Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2012 Sep 1.
Published in final edited form as: Lancet Oncol. 2011 May 12;12(9):905–912. doi: 10.1016/S1470-2045(11)70056-0

Use of antineoplastic agents in cancer patients with HIV/AIDS

Michelle A Rudek 1, Charles Flexner 2,3, Richard F Ambinder 1,3
PMCID: PMC3343721  NIHMSID: NIHMS310733  PMID: 21570912

Abstract

In the era of highly active antiretroviral therapy (HAART), patients with human immunodeficiency virus (HIV) have reduced morbidity and mortality of AIDS-related complications. However, there is an increase in the prevalence of AIDS-defining and non-AIDS-defining cancers. This article provides an up-to-date review of management of HAART pharmacotherapy in the context of cytotoxic chemotherapy or targeted antineoplastic agents.

Keywords: antiretroviral, antineoplastic agents, pharmacology

Introduction

Treatment of patients with human immunodeficiency virus (HIV) infection with highly active antiretroviral therapy (HAART) substantially restores immune function, reduces opportunistic infections, lowers plasma viral RNA load, and reduces morbidity and mortality of AIDS-related complications.1,2 However, cancer remains a significant problem in patients with HIV/AIDS. Kaposi's sarcoma, non-Hodgkin's lymphoma, and invasive cervical cancer are not typical in the immunocompetent adult population but are prevalent in those with HIV/AIDS and have thus been deemed as “AIDS-defining cancers.” Hodgkin's lymphoma, anal, lung, and testicular germ cell cancers lack the same definitive association with HIV/AIDS and are being deemed as “non-AIDS-defining cancers” when the patient also has a co-diagnosis of HIV/AIDS.

The number of non-AIDS-defining cancers has increased significantly as patients with HIV/AIDS have increased life expectancies. In the Antiretroviral Therapy Cohort Collaboration that examined 39,272 patients diagnosed with HIV who initiated antiretroviral therapy during the time period of 1996 until 2006, 1597 patients had a documented cause of death.3 A total of 236 patients (14.8% of 1,597) have died of AIDS-defining cancers from 1996 to 2006 but the proportion has declined from 20.5% (70/341) during 1996-1999 to 12.5% (78/624) during 2003-2006. While 189 patients (11.8% of 1,597) have died of non-AIDS-defining cancers during the same time period, the proportion has increased from 7.3% (25/341) during 1996-1999 to 15.4% (96/624) during 2003-2006. A similar trend was also noted when assessing the 5-year cumulative incidence of cancer in 472,378 individuals with HIV or AIDS who were cancer-free at the time of diagnosis from 1980 to 2006 in 3 distinct timeframes: 83,789 patients from 1980-1989 (prior to antiretroviral use), 213,029 patients from 1990-1995 (monotherapy/dual therapy with antiretroviral drugs), and 175,560 patients from 1996-2006 (HAART).4 There was a decline in cumulative incidence of AIDS-defining cancers from 18% (15,728/83,789) to 11% (23,603/213,029) to 4.2% (7,570/175,560) over the 3 timeframes and a rise in non-AIDS-defining cancers from 1.1% (1,056/83,789) to 1.5% (4,348/213,029) with no change noted from 1996-2006 (2,911/175,560). While there was no change noted from 1990-1995 to 1996-2006, the incidence for non-AIDS-defining cancers such as anal cancer, Hodgkins lymphoma, and liver cancer did have a continued increase in incidence in all timeframes.

While the type of cancer HIV patients are getting may be changing, the need for treatment with concurrent antineoplastic agents and HAART is increasingly common. The potential of HAART to cause drug interactions is well documented.5,6 However, little is known about the interaction potential of either cytotoxic or targeted antineoplastic agents with HAART. In addition to pharmacokinetic drug interactions, overlapping toxicities are also possible. This review will highlight what is known about potential pharmacologic interactions between antiretroviral and antineoplastic therapy. We will also consider how to combine antiretroviral and antineoplastic agents in patients with HIV who are on HAART therapy.

Antiretroviral Therapy

Antiretroviral Drug Classes

Current antiretroviral drugs classes include: nucleoside or nucleotide reverse-transcriptase inhibitors (NRTIs), non-nucleoside reverse-transcriptase inhibitor (NNRTIs), HIV-1 protease inhibitors (PI), integrase strand transfer inhibitors (INSTI), fusion inhibitors, and entry inhibitors which include chemokine receptor antagonists.7 All recommended HAART regimens include a minimum of three active drugs to prevent resistance, with initial regimens including combinations of two NRTIs with an NNRTI, a PI boosted with ritonavir, or an INSTI.8 Table 1 provides an overview of the potential drug interactions of each antiretroviral drug class with regards to the primary elimination route and alterations in drug metabolizing enzymes with emphasis on potential for interactions with anticancer agents. The potential for overlapping toxicities with anticancer agents and each antiretroviral drug class will be discussed in the toxicity section.

Table 1. Drug Interaction Potential of HAART.

Drug Route of Elimination (Substrate) Effect on CYP450/Transporters Potential for Clinically Significant Pharmacokinetic Drug Interactions Ref.
Effect on ARVs (substrate) Effect on Cancer Drugs (due to enzyme or transporter induction or inhibition)
Nucleoside reverse-transcriptase inhibitors (NRTIs)
Abacavir Renal excretion, ALDH, UGT1 None known Unlikely Unlikely 9-11
Didanosine Renal excretion, purine nucleoside phosphorylase None known Unlikely Unlikely 12
Emtricitabine Renal excretion None known Unlikely Unlikely 13
Lamivudine Renal excretion None known Unlikely Unlikely 14
Stavudine Renal excretion None known Unlikely Unlikely 15
Zidovudine UGT2B7 None known Possible Unlikely 16,17
Nucleotide reverse-transcriptase inhibitors (NtRTIs)
Tenofovir Renal excretion Weak inhibitor: CYP1A2 Unlikely Possible 18
Non-nucleoside reverse-transcriptase inhibitor (NNRTIs)
Delavirdine CYP2D6, CYP3A4 Inhibitor: CYP2C9/2C19, CYP2D6, CYP3A4 Possible Possible (inhibitor) 19,20
Efavirenz CYP2B6>CYP3A, UGT2B7 Inducer: CYP2B6, CYP3A4
Inhibitor: CYP2C9/2C19. CYP3A4
Possible Highly likely (inducer) 21-24
Etravirine CYP2C9/2C19, CYP3A4, UGT1 Weak inducer: CYP2B6, CYP3A4
Weak inhibitor: CYP2C9/2C19, ABCB1
Possible Highly likely (inducer) 25,26
Nevirapine CYP2B6, CYP3A4, UGT1 Potent inducer: CYP2B6, CYP3A4 Possible Highly likely (inducer) 26-28
Ritonavir or ritonavir–boosted HIV-1 protease inhibitors (PI)
Amprenavir CYP3A4>CYP2D6, CYP2C9, ABCB1, UGT1 Strong to weak inhibitor: CYP3A
Inducer: CYP3A4, ABCB1
Possible Definite (inhibitor)2 29-33
Darunavir CYP3A4 Inhibitor: CYP3A4 Possible Definite (inhibitor)2 34
Fosamprenavir (prodrug) Hydrolyzed to amprenavir See amprenavir Possible Definite (inhibitor)2 29,35
Indinavir CYP3A4, ABCB1, ABCC2, UGT1 Strong to weak inhibitor: CYP3A>CYP2D6, UGT1A1 Possible Definite (inhibitor)2 29,32,36-41
Lopinavir CYP3A, ABCC2 Strong to weak inhibitor: CYP3A4>UGT1A1 Possible Definite (inhibitor)2 41-44
Ritonavir CYP3A4 >CYP2D6, ABCB1, ABCC2 Inducer: CYP2B6, CYP2C9/2C19, CYP3A4, UGT1
Inhibitor: CYP3A>CYP2D6>CYP2C9
Possible Definite (inhibitor or inducer) 29,32,36, 38,39,45-49
Saquinavir CYP3A4, ABCB1, ABCC2 Weak inhibitor: CYP3A, CYP2C9>CYP2D6, UGT1A1, ABCB1, ABCC2 Possible Definite (inhibitor)2 29,32,36, 38,39,41, 44
Tipranavir CYP3A4, UGT1, ABCB1 Inducer: CYP3A4, ABCB1
Inhibitor: CYP1A2, CYP2C9/2C19, CYP2D6
Possible Definite (inhibitor or inducer)2 50-52
Non-ritonavir boosted HIV-1 protease inhibitors (PI)
Atazanavir CYP3A4, ABCB1 Inhibitor: CYP3A4>CYP2C8, UGT1A1, ABCC2 Possible Possible (inhibitor) 41,44,53-56
Nelfinavir CYP2C19> CYP3A4 CYP2D6, CYP2C9 Inducer: CYP2C9, CYP3A4, ABCB1
Inhibitor: CYP3A>CYP2D6
Possible Possible (inhibitor or inducer) 29,31,36, 38,57-59
Integrase strand transfer inhibitors
Raltegravir UGT1A1 None known Possible Unlikely 60
Fusion inhibitors
Enfuvirtide Catabolism None known Unlikely Unlikely 61
Entry inhibitors (Chemokine receptor antagonists)
Maraviroc CYP3A4, ABCB1 None known Possible Unlikely 62-65

Abbreviations: ABCB1, ATP-binding cassette sub-family B member 1 (a.k.a., P-glycoprotein); ABCC2, ATP-binding cassette sub-family C member 2 (a.k.a., CMOAT, MRP2); ALDH, alcohol dehydrogenase; ARV, antiretroviral; CYP, cytochrome P450; UGT, Uridine 5′-diphospho-glucuronosyltransferase

1

Isozyme not specified.

