Back to Journals » HIV/AIDS - Research and Palliative Care » Volume 12

Antiretroviral Therapy-Associated Metabolic Complications: Review of the Recent Studies

Authors Thet D , Siritientong T 

Received 3 August 2020

Accepted for publication 3 September 2020

Published 2 October 2020 Volume 2020:12 Pages 507—524

DOI https://doi.org/10.2147/HIV.S275314

Checked for plagiarism Yes

Review by Single anonymous peer review

Peer reviewer comments 2

Editor who approved publication: Professor Bassel Sawaya



Daylia Thet, Tippawan Siritientong

Department of Food and Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand

Correspondence: Tippawan Siritientong
Department of Food and Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Phyathai Road, Pathumwan, Bangkok 10330, Thailand
Tel +6661-956-2898
Fax +662-218-8291
Email [email protected]

Abstract: The extensive utilization of antiretroviral therapy (ART) has successfully improved human immunodeficiency virus (HIV)-associated complications. The incidence of opportunistic infections is decreased by the viral load suppression and the CD4 count promotion. However, metabolic complications, commonly bone demineralization, lipodystrophy, and lactic acidosis, are arising following the adaptation of long-term ART. The events are not drug-specific, but the severity and incidence individually vary depending upon classes of drugs. Such concerning occurrences may lead to discontinuation of current therapy or switching to another regimen with fewer adverse effects. The purpose of this review is to demonstrate the common metabolic abnormalities associated with each class of widely used ART in people living with HIV (PLHIV). Electronic databases such as PubMed, ScienceDirect, Scopus, Google Scholar, SciFinder, and Web of Science were used for the literature search. A better understanding of ART-associated metabolic adverse effects is helpful in various clinical settings so that therapists may optimize treatments in this population.

Keywords: HIV, antiretroviral therapy, lipodystrophy, metabolic, lipid profile

Introduction

Since 10 years ago, acquired immune deficiency syndrome (AIDS)-related death has declined by 39% according to the global statistics of 2020.1 The mortality related to human immunodeficiency virus (HIV) infection has been decreased with the discovery of effective antiretroviral therapy (ART).2,3 Globally, of the 67% of patients receiving treatment, 59% had successfully viral suppression in 2019.1 On the other hand, the increase in survival makes HIV infection a chronic illness which requires life-long treatment. Despite therapeutic effectiveness, ART regimens have long been accompanied with various metabolic complications.4 Consequently, ART associated metabolic abnormalities become increasing concerns among people living with HIV (PLHIV). The severity and prevalence may be varied according to the individual or the type of regimen. Highly Active Antiretroviral Therapy (HAART) are combination regimens consisting of at least three ARTs from two different mechanism of actions. Commonly used regimens are two nucleoside reverse transcriptase inhibitors (NRTIs) plus one or two protease inhibitors (PIs) and/or a non-nucleoside reverse transcriptase inhibitor (NNRTI). NRTIs such as abacavir, lamivudine, zidovudine, tenofovir, and stavudine; PIs such as lopinavir, ritonavir, saquinavir, and nelfinavir; and NNRTIs such as nevirapine, efavirenz, and etravirine have long been widely used as first or second line ART.5

Some metabolic side-effects of antiretroviral drugs include bone demineralization, increased cholesterol and triglyceride levels, and abnormalities in fat distribution.4 Generally, the prevalence of reduced bone mineral density (BMD) was higher in PI-treated HIV-infected patients. They had three times higher prevalence of osteoporosis compared with uninfected people.6 Lipodystrophy was a common metabolic adverse event which was associated with central adiposity, peripheral fat wasting, cushingoid feature, lipomas, and breast hypertrophy.7 In particular, HIV-infected patients with abnormal fat distribution or lipoatrophy may experience dyslipidemia, which has become a significant issue.8 In some HIV-infected individuals, serious metabolic events may lead to discontinuation of therapy or switching to another regimen with fewer side-effects. This review aims to better understand metabolic abnormalities associated with each class of ART, highlighting the HAART regimen. Therefore, advanced strategies for modification of therapy to mitigate complications in the HIV population can be more developed.

Methods

We performed a comprehensive literature search for the articles published between January 2000 and June 2020. Clinical trials, cross-sectional studies, and cohort studies which are available in English language were included. The articles which demonstrated metabolic abnormalities were mainly reviewed with a specific focus on lipodystrophy, abnormalities of lipid profiles, glucose levels, and bone. Other interesting side-effects related to ART were also thoroughly reviewed. Exclusion criteria were pilot studies, research protocols, conference proceedings, book chapters, review papers, and articles without full text. Terms such as human immunodeficiency virus/HIV/antiretroviral therapy/acquired immunodeficiency syndrome/metabolic/lipodystrophy/lipoatrophy/lactic acidosis/bone demineralization/osteopenia/dyslipidemia/nucleoside reverse transcriptase inhibitor/abacavir/zidovudine/lamivudine/stavudine/tenofovir disoproxil fumarate (tenofovir DF)/tenofovir alafenamide (tenofovir AF)/didanosine/emtricitabine/non-nucleoside reverse transcriptase inhibitor/nevirapine/efavirenz/etravirine/rilpivirine/protease inhibitor/lopinavir/ritonavir/atazanavir/darunavir/indinavir/nelfinavir/saquinavir/integrase inhibitor/dolutegravir/raltegravir were used to search in the electronic databases: PubMed, ScienceDirect, Scopus, Google Scholar, SciFinder, and Web of Science.

Pathophysiology

Lipodystrophy refers to the abnormal distribution of adipose tissue.9 It includes lipohypertrophy, which is fat accumulation in central areas: abdomen, breast, neck, and lipoatrophy, which is loss of fat in proximal areas: face, arms, legs, buttock. The exact etiology of lipodystrophy is still unknown.9 The issue that should be emphasized is both ART and HIV cause changes in lipid distribution. The production of inflammatory cytokines such as interleukins are provoked, which results in adipose cell destruction. Moreover, insulin signaling function and glucose transport were impaired. As a consequence, ART associated lipodystrophy increases the risk for dyslipidemia, insulin resistance, diabetes mellitus, and heart disease.10 Dyslipidemia is the condition in which total cholesterol levels above 5 mmol/L, triglycerides >1.7 mmol/L, and low-density lipoprotein cholesterol (LDL-C) levels of >3 mmol/L.11 Lactic acidosis is also common, which referred to the level of uncuffed lactate >5 mmol/L and arterial pH <7.35 or a total venous carbon dioxide <20 mmol/L.11 In some patients receiving ART, there was elevation of aspartate aminotransferase (AST) and alanine aminotransferase (ALT) (>40 U/L) which were predictive factors of metabolic complications also in the general population.12

