Next Article in Journal
Viral Shrimp Diseases Listed by the OIE: A Review
Previous Article in Journal
HBV-RNA, Quantitative HBsAg, Levels of HBV in Peripheral Lymphocytes and HBV Mutation Profiles in Chronic Hepatitis B
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Expression Profile and Biological Role of Immune Checkpoints in Disease Progression of HIV/SIV Infection

1
Beijing Key Laboratory for Animal Models of Emerging and Re-Emerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Beijing 100021, China
2
NHC Key Laboratory of Human Disease Comparative Medicine, Comparative Medicine Center, Peking Union Medical College, Beijing 100021, China
3
Center for Acquired ImmunoDeficiency Syndrome Research, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
*
Author to whom correspondence should be addressed.
Submission received: 5 February 2022 / Revised: 9 March 2022 / Accepted: 9 March 2022 / Published: 11 March 2022
(This article belongs to the Topic Infectious Diseases)

Abstract

:
During HIV/SIV infection, the upregulation of immune checkpoint (IC) markers, programmed cell death protein-1 (PD-1), cytotoxic T-lymphocyte-associated antigen-4 (CTLA-4), T cell immunoglobulin and ITIM domain (TIGIT), lymphocyte-activation gene-3 (LAG-3), T cell immunoglobulin and mucin domain-3 (Tim-3), CD160, 2B4 (CD244), and V-domain Ig suppressor of T cell activation (VISTA), can lead to chronic T cell exhaustion. These ICs play predominant roles in regulating the progression of HIV/SIV infection by mediating T cell responses as well as enriching latent viral reservoirs. It has been demonstrated that enhanced expression of ICs on CD4+ and CD8+ T cells could inhibit cell proliferation and cytokine production. Overexpression of ICs on CD4+ T cells could also format and prolong HIV/SIV persistence. IC blockers have shown promising clinical results in HIV therapy, implying that targeting ICs may optimize antiretroviral therapy in the context of HIV suppression. Here, we systematically review the expression profile, biological regulation, and therapeutic efficacy of targeted immune checkpoints in HIV/SIV infection.

1. Introduction

T cell exhaustion due to constant antigen stimulation results in chronic cellular dysfunction. It occurs during chronic infection or cancer when the same antigens repeatedly stimulate T cells, leading to a dysfunctional state [1]. During the process of chronic T cell exhaustion, T cell reactivation gradually declines along with enhanced expression of immune checkpoint (IC) markers. IC markers, primarily PD-1 (programmed cell death protein-1), CTLA-4 (cytotoxic T-lymphocyte-associated antigen-4), TIGIT (T cell immunoglobulin and ITIM domain), LAG-3 (lymphocyte-activation gene-3), Tim-3 (T cell immunoglobulin and mucin domain-3), CD160, 2B4 (CD244), BTLA (B and T lymphocyte attenuator) and VISTA (V-domain Ig suppressor of T cell activation), interact with their respective ligands to ”symphonically” switch T cell activation into T cell exhaustion [2,3]. By balancing co-stimulatory signals and co-inhibitory signals, IC markers guarantee T cell activation, proliferation, and differentiation in response to antigenic stimulation from invading microbes [4].
How increasing IC markers maintains T cell dysfunction and exhaustion has been extensively investigated in cancer [5,6]; by contrast, the mechanism of how IC markers on T cells affect the disease progression in viral infections, particularly in human immunodeficiency virus (HIV), requires more exploration [2,5,7]. In HIV infection, checkpoint molecules were reported to be overexpressed on both CD4+ T cells and CD8+ T cells in general [8]. Moreover, the regulation of IC marker expression was correlated with HIV disease progression, as reflected in viral RNA replication, T cell function, and HIV reservoir enrichment. In this review, we systematically compared the expression of T cell checkpoint markers with the stage of HIV infection to assess their potential as therapeutic targets.

2. HIV Infection Upregulates the Expression of Immune Checkpoints in T Cells

The expression of ICs on T cells varies according to the stage and the level of host immunity against HIV infection. PD-1 is a critical IC marker that is consistently upregulated during HIV infection. The percentage of PD-1+ CD4+ T cells in HIV-infected individuals (both long-term nonprogressors and common infectors) has been found to be significantly higher than that in healthy persons [9,10,11,12]. A 10% average increase in PD-1+ CD4+ T cells was seen in HIV-infected hosts as compared with HIV-naïve controls [10,13]. Among HIV-infected individuals, long-term nonprogressors (LTNPs) displayed a much lower percentage of PD-1+ CD4+ T cells (6–21%) as compared with viremic patients (14–50%) [9,11,12,14,15,16]. The percentage of PD-1+ CD4+ T cells decreased to 10–35% in peripheral blood mononuclear cells (PBMCs) from common infectors with antiretroviral therapy (ART) [9,11,12,15,17,18]. Similarly, the proportion of PD-1+ CD8+ T cells has been shown to be 5–23%, 11–20%, 21–50%, and 10–30% in healthy persons, long-term nonprogressors, viremic patients, and ART-treated HIV-infected patients, respectively [9,10,12,14,15,18,19,20,21,22,23]. These data indicate that HIV infection upregulated PD-1 expression on CD4+ and CD8+ T cells dependent of ART treatment. Except for PD-1, numerous IC markers including CTLA-4, TIGIT, LAG-3, Tim-3, CD160, 2B4, and VISTA have been found to be regulated during HIV infection in general. Moreover, the majority of these IC markers showed regulatory trends similar to those of PD-1 (data in regular type in Table 1). In contrast, Zheng Zhang et al. found that HIV infection downregulated BTLA expression. It was shown that BTLA was reduced from 85% to 60–75% on CD4+ T cells and from 69% to 21–42% on CD8+ T cells in ART-naïve, HIV-infected patients as compared with healthy controls [24].
The underlying mechanism of regulation of IC expression mediated by HIV infection is still unknown and needs further investigation. How HIV regulates PD-1/CTLA-4/Tim-3 expression in CD4+ T cells is partially understood, as the HIV Nef protein upregulates PD-1 transcription in HIV-infected CD4+ T cell by activating p38 MAPK [25]. However, the Nef protein has also been reported to downregulate CTLA-4 expression in HIV-infected CD4+ T cell to provide optimal cellular conditions for cell activation and viral replication [26]. Regarding the regulation of Tim-3 by HIV, evidence has shown that the transmembrane domain of the HIV Vpu protein could downregulate Tim-3, while the di-leucine motif (LL164/165) of the HIV Nef protein upregulated Tim-3 expression on the surface of HIV-infected CD4+ T cells in vitro [27,28].
Non-human primate animal models that are widely used in studying HIV/SIV disease have shown different results as compared with humans. For example, SIV-infected rhesus macaques have relatively stable expression of PD-1, CTLA-4, TIGIT, LAG-3, and Tim-3 on CD4+ and CD8+ T cells with little change in expression (data in bold type in Table 1).
Table 1. Expression profile of immune checkpoint markers on T cells from HIV/SIV-infected individuals.
Table 1. Expression profile of immune checkpoint markers on T cells from HIV/SIV-infected individuals.
IC
Marker
Healthy ControlsHIV-Infected IndividualsLong-Term NonprogressorsViremic IndividualsART-Treated HIV-Infected Individuals
CD4+ T CellsCD8+ T CellsCD4+ T CellsCD8+ T CellsCD4+ T CellsCD8+ T CellsCD4+ T CellsCD8+ T CellsCD4+ T CellsCD8+ T Cells
PD-15–20% [9,10,11,12]
RM: 12% [29]
5–23% [9,10,12,19,21,23,30]10% [10]10% [10]6–21% [9,11,16,31]11–20% [9,31]14–50% [9,11,12,14,15,16,31]
RM: 6% [29]
21–50% [9,12,14,15,19,20,21,23,30,31]10–35% [9,11,12,15,17,18]
RM: 10% [29]
10–30% [9,11,12,15,18,21,23,30]
TIGIT10–18% [10,11,32]30% [10,32,33]
RM: 20% [32]
20% [10]50% [10]
RM: 40% [32]
18–19% [11,32]50–57% [32,33]21–30% [11,32]65–70% [32,33]17–20% [11,17,32]45–60% [32,33]
CTLA-41–6% [11,12,34,35]
RM: 4% [29]
1–2% [12,35]6%-11% [34,35]1% [35]3–7% [11,16,31]0.67% [31]2–10% [11,12,16,31]
RM: 2% [29]
0.37, 10–12% [12]1–8% [11,12,34]
RM: 1% [29]
5% [12]
LAG-36% [36]
RM: 9% [36]
5–7% [30,36]
RM: 9% [36]
N/AN/A0.065% [31]0.047% [31]0.021,14–48% [14,31,36]
RM: 12% [36]
0.004,8–32% [14,30,31,36]
RM: 15% [36]
12% [17]4% [30]
Tim-33–15% [10,37]
RM: 1% [38]
5–29% [10,37]
RM: 5% [38]
5% [10]5% [10]22% [37]30% [37]30–41% [14,31,37]
RM: 2% [38]
14–59% [14,20,37]
RM: 5% [38]
0.8% [17]N/A
CD1603% [10]1–10% [10,21,30,39]12% [10]20% [10]N/AN/AN/A15–40% [21,30,39]1.1% [17]10–16% [21,30]
2B43% [10]40–57% [10,21,30]10% [10]65% [10]5% [31]70% [31]10% [31]75–85% [21,30,31]9.5% [17]60–70% [21,30]
BTLA85% [24]69% [24]N/AN/A75% [24]42% [24]60% [24]21% [24]N/AN/A
VISTA3% [10]5% [10]8% [10]10% [10]N/AN/AN/AN/AN/AN/A
RM, rhesus monkey; ART, active antiretroviral therapy; PD-1, programmed cell death protein-1; TIGIT, T cell immunoglobulin and ITIM domain; CTLA-4, cytotoxic T-lymphocyte-associated antigen-4; LAG-3, lymphocyte-activation gene-3, Tim-3, T cell immunoglobulin and mucin domain-3, BTLA, B and T lymphocyte attenuator; VISTA, V-domain Ig suppressor of T cell activation; N/A, not assessed.

3. Immune Checkpoints Are Associated with Progression of Disease in HIV/SIV Infection

IC expression has been found to be highly correlated with disease progression after HIV/SIV infection, involving activation, proliferation, and function of T cells (Figure 1), and formation and persistence of HIV/SIV latency (Figure 2, Table 2).