2

When used as a ritonavir-boosted PI.

NRTIs inhibit the activity of HIV reverse transcriptase, an enzyme that copies the viral single stranded RNA into a double-stranded DNA. Nucleoside NRTIs or nucleotide NRTIs (NtRTIs) compete for incorporation into DNA with naturally occurring deoxynucleotides. NRTIs have relatively short plasma half-lives but have longer intracellular half-lives thus allowing for once daily administration for most NRTIs or NtRTIs. Certain NRTI/NtRTI-based regimens are associated with anemia, dyslipidemia, gastrointestinal symptoms, insulin resistance, neutropenia, nephrotoxicity, lactic acidosis associated with hepatic steatosis, noncirrhotic portal hypertension, pancreatitis, peripheral neuropathy, and an increased risk of cardiovascular events.

NNRTIs bind to a pocket distant from the enzyme active site, and inhibit HIV reverse transcriptase by inducing conformational changes. Single point mutations in reverse transcriptase can dramatically alter virus susceptibility to first generation NNRTIs. Etravirine is a second generation NNRTI with the ability to bind to various conformations of the reverse transcriptase enzyme thus maintaining activity with single point mutations that confer resistance to first generation NNRTIs. Various NNRTI-based regimens are associated with rash, central nervous system toxicity, and hepatic transaminase elevations.

PIs block viral replication by preventing the HIV-1 protease from cleaving precursor proteins necessary to form infectious virions. Multiple mutations in the HIV protease enzyme are required to develop high level resistance to most PIs. Various PI-based regimens have been associated with dyslipidemia, fat maldistribution, gastrointestinal symptoms, insulin resistance, hepatic transaminase elevations, hyperbilirubinemia, and an increased risk of cardiovascular events.

Integrase strand transfer inhibitors block the final step in integration of viral genes into the host cell DNA. Single point mutations in integrase have been noted to cause resistance to the integrase strand transfer inhibitor, raltegravir.66,67 Raltegravir is the first approved integrase inhibitor and has been associated with creatine kinase laboratory abnormalities, headache, insomnia, myopathy, rash, and rhabdomyolysis.

Fusion inhibitors interfere with the entry or fusion of HIV-1 to the host cell by blocking one of several targets including the viral envelope protein or a chemokine co-receptor (i.e., chemokine co-receptor 5 (CCR5)). Enfuvirtide, a synthetic peptide, was the first fusion inhibitor approved is injected twice daily but is currently reserved for heavily treatment-experienced patients only. Side effects associated with enfuvirtide include diarrhea, fatigue, injection site reactions, and nausea. Maraviroc is an entry inhibitor which binds the chemokine receptor CCR5 and is approved for the use in patients who have CCR5-trophic virus. Maraviroc has been associated with dizziness, hepatotoxicity, pyrexia, rash, and upper respiratory tract infections.

Initiating and Stopping Antiretroviral Therapy

Guidelines for developed countries now recommend that treatment is offered to HIV infected patients with: 1) a history of an AIDS-defining illness or 2) a CD4 lymphocyte count of <500 cells/mm3.8 Resistance testing is recommended for patients with HIV infection prior to initiating HAART treatment. The ultimate goal of therapy is to preserve or improve immune function while decreasing HIV-associated morbidity and mortality. Initial regimens should be selected to allow for maximal compliance while taking into consideration comorbidities, pretreatment genotypic drug resistance testing, and drug-specific factors such as convenience, drug interaction potential and side effect profiles. All NNRTI-based and PI-based regimens are typically administered once or twice daily.

Due to tolerability and viral sensitivity, the initial HAART regimen can change over time with special considerations being given to starting and stopping any component in the regimen. New regimens should contain at least two, and preferably three, active drugs from multiple antiretroviral drug classes. If treatment failure is suspected, compliance, tolerability, pharmacokinetic related issues, drug resistance, immunologic and virologic failure should be considered.

Caution is warranted when stopping treatment because of the risk of creating a resistant HIV strain.68 In cases of severe or life-threatening toxicity, all components of the regimen should be stopped simultaneously. If drugs that have differing half-lives are stopped simultaneously, this may result in functional monotherapy if the drug with the longest half-life remains in circulation for a prolonged period after the short half-life drugs are eliminated (i.e., longer half-life NNRTIs with short half-life NRTIs). In this case, the strategy varies from either a staggered stop, or an exchange or replacement of an NNRTI with a PI to decrease the risk of functional monotherapy. The best approach to discontinuing therapy is unproven.

Drug Interactions with Antiretroviral Therapy

The pharmacokinetic drug interaction potential with the various antiretroviral therapies is considerable. Since each drug is metabolized by differing metabolic isozymes, generalizations based on each class are not possible (see Table 1). The drug interaction potential for NRTIs and NtRTIs is minimal but may occur if another drug alters renal clearance and/or intracellular phosphorylation. Tenofovir has been shown to cause unexpected changes in the concentration of other antiretroviral drugs, in some cases reflecting possible effects on drug transporters.69,70 There is a high potential for pharmacodynamic interactions with some NRTIs, for example those causing hematologic toxicity. For PIs and NNRTIs, which are extensively metabolized by and induce or inhibit the CYP450 system, the drug interaction potential is high. Raltegravir undergoes glucuronidation by UGT1A1 and has lower drug interaction.60 Maraviroc is a substrate of the CYP3A enzyme and ABCB1 transporter and susceptible to multiple drug interactions.62,63

For the majority of antiretroviral drugs that are CYP450 substrates, inducers, or inhibitors, coadministration with other metabolized drugs could result in drug accumulation and possible toxicity, or decreased efficacy of one or both drugs. Drug interaction resources should be consulted when determining interaction potential (see Table 2). For example, with the benzodiazepam class, PIs should not be coadministered with alprazolam, diazepam, oral midazolam, and triazolam but can be coadministered with lorazepam, oxazepam, or temazepam.8 Intravenous midazolam utilized for conscious sedation should be used at a reduced dose and with caution when combined with ritonavir PI-based therapy.71

Table 2. Antiretroviral Drug Interaction Resources.

Resource Website Source
AIDSinfo www.aidsinfo.nih.gov U.S. Department of Health and Human Services (DHHS)
AIDSMeds aidsmeds.com/ HIV+ patients and physicians
Clinical Care Options clinicalcareoptions.com/ Clinical Care Options
FDA MedWatch www.fda.gov/Safety/MedWatch/default U.S. Food and Drug Administration (FDA)
HIV InSite www.hivinsite.com University of California San Francisco
HIVMA www.hivma.org HIV Medical Association
HIV medication guide www.hivmedicationguide.com J. Antony Gagnon, Pharm.D., B.Pharm., D.P.H., CDE, CAE and Rachel Therrien, Pharm., D.P.H.,MSC
Johns Hopkins POC IT Center: HIV Guide www.hopkins-hivguide.org Johns Hopkins University
The Internet's HIV/AIDS Oral Healthcare Resource www.HIVdent.org US nonprofit dentists, nurses, pharmacists
Medscape www.medscape.com WebMD Health Professional Network
Toronto General Hospital Immunodeficiency Clinic www.tthhivclinic.com/ Toronto General Hospital
University of Liverpool HIV Drug Interactions Web site www.hiv-druginteractions.org University of Liverpool HIV Pharmacology Group

Antiretroviral Therapy and Anticancer Treatment

As HIV patients continue to live longer and develop AIDS-related malignancies or non-AIDS-defining cancers, more information on how to treat patients with anticancer treatment will be needed. Drug interactions are certainly one aspect, but overlapping toxicities are also a concern. Several antiretrovirals, including didanosine, stavudine, and zidovudine, have significant toxicities described below and are therefore not utilized in first-line HAART regimens in the developed world.

Assessing hepatic function in patients on antiretrovirals

Bilirubin is often used as a guide for dose adjustment for cancer chemotherapy agents such as docetaxel,72 doxorubicin,73 etoposide,74 imatinib,75 irinotecan,76 paclitaxel,77 sorafenib,78 vincristine,79 and vorinostat.80 Several antiretrovirals, most notably atazanavir and indinavir are associated with unconjugated hyperbilirubinemia secondary to UGT1A1 inhibition similar to that which occurs in association with Gilbert's syndrome.53,81,82 When assessing liver function in HIV patients on these antiretroviral agents, it is useful to also assess transaminases and alkaline phosphatase. Unconjugated hyperbilirubinemia in association with these agents and in the absence of other evidence of hepatic dysfunction may be ignored in dosing chemotherapeutic agents. On the other hand, didanosine, stavudine, and zidovudine may produce hepatotoxicity associated with lactic acidosis and steatosis.83 Maraviroc has been noted to rarely produce a hepatotoxicity associated with allergic features.84 Such hepatotoxicity should not be ignored and didanosine, maraviroc, stavudine, and zidovudine should be stopped or replaced before initiating cytotoxic chemotherapy with agents that have hepatic metabolism at standard doses but use reduced dosing based on the degree of hepatotoxicity. The NRTIs, abacavir, emtricitabine, lamivudine, and tenofovir, and the NNRTI efavirenz are the less likely to be hepatotoxic and may often be substituted.

Toxicity-related Concerns

Zidovudine is associated with severe neutropenia in ∼8% of patients with advanced AIDS.85 Since traditional cytotoxic chemotherapy regimens are also associated with neutropenia, this combination should be avoided and an alternative NRTI should be prescribed. If zidovudine use cannot be avoided, less myelosuppressive chemotherapy should be administered or the patient monitored closely for neutropenia.

Didanosine and stavudine have been frequently associated with peripheral neuropathy which may be irreversible.86 While the onset is typically weeks to months after initiation of therapy, patients with pre-existing neuropathy may experience this toxicity sooner. Platinums, taxanes, and vinca-alkaloids are the three classes of chemotherapeutics frequently associated with peripheral neuropathy. The first proteasome inhibitor, bortezomib, is associated with a reversible peripheral neuropathy which appears to be a class-effect.87,88 Chemotherapy-induced neuropathy is generally cumulative or dose related, with management consisting of dose-reduction or lower dose-intensity. If an HIV patient develops a malignancy and is on one of the NRTIs listed above, the following options exist: 1) select an alternative chemotherapy regimen without overlapping toxicity; 2) substitute an alternate NRTI or other appropriate antiretroviral; 3) temporarily discontinue antiretroviral therapy.

Atazanavir,89 ritonavir boosted lopinavir,90 and saquinavir91 are associated with QT prolongation. Multiple anticancer agents also are associated with QT prolongation including cytotoxic agents and the newer molecularly targeted anticancer agents such as anthracyclines,92 arsenic trioxide,92 dasatninb,93 lapatinib,94 nilotinib,95 sunitinib,96 and tamoxifen.97 Due to the potential for sudden death, combinations of these agents should be avoided.

Anticancer Drug Interaction Potential

Since many anticancer agents are also metabolized by CYP450, the potential for drug interactions with HAART is high. Anthracyclines, antimetabolite agents, antitumor antibiotics, and platinums undergo non-CYP450 routes of elimination and would be unlikely to be altered by HAART.98,99 Camptothecins undergo non-enzymatic routes of elimination, are substrates but not inhibitors or inducers of CYP450 and UGT isozymes and therefore are likely to be altered by HAART.100 Proteasome inhibitors are substrates but not inhibitors or inducers of CYP450s at clinically relevant drug concentrations.101 Bidirectional drug interactions could be anticipated by other classes of anticancer agents including alkylating agents,102 corticosteroids, epipodophyllotoxins,102 taxanes,102,103 tyrosine kinase inhibitors,104 and vinca alkaloids.102 Antoniou and Tseng recently reviewed the potential drug interactions between antiretroviral and anticancer therapy.105 As recently reviewed by Deeken and colleagues, some molecularly targeted agents may have pharmacokinetic-based drug interactions with HAART.106There is little information available from prospective drug interaction trials and so the review was largely predicted from metabolic fate of the combinations. Several case reports, small series, and clinical trials have suggested antiretroviral drug interactions with several anticancer agents including bexarotene,107 cyclophosphamide,108 docetaxel,109 irinotecan,110 and vinblastine.111 It is possible that newer classes of antiretrovirals, such as the integrase strand transfer inhibitor raltegravir,112 will have reduced interaction potential than NNRTIs and PIs.

When trying to address patient specific cancer regimens in patients with HIV/AIDS on HAART, the oncologist should partner with an infectious diseases specialist to review anticipated changes in the HAART regimen. At the current time, there is no guidance on dose adjustments of either HAART or chemotherapy. This is in part the consequence of HIV patients being excluded from early cancer drug development studies.113 In 2006, the Cancer Therapy Evaluation Program (CTEP) of the National Cancer Institute (NCI) advised that “Individuals known to be HIV-positive should not be arbitrarily excluded from participation in clinical cancer treatment trials,” without scientific justification for exclusion.114 Therefore, it will take several years before guidelines provide scientifically sound recommendations for novel agents still undergoing development.

The dilemma for the clinician becomes how to treat patients who require anticancer drug treatment given the propensity of drug interactions. Since the advent of HAART, HIV has become a chronic disease. There has been no such success with the majority of cancers especially with the deadliest forms such as esophageal, lung, metastatic melanoma, and pancreatic cancers. The maintenance of dose-schedule and dose-intensity are the primary principals which are thought to contribute to cancer cure. In some cases, cancer treatment should take priority over HIV treatment, despite the risk associated with stopping HAART.115,116 However, the oncologist must also recognize that continuous HAART therapy is recommended in order to prevent resistant HIV strains, opportunistic infections, and eventual death.116

When cancer occurs in patients with HIV who are not yet on antiretroviral therapy, many clinicians opt to initiate cancer chemotherapy first, and only to add antiretroviral therapy after side effects (e.g., nausea, vomiting, and mucositis) associated with chemotherapy are adequately managed. This avoids starting and stopping antiretroviral therapy in a way that might engender antiretroviral resistance. For some chemotherapy regimens, notably continuous infusion regimens and high dose/ablative regimens such as are used in the setting of autologous transplant, concerns with possible adverse drug interactions are such that it is routine to stop antiretroviral therapy before initiating such treatment. For example, Little and colleagues were able to successfully treat AIDS–related lymphomas (ARLs) with dose-adjusted EPOCH (etoposide, prednisone, vincristine, cyclophosphamide, and doxorubicin) with suspension of antiretroviral therapy.117 The choice to stop antiretroviral therapy was to ensure that the maximum intensity of the dose-adjusted EPOCH regimen could be achieved while minimizing drug interactions and preserving immune function. The majority (74%) of patients achieved a complete remission with viral loads decreased below baseline by 3 months and CD4+ counts recovered by 12 months after HAART was reinstituted. When EPOCH was studied by ECOG, institutional investigators were allowed to determine whether to administer or hold HAART therapy.118 No clear adverse effects were noted in patients who received combined HAART and EPOCH chemotherapy, and so it would appear that for many patients and many regimens, either approach is allowable. Most clinicians would avoid the combination of zidovudine with any myelosuppressive regimen, many would interrupt HAART for continuous or high dose chemotherapy regimens, and most would search for alternatives to ritonavir-based regimens when combination chemotherapy regimens are being administered since no clear consensus exists.

Future of Antiretroviral Therapy and Targeted Anticancer Treatment

The trend in anticancer drug development is to move from the use of cytotoxic chemotherapy which is indiscriminate to molecularly targeted agents which are more selective at killing cancer cells.119 Since molecularly targeted agents tend to have less myelosuppression and peripheral neuropathy, there may be fewer concerns about overlapping toxicity with HAART. However, the newer anticancer agents are not without toxicity such as QT prolongation or hypertension.

The AIDS Malignancy Consortium, a National Cancer Institute-supported clinical trials group, is starting to address some of these issues by conducting prospective clinical trials with molecularly targeted agents in patients on HAART. Patients are stratified according to HAART regimens: NNRTI-based HAART therapy, efavirenz- based HAART therapy, non-ritonavir-based PI therapy, and ritonavir-based PI therapy. Patients on NNRTI-based, efavirenz- based, or non-ritonavir-based PI therapy commence treatment at the FDA-approved dose of the anticancer drug, while patients on ritonavir-based PI therapy will start at a reduced dose and escalate according to a standard ‘3+3’ dose escalation. Patients on efavirenz-based therapy may be escalated above the FDA-approved dose while cautiously monitoring both drug concentrations and toxicity. As standard of care in HAART regimen change over time8, this stratification schema may shift, but in general will include considerations of drug interaction potential. This design was modeled after organ dysfunction studies conducted in cancer patients.120 A translational approach may be warranted to aid in prioritizing anticancer agents for the next clinical trials based on their propensity to interact with HAART in vitro and in animals models.

Conclusion

Detailed guidelines for dose adjustment based on clinical trials data are generally not available for anticancer and antiretroviral drugs used concurrently. We look forward to a time when the results of prospective clinical trial data will be available to guide clinical decision making. For the time being, clinicians and clinical investigators must be cognizant of the potential for interactions that may be inferred from knowledge of drug metabolism and make judicious treatment decisions. The importance of oncologists and infectious disease specialists partnering in the management of these patients and discussing the particulars of strategies that involve combinations of drugs cannot be overstressed. As patients with HIV live longer, and more develop malignancies whether HIV related or not, a better understanding of cancer chemotherapy and antiretroviral drug interactions will grow in importance.

Acknowledgments

M.A.R., C.F., and R.F.A. are supported in part by National Cancer Institute (NCI) grant U01 CA 121947 to the AIDS Malignancy Consortium which has facilitated interactions related to this review. C.F. is also supported in part by National Institute of Allergy and Infectious Disease (NIAID) grants U01 AI 069465 and AI 068636.

Footnotes

Disclosure: During the preparation of this manuscript and for 3 years prior, C.F. received research grant support from GlaxoSmithKline; served on scientific advisory boards for Boehringer–Ingelheim Inc., Bristol–Myers Squibb, GlaxoSmithKline, Merck Pharmaceuticals, Tibotec Pharmaceuticals, and Virostatics, LLC; was a paid consultant for Inhibitex Inc., and received lecture honoraria from Abbott Laboratories.

Contributions

M.A.R. performed the literature search and interpretation. M.A.R., C.F., and R.F.A. were involved with the concept and design, manuscript writing, and final approval of manuscript.

Search Strategy: Data for this review were identified by searches of Medline with the search terms “antiretroviral agents,” “cancer,” “HAART,” and individual drug names. Additional references were selected from relevant articles. Abstracts and reports from meetings were included only when they related directly to previously published work. Only papers published in English between January, 1990, and January, 2011, were included.