Animal Studies

The safety and efficacy of antiretroviral drugs have been monitored, as has the metabolic alteration during the treatment period tested in preclinical trial studies.1316 Conradie et al13 examined the effects of ART on bone metabolism and lipodystrophy in rats. Stavudine (6.2 mg/kg), tenofovir DF (26.6 mg/kg), lopinavir/ritonavir (70.8 mg/kg), and water (1.5 mL) were subjected orally to 40 male rats for 9 weeks. Under examination by dual energy X-ray absorptiometry (DEXA), stavudine monotherapy was associated with a decrease in femoral BMD (0.247±0.02 g/cm2, P<0.05). Biomechanical testing showed that the load-bearing capacity and strength of femurs of stavudine-treated rats were significantly lower than those of other groups (P<0.05). Both NRTIs (stavudine and tenofovir) resulted in significant bone marrow adiposity. Another NRTI, zidovudine, also reduced BMD in mice.14 Mice were orally treated with 0.25 mg zidovudine daily for 4 weeks while controls received water. Under examination by DEXA, zidovudine treated mice showed a significant decrease in BMD (P<0.04). Metabolic consequences of PIs administration in mice has been reported.15,16 C57BL/6 mice were assigned combined lopinavir/ritonavir (50/12.5 to 200/50 mg/kg) for 3 weeks. There were significant changes in metabolic parameters showing hyperlipidemia, hyperinsulinemia, and lipodystrophy symptoms as the dose-dependent manner. The findings indicated that PIs, particularly lopinavir/ritonavir treatment, result in lipodystrophy symptoms which are similar to those found in human.15 Tsai et al16 examined the 4-week treatment effects of lopinavir/ritonavir in C57BL/6 mice. Increased total cholesterol and high-density lipoprotein cholesterol (HDL-C) levels were observed in addition to intracellular lipid accumulation. In a mice model, daily administration of abacavir/lamivudine and lopinavir/ritonavir may probably increase metabolic disorders regardless of HIV infection.16

In vitro Studies

Lipin-1 is a gene necessary for the normal development of adipose tissues.17 HIV-infected patients with lipodystrophy or metabolic abnormalities due to ART have decreased lipin-1 mRNA expression in subcutaneous adipose tissues.18 In differentiated 3T3-L1 cells, treatments with PIs resulted in reduction in lipin-1 mRNA expression by promoting the miRNA-218 expression. Although the role of miRNA is still ambiguous, it was reported to have a relationship with glucose transporter type 4 (GLUT-4) gene expression which is the insulin-stimulated glucose transporter. During treatment with PIs, increased miRNA caused reduction in GLUT-4 which in turn resulted in insulin resistance and metabolic abnormalities. 3T3-L1 adipocytes on exposure to lopinavir: ritonavir combination showed a significant reduction in lipid (−13.2% in concentration of 12 μM:3 μM and −14.5% in 16 μM:4 μM, P<0.05).18 Caron et al19 examined the long-term effects of NRTIs on lipid accumulation and mitochondrial function of differentiating 3T3-F442A and differentiated 3T3 adipocytes. Among the treated NRTIs, zidovudine (1 μM) and stavudine (10 μM) showed changes in lipid phenotype, content, and expression of metabolic markers. Thus, in vitro after 6–11 days of zidovudine and stavudine treatment at near Cmax concentrations, there were decreases in lipid content, survival of adipose cells, and mitochondrial activity. These results seemed to be relevant to the lipodystrophy of patients treated with low dose concentrations for months or years of treatment period. Nolan et al20 explored the effect of cellular mitochondrial DNA (mtDNA) reduction in patients receiving NRTIs therapy. The amount of mtDNA was significantly decreased in NRTIs (zidovudine/stavudine) group compared to ART-naïve HIV-positive controls (P<0.001). There were also abnormalities of adipose tissue among patients receiving NRTIs.

Genetic and Ethnic Influence

The risk of metabolic abnormalities including dyslipidemia can be predicted by genetic factors. The effects of apolipoprotein C3 (APOC3), apolipoprotein E (APOE) on ART-associated lipid abnormalities have been explored.21 The effects of alleles of APOC3 and APOE on plasma cholesterol and triglycerides in patients receiving ritonavir were prospectively monitored over 3-year study period. Triglyceride levels were significantly high in patients who had alleles of APOC3-482CT, −455TC, and −3238CG (P=0.006). Meanwhile, there were elevations of non-high density lipoprotein cholesterol (non-HDL-C) levels in patients with APOE-3/4 and E-4/4 genotypes (P=0.038). Similarly, Fauvel et al22 evaluated the influence of APOC3 gene polymorphism on dyslipidemia in male patients receiving PIs. Indinavir, ritonavir, saquinavir, and nelfinavir were administered in combination with two NRTIs; stavudine and lamivudine. During a 3-month period, plasma triglycerides were elevated, and HDL-C levels were decreased in those patients (P<0.01).

The prevalence of metabolic complications was varied among different ethnic groups. Curtis et al23 highlighted the prevalence of metabolic bone abnormalities in a cohort of PLHIV under HAART. Non-African-Americans had higher risk of osteopenia (59%) than African-Americans (26%). Mean urinary N-telopeptide levels were also higher in non-African-Americans. Glucose levels were also varied among ethnicities. HIV-infected African-Americans and Hispanics had higher risk of diabetes than non-Hispanic whites.24 HbA1c and 2 hour glucose levels were significantly higher in African-Americans than Hispanics or non-Hispanic whites. Ethnicity was also related to CD4 counts in patients on HAART which affect insulin sensitivity. In the occurrence of lipodystrophy due to NRTIs therapy, the increased risk was provoked in White patients (P=0.023).25 This finding also suggested that there are differences in severity of metabolic variation among PLHIV based on ethnicity. In patients on PI regimens, the risk of dyslipidemia can be predicted by ethnicity.26 African-Americans and Hispanics had higher levels of triglycerides than White HIV-infected patients on exposure to non-ritonavir PI regimens. However, on exposure to ritonavir regimens, all groups tended to increase in triglycerides.

