Immunology
Review
1 February 2024

Evolving mechanisms and presentations of cardiovascular disease in people with HIV: implications for management

SUMMARY

People with HIV (PWH) are at elevated risk for cardiovascular diseases (CVDs), including myocardial infarction, heart failure, and sudden cardiac death, among other CVD manifestations. Chronic immune dysregulation resulting in persistent inflammation is common among PWH, particularly those with sustained viremia and impaired CD4+ T cell recovery. This inflammatory milieu is a major contributor to CVDs among PWH, in concert with common comorbidities (such as dyslipidemia and smoking) and, to a lesser extent, off-target effects of antiretroviral therapy. In this review, we discuss the clinical and mechanistic evidence surrounding heightened CVD risks among PWH, implications for specific CVD manifestations, and practical guidance for management in the setting of evolving data.

INTRODUCTION TO HIV-ASSOCIATED CARDIOVASCULAR DISEASES: SCOPE AND CONTEXT

As immediate and continuous antiretroviral therapy (ART) has become the standard of care for people with HIV (PWH), life expectancies among PWH with access to treatment have increased dramatically (1, 2). Concomitant with this increased life expectancy is the emerging epidemic of aging-associated multimorbidity among PWH (3). Cardiovascular diseases (CVDs)—which include coronary artery disease and myocardial infarction (MI), heart failure (HF), arrhythmia and sudden cardiac death, and pulmonary arterial hypertension (PAH), among others—are among the chief drivers of this HIV-associated multimorbidity (46). The burden of CVD morbidity and mortality among PWH is substantial in both relative and absolute terms. People with HIV have significantly higher risks for various CVDs compared with HIV-uninfected individuals; these include 1.5× or higher relative risks for MI, HF, stroke, pulmonary arterial hypertension, and sudden cardiac death (716). In absolute terms, disability-adjusted life years from HIV-associated cardiovascular disease increased from 0.74 million in 1990 to 2.57 million in 2015, while CVD-related mortality (as a proportion of overall causes of death among PWH) more than doubled (17). Several factors contribute to the growing and excess burden of CVD among PWH, ranging from chronic immune dysregulation and unresolving inflammation, coinciding “traditional” cardiovascular risk factors such as cigarette smoking and dyslipidemia, and off-target effects of certain antiretrovirals, among other factors (5). In this review, we will discuss the pathogenesis and scope of HIV-associated CVDs, with the goal of elucidating immunologic, clinical, and social and environmental factors that shape emerging CVD risks among PWH. We will also provide practical guidance on CVD prevention and management for PWH based on existing and emerging data.

PERSISTENT INFLAMMATION IN HIV: A NIDUS FOR CARDIOVASCULAR DISEASES

HIV is a retrovirus which infects and destroys immune cells, fundamentally altering host immune responses in a way that persists even after viral suppression to undetectable levels (18, 19). The natural history of HIV typically includes an acute phase of viral replication, a chronic phase of immune activation and continued viral replication, and, left untreated, an eventual decline in CD4+ T cells (referred to hereafter as “CD4 cells” or “CD4” when discussing levels) leading to AIDS (20), marked by opportunistic infections and high mortality (18, 21). Heterogeneity exists in viral suppression and progression of CD4 decline among untreated PWH: long-term nonprogressors remain asymptomatic with higher (>500 cells/mm3) CD4 counts even without ART (2224), while others (often termed “elite controllers”) can suppress HIV replication to undetectable levels without ART (22). Nevertheless, these individuals comprise a small minority [<1%, by some estimates (25)] of PWH, as the vast majority experience some degree of viremia and continued CD4 cell decline when untreated. Importantly, CD4 cells are not the only targets of HIV, as macrophages are also targets and express surface markers, including CCR5 and CXCR4, through which HIV entry may occur via the viral envelope protein, gp120 (26, 27).
The natural history of HIV fundamentally changed with the genesis and widespread uptake of ART, which suppresses viral replication and limits CD4 decline (19). Landmark clinical trials demonstrating benefits of continuous (28) and early (29) ART, coupled with subsequent observational and interventional studies of rapid ART initiation in various settings (3034), helped establish the current consensus approach of initiating ART as soon as possible after diagnosis (35), ideally within 7 days. The clinical trial-demonstrated benefits of early/immediate ART have also been mirrored in surveillance data: although mortality among PWH remains higher than the general population overall, mortality for PWH with minimal CD4 decline, and consistently undetectable viral loads on ART approaches that of the general population in some settings (6, 3639).
Yet, despite effective viral suppression and CD4 stabilization/recovery on ART, residual immunologic changes favoring a persistently inflammatory phenotype remain for many PWH. PWH on ART with modest CD4 recovery still experience a functional regulatory T cell (Treg) deficit (40, 41), with related impairment in homeostatic/regulatory immune responses and a shift toward more inflammatory immune repertoires. Furthermore, even with modest CD4 recovery in absolute terms, comparative expansion of the CD8+ T cell (referred to hereafter as “CD8 cell” or “CD8” when discussing counts) repertoire is common in PWH; this results from initial increases in CD8 cells coupled with CD4 decline during untreated infection, followed by variable CD4 recovery and CD8 decline after ART initiation (42, 43). Ultimately, despite the effectiveness of ART in aiding CD4 recovery, blunting exuberant CD8 expansion, and normalizing some markers of T cell-associated inflammation (44), a net inflammatory shift in immune cell repertoire persists. Even PWH who achieve undetectable viremia and CD4 recovery to >500 cells/mm3 have permanently altered T cell phenotypes, with a majority of such patients continuing to have “inverted” CD4/CD8 ratios less than 1 (45). This inverted CD4/CD8 ratio—so called because CD4/CD8 ratios are commonly 1.5–2.5 in the general population (46, 47)—reflects a general shift toward a T cell repertoire marked by excess activation as well as, ultimately, exhaustion and senescence among PWH (4853). Concomitant with this shift in T cell phenotype is a shift toward activated, inflammatory monocytes and macrophages among PWH (54, 55), which does not normalize with ART (44).
These shifts in host immune repertoire dovetail with increased gut microbial translocation and reactivation of latent viruses [including but not limited to cytomegalovirus (CMV)] to contribute to a net inflammatory state among PWH, even in the setting of sustained viral suppression (40, 5659). Unsurprisingly, PWH have elevated circulating markers of immune activation and inflammation compared with HIV-uninfected individuals, including but not limited to interleukin-6 (IL-6), high-sensitivity C-reactive protein (hsCRP), soluble CD14 and CD163 (sCD14 and sCD163), and lipopolysaccharide; importantly, several of these markers independently associate with CVD and overall mortality among PWH (44, 6064). Next, we will discuss the biology and scope of CVDs in PWH, with particular attention to ways in which chronic HIV-related immune dysregulation and inflammation contribute.

ATHERO-THROMBOTIC DISEASE IN HIV

Cholesterol, chronic inflammation, comorbidity, and clots: biological underpinnings of HIV-associated atherosclerosis and thrombosis

Inflammation is a cause and byproduct of atherosclerosis, thrombosis, and myocardial dysfunction and plays central roles in resulting clinical presentations, including MI, stroke, HF, and sudden cardiac death (6589). Atherosclerosis broadly results from accumulation of plaque due to excess retained lipids within the arterial wall, net-inflammatory immune response to these retained products, and vascular injury (hemodynamic and otherwise) (9093). Eventually, this plaque may erode or rupture, leading to thrombus formation and possible occlusion of the artery (70, 76, 78). Coronary artery occlusion, in turn, results in ischemia, acute MI, and/or sudden cardiac death (94, 95). The pathogenesis of HIV-associated atherosclerosis is incompletely described, and data are based mainly on clinical studies and a limited number of experimental studies. It is likely that HIV-mediated inflammation and dyslipidemia contribute to the severity of HIV-associated atherosclerosis, while the pathogenesis itself is largely the same as in uninfected individuals. The first step of atherogenesis remains the same: endothelial dysfunction which leads to immune activation (96, 97). This inflammation in the endothelium, which is largely mediated by IL-6 and monocyte chemoattractant protein-1 (MCP-1), two proteins commonly upregulated in HIV infection (97, 98), leads to the recruitment, migration, and transformation of macrophages into foam cells (9698). Macrophages both harbor the HIV virus and are impacted by the HIV Nef protein which impairs cholesterol efflux from the cell (99), possibly accelerating foam cell formation and influencing HIV atherogenesis. Oxidative stress, activation of the NLRP3 inflammasome, and endoplasmic reticulum (ER) stress also contribute to atherogenesis (9698). ER stress may be especially impacted by HIV infection due to the associated dyslipidemia and release of HIV proteins, such as gp120, which trigger the ER stress response and associated macrophage apoptosis (96, 98).
Dysregulated lipid profiles are common in PWH and often characterized by decreased high-density lipoprotein cholesterol (HDL-c) levels and elevated triglyceride levels in both treated and untreated PWH (100). HIV disrupts the function of HDL (5) (101, 102), and ART does not revert HDL levels back to pre-infection baseline (101), an important consideration, given the well-described atheroprotective role of functional HDL (103). Effects of ART on lipids also vary by class and specific drug. Protease inhibitors (PIs; particularly earlier generation protease inhibitors) increase total cholesterol, low-density lipoprotein cholesterol (LDL-C), and triglyceride levels, whereas effects of nucleoside and non-nucleoside reverse transcriptase inhibitors (NRTIs and NNRTIs) are more variable (104106). For instance, a recent study of over 6,000 PWH switching from tenofovir disoproxil fumarate (TDF) to tenofovir alafenamide (TAF) demonstrated significant increases in LDL-C and triglyceride levels over approximately 1 year (107). Integrase inhibitors appear to be largely lipid neutral compared with other antiretrovirals aside from protease inhibitors (108), but data are limited. In addition to changes in circulating lipids, fat distribution likewise changes both as a result of HIV and ART, resulting in redistribution syndromes marked by inflammatory visceral adiposity and varying degrees of peripheral lipodystrophy (109, 110).
Given the well-described interplay between innate immune activation and thrombosis (7882), it is not surprising that PWH exhibit both hyper-inflammatory and hyper-thrombotic profiles. Monocyte activation persists after viral suppression on ART, with hyper-inflammatory tissue factor-expressing monocytes identified as particularly harmful promoters of a thrombo-inflammatory milieu among PWH (111, 112); markers of both monocyte activation and thrombosis have likewise been associated with CVD and death in PWH (61, 113). Although markers of thrombo-inflammation appear elevated among PWH, more research is needed to delineate underlying HIV- and ART-relevant contributors as well as potential strategies to ameliorate HIV-related thrombo-inflammation (114).
In addition to dyslipidemia and pro-thrombotic features, as well as chronic immune dysregulation, concomitant cardiovascular risk factors contribute to atherogenesis and thrombosis in PWH. The global prevalence of hypertension in PWH is approximately 20%–25%, with some variability depending on setting (e.g., North America or sub-Saharan Africa) (115121). A complex interplay between unresolving inflammation, dyslipidemia (and associated atherosclerosis), and chronic kidney disease all may contribute to hypertension among PWH (122125). In addition, men with HIV on ART were found to have a fourfold increase in developing diabetes mellitus (DM) compared to men without HIV in the Multicenter AIDS Cohort Study (126). Furthermore, studies of women with HIV observed a higher risk for developing DM among women with HIV compared with men with HIV, suggesting a sex-specific risk for type 2 diabetes and HIV infection (127). This elevated DM risk among people with HIV has clear implications for cardiovascular diseases; in PWH, the risk of MI is more than doubled for those with DM than for those without DM (128). It is important to note, however, that the observed elevated risk of DM among men and women with HIV compared with people without HIV has not been universal across cohorts and may depend on both HIV and control populations studied, as several cross-sectional studies observed similar prevalence of DM for people with HIV on ART and the general population (129131).
Cigarette smoking is especially common among PWH and strongly associated with athero-thrombotic complications such as MI and stroke. Although the prevalence of current smoking in US adults declined from 20.6% to 16.8% between 2009 and 2014, these numbers were 37.6%–33.6% for PWH (132). In men with HIV, current cigarette smoking was positively associated with subclinical atherosclerosis and increased risk for MI (133). Compared to men with HIV who do not smoke, those who do smoke are estimated to have twice the risk of having a major cardiovascular event (134). Alcohol use is also common in PWH, and hazardous drinking, defined as binge drinking or more than 14 drinks per week, is associated with an increased incidence of CVD in men with HIV as determined by the Veterans Aging Cohort Study (VACS) (135). This association between hazardous drinking alcohol abuse or dependence and CVD persists after accounting for traditional CVD risk factors, CD4 count, and adherence to ART (135). Moderate drinking and other substance use, including cocaine, did not significantly increase the risk for developing HF in the VACS analysis (13). A conceptual model of HIV-associated immune dysregulation and coinciding contributors to CVD risk is included in Fig. 1.
Fig 1
Fig 1 Immunologic and clinical contributors to HIV-associated cardiovascular diseases.

Subclinical arterial disease

Given this inflammatory and metabolically dysregulated milieu, it is not surprising that PWH exhibit a higher prevalence of subclinical arterial disease as well as a more inflammatory arterial disease phenotype compared with HIV-uninfected controls. Compared with HIV-uninfected individuals, PWH have a higher prevalence and greater extent of noncalcified, high-risk coronary plaque and increased coronary artery adverse remodeling—clinically validated markers of increased MI risk that correlate with monocyte activation markers (136142). Interestingly, a recent analysis investigating associations of a large panel of inflammation-associated proteins with calcified and overall coronary plaque [as determined by computed tomography (CT)] suggested unique potential pathways underlying noncalcified versus calcified plaque presence in PWH (143).
Data investigating associations of systemic and vascular inflammation with coronary plaque among PWH are not limited to studies of blood-based biomarkers. Indeed, one study investigating coronary plaque via CT among 41 PWH also employed functional positron emission tomography (PET) imaging of the aorta to measure inflammation and revealed a significant association between aortic inflammation and high-risk coronary plaque (140). While this association was observed within PWH, data vary on whether PWH have significantly more arterial inflammation than HIV-uninfected controls. One study of 153 young to middle-aged adult (aged 18–40) PWH and 153 age- and gender-matched controls revealed higher target-to-background ratio—a marker of arterial inflammation on PET—as well as high-sensitivity C-reactive protein among PWH (144). Meanwhile, a smaller study of 26 PWH and 25 HIV-uninfected controls revealed no difference in arterial inflammation measured by PET, although controls were 9 years younger (on average) than PWH in this study (145).
In addition to CT-based measurements of coronary plaque, numerous studies have investigated carotid intima-media thickness and plaque on ultrasound—direct markers of carotid disease and proxy markers of systemic and coronary arterial disease—in PWH and controls. These likewise demonstrated a higher prevalence of carotid intima-media thickness, plaque, and high-risk features among PWH, as well as associations between elevated indices of inflammation and arterial disease in PWH (141, 146148). A recent systematic review of studies investigating subclinical cardiovascular imaging among PWH, which included several of the above-mentioned studies as well as others with variable risk of bias, revealed generally higher indices of subclinical arterial plaque and inflammation among PWH versus HIV-uninfected controls (149). Importantly, a high degree of heterogeneity across studies was noted, highlighting the non-uniformity of populations of PWH as well as controls that may be selected for such studies as well as the potential impact of variable control selection on findings related to HIV-associated subclinical disease burden (150).

