Skip to main content
The Yale Journal of Biology and Medicine logoLink to The Yale Journal of Biology and Medicine
. 2017 Jun 23;90(2):245–259.

Mechanisms of Virologic Control and Clinical Characteristics of HIV+ Elite/Viremic Controllers

Elena Gonzalo-Gil 1, Uchenna Ikediobi 1, Richard E Sutton 1,*
PMCID: PMC5482301  PMID: 28656011

Abstract

Human immunodeficiency virus type 1 (HIV-1) disease is pandemic, with approximately 36 million infected individuals world-wide. For the vast majority of these individuals, untreated HIV eventually causes CD4+ T cell depletion and profound immunodeficiency, resulting in morbidity and mortality. But for a remarkable few (0.2 to 0.5 percent), termed elite controllers (ECs), viral loads (VLs) remain suppressed to undetectable levels (< 50 copies/ml) and peripheral CD4+ T cell counts remain high (200 to 1000/μl), all in the absence of antiretroviral therapy (ART). Viremic controllers (VCs) are a similar but larger subset of HIV-1 infected individuals who have the ability to suppress their VLs to low levels. These patients have been intensively studied over the last 10 years in order to determine how they are able to naturally control HIV in the absence of medications, and a variety of mechanisms have been proposed. Defective HIV does not explain the clinical status of most ECs/VCs; rather these individuals appear to somehow control HIV infection, through immune or other unknown mechanisms. Over time, many ECs and VCs eventually lose the ability to control HIV, leading to CD4+ T cell depletion and immunologic dysfunction in the absence of ART. Elucidating novel mechanisms of HIV control in this group of patients will be an important step in understanding HIV infection. This will extend our knowledge of HIV-host interaction and may pave the way for the development of new therapeutic approaches and advance the cure agenda.

Keywords: HIV, Elite Controllers, Viremic controllers, Long-term non progressors

Introduction

Natural Progression of HIV Infection

Human immunodeficiency virus-1 (HIV-1) is transmitted via hetero or homosexual contact, exchange of infected blood via transfusion and/or the sharing of needles, breast-feeding from an infected mother to child, or trans-placentally from an infected mother to fetus [1,2]. After introduction by sexual contact, virus encounters Langerhans cells, antigen-presenting dendritic cells (DCs) that densely populate mucosal surfaces of the gut and vaginal tissue. And although these cells express low levels of cluster of differentiation 4 (CD4) and C-C Motif Chemokine Receptor 5 (CCR5) on the cell surface, they actively bind to HIV particles via these and other receptors, and facilitate attachment and fusion of viral and host cell membranes [3,4]. Langerhans cells with bound, internalized virus migrate from the mucosal surface to infect neighboring T cells expressing CD4 and CCR5 receptors, before arriving at regional lymph nodes [5]. In addition, these cells are important for priming naive CD4+ T cells into HIV-specific T helper (TH) cells. Local viral replication then occurs, followed by initial detectable plasma viremia, a process called primary infection [4]. Once within regional and draining lymph nodes, infected CD4+ T cells induce T cell activation and proliferation by stimulating HIV-specific CD8+ T cells [6] (Figure 1). The key effector functions of HIV-specific CD8+ T cells are three-fold and include (i) T cell receptor-based recognition of virally-infected cells and subsequent release of perforins and granzyme that are essential to cytotoxic function, (ii) prevention of viral entry via release of competitively binding chemokines such as macrophage inflammatory protein-1 (MIP-1)α, MIP-1β and Chemokine (C-C motif) ligand 5 (RANTES), that serve as chemo-attractants for lymphocytes and monocytes [7,8] and (iii) inhibition of viral replication via production of cytokines and activation of the interferon (IFN) signaling pathway [9] (Figure 1).

Figure 1.

Figure 1

T cell immune responses after HIV-1 infection.

During HIV infection, chemokine ligands inhibit viral entry into cells, preventing viral replication and delaying disease progression by competitively binding to the co-receptors, CCR5 and C-X-C chemokine receptor type 4 (CXCR4), on CD4+ T cells and macrophages [10,11]. The polyfunctional HIV-specific and non-specific CD8+ T cell responses induce cytotoxic killing of HIV-infected cells [12] and establish a general antiviral state by enhancing innate immunity, making cells more resistant to virus replication. This, as well as gastrointestinal microbial translocation which occurs due to depletion of gut-associated lymphoid tissue secondary to early, massive HIV replication, creates a pro-inflammatory environment that drives chronic immune activation and leads to disease progression [13]. One consequence of this, with respect to long-term clinical outcomes in HIV-infected individuals, may be a greater propensity for the development of cardiovascular (CV) and other inflammatory diseases [14]. Even in the presence of antiretroviral therapy (ART) there are low levels of viremia; whether there remains active viral replication in blood and lymphoid tissue is controversial and subject to debate and continued study [15]. But presumably the presence of virus or viral gene products is at least in part responsible for the chronic inflammatory state, which may result in a myriad of untoward consequences, with adverse effects on the health of the infected individual.

HIV-1 Viral Life Cycle and Antiretroviral Therapy


The viral life cycle begins when the envelope (env) glycoprotein gp120 binds to the cell surface receptor CD4 and the membrane co-receptors CCR5 or CXCR4 [16,17] (Figure 2). After fusion of the viral and cellular membranes, the viral particle enters into the cytosol and viral RNA is reverse transcribed into proviral double-stranded cDNA (dscDNA) [18]. Although it is not clear whether reverse transcription occurs within an intact viral capsid core, some studies suggest that post-entry at least a partial capsid core structure is required for optimal reverse transcriptase activity [19,20]. After formation of a pre-integration complex (PIC), dscDNA is imported into the cell nucleus through an intact nuclear pore [21] and the genetic material either circularizes as one or two long terminal repeat (LTR)-containing circles (considered dead-end products) or becomes incorporated irreversibly into the host genome via the catalytic activity of viral integrase [22]. Transcription of integrated provirus yields viral mRNAs of different sizes, which are exported from the nucleus [23,24]. These mRNAs serve as templates for protein production and genome-length RNA is incorporated into nascent viral particles, likely cooperatively assembled at the plasma membrane [25]. Finally the newly made viral particles bud from the plasma membrane and mature through the activity of the viral protease, which cleaves the Gag and Pol polyprotein, to produce fully infectious particles [26].

Figure 2.

Figure 2

HIV-1 viral cycle and FDA-approved therapies and targets (attachment and maturation inhibitors in late stage clinical trials).

In vitro approaches have identified a number of host genes that negatively regulate or interfere with virus replication. These potent HIV restriction factors include tripartite motif-containing protein 5 alpha (TRIM-5α) [27], multiple apolipoprotein B mRNA editing enzyme catalytic (APOBEC) family members [28], the nucleotide hydrolase SAMHD1 [29], SERINC family members, myxovirus resistance protein (MXB), and tetherin [30-32]. Each of these factors acts at distinct steps of the virus lifecycle to inhibit viral replication and yet none has been definitively implicated in viral control in humans [27,30]. On the other hand, many steps of the viral life cycle are targets for ART [33] (Figure 2), and one of the greatest success stories of the last two decades of modern medicine is the widespread use of ART to treat HIV and transform the infection, once considered a death sentence, into a chronic, very manageable disease. Despite this, ART is life-long and non-curative, and once therapy is stopped or drug resistance develops, viral rebound invariably occurs within weeks and CD4 counts then decline [34,35].

As discussed above, HIV-1 infects both activated and resting cells, allowing the viral genome to be permanently integrated into the chromosome of a host T cell or tissue macrophage, cell types that can be very long-lived [36]. Latent, cellular reservoirs of virus are established very early during primary infection, even in the presence of ART, and their very long half-life and consequent slow decay constitutes the major barrier to eradication [37]. Thus, despite the extraordinary advances that have been made in ART over the last two decades, we still have much to learn regarding how to effectively control and eventually eradicate the virus. Identifying novel mechanisms for HIV control bears critical importance to HIV research and treatment, as it will extend our knowledge of the HIV-host interaction and potentially pave the way to new therapeutic approaches.


Elite Controllers, Viremic Controllers, and Long-term Nonprogressors


Elite controllers (ECs) are a small subset of HIV-1 infected individuals (on the order of 1 in 200 to 1 in 500 or 0.2 to 0.5 percent) who have the ability to suppress viremia to undetectable levels (< 50 copies/ml), while maintaining elevated CD4 cell counts (200 to 1000/µl) in the absence of ART [38-42] (Figure 3). These ECs have been intensively investigated over the last several years in order to determine how they are able to naturally control HIV. A similar subset of HIV-infected individuals termed viremic controllers (VCs) achieve a lesser degree of virologic control (200 < VL < 2000 copies/ml), while also maintaining elevated CD4 cell counts (typically ≤ 500/µl), in the absence of ART [43]. ECs and VCs are part of a significantly larger cohort of HIV-infected persons, described as long-term non-progressors (LTNP). LTNP are characterized by their ability to maintain elevated CD4 cell counts in the absence of ART [43].

Figure 3.

Figure 3

Progression of disease after HIV-1 infection in HIV-1 progressors, EC and VC.

These individuals can be identified early during the course of HIV infection and achieve a significantly lower VL set point after sero-conversion [4,44-46]. As a result, collectively, ECs, VCs and LTNP, hereafter termed “controllers,” have more favorable outcomes compared to most HIV-infected individuals who do not have ability to achieve virologic suppression in the absence of ART, termed “non-controllers” (NC) [43]. Although clinical latency in untreated non-controllers may persist for years, because of unrelenting high level viral replication for the vast majority of these patients there is an inexorable loss of CD4+ T cells and immune system decline, eventually resulting in acquired immune deficiency syndrome(AIDS) [47] (Figure 3).


Little is known regarding the precise mechanisms that allow robust control of HIV infection, especially in ECs/VCs. Further investigation into how controllers achieve such a high degree of virologic control may help facilitate efforts directed towards a “functional cure” for HIV, in which the virus is still present in latent reservoirs but never reaches high levels of replication, all in the absence of ART.

Epidemiology and Clinical Definitions of Elite Controllers


As mentioned above the prevalence of the ECs phenotype has been estimated to be < 0.5 percent of the HIV positive patient population [48,49]. The prevalence estimates for VCs have not been entirely elucidated, but are believed to be several-fold higher compared to ECs [43]. These prevalence estimates for ECs reflect data gathered from studied HIV-1+ cohorts within the U.S and Europe. Little is known regarding the prevalence of these controller phenotypes in Sub-Saharan Africa and Asia. In general, there are no consistent demographic patterns among controllers, with respect to gender or race predilection, or differences in modes of HIV transmission between controllers and NCs [43].


As the sensitivity of VL assays improved over time, the definition of who is classified as an ECs has also changed. For example, an individual characterized as having an undetectable VL between 1995 and 2000 (VL of < 500 copies/ml) may not be identified as an ECs today, as the VL assays in routine clinical use are now able to detect less than 20 copies/ml. This is a potential confounder of longitudinal analyses that evaluate clinical outcomes over prolonged periods of time that cross generations of VL assays.


Furthermore, fluctuations in HIV VL are observed naturally during the course of HIV infection and are usually due to concurrent illness or other co-morbidities, receipt of vaccinations, variability or reproducibility of the assay, or inconsistent ART compliance (of course the latter does not pertain to those off therapy) [43]. The ability to not only achieve undetectable VLs, but to sustain them is what differentiates an EC from an NC. Therefore, widely accepted study definitions of an EC include having three or more VL determinations below the limit of assay detection (usually < 50 copies/ml), spanning 12 months or more, in the absence of ART [43,50,51]. Other studies have required a high percentage of VL values below the limit of detection (> 90 percent) over 10 years to define an EC, although these metrics have not been widely employed [48]. VCs are similar, except that 200 < VL < 2000, with an occasional higher or lower value.



Mechanisms of Control of HIV-1 in Elite Controllers


Viral and Host Cell Intrinsic Factors


Some studies have suggested that the viral control is the result of infection with defective viral strains [52,53]. However, other studies have proposed that the majority of control is due to host factors. A summary of the viral proteins and host restriction factors implicated in control in VCs/ECs and their role in viral cycle is included in Table 1 and Figure 4. Infection with highly attenuated HIV was observed in a group of recipients of blood products from a common infected donor. The transmitted virus contained a deletion in the viral accessory gene nef, and all recipients maintained virologic suppression for years [54]. This result contributed to the belief that attenuated virus played a potentially major role in achieving the controller phenotype. Eventually, however, many of these infected individuals progressed to AIDS, thus minimizing the role of defective HIV in the controller phenotype [55].

Table 1. Viral proteins and host restriction factors implicated in control in HIV-1 VCs/ECs.

Viral protein/ Host factor Mechanism of action [ref]
nef • Downregulates surface levels of MHC-I and MHC-II
 [32,138-140]
• Modulates TCR signaling by inducing/ blocking NFAT and IL-2 production in fresh/ activated T cells, respectively

• Prevents incorporation of SERINC-3 and SERINC-5 into HIV-1 virions, enhancing infectivity of the virus
vpu • Downregulates CD4, and BST-2/tetherin [30,141,142]
vif • Binds to and blocks the antiviral activity of APOBEC3 proteins, in conjunction with other host factors, inducing their proteasomal degradation [143]
TRIM-5α • Binds to and multimerizes on the viral capsid, somehow inhibiting viral replication
 [27]
• Initiates innate immune sensing of cytosolic viral capsid

• Counteracted by mutations in viral capsid
Mx2/MxB • Delays HIV-1 DNA nuclear import and integration by targeting viral capsid, exact mechanism of action uncertain
 [31,144]
• Counteracted by mutations in viral capsid
APOBEC3 family members • Inhibits viral reverse transcription and integration [28]
• Induces lethal mutations in viral cDNA

• Counteracted by vif (see above)
Tetherin • Inhibits HIV-1 release by binding virus particles that bud through the cell membrane [30,145]
• Counteracted by vpu (see above)
Serinc-3/5 • Inhibit HIV-1 particle infectivity
 [32]
• Counteracted by nef (see above)

MHC: major histocompatibility complex; TCR: T Cell Receptor; NFAT: nuclear factor of activated T-cells; BST-2: bone marrow stromal antigen 2; APOBEC: apolipoprotein B mRNA editing enzyme 3 catalytic polypeptide; Mx2/McB: myxovirus resistance protein 2; BST-2: bone marrow stromal antigen 2.

Figure 4.

Figure 4

Host restriction factors and lentiviral proteins in HIV replication.

Several reports have concluded that mutations within HIV accessory genes can lead to virus control and disease non-progression [56,57]. These studies support the idea of a relative attenuation of viral protein function in EC-derived HIV sequences. This includes a possible role of mutated HIV vif gene in reduced viral replication in ECs [58,59]. Additionally, the presence of a premature stop codon and a rare mutation in HIV nef and vif genes, respectively, was only observed in some EC patients [60]. Also, infection with attenuated forms of HIV-1, including deletions in nef, contributed to the absence of disease progression in a subset of patients [61,62], and reduced nef function has been shown in some ECs compared to progressors [63]. Recently, a modestly reduced function of HIV gene vpu has been observed in a subgroup of ECs [64]. Most certainly, in chronically infected patients, even ECs, there is a wealth of genetic diversity in HIV sequences, both in plasma and archived as integrated provirus, and how much that contributes to virologic control or viral load set point is highly variable and likely very patient-dependent.


Whether and to what extent that viral genetic diversity contributes to the controller phenotype remains a mostly unanswered question, especially since the in vivo data is strictly correlative. In some ECs, certain polymorphisms within the HIV genome were likely acquired early during the course of infection, rendering the virus somewhat devoid of genetic variability and thus yielding a relatively poorly replicating virus [53,65]. Many of these studies involved small numbers of ECs in whom replication competent viruses were not isolated, thus limiting the generalizability of the conclusions. Other work has isolated and analyzed the genomes of replication-competent virus from ECs and results have shown comparable degrees of genetic variation, replication, and evolution, compared to virus isolated from NC [52]. Thus, perhaps host factors play a more significant role in achieving and sustaining virologic control. Conistent with this idea, Buckheit et al., were able to isolate identical viruses from NC and one EC and another VC, consistent with host factors having a dominant role in the control of the HIV-1 replication [66].

Cellular Immune Responses

Studies have suggested that viral control is strongly correlated with the cellular and humoral immune responses in man [67]. A tight association has been observed between Gag-specific cytotoxic T lymphocyte responses and viral control [68,69], and most notably, HIV-1 specific-CD8+ T cell responses against viral structural proteins have been shown to correlate inversely to set point levels of viral RNA [69]. More recently, greater avidities of Gag-specific T cell and human leukocyte antigen (HLA)-B-restricted responses were seen in vivo in ECs than in NC [70], consistent with these HIV-specific CD4+ and CD8+ T cell immune responses occurring more frequently in ECs than NCs [71,72].

On the other hand, the absence of some of these HIV-specific CD4+ T cell responses has been shown to be a marker of disease progression [73]. CD8+ T cells from ECs have exhibited more polyfunctional capabilities in response to HIV antigens compared to NC, with greater degranulation and release of perforin and granzyme B [74-76]. Furthermore, CD8+ cells from HLA-B*57/5801 ECs were more efficient at eliminating potentially infected resting and activated CD4+ T cells compared to the same cells in progressors [77]. Higher frequency in memory CD8+ CD73+ cells, a subtype involved in the HIV-specific CD8+ T-cell responses, was observed in ECs compared to healthy controls and HIV+ patients, even for those on ART [78]. CD8+ T cells from ECs produced more CD107a, a marker of CD8+ T-cell degranulation following stimulation in response to HIV, compared with NC on ART [79]. Also, CD8+ T cells from ECs and VCs released more inflammatory cytokines and chemokines than NC. These soluble factors included tumor necrosis factor-alpha (TNF-α) and MIP-1β, which facilitate cytotoxic T cell lysis of HIV-infected cells [79,80]. Inhibiting the function of chemokine ligands in vitro led to loss of viral control and replication of HIV in susceptible T lymphocytes [81]. This may serve as one method by which ECs are able to achieve viral control.

Other studies performed in CD4+ T cells isolated from ECs have been aimed at understanding how these individuals are able to control viral replication. CD4+ T cells from ECs have been shown to retain their ability to proliferate and produce interleukin-2 (IL-2) in response to HIV [80,82,83]. Moreover, control of HIV replication has been associated with high levels of HIV-specific interferon-gamma (IFN-γ) CD4+ T cells and lower levels of T-cell activation and HIV-neutralizing antibodies [82-84]. However, HIV-specific CD4+ T cell responses have been suggested not play a direct role in controlling viral replication, at least in non-human primates infected with simian immunodeficiency virus [85].


CD4+ T Cell Phenotype and Susceptibility to HIV Infection


Whether CD4+ T cells from ECs are intrinsically more resistant to HIV infection has also been investigated; these results have been very controversial, dependent on the method of CD4+ T cell stimulation. Polyclonal, PHA-activated ECs, and LTNP CD4+ T cells were susceptible to HIV infection [52].

In contrast, CD3-activated CD4+ T cells from ECs were resistant to HIV infection in culture, independent of co-receptor usage [86,87]. This phenotype was associated with increased levels of the cyclin dependent kinase (CDK) inhibitor p21 [86,88]. Further investigation of the role of p21 in ECs suggests that it may indirectly block HIV reverse transcription by inhibiting CDK2-dependent phosphorylation [89]. A recent study demonstrated that a subset of ECs have CD4+ T cells that produce higher levels of MIP chemokines, suggesting that these cells may be resistant to HIV infection by blocking R5-tropic HIV viral entry [90]. Conversely, HIV infection of CD3-activated CD4+ T cells from ECs and NCs was similar [91]. Non-activated CD4+ T cells from ECs were fully susceptible to HIV infection, similar to those of progressors. Higher levels of viral particle production, however, were observed in NCs compared with ECs [92,93]. Unstimulated CD4+ T cells from ECs exhibited reduced levels of viral integration, compared to those of NCs and HIV-negative controls [94].

On the other hand, ECs do not exhibit some of the immune changes that are observed in NCs. Cytotoxic T-Lymphocyte Antigen-4 (CTLA-4) is upregulated on HIV-specific CD4+ T cells during acute HIV infection, and also correlates with progression of disease. However, this phenotype has not been observed on CD4+ T cells from ECs [95,96]. Interestingly, ECs harbor lower levels of integrated HIV DNA, but higher levels of 2-LTR circular HIV DNA, suggesting a block at genome integration, after nuclear entry [97].


Other groups have studied whether the cellular phenotypes observed in natural killer (NK) cells were associated with ECs phenotype [98]. Undetectable viremia observed in ECs was shown to correlate with a higher percentage of activated NK cells [99]. Also, it has been suggested an increased NK activity in ECs who lacks HIV-1 specific CD8+ T cell responses [100]. Recently, the maintenance of CD4+ T cells in ECs has been associated with the lack of expression of one of the natural cytotoxic receptors in NK cells [101].


Host Genetic Factors


Varied approaches have been taken to identify potential host factors and genes involved in virologic control in both ECs and LTNPs (see Table 2). Several alleles within the HLA-B/C haplotype block have been associated with control, including HLA-B*5701, HLA-C, and HCP5 alleles [39,102-109]. Furthermore, the presence of the CCR5 delta 32 (∆32) allele (a 32 base-pair deletion in CCR5 which renders the co-receptor cytosolic and non-functional) confers protection against seroconversion, with homozygotes being completely resistant to infection by R5-tropic viral strains [110,111]. HIV+ individuals who are heterozygous for the ∆32 CCR5 genotype have relatively normal levels of CD4 T cell surface CCR5 expression but delayed disease progression [112]. Specific alleles of zinc ribbon domain containing (ZNRD1, a subunit of RNA polymerase I) and ring finger protein 39 (RNF39, a poorly characterized gene) were associated with progression [105].

Table 2. Genetic alleles associated with HIV control.

Genes Author Journal, year [ref]
HLA-DRB1*13 Malhotra, U. et al
 J Clin Invest, 2001 [146]
Chen, Y. et al Hum Immunol, 1997 [147]
MICB, TNF, RDBP, BAT1-5, PSORSICI, HLA-C Limou, S. et al J Infect Dis., 2009 [148]
HLA-B57, HLA-C Fellay, J. et al
 Science, 2007 [105]

Trachtenberg, E. et al Genes Immun, 2009 [107]
HLA-B57, HLA-B27 Pereyra, F. et al Science, 2010 [51]
HLA-DRB1*13 and/or HLA-DRB1*06 Ferre, AL. et al J Virol, 2010 [149]
HCP5, HLA-C Han, Y. et al AIDS, 2008 [108]
HLA-B57 Tang, Y. et al
 AIDS, 2010 [109]

Migueles, SA. et al
 J virol, 2003 [104]

Gao, X. et al Nat Med, 2005 [150]

Kiepiela, P et al
 Nature, 2004 [102]

Bailey, J.R. et al J Exp Med, 2006 [103]
HLA-A, HLA-B, CCR3 McLaren, P.J. et al PNAS, 2015 [151]

Genome-wide association studies (GWAS) of HIV-infected cohorts evaluated associations between naturally occurring single nucleotide polymorphisms (SNPs) and particular phenotypes of interest (Table 2). In examining thousands of ECs and NC, the International HIV Controllers Study identified over 300 SNPs located within the chromosome 6 significantly associated with HIV control [51]. Specific amino acid sequences identified within the HLA-B peptide-binding groove were shown to have extremely low P values, lower than any other SNP found by GWAS, or any other HLA allele [113]. Imputed amino acids within the HLA-B peptide-binding groove, in addition to an independent HLA-C effect, explained the associations and the risk and protective alleles, suggesting that very specific interactions between HLA and viral peptides contribute to viral control. In particular, B*57:01, B*27:05, B*14/Cw08:02, B*52 and A*25 alleles were protective, whereas B*35 and C*w07 conferred risk. Importantly, however, only ~20 percent of the protective effect was explained by the identified SNPs [51], suggesting that other, unknown genes and mechanisms are responsible for the observed control.

Additionally, it has been reported that TRIM-5α expression contributes to viral control in EC patients expressing HLA-B*57 or HLA-B*27 alleles [114].

Investigators have also focused on the role of genetic and molecular factors, including those that regulate chromatin and DNA methylation, in viral control. Epigenetic modifications of the HIV promoter have been associated with control of HIV replication and transcription. ECs were shown to have higher levels of DNA methylation in the 5’-LTR compared with progressors [115]. Similarly, lower levels of ccr5 gene DNA methylation were seen in EC and HIV suppressors compared with HIV-negative individuals, indicating an association between ccr5 methylation status and HIV disease [116]. DNA demethylation of regions that regulate PD-1 gene expression in HIV-specific CD8+ T cells was also associated with HIV control, in both ECs and NC on ART [117].


Clinical Outcomes of Controllers vs. Non-controllers on ART


The long-term clinical outcomes of ECs, as compared to NCs, have been mainly focused on progression to AIDS and AIDS-related death [43]. More is now known about the non-AIDS related clinical outcomes of ECs and the role that chronic immune activation plays in their outcome.

Several retrospective studies have tried to better understand the potential benefit of early ART in modifying both AIDS-related and non-AIDS related outcomes in controllers [118]. A summary of these studies examining clinical outcomes of ECs is provided below (Table 3).

Table 3. Summary of retrospective cohort studies of clinical outcomes in ECs.

Study population Sample size (N) Primary outcome Study period Relevant results Ref
HIV+ in the military healthcare system Total (4,586) Time to develop AIDS 1986-2006 1. Time to virologic suppression was early after infection (less than 1 year from the time of sero-conversion) in most ECs/VCs [43]
• EC (25)
 2. ECs/VCs had fewer deaths and AIDS-defining events, and longer time to AIDS and death compared to NC
• VC (153) 3. Individuals achieving LTNP status for 10 years had more favorable time to AIDS and death compared to LTNP reaching their status for 7 years

• LTNP 10 (52)
• LTNP 7 (101)
HIV research network Total (34,000)
 All-cause hospitalization rates 2005-2011 1. ECs had higher rates of hospitalization rates due to CV disease and psychiatric illness, compared to NC under ART [135]
• EC (149)
 2. ECs were more likely to be hospitalized than VCs (with both high and low VL) due to CV diseases
High/ low VL (12,847/ 12,044)
• NC (9,226)
US military HIV+ natural history Total (1091)
 Non-AIDS 2000-2013 1. Non-AIDS infection was the most common reason for hospitalizations in all groups, ECs, VCs and progressors on therapy [134]
• EC (33)
 2. No differences in hospitalization rates associated with CV disease between groups, suggesting longer follow up of patients may be needed
• VC (188)

• Progressors on ART (870)
HIV+ patients from a University Hospital Total (574) Non-AIDS and AIDS events 1996-2011 1. Non-AIDS-defining malignancies were the most common reason for hospitalization, followed by CV and neuropsychiatric illnesses
 [152]
• EC (64)
 2. The risk of non-AIDS events was comparable in ECs, VCs and NCs

• VC (76)
 3. Only controllers who retained spontaneous control during the entire follow-up period had a lower risk of non-AIDS events
• NC (434)

EC: elite controller; VC: viremic controller; LTNP: long-term non-progressor; LTNP 7: LTNP through 7 years of follow-up; LTNP 10: LTNP through 10 years of follow-up; NC: non-controller; ART: antiretroviral therapy; CV: cardiovascular; VL: viral load; AIDS: acquired immune deficiency syndrome

AIDS-associated Clinical Outcomes


CD4+ count is the most well-recognized and reliable clinical indicator of HIV disease progression. For many years CD4+ number was paramount in treatment guidelines regarding timing of ART initiation [119-121].

Although early, high HIV RNA levels have been associated with CD4 decline [120,122], it is subsequent or set point viral RNA levels that have a greater prognostic impact on disease progression [123]. ECs achieve lower early baseline and set point viral RNA levels compared to NCs, and therefore have lower rates of AIDS progression and associated mortality [44,45,120,122] (Figure 3).


In a retrospective study, Okulicz et al [43] showed that among most ECs/VCs, virologic suppression occurred early after infection, and in most cases, during the first year from the time of known seroconversion. However, they did uncover differences between ECs and VCs, including more stable and higher CD4 counts in ECs than VCs. They also evaluated the time to AIDS and death among LTNP through 7 years of follow-up and 10 years of follow-up, depending on the duration of non-progression. Results showed that individuals achieving LTNP status for 10 years had more favorable time to AIDS and death compared to those achieving LTNP status earlier. Eventually, however, some VCs did progress to AIDS and death, reaffirming the notion that loss of virologic control and immune function occurs in some of these individuals. In fact, a study of more than four hundred ECs revealed that almost 30 percent of them lost viral control, resulting in reduced CD4 counts, underscoring the concept that many of these patients may eventually progress to AIDS [124].

Non AIDS-associated Clinical Outcomes


ECs and VCs have higher levels of circulating inflammatory cytokines and rates of coronary atherosclerosis compared to NC on ART [125,126], suggesting that the chronic inflammation present may account for early vascular dysfunction [127]. Several factors contribute to the heightened inflammatory cellular setting of ECs and VCs compared to NC on ART, including gut microbial translocation and chronic T cell activation [128,129]. Gut microbial translocation is measured as circulating lipopolysaccharide (LPS) levels, and its presence has been attributed to viremia and disease progression [13,130,131]. Thus, it has been clearly demonstrated that ART reduces LPS levels and reduces the degree of immune activation [132,133]. The inflammatory environment that may be responsible for maintaining strict virological control in ECs and VCs may also portend unfavorable long-term clinical outcomes. A recent retrospective study revealed that non-AIDS-defining infections were the most common reason for hospitalization in ECs, with the same rates of hospitalization due to CV disease in both progressors on ART and ECs [134]. Crowell and colleagues [135] showed, however, that compared to NCs on ART, ECs had higher rates of all-cause hospitalizations due to CV disease and psychiatric illness. In light of what is known about the association of inflammatory cytokines and coronary artery disease in controllers, perhaps this finding is not surprising. That VCs had more favorable all-cause hospitalization rates due to CV disease compared to ECs was nonetheless unanticipated and certainly counter-intuitive.

In light of these findings it was of interest to determine the clinical outcomes of ECs and VCs after beginning ART. A recent study demonstrated an increase in CD4 number after ART initiation in both ECs and VCs, although it was somewhat better in the former [136]. Treatment with ART in ECs and VCs for six months reduced levels of immune activation markers and HIV VL (the latter in VCs), indicating that the use of ART in this setting may be beneficial [137]. Whether there are long-term, meaningful, and lasting differences in clinical outcomes remains to be established. Per current DHHS guidelines, starting ECs on ART is an individualized decision; all VCs should be on ART given their higher, detectable VLs.


Conclusions

ECs and VCs are able to achieve spontaneous control of viral replication to differing degrees, in the absence of antiretroviral medications. This relatively rare ability is thought to be mediated via either viral or host immune or genetic factors. HIV-specific immune activation, a greater poly-functional CD8+ T cell response, and HIV-specific CD4+ T cell responses in ECs may indeed play a significant role in reducing VL and delaying disease progression. Also, ECs have SNPs within the HLA loci that are significantly associated with viral control and finely map to the peptide-binding groove of the class I molecule. Functional and biochemical studies, however, are required to confirm the role of these amino acid residues in virologic suppression. Additional studies are necessary to pinpoint novel pathways and causal host genes responsible for virologic control, especially since the SNPs observed in the HLA loci can only explain ~20 percent of the EC phenotype. A better understanding of the mechanisms that underlie virologic control and the long-term clinical outcomes of ECs/VCs may help inform the ‘HIV cure’ agenda and lead to a better quality of life, even for HIV+ progressors.


Glossary

HIV-1

Human immunodeficiency virus-1

DC

dendritic cell

CD4

cluster of differentiation 4

CCR5

C-C Motif Chemokine Receptor 5

TH

T helper

MIP-1

macrophage inflammatory protein-1

RANTES

Chemokine (C-C motif) ligand 5

CXCR4

C-X-C chemokine receptor type 4

CV

cardiovascular

ART

antiretroviral therapy

env

envelope

dscDNA

double-stranded cDNA

PIC

pre-integration complex

LTR

long terminal repeat

TRIM-5α

tripartite motif-containing protein-5 alpha

APOBEC

apolipoprotein B mRNA editing enzyme catalytic

EC

elite controller

VC

viremic controller

LTNP

long-term non-progressor

LTNP 7

LTNP through 7 years of follow-up

LTNP 10

LTNP through 10 years of follow-up

NC

non-controller

VL

viral load

AIDS

acquired immune deficiency syndrome

HLA

human leukocyte antigen

TNF-α

tumor necrosis factor-alpha

IL-2

interleukin-2

IFN-γ

interferon-gamma

CDK

cyclin dependent kinase

CTLA-4

Cytotoxic T-Lymphocyte Antigen-4

NK

natural killer

ZNRD1

zinc ribbon domain

RNF39

ring finger protein 39

GWAS

Genome-wide association studies

SNP

single nucleotide polymorphisms

LPS

lipopolysaccharide

Mx2/McB

myxovirus resistance protein 2

NFAT

nuclear factor of activated T-cells

BST-2

bone marrow stromal antigen 2

Author contributions

EGG, RES, and UI wrote and edited the paper. Funding sources: NIH-NIDA DP1DA036463 (RES and EGG); NIH-NIAID T32AI007517 (UI).

References

  1. Fox J, Fidler S. Sexual transmission of HIV-1. Antiviral Res. 2010;85(1):276–285. doi: 10.1016/j.antiviral.2009.10.012. [DOI] [PubMed] [Google Scholar]
  2. Hansasuta P, Rowland-Jones SL. HIV-1 transmission and acute HIV-1 infection. Br Med Bull. 2001;58:109–127. doi: 10.1093/bmb/58.1.109. [DOI] [PubMed] [Google Scholar]
  3. Wilen CB, Tilton JC, Doms RW. HIV: cell binding and entry. Cold Spring Harb Perspect Med. 2012;2(8) doi: 10.1101/cshperspect.a006866. [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Weber J. The pathogenesis of HIV-1 infection. Br Med Bull. 2001;58:61–72. doi: 10.1093/bmb/58.1.61. [DOI] [PubMed] [Google Scholar]
  5. Gupta P, Collins KB, Ratner D. et al. Memory CD4(+) T cells are the earliest detectable human immunodeficiency virus type 1 (HIV-1)-infected cells in the female genital mucosal tissue during HIV-1 transmission in an organ culture system. J Virol. 2002;76(19):9868–9876. doi: 10.1128/JVI.76.19.9868-9876.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Wilen CB, Tilton JC, Doms RW. Molecular mechanisms of HIV entry. Adv Exp Med Biol. 2012;726:223–242. doi: 10.1007/978-1-4614-0980-9_10. [DOI] [PubMed] [Google Scholar]
  7. Graw F, Regoes RR. Predicting the impact of CD8+ T cell polyfunctionality on HIV disease progression. J Virol. 2014;88(17):10134–10145. doi: 10.1128/JVI.00647-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Miyagishi R, Kikuchi S, Takayama C. et al. Identification of cell types producing RANTES, MIP-1 alpha and MIP-1 beta in rat experimental autoimmune encephalomyelitis by in situ hybridization. J Neuroimmunol. 1997;77(1):17–26. doi: 10.1016/s0165-5728(97)00040-4. [DOI] [PubMed] [Google Scholar]
  9. Norris PJ, Pappalardo BL, Custer B. et al. Elevations in IL-10, TNF-alpha, and IFN-gamma from the earliest point of HIV Type 1 infection. AIDS Res Hum Retroviruses. 2006;22(8):757–762. doi: 10.1089/aid.2006.22.757. [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Paxton WA, Martin SR, Tse D. et al. Relative resistance to HIV-1 infection of CD4 lymphocytes from persons who remain uninfected despite multiple high-risk sexual exposure. Nat Med. 1996;2(4):412–417. doi: 10.1038/nm0496-412. [DOI] [PubMed] [Google Scholar]
  11. Saha K, Bentsman G, Chess L. et al. Endogenous production of beta-chemokines by CD4+, but not CD8+, T-cell clones correlates with the clinical state of human immunodeficiency virus type 1 (HIV-1)-infected individuals and may be responsible for blocking infection with non-syncytium-inducing HIV-1 in vitro. J Virol. 1998;72(1):876–881. doi: 10.1128/jvi.72.1.876-881.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Hersperger AR, Pereyra F, Nason M. et al. Perforin expression directly ex vivo by HIV-specific CD8 T-cells is a correlate of HIV elite control. PLoS Pathog. 2010;6(5):e1000917. doi: 10.1371/journal.ppat.1000917. [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Marchetti G, Tincati C, Silvestri G. Microbial translocation in the pathogenesis of HIV infection and AIDS. Clin Microbiol Rev. 2013;26(1):2–18. doi: 10.1128/CMR.00050-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Triant VA. Cardiovascular disease and HIV infection. Curr HIV/AIDS Rep. 2013;10(3):199–206. doi: 10.1007/s11904-013-0168-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Hsue PY, Deeks SG, Hunt PW. et al. Immunologic basis of cardiovascular disease in HIV-infected adults. J Infect Dis. 2012;205(Suppl 3):S375–S382. doi: 10.1093/infdis/jis200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Choe H, Farzan M, Sun Y. et al. The beta-chemokine receptors CCR3 and CCR5 facilitate infection by primary HIV-1 isolates. Cell. 1996;85(7):1135–1148. doi: 10.1016/s0092-8674(00)81313-6. [DOI] [PubMed] [Google Scholar]
  17. Feng Y, Broder CC, Kennedy PE. et al. HIV-1 entry cofactor: functional cDNA cloning of a seven-transmembrane, G protein-coupled receptor. Science. 1996;272(5263):872–877. doi: 10.1126/science.272.5263.872. [DOI] [PubMed] [Google Scholar]
  18. Bukrinsky MI, Stanwick TL, Dempsey MP. et al. Quiescent T lymphocytes as an inducible virus reservoir in HIV-1 infection. Science. 1991;254(5030):423–427. doi: 10.1126/science.1925601. [DOI] [PMC free article] [PubMed] [Google Scholar]
  19. Iordanskiy S, Bukrinsky M. Reverse transcription complex: the key player of the early phase of HIV replication. Future Virol. 2007;2(1):49–64. doi: 10.2217/17460794.2.1.49. [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Hulme AE, Perez O, Hope TJ. Complementary assays reveal a relationship between HIV-1 uncoating and reverse transcription. Proc Natl Acad Sci U S A. 2011;108(24):9975–9980. doi: 10.1073/pnas.1014522108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  21. Bukrinsky MI, Sharova N, Dempsey MP. et al. Active nuclear import of human immunodeficiency virus type 1 preintegration complexes. Proc Natl Acad Sci U S A. 1992;89(14):6580–6584. doi: 10.1073/pnas.89.14.6580. [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. Arhel NJ, Souquere-Besse S, Munier S. et al. HIV-1 DNA Flap formation promotes uncoating of the pre-integration complex at the nuclear pore. EMBO J. 2007;26(12):3025–3037. doi: 10.1038/sj.emboj.7601740. [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. Malim MH, Hauber J, Le SY. et al. The HIV-1 rev trans-activator acts through a structured target sequence to activate nuclear export of unspliced viral mRNA. Nature. 1989;338(6212):254–257. doi: 10.1038/338254a0. [DOI] [PubMed] [Google Scholar]
  24. Felber BK, Hadzopoulou-Cladaras M, Cladaras C. et al. rev protein of human immunodeficiency virus type 1 affects the stability and transport of the viral mRNA. Proc Natl Acad Sci U S A. 1989;86(5):1495–1499. doi: 10.1073/pnas.86.5.1495. [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Swanson CM, Malim MH. Retrovirus RNA trafficking: from chromatin to invasive genomes. Traffic. 2006;7(11):1440–1450. doi: 10.1111/j.1600-0854.2006.00488.x. [DOI] [PubMed] [Google Scholar]
  26. Sundquist WI, Krausslich HG. HIV-1 assembly, budding, and maturation. Cold Spring Harb Perspect Med. 2012;2(7):a006924. doi: 10.1101/cshperspect.a006924. [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. Stremlau M, Owens CM, Perron MJ. et al. The cytoplasmic body component TRIM5alpha restricts HIV-1 infection in Old World monkeys. Nature. 2004;427(6977):848–853. doi: 10.1038/nature02343. [DOI] [PubMed] [Google Scholar]
  28. Sheehy AM, Gaddis NC, Choi JD. et al. Isolation of a human gene that inhibits HIV-1 infection and is suppressed by the viral Vif protein. Nature. 2002;418(6898):646–650. doi: 10.1038/nature00939. [DOI] [PubMed] [Google Scholar]
  29. Laguette N, Sobhian B, Casartelli N. et al. SAMHD1 is the dendritic- and myeloid-cell-specific HIV-1 restriction factor counteracted by Vpx. Nature. 2011;474(7353):654–657. doi: 10.1038/nature10117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Neil SJ, Zang T, Bieniasz PD. Tetherin inhibits retrovirus release and is antagonized by HIV-1 Vpu. Nature. 2008;451(7177):425–430. doi: 10.1038/nature06553. [DOI] [PubMed] [Google Scholar]
  31. Liu Z, Pan Q, Ding S. et al. The interferon-inducible MxB protein inhibits HIV-1 infection. Cell Host Microbe. 2013;14(4):398–410. doi: 10.1016/j.chom.2013.08.015. [DOI] [PubMed] [Google Scholar]
  32. Usami Y, Wu Y, Gottlinger HG. SERINC3 and SERINC5 restrict HIV-1 infectivity and are counteracted by Nef. Nature. 2015;526(7572):218–223. doi: 10.1038/nature15400. [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Laskey SB, Siliciano RF. A mechanistic theory to explain the efficacy of antiretroviral therapy. Nat Rev Microbiol. 2014;12(11):772–780. doi: 10.1038/nrmicro3351. [DOI] [PubMed] [Google Scholar]
  34. Strategies for Management of Antiretroviral Therapy Study Group; El-Sadr WM, Lundgren J, Neaton JD. et al. CD4+ count-guided interruption of antiretroviral treatment. N Engl J Med. 2006;355(22):2283–2296. doi: 10.1056/NEJMoa062360. [DOI] [PubMed] [Google Scholar]
  35. Holkmann Olsen C, Mocroft A, Kirk O. et al. Interruption of combination antiretroviral therapy and risk of clinical disease progression to AIDS or death. HIV Med. 2007;8(2):96–104. doi: 10.1111/j.1468-1293.2007.00436.x. [DOI] [PubMed] [Google Scholar]
  36. Chavez L, Calvanese V, Verdin E. HIV Latency Is Established Directly and Early in Both Resting and Activated Primary CD4 T Cells. PLoS Pathog. 2015;11(6):e1004955. doi: 10.1371/journal.ppat.1004955. [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. Siliciano RF, Greene WC. HIV latency. Cold Spring Harb Perspect Med. 2011;1(1):a007096. doi: 10.1101/cshperspect.a007096. [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Okulicz JF, Lambotte O. Epidemiology and clinical characteristics of elite controllers. Curr Opin HIV AIDS. 2011;6(3):163–168. doi: 10.1097/COH.0b013e328344f35e. [DOI] [PubMed] [Google Scholar]
  39. Lambotte O, Boufassa F, Madec Y. et al. HIV controllers: a homogeneous group of HIV-1-infected patients with spontaneous control of viral replication. Clin Infect Dis. 2005;41(7):1053–1056. doi: 10.1086/433188. [DOI] [PubMed] [Google Scholar]
  40. Deeks SG, Walker BD. Human immunodeficiency virus controllers: mechanisms of durable virus control in the absence of antiretroviral therapy. Immunity. 2007;27(3):406–416. doi: 10.1016/j.immuni.2007.08.010. [DOI] [PubMed] [Google Scholar]
  41. O'Connell KA, Bailey JR, Blankson JN. Elucidating the elite: mechanisms of control in HIV-1 infection. Trends Pharmacol Sci. 2009;30(12):631–637. doi: 10.1016/j.tips.2009.09.005. [DOI] [PubMed] [Google Scholar]
  42. Blankson JN. Effector mechanisms in HIV-1 infected elite controllers: highly active immune responses? Antiviral Res. 2010;85(1):295–302. doi: 10.1016/j.antiviral.2009.08.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  43. Okulicz JF, Marconi VC, Landrum ML. et al. Clinical outcomes of elite controllers, viremic controllers, and long-term nonprogressors in the US Department of Defense HIV natural history study. J Infect Dis. 2009;200(11):1714–1723. doi: 10.1086/646609. [DOI] [PubMed] [Google Scholar]
  44. Lavreys L, Baeten JM, Chohan V. et al. Higher set point plasma viral load and more-severe acute HIV type 1 (HIV-1) illness predict mortality among high-risk HIV-1-infected African women. Clin Infect Dis. 2006;42(9):1333–1339. doi: 10.1086/503258. [DOI] [PubMed] [Google Scholar]
  45. Mellors JW, Kingsley LA, Rinaldo CR, Jr. et al. Quantitation of HIV-1 RNA in plasma predicts outcome after seroconversion. Ann Intern Med. 1995;122(8):573–579. doi: 10.7326/0003-4819-122-8-199504150-00003. [DOI] [PubMed] [Google Scholar]
  46. Mellors JW, Rinaldo CR, Jr. Prognosis in HIV-1 infection predicted by the quantity of virus in plasma. Science. 1996;272(5265):1167–1170. doi: 10.1126/science.272.5265.1167. [DOI] [PubMed] [Google Scholar]
  47. Pantaleo G, Graziosi C, Fauci AS. et al. New concepts in the immunopathogenesis of human immunodeficiency virus infection. N Engl J Med. 1993;328(5):327–335. doi: 10.1056/NEJM199302043280508. [DOI] [PubMed] [Google Scholar]
  48. Lambotte O, Boufassa F, Madec Y. et al. HIV controllers: a homogeneous group of HIV-1-infected patients with spontaneous control of viral replication. Clin Infect Dis. 2005;41(7):1053–1056. doi: 10.1086/433188. [DOI] [PubMed] [Google Scholar]
  49. Grabar S, Selinger-Leneman H, Abgrall S. et al. Prevalence and comparative characteristics of long-term nonprogressors and HIV controller patients in the French Hospital Database on HIV. AIDS. 2009;23(9):1163–1169. doi: 10.1097/QAD.0b013e32832b44c8. [DOI] [PubMed] [Google Scholar]
  50. Walker BD. Elite control of HIV Infection: implications for vaccines and treatment. Top HIV Med. 2007;15(4):134–136. [PubMed] [Google Scholar]
  51. International HIVCS; Pereyra F, Jia X, McLaren PJ. et al. The major genetic determinants of HIV-1 control affect HLA class I peptide presentation. Science. 2010;330(6010):1551–1557. doi: 10.1126/science.1195271. [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. Blankson JN, Bailey JR, Thayil S. et al. Isolation and characterization of replication-competent human immunodeficiency virus type 1 from a subset of elite suppressors. J Virol. 2007;81(5):2508–2518. doi: 10.1128/JVI.02165-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  53. Wang B, Dyer WB, Zaunders JJ. et al. Comprehensive analyses of a unique HIV-1-infected nonprogressor reveal a complex association of immunobiological mechanisms in the context of replication-incompetent infection. Virology. 2002;304(2):246–264. doi: 10.1006/viro.2002.1706. [DOI] [PubMed] [Google Scholar]
  54. Deacon NJ, Tsykin A, Solomon A. et al. Genomic structure of an attenuated quasi species of HIV-1 from a blood transfusion donor and recipients. Science. 1995;270(5238):988–991. doi: 10.1126/science.270.5238.988. [DOI] [PubMed] [Google Scholar]
  55. Churchill MJ, Rhodes DI, Learmont JC. et al. Longitudinal analysis of human immunodeficiency virus type 1 nef/long terminal repeat sequences in a cohort of long-term survivors infected from a single source. J Virol. 2006;80(2):1047–1052. doi: 10.1128/JVI.80.2.1047-1052.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  56. Lum JJ, Cohen OJ, Nie Z. et al. Vpr R77Q is associated with long-term nonprogressive HIV infection and impaired induction of apoptosis. J Clin Invest. 2003;111(10):1547–1554. doi: 10.1172/JCI16233. [DOI] [PMC free article] [PubMed] [Google Scholar]
  57. Yamada T, Iwamoto A. Comparison of proviral accessory genes between long-term nonprogressors and progressors of human immunodeficiency virus type 1 infection. Arch Virol. 2000;145(5):1021–1027. doi: 10.1007/s007050050692. [DOI] [PubMed] [Google Scholar]
  58. Kikuchi T, Iwabu Y, Tada T. et al. Anti-APOBEC3G activity of HIV-1 Vif protein is attenuated in elite controllers. J Virol. 2015;89(9):4992–5001. doi: 10.1128/JVI.03464-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  59. Hassaine G, Agostini I, Candotti D. et al. Characterization of human immunodeficiency virus type 1 vif gene in long-term asymptomatic individuals. Virology. 2000;276(1):169–180. doi: 10.1006/viro.2000.0543. [DOI] [PubMed] [Google Scholar]
  60. Cruz NV, Amorim R, Oliveira FE. et al. Mutations in the nef and vif genes associated with progression to AIDS in elite controller and slow-progressor patients. J Med Virol. 2013;85(4):563–574. doi: 10.1002/jmv.23512. [DOI] [PubMed] [Google Scholar]
  61. Kirchhoff F, Greenough TC, Brettler DB. et al. Brief report: absence of intact nef sequences in a long-term survivor with nonprogressive HIV-1 infection. N Engl J Med. 1995;332(4):228–232. doi: 10.1056/NEJM199501263320405. [DOI] [PubMed] [Google Scholar]
  62. Huang Y, Zhang L, Ho DD. et al. Characterization of nef sequences in long-term survivors of human immunodeficiency virus type 1 infection. J Virol. 1995;69(1):93–100. doi: 10.1128/jvi.69.1.93-100.1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  63. Mwimanzi P, Markle TJ, Martin E. et al. Attenuation of multiple Nef functions in HIV-1 elite controllers. Retrovirology. 2013;10:1. doi: 10.1186/1742-4690-10-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. Chen J, Tibroni N, Sauter D. et al. Modest attenuation of HIV-1 Vpu alleles derived from elite controller plasma. PLoS One. 2015;10(3):e0120434. doi: 10.1371/journal.pone.0120434. [DOI] [PMC free article] [PubMed] [Google Scholar]
  65. Miura T, Brockman MA, Brumme ZL. et al. HLA-associated alterations in replication capacity of chimeric NL4-3 viruses carrying gag-protease from elite controllers of human immunodeficiency virus type 1. J Virol. 2009;83(1):140–149. doi: 10.1128/JVI.01471-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  66. Buckheit RW 3rd, Allen TG, Alme A. et al. Host factors dictate control of viral replication in two HIV-1 controller/chronic progressor transmission pairs. Nat Commun. 2012;3:716. doi: 10.1038/ncomms1697. [DOI] [PMC free article] [PubMed] [Google Scholar]
  67. Pernas M, Casado C, Arcones C. et al. Low-replicating viruses and strong anti-viral immune response associated with prolonged disease control in a superinfected HIV-1 LTNP elite controller. PLoS One. 2012;7(2):e31928. doi: 10.1371/journal.pone.0031928. [DOI] [PMC free article] [PubMed] [Google Scholar]
  68. Buseyne F, Scott-Algara D, Porrot F. et al. Frequencies of ex vivo-activated human immunodeficiency virus type 1-specific gamma-interferon-producing CD8+ T cells in infected children correlate positively with plasma viral load. J Virol. 2002;76(24):12414–12422. doi: 10.1128/JVI.76.24.12414-12422.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  69. Edwards BH, Bansal A, Sabbaj S. et al. Magnitude of functional CD8+ T-cell responses to the gag protein of human immunodeficiency virus type 1 correlates inversely with viral load in plasma. J Virol. 2002;76(5):2298–2305. doi: 10.1128/jvi.76.5.2298-2305.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  70. Berger CT, Frahm N, Price DA. et al. High-functional-avidity cytotoxic T lymphocyte responses to HLA-B-restricted Gag-derived epitopes associated with relative HIV control. J Virol. 2011;85(18):9334–9345. doi: 10.1128/JVI.00460-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  71. Ferrando-Martinez S, Casazza JP, Leal M. et al. Differential Gag-specific polyfunctional T cell maturation patterns in HIV-1 elite controllers. J Virol. 2012;86(7):3667–3674. doi: 10.1128/JVI.07034-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  72. Turk G, Ghiglione Y, Falivene J. et al. Early Gag immunodominance of the HIV-specific T-cell response during acute/early infection is associated with higher CD8+ T-cell antiviral activity and correlates with preservation of the CD4+ T-cell compartment. J Virol. 2013;87(13):7445–7462. doi: 10.1128/JVI.00865-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  73. Krowka JF, Stites DP, Jain S. et al. Lymphocyte proliferative responses to human immunodeficiency virus antigens in vitro. J Clin Invest. 1989;83(4):1198–1203. doi: 10.1172/JCI114001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  74. Betts MR, Nason MC, West SM. et al. HIV nonprogressors preferentially maintain highly functional HIV-specific CD8+ T cells. Blood. 2006;107(12):4781–4789. doi: 10.1182/blood-2005-12-4818. [DOI] [PMC free article] [PubMed] [Google Scholar]
  75. Migueles SA, Laborico AC, Shupert WL. et al. HIV-specific CD8+ T cell proliferation is coupled to perforin expression and is maintained in nonprogressors. Nat Immunol. 2002;3(11):1061–1068. doi: 10.1038/ni845. [DOI] [PubMed] [Google Scholar]
  76. Almeida JR, Price DA, Papagno L. et al. Superior control of HIV-1 replication by CD8+ T cells is reflected by their avidity, polyfunctionality, and clonal turnover. J Exp Med. 2007;204(10):2473–2485. doi: 10.1084/jem.20070784. [DOI] [PMC free article] [PubMed] [Google Scholar]
  77. Buckheit RW 3rd, Siliciano RF, Blankson JN. Primary CD8+ T cells from elite suppressors effectively eliminate non-productively HIV-1 infected resting and activated CD4+ T cells. Retrovirology. 2013;10:68. doi: 10.1186/1742-4690-10-68. [DOI] [PMC free article] [PubMed] [Google Scholar]
  78. Carriere M, Lacabaratz C, Kok A. et al. HIV "elite controllers" are characterized by a high frequency of memory CD8+ CD73+ T cells involved in the antigen-specific CD8+ T-cell response. J Infect Dis. 2014;209(9):1321–1330. doi: 10.1093/infdis/jit643. [DOI] [PubMed] [Google Scholar]
  79. Ferre AL, Hunt PW, Critchfield JW. et al. Mucosal immune responses to HIV-1 in elite controllers: a potential correlate of immune control. Blood. 2009;113(17):3978–3989. doi: 10.1182/blood-2008-10-182709. [DOI] [PMC free article] [PubMed] [Google Scholar]
  80. Emu B, Sinclair E, Hatano H. et al. HLA class I-restricted T-cell responses may contribute to the control of human immunodeficiency virus infection, but such responses are not always necessary for long-term virus control. J Virol. 2008;82(11):5398–5407. doi: 10.1128/JVI.02176-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  81. Demers KR, Reuter MA, Betts MR. CD8(+) T-cell effector function and transcriptional regulation during HIV pathogenesis. Immunol Rev. 2013;254(1):190–206. doi: 10.1111/imr.12069. [DOI] [PMC free article] [PubMed] [Google Scholar]
  82. Emu B, Sinclair E, Favre D. et al. Phenotypic, functional, and kinetic parameters associated with apparent T-cell control of human immunodeficiency virus replication in individuals with and without antiretroviral treatment. J Virol. 2005;79(22):14169–14178. doi: 10.1128/JVI.79.22.14169-14178.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  83. Pereyra F, Addo MM, Kaufmann DE. et al. Genetic and immunologic heterogeneity among persons who control HIV infection in the absence of therapy. J Infect Dis. 2008;197(4):563–571. doi: 10.1086/526786. [DOI] [PubMed] [Google Scholar]
  84. Zimmerli SC, Harari A, Cellerai C. et al. HIV-1-specific IFN-gamma/IL-2-secreting CD8 T cells support CD4-independent proliferation of HIV-1-specific CD8 T cells. Proc Natl Acad Sci U S A. 2005;102(20):7239–7244. doi: 10.1073/pnas.0502393102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  85. Mudd PA, Ericsen AJ, Price AA. et al. Reduction of CD4+ T cells in vivo does not affect virus load in macaque elite controllers. J Virol. 2011;85(14):7454–7459. doi: 10.1128/JVI.00738-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  86. Chen H, Li C, Huang J. et al. CD4+ T cells from elite controllers resist HIV-1 infection by selective upregulation of p21. J Clin Invest. 2011;121(4):1549–1560. doi: 10.1172/JCI44539. [DOI] [PMC free article] [PubMed] [Google Scholar]
  87. Saez-Cirion A, Hamimi C, Bergamaschi A. et al. Restriction of HIV-1 replication in macrophages and CD4+ T cells from HIV controllers. Blood. 2011;118(4):955–964. doi: 10.1182/blood-2010-12-327106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  88. Yu XG, Lichterfeld M. Elite control of HIV: p21 (waf-1/cip-1) at its best. Cell Cycle. 2011;10(19):3213–3214. doi: 10.4161/cc.10.19.17051. [DOI] [PMC free article] [PubMed] [Google Scholar]
  89. Leng J, Ho HP, Buzon MJ. et al. A cell-intrinsic inhibitor of HIV-1 reverse transcription in CD4(+) T cells from elite controllers. Cell Host Microbe. 2014;15(6):717–728. doi: 10.1016/j.chom.2014.05.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  90. Walker WE, Kurscheid S, Joshi S. et al. Increased Levels of Macrophage Inflammatory Proteins Result in Resistance to R5-Tropic HIV-1 in a Subset of Elite Controllers. J Virol. 2015;89(10):5502–5514. doi: 10.1128/JVI.00118-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  91. Julg B, Pereyra F, Buzon MJ. et al. Infrequent recovery of HIV from but robust exogenous infection of activated CD4(+) T cells in HIV elite controllers. Clin Infect Dis. 2010;51(2):233–238. doi: 10.1086/653677. [DOI] [PMC free article] [PubMed] [Google Scholar]
  92. O'Connell KA, Rabi SA, Siliciano RF. et al. CD4+ T cells from elite suppressors are more susceptible to HIV-1 but produce fewer virions than cells from chronic progressors. Proc Natl Acad Sci U S A. 2011;108(37):E689–E698. doi: 10.1073/pnas.1108866108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  93. Rabi SA, O'Connell KA, Nikolaeva D. et al. Unstimulated primary CD4+ T cells from HIV-1-positive elite suppressors are fully susceptible to HIV-1 entry and productive infection. J Virol. 2011;85(2):979–986. doi: 10.1128/JVI.01721-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  94. Buzon MJ, Seiss K, Weiss R. et al. Inhibition of HIV-1 integration in ex vivo-infected CD4 T cells from elite controllers. J Virol. 2011;85(18):9646–9650. doi: 10.1128/JVI.05327-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  95. Zaunders JJ, Ip S, Munier ML. et al. Infection of CD127+ (interleukin-7 receptor+) CD4+ cells and overexpression of CTLA-4 are linked to loss of antigen-specific CD4 T cells during primary human immunodeficiency virus type 1 infection. J Virol. 2006;80(20):10162–10172. doi: 10.1128/JVI.00249-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  96. Kaufmann DE, Kavanagh DG, Pereyra F. 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):1246–1254. doi: 10.1038/ni1515. [DOI] [PubMed] [Google Scholar]
  97. Graf EH, Mexas AM, Yu JJ. et al. Elite suppressors harbor low levels of integrated HIV DNA and high levels of 2-LTR circular HIV DNA compared to HIV+ patients on and off HAART. PLoS Pathog. 2011;7(2):e1001300. doi: 10.1371/journal.ppat.1001300. [DOI] [PMC free article] [PubMed] [Google Scholar]
  98. O'Connell KA, Han Y, Williams TM. et al. Role of natural killer cells in a cohort of elite suppressors: low frequency of the protective KIR3DS1 allele and limited inhibition of human immunodeficiency virus type 1 replication in vitro. J Virol. 2009;83(10):5028–5034. doi: 10.1128/JVI.02551-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  99. Barker E, Martinson J, Brooks C. et al. Dysfunctional natural killer cells, in vivo, are governed by HIV viremia regardless of whether the infected individual is on antiretroviral therapy. AIDS. 2007;21(17):2363–2365. doi: 10.1097/QAD.0b013e3282f1d658. [DOI] [PubMed] [Google Scholar]
  100. Tomescu C, Duh FM, Hoh R. et al. Impact of protective killer inhibitory receptor/human leukocyte antigen genotypes on natural killer cell and T-cell function in HIV-1-infected controllers. AIDS. 2012;26(15):1869–1878. doi: 10.1097/QAD.0b013e32835861b0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  101. Marras F, Nicco E, Bozzano F. et al. Natural killer cells in HIV controller patients express an activated effector phenotype and do not up-regulate NKp44 on IL-2 stimulation. Proc Natl Acad Sci U S A. 2013;110(29):11970–11975. doi: 10.1073/pnas.1302090110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  102. Kiepiela P, Leslie AJ, Honeyborne I. et al. Dominant influence of HLA-B in mediating the potential co-evolution of HIV and HLA. Nature. 2004;432(7018):769–775. doi: 10.1038/nature03113. [DOI] [PubMed] [Google Scholar]
  103. Bailey JR, Williams TM, Siliciano RF. et al. Maintenance of viral suppression in HIV-1-infected HLA-B*57+ elite suppressors despite CTL escape mutations. J Exp Med. 2006;203(5):1357–1369. doi: 10.1084/jem.20052319. [DOI] [PMC free article] [PubMed] [Google Scholar]
  104. Migueles SA, Laborico AC, Imamichi H. et al. The differential ability of HLA B*5701+ long-term nonprogressors and progressors to restrict human immunodeficiency virus replication is not caused by loss of recognition of autologous viral gag sequences. J Virol. 2003;77(12):6889–6898. doi: 10.1128/JVI.77.12.6889-6898.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  105. Fellay J, Shianna KV, Ge D. et al. A whole-genome association study of major determinants for host control of HIV-1. Science. 2007;317(5840):944–947. doi: 10.1126/science.1143767. [DOI] [PMC free article] [PubMed] [Google Scholar]
  106. Fellay J, Ge D, Shianna KV. et al. Common genetic variation and the control of HIV-1 in humans. PLoS Genet. 2009;5(12):e1000791. doi: 10.1371/journal.pgen.1000791. [DOI] [PMC free article] [PubMed] [Google Scholar]
  107. Trachtenberg E, Bhattacharya T, Ladner M. et al. The HLA-B/-C haplotype block contains major determinants for host control of HIV. Genes Immun. 2009;10(8):673–677. doi: 10.1038/gene.2009.58. [DOI] [PMC free article] [PubMed] [Google Scholar]
  108. Han Y, Lai J, Barditch-Crovo P. et al. The role of protective HCP5 and HLA-C associated polymorphisms in the control of HIV-1 replication in a subset of elite suppressors. AIDS. 2008;22(4):541–544. doi: 10.1097/QAD.0b013e3282f470e4. [DOI] [PubMed] [Google Scholar]
  109. Tang Y, Huang S, Dunkley-Thompson J. et al. Correlates of spontaneous viral control among long-term survivors of perinatal HIV-1 infection expressing human leukocyte antigen-B57. AIDS. 2010;24(10):1425–1435. doi: 10.1097/QAD.0b013e32833a2b5b. [DOI] [PMC free article] [PubMed] [Google Scholar]
  110. Liu R, Paxton WA, Choe S. et al. Homozygous defect in HIV-1 coreceptor accounts for resistance of some multiply-exposed individuals to HIV-1 infection. Cell. 1996;86(3):367–377. doi: 10.1016/s0092-8674(00)80110-5. [DOI] [PubMed] [Google Scholar]
  111. Samson M, Libert F, Doranz BJ. et al. Resistance to HIV-1 infection in caucasian individuals bearing mutant alleles of the CCR-5 chemokine receptor gene. Nature. 1996;382(6593):722–725. doi: 10.1038/382722a0. [DOI] [PubMed] [Google Scholar]
  112. Rappaport J, Cho YY, Hendel H. et al. 32 bp CCR-5 gene deletion and resistance to fast progression in HIV-1 infected heterozygotes. Lancet. 1997;349(9056):922–923. doi: 10.1016/S0140-6736(05)62697-9. [DOI] [PubMed] [Google Scholar]
  113. van Manen D, van 't Wout AB, Schuitemaker H. Genome-wide association studies on HIV susceptibility, pathogenesis and pharmacogenomics. Retrovirology. 2012;9:70. doi: 10.1186/1742-4690-9-70. [DOI] [PMC free article] [PubMed] [Google Scholar]
  114. Granier C, Battivelli E, Lecuroux C. et al. Pressure from TRIM5alpha contributes to control of HIV-1 replication by individuals expressing protective HLA-B alleles. J Virol. 2013;87(18):10368–10380. doi: 10.1128/JVI.01313-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  115. Palacios JA, Perez-Pinar T, Toro C. et al. Long-term nonprogressor and elite controller patients who control viremia have a higher percentage of methylation in their HIV-1 proviral promoters than aviremic patients receiving highly active antiretroviral therapy. J Virol. 2012;86(23):13081–13084. doi: 10.1128/JVI.01741-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  116. Gornalusse GG, Mummidi S, Gaitan AA. et al. Epigenetic mechanisms, T-cell activation, and CCR5 genetics interact to regulate T-cell expression of CCR5, the major HIV-1 coreceptor. Proc Natl Acad Sci U S A. 2015;112(34):E4762–E4771. doi: 10.1073/pnas.1423228112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  117. Youngblood B, Noto A, Porichis F. et al. Cutting edge: Prolonged exposure to HIV reinforces a poised epigenetic program for PD-1 expression in virus-specific CD8 T cells. J Immunol. 2013;191(2):540–544. doi: 10.4049/jimmunol.1203161. [DOI] [PMC free article] [PubMed] [Google Scholar]
  118. Crowell TA, Hatano H. Clinical outcomes and antiretroviral therapy in 'elite' controllers: a review of the literature. J Virus Erad. 2015;1(2):72–77. doi: 10.1016/S2055-6640(20)30488-X. [DOI] [PMC free article] [PubMed] [Google Scholar]
  119. Langford SE, Ananworanich J, Cooper DA. Predictors of disease progression in HIV infection: a review. AIDS Res Ther. 2007;4:11. doi: 10.1186/1742-6405-4-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  120. Goujard C, Bonarek M, Meyer L. et al. CD4 cell count and HIV DNA level are independent predictors of disease progression after primary HIV type 1 infection in untreated patients. Clin Infect Dis. 2006;42(5):709–715. doi: 10.1086/500213. [DOI] [PubMed] [Google Scholar]
  121. Phillips AN, Lundgren JD. The CD4 lymphocyte count and risk of clinical progression. Curr Opin HIV AIDS. 2006;1(1):43–49. doi: 10.1097/01.COH.0000194106.12816.b1. [DOI] [PubMed] [Google Scholar]
  122. Touloumi G, Hatzakis A, Rosenberg PS. et al. Effects of age at seroconversion and baseline HIV RNA level on the loss of CD4+ cells among persons with hemophilia. Multicenter Hemophilia Cohort Study. AIDS. 1998;12(13):1691–1697. doi: 10.1097/00002030-199813000-00018. [DOI] [PubMed] [Google Scholar]
  123. Hubert JB, Burgard M, Dussaix E. et al. Natural history of serum HIV-1 RNA levels in 330 patients with a known date of infection. The SEROCO Study Group. AIDS. 2000;14(2):123–131. doi: 10.1097/00002030-200001280-00007. [DOI] [PubMed] [Google Scholar]
  124. Leon A, Perez I, Ruiz-Mateos E. et al. Rate and predictors of progression in elite and viremic HIV-1 controllers. AIDS. 2016;30(8):1209–1220. doi: 10.1097/QAD.0000000000001050. [DOI] [PubMed] [Google Scholar]
  125. Li JZ, Arnold KB, Lo J. et al. Differential levels of soluble inflammatory markers by human immunodeficiency virus controller status and demographics. Open Forum Infect Dis. 2015;2(1):ofu117. doi: 10.1093/ofid/ofu117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  126. Pereyra F, Lo J, Triant VA. et al. Increased coronary atherosclerosis and immune activation in HIV-1 elite controllers. AIDS. 2012;26(18):2409–2412. doi: 10.1097/QAD.0b013e32835a9950. [DOI] [PMC free article] [PubMed] [Google Scholar]
  127. Hsue PY, Hunt PW, Schnell A. et al. Role of viral replication, antiretroviral therapy, and immunodeficiency in HIV-associated atherosclerosis. AIDS. 2009;23(9):1059–1067. doi: 10.1097/QAD.0b013e32832b514b. [DOI] [PMC free article] [PubMed] [Google Scholar]
  128. Hunt PW, Landay AL, Sinclair E. et al. A low T regulatory cell response may contribute to both viral control and generalized immune activation in HIV controllers. PLoS One. 2011;6(1):e15924. doi: 10.1371/journal.pone.0015924. [DOI] [PMC free article] [PubMed] [Google Scholar]
  129. Krishnan S, Wilson EM, Sheikh V. et al. Evidence for innate immune system activation in HIV type 1-infected elite controllers. J Infect Dis. 2014;209(6):931–939. doi: 10.1093/infdis/jit581. [DOI] [PMC free article] [PubMed] [Google Scholar]
  130. BenMarzouk-Hidalgo OJ, Torres-Cornejo A, Gutierrez-Valencia A. et al. Differential effects of viremia and microbial translocation on immune activation in HIV-infected patients throughout ritonavir-boosted darunavir monotherapy. Medicine (Baltimore) 2015;94(17):e781. doi: 10.1097/MD.0000000000000781. [DOI] [PMC free article] [PubMed] [Google Scholar]
  131. Nwosu FC, Avershina E, Wilson R. et al. Gut Microbiota in HIV Infection: Implication for Disease Progression and Management. Gastroenterol Res Pract. 2014;2014:803185. doi: 10.1155/2014/803185. [DOI] [PMC free article] [PubMed] [Google Scholar]
  132. d'Ettorre G, Baroncelli S, Micci L. et al. Reconstitution of intestinal CD4 and Th17 T cells in antiretroviral therapy suppressed HIV-infected subjects: implication for residual immune activation from the results of a clinical trial. PLoS One. 2014;9(10):e109791. doi: 10.1371/journal.pone.0109791. [DOI] [PMC free article] [PubMed] [Google Scholar]
  133. Jiang W, Lederman MM, Hunt P. 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):1177–1185. doi: 10.1086/597476. [DOI] [PMC free article] [PubMed] [Google Scholar]
  134. Crowell TA, Ganesan A, Berry SA. et al. Hospitalizations among HIV controllers and persons with medically controlled HIV in the U.S. Military HIV Natural History Study. J Int AIDS Soc. 2016;19(1):20524. doi: 10.7448/IAS.19.1.20524. [DOI] [PMC free article] [PubMed] [Google Scholar]
  135. Crowell TA, Gebo KA, Blankson JN. et al. Hospitalization Rates and Reasons Among HIV Elite Controllers and Persons With Medically Controlled HIV Infection. J Infect Dis. 2015;211(11):1692–1702. doi: 10.1093/infdis/jiu809. [DOI] [PMC free article] [PubMed] [Google Scholar]
  136. Boufassa F, Lechenadec J, Meyer L. et al. Blunted response to combination antiretroviral therapy in HIV elite controllers: an international HIV controller collaboration. PLoS One. 2014;9(1):e85516. doi: 10.1371/journal.pone.0085516. [DOI] [PMC free article] [PubMed] [Google Scholar]
  137. Hatano H, Yukl SA, Ferre AL. et al. Prospective antiretroviral treatment of asymptomatic, HIV-1 infected controllers. PLoS Pathog. 2013;9(10):e1003691. doi: 10.1371/journal.ppat.1003691. [DOI] [PMC free article] [PubMed] [Google Scholar]
  138. Abraham L, Fackler OT. HIV-1 Nef: a multifaceted modulator of T cell receptor signaling. Cell Commun Signal. 2012;10(1):39. doi: 10.1186/1478-811X-10-39. [DOI] [PMC free article] [PubMed] [Google Scholar]
  139. Neri F, Giolo G, Potesta M. et al. The HIV-1 Nef protein has a dual role in T cell receptor signaling in infected CD4+ T lymphocytes. Virology. 2011;410(2):316–326. doi: 10.1016/j.virol.2010.11.018. [DOI] [PubMed] [Google Scholar]
  140. Rosa A, Chande A, Ziglio S. et al. HIV-1 Nef promotes infection by excluding SERINC5 from virion incorporation. Nature. 2015;526(7572):212–217. doi: 10.1038/nature15399. [DOI] [PMC free article] [PubMed] [Google Scholar]
  141. Skasko M, Tokarev A, Chen CC. et al. BST-2 is rapidly down-regulated from the cell surface by the HIV-1 protein Vpu: evidence for a post-ER mechanism of Vpu-action. Virology. 2011;411(1):65–77. doi: 10.1016/j.virol.2010.12.038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  142. Christodoulopoulos I, Droniou-Bonzom ME, Oldenburg JE. et al. Vpu-dependent block to incorporation of GaLV Env into lentiviral vectors. Retrovirology. 2010;7:4. doi: 10.1186/1742-4690-7-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  143. Yu X, Yu Y, Liu B. et al. Induction of APOBEC3G ubiquitination and degradation by an HIV-1 Vif-Cul5-SCF complex. Science. 2003;302(5647):1056–1060. doi: 10.1126/science.1089591. [DOI] [PubMed] [Google Scholar]
  144. Kane M, Yadav SS, Bitzegeio J. et al. MX2 is an interferon-induced inhibitor of HIV-1 infection. Nature. 2013;502(7472):563–566. doi: 10.1038/nature12653. [DOI] [PMC free article] [PubMed] [Google Scholar]
  145. Perez-Caballero D, Zang T, Ebrahimi A. et al. Tetherin inhibits HIV-1 release by directly tethering virions to cells. Cell. 2009;139(3):499–511. doi: 10.1016/j.cell.2009.08.039. [DOI] [PMC free article] [PubMed] [Google Scholar]
  146. Malhotra U, Holte S, Dutta S. et al. Role for HLA class II molecules in HIV-1 suppression and cellular immunity following antiretroviral treatment. J Clin Invest. 2001;107(4):505–517. doi: 10.1172/JCI11275. [DOI] [PMC free article] [PubMed] [Google Scholar]
  147. Chen Y, Winchester R, Korber B. et al. Influence of HLA alleles on the rate of progression of vertically transmitted HIV infection in children: association of several HLA-DR13 alleles with long-term survivorship and the potential association of HLA-A*2301 with rapid progression to AIDS. Long-Term Survivor Study. Hum Immunol. 1997;55(2):154–162. doi: 10.1016/s0198-8859(97)00092-x. [DOI] [PubMed] [Google Scholar]
  148. Limou S, Le Clerc S, Coulonges C. et al. Genomewide association study of an AIDS-nonprogression cohort emphasizes the role played by HLA genes (ANRS Genomewide Association Study 02). J Infect Dis. 2009;199(3):419–426. doi: 10.1086/596067. [DOI] [PubMed] [Google Scholar]
  149. Ferre AL, Hunt PW, McConnell DH. et al. HIV controllers with HLA-DRB1*13 and HLA-DQB1*06 alleles have strong, polyfunctional mucosal CD4+ T-cell responses. J Virol. 2010;84(21):11020–11029. doi: 10.1128/JVI.00980-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  150. Gao X, Bashirova A, Iversen AK. et al. AIDS restriction HLA allotypes target distinct intervals of HIV-1 pathogenesis. Nat Med. 2005;11(12):1290–1292. doi: 10.1038/nm1333. [DOI] [PubMed] [Google Scholar]
  151. McLaren PJ, Coulonges C, Bartha I. et al. Polymorphisms of large effect explain the majority of the host genetic contribution to variation of HIV-1 virus load. Proc Natl Acad Sci U S A. 2015;112(47):14658–14663. doi: 10.1073/pnas.1514867112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  152. Lucero C, Torres B, Leon A. et al. Rate and predictors of non-AIDS events in a cohort of HIV-infected patients with a CD4 T cell count above 500 cells/mm(3). AIDS Res Hum Retroviruses. 2013;29(8):1161–1167. doi: 10.1089/aid.2012.0367. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from The Yale Journal of Biology and Medicine are provided here courtesy of Yale Journal of Biology and Medicine

RESOURCES