Abstract
Purpose of review:
We review the scope and burden of metabolic syndrome in HIV/HCV co-infected patients, risk factors and potential mechanisms driving the increased cardio-metabolic risk in this population, and discuss relevant clinical considerations for management in the era of highly effective antiretroviral therapy (ART) and curative anti-HCV direct-acting antivirals.
Recent findings:
HIV/HCV co-infected patients are at elevated risk of metabolic syndrome, attributed to (1) patient-specific factors, (2) viral-mediated effects, and (3) ART exposure. Risk factors for cardio-metabolic disorders are common in this population and include poor socioeconomic conditions, substance use, cardiovascular comorbidities, and liver/kidney disease. Chronic HIV/HCV infection induces an inflammatory and immune activated state in the host leading to alterations in glucose and lipid metabolism. Selection of life-saving ART must carefully consider the differential metabolic risk associated with each drug class and agent, such as dyslipidemia, hyperglycemia and insulin resistance, weight gain and hypertension. Emerging evidence supports metabolic derangements in chronic HCV may be improved by viral eradication with direct-acting antivirals, however, additional study in HIV/HCV co-infected patients is needed.
Summary:
Future research programs should aim to better characterize metabolic syndrome in HIV/HCV co-infected patients with the goal of improved screening, treatment and prevention.
Keywords: Human immunodeficiency virus, chronic hepatitis C virus, metabolic syndrome, age-associated comorbidities, antiretroviral therapy, direct-acting antivirals
I. Introduction
Persons with HIV (PWH) with access to care are aging. Those treated with antiretroviral therapy (ART) experience a near-normal life expectancy compared with the general population1. According to the most recent Centers for Disease Control and Prevention surveillance data, persons aged ≥65 years had the largest percentage increase in prevalence of HIV diagnoses from 2012 to 2016 (56%; from 104.4 to 162.6 per 100,000 persons)2. Age-related non-AIDS comorbidities increasingly account for morbidity and mortality in PWH3-5. Most commonly, comorbidities include cardiovascular disease (CVD), metabolic disorders, hepatic and renal disease, and psychiatric illnesses3,6,7.
In the U.S., approximately 25-30% of PWH are co-infected with hepatitis C virus (HCV), accounting for roughly one million individuals8. It is well-established that patients with HIV/HCV co-infection experience an accelerated natural history of liver disease with increased complications and fatality, when compared with HIV or HCV mono-infected patients9-12. There is emerging evidence that HIV/HCV co-infected individuals also suffer from increased cardio-metabolic risk13-16. Chronic HCV infection is associated with a multitude of metabolic complications: dyslipidemia, hepatic steatosis, insulin resistance (IR) and chronic kidney disease (CKD)17,18. Exactly how HCV may exacerbate the risk of cardio-metabolic comorbidities in PWH warrants further characterization. The advent of curative direct-acting antiviral (DAA) therapy has revolutionized the management of HCV infection for both HCV mono- and HIV/HCV co-infected patients19,20; and the metabolic impacts of achieving HCV cure after DAA treatment are under active investigation21.
The National Cholesterol Education Program Adult Treatment Panel III definition of “metabolic syndrome” (alternatively, “insulin resistance syndrome” or “syndrome X”) is the co-occurrence of (1) hyperglycemia, (2) dyslipidemia, (3) hypertension and (4) abdominal obesity22, all of which coalesce to promote the development of type 2 diabetes mellitus (T2DM) and CVD. Metabolic syndrome is uniquely prevalent in aging PWH6,23,24 and in persons with chronic HCV17,25. Accordingly, it is plausible that HIV/HCV co-infection compounds metabolic derangements in these individuals. The development of metabolic syndrome in HIV/HCV co-infection is likely multifactorial, influenced by (1) patient-specific factors, (2) HIV and HCV viral-mediated effects, and (3) ART exposure.
In this article, we review the scope and burden of metabolic syndrome in HIV/HCV co-infected patients, risk factors and potential mechanisms driving the increased cardio-metabolic risk in this population, and discuss relevant clinical considerations for management in the era of highly effective and tolerable ART and curative anti-HCV DAA therapy.
II. The scope and burden of metabolic syndrome in HIV/HCV co-infection
Data on the prevalence and incidence of ‘metabolic syndrome’ by formal definition are limited in HIV/HCV co-infected patients. However, extrapolations can be made from reports of metabolic syndrome in PWH or those with chronic HCV, as well as studies examining the syndrome’s individual diagnostic components or clinical endpoints (i.e. T2DM, CVD) in the context of HIV/HCV co-infection.
The prevalence of baseline metabolic syndrome in 881 PWH initiating ART in the Initio trial was 7.8-8.5%, depending on the criteria used26. The incidence during three years of follow-up was 8 to 12 cases per 100 person-years. Whereas only incident metabolic syndrome was associated with the development of CVD (HR 2.73, 95% CI 1.07-6.96), both baseline and incident metabolic syndrome increased the risk of T2DM (HR 4.34, 95% CI 1.83-10.25; and HR 4.89, 95% CI 2.22-10.78, respectively)26 Data from the Women’s Interagency HIV Study (WIHS) revealed a higher prevalence of metabolic syndrome in HIV-seropositive than HIV-seronegative women (33% vs 22%, respectively; aOR 1.79, 95% CI 1.48-2.16)23. This increased risk was attributed to dyslipidemia rather than higher blood pressure, glucose, or waist circumference. According to the D:A:D multi-cohort study and a recent trends analysis of claims data, the prevalence of metabolic syndrome in PWH is increasing over time3,24, which has significant and complex implications for the care management of this aging population.
A retrospective evaluation of 2003 to 2012 National Health and Nutrition Examination Survey (NHANES) data among patients with HCV found the prevalence of T2DM and metabolic syndrome to be 18% and 35%, respectively18. The association between chronic HCV infection and diabetes risk is unequivocal and interconnected27: HCV results in hyperinsulinemia and IR28, and T2DM accelerates liver fibrosis leading to cirrhosis29. Hepatic steatosis and fibrosis, both implicated in chronic HCV, are associated with metabolic syndrome and likely compound HCV-induced glucose abnormalities, further elevating metabolic risk in this population30. The link between chronic HCV and the metabolic outcome of CVD is less clear, though the evidence overall suggests an association with higher rates of cardiovascular events and death31,32.
In a cross-sectional analysis of the ICONA Study, HIV/HCV co-infected patients with HCV viremia had a three-fold higher risk of diabetes than PWH33. In this study, the following metabolic comorbidities were independently associated with diabetes in the setting of co-infection: body mass index (BMI) ≥30 kg/m2, hypertension, hypercholesteremia and hypertriglyceridemia33. Older age, HIV viremia and degree of liver fibrosis also increased diabetes risk in HIV/HCV co-infected patients. In terms of HCV modifying the cardiovascular risk in PWH, studies have yielded conflicting results, especially when controlling for potential confounders15,16,34,35. Acute myocardial infarction (AMI) and cerebrovascular event incidences were higher among 6,136 HIV/HCV co-infected veterans (4.2 and 12.5 events per 1000 person-years, respectively) compared with those HIV mono-infected (3.4 and 11.1 events per 1000 person-years, respectively)35. However, the greater risk of AMI in co-infected veterans was not statistically significant after adjusting for age, hypertension, T2DM and duration of ART.
III. Risk factors associated with metabolic syndrome in HIV/HCV co-infection
Metabolic syndrome in HIV/HCV co-infected patients arises from the interplay of traditional and nontraditional risk factors that are overrepresented compared with the general population: poor socioeconomics, substance use, cardiovascular comorbidities (e.g. dyslipidemia) and liver/kidney disease. Additionally, exposure to ART and direct viral-mediated effects of chronic HIV/HCV further elevate cardio-metabolic risk in the aging co-infected population.
III.A. Patient-specific factors
The development of metabolic syndrome in the general population is associated with age, race/ethnicity and increased body weight36,37. Other risk factors in NHANES included smoking, low household income, high carbohydrate diet and physical inactivity38, factors especially prevalent in HIV/HCV co-infected individuals. Metabolic syndrome is also associated with hepatic steatosis, fibrosis and cirrhosis39, along with CKD and microalbuminuria40, comorbidities common to those with HIV/HCV co-infection. In terms of traditional cardiovascular risk, evidence is strongest for overrepresentation of dyslipidemia in PWH compared with the general population, however, emerging data suggest other metabolic components are likely also more common in PWH; the role of HCV co-infection is less clear. Patient-specific factors contributing to the development of metabolic syndrome in HIV/HCV co-infection are summarized in Table 1.
Table 1.
Patient-specific Factors Contributing to the Development of Metabolic Syndrome in HIV/HCV Co-infection
| Risk category | Viral infection | Risk factors |
|---|---|---|
| Socioeconomics | HIV/HCV | Low household income High carbohydrate diet Physical inactivity |
| Substance use | HIV/HCV | Tobacco (increased CVD risk and mortality) Marijuana (increased CVD risk) Cocaine (subclinical coronary atherosclerosis) |
| Liver disease | HIV |
|
| HCV |
|
|
| Kidney disease | HIV/HCV |
|
| HIV |
|
|
| HCV |
|
|
| Cardiovascular comorbidities | HIV/HCV | Dyslipidemia Elevated blood pressure Impaired glucose tolerance Increased waist-to-hip ratio |
Abbreviations: ART = antiretroviral therapy; CVD = cardiovascular disease
III.A.i. Substance use
Rates of smoking in PWH are nearly twice as high than in the general population: 42% and 20% of >4,000 PWH reported current and former cigarette use, respectively, compared with 21% and 22%, respectively, of nearly 28,000 U.S. adults without HIV41. Concurrent substance use with marijuana and cocaine is common in PWH and both have been associated with CVD risk in this population42,43. Furthermore, the effects of tobacco and cocaine use have proven more deleterious in PWH, contributing to higher mortality and worsened subclinical coronary atherosclerosis, respectively, compared with the general population42,44 Drug use including smoking is also more common in HCV-infected than HCV-uninfected persons45. Given a clear predisposition to all four metabolic syndrome components, it is well-founded that the high prevalence of polysubstance abuse by persons with HIV/HCV co-infection exacerbates cardio-metabolic risk.
