Recently, Lu and Andrieu1 stated that the HIV-1 protease inhibitors (PIs) indinavir and saquinavir used in concentrations at least 30-fold lower than those needed for 90% viral inhibition apparently do not influence sensitivity of peripheral blood T cells from HIV-1–infected individuals toward apoptosis induced either by T-cell receptor/CD3 ligation or by direct triggering of the CD95 receptor in vitro.1 They conclude from their data that the beneficial effect of PI treatment on clinical and immunological parameters in HIV-1–infected patients, which can be encountered even in the absence of relevant virological effects, are not related to changes in T-cell apoptosis.

In our own study in a cohort of HIV-1–infected children and adolescents, we had observed an increased sensitivity of freshly isolated T cells toward apoptosis,2 which was rapidly down-regulated following initiation of highly active antiretroviral therapy including PIs (HAART).3 Here we report that prolonged PI treatment in vivo reduced sensitivity of peripheral blood T cells toward apoptosis in vitro even when plasma viral load levels were not decreased and susceptibility for T-cell activation in these patients was not down-regulated.

Between January 1996 and January 1999, a total of 47 HIV-1–infected children and adolescents and 28 age-matched healthy controls were studied. Patients were stratified according to treatment modalities at the time of the immunological investigation (Table1). The study was conducted according to the Declaration of Helsinki and was approved by the ethical committee of the University Hospital in Heidelberg. T-cell phenotyping, plasma viral load quantification, and assessment of T-cell sensitivity toward apoptosis and activation was performed as previously described.3 Patients and controls and/or their relatives gave informed consent prior to venipuncture.

Table 1.

Clinical data of HIV-1–infected patients stratified according to the actual antiretroviral treatment regimen at the time of the study

Patient group
(Treatment)
I
(no therapy)
II
(2 RT inhibitors)
III
(HAART)
29 20 16  
Age (y) 5 (0.5-28) 7 (1-16) 10 (2-21)  
CD4 count (%) 20 ± 2 23 ± 3  25 ± 2   
CD4 count (cells per μL) 697 ± 87 977 ± 242 766 ± 143  
“Viral load”
(log10 RNA copies per mL) 
4.54
(2.70-5.76)
[n = 14]* 
4.81
(1.90-6.53) 
5.13
(1.48-5.87) 
Duration of therapy (mo) NA 11 (7-20) 10 (7-22)  
Number of patients with “viral load” less than 3.0 log10copies per mL (%) 2 (14) 7 (35) 7 (43) 
Patient group
(Treatment)
I
(no therapy)
II
(2 RT inhibitors)
III
(HAART)
29 20 16  
Age (y) 5 (0.5-28) 7 (1-16) 10 (2-21)  
CD4 count (%) 20 ± 2 23 ± 3  25 ± 2   
CD4 count (cells per μL) 697 ± 87 977 ± 242 766 ± 143  
“Viral load”
(log10 RNA copies per mL) 
4.54
(2.70-5.76)
[n = 14]* 
4.81
(1.90-6.53) 
5.13
(1.48-5.87) 
Duration of therapy (mo) NA 11 (7-20) 10 (7-22)  
Number of patients with “viral load” less than 3.0 log10copies per mL (%) 2 (14) 7 (35) 7 (43) 

Group I had no specific antiretroviral therapy, group II had at least 6 months of treatment with 2 nucleosidic inhibitors of HIV-1 reverse transcriptase (RT inhibitors), and group III had at least 6 months of treatment with 2 RT inhibitors and at least 1 inhibitor of HIV-1 protease (HAART). Sixteen patients were studied before and after changes in their therapeutic regimen and were included in both respective patient groups; 4 patients were studied repeatedly and were included in all 3 groups. The remaining 21 patients were studied only once. Data are given either as median values (range) or as arithmetic mean ± SEM.

AZT indicates zidovudine; d4T, stavudine; ddC, zalcitabine; ddI, didanosine; 3TC, lamivudine; IDV, indinavir; NFV, nelfinavir; SQV, saquinavir; RTV, ritonavir; and NA, not applicable.

*

Plasma viral load levels were not available for all patients; the actual number is given in brackets.

Specific drug combinations: 9 patients received AZT + 3TC; 8, AZT + ddI; 2, AZT + ddC; and 1, d4T + 3TC.

Specific drug combinations: 3 patients received d4T + 3TC + IDV; 4, d4T + ddI + NFV; 3, d4T + 3TC + NFV + SQV; 1, d4T + 3TC + NFV;1, AZT + 3TC + NFV; 1, AZT + ddI + IDV; 1, d4T + NFV + SQV; 1, d4T + ddI + NFV + SQV; and 1, d4T + ddI + RTV + SQV.

We compared T-cell apoptosis and T-cell activation (CD69 expression) in patients on HAART for more than 6 months (group III; n = 16) to patients without therapy (group I; n = 29), patients treated with HIV-1 reverse transcriptase inhibitors (RTIs) alone (group II; n = 20) and healthy controls (CTRLs; n = 28) by analysis of variance using the InStat statistical analysis software package (GraphPad Software, San Diego, CA). As shown in Figure1, the sensitivity of T cells toward anti-CD95–induced apoptosis was significantly lower in group III than in group II (Figure 1B; CD4+ T cells, 3 ± 1% versus 26 ± 5%, P < .001; CD8+ T cells, 5 ± 1% versus 24 ± 5%, P < .001) despite similar plasma viral load levels in both patient groups. Spontaneous T-cell death did not differ between these groups (Figure 1A; CD4+T cells, 10 ± 2% versus 17 ± 4%, P = NS; CD8+ T cells, 12 ± 3% versus 15 ± 2%,P = NS).

Fig. 1.

Effect of different antiretroviral treatment regimens on apoptosis and CD69 expression in CD4+ and CD8+T cell subpopulations in HIV-1–infected patients and healthy controls.

(A) Spontaneous apoptosis, (B) anti-CD95–induced apoptosis, (C) anti-CD3–induced apoptosis, (D) spontaneous CD69 expression, (E) anti-CD95–induced CD69 expression, (F) anti-CD3–induced CD69 expression. ● indicates CD4+ T cell subpopulations, and ○, CD8+ T cell subpopulations. Horizontal bars indicate the group median. Data from HIV-1–infected patients receiving either no effective antiretroviral therapy (group I, n = 26) or at least 6 months of treatment with 2 inhibitors of HIV-1 reverse transcriptase (RTIs; group II, n = 20) or 2 RTIs plus at least 1 inhibitor of HIV-1 protease (HAART; group III, n = 16) were compared to 28 age-matched healthy controls (CTRLs). The respective patient numbers for measurements of CD69 expression were group I, n = 11; group II, n = 20; group III, n = 16; and CTRL, n = 12. Methods: Freshly isolated peripheral blood mononuclear cells (PBMCs) (2 × 105 cells/well) were cultured in the presence or absence of either anti-CD3 monoclonal antibodies (moAbs; OKT3; 10 μg/mL) or anti-CD95 moAb (anti–APO-1; 10 μg/mL) and protein A (5 ng/mL) as described.3 Cells were collected after 21 ± 1 hours and stained for surface expression of CD4, CD8, and CD69 using fluorochrome-labeled moAbs (Becton Dickinson, Heidelberg, Germany), and cell death was determined in CD4+ and CD8+ T cells by flow cytometry. The percentage of specific anti-CD3– and anti-CD95–induced apoptosis was calculated by 100 × (% of experimentally induced cell death − % of spontaneous cell death)/(100 − % of spontaneous cell death). Spontaneous and experimentally induced CD69 expression on viable CD4+ and CD8+ T cells was determined as a marker for T-cell activation as described,3 specific anti-CD3– and anti-CD95–induced CD69 expression on both T-cell subpopulations was calculated using the formula for specific cell death.

Fig. 1.

Effect of different antiretroviral treatment regimens on apoptosis and CD69 expression in CD4+ and CD8+T cell subpopulations in HIV-1–infected patients and healthy controls.

(A) Spontaneous apoptosis, (B) anti-CD95–induced apoptosis, (C) anti-CD3–induced apoptosis, (D) spontaneous CD69 expression, (E) anti-CD95–induced CD69 expression, (F) anti-CD3–induced CD69 expression. ● indicates CD4+ T cell subpopulations, and ○, CD8+ T cell subpopulations. Horizontal bars indicate the group median. Data from HIV-1–infected patients receiving either no effective antiretroviral therapy (group I, n = 26) or at least 6 months of treatment with 2 inhibitors of HIV-1 reverse transcriptase (RTIs; group II, n = 20) or 2 RTIs plus at least 1 inhibitor of HIV-1 protease (HAART; group III, n = 16) were compared to 28 age-matched healthy controls (CTRLs). The respective patient numbers for measurements of CD69 expression were group I, n = 11; group II, n = 20; group III, n = 16; and CTRL, n = 12. Methods: Freshly isolated peripheral blood mononuclear cells (PBMCs) (2 × 105 cells/well) were cultured in the presence or absence of either anti-CD3 monoclonal antibodies (moAbs; OKT3; 10 μg/mL) or anti-CD95 moAb (anti–APO-1; 10 μg/mL) and protein A (5 ng/mL) as described.3 Cells were collected after 21 ± 1 hours and stained for surface expression of CD4, CD8, and CD69 using fluorochrome-labeled moAbs (Becton Dickinson, Heidelberg, Germany), and cell death was determined in CD4+ and CD8+ T cells by flow cytometry. The percentage of specific anti-CD3– and anti-CD95–induced apoptosis was calculated by 100 × (% of experimentally induced cell death − % of spontaneous cell death)/(100 − % of spontaneous cell death). Spontaneous and experimentally induced CD69 expression on viable CD4+ and CD8+ T cells was determined as a marker for T-cell activation as described,3 specific anti-CD3– and anti-CD95–induced CD69 expression on both T-cell subpopulations was calculated using the formula for specific cell death.

Close modal

Although T cells from HAART-treated patients (group III) had a normal sensitivity toward anti-CD95–induced apoptosis in vitro (P = NS when compared to the control; Figure 1B), the sensitivity toward anti-CD3–induced CD69 expression in both CD4+ and CD8+ T-cell subpopulations was significantly higher in group III than in all other groups of patients and the controls (Figure 1E, P < .001). Anti-CD3–induced CD69 expression on CD4+ and CD8+ T cells tended to increase in HAART-treated patients (group III) with detectable, compared to those with undetectable, plasma viral load, although these differences were statistically not significant (CD4+ T cells, 71 ± 8% versus 51 ± 7%, P = NS; CD8+ T cells, 69 ± 8% versus 52 ± 6%,P = NS). Spontaneous CD69 expression in T cells from PI-treated patients in group III was increased compared to other treatment groups and healthy controls (28 ± 7% on CD4+and 27 ± 4% on CD8+ T cells; P < .001); see Figure 1D.

Our observations suggest that the reduced T-cell sensitivity toward CD95-induced apoptosis in vitro might partly explain the observation that an increasing number of patients with virological rebound on PI therapy maintain stable CD4 counts and do not experience clinical deterioration as fast as one could expect from the increased viral load. The CD95-resistant phenotype of peripheral blood T cells in these patients may be due to a decreased viral “fitness” or a direct inhibition of apoptosis pathways (eg, at the level of proapoptotic proteases).4 The fact that these T cells are still hypersensitive toward anti-CD3–induced activation argues against the hypothesis of Lu and Andrieu that the restoration of a normal apoptosis sensitivity is simply an epiphenomenon of the general immunological recovery in HIV-infected individuals receiving HAART.

Böhler et al's comments invoke the fundamental issue: what is the interrelation between T-cell activation, anergy, and apoptosis (AAA) in HIV-infected patients treated with highly active antiretroviral therapy (HAART). We agree that this issue is important enough to deserve a deeper discussion.

Recently, we1-1 and others1,2 have reported an apparent dissociation between T-cell recovery and plasma viral-load suppression in HIV-infected patients under HAART. In addition, Böhler et al also observed a reduced sensitivity of HAART-treated patients' T cells to apoptosis that was independent from viral-load response. This dissociation implies a direct effect of HAART on patients' immune systems rather than “a decreased viral fitness” as suggested by Böhler et al, because the immune recovery in patients experiencing virologic failure could not be maintained after withdrawal of HAART (Table 1-1).

Table 1-1.

CD4+ and CD8+ T-cell counts in 5 HIV-infected patients experiencing virologic nonresponse under 3 years of HAART and during temporary withdrawal of treatment

Patients2-1-150Peripheral blood T-cell countPlasma viral load
(log10 eq copies/mL)
CD4 cells/μLCD8 cells/μL
Patient 88    
 At entry 36 425 4.99  
 Month 12 283 979 5.04 
 Month 36 380 1656 4.68  
 Month 382-1-151 147 878 4.70  
 Month 41 385 1128 4.26 
Patient 89    
 At entry 43 225 5.22 
 Month 12 136 707 4.36  
 Month 36 233 891 5.31  
 Month 402-1-151 69 646 4.89 
 Month 42 162 915 4.77  
Patient 90    
 At entry 81 615 4.24  
 Month 12 237 1204 4.51 
 Month 36 374 1844 5.23  
 Month 392-1-151 144 943 5.28  
 Month 42 294 1544 4.79 
Patient 93    
 At entry 155 900 4.79 
 Month 12 274 1243 4.72  
 Month 36 229 2564 4.13  
 Month 432-1-151 170 1542 4.93 
 Month 45 401 2517 4.28  
Patient 96    
 At entry 213 259 4.47  
 Month 12 329 414 4.10  
 Month 36 567 873 4.53 
 Month 422-1-151 226 449 5.15  
 Month 44 549 646 5.18 
Patients2-1-150Peripheral blood T-cell countPlasma viral load
(log10 eq copies/mL)
CD4 cells/μLCD8 cells/μL
Patient 88    
 At entry 36 425 4.99  
 Month 12 283 979 5.04 
 Month 36 380 1656 4.68  
 Month 382-1-151 147 878 4.70  
 Month 41 385 1128 4.26 
Patient 89    
 At entry 43 225 5.22 
 Month 12 136 707 4.36  
 Month 36 233 891 5.31  
 Month 402-1-151 69 646 4.89 
 Month 42 162 915 4.77  
Patient 90    
 At entry 81 615 4.24  
 Month 12 237 1204 4.51 
 Month 36 374 1844 5.23  
 Month 392-1-151 144 943 5.28  
 Month 42 294 1544 4.79 
Patient 93    
 At entry 155 900 4.79 
 Month 12 274 1243 4.72  
 Month 36 229 2564 4.13  
 Month 432-1-151 170 1542 4.93 
 Month 45 401 2517 4.28  
Patient 96    
 At entry 213 259 4.47  
 Month 12 329 414 4.10  
 Month 36 567 873 4.53 
 Month 422-1-151 226 449 5.15  
 Month 44 549 646 5.18 

HIV indicates human immunodeficiency virus; HAART, highly active antiretroviral therapy.

F2-1-150

Patients whose characteristics and HAART regimen were reported previously.1-1 

F2-1-151

Patients' samples collected within 1-2 months after withdrawal of their HAART regimen.

In general, the immune recovery under HAART is characterized by an increase of CD4+ and CD8+ T-cell counts associated with an overall down-regulation of T-cell AAA in vivo. To learn what the effects of HAART on the AAA are, we have conducted an in vitro study to investigate the AAA kinetics of patients' peripheral blood mononuclear cells (PBMCs) following immune stimuli in the absence or the presence of HAART components (nucleoside reverse trascriptase inhibitors [NRTIs] or protease inhibitors [PIs]).1-1 Results showed that PIs (indinavir or saquinavir), even at nonantiviral doses, allowed an early (4 days after activation) reversal of T-cell anergy (ie, restoration of T-cell proliferation) but a later (14 days after activation) down-regulation of T-cell apoptosis. Therefore, we concluded that reversal of T-cell anergy should be the primary action of PIs responsible for the overall immune improvements observed in patients treated with HAART. This finding is in keeping with the increasing evidence that the progression of HIV disease is associated with an impaired T-cell proliferation/production (anergic mechanism) coupled with chronic immune activation and apoptosis (ie, reduced half-life of T cells) rather than with an increased T-cell turnover (overproduction or exhaustion hypothesis).1-6-1-10 

An increased proportion of HIV-infected patients' T cells undergoing apoptosis following APO-1/Fas (CD95) ligation reflects, indeed, a subpopulation of T cells chronically activated in vivo, because purified quiescent T cells (G0) showed nearly 100% resistance to apoptosis triggered by APO-1/Fas ligation while the proportion of T cells susceptible to Fas-mediated apoptosis increased gradually after 7 days of stimulation with anti-CD3 and anti-CD28 monoclonal antibodies (Lu and Andrieu, unpublished observation, 2000). Normalized sensitivity of T cells to apoptosis in PI-treated (but not NRTI-treated) patients as reported by Böhler et al could thus be the consequence of early restoration of T-cell proliferation and subsequent down-regulation of T-cell activation by PIs.

Böhler et al's key argument challenging the normalization of T-cell proliferation/activation as the primary event involved in HAART (ie, PI)–induced immune recovery is based on their observation that CD69 expression on T cells following TCR/CD3 stimulation was significantly enhanced in HAART-treated patients. In fact, this observation supports further the PI-mediated enhancement of T-cell proliferation/differentiation following immune stimuli, because CD69 expression on T cells upon to TCR/CD3 stimulation was shown to be correlated positively with proliferation and effector function of T cells.1-11 

In conclusion, anergized status of HIV-infected patients' T cells permits unresolved chronic activation and apoptosis throughout the course of infection, thus contributing to the eventual development of AIDS. PIs, even at nonantiviral concentrations, are capable of restoring T-cell proliferation and in consequence normalizing     T-cell activation and apoptosis in HAART-treated patients. These findings are instrumental in improving our understanding of HIV immunopathology and in guiding (or designing) immune-based therapeutic intervention, particularly in patients experiencing viro- logic failure and/or in those suffering permanently from side effects of antiviral drugs.

References

1-1
Lu
W
Andrieu
JM
HIV protease inhibitors restore impaired T-cell proliferative response in vivo and in vitro: a viral-suppression-independent mechanism.
Blood.
96
2000
250
258
1-2
Kaufmann
D
Pantaleo
G
Sudre
P
Telenti
A
CD4-cell count in HIV-1-infected individuals remaining viraemic with highly active antiretroviral therapy (HAART): Swiss HIV cohort study.
Lancet.
351
1998
723
724
1-3
Levitz
SM
Improvement in CD4+ cell counts despite persistently detectable HIV load.
N Engl J Med.
338
1998
1074
1075
1-4
Ledergerber
B
Egger
M
Opravil
M
et al
Clinical progression and virological failure on highly active antiretroviral therapy in HIV-1 patients: a prospective cohort study: SwissHIV cohort study.
Lancet.
353
1999
863
868
1-5
Grabar
S
Moing
VL
Goujard
C
et al
Clinical outcome of patients with HIV-1 infection according to immunologic and virologic response after 6 months of highly active antiretroviral therapy.
Ann Intern Med.
133
2000
401
410
1-6
Andersson
J
Fehniger
TE
Patterson
BK
et al
Early reduction of immune activation in lymphoid tissue following highly active HIV therapy.
AIDS.
12
1998
F123
F129
1-7
Bofill
M
Borthwick
NJ
Simmonds
HA
Novel mechanism for the impairment of cell proliferation in HIV-1 infection.
Immunol Today.
20
1999
258
261
1-8
Hellerstein
M
Hanley
MB
Cesar
D
et al
Directly measured kinetics of circulating T lymphocytes in normal and HIV-1-infected humans.
Nat Med.
5
1999
83
89
1-9
McCune
JM
Hanley
MB
Cesar
D
et al
Factors influencing T-cell turnover in HIV-1-seropositive patients.
J Clin Invest.
105
2000
R1
R8
1-10
Fleury
S
Rizzardi
GP
Chapuis
A
et al
Long-term kinetics of T cell production in HIV-infected subjects treated with highly active antiretroviral therapy.
Proc Natl Acad Sci U S A.
97
2000
5393
5398
1-11
Nielsen
SD
Afzelius
P
Ersboll
AK
Nielsen
JO
Hansen
JE
Expression of the activation antigen CD69 predicts functionality of in vitro expanded peripheral blood mononuclear cells (PBMC) from healthy donors and HIV-infected patients.
Clin Exp Immunol.
114
1998
66
72
1
Lu
W
Andrieu
JM
HIV protease inhibitors restore impaired T-cell proliferative response in vivo and in vitro: a viral-suppression-independent mechanism.
Blood.
96
2000
250
258
2
Bäumler
CB
Böhler
T
Herr
I
Benner
A
Krammer
PH
Debatin
KM
Activation of the CD95 (APO-1/Fas) system in T cells from human immunodeficiency virus type-1-infected children.
Blood.
88
1996
1741
1746
3
Böhler
T
Walcher
J
Hölzl-Wenig
G
et al
Early effects of antiretroviral combination therapy on activation, apoptosis and regeneration of T cells in HIV-1–infected children and adolescents.
AIDS.
13
1999
779
789
4
Sloand
EM
Kumar
PN
Kim
S
Chaudhuri
A
Weichold
FF
Young
NS
Human immunodeficiency virus type 1 protease inhibitor modulates activation of peripheral blood CD4+ T cells and decreases their susceptibility to apoptosis in vitro and in vivo.
Blood.
94
1999
1021
1027
Sign in via your Institution