Abstract
Introduction
Complete delineation of the HIV-1 life cycle has resulted in the development of several antiretroviral drugs. Twenty-five therapeutic agents belonging to five different classes are currently available for the treatment of HIV-1 infections. Advent of triple combination antiretroviral therapy has significantly lowered the mortality rate in HIV patients. However, fungal infections still represent major opportunistic diseases in immunocompromised patients worldwide.
Areas covered
Antiretroviral drugs that target enzymes and/or proteins indispensable for viral replication are discussed in this article. Fungal infections, causative organisms, epidemiology and preferred treatment modalities are also outlined. Finally, observed/predicted drug-drug interactions between antiretrovirals and antifungals are summarized along with clinical recommendations.
Expert opinion
Concomitant use of amphotericin B and tenofovir must be closely monitored for renal functioning. Due to relatively weak interactive potential with the CYP450 system, fluconazole is the preferred antifungal drug. High itraconazole doses (> 200 mg/day) are not advised in patients receiving booster protease inhibitor (PI) regimen. Posaconazole is contraindicated in combination with either efavirenz or fosamprenavir. Moreover, voriconazole is contraindicated with high-dose ritonavir-boosted PI. Echino-candins may aid in overcoming the limitations of existing antifungal therapy. An increasing number of documented or predicted drug-drug interactions and therapeutic drug monitoring may aid in the management of HIV-associated opportunistic fungal infections.
Keywords: antiretrovirals, azole antifungals, clinical recommendations, human immunodeficiency virus infection, opportunistic infections, pharmacodynamic, pharmacokinetic, therapeutic drug monitoring
1. Introduction
Over the past decade, AIDS-related morbidity and mortality are significantly lower due to the advent of highly active antiretroviral therapy (HAART). Notable advances in the discovery of HIV biology have resulted in the discovery of several anti-HIV drugs. Currently, 25 antiretroviral agents belonging to five different classes have been approved by the US FDA for the treatment of HIV infections. The drug classes include entry and fusion inhibitors, nucleoside/nucleotide reverse transcriptase inhibitors (NRTIs), non-nucleoside reverse transcriptase inhibitors (NNRTIs), integrase strand transfer inhibitors (INSTIs) and protease inhibitors (PIs). These drugs target different enzymes and/or proteins that are indispensable for viral replication. The guidelines for the use of antiretroviral therapy (ART) in HIV-infected adults include a combination of two NRTIs and one drug from either class: NNRTI, INSTI or boosted PI. The drug combination is based on HIV viral load, patient’s resistance testing, adverse effects, dosing frequency and pill burden, with an ultimate goal of achieving maximal viral suppression [1].
Despite the advances made in ART, opportunistic infections (OIs) resulting from immunosuppression still remains the principal cause of morbidity and mortality in HIV-infected individuals [2]. This is primarily due to three main reasons: i) 20% of HIV-infected persons are unaware of their infection; ii) infected individuals do not take ART due to social or economic factors; and iii) infected individuals who are prescribed ART show poor virological and immunological response due to unfavorable pharmacokinetics and other biological factors [3–5]. A bidirectional relation exists between HIV and OIs. HIV results in immunosuppression that permits the infection of several bacterial, fungal and viral pathogens. Further, these OIs can dramatically increase circulating viral load, accelerating HIV progression and transmission [6,7]. Among the OIs, fungal infections are frequently noticed in HIV-infected individuals. Although the incidence of opportunistic fungal infections has dramatically reduced in developed countries, they still represent a major problem in developing countries [8].
HIV patients are usually prescribed a number of medications, including antiretroviral drugs and antifungals. Drug--drug interactions (DDIs) are commonly observed in these patients, which often necessitate dosage adjustment to prevent potential adverse effects and treatment failure [9]. Based on the mechanism involved, the interactions can either be classified as pharmacodynamic or pharmacokinetic. Pharmacodynamic interactions lead to potentiating or antagonistic drug-response effects. Pharmacokinetic interactions involve the effect of one drug on the absorption, distribution, metabolism and excretion of another drug. Both interactions may result in unanticipated therapeutic outcomes. Hence, it is imperative that clinicians recognize these potential interactions between antiretroviral and antifungal drugs. This review highlights a general overview of antiretroviral and antifungal therapy, with a focus on known DDIs and recommendations for clinical management. Such information may aid in better clinical management of opportunistic fungal infections in HIV patients.
2. Antiretroviral therapy
2.1 HIV-1 life cycle
HIV-1 is a member of the Lentivirus genus of the Retroviridae family. The virus particle consists of two identical RNA copies of the genome. The HIV replication cycle can be divided into an early phase and a late phase. It can be summarized as the following steps: binding and entry, uncoating, reverse transcription, integration, viral protein synthesis, assembly and budding [10–12].
The attachment of virus particle to the host cells is considered as a first step in HIV infection. HIV-1 virus binds specifically to CD4 receptor and seven transmembrane coreceptor (CCR5 or CXCR4) expressing host cells. The process is initiated by binding of viral envelope gp120 spikes to CD4 receptor of host cell. This process triggers a change in gp120 configuration leading to coreceptor (CCR5 or CXCR4) engagement and fusion by viral gp41 protein. Following viral entry, the virus envelope dissociates and uncoating occurs. It facilitates the release of viral genetic material and other enzymes into host cells. The important early event in HIV life cycle is the reverse transcription of viral RNA to double-stranded DNA by virus-encoded reverse transcriptase enzyme. This pre-integration complex is then imported into the host nucleus via the interaction between cellular receptor and viral nuclear localization signal. The viral DNA complex integrates with the host cellular chromosomal DNA with the help of integrase enzyme. This stage is considered as the latent stage of HIV life cycle as the virus particles may remain inactive for several years.
Post integration, the virus enters a late phase of replication. The provirus utilizes various enzymes for transcription of DNA to multiple copies of full-length progeny viral RNA and mRNAs that are translated into various viral proteins. The longer HIV-1 polyproteins are cleaved by HIV protease into individual functional proteins and enzymes. Finally, various structural components are assembled together to produce a new mature HIV virion. These newly formed virions then bud off the host cell via the process of exocytosis and are able to infect other cells.
2.2 Current anti-HIV therapeutics
Comprehensive understanding about HIV-1 life cycle and the necessity to inhibit viral replication has resulted in the identification and development of several classes of anti-HIV-1 agents. Twenty-five therapeutic agents belonging to five different classes are currently available for the treatment of HIV-1 infections [13]. These classes include entry and fusion inhibitors, NRTIs, NNRTIs, INSTIs and PIs. List of drugs belonging to these different classes, their metabolic and transport characteristics and adverse effects have been summarized in Table 1.
Table 1.
List of antiretrovirals approved by US FDA for the treatment of HIV infections.
Antirviral drug |
Metabolic enzyme/transport characteristics |
Adverse reactions | ||
---|---|---|---|---|
Substrate | Inhibitor | Inducer | ||
Entry and fusion inhibitors | ||||
Enfuvirtide (T-20) | × | × | × | Pneumonia, erythema, nodules, pruritus, ecchymosis, pancreatitis, anorexia and sinusitis |
Maraviroc | CYP3A4, P-gp | × | × | Hepatotoxicity, cardiovascular events, severe skin and hypersensitivity reactions |
Nucleoside/nucleotide reverse transcriptase inhibitors | ||||
Abacavir | × | × | × | Hypersensitivity reactions, myocardial infarction, nausea, diarrhea, bronchitis, fat redistribution, lactic acidosis and hepatic steatosis |
Didanosine | × | × | × | Blood, lymphatic, ophthalmic, exocrine gland, gastrointestinal, hepatobiliary, musculoskeletal and metabolic disorder |
Emtricitabine | × | × | × | Immune reconstitution syndrome, gastrointestinal, respiratory, musculoskeletal and central nervous system disorder |
Lamivudine | × | × | × | Headache, nausea, malaise, fatigue, nasal symptoms, diarrhea and cough |
Stavudine | × | × | × | Headache, diarrhea, neuropathy, rash, nausea and vomiting |
Tenofovir disoproxil fumarate | × | × | × | Hepatotoxicity, gastrointestinal disorders, headache, pain, depression and asthenia |
Zidovudine | × | × | × | Malaise, headache, asthenia and gastrointestina disorders |
Non-nucleoside/nucleotide reverse transcriptase inhibitors | ||||
Delavirdine | CYP3A4, CYP2D6 | CYP3A4, CYP2C9, CYP2D6, CYP2C19 | × | Skin rash, digestive, nervous, musculoskeletal and respiratory disorders |
Efavirenz | CYP3A4, CYP2B6 | CYP2C9, CYP2C19, CYP3A4 | CYP3A4, CYP2B6 | Rash, dizziness, nausea, headache, fatigue, insomnia and vomiting |
Etravirine | CYP3A4, CYP2C9, CYP2C19 | CYP2C9, CYP2C19, P-gp | CYP3A4 | Skin and hypersensitivity reactions |
Nevirapine | CYP3A4, CYP2B6 | × | CYP3A4, CYP2B6 | Hepatitis, hepatic failure, Stevens-Johnson syndrome, toxic epidermal necrolysis and hypersensitivity reactions |
Rilpivirine | CYP3A4 | × | × | Hepatic, depressive and gastrointestinal disorders |
Integrase strand transfer inhibitors | ||||
Dolutegravir | UGT1A1, CYP3A4, UGT1A3, UGT1A9, BCRP, P-gp | OCT2 | × | Hypersensitivity reactions, nervous system and gastrointestinal disorders |
Elvitegravir | CYP3A4, UGT1A1, UGT1A3 | × | CYP2C9 | Bronchitis, diarrhea, nausea, upper respiratory tract and urinary tract infections |
Raltegravir | UGT1A1 | × | × | Gastrointestinal and nervous disorders |
Protease inhibitors | ||||
Atazanavir | CYP3A4 | CYP3A4, CYP2C8, UGT1A1 | × | Nausea, jaundice/scleral icterus and rash |
Darunavir | CYP3A4 | × | × | Diarrhea, headache, abdominal pain and rash |
Fosamprenavir | CYP3A4, P-gp | CYP3A4 | CYP3A4, P-gp | Diarrhea, rash, nausea, vomiting, fatigue and headache |
Indinavir | CYP3A4 | CYP3A4, CYP2D6 | × | Hyperbilirubinemia, pain, rash nephrolithiasis/urolithiasis and gastrointestinal disorders |
Lopinavir/ ritonavir | CYP3A4 | CYP3A4 | × | Diarrhea, gastrointestinal, eye, cardiac and hepatobiliary disorders |
Nelfinavir | CYP3A4, CYP2C19 | CYP3A4 | × | Diarrhea, rash, nausea, pain, fever, gastrointestinal disorders and bleeding |
Saquinavir | CYP3A4, P-gp | × | × | Nausea, vomiting, diarrhea, musculoskeletal, endocrine, skin, vascular, renal and respiratory disorders |
Tipranavir | CYP3A4, P-gp | P-gp | P-gp | Diarrhea, nausea, pyrexia, vomiting, fatigue, headache, and abdominal pain |
×: None reported; OCT2: Organic cation transporter 2; P-gp: P-glycoprotein
2.2.1 Entry and fusion inhibitors
Enfuvirtide, a fusion inhibitor, blocks gp41-mediated viral and host cellular membrane fusion. Maraviroc, a CCR5 antagonist, inhibits the ability of viral gp120 to bind to the coreceptor. These molecules have a significant potential for treating HIV infections, particularly in patients resistant to other drugs. However, utilization of these drugs is limited due to the variability of viral envelope protein and involvement of multiple coreceptors in viral entry [14]. The dose of enfuvirtide and maraviroc is 90 and 300 mg twice daily (b.i.d.), respectively. Plasma protein binding of enfuvirtide is reported to be 92%, whereas that of maraviroc is 76%.
