Literature
Home医源资料库在线期刊传染病学杂志2005年第191卷第12期

Combination of HIV-1Specific CD4 Th1 Cell Responses and IgG2 Antibodies Is the Best Predictor for Persistence of Long-Term Nonprogression

来源:传染病学杂志
摘要:Weprospectivelyfollowed66untreatedlong-termnonprogressorsandanalyzedrelationshipsbetweenHIV-1specificCD4Thelper(Th)1andCD8TcellresponsesandHIV-1specificantibodies,HIV-1RNAandproviralDNAloads,hostgenes,andCD4Th1cellcountsatentryintothestudyand4yearslat......

点击显示 收起

    Laboratoire d'Immunologie Cellulaire et Tissulaire, INSERM U, INSERM EMI
    Laboratoire de Virologie, UPRES EA, Université Pierre et Marie Curie, Hpital Pitié-Salpêtrière
    Laboratoire de Virologie, Hpital Necker-Enfants Malades, Service d'Immuno-Hématologie, Hpital Saint-Louis
    Service de Médecine Interne, Hpital Cochin, Paris, France

    Background.

    Strong T cell and antibody responses to human immunodeficiency virus (HIV), low virus production, and some genetic traits have been individually associated with nonprogression of HIV infection, but the best correlate with protection against disease progression remains unknown.

    Methods.

    We prospectively followed 66 untreated long-term nonprogressors and analyzed relationships between HIV-1specific CD4 T helper (Th) 1 and CD8 T cell responses and HIV-1specific antibodies, HIV-1 RNA and proviral DNA loads, host genes, and CD4 Th1 cell counts at entry into the study and 4 years later.

    Results.

    HIV-1 p24specific CD4 Th1 cell proliferation, interferon (IFN) production, and IFN-producing cell frequencies at entry significantly and negatively correlated with HIV-1 RNA and proviral DNA loads and were independent of CD4 Th1 cell counts and host genes. HIV-1 Gagspecific IFN-producing CD8 T cell frequencies correlated with HIV-1 proviral DNA loads but not with RNA loads. Only high frequencies of HIV-1 p24specific CD4 Th1 cells combined with HIV-1 gp41specific IgG2 antibodies significantly predicted persistence of high CD4 Th1 cell counts.

    Conclusion.

    HIV-1specific CD4 Th1 responses combined with IgG2 antibodies and IFN-producing CD4 Th1 cells are better predictors of long-term nonprogression than are virus parameters, host genes, or HIV-1specific CD4 Th1 or CD8 T cell proliferation.

    Nonprogression (NP) in HIV infection is a rare event characterized by a tight host-virus equilibrium in which there are stable, high CD4 Th1 cell counts and low viral replication in the absence of treatment [16]. Factors put forward to explain this status [35, 79] raise the question of whether long-term nonprogression (LTNP) essentially results from a particular set of genes or whether there is also a benefit from strong immune responses that are independent of these genes. Analysis of this question is relevant for the vast majority of the HIV-infected population, because neither antiretroviral therapy [10] nor the present vaccine strategies [11] can eradicate the virusthey only limit disease progression. Genetic factors that affect disease progression involve (1) HIV variants encoding for defective viruses [8, 12], which are too rare, however, to explain most cases of LTNP [13]; (2) variants of genes encoding for chemokines and chemokine receptors limiting HIV entry [9, 1417]; or (3) some HLA alleles governing immune responses [16, 1820]. A particular set of these host genetic factors can, indeed, help classify two-thirds of the cases of LTNP [16].

    Host immune responses to HIV are central in controlling the infection, as is demonstrated by the viral escape after depletion of CD8 T cells in simian immunodeficiency virusinfected macaques [21], but immune correlates of NP are still unclear. All CD8 T cell functionsincluding cytotoxicity [5, 2325] and production of soluble factors [4], chemokines [22], and cytokineshave been observed in LTNP. The vigorous antiHIV-1 responses by CD4 Th1 cells found in LTNP [2629] support a central role for CD4 Th1 cells in the control of viral replication [30, 31]. The strong frequencies of HIV-1specific CD4 Th1 or CD8 T cells that produce interferon (IFN) found in LTNP appear to be less consistently correlated with control of viral replication [32, 33] than are memory cytotoxic CD8 T cells that have proliferative potential [3436], which raises a question about the contribution of IFN- production to control of viral replication and suggests that control of viral replication might be better associated with the proliferative potential of memory T cells [37]. Similarly, the capacity of HIV-1specific CD4 Th1 cells to proliferate and produce interleukin (IL)2 in association with IFN- was shown to better correlate with NP than was their capacity to produce IFN- alone [3843]. Finally, because CD4 Th1 cells are a preferential target for HIV-1 [44, 45], the strong frequencies of these HIV-1specific cells observed in LTNP might reflect only protection against HIV-1 infection rather than control of viral replication. Therefore, a major step in understanding LTNP would be to determine if these immune responses make an independent contribution from that of the host's genes in the control of viral replication.

    Addressing these questions requires the long-term follow-up of cohorts in which these parameters have been measured at entry. We prospectively measured both HIV-1specific CD4 Th1 and CD8 T cell responses in 66 nonprogressors from the Asymptomatiques à Long Terme (ALT) cohort in which viral characteristics [13, 46], host genes [16, 17], and antibody responses to HIV-1 [47] have been extensively studied and for whom a 4-year prospective follow-up after entry into the study was available. We focused on HIV-1specific CD4 Th1 cell responses evaluated at entry into the study by proliferation of, level of production by, and quantification of IFN-producing CD4 Th1 cells, and we also measured the frequencies of HIV-1 Gagspecific CD8 T cells. A multivariate analysis incorporating the host chemokine/chemoreceptor gene variants, HLA alleles [16], and antiHIV-1 IgG2 antibodies previously associated with NP in this cohort [47] was used to determine which parameter could best predict persistence of stable CD4 Th1 cell counts over time and LTNP.

    PATIENTS, MATERIALS, AND METHODS

    Patients

    A total of 66 HIV-1infected nonprogressors from the ALT cohort [13] for whom an HIV-1specific T cell proliferation and/or IFN- enzyme-linked immunospot (ELISPOT) assay were performed at entry into the cohort were studied. Inclusion criteria in the ALT cohort were HIV-1 seropositivity for at least 8 years, CD4 Th1 cell count >600 cells/mm3 for the preceding 5 years, no clinical symptoms, and no receipt of antiretroviral therapy [13]. Plasma HIV-1 RNA load, which was not routinely available at the time of entry into the study (199495), was not an inclusion criterion. The Pitié-Salpêtrière Hospital Institutional Review Board approved the protocol, and each patient provided written, informed consent. Patient characteristics are shown in table 1. Patients were prospectively followed up through yearly evaluations of CD4 Th1 cell counts and plasma HIV-1 RNA loads. Ten patients were lost to follow-up, and 20 patients initiating antiretroviral therapy in accordance with the criteria recommended in 19962000 [4851] were excluded from follow-up (8, 6, 3, and 3 patients at years 1, 2, 3, and 4, respectively).

    CD4 Th1 Cell Counts

    All absolute CD4 Th1 cell counts were conducted in a single laboratory. Counts were performed on fresh blood by 4-color flow cytometry (Coulter) and were determined in accordance with a standard internal control (mean ± SD reference value, 858 ± 260 cells/mm3).

    Parameters of HIV-1 Production

    HIV-1 RNA.

    HIV-1 virions were quantified in fresh plasma samples by use of the NASBA assay (Organon-Teknika; limit of detection, 800 RNA copies/mL) and the ultrasensitive HIV-1 Amplicor-Monitor assay (Roche-Diagnostic Systems; limit of detection, 20 copies/mL), which, when combined, gave a limit of detection of <800 RNA copies/mL. All tests were conducted in a single laboratory [13].

    HIV-1 proviral DNA.

    The level of HIV-1 proviral DNA was determined in frozen peripheral blood mononuclear cells (PBMCs) by use of a modified Amplicor Monitor assay (Roche Laboratories) with an internal HIV-1 proviral DNA standard provided by S. Kwok [52]. All tests were conducted in a single laboratory [53], and results were expressed as copies of HIV-1 proviral DNA/106 PBMCs.

    HIV-1Specific CD4 Th1 Cell Responses

    An HIV-1 antigenspecific T cell proliferation assay was performed on fresh whole PBMCs from 41 patients after depletion by anti-CD8 monoclonal antibody (MAb) magnetic beads (Dynabeads; Dynal), as described elsewhere [54]. Cells were incubated for 6 days with the recombinant HIV-1 LAI p24 protein (0.25 g/mL; donated by Transgène), which was previously tested in 10 seronegative donors with negative results. Control antigens were purified protein derivative (Serum-Statens Institute) and candidin (Sanofi-Diagnostic-Pasteur) at appropriate concentrations. Positive controls were anti-CD3 and anti-CD28 MAbs (Immunotech). After labeling with tritiated thymidine (CEA), positive responses were defined as >3000 cpm and stimulation index 3 (counts per minute of [cells + stimuli] : [cells + medium]), as described elsewhere [54].

    Cytokine production assay.

    Fresh PBMCs from 37 of these 41 patients were stimulated for cytokine production, as described elsewhere [54, 55], by use of the same HIV-1 LAI p24 protein and anti-CD3 and anti-CD28 MAbs as described above. Culture supernatants were collected at day 2 after incubation, in accordance with previously established kinetics [55], and were cryopreserved before batch analysis with the IL-2 (Immunotech) and IFN- (Endogen) ELISA kits. The cytokine production from CD8-depleted PBMCs showed a persistence of 90% of the cytokine production from nondepleted PBMCs.

    ELISPOT Assay for Quantification of HIV-1Specific IFN-Producing CD4 Th1 Cells

    A sensitized IFN- ELISPOT assay derived from one described elsewhere [36, 54] was performed on frozen PBMCs from 55 nonprogressors with available samples that had a viability after thawing of >85% (Trypan-Blue exclusion). Briefly, recombinant HIV-1 LAI p24 protein (2 g/mL; Protein-Sciences) was added in triplicate wells for a previously optimized 40-h incubation period to ensure maximal specific IFN- release and low background (median, 2 spot-forming cells /well). Depletion of CD4 Th1 cells (Dynabeads; Dynal) abolished 85% of the IFN- production (data not shown). Negative and positive controls were medium alone and phytohemagglutinin (1 g/mL; Murex), respectively. Spots were counted using an ELISPOT reader (Zeiss), and data were expressed as sfc/106 PBMCs. Results were considered to be positive if there were >50 sfc/106 PBMCs after subtraction of the mean background value obtained without incubation with antigen. Counts of HIV-1 p24specific cells were found to be similar in fresh and frozen PBMCs from 5 patients (P = .34).

    Quantification of HIV-1Specific CD8 T Cells by IFN- ELISPOT Assay

    An IFN- ELISPOT assay derived from one described elsewhere [36, 54] was performed on frozen PBMCs from 43 nonprogressors with available samples that had a viability after thawing of >85% (Trypan-Blue exclusion). Cells in triplicate were stimulated by pools of 911 synthetic 15-mer peptides overlapping by 11 aa and spanning the entire HIV-1 gag sequence (each peptide, 2 g/mL; Neosystem). The 18-h incubation had been previously optimized under these conditions. Positive responses were defined as described above. The CD8-depleted PBMCs (anti-CD8 Dynabeads; Dynal) from 20 samples tested against 87 Gag peptide pools yielding positive results eliminated a median of 90% of the spot-forming cells.

    CCR5, CCR2, SDF1, and HLA Typing

    The chemokine receptor/chemokine gene variants (CCR5-32, CCR2-64I, and SDF1-3A) and HLA phenotypes of the ALT cohort have been reported elsewhere [16]. They were used in the statistical analysis and were incorporated into the multivariate analysis.

    Isotype Characterization of Antibodies Directed against HIV-1

    AntiHIV-1 antibody isotypes in the ALT cohort have been described elsewhere [47]. These data were incorporated into the multivariate analysis.

    Statistical Analysis

    Data were analyzed using SPSS software (version 11; SPSS). Associations between continuous variables were tested using the nonparametric Spearman's rank correlation test, the Mann-Whitney U test, or the Kruskal-Wallis test. The 2 test or Fisher's exact test were used for categorical variables.

    To investigate factors independently associated with LTNP (defined as persistent CD4 Th1 cell count >600 cells/mm3 during follow-up), we used time-to-event analysis, a method developed specifically to handle censored data. Thus, patients initiating antiretroviral therapy or lost to follow-up were considered to be censored at the time of the last evaluation before these events. We prepared univariate Cox models of time to progression, and the characteristics significantly associated (P < .20) with control of viral replication at entry into the study were entered into a multivariate backward Cox model. Continuous variables were either entered as continuous variables or converted to categorical variables. We used tertiles to test whether associations were linear (continuous model), because 3 classes represent the minimum number that can detect a deviation from linearity in limited sample sizes [56]. For example, the tertiles for HIV-1 p24specific CD4 Th1 cells were 040, 40170, and >170 sfc/106 PBMCs, with the latter tertile defining high counts of HIV-1specific CD4 Th1 cells. The variables that had the best likelihoods of an association with LTNP were used in later analyses. In addition to the parameters tested in the present study, we incorporated the chemokine/chemoreceptor gene variants and HLA alleles (A3, B5, B12, B14, B17, B27, DR6, and DR7) in this cohort that were shown elsewhere to be significantly associated (P < .20) with nonprogressors [16], when their effect in progressors was compared. Furthermore, the IgG2 antibodies against HIV-1 p55, p24, p68, gp160, gp120, and gp41 that were shown to be significantly associated with plasma HIV-1 RNA loads at entry into the study [47] were entered in a first backward multivariate analysis showing that only anti-gp41 IgG2 antibody was an independent predictor of NP (P < .05). A second backward multivariate model was built that included anti-gp41 IgG2 antibody and the other variables retained in univariate analyses. Life-table estimators, adapted for once-yearly follow-up, were used to assess proportions of LTNP at various time points in the subgroups defined on the basis of the factors independently associated with LTNP.

    RESULTS

    HIV-1 p24specific CD4 Th1 cell proliferation and IFN- production in association with low plasma HIV-1 RNA and cell-associated HIV-1 proviral DNA loads.

    Lymphoproliferative responses to HIV-1 p24 were tested on fresh samples obtained at entry into the study from 41 patients (median CD4 Th1 cell count, 676 cells/mm3 [interquartile range {IQR}, 525846 cells/mm3]; median HIV-1 RNA load, 4200 copies/mL [IQR, 19035,000 copies/mL]). Responses were detected in whole and CD8-depleted PBMCs from 6 subjects (15%) and 13 subjects (32%), respectively. These responses were independent of CD4 Th1 cell counts (r = 0.121; P = .45) (figure 1A) but were negatively correlated with plasma HIV-1 RNA loads (r = -0.396; P = .01) (figure 1B) and cell-associated HIV-1 proviral DNA loads (r = -0.363; P = .02) measured simultaneously. The proliferative responses against tuberculin and candidin, which were observed in higher proportions of samples (87% and 43%, respectively, in whole PBMCs), were not correlated with HIV-1 RNA or proviral DNA loads (data not shown).

    The levels of IFN- and IL-2 directed against HIV-1 p24 were simultaneously quantified in culture supernatants from 37 of these 41 patients, and a 10-fold higher level of IFN- than of IL-2 (median, 92 vs. 7 pg/mL) was observed. HIV-1 p24specific IFN- production, but not IL-2 production, was negatively correlated with plasma HIV-1 RNA loads (r = -0.583; P = .0001) (figure 2A) and HIV-1 proviral DNA loads (r = -0.529; P = .001) (figure 2B), but it was not negatively correlated with CD4 Th1 cell counts (r = 0.13; P = .44) measured at entry into the study. In contrast, IL-2 production, but not IFN- production, was positively correlated with the HIV-1 p24specific proliferative responses (r = 0.41; P = .014). The 5 patients with both IL-2 and IFN- production had a 25-fold lower median plasma HIV-1 RNA load (median, 187 vs. 3300 copies/mL) than the 27 patients with IFN- production only, although the difference was not statistically significant.

    To evaluate whether these correlations between responses to HIV-1 p24 and virus parameters might reflect protection of HIV-1specific CD4 Th1 cells against HIV-1 entry, we studied their relationship with the chemokine/chemoreceptor gene variants previously linked to this cohortthe CCR5-32 deletion and the CCR2-64I and SDF1-3A mutations [16]but found no association (data not shown). Altogether, these data show that the capacity of CD4 Th1 cells to proliferate and produce IFN- against HIV-1 is strongly associated with low HIV-1 RNA and proviral DNA loads and is independent of CD4 Th1 cell counts and genetic resistance to virus entry.

    HIV-1 p24specific IFN-producing CD4 Th1 cells and control of the plasma HIV-1 RNA and cell-associated HIV-1 proviral DNA loads.

    The HIV-1 p24specific IFN-producing CD4 Th1 cells were evaluated at entry into the study by ELISPOT on frozen PBMCs from 55 nonprogressors (median CD4 Th1 cell count, 679 cells/mm3 [IQR, 511812 cells/mm3]; median HIV-1 RNA load, 6600 copies/mL [IQR, 35036,000 copies/mL]) and were detected in 31 subjects (56%) with a median frequency of 188 sfc/106 PBMCs (IQR, 103317 sfc/106 PBMCs) and a tertile distribution of 0, 40, and 170 sfc/106 PBMCs. These frequencies were negatively correlated with the plasma HIV-1 RNA loads (r = -0.304; P = .026) (figure 2C) and the cell-associated HIV-1 proviral DNA loads (r = -0.461; P = .0001) (figure 2D) but not with the CD4 Th1 cell counts (r = 0.215; P = .12) measured at entry into the study.

    We further evaluated the relationship between viral parameters and combined proliferation and IFN- production in the 32 patients in whom ELISPOT and proliferation assays against HIV-1 p24 could be performed in parallel. Significantly lower HIV-1 proviral DNA loadsbut not plasma HIV-1 RNA loads, although these were lowerwere observed in the 7 patients with both proliferative responses and strong frequencies of IFN-producing cells, compared with those in the 15 patients with either proliferative responses or strong frequencies of IFN-producing cells and with those in the 10 patients with no responses (table 2). Altogether, these data show a stronger correlation between frequencies of HIV-1specific CD4 Th1 cells and frequencies of cells harboring HIV-1 proviral DNA than between frequencies of HIV-1specific CD4 Th1 cells and plasma HIV-1 RNA loads, and they also show that this association is reinforced by the presence of CD4 Th1 cell proliferative responses to HIV-1, which are independent of CD4 Th1 cell counts, at entry into the study.

    HIV-1 Gagspecific CD8 T cells and cell-associated HIV-1 proviral DNA.

    The HIV-1 Gagspecific IFN-producing CD8 T cells were quantified at entry into the study by ELISPOT on 43 available samples of frozen PBMCs (median CD4 Th1 cell count, 676 cells/mm3 [IQR, 508790 cells/mm3]; median HIV-1 RNA load, 6600 copies/mL [IQR, 35056,000 copies/mL]). We used pools of 15-mer synthetic peptides overlapping by 11 aa and spanning the entire HIV-1 gag sequence. High frequencies of HIV-1 Gagspecific CD8 T cells were observed (median, 2986 sfc/106 PBMCs [IQR, 14764663 sfc/106 PBMCs]) that significantly correlated with frequencies of HIV-1 p24specific CD4 Th1 cells (r = 0.439; P = .005) (figure 3A) but not with CD4 Th1 cell counts at entry into the study. In contrast to the CD4 Th1 cell reactivity to HIV-1 p24 that correlated with both plasma and cell-associated HIV-1 RNA loads, the frequencies of HIV-1 Gagspecific CD8 T cells did not correlate with plasma HIV-1 RNA loads (data not shown) but did correlate with cell-associated HIV-1 proviral DNA loads (r = -0.383; P = .009) (figure 3B) measured simultaneously at entry into the study.

    HIV-1 p24specific CD4 Th1 cell responses and LTNP.

    Finally, we sought to identify which of the above parameters related to control of viral replication after a median 9 years of HIV-1 seropositivity could predict persistence of LTNP status as assessed by persistence of entry criteriathat is, CD4 Th1 cell count >600 cells/mm3over the course of 4 years of follow-up.

    Neither HIV-1 p24specific proliferative responses at entry into the study nor frequencies of HIV-1specific IFN-producing CD8 T cells were predictive of persistence of LTNP. We then evaluated the predictive value of the other variables associated with NP ([16, 47] and see Patients, Materials, and Methods) in the 53 patients with HIV-1 p24 ELISPOT entry data and follow-up data available (median follow-up, 21 months [IQR, 1248 months]). Only HIV-1 p24specific CD4 Th1 cell counts >170 sfc/106 PBMCs (i.e., within the highest tertile); HIV-1 RNA and proviral DNA loads; IgG2 antibodies against HIV-1 p55, p24, p68, gp160, gp120, and gp41 [47]; and HLA allele B12 [16] yielded P < .20. A first backward multivariate analysis of the 6 patients with IgG2 antibody responses showed that only antiHIV-1 gp41specific IgG2 antibodies were independent predictors of LTNP (P < .05), and this variable was then incorporated into the multivariate analysis with the other retained variables. Only high HIV-1 p24specific cell counts and antiHIV-1 gp41specific IgG2 antibodies significantly predicted persistence of CD4 Th1 cell counts >600 cells/mm3. The probability of maintaining LTNP was 4.2-fold higher (relative risk [RR], 4.2 [95% confidence interval {CI}, 1.611.1]) in patients with antiHIV-1 gp41specific IgG2 antibodies than in those without, and it was 2.5-fold higher (RR, 2.5 [95% CI, 1.15.6]) if HIV-1 p24specific CD4 Th1 cell counts were >170 sfc/106 PBMCs. The combination of both responses had even stronger effect on CD4 Th1 cell counts, as is shown in the life-table analysis (figure 4): (1) all 5 patients with high HIV-1 p24specific CD4 Th1 cell counts and antiHIV-1 gp41specific IgG2 antibodies maintained LTNP; (2) 5 of the 11 patients with >170 sfc/106 PBMCs but without IgG2 antibodies had CD4 Th1 cell counts >600 cells/mm3; (3) 4 of the 8 patients with 170 sfc/106 PBMCs but with IgG2 antibodies had CD4 Th1 cell counts >600 cells/mm3; and (4) only 7 of the 29 patients with none of these parameters had CD4 Th1 cell counts >600/mm3 at the end of follow-up.

    DISCUSSION

    This unique overall analysis of the various aspects of immune responses to HIV-1 and viral and host gene parameters in a cohort of nonprogressors clearly demonstrates that the combination of high frequencies of antiHIV-1 p24specific CD4 Th1 cells and antiHIV-1 gp41specific IgG2 antibodies are better predictors for persistence of LTNPdefined as CD4 Th1 cell count >600 cells/mm3 for >10 yearsthan are IFN-producing CD8 T cell responses to HIV-1 Gag, plasma and cellular virus parameters, host chemokine/chemoreceptor gene variants, and HLA alleles. The data also provide novel evidence for a strong association between the frequencies of HIV-1 p24specific CD4 Th1 cells and the frequencies of HIV-1infected cells when measured simultaneously, and this association was even potentiated by an HIV-1 p24specific CD4 Th1 cell proliferation or IL-2 production that was independent of the depletion of CD4 Th1 cells.

    CD4 Th1 cell responses, which were independent of the CD4 Th1 cell counts, were strongly correlated with virus parameters at entry into the study. The large sample size of the ALT cohort and the wide ranges of viral production at entry into the study may explain the differences between our results and those of previous studies in which the selected viral parameters were in a much less broad range but did not correlate with the CD4 Th1 cell responses to HIV-1 [33, 3941]. Discrepancies with other studies, in which no such associations were found [3843], may also be explained by several technical factors, although all assays used to measure CD4 Th1 cell responses in the present study were concordant at indicating correlation with virus parameters, particularly stimulation with a whole recombinant p24 protein instead of peptides overlapping the whole gag sequence, the duration of incubation, and the threshold of detection in our ELISPOT assay.

    In accordance with the results of previous studies [42, 43], we found that both IL-2 and IFN- production against HIV-1 p24 was associated with lower levels of HIV-1 RNA in plasma and proviral DNA in PBMCs, although IFN- production alone was not. However, neither HIV-1specific proliferation nor IL-2 production predicted NP, as assessed by stable CD4 Th1 cell counts. Such features might reflect the exquisite sensitivity to HIV-1 infection of CD4 Th1 cells capable of proliferation and IL-2 production, whereas IFN-producing CD4 Th1 cells might be more resistant. However, only NSI R5 viruses of low cytopathogenicity were found in the ALT cohort [37, 46]. In addition, we have shown here for the first time that these responses were independent of the host chemokine/chemoreceptor gene variants known to limit HIV-1 entry [1420]. These data suggest that an association between strong HIV-1specific CD4 Th1 cell responses and low virus parameters does not reflect a genetic protection from HIV-1 infection in this cohort but rather reflects an immunity-based control of the virus.

    In contrast, frequencies of HIV-1 Gagspecific IFN-producing CD8 T cells were correlated with CD4 Th1 cell frequencies but not with plasma HIV-1 RNA loads. This latter finding, in accordance with the results of several studies [3334], does not exclude the possibility, however, that effector [57] or memory cytotoxic T lymphocytes (CTLs) might better correlate with LTNP than does the immediate IFN- production of CD8 T cells tested in the present study. Indeed, we reported elsewhere that the number of HIV-1specific memory CTLs, but not the number of HIV-1specific IFN-producing CD8 T cells, was significantly higher in nonprogressors from the ALT cohort than in progressors [36]. We also showed elsewhere that HIV-1specific memory CTLs were negatively correlated with HIV-1 RNA loads in progressors [35]. However, the correlation we found in the present study between the frequencies of IFN-producing CD8 T cells specific for HIV-1 proviral DNA is novel and suggests that those cells control HIV-1infected cells better than soluble virions do, and this finding is in accordance with the reactivity that major histocompatibility complex class Irestricted CD8 T cells have with cell-associated antigens.

    The multivariate analysis compared, in 55 nonprogressors, the predictive value of all parameters that were associated even roughly with control of viral replication in plasma in a univariate analysis. Viral factors and host chemokine/chemoreceptor genes and HLA alleles were less predictive than were high frequencies of HIV-1 p24specific CD4 Th1 cells combined with antiHIV-1 gp41specific IgG2 antibodies for the persistence of CD4 Th1 cell counts >600 cells/mm3 over the course of 4 years. The sample size did not allow us to test whether these 2 parameters could also predict long-term control of viral replication, because only 26 patients had an HIV-1 RNA load <10,000 copies/mL and only 14 patients had an HIV-1 RNA load <400 copies/mL at entry into the study. We cannot exclude the possibility that viral and host gene parameters would have a predictive value in a larger sample size, but our data illustrate the more potent effect of the CD4 Th1 cell parameters. Whether the predictive value of antiHIV-1 gp41specific IgG2 antibodies reveals a specific immune protection, as a recent study suggests [58], remains to be determined. In the present study, we considered IgG2 antibody levels to reflect CD4 Th1 cell functions, because, in humans, as in mice, IgG2 antibody production certainly depends on IFN- production [5962]. Indeed, although IgG1 antibodies predominate in HIV-1specific responses [63], IgG2 antibodies to various HIV-1 antigens had been previously found in patients in the ALT cohort with low HIV-1 RNA loads [47] and were positively correlated with HIV-1 p24specific CD4 Th1 cell responses, suggesting that they might be used as a soluble marker of CD4 Th1 cell responses to HIV-1, as has been proposed in mice.

    In conclusion, although progression of HIV-1 disease likely depends on several factors, the CD4 Th1 cell responses to HIV-1 are the strongest correlate of the persistence of NP, thus emphasizing the key contribution that CD4 Th1 cells and IgG2 antibodies specific for HIV-1 p24 and gp41, respectively, make to the long-term control of HIV-1 disease. The independence of the HIV-1specific CD4 Th1 cell responses from genetic factors of NP provides hope that efforts to design vaccines and immune-based treatment strategies will succeed in conferring nonprogressor status to all HIV-infected individuals.

    ADDITIONAL ASYMPTOMATIQUES à LONG TERME STUDY GROUP MEMBERS

    Additional Asymptomatiques à Long Terme Study Group Members are as follows: V. Calvez, D. Candotti, and J. M. Huraux (Virologie, Hpital Pitié-Salpêtrière, Paris), J. M. Bouley (Immuno Hématologie, Hpital Saint-Louis, Paris), and S. Chaput and G. Spiridon (Medicine Interne, Hpital Cochin, Paris).

    Acknowledgments

    We thank R. Kolbe (Transgène), for kindly providing the HIV recombinant proteins, and N. Alatrakchi and M. Almeida, for their contribution to the CD4 Th1 cell and CD8 T cell enzyme-linked immunospot analyses. We are particularly grateful to all the patients and clinicians who participated in the Asymptomatiques à Long Terme cohort: M. Kazatchkine, C. Gazengel, J. F. Delfraissy, A. Sobel, J. P. Coulaud, C. Katlama, F. Bricaire, W. Rozenbaum, J. C. Imbert, E. Oksenhendler, J. P. Escande, B. Dupont, C. Patey, L. J. Couderc, J. A. Gastaut, F. Beaufils, P. Lang, C. Choutet, F. Bazin, and F. Dellamonica.

    References

    1.  Buchbinder SP, Katz MH, Hessol NA, O'Malley PM, Holmberg SD. Long-term HIV-1 infection without immunologic progression. AIDS 1994; 8:11238. First citation in article

    2.  Hendriks JC, Medley GF, van Griensven GJ, Coutinho RA, Heisterkamp SH, van Druten HA. The treatment-free incubation period of AIDS in a cohort of homosexual men. AIDS 1993; 7:2319. First citation in article

    3.  Cao Y, Qin L, Zhang L, Safrit J, Ho DD. Virologic and immunologic characterization of long-term survivors of human immunodeficiency virus type 1 infection. N Engl J Med 1995; 332:2018. First citation in article

    4.  Barker E, Mackewicz CE, Reyes-Teran G, et al. Virological and immunological features of long-term human immunodeficiency virus-infected individuals who have remained asymptomatic compared with those who have progressed to acquired immunodeficiency syndrome. Blood 1998; 92:310514. First citation in article

    5.  Pantaleo G, Menzo S, Vaccarezza M, et al. Studies in subjects with long-term nonprogressive human immunodeficiency virus infection. N Engl J Med 1995; 332:20916. First citation in article

    6.  Salhi Y, Costagliola D. Long-term nonprogression in HIV infection. Clinical Epidemiology Group from the Centre d'Information et de Soins de l'Immunodeficience Humaine. J Acquir Immune Defic Syndr Hum Retrovirol 1997; 16:40911. First citation in article

    7.  Mellors JW, Rinaldo CR Jr, Gupta P, White RM, Todd JA, Kingsley LA. Prognosis in HIV-1 infection predicted by the quantity of virus in plasma. Science 1996; 272:116770. First citation in article

    8.  Learmont JC, Geczy AF, Mills J, et al. Immunologic and virologic status after 14 to 18 years of infection with an attenuated strain of HIV-1. A report from the Sydney Blood Bank Cohort. N Engl J Med 1999; 340:171522. First citation in article

    9.  Dean M, Carrington M, Winkler C, et al. Genetic restriction of HIV-1 infection and progression to AIDS by a deletion allele of the CKR5 structural gene. Hemophilia Growth and Development Study, Multicenter AIDS Cohort Study, Multicenter Hemophilia Cohort Study, San Francisco City Cohort, ALIVE Study. Science 1996; 273:185662. First citation in article

    10.  Finzi D, Blankson J, Siliciano JD, et al. Latent infection of CD4+ T cells provides a mechanism for lifelong persistence of HIV-1, even in patients on effective combination therapy. Nat Med 1999; 5:5127. First citation in article

    11.  Letvin NL, Barouch DH, Montefiori DC. Prospects for vaccine protection against HIV-1 infection and AIDS. Annu Rev Immunol 2002; 20:7399. First citation in article

    12.  Kirchhoff F, Greenough TC, Brettler DB, Sullivan JL, Desrosiers RC. Brief report: absence of intact nef sequences in a long-term survivor with nonprogressive HIV-1 infection. N Engl J Med 1995; 332:22832. First citation in article

    13.  Candotti D, Costagliola D, Joberty C, et al. Status of long-term asymptomatic HIV-1 infection correlates with viral load but not with virus replication properties and cell tropism. French ALT Study Group. J Med Virol 1999; 58:25663. First citation in article

    14.  Samson M, Libert F, Doranz BJ, et al. Resistance to HIV-1 infection in caucasian individuals bearing mutant alleles of the CCR-5 chemokine receptor gene. Nature 1996; 382:7225. First citation in article

    15.  Michael NL, Louie LG, Rohrbaugh AL, et al. The role of CCR5 and CCR2 polymorphisms in HIV-1 transmission and disease progression. Nat Med 1997; 3:11602. First citation in article

    16.  Magierowska M, Theodorou I, Debre P, et al. Combined genotypes of CCR5, CCR2, SDF1, and HLA genes can predict the long-term nonprogressor status in human immunodeficiency virus-1-infected individuals. Blood 1999; 93:93641. First citation in article

    17.  Faure S, Meyer L, Costagliola D, et al. Rapid progression to AIDS in HIV+ individuals with a structural variant of the chemokine receptor CX3CR1. Science 2000; 287:22747. First citation in article

    18.  Kaslow RA, Carrington M, Apple R, et al. Influence of combinations of human major histocompatibility complex genes on the course of HIV-1 infection. Nat Med 1996; 2:40511. First citation in article

    19.  Nelson GW, Kaslow R, Mann DL. Frequency of HLA allele-specific peptide motifs in HIV-1 proteins correlates with the allele's association with relative rates of disease progression after HIV-1 infection. Proc Natl Acad Sci USA 1997; 94:98027. First citation in article

    20.  Leslie AJ, Pfafferott KJ, Chetty P, et al. HIV evolution: CTL escape mutation and reversion after transmission. Nat Med 2004; 10:2829. First citation in article

    21.  Schmitz JE, Kuroda MJ, Santra S, et al. Control of viremia in simian immunodeficiency virus infection by CD8+ lymphocytes. Science 1999; 283:85760. First citation in article

    22.  Cocchi F, DeVico AL, Garzino-Demo A, Arya SK, Gallo RC, Lusso P. Identification of RANTES, MIP-1 alpha, and MIP-1 beta as the major HIV-suppressive factors produced by CD8+ T cells. Science 1995; 270:18115. First citation in article

    23.  Klein MR, van Baalen CA, Holwerda AM, et al. Kinetics of Gag-specific cytotoxic T lymphocyte responses during the clinical course of HIV-1 infection: a longitudinal analysis of rapid progressors and long-term asymptomatics. J Exp Med 1995; 181:136572. First citation in article

    24.  Harrer T, Harrer E, Kalams SA, et al. Cytotoxic T lymphocytes in asymptomatic long-term nonprogressing HIV-1 infection: breadth and specificity of the response and relation to in vivo viral quasispecies in a person with prolonged infection and low viral load. J Immunol 1996; 156:261623. First citation in article

    25.  Rinaldo C, Huang XL, Fan ZF, et al. High levels of anti-human immunodeficiency virus type 1 (HIV-1) memory cytotoxic T-lymphocyte activity and low viral load are associated with lack of disease in HIV-1-infected long-term nonprogressors. J Virol 1995; 69:583842. First citation in article

    26.  Rosenberg ES, Billingsley JM, Caliendo AM, et al. Vigorous HIV-1-specific CD4+ T cell responses associated with control of viremia. Science 1997; 278:144750. First citation in article

    27.  Kalams SA, Buchbinder SP, Rosenberg ES, et al. Association between virus-specific cytotoxic T-lymphocyte and helper responses in human immunodeficiency virus type 1 infection. J Virol 1999; 73:671520. First citation in article

    28.  Pitcher CJ, Quittner C, Peterson DM, et al. HIV-1-specific CD4+ T cells are detectable in most individuals with active HIV-1 infection, but decline with prolonged viral suppression. Nat Med 1999; 5:51825. First citation in article

    29.  Pontesilli O, Carotenuto P, Kerkhof-Garde SR, et al. Lymphoproliferative response to HIV type 1 p24 in long-term survivors of HIV type 1 infection is predictive of persistent AIDS-free infection. AIDS Res Hum Retroviruses 1999; 15:97381. First citation in article

    30.  Matloubian M, Concepcion RJ, Ahmed R. CD4+ T cells are required to sustain CD8+ cytotoxic T-cell responses during chronic viral infection. J Virol 1994; 68:805663. First citation in article

    31.  Kalams SA, Walker BD. The critical need for CD4 help in maintaining effective cytotoxic T lymphocyte responses. J Exp Med 1998; 188:2199204. First citation in article

    32.  Dalod M, Dupuis M, Deschemin JC, et al. Broad, intense anti-human immunodeficiency virus (HIV) ex vivo CD8+ responses in HIV type 1infected patients: comparison with anti-Epstein-Barr virus responses and changes during antiretroviral therapy. J Virol 1999; 73:710816. First citation in article

    33.  Betts MR, Ambrozak DR, Douek DC, et al. Analysis of total human immunodeficiency virus (HIV)-specific CD4+ and CD8+ T-cell responses: relationship to viral load in untreated HIV infection. J Virol 2001; 75:1198391. First citation in article

    34.  Migueles SA, Laborico AC, Shupert WL, et al. HIV-specific CD8+ T cell proliferation is coupled to perforin expression and is maintained in nonprogressors. Nat Immunol 2002; 3:10618. First citation in article

    35.  Chouquet C, Autran B, Gomard E, et al. Correlation between breadth of memory HIV-specific cytotoxic T cells, viral load and disease progression in HIV infection. AIDS 2002; 16:2399407. First citation in article

    36.  Sun Y, Iglesias E, Samri A, et al. A systematic comparison of methods to measure HIV-1 specific CD8 T cells. J Immunol Methods 2003; 272:2334. First citation in article

    37.  Sallusto F, Geginat J, Lanzavecchia A. Central memory and effector memory T cell subsets: function, generation, and maintenance. Annu Rev Immunol 2004; 22:74563. First citation in article

    38.  Wilson JD, Imami N, Watkins A, et al. Loss of CD4+ T cell proliferative ability but not loss of human immunodeficiency virus type 1 specificity equates with progression to disease. J Infect Dis 2000; 182:7928. First citation in article

    39.  Palmer BE, Boritz E, Blyveis N, Wilson CC. Discordance between frequency of human immunodeficiency virus type 1 (HIV-1)-specific gamma interferon-producing CD4+ T cells and HIV-1-specific lymphoproliferation in HIV-1-infected subjects with active viral replication. J Virol 2002; 76:592536. First citation in article

    40.  Imami N, Pires A, Hardy G, Wilson J, Gazzard B, Gotch F. A balanced type 1/type 2 response is associated with long-term nonprogressive human immunodeficiency virus type 1 infection. J Virol 2002; 76:901123. First citation in article

    41.  Boaz MJ, Waters A, Murad S, Easterbrook PJ, Vyakarnam A. Presence of HIV-1 Gag-specific IFN-gamma+IL-2+ and CD28+IL-2+ CD4 T cell responses is associated with nonprogression in HIV-1 infection. J Immunol 2002; 169:637685. First citation in article

    42.  Harari A, Petitpierre S, Vallelian F, Pantaleo G. Skewed representation of functionally distinct populations of virus-specific CD4 T cells in HIV-1-infected subjects with progressive disease: changes after antiretroviral therapy. Blood 2004; 103:96672. First citation in article

    43.  Younes SA, Yassine-Diab B, Dumont AR, et al. HIV-1 viremia prevents the establishment of interleukin 2-producing HIV-specific memory CD4+ T cells endowed with proliferative capacity. J Exp Med 2003; 198:190922. First citation in article

    44.  Douek DC, Brenchley JM, Betts MR, et al. HIV preferentially infects HIV-specific CD4+ T cells. Nature 2002; 417:958. First citation in article

    45.  McNeil AC, Shupert WL, Iyasere CA, et al. High-level HIV-1 viremia suppresses viral antigen-specific CD4+ T cell proliferation. Proc Natl Acad Sci USA 2001; 98:1387883. First citation in article

    46.  Candotti D, Calvez V, Autran B, Costagliola D, Rouzioux C, Agut H. Decreased peripheral circulation of HIV-infected cells in a subset of long-term nonprogressors. The French ALT Study Group. J Acquir Immune Defic Syndr 1999; 21:2535. First citation in article

    47.  Ngo-Giang-Huong N, Candotti D, Goubar A, et al. HIV type 1-specific IgG2 antibodies: markers of helper T cell type 1 response and prognostic marker of long-term nonprogression. AIDS Res Hum Retroviruses 2001; 17:143546. First citation in article

    48.  Stratégies d'utilisation des antirétroviraux dans l'infection par le VIH. Rapport d'experts 1997 sous la direction de J. Dormont. Paris: Médecine-Sciences, Flammarion, 1997. First citation in article

    49.  Stratégies d'utilisation des antirétroviraux dans l'infection par le VIH. Rapport d'experts 1998 sous la direction de J. Dormont. Paris: Médecine-Sciences, Flammarion, 1998. First citation in article

    50.  Prise en charge des personnes infectées par le VIH. Recommandations du groupe d'experts, rapport 1999 sous la direction du Professeur JF Delfraissy. Paris: Médecine-Sciences, Flammarion, 1999. First citation in article

    51.  Prise en charge des personnes infectées par le VIH. Recommandations du groupe d'experts, rapport 2000 sous la direction du Professeur JF Delfraissy. Paris: Médecine-Sciences, Flammarion, 2000. First citation in article

    52.  Christopherson C, Kidane Y, Conway B, Krowka J, Sheppard H, Kwok S. PCR-based assay to quantify human immunodeficiency virus type 1 DNA in peripheral blood mononuclear cells. J Clin Microbiol 2000; 38:6304. First citation in article

    53.  Burgard M, Izopet J, Dumon B, et al. HIV RNA and HIV DNA in peripheral blood mononuclear cells are consistent markers for estimating viral load in patients undergoing long-term potent treatment. AIDS Res Hum Retroviruses 2000; 16:193947. First citation in article

    54.  Carcelain G, Tubiana R, Samri A, et al. Transient mobilization of human immunodeficiency virus (HIV)-specific CD4 T-helper cells fails to control virus rebounds during intermittent antiretroviral therapy in chronic HIV type 1 infection. J Virol 2001; 75:23441. First citation in article

    55.  Autran B, Legac E, Blanc C, Debre P. A Th0/Th2-like function of CD4+CD7-T helper cells from normal donors and HIV-infected patients. J Immunol 1995; 154:140817. First citation in article

    56.  Kleinbaum DG, Morgenstern H. Modelling analysis strategy. In: Reinhold VN, ed. Epidemiologic research principles and quantitative methods. New York: Wiley, 1982:44756. First citation in article

    57.  Keoshkerian E, Ashton LJ, Smith DG, et al. Effector HIV-specific cytotoxic T-lymphocyte activity in long-term nonprogressors: associations with viral replication and progression. J Med Virol 2003; 71:48391. First citation in article

    58.  Hovanessian AG, Briand JP, Said EA, et al. The caveolin-1 binding domain of HIV-1 glycoprotein gp41 is an efficient B cell epitope vaccine candidate against virus infection. Immunity 2004; 21:61727. First citation in article

    59.  Snapper CM, Paul WE. Interferon-gamma and B cell stimulatory factor-1 reciprocally regulate Ig isotype production. Science 1987; 236:9447. First citation in article

    60.  Stevens TL, Bossie A, Sanders VM, et al. Regulation of antibody isotype secretion by subsets of antigen-specific helper T cells. Nature 1988; 334:2558. First citation in article

    61.  Finkelman FD, Katona IM, Mosmann TR, Coffman RL. IFN-gamma regulates the isotypes of Ig secreted during in vivo humoral immune responses. J Immunol 1988; 140:10227. First citation in article

    62.  Finkelman FD, Urban JF Jr, Beckmann MP, Schooley KA, Holmes JM, Katona IM. Regulation of murine in vivo IgG and IgE responses by a monoclonal anti-IL-4 receptor antibody. Int Immunol 1991; 3:599607. First citation in article

    63.  Sundqvist VA, Linde A, Kurth R, et al. Restricted IgG subclass responses to HTLV-III/LAV and to cytomegalovirus in patients with AIDS and lymphadenopathy syndrome. J Infect Dis 1986; 153:9703. First citation in article

作者: Valérie Martinez, Dominique Costagliola, Olivia B 2007-5-15
医学百科App—中西医基础知识学习工具
  • 相关内容
  • 近期更新
  • 热文榜
  • 医学百科App—健康测试工具