Post-treatment HIV-1 controllers with a long-term virological remission after the interruption of early initiated antiretroviral therapy ANRS VISCONTI Study

Asier Sáez-Cirión, Charline Bacchus, Laurent Hocqueloux, Véronique Avettand-Fenoel, Isabelle Girault, Camille Lecuroux, Valerie Potard, Pierre Versmisse, Adeline Melard, Thierry Prazuck, Benjamin Descours, Julien Guergnon, Jean-Paul Viard, Faroudy Boufassa, Olivier Lambotte, Cécile Goujard, Laurence Meyer, Dominique Costagliola, Alain Venet, Gianfranco Pancino, Brigitte Autran, Christine Rouzioux, ANRS VISCONTI Study Group, Asier Sáez-Cirión, Charline Bacchus, Laurent Hocqueloux, Véronique Avettand-Fenoel, Isabelle Girault, Camille Lecuroux, Valerie Potard, Pierre Versmisse, Adeline Melard, Thierry Prazuck, Benjamin Descours, Julien Guergnon, Jean-Paul Viard, Faroudy Boufassa, Olivier Lambotte, Cécile Goujard, Laurence Meyer, Dominique Costagliola, Alain Venet, Gianfranco Pancino, Brigitte Autran, Christine Rouzioux, ANRS VISCONTI Study Group

Abstract

Combination antiretroviral therapy (cART) reduces HIV-associated morbidities and mortalities but cannot cure the infection. Given the difficulty of eradicating HIV-1, a functional cure for HIV-infected patients appears to be a more reachable short-term goal. We identified 14 HIV patients (post-treatment controllers [PTCs]) whose viremia remained controlled for several years after the interruption of prolonged cART initiated during the primary infection. Most PTCs lacked the protective HLA B alleles that are overrepresented in spontaneous HIV controllers (HICs); instead, they carried risk-associated HLA alleles that were largely absent among the HICs. Accordingly, the PTCs had poorer CD8+ T cell responses and more severe primary infections than the HICs did. Moreover, the incidence of viral control after the interruption of early antiretroviral therapy was higher among the PTCs than has been reported for spontaneous control. Off therapy, the PTCs were able to maintain and, in some cases, further reduce an extremely low viral reservoir. We found that long-lived HIV-infected CD4+ T cells contributed poorly to the total resting HIV reservoir in the PTCs because of a low rate of infection of naïve T cells and a skewed distribution of resting memory CD4+ T cell subsets. Our results show that early and prolonged cART may allow some individuals with a rather unfavorable background to achieve long-term infection control and may have important implications in the search for a functional HIV cure.

Conflict of interest statement

The authors have declared that no competing interests exist.

Figures

Figure 1. Long-term control of viremia and…
Figure 1. Long-term control of viremia and stable CD4+ T cell counts in fourteen patients after interruption of antiretroviral treatment initiated in primary HIV-1 infection.
CD4+ T cell counts (in black) and plasma HIV-1 RNA viral loads (in blue) during the follow-up after PHI diagnosis in the 14 PTCs included in the study. The detectable viral loads after treatment interruption are indicated in red. The gray areas represent the periods during which the patients received cART.
Figure 2. Patients to become post-treatment controllers…
Figure 2. Patients to become post-treatment controllers have higher viral loads and lower CD4+ T cell counts than HIV controllers during primary HIV infection.
CD4+ T cell counts (A) and plasma viral load (B) during the primary infection for the patients enrolled in the ANRS PRIMO cohort who later exhibited spontaneous control of infection (preHIC; n = 8) , for the PTCs included in our study (n = 14) and for the patients in the ANRS PRIMO cohort who did not control infection (n = 1,245). The median and the 10th and 90th percentiles are shown for each group.
Figure 3. Post-treatment controllers differ from HIV…
Figure 3. Post-treatment controllers differ from HIV controllers in terms of HLA class I profile, frequency and quality of the CD8+ T cell response and activation levels of CD8+ T cells.
A. The frequencies of the protective alleles HLA-B*27 and B*57 and the risk alleles HLA-B*07 and B*35 in the general French population (n = 6094 alleles , www.allelefrequencies.net), HICs (n = 148 alleles) and PTCs (n = 28 alleles). The statistical analyses are shown in Table S2. B. The frequency of HIV-specific CD8+ T cells, estimated as the number of CD8+ T cells producing IFN-γ upon stimulation with optimal HIV-1 peptides (spot-forming cells, SFC) in untreated viremic patients (VIRs) (n = 57), treated patients (ARTs) (n = 60), HICs (n = 100) and PTCs (n = 12). C. The capacity of CD8+ T cells from VIRs (n = 22), ARTs (n = 14), HICs (n = 73) and PTCs (n = 14) to suppress the HIV-1 infection of autologous CD4+ T cells, as determined by the log-fold decrease in the level of secreted p24 (CD4 vs. CD4∶CD8 1∶1 cell cultures). D. The percentage of CD8+ T cells from ARTs (n = 5), HICs (n = 58) and PTCs (n = 8) that expressed CD38, HLA-DR or both CD38 and HLA-DR ex vivo. B, C and D. The mean and standard deviation for each group are shown.
Figure 4. Post-treatment controllers have very low…
Figure 4. Post-treatment controllers have very low levels of cell-associated HIV DNA which keep decreasing after treatment interruption for some patients.
A. Levels of cell-associated HIV-1 DNA (median and IQR) in 6 PTCs at PHI, just before or at treatment interruption (TI), and the last available value obtained at a median of 6 years after cART discontinuation (Last). B. The evolution of cell-associated HIV DNA after treatment interruption in PBMCs from 8 PTCs. The slope of the evolution of HIV-DNA levels after treatment interruption was calculated by linear regression (lines) of the available sequential measures (symbols). Five PTCs experienced a decline of their cell-associated HIV-DNA levels (left); two PTCs maintained stable levels and a positive slope was calculated for OR3 (right). C. Infection levels in various cell populations from 11 PTCs: PBMCs, CD4+ T cells and monocytes; activated and resting CD4+ T cells; resting naïve (TN), central memory (TCM), transitional memory (TTM) and effector memory (TEM) CD4+ T cell subsets (see Figure S2 for the sorting strategy). The open symbols represent values below the threshold of detection. The medians are represented. A, B, C. The results are expressed as the log10 HIV DNA copy numbers per million cells.
Figure 5. HIV replication is inducible from…
Figure 5. HIV replication is inducible from the resting memory CD4 T cell subsets from post-treatment controllers.
The cell capacity to replicate HIV was evaluated in 7 PTCs (each symbol represents one PTC) by stimulating sorted-CD4 T cell subsets with an anti-CD3/anti-CD28 co-stimulation plus IL-2 and IL-7 (filled symbols and continuous lines) or IL-7 alone (empty symbols and dashed lines). HIV RNA was quantified in the supernatants of resting TN, TCM, TTM and TEM cells during a 13-day long culture. Results are expressed as the log10 of the ratio between the HIV RNA copy numbers quantified at a given day of culture and the level of cell-associated HIV DNA in the subset measured at D0 of culture. Kinetics of HIV production in a patient has been represented with connecting lines. HIV RNA values that were under the detection threshold of the technique were arbitrarily placed at 0. ND is not done.
Figure 6. Weak contribution of long-lived resting…
Figure 6. Weak contribution of long-lived resting CD4+ T cells to the HIV reservoir in the post-treatment controllers.
A. HIV infection levels in the resting TN, TCM, TTM and TEM cells of 11 PTCs and 8 HICs. The results are expressed as the log10 HIV DNA copy numbers per million cells, and the medians are represented. The open symbols are values below the threshold of detection. ‘ns’ are non significant p values. B. CD4+ T cell subsets contribution to the resting HIV reservoir, considering both infection levels and frequency. The results are expressed as the median percentage of the resting CD4 HIV reservoir, with interquartile range [25%–75%] and minimum and maximum values. Statistical analyses were applied between all subsets from a single group as well as between each subset from the two groups.
Figure 7. Interruption of long-term treatment initiated…
Figure 7. Interruption of long-term treatment initiated at PHI leads to a significant frequency of viremia control.
Kaplan-Meier curve of the probability for patients included in the FHDH between 1997 and 2011 to lose control of viremia after interruption of a, at least, one year-long cART initiated within 6 months of HIV infection, and who had at least one viral load determination 12 months after treatment interruption (n = 74). Loss of control was defined by 2 or more viral loads above 50 RNA copies/mL or one viral load above 50 RNA copies/mL followed by resumption of cART.

References

    1. Antiretroviral-Therapy-Cohort-Collaboration (2008) Life expectancy of individuals on combination antiretroviral therapy in high-income countries: a collaborative analysis of 14 cohort studies. Lancet 372: 293–299.
    1. Wong JK, Hezareh M, Gunthard HF, Havlir DV, Ignacio CC, et al. (1997) Recovery of replication-competent HIV despite prolonged suppression of plasma viremia. Science 278: 1291–1295.
    1. Bongiovanni M, Casana M, Tincati C, d'Arminio Monforte A (2006) Treatment interruptions in HIV-infected subjects. J Antimicrob Chemother 58: 502–505.
    1. Cain LE, Logan R, Robins JM, Sterne JA, Sabin C, et al. (2011) When to initiate combined antiretroviral therapy to reduce mortality and AIDS-defining illness in HIV-infected persons in developed countries: an observational study. Ann Intern Med 154: 509–515.
    1. Yerly S, Kaiser L, Perneger TV, Cone RW, Opravil M, et al. (2000) Time of initiation of antiretroviral therapy: impact on HIV-1 viraemia. The Swiss HIV Cohort Study. AIDS 14: 243–249.
    1. Delwart E, Magierowska M, Royz M, Foley B, Peddada L, et al. (2002) Homogeneous quasispecies in 16 out of 17 individuals during very early HIV-1 primary infection. AIDS 16: 189–195.
    1. Ngo-Giang-Huong N, Deveau C, Da Silva I, Pellegrin I, Venet A, et al. (2001) Proviral HIV-1 DNA in subjects followed since primary HIV-1 infection who suppress plasma viral load after one year of highly active antiretroviral therapy. AIDS 15: 665–673.
    1. Alter G, Teigen N, Davis BT, Addo MM, Suscovich TJ, et al. (2005) Sequential deregulation of NK cell subset distribution and function starting in acute HIV-1 infection. Blood 106: 3366–3369.
    1. Moir S, Buckner CM, Ho J, Wang W, Chen J, et al. (2010) B cells in early and chronic HIV infection: evidence for preservation of immune function associated with early initiation of antiretroviral therapy. Blood 116: 5571–5579.
    1. Oxenius A, Price DA, Easterbrook PJ, O'Callaghan CA, Kelleher AD, et al. (2000) Early highly active antiretroviral therapy for acute HIV-1 infection preserves immune function of CD8+ and CD4+ T lymphocytes. Proc Natl Acad Sci U S A 97: 3382–3387.
    1. Hecht FM, Wang L, Collier A, Little S, Markowitz M, et al. (2006) A multicenter observational study of the potential benefits of initiating combination antiretroviral therapy during acute HIV infection. J Infect Dis 194: 725–733.
    1. Rosenberg ES, Altfeld M, Poon SH, Phillips MN, Wilkes BM, et al. (2000) Immune control of HIV-1 after early treatment of acute infection. Nature 407: 523–526.
    1. Seng R, Goujard C, Desquilbet L, Sinet M, Rouzioux C, et al. (2008) Rapid CD4+ cell decrease after transient cART initiated during primary HIV infection (ANRS PRIMO and SEROCO cohorts). J Acquir Immune Defic Syndr 49: 251–258.
    1. Kaufmann DE, Lichterfeld M, Altfeld M, Addo MM, Johnston MN, et al. (2004) Limited durability of viral control following treated acute HIV infection. PLoS Med 1: e36.
    1. Hocqueloux L, Prazuck T, Avettand-Fenoel V, Lafeuillade A, Cardon B, et al. (2010) Long-term immunovirologic control following antiretroviral therapy interruption in patients treated at the time of primary HIV-1 infection. AIDS 24: 1598–1601.
    1. Goujard C, Chaix ML, Lambotte O, Deveau C, Sinet M, et al. (2009) Spontaneous control of viral replication during primary HIV infection: when is “HIV controller” status established? Clin Infect Dis 49: 982–986.
    1. Pereyra F, Jia X, McLaren PJ, Telenti A, de Bakker PI, et al. (2010) The major genetic determinants of HIV-1 control affect HLA class I peptide presentation. Science 330: 1551–1557.
    1. Boufassa F, Saez-Cirion A, Lechenadec J, Zucman D, Avettand-Fenoel V, et al. (2011) CD4 Dynamics over a 15 Year-Period among HIV Controllers Enrolled in the ANRS French Observatory. PLoS ONE 6: e18726 doi:
    1. Migueles SA, Sabbaghian MS, Shupert WL, Bettinotti MP, Marincola FM, et al. (2000) HLA B*5701 is highly associated with restriction of virus replication in a subgroup of HIV-infected long term nonprogressors. Proc Natl Acad Sci U S A 97: 2709–2714.
    1. Alizadeh M, Gagne K, Moine A, Bignon JD, Bensa JC, et al. (2008) Evidence for the improvement of French Volunteer Bone Marrow Donor Registry by using HLA-SBT at the registration step. Tissue Antigens 71: 315.
    1. Gao X, Nelson GW, Karacki P, Martin MP, Phair J, et al. (2001) Effect of a single amino acid change in MHC class I molecules on the rate of progression to AIDS. The New England journal of medicine 344: 1668–1675.
    1. Saez-Cirion A, Lacabaratz C, Lambotte O, Versmisse P, Urrutia A, et al. (2007) HIV controllers exhibit potent CD8 T cell capacity to suppress HIV infection ex vivo and peculiar cytotoxic T lymphocyte activation phenotype. Proc Natl Acad Sci U S A 104: 6776–6781.
    1. Gea-Banacloche JC, Migueles SA, Martino L, Shupert WL, McNeil AC, et al. (2000) Maintenance of large numbers of virus-specific CD8+ T cells in HIV-infected progressors and long-term nonprogressors. J Immunol 165: 1082–1092.
    1. Doisne JM, Urrutia A, Lacabaratz-Porret C, Goujard C, Meyer L, et al. (2004) CD8+ T cells specific for EBV, cytomegalovirus, and influenza virus are activated during primary HIV infection. J Immunol 173: 2410–2418.
    1. Hunt PW, Brenchley J, Sinclair E, McCune JM, Roland M, et al. (2008) Relationship between T cell activation and CD4+ T cell count in HIV-seropositive individuals with undetectable plasma HIV RNA levels in the absence of therapy. J Infect Dis 197: 126–133.
    1. Saez-Cirion A, Hamimi C, Bergamaschi A, David A, Versmisse P, et al. (2011) Restriction of HIV-1 replication in macrophages and CD4+ T cells from HIV controllers. Blood 118: 955–964.
    1. Lewin SR, Rouzioux C (2011) HIV cure and eradication: how will we get from the laboratory to effective clinical trials? AIDS 25: 885–897.
    1. Chomont N, DaFonseca S, Vandergeeten C, Ancuta P, Sekaly RP (2011) Maintenance of CD4+ T-cell memory and HIV persistence: keeping memory, keeping HIV. Current opinion in HIV and AIDS 6: 30–36.
    1. Descours B, Avettand-Fenoel V, Blanc C, Samri A, Melard A, et al. (2012) Immune responses driven by protective human leukocyte antigen alleles from long-term nonprogressors are associated with low HIV reservoir in central memory CD4 T cells. Clinical infectious diseases : an official publication of the Infectious Diseases Society of America 54: 1495–1503.
    1. Lodi SMLKAD, et al. (2012) Immunovirologic control 24 months after interruption of antiretroviral therapy initiated close to hiv seroconversion. Archives of Internal Medicine 172(16): 1252–1255.
    1. Goujard C, Girault I, Rouzioux C, Lecuroux C, Deveau C, et al. (2012) HIV-1 control after transient antiretroviral treatment initiated in primary infection: role of patient characteristics and effect of therapy. Antiviral therapy 17: 1001–1009.
    1. Okulicz JF, Marconi VC, Landrum ML, Wegner S, Weintrob A, et al. (2009) Clinical outcomes of elite controllers, viremic controllers, and long-term nonprogressors in the US Department of Defense HIV natural history study. J Infect Dis 200: 1714–1723.
    1. Huang J, Goedert JJ, Sundberg EJ, Cung TD, Burke PS, et al. (2009) HLA-B*35-Px-mediated acceleration of HIV-1 infection by increased inhibitory immunoregulatory impulses. J Exp Med 206: 2959–2966.
    1. Saez-Cirion A, Sinet M, Shin SY, Urrutia A, Versmisse P, et al. (2009) Heterogeneity in HIV suppression by CD8 T cells from HIV controllers: association with Gag-specific CD8 T cell responses. J Immunol 182: 7828–7837.
    1. Emu B, Sinclair E, Hatano H, Ferre A, Shacklett B, et al. (2008) HLA class I-restricted T-cell responses may contribute to the control of human immunodeficiency virus infection, but such responses are not always necessary for long-term virus control. J Virol 82: 5398–5407.
    1. Madec Y, Boufassa F, Porter K, Meyer L (2005) Spontaneous control of viral load and CD4 cell count progression among HIV-1 seroconverters. AIDS 19: 2001–2007.
    1. Strain MC, Little SJ, Daar ES, Havlir DV, Gunthard HF, et al. (2005) Effect of treatment, during primary infection, on establishment and clearance of cellular reservoirs of HIV-1. J Infect Dis 191: 1410–1418.
    1. Chun TW, Justement JS, Murray D, Hallahan CW, Maenza J, et al. (2010) Rebound of plasma viremia following cessation of antiretroviral therapy despite profoundly low levels of HIV reservoir: implications for eradication. AIDS 24: 2803–2808.
    1. Ganesan A, Chattopadhyay PK, Brodie TM, Qin J, Gu W, et al. (2010) Immunologic and virologic events in early HIV infection predict subsequent rate of progression. J Infect Dis 201: 272–284.
    1. Brenchley JM, Hill BJ, Ambrozak DR, Price DA, Guenaga FJ, et al. (2004) T-cell subsets that harbor human immunodeficiency virus (HIV) in vivo: implications for HIV pathogenesis. J Virol 78: 1160–1168.
    1. Paiardini M, Cervasi B, Reyes-Aviles E, Micci L, Ortiz AM, et al. (2011) Low levels of SIV infection in sooty mangabey central memory CD(4)(+) T cells are associated with limited CCR5 expression. Nature medicine 17: 830–836.
    1. Beaumier CM, Harris LD, Goldstein S, Klatt NR, Whitted S, et al. (2009) CD4 downregulation by memory CD4+ T cells in vivo renders African green monkeys resistant to progressive SIVagm infection. Nat Med 15: 879–885.
    1. Assomou L, Descamps D, Yerly S, Dos Santos G, Marcelin AG, et al. (2010) Prevalence of HIV-1 drug resistance in treated patients with viral load >50 copies/ml in 2009: a French nationwide study. Antiviral Therapy 15: A185–A185.
    1. Cohen MS, Chen YQ, McCauley M, Gamble T, Hosseinipour MC, et al. (2011) Prevention of HIV-1 Infection with Early Antiretroviral Therapy. N Engl J Med 365: 493–505.
    1. Saez-Cirion A, Shin SY, Versmisse P, Barre-Sinoussi F, Pancino G (2010) Ex vivo T cell-based HIV suppression assay to evaluate HIV-specific CD8+ T-cell responses. Nat Protoc 5: 1033–1041.
    1. Avettand-Fenoel V, Chaix ML, Blanche S, Burgard M, Floch C, et al. (2009) 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 81: 217–223.
    1. Avettand-Fenoel V, Prazuck T, Hocqueloux L, Melard A, Michau C, et al. (2008) HIV-DNA in rectal cells is well correlated with HIV-DNA in blood in different groups of patients, including long-term non-progressors. AIDS 22: 1880–1882.

Source: PubMed

3
Prenumerera