Impact of Early Versus Late Antiretroviral Treatment Initiation on Naive T Lymphocytes in HIV-1-Infected Children and Adolescents - The-ANRS-EP59-CLEAC Study

Pierre Frange, Thomas Montange, Jérôme Le Chenadec, Damien Batalie, Ingrid Fert, Catherine Dollfus, Albert Faye, Stéphane Blanche, Anne Chacé, Corine Fourcade, Isabelle Hau, Martine Levine, Nizar Mahlaoui, Valérie Marcou, Marie-Dominique Tabone, Florence Veber, Alexandre Hoctin, Thierry Wack, Véronique Avettand-Fenoël, Josiane Warszawski, Florence Buseyne, Pierre Frange, Thomas Montange, Jérôme Le Chenadec, Damien Batalie, Ingrid Fert, Catherine Dollfus, Albert Faye, Stéphane Blanche, Anne Chacé, Corine Fourcade, Isabelle Hau, Martine Levine, Nizar Mahlaoui, Valérie Marcou, Marie-Dominique Tabone, Florence Veber, Alexandre Hoctin, Thierry Wack, Véronique Avettand-Fenoël, Josiane Warszawski, Florence Buseyne

Abstract

Background: The early initiation of antiretroviral therapy (ART) in HIV-1-infected infants reduces mortality and prevents early CD4 T-cell loss. However, the impact of early ART on the immune system has not been thoroughly investigated in children over five years of age or adolescents. Here, we describe the levels of naive CD4 and CD8 T lymphocytes (CD4/CD8TN), reflecting the quality of immune reconstitution, as a function of the timing of ART initiation (early (<6 months) versus late (≥24 months of age)).

Methods: The ANRS-EP59-CLEAC study enrolled 27 children (5-12 years of age) and nine adolescents (13-17 years of age) in the early-treatment group, and 19 children (L-Ch) and 21 adolescents (L-Ado) in the late-treatment group. T lymphocytes were analyzed by flow cytometry and plasma markers were analyzed by ELISA. Linear regression analysis was performed with univariate and multivariate models.

Results: At the time of evaluation, all patients were on ART and had a good immunovirological status: 83% had HIV RNA loads below 50 copies/mL and the median CD4 T-cell count was 856 cells/µL (interquartile range: 685-1236 cells/µL). In children, early ART was associated with higher CD8TN percentages (medians: 48.7% vs. 31.0%, P = 0.001), and a marginally higher CD4TN (61.2% vs. 53.1%, P = 0.33). In adolescents, early ART was associated with low CD4TN percentages and less differentiated memory CD8 T cells. CD4TN and CD8TN levels were inversely related to cellular activation and gut permeability.

Conclusion: In children and adolescents, the benefits of early ART for CD8TN were clear after long-term ART. The impact of early ART on CD4TN appears to be modest, because pediatric patients treated late respond to HIV-driven CD4 T-lymphocyte loss by the de novo production of TN cells in the thymus. Our data also suggest that current immune activation and/or gut permeability has a negative impact on TN levels.

Clinical trial registration: ClinicalTrials.gov, identifier NCT02674867.

Keywords: HIV-1; T lymphocyte; adolescents; children; early ART; naive T lymphocyte.

Conflict of interest statement

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Copyright © 2021 Frange, Montange, Le Chenadec, Batalie, Fert, Dollfus, Faye, Blanche, Chacé, Fourcade, Hau, Levine, Mahlaoui, Marcou, Tabone, Veber, Hoctin, Wack, Avettand-Fenoël, Warszawski and Buseyne.

Figures

Figure 1
Figure 1
CD4 T-cell subsets in patients treated early and late. T-cell phenotypes were assessed by flow cytometry on fresh whole blood (Supplementary Material). The percentage of each subset among total CD4 T lymphocytes is presented for children (A) and adolescents (B), on box and whiskers plots showing the minimum and maximum values. The early and late treatment groups were compared in Mann-Whitney tests; when significant, P values are shown on the graph. Subsets are presented from the least to the most differentiated, and were defined as naive TN (CD45RA+CCR7+); recent thymic emigrants, TRTE (CD45RA+CCR7+CD31+); CD31negTN (CD45RA+CCR7+CD31-); central memory, TCM (CD45RA-CCR7+); transitional memory TTM (CD45RA-CCR7-CD27+); effector memory TEM (CD45RA-CCR7-CD27-) and effector TEF (CD45RA+CCR7-CD27-CD28-) cells. (C) The correlograms present Spearman’s rank correlation coefficients as symbols (upper quadrants) and values. Significant associations are indicated by symbols (*p < .05; **p < .01; ***p < .001). CD4 T-cell counts were used for correlation analyses.
Figure 2
Figure 2
Linear regression analysis of the associations between CD4TN and demographic, virological and immunological factors in children and adolescents. Results from univariate (A, B) and multivariate (C, D) linear regressions are presented as estimates (β) and 95% confidence intervals. Significant associations are indicated by symbols (*p < .05; **p < .01; ***p < .001). Multivariate analysis included the covariables indicated on the plot. A and C: children; B and D: adolescents. Estimates are given per year, 100 CD4 T cells, and 1% activated CD4TM.
Figure 3
Figure 3
CD8 T-cell differentiation subsets in patients with early and late treatment initiation. T-cell phenotypes were assessed by flow cytometry on fresh whole blood (Supplementary Material). The percentage of each subset among total CD8 T lymphocytes is presented for children (A) and adolescents (B) on box and whiskers plots showing the minimum and maximum values. Early and late treatment groups were compared in Mann-Whitney tests; when significant, P values are shown on the graph. Subsets are presented from the least to the most differentiated, and were defined as naive TN (CD45RA+CCR7+); central memory TCM (CD45RA-CCR7+); effector memory TEM27+28+ (CD45RA-CCR7-CD27+CD28+); TEM27-28+ (CD45RA-CCR7- CD27-CD28+); TEM27+28- (CD45RA-CCR7- CD27+CD28-); TEM27-28- (CD45RA-CCR7- CD27-CD28-) and effector TEF (CD45RA+CCR7-CD27-CD28-) cells. (C) The correlograms present Spearman’s rank correlation coefficients as symbols (upper quadrants) and values. Significant associations are indicated by symbols (*p < .05; **p < .01; ***p < .001). CD4 T-cell counts were used to calculate correlations.
Figure 4
Figure 4
Linear regression analysis of the associations between CD8TN and demographic, virological and immunological factors in children and adolescents. Results from univariate (A, B) and multivariate (C, D) linear regressions are presented as estimates (β) and 95% confidence intervals. Significant associations are indicated by symbols (*p < .05; **p < .001; ***p < .0001). Multivariate analysis included the covariables indicated on the plot. Estimates are given per year, per 0.1 units of normalized viral suppression, per unit of normalized cumulative viremia, per point of CD4/CD8 ratio, and per 1% of activated CD4TM.

References

    1. Dollfus C, Le Chenadec J, Faye A, Blanche S, Briand N, Rouzioux C, et al. . Long-term outcomes in adolescents perinatally infected with HIV-1 and followed up since birth in the French Perinatal Cohort (EPF/ANRS CO10). Clin Infect Dis (2010) 51:214–24. 10.1086/653674
    1. Chiappini E, Bianconi M, Dalzini A, Petrara MR, Galli L, Giaquinto C, et al. . Accelerated aging in perinatally HIV-infected children: clinical manifestations and pathogenetic mechanisms. Aging (2018) 10:3610–25. 10.18632/aging.101622
    1. Zicari S, Sessa L, Cotugno N, Ruggiero A, Morrocchi E, Concato C, et al. . Immune activation, inflammation, and non-AIDS co-morbidities in HIV-infected patients under long-term ART. Viruses (2019) 11:200. 10.3390/v11030200
    1. Cotton MF, Violari A, Otwombe K, Panchia R, Dobbels E, Rabie H, et al. . Early time-limited antiretroviral therapy versus deferred therapy in South African infants infected with HIV: results from the children with HIV early antiretroviral (CHER) randomised trial. Lancet (2013) 382:1555–63. 10.1016/S0140-6736(13)61409-9
    1. Shiau S, Abrams EJ, Arpadi SM, Kuhn L. Early antiretroviral therapy in HIV-infected infants: can it lead to HIV remission? Lancet HIV (2018) 5:e250–e58. 10.1016/s2352-3018(18)30012-2
    1. Sabin CA, Smith CJ, Monforte AD, Battegay M, Gabiano C, Galli L, et al. . Response to combination antiretroviral therapy: variation by age - The Collaboration of Observational HIV Epidemiological Research Europe (COHERE) study group. AIDS (2008) 22:1463–73. 10.1016/S0140-6736(11)60208-0
    1. Picat MQ, Lewis J, Musiime V, Prendergast A, Nathoo K, Kekitiinwa A, et al. . Predicting patterns of long-term CD4 reconstitution in HIV-infected children starting antiretroviral therapy in sub-Saharan Africa: a cohort-based modelling study. PloS Med (2013) 10:e1001542. 10.1371/journal.pmed.1001542
    1. Azzoni L, Barbour R, Papasavvas E, Glencross DK, Stevens WS, Cotton MF, et al. . Early ART results in greater immune reconstitution benefits in HIV-infected infants: Working with data missingness in a longitudinal dataset. PloS One (2015) 10:e0145320. 10.1371/journal.pone.0145320
    1. Guillen S, Prieto L, Jimenez de Ory S, Gonzalez-Tome MI, Rojo P, Navarro ML, et al. . Prognostic factors of a lower CD4/CD8 ratio in long term viral suppression HIV infected children. PloS One (2019) 14:e0220552. 10.1371/journal.pone.0220552
    1. Buseyne F, Scott-Algara D, Bellal N, Burgard M, Rouzioux C, Blanche S, et al. . The frequency of HIV-specific interferon-gamma-producing CD8 T cells is associated with both age and level of antigenic stimulation in HIV-1-infected children. J Infect Dis (2005) 192:1781–86. 10.1086/497152
    1. Scott ZA, Chadwick EG, Gibson LL, Catalina MD, McManus MM, Yogev R, et al. . Infrequent detection of HIV-1-specific, but not cytomegalovirus-specific, CD8+ T cell responses in young HIV-1-infected infants. J Immunol (2001) 167:7134–40. 10.4049/jimmunol.167.12.7134
    1. Rinaldi S, Pallikkuth S, Cameron M, de Armas LR, Cotugno N, Dinh V, et al. . Impact of early antiretroviral therapy initiation on HIV-specific CD4 and CD8 T-cell function in perinatally infected children. J Immunol (2020) 204:540–49. 10.4049/jimmunol.1900856
    1. Aupiais C, Faye A, Le Chenadec J, Rouzioux C, Bouallag N, Laurent C, et al. . Interruption of cART in clinical practice is associated with an increase in the long-term risk of subsequent immunosuppression in HIV-1-infected children. Pediatr Infect Dis J (2014) 33:1237–45. 10.1097/inf.0000000000000450
    1. Palladino C, Briz V, Bellon JM, Climent FJ, de Ory SJ, Mellado MJ, et al. . Determinants of highly active antiretroviral therapy duration in HIV-1-infected children and adolescents in Madrid, Spain, from 1996 to 2012. PloS One (2014) 9:e96307. 10.1371/journal.pone.0096307
    1. Kakkar F, Lee T, Hawkes MT, Brophy J, Lindy S, Singer J, et al. . Challenges to achieving and maintaining viral suppression among children living with HIV. AIDS (2020) 34:687–97. 10.1097/qad.0000000000002454
    1. Avettand-Fenoel V, Le Chenadec J, Diallo M, Fillion M, Mélard A, Dollfus C, et al. . Initiating antiretroviral treatment early in infancy has long-term benefits on the HIV reservoir in late childhood and adolescence. Clin Infect Dis (2021). 10.1093/cid/ciaa1931. Advanced online publication.
    1. Gaballa A, Clave E, Uhlin M, Toubert A, Arruda LCM. Evaluating thymic function after human hematopoietic stem cell transplantation in the personalized medicine era. Front Immunol (2020) 11:1341. 10.3389/fimmu.2020.01341
    1. Avettand-Fenoel V, Chaix ML, Blanche S, Burgard A, Floch C, Toure K, et al. . LTR real-time PCR for HIV-1 DNA quantitation in blood cells for early diagnosis in infants born to seropositive mothers treated in HAART Area (ANRS CO 01). J Med Virol (2009) 81:217–23. 10.1002/jmv.21390
    1. Avettand-Fenoel V, Blanche S, Le Chenadec J, Scott-Algara D, Dollfus C, Viard JP, et al. . Relationships between HIV disease history and blood HIV-1 DNA load in perinatally infected adolescents and young adults: the ANRS-EP38-IMMIP Study. J Infect Dis (2012) 205:1520–28. 10.1093/infdis/jis233
    1. Garcia-Prat M, Álvarez-Sierra D, Aguiló-Cucurull A, Salgado-Perandrés S, Briongos-Sebastian S, Franco-Jarava C, et al. . Extended immunophenotyping reference values in a healthy pediatric population. Cytom B Clin Cytom (2019) 96:223–33. 10.1002/cyto.b.21728
    1. Blanche S, Scott-Algara D, Le Chenadec J, Didier C, Montange T, Avettand-Fenoel V, et al. . Naive T lymphocytes and recent thymic emigrants are associated with HIV-1 disease history in French adolescents and young adults infected in the perinatal period: The ANRS-EP38-IMMIP study. Clin Infect Dis (2014) 58:573–87. 10.1093/cid/cit729[doi
    1. Nobile M, Correa R, Borghans JAM, D’Agostino C, Schneider P, de Boer RJ, et al. . De novo T-cell generation in patients at different ages and stages of HIV-1 disease. Blood (2004) 104:470–77. 10.1182/blood-2003-12-4265
    1. Aguilera-Sandoval CR, Yang OO, Jojic N, Lovato P, Chen DY, Boechat MI, et al. . Supranormal thymic output up to 2 decades after HIV-1 infection. AIDS (2016) 30:701–11. 10.1097/QAD.0000000000001010
    1. Appay V, Fastenackels S, Katlama C, Ait-Mohand H, Schneider L, Guihot A, et al. . Old age and anti-cytomegalovirus immunity are associated with altered T-cell reconstitution in HIV-1-infected patients. AIDS (2011) 25:1813–22. 10.1097/QAD.0b013e32834640e6
    1. Lewis J, Walker AS, Castro H, De Rossi A, Gibb DM, Giaquinto C, et al. . Age and CD4 count at initiation of antiretroviral therapy in HIV-infected children: effects on long-term T-cell reconstitution. J Infect Dis (2012) 205:548–56. 10.1093/infdis/jir787
    1. Zakhour R, Tran DQ, Degaffe G, Bell CS, Donnachie E, Zhang W, et al. . Recent thymus emigrant CD4+ T cells predict HIV disease progression in patients with perinatally acquired HIV. Clin Infect Dis (2016) 62:1029–35. 10.1093/cid/ciw030
    1. Lewis J, Payne H, Walker AS, Otwombe K, Gibb DM, Babiker AG, et al. . Thymic output and CD4 T-cell reconstitution in HIV-infected children on early and interrupted antiretroviral treatment: Evidence from the children with HIV early antiretroviral therapy trial. Front Immunol (2017) 8:1162. 10.3389/fimmu.2017.01162
    1. Goetghebuer T, Le Chenadec J, Haelterman E, Galli L, Dollfus C, Thorne C, et al. . Short- and long-term immunological and virological outcome in HIV-infected infants according to the age at antiretroviral treatment initiation. Clin Infect Dis (2012) 54:878–81. 10.1093/cid/cir950
    1. Cohen S, Smit C, van Rossum AM, Fraaij PL, Wolfs TF, Geelen SP, et al. . Long-term response to combination antiretroviral therapy in HIV-infected children in the Netherlands registered from 1996 to 2012. AIDS (2013) 27:2567–75. 10.1097/01.aids.0000432451.75980.1b
    1. Desmonde S, Dicko F, Koueta F, Eboua T, Balestre E, Amani-Bosse C, et al. . Association between age at antiretroviral therapy initiation and 24-month immune response in West-African HIV-infected children. AIDS (2014) 28:1645–55. 10.1097/QAD.0000000000000272
    1. Novelli S, Lecuroux C, Avettand-Fenoel V, Seng R, Essat A, Morlat P, et al. . Long-term therapeutic impact of the timing of antiretroviral therapy in patients diagnosed with primary human immunodeficiency virus type 1 infection. Clin Infect Dis (2018) 66:1519–27. 10.1093/cid/cix1068
    1. Ferrando-Martinez S, De Pablo-Bernal RS, De Luna-Romero M, De Ory SJ, Genebat M, Pacheco YM, et al. . Thymic function failure is associated with human immunodeficiency virus disease progression. Clin Infect Dis (2017) 64:1191–97. 10.1093/cid/cix095
    1. Paghera S, Quiros-Roldan E, Sottini A, Properzi M, Castelli F, Imberti L. Lymphocyte homeostasis is maintained in perinatally HIV-infected patients after three decades of life. Immun Ageing (2019) 16:26. 10.1186/s12979-019-0166-7
    1. Turner JE, Campbell JP, Edwards KM, Howarth LJ, Pawelec G, Aldred S, et al. . Rudimentary signs of immunosenescence in Cytomegalovirus-seropositive healthy young adults. Age (2014) 36:287–97. 10.1007/s11357-013-9557-4
    1. van den Heuvel D, Jansen MAE, Nasserinejad K, Dik WA, van Lochem EG, Bakker-Jonges LE, et al. . Effects of nongenetic factors on immune cell dynamics in early childhood: The Generation R Study. J Allergy Clin Immunol (2017) 139:1923–34.e17. 10.1016/j.jaci.2016.10.023
    1. Bourgeois C, Hao Z, Rajewsky K, Potocnik AJ, Stockinger B. Ablation of thymic export causes accelerated decay of naive CD4 T cells in the periphery because of activation by environmental antigen. PNAS (2008) 105:8691–96. 10.1073/pnas.0803732105
    1. De Voeght A, Martens H, Renard C, Vaira D, Debruche M, Simonet J, et al. . Exploring the link between innate immune activation and thymic function by measuring sCD14 and TRECs in HIV patients living in Belgium. PloS One (2017) 12:e0185761. 10.1371/journal.pone.0185761
    1. Koay WLA, Lindsey JC, Uprety P, Bwakura-Dangarembizi M, Weinberg A, Levin MJ, et al. . Intestinal integrity biomarkers in early antiretroviral-treated perinatally HIV-1-infected infants. J Infect Dis (2018) 218:1085–89. 10.1093/infdis/jiy271
    1. Prendergast AJ, Chasekwa B, Rukobo S, Govha M, Mutasa K, Ntozini R, et al. . Intestinal damage and inflammatory biomarkers in human immunodeficiency virus (HIV)–exposed and HIV-infected Zimbabwean infants. J Inf Dis (2017) 216:651–61. 10.1093/infdis/jix367
    1. Dirajlal-Fargo S, El-Kamari V, Weiner L, Shan L, Sattar A, Kulkarni M, et al. . Altered intestinal permeability and fungal translocation in Ugandan children with human immunodeficiency virus. Clin Infect Dis (2020) 70:2413–22. 10.1093/cid/ciz561
    1. Sandler NG, Wand H, Roque A, Law M, Nason MC, Nixon DE, et al. . Plasma levels of soluble CD14 independently predict mortality in HIV infection. J Infect Dis (2011) 203:780–90. 10.1093/infdis/jiq118
    1. Fastenackels S, Sauce D, Vigouroux C, Avettand-Fenoel V, Bastard J-P, Fellahi S, et al. . HIV-mediated immune aging in young adults infected perinatally or during childhood. AIDS (2019) 33:1705–10. 10.1097/QAD.0000000000002275

Source: PubMed

3
S'abonner