III.A.ii. Liver and kidney disease
A recent trend analysis using cross-sectional MarketScan research data from 64,398 PWH evaluated prevalent non-AIDS comorbidities between 2003 and 2013. The most common comorbidities were hypertension (31-76%), hyperlipidemia (22-50%) and diabetes (11-37%) (ranges reflect payer source)3. Compared with HIV-negative matched controls, the proportions of PWH with HCV infection, liver disease and renal impairment were significantly greater3. HIV infection accelerates the natural history of HCV-mediated liver disease, resulting in higher rates of fibrosis progression, hepatic decompensation and hepatocellular carcinoma9-12. It should also be noted that PWH are at increased risk of hepatic steatosis independent of viral hepatitis. For example, other etiologies of chronic liver disease include alcoholic- and non-alcoholic fatty liver disease, drug-induced liver injury, opportunistic infections and AIDS cholangiopathy46.
HIV and HCV are independently associated with CKD47. Studies of PWH have consistently shown the risk of CKD increases dramatically (i.e. 90%) in the presence of HCV co-infection35,48 In a study of >20,000 veterans with HIV, those with HIV/HCV co-infection had significantly higher rates of CKD and mortality, even when adjusting for degree of kidney impairment49. According to NHANES data, metabolic syndrome significantly increases the risk of CKD and microalbuminuria in a “dose-dependent” fashion based on the number of metabolic components40. The high prevalence of metabolic comorbidities in HIV/HCV co-infection may exacerbate renal complications and death in this patient population.
III.A.iii. Traditional cardiovascular risk
Data from two large, prospective U.S. cohorts of women and men with and without HIV (the WIHS and the Multicenter AIDS Cohort Study [MACS], respectively) estimated 10-year risk of developing CVD by the Framingham risk score equations50. Compared with seronegative controls, men and women with HIV more frequently had reduced levels of high-density lipoprotein (HDL) cholesterol and elevated triglycerides50, a finding consistent with a similar cohort of French men and women51. In the WIHS/MACS study, ART exposure, being overweight and having a low income predicted CVD risk in PWH50. While HIV-associated dyslipidemia can be exacerbated by ART, it is also present in ART-naïve individuals52. In 50 men whom HIV-seroconverted in the MACS, significant declines in mean serum total cholesterol, HDL and low-density lipoprotein (LDL) cholesterol were observed for several years after HIV infection53. Subsequent ART initiation was associated with increased total and LDL cholesterol but minimal change in HDL cholesterol levels.
Chronic HCV is also associated with dyslipidemia17. Acquisition of HCV infection has been shown to lead to hypolipidemia, including a progressive decline in triglycerides and total, HDL and LDL cholesterol levels independent of BMI and liver fibrosis54. Further, some data suggest HCV co-infection may confer protection against HIV-associated dyslipidemia, a finding supported by a lower observed use of lipid-lowering agents in co-infected patients55,56.
Findings from the MACS following 5,578 men over 19 years revealed an association between systolic hypertension and prolonged ART use, controlling for age, BMI and smoking57. Interestingly, the odds of systolic hypertension were similar in ART-naïve men and those with ART exposure <2 years compared with HIV-seronegative men. Recently, an association between hypertension and higher waist-to-hip ratio among PWH has also been suggested58,59. In the Veterans Aging Cohort Study, baseline and incident T2DM after ART initiation were lower in PWH compared with HIV-seronegative controls, however, weight gained in the first year after initiating ART was associated with greater risk of T2DM in PWH60. While ART is a major driver of cardio-metabolic risk in PWH and those with HCV co-infection, viral-mediated effects also contribute – the roles of each are discussed below.
III.B. ART impact on metabolic syndrome in HIV/HCV co-infection
The preponderance of evidence supports an increased cardio-metabolic risk in ART-exposed compared with ART-naïve PWH13,61,62. However, the benefits of ART such as HIV virologic control, immunologic recovery, decreased inflammation and immune activation, and prolonged longevity, undoubtedly outweigh treatment-associated risks63-65. While an exhaustive analysis of ART is beyond the scope of this review, it is important to note its global impact on metabolic syndrome and the differential impact of select classes and agents on metabolic events in HIV/HCV co-infection.
III.B.i. ART and lipodystrophy syndrome
The HIV-associated lipodystrophy syndrome is characterized by lipoatrophy and/or fat accumulation. Clinical presentation is heterogeneous and estimated prevalence ranges from 10-80%66,67. While the development of lipoatrophy is primarily attributed to thymidine analogue nucleoside reverse transcriptase inhibitors (e.g. stavudine, zidovudine), no specific antiretroviral agent or class has been implicated in fat accumulation68. Other possible contributors to lipodystrophy syndrome include: lower fat mass at baseline, HIV disease severity (lipoatrophy); female sex, elevated baseline triglycerides, higher body fat percentage (fat accumulation); and, increased age (lipoatrophy and fat accumulation)69. Both lipoatrophy and fat accumulation have been associated with abnormal glucose and lipid metabolism, even in the absence of frank obesity70. Notably, lipodystrophy and associated metabolic complications may be worsened by HCV co-infection. In a cross-sectional analysis of patients with comparable age, CD4 count, HIV viral load and duration of ART, lipoatrophy was more prevalent in HIV/HCV co-infected versus HIV mono-infected patients (41% vs 14%, p=0.003)71. In multivariable analysis, IR, BMI, triglycerides and peripheral fat wasting were associated with HCV co-infection.
III.B.ii. Metabolic effects of ART
Evaluating the pathogenesis driving atherosclerosis in HIV, Piconi et al demonstrated pro-thrombotic and inflammatory mediators were lower, and metabolic factors (i.e. serum cholesterol and lipoproteins) higher, in ART-treated than ART-naïve PWH65. Data from the MACS and WIHS cohorts found that ART exposure induces an atherogenic lipoprotein profile in men and women with HIV, respectively53,72. As mentioned previously, studies have suggested HCV co-infection may blunt ART-associated hypercholesterolemia55,56,73 While the most prominent ART-induced metabolic derangement is dyslipidemia, elevated risks of hypertension and diabetes have also been reported, even when controlling for possible lipid effects74-76. Important clinical considerations for cardio-metabolic risk by ART class are summarized in Table 2, and a focused discussion on the most recent data follows.
Table 2.
Summary of Recommended Antiretroviral Regimens by Drug Class and Potential Metabolic Considerations for Persons with HIV/HCV Co-infection
| Impact on Lipids |
Cardiovascular disease |
Liver disease | Kidney disease |
PK drug interactions |
|
|---|---|---|---|---|---|
| Nucleoside Reverse Transcriptase Inhibitors (NRTI) | |||||
| ABC/3TC | - | (X) | (X) in CTP B, C | - | - |
| TAF/FTC | ↓ | - | Active against Hepatitis B | - | - |
| TDF/FTC | ↑ | - | Active against Hepatitis B | (X) | - |
| Non-nucleoside Reverse Transcriptase Inhibitors (NNRTI) | |||||
| DOR | - | - | - | - | - |
| EFV | ↓ | - | - | - | Many DDIs possible* |
| RPV | - | - | - | - | (X) with acid-lowering agents |
| Protease Inhibitors (PI) | |||||
| ATV/c, ATV/r | ↓ | ↑ | Increases indirect hyper-bilirubinemia | May cause nephrolithiasis | (X) with acid-lowering agents |
| DRV/c, DRV/r | ↓ | (X) | - | - | Many DDIs possible* |
| Integrase Strand Transfer Inhibitors (INSTI) | |||||
| BIC | - | - | - | - | (X) coadministration with polyvalent cations |
| DTG | - | - | - | - | (X) coadministration with polyvalent cations |
| EVG/c | ↓ | - | - | - | Many DDIs possible* |
| RAL | - | - | - | - | (X) coadministration with polyvalent cations |
Abbreviations: 3TC = Lamivudine; ABC = Abacavir; ATV = Atazanavir; BIC = Bictegravir; CTP = Child Turcotte Pugh; c = Cobicistat; DDIs = Drug-drug interactions; DOR = Doravirine; DRV = Darunavir; DTG = Dolutegravir; EVG = Elvitegravir; FTC = Emtricitabine; PK = pharmacokinetic; r = Ritonavir; RAL = Raltegravir; RPV = Rilpivirine; TAF = Tenofovir Alafenamide Fumarate; TDF = Tenofovir Disoproxil Fumarate
Symbols: (X) = Avoid use; (--) = No reported association; ↑ = May benefit; ↓ = May worsen
Suggest referencing a medication interaction tool (i.e. https://www.hiv-druginteractions.org/)
III.B.ii.a. Nucleoside reverse transcriptase inhibitors (NRTIs)
Tenofovir disoproxil fumarate (TDF) is associated with lower lipid levels than tenofovir alafenamide (TAF) and abacavir77-79. However, TDF use has been associated with kidney function decline, proximal renal tubulopathy (leading to proteinuria and phosphate wasting), and reduced bone mineral density (BMD). While abacavir use has fewer nephrotoxic and deleterious BMD effects than TDF, it has been associated with increased risk of cardiovascular events (including AMI) in some, but not all, observational studies80-82. Switching the NRTI backbone component from TDF to TAF given renal and bone safety advantages is increasingly common. While TAF-associated rises in triglyceride, total and LDL cholesterol levels (due to decreased plasma tenofovir levels) may be reversed by switching back to TDF83, long-term follow-up studies assessing lipid changes on cardio-metabolic clinical outcomes are needed.
III.B.ii.b. Protease inhibitors (PIs)
Hypercholesteremia and hypertriglyceridemia are common with use of PIs, though effect varies by individual PI84. Observational cohort studies have found an association between some PIs and an increased risk of cardiovascular events81, however, this has not been seen with atazanavir85,86 It is speculated that the indirect hyperbilirubinemia observed with atazanavir use may be cardioprotective, however, further study is needed, especially considering possible alteration by HCV co-infection87,88.
III.B.ii.c. Non-nucleoside reverse transcriptase inhibitors (NNRTIs)
Use of NNRTIs, including efavirenz, has been associated with increased LDL and total cholesterol levels, compared to those of ART-naïve PWH, however, this is counterbalanced by increased HDL levels89. The overall favorable lipid profile associated with NNRTIs appears most pronounced for rilpivirine and the newest agent in this class, doravirine, as demonstrated in randomized clinical trials of ART-naïve patients comparing initiation of either to efavirenz90,91.
III.B.ii.d. Integrase strand transfer inhibitors (INSTIs)
While the newest ART class has favorable effects on lipids, results from two recently published open-label, phase 3, randomized clinical trials in Africa, ADVANCE92 and NAMSAL93, have intensified concerns regarding metabolic risk associated with INSTI use. In both trials, significantly more weight gain was observed with dolutegravir-containing compared with efavirenz-based regimens (5-6 vs 2-3 kilograms, respectively); especially when combined with TAF and in female patients92. Data from 1118 WIHS participants (2008-2017) demonstrated women on ART who switched to or added an INSTI not only gained significantly more weight, but also experienced increases in BMI, body fat and blood pressure, compared with women who remained on non-INSTI ART94.
III.C. HIV and HCV viral-mediated effects on metabolic syndrome
The ongoing viral replication of chronic HIV/HCV infection induces an inflammatory and immune activated state in the host, leading to alterations in glucose and lipid metabolism14. Resultant metabolic complications – often exacerbated by ART – include hepatic steatosis, IR, dyslipidemia and visceral adiposity, which ultimately increase the risk of T2DM and CVD in HIV/HCV co-infected patients. Proposed viral mechanisms driving the development of metabolic syndrome are reviewed here and summarized in the Figure.
Figure.
Summary of mechanisms by which chronic HCV and HIV infection increase, decrease, or accelerate the risk of metabolic syndrome mediated by hepatic steatosis, insulin resistance, dyslipidemia and visceral adiposity. (ART = antiretroviral therapy)
III.C.i. Hepatic steatosis
HCV core protein, a key modulator of viral and cellular gene expression, has been shown to disrupt lipid metabolism and play a significant role in the development of hepatic steatosis95. Upon hepatocyte invasion, HCV core protein facilitates viral replication and assembly. The replication complex co-locates near hepatocyte lipid droplets, allowing HCV core protein to directly interfere with intracellular lipid metabolism by overcoming host machinery. This leads to decreased expression of peroxisome proliferator activating receptor-alpha/gamma and activation of sterol regulatory element-binding protein, ultimately triggering lipogenesis96. Pathways exploited by HCV core protein also impair lipid degradation and export, exacerbating hepatic steatosis96.
In a study evaluating PWH with elevated aminotransferases by liver biopsy, 75% had evidence of steatosis; of whom, 32% were graded moderate to severe97. ART duration ≥12 months was the greatest risk factor. When compared with HCV mono-infection, HIV/HCV co-infection hastens the trajectory of chronic liver disease as measured by prevalence, severity and necro-inflammatory scores of steatosis95. While the exact mechanism of accelerated hepatic fibrosis progression is unknown, HIV viremia likely plays a significant role. In a study of 274 HIV/HCV co-infected patients, those with any degree of HIV viremia had significantly faster progression than those virologically-suppressed; however, when those virologically-suppressed were compared with 382 HCV mono-infected patients, rates of fibrosis progression were comparable98.
III.C.ii. Insulin resistance
Inflammatory states, such as those seen in chronic viral infection, facilitate the interaction of hepatocytes and adipocytes with extracellular mediators (e.g. cytokines, free fatty acids). This initiates a signaling cascade leading to inhibition of insulin action and generation of IR through enhanced glycogenesis, gluconeogenesis and hepatic glucose production99. Chronic HCV promotes an inflammatory state mediated by host and viral factors (i.e. the production of interferon-gamma/tumor necrosis factor-alpha [TNF-a] and HCV core protein, respectively). In turn, TNF-a mediates chronic activation of monocytes/macrophages and can lead to elevated homeostatic model assessment of IR (HOMA-IR) levels and hepatic steatosis99.
In PWH, NRTI exposure was independently associated with lactate production, however, this relationship was attenuated when adjusting for HOMA-IR levels, suggesting IR may drive this association100. NRTIs are implicated in mitochondrial toxicity and formation of reactive oxidative species; this downregulates adiponectin and increases lactate production in adipocytes, culminating in IR101. Translocation of gut microbes may also contribute to HIV-associated development of IR. Depletion of gut-associated lymphoid tissue during primary HIV infection increases epithelial permeability and microbial translocation, thereby activating toll-like receptor-4 and the NF-kB pathway. Downstream inflammatory signaling activates release of interferon-gamma and lipopolysaccharides, ultimately promoting IR and atherogenesis 102,103.
Reid et al found that sCD163, a marker of monocyte/macrophage activation, may play a significant role in mediating the development of IR through inflammation triggered by gut translocation in the setting of HIV/HCV co-infection102. The authors speculate that the association between co-infection and greater HOMA-IR is likely facilitated by viral and non-viral factors affecting sCD163, and further investigation is needed.
III.C.iii. Dyslipidemia
HIV infection promotes dyslipidemia and atherogenesis through direct viral effects and ART use. HIV viremia increases inflammatory cytokines (e.g. TNF-a, interferon-alpha, interleukins, steroid hormones), which induce hypertriglyceridemia104. HIV replication modifies HDL cholesterol levels through upregulating cholesteryl ester transfer protein activity, stimulating atherogenesis105. In terms of ART, PIs directly stimulate hepatic triglyceride synthesis and decrease lipolytic activity, resulting in increased very-LDL secretion and decreased circulating HDL106. Similarly, the mechanism driving NNRTI-associated dyslipidemia appears related to increased apoA1 production promoting capacity for lipoprotein secretion, whereas NRTIs stimulate lipid production indirectly through mitochondrial toxicity107.
In contrast, HCV is associated with hypolipidemia as previously discussed54. Pathways whereby HCV contributes to atherogenesis include chronic inflammation, direct vascular invasion, disruption of lipid metabolism, and increased IR108. When compounded with HIV-associated lipid effects, co-infected patients experience an increased risk of atherosclerosis and CVD.
III.C.iv. Visceral adiposity
Both HIV- and HCV-associated chronic inflammation independently stimulate visceral adipose tissue accumulation, which likely plays a role in the development of obesity. A study of 236 HCV-infected patients showed a higher visceral adiposity index was associated with degree of HCV viremia, severity of steatosis and necroinflammation109. HIV viral proteins (i.e. Vpr, Tat) have been implicated in affecting adipogenesis through increased inflammatory cytokine expression thereby affecting adipose tissue distribution in PWH. ART may further enhance visceral adiposity development by increasing systemic inflammation and altering expression of mitochondrial DNA and transcription factors regulating lymphocyte proliferation, differentiation and activation110,111.
There are limited studies on the effect of HIV/HCV co-infection on visceral adiposity. A cross-sectional analysis comparing co-infected men and women with those HIV mono-infected suggested amounts of subcutaneous and visceral adipose tissue are comparable between groups, though subtle differences by gender and ART status may exist112. The authors speculate that HIV commonly precedes HCV infection, therefore HCV has minimal additional contribution to preexistent HIV viral- and ART-mediated effects on adiposity (e.g. mitochondrial toxicity and systemic inflammation).
IV. Clinical considerations for management of metabolic syndrome in HIV/HCV co-infection
The landscape for managing HIV/HCV co-infected patients has vastly changed in the era of highly efficacious and tolerable ART as well as curative anti-HCV DAA therapy19,20. This cohort is now aging and the cornerstone of management hinges on addressing senescent-related comorbidities and sequelae of chronic viral infection and therapy-associated side effects. The safety and efficacy of DAAs allows for treatment of co-infected patients in the same way as HCV mono-infected patients19 and confers a mortality benefit in ART-treated PWH113. Below we discuss important clinical considerations for the management of metabolic syndrome in HIV/HCV co-infected patients, including cardio-metabolic screening and risk mitigation, recognizing relevant drug-drug interactions (DDIs), and possible metabolic improvement after DAA-induced HCV eradication.
IV.A. Screening and monitoring of metabolic syndrome
Given the increased baseline risk of metabolic syndrome in PWH and in those with chronic HCV, it is imperative to screen for metabolic comorbidities during initial clinical assessment, prior to ART initiation. Primary Care Guidelines for the management of PWH recommend evaluation of renal or hepatic dysfunction with a comprehensive metabolic panel, viral hepatitis testing, and screening for dyslipidemia (fasting lipid panel) and impaired glucose tolerance (either hemoglobin A1c or fasting blood glucose) prior to and 1-3 months after starting ART114. Additionally, blood pressure and body weight (including BMI) should be measured at the initial clinic visit and each follow-up appointment. Furthermore, it is crucial to screen for comorbid substance use disorders with standardized instruments such as the AUDIT-C and counsel on harm reduction where appropriate115.
Many recommendations for cardio-metabolic screening in PWH are based on clinical guidelines for the general population. However, data demonstrating the burden of metabolic comorbidities in PWH approximates that of HIV-seronegative persons aged ten years older support the need for earlier screening in PWH116. Appropriate intervals for reassessing metabolic derangements in PWH are unknown, and data to specifically inform guidance on screening and monitoring of metabolic syndrome in HIV/HCV co-infection are lacking. An opportunity for preventive intervention before the onset of metabolic disease may be assessing trends in routinely collected clinical data. For example, Rhee et al found that at least two measurements of random plasma glucose >130 mg/dL was nearly 95% specific for T2DM onset within five years117.
IV.B. Metabolic consequences of drug-drug interactions
Selecting an ART regimen poses several important host and virologic considerations, including presence of HCV co-infection118. Specific metabolic effects by ART class are discussed previously and summarized in Table 2. Additionally, critical pharmacokinetic DDIs involving the cytochrome P450 enzyme system, specifically the CYP3A4 enzyme, are relevant in managing HIV/HCV co-infected patients given potential for metabolic consequences. Ritonavir and cobicistat, included in PI- and INSTI-based ART as a strategy to increase or “boost” antiretroviral exposure and accommodate once daily dosing, both strongly inhibit CYP3A4. It is therefore critical to recognize either pharmacologic “booster” in an ART regimen when selecting anti-HCV DAA or lipid-lowering (i.e. HMG-CoA reductase inhibitor or “statin”) therapy given possible DDIs. The potent inhibition of CYP3A4 by ritonavir or cobicistat can also lead to significantly higher levels of certain anti-psychotic drugs and worsen associated metabolic toxicities. A medication interaction tool should be used to review potential DDIs before initiating ART and/or DAAs with other concomitant medications (i.e. https://www.hiv-druginteractions.org/).
IV.C. Metabolic reversal with HCV clearance after DAA therapy
There is emerging literature describing recovery of metabolic impairment in patients who achieve HCV cure after DAA treatment21, however, reports in HIV/HCV co-infected individuals are lacking. In the era of interferon-free DAA therapy with HCV eradication rates of 94-100%19, improvements in HCV-associated alterations in glucose and lipid metabolism are attributed to viral clearance.
In a prospective case-control study of 133 HCV-infected patients with advanced fibrosis though not T2DM, DAA-induced HCV clearance improved IR and glycemic control119. In 2,435 veterans with T2DM, a significantly greater drop in hemoglobin (mean decrease of 0.98%) and reduced need for insulin was observed one year after DAAs in those who achieved sustained virologic response versus treatment failure120. A proposed mechanism is that HCV eradication by DAAs enhances insulin sensitivity, however, studies are conflicting on whether this occurs in peripheral and/or hepatic tissue121,122. Further, HCV-associated hypolipidemia and intra-hepatic lipid accumulation has been shown to normalize after DAA therapy initiation, resulting in significantly improved steatosis123. In a cohort of liver-transplant recipients, DAA-based eradication of recurrent HCV infection resulted in a decreased need for treatment of diabetes and hypertension by 38% and 22% from baseline, respectively124. The effects of HCV clearance by DAA treatment on clinical metabolic outcomes show promise in HCV mono-infected patients though data are needed in PWH.
IV.D. Clinical approach to metabolic modification in HIV/HCV co-infected patients
Clinicians caring for HIV/HCV co-infected patients should utilize regular clinic visits as opportunities to reduce modifiable metabolic risk factors including: (1) brief assessment and evidence-based intervention for substance use disorders; (2) counseling to minimize unhealthy weight gain; (3) aggressive pharmacotherapy management of hypertension, diabetes and dyslipidemia; (4) prioritizing HCV cure with DAA therapy; and (5) periodic re-assessment of ART regimen in virologically suppressed PWH to consider whether a different (potentially two-drug125) regimen may be associated with lower cardio-metabolic risk over time125. Ideally, multi-disciplinary teams comprising addiction psychiatrists, dieticians, clinical pharmacy specialists, and health coaches in addition to nurses, advanced practice practitioners, and physicians should be employed to care for this aging cohort of HIV/HCV co-infected patients at high-risk of metabolic syndrome.
V. Conclusions
Persons with HIV/HCV co-infection are aging due to the success of ART and accordingly, suffer from an increased risk of metabolic syndrome and subsequent development of T2DM and CVD. Traditional and non-traditional cardio-metabolic risk factors are overrepresented in co-infected patients compared with the general population: poor socioeconomics, substance use, cardiovascular comorbidities, liver and kidney disease. Direct viral-mediated effects of chronic HIV/HCV infection and toxicities associated with long-term ART exposure further compound risk by altering host glucose and lipid metabolic pathways. HCV eradication by DAA therapy may improve metabolic comorbidities, however, additional study in HIV/HCV co-infection is needed. Future research programs should aim to better characterize metabolic syndrome in HIV/HCV co-infected patients with the overall goal of improved screening, treatment and prevention. This will require additional investigation into optimal screening modalities and intervals, comorbidity reduction, viral effects on host metabolism, and safer ART regimens.
OPINION STATEMENT.
Persons with HIV are living longer due to highly effective antiretroviral therapy (ART). Age-related non-AIDS comorbidities (e.g. cardiovascular and metabolic disorders) increasingly account for morbidity and mortality in this population. Chronic HCV is independently associated with dyslipidemia, hepatic steatosis and insulin resistance. The role of curative anti-HCV direct-acting antiviral (DAA) therapy in reversing metabolic derangements is being actively investigated. Less is known regarding how HIV co-infection may influence HCV-induced metabolic sequelae and potential recovery with DAAs after viral cure. However, evidence consistently supports that compared with mono-infected counterparts, patients with HIV/HCV co-infection are at increased risk of metabolic syndrome, which predisposes to the development of type 2 diabetes and cardiovascular disease. This is likely multifactorial due to patient-specific factors, direct effects of chronic HIV/HCV infection on glucose and lipid metabolism, and cardio-metabolic toxicities associated with long-term ART use. Newer antiretrovirals, particularly integrase strand transfer inhibitors and tenofovir alafenamide, have been associated with weight gain and dyslipidemia, respectively. Additional investigation of the mechanisms – both viral- and ART-mediated – driving metabolic syndrome in HIV/HCV co-infection is needed; in addition to the potential for metabolic reversal following DAA therapy. Finally, further studies evaluating appropriate screening modalities and intervals, prevention tools, and therapeutics are needed to inform the clinical management of metabolic syndrome in patients with HIV/HCV co-infection.
Acknowledgments
Compliance with Ethical Standards
The authors’ research activities are supported, in part, by grants from the National Institutes of Health/National Center for Advancing Translational Sciences (NCATS) (TL1TR002382, UL1TR002378).
Footnotes
Conflict of Interest
The authors declare no conflicts of interest relevant to this manuscript.
Human and Animal Rights and Informed Consent
This article does not contain any studies with human or animal subjects performed by any of the authors.
References
Papers of particular interest, published recently, have been highlighted as:
• Of importance
•• Of major importance
- 1.Marcus JL, Chao CR, Leyden WA, et al. Narrowing the Gap in Life Expectancy Between HIV-Infected and HIV-Uninfected Individuals With Access to Care. J Acquir Immune Defic Syndr. 2016;73(1):39–46. doi: 10.1097/QAI.0000000000001014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Centers for Disease Control and Prevention. HIV Surveillance Report, 2017; vol. 29 http://www.cdc.gov/hiv/library/reports/hiv-surveillance.html. Published November 2018. Accessed [June 1,2019]. [Google Scholar]
- 3.Gallant J, Hsue PY, Shreay S, Meyer N. Comorbidities Among US Patients With Prevalent HIV Infection-A Trend Analysis. J Infect Dis. 2017;216(12):1525–1533. doi: 10.1093/infdis/jix518 [DOI] [PubMed] [Google Scholar]
- 4.Neuhaus J, Angus B, Kowalska JD, et al. Risk of all-cause mortality associated with nonfatal AIDS and serious non-AIDS events among adults infected with HIV. AIDS. 2010;24(5):697–706. doi: 10.1097/QAD.0b013e3283365356 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Antiretroviral Therapy Cohort Collaboration. Causes of death in HIV-1-infected patients treated with antiretroviral therapy, 1996-2006: collaborative analysis of 13 HIV cohort studies. Clin Infect Dis. 2010;50(10):1387–1396. doi: 10.1086/652283 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.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;59(12):1787–1797. doi: 10.1093/cid/ciu701 [DOI] [PubMed] [Google Scholar]
- 7.Cole MB, Galárraga O, Rahman M, Wilson IB. Trends in Comorbid Conditions Among Medicaid Enrollees With HIV. Open Forum Infect Dis. 2019;6(4):ofz124. doi: 10.1093/ofid/ofz124 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Staples CT, Rimland D, Dudas D. Hepatitis C in the HIV (human immunodeficiency virus) Atlanta V.A. (Veterans Affairs Medical Center) Cohort Study (HAVACS): the effect of coinfection on survival. Clin Infect Dis. 1999;29(1):150–154. doi: 10.1086/520144 [DOI] [PubMed] [Google Scholar]
- 9.Benhamou Y, Bochet M, Di Martino V, et al. Liver fibrosis progression in human immunodeficiency virus and hepatitis C virus coinfected patients. The Multivirc Group. Hepatology. 1999;30(4):1054–1058. doi: 10.1002/hep.510300409 [DOI] [PubMed] [Google Scholar]
- 10.Kirk GD, Mehta SH, Astemborski J, et al. HIV, age, and the severity of hepatitis C virus-related liver disease: a cohort study. Ann Intern Med. 2013;158(9):658–666. doi: 10.7326/0003-4819-158-9-201305070-00604 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Fierer DS, Dieterich DT, Fiel MI, et al. Rapid progression to decompensated cirrhosis, liver transplant, and death in HIV-infected men after primary hepatitis C virus infection. Clin Infect Dis. 2013;56(7):1038–1043. doi: 10.1093/cid/cis1206 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Graham CS, Baden LR, Yu E, et al. Influence of human immunodeficiency virus infection on the course of hepatitis C virus infection: a meta-analysis. Clin Infect Dis. 2001;33(4):562–569. doi: 10.1086/321909 [DOI] [PubMed] [Google Scholar]
- 13.Bedimo R, Abodunde O. Metabolic and Cardiovascular Complications in HIV/HCV-Co-infected Patients. Curr HIV/AIDS Rep. 2016;13(6):328–339. doi: 10.1007/s11904-016-0333-9 [DOI] [PubMed] [Google Scholar]
- 14.Slama L, Le Camus C, Serfaty L, Pialoux G, Capeau J, Gharakhanian S. Metabolic disorders and chronic viral disease: the case of HIV and HCV. Diabetes Metab. 2009;35(1):1–11. doi: 10.1016/j.diabet.2008.08.003 [DOI] [PubMed] [Google Scholar]
- 15.Freiberg MS, Chang C-CH, Skanderson M, et al. The risk of incident coronary heart disease among veterans with and without HIV and hepatitis C. Circ Cardiovasc Qual Outcomes. 2011;4(4):425–432. doi: 10.1161/CIRCOUTCOMES.110.957415 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.McKibben RA, Haberlen SA, Post WS, et al. A Cross-sectional Study of the Association Between Chronic Hepatitis C Virus Infection and Subclinical Coronary Atherosclerosis Among Participants in the Multicenter AIDS Cohort Study. J Infect Dis. 2016;213(2):257–265. doi: 10.1093/infdis/jiv396 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Shiffman ML, Gunn NT. Impact of hepatitis C virus therapy on metabolism and public health. Liver Int. 2017;37 Suppl 1:13–18. doi: 10.1111/liv.13282 [DOI] [PubMed] [Google Scholar]
- 18.Banks DE, Bogler Y, Bhuket T, Liu B, Wong RJ. Significant disparities in risks of diabetes mellitus and metabolic syndrome among chronic hepatitis C virus patients in the U.S. Diabetes Metab Syndr. 2017;11 Suppl 1:S153–S158. doi: 10.1016/j.dsx.2016.12.025 [DOI] [PubMed] [Google Scholar]
- 19.Marks K, Naggie S. Management of Hepatitis C in 2019. JAMA. May 2019. doi: 10.1001/jama.2019.5353 [DOI] [PubMed] [Google Scholar]
- 20.Collins LF, Chan A, Zheng J, et al. Direct-Acting Antivirals Improve Access to Care and Cure for Patients With HIV and Chronic HCV Infection. Open Forum Infect Dis. 2018;5(1):ofx264. doi: 10.1093/ofid/ofx264 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Lanini S, Scognamiglio P, Pisapia R, Minosse C, Agresta A, Ippolito G. Recovery of metabolic impairment in patients who cleared chronic hepatitis C infection after direct-acting antiviral therapy. Int J Antimicrob Agents. 2019;53(5):559–563. doi: 10.1016/j.ijantimicag.2018.11.024 [DOI] [PubMed] [Google Scholar]
- 22.Alberti KGMM, Eckel RH, Grundy SM, et al. Harmonizing the metabolic syndrome: a joint interim statement of the International Diabetes Federation Task Force on Epidemiology and Prevention; National Heart, Lung, and Blood Institute; American Heart Association; World Heart Federation; International Atherosclerosis Society; and International Association for the Study of Obesity. Circulation. 2009;120(16):1640–1645. doi: 10.1161/CIRCULATIONAHA.109.192644 [DOI] [PubMed] [Google Scholar]
- 23.Sobieszczyk ME, Hoover DR, Anastos K, et al. Prevalence and predictors of metabolic syndrome among HIV-infected and HIV-uninfected women in the Women’s Interagency HIV Study. J Acquir Immune Defic Syndr. 2008;48(3):272–280. doi: 10.1097/QAI.0b013e31817af461 [DOI] [PubMed] [Google Scholar]
- 24.Worm SW, Friis-Møller N, Bruyand M, et al. High prevalence of the metabolic syndrome in HIV-infected patients: impact of different definitions of the metabolic syndrome. AIDS. 2010;24(3):427–435. doi: 10.1097/QAD.0b013e328334344e [DOI] [PubMed] [Google Scholar]
- 25.Colaci M, Malatino L, Antonelli A, Fallahi P, Giuggioli D, Ferri C. Endocrine disorders associated with hepatitis C virus chronic infection. Rev Endocr Metab Disord. 2018;19(4):397–403. doi: 10.1007/s11154-018-9475-y [DOI] [PubMed] [Google Scholar]
- 26.Wand H, Calmy A, Carey DL, et al. Metabolic syndrome, cardiovascular disease and type 2 diabetes mellitus after initiation of antiretroviral therapy in HIV infection. AIDS. 2007;21(18):2445–2453. doi: 10.1097/QAD.0b013e3282efad32 [DOI] [PubMed] [Google Scholar]
- 27.Fabiani S, Fallahi P, Ferrari SM, Miccoli M, Antonelli A. Hepatitis C virus infection and development of type 2 diabetes mellitus: Systematic review and meta-analysis of the literature. Rev Endocr Metab Disord. 2018;19(4):405–420. doi: 10.1007/s11154-017-9440-1 [DOI] [PubMed] [Google Scholar]
- 28.Moucari R, Asselah T, Cazals-Hatem D, et al. Insulin resistance in chronic hepatitis C: association with genotypes 1 and 4, serum HCV RNA level, and liver fibrosis. Gastroenterology. 2008; 134(2):416–423. doi: 10.1053/j.gastro.2007.11.010 [DOI] [PubMed] [Google Scholar]
- 29.Li X, Gao Y, Xu H, Hou J, Gao P. Diabetes mellitus is a significant risk factor for the development of liver cirrhosis in chronic hepatitis C patients. Sci Rep. 2017;7(1):9087. doi: 10.1038/s41598-017-09825-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Marceau P, Biron S, Hould FS, et al. Liver pathology and the metabolic syndrome X in severe obesity. J Clin Endocrinol Metab. 1999;84(5):1513–1517. doi: 10.1210/jcem.84.5.5661 [DOI] [PubMed] [Google Scholar]
- 31.Butt AA, Yan P, Chew KW, et al. Risk of Acute Myocardial Infarction Among Hepatitis C Virus (HCV)-Positive and HCV-Negative Men at Various Lipid Levels: Results From ERCHIVES. Clin Infect Dis. 2017;65(4):557–565. doi: 10.1093/cid/cix359 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Petta S, Maida M, Macaluso FS, et al. Hepatitis C Virus Infection Is Associated With Increased Cardiovascular Mortality: A Meta-Analysis of Observational Studies. Gastroenterology. 2016; 150(1): 145–155.e4; quiz e15-16. doi: 10.1053/j.gastro.2015.09.007 [DOI] [PubMed] [Google Scholar]
- 33.Leone S, Lorenzini P, Cozzi-Lepri A, et al. Impact of diabetes on the risk of serious liver events and liver-related deaths in people living with HIV and hepatitis C co-infection: data from the ICONA Foundation Cohort Study. Eur J Clin Microbiol Infect Dis. June 2019. doi: 10.1007/s10096-019-03618-8 [DOI] [PubMed] [Google Scholar]
- 34.Data Collection on Adverse Events of Anti-HIV Drugs (D:A:D) Study Group, Weber R, Sabin C, et al. HBV or HCV coinfections and risk of myocardial infarction in HIV-infected individuals: the D:A:D Cohort Study. Antivir Ther (Lond). 2010; 15(8): 1077–1086. doi: 10.3851/IMP1681 [DOI] [PubMed] [Google Scholar]
- 35.Bedimo R, Westfall AO, Mugavero M, Drechsler H, Khanna N, Saag M. Hepatitis C virus coinfection and the risk of cardiovascular disease among HIV-infected patients. HIV Med. 2010;11(7):462–468. doi: 10.1111/j.1468-1293.2009.00815.x [DOI] [PubMed] [Google Scholar]
- 36.Ford ES, Giles WH, Dietz WH. Prevalence of the metabolic syndrome among US adults: findings from the third National Health and Nutrition Examination Survey. JAMA. 2002;287(3):356–359. doi: 10.1001/jama.287.3.356 [DOI] [PubMed] [Google Scholar]
- 37.Moore JX, Chaudhary N, Akinyemiju T. Metabolic Syndrome Prevalence by Race/Ethnicity and Sex in the United States, National Health and Nutrition Examination Survey, 1988–2012. Preventing Chronic Disease. 2017;14. doi: 10.5888/pcd14.160287 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Park Y-W, Zhu S, Palaniappan L, Heshka S, Carnethon MR, Heymsfield SB. The metabolic syndrome: prevalence and associated risk factor findings in the US population from the Third National Health and Nutrition Examination Survey, 1988–1994. Arch Intern Med. 2003;163(4):427–436. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Hanley AJG, Williams K, Festa A, Wagenknecht LE, D’Agostino RB, Haffner SM. Liver markers and development of the metabolic syndrome: the insulin resistance atherosclerosis study. Diabetes. 2005;54(11):3140–3147. doi: 10.2337/diabetes.54.11.3140 [DOI] [PubMed] [Google Scholar]
- 40.Chen J, Muntner P, Hamm LL, et al. The metabolic syndrome and chronic kidney disease in U.S. adults. Ann Intern Med. 2004; 140(3): 167–174. doi: 10.7326/0003-4819-140-3-200402030-00007 [DOI] [PubMed] [Google Scholar]
- 41.Mdodo R, Frazier EL, Dube SR, et al. Cigarette smoking prevalence among adults with HIV compared with the general adult population in the United States: cross-sectional surveys. Ann Intern Med. 2015;162(5):335–344. doi: 10.7326/M14-0954 [DOI] [PubMed] [Google Scholar]
- 42.Lai H, Moore R, Celentano DD, et al. HIV Infection Itself May Not Be Associated With Subclinical Coronary Artery Disease Among African Americans Without Cardiovascular Symptoms. J Am Heart Assoc. 2016;5(3):e002529. doi: 10.1161/JAHA.115.002529 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Lorenz DR, Dutta A, Mukerji SS, Holman A, Uno H, Gabuzda D. Marijuana Use Impacts Midlife Cardiovascular Events in HIV-Infected Men. Clin Infect Dis. 2017;65(4):626–635. doi: 10.1093/cid/cix391 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Helleberg M, Afzal S, Kronborg G, et al. Mortality attributable to smoking among HIV-1-infected individuals: a nationwide, population-based cohort study. Clin Infect Dis. 2013;56(5):727–734. doi: 10.1093/cid/cis933 [DOI] [PubMed] [Google Scholar]
- 45.Younossi ZM, Stepanova M, Nader F, Younossi Z, Elsheikh E. Associations of chronic hepatitis C with metabolic and cardiac outcomes. Aliment Pharmacol Ther. 2013;37(6):647–652. doi: 10.1111/apt.12234 [DOI] [PubMed] [Google Scholar]
- 46.Rafiq N, Stepanova M, Lam B, Nader F, Srishord M, Younossi ZM. Predictors of chronic liver disease in individuals with human immunodeficiency virus infection. Ann Hepatol. 2013;13(1):60–64. [PubMed] [Google Scholar]
- 47.Kupin WL. Viral-Associated GN: Hepatitis C and HIV. Clin J Am Soc Nephrol. 2017;12(8):1337–1342. doi: 10.2215/CJN.04320416 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Kalayjian RC, Lau B, Mechekano RN, et al. Risk factors for chronic kidney disease in a large cohort of HIV-1 infected individuals initiating antiretroviral therapy in routine care. AIDS. 2012;26(15):1907–1915. doi: 10.1097/QAD.0b013e328357f5ed [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Fischer MJ, Wyatt CM, Gordon K, et al. Hepatitis C and the risk of kidney disease and mortality in veterans with HIV. J Acquir Immune Defic Syndr. 2010;53(2):222–226. doi: 10.1097/QAI.0b013e3181b980d4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Kaplan RC, Kingsley LA, Sharrett AR, et al. Ten-year predicted coronary heart disease risk in HIV-infected men and women. Clin Infect Dis. 2007;45(8):1074–1081. doi: 10.1086/521935 [DOI] [PubMed] [Google Scholar]
- 51.Savès M, Chêne G, Ducimetière P, et al. Risk factors for coronary heart disease in patients treated for human immunodeficiency virus infection compared with the general population. Clin Infect Dis. 2003;37(2):292–298. doi: 10.1086/375844 [DOI] [PubMed] [Google Scholar]
- 52.Grunfeld C, Pang M, Doerrler W, Shigenaga JK, Jensen P, Feingold KR. Lipids, lipoproteins, triglyceride clearance, and cytokines in human immunodeficiency virus infection and the acquired immunodeficiency syndrome. J Clin Endocrinol Metab. 1992;74(5):1045–1052. doi: 10.1210/jcem.74.5.1373735 [DOI] [PubMed] [Google Scholar]
- 53.Riddler SA, Smit E, Cole SR, et al. Impact of HIV infection and HAART on serum lipids in men. JAMA. 2003;289(22):2978–2982. doi: 10.1001/jama.289.22.2978 [DOI] [PubMed] [Google Scholar]
- 54.Butt AA, Yan P, Simon TG, Chung RT, Abou-Samra A-B, ERCHIVES study team. Changes in circulating lipids level over time after acquiring HCV infection: results from ERCHIVES. BMC Infect Dis. 2015;15:510. doi: 10.1186/s12879-015-1268-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Cooper CL, Mills E, Angel JB. Mitigation of antiretroviral-induced hyperlipidemia by hepatitis C virus co-infection. AIDS. 2007;21(1):71–76. doi: 10.1097/QAD.0b013e3280110ada [DOI] [PubMed] [Google Scholar]
- 56.Patroni A, Torti C, Tomasoni L, et al. Effect of highly active antiretroviral therapy (HAART) and hepatitis C Co-infection on hyperlipidemia in HIV-infected patients: a retrospective longitudinal study. HIV Clin Trials. 2002;3(6):451–461. doi: 10.1310/hct.2002.3.6.002 [DOI] [PubMed] [Google Scholar]
- 57.Seaberg EC, Muñoz A, Lu M, et al. Association between highly active antiretroviral therapy and hypertension in a large cohort of men followed from 1984 to 2003. AIDS. 2005;19(9):953–960. doi: 10.1097/01.aids.0000171410.76607.f8 [DOI] [PubMed] [Google Scholar]
- 58.van Zoest RA, Wit FW, Kooij KW, et al. Higher Prevalence of Hypertension in HIV-1-Infected Patients on Combination Antiretroviral Therapy Is Associated With Changes in Body Composition and Prior Stavudine Exposure. Clin Infect Dis. 2016;63(2):205–213. doi: 10.1093/cid/ciw285 [DOI] [PubMed] [Google Scholar]
- 59.Gelpi M, Afzal S, Lundgren J, et al. Higher Risk of Abdominal Obesity, Elevated Low-Density Lipoprotein Cholesterol, and Hypertriglyceridemia, but not of Hypertension, in People Living With Human Immunodeficiency Virus (HIV): Results From the Copenhagen Comorbidity in HIV Infection Study. Clin Infect Dis. 2018;67(4):579–586. doi: 10.1093/cid/ciy146 [DOI] [PubMed] [Google Scholar]
- 60.Herrin M, Tate JP, Akgün KM, et al. Weight Gain and Incident Diabetes Among HIV-Infected Veterans Initiating Antiretroviral Therapy Compared With Uninfected Individuals. J Acquir Immune Defic Syndr. 2016;73(2):228–236. doi: 10.1097/QAI.0000000000001071 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Currier JS, Taylor A, Boyd F, et al. Coronary heart disease in HIV-infected individuals. J Acquir Immune Defic Syndr. 2003;33(4):506–512. [DOI] [PubMed] [Google Scholar]
- 62.Obel N, Thomsen HF, Kronborg G, et al. Ischemic heart disease in HIV-infected and HIV-uninfected individuals: a population-based cohort study. Clin Infect Dis. 2007;44(12):1625–1631. doi: 10.1086/518285 [DOI] [PubMed] [Google Scholar]
- 63.Drozd DR, Kitahata MM, Althoff KN, et al. Increased Risk of Myocardial Infarction in HIV-Infected Individuals in North America Compared With the General Population. J Acquir Immune Defic Syndr. 2017;75(5):568–576. doi: 10.1097/QAI.0000000000001450 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Friis-Møller N, Sabin CA, Weber R, et al. Combination antiretroviral therapy and the risk of myocardial infarction. N Engl J Med. 2003;349(21):1993–2003. doi: 10.1056/NEJMoa030218 [DOI] [PubMed] [Google Scholar]
- 65.Piconi S, Parisotto S, Rizzardini G, et al. Atherosclerosis is associated with multiple pathogenic mechanisms in HIV-infected antiretroviral-naive or treated individuals. AIDS. 2013;27(3):381–389. doi: 10.1097/QAD.0b013e32835abcc9 [DOI] [PubMed] [Google Scholar]
- 66.Mallon PW, Miller J, Cooper DA, Carr A. Prospective evaluation of the effects of antiretroviral therapy on body composition in HIV-1-infected men starting therapy. AIDS. 2003;17(7):971–979. doi: 10.1097/01.aids.0000060348.78202.74 [DOI] [PubMed] [Google Scholar]
- 67.Heath KV, Hogg RS, Chan KJ, et al. Lipodystrophy-associated morphological, cholesterol and triglyceride abnormalities in a population-based HIV/AIDS treatment database. AIDS. 2001. ;15(2):231–239. [DOI] [PubMed] [Google Scholar]
- 68.Shlay JC, Sharma S, Peng G, Gibert CL, Grunfeld C. Long-term subcutaneous tissue changes among antiretroviral-naive persons initiating stavudine, zidovudine, or abacavir with lamivudine. J Acquir Immune Defic Syndr. 2008;48(1):53–62. [DOI] [PubMed] [Google Scholar]
- 69.Jacobson DL, Knox T, Spiegelman D, Skinner S, Gorbach S, Wanke C. Prevalence of, evolution of, and risk factors for fat atrophy and fat deposition in a cohort of HIV-infected men and women. Clin Infect Dis. 2005;40(12):1837–1845. doi: 10.1086/430379 [DOI] [PubMed] [Google Scholar]
- 70.Addy CL, Gavrila A, Tsiodras S, Brodovicz K, Karchmer AW, Mantzoros CS. Hypoadiponectinemia is associated with insulin resistance, hypertriglyceridemia, and fat redistribution in human immunodeficiency virus-infected patients treated with highly active antiretroviral therapy. J Clin Endocrinol Metab. 2003;88(2):627–636. doi: 10.1210/jc.2002-020795 [DOI] [PubMed] [Google Scholar]
- 71.Duong M, Petit JM, Piroth L, et al. Association between insulin resistance and hepatitis C virus chronic infection in HIV-hepatitis C virus-coinfected patients undergoing antiretroviral therapy. J Acquir Immune Defic Syndr. 2001;27(3):245–250. [DOI] [PubMed] [Google Scholar]
- 72.Parrinello CM, Landay AL, Hodis HN, et al. Treatment-related changes in serum lipids and inflammation: clinical relevance remains unclear. Analyses from the Women’s Interagency HIV study. AIDS. 2013;27(9):1516–1519. doi: 10.1097/QAD.0b013e32835fd8a9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Lapadula G, Torti C, Paraninfo G, et al. Influence of hepatitis C genotypes on lipid levels in HIV-positive patients during highly active antiretroviral therapy. Antivir Ther (Lond). 2006;11(4):521–527. [PubMed] [Google Scholar]
- 74.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; 165(10):1179–1184. doi: 10.1001/archinte.165.10.1179 [DOI] [PubMed] [Google Scholar]
- 75.De Wit S, Sabin CA, Weber R, et al. Incidence and risk factors for new-onset diabetes in HIV-infected patients: the Data Collection on Adverse Events of Anti-HIV Drugs (D:A:D) study. Diabetes Care. 2008;31 (6): 1224–1229. doi: 10.2337/dc07-2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.DAD Study Group, Friis-Møller N, Reiss P, et al. Class of antiretroviral drugs and the risk of myocardial infarction. N Engl J Med. 2007;356(17):1723–1735. doi: 10.1056/NEJMoa062744 [DOI] [PubMed] [Google Scholar]
- 77.Arribas JR, Thompson M, Sax PE, et al. Brief Report: Randomized, Double-Blind Comparison of Tenofovir Alafenamide (TAF) vs Tenofovir Disoproxil Fumarate (TDF), Each Coformulated With Elvitegravir, Cobicistat, and Emtricitabine (E/C/F) for Initial HIV-1 Treatment: Week 144 Results. J Acquir Immune Defic Syndr. 2017;75(2):211–218. doi: 10.1097/QAI.0000000000001350•Of importance: Results at 144 weeks of two double-blind phase three trials demonstrated superiority of tenofovir alafenamide- versus tenofovir disoproxil fumarate-backed initial HIV therapy with improved renal and bone safety indices in the tenofovir alafenamide group, however, increased lipid profiles.
- 78.Gallant JE, Daar ES, Raffi F, et al. Efficacy and safety of tenofovir alafenamide versus tenofovir disoproxil fumarate given as fixed-dose combinations containing emtricitabine as backbones for treatment of HIV-1 infection in virologically suppressed adults: a randomised, double-blind, active-controlled phase 3 trial. Lancet HIV. 2016;3(4):e158–165. doi: 10.1016/S2352-3018(16)00024-2 [DOI] [PubMed] [Google Scholar]
- 79.Gupta SK, Post FA, Arribas JR, et al. Renal safety of tenofovir alafenamide vs. tenofovir disoproxil fumarate: a pooled analysis of 26 clinical trials. AIDS. 2019;33(9):1455–1465. doi: 10.1097/QAD.0000000000002223 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Young J, Xiao Y, Moodie EEM, et al. Effect of Cumulating Exposure to Abacavir on the Risk of Cardiovascular Disease Events in Patients From the Swiss HIV Cohort Study. J Acquir Immune Defic Syndr. 2015;69(4):413–421. doi: 10.1097/QAI.0000000000000662 [DOI] [PubMed] [Google Scholar]
- 81.Worm SW, Sabin C, Weber R, et al. Risk of myocardial infarction in patients with HIV infection exposed to specific individual antiretroviral drugs from the 3 major drug classes: the data collection on adverse events of anti-HIV drugs (D:A:D) study. J Infect Dis. 2010;201(3):318–330. doi: 10.1086/649897 [DOI] [PubMed] [Google Scholar]
- 82.Ding X, Andraca-Carrera E, Cooper C, et al. No association of abacavir use with myocardial infarction: findings of an FDA meta-analysis. J Acquir Immune Defic Syndr. 2012;61(4):441–447. doi: 10.1097/QAI.0b013e31826f993c [DOI] [PubMed] [Google Scholar]
- 83.Milinkovic Ana, Berger Florian, Arenas-Pinto Alejandro, Mauss Stefan. Lipid Changes Associated with TAF are Reversible by Switching Back to TDF. Presented at the: Conference on Retroviruses and Opportunistic Infections 2019; March 4, 2019; Seattle, WA. [Google Scholar]
- 84.Ofotokun I, Na LH, Landovitz RJ, et al. Comparison of the metabolic effects of ritonavir-boosted darunavir or atazanavir versus raltegravir, and the impact of ritonavir plasma exposure: ACTG 5257. Clin Infect Dis. 2015;60(12):1842–1851. doi: 10.1093/cid/civ193 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.LaFleur J, Bress AP, Rosenblatt L, et al. Cardiovascular outcomes among HIV-infected veterans receiving atazanavir. AIDS. 2017;31(15):2095–2106. doi: 10.1097/QAD.0000000000001594 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Ryom L, Lundgren JD, El-Sadr W, et al. Cardiovascular disease and use of contemporary protease inhibitors: the D:A:D international prospective multicohort study. Lancet HIV. 2018;5(6):e291–e300. doi: 10.1016/S2352-3018(18)30043-2 [DOI] [PubMed] [Google Scholar]
- 87.Muccini C, Galli L, Poli A, et al. Brief Report: Hyperbilirubinemia Is Associated With a Decreased Risk of Carotid Atherosclerosis in HIV-Infected Patients on Virological Suppression. J Acquir Immune Defic Syndr. 2018;79(5):617–623. doi: 10.1097/QAI.0000000000001854 [DOI] [PubMed] [Google Scholar]
- 88.Crane HM, Nance RM, Heckbert SR, et al. Association Between Bilirubin, Atazanavir, and Cardiovascular Disease Events Among People Living With HIV Across the United States. J Acquir Immune Defic Syndr. 2019;81(5):e141–e147. doi: 10.1097/QAI.0000000000002071 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Fontas E, van Leth F, Sabin CA, et al. Lipid profiles in HIV-infected patients receiving combination antiretroviral therapy: are different antiretroviral drugs associated with different lipid profiles? J Infect Dis. 2004; 189(6):1056–1074. doi: 10.1086/381783 [DOI] [PubMed] [Google Scholar]
- 90.Tebas P, Sension M, Arribas J, et al. Lipid levels and changes in body fat distribution in treatment-naive, HIV-1-Infected adults treated with rilpivirine or Efavirenz for 96 weeks in the ECHO and THRIVE trials. Clin Infect Dis. 2014;59(3):425–434. doi: 10.1093/cid/ciu234 [DOI] [PubMed] [Google Scholar]
- 91.Thompson M, Orkin C, Molina J-M, et al. Once-Daily Doravirine for Initial Treatment of Adults Living With HIV-1: An Integrated Safety Analysis. Clin Infect Dis. May 2019. doi: 10.1093/cid/ciz423 [DOI] [PubMed] [Google Scholar]
- 92.Venter WDF, Moorhouse M, Sokhela S, et al. Dolutegravir plus Two Different Prodrugs of Tenofovir to Treat HIV. N Engl J Med. July 2019. doi: 10.1056/NEJMoa1902824•Of major importance: This 96-week, phase three, open-label, randomized trial in South Africa comparing dolutegravir (with emtricitabine plus tenofovir alafenamide or tenofovir disoproxil fumarate) with efavirenz-based antiretroviral therapy showed significant weight gain in the dolutegravir group, especially when combined with tenofovir alafenamide and in female patients.
- 93.NAMSAL ANRS 12313 Study Group. Dolutegravir-Based or Low-Dose Efavirenz-Based Regimen for the Treatment of HIV-1. N Engl J Med. July 2019. doi: 10.1056/NEJMoa1904340 [DOI] [PubMed] [Google Scholar]
- 94.Kerchberger AM, Sheth AN, Angert CD, et al. Integrase Strand Transfer Inhibitors are Associated with Weight Gain in Women. Presented at the: Conference on Retroviruses and Opportunistic Infections 2019; March 4, 2019; Seattle, WA. [Google Scholar]
- 95.Gaslightwala I, Bini EJ. Impact of human immunodeficiency virus infection on the prevalence and severity of steatosis in patients with chronic hepatitis C virus infection. J Hepatol. 2006;44(6):1026–1032. doi: 10.1016/j.jhep.2006.02.009 [DOI] [PubMed] [Google Scholar]
- 96.Schaefer EAK, Chung RT. HCV and host lipids: an intimate connection. Semin Liver Dis. 2013;33(4):358–368. doi: 10.1055/s-0033-1358524 [DOI] [PubMed] [Google Scholar]
- 97.Morse CG, McLaughlin M, Matthews L, et al. Nonalcoholic Steatohepatitis and Hepatic Fibrosis in HIV-1-Monoinfected Adults With Elevated Aminotransferase Levels on Antiretroviral Therapy. Clin Infect Dis. 2015;60(10):1569–1578. doi: 10.1093/cid/civ101 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Bräu N, Salvatore M, Ríos-Bedoya CF, et al. Slower fibrosis progression in HIV/HCV-coinfected patients with successful HIV suppression using antiretroviral therapy. J Hepatol. 2006;44(1):47–55. doi: 10.1016/j.jhep.2005.07.006 [DOI] [PubMed] [Google Scholar]
- 99.Serfaty L, Capeau J. Hepatitis C, insulin resistance and diabetes: clinical and pathogenic data. Liver Int. 2009;29 Suppl 2:13–25. doi: 10.1111/j.1478-3231.2008.01952.x [DOI] [PubMed] [Google Scholar]
- 100.Lo JC, Kazemi MR, Hsue PY, et al. The relationship between nucleoside analogue treatment duration, insulin resistance, and fasting arterialized lactate level in patients with HIV infection. Clin Infect Dis. 2005;41 (9):1335–1340. doi: 10.1086/496981 [DOI] [PubMed] [Google Scholar]
- 101.Hadigan C Insulin resistance among HIV-infected patients: unraveling the mechanism. Clin Infect Dis. 2005;41(9):1341–1342. doi: 10.1086/496990 [DOI] [PubMed] [Google Scholar]
- 102.Reid M, Ma Y, Scherzer R, et al. Higher CD163 levels are associated with insulin resistance in hepatitis C virus-infected and HIV-infected adults. AIDS. 2017;31(3):385–393. doi: 10.1097/QAD.0000000000001345•Of importance: Generation of insulin resistance in HIV/HCV co-infection may be mediated by microbial gut translocation triggering inflammatory signaling cascades as evidenced by higher sCD163 levels.
- 103.Brenchley JM, Price DA, Schacker TW, et al. Microbial translocation is a cause of systemic immune activation in chronic HIV infection. Nat Med. 2006; 12(12):1365–1371. doi: 10.1038/nm1511 [DOI] [PubMed] [Google Scholar]
- 104.Grunfeld C, Feingold KR. The role of the cytokines, interferon alpha and tumor necrosis factor in the hypertriglyceridemia and wasting of AIDs. J Nutr. 1992;122(3 Suppl):749–753. doi: 10.1093/jn/122.suppl_3.749 [DOI] [PubMed] [Google Scholar]
- 105.Rose H, Hoy J, Woolley I, et al. HIV infection and high density lipoprotein metabolism. Atherosclerosis. 2008;199(1):79–86. doi: 10.1016/j.atherosclerosis.2007.10.018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Liang JS, Distler O, Cooper DA, et al. HIV protease inhibitors protect apolipoprotein B from degradation by the proteasome: a potential mechanism for protease inhibitor-induced hyperlipidemia. Nat Med. 2001;7(12):1327–1331. doi: 10.1038/nm1201-1327 [DOI] [PubMed] [Google Scholar]
- 107.Kotler DP. HIV and antiretroviral therapy: lipid abnormalities and associated cardiovascular risk in HIV-infected patients. J Acquir Immune Defic Syndr. 2008;49 Suppl 2:S79–85. doi: 10.1097/QAI.0b013e318186519c [DOI] [PubMed] [Google Scholar]
- 108.Babiker A, Jeudy J, Kligerman S, Khambaty M, Shah A, Bagchi S. Risk of Cardiovascular Disease Due to Chronic Hepatitis C Infection: A Review. J Clin Transl Hepatol. 2017;5(4):343–362. doi: 10.14218/JCTH.2017.00021 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Petta S, Amato M, Cabibi D, et al. Visceral adiposity index is associated with histological findings and high viral load in patients with chronic hepatitis C due to genotype 1. Hepatology. 2010;52(5):1543–1552. doi: 10.1002/hep.23859 [DOI] [PubMed] [Google Scholar]
- 110.Koethe JR, Hulgan T, Niswender K. Adipose Tissue and Immune Function: A Review of Evidence Relevant to HIV Infection. J Infect Dis. 2013;208(8):1194–1201. doi: 10.1093/infdis/jit324 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Lake JE. The Fat of the Matter: Obesity and Visceral Adiposity in Treated HIV Infection. Curr HIV/AIDS Rep. 2017;14(6):211–219. doi: 10.1007/s11904-017-0368-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Tien PC, Bacchetti P, Gripshover B, Overton ET, Rimland D, Kotler D. Association Between Hepatitis C Virus Coinfection and Regional Adipose Tissue Volume in HIV-Infected Men and Women. J Acquir Immune Defic Syndr. 2007;45(1):60–65. doi: 10.1097/QAI.0b013e3180423a95 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Breskin A, Westreich D, Cole SR, et al. The Effects of Hepatitis C Infection and Treatment on All-cause Mortality Among People Living With Human Immunodeficiency Virus. Clin Infect Dis. October 2018. doi: 10.1093/cid/ciy5•Of importance: Modeling the 10-year all-cause mortality in persons living with HIV after initiation of antiretroviral therapy showed a significant mortality benefit attributable to HCV elimination by direct-acting antivirals.
- 114.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;58(1):1–10. doi: 10.1093/cid/cit757 [DOI] [PubMed] [Google Scholar]
- 115.Moyer VA, Preventive Services Task Force. Screening and behavioral counseling interventions in primary care to reduce alcohol misuse: U.S. preventive services task force recommendation statement. Ann Intern Med. 2013;159(3):210–218. doi: 10.7326/0003-4819-159-3-201308060-00652 [DOI] [PubMed] [Google Scholar]
- 116.Guaraldi G, Orlando G, Zona S, et al. Premature age-related comorbidities among HIV-infected persons compared with the general population. Clin Infect Dis. 2011;53(11):1120–1126. doi: 10.1093/cid/cir627 [DOI] [PubMed] [Google Scholar]
- 117.Rhee MK, Ho Y-L, Raghavan S, et al. Random plasma glucose predicts the diagnosis of diabetes. PLoS ONE. 2019;14(7):e0219964. doi: 10.1371/journal.pone.0219964 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Panel on Antiretroviral Guidelines for Adults and Adolescents. Guidelines for the Use of Antiretroviral Agents in Adults and Adolescents Living with HIV. Department of Health and Human Services. Available at http://www.aidsinfo.nih.gov/ContentFiles/AdultandAdolescentGL.pdf Accessed 24 Jul 2019.
- 119.Adinolfi LE, Nevola R, Guerrera B, et al. Hepatitis C virus clearance by direct-acting antiviral treatments and impact on insulin resistance in chronic hepatitis C patients. J Gastroenterol Hepatol. 2018;33(7):1379–1382. doi: 10.1111/jgh.14067 [DOI] [PubMed] [Google Scholar]
- 120.Hum J, Jou JH, Green PK, et al. Improvement in Glycemic Control of Type 2 Diabetes After Successful Treatment of Hepatitis C Virus. Diabetes Care. 2017;40(9):1173–1180. doi: 10.2337/dc17-0485 [DOI] [PubMed] [Google Scholar]
- 121.Gastaldi G, Gomes D, Schneiter P, et al. Treatment with direct-acting antivirals improves peripheral insulin sensitivity in non-diabetic, lean chronic hepatitis C patients. PLoS ONE. 2019;14(6):e0217751. doi: 10.1371/journal.pone.0217751 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Lim TR, Hazlehurst JM, Oprescu AI, et al. Hepatitis C virus infection is associated with hepatic and adipose tissue insulin resistance that improves after viral cure. Clin Endocrinol (Oxf). 2019;90(3):440–448. doi: 10.1111/cen.13924 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Meissner EG, Lee Y-J, Osinusi A, et al. Effect of sofosbuvir and ribavirin treatment on peripheral and hepatic lipid metabolism in chronic hepatitis C virus, genotype 1-infected patients. Hepatology. 2015;61(3):790–801. doi: 10.1002/hep.27424 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Beig J, Orr D, Harrison B, Gane E. Hepatitis C Virus Eradication with New Interferon-Free Treatment Improves Metabolic Profile in Hepatitis C Virus-Related Liver Transplant Recipients. Liver Transpl. 2018;24(8):1031–1039. doi: 10.1002/lt.25060 [DOI] [PubMed] [Google Scholar]
- 125.Aboud M, Orkin C, Podzamczer D, et al. Efficacy and safety of dolutegravir-rilpivirine for maintenance of virological suppression in adults with HIV-1: 100-week data from the randomised, open-label, phase 3 SWORD-1 and SWORD-2 studies. Lancet HIV. July 2019. doi: 10.1016/S2352-3018(19)30149-3 [DOI] [PubMed] [Google Scholar]

