Вы находитесь на странице: 1из 9

See

discussions, stats, and author profiles for this publication at: https://www.researchgate.net/publication/237070034

Immune response to HIV

Article in Current opinion in HIV and AIDS · July 2013


DOI: 10.1097/COH.0b013e328361faf4 · Source: PubMed

CITATIONS READS

20 793

3 authors, including:

Matthieu Perreau Giuseppe Pantaleo


University Hospital of Lausanne University Hospital of Lausanne
60 PUBLICATIONS 1,285 CITATIONS 233 PUBLICATIONS 12,404 CITATIONS

SEE PROFILE SEE PROFILE

Some of the authors of this publication are also working on these related projects:

HIV reservoir View project

Immune responses to HCV View project

All content following this page was uploaded by Matthieu Perreau on 25 August 2017.

The user has requested enhancement of the downloaded file.


REVIEW

CURRENT
OPINION Immune response to HIV
Matthieu Perreau a, Yves Levy c, and Giuseppe Pantaleo a,b

Purpose of review
Major advances have been made in the delineation of HIV-specific immune response and in the
mechanisms of virus escape. The kinetics of the immunological and virological events occurring during
primary HIV infection indicate that the establishment of the latent HIV reservoir, the major obstacle to HIV
eradication likely occurs during the very early stages of primary infection, that is, the ‘eclipse phase’, prior
to the development of the HIV-specific immune response which has limited efficacy in the control of the
early events of infection. Therefore, the window of opportunity to develop effective interventions either to
clear HIV during primary infection or to prevent rebound of HIV in patients successfully treated who stop
antiretroviral therapy is very narrow.
Recent findings
Genetic factors most strongly associated with nonprogressive infection are human leukocyte antigen (HLA)
class I alleles and particularly HLA-B5701. CD4 and CD8 T-cell responses with polyfunctional profile are
associated with nonprogressive infection. Broader neutralizing antibodies are detected 3–4 years after
infection, generated only in 20% of individuals but show no efficacy in the control of HIV replication.
Summary
In the present review, we shall discuss the different components of the HIV-specific immune response
elicited by the infection, the kinetics of these responses during primary infection and the changes following
transition to the chronic phase of infection, and the functional profile of ‘effective’ versus ‘noneffective’
HIV-specific immune responses.
Keywords
antibodies, HIV, immune response, T cells

INTRODUCTION immunologically driven, clearance of HIV following


Primary HIV infection (PHI) is characterized by a infection. However, a small percentage (1–5% based
transient symptomatic illness (in 40–90% of cases) on case definition) of HIV-infected individuals
associated with high levels of virus replication. In experience stable disease, that is, lack of decline of
the large majority (>80%) of infected individuals, CD4 T-cell counts, and control of virus replication
the transmitted virus results from the infection of a below 1000 HIV RNA copies/ml for an extended
single virus variant as revealed by genome analysis period of time (at least 7–10 years) in the absence
of viral RNA isolated during PHI [1]. Symptoms are of antiretroviral therapy (ART) [6]. These individuals
not specific to HIV but typical of an acute viral are called long-term nonprogressors (LTNPs) [7].
syndrome and may include fever, fatigue, rash, Furthermore, an even smaller percentage (<1%) of
headache, lymphadenopathy, pharyngitis, myalgia, individuals show control of virus replication below
arthralgia, aseptic meningitis, retrorbital pain,
weight loss, depression, gastrointestinal distress
a
night sweats, and oral or genital ulcers [2]. The Division of Immunology and Allergy, bSwiss Vaccine Research Institute,
symptomatic phase generally lasts for 2–4 weeks Centre Hospitalier Universitaire Vaudois, University of Lausanne,
Lausanne, Switzerland and cINSERM U955, Université Paris Est Créteil,
in individuals with ‘normal’ rate of diseases pro-
Faculté de Médecine, Groupe Henri-Mondor Albert-Chenevier, Immuno-
gression, whereas severe and prolonged symptoms logie Clinique, Vaccine Research Institute, Creteil, France
are associated with rapid disease progression [3,4]. Correspondence to Matthieu Perreau, Division of Immunology and
Once chronic HIV infection is established, the Allergy, Centre Hospitalier Universitaire Vaudois, Rue Bugnon 46,
course of HIV disease and progression may be influ- 1011 Lausanne, Switzerland. Tel: +41 2 131 41073; e-mail: matthieu.
enced substantially by host (genetic and immuno- perreau@chuv.ch
logical) and virological factors [5]. At present, there Curr Opin HIV AIDS 2013, 8:333–340
have not been reports of spontaneous, that is, DOI:10.1097/COH.0b013e328361faf4

1746-630X ß 2013 Wolters Kluwer Health | Lippincott Williams & Wilkins www.co-hivandaids.com

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
Thirty years of HIV and AIDS

replication and spreading due to the large concen-


KEY POINTS tration of activated T cells, to the trapping of virions
 HIV-specific immune response lacks efficacy during on the extracellular surface of follicular dendritic
the early stages of HIV spreading during cells [21–23], and to the abundant production of
primary infection. pro-inflammatory cytokines such as interleukin-1
(IL-1), IL-6, or tumor necrosis factor a (TNF-a) that
 Establishment of the latent HIV reservoir occurs prior to
highly support HIV replication [24]. The immune
the development of operational immune responses.
response to HIV at the end of the ‘eclipse phase’ is
 HIV-specific neutralizing antibodies appear to have stimulated as indicated by the detection of proteins
limited efficacy in the control of virus replication during of the acute phase, by the initial detection of inflam-
chronic infection. matory cytokines resulting from the activation of
the innate immune response [24–26], and likely by
the stimulation of HIV-specific CD4 T cells. During
50 HIV RNA copies/ml regardless of the length of the the ‘eclipse phase’, no effector components of the
time of control and are called elite controllers [6]. immune response to HIV capable of containing
Rarely, the control of virus replication in LTNPs can the initial spreading of infection appear to be
be explained by the presence of defective virus [8], operational and after the amplification of the infec-
and most of them are infected with replication- tion in the draining lymph nodes, HIV viral RNA
competent virus, thus indicating that host factors becomes detectable in the plasma (Feibig Stage I)
predominantly contribute to virus control and pre- and spreading of the infection to other anatomic
vention of disease progression. compartments such as the gut-associated lymphoid
tissues (GALTs) occurs [27]. At the end of the ‘eclipse
phase’, HIV begins to spread to the GALT that is
KINETICS OF IMMUNE RESPONSE DURING predominantly populated of CD4 CCR5þ memory
PRIMARY INFECTION T cells. This results in an exponential expansion of
In order to discuss the kinetics of the different infection, with up to 20% of gut resident CD4 T cells
components of the immune response elicited by being infected and 80% depleted [27–31]. The
HIV infection, we will use as a reference the Stages large depletion of CD4 T cells occurring in the
defined by Fiebig on the basis of the kinetics of HIV lymphoid tissues during this stage is indicated by
viral RNA and the appearance of an HIV-specific the detection of a number of markers of apoptosis
antibody response [9,10]. such as TRAIL, microparticles containing CCR5,
Of particular importance is the initial period of TNFR2, and soluble FAS ligand as viremia increases
infection defined by the so-called ‘eclipse phase’. [32]. By week 3–4, the levels of viremia peak (gener-
This phase last for 10 days and defines the period ally >107 virus particles per ml of plasma) [9] and it
when the virus is not detectable in the plasma [1,11]. is just at this time that antibody response becomes
Studies performed in rhesus macaques infected with detectable.
simian immunodeficiency virus (SIV) [12,13] have The first wave of antibody response is predom-
been instrumental to delineate the events occurring inantly composed of anti-gp41 IgG antibodies,
in the ‘eclipse phase’. Virus-infected cells cannot be which form immune complexes with HIV virions
detected in the mucosa until 1–3 days after infec- [33]. Immune complexes have been found in the
tion and the first infected cells are resting memory large majority of individuals (90%) during PHI, but
CD4 T cells expressing CCR5. In addition, submu- only a minority (about 20%) of virions was opson-
cosal dendritic cells and Langerhans cells uptake the ised by anti-gp41 antibodies and forming immune
virus from the mucosa through C-type lectins such complexes [33]. The anti-gp41 antibodies bind
as DC-SIGN [14,15], which binds HIV gp120 with both infectious and noninfectious virions and lack
high affinity [14,15] and internalizes virions and neutralizing activity [33]. Anti-gp120 antibodies
subsequently expresses them on the cell surface after represent a minor component of the antibody
the dendritic cells have matured and migrated to response during PHI while become the dominant
draining lymph nodes where they encounter T cells anti-Env antibody response after the transition to
[14,16–18]. At 1 week postinfection, that is versus the chronic phase of infection. As viremia peaks and
the end of the ‘eclipse period’, the virus becomes following initial decline, antibodies specific to the
detectable in the draining lymph nodes where virus other HIV proteins such as Gag are detected by week
production may be amplified as result of spreading 3–4 of infection (Feibig Stages III/IV). Taken
of infection among activated T cells that have together, the above observations indicate that the
come in contact with dendritic cells [19,20]. The antibody response during PHI has no efficacy in the
environment of the lymphoid tissue is ideal for virus containment of HIV replication and spreading.

334 www.co-hivandaids.com Volume 8  Number 4  July 2013

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
Immune response to HIV Perreau et al.

HIV-specific CD8 T-cell response is detected with a broad neutralizing activity against a wide
during the increase in viremia levels and generally range of virus isolates [46–53] remains limited
peaks 1–2 weeks after viremia peak and the peak in and the majority of neutralizing antibodies has
CD8 T-cell response coincides with the decline in narrow breadth [54].
viremia levels [34,35]. The first wave of CD8 T-cell Numerous phenotypical and functional abnor-
response is directed predominantly against Env and malities of the B-cell compartment have been
Nef HIV proteins. The initial CD8 T-cell response described in HIV-infected individuals [55]. These
appears to have limited or no effect on HIV repli- abnormalities include abnormal B-cell activation
cation as indicated by the persistence of a homo- [56], appearance of transitional immature B cells
geneous founder virus without evidence of and exhausted B cells [57,58], and progressive loss
immune-driven selection of virus escape mutants of recall responses to vaccination [59].
[36]. It is only at the time of the peak CD8 T-cell Passive administration of neutralizing anti-
response and the decline of viremia that numerous bodies in nonhuman primates showed protection
amino acid mutations occur in epitopes of the against SIV challenge, but was dependent upon the
founder virus recognized by CD8 T cells [36]. No titer and the quality of the antibodies [60,61]. In
mutations are observed in the escape mutants [36]. particular, the injection of a cocktail of neutralizing
The initial CD8 T-cell response is associated with a antibodies was able to prevent SIV infection after
massive expansion of HIV-specific CD8 T cells [37]. mucosal virus challenge [62–67]. However, B-cell
Despite the massive expansion, the initial CD8 depletion in monkeys with chronic SIV infection
T-cell response has likely limited efficacy in the does not influence the levels of plasma viremia [68],
control of virus replication because of the rapid thus suggesting that B cells have limited protective
emergence of virus escape mutants and of the rapid role during chronic infection.
disappearance of the responding CD8 T-cell clones Slow progression of HIV disease has been found
due to the lack of recognition of the escape mutants to be associated with high titers of anti-p24 anti-
or of exhaustion resulting from high virus/Ag load bodies [69], with neutralizing antibodies directed
[36,38]. against autologous virus [70], or with antibodies
In the absence of ART, the levels of viremia start against certain gp120 epitopes [71]. Antibodies with
to decline around week 4 after infection in order to broader neutralizing activity have been found more
reach the ‘virologic’ set point generally 12–16 weeks frequently in LTNPs [72–76]. However, it is unclear
after infection, which marks the transition of the whether these antibody responses reflect mechan-
infection to the chronic phase [39]. The contri- isms of protection or rather the integrity of the
bution of the immune response to the decline of immune system in LTNPs.
viremia is only partial and predominantly mediated Finally, mucosal HIV-specific immunoglobulin
by HIV-specific cytotoxic CD8 T cells that may kill A (IgA) response has been detected in HIV-exposed
HIV-infected CD4 T cells. However, a major mech- uninfected individuals [77]. These IgA may prevent
anism of the decline may be the lack of cell targets, potentially HIV transcytosis through the epithelium
that is, CD4 T cells, for HIV due to the large [78]. IgA antibodies specific for the gp120-CD4-
depletion of CD4 T cells through direct virus cytho- binding site have also been detected in LTNPs [79].
pathic effect, killing by CD8 T cells and activation-
induced cell death. There is no evidence that the
antibody response plays any role in the partial sup- THE ROLE OF CD8 T-CELLS IN VIRUS
pression of the decline of viremia and establishment CONTROL
of the ‘virologic’ set point. CD8 T cells play a critical role in antiviral immunity
[34,35,80–84]. There are a number of observations
underscoring the importance of HIV-specific CD8
ANTIBODY RESPONSE T cells in the control of virus replication and preven-
Neutralizing antibodies against autologous virus are tion of disease progression in HIV-infected indivi-
detected in a large proportion of HIV-infected indi- duals. These include the rapid loss of control of virus
viduals about 3 months after infection [40] and their replication after CD8 T-cell depletion in SIV-infected
appearance is associated with the emergence of monkeys [80,81,85], the decline of viremia during
mutations in the Env region [41,42]. Neutralizing PHI coinciding with the peak of CD8 T-cell response
antibodies with broad neutralizing activity are [35], the presence of CD8 T-cell responses in exposed
generated only in a minority (about 20%) of indi- uninfected individuals [86–89], and the association
viduals and appear 2–4 years after infection, but between protection and HIV-specific CD8 T-cell
there is no evidence of their efficacy in the control of responses restricted by certain human leukocyte
viremia [43–45]. The number of human antibodies antigen (HLA) class I alleles [90,91].

1746-630X ß 2013 Wolters Kluwer Health | Lippincott Williams & Wilkins www.co-hivandaids.com 335

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
Thirty years of HIV and AIDS

CD8 T-cell responses from elite controllers The superior efficacy of CD8 T cells from elite
compared to progressors have superior capacity to controllers might also reflect the preservation of the
secrete multiple cytokines and chemokines (poly- CD4 TCM-cell compartment and of proliferation
functionality), particularly IL-2 [92], to proliferate capacity, which was observed in elite controllers
[93], to inhibit HIV replication in vitro and kill target as compared to untreated and treated chronically
cells [83,94], to express perforin and degranulate HIV-infected individuals [121,122,128,131].
upon HIV-specific stimulation [95–98], and to tar- In addition, CD4 T cells from elite controllers do
get Gag epitopes [99,100]. CD8 T-cell responses from not show signs of immune exhaustion such as loss of
elite controllers also inhibited HIV replication with- polyfunctionality and expression of coinhibitory
out prior activation in vitro, thus suggesting the molecules [117,132]. Therefore, these cells are able
presence of a pool of CD8 T cells with immediate to secrete IL-2, IL-21, and possibly other cytokines
or rapidly inducible antiviral effector function needed to help CD8 T cells in developing and main-
[92,101–105]. Efficient viral suppression in vitro taining their effector functions as well as their pro-
is predominantly mediated by CD8 T cells specific liferation capacity and survival [101,121,122,133].
to Gag rather than Env or Nef [106,107], consist-
ently with the observed protection associated with
Gag-specific CD8 T-cell responses detected ex vivo THE ROLE OF HLA ALLELES IN CONTROL
[99]. Whole-genome analysis has demonstrated that
the genetic factors most strongly associated with
nonprogressive infection are HLA class I alleles
THE ROLE OF CD4 T-CELLS IN VIRUS and the activating killer cell Ig-like receptor (KIR)
CONTROL allele KIR3DS [90,134–141]. In particular, the HLA-
Memory CD4 T cells are the primary targets of HIV B5701, HLA-B5801, and the HLA-B2705 are
[108], and massive depletion of CD4 T cells, particu- associated with slow disease progression, whereas
larly of HIV-specific CD4 T cells [109], occurs during HLA-B35 with faster disease progression [139,142].
PHI. Long-term ART only partially restores the pool Protective CD8 T-cell responses restricted by HLA-
of total and HIV-specific memory CD4 T cells B5701 and HLA-B27 recognize the following
[27,28,110,111]. immunodominant Gag epitopes: TSTLQEQIGW
HIV-specific CD4 T cells harbor a skewed func- (TW10), KAFSEPVIPMF (KF11), ISPRTLNAW (IW9)
tional profile associated with a reduced IL-2 pro- restricted by HLA-B5701, and KRWIILGLNK (KK10)
duction and proliferation capacity [95,112,113]. restricted by HLA-B27 [138,143]. The low viremia is
This functional impairment was later associated associated with high rate of sequence mutation
with the expression of coinhibitory molecules [pro- (reflecting the immunological pressure exerted on
grammed death 1 [114–116], cytotoxic T lympho- epitopes restricted by HLA-B57), induction of virus
cyte antigen 4 (CTLA-4) [117]]. Interestingly, a variants with reduced replication capacity, and the
recent study highlighted the functional impairment ability to recognize epitopes with escape mutations
of follicular helper T cells (Tfh) as responsible for [144,145]. The effect of HLA-B on disease outcome
inadequate B-cell help, which may explain some of seems to be mediated in part by the amino acid
the functional abnormalities in the B-cell functions position located in the peptide-binding groove,
&&
observed in HIV infection [118 ]. In this regard, it suggesting a difference in the conformation of the
has been recently shown that Tfh serve as the major peptide presentation [90]. Indeed, intrinsic differ-
cell reservoir for HIV, which, in turn, may be respon- ences in self-peptide binding among HLA molecules
&&
sible for their functional impairment [119 ]. are important during T-cell repertoire development.
Several evidence underscore the importance of As fewer self-peptides are able to bind to HLA-B57
CD4 T-cell responses in the control HIV [120]. These molecules, fewer CD8 T cells restricted by this HLA
include the enrichment in LTNPs and elite con- will undergo negative selection. HLA-B57-restricted
trollers of polyfunctional HIV-specific CD4 T cells CD8 T cells have higher tolerability for epitope
and preservation of proliferation capacity [84,121– mutations: several mutations in the peptide recog-
125], the greater production of IL-21 by HIV-specific nized do not abrogate the recognition capacity by
CD4 T cells in HIV controllers [126,127], the presence T cells. These observations explain also the associ-
of HIV-specific CD4 T cells with higher functional ation between HLA-B57 and control of hepatitis C
avidity and T-cell receptor-binding affinity in LTNPs virus (HCV) infection and the association between
[128], the association of some HLA class II alleles with HLA-B57 and HLA-B27 and enhanced risk of auto-
lower viral loads [129,130], and the lower expression immunity [146–148]. Along the same line, KK10-
of CTLA-4 and other exhaustion markers in HIV- specific CD8 T-cell clones isolated from elite con-
specific CD4 T cells in LTNPs [117]. trollers expressing HLA-B27 were characterized by

336 www.co-hivandaids.com Volume 8  Number 4  July 2013

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
Immune response to HIV Perreau et al.

8. Kirchhoff F, Greenough TC, Brettler DB, et al. Brief report: absence of intact
complementarity determining regions 3 (CDR3) nef sequences in a long-term survivor with nonprogressive HIV-1 infection.
sequences significantly closer to germline sequen- N Engl J Med 1995; 332:228–232.
9. Fiebig EW, Wright DJ, Rawal BD, et al. Dynamics of HIV viremia and antibody
ces, superior ability to control virus replication seroconversion in plasma donors: implications for diagnosis and staging of
in vitro, and superior cross-reactivity than KK10- primary HIV infection. AIDS 2003; 17:1871–1879.
10. McMichael AJ, Borrow P, Tomaras GD, et al. The immune response during
specific CD8 T-cell clones isolated from progressors acute HIV-1 infection: clues for vaccine development. Nat Rev Immunol
[105]. 2010; 10:11–23.
11. Lee HY, Giorgi EE, Keele BF, et al. Modeling sequence evolution in acute
HIV-1 infection. J Theor Biol 2009; 261:341–360.
12. Li Q, Duan L, Estes JD, et al. Peak SIV replication in resting memory CD4þ
CONCLUSION T cells depletes gut lamina propria CD4þ T cells. Nature 2005; 434:1148–
1152.
HIV-specific T-cell and antibody responses have 13. Pope M, Haase AT. Transmission, acute HIV-1 infection and the quest for
strategies to prevent infection. Nat Med 2003; 9:847–852.
been extensively characterized in HIV infection. 14. Geijtenbeek TB, Kwon DS, Torensma R, et al. DC-SIGN, a dendritic cell-
Despite the fact that the magnitude of these specific HIV-1-binding protein that enhances trans-infection of T cells. Cell
2000; 100:587–597.
responses is orders of magnitude greater as com- 15. Geijtenbeek TB, Torensma R, van Vliet SJ, et al. Identification of DC-SIGN, a
pared to those elicited by other viruses establishing novel dendritic cell-specific ICAM-3 receptor that supports primary immune
responses. Cell 2000; 100:575–585.
chronic infections, HIV-specific responses fail to 16. Kwon DS, Gregorio G, Bitton N, et al. DC-SIGN-mediated internalization of
effectively control HIV replication and disease pro- HIV is required for trans-enhancement of T cell infection. Immunity 2002;
16:135–144.
gression in the absence of ART in the large majority 17. Fong L, Mengozzi M, Abbey NW, et al. Productive infection of plasmacytoid
(>95%) of HIV-infected individuals. Several genetic dendritic cells with human immunodeficiency virus type 1 is triggered by
CD40 ligation. J Virol 2002; 76:11033–11041.
factors and functional profile of the HIV-specific 18. Fonteneau JF, Larsson M, Beignon AS, et al. Human immunodeficiency virus
immune response (particularly in the T-cell type 1 activates plasmacytoid dendritic cells and concomitantly induces the
bystander maturation of myeloid dendritic cells. J Virol 2004; 78:5223–5232.
response) are associated with protection in LTNPs 19. Pope M, Betjes MG, Romani N, et al. Conjugates of dendritic cells and
and elite controllers. These observations together memory T lymphocytes from skin facilitate productive infection with HIV-1.
Cell 1994; 78:389–398.
with the recent identification of Tfh cells as the 20. Lore K, Smed-Sorensen A, Vasudevan J, et al. Myeloid and plasmacytoid
major cell reservoir for HIV infection may guide dendritic cells transfer HIV-1 preferentially to antigen-specific CD4þ T cells.
J Exp Med 2005; 201:2023–2033.
the future development of therapeutic interven- 21. Carter CC, Onafuwa-Nuga A, McNamara LA, et al. HIV-1 infects multipotent
tions aimed at the development of strategies that progenitor cells causing cell death and establishing latent cellular reservoirs.
Nat Med 2010; 16:446–451.
may either stimulate effective T-cell immunity or 22. Embretson J, Zupancic M, Ribas JL, et al. Massive covert infection of helper
efficiently target the major HIV cell reservoir, that is, T lymphocytes and macrophages by HIV during the incubation period of
AIDS. Nature 1993; 362:359–362.
Tfh cells. 23. Pantaleo G, Graziosi C, Demarest JF, et al. HIV infection is active and
progressive in lymphoid tissue during the clinically latent stage of disease.
Nature 1993; 362:355–358.
Acknowledgements 24. Stacey AR, Norris PJ, Qin L, et al. Induction of a striking systemic
None. cytokine cascade prior to peak viremia in acute human immunodefi-
ciency virus type 1 infection, in contrast to more modest and delayed
responses in acute hepatitis B and C virus infections. J Virol 2009;
Conflicts of interest 83:3719–3733.
25. Kramer HB, Lavender KJ, Qin L, et al. Elevation of intact and proteolytic
The authors declare no conflict of interest. fragments of acute phase proteins constitutes the earliest systemic antiviral
response in HIV-1 infection. PLoS Pathog 2010; 6:e1000893.
26. Borrow P, Bhardwaj N. Innate immune responses in primary HIV-1 infection.
Curr Opin HIV AIDS 2008; 3:36–44.
REFERENCES AND RECOMMENDED 27. Brenchley JM, Schacker TW, Ruff LE, et al. CD4þ T cell depletion during all
READING stages of HIV disease occurs predominantly in the gastrointestinal tract.
Papers of particular interest, published within the annual period of review, have J Exp Med 2004; 200:749–759.
been highlighted as: 28. Mattapallil JJ, Douek DC, Hill B, et al. Massive infection and loss of memory
& of special interest CD4þ T cells in multiple tissues during acute SIV infection. Nature 2005;
&& of outstanding interest 434:1093–1097.
Additional references related to this topic can also be found in the Current 29. Veazey RS, Lackner AA. The gastrointestinal tract and the pathogenesis of
World Literature section in this issue (pp. 357–359). AIDS. AIDS 1998; 12:S35–S42.
30. Veazey RS, DeMaria M, Chalifoux LV, et al. Gastrointestinal tract as a major
1. Keele BF, Giorgi EE, Salazar-Gonzalez JF, et al. Identification and character- site of CD4þ T cell depletion and viral replication in SIV infection. Science
ization of transmitted and early founder virus envelopes in primary HIV-1 1998; 280:427–431.
infection. Proc Natl Acad Sci U S A 2008; 105:7552–7557. 31. Schindler M, Munch J, Kutsch O, et al. Nef-mediated suppression of T cell
2. Kahn JO, Walker BD. Acute human immunodeficiency virus type 1 infection. activation was lost in a lentiviral lineage that gave rise to HIV-1. Cell 2006;
N Engl J Med 1998; 339:33–39. 125:1055–1067.
3. Dorrucci M, Rezza G, Vlahov D, et al. Clinical characteristics and prognostic 32. Gasper-Smith N, Crossman DM, Whitesides JF, et al. Induction of plasma
value of acute retroviral syndrome among injecting drug users. Italian (TRAIL), TNFR-2 Fas ligand, and plasma microparticles after human immu-
Seroconversion Study. AIDS 1995; 9:597–604. nodeficiency virus type 1 (HIV-1) transmission: implications for HIV-1 vaccine
4. Henrard DR, Phillips JF, Muenz LR, et al. Natural history of HIV-1 cell-free design. J Virol 2008; 82:7700–7710.
viremia. JAMA: J Am Med Assoc 1995; 274:554–558. 33. Liu P, Overman RG, Yates NL, et al. Dynamic antibody specificities and virion
5. Liu SL, Schacker T, Musey L, et al. Divergent patterns of progression to AIDS concentrations in circulating immune complexes in acute to chronic HIV-1
after infection from the same source: human immunodeficiency virus type 1 infection. J Virol 2011; 85:11196–11207.
evolution and antiviral responses. J Virol 1997; 71:4284–4295. 34. Borrow P, Lewicki H, Hahn BH, et al. Virus-specific CD8þ cytotoxic
6. Walker BD, Korber BT. Immune control of HIV: the obstacles of HLA and viral T-lymphocyte activity associated with control of viremia in primary human
diversity. Nat Immunol 2001; 2:473–475. immunodeficiency virus type 1 infection. J Virol 1994; 68:6103–6110.
7. Pantaleo G, Menzo S, Vaccarezza M, et al. Studies in subjects with long-term 35. Koup RA, Safrit JT, Cao Y, et al. Temporal association of cellular immune
nonprogressive human immunodeficiency virus infection. N Engl J Med 1995; responses with the initial control of viremia in primary human immunodefi-
332:209–216. ciency virus type 1 syndrome. J Virol 1994; 68:4650–4655.

1746-630X ß 2013 Wolters Kluwer Health | Lippincott Williams & Wilkins www.co-hivandaids.com 337

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
Thirty years of HIV and AIDS

36. Goonetilleke N, Liu MK, Salazar-Gonzalez JF, et al. The first T cell response to 63. Ferrantelli F, Kitabwalla M, Rasmussen RA, et al. Potent cross-group
transmitted/founder virus contributes to the control of acute viremia in HIV-1 neutralization of primary human immunodeficiency virus isolates with mono-
infection. J Exp Med 2009; 206:1253–1272. clonal antibodies: implications for acquired immunodeficiency syndrome
37. Pantaleo G, Demarest JF, Soudeyns H, et al. Major expansion of CD8þ vaccine. J Infect Dis 2004; 189:71–74.
T cells with a predominant V beta usage during the primary immune response 64. Xiao P, Patterson LJ, Kuate S, et al. Replicating adenovirus-simian immuno-
to HIV. Nature 1994; 370:463–467. deficiency virus (SIV) recombinant priming and envelope protein boosting
38. Soudeyns H, Paolucci S, Chappey C, et al. Selective pressure exerted by elicits localized, mucosal IgA immunity in rhesus macaques correlated with
immunodominant HIV-1-specific cytotoxic T lymphocyte responses during delayed acquisition following a repeated low-dose rectal SIV(mac251)
primary infection drives genetic variation restricted to the cognate epitope. challenge. J Virol 2012; 86:4644–4657.
Eur J Immunol 1999; 29:3629–3635. 65. Hessell AJ, Rakasz EG, Tehrani DM, et al. Broadly neutralizing monoclonal
39. Rizzardi GP, Harari A, Capiluppi B, et al. Treatment of primary HIV-1 infection antibodies 2F5 and 4E10 directed against the human immunodeficiency
with cyclosporin A coupled with highly active antiretroviral therapy. J Clin virus type 1 gp41 membrane-proximal external region protect against mu-
Invest 2002; 109:681–688. cosal challenge by simian-human immunodeficiency virus SHIVBa-L. J Virol
40. Alter G, Moody MA. The humoral response to HIV-1: new insights, renewed 2010; 84:1302–1313.
focus. J Infect Dis 2010; 202 (Suppl 2):S315–S322. 66. Hessell AJ, Poignard P, Hunter M, et al. Effective, low-titer antibody protec-
41. Frost SD, Trkola A, Gunthard HF, et al. Antibody responses in primary HIV-1 tion against low-dose repeated mucosal SHIV challenge in macaques. Nat
infection. Curr Opin HIV AIDS 2008; 3:45–51. Med 2009; 15:951–954.
42. Richman DD, Wrin T, Little SJ, et al. Rapid evolution of the neutralizing 67. Hessell AJ, Rakasz EG, Poignard P, et al. Broadly neutralizing human anti-
antibody response to HIV type 1 infection. Proc Natl Acad Sci U S A 2003; HIV antibody 2G12 is effective in protection against mucosal SHIV chal-
100:4144–4149. lenge even at low serum neutralizing titers. PLoS Pathog 2009; 5:
43. Gray ES, Moore PL, Choge IA, et al. Neutralizing antibody responses in acute e1000433.
human immunodeficiency virus type 1 subtype C infection. J Virol 2007; 68. Schmitz JE, Kuroda MJ, Santra S, et al. Effect of humoral immune responses
81:6187–6196. on controlling viremia during primary infection of rhesus monkeys with simian
44. Shen X, Parks RJ, Montefiori DC, et al. In vivo gp41 antibodies targeting the immunodeficiency virus. J Virol 2003; 77:2165–2173.
2F5 monoclonal antibody epitope mediate human immunodeficiency virus 69. Hogervorst E, Jurriaans S, de Wolf F, et al. Predictors for non and slow
type 1 neutralization breadth. J Virol 2009; 83:3617–3625. progression in human immunodeficiency virus (HIV) type 1 infection: low viral
45. Stamatatos L, Morris L, Burton DR, et al. Neutralizing antibodies generated RNA copy numbers in serum and maintenance of high HIV-1 p24-specific but
during natural HIV-1 infection: good news for an HIV-1 vaccine? Nat Med not V3-specific antibody levels. J Infect Dis 1995; 171:811–821.
2009; 15:866–870. 70. Montefiori DC, Pantaleo G, Fink LM, et al. Neutralizing and infection-enhan-
46. Calarese DA, Scanlan CN, Zwick MB, et al. Antibody domain exchange is an cing antibody responses to human immunodeficiency virus type 1 in long-
immunological solution to carbohydrate cluster recognition. Science 2003; term nonprogressors. J Infect Dis 1996; 173:60–67.
300:2065–2071. 71. Wong MT, Warren RQ, Anderson SA, et al. Longitudinal analysis of the
47. Conley AJ, Kessler JA 2nd, Boots LJ, et al. Neutralization of divergent human humoral immune response to human immunodeficiency virus type 1 (HIV-1)
immunodeficiency virus type 1 variants and primary isolates by IAM-41-2F5, gp160 epitopes in rapidly progressing and nonprogressing HIV-1-infected
an antigp41 human monoclonal antibody. Proc Natl Acad Sci U S A 1994; subjects. J Infect Dis 1993; 168:1523–1527.
91:3348–3352. 72. Sajadi MM, Guan Y, DeVico AL, et al. Correlation between circulating HIV-1
48. Nelson JD, Brunel FM, Jensen R, et al. An affinity-enhanced neutralizing RNA and broad HIV-1 neutralizing antibody activity. J Acquir Immune Defic
antibody against the membrane-proximal external region of human immuno- Syndr 2011; 57:9–15.
deficiency virus type 1 gp41 recognizes an epitope between those of 2F5 73. Banerjee K, Klasse PJ, Sanders RW, et al. IgG subclass profiles in
and 4E10. J Virol 2007; 81:4033–4043. infected HIV type 1 controllers and chronic progressors and in uninfected
49. Roben P, Moore JP, Thali M, et al. Recognition properties of a panel of human recipients of Env vaccines. AIDS Res Hum Retroviruses 2010; 26:445–
recombinant Fab fragments to the CD4 binding site of gp120 that show 458.
differing abilities to neutralize human immunodeficiency virus type 1. J Virol 74. Bello G, Velasco-de-Castro CA, Bongertz V, et al. Immune activation and
1994; 68:4821–4828. antibody responses in nonprogressing elite controller individuals infected
50. Stiegler G, Kunert R, Purtscher M, et al. A potent cross-clade neutralizing with HIV-1. J Med Virol 2009; 81:1681–1690.
human monoclonal antibody against a novel epitope on gp41 of human 75. Pereyra F, Palmer S, Miura T, et al. Persistent low-level viremia in HIV-1 elite
immunodeficiency virus type 1. AIDS Res Hum Retroviruses 2001; controllers and relationship to immunologic parameters. J Infect Dis 2009;
17:1757–1765. 200:984–990.
51. Trkola A, Pomales AB, Yuan H, et al. Cross-clade neutralization of primary 76. Lambotte O, Ferrari G, Moog C, et al. Heterogeneous neutralizing antibody
isolates of human immunodeficiency virus type 1 by human monoclonal and antibody-dependent cell cytotoxicity responses in HIV-1 elite controllers.
antibodies and tetrameric CD4-IgG. J Virol 1995; 69:6609–6617. AIDS 2009; 23:897–906.
52. Walker LM, Phogat SK, Chan-Hui PY, et al. Broad and potent neutralizing 77. Bunders M, Pembrey L, Kuijpers T, et al. Evidence of impact of maternal HIV
antibodies from an African donor reveal a new HIV-1 vaccine target. Science infection on immunoglobulin levels in HIV-exposed uninfected children. AIDS
2009; 326:285–289. Res Hum Retroviruses 2010; 26:967–975.
53. Zwick MB, Labrijn AF, Wang M, et al. Broadly neutralizing antibodies 78. Shen R, Drelichman ER, Bimczok D, et al. GP41-specific antibody blocks
targeted to the membrane-proximal external region of human immunodefi- cell-free HIV type 1 transcytosis through human rectal mucosa and model
ciency virus type 1 glycoprotein gp41. J Virol 2001; 75:10892–10905. colonic epithelium. J Immunol 2010; 184:3648–3655.
54. Moore JP, Trkola A, Korber B, et al. A human monoclonal antibody to a 79. Planque S, Salas M, Mitsuda Y, et al. Neutralization of genetically diverse HIV-
complex epitope in the V3 region of gp120 of human immunodeficiency virus 1 strains by IgA antibodies to the gp120-CD4-binding site from long-term
type 1 has broad reactivity within and outside clade B. J Virol 1995; 69:122– survivors of HIV infection. AIDS 2010; 24:875–884.
130. 80. Jin X, Bauer DE, Tuttleton SE, et al. Dramatic rise in plasma viremia after
55. Moir S, Fauci AS. B cells in HIV infection and disease. Nat Rev Immunol CD8(þ) T cell depletion in simian immunodeficiency virus-infected maca-
2009; 9:235–245. ques. J Exp Med 1999; 189:991–998.
56. Lane HC, Masur H, Edgar LC, et al. Abnormalities of B-cell activation and 81. Schmitz JE, Kuroda MJ, Santra S, et al. Control of viremia in simian im-
immunoregulation in patients with the acquired immunodeficiency syndrome. munodeficiency virus infection by CD8þ lymphocytes. Science 1999;
N Engl J Med 1983; 309:453–458. 283:857–860.
57. Moir S, Fauci AS. Pathogenic mechanisms of B-lymphocyte dysfunction in 82. Moore CB, John M, James IR, et al. Evidence of HIV-1 adaptation to HLA-
HIV disease. J Allergy Clin Immunol 2008; 122:12–19; quiz 20 11. restricted immune responses at a population level. Science 2002; 296:
58. Moir S, Ho J, Malaspina A, et al. Evidence for HIV-associated B cell 1439–1443.
exhaustion in a dysfunctional memory B cell compartment in HIV-infected 83. Hersperger AR, Pereyra F, Nason M, et al. Perforin expression directly ex vivo
viremic individuals. J Exp Med 2008; 205:1797–1805. by HIV-specific CD8 T-cells is a correlate of HIV elite control. PLoS Patho-
59. De Milito A. B lymphocyte dysfunctions in HIV infection. Curr HIV Res 2004; gens 2010; 6:e1000917.
2:11–21. 84. Pereyra F, Addo MM, Kaufmann DE, et al. Genetic and immunologic
60. Mascola JR, Stiegler G, VanCott TC, et al. Protection of macaques against heterogeneity among persons who control HIV infection in the absence of
vaginal transmission of a pathogenic HIV-1/SIV chimeric virus by passive therapy. J Infect Dis 2008; 197:563–571.
infusion of neutralizing antibodies. Nat Med 2000; 6:207–210. 85. O’Connor DH, Allen TM, Vogel TU, et al. Acute phase cytotoxic T lymphocyte
61. Baba TW, Liska V, Hofmann-Lehmann R, et al. Human neutralizing mono- escape is a hallmark of simian immunodeficiency virus infection. Nat Med
clonal antibodies of the IgG1 subtype protect against mucosal simian-human 2002; 8:493–499.
immunodeficiency virus infection. Nat Med 2000; 6:200–206. 86. Perez CL, Hasselrot K, Bratt G, et al. Induction of systemic HIV-1-specific
62. Ferrantelli F, Rasmussen RA, Buckley KA, et al. Complete protection of cellular immune responses by oral exposure in the uninfected partner of
neonatal rhesus macaques against oral exposure to pathogenic simian- discordant couples. AIDS 2010; 24:969–974.
human immunodeficiency virus by human anti-HIV monoclonal antibodies. 87. Hasselrot K. Genital and oral mucosal immune response against HIV-1 in
J Infect Dis 2004; 189:2167–2173. exposed uninfected individuals. Crit Rev Immunol 2009; 29:369–377.

338 www.co-hivandaids.com Volume 8  Number 4  July 2013

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
Immune response to HIV Perreau et al.

88. Hasselrot K, Bratt G, Hirbod T, et al. Orally exposed uninfected individuals 115. D’Souza M, Fontenot AP, Mack DG, et al. Programmed death 1 expression
have systemic anti-HIV responses associating with partners’ viral load. AIDS on HIV-specific CD4þ T cells is driven by viral replication and associated
2010; 24:35–43. with T cell dysfunction. J Immunol 2007; 179:1979–1987.
89. Erickson AL, Willberg CB, McMahan V, et al. Potentially exposed but 116. Porichis F, Kwon DS, Zupkosky J, et al. Responsiveness of HIV-specific CD4
uninfected individuals produce cytotoxic and polyfunctional human immuno- T cells to PD-1 blockade. Blood 2011; 118:965–974.
deficiency virus type 1-specific CD8(þ) T-cell responses which can be 117. Kaufmann DE, Kavanagh DG, Pereyra F, et al. Upregulation of CTLA-4 by
defined to the epitope level. Clin Vaccine Immunol 2008; 15:1745–1748. HIV-specific CD4þ T cells correlates with disease progression and defines a
90. Pereyra F, Jia X, McLaren PJ, et al. The major genetic determinants of HIV-1 reversible immune dysfunction. Nat Immunol 2007; 8:1246–1254.
control affect HLA class I peptide presentation. Science 2010; 330:1551– 118. Cubas RA, Mudd JC, Savoye AL, et al. Inadequate T follicular cell help impairs
1557. && B cell immunity during HIV infection. Nat Med 2013; 19:494–499.

91. Fellay J, Shianna KV, Ge D, et al. A whole-genome association study of major This study demonstrates that the function of T follicular helper cells is abnormal in
determinants for host control of HIV-1. Science 2007; 317:944–947. HIV-infected individuals with chronic HIV infection. These results may help to
92. Betts MR, Nason MC, West SM, et al. HIV nonprogressors preferentially explain why B-cell and antibody responses are impaired in HIV infection.
maintain highly functional HIV-specific CD8þ T cells. Blood 2006; 119. Perreau M, Savoye AL, De Crignis E, et al. Follicular helper T cells serve as
107:4781–4789. && the major CD4 T cell compartment for HIV-1 infection, replication, and

93. Ndhlovu ZM, Chibnik LB, Proudfoot J, et al. High-dimensional immunomo- production. J Exp Med 2013; 210:143–156.
nitoring models of HIV-1-specific CD8 T-cell responses accurately identify In this study, the distribution of HIV-specific and HIV-infected CD4 T cells within
subjects achieving spontaneous viral control. Blood 2013; 121:801–811. different populations of memory CD4 T cells isolated from lymph nodes of viremic
94. Migueles SA, Laborico AC, Shupert WL, et al. HIV-specific CD8þ T cell HIV-infected individuals was examined. The study shows that the Tfh cell popula-
proliferation is coupled to perforin expression and is maintained in nonpro- tion is enriched in HIV-specific CD4 T cells, and is significantly increased in viremic
gressors. Nat Immunol 2002; 3:1061–1068. HIV-infected individuals. The Tfh cell population contained the highest percentage
95. Harari A, Petitpierre S, Vallelian F, et al. Skewed representation of functionally of CD4 T cells harboring HIV DNA and was the most efficient in supporting
distinct populations of virus-specific CD4 T cells in HIV-1-infected subjects productive infection in vitro. Furthermore, only the percentage of Tfh cells corre-
with progressive disease: changes after antiretroviral therapy. Blood 2004; lated with plasma viremia levels.
103:966–972. 120. Theze J, Chakrabarti LA, Vingert B, et al. HIV controllers: a multifactorial
96. Harari A, Dutoit V, Cellerai C, et al. Functional signatures of protective phenotype of spontaneous viral suppression. Clin Immunol 2011; 141:15–
antiviral T-cell immunity in human virus infections. Immunol Rev 2006; 30.
211:236–254. 121. Potter SJ, Lacabaratz C, Lambotte O, et al. Preserved central memory and
97. Harari A, Cellerai C, Pantaleo G. Role of HIV-1-specific CD4 T cells. Curr activated effector memory CD4þ T-cell subsets in human immunodeficiency
Opin HIV AIDS 2006; 1:22–27. virus controllers: an ANRS EP36 study. J Virol 2007; 81:13904–13915.
98. Wherry EJ, Blattman JN, Murali-Krishna K, et al. Viral persistence alters CD8 122. Younes SA, Yassine-Diab B, Dumont AR, et al. HIV-1 viremia prevents the
T-cell immunodominance and tissue distribution and results in distinct stages establishment of interleukin 2-producing HIV-specific memory CD4þ T cells
of functional impairment. J Virol 2003; 77:4911–4927. endowed with proliferative capacity. J Exp Med 2003; 198:1909–1922.
99. Kiepiela P, Ngumbela K, Thobakgale C, et al. CD8þ T-cell responses to 123. Betts MR, Ambrozak DR, Douek DC, et al. Analysis of total human immu-
different HIV proteins have discordant associations with viral load. Nat Med nodeficiency virus (HIV)-specific CD4(þ) and CD8(þ) T-cell responses:
2007; 13:46–53. relationship to viral load in untreated HIV infection. J Virol 2001; 75:11983–
100. Zuniga R, Lucchetti A, Galvan P, et al. Relative dominance of Gag p24- 11991.
specific cytotoxic T lymphocytes is associated with human immunodeficiency 124. Harari A, Cellerai C, Enders FB, et al. Skewed association of polyfunctional
virus control. J Virol 2006; 80:3122–3125. antigen-specific CD8 T cell populations with HLA-B genotype. Proc Natl
101. Saez-Cirion A, Lacabaratz C, Lambotte O, et al. HIV controllers exhibit potent Acad Sci U S A 2007; 104:16233–16238.
CD8 T cell capacity to suppress HIV infection ex vivo and peculiar cytotoxic 125. Ferre AL, Hunt PW, McConnell DH, et al. HIV controllers with HLA-DRB113
T lymphocyte activation phenotype. Proc Natl Acad Sci U S A 2007; and HLA-DQB106 alleles have strong, polyfunctional mucosal CD4þ T-cell
104:6776–6781. responses. J Virol 2010; 84:11020–11029.
102. Ferre AL, Hunt PW, Critchfield JW, et al. Mucosal immune responses to HIV- 126. Yue FY, Lo C, Sakhdari A, et al. HIV-specific IL-21 producing CD4þ T cells
1 in elite controllers: a potential correlate of immune control. Blood 2009; are induced in acute and chronic progressive HIV infection and are asso-
113:3978–3989. ciated with relative viral control. J Immunol 2010; 185:498–506.
103. Migueles SA, Weeks KA, Nou E, et al. Defective human immunodeficiency 127. Chevalier MF, Julg B, Pyo A, et al. HIV-1-specific interleukin-21þ CD4þ
virus-specific CD8þ T-cell polyfunctionality, proliferation, and cytotoxicity are T cell responses contribute to durable viral control through the modulation of
not restored by antiretroviral therapy. J Virol 2009; 83:11876–11889. HIV-specific CD8þ T cell function. J Virol 2011; 85:733–741.
104. Makedonas G, Hutnick N, Haney D, et al. Perforin and IL-2 upregulation 128. Vingert B, Perez-Patrigeon S, Jeannin P, et al. HIV controller CD4þ T cells
define qualitative differences among highly functional virus-specific human respond to minimal amounts of Gag antigen due to high TCR avidity. PLoS
CD8 T cells. PLoS Pathogens 2010; 6:e1000798. Pathog 2010; 6:e1000780.
105. Chen H, Ndhlovu ZM, Liu D, et al. TCR clonotypes modulate the protective 129. Malhotra U, Holte S, Dutta S, et al. Role for HLA class II molecules in HIV-1
effect of HLA class I molecules in HIV-1 infection. Nat Immunol 2012; suppression and cellular immunity following antiretroviral treatment. J Clin
13:691–700. Invest 2001; 107:505–517.
106. Saez-Cirion A, Sinet M, Shin SY, et al. Heterogeneity in HIV suppression by 130. Julg B, Pereyra F, Buzon MJ, et al. Infrequent recovery of HIV from but robust
CD8 T cells from HIV controllers: association with Gag-specific CD8 T cell exogenous infection of activated CD4(þ) T cells in HIV elite controllers. Clin
responses. J Immunol 2009; 182:7828–7837. Infect Dis 2010; 51:233–238.
107. Julg B, Williams KL, Reddy S, et al. Enhanced anti-HIV functional activity 131. Emu B, Sinclair E, Favre D, et al. Phenotypic, functional, and kinetic para-
associated with Gag-specific CD8 T-cell responses. J Virol 2010; 84:5540– meters associated with apparent T-cell control of human immunodeficiency
5549. virus replication in individuals with and without antiretroviral treatment. J Virol
108. Schnittman SM, Lane HC, Greenhouse J, et al. Preferential infection of 2005; 79:14169–14178.
CD4þ memory T cells by human immunodeficiency virus type 1: evidence for 132. Deeks SG, Walker BD. Human immunodeficiency virus controllers: mechan-
a role in the selective T-cell functional defects observed in infected indivi- isms of durable virus control in the absence of antiretroviral therapy. Immunity
duals. Proc Natl Acad Sci U S A 1990; 87:6058–6062. 2007; 27:406–416.
109. Douek DC, Brenchley JM, Betts MR, et al. HIV preferentially infects HIV- 133. van Grevenynghe J, Procopio FA, He Z, et al. Transcription factor FOXO3a
specific CD4þ T cells. Nature 2002; 417:95–98. controls the persistence of memory CD4(þ) T cells during HIV infection. Nat
110. Guadalupe M, Reay E, Sankaran S, et al. Severe CD4þ T-cell depletion in gut Med 2008; 14:266–274.
lymphoid tissue during primary human immunodeficiency virus type 1 infec- 134. Kiepiela P, Leslie AJ, Honeyborne I, et al. Dominant influence of HLA-B in
tion and substantial delay in restoration following highly active antiretroviral mediating the potential co-evolution of HIV and HLA. Nature 2004;
therapy. J Virol 2003; 77:11708–11717. 432:769–775.
111. Rosenberg ES, Billingsley JM, Caliendo AM, et al. Vigorous HIV-1-specific 135. Chopera DR, Mlotshwa M, Woodman Z, et al. Virological and immunological
CD4þ T cell responses associated with control of viremia. Science 1997; factors associated with HIV-1 differential disease progression in HLA-
278:1447–1450. B58:01-positive individuals. J Virol 2011; 85:7070–7080.
112. Harari A, Rizzardi GP, Ellefsen K, et al. Analysis of HIV-1- and CMV-specific 136. Peretz Y, Marra O, Thomas R, et al. Relative contribution of HIV-specific
memory CD4 T-cell responses during primary and chronic infection. Blood functional lymphocyte subsets restricted by protective and non-protective
2002; 100:1381–1387. HLA alleles. Viral Immunol 2011; 24:189–198.
113. Harari A, Vallelian F, Meylan PR, et al. Functional heterogeneity of memory 137. Payne RP, Kloverpris H, Sacha JB, et al. Efficacious early antiviral activity of
CD4 T cell responses in different conditions of antigen exposure and HIV Gag- and Pol-specific HLA-B 2705-restricted CD8þ T cells. J Virol
persistence. J Immunol 2005; 174:1037–1045. 2010; 84:10543–10557.
114. Day CL, Kaufmann DE, Kiepiela P, et al. PD-1 expression on HIV-specific T 138. Migueles SA, Sabbaghian MS, Shupert WL, et al. HLA B5701 is highly
cells is associated with T-cell exhaustion and disease progression. Nature associated with restriction of virus replication in a subgroup of HIV-infected
2006; 443:350–354. long term nonprogressors. Proc Nal Acad Sci U S A 2000; 97:2709–2714.

1746-630X ß 2013 Wolters Kluwer Health | Lippincott Williams & Wilkins www.co-hivandaids.com 339

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.
Thirty years of HIV and AIDS

139. Carrington M, O’Brien SJ. The influence of HLA genotype on AIDS. Annu Rev 144. Bailey JR, Williams TM, Siliciano RF, et al. Maintenance of viral suppression in
Med 2003; 54:535–551. HIV-1-infected HLA-B57þ elite suppressors despite CTL escape muta-
140. Martin MP, Gao X, Lee JH, et al. Epistatic interaction between KIR3DS1 tions. J Exp Med 2006; 203:1357–1369.
and HLA-B delays the progression to AIDS. Nat Genet 2002; 31:429– 145. Miura T, Brumme CJ, Brockman MA, et al. HLA-associated viral mutations are
434. common in human immunodeficiency virus type 1 elite controllers. J Virol
141. Bashirova AA, Thomas R, Carrington M. HLA/KIR restraint of HIV: surviving 2009; 83:3407–3412.
the fittest. Annu Rev Immunol 2011; 29:295–317. 146. Thio CL, Gao X, Goedert JJ, et al. HLA-Cw04 and hepatitis C virus
142. Carrington M, Walker BD. Immunogenetics of spontaneous control of HIV. persistence. J Virol 2002; 76:4792–4797.
Ann Rev Med 2012; 63:131–145. 147. Chessman D, Kostenko L, Lethborg T, et al. Human leukocyte antigen class
143. Goulder PJ, Bunce M, Krausa P, et al. Novel, cross-restricted, conserved, I-restricted activation of CD8þ T cells provides the immunogenetic basis of a
and immunodominant cytotoxic T lymphocyte epitopes in slow progres- systemic drug hypersensitivity. Immunity 2008; 28:822–832.
sors in HIV type 1 infection. AIDS Res Hum Retroviruses 1996; 12: 148. Bowness P. HLA B27 in health and disease: a double-edged sword?
1691–1698. Rheumatology 2002; 41:857–868.

340 www.co-hivandaids.com Volume 8  Number 4  July 2013

Copyright © Lippincott Williams & Wilkins. Unauthorized reproduction of this article is prohibited.

View publication stats

Вам также может понравиться