Introduction
Prior to the introduction of effective combination antiretroviral therapy (ART), human immunodeficiency virus (HIV) infection was a major cause of death among young adults in the 1980s and early 1990s. The hallmark of HIV infection is CD4+ T cell decline, and if left untreated, the infection typically progresses to acquired immunodeficiency syndrome (AIDS) 8–11 years after seroconversion [1]. The complications of AIDS are well-known and include potentially fatal opportunistic infections (OIs) and certain malignancies, mostly lymphomas, human papilloma virus associated anal or cervical cancer and Kaposi sarcoma. ART has decreased the prevalence of AIDS and lowered mortality rates [2] among people with HIV (PWH). However, even with treatment, the life expectancy of PWH is shorter compared to the general population [3], indicating unmet needs in the understanding and treatment of HIV.
Although HIV/AIDS is widely known as an immunodeficiency state, it is also, if not predominantly, a disease of immune dysregulation. This is demonstrated by the multiple defects in almost every aspect of the immune system and the occasional manifestations of HIV infection as exacerbated or atypical infectious, rheumatologic or autoimmune conditions, seemingly a paradox [4–6]. Evidence of this immune dysfunction persists even after ART-mediated viral suppression. Shortly after ART initiation, it can manifest in the form of immune reconstitution inflammatory syndrome (IRIS), a condition characterized by dysregulated inflammatory responses to co-infection. In contrast, after long-term ART, PWH experience higher rates of liver disease, cardiovascular morbidities, non-AIDS cancers, and other serious non-AIDS events (SNAEs) [7]. This elevated risk for certain chronic diseases is at least partly attributed to the damaging effects of chronic inflammation. Further, some ART-treated PWH demonstrate persistently increased risk for other infections and decreased responses to vaccination. These phenomena indicate that while viral suppression and rise in CD4 counts with ART prevents AIDS-related complications, it cannot fully reverse some of the negative consequences of HIV infection on immunological function.
In this article, we will discuss how chronic inflammation and immune dysfunction intersect and prevent full immune recovery and health among PWH even under ART. We will outline possible causes of chronic inflammation and conditions that reflect immune dysfunction, such as IRIS, increased rates of chronic diseases, and poor vaccine responses. Finally, we will discuss additional risk factors for poor health outcomes in this population and strategies for reducing immune dysfunction in PWH.
Evidence of immune dysfunction and chronic inflammation
Studies of Simian Immunodeficiency Virus (SIV) in non-human primates (NHPs) have been valuable for understanding important aspects of HIV pathogenesis. SIV infection progresses and causes an immunodeficiency disease only in Asian NHPs, which are non-natural hosts for the virus, while maintaining a milder course in the natural African NHP host. However, both natural and non-natural hosts develop high levels of viremia and CD4 depletion after SIV infection, suggesting that these factors alone cannot fully account for disease progression in non-natural hosts. Instead, pathogenic infection is distinguished in part by persistent and generalized activation of both the innate and adaptive arms of the immune system in the non-natural host, compared to a transient inflammatory response during acute infection in natural hosts, implicating chronic inflammation in the disease pathogenesis [8].
A large body of evidence indicates that HIV also causes chronic inflammation in humans. The immune activation also triggers coagulopathy, which further exacerbates inflammation [9]. Biomarkers reflecting inflammation and coagulopathy are elevated in PWH, and some strongly associate with poor outcomes. The levels of D-dimer, IL-6, C-reactive protein, and soluble CD14 (sCD14) specifically are important independent predictors of mortality even after controlling for important clinical factors such as age or HIV-related factors such as CD4 counts [10, 11]. In fact, immune activation, commonly assessed by the expression of CD38, HLA-DR, or Ki-67 among T cells, has been demonstrated to be a better predictor of mortality than viral load in some studies [12, 13]. The persistent activation of the innate immune system, often measured by monocyte activation, may be an even stronger predictor of morbidity and mortality than adaptive immune activation during virologic suppression [14]. Although these biomarkers decline with ART, some remain elevated over HIV-uninfected controls even when ART is started very early in acute HIV infection (AHI) [15]. These studies suggest that immune activation and coagulopathy are triggered early in HIV infection, persist despite virologic suppression, and are important independent drivers of poor health outcomes among PWH.
Further support for the role of chronic immune activation in adverse outcomes is derived from studies of elite controllers (ECs). ECs are PWH who are able to spontaneously control viral loads without ART, and most also maintain normal CD4+ T cell counts. However, chronic immune activation also occurs among ECs, who have decreased percentages of naïve cells [16] and elevated monocyte activation markers [17] and soluble inflammatory mediators. Further, some experience declining CD4+ T cells and can develop AIDS despite consistently low levels of viremia. Compared to those maintaining CD4+ T cell counts, ECs with CD4 decline have increased levels of adaptive immune activation, reflected by the expression of CD38 and HLA-DR among both CD4+ and CD8+ T cells [18]. Markers of innate immune activation, including higher proportions of mature CD14++CD16+ monocytes, are also found in these patients [16]. A subset of ECs remains at elevated risk for SNAEs despite low viral load, implicating persistent immune dysfunction as the driving pathogenic factor.
Causes of chronic inflammation and immune dysfunction
The pathogenesis of chronic inflammation and immune activation in HIV infection appears to be multifactorial. Several viral proteins are highly immunogenic, and acute HIV infection induces inflammatory cytokine production and inflammatory cell death via pyroptosis [19]. Even under ART, which suppresses active viral replication, HIV persists in a small subset of long-lived infected cells termed the viral reservoir. Both cellular reservoirs as well as tissue reservoirs, such as the central nervous system and other anatomical sites with poor ART penetration [20], enable the persistence of latent virus that may continue to trigger the immune system. Activated CD4+ T cells, particularly peripheral PD-1+CXCR3− T follicular helper cells, are enriched for inducible virus, demonstrating an important relationship between immune activation and viral reservoir formation and maintenance [21].
Transient elevations in viremia or “blips” occur among virally suppressed PWH [22], and very low copy numbers can be detected by ultra-sensitive methods, suggesting some degree of persistent replication [23]. Even some of the defective, replication-incompetent proviruses that accumulate under ART are transcriptionally and translationally active, producing viral products that could be the trigger of immune responses [24, 25]. Early initiation of ART results in smaller viral reservoirs as measured by HIV DNA and decreased expression of CD38 and HLA-DR on T cells, supporting a relationship between reservoir size and degree of immune activation [26]. Persistent immune stimulation by the ART-suppressed virus is further supported by the observation that most PWH remain seropositive despite long-term viral suppression. Despite this, in a cohort of early acute PWH who initiated therapy prior to seroconversion and remained seronegative, we did not find evidence of lower inflammatory markers compared to those who became seropositive having evidence of viral transcription [27].
Significant evidence also implicates disturbances to the gut, one of the earliest sites of HIV replication. HIV infection causes the preferential loss of gut CD4+ T cells [28], especially Th17 cell subsets critical for maintaining mucosal integrity. The virus drives downregulation of epithelial cell tight junctions [29] and enterocyte apoptosis, further disrupting barrier function. The result of these effects is the translocation of products of gut bacteria across the bowel wall, evidenced by increased systemic levels of bacterial products, such as lipopolysaccharide (LPS), which contribute to innate and adaptive immune activation [30, 31]. In NHP models, systemic LPS levels do not increase after infection in natural hosts, supporting the role for microbial translocation in pathological infection [30]. These changes to the gut are accompanied by microbiome disturbances, characterized by an overall loss of diversity and a shift to a more pro-inflammatory distribution of gut bacteria [32, 33]. This includes enrichments in Enterobacteriaceae and Prevotella, both of which have been linked to immune activation [31, 34, 35]. These shifts in species and low gene counts indicative of decreased diversity have been correlated with lower nadir CD4 counts [36]. In addition to this relationship with immunodeficiency, the extent of dysbiosis has further been associated with the degree of immune activation, and an increased number of comorbidities [37]. Importantly, more recently, Prevotella has been linked to sexual practices (receptive anal intercourse) regardless of HIV status [37].
Fibrosis of lymphoid tissues also occurs with HIV infection and contributes to CD4 decline and poor CD4 recovery after ART, setting the stage for immune dysfunction. Collagen deposition in the lymph nodes of PWH is believed to be triggered by TGF-β expression induced by infection. Fibrosis is also observed among ECs and those treated with ART [38], suggesting it can even be induced by very low levels of viral replication. This fibrosis causes disruption of the lymph node tissue architecture, which is hypothesized to impede T cell migration and limit cells’ access to growth factors like IL-7 made by fibroblast reticular cells [39]. These effects may be particularly detrimental to naïve populations, which indeed are preferentially lost.
High rates of co-infection with other pathogens among PWH are another important factor. The most important of these may be Cytomegalovirus (CMV), which has high rates of seropositivity and shedding among PWH. Even in HIV-uninfected individuals, CMV infection can drive massive expansion of CMV-specific CD8+ T cells [40], the result of which is restriction of the T cell receptor (TCR) repertoire and inversion of the CD4/CD8 ratio. These effects likely compound immune dysfunction in PWH. CMV shedding has been linked to both CD4+ and CD8+ T cell activation in ART-treated PWH [41, 42] and delayed reservoir decay under ART. Co-infection with hepatitis viruses is also highly prevalent among PWH, and increased immune activation, measured by CD38 expression on both CD4 and CD8 T cells, has been observed among PWH co-infected with Hepatitis C virus [43].
Defective immune reconstitution under ART
After ART initiation, CD4 counts typically continue to rise over the course of years, and the majority of ART-treated PWH achieve suppression of plasma viremia and reconstitution of CD4 T cells. However, some fail to achieve full immunologic recovery, reflected by persistently low CD4 T cell counts despite long-term ART. Roughly 60% of PWH fail to achieve normal CD4 counts of >500 cells/μL after 4 years of ART [44, 45]. Approximately 6% never even achieve CD4 counts >200 cells/μL despite continuous ART for 4 years, and this increases to 25.9% with discontinuous ART use [44]. Incomplete CD4 recovery predicts increased risk of mortality [46], and rates of both AIDS-related events and SNAEs are increased [47, 48]; these include non-AIDS malignancies [46, 49], liver disease [50], and respiratory disease [51].
In addition to discontinuous ART use, initiation of ART during late stages of HIV infection also increases the likelihood of immunological non-response. Most patients starting ART with baseline CD4 counts of >300 or >350 cells/μL achieve reconstitution [45, 52], but persistently low CD4 counts are seen among 25% of those with baseline CD4 <100 cells/μL, even after 7–10 years of ART [45]. Poor responses also correlate with increased levels of innate and adaptive immune activation [53, 54]. CD4 recovery further depends upon thymic size and function [55], and higher baseline collagen deposition predicts poor CD4 reconstitution after ART [56, 57]. Even when peripheral CD4 counts normalize, tissue CD4 populations may not fully recover. Virally suppressed PWH with normal or near-normal blood CD4 counts may exhibit persistently decreased CD4+ T cell numbers in gut-associated lymphoid tissue [58].
Other aspects of CD4+ T cell reconstitution beyond quantitative reconstitution must also be considered. The balance of naïve, effector, and memory T cell populations is also skewed by HIV infection and may not be fully restored by ART. Limited rescue of the naïve CD4+ T cell population occurs with ART among patients who already experienced severe depletion of these populations [59]. Constriction of the CD4+ TCR repertoire is also reflected by depletion of certain TCRBV subfamilies [59]. Finally, CD4+ T cells retain qualitative defects that hinder full functional immune recovery. Antigen-specific responses are impaired, seen in decreased IFN-γ production from Mycobacterium tuberculosis (TB)-specific CD4+ T cells [60] and increased CTLA-4 and PD-1 on HIV-infected cells contributing to hypo-responsiveness [61].
Due to concurrent changes among CD8+ T cells, including expansions of terminally differentiated cells [62, 63] and similar depletions of naïve populations, some ART-treated PWH fail to normalize CD4/CD8 ratios even with CD4 reconstitution. Inverted CD4/CD8 ratios and the accumulation of anergic CD28− CD8+ T cells are observed in both aging and HIV infection [64] and are part of an “immune risk” phenotype that predicts death even in the general population [65]. Among PWH, low CD4/CD8 ratios confer an increased risk of SNAEs and non-AIDS mortality, independently of CD4 count [66]. Patients with CD8 counts >2000 cells/μL after 1 year of ART have an increased risk of death compared to those with CD8 counts between 500–1500 cells/μL [67], and increasing CD8 counts may be associated with adverse health outcomes such as myocardial infarction [68]. Functional defects among CD8+ T cells, such as decreased proliferative and cytotoxic activity among HIV-specific CD8+ T cells, also persist despite viral suppression [69, 70].
Persistent disturbances in innate immunity are also apparent, with many studies addressing monocytes. Monocyte activation and inflammasome induction have been implicated in persistent CD4 decline despite ART [71] as well as poor CD4 reconstitution after ART [71, 72]. A subset of monocytes in PWH strongly upregulates expression of tissue factor (TF) and cytokines in response to LPS, even under ART, driving activation of the clotting cascade [73]. Other innate cells are also altered. The percentages of activated HLA-DR+CD38+ NK cells remain elevated despite viral suppression [74], and mature NK cells still exhibit low IFN-γ production after rescue to normal levels with ART [75]. Similarly, neutrophils remain highly activated under ART as evidenced by increased CD11b expression and generation of reactive oxygen species [76], but retain functional defects such as decreased chemotaxis and low fungicidal activity [77]. HIV infection also causes low numbers of dendritic cells (DCs) and decreased cytokine signaling from these cells, and proportions of plasmacytoid DCs may not be restored even with ART [78, 79]. The involvement of innate immune dysregulation is reflected by increased levels of innate immune activation markers in serum, including neopterin, CXCL10, sCD14, and sCD163, which are not restored to levels seen in age-matched uninfected controls by ART [80, 81].
More recent research also suggests that innate lymphoid cells (ILCs), particularly IL-17 and IL-22 producing colonic ILC3s, are depleted early in HIV infection, not fully restored by ART if initiated after cellular loss, and may contribute to persistent gut barrier dysfunction [82, 83]. Mucosal associated invariant T cells (MAIT cells), particularly those expressing IL-17, are also lost early in HIV infection and do not recover fully in number or in function under ART [84, 85]. HIV infection is further marked by alterations in γδ T cells, with decreased expression of IFN-γ and TNF-α that remains low after ART [86]. These changes may further contribute to impaired mucosal immunity as well as increased susceptibility to other infections, such as TB.
Finally, HIV infection is characterized by persistent defects in humoral immunity, including B cell activation, hypergammaglobulinemia, and changes in B cell phenotypes such as decreased memory populations and increased percentages of exhausted, immature transitional, and other atypical subsets [87]. Certain defects, including decreased B cell proliferation in response to CD4+ T cell help, are ameliorated with viral suppression under ART [88]. Other defects are not corrected despite antiretroviral therapy. Persistently low levels of memory B cell populations and loss of protective antibodies against vaccine antigens contribute to increased risk for certain diseases such as pneumococcal infection even after treatment with ART [89–91].
Inflammatory complications during immune reconstitution
It is clear that the negative consequences of HIV infection on the immune system are not fully reversed by ART. In fact, the initiation of ART itself is sometimes the catalyst that unveils this immune dysfunction. Shortly after starting ART, approximately 20% of patients develop Immune Reconstitution Inflammatory Syndrome (IRIS), a syndrome of clinical worsening following viral suppression that is attributed to the activation of dysregulated responses against underlying co-infections. Most cases occur within the first 2 months of treatment mostly in patients starting therapy with lower CD4 counts [92] and are subcategorized as “unmasking” IRIS, involving a formerly undetected pathogen, or “paradoxical” IRIS, involving a co-infection that had been seemingly adequately treated. The most common pathogens involved are Mycobacterial species, Cryptococcus, and herpesviruses including CMV. Given the role of immune hyperactivation in IRIS, glucocorticoid therapy is the mainstay of treatment.
Risk factors for the development of IRIS help to illustrate a relationship between immunosuppression, immune activation and immune dysregulation. Lower CD4 nadirs, lower CD4 counts at the time of ART initiation, and lower CD4/CD8 ratios reflective of more severe immunosuppression are all associated with increased risk of IRIS [93–95]. Excess immune activation is also apparent during the time of the IRIS event. One study of IRIS stemming from various pathogens found overall increased levels of IFN-γ during the timing of the IRIS event compared to non-IRIS controls [96]. Levels of IL-6 are also elevated during IRIS arising from TB infection (TB-IRIS) and Cryptococcal meningitis (CM-IRIS) [97, 98]. Patients with TB-IRIS have increased serum TNF and IFN-γ [99] and cells producing cytokine in response to antigen stimulation [100], illustrating involvement of adaptive responses. TB-IRIS patients have higher levels of sCD14, soluble CD163 (sCD163), and soluble tissue factor (sTF) compared to non-IRIS controls at the time of IRIS, correlating with expansion of CD14++CD16− monocytes [97], indicating simultaneous involvement of innate cells. Inflammasome activation has also been implicated in the development of TB-IRIS [101] further suggesting a major role for innate immune dysregulation in IRIS.
Other important risk factors for IRIS include shorter duration of OI treatment and shorter interval between OI treatment and ART initiation [92, 97]. Longer treatment of TB infection has been shown to decrease markers of immune activation and the incidence of IRIS [97]. In CM-IRIS, a higher antigen burden, lower levels of protective Th1 type cytokines, and accompanying increases in less protective Th17 and Th2 type cytokines are associated with increased risk of IRIS [98, 102]. These studies suggest that suboptimal antigen clearance is involved in the pathogenesis. In the context of severe lymphopenia and residual antigen, homeostatic proliferation driven by IL-7, which increases with CD4 lymphopenia and during IRIS events [96], may promote the over-representation of highly activated pathogen-specific CD4+ T cells. Even 6 months after the IRIS event, IRIS patients have increased percentages of effector memory CD4+ T cells and central memory CD8+ T cells [96].
While IRIS due to infectious causes appears relatively soon after ART initiation, autoimmune disease is another complication that can occur during the later stages of immune recovery. Autoimmune thyroid conditions may appear roughly 1.5–2 years after ART initiation and are more common with a history of more severe immunosuppression indicated by low CD4 count [103]. The recurrence of sarcoidosis among PWH with a remote history of this condition has also been reported [104]. Other autoimmune phenomena that have been documented after ART include autoimmune hepatitis [105], Sjogren’s syndrome [106], and alopecia [107]. Hypothesized causes of this autoimmunity include residual thymic dysfunction, re-biasing from a Th2 to a Th1 type response, and expansion of self-reactive clones during reconstitution of the CD4+ T cell receptor repertoire.
Major HIV morbidities: cardiovascular disease and others
More chronically, persistent immune dysfunction can manifest as increased rates of non-infectious morbidities. Cardiovascular disease is the leading cause of mortality worldwide, and the prevalence is increasing among PWH despite decreases in the general population [108]. The risk of acute myocardial infarction among PWH is 1.5-fold higher [109], and HIV infection is an independent risk factor for ischemic stroke [110, 111]. Chronic inflammation and immune dysfunction clearly play a role in the pathogenesis of these conditions. Levels of sCD163, sCD14, and CCL2 reflect monocyte/macrophage activation and are correlated with coronary artery stenosis in PWH and the general population [112]. Increased proportions of non-classical CD14+CD16++ and intermediate CD14++CD16++ monocytes, as well as upregulated TF expression on monocytes, are seen in both HIV infection and in patients with acute coronary syndrome who are HIV seronegative [113]. Expression of TF by monocytes correlates with plasma viremia and plasma sCD14 and can be induced in vitro by LPS, suggesting microbial translocation probably contributes to thrombotic disease through activation of monocytes [113]. CMV infection, which is common among PWH, is also linked to atherosclerosis [114].
Some ART medications themselves may be linked to cardiovascular morbidity. Protease inhibitors in particular are linked to hyperlipidemia and hypercholesterolemia [115], the development of atherosclerosis via increased fibrinogen [116], and increased risk of myocardial infarction in some studies [117]. Nucleoside reverse-transcriptase inhibitors (NRTIs) and non-nucleoside reverse-transcriptase inhibitors (NNRTIs) have also been associated in lipid and triglyceride alterations [118]. Certain ART regimes, particularly dolutegravir combined with tenofovir alafenamide fumarate, augment weight gain and obesity [119], contributing to metabolic disturbances that may increase the risk of cardiovascular morbidity. Other known risk factors for cardiovascular and thrombotic diseases, including smoking and comorbidities such as hypertension, are also prevalent among PWH and further contribute to the increased risk.
Liver disease is another important morbidity among PWH, accounting for about 14.5% of deaths [50]. In this population, liver disease is strongly associated with immunodeficiency and co-infection with Hepatitis B and C viruses [50]. Increasing CD4 counts are protective, suggesting beneficial effects of ART. However, unexpectedly, the risk of liver-related mortality has been shown to increase with longer exposure to ART – the exact reasons for this are unclear, and may involve antiretroviral toxicity, particularly with reverse transcriptase inhibitors, and the natural progression of hepatitis over time [50, 120]. Non-alcoholic steatohepatitis (NASH) and non-alcoholic fatty liver disease (NAFLD) also occur at high rates among PWH and are driven by multiple factors in this population, including an association with male gender, NNRTI use, and increased body mass index among others [121, 122].
Renal dysfunction is also observed in PWH, reflected in elevated cystatin C levels compared to uninfected controls [123], and has also been associated with tenofovir usage [124]. Renal and liver diseases combined with poor nutritional status may contribute to Vitamin D deficiency, which is common among PWH and is linked to various other pathologies, including IRIS, osteoporosis, atherosclerosis[125] and accelerated liver disease [126].
Non-AIDS defining cancers (NADCs) have become another leading cause of death among PWH. PWH are at an elevated risk of anal cancers, certain lymphomas, oropharyngeal cancers, and lung cancer, among others [127]. High rates of smoking are observed among PWH, and this represents the greatest risk factor for the development of lung cancer in this population [128]. However, HIV infection remains an independent risk factor for lung cancer [128, 129], and the risk appears to be inversely related to CD4 count. Local tissue damage from dysregulated CD8 responses, increased rates of pneumonia and chronic lung infections, and even direct viral oncogenesis have been proposed as contributing factors with varying degrees of supporting data [130]. Risk of NADCs such as liver cancer and anal cancer are augmented by co-infection with other pathogens, including hepatitis viruses and human papilloma viruses, respectively.
HIV infection also affects the central nervous system, causing neuronal apoptosis and synaptodendritic degeneration, leading to neuropsychiatric impairment in approximately half of PWH [131]. This impairment is common even among PWH treated with ART. While the prevalence of HIV-associated dementia has significantly decreased in the ART era, a similar percentage of PWH meets criteria for milder HIV-associated neurocognitive disorders now compared to in the pre-ART era [131, 132]. Impairment is more common with lower CD4 nadirs [132]. Although cognitive function improves over time with treatment, PWH with advanced immunosuppression still exhibit twice the rate of cognitive impairment compared to the HIV-uninfected population, even after CD4 >200 on ART [133]. These studies suggest that neuropsychiatric impairment is likely established during the peak of immunosuppression and persists despite immune reconstitution.
HIV infection also decreases bone mineral density through direct effects on osteoclasts and osteoblasts as well as inflammation-related impact, resulting in a high prevalence of bone diseases among PWH. Osteoporosis is approximately three times more common among PWH compared to the HIV uninfected population [134]. ART does not seem to ameliorate the effects of HIV on bone density, and some medications such as protease inhibitors may even accelerate the problem; osteoporosis appears to be more common among ART-treated PWH compared to ART-naïve [134], although this finding remains controversial.
Other disorders that are more common among PWH that are likely not fully reversed by ART include pulmonary arterial hypertension [135], which may arise from dysregulated fibroblast activity and growth factor expression during HIV infection [136], and frailty, a phenotype associated with weight loss, exhaustion, and weakness that is typically associated with advanced age [137].
Infections & vaccine responses
OIs decrease significantly with ART, but some PWH remain at risk, particularly those with incomplete CD4 recovery. Compared to the general population, PWH also remain at higher risk of a number of bacterial diseases even with treatment. They are more likely to develop invasive pneumococcal disease [138], a phenomenon attributed to the loss of memory B cells and serologic memory [91]. Globally, TB co-infection also remains a major obstacle. PWH are at higher risk of acquiring TB infection, which has been shown to accelerate the course of HIV [139], and this risk likely remains elevated over the HIV-uninfected population even after treatment with ART [140].
PWH also have high rates of co-infection with other chronic viruses. Co-infection with Hepatitis B and C viruses is common and confers a higher risk of SNAEs [48], in particular liver disease [141]. Infection with certain human papilloma viruses can cause anal, cervical and oropharyngeal cancers, which occur at higher rates among PWH. PWH are also at an increased risk of developing herpes zoster and complications of zoster, even at relatively young ages, a phenomenon associated with poor immune function [142]. As previously discussed, seropositivity rates for human herpesviruses such as CMV and the related Epstein-Barr Virus are also high and may contribute to immune activation and dysfunction.
PWH may also remain at an elevated risk of vaccine-preventable illnesses such as measles, tetanus, and influenza. ART fails to recover protective memory B cell populations, and PWH have poorer responses to immunizations and booster immunizations against certain pathogens [143–146]. Responses to influenza vaccine are perhaps the best studied, and it is now well-documented that low CD4 count is associated with poor responses to influenza vaccination. Low CD4 count is associated with lower peak antibody titers as well as decreased formation of memory B cell responses [147]. ART improves responses to influenza vaccination largely by increasing numbers of CD4 T cells [148]. However, even PWH with CD4 >500 are still less likely to mount antibody responses than HIV-uninfected counterparts [143], and antibody titers remain lower among PWH despite ART. Similar immunological deficiencies are present in both the elderly and among PWH, such as decreased memory B cell and peripheral T follicular helper cell expansion after vaccination [144]. Accordingly, as in the elderly, PWH have improved responses with increased vaccine doses and the addition of adjuvants as outlined below.
Defects in responses to various other important vaccinations are also observed. PWH have decreased seroconversion rates in response to primary Hepatitis B vaccination, and the duration of the response is shorter. Protective antibody levels were only present in roughly 50–66% of vaccinated PWH after 1 year [149, 150], and less than a third of initial responders after two2 years [150]. Adjuvants, higher doses, and additional immunizations can increase seroconversion rates [145, 149, 151]. Loss of protection is also seen earlier among PWH for measles vaccination, a greater proportion of whom have no evidence of measles antibodies 1 one year post-vaccination compared to HIV-uninfected controls [146]. Pediatric studies have shown that while DTaP vaccination induces protective antibodies in ART-treated children, titers may be lower and duration is shorter compared to those observed in HIV-uninfected children [152, 153]. In summary, while PWH may be able to respond to vaccination, particularly after ART treatment, the duration of this response may be limited in both magnitude and duration due to persistent dysfunction among immune cell subsets.
Demographics and lifestyle factors
Persistent immune dysfunction plays a prominent role in the morbidities and poor health outcomes that persist after ART, but lifestyle risk factors are also involved. Although continuous ART has been shown to improve outcomes, medication adherence is unfortunately suboptimal in most cases [154], indicating a critical area for improvement. High rates of tobacco use, injection drug use (IDU), and alcohol use are present among PWH and contribute to various SNAEs including lung cancer, liver disease, and more. These activities have also been associated with poorer adherence to ART and control of HIV [139, 155]. IDU further increases risk of acquiring other chronic infections such as hepatitis B or C viruses. Obesity is also present at high rates among PWH and has been independently linked to monocyte and innate immune activation in PWH [156].
Sex differences may also play a role in HIV-associated outcomes, potentially due to differences in immunological function, drug metabolism, and sociological factors related to sex. It has been frequently observed that women with HIV have lower viral loads [157], but studies on outcomes after ART are conflicting. Women with HIV may have a decreased risk of certain complications compared to men with HIV, such as dementia and Kaposi sarcoma [158], while experiencing an increased risk of others, such as myocardial infarction [109]. Some data suggest that sex plays a role in risk for disease progression and virologic control after ART, but many of these studies are complicated by differences in medication adherence, drug use, educational and socioeconomic factors.
Finally, social determinants of health also contribute to high rates of morbidity in this population. For example, despite elevated risk of colorectal cancer, PWH are less likely to be up-to-date on screening [159], revealing a need for more rigorous use of preventative health measures in this population. Insurance coverage and stable housing are two factors shown to increase ART usage and decrease mortality rates among PWH [160, 161], and programs to help patients access these resources could have tremendous positive impacts on health outcomes.
Combatting residual immune dysfunction
ART remains the single intervention with the strongest data for reducing morbidity and mortality among PWH. Early initiation of ART can promote robust immune restoration, minimize the size of the viral reservoir [162], decrease the degree of residual inflammation [15], limit mucosal damage, and reduce the extent of lymphoid tissue fibrosis [57]. Therefore, early diagnosis of HIV infection and early initiation of ART, at higher CD4 counts and before advanced disease or severe immunodeficiency, is critically important for limiting immune activation and persistent immune dysfunction.
Because the ART-suppressed virus may still be recognized by the immune system [24, 25], further reductions to the viral reservoir could theoretically minimize chronic immune activation and improve outcomes. Some teams have investigated the use of intensive ART regimens involving an increased number of drugs, but this “mega-ART” approach has not been shown to improve outcomes or accelerate reservoir decay compared to standard ART [163, 164]. The development of effective therapeutic vaccines could also potentially contribute to improved viral control, but trials of candidates in humans have only yielded mixed results [165]. Partial inhibition of glycolysis has been found to dampen viral replication and accelerate the decay of infected cells in vivo, suggesting that metabolic pathways could be a promising target to explore for the development of future novel therapies [166].
Other strategies to improve health outcomes have focused on augmenting immune recovery via cytokine therapies, which have had mixed results. Treatment with IL-2 can increase the numbers of CD4+ T cells, but two large randomized controlled trials, Subcutaneous Recombinant, Human Interleukin-2 in HIV-Infected Patients with Low CD4+ Counts under Active ART (SILCAAT) and Evaluation of Subcutaneous Proleukin in a Randomized International Trial (ESPRIT), failed to show any clinical benefit [167]. Treatment with recombinant human IL-7 can also increase numbers of both CD4+ and CD8+ T cells [168], and in particular naïve and central memory CD4+ T cells [169], but it remains unclear if this confers any clinical advantage. These cytokines have further been used as part of “shock and kill” strategies to reactivate the latent viral reservoir to increase susceptibility to therapies targeting active replication [170, 171].
Other approaches have attempted to dampen immune activation with medications directly acting against the inflammation or against its causes. The immunosuppressive agent hydroxychloroquine decreases percentages of activated T cells and monocytes and increases proportions of naïve cells among PWH [172], and the immunomodulatory drug leflunomide was similarly found to decrease markers of T cell activation in untreated PWH in a randomized trial [173]. HIV-associated gut mucosal disruption has been targeted with probiotics, which have shown some effects on reversing immune dysfunction [174]; lactoferrin, which had no effect [175]; and with sevelamer, a drug previously observed to decrease microbial translocation in hemodialysis patients. Sevelamer did not decrease various measures of microbial translocation or immune activation in PWH, but lowered levels of sTF and cholesterol, suggesting potential cardiovascular benefits [176]. Antifibrotic drugs have been investigated for their potential to counteract lymphoid collagen deposition. Pirfenidone has been shown to decrease collagen synthesis from fibroblasts in vivo [177] and improve CD4+ T cell reconstitution in SIV models [178], but lisinopril, also tested for its anti-fibrotic properties, had no effect on reversing fibrosis, dampening inflammation, or decreasing HIV reservoirs [179]. Losartan also failed to decrease inflammation or improve CD4 counts [180]. Given the role of co-infections in chronic immune activation, a study investigated the effect of treating subclinical CMV infection with valganciclovir and found that it decreased the rates of CMV DNA positivity and the levels of CD8 T cell activation [181].
Finally, because of the prominence of HIV-associated coagulopathy and its relationship with inflammation and adverse outcomes, many trials have studied the use of anticoagulants in PWH. The Targeted Anticoagulation Therapy To Reduce Inflammation and Cellular Activation in Long-Term HIV Disease (TACTICAL-HIV) trial found that giving a low dose of the direct factor Xa inhibitor edoxaban for four months significantly lowered D-dimer levels but had no effect on markers of inflammation [182]. A shorter course of the thrombin receptor antagonist vorapaxar did not significantly decrease markers of immune activation or coagulation in the Attenuation of D-dimer Using Vorapaxar to Target Inflammatory and Coagulation Endpoints (ADVICE) study [183]. Treatment of PWH with statins and aspirin have been shown to decrease markers of T cell and innate immune activation [184–186]. The Randomized Trial to Prevent Vascular Events in HIV (REPRIEVE), a large prospective study comparing pivastatin to control for prevention of major adverse cardiac events in PWH, is currently underway [187].
Although the development of ART is still one of the triumphs of modern medicine, the evidence of persistent immune dysfunction even among early treated PWH indicates that challenges in the understanding and treatment of HIV infection remain. Decades of studies have increased our knowledge of the multiple factors that drive and sustain the chronic immune activation and inflammation in ART-treated PWH, which is closely linked to the higher burden of certain non-infectious morbidities in this population. As we continue to learn more about the pathogenesis of persistent immune dysfunction under ART, health outcomes in PWH could be improved now by early diagnosis and treatment, cancer screenings and other age-appropriate monitoring, lifestyle interventions to decrease high risk behaviors, increased availability of social resources, and control of coinfections such as HCV and comorbidities.
Funding:
This work was supported by the Intramural Research program of NIAID/NIH.
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- 1.Time from HIV-1 seroconversion to AIDS and death before widespread use of highly-active antiretroviral therapy: a collaborative re-analysis. Collaborative Group on AIDS Incubation and HIV Survival including the CASCADE EU Concerted Action. Concerted Action on SeroConversion to AIDS and Death in Europe. Lancet, 2000. 355(9210): p.1131–7. [PubMed] [Google Scholar]
- 2.Palella FJ Jr., et al. , Declining morbidity and mortality among patients with advanced human immunodeficiency virus infection. HIV Outpatient Study Investigators. N Engl J Med, 1998. 338(13): p. 853–60. [DOI] [PubMed] [Google Scholar]
- 3.Antiretroviral Therapy Cohort C, Life expectancy of individuals on combination antiretroviral therapy in high-income countries: a collaborative analysis of 14 cohort studies. Lancet, 2008. 372(9635): p. 293–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Zandman-Goddard G and Shoenfeld Y, HIV and autoimmunity. Autoimmun Rev, 2002. 1(6): p. 329–37. [DOI] [PubMed] [Google Scholar]
- 5.Cuellar ML and Espinoza LR, Rheumatic manifestations of HIV-AIDS. Baillieres Best Pract Res Clin Rheumatol, 2000. 14(3): p. 579–93. [DOI] [PubMed] [Google Scholar]
- 6.Kopelman RG and Zolla-Pazner S, Association of human immunodeficiency virus infection and autoimmune phenomena. Am J Med, 1988. 84(1): p. 82–8. [DOI] [PubMed] [Google Scholar]
- 7.Deeks SG and Phillips AN, HIV infection, antiretroviral treatment, ageing, and non-AIDS related morbidity. BMJ, 2009. 338: p. a3172. [DOI] [PubMed] [Google Scholar]
- 8.Klatt NR, Silvestri G, and Hirsch V, Nonpathogenic simian immunodeficiency virus infections. Cold Spring Harb Perspect Med, 2012. 2(1): p. a007153. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Foley JH and Conway EM, Cross Talk Pathways Between Coagulation and Inflammation. Circ Res, 2016. 118(9): p. 1392–408. [DOI] [PubMed] [Google Scholar]
- 10.Kuller LH, et al. , Inflammatory and coagulation biomarkers and mortality in patients with HIV infection. PLoS Med, 2008. 5(10): p. e203. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Sandler NG, et al. , Plasma levels of soluble CD14 independently predict mortality in HIV infection. J Infect Dis, 2011. 203(6): p. 780–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Giorgi JV, et al. , Shorter survival in advanced human immunodeficiency virus type 1 infection is more closely associated with T lymphocyte activation than with plasma virus burden or virus chemokine coreceptor usage. J Infect Dis, 1999. 179(4): p. 859–70. [DOI] [PubMed] [Google Scholar]
- 13.Hazenberg MD, et al. , Persistent immune activation in HIV-1 infection is associated with progression to AIDS. AIDS, 2003. 17(13): p. 1881–8. [DOI] [PubMed] [Google Scholar]
- 14.Wilson EM, et al. , Monocyte-activation phenotypes are associated with biomarkers of inflammation and coagulation in chronic HIV infection. J Infect Dis, 2014. 210(9): p. 1396–406. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Sereti I, et al. , Persistent, Albeit Reduced, Chronic Inflammation in Persons Starting Antiretroviral Therapy in Acute HIV Infection. Clin Infect Dis, 2017. 64(2): p. 124–131. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Krishnan S, et al. , Evidence for innate immune system activation in HIV type 1-infected elite controllers. J Infect Dis, 2014. 209(6): p. 931–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Pereyra F, et al. , Increased coronary atherosclerosis and immune activation in HIV-1 elite controllers. AIDS, 2012. 26(18): p. 2409–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Bansal A, et al. , Normal T-cell activation in elite controllers with preserved CD4+ T-cell counts. AIDS, 2015. 29(17): p. 2245–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Doitsh G, et al. , Cell death by pyroptosis drives CD4 T-cell depletion in HIV-1 infection. Nature, 2014. 505(7484): p. 509–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Wong JK and Yukl SA, Tissue reservoirs of HIV. Curr Opin HIV AIDS, 2016. 11(4): p. 362–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Pallikkuth S, et al. , Peripheral T Follicular Helper Cells Are the Major HIV Reservoir within Central Memory CD4 T Cells in Peripheral Blood from Chronically HIV-Infected Individuals on Combination Antiretroviral Therapy. J Virol, 2015. 90(6): p. 2718–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Nettles RE, et al. , Intermittent HIV-1 viremia (Blips) and drug resistance in patients receiving HAART. JAMA, 2005. 293(7): p. 817–29. [DOI] [PubMed] [Google Scholar]
- 23.Dornadula G, et al. , Residual HIV-1 RNA in blood plasma of patients taking suppressive highly active antiretroviral therapy. JAMA, 1999. 282(17): p. 1627–32. [DOI] [PubMed] [Google Scholar]
- 24.Imamichi H, et al. , Defective HIV-1 proviruses produce novel protein-coding RNA species in HIV-infected patients on combination antiretroviral therapy. Proc Natl Acad Sci U S A, 2016. 113(31): p. 8783–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Imamichi H, et al. , Defective HIV-1 proviruses produce viral proteins. Proc Natl Acad Sci U S A, 2020. 117(7): p. 3704–3710. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Jain V, et al. , Antiretroviral therapy initiated within 6 months of HIV infection is associated with lower T-cell activation and smaller HIV reservoir size. J Infect Dis, 2013. 208(8): p. 1202–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Cai CW, et al. , Inflammatory Biomarkers Do Not Differ Between Persistently Seronegative vs Seropositive People With HIV After Treatment in Early Acute HIV Infection. Open Forum Infect Dis, 2020. 7(9): p. ofaa383. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Brenchley JM, et al. , CD4+ T cell depletion during all stages of HIV disease occurs predominantly in the gastrointestinal tract. J Exp Med, 2004. 200(6): p. 749–59. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Nazli A, et al. , Exposure to HIV-1 directly impairs mucosal epithelial barrier integrity allowing microbial translocation. PLoS Pathog, 2010. 6(4): p. e1000852. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Brenchley JM, et al. , Microbial translocation is a cause of systemic immune activation in chronic HIV infection. Nat Med, 2006. 12(12): p. 1365–71. [DOI] [PubMed] [Google Scholar]
- 31.Jiang W, et al. , Plasma levels of bacterial DNA correlate with immune activation and the magnitude of immune restoration in persons with antiretroviral-treated HIV infection. J Infect Dis, 2009. 199(8): p. 1177–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Mutlu EA, et al. , A compositional look at the human gastrointestinal microbiome and immune activation parameters in HIV infected subjects. PLoS Pathog, 2014. 10(2): p. e1003829. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Gootenberg DB, et al. , HIV-associated changes in the enteric microbial community: potential role in loss of homeostasis and development of systemic inflammation. Curr Opin Infect Dis, 2017. 30(1): p. 31–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Dillon SM, et al. , An altered intestinal mucosal microbiome in HIV-1 infection is associated with mucosal and systemic immune activation and endotoxemia. Mucosal Immunol, 2014. 7(4): p. 983–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Dinh DM, et al. , Intestinal microbiota, microbial translocation, and systemic inflammation in chronic HIV infection. J Infect Dis, 2015. 211(1): p. 19–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Guillen Y, et al. , Low nadir CD4+ T-cell counts predict gut dysbiosis in HIV-1 infection. Mucosal Immunol, 2019. 12(1): p. 232–246. [DOI] [PubMed] [Google Scholar]
- 37.Vujkovic-Cvijin I, et al. , HIV-associated gut dysbiosis is independent of sexual practice and correlates with noncommunicable diseases. Nat Commun, 2020. 11(1): p. 2448. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Sanchez JL, et al. , Lymphoid fibrosis occurs in long-term nonprogressors and persists with antiretroviral therapy but may be reversible with curative interventions. J Infect Dis, 2015. 211(7): p. 1068–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Estes JD, Haase AT, and Schacker TW, The role of collagen deposition in depleting CD4+ T cells and limiting reconstitution in HIV-1 and SIV infections through damage to the secondary lymphoid organ niche. Semin Immunol, 2008. 20(3): p. 181–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Koch S, et al. , Cytomegalovirus infection: a driving force in human T cell immunosenescence. Ann N Y Acad Sci, 2007. 1114: p. 23–35. [DOI] [PubMed] [Google Scholar]
- 41.Wittkop L, et al. , Effect of cytomegalovirus-induced immune response, self antigen-induced immune response, and microbial translocation on chronic immune activation in successfully treated HIV type 1-infected patients: the ANRS CO3 Aquitaine Cohort. J Infect Dis, 2013. 207(4): p. 622–7. [DOI] [PubMed] [Google Scholar]
- 42.Christensen-Quick A, et al. , Subclinical Cytomegalovirus DNA Is Associated with CD4 T Cell Activation and Impaired CD8 T Cell CD107a Expression in People Living with HIV despite Early Antiretroviral Therapy. J Virol, 2019. 93(13). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Gonzalez VD, et al. , High levels of chronic immune activation in the T-cell compartments of patients coinfected with hepatitis C virus and human immunodeficiency virus type 1 and on highly active antiretroviral therapy are reverted by alpha interferon and ribavirin treatment. J Virol, 2009. 83(21): p. 11407–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Kaufmann GR, et al. , CD4 T-lymphocyte recovery in individuals with advanced HIV-1 infection receiving potent antiretroviral therapy for 4 years: the Swiss HIV Cohort Study. Arch Intern Med, 2003. 163(18): p. 2187–95. [DOI] [PubMed] [Google Scholar]
- 45.Kelley CF, et al. , Incomplete peripheral CD4+ cell count restoration in HIV-infected patients receiving long-term antiretroviral treatment. Clin Infect Dis, 2009. 48(6): p. 787–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Pacheco YM, et al. , Increased risk of non-AIDS-related events in HIV subjects with persistent low CD4 counts despite cART in the CoRIS cohort. Antiviral Res, 2015. 117: p. 69–74. [DOI] [PubMed] [Google Scholar]
- 47.Strategies for Management of Antiretroviral Therapy Study, G., et al. , CD4+ count-guided interruption of antiretroviral treatment. N Engl J Med, 2006. 355(22): p. 2283–96. [DOI] [PubMed] [Google Scholar]
- 48.Baker JV, et al. , CD4+ count and risk of non-AIDS diseases following initial treatment for HIV infection. AIDS, 2008. 22(7): p. 841–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Monforte A, et al. , HIV-induced immunodeficiency and mortality from AIDS-defining and non-AIDS-defining malignancies. AIDS, 2008. 22(16): p. 2143–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Weber R, et al. , Liver-related deaths in persons infected with the human immunodeficiency virus: the D:A:D study. Arch Intern Med, 2006. 166(15): p. 1632–41. [DOI] [PubMed] [Google Scholar]
- 51.Marin B, et al. , Non-AIDS-defining deaths and immunodeficiency in the era of combination antiretroviral therapy. AIDS, 2009. 23(13): p. 1743–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Gras L, et al. , CD4 cell counts of 800 cells/mm3 or greater after 7 years of highly active antiretroviral therapy are feasible in most patients starting with 350 cells/mm3 or greater. J Acquir Immune Defic Syndr, 2007. 45(2): p. 183–92. [DOI] [PubMed] [Google Scholar]
- 53.Hunt PW, et al. , T cell activation is associated with lower CD4+ T cell gains in human immunodeficiency virus-infected patients with sustained viral suppression during antiretroviral therapy. J Infect Dis, 2003. 187(10): p. 1534–43. [DOI] [PubMed] [Google Scholar]
- 54.Marchetti G, et al. , Microbial translocation is associated with sustained failure in CD4+ T-cell reconstitution in HIV-infected patients on long-term highly active antiretroviral therapy. AIDS, 2008. 22(15): p. 2035–8. [DOI] [PubMed] [Google Scholar]
- 55.Smith KY, et al. , Thymic size and lymphocyte restoration in patients with human immunodeficiency virus infection after 48 weeks of zidovudine, lamivudine, and ritonavir therapy. J Infect Dis, 2000. 181(1): p. 141–7. [DOI] [PubMed] [Google Scholar]
- 56.Schacker TW, et al. , Amount of lymphatic tissue fibrosis in HIV infection predicts magnitude of HAART-associated change in peripheral CD4 cell count. AIDS, 2005. 19(18): p. 2169–71. [DOI] [PubMed] [Google Scholar]
- 57.Zeng M, et al. , Lymphoid tissue damage in HIV-1 infection depletes naive T cells and limits T cell reconstitution after antiretroviral therapy. PLoS Pathog, 2012. 8(1): p. e1002437. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Chun TW, et al. , Persistence of HIV in gut-associated lymphoid tissue despite long-term antiretroviral therapy. J Infect Dis, 2008. 197(5): p. 714–20. [DOI] [PubMed] [Google Scholar]
- 59.Connors M, et al. , HIV infection induces changes in CD4+ T-cell phenotype and depletions within the CD4+ T-cell repertoire that are not immediately restored by antiviral or immune-based therapies. Nat Med, 1997. 3(5): p. 533–40. [DOI] [PubMed] [Google Scholar]
- 60.Sutherland R, et al. , Impaired IFN-gamma-secreting capacity in mycobacterial antigen-specific CD4 T cells during chronic HIV-1 infection despite long-term HAART. AIDS, 2006. 20(6): p. 821–9. [DOI] [PubMed] [Google Scholar]
- 61.Kaufmann DE, et al. , Upregulation of CTLA-4 by HIV-specific CD4+ T cells correlates with disease progression and defines a reversible immune dysfunction. Nat Immunol, 2007. 8(11): p. 1246–54. [DOI] [PubMed] [Google Scholar]
- 62.Roederer M, et al. , CD8 naive T cell counts decrease progressively in HIV-infected adults. J Clin Invest, 1995. 95(5): p. 2061–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Effros RB, et al. , Shortened telomeres in the expanded CD28−CD8+ cell subset in HIV disease implicate replicative senescence in HIV pathogenesis. AIDS, 1996. 10(8): p. F17–22. [DOI] [PubMed] [Google Scholar]
- 64.Serrano-Villar S, et al. , The CD4:CD8 ratio is associated with markers of age-associated disease in virally suppressed HIV-infected patients with immunological recovery. HIV Med, 2014. 15(1): p. 40–9. [DOI] [PubMed] [Google Scholar]
- 65.Strindhall J, et al. , No Immune Risk Profile among individuals who reach 100 years of age: findings from the Swedish NONA immune longitudinal study. Exp Gerontol, 2007. 42(8): p. 753–61. [DOI] [PubMed] [Google Scholar]
- 66.Serrano-Villar S, et al. , Increased risk of serious non-AIDS-related events in HIV-infected subjects on antiretroviral therapy associated with a low CD4/CD8 ratio. PLoS One, 2014. 9(1): p. e85798. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Helleberg M, et al. , Course and Clinical Significance of CD8+ T-Cell Counts in a Large Cohort of HIV-Infected Individuals. J Infect Dis, 2015. 211(11): p. 1726–34. [DOI] [PubMed] [Google Scholar]
- 68.Badejo OA, et al. , CD8+ T-cells count in acute myocardial infarction in HIV disease in a predominantly male cohort. Biomed Res Int, 2015. 2015: p. 246870. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Appay V, et al. , HIV-specific CD8(+) T cells produce antiviral cytokines but are impaired in cytolytic function. J Exp Med, 2000. 192(1): p. 63–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Migueles SA, et al. , Defective human immunodeficiency virus-specific CD8+ T-cell polyfunctionality, proliferation, and cytotoxicity are not restored by antiretroviral therapy. J Virol, 2009. 83(22): p. 11876–89. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Lisco A, et al. , Identification of rare HIV-1-infected patients with extreme CD4+ T cell decline despite ART-mediated viral suppression. JCI Insight, 2019. 4(8). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Bandera A, et al. , The NLRP3 Inflammasome Is Upregulated in HIV-Infected Antiretroviral Therapy-Treated Individuals with Defective Immune Recovery. Front Immunol, 2018. 9: p. 214. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Schechter ME, et al. , Inflammatory monocytes expressing tissue factor drive SIV and HIV coagulopathy. Sci Transl Med, 2017. 9(405). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Lichtfuss GF, et al. , Virologically suppressed HIV patients show activation of NK cells and persistent innate immune activation. J Immunol, 2012. 189(3): p. 1491–9. [DOI] [PubMed] [Google Scholar]
- 75.Azzoni L, et al. , Sustained impairment of IFN-gamma secretion in suppressed HIV-infected patients despite mature NK cell recovery: evidence for a defective reconstitution of innate immunity. J Immunol, 2002. 168(11): p. 5764–70. [DOI] [PubMed] [Google Scholar]
- 76.Campillo-Gimenez L, et al. , Neutrophils in antiretroviral therapy-controlled HIV demonstrate hyperactivation associated with a specific IL-17/IL-22 environment. J Allergy Clin Immunol, 2014. 134(5): p. 1142–52 e5. [DOI] [PubMed] [Google Scholar]
- 77.Mastroianni CM, et al. , Improvement in neutrophil and monocyte function during highly active antiretroviral treatment of HIV-1-infected patients. AIDS, 1999. 13(8): p. 883–90. [DOI] [PubMed] [Google Scholar]
- 78.Chehimi J, et al. , Persistent decreases in blood plasmacytoid dendritic cell number and function despite effective highly active antiretroviral therapy and increased blood myeloid dendritic cells in HIV-infected individuals. J Immunol, 2002. 168(9): p. 4796–801. [DOI] [PubMed] [Google Scholar]
- 79.Schmidt B, et al. , Variations in plasmacytoid dendritic cell (PDC) and myeloid dendritic cell (MDC) levels in HIV-infected subjects on and off antiretroviral therapy. J Clin Immunol, 2006. 26(1): p. 55–64. [DOI] [PubMed] [Google Scholar]
- 80.Burdo TH, et al. , Soluble CD163 made by monocyte/macrophages is a novel marker of HIV activity in early and chronic infection prior to and after anti-retroviral therapy. J Infect Dis, 2011. 204(1): p. 154–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Hearps AC, et al. , HIV infection induces age-related changes to monocytes and innate immune activation in young men that persist despite combination antiretroviral therapy. AIDS, 2012. 26(7): p. 843–53. [DOI] [PubMed] [Google Scholar]
- 82.Kloverpris HN, et al. , Innate Lymphoid Cells Are Depleted Irreversibly during Acute HIV-1 Infection in the Absence of Viral Suppression. Immunity, 2016. 44(2): p. 391–405. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Kramer B, et al. , Compartment-specific distribution of human intestinal innate lymphoid cells is altered in HIV patients under effective therapy. PLoS Pathog, 2017. 13(5): p. e1006373. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Cosgrove C, et al. , Early and nonreversible decrease of CD161++/MAIT cells in HIV infection. Blood, 2013. 121(6): p. 951–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Leeansyah E, et al. , Activation, exhaustion, and persistent decline of the antimicrobial MR1-restricted MAIT-cell population in chronic HIV-1 infection. Blood, 2013. 121(7): p. 1124–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Kosub DA, et al. , Gamma/Delta T-cell functional responses differ after pathogenic human immunodeficiency virus and nonpathogenic simian immunodeficiency virus infections. J Virol, 2008. 82(3): p. 1155–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Moir S and Fauci AS, B cells in HIV infection and disease. Nat Rev Immunol, 2009. 9(4): p. 235–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Moir S, et al. , Perturbations in B cell responsiveness to CD4+ T cell help in HIV-infected individuals. Proc Natl Acad Sci U S A, 2003. 100(10): p. 6057–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Bekker V, et al. , Persistent humoral immune defect in highly active antiretroviral therapy-treated children with HIV-1 infection: loss of specific antibodies against attenuated vaccine strains and natural viral infection. Pediatrics, 2006. 118(2): p. e315–22. [DOI] [PubMed] [Google Scholar]
- 90.Titanji K, et al. , Loss of memory B cells impairs maintenance of long-term serologic memory during HIV-1 infection. Blood, 2006. 108(5): p. 1580–7. [DOI] [PubMed] [Google Scholar]
- 91.Hart M, et al. , Loss of discrete memory B cell subsets is associated with impaired immunization responses in HIV-1 infection and may be a risk factor for invasive pneumococcal disease. J Immunol, 2007. 178(12): p. 8212–20. [DOI] [PubMed] [Google Scholar]
- 92.Shelburne SA, et al. , Incidence and risk factors for immune reconstitution inflammatory syndrome during highly active antiretroviral therapy. AIDS, 2005. 19(4): p. 399–406. [DOI] [PubMed] [Google Scholar]
- 93.Ratnam I, et al. , Incidence and risk factors for immune reconstitution inflammatory syndrome in an ethnically diverse HIV type 1-infected cohort. Clin Infect Dis, 2006. 42(3): p. 418–27. [DOI] [PubMed] [Google Scholar]
- 94.Murdoch DM, et al. , Incidence and risk factors for the immune reconstitution inflammatory syndrome in HIV patients in South Africa: a prospective study. AIDS, 2008. 22(5): p. 601–10. [DOI] [PubMed] [Google Scholar]
- 95.Muller M, et al. , Immune reconstitution inflammatory syndrome in patients starting antiretroviral therapy for HIV infection: a systematic review and meta-analysis. Lancet Infect Dis, 2010. 10(4): p. 251–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Antonelli LR, et al. , Elevated frequencies of highly activated CD4+ T cells in HIV+ patients developing immune reconstitution inflammatory syndrome. Blood, 2010. 116(19): p. 3818–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Andrade BB, et al. , Mycobacterial antigen driven activation of CD14++CD16− monocytes is a predictor of tuberculosis-associated immune reconstitution inflammatory syndrome. PLoS Pathog, 2014. 10(10): p. e1004433. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Boulware DR, et al. , Clinical features and serum biomarkers in HIV immune reconstitution inflammatory syndrome after cryptococcal meningitis: a prospective cohort study. PLoS Med, 2010. 7(12): p. e1000384. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Tadokera R, et al. , Hypercytokinaemia accompanies HIV-tuberculosis immune reconstitution inflammatory syndrome. Eur Respir J, 2011. 37(5): p. 1248–59. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Meintjes G, et al. , Type 1 helper T cells and FoxP3-positive T cells in HIV-tuberculosis-associated immune reconstitution inflammatory syndrome. Am J Respir Crit Care Med, 2008. 178(10): p. 1083–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Lai RPJ, et al. , HIV-tuberculosis-associated immune reconstitution inflammatory syndrome is characterized by Toll-like receptor and inflammasome signalling. Nat Commun, 2015. 6: p. 8451. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Boulware DR, et al. , Paucity of initial cerebrospinal fluid inflammation in cryptococcal meningitis is associated with subsequent immune reconstitution inflammatory syndrome. J Infect Dis, 2010. 202(6): p. 962–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Chen F, et al. , Characteristics of autoimmune thyroid disease occurring as a late complication of immune reconstitution in patients with advanced human immunodeficiency virus (HIV) disease. Medicine (Baltimore), 2005. 84(2): p. 98–106. [DOI] [PubMed] [Google Scholar]
- 104.Lenner R, et al. , Recurrent pulmonary sarcoidosis in HIV-infected patients receiving highly active antiretroviral therapy. Chest, 2001. 119(3): p. 978–81. [DOI] [PubMed] [Google Scholar]
- 105.O’Leary JG, et al. , De novo autoimmune hepatitis during immune reconstitution in an HIV-infected patient receiving highly active antiretroviral therapy. Clin Infect Dis, 2008. 46(1): p. e12–4. [DOI] [PubMed] [Google Scholar]
- 106.Mastroianni A, Emergence of Sjogren’s syndrome in AIDS patients during highly active antiretroviral therapy. AIDS, 2004. 18(9): p. 1349–52. [DOI] [PubMed] [Google Scholar]
- 107.Sereti I, et al. , Alopecia universalis and Graves’ disease in the setting of immune restoration after highly active antiretroviral therapy. AIDS, 2001. 15(1): p. 138–40. [DOI] [PubMed] [Google Scholar]
- 108.Feinstein MJ, et al. , Patterns of Cardiovascular Mortality for HIV-Infected Adults in the United States: 1999 to 2013. Am J Cardiol, 2016. 117(2): p. 214–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Freiberg MS, et al. , HIV infection and the risk of acute myocardial infarction. JAMA Intern Med, 2013. 173(8): p. 614–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Chow FC, et al. , Comparison of ischemic stroke incidence in HIV-infected and non-HIV-infected patients in a US health care system. J Acquir Immune Defic Syndr, 2012. 60(4): p. 351–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Sico JJ, et al. , HIV status and the risk of ischemic stroke among men. Neurology, 2015. 84(19): p. 1933–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.McKibben RA, et al. , Elevated levels of monocyte activation markers are associated with subclinical atherosclerosis in men with and those without HIV infection. J Infect Dis, 2015. 211(8): p. 1219–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Funderburg NT, et al. , Shared monocyte subset phenotypes in HIV-1 infection and in uninfected subjects with acute coronary syndrome. Blood, 2012. 120(23): p. 4599–608. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Sacre K, et al. , A role for cytomegalovirus-specific CD4+CX3CR1+ T cells and cytomegalovirus-induced T-cell immunopathology in HIV-associated atherosclerosis. AIDS, 2012. 26(7): p. 805–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Fellay J, et al. , Prevalence of adverse events associated with potent antiretroviral treatment: Swiss HIV Cohort Study. Lancet, 2001. 358(9290): p. 1322–7. [DOI] [PubMed] [Google Scholar]
- 116.Madden E, et al. , Association of antiretroviral therapy with fibrinogen levels in HIV-infection. AIDS, 2008. 22(6): p. 707–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Mary-Krause M, et al. , Increased risk of myocardial infarction with duration of protease inhibitor therapy in HIV-infected men. AIDS, 2003. 17(17): p. 2479–86. [DOI] [PubMed] [Google Scholar]
- 118.Dube M and Fenton M, Lipid abnormalities. Clin Infect Dis, 2003. 36(Suppl 2): p. S79–83. [DOI] [PubMed] [Google Scholar]
- 119.Venter WDF, et al. , Dolutegravir plus Two Different Prodrugs of Tenofovir to Treat HIV. N Engl J Med, 2019. 381(9): p. 803–815. [DOI] [PubMed] [Google Scholar]
- 120.Mocroft A, et al. , Is there evidence for an increase in the death rate from liver-related disease in patients with HIV? AIDS, 2005. 19(18): p. 2117–25. [DOI] [PubMed] [Google Scholar]
- 121.Pembroke T, et al. , Hepatic steatosis progresses faster in HIV mono-infected than HIV/HCV co-infected patients and is associated with liver fibrosis. J Hepatol, 2017. 67(4): p. 801–808. [DOI] [PubMed] [Google Scholar]
- 122.Guaraldi G, et al. , Nonalcoholic fatty liver disease in HIV-infected patients referred to a metabolic clinic: prevalence, characteristics, and predictors. Clin Infect Dis, 2008. 47(2): p. 250–7. [DOI] [PubMed] [Google Scholar]
- 123.Odden MC, et al. , Cystatin C level as a marker of kidney function in human immunodeficiency virus infection: the FRAM study. Arch Intern Med, 2007. 167(20): p. 2213–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Mauss S, Berger F, and Schmutz G, Antiretroviral therapy with tenofovir is associated with mild renal dysfunction. AIDS, 2005. 19(1): p. 93–5. [DOI] [PubMed] [Google Scholar]
- 125.Ross AC, et al. , Vitamin D is linked to carotid intima-media thickness and immune reconstitution in HIV-positive individuals. Antivir Ther, 2011. 16(4): p. 555–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Terrier B, et al. , Low 25-OH vitamin D serum levels correlate with severe fibrosis in HIV-HCV co-infected patients with chronic hepatitis. J Hepatol, 2011. 55(4): p. 756–61. [DOI] [PubMed] [Google Scholar]
- 127.Patel P, et al. , Incidence of types of cancer among HIV-infected persons compared with the general population in the United States, 1992–2003. Ann Intern Med, 2008. 148(10): p. 728–36. [DOI] [PubMed] [Google Scholar]
- 128.Kirk GD, et al. , HIV infection is associated with an increased risk for lung cancer, independent of smoking. Clin Infect Dis, 2007. 45(1): p. 103–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Engels EA, et al. , Elevated incidence of lung cancer among HIV-infected individuals. J Clin Oncol, 2006. 24(9): p. 1383–8. [DOI] [PubMed] [Google Scholar]
- 130.Sigel K, Makinson A, and Thaler J, Lung cancer in persons with HIV. Curr Opin HIV AIDS, 2017. 12(1): p. 31–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Ellis R, Langford D, and Masliah E, HIV and antiretroviral therapy in the brain: neuronal injury and repair. Nat Rev Neurosci, 2007. 8(1): p. 33–44. [DOI] [PubMed] [Google Scholar]
- 132.Heaton RK, et al. , HIV-associated neurocognitive disorders persist in the era of potent antiretroviral therapy: CHARTER Study. Neurology, 2010. 75(23): p. 2087–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.McCutchan JA, et al. , HIV suppression by HAART preserves cognitive function in advanced, immune-reconstituted AIDS patients. AIDS, 2007. 21(9): p. 1109–17. [DOI] [PubMed] [Google Scholar]
- 134.Brown TT and Qaqish RB, Antiretroviral therapy and the prevalence of osteopenia and osteoporosis: a meta-analytic review. AIDS, 2006. 20(17): p. 2165–74. [DOI] [PubMed] [Google Scholar]
- 135.Sitbon O, et al. , Prevalence of HIV-related pulmonary arterial hypertension in the current antiretroviral therapy era. Am J Respir Crit Care Med, 2008. 177(1): p. 108–13. [DOI] [PubMed] [Google Scholar]
- 136.Humbert M, et al. , Platelet-derived growth factor expression in primary pulmonary hypertension: comparison of HIV seropositive and HIV seronegative patients. Eur Respir J, 1998. 11(3): p. 554–9. [PubMed] [Google Scholar]
- 137.Desquilbet L, et al. , HIV-1 infection is associated with an earlier occurrence of a phenotype related to frailty. J Gerontol A Biol Sci Med Sci, 2007. 62(11): p. 1279–86. [DOI] [PubMed] [Google Scholar]
- 138.Jordano Q, et al. , Invasive pneumococcal disease in patients infected with HIV: still a threat in the era of highly active antiretroviral therapy. Clin Infect Dis, 2004. 38(11): p. 1623–8. [DOI] [PubMed] [Google Scholar]
- 139.Badri M, et al. , Association between tuberculosis and HIV disease progression in a high tuberculosis prevalence area. Int J Tuberc Lung Dis, 2001. 5(3): p. 225–32. [PubMed] [Google Scholar]
- 140.Lawn SD, et al. , Burden of tuberculosis in an antiretroviral treatment programme in sub-Saharan Africa: impact on treatment outcomes and implications for tuberculosis control. AIDS, 2006. 20(12): p. 1605–12. [DOI] [PubMed] [Google Scholar]
- 141.Salmon-Ceron D, et al. , Liver disease as a major cause of death among HIV infected patients: role of hepatitis C and B viruses and alcohol. J Hepatol, 2005. 42(6): p. 799–805. [DOI] [PubMed] [Google Scholar]
- 142.Blank LJ, et al. , Herpes zoster among persons living with HIV in the current antiretroviral therapy era. J Acquir Immune Defic Syndr, 2012. 61(2): p. 203–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Crum-Cianflone NF, et al. , Immunogenicity of a monovalent 2009 influenza A (H1N1) vaccine in an immunocompromised population: a prospective study comparing HIV-infected adults with HIV-uninfected adults. Clin Infect Dis, 2011. 52(1): p. 138–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.George VK, et al. , HIV infection Worsens Age-Associated Defects in Antibody Responses to Influenza Vaccine. J Infect Dis, 2015. 211(12): p. 1959–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Rey D, et al. , Increasing the number of hepatitis B vaccine injections augments anti-HBs response rate in HIV-infected patients. Effects on HIV-1 viral load. Vaccine, 2000. 18(13): p. 1161–5. [DOI] [PubMed] [Google Scholar]
- 146.Belaunzaran-Zamudio PF, et al. , Early loss of measles antibodies after MMR vaccine among HIV-infected adults receiving HAART. Vaccine, 2009. 27(50): p. 7059–64. [DOI] [PubMed] [Google Scholar]
- 147.Malaspina A, et al. , Compromised B cell responses to influenza vaccination in HIV-infected individuals. J Infect Dis, 2005. 191(9): p. 1442–50. [DOI] [PubMed] [Google Scholar]
- 148.Kroon FP, et al. , Antibody response after influenza vaccination in HIV-infected individuals: a consecutive 3-year study. Vaccine, 2000. 18(26): p. 3040–9. [DOI] [PubMed] [Google Scholar]
- 149.Cooper CL, et al. , CPG 7909 adjuvant plus hepatitis B virus vaccination in HIV-infected adults achieves long-term seroprotection for up to 5 years. Clin Infect Dis, 2008. 46(8): p. 1310–4. [DOI] [PubMed] [Google Scholar]
- 150.Cruciani M, et al. , Serologic response to hepatitis B vaccine with high dose and increasing number of injections in HIV infected adult patients. Vaccine, 2009. 27(1): p. 17–22. [DOI] [PubMed] [Google Scholar]
- 151.Fonseca MO, et al. , Randomized trial of recombinant hepatitis B vaccine in HIV-infected adult patients comparing a standard dose to a double dose. Vaccine, 2005. 23(22): p. 2902–8. [DOI] [PubMed] [Google Scholar]
- 152.Abzug MJ, et al. , Pertussis booster vaccination in HIV-infected children receiving highly active antiretroviral therapy. Pediatrics, 2007. 120(5): p. e1190–202. [DOI] [PubMed] [Google Scholar]
- 153.Rosenblatt HM, et al. , Tetanus immunity after diphtheria, tetanus toxoids, and acellular pertussis vaccination in children with clinically stable HIV infection. J Allergy Clin Immunol, 2005. 116(3): p. 698–703. [DOI] [PubMed] [Google Scholar]
- 154.Golin CE, et al. , A prospective study of predictors of adherence to combination antiretroviral medication. J Gen Intern Med, 2002. 17(10): p. 756–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Mdodo R, et al. , Cigarette smoking prevalence among adults with HIV compared with the general adult population in the United States: cross-sectional surveys. Ann Intern Med, 2015. 162(5): p. 335–44. [DOI] [PubMed] [Google Scholar]
- 156.Conley LJ, et al. , Obesity is associated with greater inflammation and monocyte activation among HIV-infected adults receiving antiretroviral therapy. AIDS, 2015. 29(16): p. 2201–7. [DOI] [PubMed] [Google Scholar]
- 157.Farzadegan H, et al. , Sex differences in HIV-1 viral load and progression to AIDS. Lancet, 1998. 352(9139): p. 1510–4. [DOI] [PubMed] [Google Scholar]
- 158.Jarrin I, et al. , Gender differences in HIV progression to AIDS and death in industrialized countries: slower disease progression following HIV seroconversion in women. Am J Epidemiol, 2008. 168(5): p. 532–40. [DOI] [PubMed] [Google Scholar]
- 159.Reinhold JP, et al. , Colorectal cancer screening in HIV-infected patients 50 years of age and older: missed opportunities for prevention. Am J Gastroenterol, 2005. 100(8): p. 1805–12. [DOI] [PubMed] [Google Scholar]
- 160.Goldman DP, et al. , Effect of insurance on mortality in an HIV-positive population in care (vol 96, pg 883, 2001). Journal of the American Statistical Association, 2002. 97(460): p. 1218–1218. [Google Scholar]
- 161.Spinelli MA, et al. , Homelessness at diagnosis is associated with death among people with HIV in a population-based study of a US city. AIDS, 2019. 33(11): p. 1789–1794. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Crowell TA, et al. , Initiation of antiretroviral therapy before detection of colonic infiltration by HIV reduces viral reservoirs, inflammation and immune activation. J Int AIDS Soc, 2016. 19(1): p. 21163. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Gandhi RT, et al. , No evidence for decay of the latent reservoir in HIV-1-infected patients receiving intensive enfuvirtide-containing antiretroviral therapy. J Infect Dis, 2010. 201(2): p. 293–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Cheret A, et al. , Intensive five-drug antiretroviral therapy regimen versus standard triple-drug therapy during primary HIV-1 infection (OPTIPRIM-ANRS 147): a randomised, open-label, phase 3 trial. Lancet Infect Dis, 2015. 15(4): p. 387–96. [DOI] [PubMed] [Google Scholar]
- 165.McMichael AJ, HIV vaccines. Annu Rev Immunol, 2006. 24: p. 227–55. [DOI] [PubMed] [Google Scholar]
- 166.Valle-Casuso JC, et al. , Cellular Metabolism Is a Major Determinant of HIV-1 Reservoir Seeding in CD4(+) T Cells and Offers an Opportunity to Tackle Infection. Cell Metab, 2019. 29(3): p. 611–626 e5. [DOI] [PubMed] [Google Scholar]
- 167.Group, I.-E.S., et al. , Interleukin-2 therapy in patients with HIV infection. N Engl J Med, 2009. 361(16): p. 1548–59. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Sereti I, et al. , IL-7 administration drives T cell-cycle entry and expansion in HIV-1 infection. Blood, 2009. 113(25): p. 6304–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169.Levy Y, et al. , Enhanced T cell recovery in HIV-1-infected adults through IL-7 treatment. J Clin Invest, 2009. 119(4): p. 997–1007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Prins JM, et al. , Immuno-activation with anti-CD3 and recombinant human IL-2 in HIV-1-infected patients on potent antiretroviral therapy. AIDS, 1999. 13(17): p. 2405–10. [DOI] [PubMed] [Google Scholar]
- 171.Wang FX, et al. , IL-7 is a potent and proviral strain-specific inducer of latent HIV-1 cellular reservoirs of infected individuals on virally suppressive HAART. J Clin Invest, 2005. 115(1): p. 128–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Piconi S, et al. , Hydroxychloroquine drastically reduces immune activation in HIV-infected, antiretroviral therapy-treated immunologic nonresponders. Blood, 2011. 118(12): p. 3263–72. [DOI] [PubMed] [Google Scholar]
- 173.Read SW, et al. , The effect of leflunomide on cycling and activation of T-cells in HIV-1-infected participants. PLoS One, 2010. 5(8): p. e11937. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174.d’Ettorre G, et al. , Probiotics Reduce Inflammation in Antiretroviral Treated, HIV-Infected Individuals: Results of the “Probio-HIV” Clinical Trial. PLoS One, 2015. 10(9): p. e0137200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175.Sortino O, et al. , The Effects of Recombinant Human Lactoferrin on Immune Activation and the Intestinal Microbiome Among Persons Living with Human Immunodeficiency Virus and Receiving Antiretroviral Therapy. J Infect Dis, 2019. 219(12): p. 1963–1968. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Sandler NG, et al. , Sevelamer does not decrease lipopolysaccharide or soluble CD14 levels but decreases soluble tissue factor, low-density lipoprotein (LDL) cholesterol, and oxidized LDL cholesterol levels in individuals with untreated HIV infection. J Infect Dis, 2014. 210(10): p. 1549–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.Zeng M, et al. , Cumulative mechanisms of lymphoid tissue fibrosis and T cell depletion in HIV-1 and SIV infections. J Clin Invest, 2011. 121(3): p. 998–1008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178.Estes JD, et al. , Antifibrotic therapy in simian immunodeficiency virus infection preserves CD4+ T-cell populations and improves immune reconstitution with antiretroviral therapy. J Infect Dis, 2015. 211(5): p. 744–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.Cockerham LR, et al. , A Randomized Controlled Trial of Lisinopril to Decrease Lymphoid Fibrosis in Antiretroviral-Treated, HIV-infected Individuals. Pathog Immun, 2017. 2(3): p. 310–334. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180.Baker JV, et al. , Losartan to reduce inflammation and fibrosis endpoints in HIV disease (LIFE-HIV): results from a randomized placebo-controlled trial. AIDS, 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181.Hunt PW, 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(10): p. 1474–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182.Baker JV, et al. , Factor Xa Inhibition Reduces Coagulation Activity but Not Inflammation Among People With HIV: A Randomized Clinical Trial. Open Forum Infect Dis, 2020. 7(2): p. ofaa026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183.group, A.s., Vorapaxar for HIV-associated inflammation and coagulopathy (ADVICE): a randomised, double-blind, placebo-controlled trial. Lancet HIV, 2018. 5(10): p. e553–e559. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184.Aslangul E, et al. , High-sensitivity C-reactive protein levels fall during statin therapy in HIV-infected patients receiving ritonavir-boosted protease inhibitors. AIDS, 2011. 25(8): p. 1128–31. [DOI] [PubMed] [Google Scholar]
- 185.O’Brien M, et al. , Aspirin attenuates platelet activation and immune activation in HIV-1-infected subjects on antiretroviral therapy: a pilot study. J Acquir Immune Defic Syndr, 2013. 63(3): p. 280–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186.Funderburg NT, 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(4): p. 396–404. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187.Grinspoon SK, et al. , Rationale and design of the Randomized Trial to Prevent Vascular Events in HIV (REPRIEVE). Am Heart J, 2019. 212: p. 23–35. [DOI] [PMC free article] [PubMed] [Google Scholar]