当前位置: 首页 > 医学版 > 期刊论文 > 基础医学 > 病菌学杂志 > 2005年 > 第8期 > 正文
编号:11200366
Human Immunodeficiency Virus (HIV)-Specific Gamma
http://www.100md.com 病菌学杂志 2005年第8期
     McGill University Health Center, Montreal, Quebec, Canada

    Massachusetts General Hospital, Boston, Massachusetts

    ABSTRACT

    Immune responses to human immunodeficiency virus (HIV) are detected at all stages of infection and are believed to be responsible for controlling viremia. This study seeks to determine whether gamma interferon (IFN-)-secreting HIV-specific T-cell responses influence disease progression as defined by the rate of CD4 decline. The study population consisted of 31 subjects na?ve to antiretroviral therapy. All were monitored clinically for a median of 24 months after the time they were tested for HIV-specific responses. The rate of CD4+-T-cell loss was calculated for all participants from monthly CD4 counts. Within this population, 17 subjects were classified as typical progressors, 6 subjects were classified as fast progressors, and 8 subjects were classified as slow progressors. Peripheral blood mononuclear cells were screened for HIV-specific IFN- responses to all expressed HIV genes. Among the detected immune responses, 48% of the recognized peptides were encoded by Gag and 19% were encoded by Nef gene products. Neither the breadth nor the magnitude of HIV-specific responses correlated with the viral load or rate of CD4 decline. The breadth and magnitude of HIV-specific responses did not differ significantly among typical, fast, and slow progressors. These results support the conclusion that although diverse HIV-specific IFN--secreting responses are mounted during the asymptomatic phase, these responses do not seem to modulate disease progression rates.

    INTRODUCTION

    Human immunodeficiency virus (HIV) infection is characterized by robust virus-specific CD8+ cytotoxic T-lymphocyte (CTL) responses that are believed to play a role in the control of viral replication and dissemination. These responses are thought to be responsible for the reduction of viremia in primary infection (8, 35), maintenance of the viral set point during the asymptomatic phase (53), and control of viremia in long-term nonprogressors (27). The most direct evidence supporting a role for CD8+ CTL in viral control comes from an animal model for HIV infection, i.e., macaques infected with the simian immunodeficiency virus (SIV). In this model, depletion of CD8+ T cells results in either increased viremia that remains high until the CD8+ T cells are reconstituted or uncontrolled viremia and rapid disease progression (31, 59). CD8+ CTLs are believed to exert a selective pressure on the virus sequences they recognize, as demonstrated by the preferential emergence of viral escape mutations in sequences corresponding to CTL epitopes in SIV-infected macaques (2, 33, 52) and in HIV-infected humans (23, 24, 34, 55). However, despite the presence of HIV-specific CTLs at all stages of the infection, CTLs are unable to clear infection (40).

    HIV-specific CD8+ T cells can suppress viral replication by several mechanisms. They can lyse HIV-infected targets before mature virions can be produced, secrete factors such as MIP-1, MIP-1?, or RANTES, which bind the CCR5 coreceptor and prevent HIV entry into target cells, and secrete factors that control viral replication (38, 65). HIV-specific CD8+ and CD4+ T cells also secrete the antiviral cytokine gamma interferon (IFN-) upon activation (30). The IFN- enzyme-linked immunospot assay (ELISPOT assay) has come into common use as a high-throughput, sensitive technique for measuring the frequency of cells able to secrete a cytokine, such as IFN-, upon antigen stimulation (50, 58). The availability of peptide sets corresponding to all expressed HIV genes has led to the development of methods that permit the comprehensive screening of peripheral blood mononuclear cells (PBMCs) for responses to HIV.

    Several reports have used peptide pool matrix arrays that include sequences corresponding to all expressed HIV genes to screen for the breadth, magnitude, and specificity of HIV-specific effector function in diverse patient populations. Conflicting results on whether an association exists between the breadth and/or magnitude of HIV-specific IFN- secretion and viral load have been reported (1, 7, 12, 17, 51). Furthermore, the cross-sectional design of these studies precluded the assessment of whether breadth, magnitude, and specificity of HIV-specific immune responses predicted the subsequent rate of CD4+-T-cell loss. It is well established that the plasma viral load set point strongly predicts the rate of CD4+-T-cell decline, progression to AIDS, and death (47). While direct cytopathic effects of HIV on CD4+ T cells likely plays a role in CD4+-T-cell depletion, most of the cells in this compartment that die are not infected (18, 20). Indirect effects of viral replication, such as immune activation causing increased cell turnover, have been postulated to contribute to the progressive loss of CD4+ T cells (28, 46). Immune activation is often measured by expression levels of CD38 on CD8+ T cells (21, 26). The predictive value of this marker of immune activation for HIV disease progression, including rate of CD4+-T-cell decline, was shown to be stronger than and independent of the plasma viral load set point, demonstrating that immune activation is also an important factor in the pathogenic process (37, 41). Therefore, previous studies that assessed associations between total HIV-specific immune responses measured in IFN- ELISPOT assays and viral load evaluated correlations between these responses and one factor contributing to rate of CD4+-T-cell decline.

    In this report, we used an IFN- ELISPOT assay to screen PBMCs from 31 antiretroviral therapy-na?ve chronically infected subjects with known rates of CD4+-T-cell decline calculated from CD4+-T-cell counts measured monthly for a minimum of 1 year from the time point used for immune response assessments. The hypothesis being tested was that if HIV-specific IFN- secretion is a correlate of protection from disease progression, subjects with slower kinetics of CD4+-T-cell loss should have more intense and/or broader responses than those with typical or fast disease progression rates.

    We found that neither the magnitude nor the breadth of HIV-specific recognition was correlated with CD4+- or CD8+-T-cell counts, viral load, or rate of CD4+-T-cell decline in this untreated population in the chronic phase of HIV infection. These conclusions also applied to comparisons made among three subgroups selected from among the larger population, namely: a subgroup with a typical rate of CD4+-T-cell loss, a fast progressor group, and a slow progressor group with a rate of CD4+-T-cell decline similar to that seen in uninfected subjects. The three subgroups exhibited similar breadth and magnitude of HIV-specific IFN- secretion to all expressed HIV genes.

    MATERIALS AND METHODS

    Study population. We studied 31 HIV-infected subjects (26 males and 1 female infected through sexual exposure, 3 hemophilia patients, and 1 intravenous drug user infected parenterally) in the chronic phase of infection. All subjects were asymptomatic and na?ve to antiretroviral therapy and were monitored clinically for a median of 24 (range, 12 to 24) months (Table 1). Monthly CD4 counts taken within this period were used to calculate the rate of CD4 decline for each individual from the test date. Because monthly CD4 counts were available for these individuals, we were able to identify three subgroups from this population, based on rates of CD4+-T-cell decline, for additional analyses. Informed consent was obtained from all study subjects, and the research conformed to all ethical guidelines of the authors' institutions.

    Laboratory testing. Plasma viremia was measured by using an HIV RNA multiplex b-DNA assay, version 2.0. Those undetectable by this assay were retested with the ultrasensitive Chiron b-DNA assay (version 3.0; Chiron Corp., Emeryville, Calif.) with a detection limit of 50 HIV type 1 (HIV-1) RNA copies/ml of plasma. Flow cytometric analysis using fluorescein isothiocyanate-conjugated anti-CD3-phycoerythrin-conjugated anti-CD4 and fluorescein isothiocyanate-conjugated anti-CD3-phycoerythrin-conjugated anti-CD8 (Simultest; Becton Dickinson, Palo Alto, Calif.) was used to assess absolute CD4+- and CD8+-T-cell counts. Complete white blood cell counts were done on whole blood with an Advia 120 instrument (Bayer, Tarrytown, N.Y.).

    Cells. PBMCs were isolated from blood by density gradient centrifugation (Ficoll-Paque; Pharmacia, Uppsala, Sweden) and cryopreserved in 10% dimethyl sulfoxide (DMSO; Sigma-Aldrich, St. Louis, Mo.) with 90% fetal calf serum (FCS; Canadian Life Technologies, Burlington, Ontario, Canada).

    HLA. Subjects were typed for major histocompatibility complex (MHC) class I antigen expression by amplification refractory mutation system-PCR with 95 primer sets amplifying defined MHC class I alleles (ABC SSP Unitray; Pel-Freez Clinical Systems, Brown Deer, Wis.) (11). Genomic DNA for molecular HLA typing was prepared from either fresh blood or Epstein-Barr virus-transformed B-cell lines by using a QIAamp DNA blood kit (QIAGEN, Inc., Mississauga, Ontario, Canada).

    Design of peptide pool matrices. The HIV peptide sets used for stimulation were 15 amino acids (aa) with 11-aa overlaps (Gag, Env, Nef, Tat, Rev, Vpr, Vpu, and Vif) or 20 aa with 10-aa overlaps (Pol). The peptides were obtained from the NIH AIDS Research and Reference Reagent Program (Rockville, Md.). Lyophilized peptides (n = 620) spanning all HIV-1 gene products were dissolved to a final concentration of 10 mg/ml in DMSO and stored at –70°C. These samples included 100 Pol 20-mers and 122 Gag 15-mers corresponding to the HIV-1HXB2 clade B isolate; 49 Nef, 27 Rev, 23 Tat, 46 Vif, 22 Vpr, and 19 Vpu 15-mers corresponding to consensus HIV (Vpu) or consensus clade B sequence (Nef, Rev, Tat, Vif, and Vpr); and 212 Env 15-mers corresponding to the HIVMN clade B isolate. Pools containing 2 to 15 peptides were prepared and organized into matrices of Gag, Pol, Nef, Env, and accessory gene peptide pools such that each peptide was present in two pools within each peptide matrix.

    Matrix assay for single-cell IFN- release. IFN- secretion by HIV-specific cells was quantified by using an ELISPOT assay. Cells were resuspended in RPMI containing 10% FCS, 2 mM L-glutamine (ICN Biomedicals, Costa Mesa, Calif.), 50 μM ?-mercaptoethanol (Sigma-Aldrich), 50 IU of penicillin/ml, and 50 μg of streptomycin (ICN Biomedicals)/ml. Cells (70,000 to 100,000) were plated in 96-well polyvinylidene difluoride-backed plates (MAIPS 45; Millipore, Bedford, Mass.) precoated overnight at 4°C with 100 μl of the anti-IFN- monoclonal antibody 1-DK (5 μg/ml; Mabtech, Stockholm, Sweden). PBMCs were stimulated with peptide pools such that the final concentration of each peptide within a pool was 2 to 4 μg/ml. Medium alone with concentrations of DMSO equivalent to that in peptide pools was used as a negative control, and anti-CD3 antibody (Research Diagnostics, Inc., Flanders, N.J.) was used as a positive control stimulus. A pool of 23 peptides (NIH AIDS Research and Reference Reagent Program), derived from cytomegalovirus, Epstein-Barr virus, and influenza virus, restricted to common MHC class I alleles, was also used as a positive control stimulus (14). Sixty-one percent of the individuals tested responded to this pool with an average magnitude of 342 spot-forming cells (SFCs)/106 PBMCs. The plates were incubated at 37°C in 5% CO2 overnight. Cells were washed away with phosphate-buffered saline supplemented with 0.05% Tween 20. Spots corresponding to the footprint of IFN--secreting cells were developed by sequential addition followed by washing with phosphate-buffered saline of biotinylated anti-IFN- monoclonal antibody 7-B6-1 (Mabtech) diluted to 0.5 μg/ml for 3 h at room temperature (RT), alkaline phosphatase conjugated to streptavidin (Mabtech) at a dilution of 1:2,000 for 2 h at RT, and of 5-bromo-4-chloro-3-indolylphosphate and nitroblue tetrazolium (Bio-Rad Laboratories, Hercules, Calif.) for 10 to 15 min at RT. Spots were counted with a stereomicroscope (Carl Zeiss, London, Ontario, Canada). Results are expressed as SFCs/106 PBMCs following the subtraction of negative controls. The criteria used to identify positive responses were obtained by testing 10 low-risk HIV-uninfected individuals with the same peptide pool matrix array. Uninfected individuals produced a mean of 18.3 ± 20.2 SFCs/106 PBMCs to the stimulatory peptide pools. A positive response was defined as being 3 standard deviations above that seen for uninfected subjects, i.e., >79 SFCs/106 PBMCs and at least threefold greater than the autologous negative control wells.

    Confirmation of peptide specificity. The stimulatory capacity of candidate peptides identified in the peptide pool matrix ELISPOT assay was confirmed in a second experiment with cells from the same time point stimulated with individual peptides that were common to two stimulatory peptide pools in the initial ELISPOT screen. For single peptide verification experiments, cells were plated in triplicate with 4 μg of individual candidate peptides/ml. The criteria used for the original peptide pool matrix experiment were also used for the verification assay to identify positive responses. Responses to two adjacent overlapping peptides were counted as a single response. The higher response of two adjacent overlapping peptides was used to estimate the total magnitude. Therefore, analyses that include measures of breadth are based on a minimum estimate, as it is possible that peptides and sequential peptides contain more than one epitope. CD8 depletion was performed by using immunomagnetic beads (Miltenyi Biotec, Auburn, Calif.) in order to determine the cell population responsible for the peptide-specific response. Ninety percent of the CD3+ CD8+ cell population was depleted, as determined by flow cytometry. Among 20 peptide-specific responses detected for three individuals, all were attributed to CD3+ CD8+ lymphocytes.

    Statistical analysis. Statistical analysis and graphical presentation were performed by using GraphPad InStat 3.05 and Excel 2002. CD4+-T-lymphocyte slopes were obtained by fitting a linear regression model by the least-squares method for each patient. Analysis of variance (ANOVA) and Student's t test were used to assess differences in age, CD4+-T-cell count, CD8+-T-cell count, and viral load magnitude and breadth among the groups. Pearson's correlation analysis was used to study the relationships between the breadth and magnitude of HIV-specific responses and clinical parameters, such as viral load and rate of CD4+-T-cell decline. P values of less than 0.05 were considered significant.

    RESULTS

    Study population. Table 1 provides information on the 31 highly active retroviral therapy-na?ve chronically infected subjects studied. At the time point used for screening for HIV-specific IFN- secretion, the study population was a median of 34 (range, 18 to 51) years old and had a median CD4+-T-cell count of 572 (range, 374 to 999) cells/μl, a median CD8+-T-cell count of 1,144 (range, 720 to 2,412) cells/μl, and a median viral load of 1,452 (range, 72 to 22,065) HIV-1 RNA copies/ml. Monthly absolute CD4+-T-cell count values taken over a median of 24 (range, 12 to 24) months were used to determine the rate of CD4+-T-cell loss for this population of a median of 57 (range, 0 to 177) cells/μl/year. From this study population, three subgroups were constituted based on their rates of CD4+-T-cell decline. Subjects in the first subgroup were designated typical progressors (n = 17), those in the second subgroup were designated fast progressors (n = 6), and those in the third subgroup were designated slow progressors (n = 8) (Table 2). The rate of CD4+-T-cell loss in the slow progressor group was not significantly different from that seen in low-risk control HIV-uninfected subjects (n = 14) monitored for a median time of 36 (range, 24 to 108) months and who lost a median of 7.9 (range, 0 to 58) cells/μl/year (P values were not significant [NS]; Student's t test). All subgroups had a similar median age, CD4+-T-cell count, CD8+-T-cell count, and viral load at the time points tested (P values was NS; ANOVA). CD4+-T-cell slopes for the follow-up period after testing differed significantly among the three groups (P < 0.001; ANOVA) (Table 2).

    Characteristics of HIV-specific IFN- secretion. A peptide pool matrix IFN- ELISPOT assay was used to screen PBMCs from the 31 treatment-na?ve subjects in the chronic phase of infection for HIV-specific immune responses to all expressed HIV genes. Of the 620 peptides used for screening, 104 stimulated a response. The position of the 104 peptides (35 Gag, 19 Pol, 19 Nef, 16 Acc, and 15 Env) with respect to their location on the HIV sequence is shown in Fig. 1A. The median number of peptides recognized by this study population was 5 (range, 1 to 11), with a median cumulative magnitude of 1,933 (range, 100 to 9,176) SFCs/106 PBMCs (Fig. 1B). The relative contribution of each gene product to the total breadth and magnitude is shown in Fig. 1C and D, respectively. Determination of these values took into consideration the relative length of each gene product. Gag was the gene product most frequently recognized, with 48% of all responses and 53% of the cumulative magnitude targeting this protein (Fig. 1C and D). The magnitude and breadth of the responses targeting Gag were significantly higher than those for all other gene products (P < 0.001; ANOVA). Among Gag subunits, p24- and p17-derived peptides contributed most of the stimulatory capacity attributed to the Gag gene product. All 31 individuals responded to at least one Gag peptide (Fig. 1B). Nef was the second most frequently recognized protein; 19% of all responses detected were directed at Nef-derived peptides, whereas 21% of the cumulative magnitude targeted this gene product (Fig. 1C and D).

    Rate of CD4 decline and viral load does not correlate with the breadth and magnitude of HIV-specific IFN--secreting responses. Because at least 12 months of clinical follow-up information was available for this study population after the time of immune response assessment, we were able to determine whether either the breadth or the magnitude of HIV-specific IFN- secretion correlated with the subsequent rate of CD4+-T-cell decline. No statistically significant correlation was observed between clinical parameters, such as the rate of CD4+-T-cell decline or viral load, and the breadth or the magnitude of the immune responses to HIV (Fig. 2A to D) (P values were NS for all correlations; Pearson's correlation coefficient). Furthermore, no significant correlations were found between these clinical parameters and the breadth or magnitude of responses directed to individual HIV gene products (data not shown).

    Analysis of HIV-specific IFN--secreting responses in three groups stratified by rate of CD4 decline. Additional comparisons were made among three subgroups of the study population, which exhibited different rates of CD4+-T-cell loss. No differences among the groups in the magnitude or breadth of HIV-specific responses to all expressed HIV genes or to individual HIV gene products were detected (P values were NS for all comparisons; ANOVA) (Fig. 3A and B).

    DISCUSSION

    We present here data on total HIV-specific immune responses measured by IFN- ELISPOT for 31 treatment-na?ve subjects in the chronic phase of infection for whom a median of 24 (range, 12 to 24) months of follow-up information is available. We found that neither the breadth nor the magnitude of HIV-specific responses correlated with viral loads or with CD4+- or CD8+-T-cell counts at the time points tested. All subjects remained untreated for up to 24 months from the time they were screened for immune responses, during which monthly CD4+-T-cell counts were assessed. We used these serial CD4+-T-cell counts to calculate the rate of CD4+-T-cell loss during the 12 to 24 months immediately after the immune screening. The slope of CD4+-T-cell decline is a powerful surrogate marker for the prediction of disease progression rates (43). We found that neither the breadth nor the magnitude of HIV-specific IFN- secretion correlated with the rate of CD4+-T-cell decline. Slow, typical, and rapid progressors were selected from the study population based on rates of CD4+-T-cell loss and compared by the above immune response parameters. No significant differences among the groups with respect to breadth or magnitude of HIV-specific IFN- secretion were seen, suggesting that this function of HIV-specific cells did not play a strong role in modulating the rate of disease progression.

    The rate of disease progression in individuals infected with HIV-1 is highly variable. The median time from infection to AIDS is 8 to 10 years, and the average annual decline in CD4+-T-cell count is 60 cells/μl/year (10, 22). However, time to AIDS can vary from as few as 2 to 3 years to 15 or more years (10). Previous studies have demonstrated the prognostic value of plasma viral loads, absolute CD4+-T-cell counts, and immune activation as predictors of HIV disease progression rate (42, 43, 48). However, the use of HIV-specific immune responses as prognostic markers for disease outcome has not been addressed in the past.

    At the time the HIV-infected population studied here was assessed for HIV-specific immune responses, the subjects were fairly homogeneous and healthy in terms of CD4 count and viral load. Only five subjects had CD4 counts below 500, and four had viral loads of over 10,000 HIV copies/ml of plasma. Despite these characteristics, we were able to identify three subpopulations based on the rates of CD4+-T-cell loss; these subpopulations did not differ from one other in CD4 counts and viral loads at the initial immune response sampling date. In this population, the effects of viral load set point, CD4 count, and antiretroviral therapy on disease progression were minimized. Should the breadth and magnitude of HIV-specific IFN- secretion have an effect on CD4+-T-cell decline, it would be expected to be easier to detect in such a population.

    CTLs specific for MHC class I restricted viral epitopes are thought to be crucial for controlling several viral infections, including HIV (8, 35, 36, 44, 56, 57, 59, 68). The evidence for this idea comes from SIV-infected macaque models, where depletion of CD8+ T cells results in uncontrolled viremia. In addition, mutation in CTL epitopes leads to escape from CTL control and vaccination strategies that induce CTL slow disease progression once macaques become infected with a pathogenic challenge virus (2, 3, 5, 6, 31, 59, 60). The temporal association between induction of CTLs and reduction in viral load in primary infection and the observation that disease progression rates are associated with certain MHC class I alleles or grouping of these alleles into supertypes supports a role for CTLs in controlling HIV in humans (8, 32, 35, 62). However, CD8+ T cells have multiple antiviral functions, including CTL activity and secretion of factors such as IFN-, chemokines, and other molecules that modulate HIV spread (4, 30, 38, 65). The relationship between lytic activity and IFN- secretion was established at the level of single cells that secrete IFN- upon antigen stimulation and also develop into cells with lytic activity (16). Other studies have also reported associations between these two functions, which led to the adoption of the IFN- ELISPOT assay as a high-throughput quantitative methodology for enumerating and characterizing HIV-specific effector activity (25). However, the correlation between lytic and IFN--secreting functions of CD8+ T cells has been questioned recently. The distribution of HIV-specific memory T-cell subsets in HIV-infected individuals is skewed compared to those recognizing non-HIV antigens, such as cytomegalovirus. The development of terminally differentiated CD45RA+ CCR7– effector cells appears to be blocked at the CD45RA– CCR7– memory effector stage. Although cells in both of these memory compartments secrete IFN-, only the terminally differentiated cells produce high levels of perforin, a marker for cells with lytic capacity (13). The discordance between IFN- secretion and the ability to lyse HIV-infected target cells efficiently in preterminally differentiated memory cells, which are the predominant effector population in untreated chronically HIV-infected subjects, may contribute to the lack of an association between the breadth and magnitude of HIV-specific IFN- secretion and rate of CD4+-T-cell loss. This conclusion is further supported by the observation that slow progressors do not have higher and broader HIV-specific IFN--secreting responses than do typical or rapid progressors.

    Because assays measuring lytic activity are cumbersome and do not lend themselves to screening for responses to large numbers of peptides in a high-throughput format, such as an IFN- ELISPOT assay, quantitative measures of lytic activity have not been done on large study populations. Although HIV-specific CTLs are believed to control viremia, IFN- secretion may not be the most potent mechanism through which this control is mediated. During chronic viral infections, such as lymphocytic choriomeningitis virus infection in mice, an antigen dose-driven gradual impairment of T-cell function occurs. Secretion of effector molecules, such as IL-2, TNF-, and IFN-, is progressively lost, with IFN- being the most resistant to exhaustion (63, 64). Lichterfeld et al. have shown that HIV-specific cytotoxicity is best mediated by a subset of CD8+ T cells which maintains secretion of both TNF- and IFN- (39). Furthermore, several studies have suggested that maintenance of HIV-specific IL-2 secretion and proliferation are associated with viral control in long-term nonprogressors and individuals who start effective highly active antiretroviral therapy in early infection (29, 49, 66). Since IFN- secretion is the function of CD8+ T cells that is maintained longest in the context of HIV viremia, it is likely to detect the broadest range of peptide-specific responses. However, as shown here, the breadth and magnitude of HIV-specific IFN- secretion is not associated with viral control and disease progression.

    Others have used a similar comprehensive screening approach to test for HIV-specific IFN- secretion in HIV-infected individuals (1, 7, 15, 19, 45, 51). These studies were ideal for obtaining a global picture of immune responses to HIV recognized in Caucasian and non-Caucasian populations infected with clade B or C virus. They were also suited to identifying the regions of HIV that are targeted in the largest cross-section of the populations studied and the features of HIV sequence that are likely to harbor immunogenic epitopes (19, 67). Although data on viral load and CD4+-T-cell counts were presented at the time point studied, follow-up information was not available. Therefore, no conclusion could be drawn as to the predictive power of HIV-specific IFN- secretion for disease progression. Furthermore, any correlations observed between immune responses and viral load cannot be used to infer a relationship between these immune responses and the rate of disease progression, as viral load set point is only one factor that contributes to HIV disease progression. Both direct viral cytopathic effects and indirect effects of viral replication, such as immune activation, contribute to the rate of CD4+-T-cell decline and immunologic progression (26). Studies of two natural hosts of SIV, sooty mangabeys and African green monkeys, illustrate the dichotomy between viral load set point and disease progression. Despite high viremia, they exhibit low levels of immune activation and do not progressively lose CD4+ T cells (9). Additional observations of patients infected with HIV-1 and HIV-2 have been made. Patients infected with either virus displayed similar degrees of CD4 depletion and immune activation despite significant differences in plasma viral load (61). Taken together, these results show that viremia contributes only partially to the subsequent degree of CD4+-T-cell loss.

    Our results confirm those generated by others which showed that HIV Gag and Nef are the most frequently targeted HIV gene products in chronically infected subjects (1, 15, 19). Furthermore, similar to the results of these studies, we did not find a correlation between the detected immune responses and viral load. In one study, longitudinal data for clinical follow-up was available and immune response testing was performed at the end of the follow-up period. In this case, conclusions were drawn on the combined effect of the rate of CD4+-T-cell decline and viral load on the breadth and magnitude of HIV-specific IFN- secretion (54). The present report is the first that comprehensively screened for HIV-specific IFN- responses in a group of chronically infected, treatment-na?ve subjects monitored for up to 24 months after determination of immune response parameters. We conclude that although HIV-specific IFN--secreting responses are present in all of those tested, neither their breadth nor their magnitude correlates with viral control or the rate of CD4+-T-cell decline.

    ACKNOWLEDGMENTS

    We thank the study participants. In addition, we would like to thank the study coordinator, G. Deutsch; the laboratory personnel, Alefia Merchant and Rebecca Mullan, for HLA typing; L. Gilbert for performing cell surface phenotyping and data management; George Makedonas for critical review of the manuscript; and M. P. Johnson at Chiron Corp. for viral load assays.

    This work was supported by the Canadian Instit utes for Health Research grant HOP 15573, CANFAR grant 01 4 - 0 07, and the Fonds de Recherche en Santé du Québec AIDS and Infectious Diseases Network.

    REFERENCES

    Addo, M. M., X. G. Yu, A. Rathod, D. Cohen, R. L. Eldridge, D. Strick, M. N. Johnston, C. Corcoran, A. G. Wurcel, C. A. Fitzpatrick, M. E. Feeney, W. R. Rodriguez, N. Basgoz, R. Draenert, D. R. Stone, C. Brander, P. J. Goulder, E. S. Rosenberg, M. Altfeld, and B. D. Walker. 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.

    Allen, T. M., D. H. O'Connor, P. Jing, J. L. Dzuris, B. R. Mothe, T. U. Vogel, E. Dunphy, M. E. Liebl, C. Emerson, N. Wilson, K. J. Kunstman, X. Wang, D. B. Allison, A. L. Hughes, R. C. Desrosiers, J. D. Altman, S. M. Wolinsky, A. Sette, and D. I. Watkins. 2000. Tat-specific cytotoxic T lymphocytes select for SIV escape variants during resolution of primary viraemia. Nature 407:386-390.

    Amara, R. R., F. Villinger, J. D. Altman, S. L. Lydy, S. P. O'Neil, S. I. Staprans, D. C. Montefiori, Y. Xu, J. G. Herndon, L. S. Wyatt, M. A. Candido, N. L. Kozyr, P. L. Earl, J. M. Smith, H. L. Ma, B. D. Grimm, M. L. Hulsey, J. Miller, H. M. McClure, J. M. McNicholl, B. Moss, and H. L. Robinson. 2001. Control of a mucosal challenge and prevention of AIDS by a multiprotein DNA/MVA vaccine. Science 292:69-74.

    Appay, V., D. F. Nixon, S. M. Donahoe, G. M. Gillespie, T. Dong, A. King, G. S. Ogg, H. M. Spiegel, C. Conlon, C. A. Spina, D. V. Havlir, D. D. Richman, A. Waters, P. Easterbrook, A. J. McMichael, and S. L. Rowland-Jones. 2000. HIV-specific CD8+ T cells produce antiviral cytokines but are impaired in cytolytic function. J. Exp. Med. 192:63-75.

    Barouch, D. H., J. Kunstman, M. J. Kuroda, J. E. Schmitz, S. Santra, F. W. Peyerl, G. R. Krivulka, K. Beaudry, M. A. Lifton, D. A. Gorgone, D. C. Montefiori, M. G. Lewis, S. M. Wolinsky, and N. L. Letvin. 2002. Eventual AIDS vaccine failure in a rhesus monkey by viral escape from cytotoxic T lymphocytes. Nature 415:335-339.

    Barouch, D. H., S. Santra, J. E. Schmitz, M. J. Kuroda, T. M. Fu, W. Wagner, M. Bilska, A. Craiu, X. X. Zheng, G. R. Krivulka, K. Beaudry, M. A. Lifton, C. E. Nickerson, W. L. Trigona, K. Punt, D. C. Freed, L. Guan, S. Dubey, D. Casimiro, A. Simon, M. E. Davies, M. Chastain, T. B. Strom, R. S. Gelman, D. C. Montefiori, M. G. Lewis, E. A. Emini, J. W. Shiver, and N. L. Letvin. 2000. Control of viremia and prevention of clinical AIDS in rhesus monkeys by cytokine-augmented DNA vaccination. Science 290:486-492.

    Betts, M. R., D. R. Ambrozak, D. C. Douek, S. Bonhoeffer, J. M. Brenchley, J. P. Casazza, R. A. Koup, and L. J. Picker. 2001. Analysis of total human immunodeficiency virus (HIV)-specific CD4+ and CD8+ T-cell responses: relationship to viral load in untreated HIV infection. J. Virol. 75:11983-11991.

    Borrow, P., H. Lewicki, B. H. Hahn, G. M. Shaw, and M. B. Oldstone. 1994. Virus-specific CD8+ cytotoxic T-lymphocyte activity associated with control of viremia in primary human immunodeficiency virus type 1 infection. J. Virol. 68:6103-6110.

    Broussard, S. R., S. I. Staprans, R. White, E. M. Whitehead, M. B. Feinberg, and J. S. Allan. 2001. Simian immunodeficiency virus replicates to high levels in naturally infected African green monkeys without inducing immunologic or neurologic disease. J. Virol. 75:2262-2275.

    Buchbinder, S., and E. Vittinghoff. 1999. HIV-infected long-term nonprogressors: epidemiology, mechanisms of delayed progression, and clinical and research implications. Microbes Infect. 1:1113-1120.

    Bunce, M. 2003. PCR-sequence-specific primer typing of HLA class I and class II alleles. Methods Mol. Biol. 210:143-171.

    Cao, J., J. McNevin, S. Holte, L. Fink, L. Corey, and M. J. McElrath. 2003. Comprehensive analysis of human immunodeficiency virus type 1 (HIV-1)-specific gamma interferon-secreting CD8+ T cells in primary HIV-1 infection. J. Virol. 77:6867-6878.

    Champagne, P., G. S. Ogg, A. S. King, C. Knabenhans, K. Ellefsen, M. Nobile, V. Appay, G. P. Rizzardi, S. Fleury, M. Lipp, R. Forster, S. Rowland-Jones, R. P. Sekaly, A. J. McMichael, and G. Pantaleo. 2001. Skewed maturation of memory HIV-specific CD8 T lymphocytes. Nature 410:106-111.

    Currier, J. R., E. G. Kuta, E. Turk, L. B. Earhart, L. Loomis-Price, S. Janetzki, G. Ferrari, D. L. Birx, and J. H. Cox. 2002. A panel of MHC class I restricted viral peptides for use as a quality control for vaccine trial ELISPOT assays. J. Immunol. Methods 260:157-172.

    Draenert, R., C. L. Verrill, Y. Tang, T. M. Allen, A. G. Wurcel, M. Boczanowski, A. Lechner, A. Y. Kim, T. Suscovich, N. V. Brown, M. M. Addo, and B. D. Walker. 2004. Persistent recognition of autologous virus by high-avidity CD8 T cells in chronic, progressive human immunodeficiency virus type 1 infection. J. Virol. 78:630-641.

    Dunbar, P. R., G. S. Ogg, J. Chen, N. Rust, P. van der Bruggen, and V. Cerundolo. 1998. Direct isolation, phenotyping and cloning of low-frequency antigen-specific cytotoxic T lymphocytes from peripheral blood. Curr. Biol. 8:413-416.

    Edwards, B. H., A. Bansal, S. Sabbaj, J. Bakari, M. J. Mulligan, and P. A. Goepfert. 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.

    Finkel, T. H., G. Tudor-Williams, N. K. Banda, M. F. Cotton, T. Curiel, C. Monks, T. W. Baba, R. M. Ruprecht, and A. Kupfer. 1995. Apoptosis occurs predominantly in bystander cells and not in productively infected cells of HIV- and SIV-infected lymph nodes. Nat. Med. 1:129-134.

    Frahm, N., B. T. Korber, C. M. Adams, J. J. Szinger, R. Draenert, M. M. Addo, M. E. Feeney, K. Yusim, K. Sango, N. V. Brown, D. SenGupta, A. Piechocka-Trocha, T. Simonis, F. M. Marincola, A. G. Wurcel, D. R. Stone, C. J. Russell, P. Adolf, D. Cohen, T. Roach, A. StJohn, A. Khatri, K. Davis, J. Mullins, P. J. Goulder, B. D. Walker, and C. Brander. 2004. Consistent cytotoxic-T-lymphocyte targeting of immunodominant regions in human immunodeficiency virus across multiple ethnicities. J. Virol. 78:2187-2200.

    Gandhi, R. T., B. K. Chen, S. E. Straus, J. K. Dale, M. J. Lenardo, and D. Baltimore. 1998. HIV-1 directly kills CD4+ T cells by a Fas-independent mechanism. J. Exp. Med. 187:1113-1122.

    Giorgi, J. V., R. H. Lyles, J. L. Matud, T. E. Yamashita, J. W. Mellors, L. E. Hultin, B. D. Jamieson, J. B. Margolick, C. R. Rinaldo, Jr., J. P. Phair, and R. Detels. 2002. Predictive value of immunologic and virologic markers after long or short duration of HIV-1 infection. J. Acquir. Immune Defic. Syndr. 29:346-355.

    Goedert, J. J., C. M. Kessler, L. M. Aledort, R. J. Biggar, W. A. Andes, G. C. White, J. E. Drummond, K. Vaidya, D. L. Mann, M. E. Eyster, et al. 1989. A prospective study of human immunodeficiency virus type 1 infection and the development of AIDS in subjects with hemophilia. N. Engl. J. Med. 321:1141-1148.

    Goulder, P. J., C. Brander, Y. Tang, C. Tremblay, R. A. Colbert, M. M. Addo, E. S. Rosenberg, T. Nguyen, R. Allen, A. Trocha, M. Altfeld, S. He, M. Bunce, R. Funkhouser, S. I. Pelton, S. K. Burchett, K. McIntosh, B. T. Korber, and B. D. Walker. 2001. Evolution and transmission of stable CTL escape mutations in HIV infection. Nature 412:334-338.

    Goulder, P. J., R. E. Phillips, R. A. Colbert, S. McAdam, G. Ogg, M. A. Nowak, P. Giangrande, G. Luzzi, B. Morgan, A. Edwards, A. J. McMichael, and S. Rowland-Jones. 1997. Late escape from an immunodominant cytotoxic T-lymphocyte response associated with progression to AIDS. Nat. Med. 3:212-217.

    Goulder, P. J., Y. Tang, C. Brander, M. R. Betts, M. Altfeld, K. Annamalai, A. Trocha, S. He, E. S. Rosenberg, G. Ogg, C. A. O'Callaghan, S. A. Kalams, R. E. McKinney, Jr., K. Mayer, R. A. Koup, S. I. Pelton, S. K. Burchett, K. McIntosh, and B. D. Walker. 2000. Functionally inert HIV-specific cytotoxic T lymphocytes do not play a major role in chronically infected adults and children. J. Exp. Med. 192:1819-1832.

    Grossman, Z., M. Meier-Schellersheim, A. E. Sousa, R. M. Victorino, and W. E. Paul. 2002. CD4+ T-cell depletion in HIV infection: are we closer to understanding the cause? Nat. Med. 8:319-323.

    Harrer, T., E. Harrer, S. A. Kalams, T. Elbeik, S. I. Staprans, M. B. Feinberg, Y. Cao, D. D. Ho, T. Yilma, A. M. Caliendo, R. P. Johnson, S. P. Buchbinder, and B. D. Walker. 1996. Strong cytotoxic T cell and weak neutralizing antibody responses in a subset of persons with stable nonprogressing HIV type 1 infection. AIDS Res. Hum. Retrovir. 12:585-592.

    Hazenberg, M. D., D. Hamann, H. Schuitemaker, and F. Miedema. 2000. T cell depletion in HIV-1 infection: how CD4+ T cells go out of stock. Nat. Immunol. 1:285-289.

    Iyasere, C., J. C. Tilton, A. J. Johnson, S. Younes, B. Yassine-Diab, R. P. Sekaly, W. W. Kwok, S. A. Migueles, A. C. Laborico, W. L. Shupert, C. W. Hallahan, R. T. Davey, Jr., M. Dybul, S. Vogel, J. Metcalf, and M. Connors. 2003. Diminished proliferation of human immunodeficiency virus-specific CD4+ T cells is associated with diminished interleukin-2 (IL-2) production and is recovered by exogenous IL-2. J. Virol. 77:10900-10909.

    Jassoy, C., T. Harrer, T. Rosenthal, B. A. Navia, J. Worth, R. P. Johnson, and B. D. Walker. 1993. Human immunodeficiency virus type 1-specific cytotoxic T lymphocytes release gamma interferon, tumor necrosis factor alpha (TNF-), and TNF-? when they encounter their target antigens. J. Virol. 67:2844-2852.

    Jin, X., D. E. Bauer, S. E. Tuttleton, S. Lewin, A. Gettie, J. Blanchard, C. E. Irwin, J. T. Safrit, J. Mittler, L. Weinberger, L. G. Kostrikis, L. Zhang, A. S. Perelson, and D. D. Ho. 1999. Dramatic rise in plasma viremia after CD8+ T cell depletion in simian immunodeficiency virus-infected macaques. J. Exp. Med. 189:991-998.

    Kaslow, R. A., M. Carrington, R. Apple, L. Park, A. Mu?oz, A. J. Saah, J. J. Goedert, C. Winkler, S. J. O'Brien, C. Rinaldo, R. Detels, W. Blattner, J. Phair, H. Erlich, and D. L. Mann. 1996. Influence of combinations of human major histocompatibility complex genes on the course of HIV-1 infection. Nat. Med. 2:405-411.

    Kaur, A., L. Alexander, S. I. Staprans, L. Denekamp, C. L. Hale, H. M. McClure, M. B. Feinberg, R. C. Desrosiers, and R. P. Johnson. 2001. Emergence of cytotoxic T lymphocyte escape mutations in nonpathogenic simian immunodeficiency virus infection. Eur. J. Immunol. 31:3207-3217.

    Kelleher, A. D., C. Long, E. C. Holmes, R. L. Allen, J. Wilson, C. Conlon, C. Workman, S. Shaunak, K. Olson, P. Goulder, C. Brander, G. Ogg, J. S. Sullivan, W. Dyer, I. Jones, A. J. McMichael, S. Rowland-Jones, and R. E. Phillips. 2001. Clustered mutations in HIV-1 gag are consistently required for escape from HLA-B27-restricted cytotoxic T lymphocyte responses. J. Exp. Med. 193:375-386.

    Koup, R. A., J. T. Safrit, Y. Cao, C. A. Andrews, G. McLeod, W. Borkowsky, C. Farthing, and D. D. Ho. 1994. Temporal association of cellular immune responses with the initial control of viremia in primary human immunodeficiency virus type 1 syndrome. J. Virol. 68:4650-4655.

    Lechner, F., D. K. Wong, P. R. Dunbar, R. Chapman, R. T. Chung, P. Dohrenwend, G. Robbins, R. Phillips, P. Klenerman, and B. D. Walker. 2000. Analysis of successful immune responses in persons infected with hepatitis C virus. J. Exp. Med. 191:1499-1512.

    Leng, Q., G. Borkow, Z. Weisman, M. Stein, A. Kalinkovich, and Z. Bentwich. 2001. Immune activation correlates better than HIV plasma viral load with CD4 T-cell decline during HIV infection. J. Acquir. Immune Defic. Syndr. 27:389-397.

    Levy, J. A. 2003. The search for the CD8+ cell anti-HIV factor (CAF). Trends Immunol. 24:628-632.

    Lichterfeld, M., X. G. Yu, M. T. Waring, S. K. Mui, M. N. Johnston, D. Cohen, M. M. Addo, J. Zaunders, G. Alter, E. Pae, D. Strick, T. M. Allen, E. S. Rosenberg, B. D. Walker, and M. Altfeld. 2004. HIV-1-specific cytotoxicity is preferentially mediated by a subset of CD8+ T cells producing both interferon- and tumor necrosis factor-. Blood 104:487-494.

    Lieberman, J., P. Shankar, N. Manjunath, and J. Andersson. 2001. Dressed to kill? A review of why antiviral CD8 T lymphocytes fail to prevent progressive immunodeficiency in HIV-1 infection. Blood 98:1667-1677.

    Liu, Z., W. G. Cumberland, L. E. Hultin, A. H. Kaplan, R. Detels, and J. V. Giorgi. 1998. CD8+ T-lymphocyte activation in HIV-1 disease reflects an aspect of pathogenesis distinct from viral burden and immunodeficiency. J. Acquir. Immune Defic. Syndr. Hum. Retrovir. 18:332-340.

    Liu, Z., W. G. Cumberland, L. E. Hultin, H. E. Prince, R. Detels, and J. V. Giorgi. 1997. Elevated CD38 antigen expression on CD8+ T cells is a stronger marker for the risk of chronic HIV disease progression to AIDS and death in the Multicenter AIDS Cohort Study than CD4+ cell count, soluble immune activation markers, or combinations of HLA-DR and CD38 expression. J. Acquir. Immune Defic. Syndr. Hum. Retrovir. 16:83-92.

    Lyles, R. H., A. Mu?oz, T. E. Yamashita, H. Bazmi, R. Detels, C. R. Rinaldo, J. B. Margolick, J. P. Phair, and J. W. Mellors, et al. 2000. Natural history of human immunodeficiency virus type 1 viremia after seroconversion and proximal to AIDS in a large cohort of homosexual men. J. Infect. Dis. 181:872-880.

    Maini, M. K., C. Boni, G. S. Ogg, A. S. King, S. Reignat, C. K. Lee, J. R. Larrubia, G. J. Webster, A. J. McMichael, C. Ferrari, R. Williams, D. Vergani, and A. Bertoletti. 1999. Direct ex vivo analysis of hepatitis B virus-specific CD8+ T cells associated with the control of infection. Gastroenterology 117:1386-1396.

    Masemola, A., T. Mashishi, G. Khoury, P. Mohube, P. Mokgotho, E. Vardas, M. Colvin, L. Zijenah, D. Katzenstein, R. Musonda, S. Allen, N. Kumwenda, T. Taha, G. Gray, J. McIntyre, S. A. Karim, H. W. Sheppard, and C. M. Gray. 2004. Hierarchical targeting of subtype C human immunodeficiency virus type 1 proteins by CD8+ T cells: correlation with viral load. J. Virol. 78:3233-3243.

    McCune, J. M. 2001. The dynamics of CD4+ T-cell depletion in HIV disease. Nature 410:974-979.

    Mellors, J. W., A. Mu?oz, J. V. Giorgi, J. B. Margolick, C. J. Tassoni, P. Gupta, L. A. Kingsley, J. A. Todd, A. J. Saah, R. Detels, J. P. Phair, and C. R. Rinaldo, Jr. 1997. Plasma viral load and CD4+ lymphocytes as prognostic markers of HIV-1 infection. Ann. Intern. Med. 126:946-954.

    Mellors, J. W., C. R. Rinaldo, Jr., P. Gupta, R. M. White, J. A. Todd, and L. A. Kingsley. 1996. Prognosis in HIV-1 infection predicted by the quantity of virus in plasma. Science 272:1167-1170.

    Migueles, S. A., A. C. Laborico, W. L. Shupert, M. S. Sabbaghian, R. Rabin, C. W. Hallahan, D. van Baarle, S. Kostense, F. Miedema, M. McLaughlin, L. Ehler, J. Metcalf, S. Liu, and M. Connors. 2002. HIV-specific CD8+ T cell proliferation is coupled to perforin expression and is maintained in nonprogressors. Nat. Immunol. 3:1061-1068.

    Mwau, M., A. J. McMichael, and T. Hanke. 2002. Design and validation of an enzyme-linked immunospot assay for use in clinical trials of candidate HIV vaccines. AIDS Res. Hum. Retrovir. 18:611-618.

    Novitsky, V., P. Gilbert, T. Peter, M. F. McLane, S. Gaolekwe, N. Rybak, I. Thior, T. Ndung'u, R. Marlink, T. H. Lee, and M. Essex. 2003. Association between virus-specific T-cell responses and plasma viral load in human immunodeficiency virus type 1 subtype C infection. J. Virol. 77:882-890.

    O'Connor, D. H., T. M. Allen, T. U. Vogel, P. Jing, I. P. DeSouza, E. Dodds, E. J. Dunphy, C. Melsaether, B. Mothe, H. Yamamoto, H. Horton, N. Wilson, A. L. Hughes, and D. I. Watkins. 2002. Acute phase cytotoxic T lymphocyte escape is a hallmark of simian immunodeficiency virus infection. Nat. Med. 8:493-499.

    Ogg, G. S., X. Jin, S. Bonhoeffer, P. R. Dunbar, M. A. Nowak, S. Monard, J. P. Segal, Y. Cao, S. L. Rowland-Jones, V. Cerundolo, A. Hurley, M. Markowitz, D. D. Ho, D. F. Nixon, and A. J. McMichael. 1998. Quantitation of HIV-1-specific cytotoxic T lymphocytes and plasma load of viral RNA. Science 279:2103-2106.

    Oxenius, A., D. A. Price, M. Hersberger, E. Schlaepfer, R. Weber, M. Weber, T. M. Kundig, J. Boni, H. Joller, R. E. Phillips, M. Flepp, M. Opravil, and R. F. Speck. 2004. HIV-specific cellular immune response is inversely correlated with disease progression as defined by decline of CD4+ T cells in relation to HIV RNA load. J. Infect. Dis. 189:1199-1208.

    Price, D. A., P. J. Goulder, P. Klenerman, A. K. Sewell, P. J. Easterbrook, M. Troop, C. R. Bangham, and R. E. Phillips. 1997. Positive selection of HIV-1 cytotoxic T lymphocyte escape variants during primary infection. Proc. Natl. Acad. Sci. USA 94:1890-1895.

    Rickinson, A. B., and D. J. Moss. 1997. Human cytotoxic T lymphocyte responses to Epstein-Barr virus infection. Annu. Rev. Immunol. 15:405-431.

    Riddell, S. R., K. S. Watanabe, J. M. Goodrich, C. R. Li, M. E. Agha, and P. D. Greenberg. 1992. Restoration of viral immunity in immunodeficient humans by the adoptive transfer of T cell clones. Science 257:238-241.

    Russell, N. D., M. G. Hudgens, R. Ha, C. Havenar-Daughton, and M. J. McElrath. 2003. Moving to human immunodeficiency virus type 1 vaccine efficacy trials: defining T cell responses as potential correlates of immunity. J. Infect. Dis. 187:226-242.

    Schmitz, J. E., M. J. Kuroda, S. Santra, V. G. Sasseville, M. A. Simon, M. A. Lifton, P. Racz, K. Tenner-Racz, M. Dalesandro, B. J. Scallon, J. Ghrayeb, M. A. Forman, D. C. Montefiori, E. P. Rieber, N. L. Letvin, and K. A. Reimann. 1999. Control of viremia in simian immunodeficiency virus infection by CD8+ lymphocytes. Science 283:857-860.

    Shiver, J. W., T. M. Fu, L. Chen, D. R. Casimiro, M. E. Davies, R. K. Evans, Z. Q. Zhang, A. J. Simon, W. L. Trigona, S. A. Dubey, L. Huang, V. A. Harris, R. S. Long, X. Liang, L. Handt, W. A. Schleif, L. Zhu, D. C. Freed, N. V. Persaud, L. Guan, K. S. Punt, A. Tang, M. Chen, K. A. Wilson, K. B. Collins, G. J. Heidecker, V. R. Fernandez, H. C. Perry, J. G. Joyce, K. M. Grimm, J. C. Cook, P. M. Keller, D. S. Kresock, H. Mach, R. D. Troutman, L. A. Isopi, D. M. Williams, Z. Xu, K. E. Bohannon, D. B. Volkin, D. C. Montefiori, A. Miura, G. R. Krivulka, M. A. Lifton, M. J. Kuroda, J. E. Schmitz, N. L. Letvin, M. J. Caulfield, A. J. Bett, R. Youil, D. C. Kaslow, and E. A. Emini. 2002. Replication-incompetent adenoviral vaccine vector elicits effective anti-immunodeficiency-virus immunity. Nature 415:331-335.

    Sousa, A. E., J. Carneiro, M. Meier-Schellersheim, Z. Grossman, and R. M. Victorino. 2002. CD4 T cell depletion is linked directly to immune activation in the pathogenesis of HIV-1 and HIV-2 but only indirectly to the viral load. J. Immunol. 169:3400-3406.

    Trachtenberg, E., B. Korber, C. Sollars, T. B. Kepler, P. T. Hraber, E. Hayes, R. Funkhouser, M. Fugate, J. Theiler, Y. S. Hsu, K. Kunstman, S. Wu, J. Phair, H. Erlich, and S. Wolinsky. 2003. Advantage of rare HLA supertype in HIV disease progression. Nat. Med. 9:928-935.

    Wherry, E. J., and R. Ahmed. 2004. Memory CD8 T-cell differentiation during viral infection. J. Virol. 78:5535-5545.

    Wherry, E. J., J. N. Blattman, K. Murali-Krishna, R. van der Most, and R. Ahmed. 2003. Viral persistence alters CD8 T-cell immunodominance and tissue distribution and results in distinct stages of functional impairment. J. Virol. 77:4911-4927.

    Yang, O. O., S. A. Kalams, A. Trocha, H. Cao, A. Luster, R. P. Johnson, and B. D. Walker. 1997. Suppression of human immunodeficiency virus type 1 replication by CD8+ cells: evidence for HLA class I-restricted triggering of cytolytic and noncytolytic mechanisms. J. Virol. 71:3120-3128.

    Younes, S. A., B. Yassine-Diab, A. R. Dumont, M. R. Boulassel, Z. Grossman, J. P. Routy, and R. P. Sekaly. 2003. HIV-1 viremia prevents the establishment of interleukin 2-producing HIV-specific memory CD4+ T cells endowed with proliferative capacity. J. Exp. Med. 198:1909-1922.

    Yusim, K., C. Kesmir, B. Gaschen, M. M. Addo, M. Altfeld, S. Brunak, A. Chigaev, V. Detours, and B. T. Korber. 2002. Clustering patterns of cytotoxic T-lymphocyte epitopes in human immunodeficiency virus type 1 (HIV-1) proteins reveal imprints of immune evasion on HIV-1 global variation. J. Virol. 76:8757-8768.

    Zinkernagel, R. M., and H. Hengartner. 1996. Correlates of protective viruses damaging to HIV infection. Science 272:1362.(Yoav Peretz, Galit Alter,)