Your activity: 2 p.v.

Overview of antiretroviral agents used to treat HIV

Overview of antiretroviral agents used to treat HIV
Author:
Courtney V Fletcher, PharmD
Section Editor:
Paul E Sax, MD
Deputy Editor:
Jennifer Mitty, MD, MPH
Literature review current through: Dec 2022. | This topic last updated: Apr 06, 2022.

INTRODUCTION — Antiretroviral therapy (ART) regimens used to treat individuals with HIV have led to dramatic reductions in morbidity and mortality. Drug selection could potentially include at least 25 antiretroviral medications available in six major classes (table 1). For most individuals, an ART regimen consists of a dual nucleoside combination plus a third agent from a different class.

This topic will provide an overview of the different antiretroviral agents used to treat HIV. Topic reviews that discuss the safety of these agents in pregnancy, when to initiate therapy, how to choose a regimen, and antiretroviral resistance are presented elsewhere:

(See "When to initiate antiretroviral therapy in persons with HIV".)

(See "Selecting antiretroviral regimens for treatment-naïve persons with HIV-1: General approach".)

(See "Antiretroviral selection and management in pregnant women with HIV in resource-rich settings".)

(See "Safety and dosing of antiretroviral medications in pregnancy".)

(See "Interpretation of HIV drug resistance testing".)

(See "Evaluation of the treatment-experienced patient failing HIV therapy".)

(See "Selecting an antiretroviral regimen for treatment-experienced patients with HIV who are failing therapy".)

OVERVIEW OF HIV REPLICATION — The life cycle of HIV can be broken down into 6 steps: (1) entry (binding and fusion), (2) reverse transcription, (3) integration, (4) replication (transcription and translation), (5) assembly, and (6) budding and maturation. The identification and understanding of these processes have provided the basis for antiretroviral drug discovery.

Entry – The process of entry involves the binding of HIV to receptors on the surface of the CD4 cell. Once initial binding occurs, the intimate association of HIV with the cell is enhanced by further binding to chemokine coreceptors, which promotes membrane fusion and internalization of the viral genetic material and enzymes necessary for replication. Maraviroc, enfuvirtide, and fostemsavir are antiretroviral agents that inhibit binding and fusion, respectively. However, these agents are not commonly used for the treatment of HIV infection. (See 'Entry inhibitors' below.)

Reverse transcription – The genetic material of HIV is RNA, and the virus uses reverse transcriptase (one of three virally encoded enzymes: reverse transcriptase, integrase, and protease) to convert its RNA into DNA. Following reverse transcription, the double-stranded viral DNA product migrates into the nucleus of the cell. Nucleoside reverse transcriptase inhibitors (NRTIs) and non-nucleoside reverse transcriptase inhibitors (NNRTIs) inhibit the process of reverse transcription. There are several agents in each of these classes that are widely used for the treatment of HIV. (See 'Nucleoside/nucleotide reverse transcriptase inhibitors (NRTIs)' below and 'Non-nucleoside reverse transcriptase inhibitors (NNRTIs)' below.)

Integration – The virus uses the enzyme integrase to integrate (insert) its viral DNA into the DNA of the host CD4 cell. Once viral DNA has been integrated into the cell's DNA, the cell is infected for the remainder of its life. Integrase strand transfer inhibitors (INSTIs) inhibit the process of integration. In many countries, INSTIs are considered the preferred third agent (in combination with two nucleoside analogues) for treatment-naïve individuals. (See 'Integrase strand transfer inhibitors (INSTIs)' below.)

Replication – After integration, HIV preferentially replicates in activated cells. Proviral DNA is formed by transcription using the machinery of the host CD4 cell to create new viral RNA. This viral RNA is translated into long polyprotein chains, which will become the protein and enzyme components of new viral particles. There are no antiretroviral agents that inhibit this step of the replication cycle.

Assembly – The new viral proteins and enzymes move to the cell's outer membrane where they assemble into an immature, noninfectious HIV particle or bud. There are no antiretroviral agents that inhibit this step of the replication cycle.

Budding and maturation – The virus bud is released from the host CD4 cell. The viral enzyme, protease, cuts the long HIV polyprotein chains into smaller functional HIV proteins to make a mature, infectious viral particle. Protease inhibitors are antiretroviral agents that inhibit the HIV protease enzyme, and therefore, prevent this final step in the replication cycle. (See 'Protease inhibitors (PIs)' below.)

ENTRY INHIBITORS

CCR5 antagonists — HIV enters CD4 cells via the CD4 receptor in conjunction with one of its coreceptors: the chemokine coreceptor 5 (CCR5) or the CXC chemokine coreceptor 4 (CXCR4). Viruses that use the CCR5 coreceptor are called "R5 viruses," whereas others that use the CXCR4 coreceptor are called "X4 viruses."

Agents that block CCR5 exert their antiviral activity against HIV by blocking entry of CCR5-tropic viruses into the CD4 T cell. Maraviroc is the only approved CCR5 antagonist. Maraviroc is not commonly used for initial treatment of HIV. However, it may have a role in the treatment-experienced patients with drug-resistant virus. (See "Selecting an antiretroviral regimen for treatment-experienced patients with HIV who are failing therapy", section on 'Patients who have failed multiple regimens'.)

If maraviroc is being considered, a tropism assay is essential prior to initiation of therapy. This assay determines whether a response from a CCR5 antagonist should be expected since maraviroc is not active against X4 or dual-mixed tropic viruses. (See "Overview of HIV drug resistance testing assays", section on 'Tropism assays'.)

Maraviroc is eliminated by both renal and hepatic pathways. The dose should be adjusted in persons with a creatinine clearance <30 mL/min or on dialysis. Increased maraviroc concentrations are expected in persons with significant hepatic dysfunction, though there are no specific guidelines for dose adjustment.

In all settings, the dose of maraviroc must be adjusted when given with concomitant antiretrovirals (or other drugs) that are CYP3A enzyme inducers (eg, efavirenz) or inhibitors (eg, ritonavir). Refer to the Lexicomp drug interactions program within UpToDate for more specific information.

Fusion inhibitors — Fusion inhibitors bind to the envelope glycoprotein 41 (gp41) of HIV to prevent viral fusion to the CD4 T cell. The only approved fusion inhibitor is enfuvirtide (sometimes referred to as T-20), a 36 amino acid peptide. This is an injectable agent that is effective in treatment-experienced patients who have not been exposed to this medication. However, enfuvirtide is difficult to administer for long periods of time due to the need for twice-daily injections, which often lead to local cutaneous reactions. (See "Selecting an antiretroviral regimen for treatment-experienced patients with HIV who are failing therapy", section on 'Fusion inhibitors'.)

Attachment inhibitors — Fostemsavir is an attachment inhibitor that was approved for use in the United States in July 2020 for patients with HIV-1 infection [1]. Fostemsavir is a prodrug, which is converted to the active metabolite temsavir. Temsavir binds directly to the HIV-1 envelope glycoprotein gp120 and prevents viral attachment and subsequent entry of virus into host T cells.

Fostemsavir is dosed as 600 mg twice daily and should be used in combination with other active antiretroviral agents for treatment-experienced patients who are failing therapy and have limited treatment options [1]. It should not be used in patients taking strong cytochrome P450 (CYP)3A inducers (eg, rifampin, carbamazepine, phenytoin, St John's wort), as they decrease the level of temsavir. The most common adverse reaction to fostemsavir is nausea. Other adverse reactions include elevations in liver enzymes in those with hepatitis B or C infection, as well as QTc prolongation.

The efficacy of fostemsavir was demonstrated in a randomized trial of 272 treatment-experienced patients failing therapy, in which the addition of fostemsavir to a regimen with at least one active agent resulted in a greater reduction of HIV RNA after eight days of treatment compared with a regimen that did not include fostemsavir (0.79 versus 0.17 log10 copies, respectively) [2]. In this trial, open-label fostemsavir was given to the whole study population after the initial eight days of treatment, and 54 percent achieved an HIV viral load <40 copies/mL at 48 weeks. By 96 weeks, 60 percent of participants had a suppressed viral load [3]. In a separate cohort of 99 patients who had no active drugs but had fostemsavir added to an optimized antiretroviral therapy regimen, 38 percent had an HIV viral load <40 copies/mL at 48 weeks [2].

NUCLEOSIDE/NUCLEOTIDE REVERSE TRANSCRIPTASE INHIBITORS (NRTIs)

NRTI class characteristics — The nucleoside/nucleotide reverse transcriptase inhibitor (NRTI) class includes (listed alphabetically) (table 1):

Abacavir sulfate, a guanosine analog. (See 'Abacavir' below.)

Emtricitabine and lamivudine, which are cytosine analogs. (See 'Emtricitabine and lamivudine' below.)

Tenofovir alafenamide and tenofovir disoproxil fumarate, which are adenosine-derived NRTIs. (See 'Tenofovir' below.)

Zidovudine, a thymidine analog. (See 'Zidovudine' below.)

Other NRTI agents (eg, didanosine and stavudine) are rarely used because of adverse reactions including neuropathy, lipodystrophy, and mitochondrial toxicity.

NRTIs are usually given in pairs. The commonly used combinations are available as coformulations (eg, tenofovir alafenamide-emtricitabine [Descovy], tenofovir disoproxil fumarate-emtricitabine [Truvada], abacavir-lamivudine [Epzicom]) [4,5]. However, two-drug regimens that include an integrase strand transfer inhibitor (INSTI) or a boosted protease inhibitor (PI) in combination with lamivudine can be used in select patients. (See "Selecting antiretroviral regimens for treatment-naïve persons with HIV-1: General approach", section on 'Use of two-drug regimens in select patients' and "Switching antiretroviral therapy for adults with HIV-1 and a suppressed viral load".)

Mechanism of action – NRTIs undergo intracellular phosphorylation mediated by several host enzymes (eg, cytoplasmic or mitochondrial kinases and phosphotransferases). The active triphosphate form inhibits viral replication through competitive binding to the viral enzyme, reverse transcriptase. DNA chain elongation is terminated after the NRTI triphosphate is incorporated [6,7]. (See 'Overview of HIV replication' above.)

Spectrum of activity – The spectrum of activity of NRTIs includes HIV-1 and HIV-2 viruses.

Lamivudine, emtricitabine, and tenofovir (tenofovir disoproxil fumarate and tenofovir alafenamide) also have activity against hepatitis B virus (HBV). However, lamivudine and emtricitabine should not be used as monotherapy for the treatment of HBV given the high risk of developing HBV resistance with prolonged use. HBV flares can be seen if agents active against HBV are discontinued. A discussion of the treatment of HBV in HIV-infected patients is found elsewhere. (See "Treatment of chronic hepatitis B in patients with HIV".)

Resistance – All of the NRTIs can select for resistance mutations. Detailed discussions of HIV drug resistance are found elsewhere. (See "Interpretation of HIV drug resistance testing", section on 'Nucleoside reverse transcriptase inhibitors' and "Selecting an antiretroviral regimen for treatment-experienced patients with HIV who are failing therapy", section on 'Patients with drug-resistant virus'.)

Adverse events – The hallmark toxicity of the NRTI class is mitochondrial toxicity, which may manifest as peripheral neuropathy, pancreatitis, lipoatrophy, and/or hepatic steatosis [8-10]. Although all NRTIs have "black box" warnings in their product labeling because of the possibility of lactic acidosis syndrome, which is potentially fatal, the risk of mitochondrial toxicity is low with the commonly used agents (abacavir, emtricitabine, lamivudine, tenofovir alafenamide, tenofovir disoproxil fumarate). (See "Mitochondrial toxicity of HIV nucleoside reverse transcriptase inhibitors".)

Discussions of adverse reactions seen with specific agents are found below. (See 'Abacavir' below and 'Tenofovir' below and 'Zidovudine' below.)

Drug interactions – NRTIs have few clinically significant drug-drug interactions because they are not substrates, inhibitors, or inducers of hepatic cytochrome P450 (CYP) enzymes. However, certain important interactions do exist. As an example, tenofovir alafenamide should generally be avoided if the patient is receiving a rifamycin (eg, for the treatment of tuberculosis) since rifamycins reduce the level of tenofovir alafenamide [4]. Additional information on drug interactions can be found in the Lexicomp drug interactions program within UpToDate.

Specific agents (listed alphabetically)

Abacavir — Abacavir is administered as 300 mg twice daily or as 600 mg once daily. It is also available as part of several coformulated tablets, including abacavir-lamivudine, which is a commonly used NRTI combination, and dolutegravir-abacavir-lamivudine (table 1). Prior to initiating treatment with abacavir, all patients must be screened for HLA-B*5701. Abacavir is contraindicated in persons who test positive for HLA-B*5701, as they are at high risk for developing an abacavir hypersensitivity reaction. (See "Abacavir hypersensitivity reaction".)

Although abacavir-lamivudine is considered a first-line NRTI combination, there are concerns about its use in patients with certain comorbidities, as well as those with high viral loads (unless dolutegravir is used as the third agent).

Heart disease Abacavir should be generally avoided in individuals with coronary artery disease (CAD) as well as those with multiple risk factors for CAD. Although there is no consensus on whether there is a causal association between abacavir and the risk of myocardial infarction, data suggest that abacavir may be associated with an increased risk of hyperlipidemia and cardiovascular events [11-15]. Additionally, in one trial, more favorable changes in the total and low-density lipoprotein (LDL) cholesterol levels were observed among patients who switched from an abacavir-lamivudine to a tenofovir disoproxil fumarate-emtricitabine-containing NRTI combination [16]. However, other studies have found no association or only a weak association between abacavir and the risk for myocardial infarction [13,17-19]. Additional information on the use of abacavir in patients with, or at risk for, cardiovascular disease is presented in separate topic reviews. (See "Epidemiology of cardiovascular disease and risk factors in patients with HIV", section on 'Abacavir' and "Selecting antiretroviral regimens for treatment-naive persons with HIV-1: Patients with comorbid conditions", section on 'Cardiovascular disease'.)