2.2.2 Nucleoside/nucleotide reverse transcriptase inhibitors
NRTIs block HIV-1 reverse transcriptase, an enzyme that reverse transcribes viral RNA to cDNA. This class of drugs remains a cornerstone of ART combination therapy. Tenofovir disoproxil fumarate is the only nucleotide adenosine analog of this class, whereas others are nucleoside analogs. Among the nucleosides, abacavir is guanosine; didanosine is an adenosine; lamivudine and emtricitabine are cytidine; and zidovudine and stavudine are thymidine analogs. All NRTIs are inactive when administered and require intracellular phosphorylation to elicit antiretroviral activity. Nucleosides require three steps for phosphorylation as opposed to nucleotides, which require only two steps. The activation pathway is unique for different analogs. For instance, the phosphorylation of thymidine analogs is different from guanosine analogs. Following phosphorylation, a pool of dideoxynucleoside analog triphosphates are formed, which compete with intracellular deoxynucleotide triphosphates for binding to reverse transcriptase enzyme. During reverse transcription, phosphorylated NRTIs are incorporated into synthesized DNA, preventing further addition of nucleotides and terminating viral DNA replication [15,16].
Members of organic anion transporter (OAT) and organic cation transporter (OCT) families have been implicated in the uptake of few NRTIs [17]. With the exception of didanosine, all NRTIs can be administered without food restrictions. Didanosine is preferred to be administered in the fasted state. Most of the NRTIs exhibit a relatively high bioavailability. Emtricitabine and tenofovir have longer plasma half-lives, allowing less frequent dosing. Abacavir, lamivudine and zidovudine are 50, 36 and 34% protein-bound, respectively, whereas the other NRTIs are not highly bound to plasma proteins. Most importantly, NRTIs are neither substrates nor inhibitors of cytochrome P450 (CYP450) enzyme system, lowering the risk for potential DDIs [18,19].
2.2.3 Non-nucleoside reverse transcriptase inhibitors
NNRTIs also act by inhibiting reverse transcriptase enzyme. Unlike NRTIs, this class of drug molecules is active when administered and does not require intracellular phosphorylation. The compounds inhibit reverse transcription by binding to a hydrophobic pocket located in the p66 subunit at a distance of 10 Å from the catalytic site of viral enzyme. This binding induces a spatial conformational change and reduces polymerase activity. Studies using the crystal structures have demonstrated that the binding pocket is created only in the presence of an inhibitor. NNRTIs are non/uncompetitive inhibitors of only HIV-1 reverse transcriptase enzyme and have no activity against other retroviruses. Thus, the selectivity index for this class of drugs is very high [20,21].
NNRTIs are classified into first- and second-generation drugs. Nevirapine, delavirdine and efavirenz are first-generation inhibitors, whereas etravirine and rilpivirine belong to the second generation. First-generation molecules require only single point mutation, whereas second-generation drugs require more than one point mutation to elicit resistance. Although most NNRTIs are administered on a single daily regimen due to their longer half-lives, delavirdine is indicated three times daily. All NNRTIs showed significant protein binding (~ 98%), except for nevirapine, which exhibited 62% binding. NNRTIs are predominantly metabolized by CYP450 enzyme system, including 3A4, 2D6, 2B6, 2C9 and 2C19 isoenzymes. Also they act as inhibitors or inducers of these enzymes, further complicating the potential for significant interactions with other drugs. NNRTIs are safe and well tolerated except for nevirapine, which causes severe hepatotoxicity [22,23].
2.2.4 Integrase strand transfer inhibitors
INSTIs prevent integration of proviral DNA into the host genome catalyzed by viral integrase enzyme. These drugs bind to a specific complex between viral DNA and integrase enzyme, causing displacement of reactive viral DNA and chelation of magnesium cofactors at the active catalytic site. INSTIs are active against multiple retroviruses, including HIV-1 and HIV-2. Raltegravir is the first-generation INSTI, whereas dolutegravir is the second-generation inhibitor used for treating raltegravir-resistant strains. Raltegravir is extremely potent, safe and very well tolerated. However, this drug exhibits intrapatient and interpatient pharmacokinetic variability. Dolutegravir has low variability profile, extended half-life and more favorable resistance profile. Raltegravir is almost 83% protein-bound, whereas dolutegravir exhibits 98% protein binding [24–26]. Elvitegravir was approved by US FDA only in a fixed-dose combination tablet that includes cobicistat, tenofovir and emtricitabine. Since elvitegravir is metabolized primarily by CYP3A4, coadministration with a boosting agent (cobicistat) enhances systemic exposure of elvitegravir and facilitates once-daily dosing [27].
2.2.5 Protease inhibitors
PIs inhibit HIV protease, the enzyme responsible for processing viral gag and gag-pol polyprotein precursors into functional and structural proteins. Although, inhibition of protease does produce viral particles, they are immature and are noninfectious. Saquinavir, nelfinavir, ritonavir, indinavir and fosamprenavir are first-generation PIs and lopinavir, tipranavir, atazanavir and darunavir belong to the second generation. Tipranavir is the only non-peptidic inhibitor and the other PIs are competitive peptidomimetic inhibitors. All the PIs are administered orally and the oral bioavailability ranges from 4% for saquinavir to 70% for nelfinavir. These drugs exhibit extensive binding to plasma proteins (90 – 99%), predominantly to two proteins, albumin and α1-acid glycoprotein. Long-term side effects include visceral fat accumulation (lipodystrophy or fat redistribution syndrome) and metabolic disorders (insulin resistance, hyperlip-idemia, hypertriglyceridemia) [28]. In order to enhance their pharmacokinetic profile, all PIs (except for nelfinavir) are boosted with low-dose ritonavir. Ritonavir, a potent inhibitor of CYP450 enzyme system (mainly CYP3A4) acts as a pharmacokinetic enhancer or boosting agent. This combination increases plasma concentration of coadministered drug, allowing simplified dosage regimen, reduced pill burden and improved patient compliance [29,30].
2.3 Highly active antiretroviral therapy
With the availability of drugs that act at different stages of viral replication, combination therapy incorporating various agents has evolved. A prototypical HAART combination includes two NRTIs as the backbone regimen plus one drug from NNRTI, INSTI or boosted PI (Table 2). These combination regimens have been very effective in suppressing viral replication and progression, by acting at different stages of HIV life cycle. Studies suggest that HAART significantly improved patient’s quality of life and survival rate and lowered viral loads. More importantly, combination regimen minimized development of resistance [31,32]. The goal of combination therapy is to achieve maximal and continuous suppression of HIV replication and improvement in patient’s immunological function. Many trials have indicated that ART should be initiated at an early stage for better treatment of severe OIs [33–35].
Table 2.
Preferred and alternative regimens for ART [1].
ART regimen | Combination of antiretrovirals | ||
---|---|---|---|
NNRTI-based regimen | NNRTI | NRTI | NRTI |
Preferred | Efavirenz | Tenofovir | Emtricitabine |
Alternative | Efavirenz | Abacavir | Lamivudine |
Rilpivirine | Tenofovir | Emtricitabine | |
Rilpivirine | Abacavir | Lamivudine | |
INSTI-based regimen | INSTI | NRTI | NRTI |
Preferred | Raltegravir | Tenofovir | Emtricitabine |
Alternative | Elvitegravir/cobicistat | Tenofovir | Emtricitabine |
Raltegravir | Abacavir | Lamivudine | |
PI-based regimen | PI/r (r: ritonavir boosted) | NRTI | NRTI |
Preferred | Atazanavir/r | Tenofovir | Emtricitabine |
Darunavir/r | Tenofovir | Emtricitabine | |
Alternative | Atazanavir/r | Abacavir | Lamivudine |
Darunavir/r | Abacavir | Lamivudine | |
Fosamprenavir/r | Abacavir | Lamivudine | |
Fosamprenavir/r | Tenofovir | Emtricitabine | |
Lopinavir/r | Abacavir | Lamivudine | |
Lopinavir/r | Tenofovir | Emtricitabine |
ART: Antiretroviral therapy; INSTI: Integrase strand transfer inhibitors; NNRTI: Non-nucleoside reverse transcriptase inhibitor; NRTI: Nucleoside/ nucleotide reverse transcriptase inhibitor
3. Antifungal therapy
3.1 Opportunistic fungal infections
OIs still represent a major complication in HIV-infected individuals. Most of the life-threatening OIs occur when the CD4 count falls < 200 cells/mm3. The most common systemic fungal infections observed in AIDS patients include aspergillosis, coccidioidomycosis, cryptococcosis, esophageal and oropharyngeal candidiasis, histoplasmosis and penicilliosis [36]. Invasive aspergillosis is noticed in advanced stages of HIV infection. Predisposing risk factors for this fungal infection include neutropenia, lung diseases and corticosteroid therapy. The causative fungus, Aspergillus fumigatus, presents respiratory disorders such as necrotizing pneumonia and tracheobronchitis [37]. Coccidioidomycosis is caused by Coccidioides immitis and C. posadasii. This infection is endemic to southwestern USA and several central states. The incidence of coccidioidomycosis has significantly reduced since the advent of HAART. The common syndromes noticed with the infection include focal and diffuse pneumonia, cutaneous disease and meningitis [38]. Cryptococcal infection is a frequent life-threatening OI in both HIV-infected and uninfected patients. Although ART has significantly reduced incidence of cryptococcal meningitis in Western countries, this infection remains the leading cause of morbidity and mortality in HIV-infected patients in sub-Saharan Africa and southeast Asia [39]. The two main species responsible for the infection are Cryptococcus neoformans and C. gattii. The most common clinical indications include subacute meningitis or meningoencephalitis with malaise, headache, fever and visual disturbance and altered mental status. The infection is commonly disseminated to every organ system in the body [40].
Mucocutaneous candidiasis is the most commonly observed fungal infection in the global HIV setting. The causative organism for both esophageal and oropharyngeal candidiasis is Candida albicans. The occurrence of candidiasis is recognized as an indicator of immune suppression. The advent of HAART has caused a significant lowering in the prevalence of candidiasis. Esophageal candidiasis is characterized by retrosternal burning pain and odynophagia, whereas oropharyngeal candidiasis is characterized by painless, creamy white lesions on the buccal surface, palate, oropharyngeal mucosa or tongue [41]. The dimorphic fungus Histoplasma capsulatum is responsible for histoplasmosis. This infection is also endemic and limited to central and south USA. The common symptoms include fever, fatigue, weight loss and hepatosplenomegaly [42]. Penicilliosis, caused by the dimorphic fungus Penicillium marneffei, is endemic to southeast and eastern Asia. Such infection could be fatal if not treated appropriately. The symptoms include fever, anemia, weight loss and generalized skin papules [43]. The salient features (causative organism, epidemiology, clinical manifestations and preferred treatment) of these infections have been summarized in Table 3. The preferred mainstay treatment for the fungal infections includes the administration of amphotericin B, azole antifungals and echinocandins.
Table 3.
Summary of opportunistic fungal infections in HIV-infected individuals [2].
Opportunistic fungal infection |
Causative organism | Epidemiology | Clinical manifestations | Preferred treatment |
---|---|---|---|---|
Aspergillosis |
Aspergillus fumigatus A. flavus A. niger A. terreus |
CD4< 100 cells/mm3 | Necrotizing pneumonia, tracheobronchitis | Voriconazole |
Coccidioidomycosis |
Coccidioides immitis C. posadasii |
CD4 < 250 cells/mm3 | Focal pneumonia, cutaneous disease, meningitis | Fluconazole |
Cryptococcosis |
Cryptococcus neoformans C. gattii |
CD4< 100 cells/mm3 | Subacute meningitis, meningoencephalitis | Amphotericin B, fluconazole |
Esophageal candidiasis | Candida albicans | CD4 < 200 cells/mm3 | Retrosternal burning pain, odynophagia | Fluconazole, itraconazole |
Histoplasmosis | Histoplasma capsulatum | CD4< 150 cells/mm3 | Fever, fatigue, weight loss, hepatosplenomegaly, chest pain and dyspnea | Itraconazole |
Penicilliosis | Penicillium marneffei | CD4< 100 cells/mm3 | Fever, anemia, weight loss, generalized skin papules | Itraconazole |
Oropharyngeal candidiasis | C. albicans | CD4 < 200 cells/mm3 | Lesions on buccal surface, palate, oropharyngeal mucosa, tongue surface | Fluconazole |
3.2 Amphotericin B
Since its approval by the US FDA, amphotericin B is considered as the gold standard for invasive fungal infections. It has antifungal activity against a wide variety of yeasts and molds, including most Candida, Aspergillus, Rhodotorula, Mucor and Rhizopus species. The agent binds to ergosterol in the fungal cell membrane, increases permeability and cell death. Due to its high hydrophobicity and poor absorption through the gastrointestinal tract, it is administered intravenously. Amphotericin B is neither a substrate nor an inhibitor of the CYP450 enzyme system. It is associated with a high occurrence of nephrotoxicity and infusion-related reactions (fever, shaking chills, hypotension, anorexia, nausea, vomiting, headache, and tachypnea) [44]. Despite these side effects, the drug still remains the first line of treatment for severe and life-threatening systemic fungal infections. New amphotericin B formulations, such as liposomal AmB or lipid AmB complexes, minimize the toxic side effects associated with amphotericin B [45].
3.3 Azole antifungals
Azole antifungals have been indicated in the treatment of various topical and systemic fungal infections. These drugs inhibit lanosterol 14-α-demethylase enzyme. This CYP450 enzyme converts lanosterol to ergosterol, a vital component of fungal plasma membrane. Depletion of ergosterol and accumulation of toxic metabolites disrupt the structure and function of cell membrane, preventing the fungal cell growth and replication. Azole antifungal drugs differ in their potency and activity depending on their affinity for 14-α-demethylase enzyme. Azoles can be classified into two groups, namely, imidazoles (e.g., ketoconazole, miconazole and clotrimazole) and triazoles (e.g., fluconazole, itraconazole, posaconazole and voriconazole), based on their structural differences. Both imidazoles and triazoles have five-membered heterocyclic ring. Imidazoles have a two nitrogen azole ring, whereas triazoles have three nitrogen azole ring. Even though they share the same mechanism of action, imidazoles and triazoles differ in their spectrum of activity, pharmacokinetic profiles and toxicities. Triazoles are mainly indicated for the treatment of systemic infections since these compounds are less susceptible to metabolic degradation and exhibit favorable pharmacokinetic profiles than imidazoles. Due to the lack of enzyme specificity and adverse effects associated with systemic therapy, use of imidazoles has been restricted to the treatment of superficial fungal infections [46,47]. Many of the azoles can interact with CYP450 enzyme system, hence raising the possibility of DDIs [48]. The characteristics of few important azole antifungals have been described in the following sections.
3.3.1 Fluconazole
It is the first-generation triazole antifungal active against a majority of Candida species. Fluconazole is marginally lipophilic, highly water-soluble and weakly protein-bound. The compound does not undergo first-pass metabolism and has > 90% bioavailability. Absorption of fluconazole from the gastrointestinal tract is independent of the formulation, gastric acidity or concomitant food intake. This molecule displays low nephrotoxicity (< 2%). Fluconazole is excreted unchanged by the kidneys, with a minor part (10%) being metabolized by CYP3A4. This agent is a potent inhibitor for CYP2C9 and moderate inhibitor for CYP3A4 enzymes system [49].
3.3.2 Itraconazole
Itraconazole is the first marketed orally bioavailable first-generation triazole which is active against both Candida and Aspergillus species. It is the most lipophilic and water-insoluble azole. The compound undergoes extensive hepatic metabolism and is excreted in an inactive form via liver and kidneys. The biotransformation pathways are complex and > 30 metabolites have been reported for itraconazole. This antifungal is highly efficacious because of its primary metabolite hydroxyitraconazole that can reach higher concentrations than the parent compound. The metabolite has considerable antifungal activity relative to parent drug. Itraconazole is a substrate and inhibitor of both P-glycoprotein (P-gp) and CYP3A4 [50].
3.3.3 Ketoconazole
Ketoconazole, an imidazole antifungal, is the first orally available azole antifungal for the treatment of topical and systemic fungal infections. It has been approved by the FDA for the treatment of mucocutaneous candidiasis, oral thrush, candiduria, coccidioidomycosis, histoplasmosis, chromomycosis and paracoccidioidomycosis. It is practically insoluble in water, rendering it unsuitable for intravenous formulation. Oral absorption of ketoconazole varies from 37 to 97% and it exhibits high plasma protein binding. Common side effects include nausea, vomiting and hepatotoxicity. Recently, this regimen is often substituted with more effective triazole drugs, fluconazole and itraconazole [51].
3.3.4 Posaconazole
Posaconazole is a potent second-generation triazole with broad spectrum activity against invasive fungal infections caused by Candida, Aspergillus and Zygomycetes species. It is more active than fluconazole and itraconazole and highly effective in the prophylaxis of fungal infections in immunocompromised patients with hematological complications. Posaconazole is structurally derived from itraconazole by replacement of chlorine with fluorine in the phenyl ring and hydroxylation of the triazolone side chain. Phase II biotransformation via glucuronidation is the primary metabolic pathway. Posaconazole has an oral bioavailability of 8 – 47% and must be administered with fatty food to achieve reasonable serum levels. Unlike itraconazole or ketoconazole, absorption of posaconazole is not significantly influenced by gastric pH. Posaconazole is highly plasma protein bound (> 95%). It is an inhibitor of P-gp and CYP3A4 enzyme. The common adverse effects include gastrointestinal disturbances, hypokalemia and hepatotoxicity [52].
3.3.5 Voriconazole
Voriconazole is a second-generation triazole effective against fluconazole-resistant species. It is available for both oral and intravenous administration. This drug is the first-line treatment for invasive aspergillosis. It is lipophilic and rapidly absorbed with high oral bioavailability. The compound is metabolized in the liver and exhibits nonlinear pharmacokinetics in adults. Voriconazole has a large volume of distribution (4 1/kg) and is ~ 60% plasma protein-bound. Voriconazole is prone to many interactions primarily due to its affinity for multiple CYP enzymes (highest for CYP2C19, followed by CYP2C9 and lower for CYP3A4) [53,54].
3.4 Echinocandins
Echinocandins represent the newest class in the antifungal armamentarium. They are semisynthetic lipopeptides that inhibit the synthesis of β-(l,3)-glucan, an essential fungal cell wall component, via noncompetitive inhibition of β-1,3-d-glucan synthase enzyme [55]. Echinocandins are widely used against most Candida and Aspergillus fungi in which β-(l,3)-glucan is a prominent cell wall component. As the target enzyme is absent in mammalian cell walls, this class of molecules are less toxic compared to other antifungals [56]. All echinocandin formulations are available for intravenous use since they have low oral bioavailability. These drugs have linear or moderately linear kinetics with a terminal half-life of 8 – 13 h facilitating once-a-day dosing. The molecules exhibit high protein binding (84 – 99.8%). Echinocandins are poor substrates, inhibitors or inducers of CYP450 system and do not interact with P-gp. Therefore, any serious DDIs are not anticipated when this class of molecules is coadministered with other drugs [57]. All three echinocandins (anidulafungin, caspofungin and micafungin) are generally well tolerated, although few hematological and gastrointestinal disturbances are observed [58].
3.4.1 Anidulafungin
Several clinical studies have indicated that anidulafungin is an effective antifungal agent used in the treatment of azole-refractory esophageal candidiasis, candidemia and invasive candidiasis. This molecule exhibits potent activity against fluconazole or amphotericin B-resistant Aspergillus and Candida species. Anidulafungin undergoes slow chemical degradation by nonspecific peptidases in the blood rather than being by metabolized in the liver [59]. Population pharmacokinetic analysis has suggested that the clearance of anidulafungin is not altered in presence of substrates, inhibitors or inducers of CYP450 system. Hence, interactions with other antiretro-virals are unlikely to occur and any dosage adjustment is not recommended [60].
3.4.2 Caspofungin
Caspofungin is active against most of Candida and Aspergillus species and does not possess activity against Cryptococcus neoformans. It is approved for the treatment of esophageal or oropharyngeal candidiasis and invasive aspergillosis. Caspofungin exhibits moderate nonlinear pharmacokinetics with a terminal half-life of 9 – 11 h and is extensively protein bound (~ 97%). The compound undergoes metabolism via nonenzymatic peptide hydrolysis and/or N-acetylation into inactive metabolites in the liver [61].
3.4.3 Micafungin
Micafungin (FK463), a water soluble antifungal agent, is active against Candida (albicans and non-albicans) and Aspergillus species but inactive against Zygomycetes and Cryptococcus species. This compound is more active than amphotericin B, caspofungin, fluconazole or itraconazole against most Candida species. Micafungin is characterized by linear pharmacokinetics and a mean terminal half-life of ~ 10 – 17 h. It is highly bound to plasma proteins (> 99%). This compound is metabolized in the liver by arylsulfatase and secondary metabolism by catechol-O-methyltransferase [62,63]. Table 4 summarizes the important antifungals, their metabolic and transport characteristics and adverse effects.
Table 4.
List of important antifungals for the treatment of opportunistic fungal infections
Antifungal drug | Metabolic enzyme/transport characteristics |
Adverse reactions | |
---|---|---|---|
Substrate | Inhibitor | ||
Amphotericin B | × | × | Infusion-related reactions (fever, chills, back pain, hypotension), nephrotoxicity |
Azole antifungals | |||
Fluconazole | CYP3A4(10%), | CYP2C9, CYP3A4, CYP2C19 | Hepatotoxicity, rash, nausea, vomiting, diarrhea, abdominal discomfort, prolongation of the QT interval |
Itraconazole | CYP3A4, P-gp | CYP3A4, P-gp | Hepatotoxicity, congestive heart failure, hypokalemia, rash, nausea, vomiting, diarrhea, abdominal pain |
Ketoconazole | CYP3A4 | CYP3A4, CYP2C9 CYP1A2, UGT, P-gp | Hepatotoxicity, nausea, vomiting, diarrhea, abdominal pain |
Posaconazole | UGT1A4, P-gp | CYP3A4, P-gp | Hepatotoxicity, QTc prolongation, hypokalemia, rash, nausea, vomiting, diarrhea, abdominal pain |
Voriconazole | CYP2C19 (major), CYP3A4, CYP2C9 | CYP3A4, CYP2C9 CYP2C19 | Hepatotoxicity, QTc prolongation, rash, visual disturbances (initial dosing), optic neuritis (> 28 days treatment) |
Echinocandins | |||
Anidulafungin | × | × | Headache, histamine release, rash, liver toxicity |
Caspofungin | × | × | Fever, headache, histamine release, rash, liver toxicity |
Micafungin | × | × | Headache, histamine release, rash, liver toxicity |
×: None reported; P-gp: P-glycoprotein
4. Drug-drug interactions
DDIs are often unavoidable in HIV-infected patients. Along with antiretroviral drug therapy, patients may receive other medications for supportive care, OIs and/or immunomodulation. These multidrug regimens can often alter the pharmacodynamics or pharmacokinetics of another drug, leading to clinically relevant DDIs [64]. Pharmacodynamic interactions result from combined treatment at the site of biological activity, leading to additive, synergistic or antagonistic pharmacological effects, without a net change in drug pharmacokinetic parameters. Additive effects arise when coadministration results in combined pharmacological activity, whereas a synergistic effect leads to an enhanced response greater than the additive effect. Antagonistic effects result in diminished pharmacological activity due to multidrug regimen. Pharmacokinetic drug interactions result from altered absorption, distribution and disposition. DDIs involving absorption or metabolism are most frequently encountered in HIV and OI patients [65,66].