Clinical presentation and HIV-specific CVD risk enhancers

Clinically, PWH have approximately 1.5- to 2-fold higher risks for MI compared with HIV-uninfected individuals, a risk that persists after accounting for common risk factors and confounders. These elevated MI risks were noted in the early ART era and have persisted; furthermore, a biologic gradient exists whereby lower CD4 count is strongly associated with MI among PWH (151157). Additionally, higher viral loads are associated with endothelial dysfunction, inflammation, and the pathogenesis of atherosclerosis (158161). Such associations of viremia and CD4 decline with MI risk likely reflect putative mechanisms discussed above, ranging from net-activated/inflammatory T cell and monocyte repertoires to concomitant dyslipidemia and thrombosis (5). Interestingly, HIV-associated excess MI (and overall CVD) risk may be especially high among women with HIV (156, 162), although underlying mechanisms are incompletely defined, as are CVD risks in transgender and non-binary PWH. In addition to MI, ischemic stroke also appears to be more common among PWH than in the general population (8, 163, 164), although this comparative excess risk has not been universally observed across cohorts (165).
Yet, for PWH and providers engaged in their care, broad comparisons of MI or stroke risk by HIV serostatus are less practically informative than understanding HIV-specific factors that may be driving CVD risk at an individual level (150). Among PWH, sequelae of uncontrolled virus and immune progression—in particular, low (<200 or even <350 cells/mm3) CD4 and histories of prolonged viremia—have been consistently associated with higher MI (151157) and stroke risk (8, 163165) and are thus considered HIV-specific CVD risk enhancers (5). The HIV Outpatient Cohort Study found that recent (as opposed to nadir) CD4 counts of <500 cells/mm3 were associated with a 20% attributable risk for incident CVD and could be considered as an independent risk factor for CVD (153). Additionally, a lower CD4/CD8 ratio is independently associated with an increased risk for coronary atherosclerosis (166). Other HIV-specific CVD risk enhancers, which have likewise been associated with heightened CVD risk among PWH and reflect inflammatory pathophysiologies discussed above, include co-infection with hepatitis C virus and HIV-associated metabolic sequelae such as lipodystrophy (5). Most PWH (90% or higher in some cohorts) are co-infected with CMV (167). CMV co-infection exacerbates non-AIDS comorbidities, especially CVD and cerebrovascular events (167, 168). In HIV-uninfected CMV-infected populations, the Sacramento Area Latino Study on Aging found an association between CMV IgG levels and ischemic heart disease and increased carotid artery stiffness (169). In PWH, CMV IgG levels do not necessarily correlate with CMV replication, highlighting the important role of immune activation (169). Approximately 10% of CD4 and CD8 cells are targeted toward CMV in infected individuals, and this percentage may increase as the individual ages (169). It is hypothesized that this robust response toward CMV infection may decrease the available immune cells to respond to other infections, further stressing the immune system in PWH.

Now what? practical insights for prevention and management

For CVD primary prevention—prevention of CVD before its overt clinical onset—assessment of absolute risks is required to inform the net clinical benefit [absolute risk reduction minus absolute adverse event rate (170)] of potential CVD preventive interventions. For example, if medication A was expected to broadly achieve a relative reduction in CVD events by one-third in most populations, patient X with a 10-year predicted CVD risk of 30% would be expected to achieve a 10% absolute reduction in CVD risk (down to 20%) with medication A; for patient Y with a 10-year predicted risk of only 3%, this absolute risk reduction would be only 1%. Therefore, if medication A confers a consistent 2-5% absolute risk for serious adverse effects, the net clinical benefit of medication A would be more readily justified in patient X than patient Y, for whom adverse effects would be more likely than preventing a CVD event.
In this context, there has been considerable interest in precisely estimating CVD risk among PWH. Unfortunately, general population and HIV-specific CVD risk estimators exhibit modest at-best predictive utility in assessing CVD risk for PWH (171174), with particularly poor performance in women, Black individuals, and younger populations (175). In the absence of optimally calibrated CVD risk estimation tools for PWH, a reasonable interim approach is to use an existing CVD population risk estimator (e.g., the American Heart Association/American College of Cardiology pooled cohort equations or Framingham Risk Score, among others), with the understanding that in the presence of HIV-specific risk enhancers, an individual’s actual CVD risk is likely 1.5-fold or more higher than predicted (5). As discussed above, HIV-related risk enhancers include clinical features such as low current or nadir CD4 count (<200 or <350 cells/mm3), history of prolonged viremia and/or treatment failure, and co-infection with hepatitis C virus, among others. Whether circulating inflammatory markers such as interleukin-6 or soluble CD163 meaningfully improve risk stratification remains to be determined.
A separate question relates to the role of subclinical atherosclerotic cardiovascular disease (ASCVD) imaging—for instance, coronary artery calcium (CAC) screening—for further risk stratification in PWH. In the general population, CAC screening is a widely accepted, relatively inexpensive, and low-radiation way to evaluate calcified coronary plaque burden (which generally, though imperfectly, correlates with overall plaque burden) and thus stratify individuals’ risk for progression to clinical ASCVD such as MI (176). Several investigations adding CAC scores to clinical risk prediction models have revealed CAC to be an important—in some cases, the most important—predictor of progression to clinical events, supporting its widespread use for risk stratification (176178). While these general population data argue in favor of potential value for CVD risk stratification in PWH, there are some caveats. As discussed above, subclinical arterial disease may present with a somewhat different phenotype in PWH than in HIV-uninfected persons, with PWH having a comparative predilection for noncalcified plaque. Given these data, CAC screening in PWH may be useful insofar as observation of significant CAC [e.g., absolute CAC score >400 Agatston units, and/or at a high percentile compared to what would be age- and demographically expected based on normative cohort data (179)] is clearly indicative of plaque burden and places that individual in a higher risk group than would have been expected pretest. Meanwhile, a lower-than-expected CAC score or zero CAC, while somewhat reassuring, may not be quite as reassuring in PWH, given noncalcified plaque may still be present.
The predicted CVD risk for PWH can inform providers regarding expected net clinical benefits of established CVD preventive therapies—in particular, lipid-lowering therapies such as statins. To this end, the recently completed randomized trial to prevent vascular events in HIV (REPRIEVE), which demonstrated a higher-than-expected 35% risk reduction in major cardiac events for PWH randomized to a moderate intensity statin (pitavastatin) and similarly low serious adverse event rates as in the general population, provides timely data to inform these individual-level risk/benefit considerations (180). Non-statin lipid-lowering therapies are also under investigation in PWH, with a recent trial of a proprotein convertase subtilisin/kexin type 9-inhibiting monoclonal antibody demonstrating lipid-lowering efficacy and a tolerable side effect profile in PWH (181). The Evolocumab Effect on LDL-C Lowering in Subjects with Human Immunodeficiency Virus and Increased Cardiovascular Risk (BEIJERINCK) trial also found monthly evolocumab injections significantly reduced LDL-C levels in PWH with high cardiovascular risk. The LDL-lowering effects of omega-3 fish oil and fenofibrate in PWH still require more elucidation, with two small studies showing insignificant differences and mild improvement for each respective therapy (182, 183). An overview of clinical trials evaluating cardiovascular endpoints in PWH is provided in Table 1.
TABLE 1
TABLE 1 Randomized controlled trials evaluating cardiovascular endpoints in people with HIV
TrialNumber of participantsIntervention/observationPrimary outcome and effect sizeSecondary endpoints
OPERA Cohort (2018)6,451 PWH, at least 18 years of age, with at least 4 weeks of TDF use and direct switch from TDF to TAF between 5 September 2015 and 31 March 2018Switched from TDF to TAF, 4,328 PWH maintained their other agentsLDL-C increased by 1.40 mg/dL/mo over first 3 months on TAF and 0.33 mg/dL/mo between 3 and 9 months before plateauing. TG increased by 3.52 mg/dL/mo over first 3 months on TAF and 0.91 mg/mL/mo between 3 and 9 months.Weight increased 0.25 kg/mo over first 3 months and 0.10 kg/mo between 3 and 9 months.
Swiss HIV Cohort (2021) TDF versus TAF (184)4,375 PWH who received TDF-containing ART for at least 6 monthsSwitch therapy to TAF or continue TDF-based regimenSwitching to TAF was associated with adjusted mean weight increase of 1.7 kg after 18 months compared to 0.7 kg in the TDF group. 13.8% of the TAF group became overweight/obese compared to 8.4% of the TDF group.Switching to TAF led to an increased adjusted mean total cholesterol of 9.5 mg/dL, HDL-C of 1.9 mg/dL, LDL-C of 4.7 mg/dL, and TG of 16.1 mg/dL after 19 months.
SMARTa (2009) (6, 185, 186)5,472 PWH with CD4 counts >350 cells/µLIntermittent ART until CD4 count <250 cells/ µL or continuous ARTThe opportunistic disease/death hazard ratio (HR) was 3.5 for the drug conservation (DC) versus viral suppression (VS) groups.The hazard ratio for serious non-AIDS events was 7.0 for the DC versus VS groups.
START(29)4,658 PWH with a starting CD4 count of >500 cells/mm3Immediate initiation of ART or deferred treatment until CD4 count fell to 350 cells/mm3 or development of AIDSPrimary outcome of composite of serious AIDS-related event and serious non-AIDS-related event. 1.8% of the immediate initiation group compared to 4.1% of the deferred initiation group reached the primary endpoint. Hazard ratio of 0.43.Secondary endpoints included serious AIDS-related events, serious non-AIDS-related events, death from any cause, grade 4 events, and unscheduled hospitalizations for reasons other than AIDS. HR of 0.28 for serious AIDS-related event, 0.61 for serious non-AIDS-related events, and 0.58 for death from any cause.
BEIJERNIK(181)464 PWH with hypercholesterolemia/mixed hyperlipidemia on statin therapyMonthly subcutaneous injection of evolocumab 420 mg or placeboPrimary endpoint of percent change in LDL-C. Placebo-corrected mean percent change in LDL-C of −56.9% with evolocumab treatment.Secondary endpoints of LDL-C <70 mg/dL and percent change in other plasma lipids and lipoprotein levels. LDL-C level <70 mg/dL achieved in 73.3% in evolocumab group compared to 7.9% in placebo group.
REPRIEVE (2023) (143, 187)7,769 PWH between the ages of 40 and 75 years with low-to-moderate CVD risk on ART4 mg of daily pitavastatin calcium or placeboDaily statin use lowered risk of major adverse cardiovascular events by 35% compared to placebo group.
Major adverse cardiovascular event defined as composite of cardiovascular death, myocardial infarction, hospitalization for unstable angina, stroke, transient ischemic attack, peripheral arterial ischemia, revascularization, or death from undetermined cause.
Secondary outcomes of composite of a major adverse cardiovascular event or death from any cause, individual components of the primary outcome, death from any cause, and LDL and non-HDL cholesterol, among others. The incidence of major adverse cardiovascular events or death from any cause was 9.18 per 1,000 person-years in the pitavastatin group compared to 11.63 per 1,000 person-years in the placebo group (HR, 0.79). There were deaths from noncardiovascular causes in 82 in the pitavastatin group compared to 81 in the placebo group, and nonfatal heart-failure events in 13 of the pitavastatin group compared to 12 in the placebo group. The LDL-C levels decreased from a median of 107 to 74 mg/dL in the pitavastatin group and from 106 to 105 mg/dL in the placebo group.
a
SMART, Strategies for Management of Antiretroviral Therapy.
Fewer data exist to inform on other antithrombotic and anti-inflammatory therapies for CVD prevention in PWH. Aspirin exhibited muted antiplatelet effects in PWH compared with alternative antiplatelet agents, such as clopidogrel (188190), but the clinical relevance of these findings and comparative risk versus benefit of antiplatelet for primary prevention of CVD in PWH remain to be seen. Similarly, the role for targeted anti-inflammatory therapies, such as canakinumab, for CVD prevention remains unclear in PWH but is under investigation (191). The Antiinflamatory Therapy with Canakinumab for Atherosclerotic Disease (CANTOS) trial showed that canakinumab, a monoclonal antibody, treatment was helpful in reducing major adverse cardiovascular events in patients without HIV who achieved hsCRP levels less than 2 mg/L with the first dose (192194). Canakinumab has major effects on inflammation as shown by circulating inflammatory markers but very little effect on lipid levels, especially LDL (192, 193). However, the study did not include PLWH, leaving limited anti-inflammatory recommendations for this population. Similarly, the Colchicine Cardiovascular Outcomes Trial (COLCOT) and the second low-dose colchicine trial (LODOCO2) found that 0.5 mg of colchicine daily significantly decreased the risk of cardiovascular events in patients with recent MI and patients with chronic coronary disease, respectively (195, 196). Neither trial collected data on PWH. An overview of clinical trials evaluating inflammation-targeted therapies in the general population is provided in Table 2.
TABLE 2
TABLE 2 Randomized controlled trials evaluating inflammation targeted for cardiovascular disease in the general population
TrialNumber of participantsIntervention/observationPrimary outcome and effect sizeSecondary endpoints
CANTOS (2019) (193, 194)10,061 patients with a history of MI50, 150, or 300 mg of canakinumab or placebo subcutaneously every 3 monthsParticipants in the canakinumab treatment group who achieved IL-6 levels below 1.65 ng/L had a 32% reduction in major adverse cardiovascular events (MACE), a 30% reduction in MACE plus hospitalizations for unstable angina requiring urgent revascularization, a 52% reduction in cardiovascular mortality, and a 48% reduction in all-cause mortality compared to the placebo group.Participants in the canakinumab group who did not achieve IL-6 levels below 1.65 ng/L did not have any significant benefits for these endpoints.
LODOCO2 (195)5,522 patients between the ages of 35–82 years with evidence of coronary disease0.5 mg of colchicine oncedaily or placeboPrimary endpoint composite of cardiovascular death, spontaneous MI, ischemic stroke, or ischemia-driven coronary revascularization.
The primary endpoint occurred in 6.8% of the colchicine group and 9.6% of the placebo group.
A key secondary endpoint occurred in 4.2% of the colchicine group and 5.7% of the placebo group.
CIRT (197)4,786 patients with previous MI or multivessel coronary disease who also had type 2 diabetes or metabolic syndrome15 mg of methotrexate once weekly with increase to 20 mg at 4 months or matching placebo. All participants received 1 mg of oral folic acid dailyPrimary endpoint of first occurrence of a major adverse cardiovascular event defined as a composite of nonfatal MI, nonfatal stroke, or cardiovascular death. Hospitalization for unstable angina that led to urgent revascularization was added to the primary endpoint. This final primary endpoint occurred in 201 patients in the methotrexate group and 207 in the placebo group [hazard ratio (HR), 0.96]. The original primary endpoint occurred in 170 patients in the methotrexate group and 167 in the placebo group (HR, 1.01).Secondary endpoints of death from any cause, a composite of major adverse cardiovascular events plus coronary revascularization, hospitalization for congestive heart failure. There were no significant differences between the groups.
COLCOT (2019) (196)4,745 patients with prior MI within 30 days of trial0.5 mg once daily colchicine or placeboPrimary endpoint of composite of death from cardiovascular causes, resuscitated cardiac arrest, myocardial infarction, stroke, or urgent hospitalization for angina leading to coronary revascularization. Primary endpoint occurred in 5.5% of colchicine group compared to 7.1% of placebo group. Hazard ratios of 0.84 for death from cardiovascular causes, 0.83 for resuscitated cardiac arrest, 0.91 for myocardial infarction, 0.26 for stroke, and 0.50 for urgent hospitalization for angina needing coronary revascularization.Diarrhea reported in 9.7% of colchicine group and 8.9% of placebo group. Pneumonia reported in 0.9% of colchicine group and 0.4% of placebo group.
Multiple studies have shown that low-to-moderate dose methotrexate treatment in patients with rheumatoid arthritis lowered their CVD risk by modulating inflammation (198202), but a recent study of low-dose methotrexate showed that the drug had no significant effect on endothelial function or inflammatory markers in PWH (203). The treatment did significantly lower CD8 cells, and a subsequent study showed that the methotrexate inhibited T cell proliferation (5, 203, 204). Ultimately, while there is ongoing research on anti-inflammatory therapies and cardiovascular risk, there is an absence of data applying to PWH.
PWH also warrant different secondary prevention considerations. Although PWH have similar rates of complication following percutaneous coronary intervention compared with HIV-uninfected individuals (205), they are less likely to undergo percutaneous coronary intervention following MI. This gap in indicated coronary interventions may be especially pronounced among women (206, 207). Rates of appropriate, indicated lipid-lowering and antiplatelet therapy following athero-thrombotic CVD events (in particular, MI) likewise lag for PWH compared with HIV-uninfected individuals (208210). Given suboptimal post-MI care and a high burden of mortality post-MI among PWH (211), HIV care providers must remain vigilant to ensure patients with existing CVD receive appropriate, indicated therapies to prevent recurrent events.