Metabolic Complications Associated with ART in HIV-Infected Patients

The metabolic outcomes in HIV-infected patients receiving different classes of antiretroviral drugs in previous publications are summarized in Table 1. Questionnaires, abnormalities reports, physical measurements, and clinical assessments including evaluations of laboratory parameters such as CD4, HIV-1 RNA, lipid profiles, glucose, and serum lactate were applied in these studies.

Table 1 The Clinical Studies Reported Metabolic Effects of Antiretroviral Therapy7,11,2735,37,3949,51,52

NRTI-Associated Metabolic Complications

Several studies have reported the lipodystrophy occurrence in NRTI-treated HIV-infected patients.11,2832 Zidovudine and stavudine can cause lipodystrophy with different incidence.28 Patients in the stavudine arm had more frequent symptoms of peripheral fat depletion compared to those in the zidovudine therapy. The fat depletion was presented by atrophy on the face, lower limb, buttock, and venomegaly (P=0.011, 0.006, 0.009, and 0.001, respectively). There were elevations in cholesterol, triglyceride, and glucose levels but they were not significantly different between the two groups. Univariate analysis showed that older patients and female genders had increased risk of central fat accumulation. Consistently, Menezes et al11 reported the metabolic toxicity of stavudine during a median follow-up of 19 months in their prospective cohort study. The prevalence of abnormalities was 17.1% peripheral neuropathy, 5.7% symptomatic hyperlactatemia, 2.5% lactic acidosis, and 7.3% lipoatrophy in the study group on stavudine. They examined that patients with a body mass index (BMI) of 25–30 kg/m2 or >30 kg/m2 had higher risks of hyperlactatemia and lactic acidosis. Moreover, females had higher metabolic toxicities such as diabetes and dyslipidemia. The findings concur with the previous study.28 In contrast, male gender was associated with lipodystrophy in the study by Alikhani et al.29 The prevalence of lipodystrophy in this study was 47%, with a mean trunk/limb fat ratio of 1.87. Patients with lipodystrophy were at risk of development of diabetes and dyslipidemia following the elevations of blood glucose and triglyceride levels (P=0.002 and P<0.001, respectively). Additional factors such as duration of HIV infection, year of exposure to zidovudine or stavudine, and older age were also associated with lipodystrophy.

In a clinical trial of Gallant et al,30 where the safety and efficacy of tenofovir DF versus stavudine in ART-naïve patients were assessed, the overall incidence of laboratory abnormalities were similar. Lower mean elevations in triglycerides (+0.01 mmol/L), total cholesterol levels (+0.78 mmol/L), LDL-C levels (+0.36 mmol/L), and higher increase in HDL-C levels (+0.23 mmol/L) were found in the tenofovir DF group. All the patients with lactic acidosis were from the stavudine group. In addition, patients with abnormal AST and ALT levels were more frequent in the stavudine group. Tenofovir DF receiving patients had higher loss of limb fat compared to the stavudine arm at week 96 (7.9 kg vs 5 kg), and at week 144 (8.6 kg vs 4.5 kg) (P<0.001). Surprisingly, the proportions of bone fractures showed no difference between two groups, whereas researcher-reported lipodystrophy was more common in stavudine receiving patients (P<0.001). This finding was supported by the study of Haubrich et al.31 They reported that lipoatrophy was most frequent with stavudine-containing regimens. Median cholesterol levels were also elevated in the stavudine group (41 mg/dL, P=0.02). Regimens containing didanosine/stavudine were associated with greater limb fat loss.33 In an open-label randomized study by Gallant et al,32 limb fat mass measured by DEXA showed that patients receiving tenofovir DF/emtricitabine had higher limb fat loss compared to the zidovudine/lamivudine group (P=0.03). The fasting lipid profile showed that the zidovudine/lamivudine group had a greater mean increase in total cholesterol (0.91 mmol/L vs 0.54 mmol/L, P<0.001) and LDL-C (0.52 mmol/L vs 0.34 mmol/L, P=0.01) compared to the tenofovir DF/emtricitabine group. Patients in the zidovudine/lamivudine group experienced more frequent adverse events which resulted in discontinuation of treatment. The study found that the most frequent cause of discontinuation of zidovudine/lamivudine containing regimen was anemia.

Sax et al35 examined treatment-naïve patients who were randomly assigned to oral tablets containing 150 mg elvitegravir, 150 mg cobicistat, 200 mg emtricitabine in combination with either 10 mg tenofovir AF or 300 mg tenofovir DF. This study reported that the tenofovir DF group had significantly greater decreases in BMD at hip (mean % change −2.95 vs −0.66, P<0.0001) and at spine (−2.86 vs−1.3, P<0.001) compared to the tenofovir AF group at week 48. A decrease in BMD induced by tenofovir DF at week 48 was supported by the clinical trial of Chen et al.36

NNRTI-Associated Metabolic Complications

The use of NNRTIs showed alterations in metabolic parameters although the changes were not as much pronounced as other classes of drugs.8 In contrast to other classes, NNRTIs cause only a modest increase in lipid profiles. Martínez et al37 conducted a multicenter, randomized, open-label clinical trial in 15 centers in Spain. The efficacy of NNRTIs (nevirapine or efavirenz) or abacavir as a substitute for a protease inhibitor in HIV-infected patients was studied at baseline, 1-month, 3-month, and every 3-month period until 12-month follow-up. The proportion of patients with elevated triglycerides (>4.5 mmol/L) and cholesterol values (>6.2 mmol/L) was higher in the NNRTI group. The median fasting plasma glucose levels were also significantly higher in patients receiving efavirenz at every follow-up time (P<0.01). The efficacy and safety analysis of a randomized trial found that efavirenz had higher grade 2–4 lipid disorders than rilpivirine.38 During the week 96 treatment period, efavirenz had higher elevations of LDL, AST, and ALT levels than rilpivirine. Lazzarin et al39 performed a randomized, double-blind, placebo-controlled, Phase III trial in HIV-infected patients who failed by ART with resistance to NNRTIs and PIs. The patients were randomly assigned to receive either etravirine 200 mg or placebo, twice daily in combination with darunavir/ritonavir, NRTIs, and optional enfuvirtide. During a 24-week study period, grade 3 or 4 changes in laboratory lipid parameters (hyperglycemia, hypercholesterolemia, hypertriglyceridemia, increased AST and ALT) were observed which were generally similar between treatment arms. Fewer lipid elevations in etravirine were reported in a trial study by Fätkenheuer et al.40 Of the 157 ART-naïve patients, 79 patients were assigned to etravirine 400 mg once daily with efavirenz placebo, and 78 patients were assigned to efavirenz 600 mg once daily with etravirine placebo, plus two NRTIs in both regimens. NRTIs included in this study were abacavir/lamivudine, zidovudine/lamivudine, or tenofovir/emtricitabine. At week 48, the efavirenz group showed significant elevations of lipids compared with the etravirine group: total cholesterol (+0.61 mmol/L, P<0.0001), HDL-C (+0.15 mmol/L, P=0.004), LDL-C (+0.35 mmol/L, P=0.005), and triglycerides (+0.33 mmol/L, P=0.03). Patients receiving abacavir/lamivudine in both groups showed a high increase in total cholesterol levels compared to those taking tenofovir/emtricitabine (P=0.005). The etravirine group showed fewer grade 3 elevations of lipids than the efavirenz group. The study suggested larger, long-term trials for the assessment of safety and efficacy of etravirine 400 mg once daily. In addition to an increase in lipid parameters, NNRTIs such as nevirapine, efavirenz, and delavirdine also increased insulin resistance.41 There was a significant elevation of mean insulin levels over the median study period of 5 years (P<0.01). This supported the close monitoring of risk of diabetes and glucose levels. Moreover, efavirenz in combination with NRTI (tenofovir DF) has been examined to reduce BMD in HIV-infected ART-naïve patients.36

PI-Associated Metabolic Complications

The use of PIs was associated with a significant increase in lipid levels leading to hyperlipidemia. The lipid abnormalities due to PIs are severe and more common than other classes of drugs,8 some of which resulted in discontinuation of the treatment.42 In a clinical trial of Clumeck et al,43 patients receiving PI-based therapy for at least 6 months were examined. The only recommended PI during randomization was ritonavir/saquinavir. Increased lipid profiles (triglyceride >2.3 mmol/L and cholesterol >5.2 mmol/L) were found in these patients. Some patients discontinued treatment because of lipodystrophy and significant elevation in triglyceride and cholesterol levels. Similarly, abnormally higher triglycerides and total cholesterol levels due to PIs were also observed in a study of Walmsley et al.7 Patients on lopinavir/ritonavir regimens had higher abnormalities in laboratory profiles than those on nelfinavir regimens. During the 48-week period, the mean elevations in total cholesterol were 1.37 mmol/L vs 1.24 mmol/L, and in triglycerides were 1.4 mmol/L vs 0.5 mmol/L in these groups. Lipodystrophy or lipoatrophy were reported but the prevalence was not much pronounced. The study suggested a longer follow-up time over 48 weeks to determine the clinical significance of elevations in lipids. When compared to efavirenz (NNRTI), nelfinavir was found with greater loss of limb fat in a randomized study by Dube et al33 (P<0.003). Lipid profile measurements also showed high elevations in the nelfinavir group over 64 weeks.

Ortiz et al44 randomly subjected ART-naïve patients to receive either darunavir/ritonavir or lopinavir/ritonavir, in combination with tenofovir/emtricitabine. Grade 2–4 elevations in triglycerides and cholesterol were found more frequently in the lopinavir group than the darunavir group (11% vs 3%, and 23% vs 13%, respectively) at week 48. This finding was supported by the week 48 phase III trial of Madruga et al.45 The grade 2–4 elevations of AST and ALT were also observed in addition to the same incidence of hyperglycemia in two groups. Hypertriglyceridemia and hypercholesterolemia were the adverse events leading to permanent treatment discontinuation found in the lopinavir/ritonavir group. In a randomized trial of Molina et al,46 ART-naïve patients were subjected to receive atazanavir/ritonavir or lopinavir/ritonavir, each in combination with tenofovir/emtricitabine. Bilirubin-associated adverse events were higher in the atazanavir group. Some changes in lipid parameters (increased AST, ALT, cholesterol, and triglycerides) were significantly higher in the lopinavir group (P<0.0001) at week 96. Additionally, the patients in the lopinavir group had higher gastrointestinal intolerability rates, in which 1.6% of patients discontinued the treatment due to diarrhea. This study recommended atazanavir/ritonavir as the first-line treatment because it has a better lipid profile and high efficiency in treatment-naïve patients with high plasma HIV viral loads and low CD4 counts at baseline.

Arathoon et al42 conducted a 192-week, randomized, Phase III, open-label trial to compare the efficacy and safety of darunavir/ritonavir and lopinavir/ritonavir, each in combination with fixed-dose tenofovir/emtricitabine in ART-naïve patients. Metabolic-related adverse effects were observed at week 96. Two patients on lopinavir-based regimens discontinued treatment due to hypercholesterolemia and hypertriglyceridemia. The occurrence of metabolic and nutritional disorders was higher in the lopinavir group. The most frequent lipid-related adverse events were hypertriglyceridemia (2% vs 5.8%), hypercholesterolemia (1.5% vs 4%), hyperlipidemia (0.6% vs 3.2%), and increased LDL-C (2.3% vs 1.2%) in the darunavir and lopinavir groups. Some anthropometric changes such as body weight or mid-waist/hip ratio were observed in week 96, but these were considered clinically irrelevant. PI containing regimens increased insulin resistance prevalence and glucose level in HIV-infected patients in previous randomized studies.41,47 AIDS Clinical Trials Group (ACTG) A5260s, a trial study, examined the changes in insulin resistance after treatment with raltegravir, darunavir/ritonavir, or atazanavir/ritonavir in ART-naïve HIV-infected patients.47 At the follow-up period of week 96, there were increases in homeostatic model assessment of insulin resistance (HOMA-IR) in all study groups. It was also observed that such an increase in insulin resistance was associated with changes in BMI, interleukin-6, and high-sensitivity C-reactive protein at week 48 and 96. Bone abnormalities were also pronounced in patients receiving PIs. In a randomized study of ART-naïve patients, BMD changes were monitored at baseline and week 48 by DEXA.48 The randomized regimens were PIs plus NNRTI and PIs plus NRTI or NNRTI plus NRTIs. At week 48, there were significant decreases in BMD at lumbar spine and hip (mean −4.1 and −2.8, P≤0.001, respectively). BMD at lumbar spine was decreased more prominently in PI receiving patients compared to regimens without PIs.

Other ART-Associated Metabolic Complications

Lipodystrophy was associated with the cumulative use of antiretroviral drugs such as raltegravir.29 Patients with fat abnormalities were more likely to develop diabetes and dyslipidemia. However, raltegravir showed better safety and clinical profiles in a trial reported by Lennox et al.49 ART-naïve patients were randomized to receive either 400 mg raltegravir twice daily or 600 mg efavirenz once daily, both in combination with tenofovir and emtricitabine. Fasting LDL-C >4.92 mmol/L and triglycerides >3.48 mmol/L were found more frequently in the efavirenz group. Mean LDL-C and triglyceride levels were higher in the efavirenz than the raltegravir group after week 48.

