HIV reservoirs and the possibility of a cure for HIV infection

Page created by Clifford Barton
 
CONTINUE READING
Symposium            |                                                                                     doi: 10.1111/j.1365-2796.2011.02457.x

HIV reservoirs and the possibility of a cure for HIV infection
   S. Palmer1,2, L. Josefsson1,2 & J. M. Coffin3
From the 1Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet; 2Department of Virology, Swedish Institute for Infectious
Disease Control, Solna, Sweden, and 3Department of Molecular Biology and Microbiology, Tufts University, Boston, MA, USA

Abstract. Palmer S, Josefsson L, Coffin JM (Karolinska                        suboptimal; and ⁄ or (iii) proliferation of latently in-
Institutet, Solna; Swedish Institute for Infectious Dis-                      fected cells with regeneration of a stable reservoir of
ease Control, Solna, Sweden; and Tufts University,                            slowly dividing infected cells. A well-defined latent
Boston, MA, USA). HIV reservoirs and the possibility                          reservoir of HIV is memory CD4+ T-cells where la-
of a cure for HIV infection (Symposium). J Intern Med                         tency is established when an activated CD4+ T-cell
2011; doi: 10.1111/j.1365-2796.2011.02457.x.                                  becomes infected by HIV, but transitions to a termi-
                                                                              nally differentiated memory cell before it is elimi-
Recent studies demonstrate that suppressive ther-                             nated. This review examines the dynamics and possi-
apy can drive HIV-1 RNA levels to less than 50 cop-                           ble reservoirs of persistent HIV in patients on
ies mL)1 in patient plasma. Yet, ultrasensitive assays                        suppressive therapy, the mechanisms promoting
show that most patients continue to harbour low-                              viral latency and strategies to purge latent viral reser-
level persistent viremia. Treatment intensification                           voirs. The promising research described here takes a
studies indicate that low-level viremia could arise                           number of steps forward to seriously address HIV
from several different sources. These sources in-                             remission and ⁄ or eradication.
clude: (i) long-lived HIV-infected cells that replicate
and produce virus; (ii) ongoing replication cycles in                         Keywords: HIV eradication, HIV latency, HIV persis-
cells located in sanctuary sites where drug levels are                        tence, HIV reservoirs.

                                                                              development and many other related fields. In this
Introduction
                                                                              article we review research into the source and dynam-
The development of combination antiretroviral ther-                           ics of persistent viremia. We will focus on studies
apy remains one of the great triumphs of modern                               describing the levels and genetic make-up of persis-
medicine. Despite its unquestioned success, current                           tent viremia during suppressive therapy and what
therapy for the treatment of human immunodefi-                                this research can tell us about viral reservoirs and
ciency virus (HIV) has a number of limitations. For                           mechanisms of HIV latency. In the final section we
example, although effective and life saving, it is not                        will discuss current strategies for reactivation of the
curative and does not eradicate infection. During                             latent HIV reservoir, a step likely to be crucial for its
combination antiretroviral therapy, reduction of HIV                          eradication.
RNA levels to less than 50 copies mL)1 is frequently
achieved; however, residual low-level viremia can be
                                                                              Persistent HIV viremia
detected in most patients using ultrasensitive assays
[1–4]. Furthermore, interruption of treatment results                         Highly active antiretroviral therapy (HAART) effec-
in a rapid viral rebound arising from the persistent,                         tively suppresses HIV RNA levels from an average of
residual viremia. Notably, this persistent viremia is                         about 30 000 to below 50 copies mL)1 (the lower
evident even after seven years of therapy [5]. There-                         detection limit of most commercial assays used in
fore, effective therapy requires life-long adherence,                         clinical practice) in the plasma of infected patients.
which many patients find difficult to achieve.                                However, if treatment is interrupted, the virus will re-
                                                                              bound and become detectable again, often within two
The eradication of human immunodeficiency virus                               weeks [6]. Therefore, HAART does not eliminate HIV
(HIV) from infected individuals is one of the major                           infection and residual low-level viremia has been de-
medical challenges of our time. For the past 25 years,                        tected using ultrasensitive assays [2, 3, 7–9] in 80%
this effort has attracted researchers in the fields of                        of treated patients. Notably, this viremia can persist
infectious disease, virology, molecular biology, drug                         for at least 7 years of suppressive therapy [5].

                                                                                    ª 2011 The Association for the Publication of the Journal of Internal Medicine   1
S. Palmer et al.
                                    |                                                                           Symposium: HIV reservoirs and HIV infection

Owing to the fact that HAART prevents the transmis-                                                  ing viral replication is contributing to persistent vire-
sion of infection to new cells and the half-life of plas-                                            mia in patients on effective HAART regimens. At least
ma virions is 6 h, the levels of HIV RNA in the plasma                                               four studies, using a number of different treatment
are strongly correlated to the half-life of the cells pro-                                           protocols, have shown no perceptible change in per-
ducing the virus [10, 11]. After introduction of HA-                                                 sistent viremia in patients receiving intensified treat-
ART, viremia declines rapidly and mathematical                                                       ment or statistical difference between the intensified
modelling has revealed at least four phases of viral de-                                             and placebo arms of the study, implicating long-lived
cay corresponding to the half lives of different popula-                                             cells as the probable source [14–17]. This conclusion
tions of HIV-infected cells (Fig. 1). As HAART prevents                                              is supported by studies that demonstrate the lack of
new cycles of HIV infection, but has no effect on the                                                further evolution in the persistent virus population.
death rate or viral production of already infected cells,                                            Indeed, the emergence of genetically homogeneous
each phase of the decline in viremia reflects the death                                              subpopulations can often be observed in patients un-
or elimination of a different population of virus-in-                                                der long-term treatment, as well as in the viral popu-
fected cells. During the fourth phase, there is little or                                            lation that rebounds during treatment interruption
no viral decay and the levels of plasma HIV RNA re-                                                  [18, 19]. The genetic homogeneity of these viral popu-
main stable ranging from £1–5 copies mL)1 with an                                                    lations suggests that persistent viremia arises from
overall median viral load of 3 copies mL)1 [1–3, 5].                                                 long-lived latently infected cells rather than ongoing
                                                                                                     cycles of replicating virus. Further support for this
There are two different views regarding the source an-                                               comes from earlier studies which revealed that the le-
d ⁄ or dynamics of persistent viremia in patients on                                                 vel of persistent viremia is not related to treatment
long-term suppressive therapy. One view holds that                                                   regimen but to pretreatment viral RNA levels, sug-
persistent viremia is the result of ongoing cycles of vir-                                           gesting that the pretreatment viral set-point is corre-
al replication [2, 12, 13]. The other view maintains                                                 lated to the number of long-lived HIV-infected cells
that this residual viremia is from long-lived cells that                                             [3].
are infected prior to treatment initiation [3]. To settle
this debate, the source of persistent viremia was re-                                                However, additional studies of treatment intensifica-
cently characterized by quantifying the levels of vire-                                              tion with raltegravir, which blocks integration of HIV
mia in patients on suppressive regimens (viral                                                       DNA into chromosomal DNA, have produced appar-
load < 50 copies mL)1) who ‘intensify’ their standard                                                ent conflicting results. In a minority (30%) of patients,
regimen of three drugs by adding a fourth drug such                                                  an increase in episomal (unintegrated) HIV DNA was
as raltegravir. If persistent viremia decreases during                                               observed leading to the conclusion that active replica-
this treatment intensification, it would suggest ongo-                                               tion persists in some infected individuals on suppres-

                           HAART
        HIV-1 RNA

                                                                                                                                 Fig. 1 The four phases of viral
                                                                                                                                 decay after the initiation of
                                                                                                                                 highly active antiretroviral ther-
                        < 50 copies/mL
                                                                                                                                 apy and the virion producing cell
                                                                                                                                 populations related to each
                                                                                                                                 phase. The yellow cells represent
                                                                                                                                 activated CD4+ T-cells, the green
                                                                                                                                 cells represent partially acti-
                    Time                                                                                                         vated CD4+ T-cells or other cell
Phase                       1                       2                          3                            4                    types, the blue cells represent
Half time               1 – 2 days               2 weeks                   39 weeks                                              resting memory CD4+ T-cell sub-
Cell type           Activated CD4+ T-cells   Partially activated        Resting memory              Resting memory
                                                                                                                                 sets or other cells and the red
                                             CD4+ T-cells,              CD4+ T-cells                CD4+ T-cells
                                             macrophages,               - central                   - central                    cells represent long-lived HIV-in-
                                             dendritic cells            - transitional              - transitional               fected cells which could be rest-
                                                                        - effector                  - effector
                                                                        - terminally                - terminally                 ing memory CD4+ T-cells, or sub-
                                                                        differentiated              differentiated               sets of these cells.

2    ª 2011 The Association for the Publication of the Journal of Internal Medicine Journal of Internal Medicine
S. Palmer et al.
                          |                                                          Symposium: HIV reservoirs and HIV infection

                                            (a)                                                           (b)
                                                                                                                                  Central nervous system/CSF

                                                          Activated CD4+ T-cell
Fig. 2 Possible reservoirs of
persistent HIV virus. HIV persis-                                                                                                     Peripheral blood
tence in resting long-lived mem-                                                                            Lymph node tissue
                                                         Becomes HIV infected
ory CD4+ T-cells: an activated
CD4+ T-cell becomes infected
and then subsequently transi-                                            HIV integrates
tions to a resting memory pheno-                                         into cellular genome                                   Gut associated lymphoid tissue
type (a). A number of findings
suggest that the reservoir is lar-                                                                          Bone marrow tissue
gely established and maintained                                 Transition to
in human tissues (b). Determin-                                 memory CD4+ T-cell
                                             Antigen
ing the type of cells and reservoir
producing persistent HIV is cru-
cial for future efforts aimed at
HIV eradication as these cells                    Cellular activation and viral production
could be targeted by new thera-
pies.

sive therapy [20]. In addition, a recent raltegravir                       eral different types of HIV-infected cells may contrib-
intensification study with anatomic analysis of HIV                        ute to this viral decay including macrophages, par-
infection in the gastrointestinal tract revealed no de-                    tially activated CD4+ T-cells and different types of
creases in peripheral viremia but decreases were                           dendritic cells. Macrophages are less susceptible to
measured in unspliced HIV RNA in CD4+ T-cells ob-                          the viral cytopathic effects than activated CD4+
tained from the terminal ileum [21]. These results                         T-cells and the half-life of tissue macrophages can be
indicate that low-level viremia might arise from sev-                      several weeks [24, 25]. Partially activated HIV-
eral different sources. These sources may include: (i)                     infected CD4+ T-cells have been reported to have a
long-lived HIV-infected cells that produce virus; (ii)                     longer turnover time than fully activated CD4+ T-cells
ongoing replication cycles in cells located in sanctu-                     [9]. Although it is unknown how long dendritic cells
ary sites where drug levels are suboptimal such as a                       (DCs) can survive after HIV infection, in vitro studies
tissue-based foci of viral replication within CD4+ T-                      show that infected myeloid dendritic cells can survive
cells and ⁄ or myeloid cells; and ⁄ or (iii) proliferation of              for more than 45 days [26]. Langerhans cells
latently infected cells with regeneration of a stable                      (dendritic cells in the skin) have been shown to have a
reservoir of slowly dividing infected cells [1, 4, 9–11].                  half-life of about 15 days when infected [27, 28],
                                                                           whilst DCs in the mucosa have a shorter half-life of
                                                                           only 2–3 days [29]. However, HIV has not been found
Viral reservoirs and sanctuary sites
                                                                           in DCs from the peripheral blood following 6 weeks of
During untreated infection, HIV predominantly repli-                       HAART which suggests that these cells do not con-
cates in activated CD4+ T-cells which die quickly                          tribute to long-term HIV persistence [30]. Follicular
either from viral cytopathic effect or from immune re-                     dendritic cells trap virions on their surface and these
sponse [9]. After suppressive HAART is initiated,                          virions can remain infectious for at least 9 months
infection of new cells is prevented. Cells that are al-                    [31] but it is unclear whether their slow release con-
ready infected produce virus and die as they would                         tributes to the different phases of decay. It has also
have without therapy. Measurement of decay of virus                        been observed that in HIV-infected patients treated
in plasma as a function of time since initiation of ther-                  with a raltegravir-containing regimen, the proportion
apy thus reveals the kinetics of death of infected cells.                  of virus attributable to the second phase is greatly
The bulk of the plasma virus – 90% or so – decays in                       reduced, suggesting that integration is delayed in the
the first phase with a half-life of 1–2 days, and almost                   cells that make-up this phase [32, 33].
certainly reflects infection and killing of activated
CD4+ T-cells (Fig. 1) [22, 23].                                            The cellular sources of the third and fourth phases of
                                                                           decay have not been fully identified but these phases
The kinetics of the second phase of decay is slower                        most likely represent additional classes of long-lived
with a half-life of 12–14 days. The elimination of sev-                    virus-producing cells. One well-accepted latent

                                                        ª 2011 The Association for the Publication of the Journal of Internal Medicine Journal of Internal Medicine   3
S. Palmer et al.
                                 |                                                                             Symposium: HIV reservoirs and HIV infection

reservoir of HIV is the stable infection of resting long-                                            fraction (perhaps 1% or so) of the total HIV DNA–
lived memory CD4+ T-cells. These cells may become                                                    positive cells in patients on long-term suppressive
infected directly or, more likely, when an activated                                                 therapy that is capable of being induced to make
CD4+ T-cell becomes infected and then subse-                                                         infectious virus in vitro. The large majority of provi-
quently transitions to a resting memory phenotype                                                    ruses are apparently incapable of activation, owing to
before HIV infection eliminates the cell (Fig. 2a).                                                  accumulation of inactivating mutations, unfavour-
Transition to a memory cell is associated with a                                                     able integration sites or some other cellular feature.
reduction in transcription factors required for HIV                                                  At the moment, there is no way to physically distin-
replication. Thus, switching to a memory cell allows                                                 guish amongst these populations.
this infected host cell to persist for years until it re-
ceives a stimulatory signal that activates the cell and                                              Although the bulk of evidence favours the conclusion
concomitantly induces viral production. Resting                                                      that persistent viremia is owing to virus production
memory CD4+ T-cells isolated from infected individ-                                                  from cells infected prior to initiation of therapy, it re-
uals on suppressive therapy can be activated by                                                      mains possible that low-level viral replication may oc-
agents like anti-CD3 or by the combination of cyto-                                                  cur in anatomical compartments or tissues where
kines TNF-a, IL-2 and IL-6 to produce infectious                                                     drug levels are suboptimal (Fig. 2b) [21]. Only about
virus [34, 35]. During antiretroviral therapy, these                                                 2% of lymphocytes are in the circulation, the remain-
inducible cells decay very slowly with an average                                                    der are spread throughout the body, especially in
half-life of 44 months, indicating that under current                                                lymphoid organs such as the spleen, lymph nodes
treatment it will take over 60 years to deplete this                                                 and gut-associated lymphoid tissue (GALT) where
reservoir [36]. The frequency of infected resting                                                    the majority of viral replication takes place during un-
memory CD4+ T-cells is low, about one infected cells                                                 treated infection [41–44]. In fact, large numbers of
per 106 resting memory CD4+ T-cells (about 100-fold                                                  lymphocytes are sequestered in the gastrointestinal
less than total HIV DNA–positive cells), and this res-                                               (GI) tract. During the acute phase of HIV infection,
ervoir is established during primary HIV infection                                                   CD4+ lymphocytes located in the GI tract are depleted
[37, 38].                                                                                            and remain so throughout the course of the disease
                                                                                                     [13–15]. During suppressive therapy, the CD4+ T-
Recently, Chomont et al. have shown that during                                                      cells in the GI tract are slowly but incompletely re-
HAART, integrated HIV DNA can be found in two                                                        stored [45]. Although antiretroviral therapy greatly
different subsets of CD4+ T-cells: central memory                                                    reduces HIV replication and immune activation, it is
T-cells (TCM) and transitional memory T-cells (TTM).                                                 unclear whether low-level ongoing HIV replication is
They found that TCM cells are the major viral reservoir                                              continuing in the gut of treated patients.
for patients with relatively high CD4+ T-cell counts,
and the low proliferation rate of these cells allows                                                 Also of note, patients infected with HIV-1 present
them to persist at adequate levels for years. In                                                     hematopoietic abnormalities which are caused by
patients with low CD4+ counts, the major reservoir is                                                HIV infection of the bone marrow [16]. A recent study
TTM cells and these cells persist by homoeostatic pro-                                               has shown that HIV infects multipotent hematopoi-
liferation making them a very stable viral reservoir.                                                etic progenitor cells (HPCs) and that latent HIV infec-
Furthermore, they observed that in individuals who                                                   tion was established in some of these HPCs [17]. The
have a lower CD4+ count the stability of the TTM reser-                                              authors note in this study that further research is
voir is mediated through IL-7 induced proliferation                                                  now needed to test whether persistent circulating
and that in these individuals plasma levels of IL-7                                                  virus in patients on suppressive therapy is partially
correlated inversely to the rate of decrease of the                                                  derived from HPCs as was shown for resting memory
reservoir. This study suggests that there are at least                                               T-cells.
two mechanisms by which the reservoir of infected
resting memory CD4+ T-cells is maintained [39].                                                      In addition to the lymphoid system, the central ner-
Recent studies indicate that the persistent viremia in                                               vous system (CNS) is a target for HIV-1 infection; and
50% of patients is derived from memory CD4+ T-cells,                                                 during primary infection, viral RNA has been detected
which suggests that other virus-producing cellular                                                   in the cerebrospinal fluid (CSF) of patients [46, 47].
sources may be responsible for the persistent viremia                                                This infection causes a local inflammatory response
during the fourth phase of decay during HAART [18,                                                   which is detectable in the CSF of some HIV-infected
40]. An important caveat in the interpretation of these                                              individuals, although, in many, HIV-1 infection of the
studies is that they assume that the properties of the                                               CNS is not clinically apparent [48]. In a subset of pa-
bulk proviral DNA are representative of the small                                                    tients during the late stages of disease, HIV-1 infec-

4    ª 2011 The Association for the Publication of the Journal of Internal Medicine Journal of Internal Medicine
S. Palmer et al.
                           |                                                   Symposium: HIV reservoirs and HIV infection

tion of the CNS evolves to encephalitis and dementia.                delve into mechanisms of latency have, for the most
In addition, opportunistic infections of the CNS in-                 part, been confined to cells infected and manipulated
crease during late stage HIV infection. Recent studies               ex vivo.
suggest the CNS is a viral sanctuary site, as the HIV
genetic populations in plasma and CSF are nearly                     As the chromatin environment influences the expres-
identical during primary infection but as the disease                sion of HIV, mechanisms which promote HIV latency
progresses, these populations diverge [49, 50]. More-                in infected CD4+ T-cells include histone and DNA
over, patients with HIV-associated dementia have the                 modification in the viral long terminal repeat (LTR).
most divergent HIV populations between plasma and                    The recruitment of cellular histone deacetylases
CSF. The advent of HAART has reduced the levels of                   (HDACs) and CpG (CG-rich DNA) methylation, which
HIV RNA in the CSF and the incidence of dementia in                  closes the LTR chromatin structure, prevent HIV
HIV-infected patients has also greatly decreased [51,                transcription and expression [60, 61]. The extent of
52]. However, many drugs have suboptimal penetra-                    DNA methylation has been studied in infected indi-
tion of the CNS owing to restrictions of these drugs in              viduals and more hypermethylation was found in pa-
crossing the blood-brain and blood-CSF barriers                      tients with 50 copies mL)1, and a recent study
drugs allows the continual viral replication during                  demonstrated that CpG methylation is greater in la-
HAART not only resulting in HIV drug resistance but                  tently infected than in productively infected primary
is also the development of HIV-associated cognitive                  human T- cells [62]. Although CpG methylation once
impairments. It is unknown whether this persistent                   established is perhaps the major factor suppressing
virus in the CNS is contributing to the persistent vire-             transcription of much of the genome, its establish-
mia found in plasma of patients on suppressive ther-                 ment may depend on other factors which block gene
apy, although the genetic similarity of both the persis-             expression in the first place. Thus, in the case of HIV,
tent virus population and the virus that appears                     this modification is secondary to some other suppres-
following discontinuation of therapy to the prethera-                sive mechanism, such as lack of an appropriate acti-
py plasma virus argues against this possibility. In                  vator. Furthermore, it is unclear whether reactivation
addition, a recent treatment intensification study re-               of hypermethylated proviruses is a feasible mecha-
vealed no decreases in plasma or CSF HIV RNA levels                  nism to explain persistent viremia.
or changes in soluble inflammatory markers such as
neopterin [54]. These results suggest that persistent                Several host transcription factors are maintained at
viremia may arise from sources other than the CNS                    low levels in resting CD4+ T-cells including nuclear
compartment.                                                         factor kappa B (NF-kB), nuclear factor of activated
                                                                     T-cells (NFAT) and positive elongation factor b (P-
In untreated patients, there is compartmentalization                 TEFb). The up-regulation of these host factors may
of virus between genital secretions and plasma [55,                  activate latently HIV-infected cells and increase HIV
56]. During HAART, HIV can be found in the semen,                    expression. In addition, low concentrations or im-
both as free virus and integrated DNA [57]. Interest-                paired activity of the viral activator protein Tat and
ingly, HIV has also been found in renal epithelium                   low levels of the cellular protein PTB, which causes
[58]. It remains to be determined to what extent                     a post-transcriptional block and degradation or
GALT, CNS and the genitourinary tract contributes to                 translational suppression of viral mRNA, have been
the production of virions and the persistence of HIV                 shown to be important mechanisms of HIV latency.
during HAART.                                                        (Reviewed in [63]).

                                                                     The site of viral integration into the host genome can
Mechanism of HIV latency
                                                                     affect the transcription rate of HIV. The integration of
The best-defined HIV reservoir is a small pool of la-                HIV proviruses has been found to occur in both inac-
tently infected resting memory CD4+ T-cells carrying                 tively and actively transcribed genes [64, 65]. The
an integrated form of the viral genome [59]. The pre-                reduction of viral transcription in actively transcribed
cise molecular mechanisms contributing to the gen-                   gene regions of resting memory CD4+ T-cells may be
eration and maintenance of this reservoir, however,                  owing to transcriptional interference [66]. Transcrip-
remain to be elucidated. As noted before, the scarcity               tional interference takes place when cellular tran-
of such cells, even amongst those carrying integrated                scripts initiated outside the integration site of HIV
HIV DNA, makes it virtually impossible to identify,                  read through the HIV genome and disrupt the tran-
isolate and study them directly. Therefore, studies to               scription of HIV by displacing the viral transcription

                                                  ª 2011 The Association for the Publication of the Journal of Internal Medicine Journal of Internal Medicine   5
S. Palmer et al.
                                 |                                                                             Symposium: HIV reservoirs and HIV infection

factors assembled at the HIV promoter causing a de-                                                  Strategies for purging the latent HIV reservoir
crease in HIV expression.
                                                                                                     The mechanisms behind HIV latency are extremely
                                                                                                     complex; and to efficiently eliminate the viral reser-
Owing to difficulties in using primary human T-cells
                                                                                                     voir, many different strategies have to be explored.
for investigation of HIV latency, most studies to date
                                                                                                     Reactivation of these latently infected cells which
have utilized infected cell lines for modelling viral la-
                                                                                                     would eliminate the HIV-infected cell either by viral
tency [65, 67, 68]. Studies based on these cell lines
                                                                                                     cytopathic effect or natural immune response in com-
have revealed many important mechanisms of HIV la-
                                                                                                     bination with antiretroviral therapy to prevent new
tency but an infected cell line may not truly reflect the
                                                                                                     infections is currently considered the best strategy,
latent state in vivo. Therefore, new ex vivo models
                                                                                                     but not the only, for eradication of HIV.
based on primary human CD4+ T-cells have been
developed by several researchers and most of these
                                                                                                     Earlier studies have shown a nonspecific activation
promising ex vivo latency models use human primary
                                                                                                     of resting memory T-cells and an increase in HIV
CD4+ T-cells infected with HIV [62, 69–72]. These
                                                                                                     replication by CD3 monoclonal antibodies or by
new models have allowed for detailed studies of the
                                                                                                     combination of cytokines TNF-a, IL-2 and IL-6 [34,
signalling pathways which can reactivate latent HIV.
                                                                                                     35]. In addition, the cytokine IL-7 has been shown
They underline the importance of NFAT, the tyrosine
                                                                                                     to reactivate HIV primary human T-cells and thy-
kinase Lck and cellular transcription factor binding
                                                                                                     mocytes in vitro and is well tolerated in HIV-infected
sites such as Sp1 and kB ⁄ NFAT for optimal reactiva-
                                                                                                     patients [76–79]. Patients infected with HIV are cur-
tion of latent HIV in memory CD4+ T-cells. In addi-
                                                                                                     rently receiving IL-7 as part of an ongoing clinical
tion, these relatively new ex vivo models revealed that
                                                                                                     trial to determine if IL-7 reduces their latent HIV
HIV was not reactivated after incubation with com-
                                                                                                     reservoir (ERAMUNE, http://www.clinicaltrials.
pounds such as the HDAC inhibitor valproic acid; the
                                                                                                     gov). The outcomes of this trial could help in the
NF-kB activating agent prostratin; PMA; and TNF-a.
                                                                                                     development of new and important therapies for
These compounds had previously been shown to
                                                                                                     HIV eradication. Upregulating cellular transcription
reactivate HIV in cell line-based models [70]. These
                                                                                                     to induce HIV gene expression has been proposed
discrepancies further stress the importance of using
                                                                                                     as another strategy for reducing latent HIV reser-
ex vivo models instead of models based on infected
                                                                                                     voirs. This includes inhibiting class I HDACs be-
cell lines. Moreover, these ex vivo models did demon-
                                                                                                     cause these HDACs promote latency by regulating
strate the importance of heterochromatic structures
                                                                                                     genome structure and transcriptional activity. A re-
owing to histone deacetylation and DNA methylation
                                                                                                     cent report has shown that synthetic class I HDAC
for promoting HIV latency.
                                                                                                     inhibitors can reactivate latent HIV in a Jurkat cell
                                                                                                     model of latency and in resting CD4+ T-cells iso-
A recent study revealed that HIV integration can hap-
                                                                                                     lated from patients better than class II HDAC inhib-
pen in both active and resting primary human T-cells
                                                                                                     itors by inducing LTR expression [67]. However, the
and that in both cell types, integration was favoured
                                                                                                     coadministration of the HDAC inhibitor, valproic
in active transcription units [73]. A modest difference
                                                                                                     acid and HAART gave mixed results [80, 81].
between resting and active cells was found and inte-
gration into actively transcribing genes was reduced
                                                                                                     Similar to multiple-drug HIV therapy which interferes
in resting cells. The finding that latency can be estab-
                                                                                                     with different stages in the viral life cycle, antilatency
lished in resting T-cells conflicts with commonly held
                                                                                                     compounds have been combined to purge and reacti-
beliefs that HIV latency is established only in acti-
                                                                                                     vate latent HIV. A recent study revealed that in
vated T-cells. Additional studies are needed to inves-
                                                                                                     patients on suppressive therapy (HIV RNA < 50 cop-
tigate the contribution of resting T-cells to persistent
                                                                                                     ies mL)1), a combination of prostratin with either
viremia.
                                                                                                     valproic acid (long approved – at low doses – for the
                                                                                                     treatment of epilepsy) or suberoylanilide hydroxamic
In addition, the role of myeloid cells in maintaining
                                                                                                     acid (SAHA) (a recently approved HDAC inhibitor for
the latent reservoir should be investigated as it is gen-
                                                                                                     clinical use as cancer therapy) was more efficient
erally assumed that HIV persists in both lymphoid
                                                                                                     than each compound alone in reactivating latent HIV
and myeloid cells [74, 75]. For example, activated
                                                                                                     in CD8 depleted PBMCs [82]. Synergistic reactivation
macrophages residing within tissues interact with
                                                                                                     of latent HIV was also achieved by combining HDAC
CD4+ T-cells via cell-to-cell contact, and via release of
                                                                                                     inhibitors and NF-kB activating agents [83]. This
inflammatory and regulatory cytokines. These inter-
                                                                                                     study demonstrated that in PBMCs and Jurkat-cell-
actions may contribute to HIV persistence.

6    ª 2011 The Association for the Publication of the Journal of Internal Medicine Journal of Internal Medicine
S. Palmer et al.
                     |                                                          Symposium: HIV reservoirs and HIV infection

based systems, a combination of SAHA and prostrat-                    HAART making this an attractive macaque model
in synergistically reactivates latent HIV. This SAHA                  for studying HIV latency [89, 90]. However, the
and prostratin combination targeted a wide range of                   macaque model has two major disadvantages:
latent integration sites, acted effectively against viral             (i) rhesus macaques are expensive as they have to
variants that have acquired mutations in their pro-                   be maintained on therapy for a long time for studies
moter regions and functioned across five different                    of viral persistence, and (ii) there are important ge-
HIV-1 subtypes. These results emphasize the possi-                    netic differences between SIV and HIV. Therefore, a
bilities of using combination antilatency therapy and                 ‘humanized’ bone marrow ⁄ liver ⁄ thymus mouse
may explain earlier disappointing results from a clini-               model populated with both human B- and T-cells
cal trial using valproic acid alone to treat patients on              that can be infected with HIV is an interesting alter-
suppressive HAART [80]. Moreover, SAHA by itself                      native animal model for researching viral reservoirs
reactivated latent HIV in cell lines and CD4+ T-cells                 and compounds that purge these reservoirs [91].
isolated from patients on suppressive HAART. SAHA
is approved by the US Food and Drug Administration
                                                                      Clinical approaches to HIV eradication
for clinical use, making it a promising candidate for
future clinical trials [84, 85]. Currently there are                  The observation that an increase in viral RNA was
large-scale screening efforts ongoing to find novel                   measured in HIV-infected individuals treated with
compounds to reactivate latent HIV using latently                     intravenous immunoglobulin (IVIGs) for Guillan-
HIV-1-infected reporter cell lines that allow for high                Barrés syndrome resulted in a study of the possible
throughput drug screening [86]. In addition, resting                  use of IVIGs to eradicate HIV [92]. The administra-
memory CD4+ T-cells from patients on suppressive                      tion of high dosage of IVIGs to patients on HAART
HIV therapy are being used to screen potential reacti-                decreased the latent reservoir, as measured by a
vating agents [67].                                                   decrease in quantifiable HIV in resting memory
                                                                      CD4+ T-cells, in about half of the patients revealing
NF-kB inducing agents such as prostratin have                         how bedside observations may point the way to
shown promise in reactivating HIV from latently in-                   new important anti-latency therapies. It is assumed
fected cells. However, NF-kB promotes global T-cell                   the mechanism for how IVIGs decrease the latent
activation running the risk of systemic inflammatory                  reservoir in HIV patients is related to the effects of
response in patients. Recently, two transcription fac-                IVIGs have on the activation, differentiation and
tors Ets-1 and VII-Ets-I have been found which reac-                  effector functions of dendritic cells, and T- and
tivate latent HIV in a NF-kb-independent manner                       B-cells [93].
[87]. In addition, a compound, 5-hydroxynaphtha-
lene-1,4-dione (5HN), has been found which reacti-                    Also of note is the interesting case of a HIV-infected
vates latent HIV without causing global T-cell activa-                patient in Germany, apparently now cured of the
tion[72]. In a related study, a protein secreted by the               disease following a stem cell transplant from a
nonpathogenic bacterium, Massilia timonae, called                     donor, homozygous for the CCR5D32 mutation,
HIV-1-reactivating factor, efficiently reactivated                    which prevents expression of this HIV-1 coreceptor.
latent HIV-1 infection by producing a strong but short                Despite receiving no antiretroviral therapy in the
burst of NF-jB activity. This NF-jB activity was suffi-               3.5 years since receiving the transplant, this pa-
cient to trigger initial HIV-1 Tat production, which                  tient has experienced no viral rebound [94]. Such
then induced HIV-1 expression [88].                                   procedures are not presently a viable solution for
                                                                      tens of millions of HIV-infected individuals world-
To further elucidate where HIV persists during ther-                  wide. Nevertheless, it is an intriguing and promis-
apy and to test compounds for the eradication of                      ing result which informs efforts to develop a cure
HIV, an appropriate animal model is necessary. Ide-                   for HIV infection.
ally, this model should show similar patterns of viral
decay and CD4+ T-cell increase during therapy as                      The CCR5D32 phenotype is being replicated in HIV-
HIV-infected humans starting HAART. The therapy                       infected individuals through the use of gene modifi-
used in the animal model should also simulate HA-                     cation. The expression of the CCR5 coreceptor is
ART. A recent study revealed that SIV-infected ma-                    eliminated from CD4+ T-cells and hematopoietic
caques treated with a combination of four antiretro-                  stem or progenitor cells (HSC) by zinc-finger nucleas-
viral drugs revealed a similar pattern of viral decay                 es (ZFNs) which permanently disrupt the CCR5
and reduced frequencies of circulating resting CD4+                   open-reading frame [95, 96]. The use of adenovirus
T-cells similar to HIV-infected humans starting                       vectors to deliver these CCR5-targeted ZFNs to pri-

                                                   ª 2011 The Association for the Publication of the Journal of Internal Medicine Journal of Internal Medicine   7
S. Palmer et al.
                                 |                                                                             Symposium: HIV reservoirs and HIV infection

mary human CD4+ T-cells resulted in HIV-resistant                                                    Conclusion
primary CD4+ T-cells that expanded stably in cul-
                                                                                                     Under the most effective current antiretroviral ther-
ture [96]. The transplantation of these cells into
                                                                                                     apy, HIV infection is well suppressed but not eradi-
immunodeficient mice and subsequent challenge
                                                                                                     cated. During combination antiretroviral therapy,
with CCR5-tropic HIV-1 resulted in a statistically
                                                                                                     reduction of HIV RNA levels to less than 50 cop-
significant reduction in plasma viral RNA in the mice
                                                                                                     ies mL)1 is frequently achieved; however, ultrasensi-
receiving the ZFN-modified T-cells compared to the
                                                                                                     tive assays detect the persistence of viremia in
mice which did not receive these cells. Additional
                                                                                                     approximately 80% of patients on suppressive
studies of immunodeficient mice transplanted with
                                                                                                     HAART for periods exceeding 7 years. The source
ZFN-modified HSC revealed that 6 weeks after infec-
                                                                                                     and dynamics of this persistent viremia are currently
tion with a CCR5-tropic HIV-1, viral loads decreased
                                                                                                     under investigation. The important conclusion that
and their transplanted human CD4+ T-cells in-
                                                                                                     emerged from this work is that antiviral strategies
creased to preinfection levels. In fact, HIV-1 could
                                                                                                     alone will never be adequate to eliminate HIV infec-
not be detected by PCR quantification 10–12 weeks
                                                                                                     tion; to accomplish this goal, we must address HIV
postinfection at necropsy. In addition, the levels of
                                                                                                     latency and its implications. A well-defined latent
CCR5-negative human cells had expanded indicat-
                                                                                                     reservoir of HIV is resting memory CD4+ T-cells,
ing a selective advantage for these cells [95]. These
                                                                                                     which might be infected directly at low frequency, or
CCR5-modified T-cells remain susceptible to
                                                                                                     arise when an activated CD4+ T-cell becomes
CXCR4-tropic strains of HIV-1 and, therefore, phe-
                                                                                                     infected by HIV, but transitions to a terminally differ-
nocopy the HIV-1 resistance in patients with the nat-
                                                                                                     entiated memory T-cell before HIV infection elimi-
urally occurring CCR5D32 allele. Based on the
                                                                                                     nates the cell. However, other cell types such as
positive results from these preclinical studies, the
                                                                                                     hematopoietic stem cells or cells of the mono-
therapeutic value of CCR5-knockout T-cells is being
                                                                                                     cyte ⁄ macrophage linage may also contribute to the
evaluated in a phase 1 clinical trial. Three different
                                                                                                     latent reservoir. A number of mechanisms, such as
patient cohorts are being enrolled in this trial:
                                                                                                     the chromatin environment, the viral integration
patients infected with multidrug resistant virus; pa-
                                                                                                     site, down-regulated transcription factors, impaired
tients with controlled viral infection who have normal
                                                                                                     activity of Tat and suppression of viral mRNA leading
CD4+ T-cell levels; and patients infected with drug-
                                                                                                     to reduced viral protein translation, have been
sensitive virus but who have low CD4+ T-cell levels
                                                                                                     shown to play a role in maintaining HIV latency.
(ClinicalTrials.govNCT00842634). CD4+ T-cells
                                                                                                     Whilst further research is needed, recent studies
from these patients will be exposed to CCR5-targeted
                                                                                                     have introduced new insights into the mechanisms
ZFNs ex vivo. The resulting CCR5-knockout cells will
                                                                                                     of latency and persistent HIV reservoirs. Consider-
be expanded for 10 days and then transferred to the
                                                                                                     able effort is now underway to apply the findings of
patient. The overall aim of this trial is to replicate the
                                                                                                     these mechanistic studies to the clinical problem.
CCR5D32 phenotype in HIV-infected individuals and
                                                                                                     The problem of HIV remission and ⁄ or eradication
it will be completed in 2011.
                                                                                                     from an infected patient is an extremely difficult one,
                                                                                                     but well worth the effort.
Obviously, caution in approaching therapies that
meddle with control of immune activation is war-
ranted. The very nature of the virus-host relation-                                                  Conflict of interest statement
ship means that seeking to purge the latent HIV res-
                                                                                                     No conflict of interest was declared.
ervoir runs the risk of triggering a systemic patient
immune reaction. In addition, any future clinical ap-
proaches to eradicating latent HIV reservoirs must                                                   References
avoid the generation of new HIV infection resulting                                                   1 Havlir DV et al. Prevalence and predictive value of intermittent
from the activation of these latently infected cells. In                                                viremia with combination HIV therapy. JAMA 2001; 286: 171–9.
carrying out initial exploration of these methods,                                                    2 Havlir DV et al. Predictors of residual viremia in HIV-infected pa-
researchers will need to integrate appropriate animal                                                   tients successfully treated with efavirenz and lamivudine plus
modelling first to avoid such risks arising in hu-                                                      either tenofovir or stavudine. J Infect Dis 2005; 191: 1164–8.
                                                                                                      3 Maldarelli F et al. ART suppresses plasma HIV-1 RNA to a stable
mans. Even then, experience with potent immune
                                                                                                        set point predicted by pretherapy viremia. PLoS Pathog 2007; 3:
activators in other fields has shown that animal                                                        e46.
models may not successfully predict the human                                                         4 Palmer S et al. New real-time reverse transcriptase-initiated PCR
reaction to therapies that can affect the immune                                                        assay with single-copy sensitivity for human immunodeficiency
response at a basic level.                                                                              virus type 1 RNA in plasma. J Clin Microbiol 2003; 41: 4531–6.

8    ª 2011 The Association for the Publication of the Journal of Internal Medicine Journal of Internal Medicine
S. Palmer et al.
                           |                                                                  Symposium: HIV reservoirs and HIV infection

 5 Palmer S et al. Low-level viremia persists for at least 7 years in pa-          23 Wei X et al. Viral dynamics in human immunodeficiency virus
   tients on suppressive antiretroviral therapy. Proc Natl Acad Sci U                 type 1 infection. Nature 1995; 373: 117–22.
   S A 2008; 105: 3879–84.                                                         24 Ho DD, Rota TR, Hirsch MS. Infection of monocyte ⁄ macrophages
 6 Davey Jr RT et al. HIV-1 and T cell dynamics after interruption of                 by human T-lymphotrophic virus type III. J Clin Invest 1986; 77:
   highly active antiretroviral therapy (HAART) in patients with a                    1712–5.
   history of sustained viral suppression. Proc Natl Acad Sci U S A                25 Nicholson JK et al. In vitro infection of human monocytes
   1999; 96: 15109–14.                                                                with human T lymphotropic virus type III ⁄ lymphadenop-
 7 Dornadula G et al. Residual HIV-1 RNA in blood plasma of                           athyassociated virus (HTLV-III ⁄ LAV). J Immunol 1986; 137:
   patients taking suppressive highly active antiretroviral therapy.                  323–9.
   JAMA 1999; 282: 1627–32.                                                        26 Popov S et al. Long-term productive human immunodeficiency
 8 Lewin SR et al. Use of real-time PCR and molecular beacons to de-                  virus infection of CD1a-sorted myeloid dendritic cells. J Virol
   tect virus replication in human immunodeficiency virus type 1-                     2005; 79: 602–8.
   infected individuals on prolonged effective antiretroviral therapy.             27 Kalter DC, Gendelman HE, Meltzer MS. Monocytes, dendritic
   J Virol 1999; 73: 6099–103.                                                        cells, and Langerhans cells in human immunodeficiency virus
 9 Zhang L et al. Quantifying residual HIV-1 replication in patients                  infection. Dermatol Clin 1991; 9: 415–28.
   receiving combination antiretroviral therapy. N Engl J Med 1999;                28 Kalter DC et al. Epidermal Langerhans cells are not principal
   340: 1605–13.                                                                      reservoirs of virus in HIV disease. J Immunol 1991; 146: 3396–
10 Coffin JM. HIV population dynamics in vivo: implications for ge-                   404.
   netic variation, pathogenesis, and therapy. Science 1995; 267:                  29 Pugh CW, MacPherson GG, Steer HW. Characterization of non-
   483–9.                                                                             lymphoid cells derived from rat peripheral lymph. J Exp Med
11 Perelson AS et al. Decay characteristics of HIV-1-infected                         1983; 157: 1758–79.
   compartments during combination therapy. Nature 1997; 387:                      30 Otero M et al. Peripheral blood dendritic cells are not a major res-
   188–91.                                                                            ervoir for HIV type 1 in infected individuals on virally suppressive
12 Havlir DV et al. Productive infection maintains a dynamic steady                   HAART. AIDS Res Hum Retroviruses 2003; 19: 1097–103.
   state of residual viremia in human immunodeficiency virus type                  31 Smith BA et al. Persistence of infectious HIV on follicular den-
   1-infected persons treated with suppressive antiretroviral ther-                   dritic cells. J Immunol 2001; 166: 690–6.
   apy for five years. J Virol 2003; 77: 11212–9.                                  32 Murray JM et al. Antiretroviral therapy with the integrase inhibi-
13 Ramratnam B et al. Intensification of antiretroviral therapy                       tor raltegravir alters decay kinetics of HIV, significantly reducing
   accelerates the decay of the HIV-1 latent reservoir and decreases,                 the second phase. AIDS 2007; 21: 2315–21.
   but does not eliminate, ongoing virus replication. J Acquir                     33 Spivak AM et al. Dynamic constraints on the second phase com-
   Immune Defic Syndr 2004; 35: 33–7.                                                 partment of HIV-infected cells. AIDS Res Hum Retroviruses 2011;
14 Dinoso JB et al. Treatment intensification does not reduce                         27: 759–61.
   residual HIV-1 viremia in patients on highly active antiretroviral              34 Chun TW et al. Induction of HIV-1 replication in latently infected
   therapy. Proc Natl Acad Sci U S A 2009; 106: 9403–8.                               CD4+ T cells using a combination of cytokines. J Exp Med 1998;
15 Gandhi RT et al. The effect of raltegravir intensification on                      188: 83–91.
   low-level residual viremia in HIV-infected patients on antiretro-               35 Prins JM et al. Immuno-activation with anti-CD3 and recombi-
   viral therapy: a randomized controlled trial. PLoS Med 2010; 7:                    nant human IL-2 in HIV-1-infected patients on potent antiretro-
   1–11.                                                                              viral therapy. AIDS 1999; 13: 2405–10.
16 Hatano H et al. A randomized, controlled trial of raltegravir inten-            36 Siliciano JD et al. Long-term follow-up studies confirm the stabil-
   sification in antiretroviral-treated, HIV-infected patients with a                 ity of the latent reservoir for HIV-1 in resting CD4+ T cells. Nat
   suboptimal CD4+ T cell response. J Infect Dis 2011; 203: 960–8.                    Med 2003; 9: 727–8.
17 McMahon D et al. Short-course raltegravir intensification does                  37 Chun TW et al. Early establishment of a pool of latently infected,
   not reduce persistent low-level viremia in patients with HIV-1                     resting CD4(+) T cells during primary HIV-1 infection. Proc Natl
   suppression during receipt of combination antiretroviral ther-                     Acad Sci U S A 1998; 95: 8869–73.
   apy. Clin Infect Dis 2010; 50: 912–9.                                           38 Chun TW et al. Presence of an inducible HIV-1 latent reservoir
18 Bailey JR et al. Residual human immunodeficiency virus type 1                      during highly active antiretroviral therapy. Proc Natl Acad Sci U S
   viremia in some patients on antiretroviral therapy is dominated                    A 1997; 94: 13193–7.
   by a small number of invariant clones rarely found in circulating               39 Chomont N et al. HIV reservoir size and persistence are driven by
   CD4+ T cells. J Virol 2006; 80: 6441–57.                                           T cell survival and homeostatic proliferation. Nat Med 2009; 15:
19 Joos B et al. HIV rebounds from latently infected cells, rather                    893–900.
   than from continuing low-level replication. Proc Natl Acad Sci U S              40 Brennan TP et al. Analysis of human immunodeficiency virus
   A 2008; 105: 16725–30.                                                             type 1 viremia and provirus in resting CD4+ T cells reveals a novel
20 Buzon MJ et al. HIV-1 replication and immune dynamics are af-                      source of residual viremia in patients on antiretroviral therapy.
   fected by raltegravir intensification of HAART-suppressed sub-                     J Virol 2009; 83: 8470–81.
   jects. Nat Med 2010; 16: 460–5.                                                 41 Gunthard HF et al. Residual human immunodeficiency virus
21 Yukl SA et al. Effect of raltegravir-containing intensification on                 (HIV) Type 1 RNA and DNA in lymph nodes and HIV RNA in genital
   HIV burden and T-cell activation in multiple gut sites of HIV-posi-                secretions and in cerebrospinal fluid after suppression of viremia
   tive adults on suppressive antiretroviral therapy. AIDS 2010; 24:                  for 2 years. J Infect Dis 2001; 183: 1318–27.
   2451–60.                                                                        42 Pantaleo G, Graziosi C, Fauci AS. New concepts in the immuno-
22 Ho DD et al. Rapid turnover of plasma virions and CD4 lympho-                      pathogenesis of human immunodeficiency virus infection. N Engl
   cytes in HIV-1 infection. Nature 1995; 373: 123–6.                                 J Med 1993; 328: 327–35.

                                                                 ª 2011 The Association for the Publication of the Journal of Internal Medicine Journal of Internal Medicine   9
S. Palmer et al.
                                 |                                                                             Symposium: HIV reservoirs and HIV infection

43 Veazey RS et al. Gastrointestinal tract as a major site of CD4+ T                                64 Han Y et al. Resting CD4+ T cells from human immunodeficiency
   cell depletion and viral replication in SIV infection. Science 1998;                                virus type 1 (HIV-1)-infected individuals carry integrated HIV-1
   280: 427–31.                                                                                        genomes within actively transcribed host genes. J Virol 2004; 78:
44 Veazey RS, Lackner AA. The gastrointestinal tract and the patho-                                    6122–33.
   genesis of AIDS. AIDS 1998; 12(Suppl A): S35–42.                                                 65 Jordan A, Bisgrove D, Verdin E. HIV reproducibly establishes a
45 Guadalupe M et al. Viral suppression and immune restoration in                                      latent infection after acute infection of T cells in vitro. EMBO J
   the gastrointestinal mucosa of human immunodeficiency virus                                         2003; 22: 1868–77.
   type 1-infected patients initiating therapy during primary or                                    66 Lenasi T, Contreras X, Peterlin BM. Transcriptional interference
   chronic infection. J Virol 2006; 80: 8236–47.                                                       antagonizes proviral gene expression to promote HIV latency. Cell
46 Gisslen M et al. Cerebrospinal fluid viral load, intrathecal immu-                                  Host Microbe 2008; 4: 123–33.
   noactivation, and cerebrospinal fluid monocytic cell count in                                    67 Archin NM et al. Expression of latent human immunodeficiency
   HIV-1 infection. J Acquir Immune Defic Syndr 1999; 21: 271–6.                                       type 1 is induced by novel and selective histone deacetylase
47 Hagberg L et al. Kinetics of HIV-1 in cerebrospinal fluid and ser-                                  inhibitors. AIDS 2009; 23: 1799–806.
   um after zidovudine treatment. J Neuro AIDS 1999; 2: 29–35.                                      68 Pomerantz RJ et al. Cells nonproductively infected with HIV-1 ex-
48 Kaul M. HIV-1 associated dementia: update on pathological                                           hibit an aberrant pattern of viral RNA expression: a molecular
   mechanisms and therapeutic approaches. Curr Opin Neurol                                             model for latency. Cell 1990; 61: 1271–6.
   2009; 22: 315–20.                                                                                69 Blazkova J et al. CpG methylation controls reactivation of HIV
49 Strain MC et al. Effect of treatment, during primary infection, on                                  from latency. PLoS Pathog 2009; 5: e1000554.
   establishment and clearance of cellular reservoirs of HIV-1.                                     70 Bosque A, Planelles V. Induction of HIV-1 latency and reactiva-
   J Infect Dis 2005; 191: 1410–8.                                                                     tion in primary memory CD4+ T cells. Blood 2009; 113: 58–65.
50 Wong JK et al. In vivo compartmentalization of human immuno-                                     71 Tyagi M, Pearson RJ, Karn J. Establishment of HIV latency in pri-
   deficiency virus: evidence from the examination of pol sequences                                    mary CD4+ cells is due to epigenetic transcriptional silencing
   from autopsy tissues. J Virol 1997; 71: 2059–71.                                                    and P-TEFb restriction. J Virol 2010; 84: 6425–37.
51 Aquaro S et al. HIV-1-associated dementia during HAART                                           72 Yang HC et al. Small-molecule screening using a human primary
   therapy. Recent Pat CNS Drug Discov 2008; 3: 23–33.                                                 cell model of HIV latency identifies compounds that reverse la-
52 Price RW, Spudich S. Antiretroviral therapy and central nervous                                     tency without cellular activation. J Clin Invest 2009; 119: 3473–
   system HIV type 1 infection. J Infect Dis 2008; 197(Suppl 3):                                       86.
   S294–306.                                                                                        73 Brady T et al. HIV integration site distributions in resting and
53 Varatharajan L, Thomas SA. The transport of anti-HIV drugs                                          activated CD4+ T cells infected in culture. AIDS 2009; 23: 1461–
   across blood-CNS interfaces: summary of current knowledge                                           71.
   and recommendations for further research. Antiviral Res 2009;                                    74 Crowe S, Zhu T, Muller WA. The contribution of monocyte
   84: A99–109.                                                                                        infection and trafficking to viral persistence, and maintenance of
54 Yilmaz A et al. Treatment intensification has no effect on the                                      the viral reservoir in HIV infection. J Leukoc Biol 2003; 74: 635–
   HIV-1 central nervous system infection in patients on suppres-                                      41.
   sive antiretroviral therapy. J Acquir Immune Defic Syndr 2010;                                   75 Crowe SM, Sonza S. HIV-1 can be recovered from a variety of cells
   55: 590–6.                                                                                          including peripheral blood monocytes of patients receiving highly
55 Kiessling AA et al. Human immunodeficiency virus in semen                                           active antiretroviral therapy: a further obstacle to eradication.
   arises from a genetically distinct virus reservoir. AIDS Res Hum                                    J Leukoc Biol 2000; 68: 345–50.
   Retroviruses 1998; 14(Suppl 1): S33–41.                                                          76 Levy Y et al. Enhanced T cell recovery in HIV-1-infected adults
56 Vernazza PL et al. Detection and biologic characterization of                                       through IL-7 treatment. J Clin Invest 2009; 119: 997–1007.
   infectious HIV-1 in semen of seropositive men. AIDS 1994; 8:                                     77 Scripture-Adams DD et al. Interleukin-7 induces expression of
   1325–9.                                                                                             latent human immunodeficiency virus type 1 with minimal ef-
57 Zhang H et al. Human immunodeficiency virus type 1 in the se-                                       fects on T-cell phenotype. J Virol 2002; 76: 13077–82.
   men of men receiving highly active antiretroviral therapy. N Engl                                78 Sereti I et al. IL-7 administration drives T cell-cycle entry and
   J Med 1998; 339: 1803–9.                                                                            expansion in HIV-1 infection. Blood 2009; 113: 6304–14.
58 Bruggeman LA et al. Renal epithelium is a previously unrecog-                                    79 Wang FX et al. IL-7 is a potent and proviral strain-specific inducer
   nized site of HIV-1 infection. J Am Soc Nephrol 2000; 11: 2079–                                     of latent HIV-1 cellular reservoirs of infected individuals on virally
   87.                                                                                                 suppressive HAART. J Clin Invest 2005; 115: 128–37.
59 Chun TW et al. In vivo fate of HIV-1-infected T cells: quantitative                              80 Lehrman G et al. Depletion of latent HIV-1 infection in vivo: a
   analysis of the transition to stable latency. Nat Med 1995; 1:                                      proof-of-concept study. Lancet 2005; 366: 549–55.
   1284–90.                                                                                         81 Siliciano JD et al. Stability of the latent reservoir for HIV-1 in
60 Bednarik DP, Mosca JD, Raj NB. Methylation as a modulator of                                        patients receiving valproic acid. J Infect Dis 2007; 195: 833–6.
   expression of human immunodeficiency virus. J Virol 1987; 61:                                    82 Reuse S et al. Synergistic activation of HIV-1 expression by
   1253–7.                                                                                             deacetylase inhibitors and prostratin: implications for treatment
61 Ylisastigui L et al. Coaxing HIV-1 from resting CD4 T cells: his-                                   of latent infection. PLoS ONE 2009; 4: e6093.
   tone deacetylase inhibition allows latent viral expression. AIDS                                 83 Burnett JC et al. Combinatorial latency reactivation for HIV-1
   2004; 18: 1101–8.                                                                                   subtypes and variants. J Virol 2010; 84: 5958–74.
62 Kauder SE et al. Epigenetic regulation of HIV-1 latency by cyto-                                 84 Archin NM et al. Expression of latent HIV induced by the potent
   sine methylation. PLoS Pathog 2009; 5: e1000495.                                                    HDAC inhibitor suberoylanilide hydroxamic acid. AIDS Res Hum
63 Dahl V, Josefsson L, Palmer S. HIV reservoirs, latency, and                                         Retroviruses 2009; 25: 207–12.
   reactivation: prospects for eradication. Antiviral Res 2010; 85:                                 85 Contreras X et al. Suberoylanilide hydroxamic acid reactivates
   286–94.                                                                                             HIV from latently infected cells. J Biol Chem 2009; 284: 6782–9.

10    ª 2011 The Association for the Publication of the Journal of Internal Medicine Journal of Internal Medicine
S. Palmer et al.
                          |                                                                 Symposium: HIV reservoirs and HIV infection

86 Jones J et al. High throughput drug screening for human immu-                93 Negi VS et al. Intravenous immunoglobulin: an update on the
   nodeficiency virus type 1 reactivating compounds. Assay Drug                    clinical use and mechanisms of action. J Clin Immunol 2007; 27:
   Dev Technol 2007; 5: 181–9.                                                     233–45.
87 Yang HC et al. Isolation of a cellular factor that can reactivate            94 Hutter G et al. Long-term control of HIV by CCR5 Delta32 ⁄
   latent HIV-1 without T cell activation. Proc Natl Acad Sci U S A                Delta32 stem-cell transplantation. N Engl J Med 2009; 360:
   2009; 106: 6321–6.                                                              692–8.
88 Wolschendorf F et al. Hit-and-run stimulation: a novel concept to            95 Holt N et al. Human hematopoietic stem ⁄ progenitor cells modi-
   reactivate latent HIV-1 infection without cytokine gene induc-                  fied by zinc-finger nucleases targeted to CCR5 control HIV-1 in
   tion. J Virol 2010; 84: 8712–20.                                                vivo. Nat Biotechnol 2010; 28: 839–47.
89 Dinoso JB et al. A simian immunodeficiency virus-infected maca-              96 Perez EE et al. Establishment of HIV-1 resistance in CD4+ T cells
   que model to study viral reservoirs that persist during highly                  by genome editing using zinc-finger nucleases. Nat Biotechnol
   active antiretroviral therapy. J Virol 2009; 83: 9247–57.                       2008; 26: 808–16.
90 North TW et al. Viral sanctuaries during highly active antiretrovi-
   ral therapy in a nonhuman primate model for AIDS. J Virol 2010;               Correspondence: Sarah Palmer, Department of Virology, Swedish
   84: 2913–22.                                                                  Institute for Infectious Disease Control, Nobels väg 18, 171 82 Solna,
91 Sun Z et al. Intrarectal transmission, systemic infection, and                Sweden.
   CD4+ T cell depletion in humanized mice infected with HIV-1.                  (fax: 46-08-33 7272; e-mail: Sarah.Palmer@smi.se).
   J Exp Med 2007; 204: 705–14.
92 Lindkvist A et al. Reduction of the HIV-1 reservoir in resting CD4+
   T-lymphocytes by high dosage intravenous immunoglobulin
   treatment: a proof-of-concept study. AIDS Res Ther 2009; 6: 15.

                                                             ª 2011 The Association for the Publication of the Journal of Internal Medicine Journal of Internal Medicine   11
You can also read