References

  • 1.Palella FJ, Jr, Delaney KM, Moorman AC, Loveless MO, Fuhrer J, Satten GA, et al. Declining morbidity and mortality among patients with advanced human immunodeficiency virus infection. HIV Outpatient Study Investigators. N Engl J Med. 1998;338(13):853–60. doi: 10.1056/NEJM199803263381301. [DOI] [PubMed] [Google Scholar]
  • 2.Lima VD, Hogg RS, Harrigan PR, Moore D, Yip B, Wood E, et al. Continued improvement in survival among HIV-infected individuals with newer forms of highly active antiretroviral therapy. Aids. 2007;21(6):685–92. doi: 10.1097/QAD.0b013e32802ef30c. [DOI] [PubMed] [Google Scholar]
  • 3.Causes of death in HIV-1-infected patients treated with antiretroviral therapy, 1996-2006: collaborative analysis of 13 HIV cohort studies. Clin Infect Dis. 2010;50(10):1387–96. doi: 10.1086/652283. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Simard EP, Pfeiffer RM, Engels EA. Cumulative incidence of cancer among individuals with acquired immunodeficiency syndrome in the United States. Cancer. 2010 Oct 19; doi: 10.1002/cncr.25547. Epub ahead of print. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Seden K, Back D, Khoo S. Antiretroviral drug interactions: often unrecognized, frequently unavoidable, sometimes unmanageable. J Antimicrob Chemother. 2009;64(1):5–8. doi: 10.1093/jac/dkp152. [DOI] [PubMed] [Google Scholar]
  • 6.Piscitelli SC, Gallicano KD. Interactions among drugs for HIV and opportunistic infections. N Engl J Med. 2001;344(13):984–96. doi: 10.1056/NEJM200103293441307. [DOI] [PubMed] [Google Scholar]
  • 7.Flexner C. HIV drug development: the next 25 years. Nat Rev Drug Discov. 2007;6(12):959–66. doi: 10.1038/nrd2336. [DOI] [PubMed] [Google Scholar]
  • 8.Department of Health and Human Services. Panel on Antiretroviral Guidelines for Adults and Adolescents. Guidelines for the use of antiretroviral agents in HIV-1-infected adults and adolescents. [Accessed January 15, 2011];2011 Jan 10;:1–166. Available at http://www.aidsinfo.nih.gov/ContentFiles/AdultandAdolescentGL.pdf.
  • 9.Walsh JS, Reese MJ, Thurmond LM. The metabolic activation of abacavir by human liver cytosol and expressed human alcohol dehydrogenase isozymes. Chem Biol Interact. 2002;142(1-2):135–54. doi: 10.1016/s0009-2797(02)00059-5. [DOI] [PubMed] [Google Scholar]
  • 10.Yuen GJ, Weller S, Pakes GE. A review of the pharmacokinetics of abacavir. Clin Pharmacokinet. 2008;47(6):351–71. doi: 10.2165/00003088-200847060-00001. [DOI] [PubMed] [Google Scholar]
  • 11.McDowell JA, Chittick GE, Ravitch JR, Polk RE, Kerkering TM, Stein DS. Pharmacokinetics of [(14)C]abacavir, a human immunodeficiency virus type 1 (HIV-1) reverse transcriptase inhibitor, administered in a single oral dose to HIV-1-infected adults: a mass balance study. Antimicrob Agents Chemother. 1999;43(12):2855–61. doi: 10.1128/aac.43.12.2855. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Ray AS, Olson L, Fridland A. Role of purine nucleoside phosphorylase in interactions between 2′,3′-dideoxyinosine and allopurinol, ganciclovir, or tenofovir. Antimicrob Agents Chemother. 2004;48(4):1089–95. doi: 10.1128/AAC.48.4.1089-1095.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Zong J, Chittick GE, Wang LH, Hui J, Begley JA, Blum MR. Pharmacokinetic evaluation of emtricitabine in combination with other nucleoside antivirals in healthy volunteers. J Clin Pharmacol. 2007;47(7):877–89. doi: 10.1177/0091270007300808. [DOI] [PubMed] [Google Scholar]
  • 14.Johnson MA, Moore KH, Yuen GJ, Bye A, Pakes GE. Clinical pharmacokinetics of lamivudine. Clin Pharmacokinet. 1999;36(1):41–66. doi: 10.2165/00003088-199936010-00004. [DOI] [PubMed] [Google Scholar]
  • 15.Grasela DM, Stoltz RR, Barry M, Bone M, Mangold B, O'Grady P, et al. Pharmacokinetics of single-dose oral stavudine in subjects with renal impairment and in subjects requiring hemodialysis. Antimicrob Agents Chemother. 2000;44(8):2149–53. doi: 10.1128/aac.44.8.2149-2153.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Court MH, Krishnaswamy S, Hao Q, Duan SX, Patten CJ, Von Moltke LL, et al. Evaluation of 3′-azido-3′-deoxythymidine, morphine, and codeine as probe substrates for UDP-glucuronosyltransferase 2B7 (UGT2B7) in human liver microsomes: specificity and influence of the UGT2B7*2 polymorphism. Drug Metab Dispos. 2003;31(9):1125–33. doi: 10.1124/dmd.31.9.1125. [DOI] [PubMed] [Google Scholar]
  • 17.Blum MR, Liao SH, Good SS, de Miranda P. Pharmacokinetics and bioavailability of zidovudine in humans. Am J Med. 1988;85(2A):189–94. [PubMed] [Google Scholar]
  • 18.VIREAD® (tenofovir disoproxil fumarate) tablets [Package Insert] Foster City, CA: Gilead Sciences Inc; Oct, 2010. [Google Scholar]
  • 19.Voorman RL, Payne NA, Wienkers LC, Hauer MJ, Sanders PE. Interaction of delavirdine with human liver microsomal cytochrome P450: inhibition of CYP2C9, CYP2C19, and CYP2D6. Drug Metab Dispos. 2001;29(1):41–7. [PubMed] [Google Scholar]
  • 20.Voorman RL, Maio SM, Hauer MJ, Sanders PE, Payne NA, Ackland MJ. Metabolism of delavirdine, a human immunodeficiency virus type-1 reverse transcriptase inhibitor, by microsomal cytochrome P450 in humans, rats, and other species: probable involvement of CYP2D6 and CYP3A. Drug Metab Dispos. 1998;26(7):631–9. [PubMed] [Google Scholar]
  • 21.Hariparsad N, Nallani SC, Sane RS, Buckley DJ, Buckley AR, Desai PB. Induction of CYP3A4 by efavirenz in primary human hepatocytes: comparison with rifampin and phenobarbital. J Clin Pharmacol. 2004;44(11):1273–81. doi: 10.1177/0091270004269142. [DOI] [PubMed] [Google Scholar]
  • 22.Robertson SM, Maldarelli F, Natarajan V, Formentini E, Alfaro RM, Penzak SR. Efavirenz induces CYP2B6-mediated hydroxylation of bupropion in healthy subjects. J Acquir Immune Defic Syndr. 2008;49(5):513–9. doi: 10.1097/QAI.0b013e318183a425. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Ward BA, Gorski JC, Jones DR, Hall SD, Flockhart DA, Desta Z. The cytochrome P450 2B6 (CYP2B6) is the main catalyst of efavirenz primary and secondary metabolism: implication for HIV/AIDS therapy and utility of efavirenz as a substrate marker of CYP2B6 catalytic activity. J Pharmacol Exp Ther. 2003;306(1):287–300. doi: 10.1124/jpet.103.049601. [DOI] [PubMed] [Google Scholar]
  • 24.Belanger AS, Caron P, Harvey M, Zimmerman PA, Mehlotra RK, Guillemette C. Glucuronidation of the antiretroviral drug efavirenz by UGT2B7 and an in vitro investigation of drug-drug interaction with zidovudine. Drug Metab Dispos. 2009;37(9):1793–6. doi: 10.1124/dmd.109.027706. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Scholler-Gyure M, Kakuda TN, Raoof A, De Smedt G, Hoetelmans RM. Clinical pharmacokinetics and pharmacodynamics of etravirine. Clin Pharmacokinet. 2009;48(9):561–74. doi: 10.2165/10895940-000000000-00000. [DOI] [PubMed] [Google Scholar]
  • 26.Faucette SR, Zhang TC, Moore R, Sueyoshi T, Omiecinski CJ, LeCluyse EL, et al. Relative activation of human pregnane X receptor versus constitutive androstane receptor defines distinct classes of CYP2B6 and CYP3A4 inducers. J Pharmacol Exp Ther. 2007;320(1):72–80. doi: 10.1124/jpet.106.112136. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Erickson DA, Mather G, Trager WF, Levy RH, Keirns JJ. Characterization of the in vitro biotransformation of the HIV-1 reverse transcriptase inhibitor nevirapine by human hepatic cytochromes P-450. Drug Metab Dispos. 1999;27(12):1488–95. [PubMed] [Google Scholar]
  • 28.Riska P, Lamson M, MacGregor T, Sabo J, Hattox S, Pav J, et al. Disposition and biotransformation of the antiretroviral drug nevirapine in humans. Drug Metab Dispos. 1999;27(8):895–901. [PubMed] [Google Scholar]
  • 29.Granfors MT, Wang JS, Kajosaari LI, Laitila J, Neuvonen PJ, Backman JT. Differential inhibition of cytochrome P450 3A4, 3A5 and 3A7 by five human immunodeficiency virus (HIV) protease inhibitors in vitro. Basic Clin Pharmacol Toxicol. 2006;98(1):79–85. doi: 10.1111/j.1742-7843.2006.pto_249.x. [DOI] [PubMed] [Google Scholar]
  • 30.Decker CJ, Laitinen LM, Bridson GW, Raybuck SA, Tung RD, Chaturvedi PR. Metabolism of amprenavir in liver microsomes: role of CYP3A4 inhibition for drug interactions. J Pharm Sci. 1998;87(7):803–7. doi: 10.1021/js980029p. [DOI] [PubMed] [Google Scholar]
  • 31.Huang L, Wring SA, Woolley JL, Brouwer KR, Serabjit-Singh C, Polli JW. Induction of P-glycoprotein and cytochrome P450 3A by HIV protease inhibitors. Drug Metab Dispos. 2001;29(5):754–60. [PubMed] [Google Scholar]
  • 32.Polli JW, Jarrett JL, Studenberg SD, Humphreys JE, Dennis SW, Brouwer KR, et al. Role of P-glycoprotein on the CNS disposition of amprenavir (141W94), an HIV protease inhibitor. Pharm Res. 1999;16(8):1206–12. doi: 10.1023/a:1018941328702. [DOI] [PubMed] [Google Scholar]
  • 33.Singh R, Chang SY, Taylor LC. In vitro metabolism of a potent HIV-protease inhibitor (141W94) using rat, monkey and human liver S9. Rapid Commun Mass Spectrom. 1996;10(9):1019–26. doi: 10.1002/(SICI)1097-0231(19960715)10:9<1019::AID-RCM618>3.0.CO;2-J. [DOI] [PubMed] [Google Scholar]
  • 34.Rittweger M, Arasteh K. Clinical pharmacokinetics of darunavir. Clin Pharmacokinet. 2007;46(9):739–56. doi: 10.2165/00003088-200746090-00002. [DOI] [PubMed] [Google Scholar]
  • 35.Furfine ES, Baker CT, Hale MR, Reynolds DJ, Salisbury JA, Searle AD, et al. Preclinical pharmacology and pharmacokinetics of GW433908, a water-soluble prodrug of the human immunodeficiency virus protease inhibitor amprenavir. Antimicrob Agents Chemother. 2004;48(3):791–8. doi: 10.1128/AAC.48.3.791-798.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.von Moltke LL, Greenblatt DJ, Grassi JM, Granda BW, Duan SX, Fogelman SM, et al. Protease inhibitors as inhibitors of human cytochromes P450: high risk associated with ritonavir. J Clin Pharmacol. 1998;38(2):106–11. doi: 10.1002/j.1552-4604.1998.tb04398.x. [DOI] [PubMed] [Google Scholar]
  • 37.Zucker SD, Qin X, Rouster SD, Yu F, Green RM, Keshavan P, et al. Mechanism of indinavir-induced hyperbilirubinemia. Proc Natl Acad Sci U S A. 2001;98(22):12671–6. doi: 10.1073/pnas.231140698. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.von Moltke LL, Greenblatt DJ, Duan SX, Daily JP, Harmatz JS, Shader RI. Inhibition of desipramine hydroxylation (Cytochrome P450-2D6) in vitro by quinidine and by viral protease inhibitors: relation to drug interactions in vivo. J Pharm Sci. 1998;87(10):1184–9. doi: 10.1021/js980197h. [DOI] [PubMed] [Google Scholar]
  • 39.Eagling VA, Back DJ, Barry MG. Differential inhibition of cytochrome P450 isoforms by the protease inhibitors, ritonavir, saquinavir and indinavir. Br J Clin Pharmacol. 1997;44(2):190–4. doi: 10.1046/j.1365-2125.1997.00644.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Balani SK, Woolf EJ, Hoagland VL, Sturgill MG, Deutsch PJ, Yeh KC, et al. Disposition of indinavir, a potent HIV-1 protease inhibitor, after an oral dose in humans. Drug Metab Dispos. 1996;24(12):1389–94. [PubMed] [Google Scholar]
  • 41.Ye ZW, Camus S, Augustijns P, Annaert P. Interaction of eight HIV protease inhibitors with the canalicular efflux transporter ABCC2 (MRP2) in sandwich-cultured rat and human hepatocytes. Biopharm Drug Dispos. 2010;31(2-3):178–88. doi: 10.1002/bdd.701. [DOI] [PubMed] [Google Scholar]
  • 42.Kumar GN, Dykstra J, Roberts EM, Jayanti VK, Hickman D, Uchic J, et al. Potent inhibition of the cytochrome P-450 3A-mediated human liver microsomal metabolism of a novel HIV protease inhibitor by ritonavir: A positive drug-drug interaction. Drug Metab Dispos. 1999;27(8):902–8. [PubMed] [Google Scholar]
  • 43.Kumar GN, Jayanti VK, Johnson MK, Uchic J, Thomas S, Lee RD, et al. Metabolism and disposition of the HIV-1 protease inhibitor lopinavir (ABT-378) given in combination with ritonavir in rats, dogs, and humans. Pharm Res. 2004;21(9):1622–30. doi: 10.1023/b:pham.0000041457.64638.8d. [DOI] [PubMed] [Google Scholar]
  • 44.Zhang D, Chando TJ, Everett DW, Patten CJ, Dehal SS, Humphreys WG. In vitro inhibition of UDP glucuronosyltransferases by atazanavir and other HIV protease inhibitors and the relationship of this property to in vivo bilirubin glucuronidation. Drug Metab Dispos. 2005;33(11):1729–39. doi: 10.1124/dmd.105.005447. [DOI] [PubMed] [Google Scholar]
  • 45.Kumar GN, Rodrigues AD, Buko AM, Denissen JF. Cytochrome P450-mediated metabolism of the HIV-1 protease inhibitor ritonavir (ABT-538) in human liver microsomes. J Pharmacol Exp Ther. 1996;277(1):423–31. [PubMed] [Google Scholar]
  • 46.Lim ML, Min SS, Eron JJ, Bertz RJ, Robinson M, Gaedigk A, et al. Coadministration of lopinavir/ritonavir and phenytoin results in two-way drug interaction through cytochrome P-450 induction. J Acquir Immune Defic Syndr. 2004;36(5):1034–40. doi: 10.1097/00126334-200408150-00006. [DOI] [PubMed] [Google Scholar]
  • 47.Kharasch ED, Mitchell D, Coles R, Blanco R. Rapid clinical induction of hepatic cytochrome P4502B6 activity by ritonavir. Antimicrob Agents Chemother. 2008;52(5):1663–9. doi: 10.1128/AAC.01600-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Greenblatt DJ, von Moltke LL, Daily JP, Harmatz JS, Shader RI. Extensive impairment of triazolam and alprazolam clearance by short-term low-dose ritonavir: the clinical dilemma of concurrent inhibition and induction. J Clin Psychopharmacol. 1999;19(4):293–6. doi: 10.1097/00004714-199908000-00001. [DOI] [PubMed] [Google Scholar]
  • 49.Ouellet D, Hsu A, Qian J, Locke CS, Eason CJ, Cavanaugh JH, et al. Effect of ritonavir on the pharmacokinetics of ethinyl oestradiol in healthy female volunteers. Br J Clin Pharmacol. 1998;46(2):111–6. doi: 10.1046/j.1365-2125.1998.00749.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Vourvahis M, Kashuba AD. Mechanisms of pharmacokinetic and pharmacodynamic drug interactions associated with ritonavir-enhanced tipranavir. Pharmacotherapy. 2007;27(6):888–909. doi: 10.1592/phco.27.6.888. [DOI] [PubMed] [Google Scholar]
  • 51.King JR, Acosta EP. Tipranavir: a novel nonpeptidic protease inhibitor of HIV. Clin Pharmacokinet. 2006;45(7):665–82. doi: 10.2165/00003088-200645070-00003. [DOI] [PubMed] [Google Scholar]
  • 52.Mukwaya G, MacGregor T, Hoelscher D, Heming T, Legg D, Kavanaugh K, et al. Interaction of ritonavir-boosted tipranavir with loperamide does not result in loperamide-associated neurologic side effects in healthy volunteers. Antimicrob Agents Chemother. 2005;49(12):4903–10. doi: 10.1128/AAC.49.12.4903-4910.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Lankisch TO, Moebius U, Wehmeier M, Behrens G, Manns MP, Schmidt RE, et al. Gilbert's disease and atazanavir: from phenotype to UDP-glucuronosyltransferase haplotype. Hepatology. 2006;44(5):1324–32. doi: 10.1002/hep.21361. [DOI] [PubMed] [Google Scholar]
  • 54.Colombo S, Buclin T, Franc C, Guignard N, Khonkarly M, Tarr PE, et al. Ritonavir-boosted atazanavir-lopinavir combination: a pharmacokinetic interaction study of total, unbound plasma and cellular exposures. Antivir Ther. 2006;11(1):53–62. [PubMed] [Google Scholar]
  • 55.REYATAZ® (atazanavir sulfate) capsules [Package Insert] Princeton, NJ: Bristol-Myers Squibb; Apr, 2010. [Google Scholar]
  • 56.Friedland G, Andrews L, Schreibman T, Agarwala S, Daley L, Child M, et al. Lack of an effect of atazanavir on steady-state pharmacokinetics of methadone in patients chronically treated for opiate addiction. AIDS. 2005;19(15):1635–41. doi: 10.1097/01.aids.0000183628.20041.f2. [DOI] [PubMed] [Google Scholar]
  • 57.Wu E, Sandoval T, Zhang K, Grettenberger H, Hee B, Lee C, et al. Cytochrome P450 isoforms involved in the metabolism of nelfinavir mesylate, an HIV-1 protease inhibitor. ISSX Proc. 1998;13:110. [Google Scholar]
  • 58.Lillibridge JH, Liang BH, Kerr BM, Webber S, Quart B, Shetty BV, et al. Characterization of the selectivity and mechanism of human cytochrome P450 inhibition by the human immunodeficiency virus-protease inhibitor nelfinavir mesylate. Drug Metab Dispos. 1998;26(7):609–16. [PubMed] [Google Scholar]