Switching Strategies

Several studies have recommended to switch or simplify the regimens in order to reverse the treatment-related abnormalities such as metabolic disorders, fat redistribution, or other manageable side-effects.30,34,37,43,5053 Clumeck et al43 demonstrated that switching from a PI-based regimen (ritonavir/saquinavir) to abacavir-based regimen provides prolonged viral suppression and significant improvements in lipid abnormalities. Switching from a stable PI regimen to atazanavir/ritonavir was found to improve insulin resistance and dyslipidemia in HIV-infected men.54 After 3 months, there were increases in glucose disposal rate (P=0.008), and a significant decrease in triglyceride levels also (P=0.023). Carr et al53 observed that switching from nucleoside analogs such as zidovudine or stavudine to abacavir resulted in improvement in HIV-associated lipoatrophy. There was a significant increase in limb fat mass on DEXA in abacavir receiving patients (0.39 kg) compared to the zidovudine or stavudine arm (0.08 kg), 95% CI= 0.06–0.57, P=0.02. Nonetheless, a larger study population and longer follow-up period were recommended to decide whether lipoatrophy can be improved clinically or resolved. ACTG 5125s studied the effects of PIs and NRTIs on the fat distribution, BMD, and metabolic parameters in patients with advance HIV (CD4 ≤200 cells/mm3 or viral load ≥80,000 copies/mL) and undetectable viral load.51 Patients were randomized to switch their initial regimen to open label lopinavir/ritonavir plus efavirenz or two NRTIs plus efavirenz. During a median observation period of 102 weeks, the PIs group had a greater increase in limb fat, meanwhile the NRTIs arm had loss of limb fat. However, at week 48, the PIs group had greater increases in triglycerides (85 vs 11 mg/dL, P=0.01), total cholesterol (19 vs −7 mg/dL, P=0.009), and non HDL-C (13 vs −10 mg/dL) compared to NRTIs group. It was also found that, at week 48, treatment changes in body mass were directly related to changes in fasting insulin concentration. On the other hand, changes in total cholesterol and triglyceride levels were inversely related to baseline BMI and lean mass. After a median follow-up of 2.1 years, the PIs group showed a higher treatment discontinuation rate than the NRTIs group (40 vs 19, P<0.001, respectively). This rate was mainly forced by increased triglyceride levels. Although the lopinavir/ritonavir arm improved limb fat mass, their significant increase in lipid parameters suggested careful therapeutic management.

Martin et al34 conducted a randomized, open-label, 96-week trial in HIV-infected patients by switching from the existing NRTIs to either tenofovir/emtricitabine or abacavir/lamivudine. The baseline NRTIs included abacavir without tenofovir, tenofovir without abacavir, lamivudine, emtricitabine, zidovudine, didanosine, and stavudine. In the overall 357 patients, the abacavir group had more common lipid-related events compared to the tenofovir group (P=0.003). However, bone events such as osteopenia or osteoporosis were more common in the tenofovir group (P=0.032). In addition, the tenofovir regimen was not only associated with rapid decreases in body weight and lean body mass but also led to significant loss of hip and spine BMD. In a trial of Perez-Molina et al,52 ART-experienced patients were randomized into either dual therapy with atazanavir/ritonavir plus lamivudine or triple therapy with atazanavir/ritonavir plus two NRTIs (tenofovir/emtricitabine or abacavir/lamivudine or zidovudine/lamivudine). From baseline to 96 weeks, analysis showed that two patients per group discontinued ART because of hyperbilirubinemia but there was no new discontinuation beyond 48-week duration. Increased total cholesterol and triglyceride levels were observed in the dual therapy arm. A decrease in BMD was also observed but there were no significant different changes between groups. Mills et al27 conducted a multicenter, randomized, open-label, phase III clinical trial in treatment experienced HIV-infected adults. Participants were randomly assigned at a 2:1 ratio into either switching into single tablet tenofovir AF (n=959) 10 mg plus elvitegravir 150 mg, cobicistat 150 mg and emtricitabine 200 mg or continuing previous tenofovir DF regimens (n=477). At week 48, mean BMD at the hip and spine remained stable or decreased in the tenofovir DF group, whereas there were increases in the tenofovir AF group (P<0.0001). Patients on previous tenofovir DF regimens tended to improve BMD after switching to tenofovir AF regimens. Consistent with this finding, Maggiolo et al55 observed the significant improvement in BMD after switching from tenofovir DF to tenofovir AF containing regimens in HIV-infected patients aged 60 years or older.

According to previous studies, PLHIV receiving to ART exhibited the changes in subcutaneous adipose tissues, commonly lipodystrophy and abnormal lipid profiles. Lists of antiretroviral drugs in each class associated with lipodystrophy are summarized in Table 2. Although lipodystrophy in patients receiving abacavir, nevirapine, or efavirenz had been decreased, newer cases were arising. Therefore, switching from PIs to these drugs in order to reduce tendency for discontinuation due to lipid abnormalities is still not an applicable strategy.37 Individual drugs with their effects on changes in metabolic markers are summarized in Table 3. Types of ART with different metabolic profiles can be differentiated. Although some known drugs do not have optimal safety and tolerability profiles, their side-effects do not limit the efficacy in clinical settings. NRTIs (abacavir, lamivudine, zidovudine, emtricitabine, tenofovir), NNRTIs (nevirapine, efavirenz), and PIs (lopinavir, ritonavir, atazanavir) are commonly recommended in the first-line HAART regimens.5 Each antiretroviral drug with their common metabolic complications examined in previous studies are summarized in Table 4.

