metricas
covid
Buscar en
Enfermedades Infecciosas y Microbiología Clínica
Toda la web
Inicio Enfermedades Infecciosas y Microbiología Clínica Efficacy of recombinant interleukin-2 (rIL-2) in patients with advanced HIV-1 in...
Información de la revista
Vol. 26. Núm. 1.
Páginas 27-31 (enero 2008)
Compartir
Compartir
Descargar PDF
Más opciones de artículo
Visitas
4062
Vol. 26. Núm. 1.
Páginas 27-31 (enero 2008)
ORIGINALES BREVES
Acceso a texto completo
Efficacy of recombinant interleukin-2 (rIL-2) in patients with advanced HIV-1 infection and blunted immune response to HAART
Eficacia de la interleucina-2 recombinante (rIL-2) en pacientes con infección avanzada por VIH-1 y escasa respuesta inmunológica al TARGA
Visitas
4062
Manuel Crespoa,
Autor para correspondencia
mcrespo@vhebron.net

Dr. M. Crespo. Infectious Diseases Department. Hospital Universitari Vall d’Hebron. P.° Vall d’Hebron, 119-129. 08035 Barcelona. Spain.
, Isabel Caragolb, Vicenç Falcóa, Esteban Riberaa, Susana Urbanb, Albert Pahissaa
a Infectious Diseases Department. Hospital Universitari Vall d’Hebron, Universitat Autònoma de Barcelona. Spain
b Immunology Unit. Hospital Universitari Vall d’Hebron, Universitat Autònoma de Barcelona. Spain
Este artículo ha recibido
Información del artículo
Resumen
Texto completo
Bibliografía
Descargar PDF
Estadísticas
Tablas (2)
Table 1. Baseline characteristics of 19 HlV-infected patients with poor immune-recovery despite persistent viral suppression under HAART
Table 2. Changes in lymphocyte subsets over the study period
Mostrar másMostrar menos
Objective

The efficacy of recombinant interleukin-2 (rIL-2) was assessed in HIV-infected patients with advanced immune suppression and a discordant immune response to highly active antiretroviral therapy (HAART). The primary endpoint was median change in CD4 + T-cell counts at the end of treatment as compared to baseline. Secondary endpoints were safety and changes in the various T-cell subpopulations.

Material and methods

In a prospective cohort study, 19 patients with HIV-RNA < 50 copies/mL and < 200 CD4 + T cells/mm3 without a significant increase in the previous 12 months were scheduled to receive 6 cycles of 4.5 × 106 IU subcutaneous rIL-2 daily for 5 consecutive days, every 4 weeks.

Results

Median age was 43 years, and 64% had a previous AIDS-defining event. Median nadir and baseline CD4 + cell counts were 36 and 99 cells/mm3, respectively. Three patients discontinued treatment and one experienced grade 4 side effects. CD4 + T-cell counts increased to 147 cells/mm3 (range, 24-285) at 1 month following completion of treatment (P = 0.002), and 180 cells/mm3 (range, 38-280) at 18 months (P < 0.001). This improvement was associated with a significant decrease in expression rates of the activation markers, HLA-DR and CD38.

Conclusion

Our results suggest that in patients with advanced HIV-infection showing a blunted immune response to HAART, rIL-2 might increase the pool of CD4 + T-cells by down-regulating the status of immune activation.

Key words:
Recombinant IL-2
Discordant immune response
Regulatory CD4 + T cells
Objetivo

Evaluamos la eficacia de la interleucina-2 recombinante (rIL-2) en pacientes infectados por VIH, con inmunodepresión avanzada y respuesta immune discordante al tratamiento antirretroviral de gran actividad (TARGA). El objetivo primario de evaluación fue el aumento de los linfocitos CD4 + al final del tratamiento respecto al valor basal. Se evaluó, además, la seguridad y el efecto de la rIL-2 sobre las diferentes poblaciones linfocitarias.

Material y métodos

Estudio de cohorte prospective en el que se incluyeron 19 pacientes con ARN-VIH < 50 y linfocitos CD4 + < 200/mm3, estables en los 12 meses previos. Se administró rIL-2 a dosis de 4,5 × 106 mUI/día por vía subcutánea, cada 4 semanas, hasta completer 6 ciclos.

Resultados

La edad media de los pacientes fue 43 años, el 64% con diagnóstico previo de sida. La media de linfocitos CD4 + basal (99 células/mm3) aumentó hasta 147 células/mm3 (intervalo, 24-285) al final del tratamiento (p = 0,002) y 180 células/mm3 (intervalo, 38-280) 18 meses después de la última dosis de rIL-2. Este aumento se asoció con una disminución en la expresión de los marcadores de activación linfocitaria, HLA-DR y CD38.

Conclusión

Estos resultados sugieren que la rIL-2, en pacientes con infección por VIH avanzada y respuesta inmune discordante al TARGA, puede contribuir a la expansión de los linfocitos CD4 + a través de una disminución de la activación inmunológica.

Palabras claves:
IL-2 recombinante
Respuesta immune discordante
Linfocitos CD4 + reguladores
Texto completo
Introduction

A significant proportion of patients with advanced HIV-infection show incomplete immune reconstitution after initiation of highly active antiretroviral therapy (HAART) and may be at risk for opportunistic infections and clinical progression1. Several trials have shown that various schedules of recombinant interleukin-2 (rIL-2) in combination with HAART safely increase CD4+ cell counts in patients with relatively high baseline CD4+ values2–4. Nevertheless, there is little data on the usefulness of rIL-2 in patients with advanced immune deficiency5–8. Furthermore, the impact on repopulation of various T-cell subsets, the long-term outcome of these patients, and the mechanism of rIL-2-induced immune restoration have not been completely elucidated.

This study assesses the efficacy and safety of intermittent subcutaneous rIL-2 administration in a group of patients with chronic HIV-1 infection on stable HAART and CD4+ T-cell counts persistently less than 200/mm3 despite undetectable plasma HIV-RNA.

Materials and MethodsStudy design

Nineteen Caucasian patients were enrolled in an open, prospective study from January 2001 to June 2003. Eligibility criteria included age 18 to 60 years, plasma HIV-RNA < 50 copies/mL, stable HAART, CD4+ < 200 cells/ mm3 with no significant increase in the last 12 months, and no AIDS-defining illnesses or steroid administration for at least one year. Patients were scheduled to receive 6 cycles of subcutaneous rIL-2 (Macrolin®, Chiron, France), consisting of 4.5 x 106 IU daily for 5 consecutive days, every 4 weeks. Patients receiving at least one dose of rIL-2 were included in the safety analysis, based on changes in plasma HIV-RNA and laboratory parameters as well as clinical safety and tolerability. Blood samples for measuring plasma HIV-RNA, lymphocyte subset analysis, and T-cell production of intracellular cytokines were obtained on the first day of each cycle prior to dose administration, and one month after the last cycle. After completion of therapy, blood samples were obtained every three months. Efficacy was evaluated in patients receiving at least three cycles. The primary study endpoint was the median change in CD4+ lymphocyte counts at the end of treatment from baseline. Secondary endpoints were the percentage of patients who attained more than 200 CD4+ T-cells/mm3, changes in the percentage of lymphocyte phenotype subsets, assessment of intracellular IL-2, IFN-γ, IL-4 and TNF-α production, and the tolerance and safety of rIL-2 therapy.

Laboratory testing

Lymphocyte subpopulations were analyzed in lysed fresh whole blood samples by flow cytometry (FACScalibur; Becton Dickinson, San Jose, CA), and Cell-Quest software for acquisition and analysis. Intracellular production of IFN-γ, TNF-α, IL-4 and IL-2 was detected by flow cytometry after stimulating whole blood samples with PMA and ionomycin during 4 h in the presence of monensin, plus further lysis, permeabilization and cell staining with specific monoclonal antibodies. Non-stimulated whole blood was used as a negative control.

Statistical analyses

The data set was closed on May 31, 2005. A paired Wilcoxon signed-rank test was used to assess whether changes over time were different from zero. Correlations were studied with the non-parametric Spearman correlation coefficient. A two-sided P-value less than 0.05 was considered statistically significant. Multivariate analysis was not performed because of the small number of patients. Data analysis was performed with SPSS, v. 12.0 (SPSS Inc.; Chicago, IL). All patients provided written informed consent for participation.

Results

Demographic and clinical characteristics of the patients are summarized in table 1.

Table 1.

Baseline characteristics of 19 HlV-infected patients with poor immune-recovery despite persistent viral suppression under HAART

Characteristic  Values 
Sex M/F  15/4 
Median age, years (range)  43 (27-58) 
Route of HIV infection (%)   
Intravenous drug use  7 (37) 
Sexual intercourse  10 (53) 
Transfusion  2 (10) 
Previous AIDS diagnosis (%)  12 (64) 
Median CD4 T-cell count/mm3 (range)   
Nadir  36 (3-90) 
At study enrollment  99 (36-195) 
Median time on HAART, months (range)  56.5 (12-69) 
Antiretroviral treatment (%)   
2 NRTI + 1NNRTI  11 (58) 
2 NRTI + 1 PI  5 (26) 
3 NRTI  3 (16) 
HCV/HIV co-infection, n (%)  8 (42) 

M: male; F: female; HAART: highly active antiretroviral therapy; HCV: hepatitis C virus; NRTI: nucleoside reverse-transcriptase inhibitor; NNRTI: non-nucleoside reverse-transcriptase inhibitor; PI: protease inhibitor.

Changes in CD4+ and CD8+ T lymphocytes

Overall, rIL-2 led to an increase in CD4+ T-cell counts from a median of 99 cells/mm3 (range, 36-195 cells/mm3) to 147 cells/mm3 (range, 24-285 cells/mm3) after the last dose of treatment (P = 0.002), and 180 cells/mm3 (range, 38-280 cells/mm3) 18 months after the last dose (P < 0.001) (table 2). The CD4+ T-cell percentage increased from 10% (range, 4-19) to 13% (range, 3-21) after the third cycle (P = 0.001) and remained stable afterwards (table 2). However, an individual-based analysis of CD4+ T-cell count change over time showed marked heterogeneity. Among 18 patients receiving at least 3 cycles of rIL-2, 5 (27.7%) experienced an increase in CD4+ cell counts lower than 25%, and 3 (17%) (P = 0.50) and 8 (44%) (P = 0.016) patients achieved > 200 CD4+ lymphocytes/mm3 at the end of treatment and at month 18 post-treatment, respectively. Pre-treatment CD4+ T-cell count differed significantly between patients who achieved > 200 CD4+ lymphocytes/mm3 at the end of treatment (166 ± 46 cells/mm3) and those who did not achieve this threshold (88 ± 26 cells/ mm3; P = 0.001), whereas age, sex, HCV/HIV co-infection and a previous AIDS-defining event did not correlate with CD4+ T-cell count at treatment end. CD8+ T-cell percentage and absolute count remained stable during the study (table 2).

Table 2.

Changes in lymphocyte subsets over the study period

  Baseline Median (range)  Cycle 3 Median (range)  Cycle 6 Median (range)  Follow-up* Median (range)  P 
CD4+ T cells
Cells/mm3  18  99 (36-195)  18  139 (39-225)  0.001  18  147 (24-285)  0.002  18  180 (38-280)  < 0.001 
Percentage  18  10 (4-19)  18  13 (3-21)  0.007  18  13 (2-21)  0.004  18  13 (3-21)  0.003 
CD45RA+ (%)  17  6 (0-26)  14  9(0-16)  0.175  12  8 (1-23)  0.636  11  13 (0-32)  0.192 
CD45R0+ (%)  17  68 (27-91)  14  71 (39-90)  0.552  12  57 (19-87)  0.158  11  69 (33-82)  0.185 
HLA-DR+ (%)  17  25 (8-65)  14  24 (7-65)  0.289  12  17 (8-37)  0.037  11  17 (7-53)  0.005 
CD62+ (%)  17  72 (43-93)  14  71 (51-87)  0.682  12  73 (53-84)  0.783  11  77 (51-82)  0.790 
CD25+ (%)  17  26 (10-46)  14  33 (15-61)  0.005  12  32 (14-56)  0.008  11  28 (13-47)  0.166 
CD28+ (%)  17  98 (77-100)  14  97 (78-100)  0.246  12  98 (86-100)  0.150  11  96 (50-100)  0.889 
CD95+ (%)  17  92 (80-100)  14  80 (54-100)  0.045  12  86 (25-99)  0.270  11  81 (68-98)  0.075 
CD8+ T cells
Cells/mm3  18  413 (135-1700)  18  548(165-1825)  0.117  18  553 (165-1775)  0.133  18  560 (177-1400)  0.064 
Percentage  18  52 (15-72)  18  56 (15-73)  0.585  18  52 (15-71)  0.648  18  52 (16-71)  0.959 
CD45RA+ (%)  17  17 (0-54)  14  19 (7-43)  0.964  12  14 (1-41)  0.283  11  19 (3-52)  0.074 
CD45RO+ (%)  17  39 (14-77)  14  39 (3-69)  0.826  12  36 (17-60)  0.053  11  45 (11-81)  0.893 
HLA-DR+ (%)  17  42 (21-73)  14  38 (17-70)  0.084  12  30 (10-72)  0.003  11  34 (9-74)  0.008 
CD38+ (%)**  17  68 (36-99)  14  52 (22-90)  0.011  12  46 (24-84)  0.020  11  46 (14-87)  0.013 
CD62+ (%)  17  49 (15-87)  14  53 (13-63)  0.258  12  52 (30-79)  0.081  11  48 (11-88)  0.612 
CD25+ (%)  17  4 (1-27)  14  5(1-18)  0.152  12  7 (1-31)  0.099  11  3 (0-25)  0.237 
CD28+ (%)  17  51 (28-84)  14  58 (22-87)  0.040  12  60 (28-88)  0.005  11  60 (32-86)  0.062 
CD95+ (%)  17  83 (70-95)  13  70 (28-99)  0.099  12  80 (46-99)  0.666  11  84 (44-99)  0.075 

N denotes the number of patients tested. P = Wilcoxon signed-rank test value for comparison with baseline values. Significant P values are highlighted.

*

Eighteen months following the last dose.

**

Percentage of CD8+ cells with moderate to intense CD38 expression.

Changes in T lymphocyte subpopulations

No statistically significant changes were observed in the percentage of naïve and memory CD4+ T-cells over time (table 2). As T-cell counts increased during treatment, a significant increase in the absolute counts of both naive and memory CD4+ T-cells was observed at the end of treatment (data not shown). Notably, rIL-2 treatment led to a significant decrease in cell surface expression of HLA-DR and CD95 on CD4+ T-cells, and HLA-DR and CD38 on CD8+ T-cells (table 2). Furthermore, there was an increase in the expression of the alpha-chain of IL-2 receptor (CD25) on CD4+ T lymphocytes, and the effector cell marker (CD28) on CD8+ T lymphocytes (table 2). A trend toward a correlation was found between the HLA-DR expression rate on CD4+ T-cells and CD4+ T-cell counts at baseline (r = -0.30; P = 0.12), and between the decrease in HLA-DR expression on CD4+ T-cells and the increase in CD4+ T lymphocyte counts observed one month after the last dose, as compared to baseline (r = 0.345; P = 0.09).

Assessment of cytokine production

The percentage of IL-2-producing CD4+ T-cells increased from a median of 9.8% (range, 3.0-31) at baseline to 22.8% (range, 3.2-32.7) after the third cycle (P = 0.003) and 27% (range, 15.5-61.4) at the end of treatment (P = 0.005). Similarly, the percentage of IL-2-producing CD8+ T-cells significantly increased at the end of treatment as compared to baseline (P = 0.003). Intracellular expression of IFN-γ, TNF-α and IL-4 in both CD4+ and CD8+ T-cells remained unchanged during the study.

Safety

Mild constitutional symptoms and local erythema at the injection site occurred in all patients. Three patients discontinued treatment because of intolerance, one of them after the first dose, and two others after the third and the fifth cycle, respectively. Only one patient suffered severe side effects. In brief, after the third dose of the fifth cycle he was admitted to another hospital because of high fever (41 °C), chest tightness and erythroderma. Oxygen and 6-methylprednisolone were administered, rIL-2 was definitively stopped, and he experienced a rapid clinical improvement. A transitory increase in HIV-RNA was seen in 50% of patients (9/18) during the treatment period.

Discussion

Overall, subcutaneous rIL-2 significantly increased both absolute count and percentage of the CD4+ lymphocyte pool. Our results confirm that rIL-2 might improve the immunological status of patients who start HAART with advanced HIV infection and persist with < 200 CD4+ lymphocytes/mm3, despite viral suppression to undetectable levels5–8. However, in agreement with previous reports9, individual response to rIL-2 varied widely, and almost 30% (5/18) of patients receiving at least 3 cycles of rIL-2 had a minimal or null response (CD4+ T-cell increase < 25%). The lower percentage of patients who achieved 200 CD4+ T-cells/mm3 in our study in comparison with previous reports5–8 may be related to a more advanced disease status in our population, as reflected by a lower CD4+ T-cell count at baseline (99 cells/mm3) and the high rate of patients with a previous AIDS-defining clinical event (64%), or to the lower daily dose of rIL-2 administered in our study. We tried this dose, which provides the same accumulated monthly amount of rIL-2 as the standard dose, because lower daily doses of rIL-2 have been proven effective, are better tolerated, and allow for a better quality of life5. In keeping with previous studies, the treatment was in general well tolerated, yet one patient suffered a life-threatening event.

Poor immune reconstitution in HIV-infected patients on HAART has been related to low nadir CD4+ T-cell counts10,11 and markers of limited thymopoiesis, including older age, decreased thymic mass and smaller number of peripheral nai've CD4+ T-cells12,13. Furthermore, persistent immune activation and spontaneous apoptosis were found to be a consistent feature in patients with a discordant immune response, supporting a role for immune activation in the pathogenesis of CD4+ T-cell depletion and immune reconstitution following HAART14–16.

Several hypotheses such as cellular proliferation17, prevention of apoptosis18, and neo-thymic production7 have been proposed to explain rIL-2-driven CD4 T-cell expansion, but the mechanisms are still not completely understood. Recently reported results suggest that intermittent rIL-2 leads to expansion of the CD4+ T-cell pool by downregulation of immune activation and T-cell proliferation rates19. Additionally, in vivo labeling studies of the CD4+ T-cell pool before and after rIL-2 therapy identified the emergence of a long-lived CD4+ T-cell subpopulation (CD4+ CD25+) on both naïve (CD45RA + CD27+) and recall memory phenotypes (CD45RO + CD27+)20. In vitro study has shown that IL-2 production and CD25 expression shape a population of regulatory T-cells (CD4+ CD25+ CD62+) that accounts for 1% to 2% of the entire CD4+ T-cell pool and plays an essential role in peripheral CD4+ T-cell homeostasis21. Loss of these regulatory cells has been associated with higher levels of immune activation and decreases in the number of peripheral CD4+ T-cells22. In keeping with these results, we observed a sustained increase of CD25 expression on CD4+ T-cells (although we did not further characterize them as regulatory cells) and a significant decrease in the expression of immune activation markers on both CD4+ and CD8+ T-cells. In addition, there was an increase in co-stimulatory molecule CD28 expression on CD8+ lymphocytes, which also suggests a decreased immune activation state. It has been shown that CD8+ T-cells lacking CD28 surface expression represent a subset of short-lived activated and differentiated cytotoxic effector cells, driven by viral replication23. Indeed, the percentage of both CD4+ and CD8+ T-cells expressing intracellular IL-2 did increase significantly.

Interestingly, immunological recovery continued in our study after rIL-2 was discontinued. This long-lasting effect has been observed in patients with advanced HIV infection5,7 and can be explained by the persistence of rIL-2-driven homeostatic changes.

The main limitation of our study is the lack of a control group. The immunological improvement could be explained alternatively by persistent HAART-induced viral suppression24, which could be related to a slow but ongoing decay in viremia over time. Another potential limitation is the small size of the population studied. Assessment of T lymphocyte subpopulation changes over time was only performed in a subset of patients. This fact could explain the lack of significant changes in naïve and memory CD4+ T-cell percentages, as well as the lack of a significant correlation between the CD4+ T-cell count increase and decreased expression of immune activation markers. Finally, although the use of rIL-2 makes particular sense in patients with advanced infection such as those included in our study, its clinical benefit in this subpopulation has yet to be definitively proven.

In summary, although high interindividual variability was observed, our data suggest that rIL-2 therapy might expand the CD4+ T-cell pool in some patients with advanced disease and a blunted immune response to HAART. These rIL-2-driven changes could be mediated by down-regulation of immune activation status.

The results of the ongoing SILCAAT trial will determine whether the changes in the number and function of rIL-2-induced CD4+ T-cells translate into clinical benefit.

Acknowledgments

The authors thank Celine Cavallo for English language editing.

References
[1.]
H.M. Lederman, P.L. Williams, J.W. Wu, T.G. Evans, S.E. Cohn, J.A. McCutchan, et al.
Incomplete immune reconstitution after initiation of highly active antiretroviral therapy in human immunodeficiency virus-infected patients with severe CD4 + cell depletion.
J Infect Dis, 188 (2003), pp. 1794-1803
[2.]
Y. Levy, C. Durier, R. Krzysiek, C. Rabian, C. Capitant, A.S. Lascaux, et al.
Effects of interleukin-2 therapy combined with highly active antiretroviral therapy on immune restoration in HIV-1 infection: a randomized controlled trial.
[3.]
M.H. Losso, W.H. Belloso, S. Emery, J.A. Benetucci, P.E. Cahn, M.C. Lasala, et al.
A randomized, controlled, phase II trial comparing escalating doses of subcutaneous interleukin-2 plus antiretrovirals versus antiretrovirals alone in human immunodeficiency virus-infected patients with CD4 + cell counts ≥ 350/mm3.
J Infect Dis, 181 (2000), pp. 1614-1621
[4.]
R.T. Davey Jr, R.L. Murphy, F.M. Graziano, S.L. Boswell, A.T. Pavia, M. Cancio, et al.
Immunologic and virologic effects of subcutaneous interleukin 2 in combination with antiretroviral therapy: A randomized controlled trial.
JAMA, 284 (2000), pp. 183-189
[5.]
A. Arnó, L. Ruiz, M. Juan, A. Jon, M. Balagué, M.K. Zayat, et al.
Efficacy of low-dose subcutaneous interleukin-2 to treat advanced human immunodeficiency virus type 1 in persons with ≤ 250/μL CD4 T cells and undetectable plasma virus load.
J Infect Dis, 180 (1999), pp. 56-60
[6.]
D. David, L. Nait-Ighil, B. Dupont, J. Maral, B. Gachot, J. Theze.
Rapid effect of interleukin-2 therapy in human immunodeficiency virus-infected patients whose CD4 cell counts increase only slightly in response to combined antiretroviral treatment.
J Infect Dis, 183 (2001), pp. 730-735
[7.]
G. Marchetti, L. Meroni, S. Varchetta, V. Terzieva, A. Bandera, D. Manganaro, et al.
Low-dose prolonged intermittent interleukin-2 adjuvant therapy: results of a randomized trial among human immunodeficiency virus-positive patients with advanced immune impairment.
J Infect Dis, 186 (2002), pp. 606-616
[8.]
C. Katlama, G. Carcelain, C. Duvivier, C. Chouguet, R. Tubiana, M. De Sa, et al.
Interleukin-2 accelerates CD4 cell reconstitution in HIV-infected patients with severe immunosuppression despite highly active antiretroviral therapy: the ILSTIM study - ANRS 082.
AIDS, 16 (2002), pp. 2027-2034
[9.]
C.E. Farel, D.G. Chaitt, B.K. Hahn, J.A. Tavel, J.A. Kovacs, M.A. Polis, et al.
Induction and maintenance therapy with intermittent interleukin-2 in HIV-1 infection.
Blood, 103 (2004), pp. 3282-3286
[10.]
M. Renaud, C. Katlama, A. Mallet, V. Calvez, G. Carcelain, R. Tubiana, et al.
Determinants of paradoxical CD4 cell reconstitution after protease inhibitor-containing antiretroviral regimen.
AIDS, 13 (1999), pp. 669-676
[11.]
F. García, L.E. de Lazzari, M. Plana, P. Castro, G. Mestre, M. Nomdedeu, et al.
Long-term CD4 + T-cell response to highly active antiretroviral therapy according to baseline CD4 + T-cell count.
J Acquir Immune Defic Syndr, 36 (2004), pp. 702-713
[12.]
D.W. Notermans, N.G. Pakker, D. Hamann, N.A. Fondraine, R.H. Kauffmann, P.L. Meenhorst, et al.
Immune reconstitution after 2 years of successful potent antiretroviral therapy in previously untreated human immunodeficiency virus type 1-infected adults.
J Infect Dis, 180 (1999), pp. 1050-1056
[13.]
R.C. Kalayjian, J. Spritzler, M. Pu, A. Landay, R.B. Pollard, V. Stocker, et al.
Distinct mechanisms of T cell reconstitution can be identified by estimating thymic volume in adult HIV-1 disease.
J Infect Dis, 192 (2005), pp. 1577-1587
[14.]
O. Benveniste, A. Flahault, F. Rollot, C. Elbim, J. Estaquier, B. Pédron, et al.
Mechanisms involved in the low-level regeneration of CD4 + cells in HIV-1-infected patients receiving highly active antiretroviral therapy who have prolonged undetectable plasma viral loads.
J Infect Dis, 191 (2005), pp. 1670-1679
[15.]
N. Hansjee, G.R. Kaufmann, C. Strub, R. Weber, M. Battegay, P. Erb.
Persistent apoptosis in HIV-1-infected individuals receiving potent antiretroviral therapy is associated with poor recovery of CD4 T lymphocytes.
J Acquir Immune Defic Syndr, 36 (2004), pp. 671-677
[16.]
K.B. Anthony, C. Yoder, J.A. Metcalf, R. Der Simonian, J.M. Orenstein, R.A. Stevens, 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, 33 (2003), pp. 125-133
[17.]
U.R. Hengge, C. Borchard, S. Esser, M. Schroder, A. Mirmohammadsadegh, M. Goos.
Lymphocytes proliferate in blood and lymph nodes following inter-leukin-2 therapy in addition to highly active antiretroviral therapy.
AIDS, 16 (2002), pp. 151-160
[18.]
F. Pandolfi, M. Pierdominici, M. Marziali, M. Liria Bernardi, G. Antonelli, V. Galati, et al.
Low-dose IL-2 reduces lymphocyte apoptosis and increases naive CD4 cells in HIV-1 patients treated with HAART.
Clin Immunol, 94 (2000), pp. 153-159
[19.]
I. Sereti, K.B. Anthony, H. Martinez-Wilson, R. Lempicki, J. Adelsberger, J.A. Metcalf, et al.
IL-2-induced CD4 + T-cell expansion in HIV-infected patients is associated with long-term decreases in T-cell proliferation.
Blood, 104 (2004), pp. 775-780
[20.]
P.P. De Paoli, S. Zanussi, C. Simonelli, M.T. Bortolin, M. D’Andrea, C. Crepaldi, et al.
Effects of subcutaneous interleukin-2 therapy on CD4 subsets and in vitro cytokine production in HIV + subjects.
J Clin Invest, 100 (1997), pp. 2737-2743
[21.]
A.R. Almeida, N. Legrand, M. Papiernik, A.A. Freitas.
Homeostasis of peripheral CD4 + T cells: IL-2R alpha and IL-2 shape a population of regulatory cells that controls CD4 + T cell numbers.
J Immunol, 169 (2002), pp. 4850-4860
[22.]
M.P. Eggena, B. Barugahare, N. Jones, M. Okello, S. Mutalya, C. Kityo, et al.
Depletion of regulatory T cells in HIV infection is associated with immune activation.
J Immunol, 174 (2005), pp. 4407-4414
[23.]
N.J. Borthwick, M. Bofill, W.M. Gombert, A.N. Akbar, E. Medina, K. Sagawa, et al.
Lymphocyte activation in HIV-1 infection. II. Functional defects of CD28-T cells.
AIDS, 8 (1994), pp. 431-441
[24.]
S.L. Koletar, P.L. Williams, J. Wu, J.A. McCutchan, S.E. Cohn, R.L. Murphy, et al.
Long-term follow-up of HIV-infected individuals who have significant increases in CD4 + cell counts during antiretroviral therapy.
Clin Infect Dis, 39 (2004), pp. 1500-1506
Copyright © 2008. Elsevier España, S.L.. All rights reserved
Opciones de artículo
es en pt

¿Es usted profesional sanitario apto para prescribir o dispensar medicamentos?

Are you a health professional able to prescribe or dispense drugs?

Você é um profissional de saúde habilitado a prescrever ou dispensar medicamentos