Skip to main content
Journal of Virology logoLink to Journal of Virology
. 2015 Aug 12;89(21):10735–10747. doi: 10.1128/JVI.01527-15

The Breadth of Expandable Memory CD8+ T Cells Inversely Correlates with Residual Viral Loads in HIV Elite Controllers

Zaza M Ndhlovu a,d, Eleni Stampouloglou a, Kevin Cesa a, Orestes Mavrothalassitis a, Donna Marie Alvino a, Jonathan Z Li c, Shannon Wilton a, Daniel Karel a, Alicja Piechocka-Trocha a, Huabiao Chen e, Florencia Pereyra c, Bruce D Walker a,b,d,
Editor: G Silvestri
PMCID: PMC4621138  PMID: 26269189

ABSTRACT

Previous studies have shown that elite controllers with minimal effector T cell responses harbor a low-frequency, readily expandable, highly functional, and broadly directed memory population. Here, we interrogated the in vivo relevance of this cell population by investigating whether the breadth of expandable memory responses is associated with the magnitude of residual viremia in individuals achieving durable suppression of HIV infection. HIV-specific memory CD8+ T cells were expanded by using autologous epitopic and variant peptides. Viral load was measured by an ultrasensitive single-copy PCR assay. Following expansion, controllers showed a greater increase in the overall breadth of Gag responses than did untreated progressors (P = 0.01) as well as treated progressors (P = 0.0003). Nef- and Env-specific memory cells expanded poorly for all groups, and their expanded breadths were indistinguishable among groups (P = 0.9 for Nef as determined by a Kruskal-Wallis test; P = 0.6 for Env as determined by a Kruskal-Wallis test). More importantly, we show that the breadth of expandable, previously undetectable Gag-specific responses was inversely correlated with residual viral load (r = −0.6; P = 0.009). Together, these data reveal a direct link between the abundance of Gag-specific expandable memory responses and prolonged maintenance of low-level viremia. Our studies highlight a CD8+ T cell feature that would be desirable in a vaccine-induced T cell response.

IMPORTANCE Many studies have shown that the rare ability of some individuals to control HIV infection in the absence of antiretroviral therapy appears to be heavily dependent upon special HIV-specific killer T lymphocytes that are able to inhibit viral replication. The identification of key features of these immune cells has the potential to inform rational HIV vaccine design. This study shows that a special subset of killer lymphocytes, known as central memory CD8+ T lymphocytes, is at least partially involved in the durable control of HIV replication. HIV controllers maintain a large proportion of Gag-specific expandable memory CD8+ T cells involved in ongoing viral suppression. These data suggest that induction of this cell subset by future HIV vaccines may be important for narrowing possible routes of rapid escape from vaccine-induced CD8+ T cell responses.

INTRODUCTION

Most human immunodeficiency virus (HIV)-infected individuals have continuous viral replication and, if left untreated, eventually progress to AIDS (14). Only a very small group of infected individuals, referred to as elite controllers (EC) or elite suppressors, achieves spontaneous control of viral replication for prolonged periods in the absence of treatment (58). This remarkable control of viral replication among elite controllers is believed to be mediated largely by major histocompatibility complex (MHC) class I-restricted CD8+ T cell responses (913). These individuals therefore present a unique model for understanding the in vivo mechanisms of T cell-mediated immune control (14).

Most studies examining the relationship between CD8+ T cell responses and viral load in elite controllers have focused on assays that measure effector memory rather than central memory responses. Using gamma interferon (IFN-γ) enzyme-linked immunosorbent spot (ELISPOT) assays, the most frequent and robust CD8+ T cell responses in elite controllers are directed toward the HIV Gag protein; particularly, p24 capsid protein targeting has been repeatedly shown to be associated with enhanced control of viremia in vivo (15) and in vitro (16). In contrast, preferential targeting of the HIV envelope (Env) protein has been associated with higher viral loads in both human and monkey studies (1720). Although the precise mechanisms responsible for the enhanced antiviral function associated with Gag-specific responses are not fully understood, fitness costs associated with escape mutations from CD8+ T cell responses directed at the highly conserved Gag protein have been implicated in both humans infected with HIV and primates infected with simian immunodeficiency virus (SIV) (2127).

Despite extensive evidence supporting a specific role for CD8+ T cells in immune-mediated control of HIV, not all elite controllers exhibit readily detectable CD8+ T cell responses. Measurements of CD8+ T cell responses by ex vivo cytokine secretion assays fail to accurately measure central memory responses (18). Consequently, the role of this cell subset in the immune-mediated control of HIV remains ill defined. A recent analysis of HIV-specific CD8+ T cells following ex vivo enrichment and after expansion in culture defined the phenotype and functional features of HIV-specific central memory CD8+ T cells (28). These studies show that in addition to the readily detectable responses, most elite controllers harbor a wide range of low-frequency but highly functional and readily expandable Gag-specific memory cells, which are able to inhibit virus replication in vitro (28). However, it is not known whether this population contributes to durable viral suppression. Moreover, it is not known if this is a property that is limited to responses targeting the Gag protein or whether expandable central memory responses to other HIV protein targets are also involved.

In this study, we interrogated the in vivo relevance of the expandable memory population based on the premise that there are two possibilities for the role of this population in HIV elite controllers. They may be directly responsible for ongoing active suppression of the virus, or they could be footprints of immune responses to epitopes that have escaped or were completely suppressed. We measured the breadth, specificity, and functional characteristics of expandable memory cells in elite controllers and chronic progressors (CP). We also investigated whether there was a relationship between the breadth of the expandable responses and viral load. Our data demonstrate that expandable responses in HIV controllers are directed predominantly against the HIV Gag protein and show a link between the abundance of expandable responses to epitope variants and durable virus suppression. Importantly, we show an inverse relationship between the breadth of Gag-specific expandable responses and the level of plasma viremia. These data shed light on immune responses that are most associated with sustained viral control and therefore desirable to induce through HIV vaccination.

MATERIALS AND METHODS

Study subjects.

HIV-infected individuals were recruited from outpatient clinics at Massachusetts General Hospital and affiliated Boston-area hospitals. The respective institutional review boards approved this study, and all subjects gave written informed consent. A total of 16 EC, 2 viremic controllers (VC), 13 untreated CP, and 15 treated CP were studied. Detailed definitions of controllers and chronic progressors were described previously (29). In brief, EC were defined as having plasma HIV RNA levels of <50 copies/ml in the absence of antiretroviral therapy, on at least three determinations over at least a year of follow-up. VC had detectable HIV-1 RNA levels of <2,000 copies/ml. CP were defined as subjects having untreated HIV infection for >1 year with plasma viral loads of >2,000 copies/ml for at least 1 year of follow-up; treated (antiretroviral therapy [ART]) chronic progressors had HIV RNA levels below the limit of detection for the respective available standard assays (e.g., <75 RNA copies/ml by branched DNA assay or <50 copies by PCR). All subjects selected for this study had absolute CD4+ T cell counts of >400 cells/mm3. All experiments were performed on cryopreserved peripheral blood mononuclear cells (PBMCs).

IFN-γ ELISPOT assays.

Gamma interferon (IFN-γ) enzyme-linked immunospot (ELISPOT) assays were performed as described previously (28), using a final concentration of 100 ng/ml (20 ng of the peptide mix in a 200-μl volume) of overlapping peptides (OLPs) and incubation overnight. We used OLPs corresponding to the HIV clade B sequence from 2001 (17). OLPs averaged 18 amino acids in length, overlapped by 10 amino acids, and spanned the entire HIV Gag, Nef, and Env proteins. The number of input cells ranged from 50,000 to 100,000 cells per well, depending on cell availability. The number of specific spot-forming cells (SFC) was calculated by subtracting the number of spots in the negative-control wells from the number of spots in each experimental well. A positive response was defined as a well having at least three times the mean number of SFC in the three negative-control wells. Wells with positive responses also had to have at least 50 SFC/106 PBMCs (28). The magnitude of the epitope-specific response was reported as the number of SFC per million PBMCs.

Cultured IFN-γ ELISPOT assays.

Peptide-stimulated cells were cultured at 37°C with 5% CO2 for 12 days in RPMI medium containing 10% heat-inactivated fetal calf serum (R10 medium) and in R10 medium supplemented with 50 U/ml of recombinant human interleukin-2 (IL-2) (R10/50 medium). A 100-ng/ml final peptide concentration was used for cultured stimulation because it was determined to be the optimal concentration at which pooled overlapping HIV peptides stimulated the greatest number of responses (28). Cultures were supplemented with fresh R10/50 medium at 3-day intervals or as needed. On day 12, cells were washed three times with fresh R10 medium and rested at 37°C with 5% CO2 overnight in fresh R10 medium. The cells were then retested against OLPs spanning HIV Gag, Nef, and Env proteins in an overnight ELISPOT assay, as described above.

Flow cytometry.