Table 2 Antiretroviral Therapy Associated with Lipodystrophy7,11,2833,37,43

Table 3 Summary of the Effects of Antiretroviral Therapy on Metabolic Parameters7,11,27,29,30,3235,37,39,40,4246,49,51,52

Table 4 The Common Metabolic Effects of Antiretroviral Therapy7,11,2735,3740,4246,4853,56

Recommendations

The prevalence or severity of metabolic abnormalities are varied among individuals or classes of drugs; some of which may be developed over time. Therefore, a longer study period is required to better recognize long-term abnormalities related to different classes of drugs. One more point to be noted is that the nutritional status of HIV-infected patients is affected by several metabolic complications due to both HIV infection and ART. Therefore, it is required to manage ART-associated metabolic abnormalities because HIV-infected patients require life-long therapy. Otherwise, patients with poor nutritional status will be exacerbated. Lifestyle management and pharmaceutical interventions are necessary for patients on long-term therapy. Nutritional counseling and intervention may be required especially in the patients who are at risk of malnutrition. In advance, dietary consideration should be provided to optimize treatment associated complications such as dyslipidemia, glucose abnormalities, and limb fat loss.57 Individualization is also critical in relation to the treatment of metabolic abnormalities in combination with giving interventions according to each patient’s needs.

Conclusion

Some drugs with high metabolic toxicity are still being used in resource-limited countries due to cost effectiveness. Thus, appropriate pharmacologic development and protocols are necessary to minimize toxicity. Nutritional counseling and management in PLHIV have been strongly recommended. The occurrence of ART-associated metabolic complication may be applied as predicting factors for either modification of therapy in PLHIV or finding new regimens with fewer side-effects. Active pharmacovigilance programs are recommended in HIV care programs to properly identify ART-related adverse events so that serious complications can be improved.56 Moreover, strategies which differentiate PLHIV at high risk of metabolic complications may be helpful in exploring the appropriate regimens. At the beginning of ART in PLHIV, it has been highly recommended to perform genotyping.21 Genetic polymorphism and ethnicities in some way may be related to the effects of ART. The better choice of ART regimen based on genetic analysis may help reducing the potential risk of metabolic abnormalities. Because of the multifactorial nature of metabolic complications, it is advisable to adjust the benefits and risks, including virologic success and potential toxicities of new regimens when switching or adding to the existing regimens. Prospective studies evolving the safety and efficacy of individual antiretroviral therapy or combination regimens in a large population are warranted. To get a sustained clinical benefit of therapy, pharmacologic improvements in administration of regimens have been recommended. The particular adverse effects of each drug or regimen should be clarified to achieve the effectiveness of therapy and improve the tolerability of treatment in HIV-infected patients.

Acknowledgment

This review was supported by Thailand International Cooperation Agency Scholarship.

Disclosure

The authors report no conflicts of interest in this work.

References

1. UNAIDS. Global HIV & AIDS statistics_2020 fact sheet. 2020. Available from: https://www.unaids.org/en/resources/fact-sheet. Accessed July 24, 2020.

2. 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;15(7):810–818. doi:10.1016/S1473-3099(15)00056-0

3. Poorolajal J, Hooshmand E, Mahjub H, Esmailnasab N, Jenabi E. Survival rate of AIDS disease and mortality in HIV-infected patients: a meta-analysis. Public Health. 2016;139:3–12. doi:10.1016/j.puhe.2016.05.004

4. Jain RG, Furfine ES, Pedneault L, White AJ, Lenhard JM. Metabolic complications associated with antiretroviral therapy. Antiviral Res. 2001;51(3):151–177. doi:10.1016/S0166-3542(01)00148-6

5. World Health Organization. Update of recommendations on first-and second-line antiretroviral regimens. 2019. Available from: https://apps.who.int/iris/bitstream/handle/10665/325892/WHO-CDS-HIV-19.15-eng.pdf?ua=1. Accessed June 13, 2019.

6. Brown TT, Qaqish RB. Antiretroviral therapy and the prevalence of osteopenia and osteoporosis: a meta-analytic review. AIDS. 2006;20(17):2165–2174. doi:10.1097/QAD.0b013e32801022eb

7. Walmsley S, Bernstein B, King M, et al. Lopinavir–ritonavir versus nelfinavir for the initial treatment of HIV infection. N Engl J Med. 2002;346(26):2039–2046. doi:10.1056/NEJMoa012354

8. Dubé MP, Stein JH, Aberg JA, et al. Guidelines for the evaluation and management of dyslipidemia in human immunodeficiency virus (HIV)-infected adults receiving antiretroviral therapy: recommendations of the HIV Medicine Association of the Infectious Disease Society of America and the Adult AIDS Clinical Trials Group. Clin Infect Dis. 2003;37(5):613–627. doi:10.1086/378131

9. Guzman N, Vijayan V. HIV-Associated Lipodystrophy. StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2019. Available from: https://www.ncbi.nlm.nih.gov/books/NBK493183/s. Accessed September 24, 2020.

10. Fiorenza CG, Chou SH, Mantzoros CS. Lipodystrophy: pathophysiology and advances in treatment. Nat Rev Endocrinol. 2011;7(3):137–150. doi:10.1038/nrendo.2010.199

11. Menezes CN, Maskew M, Sanne I, Crowther NJ, Raal FJ. A longitudinal study of stavudine-associated toxicities in a large cohort of South African HIV infected subjects. BMC Infect Dis. 2011;11(1):244. doi:10.1186/1471-2334-11-244

12. Kim HR, Han MA. Association between serum liver enzymes and metabolic syndrome in Korean adults. Int J Environ Res Public Health. 2018;15(8):1658. doi:10.3390/ijerph15081658

13. Conradie M, van de Vyver M, Andrag E, Conradie M, Ferris W. A direct comparison of the effects of the antiretroviral drugs stavudine, tenofovir and the combination lopinavir/ritonavir on bone metabolism in a rat model. Calcif Tissue Int. 2017;101(4):422–432. doi:10.1007/s00223-017-0290-3

14. Pan G, Wu X, McKenna MA, Feng X, Nagy TR, McDonald JM. AZT enhances osteoclastogenesis and bone loss. AIDS Res Hum Retroviruses. 2004;20(6):608–620. doi:10.1089/0889222041217482

15. Pistell PJ, Gupta S, Knight AG, et al. Metabolic and neurologic consequences of chronic lopinavir/ritonavir administration to C57BL/6 mice. Antiviral Res. 2010;88(3):334–342. doi:10.1016/j.antiviral.2010.10.006

16. Tsai F-J, Ho M-W, Lai C-H, et al. Evaluation of oral antiretroviral drugs in mice with metabolic and neurologic complications. Front Pharmacol. 2018;9:1004. doi:10.3389/fphar.2018.01004

17. Sembongi H, Miranda M, Han G-S, et al. Distinct roles of the phosphatidate phosphatases lipin 1 and 2 during adipogenesis and lipid droplet biogenesis in 3T3-L1 cells. J Biol Chem. 2013;288(48):34502–34513. doi:10.1074/jbc.M113.488445