3.1. Correlation between ICs from CD28 Superfamily and Progression of Disease in HIV/SIV Infection

PD-1, CTLA-4, and BTLA, members of the CD28 superfamily, are characterized by an immune receptor tyrosine-based inhibitory motif (ITIM) domain in the cytoplasm; PD-1, CTLA-4, and BTLA bind to PD-L1/2, B7, and herpes virus entry mediator (HVEM), respectively, to regulate progression of disease from HIV/SIV infection via transmission of inhibitory signals. PD-1 expressed on CD4+ and CD8+ T cells is positively correlated with plasma viral RNA (vRNA) replication and negatively correlated with CD4+ T cell count in HIV infection [9,13,15,20,40,41,42]. In HIV-infected children, a high proportion of PD-1+ memory CD4+ and CD8+ T cells was also observed, correlating with vRNA, CD4/CD8 ratio, and immune activation [43]. PD-1 is also highly expressed on HIV-specific CD8+ T cells, with significant correlation to plasma viral load and CD4 T cell count [9,19,30,44].
It has been reported that overexpression of PD-1 impaired T cell proliferation and caused overactivation and exhaustion of T cells, ultimately leading to HIV progression [2]. In ART-treated HIV patients, both PD-1hi CD4+ T cells and PD-1hi CD8+ T cells were negatively regulated by HIV-specific T cell immune responses characterized by reduced IFN-γ secretion in response to HIV Gag and Env peptides [15]. Blockade of the PD-1/PD-L1 pathway, especially simultaneous blocking of PD-L1 and stimulation through CD28, significantly enhanced the proliferation of HIV-specific CD4+ T cells in ART-naïve, HIV-infected patients [40,45], indicating a synergistic effect by targeting both the inhibitory and costimulatory pathways [45]. In addition, upregulated PD-1 has been observed in HIV-specific CD8+ T cells isolated from typical progressors (TPs) but not in long-term nonprogressors (LTNPs), leading to lower IFN-γ levels from HIV-specific CD8+ T cells and impaired proliferation of HIV-specific effector memory CD8+ T cells [9]. PD-1hi HIV-specific CD8+ T cells were shown to accumulate poorly differentiated phenotypes (CD27hi, CD28lo, CD57lo, CD127lo, CCR7 and CD45RA) and showed lower production of IFN-γ, IL-2 and TNF-α in response to HIV peptides [44]. Blockade of the PD-1 pathway enhanced proliferation [9,40,44,46] and increased IFN-γ, IL-2 [23,40], TNF-α, granzyme B, and lymphotoxin-α production by HIV-specific CD8+ T cells [44]. Polyfunctional HIV Gag-specific CD8+ T cell responses to PD-1 blockade could be improved by metformin therapy [47].
In an SIV-infected macaque model, PD-1 was also found to be an essential checkpoint marker during SIV infection. Low PD-1 expression was found in memory and naive CD8+ T cells, whereas high PD-1 levels were seen in SIV-specific CD8+ T cells isolated from PBMCs, lymph nodes, spleen, and mucosa, which might be attributed to the differences in TCR response [48]. Proliferation was impaired in SIV-specific PD-1hi CD8+ T cells [48]. Blocking PD-1 by recombinant rhesus macaque PD-1 (rMamu-PD-1) or rMamu-PD-1-immunoglobulin G (IgG) on PBMCs isolated from SIV-infected rhesus macaques increased the SIV-specific proliferative responses of both CD4+ and CD8+ T cells [49].
CTLA-4 expression on CD4+ T cells has been shown to be closely correlated with HIV progression, and CTLA-4 inhibited cytokine production and proliferation of T cells in chronic HIV infection. The proportion of CTLA-4+ CD4+ T cells was inversely correlated with CD4+ T cell counts and the CD4/CD8 ratio in both ART-naive and ART-treated HIV-1-infected patients [11,34]. The higher proportion of CTLA-4+ CD4+ T cells co-expressing high levels of CCR5 and Ki-67 resulted in increased susceptibility to HIV infection [34]. A subset of CTLA-4+ memory CD4+ T cells was also positively correlated with T cell activation (single and dual CD38 and HLA-DR-expressing memory CD4+ T cells) during HIV infection, reconfirming that CTLA-4 is a hallmark of HIV disease [11]. Moreover, expression of CTLA-4 was upregulated in HIV-specific CD4+ T cells, but could be downregulated by ART [45,50]. The increase in CTLA-4+ HIV-specific CD4+ T cells was correlated with higher vRNA levels and lower CD4+ T cell counts in PBMCs [45,50]. The blockade of CTLA-4 partially rescued cytokine production and proliferation of CD4+ and CD8+ T cell subsets, as upregulated expression of IFN-γ, CD40L, IL-2, and TNF-α in both CD4+ T cells and CD8+ T cells [31], and restored the proliferation of HIV-specific CD4+ T cell producing IFN-γ and IL-2 [50].
The proportion of BTLAlow T cells has been shown to be positively correlated with HIV disease progression of HIV infection. Lower expression of BTLA on both T cells and HIV-specific CD8+ T cells was commonly found in HIV-infected patients either as TP or with AIDS, but rarely found in LTNP [24]. The proportion of BTLA+ CD4+ T cells was positively correlated with CD4+ T cell count, yet inversely correlated with vRNA and Ki-67 expression, respectively. Moreover, BTLA-mediated inhibitory signaling of CD4+ T cell activation as well as cytokine production was severely impaired in HIV-infected patients [24].

3.2. Correlation between ICs from CD4 Family Members and Disease Progression in HIV Infection

LAG-3, a member of the CD4 family, promotes T cell exhaustion by ligating to MHC II, and participates in HIV pathogenesis as a new predictor for disease progression of HIV infection. Tian et al. showed that the numbers of LAG-3+ CD4+ T cells and LAG-3+ CD8+ T cells were inversely correlated with CD4+ T cell counts, while the number of LAG-3+ CD4+ T cells was positively correlated with vRNA in HIV-positive individuals [36]. Another study found that the proportion of LAG-3+ CD8+ T cells was positively correlated with vRNA, but not correlated with CD4 T cell count [20]. A high proportion of LAG-3+ CD4+ and CD8+ T cells was also found to be related to continuous T cell activation, as indicated by co-expression with CD38 in HIV-1-infected individuals [36]. The putative correlation between LAG-3 expression and clinical indicators needs more data to support it.
The increased inhibitory signals transduced by LAG-3 have been shown to be related to functional T cell impairment, including enhanced production of IFN-γ, IL-2, IL-10, and TNF-α in HIV-1-infected subjects in the presence of a LAG-3 blocker, and increased HIV Gag-stimulated proliferation of HIV-specific IFN-γ+ CD8+ T and IFN-γ+ CD4+ T cells. Overexpression of LAG-3 consistently validated the inhibitory regulation of LAG-3 during HIV infection. The synergistic enhancement between the LAG-3/MHC II pathway and the PD-1/PD-L1 pathway strongly promoted the T cell-mediated immune response against HIV [36].

3.3. Correlation between TIGIT and Disease Progression in HIV/SIV Infection

TIGIT is a common immune receptor linked to PVR/Nectin2 that induces potent immunosuppression, but the correlation between TIGIT expression and HIV infection is controversial. Glen et al. showed that the number of TIGIT+ CD8+ T cells and TIGIT+ CD4+ T cells was increased in HIV-infected patients during all stages of AIDS [32]. However, the frequency of TIGIT+ CD8+ T cells and TIGIT+ CD4+ T cells was also increased in ART-naïve and long-term ART-treated patients with HIV infection as compared with elite controllers and healthy subjects [11,33]. In HIV-infected patients, the proportion of TIGIT+ CD8+ T cells was inversely correlated with the CD4 T cell count and the CD4/CD8 ratio [32,33]. Expression of TIGIT on T cells may be related to HIV pathogenesis by mediating HIV replication and host immune activation during HIV infection [32]. Among long-term ART-treated, HIV-infected patients, a positive correlation between the numbers of TIGIT+ CD4+ T cells and CD4+ T cell-associated HIV DNA was found. The frequencies of TIGIT+ CD8+ T cells and TIGIT+ CD4+ T cells were positively correlated with the frequency of CD38+ HLA-DR+ cells in HIV-infected individuals [11,32,33]. In vitro studies indicated that TIGIT might limit CD8+ T cell proliferation and function against HIV by a unique pathway [32,33]. However, a recent study proposed that TIGIT upregulated genes associated with antiviral immunity in CD8+ T cells, suggesting that TIGIT+ CD8+ T cells maintain an intrinsic cytotoxicity rather than succumbing to a state of immune exhaustion in HIV-infected individuals [51]. Rhesus TIGIT (rhTIGIT) partially mimics human TIGIT expression and function during HIV infection. In SIV-infected rhesus macaques, expression of rhTIGIT was significantly increased on CD8+ T cells derived from lymph nodes (LNs) and spleen but not PBMCs significantly [32]. RhTIGIT inhibited the proliferation and cytokine secretion of SIV-specific CD8+ T cells [32].

3.4. Correlation between Tim-3 and Disease Progression in HIV/SIV Infection

The numbers of Tim-3+ CD4+ and CD8+ T cells have been shown to be elevated in individuals with acute and chronic progression of HIV-1 infection, but not in patients with viral control, as compared with noninfected individuals. Expression of Tim-3 was upregulated on HIV-specific CD4+ and CD8+ T cells from HIV-1-infected patients with chronic progression [37,45,52], but this could be effectively reduced by ART. However, no significant increase in Tim-3 expression was found in an HIV-2 cohort [53]. As a potential predictor of HIV-1 disease progression, the expression of Tim-3 on CD4+ and CD8+ T cells correlated positively with vRNA and CD38 expression, but was inversely correlated with CD4+ T cell counts in ART-free HIV infectors [37]. ART strongly attenuated the positive correlation between Tim-3 expression on CD8+ T cells, vRNA, and CD4+ T cell count [37]; but, the significant association between Tim-3 and CD38 was unaffected by ART [37]. These linear correlations were also observed between plasma HIV viral load and expression of Tim-3 on HIV-specific CD4+ T cells [45].
Tim-3-expressing T cells are dysfunctional because of their low capacity to proliferate, produce cytokines, and degranulate in response to ligation of Tim-3 (galectin-9). In both HIV-1 infected patients and healthy individuals, Tim-3 was shown to moderately inhibit IFN-γ, TNF-α, and CD107a production in total and HIV-specific Tim-3+ CD4+ T cells and Tim-3+ CD8+ T cells [37,54]. In chronic persistent HIV infection, the proliferation of Tim-3low CD8+ T cells was higher than that of their Tim-3hi counterparts [37]. Blocking Tim-3 enhanced proliferation and cytotoxicity in both HIV-specific CD4+ and CD8+ T cells [37,46,54]. Recently, Tim-3 was found to be a natural immune inhibitor against viral spread [27].
In SIV-infected rhesus monkeys, the percentage of Tim-3+ CD8+ T cells was significantly higher in LNs than in PBMCs [38]. Amancha et al. found that Tim-3 expression on CD4+ and CD8+ T cells followed a ”reversed U” curve in PBMCs during SIV infection [55]. Therefore, there was little linear correlation between PBMC/LN-derived Tim-3+ T cells and SIV viremia [38,55]. Interestingly, using Tim-3+/PD-1+ double markers, the expression of these dual ICs on LN-derived CD8+ T cells was positively correlated with SIV plasma viremia [38]. The mechanism of Tim-3 regulation of HIV-infected T cells remains controversial and further studies are needed to clarify the pathway [38,55].

3.5. Correlation between CD160 and Disease Progression in HIV/SIV Infection

The expression of CD160 has been shown to be increased in CD8+ T cells in ART-naïve HIV-1-infected individuals [39,56], which could be remarkably reduced by ART, particularly in naïve CD8+ T cells [21,30,56]. Unexpectedly, the subset of CD160+ CD8+ T cells was higher in HIV-1-infected slow progressors (SPs) and ECs as compared with TPs. These results differed from those obtained for PD-1+ and CTLA-4+ CD8+ T cells in HIV infection [39,57]. Expression of CD160 on CD8+ T cells was associated with slow progression of HIV infection, and showed a positive correlation with CD4 T cell counts, but a negative correlation with vRNA. It was concluded that enhanced CD160 expression on CD8+ T cells promoted cytotoxicity against HIV [39]. HIV-specific CD160+ CD8+ T cells were dominantly proliferative and highly activated in ART-naïve, HIV-infected individuals, with increasing expression of granzyme B and CD107a [39]. In contrast, CD160 signaling could attenuate CD8+ T cell immunity against HIV chronic infection, largely through immunosuppression of CD160-HVEM (herpes virus entry mediator). Blockade of the CD160/HVEM pathway restored proliferation and cytokine production from HIV-specific CD8+ T cells [56]. Moreover, HIV-specific CD8+ T cells co-expressing PD-1, CD160, or 2B4, which have been defined as exhausted phenotypes, presented poor activation, reduced intracellular cytokine production, and attenuated responses [30,56,57].
The expression of 2B4 on CD8+ T cells has been shown to be higher in ART-naïve HIV patients, and the expression could be reduced by ART [21,30,57]. The cross-linkage of 2B4 with CD48 was considered an inhibitory signal, which downregulated the functionality of CD4+ and CD8+ T cells during disease progression associated with HIV infection. T cells against HIV could be cooperatively attenuated by co-expression of 2B4 with PD-1 and/or CD160, which was inversely correlated with production of IFN-γ, IL-2, and TNF-α from HIV-specific CD8+ T cells [30]. The percentage of HIV-specific CD8+ T cells expressing both 2B4 and PD-1 was negatively correlated with perforin production [57]. Blockade of 2B4 elevated the CD40L responses on both CD4+ and CD8+ T cells, which could be exponentially augmented by PD-1 co-blockade in HIV-positive patients [31]. Co-blockade of PD-1/PD-L1 and 2B4/CD48 synergistically restored the proliferation of HIV-specific CD8+ T cells [30]. The findings indicated that 2B4, PD-1, and/or CD160 cooperatively exhausted CD8+ T cells against chronic HIV stimulation.
The expression of VISTA was significantly higher on both CD4+ and CD8+ T cells, with attenuated IFN-γ and TNF-α production in HIV-infected individuals [10].

3.6. Correlation between Co-Expression of Checkpoint Markers and Disease Progression in HIV Infection

PD-1 is an essential immune checkpoint marker, involved in multiple synergistic co-functions related to HIV-specific immune regulation. Numerous co-expressing phenotypes of checkpoint markers including PD-1+ TIGIT+ CD8+ T cells, PD-1+ TIGIT+ CD160+ 2B4+ CD8+ T cells, PD-1+ 2B4+ CD160+ Tim-3 CD8+ T cells, and PD-1+ TIGIT+ CTLA-4+ CD4+ T cells have been shown to be significantly increased in HIV-infected patients [21,22,32,33]. CTLA-4 was linked to PD-1 expression on HIV-specific CD4+ T cells [45,50]. HIV-specific CD4+ T cells co-expressing PD-1 and CTLA-4 were found at much higher percentages in viremic patients as compared with ECs [45,50]. Co-expressed checkpoints exhibited stronger correlations with HIV viral load and CD4 T cell counts. The percentage of memory CD4+ T cells co-expressing PD-1, CTLA-4, and TIGIT was robustly correlated with CD4 T cell counts, vRNA, CD4/CD8 ratio, and CD4+ T cell activation during HIV infection [11]. In addition, PD-1+ TIGIT+ CD8+ T cells were inversely correlated with CD4 T cell counts, but positively correlated with vRNA [32]. HIV-specific CD4+ T cells co-expressing PD-1, CTLA-4, and Tim-3 exhibited greater aggregate correlation with vRNA [45]. A modest positive correlation between the percentage of PD-1+ CD160+ 2B4+ LAG-3 HIV-specific CD8+ T cells and virus load was shown during chronic HIV infection [30]. In T cells from HIV-viremic individuals, multiple expression of checkpoint markers including PD-1+ CD160+ CD8+ T cells, PD-1+ CD160+ 2B4+ CD8+ T cells, PD-1+ TIGIT+ CD8+ T cells, and PD-1+ Tim-3+ T cells were found to be associated with lower expression of cytokines against HIV as compared with T cells expressing single checkpoint markers [30,32,52,56]. In particular, PD-1+ CD160+ double pathways co-enhanced in CD8+ T cells resulted in poor survival [56]. The in vitro response of HIV-specific CD8+ T cells was better restored via blockade of PD-1/PD-L1 synergizing with TIGIT/CD155, 2B4/CD48, Tim-3/galectin-9, or BTLA/HVEM as compared with that from single blockade of PD-1/PD-L1 [30,32,46,52]. HIV-specific CD4+ T cell function was improved through co-blockade of PD-L1 with Tim-3 [52]. In summary, the synergistic effects of co-inhibition of these checkpoint markers were remarkable, but more investigation is warranted.

3.7. Correlation between Immune Checkpoint Markers and HIV/SIV Latency

HIV reservoirs remain largely understudied. Accumulating proof has shown that checkpoint markers play fundamental roles in maintaining HIV latency (Figure 2). The baseline levels of the checkpoint markers PD-1, Tim-3, and LAG-3 have been shown to be linearly correlated with total baseline HIV-1 DNA, which was predictive of the HIV DNA reservoir after ART, as well as time to rebound after ART withdrawal [14,58]. Such a correlation indicated that checkpoint markers represented the exhaustive dynamics of T cells interacting with HIV reservoirs. Expression of PD-1, TIGIT, and LAG-3 on CD4+ T cells was assumed to be positively correlated with the number of persistently infected CD4+ T cells considered to be the essential part of the HIV reservoir in patients controlled with successful ART [17,59,60]. PD-1 expressed on lymph node CD4+ T cells was characterized as a major source of replication-competent HIV, which presents a serious dilemma for achieving a functional cure [61,62,63].
In SIV-infected rhesus macaques, CTLA-4+ PD-1 memory CD4+ T cells were found with a large majority of cell-associated SIV Gag DNA in PBMCs, lymph nodes, spleen, and gut tissues in monkeys successfully controlled with ART. CTLA-4+ PD-1 memory CD4+ T cells isolated from lymph nodes were considered to be a major part of the persistent viral reservoir [64]. In contrast, we recently reported that LP-98 (a HIV fusion-inhibitory lipopeptide) monotherapy showed extremely potent antiviral efficacy in SHIVSF162P3-infected rhesus macaques. Interestingly, we found that a higher proportion of PD-1+ resting CD4+ central memory T cells was measured on both superficial lymph nodes and deep lymph nodes of stable virologic rebound (SVR) macaques as compared with that of stable virologic control (SVC) after withdrawal from LP-98 treatment, highlighting the close relationship between PD-1 and the LN viral reservoir [65]. In HIV-infected humanized mice, PD-1+ CD4+ T cells and TIGIT+ CD4+ T cells originating from spleens were enriched with latent and reactivatable HIV-1 [66]. In vitro experiments also indicated that HIV latency was enriched in PD-1, CTLA-4, Tim-3, or BTLA expressing CD4+ T cells, which could also be reversed by blockade of ICs [67,68,69,70]. The biological effects of ICs on HIV latency have been investigated with various experimental models, but need to be further verified, and the precise conclusions should be summarized for clinical application.
Table 2. Biological role of immune checkpoint markers on T cell function in HIV/SIV infection.
Table 2. Biological role of immune checkpoint markers on T cell function in HIV/SIV infection.
IC MarkerFunction Mediated by HIV/SIV Infection
CD4+ T CellCD8+ T Cell
PD-1Proliferation
  • Impairs proliferation of HIV-specific CD4+ T cells [2,40,45].
  • Impairs proliferation of SIV-specific CD4+ T cells [49].
Proliferation
  • Impairs proliferation of HIV-specific effector memory CD8+ T cells and HIV-specific CD8+ T cells [2,9,40,44,46].
  • Impairs proliferation of SIV-specific CD8+ T cells [48,49].
Cytokine secretion
  • Reduces IFN-γ secretion by HIV-specific CD4+ T cells [15].
Cytokine secretion
  • Reduces IFN-γ, IL-2, TNF-α, granzyme B, and lymphotoxin-α secretion by HIV-specific CD8+ T cells [9,15,44].
Virus reservoirs
  • Contributes to HIV reservoirs in CD4+ T cells [14,17,59,60,61,62,63,67,68,69].
  • Contributes to SIV reservoirs in resting CD4+ central memory T cells in LNs [65].
TIGITVirus reservoirs
  • Contributes to HIV reservoirs in CD4+ T cells [17,59,60,68].
Proliferation
  • Impairs proliferation of HIV-specific CD8+ T cell [32].
  • Inhibits the proliferation of SIV-specific CD8+ T cells [32].
Cytokine secretion
  • Reduces IFN-γ, TNF-α, and IL-2 production in HIV-specific CD8+ T cells [32].
  • Increases perforin and granzyme B production in CD8+ T cells [32].
  • Decreases the frequency of polyfunctional CD107a+GrzB+IFN-γ+TNF+ as well CD107aGrzBIFN-γ+TNF HIV-specific CD8+ T cells [33].
  • Upregulates genes associated with antiviral immunity in CD8+ T cells [51].
  • Reduces IFN-γ secretion by SIV-specific CD8+ T cells [32].
CTLA-4Proliferation
  • Reduces the proliferation of HIV-specific CD4+ T cell producing IFN-γ and IL-2 [50].
Cytokine secretion
  • Reduces IFN-γ, CD40L, IL-2, and TNF-α by CD8+ T cells [31].
Cytokine secretion
  • Reduces IFN-γ, CD40L, IL-2 and TNF-α by CD4+ T cells [31].
Virus reservoirs
  • Contributes to SIV reservoirs in memory CD4+ T cells [64,68].
LAG-3Proliferation
  • Impairs HIV-specific CD4+ T cell proliferation [36].
Proliferation
  • Impairs HIV-specific CD8+ T cell proliferation [36].
Cytokine secretion
  • Reduces IFN-γ secretion by HIV-specific CD4+ T cell [36].
Cytokine secretion
  • Reduces IFN-γ secretion by HIV-specific CD8+ T cell [36].
Virus reservoirs
  • Contributes to HIV reservoirs in CD4+ T cells [14,17,59].
Tim-3Proliferation
  • Impairs proliferation of HIV-specific CD4+ T cells [37,46].
Proliferation
  • Impairs proliferation of CD8+ T cells and HIV-specific CD8+ T cells [37,38,46].
Cytokine secretion
  • Inhibits IFN-γ, TNF-α, and CD107a production in total and HIV-specific CD4+ T cells [37,54].
  • Increases IFN-γ, TNF-α, IL-2, and CD107a production in SIV-specific CD4+ T cells [55].
Cytokine secretion
  • Inhibits IFN-γ, TNF-α, and CD107a production in total and HIV-specific CD8+ T cells [37,54].
  • Decreases ability to release perforin and granzymes by HIV-specific CD8+ T cells [54].
  • Inhibits IFN-γ production in total and SIV-specific CD8+ T cells [38].
  • Increases IFN-γ, TNF-α, IL-2, and CD107a production in SIV-specific CD8+ T cells [55].
Virus reservoirs
  • Contributes to HIV reservoirs in CD4+ T cells [14,58,68].
CD160N/AProliferation
  • Impairs proliferation of HIV-specific CD8+ T cells [56].
  • Enhances proliferation of HIV-specific CD8+ T cells [39].
Cytokine secretion
  • Inhibits IFN-γ, TNF-α, and CD107a production in HIV-specific CD8+ T cells [56].
  • Increases CD107a and granzyme B expression in HIV-specific CD8+ T cells [39].
2B4Cytokine secretion
  • Reduces CD40L production in CD4+ T cells [31].
Proliferation
  • Impairs proliferation of HIV-specific CD8+ T cells [30].
Cytokine secretion
  • Reduces IFN-γ, TNF-α, IL-2, and perforin in HIV-specific CD8+ T cells [30,57].
  • Reduces CD40L in CD8+ T cells [31].
BTLACytokine secretion
  • Impairs BTLA-mediated suppression of IL-2 and IFN-γ production in CD4+ T cells [24].
N/A
Virus reservoirs
  • Contributes to HIV reservoirs in CD4+ T cells [68].
VISTACytokine secretion
  • Attenuates IFN-γ and TNF-α production in CD4+ T cells [10].
Cytokine secretion
  • Attenuates IFN-γ and TNF-α production in CD8+ T cells [10].

4. Therapeutic Effects of Immune Checkpoint Blockers on HIV/SIV Infection

Immune checkpoint blockers (ICBs) targeting PD-1 and CTLA-4 pathways combined with anti-HIV drugs have been tested in clinical and experimental trials to develop an effective therapeutic regimen (Table 3 and Table 4).