  • 59.Dixit V, Hariparsad N, Li F, Desai P, Thummel KE, Unadkat JD. Cytochrome P450 enzymes and transporters induced by anti-human immunodeficiency virus protease inhibitors in human hepatocytes: implications for predicting clinical drug interactions. Drug Metab Dispos. 2007;35(10):1853–9. doi: 10.1124/dmd.107.016089. [DOI] [PubMed] [Google Scholar]
  • 60.Kassahun K, McIntosh I, Cui D, Hreniuk D, Merschman S, Lasseter K, et al. Metabolism and disposition in humans of raltegravir (MK-0518), an anti-AIDS drug targeting the human immunodeficiency virus 1 integrase enzyme. Drug Metab Dispos. 2007;35(9):1657–63. doi: 10.1124/dmd.107.016196. [DOI] [PubMed] [Google Scholar]
  • 61.Ruxrungtham K, Boyd M, Bellibas SE, Zhang X, Dorr A, Kolis S, et al. Lack of interaction between enfuvirtide and ritonavir or ritonavir-boosted saquinavir in HIV-1-infected patients. J Clin Pharmacol. 2004;44(7):793–803. doi: 10.1177/0091270004266489. [DOI] [PubMed] [Google Scholar]
  • 62.Hyland R, Dickins M, Collins C, Jones H, Jones B. Maraviroc: in vitro assessment of drug-drug interaction potential. Br J Clin Pharmacol. 2008;66(4):498–507. doi: 10.1111/j.1365-2125.2008.03198.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Walker DK, Abel S, Comby P, Muirhead GJ, Nedderman AN, Smith DA. Species differences in the disposition of the CCR5 antagonist, UK-427,857, a new potential treatment for HIV. Drug Metab Dispos. 2005;33(4):587–95. doi: 10.1124/dmd.104.002626. [DOI] [PubMed] [Google Scholar]
  • 64.Boffito M, Abel S. A review of the clinical pharmacology of maraviroc. Introduction. Br J Clin Pharmacol. 2008;65(1):1–4. doi: 10.1111/j.1365-2125.2008.03131.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Abel S, van der Ryst E, Rosario MC, Ridgway CE, Medhurst CG, Taylor-Worth RJ, et al. Assessment of the pharmacokinetics, safety and tolerability of maraviroc, a novel CCR5 antagonist, in healthy volunteers. Br J Clin Pharmacol. 2008;65(1):5–18. doi: 10.1111/j.1365-2125.2008.03130.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Mouscadet JF, Arora R, Andre J, Lambry JC, Delelis O, Malet I, et al. HIV-1 IN alternative molecular recognition of DNA induced by raltegravir resistance mutations. J Mol Recognit. 2009 doi: 10.1002/jmr.970. [DOI] [PubMed] [Google Scholar]
  • 67.Cooper DA, Steigbigel RT, Gatell JM, Rockstroh JK, Katlama C, Yeni P, et al. Subgroup and resistance analyses of raltegravir for resistant HIV-1 infection. N Engl J Med. 2008;359(4):355–65. doi: 10.1056/NEJMoa0708978. [DOI] [PubMed] [Google Scholar]
  • 68.Taylor S, Boffito M, Khoo S, Smit E, Back D. Stopping antiretroviral therapy. Aids. 2007;21(13):1673–82. doi: 10.1097/QAD.0b013e3281c61394. [DOI] [PubMed] [Google Scholar]
  • 69.Bousquet L, Pruvost A, Guyot AC, Farinotti R, Mabondzo A. Combination of tenofovir and emtricitabine plus efavirenz: in vitro modulation of ABC transporter and intracellular drug accumulation. Antimicrob Agents Chemother. 2009;53(3):896–902. doi: 10.1128/AAC.00733-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Moss DM, Kwan WS, Liptrott NJ, Smith DL, Siccardi M, Khoo SH, et al. Raltegravir Is a Substrate for SLC22A6: a Putative Mechanism for the Interaction between Raltegravir and Tenofovir. Antimicrob Agents Chemother. 2011;55(2):879–87. doi: 10.1128/AAC.00623-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Knox TA, Oleson L, von Moltke LL, Kaufman RC, Wanke CA, Greenblatt DJ. Ritonavir greatly impairs CYP3A activity in HIV infection with chronic viral hepatitis. J Acquir Immune Defic Syndr. 2008;49(4):358–68. doi: 10.1097/qai.0b013e31818c7efe. [DOI] [PubMed] [Google Scholar]
  • 72.Taxotere (docetaxel_ injection concentrate [Package Insert] Bridgewater, NJ: Sanofi-Aventis U.S. LLC; Apr, 2010. [Google Scholar]
  • 73.ADRIAMYCIN (doxorubicin HCl) [Package Insert] Bedford, OH: Bedford Laboratories; Sep, 2010. [Google Scholar]
  • 74.Joel SP, Shah R, Clark PI, Slevin ML. Predicting etoposide toxicity: relationship to organ function and protein binding. J Clin Oncol. 1996;14(1):257–67. doi: 10.1200/JCO.1996.14.1.257. [DOI] [PubMed] [Google Scholar]
  • 75.Ramanathan RK, Egorin MJ, Takimoto CH, Remick SC, Doroshow JH, LoRusso PA, et al. Phase I and pharmacokinetic study of imatinib mesylate in patients with advanced malignancies and varying degrees of liver dysfunction: a study by the National Cancer Institute Organ Dysfunction Working Group. J Clin Oncol. 2008;26(4):563–9. doi: 10.1200/JCO.2007.11.0304. [DOI] [PubMed] [Google Scholar]
  • 76.Venook AP, Enders Klein C, Fleming G, Hollis D, Leichman CG, Hohl R, et al. A phase I and pharmacokinetic study of irinotecan in patients with hepatic or renal dysfunction or with prior pelvic radiation: CALGB 9863. Ann Oncol. 2003;14(12):1783–90. doi: 10.1093/annonc/mdg493. [DOI] [PubMed] [Google Scholar]
  • 77.Venook AP, Egorin MJ, Rosner GL, Brown TD, Jahan TM, Batist G, et al. Phase I and pharmacokinetic trial of paclitaxel in patients with hepatic dysfunction: Cancer and Leukemia Group B 9264. J Clin Oncol. 1998;16(5):1811–9. doi: 10.1200/JCO.1998.16.5.1811. [DOI] [PubMed] [Google Scholar]
  • 78.Miller AA, Murry DJ, Owzar K, Hollis DR, Kennedy EB, Abou-Alfa G, et al. Phase I and pharmacokinetic study of sorafenib in patients with hepatic or renal dysfunction: CALGB 60301. J Clin Oncol. 2009;27(11):1800–5. doi: 10.1200/JCO.2008.20.0931. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Vincristine sulfate injection, solution [Package Insert] Lake Forest, IL: Hospira Inc; Dec, 2007. [Google Scholar]
  • 80.Ramalingam SS, Kummar S, Sarantopoulos J, Shibata S, LoRusso P, Yerk M, et al. Phase I study of vorinostat in patients with advanced solid tumors and hepatic dysfunction: a National Cancer Institute Organ Dysfunction Working Group study. J Clin Oncol. 2010;28(29):4507–12. doi: 10.1200/JCO.2010.30.2307. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Rotger M, Taffe P, Bleiber G, Gunthard HF, Furrer H, Vernazza P, et al. Gilbert syndrome and the development of antiretroviral therapy-associated hyperbilirubinemia. J Infect Dis. 2005;192(8):1381–6. doi: 10.1086/466531. [DOI] [PubMed] [Google Scholar]
  • 82.Strassburg CP. Pharmacogenetics of Gilbert's syndrome. Pharmacogenomics. 2008;9(6):703–15. doi: 10.2217/14622416.9.6.703. [DOI] [PubMed] [Google Scholar]
  • 83.Walker UA, Bauerle J, Laguno M, Murillas J, Mauss S, Schmutz G, et al. Depletion of mitochondrial DNA in liver under antiretroviral therapy with didanosine, stavudine, or zalcitabine. Hepatology. 2004;39(2):311–7. doi: 10.1002/hep.20074. [DOI] [PubMed] [Google Scholar]
  • 84.Hughes CA, Robinson L, Tseng A, Macarthur RD. New antiretroviral drugs: a review of the efficacy, safety, pharmacokinetics, and resistance profile of tipranavir, darunavir, etravirine, rilpivirine, maraviroc, and raltegravir. Expert Opin Pharmacother. 2009 doi: 10.1517/14656560903176446. [DOI] [PubMed] [Google Scholar]
  • 85.RETROVIR (zidovudine) tablets, capsules, and syrup [Package Insert] Research Triangle Park, NC: GlaxoSmithKline; Jan, 2011. [Google Scholar]
  • 86.Moyle GJ, Sadler M. Peripheral neuropathy with nucleoside antiretrovirals: risk factors, incidence and management. Drug Saf. 1998;19(6):481–94. doi: 10.2165/00002018-199819060-00005. [DOI] [PubMed] [Google Scholar]
  • 87.Richardson PG, Briemberg H, Jagannath S, Wen PY, Barlogie B, Berenson J, et al. Frequency, characteristics, and reversibility of peripheral neuropathy during treatment of advanced multiple myeloma with bortezomib. J Clin Oncol. 2006;24(19):3113–20. doi: 10.1200/JCO.2005.04.7779. [DOI] [PubMed] [Google Scholar]
  • 88.Cavaletti G, Jakubowiak AJ. Peripheral neuropathy during bortezomib treatment of multiple myeloma: a review of recent studies. Leuk Lymphoma. 2010;51(7):1178–87. doi: 10.3109/10428194.2010.483303. [DOI] [PubMed] [Google Scholar]
  • 89.Ly T, Ruiz ME. Prolonged QT interval and torsades de pointes associated with atazanavir therapy. Clin Infect Dis. 2007;44(6):e67–8. doi: 10.1086/511875. [DOI] [PubMed] [Google Scholar]