18. Bresciani E, Saletti C, Squillace N, et al. miRNA-218 targets Lipin-1 and glucose transporter type 4 genes in 3T3-L1 cells treated with lopinavir/itonavir. Front Pharmacol. 2019;10(461). doi:10.3389/fphar.2019.00461

19. Caron M, Auclair M, Lagathu C, et al. The HIV-1 nucleoside reverse transcriptase inhibitors stavudine and zidovudine alter adipocyte functions in vitro. AIDS. 2004;18(16):2127–2136. doi:10.1097/00002030-200411050-00004

20. Nolan D, Hammond E, Martin A, et al. Mitochondrial DNA depletion and morphologic changes in adipocytes associated with nucleoside reverse transcriptase inhibitor therapy. AIDS. 2003;17(9):1329–1338. doi:10.1097/00002030-200306130-00007

21. Tarr PE, Taffé P, Bleiber G, et al. Modeling the influence of APOC3, APOE and TNF polymorphisms on the risk of antiretroviral therapy–associated lipid disorders. J Infect Dis. 2005;191(9):1419–1426. doi:10.1086/430627

22. Fauvel J, Bonnet E, Ruidavets J-B, et al. An interaction between apo C-III variants and protease inhibitors contributes to high triglyceride/low HDL levels in treated HIV patients. AIDS. 2001;15(18):2397–2406. doi:10.1097/00002030-200112070-00007

23. Curtis JR, Smith B, Weaver M, et al. Ethnic variations in the prevalence of metabolic bone disease among HIV-positive patients with lipodystrophy. AIDS Res Hum Retroviruses. 2006;22(2):125–131. doi:10.1089/aid.2006.22.125

24. Misra R, Chandra P, Riechman SE, et al. Relationship of ethnicity and CD4 count with glucose metabolism among HIV patients on Highly-Active Antiretroviral Therapy (HAART). BMC Endocr Disord. 2013;13(1):13. doi:10.1186/1472-6823-13-13

25. Mallal SA, John M, Moore CB, James IR, McKinnon EJ. Contribution of nucleoside analogue reverse transcriptase inhibitors to subcutaneous fat wasting in patients with HIV infection. AIDS. 2000;14(10):1309–1316. doi:10.1097/00002030-200007070-00002

26. Foulkes AS, Wohl DA, Frank I, et al. Associations among race/ethnicity, apoC-III genotypes, and lipids in HIV-1-infected individuals on antiretroviral therapy. PLoS Med. 2006;3(3):337–347. doi:10.1371/journal.pmed.0030052

27. Mills A, Arribas JR, Andrade-Villanueva J, et al. Switching from tenofovir disoproxil fumarate to tenofovir alafenamide in antiretroviral regimens for virologically suppressed adults with HIV-1 infection: a randomised, active-controlled, multicentre, open-label, phase 3, non-inferiority study. Lancet Infect Dis. 2016;16(1):43–52. doi:10.1016/S1473-3099(15)00348-5

28. Joly V, Flandre P, Meiffredy V, et al. Increased risk of lipoatrophy under stavudine in HIV-1-infected patients: results of a substudy from a comparative trial. AIDS. 2002;16(18):2447–2454. doi:10.1097/00002030-200212060-00010

29. Alikhani A, Morin H, Matte S, Alikhani P, Tremblay C, Durand M. Association between lipodystrophy and length of exposure to ARTs in adult HIV-1 infected patients in Montreal. BMC Infect Dis. 2019;19(1):820. doi:10.1186/s12879-019-4446-9

30. Gallant JE, Staszewski S, Pozniak AL, et al. Efficacy and safety of tenofovir DF vs stavudine in combination therapy in antiretroviral-naive patients: a 3-year randomized trial. JAMA. 2004;292(2):191–201. doi:10.1001/jama.292.2.191

31. Haubrich RH, Riddler SA, DiRienzo AG, et al. Metabolic outcomes in a randomized trial of nucleoside, nonnucleoside and protease inhibitor-sparing regimens for initial HIV treatment. AIDS. 2009;23(9):1109. doi:10.1097/QAD.0b013e32832b4377

32. Gallant JE, DeJesus E, Arribas JR, et al. Tenofovir DF, emtricitabine, and efavirenz vs. zidovudine, lamivudine, and efavirenz for HIV. N Engl J Med. 2006;354(3):251–260. doi:10.1056/NEJMoa051871

33. Dube MP, Parker RA, Tebas P, et al. Glucose metabolism, lipid, and body fat changes in antiretroviral-naive subjects randomized to nelfinavir or efavirenz plus dual nucleosides. AIDS. 2005;19(16):1807–1818. doi:10.1097/01.aids.0000183629.20041.bb

34. Martin A, Bloch M, Amin J, et al. Simplification of antiretroviral therapy with tenofovir-emtricitabine or abacavir-lamivudine: a randomized, 96-week trial. Clin Infect Dis. 2009;49(10):1591–1601. doi:10.1086/644769

35. Sax PE, Wohl D, Yin MT, et al. Tenofovir alafenamide versus tenofovir disoproxil fumarate, coformulated with elvitegravir, cobicistat, and emtricitabine, for initial treatment of HIV-1 infection: two randomised, double-blind, Phase 3, non-inferiority trials. Lancet. 2015;385(9987):2606–2615. doi:10.1016/S0140-6736(15)60616-X

36. Chen J, Chen R, Shen Y, et al. Efficacy and safety of lower dose tenofovir disoproxil fumarate and efavirenz versus standard dose in HIV-infected, antiretroviral-naive adults: a multicentre, randomized, noninferiority trial. Emerg Microbes Infect. 2020;9(s1):843–850. doi:10.1080/22221751.2020.1752609

37. Martínez E, Arnaiz JA, Podzamczer D, et al. Substitution of nevirapine, efavirenz, or abacavir for protease inhibitors in patients with human immunodeficiency virus infection. N Engl J Med. 2003;349(11):1036–1046. doi:10.1056/NEJMoa021589

38. Cohen CJ, Molina J-M, Cassetti I, et al. Week 96 efficacy and safety of rilpivirine in treatment-naive, HIV-1 patients in two Phase III randomized trials. AIDS. 2013;27(6):939–950. doi:10.1097/QAD.0b013e32835cee6e

39. Lazzarin A, Campbell T, Clotet B, et al. Efficacy and safety of TMC125 (etravirine) in treatment-experienced HIV-1-infected patients in DUET-2: 24-week results from a randomised, double-blind, placebo-controlled trial. Lancet. 2007;370(9581):39–48. doi:10.1016/S0140-6736(07)61048-4

40. Fätkenheuer G, Duvivier C, Rieger A, et al. Lipid profiles for etravirine versus efavirenz in treatment-naive patients in the randomized, double-blind SENSE trial. J Antimicrob Chemother. 2012;67(3):685–690. doi:10.1093/jac/dkr533

41. Shlay JC, Bartsch G, Peng G, et al. Long-term body composition and metabolic changes in antiretroviral naive persons randomized to protease inhibitor-, nonnucleoside reverse transcriptase inhibitor-, or protease inhibitor plus nonnucleoside reverse transcriptase inhibitor-based strategy. J Acquir Immune Defic Syndr. 2007;44(5):506–517. doi:10.1097/QAI.0b013e31804216cf

42. Arathoon E, Schneider S, Baraldi E, et al. Effects of once-daily darunavir/ritonavir versus lopinavir/ritonavir on metabolic parameters in treatment-naïve HIV-1-infected patients at week 96: ARTEMIS. Int J STD AIDS. 2013;24(1). doi:10.1258/ijsa.2012.012120

43. Clumeck N, Goebel F, Rozenbaum W, et al. Simplification with abacavir-based triple nucleoside therapy versus continued protease inhibitor-based highly active antiretroviral therapy in HIV-1-infected patients with undetectable plasma HIV-1 RNA. AIDS. 2001;15(12):1517–1526. doi:10.1097/00002030-200108170-00009

44. Ortiz R, DeJesus E, Khanlou H, et al. Efficacy and safety of once-daily darunavir/ritonavir versus lopinavir/ritonavir in treatment-naive HIV-1-infected patients at week 48. AIDS. 2008;22(12):1389–1397. doi:10.1097/QAD.0b013e32830285fb

45. Madruga JV, Berger D, McMurchie M, et al. Efficacy and safety of darunavir-ritonavir compared with that of lopinavir-ritonavir at 48 weeks in treatment-experienced, HIV-infected patients in TITAN: a randomised controlled phase III trial. Lancet. 2007;370(9581):49–58. doi:10.1016/S0140-6736(07)61049-6

46. Molina J-M, Andrade-Villanueva J, Echevarria J, et al. Once-daily atazanavir/ritonavir compared with twice-daily lopinavir/ritonavir, each in combination with tenofovir and emtricitabine, for management of antiretroviral-naive HIV-1-infected patients: 96-week efficacy and safety results of the CASTLE study. J Acquir Immune Defic Syndr. 2010;53(3):323–332. doi:10.1097/QAI.0b013e3181c990bf

47. Dirajlal-Fargo S, Moser C, Brown TT, et al. Changes in insulin resistance after initiation of raltegravir or protease inhibitors with tenofovir-emtricitabine: AIDS Clinical Trials Group A5260s. Open Forum Infect Dis. 2016;3(3):ofw174. doi:10.1093/ofid/ofw174

48. Duvivier C, Kolta S, Assoumou L, et al. Greater decrease in bone mineral density with protease inhibitor regimens compared with nonnucleoside reverse transcriptase inhibitor regimens in HIV-1 infected naive patients. AIDS. 2009;23(7):817–824. doi:10.1097/QAD.0b013e328328f789

49. Lennox JL, DeJesus E, Lazzarin A, et al. Safety and efficacy of raltegravir-based versus efavirenz-based combination therapy in treatment-naive patients with HIV-1 infection: a multicentre, double-blind randomised controlled trial. Lancet. 2009;374(9692):796–806. doi:10.1016/S0140-6736(09)60918-1

50. Elzi L, Erb S, Furrer H, et al. Adverse events of raltegravir and dolutegravir. AIDS. 2017;31(13):1853. doi:10.1097/QAD.0000000000001590

51. Tebas P, Zhang J, Yarasheski K, et al. Switching to a protease inhibitor-containing, nucleoside-sparing regimen (lopinavir/ritonavir plus efavirenz) increases limb fat but raises serum lipid levels: results of a prospective randomized trial (AIDS clinical trial group 5125s). J Acquir Immune Defic Syndr. 2007;45(2):193. doi:10.1097/QAI.0b013e318042e204

52. Perez-Molina J, Rubio R, Rivero A, et al. Simplification to dual therapy (atazanavir/ritonavir+ lamivudine) versus standard triple therapy [atazanavir/ritonavir+ two nucleos (t) ides] in virologically stable patients on antiretroviral therapy: 96 week results from an open-label, non-inferiority, randomized clinical trial (SALT study). J Antimicrob Chemother. 2016;72(1):246–253. doi:10.1093/jac/dkw379

53. Carr A, Workman C, Smith DE, et al. Abacavir substitution for nucleoside analogs in patients with HIV lipoatrophy: a randomized trial. JAMA. 2002;288(2):207–215. doi:10.1001/jama.288.2.207

54. Busti AJ, Bedimo R, Margolis DM, Hardin DS. Improvement in insulin sensitivity and dyslipidemia in protease inhibitor-treated adult male patients after switch to atazanavir/ritonavir. J Investig Med. 2008;56(2):539–544. doi:10.2310/JIM.0b013e3181641b26

55. Maggiolo F, Rizzardini G, Raffi F, et al. Bone mineral density in virologically suppressed people aged 60 years or older with HIV-1 switching from a regimen containing tenofovir disoproxil fumarate to an elvitegravir, cobicistat, emtricitabine, and tenofovir alafenamide single-tablet regimen: a multicentre, open-label, phase 3b, randomised trial. Lancet HIV. 2019;6(10):e655–e666. doi:10.1016/S2352-3018(19)30195-X

56. Tetteh RA, Nartey ET, Lartey M, et al. Association between the occurrence of adverse drug events and modification of first-line highly active antiretroviral therapy in Ghanaian HIV patients. Drug Saf. 2016;39(11):1139–1149. doi:10.1007/s40264-016-0460-7

57. Clark WA, Cress EM. Nutritional issues and positive living in human immunodeficiency virus/AIDS. Nurs Clin. 2018;53(1):13–24. doi:10.1016/j.cnur.2017.10.002

Creative Commons License © 2020 The Author(s). This work is published and licensed by Dove Medical Press Limited. The full terms of this license are available at https://www.dovepress.com/terms.php and incorporate the Creative Commons Attribution - Non Commercial (unported, v3.0) License. By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted without any further permission from Dove Medical Press Limited, provided the work is properly attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms.