Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2011 Aug 4.
Published in final edited form as: J Acquir Immune Defic Syndr. 2008 Jun 1;48(2):149–155. doi: 10.1097/QAI.0b013e31816d9c3b

Early Changes in T-Cell Activation Predict Antiretroviral Success in Salvage Therapy of HIV Infection

Brett D Shepard *, Mona R Loutfy †,, Janet Raboud †,§, Frank Mandy ||, Colin M Kovacs †,, Christina Diong §, Michele Bergeron ||, Victoria Govan , Stacey A Rizza *, Jonathan B Angel , Andrew D Badley *
PMCID: PMC3149796  NIHMSID: NIHMS313401  PMID: 18360289

Abstract

Objective

Because effective antiretroviral therapy (ART) reduces immune activation, we hypothesize that early changes in immune activation are associated with subsequent virologic response to therapy.

Design

Observational cohort study.

Setting

Institutional HIV clinic.

Subjects

Thirty-four adult HIV patients with virologic failure on their current antiretroviral regimen.

Intervention

Change to salvage regimen selected by patient’s physician.

Main Outcome Measures

Measures of immune activation at baseline and at 2, 4, 8, and 24 weeks after enrollment. Data were analyzed by proportional hazards (PH) models.

Results

PH models showed that reductions between baseline and week 2 in expression of CD38 (P = 0.02) or CD95 (P = 0.02) on CD4+ T cells were associated with increased likelihood of achieving virologic suppression. Kaplan-Meier analysis demonstrated that patients who had reductions within the first 2 weeks of therapy in CD4+ T-cell expression of CD38 (P = 0.003) or CD95 (P = 0.08) were more likely to achieve viral suppression than those who did not.

Conclusions

Reduced CD4+ T-cell expression of CD38 and CD95 occurring within 2 weeks of salvage therapy is associated with subsequent viral suppression. Monitoring CD38 and CD95 may allow earlier assessment of the response to ART.

Keywords: antiretroviral therapy, clinical trial, HIV, immune activation, prognosis, virologic failure


The management of HIV-infected patients is guided largely by 2 key measures: the amount of viral replication, which predicts the rate of disease progression, and the degree of immune system dysfunction as measured by absolute CD4+ T-cell number.1 The continuing development of effective anti-retroviral therapies (ARTs) has allowed pharmacologic suppression of HIV replication in many infected patients, often leading to quantitative and qualitative reconstitution of the immune system. Ongoing viral replication in the presence of incompletely suppressive drugs, when coupled with the labile genetic nature of HIV, may lead to resistance mutations, loss of virologic control,25 decreased CD4+ T-cell number, and significant morbidity and mortality, however.6,7 Moreover, once resistance occurs, the likelihood of extended resuppression of viral replication with subsequent therapies is substantially lower.8,9 With the current practice of monitoring only viral replication and CD4+ T-cell number, it may take several months to determine whether or not a new antiretroviral regimen is effective.1 Therefore, identifying markers that accurately predict at early time points whether there is likely to be eventual viral suppression, particularly in situations in which salvage drug regimens are required, would be of great clinical utility.

There has been considerable interest in evaluating markers of immune function to aid in the management of patients infected with HIV. Several immunologic markers have been proposed as predictors of disease outcome, including serum p24 antigen levels, serum tumor necrosis factor (TNF)-α and TNFγ levels, CD38 antigen intensity on CD8+ T cells, and a variety of other T-cell activation markers (eg, CD95, human leukocyte antigen D-related [HLA-DR], CD28).1021 Not all the previous studies evaluated patients who had previously received ART,11,13,16 however, and, often, only late time points (eg, ≥24 weeks after therapy) were analyzed.10,12,14,15,17,18,20 Although a study that analyzed earlier time points (beginning at 12 weeks) showed that the CD8+CD38+ T-cell count is a prognostic marker of therapeutic failure in HIV-infected children,22 no studies have evaluated the prognostic value of early changes in these immune markers in adult HIV-positive patients over time. Thus, it is unknown whether early changes in such markers in adult patients add prognostic information to the current standard clinical practice of monitoring CD4+ T-cell counts and HIV viral load at intervals of every few months.

With the goal of identifying whether changes in immunologic markers that occur early after initiation of salvage therapy are associated with subsequent virologic response to therapy, a single-center cohort study was initiated. Multivariate flow cytometry was used to assess immune activation and T-cell subsets before and during salvage therapy to identify immunologic markers that predict within the first few weeks the subsequent restoration of virologic control.

METHODS

Study Population and Intervention

Patients were enrolled from September 1, 2001 through December 1, 2001 from the Maple Leaf Medical Clinic (Toronto, Ontario, Canada). The study (Clinical Trials Registry NCT00440206) was reviewed and approved by the respective Institutional Review Board (IRB), IRB Services, and the Ottawa Hospital. Patients providing written informed consent and meeting the following inclusion criteria were enrolled in the study: (1) previously documented HIV infection, (2) age ≥18 years, (3) treatment with the same anti-retroviral regimen (including ≥3 medications) for at least 3 months, (4) virologic failure defined as viral load ≥50 copies/mL on 2 occasions at least 2 weeks apart, and (5) plans by the patient and patient’s attending physician to switch from the current antiretroviral regimen to a salvage regimen (ie, ART initiated after failure of a prior regimen). Exclusion criteria included patients with (1) hemoglobin <100 g/L, (2) platelet count <20,000 cells/μL, (3) international normalized ratio (INR) ≥3.5, or (4) partial thromboplastin time (PTT) ≥60 IU. For each patient, the study concluded when either of the following was met: (1) 12 months had elapsed since the date of enrollment or (2) the salvage antiretroviral regimen was discontinued or changed at the discretion of the patient and/or the patient’s physician.

Laboratory Measurements and Clinical Evaluation

Viral load assessment was determined by measuring the plasma HIV-1 RNA level using the branched-DNA Chiron 3.0 assay (Chiron, Emeryville, CA) with a lower limit of detection of 50 copies/mL. For each patient, viral load measurements were made at the time of the study enrollment (before initiation of salvage regimen, week 0) and at 2, 4, 8, 24, and 48 weeks after initiation of the new regimen. A clinical evaluation and safety and toxicity laboratory evaluations for monitoring ART were performed by the patient’s physician according to standard clinical care guidelines at the time of the study.

Immunophenotypic Assays

Immunophenotypic analysis was performed using 2 4-color immunophenotyping panels at baseline and at 2, 4, 8, and 24 weeks of follow-up. The first panel (panel A) consisted of the following commercially premixed antibodies: CD45-fluorescein isothiocyanate (FITC)/CD4-RD1/CD8-ECD/CD3-PC5 and CD45-FITC/CD56-RD1/CD19-PE-TxRed (ECD)/CD3-PC5 (Beckman Coulter, Fullerton, CA). The second panel (panel B) was prepared with the following antibodies: CD45RA-FITC, HLA-DR-FITC (BD Biosciences, Fullerton, CA) and CD4-ECD, CD8-PC5, CD45RO-phycoerythrin (PE), CD28-FITC, CD62L-PE, CD69-PE, CD38-PE, and CD45RO-PE (Beckman Coulter).

Samples were prepared by mixing each venous blood sample with antibodies from panel A or panel B and incubating for 10 minutes at room temperature before lysing with the automated ImmunoPrep reagent system (Beckman Coulter). Samples were fixed with 0.5 mL of 2% paraformaldehyde solution. For measurements with panel A, 100 μL of Flow-Count (Beckman Coulter) absolute count beads was added before analysis to allow for measurement of absolute cell counts as a single-platform procedure.

Flow cytometry was performed with Coulter Epics XL-MCL (Beckman Coulter). The instrument was prepared using QC3 reference bead standards (Bangs Laboratories, Fishers, IN) to establish target channels and standardize the fluorescence intensity analysis range throughout the study. Lymphocytes gates were set in panel A using the universal template CD45 and CD4 double-anchor gate.23 Absolute and relative cell values were measured. The CD4+ and CD8+ T-cell gates in panel B were defined with the heterogeneous gating strategy, where CD4 and CD8 bright fluorescence expression was expressed against the side-scattered light. All results were expressed as percentages of CD4 and bright CD8 cells. In addition, the mean fluorescence intensity of each of the activation markers HLA-DR, CD38, CD28, CD95, and CD69 was measured.

Statistical Analysis

Associations between baseline characteristics and subsequent virologic suppression were calculated using the χ2 test or Fisher exact test for categoric covariates and the Wilcoxon rank sum test for continuous covariates. Virologic suppression was defined to be a viral load <50 copies/mL at any time during the study. Patients were classified as virologic “responders” if they achieved virologic suppression during the study period; otherwise, they were classified as “nonresponders.” Cox proportional hazards (PR) models with fixed covariates and time-dependent covariates and Kaplan-Meier analyses were used to determine the associations of covariates with time to achieve virologic suppression.

RESULTS

Thirty-four patients were enrolled in the study. Two patients did not complete follow-up. All patients were male, 27 (84%) of the 32 patients were white, and the median age was 43 years (interquartile range: 39 to 48 years). Nineteen (59%) of the 32 patients achieved virologic suppression during the study. Of these 19, 14 had evaluable viral load data at week 2, and 5 of 14 achieved viral suppression by that time point. Nine (64%) of 14 patients who ultimately achieved virologic suppression did not achieve virologic suppression by week 2. The baseline demographics and clinical characteristics of the study population are shown in Table 1. Patients who achieved virologic suppression were less likely to have a history of an AIDS-defining illness (P < 0.01), had a lower baseline viral load (P < 0.01), and had a higher baseline CD4+ T-cell count (P = 0.03). Higher baseline expression of CD38 on CD4 (P = 0.04) and CD8 (P = 0.01), higher expression of CD38 mean fluorescence intensity on CD8 (P = 0.03), and lower expression of CD28 on CD4 (P = 0.02) were associated with a decreased likelihood of virologic suppression. Other baseline demographic and clinical covariates were not different between groups.

TABLE 1.

Comparison of Demographic and Clinical Characteristics by Virologic Responder Status

Characteristics Nonresponder (n = 13) Responder (n = 19) Total (n = 32) P
Male 13 (100.0%) 19 (100.0%) 32 (100.0%)
Age, y 40 (39 to 42) 45 (41 to 53) 43 (39 to 48) 0.05
Race 0.85
 White 11 (84.6%) 16 (84.2%) 27 (84.4%)
 Black 1 (7.7%) 2 (10.5%) 3 (9.4%)
 Asian 1 (7.7%) 0 (0.0%) 1 (3.1%)
 Other 0 (0.0%) 1 (5.3%) 1 (3.1%)
HIV risk factor 0.46
 MSM 11 (91.7%) 15 (83.3%) 26 (86.7%)
 Heterosexual 1 (8.3%) 0 (0.0%) 1 (3.3%)
 Blood transfusion 0 (0.0%) 1 (5.6%) 1 (3.3%)
 MSM + IVDU 0 (0.0%) 2 (11.1%) 2 (6.7%)
 Missing 1 1
AIDS-defining illness 12 (92.3%) 8 (42.1%) 20 (62.5%) 0.01
Hepatitis B 1 (7.7%) 1 (5.3%) 2 (6.3%) 0.99
Hepatitis C 1 (7.7%) 3 (15.8%) 4 (12.5%) 0.63
No. years using ART 10 (8 to 14) 11.5 (8 to 13) 11 (8 to 13) 0.95
No. previous antiretroviral drugs 11 (10 to 12) 8 (7 to 11) 10 (7 to 12) 0.07
No. previous regimens 7 (6 to 11) 6.0 (4.0 to 7.0) 6.0 (5.0 to 9.5) 0.12
Present regimen includes PI 13 (100.0%) 13 (68.4%) 26 (81.3%) 0.06
Present regimen includes NNRTI 8 (61.5%) 11 (57.9%) 19 (59.4%) 0.84
Baseline viral load, log10 copies/mL 5.0 (4.9 to 5.5) 3.7 (2.7 to 4.7) 4.3 (3.2 to 5.0) <0.01
Baseline CD4 count, cells/mm3 43 (20 to 181) 230 (132 to 350) 191 (50 to 276) 0.03
Baseline CD8 count, cells/mm3 560 (421 to 861) 748 (420 to 1240) 710 (421 to 965) 0.39
CD3+ mean 75.5 (74.4 to 81.6) 75.8 (69.4 to 82.9) 75.7 (72.2 to 82.3) 0.89
CD38+ on CD4 85.6 (66.8 to 87.0) 64.6 (48.4 to 78.8) 71.4 (55.7 to 85.0) 0.04
HLA-DR on CD4 12.7 (7.2 to 16.7) 7.8 (6.4 to 13.5) 10.4 (6.9 to 16.2) 0.39
CD38+ on CD8 94.1 (89.1 to 95.6) 70.0 (46.7 to 84.1) 77.4 (48.2 to 94.3) 0.01
HLA-DR on CD8 22.6 (19.9 to 32.5) 18.6 (13.4 to 27.9) 21.1 (16.4 to 28.0) 0.15
CD28+ on CD4 51.9 (48.5 to 65.6) 76.9 (57.5 to 89.4) 65.2 (50.1 to 86.5) 0.02
CD95+ on CD4 62.1 (58.5 to 70.3) 63.5 (56.1 to 83.6) 62.8 (57.2 to 76.2) 0.82
CD95+ on CD8 60.1 (40.6 to 67.4) 56.4 (34.4 to 61.3) 56.8 (36.9 to 63.6) 0.37
CD69+ on CD4 12.2 (6.7 to 14.9) 15.4 (7.3 to 21.8) 13.9 (6.8 to 19.5) 0.35
CD69+ on CD8 21.8 (13.4 to 26.2) 17.8 (12.5 to 20.6) 18.2 (12.8 to 23.8) 0.29
CD38 MnI on CD4 21.8 (15.6 to 26.0) 16.9 (8.7 to 22.5) 18.5 (12.8 to 24.6) 0.22
CD38 MnI on CD8 36.6 (18.6 to 43.1) 14.4 (5.3 to 24.2) 20.5 (9.2 to 36.9) 0.03
CD95 MnI on CD4 6.0 (5.2 to 7.9) 6.1 (4.9 to 6.9) 6.0 (4.9 to 7.4) 0.70

Quantities in N (%) or median.

Interquartile ranges are reported with medians.

Percentages are calculated based on valid data only.

IVDU indicates intravenous drug user; MnI mean intensity; MSM, men who have sex with men; NNRTI, nonnucleoside reverse transcriptase inhibitor.

Univariate PH models were used to determine the associations of baseline demographic and clinical characteristics with time to virologic suppression (Table 2). Patients with a history of an AIDS-defining illness (hazard ratio [HR] = 0.32; P = 0.01) and those with a higher baseline viral load (HR = 0.56 per log10 copies/mL; P < 0.01) were less likely to achieve virologic suppression. Patients with a CD4+ T-cell count ≥100 cells/mm3 at baseline were more likely to achieve virologic suppression (HR = 5.00; P = 0.01) than patients with a CD4+ T-cell count <100 cells/mm3. Univariate PH models with time-dependent covariates were used to estimate the HR of virologic suppression associated with immune markers (Table 3). Continued high expression of CD38 on CD4+ and CD8+ T cells (HR = 0.73 per 10 cells/mm3; P = 0.01 and HR = 0.80 per 10 cells/mm3; P = 0.02, respectively) and increased expression of CD95 on CD4+ T cells (as measured by mean fluorescence intensity, HR = 0.81 per cell/mm3; P = 0.05) during the course of the study were associated with a decreased likelihood of virologic suppression.

TABLE 2.

Univariate PH Models of Time to Virologic Suppression

Covariate HR P
Age, per y 1.04 (0.99 to 1.10) 0.13
White race 1.00 (0.29 to 3.43) 0.99
Men who have sex with men 0.61 (0.17 to 2.14) 0.44
AIDS-defining illness 0.32 (0.13 to 0.79) 0.01
Hepatitis B 0.87 (0.12 to 6.51) 0.89
Hepatitis C 1.60 (0.46 to 5.56) 0.46
Baseline viral load, log10 copies/mL 0.56 (0.39 to 0.80) <0.01
Baseline CD4 count, per 100 cells/mm3 1.16 (0.96 to 1.40) 0.13
Baseline CD4 count >200 cells/mm3 3.47 (1.34 to 8.96) 0.01
Baseline CD4 count >100 cells/mm3 5.00 (1.45 to 17.27) 0.01
No. years using ART 0.97 (0.84 to 1.12) 0.66
No. previous antiretroviral drugs 0.81 (0.67 to 0.98) 0.03
No. previous regimens 0.91 (0.79 to 1.05) 0.18
Present regimen includes PI 0.41 (0.16 to 1.09) 0.07
Present regimen includes NNRTI 0.91 (0.36 to 2.26) 0.83

NNRTI indicates nonnucleoside reverse transcriptase inhibitor.

TABLE 3.

Univariate PH Models of Time to Virologic Suppression With Time-Dependent Covariates

Covariate Unit (Cells/mm3) HR P
CD4 count 100 1.17 (0.90 to 1.52) 0.23
CD8 count 100 1.06 (0.97 to 1.16) 0.16
CD3+ mean 10 1.32 (0.82 to 2.12) 0.26
CD38+ on CD4 10 0.73 (0.57 to 0.94) 0.01
HLA-DR on CD4 10 0.68 (0.37 to 1.25) 0.21
CD38+ on CD8 10 0.80 (0.67 to 0.96) 0.02
HLA-DR on CD8 10 0.76 (0.49 to 1.16) 0.20
CD28+ on CD4 10 1.17 (0.92 to 1.48) 0.20
CD95+ on CD4 10 0.84 (0.61 to 1.16) 0.29
CD95+ on CD8 10 0.84 (0.64 to 1.08) 0.18
CD69+ on CD4 10 0.85 (0.56 to 1.30) 0.46
CD69+ on CD8 10 0.68 (0.43 to 1.09) 0.11
CD38 MnI on CD4 10 0.65 (0.40 to 1.06) 0.08
CD38 MnI on CD8 10 0.68 (0.47 to 0.98) 0.04
CD95 MnI on CD4 1 0.81 (0.66 to 1.00) 0.05

MnI indicates mean intensity.

Univariate PH models with covariates for changes in immunologic markers from baseline to week 2 identified associations between changes within the first 2 weeks of salvage therapy and virologic suppression (Table 4). Decreased expression of HLA-DR and CD95 (the latter measured by mean fluorescence intensity) on CD4+ T cells during the first 2 weeks of salvage therapy was associated with increased likelihood of virologic suppression (HR = 0.33 per 10 cells/mm3; P = 0.02 and HR = 0.78 per 1 cell/mm3; P = 0.02, respectively). As at baseline, decreased levels of CD38 on CD4+ T cells (as measured by mean fluorescence intensity) continued to be associated with an increased likelihood of achieving virologic suppression (HR = 0.66 per 10 cells/mm3; P = 0.02).

TABLE 4.

Univariate PH Models of Time to Virologic Suppression, With Immune Marker Changes from Baseline to Week 2 as Covariates

Covariate Unit (Cells/mm3) HR P
Change in CD4 count 100 0.51 (0.19 to 1.40) 0.19
Change in CD8 count 100 0.87 (0.69 to 1.12) 0.28
Change in CD3+ mean 10 2.30 (0.63 to 8.39) 0.21
Change in CD38+ on CD4 10 0.83 (0.46 to 1.50) 0.55
Change in HLA-DR on CD4 10 0.33 (0.14 to 0.82) 0.02
Change in CD38+ on CD8 10 1.00 (0.46 to 2.16) 1.00
Change in HLA-DR on CD8 10 0.98 (0.52 to 1.86) 0.95
Change in CD28+ on CD4 10 0.91 (0.71 to 1.15) 0.42
Change in CD95+ on CD4 10 0.73 (0.47 to 1.12) 0.15
Change in CD95+ on CD8 10 0.95 (0.68 to 1.32) 0.76
Change in CD69+ on CD4 10 0.75 (0.54 to 1.04) 0.08
Change in CD69+ on CD8 10 0.70 (0.36 to 1.35) 0.29
Change in CD38 MnI on CD4 10 0.66 (0.46 to 0.95) 0.02
Change in CD38 MnI on CD8 10 1.35 (0.66 to 2.77) 0.41
Change in CD95 MnI on CD4 1 0.78 (0.63 to 0.97) 0.02

In a multivariate PH model, patients with baseline CD4+ T cells ≥200 cells/mm3 remained much more likely to achieve virologic suppression (HR = 11.93; P = 0.003). After controlling for baseline CD4+ T-cell counts in multivariate modeling, decreases in expression of CD38 on CD4+ T cells between baseline and week 2 were associated with an increased likelihood of virologic suppression (HR = 2.13 per 10 cells/mm3; P = 0.007). Moreover, a decrease in expression of CD95 on CD4+ T cells between baseline and 2 weeks of salvage therapy was associated with an increased likelihood of subsequent virologic suppression (HR = 1.54 per cell/mm3; P = 0.007). Kaplan-Meier analysis confirmed that changes in CD38 on CD4+ T cells within the first 2 weeks of salvage ART are associated with subsequent viral suppression (P = 0.003; Fig. 1A). Although Kaplan-Meier analysis demonstrated a similar trend for changes in CD95 on CD4+ T cells within the first 2 weeks of salvage ART, the trend was not statistically significant (P = 0.08; see Fig. 1B).

FIGURE 1.

FIGURE 1

A, Probability of viral suppression stratified by change in CD38 expression on CD4+ T cells between baseline and week 2. B, Probability of viral suppression stratified by change in CD95 expression on CD4+ T cells between baseline and week 2.

DISCUSSION

Although other studies have highlighted an association between decreased immune activation and virologic suppression and/or immune reconstitution, it has been unknown how early such changes occur and whether they can be used to predict antiviral response. The present study is the first to show that decreased immune activation within the first 2 weeks of salvage therapy is associated with, and predictive of, subsequent virologic suppression. We show in a multivariate model controlling for baseline CD4+ T-cell count that decreases in CD38 expression on CD4+ T cells (with a similar trend for decreased expression of CD95 on CD4+ T cells) after 2 weeks of salvage therapy were predictive of an increased likelihood of achieving virologic suppression. Importantly, these changes in activation occurred independent of viral suppression, because, at week 2, when measures of immune activation were improved, 64% of patients still had detectable viral replication. These data support the hypothesis that early measures of immune activation can be used to predict patients who ultimately suppress viral replication after initiation of salvage therapy. When compared with measures of CD4+ T-cell count and viral load, which are measured at much later time points after initiation of salvage therapy (commonly 1 to 3 months later), the prognostic value of early measures of CD38 and CD95 expression by CD4+ T cells may provide a distinct clinical advantage by allowing earlier assessment of the response to changes in ART.

The results of this study further highlight the relation between HIV disease progression, immune dysfunction, and T-cell activation, because we show that persistent immune activation predicts ongoing viral replication and progressive dysfunction of the CD4+ T-cell population.19,2426 In a study of CD4+ T-cell recovery in HIV-1–infected patients after 12 months of highly active antiretroviral therapy (HAART), incomplete CD4+ T-cell recovery, despite suppression of viral replication with HAART, was associated with ongoing CD4+ T-cell activation.27 Persistent CD4+ but not CD8+ T-cell activation was associated with decreased CD4+ T-cell recovery during the first 3 months of HAART.28 Similarly, several recent studies have further confirmed the relation between limited immune restoration and higher levels of CD4+ T-cell activation.11,17,29,30 Each of these studies examined the relation between T-cell activation and subsequent immunologic recovery, however, without specifically examining direct relations between T-cell activation and virologic suppression.

Our data, and those of previous reports, suggest a link between ongoing viremia and persistent T-cell activation. Several studies have highlighted mechanisms mediating this link. A recent study identified bacterial translocation as a mechanism of chronic immune activation in the context of a compromised gastrointestinal mucosal surface during chronic HIV infection.31 It is also likely that the association between ongoing viremia and persistent T-cell activation occurs in response to virally produced proteins and/or host-produced cytokines that are elaborated during virus-host interactions. Consistent with that hypothesis, CD38- and CD95-mediated T-cell activation can be induced by Tat,3234 Nef,35,36 and gp12034 as well as by altered production of a wide variety of cytokines, such as TNFα, interferon (IFN)-γ, interleukin (IL)-1β, IL-2, IL-6, IL-10, and IL-12, produced by chronically activated CD8+ T cells and macrophages during HIV infection.3742 Consequently, as levels of these immunomodulators decrease in conjunction with effective therapy, the levels of CD38 and CD95 decrease.18 Taken together, these data demonstrate the potential clinical utility of early measures of activation (changes in CD95 and/or CD38 between baseline and week 2) as predictors of virologic suppression after initiation of salvage therapy.

Similarly, different levels of CD4+ T-cell activation at baseline were recently reported in another study. Baseline proportions of CD4+ T-cell activation were not significantly different among immunologically concordant versus discordant patients before initiation of ART.17 When immune cell subsets were measured by absolute cell counts, however, discordant patients showed increased activation of CD4+ and CD8+ T cells at baseline, suggesting that baseline measurements of activation may prove clinically useful in predicting CD4+ T-cell recovery.17 Follow-up immunophenotypic data were measured after 48 weeks of successful (HIV viral load ≤50 copies/mL) ART,17 and there was no comparison of those patients who achieved virologic suppression versus those who did not. Similarly, AIDS Clinical Trials Group (ACTG) 384 showed that persistent T-cell activation was associated with decreased T-cell recovery after ART in treatment-naive patients.43 The authors acknowledge that because CD4+ T-cell subsets were first assessed 6 months after treatment initiation, they “may have missed early changes”.43

Several limitations must be considered in appropriate interpretation of the data, some of which suggest further areas of research. Two of 34 patients did not complete any follow-up evaluations during the study and were thus excluded from the analysis. The study population was homogeneous, including only middle-aged, primarily white, men. Nonetheless, the results of the study are consistent with those reported in a pediatric study, which showed that the CD8+CD38+ T-cell count is a prognostic marker of therapeutic failure in HIV-infected children.22 There was a trend toward more frequent use of protease inhibitors (PIs) in those therapies that ultimately failed to achieve virologic suppression. Although the present study demonstrated a decline in immune activation markers during early effective salvage therapy, further studies should be initiated to define the generalizability of these findings in diverse patient populations, who are treated with a range of salvage therapies, and to determine whether the predictive ability of these changes are present in naive patients initiating therapy

The present study adds to the body of data that highlights the importance of persistent T-cell activation in HIV pathogenesis.28,44,45 Three features make the present study unique. First, although other studies have focused on the impact of persistent T-cell activation on eventual T-cell recovery, the present study investigated the relation between T-cell activation and eventual virologic suppression. Second, the present study focused on patients undergoing salvage therapy, whereas other studies of T-cell activation and HIV disease progression have typically included only treatment-naive patients. Third, the present study examined early time points (2, 4, 8, 24, and 48 weeks) after initiation of salvage therapy, whereas other studies have generally focused on time points beginning several months after the initiation of an initial anti-retroviral regimen. Based on these early data points, the present study reports that measures comparing T-cell activation between baseline and 2 weeks of salvage ART are sufficient for predicting those individuals who eventually achieve control of HIV replication, which is the true measure of disease progression or control.

Acknowledgments

S. A. Rizza is supported by a grant from the Mayo Program in Translational Immunovirology and Biodefense and a Robert and Arlene Kogod Program on Aging Award from the Mayo Foundation. A. D. Badley is supported by National Institutes of Health grants RO1-AI62261 and RO1-AI40384, and a Burroughs Wellcome Award identification number 1005160. J. B. Angel is an Ontario HIV Treatment Network (OHTN) Career Scientist, and funds from the OHTN were used to perform this study.

Footnotes

AUTHOR CONTRIBUTIONS

A. D. Badley, M. R. Loutfy, F. Mandy, C. M. Kovacs, and M. Bergeron participated in the design and implementation of the study; the collection, compilation, and interpretation of data; and the preparation of the manuscript. V. Govan and J. B. Angel participated in the implementation of the study; the collection, compilation, and interpretation of data; and the preparation of the manuscript. B. D. Shepard and J. Raboud participated in the compilation and interpretation of data and the preparation of the manuscript. C. Diong and S. A. Rizza participated in the data interpretation and manuscript preparation.

A. D. Badley had full access to all the data in the study and had final responsibility for the decision to submit for publication. None of the authors have a commercial association or other association that might pose a conflict of interest. The funding sources had no involvement in the study design; in the collection, analysis, or interpretation of data; in the writing of the manuscript; or in the decision to submit the manuscript for publication.

References

  • 1.Hammer SM, Saag MS, Schechter M, et al. Treatment for adult HIV infection: 2006 recommendations of the International AIDS Society-USA Panel. JAMA. 2006;296:827–843. doi: 10.1001/jama.296.7.827. [DOI] [PubMed] [Google Scholar]
  • 2.Clavel F, Hance AJ. HIV drug resistance. N Engl J Med. 2004;350:1023–1035. doi: 10.1056/NEJMra025195. [DOI] [PubMed] [Google Scholar]
  • 3.Coffin JM. HIV population dynamics in vivo: implications for genetic variation, pathogenesis, and therapy. Science. 1995;267:483–489. doi: 10.1126/science.7824947. [DOI] [PubMed] [Google Scholar]
  • 4.Hatano H, Hunt P, Weidler J, et al. Rate of viral evolution and risk of losing future drug options in heavily pretreated, HIV-infected patients who continue to receive a stable, partially suppressive treatment regimen. Clin Infect Dis. 2006;43:1329–1336. doi: 10.1086/508655. [DOI] [PubMed] [Google Scholar]
  • 5.Hirsch MS, Brun-Vezinet F, Clotet B, et al. Antiretroviral drug resistance testing in adults infected with human immunodeficiency virus type 1: 2003 recommendations of an International AIDS Society-USA Panel. Clin Infect Dis. 2003;37:113–128. doi: 10.1086/375597. [DOI] [PubMed] [Google Scholar]
  • 6.Ormaasen V, Sandvik L, Asjo B, et al. An algorithm-based genotypic resistance score is associated with clinical outcome in HIV-1-infected adults on antiretroviral therapy. HIV Med. 2004;5:400–406. doi: 10.1111/j.1468-1293.2004.00244.x. [DOI] [PubMed] [Google Scholar]
  • 7.Zaccarelli M, Tozzi V, Lorenzini P, et al. Multiple drug class-wide resistance associated with poorer survival after treatment failure in a cohort of HIV-infected patients. AIDS. 2005;19:1081–1089. doi: 10.1097/01.aids.0000174455.01369.ad. [DOI] [PubMed] [Google Scholar]
  • 8.Lorenzi P, Opravil M, Hirschel B, et al. Impact of drug resistance mutations on virologic response to salvage therapy. Swiss HIV Cohort Study. AIDS. 1999;13:F17–F21. doi: 10.1097/00002030-199902040-00001. [DOI] [PubMed] [Google Scholar]
  • 9.Temesgen Z, Cainelli F, Poeschla EM, et al. Approach to salvage antiretroviral therapy in heavily antiretroviral-experienced HIV-positive adults. Lancet Infect Dis. 2006;6:496–507. doi: 10.1016/S1473-3099(06)70550-3. [DOI] [PubMed] [Google Scholar]
  • 10.Aukrust P, Muller F, Lien E, et al. Tumor necrosis factor (TNF) system levels in human immunodeficiency virus-infected patients during highly active antiretroviral therapy: persistent TNF activation is associated with virologic and immunologic treatment failure. J Infect Dis. 1999;179:74–82. doi: 10.1086/314572. [DOI] [PubMed] [Google Scholar]
  • 11.Benito JM, Lopez M, Lozano S, et al. Differential upregulation of CD38 on different T-cell subsets may influence the ability to reconstitute CD4+ T cells under successful highly active antiretroviral therapy. J Acquir Immune Defic Syndr. 2005;38:373–381. doi: 10.1097/01.qai.0000153105.42455.c2. [DOI] [PubMed] [Google Scholar]
  • 12.Beran O, Holub M, Spala J, et al. CD38 expression on CD8+ T cells in human immunodeficiency virus 1-positive adults treated with HAART. Acta Virol. 2003;47:121–124. [PubMed] [Google Scholar]
  • 13.Bofill M, Mocroft A, Lipman M, et al. Increased numbers of primed activated CD8+CD38+CD45RO+ T cells predict the decline of CD4+ T cells in HIV-1-infected patients. AIDS. 1996;10:827–834. doi: 10.1097/00002030-199607000-00005. [DOI] [PubMed] [Google Scholar]
  • 14.Brazille P, Dereuddre-Bosquet N, Leport C, et al. Decreases in plasma TNF-alpha level and IFN-gamma mRNA level in peripheral blood mononuclear cells (PBMC) and an increase in IL-2 mRNA level in PBMC are associated with effective highly active antiretroviral therapy in HIV-infected patients. Clin Exp Immunol. 2003;131:304–311. doi: 10.1046/j.1365-2249.2003.02064.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Burgisser P, Hammann C, Kaufmann D, et al. Expression of CD28 and CD38 by CD8+ T lymphocytes in HIV-1 infection correlates with markers of disease severity and changes towards normalization under treatment. The Swiss HIV Cohort Study. Clin Exp Immunol. 1999;115:458–463. doi: 10.1046/j.1365-2249.1999.00818.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Chun TW, Justement JS, Sanford C, et al. Relationship between the frequency of HIV-specific CD8+ T cells and the level of CD38+CD8+ T cells in untreated HIV-infected individuals. Proc Natl Acad Sci USA. 2004;101:2464–2469. doi: 10.1073/pnas.0307328101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Goicoechea M, Smith DM, Liu L, et al. Determinants of CD4+ T cell recovery during suppressive antiretroviral therapy: association of immune activation, T cell maturation markers, and cellular HIV-1 DNA. J Infect Dis. 2006;194:29–37. doi: 10.1086/504718. [DOI] [PubMed] [Google Scholar]
  • 18.Koblavi-Deme S, Maran M, Kabran N, et al. Changes in levels of immune activation and reconstitution markers among HIV-1-infected Africans receiving antiretroviral therapy. AIDS. 2003;17(Suppl 3):S17–S22. doi: 10.1097/00002030-200317003-00003. [DOI] [PubMed] [Google Scholar]
  • 19.Liu Z, Cumberland WG, Hultin LE, et al. 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 Retrovirol. 1997;16:83–92. doi: 10.1097/00042560-199710010-00003. [DOI] [PubMed] [Google Scholar]
  • 20.Ondoa P, Koblavi-Deme S, Borget MY, et al. Assessment of CD8 T cell immune activation markers to monitor response to antiretroviral therapy among HIV-1 infected patients in Cote d’Ivoire. Clin Exp Immunol. 2005;140:138–148. doi: 10.1111/j.1365-2249.2005.02734.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Schupbach J, Boni J, Bisset LR, et al. HIV-1 p24 antigen is a significant inverse correlate of CD4 T-cell change in patients with suppressed viremia under long-term antiretroviral therapy. J Acquir Immune Defic Syndr. 2003;33:292–299. doi: 10.1097/00126334-200307010-00002. [DOI] [PubMed] [Google Scholar]
  • 22.Resino S, Bellon JM, Gurbindo MD, et al. CD38 expression in CD8+ T cells predicts virological failure in HIV type 1-infected children receiving antiretroviral therapy. Clin Infect Dis. 2004;38:412–417. doi: 10.1086/380793. [DOI] [PubMed] [Google Scholar]
  • 23.Bergeron M, Faucher S, Ding T, et al. Evaluation of a universal template for single-platform absolute T-lymphocyte subset enumeration. Cytometry. 2002;50:62–68. doi: 10.1002/cyto.10089. [DOI] [PubMed] [Google Scholar]
  • 24.Froebel KS, Raab GM, D’Alessandro C, et al. A single measurement of CD38CD8 cells in HIV+, long-term surviving injecting drug users distinguishes those who will progress to AIDS from those who will remain stable. Clin Exp Immunol. 2000;122:72–78. doi: 10.1046/j.1365-2249.2000.01348.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Giorgi JV, Hultin LE, McKeating JA, et al. Shorter survival in advanced human immunodeficiency virus type 1 infection is more closely associated with T lymphocyte activation than with plasma virus burden or virus chemokine coreceptor usage. J Infect Dis. 1999;179:859–870. doi: 10.1086/314660. [DOI] [PubMed] [Google Scholar]
  • 26.Plaeger S, Bass HZ, Nishanian P, et al. The prognostic significance in HIV infection of immune activation represented by cell surface antigen and plasma activation marker changes. Clin Immunol. 1999;90:238–246. doi: 10.1006/clim.1998.4646. [DOI] [PubMed] [Google Scholar]
  • 27.Anthony KB, Yoder C, Metcalf JA, et al. Incomplete CD4 T-cell recovery in HIV-1 infection after 12 months of highly active antiretroviral therapy is associated with ongoing increased CD4 T-cell activation and turnover. J Acquir Immune Defic Syndr. 2003;33:125–133. doi: 10.1097/00126334-200306010-00002. [DOI] [PubMed] [Google Scholar]
  • 28.Hunt PW, Martin JN, Sinclair E, et al. T cell activation is associated with lower CD4+ T cell gains in human immunodeficiency virus-infected patients with sustained viral suppression during antiretroviral therapy. J Infect Dis. 2003;187:1534–1543. doi: 10.1086/374786. [DOI] [PubMed] [Google Scholar]
  • 29.Benito JM, Lopez M, Lozano S, et al. CD4+ T cell recovery beyond the first year of complete suppression of viral replication during highly active antiretroviral therapy is not influenced by CD8+ T cell activation. J Infect Dis. 2005;192:2142–2146. doi: 10.1086/498168. [DOI] [PubMed] [Google Scholar]
  • 30.Delobel P, Nugeyre MT, Cazabat M, et al. Naive T-cell depletion related to infection by X4 human immunodeficiency virus type 1 in poor immunological responders to highly active antiretroviral therapy. J Virol. 2006;80:10229–10236. doi: 10.1128/JVI.00965-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Brenchley JM, Price DA, Schacker TW, et al. Microbial translocation is a cause of systemic immune activation in chronic HIV infection. Nat Med. 2006;12:1365–1371. doi: 10.1038/nm1511. [DOI] [PubMed] [Google Scholar]
  • 32.Li-Weber M, Laur O, Dern K, et al. T cell activation-induced and HIV tat-enhanced CD95(APO-1/Fas) ligand transcription involves NF-kappaB. Eur J Immunol. 2000;30:661–670. doi: 10.1002/1521-4141(200002)30:2<661::AID-IMMU661>3.0.CO;2-L. [DOI] [PubMed] [Google Scholar]
  • 33.Li CJ, Friedman DJ, Wang C, et al. Induction of apoptosis in uninfected lymphocytes by HIV-1 Tat protein. Science. 1995;268:429–431. doi: 10.1126/science.7716549. [DOI] [PubMed] [Google Scholar]
  • 34.Westendorp MO, Frank R, Ochsenbauer C, et al. Sensitization of T cells to CD95-mediated apoptosis by HIV-1 Tat and gp120. Nature. 1995;375:497–500. doi: 10.1038/375497a0. [DOI] [PubMed] [Google Scholar]
  • 35.Zauli G, Gibellini D, Secchiero P, et al. Human immunodeficiency virus type 1 Nef protein sensitizes CD4(+) T lymphoid cells to apoptosis via functional upregulation of the CD95/CD95 ligand pathway. Blood. 1999;93:1000–1010. [PubMed] [Google Scholar]
  • 36.Xu XN, Laffert B, Screaton GR, et al. Induction of Fas ligand expression by HIV involves the interaction of Nef with the T cell receptor zeta chain. J Exp Med. 1999;189:1489–1496. doi: 10.1084/jem.189.9.1489. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Boyle MJ, Berger MF, Tschuchnigg M, et al. Increased expression of interferon-gamma in hyperplastic lymph nodes from HIV-infected patients. Clin Exp Immunol. 1993;92:100–105. doi: 10.1111/j.1365-2249.1993.tb05954.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Cohen OJ, Kinter A, Fauci AS. Host factors in the pathogenesis of HIV disease. Immunol Rev. 1997;159:31–48. doi: 10.1111/j.1600-065x.1997.tb01005.x. [DOI] [PubMed] [Google Scholar]
  • 39.Esser R, von Briesen H, Brugger M, et al. Secretory repertoire of HIV-infected human monocytes/macrophages. Pathobiology. 1991;59:219–222. doi: 10.1159/000163649. [DOI] [PubMed] [Google Scholar]
  • 40.Graziosi C, Pantaleo G, Gantt KR, et al. Lack of evidence for the dichotomy of TH1 and TH2 predominance in HIV-infected individuals. Science. 1994;265:248–252. doi: 10.1126/science.8023143. [DOI] [PubMed] [Google Scholar]
  • 41.Jassoy C, Walker BD. HIV-1-specific cytotoxic T lymphocytes and the control of HIV-1 replication. Springer Semin Immunopathol. 1997;18:341–354. doi: 10.1007/BF00813502. [DOI] [PubMed] [Google Scholar]
  • 42.Poli G, Fauci AS. Role of cytokines in the pathogenesis of human immunodeficiency virus infection. In: Aggarwal B, Puri R, editors. Human Cytokines: Their Role in Disease and Therapy. Cambridge, United Kingdom: Blackwell Science; 1995. pp. 421–449. [Google Scholar]
  • 43.Gandhi RT, Spritzler J, Chan E, et al. Effect of baseline- and treatment-related factors on immunologic recovery after initiation of antiretroviral therapy in HIV-1–positive subjects: results from ACTG 384. J Acquir Immune Defic Syndr. 2006;42:426–434. doi: 10.1097/01.qai.0000226789.51992.3f. [DOI] [PubMed] [Google Scholar]
  • 44.Hazenberg MD, Hamann D, Schuitemaker H, et al. T cell depletion in HIV-1 infection: how CD4+ T cells go out of stock. Nat Immunol. 2000;1:285–289. doi: 10.1038/79724. [DOI] [PubMed] [Google Scholar]
  • 45.McCune JM. The dynamics of CD4+ T-cell depletion in HIV disease. Nature. 2001;410:974–979. doi: 10.1038/35073648. [DOI] [PubMed] [Google Scholar]

RESOURCES