Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2022 May 1.
Published in final edited form as: Curr Opin HIV AIDS. 2021 May 1;16(3):168–176. doi: 10.1097/COH.0000000000000678

CMV and Persistent Immune Activation in HIV

Samuel R Schnittman 1, Peter W Hunt 1
PMCID: PMC8238090  NIHMSID: NIHMS1684836  PMID: 33833209

Abstract

Purpose of review:

Despite antiretroviral therapy (ART)-mediated viral suppression, people with HIV (PWH) have increased morbidity and mortality. Immune activation and inflammation persist on antiretroviral therapy and predicts these complications. Over 90% of PWH have cytomegalovirus (CMV) co-infection, and CMV is considered a plausible contributor to this persistent immune activation.

Recent findings:

A detailed understanding of the link between CMV and multimorbidity is needed, particularly as research moves toward identifying potential targeted therapeutics to attenuate inflammation-mediated morbidity and mortality in treated HIV. We review the literature on the association between CMV and immune activation as well as multiple end-organ complications including cardiovascular disease, venous thromboembolic disease, metabolic complications, gastrointestinal dysfunction, central nervous system involvement, birth sex-related differences, and the relation to the HIV reservoir. We conclude with a discussion of ongoing therapeutic efforts to target CMV.

Summary:

As CMV is a plausible driver of multiple comorbidities through persistent immune activation in treated HIV, future research is needed and planned to address its causal role as well as to test novel therapeutics in this setting.

Keywords: CMV, inflammation, immune activation, cardiovascular disease, letermovir

Introduction

Among viruses, cytomegalovirus (CMV) has one of the most dynamic and comprehensive interactions with the human immune system. CMV elicits and maintains a high frequency of CMV-specific T cells – nearly 10% of the entire memory T cell repertoire - that are engaged in a lifelong effort to restrict CMV replication and prevent life-threatening disease [13]. As people age with CMV, repeated immune activation and response to CMV can drive T cells toward a more differentiated and senescent phenotype [46]. Senescent T cells are enriched for CMV-specific cells, and the abundance of senescent T cells has been associated with negative outcomes in older adults including frailty, cardiovascular disease, and systemic infections [713]. These inflammatory effector CD8+ T cells typically migrate to tissues where they—along with low-level CMV replication itself—may contribute to local inflammation and chronic disease. A recent review has addressed CMV and immune dysfunction, particularly with CMV’s interaction with human immunodeficiency virus (HIV) as a coinfection, which is further relevant given co-infection prevalence in HIV over 90% [14]. This review will focus on the various end-organ disease manifestations associated with CMV in treated HIV, as well as prior and ongoing interventional approaches aimed at CMV as one of the putative drivers of persistent immune activation.

CMV and Immune Activation

Asymptomatic CMV replication is induced by inflammation in vivo. It has long been recognized that inflammation can trigger lytic CMV replication in latently CMV-infected myeloid cells. Inflammatory signaling through TNFα receptors in latently CMV-infected myeloid cells activates the NF-κB pathway, which in turn binds to the immediate early enhancer region of CMV, initiating virus transcription [15, 16]. This phenomenon appears to explain why CMV DNA, which is typically undetectable in the peripheral blood in immunocompetent CMV-seropositive individuals, becomes detectable in inflammatory states such as sepsis, myocardial infarction, and even atopic dermatitis [15, 1721]. That said, asymptomatic CMV replication is also a cause of systemic inflammation in vivo. Even asymptomatic CMV infection in immunocompetent adults is associated with measurable increases in systemic inflammation [22]. One study in critically ill HIV-negative CMV-seropositive participants with sepsis and acute respiratory distress syndrome found that while ganciclovir therapy improved oxygenation and shortened the time requiring mechanical ventilation, even though it failed to significantly reduce plasma IL-6 levels (though this may not have been the most appropriate biomarker to reflect CMV-induced inflammation) [23]. In the context of treated HIV, CMV seropositive adults also have higher markers of inflammation, greater CD8+ T cell expansion, and lower CD4+/CD8+ T cell ratios than CMV-seronegative adults [2329]. We also previously assessed the contribution of asymptomatic CMV replication to systemic inflammation in a randomized placebo-controlled trial of valganciclovir in asymptomatic CMV-seropositive people with HIV (PWH) with incomplete CD4+ T cell recovery on antiretroviral therapy (ART) [30]. The trial showed a reduction in T cell immune activation in those randomized to valganciclovir after 8 weeks of therapy that was sustained for 4 weeks following treatment cessation. Recent analysis of samples from this trial further revealed nearly an entire quartile reduction in sTNFR2 as well as significant reductions in sCD163 and sCD14 levels in the valganciclovir arm [31, 32].

CMV and Cardiovascular Disease

CMV may play a modest role in cardiovascular disease (CVD) in the general population but is likely to play a far greater role in immunocompromised populations, including those with HIV. Mechanistically, CMV can replicate in macrophages and endothelial and smooth muscle cells within the vasculature, attracting inflammatory CX3CR1+ T cells and monocytes, which may all contribute to atherogenesis [24, 33]. Indeed, activated CD8+ T cells may promote not just atherosclerosis, but also monocyte tissue factor expression and coagulation via a TNF-dependent mechanism in non-human primate models of HIV infection [34]. In the general population, 82% of atherosclerotic plaques obtained from HIV-uninfected individuals undergoing endarterectomy had detectable CMV DNA [18, 35]. Observational data also suggest a modest role of CMV in increasing CVD risk in the general population; a recent meta-analysis including nearly 35,000 participants reported that CMV serostatus was associated with a 1.22-fold increased risk of incident CVD after adjustment for traditional risk factors [36]. A more recently published meta-analysis of 5 European cohorts did not find CMV to be associated with all-cause or cardiovascular mortality and more limited evidence of higher CMV IgG quartile as associated with all-cause mortality compared to seronegative controls [37].

The evidence for a role of CMV in contributing to vascular disease in immunocompromised populations – particularly those with T cell defects - is much stronger. In one clinical trial, just 28 days of ganciclovir appeared to cause a 2-fold decreased risk of post-transplant atherosclerosis 48 weeks after heart transplantation [38]. While associated with more subtle degrees of immunodeficiency than solid organ transplantation, treated HIV infection is associated with extremely high CMV prevalence (>90%) and much higher levels of mucosal CMV shedding than in HIV-uninfected CMV-seropositive individuals [39, 40]. Higher levels of CMV-specific CD8+ T cells and CMV-specific IgG are associated with atherosclerosis in treated HIV infection [11, 41, 42]. In fact, after adjusting for CMV-specific CD8+ T cell frequencies, HIV-infected participants no longer had greater atherosclerosis (by carotid intima-media thickness) than HIV-uninfected individuals, an early clue that CMV may mediate the increased CVD risk in HIV infection [42]. ART-suppressed CMV-seropositive PWH also had a 2.3-fold higher incidence of cardiovascular events than CMV-seronegative PWH in the ICONA cohort [43]. Recent work has demonstrated that not just CMV serostatus, but CMV IgG titer is associated with type 1 and type 2 myocardial infarction (MI), with a stepwise increase in hazard by IgG quartile [44]. It is also important to note that valganciclovir-mediated suppression of asymptomatic CMV replication (and potentially other herpesviruses) in our recent clinical trial of PWH with incomplete CD4+ T cell recovery also decreased inflammatory biomarkers that strongly predict MI and arterial inflammation by FDG-PET/CT in other studies of ART-suppressed PWH (i.e., sTNFR2, sCD163, and sICAM-1) [31, 4548].

CMV and Venous Thromboembolic Disease

CMV also stimulates thrombogenic activity on cells independent of vascular damage as well as cell-independent thrombin production, converting resting cells from an anticoagulant to a procoagulant state via factor Xa generation [9, 4951]. This may plausibly lead to a higher risk of venous thromboembolism (VTE) in treated HIV. In one published study on predictors of VTE in HIV addressing CMV, a prior history of CMV end-organ disease and CMV viremia were strong predictors of subsequent VTE in participants with AIDS on suppressive ART [52]. In our more recent case-cohort study of ART-suppressed PWH, CMV IgG titer was associated with incident VTE, with a stepwise increase in hazard by IgG quartile similar to MI above [44].

CMV and Metabolic Complications

CMV may also plausibly contribute to adipose tissue inflammation and fibrosis, insulin resistance, and visceral obesity. Type 2 diabetes (T2DM) is one of the most frequently occurring comorbidities in treated HIV infection, occurring at a rate of approximately one case for every 100 person-years in the NA-ACCORD cohort [53], much higher than the adult population without HIV [5457]. Several mechanistic studies in animal models suggest a causal role of CMV in contributing to these complications: in both murine and non-human primate models, CMV infects adipose tissue and induces inflammatory and fibrotic responses and insulin resistance [5862]. CMV is also known to preferentially replicate in adipose tissue in humans [63]. Inflammation in adipose tissue and the resultant fibrosis restricts adipocyte growth, preventing these cells from accumulating lipid during states of nutrient excess; consequently, lipids that should be stored in subcutaneous fat stores are forced into visceral adipose tissue, the liver, and epicardium—all harbingers of insulin resistance and the metabolic syndrome [64]. Less is known about the causal role of CMV in insulin resistance and visceral adiposity in humans. A recent meta-analysis including over 1,300 kidney transplant recipients found CMV infection carried a nearly 2-fold increased risk of post-transplant new-onset diabetes [65]. CMV has also been associated with metabolic syndrome in otherwise healthy normal weight women [66]. The plasma inflammatory markers that most strongly predict incident diabetes in treated HIV (sTNFR1, sTNFR2, sCD163) are also the same markers that were profoundly suppressed by treating asymptomatic CMV replication in our prior valganciclovir trial [30]. In a recent adipose tissue sampling study among people with treated HIV and CMV co-infection, a striking infiltration of subcutaneous fat by terminally differentiated effector T cells expressing CX3CR1+ and GPR56+, two markers frequently seen on CMV-specific T cells, was observed [67]. These cells tend to express a highly cytotoxic and proinflammatory transcriptome. While more data is needed to link CMV to these metabolic complications in PWH, these studies suggest that CMV may plausibly be associated with multiple metabolic complications in this setting.

CMV and Gut Dysfunction

The GI tract is a major site of CMV disease in immunocompromised individuals, including those with HIV. While CMV esophagitis and colitis are more common manifestations in those profoundly immunosuppressed, even asymptomatic CMV replication may contribute to microbial translocation across a leaky gut barrier, leading to other end-organ dysfunction [27]. CMV targets endothelial, stromal, and intestinal epithelial cells, weakening intercellular tight junctions that maintain barrier function. CMV persistence in rectosigmoid tissues of asymptomatic CMV seropositive individuals with both untreated and ART-suppressed HIV has been associated with gut epithelial barrier dysfunction [68]. In fact, in model systems independent of HIV, CMV disrupted tight junctions of polarized epithelial cells, enhancing barrier permeability and leading to immune cell infiltration [68]. Activated immune cells accelerate further disruption of the intestinal barrier, leading to persistent inflammation in the gut of ART-suppressed PWH [27, 69, 70]. This effect was mediated in part by CMV-induced inflammation and letermovir, an anti-CMV-specific drug, preserved barrier function [68]. In other observational studies, CMV seropositivity is strongly associated with circulating markers of microbial translocation, which have been linked to end-organ dysfunction [27]. Intriguingly, although CMV infection elicits a large and expanding pool of CMV-specific CD4+ and CD8+ T cells, CMV-specific CD8+ T cells appear to be quite rare in colorectal mucosa [5, 71]. The basis for the failure of CMV-specific T cells to traffic to the gut is unclear, but their near-absence or dysfunction likely contributes to CMV persistence and its associated systemic immune activation in this setting.

CMV and the Central Nervous System

The central nervous system (CNS) is a long-recognized site of CMV end-organ disease in advanced AIDS (e.g., retinitis, periventriculitis). CMV may also be relevant to CNS disease in the general population. CMV-infected individuals in the early stages of multiple sclerosis showed greater generalized brain atrophy over time than CMV-negative participants [72]. CMV may also play a role in psychiatric diseases like depression and sleep disturbance [73], and several earlier studies have linked psychological stress to CMV reactivation [7479]. In people with HIV, higher anti-CMV IgG titer was associated with greater neurocognitive impairment, similar to findings in HIV-uninfected elderly individuals [8083]. CMV DNA was detected in several cerebrospinal fluid (CSF) samples in the small, cross-sectional HIV study, but the role for migrating CMV-specific CD8+ T cells or low-level CMV replication in the brain has not been excluded. Additionally, higher plasma anti-CMV IgG concentrations were associated with higher HIV RNA and sCD163 in the CSF, indicating that the CMV-related immune response at least indirectly influences pathologic events in the CNS, perhaps driven by migration of activated monocytes or infected CD4 T-cells into this compartment. Some additional data from PWH has linked the immune activation pathways thought to be driven by CMV to depression in HIV [84]. Even in the absence of advanced immune suppression, mental health complications such as depression and neurocognitive impairment still occur more commonly in PWH than in the general population and seem to be less responsive to standard therapy [67, 8588]. Further work is needed to determine whether inhibiting low-level CMV shedding improves neuroinflammation, neuronal injury, neurocognitive performance, and mood.

