease inhibitor saquinavir by human small-intestinal cyto-
6 chrome P4503A4: potential contribution to high first-pass metabolism. Drug
Metab Dispos 1997; 25: 256-66
48. Chang J, Towner W, Kerrigan HL, et al. Evaluation of concomitant use of protease
inhibitor (PI)-atazanavir (ATV) with proton pump inhibitor (proton pump 66. Lötsch J, Harder S, Sturmer M, et al. Association of saquinavir plasma concentra-
inhibitor) [abstract no. WePe3.3C04]. 3rd International AIDS Society Confer- tions with side effects but not with antiretroviral outcome in patients infected
ence on HIV Pathogenesis and Treatment; 2005 Jul 24-27; Rio de Janeiro with protease inhibitor-susceptible human immunodeficiency virus type 1.
49. Sahloff EG, Duggan JM. Clinical outcomes associated with concomitant use of Antimicrob Agents Chemother 2007; 51: 3264-72
atazanavir and proton pump inhibitors. Ann Pharmacother 2006; 40: 1731-6 67. Invirase® 500 (saquinavir mesylate) capsules and tablets [prescribing information].
50. Chan-Tack KM, Edozien A. Ritonavir-boosted atazanavir may be efficacious in Nutley (NJ): Roche Laboratories Inc., 2005 Sep
HIV-infected patients concurrently receiving omeprazole [letter]. Clin Infect
Dis 2006; 42: 1344
51. Winston A, Bloch M, Carr A, et al. Atazanavir trough plasma concentration Correspondence: Dr Ronald W. Falcon, Tibotec Therapeutics, 430 Route 22
monitoring in a cohort of HIV-1-positive individuals receiving highly active East, Bridgewater, NJ 08807, USA.
antiretroviral therapy. J Antimicrob Chemother 2005; 56: 380-7 E-mail: rfalcon@tttus.jnj.com

© 2008 Adis Data Information BV. All rights reserved. Clin Pharmacokinet 2008; 47 (2)

You might also like