ART-specific considerations in ASCVD: past, present, and future

ART has transformed the natural history of HIV, and evolutions in ART have likewise impacted the natural history of HIV-associated aging and comorbidity. Several landmark trials elucidated the benefits of long-term antiretroviral (ARV) use, as well as the importance of early and continuous ART. The Strategies for Management of Antiretroviral Therapy trial demonstrated the significant benefit of continuous ART over episodic use (185), even with the side effects of early antiretrovirals; in addition to improving HIV/AIDS-related outcomes, continuous ART actually reduced risk for myocardial infarction. Subsequently, the START (initiation of antiretroviral therapy in early asymptomatic HIV infection) demonstrated a clear clinical benefit of immediate and continuous ART initiation (29), though without a significant reduction in cardiovascular events with immediate ART. Thus, although one might extrapolate a reduction in overall cardiovascular risk with immediate ART based on the potential to reduce viremia and early CD4 decline, the clinical trial data from START do not resoundingly indicate cardiovascular-specific benefit of immediate ART.
In the current era, there are seven classes of antiretroviral drugs approved by the Food and Drug Administration: NRTIs, NNRTIs, PIs, INSTIs (integrase strand transfer inhibitors), fusion inhibitors, CCR5 antagonists, and post-attachment inhibitors. People with HIV are commonly on three drugs from two different classes, although two-drug regimens are becoming increasingly common. The 2022 recommendations from the International Antiviral Society, USA Panel for preferred drug regimens pair one INSTI with one or more NRTIs (35). The formulations of antiretroviral drugs have changed from their first iterations to modern usages. Older variations of NRTIs, such as azidothymidine, were associated with higher mitochondrial toxicity leading to myopathies and neuropathies (212, 213) As mentioned previously, TDF and TAF have varying effects on lipid profiles (107). While more studies are needed to conclude their effect on CVD risk, it is possible that TDF-based regimens have positive effects on lipid profiles and, therefore, lessen CVD risk compared to TAF-based regimens. Despite the cardiometabolic conditions associated with INSTIs, a study based on the Swiss HIV Cohort Study just concluded that there is no difference in short-term or long-term risk for CVD in treatment-naïve PWH who start an INSTI-based ART compared to those who start another ART (184).
Overall, drug-/class-specific data are too inconsistent to inform a blanket recommendation for a particular drug or class as preferable to others. Protease inhibitors have been associated with modest increases in MI risk (214), but these findings do not appear to be a universal class effect: among protease inhibitors, ritonavir-boosted darunavir is associated with increased CVD risk, whereas ritonavir-boosted atazanavir is not (215). Meanwhile, abacavir has been associated with elevated MI risk in some (214, 216221) but not all studies (222225). Therefore, while ART is clearly preferable to no ART for control of HIV as well as blunting-related immunologic changes driving persistent inflammation among PWH, there is currently no single, clearly superior “CVD-friendly” ART regimen to recommend above others. In the presence of boosters, it is important for clinicians to likewise understand potential effects on other medications, as boosters such as cobicistat and ritonavir can substantially increase levels of other drugs metabolized by the cytochrome P450 system, including statins (105). An overview of statin-related considerations for PWH on ART is provided in Table 3.
TABLE 3
TABLE 3 Statin dosing-related considerations for people with HIV on ART
StatinIntensityMetabolism (226229)ARV-specific data
AtorvastatinHigh (40 or 80 mg daily dose)
Lower doses considered moderate (226, 230232)
CYP3A4
Pgp
OATP1B1
Interactions with PIs and NNRTIs which may require statin dose adjustments. Interacts with boosters including cobicistat given CYP3A4 metabolism; in these cases, atorvastatin downward dose adjustment recommended (105, 233239).
PitavastatinModerate (2–4 mg) (226)Minimally metabolized (240)No significant or severe noted interactions with antiretrovirals (105, 227, 233, 238, 241).
LovastatinModerate (40 mg) (226, 242, 243)CYP3A4
Pgp
Interactions with PIs and NNRTIs which may contraindicate use. Rarely used in modern era due to superior efficacy/safety profiles of other statins (105, 233, 244).
RosuvastatinHigh (20–40 mg) (226, 245, 246)
Lower doses considered moderate
CYP2C9
OATP1B1
OATP1B3
Interactions with some PIs which may require downward dose adjustment (105, 233, 238, 241, 245).
SimvastatinModerate (20–40 mg) (226, 231, 247, 248)CYP3A4
Pgp
OATP1B1
Heavily metabolized by CYP3A4 system; strong interactions with PIs and NNRTIs which may contraindicate use. Rarely used in modern era due to superior safety profiles of alternative moderate-intensity statins (pravastatin, pitavastatin, and atorvastatin or rosuvastatin at moderate intensity) (105, 233235, 239, 244).
PravastatinModerate (40–80 mg) (226, 249)OATP1B1
OATP1B3
Interactions with PIs and NNRTIs which may require modest downward pravastatin dose adjustments (105, 233235, 237, 250, 251).
FluvastatinModerate (80 mg) (226, 252)CYP2C9
OATP1B3
Interactions with PIs which may require modest downward fluvastatin dose adjustments (105, 233, 253, 254)

OTHER CARDIOVASCULAR MANIFESTATIONS OF HIV: AN INCOMPLETE PICTURE

As discussed above, atherosclerosis has a largely stereotyped pathogenesis and presentation, which, despite some heterogeneity (255), result from a finite and largely predictable combination of triggers. Meanwhile, other CVDs for which PWH are at heightened risk—in particular, HF and fatal arrhythmia/sudden cardiac death—represent often later-stage sequelae of diverse CVD pathologies. Accordingly, mechanisms of HIV-associated HF, for instance, are challenging to define in a singular manner, as are HIV-specific preventive and therapeutic approaches. These challenges mirror those observed in the general population, where heterogeneity of HF mechanisms and manifestations has spurred more specific phenotype- and etiology-focused interventions (256, 257).

Heart failure in HIV: evolution of a final common pathway

Heart failure is a complex clinical syndrome with varying manifestations, resulting from underlying myocardial tissue abnormalities coupled with extracardiac contributors such as neurohormonal activation, pulmonary and renal disease, and morbid obesity (258). Given this diversity of contributors and manifestations, the nature of HIV-associated HF has unsurprisingly evolved as the natural history of HIV has changed (158). In the pre-ART era, PWH commonly experienced overt severe systolic dysfunction, resulting, in many cases, in HF with reduced ejection fraction (HFrEF) (259)—a manifestation particularly common among PWH with high viremia, overt immunodeficiency, and AIDS (259262). In the modern ART era, rates of severe systolic dysfunction have declined, but systolic dysfunction remains common among PWH as does, increasingly, diastolic dysfunction (a common underlying contributor to HF with preserved ejection fraction, HFpEF) (5, 259, 263). These underlying abnormalities in myocardial function predispose to HF, for which PWH have 1.5- to 2-fold higher risk than HIV-uninfected individuals in the modern ART era, even after adjustment for clinical and other confounders (12, 13). In both of these studies investigating HF risk, conducted in distinct cohorts and with distinct HF ascertainment methods, PWH with higher viral load and lower CD4 counts were at especially elevated risk—mirroring the elevated risk of MI and stroke observed among PWH with viremia and/or low CD4.
Although the scope and presentation (e.g., HFrEF versus HFpEF) are increasingly defined among PWH, several major questions remain. Little data from the modern ART exist on specific etiologies of HF (e.g., post-MI damage versus valvular origin versus stimulant-induced HF, among other potential causes) among PWH. This represents a major gap, given the vast diversity of potential HF etiologies (264). As a result of this gap, biologic insights into HIV-associated HF have depended largely on small imaging studies investigating intermediate subclinical manifestations on cardiac magnetic resonance imaging (CMRI) of myocardial disease, such as fibrosis and mechanical dysfunction. These studies suggest that PWH have more myocardial inflammation/edema, fibrosis, steatosis, and mechanical dysfunction than HIV-uninfected controls (265267). The myocardial substrate may also be especially vulnerable to ischemia and damage following ischemia/MI among PWH (268, 269); one study observed twice the extent of myocardial fibrosis following MI for PWH versus controls with similar coronary artery disease extent and MI culprit lesions (269), with the result being more myocardial area at risk and resulting dysfunction during ischemic events. Nevertheless, despite these interesting data, the optimal role for cardiac imaging—whether with echocardiography, CMRI, and/or computed tomography—in PWH without existing cardiac symptoms is unclear. At a minimum, it is important that clinicians maintain a high index of suspicion for coronary ischemia and/or myocardial dysfunction in their PWH presenting with potential cardiopulmonary symptoms (in particular, chest pain or shortness of breath), with subsequent symptom-directed imaging studies informing early diagnosis and treatment.
Despite these emerging data, limited HF etiology-specific knowledge among PWH limits HIV-specific insights into HF prevention and treatment. For HIV care providers, understanding that PWH have heightened HF risk—particularly in the setting of current or historical sustained viremia and/or CD4 decline—is an essential first step that should raise the index of suspicion for HF in PWH experiencing potential HF symptoms such as dyspnea or lower extremity edema. In 2013, The HIV Organ Policy Equity Act allowed for HIV-infected patients to receive organ transplants from HIV-infected donors in order to further research (270). While most data come from kidney and liver transplants, there are studies to show the 5-year survival rate for PWH receiving heart transplants is similar to that of the HIV-uninfected population (270). Similar studies have shown the effectiveness of ventricular assist devices (VAD) in PWH suffering from advanced HF, even in the setting of higher rates of preimplant comorbidities than the HIV-uninfected population (271). Access to both heart transplants and VAD is currently limited for PWH due to persisting views that HIV infection is a contraindication for these interventions (272, 273). In the coming years, as more HF etiology-specific knowledge emerges, more targeted insights for HF prevention and therapy among PWH may likewise emerge.

HIV-associated arrhythmias: inconsistent data—except for excess sudden cardiac death

As with HF, cardiac rhythm disturbances (arrhythmias) can result from a diversity of intrinsic myocardial (e.g., tissue fibrosis) and extracardiac (e.g., adrenergic over-activation) stimuli. Arrhythmias likewise vary in their anatomic sites of origin, with implications for clinical severity. Generally speaking, supraventricular arrhythmias—ranging from atrial fibrillation or flutter to atrial tachycardia, among others—have a more benign clinical profile than sustained ventricular arrhythmias such as ventricular tachycardia or fibrillation, the latter of which is usually fatal. Data vary on whether PWH have higher risk for atrial fibrillation compared with controls and may depend on cohort and atrial fibrillation ascertainment methods (274276). Regarding ventricular arrhythmias, PWH do not appear to have elevated risk for isolated (usually benign) ventricular ectopy or nonsustained ventricular tachycardia (277, 278). However, the risk for sudden cardiac death—often resulting from fatal arrhythmias such as sustained ventricular tachycardia degenerating into ventricular fibrillation (279)—is higher among PWH than HIV-uninfected individuals and may be driven by more fibrosis among PWH (11, 14, 280). Whether and how these findings should alter thresholds for arrhythmia screening and preventive therapy (in the case of sudden cardiac death, via implantable cardioverter-defibrillator for very high-risk individuals based on HF and/or arrhythmic history) remains to be seen (281). In the meantime, a reasonable approach is for HIV care providers to maintain a high index of suspicion in the presence of concerning symptoms (particularly syncope or presyncope without prodrome) and a low threshold for electrocardiographic screening and/or referral to a cardiologist. Furthermore, although the role for broad screening of asymptomatic PWH with electrocardiograms is uncertain, a low threshold for screening is supported by (i) the relatively common presence of abnormal baseline electrocardiogram findings in PWH (282, 283) and (ii) the potential value of screening for QT prolongation in the setting of polypharmacy.

HIV and pulmonary arterial hypertension

PAH is one of the earliest described HIV-associated CVD manifestations and remains relevant in the modern era, with several-fold higher prevalence of PAH remaining among PWH compared with HIV-uninfected individuals (15, 16, 284286). HIV-related PAH has a significantly higher prevalence than PAH in the general population and has been well described since the 1990s. The effect of ART on HIV-related PAH remains controversial (284, 287), and the common underlying mechanism is hypothesized to be a combination of vascular injury, pulmonary arteriole remodeling, and increased inflammation (284, 287).

CONCLUSIONS, LIMITATIONS, AND FUTURE DIRECTIONS

People with HIV have higher risks for cardiovascular diseases than people without HIV, and these heightened risks apply to varied manifestations including myocardial infarction, stroke, heart failure, and sudden death. Putative HIV-specific mechanisms for these manifestations differ and are incompletely defined, particularly for heart failure and sudden cardiac death. Furthermore, an important limitation of current data on HIV-associated cardiovascular diseases is that the vast majority come from high-income settings, particularly in North America and Europe. This knowledge gap may have important implications for understanding and curbing cardiovascular pathologies in the global HIV population. For instance, the long-term immunologic effects of perinatal HIV infection—much more common in regions with higher HIV endemicity than North America and Europe—on athero-thrombotic and myocardial disease likely differ dramatically from the cardiovascular effects of HIV acquired at middle age or later. More research is needed to inform on such questions and generate more precise knowledge on how diverse immune phenotypes may differentially drive HIV-associated cardiovascular risk (150).
Despite these knowledge gaps, a common theme is that prolonged viremia and low CD4 count—current or historical—are consistently associated with higher risk for cardiovascular diseases among people with HIV. From a management perspective, an emphasis on immediate and continuous ART to control viremia and assist with CD4 recovery is therefore essential not only for HIV control but for reduction in cardiovascular risk. Recent data also suggest people with HIV receive particular athero-protective benefit from lipid-lowering therapy with statins, which may justify a lower clinical threshold to consider these therapies among PWH when perceived benefit exceeds risk. Considerably, more work is needed to define etiologies and underlying mechanisms of HIV-associated heart failure and sudden cardiac death, with the ultimate goal that these data will inform screening, prevention, and, perhaps, therapy.
Importantly, given the evolving natural history of HIV and available therapies for PWH, even these data will represent an incomplete picture. With increasing availability of long-acting therapies such as lenacapavir (288) and other novel approaches to treating HIV, natural histories of HIV viremia, immune progression, and related systemic and tissue-specific inflammation may evolve, with potential implications for CVD pathogenesis and burden. Of course, factors such as age and duration of infection also vary widely by geographic region; for instance, half of PWH in the United States or more are over age 50, whereas this proportion is considerably lower in sub-Saharan Africa, though expected to increase several-fold in the coming years (289). These considerations ensure that HIV-related CVD pathogenesis and burden will likely evolve in somewhat setting-specific manners in the decades ahead. Careful clinical and mechanistic research as well as surveillance studies will remain essential in the coming decades as HIV-associated cardiovascular diseases continue to evolve.

ACKNOWLEDGMENTS

The work for this review was funded in part by National Institutes of Health awards R01HL156792 and R01HL154862.

REFERENCES

1.
Palella FJJ, Baker RK, Moorman AC, Chmiel JS, Wood KC, Brooks JT, Holmberg SD, Investigators HOS. 2006. Mortality in the highly active antiretroviral therapy era: changing causes of death and disease in the HIV outpatient study. J Acquir Immune Defic Syndr 43:27–34.
2.
Palella FJ, Delaney KM, Moorman AC, Loveless MO, Fuhrer J, Satten GA, Aschman DJ, Holmberg SD. 1998. Declining morbidity and mortality among patients with advanced human immunodeficiency virus infection. N Engl J Med 338:853–860.
3.
Montano M, Oursler KK, Xu K, Sun YV, Marconi VC. 2022. Biological ageing with HIV infection: evaluating the geroscience hypothesis. Lancet Healthy Longev 3:e194–e205.
4.
Ntsekhe M, Baker JV. 2023. Cardiovascular disease among persons living with HIV: new insights into pathogenesis and clinical manifestations in a global context. Circulation 147:83–100.
5.
Feinstein MJ, Hsue PY, Benjamin LA, Bloomfield GS, Currier JS, Freiberg MS, Grinspoon SK, Levin J, Longenecker CT, Post WS. 2019. Characteristics, prevention, and management of cardiovascular disease in people living with HIV: a scientific statement from the American heart Association. Circulation 140:e98–e124.
6.
Rodger AJ, Lodwick R, Schechter M, Deeks S, Amin J, Gilson R, Paredes R, Bakowska E, Engsig FN, Phillips A, INSIGHT SMART, ESPRIT Study Groups. 2013. Mortality in well controlled HIV in the continuous antiretroviral therapy arms of the SMART and ESPRIT trials compared with the general population. AIDS 27:973–979.
7.
Shah ASV, Stelzle D, Lee KK, Beck EJ, Alam S, Clifford S, Longenecker CT, Strachan F, Bagchi S, Whiteley W, Rajagopalan S, Kottilil S, Nair H, Newby DE, McAllister DA, Mills NL. 2018. Global burden of atherosclerotic cardiovascular disease in people living with HIV: systematic review and meta-analysis. Circulation 138:1100–1112.
8.
Chow FC, Regan S, Feske S, Meigs JB, Grinspoon SK, Triant VA. 2012. Comparison of ischemic stroke incidence in HIV-infected and non-HIV-infected patients in a US health care system. J Acquir Immune Defic Syndr 60:351–358.
9.
Triant VA, Meigs JB, Grinspoon SK. 2009. Association of C-reactive protein and HIV infection with acute myocardial infarction. J Acquir Immune Defic Syndr 51:268–273.
10.
Butt AA, Chang CC, Kuller L, Goetz MB, Leaf D, Rimland D, Gibert CL, Oursler KK, Rodriguez-Barradas MC, Lim J, Kazis LE, Gottlieb S, Justice AC, Freiberg MS. 2011. Risk of heart failure with human immunodeficiency virus in the absence of prior diagnosis of coronary heart disease. Arch Intern Med 171:737–743.
11.
Tseng ZH, Secemsky EA, Dowdy D, Vittinghoff E, Moyers B, Wong JK, Havlir DV, Hsue PY. 2012. Sudden cardiac death in patients with human immunodeficiency virus infection. J Am Coll Cardiol 59:1891–1896.
12.
Feinstein MJ, Steverson AB, Ning H, Pawlowski AE, Schneider D, Ahmad FS, Sanders JM, Sinha A, Nance RM, Achenbach CJ, Christopher Delaney JA, Heckbert SR, Shah SJ, Hanna DB, Hsue PY, Bloomfield GS, Longenecker CT, Crane HM, Lloyd-Jones DM. 2018. Adjudicated heart failure in HIV-infected and uninfected men and women. J Am Heart Assoc 7:e009985.
13.
Freiberg MS, Chang C-CH, Skanderson M, Patterson OV, DuVall SL, Brandt CA, So-Armah KA, Vasan RS, Oursler KA, Gottdiener J, et al. 2017. Association between HIV infection and the risk of heart failure with reduced ejection fraction and preserved ejection fraction in the antiretroviral therapy era: results from the veterans aging cohort study. JAMA Cardiol 2:536–546.
14.
Tseng ZH, Moffatt E, Kim A, Vittinghoff E, Ursell P, Connolly A, Olgin JE, Wong JK, Hsue PY. 2021. Sudden cardiac death and myocardial fibrosis, determined by autopsy, in persons with HIV. N Engl J Med 384:2306–2316.
15.
Barnett CF, Hsue PY. 2013. Human immunodeficiency virus-associated pulmonary arterial hypertension. Clin Chest Med 34:283–292.
16.
Sitbon O, Lascoux-Combe C, Delfraissy J-F, Yeni PG, Raffi F, De Zuttere D, Gressin V, Clerson P, Sereni D, Simonneau G. 2008. Prevalence of HIV-related pulmonary arterial hypertension in the current antiretroviral therapy era. Am J Respir Crit Care Med 177:108–113.
17.
Feinstein MJ, Bahiru E, Achenbach C, Longenecker CT, Hsue P, So-Armah K, Freiberg MS, Lloyd-Jones DM. 2016. Patterns of cardiovascular mortality for HIV-infected adults in the United States: 1999 to 2013. Am J Cardiol 117:214–220.
18.
Naif HM. 2013. Pathogenesis of HIV infection. Infect Dis Rep 5:e6.
19.
Chun T-W, Fauci AS. 2012. HIV reservoirs: pathogenesis and obstacles to viral eradication and cure. AIDS 26:1261–1268.
20.
Moir S, Chun TW, Fauci AS. 2011. Pathogenic mechanisms of HIV disease. Annu Rev Pathol 6:223–248.
21.
El-Atrouni W, Berbari E, Temesgen Z. 2006. HIV-associated opportunistic infections. Bacterial infections. J Med Liban 54:80–83.
22.
Sabin CA, Lundgren JD. 2013. The natural history of HIV infection. Curr Opin HIV AIDS 8:311–317.
23.
Gaardbo JC, Hartling HJ, Gerstoft J, Nielsen SD. 2012. Thirty years with HIV infection-nonprogression is still puzzling: lessons to be learned from controllers and long-term nonprogressors. AIDS Res Treat 2012:161584.
24.
Poropatich K, Sullivan DJ. 2011. Human immunodeficiency virus type 1 long-term non-progressors: the viral, genetic and immunological basis for disease non-progression. J Gen Virol 92:247–268.
25.
Grabar S, Selinger-Leneman H, Abgrall S, Pialoux G, Weiss L, Costagliola D. 2009. Prevalence and comparative characteristics of long-term nonprogressors and HIV controller patients in the French hospital database on HIV. AIDS 23:1163–1169.
26.
Rodrigues V, Ruffin N, San-Roman M, Benaroch P. 2017. Myeloid cell interaction with HIV: a complex relationship. Front Immunol 8:1698.
27.
Cunningham AL, Li S, Juarez J, Lynch G, Alali M, Naif H. 2000. The level of HIV infection of macrophages is determined by interaction of viral and host cell genotypes. J Leukoc Biol 68:311–317.
28.
El-Sadr WM, Lundgren JD, Neaton JD, Gordin F, Abrams D, Arduino RC, Babiker A, Burman W, Clumeck N, Cohen CJ, Cohn D, Cooper D, Darbyshire J, Emery S, Fätkenheuer G, Gazzard B, Grund B, Hoy J, Klingman K, Losso M, Markowitz N, Neuhaus J, Phillips A, Rappoport C, Strategies for Management of Antiretroviral Therapy (SMART) Study Group. 2006. CD4+ count-guided interruption of antiretroviral treatment. N Engl J Med 355:2283–2296.
29.
Lundgren JD, Babiker AG, Gordin F, Emery S, Grund B, Sharma S, Avihingsanon A, Cooper DA, Fätkenheuer G, Llibre JM, Molina J-M, Munderi P, Schechter M, Wood R, Klingman KL, Collins S, Lane HC, Phillips AN, Neaton JD, INSIGHT START Study Group. 2015. Initiation of antiretroviral therapy in early asymptomatic HIV infection. N Engl J Med 373:795–807.
30.
Bacon O, Chin J, Cohen SE, Hessol NA, Sachdev D, Coffey S, Scheer S, Buchbinder S, Havlir DV, Hsu L. 2021. Decreased time from human immunodeficiency virus diagnosis to care, antiretroviral therapy initiation, and virologic suppression during the citywide RAPID initiative in San Francisco. Clin Infect Dis 73:e122–e128.
31.
Bai R, Du J, Lv S, Hua W, Dai L, Wu H. 2022. Benefits and risks of rapid initiation of antiretroviral therapy: a systematic review and meta-analysis. Front Pharmacol 13:898449.
32.
Dah TTE, Yaya I, Mensah E, Coulibaly A, Kouamé J-B, Traoré I, Mora M, Palvadeau P, Anoma C, Keita BD, Spire B, Laurent C, CohMSM Study Group. 2021. Rapid antiretroviral therapy initiation and its effect on treatment response in MSM in West Africa. AIDS 35:2201–2210.
33.
Lebelonyane R, Bachanas P, Block L, Ussery F, Abrams W, Roland M, Theu J, Kapanda M, Matambo S, Lockman S, Gaolathe T, Makhema J, Moore J, Jarvis JN. 2020. Rapid antiretroviral therapy initiation in the Botswana combination prevention project: a quasi-experimental before and after study. Lancet HIV 7:e545–e553.
34.
Michienzi SM, Barrios M, Badowski ME. 2021. Evidence regarding rapid initiation of antiretroviral therapy in patients living with HIV. Curr Infect Dis Rep 23:7.
35.
Gandhi RT, Bedimo R, Hoy JF, Landovitz RJ, Smith DM, Eaton EF, Lehmann C, Springer SA, Sax PE, Thompson MA, Benson CA, Buchbinder SP, Del Rio C, Eron JJ, Günthard HF, Molina J-M, Jacobsen DM, Saag MS. 2023. Antiretroviral drugs for treatment and prevention of HIV infection in adults: 2022 recommendations of the international antiviral society-USA panel. JAMA 329:63–84.
36.
Wandeler G, Johnson LF, Egger M. 2016. Trends in life expectancy of HIV-positive adults on antiretroviral therapy across the globe: comparisons with general population. Curr Opin HIV AIDS 11:492–500.
37.
Simmons RD, Ciancio BC, Kall MM, Rice BD, Delpech VC. 2013. Ten-year mortality trends among persons diagnosed with HIV infection in England and Wales in the era of antiretroviral therapy: AIDS remains a silent killer. HIV Med 14:596–604.
38.
Croxford S, Kitching A, Desai S, Kall M, Edelstein M, Skingsley A, Burns F, Copas A, Brown AE, Sullivan AK, Delpech V. 2017. Mortality and causes of death in people diagnosed with HIV in the era of highly active antiretroviral therapy compared with the general population: an analysis of a national observational cohort. Lancet Public Health 2:e35–e46.
39.
Fontela C, Aguinaga A, Moreno-Iribas C, Repáraz J, Rivero M, Gracia M, Floristán Y, Fresán U, Miguel RS, Ezpeleta C, Castilla J. 2020. Trends and causes of mortality in a population-based cohort of HIV-infected adults in Spain: comparison with the general population. Sci Rep 10:8922.
40.
Lv T, Cao W, Li T. 2021. HIV-related immune activation and inflammation: current understanding and strategies. J Immunol Res 2021:7316456.
41.
Moreno-Fernandez ME, Zapata W, Blackard JT, Franchini G, Chougnet CA. 2009. Human regulatory T cells are targets for human immunodeficiency virus (HIV) infection, and their susceptibility differs depending on the HIV type 1 strain. J Virol 83:12925–12933.
42.
Margolick JB, Muñoz A, Donnenberg AD, Park LP, Galai N, Giorgi JV, O’Gorman MR, Ferbas J. 1995. Failure of T-cell homeostasis preceding AIDS in HIV-1 infection. The multicenter AIDS cohort study. Nat Med 1:674–680.
43.
Deeks SG, Kitchen CMR, Liu L, Guo H, Gascon R, Narváez AB, Hunt P, Martin JN, Kahn JO, Levy J, McGrath MS, Hecht FM. 2004. Immune activation set point during early HIV infection predicts subsequent CD4+ T-cell changes independent of viral load. Blood 104:942–947.
44.
Wada NI, Jacobson LP, Margolick JB, Breen EC, Macatangay B, Penugonda S, Martínez-Maza O, Bream JH. 2015. The effect of HAART-induced HIV suppression on circulating markers of inflammation and immune activation. AIDS 29:463–471.
45.
Caby F, Guihot A, Lambert-Niclot S, Guiguet M, Boutolleau D, Agher R, Valantin MA, Tubiana R, Calvez V, Marcelin AG, Carcelain G, Autran B, Costagliola D, Katlama C. 2016. Determinants of a low CD4/CD8 ratio in HIV-1-infected individuals despite long-term viral suppression. Clin Infect Dis 62:1297–1303.
46.
Howard RR, Fasano CS, Frey L, Miller CH. 1996. Reference intervals of CD3, CD4, CD8, CD4/CD8, and absolute CD4 values in Asian and non-Asian populations. Cytometry 26:231–232.
47.
Amadori A, Zamarchi R, De Silvestro G, Forza G, Cavatton G, Danieli GA, Clementi M, Chieco-Bianchi L. 1995. Genetic control of the CD4/CD8 T-cell ratio in humans. Nat Med 1:1279–1283.
48.
Zheng L, Taiwo B, Gandhi RT, Hunt PW, Collier AC, Flexner C, Bosch RJ. 2014. Factors associated with CD8+ T-cell activation in HIV-1-infected patients on long-term antiretroviral therapy. J Acquir Immune Defic Syndr 67:153–160.
49.
Sainz T, Serrano-Villar S, Díaz L, González Tomé MI, Gurbindo MD, de José MI, Mellado MJ, Ramos JT, Zamora J, Moreno S, Muñoz-Fernández MA. 2013. The CD4/CD8 ratio as a marker T-cell activation, senescence and activation/exhaustion in treated HIV-infected children and young adults. AIDS 27:1513–1516.
50.
Ron R, Moreno E, Martinez-Sanz J, Branas F, Sainz T, Moreno S, Serrano-Villar S. 2023. CD4/CD8 ratio during HIV treatment: time for routine monitoring? Clin Infect Dis.
51.
Serrano-Villar S, Sainz T, Lee SA, Hunt PW, Sinclair E, Shacklett BL, Ferre AL, Hayes TL, Somsouk M, Hsue PY, Van Natta ML, Meinert CL, Lederman MM, Hatano H, Jain V, Huang Y, Hecht FM, Martin JN, McCune JM, Moreno S, Deeks SG. 2014. HIV-infected individuals with low CD4/CD8 ratio despite effective antiretroviral therapy exhibit altered T cell subsets, heightened CD8+ T cell activation, and increased risk of non-AIDS morbidity and mortality. PLoS Pathog 10:e1004078.
52.
Margolick JB, Gange SJ, Detels R, O’Gorman MRG, Rinaldo CR, Lai S. 2006. Impact of inversion of the CD4/CD8 ratio on the natural history of HIV-1 infection. J Acquir Immune Defic Syndr 42:620–626.
53.
Serrano-Villar S, Moreno S, Fuentes-Ferrer M, Sánchez-Marcos C, Avila M, Sainz T, de Villar NGP, Fernández-Cruz A, Estrada V. 2014. The CD4:CD8 ratio is associated with markers of age-associated disease in virally suppressed HIV-infected patients with immunological recovery. HIV Med 15:40–49.
54.
Novelli S, Lécuroux C, Goujard C, Reynes J, Villemant A, Blum L, Essat A, Avettand-Fenoël V, Launay O, Molina J-M, Bourgeois C, Meyer L. 2020. Persistence of monocyte activation under treatment in people followed since acute HIV-1 infection relative to participants at high or low risk of HIV infection. EBioMedicine 62:103129.
55.
Liang H, Xie Z, Shen T. 2017. Monocyte activation and cardiovascular disease in HIV infection. Cell Mol Immunol 14:960–962.
56.
Margolick JB, Bream JH, Nilles TL, Li H, Langan SJ, Deng S, Wang R, Wada N, Leng SX. 2018. Markers of inflammation, and frailty in HIV-uninfected and HIV-infected men in the multicenter AIDS cohort study. J Infect Dis 218:249–258.
57.
Dinh DM, Volpe GE, Duffalo C, Bhalchandra S, Tai AK, Kane AV, Wanke CA, Ward HD. 2015. Intestinal microbiota, microbial translocation, and systemic inflammation in chronic HIV infection. J Infect Dis 211:19–27.
58.
Nowak P, Troseid M, Avershina E, Barqasho B, Neogi U, Holm K, Hov JR, Noyan K, Vesterbacka J, Svärd J, Rudi K, Sönnerborg A. 2015. Gut microbiota diversity predicts immune status in HIV-1 infection. AIDS 29:2409–2418.
59.
Zevin AS, McKinnon L, Burgener A, Klatt NR. 2016. Microbial translocation and microbiome dysbiosis in HIV-associated immune activation. Curr Opin HIV AIDS 11:182–190.
60.
Grund B, Baker JV, Deeks SG, Wolfson J, Wentworth D, Cozzi-Lepri A, Cohen CJ, Phillips A, Lundgren JD, Neaton JD, Group I. 2016. Relevance of Interleukin-6 and D-Dimer for serious non-AIDS morbidity and death among HIV-positive adults on suppressive antiretroviral therapy. PLoS One 11:e0155100.
61.
Duprez DA, Neuhaus J, Kuller LH, Tracy R, Belloso W, De Wit S, Drummond F, Lane HC, Ledergerber B, Lundgren J, Nixon D, Paton NI, Prineas RJ, Neaton JD, INSIGHT SMART Study Group. 2012. Inflammation, coagulation and cardiovascular disease in HIV-infected individuals. PLoS One 7:e44454.
62.
Neuhaus J, Jacobs DR, Baker JV, Calmy A, Duprez D, La Rosa A, Kuller LH, Pett SL, Ristola M, Ross MJ, Shlipak MG, Tracy R, Neaton JD. 2010. Markers of inflammation, coagulation, and renal function are elevated in adults with HIV infection. J Infect Dis 201:1788–1795.
63.
Marchetti G, Cozzi-Lepri A, Merlini E, Bellistrì GM, Castagna A, Galli M, Verucchi G, Antinori A, Costantini A, Giacometti A, di Caro A, D’arminio Monforte A, ICONA Foundation Study Group. 2011. Microbial translocation predicts disease progression of HIV-infected antiretroviral-naive patients with high CD4+ cell count. AIDS 25:1385–1394.
64.
Méndez-Lagares G, Romero-Sánchez MC, Ruiz-Mateos E, Genebat M, Ferrando-Martínez S, Muñoz-Fernández MÁ, Pacheco YM, Leal M. 2013. Long-term suppressive combined antiretroviral treatment does not normalize the serum level of soluble CD14. J Infect Dis 207:1221–1225.
65.
Ross R. 1999. Atherosclerosis--an inflammatory disease. N Engl J Med 340:115–126.
66.
Ridker PM, Rifai N, Stampfer MJ, Hennekens CH. 2000. Plasma concentration of interleukin-6 and the risk of future myocardial infarction among apparently healthy men. Circulation 101:1767–1772.
67.
Pradhan AD, Aday AW, Rose LM, Ridker PM. 2018. Residual inflammatory risk on treatment with PCSK9 inhibition and statin therapy. Circulation 138:141–149.
68.
Lu Y, Zhou S, Dreyer RP, Spatz ES, Geda M, Lorenze NP, D’Onofrio G, Lichtman JH, Spertus JA, Ridker PM, Krumholz HM. 2017. Sex differences in inflammatory markers and health status among young adults with acute myocardial infarction: results from the VIRGO. Circ Cardiovasc Qual Outcomes 10.
69.
Bermudez EA, Rifai N, Buring J, Manson JE, Ridker PM. 2002. Interrelationships among circulating Interleukin-6, C-reactive protein, and traditional cardiovascular risk factors in women. Arterioscler Thromb Vasc Biol 22:1668–1673.
70.
Virmani R, Burke AP, Farb A, Kolodgie FD. 2006. Pathology of the vulnerable plaque. J Am Coll Cardiol 47:C13–C18.
71.
Herder C, de las Heras Gala T, Carstensen-Kirberg M, Huth C, Zierer A, Wahl S, Sudduth-Klinger J, Kuulasmaa K, Peretz D, Ligthart S, Bongaerts BWC, Dehghan A, Ikram MA, Jula A, Kee F, Pietilä A, Saarela O, Zeller T, Blankenberg S, Meisinger C, Peters A, Roden M, Salomaa V, Koenig W, Thorand B. 2017. Circulating levels of interleukin 1-receptor antagonist and risk of cardiovascular disease. ATVB 37:1222–1227.
72.
Schnabel RB, Yin X, Larson MG, Yamamoto JF, Fontes JD, Kathiresan S, Rong J, Levy D, Keaney JF, Wang TJ, Murabito JM, Vasan RS, Benjamin EJ. 2013. Multiple inflammatory biomarkers in relation to cardiovascular events and mortality in the community. Arterioscler Thromb Vasc Biol 33:1728–1733.
73.
Welsh C, Welsh P, Mark PB, Celis-Morales CA, Lewsey J, Gray SR, Lyall DM, Iliodromiti S, Gill JMR, Pell J, Jhund PS, Sattar N. 2018. Association of total and differential leukocyte counts with cardiovascular disease and mortality in the UK biobank. Arterioscler Thromb Vasc Biol 38:1415–1423.
74.
Ridker PM. 2017. How common is residual inflammatory risk. Circ Res 120:617–619.
75.
Libby P. 2012. Inflammation in atherosclerosis. Arterioscler Thromb Vasc Biol 32:2045–2051.
76.
Libby P. 2021. The changing landscape of atherosclerosis. Nature 592:524–533.
77.
Matsuura Y, Kanter JE, Bornfeldt KE. 2019. Highlighting residual atherosclerotic cardiovascular disease risk. Arterioscler Thromb Vasc Biol 39:e1–e9.
78.
Stark K, Massberg S. 2021. Interplay between inflammation and thrombosis in cardiovascular pathology. Nat Rev Cardiol 18:666–682.
79.
Vieira-de-Abreu A, Campbell RA, Weyrich AS, Zimmerman GA. 2012. Platelets: versatile effector cells in hemostasis, inflammation, and the immune continuum. Semin Immunopathol 34:5–30.
80.
Semple JW, Italiano JE, Freedman J. 2011. Platelets and the immune continuum. Nat Rev Immunol 11:264–274.
81.
Hottz ED, Lopes JF, Freitas C, Valls-de-Souza R, Oliveira MF, Bozza MT, Da Poian AT, Weyrich AS, Zimmerman GA, Bozza FA, Bozza PT. 2013. Platelets mediate increased endothelium permeability in dengue through NLRP3-inflammasome activation. Blood 122:3405–3414.
82.
Engelmann B, Massberg S. 2013. Thrombosis as an intravascular effector of innate immunity. Nat Rev Immunol 13:34–45.
83.
Shah SJ, Kitzman DW, Borlaug BA, van Heerebeek L, Zile MR, Kass DA, Paulus WJ. 2016. Phenotype-specific treatment of heart failure with preserved ejection fraction: a multiorgan roadmap. Circulation 134:73–90.
84.
Dick SA, Epelman S. 2016. Chronic heart failure and inflammation: what do we really know? Circ Res 119:159–176.
85.
Levine B, Kalman J, Mayer L, Fillit HM, Packer M. 1990. Elevated circulating levels of tumor necrosis factor in severe chronic heart failure. N Engl J Med 323:236–241.
86.
Givertz MM, Colucci WS. 1998. New targets for heart-failure therapy: endothelin, inflammatory cytokines, and oxidative stress. Lancet 352:SI34–8.
87.
Vasan RS, Sullivan LM, Roubenoff R, Dinarello CA, Harris T, Benjamin EJ, Sawyer DB, Levy D, Wilson PWF, D’Agostino RB, Framingham Heart Study. 2003. Inflammatory markers and risk of heart failure in elderly subjects without prior myocardial infarction: the Framingham heart study. Circulation 107:1486–1491.
88.
Albert CM, Ma J, Rifai N, Stampfer MJ, Ridker PM. 2002. Prospective study of C-reactive protein, homocysteine, and plasma lipid levels as predictors of sudden cardiac death. Circulation 105:2595–2599.
89.
Hussein AA, Gottdiener JS, Bartz TM, Sotoodehnia N, DeFilippi C, See V, Deo R, Siscovick D, Stein PK, Lloyd-Jones D. 2013. Inflammation and sudden cardiac death in a community-based population of older adults: the cardiovascular health study. Heart Rhythm 10:1425–1432.
90.
Palasubramaniam J, Wang X, Peter K. 2019. Myocardial infarction—from atherosclerosis to thrombosis. Arterioscler Thromb Vasc Biol 39:e176–e185.
91.
Lu H, Daugherty A. 2015. Atherosclerosis. Arterioscler Thromb Vasc Biol 35:485–491.
92.
Moroni F, Ammirati E, Norata GD, Magnoni M, Camici PG. 2019. The role of monocytes and macrophages in human atherosclerosis, plaque neoangiogenesis, and atherothrombosis. Mediators Inflamm 2019:7434376.
93.
Bonetti PO, Lerman LO, Lerman A. 2003. Endothelial dysfunction. ATVB 23:168–175.
94.
Otsuka F, Yasuda S, Noguchi T, Ishibashi-Ueda H. 2016. Pathology of coronary atherosclerosis and thrombosis. Cardiovasc Diagn Ther 6:396–408.
95.
Davies MJ, Thomas A. 1984. Thrombosis and acute coronary-artery lesions in sudden cardiac ischemic death. N Engl J Med 310:1137–1140.
96.
Kearns A, Gordon J, Burdo TH, Qin X. 2017. HIV-1-associated atherosclerosis: unraveling the missing link. J Am Coll Cardiol 69:3084–3098.
97.
Libby P, Ridker PM, Hansson GK. 2011. Progress and challenges in translating the biology of atherosclerosis. Nature 473:317–325.
98.
Shrestha S, Irvin MR, Grunfeld C, Arnett DK. 2014. HIV, inflammation, and calcium in atherosclerosis. Arterioscler Thromb Vasc Biol 34:244–250.
99.
Mujawar Z, Rose H, Morrow MP, Pushkarsky T, Dubrovsky L, Mukhamedova N, Fu Y, Dart A, Orenstein JM, Bobryshev YV, Bukrinsky M, Sviridov D. 2006. Human immunodeficiency virus impairs reverse cholesterol transport from macrophages. PLoS Biol 4:e365.
100.
Funderburg NT, Mehta NN. 2016. Lipid abnormalities and inflammation in HIV inflection. Curr HIV/AIDS Rep 13:218–225.
101.
Baker J, Ayenew W, Quick H, Hullsiek KH, Tracy R, Henry K, Duprez D, Neaton JD. 2010. High-density lipoprotein particles and markers of inflammation and thrombotic activity in patients with untreated HIV infection. J Infect Dis 201:285–292.
102.
Kelesidis T, Oda MN, Borja MS, Yee Y, Ng KF, Huynh D, Elashoff D, Currier JS. 2017. Predictors of impaired HDL function in HIV-1 infected compared to uninfected individuals. J Acquir Immune Defic Syndr 75:354–363.
103.
Navab M, Reddy ST, Van Lenten BJ, Fogelman AM. 2011. HDL and cardiovascular disease: atherogenic and atheroprotective mechanisms. Nat Rev Cardiol 8:222–232.
104.
El-Sadr WM, Mullin CM, Carr A, Gibert C, Rappoport C, Visnegarwala F, Grunfeld C, Raghavan SS. 2005. Effects of HIV disease on lipid, glucose and insulin levels: results from a large antiretroviral-naive cohort. HIV Med 6:114–121.
105.
Feinstein MJ, Achenbach CJ, Stone NJ, Lloyd-Jones DM. 2015. A systematic review of the usefulness of statin therapy in HIV-infected patients. Am J Cardiol 115:1760–1766.
106.
Waters DD, Hsue PY. 2019. Lipid abnormalities in persons living with HIV infection. Can J Cardiol 35:249–259.
107.
Mallon PWG, Brunet L, Fusco JS, Prajapati G, Beyer A, Fusco GP, Wohlfeiler MB. 2022. Lipid changes after switch from TDF to TAF in the OPERA cohort: LDL cholesterol and triglycerides. Open Forum Infect Dis 9:ofab621.
108.
Taramasso L, Tatarelli P, Ricci E, Madeddu G, Menzaghi B, Squillace N, De Socio GV, Martinelli C, Gulminetti R, Maggi P, Orofino G, Vichi F, Di Biagio A, Bonfanti P, CISAI Study Group. 2018. Improvement of lipid profile after switching from efavirenz or ritonavir-boosted protease inhibitors to rilpivirine or once-daily integrase inhibitors: results from a large observational cohort study (SCOLTA). BMC Infect Dis 18:357.
109.
Koethe JR, Lagathu C, Lake JE, Domingo P, Calmy A, Falutz J, Brown TT, Capeau J. 2020. HIV and antiretroviral therapy-related fat alterations. Nat Rev Dis Primers 6:48.
110.
Guaraldi G, Stentarelli C, Zona S, Santoro A, Beghetto B, Carli F, Orlando G, Franceschetto A, Casolo A, Mussini C. 2014. The natural history of HIV-associated lipodystrophy in the changing scenario of HIV infection. HIV Med 15:587–594.
111.
Schechter ME, Andrade BB, He T, Richter GH, Tosh KW, Policicchio BB, Singh A, Raehtz KD, Sheikh V, Ma D, Brocca-Cofano E, Apetrei C, Tracy R, Ribeiro RM, Sher A, Francischetti IMB, Pandrea I, Sereti I. 2017. Inflammatory monocytes expressing tissue factor drive SIV and HIV coagulopathy. Sci Transl Med 9:eaam5441.
112.
Funderburg NT, Mayne E, Sieg SF, Asaad R, Jiang W, Kalinowska M, Luciano AA, Stevens W, Rodriguez B, Brenchley JM, Douek DC, Lederman MM. 2010. Increased tissue factor expression on circulating monocytes in chronic HIV infection: relationship to in vivo coagulation and immune activation. Blood 115:161–167.
113.
Kuller LH, Tracy R, Belloso W, De Wit S, Drummond F, Lane HC, Ledergerber B, Lundgren J, Neuhaus J, Nixon D, Paton NI, Neaton JD, INSIGHT SMART Study Group. 2008. Inflammatory and coagulation biomarkers and mortality in patients with HIV infection. PLoS Med 5:e203.
114.
Perkins MV, Joseph SB, Dittmer DP, Mackman N. 2023. Cardiovascular disease and thrombosis in HIV infection. Arterioscler Thromb Vasc Biol 43:175–191.
115.
Badru O, Oduola T, Abdulrazaq A, Peter C. 2022. Prevalence and predictive factors of hypertension among people living with HIV in Kebbi state. J Assoc Nurses AIDS Care 33:e6–e14.
116.
Rajagopaul A, Naidoo M. 2021. Prevalence of diabetes mellitus and hypertension amongst the HIV-positive population at a district hospital in eThekwini, South Africa. Afr J Prim Health Care Fam Med 13:e1–e6.
117.
Gonah L, Moodley I, Hlongwana K. 2020. Prevalence, healthcare costs and management of non-communicable diseases in people living with human immunodeficiency virus: a scoping review. Afr J Prim Health Care Fam Med 12:e1–e8.
118.
Musekwa R, Hamooya BM, Koethe JR, Nzala S, Masenga SK. 2021. Prevalence and correlates of hypertension in HIV-positive adults from the Livingstone central hospital, Zambia. Pan Afr Med J 39:237.
119.
Manavalan P, Madut DB, Hertz JT, Thielman NM, Okeke NL, Mmbaga BT, Watt MH. 2020. Hypertension burden and challenges across the hypertension treatment cascade among adults enrolled in HIV care in northern Tanzania. J Clin Hypertens (Greenwich) 22:1518–1522.
120.
Bigna JJ, Ndoadoumgue AL, Nansseu JR, Tochie JN, Nyaga UF, Nkeck JR, Foka AJ, Kaze AD, Noubiap JJ. 2020. Global burden of hypertension among people living with HIV in the era of increased life expectancy: a systematic review and meta-analysis. J Hypertens 38:1659–1668.
121.
Harimenshi D, Niyongabo T, Preux P-M, Aboyans V, Desormais I. 2022. Hypertension and associated factors in HIV-infected patients receiving antiretroviral treatment in Burundi: a cross-sectional study. Sci Rep 12:20509.
122.
Xu Y, Chen X, Wang K. 2017. Global prevalence of hypertension among people living with HIV: a systematic review and meta-analysis. J Am Soc Hypertens 11:530–540.
123.
Fahme SA, Bloomfield GS, Peck R. 2018. Hypertension in HIV-infected adults. Hypertension 72:44–55.
124.
Crane HM, Grunfeld C, Harrington RD, Kitahata MM. 2009. Lipoatrophy and lipohypertrophy are independently associated with hypertension. HIV Med 10:496–503.
125.
Srinivasa S, Fitch KV, Wong K, Torriani M, Mayhew C, Stanley T, Lo J, Adler GK, Grinspoon SK. 2015. RAAS activation is associated with visceral adiposity and insulin resistance among HIV-infected patients. J Clin Endocrinol Metab 100:2873–2882.
126.
Brown TT, Cole SR, Li X, Kingsley LA, Palella FJ, Riddler SA, Visscher BR, Margolick JB, Dobs AS. 2005. Antiretroviral therapy and the prevalence and incidence of diabetes mellitus in the multicenter AIDS cohort study. Arch Intern Med 165:1179–1184.
127.
Birabaharan M, Strunk A, Kaelber DC, Smith DM, Martin TCS. 2022. Sex differences in type 2 diabetes mellitus prevalence among persons with HIV. AIDS 36:383–389.
128.
Worm SW, De Wit S, Weber R, Sabin CA, Reiss P, El-Sadr W, Monforte AD, Kirk O, Fontas E, Dabis F, Law MG, Lundgren JD, Friis-Møller N. 2009. Diabetes mellitus, preexisting coronary heart disease, and the risk of subsequent coronary heart disease events in patients infected with human immunodeficiency virus. Circulation 119:805–811.
129.
Sarkar S, Brown TT. 2021. Diabetes in people with HIV. Curr Diab Rep 21:13.
130.
Buchacz K, Baker RK, Palella FJ, Shaw L, Patel P, Lichtenstein KA, Chmiel JS, Vellozzi C, Debes R, Henry K, Overton ET, Bush TJ, Tedaldi E, Carpenter C, Mayer KH, Brooks JT, HIV Outpatient Study Investigators. 2013. Disparities in prevalence of key chronic diseases by gender and race/ethnicity among antiretroviral-treated HIV-infected adults in the US. Antivir Ther 18:65–75.
131.
Warriner AH, Burkholder GA, Overton ET. 2014. HIV-related metabolic comorbidities in the current ART era. Infect Dis Clin North Am 28:457–476.
132.
Frazier EL, Sutton MY, Brooks JT, Shouse RL, Weiser J. 2018. Trends in cigarette smoking among adults with HIV compared with the general adult population, United States - 2009–2014. Prev Med 111:231–234.
133.
Rasmussen LD, Helleberg M, May MT, Afzal S, Kronborg G, Larsen CS, Pedersen C, Gerstoft J, Nordestgaard BG, Obel N. 2015. Myocardial infarction among Danish HIV-infected individuals: population-attributable fractions associated with smoking. Clin Infect Dis 60:1415–1423.
134.
Kelly SG, Plankey M, Post WS, Li X, Stall R, Jacobson LP, Witt MD, Kingsley L, Cox C, Budoff M, Palella FJ. 2016. Associations between tobacco alcohol, and drug use with coronary artery plaque among HIV-infected and uninfected men in the multicenter AIDS cohort study. PLoS One 11:e0147822.
135.
Freiberg MS, McGinnis KA, Kraemer K, Samet JH, Conigliaro J, Curtis Ellison R, Bryant K, Kuller LH, Justice AC, VACS Project Team. 2010. The association between alcohol consumption and prevalent cardiovascular diseases among HIV-infected and HIV-uninfected men. J Acquir Immune Defic Syndr 53:247–253.
136.
Miller PE, Haberlen SA, Metkus T, Rezaeian P, Palella F, Kingsley LA, Witt MD, George RT, Jacobson LP, Brown TT, Budoff M, Post WS. 2015. HIV and coronary arterial remodeling from the multicenter AIDS cohort study (MACS). Atherosclerosis 241:716–722.
137.
Zanni MV, Abbara S, Lo J, Wai B, Hark D, Marmarelis E, Grinspoon SK. 2013. Increased coronary atherosclerotic plaque vulnerability by coronary computed tomography angiography in HIV-infected men. AIDS 27:1263–1272.
138.
Post WS, Budoff M, Kingsley L, Palella FJ, Witt MD, Li X, George RT, Brown TT, Jacobson LP. 2014. Associations between HIV infection and subclinical coronary atherosclerosis. Ann Intern Med 160:458–467.
139.
Bahrami H, Budoff M, Haberlen SA, Rezaeian P, Ketlogetswe K, Tracy R, Palella F, Witt MD, McConnell MV, Kingsley L, Post WS. 2016. Inflammatory markers associated with subclinical coronary artery disease: the multicenter AIDS cohort study. J Am Heart Assoc 5:e003371.
140.
Tawakol A, Lo J, Zanni MV, Marmarelis E, Ihenachor EJ, MacNabb M, Wai B, Hoffmann U, Abbara S, Grinspoon S. 2014. Increased arterial inflammation relates to high-risk coronary plaque morphology in HIV-infected patients. J Acquir Immune Defic Syndr 66:164–171.
141.
Hanna DB, Lin J, Post WS, Hodis HN, Xue X, Anastos K, Cohen MH, Gange SJ, Haberlen SA, Heath SL, Lazar JM, Liu C, Mack WJ, Ofotokun I, Palella FJ, Tien PC, Witt MD, Landay AL, Kingsley LA, Tracy RP, Kaplan RC. 2017. Association of macrophage inflammation biomarkers with progression of subclinical carotid artery atherosclerosis in HIV-infected women and men. J Infect Dis 215:1352–1361.
142.
Werede AT, Terry JG, Nair S, Temu TM, Shepherd BE, Bailin SS, Mashayekhi M, Gabriel CL, Lima M, Woodward BO, Hannah L, Mallal SA, Beckman JA, Li JZ, Fajnzylber J, Harrison DG, Carr JJ, Koethe JR, Wanjalla CN. 2022. Mean coronary cross-sectional area as a measure of arterial remodeling using noncontrast CT imaging in persons with HIV. J Am Heart Assoc 11:e025768.
143.
Kolossváry M, deFilippi C, Lu MT, Zanni MV, Fulda ES, Foldyna B, Ribaudo H, Mayrhofer T, Collier AC, Bloomfield GS, Fichtenbaum C, Overton ET, Aberg JA, Currier J, Fitch KV, Douglas PS, Grinspoon SK. 2022. Proteomic signature of subclinical coronary artery disease in people with HIV: analysis of the REPRIEVE mechanistic substudy. J Infect Dis 226:1809–1822.
144.
Lawal IO, Ankrah AO, Popoola GO, Lengana T, Sathekge MM. 2019. Arterial inflammation in young patients with human immunodeficiency virus infection: a cross-sectional study using F-18 FDG PET/CT. J Nucl Cardiol 26:1258–1265.
145.
Knudsen A, Hag AMF, Loft A, von Benzon E, Keller SH, Møller HJ, Lebech A-M, Ripa RS, Kjær A. 2015. HIV infection and arterial inflammation assessed by (18)F-fluorodeoxyglucose (FDG) positron emission tomography (PET): a prospective cross-sectional study. J Nucl Cardiol 22:372–380.
146.
Janjua SA, Staziaki PV, Szilveszter B, Takx RAP, Mayrhofer T, Hennessy O, Emami HA, Park J, Ivanov A, Hallett TR, Lu MT, Romero JM, Grinspoon SK, Hoffmann U, Zanni MV, Neilan TG. 2017. Presence, characteristics, and prognostic associations of carotid plaque among people living with HIV. Circ Cardiovasc Imaging 10:e005777.
147.
Hanna DB, Guo M, Bůžková P, Miller TL, Post WS, Stein JH, Currier JS, Kronmal RA, Freiberg MS, Bennett SN, Shikuma CM, Anastos K, Li Y, Tracy RP, Hodis HN, Delaney JA, Kaplan RC. 2016. HIV infection and carotid artery intima-media thickness: pooled analyses across 5 cohorts of the NHLBI HIV-CVD collaborative. Clin Infect Dis 63:249–256.
148.
Hsue PY, Ordovas K, Lee T, Reddy G, Gotway M, Schnell A, Ho JE, Selby V, Madden E, Martin JN, Deeks SG, Ganz P, Waters DD. 2012. Carotid intima-media thickness among human immunodeficiency virus-infected patients without coronary calcium. Am J Cardiol 109:742–747.
149.
Hudson JA, Majonga ED, Ferrand RA, Perel P, Alam SR, Shah ASV. 2022. Association of HIV infection with cardiovascular pathology based on advanced cardiovascular imaging: a systematic review. JAMA 328:951.