Viral load The use of abacavir-lamivudine in patients with a high viral load depends upon which third agent is used. As an example, the use of a regimen containing abacavir-lamivudine plus dolutegravir in patients with high viral loads is supported by a randomized, placebo-controlled trial of 833 participants treated with abacavir-lamivudine-dolutegravir versus efavirenz-tenofovir disoproxil fumarate-lamivudine [20]. The abacavir-lamivudine-dolutegravir arm had a significantly greater percentage of participants achieve viral suppression to <50 copies/mL (88 versus 81 percent), and higher CD4 counts (267 versus 208 cells/microL), even among individuals with a baseline HIV RNA >100,000 copies/mL.

In contrast, there are conflicting data about the efficacy of abacavir-lamivudine in patients with high HIV RNA levels when dolutegravir is not the third agent [11,21,22]. Clinical trial data indicate that patients receiving a nucleoside combination of abacavir-lamivudine are less likely to achieve virologic suppression compared with those receiving tenofovir disoproxil fumarate-emtricitabine when the third drug is efavirenz or ritonavir-boosted atazanavir [11]. There are limited data with the use of other third agents in combination with abacavir-lamivudine.

Emtricitabine and lamivudine — Emtricitabine and lamivudine are both dideoxycytidine analogues, which have very similar pharmacokinetic and pharmacodynamic characteristics, and are considered interchangeable. Structurally, emtricitabine is very similar to lamivudine, only differing in the addition of a fluorine to emtricitabine. Lamivudine and emtricitabine should not be combined because they will compete for intracellular phosphorylation [23].

These agents are used as components of the preferred dual NRTI combination (tenofovir-emtricitabine, abacavir-lamivudine). In addition, lamivudine has been used in combination with dolutegravir or a boosted PI in certain treatment-naïve and -experienced patients. (See "Selecting antiretroviral regimens for treatment-naïve persons with HIV-1: General approach", section on 'Approach for most patients' and "Selecting antiretroviral regimens for treatment-naïve persons with HIV-1: General approach", section on 'Use of two-drug regimens in select patients' and "Switching antiretroviral therapy for adults with HIV-1 and a suppressed viral load".)

Both emtricitabine and lamivudine are extremely well tolerated. Rare side effects include:

Pancreatitis – Lamivudine has been associated with pancreatitis, although the risk in adults is quite low at 0.3 percent [24]. The risk of pancreatitis may be higher in children [24].

Skin changes – Emtricitabine can cause skin discoloration that presents as hyperpigmentation usually on the palms and/or soles. The incidence appears to be approximately 2 percent [25]. The mechanism and clinical significance is unknown.

Tenofovir — Tenofovir is an acyclic nucleotide diester analog of adenosine monophosphate, which is administered orally as the prodrug tenofovir alafenamide (TAF) or tenofovir disoproxil fumarate (TDF). Both are converted intracellularly to the pharmacologically active moiety, tenofovir-diphosphate.

TAF is administered at a lower dose than TDF, and plasma concentrations of tenofovir are lower; however, intracellular concentrations of tenofovir-diphosphate are higher. Because of differences in drug transporters involved in TAF versus TDF absorption, there are drug interactions (eg, certain anticonvulsants, rifamycins), which can lower TAF, but not TDF levels. Specific information on drug interactions can be found in the Lexicomp drug interactions program within UpToDate.

Tenofovir alafenamide — Tenofovir alafenamide (TAF) is available as part of several coformulated tablets: elvitegravir-cobicistat-emtricitabine-tenofovir alafenamide (EVG/c/FTC/TAF), rilpivirine-emtricitabine-tenofovir alafenamide (RPV/FTC/TAF), tenofovir alafenamide-emtricitabine (TAF/FTC), bictegravir-emtricitabine-tenofovir alafenamide (BIC/FTC/TAF), and darunavir-cobicistat-emtricitabine-tenofovir alafenamide (DRC/c/FTC/TAF). These coformulated tablets are approved for use in patients with an estimated glomerular filtration rate (eGFR) ≥30 mL/min/1.73 m2. There are insufficient data for dosing recommendations in patients with an eGFR <30 mL/min/1.73 m2, unless they are on hemodialysis. Special considerations for patients with HIV/HBV coinfection are presented elsewhere. (See "Treatment of chronic hepatitis B in patients with HIV", section on 'Tenofovir in patients with reduced kidney function'.)

In these coformulated tablets, TAF is used in two different doses: either 10 or 25 mg. When TAF is combined within a pharmacokinetic boosting agent (eg, cobicistat or ritonavir) as in elvitegravir-cobicistat-emtricitabine-tenofovir alafenamide, the reduced dose of 10 mg is used. When TAF is not given with a pharmacokinetic booster, as in rilpivirine-emtricitabine-tenofovir alafenamide and bictegravir-emtricitabine-tenofovir alafenamide, the dose is 25 mg. Pharmacologic boosting agents (eg, cobicistat, ritonavir) inhibit intestinal P-glycoprotein, thereby increasing TAF bioavailability and tenofovir plasma concentrations; the reduced TAF dose of 10 mg is used to manage this interaction. The long-term safety of TAF at the 25 mg dose given with a pharmacokinetic booster has not been established.

Tenofovir can lead to kidney injury and bone loss; however TAF is associated with less renal and bone toxicity compared with TDF since it achieves lower plasma tenofovir concentrations [26]. In two randomized studies that included 1733 patients, elvitegravir-cobicistat-emtricitabine-tenofovir alafenamide was compared with elvitegravir-cobicistat-emtricitabine-tenofovir disoproxil fumarate (EVG/c/FTC/TDF) [27]. More than 90 percent of patients achieved a plasma HIV-RNA <50 copies/mL after 48 weeks, regardless of which agent they received. However, those who received the TAF-containing regimen had significantly smaller increases in serum creatinine (0.08 versus 0.12 mg/dL), less proteinuria (median percent change –3 versus 20), and smaller decreases in bone mineral density (median percent change spine: –1·30 versus –2.86; hip –0.66 versus –2.95).

Although TAF has several advantages over TDF, TAF may be associated with greater weight gain [28-30]. In addition, levels of LDL and high-density lipoprotein (HDL) cholesterol, as well as triglycerides, have been found to be higher in patients receiving TAF compared with those receiving TDF in randomized trials [27,28]. However, the total cholesterol-to-HDL ratio did not differ between patients receiving TAF and TDF; thus, the clinical significance of these findings is unclear.

Tenofovir disoproxil fumarate — Tenofovir disoproxil fumarate (TDF) is administered once daily and taken without regard to food. TDF is usually combined with emtricitabine (FTC), which is available as a coformulated tablet (TDF [300 mg]/FTC [200 mg]) [4]. For treatment of HIV infection, tenofovir disoproxil fumarate-emtricitabine must be given with a third agent from a different class, and several combinations of these drugs are available as coformulated tablets (table 1). There are also generic combination tablets that include TDF, such as TDF (300 mg) with lamivudine (300 mg), which are available in resource-limited countries.

The main toxicities of TDF include kidney injury and bone loss:

Kidney injury TDF can lead to renal impairment, characterized by increases in serum creatinine, proteinuria, glycosuria, hypophosphatemia, and acute tubular necrosis [31]. Patterns of kidney injury include proximal tubular dysfunction (Fanconi syndrome), acute kidney injury, chronic kidney disease, and nephrogenic diabetes insipidus [32].

Baseline risk factors for the development of kidney injury from TDF include elevated serum creatinine, older age, advanced HIV infection, low body weight, hepatitis C coinfection, and prescription of concurrent nephrotoxic drugs [33]. Concomitant use with a pharmacokinetically boosted regimen (eg, a protease inhibitor plus ritonavir or elvitegravir plus cobicistat) may also increase the risk of renal dysfunction [4]. An additional discussion of tenofovir nephrotoxicity is found elsewhere. (See "Overview of kidney disease in patients with HIV", section on 'Medication nephrotoxicity'.)

Given the potential for nephrotoxicity:

For patients with HIV, we generally avoid administering TDF to those with a baseline eGFR <60 mL/min/1.73 m2. However, if TDF must be used in this setting, the manufacturer recommends the dose be adjusted when the eGFR falls below 50 mL/min/1.73 m2. If cobicistat is used as part of the ART regimen, TDF should not be administered to patients with a pretreatment eGFR <70 mL/min/1.73 m2. (See "Selecting antiretroviral regimens for treatment-naive persons with HIV-1: Patients with comorbid conditions", section on 'Reduced kidney function'.)

Renal function and proteinuria should be monitored during the use of this agent. (See "Administration of pre-exposure prophylaxis against HIV infection", section on 'Patient monitoring' and "Patient monitoring during HIV antiretroviral therapy".)

In patients with end-stage kidney disease on hemodialysis in whom recovery of renal function is not anticipated, TDF may be a convenient choice because it can be dosed at 300 mg once weekly (every seven days). (See 'Timing of doses for patients on hemodialysis' below.)

Bone loss – TDF is associated with decreased bone mineral density, which usually stabilizes with continued use [34-36]. In one randomized trial, TDF was associated with a greater reduction in bone mineral density compared with abacavir [36]. (See "Bone and calcium disorders in patients with HIV", section on 'Risk with specific agents'.)

Overall, tenofovir disoproxil fumarate-emtricitabine is very well tolerated, and a number of well-designed studies have found that this combination is effective in suppressing HIV RNA when used with a variety of different third agents. Although data are limited, tenofovir disoproxil fumarate-emtricitabine has generally been superior to other nucleoside combinations [12,21,22,25,37-39]. In addition, the combination of tenofovir disoproxil fumarate-emtricitabine has been found to be effective in reducing HIV transmission when administered as pre-exposure prophylaxis (PrEP) to patients without HIV. A detailed discussion of tenofovir disoproxil fumarate-emtricitabine for PrEP is found elsewhere. (See "Administration of pre-exposure prophylaxis against HIV infection".)

Zidovudine — Zidovudine (also known as AZT) was the first antiretroviral medication used for the treatment of HIV that demonstrated a reduction in the morbidity and mortality associated with HIV infection. Zidovudine is dosed twice daily, as pharmacokinetic and pharmacodynamic studies with once-daily dosing suggest that intracellular trough levels are lower and HIV RNA declines are less robust than with twice-daily dosing [40,41].

Adverse reactions associated with zidovudine include headache, malaise, anorexia, nausea, vomiting, lactic acidosis, and loss of limb fat. Administration of zidovudine with food improves gastrointestinal tolerability. The major dose-limiting toxicities of zidovudine are related to bone marrow suppression (eg, anemia and/or neutropenia).

The coformulation of zidovudine (300 mg) and lamivudine (150 mg) is dosed as one tablet twice daily. This NRTI combination was a preferred choice when it was first introduced in 1997, but is now rarely used. A tenofovir-containing nucleoside combination or abacavir-lamivudine is administered instead of zidovudine-lamivudine for the following reasons:

Zidovudine requires twice-daily dosing whereas tenofovir disoproxil fumarate-emtricitabine, tenofovir alafenamide-emtricitabine, and abacavir-lamivudine are dosed once daily.

Zidovudine is associated with more severe adverse reactions [38,42].

Zidovudine-lamivudine has been shown to have inferior virologic potency compared with tenofovir disoproxil fumarate-emtricitabine-based ART [25,38].

However, there are certain situations where zidovudine may still be indicated. As an example, there may be a benefit of using this drug in the setting of the K65R mutation when patients have difficulty achieving viral suppression with more commonly used regimens. In addition, the intravenous formulation of zidovudine (zidovudine is one of the only agents available in a parenteral form) is administered to certain pregnant women at the time of delivery. More detailed discussions of the management of patients with drug-resistant virus and pregnant women are found elsewhere. (See "Selecting an antiretroviral regimen for treatment-experienced patients with HIV who are failing therapy", section on 'Patients who have failed multiple regimens' and "Antiretroviral selection and management in pregnant women with HIV in resource-rich settings" and "Intrapartum management of pregnant women with HIV and infant prophylaxis in resource-rich settings", section on 'Intrapartum antiretrovirals'.)

NON-NUCLEOSIDE REVERSE TRANSCRIPTASE INHIBITORS (NNRTIs)

NNRTI class characteristics — The non-nucleoside reverse transcriptase inhibitors (NNRTIs) are typically administered with a dual nucleoside reverse transcriptase inhibitor (NRTI) combination, although the combination of dolutegravir-rilpivirine is an option for select patients who are virologically suppressed and are switching regimens. (See "Switching antiretroviral therapy for adults with HIV-1 and a suppressed viral load".)

First-generation NNRTIs include efavirenz and nevirapine; other NNRTIs (doravirine, etravirine, and rilpivirine) have resistance profiles that are distinct from efavirenz and nevirapine. Most NNRTIs are used for treatment-naïve patients and those with suppressed viral loads switching regimens, except for etravirine, which is generally used for individuals with evidence of drug-resistant virus.

Mechanism of action – NNRTIs prevent HIV-1 reverse transcriptase from adding new nucleotides to the growing DNA chain. The NNRTIs block viral cDNA elongation at a site that is separate from the active site targeted by the NRTI class. The NNRTIs bind to a hydrophobic pocket in the palm subdomain of p66, approximately 10 angstroms from the polymerase active site [43]. This causes a stereochemical change in the protein, which reduces the ability of naturally occurring nucleosides to bind to the active site pocket. Reverse transcriptase becomes less flexible and DNA polymerization is inhibited. In addition, cDNA elongation is reduced, which ultimately leads to a decline in viral replication [44]. (See 'Overview of HIV replication' above.)

Spectrum of activity – The NNRTIs are active against HIV-1 but are not active against HIV-2.

Resistance – The first-generation NNRTIs (ie, efavirenz and nevirapine) have a low barrier to resistance and require only a single mutation to confer drug resistance. A major mutation (eg, K103N) leads to cross resistance to other first-generation NNRTIs. In some cases, the second-generation NNRTI, etravirine, can be used in the treatment of patients with resistance to first-generation NNRTIs [4,45]. Virologic failure on rilpivirine may also compromise the anti-HIV activity of other NNRTIs, including etravirine. (See "Interpretation of HIV drug resistance testing", section on 'Non-nucleoside reverse transcriptase inhibitors' and "Selecting an antiretroviral regimen for treatment-experienced patients with HIV who are failing therapy", section on 'NNRTI-containing regimens'.)

Oral NNRTIs have a long plasma half-life, typically >24 hours in adults, with the exception of doravirine, which has a half-life of 15 to 21 hours. The long half-life allows all NNRTIs to be administered once daily. However, when an NNRTI-containing antiretroviral regimen is discontinued, a period of functional NNRTI monotherapy may occur if the concentrations of the NNRTI decline slower than those of the other antiretrovirals; this period of functional monotherapy or "NNRTI tail" increases the risk of developing NNRTI resistance [46,47]. Thus, some experts suggest that if a regimen containing an NNRTI is stopped, the concomitant nucleoside backbone should be continued for three to seven days after the NNRTI is discontinued. An alternative strategy is for a boosted protease inhibitor to be substituted for the NNRTI for one week before discontinuing the nucleoside backbone. These relatively cumbersome stop strategies have not been well studied, and it is difficult to recommend any one approach. Specific considerations regarding the injectable formulation are presented elsewhere. (See "Switching antiretroviral therapy for adults with HIV-1 and a suppressed viral load".)

Drug interactions – Drug interactions can result in increased or decreased NNRTI levels, and NNRTIs may be a perpetrator of drug interactions. Detailed information about specific interactions is available by using the Lexicomp drug interactions program included with UpToDate.

Adverse events – There are no specific class-related adverse events. However, the two most commonly used NNRTIs (efavirenz and rilpivirine) can result in neurologic and psychiatric side effects. They are also associated with QT prolongation.

Specific agents (listed alphabetically)

Doravirine — Doravirine became available in the United States in August of 2018. This agent is approved for use in treatment-naïve patients and is available alone or in combination with tenofovir disoproxil fumarate-lamivudine. Doravirine may be administered with or without food, and there are no viral load or CD4 count parameters to initiate therapy.

Doravirine is metabolized by CYP3A, and drugs that induce or inhibit CYP3A may affect plasma concentrations. As an example, rifampin decreases doravirine exposure by >70 percent, and coadministration is contraindicated. By contrast, rifabutin decreases doravirine exposure by 50 percent, and this interaction may be managed by increasing the doravirine dose from 100 mg once daily to 100 mg twice daily. Detailed information about specific interactions is available by using the Lexicomp drug interactions program within UpToDate.

Doravirine was found to be non-inferior to efavirenz- and darunavir-based regimens in randomized trials [48,49]. In addition, it appears to be well tolerated. Although it may cause neurologic and psychiatric side effects similar to efavirenz (eg, dizziness, sleep disturbances, and altered sensorium), these are much less common. In the trial that compared doravirine-lamivudine- tenofovir disoproxil fumarate with efavirenz-emtricitabine-tenofovir disoproxil fumarate, the rates of neuropsychiatric adverse events at 48 weeks were 12 versus 22 percent, respectively [48]. Clinical trial data also suggest that it has better lipid profiles than both efavirenz and darunavir [48,49]. Additional information on the adverse events associated with efavirenz and darunavir are presented elsewhere. (See 'Efavirenz' below and 'Darunavir' below.)

Despite the favorable clinical trial data, there is limited experience with this agent, and doravirine has never been directly compared with INSTIs, the preferred third agent for most patients. (See "Selecting antiretroviral regimens for treatment-naïve persons with HIV-1: General approach", section on 'Approach for most patients'.)

Efavirenz — Efavirenz is given at a dose of 600 mg once daily. It is also available as part of coformulated tablets (efavirenz-emtricitabine-tenofovir disoproxil fumarate and efavirenz-lamivudine-tenofovir disoproxil fumarate) (table 1). Since efavirenz is a potent inducer of hepatic cytochrome P450 (CYP) enzymes, drug interactions can be seen with certain medications that are commonly used in patients with HIV. These include rifabutin, clarithromycin, atorvastatin, simvastatin, methadone, carbamazepine, and other anticonvulsants. Detailed information about specific interactions is available by using the Lexicomp drug interactions program included with UpToDate.

Efavirenz has been used extensively and has excellent potency and durability in treatment-naïve patients, including those with high pretreatment viral loads and low CD4 cell counts [10,25,50-57]. However, integrase strand transfer inhibitors are generally preferred, due in part to fewer adverse events [4,5,20,58-60]. (See 'Integrase strand transfer inhibitors (INSTIs)' below.)

Side effects seen with efavirenz include central nervous system (CNS) toxicity, elevated hepatic transaminases (including fulminant hepatitis), and QT interval prolongation. Thus, this agent is typically avoided in patients with psychiatric illness, moderate to severe liver disease (Child Pugh B and C), and those at risk for torsade de pointes. Other side effects include rash and hyperlipidemia. The safety of efavirenz during pregnancy is discussed elsewhere. (See "Safety and dosing of antiretroviral medications in pregnancy", section on 'Efavirenz'.)

The CNS symptoms, which are one of the most frequently seen side effects, commonly include vivid dreams, confusion, dizziness, and a "hungover" feeling the following day. Higher rates of intolerance are noted in Black patients due to slower metabolism that is genetically determined. Taking efavirenz on an empty stomach may reduce the risk of these side effects since high-fat/high-caloric meals increase the absorption of efavirenz.

Although many CNS side effects are restricted to the first few weeks after initiation, there are data that suggest long-term mental health consequences (eg, irritability, anxiety, mood changes, depression) and increased suicidality (combined endpoint of suicidal ideation, suicide attempts, and completed suicides) can be seen with efavirenz [61-64]. A combined analysis of 5332 HIV-infected individuals from four clinical trials evaluated the risk of suicidality among those receiving an efavirenz-containing regimen [63]. There was a significantly increased incidence of suicidality among those who received efavirenz compared with those who did not (8.08 versus 3.66 per 1000 person-years; hazard ratio 2.28; 95% CI, 1.27-4.10). The increased risk of suicidality appears to be greatest in patients who have genotypes that predict higher plasma efavirenz exposure [65]. This may help explain why an increase in suicidality is not seen in all patient groups [66].

A fixed-dose combination of tenofovir disoproxil fumarate-lamivudine-efavirenz using a lower dose of efavirenz (400 mg) is available. Clinical trial data in antiretroviral-naïve persons suggest that this lower dose of efavirenz is noninferior to the 600 mg dose with regards to virologic efficacy [67,68] and may be associated with fewer adverse events [67]. In one study, the 400 mg dose of efavirenz appeared to be noninferior to dolutegravir at 48 weeks, with 69 and 75 percent of patients achieving a viral load <50 copies, respectively [69]. However, the 400 mg dose of efavirenz is not routinely used in clinical practice.

Etravirine — The most commonly used NNRTI for patients with drug-resistant virus is etravirine [70]. The likelihood that etravirine will be active against a particular virus can be determined via a weighted mutation score based upon the specific NNRTI mutations (table 2). (See "Interpretation of HIV drug resistance testing", section on 'Non-nucleoside reverse transcriptase inhibitors'.)

If etravirine is part of the new regimen, it should be used with at least one, and ideally two, other fully active drugs; if this is not possible, it should be used with multiple partially active agents. Increasing the number of active antiretroviral agents used in combination with etravirine increases the likelihood of obtaining the desired treatment response [16,17]. As an example, in a study of 243 patients with multidrug resistance, the use of two or more active agents in combination with etravirine increased the likelihood of achieving HIV RNA suppression by more than eightfold [17].

The most common adverse event after etravirine administration is rash, which is generally mild. In clinical trials, rash occurred primarily in the second week of therapy and was infrequent after week four. Rash generally resolved within one to two weeks of continued dosing. As with other NNRTIs, severe rashes (including Stevens-Johnson Syndrome, toxic epidermal necrolysis, and erythema multiforme) have been reported in the postmarketing experience. Etravirine should be immediately discontinued when signs and symptoms of severe skin or hypersensitivity reactions develop.

There are significant drug-drug interactions with etravirine and other antiretroviral medications that can limit its use. As an example, if etravirine is used with dolutegravir, the regimen must also include a boosted PI. A detailed review of drug interactions can be found in the Lexicomp drug interactions program within UpToDate.

Nevirapine — In the United States, nevirapine is no longer recommended for the initial treatment of HIV infection in treatment-naïve patients [4,5]. Compared with other NNRTIs, nevirapine is associated with increased toxicity, especially during the first three months of therapy. Complications include hepatic necrosis and serious cutaneous reactions, including deaths due to hepatic necrosis or Stevens-Johnson Syndrome [71-78].

If nevirapine is being considered, it should not be initiated in females with a baseline CD4 count >250 cells/microL or in males with a CD4 count >400 cells/microL since these CD4 cell thresholds represent major risk factors for hepatotoxicity. Caution should also be used when nevirapine is initiated in males with CD4 cell counts between 250 to 400 cells/microL. A study of 566 patients from China showed that a CD4 cell count of >250 cells/microL was an independent predictor of hepatotoxicity in both males and females initiated on a nevirapine-containing regimen [79].

Nevirapine should also be avoided in patients with moderate to severe liver disease (Child-Pugh Class B or C) (calculator 1). As described above, nevirapine has been associated with hepatotoxicity, and data on its use in this population are limited.

If administered, nevirapine should be started at a dose of 200 mg once daily for 14 days; the dose is then increased to the maintenance dose of 200 mg twice daily. We monitor serum aminotransferases at baseline, prior to and two weeks after dose escalation, then monthly for the first three months. Patients should also be counseled about symptoms and signs of hepatotoxicity (eg, nausea, jaundice).

Rilpivirine — Rilpivirine is formulated both as an individual tablet (25 mg once daily) and in three fixed-dose combination tablets (rilpivirine-emtricitabine-tenofovir disoproxil fumarate, rilpivirine-emtricitabine-tenofovir alafenamide, and dolutegravir-rilpivirine), all of which can be administered once daily (table 1). It is also available in an injectable formulation that must be used with cabotegravir. (See 'Cabotegravir' below.)

Rilpivirine can be used in combination with tenofovir-emtricitabine in treatment-naïve patients with a baseline viral load <100,000 copies/mL and a CD4 cell count ≥200 cells/microL [4]. This combination, as well as the two-drug combination of dolutegravir-rilpivirine, can also be used in select virologically suppressed patients who are switching regimens. (See "Switching antiretroviral therapy for adults with HIV-1 and a suppressed viral load".)

Rilpivirine should be used cautiously or avoided in patients with long QT syndrome or other risks for torsade de pointes [4]. High doses of rilpivirine (ie, 75 mg and 300 mg once daily) were associated with QT interval prolongation [80].

Gastric acid and food are required for optimal absorption; thus, rilpivirine must be administered with a meal, and concomitant use of rilpivirine with proton pump inhibitors is contraindicated. In addition, other drugs that alter the stomach pH (eg, H2 antagonists, antacids) may impair absorption and should be avoided if possible; however, if they are used, they should be dosed separately from rilpivirine.

Important drug interactions can also be seen with certain anti-tuberculosis medications (eg, rifampin and rifabutin), anti-epileptics (eg, phenytoin, phenobarbital), and drugs that may inhibit rilpivirine metabolism and increase plasma concentrations (eg, azole antifungals, macrolide antibiotics). Detailed information about specific interactions is available by using the Lexicomp drug interactions program within UpToDate.

Rilpivirine is generally well tolerated [81]. The most common adverse drug reactions to rilpivirine include depression, insomnia, headache, and rash. Although rilpivirine may cause neurologic and psychiatric side effects similar to efavirenz, they are much less common [82]. In addition, rash led to drug discontinuation in less than 1 percent of study participants in the phase 3 clinical trials [83].

Two controlled trials (ECHO and THRIVE) compared rilpivirine with efavirenz in treatment-naïve patients in combination with two NRTIs; most received tenofovir disoproxil fumarate-emtricitabine as the NRTI combination [84-86]. The pooled results demonstrated that the proportion of patients with viral suppression at 48 weeks was comparable in the rilpivirine and efavirenz-containing arms. Rilpivirine was also well tolerated and associated with minimal changes in lipids.

In a 96-week analysis of 1368 participants, rilpivirine had a lower incidence of any neurologic or psychiatric adverse event compared with efavirenz (38 versus 17 percent and 24 versus 16 percent, respectively) [85]. However, among those subjects with a baseline viral load >100,000 copies/mL, the risk of virologic failure was higher in the rilpivirine arm compared with the efavirenz arm. An increased risk of nucleoside analog resistance mutations (eg, M184V/I) was also noted in these trials [84-86].

The combination of rilpivirine-emtricitabine-tenofovir alafenamide was approved on the basis of pharmacokinetic bioequivalence; comparative clinical trial data have not yet been reported.

INTEGRASE STRAND TRANSFER INHIBITORS (INSTIs)

INSTI class characteristics — Regimens using integrase strand transfer inhibitors (INSTIs), such as raltegravir, elvitegravir, dolutegravir, and bictegravir, are effective and well tolerated. Most INSTIs are administered orally. An injectable, long-acting INSTI (cabotegravir) is also available and must be used in combination with injectable rilpivirine.

In many countries, including the United States, INSTIs are considered the preferred third agent for treatment-naïve individuals when used in combination with two nucleoside analogues. These agents are a particularly attractive option for patients with either abnormal lipid profiles or risk factors for coronary artery disease, since they have a neutral effect on cholesterol and triglycerides compared with efavirenz and protease inhibitors. (See "Selecting antiretroviral regimens for treatment-naïve persons with HIV-1: General approach".)

Mechanism of action – HIV integrase is one of three enzymes (reverse transcriptase, protease, and integrase) that are encoded by the virus and are essential for HIV replication. After entry into CD4+ T cells, viral RNA is reverse transcribed into DNA by HIV reverse transcriptase. The integrase enzyme catalyzes the process by which viral DNA is integrated into the genome of the host cell. This process is essential for maintenance of the viral genome and viral gene expression [87-92].

Integrase inhibitors target the strand transfer step of viral DNA integration and are sometimes referred to as an "INSTI." These drugs prevent or inhibit the binding of the preintegration complex (PIC) to host cell DNA, thus terminating the integration step of HIV replication. (See 'Overview of HIV replication' above.)

Spectrum of activity – The integrase inhibitors are active against HIV-1 and HIV-2.

Resistance – HIV-infected patients who fail an antiretroviral therapy (ART) regimen that contains raltegravir or elvitegravir have a relatively high risk of developing integrase resistance. In addition, there appears to be a high level of cross resistance between raltegravir and elvitegravir, precluding their sequential use [93-95]. As an example, in one study, 13 of the 23 patients who experienced virologic failure while receiving elvitegravir developed the following drug resistance mutations: T66I, E92Q, Q148R, and N155H [96]; three of these mutations (E92Q, Q148R, and N155H) also conferred significant resistance to raltegravir.

By contrast, dolutegravir and bictegravir resistance is very uncommon among patients failing an initial regimen that contains one of these agents [97,98]. It is also uncommon for such individuals to develop resistance to the nucleoside reverse transcriptase inhibitors (NRTIs) that are administered in combination with dolutegravir or bictegravir.

Drug interactions – A decrease in INSTI concentrations is possible when administered with polyvalent cation-containing antacids and supplements. With regards to specific agents, there are relatively few problematic drug-drug interactions with raltegravir and dolutegravir because they are primarily metabolized via glucuronidation. Bictegravir, which is cleared by both glucuronidation and hepatic cytochrome P450, also has few problematic drug interactions. By contrast, elvitegravir is associated with more drug-drug interactions since it is administered with the boosting agent cobicistat, which is a potent inhibitor of CYP3A. More specific information on drug-drug interactions is found in the sections below and in the Lexicomp drug interactions program within UpToDate.

Adverse events – All INSTIs are generally well tolerated. However, INSTIs may be associated with increased weight gain compared with other agents [30,69,99-101]. In one study that evaluated data from eight randomized trials, weight gain at 96 weeks was approximately 3.5 kg, and was similar in those that received bictegravir- and dolutegravir-based regimens. The mechanism for this weight gain is not known.

Insomnia and dizziness can also occur in some patients receiving INSTIs [102,103]. In addition, depression and suicidal ideation have been reported on rare occasion, primarily in patients with a history of psychiatric illnesses [4]. On occasion, elevations in creatine phosphokinase (CPK), rhabdomyolysis, and myopathy or myositis have been reported in patients taking raltegravir and dolutegravir [4].

Considerations in persons of childbearing potential – When deciding whether to use a regimen that includes an INSTI in individuals who are of childbearing potential, it is important to determine if the person is pregnant and, if not, if they are committed to using effective birth control (eg, oral, injectable, or intrauterine contraception). Pregnancy, as well as the desire to become pregnant, may impact the selection of ART agents used for treatment of HIV infection. More detailed information on the use of INSTIs in persons who are of childbearing potential and in those who are pregnant is discussed elsewhere. (See "HIV and women" and "Safety and dosing of antiretroviral medications in pregnancy" and "Antiretroviral selection and management in pregnant women with HIV in resource-rich settings", section on 'ART selection and management' and "Prevention of vertical HIV transmission in resource-limited settings".)

Specific agents (listed alphabetically)

Bictegravir — Bictegravir was approved for use in February 2018 and is available as a coformulated tablet with emtricitabine and tenofovir alafenamide. It has been approved for use in treatment-naïve patients and in those who are virologically suppressed and are considering a change in their regimen. (See "Selecting antiretroviral regimens for treatment-naïve persons with HIV-1: General approach" and "Switching antiretroviral therapy for adults with HIV-1 and a suppressed viral load".)

There are limited data using this drug in treatment-experienced patients with a history of virologic failure. (See "Selecting an antiretroviral regimen for treatment-experienced patients with HIV who are failing therapy".)

Overall, bictegravir-emtricitabine-tenofovir alafenamide appears well tolerated. Notable drug interactions include rifampin and dofetilide, which are contraindicated for coadministration with bictegravir.

Bictegravir-emtricitabine-tenofovir alafenamide can be used in patients on hemodialysis; it is approved for use in treatment-experienced patients who are virologically suppressed. Similar to most other antiretroviral agents, it has not been evaluated in treatment-naïve patients on hemodialysis.

Cabotegravir — Cabotegravir is available as an oral and injectable preparation, which must be used in conjunction with rilpivirine [4,5]. The oral formulation can be used as lead-in therapy prior to initiating injectable therapy. Injectable cabotegravir-rilpivirine should only be used in treatment-experienced patients who are virologically suppressed, able to tolerate 28 days of lead-in therapy with oral cabotegravir plus rilpivirine, have no resistance to either drug, do not have chronic hepatitis B infection, have no significant drug-drug interactions that might reduce either drug's concentration, and are able to adhere to monthly clinic visits for intramuscular injections. (See "Switching antiretroviral therapy for adults with HIV-1 and a suppressed viral load".)

The injectable regimen is administered every four or eight weeks. Overall, cabotegravir-rilpivirine is well tolerated; however, when the injectable formulation is used, mild to moderate injection site reactions are common. The most common other adverse reactions are mild and nonspecific, such as pyrexia, fatigue, headache, muscle aches, nausea, sleep disorders, dizziness, and rash. On rare occasion, serious post-injection reactions due to rilpivirine have been reported (dyspnea, flushing, sweating); however, these typically resolve within minutes.

Some drugs are contraindicated for coadministration with intramuscular cabotegravir-rilpivirine, such as the rifamycins (eg, rifampin) and certain anticonvulsants (eg, phenytoin). Information on specific drug-drug interactions can be found in the Lexicomp drug interactions program within UpToDate.

A more detailed discussion of cabotegravir-rilpivirine, including dosing regimens, the approach to missed doses, and the data supporting its efficacy, are presented in the Lexicomp drug information topic and a separate topic review. (See "Switching antiretroviral therapy for adults with HIV-1 and a suppressed viral load".)

Dolutegravir — Dolutegravir is approved for use in treatment-naïve and most treatment-experienced individuals [104]. Patients receiving dolutegravir are unlikely to develop drug-resistant virus [105].

For most patients, dolutegravir is given at a dose of 50 mg once daily without regard to food. It can be administered with a tenofovir-containing nucleoside combination or abacavir-lamivudine. Topic reviews that discuss regimen selection are presented separately. (See "Selecting antiretroviral regimens for treatment-naïve persons with HIV-1: General approach" and "Switching antiretroviral therapy for adults with HIV-1 and a suppressed viral load" and "Selecting an antiretroviral regimen for treatment-experienced patients with HIV who are failing therapy".)

Dolutegravir is available as part of three coformulated tablets (dolutegravir-abacavir-lamivudine, dolutegravir-lamivudine, and dolutegravir-rilpivirine) (table 1).

Dolutegravir-rilpivirine is only approved for use in virologically suppressed individuals switching regimens. Detailed discussions of the use of these regimens, including contraindications to their use, are presented elsewhere. (See "Selecting antiretroviral regimens for treatment-naïve persons with HIV-1: General approach" and "Switching antiretroviral therapy for adults with HIV-1 and a suppressed viral load".)

The dose of dolutegravir should be increased to 50 mg twice daily for treatment-experienced patients who have previously taken integrase inhibitors and have confirmed or suspected integrase resistance. (See "Selecting an antiretroviral regimen for treatment-experienced patients with HIV who are failing therapy", section on 'Patients with drug-resistant virus'.)

Dolutegravir should not be used for patients who have the Q148 mutation in addition to two or more secondary integrase mutations, since such patients respond less well to a dolutegravir-containing regimen [106-108]. (See "Selecting an antiretroviral regimen for treatment-experienced patients with HIV who are failing therapy", section on 'Integrase strand transfer inhibitors'.)

Dolutegravir can block tubular secretion of creatinine without reducing renal glomerular function. In some patients, this can lead to an expected small increase in serum creatinine (and decrease in eGFR) [98].

Several trials have shown that dolutegravir is well tolerated [20,109,110]. However, dolutegravir may be associated with more central nervous side effects (CNS) compared with other INSTIs [102]. This was demonstrated in a retrospective review of 1704 patients taking 1950 INSTI-based therapies, where neuropsychiatric adverse events (eg, insomnia and dizziness) resulted in treatment discontinuation more frequently in those receiving dolutegravir versus a different INSTI (5.6 percent for dolutegravir versus 0.7 and 1.9 percent for elvitegravir and raltegravir, respectively). These CNS side effects occurred significantly more frequently in females, and in patients older than 60 years.

Dolutegravir can have significant interactions with agents, such as metformin, rifampin, and antiepileptics (eg, phenytoin, phenobarbital, and carbamazepine). Detailed information can be found in the Lexicomp drug interactions program within UpToDate.

There are no clinically significant drug interactions between dolutegravir and NRTIs or the boosted protease inhibitors atazanavir and lopinavir. This is because dolutegravir does not induce or inhibit CYP or UGT isoenzymes at clinically relevant concentrations [111]. Although dolutegravir trough concentrations are reduced by approximately 38 percent when given with darunavir/ritonavir, this is not considered clinically significant. In addition, it appears there is not a clinically significant adverse interaction if dolutegravir is given with darunavir/cobicistat [112]. However, there are very limited data assessing possible interactions between dolutegravir and cobicistat.

The concentration of dolutegravir can be affected when dolutegravir is administered with drugs that inhibit or induce the UGT family of enzymes and CYP3A4. As an example, the non-nucleoside reverse transcriptase inhibitors (NNRTIs) efavirenz and etravirine reduce dolutegravir trough concentrations by 75 and 88 percent, respectively. Dose adjustments or coadministration with a ritonavir-boosted protease inhibitor are required when these agents are used together; specific recommendations can be found at the United States Department of Health and Human Services (DHHS) Antiretroviral Guidelines for Adults and Adolescents.

Clinical trial data supporting the efficacy of dolutegravir are summarized below:

Studies of dolutegravir have used both tenofovir disoproxil fumarate-emtricitabine and abacavir-lamivudine as nucleoside backbone therapy [109]. The virologic response rates were comparable to those seen in patients treated with raltegravir, even among patients with baseline HIV RNA >100,000 copies/mL, and there were no differences in the rates of discontinuation for adverse events.

At 48 weeks, dolutegravir-abacavir-lamivudine was associated with significantly more frequent viral suppression to <50 copies/mL (88 versus 81 percent) and higher CD4 counts (267 versus 208 cells/microL) when compared with efavirenz-emtricitabine-tenofovir disoproxil fumarate [20]. This was seen even among individuals with a baseline HIV RNA >100,000 copies/mL.

Patients receiving dolutegravir were more likely to have an HIV RNA level <50 copies/mL (90 versus 83 percent) compared with darunavir/ritonavir (800/100 mg once daily) in an open-label, randomized clinical trial [113]. This difference was greatest in those with a baseline HIV RNA >100,000 copies/mL.

Elvitegravir — Elvitegravir can be used for treatment-naïve or treatment-experienced patients. However, it must be administered with a boosting agent (eg, cobicistat or ritonavir), which is a potent inhibitor of CYP3A and thus causes more potential drug-drug interactions. A discussion of pharmacokinetic boosting is found below. (See 'Pharmacokinetic boosting' below.)

Elvitegravir was initially approved for use in treatment-naïve HIV-infected patients as part of a fixed-dose antiretroviral combination, elvitegravir-cobicistat-emtricitabine-tenofovir disoproxil fumarate (EVG/c/FTC/TDF). This combination can be administered once daily, but only to patients with a pre-treatment eGFR >70 mL/min/1.73 m2. In November 2015, the coformulation of elvitegravir-cobicistat-emtricitabine-tenofovir alafenamide (EVG/c/FTC/TAF) became available for treatment-naïve patients. Elvitegravir-cobicistat-emtricitabine-tenofovir alafenamide can be administered to patients with reduced kidney function who have an eGFR ≥30 mL/min/m2.

Elvitegravir, like raltegravir, has a low genetic barrier to resistance. In addition, drug-drug interactions should be thoroughly assessed as it must be administered with a pharmacologic boosting agent. (See 'Pharmacokinetic boosting' below.)

Clinical trials supporting the use of elvitegravir include:

Two trials that enrolled 1408 treatment-naïve patients with a pre-treatment eGFR >70 mL/min/1.73 m2 found that elvitegravir-cobicistat-emtricitabine-tenofovir disoproxil fumarate was noninferior to coformulated efavirenz-emtricitabine-tenofovir disoproxil fumarate or ritonavir-boosted atazanavir plus tenofovir disoproxil fumarate-emtricitabine in achieving viral suppression at 48 weeks of therapy [58,114]. The most common adverse events on the elvitegravir regimen were headache and transient diarrhea and nausea; four patients developed proximal tubulopathy in these phase 3 clinical trials.

Trials have shown elvitegravir-cobicistat-emtricitabine-tenofovir alafenamide has equal efficacy and less nephrotoxicity and bone loss compared with elvitegravir-cobicistat-emtricitabine-tenofovir disoproxil fumarate [27,115]. In addition, patients who are virologically suppressed on a tenofovir disoproxil fumarate-containing regimen were able to safely switch to elvitegravir-cobicistat-emtricitabine-tenofovir alafenamide. In an open-label, randomized trial of 1443 patients who were virologically suppressed on a regimen containing tenofovir disoproxil fumarate for 96 weeks, patients were switched to elvitegravir-cobicistat-emtricitabine-tenofovir alafenamide or were continued on their regimen [115]. By 48 weeks, the tenofovir alafenamide regimen was found to be noninferior to a tenofovir disoproxil fumarate-containing regimen, and 97 percent of the 959 patients who were assigned to elvitegravir-cobicistat-emtricitabine-tenofovir alafenamide had an HIV RNA <50 copies/mL.

Raltegravir — Raltegravir is approved for use in treatment-naïve and treatment-experienced patients with HIV as part of a combination ART regimen [59,60,116-118]. It is available in 400 mg and 600 mg tablets. A discussion of how to select an ART regimen is presented separately. (See "Selecting antiretroviral regimens for treatment-naïve persons with HIV-1: General approach".)

Raltegravir is generally administered as 400 mg twice daily (800 mg total daily dose). However, once daily dosing with two 600 mg tablets (1200 mg total daily dose) is an option for patients who are treatment naïve or who are virologically suppressed (eg, HIV RNA <50 copies/mL) on an initial regimen of raltegravir 400 mg twice daily. If raltegravir is coadministered with rifampin, it should be administered as 800 mg twice daily (1600 mg total daily dose) regardless of prior ART experience.

Although rare, side effects can be seen with raltegravir, and include nausea, dizziness, and headache. Rare cases of skeletal muscle toxicity and severe systemic cutaneous reaction resembling Stevens-Johnson syndrome have also been reported [119-121].

As with other INSTIs, clinical trial data have found that raltegravir, when used in combination with tenofovir disoproxil fumarate-emtricitabine, is among the most effective agents in suppressing the HIV viral load. Raltegravir has demonstrated superior viral suppression compared with efavirenz [59,60] and was found to be superior to first-line protease inhibitors (ie, boosted atazanavir and boosted darunavir) when combined tolerability and virologic efficacy endpoints were evaluated [122]. In addition, raltegravir achieves meaningful drug concentrations in cerebral spinal fluid and genital compartments [123,124].

One of the main disadvantages of raltegravir is its lower genetic barrier to resistance compared with dolutegravir and protease inhibitors (N155H is its signature mutation) [97,122]. As an example, in one study virologic resistance developed in 3 percent of patients receiving raltegravir versus ≤1.5 percent in those receiving a protease inhibitor [122]. In addition, raltegravir has been studied only in combination with tenofovir disoproxil fumarate-emtricitabine as the NRTI pair.

PROTEASE INHIBITORS (PIs) — Protease inhibitors (PIs) are typically administered in combination with a dual nucleoside combination; however, they can also be used as part of a nucleoside-sparing/limiting regimen. PIs should be administered with a boosting agent, either ritonavir or cobicistat. They can be used for patients who are treatment-naïve, and are often the preferred agent for patients failing their initial antiretroviral therapy (ART) regimen. (See 'Pharmacokinetic boosting' below and "Selecting antiretroviral regimens for treatment-naïve persons with HIV-1: General approach" and "Selecting an antiretroviral regimen for treatment-experienced patients with HIV who are failing therapy" and "Switching antiretroviral therapy for adults with HIV-1 and a suppressed viral load".)

Although boosted darunavir and boosted atazanavir are effective as initial therapy in combination with a nucleoside backbone, boosted darunavir is generally preferred since darunavir appears to be better tolerated [4,125,126]. Ritonavir-boosted darunavir, ritonavir-boosted atazanavir, or raltegravir given with tenofovir disoproxil fumarate-emtricitabine were compared in a randomized study of 1809 ART-naïve patients [122]. There was a 9 percent higher incidence of treatment discontinuation in patients who received boosted atazanavir compared with boosted darunavir (97.5% CI 5.5 to 12.9 percent). This difference was due in large part to clinical jaundice and hyperbilirubinemia associated with atazanavir.

PI class characteristics

Mechanism of action – Protease inhibitors (PIs) competitively inhibit the cleavage of the Gag-Pol polyproteins in HIV-infected cells, which is a crucial step in the viral maturation process, thereby resulting in the production of immature virions that are not infectious. (See 'Overview of HIV replication' above.)

Spectrum of activity – PIs are active against HIV-1 and HIV-2.

Resistance – PIs have a relatively high genetic barrier to resistance compared with non-nucleoside reverse transcriptase inhibitors (NNRTIs) and the integrase inhibitors (INSTIs) raltegravir and elvitegravir. The development of resistance to pharmacologically boosted PIs in previously antiretroviral-naïve individuals is uncommon, even with less than optimal adherence [127]. In clinical trials evaluating the frequency of PI resistance in patients with virologic failure on their initial ART regimen, protease resistance developed in 0 of 76 cases with lopinavir/ritonavir, 0 of 140 cases with atazanavir/ritonavir, and 0 of 46 cases with darunavir/ritonavir. Thus, protease inhibitor-based regimens are a good choice for patients in whom adherence is a concern. (See "Interpretation of HIV drug resistance testing", section on 'Protease inhibitors' and "Selecting an antiretroviral regimen for treatment-experienced patients with HIV who are failing therapy", section on 'Patients without drug-resistant virus'.)

Adverse events – There are several side effects that appear to be PI class effects while others are agent-specific. Some of the class side effects are insulin resistance, hyperglycemia, diabetes, hyperlipidemia, lipodystrophy, hepatotoxicity, bleeding in patients with hemophilia, and PR interval prolongation [128-130]. Adverse reactions can also be a result of drug interactions with other hepatically metabolized drugs [128,131]. Specific adverse reactions related to the commonly used agents are described below. (See 'Darunavir' below and 'Atazanavir' below and 'Other protease inhibitors' below.)

Drug interactions – It is important to carefully check for potential drug interactions between the PI being considered and other concomitant medications [132]. Under-recognition of these interactions may result in loss of therapeutic effect or excess toxicity. Some studies have demonstrated that unsuspected drug-drug interactions were more common among HIV-infected patients taking PIs compared with those taking antiretroviral medications within other drug classes [132]. Mechanistically, most drug interactions with PIs arise from the pharmacological boosting agents. (See 'Pharmacokinetic boosting' below.)

For detailed drug interaction information, see the United States Department of Health and Human Services (DHHS) Antiretroviral Guidelines for Adults and Adolescents (tables 17, 18, and 19) and the Lexicomp drug interactions program within UpToDate.

Specific agents (listed alphabetically)

Atazanavir — Atazanavir should be administered as atazanavir 300 mg once daily plus ritonavir 100 mg once daily or atazanavir 300 mg once daily plus cobicistat 150 mg once daily. Atazanavir and cobicistat are coformulated as a single tablet (table 1). When atazanavir is administered with cobicistat, it should be avoided in patients with an eGFR <70 mL/min/1.73 m2 if tenofovir disoproxil fumarate-emtricitabine is used as the nucleoside combination. Although the US Food and Drug Administration (FDA) has approved unboosted atazanavir at 400 mg (two 200 mg capsules) once a day, unboosted atazanavir is not recommended except in select situations [4,5].

Atazanavir has the advantage of better gastrointestinal tolerance than most other PIs, once-daily dosing, and good potency. Atazanavir is not associated with the development of insulin resistance when compared with some other PIs such as lopinavir-ritonavir. Lipid profiles show mild increases in cholesterol and triglycerides that are mainly related to ritonavir [133].

Disadvantages with atazanavir include the need for gastric acid and food for optimal absorption; thus, concomitant use of drugs that alter stomach pH (eg, proton pump inhibitors) may impair absorption of the drug. Atazanavir should not be used in patients who require a proton pump inhibitor dose equivalent to >20 mg omeprazole per day.

Atazanavir has been associated with the development of renal stones and kidney injury (eg, proximal tubular dysfunction, interstitial nephritis, and acute and chronic kidney injury) [32,134-139]. (See "Overview of kidney disease in patients with HIV", section on 'Causes of AKI in patients with HIV'.)

Adverse reactions also include an increase in the indirect serum bilirubin concentration, which is benign but may cause jaundice. However, in antiretroviral-naïve persons with low cardiovascular risk, the hyperbilirubinemia associated with atazanavir plus ritonavir may be responsible for slower progression of carotid artery intima-media thickening compared with those who received darunavir boosted with ritonavir [140]. In addition, unlike other PIs, atazanavir is not associated with an increased risk of cardiovascular disease [141-143]. (See "Epidemiology of cardiovascular disease and risk factors in patients with HIV".)

Darunavir — Dosing of darunavir for treatment-naïve patients is 800 mg once daily plus ritonavir 100 mg once daily or darunavir 800 mg once daily plus cobicistat 150 mg once daily. Darunavir is available in two single coformulated tablets, darunavir-cobicistat and darunavir-cobicistat-emtricitabine-tenofovir alafenamide (table 1).

For patients with drug-resistant virus, the dose of darunavir depends upon which drug resistance mutations are present.

We administer darunavir twice daily (600 mg darunavir plus 100 mg ritonavir) if any of the following darunavir resistance mutations are present: V11I, V32I, L33F, I47V, I50V, I54L/M, T74P, L76V, I84V, and L89V). Cobicistat should not be used as a boosting agent for such patients who require twice-daily dosing of darunavir.

Darunavir can be given once daily (800 mg darunavir with 100 mg ritonavir or 150 mg cobicistat) if there are no darunavir mutations, even if there is resistance to other agents [144].

Darunavir appears to be the best tolerated PI in randomized studies [122,126]. The principal adverse effects associated with darunavir include nausea, diarrhea, increased transaminases, headache, and rash. However, severe reactions can occur, and darunavir should be discontinued in patients who develop a rash associated with fever, transaminase elevations, eosinophilia, and/or systemic symptoms, as well as those who have severe skin reactions (eg, Stevens-Johnson syndrome, toxic epidermal necrolysis, exanthematous pustulosis) [145-149].

Darunavir, whether given with ritonavir or cobicistat, should not be used in patients with severe liver disease, as infrequent cases of drug-induced liver injury have been reported. In addition, we generally avoid administering darunavir to patients with a sulfonamide allergy, especially those with a severe allergy (eg, rash with systemic symptoms, immediate-type allergy, Stevens-Johnson syndrome) since darunavir contains a sulfonamide moiety. Although most patients with a sulfonamide allergy are able to tolerate darunavir (it lacks one or both essential functional groups implicated in sulfonamide antibiotic hypersensitivity), we prefer using a different third drug for such patients if possible, given the availability of multiple effective alternatives.

Other protease inhibitors

Lopinavir – Lopinavir boosted with ritonavir is not typically recommended because of issues related to potency, toxicity, and convenience [4,5,125,126]. However, when it is used, a tenofovir-containing nucleoside reverse transcriptase inhibitor (NRTI) combination is preferred.

Lopinavir-ritonavir can also be given with lamivudine alone for patients who are unable to tolerate a tenofovir or abacavir-containing backbone [4]. In a randomized study of 426 patients, individuals received lopinavir-ritonavir in combination with lamivudine (dual therapy) or tenofovir disoproxil fumarate-emtricitabine (triple therapy). After 48 weeks of treatment, the number of patients who achieved a viral load <50 copies/mL was similar between the two groups (88 versus 83 percent) [150]. However, a nucleoside-limiting regimen such as lopinavir-ritonavir plus lamivudine may not be suitable for all patients. (See "Selecting antiretroviral regimens for treatment-naïve persons with HIV-1: General approach", section on 'Use of two-drug regimens in select patients'.)

PIs rarely used – Several older PIs that were commonly used are either no longer administered or are rarely used because of poor efficacy and/or toxicity. Such agents include indinavir, fosamprenavir, nelfinavir, saquinavir, and tipranavir. As an example, although tipranavir is active against strains of HIV that are resistant to other protease inhibitors, it is avoided when possible since it has high rates of hepatotoxicity and has been associated with intracranial hemorrhage [151-154]. (See "Selecting an antiretroviral regimen for treatment-experienced patients with HIV who are failing therapy", section on 'Protease inhibitors'.)

POST-ATTACHMENT INHIBITOR — Ibalizumab is a monoclonal antibody that is given as an intravenous infusion every two weeks and binds the CD4 molecule in such a way that it does not block viral attachment, but does block entry [155]. It is indicated for the treatment of heavily antiretroviral-experienced HIV-1-infected adults with multidrug-resistant virus who are failing their current regimen. It should be used as part of a regimen with other potentially active agents whenever possible. (See "Selecting an antiretroviral regimen for treatment-experienced patients with HIV who are failing therapy", section on 'If a fully active PI is NOT available'.)

SPECIAL CONSIDERATIONS

Pharmacokinetic boosting — Most protease inhibitors (PIs), as well as the integrase inhibitor elvitegravir, are administered in combination with another agent (eg, low-dose ritonavir or cobicistat) to increase trough plasma drug concentrations, increase drug half-lives, and increase maximum plasma concentrations (CMAX). This improves the potency of the antiviral agent and enables lower and less frequent dosing of the parent drug, thereby decreasing pill burden.

For PIs, pharmacokinetic boosting is done in combination with cobicistat or low-dose ritonavir. Although ritonavir is also a PI, it is not used as an antiviral agent when administered at a low dose but rather as a boosting agent for other PIs. Both cobicistat and ritonavir inhibit the hepatic microsomal enzyme CYP (450) 3A4, a characteristic used to "boost" the concentration of the coadministered PI.

Ritonavir boosting is required for darunavir and lopinavir, and is preferred for atazanavir. As a boosting agent for darunavir and atazanavir, ritonavir is dosed as 100 mg daily; for lopinavir, ritonavir is dosed at 200 mg per day. Ritonavir is available as a tablet, gel cap, and oral solution. Tablets and oral solution do not require refrigeration. The gel capsules should be refrigerated but may be left at room temperature (77°F) for up to 30 days. Full-dose ritonavir (600 mg twice daily) is never used to treat HIV infection because of significant adverse effects at this dose. The major drawbacks of using ritonavir boosting are related to adverse drug interactions, an increased risk of gastrointestinal intolerance, and hyperlipidemia. All of these adverse effects are dose related.

Cobicistat is chemically similar to ritonavir, but has no antiviral activity. Overall, it is well tolerated, but it has been associated with modest increases in serum creatinine (it inhibits tubular secretion of creatinine), and has gastrointestinal toxicity, lipid changes, and drug interactions comparable with ritonavir. It is also used to boost the integrase inhibitor, elvitegravir, and the protease inhibitors, atazanavir and darunavir. Coformulated tablets of cobicistat with these agents are available. Cobicistat-boosted regimens should not be used in females of childbearing age who are not committed to effective birth control and in pregnant females due to decreased drug levels during pregnancy. (See "HIV and women", section on 'Selection of antiretroviral regimens'.)

