Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2022 Mar 1.
Published in final edited form as: J Neuroimmune Pharmacol. 2019 Dec 10;16(1):130–143. doi: 10.1007/s11481-019-09886-7

CNS Neurotoxicity of Antiretrovirals

Tyler Lanman 1, Scott Letendre 2, Qing Ma 3, Anne Bang 4, Ronald Ellis 1
PMCID: PMC7282963  NIHMSID: NIHMS1546237  PMID: 31823251

Abstract

The development of novel antiretroviral treatments has led to a significant turning point in the fight against HIV. Although therapy leads to virologic suppression and prolonged life expectancies, HIV-associated neurocognitive disorder (HAND) remains prevalent. While various hypotheses have been proposed to explain this phenomenon, a growing body of literature explores the neurotoxic effects of antiretroviral therapy. Research to date brings into question the potential role of such medications in neurocognitive and neuropsychiatric impairment seen in HIV-positive patients. This review highlights recent findings and controversies in cellular, molecular, and clinical neurotoxicity of antiretrovirals. It explores the pathogenesis of such toxicity and relates it to clinical manifestations in each medication class. The concept of accelerated aging in persons living with HIV (PLWH) as well as potential treatments for HAND are also discussed. Ultimately, this article hopes to educate clinicians and basic scientists about the neurotoxic effects of antiretrovirals and spur future scientific investigation into this important topic.

Graphical Abstract

graphic file with name nihms-1546237-f0001.jpg

Introduction

The HIV epidemic led to the development of a myriad of antiretroviral therapies. First discovered was azidothymidine (AZT), and after patterns of AZT-resistance emerged, other nucleoside reverse transcriptase inhibitors (NRTIs) were developed. Next came non-nucleoside reverse transcriptase inhibitors (NNRTIs) and protease inhibitors (PIs). Later came integrase inhibitors, fusion inhibitors, and entry inhibitors. A pharmacokinetic enhancer class (cobicistat) was recently introduced designed to improve the pharmacokinetics and increase effectiveness of HIV medications. Today, a regimen combining two NRTIs and one integrase inhibitor is typically recommended, though a multitude of other options exist based on individual circumstances such as genotypic resistance, prior exposure and demonstrated medication intolerance (Saag, Benson et al. 2018). With the advent of combination antiretroviral therapy (cART; sometimes referred to as highly active antiretroviral therapy or HAART), a once fatal disease has become indefinitely controllable, leading to drastically increased life expectancies in affected patients (Marcus, Chao et al. 2016). Since a definitive cure is not yet available, patients require life-long therapy, and with such a prolonged exposure to medications (in addition to long-term toxicity from the first-generation medications), a careful consideration of neurological adverse effects is warranted.

In particular, antiretroviral use has been associated with a range of neurological toxicity, from peripheral neuropathy to neuropsychiatric and neurocognitive deficits in the central nervous system (CNS) (Meeker, Robertson et al. 2014). However, it is often difficult to distinguish certain adverse effects caused by HIV medications from direct and indirect deleterious effects from the virus itself (Treisman and Soudry 2016). One such instance is HIV-associated neurocognitive disorder (HAND), a term which describes several disorders based on severity of neurocognitive impairment. They are asymptomatic neurocognitive impairment (ANI), mild neurocognitive disorder (MND), and HIV-associated dementia (HAD), a progressive and life-threatening form of dementia (Antinori, Arendt et al. 2007, Letendre 2011). To date, no specific treatment exists for HAND nor is a diagnostic biomarker available (Saylor, Dickens et al. 2016). Although other non-neurological conditions have declined in prevalence due to the efficacy of cART, HAND remains common in the cART era. It is estimated that about one third of HIV+ patients have a HAND diagnosis and over half have neuropsychological impairment (Heaton, Clifford et al. 2010, Sacktor, Skolasky et al. 2016). Interestingly, the compositional prevalence of its subgroups has changed in the cART era. HAD has become increasingly uncommon (2%) while rates of ANI and MND actually increased (Heaton, Franklin et al. 2011, Singer and Nemanim 2017). This suggests that either cART is unable to adequately suppress HIV in the nervous system or that cART use is contributing to the development of HAND (Etherton, Lyons et al. 2015).

In this article, we evaluate each of the classes of HIV therapy, reviewing the latest concepts and controversies regarding the clinical manifestations and cellular mechanisms of ART-induced CNS neurotoxicity. Where applicable, we include antiretroviral routes of administration in in vivo studies (intraperitoneal, CSF, etc.), and mention when medications used in studies are clinically relevant. However, note that estimating clinically-relevant concentrations is difficult, given lack of data on antiretroviral CSF:plasma area under the curve, predictions that parenchymal concentrations can reach greater levels than in the CSF, and the fact that HIV disrupts the blood brain barrier (BBB), allowing for increased antiretroviral CSF accessibility (Decloedt, Rosenkranz et al. 2015, Jensen, Monnerie et al. 2015). We discuss how CNS penetrance by ART may affect neurotoxicity, explore the concept of accelerated aging in PLWH (persons living with HIV), and highlight recent advancements in the possible treatment of HAND. Peripheral nervous system toxicity is beyond the scope of this review and only briefly covered.

Nucleoside reverse transcriptase inhibitors (NRTIs)

NRTIs, the first class of HIV medications discovered, work by blocking reverse transcriptase, thereby preventing the virus from generating functional cDNA via premature DNA strand termination (Shah, Gangwani et al. 2016). In ascending order of approval date, the NRTIs are azidothymidine/zidovudine (AZT), didanosine (ddl), stavudine (d4T), lamivudine (3TC), abacavir (ABC), tenofovir disoproxil fumarate (TDF), emtricitabine (FTC), and tenofovir alafenamide fumarate (TAF). Older NRTIs such as AZT were found to have more off-target effects, limiting their clinical use relative to newer agents (Schweinsburg, Taylor et al. 2005).

Although potent inhibitors of reverse transcriptase, NRTIs also cause off-target inhibition of mitochondrial polymerase γ, the enzyme responsible for normal mitochondrial DNA replication (Kakuda 2000). Through this inhibition, the primary mechanism of NRTI toxicity appears to be mitochondrial toxicity, energy depletion, and oxidative stress, which have been demonstrated both in vitro and in vivo (Lewis, Day et al. 2003, Kohler and Lewis 2007, Nooka and Ghorpade 2018). The extent of mitochondrial polymerase γ inhibition among NRTIs is ddl > d4T >> 3TC > TDF ≥ FTC ≥ AZT ≥ ABC (Bienstock and Copeland 2004). This type of mitochondrial toxicity is considerably cell/tissue-dependent. Stavudine impairs mitochondria in axons and Schwann cells causing peripheral neuropathy, AZT impairs mitochondria in skeletal muscles and causes myopathy, and others can cause lipoatrophy and lactic acidosis (White 2001, Abers, Shandera et al. 2014, Margolis, Heverling et al. 2014). Mitochondrial DNA (mtDNA) depletion from NRTI exposure is also persistent, dependent on cumulative exposure, and can cause long-term effects even after discontinuation (Poirier, Divi et al. 2003, Underwood, Robertson et al. 2015). TAF, a prodrug of tenofovir and a component of the vast majority of modern regimens, produces greater intracellular concentrations than TDF, which might lead to worse neurotoxicity.

It was previously thought that NRTI neurotoxicity was limited to the periphery, but emerging evidence has called this into question. From a clinical standpoint, AZT is known to cause insomnia, nausea, and severe headaches, and in high doses can cause seizures (Richman, Fischl et al. 1987, Saracchini, Vaccher et al. 1989). Other NRTIs have been linked to retinal atrophy, and dose-dependent psychiatric disturbances (Turjanski and Lloyd 2005, Gabrielian, MacCumber et al. 2013). One study used magnetic resonance spectroscopy in patients as a proxy for brain mitochondrial integrity and their results suggested that didanosine and/or stavudine may cause depleted brain mitochondria (Schweinsburg, Taylor et al. 2005). On a cellular level too, NRTIs have been implicated in CNS toxicity. Abacavir induced endoplasmic reticulum (ER) stress in human astrocytes at therapeutic doses, activating all three unfolded protein response (UPR) pathways in vitro (Nooka and Ghorpade 2017, Nooka and Ghorpade 2018). Oligodendrocyte dysfunction (both in vitro and in vivo with intravenous administration) seen with other ART drugs (such as ritonavir and lopinavir) was not observed in NRTIs (Jensen, Monnerie et al. 2015). In mice, long-term intraperitoneal NRTI administration at clinically relevant concentrations led to mtDNA deletion and mitochondrial toxicity in cortical neurons (Zhang, Song et al. 2014, Hung, Chen et al. 2017). Additionally, TDF has been associated with increased risk of developing chronic kidney disease (Scherzer and Shlipak 2015) (presumably through mitochondrial nephrotoxicity (Rodriguez-Nóvoa, Alvarez et al. 2010)) which, in itself, is known to cause cognitive decline (Etgen, Chonchol et al. 2012). Overall, given the link between mitochondrial dysfunction and cognitive impairment (Finsterer 2012), researchers have suggested that although no direct clinical association has been found, NRTI-related mitochondrial toxicity may directly or indirectly contribute to the development of HAND (Hung, Chen et al. 2017).

Non-nucleoside reverse transcriptase inhibitors (NNRTI)

NNRTIs include, in order of approval, nevirapine, delavirdine, efavirenz, etravirine, rilpivirine and doravirine. Unlike NRTIs, these drugs do not resemble nucleotides/nucleosides and act on reverse transcriptase noncompetitively to impair cDNA synthesis. Although this class is generally better tolerated than NRTIs, resistant HIV strains became problematic, necessitating that NNRTIs be used in combination with other antiretrovirals (hence cART) (Margolis, Heverling et al. 2014). As a class, the most common adverse event is rash, though individual drugs in this class have their own specific side effect profiles (Drake 2000).

Of the NNRTIs, the most infamous for CNS toxicity is efavirenz, which in the past was also one of the most commonly prescribed cART components due to its efficacy and favorable pharmacokinetics (Shah, Gangwani et al. 2016). Efavirenz has been associated with both neurological (dizziness, insomnia, vivid dreams, headache, and impaired concentration) and psychiatric (paranoia, hallucinations, anxiety, mania, and depression) adverse effects (Apostolova, Funes et al. 2015). These adverse effects occur in upwards of half of patients taking efavirenz and although they typically resolve after several weeks, some can be more persistent (Arendt, de Nocker et al. 2007). The adverse effect most classically associated with efavirenz is vivid dreams. An ambulatory electroencephalogram (EEG) study found that patients taking efavirenz (in a dose-dependent manner) had longer sleep latencies and shorter duration of rapid eye movement (REM) sleep, which was theorized to result in more intense REM periods (i.e. vivid dreams) (Gallego, Barreiro et al. 2004). This lack of sleep efficacy (which typically persists for over 3 months of therapy) also would explain the daytime fatigue and somnolence experienced by patients on the medication (Moyle, Fletcher et al. 2006). Psychiatric symptoms caused by efavirenz exposure can be even more disabling for certain patients. The population of PLWH already have higher rates of psychiatric disorders than the general population (with nearly half of PLWH screening positive) (Bing, Burnam et al. 2001). Clinicians therefore need to carefully screen and monitor their patients when prescribing efavirenz, especially since it may cause increased rates of suicidality (Mollan, Smurzynski et al. 2014), although this remains controversial (Kenedi and Goforth 2011). However, when mental illness contraindicates this drug, using alternative regimens which have less convenient dosing schedules could lead to decreased ART adherence (Kenedi and Goforth 2011).

The mechanisms responsible for efavirenz neurotoxicity (or more relevantly, its main metabolite, 8-hydroxy-efavirenz, a more potent neurotoxin than the parent drug) are currently not well elucidated (Apostolova, Funes et al. 2015, Grilo, Joao Correia et al. 2017). Recently, there has been considerable scientific interest in understanding how pharmacogenetics impacts its CNS side effects. Research suggests that, similar to NRTIs, the toxicity of efavirenz is mediated by oxidative stress and consequent mitochondrial dysfunction (in addition to elevating intracellular pro-inflammatory factors) (Shah, Gangwani et al. 2016, Ciavatta, Bichler et al. 2017). Furthermore, efavirenz is consistently found to be more neurotoxic than other ART drugs tested, consistent with its clinical side effect profile. In one experiment of four antiretrovirals in primary rat neurons, efavirenz was the only one to cause ER stress and mitochondrial toxicity at clinically-relevant concentrations (Blas-García, Polo et al. 2014). In an in vitro study, efavirenz elicited a dose-dependent (encompassing the range of clinical concentrations) impairment in striatal nerve terminal mitochondrial respiration, leading to depleted ATP levels at the synapse (Stauch, Emanuel et al. 2017). In a recent in vitro and ex vivo study, efavirenz was the only NNRTI (and more potently than ART drugs in other classes) that demonstrated detrimental effects on neuronal viability, morphology, respiration, and excitability when exposed to rat cortical neurons at target plasma concentrations (Ciavatta, Bichler et al. 2017).

Given the well-characterized CNS side effect profile of efavirenz and the persistence of HAND in the cART era, researchers were interested in its effect on cognitive function. As expected, efavirenz is associated with long-term cognitive impairment. In a recent large cohort study, patients taking long-term efavirenz had significant neurocognitive impairment in many domains compared to those taking lopinavir-ritonavir. This effect was less among HCV seropositive individuals (Ma, Vaida et al. 2016). Another large study observed efavirenz use was associated with HAND, with higher education acting as a protective factor (Ciccarelli, Fabbiani et al. 2011). Switching patients from efavirenz to an alternative regimen did not lead to improvement in neurocognitive measures after 10 weeks, suggesting that efavirenz likely leads to persistent neurocognitive dysfunction (Payne, Chadwick et al. 2017).

Other drugs in the NNRTI class in addition to efavirenz are known to have CNS toxicity, with nevirapine being more toxic than the remaining NNRTIs (Shah, Gangwani et al. 2016). However, compared to efavirenz, these drugs’ CNS side effects are less studied, less frequent, and less significant in clinical practice (Abers, Shandera et al. 2014).

Protease Inhibitors (PI)

In the HIV life cycle, once mRNA is translated into protein precursors, a virally-encoded protease is required to cleave these into mature proteins (Flexner 1998, Brik and Wong 2003). The protease enzyme as a therapeutic target led to the development of protease inhibitors, including saquinavir mesylate, ritonavir, indinavir, nelfinavir mesylate, lopinavir, atazanavir sulfate, fosamprenavir calcium, tipranavir, and darunavir. Of note, after discovering the cytochrome P450-inhibiting effects of ritonavir, it is now used mostly as a pharmacokinetic booster, allowing for less-frequent dosing of PI-containing regimens (Lv, Chu et al. 2015). In comparison to NNRTI-containing regimens, PI-based regimens were found to have lower rates of resistance (Riddler, Haubrich et al. 2008), though the use of PIs has been limited by their drug-drug interactions and off-target toxicities. In particular, they can cause lipodystrophy syndrome (due to homology between protease enzyme and two lipid metabolism enzymes) and insulin resistance (which in some cases, can lead to the development of diabetes), in addition to cardiovascular disease (Carr 2000, Brown, Cole et al. 2005, Lv, Chu et al. 2015). Newer PIs, such as darunavir, have been designed specifically to minimize these off-target effects (Pokorná, Machala et al. 2009).

Results from cell and animal studies of PI neurotoxicity have been mixed. In one in vitro study, darunavir did not cause mitochondrial toxicity in rat neurons at clinically relevant concentrations, unlike efavirenz (Blas-García, Polo et al. 2014). Lopinavir and to a lesser extent, amprenavir, caused disruption of astrocytic glutamate homeostasis in vitro and were associated with gliosis and neurobehavioral deficits in mice exposed to oral doses (Vivithanaporn, Asahchop et al. 2016). Lopinavir, but not darunavir, was neurotoxic to primary rat neuroglial cultures. This was thought to be mediated by oxidative stress (Stern, Lee et al. 2018). In another in vitro study, darunavir caused reactive oxygen species (ROS) production in astrocytes although not at clinically relevant concentrations (Latronico, Pati et al. 2018). Intravenous ritonavir and lopinavir (at doses based on human plasma and CSF levels) had detrimental effects on mice oligodendrocyte maturation in vivo which was reversed with drug cessation (Jensen, Monnerie et al. 2015). Investigators studying the effects of ART on neurotransmitter release found that indinavir reduced in vitro synaptic acetylcholine transmission at plasma-level concentrations (Ekins, Mathews et al. 2017).

PIs also appear to cause certain CNS effects on a clinical level. Ritonavir was shown to be more neurotoxic than other PIs and can cause nausea, dizziness, and circumoral paresthesia (Markowitz, Saag et al. 1995). However, using ritonavir as an pharmacokinetic enhancer allows for lower doses, which reduces the frequency of adverse events (Hill, van der Lugt et al. 2009). Several studies (Bacellar, Muñoz et al. 1994, Pettersen, Jones et al. 2006) have found increased risk of peripheral neuropathy with PI use (although a recent analysis found the independent risk from PIs is small (Ellis, Marquie-Beck et al. 2008)). Based on results from aforementioned cell and animal studies, it is feasible that PI use could contribute to neurocognitive dysfunction. HAND has been associated with myelin disruption (with reduced levels of myelin basic protein) and structural white matter deterioration on imaging (ritonavir and lopinavir have oligodendrocyte toxicity (Jensen, Monnerie et al. 2015)). Furthermore, since neurotransmitter system dysfunction could help explain ART CNS toxicity, the authors who found impaired synaptic acetylcholine transmission with indinavir suggested that this may contribute to cognitive dysfunction (Ekins, Mathews et al. 2017). An autopsy study found that PI exposure increased the risk of cerebral small vessel disease, which was, in turn, associated with neurocognitive impairment (Soontornniyomkij, Umlauf et al. 2014). A large study did not find differences in neurocognitive performance with PI use, in comparison to triple therapy, after several years (Arenas-Pinto, Stöhr et al. 2016). Another study found that CSF viral escape (when HIV is detectable in CSF but not in the serum) is associated with PI use, but did not lead to worse neurocognitive performance (Pérez-Valero, Ellis et al. 2019). PI use is associated with hyperbilirubinemia, but this was not shown to affect neurocognitive function (Barber, Moyle et al. 2016). Despite the link between PIs and certain neurologic adverse effects, there is little, if any, clinical or preclinical evidence of a link between their use and HAND.

Integrase Inhibitors

Integrase is an HIV-encoded protein necessary for integration of viral cDNA into host DNA and after 12 years of development, the first agent in the integrase inhibitor class, raltegravir, was introduced in 2007 (Pommier, Johnson et al. 2005), followed by dolutegravir, elvitegravir, and most recently approved, bictegravir in 2018. In general, these drugs are some of the most efficacious among antiretrovirals, have low rates of resistance, and are relatively tolerable in the clinical setting (Patel P. 2018). The most common side effects of this class include diarrhea, nausea, and headache (del Mar Gutierrez, Mateo et al. 2014). In clinical trials, raltegravir had lower rates of CNS adverse events than efavirenz and similar rates of severe adverse effects relative to placebo (Lennox, DeJesus et al. 2010, Steigbigel, Cooper et al. 2010, Nguyen, Isaacs et al. 2011). Subsequent studies found higher rates of myalgia in patients taking raltegravir although this was rarely a cause for discontinuation (Lee, Amin et al. 2013). A large study in Botswana found evidence for neural tube defects associated with dolutegravir use during pregnancy (Zash, Jacobson et al. 2017, Zash, Makhema et al. 2018). The most common neuropsychiatric effect reported with raltegravir and dolutegravir is insomnia which was reversible after drug cessation and can be improved by switching to morning dosing schedules (Gray and Young 2009, Capetti, Di Giambenedetto et al. 2017). Other neuropsychiatric effects linked to integrase inhibitors include depression and anxiety and have been found to have higher rates than initially suggested by clinical trials (Harris, Larsen et al. 2008, Curtis, Nichols et al. 2014, Fettiplace, Stainsby et al. 2017, Harris 2018). A large clinical study found that the discontinuation rates due to adverse events for raltegravir, dolutegravir, and elvitegravir were 3.6, 3.8, and 5.0% (Penafiel, de Lazzari et al. 2017). Dolutegravir had higher rates of discontinuation due to neuropsychiatric effects compared to raltegravir and elvitegravir. These results were consistent with findings from a previous cohort study which additionally showed an almost three-fold increase in discontinuation rate in female patients and older patients (Hoffmann, Welz et al. 2017). When bictegravir was introduced, trials found rates of neuropsychiatric effects comparable to dolutegravir, suggesting a class effect of integrase inhibitors (Gallant, Lazzarin et al. 2017, Sax, Pozniak et al. 2017).

Although reports of neuropsychiatric effects from integrase inhibitors suggest neurotoxicity, underlying mechanisms for such toxicity are not fully understood. In one in vitro study, raltegravir did not cause mitochondrial toxicity in rat neurons at clinically-relevant concentrations, unlike efavirenz (Blas-García, Polo et al. 2014). In another, raltegravir caused ROS production in astrocytes, although not at clinically relevant concentrations (Latronico, Pati et al. 2018). However, an in vitro study found that elvitegravir but not raltegravir nor dolutegravir was neurotoxic to primary rat neuroglial cultures at clinically relevant plasma level concentrations. This effect was thought to be mediated by the integrated stress response (ISR) rather than strictly oxidative stress (Stern, Lee et al. 2018). The ISR is normally an adaptive response to cellular stressors which restores homeostasis but with prolonged exposure to certain insults, this response activates pathways that lead to cell death (Pakos-Zebrucka, Koryga et al. 2016).

A clinical study of dolutegravir-containing ART found high dolutegravir concentrations in the CSF, suggesting a possible mechanism by which concentration-dependent neurotoxicity causes CNS adverse effects (Letendre, Mills et al. 2014). Other than neuropsychiatric effects, integrase inhibitors do not appear to cause significant neurocognitive impairment. On the contrary, dolutegravir is being studied as a possible treatment for HAND, as discussed later.

Entry Inhibitors

To infect a host cell, the HIV envelope proteins gp41 and gp120 bind to host CD4 and then to a co-receptor, typically CCR5 or CXCR4. In 2003, enfuvirtide, a gp41 inhibitor was approved and later maraviroc, a CCR5 antagonist, gained FDA approval. Very recently, ibalizumab, a monoclonal antibody against CD4, gained approval in 2018. These drugs prevent viral entry into host cells. Of note, HIV-2 uses different chemokine receptors and therefore this class is only effective with HIV-1 (Saraiya, Kanagala et al. 2018).

Enfuvirtide use in ART is limited by its requirement of twice-daily parenteral administration due to poor solubility and rapid removal from circulation (although research shows that conjugating it with polyethylene glycol may help with this problem) (Cheng, Wang et al. 2016). However, it remains an effective therapy for drug-resistant HIV when other regimens have been exhausted (Lalezari, Henry et al. 2003. Enfuvirtide was initially thought to have increased rates of peripheral neuropathy (Fung and Guo 2004, yet subsequent studies found no clear evidence of this link (Cherry, Duncan et al. 2008). To date, there have been no significant reports of CNS toxicity in enfuvirtide, and in general, it has a favorable safety profile with adverse events mostly limited to injection-site reactions (LaBonte, Lebbos et al. 2003, Oldfield, Keating et al. 2005, Manfredi and Sabbatani 2006, Treisman and Soudry 2016).

Maraviroc is a slowly reversible, noncompetitive CCR5 antagonist. Similar to enfuvirtide, maraviroc has favorable tolerability, a limited resistance pattern, and is a potent agent in virologic failure cases (Emmelkamp and Rockstroh 2007, Emmelkamp and Rockstroh 2008). In clinical trials, maraviroc monotherapy achieved rapid viral load reduction in a matter of days (Fatkenheuer, Pozniak et al. 2005), and the most common side effects were similar between maraviroc and placebo (Yost, Pasquale et al. 2009). However, maraviroc is only effective in patients with CCR5-tropic HIV-1, a feature that limits its use and requires tropism testing prior to use (Emmelkamp and Rockstroh 2008). Unfortunately, all trials on CXCR4 inhibitors have failed due to peripheral toxicity (Shah, Gangwani et al. 2016). In in vitro toxicology studies, maraviroc was the least toxic to astrocytes compared to a number of ART drugs from other classes, with a TC50 10,000-fold higher than CSF concentrations (Latronico, Pati et al. 2018). One in vitro study showed that it may cause pro-inflammatory activation of microglia cells in rats (Lisi, Tramutola et al. 2012). However, a subsequent study provided evidence against this claim, showing that by blocking CCR5 in the CNS, maraviroc could ameliorate neuropathic pain (when administered intrathecally in rats) by restoring the balance of pro- and antinociceptive factors in astrocytes and microglia (Piotrowska, Kwiatkowski et al. 2016). There have been no substantial clinical reports of neurocognitive impairment with maraviroc. Rather, maraviroc and a similar investigational drug, cenicriviroc, are being studied as potential treatment options for HAND, as discussed below.

Ibalizumab, the most recent entry inhibitor, has advantages over others in the class. Its weekly dosing could improve adherence and its unique mechanism of action could prevent cross-resistance of HIV. Although data on neurotoxicity screening in this medication is sparse, it has also been fairly well-tolerated with no significant neurological effects reported (Jacobson, Kuritzkes et al. 2009, Bruno and Jacobson 2010).

Pharmacokinetic Enhancers

When ritonavir was initially approved at a 600mg twice daily dose, toxicity (nausea, vomiting, diarrhea, etc.) led to discontinuation in up to a third of patients (Rublein, Eron Jr et al. 1999, Monforte, Lepri et al. 2000). Additionally, it led to many drug-drug interactions due to its cytochrome P450 inhibiting effects (predominantly CYP3A4 but also CYP2D6) (Kumar, Rodrigues et al. 1996, Rathbun and Rossi 2002). In humans, ritonavir increased the area under the curve (AUC) of CYP3A-metabolized drugs by up to 20-fold in humans and increased AUC of CYP2D6-metabolized drugs by 145% (Hsu, Granneman et al. 1998). Given that most PIs undergo metabolism through the CYP3A pathway, researchers quickly realized the potential of using ritonavir to “boost” levels of these drugs. Trials comparing ritonavir to dual protease inhibition with ritonavir and another drug led to substantial improvements in viral suppression and allowed ritonavir to be used at less toxic doses (Yu and Daar 2000, Michelet, Ruffault et al. 2001). With this discovery, the pharmacokinetic enhancer class was incidentally created. Adding an enhancer to an ART regimen allows for reduced pill burden, simpler regimens, and improved adherence, which all lead to increased antiviral efficacy (Xu and Desai 2009). Ritonavir itself does not appear to have serious CNS effects although by boosting levels of other drugs, it theoretically has the potential to indirectly propagate such neurotoxic effects of antiretrovirals (Danner, Carr et al. 1995, Carr and Cooper 2000).

Cobicistat is a CYP3A inhibitor designed to enhance the activity of antiretrovirals similar to ritonavir, but holds several unique advantages such as an easier dosing schedule and a more favorable side effect profile (Xu, Liu et al. 2010, Larson, Wang et al. 2014, Marzolini, Gibbons et al. 2016, Tseng, Hughes et al. 2017). Similar to ritonavir, it is possible that it could promote potential neurotoxic effects of the medications it enhances. Although no evidence of neurotoxicity has been reported, it has not been extensively tested relative to other HIV medications.

Blood Brain Barrier (BBB)

HIV invasion of the CNS occurs early in disease progression, with the virus being detected in CSF as early as 8 days after initial exposure, leading to activation of pro-inflammatory responses in the CSF and brain parenchyma (Valcour, Chalermchai et al. 2012). In around 5-20% of HIV+ patients on ART, HIV is detected in the CSF despite elimination in the plasma below detectable limits, a term called CSF viral escape (Canestri, Lescure et al. 2010, Joseph, Cinque et al. 2016). This entity can be divided into three categories- asymptomatic, neuro-symptomatic (clinical and progressive CNS disease), and secondary (increased CSF virus resulting from a secondary infection) (Ferretti, Gisslen et al. 2015). The CSF reservoir created by this escape is associated with elevated CSF levels of neopterin (a marker of macrophage activation), and is thought to increase the risk of HAND (Chen, Gill et al. 2014, Gisslén and Hunt 2019). It was theorized that if antiretroviral drugs could penetrate the BBB, this HIV reservoir could be effectively reduced, leading to improvement in CNS insult. To estimate exposure to the CNS by antiretrovirals, researchers developed the CNS penetration effectiveness (CPE) scale. Each drug is ranked from one (lowest penetrance) to four (highest penetrance) based on factors such as CSF concentration and drug pharmacology (Letendre, Ellis et al. 2010). The CPE scale’s negative correlation with viral RNA in the CNS (the higher the score, the lower the viral load) was validated in several studies (Letendre, Marquie-Beck et al. 2008, Marra, Zhao et al. 2009). CPE correlation with neurocognitive performance is less clear.

Several studies found that regimens with higher CPE were associated with better neurocognitive function in addition to lower CNS levels of TNF-α, a prominent inflammatory marker (Cysique, Waters et al. 2011, Smurzynski, Wu et al. 2011, Tiraboschi, Muñoz-Moreno et al. 2015, Carvalhal, Gill et al. 2016). In contrast, other studies found either no effect or the opposite effect with higher CPE scores correlating with lower neurocognitive performance or higher risk of dementia (Marra, Zhao et al. 2009, Cross, Combrinck et al. 2013, Caniglia, Cain et al. 2014). Some found that ART intensification with high-CPE medications did not a translate to reduced intrathecal immunoactivation (Yilmaz, Verhofstede et al. 2010, Dahl, Lee et al. 2011). Furthermore, one study found that interrupting ART is associated with improved neurocognitive performance (Robertson, Su et al. 2010). Participants in this study took older, more toxic regimens, so the relevance of this finding for newer ART is unclear. Another study found that placing patients on higher CPE regimens only improved neurocognition in patients who were impaired at baseline (Tozzi, Balestra et al. 2009). Authors of these studies suggest that although highly-penetrating regimens are effective at reducing the CNS viral reservoir, they also have higher potential to exert neurotoxicity. Future investigation is required to determine which regimens can optimally suppress HIV in the CNS while simultaneously minimizing neurotoxicity, in the hopes of stabilizing or improving neurocognition.

Aging and Antiretrovirals

With the advent of ART, HIV+ patients have been living longer, and while this is a step in the right direction, the graying of this population brings with it certain clinical ramifications (Kirk and Goetz 2009). For instance, age-related multimorbidity in PLWH (including metabolic syndrome and vascular disease) may also contribute to neurotoxicity, with the resulting polypharmacy increasing the risk of drug-drug interactions that could cause CNS injury (Alonso-Villaverde, Aragonès et al. 2010, Tarr and Telenti 2010). Although the underlying mechanisms remain largely unclear, HIV and aging appear to independently contribute to neurocognitive decline and HAND development (Cañizares, Cherner et al. 2014, Seider, Luo et al. 2014, Coban, Robertson et al. 2017). This suggests that HIV patients experience premature and accelerated aging, although some researchers question whether the root cause is HIV itself or rather the deleterious effects from therapy (Smith, de Boer et al. 2012).

A working hypothesis to explain the accelerated aging phenomenon is that age-related CNS injury resulting from toxicity of ART and concomitant drugs enhance vulnerability to CNS complications, even in those with virologic control. Aging-related changes in drug distribution, binding proteins, metabolism and elimination can lead to greater ART drug exposure in the elderly (Mangoni and Jackson 2004, Klotz 2009, Winston, Jose et al. 2013). Aging causes structural and functional changes in the BBB, such as decreased endothelial cell counts, choroid plexus epithelium flattening and calcification, as well as thickening of basement and arachnoid membranes. These changes result in increased BBB permeability which may likely affect ART CNS pharmacokinetics (Erdő, Denes et al. 2017). PI distribution in the CNS seems to be particularly affected by age, with studies showing that elderly HIV+ patients have decreased clearance of lopinavir and darunavir, longer half-life of indinavir, and higher total exposure of atazanavir (Zhou, Havlir et al. 2000, Crawford, Spritzler et al. 2010, Avihingsanon, Kerr et al. 2013, Winston, Jose et al. 2013, Calza, Colangeli et al. 2017).

Current research is investigating ways to mitigate accelerated cognitive aging in PLWH. One trial ( NCT02936401) is currently assessing the use of Mindfulness Based Stress Reduction as a method to improve function in patients older than 60 with HAND. Another ( NCT03483740) is testing cognitive remediation group therapy in a similar cohort of older individuals with HAND. A comprehensive review of potential HAND treatment is discussed below.

Experimental HAND Treatment

Given the persistence of HAND in the cART era and the possible contribution from antiretroviral neurotoxicity, a number of previous and current trials have investigated possible therapeutic options to combat HAND (Cross and Kolson 2017). These include drugs already approved for treating other neurodegenerative diseases (selegiline and memantine) (Schifitto, Navia et al. 2007, Schifitto, Zhang et al. 2007), drugs predominantly used for nonneurologic conditions (minocycline, fluconazole, intranasal insulin [ NCT03277222], and statins [ NCT01600170]) (Rezaie-Majd, Maca et al. 2002, Sacktor, Miyahara et al. 2011, Gerena, Skolasky et al. 2012, Nakasujja, Miyahara et al. 2013, Meulendyke, Queen et al. 2014, Sacktor, Skolasky et al. 2018), and antioxidants (Coenzyme Q10, heme oxygenase-1, and dimethyl fumarate) (Cross, Cook et al. 2011, Louboutin and Strayer 2018, Velichkovska, Surnar et al. 2018).

Although some ART drugs are associated with neurotoxicity, several ongoing trials are testing treatment intensification approach for cognitive improvement. One trial ( NCT01448486) investigated the effects of raltegravir intensification on neurocognitive performance but was unfortunately stopped prematurely due to insufficient patient recruitment. Maraviroc intensification in humans caused an improvement in neuropsychiatric performance, hypothesized to result from reducing the HIV burden in monocytes, leading to two current clinical trials ( NCT02159027 and NCT02519777) (Burdo, Weiffenbach et al. 2013, Ndhlovu, Umaki et al. 2014). Cenicriviroc, when given to HAND patients, led to decreased inflammatory monocyte activation and subtle improvement in cognitive performance (D'Antoni, Paul et al. 2018).

Apart from a few mild successes in trials listed above, we still have not discovered a consistent and efficacious treatment or prevention of HAND. The explanation for this lack of effectiveness is multifactorial. Inherently, clinical trials frequently fail despite promising preclinical results, due to inadequate patient recruitment/retention, fundamental differences between animal models and human subjects, unforeseen adverse effects, etc. More specifically, the underlying epidemiology, natural progression, and pathogenesis behind HAND still eludes us. Does persistent HAND despite virologic suppression result from incomplete antiretroviral CSF penetration, direct or indirect neurotoxicity from antiretrovirals, or something else entirely? Without a clear pathological target, developing specific treatment modalities becomes exceptionally challenging. This is why the impetus for the aforementioned clinical trials came either from medications that showed neuroprotection in other diseases or simply came from incidental findings in the clinic. As such, it is unlikely that these therapies could actually reverse ART-induced specific neurotoxicities rather than simply imparting general neuroprotection. In order to properly confront this disease entity, more research to provide answers to preclinical questions about HAND is essential.

Conclusions

Antiretroviral neurotoxicity is a growing body of research, with novel molecular, cellular, and animal studies uncovering the pathogenesis of such toxicity and relating it to clinical manifestations seen in patients. Each medication has a unique side effect profile, but understanding their long-term effects is becoming increasingly relevant, as the development of new therapy extends the average lifespan of PLWH. New challenges are being uncovered with this aging population, given that they experience longer cumulative ART exposure, have more comorbidities, and develop changes in their pharmacokinetic responses to such drugs (Erdő, Denes et al. 2017). Although HIV exerts neurotoxic effects on the brain and can use the CNS as a reservoir for replication, the fact that regimens with higher CPE do not necessarily lead to cognitive improvement has led researchers to hypothesize that ART itself may, in part, contribute to neurotoxicity (Caniglia, Cain et al. 2014). This theory is supported by the persistence of HAND in the cART era (Heaton, Clifford et al. 2010).

Despite the potential for ART-induced neurotoxicity, viral load reduction in the plasma and CNS should remain the principal objective of antiretroviral treatment. Moving forward, we advocate for the following: 1) clinicians maintain a high level of suspicion of HAND (even when sufficiently treated), 2) scientists continue to unravel the epidemiology and pathogenesis of ART-induced neurotoxicity with rigorous studies, and 3) researchers develop and assess novel treatment options for such neurotoxicity, including HAND.

Table 1.

Antiretroviral medications by class, including year of approval and CNS penetration effectiveness (CPE) score, a measurement of how well medications penetrate the CNS

Abbreviation Approval
Year
CPE
Score
NRTI
azidothymidine/zidovudine AZT/ZDV 1987 4
didanosine ddI 1991 2
stavudine d4T 1994 2
lamivudine 3TC 1995 2
abacavir ABC 1998 3
tenofovir disoproxil fumarate TDF 2001 1
emtricitabine FTC 2003 3
tenofovir alafenamide TAF 2015 1
NNRTI
nevirapine NVP 1996 4
delavirdine DLV 1997 3
efavirenz EFV 1998 3
etravirine ETR 2008 2
rilpivirine RPV 2011 -
doravirine DOR 2018 -
Protease Inhibitors
saquinavir mesylate SQV 1995 1
ritonavir* RTV 1996 1
indinavir IDV 1996 3
nelfinavir mesylate NFV 1997 1
lopinavir LPV 2000 3
atazanavir sulfate ATV 2003 2
fosamprenavir calcium FOS 2003 2
tipranavir TPV 2005 1
darunavir DRV 2006 3
Fusion Inhibitors
enfuvirtide T-20 2003 1
CCR5 Co-receptor Antagonists
maraviroc MVC 2007 3
Integrase Inhibitors
raltegravir RAL 2007 3
dolutegravir DTG 2013 -
elvitegravir EVG 2014 -
bictegravir BIC 2018 -
Post-Attachment Inhibitors
ibalizumab IBA 2018 -
Pharmacokinetic Enhancers
ritonavir* RTV 1996 1
cobicistat COBI 2014 -
*

Ritonavir is used clinically as a PK enhancer rather than an antiretroviral.

Footnotes

Conflict of Interest: The authors declare that they have no conflict of interest.

Publisher's Disclaimer: This Author Accepted Manuscript is a PDF file of a an unedited peer-reviewed manuscript that has been accepted for publication but has not been copyedited or corrected. The official version of record that is published in the journal is kept up to date and so may therefore differ from this version.

References

  1. Abers MS, Shandera WX and Kass JS (2014). "Neurological and psychiatric adverse effects of antiretroviral drugs." CNS drugs 28(2): 131–145. [DOI] [PubMed] [Google Scholar]
  2. Alonso-Villaverde C, Aragonès G, Beltrán-Debón R, Fernández-Sender L, Rull A, Rodríguez-Sanabria F, Marsillach J, Pardo-Reche P, Camps J and Joven J (2010). "Host–pathogen interactions in the development of metabolic disturbances and atherosclerosis in HIV infection: The role of CCL2 genetic variants." Cytokine 51(3): 251–258. [DOI] [PubMed] [Google Scholar]
  3. Antinori A, Arendt G, Becker J, Brew B, Byrd D, Cherner M, Clifford D, Cinque P, Epstein L and Goodkin K (2007). "Updated research nosology for HIV-associated neurocognitive disorders." Neurology 69(18): 1789–1799. [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Apostolova N, Funes HA, Blas-Garcia A, Galindo MJ, Alvarez A and Esplugues JV (2015). "Efavirenz and the CNS: what we already know and questions that need to be answered." Journal of Antimicrobial Chemotherapy 70(10): 2693–2708. [DOI] [PubMed] [Google Scholar]
  5. Arenas-Pinto A, Stöhr W, Jäger HR, Haddow L, Clarke A, Johnson M, Chen F, Winston A, Godi C and Thust S (2016). "Neurocognitive function and neuroimaging markers in virologically suppressed HIV-positive patients randomized to ritonavir-boosted protease inhibitor monotherapy or standard combination ART: a cross-sectional substudy from the PIVOT trial." Clinical Infectious Diseases 63(2): 257–264. [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Arendt G, de Nocker D, von Giesen H-J and Nolting T (2007). "Neuropsychiatric side effects of efavirenz therapy." Expert Opinion on drug safety 6(2): 147–154. [DOI] [PubMed] [Google Scholar]
  7. Avihingsanon A, Kerr SJ, Punyawudho B, van der Lugt J, Gorowara M, Ananworanich J, Lange JM, Cooper DA, Phanuphak P and Burger DM (2013). "Aging Not Gender Is Associated with High Atazanavir Plasma Concentrations in Asian HIV-Infected Patients." AIDS research and human retroviruses 29(12): 1541–1546. [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Bacellar H, Muñoz A, Miller E, Cohen BA, Besley D, Seines O, Becker J and McArthur JC (1994). "Temporal trends in the incidence of HTV-l-related neurologic diseases: Multicenter AIDS Cohort Study, 1985-1992." Neurology 44(10): 1892–1892. [DOI] [PubMed] [Google Scholar]
  9. Barber TJ, Moyle G, Hill A, Jagjit Singh G, Scourfield A, Yapa HM, Waters L, Asboe D, Boffito M and Nelson M (2016). "A cross-sectional study to evaluate the association of hyperbilirubinaemia on markers of cardiovascular disease, neurocognitive function, bone mineral density and renal markers in HIV-1 infected subjects on protease inhibitors." HIV Clin Trials 17(3): 123–130. [DOI] [PubMed] [Google Scholar]
  10. Bienstock RJ and Copeland WC (2004). "Molecular insights into NRTI inhibition and mitochondrial toxicity revealed from a structural model of the human mitochondrial DNA polymerase." Mitochondrion 4(2-3): 203–213. [DOI] [PubMed] [Google Scholar]
  11. Bing EG, Burnam MA, Longshore D, Fleishman JA, Sherbourne CD, London AS, Turner BJ, Eggan F, Beckman R and Vitiello B (2001). "Psychiatric disorders and drug use among human immunodeficiency virus–infected adults in the United States." Archives of general psychiatry 58(8): 721–728. [DOI] [PubMed] [Google Scholar]
  12. Blas-García A, Polo M, Alegre F, Funes HA, Martínez E, Apostolova N and Esplugues JV (2014). "Lack of mitochondrial toxicity of darunavir, raltegravir and rilpivirine in neurons and hepatocytes: a comparison with efavirenz." Journal of Antimicrobial Chemotherapy 69(11): 2995–3000. [DOI] [PubMed] [Google Scholar]
  13. Brik A and Wong C-H (2003). "HIV-1 protease: mechanism and drug discovery." Organic & biomolecular chemistry 1(1): 5–14. [DOI] [PubMed] [Google Scholar]
  14. Brown TT, Cole SR, Li X, Kingsley LA, Palella FJ, Riddler SA, Visscher BR, Margolick JB and Dobs AS (2005). "Antiretroviral therapy and the prevalence and incidence of diabetes mellitus in the multicenter AIDS cohort study." Archives of internal medicine 165(10): 1179–1184. [DOI] [PubMed] [Google Scholar]
  15. Bruno CJ and Jacobson JM (2010). "Ibalizumab: an anti-CD4 monoclonal antibody for the treatment of HIV-1 infection." Journal of antimicrobial chemotherapy 65(9): 1839–1841. [DOI] [PubMed] [Google Scholar]
  16. Burdo TH, Weiffenbach A, Woods SP, Letendre S, Ellis RJ and Williams KC (2013). "Elevated sCD163 in plasma but not cerebrospinal fluid is a marker of neurocognitive impairment in HIV infection." AIDS (London, England) 27(9). [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Calza L, Colangeli V, Magistrelli E, Bussini L, Conti M, Ramazzotti E, Mancini R and Viale P (2017). "Plasma trough concentrations of darunavir/ritonavir and raltegravir in older patients with HIV-1 infection." HIV medicine 18(7): 474–481. [DOI] [PubMed] [Google Scholar]
  18. Canestri A, Lescure F-X, Jaureguiberry S, Moulignier A, Amiel C, Marcelin A, Peytavin G, Tubiana R, Pialoux G and Katlama C (2010). "Discordance between cerebral spinal fluid and plasma HIV replication in patients with neurological symptoms who are receiving suppressive antiretroviral therapy." Clinical Infectious Diseases 50(5): 773–778. [DOI] [PubMed] [Google Scholar]
  19. Caniglia EC, Cain LE, Justice A, Tate J, Logan R, Sabin C, Winston A, van Sighem A, Miro JM and Podzamczer D (2014). "Antiretroviral penetration into the CNS and incidence of AIDS-defining neurologic conditions." Neurology 83(2): 134–141. [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Cañizares S, Cherner M and Ellis RJ (2014). HIV and aging: effects on the central nervous system. Seminars in neurology, Thieme Medical Publishers. [DOI] [PMC free article] [PubMed] [Google Scholar]
  21. Capetti A, Di Giambenedetto S, Latini A, Sterrantino G, De Benedetto I, Cossu M and Gori A (2017). "Morning dosing for dolutegravir-related insomnia and sleep disorders." HIV Med 838: 1–2. [DOI] [PubMed] [Google Scholar]
  22. Carr A (2000). "HIV protease inhibitor-related lipodystrophy syndrome." Clinical Infectious Diseases 30(Supplement_2): S135–S142. [DOI] [PubMed] [Google Scholar]
  23. Carr A and Cooper DA (2000). "Adverse effects of antiretroviral therapy." The Lancet 356(9239): 1423–1430. [DOI] [PubMed] [Google Scholar]
  24. Carvalhal A, Gill MJ, Letendre SL, Rachlis A, Bekele T, Raboud J, Burchell A and Rourke SB (2016). "Central nervous system penetration effectiveness of antiretroviral drugs and neuropsychological impairment in the Ontario HIV Treatment Network Cohort Study." Journal of neurovirology 22(3): 349–357. [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Chen MF, Gill AJ and Kolson DL (2014). "Neuropathogenesis of HIV-associated neurocognitive disorders: roles for immune activation, HIV blipping and viral tropism." Curr Opin HIV AIDS 9(6): 559–564. [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Cheng S, Wang Y, Zhang Z, Lv X, Gao GF, Shao Y, Ma L and Li X (2016). "Enfuvirtide– PEG conjugate: A potent HIV fusion inhibitor with improved pharmacokinetic properties." European journal of medicinal chemistry 121: 232–237. [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. Cherry CL, Duncan AJ, Mackie KF, Wesselingh SL and Brew BJ (2008). "A report on the effect of commencing enfuvirtide on peripheral neuropathy." AIDS research and human retroviruses 24(8): 1027–1030. [DOI] [PubMed] [Google Scholar]
  28. Ciavatta VT, Bichler EK, Speigel IA, Elder CC, Teng SL, Tyor WR and García PS (2017). "In vitro and Ex vivo Neurotoxic Effects of Efavirenz are Greater than Those of Other Common Antiretrovirals." Neurochemical research 42(11): 3220–3232. [DOI] [PubMed] [Google Scholar]
  29. Ciccarelli N, Fabbiani M, Di Giambenedetto S, Fanti I, Baldonero E, Bracciale L, Tamburrini E, Cauda R, De Luca A and Silveri MC (2011). "Efavirenz associated with cognitive disorders in otherwise asymptomatic HIV-infected patients." Neurology 76(16): 1403–1409. [DOI] [PubMed] [Google Scholar]
  30. Coban H, Robertson K, Smurzynski M, Krishnan S, Wu K, Bosch RJ, Collier AC and Ellis RJ (2017). "Impact of aging on neurocognitive performance in previously antiretroviral-naive HIV-infected individuals on their first suppressive regimen." AIDS (London, England) 31(11): 1565–1571. [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Crawford KW, Spritzler J, Kalayjian RC, Parsons T, Landay A, Pollard R, Stocker V, Lederman MM and Flexner C (2010). "Age-related changes in plasma concentrations of the HIV protease inhibitor lopinavir." AIDS research and human retroviruses 26(6): 635–643. [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Cross HM, Combrinck MI and Joska JA (2013). "HIV-associated neurocognitive disorders: antiretroviral regimen, central nervous system penetration effectiveness, and cognitive outcomes." South African Medical Journal 103(10): 758–762. [DOI] [PubMed] [Google Scholar]
  33. Cross SA, Cook DR, Chi AW, Vance PJ, Kolson LL, Wong BJ, Jordan-Sciutto KL and Kolson DL (2011). "Dimethyl fumarate, an immune modulator and inducer of the antioxidant response, suppresses HIV replication and macrophage-mediated neurotoxicity: a novel candidate for HIV neuroprotection." The Journal of Immunology 187(10): 5015–5025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Cross SA and Kolson DL (2017). Therapeutic Considerations in HIV-Associated Neurocognitive Disorders. Neuroimmune Pharmacology, Springer: 737–751. [Google Scholar]
  35. Curtis L, Nichols G, Stainsby C, Lim J, Aylott A, Wynne B, Clark A, Bloch M, Maechler G and Martin-Carpenter L (2014). "Dolutegravir: clinical and laboratory safety in integrase inhibitor–naive patients." HIV clinical trials 15(5): 199–208. [DOI] [PubMed] [Google Scholar]
  36. Cysique LA, Waters EK and Brew BJ (2011). "Central nervous system antiretroviral efficacy in HIV infection: a qualitative and quantitative review and implications for future research." BMC neurology 11(1): 148. [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. D'Antoni ML, Paul RH, Mitchell BI, Kohorn L, Fischer L, Lefebvre E, Seyedkazemi S, Nakamoto BK, Walker M, Kallianpur KJ, Ogata-Arakaki D, Ndhlovu LC and Shikuma C (2018). "Improved Cognitive Performance and Reduced Monocyte Activation in Virally Suppressed Chronic HIV After Dual CCR2 and CCR5 Antagonism." J Acquir Immune Defic Syndr 79(1): 108–116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Dahl V, Lee E, Peterson J, Spudich SS, Leppla I, Sinclair E, Fuchs D, Palmer S and Price RW (2011). "Raltegravir treatment intensification does not alter cerebrospinal fluid HIV-1 infection or immunoactivation in subjects on suppressive therapy." The Journal of infectious diseases 204(12): 1936–1945. [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. Danner SA, Carr A, Leonard JM, Lehman LM, Gudiol F, Gonzales J, Raventos A, Rubio R, Bouza E and Pintado V (1995). "A short-term study of the safety, pharmacokinetics, and efficacy of ritonavir, an inhibitor of HIV-1 protease." New England Journal of Medicine 333(23): 1528–1534. [DOI] [PubMed] [Google Scholar]
  40. Decloedt EH, Rosenkranz B, Maartens G and Joska J (2015). "Central nervous system penetration of antiretroviral drugs: pharmacokinetic, pharmacodynamic and pharmacogenomic considerations." Clinical pharmacokinetics 54(6): 581–598. [DOI] [PubMed] [Google Scholar]
  41. del Mar Gutierrez M, Mateo MG, Vidal F and Domingo P (2014). "Drug safety profile of integrase strand transfer inhibitors." Expert opinion on drug safety 13(4): 431–445. [DOI] [PubMed] [Google Scholar]
  42. Drake SM (2000). "NNRTIs–a new class of drugs for HIV." Journal of Antimicrobial Chemotherapy 45(4): 417–420. [DOI] [PubMed] [Google Scholar]
  43. Ekins S, Mathews P, Saito EK, Diaz N, Naylor D, Chung J and McMurtray AM (2017). "α7-Nicotinic acetylcholine receptor inhibition by indinavir: Implications for cognitive dysfunction in treated HIV disease." AIDS 31(8): 1083–1089. [DOI] [PubMed] [Google Scholar]
  44. Ellis RJ, Marquie-Beck J, Delaney P, Alexander T, Clifford DB, McArthur JC, Simpson DM, Ake C, Collier AC and Gelman BB (2008). "Human immunodeficiency virus protease inhibitors and risk for peripheral neuropathy." Annals of Neurology: Official Journal of the American Neurological Association and the Child Neurology Society 64(5): 566–572. [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. Emmelkamp J and Rockstroh J (2007). "CCR5 antagonists: comparison of efficacy, side effects, pharmacokinetics and interactions–review of the literature." Eur J Med Res 12(9): 409–417. [PubMed] [Google Scholar]
  46. Emmelkamp JM and Rockstroh JK (2008). "Maraviroc, risks and benefits: a review of the clinical literature." Expert opinion on drug safety 7(5): 559–569. [DOI] [PubMed] [Google Scholar]
  47. Erdő F, Denes L and de Lange E (2017). "Age-associated physiological and pathological changes at the blood–brain barrier: a review." Journal of Cerebral Blood Flow & Metabolism 37(1): 4–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. Etgen T, Chonchol M, Förstl H and Sander D (2012). "Chronic kidney disease and cognitive impairment: a systematic review and meta-analysis." American journal of nephrology 35(5): 474–482. [DOI] [PubMed] [Google Scholar]
  49. Etherton MR, Lyons JL and Ard KL (2015). "HIV-associated neurocognitive disorders and antiretroviral therapy: current concepts and controversies." Current infectious disease reports 17(6): 28. [DOI] [PubMed] [Google Scholar]
  50. Fatkenheuer G, Pozniak AL, Johnson MA, Plettenberg A, Staszewski S, Hoepelman AI, Saag MS, Goebel FD, Rockstroh JK, Dezube BJ, Jenkins TM, Medhurst C, Sullivan JF, Ridgway C, Abel S, James IT, Youle M and van der Ryst E (2005). "Efficacy of short-term monotherapy with maraviroc, a new CCR5 antagonist, in patients infected with HIV-1." Nat Med 11(11): 1170–1172. [DOI] [PubMed] [Google Scholar]
  51. Ferretti F, Gisslen M, Cinque P and Price RW (2015). "Cerebrospinal fluid HIV escape from antiretroviral therapy." Current HIV/AIDS Reports 12(2): 280–288. [DOI] [PubMed] [Google Scholar]
  52. Fettiplace A, Stainsby C, Winston A, Givens N, Puccini S, Vannappagari V, Hsu R, Fusco J, Quercia R, Aboud M and Curtis L (2017). "Psychiatric Symptoms in Patients Receiving Dolutegravir." J Acquir Immune Defic Syndr 74(4): 423–431. [DOI] [PMC free article] [PubMed] [Google Scholar]
  53. Finsterer J (2012). "Cognitive dysfunction in mitochondrial disorders." Acta Neurologica Scandinavica 126(1): 1–11. [DOI] [PubMed] [Google Scholar]
  54. Flexner C (1998). "HIV-protease inhibitors." New England Journal of Medicine 338(18): 1281–1293. [DOI] [PubMed] [Google Scholar]
  55. Fung HB and Guo Y (2004). "Enfuvirtide: a fusion inhibitor for the treatment of HIV infection." Clinical therapeutics 26(3): 352–378. [DOI] [PubMed] [Google Scholar]
  56. Gabrielian A, MacCumber MM, Kukuyev A, Mitsuyasu R, Holland GN and Sarraf D (2013). "Didanosine-associated retinal toxicity in adults infected with human immunodeficiency virus." JAMA ophthalmology 131(2): 255–259. [DOI] [PubMed] [Google Scholar]
  57. Gallant J, Lazzarin A, Mills A, Orkin C, Podzamczer D, Tebas P, Girard P-M, Brar I, Daar ES and Wohl D (2017). "Bictegravir, emtricitabine, and tenofovir alafenamide versus dolutegravir, abacavir, and lamivudine for initial treatment of HIV-1 infection (GS-US-380-1489): a double-blind, multicentre, phase 3, randomised controlled non-inferiority trial." The Lancet 390(10107): 2063–2072. [DOI] [PubMed] [Google Scholar]
  58. Gallego L, Barreiro P, del Rio R, Gonzalez de Requena D, Rodriguez-Albarino A, Gonzalez-Lahoz J and Soriano V (2004). "Analyzing sleep abnormalities in HIV-infected patients treated with Efavirenz." Clin Infect Dis 38(3): 430–432. [DOI] [PubMed] [Google Scholar]
  59. Gerena Y, Skolasky RL, Velez JM, Toro-Nieves D, Mayo R, Nath A and Wojna V (2012). "Soluble and cell-associated insulin receptor dysfunction correlates with severity of HAND in HIV-infected women." PLoS One 7(5): e37358. [DOI] [PMC free article] [PubMed] [Google Scholar]
  60. Gisslén M and Hunt PW (2019). "Antiretroviral Treatment of Acute HIV Infection Normalizes Levels of Cerebrospinal Fluid Markers of Central Nervous System (CNS) Inflammation: A Consequence of a Reduced CNS Reservoir?” The Journal of Infectious Diseases. [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Gray J and Young B (2009). "Acute onset insomnia associated with the initiation of raltegravir: a report of two cases and literature review." AIDS Patient Care and STDs 23(9): 689–690. [DOI] [PubMed] [Google Scholar]
  62. Grilo NM, Joao Correia M, Miranda JP, Cipriano M, Serpa J, Matilde Marques M, Monteiro EC, Antunes AMM, Diogo LN and Pereira SA (2017). "Unmasking efavirenz neurotoxicity: Time matters to the underlying mechanisms." Eur J Pharm Sci 105: 47–54. [DOI] [PubMed] [Google Scholar]
  63. Harris M (2018). "What did we learn from the bictegravir switch studies?” Lancet HIV 5(7): e336–e337. [DOI] [PubMed] [Google Scholar]
  64. Harris M, Larsen G and Montaner JS (2008). "Exacerbation of depression associated with starting raltegravir: a report of four cases." Aids 22(14): 1890–1892. [DOI] [PubMed] [Google Scholar]
  65. Heaton R, Clifford D, Franklin D, Woods S, Ake C, Vaida F, Ellis R, Letendre S, Marcotte T and Atkinson J (2010). "HIV-associated neurocognitive disorders persist in the era of potent antiretroviral therapy CHARTER Study." Neurology 75(23): 2087–2096. [DOI] [PMC free article] [PubMed] [Google Scholar]
  66. Heaton RK, Franklin DR, Ellis RJ, McCutchan JA, Letendre SL, LeBlanc S, Corkran SH, Duarte NA, Clifford DB and Woods SP (2011). "HIV-associated neurocognitive disorders before and during the era of combination antiretroviral therapy: differences in rates, nature, and predictors." Journal of neurovirology 17(1): 3–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  67. Hill A, van der Lugt J, Sawyer W and Boffito M (2009). "How much ritonavir is needed to boost protease inhibitors? Systematic review of 17 dose-ranging pharmacokinetic trials." Aids 23(17): 2237–2245. [DOI] [PubMed] [Google Scholar]
  68. Hoffmann C, Welz T, Sabranski M, Kolb M, Wolf E, Stellbrink HJ and Wyen C (2017). "Higher rates of neuropsychiatric adverse events leading to dolutegravir discontinuation in women and older patients." HIV medicine 18(1): 56–63. [DOI] [PubMed] [Google Scholar]
  69. Hsu A, Granneman GR and Bertz RJ (1998). "Ritonavir." Clinical pharmacokinetics 35(4): 275–291. [DOI] [PubMed] [Google Scholar]
  70. Hung K-M, Chen P-C, Hsieh H-C and Calkins MJ (2017). "Mitochondrial defects arise from nucleoside/nucleotide reverse transcriptase inhibitors in neurons: Potential contribution to HIV-associated neurocognitive disorders." Biochimica et Biophysica Acta (BBA)-Molecular Basis of Disease 1863(2): 406–413. [DOI] [PubMed] [Google Scholar]
  71. Jacobson JM, Kuritzkes DR, Godofsky E, DeJesus E, Larson JA, Weinheimer SP and Lewis ST (2009). "Safety, pharmacokinetics, and antiretroviral activity of multiple doses of ibalizumab (formerly TNX-355), an anti-CD4 monoclonal antibody, in human immunodeficiency virus type 1-infected adults." Antimicrobial agents and chemotherapy 53(2): 450–457. [DOI] [PMC free article] [PubMed] [Google Scholar]
  72. Jensen BK, Monnerie H, Mannell MV, Gannon PJ, Espinoza CA, Erickson MA, Bruce-Keller AJ, Gelman BB, Briand LA and Pierce RC (2015). "Altered oligodendrocyte maturation and myelin maintenance: The role of antiretrovirals in HIV-associated neurocognitive disorders." Journal of Neuropathology & Experimental Neurology 74(11): 1093–1118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  73. Joseph J, Cinque P, Colosi D, Dravid A, Ene L, Fox H, Gabuzda D, Gisslen M, Joseph SB and Letendre S (2016). "Highlights of the global HIV-1 CSF escape consortium meeting, 9 June 2016, Bethesda, MD, USA." Journal of virus eradication 2(4): 243. [PMC free article] [PubMed] [Google Scholar]
  74. Kakuda TN (2000). "Pharmacology of nucleoside and nucleotide reverse transcriptase inhibitor-induced mitochondrial toxicity." Clin Ther 22(6): 685–708. [DOI] [PubMed] [Google Scholar]
  75. Kenedi CA and Goforth HW (2011). "A systematic review of the psychiatric side-effects of efavirenz." AIDS Behav 15(8): 1803–1818. [DOI] [PubMed] [Google Scholar]
  76. Kirk JB and Goetz MB (2009). "Human immunodeficiency virus in an aging population, a complication of success." J Am Geriatr Soc 57(11): 2129–2138. [DOI] [PubMed] [Google Scholar]
  77. Klotz U (2009). "Pharmacokinetics and drug metabolism in the elderly." Drug metabolism reviews 41(2): 67–76. [DOI] [PubMed] [Google Scholar]
  78. Kohler JJ and Lewis W (2007). "A brief overview of mechanisms of mitochondrial toxicity from NRTIs." Environ Mol Mutagen 48(3-4): 166–172. [DOI] [PubMed] [Google Scholar]
  79. Kumar GN, Rodrigues AD, Buko AM and Denissen JF (1996). "Cytochrome P450-mediated metabolism of the HIV-1 protease inhibitor ritonavir (ABT-538) in human liver microsomes." Journal of Pharmacology and Experimental Therapeutics 277(1): 423–431. [PubMed] [Google Scholar]
  80. LaBonte J, Lebbos J and Kirkpatrick P (2003). Enfuvirtide, Nature Publishing Group. [DOI] [PubMed] [Google Scholar]
  81. Lalezari JP, Henry K, O'Hearn M, Montaner JS, Piliero PJ, Trottier B, Walmsley S, Cohen C, Kuritzkes DR and Eron JJ Jr (2003). "Enfuvirtide, an HIV-1 fusion inhibitor, for drug-resistant HIV infection in North and South America." New England Journal of Medicine 348(22): 2175–2185. [DOI] [PubMed] [Google Scholar]
  82. Larson KB, Wang K, Delille C, Otofokun I and Acosta EP (2014). "Pharmacokinetic enhancers in HIV therapeutics." Clinical pharmacokinetics 53(10): 865–872. [DOI] [PubMed] [Google Scholar]
  83. Latronico T, Pati I, Ciavarella R, Fasano A, Mengoni F, Lichtner M, Vullo V, Mastroianni CM and Liuzzi GM (2018). "In vitro effect of antiretroviral drugs on cultured primary astrocytes: analysis of neurotoxicity and matrix metalloproteinase inhibition." Journal of neurochemistry 144(3): 271–284. [DOI] [PubMed] [Google Scholar]
  84. Lee FJ, Amin J, Bloch M, Pett SL, Marriott D and Carr A (2013). "Skeletal muscle toxicity associated with raltegravir-based combination antiretroviral therapy in HIV-infected adults." J Acquir Immune Defic Svndr 62(5): 525–533. [DOI] [PubMed] [Google Scholar]
  85. Lennox JL, DeJesus E, Berger DS, Lazzarin A, Pollard RB, Madruga JVR, Zhao J, Wan H, Gilbert CL and Teppler H (2010). "Raltegravir versus efavirenz regimens in treatment-naive HIV-l–infected patients: 96-week efficacy, durability, subgroup, safety, and metabolic analyses." Journal of acquired immune deficiency syndromes (1999) 55(1): 39. [DOI] [PMC free article] [PubMed] [Google Scholar]
  86. Letendre S (2011). "Central nervous system complications in HIV disease: HIV-associated neurocognitive disorder." Topics in antiviral medicine 19(4): 137–142. [PMC free article] [PubMed] [Google Scholar]
  87. Letendre S, Marquie-Beck J, Capparelli E, Best B, Clifford D, Collier AC, Gelman BB, McArthur JC, McCutchan JA and Morgello S (2008). "Validation of the CNS penetration-effectiveness rank for quantifying antiretroviral penetration into the central nervous system." Archives of neurology 65(1): 65–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
  88. Letendre SL, Ellis RJ, Ances BM and McCutchan JA (2010). "Neurologic complications of HIV disease and their treatment." Topics in HIV medicine: a publication of the International AIDS Society, USA 18(2): 45. [PMC free article] [PubMed] [Google Scholar]
  89. Letendre SL, Mills AM, Tashima KT, Thomas DA, Min SS, Chen S, Song IH and Piscitelli SC (2014). "ING116070: A Study of the Pharmacokinetics and Antiviral Activity of Dolutegravir in Cerebrospinal Fluid in HIV-1–Infected, Antiretroviral Therapy–Naive Subjects." Clinical Infectious Diseases 59(7): 1032–1037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  90. Lewis W, Day BJ and Copeland WC (2003). "Mitochondrial toxicity of NRTI antiviral drugs: an integrated cellular perspective." Nat Rev Drug Discov 2(10): 812–822. [DOI] [PubMed] [Google Scholar]
  91. Lisi L, Tramutola A, De Luca A, Navarra P and Dello Russo C (2012). "Modulatory effects of the CCR5 antagonist maraviroc on microglial pro-inflammatory activation elicited by gp120." Journal of neurochemistry 120(1): 106–114. [DOI] [PubMed] [Google Scholar]
  92. Louboutin J-P and Strayer DS (2018). Gene Delivery of Antioxidant Enzymes in HIV-1-Associated Neurocognitive Disorder. HIV/AIDS, Elsevier: 107–123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  93. Lv Z, Chu Y and Wang Y (2015). "HIV protease inhibitors: a review of molecular selectivity and toxicity." HIV/AIDS (Auckland, NZ) 7: 95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  94. Ma Q, Vaida F, Wong J, Sanders CA, Kao Y.-t., Croteau D, Clifford DB, Collier AC, Gelman BB and Marra CM (2016). "Long-term efavirenz use is associated with worse neurocognitive functioning in HIV-infected patients." Journal of neurovirology 22(2): 170–178. [DOI] [PMC free article] [PubMed] [Google Scholar]
  95. Manfredi R and Sabbatani S (2006). "A novel antiretroviral class (fusion inhibitors) in the management of HIV infection. Present features and future perspectives of enfuvirtide (T-20)." Current medicinal chemistry 13(20): 2369–2384. [DOI] [PubMed] [Google Scholar]
  96. Mangoni AA and Jackson SH (2004). "Age-related changes in pharmacokinetics and pharmacodynamics: basic principles and practical applications." British journal of clinical pharmacology 57(1): 6–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  97. Marcus JL, Chao CR, Leyden WA, Xu L, Quesenberry CP Jr, Klein DB, Towner WJ, Horberg MA and Silverberg MJ (2016). "Narrowing the gap in life expectancy between HIV-infected and HIV-uninfected individuals with access to care." Journal of acquired immune deficiency syndromes (1999) 73(1): 39. [DOI] [PMC free article] [PubMed] [Google Scholar]
  98. Margolis AM, Heverling H, Pham PA and Stolbach A (2014). "A review of the toxicity of HIV medications." Journal of Medical Toxicology 10(1): 26–39. [DOI] [PMC free article] [PubMed] [Google Scholar]
  99. Markowitz M, Saag M, Powderly WG, Hurley AM, Hsu A, Valdes JM, Henry D, Sattler F, Marca AL and Leonard JM (1995). "A preliminary study of ritonavir, an inhibitor of HIV-1 protease, to treat HIV-1 infection." New England Journal of Medicine 333(23): 1534–1540. [DOI] [PubMed] [Google Scholar]
  100. Marra CM, Zhao Y, Clifford DB, Letendre S, Evans S, Henry K, Ellis RJ, Rodriguez B, Coombs RW and Schifitto G (2009). "Impact of combination antiretroviral therapy on cerebrospinal fluid HIV RNA and neurocognitive performance." AIDS (London, England) 23(11): 1359. [DOI] [PMC free article] [PubMed] [Google Scholar]
  101. Marzolini C, Gibbons S, Khoo S and Back D (2016). "Cobicistat versus ritonavir boosting and differences in the drug–drug interaction profiles with co-medications." Journal of Antimicrobial Chemotherapy 71(7): 1755–1758. [DOI] [PubMed] [Google Scholar]
  102. Meeker RB, Robertson K and Power C (2014). "Neurotoxic Consequences of Antiretroviral Therapies." Encyclopedia of AIDS: 1–7. [Google Scholar]
  103. Meulendyke KA, Queen SE, Engle EL, Shirk EN, Liu J, Steiner JP, Nath A, Tarwater PM, Graham DR and Mankowski JL (2014). "Combination fluconazole/paroxetine treatment is neuroprotective despite ongoing neuroinflammation and viral replication in an SIV model of HIV neurological disease." Journal of neurovirology 20(6): 591–602. [DOI] [PMC free article] [PubMed] [Google Scholar]
  104. Michelet C, Ruffault A, Sébille V, Arvieux C, Jaccard P, Raffi F, Bazin C, Chapplain J-M, Chauvin J-P and Dohin E (2001). "Ritonavir-saquinavir dual protease inhibitor compared to ritonavir alone in human immunodeficiency virus-infected patients." Antimicrobial agents and chemotherapy 45(12): 3393–3402. [DOI] [PMC free article] [PubMed] [Google Scholar]
  105. Mollan KR, Smurzynski M, Eron JJ, Daar ES, Campbell TB, Sax PE, Gulick RM, Na L, O'keefe L and Robertson KR (2014). "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." Annals of internal medicine 161(1): 1–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  106. Monforte A. d. A., Lepri AC, Rezza G, Pezzotti P, Antinori A, Phillips AN, Angarano G, Colangeli V, De Luca A and Ippolito G (2000). "Insights into the reasons for discontinuation of the first highly active antiretroviral therapy (HAART) regimen in a cohort of antiretroviral naive patients." Aids 14(5): 499–507. [DOI] [PubMed] [Google Scholar]
  107. Moyle G, Fletcher C, Brown H, Mandalia S and Gazzard B (2006). "Changes in sleep quality and brain wave patterns following initiation of an efavirenz-containing triple antiretroviral regimen." HIV Med 7(4): 243–247. [DOI] [PubMed] [Google Scholar]
  108. Nakasujja N, Miyahara S, Evans S, Lee A, Musisi S, Katabira E, Robertson K, Ronald A, Clifford DB and Sacktor N (2013). "Randomized trial of minocycline in the treatment of HIV-associated cognitive impairment." Neurology 80(2): 196–202. [DOI] [PMC free article] [PubMed] [Google Scholar]
  109. Ndhlovu LC, Umaki T, Chew GM, Chow DC, Agsalda M, Kallianpur KJ, Paul R, Zhang G, Ho E and Hanks N (2014). "Treatment intensification with maraviroc (CCR5 antagonist) leads to declines in CD16-expressing monocytes in cART-suppressed chronic HIV-infected subjects and is associated with improvements in neurocognitive test performance: implications for HIV-associated neurocognitive disease (HAND)." Journal of neurovirology 20(6): 571–582. [DOI] [PMC free article] [PubMed] [Google Scholar]
  110. Nguyen BYT, Isaacs RD, Teppler H, Leavitt RY, Sklar P, Iwamoto M, Wenning LA, Miller MD, Chen J and Kemp R (2011). "Raltegravir: the first HIV-1 integrase strand transfer inhibitor in the HIV armamentarium." Annals of the New York Academy of Sciences 1222(1): 83–89. [DOI] [PubMed] [Google Scholar]
  111. Nooka S and Ghorpade A (2017). "HIV-1-associated inflammation and antiretroviral therapy regulate astrocyte endoplasmic reticulum stress responses." Cell death discovery 3: 17061. [DOI] [PMC free article] [PubMed] [Google Scholar]
  112. Nooka S and Ghorpade A (2018). "Organellar stress intersects the astrocyte endoplasmic reticulum, mitochondria and nucleolus in HIV associated neurodegeneration." Cell death & disease 9(3): 317. [DOI] [PMC free article] [PubMed] [Google Scholar]
  113. Oldfield V, Keating GM and Plosker G (2005). "Enfuvirtide: a review of its use in the management of HIV infection." Drugs 65(8): 1139–1160. [DOI] [PubMed] [Google Scholar]
  114. Pakos-Zebrucka K, Koryga I, Mnich K, Ljujic M, Samali A and Gorman AM (2016). "The integrated stress response." EMBO reports 17(10): 1374–1395. [DOI] [PMC free article] [PubMed] [Google Scholar]
  115. Patel P, L. S (2018). Drug Interactions in HIV: Protease and Integrase Inhibitors. Drug Interactions in Infectious Diseases: Antimicrobial Drug Interactions. K. J. Pai M., Gubbins P, Rodvold K, Humana Press, Cham. [Google Scholar]
  116. Payne B, Chadwick T, Blamire A, Anderson K, Parikh J, Qian J, Hynes A, Wilkinson J, Price D and E. o. S. t. L. R. i. I. C. F. i. E. t. P. s. team (2017). "Does efavirenz replacement improve neurological function in treated HIV infection?” HIV medicine 18(9): 690–695. [DOI] [PMC free article] [PubMed] [Google Scholar]
  117. Penafiel J, de Lazzari E, Padilla M, Rojas J, Gonzalez-Cordon A, Blanco JL, Blanch J, Marcos MA, Lonca M, Martinez-Rebollar M, Laguno M, Tricas A, Rodriguez A, Mallolas J, Gatell JM and Martinez E (2017). "Tolerability of integrase inhibitors in a real-life setting." J Antimicrob Chemother 72(6): 1752–1759. [DOI] [PubMed] [Google Scholar]
  118. Pérez-Valero I, Ellis R, Heaton R, Deutsch R, Franklin D, Clifford DB, Collier A, Gelman B, Marra C and McCutchan JA (2019). "Cerebrospinal fluid viral escape in aviremic HIV-infected patients receiving antiretroviral therapy: prevalence, risk factors and neurocognitive effects." AIDS 33(3): 475–481. [DOI] [PMC free article] [PubMed] [Google Scholar]
  119. Pettersen JA, Jones G, Worthington C, Krentz HB, Keppler OT, Hoke A, Gill MJ and Power C (2006). "Sensory neuropathy in human immunodeficiency virus/acquired immunodeficiency syndrome patients: Protease inhibitor–mediated neurotoxicity." Annals of Neurology: Official Journal of the American Neurological Association and the Child Neurology Society 59(5): 816–824. [DOI] [PubMed] [Google Scholar]
  120. Piotrowska A, Kwiatkowski K, Rojewska E, Makuch W and Mika J (2016). "Maraviroc reduces neuropathic pain through polarization of microglia and astroglia–evidence from in vivo and in vitro studies." Neuropharmacology 108: 207–219. [DOI] [PubMed] [Google Scholar]
  121. Poirier MC, Divi RL, Al-Harthi L, Olivero OA, Nguyen V, Walker B, Landay AL, Walker VE, Charurat M, Blattner WA, Women and G Infants Transmission Study (2003). "Long-term mitochondrial toxicity in HIV-uninfected infants born to HIV-infected mothers." J Acquir Immune Defic Syndr 33(2): 175–183. [DOI] [PubMed] [Google Scholar]
  122. Pokorná J, Machala L, Řezáčová P and Konvalinka J (2009). "Current and novel inhibitors of HIV protease." Viruses 1(3): 1209–1239. [DOI] [PMC free article] [PubMed] [Google Scholar]
  123. Pommier Y, Johnson AA and Marchand C (2005). "Integrase inhibitors to treat HIV/AIDS." Nature reviews Drug discovery 4(3): 236. [DOI] [PubMed] [Google Scholar]
  124. Rathbun RC and Rossi DR (2002). "Low-dose ritonavir for protease inhibitor pharmacokinetic enhancement." Annals of Pharmacotherapy 36(4): 702–706. [DOI] [PubMed] [Google Scholar]
  125. Rezaie-Majd A, Maca T, Bucek RA, Valent P, Müller MR, Husslein P, Kashanipour A, Minar E and Baghestanian M (2002). "Simvastatin reduces expression of cytokines interleukin-6, interleukin-8, and monocyte chemoattractant protein-1 in circulating monocytes from hypercholesterolemic patients." Arteriosclerosis, thrombosis, and vascular biology 22(7): 1194–1199. [DOI] [PubMed] [Google Scholar]
  126. Richman DD, Fischl MA, Grieco MH, Gottlieb MS, Volberding PA, Laskin OL, Leedom JM, Groopman JE, Mildvan D and Hirsch MS (1987). "The toxicity of azidothymidine (AZT) in the treatment of patients with AIDS and AIDS-related complex." New England Journal of Medicine 317(4): 192–197. [DOI] [PubMed] [Google Scholar]
  127. Riddler SA, Haubrich R, DiRienzo AG, Peeples L, Powderly WG, Klingman KL, Garren KW, George T, Rooney JF, Brizz B, Lalloo UG, Murphy RL, Swindells S, Havlir D, Mellors JW and A. C. T. G. S. A. Team (2008). "Class-sparing regimens for initial treatment of HIV-1 infection." N Engl J Med 358(20): 2095–2106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  128. Robertson K, Su Z, Margolis D, Krambrink A, Havlir D, Evans S, Skiest D and A. S. Team (2010). "Neurocognitive effects of treatment interruption in stable HIV-positive patients in an observational cohort." Neurology 74(16): 1260–1266. [DOI] [PMC free article] [PubMed] [Google Scholar]
  129. Rodriguez-Nóvoa S, Alvarez E, Labarga P and Soriano V (2010). "Renal toxicity associated with tenofovir use." Expert opinion on drug safety 9(4): 545–559. [DOI] [PubMed] [Google Scholar]
  130. Rublein JC, Eron JJ Jr, Butts JD and Raasch RH (1999). "Discontinuation rates for protease inhibitor regimens containing ritonavir 600 mg versus ritonavir 400 mg plus saquinavir 400 mg." Annals of Pharmacotherapy 33(9): 899–905. [DOI] [PubMed] [Google Scholar]
  131. Saag MS, Benson CA, Gandhi RT, Hoy JF, Landovitz RJ, Mugavero MJ, Sax PE, Smith DM, Thompson MA, Buchbinder SP, Del Rio C, Eron JJ Jr., Fatkenheuer G, Gunthard HF, Molina JM, Jacobsen DM and Volberding PA (2018). "Antiretroviral Drugs for Treatment and Prevention of HIV Infection in Adults: 2018 Recommendations of the International Antiviral Society-USA Panel." JAMA 320(4): 379–396. [DOI] [PMC free article] [PubMed] [Google Scholar]
  132. Sacktor N, Miyahara S, Deng L, Evans S, Schifitto G, Cohen BA, Paul R, Robertson K, Jarocki B and Scarsi K (2011). "Minocycline treatment for HIV-associated cognitive impairment: results from a randomized trial." Neurology 77(12): 1135–1142. [DOI] [PMC free article] [PubMed] [Google Scholar]
  133. Sacktor N, Skolasky RL, Moxley R, Wang S, Mielke MM, Munro C, Steiner J, Nath A, Haughey N and McArthur J (2018). "Paroxetine and fluconazole therapy for HIV-associated neurocognitive impairment: results from a double-blind, placebo-controlled trial." J Neurovirol 24(1): 16–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
  134. Sacktor N, Skolasky RL, Seaberg E, Munro C, Becker JT, Martin E, Ragin A, Levine A and Miller E (2016). "Prevalence of HIV-associated neurocognitive disorders in the Multicenter AIDS Cohort Study." Neurology 86(4): 334–340. [DOI] [PMC free article] [PubMed] [Google Scholar]
  135. Saracchini S, Vaccher E, Covezzi E, Tortorici G, Carbone A and Tirelli U (1989). "Lethal neurotoxicity associated to azidothymidine therapy." Journal of neurology, neurosurgery, and psychiatry 52(4): 544. [DOI] [PMC free article] [PubMed] [Google Scholar]
  136. Saraiya N, Kanagala V and Corpuz M (2018). "HIV-2 in the United States: rare but not forgotten." Aids 32(11): 1547–1549. [DOI] [PubMed] [Google Scholar]
  137. Sax PE, Pozniak A, Montes ML, Koenig E, DeJesus E, Stellbrink H-J, Antinori A, Workowski K, Slim J and Reynes J (2017). "Coformulated bictegravir, emtricitabine, and tenofovir alafenamide versus dolutegravir with emtricitabine and tenofovir alafenamide, for initial treatment of HIV-1 infection (GS-US-380–1490): a randomised, double-blind, multicentre, phase 3, non-inferiority trial." The Lancet 390(10107): 2073–2082. [DOI] [PubMed] [Google Scholar]
  138. Saylor D, Dickens AM, Sacktor N, Haughey N, Slusher B, Pletnikov M, Mankowski JL, Brown A, Volsky DJ and McArthur JC (2016). "HIV-associated neurocognitive disorder–pathogenesis and prospects for treatment." Nature Reviews Neurology 12(4): 234. [DOI] [PMC free article] [PubMed] [Google Scholar]
  139. Scherzer R and Shlipak MG (2015). "Risk factors: Individual assessment of CKD risk in HIV-positive patients." Nature Reviews Nephrology 11(7): 392. [DOI] [PubMed] [Google Scholar]
  140. Schifitto G, Navia BA, Yiannoutsos CT, Marra CM, Chang L, Ernst T, Jarvik JG, Miller EN, Singer EJ and Ellis RJ (2007). "Memantine and HIV-associated cognitive impairment: a neuropsychological and proton magnetic resonance spectroscopy study." Aids 21(14): 1877–1886. [DOI] [PubMed] [Google Scholar]
  141. Schifitto G, Zhang J, Evans S, Sacktor N, Simpson D, Millar L, Hung V, Miller E, Smith E and Ellis R (2007). "A multicenter trial of selegiline transdermal system for HIV-associated cognitive impairment." Neurology 69(13): 1314–1321. [DOI] [PubMed] [Google Scholar]
  142. Schweinsburg BC, Taylor MJ, Alhassoon ΟM, Gonzalez R, Brown GG, Ellis RJ, Letendre S, Videen JS, McCutchan JA and Patterson TL (2005). "Brain mitochondrial injury in human immunodeficiency virus-seropositive (HIV+) individuals taking nucleoside reverse transcriptase inhibitors." Journal of neurovirology 11(4): 356–364. [DOI] [PubMed] [Google Scholar]
  143. Seider TR, Luo X, Gongvatana A, Devlin KN, de la Monte SM, Chasman JD, Yan P, Tashima KT, Navia B and Cohen RA (2014). "Verbal memory declines more rapidly with age in HIV infected versus uninfected adults." Journal of clinical and experimental neuropsychology 36(4): 356–367. [DOI] [PMC free article] [PubMed] [Google Scholar]
  144. Shah A, Gangwani MR, Chaudhari NS, Glazyrin A, Bhat HK and Kumar A (2016). "Neurotoxicity in the post-HAART era: caution for the antiretroviral therapeutics." Neurotoxicity research 30(4): 677–697. [DOI] [PMC free article] [PubMed] [Google Scholar]
  145. Singer EJ and Nemanim NM (2017). The Persistence of HIV-Associated Neurocognitive Disorder (HAND) in the Era of Combined Antiretroviral Therapy (cART). Global Virology II-HIV and NeuroAIDS. Springer: 375–403. [Google Scholar]
  146. Smith RL, de Boer R, Brul S, Budovskaya Y and van Spek H (2012). "Premature and accelerated aging: HIV or HAART?” Front Genet 3: 328. [DOI] [PMC free article] [PubMed] [Google Scholar]
  147. Smurzynski M, Wu K, Letendre S, Robertson K, Bosch RJ, Clifford DB, Evans S, Collier AC, Taylor M and Ellis R (2011). "Effects of central nervous system antiretroviral penetration on cognitive functioning in the ALLRT cohort." AIDS (London. England) 25(3): 357. [DOI] [PMC free article] [PubMed] [Google Scholar]
  148. Soontornniyomkij V, Umlauf A, Chung SA, Cochran ML, Soontornniyomkij B, Gouaux B, Toperoff W, Moore DJ, Masliah E and Ellis RJ (2014). "HIV protease inhibitor exposure predicts cerebral small vessel disease." AIDS (London, England) 28(9): 1297. [DOI] [PMC free article] [PubMed] [Google Scholar]
  149. Stauch KL, Emanuel K, Lamberty BG, Morsey B and Fox HS (2017). "Central nervous system-penetrating antiretrovirals impair energetic reserve in striatal nerve terminals." Journal of neurovirology 23(6): 795–807. [DOI] [PMC free article] [PubMed] [Google Scholar]
  150. Steigbigel RT, Cooper DA, Teppler H, Eron JJ, Gatell JM, Kumar PN, Rockstroh JK, Schechter M, Katlama C and Markowitz M (2010). "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." Clinical Infectious Diseases 50(4): 605–612. [DOI] [PMC free article] [PubMed] [Google Scholar]
  151. Stern AL, Lee RN, Panvelker N, Li J, Harowitz J, Jordan-Sciutto KL and Akay-Espinoza C (2018). "Differential Effects of Antiretroviral Drugs on Neurons In Vitro: Roles for Oxidative Stress and Integrated Stress Response." Journal of Neuroimmune Pharmacology 13(1): 64–76. [DOI] [PMC free article] [PubMed] [Google Scholar]
  152. Tarr PE and Telenti A (2010). "Genetic screening for metabolic and age-related complications in HIV-infected persons." F1000 medicine reports 2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  153. Tiraboschi JM, Muñoz-Moreno JA, Puertas M, Alonso-Villaverde C, Prats A, Ferrer E, Rozas N, Masó M, Ouchi D and Martinez-Picado J (2015). "Viral and inflammatory markers in cerebrospinal fluid of patients with HIV-l-associated neurocognitive impairment during antiretroviral treatment switch." HIV medicine 16(6): 388–392. [DOI] [PubMed] [Google Scholar]
  154. Tozzi V, Balestra P, Salvatori MF, Vlassi C, Liuzzi G, Giancola ML, Giulianelli M, Narciso P and Antinori A (2009). "Changes in cognition during antiretroviral therapy: comparison of 2 different ranking systems to measure antiretroviral drug efficacy on HIV-associated neurocognitive disorders." JAIDS Journal of Acquired Immune Deficiency Syndromes 52(1): 56–63. [DOI] [PubMed] [Google Scholar]
  155. Treisman GJ and Soudry O (2016). "Neuropsychiatric effects of HIV antiviral medications." Drug safety 39(10): 945–957. [DOI] [PubMed] [Google Scholar]
  156. Tseng A, Hughes CA, Wu J, Seet J and Phillips EJ (2017). "Cobicistat versus ritonavir: similar pharmacokinetic enhancers but some important differences." Annals of Pharmacotherapy 51(11): 1008–1022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  157. Turjanski N and Lloyd GG (2005). "Psychiatric side-effects of medications: recent developments." Advances in Psychiatric Treatment 11(1): 58–70. [Google Scholar]
  158. Underwood J, Robertson KR and Winston A (2015). "Could antiretroviral neurotoxicity play a role in the pathogenesis of cognitive impairment in treated HIV disease?” Aids 29(3): 253–261. [DOI] [PubMed] [Google Scholar]
  159. Valcour V, Chalermchai T, Sailasuta N, Marovich M, Lerdlum S, Suttichom D, Suwanwela NC, Jagodzinski L, Michael N and Spudich S (2012). "Central nervous system viral invasion and inflammation during acute HIV infection." The Journal of infectious diseases 206(2): 275–282. [DOI] [PMC free article] [PubMed] [Google Scholar]
  160. Velichkovska M, Surnar B, Nair M, Dhar S and Toborek M (2018). "TARGETED MITOCHONDRIAL COQ10 DELIVERY ATTENUATES ANTIRETROVIRAL DRUG-INDUCED SENESCENCE OF NEURAL PROGENITOR CELLS." Molecular pharmaceutics. [DOI] [PMC free article] [PubMed] [Google Scholar]
  161. Vivithanaporn P, Asahchop EL, Acharjee S, Baker GB and Power C (2016). "HIV protease inhibitors disrupt astrocytic glutamate transporter function and neurobehavioral performance." AIDS (London, England) 30(4): 543. [DOI] [PMC free article] [PubMed] [Google Scholar]
  162. White AJ (2001). "Mitochondrial toxicity and HIV therapy." Sexually transmitted infections 77(3): 158–173. [DOI] [PMC free article] [PubMed] [Google Scholar]
  163. Winston A, Jose S, Gibbons S, Back D, Stohr W, Post F, Fisher M, Gazzard B, Nelson M and Gilson R (2013). "Effects of age on antiretroviral plasma drug concentration in HIV-infected subjects undergoing routine therapeutic drug monitoring." Journal of Antimicrobial Chemotherapy 68(6): 1354–1359. [DOI] [PubMed] [Google Scholar]
  164. Xu L and Desai MC (2009). "Pharmacokinetic enhancers for HIV drugs." Current opinion in investigational drugs (London, England: 2000) 10(8): 775–786. [PubMed] [Google Scholar]
  165. Xu L, Liu H, Murray BP, Callebaut C, Lee MS, Hong A, Strickley RG, Tsai LK, Stray KM and Wang Y (2010). "Cobicistat (GS-9350): a potent and selective inhibitor of human CYP3A as a novel pharmacoenhancer." ACS medicinal chemistry letters 1(5): 209–213. [DOI] [PMC free article] [PubMed] [Google Scholar]
  166. Yilmaz A, Verhofstede C, D'Avolio A, Watson V, Hagberg L, Fuchs D, Svennerholm B and Gisslén M (2010). "Treatment intensification has no effect on the HIV-1 central nervous system infection in patients on suppressive antiretroviral therapy." JAIDS Journal of Acquired Immune Deficiency Syndromes 55(5): 590–596. [DOI] [PubMed] [Google Scholar]
  167. Yost R, Pasquale TR and Sahloff EG (2009). "Maraviroc: a coreceptor CCR5 antagonist for management of HIV infection." American Journal of Health-System Pharmacy 66(8): 715–726. [DOI] [PubMed] [Google Scholar]
  168. Yu K and Daar ES (2000). "Dual protease inhibitor therapy in the management of the HIV-1." Expert opinion on pharmacotherapy 1(7): 1331–1342. [DOI] [PubMed] [Google Scholar]
  169. Zash R, Jacobson DL, Diseko M, Mayondi G, Mmalane M, Essex M, Petlo C, Lockman S, Makhema J and Shapiro RL (2017). "Comparative safety of antiretroviral treatment regimens in pregnancy." JAMA pediatrics 171(10): e172222–e172222. [DOI] [PMC free article] [PubMed] [Google Scholar]
  170. Zash R, Makhema J and Shapiro RL (2018). "Neural-tube defects with dolutegravir treatment from the time of conception." New England Journal of Medicine 379(10): 979–981. [DOI] [PMC free article] [PubMed] [Google Scholar]
  171. Zhang Y, Song F, Gao Z, Ding W, Qiao L, Yang S, Chen X, Jin R and Chen D (2014). "Long-term exposure of mice to nucleoside analogues disrupts mitochondrial DNA maintenance in cortical neurons." PloS one 9(1): e85637. [DOI] [PMC free article] [PubMed] [Google Scholar]
  172. Zhou X-J, Havlir DV, Richman DD, Acosta EP, Hirsch M, Collier AC, Tebas P, Sommadossi J-P and A. C. T. G. S. Investigators (2000). "Plasma population pharmacokinetics and penetration into cerebrospinal fluid of indinavir in combination with zidovudine and lamivudine in HIV-1-infected patients." Aids 14(18): 2869–2876. [DOI] [PubMed] [Google Scholar]

RESOURCES