Abstract
Introduction:
Modern antiretroviral therapy (ART) has revolutionized HIV treatment. ART regimens are now highly efficacious, well-tolerated, safe, often with one multi-drug pill, once-daily regimens available. However, clinical challenges persist in managing ART in persons with HIV (PWH), such as drug-drug interactions, side effects, pregnancy, co-morbidities and adherence.
Areas Covered:
In this review, we discuss the ongoing challenges of ART for adults in the United States. We review the difficulties of initiating ART and maintaining therapy throughout adulthood and discuss new agents and strategies under investigation to address these issues. A PubMed search was utilized to identify relevant publications and guidelines through July 2019.
Expert Opinion:
Challenges persist in initiation and maintenance of ART. An individual’s coexisting medical, social and personal factors must be considered in selecting and continuing ART to ensure safety, tolerability, and efficacy throughout adulthood. Continued development of new therapeutics and novel approaches to ART, such as long acting drugs or dual therapy, are needed to respond many of these challenges. In addition, future research must address therapeutic disparities for populations historically underrepresented in clinical trials, including women, people aging with HIV, and those with complex comorbidities.
Keywords: HIV, Antiretroviral therapy, women, aging, drug-drug interactions, non-AIDS-related comorbidities, Adults, AIDS
1. Introduction
The advent of suppressive combination antiretroviral therapy (ART) has transformed HIV into a controllable, chronic disease. With modern ART, life expectancy for persons with HIV (PWH) now nears that of the general population[1]. Despite these advances, current optimal management of HIV requires that PWH remain on lifetime ART. Initiation and maintenance of ART throughout adulthood provide unique challenges for PWH and their care teams. In this paper, we provide an overview of the major challenges of ART management for adults with HIV in the United States. Our review and recommendations are based on the most recent United States ART guidelines and a PubMed search of articles available through July 2019.
2. History of Antiretroviral Therapy
Since the AIDS epidemic was first recognized in the United States in the early 1980s[2, 3], advances in ART have revolutionized care for PWH. Four decades of research have led to development of various antiretroviral medications that target key points in the HIV life cycle (Figure 1)[4]. Nucleoside analogues, which serve to terminate DNA synthesis, were the first class of medications to demonstrate activity against HIV [5]. Azidothymidine (AZT), originally created as a cancer drug, displayed potent activity against HIV but when used alone was associated with rapid development of viral resistance[6]. Additional nucleoside/nucleotide reverse transcriptase inhibitors (NRTIs) were developed, but viral resistance continued to limit their duration of activity. Over the next decade, new classes of medications including protease inhibitors (PIs), first approved in 1995, and non-nucleoside reverse transcriptase inhibitors (NNRTIs), first approved in 1996, became available[7]. By 1996, triple-drug combination, highly active antiretroviral therapy (HAART) comprised of 2 NRTIs plus either a PI or NNRTI became the new treatment standard with a subsequent marked decline in HIV-associated mortality rates [8, 9].
Figure 1.
Reproductive Cycle of Human Immunodeficiency Virus and Sites of Action of the Major Classes of Antiretroviral Medications.
Source: (Ref 4) Gandhi M, Gandhi RT. Single-pill combination regimens for treatment of HIV-1 infection. N Engl J Med. 371:248-59, 2014. © 2014 Massachusetts Medical Society. Figure and Legend Reproduced with permission.
Additional medication classes including fusion/entry inhibitors, integrase strand transfer inhibitors (INSTIs), and post-attachment inhibitors continue to strengthen the HIV treatment armamentarium. The development of INSTIs, which are well-tolerated and have a high barrier to resistance, inaugurated another revolution in ART management. INSTI-containing three-drug regimens are now recommended for initial ART by US guidelines[10, 11].
3. Goals of ART
The goals of antiretroviral therapy are to achieve and maintain viral suppression to preserve immunity, reduce transmission, and prevent AIDS related morbidity and mortality. Early treatment of HIV has been associated with a higher likelihood of restoring CD4 counts and long-term preservation of T lymphocytes[12, 13]. The mortality benefits of early ART in HIV are now well established as detailed in sections below.
Treatment of PWH also serves an important public health role by reducing the risk of sexual, perinatal, and parenteral transmission of HIV. The landmark Pediatric AIDS Clinical Trials Group (PACTG) study 076 established that treating mothers with HIV and their newborn infants with ART dramatically reduced perinatal transmission of HIV[14]. Several large trials have concluded that effective ART prevents sexual transmission of HIV. In the HPTN 052 randomized controlled trial among HIV serostatus-discordant, mostly heterosexual couples, the partners with HIV were randomized to early versus delayed start of ART[15]. The trial demonstrated a 93% reduction of HIV transmission among couples in the early ART group. There were 8 transmissions in the early start group, and all occurred when the partner with HIV had a positive viral load (prior to suppression or during virologic failure). No transmissions were detected when viral loads were completely suppressed. The PARTNER study evaluated gay serostatus-discordant male couples over an average of 2 years with greater than 76,000 condomless sexual encounters reported. During this study there were no phylogenetically linked within-couple transmissions[16].
4. When to Start ART
Current guidelines recommend initiation of ART in almost all PWH as soon as possible after diagnosis regardless of CD4 count[10, 11]. Historically, concern for development of drug resistance and the toxicities of early regiments often delayed ART initiation for PWH with a normal CD4 count. Development of safer, more efficacious ART has shifted this risk to benefit ratio in favor of treatment. Initiation of ART reduces morbidity and mortality and prevents progression to AIDS in PWH[17]. Two major trials have since demonstrated benefits of ART therapy irrespective of CD4 count[18, 19]. In the START trial, treatment-naïve PWH initiated on therapy with a CD4 count > 500 cells/microL had a decrease in serious AIDS and serious non-AIDs related events at interim analysis compared to those with a delayed therapy (<350 cells/microL). These dramatic findings resulted in early termination of the study[18]. Similarly, the TEMPRANO study randomized patients to either early verses delayed initiation of ART. Persons in the early start group demonstrated a decreased risk of death and severe HIV-related illnesses[11].
Immediate ART or rapid start--defined as initiation of antiretrovirals on the same day or up to 14 days after diagnosis--has shown success in multiple cohort and randomized controlled studies[20, 21, 22, 23, 24, 25, 26]. Immediate start protocols have been associated with significant improvement in retention of care, time to viral suppression, and viral suppression up to a year after initiation[20, 21, 22, 24, 25, 26, 27]. Among persons with acute HIV, rapid start has additional theoretical benefits of decreasing the symptoms of acute HIV, reducing transmission in setting of high-level viremia, and preventing seeding of viral reservoirs[15, 28, 29, 30, 31, 32, 33]. However, rapid start does require coordination between testing and treatment centers and resources to ensure immediate access to medications and timely follow-up care with an HIV specialist. While many care settings in the U.S. do not have adequate infrastructure to support immediate ART, current guidelines recommend initiating ART as soon as possible after diagnosis as long as the PWH is ready to commit to treatment[11].
Despite the shift toward earlier ART initiation for all PWH, certain opportunistic infections (OIs) must still delay HIV therapy. Persons with most OIs should be started on antiretroviral therapy within 2 weeks of diagnosis and initiation of appropriate treatment for the OI with close monitoring for the immune reconstitution inflammatory syndrome (IRIS) [10, 11, 34]. Exceptions include certain persons with tuberculosis (TB) and those with cryptococcal meningitis due to potential complications from IRIS seen shortly after initiating ART. Individuals with TB and CD4 <50 cells/mm3 should be started on ART within 2 weeks[35] and those with TB and CD4 ≥50 cells/mm3 should be started on ART within 2–8 weeks[36]. ART should not be delayed until TB therapy is completed as this was associated with higher mortality[37]. PWH with tuberculous meningitis randomized to immediate ART did not have improved mortality and were noted to have more adverse events compared to delayed ART. Therefore, caution with early ART is recommended in this population[38]. For persons with HIV and cryptococcal meningitis, the optimal timing for ART initiation is unknown, but early ART (started within 1-2 weeks of diagnosis) has been associated with increased mortality compared to deferred therapy (after 5 weeks)[39]. ART in cryptococcal meningitis should be delayed at least 2 weeks until induction therapy is completed and possibly until completion of entire consolidation phase[10].
5. Choosing an Initial ART Regimen
HIV providers and PWH must consider many factors when selecting an ART regimen. For most PWH who are treatment naïve, guidelines recommend the initiation of an INSTI and 2 NRTIs[10, 11]. The newer, second-generation INSTIs, dolutegravir (DTG) and bictegravir (BIC), are generally preferred due to their high barrier to resistance, favorable side effect profile, and lack of a need for a pharmacologic booster. Recommended NRTIs are tenofovir disoproxil fumarate (TDF) or tenofovir alafenamide (TAF), emtricitabine (FTC), abacavir (ABC), or lamivudine (3TC). Alternative regimens include a 2 NRTIs plus either a boosted PI or an NNRTI. Certain scenarios may warrant use or avoidance of specific antiretrovirals. Important considerations for selecting an ART regimen include characteristics of the HIV infection itself, co-infections, co-morbidities and drug interactions, reproductive plans in women of child-bearing potential, access, and convenience (e.g., food requirements, pill burden).
5.1. HIV Characteristics
Potential antiretroviral resistance and HIV viral load must be considered prior to ART initiation. Baseline genotypic resistance testing to assess for transmitted drug resistance (TDR) is recommended for all PWH prior to starting ART. Rates of baseline resistance among treatment naïve individuals are variable based on region and risk factors for HIV acquisition. ART prescribing patterns can contribute to regional differences in TDR[40]. A recent observational cohort study assessed baseline resistance in over 4,000 treatment naïve individuals spanning 14 years and found that the overall rate of TDR was increasing, but the drug resistance mutations recovered did not affect the efficacy of current first line regimens[41]. Use of TDF/FTC as pre-exposure prophylaxis (PrEP) initially raised concerns that it may generate resistant HIV infections. However, transmitted drug-resistance mutations are rare in PrEP and have been primarily noted in individuals who were started on TDF/FTC when they in fact had acute HIV infection but had negative findings on screening antibody tests[42].
When starting ART prior to the availability of genotype results or HIV viral load, boosted PI- or INSTI-based regimens are favored for their high barrier to resistance and rapid efficacy. For immediate start, favored regimens include DTG+(TDF or TAF)/FTC, BIC/TAF/FTC, or darunavir (DRV) boosted with ritonavir (r) or cobicistat (c) +TAF/FTC or TDF/FTC or TDF/3TC due to their higher barriers against resistance[11]. Prior to use of ABC, PWH must have a negative test for HLA B*5701 which evaluates for predisposition to development of abacavir hypersensitivity syndrome, a potentially life-threatening condition which can occur during the first few weeks of initiation. As a result, INSTI-based single tablet regimen DTG/ABC/3TC is not appropriate for use as a rapid start ART choice.
Pretreatment HIV RNA level and CD4 count also must be factored into ART selection. Certain regimens have shown higher rates of virologic failure—i.e. failure of the ART regimen to suppress the viral load below 200 copies/mL—in the setting of high HIV RNA levels or low CD4 levels pretreatment. As a result, rilpivirine (RPV)-based regimens, DRV/r + raltegravir (RAL), and ABC/3TC with efavirenz (EFV) or ritonavir-boosted atazanavir (ATV) are not recommended for individuals with a pretreatment HIV RNA level >100,000 copies/mL. Similarly, RPV-based regimens and DRV/r + RAL are not recommended for PWH with a pretreatment CD4 count less than 200 copies/mL[10].
5.2. Co-infections
Treatment for co-infections has a significant impact on ART selection. Globally, 5-20% of PWH are also co-infected with hepatitis B virus (HBV)[43]. Persons with HIV-HBV coinfections experience higher morbidity and mortality than those with HIV infection alone[43]. Lamivudine, emtricitabine, and tenofovir (both TDF and TAF) are active against both HBV and HIV. In individuals with HIV-HBV co-infection, 2 drugs in the ART regimen must be HBV-active to treat adequately HBV[10].
Up to 30% of PWH in the US also are co-infected with HCV[44]. Individuals with dual HIV-HCV infection are at increased risk of progressive liver disease, particularly if they have low CD4 T cell lymphocyte counts. While use of direct acting antivirals (DAAs) for the treatment of HCV has been equally efficacious in achieving sustained virologic response in HIV/HCV coinfected populations[45], providers must be cognizant of some drug-drug interactions between DAAs and ART. Use of PIs and pharmacoenhancers are contraindicated with many DAAs, but other ART classes can be co-administered[10, 46].
Treatment of active tuberculosis (TB), latent TB and nontuberculous mycobacterium (NTM) infections in PWH may have a substantial impact on ART selection. Rifamycins are the backbone of TB therapy and potent inducers of the CYP450, P-glycoprotein and uridine diphosphate glucuronosyltransferase (UGT) 1A1 enzymes which cause increased metabolism of antiretrovirals. The current US Department of Health and Human Services (DHHS) Guidelines do not recommend use of rifampin and rifapentine with PIs, both boosted and unboosted, because of the potential for reduced levels of antiretroviral activity[10]. On the other hand, rifabutin, which is a substrate of the CYP450 enzyme system, may be used with PIs but requires dose-reduction (rifabutin 150 mg daily). Among the NNRTIs, only efavirenz (EFV) is recommended for use with rifamycins. With regards to INSTIs, BIC and cobicistat-boosted elvitegravir (EVG) are contraindicated with rifamycins. However, RAL can be administered with rifamycins[47]. Raltegravir should be increased to 800 mg twice daily if used with rifampin but can be given at 400 mg twice daily when used with a rifabutin-containing TB regimen[10]. Dolutegravir can also be used but current data suggest that increasing the dose to 50 mg twice a day is recommended when used with rifampin but no dose increased was needed with rifabutin even at 300 mg daily[1, 10, 48, 49]. Within the NRTIs, TDF, ABC, 3TC, and FTC can be given together with rifampin-containing TB treatment without dose adjustment. TAF should be used with caution since PK and virologic efficacy with ART and TB regimens have not yet been completed[10].
5.3. Co-morbidities
The presence of chronic, non-infectious conditions also impact ART selection. While the safety and tolerability of modern ART has substantially improved compared to historical regimens, significant toxicities persist. In particular, ART effects on renal function, bone density, neuropsychiatric symptoms, and cardiometabolic risk have been seen[10].
HIV is an independent risk factor for renal disease[50]. ART can both decrease the risk of renal disease by treating HIV and contribute to nephrotoxicity[51, 52]. Tenofovir disoproxil fumarate (TDF) has been associated with nephrotoxicity, including proximal renal tubulopathy (Fanconi’s Syndrome)[53]. These adverse effects of TDF are attributed to high circulating plasma levels of tenofovir. The newly formulated tenofovir alafenamide (TAF) is a tenofovir prodrug, which has increased intracellular delivery of the active moiety and lowers plasma circulating levels of tenofovir by 90%[54]. As a result, TAF has an improved safety profile and has demonstrated improved laboratory measures of renal function than TDF in a large non-inferiority study[55]. In PWH who have a pretreatment estimated glomerular filtration rate (eGFR) < 60 mL/min, TDF should be avoided. TAF can be used with an expanded eGFR >30 mL/min[10].
Compared to seronegative individuals, PWH have higher rates of osteopenia, osteoporosis and increased risk of fragility fractures[56]. In addition to its nephrotoxicity, TDF can cause decreased bone mineral density leading to osteopenia and osteoporosis[57]. As a result, TDF-containing regimens should be avoided in PWH starting ART who have known osteoporosis[10]. However, TAF has a favorable bone mineral density safety profile and can be used for these individuals[55].
PWH also suffer from increased rates of psychiatric illness and are at heightened risk for neurocognitive impairment (NCI)[58, 59]. Efavirenz and, to some extent, rilpivirine have been associated with neuropsychiatric symptoms and suicidality[60]. Therefore, EFV- and RPV- containing regimens are often avoided for PWH with known psychiatric illness. Recent data from cohort studies has indicated increased rates of neuropsychiatric adverse events with DTG[61, 62, 63]. Ongoing prospective studies are examining the mechanisms and clinical significance of this effect.
In addition, PWH experience higher rates cardiovascular disease than the general population[64, 65, 66]. This excess risk has been attributed to HIV-associated chronic inflammation and immune activation, ART-associated dyslipidemia and other cardiometabolic comorbidities, healthcare disparities, and increased tobacco use among PWH[64]. Dyslipidemia independently contributes to risk of atherosclerotic cardiovascular disease (ASCVD) in PWH[67]. Boosted-PIs, cobicistat and EFV are known to cause dyslipidemia and may need to be avoided in individuals with known hyperlipidemia. In contrast, INSTIs are considered lipid neutral and TDF has a favorable lipid profile[10]. Furthermore, PWH experience high rates of metabolic comorbidities including diabetes[68], non-alcoholic fatty liver disease (NAFLD)[69], and body composition changes that can confer increased risk of cardiovascular disease[70]. PIs (particularly ritonavir, lopinavir, and indinavir) and, more recently, INSTIs have been associated with hyperglycemia and development of insulin resistance and diabetes[71, 72, 73]. Early PIs and NRTIs were associated with subcutaneous fat and increased visceral adiposity resulting in lipodystrophy and insulin resistance[74]. While modern first-line ART regimens are felt to have more favorable metabolic profiles, there is increasing concern about excess weight gain related to INSTIs and TAF and potential obesity-related complications. Newer ART, particularly second generation INSTIs (BIC and DTG) and TAF, has been associated with more weight gain than other regimens[75, 76, 77, 78]. Investigation into the cardiometabolic risks associated with such weight gain are ongoing.
Some ART regimens have also been independently associated with increased cardiovascular risk. In particular, ABC has been linked to cardiovascular events which increasingly deters its use in high risk individuals. A large observational study comes from the Data Collection on Adverse Events in Anti-HIV Drugs that demonstrated a two-fold increased risk of myocardial infarction (MI) with ABC use[79]. More recently, the NA-ACCORD, a consortium of HIV cohorts in the US and Canada, evaluated risk of MI in PWH on ABC compared to those on alternate regimens. After adjusting for confounding variables, ABC use within the prior 6 months was associated with an increased risk of both type 1 and type 2 MI[80]. However, it remains up to the discretion of the clinician to assess the individual patient risk of MI compared to the potential benefit of ABC-based therapy. Beyond selecting ART with a favorable cardiovascular profile, mitigating cardiovascular risk with statin therapy presents unique challenges in PWH. Cardiovascular risk assessment tools for the general population underestimate risk for PWH[81, 82, 83, 84]. Guidelines for when to start statin therapy in PWH are extrapolated from the recommendations for the general population but may be expanded as new data becomes available from the ongoing Randomized Trial to Prevent Vascular Events in HIV (REPRIEVE) study[64]. When initiating statin therapy, providers must be cognizant that statins and some antiretrovirals are metabolized via the CYP system and assess for drug-drug interactions. PIs and pharmacokinetic boosters are at highest risk of interaction with statins. With these regimens, simvastatin and lovastatin should to be avoided; atorvastatin, pravastatin and rosuvastatin may need dose reductions; and pitavastatin may be used without restriction[85].
5.4. Special considerations for women with HIV
Prior to starting ART, healthcare providers and women of childbearing age with HIV should discuss her reproductive goals. For women who are or desire pregnancy, special consideration must be taken to select ART that is safe and effective for both the mother and her child. Poor HIV control is associated with poor outcomes for the mother and increased risk of perinatal transmission of HIV[86]. Unfortunately, the pharmacokinetic and safety of ART during pregnancy remain understudied.. Physiologic changes during pregnancy can affect ART absorption, distribution, metabolism, and excretion[88]. Pregnant women experience decreased gastrointestinal motility due to progesterone and may alter their food intake due to nausea or vomiting resulting in decreased absorption of ART and other medications. During pregnancy plasma volume expands thereby diluting plasma proteins and leading to increased free drug levels and clearance[89]. Increase cardiac output and glomerular filtration rate lead to increased renal elimination of medication. In addition, pregnancy can differentially increase or decrease drug metabolism via CYP enzymes[90].
For ART-naïve women, only RAL +TDF/FTC is considered preferred in both the DHHS Adult and Perinatal guidelines[10, 87]. Women who are virally suppressed on ART at the time of pregnancy planning or conception should remain on their ART regimen with few exceptions. TAF is not recommended in this population due to insufficient data regarding its use during pregnancy[87]. Recently, concern has been raised about the safety of DTG at the time of conception and during the first trimester in pregnant women with HIV. A slightly increased rate of neural tube defects has been identified in infants born to women who started DTG-containing regimens before the time of conception[91]. A multicenter surveillance program in Botswana initially noted a 0.94% rate of neural tube defects in pregnant women with HIV taking DTG since the time of conception. In a follow-up analysis from July 2018, 0.67% (4/596) had NTD compared to 0.12% of infants born to mother on non-DTG based regimens[91]. As of March 2019 in Botswana with a larger sample size, among 1683 deliveries among women on DTG at conception, 5 NTD were identified (0.30%) compared with 15 NTD among 14,792 deliveries (0.10%) in mothers taking any non-dolutegravir ART at conception[92]. In the same surveillance data, there was only 1 NTD among 3849 (0.03%) in which the mother started DTG during pregnancy typically after the first trimester, and 70 among 89,372 (0.08%) among HIV-uninfected mothers. Given this slightly increased risk of NTD among women on DTG at conception, it is important for providers to discuss the potential risks and benefits of available ART options with women considering pregnancy.
Cobicistat is not recommended during pregnancy. Cobicistat’s reliance on metabolic inhibition raised concern that metabolic changes in pregnancy might affect its efficacy. Initial case reports of pregnant women treated with elvitegravir/cobicistat containing ART revealed 44% lower cobicistat exposure during pregnancy than in the postpartum period[93]. Subsequently, a multicenter phase IV trial evaluating cobicistat and elvitegravir pharmacokinetics during the second trimester, third trimester, and postpartum period found that elvitegravir exposure was lower during pregnancy than during the postpartum period[94]. In this study, the lowest values were noted during the third trimester. In fact, only 47% of participants in the 2nd trimester and 38% in the 3rd trimester met the target area under the concentration curve (AUC) goal. Similarly, cobicistat exposure was found to be lower during pregnancy than during the postpartum period. Such unfavorable pharmacokinetics preclude use of cobicistat-containing regimens during pregnancy. Presently ritonavir is the preferred boosting agent during pregnancy[87].
Women of childbearing age who do not desire pregnancy should be offered contraceptive counseling as part of routine HIV care. Combined oral contraceptives (COCs) and implanted hormonal contraceptives can be affected by ART leading to ineffective contraception. However, limited data exist to fully characterize the interactions and clinical implications. Most notable is the interaction of EFV with implanted levonorgestrel in which women on EFV-based regimens with implanted levonorgestrel were shown to have decreased levels of levonorgestrel, a progesterone, and increased rates of contraception failures[95]. Even with addition of a second levonorgestrel implant, this interaction persisted[96]. Boosted PI based regimens can interfere with ethinyl estradiol and lead to decreased levels and increased risk of side effects (e.g., vaginal bleeding), but ultimate impact on pregnancy prevention may be limited. Depot delivery of hormonal contraception utilizing medroxyprogesterone acetate (MPA) leads to higher levels of MPA and hence, has not been shown to be affected by ART. The DHHS perinatal HIV guidelines include Table 3 “Drug Interactions Between Antiretroviral Agents and Hormonal Contraceptives” that summarizes the available data and expert recommendations for prescribing hormonal contraceptives with ART[10]. This potential for decreased contraceptive efficacy should prompt clinicians to encourage additional methods of pregnancy prevention, but not prevent prescribing of hormonal contraception.
Among transgender women with HIV, gender-affirming hormonal therapy (HT) may also be impacted by ARVs, but most data has been extrapolated from COCs. Gender affirming hormone therapy usually consists of an androgen blocking agent and an estrogen agent. Spironolactone is the most frequently used anti-androgen medication in the US and no drug interactions with ARVs have been identified[97]. PIs and NNRTIs may decrease levels of circulating estrogen, requiring closer monitoring to achieve a clinically efficacious dose of estrogen. The concern for drug-drug interactions and decreased efficacy of HT may lead to reduced adherence to ARVs in favor of HT if not monitored and addressed by the provider[98].
6. Challenges with ART maintenance
6.1. Maintaining Engagement in Care
Successful ART requires that PWH remain engaged in care and adherent to ART for the remainder of their life. Among adults and adolescents living with HIV in the United Sates in 2015, only 49% were engaged in continuous HIV care and 51% had achieved virologic suppression[99]. Numerous barriers across the HIV care continuum--from intrapersonal patient obstacles to issues of national health policy--pose challenges for engagement in and adherence to continued HIV therapy[100, 101].
Psychosocial factors are a significant contributor to ART nonadherence. Effective management of HIV must address social barriers such as stigma[102, 103], loneliness[104, 105], unstable housing[106], and intimate partner violence[107] that can contribute to decreased medication adherence. Intensive social work and case management support can help to identify and direct appropriate interventions to facilitate continuous HIV care, such as housing, transportation, insurance coverage, and mental health treatment[108, 109]. For PWH who face significant financial and structural barriers, more intensive outreach and peer navigator interventions may help to determine needed supportive services[110, 111].
HIV care providers must also actively address and manage comorbidities that affect medication adherence. Rates of mental health disorders, neurocognitive impairment, and substance abuse are increased among PWH compared to the general population[59, 112, 113]. Ongoing depressive symptoms adversely affect engagement in HIV care[114, 115]. Integrated treatment of opioid dependence and HIV has been shown to increase number of visits, ART initiation, and rates of virologic suppression[116, 117]. Providers offering pharmacologic therapies for opioid dependence and mood disorders in PWH must be cognizant of potential drug interactions with ART. For example, all ritonavir-boosted PIs as well as efavirenz decrease methadone’s AUC, so providers must adjust methadone dosing and/or monitor patients for signs of opioid withdrawal with co-administration of these mediations [10].
Among PWH already established on virally suppressive ART, some regimen switches may be considered to improve adherence, decrease medication side effects and long-term toxicities, or avoid drug-drug interactions. To maintain viral suppression, it is imperative to consider any prior resistance mutations as well as the new regimen’s barrier to resistance when making changes in ART. Within class switches to an agent with a similar barrier to resistance--such as changing TDF to TAF to reduce bone and renal toxicity—is a reasonable strategy. Similarly, between class switches to an agent with a similar barrier to resistance-such as changing a boosted PI to DTG—are also effective. However, switching to an agent with a lower barrier to resistance either in the same (e.g., DTG to RAL) or a different (e.g., DTG to RPV) class should only be considered if there has not been any prior resistance noted[10].
Maximizing ART convenience can promote adherence and is a common reason for switching regimens. Food requirements, excess pill burden, and higher dosing frequencies can contribute to treatment-fatigue. Single tablet regimens (STRs) can increase adherence and virologic suppression as well as decrease rates of discontinuation and laboratory abnormalities[118]. There are a growing number of STRs available. However, for certain populations, like individuals on hemodialysis, fixed dose combinations may not be appropriate. One case series has demonstrated the safety and efficacy of a single tablet regimen with DTG/ABC/3TC in patients with HIV and ESRD on hemodialysis but has not been studied on a larger scale[119].
Virologic failure (or the inability to maintain viral suppression below 200 copies/mL) also warrants re-evaluation of the individual’s ART regimen. Prior to switching therapies, providers and PWH should address and intervene on any factors that may limit adherence to and efficacy of the current regimen, such as improper dosing, cost, or drug-drug interactions. In the setting of virologic failure, resistance testing should be performed—including assessment for integrase resistance if prior ART included an INSTI. When selecting a new ART regimen, current and historical resistance must be considered. For PWH with virologic failure, switch regimens must contain at least 2 but ideally 3 active drugs[10]. In some cases, there may still be active drug options in the same ART classes as the failing regimen that has a higher barrier to resistance. For example, in the setting of a K103N mutation conferring resistance to EFV, other NNRTIs such as doravirine, rilpivirine, and etravirine may still retain activity. In the setting of multidrug resistant HIV, providers may consider addition of an active drug with a different mechanism of action, such as a CCR5 antagonist, fusion inhibitor, or the recently approved post-attachment inhibitor ibalizumab[120]. When constructing a salvage regimen for PWH with extensive drug resistance, consultation with an HIV expert is recommended.
6.2. Challenges in a young adult population
For adolescents living with HIV, the transition to adult care represents a particularly vulnerable period for retention in care and virologic suppression. In 2017, youth aged 13 to 24 years old accounted for 21% of new HIV diagnoses in the United States. Moreover, there are a growing number of adolescent and young adult long-term survivors of perinatal HIV infection[121]. ART adherence can be a particular challenge for adolescents with either recently or perinatally acquired HIV infection[122, 123]. Furthermore, teens and young adults living with HIV have poorer virologic outcomes and follow-up rates[124]. As such, HIV clinics focused on adult care must be attuned to the specific needs of adolescents transitioning into their care.
While the general factors that affect retention in care and ART adherence in all PWH apply to adolescents and young adults living with HIV, such challenges can be magnified in this population. Youth transitioning to adult HIV care may be more likely to have gaps in health insurance, concerns about disclosure of their HIV status if they remain on their parents’ insurance, and less experience navigating complex health systems[125]. Many young PWH also have decreased social support[126] and experience high rates of mood disorders[127] that can impact adherence and follow-up. Therefore, comprehensive clinic models that are easily navigable, offer intensive mental health and social support, and include HIV providers with significant experience in care transitions can improve outcomes for young PWH.
Moreover, adolescents tend to be less risk averse and are still developing concrete thinking skills which can diminish adherence to ART or any chronic medication. To prevent development of resistance, ART may sometimes be deferred until adherence is more likely and the most potent and easily dosed regimen should be selected[10]. However, long-term survivors of perinatal infection are often highly ART experienced, with higher likelihood of harboring HIV resistance mutations, and tend to be on more complex regimens[128]. User-friendly, inconspicuous support systems such as apps, reminder calls, and even pill boxes can help to bolster ART adherence among adolescent and young adults living with HIV[129, 130].
6.3. Challenges in an aging population
Management of HIV in older adults poses a special set of challenges. With successful ART, PWH are living longer. In addition, HIV risk persists in older ages. In 2015, adults over the age of 50 accounted for 47% of known as well as 17% of new HIV diagnoses in the United States. Older patients tend to present with lower CD4 counts at time of diagnosis and have steeper declines in CD4 count over time[131, 132, 133]. With initiation of ART, elderly PWH tend to have improved adherence compared with younger adults but have decrease immune recovery compared to younger patients [134, 135, 136]. Due to their depressed immune recovery and increased risk of other adverse outcomes, older PWH benefit the most from early initiation of ART[137]. Despite increased immunosenescence, older patients have a virologic response similar to that of younger individuals on ART and, in some studies, even higher rates of viral suppression presumably from improved adherence rates[136, 138].
While initial ART recommendations remain the same for older patients, providers must be cognizant of this population’s specific clinical characteristics. Older individuals are more likely to have comorbidities--such as decreased renal function, cardiovascular disease, or reduced bone mineral density—that may impact choice of ART. For example, dosage adjustments of NRTIs may be required for patients with kidney disease. Similarly, associations between ABC and increased risk of cardiovascular events may lend caution to its use in older patients, who may already have heightened cardiovascular disease risks[80]. Studies of the pharmacokinetics, efficacy, and adverse effects of ART in the elderly have been limited. Following initiation of ART, older patients should be monitored for development of adverse drug effects.
Older adults living with chronic HIV infection experience both earlier onset and higher rates of chronic non-infectious aging-related comorbidities than the general population[139, 140, 141]. The mechanisms underlying these differences are not fully understood but have been attributed to chronic inflammation and immune activation, ART toxicities, and increased prevalence of high-risk behaviors (e.g., smoking) among PWH. Development of additional comorbidities may prompt changes to ART to minimize toxicity. If a patient on a TDF- based regimen develops osteoporosis, for example, that individual and their provider may consider switching to TAF or a NRTI-sparing regimen. In addition, older PWH with multiple comorbidities are at increased risk for polypharmacy[142, 143]. Such individuals must be carefully monitored for potential drug-drug interactions.
Among other comorbidities, persons aging with HIV are at increased risk for geriatric syndromes such as frailty and cognitive impairment[144, 145, 146]. As a result, this population is particularly vulnerable to poor health outcomes such as disability, reduced quality of life, and social isolation[147, 148, 149, 150]. These age-related comorbidities can also contribute to non-adherence. Similarly, depression, social isolation and neurocognitive impairment can all decrease adherence and engagement in HIV care[151, 152]. Increasing pill burden and, as a result, medication costs, may decrease adherence and contribute to virologic failure in older PWH. Attention to these complex, age-related issues is essential not just to maintain virologic control but also to maximize quality of life for PWH across the age continuum.
7. Future of ART
Current ART regimens are now safer, more convenient, and more efficacious than ever. First line ART with INSTI-containing three-drug regimens are well-tolerated, highly effective with a high resistance barrier, and often available as single tablet regimens. Novel ART agents and strategies offer the promise of further reductions in medication toxicity, more convenient administration, and improved efficacy against resistant virus.
7.1. Dual ART
There is growing evidence that dual therapy is efficacious and may reduce cost and drug toxicity. Some guidelines include DTG + 3TC, DRV/r + RAL twice daily and DRV/r+ 3TC as potential initial ART in special situations, such as when tenofovir or abacavir cannot be used[10]. DRV/r +RAL is further restricted to patients with VL < 100,000 copies/mL and CD4 > 200 cells/μL.
There is greater evidence supporting a switch to dual therapy following virologic suppression with three-drug ART. SWORD-1 and 2 demonstrated that a once-daily combination of DTG plus RPV maintained virologic suppression in greater than 95% of participants switched off of their previous combination ART[153]. DTG/RPV is now available as a co-formulated tablet. Dual therapy with a PI/r and emtricitabine also has growing supportive evidence[154, 155, 156, 157]. However, PI-based dual therapy has had limited uptake due to its propensity for drug interactions and adverse effects, such as metabolic derangements.
Dual therapy with a boosted-PI plus an INSTI or DTG plus 3TC also has some supporting evidence as switch therapy but are still not recommended in guidelines. Small studies have shown that viral suppression was maintained in 97% of patients switched to DRV/R plus DTG[158, 159]. The combination of DTG plus 3TC maintained virologic suppression in the LAMIDOL and ASPIRE studies[160]. Recently, GEMINI 1 and GEMINI 2 demonstrated non-inferiority of DTG + 3TC to DTG + TDF/FTC as initial ART[161]. In addition, DTG/3TC is also now available as a co-formulated tablet.
Though dual therapy shows increasing promise and offers some potential advantages, significant limitations constrain its use. Dual regimens all lack the two NRTIs recommended for treatment of HIV and HBV co-infection and are therefore contraindicated in this population. Furthermore, dual regimens have not been investigated in the setting of tuberculosis co-infection, pregnancy, and renal disease. The lack of data in these key populations limits dual regimens’ widespread uptake.
7.2. Long-acting ART
While dual regimens may decrease toxicity and cost, other novel therapies offer innovative delivery techniques to improve adherence. Long-acting antiretroviral therapies including parenteral agents, implants, and patches may offer promising treatment alternatives for PWH who have difficulty adhering to a daily pill-based regimen. Long-acting injectable nanoformulated antiretrovirals are farthest along in clinical trials. The Phase II LATTE-2 trial showed that after a 20 week oral lead in, an all injectable regimen of cabotegravir, a novel INSTI, and rilpivirine monthly or every 2 months was as effective as three-drug oral ART at maintaining viral suppression[162]. Phase III clinical trials are ongoing to evaluate this combination as initial ART (FLAIR), switch therapy for PWH with viral suppression on an oral ART regimen (ATLAS), and in PWH with a history of poor adherence to ART and poorly controlled HIV (LATITUDE/ACTG A5359). Development of additional new long-acting injectable antiretrovirals is also underway. The novel NNRTI, elsulfavirine (VM1500A), is distinguished by its tolerability and long half-life. Preclinical pharmacokinetic studies have shown the potential to develop VM1500A as long-acting injectable agent and a Phase 1b trial investigating its weekly dosing is underway (ClinicalTrials.gov NCT03730311).Bichko V, Rogovoy B, Koryakova A, et al. Pre-clinical pharmacokinetics of elsufavirine/VM1500A long acting injectable formulations. International Antiviral Society-USA. 2017. Poster WEPEA0190.. A new long-acting fusion inhibitor, albuvirtide, has been approved in China and is well-tolerated[163]. There are ongoing efforts to develop a subcutaneous formulation of albuviritide that would allow self-administration every 2–4 weeks.
Broadly neutralizing antibodies (bNAbs) offer another promising new class of long-acting parental therapy for both HIV treatment and prevention[164, 165]. HIV bNAbs were originally isolated from individuals with high levels of anti-HIV neutralizing activity and target antigens on the HIV external membrane glycoprotein 120 (gp120). Early clinical studies have shown that bNAbs can reduce viral load, stimulate immune response to HIV and HIV-infected cells, and are well-tolerated[166, 167]. Clinical trials are now underway to test combinations of bNAbs as well as bNAbs combined with a long-acting antiretroviral such as albuvirtide (3BNC117 ) which is being investigated as long-acting maintenance therapy in ClinicalTrials.gov NCT03719664.
Antiretroviral implants and patches could offer alternative long-acting treatment approaches. In contrast to parenteral therapies, implants and patches would not require more clinic visits for infusions and therefore may be more applicable to resource poor settings. A novel nucleoside reverse transcriptase translocation inhibitor (NRTTI), MK 8591 (islatravir), has a prolonged half-life of 150 to 160 hours and potent activity against HIV-1, -2 , as well as multidrug resistant strains[168]. As a result of its these properties, MK 8591 has been formulated in a drug-eluting implant[169] and is an attractive candidate for long-acting pre-exposure prophylaxis (PrEP) and as well as HIV treatment. Other novel drug delivery technology, including microneedle patches for transdermal delivery[170] and biodegradable implants[171], may offer promising future long-acting treatment strategies as well.
7.3. The ART Pipeline
Entirely new classes of antiretrovirals are also under development and may offer more therapeutic options especially for highly treatment experienced PWH. The first CD4 attachment inhibitor, fostemsavir, acts by binding to HIV envelope glycoprotein 120. It is currently under investigation in the Phase III BRIGHTE study for heavily treatment experienced PWH with multidrug resistance (ClinicalTrials.gov NCT02362503). Similarly, the novel class of capsid inhibitors have been shown to be potent against HIV-1 at both early and late stages of viral replication in vitro[174]. These agents, which include GS-CA1 and its modified version GS-6207, interfere with capsid disassembly and render virions non-infectious. Moreover, GS-6207 has unique pharmacokinetic properties that may allow for long-acting subcutaneous administration and is currently in Phase I trials (ClinicalTrials.gov NCT03739866). Finally, ABX464 is an orally available small molecule in the novel class of Rev inhibitors and blocks viral replication by preventing the export of viral RNA from the nucleus to the cytoplasm in HIV-infected cells. By interfering with the HIV protein Rev which is essential for this transport out of the nucleus, this drug prevents production of new virus and also triggers immune activation to eliminate HIV-infected cells. As such, this agent is thought to reduce the HIV reservoir and has the potent to be part of functional cure strategies[175].
8. Expert Opinion
Effective ART has transformed HIV from a uniformly fatal illness to a chronic, manageable condition. On modern ART, a young adult in North America diagnosed with HIV today has a life expectancy nearing that of the general population[1]. Such effective treatment also prevents the spread of HIV and is crucial for ending the epidemic[14, 15, 176]. However, attaining these benefits of ART currently requires that PWH be adherent to lifelong, daily ART. While modern ART is increasingly safe, effective, and convenient, challenges persist in initiating and maintaining ART throughout adulthood.
When selecting initial ART, an individual’s coexisting medical, social and personal factors must be considered. While several first-line single table regimens are now available, these once daily regimens may be contraindicated in persons with certain comorbidities. In particular, fixed dose combination pills may not be appropriate for those with renal disease or during pregnancy. As a result, some persons with HIV must take less convenient multi-tablet regimens, which are associated with increased risk of non-adherence.
Maintaining ART across changing life conditions often proves more challenging than initiating therapy itself. For women living with HIV, the risks and benefits of various antiretrovirals change across their reproductive years. ART can have significant drug interactions with hormonal contraception. Many first-line ART regimens have safety and efficacy concerns during pregnancy. Therefore, it is imperative that providers continually discuss reproductive goals with all PWH and especially those who are able to become pregnant to ensure that their ART is concordant with their parenting desires[177]. In addition, as PWH live longer on effective ART, providers must be cognizant of therapeutic issues in the growing population of persons aging with HIV. Polypharmacy, medical comorbidities, cognitive difficulties and social isolation can all increase with age and lead to greater drug toxicity and difficulties with adherence. Moreover, for PWH of all ages and genders, providers must watch for and actively manage factors that may lead to ART non-adherence, such as pill burden, ART side effects, cost of medications, homelessness, intimate partner violence, stigma, opiate addiction, and mental health concerns. Furthermore, providers must watch for virologic failure and emergence of antiretroviral resistance that may require ART modification.
Emerging therapies and novel approaches to ART delivery will address many of these challenges but may also raise new complications. Long-acting ART, administered as injections, patches or implants, has the potential to increase convenience and adherence. However, these therapies may be more expensive and place increased burden on clinics and PWH. In the event of a missed injection, for example, such long-acting administrations can have months-long low-level drug concentrations that could lead to drug resistance. Dual therapy has the potential to decrease ART toxicity and cost. But currently two-drug ART regimens can only safely be used in a subset of PWH. Other therapies under development may offer new antiretrovirals in existing and novel drug classes that may further combat continued resistance, increase efficacy, and, hopefully, bring the field closer to functional cure.
Continued HIV research must address ART in populations historically underrepresented in clinical trials for whom therapeutic disparities persist. Further studies are needed to better understand potential complications in and improve ART for female, aging, medically complex, and transgender PWH. Over the last 4 decades, advances in ART have revolutionized HIV care. Until HIV cure arrives, future research must continue to improve the efficacy, safety, convenience, and equitability of ART for all persons living with HIV.
Article Highlights:
Antiretroviral therapy (ART) has transformed HIV into a chronic, manageable illness. Virally suppressive ART extends the life-expectancy of PWH to near that of the general population but requires life-long adherence.
Selection of an initial ART regimen requires multiple considerations including baseline resistance, co-infections, and co-morbidities.
ART remains understudied in women and during pregnancy. Women living with HIV and their providers must discuss her reproductive goals to ensure safe and effective ART.
Multidisciplinary approaches are essential for maintaining engagement in care and viral suppression across the adult lifespan of PWH.
Emerging ART—including dual therapy and long-acting agents—offer the promise of safer, more convenient, and more efficacious treatments.
Acknowledgments
Funding
This work was funded by the National Institute of Diabetes and Digestive and Kidney Diseases T32 DK007169 (to MCM). The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health.
Footnotes
Declaration of Interest
SE Cohn has served as a scientific advisor for Merck & Co. The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.
Reviewer Disclosures
Peer reviewers on this manuscript have no relevant financial or other relationships to disclose.
References and Recommended reading
Papers of particular interest have been highlighted as:
* of special interest
** of outstanding interest
- 1.Antiretroviral Therapy Cohort C. Survival of HIV-positive patients starting antiretroviral therapy between 1996 and 2013: a collaborative analysis of cohort studies. Lancet HIV. 2017. August;4(8):e349–e356. doi: 10.1016/S2352-3018(17)30066-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Centers for Disease C. Revision of the case definition of acquired immunodeficiency syndrome for national reporting--United States. MMWR Morb Mortal Wkly Rep. 1985. June 28;34(25):373–5. [PubMed] [Google Scholar]
- 3.Centers for Disease C. Pneumocystis pneumonia--Los Angeles. MMWR Morb Mortal Wkly Rep. 1981. June 5;30(21):250–2. [PubMed] [Google Scholar]
- 4.Gandhi M, Gandhi RT. Single-pill combination regimens for treatment of HIV-1 infection. N Engl J Med. 2014. July 17;371(3):248–59. doi: 10.1056/NEJMct1215532. [DOI] [PubMed] [Google Scholar]
- 5.Broder S The development of antiretroviral therapy and its impact on the HIV-1/AIDS pandemic. Antiviral Res. 2010. January;85(1):1–18. doi: 10.1016/j.antiviral.2009.10.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Fischl MA, Richman DD, Grieco MH, et al. The efficacy of azidothymidine (AZT) in the treatment of patients with AIDS and AIDS-related complex. A double-blind, placebo-controlled trial. N Engl J Med. 1987. July 23;317(4):185–91. doi: 10.1056/NEJM198707233170401. [DOI] [PubMed] [Google Scholar]
- 7.Vella S, Schwartlander B, Sow SP, et al. The history of antiretroviral therapy and of its implementation in resource-limited areas of the world. AIDS. 2012. June 19;26(10):1231–41. doi: 10.1097/QAD.0b013e32835521a3. [DOI] [PubMed] [Google Scholar]
- 8.Hammer SM, Squires KE, Hughes MD, et al. A controlled trial of two nucleoside analogues plus indinavir in persons with human immunodeficiency virus infection and CD4 cell counts of 200 per cubic millimeter or less. AIDS Clinical Trials Group 320 Study Team. N Engl J Med. 1997. September 11;337(11):725–33. doi: 10.1056/NEJM199709113371101. [DOI] [PubMed] [Google Scholar]
- 9.Gulick RM, Mellors JW, Havlir D, et al. 3-year suppression of HIV viremia with indinavir, zidovudine, and lamivudine. Ann Intern Med. 2000. July 4;133(1):35–9. doi: 10.7326/0003-4819-133-1-200007040-00007. [DOI] [PubMed] [Google Scholar]
- 10.Guidelines for the use of antiretroviral agents in adults and adolescents living with HIV. Available at: https://aidsinfo.nih.gov/guidelines Accessed 7/1/19.; ** The U.S. Department of Health and Human Services clinical guidelines for antiretroviral therapy for adults living with HIV.
- 11.Saag MS, Benson CA, Gandhi RT, et al. Antiretroviral Drugs for Treatment and Prevention of HIV Infection in Adults: 2018 Recommendations of the International Antiviral Society-USA Panel. JAMA. 2018. July 24;320(4):379–396. doi: 10.1001/jama.2018.8431. [DOI] [PMC free article] [PubMed] [Google Scholar]; ** Guidelines from the International Antiviral Society-USA for antitretroviral therpay for adults living with HIV.
- 12.Le T, Wright EJ, Smith DM, et al. Enhanced CD4+ T-cell recovery with earlier HIV-1 antiretroviral therapy. N Engl J Med. 2013. January 17;368(3):218–30. doi: 10.1056/NEJMoa1110187. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Oxenius A, Price DA, Easterbrook PJ, et al. Early highly active antiretroviral therapy for acute HIV-1 infection preserves immune function of CD8+ and CD4+ T lymphocytes. Proc Natl Acad Sci U S A. 2000. March 28;97(7):3382–7. doi: 10.1073/pnas.97.7.3382. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Connor EM, Sperling RS, Gelber R, et al. Reduction of maternal-infant transmission of human immunodeficiency virus type 1 with zidovudine treatment. Pediatric AIDS Clinical Trials Group Protocol 076 Study Group. N Engl J Med. 1994. November 3;331(18):1173–80. doi: 10.1056/NEJM199411033311801. [DOI] [PubMed] [Google Scholar]
- 15.Cohen MS, Chen YQ, McCauley M, et al. Prevention of HIV-1 infection with early antiretroviral therapy. N Engl J Med. 2011. August 11;365(6):493–505. doi: 10.1056/NEJMoa1105243. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Rodger AJ, Cambiano V, Bruun T, et al. Risk of HIV transmission through condomless sex in serodifferent gay couples with the HIV-positive partner taking suppressive antiretroviral therapy (PARTNER): final results of a multicentre, prospective, observational study. Lancet. 2019. June 15;393(10189):2428–2438. doi: 10.1016/S0140-6736(19)30418-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Palella FJ Jr., Delaney KM, Moorman AC, et al. Declining morbidity and mortality among patients with advanced human immunodeficiency virus infection. HIV Outpatient Study Investigators. N Engl J Med. 1998. March 26;338(13):853–60. doi: 10.1056/NEJM199803263381301. [DOI] [PubMed] [Google Scholar]
- 18.Group ISS, Lundgren JD, Babiker AG, et al. Initiation of Antiretroviral Therapy in Early Asymptomatic HIV Infection. N Engl J Med. 2015. August 27;373(9):795–807. doi: 10.1056/NEJMoa1506816. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Group TAS, Danel C, Moh R, et al. A Trial of Early Antiretrovirals and Isoniazid Preventive Therapy in Africa. N Engl J Med. 2015. August 27;373(9):808–22. doi: 10.1056/NEJMoa1507198. [DOI] [PubMed] [Google Scholar]
- 20.Rosen S, Maskew M, Fox MP, et al. Initiating Antiretroviral Therapy for HIV at a Patient’s First Clinic Visit: The RapIT Randomized Controlled Trial. PLoS Med. 2016. May;13(5):e1002015. doi: 10.1371/journal.pmed.1002015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Koenig SP, Dorvil N, Devieux JG, et al. Same-day HIV testing with initiation of antiretroviral therapy versus standard care for persons living with HIV: A randomized unblinded trial. PLoS Med. 2017. July;14(7):e1002357. doi: 10.1371/journal.pmed.1002357. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Labhardt ND, Ringera I, Lejone TI, et al. Effect of Offering Same-Day ART vs Usual Health Facility Referral During Home-Based HIV Testing on Linkage to Care and Viral Suppression Among Adults With HIV in Lesotho: The CASCADE Randomized Clinical Trial. JAMA. 2018. March 20;319(11):1103–1112. doi: 10.1001/jama.2018.1818. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Amanyire G, Semitala FC, Namusobya J, et al. Effects of a multicomponent intervention to streamline initiation of antiretroviral therapy in Africa: a stepped-wedge cluster-randomised trial. Lancet HIV. 2016. November;3(11):e539–e548. doi: 10.1016/S2352-3018(16)30090-X. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Pilcher CD, Ospina-Norvell C, Dasgupta A, et al. The Effect of Same-Day Observed Initiation of Antiretroviral Therapy on HIV Viral Load and Treatment Outcomes in a US Public Health Setting. J Acquir Immune Defic Syndr. 2017. January 1;74(1):44–51. doi: 10.1097/QAI.0000000000001134. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Colasanti J, Sumitani J, Mehta CC, et al. Implementation of a Rapid Entry Program Decreases Time to Viral Suppression Among Vulnerable Persons Living With HIV in the Southern United States. Open Forum Infect Dis. 2018. June;5(6):ofy104. doi: 10.1093/ofid/ofy104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Coffey S, Bacchetti P, Sachdev D, et al. RAPID ART: High virologic suppression rates with immediate ART initiation in a vulnerable urban clinic population. AIDS. 2018. December 21. doi: 10.1097/QAD.0000000000002124. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Ford N, Migone C, Calmy A, et al. Benefits and risks of rapid initiation of antiretroviral therapy. AIDS. 2018. January 2;32(1):17–23. doi: 10.1097/QAD.0000000000001671. [DOI] [PMC free article] [PubMed] [Google Scholar]; * This meta-analysis assessed the impact of rapid antiretroviral therapy (ART) on patient outcomes. It found that accelerated iniation of ART was associated with improved clinical outcomes.
- 28.Strain MC, Little SJ, Daar ES, et al. Effect of treatment, during primary infection, on establishment and clearance of cellular reservoirs of HIV-1. J Infect Dis. 2005. May 1;191(9):1410–8. doi: 10.1086/428777. [DOI] [PubMed] [Google Scholar]
- 29.Chun TW, Justement JS, Moir S, et al. Decay of the HIV reservoir in patients receiving antiretroviral therapy for extended periods: implications for eradication of virus. J Infect Dis. 2007. June 15;195(12):1762–4. doi: 10.1086/518250. [DOI] [PubMed] [Google Scholar]
- 30.Jain V, Hartogensis W, Bacchetti P, et al. Antiretroviral therapy initiated within 6 months of HIV infection is associated with lower T-cell activation and smaller HIV reservoir size. J Infect Dis. 2013. October 15;208(8):1202–11. doi: 10.1093/infdis/jit311. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Hey-Cunningham WJ, Murray JM, Natarajan V, et al. Early antiretroviral therapy with raltegravir generates sustained reductions in HIV reservoirs but not lower T-cell activation levels. AIDS. 2015. May 15;29(8):911–9. doi: 10.1097/QAD.0000000000000625. [DOI] [PubMed] [Google Scholar]
- 32.Martinez-Bonet M, Puertas MC, Fortuny C, et al. Establishment and Replenishment of the Viral Reservoir in Perinatally HIV-1-infected Children Initiating Very Early Antiretroviral Therapy. Clin Infect Dis. 2015. October 1;61(7):1169–78. doi: 10.1093/cid/civ456. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Novelli S, Lecuroux C, Avettand-Fenoel V, et al. Long-term Therapeutic Impact of the Timing of Antiretroviral Therapy in Patients Diagnosed With Primary Human Immunodeficiency Virus Type 1 Infection. Clin Infect Dis. 2018. May 2;66(10):1519–1527. doi: 10.1093/cid/cix1068. [DOI] [PubMed] [Google Scholar]
- 34.Aberg JA, Gallant JE, Ghanem KG, et al. Primary care guidelines for the management of persons infected with HIV: 2013 update by the HIV Medicine Association of the Infectious Diseases Society of America. Clin Infect Dis. 2014. January;58(1):1–10. doi: 10.1093/cid/cit757. [DOI] [PubMed] [Google Scholar]
- 35.Abdool Karim SS, Naidoo K, Grobler A, et al. Integration of antiretroviral therapy with tuberculosis treatment. N Engl J Med. 2011. October 20;365(16):1492–501. doi: 10.1056/NEJMoa1014181. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Blanc FX, Sok T, Laureillard D, et al. Earlier versus later start of antiretroviral therapy in HIV-infected adults with tuberculosis. N Engl J Med. 2011. October 20;365(16):1471–81. doi: 10.1056/NEJMoa1013911. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Abdool Karim SS, Naidoo K, Grobler A, et al. Timing of initiation of antiretroviral drugs during tuberculosis therapy. N Engl J Med. 2010. February 25;362(8):697–706. doi: 10.1056/NEJMoa0905848. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Torok ME, Yen NT, Chau TT, et al. Timing of initiation of antiretroviral therapy in human immunodeficiency virus (HIV)--associated tuberculous meningitis. Clin Infect Dis. 2011. June;52(11):1374–83. doi: 10.1093/cid/cir230. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Boulware DR, Meya DB, Muzoora C, et al. Timing of antiretroviral therapy after diagnosis of cryptococcal meningitis. N Engl J Med. 2014. June 26;370(26):2487–98. doi: 10.1056/NEJMoa1312884. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Panichsillapakit T, Smith DM, Wertheim JO, et al. Prevalence of Transmitted HIV Drug Resistance Among Recently Infected Persons in San Diego, CA 1996-2013. J Acquir Immune Defic Syndr. 2016. February 1;71(2):228–36. doi: 10.1097/QAI.0000000000000831. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Rhee SY, Clutter D, Fessel WJ, et al. Trends in the Molecular Epidemiology and Genetic Mechanisms of Transmitted Human Immunodeficiency Virus Type 1 Drug Resistance in a Large US Clinic Population. Clin Infect Dis. 2019. January 7;68(2):213–221. doi: 10.1093/cid/ciy453. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Riddell Jt, Amico KR, Mayer KH. HIV Preexposure Prophylaxis: A Review. JAMA. 2018. March 27;319(12):1261–1268. doi: 10.1001/jama.2018.1917. [DOI] [PubMed] [Google Scholar]
- 43.Singh KP, Crane M, Audsley J, et al. HIV-hepatitis B virus coinfection: epidemiology, pathogenesis, and treatment. AIDS. 2017. September 24;31(15):2035–2052. doi: 10.1097/QAD.0000000000001574. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Gemtessa TA, Chirch LM. Update on Hepatitis C Virus and HIV Coinfection. J Clin Transl Hepatol. 2013. December;1(2):109–15. doi: 10.14218/JCTH.2013.00018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Menard A, Colson P, Catherine D, et al. First Real Life Evidence of New Direct-acting Antivirals (DAA) in Co-infected HIV HCV Patients: Better than Ever. Clin Infect Dis. 2016. April 1;62(7):947–9. doi: 10.1093/cid/civ1215. [DOI] [PubMed] [Google Scholar]
- 46.Panel A-IHG. Hepatitis C Guidance 2018 Update: AASLD-IDSA Recommendations for Testing, Managing, and Treating Hepatitis C Virus Infection. Clin Infect Dis. 2018. October 30;67(10):1477–1492. doi: 10.1093/cid/ciy585. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Wenning LA, Hanley WD, Brainard DM, et al. Effect of rifampin, a potent inducer of drug-metabolizing enzymes, on the pharmacokinetics of raltegravir. Antimicrob Agents Chemother. 2009. July;53(7):2852–6. doi: 10.1128/AAC.01468-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Dooley KE, Sayre P, Borland J, et al. Safety, tolerability, and pharmacokinetics of the HIV integrase inhibitor dolutegravir given twice daily with rifampin or once daily with rifabutin: results of a phase 1 study among healthy subjects. J Acquir Immune Defic Syndr. 2013. January 1;62(1):21–7. doi: 10.1097/QAI.0b013e318276cda9. [DOI] [PubMed] [Google Scholar]
- 49.Dooley KE, Kaplan R, Mwelase N, et al. Dolutegravir-Based Antiretroviral Therapy for Patients Co-Infected with Tuberculosis and Hiv: A Multicenter, Noncomparative, Open-Label, Randomized Trial. Clin Infect Dis. 2019. March 28. doi: 10.1093/cid/ciz256. [DOI] [PubMed] [Google Scholar]
- 50.Flandre P, Pugliese P, Cuzin L, et al. Risk factors of chronic kidney disease in HIV-infected patients. Clin J Am Soc Nephrol. 2011. July;6(7):1700–7. doi: 10.2215/CJN.09191010. [DOI] [PubMed] [Google Scholar]
- 51.Doshi S, Ucanda M, Hart R, et al. Incidence and Risk Factors for Renal Disease in an Outpatient Cohort of HIV-Infected Patients on Antiretroviral Therapy. Kidney Int Rep. 2019. August;4(8):1075–1084. doi: 10.1016/j.ekir.2019.04.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Roling J, Schmid H, Fischereder M, et al. HIV-associated renal diseases and highly active antiretroviral therapy-induced nephropathy. Clin Infect Dis. 2006. May 15;42(10):1488–95. doi: 10.1086/503566. [DOI] [PubMed] [Google Scholar]
- 53.Woodward CL, Hall AM, Williams IG, et al. Tenofovir-associated renal and bone toxicity. HIV Med. 2009. September;10(8):482–7. doi: 10.1111/j.1468-1293.2009.00716.x. [DOI] [PubMed] [Google Scholar]
- 54.Ray AS, Fordyce MW, Hitchcock MJ. Tenofovir alafenamide: A novel prodrug of tenofovir for the treatment of Human Immunodeficiency Virus. Antiviral Res. 2016. January;125:63–70. doi: 10.1016/j.antiviral.2015.11.009. [DOI] [PubMed] [Google Scholar]
- 55.DeJesus E, Haas B, Segal-Maurer S, et al. Superior Efficacy and Improved Renal and Bone Safety After Switching from a Tenofovir Disoproxil Fumarate- to a Tenofovir Alafenamide-Based Regimen Through 96 Weeks of Treatment. AIDS Res Hum Retroviruses. 2018. April;34(4):337–342. doi: 10.1089/AID.2017.0203. [DOI] [PubMed] [Google Scholar]; * This randomized controlled trial assessed antiretroviral therapy switch to a TDF- or TAF-containing regimens. Improved bone and renal parameters were observed among participants on TAF-containing regimens.
- 56.Gonciulea A, Wang R, Althoff KN, et al. An increased rate of fracture occurs a decade earlier in HIV+ compared with HIV− men. AIDS. 2017. June 19;31(10):1435–1443. doi: 10.1097/QAD.0000000000001493. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Grant PM, Cotter AG. Tenofovir and bone health. Curr Opin HIV AIDS. 2016. May;11(3):326–32. doi: 10.1097/COH.0000000000000248. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Owe-Larsson B, Sall L, Salamon E, et al. HIV infection and psychiatric illness. Afr J Psychiatry (Johannesbg). 2009. May;12(2):115–28. [DOI] [PubMed] [Google Scholar]
- 59.Robertson KR, Smurzynski M, Parsons TD, et al. The prevalence and incidence of neurocognitive impairment in the HAART era. AIDS. 2007. September 12;21(14):1915–21. doi: 10.1097/QAD.0b013e32828e4e27. [DOI] [PubMed] [Google Scholar]
- 60.Arendt G, de Nocker D, von Giesen HJ, et al. Neuropsychiatric side effects of efavirenz therapy. Expert Opin Drug Saf. 2007. March;6(2):147–54. doi: 10.1517/14740338.6.2.147. [DOI] [PubMed] [Google Scholar]
- 61.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. January;18(1):56–63. doi: 10.1111/hiv.12468. [DOI] [PubMed] [Google Scholar]
- 62.Hoffmann C, Llibre JM. Neuropsychiatric Adverse Events with Dolutegravir and Other Integrase Strand Transfer Inhibitors. AIDS Rev. 2019;21(1):4–10. doi: 10.24875/AIDSRev.19000023. [DOI] [PubMed] [Google Scholar]
- 63.O’Halloran JA, Cooley SA, Strain JF, et al. Altered neuropsychological performance and reduced brain volumetrics in people living with HIV on integrase strand transfer inhibitors. AIDS. 2019. July 15;33(9):1477–1483. doi: 10.1097/QAD.0000000000002236. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Feinstein MJ, Hsue PY, Benjamin LA, et al. Characteristics, Prevention, and Management of Cardiovascular Disease in People Living With HIV: A Scientific Statement From the American Heart Association. Circulation. 2019. July 9;140(2):e98–e124. doi: 10.1161/CIR.0000000000000695. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Feinstein MJ, Bahiru E, Achenbach C, et al. Patterns of Cardiovascular Mortality for HIV-Infected Adults in the United States: 1999 to 2013. Am J Cardiol. 2016. January 15;117(2):214–20. doi: 10.1016/j.amjcard.2015.10.030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Palella FJ Jr., Baker RK, Moorman AC, et al. Mortality in the highly active antiretroviral therapy era: changing causes of death and disease in the HIV outpatient study. J Acquir Immune Defic Syndr. 2006. September;43(1):27–34. doi: 10.1097/01.qai.0000233310.90484.16. [DOI] [PubMed] [Google Scholar]
- 67.Friis-Moller N, Sabin CA, Weber R, et al. Combination antiretroviral therapy and the risk of myocardial infarction. N Engl J Med. 2003. November 20;349(21):1993–2003. doi: 10.1056/NEJMoa030218. [DOI] [PubMed] [Google Scholar]
- 68.Brown TT, Cole SR, Li X, et al. Antiretroviral therapy and the prevalence and incidence of diabetes mellitus in the multicenter AIDS cohort study. Arch Intern Med. 2005. May 23;165(10):1179–84. doi: 10.1001/archinte.165.10.1179. [DOI] [PubMed] [Google Scholar]
- 69.van Welzen BJ, Mudrikova T, El Idrissi A, et al. A Review of Non-Alcoholic Fatty Liver Disease in HIV-Infected Patients: The Next Big Thing? Infect Dis Ther. 2019. March;8(1):33–50. doi: 10.1007/s40121-018-0229-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Joy T, Keogh HM, Hadigan C, et al. Relation of body composition to body mass index in HIV-infected patients with metabolic abnormalities. J Acquir Immune Defic Syndr. 2008. February 1;47(2):174–84. doi: 10.1097/QAI.0b013e31815b0792. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Murata H, Hruz PW, Mueckler M. The mechanism of insulin resistance caused by HIV protease inhibitor therapy. J Biol Chem. 2000. July 7;275(27):20251–4. doi: 10.1074/jbc.C000228200. [DOI] [PubMed] [Google Scholar]
- 72.McLaughlin M, Walsh S, Galvin S. Dolutegravir-induced hyperglycaemia in a patient living with HIV. J Antimicrob Chemother. 2018. January 1;73(1):258–260. doi: 10.1093/jac/dkx365. [DOI] [PubMed] [Google Scholar]
- 73.Fong PS, Flynn DM, Evans CD, et al. Integrase strand transfer inhibitor-associated diabetes mellitus: A case report. Int J STD AIDS. 2017. May;28(6):626–628. doi: 10.1177/0956462416675107. [DOI] [PubMed] [Google Scholar]
- 74.Feeney ER, Mallon PW. Insulin resistance in treated HIV infection. Best Pract Res Clin Endocrinol Metab. 2011. June;25(3):443–58. doi: 10.1016/j.beem.2010.11.002. [DOI] [PubMed] [Google Scholar]
- 75.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. 2019. October 14. doi: 10.1093/cid/ciz999. [DOI] [PMC free article] [PubMed] [Google Scholar]; * This pooled analysis of randomized controlled clinical trials of antiretroviral therapy (ART) initiation found that modern ART regimens are all associated with weight gain, but that integrase strand transfer inhibitors were associated with more weight gain than protease inhibitors or non-nucleoside reverse transcriptase inhibitors.
- 76.Bourgi K, Rebeiro PF, Turner M, et al. Greater Weight Gain in Treatment Naive Persons Starting Dolutegravir-Based Antiretroviral Therapy. Clin Infect Dis. 2019. May 17. doi: 10.1093/cid/ciz407. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Kerchberger AM, Sheth AN, Angert CD, et al. Weight Gain Associated with Integrase Stand Transfer Inhibitor Use in Women. Clin Infect Dis. 2019. August 28. doi: 10.1093/cid/ciz853. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.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. December 15;76(5):527–531. doi: 10.1097/QAI.0000000000001525. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Strategies for Management of Anti-Retroviral Therapy I, Groups DADS. Use of nucleoside reverse transcriptase inhibitors and risk of myocardial infarction in HIV-infected patients. AIDS. 2008. September 12;22(14):F17–24. doi: 10.1097/QAD.0b013e32830fe35e. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Elion RA, Althoff KN, Zhang J, et al. Recent Abacavir Use Increases Risk of Type 1 and Type 2 Myocardial Infarctions Among Adults With HIV. J Acquir Immune Defic Syndr. 2018. May 1;78(1):62–72. doi: 10.1097/QAI.0000000000001642. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Feinstein MJ. Cardiovascular disease risk assessment in HIV: navigating data-sparse zones. Heart. 2016. August 1;102(15):1157–8. doi: 10.1136/heartjnl-2016-309752. [DOI] [PubMed] [Google Scholar]
- 82.Feinstein MJ, Nance RM, Drozd DR, et al. Assessing and Refining Myocardial Infarction Risk Estimation Among Patients With Human Immunodeficiency Virus: A Study by the Centers for AIDS Research Network of Integrated Clinical Systems. JAMA Cardiol. 2017. February 1;2(2):155–162. doi: 10.1001/jamacardio.2016.4494. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Thompson-Paul AM, Lichtenstein KA, Armon C, et al. Cardiovascular Disease Risk Prediction in the HIV Outpatient Study. Clin Infect Dis. 2016. December 1;63(11):1508–1516. doi: 10.1093/cid/ciw615. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Triant VA, Perez J, Regan S, et al. Cardiovascular Risk Prediction Functions Underestimate Risk in HIV Infection. Circulation. 2018. May 22;137(21):2203–2214. doi: 10.1161/CIRCULATIONAHA.117.028975. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Wiggins BS, Lamprecht DG Jr., Page RL 2nd, et al. Recommendations for Managing Drug-Drug Interactions with Statins and HIV Medications. Am J Cardiovasc Drugs. 2017. Oct;17(5):375–389. doi: 10.1007/s40256-017-0222-7. [DOI] [PubMed] [Google Scholar]
- 86.Brocklehurst P, French R. The association between maternal HIV infection and perinatal outcome: a systematic review of the literature and meta-analysis. Br J Obstet Gynaecol. 1998. August;105(8):836–48. doi: 10.1111/j.1471-0528.1998.tb10227.x. [DOI] [PubMed] [Google Scholar]
- 87.Recommendations for the Use of Antiretroviral Drugs in Pregnant Women with HIV Infection and Interventions to Reduce Perinatal HIV Transmission in the United States. Available at: https://aidsinfo.nih.gov/contentfiles/lvguidelines/PerinatalGL.pdf Accessed 7/1/19.
- 88.Jeong H Altered drug metabolism during pregnancy: hormonal regulation of drug-metabolizing enzymes. Expert Opin Drug Metab Toxicol. 2010. June;6(6):689–99. doi: 10.1517/17425251003677755. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Notarianni LJ. Plasma protein binding of drugs in pregnancy and in neonates. Clin Pharmacokinet. 1990. January;18(1):20–36. doi: 10.2165/00003088-199018010-00002. [DOI] [PubMed] [Google Scholar]
- 90.Hodge LS, Tracy TS. Alterations in drug disposition during pregnancy: implications for drug therapy. Expert Opin Drug Metab Toxicol. 2007. August;3(4):557–71. doi: 10.1517/17425225.3.4.557. [DOI] [PubMed] [Google Scholar]
- 91.Zash R, Makhema J, Shapiro RL. Neural-Tube Defects with Dolutegravir Treatment from the Time of Conception. N Engl J Med. 2018. September 6;379(10):979–981. doi: 10.1056/NEJMc1807653. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Zash R, Holmes L, Diseko M, et al. Neural-Tube Defects and Antiretroviral Treatment Regimens in Botswana. N Engl J Med. 2019. July 22. doi: 10.1056/NEJMoa1905230. [DOI] [PMC free article] [PubMed] [Google Scholar]; * Using Botswanan birth-outcomes surveillance data, this study assessed the prevelance of neural tube defects (NTDs) by maternal ART exposure. Slightly higher prevelance of NTDs was associated with dolutegravir exposure at the time of conception compared to other regimens.
- 93.Schalkwijk S, Colbers A, Konopnicki D, et al. First reported use of elvitegravir and cobicistat during pregnancy. AIDS. 2016. March 13;30(5):807–8. doi: 10.1097/QAD.0000000000000976. [DOI] [PubMed] [Google Scholar]
- 94.Momper JD, Best BM, Wang J, et al. Elvitegravir/cobicistat pharmacokinetics in pregnant and postpartum women with HIV. AIDS. 2018. November 13;32(17):2507–2516. doi: 10.1097/QAD.0000000000001992. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Scarsi KK, Darin KM, Nakalema S, et al. Unintended Pregnancies Observed With Combined Use of the Levonorgestrel Contraceptive Implant and Efavirenz-based Antiretroviral Therapy: A Three-Arm Pharmacokinetic Evaluation Over 48 Weeks. Clin Infect Dis. 2016. March 15;62(6):675–682. doi: 10.1093/cid/civ1001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Scarsi K et al. Double-dose levonorgestrel implant does not fully overcome interaction with efavirenz. CROI 2019 Seattle 4–7 March 2019 Oral abstract 51. [Google Scholar]
- 97.Radix A, Sevelius J, Deutsch MB. Transgender women, hormonal therapy and HIV treatment: a comprehensive review of the literature and recommendations for best practices. J Int AIDS Soc. 2016;19(3 Suppl 2):20810. doi: 10.7448/IAS.19.3.20810. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Braun HM, Candelario J, Hanlon CL, et al. Transgender Women Living with HIV Frequently Take Antiretroviral Therapy and/or Feminizing Hormone Therapy Differently Than Prescribed Due to Drug-Drug Interaction Concerns. LGBT Health. 2017. October;4(5):371–375. doi: 10.1089/lgbt.2017.0057. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Announcement: Monitoring Selected National HIV Prevention and Care Objectives. MMWR Morb Mortal Wkly Rep. 2017. July 28;66(29):794. doi: 10.15585/mmwr.mm6629a6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Risher KA, Kapoor S, Daramola AM, et al. Challenges in the Evaluation of Interventions to Improve Engagement Along the HIV Care Continuum in the United States: A Systematic Review. AIDS Behav. 2017. July;21(7):2101–2123. doi: 10.1007/s10461-017-1687-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Mugavero MJ, Amico KR, Horn T, et al. The state of engagement in HIV care in the United States: from cascade to continuum to control. Clin Infect Dis. 2013. October;57(8):1164–71. doi: 10.1093/cid/cit420. [DOI] [PubMed] [Google Scholar]
- 102.Rao D, Feldman BJ, Fredericksen RJ, et al. A structural equation model of HIV-related stigma, depressive symptoms, and medication adherence. AIDS Behav. 2012. April;16(3):711–6. doi: 10.1007/s10461-011-9915-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Golin C, Isasi F, Bontempi JB, et al. Secret pills: HIV-positive patients’ experiences taking antiretroviral therapy in North Carolina. AIDS Educ Prev. 2002. August;14(4):318–29. [DOI] [PubMed] [Google Scholar]
- 104.Warren-Jeanpiere L, Dillaway H, Hamilton P, et al. Taking it one day at a time: African American women aging with HIV and co-morbidities. AIDS Patient Care STDS. 2014. July;28(7):372–80. doi: 10.1089/apc.2014.0024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Turan B, Smith W, Cohen MH, et al. Mechanisms for the Negative Effects of Internalized HIV-Related Stigma on Antiretroviral Therapy Adherence in Women: The Mediating Roles of Social Isolation and Depression. J Acquir Immune Defic Syndr. 2016. June 1;72(2):198–205. doi: 10.1097/QAI.0000000000000948. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Aidala AA, Wilson MG, Shubert V, et al. Housing Status, Medical Care, and Health Outcomes Among People Living With HIV/AIDS: A Systematic Review. Am J Public Health. 2016. January;106(1):e1–e23. doi: 10.2105/AJPH.2015.302905. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Sullivan TP. The intersection of intimate partner violence and HIV: detection, disclosure, discussion, and implications for treatment adherence. Top Antivir Med. 2019. May;27(2):84–87. [PMC free article] [PubMed] [Google Scholar]
- 108.Wohl AR, Garland WH, Wu J, et al. A youth-focused case management intervention to engage and retain young gay men of color in HIV care. AIDS Care. 2011. August;23(8):988–97. doi: 10.1080/09540121.2010.542125. [DOI] [PubMed] [Google Scholar]
- 109.Terzian AS, Irvine MK, Hollod LM, et al. Effect of HIV Housing Services on Engagement in Care and Treatment, New York City, 2011. AIDS Behav. 2015. November;19(11):2087–96. doi: 10.1007/s10461-015-1003-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Bradford JB, Coleman S, Cunningham W. HIV System Navigation: an emerging model to improve HIV care access. AIDS Patient Care STDS. 2007;21 Suppl 1:S49–58. doi: 10.1089/apc.2007.9987. [DOI] [PubMed] [Google Scholar]
- 111.Naar-King S, Bradford J, Coleman S, et al. Retention in care of persons newly diagnosed with HIV: outcomes of the Outreach Initiative. AIDS Patient Care STDS. 2007;21 Suppl 1:S40–8. doi: 10.1089/apc.2007.9988. [DOI] [PubMed] [Google Scholar]
- 112.Chander G, Himelhoch S, Moore RD. Substance abuse and psychiatric disorders in HIV-positive patients: epidemiology and impact on antiretroviral therapy. Drugs. 2006;66(6):769–89. doi: 10.2165/00003495-200666060-00004. [DOI] [PubMed] [Google Scholar]
- 113.Whetten K, Reif S, Whetten R, et al. Trauma, mental health, distrust, and stigma among HIV-positive persons: implications for effective care. Psychosom Med. 2008. June;70(5):531–8. doi: 10.1097/PSY.0b013e31817749dc. [DOI] [PubMed] [Google Scholar]
- 114.Bengtson AM, Pence BW, Mimiaga MJ, et al. Depressive Symptoms and Engagement in Human Immunodeficiency Virus Care Following Antiretroviral Therapy Initiation. Clin Infect Dis. 2019. January 18;68(3):475–481. doi: 10.1093/cid/ciy496. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Pence BW, O’Donnell JK, Gaynes BN. Falling through the cracks: the gaps between depression prevalence, diagnosis, treatment, and response in HIV care. AIDS. 2012. March 13;26(5):656–8. doi: 10.1097/QAD.0b013e3283519aae. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Altice FL, Bruce RD, Lucas GM, et al. HIV treatment outcomes among HIV-infected, opioid-dependent patients receiving buprenorphine/naloxone treatment within HIV clinical care settings: results from a multisite study. J Acquir Immune Defic Syndr. 2011. March 1;56 Suppl 1:S22–32. doi: 10.1097/QAI.0b013e318209751e. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Cunningham CO, Sohler NL, Cooperman NA, et al. Strategies to improve access to and utilization of health care services and adherence to antiretroviral therapy among HIV-infected drug users. Subst Use Misuse. 2011;46(2–3):218–32. doi: 10.3109/10826084.2011.522840. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Clay PG, Yuet WC, Moecklinghoff CH, et al. A meta-analysis comparing 48-week treatment outcomes of single and multi-tablet antiretroviral regimens for the treatment of people living with HIV. AIDS Res Ther. 2018. October 30;15(1):17. doi: 10.1186/s12981-018-0204-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Michienzi SM, Schriever CA, Badowski ME. Abacavir/lamivudine/dolutegravir single tablet regimen in patients with human immunodeficiency virus and end-stage renal disease on hemodialysis. Int J STD AIDS. 2019. February;30(2):181–187. doi: 10.1177/0956462418800865. [DOI] [PubMed] [Google Scholar]
- 120.Emu B, Fessel J, Schrader S, et al. Phase 3 Study of Ibalizumab for Multidrug-Resistant HIV-1. N Engl J Med. 2018. August 16;379(7):645–654. doi: 10.1056/NEJMoa1711460. [DOI] [PubMed] [Google Scholar]
- 121.Hazra R, Siberry GK, Mofenson LM. Growing up with HIV: children, adolescents, and young adults with perinatally acquired HIV infection. Annu Rev Med. 2010;61:169–85. doi: 10.1146/annurev.med.050108.151127. [DOI] [PubMed] [Google Scholar]
- 122.Rudy BJ, Murphy DA, Harris DR, et al. Prevalence and interactions of patient-related risks for nonadherence to antiretroviral therapy among perinatally infected youth in the United States. AIDS Patient Care STDS. 2010. February;24(2):97–104. doi: 10.1089/apc.2009.0198. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.MacDonell K, Naar-King S, Huszti H, et al. Barriers to medication adherence in behaviorally and perinatally infected youth living with HIV. AIDS Behav. 2013. January;17(1):86–93. doi: 10.1007/s10461-012-0364-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Ryscavage P, Anderson EJ, Sutton SH, et al. Clinical outcomes of adolescents and young adults in adult HIV care. J Acquir Immune Defic Syndr. 2011. October 1;58(2):193–7. doi: 10.1097/QAI.0b013e31822d7564. [DOI] [PubMed] [Google Scholar]
- 125.Straub DM, Tanner AE. Health-care transition from adolescent to adult services for young people with HIV. Lancet Child Adolesc Health. 2018. March;2(3):214–222. doi: 10.1016/S2352-4642(18)30005-1. [DOI] [PubMed] [Google Scholar]
- 126.Naar-King S, Templin T, Wright K, et al. Psychosocial factors and medication adherence in HIV-positive youth. AIDS Patient Care STDS. 2006. January;20(1):44–7. doi: 10.1089/apc.2006.20.44. [DOI] [PubMed] [Google Scholar]
- 127.Mellins CA, Brackis-Cott E, Leu CS, et al. Rates and types of psychiatric disorders in perinatally human immunodeficiency virus-infected youth and seroreverters. J Child Psychol Psychiatry. 2009. September;50(9):1131–8. doi: 10.1111/j.1469-7610.2009.02069.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Van Dyke RB, Patel K, Siberry GK, et al. Antiretroviral treatment of US children with perinatally acquired HIV infection: temporal changes in therapy between 1991 and 2009 and predictors of immunologic and virologic outcomes. J Acquir Immune Defic Syndr. 2011. June 1;57(2):165–73. doi: 10.1097/QAI.0b013e318215c7b1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Belzer ME, Kolmodin MacDonell K, Clark LF, et al. Acceptability and Feasibility of a Cell Phone Support Intervention for Youth Living with HIV with Nonadherence to Antiretroviral Therapy. AIDS Patient Care STDS. 2015. June;29(6):338–45. doi: 10.1089/apc.2014.0282. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Mimiaga MJ, Kuhns LM, Biello KB, et al. Positive STEPS - a randomized controlled efficacy trial of an adaptive intervention for strengthening adherence to antiretroviral HIV treatment among youth: study protocol. BMC Public Health. 2018. July 13;18(1):867. doi: 10.1186/s12889-018-5815-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Lodi S, Phillips A, Touloumi G, et al. Time from human immunodeficiency virus seroconversion to reaching CD4+ cell count thresholds <200, <350, and <500 Cells/mm(3): assessment of need following changes in treatment guidelines. Clin Infect Dis. 2011. October;53(8):817–25. doi: 10.1093/cid/cir494. [DOI] [PubMed] [Google Scholar]
- 132.Collaboration of Observational HIVERESG, Sabin CA, Smith CJ, et al. Response to combination antiretroviral therapy: variation by age. AIDS. 2008. July 31;22(12):1463–73. doi: 10.1097/QAD.0b013e3282f88d02. [DOI] [PubMed] [Google Scholar]
- 133.Nogueras M, Navarro G, Anton E, et al. Epidemiological and clinical features, response to HAART, and survival in HIV-infected patients diagnosed at the age of 50 or more. BMC Infect Dis. 2006. November 6;6:159. doi: 10.1186/1471-2334-6-159. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Grabar S, Kousignian I, Sobel A, et al. Immunologic and clinical responses to highly active antiretroviral therapy over 50 years of age. Results from the French Hospital Database on HIV. AIDS. 2004. October 21;18(15):2029–38. [DOI] [PubMed] [Google Scholar]
- 135.Viard JP, Mocroft A, Chiesi A, et al. Influence of age on CD4 cell recovery in human immunodeficiency virus-infected patients receiving highly active antiretroviral therapy: evidence from the EuroSIDA study. J Infect Dis. 2001. April 15;183(8):1290–4. doi: 10.1086/319678. [DOI] [PubMed] [Google Scholar]
- 136.Tracy LA, Struble K, Firnhaber C, et al. Age Differences by Sex in Antiretroviral-Naive Participants: Pooled Analysis from Randomized Clinical Trials. J Assoc Nurses AIDS Care. 2018. May-Jun;29(3):371–382. doi: 10.1016/j.jana.2018.01.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Edwards JK, Cole SR, Westreich D, et al. Age at Entry Into Care, Timing of Antiretroviral Therapy Initiation, and 10-Year Mortality Among HIV-Seropositive Adults in the United States. Clin Infect Dis. 2015. October 1;61(7):1189–95. doi: 10.1093/cid/civ463. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Horberg MA, Hurley LB, Klein DB, et al. The HIV Care Cascade Measured Over Time and by Age, Sex, and Race in a Large National Integrated Care System. AIDS Patient Care STDS. 2015. November;29(11):582–90. doi: 10.1089/apc.2015.0139. [DOI] [PubMed] [Google Scholar]
- 139.Deeks SG. HIV infection, inflammation, immunosenescence, and aging. Annu Rev Med. 2011;62:141–55. doi: 10.1146/annurev-med-042909-093756. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Hasse B, Ledergerber B, Furrer H, et al. Morbidity and aging in HIV-infected persons: the Swiss HIV cohort study. Clin Infect Dis. 2011. December;53(11):1130–9. doi: 10.1093/cid/cir626. [DOI] [PubMed] [Google Scholar]
- 141.Smit M, Brinkman K, Geerlings S, et al. Future challenges for clinical care of an ageing population infected with HIV: a modelling study. Lancet Infect Dis. 2015. July;15(7):810–8. doi: 10.1016/S1473-3099(15)00056-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Ware D, Palella FJ Jr., Chew KW, et al. Prevalence and trends of polypharmacy among HIV-positive and -negative men in the Multicenter AIDS Cohort Study from 2004 to 2016. PLoS One. 2018;13(9):e0203890. doi: 10.1371/journal.pone.0203890. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Schouten J, Wit FW, Stolte IG, et al. Cross-sectional comparison of the prevalence of age-associated comorbidities and their risk factors between HIV-infected and uninfected individuals: the AGEhIV cohort study. Clin Infect Dis. 2014. December 15;59(12):1787–97. doi: 10.1093/cid/ciu701. [DOI] [PubMed] [Google Scholar]
- 144.Desquilbet L, Jacobson LP, Fried LP, et al. HIV-1 infection is associated with an earlier occurrence of a phenotype related to frailty. J Gerontol A Biol Sci Med Sci. 2007. November;62(11):1279–86. [DOI] [PubMed] [Google Scholar]
- 145.Schrack JA, Althoff KN, Jacobson LP, et al. Accelerated Longitudinal Gait Speed Decline in HIV-Infected Older Men. J Acquir Immune Defic Syndr. 2015. December 1;70(4):370–6. doi: 10.1097/QAI.0000000000000731. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Schrack JA, Jacobson LP, Althoff KN, et al. Effect of HIV-infection and cumulative viral load on age-related decline in grip strength. AIDS. 2016. November 13;30(17):2645–2652. doi: 10.1097/QAD.0000000000001245. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Johs NA, Wu K, Tassiopoulos K, et al. Disability Among Middle-Aged and Older Persons With Human Immunodeficiency Virus Infection. Clin Infect Dis. 2017. July 1;65(1):83–91. doi: 10.1093/cid/cix253. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Sharma A, Hoover DR, Shi Q, et al. Longitudinal study of falls among HIV-infected and uninfected women: the role of cognition. Antivir Ther. 2018;23(2):179–190. doi: 10.3851/IMP3195. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Tassiopoulos K, Abdo M, Wu K, et al. Frailty is strongly associated with increased risk of recurrent falls among older HIV-infected adults. AIDS. 2017. October 23;31(16):2287–2294. doi: 10.1097/QAD.0000000000001613. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Erlandson KM, Allshouse AA, Jankowski CM, et al. Relationship of physical function and quality of life among persons aging with HIV infection. AIDS. 2014. August 24;28(13):1939–43. doi: 10.1097/QAD.0000000000000384. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Jacks A, Wainwright DA, Salazar L, et al. Neurocognitive deficits increase risk of poor retention in care among older adults with newly diagnosed HIV infection. AIDS. 2015. August 24;29(13):1711–4. doi: 10.1097/QAD.0000000000000700. [DOI] [PubMed] [Google Scholar]
- 152.Grov C, Golub SA, Parsons JT, et al. Loneliness and HIV-related stigma explain depression among older HIV-positive adults. AIDS Care. 2010. May;22(5):630–9. doi: 10.1080/09540120903280901. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Llibre JM, Hung CC, Brinson C, et al. Efficacy, safety, and tolerability of dolutegravir-rilpivirine for the maintenance of virological suppression in adults with HIV-1: phase 3, randomised, non-inferiority SWORD-1 and SWORD-2 studies. Lancet. 2018. March 3;391(10123):839–849. doi: 10.1016/S0140-6736(17)33095-7. [DOI] [PubMed] [Google Scholar]
- 154.Perez-Molina JA, Rubio R, Rivero A, et al. Dual treatment with atazanavir-ritonavir plus lamivudine versus triple treatment with atazanavir-ritonavir plus two nucleos(t)ides in virologically stable patients with HIV-1 (SALT): 48 week results from a randomised, open-label, non-inferiority trial. Lancet Infect Dis. 2015. July;15(7):775–84. doi: 10.1016/S1473-3099(15)00097-3. [DOI] [PubMed] [Google Scholar]
- 155.Fabbiani M, Gagliardini R, Ciccarelli N, et al. Atazanavir/ritonavir with lamivudine as maintenance therapy in virologically suppressed HIV-infected patients: 96 week outcomes of a randomized trial. J Antimicrob Chemother. 2018. July 1;73(7):1955–1964. doi: 10.1093/jac/dky123. [DOI] [PubMed] [Google Scholar]
- 156.Pulido F, Ribera E, Lagarde M, et al. Dual Therapy With Darunavir and Ritonavir Plus Lamivudine vs Triple Therapy With Darunavir and Ritonavir Plus Tenofovir Disoproxil Fumarate and Emtricitabine or Abacavir and Lamivudine for Maintenance of Human Immunodeficiency Virus Type 1 Viral Suppression: Randomized, Open-Label, Noninferiority DUAL-GESIDA 8014-RIS-EST45 Trial. Clin Infect Dis. 2017. November 29;65(12):2112–2118. doi: 10.1093/cid/cix734. [DOI] [PubMed] [Google Scholar]
- 157.Arribas JR, Girard PM, Landman R, et al. Dual treatment with lopinavir-ritonavir plus lamivudine versus triple treatment with lopinavir-ritonavir plus lamivudine or emtricitabine and a second nucleos(t)ide reverse transcriptase inhibitor for maintenance of HIV-1 viral suppression (OLE): a randomised, open-label, non-inferiority trial. Lancet Infect Dis. 2015. July;15(7):785–92. doi: 10.1016/S1473-3099(15)00096-1. [DOI] [PubMed] [Google Scholar]
- 158.Capetti AF, Cossu MV, Orofino G, et al. A dual regimen of ritonavir/darunavir plus dolutegravir for rescue or simplification of rescue therapy: 48 weeks’ observational data. BMC Infect Dis. 2017. September 30;17(1):658. doi: 10.1186/s12879-017-2755-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Wheeler J, Chan S, Harrigan PR, et al. Dolutegravir with boosted darunavir treatment simplification for the transmitted HIV thymidine analog resistance in Manitoba, Canada. Int J STD AIDS. 2018. April;29(5):520–522. doi: 10.1177/0956462418760426. [DOI] [PubMed] [Google Scholar]
- 160.Taiwo BO, Marconi VC, Berzins B, et al. Dolutegravir Plus Lamivudine Maintains Human Immunodeficiency Virus-1 Suppression Through Week 48 in a Pilot Randomized Trial. Clin Infect Dis. 2018. May 17;66(11):1794–1797. doi: 10.1093/cid/cix1131. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Cahn P, Madero JS, Arribas JR, et al. Dolutegravir plus lamivudine versus dolutegravir plus tenofovir disoproxil fumarate and emtricitabine in antiretroviral-naive adults with HIV-1 infection (GEMINI-1 and GEMINI-2): week 48 results from two multicentre, double-blind, randomised, non-inferiority, phase 3 trials. Lancet. 2019. January 12;393(10167):143–155. doi: 10.1016/S0140-6736(18)32462-0. [DOI] [PubMed] [Google Scholar]
- 162.Margolis DA, Gonzalez-Garcia J, Stellbrink HJ, et al. Long-acting intramuscular cabotegravir and rilpivirine in adults with HIV-1 infection (LATTE-2): 96-week results of a randomised, open-label, phase 2b, non-inferiority trial. Lancet. 2017. September 23;390(10101):1499–1510. doi: 10.1016/S0140-6736(17)31917-7. [DOI] [PubMed] [Google Scholar]
- 163.Gulick RM, Flexner C. Long-Acting HIV Drugs for Treatment and Prevention. Annu Rev Med. 2019. January 27;70:137–150. doi: 10.1146/annurev-med-041217-013717. [DOI] [PubMed] [Google Scholar]
- 164.Cohen YZ, Caskey M. Broadly neutralizing antibodies for treatment and prevention of HIV-1 infection. Curr Opin HIV AIDS. 2018. July;13(4):366–373. doi: 10.1097/COH.0000000000000475. [DOI] [PubMed] [Google Scholar]
- 165.Kumar R, Qureshi H, Deshpande S, et al. Broadly neutralizing antibodies in HIV-1 treatment and prevention. Ther Adv Vaccines Immunother. 2018. August;6(4):61–68. doi: 10.1177/2515135518800689. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Mendoza P, Gruell H, Nogueira L, et al. Combination therapy with anti-HIV-1 antibodies maintains viral suppression. Nature. 2018. September;561(7724):479–484. doi: 10.1038/s41586-018-0531-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Gautam R, Nishimura Y, Gaughan N, et al. A single injection of crystallizable fragment domain-modified antibodies elicits durable protection from SHIV infection. Nat Med. 2018. May;24(5):610–616. doi: 10.1038/s41591-018-0001-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Nakata H, Amano M, Koh Y, et al. Activity against human immunodeficiency virus type 1, intracellular metabolism, and effects on human DNA polymerases of 4’-ethynyl-2-fluoro-2’-deoxyadenosine. Antimicrob Agents Chemother. 2007. August;51(8):2701–8. doi: 10.1128/AAC.00277-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169.Barrett SE, Teller RS, Forster SP, et al. Extended-Duration MK-8591-Eluting Implant as a Candidate for HIV Treatment and Prevention. Antimicrob Agents Chemother. 2018. October;62(10). doi: 10.1128/AAC.01058-18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Moffatt K, Wang Y, Raj Singh TR, et al. Microneedles for enhanced transdermal and intraocular drug delivery. Curr Opin Pharmacol. 2017. October;36:14–21. doi: 10.1016/j.coph.2017.07.007. [DOI] [PubMed] [Google Scholar]
- 171.Stewart SA, Dominguez-Robles J, Donnelly RF, et al. Implantable Polymeric Drug Delivery Devices: Classification, Manufacture, Materials, and Clinical Applications. Polymers (Basel). 2018. December 12;10(12). doi: 10.3390/polym10121379. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.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. Clinical infectious diseases : an official publication of the Infectious Diseases Society of America. 2018. August 31. doi: 10.1093/cid/ciy540. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.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. The lancet HIV. 2018. May;5(5):e211–e220. doi: 10.1016/s2352-3018(18)30021-3. [DOI] [PubMed] [Google Scholar]
- 174.Novikova M, Zhang Y, Freed EO, et al. Multiple Roles of HIV-1 Capsid during the Virus Replication Cycle. Virol Sin. 2019. April;34(2):119–134. doi: 10.1007/s12250-019-00095-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175.Rutsaert S, Steens JM, Gineste P, et al. Safety, tolerability and impact on viral reservoirs of the addition to antiretroviral therapy of ABX464, an investigational antiviral drug, in individuals living with HIV-1: a Phase IIa randomised controlled study. J Virus Erad. 2019. January 1;5(1):10–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Cohen MS, Chen YQ, McCauley M, et al. Antiretroviral Therapy for the Prevention of HIV-1 Transmission. N Engl J Med. 2016. September 1;375(9):830–9. doi: 10.1056/NEJMoa1600693. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.Cohn SE, Haddad LB, Sheth AN, et al. Parenting Desires Among Individuals Living With Human Immunodeficiency Virus in the United States. Open Forum Infect Dis. 2018. October;5(10):ofy232. doi: 10.1093/ofid/ofy232. [DOI] [PMC free article] [PubMed] [Google Scholar]