  • 90.KALETRA® (lopinavir/ritonavir) tablets and oral solution [Package Insert] North Chicago, IL: Abbott Laboratories; Jun, 2010. [Google Scholar]
  • 91.INVIRASE® (saquinavir mesylate) capsules and tablets [Package Insert] South San Francisco, CA: Genetech USA, Inc; Oct, 2010. [Google Scholar]
  • 92.Raschi E, Vasina V, Ursino MG, Boriani G, Martoni A, De Ponti F. Anticancer drugs and cardiotoxicity: Insights and perspectives in the era of targeted therapy. Pharmacol Ther. 2010;125(2):196–218. doi: 10.1016/j.pharmthera.2009.10.002. [DOI] [PubMed] [Google Scholar]
  • 93.Johnson FM, Agrawal S, Burris H, Rosen L, Dhillon N, Hong D, et al. Phase 1 pharmacokinetic and drug-interaction study of dasatinib in patients with advanced solid tumors. Cancer. 2010;116(6):1582–91. doi: 10.1002/cncr.24927. [DOI] [PubMed] [Google Scholar]
  • 94.Lee HA, Kim EJ, Hyun SA, Park SG, Kim KS. Electrophysiological effects of the anti-cancer drug lapatinib on cardiac repolarization. Basic Clin Pharmacol Toxicol. 2010;107(1):614–8. doi: 10.1111/j.1742-7843.2010.00556.x. [DOI] [PubMed] [Google Scholar]
  • 95.Deremer DL, Ustun C, Natarajan K. Nilotinib: a second-generation tyrosine kinase inhibitor for the treatment of chronic myelogenous leukemia. Clin Ther. 2008;30(11):1956–75. doi: 10.1016/j.clinthera.2008.11.014. [DOI] [PubMed] [Google Scholar]
  • 96.Bello CL, Mulay M, Huang X, Patyna S, Dinolfo M, Levine S, et al. Electrocardiographic characterization of the QTc interval in patients with advanced solid tumors: pharmacokinetic- pharmacodynamic evaluation of sunitinib. Clin Cancer Res. 2009;15(22):7045–52. doi: 10.1158/1078-0432.CCR-09-1521. [DOI] [PubMed] [Google Scholar]
  • 97.Liu BA, Juurlink DN. Drugs and the QT interval - caveat doctor. N Engl J Med. 2004;351(11):1053–6. doi: 10.1056/NEJMp048192. [DOI] [PubMed] [Google Scholar]
  • 98.Lennard L. Therapeutic drug monitoring of antimetabolic cytotoxic drugs. Br J Clin Pharmacol. 1999;47(2):131–43. doi: 10.1046/j.1365-2125.1999.00884.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Burger H, Loos WJ, Eechoute K, Verweij J, Mathijssen RH, Wiemer EA. Drug transporters of platinum-based anticancer agents and their clinical significance. Drug Resist Updat. 2011 doi: 10.1016/j.drup.2010.12.002. [DOI] [PubMed] [Google Scholar]
  • 100.Fujita K, Sparreboom A. Pharmacogenetics of irinotecan disposition and toxicity: a review. Curr Clin Pharmacol. 2010;5(3):209–17. doi: 10.2174/157488410791498806. [DOI] [PubMed] [Google Scholar]
  • 101.Schwartz R, Davidson T. Pharmacology, pharmacokinetics, and practical applications of bortezomib. Oncology (Williston Park) 2004;18(14 Suppl 11):14–21. [PubMed] [Google Scholar]
  • 102.van Schaik RH. CYP450 pharmacogenetics for personalizing cancer therapy. Drug Resist Updat. 2008;11(3):77–98. doi: 10.1016/j.drup.2008.03.002. [DOI] [PubMed] [Google Scholar]
  • 103.Nallani SC, Goodwin B, Buckley AR, Buckley DJ, Desai PB. Differences in the induction of cytochrome P450 3A4 by taxane anticancer drugs, docetaxel and paclitaxel, assessed employing primary human hepatocytes. Cancer Chemother Pharmacol. 2004;54(3):219–29. doi: 10.1007/s00280-004-0799-9. [DOI] [PubMed] [Google Scholar]
  • 104.Baker SD, Hu S. Pharmacokinetic considerations for new targeted therapies. Clin Pharmacol Ther. 2009;85(2):208–11. doi: 10.1038/clpt.2008.242. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Antoniou T, Tseng AL. Interactions between antiretrovirals and antineoplastic drug therapy. Clin Pharmacokinet. 2005;44(2):111–45. doi: 10.2165/00003088-200544020-00001. [DOI] [PubMed] [Google Scholar]
  • 106.Deeken JF, Pantanowitz L, Dezube BJ. Targeted therapies to treat non-AIDS-defining cancers in patients with HIV on HAART therapy: treatment considerations and research outlook. Curr Opin Oncol. 2009;21(5):445–54. doi: 10.1097/CCO.0b013e32832f3e04. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Desnoyer A, Kaied FA, Descamps D, Yeni P, Descamps V, Le Beller C, et al. Deleterious pharmacokinetic interaction between bexarotene and efavirenz. AIDS. 2010;24(14):2296–8. doi: 10.1097/QAD.0b013e32833d1243. [DOI] [PubMed] [Google Scholar]
  • 108.Ratner L, Lee J, Tang S, Redden D, Hamzeh F, Herndier B, et al. Chemotherapy for human immunodeficiency virus-associated non-Hodgkin's lymphoma in combination with highly active antiretroviral therapy. J Clin Oncol. 2001;19(8):2171–8. doi: 10.1200/JCO.2001.19.8.2171. [DOI] [PubMed] [Google Scholar]
  • 109.Mir O, Dessard-Diana B, Louet AL, Loulergue P, Viard JP, Langlois A, et al. Severe toxicity related to a pharmacokinetic interaction between docetaxel and ritonavir in HIV-infected patients. Br J Clin Pharmacol. 2010;69(1):99–101. doi: 10.1111/j.1365-2125.2009.03555.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Corona G, Vaccher E, Sandron S, Sartor I, Tirelli U, Innocenti F, et al. Lopinavir-ritonavir dramatically affects the pharmacokinetics of irinotecan in HIV patients with Kaposi's sarcoma. Clin Pharmacol Ther. 2008;83(4):601–6. doi: 10.1038/sj.clpt.6100330. [DOI] [PubMed] [Google Scholar]
  • 111.Kotb R, Vincent I, Dulioust A, Peretti D, Taburet AM, Delfraissy JF, et al. Life-threatening interaction between antiretroviral therapy and vinblastine in HIV-associated multicentric Castleman's disease. Eur J Haematol. 2006;76(3):269–71. doi: 10.1111/j.0902-4441.2005.t01-1-EJH2435.x. [DOI] [PubMed] [Google Scholar]
  • 112.Fulco PP, Hynicka L, Rackley D. Raltegravir-based HAART regimen in a patient with large B-cell lymphoma. Ann Pharmacother. 2010;44(2):377–82. doi: 10.1345/aph.1M370. [DOI] [PubMed] [Google Scholar]
  • 113.Persad GC, Little RF, Grady C. Including persons with HIV infection in cancer clinical trials. J Clin Oncol. 2008;26(7):1027–32. doi: 10.1200/JCO.2007.14.5532. [DOI] [PubMed] [Google Scholar]
  • 114.Bethesda, MD: National Cancer Institute Cancer Therapy Evaluation Program: Guidelines regarding the inclusion of cancer survivors and HIV-positive individuals on clinical trials. Available from : http://ctep.cancer.gov/protocoldevelopment/policies_hiv.htm. Last updated May 29, 2008. [Google Scholar]
  • 115.El-Sadr WM, Lundgren JD, Neaton JD, Gordin F, Abrams D, Arduino RC, et al. CD4+ count-guided interruption of antiretroviral treatment. N Engl J Med. 2006;355(22):2283–96. doi: 10.1056/NEJMoa062360. [DOI] [PubMed] [Google Scholar]
  • 116.El-Sadr WM, Grund B, Neuhaus J, Babiker A, Cohen CJ, Darbyshire J, et al. Risk for opportunistic disease and death after reinitiating continuous antiretroviral therapy in patients with HIV previously receiving episodic therapy: a randomized trial. Ann Intern Med. 2008;149(5):289–99. doi: 10.7326/0003-4819-149-5-200809020-00003. [DOI] [PubMed] [Google Scholar]
  • 117.Little RF, Pittaluga S, Grant N, Steinberg SM, Kavlick MF, Mitsuya H, et al. Highly effective treatment of acquired immunodeficiency syndrome-related lymphoma with dose-adjusted EPOCH: impact of antiretroviral therapy suspension and tumor biology. Blood. 2003;101(12):4653–9. doi: 10.1182/blood-2002-11-3589. [DOI] [PubMed] [Google Scholar]
  • 118.Sparano JA, Lee S, Chen MG, Nazeer T, Einzig A, Ambinder RF, et al. Phase II trial of infusional cyclophosphamide, doxorubicin, and etoposide in patients with HIV-associated non-Hodgkin's lymphoma: an Eastern Cooperative Oncology Group Trial (E1494) J Clin Oncol. 2004;22(8):1491–500. doi: 10.1200/JCO.2004.08.195. [DOI] [PubMed] [Google Scholar]
  • 119.Sparreboom A, Verweij J. Advances in cancer therapeutics. Clin Pharmacol Ther. 2009;85(2):113–7. doi: 10.1038/clpt.2008.259. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Ratain MJ, Miller AA, McLeod HL, Venook AP, Egorin MJ, Schilsky RL. The cancer and leukemia group B pharmacology and experimental therapeutics committee: a historical perspective. Clin Cancer Res. 2006;12(11 Pt 2):3612s–6s. doi: 10.1158/1078-0432.CCR-06-9008. [DOI] [PubMed] [Google Scholar]

RESOURCES