Patients who have trouble swallowing pills — Certain patients may have trouble taking combination regimens if they have difficulty swallowing due to other comorbidities, such as candidal esophagitis, or more permanent disabilities, such as stroke. Fortunately, many antiretroviral medications are available as liquid preparations; in addition, for some medications, there are methods for dissolving or crushing tablets without losing drug potency [156,157]. The milligram dosing of a liquid preparation may differ from the milligram dose of a tablet formulation, so a direct conversion should not be expected. In addition, some chewable, powder, and liquid formulations are not approved for or conducive to administration to adults. Antiretroviral agents for patients who have trouble swallowing pills are described in the table (table 3).

Timing of doses for patients on hemodialysis — For patients undergoing hemodialysis, we schedule medications so that at least one of the daily ART doses is taken soon after dialysis on dialysis days. As an example, bictegravir-emtricitabine-tenofovir alafenamide, emtricitabine, lamivudine, tenofovir alafenamide, tenofovir disoproxil fumarate, and zidovudine specifically be taken after dialysis on dialysis days [4,158]. Although some antiretroviral agents are probably not dialyzed, the logic behind dosing after dialysis is to guarantee therapeutic plasma concentrations in case dialysis removes some of the drug or an important metabolite.

More detailed information on the use of specific antiretroviral agents in the setting of dialysis can be found in the Lexicomp drug monographs included with UpToDate and in the DHHS guidelines [4].

SOCIETY GUIDELINE LINKS — Links to society and government-sponsored guidelines from selected countries and regions around the world are provided separately. (See "Society guideline links: HIV treatment in nonpregnant adults and adolescents".)

SUMMARY AND RECOMMENDATIONS

The life cycle of HIV can be broken down into 6 steps: (1) entry (binding and fusion), (2) reverse transcription, (3) integration, (4) replication (transcription and translation), (5) assembly, and (6) budding and maturation. The identification and understanding of these processes have provided the basis for antiretroviral agents used to treat HIV. (See 'Overview of HIV replication' above.)

Maraviroc and enfuvirtide are antiretroviral agents that inhibit binding and fusion, respectively. However, these agents are not commonly used for the treatment of HIV infection. (See 'Entry inhibitors' above.)

Nucleoside reverse transcriptase inhibitors (NRTIs) are often referred to as the "backbone" of antiretroviral therapy (ART) regimens and are usually given in pairs. The commonly used combinations are available as coformulations (tenofovir alafenamide-emtricitabine, tenofovir disoproxil fumarate-emtricitabine, abacavir-lamivudine). (See 'Nucleoside/nucleotide reverse transcriptase inhibitors (NRTIs)' above.)

The non-nucleoside reverse transcriptase inhibitors (NNRTIs) are typically administered with an NRTI combination. For treatment-naïve patients, options include efavirenz, rilpivirine, and doravirine. Etravirine is generally used for individuals with evidence of drug-resistant virus. (See 'Non-nucleoside reverse transcriptase inhibitors (NNRTIs)' above.)

Regimens using integrase strand transfer inhibitors (INSTIs; eg, bictegravir, dolutegravir, elvitegravir, and raltegravir) are effective and well tolerated. In many countries, including the United States, INSTIs are considered the preferred third agent for treatment-naïve individuals in combination with two nucleoside analogues. In addition, a once-monthly, injectable regimen (cabotegravir-rilpivirine) is available as switch therapy for treatment-experienced patients who are virologically suppressed on an oral regimen. (See 'Integrase strand transfer inhibitors (INSTIs)' above.)

Protease inhibitors (PIs) are typically administered with an NRTI combination; however, they can also be used as part of a nucleoside-sparing/limiting regimen. PIs should be administered with a boosting agent such as ritonavir or cobicistat. They can be used for patients who are treatment-naïve, and are often the preferred agent for patients failing their initial ART regimen. (See 'Protease inhibitors (PIs)' above and 'Pharmacokinetic boosting' above.)

Certain patients may have trouble taking combination regimens if they have difficulty swallowing due to other comorbidities, such as candidal esophagitis, or more permanent disabilities, such as stroke. For such patients, alternative antiretroviral agents may be available (table 3). (See 'Patients who have trouble swallowing pills' above.)

ACKNOWLEDGMENTS — The UpToDate editorial staff acknowledges Amy Graziani, PharmD, and Charles Hicks, MD, who contributed to an earlier version of this topic review.

We are saddened by the death of John G Bartlett, MD, who passed away in January 2021. UpToDate gratefully acknowledges Dr. Bartlett's role as section editor on this topic, his tenure as the founding Editor-in-Chief for UpToDate in Infectious Diseases, and his dedicated and longstanding involvement with the UpToDate program.

  1. US Food and Drug Administration. FDA Approves New HIV Treatment for Patients With Limited Treatment Options https://www.fda.gov/news-events/press-announcements/fda-approves-new-hiv-treatment-patients-limited-treatment-options (Accessed on August 06, 2020).
  2. Kozal M, Aberg J, Pialoux G, et al. Fostemsavir in Adults with Multidrug-Resistant HIV-1 Infection. N Engl J Med 2020; 382:1232.
  3. Lataillade JM et al. Week 96 safety and efficacy of the novel HIV-1 attachment inhibitor prodrug fostemsavir in heavily treatment-experienced participants infected with multi-drug resistant HIV-1 (BRIGHTE study). 10th International AIDS Society Conference on HIV Science, Mexico City, abstract MOAB0102, 2019. https://onlinelibrary.wiley.com/doi/full/10.1002/jia2.25327 (Accessed on September 25, 2020).
  4. Panel on Antiretroviral Guidelines for Adults and Adolescents. Guidelines for the use of antiretroviral agents in HIV-1-infected adults and adolescents. Department of Health and Human Services. Available at https://aidsinfo.nih.gov/contentfiles/lvguidelines/adultandadolescentgl.pdf (Accessed on March 03, 2021).
  5. Saag MS, Gandhi RT, Hoy JF, et al. Antiretroviral Drugs for Treatment and Prevention of HIV Infection in Adults: 2020 Recommendations of the International Antiviral Society-USA Panel. JAMA 2020; 324:1651.
  6. Anderson PL, Kakuda TN, Kawle S, Fletcher CV. Antiviral dynamics and sex differences of zidovudine and lamivudine triphosphate concentrations in HIV-infected individuals. AIDS 2003; 17:2159.
  7. Kakuda TN. Pharmacology of nucleoside and nucleotide reverse transcriptase inhibitor-induced mitochondrial toxicity. Clin Ther 2000; 22:685.
  8. Boubaker K, Flepp M, Sudre P, et al. Hyperlactatemia and antiretroviral therapy: the Swiss HIV Cohort Study. Clin Infect Dis 2001; 33:1931.
  9. Coghlan ME, Sommadossi JP, Jhala NC, et al. Symptomatic lactic acidosis in hospitalized antiretroviral-treated patients with human immunodeficiency virus infection: a report of 12 cases. Clin Infect Dis 2001; 33:1914.
  10. Robbins GK, De Gruttola V, Shafer RW, et al. Comparison of sequential three-drug regimens as initial therapy for HIV-1 infection. N Engl J Med 2003; 349:2293.
  11. Sax PE, Tierney C, Collier AC, et al. Abacavir/lamivudine versus tenofovir DF/emtricitabine as part of combination regimens for initial treatment of HIV: final results. J Infect Dis 2011; 204:1191.
  12. Sax PE, Tierney C, Collier AC, et al. Abacavir-lamivudine versus tenofovir-emtricitabine for initial HIV-1 therapy. N Engl J Med 2009; 361:2230.
  13. Strategies for Management of Anti-Retroviral Therapy/INSIGHT, DAD Study Groups. Use of nucleoside reverse transcriptase inhibitors and risk of myocardial infarction in HIV-infected patients. AIDS 2008; 22:F17.
  14. D:A:D Study Group, Sabin CA, Worm SW, et al. Use of nucleoside reverse transcriptase inhibitors and risk of myocardial infarction in HIV-infected patients enrolled in the D:A:D study: a multi-cohort collaboration. Lancet 2008; 371:1417.
  15. Marcus JL, Neugebauer RS, Leyden WA, et al. Use of Abacavir and Risk of Cardiovascular Disease Among HIV-Infected Individuals. J Acquir Immune Defic Syndr 2016; 71:413.
  16. Campo R, DeJesus E, Bredeek UF, et al. SWIFT: prospective 48-week study to evaluate efficacy and safety of switching to emtricitabine/tenofovir from lamivudine/abacavir in virologically suppressed HIV-1 infected patients on a boosted protease inhibitor containing antiretroviral regimen. Clin Infect Dis 2013; 56:1637.
  17. Durand M, Sheehy O, Baril JG, et al. Association between HIV infection, antiretroviral therapy, and risk of acute myocardial infarction: a cohort and nested case-control study using Québec's public health insurance database. J Acquir Immune Defic Syndr 2011; 57:245.
  18. Choi AI, Vittinghoff E, Deeks SG, et al. Cardiovascular risks associated with abacavir and tenofovir exposure in HIV-infected persons. AIDS 2011; 25:1289.
  19. Ribaudo HJ, Benson CA, Zheng Y, et al. No risk of myocardial infarction associated with initial antiretroviral treatment containing abacavir: short and long-term results from ACTG A5001/ALLRT. Clin Infect Dis 2011; 52:929.
  20. Walmsley SL, Antela A, Clumeck N, et al. Dolutegravir plus abacavir-lamivudine for the treatment of HIV-1 infection. N Engl J Med 2013; 369:1807.
  21. Smith KY, Patel P, Fine D, et al. Randomized, double-blind, placebo-matched, multicenter trial of abacavir/lamivudine or tenofovir/emtricitabine with lopinavir/ritonavir for initial HIV treatment. AIDS 2009; 23:1547.
  22. Bansi L, Sabin C, Gilson R, et al. Virological response to initial antiretroviral regimens containing abacavir or tenofovir. J Infect Dis 2009; 200:710.
  23. Holdich T, Shiveley LA, Sawyer J. Effect of Lamivudine on the plasma and intracellular pharmacokinetics of apricitabine, a novel nucleoside reverse transcriptase inhibitor, in healthy volunteers. Antimicrob Agents Chemother 2007; 51:2943.
  24. GlaxoSmithKline. Epivir (lamivudine) tablets and oral solution. Research Triangle Park, NC. 2008.
  25. Gallant JE, DeJesus E, Arribas JR, et al. Tenofovir DF, emtricitabine, and efavirenz vs. zidovudine, lamivudine, and efavirenz for HIV. N Engl J Med 2006; 354:251.
  26. Ruane PJ, DeJesus E, Berger D, et al. Antiviral activity, safety, and pharmacokinetics/pharmacodynamics of tenofovir alafenamide as 10-day monotherapy in HIV-1-positive adults. J Acquir Immune Defic Syndr 2013; 63:449.
  27. Sax PE, Wohl D, Yin MT, et al. Tenofovir alafenamide versus tenofovir disoproxil fumarate, coformulated with elvitegravir, cobicistat, and emtricitabine, for initial treatment of HIV-1 infection: two randomised, double-blind, phase 3, non-inferiority trials. Lancet 2015; 385:2606.
  28. Schafer JJ, Sassa KN, O'Connor JR, et al. Changes in Body Mass Index and Atherosclerotic Disease Risk Score After Switching From Tenofovir Disoproxil Fumarate to Tenofovir Alafenamide. Open Forum Infect Dis 2019; 6:ofz414.
  29. Gomez M, Seybold U, Roider J, et al. A retrospective analysis of weight changes in HIV-positive patients switching from a tenofovir disoproxil fumarate (TDF)- to a tenofovir alafenamide fumarate (TAF)-containing treatment regimen in one German university hospital in 2015-2017. Infection 2019; 47:95.
  30. Sax PE, Erlandson KM, Lake JE, et al. Weight Gain Following Initiation of Antiretroviral Therapy: Risk Factors in Randomized Comparative Clinical Trials. Clin Infect Dis 2020; 71:1379.
  31. Zimmermann AE, Pizzoferrato T, Bedford J, et al. Tenofovir-associated acute and chronic kidney disease: a case of multiple drug interactions. Clin Infect Dis 2006; 42:283.
  32. Lucas GM, Ross MJ, Stock PG, et al. Clinical practice guideline for the management of chronic kidney disease in patients infected with HIV: 2014 update by the HIV Medicine Association of the Infectious Diseases Society of America. Clin Infect Dis 2014; 59:e96.
  33. Tourret J, Deray G, Isnard-Bagnis C. Tenofovir effect on the kidneys of HIV-infected patients: a double-edged sword? J Am Soc Nephrol 2013; 24:1519.
  34. Huang JS, Hughes MD, Riddler SA, et al. Bone mineral density effects of randomized regimen and nucleoside reverse transcriptase inhibitor selection from ACTG A5142. HIV Clin Trials 2013; 14:224.
  35. Yin MT, Overton ET. Increasing clarity on bone loss associated with antiretroviral initiation. J Infect Dis 2011; 203:1705.
  36. McComsey GA, Kitch D, Daar ES, et al. Bone mineral density and fractures in antiretroviral-naive persons randomized to receive abacavir-lamivudine or tenofovir disoproxil fumarate-emtricitabine along with efavirenz or atazanavir-ritonavir: Aids Clinical Trials Group A5224s, a substudy of ACTG A5202. J Infect Dis 2011; 203:1791.
  37. Arribas JR, Pozniak AL, Gallant JE, et al. Tenofovir disoproxil fumarate, emtricitabine, and efavirenz compared with zidovudine/lamivudine and efavirenz in treatment-naive patients: 144-week analysis. J Acquir Immune Defic Syndr 2008; 47:74.
  38. Pozniak AL, Gallant JE, DeJesus E, et al. Tenofovir disoproxil fumarate, emtricitabine, and efavirenz versus fixed-dose zidovudine/lamivudine and efavirenz in antiretroviral-naive patients: virologic, immunologic, and morphologic changes--a 96-week analysis. J Acquir Immune Defic Syndr 2006; 43:535.
  39. Post FA, Moyle GJ, Stellbrink HJ, et al. Randomized comparison of renal effects, efficacy, and safety with once-daily abacavir/lamivudine versus tenofovir/emtricitabine, administered with efavirenz, in antiretroviral-naive, HIV-1-infected adults: 48-week results from the ASSERT study. J Acquir Immune Defic Syndr 2010; 55:49.
  40. Flynn PM, Rodman J, Lindsey JC, et al. Intracellular pharmacokinetics of once versus twice daily zidovudine and lamivudine in adolescents. Antimicrob Agents Chemother 2007; 51:3516.
  41. Ruane PJ, Richmond GJ, DeJesus E, et al. Pharmacodynamic effects of zidovudine 600 mg once/day versus 300 mg twice/day in therapy-naïve patients infected with human immunodeficiency virus. Pharmacotherapy 2004; 24:307.
  42. Berenguer J, González J, Ribera E, et al. Didanosine, lamivudine, and efavirenz versus zidovudine, lamivudine, and efavirenz for the initial treatment of HIV type 1 infection: final analysis (48 weeks) of a prospective, randomized, noninferiority clinical trial, GESIDA 3903. Clin Infect Dis 2008; 47:1083.
  43. Grobler JA, Dornadula G, Rice MR, et al. HIV-1 reverse transcriptase plus-strand initiation exhibits preferential sensitivity to non-nucleoside reverse transcriptase inhibitors in vitro. J Biol Chem 2007; 282:8005.
  44. Zdanowicz MM. The pharmacology of HIV drug resistance. Am J Pharm Educ 2006; 70:100.
  45. Anta L, Llibre JM, Poveda E, et al. Rilpivirine resistance mutations in HIV patients failing non-nucleoside reverse transcriptase inhibitor-based therapies. AIDS 2013; 27:81.
  46. Hare CB, Mellors J, Krambrink A, et al. Detection of nonnucleoside reverse-transcriptase inhibitor-resistant HIV-1 after discontinuation of virologically suppressive antiretroviral therapy. Clin Infect Dis 2008; 47:421.
  47. Kilkaire B, Walker S, Khoo S, et al. Plasma levels of nevirapine following interruption of ZDV/3Tc/nvp in African adults within the DART trial. Program and abstracts of the XVI International AIDS Conference; August 13-18, 2006; Toronto, Ontario, Canada. Abstract WEAB0201.
  48. Orkin C, Squires KE, Molina JM, et al. Doravirine/Lamivudine/Tenofovir Disoproxil Fumarate is Non-inferior to Efavirenz/Emtricitabine/Tenofovir Disoproxil Fumarate in Treatment-naive Adults With Human Immunodeficiency Virus-1 Infection: Week 48 Results of the DRIVE-AHEAD Trial. Clin Infect Dis 2019; 68:535.
  49. Molina JM, Squires K, Sax PE, et al. Doravirine versus ritonavir-boosted darunavir in antiretroviral-naive adults with HIV-1 (DRIVE-FORWARD): 48-week results of a randomised, double-blind, phase 3, non-inferiority trial. Lancet HIV 2018; 5:e211.
  50. Shafer RW, Smeaton LM, Robbins GK, et al. Comparison of four-drug regimens and pairs of sequential three-drug regimens as initial therapy for HIV-1 infection. N Engl J Med 2003; 349:2304.
  51. van Leth F, Phanuphak P, Ruxrungtham K, et al. Comparison of first-line antiretroviral therapy with regimens including nevirapine, efavirenz, or both drugs, plus stavudine and lamivudine: a randomised open-label trial, the 2NN Study. Lancet 2004; 363:1253.
  52. Squires K, Lazzarin A, Gatell JM, et al. Comparison of once-daily atazanavir with efavirenz, each in combination with fixed-dose zidovudine and lamivudine, as initial therapy for patients infected with HIV. J Acquir Immune Defic Syndr 2004; 36:1011.
  53. Staszewski S, Morales-Ramirez J, Tashima KT, et al. Efavirenz plus zidovudine and lamivudine, efavirenz plus indinavir, and indinavir plus zidovudine and lamivudine in the treatment of HIV-1 infection in adults. Study 006 Team. N Engl J Med 1999; 341:1865.
  54. Lucas GM, Chaisson RE, Moore RD. Comparison of initial combination antiretroviral therapy with a single protease inhibitor, ritonavir and saquinavir, or efavirenz. AIDS 2001; 15:1679.
  55. Montaner JS, Saag MS, Barylski C, Siemon-Hryczyk P. FOCUS study: saquinavir QD reigmen versus efavirenz QD regimen 48 week analysis in HIV infected patients. In: 42nd Interscience Conference on Antimicrobial Agents and Chemotherapy; 2002; San Diego. (Abstract).
  56. Bartlett JA, Johnson J, Herrera G, et al. Abacavir/lamivudine (ABC/3TC) in combination with efavirenz (NNRTI), amprenavir/ritonavir (PI) or stavudine (NRTI): ESS40001 (CLASS) preliminary 48 week results. In 14th International AIDS Conference; 2002; Barcelona. (Abstract).
  57. Ribaudo HJ, Kuritzkes DR, Lalama CM, et al. Efavirenz-based regimens in treatment-naive patients with a range of pretreatment HIV-1 RNA levels and CD4 cell counts. J Infect Dis 2008; 197:1006.
  58. Sax PE, DeJesus E, Mills A, et al. Co-formulated elvitegravir, cobicistat, emtricitabine, and tenofovir versus co-formulated efavirenz, emtricitabine, and tenofovir for initial treatment of HIV-1 infection: a randomised, double-blind, phase 3 trial, analysis of results after 48 weeks. Lancet 2012; 379:2439.
  59. Rockstroh JK, DeJesus E, Lennox JL, et al. Durable efficacy and safety of raltegravir versus efavirenz when combined with tenofovir/emtricitabine in treatment-naive HIV-1-infected patients: final 5-year results from STARTMRK. J Acquir Immune Defic Syndr 2013; 63:77.
  60. Lennox JL, DeJesus E, Lazzarin A, et al. Safety and efficacy of raltegravir-based versus efavirenz-based combination therapy in treatment-naive patients with HIV-1 infection: a multicentre, double-blind randomised controlled trial. Lancet 2009; 374:796.
  61. Rihs TA, Begley K, Smith DE, et al. Efavirenz and chronic neuropsychiatric symptoms: a cross-sectional case control study. HIV Med 2006; 7:544.
  62. Leutscher PD, Stecher C, Storgaard M, Larsen CS. Discontinuation of efavirenz therapy in HIV patients due to neuropsychiatric adverse effects. Scand J Infect Dis 2013; 45:645.
  63. Mollan KR, Smurzynski M, Eron JJ, et al. Association between efavirenz as initial therapy for HIV-1 infection and increased risk for suicidal ideation or attempted or completed suicide: an analysis of trial data. Ann Intern Med 2014; 161:1.
  64. Clifford DB, Evans S, Yang Y, et al. Impact of efavirenz on neuropsychological performance and symptoms in HIV-infected individuals. Ann Intern Med 2005; 143:714.
  65. Mollan KR, Tierney C, Hellwege JN, et al. Race/Ethnicity and the Pharmacogenetics of Reported Suicidality With Efavirenz Among Clinical Trials Participants. J Infect Dis 2017; 216:554.
  66. Chang JL, Tsai AC, Musinguzi N, et al. Depression and Suicidal Ideation Among HIV-Infected Adults Receiving Efavirenz Versus Nevirapine in Uganda: A Prospective Cohort Study. Ann Intern Med 2018; 169:146.
  67. ENCORE1 Study Group, Carey D, Puls R, et al. Efficacy and safety of efavirenz 400 mg daily versus 600 mg daily: 96-week data from the randomised, double-blind, placebo-controlled, non-inferiority ENCORE1 study. Lancet Infect Dis 2015; 15:793.
  68. Lamorde M, Wang X, Neary M, et al. Pharmacokinetics, Pharmacodynamics, and Pharmacogenetics of Efavirenz 400 mg Once Daily During Pregnancy and Post-Partum. Clin Infect Dis 2018; 67:785.
  69. NAMSAL ANRS 12313 Study Group, Kouanfack C, Mpoudi-Etame M, et al. Dolutegravir-Based or Low-Dose Efavirenz-Based Regimen for the Treatment of HIV-1. N Engl J Med 2019; 381:816.
  70. Johnson LB, Saravolatz LD. Etravirine, a next-generation nonnucleoside reverse-transcriptase inhibitor. Clin Infect Dis 2009; 48:1123.
  71. Sanne I, Mommeja-Marin H, Hinkle J, et al. Severe hepatotoxicity associated with nevirapine use in HIV-infected subjects. J Infect Dis 2005; 191:825.
  72. Stern JO, Robinson PA, Love J, et al. A comprehensive hepatic safety analysis of nevirapine in different populations of HIV infected patients. J Acquir Immune Defic Syndr 2003; 34 Suppl 1:S21.
  73. Kesselring AM, Wit FW, Sabin CA, et al. Risk factors for treatment-limiting toxicities in patients starting nevirapine-containing antiretroviral therapy. AIDS 2009; 23:1689.
  74. Chantarangsu S, Mushiroda T, Mahasirimongkol S, et al. Genome-wide association study identifies variations in 6p21.3 associated with nevirapine-induced rash. Clin Infect Dis 2011; 53:341.
  75. Aaron E, Kempf MC, Criniti S, et al. Adverse events in a cohort of HIV infected pregnant and non-pregnant women treated with nevirapine versus non-nevirapine antiretroviral medication. PLoS One 2010; 5:e12617.
  76. van Leth F, Andrews S, Grinsztejn B, et al. The effect of baseline CD4 cell count and HIV-1 viral load on the efficacy and safety of nevirapine or efavirenz-based first-line HAART. AIDS 2005; 19:463.
  77. Carr DF, Chaponda M, Jorgensen AL, et al. Association of human leukocyte antigen alleles and nevirapine hypersensitivity in a Malawian HIV-infected population. Clin Infect Dis 2013; 56:1330.
  78. Dodi F, Alessandrini A, Camera M, et al. Stevens-Johnson syndrome in HIV patients treated with nevirapine: two case reports. AIDS 2002; 16:1197.
  79. Zhang C, Wang W, Zhou M, et al. The interaction of CD4 T-cell count and nevirapine hepatotoxicity in China: a change in national treatment guidelines may be warranted. J Acquir Immune Defic Syndr 2013; 62:540.
  80. Product labeling. Edurant (rilpivirine). Tibotec, May 2015.
  81. Pozniak AL, Morales-Ramirez J, Katabira E, et al. Efficacy and safety of TMC278 in antiretroviral-naive HIV-1 patients: week 96 results of a phase IIb randomized trial. AIDS 2010; 24:55.
  82. Cohen C, Wohl D, Arribas JR, et al. Week 48 results from a randomized clinical trial of rilpivirine/emtricitabine/tenofovir disoproxil fumarate vs. efavirenz/emtricitabine/tenofovir disoproxil fumarate in treatment-naive HIV-1-infected adults. AIDS 2014; 28:989.
  83. Schöller-Gyüre M, Kakuda TN, De Smedt G, et al. Effects of hepatic impairment on the steady-state pharmacokinetics of etravirine 200 mg BID: an open-label, multiple-dose, controlled Phase I study in adults. Clin Ther 2010; 32:328.
  84. Molina JM, Cahn P, Grinsztejn B, et al. Rilpivirine versus efavirenz with tenofovir and emtricitabine in treatment-naive adults infected with HIV-1 (ECHO): a phase 3 randomised double-blind active-controlled trial. Lancet 2011; 378:238.
  85. Cohen CJ, Andrade-Villanueva J, Clotet B, et al. Rilpivirine versus efavirenz with two background nucleoside or nucleotide reverse transcriptase inhibitors in treatment-naive adults infected with HIV-1 (THRIVE): a phase 3, randomised, non-inferiority trial. Lancet 2011; 378:229.
  86. Cohen CJ, Molina JM, Cassetti I, et al. Week 96 efficacy and safety of rilpivirine in treatment-naive, HIV-1 patients in two Phase III randomized trials. AIDS 2013; 27:939.
  87. Hazuda DJ, Felock P, Witmer M, et al. Inhibitors of strand transfer that prevent integration and inhibit HIV-1 replication in cells. Science 2000; 287:646.
  88. Hazuda DJ, Felock PJ, Hastings JC, et al. Differential divalent cation requirements uncouple the assembly and catalytic reactions of human immunodeficiency virus type 1 integrase. J Virol 1997; 71:7005.
  89. Yoder KE, Bushman FD. Repair of gaps in retroviral DNA integration intermediates. J Virol 2000; 74:11191.
  90. Zhu K, Dobard C, Chow SA. Requirement for integrase during reverse transcription of human immunodeficiency virus type 1 and the effect of cysteine mutations of integrase on its interactions with reverse transcriptase. J Virol 2004; 78:5045.
  91. Pommier Y, Johnson AA, Marchand C. Integrase inhibitors to treat HIV/AIDS. Nat Rev Drug Discov 2005; 4:236.
  92. Schafer JJ, Squires KE. Integrase inhibitors: a novel class of antiretroviral agents. Ann Pharmacother 2010; 44:145.
  93. Powderly WG. Integrase inhibitors in the treatment of HIV-1 infection. J Antimicrob Chemother 2010; 65:2485.
  94. Blanco JL, Varghese V, Rhee SY, et al. HIV-1 integrase inhibitor resistance and its clinical implications. J Infect Dis 2011; 203:1204.
  95. Garrido C, Villacian J, Zahonero N, et al. Broad phenotypic cross-resistance to elvitegravir in HIV-infected patients failing on raltegravir-containing regimens. Antimicrob Agents Chemother 2012; 56:2873.
  96. Stribild [package insert]. Foster City, CA: Gilead Sciences, Inc; August 2012
  97. Llibre JM, Pulido F, García F, et al. Genetic barrier to resistance for dolutegravir. AIDS Rev 2015; 17:56.
  98. Gutiérrez F, Fulladosa X, Barril G, Domingo P. Renal tubular transporter-mediated interactions of HIV drugs: implications for patient management. AIDS Rev 2014; 16:199.
  99. Norwood J, Turner M, Bofill C, et al. Brief Report: Weight Gain in Persons With HIV Switched From Efavirenz-Based to Integrase Strand Transfer Inhibitor-Based Regimens. J Acquir Immune Defic Syndr 2017; 76:527.
  100. Venter WDF, Moorhouse M, Sokhela S, et al. Dolutegravir plus Two Different Prodrugs of Tenofovir to Treat HIV. N Engl J Med 2019; 381:803.
  101. Bourgi K, Rebeiro PF, Turner M, et al. Greater Weight Gain in Treatment-naive Persons Starting Dolutegravir-based Antiretroviral Therapy. Clin Infect Dis 2020; 70:1267.
  102. Hoffmann C, Welz T, Sabranski M, et al. Higher rates of neuropsychiatric adverse events leading to dolutegravir discontinuation in women and older patients. HIV Med 2017; 18:56.
  103. Fettiplace A, Stainsby C, Winston A, et al. Psychiatric Symptoms in Patients Receiving Dolutegravir. J Acquir Immune Defic Syndr 2017; 74:423.
  104. Osterholzer DA, Goldman M. Dolutegravir: a next-generation integrase inhibitor for treatment of HIV infection. Clin Infect Dis 2014; 59:265.
  105. Oliveira M, Mesplède T, Quashie PK, et al. Resistance mutations against dolutegravir in HIV integrase impair the emergence of resistance against reverse transcriptase inhibitors. AIDS 2014; 28:813.
  106. van Lunzen J, Maggiolo F, Arribas JR, et al. Once daily dolutegravir (S/GSK1349572) in combination therapy in antiretroviral-naive adults with HIV: planned interim 48 week results from SPRING-1, a dose-ranging, randomised, phase 2b trial. Lancet Infect Dis 2012; 12:111.
  107. Eron JJ, Clotet B, Durant J, et al. Safety and efficacy of dolutegravir in treatment-experienced subjects with raltegravir-resistant HIV type 1 infection: 24-week results of the VIKING Study. J Infect Dis 2013; 207:740.
  108. Nichols G, et al. Phase 3 assessment of dolutegravir (DTG) 50mg twice daily in HIV-1-infected subjects with raltegravir (RAL) and/or elvitegravir (EVG) resistance in VIKING-3: week 24 results of all 183 patients enrolled. 7th IAS, Kuala Lumpur, 2013, Abstract TULBPE19
  109. Raffi F, Rachlis A, Stellbrink HJ, et al. Once-daily dolutegravir versus raltegravir in antiretroviral-naive adults with HIV-1 infection: 48 week results from the randomised, double-blind, non-inferiority SPRING-2 study. Lancet 2013; 381:735.
  110. Stellbrink HJ, Reynes J, Lazzarin A, et al. Dolutegravir in antiretroviral-naive adults with HIV-1: 96-week results from a randomized dose-ranging study. AIDS 2013; 27:1771.
  111. Cottrell ML, Hadzic T, Kashuba AD. Clinical pharmacokinetic, pharmacodynamic and drug-interaction profile of the integrase inhibitor dolutegravir. Clin Pharmacokinet 2013; 52:981.
  112. Gervasoni C, Riva A, Capetti A, et al. Increased dolutegravir exposure in HIV patients switched from ritonavir to cobicistat. Presented at CROI 2017. Seattle, Washington. Abstract #410.
  113. Clotet B, Feinberg J, van Lunzen J, et al. Once-daily dolutegravir versus darunavir plus ritonavir in antiretroviral-naive adults with HIV-1 infection (FLAMINGO): 48 week results from the randomised open-label phase 3b study. Lancet 2014; 383:2222.
  114. DeJesus E, Rockstroh JK, Henry K, et al. Co-formulated elvitegravir, cobicistat, emtricitabine, and tenofovir disoproxil fumarate versus ritonavir-boosted atazanavir plus co-formulated emtricitabine and tenofovir disoproxil fumarate for initial treatment of HIV-1 infection: a randomised, double-blind, phase 3, non-inferiority trial. Lancet 2012; 379:2429.
  115. Mills A, Arribas JR, Andrade-Villanueva J, et al. Switching from tenofovir disoproxil fumarate to tenofovir alafenamide in antiretroviral regimens for virologically suppressed adults with HIV-1 infection: a randomised, active-controlled, multicentre, open-label, phase 3, non-inferiority study. Lancet Infect Dis 2016; 16:43.
  116. Cooper DA, Steigbigel RT, Gatell JM, et al. Subgroup and resistance analyses of raltegravir for resistant HIV-1 infection. N Engl J Med 2008; 359:355.
  117. Steigbigel RT, Cooper DA, Kumar PN, et al. Raltegravir with optimized background therapy for resistant HIV-1 infection. N Engl J Med 2008; 359:339.
  118. Steigbigel RT, Cooper DA, Teppler H, et al. Long-term efficacy and safety of Raltegravir combined with optimized background therapy in treatment-experienced patients with drug-resistant HIV infection: week 96 results of the BENCHMRK 1 and 2 Phase III trials. Clin Infect Dis 2010; 50:605.
  119. Perry ME, Almaani N, Desai N, et al. Raltegravir-induced Drug Reaction with Eosinophilia and Systemic Symptoms (DRESS) syndrome - implications for clinical practice and patient safety. Int J STD AIDS 2013; 24:639.
  120. Lee FJ, Amin J, Bloch M, et al. Skeletal muscle toxicity associated with raltegravir-based combination antiretroviral therapy in HIV-infected adults. J Acquir Immune Defic Syndr 2013; 62:525.
  121. Thomas M, Hopkins C, Duffy E, et al. Association of the HLA-B*53:01 Allele With Drug Reaction With Eosinophilia and Systemic Symptoms (DRESS) Syndrome During Treatment of HIV Infection With Raltegravir. Clin Infect Dis 2017; 64:1198.
  122. Lennox JL, Landovitz RJ, Ribaudo HJ, et al. Efficacy and tolerability of 3 nonnucleoside reverse transcriptase inhibitor-sparing antiretroviral regimens for treatment-naive volunteers infected with HIV-1: a randomized, controlled equivalence trial. Ann Intern Med 2014; 161:461.
  123. Barau C, Delaugerre C, Braun J, et al. High concentration of raltegravir in semen of HIV-infected men: results from a substudy of the EASIER-ANRS 138 trial. Antimicrob Agents Chemother 2010; 54:937.
  124. Yilmaz A, Gisslén M, Spudich S, et al. Raltegravir cerebrospinal fluid concentrations in HIV-1 infection. PLoS One 2009; 4:e6877.
  125. Molina JM, Andrade-Villanueva J, Echevarria J, et al. Once-daily atazanavir/ritonavir versus twice-daily lopinavir/ritonavir, each in combination with tenofovir and emtricitabine, for management of antiretroviral-naive HIV-1-infected patients: 48 week efficacy and safety results of the CASTLE study. Lancet 2008; 372:646.
  126. Mills AM, Nelson M, Jayaweera D, et al. Once-daily darunavir/ritonavir vs. lopinavir/ritonavir in treatment-naive, HIV-1-infected patients: 96-week analysis. AIDS 2009; 23:1679.
  127. Gardner EM, Hullsiek KH, Telzak EE, et al. Antiretroviral medication adherence and class- specific resistance in a large prospective clinical trial. AIDS 2010; 24:395.
  128. Eastone JA, Decker CF. New-onset diabetes mellitus associated with use of protease inhibitor. Ann Intern Med 1997; 127:948.
  129. Tsiodras S, Perelas A, Wanke C, Mantzoros CS. The HIV-1/HAART associated metabolic syndrome - novel adipokines, molecular associations and therapeutic implications. J Infect 2010; 61:101.
  130. Soliman EZ, Lundgren JD, Roediger MP, et al. Boosted protease inhibitors and the electrocardiographic measures of QT and PR durations. AIDS 2011; 25:367.
  131. Visnegarwala F, Krause KL, Musher DM. Severe diabetes associated with protease inhibitor therapy. Ann Intern Med 1997; 127:947.
  132. Evans-Jones JG, Cottle LE, Back DJ, et al. Recognition of risk for clinically significant drug interactions among HIV-infected patients receiving antiretroviral therapy. Clin Infect Dis 2010; 50:1419.
  133. Squires KE, Young B, Dejesus E, et al. Similar efficacy and tolerability of atazanavir compared with atazanavir/ritonavir, each with abacavir/lamivudine after initial suppression with abacavir/lamivudine plus ritonavir-boosted atazanavir in HIV-infected patients. AIDS 2010; 24:2019.
  134. Ryom L, Mocroft A, Kirk O, et al. Association between antiretroviral exposure and renal impairment among HIV-positive persons with normal baseline renal function: the D:A:D study. J Infect Dis 2013; 207:1359.
  135. Ryom L, Mocroft A, Lundgren J. HIV therapies and the kidney: some good, some not so good? Curr HIV/AIDS Rep 2012; 9:111.
  136. Rodríguez-Nóvoa S, Martín-Carbonero L, Barreiro P, et al. Genetic factors influencing atazanavir plasma concentrations and the risk of severe hyperbilirubinemia. AIDS 2007; 21:41.
  137. Flexner C, Tierney C, Gross R, et al. Comparison of once-daily versus twice-daily combination antiretroviral therapy in treatment-naive patients: results of AIDS clinical trials group (ACTG) A5073, a 48-week randomized controlled trial. Clin Infect Dis 2010; 50:1041.
  138. Ribaudo HJ, Daar ES, Tierney C, et al. Impact of UGT1A1 Gilbert variant on discontinuation of ritonavir-boosted atazanavir in AIDS Clinical Trials Group Study A5202. J Infect Dis 2013; 207:420.
  139. Hamada Y, Nishijima T, Watanabe K, et al. High incidence of renal stones among HIV-infected patients on ritonavir-boosted atazanavir than in those receiving other protease inhibitor-containing antiretroviral therapy. Clin Infect Dis 2012; 55:1262.
  140. Stein JH, Ribaudo HJ, Hodis HN, et al. A prospective, randomized clinical trial of antiretroviral therapies on carotid wall thickness. AIDS 2015; 29:1775.
  141. Monforte Ad, Reiss P, Ryom L, et al. Atazanavir is not associated with an increased risk of cardio- or cerebrovascular disease events. AIDS 2013; 27:407.
  142. LaFleur J, Bress AP, Rosenblatt L, et al. Cardiovascular outcomes among HIV-infected veterans receiving atazanavir. AIDS 2017; 31:2095.
  143. Ryom L et al. Association between cardiovascular disease and contemporarily used protease inhibitors. Conference on Retroviruses and Opportunistic Infections (CROI 2017), Seattle, 2017.
  144. Cahn P, Fourie J, Grinsztejn B, et al. Week 48 analysis of once-daily vs. twice-daily darunavir/ritonavir in treatment-experienced HIV-1-infected patients. AIDS 2011; 25:929.
  145. Nishijima T, Gatanaga H, Teruya K, et al. Skin rash induced by ritonavir-boosted darunavir is common, but generally tolerable in an observational setting. J Infect Chemother 2014; 20:285.
  146. http://www.accessdata.fda.gov/drugsatfda_docs/label/2014/021976s034,202895s011lbl.pdf (Accessed on April 29, 2014).
  147. Vispo E. Warning on hepatotoxicity of darunavir. AIDS Rev 2008; 10:63.
  148. Currier J, Averitt Bridge D, Hagins D, et al. Sex-based outcomes of darunavir-ritonavir therapy: a single-group trial. Ann Intern Med 2010; 153:349.
  149. Introcaso CE, Hines JM, Kovarik CL. Cutaneous toxicities of antiretroviral therapy for HIV: part I. Lipodystrophy syndrome, nucleoside reverse transcriptase inhibitors, and protease inhibitors. J Am Acad Dermatol 2010; 63:549.
  150. Cahn P, Andrade-Villanueva J, Arribas JR, et al. Dual therapy with lopinavir and ritonavir plus lamivudine versus triple therapy with lopinavir and ritonavir plus two nucleoside reverse transcriptase inhibitors in antiretroviral-therapy-naive adults with HIV-1 infection: 48 week results of the randomised, open label, non-inferiority GARDEL trial. Lancet Infect Dis 2014; 14:572.
  151. Walmsley SL, Katlama C, Lazzarin A, et al. Pharmacokinetics, safety, and efficacy of tipranavir boosted with ritonavir alone or in combination with other boosted protease inhibitors as part of optimized combination antiretroviral therapy in highly treatment-experienced patients (BI Study 1182.51). J Acquir Immune Defic Syndr 2008; 47:429.
  152. Temesgen Z, Feinberg J. Tipranavir: a new option for the treatment of drug-resistant HIV infection. Clin Infect Dis 2007; 45:761.
  153. Cahn P, Villacian J, Lazzarin A, et al. Ritonavir-boosted tipranavir demonstrates superior efficacy to ritonavir-boosted protease inhibitors in treatment-experienced HIV-infected patients: 24-week results of the RESIST-2 trial. Clin Infect Dis 2006; 43:1347.
  154. Cooper D, Zajdenverg R, Ruxrungtham K, et al. Efficacy and safety of two doses of tipranavir/ritonavir versus lopinavir/ritonavir-based therapy in antiretroviral-naive patients: results of BI 1182.33 (Abstract PL13.4]. In: Program and abstracts of the 8th International Congress on Drug Therapy in HIV infection 2006.
  155. Emu B, Fessel J, Schrader S, et al. Phase 3 Study of Ibalizumab for Multidrug-Resistant HIV-1. N Engl J Med 2018; 379:645.
  156. Huesgen E, DeSear KE, Egelund EF, et al. A HAART-Breaking Review of Alternative Antiretroviral Administration: Practical Considerations with Crushing and Enteral Tube Scenarios. Pharmacotherapy 2016; 36:1145.
  157. Fulco PP. Crushed bictegravir/emtricitabine/tenofovir alafenamide in a human immunodeficiency virus-positive patient with esophageal cancer. Am J Health Syst Pharm 2020; 77:509.
  158. US Food and Drug Administration. Biktarvy: prescribing information. https://www.accessdata.fda.gov/drugsatfda_docs/label/2021/210251s007lbl.pdf (Accessed on March 03, 2021).
Topic 13978 Version 42.0

References