After 12 days of culture stimulation, cells were rested overnight in R10 medium and then restimulated with Gag OLPs at a 20-ng/ml final concentration or optimal peptides for 1 h at 37°C with 5% CO2. After 1 h, 10 ng/ml of brefeldin A (Sigma-Aldrich, St. Louis MO, USA) was added, and the cells were incubated for another 5 h. At the end of the stimulation period, intracellular cytokine staining (ICS) was performed according to the BD Biosciences ICS protocol. Briefly, cells were first stained with dead cell dye for 10 min and then washed and surface stained with anti-CD3, -CD4, and -CD8 and exclusion channel antibodies (CD14, CD19, and CD56). The cells were then fixed and permeabilized with Cytofix/Cytoperm solution and stained with anti-IFN-γ antibody (BD Biosciences, San Jose, CA, USA). Following staining, the cells were resuspended in phosphate-buffered saline (PBS) containing 2% paraformaldehyde. The cells were acquired on a BD LSRFortessa cytometer (BD Biosciences, San Jose, CA, USA). Flow cytometry data were analyzed with the FlowJo software package (Treestar, Ashland, OR).

Virus inhibition assay.

The ability of CD8+ T cells to inhibit virus replication in autologous primary CD4+ T cells was assessed by measuring p24 antigen production, as described previously, with modifications (16). Briefly, primary CD4+ and CD8+ T cells were isolated by using CD4 MicroBeads and CD8 MicroBeads, respectively (Miltenyi Biotech, San Diego, CA, USA). Enriched CD8+ T cells were cultured for 9 days with Gag, Nef, or Env OLP pools. IFN-γ ICS was used to assess the frequency of expandable cells specific for each of the three HIV genes. CD4+ T cell targets were prepared by stimulating the magnetic bead-enriched CD4+ T cells with a CD3/CD8-bispecific antibody and infecting the cells on day 3 with the NL4-3 laboratory-adapted HIV strain at a multiplicity of infection (MOI) of 0.001 for 4 h at 37°C (30). The virus-infected CD4+ T cells were then incubated in the presence or absence of expanded CD8+ T cells at an adjusted effector-to-target-cell ratio of 1:1. The number of input CD8+ T cells for each cell culture was adjusted based on the frequency of IFN-γ-secreting cells, such that equivalent numbers of HIV-specific CD8+ T cells were added to each culture. The cultures were fed at regular intervals by removing and replacing one half of the culture supernatant with fresh medium. Supernatants harvested at days 3, 5, and 7 were cryopreserved for later p24 antigen quantification by an enzyme-linked immunosorbent assay (ELISA) (PerkinElmer, Boston, MA). Log inhibition values were calculated by subtracting log10 p24 values for cultures with CD8+ T cells from log10 p24 values for cultures without CD8+ T cells at day 7.

Determination of HIV-1 RNA levels by single-copy assays.

To accurately quantify low-level viremia in elite controllers and in patients receiving antiretroviral therapy, we used a highly sensitive quantitative real-time reverse transcriptase (RT)-initiated PCR (RT-PCR) assay that detects and quantifies HIV-1 RNA levels down to 1 copy/ml. The assay was performed as described previously (31). Briefly, the assay was performed in three steps: RNA extraction, reverse transcription, and quantitative real-time PCR. A key internal control aimed at monitoring the recovery of HIV-1 from plasma samples involved the spiking of plasma samples with a known transcript copy of RNA isolated from an avian sarcoma leukosis retroviral vector (RCAS). Seven milliliters of plasma from each subject was mixed with 200 μl of the RCAS stock (300,000 copies of RNA) before RNA extraction and quantification by RT-PCR. PCR amplification of the HIV Gag region was performed on either plasma or PBMC samples from each subject, as previously described (31).

Real-time PCR plate setup.

For each real-time PCR run, two standard curves were generated, one for HIV and the other for RCAS. To generate the standard curves, a prequantified HIV RNA stock serially diluted to between 1 million copies and 0.3 copies/10 ml was used. Similarly, RCAS standards were generated in duplicate by using a serially diluted RCAS RNA stock. To ensure that there was no contamination of the PCR reagents, a template control was tested in duplicate for both RCAS and HIV primers/probes with no plasma samples added (31). For each specimen, three replicate reactions were performed for HIV quantification. The number of copies of HIV was derived from the calculated number of copy equivalents per reaction mixture and was expressed as the number of copies per milliliter of the starting plasma sample.

Assay reproducibility and limit of quantitation.

The assay was validated by running 6 independent real-time PCRs in duplicate, using previously determined HIV-1 RNA amounts ranging from 100,000 to 0.4 copies per reaction mixture. Linear regression analyses revealed that the assay was sensitive enough to detect as little as a single copy of HIV-1 RNA in the linear dynamic range of between 0.4 copies and 100,000 HIV RNA copies per reaction mixture (R2 = 0.94). The assay also showed excellent agreement between the expected values and the measured values for 6 independent serial dilutions (run in duplicate) of the control HIV RNA transcript (R2 = 0.96).

Statistical analyses.

Spearman rank correlation simple linear regression and Mann-Whitney tests were performed by using GraphPad Prism version 5.0b. All tests were two tailed, and P values of <0.05 were considered significant.

RESULTS

We studied 46 subjects chronically infected with HIV, including 16 HIV EC, 2 VC, 13 CP, and 15 individuals treated with ART. Table 1 provides information on age, sex, absolute CD4 cell counts, and virus loads at the time when samples were tested for HIV-specific immune responses. Table 2 shows HLA class I alleles of the study participants and indicates the experiments performed on samples from each subject.

TABLE 1.

Baseline characteristics of study participants

Patient characteristica Value for group (n = 46)
Controllers (EC, n = 16; VC, n = 2) Progressors (CP, n = 13) Treated subjects (ART, n = 15)
No. (%) of patients of gender
    Male 16 (88.9) 12 (92.3) 12 (80)
    Female 4 (11.1) 1 (7.7) 3 (20)
Age (yr)
    Median 56 50 50.5
    Q1–Q3 46–73 38–64 38–64
No. of CD4+ T cells/mm3
    Median 733 559 508.5
    Q1–Q3 486–1,786 436–1,320 443.3–1,028
Log10 HIV copies/ml determined by a standard assay
    Median 1.7 3.8 1.7
    Q1–Q3 1.7–2.4 3.6–4.6 1.7–3.5
a

Q1–Q3 denotes interquartile ranges for the parameters indicated.

TABLE 2.

Study participants, class I HLA alleles, and experimentsa

Subject HLA class I allele
Subject used for exptb
HLA-A1 HLA-A2 HLA-B1 HLA-B2 HLA-C1 HLA-C2 Cultured ELISPOT SCA VIA TW10, T3N, G9D responses
Elite controllers
    586181 0201 0302 0801 5701 0602 0701
    108896 0201 2402 0801 4001 0304 0304
    835698 0201 3001 1302 5701 0602 0602
    255675 0201 0201 2705 5701 0102 0602
    540772 0101 6801 5501 5701 0303 0602
    731849 0101 7400 3701 8100 0602 1800
    701554 1101 3201 3501 5001 0401 0602
    194133 0201 3004 3901 4101 1203 1700
    382086 3001 3201 1302 4001 0304 0602
    849151 0101 2601 2705 5701 0202 0602
    473516 0101 3402 0801 5701 0701 0602
    595424 0201 1101 1402 5101 0802 1402
    555477 0301 2601 1401 5701 0602 0802
    275432 0103 3201 3508 7301 0401 1505
    553064 0201 0301 1501 2705 0102 0304
    847733 0201 2301 4403 5106 1601 1601
    164007 0101 0301 5701 4102 0602 1202
    831969 0301 2601 4501 5701 0602 0602
Chronic progressors
    387879 2902 3002 0702 3501 0401 1505
    950005 0101 3101 4001 5801 0302 0304
    690641 0201 3303 1302 2705 0401 0202
    185075 0101 0201 4402 5701 0102 0602
    805181 0201 0201 0702 4403 0401 0702
    667335 0201 2902 4403 4403 1601 1601
    156261 0101 0206 1501 5701 0304 0602
    703459 2902 2902 4403 4501 0602 1601
    743887 0201 0301 1801 3501 0401 0701
    930133 0101 2402 1302 5701 0602 0602
    705357 3301 3601 1402 5301 0401 0802
    930024 0101 6801 0801 5701 0602 0701
    388555 0101 3201 4402 5701 0501 0602
ARV treated
    930213 0201 0201 4001 4001 0304 0501
    453230 0301 3601 3502 5703 0401 0701
    508473 0217 2902 4403 5101 1502 1601
    540319 0101 6801 1517 2705 0102 0702
    289151 0101 0201 2705 5101 0102 1502
    985170 3001 3303 4201 4201 1700 1700
    516917 0101 0101 0801 1501 0401 0701
    672068 0301 1101 4001 4402 0304 0501
    409231 0101 0201 0801 5101 0701 1502
    672068 0301 0110 4001 4402 0304 0501
    608520 3101 7400 3501 5701 0602 1601
    350103 0201 0301 1501 5701 0401 0602
    898049 0201 0301 4102 5701 0602 1700
    922403 0101 0301 0702 5701 0602 0702
    403998 0101 0201 4402 5701 0602 1203
a

Shown is a list of all subjects studied, including class I HLA information and the all the experiments performed. SCA, single copy PCR assay; VIA, virus inhibition assay.

b

✓ indicates that the subject was used for the indicated experiment.

Expandable memory CD8+ T cells from elite controllers preferentially target Gag rather than Env and Nef.

Given that EC maintain prolonged control of HIV viremia, it is important to understand the immune responses that are associated with this equilibrium. To determine the potential contribution of expandable responses to various epitopic regions in chronically infected individuals, we examined HIV protein targeting and the breadth of the expandable responses. Our analysis focused on memory responses to three HIV proteins: Gag, Nef, and Env. We chose these three HIV proteins for the following reasons: (i) T cell responses directed to the HIV Gag protein have consistently been associated with lower-level viremia (17, 3235); (ii) Nef responses are immunodominant early during acute HIV infection and may be involved in the initial control of viremia, and therefore, their persistence may be relevant to long-term viral control (3639); and (iii) responses targeting the Env region have been shown to be important for both T cell and B cell immune responses (4042), and the breadth of Env-specific responses has been positively associated with viral load (17, 43). Thus, elucidating the role of long-lived responses to these HIV proteins may be relevant for vaccines.

We first carried out a comparative analysis of expandable responses in three subject groups using cultured ELISPOTs. Overlapping peptides (OLPs) were used to screen for HIV-specific T cell responses. The peptides spanned HIV Gag, Nef, and Env proteins and were based on the consensus clade B sequence. The peptides were 15 to 20 amino acids in length, overlapping by 10 amino acids. PBMCs were initially cultured for 12 days in the presence of OLPs. An IFN-γ ELISPOT assay was then performed on the cultured cells. To investigate whether there were quantitative differences in the frequencies of expandable responses directed against the three HIV proteins, we compared the breadths of Gag, Nef, and Env responses in cultures. Breadth of response in this study is defined as the sum of IFN-γ-positive OLP responses for the entire HIV protein. Although the breadths of ex vivo Gag responses determined by IFN-γ ELISPOT assays were comparable between HIV controllers and progressors (P = 0.28, as determined by a Kruskal-Wallis test) (Fig. 1A), upon peptide stimulation, controllers showed a greater breadth of Gag responses than did untreated progressors (P = 0.01) as well as treated progressors (P = 0.0003) (Fig. 1B). Furthermore, stimulated expansion of controller PBMCs with Gag peptides resulted in a significantly larger number of responses that were not detectable before expansion (new responses) than those of untreated progressors (P = 0.03, as determined by a Kruskal-Wallis test) (Fig. 1C). The breadths of ex vivo Nef responses were indistinguishable among the groups (P = 0.1, as determined by a Kruskal-Wallis test) (Fig. 1D), and the responses expanded poorly upon culture stimulation in all groups (P = 0.9 for Nef, as determined by a Kruskal-Wallis test) (Fig. 1E). Similarly, ex vivo Env responses (Fig. 1G) and expanded Env responses (Fig. 1H) were indistinguishable among the groups (P = 0.07 for ex vivo Env, as determined by a Kruskal-Wallis test; P = 0.6 for expanded Env, as determined by a Kruskal-Wallis test). The breadth of expanded responses that were not detectable ex vivo (new responses) was small for both Nef and Env stimulations (Fig. 1F and I) for all groups. Together, these data demonstrate that the majority of the HIV-specific expandable memory pool maintained by HIV controllers is directed predominantly against Gag.

FIG 1.

FIG 1

Expandable memory responses in elite controllers are directed predominantly toward the HIV Gag protein. (A and B) Gag-specific responses of controllers and treated and untreated progressors measured ex vivo (A) and in cultures (B). (C) Gag-specific responses, undetectable at baseline, become detectable following stimulation of cultures. (D and E) Nef-specific responses in controllers and treated and untreated progressors measured ex vivo (D) and in cultures (E). (F) Nef-specific responses are undetectable at baseline but become detectable following stimulation of cultures. (G and H) Env-specific responses in controllers and treated and untreated progressors measured ex vivo (G) and in cultures (H). (I) Env responses are low at baseline but increase following stimulation of cultures. For all panels, horizontal bars denote mean values. P values were calculated by using the two-tailed Mann-Whitney test and the Kruskal-Wallis one-way analysis of variance.

Expandable responses of HIV controllers to Gag retain a superior virus-inhibitory capacity compared to those for Env and Nef.

Previous studies have shown that qualitative features of host CD8+ T cell responses are more strongly associated with immune-mediated control of HIV replication than quantitative parameters (10, 4447). We therefore compared the virus inhibition capacities of memory CD8+ T cells directed against the three HIV proteins. We chose to examine this function because we had previously shown that Gag-specific central memory cells preferentially retain the capacity to inhibit virus replication upon stimulated expansion with overlapping Gag peptides (28). We directly compared the inhibitory capacities of Gag-, Nef-, and Env-specific expanded CD8+ T cells by measuring their ability to inhibit HIV replication in autologous CD4 T cells infected in vitro. We selected donors with no detectable inhibitory activity before culture (data not shown). Representative data for one elite controller (Fig. 2A) show that CD8+ T cells that expanded following HIV Gag, Nef, and Env stimulations resulted in a reduction in p24 antigen production over a 7-day culture period, with the most effective suppression being shown by Gag-specific CD8+ T cells. We next compared the inhibitory capacities of expandable responses in 6 elite controllers and 3 treated and 3 untreated chronic progressors. Subjects with at least five expandable responses to Gag, Nef, and Env were selected for these studies. On a per-cell basis, Gag responses of HIV controllers had a significantly greater inhibition capacity than did those of progressors (P = 0.009) (Fig. 2B), whereas no significant differences were observed for Nef responses (P = 0.1) (Fig. 2C) or Env responses (P = 0.2) (Fig. 2D) among groups. The expansion data and the virus inhibition data collectively suggest that HIV controllers have a greater capacity to maintain a larger breadth of functionally superior expandable responses directed against HIV Gag.

FIG 2.

FIG 2

Virus inhibition of NL4-3-infected autologous CD4+ T cells by memory responses. (A) NL4-3-infected CD4+ T cells were cultured at an adjusted ratio of 1:1 with unstimulated PBMCs or PBMCs stimulated with HIV Gag, Nef, or Env peptide pools. (The adjusted ratio is described in Materials and Methods.) Control conditions included infected CD4+ T cells alone (positive control) and uninfected CD4+ T cells (negative control). Log10 differences in p24 values between CD4+ T cells alone and those cocultured with CD8+ T cells at days 3, 5, and 7 for a representative elite controller are displayed. (B to D) Virus inhibition data for 6 elite controllers and 6 progressors, reported as fold reductions in log10 p24 antigen concentrations in supernatants from Gag (B)-, Nef (D)-, and Env (E)-expanded CD8+ T cells cocultured with NL4-3-infected autologous CD4+ T cells. Data are calculated as fold reductions in log10 p24 antigen concentrations at day 7.

Correlation between the breadth of HIV-specific memory CD8+ T cell responses and HIV plasma viral load.

In order to ascertain the in vivo relevance of expandable memory responses, we next set out to examine the relationship between the breadth of this cell population and viral load, which is a well-defined prognostic marker of HIV disease progression and has been associated with CD4+ T cell loss in elite controllers (4850). We used an internally controlled real-time reverse transcriptase PCR assay that quantifies the HIV RNA concentration down to 1 copy per ml of plasma to accurately quantify viral loads in elite controllers and in progressors whose plasma HIV RNA levels are suppressed to below 75 copies/ml. We first used the single-copy assay to measure HIV RNA levels in plasma samples obtained from elite controllers and ART-suppressed HIV-infected patients. Our analysis revealed comparable viral load levels between elite controllers and ART-suppressed subjects (P = 0.11, as determined by a Mann-Whitney test) (Fig. 3A). Two treated progressors had 100 copies/ml of HIV RNA as determined by the single-copy assay, even though they had levels that were below the limit of detection according to standard viral load measurements, highlighting the greater sensitivity of this assay. We next investigated if residual viremia influences HIV disease progression. Here, residual viremia refers to low-level viremia that can be detected by the ultrasensitive single-copy viral load assay but not standard viral load assays. Interestingly, the residual plasma viral load in controllers was inversely correlated with the breadth of new (absent before expansion) Gag memory responses (Spearmen's r = −0.6; P = 0.009) (Fig. 3C), whereas ex vivo responses (Spearman's r = −0.02; P = 0.9) (Fig. 3B) and total expanded responses (data not shown) did not. Neither ex vivo nor expanded (new) Nef responses (Spearman's r = 0.4 and P = 0.2 for ex vivo responses; Spearman's r = 0.3 and P = 0.3 for expanded responses) correlated with viral load (Fig. 3D and E). Similarly, Env responses (Spearman's r = −0.1 and P = 0.7 for ex vivo responses; Spearman's r = −0.07 and P = 0.8 for expanded responses) did not correlate with residual viral load (Fig. 3F and G). These data show an association between the breadth of new Gag-specific (previously undetectable) responses and viral load. We next carried out similar analyses for ART-suppressed subjects, but we were unable to detect significant correlations for either ex vivo responses (Spearman's r = −0.1; P = 0.7) (Fig. 3H) or expanded (new) Env responses (Spearman's r = 0.1; P = 0.7) (Fig. 3I). Collectively, our data demonstrate a strong association between a larger breadth of the Gag-specific memory pool and effective viral suppression in the absence of antiretroviral therapy. These data also support a model in which very-low-frequency but highly expandable HIV-specific memory cells may be directly involved in sustained viral suppression in vivo and that broad Gag-specific memory responses are important for sustained HIV suppression.

FIG 3.

FIG 3

The breadth of Gag-specific memory responses is inversely correlated with residual plasma viral load in HIV elite controllers. (A) Plasma viral RNA copy numbers measured by a single-copy PCR assay for elite controllers and ART-suppressed chronic progressors whose HIV RNA levels were below the limit of detection by a standard viral load (VL) assay. (B and C) Elite controller plasma viral loads measured by a single-copy assay, plotted against the breadth of ex vivo Gag-specific responses (B) or expandable, previously undetectable (new) Gag-specific responses (C). (D and E) Elite controller plasma viral loads measured by a single-copy assay, plotted against the breadth of ex vivo Nef-specific responses (D) or new Nef-specific responses in cultures (E). (F and G) Elite controller plasma viral loads measured by a single-copy assay, plotted against the breadth of ex vivo Env-specific responses (F) or (new) Env-specific responses in cultures (G). (H and I) Plasma viral loads of ART-suppressed subjects measured by a single-copy assay, plotted against the breadth of ex vivo Gag-specific responses (H) or new Gag-specific responses in cultures (I). The correlation coefficient (r) values and P values were determined by the Spearman rank correlation test.

HIV controllers maintain a large pool of epitope variant (type)-specific expandable memory CD8+ T cells directed against in vivo-occurring viral variants.

Previous studies have demonstrated that accumulation of viral escape mutants contributes to higher viral loads during chronic AIDS virus infection (5155). In addition, the ability to mount de novo CD8+ T cell responses with sufficient cross-reactivity to potential escape variants may be one mechanism by which elite controllers maintain lower viral loads despite ongoing viral evolution (47, 56). We hypothesized that maintenance of a greater breadth of memory responses against escape variants might also contribute to sustained suppression of HIV. We addressed this issue by determining whether EC maintain immune responses to a wide range of epitope variants known to arise in vivo. To simplify the analysis, we focused on a well-characterized protective Gag240–249 TW10 epitope and two of its in vivo variants (57). We chose the TW10 epitope because the T242N (T3N) mutation in this epitope is the most frequently detected mutation, which occurs rapidly after acute infection in HLA-B*57/B*58-positive persons (58). The T3N mutation is also associated with viral replicative fitness cost, and the arising variants can be targeted by specific immune responses (26, 59).

We performed cultured expansions of HIV-specific CD8+ T cells using the wild-type (WT) TW10 epitope as well as the T3N and G248D (G9D) variant epitopes. The no-peptide-stimulation culture served as the negative control. Sixteen HLA-B*5701 donors, comprised of 6 elite controllers, 5 chronic progressors, and 5 treated progressors, were used for these studies (Table 2). To determine the specificity of the expanded memory CD8+ T cells, we tested the reactivity of each expanded population against the peptide used to generate the cell line as well as against the other two TW10 variant peptides. Culture stimulation of PBMCs from one elite controller (known to have the G9D mutation in plasma viruses) in the presence of TW10 peptide variants resulted in a significant expansion of the G9D-, WT-, and T3N-specific cells populations (Fig. 4A). In contrast, a chronically infected untreated B*5701 donor with the T3N mutation in plasma viruses expanded only T3N-reactive cells (Fig. 4B). Interestingly, each expanded memory cell population was more strongly reactive to the peptide used for its expansion (Fig. 4A and B). Figure 4C to E show summary data for all 16 donors studied. Figure 4C shows data for 6 elite controllers. PBMCs were expanded with the TW10 (Fig. 4C, left), T3N (middle), and G9D (right) epitopes and tested for reactivity to the three variant peptides. The data show that elite controllers maintained significantly higher frequencies of memory CD8+ T cells with the wild-type TW10 and G9D variant epitopes. Figure 4D shows data from a similar analysis for untreated chronic progressors. The data reveal that memory responses of chronic progressors were narrowly directed to T3N (Fig. 4D, middle). Figure 4E shows that ART-suppressed subjects also maintained only T3N memory responses. Overall, these data show that elite controllers maintain larger pools of expandable memory responses to epitope variants, at least in the context of HLA-B*5701, than do chronic progressors.

FIG 4.

FIG 4

Elite controllers maintain a large pool of expandable memory responses specific for the TW10 epitope and its in vivo-occurring variants. PBMCs from HLA-B*57 donors were stimulated with either wild-type TW10 or variant peptides. Each expanded population was then tested for reactivity to other variants by using IFN-γ ICS. (A) Representative flow cytometry plot for one elite controller. Numbers in the gates represent the proportions of IFN-γ-secreting CD8+ T cells. (B) Representative flow cytometry plot for a chronic progressor. (C to E) Summary data for 6 controllers (C), 5 progressors (D), and 5 ART-suppressed subjects (E). Data show how each population expands when a specific epitope cross-reacts with variant epitopes. ns, not significant.

DISCUSSION

This study confirms and extends data from a previous study showing that maintenance of a greater breadth of central memory T cell (Tcm) responses targeting the HIV Gag protein is associated with durable immune-mediated control in the setting of natural HIV infection. These data support the hypothesis that non-Gag-specific CD8+ T cell responses, particularly Nef- and Env-specific responses, contribute less to durable HIV suppression. Although the importance of Gag-specific CD8+ T cell responses for the elite control of HIV has been demonstrated in many studies, we now demonstrate that Gag-specific new (absent before expansion) memory responses are associated with sustained control of plasma HIV viremia. More importantly, the observed effect of expandable responses on residual viremia strongly suggests that immune control in chronic HIV infection is an active and ongoing process.

In this study, we further elucidated the superiority of Gag responses by showing that under conditions of very low antigen loads, HIV controllers preserve larger breadths of Gag responses to viral variants. We also show that on a per-cell basis, the expandable Gag responses retain superior antiviral qualities compared to the Nef and Env responses, even within individual subjects. Furthermore, the breadth of Gag memory responses was associated with lower residual viral loads, whereas no association was observed for either Nef or Env responses. We also showed that the superiority of Gag memory responses in terms of virus-inhibitory capacity is lost during progressive HIV infection, and complete suppression of viral replication with combination ART did not restore this. More importantly, in ART-treated individuals, the breadth of memory responses (including Gag responses) did not correlate with viral load, although there was a trend toward a negative association. Even though cross-sectional analyses cannot unequivocally provide a direct proof of causality, these data strongly suggest that low-frequency but readily expandable memory responses play an active role in sustained suppression of HIV. These data also indicate that there are qualitative differences in memory responses in controllers compared to those in progressors in terms of antiviral function, and a lack of effective expandable memory responses in progressors might allow rapid viral rebound upon cessation of antiviral therapy.

Other features associated with the superiority of Gag responses in HIV controllers are the capacity to cross-recognize viral variants and the ability to induce de novo responses to mutant viruses (9, 47, 56). Both HIV and SIV infections exhibit selection of escape variants during both primary and chronic infections. It has also been shown that the selection of viral escape variants during chronic SIV and HIV infections can result in a loss of immune control and disease progression (53, 6062). Thus, the ability of HIV to escape from virus-specific CD8+ T cell responses has been proposed to be an important obstacle for the maintenance of protective immune responses, and likewise, the viral diversity resulting from CD8+ T cell-driven viral evolution represents a major hurdle for HIV vaccine design (63). Interestingly, elite controllers appear to be able to control the virus despite ongoing evolution and the development of escape mutations in key CD8+ T cell epitopes. Two main mechanisms of control of virus replication by CD8+ T cells in the face of viral variation include the generation of a repertoire of effective T cell clonotypes and the ability to recognize individual viral epitope variants (47, 57). In this study, recognition of variant epitopes was measured by an IFN-γ ICS assay, which was shown in a previous SIV study to not necessarily correlate with the ability to suppress escape mutants (64). However, consistent with our findings, a recent study used CD4+ T cells infected with mutant viruses as target cells in virus inhibition assays and also showed that CD8+ T cells of elite controllers can suppress mutant viruses (65). Our data suggest that the capacity to maintain highly functional expandable memory responses with antiviral activity against viral variants is one possible mechanism contributing to durable virus suppression in HIV controllers.

The precise mechanisms that render Gag responses more effective in controlling HIV are unclear. Some proposed mechanisms include a rapid expression of epitopes derived from the Gag protein contained in the infecting viral particles and structural constraints of the Gag protein that impede CD8+ T cell escape (6670). However, these suggestions are inconsistent with the fact that beneficial effects of Gag-specific responses are not universal among elite controllers, and indeed, most progressors also mount detectable responses against Gag (71, 72). This discrepancy can partly be explained by our recent data showing that only a subset of Gag responses, particularly those restricted by protective alleles, exhibits superior antiviral function (10).

It is important to note that although we show strong evidence for the role of expandable memory responses in durable virus suppression, other cell subsets may play equally important roles in different stages of HIV infection. Indeed, several studies have shown that a wide range of phenotypically distinct memory subsets possess virus-inhibitory potential. In these in vitro studies, effector populations demonstrate an inhibitory capacity in short-term stimulations, whereas long-term stimulation results in increased inhibitory activity by central memory cells (73, 74). Effector memory T cells (Tem) and Tcm play complementary roles in the immune-mediated suppression of HIV replication. Tem are much more effective at controlling new infections because they are endowed with more immediate effector functions and can populate lymphoid and extralymphoid sites, which are the initial sites of HIV exposure (75). A case in point is a recent study in which a Tem-based vaccine blocked the establishment of chronic infection following challenge with a highly pathogenic SIV strain (7678). However, Tem have a limited proliferative capacity, are dependent on persistent antigens, and therefore may be less effective at controlling a fully established systemic infection. On the other hand, central memory cells are much more effective at suppressing systemic infections because they provide a readily expandable reserve force of HIV-specific CD8+ T cells with diverse specificities. Additionally, because central memory cells have a high proliferative potential, a relatively small population of pathogen-specific central memory cells can quickly mount greater anamnestic effector responses than those of Tem.

Several limitations of this study should be noted. First, this work focused on three HIV proteins, Gag, Nef, and Env. While we believe that the approach used provides a robust assessment of relevant expandable memory populations, we did not assess the potential roles of this population in responses against other HIV proteins. Second, the use of consensus peptides for our screening assays may have reduced our ability to detect some memory responses directed against autologous viruses. However, this should have affected the different cohorts equally, and yet we saw significant differences only in comparisons of the Gag-specific responses of EC to those of the other groups. Despite these limitations, our data suggest that in-depth interrogation of this previously underappreciated cell population can lead to a clearer understanding of the role that CD8+ T cell responses play during successful containment of HIV replication.

In conclusion, our data support a superior role for Gag-specific CD8+ T cell responses in immune-mediated control of HIV infection compared to those directed at Env and Nef. Furthermore, our data reinforce the concept that not all CD8+ T cell responses are equally beneficial. Together with data from previous reports, the results here link expandable memory responses to sustained virus suppression in the elite control of HIV. The demonstration of a negative correlation between expandable Gag-specific memory T cell responses and residual viremia strongly suggests that this cell population is not a mere footprint of escaped viruses but rather actively contributes to the sustained suppression of virus replication. Importantly, these data also show that HIV controllers possess a greater ability to mount responses to mutant viruses. Finally, the presence among elite controllers of large proportions of Gag-specific memory cells that are more reactive to epitope variants suggests that their induction by future HIV vaccines may be important for narrowing possible routes of rapid escape from vaccine-induced CD8+ T cell responses.

ACKNOWLEDGMENTS

We thank Thumbi Ndung'u, Filippos Porichis, and Philomena Kamya for their input in this study.

The International HIV Controllers Study (IHCS) (www.hivcontrollers.org), Collaboration for AIDS Vaccine Discovery of the Bill and Melinda Gates Foundation, and the AIDS Healthcare Foundation funded the cohort. Other funders include the Harvard University Center for AIDS Research (CFAR), an NIH-funded program (P30 AI060354) which is supported by NIH cofunding and participating institutes and centers, including the NIAID, NCI, NICHD, NHLBI, NIDA, NIMH, NIA, FIC, and OAR (F.P. and B.D.W); the Howard Hughes Medical Institute (B.D.W.); the Mark and Lisa Schwartz Foundation (B.D.W.); and the Dan and Marjorie Sullivan Foundation (Z.M.N.).

REFERENCES

  • 1.Rowland-Jones SL. 2003. Timeline: AIDS pathogenesis. What have two decades of HIV research taught us? Nat Rev Immunol 3:343–348. doi: 10.1038/nri1058. [DOI] [PubMed] [Google Scholar]
  • 2.Hazenberg MD, Otto SA, van Benthem BH, Roos MT, Coutinho RA, Lange JM, Hamann D, Prins M, Miedema F. 2003. Persistent immune activation in HIV-1 infection is associated with progression to AIDS. AIDS 17:1881–1888. doi: 10.1097/00002030-200309050-00006. [DOI] [PubMed] [Google Scholar]
  • 3.Spijkerman IJ, Prins M, Goudsmit J, Veugelers PJ, Coutinho RA, Miedema F, de Wolf F. 1997. Early and late HIV-1 RNA level and its association with other markers and disease progression in long-term AIDS-free homosexual men. AIDS 11:1383–1388. doi: 10.1097/00002030-199711000-00013. [DOI] [PubMed] [Google Scholar]
  • 4.Lyles RH, Munoz A, Yamashita TE, Bazmi H, Detels R, Rinaldo CR, Margolick JB, Phair JP, Mellors JW. 2000. Natural history of human immunodeficiency virus type 1 viremia after seroconversion and proximal to AIDS in a large cohort of homosexual men. Multicenter AIDS Cohort Study. J Infect Dis 181:872–880. [DOI] [PubMed] [Google Scholar]
  • 5.Cao Y, Qin L, Zhang L, Safrit J, Ho DD. 1995. Virologic and immunologic characterization of long-term survivors of human immunodeficiency virus type 1 infection. N Engl J Med 332:201–208. doi: 10.1056/NEJM199501263320401. [DOI] [PubMed] [Google Scholar]
  • 6.Cao Y, Qin L, Zhang L, Safrit J, Ho DD. 1996. Characterization of long-term survivors of human immunodeficiency virus type 1 infection. Immunol Lett 51:7–13. doi: 10.1016/0165-2478(96)02548-5. [DOI] [PubMed] [Google Scholar]
  • 7.Deeks SG, Walker BD. 2007. Human immunodeficiency virus controllers: mechanisms of durable virus control in the absence of antiretroviral therapy. Immunity 27:406–416. doi: 10.1016/j.immuni.2007.08.010. [DOI] [PubMed] [Google Scholar]
  • 8.Walker BD, Yu XG. 2013. Unravelling the mechanisms of durable control of HIV-1. Nat Rev Immunol 13:487–498. doi: 10.1038/nri3478. [DOI] [PubMed] [Google Scholar]
  • 9.Mothe B, Llano A, Ibarrondo J, Zamarreno J, Schiaulini M, Miranda C, Ruiz-Riol M, Berger CT, Herrero MJ, Palou E, Plana M, Rolland M, Khatri A, Heckerman D, Pereyra F, Walker BD, Weiner D, Paredes R, Clotet B, Felber BK, Pavlakis GN, Mullins JI, Brander C. 2012. CTL responses of high functional avidity and broad variant cross-reactivity are associated with HIV control. PLoS One 7:e29717. doi: 10.1371/journal.pone.0029717. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Ndhlovu ZM, Chibnik LB, Proudfoot J, Vine S, McMullen A, Cesa K, Porichis F, Jones RB, Alvino DM, Hart MG, Stampouloglou E, Piechocka-Trocha A, Kadie C, Pereyra F, Heckerman D, De Jager PL, Walker BD, Kaufmann DE. 2013. High-dimensional immunomonitoring models of HIV-1-specific CD8 T-cell responses accurately identify subjects achieving spontaneous viral control. Blood 121:801–811. doi: 10.1182/blood-2012-06-436295. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Migueles SA, Sabbaghian MS, Shupert WL, Bettinotti MP, Marincola FM, Martino L, Hallahan CW, Selig SM, Schwartz D, Sullivan J, Connors M. 2000. HLA B*5701 is highly associated with restriction of virus replication in a subgroup of HIV-infected long term nonprogressors. Proc Natl Acad Sci U S A 97:2709–2714. doi: 10.1073/pnas.050567397. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Migueles SA, Laborico AC, Shupert WL, Sabbaghian MS, Rabin R, Hallahan CW, Van Baarle D, Kostense S, Miedema F, McLaughlin M, Ehler L, Metcalf J, Liu S, Connors M. 2002. HIV-specific CD8+ T cell proliferation is coupled to perforin expression and is maintained in nonprogressors. Nat Immunol 3:1061–1068. doi: 10.1038/ni845. [DOI] [PubMed] [Google Scholar]
  • 13.International HIV Controllers Study, Pereyra F, Jia X, McLaren PJ, Telenti A, de Bakker PI, Walker BD, Ripke S, Brumme CJ, Pulit SL, Carrington M, Kadie CM, Carlson JM, Heckerman D, Graham RR, Plenge RM, Deeks SG, Gianniny L, Crawford G, Sullivan J, Gonzalez E, Davies L, Camargo A, Moore JM, Beattie N, Gupta S, Crenshaw A, Burtt NP, Guiducci C, Gupta N, Gao X, Qi Y, Yuki Y, Piechocka-Trocha A, Cutrell E, Rosenberg R, Moss KL, Lemay P, O'Leary J, Schaefer T, Verma P, Toth I, Block B, Baker B, Rothchild A, Lian J, Proudfoot J, Alvino DM, Vine S, Addo MM, et al. 2010. The major genetic determinants of HIV-1 control affect HLA class I peptide presentation. Science 330:1551–1557. doi: 10.1126/science.1195271. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Autran B, Descours B, Avettand-Fenoel V, Rouzioux C. 2011. Elite controllers as a model of functional cure. Curr Opin HIV AIDS 6:181–187. doi: 10.1097/COH.0b013e328345a328. [DOI] [PubMed] [Google Scholar]
  • 15.Julg B, Williams KL, Reddy S, Bishop K, Qi Y, Carrington M, Goulder PJ, Ndung'u T, Walker BD. 2010. Enhanced anti-HIV functional activity associated with Gag-specific CD8 T-cell responses. J Virol 84:5540–5549. doi: 10.1128/JVI.02031-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Chen H, Piechocka-Trocha A, Miura T, Brockman MA, Julg BD, Baker BM, Rothchild AC, Block BL, Schneidewind A, Koibuchi T, Pereyra F, Allen TM, Walker BD. 2009. Differential neutralization of human immunodeficiency virus (HIV) replication in autologous CD4 T cells by HIV-specific cytotoxic T lymphocytes. J Virol 83:3138–3149. doi: 10.1128/JVI.02073-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Kiepiela P, Ngumbela K, Thobakgale C, Ramduth D, Honeyborne I, Moodley E, Reddy S, de Pierres C, Mncube Z, Mkhwanazi N, Bishop K, van der Stok M, Nair K, Khan N, Crawford H, Payne R, Leslie A, Prado J, Prendergast A, Frater J, McCarthy N, Brander C, Learn GH, Nickle D, Rousseau C, Coovadia H, Mullins JI, Heckerman D, Walker BD, Goulder P. 2007. CD8+ T-cell responses to different HIV proteins have discordant associations with viral load. Nat Med 13:46–53. doi: 10.1038/nm1520. [DOI] [PubMed] [Google Scholar]
  • 18.Staprans SI, Barry AP, Silvestri G, Safrit JT, Kozyr N, Sumpter B, Nguyen H, McClure H, Montefiori D, Cohen JI, Feinberg MB. 2004. Enhanced SIV replication and accelerated progression to AIDS in macaques primed to mount a CD4 T cell response to the SIV envelope protein. Proc Natl Acad Sci U S A 101:13026–13031. doi: 10.1073/pnas.0404739101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Ngumbela KC, Day CL, Mncube Z, Nair K, Ramduth D, Thobakgale C, Moodley E, Reddy S, de Pierres C, Mkhwanazi N, Bishop K, van der Stok M, Ismail N, Honeyborne I, Crawford H, Kavanagh DG, Rousseau C, Nickle D, Mullins J, Heckerman D, Korber B, Coovadia H, Kiepiela P, Goulder PJ, Walker BD. 2008. Targeting of a CD8 T cell env epitope presented by HLA-B*5802 is associated with markers of HIV disease progression and lack of selection pressure. AIDS Res Hum Retroviruses 24:72–82. doi: 10.1089/aid.2007.0124. [DOI] [PubMed] [Google Scholar]
  • 20.Peut V, Kent SJ. 2009. Substantial envelope-specific CD8 T-cell immunity fails to control SIV disease. Virology 384:21–27. doi: 10.1016/j.virol.2008.11.030. [DOI] [PubMed] [Google Scholar]
  • 21.Miura T, Brockman MA, Brumme ZL, Brumme CJ, Pereyra F, Trocha A, Block BL, Schneidewind A, Allen TM, Heckerman D, Walker BD. 2009. 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 83:140–149. doi: 10.1128/JVI.01471-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Miura T, Brumme CJ, Brockman MA, Brumme ZL, Pereyra F, Block BL, Trocha A, John M, Mallal S, Harrigan PR, Walker BD. 2009. HLA-associated viral mutations are common in human immunodeficiency virus type 1 elite controllers. J Virol 83:3407–3412. doi: 10.1128/JVI.02459-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Miura T, Brumme ZL, Brockman MA, Rosato P, Sela J, Brumme CJ, Pereyra F, Kaufmann DE, Trocha A, Block BL, Daar ES, Connick E, Jessen H, Kelleher AD, Rosenberg E, Markowitz M, Schafer K, Vaida F, Iwamoto A, Little S, Walker BD. 2010. Impaired replication capacity of acute/early viruses in persons who become HIV controllers. J Virol 84:7581–7591. doi: 10.1128/JVI.00286-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Wright JK, Naidoo VL, Brumme ZL, Prince JL, Claiborne DT, Goulder PJ, Brockman MA, Hunter E, Ndung'u T. 2012. Impact of HLA-B*81-associated mutations in HIV-1 Gag on viral replication capacity. J Virol 86:3193–3199. doi: 10.1128/JVI.06682-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Wright JK, Novitsky V, Brockman MA, Brumme ZL, Brumme CJ, Carlson JM, Heckerman D, Wang B, Losina E, Leshwedi M, van der Stok M, Maphumulo L, Mkhwanazi N, Chonco F, Goulder PJ, Essex M, Walker BD, Ndung'u T. 2011. Influence of Gag-protease-mediated replication capacity on disease progression in individuals recently infected with HIV-1 subtype C. J Virol 85:3996–4006. doi: 10.1128/JVI.02520-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Martinez-Picado J, Prado JG, Fry EE, Pfafferott K, Leslie A, Chetty S, Thobakgale C, Honeyborne I, Crawford H, Matthews P, Pillay T, Rousseau C, Mullins JI, Brander C, Walker BD, Stuart DI, Kiepiela P, Goulder P. 2006. Fitness cost of escape mutations in p24 Gag in association with control of human immunodeficiency virus type 1. J Virol 80:3617–3623. doi: 10.1128/JVI.80.7.3617-3623.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Mudd PA, Ericsen AJ, Walsh AD, Leon EJ, Wilson NA, Maness NJ, Friedrich TC, Watkins DI. 2011. CD8+ T cell escape mutations in simian immunodeficiency virus SIVmac239 cause fitness defects in vivo, and many revert after transmission. J Virol 85:12804–12810. doi: 10.1128/JVI.05841-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Ndhlovu ZM, Proudfoot J, Cesa K, Alvino DM, McMullen A, Vine S, Stampouloglou E, Piechocka-Trocha A, Walker BD, Pereyra F. 2012. Elite controllers with low to absent effector CD8+ T cell responses maintain highly functional, broadly directed central memory responses. J Virol 86:6959–6969. doi: 10.1128/JVI.00531-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Pereyra F, Addo MM, Kaufmann DE, Liu Y, Miura T, Rathod A, Baker B, Trocha A, Rosenberg R, Mackey E, Ueda P, Lu Z, Cohen D, Wrin T, Petropoulos CJ, Rosenberg ES, Walker BD. 2008. Genetic and immunologic heterogeneity among persons who control HIV infection in the absence of therapy. J Infect Dis 197:563–571. doi: 10.1086/526786. [DOI] [PubMed] [Google Scholar]
  • 30.Wilson CC, Wong JT, Girard DD, Merrill DP, Dynan M, An DD, Kalams SA, Johnson RP, Hirsch MS, D'Aquila RT, Walker BD. 1995. Ex vivo expansion of CD4 lymphocytes from human immunodeficiency virus type 1-infected persons in the presence of combination antiretroviral agents. J Infect Dis 172:88–96. doi: 10.1093/infdis/172.1.88. [DOI] [PubMed] [Google Scholar]
  • 31.Palmer S, Wiegand AP, Maldarelli F, Bazmi H, Mican JM, Polis M, Dewar RL, Planta A, Liu S, Metcalf JA, Mellors JW, Coffin JM. 2003. New real-time reverse transcriptase-initiated PCR assay with single-copy sensitivity for human immunodeficiency virus type 1 RNA in plasma. J Clin Microbiol 41:4531–4536. doi: 10.1128/JCM.41.10.4531-4536.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Geldmacher C, Currier JR, Herrmann E, Haule A, Kuta E, McCutchan F, Njovu L, Geis S, Hoffmann O, Maboko L, Williamson C, Birx D, Meyerhans A, Cox J, Hoelscher M. 2007. CD8 T-cell recognition of multiple epitopes within specific Gag regions is associated with maintenance of a low steady-state viremia in human immunodeficiency virus type 1-seropositive patients. J Virol 81:2440–2448. doi: 10.1128/JVI.01847-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Edwards BH, Bansal A, Sabbaj S, Bakari J, Mulligan MJ, Goepfert PA. 2002. 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 76:2298–2305. doi: 10.1128/jvi.76.5.2298-2305.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Ranasinghe S, Cutler S, Davis I, Lu R, Soghoian DZ, Qi Y, Sidney J, Kranias G, Flanders MD, Lindqvist M, Kuhl B, Alter G, Deeks SG, Walker BD, Gao X, Sette A, Carrington M, Streeck H. 2013. Association of HLA-DRB1-restricted CD4(+) T cell responses with HIV immune control. Nat Med 19:930–933. doi: 10.1038/nm.3229. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Jia M, Hong K, Chen J, Ruan Y, Wang Z, Su B, Ren G, Zhang X, Liu Z, Zhao Q, Li D, Peng H, Altfeld M, Walker BD, Yu XG, Shao Y. 2012. Preferential CTL targeting of Gag is associated with relative viral control in long-term surviving HIV-1 infected former plasma donors from China. Cell Res 22:903–914. doi: 10.1038/cr.2012.19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Radebe M, Nair K, Chonco F, Bishop K, Wright JK, van der Stok M, Bassett IV, Mncube Z, Altfeld M, Walker BD, Ndung'u T. 2011. Limited immunogenicity of HIV CD8+ T-cell epitopes in acute clade C virus infection. J Infect Dis 204:768–776. doi: 10.1093/infdis/jir394. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Mlotshwa M, Riou C, Chopera D, de Assis Rosa D, Ntale R, Treunicht F, Woodman Z, Werner L, van Loggerenberg F, Mlisana K, Abdool Karim S, Williamson C, Gray CM, CAPRISA 002 Study Team . 2010. Fluidity of HIV-1-specific T-cell responses during acute and early subtype C HIV-1 infection and associations with early disease progression. J Virol 84:12018–12029. doi: 10.1128/JVI.01472-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Lichterfeld M, Yu XG, Cohen D, Addo MM, Malenfant J, Perkins B, Pae E, Johnston MN, Strick D, Allen TM, Rosenberg ES, Korber B, Walker BD, Altfeld M. 2004. HIV-1 Nef is preferentially recognized by CD8 T cells in primary HIV-1 infection despite a relatively high degree of genetic diversity. AIDS 18:1383–1392. doi: 10.1097/01.aids.0000131329.51633.a3. [DOI] [PubMed] [Google Scholar]
  • 39.Kim GJ, Lee HS, Hong KJ, Kim SS. 2010. Dynamic correlation between CTL response and viral load in primary human immunodeficiency virus-1 infected Koreans. Virol J 7:239. doi: 10.1186/1743-422X-7-239. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Letvin NL, Montefiori DC, Yasutomi Y, Perry HC, Davies ME, Lekutis C, Alroy M, Freed DC, Lord CI, Handt LK, Liu MA, Shiver JW. 1997. Potent, protective anti-HIV immune responses generated by bimodal HIV envelope DNA plus protein vaccination. Proc Natl Acad Sci U S A 94:9378–9383. doi: 10.1073/pnas.94.17.9378. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Bell SJ, Cooper DA, Kemp BE, Doherty RR, Penny R. 1992. Definition of an immunodominant T cell epitope contained in the envelope gp41 sequence of HIV-1. Clin Exp Immunol 87:37–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Belyakov IM, Ahlers JD, Brandwein BY, Earl P, Kelsall BL, Moss B, Strober W, Berzofsky JA. 1998. The importance of local mucosal HIV-specific CD8(+) cytotoxic T lymphocytes for resistance to mucosal viral transmission in mice and enhancement of resistance by local administration of IL-12. J Clin Invest 102:2072–2081. doi: 10.1172/JCI5102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Zhuang Y, Sun Y, Zhai S, Huang D, Zhao S, Wang S, Kang W, Li X, Walker BD, Altfeld M, Yu XG. 2008. Relative dominance of Env-gp41-specific cytotoxic T lymphocytes responses in HIV-1 advanced infection. Curr HIV Res 6:239–245. doi: 10.2174/157016208784324921. [DOI] [PubMed] [Google Scholar]
  • 44.Migueles SA, Osborne CM, Royce C, Compton AA, Joshi RP, Weeks KA, Rood JE, Berkley AM, Sacha JB, Cogliano-Shutta NA, Lloyd M, Roby G, Kwan R, McLaughlin M, Stallings S, Rehm C, O'Shea MA, Mican J, Packard BZ, Komoriya A, Palmer S, Wiegand AP, Maldarelli F, Coffin JM, Mellors JW, Hallahan CW, Follman DA, Connors M. 2008. Lytic granule loading of CD8+ T cells is required for HIV-infected cell elimination associated with immune control. Immunity 29:1009–1021. doi: 10.1016/j.immuni.2008.10.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Migueles SA, Weeks KA, Nou E, Berkley AM, Rood JE, Osborne CM, Hallahan CW, Cogliano-Shutta NA, Metcalf JA, McLaughlin M, Kwan R, Mican JM, Davey RT Jr, Connors M. 2009. Defective human immunodeficiency virus-specific CD8+ T-cell polyfunctionality, proliferation, and cytotoxicity are not restored by antiretroviral therapy. J Virol 83:11876–11889. doi: 10.1128/JVI.01153-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Hersperger AR, Migueles SA, Betts MR, Connors M. 2011. Qualitative features of the HIV-specific CD8+ T-cell response associated with immunologic control. Curr Opin HIV AIDS 6:169–173. doi: 10.1097/COH.0b013e3283454c39. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Chen H, Ndhlovu ZM, Liu D, Porter LC, Fang JW, Darko S, Brockman MA, Miura T, Brumme ZL, Schneidewind A, Piechocka-Trocha A, Cesa KT, Sela J, Cung TD, Toth I, Pereyra F, Yu XG, Douek DC, Kaufmann DE, Allen TM, Walker BD. 2012. TCR clonotypes modulate the protective effect of HLA class I molecules in HIV-1 infection. Nat Immunol 13:691–700. doi: 10.1038/ni.2342. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Pereyra F, Palmer S, Miura T, Block BL, Wiegand A, Rothchild AC, Baker B, Rosenberg R, Cutrell E, Seaman MS, Coffin JM, Walker BD. 2009. Persistent low-level viremia in HIV-1 elite controllers and relationship to immunologic parameters. J Infect Dis 200:984–990. doi: 10.1086/605446. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Mellors JW, Munoz A, Giorgi JV, Margolick JB, Tassoni CJ, Gupta P, Kingsley LA, Todd JA, Saah AJ, Detels R, Phair JP, Rinaldo CR Jr. 1997. Plasma viral load and CD4+ lymphocytes as prognostic markers of HIV-1 infection. Ann Intern Med 126:946–954. doi: 10.7326/0003-4819-126-12-199706150-00003. [DOI] [PubMed] [Google Scholar]
  • 50.Arnaout RA, Lloyd AL, O'Brien TR, Goedert JJ, Leonard JM, Nowak MA. 1999. A simple relationship between viral load and survival time in HIV-1 infection. Proc Natl Acad Sci U S A 96:11549–11553. doi: 10.1073/pnas.96.20.11549. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Barouch DH, Kunstman J, Kuroda MJ, Schmitz JE, Santra S, Peyerl FW, Krivulka GR, Beaudry K, Lifton MA, Gorgone DA, Montefiori DC, Lewis MG, Wolinsky SM, Letvin NL. 2002. Eventual AIDS vaccine failure in a rhesus monkey by viral escape from cytotoxic T lymphocytes. Nature 415:335–339. doi: 10.1038/415335a. [DOI] [PubMed] [Google Scholar]
  • 52.Phillips RE, Rowland-Jones S, Nixon DF, Gotch FM, Edwards JP, Ogunlesi AO, Elvin JG, Rothbard JA, Bangham CR, Rizza CR, McMichael AJ. 1991. Human immunodeficiency virus genetic variation that can escape cytotoxic T cell recognition. Nature 354:453–459. doi: 10.1038/354453a0. [DOI] [PubMed] [Google Scholar]
  • 53.Koenig S, Conley AJ, Brewah YA, Jones GM, Leath S, Boots LJ, Davey V, Pantaleo G, Demarest JF, Carter C, Wannebo C, Yannelli JR, Rosenberg SA, Lane HC. 1995. Transfer of HIV-1-specific cytotoxic T lymphocytes to an AIDS patient leads to selection for mutant HIV variants and subsequent disease progression. Nat Med 1:330–336. doi: 10.1038/nm0495-330. [DOI] [PubMed] [Google Scholar]
  • 54.Goulder PJ, Phillips RE, Colbert RA, McAdam S, Ogg G, Nowak MA, Giangrande P, Luzzi G, Morgan B, Edwards A, McMichael AJ, Rowland-Jones S. 1997. Late escape from an immunodominant cytotoxic T-lymphocyte response associated with progression to AIDS. Nat Med 3:212–217. doi: 10.1038/nm0297-212. [DOI] [PubMed] [Google Scholar]
  • 55.Goulder PJ, Brander C, Tang Y, Tremblay C, Colbert RA, Addo MM, Rosenberg ES, Nguyen T, Allen R, Trocha A, Altfeld M, He S, Bunce M, Funkhouser R, Pelton SI, Burchett SK, McIntosh K, Korber BT, Walker BD. 2001. Evolution and transmission of stable CTL escape mutations in HIV infection. Nature 412:334–338. doi: 10.1038/35085576. [DOI] [PubMed] [Google Scholar]
  • 56.Allen TM, Yu XG, Kalife ET, Reyor LL, Lichterfeld M, John M, Cheng M, Allgaier RL, Mui S, Frahm N, Alter G, Brown NV, Johnston MN, Rosenberg ES, Mallal SA, Brander C, Walker BD, Altfeld M. 2005. De novo generation of escape variant-specific CD8+ T-cell responses following cytotoxic T-lymphocyte escape in chronic human immunodeficiency virus type 1 infection. J Virol 79:12952–12960. doi: 10.1128/JVI.79.20.12952-12960.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Miura T, Brockman MA, Schneidewind A, Lobritz M, Pereyra F, Rathod A, Block BL, Brumme ZL, Brumme CJ, Baker B, Rothchild AC, Li B, Trocha A, Cutrell E, Frahm N, Brander C, Toth I, Arts EJ, Allen TM, Walker BD. 2009. HLA-B57/B*5801 human immunodeficiency virus type 1 elite controllers select for rare Gag variants associated with reduced viral replication capacity and strong cytotoxic T-lymphotye [sic] recognition. J Virol 83:2743–2755. doi: 10.1128/JVI.02265-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Brumme ZL, Brumme CJ, Carlson J, Streeck H, John M, Eichbaum Q, Block BL, Baker B, Kadie C, Markowitz M, Jessen H, Kelleher AD, Rosenberg E, Kaldor J, Yuki Y, Carrington M, Allen TM, Mallal S, Altfeld M, Heckerman D, Walker BD. 2008. Marked epitope- and allele-specific differences in rates of mutation in human immunodeficiency type 1 (HIV-1) Gag, Pol, and Nef cytotoxic T-lymphocyte epitopes in acute/early HIV-1 infection. J Virol 82:9216–9227. doi: 10.1128/JVI.01041-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Feeney ME, Tang Y, Pfafferott K, Roosevelt KA, Draenert R, Trocha A, Yu XG, Verrill C, Allen T, Moore C, Mallal S, Burchett S, McIntosh K, Pelton SI, St John MA, Hazra R, Klenerman P, Altfeld M, Walker BD, Goulder PJ. 2005. HIV-1 viral escape in infancy followed by emergence of a variant-specific CTL response. J Immunol 174:7524–7530. doi: 10.4049/jimmunol.174.12.7524. [DOI] [PubMed] [Google Scholar]
  • 60.Streeck H, Schweighardt B, Jessen H, Allgaier RL, Wrin T, Stawiski EW, Jessen AB, Allen TM, Walker BD, Altfeld M. 2007. Loss of HIV-1-specific T-cell responses associated with very rapid HIV-1 disease progression. AIDS 21:889–891. doi: 10.1097/QAD.0b013e3280f77439. [DOI] [PubMed] [Google Scholar]
  • 61.Boutwell CL, Carlson JM, Lin TH, Seese A, Power KA, Peng J, Tang Y, Brumme ZL, Heckerman D, Schneidewind A, Allen TM. 2013. Frequent and variable cytotoxic-T-lymphocyte escape-associated fitness costs in the human immunodeficiency virus type 1 subtype B Gag proteins. J Virol 87:3952–3965. doi: 10.1128/JVI.03233-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Borrow P, Lewicki H, Wei X, Horwitz MS, Peffer N, Meyers H, Nelson JA, Gairin JE, Hahn BH, Oldstone MB, Shaw GM. 1997. Antiviral pressure exerted by HIV-1-specific cytotoxic T lymphocytes (CTLs) during primary infection demonstrated by rapid selection of CTL escape virus. Nat Med 3:205–211. doi: 10.1038/nm0297-205. [DOI] [PubMed] [Google Scholar]
  • 63.Leslie AJ, Pfafferott KJ, Chetty P, Draenert R, Addo MM, Feeney M, Tang Y, Holmes EC, Allen T, Prado JG, Altfeld M, Brander C, Dixon C, Ramduth D, Jeena P, Thomas SA, St John A, Roach TA, Kupfer B, Luzzi G, Edwards A, Taylor G, Lyall H, Tudor-Williams G, Novelli V, Martinez-Picado J, Kiepiela P, Walker BD, Goulder PJ. 2004. HIV evolution: CTL escape mutation and reversion after transmission. Nat Med 10:282–289. doi: 10.1038/nm992. [DOI] [PubMed] [Google Scholar]
  • 64.Valentine LE, Piaskowski SM, Rakasz EG, Henry NL, Wilson NA, Watkins DI. 2008. Recognition of escape variants in ELISPOT does not always predict CD8+ T-cell recognition of simian immunodeficiency virus-infected cells expressing the same variant sequences. J Virol 82:575–581. doi: 10.1128/JVI.00275-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Pohlmeyer CW, Buckheit RW III, Siliciano RF, Blankson JN. 2013. CD8+ T cells from HLA-B*57 elite suppressors effectively suppress replication of HIV-1 escape mutants. Retrovirology 10:152. doi: 10.1186/1742-4690-10-152. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Dahirel V, Shekhar K, Pereyra F, Miura T, Artyomov M, Talsania S, Allen TM, Altfeld M, Carrington M, Irvine DJ, Walker BD, Chakraborty AK. 2011. Coordinate linkage of HIV evolution reveals regions of immunological vulnerability. Proc Natl Acad Sci U S A 108:11530–11535. doi: 10.1073/pnas.1105315108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Tang Y, Huang S, Dunkley-Thompson J, Steel-Duncan JC, Ryland EG, St John MA, Hazra R, Christie CD, Feeney ME. 2010. Correlates of spontaneous viral control among long-term survivors of perinatal HIV-1 infection expressing human leukocyte antigen-B57. AIDS 24:1425–1435. doi: 10.1097/QAD.0b013e32833a2b5b. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Balamurugan A, Ali A, Boucau J, Le Gall S, Ng HL, Yang OO. 2013. HIV-1 Gag cytotoxic T lymphocyte epitopes vary in presentation kinetics relative to HLA class I downregulation. J Virol 87:8726–8734. doi: 10.1128/JVI.01040-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Ali A, Lubong R, Ng H, Brooks DG, Zack JA, Yang OO. 2004. Impacts of epitope expression kinetics and class I downregulation on the antiviral activity of human immunodeficiency virus type 1-specific cytotoxic T lymphocytes. J Virol 78:561–567. doi: 10.1128/JVI.78.2.561-567.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Troyer RM, McNevin J, Liu Y, Zhang SC, Krizan RW, Abraha A, Tebit DM, Zhao H, Avila S, Lobritz MA, McElrath MJ, Le Gall S, Mullins JI, Arts EJ. 2009. Variable fitness impact of HIV-1 escape mutations to cytotoxic T lymphocyte (CTL) response. PLoS Pathog 5:e1000365. doi: 10.1371/journal.ppat.1000365. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Fu TM, Dubey SA, Mehrotra DV, Freed DC, Trigona WL, Adams-Muhler L, Clair JH, Evans TG, Steigbigel R, Jacobson JM, Goepfert PA, Mulligan MJ, Kalams SA, Rinaldo C, Zhu L, Cox KS, Guan L, Long R, Persaud N, Caulfield MJ, Sadoff JC, Emini EA, Thaler S, Shiver JW. 2007. Evaluation of cellular immune responses in subjects chronically infected with HIV type 1. AIDS Res Hum Retroviruses 23:67–76. doi: 10.1089/aid.2006.0114. [DOI] [PubMed] [Google Scholar]
  • 72.Addo MM, Yu XG, Rathod A, Cohen D, Eldridge RL, Strick D, Johnston MN, Corcoran C, Wurcel AG, Fitzpatrick CA, Feeney ME, Rodriguez WR, Basgoz N, Draenert R, Stone DR, Brander C, Goulder PJ, Rosenberg ES, Altfeld M, Walker BD. 2003. Comprehensive epitope analysis of human immunodeficiency virus type 1 (HIV-1)-specific T-cell responses directed against the entire expressed HIV-1 genome demonstrate broadly directed responses, but no correlation to viral load. J Virol 77:2081–2092. doi: 10.1128/JVI.77.3.2081-2092.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Freel SA, Lamoreaux L, Chattopadhyay PK, Saunders K, Zarkowsky D, Overman RG, Ochsenbauer C, Edmonds TG, Kappes JC, Cunningham CK, Denny TN, Weinhold KJ, Ferrari G, Haynes BF, Koup RA, Graham BS, Roederer M, Tomaras GD. 2010. Phenotypic and functional profile of HIV-inhibitory CD8 T cells elicited by natural infection and heterologous prime/boost vaccination. J Virol 84:4998–5006. doi: 10.1128/JVI.00138-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Buckheit RW III, Salgado M, Silciano RF, Blankson JN. 2012. Inhibitory potential of subpopulations of CD8+ T cells in HIV-1-infected elite suppressors. J Virol 86:13679–13688. doi: 10.1128/JVI.02439-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Masopust D, Picker LJ. 2012. Hidden memories: frontline memory T cells and early pathogen interception. J Immunol 188:5811–5817. doi: 10.4049/jimmunol.1102695. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Hansen SG, Ford JC, Lewis MS, Ventura AB, Hughes CM, Coyne-Johnson L, Whizin N, Oswald K, Shoemaker R, Swanson T, Legasse AW, Chiuchiolo MJ, Parks CL, Axthelm MK, Nelson JA, Jarvis MA, Piatak M Jr, Lifson JD, Picker LJ. 2011. Profound early control of highly pathogenic SIV by an effector memory T-cell vaccine. Nature 473:523–527. doi: 10.1038/nature10003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Hansen SG, Sacha JB, Hughes CM, Ford JC, Burwitz BJ, Scholz I, Gilbride RM, Lewis MS, Gilliam AN, Ventura AB, Malouli D, Xu G, Richards R, Whizin N, Reed JS, Hammond KB, Fischer M, Turner JM, Legasse AW, Axthelm MK, Edlefsen PT, Nelson JA, Lifson JD, Fruh K, Picker LJ. 2013. Cytomegalovirus vectors violate CD8+ T cell epitope recognition paradigms. Science 340:1237874. doi: 10.1126/science.1237874. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Hansen SG, Vieville C, Whizin N, Coyne-Johnson L, Siess DC, Drummond DD, Legasse AW, Axthelm MK, Oswald K, Trubey CM, Piatak M Jr, Lifson JD, Nelson JA, Jarvis MA, Picker LJ. 2009. Effector memory T cell responses are associated with protection of rhesus monkeys from mucosal simian immunodeficiency virus challenge. Nat Med 15:293–299. doi: 10.1038/nm.1935. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Journal of Virology are provided here courtesy of American Society for Microbiology (ASM)

RESOURCES