150.
Feinstein MJ. 2022. HIV, subclinical cardiovascular disease, and clinical progression: Insights from immunologic heterogeneity. JAMA 328:931–932.
151.
Freiberg MS, Chang C-CH, Kuller LH, Skanderson M, Lowy E, Kraemer KL, Butt AA, Bidwell Goetz M, Leaf D, Oursler KA, et al. 2013. HIV infection and the risk of acute myocardial infarction. JAMA Intern Med 173:614–622.
152.
Drozd DR, Kitahata MM, Althoff KN, Zhang J, Gange SJ, Napravnik S, Burkholder GA, Mathews WC, Silverberg MJ, Sterling TR, et al. 2017. Increased risk of myocardial infarction in HIV-infected individuals in North America compared with the general population. J Acquir Immune Defic Syndr 75:568–576.
153.
Lichtenstein KA, Armon C, Buchacz K, Chmiel JS, Buckner K, Tedaldi EM, Wood K, Holmberg SD, Brooks JT, Investigators H. 2010. Low CD4+ T cell count is a risk factor for cardiovascular disease events in the HIV outpatient study. Clin Infect Dis 51:435–447.
154.
Silverberg MJ, Leyden WA, Xu L, Horberg MA, Chao CR, Towner WJ, Hurley LB, Quesenberry CP, Klein DB. 2014. Immunodeficiency and risk of myocardial infarction among HIV-positive individuals with access to care. J Acquir Immune Defic Syndr 65:160–166.
155.
Triant VA, Lee H, Hadigan C, Grinspoon SK. 2007. Increased acute myocardial infarction rates and cardiovascular risk factors among patients with human immunodeficiency virus disease. J Clin Endocrinol Metab 92:2506–2512.
156.
Triant VA, Regan S, Lee H, Sax PE, Meigs JB, Grinspoon SK. 2010. Association of immunologic and virologic factors with myocardial infarction rates in a US healthcare system. J Acquir Immune Defic Syndr 55:615–619.
157.
Lang S, Mary-Krause M, Cotte L, Gilquin J, Partisani M, Simon A, Boccara F, Bingham A, Costagliola D, French Hospital Database on HIV-ANRS CO4. 2010. Increased risk of myocardial infarction in HIV-infected patients in France, relative to the general population. AIDS 24:1228–1230.
158.
Feinstein MJ, Bogorodskaya M, Bloomfield GS, Vedanthan R, Siedner MJ, Kwan GF, Longenecker CT. 2016. Cardiovascular complications of HIV in endemic countries. Curr Cardiol Rep 18:113.
159.
Ho JE, Deeks SG, Hecht FM, Xie Y, Schnell A, Martin JN, Ganz P, Hsue PY. 2010. Initiation of antiretroviral therapy at higher nadir CD4+ T-cell counts is associated with reduced arterial stiffness in HIV-infected individuals. AIDS 24:1897–1905.
160.
Ho JE, Scherzer R, Hecht FM, Maka K, Selby V, Martin JN, Ganz P, Deeks SG, Hsue PY. 2012. The association of CD4+ T-cell counts and cardiovascular risk in treated HIV disease. AIDS 26:1115–1120.
161.
Blum A, Hadas V, Burke M, Yust I, Kessler A. 2005. Viral load of the human immunodeficiency virus could be an independent risk factor for endothelial dysfunction. Clin Cardiol 28:149–153.
162.
Womack JA, Brandt CA, Justice AC. 2014. Women, aging, and HIV: clinical issues and management strategies. J Nurse Pract 10:409–416.
163.
Rasmussen LD, Engsig FN, Christensen H, Gerstoft J, Kronborg G, Pedersen C, Obel N. 2011. Risk of cerebrovascular events in persons with and without HIV: a Danish nationwide population-based cohort study. AIDS 25:1637–1646.
164.
Vinikoor MJ, Napravnik S, Floris-Moore M, Wilson S, Huang DY, Eron JJ. 2013. Incidence and clinical features of cerebrovascular disease among HIV-infected adults in the southeastern United States. AIDS Res Hum Retroviruses 29:1068–1074.
165.
Marcus JL, Leyden WA, Chao CR, Chow FC, Horberg MA, Hurley LB, Klein DB, Quesenberry CP, Towner WJ, Silverberg MJ. 2014. HIV infection and incidence of ischemic stroke. AIDS 28:1911–1919.
166.
Lo J, Abbara S, Shturman L, Soni A, Wei J, Rocha-Filho JA, Nasir K, Grinspoon SK. 2010. Increased prevalence of subclinical coronary atherosclerosis detected by coronary computed tomography angiography in HIV-infected men. AIDS 24:243–253.
167.
Vasilieva E, Gianella S, Freeman ML. 2020. Novel strategies to combat CMV-related cardiovascular disease. Pathog Immun 5:240–274.
168.
Lichtner M, Cicconi P, Vita S, Cozzi-Lepri A, Galli M, Lo Caputo S, Saracino A, De Luca A, Moioli M, Maggiolo F, Marchetti G, Vullo V, d’Arminio Monforte A, ICONA Foundation Study. 2015. Cytomegalovirus coinfection is associated with an increased risk of severe non-AIDS-defining events in a large cohort of HIV-infected patients. J Infect Dis 211:178–186.
169.
Gianella S, Massanella M, Wertheim JO, Smith DM. 2015. The sordid affair between human herpesvirus and HIV. J Infect Dis 212:845–852.
170.
Lloyd-Jones DM, Huffman MD, Karmali KN, Sanghavi DM, Wright JS, Pelser C, Gulati M, Masoudi FA, Goff DC. 2017. Estimating longitudinal risks and benefits from cardiovascular preventive therapies among medicare patients: the million hearts longitudinal ASCVD risk assessment tool: a special report from the American heart association and American college of cardiology. J Am Coll Cardiol 69:1617–1636.
171.
Feinstein MJ, Nance RM, Drozd DR, Ning H, Delaney JA, Heckbert SR, Budoff MJ, Mathews WC, Kitahata MM, Saag MS, Eron JJ, Moore RD, Achenbach CJ, Lloyd-Jones DM, Crane HM. 2017. Assessing and refining myocardial infarction risk estimation among patients with human immunodeficiency virus: a study by the centers for AIDS research network of integrated clinical systems. JAMA Cardiol 2:155–162.
172.
Friis-Møller N, Ryom L, Smith C, Weber R, Reiss P, Dabis F, De Wit S, Monforte AD, Kirk O, Fontas E, Sabin C, Phillips A, Lundgren J, Law M, D:A:D study group. 2016. An updated prediction model of the global risk of cardiovascular disease in HIV-positive persons: the data-collection on adverse effects of anti-HIV drugs (D:A:D) study. Eur J Prev Cardiol 23:214–223.
173.
Thompson-Paul AM, Lichtenstein KA, Armon C, Palella FJ, Skarbinski J, Chmiel JS, Hart R, Wei SC, Loustalot F, Brooks JT, Buchacz K. 2016. Cardiovascular disease risk prediction in the HIV outpatient study. Clin Infect Dis 63:1508–1516.
174.
Triant VA, Perez J, Regan S, Massaro JM, Meigs JB, Grinspoon SK, D’Agostino RB. 2018. Cardiovascular risk prediction functions underestimate risk in HIV infection. Circulation 137:2203–2214.
175.
Achhra AC, Lyass A, Borowsky L, Bogorodskaya M, Plutzky J, Massaro JM, D’Agostino RB, Triant VA. 2021. Assessing cardiovascular risk in people living with HIV: current tools and limitations. Curr HIV/AIDS Rep 18:271–279.
176.
Greenland P, Blaha MJ, Budoff MJ, Erbel R, Watson KE. 2018. Coronary calcium score and cardiovascular risk. J Am Coll Cardiol 72:434–447.
177.
Ambale-Venkatesh B, Yang X, Wu CO, Liu K, Hundley WG, McClelland R, Gomes AS, Folsom AR, Shea S, Guallar E, Bluemke DA, Lima JAC. 2017. Cardiovascular event prediction by machine learning: the multi-ethnic study of atherosclerosis. Circ Res 121:1092–1101.
178.
McClelland RL, Jorgensen NW, Budoff M, Blaha MJ, Post WS, Kronmal RA, Bild DE, Shea S, Liu K, Watson KE, Folsom AR, Khera A, Ayers C, Mahabadi A-A, Lehmann N, Jöckel K-H, Moebus S, Carr JJ, Erbel R, Burke GL. 2015. 10-year coronary heart disease risk prediction using coronary artery calcium and traditional risk factors: derivation in the MESA (multi-ethnic study of atherosclerosis) with validation in the HNR. J Am Coll Cardiol 66:1643–1653.
179.
MESA. MESA 10-year CHD risk with coronary artery calcification. Available from: https://www.mesa-nhlbi.org/MESACHDRisk/MesaRiskScore/RiskScore.aspx. Retrieved 15 Sep 2023.
181.
Boccara F, Kumar PN, Caramelli B, Calmy A, López JAG, Bray S, Cyrille M, Rosenson RS, BEIJERINCK Investigators. 2020. Evolocumab in HIV-infected patients with dyslipidemia: Primary results of the randomized, double-blind BEIJERINCK study. J Am Coll Cardiol 75:2570–2584.
182.
Swanson B, Keithley J, Baum L, Leurgans S, Adeyemi O, Barnes LL, Mata M, Rosdil A. 2018. Effects of fish oil on HIV-related inflammation and markers of immunosenescence: a randomized clinical trial. J Altern Complement Med 24:709–716.
183.
Samineni D, Fichtenbaum CJ. 2010. Fenofibrate in the treatment of dyslipidemia associated with HIV infection. Expert Opin Drug Metab Toxicol 6:995–1004.
184.
Surial B, Chammartin F, Damas J, Calmy A, Haerry D, Stöckle M, Schmid P, Bernasconi E, Fux CA, Tarr PE, Günthard HF, Wandeler G, Rauch A, tSHC S. 2023. Impact of Integrase inhibitors on cardiovascular disease events in people with human immunodeficiency virus starting antiretroviral therapy. Clin Infect Dis 77:729–737.
185.
CD4+ count–guided interruption of antiretroviral treatment. 2006. N Engl J Med 355:2283–2296.
186.
Strategies for Management of Antiretroviral Therapy (SMART) Study Group, Emery S, Neuhaus JA, Phillips AN, Babiker A, Cohen CJ, Gatell JM, Girard P-M, Grund B, Law M, Losso MH, Palfreeman A, Wood R. 2008. Major clinical outcomes in antiretroviral therapy (ART)–naive participants and in those not receiving ART at baseline in the SMART study. J Infect Dis 197:1133–1144.
187.
NIo H. 2023. Daily statin reduces the risk of cardiovascular disease in people living with HIV, large NIH study finds. Available from: https://www.nih.gov/news-events/news-releases/daily-statin-reduces-risk-cardiovascular-disease-people-living-hiv-large-nih-study-finds. Retrieved 11 Apr 2023.
188.
Marcantoni E, Allen N, Cambria MR, Dann R, Cammer M, Lhakhang T, O’Brien MP, Kim B, Worgall T, Heguy A, Tsirigos A, Berger JS. 2018. Platelet transcriptome profiling in HIV and ATP-binding cassette subfamily C member 4 (ABCC4) as a mediator of platelet activity. JACC Basic Transl Sci 3:9–22.
189.
Marcantoni E, Garshick MS, Schwartz T, Ratnapala N, Cambria M, Dann R, O’Brien M, Heguy A, Berger JS. 2022. Antiplatelet effects of clopidogrel vs aspirin in virologically controlled HIV: a randomized controlled trial. JACC Basic Transl Sci 7:1086–1097.
190.
O’Brien MP, Hunt PW, Kitch DW, Klingman K, Stein JH, Funderburg NT, Berger JS, Tebas P, Clagett B, Moisi D, Utay NS, Aweeka F, Aberg JA. 2017. A randomized placebo controlled trial of aspirin effects on immune activation in chronically human immunodeficiency virus-infected adults on virologically suppressive antiretroviral therapy. Open Forum Infect Dis 4:ofw278.
191.
Hsue PY, Li D, Ma Y, Ishai A, Manion M, Nahrendorf M, Ganz P, Ridker PM, Deeks SG, Tawakol A. 2018. IL-1β inhibition reduces atherosclerotic inflammation in HIV infection. J Am Coll Cardiol 72:2809–2811.
192.
Ridker PM, Everett BM, Thuren T, MacFadyen JG, Chang WH, Ballantyne C, Fonseca F, Nicolau J, Koenig W, Anker SD, et al. 2017. Antiinflammatory therapy with canakinumab for atherosclerotic disease. N Engl J Med 377:1119–1131.
193.
Ridker PM, MacFadyen JG, Everett BM, Libby P, Thuren T, Glynn RJ, Ridker PM, MacFadyen JG, Everett BM, Libby P. 2018. Relationship of C-reactive protein reduction to cardiovascular event reduction following treatment with canakinumab: a secondary analysis from the CANTOS randomised controlled trial. Lancet 391:319–328.
194.
Ridker PM, Libby P, MacFadyen JG, Thuren T, Ballantyne C, Fonseca F, Koenig W, Shimokawa H, Everett BM, Glynn RJ. 2018. Modulation of the interleukin-6 signalling pathway and incidence rates of atherosclerotic events and all-cause mortality: analyses from the canakinumab anti-inflammatory thrombosis outcomes study (CANTOS). Eur Heart J 39:3499–3507.
195.
Tardif J-C, Kouz S, Waters DD, Bertrand OF, Diaz R, Maggioni AP, Pinto FJ, Ibrahim R, Gamra H, Kiwan GS, Berry C, López-Sendón J, Ostadal P, Koenig W, Angoulvant D, Grégoire JC, Lavoie M-A, Dubé M-P, Rhainds D, Provencher M, Blondeau L, Orfanos A, L’Allier PL, Guertin M-C, Roubille F. 2019. Efficacy and safety of low-dose colchicine after myocardial infarction. N Engl J Med 381:2497–2505.
196.
Nidorf SM, Fiolet ATL, Mosterd A, Eikelboom JW, Schut A, Opstal TSJ, The SHK, Xu X-F, Ireland MA, Lenderink T, et al. 2020. Colchicine in patients with chronic coronary disease. N Engl J Med 383:1838–1847.
197.
Ridker PM, Everett BM, Pradhan A, MacFadyen JG, Solomon DH, Zaharris E, Mam V, Hasan A, Rosenberg Y, Iturriaga E, et al. 2019. Low-dose methotrexate for the prevention of atherosclerotic events. N Engl J Med 380:752–762.
198.
van Halm VP, Nurmohamed MT, Twisk JWR, Dijkmans BAC, Voskuyl AE. 2006. Disease-modifying antirheumatic drugs are associated with a reduced risk for cardiovascular disease in patients with rheumatoid arthritis: a case control study. Arthritis Res Ther 8:R151.
199.
Prodanovich S, Ma F, Taylor JR, Pezon C, Fasihi T, Kirsner RS. 2005. Methotrexate reduces incidence of vascular diseases in veterans with psoriasis or rheumatoid arthritis. J Am Acad Dermatol 52:262–267.
200.
Solomon DH, Avorn J, Katz JN, Weinblatt ME, Setoguchi S, Levin R, Schneeweiss S. 2006. Immunosuppressive medications and hospitalization for cardiovascular events in patients with rheumatoid arthritis. Arthritis & Rheumatism 54:3790–3798.
201.
Naranjo A, Sokka T, Descalzo MA, Calvo-Alén J, Hørslev-Petersen K, Luukkainen RK, Combe B, Burmester GR, Devlin J, Ferraccioli G, Morelli A, Hoekstra M, Majdan M, Sadkiewicz S, Belmonte M, Holmqvist A-C, Choy E, Tunc R, Dimic A, Bergman M, Toloza S, Pincus T, QUEST-RA Group. 2008. Cardiovascular disease in patients with rheumatoid arthritis: results from the QUEST-RA study. Arthritis Res Ther 10:R30.
202.
Choi HK, Hernán MA, Seeger JD, Robins JM, Wolfe F. 2002. Methotrexate and mortality in patients with rheumatoid arthritis: a prospective study. Lancet 359:1173–1177.
203.
Hsue PY, Ribaudo HJ, Deeks SG, Bell T, Ridker PM, Fichtenbaum C, Daar ES, Havlir D, Yeh E, Tawakol A, Lederman M, Currier JS, Stein JH. 2019. Safety and impact of low-dose methotrexate on endothelial function and inflammation in individuals with treated human immunodeficiency virus: AIDS clinical trials group study A5314. Clin Infect Dis 68:1877–1886.
204.
Freeman ML, Clagett BM, Moisi D, Yeh E, Morris CD, Ryu A, Rodriguez B, Stein JH, Deeks SG, Currier JS, Hsue PY, Anthony DD, Calabrese LH, Ribaudo HJ, Lederman MM. 2022. Methotrexate inhibits T cell proliferation but not inflammatory cytokine expression to modulate immunity in people living with HIV. Front Immunol 13:924718.
205.
Bundhun PK, Pursun M, Huang WQ. 2017. Does infection with human immunodeficiency virus have any impact on the cardiovascular outcomes following percutaneous coronary intervention?: a systematic review and meta-analysis. BMC Cardiovasc Disord 17:190.
206.
Hatleberg CI, Ryom L, El-Sadr W, Mocroft A, Reiss P, De Wit S, Dabis F, Pradier C, d’Arminio Monforte A, Kovari H, Law M, Lundgren JD, Sabin CA, Data Collection of Adverse Events of Anti-HIV drugs (D:A:D) Study group. 2018. Gender differences in the use of cardiovascular interventions in HIV-positive persons; the D:A:D study. J Int AIDS Soc 21:e25083.
207.
Singh V, Mendirichaga R, Savani GT, Rodriguez AP, Dabas N, Munagala A, Alfonso CE, Cohen MG, Elmariah S, Palacios IF. 2017. Coronary revascularization for acute myocardial infarction in the HIV population. J Interven Cardiology 30:405–414.
208.
Suchindran S, Regan S, Meigs JB, Grinspoon SK, Triant VA. 2014. Aspirin use for primary and secondary prevention in human immunodeficiency virus (HIV)-infected and HIV-uninfected patients. Open Forum Infect Dis 1:ofu076.
209.
van Zoest RA, van der Valk M, Wit FW, Vaartjes I, Kooij KW, Hovius JW, Prins M, Reiss P, AGEhIV Cohort Study Group. 2017. Suboptimal primary and secondary cardiovascular disease prevention in HIV-positive individuals on antiretroviral therapy. Eur J Prev Cardiol 24:1297–1307.
210.
Boccara F, Miantezila Basilua J, Mary-Krause M, Lang S, Teiger E, Steg PG, Funck-Brentano C, Girard P-M, Costagliola D, Cohen A, Guiguet M, on behalf the PACS-HIV investigators (Prognosis of Acute Coronary Syndrome in HIV-infected patients). 2017. Statin therapy and low-density lipoprotein cholesterol reduction in HIV-infected individuals after acute coronary syndrome: results from the PACS-HIV lipids Substudy. Am Heart J 183:91–101.
211.
Feinstein MJ, Nance RM, Delaney JAC, Heckbert SR, Budoff MJ, Drozd DR, Burkholder GA, Willig JH, Mugavero MJ, Mathews WC, Moore RD, Eron JJ, Napravnik S, Hunt PW, Geng E, Hsue P, Peter I, Lober WB, Crothers K, Grunfeld C, Saag MS, Kitahata MM, Lloyd-Jones DM, Crane HM. 2019. Mortality following myocardial infarction among HIV-infected persons: the center for AIDS research network of integrated clinical systems. (CNICS). BMC Med 17:149.
212.
Varga ZV, Ferdinandy P, Liaudet L, Pacher P. 2015. Drug-induced mitochondrial dysfunction and cardiotoxicity. Am J Physiol Heart Circ Physiol 309:H1453–67.
213.
Lewis W, Day BJ, Copeland WC. 2003. Mitochondrial toxicity of NRTI antiviral drugs: an integrated cellular perspective. Nat Rev Drug Discov 2:812–822.
214.
DAD Study Group, Friis-Møller N, Reiss P, Sabin CA, Weber R, Monforte A d, El-Sadr W, Thiébaut R, De Wit S, Kirk O, Fontas E, Law MG, Phillips A, Lundgren JD. 2007. Class of antiretroviral drugs and the risk of myocardial infarction. N Engl J Med 356:1723–1735.
215.
Ryom L, Lundgren JD, El-Sadr W, Reiss P, Kirk O, Law M, Phillips A, Weber R, Fontas E, d’ Arminio Monforte A, De Wit S, Dabis F, Hatleberg CI, Sabin C, Mocroft A, D:A:D study group. 2018. Cardiovascular disease and use of contemporary protease inhibitors: the D:A:D international prospective multicohort study. Lancet HIV 5:e291–e300.
216.
Elion RA, Althoff KN, Zhang J, Moore RD, Gange SJ, Kitahata MM, Crane HM, Drozd DR, Stein JH, Klein MB, Eron JJ, Silverberg MJ, Mathews WC, Justice AC, Sterling TR, Rabkin CS, Mayor AM, Klein DB, Horberg MA, Bosch RJ, Eyawo O, Palella FJ, North American AIDS Cohort Collaboration on Research and Design of IeDEA. 2018. Recent abacavir use increases risk of type 1 and type 2 myocardial Infarctions among adults with HIV. J Acquir Immune Defic Syndr 78:62–72.
217.
D:A:D Study Group, Sabin CA, Worm SW, Weber R, Reiss P, El-Sadr W, Dabis F, De Wit S, Law M, D’Arminio Monforte A, Friis-Møller N, Kirk O, Pradier C, Weller I, Phillips AN, Lundgren JD. 2008. Use of nucleoside reverse transcriptase inhibitors and risk of myocardial infarction in HIV-infected patients enrolled in the D:A:D study: a multi-cohort collaboration. Lancet 371:1417–1426.
218.
Marcus JL, Neugebauer RS, Leyden WA, Chao CR, Xu L, Quesenberry CP, Klein DB, Towner WJ, Horberg MA, Silverberg MJ. 2016. Use of abacavir and risk of cardiovascular disease among HIV-infected individuals. J Acquir Immune Defic Syndr 71:413–419.
219.
Sabin CA, Reiss P, Ryom L, Phillips AN, Weber R, Law M, Fontas E, Mocroft A, de Wit S, Smith C, Dabis F, d’Arminio Monforte A, El-Sadr W, Lundgren JD, D:A:D Study Group. 2016. Is there continued evidence for an association between abacavir usage and myocardial infarction risk in individuals with HIV? A cohort collaboration. BMC Med 14:61.
220.
Desai M, Joyce V, Bendavid E, Olshen RA, Hlatky M, Chow A, Holodniy M, Barnett P, Owens DK. 2015. Risk of cardiovascular events associated with current exposure to HIV antiretroviral therapies in a US veteran population. Clin Infect Dis 61:445–452.
221.
Choi AI, Vittinghoff E, Deeks SG, Weekley CC, Li Y, Shlipak MG. 2011. Cardiovascular risks associated with abacavir and tenofovir exposure in HIV-infected persons. AIDS 25:1289–1298.
222.
Bedimo RJ, Westfall AO, Drechsler H, Vidiella G, Tebas P. 2011. Abacavir use and risk of acute myocardial infarction and cerebrovascular events in the highly active antiretroviral therapy era. Clin Infect Dis 53:84–91.
223.
Ding X, Andraca-Carrera E, Cooper C, Miele P, Kornegay C, Soukup M, Marcus KA. 2012. No association of abacavir use with myocardial infarction: findings of an FDA meta-analysis. J Acquir Immune Defic Syndr 61:441–447.
224.
Sabin CA, Ryom L, d’Arminio Monforte A, Hatleberg CI, Pradier C, El-Sadr W, Kirk O, Weber R, Phillips AN, Mocroft A, Bonnet F, Law M, de Wit S, Reiss P, Lundgren JD. 2018. Abacavir use and risk of recurrent myocardial infarction. AIDS 32:79–88.
225.
Nan C, Shaefer M, Urbaityte R, Oyee J, Hopking J, Ragone L, Perger T, Win B, Vangerow H, McCoig C, Vannappagari V. 2018. Abacavir use and risk for myocardial infarction and cardiovascular events: pooled analysis of data from clinical trials. Open Forum Infect Dis 5:ofy086.
226.
Kheloussi S. 2018. Considerations in the approach to appropriate statin selection, on U.S. pharmacist. Available from: https://www.uspharmacist.com/article/considerations-in-the-approach-to-appropriate-statin-selection. Retrieved 11 Nov 2023.
227.
Wiggins BS, Saseen JJ, Page RL II, Reed BN, Sneed K, Kostis JB, Lanfear D, Virani S, Morris PB. 2016. Recommendations for management of clinically significant drug-drug interactions with statins and select agents used in patients with cardiovascular disease: a scientific statement from the American heart association. Circulation 134:e468–e495.
228.
Stone NJ, Robinson JG, Lichtenstein AH, Bairey Merz CN, Blum CB, Eckel RH, Goldberg AC, Gordon D, Levy D, Lloyd-Jones DM, et al. 2014. 2013 ACC/AHA guideline on the treatment of blood cholesterol to reduce atherosclerotic cardiovascular risk in adults: a report of the American college of cardiology/American heart association task force on practice guidelines. Circulation 129:S1–45.
229.
Williams D, Feely J. 2002. Pharmacokinetic-pharmacodynamic drug interactions with HMG-Coa reductase inhibitors. Clinical Pharmacokinetics 41:343–370.
230.
LaRosa JC, Grundy SM, Waters DD, Shear C, Barter P, Fruchart J-C, Gotto AM, Greten H, Kastelein JJP, Shepherd J, Wenger NK, Treating to New Targets (TNT) Investigators. 2005. Intensive lipid lowering with atorvastatin in patients with stable coronary disease. N Engl J Med 352:1425–1435.
231.
Pedersen TR, Faergeman O, Kastelein JJP, Olsson AG, Tikkanen MJ, Holme I, Larsen ML, Bendiksen FS, Lindahl C, Szarek M, Tsai J, Incremental Decrease in End Points Through Aggressive Lipid Lowering (IDEAL) Study Group. 2005. High-dose atorvastatin vs usual-dose simvastatin for secondary prevention after myocardial Infarctionthe IDEAL study: a randomized controlled trial. JAMA 294:2437–2445.
232.
Lipitor (atorvastatin) package insert. Pfizer, Inc, New York, NY.
233.
Chastain DB, Stover KR, Riche DM. 2017. Evidence-based review of statin use in patients with HIV on antiretroviral therapy. J Clin Transl Endocrinol 8:6–14.
234.
Myerson M, Malvestutto C, Aberg JA. 2015. Management of lipid disorders in patients living with HIV. J Clin Pharmacol 55:957–974.
235.
Fichtenbaum CJ, Gerber JG, Rosenkranz SL, Segal Y, Aberg JA, Blaschke T, Alston B, Fang F, Kosel B, Aweeka F. 2002. Pharmacokinetic interactions between protease inhibitors and statins in HIV seronegative volunteers: ACTG study A5047. AIDS 16:569–577.
236.
Sherman EM, Worley MV, Unger NR, Gauthier TP, Schafer JJ. 2015. Cobicistat: review of a pharmacokinetic enhancer for HIV infection. Clin Ther 37:1876–1893.
237.
Gerber JG, Rosenkranz SL, Fichtenbaum CJ, Vega JM, Yang A, Alston BL, Brobst SW, Segal Y, Aberg JA, AIDS Clinical Trials Group A5108 Team. 2005. Effect of efavirenz on the pharmacokinetics of simvastatin, atorvastatin, and pravastatin: results of AIDS clinical trials group 5108 study. J Acquir Immune Defic Syndr 39:307–312.
238.
Chastain DB, Henderson H, Stover KR. 2015. Epidemiology and management of antiretroviral-associated cardiovascular disease. Open AIDS J 9:23–37.
239.
Hsyu PH, Schultz-Smith MD, Lillibridge JH, Lewis RH, Kerr BM. 2001. Pharmacokinetic interactions between nelfinavir and 3-hydroxy-3-methylglutaryl coenzyme a reductase inhibitors atorvastatin and simvastatin. Antimicrob Agents Chemother 45:3445–3450.
240.
Saito Y. 2011. Pitavastatin: an overview. Atheroscler Suppl 12:271–276.
241.
Chauvin B, Drouot S, Barrail-Tran A, Taburet AM. 2013. Drug-drug interactions between HMG-Coa reductase inhibitors (statins) and antiviral protease inhibitors. Clin Pharmacokinet 52:815–831.
242.
Mevacor (Lovasatin) package insert. Merck & Co, Inc, Whitehouse Station, NJ.
243.
Downs JR, Clearfield M, Weis S, Whitney E, Shapiro DR, Beere PA, Langendorfer A, Stein EA, Kruyer W, Gotto AM. 1998. Primary prevention of acute coronary events with lovastatin in men and women with average cholesterol levelsresults of AFCAPS/Texcaps. JAMA 279:1615–1622.
244.
Wiggins BS, Lamprecht DG, Page RL, Saseen JJ. 2017. Recommendations for managing drug–drug interactions with statins and HIV medications. Am J Cardiovasc Drugs 17:375–389.
245.
Crestor (Rosuvastatin) package insert. AstraZeneca Pharmaceuticals LP, Wilmington, DE.
246.
Ridker PM, Danielson E, Fonseca FAH, Genest J, Gotto AM Jr, Kastelein JJP, Koenig W, Libby P, Lorenzatti AJ, MacFadyen JG, Nordestgaard BG, Shepherd J, Willerson JT, Glynn RJ, JUPITER Study Group. 2008. Rosuvastatin to prevent vascular events in men and women with elevated C-reactive protein. N Engl J Med 359:2195–2207.
247.
Zocor (simvastatin) package insert. Merck & Co, Inc, Whitehouse Station, NJ.
248.
Heart Protection Study Collaborative Group. 2002. MRC/BHF heart protection study of cholesterol lowering with simvastatin in 20 536 high-risk individuals: a randomised placebocontrolled trial. Lancet 360:7–22.
249.
Pravachol (pravastatin) package insert. Bristol-Myers Squibb Company, Princeton, NJ.
250.
Nakamura H, Arakawa K, Itakura H, Kitabatake A, Goto Y, Toyota T, Nakaya N, Nishimoto S, Muranaka M, Yamamoto A, Mizuno K, Ohashi Y, MEGA Study Group. 2006. Primary prevention of cardiovascular disease with pravastatin in Japan (MEGA study): a prospective randomised controlled trial. Lancet 368:1155–1163.
251.
Cannon CP, Braunwald E, McCabe CH, Rader DJ, Rouleau JL, Belder R, Joyal SV, Hill KA, Pfeffer MA, Skene AM, Pravastatin or Atorvastatin Evaluation and Infection Therapy-Thrombolysis in Myocardial Infarction 22 Investigators. 2004. Intensive versus moderate lipid lowering with statins after acute coronary syndromes. N Engl J Med 350:1495–1504.
252.
Lescol/ Lescol XL (Fluvastatin) package insert. Novartis Pharmaceuticals Corp, East Hanover, NJ.
253.
Hsu A, Granneman GR, Bertz RJ. 1998. Ritonavir. clinical pharmacokinetics and interactions with other anti-HIV agents. Clin Pharmacokinet 35:275–291.
254.
Long MC, King JR, Acosta EP. 2009. Pharmacologic aspects of new antiretroviral drugs. Curr HIV/AIDS Rep 6:43–50.
255.
Pasterkamp G, den Ruijter HM, Giannarelli C. 2022. False Utopia of one unifying description of the vulnerable atherosclerotic plaque: a call for recalibration that appreciates the diversity of mechanisms leading to atherosclerotic disease. Arterioscler Thromb Vasc Biol 42:e86–e95.
256.
Iorio A, Pozzi A, Senni M. 2017. Addressing the heterogeneity of heart failure in future randomized trials. Curr Heart Fail Rep 14:197–202.
257.
Shah SJ, Katz DH, Selvaraj S, Burke MA, Yancy CW, Gheorghiade M, Bonow RO, Huang CC, Deo RC. 2015. Phenomapping for novel classification of heart failure with preserved ejection fraction. Circulation 131:269–279.
258.
Wolsk E, Claggett B, Køber L, Pocock S, Yusuf S, Swedberg K, McMurray JJV, Granger CB, Pfeffer MA, Solomon SD. 2018. Contribution of cardiac and extra-cardiac disease burden to risk of cardiovascular outcomes varies by ejection fraction in heart failure. Eur J Heart Fail 20:504–510.
259.
Shah MR, Wong RP. 2015. The changing paradigm of HIV-related heart failure. Glob Heart 10:241–244.
260.
Cohen IS, Anderson DW, Virmani R, Reen BM, Macher AM, Sennesh J, DiLorenzo P, Redfield RR. 1986. Congestive cardiomyopathy in association with the acquired immunodeficiency syndrome. N Engl J Med 315:628–630.
261.
De Castro S, Migliau G, Silvestri A, D’amati G, Giannantoni P, Cartoni D, Kol A, Vullo V, Cirelli A. 1992. Heart involvement in AIDS: a prospective study during various stages of the disease. Eur Heart J 13:1452–1459.
262.
Herskowitz A, Vlahov D, Willoughby S, Chaisson RE, Schulman SP, Neumann DA, Baughman KL. 1993. Prevalence and incidence of left ventricular dysfunction in patients with human immunodeficiency virus infection. Am J Cardiol 71:955–958.
263.
Pugliese A, Isnardi D, Saini A, Scarabelli T, Raddino R, Torre D. 2000. Impact of highly active antiretroviral therapy in HIV-positive patients with cardiac involvement. J Infect 40:282–284.
264.
Ziaeian B, Fonarow GC. 2016. Epidemiology and aetiology of heart failure. Nat Rev Cardiol 13:368–378.
265.
Thiara DK, Liu CY, Raman F, Mangat S, Purdy JB, Duarte HA, Schmidt N, Hur J, Sibley CT, Bluemke DA, Hadigan C. 2015. Abnormal myocardial function is related to myocardial steatosis and diffuse myocardial fibrosis in HIV-infected adults. J Infect Dis 212:1544–1551.
266.
Holloway CJ, Ntusi N, Suttie J, Mahmod M, Wainwright E, Clutton G, Hancock G, Beak P, Tajar A, Piechnik SK, Schneider JE, Angus B, Clarke K, Dorrell L, Neubauer S. 2013. Comprehensive cardiac magnetic resonance imaging and spectroscopy reveal a high burden of myocardial disease in HIV patients. Circulation 128:814–822.
267.
Ntusi N, O’Dwyer E, Dorrell L, Wainwright E, Piechnik S, Clutton G, Hancock G, Ferreira V, Cox P, Badri M, Karamitsos T, Emmanuel S, Clarke K, Neubauer S, Holloway C. 2016. HIV-1-related cardiovascular disease is associated with chronic inflammation, frequent pericardial effusions, and probable myocardial edema. Circ Cardiovasc Imaging 9:e004430.
268.
Feinstein MJ, Poole B, Engel Gonzalez P, Pawlowski AE, Schneider D, Provias TS, Palella FJ, Achenbach CJ, Lloyd-Jones DM. 2018. Differences by HIV serostatus in coronary artery disease severity and likelihood of percutaneous coronary intervention following stress testing. J Nucl Cardiol 25:872–883.
269.
Feinstein MJ, Mitter SS, Yadlapati A, Achenbach CJ, Palella FJ, Gonzalez PE, Meyers S, Collins JD, Shah SJ, Lloyd-Jones DM. 2016. HIV-related myocardial vulnerability to infarction and coronary artery disease. J Am Coll Cardiol 68:2026–2027.
270.
Osobamiro O, Stempein-Otero A, Ssinabulya I, Longenecker C. 2022. Cardiac transplantation in people living with HIV: the global context. Heart 108:573–574.
271.
Birk SE, Baran DA, Campbell R, Herre JM, Sadatsafavi H, Yehya A. 2023. Clinical outcomes of ventricular assist device support by HIV infection status: an STS-INTERMACS analysis. J Heart Lung Transplant 42:1185–1193.
272.
Uriel N, Nahumi N, Colombo PC, Yuzefpolskaya M, Restaino SW, Han J, Thomas SS, Garan AR, Takayama H, Mancini DM, Naka Y, Jorde UP. 2014. Advanced heart failure in patients infected with human immunodeficiency virus: is there equal access to care? J Heart Lung Transplant 33:924–930.
273.
Hernandez RJ, Gilchrist J, Maning J, Grant J, Vincent LT, Ebner B, Olarte N, Olorunfemi O, Colombo RA. 2021. Abstract 14240: disparities in implementation of advanced heart failure therapies among patients with HIV: an analysis of the NIS database. Circulation 144:A14240–A14240.
274.
Hsu JC, Li Y, Marcus GM, Hsue PY, Scherzer R, Grunfeld C, Shlipak MG. 2013. Atrial fibrillation and atrial flutter in human immunodeficiency virus-infected persons: incidence, risk factors, and association with markers of HIV disease severity. J Am Coll Cardiol 61:2288–2295.
275.
Osuji N, Haberlen SA, Ashikaga H, Brown TT, Feinstein MJ, Witt MD, Magnani JW, Soliman EZ, Wu KC, Post WS. 2021. Association between human immunodeficiency virus serostatus and the prevalence of atrial fibrillation. Medicine (Baltimore) 100:e26663.
276.
Sanders JM, Steverson AB, Pawlowski AE, Schneider D, Achenbach CJ, Lloyd-Jones DM, Feinstein MJ, De Socio GV. 2018. Atrial arrhythmia prevalence and characteristics for human immunodeficiency virus-infected persons and matched uninfected controls. PLoS One 13:e0194754.
277.
Feinstein MJ, Haberlen SA, Ashikaga H, Palella FJ, Magnani JW, Budoff M, Berlacher K, D’Souza G, Brown T, Post WS, Wu KC. 2021. Ventricular ectopy and arrhythmia by HIV serostatus, viremia, and CD4+ cell count. Aids 35:846–849.
278.
Meyer A, Dandamudi S, Achenbach C, Lloyd-Jones D, Feinstein M. 2019. Ventricular ectopy and arrhythmia characteristics for persons living with HIV and uninfected controls. J Int Assoc Provid AIDS Care 18:2325958219852123.
279.
Koplan BA, Stevenson WG. 2009. Ventricular tachycardia and sudden cardiac death. Mayo Clin Proc 84:289–297.
280.
Tseng ZH, Moyers B, Secemsky EA, Havlir DV, Hsue PY. 2013. PR interval and sudden cardiac death in patients with HIV infection. J Infect Dis 207:199–200.
281.
Tseng ZH. 2019. Presumed sudden cardiac deaths among persons with HIV and heart failure. JACC Heart Fail 7:768–770.
282.
Ding Y, Zhu B, Lin H, Chen X, Shen W, Xu X, Shi R, Xu X, Zhao G, He N. 2020. HIV infection and electrocardiogram abnormalities: baseline assessment from the CHART cohort. Clin Microbiol Infect 26:1689.
283.
Semulimi AW, Kyazze AP, Kyalo E, Mukisa J, Batte C, Bongomin F, Ssinabulya I, Kirenga BJ, Okello E, Tibebu NS. 2023. Review of electrocardiographic abnormalities among people living with HIV in sub-Saharan Africa: s systematic review. PLoS One 18:e0283419.
284.
Butrous G. 2015. Human immunodeficiency virus–associated pulmonary arterial hypertension. Circulation 131:1361–1370.
285.
Coplan NL, Shimony RY, Ioachim HL, Wilentz JR, Posner DH, Lipschitz A, Ruden RA, Bruno MS, Sherrid MV, Gaetz H, Englard A, Kukin M, Packer M. 1990. Primary pulmonary hypertension associated with human immunodeficiency viral infection. Am J Med 89:96–99.
286.
Speich R, Jenni R, Opravil M, Pfab M, Russi EW. 1991. Primary pulmonary hypertension in HIV infection. Chest 100:1268–1271.
287.
Ding Y, He N. 2021. HIV and pulmonary hypertension: CD4 and viral load matter. Lancet Healthy Longev 2:e389–e390.
288.
Ogbuagu O, Segal-Maurer S, Ratanasuwan W, Avihingsanon A, Brinson C, Workowski K, Antinori A, Yazdanpanah Y, Trottier B, Wang H, Margot N, Dvory-Sobol H, Rhee MS, Baeten JM, Molina J-M, GS-US-200-4625 investigators. 2023. Efficacy and safety of the novel capsid inhibitor lenacapavir to treat multidrug-resistant HIV: week 52 results of a phase 2/3 trial. Lancet HIV 10:e497–e505.
289.
Autenrieth CS, Beck EJ, Stelzle D, Mallouris C, Mahy M, Ghys P, Lima VD. 2018. Global and regional trends of people living with HIV aged 50 and over: estimates and projections for 2000-2020. PLoS One 13:e0207005.

Author Bios

Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
Ilana Nazari is a second year student in the Northwestern University Medical Scientist Training Program. She received her Bachelor of Science degree from the Massachusetts Institute of Technology department of biological engineering. She has a budding clinical and research interest in cardiovascular disease and inflammatory processes.
Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
Division of Cardiology in the Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
Dr. Matthew Feinstein is a cardiologist, translational researcher, and Associate Professor at Northwestern University Feinberg School of Medicine (NUFSM). Dr. Feinstein’s roles at NUFSM also include Director of Student Research and Director of the Clinical and Translational Immunocardiology Program. His translational research laboratory focuses on immune plasticity in cardiovascular diseases. Dr. Feinstein was writing chair of the American Heart Association (AHA)’s Scientific Statement on HIV and Cardiovascular Disease. Other scientific leadership roles include immediate past chair of the AHA Prevention Science Committee, membership on the AHA Epidemiology and Prevention Council Leadership Committee, and service on the Inflammation Transformative Working Group for the AIDS Clinical Trials Group. Dr. Feinstein received an American Society of Clinical Investigation Young Physician-Scientist Award in 2019 and is PI on several active NIH project grants; he has served on several NIH study sections given expertise on inflammation and cardiovascular disease and HIV-associated cardiovascular diseases.

Information & Contributors

Information

Published In

cover image Clinical Microbiology Reviews
Clinical Microbiology Reviews
Volume 37Number 114 March 2024
eLocator: e00098-22
Editor: Graeme N. Forrest, Rush University, Chicago, Illinois, USA
PubMed: 38299802

History

Published online: 1 February 2024

Permissions

Request permissions for this article.

Keywords

  1. human immunodeficiency virus
  2. inflammation
  3. cardiovascular disease

Contributors

Authors

Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
Author Contributions: Conceptualization, Investigation, Methodology, Project administration, Writing – original draft, and Writing – review and editing.
Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
Division of Cardiology in the Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
Author Contributions: Conceptualization, Data curation, Formal analysis, Funding acquisition, Methodology, Project administration, Resources, Software, Supervision, Visualization, Writing – original draft, and Writing – review and editing.

Editor

Graeme N. Forrest
Editor
Rush University, Chicago, Illinois, USA

Notes

The authors declare no relationships with industry relevant to this work and no conflicts of interest.

Metrics & Citations

Metrics

Note:

  • For recently published articles, the TOTAL download count will appear as zero until a new month starts.
  • There is a 3- to 4-day delay in article usage, so article usage will not appear immediately after publication.
  • Citation counts come from the Crossref Cited by service.

Citations

If you have the appropriate software installed, you can download article citation data to the citation manager of your choice. For an editable text file, please select Medlars format which will download as a .txt file. Simply select your manager software from the list below and click Download.

View Options

Figures

Tables

Media

Share

Share

Share the article link

Share with email

Email a colleague

Share on social media

American Society for Microbiology ("ASM") is committed to maintaining your confidence and trust with respect to the information we collect from you on websites owned and operated by ASM ("ASM Web Sites") and other sources. This Privacy Policy sets forth the information we collect about you, how we use this information and the choices you have about how we use such information.
FIND OUT MORE about the privacy policy