You are on page 1of 13

REVIEWS

Paediatric HIV infection: the potential


for cure
Philip J. Goulder1, Sharon R. Lewin2,3 and Ellen M. Leitman1
Abstract | Recent anecdotal reports of HIV-infected children who received early antiretroviral
therapy (ART) and showed sustained control of viral replication even after ART discontinuation
have raised the question of whether there is greater intrinsic potential for HIV remission, or even
eradication (‘cure’), in paediatric infection than in adult infection. This Review describes the
influence of early initiation of ART, of immune ontogeny and of maternal factors on the potential
for HIV cure in children and discusses the unique immunotherapeutic opportunities and
obstacles that paediatric infection may present.

Antiretroviral therapy
The pattern of immune ontogeny in humans has maintenance of normal CD4+ T cell counts in the absence
(ART). The combination of evolved to meet the changing and distinct challenges of ART, and long-term survivors, who typically have low
usually a minimum of three to the immune system that arise in utero and in early CD4+ T cell counts and high levels of immune activation7,
anti-HIV drugs that aim to life, through childhood and into adolescence and but may be asymptomatic (presenting late) and, in the
suppress viral replication, slow
adulthood. The differences that are observed in terms case of children, are usually stunted9,10. Non-progressing
disease progression and
minimize the risk of of disease outcome for a range of infectious diseases, adult and paedi­atric infection are both charac­terized by
transmission. Pre-exposure according to the age at which the particular infections low levels of immune activation but through distinct
prophylactic ART has also been arise, illustrate the impact of the distinct phases of mechanisms. In non-progressing adult infection, this is
shown to protect against HIV immune system development on disease susceptibility. achieved through HLA-mediated suppression of viral
infection.
HIV infection is a case in point (FIG. 1). In the absence of replication. In non-progressing paediatric infection, the
CD4+ T cell counts antiretroviral therapy (ART), >50% of HIV-infected chil- mechanism by which immune activation remains low
The number of T cells dren die by 2 years of age1, whereas the median survival in some patients despite persistent high levels of virae-
expressing the CD4 receptor time is 11 years for ART-naive HIV-positive adults2. In mia7 is unknown. The phenotype of normal CD4+ T cell
on their surface in a microlitre
the small subset of ART-naive individuals infected counts and low levels of immune activation, despite high
of blood. These white blood
cells ‘help’ the immune system with HIV who maintain normal CD4+ T cell counts and viral loads, is reminiscent of the natural hosts of sim-
to mount a response against immune function and, therefore, do not progress to dis- ian immuno­deficiency virus (SIV), such as the sooty
pathogens and are the main ease, the mechanisms operating in children and adults mangabey and African green monkey, which live normal
target cells of HIV infection. are markedly different. Non-progressing adult infection, lifespans with normal CD4+ T cell counts and low l­evels
in so‑called elite ­controllers, is typically characterized by of immune activation but have median viral loads of ~105
a strong HIV-specific CD8+ T cell response restricted SIV RNA copies per ml (REF. 11).
by ‘protective’ MHC class I molecules (for example, The differences that exist between adult and paediat-
HLA‑B27 and HLA‑B57)3–5, such that high CD4+ T cell ric HIV infection, not only in terms of disease outcome
1
Department of Paediatrics, counts are maintained as a consequence of successful and immune response to HIV but also in terms of mode
University of Oxford, Oxford suppression of viraemia beneath the levels of detec- of transmission, have provided opportunities to improve
OX1 3SY, UK. tion (normally <50 HIV RNA copies per ml). By con- our understanding of the mechanisms of immune con-
2
The Peter Doherty Institute
for Infection and Immunity,
trast, non-­progressing HIV-infected children (who are trol of HIV. The possibility that these differences might
University of Melbourne, defined here as healthy, ART-naive children aged more also provide opportunities to learn about mechanisms
Melbourne 3000, Australia. than 5 years who maintain absolute CD4+ T cell counts of HIV eradication or ‘cure’ was initi­ally prompted
3
Department of Infectious in the normal range for age-matched uninfected children by the report of the ‘Mississippi child’ (REFS  12,13)
Diseases, Alfred Hospital
(above 750 cells per mm3 of blood))6 maintain normal (BOX 1). Although virus levels ultimately rebounded
and Monash University,
Melbourne 3004, Australia. CD4+ T cell counts despite median viral loads of ~30,000 in the Mississippi child 28 months after ART discon-
Correspondence to P.J.G.
copies per ml of plasma7, and the HLA‑B alleles that tinuation, the case was sufficiently unusual to prompt
philip.goulder@ strongly influence disease outcome in adult infection do the hypothesis that the early initiation of ART during
paediatrics.ox.ac.uk not have a significant role in these children8. It is impor- paediatric infection might provide the setting in which
doi:10.1038/nri.2016.19 tant to clarify the distinction between non-­progressing HIV ­eradication, or remission, could most readily
Published online 14 Mar 2016 adult and paedi­atric infection, which is defined by the be achieved.

NATURE REVIEWS | IMMUNOLOGY ADVANCE ONLINE PUBLICATION | 1


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Declining CD4+ T cell count; high levels of immune activation

Typical (progressing) adult infection Typical (progressing) paediatric infection


107

Viral load (copies per ml)

Viral load (copies per ml)


106
Viral set point
Viral set point 105
30,000

Limit of detection
50
Elite controllers
A rare subset (less than 1%) 6w 10 y 11 y 1y 2y 5y 10 y
of antiretroviral therapy Viral set point AIDS Death AIDS Death Viral set point
(ART)-naive, HIV-infected reached (median) (median) (median) (median) reached
individuals who have Time since infection Time since infection
undetectable plasma viral load
(by standard assays) and
remain clinically healthy in the
long term. HIV controllers is Stable, normal CD4+ T cell counts; low levels of immune activation
sometimes used synonymously
with elite controllers. Non-progressing adult infection Non-progressing paediatric infection
107
VISCONTI cohort
Viral load (copies per ml)

Viral load (copies per ml)


(Viro-Immunologic Sustained
106
Control after Treatment
Interruption cohort). A French
cohort of 14 adults in whom 30,000 30,000
antiretroviral therapy (ART) was
initiated during primary HIV
infection and interrupted after
a median of 36.5 months.
For a median of 7.4 years after Limit of detection
treatment interruption, the 50
viral load remained at less than
400 copies per ml; these 14 6w 10 y 5y 10 y
individuals are therefore
referred to as post-treatment
Time since infection Time since infection
controllers.
Figure 1 | Changes in viral load following HIV infection in paediatric and adult cases. Typical, progressing adult and
paediatric infection (upper panels), and non-progressing adult and paediatric infection (lower Nature Reviews
panels) 2,7,8,161 | Immunology
. Typically, in
CHER study
(Children with HIV Early HIV infection of antiretroviral therapy (ART)-naive adults, a viral set point is reached after 6 weeks, and the median time to
Antiretroviral Therapy study). AIDS is 10 years. In typical, ART-naive paediatric infection with HIV, a viral set point is reached after 5 years, and the
A randomized Phase III study median time to AIDS is 1 year. Non-progressing adult infection is characterized by low or undetectable viral loads and low
that enrolled 6–12‑week-old levels of immune activation. The prevalence of non-progressing paediatric infection is 5–10%7,162,163 and of adult elite
HIV-positive infants in South controllers is ~0.3%146. Non-progressing paediatric infection also features low levels of immune activation but in the
Africa with a percentage of setting of high viral loads (~30,000 copies per ml).
CD4+ lymphocytes of more
than 25%, who were assigned
either to receive immediate
A further anecdotal example of post-treatment con- a median age of 7 weeks19, and in the PEHSS study20, in
antiretroviral therapy (ART) for
40 or 96 weeks or to defer trol of viraemia was recently described in a French HIV- which 19 infants received 12 months of ART from a
treatment until clinical criteria infected child (BOX 1). Although anecdotal cases can be median age of 4 weeks, all patients rebounded soon after
were met. misleading (BOX 2), this paediatric example follows the ART discontinuation (M. F. Cotton, personal communi-
description of post-treatment control in the VISCONTI cation; P.J.G., unpublished observations). In summary, it
PEHSS study
cohort of adults who received ART during acute infection is not currently known whether early initiation of ART
(Paediatric Early HAART
and Strategic Treatment (within months, not days, of infection) and who discon- in paediatric infection is more or less likely to facilitate
Interruption study). A tinued ART after a median of 3 years14. It is estimated post-treatment control than early initiation of ART in
feasibility study of 63 that 5–15% of adults who initiate ART in early infection adult infection.
perinatally infected infants in
and continue for this duration become post-­treatment This Review considers the probable impact of early
KwaZulu-Natal, South Africa,
who were randomized to controllers, but the equivalent figure in paediatric initiation of ART on the potential for HIV eradication
receive either immediate infection following early initiation of ART is unknown. in children versus adults and discusses the influence
12‑month uninterrupted Several reports have described viraemic rebound within of immune ontogeny and maternal transmission in
antiretroviral therapy (ART), days of ART discontinuation in children in whom ART paedi­atric infection. Finally, we consider the particular
immediate 18‑month
treatment with structured
was initiated in the first week of life and continued for opportunities that these features of paediatric infection
interruptions or deferred 2–4 years15–18. In the much larger CHER study, in which present in terms of strategies to promote HIV remission
treatment. 250 children were treated for either 40 or 96 weeks from or eradication.

2 | ADVANCE ONLINE PUBLICATION www.nature.com/nri


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Viral reservoir Early ART in paediatric versus adult infection very rapid seeding of the viral reservoir, within 3 days of
Virus that persists in patients Feasibility. In adult infection, outside of expensive trial infection26. On the other hand, in the VISCONTI cohort
on antiretroviral therapy (ART) settings21,22, the identification of early HIV infection is subjects, ART was initiated within the first 2–3 months
in the form of latent or problematic. By contrast, in utero paediatric infection, of infection — which is categorized on the basis of
productively infected cells.
Latency is established in
which mainly occurs late in the third trimester 23,24, can Fiebig stages  ­I–VI27 — ­similarly as in the VISCONTI
long-lived resting memory be diagnosed using point‑of‑care testing 25 within hours teenager 28, which indicates that ART initi­ation after the
T cells as integrated virus that of birth in the children of chronically HIV-infected first few days of infection does not preclude subsequent
is replication competent but mothers, and ART can be initiated in these children post-treatment control in HIV infection.
transcriptionally silent.
within minutes of birth, pending the results of diagnostic
Latently infected cells persist
for decades and are more
tests. In most cases, therefore, treatment can be started Reduction of the viral reservoir. It is evident that early
frequent in tissue compared earlier in paediatric infection than in adult infection. initi­ation of ART reduces the frequency of latently
with blood. Reactivation of However, in some cases — for example, when the mother infected T cells in both adult and paediatric infection. If
virus in these cells (for develops acute infection during pregnancy 16 — in utero ART is initiated during chronic infection in adults, the
example, upon ART
interruption) is the major
transmission can occur several weeks before birth latently infected resting CD4+ T cells persist with a half-
obstacle to HIV eradication. (FIG. 2). In addition, paediatric infection can arise from life of 44 months29–31. However, treatment of adults during
intra-­partum or post-partum mother-to‑child trans- acute infection results in an approximately 10–100‑fold
Fiebig stages I–VI mission (MTCT), and in neither case are these infec- lower frequency of latently infected resting CD4+ T cells
Categorization of primary HIV
tions detectable at birth. Furthermore, there are added compared with chronic infection, when measured in
infection in six stages based on
the detection of different HIV
challenges to detecting intra-partum or post-­partum terms of either HIV DNA or replication-competent
markers. Staging begins with infection for the practical reason that the mother and virus32–34. Similar results have been demonstrated in SIV-
viral detection by PCR (stage I), child have long departed the hospital. Therefore, initi­ infected macaques when treatment is initiated within
then ELISA detection of Gag ating ART soon after infection following intra-partum 3 days compared with 7 days after infection26. In addition,
p24 (stage II) or HIV-specific
antibody (stage III), and then
or post-partum transmission of HIV may be substan- the amount of cell-associated unspliced HIV RNA, which
sequential stages of tially more difficult than after in utero transmission that reflects basal levels of HIV transcription, is significantly
HIV-specific antibody detection mostly arises late in the third trimester and is detectable lower in individuals who are treated in acute infection
by western blot (stages IV–VI). at birth. However, it is not known whether the timing compared with chronic infection35. Much of the decrease
of early initiation of ART during infection is crucial to in HIV DNA and unspliced HIV RNA levels occurs in
the outcome in terms of post-treatment control. On the the first few years of therapy 36,37. Latently infected CD4+
one hand, SIV–macaque studies indicate that there is a T cell subsets have different decay rates, and following
long-term ART (more than 10 years), long-lived central
memory T cells, memory stem cells and naive T cells are
Box 1 | Case studies of the Mississippi child and the VISCONTI child the main T cell subsets enriched with HIV36,38,39.
In infants who received ART at a median age of
The Mississippi child 8 weeks, the half-life of latently infected resting CD4+
The ‘Mississippi child’ (REFS 12,13) was infected with HIV in utero, and antiretroviral
T cells was 11 months during the first 2 years on ART40.
therapy (ART) was initiated at 30 hours of age and continued for the first 18 months of life;
Levels of both cell-associated HIV DNA and cell-­
viral load remained undetectable for more than 2 years following ART discontinuation.
Other unusual aspects of this case were: associated HIV RNA are lower in children in whom ART
was initiated earlier after infection17,41–44. In the CHER
• The healthy CD4+ T cell count (644 cells per mm3) and low viral load (2,423 copies per ml)
of the ART-naive mother (the median viral load of in utero-transmitting HIV-infected study, for example, in which levels of both HIV DNA and
mothers is ~90,000 copies per ml (REF.143)). cell-­associated HIV RNA were measured, initiation of
• The low viral load (19,812 copies per ml at 30 hours post-delivery) of the child (the ART in children before 2 months of age (at a median age
median viral load of in utero-infected infants is ~150,000 copies per ml on the first day of 53 days) resulted in a lower frequency of HIV-infected
of life)143. CD4+ T cells 7–8 years later, compared with children in
• The high percentage (69%) of lymphocytes that are CD4+ T cells 8 days post-delivery whom ART was started at more than 2 months of age
(in HIV-uninfected infants, the median percentage of CD4+ T cells is 52%)6,143. (at a median age of 170 days)43. Furthermore, whereas
• Viral rebound in the absence of HIV-specific immune responses in the child to a steady- the levels of cell-associated HIV DNA seem to plateau
state value of only 16,750 copies per ml, which is similar to the pre-ART viral load. after ~4 years in adult infection45, in children these levels
continue to decline for more than a decade41. Treatment
The VISCONTI child
of HIV infection in children at a median age of 2 months
The ‘VISCONTI child’ (REF. 28) received ART for the first 6 years of life, probably following resulted in a significant decrease in HIV DNA, but the
intra-partum infection (HIV DNA and HIV RNA were undetectable at 3 days of age) and persistence of 2‑LTR ­circles (2‑long-­terminal repeat cir-
has remained aviraemic (less than 4 copies per ml) for 12 years so far since ART
cles) and cell-associated unspliced RNA after 96 weeks,
discontinuation.
and the frequency of HIV DNA was more than 148‑fold
Other unusual or notable aspects of this case were:
higher than the frequency of infectious genomes46, as
• ART (3–4 drugs) was initiated at 3 months of age, at which time the viral load was
previously described following treatment of HIV in
2.17 × 106 copies per ml.
adults47. Thus, ART can generally be initiated earlier
• Two separate episodes of ART non-adherence in the second year of life resulted in viral
in paediatric in utero infection than in adult infection,
rebounds to 50,000–100,000 copies per ml.
the half-life of latently infected resting CD4+ T cells
• After ART discontinuation, there were no apparent increases in viral load other than to
is shorter in children, and initial ­studies indicate that
510 copies per ml at 11 years old and 48 copies per ml at 13 years old.
decline in the viral reservoir seems to continue for longer
• Infection was with a rare clade of HIV, subtype H, the significance of which is unknown.
in children than in adults on ART.

NATURE REVIEWS | IMMUNOLOGY ADVANCE ONLINE PUBLICATION | 3


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Box 2 | Anecdotal evidence of post-treatment control: two caveats low levels of immune activation in early life through
increased IL‑6 synthesis and decreased TNF produc-
Long-term aviraemia following early ART and ART discontinuation in an HIV-infected tion51. The immune response in utero and in early
adult. The original ‘Berlin patient’ was described in 1999 (REF. 144) (not to be confused life therefore establishes a tolerogenic environment
with the more famous Berlin patient, Timothy Ray Brown, who was cured of HIV that is broadly anti-­inflammatory and that promotes
infection following a stem cell transplant from a donor homozygous for the
TH17 cell-­mediated host defence against extracellular
CC-chemokine receptor 5 (CCR5) Δ32 deletion); suppression of viraemia was achieved
after initiation of antiretroviral therapy (ART) during acute HIV infection, and for bac­terial and fungal pathogens, at the expense of a
15 years after ART discontinuation, the viral load remained at low to undetectable strong TH1 cell-driven response which, by contrast, is
levels (median viral load of 399 copies per ml)145. Only recently has it emerged that this a feature of a­ ntiviral cell-mediated i­ mmunity in adults.
subject expressed HLA‑B57. The existence of a subset of ART-naive, HIV-infected adults In the setting of HIV infection in early life, one effect
termed elite controllers (FIG. 1) was not well described in the 1990s; however, it is now of these immune adaptations is to eliminate the possi-
known that 40–50% of elite controllers express HLA‑B57 (REF. 146). Therefore, a major bility of HLA class I‑restricted CD8+ T cell-mediated
factor in immune control in the case of the original Berlin patient may have been the suppression of viraemia that is observed in adult elite
expression of HLA‑B57. controllers. However, the tolerogenic environment
Long-term aviraemia following early ART and ART discontinuation in an HIV-infected in early life favours low levels of immune activation,
child. ART (with didanosine and zidovudine) was initiated at 18 months of age in a child which could interfere with the vicious cycle that nor-
testing HIV-positive in 1991. For the next 8.5 years, viral loads were undetectable (less mally occurs during adult infection of immune activa-
than 400 copies per ml) while the patient was on therapy. ART was discontinued at the tion, increased bystander cell death through apoptosis
age of 10 years, and, with the exception of intermittent increases in viraemia that never and pyroptosis55,56, increased susceptibility of activated
exceeded 1,000 copies per ml, viral load remained undetectable for the next 5 years
CD4+ T cells to further infection and increased virus
until the case was reported147. The HLA type of this child did not include ‘protective’
HLA alleles. However, the child was heterozygous for the CCR5Δ32 mutation, which
production (FIG. 3). For similar reasons to those hypoth-
has been shown to delay disease progression in both adult and paediatric HIV infection. esized in adult post-treatment controllers who express
disease-susceptible HLA alleles such as HLA‑B35
(REF.  11) (discussed further below), the tolerogenic
Impact of immune ontogeny on cure potential environ­ment of early life may interfere with the estab-
Several aspects of normal development will have an lishment of latent infection in the long-lived naive, stem
important influence on the establishment and size of cell-like memory and central memory CD4+ T cells.
the viral reservoir, some of which are likely to increase, There is some suggestion that early treatment of chil-
and others to decrease, the potential for HIV cure in dren, in contrast to adults, can result in lower levels
infected children. The overall balance of these effects of these latently infected long-lived T cell subsets36,41.
may depend crucially on the timing of ART initiation Reduced infection of long-lived T cell subsets has also
following birth. been observed in adult post-treatment controllers and
elite controllers14.
Tolerogenic environment in early life. The immune
system in utero and in early life is exquisitely adapted Role of TH17 cells. A factor that may also affect the abil-
to meet the particular challenges of that phase of ity of HIV to establish long-lived latency is the high
life. In utero, the need to avoid making inflamma- proportion of naive CD4+ T cells in utero and in new-
tory responses during the frequent exposure to non-­ borns. Indeed, in cord blood, there are almost no acti-
inherited maternal antigens is facilitated by the high vated CC-chemokine receptor 5 (CCR5)‑expressing
Cell-associated unspliced levels of transforming growth factor-β (TGFβ) in memory CD4+ T cells57. CCR5 is the co‑receptor by
HIV RNA
Unspliced HIV RNA is detected
the fetus that direct the peripheral differentiation of which HIV gains entry into CD4+ T cells, and activated
in cells of individuals on naive T cells, in the presence of the cognate antigen, memory CD4+ T cells expressing CCR5 are the most
antiretroviral therapy (ART) into ‘adaptive’ regulatory T cells (TReg cells)48. These susceptible targets for HIV infection58–60. By contrast,
from the initial first step of memory TReg cells are long-lived and can be found in in the intestinal epithelium in utero and at birth, almost
long-terminal repeat-mediated
children aged 7–17 years48. TReg cells make up 15% of one-third of the CD4+ T cells express CCR5, many of
HIV transcription before
splicing, host fetal blood T cells, compared with ~5% of adult blood which also express CCR6 (REF. 54), which is character-
promoter-initiated HIV T cells49. Moreover, innate immune cells at birth, com- istic of TH17 cells that are highly permissive for HIV
transcription (or pared with adulthood, respond to a panel of Toll-like infection61,62. Even resting CD4+ T cells that express
read-through transcription) or receptor (TLR) agonists by secreting higher levels of CCR6, in the presence of the CCR6 ligand CCL20, have
virus production. By contrast,
plasma HIV RNA measures
­interleukin‑6 (IL‑6), IL‑1β and IL‑23, which support an increased susceptibility to direct infection and estab-
RNA in mature virus particles. T helper 17 (TH17) cell differentiation, and lower levels lishment of HIV latency 63. Thus, the theoretical paucity
of pro-inflammatory cytokines such as type I interfer- of CD4+ T cell targets for HIV infection in utero and
2‑LTR circles ons (IFNs), and hence lower levels of IFNγ, tumour early life is at least partly compensated for by the high
(2‑long-terminal repeat circles).
necrosis factor (TNF) and IL‑12, which support TH1 cell numbers of TH17 cells and other CD4+ T cell targets
Circularized forms of
unintegrated viral DNA that are differentiation50–52. The higher levels in neonates of available in the intestinal epithelium.
by‑products of HIV DNA immunoregulatory and anti-inflammatory cytokines It is of note that the immune system at birth has a
integration into the host (such as IL‑10 and TGFβ) and of TH2 cell-­supporting strong TH17 cell bias, given the importance of TH17 cells
genome and exist only in the cytokines (IL‑4, IL‑5, IL‑9 and IL‑13) 53,54 further both functionally, in host defence against extra­cellular
nucleus. The stability of 2‑LTR
circles is controversial, but they
function to dampen down TH1 cell responses. Finally, pathogens, and structurally, in maintaining the integrity
are generally thought to be plasma levels of adenosine are higher in neonates than of the intestinal mucosal barrier 64. Microbial transloca-
short-lived. in adults and may contribute to the TH17 cell bias and tion has a central role in the vicious cycle of immune

4 | ADVANCE ONLINE PUBLICATION www.nature.com/nri


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

25
activation that results in CD4+ T cell decline during HIV
infection, and the preferential loss of TH17 cells observed 20
in HIV infection65 is therefore likely to have a dispropor-

MTCT rate (%)


tionate impact on paediatric infection. It is noteworthy 15
that, despite substantial loss of intestinal CD4+ T cells
following acute SIV infection in both natural66,67 and 10
experimental hosts68–70, TH17 cell numbers in the gut
are maintained at normal levels in non-pathogenic SIV 5
infection71. The presence of higher levels of adenosine in
mucosal tissues in non-pathogenic infection72 indicates 0
No prophylaxis With prophylaxis No prophylaxis
that the adenosine pathway may contribute to limiting
Chronic maternal infection Acute maternal
TH17 cell loss by reduced immune activation and the infection
promotion of TH17 cell differentiation. In adult HIV
infection, if ART is initiated sufficiently early, TH17 cell In utero In utero
numbers are preserved and/or restored, and ­levels of
Intra-partum Post-partum
systemic immune activation are normalized22. Thus, the
preferential loss of TH17 cells in the intestinal mucosa Post-partum
during HIV infection in utero and in early life may have
a disproportionate impact on that age group, leading to Figure 2 | Risk of MTCT in utero and in early life.
increased susceptibility to infections with extracellular Naturerisk
Mother-to‑child transmission (MTCT) Reviews Immunology
of HIV| in the
pathogens, microbial translocation and immune activa- absence of prophylactic measures is approximately 7% from
in utero infection, 18% from intra-partum infection and 15%
tion. Therefore, the timing of early ART in paediatric
from post-partum transmission (through breast milk from
infection may be particularly important. breastfeeding for 18–24 months)164–167. In utero HIV-infected
infants are viraemic and HIV DNA-positive at birth, whereas
Increased immune activation in early life. Working transmission arising at the time of delivery (intra-partum)
against the tolerogenic environment are factors that results in viraemia approximately 4 weeks later. Treatment
predispose to increased immune activation in early life. with antiretroviral therapy (ART) of all mothers testing
This is directly relevant to the reservoir size because HIV-positive in pregnancy has reduced combined in utero
increased immune activation increases viral repli- and intra-partum MTCT from 25% to 1–2%102, and prevention
cation (FIG. 3). For example, microbial translocation of breast-milk transmission through ART of the mother has
across the intestinal barrier is increased in neonates, reduced post-partum MTCT rates to 1–2%168. The remaining
1–2% MTCT rate results either from ART non-adherence or
whether HIV infected or not 73,74. This is particularly
from acute infection during pregnancy or after birth (in utero
true for premature infants, and more than one-third MTCT risk of ~20%167,169,170 or post-partum MTCT risk of
of babies born to HIV-infected mothers are premature 20–25%171) as tests to detect HIV antibody are negative
(that is, born before 37 weeks of gestation). The chan­ during acute maternal infection when peak viraemia is
ging composition of the intestinal microbiota also con- ~107 copies per ml and transmission risk is high.
tributes to higher levels of immune activation in early
life73,75. It is proposed that mixed feeding, as opposed to
exclusive breastfeeding, predisposes to increased risk of that result from BCG vaccination increase HIV infec-
gastrointestinal infections and mucosal inflammation tion risk in uninfected infants and increase disease pro-
and, therefore, an increased risk of HIV transmis- gression in infected infants78. Among the large number
sion through breast milk76,77. Microbial translocation, of infections to which newborns are exposed, cyto-
leading to increased immune activation, is similarly megalovirus (CMV) is noteworthy both for its effect
increased by breaches in the intestinal mucosa, and in increasing immune activation81 and its prevalence
thus it is reasonable to assume that the type of feeding in countries in which HIV is abundant. By 3 months
received by the child may in this way strongly influence of age, almost two-thirds of children in sub-Saharan
the establishment and size of the viral reservoir before Africa have become infected with CMV81. CMV co-­
ART initiation in the infant. infection in HIV-infected children results in more
Immune activation can also be increased by vaccina- rapid HIV disease progression82. All of these factors
tions and new infections that are common in infancy. that function to increase immune activation in early
The immunization schedule provides crucial protection life may oppose the benefits in terms of HIV infection
against diseases to which HIV-infected children may be and disease progression that would be expected to result
particularly susceptible, but certain vaccines may also from the tolerogenic immune environment in early life
have unwanted side effects. Mycobacterium bovis bacille described above. However, initiation of ART in the first
Calmette–Guérin (BCG) is a particular case in point 78. 24–48 hours of life, before many of these factors have
BCG, which is given at birth to all South African infants maximum effect, would markedly reduce their impact.
on the first day of life, protects against miliary ­tuberculosis
Miliary tuberculosis (miliary TB) and TB meningitis79, and it may also Differential localization of viral reservoirs. In adults on
(Miliary TB). Dissemination of
Mycobacterium tuberculosis
have non-specific benefits in terms of reducing mor- ART, HIV-infected T cells are found at higher frequency
most often throughout the bidity resulting from non-mycobacterial infections80. in the gastrointestinal tract and lymph nodes than in the
body, including the brain. However, the increased levels of immune activation blood83,84. A detailed comparative analysis of other tissue

NATURE REVIEWS | IMMUNOLOGY ADVANCE ONLINE PUBLICATION | 5


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

• Decreased IL-12 (TH1 cells)


• Increased IL-6 and IL-23 (TH17 cells) Innate
• Decreased type I IFNs and TNF immune cells
Tolerogenic immune • Increased IL-10
environment in utero
and early life
Increased number
of TReg cells Placental TGFβ

HIV
replication
CD4+ T cell loss Large numbers of TH17 cells
through apoptosis in the intestine in utero and
HIV infection of and pyroptosis
CD4+ T cell targets early life, predisposition to
HIV infection
Increased Structural damage
immune Increased to intestinal
activation co-infections mucosal barrier
• Gut immaturity
• Low birth weight
High proportion of • Feeding changes altering
Increased microbial
naive CD4+ T cells gut microbiota
translocation across
in utero and early life the intestinal barrier

Exclusive Expression of HLA class I


breastfeeding alleles that bind LILRB2
Vicious cycle of immune
with high avidity
activation in adult HIV
infection
BCG vaccination;
co-infections such as Impact of factors associated
CMV and TB transmitted with infection occuring in utero
from HIV-infected mother and in early life

Figure 3 | Paediatric factors affecting immune activation and size of the HIV reservoir. The vicious cycle of HIV infection
— resulting in intestinal mucosal damage, microbial translocation across the intestinal barrier and increased
Nature Reviewsimmune
| Immunology
activation, leading to increased CD4+ T cell decline, which predisposes to further co‑infections, increased immune activation,
HIV replication and CD4+ T cell loss — can be prevented by the initiation of antiretroviral therapy (ART) sufficiently early in
adult and paediatric infection. There are factors specific to paediatric infection that tend to interfere with this vicious cycle,
and others that contribute to it, the balance of which may vary between individuals and also may depend crucially on the
timing of ART initiation. Aspects of immune ontogeny that favour decreased immune activation include the tolerogenic
immune environment in utero and in early life and the high proportion of naive CD4+ T cells. Aspects of immune ontogeny
that tend to decrease paediatric cure potential include the large number of T helper 17 (TH17) cells in the intestinal mucosa.
Maternal factors that favour paediatric cure potential include maternal health, exclusive breastfeeding, lack of co‑infections
and expression of HLA class I molecules that bind leukocyte immunoglobulin-like receptor subfamily B member 2 (LILRB2)
with high avidity. BCG, bacille Calmette–Guérin; CMV, cytomegalovirus; IFN, interferon; IL, interleukin; TB, tuberculosis;
TGFβ, transforming growth factor-β; TNF, tumour necrosis factor; TReg cell, regulatory T cell.

sites, such as the spleen, liver and genital tract, has not persistence in children on ART. This is an important
been carried out, but latently infected cells are found in feature to understand, as the mechanisms that lead to
all of these tissues85. Recent findings also suggest that HIV persistence in resting T cells and macrophages dif-
latently infected cells can be detected in adipose tis- fer 93, and, therefore, strategies to activate or eliminate
sue86,87. Other long-lived infected cells can also persist infected cells will need to be different. This has recently
on ART, including tissue macrophages — as Kupffer cells been demonstrated in terms of the ex vivo response to
in the liver88, as alveolar macrophages in the lung89 and histone deacetylase (HDAC) inhibitors of virus derived
as microglia or perivascular macrophages in the cen- from brain tissue compared with spleen94.
tral nervous system (CNS)90. HIV latency has also been Finally, many recent papers have shown that the
demonstrated in astrocytes in the CNS91. It is not clear majority of HIV DNA measured in blood CD4+ T cells
whether HIV DNA that persists in tissue macro­phages is from HIV-infected individuals on ART is replication
replication competent and contributes to viral rebound defective47,95. Furthermore, the conventional viral out-
following cessation of ART. growth assay may underestimate the true number of cells
In HIV-infected children, the location of latently that carry replication-competent virus owing to the pres-
infected T cells or tissue macrophages has not been ence of intact proviruses that are not induced through
explored but may be quite different, given the low fre- T cell stimulation96. In a single recent study in children
quency of memory T cells and different levels of expres- treated with ART, it seems that there is a similar rela-
sion of key chemokine receptors. Although detecting tionship between intact and defective viruses in children
HIV persistence in tissues is challenging, recent novel as in adults46, but more studies are required in children
methods developed in the macaque model, such as using newer molecular strat­egies to define the relation-
total-body imaging 92 and RNAscope or DNAscope ship between defective and r­ eplication-competent virus
in situ hybridization, should aid future studies of HIV in this setting.

6 | ADVANCE ONLINE PUBLICATION www.nature.com/nri


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

T cell vaccine potential. Children who have received settings in particular, the outcome of the child may have
ART in the first few days of life often subsequently lack less to do with immunology than with the physical and
detectable HIV-specific immune responses. Western mental well-being of the mother and the presence or
blot non-reactivity has been proposed as a marker of absence of a social support network. There are there-
early and profound suppression of viral replication41, fore practical aspects of successful HIV treatment that
and lack of HIV-specific cellular immunity may be differ between adults and children, and the dependence
similarly indicative. However, it is well established of an infected child on the mother to administer ART,
that HIV-specific CD8+ T cell responses are evident at in addition to the other needs of childhood, is an added
birth in the majority of children infected in utero 97,98. challenge that should not be underestimated.
This suggests that rapid removal of the HIV antigen
in infancy or early childhood may result in loss of the Maternal HLA type. The risk of MTCT is strongly
memory response even though an immune response related to maternal viral load103,104, and therefore trans-
was induced. Thus, in early ART-treated HIV-infected mitting mothers are more likely to express HLA class I
children, there may be the potential for a T cell vaccine molecules that are known to be associated with high
to skew the CD8+ T cell immunodominance pattern viral set points and rapid progression to disease105,106.
in a more favourable direction — such as towards a In African populations, these are HLA‑B*1801,
broad Gag-specific response — than would be the case HLA‑B*4501 and HLA‑B*5802. In Caucasian popula-
if the pre-existing, suboptimal T cell responses that are tions, the best-studied example is HLA‑B35. As children
­generated in natural infection are predominant. inherit one HLA haplotype from their mother, in utero
Successful skewing of the natural immunodomi- HIV-infected children, as a group, are enriched with the
nance pattern of the CD8+ T cell response by an HIV HLA class I molecules that are associated with rapid
T cell vaccine has been demonstrated in recent analyses progression in adult infection106. However, as described
of participants in the South African Phambili trial99 who above, the HLA‑B alleles that have a substantial impact
subsequently became infected. In individuals express- on adult infection do not significantly influence paedi­
ing the disease-susceptible HLA‑B*5802 allele, recipi- atric disease outcome8. Indeed, it has been proposed
ents of the MRKAd5 HIV‑1 Gag/Pol/Nef vaccine made that HLA class I molecules such as HLA‑B35 that are
broader and higher magnitude Gag-specific responses associated with rapid disease progression in adults may
than HLA‑B*5802‑positive recipients of a placebo100. The actively facilitate post-treatment control14. A possible
placebo recipients showed the Env-dominated response mechanism is suggested by the fact that HLA‑B35
pattern that is typical of natural infection101. In association and HLA‑B*4501 bind leukocyte immunoglobulin-­
with skewing of the CD8+ T cell response towards tar- like receptor subfamily B member 2 (LILRB2) with
geting Gag, viral set points in the HLA‑B*5802‑positive high avidity, whereas protective HLA alleles such as
vaccine recipients were significantly lower than in the HLA‑B27, HLA‑B57 and HLA-B*5801 bind LILRB2
HLA‑B*5802‑positive placebo recipients100. with low avidity 107. LILRB2 is expressed on cells of the
In summary, a range of factors associated with the myeloid lineage, such as conventional dendritic cells,
developmental stage of the child at which HIV infec- and high-avidity HLA class I binding to LILRB2 inhibits
tion occurs, before ART is initiated, are likely to affect dendritic cell function. The failure of disease-suscepti-
markedly the ultimate size of the HIV reservoir. The ble HLA class I molecules such as HLA‑B35 to induce
overall effect of these mixed influences might depend broad HIV-specific CD8+ T cell responses in adult
crucially on the exact timing of in utero HIV transmis- infection, leading to high viral set points, may there-
sion and of ART initiation. It is probable that if ART can fore in part be due to the inhibitory action of LILRB2
be initiated within the first 24–48 hours of life, then the on dendritic cell activity. However, in the setting of
factors predisposing to increased microbial transloca- early initiation of ART, factors such as these that reduce
tion and increased immune activation would be largely antiviral T cell immunity may tend to reduce immune
mitigated, thereby providing children with an inherent activation and thereby influence the establishment and
advantage compared with adults in terms of the potential cellular localization of the latent HIV reservoir. If this
for HIV cure. proves to be the case, the apparent maternally inher-
Phambili trial ited HLA ‘handi­cap’ in infected children may in fact
A trial that tested the efficacy Impact of maternal factors on cure potential predispose towards post-treatment control following
of the MRKAd5 subtype B
HIV‑1 Gag/Pol/Nef vaccine in
In utero and intra-partum MTCT are effectively pre- early ART.
South Africa, where subtype C vented entirely if viraemia is suppressed by ART during
virus is dominant. The vaccine pregnancy. In countries such as South Africa, prevention Virus type transmitted through MTCT. One of the
increased the risk of HIV strategies have reduced MTCT rates that arise in utero bene­fits of early initiation of ART in paediatric and
acquisition and did not reduce
or intra-partum from 25–30% without any therapy to adult infection is to suppress viral replication before
early viraemia.
1–2% currently 102. When transmission does occur, this the selection of autologous escape mutants. The early
Escape mutants tends to be associated with challenging circumstances. initiation of ART might be particularly important in
Viral variants that reduce For example, more than 60% of transmitting mothers in adults, in whom cytotoxic T lymphocyte (CTL) escape
recognition by the immune South Africa have a daily income of less than US$1, the mutants are selected from the first few weeks of infec-
system, typically arising in
epitopes that are targeted by
source of water in more than one-third of cases is either tion108 and persist in latently infected T cells109. By con-
HIV-specific CD8+ T cells or a shared standpipe or river water, and toilet facilities for trast, CD8+ T cell responses in in utero HIV-infected
neutralizing antibodies. almost one-half of mothers are a pit latrine20. In these infants, even when restricted by protective HLA alleles,

NATURE REVIEWS | IMMUNOLOGY ADVANCE ONLINE PUBLICATION | 7


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

such as HLA‑B57, HLA-B*5801 and HLA-B*8101, seem of post-treatment control, as has been proposed in the
to be insufficiently functional in the first few months VISCONTI adult cohort in relation to HLA class I type.
of infection to select virus escape mutants8. However, Defining these factors will require large studies to initiate
in MTCT, the virus that is transmitted is in many cases early ART in sufficient numbers of in utero HIV-­infected
pre-adapted to T cell responses available in the child, infants to eliminate some of the misleading conclusions
as well as to maternal antibodies crossing the placenta. that can arise from anecdotal cases (BOX 2).
As the child inherits half of the mother’s HLA alleles, in
theory the virus transmitted from mother to child may Broadly neutralizing antibodies at birth. Broadly
be pre-adapted to all of the CD8+ T cell epitopes that are ­neutralizing antibodies do not typically neutralize contem-
restricted by the HLA alleles shared with the mother 110. poraneous autologous virus115 and tend to be isolated
However, for two reasons, this may be less of a problem in HIV-infected individuals who have high viral set
in reality. First, the mothers of children infected with points and are progressing to AIDS116. Maternal anti-
HIV tend to express disease-susceptible HLA alleles106 bodies crossing the placenta do not neutralize trans-
(as described above), and these typically do not select mitted virus or affect paediatric disease progression117.
any virus escape mutants in the crucial Gag epitopes111. However, heterologous broadly neutralizing antibodies
Second, as prevention of MTCT becomes increasingly can both prevent infection118,119 and enhance subsequent
effective, the proportion of in utero transmissions that B cell responses in infected infants120, and they also sub-
result from acute infection of the mother during preg- stantially reduce viral replication in simian–­human
nancy becomes greater (FIG. 2). In this scenario, in utero immuno­deficiency virus (SHIV)-infected macaques
transmission may occur before escape mutants have (PGT121 antibody)121 and in HIV-infected humans
been selected in maternal virus. (3BNC117 antibody)122. Expression of broadly neutral-
In summary, the mother has a crucial role in deter- izing antibodies by adeno-associated virus vectors123 has
mining morbidity and survival of the child. However, the potential to prevent infection for life, although long-
if an HIV-infected mother is healthy and sufficient term protection against infection using this approach has
health-care support is available for mother and child, only been demonstrated in animal models so far 124,125.
there are reasons to believe that the inheritance by the In paediatric infection, broadly neutralizing anti­
child of maternal disease-susceptible HLA alleles and bodies could be used at birth at the same time as ART
the transmission of HIV adapted to maternal immune is initiated (FIG. 3). Human monoclonal antibodies pro-
responses may not be a major disadvantage in relation tected against SHIV infection following oral challenge
to the potential for HIV cure in children versus adults. in neonatal macaques126. This approach, in combination
with ART, may potentially reduce viral loads by 1–2 log10
Opportunities for effective cure strategies over 7–10 days after infusion127 in addition to the effect
Several factors discussed here provide a rationale of ART. ART alone can reduce a typical starting viral
for the proposition that unique possibilities exist to load of 105–106 copies per ml to less than 50 copies per
facilitate the eradication or remission of HIV in chil- ml in 3–6 months20, whereas ART in combination with
dren, namely: the feasibility of initiating ART within a broadly neutralizing antibody might achieve aviraemia
24–48 hours of birth; the ability of ART to make faster in days or weeks. The timing of ART initiation and the
reductions in the size of the viral reservoir that con- time to viral suppression are both important indepen­
tinue for longer in infants compared with adults; and dent factors that influence the ultimate size of the latent
the ability of ART to minimize the effect of factors in viral reservoir 44. Early suppression of viraemia clearly
early life that would otherwise counteract the reduced limits the size of the reservoir but importantly may also
immune activation resulting from a tolerogenic envi- limit viral diversity, which thereby might increase the
ronment. However, there are also specific obstacles that efficacy of any immunotherapeutic approaches that are
narrow both the window of opportunity and the range used later, as described below.
of immunotherapeutic interventions that can be used
in children. In particular, the very poor levels of ART Timing of further immunotherapeutic interventions.
Broadly neutralizing adherence in adolescence112, coupled with the normal Having initiated ART early and achieved suppression of
antibodies onset of adolescence as early as 8 and 9 years of age in viraemia, sufficient time should then elapse before the
HIV-specific antibodies that females and males, respectively 113,114, effectively place introduction of further immune interventions: first, to
arise during natural infection in
10–30% of individuals and can
a time limit on the age by which immunotherapeutic enable the latent viral reservoir to be optimally reduced
effectively neutralize diverse interventions need to be in place (FIG. 4). to levels beneath the level of detection of the most sen-
viral isolates. sitive assays; and, second, to allow TH1 cell-polarizing
Early ART alone. The universal failure, so far, of innate immune responses to develop more fully, for the
Step trial
ART initiated in the first few weeks of life to result in reason that cell-mediated immunity is likely to have a
A trial that tested the efficacy
of the MRKAd5 subtype B post-treatment HIV control in paediatric infection, let key role in eradication of the viral reservoir 109. Although
HIV‑1 Gag/Pol/Nef vaccine in alone cure, may indicate that ART should be initiated IL‑12, a TH1 cell-polarizing cytokine, remains at lower
North and South Americas, even earlier and/or for a longer duration and that there levels in infants than in adults (even in 12‑year-old chil-
Australia and the Caribbean, may be additional interventions, other than those related dren)52, by 2 years of age levels of type I IFNs are similar
where subtype B virus is
dominant. The vaccine
to the timing of onset and duration of ART, that are to those in adults, levels of IL‑6 and IL‑23 (TH17 cell-­
increased the risk of HIV likely to be required to achieve post-treatment control. polarizing cytokines) and of IL‑10 (an immuno­
acquisition. Host genetic factors may contribute to the likelihood regulatory cytokine) are lower than in neonates, and

8 | ADVANCE ONLINE PUBLICATION www.nature.com/nri


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Vaccine (T cell and/or B cell)

Broadly neutralizing antibodies


(passive or virus-vectored)

Anti-latency agents

Anti-inflammatory agents

ART interruption

Broadly
ART • Onset of adolescence
neutralizing
initiation • Poor ART adherence
antibodies
Main window of opportunity for • Multidrug resistance
immunotherapeutic intervention • Sexually active
in paediatric infection
• Suppression of viraemia through ART
• Minimization of latent reservoir
• Development of pro-inflammatory
Early and TH1 cell immune response
intervention 8–9 years

2–5 years

Birth

Figure 4 | Principal opportunities for immunotherapeutic interventions to maximize theNature potential for HIV
Reviews cure or
| Immunology
remission in paediatric infection. In utero paediatric infection provides particular opportunities for antiretroviral
therapy (ART) to be initiated with or without additional interventions (such as broadly neutralizing antibodies) very early
in the course of infection. As cell-mediated immunity is likely to have a key role in eradication of the viral reservoir109, and
T  helper 1 (TH1) cell responses take some years to develop fully, a period of time on ART alone may be useful to minimize
the size of the viral reservoir before further interventions that are designed to eradicate infection. In late childhood,
approximately between the ages of 3 and 9 years, immune ontogeny favours the generation of effective CD8+ T cell
immunity, which provides a window of opportunity for therapeutic intervention before the onset of puberty with
associated behavioural changes, including loss of ART adherence and onset of sexual activity.

TReg cell numbers are similar to those in adults128. Indeed, The safety and immunogenicity of these prime–boost
there is evidence that the middle years of childhood rep- approaches have been established in Phase I and Phase II
resent a ‘honeymoon period’ (REF. 129), during which the trials of malaria vaccination in children ranging in age
immune response is optimally adapted against a range from infants (10 weeks old and 5–12 months old) to
of infectious agents, including TB, influenza, malaria, mid-childhood (2–6 years old) (K. Ewer, personal com-
Epstein–Barr virus, varicella zoster virus, mumps, mea- munication). The induction of protective and broad
sles and perhaps also HIV, such that it is sufficient to Gag-specific CD8+ T cell responses in children treated
control the infection without causing immuno­pathology. early by vaccination and/or the use of broadly neutral-
This period — from approximately 3 to 9 years of age — izing antibodies could provide the long-term antiviral
may therefore be the optimal window of opportunity for immunity that is necessary to contain or even to elimi­
immunotherapeutic interventions designed to facilitate nate viral replication from previously latently infected
HIV eradication, before the onset of a­ dolescence (FIG. 3). cells, once ART is discontinued.

T cell vaccines. Early generation T cell vaccines did Latency reversal. Several compounds have now been
not reduce viral loads overall in vaccine recipients, and shown to activate latent HIV infection in vitro133. These
the adenovirus type 5 (Ad5) vector used in the Step include epigenetic modifiers, protein kinase C agonists
trial and Phambili trial was associated with increased and TLR agonists. HDAC inhibitors were the first latency
risk of infection130. However, current approaches such activators to move into clinical trials in HIV-infected
as those using prime–boost regimens of chimpanzee individuals on ART; these studies all demonstrated an
adeno­virus and modified vaccinia virus Ankara result increase in cell-associated HIV RNA and, in some studies,
in T cell responses that are 1 log10 higher in magni- an increase in plasma HIV RNA, but no study reported
tude and 5–10‑fold greater in breadth compared with a change in the frequency of latently infected cells134–138.
the T cell responses achieved by MRKAd5 HIV‑1 Gag/ Although HDAC inhibitors have been widely used for the
Pol/Nef 131,132 (B. Mothe, personal communication). management of malignancy in adults, there is very little

NATURE REVIEWS | IMMUNOLOGY ADVANCE ONLINE PUBLICATION | 9


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

Box 3 | Other potential immunotherapeutic strategies in children in ex vivo studies to ensure that there are no inherent
differences in the stability of main­tenance of HIV latency
In addition to the early initiation of antiretroviral therapy (ART) and the use of broadly in children compared with adults, before the initiation of
neutralizing antibodies, T cell vaccines and latency reversal agents, other approaches ­clinical trials of latency reversal in children.
are currently being developed in adults and may be considered in the future for
paediatric use.
Other immunotherapeutic strategies. Several additional
Immune checkpoint blockade. Blockade of the immune checkpoint molecules approaches are currently being developed in adults;
programmed cell death protein 1 (PD1) and cytotoxic T lymphocyte antigen 4 (CTLA4) some of these show great promise, but their efficacy
boosts HIV-specific T cell responses ex vivo148,149. In a recent case study of CTLA4‑specific and safety profile in children have yet to be determined.
antibody (ipilimumab) in an HIV-infected adult on ART with metastatic melanoma, there
These are summarized in BOX 3.
was a significant increase in cell-associated HIV RNA and a cyclical decline in plasma HIV
RNA, which is consistent with enhanced virus clearance150.
Conclusions
Treatment intensification. Studies in adults with HIV infection show that treatment The immune system in utero and in early life favours
intensification has minimal effects on HIV persistence in patients on ART151. Two studies
low levels of immune activation, at least initially, follow-
that intensified standard ART with raltegravir demonstrated residual virus replication in
approximately 30% of individuals152,153.
ing HIV infection. However, in most cases, it seems that
ART would need to be initiated as early as possible to
Anti-inflammatory drugs. Anti-inflammatory agents are being evaluated in minimize the size of the latent viral reservoir and provide
HIV-infected adults to determine whether reducing inflammation will reduce HIV
a realistic base from which additional immunothera­
persistence — by decreasing either residual viral replication or the number of latently
peutic interventions can be introduced. The timing of
infected cells. Current clinical trials in adults include the anti-inflammatory drugs
rapamycin, lisinopril and methotrexate, and antibodies targeting interleukin‑1β154–157. their introduction and of a subsequent treatment inter-
ruption remain very much open to discussion. In chil-
Gene therapy. Gene therapy that uses zinc finger nucleases to eliminate CC-chemokine dren, as distinct from adults, there is a relatively narrow
receptor 5 (CCR5) expression in peripheral circulating T cells was recently shown to be
window of opportunity between the development of an
safe in adults, and approximately 6% of the gene-modified cells persisted in blood and
tissue158. Given safety concerns, it seems unlikely that CCR5 gene therapy will be immune response that is sufficient to eliminate activated
evaluated in children in the near future. Opportunistic stem cell transplantation from a HIV-infected cells that previously comprised the latent
donor with the CCR5Δ32 mutation could potentially be considered in a child with the reservoir and the onset of puberty with its accompanying
appropriate clinical indication, given the success following transplantation of Timothy high levels of ART non-­adherence. Recent data suggest
Brown (the Berlin patient)159. However, so far this procedure has not been successful in a that even brief ART interruptions in the absence of HIV-
further six adults with HIV infection160. specific immunity can lead to a rapid and irreversible
increase in the size of the HIV reservoir 43, underlying
the observation that reservoir size alone does not nec-
Treatment intensification experience in paediatric cases, and the sustained changes essarily predict the time to viral rebound33,140. Defining
Introduction of additional in host gene expression that result from their use would the factors that determine post-treatment control and
antiretroviral drugs to a be a serious safety concern in children136. More recently, identifying biomarkers that can predict which individ-
standard three-drug disulfiram, which has been in clinical use for decades and uals will have prolonged antiretroviral-free remission141
suppressive regimen.
with an excellent safety profile, was shown to also acti- will require additional work, including clinical trials
vate HIV latent infection139. This agent could potentially in children. So far, HIV vaccine trials have been rarely
be evaluated in children for latency reversal. It remains undertaken in children, despite evidence of promising
unknown whether there are factors that would alter the immune responses in children who have been vacci-
susceptibility of latently infected cells from HIV-infected nated142. Evaluation of early treated children who receive
children on ART to activation. This needs to be evaluated immunotherapeutic interventions should be a priority.

1. Newell, M. L. et al. Mortality of infected and uninfected International AIDS Conference, Abstr. http://pag. 14. Saez-Cirion, A. et al. Post-treatment HIV‑1 controllers
infants born to HIV-infected mothers in Africa: a pooled aids2014.org/abstracts.aspx?aid=5075 (2014). with a long-term virological remission after the
analysis. Lancet 364, 1236–1243 (2004). 8. Adland, E. et al. Discordant impact of HLA on viral interruption of early initiated antiretroviral therapy
2. Collaborative Group on AIDS Incubation & HIV replicative capacity and disease progression in ANRS VISCONTI Study. PLoS Pathog. 9, e1003211
Survival including the CASCADE EU Concerted Action. pediatric and adult HIV infection. PLoS Pathog. 11, (2013).
Concerted Action on SeroConversion to AIDS and e1004954 (2015). 15. Vigano, A. et al. Failure to eradicate HIV despite fully
Death in Europe. Time from HIV‑1 seroconversion to 9. Ferrand, R. A. et al. HIV infection presenting in successful HAART initiated in the first days of life.
AIDS and death before widespread use of highly-active older children and adolescents: a case series from J. Pediatr. 148, 389–391 (2006).
antiretroviral therapy: a collaborative re‑analysis. Harare, Zimbabwe. Clin. Infect. Dis. 44, 874–878 16. Butler, K. M. et al. Rapid viral rebound after 4 years of
Lancet 355, 1131–1137 (2000). (2007). suppressive therapy in a seronegative HIV‑1 infected
3. Kaslow, R. A. et al. Influence of combinations of 10. Judd, A. et al. Vertically acquired HIV diagnosed in infant treated from birth. Pediatr. Infect. Dis. J. 34,
human major histocompatibility complex genes on adolescence and early adulthood in the United e48–e51 (2015).
the course of HIV‑1 infection. Nat. Med. 2, 405–411 Kingdom and Ireland: findings from national 17. Bitnun, A. et al. Early initiation of combination
(1996). surveillance. HIV Med. 10, 253–256 (2009). antiretroviral therapy in HIV‑1‑infected newborns
4. Pereyra, F. et al. The major genetic determinants of 11. Silvestri, G. et al. Nonpathogenic SIV infection of sooty can achieve sustained virologic suppression with
HIV‑1 control affect HLA class I peptide presentation. mangabeys is characterized by limited bystander low frequency of CD4+ T cells carrying HIV in
Science 330, 1551–1557 (2010). immunopathology despite chronic high-level viremia. peripheral blood. Clin. Infect. Dis. 59, 1012–1019
5. Kiepiela, P. et al. CD8+ T‑cell responses to different Immunity 18, 441–452 (2003). (2014).
HIV proteins have discordant associations with viral 12. Persaud, D. et al. Absence of detectable HIV‑1 viremia 18. Giacomet, V. et al. No cure of HIV infection in a child
load. Nat. Med. 13, 46–53 (2007). after treatment cessation in an infant. N. Engl. J. Med. despite early treatment and apparent viral clearance.
6. Shearer, W. T. et al. Lymphocyte subsets in healthy 369, 1828–1835 (2013). Lancet 384, 1320 (2014).
children from birth through 18 years of age: the This is the first paper to describe the Mississippi 19. Cotton, M. F. et al. Early time-limited antiretroviral
Pediatric AIDS Clinical Trials Group P1009 study. child. therapy versus deferred therapy in South African
J. Allergy Clin. Immunol. 112, 973–980 (2003). 13. Luzuriaga, K. et al. Viremic relapse after HIV‑1 infants infected with HIV: results from the children
7. Adland, E. et al. Mechanisms of non-pathogenicity remission in a perinatally infected child. N. Engl. with HIV early antiretroviral (CHER) randomised trial.
in  HIV: lessons from paediatric infection. IAS, 20th J. Med. 372, 786–788 (2015). Lancet 382, 1555–1563 (2013).

10 | ADVANCE ONLINE PUBLICATION www.nature.com/nri


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

20. Prendergast, A. et al. Early virological suppression 42. Ananworanich, J. et al. Reduced markers of HIV changes in the actin cytoskeleton. Proc. Natl Acad.
with three-class antiretroviral therapy in HIV- persistence and restricted HIV-specific immune Sci. USA 107, 16934–16939 (2010).
infected African infants. AIDS 22, 1333–1343 (2008). responses after early antiretroviral therapy in children. 64. Brenchley, J. M. et al. Differential Th17 CD4 T‑cell
21. Ndhlovu, Z. M. et al. Magnitude and kinetics of CD8+ AIDS 28, 1015–1020 (2014). depletion in pathogenic and nonpathogenic lentiviral
T cell activation during hyperacute HIV infection 43. van Zyl, G. U. et al. Early antiretroviral therapy in South infections. Blood 112, 2826–2835 (2008).
impact viral set point. Immunity 43, 591–604 (2015). African children reduces HIV‑1‑infected cells and cell- 65. Alvarez, Y. et al. Preferential HIV infection of CCR6+
22. Schuetz, A. et al. Initiation of ART during early acute associated HIV‑1 RNA in blood mononuclear cells. Th17 cells is associated with higher levels of virus
HIV infection preserves mucosal Th17 function and J. Infect. Dis. 212, 39–43 (2015). receptor expression and lack of CCR5 ligands. J. Virol.
reverses HIV-related immune activation. PLoS Pathog. 44. Martinez-Bonet, M. et al. Establishment and 87, 10843–10854 (2013).
10, e1004543 (2014). replenishment of the viral reservoir in perinatally 66. Gordon, S. N. et al. Severe depletion of mucosal CD4+
23. Brossard, Y. et al. Frequency of early in utero HIV‑1 HIV‑1‑infected children initiating very early T cells in AIDS-free simian immunodeficiency virus-
infection: a blind DNA polymerase chain reaction study antiretroviral therapy. Clin. Infect. Dis. 61, 1169–1178 infected sooty mangabeys. J. Immunol. 179,
on 100 fetal thymuses. AIDS 9, 359–366 (1995). (2015). 3026–3034 (2007).
24. Rouzioux, C. et al. Estimated timing of mother-to‑child 45. Besson, G. J. et al. HIV‑1 DNA decay dynamics in blood 67. Pandrea, I. V. et al. Acute loss of intestinal CD4+
human immunodeficiency virus type 1 (HIV‑1) during more than a decade of suppressive antiretroviral T cells is not predictive of simian immunodeficiency
transmission by use of a Markov model. The HIV therapy. Clin. Infect. Dis. 59, 1312–1321 (2014). virus virulence. J. Immunol. 179, 3035–3046
Infection in Newborns French Collaborative Study 46. Uprety, P. et al. Cell-associated HIV‑1 DNA and RNA (2007).
Group. Am. J. Epidemiol. 142, 1330–1337 (1995). decay dynamics during early combination antiretroviral 68. Li, Q. et al. Peak SIV replication in resting memory
25. Jani, I. V. et al. Accurate early infant HIV diagnosis in therapy in HIV‑1‑infected infants. Clin. Infect. Dis. 61, CD4+ T cells depletes gut lamina propria CD4+ T cells.
primary health clinics using a point‑of‑care nucleic acid 1862–1870 (2015). Nature 434, 1148–1152 (2005).
test. J. Acquir. Immune Defic. Syndr. 67, e1–e4 47. Eriksson, S. et al. Comparative analysis of measures 69. Wang, X. et al. Massive infection and loss of CD4+
(2014). of viral reservoirs in HIV‑1 eradication studies. T cells occurs in the intestinal tract of neonatal
26. Whitney, J. B. et al. Rapid seeding of the viral reservoir PLoS Pathog. 9, e1003174 (2013). rhesus macaques in acute SIV infection. Blood 109,
prior to SIV viraemia in rhesus monkeys. Nature 512, 48. Mold, J. E. et al. Maternal alloantigens promote the 1174–1181 (2007).
74–77 (2014). development of tolerogenic fetal regulatory T cells 70. Mattapallil, J. J. et al. Massive infection and loss of
27. Fiebig, E. W. et al. Dynamics of HIV viremia and in utero. Science 322, 1562–1565 (2008). memory CD4+ T cells in multiple tissues during acute
antibody seroconversion in plasma donors: This paper describes the vigorous response of SIV infection. Nature 434, 1093–1097 (2005).
implications for diagnosis and staging of primary HIV CD4+ T cells in utero to non-inherited maternal 71. Favre, D. et al. Critical loss of the balance between
infection. AIDS 17, 1871–1879 (2003). antigens; these T cells develop in the presence of Th17 and T regulatory cell populations in pathogenic
28. Frange, P. et al. HIV‑1 virological remission lasting TGFβ into long-lived TReg cells. SIV infection. PLoS Pathog. 5, e1000295 (2009).
more than 12 years after interruption of early 49. Takahata, Y. et al. CD25+CD4+ T cells in human cord The central role of TH17 cell maintenance in the
antiretroviral therapy in a perinatally infected blood: an immunoregulatory subset with naive non-pathogenicity of SIV infection is illustrated in
teenager enrolled in the French ANRS EPF‑CO10 phenotype and specific expression of forkhead box p3 African green monkeys compared with pig-tailed
paediatric cohort: a case report. Lancet HIV 3, (Foxp3) gene. Exp. Hematol. 32, 622–629 (2004). macaques.
e49–e54 (2016). 50. Kollmann, T. R. et al. Neonatal innate TLR-mediated 72. He, T. et al. Critical role for the adenosine pathway
This is the first paper to describe the VISCONTI responses are distinct from those of adults. J. Immunol. in controlling simian immunodeficiency virus-related
child. 183, 7150–7160 (2009). immune activation and inflammation in gut mucosal
29. Finzi, D. et al. Latent infection of CD4+ T cells provides This study shows the marked differences between tissues. J. Virol. 89, 9616–9630 (2015).
a mechanism for lifelong persistence of HIV‑1, even in newborns and adults in terms of the cytokines 73. Kourtis, A. P. et al. Role of intestinal mucosal integrity
patients on effective combination therapy. Nat. Med. produced by innate immune cells in response to a in HIV transmission to infants through breast-feeding:
5, 512–517 (1999). panel of TLR agonists. the BAN study. J. Infect. Dis. 208, 653–661 (2013).
30. Siliciano, J. D. et al. Long-term follow‑up studies 51. Levy, O. et al. The adenosine system selectively inhibits 74. Papasavvas, E. et al. Increased microbial translocation
confirm the stability of the latent reservoir for HIV‑1 TLR-mediated TNF-α production in the human in ≤180 days old perinatally human immunodeficiency
in resting CD4+ T cells. Nat. Med. 9, 727–728 (2003). newborn. J. Immunol. 177, 1956–1966 (2006). virus-positive infants as compared with human
31. Crooks, A. M. et al. Precise quantitation of the latent 52. Upham, J. W. et al. Development of immunodeficiency virus-exposed uninfected infants of
HIV‑1 reservoir: implications for eradication strategies. interleukin‑12‑producing capacity throughout similar age. Pediatr. Infect. Dis. J. 30, 877–882
J. Infect. Dis. 212, 1361–1365 (2015). childhood. Infect. Immun. 70, 6583–6588 (2002). (2011).
32. Zhang, L. et al. Quantifying residual HIV‑1 replication 53. Nowak, E. C. et al. IL‑9 as a mediator of Th17‑driven 75. Wallet, M. A. et al. Microbial translocation induces
in patients receiving combination antiretroviral inflammatory disease. J. Exp. Med. 206, 1653–1660 persistent macrophage activation unrelated to HIV‑1
therapy. N. Engl. J. Med. 340, 1605–1613 (1999). (2009). levels or T‑cell activation following therapy. AIDS 24,
33. Chun, T. W. et al. Rebound of plasma viremia following This study highlights the striking contrast between 1281–1290 (2010).
cessation of antiretroviral therapy despite profoundly the lack of target cells for HIV infection in the 76. Coovadia, H. M. et al. Mother-to‑child transmission of
low levels of HIV reservoir: implications for eradication. peripheral blood in utero and the high frequency HIV‑1 infection during exclusive breastfeeding in the
AIDS 24, 2803–2808 (2010). of target cells in the fetal gut. first 6 months of life: an intervention cohort study.
34. Hocqueloux, L. et al. Long-term antiretroviral therapy 54. Elyaman, W. et al. IL‑9 induces differentiation of Lancet 369, 1107–1116 (2007).
initiated during primary HIV‑1 infection is key to TH17 cells and enhances function of FoxP3+ natural 77. Iliff, P. J. et al. Early exclusive breastfeeding reduces
achieving both low HIV reservoirs and normal T cell regulatory T cells. Proc. Natl Acad. Sci. USA 106, the risk of postnatal HIV‑1 transmission and increases
counts. J. Antimicrob. Chemother. 68, 1169–1178 12885–12890 (2009). HIV-free survival. AIDS 19, 699–708 (2005).
(2013). 55. Doitsh, G. et al. Cell death by pyroptosis drives CD4 78. Tchakoute, C. T. et al. Delaying BCG vaccination until
35. Schmid, A. et al. Profound depletion of HIV‑1 T‑cell depletion in HIV‑1 infection. Nature 505, 8 weeks of age results in robust BCG-specific T‑cell
transcription in patients initiating antiretroviral 509–514 (2014). responses in HIV-exposed infants. J. Infect. Dis. 211,
therapy during acute infection. PLoS ONE 5, e13310 56. Munoz-Arias, I. et al. Blood-derived CD4 T Cells 338–346 (2015).
(2010). naturally resist pyroptosis during abortive HIV‑1 79. Roy, A. et al. Effect of BCG vaccination against
36. Buzon, M. J. et al. Long-term antiretroviral treatment infection. Cell Host Microbe 18, 463–470 (2015). Mycobacterium tuberculosis infection in children:
initiated at primary HIV‑1 infection affects the size, 57. Bunders, M. J. et al. Memory CD4+CCR5+ T cells are systematic review and meta-analysis. BMJ 349,
composition, and decay kinetics of the reservoir abundantly present in the gut of newborn infants to g4643 (2014).
of HIV‑1‑infected CD4 T cells. J. Virol. 88, facilitate mother-to‑child transmission of HIV‑1. Blood 80. Aaby, P. et al. Randomized trial of BCG vaccination
10056–10065 (2014). 120, 4383–4390 (2012). at birth to low-birth-weight children: beneficial
37. Archin, N. M. et al. Immediate antiviral therapy 58. Veazey, R. S. et al. Identifying the target cell in primary nonspecific effects in the neonatal period? J. Infect.
appears to restrict resting CD4+ cell HIV‑1 infection simian immunodeficiency virus (SIV) infection: highly Dis. 204, 245–252 (2011).
without accelerating the decay of latent infection. activated memory CD4+ T cells are rapidly eliminated 81. Miles, D. J. et al. Cytomegalovirus infection in
Proc. Natl Acad. Sci. USA 109, 9523–9528 (2012). in early SIV infection in vivo. J. Virol. 74, 57–64 Gambian infants leads to profound CD8 T‑cell
38. Wightman, F. et al. Both CD31+ and CD31– naive (2000). differentiation. J. Virol. 81, 5766–5776 (2007).
CD4+ T cells are persistent HIV type 1‑infected 59. Bleul, C. C., Wu, L., Hoxie, J. A., Springer, T. A. & 82. Kovacs, A. et al. Cytomegalovirus infection and HIV‑1
reservoirs in individuals receiving antiretroviral Mackay, C. R. The HIV coreceptors CXCR4 and CCR5 disease progression in infants born to HIV‑1‑infected
therapy. J. Infect. Dis. 202, 1738–1748 (2010). are differentially expressed and regulated on human women. Pediatric Pulmonary and Cardiovascular
39. Jaafoura, S. et al. Progressive contraction of the latent T lymphocytes. Proc. Natl Acad. Sci. USA 94, Complication of Vertically Transmitted HIV Infection
HIV reservoir around a core of less-differentiated 1925–1930 (1997). Study Group. N. Engl. J. Med. 341, 77–84 (1999).
CD4+ memory T Cells. Nat. Commun. 5, 5407 (2014). 60. Wu, L. et al. CCR5 levels and expression pattern 83. Chun, T. W. et al. Persistence of HIV in gut-associated
40. Persaud, D. et al. Dynamics of the resting CD4+ T‑cell correlate with infectability by macrophage-tropic HIV‑1, lymphoid tissue despite long-term antiretroviral
latent HIV reservoir in infants initiating HAART less in vitro. J. Exp. Med. 185, 1681–1691 (1997). therapy. J. Infect. Dis. 197, 714–720 (2008).
than 6 months of age. AIDS 26, 1483–1490 (2012). 61. Monteiro, P. et al. Memory CCR6+CD4+ T cells are 84. Fletcher, C. V. et al. Persistent HIV‑1 replication is
41. Luzuriaga, K. et al. HIV type 1 (HIV‑1) proviral preferential targets for productive HIV type 1 infection associated with lower antiretroviral drug concentrations
reservoirs decay continuously under sustained regardless of their expression of integrin β7. in lymphatic tissues. Proc. Natl Acad. Sci. USA 111,
virologic control in HIV‑1‑infected children who J. Immunol. 186, 4618–4630 (2011). 2307–2312 (2014).
received early treatment. J. Infect. Dis. 210, 62. Gosselin, A. et al. Peripheral blood CCR4+CCR6+ 85. Saksena, N. K. et al. HIV reservoirs in vivo and new
1529–1538 (2014). and CXCR3+CCR6+CD4+ T cells are highly permissive strategies for possible eradication of HIV from the
This paper indicates that decay of the viral to HIV‑1 infection. J. Immunol. 184, 1604–1616 reservoir sites. HIV AIDS (Auckl.) 2, 103–122 (2010).
reservoir not only occurs faster in early treated (2010). 86. Couturier, J. et al. Human adipose tissue as a reservoir
paediatric infection but also continues throughout 63. Cameron, P. U. et al. Establishment of HIV‑1 latency in for memory CD4+ T cells and HIV. AIDS 29, 667–674
childhood and possibly into adolescence. resting CD4+ T cells depends on chemokine-induced (2015).

NATURE REVIEWS | IMMUNOLOGY ADVANCE ONLINE PUBLICATION | 11


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

87. Damouche, A. et al. Adipose tissue is a neglected viral 106. Kiepiela, P. et al. Dominant influence of HLA‑B in test‑of‑concept trial. Lancet 372, 1881–1893
reservoir and an inflammatory site during chronic HIV mediating the potential co‑evolution of HIV and HLA. (2008).
and SIV infection. PLoS Pathog. 11, e1005153 Nature 432, 769–775 (2004). 131. Borthwick, N. et al. Vaccine-elicited human T cells
(2015). 107. Bashirova, A. A. et al. LILRB2 interaction with HLA recognizing conserved protein regions inhibit HIV‑1.
88. Kandathil, A. J. et al. Liver macrophages and HIV‑1 class I correlates with control of HIV‑1 infection. Mol. Ther. 22, 464–475 (2014).
persistence. Conference on Retroviruses and PLoS Genet. 10, e1004196 (2014). 132. Frahm, N. et al. Human adenovirus-specific T cells
Opportunistic Infections, Abstr. 281 http:// 108. Goonetilleke, N. et al. The first T cell response to modulate HIV-specific T cell responses to an
www.croiconference.org/sites/default/files/uploads/ transmitted/founder virus contributes to the control Ad5‑vectored HIV‑1 vaccine. J. Clin. Invest. 122,
croi2015-program-abstracts.pdf (2015). of acute viremia in HIV‑1 infection. J. Exp. Med. 206, 359–367 (2012).
89. Cribbs, S. K. et al. Metabolomics of bronchoalveolar 1253–1272 (2009). 133. Archin, N. M., Sung, J. M., Garrido, C.,
lavage differentiate healthy HIV‑1‑infected subjects 109. Deng, K. et al. Broad CTL response is required to clear Soriano-Sarabia, N. & Margolis, D. M. Eradicating
from controls. AIDS Res. Hum. Retroviruses 30, latent HIV‑1 due to dominance of escape mutations. HIV‑1 infection: seeking to clear a persistent
579–585 (2014). Nature 517, 381–385 (2015). pathogen. Nat. Rev. Microbiol. 12, 750–764
90. Sturdevant, C. B. et al. Compartmentalized replication 110. Goulder, P. J. et al. Evolution and transmission of (2014).
of R5 T cell-tropic HIV‑1 in the central nervous system stable CTL escape mutations in HIV infection. Nature 134. Archin, N. M. et al. Administration of vorinostat
early in the course of infection. PLoS Pathog. 11, 412, 334–338 (2001). disrupts HIV‑1 latency in patients on antiretroviral
e1004720 (2015). 111. Matthews, P. C. et al. Central role of reverting therapy. Nature 487, 482–485 (2012).
91. Churchill, M. J. et al. Extensive astrocyte infection is mutations in HLA associations with human 135. Archin, N. M. et al. HIV‑1 expression within resting
prominent in human immunodeficiency virus- immunodeficiency virus set point. J. Virol. 82, CD4+ T cells after multiple doses of vorinostat.
associated dementia. Ann. Neurol. 66, 253–258 8548–8559 (2008). J. Infect. Dis. 210, 728–735 (2014).
(2009). 112. Nsheha, A. H., Dow, D. E., Kapanda, G. E., 136. Elliott, J. H. et al. Activation of HIV transcription with
92. Santangelo, P. J. et al. Whole-body immunoPET reveals Hamel, B. C. & Msuya, L. J. Adherence to antiretroviral short-course vorinostat in HIV-infected patients on
active SIV dynamics in viremic and antiretroviral therapy among HIV-infected children receiving care at suppressive antiretroviral therapy. PLoS Pathog. 10,
therapy-treated macaques. Nat. Methods 12, Kilimanjaro Christian Medical Centre (KCMC), e1004473 (2014).
427–432 (2015). Northern Tanzania: a cross-sectional analytical study. 137. Rasmussen, T. A. et al. Panobinostat, a histone
93. Gorry, P. R., Francella, N., Lewin, S. R. & Pan Afr. Med. J. 17, 238 (2014). deacetylase inhibitor, for latent-virus reactivation in
Collman, R. G. HIV‑1 envelope-receptor interactions 113. Biro, F. M. et al. Onset of breast development in a HIV-infected patients on suppressive antiretroviral
required for macrophage infection and implications for longitudinal cohort. Pediatrics 132, 1019–1027 therapy: a phase 1/2, single group, clinical trial.
current HIV‑1 cure strategies. J. Leukoc. Biol. 95, (2013). Lancet HIV 1, e13–e21 (2014).
71–81 (2014). 114. Herman-Giddens, M. E., Wang, L. & Koch, G. 138. Sogaard, O. S. et al. The depsipeptide romidepsin
94. Gray, L. R. et al. CNS-specific regulatory elements in Secondary sexual characteristics in boys: estimates reverses HIV‑1 latency in vivo. PLoS Pathog. 11,
brain-derived HIV‑1 strains affect responses to latency- from the national health and nutrition examination e1005142 (2015).
reversing agents with implications for cure strategies. survey III, 1988–1994. Arch. Pediatr. Adolesc. Med. 139. Elliott, J. H. et al. Short-term administration of
Mol. Psychiatry http://dx.doi.org/10.1038/ 155, 1022–1028 (2001). disulfiram for reversal of latent HIV infection: a phase
mp.2015.111 (2015). 115. Wei, X. et al. Antibody neutralization and escape by 2 dose-escalation study. Lancet HIV 2, e520–e529
95. Josefsson, L. et al. The HIV‑1 reservoir in eight HIV‑1. Nature 422, 307–312 (2003). (2015).
patients on long-term suppressive antiretroviral 116. Moore, P. L. et al. Evolution of an HIV glycan- 140. Henrich, T. J. et al. Antiretroviral-free HIV‑1 remission
therapy is stable with few genetic changes over dependent broadly neutralizing antibody epitope and viral rebound after allogeneic stem cell
time. Proc. Natl Acad. Sci. USA 110, E4987–E4996 through immune escape. Nat. Med. 18, 1688–1692 transplantation: report of 2 cases. Ann. Intern. Med.
(2013). (2012). 161, 319–327 (2014).
96. Ho, Y. C. et al. Replication-competent noninduced 117. Wu, X. et al. Neutralization escape variants of human 141. Hurst, J. et al. Immunological biomarkers predict
proviruses in the latent reservoir increase barrier to immunodeficiency virus type 1 are transmitted from HIV‑1 viral rebound after treatment interruption.
HIV‑1 cure. Cell 155, 540–551 (2013). mother to infant. J. Virol. 80, 835–844 (2006). Nat. Commun. 6, 8495 (2015).
97. Luzuriaga, K. et al. HIV‑1‑specific cytotoxic T 118. Moldt, B. et al. Highly potent HIV-specific antibody 142. Klein, N. et al. Early antiretroviral therapy in children
lymphocyte responses in the first year of life. neutralization in vitro translates into effective perinatally infected with HIV: a unique opportunity to
J. Immunol. 154, 433–443 (1995). protection against mucosal SHIV challenge in vivo. implement immunotherapeutic approaches to prolong
98. Thobakgale, C. F. et al. Human immunodeficiency Proc. Natl Acad. Sci. USA 109, 18921–18925 viral remission. Lancet Infect. Dis. 15, 1108–1114
virus-specific CD8+ T‑cell activity is detectable from (2012). (2015).
birth in the majority of in utero-infected infants. 119. Mascola, J. R. et al. Protection of macaques against 143. Mphatswe, W. et al. High frequency of rapid
J. Virol. 81, 12775–12784 (2007). vaginal transmission of a pathogenic HIV‑1/SIV immunological progression in African infants infected
99. Gray, G. E. et al. Recombinant adenovirus type 5 chimeric virus by passive infusion of neutralizing in the era of perinatal HIV prophylaxis. AIDS 21,
HIV gag/pol/nef vaccine in South Africa: unblinded, antibodies. Nat. Med. 6, 207–210 (2000). 1253–1261 (2007).
long-term follow‑up of the phase 2b HVTN 503/ 120. Ng, C. T. et al. Passive neutralizing antibody controls 144. Lisziewicz, J. et al. Control of HIV despite the
Phambili study. Lancet Infect. Dis. 14, 388–396 SHIV viremia and enhances B cell responses in discontinuation of antiretroviral therapy. N. Engl.
(2014). infant macaques. Nat. Med. 16, 1117–1119 J. Med. 340, 1683–1684 (1999).
100. Leitman, E. M. et al. HLA‑B*58:02‑specific benefit (2010). 145. Jessen, H., Allen, T. M. & Streeck, H. How a single
of MRKAd5 Gag/Pol/Nef vaccine in an African 121. Barouch, D. H. et al. Therapeutic efficacy of potent patient influenced HIV research—15‑year follow‑up.
population. J. Int. AIDS Soc. Abstr. 18 (5 Suppl. 4), neutralizing HIV‑1‑specific monoclonal antibodies in N. Engl. J. Med. 370, 682–683 (2014).
20479 (2015). SHIV-infected rhesus monkeys. Nature 503, 224–228 146. Pereyra, F. et al. Genetic and immunologic
This analysis of Phambili study participants who (2013). heterogeneity among persons who control HIV
subsequently became HIV-infected shows the 122. Caskey, M. et al. Viraemia suppressed in infection in the absence of therapy. J. Infect. Dis. 197,
ability of a human T cell vaccine to switch the CD8+ HIV‑1‑infected humans by broadly neutralizing 563–571 (2008).
T cell immunodominance pattern from an antibody 3BNC117. Nature 522, 487–491 (2015). 147. Feeney, M. E., Tang, Y., Rathod, A., Kneut, C. &
Env-dominated response associated with high 123. Johnson, P. R. et al. Vector-mediated gene transfer McIntosh, K. Absence of detectable viremia in a
viraemia to a broad Gag-specific response engenders long-lived neutralizing activity and perinatally HIV‑1‑infected teenager after
associated with a lower viral set point. protection against SIV infection in monkeys. Nat. Med. discontinuation of antiretroviral therapy. J. Allergy
101. Ngumbela, K. C. et al. Targeting of a CD8 T cell env 15, 901–906 (2009). Clin. Immunol. 118, 324–330 (2006).
epitope presented by HLA‑B*5802 is associated with 124. Balazs, A. B. et al. Antibody-based protection against 148. Day, C. L. et al. PD‑1 expression on HIV-specific T cells
markers of HIV disease progression and lack of HIV infection by vectored immunoprophylaxis. Nature is associated with T‑cell exhaustion and disease
selection pressure. AIDS Res. Hum. Retroviruses 24, 481, 81–84 (2012). progression. Nature 443, 350–354 (2006).
72–82 (2008). 125. Gardner, M. R. et al. AAV-expressed eCD4‑Ig provides 149. Kaufmann, D. E. et al. Upregulation of CTLA‑4 by
102. Moodley, P., Parboosing, R. & Moodley, D. Reduction durable protection from multiple SHIV challenges. HIV-specific CD4+ T cells correlates with disease
in perinatal HIV infections in KwaZulu-Natal, South Nature 519, 87–91 (2015). progression and defines a reversible immune
Africa, in the era of more effective prevention of 126. Baba, T. W. et al. Human neutralizing monoclonal dysfunction. Nat. Immunol. 8, 1246–1254
mother to child transmission interventions antibodies of the IgG1 subtype protect against (2007).
(2004‑2012). J. Acquir. Immune Defic. Syndr. 63, mucosal simian-human immunodeficiency virus 150. Wightman, F. et al. Effect of ipilimumab on the HIV
410–415 (2013). infection. Nat. Med. 6, 200–206 (2000). reservoir in an HIV-infected individual with
103. Dickover, R. E. et al. Identification of levels of 127. Hayden, E. C. Almighty antibodies? A new wave of metastatic melanoma. AIDS 29, 504–506 (2015).
maternal HIV‑1 RNA associated with risk of antibody-based approaches aims to combat HIV. 151. Schulze zur Wiesch, J. & van Lunzen, J. Hide and
perinatal transmission. Effect of maternal zidovudine Nat. Med. 21, 657–659 (2015). seek… can we eradicate HIV by treatment
treatment on viral load. JAMA 275, 599–605 128. Kollmann, T. R., Levy, O., Montgomery, R. R. & intensification? J. Infect. Dis. 203, 894–897
(1996). Goriely, S. Innate immune function by Toll-like (2011).
104. Sperling, R. S. et al. Maternal viral load, zidovudine receptors: distinct responses in newborns and the 152. Buzon, M. J. et al. The HIV‑1 integrase genotype
treatment, and the risk of transmission of human elderly. Immunity 37, 771–783 (2012). strongly predicts raltegravir susceptibility but not viral
immunodeficiency virus type 1 from mother to infant. 129. Ahmed, R., Oldstone, M. B. & Palese, P. Protective fitness of primary virus isolates. AIDS 24, 17–25
Pediatric AIDS Clinical Trials Group Protocol 076 immunity and susceptibility to infectious diseases: (2010).
Study Group. N. Engl. J. Med. 335, 1621–1629 lessons from the 1918 influenza pandemic. 153. Hatano, H. et al. Increase in 2‑long terminal repeat
(1996). Nat. Immunol. 8, 1188–1193 (2007). circles and decrease in D‑dimer after raltegravir
105. Goulder, P. J. & Walker, B. D. HIV and HLA class I: 130. Buchbinder, S. P. et al. Efficacy assessment of a cell- intensification in patients with treated HIV infection:
an evolving relationship. Immunity 37, 426–440 mediated immunity HIV‑1 vaccine (the Step Study): a randomized, placebo-controlled trial. J. Infect. Dis.
(2012). a double-blind, randomised, placebo-controlled, 208, 1436–1442 (2013).

12 | ADVANCE ONLINE PUBLICATION www.nature.com/nri


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
REVIEWS

154. US National Library of Medicine. ClinicalTrials.gov, and European Collaborative Study. J. Acquir. transmission in South Africa. J. Infect. Dis. 203,
https://clinicaltrials.gov/ct2/show/NCT01949116 Immune Defic. Syndr. Hum. Retrovirol. 14, 442–450 1231–1234 (2011).
(2016). (1997). 170. Drake, A. L., Wagner, A., Richardson, B. &
155. US National Library of Medicine. ClinicalTrials.gov, 163. Paul, M. E. et al. Predictors of immunologic long- John-Stewart, G. Incident HIV during pregnancy
https://clinicaltrials.gov/ct2/show/NCT01535235 term nonprogression in HIV-infected children: and postpartum and risk of mother-to‑child HIV
(2015). implications for initiating therapy. J. Allergy Clin. transmission: a systematic review and meta-analysis.
156. US National Library of Medicine. ClinicalTrials.gov, Immunol. 115, 848–855 (2005). PLoS Med. 11, e1001608 (2014).
https://clinicaltrials.gov/ct2/show/NCT00000834 164. Connor, E. M. et al. Reduction of maternal-infant 171. Humphrey, J. H. et al. Mother to child transmission of
(2012). transmission of human immunodeficiency virus HIV among Zimbabwean women who seroconverted
157. US National Library of Medicine. ClinicalTrials.gov, type 1 with zidovudine treatment. Pediatric AIDS postnatally: prospective cohort study. BMJ 341,
https://clinicaltrials.gov/ct2/show/NCT02272946 Clinical Trials Group Protocol 076 Study Group. c6580 (2010).
(2016). N. Engl. J. Med. 331, 1173–1180 (1994).
158. Tebas, P. et al. Gene editing of CCR5 in autologous 165. De Cock, K. M. et al. Prevention of mother-to‑child HIV Acknowledgements
CD4 T cells of persons infected with HIV. N. Engl. transmission in resource-poor countries: translating P.J.G. is funded by the Wellcome Trust (WT104748 MA). S.R.L.
J. Med. 370, 901–910 (2014). research into policy and practice. JAMA 283, is supported by the National Institutes of Health Delaney AIDS
159. Hutter, G. et al. Long-term control of HIV by CCR5 1175–1182 (2000). Research Enterprise (Grant U19 AI096109) and a National
Δ32/Δ32 stem-cell transplantation. N. Engl. J. Med. 166. Marinda, E. et al. Child mortality according to Health and Medical Research Council (NHMRC) of Australia
360, 692–698 (2009). maternal and infant HIV status in Zimbabwe. Pediatr. Practitioner Fellowship. E.M.L. is supported by the
160. Hutter, G. More on shift of HIV tropism in stem- Infect. Dis. J. 26, 519–526 (2007). Clarendon Foundation.
cell transplantation with CCR5 Δ32/Δ32 167. Taha, T. E. et al. Association of recent HIV infection
mutation. N. Engl. J. Med. 371, 2437–2438 and in utero HIV‑1 transmission. AIDS 25, Competing interests statement
(2014). 1357–1364 (2011). The authors declare no competing interests.
161. Mori, M. et al. Sex differences in antiretroviral 168. Mofenson, L. M. Prevention in neglected
therapy initiation in pediatric HIV infection. subpopulations: prevention of mother-to‑child
PLoS ONE 10, e0131591 (2015). transmission of HIV infection. Clin. Infect. Dis. 50 DATABASES
162. Blanche, S. et al. Morbidity and mortality in (Suppl. 3), 130–148 (2010). ClinicalTrials.gov: http://www.clinicaltrials.gov
European children vertically infected by HIV‑1. 169. Moodley, D. et al. Incident HIV infection in pregnant ALL LINKS ARE ACTIVE IN THE ONLINE PDF
The French Pediatric HIV Infection Study Group and lactating women and its effect on mother-to‑child

NATURE REVIEWS | IMMUNOLOGY ADVANCE ONLINE PUBLICATION | 13


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.

You might also like