Many membrane transporters, including P-gp, multidrug-resistant proteins, breast cancer-resistance protein (BCRP), OCTs, OATs and organic anion-transporting polypeptides are involved in the absorption and distribution of drug molecules [67]. Interactions involving metabolism can occur if a drug acts as a substrate, inhibitor or inducer of CYP450 enzyme system [68]. Plasma levels of drug molecules metabolized by CYP450 enzymes can be modulated by other inhibitors or inducers. Inhibition of CYP enzymes can either be competitive, noncompetitive or mechanism-based. In most cases, both substrate and inhibitor compete for the same catalytic site on the enzyme. Induction occurs when molecules bind to nuclear hormone receptors (pregnane X receptor and constitutive androstane receptor), activate transcriptional factors and increase translation of metabolic enzymes [69]. Also, a close overlap of tissue distribution and substrate specificity of metabolic enzymes and transporters further enhance the potential interactions of various drug combinations [70,71].
Given the above mechanistic basis, it is very important to assess the nature and magnitude of DDIs, since either type of interaction can result in altered safety and efficacy profile of a drug. A study/report of the potential anticipated interactions may facilitate any dosage adjustment, additional therapeutic monitoring and contraindication in multidrug therapy. The following subsections describe observed or predicted DDIs among different classes of antiretrovirals with important antifungals recommended in the treatment of major OIs. Although all the interactions have been summarized in Table 5, only few of them are described below.
Table 5.
Summary of observed/predicted drug-drug interactions between antiretrovirals and azole antifungals.
Anti retroviral | Antifungal | Interactions | Dosing recommendation/ clinical comments |
Ref. |
---|---|---|---|---|
Entry and fusion inhibitors | ||||
Enfuvirtide | Fluconazole | ↔ | ↔ | [72] |
Itraconazole | ↔ | ↔ | [72] | |
Ketoconazole | ↔ | ↔ | [72] | |
Posaconazole | ↔ | ↔ | [72] | |
Voriconazole | ↔ | ↔ | [72] | |
Maraviroc | Fluconazole | Fluconazole may ↑ maraviroc concentration via CYP3A4 inhibition |
Although dosage adjustment is not necessary maraviroc-related toxicity should be closely monitored |
[78] |
Itraconazole | Itraconazole can ↑ maraviroc concentration via CYP3A4 inhibition |
Maraviroc dose is recommended to be reduced to 150 mg b.i.d. |
[77] | |
Ketoconazole | Maraviroc AUC and Cmax ↑ by 5- and 3.4-fold |
Maraviroc dose is recommended to be reduced to 150 mg b.i.d. |
[77] | |
Posaconazole | Posaconazole may ↑ maraviroc concentration via CYP3A4 inhibition |
lthough dosage adjustment is not necessary maraviroc-related toxicity should be closely monitored |
[78] | |
Voriconazole | Voriconazole may ↑ maraviroc concentration via CYP3A4 inhibition |
Although dosage adjustment is not necessary maraviroc-related toxicity should be closely monitored |
[78] | |
Nucleoside/nucleotide reverse transcriptase inhibitors | ||||
Fluconazole | ↔ | ↔ | [80,81,99–103] | |
Itraconazole | ↔ | ↔ | ||
Ketoconazole | ↔ | ↔ | ||
Posaconazole | ↔ | ↔ | ||
Voriconazole | ↔ | ↔ | ||
Non-nucleoside reverse transcriptase inhibitors | ||||
Delavirdine | Fluconazole | ↔ | ↔ | [82] |
Itraconazole | ↔ | ↔ | [82] | |
Ketoconazole | Delavirdine Cmin ↑ by 50% | ↔ | [82] | |
Posaconazole | ↔ | ↔ | [82] | |
Voriconazole | ↔ | ↔ | [82] | |
Efavirenz | Fluconazole | Efavirenz AUC ↑ by 16%; no change in fluconazole AUC | Dosage adjustment is not necessary |
[83] |
Itraconazole | No change in efavirenz AUC; itraconazole AUC and Cmax ↓. by 39 and 37% |
Coadministration should be avoided. If used concomitantly, therapeutic monitoring of itraconazole is needed |
[84] | |
Ketoconazole | No change in efavirenz AUC; ketoconazole AUC and Cmax ↓ by 72 and 44% |
Coadministration should be avoided |
[86] | |
Posaconazole | No change in efavirenz AUC; posaconazole AUC and Cmax ↓ by 50 and 45% |
Coadministration should be avoided unless benefits outweigh the risks. If used concomitantly therapeutic monitoring of posaconazole is needed |
[85] | |
Voriconazole | Efavirenz AUC and Cmax ↑ by 44 and 38%; voriconazole AUC and Cmax ↓. by 77 and 61 % |
Dosage adjustment is necessary. Reduce efavirenz dose to 300 mg/day and increase voriconazole dose to 400 mg b.i.d. Dosage adjustment is not |
[87] | |
Etravirine | Fluconazole | Etravirine AUC ↑ by 86%; no | [104] | |
Itraconazole | change in fluconazole AUC | necessary; use with caution | [104] | |
Concomitant use may result in ↑ plasma concentration of etravirine and ↓. plasma concentration of itraconazole |
Dose adjustments for itraconazole may be necessary; consider therapeutic monitoring |
|||
Ketoconazole | Concomitant use may result in ↑ plasma concentration of etravirine and ↓. plasma concentration of ketoconazole |
Dose adjustments for ketoconazole may be necessary; consider therapeutic monitoring |
[104] | |
Posaconazole | Posaconazole may ↑ plasma concentration of etravirine |
Dose adjustments for posaconazole may be necessary; consider therapeutic monitoring |
[104] | |
Voriconazole | Etravirine AUC ↑ by 36%; voriconazole AUC ↑ by 14% |
Dosage adjustment is not necessary; use with caution |
[104] | |
Nevirapine | Fluconazole | Nevirapine exposure ↑ by 100%; no change in fluconazole AUC |
Use with caution. Increased risk of nevirapine-associated toxicities |
[105] |
Itraconazole | No change in nevirapine AUC; itraconazole AUC and Cmax ↓. by 61 and 38% |
Consider itraconazole monitoring |
[106] | |
Ketoconazole | Nevirapine AUC ↑ by 15 –20%; ketoconazole AUC and Cmax ↓ by 72 and 44% |
Avoid concomitant use; consider other antiretrovirals or antifungals |
[105] | |
Posaconazole | ↔ | ↔ | [105] | |
Voriconazole | ↔ | ↔ | [105] | |
Rilpivirine | Fluconazole | Fluconazole may ↑ rilpivirine concentration via CYP3A4 inhibition |
Dosage adjustment is not necessary; monitor for breakthrough fungal infections |
[107] |
Itraconazole | Itraconazole may ↑ rilpivirine concentration via CYP3A4 inhibition |
Dosage adjustment is not necessary; monitor for breakthrough fungal infections |
[107] | |
Ketoconazole | Rilpivirine AUC and Cmax ↑ by 49 and 30%; ketoconazole AUC and Cmax ↓. by 24 and 15% |
Dosage adjustment is not necessary; monitor for breakthrough fungal infections |
[107] | |
Posaconazole | Posaconazole may ↑ rilpivirine concentration via CYP3A4 inhibition |
Dosage adjustment is not necessary; monitor for breakthrough fungal infections |
[107] | |
Voriconazole | Voriconazole may ↑ rilpivirine concentration via CYP3A4 inhibition |
Dosage adjustment is not necessary; monitor for breakthrough fungal infections |
[107] | |
Integrase strand transfer inhibitors | ||||
Dolutegravir | Fluconazole | ↔ | ↔ | [88] |
Itraconazole | ↔ | ↔ | [88] | |
Ketoconazole | ↔ | ↔ | [88] | |
Posaconazole | ↔ | ↔ | [88] | |
Voriconazole | ↔ | ↔ | [88] | |
Elvitegravir/ | Fluconazole | ↔ | ↔ | [108] |
cobicistat | Itraconazole | Concomitant use may result in ↑ itraconazole and/or elvitegravir and cobicistat concentrations |
The maximum daily dose of itraconazole should not exceed 200 mg |
[108] |
Ketoconazole | Concomitant use may result in ↑ ketoconazole and/or elvitegravir and cobicistat concentrations |
The maximum daily dose of ketoconazole should not exceed 200 mg |
[108] | |
Posaconazole | ↔ | ↔ | [108] | |
Voriconazole | Concomitant use may result in ↑ voriconazole and/or elvitegravir and cobicistat concentrations |
An assessment of benefit:risk ratio is recommended to justify use of voriconazole with elvitegravir/cobicistat |
[108] | |
Raltegravir | Fluconazole | ↔ | ↔ | [89] |
Itraconazole | ↔ | ↔ | [89] | |
Ketoconazole | ↔ | ↔ | [89] | |
Posaconazole | ↔ | ↔ | [89] | |
Voriconazole | ↔ | ↔ | [89] | |
Protease inhibitors (Pls)(/r represents ritonavir-boosted) | ||||
Atazanavir | Fluconazole | ↔ | ↔ | [109] |
Itraconazole | Concomitant use may result in negligible increase in atazanavir; ↑ concentration of itraconazole. Concomitant use of atazanavir/r may result in ↑ concentration of both PI and itraconazole |
Dosage adjustment is not necessary; monitor for itraconazole-related toxicities |
[109] | |
Ketoconazole | Concomitant use may result in negligible increase in atazanavir; ↑ concentration of ketoconazole. Concomitant use of atazanavir/r may result in ↑ concentration of both PI and ketoconazole |
Dosage adjustment is not necessary; monitor for ketoconazole-related toxicities |
[109] | |
Posaconazole | Concomitant use ↑ AUC of atazanavir by 268%. Concomitant use of atazanavir/r ↑ AUC of atazanavir by 146% |
Dosage adjustment is not necessary; monitor for atazanavir-related toxicities |
[85] | |
Voriconazole | Concomitant use may affect atazanavir concentration. Concomitant use of atazanavir/r in patients with a functional CYP2C19 results in ↓. concentration of atazanavir and voriconazole; patients without a functional CYP2C19 results in ↓ concentration of atazanavir and ↑ voriconazole |
Coadministration of atazanavir/r and voriconazole should be avoided unless benefits outweigh the risks. If used concomitantly, therapeutic monitoring is needed |
[109] | |
Darunavir | Fluconazole | ↔ | ↔ | [110] |
Itraconazole | Concomitant use of darunavir/r and itraconazole may ↑ plasma concentration of darunavir and itraconazole |
While administering with darunavir/r, the daily dose of itraconazole should not exceed 200 mg |
[110] | |
Ketoconazole | Concomitant use ↑ AUC and Cmax of darunavir by 155 and 78%; no change in ketoconazole concentration. Concomitant use of darunavir/r ↑ AUC and Cmax of darunavir by 42 and 21 %; ↑ AUC and Cmax of ketoconazole by 212 and 111 % |
While administering with darunavir/r, the daily dose of ketoconazole should not exceed 200 mg |
[111] | |
Posaconazole | ↔ | ↔ | [110] | |
Voriconazole | Concomitant use of darunavir/r results in ↓. concentration of voriconazole |
Coadministration of darunavir/r and voriconazole should be avoided unless benefits outweigh the risks |
[94] | |
Fosamprenavir | Fluconazole | ↔ | ↔ | [91] |
Itraconazole | Concomitant use may result in ↑ concentration of amprenavir and itraconazole. |
Dosage adjustment is not necessary; monitor for dose- related toxicities. |
[91] | |
Concomitant use of fosamprenavir/r may result in ↑ concentration of both PI and itraconazole |
While administering with fosamprenavir/r, the daily dose of itraconazole should not exceed 200 mg |
|||
Ketoconazole | Concomitant use ↑ AUC and Cmax of amprenavir by 31 and 16%; ↑ AUC and Cmax of ketoconazole by 44 and 19%. |
Dosage adjustment is not necessary; monitor for dose- related toxicities. While administering with fosamprenavir/r, the daily dose of ketoconazole should not exceed 200 mg |
[92] | |
Posaconazole | Concomitant use ↓. AUC and Cmax of posaconazole by 23 and 21% |
Do not coadminister | [93] | |
Voriconazole | Concomitant use may ↑ fosamprenavir and voriconazole concentration via CYP3A4 inhibition Concomitant use of fosamprenavir/r results in ↓ concentration of voriconazole |
Coadministration of fosamprenavir/r and voriconazole should be avoided unless benefits outweigh the risks |
[94] | |
Indinavir | Fluconazole | ↔ | ↔ | [112] |
Itraconazole | Concomitant use may result in ↑ concentration of indinavir |
Dosage adjustment is necessary. Reduce indinavir dose to 600 mg every 8 h |
[112] | |
Ketoconazole | Concomitant use may result in ↑ concentration of indinavir |
Dosage adjustment is necessary. Reduce indinavir dose to 600 mg every 8 h |
[112] | |
Posaconazole Voriconazole | ↔ Concomitant use of indinavir/r results in ↓. concentration of voriconazole |
↔ Coadministration of indinavir/r and voriconazole should be avoided unless benefits outweigh the risks |
[112] [94] | |
Lopinavir/r | Fluconazole | ↔ | ↔ | [95] |
Itraconazole | Concomitant use of lopinavir/r may result in ↑ concentration of itraconazole |
While administering with lopinavir/r, the daily dose of itraconazole should not exceed 200 mg |
[95] | |
Ketoconazole | Concomitant use of lopinavir/r may result in ↑ concentration of ketoconazole |
While administering with lopinavir/r, the daily dose of ketoconazole should not exceed 200 mg |
[95] | |
Posaconazole Voriconazole |
↔ Concomitant use of lopinavir/r results in ↓. concentration of voriconazole |
Coadministration of lopinavir/r and voriconazole should be avoided unless benefits outweigh the risks |
[95] [94] |
|
Nelfinavir | Fluconazole | Concomitant use may result in ↑ concentration of nelfinavir and fluconazole |
Dosage adjustment is not necessary; monitor for dose- related toxicities |
[113] |
Itraconazole | Concomitant use may result in ↑ concentration of nelfinavir and itraconazole |
Dosage adjustment is not necessary; monitor for dose- related toxicities |
[113] | |
Ketoconazole Posaconazole |
Concomitant use ↑ AUC and Cmax of nelfinavir by 35 and 25% |
Dosage adjustment is not necessary; monitor for dose- related toxicities |
[113] [113] |
|
Concomitant use may result in ↑ concentration of nelfinavir and posaconazole |
Dosage adjustment is not necessary; monitor for dose- related toxicities |
|||
Voriconazole | Concomitant use of nelfinavir/r results in ↓. concentration of voriconazole |
Coadministration of nelfinavir/r and voriconazole should be avoided unless benefits outweigh the risks |
[94] | |
Saquinavir | Fluconazole | Concomitant use may result in ↑ concentration of saquinavir and fluconazole |
Dosage adjustment is not necessary; monitor for dose- related toxicities |
[114] |
Itraconazole | Concomitant use ↑ exposure of saquinavir and itraconazole |
Dosage adjustment is not necessary; monitor for dose- related toxicities |
[115,116] | |
Ketoconazole | Concomitant use of saquinavir/r ↑ AUC and Cmax of ketoconazole by 45 and 168% |
While administering with saquinavir/r, the daily dose of ketoconazole should not exceed 200 mg |
[117] | |
Posaconazole | ↔ | ↔ | [114] | |
Voriconazole | Concomitant use of saquinavir/r results in ↓. concentration of voriconazole |
Coadministration of saquinavir/r and voriconazole should be avoided, unless benefits outweigh the risks |
[94] | |
Tipranavir | Fluconazole | Concomitant use ↑ AUC and Cmax of tipranavir by 56 and 46%; no change in fluconazole AUC |
Dosage adjustment is not necessary; however doses > 200 mg/day are not recommended. |
[118] |
Itraconazole | Possible ↑ itraconazole concentration |
The daily dose of itraconazole should not exceed 200 mg |
[119] | |
Ketoconazole | Possible ↑ ketoconazole concentration |
The daily dose of ketoconazole should not exceed 200 mg |
[119] | |
Posaconazole | ↔ | ↔ | [119] | |
Voriconazole | Difficult to predict due to involvement of multiple CYP enzymes |
Difficult to predict due to involvement of multiple CYP enzymes |
[119] |
4.1 Entry and fusion inhibitors
4.1.1 Enfuvirtide (T-20)
Majority of the drug interactions result due to changes in metabolic enzyme or transport characteristics. Enfuvirtide undergoes catabolic metabolism to its constituent amino acids. Due to further recycling of the amino acids, drug interactions appear unlikely. In vitro human microsomal studies have indicated that enfuvirtide is not an inhibitor of CYP450 enzymes. In vivo human metabolism studies have also revealed that enfuvirtide does not affect the metabolism of other coadministered drugs. Based on these data, it is recommended that no dosage adjustments are required when enfuvirtide is coadministered with antifungal drugs [72–75].
4.1.2 Maraviroc
Human liver and recombinant microsomal studies revealed that CYP3A4 is the major metabolizing enzyme involved in N-dealkylation of maraviroc [76]. This drug does not inhibit any of the seven major CYP isoforms (1A2, 2B6, 2C9, 2C19, 2D6 and 3A4) at clinically relevant concentrations. Hence, it is unlikely to alter the plasma concentrations of other coadministered drugs. However, being a substrate of CYP3A4, maraviroc’s metabolism can be modulated by CYP3A4 inhibitors. Open, randomized, placebo-controlled studies were conducted in healthy volunteers for pharmacokinetic assessments of maraviroc alone (100 mg b.i.d.) and in the presence of ketoconazole (400 mg/day). The pharmacokinetic parameters, that is, AUCτ and Cmax of maraviroc alone were reported to be 619 ± 54 ng × h/ml and 155 ± 74 ng/ml. AUCτ and Cmax were elevated by 5- and 3.4-fold, respectively in the presence of ketoconazole. This study recommended that the dose of maraviroc be reduced to almost 50% when coadministered with potent CYP3A4 inhibitors, including ketoconazole and itraconazole [77]. Fluconazole and voriconazole are moderate CYP3A4 inhibitors and the magnitude of their interaction with maraviroc is not reported. Although dosage adjustment is not recommended, maraviroc-related toxicity should be closely monitored [78].
Absorption of maraviroc is assessed in vitro using Caco-2 cell culture monolayers. Maraviroc exhibited poor apical to basolateral permeation, but higher transport from basolateral to apical direction. Transcellular efflux ratio was calculated to be > 10. This ratio was significantly diminished in presence of P-gp inhibitors, suggesting the role of P-gp in transport of maraviroc. Further evidence is provided by assessing the pharmacokinetic parameters in wild-type and P-gp knockout mice (mdr1a/mdr1b knockout). Following oral administration, the knockout mice exhibited threefold higher AUC compared to control mice [79]. Although the data suggests involvement of P-gp in oral absorption of maraviroc, DDIs by this transport mechanism have not been conclusively established.
4.2 Nucleoside/nucleotide reverse transcriptase inhibitors
NRTIs are primarily eliminated by the kidneys. They are neither substrates of P-gp nor CYP3A4. Also, this class of antiretrovirals does not cause any inhibition or induction of the CYP450 system. Hence, any clinically relevant DDIs between NRTIs and antifungals may not occur. However, both tenofovir and amphotericin B can cause nephrotoxicity. Concomitant or sequential administration of these drugs must be closely monitored [80]. Similarly, combination therapy of zidovudine and amphotericin B may result in additive myelo-suppression (anemia, neutropenia). Close therapeutic monitoring of hematological function is recommended [81].
4.3 Non-nucleoside reverse transcriptase inhibitors
4.3.1 Delavirdine
Delavirdine is metabolized via CYP3A4 and inhibits 3A4, 2D6, 2C9 as well as 2C19. However, it does not induce any CYP450 enzymes. The pharmacokinetic parameters of delavirdine were assessed in the presence of potent CYP3A4 inhibitors, fluconazole and ketoconazole. No significant interactions have been observed between this NNRTI and antifungals. However, the Cmin of delavirdine was increased by 50% in presence of ketoconazole. Dosage adjustment of delavirdine in the presence of antifungals is not recommended [82].
4.3.2 Efavirenz
In vitro studies with human liver microsomes have suggested that efavirenz is primarily metabolized by CYP3A4 and CYP2B6 enzymes. Hence, plasma concentration can be altered by substrates, inhibitors or inducers of these enzymes. Also, studies have revealed that efavirenz can inhibit CYP2C9, 2C19 and 3A4. These CYP enzymes can alter plasma concentrations of the coadministered drug. Efavirenz can induce the CYP enzymes, 3A4 and 2B6, resulting in induction of its own metabolism. Likewise, this induction may alter the concentration of coadministered drugs that are metabolized by CYP3A4 and CYP2B6 [83].
When given simultaneously with fluconazole, the Cmax of efavirenz remained unaltered with 16% rise in AUC. However, the pharmacokinetic parameters of fluconazole indicated no change compared to control. These interaction studies suggested that no dosage adjustment is required when efavirenz is given with fluconazole concomitantly. On coadministration with itraconazole, the plasma concentrations of efavirenz remained unaltered. Yet, AUC and Cmax of itraconazole were diminished by 39 and 37%, respectively. Hence, alternative antifungal treatment should be considered in combination with efavirenz [84]. A similar trend was also observed in interaction studies of efavirenz with ketoconazole or posaconazole. AUC and Cmax of efavirenz remained unaltered, whereas that of the antifungal agent reduced significantly. These studies clearly reinforce that coadministration should be avoided unless benefits outweigh the associated risks [85,86]. An open-label, multiple dose study was performed in healthy volunteers to study the interaction between efavirenz and voriconazole. Steady-state pharmacokinetic data revealed that AUC and Cmax of efavirenz were elevated by 44 and 38% respectively, with a significant reduction in voriconazole AUC and Cmax (77 and 61%). This interaction study demonstrates that dosage adjustment is necessary when efavirenz is indicated in combination with voriconazole. The dose of efavirenz should be reduced to 300 mg/day, whereas the dose of voriconazole is raised to 400 mg b.i.d. [87].
4.4 Integrase strand transfer inhibitors
4.4.1 Dolutegravir
Dolutegravir is primarily metabolized by UGT1A1 with minor contribution from CYP3A4 (10 –15%). Since the azole antifungals are moderate to potent inhibitors of CYP3A4, coadministration of dolutegravir with these drugs may elevate dolutegravir plasma concentration. Dolutegravir is a potent inhibitor of OCT-2. Studies have demonstrated that dolutegravir does not inhibit or induce any CYP isoforms. According to this report, dolutegravir is not expected to affect the pharmacokinetics of azole antifungals [88].
4.4.2 Raltegravir
Raltegravir is eliminated via UGTIA1-mediated glucuronidation pathway. This antiretroviral is neither a substrate nor an inhibitor/inducer of CYP enzymes. Therefore, interactions of this INSTI with azole antifungals are unlikely to occur [89].
4.5 Protease inhibitors
4.5.1 Fosamprenavir
Fosamprenavir is a water-soluble phosphate prodrug of the PI amprenavir. Upon oral administration, fosamprenavir is rapidly hydrolyzed to the parent drug prior to reaching the systemic circulation. Amprenavir is metabolized in the liver by CYP3A4 enzyme. Hence coadministration of drugs that induce CYP3A4 may lower plasma concentration of amprenavir and vice versa. Studies also report that amprenavir induces CYP3A4. Also, this PI is a substrate and inducer for the efflux transporter, P-gp [90].
Fosamprenavir is not predicted to have any potential drug interactions with fluconazole. However, when administered simultaneously with itraconazole, the plasma concentrations of both amprenavir and itraconazole are expected to rise. Hence, any dose-related toxicity should be monitored. When fosamprenavir is boosted with ritonavir, the maximum dose of itraconazole should not exceed 200 mg/day [91]. An open-label pharmacokinetic interaction study was performed in 12 healthy volunteers to assess the interaction between amprenavir and ketoconazole. Simultaneous administration elevated AUC and Cmax of amprenavir by 31 and 16%, respectively. Also, AUC and Cmax of ketoconazole were elevated by 44 and 19%, respectively. This study demonstrated that simultaneous administration of amprenavir and ketoconazole results in higher concentrations of both drugs. Clinical observation is recommended for monitoring dose-related toxicities [92].