CMV and Frailty

In addition to the above neurocognitive effects associated with CMV, chronic CMV infection has also been associated with frailty, in part through greater CMV-associated inflammation [8991]. Even with long-term effective ART, impairments in physical function and frailty are more common than expected among people aging with HIV and have been associated with an increased risk of falls, hospitalizations, and mortality [9294]. Furthermore, the combination of both HIV infection and impaired physical function is associated with a greater risk of mortality than the presence of HIV infection or impaired function alone [95, 96]. CMV seropositivity, as measured by qualitative or quantitative CMV IgG titers or CMV-associated T-cell responses have been associated with frailty in the general geriatric population, and frailty and physical function impairment in PWH [8991, 97].

Sex Differences in Asymptomatic CMV Infection

Women tend to have more robust innate immune responses to viral infections, with increased type I interferon production related to the X-chromosome’s TLR7 [98, 99]. Little is known about sex differences related to CMV in particular. Two US-based studies in women with HIV demonstrated relatively low levels of detection of CMV DNA in cervicovaginal lavage (3–7%) compared to men, but those rates have varied in studies outside the US [100103]. Women with ART-suppressed HIV have been found to have lower levels of CMV DNA (from oral, vaginal, or urine samples) compared to ART-suppressed men who have sex with men (in oral or semen samples) but without associated differences in cellular HIV DNA [104]. In a more recent study, women were observed to have significantly higher plasma CMV IgG titer compared to men after multivariate adjustment [44]. We do not know whether higher CMV IgG titers reflect a greater burden of CMV replication in tissues or a greater inflammatory response to it, but it is likely that there are clinically relevant sex-based differences in CMV and subsequent immune activation.

CMV and the HIV Reservoir

Chronic inflammation and immune activation contribute to maintenance of the HIV reservoir during ART [105, 106]. Recent studies have demonstrated that asymptomatic CMV seminal shedding is associated with higher levels of total HIV DNA in both ART-naïve individuals and in individuals suppressed on long-term ART [26, 107]. In a large longitudinal study of individuals followed since the earliest phase of HIV infection, CD4+ T cell associated HIV DNA in blood was associated with the frequency of detectable CMV DNA in blood cells over time [108]. A recent study has even linked subclinical CMV shedding measured in peripheral blood mononuclear cells to increasing molecular diversity of the HIV DNA reservoir [109]. Although the observational design of these studies does not allow causal inference, the findings do support the possibility that asymptomatic CMV replication drives local and systemic immune activation with a subsequent maintenance or increase in the latent HIV reservoir. Targeting drivers of chronic inflammation like CMV could thus potentially contribute to HIV curative strategies.

Therapeutic Approaches to Asymptomatic CMV Infection

Given the growing evidence for CMV as a plausible driver of persistent immune activation and associated end-organ disease, particularly in treated HIV, multiple therapeutic approaches to treat asymptomatic CMV infection are being evaluated in this setting. One prior trial to date has assessed the causal role of asymptomatic CMV infection in systemic inflammation in treated HIV [30]. In this study of treated PWH with incomplete CD4 recovery (all CD4 <350), participants (n=30) randomized to valganciclovir for 8 weeks were observed to have a nearly entire quartile reduction in sTNFR2 and significant reductions in sCD163 and sCD14 compared to placebo [30, 31]. Moreover, administration of valganciclovir led to a sustained reduction in CMV shedding after just 8 weeks that persisted for at least 4 weeks after discontinuation. Despite these findings, the study was limited in that only 70% of the participants had HIV RNA suppression on ART (though there was a significant effect on immune activation even in the ART-suppressed subset) and the sample size was small. There were several limitations that precluded moving forward with clinical endpoint trials of valganciclovir in this setting. First, valganciclovir’s toxicity (cytopenias and teratogenicity) creates challenges for long-term use. Second, insufficient evidence was generated to definitively conclude that key inflammatory pathways that predict disease outcome or cardiovascular surrogate markers were affected. The trial’s short 8-week duration also limited the analysis of longer-lived T cell subsets like effector and senescent T cells. TEMRA CD8+ T cells appear quite long-lived in simian immunodeficiency virus models using similar direct measures of cellular turnover [110, 111]. Thus, many months of suppression of CMV replication may be required to stop or reverse the negative contribution of CMV-specific effector CD8+ T cells to end-organ disease. Additionally, the trial was limited to individuals with incomplete CD4+ T cell recovery on ART, and it remains unclear whether inhibiting CMV shedding would benefit immune activation in individuals with normal CD4+ T cell count recovery on ART. Lastly, there were very few female participants in the trial, and as the effects of chronic viral infections on the immune system likely differ by sex with recent evidence revealing significantly higher CMV IgG titer in women after multivariate adjustment, it is unclear whether treating CMV would have the same effect in women [44].

The above issues have been specifically addressed in the forthcoming ACTG trial (A5383) of letermovir, a CMV DNA terminase complex inhibitor that is FDA-approved for prophylaxis of CMV infection and disease in adult CMV-seropositive recipients of allogeneic hematopoietic stem cell transplant (HSCT) [68, 112, 113]. Letermovir demonstrated significant benefit compared to placebo in regard to clinically significant CMV disease through week 24 post-HSCT (18.9% vs 44.3% cumulative rate, stratified log-rank p<0.001). Letermovir also has a better safety profile than valganciclovir from this existing data, and fewer adverse events in the letermovir arm were observed versus placebo in the trial among HSCT recipients. In a pending phase II, double-blind, randomized, placebo-controlled multicenter US trial, total of 180 participants 40 years and older with ART-mediated viral suppression for at least 4 years and who meet eligibility criteria will be randomized to receive letermovir or placebo for 48 weeks, followed by 12 weeks of observation on ART alone. Randomization will be stratified by CD4 count (<350 and >350) to allow for a more direct comparison to the earlier valganciclovir trial and to explore whether there may be a role for CMV suppression to reduce immune activation not just among those with incomplete CD4 T cell recovery, but also among those with preserved CD4 T cell counts. Participants will also be stratified by sex since sex differences in immune response to viral infections have been described. The primary outcome for this trial is a reduction in sTNFR2 with secondary outcomes related to drug tolerability, reduction in CMV mucosal shedding, and reduction in sCD163. Multiple sub-studies are planned to include measures of the proviral HIV reservoir, proteomics, adipose tissue aspiration, FDG-PET/CT assessment of arterial inflammation, lumbar puncture for markers of neuronal injury, and neurocognitive assessments.

A trial of a therapeutic CMV vaccine will also start soon in the ACTG to characterize the safety and immunogenicity of a modified vaccinia Ankara-based anti-CMV vaccine (A5355) [114]. A total of 90 participants will be randomized in a 2:1 ratio to receive either two injections of the trial vaccine or placebo at trial entry and at week 4. Eligibility criteria includes adults ages 18–65 years with both HIV and asymptomatic CMV co-infection who are ART-suppressed with current and nadir CD4 T cell counts >250 and >100, respectively. Primary endpoints will include cellular immunogenicity (anti-CMV CD8+ T cell responses between entry and study week 12) and week 48 plasma sTNFR2, with the hypothesis that the vaccine will stimulate B cell and T cell responses to generate a CMV-specific immune response that will decrease sub-clinical shedding, and thus lead to lower levels of systemic immune activation. Other cellular biomarkers will also be assessed.

Conclusion

CMV has learned to adapt to evade the human immune system, and while it may have modest or minimal clinical implications in the general population, it has been found to be significantly associated with numerous end-organ disease manifestations in those with varying degrees of immunosuppression. While more work is needed to link serum markers of CMV infection with the true burden of CMV disease in site-specific tissues, varying measures of CMV (serostatus, IgG titer, specific T cells, etc.) have been associated with numerous end-organ disease manifestations in PWH despite ART-mediated viral suppression, including cardiovascular disease, VTE, gastrointestinal and metabolic dysfunction, neurocognitive impairment, and other CNS disease. The one randomized trial to date assessing the causal role for CMV in persistent immune activation found evidence that treating CMV co-infection may lead to reduced levels of inflammation. Given this trial’s design and participant recruitment, more questions remain about the possibility of differential effects by sex as well as by degree of immune recovery. With this trial and the growing evidence base supporting CMV’s role as a putative driver in inflammation and end-organ disease in PWH, future trials are planned to more definitively assess this relationship as well as guide the development of pragmatic surrogate clinical endpoints, given CMV’s manifold effects. Finally, given the findings of CMV’s impact in the immunosuppressed population with more significant T cell immunodeficiency than treated HIV, these ongoing studies will be broadly pertinent to the solid organ transplant setting as well.

Key Points.

  • While antiretroviral therapy reduces morbidity and mortality in people with HIV, they face elevated rates of multiple comorbidities due at least in part to systemic inflammation.

  • CMV is a plausible root driver of persistent immune activation and inflammation in treated HIV.

  • Asymptomatic CMV infection in treated HIV is associated with numerous end-organ diseases, including cardiovascular disease, VTE, gastrointestinal and metabolic dysfunction, and neurocognitive impairment.

  • Only one trial to date of valganciclovir has assessed the causal role of asymptomatic CMV infection in treated HIV but was limited by drug toxicity, limited duration of treatment, and limited enrollment of key subgroups.

  • Future trials are planned to assess the causal role of asymptomatic CMV infection in treated HIV and to test novel oral and vaccine therapeutics in this setting.

Acknowledgements

Financial support and sponsorship

This study was supported by the following NIH grants: R38HL143581 (SRS), R01HL152957 (PWH), K24 AI145806 (PWH), R01 HL128156 (PWH), R01DK112304 (PWH), and P30 AI027763 (PWH).

Conflicts of interest

SRS has no conflicts of interest. PWH has received research funding from Gilead Sciences, honoraria from Gilead, Viiv, and Janssen, and consulting fees from Biotron and Viiv.

Abbreviations:

CMV

cytomegalovirus

HIV

human immunodeficiency virus

PWH

people with HIV

ART

antiretroviral therapy

CVD

cardiovascular disease

MI

myocardial infarction

VTE

venous thromboembolism

T2DM

type 2 diabetes mellitus

CNS

central nervous system

CSF

cerebrospinal fluid

HSCT

hematopoietic stem cell transplant

References

  • 1.Sylwester AW, Mitchell BL, Edgar JB, et al. Broadly targeted human cytomegalovirus-specific CD4+ and CD8+ T cells dominate the memory compartments of exposed subjects. J Exp Med 2005; 202:673–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Waldrop SL, Pitcher CJ, Peterson DM, Maino VC, Picker LJ. Determination of antigen-specific memory/effector CD4+ T cell frequencies by flow cytometry: evidence for a novel, antigen-specific homeostatic mechanism in HIV-associated immunodeficiency. J Clin Invest 1997; 99:1739–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3*.Mozzi A, Biolatti M, Cagliani R, et al. Past and ongoing adaptation of human cytomegalovirus to its host. PLoS Pathog 2020; 16:e1008476. [DOI] [PMC free article] [PubMed] [Google Scholar]; This study analyzed CMV evolution, showing that distinct genes were targeted by natural selection and generating a catalog of these adaptive variants.
  • 4.Di Benedetto S, Derhovanessian E, Steinhagen-Thiessen E, Goldeck D, Muller L, Pawelec G. Impact of age, sex and CMV-infection on peripheral T cell phenotypes: results from the Berlin BASE-II Study. Biogerontology 2015; 16:631–43. [DOI] [PubMed] [Google Scholar]
  • 5.Gordon CL, Miron M, Thome JJ, et al. Tissue reservoirs of antiviral T cell immunity in persistent human CMV infection. J Exp Med 2017; 214:651–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Fletcher JM, Vukmanovic-Stejic M, Dunne PJ, et al. Cytomegalovirus-specific CD4+ T cells in healthy carriers are continuously driven to replicative exhaustion. J Immunol 2005; 175:8218–25. [DOI] [PubMed] [Google Scholar]
  • 7.Appay V, Fastenackels S, Katlama C, et al. Old age and anti-cytomegalovirus immunity are associated with altered T-cell reconstitution in HIV-1-infected patients. AIDS 2011; 25:1813–22. [DOI] [PubMed] [Google Scholar]
  • 8.Deeks SG, Verdin E, McCune JM. Immunosenescence and HIV. Curr Opin Immunol 2012; 24:501–6. [DOI] [PubMed] [Google Scholar]
  • 9.Looney RJ, Falsey A, Campbell D, et al. Role of cytomegalovirus in the T cell changes seen in elderly individuals. Clin Immunol 1999; 90:213–9. [DOI] [PubMed] [Google Scholar]
  • 10.Deeks SG, Phillips AN. HIV infection, antiretroviral treatment, ageing, and non-AIDS related morbidity. Bmj 2009; 338:a3172. [DOI] [PubMed] [Google Scholar]
  • 11.Kaplan RC, Sinclair E, Landay AL, et al. T cell activation and senescence predict subclinical carotid artery disease in HIV-infected women. J Infect Dis 2011; 203:452–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Nieto FJ, Adam E, Sorlie P, et al. Cohort study of cytomegalovirus infection as a risk factor for carotid intimal-medial thickening, a measure of subclinical atherosclerosis. Circulation 1996; 94:922–7. [DOI] [PubMed] [Google Scholar]
  • 13.Spyridopoulos I, Martin-Ruiz C, Hilkens C, et al. CMV seropositivity and T-cell senescence predict increased cardiovascular mortality in octogenarians: results from the Newcastle 85+ study. Aging Cell 2016; 15:389–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Freeman ML, Lederman MM, Gianella S. Partners in Crime: The Role of CMV in Immune Dysregulation and Clinical Outcome During HIV Infection. Curr HIV/AIDS Rep 2016; 13:10–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Docke WD, Prosch S, Fietze E, et al. Cytomegalovirus reactivation and tumour necrosis factor. Lancet 1994; 343:268–9. [DOI] [PubMed] [Google Scholar]
  • 16.Stein J, Volk HD, Liebenthal C, Kruger DH, Prosch S. Tumour necrosis factor alpha stimulates the activity of the human cytomegalovirus major immediate early enhancer/promoter in immature monocytic cells. J Gen Virol 1993; 74 ( Pt 11):2333–8. [DOI] [PubMed] [Google Scholar]
  • 17.Docke WD, Kiessling C, Worm M, et al. Subclinical activation of latent cytomegalovirus (CMV) infection and anti-CMV immune response in patients with atopic dermatitis. Br J Dermatol 2003; 148:954–63. [DOI] [PubMed] [Google Scholar]
  • 18.Nikitskaya E, Lebedeva A, Ivanova O, et al. Cytomegalovirus-Productive Infection Is Associated With Acute Coronary Syndrome. J Am Heart Assoc 2016; 5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Kutza AS, Muhl E, Hackstein H, Kirchner H, Bein G. High incidence of active cytomegalovirus infection among septic patients. Clin Infect Dis 1998; 26:1076–82. [DOI] [PubMed] [Google Scholar]
  • 20.von Muller L, Klemm A, Durmus N, et al. Cellular immunity and active human cytomegalovirus infection in patients with septic shock. J Infect Dis 2007; 196:1288–95. [DOI] [PubMed] [Google Scholar]
  • 21.Limaye AP, Kirby KA, Rubenfeld GD, et al. Cytomegalovirus reactivation in critically ill immunocompetent patients. Jama 2008; 300:413–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Simanek AM, Dowd JB, Pawelec G, Melzer D, Dutta A, Aiello AE. Seropositivity to cytomegalovirus, inflammation, all-cause and cardiovascular disease-related mortality in the United States. PLoS One 2011; 6:e16103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Limaye AP, Stapleton RD, Peng L, et al. Effect of Ganciclovir on IL-6 Levels Among Cytomegalovirus-Seropositive Adults With Critical Illness: A Randomized Clinical Trial. JAMA 2017; 318:731–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Freeman ML, Mudd JC, Shive CL, et al. CD8 T-Cell Expansion and Inflammation Linked to CMV Coinfection in ART-treated HIV Infection. Clin Infect Dis 2016; 62:392–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Smith DM, Nakazawa M, Freeman ML, et al. Asymptomatic CMV Replication During Early Human Immunodeficiency Virus (HIV) Infection Is Associated With Lower CD4/CD8 Ratio During HIV Treatment. Clin Infect Dis 2016; 63:1517–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Gianella S, Massanella M, Richman DD, et al. Cytomegalovirus replication in semen is associated with higher levels of proviral HIV DNA and CD4+ T cell activation during antiretroviral treatment. J Virol 2014; 88:7818–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27*.Ramendra R, Isnard S, Lin J, et al. CMV seropositivity is associated with increased microbial translocation in people living with HIV and uninfected controls. Clin Infect Dis 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]; This study found that CMV serostatus and CMV IgG titer were associated with markers of gut microbial translocation after multivariate adjustment in people with HIV compared to HIV-negative controls.
  • 28*.Fabbiani M, Borghetti A, Squillace N, et al. Integrase Inhibitors Use and Cmv Infection Predict Immune Recovery in People Living with Hiv Starting First-Line Therapy. J Acquir Immune Defic Syndr 2020. [DOI] [PubMed] [Google Scholar]; This study observed that those with CMV serostatus was associated with a lower probability of CD4/CD8 ratio normalization.
  • 29*.Isnard S, Ramendra R, Lin J, et al. Anti-CMV IgG is linked to CD4 T-cell count decay in HIV elite controllers. Clin Infect Dis 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]; This study found that even in elite controllers, CMV IgG titer was associated with annual CD4+ T-cell count decay.
  • 30.Hunt PW, Martin JN, Sinclair E, et al. Valganciclovir reduces T cell activation in HIV-infected individuals with incomplete CD4+ T cell recovery on antiretroviral therapy. J Infect Dis 2011; 203:1474–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Beck-Engeser G, Maldarelli F, York VA. Valganciclovir reduces sTNF-R2 and vascular dysfunction markers in treated HIV. Abstract 643. Conference on Retroviruses and Opportunistic Infection (CROI); Seattle, WA: IAS-USA; 2019. [Google Scholar]
  • 32*.Sevilla S, Beck-Engeser G, York V, et al. Valganciclovir effects on gut and pulmonary epithelial barrier markers in treated HIV. Abstract 236. Conference on Retroviruses and Opportunistic Infection (CROI); Boston, MA: IAS-USA; 2020. [Google Scholar]; This abstract reported that treating asymptomatic CMV infection in treated HIV (with poor immune recovery) for 8 weeks led to significant reductions in sCD14 and LBP.
  • 33.Sacre K, Hunt PW, Hsue PY, et al. A role for cytomegalovirus-specific CD4+CX3CR1+ T cells and cytomegalovirus-induced T-cell immunopathology in HIV-associated atherosclerosis. AIDS 2012; 26:805–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34*.Freeman ML, Panigrahi S, Chen B, et al. CD8+ T-Cell-Derived Tumor Necrosis Factor Can Induce Tissue Factor Expression on Monocytes. J Infect Dis 2019; 220:73–7. [DOI] [PMC free article] [PubMed] [Google Scholar]; This study found that activated CD8+ T cells induced monocytes to express tissue factor, presenting a role for T-cell activation in cardiovascular disease in treated HIV.
  • 35.Xenaki E, Hassoulas J, Apostolakis S, Sourvinos G, Spandidos DA. Detection of cytomegalovirus in atherosclerotic plaques and nonatherosclerotic arteries. Angiology 2009; 60:504–8. [DOI] [PubMed] [Google Scholar]
  • 36.Wang H, Peng G, Bai J, et al. Cytomegalovirus Infection and Relative Risk of Cardiovascular Disease (Ischemic Heart Disease, Stroke, and Cardiovascular Death): A Meta-Analysis of Prospective Studies Up to 2016. J Am Heart Assoc 2017; 6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37*.Chen S, Pawelec G, Trompet S, et al. Associations of Cytomegalovirus Infection With All-Cause and Cardiovascular Mortality in Multiple Observational Cohort Studies of Older Adults. J Infect Dis 2021; 223:238–46. [DOI] [PMC free article] [PubMed] [Google Scholar]; In this large study of HIV-negative individuals from Europe, CMV serostatus was not associated with all-cause or cardiovascular mortality after multivariate adjustment, while those in the highest CMV IgG quartile may have a modestly elevated risk of mortality compared to CMV seronegatives, concluding that CMV may play a negligible role in the general population.
  • 38.Valantine HA, Gao SZ, Menon SG, et al. Impact of prophylactic immediate posttransplant ganciclovir on development of transplant atherosclerosis: a post hoc analysis of a randomized, placebo-controlled study. Circulation 1999; 100:61–6. [DOI] [PubMed] [Google Scholar]
  • 39.Gianella S, Smith DM, Vargas MV, et al. Shedding of HIV and human herpesviruses in the semen of effectively treated HIV-1-infected men who have sex with men. Clin Infect Dis 2013; 57:441–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Gianella S, Massanella M, Wertheim JO, Smith DM. The Sordid Affair Between Human Herpesvirus and HIV. J Infect Dis 2015; 212:845–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Kaplan RC, Sinclair E, Landay AL, et al. T cell activation predicts carotid artery stiffness among HIV-infected women. Atherosclerosis 2011; 217:207–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Hsue PY, Hunt PW, Sinclair E, et al. Increased carotid intima-media thickness in HIV patients is associated with increased cytomegalovirus-specific T-cell responses. AIDS 2006; 20:2275–83. [DOI] [PubMed] [Google Scholar]
  • 43.Lichtner M, Cicconi P, Vita S, et al. 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 2015; 211:178–86. [DOI] [PubMed] [Google Scholar]
  • 44*.Schnittman SR, Beck-Engeser G, Shigenaga JK. Sex modifies the association between inflammation and vascular events in treated HIV. Abstract 98. Conference on Retroviruses and Opportunistic Infections (CROI); Virtual: IAS-USA; 2021. [Google Scholar]; In this abstract, the authors found a CMV seropositivity rate of 97% in participants with treated HIV across the US, CMV IgG titer to be significantly elevated in women compared to men with treated HIV, CMV IgG titer to be associated with type 1 myocardial infarction and venous thromboembolism, with increasing risk for every IgG quartile increase compared to the lowest CMV IgG quartile.
  • 45.Subramanian S, Tawakol A, Burdo TH, et al. Arterial inflammation in patients with HIV. JAMA 2012; 308:379–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Tenorio AR, Zheng Y, Bosch RJ, et al. Soluble markers of inflammation and coagulation but not T-cell activation predict non-AIDS-defining morbid events during suppressive antiretroviral treatment. J Infect Dis 2014; 210:1248–59. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Funderburg NT, Jiang Y, Debanne SM, et al. Rosuvastatin treatment reduces markers of monocyte activation in HIV-infected subjects on antiretroviral therapy. Clin Infect Dis 2014; 58:588–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Funderburg NT, Jiang Y, Debanne SM, et al. Rosuvastatin Reduces Vascular Inflammation and T-cell and Monocyte Activation in HIV-Infected Subjects on Antiretroviral Therapy. J Acquir Immune Defic Syndr 2015; 68:396–404. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Etingin OR, Silverstein RL, Friedman HM, Hajjar DP. Viral activation of the coagulation cascade: molecular interactions at the surface of infected endothelial cells. Cell 1990; 61:657–62. [DOI] [PubMed] [Google Scholar]
  • 50.Visser MR, Tracy PB, Vercellotti GM, Goodman JL, White JG, Jacob HS. Enhanced thrombin generation and platelet binding on herpes simplex virus-infected endothelium. Proc Natl Acad Sci U S A 1988; 85:8227–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Sutherland MR, Raynor CM, Leenknegt H, Wright JF, Pryzdial EL. Coagulation initiated on herpesviruses. Proc Natl Acad Sci U S A 1997; 94:13510–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Musselwhite LW, Sheikh V, Norton TD, et al. Markers of endothelial dysfunction, coagulation and tissue fibrosis independently predict venous thromboembolism in HIV. Aids 2011; 25:787–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53*.Rebeiro PF, Jenkins CA, Bian A, et al. Risk of Incident Diabetes Mellitus, Weight Gain, and Their Relationships With Integrase Inhibitor–Based Initial Antiretroviral Therapy Among Persons With Human Immunodeficiency Virus in the United States and Canada. Clinical Infectious Diseases 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]; This study in the NA-ACCORD cohort found type 2 diabetes mellitus to occur at a rate of one case for every 100 person-years in people with HIV, significantly higher than the adult population without HIV.
  • 54.Hernandez-Romieu AC, Garg S, Rosenberg ES, Thompson-Paul AM, Skarbinski J. Is diabetes prevalence higher among HIV-infected individuals compared with the general population? Evidence from MMP and NHANES 2009–2010. BMJ Open Diabetes Res Care 2017; 5:e000304. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Brown TT, Cole SR, Li X, et al. Antiretroviral therapy and the prevalence and incidence of diabetes mellitus in the multicenter AIDS cohort study. Arch Intern Med 2005; 165:1179–84. [DOI] [PubMed] [Google Scholar]
  • 56.De Wit S, Sabin CA, Weber R, et al. Incidence and risk factors for new-onset diabetes in HIV-infected patients: the Data Collection on Adverse Events of Anti-HIV Drugs (D:A:D) study. Diabetes Care 2008; 31:1224–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Capeau J, Bouteloup V, Katlama C, et al. Ten-year diabetes incidence in 1046 HIV-infected patients started on a combination antiretroviral treatment. AIDS 2012; 26:303–14. [DOI] [PubMed] [Google Scholar]
  • 58.Papadimitriou JM, Shellam GR, Allan JE. The effect of the beige mutation on infection with murine cytomegalovirus: histopathologic studies. Am J Pathol 1982; 108:299–309. [PMC free article] [PubMed] [Google Scholar]
  • 59.Iwasaki T, Tashiro A, Satodate R, Sata T, Kurata T. Acute pancreatitis with cytomegalovirus infection. Acta Pathol Jpn 1987; 37:1661–8. [DOI] [PubMed] [Google Scholar]
  • 60.Price P, Eddy KS, Papadimitriou JM, Robertson TA, Shellam GR. Cytomegalovirus infection of adipose tissues induces steatitis in adult mice. Int J Exp Pathol 1990; 71:557–71. [PMC free article] [PubMed] [Google Scholar]
  • 61*.Contreras NA, Sitnik KM, Jeftic I, Coplen CP, Cicin-Sain L, Nikolich-Zugich J. Life-long control of cytomegalovirus (CMV) by T resident memory cells in the adipose tissue results in inflammation and hyperglycemia. PLoS Pathog 2019; 15:e1007890. [DOI] [PMC free article] [PubMed] [Google Scholar]; CMV was found to infect the adipose tissue during acute CMV infection in mice, leading to a both lifelong CD8 T-cell immune response largely driven by tissue-resident T cells as well as evidence of metabolic changes within the adipose tissue itself that could plausibly lead to systemic metabolic complications.
  • 62.Senchenkov E, Khoretonenko MV, Leskov IL, Ostanin DV, Stokes KY. P-selectin mediates the microvascular dysfunction associated with persistent cytomegalovirus infection in normocholesterolemic and hypercholesterolemic mice. Microcirculation 2011; 18:452–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Shnayder M, Nachshon A, Krishna B, et al. Defining the Transcriptional Landscape during Cytomegalovirus Latency with Single-Cell RNA Sequencing. MBio 2018; 9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Hepler C, Gupta RK. The expanding problem of adipose depot remodeling and postnatal adipocyte progenitor recruitment. Mol Cell Endocrinol 2017; 445:95–108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Einollahi B, Motalebi M, Salesi M, Ebrahimi M, Taghipour M. The impact of cytomegalovirus infection on new-onset diabetes mellitus after kidney transplantation: a review on current findings. J Nephropathol 2014; 3:139–48. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Fleck-Derderian S, McClellan W, Wojcicki JM. The association between cytomegalovirus infection, obesity, and metabolic syndrome in U.S. adult females. Obesity (Silver Spring) 2017; 25:626–33. [DOI] [PubMed] [Google Scholar]
  • 67*.Wanjalla CN, McDonnell WJ, Barnett L, et al. Adipose Tissue in Persons With HIV Is Enriched for CD4(+) T Effector Memory and T Effector Memory RA(+) Cells, Which Show Higher CD69 Expression and CD57, CX3CR1, GPR56 Co-expression With Increasing Glucose Intolerance. Front Immunol 2019; 10:408. [DOI] [PMC free article] [PubMed] [Google Scholar]; In a small cohort of people with treated HIV, the authors identified that the adipose tissue is a reservoir of pro-inflammatory CD4+ and CD8+ T-cell subsets (TEM and TEMRA) as well as a specific population of CD4+ T cells enriched in the subcutaneous adipose tissue of those with HIV and type 2 diabetes that may contribute to inflammation in adipose tissue and lead to metabolic complications in people with HIV.
  • 68.Maidji E, Somsouk M, Rivera JM, Hunt PW, Stoddart CA. Replication of CMV in the gut of HIV-infected individuals and epithelial barrier dysfunction. PLoS Pathog 2017; 13:e1006202. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Brenchley JM, Price DA, Schacker TW, et al. Microbial translocation is a cause of systemic immune activation in chronic HIV infection. Nat Med 2006; 12:1365–71. [DOI] [PubMed] [Google Scholar]
  • 70*.Mehraj V, Ramendra R, Isnard S, et al. Circulating (1-->3)-beta-D-glucan Is Associated With Immune Activation During Human Immunodeficiency Virus Infection. Clin Infect Dis 2020; 70:232–41. [DOI] [PMC free article] [PubMed] [Google Scholar]; This cross-sectional analysis observed elevated beta-d-glucan levels that correlated with other inflammatory biomarkers, suggesting fungal microbial translocation may contribute to persistent immune activation in treated HIV, which could be limited by early ART initiation.
  • 71.Shacklett BL, Cox CA, Sandberg JK, Stollman NH, Jacobson MA, Nixon DF. Trafficking of human immunodeficiency virus type 1-specific CD8+ T cells to gut-associated lymphoid tissue during chronic infection. J Virol 2003; 77:5621–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Zivadinov R, Chin J, Horakova D, et al. Humoral responses to herpesviruses are associated with neurodegeneration after a demyelinating event: results from the multi-center set study. J Neuroimmunol 2014; 273:58–64. [DOI] [PubMed] [Google Scholar]
  • 73*.Zheng H, Bergamino M, Ford BN, et al. Replicable association between human cytomegalovirus infection and reduced white matter fractional anisotropy in major depressive disorder. Neuropsychopharmacology 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]; Among HIV-negative individuals, this study observed that CMV infection is associated with cortical white matter damage that is associated with major depressive disorder, suggesting CMV as a possible target in the subgroup of individuals who are CMV seropositive and suffer from major depressive disorder.
  • 74.Simanek AM, Cheng C, Yolken R, Uddin M, Galea S, Aiello AE. Herpesviruses, inflammatory markers and incident depression in a longitudinal study of Detroit residents. Psychoneuroendocrinology 2014; 50:139–48. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Glaser R, Kiecolt-Glaser JK, Speicher CE, Holliday JE. Stress, loneliness, and changes in herpesvirus latency. J Behav Med 1985; 8:249–60. [DOI] [PubMed] [Google Scholar]
  • 76.Janicki-Deverts D, Cohen S, Doyle WJ, Marsland AL, Bosch J. Childhood environments and cytomegalovirus serostatus and reactivation in adults. Brain Behav Immun 2014; 40:174–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Shirtcliff EA, Coe CL, Pollak SD. Early childhood stress is associated with elevated antibody levels to herpes simplex virus type 1. Proc Natl Acad Sci U S A 2009; 106:2963–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Fagundes CP, Glaser R, Malarkey WB, Kiecolt-Glaser JK. Childhood adversity and herpesvirus latency in breast cancer survivors. Health Psychol 2013; 32:337–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79*.Zheng H, Ford BN, Bergamino M, et al. A hidden menace? Cytomegalovirus infection is associated with reduced cortical gray matter volume in major depressive disorder. Mol Psychiatry 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]; Among HIV-negative individuals, this study observed an association between CMV serostatus and reduced cortical gray matter in those with major depressive disorder compared to CMV seronegative participants.
  • 80.Letendre S, Bharti A, Perez-Valero I, et al. Higher Anti-Cytomegalovirus Immunoglobulin G Concentrations Are Associated With Worse Neurocognitive Performance During Suppressive Antiretroviral Therapy. Clin Infect Dis 2018; 67:770–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Roberts ET, Haan MN, Dowd JB, Aiello AE. Cytomegalovirus antibody levels, inflammation, and mortality among elderly Latinos over 9 years of follow-up. Am J Epidemiol 2010; 172:363–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Couturier J, Agarwal N, Nehete PN, et al. Infectious SIV resides in adipose tissue and induces metabolic defects in chronically infected rhesus macaques. Retrovirology 2016; 13:30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Brunt SJ, Cysique LA, Lee S, Burrows S, Brew BJ, Price P. Short Communication: Do Cytomegalovirus Antibody Levels Associate with Age-Related Syndromes in HIV Patients Stable on Antiretroviral Therapy? AIDS Res Hum Retroviruses 2016; 32:567–72. [DOI] [PubMed] [Google Scholar]
  • 84.Martinez P, Tsai AC, Muzoora C, et al. Reversal of the Kynurenine pathway of tryptophan catabolism may improve depression in ART-treated HIV-infected Ugandans. J Acquir Immune Defic Syndr 2014; 65:456–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Gray KS, Collins CM, Speck SH. Characterization of omental immune aggregates during establishment of a latent gammaherpesvirus infection. PLoS One 2012; 7:e43196. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Heaton RK, Clifford DB, Franklin DR Jr., et al. HIV-associated neurocognitive disorders persist in the era of potent antiretroviral therapy: CHARTER Study. Neurology 2010; 75:2087–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Ellis RJ, Letendre S, Vaida F, et al. Randomized trial of central nervous system-targeted antiretrovirals for HIV-associated neurocognitive disorder. Clin Infect Dis 2014; 58:1015–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Sacktor N, Miyahara S, Deng L, et al. Minocycline treatment for HIV-associated cognitive impairment: results from a randomized trial. Neurology 2011; 77:1135–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Margolick JB, Bream JH, Nilles TL, et al. Relationship Between T-Cell Responses to CMV, Markers of Inflammation, and Frailty in HIV-uninfected and HIV-infected Men in the Multicenter AIDS Cohort Study. J Infect Dis 2018; 218:249–58. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Schmaltz HN, Fried LP, Xue QL, Walston J, Leng SX, Semba RD. Chronic cytomegalovirus infection and inflammation are associated with prevalent frailty in community-dwelling older women. J Am Geriatr Soc 2005; 53:747–54. [DOI] [PubMed] [Google Scholar]
  • 91.Wang GC, Kao WH, Murakami P, et al. Cytomegalovirus infection and the risk of mortality and frailty in older women: a prospective observational cohort study. Am J Epidemiol 2010; 171:1144–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Hawkins KL, Zhang L, Ng DK, et al. Abdominal obesity, sarcopenia, and osteoporosis are associated with frailty in men living with and without HIV. Aids 2018; 32:1257–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Tassiopoulos K, Abdo M, Wu K, et al. Frailty is strongly associated with increased risk of recurrent falls among older HIV-infected adults. AIDS 2017; 31:2287–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Erlandson KM, Perez J, Abdo M, et al. Frailty, Neurocognitive Impairment, or Both in Predicting Poor Health Outcomes Among Adults Living With Human Immunodeficiency Virus. Clin Infect Dis 2019; 68:131–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Greene M, Covinsky K, Astemborski J, et al. The relationship of physical performance with HIV disease and mortality. AIDS 2014; 28:2711–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Piggott DA, Varadhan R, Mehta SH, et al. Frailty, Inflammation, and Mortality Among Persons Aging With HIV Infection and Injection Drug Use. J Gerontol A Biol Sci Med Sci 2015; 70:1542–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Erlandson KM, Allshouse AA, Rapaport E, et al. Physical function impairment of older, HIV-infected adults is associated with cytomegalovirus immunoglobulin response. AIDS Res Hum Retroviruses 2015; 31:905–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Meier A, Chang JJ, Chan ES, et al. Sex differences in the Toll-like receptor-mediated response of plasmacytoid dendritic cells to HIV-1. Nat Med 2009; 15:955–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Chang JJ, Woods M, Lindsay RJ, et al. Higher expression of several interferon-stimulated genes in HIV-1-infected females after adjusting for the level of viral replication. J Infect Dis 2013; 208:830–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Schoenfisch AL, Dollard SC, Amin M, et al. Cytomegalovirus (CMV) shedding is highly correlated with markers of immunosuppression in CMV-seropositive women. J Med Microbiol 2011; 60:768–74. [DOI] [PubMed] [Google Scholar]
  • 101.Mitchell C, Hitti J, Paul K, et al. Cervicovaginal shedding of HIV type 1 is related to genital tract inflammation independent of changes in vaginal microbiota. AIDS Res Hum Retroviruses 2011; 27:35–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Mostad SB, Kreiss JK, Ryncarz AJ, et al. Cervical shedding of cytomegalovirus in human immunodeficiency virus type 1-infected women. J Med Virol 1999; 59:469–73. [PubMed] [Google Scholar]
  • 103.Gianella S, Redd AD, Grabowski MK, et al. Vaginal Cytomegalovirus Shedding Before and After Initiation of Antiretroviral Therapy in Rakai, Uganda. J Infect Dis 2015; 212:899–903. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104*.Gianella S, Tran SM, Morris S, et al. Sex Differences in CMV Replication and HIV Persistence During Suppressive ART. Open Forum Infect Dis 2020; 7:ofaa289. [DOI] [PMC free article] [PubMed] [Google Scholar]; Among men who have sex with men with treated HIV, the study identified that women had reduced cellular HIV RNA and less CMV DNA but similar HIV DNA levels compared to men, suggesting sex-specific differences in CMV and possibly in relation to the HIV reservoir.
  • 105.Hatano H Immune activation and HIV persistence: considerations for novel therapeutic interventions. Curr Opin HIV AIDS 2013; 8:211–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Christensen-Quick A, Vanpouille C, Lisco A, Gianella S. Cytomegalovirus and HIV Persistence: Pouring Gas on the Fire. AIDS Res Hum Retroviruses 2017; 33:S23–S30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Gianella S, Mehta SR, Strain MC, et al. Impact of seminal cytomegalovirus replication on HIV-1 dynamics between blood and semen. J Med Virol 2012; 84:1703–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Gianella S, Anderson CM, Var SR, et al. Replication of Human Herpesviruses Is Associated with Higher HIV DNA Levels during Antiretroviral Therapy Started at Early Phases of HIV Infection. J Virol 2016; 90:3944–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109*.Chaillon A, Nakazawa M, Rawlings SA, et al. Subclinical CMV and EBV Shedding is associated with increasing HIV DNA Molecular Diversity in Peripheral Blood during Suppressive Antiretroviral Therapy. J Virol 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]; This study observed that in those with HIV on ART with detectable CMV DNA, HIV DNA molecular diversity increased over time, suggesting a contribution of asymptomatic CMV infection to persistence and dynamic changes in the HIV DNA reservoir.
  • 110.Hansen SG, Piatak M Jr., Ventura AB, et al. Immune clearance of highly pathogenic SIV infection. Nature 2013; 502:100–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Hansen SG, Sacha JB, Hughes CM, et al. Cytomegalovirus vectors violate CD8+ T cell epitope recognition paradigms. Science 2013; 340:1237874. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Bowman LJ, Melaragno JI, Brennan DC. Letermovir for the management of cytomegalovirus infection. Expert Opin Investig Drugs 2017; 26:235–41. [DOI] [PubMed] [Google Scholar]
  • 113.Marty FM, Ljungman P, Chemaly RF, et al. Letermovir Prophylaxis for Cytomegalovirus in Hematopoietic-Cell Transplantation. N Engl J Med 2017; 377:2433–44. [DOI] [PubMed] [Google Scholar]
  • 114.La Rosa C, Longmate J, Martinez J, et al. MVA vaccine encoding CMV antigens safely induces durable expansion of CMV-specific T cells in healthy adults. Blood 2017; 129:114–25. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES