You are on page 1of 18

Article

An active HIV reservoir during ART is associated with


maintenance of HIV-specific CD8+ T cell magnitude
and short-lived differentiation status
Graphical abstract Authors
Hiroshi Takata, Julie L. Mitchell,
Julian Pacheco, ..., Lydie Trautmann,
RV254/SEARCH010, RV304/
SEARCH013

Correspondence
ltrautmann@global-id.org

In brief
Takata et al. found that the residual
antigen expression by the active HIV
reservoir during suppressive
antiretroviral therapy correlated with
increased HIV-specific CD8+ T cell
magnitude but prevented differentiation
into functional cells. Thus, targeting the
residual HIV reservoir may improve HIV-
specific CD8+ T cell functionality needed
for an HIV remission.

Highlights
d Magnitude of CD8+ T cell response associated with active HIV
reservoir size on therapy

d Sustained CD8+ T cell responses correlated with a greater HIV


provirus decay

d Residual HIV reservoir levels associated with maintenance of


short-lived CD8+ T cells

d Low HIV reservoir associated with generation of functional


HIV-specific CD8+ T cells

Takata et al., 2023, Cell Host & Microbe 31, 1494–1506


September 13, 2023 ª 2023 The Author(s). Published by Elsevier Inc.
https://doi.org/10.1016/j.chom.2023.08.012 ll
ll
OPEN ACCESS

Article
An active HIV reservoir during ART is associated
with maintenance of HIV-specific CD8+ T cell
magnitude and short-lived differentiation status
Hiroshi Takata,1,2,3 Julie L. Mitchell,1,2,3 Julian Pacheco,1 Amélie Pagliuzza,4 Suteeraporn Pinyakorn,2,3
Supranee Buranapraditkun,5 Carlo Sacdalan,6,5 Louise Leyre,4 Sam Nathanson,1 Juyeon C. Kakazu,2,3 Jintana Intasan,6
Peeriya Prueksakaew,6 Nitiya Chomchey,6 Nittaya Phanuphak,7 Mark de Souza,6 Elias K. Haddad,8 Morgane Rolland,2,3
Sodsai Tovanabutra,2,3 Sandhya Vasan,2,3 Denise C. Hsu,2,3 Nicolas Chomont,4 Lydie Trautmann,1,2,3,9,*
and RV254/SEARCH010, RV304/SEARCH013
1Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR 97006, USA
2U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
3Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD 20817, USA
4Centre de Recherche du CHUM and Department of Microbiology, Infectiology and Immunology, Université de Montréal, Montréal, QC,

Canada
5Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
6SEARCH Research Foundation, Bangkok, Thailand
7Institute of HIV Research and Innovation, Bangkok, Thailand
8Department of Medicine, Division of Infectious Diseases and HIV Medicine, Drexel University, Philadelphia, PA 19102, USA
9Lead contact

*Correspondence: ltrautmann@global-id.org
https://doi.org/10.1016/j.chom.2023.08.012

SUMMARY

Before initiation of antiretroviral therapy (ART), HIV-specific CD8+ T cells are dysfunctional and short lived. To
better understand the relationship between the HIV reservoir in CD4+ T cells and the magnitude and differen-
tiation status of HIV-specific CD8+ T cells, we investigated these cells from acute and chronic HIV-infected
individuals after 2 years of ART. Although both the HIV reservoir and the CD8+ T cell responses declined
significantly after 2 years of ART, sustained HIV-specific CD8+ T cell responses correlated with a greater
reduction of integrated HIV provirus. However, the magnitude of CD8+ T cells specific for HIV Gag, Pol,
Nef, and Vif proteins positively associated with the active reservoir size during ART, measured as cell-asso-
ciated RNA. Importantly, high HIV DNA levels strongly associate with maintenance of short-lived HIV-specific
CD8+ T cells, regardless of ART initiation time. Our data suggest that the active reservoir maintains HIV-spe-
cific CD8+ T cell magnitude but prevents their differentiation into functional cells.

INTRODUCTION ing of the HIV reservoir.11 However, HIV-specific CD8+ T cells


become dysfunctional in most PWH during untreated chronic
Although antiretroviral therapy (ART) can lead to viral suppression HIV infection (CHI) because of continuous HIV antigen stimula-
below the limit of detection of clinical assays, life-long ART is tion, which induces the overexpression of checkpoint proteins
associated with significant burdens and comorbidities.1 ART- transducing inhibitory signals and drives cell exhaustion.12–15
free human immunodeficiency virus (HIV) remission, while desir- ART initiated during CHI partially restores the defect in HIV-
able, has only been achieved in a few individuals, including five specific CD8+ T cells by decreasing activation and expression
bone-marrow transplant recipients and at least two natural con- of checkpoint proteins, increasing cytokine production, and
trollers with extremely low HIV reservoir.2 Rare people with HIV increasing expression of the interleukin (IL)-7 receptor (IL-7R),
(PWH), termed ‘‘elite controllers,’’ can naturally control HIV repli- which reflects long-lived potential.16–21 However, the cells are
cation below the limit of routine clinical assays in the absence of still dysfunctional compared with those in elite controllers, sug-
ART.3,4 HIV-specific CD8+ T cells with high functionality have gesting that ART is not enough to fully reverse the defects.5–7,22
been considered key to achieving HIV control in elite control- We recently showed that initiation of ART in AHI, but not CHI,
lers.5–8 During acute HIV infection (AHI), HIV-specific CD8+ promoted the differentiation of HIV-specific CD8+ T cells with
T cells have been shown to limit viral replication,9–11 and we self-renewal and long-lived phenotypes.23 Although ART low-
have also previously shown that when ART was initiated in AHI ered the checkpoint protein programmed cell death 1 (PD-1)
HIV-specific CD8+ T cells are functional and able to reduce seed- expression on ex vivo HIV-specific CD8+ T cells in people who

1494 Cell Host & Microbe 31, 1494–1506, September 13, 2023 ª 2023 The Author(s). Published by Elsevier Inc.
This is an open access article under the CC BY-NC license (http://creativecommons.org/licenses/by-nc/4.0/).
ll
Article OPEN ACCESS

Table 1. Clinical characteristics of study participants analyzed on ART


Chronic treated for Chronic treated for Acute treated for
Pre-ART HIV infection stage 24 weeks (n = 17) 96 weeks (n = 17) 96 weeks (n = 48)
Percentage of male 100% 100% 98%
Age (years), median (interquartile range [IQR]) 29 (26–32) 30 (28–34) 27 (24–31)
Pre-ART plasma viral load copies/mL, median (IQR) 28,962 (7,127–92,155) 28,962 (7,127–92,155) 1,001,493 (64,465–7,349,735)
CD4 count cells/mm3 in plasma, median (IQR) 793 (603–949) 680 (548–858) 618 (489–806)
CD8 count cells/mm3 in plasma, median (IQR) 1,283 (780–1,658) 974 (599–1,195) 644 (462–785)

initiated ART during CHI, it remained higher relative to those in whether large or active HIV reservoirs prevent the differentiation
people treated from AHI, and the overexpression of PD-1 was of functional HIV-specific CD8+ T cells under suppressive ART.
further enhanced after in vitro antigen recall. Consistent with
this, recalled HIV-specific CD8+ T cells from people treated RESULTS
from CHI showed dampened cell expansion and cytotoxic ca-
pacities compared with those from people treated from AHI, indi- Frequencies of HIV-specific CD8+ T cells and HIV
cating that HIV-specific CD8+ T cells retain functional defects reservoir size declined during ART
upon recall.23 Persistence of this residual dysfunction on ART To understand the dynamics of the HIV reservoir and HIV-specific
could be due to its imprinting in HIV-specific CD8+ T cells prior CD8+ T cell response after ART initiation, we analyzed longitudi-
to ART initiation or due to ongoing mechanisms that maintain nal samples from participants in the RV304/SEARCH013 cohort
this dysfunction despite suppressive ART, though these have after 24 and 96 weeks of ART initiated during CHI (Table 1). All
not been demonstrated yet in HIV-specific CD8+ T cells. participants were virologically suppressed with plasma viral
Although the majority of HIV proviruses are defective in peo- load below 50 copies/mL at these visits as well as at weeks 48
ple on ART,24 these defective proviruses still produce HIV anti- and 72. Both total HIV DNA and integrated HIV DNA copy
gens, which are recognized by HIV-specific CD8+ T cells.25,26 numbers in ex vivo CD4+ T cells significantly declined between
Indeed, HIV Nef-specific CD8+ T cell responses positively asso- 24 and 96 weeks of ART (Figure 1A). We analyzed frequencies
ciated with the HIV reservoir size on long-term ART.27 The of HIV-specific CD8+ T cells recognizing all HIV proteins by stim-
change in magnitude of this Nef-specific response also corre- ulating them with peptide pools tailored to the Thai cohort
lated with the HIV reservoir size on ART.28 These data suggest (Table S1) and performing a new, more sensitive activation
that HIV-specific CD8+ T cells recognize HIV antigens but do induced marker (AIM)/intracellular cytokine staining (ICS) assay,
not lead to the elimination of the HIV reservoir under ART. which consists of simultaneous ICS for interferon (IFN)-g and
This is in line with the hypothesis that reactivated latently in- detection of the AIMs CD69 and 4-1BB (Figure S1).36,37 HIV-spe-
fected cells are resistant to killing by HIV-specific CD8+ cific CD8+ T cells from PWH on ART for 24 weeks targeted pre-
T cells29 and those with intact proviruses are enriched in cells dominantly Env, Gag, Pol, and Nef, and the hierarchy of targeted
expressing immune checkpoint markers.30 However, the HIV HIV proteins was maintained after 96 weeks of ART (Figures 1B
reservoir is defined as a compartment with a diverse virologic and S2A). When normalized for the number of peptides included
profile, ranging from latency through viral protein expression for each protein, responses were dominant for Gag and Nef
to virion production. It is unclear whether HIV-specific CD8+ (Figures S2B and S2C). Frequencies of Gag-, Nef-, Rev/Tat-,
T cells can recognize these different forms of HIV-infected cells and total HIV-specific CD8+ T cells significantly declined between
and eliminate them. 24 and 96 weeks of ART, with a similar trend toward reduction in
ART initiation during AHI leads to a lower HIV reservoir size31,32 the response to Env and Vif proteins (Figure 1C). HIV-specific
and induces functionally superior HIV-specific CD8+ T cells CD4+ T cells detected as CD69+CD40L+IFN-g+ cells from PWH
compared with those generated when ART is initiated during on ART prominently targeted Gag and Nef (Figures S1 and
CHI.23 As HIV-specific CD8+ T cells are short-lived effectors S3A–S3D), and their responses did not exhibit a clear decline be-
with a transitional memory phenotype prior to ART initiation in tween 24 and 96 weeks of ART, suggesting that HIV-specific
both AHI and CHI, we hypothesized that it is the HIV reservoir CD4+ T cells are maintained differently on ART compared with
that persists during ART that contributes to the residual dysfunc- HIV-specific CD8+ T cells (Figure S3E). Taken together, these re-
tion of HIV-specific CD8+ T cells and prevents their differentiation sults indicate that the size of the HIV reservoir and the frequency
into long-lived cells under ART. To better understand the of HIV-specific CD8+ T cells sharply decline during the first 2
relationship between the HIV reservoir in CD4+ T cells and the years of suppressive therapy initiated during CHI.
magnitude and differentiation status of HIV-specific CD8+
T cells, we investigated these cells after 2 years of ART in people Sustained HIV-specific CD8+ T cell responses
who initiated ART during AHI and CHI from the RV254/ associated with a reduction in the proviral HIV reservoir
SEARCH010 AHI cohort11,33,34 and the RV304/SEARCH013 To determine whether HIV-specific CD8+ T cells persisting dur-
CHI cohort,35 respectively, in Bangkok, Thailand. We measured ing ART contribute to the reduction in cells harboring HIV, we
total and integrated HIV DNA and cell-associated RNA levels in analyzed the association between the frequency of HIV-specific
CD4+ T cells as well as the long-lived and stemness phenotypes CD8+ T cells at 96 weeks of ART and the fold reduction in HIV
of HIV-specific CD8+ T cells within the same participants to define reservoir size (total HIV DNA and integrated HIV DNA) between

Cell Host & Microbe 31, 1494–1506, September 13, 2023 1495
ll
OPEN ACCESS Article

A B

Figure 1. HIV reservoir size and frequency of HIV-specific CD8+ T cell decay after ART initiation in people treated from CHI
(A) Total and integrated HIV DNA copy number in CD4+ T cells from paired samples 24 and 96 weeks after ART initiation in people treated from CHI.
(B) Frequency of CD69+4-1BB+IFN-g+ HIV-specific CD8+ T cells in total CD8+ T cells 96 weeks after ART initiation.
(C) Frequency of CD69+4-1BB+IFN-g+ HIV-specific CD8+ T cells in total CD8+ T cells 24 and 96 weeks after ART initiation. Differences between 24 and 96 weeks
after ART initiation were analyzed by Wilcoxon test. **p < 0.01; ***p < 0.001.

24 and 96 weeks of ART. Strikingly, higher reduction of inte- this assay captures long terminal repeat (LTR) circles and uninte-
grated HIV DNA levels correlated with a higher frequency of grated HIV DNA38,39 that cannot generate viral proteins (Fig-
Nef-specific CD8+ T cells and a trend toward higher frequencies ure S4). These data suggest that only cells harboring integrated,
of Gag- and total HIV-specific CD8+ T cells (Figures 2A and 2B). and potentially translation competent genomes are sensitive to
Moreover, the fold reduction in the HIV reservoir size also corre- the pressure exerted by HIV-specific CD8+ T cells.
lated with a smaller decline in the frequency of Gag-, Nef-, Vif-,
Rev/Tat-, and total HIV-specific CD8+ T cells (Figures 2C and People treated in AHI have lower HIV reservoir but
2D), calculated as fold change in the frequency of HIV-specific comparable frequencies of HIV-specific CD8+ T cells
CD8+ T cells between 24 and 96 weeks of ART. Similarly, the responding to antigen stimulation compared with
fold reduction in the HIV reservoir size also correlated with fold individuals treated in chronic infection
change in the frequencies of Vif- and total HIV-specific CD4+ To better assess the virologic profile of HIV-infected cells that
T cells (Figure 2E). These results suggest that persistent HIV- can be recognized by HIV-specific CD8+ T cells, we measured
specific CD8+ and CD4+ T cells contribute to the elimination of cell-associated HIV RNA (LTR-gag transcripts) as a surrogate
HIV-infected cells during suppressive ART. Of note, most of of the active reservoir. As most people who initiated ART during
these associations were lost when total HIV DNA was used as CHI typically display a high but narrow range in the frequency of
a marker of the HIV reservoir, which may reflect the fact that infected cells (100–1,000 copies of HIV DNA per million CD4+

1496 Cell Host & Microbe 31, 1494–1506, September 13, 2023
ll
Article OPEN ACCESS

A Figure 2. Higher reduction of the HIV reservoir associated


with higher and stable frequencies of HIV-specific CD8+
and CD4+ T cells on suppressive ART
(A and B) Correlation between the fold change in integrated HIV DNA
copy number and the frequency of CD69+4-1BB+IFN-g+ cells
directed against Nef and Gag (A) as well as total HIV-specific CD8+
T cells (B) in total CD8+ T cells from 96 weeks after ART initiation.
(C and D) Correlation between the fold change in integrated HIV
DNA copy number and the fold change in frequency of CD69+4-
1BB+IFN-g+ total HIV-specific CD8+ T cells in total CD8+ T cells
(C) and HIV-specific CD8+ T cells directed against Gag, Nef, Vif, and
Rev/Tat (D) from 24 to 96 weeks after ART initiation.
(E) Correlation between the fold change in integrated HIV DNA copy
B C number and the fold change in frequency of CD69+CD40L+IFN-g+
total HIV-specific CD4+ T cells in total CD4+ T cells and HIV-specific
CD4+ T cells directed against Vif from 24 to 96 weeks after ART
initiation. Correlations were analyzed by Spearman correlation with
the Benjamini-Hochberg procedure for multiple comparisons (false
discovery rate [FDR] < 0.1). Underlined p value represents
FDR > 0.1. *p < 0.05; **p < 0.01; ****p < 0.0001.

Cell Host & Microbe 31, 1494–1506, September 13, 2023 1497
ll
OPEN ACCESS Article

A B

Figure 3. HIV reservoir size and frequency of HIV-specific CD8+ T cells under suppressive ART in people treated from AHI and CHI
(A) Total HIV DNA and cell-associated RNA copy number in CD4+ T cells 96 weeks after ART initiation in people treated from AHI (Tx AHI) and CHI (Tx CHI). Open
circles represent values under the detection limit.
(B) Correlation between the total HIV DNA and cell-associated RNA copy numbers in CD4+ T cells 96 weeks after ART initiation in people treated from AHI
and CHI.

(legend continued on next page)

1498 Cell Host & Microbe 31, 1494–1506, September 13, 2023
ll
Article OPEN ACCESS

T cells), we analyzed samples from people who initiated ART HIV reservoir size associated with the differentiation
during AHI to provide a wider range of HIV reservoir size. As pre- status of HIV-specific CD8+ T cells persisting on ART
viously reported, people who initiated ART during AHI showed To characterize the differentiation status of HIV-specific CD8+
significantly lower levels of total HIV DNA31,32 and cell-associ- T cells persisting on ART, we used peptide major histocompati-
ated HIV RNA levels compared with people who initiated ART bility complex (MHC) class I tetramers specific for 5 different
during CHI (Figure 3A). Of note, these two HIV reservoir mea- HIV epitopes restricted by human leukocyte antigen (HLA)-
sures strongly correlated (Figure 3B). Unfortunately, measure- A*1101 and Cw*0102, dominant alleles in the Thai popula-
ment of integrated HIV DNA levels could not be done concom- tion.11,23,40 Tetramer+ HIV-specific CD8+ T cells after 96 weeks
itant with measurement of the active reservoir, as the nucleic of ART did not associate with pre-ART viral load (Figure 5A) or
acid extraction process causes shearing of the DNA into pre-ART total HIV DNA (Figure S6A). However, the frequency
fragments too small for the long-range Alu PCR used for HIV of HIV-specific CD8+ T cells detected by tetramer significantly
integrated DNA. Although more than half of the participants correlated with total HIV DNA on ART, while the association
who initiated ART during AHI had no detectable levels of and strong trend were also confirmed within people treated
cell-associated HIV RNA (Figure 3A), HIV-specific CD8+ from AHI and CHI, respectively (Figure 5B). Two transcription
T cells were detected in all participants with a dominance for factors, T cell factor 1 (TCF-1) and thymocyte selection-associ-
Env and Pol HIV proteins, but no protein dominance when ad- ated high-mobility group box (TOX), have been shown to play
justing for the number of peptides per protein (Figure 3C). a role in self-renewal capacity41–43 and cell exhaustion,44–48
Though people treated from AHI had lower HIV reservoir levels, respectively, during chronic viral infections, and their expression
the frequency of HIV-specific CD8+ T cells responding to anti- is used to classify CD8+ T cells as self-renewing cells (TCF-1high
gen stimulation was comparable to those from people treated TOXlow) and short-lived cells (TCF-1lowTOXhigh).49 Prior to ART
from CHI (Figure 3D). initiation, HIV-specific CD8+ T cells were almost exclusively
TCF-1lowTOXhigh short-lived effector cells, regardless of the
HIV transcription associated with the frequency of HIV- stage of infection (Figures S6B and S6C). After ART initiation,
specific CD8+ T cells during ART the majority of HIV-specific CD8+ T cells still remained TCF-1low
To analyze whether the HIV reservoir burden affects HIV-specific TOXhigh short-lived effector cells in people who initiated ART
CD8+ T cell frequencies and further understand the extent during CHI, whereas the cells from people treated from AHI
to which the HIV reservoir size affects HIV-specific CD8+ T cell differentiated into TCF-1highTOXlow stem-like self-renewing cells
responses during ART, we performed correlative analyses be- (Figures 5C and 5D). Similar to the frequency of HIV-specific
tween the different measures of HIV reservoir and the magnitude CD8+ T cells, the frequency of TCF-1highTOXlow cells did not
of total HIV- or HIV protein-specific CD8+ T cell responses. Total correlate with pre-ART HIV DNA (Figure S6D). In contrast, total
HIV DNA levels positively correlated with Gag-, Pol-, and total HIV DNA levels on ART negatively correlated with the fre-
HIV-specific CD8+ T cells with R values similar to those previ- quencies of TCF-1highTOXlow HIV-specific CD8+ T cells, while
ously reported for Nef responses (Figures 4A and S5A).27 The the same correlation and trend were also found in people treated
correlations were further adjusted for the groups (Tx AHI and from CHI and AHI, respectively (Figure 5E). These data suggest
Tx CHI) and pre-ART plasma viral load because those potentially that the HIV reservoir size affects the balance of these popula-
bias the HIV-specific CD8+ T cell responses and the HIV reservoir tions, with a higher HIV reservoir size associated with mainte-
size, respectively. Pol-specific CD8+ T cells were still weakly nance of a short-lived effector phenotype and a lower HIV reser-
associated with total HIV DNA copies after the adjustment. voir size associated with more differentiation toward stem-like
Moreover, cell-associated HIV RNA levels correlated with the self-renewing cells. We further investigated the cell exhaustion/
frequencies of HIV-specific CD8+ T cells recognizing all HIV pro- activation status, measured by PD-1 expression, and long-lived
teins except for Env as well as total HIV-specific CD8+ T cell fre- capacity, measured by IL-7R expression, of HIV-specific CD8+
quencies, with stronger associations compared with those for T cells during ART. Expression of these markers after ART did
total HIV DNA, and the correlations with Gag-, Pol-, Nef-, Vif-, not associate with pre-ART HIV DNA levels (Figure S6E).
and total HIV-specific CD8+ T cells remained significant after However, higher HIV DNA levels on ART associated with higher
adjustment for group and pre-ART viral load (Figures 4B and PD-1 expression on HIV-specific CD8+ T cells, and the associa-
S5B). Consistently, transcriptional activity of the HIV reservoir, tion and strong trend were also found in people treated from
calculated as the ratio of cell-associated HIV RNA to HIV DNA CHI and AHI, respectively (Figure 5F), although this correlation
copy number, showed similar associations with frequencies of was not significant after adjustment. Conversely, lower HIV
HIV-specific CD8+ T cells as the cell-associated RNA levels DNA levels associated with higher expression of IL-7R on
(Figures 4C and S5C). These results demonstrate that the magni- HIV-specific CD8+ T cells, and the association and strong trend
tude of HIV-specific CD8+ T cells persisting on ART is associated were also confirmed in people treated from CHI and AHI,
with the active HIV reservoir size, suggesting that they may be respectively. Most of these correlations were maintained when
maintained by residual viral transcription from the active HIV cell-associated HIV RNA was used instead of total HIV DNA,
reservoir during ART. although they lost significance when they were adjusted

(C) Frequency of CD69+4-1BB+IFN-g+ HIV-specific CD8+ T cells in total CD8+ T cells from people treated from acute HIV infection 96 weeks after ART initiation,
with and without normalization per number of peptides corresponding to each HIV protein.
(D) Comparison of the frequency of CD69+4-1BB+IFN-g+ HIV-specific CD8+ T cells in people treated from CHI and AHI. Differences between people treated from
AHI and CHI were analyzed by Mann-Whitney test. Correlation was analyzed by Spearman correlation. *p < 0.05; **p < 0.01; ****p < 0.0001.

Cell Host & Microbe 31, 1494–1506, September 13, 2023 1499
ll
OPEN ACCESS Article

A Figure 4. Frequency of HIV-specific CD8+


T cells associated with size of active HIV
reservoir on ART
Correlation between the frequency of CD69+4-
1BB+IFN-g+ HIV-specific CD8+ T cells in total
CD8+ T cells and the total HIV DNA (A), cell-
associated RNA (B), or ratio of cell-associated
RNA to HIV DNA (C) copy number in CD4+ T cells
96 weeks after ART initiation. Open circles repre-
sent values under the detection limit. Correlations
were analyzed by Spearman correlation with the
Benjamini-Hochberg procedure for multiple com-
parisons (FDR < 0.1). Underlined p value repre-
sents FDR > 0.1. p values adjusted for group (Tx
AHI and Tx CHI) and pre-ART plasma viral load are
also shown. *p < 0.05; **p < 0.01.

grated HIV DNA. This is consistent with our


previous report that HIV-specific CD8+
T cells persisting shortly after ART initiation
in AHI more efficiently restrict the seeding
of the HIV reservoir.11 These data suggest
that strategies aimed at maintaining HIV-
specific CD8+ T cells after ART initiation,
either early or later in infection, may lead
to a greater reduction of the pool of in-
fected cells.50 Importantly, we found sig-
nificant correlations between the size of
the transcriptionally active reservoir,
C
measured by cell-associated RNA, and
the magnitude of HIV-specific CD8+ T cell
responses directed against total HIV pro-
teins as well as individual Gag, Pol, Nef,
and Vif proteins. These data suggest that
transcriptionally active HIV-infected cells
may still produce antigens from all HIV
proteins, possibly in viral sanctuary sites
such as the genital tract and lymphoid
tissues.51,52
In chronic infections and cancer, CD8+
T cells with stem-like self-renewal capac-
ity play a critical role in disease outcome.
These cells express the transcriptional
factor TCF-1 and can give rise to
for timing of ART initiation and pre-ART plasma viral load more differentiated TCF-1low short-lived cells.43 Indeed, these
(Figures S7A–S7C). Altogether, these findings suggest that a TCF-1+ cells are the predominant cell type that proliferates in
larger HIV reservoir prevents the differentiation of HIV-specific response to anti-PD-1 immune checkpoint blockade ther-
CD8+ T cells into long-lived HIV-specific CD8+ T cells with self- apy53,54 and are associated with longer survival in cancer pa-
renewal capacity on ART. tients.55 Moreover, HIV-specific CD8+ T cells in elite controllers
have higher proliferative capacity5 and elevated TCF-1 expres-
DISCUSSION sion compared with non-controllers.22,47 We observed that a
larger HIV reservoir size is associated with reduced differentia-
In this study, we measured total and integrated HIV DNA as well as tion of TCF-1highTOXlow stem-like self-renewing cells and main-
cell-associated HIV RNA through 96 weeks of ART to identify asso- tenance of TCF-1lowTOXhigh short-lived effector cells. Similarly,
ciations with HIV-specific CD8+ T cell responses in people treated we found that higher total HIV DNA levels were associated with
in AHI and CHI. We found that although both the HIV reservoir lower expression of IL-7R and maintenance of PD-1 expression
levels and HIV-specific CD8+ T cell responses decline between on HIV-specific CD8+ T cells. Although we detected very strong
weeks 24 and 96 in people treated in CHI, sustained HIV-specific correlations between the differentiation status of HIV-specific
CD8+ T cell responses on ART correlated with a reduction in inte- CD8+ T cells on ART and the HIV reservoir size on ART, we did

1500 Cell Host & Microbe 31, 1494–1506, September 13, 2023
ll
Article OPEN ACCESS

A B Figure 5. The HIV reservoir size associated with differ-


entiation status of HIV-specific CD8+ T cells on ART
(A and B) Correlation between frequency of ex vivo pMHC
tetramer+ HIV-specific CD8+ T cells in total CD8+ T cells from
people treated for more than 1.5 years from AHI and CHI and
Pre-ART plasma viral load (A) or total HIV DNA copies per 106
CD4+ T cells (B) at the sampling of HIV-specific CD8+ T cells
analyzed.
(C and D) Frequency of TCF-1highTOXlow and TCF-1lowTOXhigh
cells in HIV-specific CD8+ T cells from people treated from AHI
and CHI.
(E) Correlation between total HIV DNA copies and frequency of
TCF-1highTOXlow cells in HIV-specific CD8+ T cells on ART.
(F) Correlation between total HIV DNA copies and expression
of PD-1 and IL-7R on ex vivo HIV-specific CD8+ T cells on ART.
Correlations were analyzed by Spearman correlation with the
Benjamini-Hochberg procedure for multiple comparisons
C (FDR < 0.1). Differences between people treated from AHI and
CHI were analyzed by Wilcoxon test. p values adjusted for
group (Tx AHI and Tx CHI) and pre-ART plasma viral load are
also shown. *p < 0.05; **p < 0.01; ***p < 0.001; ****p < 0.0001.

D E

Cell Host & Microbe 31, 1494–1506, September 13, 2023 1501
ll
OPEN ACCESS Article

not observe these associations with HIV DNA levels prior to ART from differences in the HIV-specific CD8+ T cells detected by
initiation. This suggests that it was not an imprinting of the anti- the 2 assays. The AIM/ICS assay detects HIV-specific CD8+
gen burden before ART initiation but rather an active recognition T cells that are able to respond to antigen stimulation by express-
of HIV antigens by CD8+ T cells during ART that was responsible ing the AIM markers and producing cytokines, whereas cells de-
for these associations, although PD-1 expression could poten- tected by tetramers also contain HIV-specific CD8+ T cells that
tially be regulated by epigenetic programming.19 Together, these are unable to respond due to possible cell exhaustion. These
data suggest that higher HIV reservoir size on ART prevents the observations suggest that the active HIV reservoir maintains
differentiation of functional stem-like self-renewing HIV-specific HIV-specific CD8+ T cells that are able to respond to HIV anti-
CD8+ T cells, which in turn may prevent a functional recall gens. Another possibility would be differences in specificity of
response by HIV-specific CD8+ T cells after ART interruption cells detected by the 2 assays, as tetramers only identify cells
by sustaining a skewed differentiation toward short-lived cells. recognizing a single epitope, whereas the AIM/ICS was de-
Previous studies which measured HIV-specific CD8+ T cell fre- signed to identify any cells recognizing any epitopes in HIV.
quencies during ART showed that they predominantly target Further, the cell-associated RNA assay detects LTR-gag RNA
Gag, Pol, and Nef proteins27,56,57 and have reported an associa- and, while the AIM/ICS assay would detect HIV-specific CD8+
tion with the HIV reservoir size only for Nef responses.27,28 In our T cells targeting all HIV proteins, including Gag, only 20% of
study, Nef- and Pol-specific CD8+ T cell responses were more HIV-specific CD8+ T cells detected by the tetramers were Gag-
strongly associated with cell-associated RNA level than Gag- specific, with the majority of cells being Nef-specific CD8+
specific CD8+ T cell responses, suggesting that Gag-specific T cells, which are immunodominant in this cohort.11,23,40 Future
CD8+ T cells are less efficient at sensing ongoing HIV activation studies are needed to evaluate direct associations between HIV
than Nef- or Pol-specific CD8+ T cells. If this finding is confirmed RNA expression and HIV-specific CD8+ T cells targeting the HIV
by additional studies, it may have important implications on protein corresponding to the RNA measured.
therapeutic vaccines that may need to target CD8+ T cells In this study, we included people treated in AHI and CHI to
specific for Nef and Pol in addition to Gag to contain residual analyze a wider range of HIV reservoir size than studies analyzing
HIV activation under ART. We also found a dominance of Env re- only people treated in CHI. However, even within people treated
sponses in our cohorts and associations between cell-associ- in AHI, the HIV reservoir size varies greatly, from undetectable
ated HIV RNA with several HIV proteins. As Env is more variable HIV DNA levels to levels comparable to people treated in CHI.
across individuals than the other proteins,58 our use of a tailored As we previously showed that sustained HIV-specific CD8+
peptide pool covering the entire HIV consensus sequence in the T cell responses shortly after ART initiation in AHI limited HIV
Thai cohorts as well as the major variants may have allowed for reservoir seeding,11 these differences in HIV reservoir size might
detection of these Env responses. This difference of dominance be due to differences in the potency of the immune response. We
in responses could also be explained by the cohorts analyzed, chose to analyze HIV reservoir after 24 and 96 weeks of ART as
with our cohorts predominantly composed of Thai individuals this corresponds to a time after the initial rapid decay of HIV
with different HLAs and infected with a clade CRF01_AE virus. reservoir after ART initiation but before stabilization of the reser-
Finally, the assays used in previous studies and our study are voir size after 4 years of ART.59,60 As we previously reported that
also different. The ELISpot, ICS, and AIM assays used in previ- HIV-specific CD8+ T cell responses remain stable on long-term
ous studies have a high limit of detection and may not be sensi- ART over 3–5 years,61 this time frame also allowed us to measure
tive enough to detect the low frequencies of HIV-specific CD8+ a decay in HIV-specific CD8+ T cell frequency prior to stabiliza-
T cells that persist on ART.23 We designed a new assay that tion of these responses. Previous studies, which only detected
combines AIM and ICS to lower the limit of detection and detect an association between Nef-specific CD8+ T cell responses
these low frequencies of HIV-specific CD8+ T cells with higher and changes in the HIV reservoir size, were conducted using
accuracy. This enhanced accuracy in measuring responses for samples collected after more than 4 years of ART, when the
all HIV proteins may explain our ability to detect correlations be- decay of both HIV reservoir and HIV-specific CD8+ T cell re-
tween HIV reservoir and HIV-specific CD8+ T cell responses sponses is much smaller.27,28 In this study, the active reservoir
directed to numerous HIV proteins. We used MHC class I tetra- was measured by cell-associated HIV RNA. We acknowledge
mers tailored to the Thai population to detect low frequencies of that a large fraction of proviruses producing HIV transcripts are
HIV-specific CD8+ T cell responses. This method is the most likely to be defective. However, this form is more likely to pro-
sensitive measurement of HIV-specific CD8+ T cells, detecting duce HIV antigens that can be recognized by HIV-specific
all T cells specific for the tetramer regardless of their ability to CD8+ T cells,25 possibly explaining the stronger correlations
respond to peptide stimulation, and in turn provided the most we measured between HIV-specific CD8+ T cells and cell-asso-
robust correlation between the magnitude of HIV-specific ciated RNA.
CD8+ T cells and HIV reservoir. The use of different and novel as- Our study suggests that residual immune dysfunction driven
says to detect HIV-specific CD8+ T cells provided complemen- by the active HIV reservoir on ART could contribute to the lack
tary data to draw a more complete picture of the association be- of viral control after analytical treatment interruption by prevent-
tween the HIV reservoir persisting on ART and these responses. ing the differentiation of functional stem-like self-renewing HIV-
We observed that the active HIV reservoir, measured as cell- specific CD8+ T cells that can mount efficient rapid recall re-
associated RNA, strongly associated with frequencies of HIV- sponses. Therefore, HIV remission strategies will likely need to
specific CD8+ T cells detected by the AIM/ICS assay but weakly target transcriptionally active proviruses producing viral proteins
associated with frequencies of cells detected by the peptide during ART to harness HIV-specific CD8+ T cells to control re-
MHC (pMHC) tetramers. This discordance potentially came bounding of the virus after ART cessation. Future studies are

1502 Cell Host & Microbe 31, 1494–1506, September 13, 2023
ll
Article OPEN ACCESS

needed to define strategies to reduce or silence the active HIV B HIV DNA and HIV RNA measures
reservoir on ART and define whether those would be able to B Statistical analysis
reverse the residual dysfunction of HIV-specific CD8+ T cells per-
sisting on ART. SUPPLEMENTAL INFORMATION

Supplemental information can be found online at https://doi.org/10.1016/j.


Limitations of the study chom.2023.08.012.
This study has several limitations. It would have been informative
to determine correlations between HIV-specific CD8+ T cell re- ACKNOWLEDGMENTS
sponses and intact HIV DNA levels. Most participants in our
study were infected with HIV-1 CRF01_AE or CRF01_AE/B re- We thank our study participants and staff from the Thai Red Cross AIDS
Research Centre (TRC-ARC), Chulalongkorn University, and Armed Forces
combinants, and the intact proviral DNA assay (IPDA) has not
Research Institute of Medical Sciences (AFRIMS) for their valuable contribu-
yet been adapted to CRF01_AE. In addition, the total numbers
tions to this study. We want to thank D. Brooks and B. Colton for technical
of HIV genomes are very low in people treaded from AHI.31,61 assistance. We are grateful to the Government Pharmaceutical Organization
Therefore, intact HIV genomes are present at extremely low fre- of Thailand, ViiV Healthcare, Gilead Sciences, and Merck for providing the anti-
quencies in the majority of these participants,62 suggesting the retrovirals for this study. We also thank the RV254/SEARCH010 and RV304/
IPDA assay might not be an optimal assay for this population. SEARCH013 study group members at TRC-ARC, AFRIMS, and MHRP. All
However, both total and integrated HIV DNA levels have been MHC class I peptide monomers were obtained through the NIH Tetramer
Core Facility. This study was supported by the following sources: NIH grants
shown to strongly correlate with intact proviruses measured by
R01AI108433, R01MH106466, and UM1AI64560, a cooperative agreement
IPDA.63 Additionally, we believe that non-intact HIV-infected (W81XWH-07-2-0067, W81XWH-11-2-0174, W81XWH-18-2-0040) between
cells expressing HIV protein,25,26 which would not be captured the Henry M. Jackson Foundation for the Advancement of Military Medicine
by the IPDA assay, could also contribute to the altered differen- Inc. and the U.S. Department of Defense, and, in part, by the Division of
tiation of functional HIV-specific CD8+ T cells on ART. A second AIDS, National Institute of Allergy and Infectious Diseases, National Institute
limitation of this study was the small sample size due to the lon- of Health (AAI20052001). The content of this manuscript is solely the responsi-
bility of the authors and does not necessarily represent the official views of any
gitudinal sampling and identified number of participants positive
of the institutions mentioned above, the U.S. Department of the Army or the
for the pMHC tetramers used. It would be informative to analyze U.S. Department of Defense, the Henry M. Jackson Foundation for the
correlations between HIV-specific CD8+ T cell responses and Advancement of Military Medicine, Inc., the National Institutes of Health,
HIV reservoir size in people treated from AHI and CHI separately; the Department of Health and Human Services, or the United States Govern-
however, the limited sample size in each participant group did ment, nor does the mention of trade names, commercial products, or organiza-
not confer enough statistical power to perform the analysis tions imply endorsement by the Thai Red Cross AIDS Research Centre. The
investigators have adhered to the policies for protection of human participants
with the separate groups. Therefore, we performed careful sta-
as prescribed in AR-70-25.
tistical analyses to eliminate possible biases by adjusting for
the groups and pre-ART plasma viral load. Nevertheless, we AUTHOR CONTRIBUTIONS
were able to confirm that the majority of the correlations re-
mained significant after adjusting for participant group and H.T. designed and performed the experiments, analyzed the data, and wrote
the manuscript. J.L.M., J.P., S.N., and J.C.K. performed the experiments and
pre-ART plasma viral load using a multivariate linear regression
helped analyze the data and write the manuscript. A.P. and L.L. performed
model. Additional studies are warranted to understand how the HIV DNA and RNA quantifications. S.B., M.R., and S.T. performed the
timing of ART initiation affects the association between HIV-spe- sequence and alignment of HIV variants for designing the HIV peptide pools.
cific CD8+ T cells and HIV reservoir persistence under ART. S.P. provided help in statistical analyses. C.S., J.I., S.T., P.P., and N. Chomchey
managed the participant recruitment and follow-up in the studies. N.P. and
M.d.S. provided support for the clinical studies. N. Chomont and E.K.H. pro-
STAR+METHODS
vided conceptual advice and edited the manuscript. S.V. and D.C.H. designed
the clinical study, provided conceptual advice, and edited the manuscript. L.T.
Detailed methods are provided in the online version of this paper designed the experiments, analyzed the data, and wrote the manuscript.
and include the following:
DECLARATION OF INTERESTS
d KEY RESOURCES TABLE
d RESOURCE AVAILABILITY The authors declare no competing interests.
B Lead contact
Received: January 20, 2023
B Materials availability
Revised: June 2, 2023
B Data and code availability
Accepted: August 14, 2023
d EXPERIMENTAL MODEL AND STUDY PARTICIPANT Published: September 13, 2023
DETAILS
B Study design REFERENCES
B Study approval
1. Chawla, A., Wang, C., Patton, C., Murray, M., Punekar, Y., de Ruiter, A.,
d METHOD DETAILS
and Steinhart, C. (2018). A review of long-term toxicity of antiretroviral
B Antibodies and Reagents for Flow cytometry analysis
treatment regimens and implications for an aging population. Infect. Dis.
B HIV peptide pools for intracellular cytokine staining Ther. 7, 183–195. https://doi.org/10.1007/s40121-018-0201-6.
+ +
B Ex vivo detection of HIV-specific CD8 and CD4 2. Gupta, P.K., and Saxena, A. (2021). HIV/AIDS: current updates on the dis-
T cells by intracellular cytokine staining ease, treatment and prevention. Proc. Natl. Acad. Sci. India Sect. B Biol.
B Flow cytometry analysis Sci. 91, 495–510. https://doi.org/10.1007/s40011-021-01237-y.

Cell Host & Microbe 31, 1494–1506, September 13, 2023 1503
ll
OPEN ACCESS Article
3. Lambotte, O., Boufassa, F., Madec, Y., Nguyen, A., Goujard, C., Meyer, L., antiretroviral therapy. J. Virol. 82, 3391–3404. https://doi.org/10.1128/
Rouzioux, C., Venet, A., and Delfraissy, J.F.; SEROCO-HEMOCO Study JVI.02383-07.
Group (2005). HIV controllers: a homogeneous group of HIV-1-infected 17. Streeck, H., Brumme, Z.L., Anastario, M., Cohen, K.W., Jolin, J.S., Meier,
patients with spontaneous control of viral replication. Clin. Infect. Dis. A., Brumme, C.J., Rosenberg, E.S., Alter, G., Allen, T.M., et al. (2008).
41, 1053–1056. https://doi.org/10.1086/433188. Antigen load and viral sequence diversification determine the functional
4. Migueles, S.A., and Connors, M. (2010). Long-term nonprogressive dis- profile of HIV-1-specific CD8+ T cells. PLoS Med. 5, e100. https://doi.
ease among untreated HIV-infected individuals: clinical implications of un- org/10.1371/journal.pmed.0050100.
derstanding immune control of HIV. JAMA 304, 194–201. https://doi.org/
18. Janbazian, L., Price, D.A., Canderan, G., Filali-Mouhim, A., Asher, T.E.,
10.1001/jama.2010.925.
Ambrozak, D.R., Scheinberg, P., Boulassel, M.R., Routy, J.P., Koup,
5. Migueles, S.A., Laborico, A.C., Shupert, W.L., Sabbaghian, M.S., Rabin, R.A., et al. (2012). Clonotype and repertoire changes drive the functional
R., Hallahan, C.W., Van Baarle, D., Kostense, S., Miedema, F., improvement of HIV-specific CD8 T cell populations under conditions of
McLaughlin, M., et al. (2002). HIV-specific CD8+ T cell proliferation is limited antigenic stimulation. J. Immunol. 188, 1156–1167. https://doi.
coupled to perforin expression and is maintained in nonprogressors. org/10.4049/jimmunol.1102610.
Nat. Immunol. 3, 1061–1068. https://doi.org/10.1038/ni845.
19. Youngblood, B., Noto, A., Porichis, F., Akondy, R.S., Ndhlovu, Z.M.,
6. Migueles, S.A., Osborne, C.M., Royce, C., Compton, A.A., Joshi, R.P., Austin, J.W., Bordi, R., Procopio, F.A., Miura, T., Allen, T.M., et al.
Weeks, K.A., Rood, J.E., Berkley, A.M., Sacha, J.B., Cogliano-Shutta, (2013). Cutting edge: prolonged exposure to HIV reinforces a poised
N.A., et al. (2008). Lytic granule loading of CD8+ T cells is required for epigenetic program for PD-1 expression in virus-specific CD8 T cells.
HIV-infected cell elimination associated with immune control. Immunity J. Immunol. 191, 540–544. https://doi.org/10.4049/jimmunol.1203161.
29, 1009–1021. https://doi.org/10.1016/j.immuni.2008.10.010.
20. Vigano, S., Negron, J., Ouyang, Z., Rosenberg, E.S., Walker, B.D.,
7. Migueles, S.A., Weeks, K.A., Nou, E., Berkley, A.M., Rood, J.E., Osborne, Lichterfeld, M., and Yu, X.G. (2015). Prolonged antiretroviral therapy pre-
C.M., Hallahan, C.W., Cogliano-Shutta, N.A., Metcalf, J.A., McLaughlin, serves HIV-1-specific CD8 T cells with stem cell-like properties. J. Virol.
M., et al. (2009). Defective human immunodeficiency virus-specific 89, 7829–7840. https://doi.org/10.1128/JVI.00789-15.
CD8+ T-cell polyfunctionality, proliferation, and cytotoxicity are not
restored by antiretroviral therapy. J. Virol. 83, 11876–11889. https://doi. 21. Mahnke, Y.D., Fletez-Brant, K., Sereti, I., and Roederer, M. (2016).
org/10.1128/JVI.01153-09. Reconstitution of peripheral T cells by tissue-derived CCR4+ central mem-
ory cells following HIV-1 antiretroviral therapy. Pathog. Immun. 1,
8. Rutishauser, R.L., and Trautmann, L. (2022). CD8 + T-cell responses in HIV
260–290. https://doi.org/10.20411/pai.v1i2.129.
controllers: potential implications for novel HIV remission strategies. Curr.
Opin. HIV AIDS 17, 315–324. https://doi.org/10.1097/COH.0000000000 22. Rutishauser, R.L., Deguit, C.D.T., Hiatt, J., Blaeschke, F., Roth, T.L.,
000748. Wang, L., Raymond, K.A., Starke, C.E., Mudd, J.C., Chen, W., et al.
(2021). TCF-1 regulates HIV-specific CD8+ T cell expansion capacity.
9. Koup, R.A., Safrit, J.T., Cao, Y., Andrews, C.A., McLeod, G., Borkowsky,
JCI Insight 6, e136648. https://doi.org/10.1172/jci.insight.136648.
W., Farthing, C., and Ho, D.D. (1994). Temporal association of cellular im-
mune responses with the initial control of viremia in primary human immu- 23. Takata, H., Kakazu, J.C., Mitchell, J.L., Kroon, E., Colby, D.J., Sacdalan,
nodeficiency virus type 1 syndrome. J. Virol. 68, 4650–4655. C., Bai, H., Ehrenberg, P.K., Geretz, A., Buranapraditkun, S., et al.
(2022). Long-term antiretroviral therapy initiated in acute HIV infection pre-
10. Ndhlovu, Z.M., Kamya, P., Mewalal, N., Kløverpris, H.N., Nkosi, T.,
vents residual dysfunction of HIV-specific CD8(+) T cells. EBioMedicine.
Pretorius, K., Laher, F., Ogunshola, F., Chopera, D., Shekhar, K., et al.
EBioMedicine 84, 104253. https://doi.org/10.1016/j.ebiom.2022.104253.
(2015). Magnitude and kinetics of CD8+ T cell activation during hyperacute
HIV infection impact viral set point. Immunity 43, 591–604. https://doi.org/ 24. Ho, Y.C., Shan, L., Hosmane, N.N., Wang, J., Laskey, S.B., Rosenbloom,
10.1016/j.immuni.2015.08.012. D.I., Lai, J., Blankson, J.N., Siliciano, J.D., and Siliciano, R.F. (2013).
Replication-competent noninduced proviruses in the latent reservoir in-
11. Takata, H., Buranapraditkun, S., Kessing, C., Fletcher, J.L., Muir, R.,
crease barrier to HIV-1 cure. Cell 155, 540–551. https://doi.org/10.1016/
Tardif, V., Cartwright, P., Vandergeeten, C., Bakeman, W., Nichols, C.N.,
j.cell.2013.09.020.
et al. (2017). Delayed differentiation of potent effector CD8+ T cells
reducing viremia and reservoir seeding in acute HIV infection. Sci. 25. Pollack, R.A., Jones, R.B., Pertea, M., Bruner, K.M., Martin, A.R., Thomas,
Transl. Med. 9, eaag1809. https://doi.org/10.1126/scitranslmed.aag1809. A.S., Capoferri, A.A., Beg, S.A., Huang, S.H., Karandish, S., et al. (2017).
12. Appay, V., Nixon, D.F., Donahoe, S.M., Gillespie, G.M., Dong, T., King, A., Defective HIV-1 proviruses are expressed and can be recognized by cyto-
Ogg, G.S., Spiegel, H.M., Conlon, C., Spina, C.A., et al. (2000). HIV-spe- toxic T lymphocytes, which shape the proviral landscape. Cell Host
cific CD8(+) T cells produce antiviral cytokines but are impaired in cytolytic Microbe 21, 494–506.e4. https://doi.org/10.1016/j.chom.2017.03.008.
function. J. Exp. Med. 192, 63–75. https://doi.org/10.1084/jem.192.1.63. 26. Imamichi, H., Smith, M., Adelsberger, J.W., Izumi, T., Scrimieri, F.,
13. Day, C.L., Kaufmann, D.E., Kiepiela, P., Brown, J.A., Moodley, E.S., Sherman, B.T., Rehm, C.A., Imamichi, T., Pau, A., Catalfamo, M., et al.
Reddy, S., Mackey, E.W., Miller, J.D., Leslie, A.J., DePierres, C., et al. (2020). Defective HIV-1 proviruses produce viral proteins. Proc. Natl.
(2006). PD-1 expression on HIV-specific T cells is associated with T-cell Acad. Sci. USA 117, 3704–3710. https://doi.org/10.1073/pnas.
exhaustion and disease progression. Nature 443, 350–354. https://doi. 1917876117.
org/10.1038/nature05115. 27. Thomas, A.S., Jones, K.L., Gandhi, R.T., McMahon, D.K., Cyktor, J.C.,
14. Trautmann, L., Janbazian, L., Chomont, N., Said, E.A., Gimmig, S., Chan, D., Huang, S.H., Truong, R., Bosque, A., Macedo, A.B., et al.
Bessette, B., Boulassel, M.R., Delwart, E., Sepulveda, H., Balderas, (2017). T-cell responses targeting HIV Nef uniquely correlate with infected
R.S., et al. (2006). Upregulation of PD-1 expression on HIV-specific cell frequencies after long-term antiretroviral therapy. PLoS Pathog. 13,
CD8+ T cells leads to reversible immune dysfunction. Nat. Med. 12, e1006629. https://doi.org/10.1371/journal.ppat.1006629.
1198–1202. https://doi.org/10.1038/nm1482. 28. Stevenson, E.M., Ward, A.R., Truong, R., Thomas, A.S., Huang, S.H.,
15. Trautmann, L., Mbitikon-Kobo, F.M., Goulet, J.P., Peretz, Y., Shi, Y., Van Dilling, T.R., Terry, S., Bui, J.K., Mota, T.M., Danesh, A., et al. (2021).
Grevenynghe, J., Procopio, F.A., Boulassel, M.R., Routy, J.P., Chomont, HIV-specific T cell responses reflect substantive in vivo interactions with
N., et al. (2012). Profound metabolic, functional, and cytolytic differences antigen despite long-term therapy. JCI Insight 6, e142640. https://doi.
characterize HIV-specific CD8 T cells in primary and chronic HIV infection. org/10.1172/jci.insight.142640.
Blood 120, 3466–3477. https://doi.org/10.1182/blood-2012-04-422550. 29. Huang, S.H., Ren, Y., Thomas, A.S., Chan, D., Mueller, S., Ward, A.R.,
16. Rehr, M., Cahenzli, J., Haas, A., Price, D.A., Gostick, E., Huber, M., Karrer, Patel, S., Bollard, C.M., Cruz, C.R., Karandish, S., et al. (2018). Latent
U., and Oxenius, A. (2008). Emergence of polyfunctional CD8+ T cells after HIV reservoirs exhibit inherent resistance to elimination by CD8+ T cells.
prolonged suppression of human immunodeficiency virus replication by J. Clin. Invest. 128, 876–889. https://doi.org/10.1172/JCI97555.

1504 Cell Host & Microbe 31, 1494–1506, September 13, 2023
ll
Article OPEN ACCESS

30. Sun, W., Gao, C., Hartana, C.A., Osborn, M.R., Einkauf, K.B., Lian, X., 43. Kratchmarov, R., Magun, A.M., and Reiner, S.L. (2018). TCF1 expression
Bone, B., Bonheur, N., Chun, T.W., Rosenberg, E.S., et al. (2023). marks self-renewing human CD8(+) T cells. Blood Adv. 2, 1685–1690.
Phenotypic signatures of immune selection in HIV-1 reservoir cells. https://doi.org/10.1182/bloodadvances.2018016279.
Nature 614, 309–317. https://doi.org/10.1038/s41586-022-05538-8. 44. Khan, O., Giles, J.R., McDonald, S., Manne, S., Ngiow, S.F., Patel, K.P.,
31. Leyre, L., Kroon, E., Vandergeeten, C., Sacdalan, C., Colby, D.J., Werner, M.T., Huang, A.C., Alexander, K.A., Wu, J.E., et al. (2019). TOX
Buranapraditkun, S., Schuetz, A., Chomchey, N., de Souza, M., transcriptionally and epigenetically programs CD8(+) T cell exhaustion.
Bakeman, W., et al. (2020). Abundant HIV-infected cells in blood and tis- Nature 571, 211–218. https://doi.org/10.1038/s41586-019-1325-x.
sues are rapidly cleared upon ART initiation during acute HIV infection.
45. Alfei, F., Kanev, K., Hofmann, M., Wu, M., Ghoneim, H.E., Roelli, P.,
Sci. Transl. Med. 12, eaav3491. https://doi.org/10.1126/scitranslmed.
Utzschneider, D.T., von Hoesslin, M., Cullen, J.G., Fan, Y., et al. (2019).
aav3491.
TOX reinforces the phenotype and longevity of exhausted T cells in chronic
32. Massanella, M., Bender Ignacio, R.A.B., Lama, J.R., Pagliuzza, A., viral infection. Nature 571, 265–269. https://doi.org/10.1038/s41586-019-
Dasgupta, S., Alfaro, R., Rios, J., Ganoza, C., Pinto-Santini, D., Gilada, 1326-9.
T., et al. (2021). Long-term effects of early antiretroviral initiation on HIV
46. Scott, A.C., Du€ndar, F., Zumbo, P., Chandran, S.S., Klebanoff, C.A.,
reservoir markers: a longitudinal analysis of the MERLIN clinical study.
Shakiba, M., Trivedi, P., Menocal, L., Appleby, H., Camara, S., et al.
Lancet Microbe 2, e198–e209. https://doi.org/10.1016/S2666-5247(21)
(2019). TOX is a critical regulator of tumour-specific T cell differentiation.
00010-0.
Nature 571, 270–274. https://doi.org/10.1038/s41586-019-1324-y.
33. Ananworanich, J., Fletcher, J.L., Pinyakorn, S., van Griensven, F.,
47. Sekine, T., Perez-Potti, A., Nguyen, S., Gorin, J.B., Wu, V.H., Gostick, E.,
Vandergeeten, C., Schuetz, A., Pankam, T., Trichavaroj, R., Akapirat, S.,
Llewellyn-Lacey, S., Hammer, Q., Falck-Jones, S., Vangeti, S., et al.
Chomchey, N., et al. (2013). A novel acute HIV infection staging system
(2020). TOX is expressed by exhausted and polyfunctional human effector
based on 4th generation immunoassay. Retrovirology 10, 56. https://doi.
memory CD8(+) T cells. Sci. Immunol. 5, eaba7918. https://doi.org/10.
org/10.1186/1742-4690-10-56.
1126/sciimmunol.aba7918.
34. Ananworanich, J., Sacdalan, C.P., Pinyakorn, S., Chomont, N., de Souza,
48. Yao, C., Sun, H.W., Lacey, N.E., Ji, Y., Moseman, E.A., Shih, H.Y.,
M., Luekasemsuk, T., Schuetz, A., Krebs, S.J., Dewar, R., Jagodzinski, L.,
Heuston, E.F., Kirby, M., Anderson, S., Cheng, J., et al. (2019). Single-
et al. (2016). Virological and immunological characteristics of HIV-infected
cell RNA-seq reveals TOX as a key regulator of CD8(+) T cell persistence
individuals at the earliest stage of infection. J. Virus Erad. 2, 43–48.
in chronic infection. Nat. Immunol. 20, 890–901. https://doi.org/10.1038/
35. Schuetz, A., Deleage, C., Sereti, I., Rerknimitr, R., Phanuphak, N., Phuang- s41590-019-0403-4.
Ngern, Y., Estes, J.D., Sandler, N.G., Sukhumvittaya, S., Marovich, M.,
49. Chen, Z., Ji, Z., Ngiow, S.F., Manne, S., Cai, Z., Huang, A.C., Johnson, J.,
et al. (2014). Initiation of ART during early acute HIV infection preserves
Staupe, R.P., Bengsch, B., Xu, C., et al. (2019). TCF-1-centered transcrip-
mucosal Th17 function and reverses HIV-related immune activation.
tional network drives an effector versus exhausted CD8 T cell-fate deci-
PLoS Pathog. 10, e1004543. https://doi.org/10.1371/journal.ppat.
sion. Immunity 51, 840–855.e5. https://doi.org/10.1016/j.immuni.2019.
1004543.
09.013.
36. Wolfl, M., Kuball, J., Ho, W.Y., Nguyen, H., Manley, T.J., Bleakley, M., and
Greenberg, P.D. (2007). Activation-induced expression of CD137 permits 50. Chomont, N., Okoye, A.A., Favre, D., and Trautmann, L. (2018). Wake me
detection, isolation, and expansion of the full repertoire of CD8+ T cells re- up before you go: a strategy to reduce the latent HIV reservoir. AIDS 32,
sponding to antigen without requiring knowledge of epitope specificities. 293–298. https://doi.org/10.1097/QAD.0000000000001695.
Blood 110, 201–210. https://doi.org/10.1182/blood-2006-11-056168. 51. Lafeuillade, A., Chollet, L., Hittinger, G., Profizi, N., Costes, O., and Poggi,
37. Reiss, S., Baxter, A.E., Cirelli, K.M., Dan, J.M., Morou, A., Daigneault, A., C. (1998). Residual human immunodeficiency virus type 1 RNA in lymphoid
Brassard, N., Silvestri, G., Routy, J.P., Havenar-Daughton, C., et al. (2017). tissue of patients with sustained plasma RNA of <200 copies/mL. J. Infect.
Comparative analysis of activation induced marker (AIM) assays for sensi- Dis. 177, 235–238. https://doi.org/10.1086/517362.
tive identification of antigen-specific CD4 T cells. PLoS One 12, e0186998. 52. Gu€nthard, H.F., Havlir, D.V., Fiscus, S., Zhang, Z.Q., Eron, J., Mellors, J.,
https://doi.org/10.1371/journal.pone.0186998. Gulick, R., Frost, S.D., Brown, A.J., Schleif, W., et al. (2001). Residual hu-
38. Avettand-Fènoe €l, V., Hocqueloux, L., Ghosn, J., Cheret, A., Frange, P., man immunodeficiency virus (HIV) Type 1 RNA and DNA in lymph nodes
Melard, A., Viard, J.P., and Rouzioux, C. (2016). Total HIV-1 DNA, a marker and HIV RNA in genital secretions and in cerebrospinal fluid after suppres-
of viral reservoir dynamics with clinical implications. Clin. Microbiol. Rev. sion of viremia for 2 years. J. Infect. Dis. 183, 1318–1327. https://doi.org/
29, 859–880. https://doi.org/10.1128/CMR.00015-16. 10.1086/319864.

39. Ananworanich, J., Chomont, N., Eller, L.A., Kroon, E., Tovanabutra, S., 53. Siddiqui, I., Schaeuble, K., Chennupati, V., Fuertes Marraco, S.A.,
Bose, M., Nau, M., Fletcher, J.L.K., Tipsuk, S., Vandergeeten, C., et al. Calderon-Copete, S., Pais Ferreira, D., Carmona, S.J., Scarpellino, L.,
(2016). HIV DNA set point is rapidly established in acute HIV infection Gfeller, D., Pradervand, S., et al. (2019). Intratumoral Tcf1(+)PD-1(+)
and dramatically reduced by early ART. EBioMedicine 11, 68–72. CD8(+) T cells with stem-like properties promote tumor control in response
https://doi.org/10.1016/j.ebiom.2016.07.024. to vaccination and checkpoint blockade immunotherapy. Immunity 50,
195–211.e10. https://doi.org/10.1016/j.immuni.2018.12.021.
40. Buranapraditkun, S., Hempel, U., Pitakpolrat, P., Allgaier, R.L.,
Thantivorasit, P., Lorenzen, S.I., Sirivichayakul, S., Hildebrand, W.H., 54. Kurtulus, S., Madi, A., Escobar, G., Klapholz, M., Nyman, J., Christian, E.,
Altfeld, M., Brander, C., et al. (2011). A novel immunodominant CD8+ Pawlak, M., Dionne, D., Xia, J., Rozenblatt-Rosen, O., et al. (2019).
T cell response restricted by a common HLA-C allele targets a conserved Checkpoint blockade immunotherapy induces dynamic changes in PD-
region of Gag HIV-1 clade CRF01_AE infected Thais. PLoS One 6, e23603. 1(-)CD8(+) tumor-infiltrating T cells. Immunity 50, 181–194.e6. https://
https://doi.org/10.1371/journal.pone.0023603. doi.org/10.1016/j.immuni.2018.11.014.
41. Zhou, X., Yu, S., Zhao, D.M., Harty, J.T., Badovinac, V.P., and Xue, H.H. 55. Sade-Feldman, M., Yizhak, K., Bjorgaard, S.L., Ray, J.P., de Boer, C.G.,
(2010). Differentiation and persistence of memory CD8(+) T cells depend Jenkins, R.W., Lieb, D.J., Chen, J.H., Frederick, D.T., Barzily-Rokni, M.,
on T cell factor 1. Immunity 33, 229–240. https://doi.org/10.1016/j.im- et al. (2018). Defining T cell states associated with response to checkpoint
muni.2010.08.002. immunotherapy in melanoma. Cell 175, 998–1013.e20. https://doi.org/10.
42. Wieland, D., Kemming, J., Schuch, A., Emmerich, F., Knolle, P., Neumann- 1016/j.cell.2018.10.038.
Haefelin, C., Held, W., Zehn, D., Hofmann, M., and Thimme, R. (2017). 56. Achenbach, C.J., Assoumou, L., Deeks, S.G., Wilkin, T.J., Berzins, B.,
TCF1(+) hepatitis C virus-specific CD8(+) T cells are maintained after Casazza, J.P., Lambert-Niclot, S., Koup, R.A., Costagliola, D., Calvez,
cessation of chronic antigen stimulation. Nat. Commun. 8, 15050. V., et al. (2015). Effect of therapeutic intensification followed by HIV DNA
https://doi.org/10.1038/ncomms15050. prime and rAd5 boost vaccination on HIV-specific immunity and HIV

Cell Host & Microbe 31, 1494–1506, September 13, 2023 1505
ll
OPEN ACCESS Article
reservoir (EraMune 02): a multicentre randomised clinical trial. Lancet HIV sons durably suppressed in Fiebig I acute HIV infection. Nat. Med. 24,
2, e82–e91. https://doi.org/10.1016/S2352-3018(15)00026-0. 923–926. https://doi.org/10.1038/s41591-018-0026-6.
57. Rosás-Umbert, M., Gunst, J.D., Pahus, M.H., Olesen, R., Schleimann, M., 62. Gantner, P., Buranapraditkun, S., Pagliuzza, A., Dufour, C., Pardons, M.,
Denton, P.W., Ramos, V., Ward, A., Kinloch, N.N., Copertino, D.C., et al. Mitchell, J.L., Kroon, E., Sacdalan, C., Tulmethakaan, N., Pinyakorn, S.,
(2022). Administration of broadly neutralizing anti-HIV-1 antibodies at et al. (2023). HIV rapidly targets a diverse pool of CD4(+) T cells to establish
ART initiation maintains long-term CD8(+) T cell immunity. Nat. productive and latent infections. Immunity 56, 653–668.e5. https://doi.
Commun. 13, 6473. https://doi.org/10.1038/s41467-022-34171-2. org/10.1016/j.immuni.2023.01.030.
58. Shankarappa, R., Margolick, J.B., Gange, S.J., Rodrigo, A.G., Upchurch,
63. Papasavvas, E., Azzoni, L., Ross, B.N., Fair, M., Yuan, Z., Gyampoh, K.,
D., Farzadegan, H., Gupta, P., Rinaldo, C.R., Learn, G.H., He, X., et al.
Mackiewicz, A., Sciorillo, A.C., Pagliuzza, A., Lada, S.M., et al. (2021).
(1999). Consistent viral evolutionary changes associated with the progres-
Intact human immunodeficiency virus (HIV) reservoir estimated by the
sion of human immunodeficiency virus type 1 infection. J. Virol. 73, 10489–
intact proviral DNA assay correlates with levels of total and integrated
10502. https://doi.org/10.1128/JVI.73.12.10489-10502.1999.
DNA in the blood during suppressive antiretroviral therapy. Clin. Infect.
59. Besson, G.J., Lalama, C.M., Bosch, R.J., Gandhi, R.T., Bedison, M.A.,
Dis. 72, 495–498. https://doi.org/10.1093/cid/ciaa809.
Aga, E., Riddler, S.A., McMahon, D.K., Hong, F., and Mellors, J.W.
(2014). HIV-1 DNA decay dynamics in blood during more than a decade 64. Vandergeeten, C., Fromentin, R., Merlini, E., Lawani, M.B., DaFonseca, S.,
of suppressive antiretroviral therapy. Clin. Infect. Dis. 59, 1312–1321. Bakeman, W., McNulty, A., Ramgopal, M., Michael, N., Kim, J.H., et al.
https://doi.org/10.1093/cid/ciu585. (2014). Cross-clade ultrasensitive PCR-based assays to measure HIV
60. Peluso, M.J., Bacchetti, P., Ritter, K.D., Beg, S., Lai, J., Martin, J.N., Hunt, persistence in large-cohort studies. J. Virol. 88, 12385–12396. https://
P.W., Henrich, T.J., Siliciano, J.D., Siliciano, R.F., et al. (2020). Differential doi.org/10.1128/JVI.00609-14.
decay of intact and defective proviral DNA in HIV-1-infected individuals on 65. Fiebig, E.W., Wright, D.J., Rawal, B.D., Garrett, P.E., Schumacher, R.T.,
suppressive antiretroviral therapy. JCI Insight 5, e132997. https://doi.org/ Peddada, L., Heldebrant, C., Smith, R., Conrad, A., Kleinman, S.H., and
10.1172/jci.insight.132997. Busch, M.P. (2003). Dynamics of HIV viremia and antibody seroconversion
61. Colby, D.J., Trautmann, L., Pinyakorn, S., Leyre, L., Pagliuzza, A., Kroon, in plasma donors: implications for diagnosis and staging of primary
E., Rolland, M., Takata, H., Buranapraditkun, S., Intasan, J., et al. (2018). HIV infection. AIDS 17, 1871–1879. https://doi.org/10.1097/00002030-
Rapid HIV RNA rebound after antiretroviral treatment interruption in per- 200309050-00005.

1506 Cell Host & Microbe 31, 1494–1506, September 13, 2023
ll
Article OPEN ACCESS

STAR+METHODS

KEY RESOURCES TABLE

REAGENT or RESOURCE SOURCE IDENTIFIER


Antibodies
BV510 anti-human CD14 (clone M5E2) BioLegend Cat# 301842; RRID: AB_2561946
BV510 anti-human CD19 (clone SJ25C1) BioLegend Cat# 363020; RRID: AB_2564229
PE Cy5 anti-human CD127 (clone A019D5) BioLegend Cat# 351324; RRID: AB_10915554
Pacific Blue labelled anti-human IFN-g BioLegend Cat# 502522; RRID: AB_893525
(clone 4S.B3)
APC labelled anti-human 4-1BB BioLegend Cat# 309810; RRID: AB_830672
(clone 4B4-1)
Alexa Fluor 488 anti-human CD69 BioLegend Cat# 310916; RRID: AB_528869
(clone FN50)
APC Fire810 anti-human CD4 (clone SK3) BioLegend Cat# 344662; RRID: AB_2860884
Brilliant Violet 785 anti-human CD8 (clone BioLegend Cat# 301046; RRID: AB_2563264
RPA-T8)
PE labelled anti-human TCF-1 (clone BioLegend Cat# 655208; RRID: AB_2728492
7F11A10)
BV510 anti-human CD3 (clone UCHT 1) BD Biosciences Cat# 566105; RRID: AB_2739563
BUV496 anti-human CD8 (clone RPA-T8) BD Biosciences Cat# 612942; RRID: AB_2870223
BUV563 anti-human CD4 (clone SK3) BD Biosciences Cat# 612912; RRID: AB_2739451
BUV737 anti-human PD-1 (clone EH12.1) BD Biosciences Cat# 612791; RRID: AB_2870118
Alexa Fluor 488 anti-TOX Cell Signaling Technology Cat# 44682S
BD FastImmune Co-Stimulatory Antibodies BD Biosciences Cat# 347690; RRID: AB_647457
(CD28/CD49d)
Biological samples
Human peripheral blood mononuclear U.S. Military HIV Research Program RV254/ SEARCH010
cells (PBMCs)
Human peripheral blood mononuclear U.S. Military HIV Research Program RV304/ SEARCH013
cells (PBMCs)
Chemicals, peptides, and recombinant proteins
BD GolgiPlug Protein Transport Inhibitor BD Biosciences Cat# 555029
(Brefeldin A)
BD GolgiStop Protein Transport Inhibitor BD Biosciences Cat# 554724
(Monensin)
BD Perm/Wash Perm/Wash Buffer BD Biosciences Cat# 554723
BD Horizon Brilliant Stain Buffer BD Biosciences Cat# 566349
BD Horizon Brilliant Stain Buffer Plus BD Biosciences Cat# 566385
Foxp3 / Transcription Factor Staining Thermo Fisher Scientific Cat# 00-5523-00
Buffer Set
LIVE/DEAD Fixable Aqua Dead Cell Thermo Fisher Scientific Cat# L34957
Stain Kit
LIVE/DEAD Fixable Yellow Dead Cell Thermo Fisher Scientific Cat# L34959
Stain Kit
BV650 Streptavidin BioLegend Cat# 405232
HIV-1 CRF01_AE Fifteen-mer peptides Anaspec N/A, Custom
Critical commercial assays
EasySep Human CD4+ T Cell STEMCELL Technologies Cat# 19052
Enrichment Kit
QuantiNova Probe PCR Kit QIAGEN Cat# 80224
(Continued on next page)

Cell Host & Microbe 31, 1494–1506.e1–e4, September 13, 2023 e1


ll
OPEN ACCESS Article

Continued
REAGENT or RESOURCE SOURCE IDENTIFIER
SuperScript III One-Step RT-PCR System Invitrogen (ThermoFisher) Cat# 12574026
with Platinum Taq DNA Polymerase
AllPrep DNA/RNA/miRNA Universal KIT QIAGEN Cat# 80224
MEGAscript T7 Transcription Kit Ambion Cat# AMB13345
Oligonucleotides
ULF1 (Forward Pre-PCR): 5’- ATG CCA Vandergeeten et al.64 N/A
CGT AAG CGA AAC TCT GGG TCT CTC
TDG TTA GAC-3’
UR1 (Reverse Pre-PCR): 5’-CCA TCT CTC Vandergeeten et al.64 N/A
TCC TTC TAG C-3’
Lambda (l) T (Forward Post-PCR): 5’-ATG Vandergeeten et al.64 N/A
CCA CGT AAG CGA AAC T-3’
UR2 (Reverse Post-PCR): 5’-CTG AGG Vandergeeten et al.64 N/A
GAT CTC TAG TTA CC-3’
Software and algorithms
FlowJo v10.8.1 FlowJo, LLC N/A
Prism 9 GraphPad Software, LLC. N/A
Other
Northern Lights 3000 cytometer Cytek Biosciences NL-3000
In vitro synthetized RNA transcript This paper N/A
Rotor-Gene Q QIAGEN Cat# 9001620

RESOURCE AVAILABILITY

Lead contact
Further information and requests for resources and reagents should be directed to and will be fulfilled by the lead contact, Lydie
Trautmann (ltrautmann@global-id.org).

Materials availability
This study did not generate new unique reagents.

Data and code availability


Any additional information required to reanalyze the data reported in this paper is available from the lead contact upon request. This
paper does not report original code.

EXPERIMENTAL MODEL AND STUDY PARTICIPANT DETAILS

Study design
Forty-eight people with HIV (PWH) from the RV254/ SEARCH010 study who initiated ART during AHI and 18 participants who
initiated ART during CHI from the RV304/ SEARCH013 study, all conducted in Bangkok, Thailand, were included in these
analyses (Table 1). In the RV254/ SEARCH010 study, samples from HIV test clients were tested by fourth- and third-generation
immunoassays and HIV nucleic acid test. The participants were classified into Fiebig I to V based on HIV nucleic acid test,
p24 antigen, and third-generation and Western blot immunoassays as previously described.33,65 For all of the experiments, lab
investigators were blinded to the participant ID and HIV infection stage at ART initiation (AHI stages or CHI) of specimens during
procedures so as not to bias measurements. The specimen information was recovered to perform statistical analyses. Outliers
were not excluded from the analyses. All participants were pre-screened for HLA-A1101/ Cw0102 restricted HIV-specific CD8+
T cells.

Study approval
All study participants provided written informed consent approved by the Chulalongkorn University and Walter Reed Army Institute of
Research institutional review boards.

e2 Cell Host & Microbe 31, 1494–1506.e1–e4, September 13, 2023


ll
Article OPEN ACCESS

METHOD DETAILS

Antibodies and Reagents for Flow cytometry analysis


BV510 labelled anti-CD14, BV510 labelled anti-CD19, PE Cy5 labelled anti-CD127, Pacific Blue labelled anti-IFN-g, APC labelled
anti-4-1BB, Alexa 488 labelled anti-CD69, APC Fire810 labelled anti-CD4, BV785 labelled anti-CD8, and PE labelled anti-TCF-1
came from Biolegend. BV480 labelled anti-CD3, BUV496 labelled anti-CD8, BUV563 labelled anti-CD4, BUV737 labelled anti-
PD-1, GolgiPlug, GolgiStop, Perm Wash Buffer, Brilliant Stain Buffer, and Brilliant Stain Buffer Plus came from BD Biosciences. Alexa
Fluor 488 labelled anti-TOX came from Cell Signaling Technology. LIVE/DEAD Fixable Dead Cell Stain Kits (Aqua and Yellow) and
FoxP3 / Transcription Factor Staining Buffer Set came from Thermo Fisher Scientific. HLA-A*1101 HIV-1 NEF (GAFDLSFFLK and
QVPLRPMTYK), HLA-A*1101 HIV-1 POL (AIFQSSMTKIL), HLA-Cw*0102 HIV-1 GAG (YSPVSILDI), and HLA-Cw*0102 HIV-1 ENV
(CTPAGYAIL) soluble biotinylated monomers were produced at NIH Tetramer Core Facility as previously described53 and tetramer-
ized with BV650-labeled Streptavidin (BioLegend).

HIV peptide pools for intracellular cytokine staining


A large collection of 1,057 overlapping peptides spanning the entire HIV proteome were designed based on available HIV sequence
alignments derived from the SEARCH10/RV254 and RV144 Thai cohorts and encompassing the consensus sequence and the most
frequent HIV variants. The detailed description of the 1,057 peptides is provided in Table S1. Fifteen-mer peptides, overlapping by 11
amino acids, were synthesized by Anaspec (Fremont, CA). Lyophilized peptides were reconstituted in dimethyl sulfoxide (DMSO) at
10mg/ml, aliquoted and stored at -80C. Single peptides were pooled in 13 pools corresponding to Nef (unique pool, n=91 peptides),
Gag (2 pools, n= 72 peptides each pool), Pol (3 pools, n=92 peptides each pool), Rev/Tat (unique pool, n=89 peptides), Vif (unique
pool, n=71 peptides), Vpu/Vpr (unique pool, n=70 peptides), Env (4 pools, 2 pools including n=80 peptides, 2 pools including n=78
peptides).

Ex vivo detection of HIV-specific CD8+ and CD4+ T cells by intracellular cytokine staining
Thawed peripheral blood mononuclear cells (PBMCs) from PWH were plated at 2x106 cells/well in 96 well deep well plate in RPMI
1640 media containing 10% FBS and Penicillin/Streptomycin (R10 media). The cells were stimulated with HIV peptide pools (1 mg of
each peptide/mL) in the presence of anti-CD28/49d (0.5 mg/mL) in R10 media and incubated for 1 hour at 37 C. The same amount of
DMSO as the HIV peptide pool was added for a negative control. GolgiPlug (1 mL/mL) and GolgiStop (0.5 mL/mL) were added to each
well and the cells were incubated at 37 C overnight (16 hrs).

Flow cytometry analysis


For analysis of cytokine producing T cells, peptide stimulated PBMCs were first stained with LIVE/DEAD Aqua for 10 minutes at room
temperature. Cells were stained for other cell surface markers in Brilliant Stain Buffer at 4 C for 20 minutes followed by 2 times wash
with PBS containing 2% FBS (washing buffer). Cells were fixed in PBS containing 2% formaldehyde (fix buffer) for 20 minutes at room
temperature. Cells were washed and resuspended in Perm/Wash buffer for 15 minutes at room temperature. Cells were stained for
intracellular cytokine markers in Perm/Wash buffer at room temperature for 30 minutes, followed by 2 washes with washing buffer
and then fixation in fix buffer. Fixed cells were analyzed using a Northern Lights 3000 cytometer (Cytek Biosciences). The frequency
of CD69+4-1BB+IFN-g+ HIV-specific CD8+ T cells in total CD8+ T cells and CD69+CD40L+IFN-g+ HIV-specific CD4+ T cells in total
CD4+ T cells was reported after subtracting the frequency of those cells in the culture with DMSO only.
For phenotypic analysis with peptide-MHC class I complex tetramer (pMHC tetramer), thawed PBMCs were first stained for pMHC
tetramer, cell surface markers, and then fixed/permeabilized for intracellular/intranuclear staining with FoxP3 / Transcription Factor
Staining Buffer Set as previously described.11,23
All flow cytometry data was analyzed with FlowJo v10.8.1 (FlowJo, LLC)

HIV DNA and HIV RNA measures


The frequencies of CD4+ T cells harboring integrated HIV DNA was measured by real time nested PCR using Alu primers together with
a primer annealing to the HIV LTR in purified CD4+ T cells obtained by negative magnetic selection (StemCell), as previously
described.64 Serial dilutions of ACH2 cells carrying a single copy of the HIV genome were used as a standard.
To measure total HIV DNA and HIV RNA, cellular DNA and RNA were co-extracted from isolated CD4+ T cells, using the AllPrep
DNA/RNA Mini Kit (Qiagen) according to the manufacturer’s instructions. The frequency of cells harbouring total HIV DNA (LTR-gag)
was quantified by real-time PCR on the extracted genomic DNA, as previously described.64 Serial dilutions of ACH2 cells were used
as a standard. Cell-associated RNA (LTR-gag) was quantified by real-time nested RT-PCR, using the same primers as in the DNA
assay. In a first PCR reaction, RNA were reverse-transcribed and pre-amplified using the Superscript III one-step RT-PCR System
(ThermoFisher) in a 25mL reaction containing 0.5mM of each primers, and 8.5mL of extracted RNA. The reverse transcription was per-
formed at 50 C for 30min, followed by a denaturation step at 94 C for 2min, and 16 cycles of: 15s at 94 C, 30s at 55 C, 1min at 68 C,
with a final elongation step of 5min at 68 C. For the nested PCR, the pre-amplified cDNA was amplified by real-time PCR on a Rotor-
Gene Q instrument, using the same protocol than for the HIV DNA PCR.64 In vitro transcribed RNA (generated from plasmids with

Cell Host & Microbe 31, 1494–1506.e1–e4, September 13, 2023 e3


ll
OPEN ACCESS Article

MEGAscript T7 Transcription Kit, ThermoFisher) were used as a standard for absolute quantification. LTR-gag transcripts were
expressed as copies per 106 cells using the CD3 copy numbers measured in the HIV DNA assay or as copies per viral genome using
the total HIV DNA quantification results.
The sample detection limit for the cell-associated RNA assay is provided as Table S2. All real time nested PCR reactions were per-
formed in triplicate. The means of triplicates were used for data analysis.

Statistical analysis
Statistical analyses were performed using the nonparametric Mann-Whitney test (2 groups) for group comparisons, and the Wilcoxon
matched-pairs signed-rank test to compare CD8+ T cell responses or HIV reservoir measurements from same participant between
2 visits. The nonparametric Spearman test was used for all of the correlation analyses, and the Benjamini-Hochberg procedure was
used to correct for multiple comparisons (FDR < 0.1). P-values were adjusted by group (Tx AHI and Tx CHI) and pre-ART viral load,
using a multivariate linear regression model.

e4 Cell Host & Microbe 31, 1494–1506.e1–e4, September 13, 2023

You might also like