Pharmacokinetics of fosamprenavir in the presence of posaconazole was assessed in an open-label, randomized interaction study in 20 healthy subjects. When administered along with unboosted fosamprenavir, AUC and Cmax of posaconazole were lowered by 23 and 21%. This report indicated that fosamprenavir and posaconazole should not be coadministered [93]. A combination of fosamprenavir and voriconazole may lead to elevated exposure of both the drugs via dual CYP3A4 inhibition. Therapeutic monitoring of both the drugs is required. Voriconazole is contraindicated in patients on fosamprenavir boosted with low-dose ritonavir. This interaction may occur due to CYP2C19/2C9 induction by ritonavir. As a result, voriconazole plasma concentration can be lower leading to loss of antifungal response [94].
4.5.2 Lopinavir/ritonavir
Lopinavir/ritonavir (LPV/r) is metabolized in the liver by CYP3A4. Therefore, drugs that induce CYP3A4 may lower plasma concentration of lopinavir. Similarly, drugs that inhibit CYP3A4 (e.g., azole antifungal) are expected to elevate lopinavir plasma concentrations. LPV/r can also inhibit CYP3A4, altering the plasma concentration of coadministered drugs. Clinically relevant drug interactions are not predicted to occur between LPV/r and fluconazole. Coadministration of LPV/r and itraconazole/ketoconazole are expected to increase plasma concentration of the antifungal drug. Hence, doses of itraconazole/ketoconazole > 200 mg/day are not recommended with LPV/r. No significant interactions have been reported with the antifungal drug posaconazole [95]. A randomized two-period study was performed in healthy male subjects to assess the steady-state pharmacokinetic and safety profile of ritonavir and voriconazole. Low-dose ritonavir (100 mg b.i.d.) lowered AUC and Cmax of voriconazole by 39 and 24%, respectively. High-dose ritonavir (400 mg b.i.d.) significantly lowered AUC and Cmax of voriconazole by 82 and 66%, respectively. These studies recommend that coadministration of voriconazole with low-dose ritonavir should be prescribed with caution and high-dose ritonavir is contraindicated [96].
5. Conclusion
Introduction of HAART has not only suppressed the viral load in HIV-infected individuals but has also greatly improved their quality of life. However, opportunistic fungal infections still remain the primary cause of morbidity and mortality in these individuals. Multidrug oral regimens are often prone to clinically relevant interactions due to metabolism between CYP450 enzyme systems along with efflux by membrane transporters. It is very crucial that clinicians and healthcare providers be familiar with the possible interactions between various antiretrovirals and antifungals. The mechanistic basis for drug interactions facilitates various clinical management options, including dosage adjustment, contraindication for concomitant use or therapeutic drug monitoring (TDM). In the absence of specific interaction potential data, it is imperative that clinicians familiarize with the pharmacodynamic and pharmacokinetic characteristics of these drugs that assist them in predicting possible interactions and measures to mitigate the associated risks.
6. Expert opinion
The current antifungal arsenal in the treatment of OIs includes amphotericin B, azole antifungals and echinocandins. Amphotericin B is neither a substrate nor an inhibitor of the CYP450 enzymes system. Hence, pharmacokinetic interactions are not expected to occur between this antifungal and other antiretrovirals. However, amphotericin B causes significant nephrotoxicity. Therefore, concomitant use with other nephrotoxic drugs (such as tenofovir) must be closely monitored for renal function. Given its relatively weak interaction potential with CYP450 system, fluconazole is the preferred antifungal drug. It may increase the plasma concentrations of coadministered antiretrovirals moderately via CYP3A4 inhibition. Although dosage adjustments are not required; simultaneous administration of fluconazole and nevirapine must be monitored for nevirapine-associated toxicities. Also, high dose of fluconazole (> 200 mg/day) is not recommended along with tipranavir. Itraconazole is a highly effective therapeutic, since the primary metabolite has considerable antifungal activity compared to the parent drug. One important clinical recommendation is that high itraconazole doses (> 200 mg/day) are not advised in patients receiving boosted PI regimen.
Ketoconazole, the imidazole antifungal, has been mostly replaced by newer triazoles. Not many clinically relevant interactions have been reported with posaconazole, the second-generation triazole antifungal. Due to significant reduction in the plasma concentration of posaconazole, it is contraindicated in combination with efavirenz or fosamprenavir. The dose of voriconazole and efavirenz needs to be adjusted when prescribed simultaneously. The use of voriconazole must be avoided with a low-dose ritonavir-boosted PI, unless benefits outweigh the associated risks. Moreover, voriconazole is contraindicated with high-dose ritonavir-boosted PI. Echinocandins are less prone to DDIs since they are neither substrate of P-gp nor CYP450 enzymes. Therefore, these antifungals may substitute the current treatment options and aid in overcoming the limitations of existing antifungal therapy. These increasing numbers of documented or predicted DDIs may be considered in clinical management of HIV and opportunistic fungal infections. Early diagnosis of fungal infections, standardized fungal susceptibility testing and adjunctive immune therapies may lead to effective management of invasive fungal infections in HIV patients.
Other area of management of DDIs includes TDM. TDM enables modification of dosing regimen in response to their plasma concentrations achieved. TDM can serve as a promising therapeutic modality for optimizing clinical efficacy of a medication, while lowering dose-dependent toxicities. However, significant barriers exist to widespread clinical application of TDM [97,98]. In the next few years, we may witness extensive research and well-designed trials to identify the role of TDM in management of ART. The ultimate goal is to follow a multidisciplinary approach to improve the quality of HIV-infected patients.
Article highlights.
The guidelines for the use of antiretroviral therapy in HIV-infected adults include a combination of two nucleoside/nucleotide reverse transcriptase inhibitors and one drug from either of the classes: non-nucleoside reverse transcriptase inhibitor, integrase strand transfer inhibitor or boosted protease inhibitor (PI).
The most common systemic fungal infections observed in HIV patients include aspergillosis, coccidioidomycosis, cryptococcosis, esophageal and oropharyngeal candidiasis, histoplasmosis and penicilliosis. The preferred treatment regimen for these infections includes the administration of amphotericin B or azole antifungals.
Pharmacodynamic interactions may lead to additive, synergistic or antagonistic pharmacological effects, and pharmacokinetic drug interactions result from altered absorption, distribution or disposition characteristics.
A study/report of the potential interactions can trigger a dosage adjustment, additional therapeutic monitoring and contraindication to concomitant use.
Tenofovir and amphotericin B can cause nephrotoxicity. Concomitant or sequential administration of these drugs must be closely monitored for renal function.
When efavirenz and voriconazole are administered concomitantly, efavirenz exposure is elevated, whereas that of voriconazole is diminished. Hence, the dose of efavirenz should be reduced to 300 mg/day and the dose of voriconazole should be raised to 400 mg b.i.d.
Voriconazole is contraindicated in patients taking high-dose ritonavir-boosted PI. It may be due to the fact that ritonavir induces CYP2C19/2C9 and thereby lowers voriconazole plasma concentration leading to loss of antifungal activity.
Therapeutic drug monitoring can serve as a useful tool for optimizing clinical efficacy of a medication, while lowering dose-dependent toxicities.
This box summarizes key points contained in the article.
Acknowledgments
This work was supported by a National Institutes of Health grant R01AI071199.
Footnotes
Declaration of interest
The authors have no conflict of interest to declare.
Bibliography
Papers of special note have been highlighted as either of interest (•) or of considerable interest (••) to readers.
- 1. HHS Panel on Antiretroviral Guidelines for Adults and Adolescents - A Working Group of the Office of AIDS Research Advisory Council (OARAC) Guidelines for the use of antiretroviral agents in HIV-1-infected adults and adolescents. 2013:A1–A4. Available from: http://aidsinfo.nih.gov/contentfiles/lvguidelines/adultandadolescentgl.pdf. •• An excellent resource for HIV treatment guidelines.
- 2. Centers for Disease Control and Prevention, the National Institutes of Health, and the HIV Medicine Association of the Infectious Diseases Society of America. Guidelines for the prevention and treatment of opportunistic infections in HIV-infected adults and adolescents. 2013 Available from: http://aidsinfo.nih.gov/contentfiles/adult_oi.pdf. •• An excellent resource for management of OIs.
- 3.Centers for Disease Control and Prevention. Vital signs: HIV prevention through care and treatment-United States. MMWR Morb Mortal Wkly Rep. 2011;60(47):618–623. [PubMed] [Google Scholar]
- 4.Perbost I, Malafronte B, Pradier C, et al. In the era of highly active antiretroviral therapy, why are HIV-infected patients still admitted to hospital for an inaugural opportunistic infection? HIV Med. 2005;6(4):232–239. doi: 10.1111/j.1468-1293.2005.00282.x. [DOI] [PubMed] [Google Scholar]
- 5.Seal PS, Jackson DA, Chamot E, et al. Temporal trends in presentation for outpatient HIV medical care 2000–2010: implications for short-term mortality. J Gen Intern Med. 2011;26(7):745–750. doi: 10.1007/s11606-011-1693-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Bentwich Z. Concurrent infections that rise the HIV viral load. J HIV Ther. 2003;8(3):72–75. [PubMed] [Google Scholar]
- 7.Lawn SD, Butera ST, Folks TM. Contribution of immune activation to the pathogenesis and transmission of human immunodeficiency virus type 1 infection. Clin Microbiol Rev. 2001;14(4):753–777. doi: 10.1128/CMR.14.4.753-777.2001. table of contents. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Warnock DW. Trends in the epidemiology of invasive fungal infections. Nihon Ishinkin Gakkai Zasshi. 2007;48(1):1–12. doi: 10.3314/jjmm.48.1. [DOI] [PubMed] [Google Scholar]
- 9.Tseng A, Foisy M. Important drug-drug interactions in HIV-infected persons on antiretroviral therapy: an update on new interactions between HIV and non-HIV drugs. Curr Infect Dis Rep. 2012;14(1):67–82. doi: 10.1007/s11908-011-0229-1. [DOI] [PubMed] [Google Scholar]
- 10.Moss JA. HIV/AIDS review. Radiol Technol. 2013;84(3):247–267. quiz 68–70. [PubMed] [Google Scholar]
- 11.Engelman A, Cherepanov P. The structural biology of HIV-1: mechanistic and therapeutic insights. Nat Rev Microbiol. 2012;10(4):279–290. doi: 10.1038/nrmicro2747. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Fanales-Belasio E, Raimondo M, Suligoi B, Butto S. HIV virology and pathogenetic mechanisms of infection: a brief overview. Ann 1st Super Sanita. 2010;46(1):5–14. doi: 10.4415/ANN_10_01_02. [DOI] [PubMed] [Google Scholar]
- 13. Flexner C. HIV drug development: the next 25 years. Nat Rev Drug Discov. 2007;6(12):959–966. doi: 10.1038/nrd2336. • A very good resource for HIV drugs.
- 14.Haqqani AA, Tilton JC. Entry inhibitors and their use in the treatment of HIV-1 infection. Antiviral Res. 2013;98(2):158–170. doi: 10.1016/j.antiviral.2013.03.017. [DOI] [PubMed] [Google Scholar]
- 15.Bonora S, Calcagno A, Gonzalez de Requena D, et al. Clinical pharmacology of nucleoside and nucleotide reverse transcriptase inhibitors. Infez Med. 2006;14(2):61–70. [PubMed] [Google Scholar]
- 16.Back DJ, Burger DM, Flexner CW, Gerber JG. The pharmacology of antiretroviral nucleoside and nucleotide reverse transcriptase inhibitors: implications for once-daily dosing. J Acquir Immune Defic Syndr. 2005;39(Suppl 1):S1–S23. doi: 10.1097/01.qai.0000168882.67942.3f. quiz S24–5. [DOI] [PubMed] [Google Scholar]
- 17.Varatharajan L, Thomas SA. The transport of anti-HIV drugs across blood-CNS interfaces: summary of current knowledge and recommendations for further research. Antiviral Res. 2009;82(2):A99–A109. doi: 10.1016/j.antiviral.2008.12.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Ruane PJ, Dejesus E. New nucleoside/ nucleotide backbone options: a review of recent studies. J Acquir Immune Defic Syndr. 2004;37(Suppl 1):S21–S29. doi: 10.1097/01.qai.0000137003.25258.76. [DOI] [PubMed] [Google Scholar]
- 19.Piliero PJ. Pharmacokinetic properties of nucleoside/nucleotide reverse transcriptase inhibitors. J Acquir Immune Defic Syndr. 2004;37(Suppl 1):S2–S12. doi: 10.1097/01.qai.0000137001.40505.56. [DOI] [PubMed] [Google Scholar]
- 20.Usach I, Melis V, Peris JE. Non-nucleoside reverse transcriptase inhibitors: a review on pharmacokinetics, pharmacodynamics, safety and tolerability. J Int AIDS Soc. 2013;16(1):1–14. doi: 10.7448/IAS.16.1.18567. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.de Bethune MP. Non-nucleoside reverse transcriptase inhibitors (NNRTIs), their discovery, development, and use in the treatment of HIV-1 infection: a review of the last 20 years (1989–2009) Antiviral Res. 2010;85(1):75–90. doi: 10.1016/j.antiviral.2009.09.008. [DOI] [PubMed] [Google Scholar]
- 22.Ma Q, Okusanya OO, Smith PF, et al. Pharmacokinetic drug interactions with non-nucleoside reverse transcriptase inhibitors. Expert Opin Drug Metab Toxicol. 2005;1(3):473–485. doi: 10.1517/17425255.1.3.473. [DOI] [PubMed] [Google Scholar]
- 23.Smith PF, DiCenzo R, Morse GD. Clinical pharmacokinetics of non-nucleoside reverse transcriptase inhibitors. Clin Pharmacokinet. 2001;40(12):893–905. doi: 10.2165/00003088-200140120-00002. [DOI] [PubMed] [Google Scholar]
- 24.Raffi F, Wainberg MA. Multiple choices for HIV therapy with integrase strand transfer inhibitors. Retrovirology. 2012;9:110. doi: 10.1186/1742-4690-9-110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Malet I, Calvez V, Marcelin AG. The future of integrase inhibitors of HIV-1. Curr Opin Virol. 2012;2(5):580–587. doi: 10.1016/j.coviro.2012.08.005. [DOI] [PubMed] [Google Scholar]
- 26.Adams JL, Greener BN, Kashuba AD. Pharmacology of HIV integrase inhibitors. Curr Opin HIV AIDS. 2012;7(5):390–400. doi: 10.1097/COH.0b013e328356e91c. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Ramanathan S, Mathias AA, German P, Kearney BP. Clinical pharmacokinetic and pharmacodynamic profile of the HIV integrase inhibitor elvitegravir. Clin Pharmacokinet. 2011;50(4):229–244. doi: 10.2165/11584570-000000000-00000. [DOI] [PubMed] [Google Scholar]
- 28.Patel M, Mandava NK, Vadlapatla RK, Mitra AK. Recent patents and emerging therapeutics for HIV infections: a focus on protease inhibitors. Pharm Pat Anal. 2013;2(4):513–538. doi: 10.4155/ppa.13.33. [DOI] [PubMed] [Google Scholar]
- 29.Qiu X, Liu ZP. Recent developments of peptidomimetic HIV-1 protease inhibitors. Curr Med Chem. 2011;18(29):4513–4537. doi: 10.2174/092986711797287566. [DOI] [PubMed] [Google Scholar]
- 30.Pokorna J, Machala L, Rezacova P, Konvalinka J. Current and novel inhibitors of HIV protease. Viruses. 2009;1(3):1209–1239. doi: 10.3390/v1031209. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Thompson MA, Aberg JA, Hoy JF, et al. Antiretroviral treatment of adult HIV infection: 2012 recommendations of the International Antiviral Society-USA panel. JAMA. 2012 Jul;308(4):387–402. doi: 10.1001/jama.2012.7961. •• Up-to-date recommendations.
- 32.Piacenti FJ. An update and review of antiretroviral therapy. Pharmacotherapy. 2006;26(8):1111–1133. doi: 10.1592/phco.26.8.1111. [DOI] [PubMed] [Google Scholar]
- 33.Lawn SD, Torok ME, Wood R. Optimum time to start antiretroviral therapy during HIV-associated opportunistic infections. Curr Opin Infect Dis. 2011;24(1):34–42. doi: 10.1097/QCO.0b013e3283420f76. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Sax PE, Sloan CE, Schackman BR, et al. Early antiretroviral therapy for patients with acute aids-related opportunistic infections: a cost-effectiveness analysis of ACTG A5164. HIV Clin Trials. 2010;11(5):248–259. doi: 10.1310/hct1105-248. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Zolopa A, Andersen J, Powderly W, et al. Early antiretroviral therapy reduces AIDS progression/death in individuals with acute opportunistic infections: a multicenter randomized strategy trial. PLoS One. 2009;4(5):e5575. doi: 10.1371/journal.pone.0005575. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Marty F, Mylonakis E. Antifungal use in HIV infection. Expert Opin Pharmacother. 2002;3(2):91–102. doi: 10.1517/14656566.3.2.91. [DOI] [PubMed] [Google Scholar]
- 37.Pupaibool J, Limper AH. Other HIV-associated pneumonias. Clin Chest Med. 2013;34(2):243–254. doi: 10.1016/j.ccm.2013.01.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Ampel NM. Coccidioidomycosis in persons infected with HIV-1. Ann NY Acad Sci. 2007;1111:336–342. doi: 10.1196/annals.1406.033. [DOI] [PubMed] [Google Scholar]
- 39.Jarvis JN, Harrison TS. HIV-associated cryptococcal meningitis. Aids. 2007;21(16):2119–2129. doi: 10.1097/QAD.0b013e3282a4a64d. [DOI] [PubMed] [Google Scholar]
- 40.Antinori S. New insights into HIV/ AIDS-associated cryptococcosis. ISRN AIDS. 2013;2013 doi: 10.1155/2013/471363. 471363. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Cassone A, Cauda R. Candida and candidiasis in HIV-infected patients: where commensalism, opportunistic behavior and frank pathogenicity lose their borders. AIDS. 2012;26(12):1457–1472. doi: 10.1097/QAD.0b013e3283536ba8. [DOI] [PubMed] [Google Scholar]
- 42.Ramos-e-Silva M, Lima CM, Schechtman RC, et al. Systemic mycoses in immunodepressed patients (AIDS) Clin Dermatol. 2012;30(6):616–627. doi: 10.1016/j.clindermatol.2012.01.008. [DOI] [PubMed] [Google Scholar]
- 43.Chakrabarti A, Slavin MA. Endemic fungal infections in the Asia-Pacific region. Med Mycol. 2011;49(4):337–44. doi: 10.3109/13693786.2010.551426. [DOI] [PubMed] [Google Scholar]
- 44.Klepser M. The value of amphotericin B in the treatment of invasive fungal infections. J Crit Care. 2011;26(2):225. doi: 10.1016/j.jcrc.2010.08.005. e1–10. [DOI] [PubMed] [Google Scholar]
- 45.Povoa P, Goncalves-Pereira J, Salluh JI. Amphotericin B in severe fungal infections: a critical reappraisal of the evidence. Am J Respir Crit Care Med. 2013;188(8):1032. doi: 10.1164/rccm.201304-0769LE. [DOI] [PubMed] [Google Scholar]
- 46.Vanden Bossche H, Engelen M, Rochette F. Antifungal agents of use in animal health-chemical, biochemical and pharmacological aspects. J Vet Pharmacol Ther. 2003;26(1):5–29. doi: 10.1046/j.1365-2885.2003.00456.x. [DOI] [PubMed] [Google Scholar]
- 47.Abuhammour W, Habte-Gabr E. Systemic antifungal agents. Indian J Pediatr. 2001;68(7):655–668. doi: 10.1007/BF02752281. [DOI] [PubMed] [Google Scholar]
- 48.Lass-Florl C. Triazole antifungal agents in invasive fungal infections: a comparative review. Drugs. 2011;71(18):2405–2419. doi: 10.2165/11596540-000000000-00000. [DOI] [PubMed] [Google Scholar]
- 49.Diflucan (Fluconazole) Package Insert. New York, NY: Pfizer, Inc; 2013. [Google Scholar]
- 50.Sporanox (Itraconazole) Package Insert. Titusville. NJ: Janssen Pharmaceuticals, Inc; 2012. [Google Scholar]
- 51.Scheinfeld N. Ketoconazole: a review of a workhorse antifungal molecule with a focus on new foam and gel formulations. Drugs Today (Bare) 2008;44(5):369–380. doi: 10.1358/dot.2008.44.5.1216598. [DOI] [PubMed] [Google Scholar]
- 52.Stevens DA. Advances in systemic antifungal therapy. Clin Dermatol. 2012;30(6):657–661. doi: 10.1016/j.clindermatol.2011.09.018. [DOI] [PubMed] [Google Scholar]
- 53.Mikulska M, Novelli A, Aversa F, et al. Voriconazole in clinical practice. J Chemother. 2012;24(6):311–327. doi: 10.1179/1973947812Y.0000000051. [DOI] [PubMed] [Google Scholar]
- 54.Vfend (Voriconazole) Package Insert. New York, NY: Pfizer, Inc; 2011. [Google Scholar]
- 55.Walker LA, Gow NA, Munro CA. Fungal echinocandin resistance. Fungal Genet Biol. 2010;47(2):117–126. doi: 10.1016/j.fgb.2009.09.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Denning DW. Echinocandin antifungal drugs. Lancet. 2003;362(9390):1142–1151. doi: 10.1016/S0140-6736(03)14472-8. [DOI] [PubMed] [Google Scholar]
- 57.Kauffman CA, Carver PL. Update on echinocandin antifungals. Semin Respir Crit Care Med. 2008;29(2):211–219. doi: 10.1055/s-2008-1063859. [DOI] [PubMed] [Google Scholar]
- 58.Eschenauer G, Depestel DD, Carver PL. Comparison of echinocandin antifungals. Ther Clin Risk Manag. 2007;3(1):71–97. doi: 10.2147/tcrm.2007.3.1.71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Vazquez JA, Sobel JD. Anidulafungin: a novel echinocandin. Clin Infect Dis. 2006;43(2):215–222. doi: 10.1086/505204. [DOI] [PubMed] [Google Scholar]
- 60.Dowell JA, Knebel W, Ludden T, et al. Population pharmacokinetic analysis of anidulafungin, an echinocandin antifungal. J Clin Pharmacol. 2004;44(6):590–598. doi: 10.1177/0091270004265644. [DOI] [PubMed] [Google Scholar]
- 61.McCormack PL, Perry CM. Caspofungin: a review of its use in the treatment of fungal infections. Drugs. 2005;65(14):2049–2068. doi: 10.2165/00003495-200565140-00009. [DOI] [PubMed] [Google Scholar]
- 62.Chandrasekar PH, Sobel JD. Micafungin: a new echinocandin. Clin Infect Dis. 2006;42(8):1171–1178. doi: 10.1086/501020. [DOI] [PubMed] [Google Scholar]
- 63.Jarvis B, Figgitt DP, Scott LJ. Micafungin. Drugs. 2004;64(9):969–982. doi: 10.2165/00003495-200464090-00004. discussion 83–4. [DOI] [PubMed] [Google Scholar]
- 64.Josephson F. Drug-drug interactions in the treatment of HIV infection: focus on pharmacokinetic enhancement through CYP3A inhibition. J Intern Med. 2010;268(6):530–539. doi: 10.1111/j.1365-2796.2010.02301.x. [DOI] [PubMed] [Google Scholar]
- 65. Pal D, Kwatra D, Minocha M, et al. Efflux transporters- and cytochrome P-450-mediated interactions between drugs of abuse and antiretrovirals. Life Sci. 2011;88(21–22):959–971. doi: 10.1016/j.lfs.2010.09.012. • An excellent review of interactions between drugs of abuse and antiretrovirals.
- 66. Pal D, Mitra AK. MDR- and CYP3A4-mediated drug-drug interactions. J Neuroimmune Pharmacol. 2006;1(3):323–339. doi: 10.1007/s11481-006-9034-2. • An excellent review on DDIs.
- 67.Giacomini KM, Huang SM, Tweedie DJ, et al. Membrane transporters in drug development. Nat Rev Drug Discov. 2010;9(3):215–236. doi: 10.1038/nrd3028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Pelkonen O, Turpeinen M, Hakkola J, et al. Inhibition and induction of human cytochrome P450 enzymes: current status. Arch Toxicol. 2008;82(10):667–715. doi: 10.1007/s00204-008-0332-8. [DOI] [PubMed] [Google Scholar]
- 69.Jana S, Paliwal J. Molecular mechanisms of cytochrome p450 induction: potential for drug-drug interactions. Curr Protein Pept Sci. 2007;8(6):619–628. doi: 10.2174/138920307783018668. [DOI] [PubMed] [Google Scholar]
- 70.Vadlapatla RK, Vadlapudi AD, Pal D, Mitra AK. Mechanisms of drug resistance in cancer chemotherapy: coordinated role and regulation of efflux transporters and metabolizing enzymes. Curr Pharm Des. 2013;19(40):7126–7140. doi: 10.2174/13816128113199990493. [DOI] [PubMed] [Google Scholar]
- 71. 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. •• A very good resource for reported interactions between antiretrovirals and other dugs.
- 72.Fuzeon (Enfuvirtide) Package Insert. South San Francisco, CA: Genentech USA, Inc; 2013. [Google Scholar]
- 73.Joly V, Jidar K, Tatay M, Yeni P. Enfuvirtide: from basic investigations to current clinical use. Expert Opin Pharmacother. 2010;11(16):2701–2713. doi: 10.1517/14656566.2010.522178. [DOI] [PubMed] [Google Scholar]
- 74.Makinson A, Reynes J. The fusion inhibitor enfuvirtide in recent antiretroviral strategies. Curr Opin HIV AIDS. 2009;4(2):150–158. doi: 10.1097/COH.0b013e32832498d8. [DOI] [PubMed] [Google Scholar]
- 75.Patel IH, Zhang X, Nieforth K, et al. Pharmacokinetics, pharmacodynamics and drug interaction potential of enfuvirtide. Clin Pharmacokinet. 2005;44(2):175–186. doi: 10.2165/00003088-200544020-00003. [DOI] [PubMed] [Google Scholar]
- 76.Hyland R, Dickins M, Collins C, et al. 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]
- 77.Abel S, Russell D, Taylor-Worth RJ, et al. Effects of CYP3A4 inhibitors on the pharmacokinetics of maraviroc in healthy volunteers. Br J Clin Pharmacol. 2008;65(Suppl 1):27–37. doi: 10.1111/j.1365-2125.2008.03133.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Selzentry (Maraviroc) Package Insert. Research Triangle Park. NC: ViiV Healthcare; 2013. [Google Scholar]
- 79.Walker DK, Abel S, Comby P, et al. Species differences in the disposition of the CCR5 antagonist, UK-427,857, a new potential treatment for HIV. Drug Metab Dispos. 2005;33(4):587–595. doi: 10.1124/dmd.104.002626. [DOI] [PubMed] [Google Scholar]
- 80.Viread (Tenofovir disoproxil fumarate) Package Insert. Foster City, CA: Gilead Sciences, Inc; 2013. [Google Scholar]
- 81.Retrovir (Zidovudine) Package Insert. Research Triangle Park, NC: Glaxo SmithKline; 2013. [Google Scholar]
- 82.Rescriptor (Delavirdine mesylate) Packge Insert. Research Triangle Park, NC: ViiV Healthcare; 2012. [Google Scholar]
- 83.Sustiva (Efavirenz) Package Insert. Princeton, NJ: Bristol-Myers Squibb; 2013. [Google Scholar]
- 84.Koo HL, Hamill RJ, Andrade RA. Drug-drug interaction between itraconazole and efavirenz in a patient with AIDS and disseminated histoplasmosis. Clin Infect Dis. 2007;45(6):e77–e79. doi: 10.1086/520978. [DOI] [PubMed] [Google Scholar]
- 85.Krishna G, Moton A, Ma L, et al. Effects of oral posaconazole on the pharmacokinetics of atazanavir alone and with ritonavir or with efavirenz in healthy adult volunteers. J Acquir Immune Defic Syndr. 2009;51(4):437–444. doi: 10.1097/qai.0b013e3181acb51b. [DOI] [PubMed] [Google Scholar]
- 86.Sriwiriyajan S, Mahatthanatrakul W, Ridtitid W, Jaruratanasirikul S. Effect of efavirenz on the pharmacokinetics of ketoconazole in HIV-infected patients. Eur J Clin Pharmacol. 2007;63(5):479–483. doi: 10.1007/s00228-007-0282-8. [DOI] [PubMed] [Google Scholar]
- 87.Damle B, LaBadie R, Crownover P, Glue P. Pharmacokinetic interactions of efavirenz and voriconazole in healthy volunteers. Br J Clin Pharmacol. 2008;65(4):523–530. doi: 10.1111/j.1365-2125.2007.03085.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Tivicay (Dolutegravir) Package Insert. Research Triangle Park, NC: GlaxoSmithKline; 2013. [Google Scholar]
- 89.Isentress (Raltegravir) Package Insert. Whitehouse Station. NJ: Merck & Co., Inc; 2013. [Google Scholar]
- 90.Chapman TM, Plosker GL, Perry CM. Fosamprenavir: a review of its use in the management of antiretroviral therapy-naive patients with HIV infection. Drugs. 2004;64(18):2101–2124. doi: 10.2165/00003495-200464180-00014. [DOI] [PubMed] [Google Scholar]
- 91.Lexiva (Fosamprenavir calcium) Package Insert. Research Triangle Park, NC: GlaxoSmithKline; 2013. [Google Scholar]
- 92.Polk RE, Crouch MA, Israel DS, et al. Pharmacokinetic interaction between ketoconazole and amprenavir after single doses in healthy men. Pharmacotherapy. 1999;19(12):1378–1384. doi: 10.1592/phco.19.18.1378.30905. [DOI] [PubMed] [Google Scholar]
- 93.Bruggemann RJ, van Luin M, Colbers EP, et al. Effect of posaconazole on the pharmacokinetics of fosamprenavir and vice versa in healthy volunteers. J Antimicrob Chemother. 2010;65(10):2188–2194. doi: 10.1093/jac/dkq280. [DOI] [PubMed] [Google Scholar]
- 94.Norvir (Ritonavir) Package Insert. North Chicago, IL: Abb Vie, Inc; 2013. [Google Scholar]
- 95.Kaletra (Lopinavir/Ritonavir) Package Insert. North Chicago, IL: AbbVie, Inc; 2013. [Google Scholar]
- 96.Liu P, Foster G, Gandelman K, et al. Steady-state pharmacokinetic and safety profiles of voriconazole and ritonavir in healthy male subjects. Antimicrob Agents Chemother. 2007;51(10):3617–3626. doi: 10.1128/AAC.00526-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Kredo T, Van der Walt JS, Siegfried N, Cohen K. Therapeutic drug monitoring of antiretrovirals for people with HIV. Cochrane Database Syst Rev. 2009;(3) doi: 10.1002/14651858.CD007268.pub2. CD007268. [DOI] [PubMed] [Google Scholar]
- 98.Gerber JG, Acosta EP. Therapeutic drug monitoring in the treatment of HIV-infection. J Clin Virol. 2003;27(2):117–128. doi: 10.1016/s1386-6532(03)00033-7. [DOI] [PubMed] [Google Scholar]
- 99.Ziagen (Abacavir sulfate) Package Insert. Research Triangle Park, NC: GlaxoSmithKline; 2013. [Google Scholar]
- 100.Videx EC (Didanosine) Package Insert. Princeton, NJ: Bristol-Myers Squibb; 2012. [Google Scholar]
- 101.Emtriva (Emtricitabine) Package Insert. Foster City, CA: Gilead Sciences, Inc; 2012. [Google Scholar]
- 102.Epivir (Lamivudine) Package Insert. Research Triangle Park, NC: GlaxoSmithKline; 2013. [Google Scholar]
- 103.Zerit (Stavudine) Package Insert. Princeton, NJ: Bristol-Myers Squibb; 2012. [Google Scholar]
- 104.Intelence (Etravirine) Package Insert. Titusville. NJ: Janssen Pharmaceuticals, Inc; 2013. [Google Scholar]
- 105.Viramune (Nevirapine) Package Insert. Ridgefield, CT: Boehringer Ingelheim Pharmaceuticals. Inc; 2013. [Google Scholar]
- 106.Jaruratanasirikul S, Sriwiriyajan S. Pharmacokinetic study of the interaction between itraconazole and nevirapine. Eur J Clin Pharmacol. 2007;63(5):451–456. doi: 10.1007/s00228-007-0280-x. [DOI] [PubMed] [Google Scholar]
- 107.Edurant (Rilpivirine) Package Insert. Titusville, NJ: Janssen Therapeutics; 2013. [Google Scholar]
- 108.Stribild (Elvitegravir/Cobicistat/ Emtricitabine/Tenofovir Disoproxil Fumarate) Package Insert. Foster City, CA: Gilead Sciences, Inc; 2013. [Google Scholar]
- 109.Reyataz (Atazanavir sulfate) Package Insert. Princeton. NJ: Bristol-Myers Squibb; 2013. [Google Scholar]
- 110.Prezista (Darunavir) Package Insert. Titusville. NJ: Janssen Pharmaceuticals, Inc; 2013. [Google Scholar]
- 111.Sekar VJ, Lefebvre E, De Pauw M, et al. Pharmacokinetics of darunavir/ritonavir and ketoconazole following coadministration in HIV-healthy volunteers. Br J Clin Pharmacol. 2008;66(2):215–221. doi: 10.1111/j.1365-2125.2008.03191.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Crixivan (Indinavir sulfate) Package Insert. Whitehouse Station, NJ: Merck & Co., Inc; 2012. [Google Scholar]
- 113.Viracept (Nelfinavir mesylate) Package Insert. Lajolla, CA: Agouron Pharmaceuticals, Inc; 2013. [Google Scholar]
- 114.Invirase (Saquinavir mesylate) Package Insert. South San Francisco, CA: Genentech USA, Inc; 2012. [Google Scholar]
- 115.Koks CH, van Heeswijk RP, Veldkamp Al, et al. Itraconazole as an alternative for ritonavir liquid formulation when combined with saquinavir. AIDS. 2000;14(1):89–90. doi: 10.1097/00002030-200001070-00011. [DOI] [PubMed] [Google Scholar]
- 116.MacKenzie-Wood AR, Whitfeld MJ, Ray JE. Itraconazole and HIV protease inhibitors: an important interaction. Med J Aust. 1999;170(1):46–47. doi: 10.5694/j.1326-5377.1999.tb126872.x. [DOI] [PubMed] [Google Scholar]
- 117.Kaeser B, Zandt H, Bour F, et al. Drug-drug interaction study of ketoconazole and ritonavir-boosted saquinavir. Antimicrob Agents Chemother. 2009;53(2):609–614. doi: 10.1128/AAC.00769-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.la Porte CJ, Sabo JP, Elgadi M, Cameron DW. Interaction studies of tipranavir-ritonavir with clarithromycin, fluconazole, and rifabutin in healthy volunteers. Antimicrob Agents Chemother. 2009;53(1):162–173. doi: 10.1128/AAC.00534-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Aptivus (Tipranavir) Package Insert. Ridgefield, CT: Boehringer Ingelheim Pharmaceuticals. Inc; 2012. [Google Scholar]