4.1. Immune Checkpoint Blockers Confer Partial Protection against Progression of HIV Infection

The efficacy and safety of ICBs against HIV infection have been reported to vary with pathological conditions [7,71,72,73,74,75,76] (Table 3). In HIV-infected patients with metastatic melanoma, a PD-1 blocker alone or combined with CTLA-4 blocker enhanced the function of HIV-1-specific CD8+ T cells and reversed HIV latency in CD4+ T cells. The CTLA-4 blocker increased CD4 or CD8 T cell counts, elevated the level of CD4+ T cell activation, and reversed HIV latency following drug infusions [67,77,78,79]. In HIV-infected patients with non-small cell lung cancer (NSCLC), a PD-1 blocker helped to restore HIV-specific CD8+ T cell function and maintain HIV viremia. However, the PD-1 blocker could not consistently improve CD4 T cell counts and reduce HIV reservoirs [80,81,82,83]. In ART-treated HIV-infected patients with other advanced-stage malignancies, the HIV suppression was inconsistent when used with PD-1 blocker alone [79,84,85]. It is worth noting that anti-PD-1 and anti-CTLA-4 combination therapy moderately reversed HIV latency and potentially eliminated the HIV reservoir in ART-suppressed people living with HIV with advanced malignancies [86]. Moreover, ICBs, including pembrolizumab, nivolumab, ipilimumab, and durvalumab caused no serious adverse events (AEs) to patients, suggesting that patients with HIV should no longer be excluded from clinical trials testing checkpoint inhibitor therapies for various cancers [67,78,80,81,82,83,84,85,87,88,89,90,91,92,93,94]. In HIV-1 infected patients without malignancies, the PD-L1 blocker was shown to enhance the HIV-1 Gag-specific CD8+ T cell response [89]. The CTLA-4 blocker reduced plasma viremia in proportion to drug concentration [88]. Safety is a significant challenge, as indicated by the immune-related adverse events (irAEs) that appeared in two clinical trials using a PD-1 blocker for HIV-infected individuals without malignancies [89,95]. Multiple injections of low doses of IC blocker might alleviate the side effects. At present, the differences in therapeutic regimens and the relatively small number of clinical cases might be the cause of the inconsistent outcomes. The tolerance and efficacy of IC blockers vary according to physiological state for HIV infectors with or without malignancies. Given that only clinical trial data for PD-1 and CTLA-4 blockers are available, the data from testing blockers against other ICs (TIGIT, LAG-3, Tim-3, CD160, 2B4, BTLA, and VISTA) may prove highly useful in treatment of AIDs. Therefore, how to combine specific IC blockers and other treatment strategies to design an effective therapeutic program for HIV infectors with or without malignancies is a problem worth studying. More tests of drug combinations in vitro, ex vivo, and in vivo are necessary, to provide the missing information on clinical efficacy for curing AIDS.
Table 3. Summary of therapeutic effects of IC blockers in HIV-infected patients.
Table 3. Summary of therapeutic effects of IC blockers in HIV-infected patients.
ReferenceIC blockerTargetObjectiveTreatmentOutcomes
Oscar Blanch-Lombarte [78]PembrolizumabPD-1ART HIV-1-infected individual with metastatic melanomaPembrolizumab (2 mg/kg/3 weeks)
  • Enhances HIV-1 specific-CD8+ T cell function.
  • Reduces HIV DNA transiently.
Vanessa A Evans [67]NivolumabPD-1ART HIV-infected individual with metastatic melanomaSingle intravenous infusion of
nivolumab (3 mg/kg)
  • Increases cell-associated HIV RNA in CD4+ T cells.
  • No change of HIV DNA or plasma HIV RNA.
Jillian S.Y. Lau [79]Nivolumab IpilimumabPD-1
CTLA-4
ART HIV-infected individual with metastatic melanomaIpilimumab (1 mg/kg/3 weeks) and
Nivolumab (3 mg/kg/3 weeks)
  • Increases cell-associated unspliced HIV RNA.
  • Increases HIV-specific central and effector memory CD8+ T cell function.
  • No consistent change of HIV DNA or the proportion of cells with inducible MS HIV RNA.
Fiona Wightman [77]IpilimumabCTLA-4 ART HIV-infected individual with metastatic melanomaIpilimumab (3 mg/kg, four doses/3 week)
  • No change in plasma HIV RNA overall.
  • Decreases plasma HIV RNA, increases CD4 T cell/CD8 T cell counts and enhances CD4+ T cell activation cyclically following each ipilimumab infusion.
  • Increases cell-associated unspliced HIV RNA following the first and second infusions.
A Guihot [80]NivolumabPD-1ART HIV-infected individual with NSCLCNivolumab (15 injections/14 days)
  • Increases plasma HIV RNA transiently.
  • Restores HIV Nef specific CD8+ T cells function.
  • Diminishes HIV reservoir.
M Hentrich [81]NivolumabPD-1ART HIV-infected individual with NSCLCChemoradiotherapy and surgical resection
Nivolumab (3 mg/kg)
  • Decreases CD4 T cell counts.
  • HIV-RNA remains undetectable.
  • Slows progressive disease.
Brennan McCullar [82]NivolumabPD-1ART HIV-infected individual with NSCLCOne cycle of carboplatin/paclitaxel
Definitive chemo-radiation with cisplatin and etoposide
Start nivolumab
  • CD4 T cell counts remain stable.
  • Viral load remains undetectable.
Gwenaëlle Le Garff [83]NivolumabPD-1ART HIV-infected individual with NSCLCDecompressive radiotherapy
Six cisplatin/gemcitabine and four Taxotere chemotherapy treatments
Start nivolumab
  • HIV viral load remains undetectable.
  • CD4 T cell and CD8 T cell counts transiently increased.
  • HIV-specific IFN-γ+ CD8+ T cells slightly increased.
  • Almost no change in HIV replication or reservoirs.
E P Scully [84]Nivolumab PembrolizumabPD-1ART HIV-1-infected individuals with malignanciesNivolumab (participant 1 with head and neck SCC, standard dosing, for 18 months)
Nivolumab (participant 2 with head and neck SCC, four doses)
Pembrolizumab (participant 3 with squamous cell carcinoma of the skin)
  • No consistent change in CD4 T cell counts or CD4+ T cell cell-associated HIV-1 RNA/DNA or plasma virus load.
  • No change in HIV-1 Gag specific T cell responses.
Neil J Shah [85]Nivolumab, Pembrolizumab, Atezolizumab, Durvalumab and AvelumabPD-1/PD-L1HIV-infected individuals with advanced-stage
cancers
Anti-PD-(L)1 monotherapy or anti-PD-(L)1 monotherapy combined with chemotherapy
  • No change in HIV viral load or CD4 T cell counts.
  • Toxicity and efficacy rates are similar to those observed in patients without HIV.
Thomas A. Rasmussen [86]Nivolumab IpilimumabPD-1
CTLA-4
ART HIV-infected individual with advanced malignanciesNivolumab (240 mg every 2 weeks) in combination with ipilimumab (1 mg/kg every 6 weeks)
  • Increases cell-associated unspliced HIV RNA.
  • Eliminates cells containing replication-competent HIV.
Cynthia L Gay [89]BMS-936559PD-L1ART HIV-1-infected adultsSingle infusions of BMS-936559 (0.3 mg/kg)
  • Increases HIV-1 Gag-specific CD8+ responses.
  • CD4 T cell counts remain unchanged.
  • No consistent change in HIV-1 DNA or RNA/DNA ratio.
Elizabeth Colston [88]IpilimumabCTLA-4 Chronic HIV-1
-infected individuals
Ipilimumab, 0.1, 1, or 3 mg/kg, two doses every 28 days; or 5 mg/kg, four doses every 28 days
  • Decreases plasma HIV-1 RNA of two patients treated with 0.1 mg/kg and 1 mg/kg ipilimumab.
  • Increases plasma HIV-1 RNA in 14 patients treated with 3 or 5 mg/kg ipilimumab.
  • No change in CD4 and CD8 T cell counts.

4.2. Immune Checkpoint Blockers Confer Partial Protection against Progression of SIV Infection

SIV-infected rhesus macaques are the most widely used nonhuman primate (NHP) models for exploring the molecular mechanism and the potential application of novel HIV therapies. Analysis of ICB outcomes in NHP models has provided an experimental foundation and theoretical basis for HIV clinical research (Table 4). In SIV-infected rhesus macaques, the PD-1 blocker enhanced proliferation and function of SIV-specific CD4+ and CD8+ T cells, as well as increased the proliferation of memory B cells with elevated SIV Env-specific antibody [96,97,98,99]. A PD-1 blocker together with ART reduced viremia to a low level, increased the CD8+ T cell response, and in general, decreased the rebound after ART withdrawal [98,100,101,102]. The CTLA-4 blocker decreased vRNA levels in lymph nodes and restored the effector function of SIV-specific CD4+ and CD8+ T cells during ART without impeding rebound after ART suspension [103]. However, Justin Harper et al. recently reported that the combination of PD-1 blocker and CTLA-4 blocker was more effective than single blockade alone in robust latency reversal, while neither PD-1 blocker nor CTLA blocker could enhance SIV-specific CD8+ T cell function [104].
Using a prophylactic model with PD-1 blocker, the vaccine-induced, SIV-specific CD8+ T cell and B cell response has been augmented, resulting in better control of pathogenic SIV infection [105,106,107]. However, PD-1 blockade combined with therapeutic vaccination might be a double-edged sword for SIV therapy. In chronically SIV-infected rhesus macaques under ART, Sheikh et al. discovered that a PD-1 blocker could be used to enhance the therapeutic effects of SIV vaccination by improving vaccine-induced CD8+ T cell function, sustaining B cell homing into follicles for vaccine-induced CD8+ T cell responses, reducing the viral reservoir in lymphoid tissue and, thereby, controlling HIV rebound upon ART interruption [108]. Surprisingly, Wu et al. found the opposite outcome: PD-1 blockade in combination with therapeutic vaccination led to viral reservoir activation that accelerated viral rebound in chronically SIV-infected macaques after ART interruption [109]. Therefore, the therapeutic potential of vaccine + PD-1 blockade might be closely related to ART treatment and the use of this drug combination still needs to be optimized in the SIV-infected rhesus macaque model for developing an HIV-1 cure in the future. Activation of the viral reservoir and enhancement of SIV-specific CD8+ T cell functions are the two dominant factors that need to be considered in the design of an immunotherapeutic strategy. Combining IC blockers with vaccine might produce better outcomes for HIV therapy, but may also raise more safety concerns. Much work is required to determine the best combination to control virus without excessive toxicity.
Table 4. Summary of therapeutic effects of IC blockers in macaque model.
Table 4. Summary of therapeutic effects of IC blockers in macaque model.
ReferenceIC BlockerTargetObjectiveTreatmentOutcomes
Vijayakumar
Velu [96]
Humanized mouse anti-human PD-1 Ab (clone EH12-1540)PD-1SIV251/SIVmac239-infected Indian rhesus macaquesAnti-PD-1 Ab (3 mg/kg) in early chronic phase and in late chronic phase on days 0, 3, 7, and 10
  • Increases SIV-specific CD8+ T cells with improved functionality.
  • Increases proliferation of memory B cells and SIV envelope-specific antibodies.
  • Reduces plasma viral load.
  • Prolongs survival of SIV-infected RMs.
Adam C
Finnefrock [107]
Anti-human
PD-1 Ab
(clone 1B8)
PD-1SIVmac239-infected rhesus macaquesTherapeutic model: single infusion
of anti-PD-1 Ab 1B8 (5 mg/kg) to chronic SIV-infected macaques before or during ART
Prophylactic model: anti-PD-1 Ab 1B8 (5 mg/kg) to naive macaques immunized with an SIV-Gag adenovirus vector vaccine
  • Therapeutic model: Transiently increases viral load; PD-1 blockade during ART has no discernible effect on SIV Gag-specific CD8+ T cells and CD4 T cell counts.
  • Prophylactic model: Enhances SIV Gag-specific CD8+ T responses.
Ravi Dyavar
Shetty [97]
Mouse anti-
human PD-1 Ab
PD-1SIV-infected rhesus macaques Anti-PD-1 Ab (3 mg/kg) at either
10 or 90 weeks after SIV infection
on 0, 3, 7, and 10 days
  • Downregulates type I IFN responses.
  • Enhances gut junction-associated gene expression and reduces microbial translocation.
  • Enhances immunity to pathogenic gut bacteria.
  • Prolongs survival of SIV-infected RMs.
Praveen K
Amancha [99]
Recombinant macaque PD-1 fused to macaque Ig-Fc (rPD-1-Fc)PD-1SIVmac239-infected rhesus macaquesrPD-1-Fc (50 mg/kg) alone or in combination with ART during the early chronic phase
  • Enhances SIV-specific CD4+ and CD8+ T cell proliferation and function.
  • Fails to alter plasma viremia.
  • Induces a significant delay in viral load rebound after ART interruption.
Geetha H
Mylvagana [98]
Primatized anti–PD-1 Ab
(clone EH12-
2132/2133)
PD-1Chronic SIVmac251-infected rhesus macaquesStage I: anti-PD-1 Ab (3 mg/kg/dose, 5 doses) between 24 and 30 weeks after infection on days 0, 3, 7, 10, and 14.
Stage II: RMs again treated with anti-PD-1 Ab (10 mg/kg/dose, three monthly, 3 doses) at 26–30 weeks following ART initiation
  • Improves SIV Gag-specific CD8+ T cell functions prior to ART.
  • Reduces the viral reservoir after ART initiation.
  • Enhances viral control after ART interruption.
Diego A Vargas- Inchaustegui [102]B7-DC-Ig
fusion protein
PD-1Chronic SIVmac251- infected rhesus macaquesART plus B7-DC-Ig (10 mg/kg, weekly, 11 weeks), then B7-DC-Ig alone for 12 weeks
  • Maintains lower viremia, favorable T cell/Treg repertoire, and lower SIV-specific responses.
Elena
Bekerman [110]
Human/rhesus chimeric anti-
PD-1 antibody
PD-1ART SIVmac251-infected rhesus macaquesAnti-PD-1 chimeric Ab (10 mg/kg, every other week, four doses) with or without TLR7 agonist vesatolimod (0.15 mg/kg, every other week, 10 doses)
  • No change in viral rebound kinetics following ART interruption.
  • No change in SIV reservoir size.
  • No change in frequency and function of SIV-specific T cells.
Sheikh Abdul Rahman [108]Primatized anti–PD-1 antibody (clone EH12)PD-1Chronical SIVmac239-infected rhesus macaquesImmunized with a CD40L plus TLR7 agonist–adjuvanted DNA/MVA SIV239 vaccine (DNA vaccine: 1 mg/333 µL, 600 µL/dose, at weeks 38 and 42 MVA vaccine: 1 mL/dose, at weeks 46 and 60) during ART. Received anti–PD-1 treatment on days 0, 3, 7, 10, and 14, starting 10 days before the initiation of ART (3 mg/kg) and on week 38–44 starting with the first DNA prime during ART (10 mg/kg, 3 doses, every 3 weeks)
  • Increases the frequency of cytolytic CD8+ T cells in the blood and LN.
  • Enhanced cytolytic CD8+ T cells localization in germinal centers of B cell follicles.
  • Reduces viral reservoirs in lymphoid tissue.
  • Controls viral rebound upon ART discontinuation.
ChunxiuWu [109]Anti-PD-1 antibody (GB226)PD-1ChronicallySIV-infected macaqueAnti-PD-1 antibody injection (20 mg/kg) every 2 weeks from 1 to 7 weeks and rAd5-SIVgpe (1011vp in 1 mL PBS) at weeks 0 and 4 post ART discontinuation; ART treatment begins at week 3 before the initial vaccination
  • Exacerbates viral rebound after ART interruption.
  • Accelerates the reactivation of the latent reservoir and AIDS progression.
  • Increased CD4 and CD8 T cell counts.
  • Improve SIV-specific CD4+ and CD8+ T cell immune function.
Enxiang Pan [106]GenolimzumabPD-1Chinese rhesus monkeysGenolimzumab injection (20 mg/kg, every two weeks) at weeks −1, 1, 3, 5, and 7 and rAd5-SIVgpe (1011vp) injection at week 0 and 4; at week 42 after the initial vaccination, animals were challenged with repeated low-dose SIVmac239
  • Augments and sustains vaccine-induced SIV-specific CD8+ T cell responses.
  • Confers better control of pathogenic SIVmac239 infection.
Ping Che [101]AvelumabPD-L1ART SIVmac239-infected rhesus macaquesAvelumab (20 mg/kg, weekly, for 24 weeks) and rhIL-15 (10 µg/kg, daily, continuous infusion for 10 days, two cycles), then, ART was discontinued and avelumab treatment continued until completion of the 24-week treatment
  • Transiently increases proliferation of natural killer and CD8+ T cells.
  • Expands CXCR3+PD-1−/low CD8+ T cell subset with ability to secrete cytokines.
  • No change in plasma viremia after ART interruption.
Amanda L Gill [100]AvelumabPD-L1ART SIVmac239-infected rhesus macaquesAvelumab (20 mg/kg, weekly)
At week 24, all treatments were discontinued
  • Leads to a trend of transient viral control after discontinuation of treatment.
Anna Hryniewicz [103]MDX-010CTLA-4ART SIVmac251-infected rhesus macaquesAdministered MDX-010 (10 mg/kg/injection) after ART initiation at weeks 5 and 8.
  • No impact on viral rebound following ART suspension.
  • No impact on CD4 T cell and CD8 T cell counts.
  • Decreases viral RNA level in LNs.
  • Increases effector function of SIV-specific CD4+ and CD8+ T cells.
Todd Bradley [105]IpilimumabCTLA-4 Cynomolgus
macaques
Immunized with recombinant CH505 HIV Env gp120 (100 µg every 4 weeks) and ipilimumab (10 mg/kg) during the immunization 1–3
  • Promotes germinal center activity.
  • Enhances HIV-1 Env antibody responses.
Justin Harper [104]Nivolumab
Ipilimumab
PD-1
CTLA-4
SIVmac239-infected rhesus macaquesWeekly nivolumab and ipilimumab over four weeks during ART, then, ART was interrupted two weeks afterwards with a seven-month follow-up
  • Enhances T cell proliferation and response in LNs and PBMCs.
  • Induces robust viral reactivation in plasma.
  • Decreases total and intact SIV-DNA in CD4+ T cells during ART in LNs.
  • No change in SIV-specific CD8+ T cell responses during ART.
  • No control of viremia after ART interruption.
In summary, the blockade of checkpoint targets is a promising strategy for controlling the disease progression of HIV or SIV infection functionally and clinically. More studies are on the way for translating this treatment into new patient regimens, hopefully soon.

5. Conclusions

Immune checkpoint markers serve as novel biomarkers for HIV infection. ICs control the switching of T cells between the states of activation and exhaustion, and are closely related to HIV pathogenesis. IC overexpression in CD4+ and CD8+ T cells attenuated the HIV-specific T cell response. Moreover, ICs are preferentially expressed on the surface of persistently HIV-infected T cells. CD4+ T cells overexpressing ICs are commonly found to serve as HIV reservoirs, and the ICs might play an essential role in HIV latency. Correspondingly, the blockade of ICs was shown to reverse HIV latency and restore the T cell function, providing promising therapeutic opportunities for HIV therapy both experimentally and clinically. However, the underlying regulatory mechanism of IC expression triggered by HIV infection and the cellular immune pathways mediated by IC blockers are still unknown and need further investigation. The optimum therapeutic design of ICs blockers alone or combined with other robust antiviral strategies under study offer novel strategies for the achievement of an HIV cure.

Author Contributions

Writing—original draft preparation, J.X. and Y.S.; writing—review and editing, J.X. and Y.S. All authors have read and agreed to the published version of the manuscript.

Funding

This work was funded by the CAMS Innovation Fund for Medical Sciences (grant number 2021-I2M-1-037), and the National Natural Science Foundation of China (grant number 81971944).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

We thank Yuhong Wang for her advice and help proofreading the article.

Conflicts of Interest

The authors declare no competing interest.

References

  1. Wherry, E.J. T cell exhaustion. Nat. Immunol. 2011, 12, 492–499. [Google Scholar] [CrossRef] [PubMed]
  2. Fenwick, C.; Joo, V.; Jacquier, P.; Noto, A.; Banga, R.; Perreau, M.; Pantaleo, G. T-cell exhaustion in HIV infection. Immunol. Rev. 2019, 292, 149–163. [Google Scholar] [CrossRef] [PubMed]
  3. Kraehenbuehl, L.; Weng, C.H.; Eghbali, S.; Wolchok, J.D.; Merghoub, T. Enhancing immunotherapy in cancer by targeting emerging immunomodulatory pathways. Nat. Rev. Clin. Oncol. 2022, 19, 37–50. [Google Scholar] [CrossRef] [PubMed]
  4. Qin, S.; Xu, L.; Yi, M.; Yu, S.; Wu, K.; Luo, S. Novel immune checkpoint targets: Moving beyond PD-1 and CTLA-4. Mol. Cancer 2019, 18, 155. [Google Scholar] [CrossRef] [PubMed]
  5. Dyck, L.; Mills, K.H.G. Immune checkpoints and their inhibition in cancer and infectious diseases. Eur. J. Immunol. 2017, 47, 765–779. [Google Scholar] [CrossRef] [PubMed]
  6. Kalbasi, A.; Ribas, A. Tumour-intrinsic resistance to immune checkpoint blockade. Nat. Rev. Immunol. 2020, 20, 25–39. [Google Scholar] [CrossRef] [PubMed]
  7. Wykes, M.N.; Lewin, S.R. Immune checkpoint blockade in infectious diseases. Nat. Rev. Immunol. 2018, 18, 91–104. [Google Scholar] [CrossRef]
  8. Chen, H.; Moussa, M.; Catalfamo, M. The Role of Immunomodulatory Receptors in the Pathogenesis of HIV Infection: A Therapeutic Opportunity for HIV Cure? Front. Immunol. 2020, 11, 1223. [Google Scholar] [CrossRef]
  9. Zhang, J.Y.; Zhang, Z.; Wang, X.; Fu, J.L.; Yao, J.; Jiao, Y.; Chen, L.; Zhang, H.; Wei, J.; Jin, L.; et al. PD-1 up-regulation is correlated with HIV-specific memory CD8+ T-cell exhaustion in typical progressors but not in long-term nonprogressors. Blood 2007, 109, 4671–4678. [Google Scholar] [CrossRef] [PubMed]
  10. Shahbaz, S.; Dunsmore, G.; Koleva, P.; Xu, L.; Houston, S.; Elahi, S. Galectin-9 and VISTA Expression Define Terminally Exhausted T Cells in HIV-1 Infection. J. Immunol. 2020, 204, 2474–2491. [Google Scholar] [CrossRef]
  11. Noyan, K.; Nguyen, S.; Betts, M.R.; Sonnerborg, A.; Buggert, M. Human Immunodeficiency Virus Type-1 Elite Controllers Maintain Low Co-Expression of Inhibitory Receptors on CD4+ T Cells. Front. Immunol. 2018, 9, 19. [Google Scholar] [CrossRef] [Green Version]
  12. Nakayama, K.; Nakamura, H.; Koga, M.; Koibuchi, T.; Fujii, T.; Miura, T.; Iwamoto, A.; Kawana-Tachikawa, A. Imbalanced production of cytokines by T cells associates with the activation/exhaustion status of memory T cells in chronic HIV type 1 infection. AIDS Res. Hum. Retrovir. 2012, 28, 702–714. [Google Scholar] [CrossRef] [PubMed]
  13. Chen, H.; Ren, C.; Song, H.; Ma, L.L.; Chen, S.F.; Wu, M.J.; Zhang, H.; Xu, J.C.; Xu, P. Temporal and spatial characterization of negative regulatory T cells in HIV-infected/AIDS patients raises new diagnostic markers and therapeutic strategies. J. Clin. Lab. Anal. 2021, 35, e23831. [Google Scholar] [CrossRef] [PubMed]
  14. Hurst, J.; Hoffmann, M.; Pace, M.; Williams, J.P.; Thornhill, J.; Hamlyn, E.; Meyerowitz, J.; Willberg, C.; Koelsch, K.K.; Robinson, N.; et al. Immunological biomarkers predict HIV-1 viral rebound after treatment interruption. Nat. Commun. 2015, 6, 8495. [Google Scholar] [CrossRef]
  15. Macatangay, B.J.C.; Gandhi, R.T.; Jones, R.B.; McMahon, D.K.; Lalama, C.M.; Bosch, R.J.; Cyktor, J.C.; Thomas, A.S.; Borowski, L.; Riddler, S.A.; et al. T cells with high PD-1 expression are associated with lower HIV-specific immune responses despite long-term antiretroviral therapy. AIDS 2020, 34, 15–24. [Google Scholar] [CrossRef] [PubMed]
  16. Whittall, T.; Peters, B.; Rahman, D.; Kingsley, C.I.; Vaughan, R.; Lehner, T. Immunogenic and tolerogenic signatures in human immunodeficiency virus (HIV)-infected controllers compared with progressors and a conversion strategy of virus control. Clin. Exp. Immunol. 2011, 166, 208–217. [Google Scholar] [CrossRef] [PubMed]
  17. Fromentin, R.; Bakeman, W.; Lawani, M.B.; Khoury, G.; Hartogensis, W.; DaFonseca, S.; Killian, M.; Epling, L.; Hoh, R.; Sinclair, E.; et al. CD4+ T Cells Expressing PD-1, TIGIT and LAG-3 Contribute to HIV Persistence during ART. PLoS Pathog. 2016, 12, e1005761. [Google Scholar] [CrossRef]
  18. Khoury, G.; Fromentin, R.; Solomon, A.; Hartogensis, W.; Killian, M.; Hoh, R.; Somsouk, M.; Hunt, P.W.; Girling, V.; Sinclair, E.; et al. Human Immunodeficiency Virus Persistence and T-Cell Activation in Blood, Rectal, and Lymph Node Tissue in Human Immunodeficiency Virus-Infected Individuals Receiving Suppressive Antiretroviral Therapy. J. Infect. Dis. 2017, 215, 911–919. [Google Scholar] [CrossRef]
  19. Petrovas, C.; Casazza, J.P.; Brenchley, J.M.; Price, D.A.; Gostick, E.; Adams, W.C.; Precopio, M.L.; Schacker, T.; Roederer, M.; Douek, D.C.; et al. PD-1 is a regulator of virus-specific CD8+ T cell survival in HIV infection. J. Exp. Med. 2006, 203, 2281–2292. [Google Scholar] [CrossRef]
  20. Hoffmann, M.; Pantazis, N.; Martin, G.E.; Hickling, S.; Hurst, J.; Meyerowitz, J.; Willberg, C.B.; Robinson, N.; Brown, H.; Fisher, M.; et al. Exhaustion of Activated CD8 T Cells Predicts Disease Progression in Primary HIV-1 Infection. PLoS Pathog. 2016, 12, e1005661. [Google Scholar] [CrossRef] [Green Version]
  21. Jensen, S.S.; Fomsgaard, A.; Larsen, T.K.; Tingstedt, J.L.; Gerstoft, J.; Kronborg, G.; Pedersen, C.; Karlsson, I. Initiation of Antiretroviral Therapy (ART) at Different Stages of HIV-1 Disease Is Not Associated with the Proportion of Exhausted CD8+ T Cells. PLoS ONE 2015, 10, e0139573. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Tailor, J.; Foldi, J.; Generoso, M.; McCarty, B.; Alankar, A.; Kilberg, M.; Mwamzuka, M.; Marshed, F.; Ahmed, A.; Liu, M.; et al. Disease Progression in Children with Perinatal HIV Correlates with Increased PD-1+ CD8 T Cells that Coexpress Multiple Immune Checkpoints. J. Infect. Dis. 2021, 224, 1785–1795. [Google Scholar]
  23. Li, J.; Huang, H.H.; Tu, B.; Zhou, M.J.; Hu, W.; Fu, Y.L.; Li, X.Y.; Yang, T.; Song, J.W.; Fan, X.; et al. Reversal of the CD8(+) T-Cell Exhaustion Induced by Chronic HIV-1 Infection Through Combined Blockade of the Adenosine and PD-1 Pathways. Front. Immunol. 2021, 12, 687296. [Google Scholar] [CrossRef] [PubMed]
  24. Zhang, Z.; Xu, X.; Lu, J.; Zhang, S.; Gu, L.; Fu, J.; Jin, L.; Li, H.; Zhao, M.; Zhang, J.; et al. B and T lymphocyte attenuator down-regulation by HIV-1 depends on type I interferon and contributes to T-cell hyperactivation. J. Infect. Dis. 2011, 203, 1668–1678. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Muthumani, K.; Choo, A.Y.; Shedlock, D.J.; Laddy, D.J.; Sundaram, S.G.; Hirao, L.; Wu, L.; Thieu, K.P.; Chung, C.W.; Lankaraman, K.M.; et al. Human immunodeficiency virus type 1 Nef induces programmed death 1 expression through a p38 mitogen-activated protein kinase-dependent mechanism. J. Virol. 2008, 82, 11536–11544. [Google Scholar] [CrossRef] [Green Version]
  26. El-Far, M.; Ancuta, P.; Routy, J.P.; Zhang, Y.; Bakeman, W.; Bordi, R.; DaFonseca, S.; Said, E.A.; Gosselin, A.; Tep, T.S.; et al. Nef promotes evasion of human immunodeficiency virus type 1-infected cells from the CTLA-4-mediated inhibition of T-cell activation. J. Gen. Virol. 2015, 96, 1463–1477. [Google Scholar] [CrossRef] [Green Version]
  27. Prevost, J.; Edgar, C.R.; Richard, J.; Trothen, S.M.; Jacob, R.A.; Mumby, M.J.; Pickering, S.; Dube, M.; Kaufmann, D.E.; Kirchhoff, F.; et al. HIV-1 Vpu Downregulates Tim-3 from the Surface of Infected CD4(+) T Cells. J. Virol. 2020, 94, e01999-19. [Google Scholar] [CrossRef]
  28. Jacob, R.A.; Edgar, C.R.; Prevost, J.; Trothen, S.M.; Lurie, A.; Mumby, M.J.; Galbraith, A.; Kirchhoff, F.; Haeryfar, S.M.M.; Finzi, A.; et al. The HIV-1 accessory protein Nef increases surface expression of the checkpoint receptor Tim-3 in infected CD4(+) T cells. J. Biol. Chem. 2021, 297, 101042. [Google Scholar] [CrossRef]
  29. Hoang, T.N.; Harper, J.L.; Pino, M.; Wang, H.; Micci, L.; King, C.T.; McGary, C.S.; McBrien, J.B.; Cervasi, B.; Silvestri, G.; et al. Bone Marrow-Derived CD4(+) T Cells Are Depleted in Simian Immunodeficiency Virus-Infected Macaques and Contribute to the Size of the Replication-Competent Reservoir. J. Virol. 2019, 93, e01344-18. [Google Scholar] [CrossRef] [Green Version]
  30. Yamamoto, T.; Price, D.A.; Casazza, J.P.; Ferrari, G.; Nason, M.; Chattopadhyay, P.K.; Roederer, M.; Gostick, E.; Katsikis, P.D.; Douek, D.C.; et al. Surface expression patterns of negative regulatory molecules identify determinants of virus-specific CD8+ T-cell exhaustion in HIV infection. Blood 2011, 117, 4805–4815. [Google Scholar] [CrossRef] [Green Version]
  31. Teigler, J.E.; Zelinskyy, G.; Eller, M.A.; Bolton, D.L.; Marovich, M.; Gordon, A.D.; Alrubayyi, A.; Alter, G.; Robb, M.L.; Martin, J.N.; et al. Differential Inhibitory Receptor Expression on T Cells Delineates Functional Capacities in Chronic Viral Infection. J. Virol. 2017, 91, e01263-17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Chew, G.M.; Fujita, T.; Webb, G.M.; Burwitz, B.J.; Wu, H.L.; Reed, J.S.; Hammond, K.B.; Clayton, K.L.; Ishii, N.; Abdel-Mohsen, M.; et al. TIGIT Marks Exhausted T Cells, Correlates with Disease Progression, and Serves as a Target for Immune Restoration in HIV and SIV Infection. PLoS Pathog. 2016, 12, e1005349. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Tauriainen, J.; Scharf, L.; Frederiksen, J.; Naji, A.; Ljunggren, H.G.; Sonnerborg, A.; Lund, O.; Reyes-Teran, G.; Hecht, F.M.; Deeks, S.G.; et al. Perturbed CD8(+) T cell TIGIT/CD226/PVR axis despite early initiation of antiretroviral treatment in HIV infected individuals. Sci. Rep. 2017, 7, 40354. [Google Scholar] [CrossRef] [Green Version]
  34. Leng, Q.; Bentwich, Z.; Magen, E.; Kalinkovich, A.; Borkow, G. CTLA-4 upregulation during HIV infection: Association with anergy and possible target for therapeutic intervention. AIDS 2002, 16, 519–529. [Google Scholar] [CrossRef]
  35. Steiner, K.; Waase, I.; Rau, T.; Dietrich, M.; Fleischer, B.; Broker, B.M. Enhanced expression of CTLA-4 (CD152) on CD4+ T cells in HIV infection. Clin. Exp. Immunol. 1999, 115, 451–457. [Google Scholar] [CrossRef]
  36. Tian, X.; Zhang, A.; Qiu, C.; Wang, W.; Yang, Y.; Qiu, C.; Liu, A.; Zhu, L.; Yuan, S.; Hu, H.; et al. The upregulation of LAG-3 on T cells defines a subpopulation with functional exhaustion and correlates with disease progression in HIV-infected subjects. J. Immunol. 2015, 194, 3873–3882. [Google Scholar] [CrossRef] [Green Version]
  37. Jones, R.B.; Ndhlovu, L.C.; Barbour, J.D.; Sheth, P.M.; Jha, A.R.; Long, B.R.; Wong, J.C.; Satkunarajah, M.; Schweneker, M.; Chapman, J.M.; et al. Tim-3 expression defines a novel population of dysfunctional T cells with highly elevated frequencies in progressive HIV-1 infection. J. Exp. Med. 2008, 205, 2763–2779. [Google Scholar] [CrossRef]
  38. Fujita, T.; Burwitz, B.J.; Chew, G.M.; Reed, J.S.; Pathak, R.; Seger, E.; Clayton, K.L.; Rini, J.M.; Ostrowski, M.A.; Ishii, N.; et al. Expansion of dysfunctional Tim-3-expressing effector memory CD8+ T cells during simian immunodeficiency virus infection in rhesus macaques. J. Immunol. 2014, 193, 5576–5583. [Google Scholar] [CrossRef] [Green Version]
  39. Zhang, L.; Zhang, A.; Xu, J.; Qiu, C.; Zhu, L.; Qiu, C.; Fu, W.; Wang, Y.; Ye, L.; Fu, Y.X.; et al. CD160 Plays a Protective Role During Chronic Infection by Enhancing Both Functionalities and Proliferative Capacity of CD8+ T Cells. Front. Immunol. 2020, 11, 2188. [Google Scholar] [CrossRef]
  40. Day, C.L.; Kaufmann, D.E.; Kiepiela, P.; Brown, J.A.; Moodley, E.S.; Reddy, S.; Mackey, E.W.; Miller, J.D.; Leslie, A.J.; DePierres, C.; et al. PD-1 expression on HIV-specific T cells is associated with T-cell exhaustion and disease progression. Nature 2006, 443, 350–354. [Google Scholar] [CrossRef]
  41. Cockerham, L.R.; Jain, V.; Sinclair, E.; Glidden, D.V.; Hartogenesis, W.; Hatano, H.; Hunt, P.W.; Martin, J.N.; Pilcher, C.D.; Sekaly, R.; et al. Programmed death-1 expression on CD4(+) and CD8(+) T cells in treated and untreated HIV disease. AIDS 2014, 28, 1749–1758. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Rallon, N.; Garcia, M.; Garcia-Samaniego, J.; Cabello, A.; Alvarez, B.; Restrepo, C.; Nistal, S.; Gorgolas, M.; Benito, J.M. Expression of PD-1 and Tim-3 markers of T-cell exhaustion is associated with CD4 dynamics during the course of untreated and treated HIV infection. PLoS ONE 2018, 13, e0193829. [Google Scholar] [CrossRef] [PubMed]
  43. Foldi, J.; Kozhaya, L.; McCarty, B.; Mwamzuka, M.; Marshed, F.; Ilmet, T.; Kilberg, M.; Kravietz, A.; Ahmed, A.; Borkowsky, W.; et al. HIV-Infected Children Have Elevated Levels of PD-1+ Memory CD4 T Cells With Low Proliferative Capacity and High Inflammatory Cytokine Effector Functions. J. Infect. Dis. 2017, 216, 641–650. [Google Scholar] [CrossRef] [PubMed]
  44. Trautmann, L.; Janbazian, L.; Chomont, N.; Said, E.A.; Gimmig, S.; Bessette, B.; Boulassel, M.R.; Delwart, E.; Sepulveda, H.; Balderas, R.S.; et al. Upregulation of PD-1 expression on HIV-specific CD8+ T cells leads to reversible immune dysfunction. Nat. Med. 2006, 12, 1198–1202. [Google Scholar] [CrossRef] [PubMed]
  45. Kassu, A.; Marcus, R.A.; D’Souza, M.B.; Kelly-McKnight, E.A.; Golden-Mason, L.; Akkina, R.; Fontenot, A.P.; Wilson, C.C.; Palmer, B.E. Regulation of virus-specific CD4+ T cell function by multiple costimulatory receptors during chronic HIV infection. J. Immunol. 2010, 185, 3007–3018. [Google Scholar] [CrossRef] [Green Version]
  46. Grabmeier-Pfistershammer, K.; Stecher, C.; Zettl, M.; Rosskopf, S.; Rieger, A.; Zlabinger, G.J.; Steinberger, P. Antibodies targeting BTLA or TIM-3 enhance HIV-1 specific T cell responses in combination with PD-1 blockade. Clin. Immunol. 2017, 183, 167–173. [Google Scholar] [CrossRef]
  47. Chew, G.M.; Padua, A.J.P.; Chow, D.C.; Souza, S.A.; Clements, D.M.; Corley, M.J.; Pang, A.P.S.; Alejandria, M.M.; Gerschenson, M.; Shikuma, C.M.; et al. Effects of Brief Adjunctive Metformin Therapy in Virologically Suppressed HIV-Infected Adults on Polyfunctional HIV-Specific CD8 T Cell Responses to PD-L1 Blockade. AIDS Res. Hum. Retrovir. 2021, 37, 24–33. [Google Scholar] [CrossRef]
  48. Petrovas, C.; Price, D.A.; Mattapallil, J.; Ambrozak, D.R.; Geldmacher, C.; Cecchinato, V.; Vaccari, M.; Tryniszewska, E.; Gostick, E.; Roederer, M.; et al. SIV-specific CD8+ T cells express high levels of PD1 and cytokines but have impaired proliferative capacity in acute and chronic SIVmac251 infection. Blood 2007, 110, 928–936. [Google Scholar] [CrossRef]
  49. Onlamoon, N.; Rogers, K.; Mayne, A.E.; Pattanapanyasat, K.; Mori, K.; Villinger, F.; Ansari, A.A. Soluble PD-1 rescues the proliferative response of simian immunodeficiency virus-specific CD4 and CD8 T cells during chronic infection. Immunology 2008, 124, 277–293. [Google Scholar] [CrossRef]
  50. Kaufmann, D.E.; Kavanagh, D.G.; Pereyra, F.; Zaunders, J.J.; Mackey, E.W.; Miura, T.; Palmer, S.; Brockman, M.; Rathod, A.; Piechocka-Trocha, A.; et al. Upregulation of CTLA-4 by HIV-specific CD4+ T cells correlates with disease progression and defines a reversible immune dysfunction. Nat. Immunol. 2007, 8, 1246–1254. [Google Scholar] [CrossRef]
  51. Blazkova, J.; Huiting, E.D.; Boddapati, A.K.; Shi, V.; Whitehead, E.J.; Justement, J.S.; Nordstrom, J.L.; Moir, S.; Lack, J.; Chun, T.W. Correlation Between TIGIT Expression on CD8+ T Cells and Higher Cytotoxic Capacity. J. Infect. Dis. 2021, 224, 1599–1604. [Google Scholar] [CrossRef] [PubMed]
  52. Zhang, Z.N.; Zhu, M.L.; Chen, Y.H.; Fu, Y.J.; Zhang, T.W.; Jiang, Y.J.; Chu, Z.X.; Shang, H. Elevation of Tim-3 and PD-1 expression on T cells appears early in HIV infection, and differential Tim-3 and PD-1 expression patterns can be induced by common gamma -chain cytokines. Biomed. Res. Int. 2015, 2015, 916936. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Tendeiro, R.; Foxall, R.B.; Baptista, A.P.; Pinto, F.; Soares, R.S.; Cavaleiro, R.; Valadas, E.; Gomes, P.; Victorino, R.M.; Sousa, A.E. PD-1 and its ligand PD-L1 are progressively up-regulated on CD4 and CD8 T-cells in HIV-2 infection irrespective of the presence of viremia. AIDS 2012, 26, 1065–1071. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Sakhdari, A.; Mujib, S.; Vali, B.; Yue, F.Y.; MacParland, S.; Clayton, K.; Jones, R.B.; Liu, J.; Lee, E.Y.; Benko, E.; et al. Tim-3 negatively regulates cytotoxicity in exhausted CD8+ T cells in HIV infection. PLoS ONE 2012, 7, e40146. [Google Scholar] [CrossRef]
  55. Amancha, P.K.; Hong, J.J.; Ansari, A.A.; Villinger, F. Up-regulation of Tim-3 on T cells during acute simian immunodeficiency virus infection and on antigen specific responders. AIDS 2015, 29, 531–536. [Google Scholar] [CrossRef] [Green Version]
  56. Peretz, Y.; He, Z.; Shi, Y.; Yassine-Diab, B.; Goulet, J.P.; Bordi, R.; Filali-Mouhim, A.; Loubert, J.B.; El-Far, M.; Dupuy, F.P.; et al. CD160 and PD-1 co-expression on HIV-specific CD8 T cells defines a subset with advanced dysfunction. PLoS Pathog. 2012, 8, e1002840. [Google Scholar] [CrossRef]
  57. Pombo, C.; Wherry, E.J.; Gostick, E.; Price, D.A.; Betts, M.R. Elevated Expression of CD160 and 2B4 Defines a Cytolytic HIV-Specific CD8+ T-Cell Population in Elite Controllers. J. Infect. Dis. 2015, 212, 1376–1386. [Google Scholar] [CrossRef] [Green Version]
  58. Martin, G.E.; Pace, M.; Shearer, F.M.; Zilber, E.; Hurst, J.; Meyerowitz, J.; Thornhill, J.P.; Lwanga, J.; Brown, H.; Robinson, N.; et al. Levels of Human Immunodeficiency Virus DNA Are Determined Before ART Initiation and Linked to CD8 T-Cell Activation and Memory Expansion. J. Infect. Dis. 2020, 221, 1135–1145. [Google Scholar] [CrossRef] [Green Version]
  59. Pardons, M.; Baxter, A.E.; Massanella, M.; Pagliuzza, A.; Fromentin, R.; Dufour, C.; Leyre, L.; Routy, J.P.; Kaufmann, D.E.; Chomont, N. Single-cell characterization and quantification of translation-competent viral reservoirs in treated and untreated HIV infection. PLoS Pathog. 2019, 15, e1007619. [Google Scholar] [CrossRef]
  60. Rinaldi, S.; de Armas, L.; Dominguez-Rodriguez, S.; Pallikkuth, S.; Dinh, V.; Pan, L.; Grtner, K.; Pahwa, R.; Cotugno, N.; Rojo, P.; et al. T cell immune discriminants of HIV reservoir size in a pediatric cohort of perinatally infected individuals. PLoS Pathog. 2021, 17, e1009533. [Google Scholar] [CrossRef]
  61. Banga, R.; Procopio, F.A.; Noto, A.; Pollakis, G.; Cavassini, M.; Ohmiti, K.; Corpataux, J.M.; de Leval, L.; Pantaleo, G.; Perreau, M. PD-1(+) and follicular helper T cells are responsible for persistent HIV-1 transcription in treated aviremic individuals. Nat. Med. 2016, 22, 754–761. [Google Scholar] [CrossRef] [PubMed]
  62. Pantaleo, G.; Levy, Y. Therapeutic vaccines and immunological intervention in HIV infection: A paradigm change. Curr. Opin. HIV AIDS 2016, 11, 576–584. [Google Scholar] [CrossRef] [PubMed]
  63. Boyer, Z.; Palmer, S. Targeting Immune Checkpoint Molecules to Eliminate Latent HIV. Front. Immunol. 2018, 9, 2339. [Google Scholar] [CrossRef] [PubMed]
  64. McGary, C.S.; Deleage, C.; Harper, J.; Micci, L.; Ribeiro, S.P.; Paganini, S.; Kuri-Cervantes, L.; Benne, C.; Ryan, E.S.; Balderas, R.; et al. CTLA-4(+)PD-1(-) Memory CD4(+) T Cells Critically Contribute to Viral Persistence in Antiretroviral Therapy-Suppressed, SIV-Infected Rhesus Macaques. Immunity 2017, 47, 776–788.e5. [Google Scholar] [CrossRef] [Green Version]
  65. Xue, J.; Chong, H.; Zhu, Y.; Zhang, J.; Tong, L.; Lu, J.; Chen, T.; Cong, Z.; Wei, Q.; He, Y. Efficient treatment and pre-exposure prophylaxis in rhesus macaques by an HIV fusion-inhibitory lipopeptide. Cell 2022, 185, 131–144.e18. [Google Scholar] [CrossRef]
  66. Llewellyn, G.N.; Seclen, E.; Wietgrefe, S.; Liu, S.; Chateau, M.; Pei, H.; Perkey, K.; Marsden, M.D.; Hinkley, S.J.; Paschon, D.E.; et al. Humanized Mouse Model of HIV-1 Latency with Enrichment of Latent Virus in PD-1(+) and TIGIT(+) CD4 T Cells. J. Virol. 2019, 93, e02086-18. [Google Scholar] [CrossRef] [Green Version]
  67. Evans, V.A.; van der Sluis, R.M.; Solomon, A.; Dantanarayana, A.; McNeil, C.; Garsia, R.; Palmer, S.; Fromentin, R.; Chomont, N.; Sekaly, R.P.; et al. Programmed cell death-1 contributes to the establishment and maintenance of HIV-1 latency. AIDS 2018, 32, 1491–1497. [Google Scholar] [CrossRef]
  68. Van der Sluis, R.M.; Kumar, N.A.; Pascoe, R.D.; Zerbato, J.M.; Evans, V.A.; Dantanarayana, A.I.; Anderson, J.L.; Sekaly, R.P.; Fromentin, R.; Chomont, N.; et al. Combination Immune Checkpoint Blockade to Reverse HIV Latency. J. Immunol. 2020, 204, 1242–1254. [Google Scholar] [CrossRef]
  69. Fromentin, R.; DaFonseca, S.; Costiniuk, C.T.; El-Far, M.; Procopio, F.A.; Hecht, F.M.; Hoh, R.; Deeks, S.G.; Hazuda, D.J.; Lewin, S.R.; et al. PD-1 blockade potentiates HIV latency reversal ex vivo in CD4(+) T cells from ART-suppressed individuals. Nat. Commun. 2019, 10, 814. [Google Scholar] [CrossRef] [Green Version]
  70. Bui, J.K.; Cyktor, J.C.; Fyne, E.; Campellone, S.; Mason, S.W.; Mellors, J.W. Blockade of the PD-1 axis alone is not sufficient to activate HIV-1 virion production from CD4+ T cells of individuals on suppressive ART. PLoS ONE 2019, 14, e0211112. [Google Scholar] [CrossRef]
  71. Cook, M.R.; Kim, C. Safety and Efficacy of Immune Checkpoint Inhibitor Therapy in Patients With HIV Infection and Advanced-Stage Cancer: A Systematic Review. JAMA Oncol. 2019, 5, 1049–1054. [Google Scholar] [CrossRef] [PubMed]
  72. Tapia Rico, G.; Chan, M.M.; Loo, K.F. The safety and efficacy of immune checkpoint inhibitors in patients with advanced cancers and pre-existing chronic viral infections (Hepatitis B/C, HIV): A review of the available evidence. Cancer Treat. Rev. 2020, 86, 102011. [Google Scholar] [CrossRef]
  73. Abbar, B.; Baron, M.; Katlama, C.; Marcelin, A.G.; Veyri, M.; Autran, B.; Guihot, A.; Spano, J.P. Immune checkpoint inhibitors in people living with HIV: What about anti-HIV effects? AIDS 2020, 34, 167–175. [Google Scholar] [CrossRef] [PubMed]
  74. Guaitoli, G.; Baldessari, C.; Maur, M.; Mussini, C.; Meschiari, M.; Barbieri, F.; Cascinu, S.; Bertolini, F. Treating cancer with immunotherapy in HIV-positive patients: A challenging reality. Crit. Rev. Oncol. Hematol. 2020, 145, 102836. [Google Scholar] [CrossRef] [PubMed]
  75. Zerbato, J.M.; Purves, H.V.; Lewin, S.R.; Rasmussen, T.A. Between a shock and a hard place: Challenges and developments in HIV latency reversal. Curr. Opin. Virol. 2019, 38, 1–9. [Google Scholar] [CrossRef] [PubMed]
  76. Pham, H.T.; Yoo, S.; Mesplede, T. Combination therapies currently under investigation in phase I and phase II clinical trials for HIV-1. Expert Opin. Investig. Drugs 2020, 29, 273–283. [Google Scholar] [CrossRef] [PubMed]
  77. Wightman, F.; Solomon, A.; Kumar, S.S.; Urriola, N.; Gallagher, K.; Hiener, B.; Palmer, S.; McNeil, C.; Garsia, R.; Lewin, S.R. Effect of ipilimumab on the HIV reservoir in an HIV-infected individual with metastatic melanoma. AIDS 2015, 29, 504–506. [Google Scholar] [CrossRef] [Green Version]
  78. Blanch-Lombarte, O.; Galvez, C.; Revollo, B.; Jimenez-Moyano, E.; Llibre, J.M.; Manzano, J.L.; Boada, A.; Dalmau, J.; Daniel, E.S.; Clotet, B.; et al. Enhancement of Antiviral CD8(+) T-Cell Responses and Complete Remission of Metastatic Melanoma in an HIV-1-Infected Subject Treated with Pembrolizumab. J. Clin. Med. 2019, 8, 2089. [Google Scholar] [CrossRef] [Green Version]
  79. Lau, J.S.Y.; McMahon, J.H.; Gubser, C.; Solomon, A.; Chiu, C.Y.H.; Dantanarayana, A.; Chea, S.; Tennakoon, S.; Zerbato, J.M.; Garlick, J.; et al. The impact of immune checkpoint therapy on the latent reservoir in HIV-infected individuals with cancer on antiretroviral therapy. AIDS 2021, 35, 1631–1636. [Google Scholar] [CrossRef]
  80. Guihot, A.; Marcelin, A.G.; Massiani, M.A.; Samri, A.; Soulie, C.; Autran, B.; Spano, J.P. Drastic decrease of the HIV reservoir in a patient treated with nivolumab for lung cancer. Ann. Oncol. 2018, 29, 517–518. [Google Scholar] [CrossRef] [Green Version]
  81. Hentrich, M.; Schipek-Voigt, K.; Jager, H.; Schulz, S.; Schmid, P.; Stotzer, O.; Bojko, P. Nivolumab in HIV-related non-small-cell lung cancer. Ann. Oncol. 2017, 28, 2890. [Google Scholar] [CrossRef] [PubMed]
  82. McCullar, B.; Alloway, T.; Martin, M. Durable complete response to nivolumab in a patient with HIV and metastatic non-small cell lung cancer. J. Thorac. Dis. 2017, 9, E540–E542. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. McLean, F.E.; Gathogo, E.; Goodall, D.; Jones, R.; Kinloch, S.; Post, F.A. Alemtuzumab induction therapy in HIV-positive renal transplant recipients. AIDS 2017, 31, 1047–1048. [Google Scholar] [CrossRef] [PubMed]
  84. Scully, E.P.; Rutishauser, R.L.; Simoneau, C.R.; Delagreverie, H.; Euler, Z.; Thanh, C.; Li, J.Z.; Hartig, H.; Bakkour, S.; Busch, M.; et al. Inconsistent HIV reservoir dynamics and immune responses following anti-PD-1 therapy in cancer patients with HIV infection. Ann. Oncol. 2018, 29, 2141–2142. [Google Scholar] [CrossRef] [PubMed]
  85. Shah, N.J.; Al-Shbool, G.; Blackburn, M.; Cook, M.; Belouali, A.; Liu, S.V.; Madhavan, S.; He, A.R.; Atkins, M.B.; Gibney, G.T.; et al. Safety and efficacy of immune checkpoint inhibitors (ICIs) in cancer patients with HIV, hepatitis B, or hepatitis C viral infection. J. Immunother. Cancer 2019, 7, 353. [Google Scholar] [CrossRef]
  86. Rasmussen, T.A.; Rajdev, L.; Rhodes, A.; Dantanarayana, A.; Tennakoon, S.; Chea, S.; Spelman, T.; Lensing, S.; Rutishauser, R.; Bakkour, S.; et al. Impact of anti-PD-1 and anti-CTLA-4 on the HIV reservoir in people living with HIV with cancer on antiretroviral therapy: The AIDS Malignancy Consortium-095 study. Clin. Infect. Dis. 2021, 73, e1973–e1981. [Google Scholar] [CrossRef] [PubMed]
  87. Chughlay, M.F.; Njuguna, C.; Cohen, K.; Maartens, G. Acute interstitial nephritis caused by lopinavir/ritonavir in a surgeon receiving antiretroviral postexposure prophylaxis. AIDS 2015, 29, 503–504. [Google Scholar] [CrossRef] [PubMed]
  88. Colston, E.; Grasela, D.; Gardiner, D.; Bucy, R.P.; Vakkalagadda, B.; Korman, A.J.; Lowy, I. An open-label, multiple ascending dose study of the anti-CTLA-4 antibody ipilimumab in viremic HIV patients. PLoS ONE 2018, 13, e0198158. [Google Scholar] [CrossRef] [Green Version]
  89. Gay, C.L.; Bosch, R.J.; Ritz, J.; Hataye, J.M.; Aga, E.; Tressler, R.L.; Mason, S.W.; Hwang, C.K.; Grasela, D.M.; Ray, N.; et al. Clinical Trial of the Anti-PD-L1 Antibody BMS-936559 in HIV-1 Infected Participants on Suppressive Antiretroviral Therapy. J. Infect. Dis. 2017, 215, 1725–1733. [Google Scholar] [CrossRef] [Green Version]
  90. Anti-PD-1 Therapy OK for Most with HIV. Cancer Discov. 2018, 8, 130–131.
  91. Gonzalez-Cao, M.; Moran, T.; Dalmau, J.; Garcia-Corbacho, J.; Bracht, J.W.P.; Bernabe, R.; Juan, O.; de Castro, J.; Blanco, R.; Drozdowskyj, A.; et al. Assessment of the Feasibility and Safety of Durvalumab for Treatment of Solid Tumors in Patients With HIV-1 Infection: The Phase 2 DURVAST Study. JAMA Oncol. 2020, 6, 1063–1067. [Google Scholar] [CrossRef] [PubMed]
  92. Spano, J.P.; Veyri, M.; Gobert, A.; Guihot, A.; Perre, P.; Kerjouan, M.; Brosseau, S.; Cloarec, N.; Montaudie, H.; Helissey, C.; et al. Immunotherapy for cancer in people living with HIV: Safety with an efficacy signal from the series in real life experience. AIDS 2019, 33, F13–F19. [Google Scholar] [CrossRef] [PubMed]
  93. Adashek, J.J.; Junior, P.N.A.; Galanina, N.; Kurzrock, R. Remembering the forgotten child: The role of immune checkpoint inhibition in patients with human immunod eficiency virus and cancer. J. Immunother. Cancer 2019, 7, 130. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Lurain, K.; Ramaswami, R.; Mangusan, R.; Widell, A.; Ekwede, I.; George, J.; Ambinder, R.; Cheever, M.; Gulley, J.L.; Goncalves, P.H.; et al. Use of pembrolizumab with or without pomalidomide in HIV-associated non-Hodgkin’s lymphoma. J. Immunother. Cancer 2021, 9, e002097. [Google Scholar] [CrossRef] [PubMed]
  95. Gay, C.L.; Bosch, R.J.; McKhann, A.; Moseley, K.F.; Wimbish, C.L.; Hendrickx, S.M.; Messer, M.; Furlong, M.; Campbell, D.M.; Jennings, C.; et al. Suspected Immune-Related Adverse Events With an Anti-PD-1 Inhibitor in Otherwise Healthy People with HIV. J. Acquir. Immune. Defic. Syndr. 2021, 87, e234–e236. [Google Scholar] [CrossRef] [PubMed]
  96. Velu, V.; Titanji, K.; Zhu, B.; Husain, S.; Pladevega, A.; Lai, L.; Vanderford, T.H.; Chennareddi, L.; Silvestri, G.; Freeman, G.J.; et al. Enhancing SIV-specific immunity in vivo by PD-1 blockade. Nature 2009, 458, 206–210. [Google Scholar] [CrossRef]
  97. Dyavar Shetty, R.; Velu, V.; Titanji, K.; Bosinger, S.E.; Freeman, G.J.; Silvestri, G.; Amara, R.R. PD-1 blockade during chronic SIV infection reduces hyperimmune activation and microbial translocation in rhesus macaques. J. Clin. Investig. 2012, 122, 1712–1716. [Google Scholar] [CrossRef]
  98. Mylvaganam, G.H.; Chea, L.S.; Tharp, G.K.; Hicks, S.; Velu, V.; Iyer, S.S.; Deleage, C.; Estes, J.D.; Bosinger, S.E.; Freeman, G.J.; et al. Combination anti-PD-1 and antiretroviral therapy provides therapeutic benefit against SIV. JCI Insight 2018, 3, e122940. [Google Scholar] [CrossRef]
  99. Amancha, P.K.; Hong, J.J.; Rogers, K.; Ansari, A.A.; Villinger, F. In vivo blockade of the programmed cell death-1 pathway using soluble recombinant PD-1-Fc enhances CD4+ and CD8+ T cell responses but has limited clinical benefit. J Immunol. 2013, 191, 6060–6070. [Google Scholar] [CrossRef] [Green Version]
  100. Gill, A.L.; Green, S.A.; Abdullah, S.; Le Saout, C.; Pittaluga, S.; Chen, H.; Turnier, R.; Lifson, J.; Godin, S.; Qin, J.; et al. Programed death-1/programed death-ligand 1 expression in lymph nodes of HIV infected patients: Results of a pilot safety study in rhesus macaques using anti-programed death-ligand 1 (Avelumab). AIDS 2016, 30, 2487–2493. [Google Scholar] [CrossRef]
  101. Chen, P.; Chen, H.; Moussa, M.; Cheng, J.; Li, T.; Qin, J.; Lifson, J.D.; Sneller, M.C.; Krymskaya, L.; Godin, S.; et al. Recombinant Human Interleukin-15 and Anti-PD-L1 Combination Therapy Expands a CXCR3+PD1-/low CD8 T-Cell Subset in Simian Immunodeficiency Virus-Infected Rhesus Macaques. J. Infect. Dis. 2020, 221, 523–533. [Google Scholar] [CrossRef] [PubMed]
  102. Vargas-Inchaustegui, D.A.; Xiao, P.; Hogg, A.E.; Demberg, T.; McKinnon, K.; Venzon, D.; Brocca-Cofano, E.; Dipasquale, J.; Lee, E.M.; Hudacik, L.; et al. Immune targeting of PD-1(hi) expressing cells during and after antiretroviral therapy in SIV-infected rhesus macaques. Virology 2013, 447, 274–284. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Hryniewicz, A.; Boasso, A.; Edghill-Smith, Y.; Vaccari, M.; Fuchs, D.; Venzon, D.; Nacsa, J.; Betts, M.R.; Tsai, W.P.; Heraud, J.M.; et al. CTLA-4 blockade decreases TGF-beta, IDO, and viral RNA expression in tissues of SIVmac251-infected macaques. Blood 2006, 108, 3834–3842. [Google Scholar] [CrossRef] [Green Version]
  104. Harper, J.; Gordon, S.; Chan, C.N.; Wang, H.; Lindemuth, E.; Galardi, C.; Falcinelli, S.D.; Raines, S.L.M.; Read, J.L.; Nguyen, K.; et al. CTLA-4 and PD-1 dual blockade induces SIV reactivation without control of rebound after antiretroviral therapy interruption. Nat. Med. 2020, 26, 519–528. [Google Scholar] [CrossRef] [PubMed]
  105. Bradley, T.; Kuraoka, M.; Yeh, C.H.; Tian, M.; Chen, H.; Cain, D.W.; Chen, X.; Cheng, C.; Ellebedy, A.H.; Parks, R.; et al. Immune checkpoint modulation enhances HIV-1 antibody induction. Nat. Commun. 2020, 11, 948. [Google Scholar] [CrossRef] [Green Version]
  106. Pan, E.; Feng, F.; Li, P.; Yang, Q.; Ma, X.; Wu, C.; Zhao, J.; Yan, H.; Chen, R.; Chen, L.; et al. Immune Protection of SIV Challenge by PD-1 Blockade During Vaccination in Rhesus Monkeys. Front. Immunol. 2018, 9, 2415. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Finnefrock, A.C.; Tang, A.; Li, F.; Freed, D.C.; Feng, M.; Cox, K.S.; Sykes, K.J.; Guare, J.P.; Miller, M.D.; Olsen, D.B.; et al. PD-1 blockade in rhesus macaques: Impact on chronic infection and prophylactic vaccination. J. Immunol. 2009, 182, 980–987. [Google Scholar] [CrossRef] [Green Version]
  108. Rahman, S.A.; Yagnik, B.; Bally, A.P.; Morrow, K.N.; Wang, S.; Vanderford, T.H.; Freeman, G.J.; Ahmed, R.; Amara, R.R. PD-1 blockade and vaccination provide therapeutic benefit against SIV by inducing broad and functional CD8(+) T cells in lymphoid tissue. Sci. Immunol. 2021, 6, eabh3034. [Google Scholar] [CrossRef]
  109. Wu, C.; He, Y.; Zhao, J.; Luo, K.; Wen, Z.; Zhang, Y.; Li, M.; Cui, Y.; Liu, Z.; Wang, C.; et al. Exacerbated AIDS Progression by PD-1 Blockade during Therapeutic Vaccination in Chronically Simian Immunodeficiency Virus-Infected Rhesus Macaques after Interruption of Antiretroviral Therapy. J. Virol. 2022, 96, e0178521. [Google Scholar] [CrossRef]
  110. Bekerman, E.; Hesselgesser, J.; Carr, B.; Nagel, M.; Hung, M.; Wang, A.; Stapleton, L.; von Gegerfelt, A.; Elyard, H.A.; Lifson, J.D.; et al. PD-1 Blockade and TLR7 Activation Lack Therapeutic Benefit in Chronic Simian Immunodeficiency Virus-Infected Macaques on Antiretroviral Therapy. Antimicrob. Agents Chemother. 2019, 63, e01163-19. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Immune checkpoint proteins impair proliferation and multi-functionality of T cells. Interaction between immune checkpoint markers and their ligands transmits inhibitory signals to CD4+ and CD8+ T cells. These inhibitory signals limit TCR- and CD28-mediated co-stimulatory pathway, including the NF-κB, NFAT, and AP-1 signaling pathways, which suppress proliferation, cytokine production, and cytotoxic response. IC blockers are able to cause ligand dissociation from the ICs and restore T cell activation.
Figure 1. Immune checkpoint proteins impair proliferation and multi-functionality of T cells. Interaction between immune checkpoint markers and their ligands transmits inhibitory signals to CD4+ and CD8+ T cells. These inhibitory signals limit TCR- and CD28-mediated co-stimulatory pathway, including the NF-κB, NFAT, and AP-1 signaling pathways, which suppress proliferation, cytokine production, and cytotoxic response. IC blockers are able to cause ligand dissociation from the ICs and restore T cell activation.
Viruses 14 00581 g001
Figure 2. Immune checkpoint proteins contribute to HIV/SIV latency. PD-1, TIGIT, LAG-3, CTLA-4, Tim-3, and BTLA signaling pathways are involved in generating an HIV reservoir and its persistence. The binding of these ICs and their ligands enforces silencing of integrated HIV DNA at the transcriptional and/or translational level. IC blockers can reactivate HIV from the cellular reservoir and produce new virions by blocking IC-ligand interactions in CD4+ T cells.
Figure 2. Immune checkpoint proteins contribute to HIV/SIV latency. PD-1, TIGIT, LAG-3, CTLA-4, Tim-3, and BTLA signaling pathways are involved in generating an HIV reservoir and its persistence. The binding of these ICs and their ligands enforces silencing of integrated HIV DNA at the transcriptional and/or translational level. IC blockers can reactivate HIV from the cellular reservoir and produce new virions by blocking IC-ligand interactions in CD4+ T cells.
Viruses 14 00581 g002
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Sun, Y.; Xue, J. Expression Profile and Biological Role of Immune Checkpoints in Disease Progression of HIV/SIV Infection. Viruses 2022, 14, 581. https://0-doi-org.brum.beds.ac.uk/10.3390/v14030581

AMA Style

Sun Y, Xue J. Expression Profile and Biological Role of Immune Checkpoints in Disease Progression of HIV/SIV Infection. Viruses. 2022; 14(3):581. https://0-doi-org.brum.beds.ac.uk/10.3390/v14030581

Chicago/Turabian Style

Sun, Yuting, and Jing Xue. 2022. "Expression Profile and Biological Role of Immune Checkpoints in Disease Progression of HIV/SIV Infection" Viruses 14, no. 3: 581. https://0-doi-org.brum.beds.ac.uk/10.3390/v14030581

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop