In people living with HIV on antiretroviral therapy, HIV latency is the major barrier to a cure. HIV persists preferentially in CD4+ T cells expressing multiple immune checkpoint (IC) molecules, including programmed death (PD)-1, T cell Ig and mucin domain-containing protein 3 (TIM-3), lymphocyte associated gene 3 (LAG-3), and T cell immunoreceptor with Ig and ITIM domains (TIGIT). We aimed to determine whether these and other IC molecules have a functional role in maintaining HIV latency and whether blocking IC molecules with Abs reverses HIV latency. Using an in vitro model that establishes latency in both nonproliferating and proliferating human CD4+ T cells, we show that proliferating cells express multiple IC molecules at high levels. Latent infection was enriched in proliferating cells expressing PD-1. In contrast, nonproliferating cells expressed IC molecules at significantly lower levels, but latent infection was enriched in cells expressing PD-1, TIM-3, CTL-associated protein 4 (CTLA-4), or B and T lymphocyte attenuator (BTLA). In the presence of an additional T cell–activating stimulus, staphylococcal enterotoxin B, Abs to CTLA-4 and PD-1 reversed HIV latency in proliferating and nonproliferating CD4+ T cells, respectively. In the absence of staphylococcal enterotoxin B, only the combination of Abs to PD-1, CTLA-4, TIM-3, and TIGIT reversed latency. The potency of latency reversal was significantly higher following combination IC blockade compared with other latency-reversing agents, including vorinostat and bryostatin. Combination IC blockade should be further explored as a strategy to reverse HIV latency.

Antiretroviral therapy (ART) has revolutionized the treatment of HIV infection and has dramatically reduced mortality and morbidity. However, ART is lifelong, expensive, and often has side effects, so there is an urgent need to identify strategies to cure HIV or induce “remission” to avoid lifelong treatment (1). The major barrier to a cure for HIV infection is the persistence of latent infection in long-lived resting and proliferating CD4+ T cells (25) that are more frequently detected in lymphoid tissue and the gastrointestinal tract (68). It is highly likely that the mechanisms maintaining HIV latency differ in nonproliferating and proliferating T cells, suggesting that multiple interventions may be needed to eliminate latency.

One approach being tested to eliminate latently infected cells in people living with HIV (PLWH) on ART is to activate latent virus and thereby induce death of the infected cell through immune clearance or virus-induced cytolysis (9, 10). To date, clinical trials that have examined latency-reversing agents (LRA), such as histone deacetylase inhibitors (HDACi), disulfiram, or TLR agonists, have shown modest latency reversal but without clearance of infected cells (1115). Furthermore, HDACi have been shown to induce adverse effects on adaptive immune function in vitro and have multiple off target effects (11, 1621). Therefore, alternative LRAs that are more potent and have a beneficial effect on adaptive immune function to enhance immune-mediated clearance of infected cells are needed.

Latent infection is enriched in CD4+ T cells expressing immune checkpoint (IC) molecules, first described for programmed death 1 (PD-1) in circulating CD4+ T cells in blood (22, 23), and more recently in T follicular helper cells in lymphoid tissue (7). In SIV-infected macaques on ART, there is also enrichment of virus in CD4+ T cells expressing CTL-associated protein 4 (CTLA-4) and PD-1 in the extrafollicular and follicular lymphoid compartments, respectively (24). We previously demonstrated that coexpression of multiple IC molecules, including PD-1, lymphocyte activation gene 3 (LAG-3), and T cell immunoreceptor with Ig and ITIM domains (TIGIT), on CD4+ T cells from blood from PLWH on ART were highly enriched for HIV infection compared with cells that expressed fewer than three IC markers (23).

HIV transcription and virus production is largely dependent on host transcription factors that increase and localize to the nucleus following T cell activation (reviewed in Ref. 1). Ligation of IC molecules can actively suppress these pathways (25). We recently demonstrated that engagement of PD-1 in vitro inhibits the establishment of HIV latency in resting CD4+ T cells (26), and in latently infected cells isolated from PLWH on ART, programmed death ligand (PD-L) 1 can block viral production at the transcriptional level by abrogating TCR-induced HIV reactivation (27). Conversely, PD-1 blockade with the mAb pembrolizumab in combination with the LRA bryostatin enhances HIV production ex vivo without increasing T cell activation (27). Together, these data suggest that IC molecules alone or in combination can define latently infected cells, and PD-1 has a functional role in the establishment, maintenance, and reversal of HIV latency in CD4+ T cells (reviewed in Ref. 28), in addition to the more global effects PD-1 might have on Ag-specific T cell function.

The administration of Abs to PD-1, PD-L1, or CTLA-4, either alone or in combination, are now licensed in many countries for the treatment of malignancy (29), and multiple small case series have demonstrated that the safety of anti–PD-1 and anti–PD-L1 in PLWH is similar to individuals without HIV infection (3037). Abs to other IC molecules, such LAG-3 and TIGIT, are also now in clinical development and early phase studies (38).

In a single case report of a person living with HIV on ART with metastatic melanoma, we demonstrated that the administration of anti–CTLA-4 (ipilimumab) led to a significant increase in cell-associated unspliced HIV RNA, potentially consistent with latency reversal (39). Additionally, in the same person, a single dose of anti–PD-1 (nivolumab) increased cell-associated HIV RNA in CD4+ T cells (26). Other case reports of anti–PD-1 (pembrolizumab) have not shown a change in cell-associated unspliced HIV RNA, although the timing of sampling blood differed in each of these studies (40). Finally, repeated dosing of anti–PD-1 has been associated with a decline in cell-associated HIV DNA, potentially consistent with clearance of infected cells (27, 41). We therefore hypothesized that IC molecules provide an attractive target to eliminate latent infection and that administration of Abs to multiple IC molecules in combination could synergistically reverse inhibitory signals required to maintain latency, thereby leading to latency reversal.

The HEK 293T and TZM-bl cell lines cells were grown as a monolayer DMEM (Life Technologies) supplemented with 10% (v/v) heat-inactivated FCS (Bovogen Biologicals, Keilor East, Australia), 100 U/ml penicillin, 100 μg/ml streptomycin, and 292 μg/ml glutamine (Life Technologies) at 37°C and 5% CO2.

PBMC were isolated by Ficoll-Paque density gradient centrifugation (GE Healthcare, Chalfont St. Giles, U.K.) from buffy coats obtained from the Australian Red Cross Blood Service (Melbourne, Australia). Resting CD4+ T cells were negatively selected using magnetic cell sorting with an in house mixture of Abs to CD8 (clone OKT8), CD11b (OKM1), CD16 (3G8), HLA-DR (2-06), CD19 (FMC63), CD14 (FMC17) and CD69 (L78), goat anti-mouse IgG magnetic beads (Miltenyi Biotec), and the AutoMACS Pro (Miltenyi Biotec), as previously described (4244). Purity (≥96%) was based on CD3+ and CD4+ expression with mouse anti-human(m-a-h)CD3-PE (HIT3a) and m-a-hCD4-FITC (M5E2; BD Biosciences).

Monocytes were positively selected from PBMC using magnetic cell sorting using CD14 Abs (FMC17), as previously described (45). Purity (≥98%) was based on CD14+ expression with m-a-hCD14-FITC (M5E2; BD Biosciences). Primary cells were cultured in RPMI 1640 (Life Technologies) supplemented with 10% (v/v) heat-inactivated FCS, 100 U/ml penicillin, 100 μg/ml streptomycin, and 292 μg/ml glutamine (RF10) at 37°C and 5% CO2.

Plasmid DNA encoding a NL4.3 backbone with an AD8 envelope and EGFP inserted one base downstream of the env open reading frame (46) followed by an IRES-nef coding sequence, kindly provided by Y. Tsunetsugu-Yokota (National Institute of Infectious Diseases, Tokyo, Japan), was transiently transfected into HEK 293T cells with calcium phosphate precipitation (47). Virus supernatant was harvested after 2 d, filtered by passage through a 0.2-μm filter, concentrated using 20% sucrose density ultracentrifugation, and stored in aliquots at −80°C. Cells were infected for 2–3 h at a multiplicity of infection of 0.5 as determined by limiting dilution on TZM-bl using the Reed and Muench method (48), followed by a wash step to remove unbound virus.

Resting CD4+ T cells were labeled with proliferation dye eFluor 670 (eBiosciences) per the manufacturer’s instructions. Labeled T cells were cultured with or without syngeneic-sorted monocytes (monocyte/T cell ratio of 1:10) with 20 ng/ml staphylococcal enterotoxin B (SEB; Sigma-Aldrich) and 10 U/ml recombinant human (r-h) IL-2 (Roche Diagnostics) for 24 h, infected with full length nef-competent EGFP reporter virus for 2–3 h, after which excess virus was washed away and cells were cultured for 5 d in media supplemented with SEB and 10 U/ml r-h IL-2. Productive infection was determined day 5 postinfection by detecting EGFP+ cells using flow cytometry. Subsequently, the nonproductively infected (EGFP), CD3+, HLA-DR (m-a-hCD3-PB, UCHT1, m-a-hHLA-DR-BV650, G46-6; from BD Biosciences) nonproliferating (eFluor 670HI) and proliferating (eFluor 670LO) CD4+ T cells were sorted using a FACS-Aria, MoFlo Astrios, or FACSAria Fusion cell sorters (BD Biosciences). In some experiments, the CD3+HLA-DREGFP nonproliferating and proliferating cells were further sorted based on IC expression using m-a-hCD279/PD-1-PE (EH12.1; BD Biosciences), m-a-hTIM-3-PE (F38-2E2; BioLegend), m-a-hCD152/CTLA-4-PE (BNI3; BD Biosciences), m-a-hCD272/BTLA-PE (MIH26; BioLegend), m-a-hTIGIT–eFluor 710 (MBSA43; eBioscience). In other experiments, nonproliferating and proliferating T cells were sorted based on the expression of any of the following molecules: PD-1, T cell Ig and mucin domain-containing protein 3 (TIM-3), CTLA-4, and B and T lymphocyte attenuator (BTLA). For these experiments, all four PE-conjugated Abs were added to the cells in combination with m-a-hCD45RA-V500 (Hl100; BD Biosciences) to exclude CD45RA+ from the nonproliferating cell population prior to IC gating.

To determine latent infection, 100,000–200,000 sorted T cells were cultured in a flat-bottom 96-well plate with 200 μl RF10 supplemented with integrase inhibitor L870812 (Merck, Whitehouse Station, NJ) or raltegravir (AIDS Research and Reference Reagent Program, Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health or SelleckChem), both used at a final concentration of 1 μM in the presence (positive control) or absence (negative control) of activation stimuli composed of 3 μg/ml plate-bound m-a-hCD3 (UCHT1; BD Biosciences), 2 μg/ml soluble m-a-hCD28 (L293; BD Biosciences), 50 ng/ml r-hIL-7 (Sigma-Aldrich) and 10 U/ml r-hIL-2. Cells were harvested 72 h after stimulation, and EGFP expression was quantified using the FACSCalibur or FACSCanto II (BD Biosciences). Results were analyzed using Weasel Software (Walter and Elisa Hall Institute of Medical Research, Melbourne, Australia), and to quantify latent infection, the number of EGFP+ cells in the unstimulated culture (background) was subtracted from the number of EGFP+ cells in the stimulated culture. In some experiments, Abs blocking IC molecules; humanized anti–PD-1 (nivolumab; a kind gift from Bristol-Myers Squibb, New York, NY), humanized anti–CTLA-4 (ipilimumab; a kind gift from Bristol-Myers Squibb), m-a-hTIM-3 (clone F38-2E2; BioLegend), m-a-hTIGIT (clone MBSA43; eBioscience), or isotype controls (hIgG4 [clone ET904; BioLegend], hIgG1 [clone ET901; BioLegend], mIgG1 [clone MOPC-21; BioLegend]), were added to activate latent infection. For these experiments 10 μg/ml IC blocker (ICB) or isotype control was added to sorted cells with back-added thawed syngeneic monocytes (monocyte/T cell ratio of 1:10) in the presence or absence of SEB and with the standard activation stimuli in a parallel culture (positive control) for 72 h. When multiple ICB were added, equal amounts of the appropriate isotype control Ab were added to the control culture.

To calculate the ICB-induced latency reversal expressed as a percentage of maximal stimulation with anti-CD3/CD28 plus IL-7 plus IL-2, the number of EGFP+ cells in the unstimulated culture was subtracted from the number of EGFP+ cells in the anti-CD3/CD28 stimulated culture, and this was set at 100% (maximal stimulation). To calculate the effect of ICB-induced reversing latency, the number of EGFP+ cells in the unstimulated control was subtracted from the number of EGFP+ cells in the ICB-treated culture and expressed as a percentage of maximal stimulation.

In other experiments, LRAs romidepsin (FK288, depsipeptide, 1 or 0.1 nM; SelleckChem), JQ1 (1 or 0.1 μM; SelleckChem), bryostatin (10 or 1 nM; Sigma-Aldrich), PMA plus ionomycin (2 nM and 0.5 μM, respectively; both from Sigma-Aldrich), or DMSO (as reconstitution solvent control; Sigma-Aldrich) were added to sorted cells with the standard activation stimuli in a parallel culture (positive control) for 72 h.

In all experiments, live and single cells were gated using forward and side scatter plots. IC ligand expression on myeloid DC and monocytes was determined on CD3HLA-DR+ cells using m-a-hCD3-PB (UCHT1; BD Biosciences), m-a-hHLA-DR-PerCP (L243; BD Biosciences), m-a-hCD274/PD-L1-PE (MIHI; BD Biosciences), m-a-hCD273/PD-L2-PE (MIH18; BD Biosciences), m-a-hCD80-PE (L307.4; BD Biosciences), m-a-hCD86-PE (2331; BD Biosciences), m-a-hCD275/ICOSLigand-PE (2D3/B7-H2; BD Biosciences), m-a-hGal9-PE (9M1-3; BioLegend), m-a-hCD112/Nectin-2/PVRL-2-PE (TX31; BioLegend), m-a-hCD155/PVR-PE (TX24; BioLegend), m-a-hCD270/HVEM-PE (eBioHVEM-122; eBioscience), or mIgG2a-PE isotype control (MOPC-173; BioLegend).

Single IC expression on T cells was determined by gating on CD3+HLA-DREGFP cells, which were subsequently separated into eFluor 670HI (nonproliferating) and eFluor 670LO (proliferating) populations using m-a-hCD3-PB (UCHT1; BD Biosciences), m-a-hHLA-DR-PE-TexR (TU36; BD Biosciences), m-a-hCD279/PD-1–PE (EH12.1; BD Biosciences), m-a-hTIM-3-PE (F38-2E2; BioLegend), m-a-hCD152/CTLA-4–PE (BNI3; BD Biosciences), m-a-hCD272/BTLA-PE (MIH26; BioLegend), m-a-hTIGIT–eFluor 710 (MBSA43; eBioscience), mIgG2a-PE isotype control (MOPC-173; BioLegend) or mIgG1–eFluor 710 isotype control (P36281; eBioscience).

Coexpression of PD-1, TIM-3, and TIGIT on CD3+HLA-DREGFP nonproliferating and proliferating cells was done using m-a-hCD3-PB, m-a-hHLA-DR-PE-TexR, or m-a-hHLA-DR-BV650 (G46-6; BD Biosciences), m-a-hCD279/PD-1–PE, m-a-hTIM-3-PE-Cy7 (F38-2E2; BioLegend), m-a-hTIGIT–eFluor 710, mIgG1–PE isotype control (MOPC-21; BD Biosciences), mIgG1-PE-Cy7 isotype control (MOPC-21; BioLegend) and mIgG1–eFluor 710 isotype control.

Coexpression of PD-1, CTLA-4, TIM-3, and TIGIT on CD3+HLA-DREGFP nonproliferating and proliferating cells was done using m-a-hCD3-BV711 (UCHT1; BD Biosciences), m-a-hHLA-DR-BV650, m-a-hCD279/PD-1–BV421 (EH12.1; BD Biosciences), m-a-hCD152/CTLA-4–PE, m-a-hTIM-3-PE-Cy7 and m-a-hTIGIT–eFluor 710.

The percentage of positive cells was calculated by subtracting fluorescence minus one (FMO) or isotype control values.

The use of blood samples from HIV-negative donors for this study was approved by the Human Research and Ethics Committees from the Alfred Hospital (HREC156/11), Monash University (CF11/1888) and the University of Melbourne (1443071). Donors were recruited by the Red Cross Blood Transfusion Service, and all provided written informed consent for the use of their blood products for the research.

Differences between experimental conditions were analyzed using Wilcoxon matched-pairs signed-rank test (n > 5) or paired Student t test (n ≤ 5) on GraphPad Prism (version 6). All p values ≤ 0.05 were considered significant.

To study the role of IC molecules in HIV latency, we first assessed the expression of multiple IC molecules: PD-1, CTLA-4, TIGIT, TIM-3, BTLA, and their ligands in an in vitro model of latency (44, 45). The model involves coculture of resting CD4+ T cells with monocytes in the presence of SEB, which are known to express high levels of IC ligands, followed by HIV infection with an EGFP-expressing virus (49) and separation of proliferating and nonproliferating T cells based on expression of the proliferation dye eFluor 670 (50) (Fig. 1A). IC molecule expression was determined on T cells before infection and day 1–4 postinfection and coculture. Monocytes were excluded from the analysis by gating for CD3+HLA-DR cells, and IC molecule expression was measured by flow cytometry on the nonproductively infected (EGFP), nonproliferating (eFLuorHI), and proliferating (eFLuorLO) T cells (Fig. 1B–G); the percentage of cells expressing IC markers was calculated over the 4 d (Fig. 1H). Postinfection data from the 3 d time point are summarized in Fig. 1I. In the nonproliferating T cell population, the mean percentage of T cells expressing PD-1, TIM-3, BTLA, and TIGIT all significantly increased, but there was no change in expression of CTLA-4 (Fig. 1I). In the proliferating T cells, the increase in expression of all IC molecules was substantial, with up to 30–40% of cells expressing at least one IC molecule (Fig. 1I).

FIGURE 1.

Expression of IC molecules on nonproliferating and proliferating CD4+ T cells cocultured with monocytes. (A) Schematic representation of the experimental set up. Resting CD4+ T cells isolated from PBMC of HIV-uninfected individuals were cocultured with syngeneic monocytes in the presence of SEB and infected with full-length nef competent CCR5-tropic EGFP reporter HIV. (B) Cells were harvested for flow cytometry analysis, and single cells were selected by assessment of forward scatter (FSC) and side scatter (SSC). T cells were identified by gating for CD3+HLA-DR cells and then gated for EGFP, eFluor 670HI (nonproliferating) and eFluor 670LO (proliferating) T cells. Representative dot plots showing gating strategy for (C) PD-1, (D) TIM-3, (E) CTLA-4, (F) BTLA, and (G) TIGIT. All dot plots shown were from 3 d postinfection. (H) Percentage of cells expressing PD-1 (n = 4), TIM-3 (n = 8), CTLA-4 (n = 3), BTLA (n = 5), and TIGIT (n = 9) on EGFP nonproliferating (left) and proliferating (right) T cells cultured alone (black circles) or with syngeneic monocytes (gray squares) was assessed by flow cytometry. Data points represent mean values ± SEM. Open circles indicate that fewer donors were used for the analysis of IC marker expression on proliferating T cells, as some donors did not have sufficient (>50,000 cells) proliferating T cells at that time point for analysis. (I) The percentage of cells expressing PD-1, TIM-3, CTLA-4, BTLA, and TIGIT 3 d postinfection on T cells cultured alone (gray bars) or cocultured with syngeneic monocytes (blue bars). Data points represent mean ± SEM. Individual donor results are shown as black symbols. *p < 0.05, **p < 0.01 as determined by Student’s t test (n ≤ 5) or Wilcoxon matched-pairs signed-rank test (n > 5).

FIGURE 1.

Expression of IC molecules on nonproliferating and proliferating CD4+ T cells cocultured with monocytes. (A) Schematic representation of the experimental set up. Resting CD4+ T cells isolated from PBMC of HIV-uninfected individuals were cocultured with syngeneic monocytes in the presence of SEB and infected with full-length nef competent CCR5-tropic EGFP reporter HIV. (B) Cells were harvested for flow cytometry analysis, and single cells were selected by assessment of forward scatter (FSC) and side scatter (SSC). T cells were identified by gating for CD3+HLA-DR cells and then gated for EGFP, eFluor 670HI (nonproliferating) and eFluor 670LO (proliferating) T cells. Representative dot plots showing gating strategy for (C) PD-1, (D) TIM-3, (E) CTLA-4, (F) BTLA, and (G) TIGIT. All dot plots shown were from 3 d postinfection. (H) Percentage of cells expressing PD-1 (n = 4), TIM-3 (n = 8), CTLA-4 (n = 3), BTLA (n = 5), and TIGIT (n = 9) on EGFP nonproliferating (left) and proliferating (right) T cells cultured alone (black circles) or with syngeneic monocytes (gray squares) was assessed by flow cytometry. Data points represent mean values ± SEM. Open circles indicate that fewer donors were used for the analysis of IC marker expression on proliferating T cells, as some donors did not have sufficient (>50,000 cells) proliferating T cells at that time point for analysis. (I) The percentage of cells expressing PD-1, TIM-3, CTLA-4, BTLA, and TIGIT 3 d postinfection on T cells cultured alone (gray bars) or cocultured with syngeneic monocytes (blue bars). Data points represent mean ± SEM. Individual donor results are shown as black symbols. *p < 0.05, **p < 0.01 as determined by Student’s t test (n ≤ 5) or Wilcoxon matched-pairs signed-rank test (n > 5).

Close modal

Next, we determined whether the cultured monocytes expressed ligands for IC molecules, including PD-L1 and PD-L2 (ligands for PD-1), galectin 9 (Gal9; ligand for TIM-3), herpes virus entry mediator (HVEM, ligand for BTLA), CD80 and CD86 (ligands for CTLA-4), poliovirus receptor (PVR) and PVR-like 2 (PVRL2, ligands for TIGIT). The same approach was used as described in Fig. 1A, except this time we measured the expression of the IC ligands 1 d following infection, and T cells were excluded from the analysis by gating for CD3HLA-DR+ cells (Fig. 2A). IC ligand expression was compared with FMO or isotype control (Fig. 2B), and the percentage of cells expressing the IC ligands was calculated (Fig. 2C). Monocytes expressed all IC ligands tested, but there was a lower frequency of cells expressing PD-L1, PD-L2, CD80, and PVR compared with the other ligands (Fig. 2C). There were no differences in IC ligand expression in the presence and absence of HIV (Fig. 2C).

FIGURE 2.

Expression of IC molecule ligands on monocytes. Cells were cultured as described in Fig. 1A, except IC ligands were measured one day following infection. (A) Single cells were selected by side scatter (SSC) and forward scatter (FSC), and monocytes were defined by gating for CD3HLA-DR+ cells. (B) Representative dot plots showing gating strategy for PD-L1, PD-L2, Gal9, HVEM, CD80, CD86, PVR, and PVRL2. All dot plots show samples at 1 d postinfection and are compared with FMO control (ctrl) except the dot plot for PVR, which represents a baseline sample compared with isotype (iso) control. (C) Percentage of monocytes expressing ligands for IC markers in the presence (blue) and absence (gray) of HIV, as determined by flow cytometry. Columns represent mean values (n = 3–5), and each symbol represents individual donors.

FIGURE 2.

Expression of IC molecule ligands on monocytes. Cells were cultured as described in Fig. 1A, except IC ligands were measured one day following infection. (A) Single cells were selected by side scatter (SSC) and forward scatter (FSC), and monocytes were defined by gating for CD3HLA-DR+ cells. (B) Representative dot plots showing gating strategy for PD-L1, PD-L2, Gal9, HVEM, CD80, CD86, PVR, and PVRL2. All dot plots show samples at 1 d postinfection and are compared with FMO control (ctrl) except the dot plot for PVR, which represents a baseline sample compared with isotype (iso) control. (C) Percentage of monocytes expressing ligands for IC markers in the presence (blue) and absence (gray) of HIV, as determined by flow cytometry. Columns represent mean values (n = 3–5), and each symbol represents individual donors.

Close modal

CD4+ T cells from PLWH on ART that express PD-1 (22) or multiple IC molecules are enriched for HIV DNA (23). We first confirmed whether these findings could be reproduced in our in vitro model of HIV latency. T cells were cultured with syngeneic monocytes in the presence of SEB and infected with the EGFP reporter virus. On day 5 postinfection, EGFP nonproductively infected cells (i.e., potentially latently infected) in nonproliferating and proliferating T cell populations were sorted based on high and low eFluor 670 expression, respectively. These two populations were then sorted on the basis of IC molecule expression (Supplemental Fig. 1), and inducible latent virus was quantified, as previously described (44, 45) (Fig. 3A). Briefly, sorted cells were activated with anti-CD3/anti-CD28 plus IL-7 plus IL-2 in the presence of raltegravir (an HIV integrase inhibitor) to prevent subsequent rounds of infection. An increase in EGFP expression following stimulation was consistent with postintegration-inducible latent infection (44). Postintegration latency was significantly enriched in nonproliferating T cells expressing high levels of PD-1, TIM-3, CTLA-4, or BTLA but not TIGIT when compared with cells that did not express these IC molecules (p = 0.01, 0.03, 0.01, 0.03, and 0.24, respectively) (Fig. 3B, Table I). In proliferating T cells, postintegration latency was significantly enriched in PD-1HI compared with PD-1LO cells (p = 0.04, Fig. 3B). No enrichment for HIV-infected cells in IC-expressing cells was detected when assessed for all other IC molecules (Fig. 3B).

FIGURE 3.

HIV latency is enriched in cells expressing high levels of IC molecules in nonproliferating and proliferating CD4+ T cells. (A) Resting CD4+ T cells were cultured with syngeneic monocytes in the presence of SEB and infected with EGFP reporter virus. At day 5 postinfection, CD3+HLA-DR EGFP (not productively infected), eFluor 670HI (nonproliferating) and eFluor 670LO (proliferating) T cells were further sorted, based on IC molecule expression into low/negative (−) and high (+) expressing populations compared with FMO controls (see also Supplemental Fig. 1) or the expression of PD-1 plus CTLA-4 plus TIM-3 plus BTLA (multiple IC molecules) or none of these molecules. Sorted cells were cultured in the presence of the integrase inhibitor raltegravir with and without anti-CD3/CD28 plus IL-7 plus IL-2. (B) On day 8, EGFP+ cells were measured by flow cytometry, and latent infection was calculated by subtracting the number of EGFP+ cells in the untreated culture from the number of EGFP+ cells in the stimulated culture. Red lines indicate median values, and dots represent individual donors (n = 4–6). MFC denotes the median fold change between the samples with and without IC expression. *p < 0.05 as determined by Student t test (n ≤ 5) or Wilcoxon matched-pairs signed-rank test (n > 5).

FIGURE 3.

HIV latency is enriched in cells expressing high levels of IC molecules in nonproliferating and proliferating CD4+ T cells. (A) Resting CD4+ T cells were cultured with syngeneic monocytes in the presence of SEB and infected with EGFP reporter virus. At day 5 postinfection, CD3+HLA-DR EGFP (not productively infected), eFluor 670HI (nonproliferating) and eFluor 670LO (proliferating) T cells were further sorted, based on IC molecule expression into low/negative (−) and high (+) expressing populations compared with FMO controls (see also Supplemental Fig. 1) or the expression of PD-1 plus CTLA-4 plus TIM-3 plus BTLA (multiple IC molecules) or none of these molecules. Sorted cells were cultured in the presence of the integrase inhibitor raltegravir with and without anti-CD3/CD28 plus IL-7 plus IL-2. (B) On day 8, EGFP+ cells were measured by flow cytometry, and latent infection was calculated by subtracting the number of EGFP+ cells in the untreated culture from the number of EGFP+ cells in the stimulated culture. Red lines indicate median values, and dots represent individual donors (n = 4–6). MFC denotes the median fold change between the samples with and without IC expression. *p < 0.05 as determined by Student t test (n ≤ 5) or Wilcoxon matched-pairs signed-rank test (n > 5).

Close modal
Table I.
Enrichment of HIV in cells expressing IC molecules
NonproliferatingProliferating
PD-1 48.0a 2.8a 
TIM-3 4.6a 1.4 
CTLA-4 4.9a 0.7 
BTLA 16.4a 1.6 
TIGIT 2.6 0.6a 
PD-1 plus TIM-3 plus CTLA-4 plus BTLA 42.0a 6.5a 
NonproliferatingProliferating
PD-1 48.0a 2.8a 
TIM-3 4.6a 1.4 
CTLA-4 4.9a 0.7 
BTLA 16.4a 1.6 
TIGIT 2.6 0.6a 
PD-1 plus TIM-3 plus CTLA-4 plus BTLA 42.0a 6.5a 

Mean fold change in enrichment of HIV in cells that express IC molecules alone or in combination compared with no expression of ICs. Cells were sorted following in vitro infection as described in Fig. 3. Similar experiments were performed for nonproliferating and proliferating T cells.

a

There was a significant increase of latent HIV in cells expressing high levels of IC molecules compared with cells expressing low to no IC molecules, defined as p < 0.05 using a Student t test (n ≤ 5) or Wilcoxon matched-pairs signed-rank test (n > 5).

To determine whether coexpression of IC molecules resulted in a greater enrichment of HIV within our model, we performed the same experiments as described above, but we sorted cells that expressed any combination of PD-1, TIM-3, CTLA-4, BTLA, or none of these. Postintegration latency was significantly enriched in both nonproliferating (mean fold change [MFC] = 42; p = 0.03) and proliferating (MFC = 48; p = 0.03) T cells expressing one or more of the IC molecules as compared with cells that expressed none of the IC molecules (Fig. 3B, Table I). However, for both nonproliferating and proliferating T cells, latency was still detected in the cells expressing low/none of the IC molecules. Finally, we observed no statistically significant correlations when comparing the frequency of IC-expressing cells and the frequency of latent infection in either the nonproliferating or proliferating T cells (Supplemental Fig. 2). BTLA was excluded from these last experiments, as there is currently no blocking Ab available, and we aimed to investigate IC that have blocking Abs available or in development for clinical use. Taken together, these data demonstrate enrichment of latent HIV infection in proliferating and nonproliferating cells expressing multiple IC molecules alone or in combination.

Given that different cells can express one or more IC molecules, we next defined coexpression of IC molecules on T cells in this in vitro model for HIV latency. We first assessed the coexpression of PD-1, TIM-3, and TIGIT, as some of these IC molecules have been shown to be expressed on cells enriched for HIV using CD4+ T cells from PLWH on ART (23), and we had previously shown that PD-1 and TIM-3 were important in the establishment of latency (26). Resting CD4+ T cells were cocultured with monocytes and analyzed for IC expression by flow cytometry. The percentage of cells positive for one IC (i.e., PD-1 or TIM-3 or TIGIT), two IC, or three IC in combination was calculated (Fig. 4A). Three days postinfection, nonproliferating cells more frequently expressed a single IC than any combination of two IC (n = 4; p < 0.001, compared with single IC) or three IC (n = 4; p < 0.001, compared with single IC) (Fig. 4B). Concomitant expression of two or three IC was far more common on proliferating than on nonproliferating cells. No statistically significant difference was observed in the frequency of cells expressing a single IC between nonproliferating and proliferating T cells. On day 3 postinfection, at the peak of IC expression compared with nonproliferating T cells, proliferating T cells expressed two and three IC more frequently (p = 0.0017, 0.002, respectively, for both) (Fig. 4B). Overall, these results show that nonproliferating T cells more often express a single IC molecule, whereas proliferating T cells more commonly coexpress two or more IC molecules. These results were similar in the presence and absence of HIV infection (data not shown).

FIGURE 4.

Coexpression of multiple IC molecules is more frequent on proliferating CD4+ T cells. (A) Coexpression of IC molecules PD-1, TIM-3, and TIGIT on T cells was determined in CD3+HLA-DREGFP nonproliferating (left) and proliferating (right) T cells cultured with syngeneic monocytes by flow cytometry at the day of infection (day 0) and on each day postinfection (day1–4), compared with isotype control. The results are grouped to show the distribution of cells expressing no IC (gray), one IC (yellow), any combination of two IC (blue), or all three IC (red). Data points represent the mean of four donors ± SEM. *p < 0.05, **p < 0.01, ***p < 0.001 as determined by Student t test. (B) Pie chart showing the distribution of IC molecule expression on nonproliferating (top) and proliferating (bottom) T cells on day 3 postinfection. Coexpression of IC molecules PD-1, CTLA-4, TIM-3, and TIGIT on T cells was determined in CD3+HLA-DREGFP nonproliferating (C) and proliferating (D) T cells cultured with syngeneic monocytes by flow cytometry at day 5 postinfection compared with isotype control. Black lines indicate mean values of six donors ± SEM, and equal colors represent equal donors across the panels. (E) Pie chart showing the distribution of IC expression on nonproliferating (left) and proliferating (right) T cells day 5 postinfection. The results are grouped to show the distribution of cells expressing no IC (gray), one IC (yellow), any combination of two IC (blue), any combination of three IC (red), or four IC (green).

FIGURE 4.

Coexpression of multiple IC molecules is more frequent on proliferating CD4+ T cells. (A) Coexpression of IC molecules PD-1, TIM-3, and TIGIT on T cells was determined in CD3+HLA-DREGFP nonproliferating (left) and proliferating (right) T cells cultured with syngeneic monocytes by flow cytometry at the day of infection (day 0) and on each day postinfection (day1–4), compared with isotype control. The results are grouped to show the distribution of cells expressing no IC (gray), one IC (yellow), any combination of two IC (blue), or all three IC (red). Data points represent the mean of four donors ± SEM. *p < 0.05, **p < 0.01, ***p < 0.001 as determined by Student t test. (B) Pie chart showing the distribution of IC molecule expression on nonproliferating (top) and proliferating (bottom) T cells on day 3 postinfection. Coexpression of IC molecules PD-1, CTLA-4, TIM-3, and TIGIT on T cells was determined in CD3+HLA-DREGFP nonproliferating (C) and proliferating (D) T cells cultured with syngeneic monocytes by flow cytometry at day 5 postinfection compared with isotype control. Black lines indicate mean values of six donors ± SEM, and equal colors represent equal donors across the panels. (E) Pie chart showing the distribution of IC expression on nonproliferating (left) and proliferating (right) T cells day 5 postinfection. The results are grouped to show the distribution of cells expressing no IC (gray), one IC (yellow), any combination of two IC (blue), any combination of three IC (red), or four IC (green).

Close modal

Given the availability of anti–CTLA-4 in the clinic for management of melanoma (51), we also examined the coexpression of the same three IC together with CTLA-4. Proportions of cells expressing CTLA-4 was less frequent compared with the other IC molecules (i.e., PD-1 and TIGIT on nonproliferating and PD-1 and TIM-3 on proliferating T cells) (Fig. 4C). Again, we found that there was more frequent expression of IC molecules in proliferating cells compared with nonproliferating cells, (expression of more than one IC was found in 46 and 7% and a single IC molecule in 33 and 15% of cells, respectively) (Fig. 4C–E). Coexpression of all four IC molecules was the rarest population within either proliferating or nonproliferating cells (Fig. 4E). Collectively, this data shows that the distribution of PD-1, CTLA-4, TIM-3, and TIGIT, expressed individually and in combination, differs between nonproliferating and proliferating T cells, and that expression of more than one IC molecule was more frequent in proliferating T cells.

To examine whether blocking IC, either alone or in combination, can reverse HIV latency in vitro, resting CD4+ T cells were cocultured with monocytes in the presence of SEB and sorted into EGFP nonproliferating and proliferating populations, as described above. ICB were added to the sorted cells in the presence of raltegravir and T20 (an integrase and fusion inhibitor, respectively). As we exclude HLA-DR+ T cells during the sort on day 5 postinfection and select for CD3+ cells, the monocytes were excluded from the sorted population. Therefore, for the IC ligand to be present in the blocking experiment, syngeneic monocytes were added back to the cultures postsort (Fig. 5A). Additionally, sorted cells were cultured in the absence of monocytes with anti-CD3/CD28 plus IL-7 plus IL-2 to induce maximal HIV production and to which all conditions were compared.

FIGURE 5.

PD-1 and CTLA-4 blockade reverses latent HIV but only in the presence of SEB or other ICB. (A) Schematic representation of experimental set up to test whether ICB can reverse HIV latency in vitro. Resting CD4+ T cells were cultured with syngeneic monocytes in the presence of SEB and infected with EGFP reporter virus. At day 5 postinfection, CD3+HLA-DREGFP eFluor 670HI (nonproliferating) and eFluor 670LO (proliferating) T cells were sorted, cultured with both an integrase inhibitor (raltegravir) and a fusion inhibitor (T20), and left untreated or stimulated with anti-CD3/CD28 plus IL-7 plus IL-2 (negative and positive control) or cocultured with add-back syngeneic monocytes (mo) with the ICB or combinations thereof or isotype controls in the absence or presence of SEB. The cells were harvested 3 d postsort (day 8 postinfection), and the number of EGFP+ cells was determined by flow cytometry (see also Supplemental Fig. 3A, 3B). Latency reversal following IC blockade is shown as a percentage of maximal stimulation with anti-CD3/CD28 plus IL-7 plus IL-2 in nonproliferating (B) or proliferating (C) T cells. To calculate the percentage, the number of EGFP+ cells in unstimulated (mock) condition was subtracted from the number of EGFP+ cells following a maximal stimulus or stimulus from the ICB. Black lines indicate mean values of six donors ± SEM, and equal colors represent equal donors across the panels. *p < 0.05 as Wilcoxon matched-pairs signed-rank test determined using the number of EGFP+ cells (shown in Supplemental Fig. 3A, 3B) in the presence of the ICB compared with the corresponding isotype control.

FIGURE 5.

PD-1 and CTLA-4 blockade reverses latent HIV but only in the presence of SEB or other ICB. (A) Schematic representation of experimental set up to test whether ICB can reverse HIV latency in vitro. Resting CD4+ T cells were cultured with syngeneic monocytes in the presence of SEB and infected with EGFP reporter virus. At day 5 postinfection, CD3+HLA-DREGFP eFluor 670HI (nonproliferating) and eFluor 670LO (proliferating) T cells were sorted, cultured with both an integrase inhibitor (raltegravir) and a fusion inhibitor (T20), and left untreated or stimulated with anti-CD3/CD28 plus IL-7 plus IL-2 (negative and positive control) or cocultured with add-back syngeneic monocytes (mo) with the ICB or combinations thereof or isotype controls in the absence or presence of SEB. The cells were harvested 3 d postsort (day 8 postinfection), and the number of EGFP+ cells was determined by flow cytometry (see also Supplemental Fig. 3A, 3B). Latency reversal following IC blockade is shown as a percentage of maximal stimulation with anti-CD3/CD28 plus IL-7 plus IL-2 in nonproliferating (B) or proliferating (C) T cells. To calculate the percentage, the number of EGFP+ cells in unstimulated (mock) condition was subtracted from the number of EGFP+ cells following a maximal stimulus or stimulus from the ICB. Black lines indicate mean values of six donors ± SEM, and equal colors represent equal donors across the panels. *p < 0.05 as Wilcoxon matched-pairs signed-rank test determined using the number of EGFP+ cells (shown in Supplemental Fig. 3A, 3B) in the presence of the ICB compared with the corresponding isotype control.

Close modal

The effect of anti–PD-1 (nivolumab) and anti–CTLA-4 (ipilimumab) were tested alone or in combination, as well as in combination with commercially available Abs to TIM-3 and TIGIT. BTLA was not further examined, as blocking agents are currently unavailable. Effects of ICB were assessed in the presence or absence of SEB. SEB is a super-Ag that will drive proliferation in a subset of cells that express a specific Vb repertoire (52). We assumed that SEB will drive proliferation of both uninfected and latently infected cells in this model. Latency reversal was quantified by measuring the increase in absolute number of cells that express EGFP following stimulation (Supplemental Fig. 3A) or as a proportion of maximal stimulation with anti-CD3/CD28 plus IL-7 plus IL-2 (Fig. 5B). The mean (range) of EGFP+ cells per 104 cells in the untreated control was 10.8 (8.4–17.1)/104 cells, and this increased to 25.9 (12.0–37.2)/104 cells following stimulation of the sorted nonproliferating T cells using a maximal stimulus (Supplemental Fig. 3A). For the sorted proliferating T cells, the number of EGFP+ cells increased from 22.3 (3.5–47.5)/104 to 46.2 (25.6–73.3)/104 using anti-CD3/CD28 plus IL-7 plus IL-2 (Supplemental Fig. 3B).

In nonproliferating CD4+ T cells, there was an increase in the number of EGFP+ cells following addition of anti-CD3/CD28 plus IL-7 plus IL-2, as expected (MFC 2.5; n = 6; p = 0.031) (Supplemental Fig. 3A), although there was quite considerable variation in inducible EGFP expression between donors. In the absence of SEB, a significant increase in EGFP+ cells was only observed when all four ICB were added (p = 0.03; Fig. 5B, Supplemental Fig. 3A). In the presence of SEB, the number of EGFP+ cells increased with anti–PD-1 alone (p = 0.031) and in combination with anti–CTLA-4 (p = 0.031) but not with anti–CTLA-4 alone (p = 0.22), consistent with only anti–PD-1 inducing latency reversal. The addition of Abs to TIM-3 (p = 0.16) and TIGIT (p = 0.22) had no further effect in enhancing EGFP expression (Fig. 5B).

In the proliferating T cells, there was an increase in EGFP+ cells following the addition of anti-CD3/CD28 plus IL-7 plus IL-2 addition as expected (n = 6; MFC 3.38; p = 0.031) (Supplemental Fig. 3B). In the absence of SEB, the results were similar to the nonproliferating cells, with an increase in EGFP only observed following the addition of all four ICB. In the presence of SEB, we observed that only anti–CTLA-4 induced a modest increase in the number of EGFP+ cells (p = 0.031) (Fig. 5C, Supplemental Fig. 3B). Interestingly, there was a decrease in the number of EGFP+ cells in the presence of anti–PD-1 (MFC 0.83; p = 0.031) which was not related to a decrease in cell viability (data not shown).

To understand what factors might be associated with efficient latency reversal, we assessed whether there was a correlation between the induction of EGFP expression and the degree of T cell proliferation, as measured by an increase in the proportion of eFluor 670LO cells for the nonproliferating cell cultures or a decrease in eFluor 670 mean fluorescent intensity in the proliferating cells, given the dye is diluted with each cell division. Following the addition of the ICB, no relationship was observed between cell proliferation and induction of EGFP expression (data not shown).

In summary, these data demonstrate that latency reversal could be achieved in both nonproliferating and proliferating latently infected cells when multiple ICB were used in combination without SEB. The degree of latency reversal was high; approaching 45% of what was achieved with maximal stimulation. In the presence of SEB, we found that latency reversal was possible using a single ICB in nonproliferating latently infected cells (using anti–PD-1) and in proliferating cells (using anti–CTLA-4) (Table II). These data suggest that the use of a single ICB is unlikely to sufficiently activate latent infection in either proliferating or nonproliferating cells, and an additional stimulus is required.

Table II.
HIV latency reversal induced by IC blockade or LRAs in vitro
Nonproliferating
Proliferating
No SEBPlus SEBNo SEBPlus SEB
ICB     
 aPD-1 23.4 63.3a 31.5 22.4 
 Isot ctrl aPD-1 25.3 33.8 39.6 43.2 
 aCTLA-4 38.6 66.3 39.3 53.2a 
 Isot ctrl aCTLA-4 27.0 52.3 47.8 37.1 
 aPD-1 plus aCTLA-4 33.2 75.2a 42.9 34.2 
 Isot ctrl 2 ICB 21.6 39.4 41.9 37.2 
 aPD-1 plus aCTLA-4 plus aTIM-3 40.5 65.3 49.6 40.7 
 Isot ctrl 3 ICB 22.9 30.2 31.7 36.8 
 aPD-1 plus aCTLA-4 plus aTIM-3 plus aTIGIT 44.6a 74.1 48.2a 35.1 
 Isot ctrl 4 ICB 22.8 56.8 19.2 39.8 
Nonproliferating
Proliferating
No SEBPlus SEBNo SEBPlus SEB
ICB     
 aPD-1 23.4 63.3a 31.5 22.4 
 Isot ctrl aPD-1 25.3 33.8 39.6 43.2 
 aCTLA-4 38.6 66.3 39.3 53.2a 
 Isot ctrl aCTLA-4 27.0 52.3 47.8 37.1 
 aPD-1 plus aCTLA-4 33.2 75.2a 42.9 34.2 
 Isot ctrl 2 ICB 21.6 39.4 41.9 37.2 
 aPD-1 plus aCTLA-4 plus aTIM-3 40.5 65.3 49.6 40.7 
 Isot ctrl 3 ICB 22.9 30.2 31.7 36.8 
 aPD-1 plus aCTLA-4 plus aTIM-3 plus aTIGIT 44.6a 74.1 48.2a 35.1 
 Isot ctrl 4 ICB 22.8 56.8 19.2 39.8 
NonproliferatingProliferating
Other LRAs   
 DMSO 7.0 2.7 
 Romi 1 nM 1.2 6.3 
 Romi 0.1 nM 5.1 7.2 
 JQ1 1 μM 2.7 2.0 
 JQ1 0.1 μM 5.2 7.8 
 Bryo 10 nM 50.9a 42.6a 
 Bryo 1 nM 64.8a 47.4 
 PMA plus iono 26.5 87.2 
NonproliferatingProliferating
Other LRAs   
 DMSO 7.0 2.7 
 Romi 1 nM 1.2 6.3 
 Romi 0.1 nM 5.1 7.2 
 JQ1 1 μM 2.7 2.0 
 JQ1 0.1 μM 5.2 7.8 
 Bryo 10 nM 50.9a 42.6a 
 Bryo 1 nM 64.8a 47.4 
 PMA plus iono 26.5 87.2 

The percentage of maximal response induced following stimulation with anti-CD3 and anti-CD28 is shown. Increase in EGFP expression (latency reversal) is shown as the mean percentage of maximal activation with anti-CD3/28 plus IL-7 plus IL-2 following addition of ICB Abs or isot ctrl alone or in combination (top panels) or LRA (bottom panels).

a

There was a significant increase compared with isot ctrl (for IC blockade or DMSO [for other LRAs]), defined as p < 0.05, using the frequency of EGFP+ cells shown in Supplemental Fig. 3.

a, anti; bryo, bryostatin; iono, ionomycin; isot ctrl, isotype control; Romi, romidepsin.

To investigate the relative potency of IC blockade for latency reversal compared with other LRAs that have been shown to reverse latent HIV, we next assessed commonly used LRAs in the same in vitro model in separate experiments (Fig. 6A). We tested three different classes of LRAs: the HDACi romidepsin, the bromodomain inhibitor JQ1, and an activator of protein kinase C (PKC), bryostatin. Of note, lower doses of romidepsin were used for these cell cultures that were maintained for 3 d. We did this to avoid the toxic effects of 40 nM romidepsin observed in continuous T cell cultures elsewhere (data not shown). As positive controls, we included anti-CD3/CD28 plus IL-7 plus IL-2 and the combination of PMA and ionomycin that mimic T cell activation, activating the transcription factors NF-κB and NFAT to initiate HIV transcription (53). Of the LRAs tested, only bryostatin led to a statistically significant increase in the frequency of EGFP+ cells in both nonproliferating and proliferating latently infected cells, reaching 65 and 43% of maximal stimulation induced by anti-CD3/CD28 plus IL-7 plus IL-2 (Fig. 6, Supplemental Fig. 3, Table II). In this model of latency, we demonstrate that IC blockade in combination or using SEB with either anti–PD-1 or anti–CTLA-4 was more potent than commonly used LRAs such as romidepsin (tested at continuous low doses) or JQ1.

FIGURE 6.

Potency of commonly used LRAs in this in vitro model. To determine the relative potency of latency reversal by commonly used LRAs in the same in vitro model as with IC blockade (Fig. 5), cells were cultured as described in Fig. 5A, with the exception that after day 5, monocytes were not added back to the sorted cells. Cells were then cultured with different LRAs in the presence of an integrase inhibitor. The cells were harvested 3 d after addition of an LRA (day 8 postinfection), and the number of EGFP+ cells was determined by flow cytometry (see also Supplemental Fig. 3C, 3D). EGFP expression following stimulation with an LRA is shown as a percentage of the maximal stimulation by anti-CD3/CD28 plus IL-7 plus IL-2 (set at 100%) in nonproliferating (A) and proliferating (B) CD4+ T cells. Black lines indicate mean values of five donors ± SEM, and equal colors represent equal donors across the panels. *p < 0.05, **p < 0.01 as determined by Student t test. bryo, bryostatin; iono, ionomycin; Romi, romidepsin.

FIGURE 6.

Potency of commonly used LRAs in this in vitro model. To determine the relative potency of latency reversal by commonly used LRAs in the same in vitro model as with IC blockade (Fig. 5), cells were cultured as described in Fig. 5A, with the exception that after day 5, monocytes were not added back to the sorted cells. Cells were then cultured with different LRAs in the presence of an integrase inhibitor. The cells were harvested 3 d after addition of an LRA (day 8 postinfection), and the number of EGFP+ cells was determined by flow cytometry (see also Supplemental Fig. 3C, 3D). EGFP expression following stimulation with an LRA is shown as a percentage of the maximal stimulation by anti-CD3/CD28 plus IL-7 plus IL-2 (set at 100%) in nonproliferating (A) and proliferating (B) CD4+ T cells. Black lines indicate mean values of five donors ± SEM, and equal colors represent equal donors across the panels. *p < 0.05, **p < 0.01 as determined by Student t test. bryo, bryostatin; iono, ionomycin; Romi, romidepsin.

Close modal

In PLWH on ART, HIV persists preferentially in CD4+ T cells expressing multiple IC molecules, in particular PD-1 (7, 22, 23). To determine the role of multiple IC molecules and blockers in the maintenance and reversal of HIV latency, we used an in vitro model that can establish latency in both nonproliferating and proliferating T cells (44, 45, 50). These two populations of latently infected cells cannot be distinguished when evaluating CD4+ T cells ex vivo from PLWH on ART. We found HIV latency to be enriched in nonproliferating and proliferating T cells expressing IC molecules, and the enrichment was greater in nonproliferating cells that expressed multiple IC molecules. Using multiple ICB that are licensed for the treatment of malignancy (anti–PD-1 and anti–CTLA-4) and in preclinical development (anti–TIM-3 and anti-TIGIT), we demonstrated that reversal of HIV latency was possible with a single IC Ab in combination with a T cell activation stimulus (SEB in this study) or with a combination of multiple Abs to IC molecules without another stimulus. In this latency model, the potency of IC blockade in reversing latency was comparable to reversing HIV latency with the PKC activator bryostatin and greater than commonly used LRAs, such as the HDACi romidepsin (used at low dose) or the bromodomain inhibitor JQ1. Given the additional potential beneficial effects of IC blockade in enhancing the function of HIV-specific CD4+ and CD8+ T cells (54, 55), IC blockade is an attractive option to move forward into clinical evaluation as both a potential LRA and as a strategy to boost HIV-specific T cell function.

Using an in vitro model of HIV latency, latent infection was found to be enriched in nonproliferating CD4+ T cells expressing PD-1, TIM-3, CTLA-4, or BTLA, but in proliferating T cells, this enrichment was only found in T cells expressing PD-1. IC molecules were expressed at high levels but were often coexpressed on proliferating T cells, whereas nonproliferating T cells more commonly expressed a single IC molecule. The mechanisms behind the need for multiple ICB to reverse latent infection in nonproliferating and proliferating cells may be different. For the nonproliferating cells, multiple ICB may be required to target different cells that express a single IC molecule, whereas for the proliferating cells, multiple ICB are needed because of the high frequency of coexpression of different types of ICs on the same cell.

Blocking IC molecules inhibits the negative signaling in the T cell activation process, effectively releasing the breaks on T cell activation. TCR and superantigen signaling induce multiple transcription factors to become active and/or translocate to the nucleus leading to induction of HIV transcription and virus production (56, 57), among many other changes in the host cell. These host transcription factors predominantly include NF-κB, NFAT, specificity protein 1 (SP-1) and AP-1 (reviewed in Ref. 58). We recently showed that PD-1 signaling prevents activation of the positive transcription elongation factor b (P-TEFb), a master regulator of HIV transcription, through inhibition of TCR-induced CDK9 phosphorylation and cyclin T1 expression (27). Whether these or other pathways are important for latency reversal following combination IC blockade remains to be determined.

In our in vitro model, we found that a single ICB could reverse latency but only in the presence of SEB (see Fig. 5). Furthermore, anti–PD-1 was the only Ab that could reverse latency in nonproliferating T cells, whereas only anti–CTLA-4 reversed latency in proliferating T cells. The lack of activity of anti–CTLA-4 in nonproliferating cells may be a result of the low expression of CTLA-4 on these cells (see Fig. 1H). This is expected because CTLA-4 is upregulated on cells following TCR priming (59) and these cells had not undergone proliferation in response to the superantigen SEB. In contrast, the proliferating T cells expressed high levels of CTLA-4 (see Fig. 1H), as expected, following stimulation with a superantigen or TCR-mediated stimulation. Similarly, the failure of anti–PD-1 to reverse HIV latency in proliferating cells when used on its own would be a major limitation as an LRA. Although administration of the combination of anti–PD-1 and anti–CTLA-4 is now recommended in the management of some malignancies, toxicity was significantly increased (29). An increase in toxicity would significantly limit the use of combination IC blockade as an HIV cure intervention, given that PLWH now have a near normal life expectancy on long term ART.

Blocking IC molecules can also induce T cell proliferation. It is therefore feasible that driving T cell proliferation could increase the number of latently infected cells in vivo, which would not be desirable. In our in vitro model, we observed enhanced T cell proliferation upon IC blockade when combined with superantigen SEB, as expected, but no correlation was observed between the degree of proliferation and reversal of latency (data not shown). In our model, we did not directly measure the number of latently infected cells but, rather, the number of cells with inducible virus. It is certainly possible that the cultures also contain cells infected with intact virus that do not respond to stimulation. This has been well described in CD4+ T cells from PLWH on ART and referred to as noninduced intact proviruses (60). Further work is needed to also measure the total number of infected cells prior to and following IC blockade to exclude the possibility of proliferation of infected cells that contain noninduced intact provirus.

There were several limitations to this study. First, we used an in vitro model to probe the effects of different IC molecules. These results need confirmation using CD4+ T cells from PLWH on ART ex vivo and/or in vivo administration of an ICB Ab. We are currently studying changes in cell-associated HIV RNA and plasma HIV RNA in the context of clinical trials of anti–PD-1 and anti–CTLA-4 in PLWH with cancer (https//clinicaltrials.gov NCT02408861). Despite this limitation, our model has several advantages, including the following: 1) the ability to separate proliferating and nonproliferating latently infected cells, which is currently not possible with cells from PLWH; 2) the measurement of inducible virus rather than cell-associated RNA, which allows quantification of the number of infected cells that undergo latency reversal; and 3) the incorporation of a superantigen to drive proliferation. A second limitation of this study is that we did not assess every possible combination of two or three ICB. Instead, we focused on the clinically relevant combination of anti–PD-1 and anti–CTLA-4 and added other ICB (TIM-3, TIGIT) to this well-established, clinically tested combination. A third limitation is that the use of SEB is not a physiological stimulus but can mimic an Ag-specific response. Other costimuli should be further evaluated. A fourth limitation of our study is that we did not evaluate the role of LAG-3 because we were unable to detect an increase in LAG-3 expression upon coculturing the CD4+ T cells with monocytes. Furthermore, we did not have access to a clinically relevant anti-LAG3 Ab to test in vitro. Finally, we only quantified inducible EGFP expression as a marker of latency reversal. Other proximal markers, such as cell-associated unspliced and multiply spliced RNA, were not quantified; however, given there are blocks in transcriptional elongation and splicing described in resting CD4+ T cells (61) and that protein expression will be required for either immune-mediated clearance or virus-induced cytolysis, we felt that quantification of protein expression would be most informative.

In conclusion, using an in vitro model that can establish latency in both nonproliferating and proliferating T cells, we show that Abs blocking signaling of specific IC molecules could reverse HIV latency in both proliferating and nonproliferating T cells, but this was only achieved in the presence of an additional T cell–activating stimulus or when four Abs were used in combination. Given that SEB is not a physiological stimulus and not feasible to deliver in vivo (62), further studies are needed to identify alternative secondary stimulus to replace SEB. One approach could be therapeutic vaccination for HIV (63), which could drive T cell proliferation but also enhance the generation of Ag-specific T cells, as recently demonstrated in the setting of cancer (64). Alternatively, the PKC agonist bryostatin or a related compound (65) might be a complimentary stimulus, given that bryostatin appears to be safe in vivo (66), induces T cell activation, and acts as an LRA alone (53) or in combination with anti–PD-1 (27). The administration of a combination of ICB should be further explored as a path to HIV remission following cessation of ART.

We thank Damian Purcell (University of Melbourne, Parkville, Australia) and Yasuko Tsunetsugu-Yokota (National Institute of Infectious Diseases, Tokyo, Japan) for providing the EGFP reporter virus. We thank G. Paukovics, J. Le Masurier, and P. Donaldson (Alfred Medical Research and Educational Precinct Flow Cytometry Core Facility, Melbourne, Australia) and T. Luka, C. Li, A. Hind, and D. Blashki (University of Melbourne Flow Cytometry Facility, Melbourne, Australia) for flow cytometric cell sorting. We thank Ashish Nair (University of Melbourne) for help processing the human buffy coats. We thank Alan Korman from Bristol Myers Squibb for providing nivolumab (anti–PD-1) and ipilimumab (anti–CTLA-4).

This work was supported by funds from The Foundation for AIDS Research (Grants 108237-51-RGRL and 109226-58-RGRL), the National Health and Medical Research Council (NHMRC) of Australia (Grants APP1041795 and APP3162044), and the National Institutes of Health Delaney AIDS Research Enterprise to Find a Cure Collaboratory (Grant UM1AI126611-01). S.R.L. is an NHMRC Practitioner Fellow.

The online version of this article contains supplemental material.

Abbreviations used in this article:

ART

antiretroviral therapy

BTLA

B and T lymphocyte attenuator

CTLA-4

CTL-associated protein 4

FMO

fluorescence minus one

HDACi

histone deacetylase inhibitor

IC

immune checkpoint

ICB

IC blocker, IC blocking

LAG-3

lymphocyte activation gene 3

LRA

latency-reversing agent

MFC

mean fold change

PD-1

programmed death 1

PD-L

programmed death ligand

PKC

protein kinase C

PLWH

people living with HIV

PVR

poliovirus receptor

r-h

recombinant human

SEB

staphylococcal enterotoxin B

TIGIT

T cell immunoreceptor with Ig and ITIM domains

TIM-3

T cell Ig and mucin domain-containing protein 3.

1
Deeks
,
S. G.
,
S. R.
Lewin
,
A. L.
Ross
,
J.
Ananworanich
,
M.
Benkirane
,
P.
Cannon
,
N.
Chomont
,
D.
Douek
,
J. D.
Lifson
,
Y. R.
Lo
, et al
International AIDS Society Towards a Cure Working Group
.
2016
.
International AIDS Society global scientific strategy: towards an HIV cure 2016.
Nat. Med.
22
:
839
850
.
2
Chun
,
T. W.
,
L.
Carruth
,
D.
Finzi
,
X.
Shen
,
J. A.
DiGiuseppe
,
H.
Taylor
,
M.
Hermankova
,
K.
Chadwick
,
J.
Margolick
,
T. C.
Quinn
, et al
.
1997
.
Quantification of latent tissue reservoirs and total body viral load in HIV-1 infection.
Nature
387
:
183
188
.
3
Hosmane
,
N. N.
,
K. J.
Kwon
,
K. M.
Bruner
,
A. A.
Capoferri
,
S.
Beg
,
D. I.
Rosenbloom
,
B. F.
Keele
,
Y. C.
Ho
,
J. D.
Siliciano
,
R. F.
Siliciano
.
2017
.
Proliferation of latently infected CD4+ T cells carrying replication-competent HIV-1: potential role in latent reservoir dynamics.
J. Exp. Med.
214
:
959
972
.
4
Lorenzi
,
J. C.
,
Y. Z.
Cohen
,
L. B.
Cohn
,
E. F.
Kreider
,
J. P.
Barton
,
G. H.
Learn
,
T.
Oliveira
,
C. L.
Lavine
,
J. A.
Horwitz
,
A.
Settler
, et al
.
2016
.
Paired quantitative and qualitative assessment of the replication-competent HIV-1 reservoir and comparison with integrated proviral DNA. [Published erratum appears in 2017 Proc. Natl. Acad. Sci. USA 114: E648–E649.]
Proc. Natl. Acad. Sci. USA
113
:
E7908
E7916
.
5
Bui
,
J. K.
,
E. K.
Halvas
,
E.
Fyne
,
M. D.
Sobolewski
,
D.
Koontz
,
W.
Shao
,
B.
Luke
,
F. F.
Hong
,
M. F.
Kearney
,
J. W.
Mellors
.
2017
.
Ex vivo activation of CD4+ T-cells from donors on suppressive ART can lead to sustained production of infectious HIV-1 from a subset of infected cells.
PLoS Pathog.
13
: e1006230.
6
Chun
,
T. W.
,
D. C.
Nickle
,
J. S.
Justement
,
J. H.
Meyers
,
G.
Roby
,
C. W.
Hallahan
,
S.
Kottilil
,
S.
Moir
,
J. M.
Mican
,
J. I.
Mullins
, et al
.
2008
.
Persistence of HIV in gut-associated lymphoid tissue despite long-term antiretroviral therapy.
J. Infect. Dis.
197
:
714
720
.
7
Banga
,
R.
,
F. A.
Procopio
,
A.
Noto
,
G.
Pollakis
,
M.
Cavassini
,
K.
Ohmiti
,
J. M.
Corpataux
,
L.
de Leval
,
G.
Pantaleo
,
M.
Perreau
.
2016
.
PD-1(+) and follicular helper T cells are responsible for persistent HIV-1 transcription in treated aviremic individuals.
Nat. Med.
22
:
754
761
.
8
Estes
,
J. D.
,
C.
Kityo
,
F.
Ssali
,
L.
Swainson
,
K. N.
Makamdop
,
G. Q.
Del Prete
,
S. G.
Deeks
,
P. A.
Luciw
,
J. G.
Chipman
,
G. J.
Beilman
, et al
.
2017
.
Defining total-body AIDS-virus burden with implications for curative strategies.
Nat. Med.
23
:
1271
1276
.
9
Kim
,
Y.
,
J. L.
Anderson
,
S. R.
Lewin
.
2018
.
Getting the “kill” into “shock and kill”: strategies to eliminate latent HIV.
Cell Host Microbe
23
:
14
26
.
10
Zerbato
,
J. M.
,
H. V.
Purves
,
S. R.
Lewin
,
T. A.
Rasmussen
.
2019
.
Between a shock and a hard place: challenges and developments in HIV latency reversal.
Curr. Opin. Virol.
38
:
1
9
.
11
Elliott
,
J. H.
,
F.
Wightman
,
A.
Solomon
,
K.
Ghneim
,
J.
Ahlers
,
M. J.
Cameron
,
M. Z.
Smith
,
T.
Spelman
,
J.
McMahon
,
P.
Velayudham
, et al
.
2014
.
Activation of HIV transcription with short-course vorinostat in HIV-infected patients on suppressive antiretroviral therapy.
PLoS Pathog.
10
: e1004473.
12
Rasmussen
,
T. A.
,
M.
Tolstrup
,
C. R.
Brinkmann
,
R.
Olesen
,
C.
Erikstrup
,
A.
Solomon
,
A.
Winckelmann
,
S.
Palmer
,
C.
Dinarello
,
M.
Buzon
, et al
.
2014
.
Panobinostat, a histone deacetylase inhibitor, for latent-virus reactivation in HIV-infected patients on suppressive antiretroviral therapy: a phase 1/2, single group, clinical trial.
Lancet HIV
1
:
e13
e21
.
13
Søgaard
,
O. S.
,
M. E.
Graversen
,
S.
Leth
,
R.
Olesen
,
C. R.
Brinkmann
,
S. K.
Nissen
,
A. S.
Kjaer
,
M. H.
Schleimann
,
P. W.
Denton
,
W. J.
Hey-Cunningham
, et al
.
2015
.
The depsipeptide romidepsin reverses HIV-1 latency in vivo.
PLoS Pathog.
11
: e1005142.
14
Vibholm
,
L.
,
M. H.
Schleimann
,
J. F.
Højen
,
T.
Benfield
,
R.
Offersen
,
K.
Rasmussen
,
R.
Olesen
,
A.
Dige
,
J.
Agnholt
,
J.
Grau
, et al
.
2017
.
Short-course toll-like receptor 9 agonist treatment impacts innate immunity and plasma viremia in individuals with human immunodeficiency virus infection.
Clin. Infect. Dis.
64
:
1686
1695
.
15
Archin
,
N. M.
,
A. L.
Liberty
,
A. D.
Kashuba
,
S. K.
Choudhary
,
J. D.
Kuruc
,
A. M.
Crooks
,
D. C.
Parker
,
E. M.
Anderson
,
M. F.
Kearney
,
M. C.
Strain
, et al
.
2012
.
Administration of vorinostat disrupts HIV-1 latency in patients on antiretroviral therapy. [Published erratum appears in 2012 Nature 489: 460.]
Nature
487
:
482
485
.
16
Jones
,
R. B.
,
S.
Mueller
,
R.
O’Connor
,
K.
Rimpel
,
D. D.
Sloan
,
D.
Karel
,
H. C.
Wong
,
E. K.
Jeng
,
A. S.
Thomas
,
J. B.
Whitney
, et al
.
2016
.
A subset of latency-reversing agents expose HIV-infected resting CD4+ T-cells to recognition by cytotoxic T-lymphocytes.
PLoS Pathog.
12
: e1005545.
17
Beliakova-Bethell
,
N.
,
A.
Mukim
,
C. H.
White
,
S.
Deshmukh
,
H.
Abewe
,
D. D.
Richman
,
C. A.
Spina
.
2019
.
Histone deacetylase inhibitors induce complex host responses that contribute to differential potencies of these compounds in HIV reactivation.
J. Biol. Chem.
294
:
5576
5589
.
18
Brinkmann
,
C. R.
,
J. F.
Højen
,
T. A.
Rasmussen
,
A. S.
Kjær
,
R.
Olesen
,
P. W.
Denton
,
L.
Østergaard
,
Z.
Ouyang
,
M.
Lichterfeld
,
X.
Yu
, et al
.
2018
.
Treatment of HIV-infected individuals with the histone deacetylase inhibitor panobinostat results in increased numbers of regulatory T cells and limits ex vivo lipopolysaccharide-induced inflammatory responses.
mSphere
3
:
e00616
e00630
.
19
Clutton
,
G.
,
Y.
Xu
,
P. L.
Baldoni
,
K. R.
Mollan
,
J.
Kirchherr
,
W.
Newhard
,
K.
Cox
,
J. D.
Kuruc
,
A.
Kashuba
,
R.
Barnard
, et al
.
2016
.
The differential short- and long-term effects of HIV-1 latency-reversing agents on T cell function. [Published erratum appears in 2016 Sci. Rep. 6: 34430.]
Sci. Rep.
6
:
30749
.
20
Garrido
,
C.
,
M.
Tolstrup
,
O. S.
Søgaard
,
T. A.
Rasmussen
,
B.
Allard
,
N.
Soriano-Sarabia
,
N. M.
Archin
,
D. M.
Margolis
.
2019
.
In-vivo administration of histone deacetylase inhibitors does not impair natural killer cell function in HIV+ individuals.
AIDS
33
:
605
613
.
21
Pace
,
M.
,
J.
Williams
,
A.
Kurioka
,
A. B.
Gerry
,
B.
Jakobsen
,
P.
Klenerman
,
N.
Nwokolo
,
J.
Fox
,
S.
Fidler
,
J.
Frater
;
CHERUB Investigators
.
2016
.
Histone deacetylase inhibitors enhance CD4 T cell susceptibility to NK cell killing but reduce NK cell function.
PLoS Pathog.
12
: e1005782.
22
Chomont
,
N.
,
M.
El-Far
,
P.
Ancuta
,
L.
Trautmann
,
F. A.
Procopio
,
B.
Yassine-Diab
,
G.
Boucher
,
M. R.
Boulassel
,
G.
Ghattas
,
J. M.
Brenchley
, et al
.
2009
.
HIV reservoir size and persistence are driven by T cell survival and homeostatic proliferation.
Nat. Med.
15
:
893
900
.
23
Fromentin
,
R.
,
W.
Bakeman
,
M. B.
Lawani
,
G.
Khoury
,
W.
Hartogensis
,
S.
DaFonseca
,
M.
Killian
,
L.
Epling
,
R.
Hoh
,
E.
Sinclair
, et al
.
2016
.
CD4+ T cells expressing PD-1, TIGIT and LAG-3 contribute to HIV persistence during ART.
PLoS Pathog.
12
: e1005761.
24
McGary
,
C. S.
,
C.
Deleage
,
J.
Harper
,
L.
Micci
,
S. P.
Ribeiro
,
S.
Paganini
,
L.
Kuri-Cervantes
,
C.
Benne
,
E. S.
Ryan
,
R.
Balderas
, et al
.
2017
.
CTLA-4+PD-1- memory CD4+ T cells critically contribute to viral persistence in antiretroviral therapy-suppressed, SIV-infected rhesus macaques.
Immunity
47
:
776
788.e5
.
25
Wherry
,
E. J.
,
M.
Kurachi
.
2015
.
Molecular and cellular insights into T cell exhaustion.
Nat. Rev. Immunol.
15
:
486
499
.
26
Evans
,
V. A.
,
R. M.
van der Sluis
,
A.
Solomon
,
A.
Dantanarayana
,
C.
McNeil
,
R.
Garsia
,
S.
Palmer
,
R.
Fromentin
,
N.
Chomont
,
R. P.
Sékaly
, et al
.
2018
.
Programmed cell death-1 contributes to the establishment and maintenance of HIV-1 latency.
AIDS
32
:
1491
1497
.
27
Fromentin
,
R.
,
S.
DaFonseca
,
C. T.
Costiniuk
,
M.
El-Far
,
F. A.
Procopio
,
F. M.
Hecht
,
R.
Hoh
,
S. G.
Deeks
,
D. J.
Hazuda
,
S. R.
Lewin
, et al
.
2019
.
PD-1 blockade potentiates HIV latency reversal ex vivo in CD4+ T cells from ART-suppressed individuals.
Nat. Commun.
10
:
814
821
.
28
Wykes
,
M. N.
,
S. R.
Lewin
.
2018
.
Immune checkpoint blockade in infectious diseases.
Nat. Rev. Immunol.
18
:
91
104
.
29
Wolchok
,
J. D.
,
V.
Chiarion-Sileni
,
R.
Gonzalez
,
P.
Rutkowski
,
J. J.
Grob
,
C. L.
Cowey
,
C. D.
Lao
,
J.
Wagstaff
,
D.
Schadendorf
,
P. F.
Ferrucci
, et al
.
2017
.
Overall survival with combined nivolumab and ipilimumab in advanced melanoma.
N. Engl. J. Med.
377
:
1345
1356
.
30
Uldrick
,
T. S.
,
P. H.
Goncalves
,
M.
Abdul-Hay
,
A. J.
Claeys
,
B.
Emu
,
M. S.
Ernstoff
,
S. P.
Fling
,
L.
Fong
,
J. C.
Kaiser
,
A. M.
Lacroix
, et al
Cancer Immunotherapy Trials Network (CITN)-12 Study Team
.
2019
.
Assessment of the safety of pembrolizumab in patients with HIV and advanced cancer-a phase 1 study.
JAMA Oncol
. DOI: 10.1001/jamaoncol.2019.2244.
31
Husnain
,
M.
,
W.
Park
,
J. C.
Ramos
,
T. E.
Johnson
,
J.
Chan
,
A.
Dasari
,
R.
Mudad
,
P. J.
Hosein
.
2018
.
Complete response to ipilimumab and nivolumab therapy in a patient with extensive extrapulmonary high-grade small cell carcinoma of the pancreas and HIV infection.
J. Immunother. Cancer
6
:
66
71
.
32
Colston
,
E.
,
D.
Grasela
,
D.
Gardiner
,
R. P.
Bucy
,
B.
Vakkalagadda
,
A. J.
Korman
,
I.
Lowy
.
2018
.
An open-label, multiple ascending dose study of the anti-CTLA-4 antibody ipilimumab in viremic HIV patients.
PLoS One
13
: e0198158.
33
Cook
,
M. R.
,
C.
Kim
.
2019
.
Safety and efficacy of immune checkpoint inhibitor therapy in patients with HIV infection and advanced-stage cancer: a systematic review.
JAMA Oncol.
5
:
1049
1054
.
34
Chang
,
E.
,
A. L.
Sabichi
,
J. R.
Kramer
,
C.
Hartman
,
K. E.
Royse
,
D. L.
White
,
N. R.
Patel
,
P.
Richardson
,
S. V.
Yellapragada
,
J. M.
Garcia
,
E. Y.
Chiao
.
2018
.
Nivolumab treatment for cancers in the HIV-infected population.
J. Immunother.
41
:
379
383
.
35
Al Homsi
,
M. U.
,
M.
Mostafa
,
K.
Fahim
.
2018
.
Favorable response to treatment with avelumab in an HIV-positive patient with advanced merkel cell carcinoma previously refractory to chemotherapy.
Case Rep. Oncol.
11
:
467
475
.
36
Bari
,
S.
,
J.
Muzaffar
,
A.
Chan
,
S. R.
Jain
,
A. M.
Haider
,
M.
Adams Curry
,
C. J.
Hostler
.
2019
.
Outcomes of programmed cell death protein 1 (PD-1) and programmed death-ligand 1(PD-L1) inhibitor therapy in HIV patients with advanced cancer. [Published erratum appears in 2019 J. Oncol. 2019: 7921582.]
J. Oncol.
2019
: 2989048.
37
Spano
,
J. P.
,
M.
Veyri
,
A.
Gobert
,
A.
Guihot
,
P.
Perré
,
M.
Kerjouan
,
S.
Brosseau
,
N.
Cloarec
,
H.
Montaudié
,
C.
Helissey
, et al
.
2019
.
Immunotherapy for cancer in people living with HIV: safety with an efficacy signal from the series in real life experience.
AIDS
33
:
F13
F19
.
38
Webster
,
R. M.
2014
.
The immune checkpoint inhibitors: where are we now?
Nat. Rev. Drug Discov.
13
:
883
884
.
39
Wightman
,
F.
,
A.
Solomon
,
S. S.
Kumar
,
N.
Urriola
,
K.
Gallagher
,
B.
Hiener
,
S.
Palmer
,
C.
Mcneil
,
R.
Garsia
,
S. R.
Lewin
.
2015
.
Effect of ipilimumab on the HIV reservoir in an HIV-infected individual with metastatic melanoma.
AIDS
29
:
504
506
.
40
Scully
,
E. P.
,
R. L.
Rutishauser
,
C. R.
Simoneau
,
H.
Delagrèverie
,
Z.
Euler
,
C.
Thanh
,
J. Z.
Li
,
H.
Hartig
,
S.
Bakkour
,
M.
Busch
, et al
.
2018
.
Inconsistent HIV reservoir dynamics and immune responses following anti-PD-1 therapy in cancer patients with HIV infection.
Ann. Oncol.
29
:
2141
2142
.
41
Guihot
,
A.
,
A. G.
Marcelin
,
M. A.
Massiani
,
A.
Samri
,
C.
Soulié
,
B.
Autran
,
J. P.
Spano
.
2018
.
Drastic decrease of the HIV reservoir in a patient treated with nivolumab for lung cancer.
Ann. Oncol.
29
:
517
518
.
42
Cameron
,
P. U.
,
S.
Saleh
,
G.
Sallmann
,
A.
Solomon
,
F.
Wightman
,
V. A.
Evans
,
G.
Boucher
,
E. K.
Haddad
,
R. P.
Sekaly
,
A. N.
Harman
, et al
.
2010
.
Establishment of HIV-1 latency in resting CD4+ T cells depends on chemokine-induced changes in the actin cytoskeleton.
Proc. Natl. Acad. Sci. USA
107
:
16934
16939
.
43
Saleh
,
S.
,
F.
Wightman
,
S.
Ramanayake
,
M.
Alexander
,
N.
Kumar
,
G.
Khoury
,
C.
Pereira
,
D.
Purcell
,
P. U.
Cameron
,
S. R.
Lewin
.
2011
.
Expression and reactivation of HIV in a chemokine induced model of HIV latency in primary resting CD4+ T cells.
Retrovirology
8
:
80
91
.
44
Evans
,
V. A.
,
N.
Kumar
,
A.
Filali
,
F. A.
Procopio
,
O.
Yegorov
,
J. P.
Goulet
,
S.
Saleh
,
E. K.
Haddad
,
C.
da Fonseca Pereira
,
P. C.
Ellenberg
, et al
.
2013
.
Myeloid dendritic cells induce HIV-1 latency in non-proliferating CD4+ T cells. [Published erratum appears in 2014 PLoS Pathog. 10: e1004486.]
PLoS Pathog.
9
: e1003799.
45
Kumar
,
N. A.
,
K.
Cheong
,
D. R.
Powell
,
C.
da Fonseca Pereira
,
J.
Anderson
,
V. A.
Evans
,
S. R.
Lewin
,
P. U.
Cameron
.
2015
.
The role of antigen presenting cells in the induction of HIV-1 latency in resting CD4(+) T-cells.
Retrovirology
12
:
76
92
.
46
Yamamoto
,
T.
,
Y.
Tsunetsugu-Yokota
,
Y. Y.
Mitsuki
,
F.
Mizukoshi
,
T.
Tsuchiya
,
K.
Terahara
,
Y.
Inagaki
,
N.
Yamamoto
,
K.
Kobayashi
,
J.
Inoue
.
2009
.
Selective transmission of R5 HIV-1 over X4 HIV-1 at the dendritic cell-T cell infectious synapse is determined by the T cell activation state.
PLoS Pathog.
5
: e1000279.
47
Das
,
A. T.
,
B.
Klaver
,
B.
Berkhout
.
1999
.
A hairpin structure in the R region of the human immunodeficiency virus type 1 RNA genome is instrumental in polyadenylation site selection.
J. Virol.
73
:
81
91
.
48
Reed
,
L. J.
,
H.
Muench
.
1938
.
A simple method of estimating fifty per cent endpoints.
Am. J. Epidemiol.
27
:
493
497
.
49
Pardoll
,
D. M.
2012
.
The blockade of immune checkpoints in cancer immunotherapy.
Nat. Rev. Cancer
12
:
252
264
.
50
Kumar
,
N. A.
,
R. M.
van der Sluis
,
T.
Mota
,
R.
Pascoe
,
V. A.
Evans
,
S. R.
Lewin
,
P. U.
Cameron
.
2018
.
Myeloid dendritic cells induce HIV latency in proliferating CD4+ T cells.
J. Immunol.
201
:
1468
1477
.
51
Kyi
,
C.
,
M. A.
Postow
.
2016
.
Immune checkpoint inhibitor combinations in solid tumors: opportunities and challenges.
Immunotherapy
8
:
821
837
.
52
Ferry
,
T.
,
D.
Thomas
,
T.
Perpoint
,
G.
Lina
,
G.
Monneret
,
I.
Mohammedi
,
C.
Chidiac
,
D.
Peyramond
,
F.
Vandenesch
,
J.
Etienne
.
2008
.
Analysis of superantigenic toxin Vbeta T-cell signatures produced during cases of staphylococcal toxic shock syndrome and septic shock.
Clin. Microbiol. Infect.
14
:
546
554
.
53
Spina
,
C. A.
,
J.
Anderson
,
N. M.
Archin
,
A.
Bosque
,
J.
Chan
,
M.
Famiglietti
,
W. C.
Greene
,
A.
Kashuba
,
S. R.
Lewin
,
D. M.
Margolis
, et al
.
2013
.
An in-depth comparison of latent HIV-1 reactivation in multiple cell model systems and resting CD4+ T cells from aviremic patients.
PLoS Pathog.
9
: e1003834.
54
Trautmann
,
L.
,
L.
Janbazian
,
N.
Chomont
,
E. A.
Said
,
S.
Gimmig
,
B.
Bessette
,
M. R.
Boulassel
,
E.
Delwart
,
H.
Sepulveda
,
R. S.
Balderas
, et al
.
2006
.
Upregulation of PD-1 expression on HIV-specific CD8+ T cells leads to reversible immune dysfunction. [Published erratum appears in 2006 Nat. Med. 12: 1329.]
Nat. Med.
12
:
1198
1202
.
55
Kaufmann
,
D. E.
,
B. D.
Walker
.
2009
.
PD-1 and CTLA-4 inhibitory cosignaling pathways in HIV infection and the potential for therapeutic intervention.
J. Immunol.
182
:
5891
5897
.
56
Tong-Starkesen
,
S. E.
,
P. A.
Luciw
,
B. M.
Peterlin
.
1989
.
Signaling through T lymphocyte surface proteins, TCR/CD3 and CD28, activates the HIV-1 long terminal repeat.
J. Immunol.
142
:
702
707
.
57
Stevenson
,
M.
,
T. L.
Stanwick
,
M. P.
Dempsey
,
C. A.
Lamonica
.
1990
.
HIV-1 replication is controlled at the level of T cell activation and proviral integration.
EMBO J.
9
:
1551
1560
.
58
Mbonye
,
U.
,
J.
Karn
.
2017
.
The molecular basis for human immunodeficiency virus latency.
Annu. Rev. Virol.
4
:
261
285
.
59
Krummel
,
M. F.
,
J. P.
Allison
.
1995
.
CD28 and CTLA-4 have opposing effects on the response of T cells to stimulation.
J. Exp. Med.
182
:
459
465
.
60
Ho
,
Y. C.
,
L.
Shan
,
N. N.
Hosmane
,
J.
Wang
,
S. B.
Laskey
,
D. I.
Rosenbloom
,
J.
Lai
,
J. N.
Blankson
,
J. D.
Siliciano
,
R. F.
Siliciano
.
2013
.
Replication-competent noninduced proviruses in the latent reservoir increase barrier to HIV-1 cure.
Cell
155
:
540
551
.
61
Yukl
,
S. A.
,
P.
Kaiser
,
P.
Kim
,
S.
Telwatte
,
S. K.
Joshi
,
M.
Vu
,
H.
Lampiris
,
J. K.
Wong
.
2018
.
HIV latency in isolated patient CD4+ T cells may be due to blocks in HIV transcriptional elongation, completion, and splicing.
Sci. Transl. Med.
10
:
eaap9927
9946
.
62
Fraser
,
J. D.
2011
.
Clarifying the mechanism of superantigen toxicity.
PLoS Biol.
9
: e1001145.
63
Mylvaganam
,
G. H.
,
G.
Silvestri
,
R. R.
Amara
.
2015
.
HIV therapeutic vaccines: moving towards a functional cure.
Curr. Opin. Immunol.
35
:
1
8
.
64
McNeel
,
D. G.
,
J. C.
Eickhoff
,
E.
Wargowski
,
C.
Zahm
,
M. J.
Staab
,
J.
Straus
,
G.
Liu
.
2018
.
Concurrent, but not sequential, PD-1 blockade with a DNA vaccine elicits anti-tumor responses in patients with metastatic, castration-resistant prostate cancer.
Oncotarget
9
:
25586
25596
.
65
Marsden
,
M. D.
,
B. A.
Loy
,
X.
Wu
,
C. M.
Ramirez
,
A. J.
Schrier
,
D.
Murray
,
A.
Shimizu
,
S. M.
Ryckbosch
,
K. E.
Near
,
T. W.
Chun
, et al
.
2017
.
In vivo activation of latent HIV with a synthetic bryostatin analog effects both latent cell “kick” and “kill” in strategy for virus eradication.
PLoS Pathog.
13
: e1006575.
66
Gutiérrez
,
C.
,
S.
Serrano-Villar
,
N.
Madrid-Elena
,
M. J.
Pérez-Elías
,
M. E.
Martín
,
C.
Barbas
,
J.
Ruipérez
,
E.
Muñoz
,
M. A.
Muñoz-Fernández
,
T.
Castor
,
S.
Moreno
.
2016
.
Bryostatin-1 for latent virus reactivation in HIV-infected patients on antiretroviral therapy.
AIDS
30
:
1385
1392
.

S.R.L. has received honoraria to her institution for participation in advisory boards and or educational seminars from Gilead Sciences, Merck, Viiv Healthcare and Abbvie. S.R.L. has received investigator initiated research grant support from Gilead Sciences, Merck and Viiv Healthcare. The other authors have no financial conflicts of interest.

Supplementary data