A disease of more than 39.6 million people worldwide, HIV-1 infection has no curative therapy. To date, one man has achieved a sterile cure, with millions more hoping to avoid the potential pitfalls of lifelong antiretroviral therapy and other HIV-related disorders, including neurocognitive decline. Recent developments in immunotherapies and gene therapies provide renewed hope in advancing efforts toward a sterilizing or functional cure.
KEYWORDS: HIV-1, reservoir, latency, functional cure, checkpoint inhibitors, CRISPR/Cas9, brain
ABSTRACT
A disease of more than 39.6 million people worldwide, HIV-1 infection has no curative therapy. To date, one man has achieved a sterile cure, with millions more hoping to avoid the potential pitfalls of lifelong antiretroviral therapy and other HIV-related disorders, including neurocognitive decline. Recent developments in immunotherapies and gene therapies provide renewed hope in advancing efforts toward a sterilizing or functional cure. On the horizon is research concentrated in multiple separate but potentially complementary domains: vaccine research, viral transcript editing, T-cell effector response targeting including checkpoint inhibitors, and gene editing. Here, we review the concept of targeting the HIV-1 tissue reservoirs, with an emphasis on the central nervous system, and describe relevant new work in functional cure research and strategies for HIV-1 eradication.
INTRODUCTION
Control of human immunodeficiency virus (HIV) infection can be achieved with optimum antiretroviral therapy (1), but this does not provide a disease cure. HIV persists in latently infected CD4+ T cells and is integrated into the host genome until cell death. The introduction of the first antiretroviral drug, zidovudine, in 1987, followed by combination antiretroviral therapy (ART) in 1996, transformed the epidemic by increasing survival and converting HIV infection from a life-threatening illness into a chronic infection. However, ART is not available to all, and compliance rates vary. Even in carefully managed patients, the consequences of lifelong medication side effects are challenging, and disease trajectory is variable. There is concern about frailty and accelerated aging with end-organ involvement. This includes HIV-associated neurocognitive disorders (HAND) and major depressive disorder (MDD), which can lead to gradual but significant functional deterioration that complicates their management (2). For these reasons, much hope has rested on finding a cure for the infection.
Despite massive efforts to eradicate HIV, challenges persist in achieving a “cure.” The history of cure strategies is one of repeated failures and false hopes. When ART was first introduced, it was thought that the size of the HIV DNA reservoir would decrease over time on prolonged therapy and the virus would eventually be cleared. Early mathematical models, such as the Ho-Shaw model, were published in high-profile journals and received wide publicity (3 – 5). Unfortunately, the ability of HIV to persist in resting CD4 T cells and tissue reservoirs almost indefinitely despite ART was underappreciated and only later fully recognized (6 – 8).
A major shift in HIV research occurred when the “Berlin patient” achieved a sterile cure following radiation treatment, chemotherapy, and a hematopoietic stem cell transplant (HSCT) from a donor with a homozygous deletion in the gene encoding the HIV coreceptor C-C chemokine receptor type 5 (CCR5Δ32) (9). Although subsequent studies have been unable to reproduce the success using homozygous CCR5Δ32 transplants (10 – 14), there have been isolated cases of suspected “cures,” such as the “Mississippi baby,” a perinatally infected child who had an undetectable viral load for 27 months after stopping ART before rebound (15, 16); this case led to the hypothesis that if children received ART at birth, treatment could be safely stopped after some time. In each case, viral rebound eventually occurred, suggesting that tissue reservoirs are established early in the disease and cannot be eradicated by ART alone. Recently, a second potential sterile-cure case, the “London patient,” was identified (17). This patient underwent a much less aggressive preconditioning for HSCT from a donor homozygous for the CCR5Δ32 allele after receiving chemotherapy for relapsed nodular sclerosing Hodgkin’s lymphoma. Unlike the Berlin patient, the London patient was homozygous for wild-type CCR5 prior to transplant and did not receive whole-body irradiation. The London patient has remained in HIV-1 remission for more than 18 months.
The term “HIV reservoir” has been variably used to describe the cells and tissues that continue to harbor HIV under optimum therapy. Some definitions include all proviruses that participate in HIV pathogenesis regardless of replication competence (18), while others include only replication-competent forms of HIV that are capable of reestablishing infection in the absence of ART (19). We prefer the former definition, since mutations and recombination events can convert a replication-incompetent viral sequence to a replication-competent virus. Further, a defective viral sequence may harbor open reading frames for individual viral proteins or produce noncoding RNA which may have biological effects. HIV reservoirs can further be divided according to scale into anatomical, cellular, and molecular reservoirs (Fig. 1), which are comprised of the organs/tissues that harbor infected cells, the HIV-containing cells themselves, and the characteristics of the HIV genomes contained within those cells, respectively. At the molecular level, the reservoir can be further subdivided into latent, persistent, and replication-defective categories. In a latent reservoir, the virus achieves complete transcriptional silencing and must be reactivated to release a replicating viral particle. HIV infection of the resting memory CD4 T cells fulfills this definition (20). However, any long-lived cell infected with the virus could serve as a persistent viral reservoir where small amounts of virus are continually or intermittently released. Macrophages likely fall into this category, though their exact contribution to chronic HIV infection is undetermined. While monocyte-derived macrophages can be persistently infected in vitro (21, 22), there is considerable debate about whether myeloid cells can support latent HIV infection (23 – 28). However, macrophages can harbor transcriptionally active unintegrated HIV DNA for more than 30 days in culture (29).
FIG 1.
Summary of types of HIV reservoirs. (A and B) There are several anatomical compartments (A) that are populated by HIV-infected cells (B). (C) The integrated provirus contained within these cells may be transcriptionally silent (latent), transcriptionally active and capable of producing infectious virions (persistent), or transcriptionally active but replication defective due to mutations or deletions in the HIV genome, leading to translation of specific viral proteins for which an open reading frame remains intact.
Data from simian immunodeficiency virus (SIV) models suggest that viral DNA (vDNA) within tissue-resident macrophages is often due to phagocytosis of infected CD4+ T cells rather than true infection (30, 31). The researchers observed that vDNA was contained in macrophages only in tissues that were not depleted of CD4+ cells (30) and that no replication-competent virus could be detected from macrophages of animals treated with ART (31). Similarly, vDNA could not be detected in alveolar macrophages isolated from HIV-positive patients on long-term ART with undetectable viral loads (31). However, others have shown that phagocytosis of infected CD4+ T cells can yield productive macrophage infection (32). In humanized myeloid-only mice (MoM) infected with HIV and suppressed with ART, viral rebound occurred in 3/9 (33%) mice 7 weeks after treatment was removed (33). Further, macrophages isolated from the urethras of three individuals on suppressive ART contained not only integrated vDNA but also HIV RNA, proteins, and viral particles, and they could produce replication-competent virus when stimulated with lipopolysaccharide (34). Together, these findings support the establishment of a myeloid reservoir in some HIV-infected individuals. Microglial cells and perivascular macrophages containing integrated vDNA have also been detected in postmortem central nervous system (CNS) tissue (35), which supports a myeloid reservoir in the brain. It is now important to better elucidate the characteristics of the macrophage reservoir, particularly because these cells are long-lived and resist the cytopathic effects of HIV (36).
Some cells harbor defective viral sequences. These cells, while incapable of producing infectious virus, may have open reading frames for viral proteins which may play a role in disease pathogenesis (37). There is also the possibility that either through a recombination event or via DNA repair mechanisms, viral production may occur. While these replication-defective viral sequences are poorly studied in the context of HIV infection, they have been extensively studied in the context of endogenous retroviruses, where the vast majority of the viruses are defective and may play a pathogenic role in neurodegenerative diseases and cancer (38). Hence a “sterilizing cure” should eradicate all three forms of molecular reservoirs. The terms “functional cure” and “remission” are used to describe approaches that prevent the production of infectious virus. However, it may be necessary to also control the production of all viral proteins to achieve a functional cure.
BRAIN RESERVOIR
While much is known about the lymphoid reservoirs in major end organs, the brain is difficult to study. Tissue is accessible only at autopsy, and inference during life is made by study of the cerebrospinal fluid (CSF) that bathes the brain. Substances that are unique to the CSF, such as divergence of viral strains between blood and CSF, or those found in higher concentrations in CSF than in blood are considered to be derived from the brain.
In well-controlled HIV-positive patients, immune activation can be present even when HIV is undetectable in the CSF, indicating a persistent response to the underlying infection (39). HIV proteins such as Tat have been found in the CSF, and antibody responses against Tat correlate with CSF viral load and inversely correlate with CD4 cell count (40). Interestingly, the presence and abundance of antibodies against HIV proteins in blood and CSF, especially p24, have been implicated as a representative measure of curative interventions and could potentially be used to evaluate reservoir eradication in the brain (41, 42). Studies on CSF viral escape have shown divergent CSF sequences compared to those in blood, suggesting compartmentalization of the brain reservoir in those patients (43 – 45).
While studies from the preantiretroviral era clearly show that HIV can infect cells within the brain, including microglia, astrocytes, macrophages, and even rare neurons (46, 47), it remains unknown if the reservoir persists in patients on ART. In SIV macaque models, myeloid cells in the brain are known to be a reservoir for replication-competent virus (48 – 50).
Studies using viral next-generation sequencing in the CSF and periphery have demonstrated distinct compartmentalization of viral subspecies, especially in patients with HIV-associated dementia (51). Up to 10% of patients on suppressive antiretroviral therapy experience low levels of CSF escape, characterized by detectable HIV RNA in CSF but not in blood (52). Our laboratory has found that HIV Tat protein can be detected in the CSF from >35% of HIV-positive individuals on suppressive ART (53), indicating that Tat levels may be an indirect measure of the brain viral reservoir. Collectively, these findings highlight the importance of considering multiple tissues, including the brain, in cure research strategies. Persistent Tat expression and CSF escape in otherwise well-controlled patients strongly suggest that a CNS reservoir exists in a subset of patients and may be capable of reseeding the periphery even if the memory T-cell reservoir is eradicated or silenced.
NOVEL DIRECTIONS IN HIV CURE STRATEGIES
The goal of HIV-1 remission strategies is to decrease the size of the replication-competent reservoir by (i) reactivating latent virus, termed “kick,” while improving anti-HIV immune responses to eliminate infected cells, termed “kill,” (ii) genetically engineering cells to make them resistant to HIV infection, or (iii) inducing epigenetic changes or chromatin remodeling to permanently prevent transcriptional activation of the viral genome, termed “block and lock.”
“Kick-and-kill” approaches to eliminate viral reservoirs.
Several strategies have been proposed to reactivate HIV expression from latently infected CD4+ T cells. Latency-reversing agents (LRAs) include the following: (i) epigenetic modifiers, including histone deacetylase inhibitors (HDACi) and histone methyltransferase inhibitors, which induce chromatin remodeling at the HIV LTR; (ii) protein kinase C (PKC) agonists and activators of the NFκB pathway; (iii) bromodomain extraterminal (BET) motif inhibitors, which enhance recruitment of P-TEFb to the HIV promoter; and (iv) cytokine/chemokine and Toll-like receptor (TLR) agonists, which activate immune signaling pathways in T cells. All of these are reviewed elsewhere (54 – 56), so our discussion is limited to agents being actively investigated in clinical trials or in early stages of development (Table 1).
TABLE 1.
Summary of active clinical trials involving latency-reversing agentsa
LRA | Secondary agent(s) | No. of patients | Status | Identifier |
---|---|---|---|---|
Nicotinamide (SIRT1 inhibitor) | Dendritic cell vaccine + auranofin + ART intensification | 30 | Active | NCT02961829 |
Vorinostat (HDACi) | ChAdV63.HIVconsv (ChAd) prime and MVA.HIVconsv boost vaccines | 60 | Active | NCT02336074 |
Disulfiram | 15 | Terminated due to AE | NCT03198559 | |
HXTCb | 12 | Recruiting | NCT03212989 | |
Tamoxifen | 30 | Active | NCT03382834 | |
AGS-004 DC therapy | 6 | Terminated (AGS-004 supply unavailable) | NCT02707900 (VOR-VAX) | |
VRC07-523LS | 12 | Recruiting | NCT03803605 | |
Panobinostat (HDACi) | Pegylated IFN-α2a | 34 | Recruiting | NCT02471430 |
Romidepsin (HDACi) | 3BNC117 | 30 | Active | NCT02850016c |
MVA vector HIV vaccine + HIVACAR01 (personalized HIV vaccine) + 10-1074 | 56 | Not yet recruiting | NCT03619278 | |
3BNC117 ab | 60 | Recruiting | NCT03041012 | |
3BNC117 ab | 42 | Not yet recruiting | RV 438 | |
Valproic acid (HDACi) | Pyrimethamine | 28 | Recruiting | NCT03525730 |
Chidamide (HDACi) | None | 60 | Active | NCT02902185 |
CAR-T or TCR-T-cell therapy | 40 | Recruiting | NCT03980691 | |
Euphorbia kansui (ingenol) (PKC agonist) | None | 9 | Recruiting | NCT02531295 |
Lefitolimod (MGN1703) (TLR9 agonist) | 10-1047 + 3BNC117 | 48 | Recruiting | NCT03837756c |
GS-9620 (TLR7 agonist) | None | 28 | Active | NCT03060447c |
Pegylated IFN-α2a | None | 54 | Active | NCT02227277 |
3BNC117 + 10-1074 | 21 | Not yet recruiting | NCT03588715c | |
Recombinant human superagonist IL-15 (ALT-803/N-803) | None | 10 | Active | NCT02191098 |
Haploidentical NK cell adoptive transfer | 8 | Recruiting | NCT03899480 |
HIV transcriptional latency is mediated largely by epigenetic silencing (57, 58); hence, HIV cure research over the past decade has focused on HDACi (59, 60). There are currently 15 clinical trials using epigenetic modifiers in HIV-infected individuals, either alone or in combination with another LRA or immune-boosting therapy (Table 1). To date, none of these studies has exhibited a significant decrease in integrated DNA levels in the CD4+ T-cell reservoir or delay in viral rebound following treatment interruption, despite immune activation and increases in cell-associated and plasma HIV RNA (54 – 56).
TLR agonists have shown promise in nonhuman primate studies, but their use in humans is in early stages. The TLR7 agonist GS-9620, in combination with an Ad26/modified vaccinia virus Ankara (MVA) therapeutic vaccination, enhanced antiviral immune responses and delayed viral rebound in SIV-infected macaques (61). Similarly, coadministration of GS-9620 and a broadly neutralizing antibody (PGT121) prevented viral rebound following treatment interruption in simian-human immunodeficiency virus (SHIV)-infected macaques (62). This was not observed with either GS-9620 or PGT121 alone, indicating that TLR agonists require pairing with other therapeutics. A phase 1 clinical trial investigating GS-9620 in HIV-infected individuals on combination ART (NCT03060447) is under way (Table 1).
The interleukin-15 (IL-15) superagonist ALT-803, which has been shown to reactivate HIV in latently infected T cells and enhance cytotoxic T-cell responses ex vivo (63), is being evaluated in individuals on long-term ART (NCT02191098) (54). Another cytokine, polyethylene glycol-alpha interferon 2a (polyethylene glycol-IFN-α2a), decreased HIV-1 DNA levels and delayed viral rebound after treatment interruption (64). A trial comparing the effects of a repeated low dose (1 μg/kg per week) versus a higher dose (180 mg per week) on viral reservoirs is under way (NCT02227277); it is also being investigated in combination with HIV-targeted broadly neutralizing antibodies (NCT03588715) (54).
Clinical data suggest that LRAs are insufficient to reduce the size of the viral reservoir when delivered as a monotherapy. Thus, ongoing studies combine different classes of LRAs or pair an LRA with an immune-boosting intervention such as therapeutic vaccination (Table 1) (54 – 56). Further, the antiviral immune response is impaired in chronic HIV infection due to chronic inflammation and immune exhaustion. Cytotoxic CD8+ T cells are unable to clear HIV-infected cells even with latency reversal, which contributes to the failure of “kick-and-kill” strategies (55, 65). Additionally, certain LRAs, such as HDACi, suppress cytotoxic T-cell responses (66). Therefore, immunomodulatory agents may be vital in successfully eliminating or reducing the viral reservoir.
Checkpoint inhibitor therapies.
Immune checkpoint molecules are cell surface receptors that regulate immune responses. Stimulatory immune checkpoint proteins provide costimulatory signals that enhance immune activation, while inhibitory immune checkpoint molecules negatively regulate immune cell function and play an important role in resolution of immune responses and maintenance of self-tolerance. During chronic viral infections, including HIV infection, upregulation of inhibitory checkpoint molecules on immune cells contributes to T-cell exhaustion characterized by loss of effector functions and failure to proliferate in response to antigen (67). Checkpoint molecules are enriched on the surface of HIV-infected CD4+ T cells in ART-treated individuals, and the number of cells expressing these molecules is tightly correlated with the size of the T-cell viral reservoir (68 – 70). Additionally, coinhibitory molecules such as programmed cell death 1 (PD-1) and cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) function as biomarkers for low-level HIV/SIV viral replication, and in vitro blockade of these molecules restores CD4+ T-cell function (71, 72). HIV-infected individuals receiving checkpoint inhibitors as immunotherapy for comorbid cancer have increased immune function and stable to decreased cell-associated HIV DNA (69, 73 – 77). These reports suggest that T-cell checkpoints hold promise for the treatment of chronic infections (78, 79), including use in cure strategies for HIV reservoirs (Fig. 2).
FIG 2.
Summary of immune checkpoint receptor interactions and gene editing strategies. (1) Coinhibitory checkpoint molecules such as PD-1, CTLA-4, TIGIT, Tim-3, LAG3, BTLA-4, CD160, and 2B4 are upregulated on T cells during HIV infection and negatively regulate T-cell proliferation and effector function when bound to their cognate ligands. In contrast, the costimulatory molecules CD28, CD226, and GITR enhance T-cell activation when ligated in conjunction with TCR-mediated stimulation. Checkpoint inhibitors that block binding of the indicated molecule to its target to enhance immune function are indicated in blue boxes, while the agonist TRX518, which mimics the interaction of GITR with its ligand, is indicated in yellow. Other potential HIV cure strategies include (2a) editing of the host CCR5 gene to generate resistance to HIV infection or (2b) deletion or indel-mediated transcriptional silencing of the HIV provirus by the CRISPR/Cas9 system, (3a) posttranscriptional silencing of HIV gene expression by antisense oligonucleotides complementary to viral mRNA or novel compounds, and (3b) inhibition of Tat-mediated LTR transactivation by small-molecule inhibitors identified by high-throughput screening.
(i) PD-1 and PD-L1.
PD-1 is an immune checkpoint molecule that induces tolerance and modulates antigen-specific immune responses to infection along with its two ligands, PD-L1 and PD-L2 (80). Upregulation of PD-1 on T cells correlates with HIV disease progression, and anti-PD-1 restores T-cell effector responses against the virus (73). In an SIV model, anti-PD-1 with ART augmented antiviral T-cell responses and decreased the CD4+ T-cell viral reservoir (81). In vivo, exposure to anti-PD-1 decreased the number of latently infected cells, and in another HIV-positive individual, anti-PD-1 therapy reversed HIV latency (82).
Follicular helper T (Tfh) (CXCR5+ PD-1+) cells are important in providing support to maturing germinal center B cells in lymph node (LN) tissue reservoirs. These cells exhibit high PD-1 surface expression and are a major source of replication-competent latent HIV (83). Blockade of PD-1 in SIV-infected macaques enhanced anti-HIV B-cell responses, suggesting some restoration of Tfh function (84). In aviremic individuals on long-term combination ART, PD-1+ Tfh cells accounted for 46% of inducible replication-competent virus and 96% of infectious virus arising from the memory CD4 T-cell reservoir, suggesting a potential target for cure therapy (68).
Pembrolizumab was the first anti-PD-1 agent approved by the FDA, in 2014, as a humanized IgG4κ monoclonal antibody (80). Since then, another anti-PD-1 IgG4 human monoclonal antibody and three anti-PD-L1 monoclonal antibodies have been approved (Table 2).
TABLE 2.
Brief survey of on-market or in-development checkpoint inhibitors with HIV-targeting potential
Stage | Checkpoint | Inhibitor(s) | Trial(s) (phase) |
---|---|---|---|
On market | PD-1 | Pembrolizumab (Keytruda), nivolumab (Opdivo) | |
PD-L1 | Atezolizumab (Tecentriq), avelumab (Bavencio), durvalumab (Imfinzi) | ||
CTLA-4 | Ipilimumab, MDX-010 (Yervoy), tremelimumab (orphan designation) | ||
In trials | TIGIT | BMS-986207 | NCT02913313 (1/2) |
OMP-313M32 | NCT03119428 (1) | ||
MTIG7192A | NCT02794571 (1) | ||
Tim-3 | TSR-022 | NCT02817633 (1) | |
LAG3 | Relatlimab (previously BMS-986016) | 11 clinical trials (1, 1/2, 2/3) | |
GITR | TRX518 | NCT01239134 (1), NCT02628574 (1) | |
Preclinical | Anti-CD160 | ELB021 (Elsalys Biotech) | |
Anti-BTLA | 40E4 chimericIgG1 (Merck & Co.) |
The ligand to PD-1, PD-L1, decreases the proliferation of antigen-specific effector T cells. It also has affinity to B7.1 and uses this pathway to inhibit T-cell responses (85). An anti-PD-L1 monoclonal antibody, BMS-936559, was tested in a phase 1, randomized, placebo-controlled, dose-escalating trial in HIV-infected participants on antiretroviral therapy (NCT02028403). The trial was halted prematurely after enrolling one low-dose arm (0.3 mg/kg), due to the development of retinal toxicity in macaques with higher doses of BMS-936559 (86). However, HIV Gag-specific CD8+ T cells increased from baseline through the 28-day trial in two of the six patients who received active drug (87), suggesting enhancement of antiviral cytotoxic T-lymphocyte (CTL) responses.
Current clinical trials are focusing on PD-1 due to the presumption of a lower risk of adverse events (AE) for this target compared to PD-L1. There are three actively recruiting trials using pembrolizumab in HIV-positive individuals without cancer; we are investigators on two of these trials. The first (NCT03367754) is a double-blind, placebo-controlled phase 1 trial targeting individuals with HIV-1 infection on ART but low CD4+ T-cell counts (<350 cells/mm3) (Table 3). The second (NCT03239899) is an open-label, phase 1 trial targeting the HIV CNS reservoir using pembrolizumab. The third (NCT03787095) is an AIDS Clinical Trials Group (ACTG) placebo-controlled phase 1/2 dose escalation trial targeting individuals with HIV on ART who have normal CD4+ T-cell counts, assessing HIV-1 Gag-specific CD8+ T-cell responses (87).
TABLE 3.
Clinical trials using checkpoint inhibitors in HIV-infected populations
Trial | Study drug | Target(s) | Population | Phase |
---|---|---|---|---|
NCT03787095 | Cemiplimab | PD1 | Suppressed HIV on ART | 1/2 |
NCT03239899 | Pembrolizumab | PD-1 | CNS HIV reservoir | 1 |
NCT03367754 | Pembrolizumab | PD-1 | HIV with low CD4+ cell count | 1 |
NCT02595866 | Pembrolizumab | PD-1 | HIV and malignant neoplasms | 1 |
NCT03304093 | Nivolumab | PD-1 | HIV and non-small-cell lung cancer | 2 |
NCT02408861 | Nivolumab and ipilimumab | PD-1, CTLA-4 | HIV and malignant neoplasms | 1 |
NCT03316274 | Nivolumab | PD-1 | HIV and Kaposi sarcoma | 1 |
NCT03407105 | Ipilimumab | CTLA-4 | HIV | 1 |
NCT03094286 | Durvalumab | PD-L1 | HIV and solid tumors | 2 |
(ii) CTLA-4.
Cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) is a homolog of the costimulatory molecule CD28 that negatively regulates T-cell activation when bound to its receptor B7. Since CTLA-4 and CD28 directly compete for binding to B7, the relative expression of these molecules determines whether a T cell will undergo activation or anergy. CTLA-4 expression on the cell surface is upregulated by T-cell receptor (TCR) and CD28 signaling and attenuates T-cell responses during the resolution phase of an immune response (88). CTLA-4 expression is moderately increased in HIV-specific CD4+ T cells from patients and is associated with higher viral loads and inversely correlated with CD4 counts (89).
In a case report of an HIV-infected patient receiving the anti-CTLA-4 therapy ipilimumab (MDX-010) for cancer, there was a decrease in plasma HIV RNA following each infusion and a slight increase in CD4 T-cell count and T-cell activation markers (90). A phase 1, open-label, dose escalation study of MDX-010 as immunotherapy in HIV-infected individuals (NCT03407105) was conducted in 2006. The results remain unpublished but have been cited as not having safety concerns (91). Combination and individual immunotherapy trials targeting CTLA-4 and PD-1 are in progress for HIV-1 related malignancies (Table 3).
In ART-treated, SIV-infected macaques CTLA-4+ PD1+ Tfh cells were more frequent than PD1+ CTLA4− Tfh cells within LNs (70). Additionally, there was a population of CTLA-4+ PD1− latently infected, replication-competent cells in the LN and splenic T-cell zone that persisted after ART, which was confirmed in HIV-infected individuals on ART, supporting that multiple checkpoint targets are necessary in cure strategies.
(iii) TIGIT, LAG3, and Tim-3.
Additional checkpoints such as TIGIT (T-cell immunoreceptor with Ig and ITIM domains), LAG3 (lymphocyte-activation gene 3), and Tim-3 (T-cell immunoglobulin and mucin domain containing-3) have been identified as targets of T-cell function enhancement in antiviral targeting strategies.
Tim-3 is a transmembrane protein that binds to galectin-9 and phosphatidylserine (92 – 94). It is a marker of T-cell exhaustion that is upregulated on the CD8+ T cells of HIV-infected individuals, correlating with HIV-1 viral load and disease progression (95, 96). CD8+ T cells expressing Tim-3 have impaired cytotoxic function, cytokine production, and proliferative capability (97). Blockade of Tim-3 using a recombinant Tim-3 glycoprotein or an anti-Tim-3 monoclonal antibody restores the proliferation of specific CD8+ T cells in response to HIV antigens (97). In HIV infection, Tim-3 expression is induced on plasmacytoid dendritic cells, resulting in reduced production of IFN-α (98). Interestingly, treating activated CD4+ T cells with a soluble form of the ligand for Tim-3, galectin-9, conferred resistance to HIV infection due to decreased expression of HIV coreceptors (CCR5 and CXCR4) and upregulation of the host restriction factor p21 (99). Conversely, in resting CD4+ T cells, which do not express high levels of Tim-3, galectin-9 enhanced HIV infection by binding to protein disulfide isomerase to alter the cell surface redox state and promote viral entry (99, 100). Thus, Tim-3 ligands must be evaluated cautiously as therapies for HIV infection.
Coexpression of PD-1, TIGIT, and LAG3 was associated with an increased frequency of CD4+ T cells harboring integrated HIV DNA (69), suggesting a clonal expansion of the cells. TIGIT suppresses T-cell activation by interaction with its ligand, poliovirus receptor (PVR), on dendritic cells in a manner analogous to that for CTLA-4, PD-1, and B- and T-lymphocyte attenuator (BTLA) (101).
LAG3 is a major histocompatibility complex (MHC) class II ligand, related to the CD4 superfamily, that is a coinhibitory stimulus (102). LAG3 expression on HIV-specific CD8+ T cells is negatively correlated with HIV RNA (102). LAG3 suppression increases T-cell proliferation and effector cytokine production. Expression of LAG3 in conjunction with Tim-3 on CD4+ CD25+ cells suppresses proinflammatory macrophage activation (103, 104). The exact mechanism of action of LAG3, like that of many of the T-cell cosignals, is likely to be influenced by the expression of other cell surface molecules. Tim-3-, TIGIT-, and LAG3-targeting immunotherapies are being evaluated in clinical trials (Table 2), though not with antiviral intent.
(iv) CD160, CD244/2B4, BTLA, and GITR.
Other targets for antibody-based therapies include the T-cell coinhibitory molecule CD160. Preclinical studies are under way in the European Union. Epstein-Barr virus (EBV)-, cytomegalovirus (CMV)- and influenza virus-specific CD8+ T cells expressing CD160 have reduced cytotoxic activity in response to viral antigens (105). Regulation of T-cell function by CD160 is independent of PD-1 expression, highlighting an alternate pathway to increase T-cell proliferation and CD8+ T-cell-mediated cell killing (Table 2) (105).
2B4 (CD244), a member of the CD2/SLAM signaling family, is an immune checkpoint molecule that is a marker of disease progression and exhaustion in HIV infection (106, 107). Abrogating 2B4 binding to its ligand CD48 using an anti-CD48 antibody increased proliferation of HIV-specific CD8+ T cells in response to HIV antigens, and blockade of both PD-1 and 2B4 further increased proliferation (107). However, the downstream signaling effects can be unpredictable and are dependent on cell type, ligand density, and availability of downstream signaling molecules (108).
B- and T-lymphocyte attenuator (BTLA) (CD272) is member of the immunoglobulin superfamily that negatively regulates T-cell proliferation and effector function. BTLA expression is downregulated during HIV infection, which correlates with disease progression (109). An anti-BTLA blocking antibody increased proliferation of CD8+ T cells from HIV-infected patients in response to HIV antigens, which was further enhanced by blockade of both PD-1 and BTLA (110). Similar effects were observed with dual blockade of PD-1 and Tim-3, and triple treatment with blocking antibodies against PD-1, BTLA, and CD160 increased production of proinflammatory cytokines compared to that with anti-PD-1 alone (110). A BTLA-targeting monoclonal IgG1 is being developed by Merck to supplement PD-1 therapies (Table 2) (111).
Costimulatory targets, such as glucocorticoid-induced tumor necrosis factor receptor (GITR), enhance CD8+ T-cell response and resolve chronic lymphocytic choriomeningitis virus (LCMV) infection in mice, signaling a potential role for immunotherapy in HIV disease (112). TRX518 is an agonist monoclonal antibody that mimics interaction of GITR with its ligand to activate antigen-specific T cells and suppress regulatory T-cell responses. Two phase 1 clinical studies with TRX518 in solid tumors (NCT01239134 and NCT02628574) are under way (Table 2). Other coinhibitory and costimulatory checkpoints have been identified as molecular signatures of immune exhaustion in HIV infection and are potential targets for immunological therapies (113, 114).
Gene editing.
Gene editing is a rapidly evolving field, and significant progress has been made in optimizing delivery and efficacy of these tools. However, the possibility of off-target effects due to binding at homologous sequences remains a serious concern, and there are continued challenges in increasing efficiency of mutation/excision at the targeted site and ensuring delivery to all relevant anatomical compartments. We include here a summary of current gene editing approaches that have been applied to HIV cure strategies.
(i) Genome cutting/excision.
Several genome engineering approaches have been used to (i) excise all or part of the provirus from the human genome, (ii) introduce mutations to render HIV incapable of replication, or (iii) modify the structure of HIV coreceptors on immune cells to make them resistant to infection. These approaches include zinc finger nucleases (ZFNs), transcription activator-like effector nucleases (TALENs), and the clustered regularly interspaced short palindromic repeat (CRISPR) and CRISPR-associated (Cas) system. ZFNs and TALENS are artificial restriction enzymes composed of an endonuclease domain fused to a DNA binding domain. The DNA binding specificity of ZFNs is determined by a series of zinc finger repeats, each of which recognizes a specific 3-bp DNA sequence. TALENs utilize 18 repeats of a 34-amino-acid TAL effector DNA binding domain (TALE), which dictates the nucleotide it recognizes (115, 116).
The discovery of CRISPR/Cas genes has reinvigorated the field of gene editing (117, 118). This approach utilizes a short guide RNA (gRNA) containing a 20-nucleotide sequence in the 5′ end that is complementary to the target DNA and a conserved region in the 3′ end that recruits Cas9 endonuclease, which cleaves target genomic DNA. Unlike ZFNs and TALENs, the CRISPR system does not require a unique enzyme for each nucleic acid target; rather, a reusable Cas9 enzyme is paired with a variety of gRNAs that bind to different regions of the target DNA. All three nucleases silence expression primarily through insertions or deletions (indels) that are introduced during nonhomologous end joining (NHEJ)-mediated repair of double-strand DNA breaks at the target site.
It is important to consider the relative advantages and technical challenges of each of the approaches outlined above when applying these technologies to HIV therapy. For example, the small size of the genes encoding ZFNs (∼1 kb) is an advantage in terms of effective gene delivery compared to TALENs (∼3 kb) or Cas9 (∼4 kb), but ZFNs are more prone to off-target cleavage than both TALENs and CRISPR/Cas9 due to binding to genomic sites that are similar to the target sequence. TALENs offer more predictable DNA binding than ZFNs and are substantially more cost-effective to generate because they do not require extensive screening to ensure optimal binding to the target without significant off-target toxicity. However, the presence of repetitive sequences in TALENS make them unsuitable for lentiviral vectors, and constructs encoding TALENs are generally too large for incorporation into adeno-associated virus (AAV) vectors. The gRNA-guided Cas9 system, in contrast, is compatible with AAV-mediated delivery and provides significant advantages over other methods in terms of target site selection and the possibility of multiplexing gRNAs to target a single Cas9 nuclease to multiple sites. Newer versions of Cas protein are now being developed that are smaller and have multiple other advantages over Cas9.
(ii) CCR5.
The most pursued genome manipulation strategy in HIV research involves disruption of the coreceptor C-C chemokine receptor type 5 (CCR5). The CCR5Δ32 allele correlates with resistance to HIV infection, decreased viral loads, and long-term survival. The “Berlin patient,” who received cells from a donor with a homozygous CCR5Δ32 for treatment of acute myeloid leukemia, has remained HIV free for almost a decade and achieved a sterilizing cure (9, 119). Similarly, the “London patient,” who received a hematopoietic stem cell transplant (HSCT) from a CCR5Δ32 donor as part of treatment for Hodgkin’s lymphoma, has remained in remission after more than 18 months (17). Due to the low number of homozygous CCR5Δ32 donors in the population, HSCT is not a viable option for most HIV-infected patients. However, gene editing strategies have targeted CCR5. It is necessary for both alleles of the CCR5 gene to be disrupted to generate resistance to HIV infection, though patients heterozygous for the CCR5Δ32 mutation exhibit slower disease progression (120). In a phase 1/2 study in which HIV-infected patients received autologous CD4+ T cells transduced with a CCR5-targeting ZFN adenoviral construct, only 10 to 27% of the reinfused cells harbored the modified ccr5 gene, which was insufficient to control viral rebound during treatment interruption (121). SB-728-T and its derivatives are adenoviral CCR5-targeted ZFN constructs that are in clinical studies focused on CCR5 editing in CD4 T cells (NCT02388594, phase 1/2; NCT02225665, phase 1/2) or hematopoietic stem cells (NCT02500849, phase 1) (121). Additionally, a 5-patient study (NCT03164135) is investigating the safety and feasibility of allogeneic transplantation of CRISPR/Cas9-modified CCR5Δ32 CD34+ cells into HIV-positive individuals with hematological malignancies.
Effective delivery of gene editing machinery and homozygous gene disruption are the challenges of a CCR5 gene editing approach, with only a small number of cells delivered in the absence of bone marrow suppression. Even if these challenges are overcome, this strategy does not take CXCR4-tropic (X4) HIV quasispecies into account; while the vast majority of transmitted founder (TF) viruses that establish infection utilize CCR5 for entry and CCR5-tropic (R5) HIV dominates early stages of the disease (122), the virus may evolve to utilize CXCR4 (X4) in response to selective pressures such as availability of specific target cells (123, 124). This coreceptor switching (R5 to X4) occurs in approximately 50% of individuals chronically infected with HIV (124). It is important to note that in both the Berlin and London cases, CXCR4-tropic virus was not identified as a component of the patient’s reservoir, which was likely a crucial factor in achieving HIV remission. In contrast, the “Essen patient,” who received HSCT from a CCR5Δ32 donor following treatment for T-cell lymphoma, experienced rapid rebound of CXCR4-tropic virus that was present before transplantation (14). Additionally, HIV can be transmitted by cell-to-cell contact in the absence of CD4 and CCR5, as is the case for infection of astrocytes in the brain by HIV-infected lymphocytes (125). It is therefore unlikely that CCR5 editing will be an effective strategy to eliminate HIV in chronically infected individuals with a stable reservoir unless it is paired with a highly efficient ablative therapy and/or is employed only after thorough screening to ensure the absence of CXCR4-utilizing HIV isolates. Alternatively, reactivation of the viral reservoir using LRAs in combination with transplantation of HIV-resistant immune cells could eventually lead to HIV eradication; however, the presence of reactivation-resistant proviruses and inefficient killing observed in “kick-and-kill” approaches to date suggest that further optimization may be required. In terms of preventative therapy, an ethical debate surrounds the possibility of using gene editing techniques at the time of fertilization to create humans with mutations in CCR5 who might be resistant to HIV infection. CCR5-targeting strategies hold promise for a subset of patients with CCR5-restricted reservoirs but are unlikely to be broadly applicable to the majority of people living with HIV.
(iii) HIV inactivation by indel mutation or excision.
One advantage of using gene editing strategies to remove integrated HIV proviruses is that the long terminal repeat (LTR) is present at both ends of the viral genome. Therefore, a single gRNA can target both LTRs, as shown in in vitro studies using CRISPR/Cas9 (126 – 134). This excises the full integrated provirus between the target sequences, though at a much lower frequency than silencing via insertions or deletions (indels) introduced by NHEJ (126, 127, 129). Employment of two different LTR-targeting gRNAs that recognize unique sequences in the HIV promoter significantly enhances Cas9 cleavage (128, 130, 132). There is a risk of viral escape with a single gRNA due to nonlethal NHEJ-induced mutations around the Cas9 cleavage site (130, 131). Sustained inhibition of viral replication requires dual or multiplex gRNAs targeted against essential conserved regions of the HIV genome (130).
The CRISPR/Cas9 system can also cleave HIV sequences within latently infected cell lines, suggesting that Cas9 may be effective even when target sequences are partially occluded by repressive chromatin remodeling (134, 135). Delivery of AAV vectors encoding anti-HIV Cas9 constructs into rodent models of HIV gave similar results, with occasional excision of the full provirus as well as indel formation and reduced susceptibility to new infection (135). Importantly, Cas9-mediated cleavage of HIV occurred in multiple tissues, including brain, heart, kidney, lung, spleen, and liver (135). One study reported complete eradication of the latent HIV reservoir in 2/7 (29%) humanized mice following administration of long-acting, slow-effective ART (LASER ART) and HIV-targeting AAV9-CRISPR/Cas9; no virus could be detected from lymphoid tissues, bone marrow, blood, or brain, as measured by nested PCR, digital droplet PCR, and RNAscope, and no viral rebound occurred after treatment was removed in these two animals (136). A decrease in HIV DNA was observed in the remaining 5 animals, indicating reduction in the size of blood and tissue reservoirs. In contrast, viral rebound occurred in all animals who received LASER ART or CRISPR/Cas9 treatment alone, highlighting the importance of combination therapy. There are currently no ongoing clinical trials utilizing HIV-targeted CRISPR/Cas9 constructs.
CRISPR/Cas9 technology is an incredibly valuable tool with broad applications that range from HIV research to onco-therapeutics and treatment of genetic disease. Since current methods have been insufficient to completely clear virus in animal models (135 – 137), there is a need for ways to optimize delivery of anti-HIV Cas9 constructs to all infected cells, as well as strategies to enhance the frequency of lethal indels and/or provirus excision. There is concern about the potential immunogenicity of Cas9, given its bacterial origin, when the protein is stably expressed (138). To address this, a Tat-inducible Cas9 construct has been devised that is expressed only during active HIV replication to minimize off-target effects (139). The synergy between HIV-targeting CRISPR/Cas9 constructs and slow-release ART to eradicate virus in a subset of animals (136) provides strong proof of concept for this approach. Nevertheless, the efficiency of Cas9-mediated HIV excision is likely still insufficient to remove all integrated provirus from all reservoirs in all subjects at this stage. As this technology continues to improve over time, it may be necessary to develop other therapeutic strategies to promote HIV remission in order to fill the gaps left by current ART (the “block-and-lock” strategy).
Tat antagonists.
The HIV trans-activator of transcription (Tat) is an HIV accessory protein that plays pivotal roles in HIV replication and pathogenesis. Tat is an early viral protein that binds to the transactivation response (TAR) element, an RNA structure that is found in the 5′ region of all HIV mRNA and recruits the positive transcription elongation factor (P-TEFb) to the HIV promoter (140). Tat-mediated transactivation of the HIV LTR enhances HIV transcription and is essential for productive infection (141 – 143). Intracellular Tat levels regulate the cycle of active HIV transcription and entry into latency (144). Furthermore, Tat can be released from infected cells and taken up by uninfected bystanders (145). Tat has also been shown to be directly neurotoxic (146 – 148) and to stimulate production of reactive oxygen species (ROS) (149) and proinflammatory cytokines and chemokines (150 – 155).
In vitro modeling has predicted that the disruption of Tat-TAR binding can generate stable HIV latency (156). Since current antiretroviral therapies do not target the early viral products such as Tat, experimental strategies to develop Tat antagonists are being considered. Our laboratory has developed a cell-based assay for high-throughput screening of compounds for identifying Tat antagonists that block LTR transactivation. Many putative Tat antagonists have been identified by different high-throughput screening techniques, including several candidates that have been used in phase 1/2a clinical trials. Since several of these antagonists have been reviewed extensively elsewhere (157), this review will focus on recently discovered compounds that show significant promise. A summary of known Tat antagonists and their stages of development is included in Table 4.
TABLE 4.
Putative Tat antagonists that significantly reduce HIV transcriptiona
Compound(s) | Infection stage(s) tested | Cell type(s) | Screen | Development status | Reference(s) |
---|---|---|---|---|---|
VRX496 (Lexgenleucel-T) | Acute, chronic | CD4+ T cells, CD34+ hematopoietic stem cells | NA | Phase 2 | 121, 171, 174 – 178 |
Rev-TD-anti-TAR | Acute, chronic | CD4+ T cells | NA | Phase 1/2 | 172, 173 |
dCA | Acute, chronic | HeLa-CD4, T-cell lines, PBMCs, primary CD4+ T cells | NA | Preclinical | 158 – 160, 164 |
Durhamycin A | Acute | HeLa-CD4, T-cell lines | LTR-reporter | Preclinical | 179 |
WM5 | Acute, chronic | HeLa, T-cell lines, PBMCs | SAR | Preclinical | 180 – 183 |
NM13 | Acute | T-cell line | SAR | Preclinical | 184, 185 |
HM13N | Acute, chronic, latent | T-cell lines, monocytic cell lines, PBMCs | SAR | Preclinical | 186 |
Temacrazine | Acute, chronic, latent | T-cell line, monocytic cell lines | NA | Preclinical | 187 |
NeoR | Acute, chronic | T-cell line, promonocytic cell line, PBMCs | SAR | Preclinical | 188 |
3-(4-Chlorophenyl)-5-methyl-N-(3-pyridinyl-methyl)pyrazolo[1,5-a]pyrimidin-7-amine (compound 791), N-{[2-(2-hydroxybenzoyl)hydrazine]carbonothiol}-4-biphenylcarboxamide (compound 833), 2-hydroxy-N′-(3-hydroxy-4-methoxybenzylidene)benzohydrazide (compound 892) | Acute, chronic, latent | T-cell lines, CEM-GXR, PBMCs, HeLa HIVrtTAΔMls (189) | SMN2 minigene reporter | Preclinical | 168 |
Based on data from reference 157. Abbreviations: NA, not available; SAR, structure-activity relationship; PBMCs, peripheral blood mononuclear cells.
One of these drugs, didehydro-cortistatin A (dCA), specifically binds to Tat in the TAR binding region to inhibit transactivation of the HIV LTR (158). dCA substantially delays and reduces rebound in ex vivo cultures of CD4+ T cells from HIV-infected individuals even in the presence of strong stimuli such as CD3/CD28 ligation, phytohemagglutinin (PHA), and the latency-reversing agent prostratin (159, 160). dCA also induces rapid repressive chromatin remodeling of the HIV promoter that is maintained even when treatment is removed (159). Similar results have been obtained using dCA against SIV in vitro and ex vivo, which suggests that this drug is appropriate for studies in nonhuman primates (161). One of the major challenges in HIV therapy is residual low-level viremia or transient reactivation events (“blips”) that can be detected only by highly sensitive assays and persist despite ART (6, 8, 162, 163). These findings suggest that it may be possible to completely silence the HIV provirus over time by blocking Tat interaction with TAR; Tat antagonists may induce a perpetual latent state without residual viremia to achieve a functional cure via a “block-and-lock” mechanism (159). Such a strategy is especially attractive for controlling viral replication in the CNS reservoir, where delivery of gene editing therapy may be suboptimal or a more conventional “kick-and-kill” approach may lead to undesirable inflammation and neural injury. Importantly, dCA also inhibits Tat uptake and production of proinflammatory cytokines from Tat-transfected astrocytes (164). However, dCA does not bind to the cysteine-rich region of Tat, which contributes to Tat-mediated neurotoxicity through association with the N-methyl-d-aspartate (NMDA) receptor (165); therefore, it is currently unknown whether dCA can directly block Tat-mediated neurotoxicity. In vitro evidence indicates that treatment with dCA can induce mutations in the HIV promoter that increase basal transcription and reduce dependence on Tat transactivation via selective pressure; however, these mutated proviruses are less likely to enter latency and are sensitive to virus-induced cell death and immune-mediated clearance (166).
Given that Tat has a variety of functions and interacts with many cellular proteins (167), identifying compounds that selectively block Tat-TAR or Tat-PTEFb interactions may be insufficient unless the inhibitor completely shuts down proviral transcription from all reservoirs. To block other Tat functions that contribute to HIV pathogenesis, it may be necessary to identify therapeutics that directly degrade Tat protein or target Tat production at the posttranscriptional level. Recently three RNA splicing modulators, 3-(4-chlorophenyl)-5-methyl-N-(3-pyridinyl-methyl)pyrazolo[1,5-a]pyrimidin-7-amine (compound 791), N-{[2-(2-hydroxybenzoyl)hydrazine]carbonothiol}-4-biphenylcarboxamide (compound 833), and 2-hydroxy-N′-(3-hydroxy-4-methoxybenzylidene)benzohydrazide (compound 892), which reduced HIV viral output by 80 to 90% in low-micromolar ranges in vitro were identified (168). All three compounds diminished levels of HIV Gag, Env, Rev, and Tat proteins and decreased relative amounts of unspliced and singly spliced HIV transcripts. Compound 833 and compound 892 had limited off-target effects on exon inclusion of some genomic transcripts and global protein synthesis, while compound 791 significantly decreased viral replication and Tat protein production without notable off-target effects (168).
Another strategy is the use of molecules that are antisense to Tat. Several groups have used antisense oligonucleotides (ASOs) complementary to HIV sequences to either block translation or degrade viral transcripts. Recently, a 937-base antisense gene targeting HIV env under the control of an HIV promoter was incorporated into a lentiviral vector (VRX496) and transduced into CD34+ hematopoietic stem cells and autologous ex vivo-expanded CD4+ T cells. These modified T cells stably expressed the antisense gene and have been safely delivered to 65 HIV-infected subjects at six institutions to date without significant adverse events (169, 170). One study on a subset of patients who underwent ART treatment interruption reported a decrease in viral load set points in 6/8 (75%) patients who received VRX496, as well as enrichment of A→G mutations in HIV sequences at the antisense target region (171). Since tat and env have overlapping mRNA sequences, VRX496 may also inhibit Tat production. VRX496 is currently in phase 2 clinical trials (NCT00131560, NCT00295477, and NCT00622232) (Table 4).
A retroviral Rev-TD-anti-TAR construct, which expresses both a dominant-negative Rev construct and antisense TAR RNA (172), was evaluated in a phase 1/2 pilot study, but the results have not been published (NCT00001535) (Table 4) (173). Since all HIV transcripts contain the TAR sequence and Tat-TAR interaction is required for transactivation of the HIV promoter, it is likely that Rev-TD-anti-TAR would also act on Tat function and generation of tat mRNA. However, no ASOs that specifically target Tat transcripts alone have entered clinical trials.
It is worth noting that Tat antagonists, like many other potential eradication/remission strategies, are best used as combination therapy to accomplish their goal. While the fact that Tat inhibition alone in the presence of combination ART can induce additional repressive epigenetic changes to the HIV LTR and delay/prevent viral rebound in murine models (159) lends proof of concept to this strategy, further work must be done in vitro and in vivo to determine the feasibility of generating true remission via a permanently silenced (“locked”) HIV promoter.
CONCLUSION
After decades of intense research, HIV remains a chronic infection, and the number of individuals living with HIV continues to rise every year. Recent advances in gene therapy and immune therapies have provided renewed hope that a sterilizing HIV cure might be possible. Some preclinical studies for refinement of these approaches to ensure delivery to all the reservoirs and prevention of off-target effects are still necessary. Identification of pharmacological approaches that block early viral transcripts provides hope for a functional cure by providing long-term silencing of HIV. Further safety studies and means for delivery to all tissue reservoirs are still needed before clinical trials can be undertaken. A common feature in each of these approaches is the challenge posed by the CNS reservoirs due to the blood-brain barrier and the cell types infected in the brain that are different than the lymphoid reservoirs. Further, the brain cannot sustain prolonged inflammation due to bystander damage which can result in permanent neuronal injury. Nonetheless, development of immune checkpoint molecule inhibitors and other strategies that combat immune exhaustion/dysfunction, especially in combination with novel antiretroviral therapies, complementary immune therapies, vaccine work, gene editing of viral and/or host genomes, and novel anti-Tat molecules, may also provide better long-term health outcomes for HIV-infected individuals and avenues toward the goal of achieving a sterile cure.
SEARCH STRATEGY
We searched PubMed, international HIV meeting abstracts, and www.clinicaltrials.gov for HIV trial data up to and including July 2019. We also used multiple spellings, truncated nomenclatures, and abbreviations as search terms. We reviewed articles published in English resulting from these searches and the most relevant references cited in these articles.
ACKNOWLEDGMENTS
This work was supported by funding from the Office of AIDS Research, National Institutes of Health, to L.J.H. and by intramural funds from the Institute of Neurological Disorders and Stroke to A.N.
We are investigators on the pembrolizumab-based clinical trials NCT03367754 and NCT03239899 described in this work. There are no other interests to disclose.
L.J.H. and L.B.R. performed literature searches and wrote and edited the manuscript. J.A.K. provided a substantial editorial review as well as conceptual interpretation. A.N. provided conceptual support as well as the design of the work, with a substantial contribution to the revision of the final draft. J.A.K. and A.N. edited the manuscript and provided expertise and feedback.
REFERENCES
- 1.Walmsley SL, Antela A, Clumeck N, Duiculescu D, Eberhard A, Gutierrez F, Hocqueloux L, Maggiolo F, Sandkovsky U, Granier C, Pappa K, Wynne B, Min S, Nichols G. 2013. Dolutegravir plus abacavir-lamivudine for the treatment of HIV-1 infection. N Engl J Med 369:1807–1818. doi: 10.1056/NEJMoa1215541. [DOI] [PubMed] [Google Scholar]
- 2.Heaton RK, Franklin DR Jr, Deutsch R, Letendre S, Ellis RJ, Casaletto K, Marquine MJ, Woods SP, Vaida F, Atkinson JH, Marcotte TD, McCutchan JA, Collier AC, Marra CM, Clifford DB, Gelman BB, Sacktor N, Morgello S, Simpson DM, Abramson I, Gamst AC, Fennema-Notestine C, Smith DM, Grant I, Group C. 2015. Neurocognitive change in the era of HIV combination antiretroviral therapy: the longitudinal CHARTER study. Clin Infect Dis 60:473–480. doi: 10.1093/cid/ciu862. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Elmer-Dewitt P. 30 December 1996. Time man of the year: Dr. David Ho. Time Magazine [Google Scholar]
- 4.Ho DD, Neumann AU, Perelson AS, Chen W, Leonard JM, Markowitz M. 1995. Rapid turnover of plasma virions and CD4 lymphocytes in HIV-1 infection. Nature 373:123–126. doi: 10.1038/373123a0. [DOI] [PubMed] [Google Scholar]
- 5.Sullivan A. 10 November 1996. When plagues end. New York Times Magazine. [Google Scholar]
- 6.Chun TW, Stuyver L, Mizell SB, Ehler LA, Mican JA, Baseler M, Lloyd AL, Nowak MA, Fauci AS. 1997. Presence of an inducible HIV-1 latent reservoir during highly active antiretroviral therapy. Proc Natl Acad Sci U S A 94:13193–13197. doi: 10.1073/pnas.94.24.13193. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Finzi D, Blankson J, Siliciano JD, Margolick JB, Chadwick K, Pierson T, Smith K, Lisziewicz J, Lori F, Flexner C, Quinn TC, Chaisson RE, Rosenberg E, Walker B, Gange S, Gallant J, Siliciano RF. 1999. Latent infection of CD4+ T cells provides a mechanism for lifelong persistence of HIV-1, even in patients on effective combination therapy. Nat Med 5:512–517. doi: 10.1038/8394. [DOI] [PubMed] [Google Scholar]
- 8.Wong JK, Hezareh M, Gunthard HF, Havlir DV, Ignacio CC, Spina CA, Richman DD. 1997. Recovery of replication-competent HIV despite prolonged suppression of plasma viremia. Science 278:1291–1295. doi: 10.1126/science.278.5341.1291. [DOI] [PubMed] [Google Scholar]
- 9.Hutter G, Nowak D, Mossner M, Ganepola S, Mussig A, Allers K, Schneider T, Hofmann J, Kucherer C, Blau O, Blau IW, Hofmann WK, Thiel E. 2009. Long-term control of HIV by CCR5 Delta32/Delta32 stem-cell transplantation. N Engl J Med 360:692–698. doi: 10.1056/NEJMoa0802905. [DOI] [PubMed] [Google Scholar]
- 10.Henrich TJ, Hanhauser E, Marty FM, Sirignano MN, Keating S, Lee TH, Robles YP, Davis BT, Li JZ, Heisey A, Hill AL, Busch MP, Armand P, Soiffer RJ, Altfeld M, Kuritzkes DR. 2014. Antiretroviral-free HIV-1 remission and viral rebound after allogeneic stem cell transplantation: report of 2 cases. Ann Intern Med 161:319–327. doi: 10.7326/M14-1027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Smoleń-Dzirba J, Rosińska M, Janiec J, Beniowski M, Cycoń M, Bratosiewicz-Wąsik J, Wąsik TJ. 2017. HIV-1 infection in persons homozygous for CCR5-Delta32 allele: the next case and the review. AIDS Rev 19:219–230. [PubMed] [Google Scholar]
- 12.Duarte RF, Salgado M, Sánchez-Ortega I, Arnan M, Canals C, Domingo-Domenech E, Fernández-de-Sevilla A, González-Barca E, Morón-López S, Nogues N, Patiño B, Puertas MC, Clotet B, Petz LD, Querol S, Martinez-Picado J. 2015. CCR5 Delta32 homozygous cord blood allogeneic transplantation in a patient with HIV: a case report. Lancet HIV 2:e236–e242. doi: 10.1016/S2352-3018(15)00083-1. [DOI] [PubMed] [Google Scholar]
- 13.Rothenberger M, Wagner JE, Haase A, Richman D, Grzywacz B, Strain M, Lada S, Estes J, Fletcher CV, Podany AT, Anderson J, Schmidt T, Wietgrefe S, Schacker T, Verneris MR. 2018. Transplantation of CCR532 homozygous umbilical cord blood in a child with acute lymphoblastic leukemia and perinatally acquired HIV infection. Open Forum Infect Dis 5:ofy090. doi: 10.1093/ofid/ofy090. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Verheyen J, Thielen A, Lubke N, Dirks M, Widera M, Dittmer U, Kordales L, Daumer M, Jong TCM, Wensing AMJ, Kaiser R, Nijhuis M, Esser S. 2018. Rapid rebound of a preexisting CXCR4-tropic HIV variant after allogeneic transplantation with CCR5 delta32 homozygous stem cells. Clin Infect Dis doi: 10.1093/cid/ciy565. [DOI] [PubMed] [Google Scholar]
- 15.Luzuriaga K, Gay H, Ziemniak C, Sanborn KB, Somasundaran M, Rainwater-Lovett K, Mellors JW, Rosenbloom D, Persaud D. 2015. Viremic relapse after HIV-1 remission in a perinatally infected child. N Engl J Med 372:786–788. doi: 10.1056/NEJMc1413931. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Persaud D, Gay H, Ziemniak C, Chen YH, Piatak M Jr, Chun TW, Strain M, Richman D, Luzuriaga K. 2013. Absence of detectable HIV-1 viremia after treatment cessation in an infant. N Engl J Med 369:1828–1835. doi: 10.1056/NEJMoa1302976. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Gupta RK, Abdul-Jawad S, McCoy LE, Mok HP, Peppa D, Salgado M, Martinez-Picado J, Nijhuis M, Wensing AMJ, Lee H, Grant P, Nastouli E, Lambert J, Pace M, Salasc F, Monit C, Innes AJ, Muir L, Waters L, Frater J, Lever A, Edwards SG, Gabriel IH, Olavarria E. 2019. HIV-1 remission following CCR5Delta32/Delta32 haematopoietic stem-cell transplantation. Nature 568:244–248. doi: 10.1038/s41586-019-1027-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Avettand-Fenoel V, Hocqueloux L, Ghosn J, Cheret A, Frange P, Melard A, Viard JP, Rouzioux C. 2016. Total HIV-1 DNA, a marker of viral reservoir dynamics with clinical implications. Clin Microbiol Rev 29:859–880. doi: 10.1128/CMR.00015-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Blankson JN, Persaud D, Siliciano RF. 2002. The challenge of viral reservoirs in HIV-1 infection. Annu Rev Med 53:557–593. doi: 10.1146/annurev.med.53.082901.104024. [DOI] [PubMed] [Google Scholar]
- 20.Eisele E, Siliciano RF. 2012. Redefining the viral reservoirs that prevent HIV-1 eradication. Immunity 37:377–388. doi: 10.1016/j.immuni.2012.08.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Brown A, Zhang H, Lopez P, Pardo CA, Gartner S. 2006. In vitro modeling of the HIV-macrophage reservoir. J Leukoc Biol 80:1127–1135. doi: 10.1189/jlb.0206126. [DOI] [PubMed] [Google Scholar]
- 22.Gartner S, Markovits P, Markovitz DM, Kaplan MH, Gallo RC, Popovic M. 1986. The role of mononuclear phagocytes in HTLV-III/LAV infection. Science 233:215–219. doi: 10.1126/science.3014648. [DOI] [PubMed] [Google Scholar]
- 23.Barton K, Hiener B, Winckelmann A, Rasmussen TA, Shao W, Byth K, Lanfear R, Solomon A, McMahon J, Harrington S, Buzon M, Lichterfeld M, Denton PW, Olesen R, Ostergaard L, Tolstrup M, Lewin SR, Sogaard OS, Palmer S. 2016. Broad activation of latent HIV-1 in vivo. Nat Commun 7:12731. doi: 10.1038/ncomms12731. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Kandathil AJ, Sugawara S, Balagopal A. 2016. Are T cells the only HIV-1 reservoir? Retrovirology 13:86. doi: 10.1186/s12977-016-0323-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Kulpa DA, Chomont N. 2015. HIV persistence in the setting of antiretroviral therapy: when, where and how does HIV hide? J Virus Erad 1:59–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Mzingwane ML, Tiemessen CT. 2017. Mechanisms of HIV persistence in HIV reservoirs. Rev Med Virol 27:e1924. doi: 10.1002/rmv.1924. [DOI] [PubMed] [Google Scholar]
- 27.Stevenson M. 2015. Role of myeloid cells in HIV-1-host interplay. J Neurovirol 21:242–248. doi: 10.1007/s13365-014-0281-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Wong JK, Yukl SA. 2016. Tissue reservoirs of HIV. Curr Opin HIV AIDS 11:362–370. doi: 10.1097/COH.0000000000000293. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Kelly J, Beddall MH, Yu D, Iyer SR, Marsh JW, Wu Y. 2008. Human macrophages support persistent transcription from unintegrated HIV-1 DNA. Virology 372:300–312. doi: 10.1016/j.virol.2007.11.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Calantone N, Wu F, Klase Z, Deleage C, Perkins M, Matsuda K, Thompson EA, Ortiz AM, Vinton CL, Ourmanov I, Lore K, Douek DC, Estes JD, Hirsch VM, Brenchley JM. 2014. Tissue myeloid cells in SIV-infected primates acquire viral DNA through phagocytosis of infected T cells. Immunity 41:493–502. doi: 10.1016/j.immuni.2014.08.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.DiNapoli SR, Ortiz AM, Wu F, Matsuda K, Twigg HL III, Hirsch VM, Knox K, Brenchley JM. 2017. Tissue-resident macrophages can contain replication-competent virus in antiretroviral-naive, SIV-infected Asian macaques. JCI Insight 2:e91214. doi: 10.1172/jci.insight.91214. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Baxter AE, Russell RA, Duncan CJ, Moore MD, Willberg CB, Pablos JL, Finzi A, Kaufmann DE, Ochsenbauer C, Kappes JC, Groot F, Sattentau QJ. 2014. Macrophage infection via selective capture of HIV-1-infected CD4+ T cells. Cell Host Microbe 16:711–721. doi: 10.1016/j.chom.2014.10.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Honeycutt JB, Thayer WO, Baker CE, Ribeiro RM, Lada SM, Cao Y, Cleary RA, Hudgens MG, Richman DD, Garcia JV. 2017. HIV persistence in tissue macrophages of humanized myeloid-only mice during antiretroviral therapy. Nat Med 23:638–643. doi: 10.1038/nm.4319. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Ganor Y, Real F, Sennepin A, Dutertre CA, Prevedel L, Xu L, Tudor D, Charmeteau B, Couedel-Courteille A, Marion S, Zenak AR, Jourdain JP, Zhou Z, Schmitt A, Capron C, Eugenin EA, Cheynier R, Revol M, Cristofari S, Hosmalin A, Bomsel M. 2019. HIV-1 reservoirs in urethral macrophages of patients under suppressive antiretroviral therapy. Nat Microbiol 4:633–644. doi: 10.1038/s41564-018-0335-z. [DOI] [PubMed] [Google Scholar]
- 35.Mitchell BI, Laws EI, Ndhlovu LC. 2019. Impact of myeloid reservoirs in HIV cure trials. Curr HIV/AIDS Rep 16:129–140. doi: 10.1007/s11904-019-00438-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Carter CA, Ehrlich LS. 2008. Cell biology of HIV-1 infection of macrophages. Annu Rev Microbiol 62:425–443. doi: 10.1146/annurev.micro.62.081307.162758. [DOI] [PubMed] [Google Scholar]
- 37.Imamichi H, Dewar RL, Adelsberger JW, Rehm CA, O'Doherty U, Paxinos EE, Fauci AS, Lane HC. 2016. Defective HIV-1 proviruses produce novel protein-coding RNA species in HIV-infected patients on combination antiretroviral therapy. Proc Natl Acad Sci U S A 113:8783–8788. doi: 10.1073/pnas.1609057113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Garcia-Montojo M, Doucet-O'Hare T, Henderson L, Nath A. 2018. Human endogenous retrovirus-K (HML-2): a comprehensive review. Crit Rev Microbiol 44:715–738. doi: 10.1080/1040841X.2018.1501345. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Dahl V, Gisslen M, Hagberg L, Peterson J, Shao W, Spudich S, Price RW, Palmer S. 2014. An example of genetically distinct HIV type 1 variants in cerebrospinal fluid and plasma during suppressive therapy. J Infect Dis 209:1618–1622. doi: 10.1093/infdis/jit805. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Bachani M, Sacktor N, McArthur JC, Nath A, Rumbaugh J. 2013. Detection of anti-tat antibodies in CSF of individuals with HIV-associated neurocognitive disorders. J Neurovirol 19:82–88. doi: 10.1007/s13365-012-0144-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Burbelo PD, Bayat A, Rhodes CS, Hoh R, Martin JN, Fromentin R, Chomont N, Hutter G, Kovacs JA, Deeks SG. 2014. HIV antibody characterization as a method to quantify reservoir size during curative interventions. J Infect Dis 209:1613–1617. doi: 10.1093/infdis/jit667. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Burbelo PD, Price RW, Hagberg L, Hatano H, Spudich S, Deeks SG, Gisslen M. 2018. Anti-human immunodeficiency virus antibodies in the cerebrospinal fluid: evidence of early treatment impact on central nervous system reservoir? J Infect Dis 217:1024–1032. doi: 10.1093/infdis/jix662. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Dahl V, Peterson J, Fuchs D, Gisslen M, Palmer S, Price RW. 2014. Low levels of HIV-1 RNA detected in the cerebrospinal fluid after up to 10 years of suppressive therapy are associated with local immune activation. AIDS 28:2251–2258. doi: 10.1097/QAD.0000000000000400. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Bavaro DF, Calamo A, Lepore L, Fabrizio C, Saracino A, Angarano G, Monno L. 2019. Cerebrospinal fluid compartmentalization of HIV-1 and correlation with plasma viral load and blood-brain barrier damage. Infection 47:441–446. doi: 10.1007/s15010-019-01268-8. [DOI] [PubMed] [Google Scholar]
- 45.Joseph SB, Kincer LP, Bowman NM, Evans C, Vinikoor MJ, Lippincott CK, Gisslen M, Spudich S, Menezes P, Robertson K, Archin N, Kashuba A, Eron JJ, Price RW, Swanstrom R. 2018. HIV-1 RNA detected in the CNS after years of suppressive antiretroviral therapy can originate from a replicating CNS reservoir or clonally expanded cells. Clin Infect Dis doi: 10.1093/cid/ciy1066. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Williams DW, Veenstra M, Gaskill PJ, Morgello S, Calderon TM, Berman JW. 2014. Monocytes mediate HIV neuropathogenesis: mechanisms that contribute to HIV associated neurocognitive disorders. Curr HIV Res 12:85–96. doi: 10.2174/1570162X12666140526114526. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Canto-Nogues C, Sanchez-Ramon S, Alvarez S, Lacruz C, Munoz-Fernandez MA. 2005. HIV-1 infection of neurons might account for progressive HIV-1-associated encephalopathy in children. J Mol Neurosci 27:79–89. doi: 10.1385/JMN:27:1:079. [DOI] [PubMed] [Google Scholar]
- 48.Gama L, Abreu C, Shirk EN, Queen SE, Beck SE, Metcalf Pate KA, Bullock BT, Zink MC, Mankowski JL, Clements JE. 2018. SIV latency in macrophages in the CNS. Curr Top Microbiol Immunol 417:111–130. doi: 10.1007/82_2018_89. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Avalos CR, Abreu CM, Queen SE, Li M, Price S, Shirk EN, Engle EL, Forsyth E, Bullock BT, Mac Gabhann F, Wietgrefe SW, Haase AT, Zink MC, Mankowski JL, Clements JE, Gama L. 2017. Brain macrophages in simian immunodeficiency virus-infected, antiretroviral-suppressed macaques: a functional latent reservoir. mBio 8:e01186-17. doi: 10.1128/mBio.01186-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Gama L, Abreu CM, Shirk EN, Price SL, Li M, Laird GM, Pate KAM, Wietgrefe SW, O'Connor SL, Pianowski L, Haase AT, Van Lint C, Siliciano RF, Clements JE. 2017. Reactivation of simian immunodeficiency virus reservoirs in the brain of virally suppressed macaques. AIDS 31:5–14. doi: 10.1097/QAD.0000000000001267. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Harrington PR, Schnell G, Letendre SL, Ritola K, Robertson K, Hall C, Burch CL, Jabara CB, Moore DT, Ellis RJ, Price RW, Swanstrom R. 2009. Cross-sectional characterization of HIV-1 env compartmentalization in cerebrospinal fluid over the full disease course. AIDS 23:907–915. doi: 10.1097/QAD.0b013e3283299129. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Eden A, Nilsson S, Hagberg L, Fuchs D, Zetterberg H, Svennerholm B, Gisslen M. 2016. Asymptomatic cerebrospinal fluid HIV-1 viral blips and viral escape during antiretroviral therapy: a longitudinal study. J Infect Dis 214:1822–1825. doi: 10.1093/infdis/jiw454. [DOI] [PubMed] [Google Scholar]
- 53.Henderson LJ, Johnson TP, Smith B, Bowen LR, Santamaria UA, Bachani M, Demarino C, Barclay RA, Snow J, Sacktor N, McArthur JC, Letendre S, Steiner J, Kashanchi F, Nath A. Presence of Tat and trans-activation response (TAR) element in spinal fluid despite antiretroviral therapy. AIDS, in press. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Delagreverie HM, Delaugerre C, Lewin SR, Deeks SG, Li JZ. 2016. Ongoing clinical trials of human immunodeficiency virus latency-reversing and immunomodulatory agents. Open Forum Infect Dis 3:ofw189. doi: 10.1093/ofid/ofw189. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Thorlund K, Horwitz MS, Fife BT, Lester R, Cameron DW. 2017. Landscape review of current HIV ‘kick and kill’ cure research—some kicking, not enough killing. BMC Infect Dis 17:595. doi: 10.1186/s12879-017-2683-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Zerbato JM, Purves HV, Lewin SR, Rasmussen TA. 2019. Between a shock and a hard place: challenges and developments in HIV latency reversal. Curr Opin Virol 38:1–9. doi: 10.1016/j.coviro.2019.03.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Pearson R, Kim YK, Hokello J, Lassen K, Friedman J, Tyagi M, Karn J. 2008. Epigenetic silencing of human immunodeficiency virus (HIV) transcription by formation of restrictive chromatin structures at the viral long terminal repeat drives the progressive entry of HIV into latency. J Virol 82:12291–12303. doi: 10.1128/JVI.01383-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Van Lint C, Bouchat S, Marcello A. 2013. HIV-1 transcription and latency: an update. Retrovirology 10:67. doi: 10.1186/1742-4690-10-67. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Deeks SG. 2012. HIV: shock and kill. Nature 487:439–440. doi: 10.1038/487439a. [DOI] [PubMed] [Google Scholar]
- 60.Hamer DH. 2004. Can HIV be cured? Mechanisms of HIV persistence and strategies to combat it. Curr HIV Res 2:99–111. doi: 10.2174/1570162043484915. [DOI] [PubMed] [Google Scholar]
- 61.Borducchi EN, Cabral C, Stephenson KE, Liu J, Abbink P, Ng'ang'a D, Nkolola JP, Brinkman AL, Peter L, Lee BC, Jimenez J, Jetton D, Mondesir J, Mojta S, Chandrashekar A, Molloy K, Alter G, Gerold JM, Hill AL, Lewis MG, Pau MG, Schuitemaker H, Hesselgesser J, Geleziunas R, Kim JH, Robb ML, Michael NL, Barouch DH. 2016. Ad26/MVA therapeutic vaccination with TLR7 stimulation in SIV-infected rhesus monkeys. Nature 540:284–287. doi: 10.1038/nature20583. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Borducchi EN, Liu J, Nkolola JP, Cadena AM, Yu WH, Fischinger S, Broge T, Abbink P, Mercado NB, Chandrashekar A, Jetton D, Peter L, McMahan K, Moseley ET, Bekerman E, Hesselgesser J, Li W, Lewis MG, Alter G, Geleziunas R, Barouch DH. 2018. Antibody and TLR7 agonist delay viral rebound in SHIV-infected monkeys. Nature 563:360–364. doi: 10.1038/s41586-018-0600-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Jones RB, Mueller S, O'Connor R, Rimpel K, Sloan DD, Karel D, Wong HC, Jeng EK, Thomas AS, Whitney JB, Lim SY, Kovacs C, Benko E, Karandish S, Huang SH, Buzon MJ, Lichterfeld M, Irrinki A, Murry JP, Tsai A, Yu H, Geleziunas R, Trocha A, Ostrowski MA, Irvine DJ, Walker BD. 2016. A subset of latency-reversing agents expose HIV-infected resting CD4+ T-cells to recognition by cytotoxic T-lymphocytes. PLoS Pathog 12:e1005545. doi: 10.1371/journal.ppat.1005545. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Azzoni L, Foulkes AS, Papasavvas E, Mexas AM, Lynn KM, Mounzer K, Tebas P, Jacobson JM, Frank I, Busch MP, Deeks SG, Carrington M, O'Doherty U, Kostman J, Montaner LJ. 2013. Pegylated Interferon alfa-2a monotherapy results in suppression of HIV type 1 replication and decreased cell-associated HIV DNA integration. J Infect Dis 207:213–222. doi: 10.1093/infdis/jis663. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Shan L, Deng K, Shroff NS, Durand CM, Rabi SA, Yang HC, Zhang H, Margolick JB, Blankson JN, Siliciano RF. 2012. Stimulation of HIV-1-specific cytolytic T lymphocytes facilitates elimination of latent viral reservoir after virus reactivation. Immunity 36:491–501. doi: 10.1016/j.immuni.2012.01.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Jones RB, O'Connor R, Mueller S, Foley M, Szeto GL, Karel D, Lichterfeld M, Kovacs C, Ostrowski MA, Trocha A, Irvine DJ, Walker BD. 2014. Histone deacetylase inhibitors impair the elimination of HIV-infected cells by cytotoxic T-lymphocytes. PLoS Pathog 10:e1004287. doi: 10.1371/journal.ppat.1004287. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Boyer Z, Palmer S. 2018. Targeting immune checkpoint molecules to eliminate latent HIV. Front Immunol 9:2339. doi: 10.3389/fimmu.2018.02339. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Banga R, Procopio FA, Noto A, Pollakis G, Cavassini M, Ohmiti K, Corpataux JM, de Leval L, Pantaleo G, Perreau M. 2016. PD-1(+) and follicular helper T cells are responsible for persistent HIV-1 transcription in treated aviremic individuals. Nat Med 22:754–761. doi: 10.1038/nm.4113. [DOI] [PubMed] [Google Scholar]
- 69.Fromentin R, Bakeman W, Lawani MB, Khoury G, Hartogensis W, DaFonseca S, Killian M, Epling L, Hoh R, Sinclair E, Hecht FM, Bacchetti P, Deeks SG, Lewin SR, Sekaly RP, Chomont N. 2016. CD4+ T cells expressing PD-1, TIGIT and LAG-3 contribute to HIV persistence during ART. PLoS Pathog 12:e1005761. doi: 10.1371/journal.ppat.1005761. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.McGary CS, Deleage C, Harper J, Micci L, Ribeiro SP, Paganini S, Kuri-Cervantes L, Benne C, Ryan ES, Balderas R, Jean S, Easley K, Marconi V, Silvestri G, Estes JD, Sekaly RP, Paiardini M. 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. doi: 10.1016/j.immuni.2017.09.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Kassu A, Marcus RA, D’Souza MB, Kelly-McKnight EA, Golden-Mason L, Akkina R, Fontenot AP, Wilson CC, Palmer BE. 2010. Regulation of virus-specific CD4+ T cell function by multiple costimulatory receptors during chronic HIV infection. J Immunol 185:3007–3018. doi: 10.4049/jimmunol.1000156. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Salisch NC, Kaufmann DE, Awad AS, Reeves RK, Tighe DP, Li Y, Piatak M Jr, Lifson JD, Evans DT, Pereyra F, Freeman GJ, Johnson RP. 2010. Inhibitory TCR coreceptor PD-1 is a sensitive indicator of low-level replication of SIV and HIV-1. J Immunol 184:476–487. doi: 10.4049/jimmunol.0902781. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Chew GM, Fujita T, Webb GM, Burwitz BJ, Wu HL, Reed JS, Hammond KB, Clayton KL, Ishii N, Abdel-Mohsen M, Liegler T, Mitchell BI, Hecht FM, Ostrowski M, Shikuma CM, Hansen SG, Maurer M, Korman AJ, Deeks SG, Sacha JB, Ndhlovu LC. 2016. TIGIT marks exhausted T Cells, correlates with disease progression, and serves as a target for immune restoration in HIV and SIV infection. PLoS Pathog 12:e1005349. doi: 10.1371/journal.ppat.1005349. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Guihot A, Marcelin AG, Massiani MA, Samri A, Soulie C, Autran B, Spano JP. 2018. Drastic decrease of the HIV reservoir in a patient treated with nivolumab for lung cancer. Ann Oncol 29:517–518. doi: 10.1093/annonc/mdx696. [DOI] [PubMed] [Google Scholar]
- 75.Hoffmann M, Pantazis N, Martin GE, Hickling S, Hurst J, Meyerowitz J, Willberg CB, Robinson N, Brown H, Fisher M, Kinloch S, Babiker A, Weber J, Nwokolo N, Fox J, Fidler S, Phillips R, Frater J, SPARTAC and CHERUB Investigators. 2016. Exhaustion of activated CD8 T cells predicts disease progression in primary HIV-1 infection. PLoS Pathog 12:e1005661. doi: 10.1371/journal.ppat.1005661. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Hurst J, Hoffmann M, Pace M, Williams JP, Thornhill J, Hamlyn E, Meyerowitz J, Willberg C, Koelsch KK, Robinson N, Brown H, Fisher M, Kinloch S, Cooper DA, Schechter M, Tambussi G, Fidler S, Babiker A, Weber J, Kelleher AD, Phillips RE, Frater J. 2015. Immunological biomarkers predict HIV-1 viral rebound after treatment interruption. Nat Commun 6:8495. doi: 10.1038/ncomms9495. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Le Garff G, Samri A, Lambert-Niclot S, Even S, Lavole A, Cadranel J, Spano JP, Autran B, Marcelin AG, Guihot A. 2017. Transient HIV-specific T cells increase and inflammation in an HIV-infected patient treated with nivolumab. AIDS 31:1048–1051. doi: 10.1097/QAD.0000000000001429. [DOI] [PubMed] [Google Scholar]
- 78.Cortese I, Muranski P, Enose-Akahata Y, Ha SK, Smith B, Monaco M, Ryschkewitsch C, Major EO, Ohayon J, Schindler MK, Beck E, Reoma LB, Jacobson S, Reich DS, Nath A. 2019. Pembrolizumab treatment for progressive multifocal leukoencephalopathy. N Engl J Med 380:1597–1605. doi: 10.1056/NEJMoa1815039. [DOI] [PubMed] [Google Scholar]
- 79.Gardiner D, Lalezari J, Lawitz E, DiMicco M, Ghalib R, Reddy KR, Chang KM, Sulkowski M, Marro SO, Anderson J, He B, Kansra V, McPhee F, Wind-Rotolo M, Grasela D, Selby M, Korman AJ, Lowy I. 2013. A randomized, double-blind, placebo-controlled assessment of BMS-936558, a fully human monoclonal antibody to programmed death-1 (PD-1), in patients with chronic hepatitis C virus infection. PLoS One 8:e63818. doi: 10.1371/journal.pone.0063818. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Khoja L, Butler MO, Kang SP, Ebbinghaus S, Joshua AM. 2015. Pembrolizumab. J Immunother Cancer 3:36. doi: 10.1186/s40425-015-0078-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Mylvaganam GH, Chea LS, Tharp GK, Hicks S, Velu V, Iyer SS, Deleage C, Estes JD, Bosinger SE, Freeman GJ, Ahmed R, Amara RR. 2018. Combination anti-PD-1 and antiretroviral therapy provides therapeutic benefit against SIV. JCI Insight 3:122940. doi: 10.1172/jci.insight.122940. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Evans VA, van der Sluis RM, Solomon A, Dantanarayana A, McNeil C, Garsia R, Palmer S, Fromentin R, Chomont N, Sekaly RP, Cameron PU, Lewin SR. 2018. Programmed cell death-1 contributes to the establishment and maintenance of HIV-1 latency. AIDS 32:1491–1497. doi: 10.1097/QAD.0000000000001849. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Perreau M, Savoye AL, De Crignis E, Corpataux JM, Cubas R, Haddad EK, De Leval L, Graziosi C, Pantaleo G. 2013. Follicular helper T cells serve as the major CD4 T cell compartment for HIV-1 infection, replication, and production. J Exp Med 210:143–156. doi: 10.1084/jem.20121932. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Velu V, Titanji K, Zhu B, Husain S, Pladevega A, Lai L, Vanderford TH, Chennareddi L, Silvestri G, Freeman GJ, Ahmed R, Amara RR. 2009. Enhancing SIV-specific immunity in vivo by PD-1 blockade. Nature 458:206–210. doi: 10.1038/nature07662. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Yang J, Riella LV, Chock S, Liu T, Zhao X, Yuan X, Paterson AM, Watanabe T, Vanguri V, Yagita H, Azuma M, Blazar BR, Freeman GJ, Rodig SJ, Sharpe AH, Chandraker A, Sayegh MH. 2011. The novel costimulatory programmed death ligand 1/B7.1 pathway is functional in inhibiting alloimmune responses in vivo. J Immunol 187:1113–1119. doi: 10.4049/jimmunol.1100056. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Gay F, D'Agostino M, Giaccone L, Genuardi M, Festuccia M, Boccadoro M, Bruno B. 2017. Immuno-oncologic approaches: CAR-T cells and checkpoint inhibitors. Clin Lymphoma Myeloma Leuk 17:471–478. doi: 10.1016/j.clml.2017.06.014. [DOI] [PubMed] [Google Scholar]
- 87.Gay CL, Bosch RJ, Ritz J, Hataye JM, Aga E, Tressler RL, Mason SW, Hwang CK, Grasela DM, Ray N, Cyktor JC, Coffin JM, Acosta EP, Koup RA, Mellors JW, Eron JJ, AIDS Clinical Trials 5326 Study Team. 2017. Clinical trial of the anti-PD-L1 antibody BMS-936559 in HIV-1 infected participants on suppressive antiretroviral therapy. J Infect Dis 215:1725–1733. doi: 10.1093/infdis/jix191. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Kaufmann DE, Walker BD. 2009. PD-1 and CTLA-4 inhibitory cosignaling pathways in HIV infection and the potential for therapeutic intervention. J Immunol 182:5891–5897. doi: 10.4049/jimmunol.0803771. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Leng Q, Bentwich Z, Borkow G. 2006. Increased TGF-beta, Cbl-b and CTLA-4 levels and immunosuppression in association with chronic immune activation. Int Immunol 18:637–644. doi: 10.1093/intimm/dxh375. [DOI] [PubMed] [Google Scholar]
- 90.Wightman F, Solomon A, Kumar SS, Urriola N, Gallagher K, Hiener B, Palmer S, McNeil C, Garsia R, Lewin SR. 2015. Effect of ipilimumab on the HIV reservoir in an HIV-infected individual with metastatic melanoma. AIDS 29:504–506. doi: 10.1097/QAD.0000000000000562. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Burke MM, Kluger HM, Golden M, Heller KN, Hoos A, Sznol M. 2011. Case report: response to ipilimumab in a patient with HIV with metastatic melanoma. J Clin Oncol 29:e792. doi: 10.1200/JCO.2011.36.9199. [DOI] [PubMed] [Google Scholar]
- 92.Hu CC, Jeng WJ, Chen YC, Fang JH, Huang CH, Teng W, Hsieh YC, Lin YC, Chien RN, Sheen IS, Lin CY. 2017. Memory regulatory T cells increase only in inflammatory phase of chronic hepatitis B infection and related to galectin-9/Tim-3 interaction. Sci Rep 7:15280. doi: 10.1038/s41598-017-15527-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Hu XH, Tang MX, Mor G, Liao AH. 2016. Tim-3: expression on immune cells and roles at the maternal-fetal interface. J Reprod Immunol 118:92–99. doi: 10.1016/j.jri.2016.10.113. [DOI] [PubMed] [Google Scholar]
- 94.Merani S, Chen W, Elahi S. 2015. The bitter side of sweet: the role of galectin-9 in immunopathogenesis of viral infections. Rev Med Virol 25:175–186. doi: 10.1002/rmv.1832. [DOI] [PubMed] [Google Scholar]
- 95.Clayton KL, Douglas-Vail MB, Nur-Ur Rahman AK, Medcalf KE, Xie IY, Chew GM, Tandon R, Lanteri MC, Norris PJ, Deeks SG, Ndhlovu LC, Ostrowski MA. 2015. Soluble T cell immunoglobulin mucin domain 3 is shed from CD8+ T cells by the sheddase ADAM10, is increased in plasma during untreated HIV infection, and correlates with HIV disease progression. J Virol 89:3723–3736. doi: 10.1128/JVI.00006-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Jones RB, Ndhlovu LC, Barbour JD, Sheth PM, Jha AR, Long BR, Wong JC, Satkunarajah M, Schweneker M, Chapman JM, Gyenes G, Vali B, Hyrcza MD, Yue FY, Kovacs C, Sassi A, Loutfy M, Halpenny R, Persad D, Spotts G, Hecht FM, Chun TW, McCune JM, Kaul R, Rini JM, Nixon DF, Ostrowski MA. 2008. Tim-3 expression defines a novel population of dysfunctional T cells with highly elevated frequencies in progressive HIV-1 infection. J Exp Med 205:2763–2779. doi: 10.1084/jem.20081398. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Sakhdari A, Mujib S, Vali B, Yue FY, MacParland S, Clayton K, Jones RB, Liu J, Lee EY, Benko E, Kovacs C, Gommerman J, Kaul R, Ostrowski MA. 2012. Tim-3 negatively regulates cytotoxicity in exhausted CD8+ T cells in HIV infection. PLoS One 7:e40146. doi: 10.1371/journal.pone.0040146. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Schwartz JA, Clayton KL, Mujib S, Zhang H, Rahman AK, Liu J, Yue FY, Benko E, Kovacs C, Ostrowski MA. 2017. Tim-3 is a marker of plasmacytoid dendritic cell dysfunction during HIV infection and is associated with the recruitment of IRF7 and p85 into lysosomes and with the submembrane displacement of TLR9. J Immunol 198:3181–3194. doi: 10.4049/jimmunol.1601298. [DOI] [PubMed] [Google Scholar]
- 99.Elahi S, Niki T, Hirashima M, Horton H. 2012. Galectin-9 binding to Tim-3 renders activated human CD4+ T cells less susceptible to HIV-1 infection. Blood 119:4192–4204. doi: 10.1182/blood-2011-11-389585. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Bi S, Hong PW, Lee B, Baum LG. 2011. Galectin-9 binding to cell surface protein disulfide isomerase regulates the redox environment to enhance T-cell migration and HIV entry. Proc Natl Acad Sci U S A 108:10650–10655. doi: 10.1073/pnas.1017954108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Yu X, Harden K, Gonzalez LC, Francesco M, Chiang E, Irving B, Tom I, Ivelja S, Refino CJ, Clark H, Eaton D, Grogan JL. 2009. The surface protein TIGIT suppresses T cell activation by promoting the generation of mature immunoregulatory dendritic cells. Nat Immunol 10:48–57. doi: 10.1038/ni.1674. [DOI] [PubMed] [Google Scholar]
- 102.Pena J, Jones NG, Bousheri S, Bangsberg DR, Cao H. 2014. Lymphocyte activation gene-3 expression defines a discrete subset of HIV-specific CD8+ T cells that is associated with lower viral load. AIDS Res Hum Retroviruses 30:535–541. doi: 10.1089/aid.2012.0195. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Zhou G, Noordam L, Sprengers D, Doukas M, Boor PPC, van Beek AA, Erkens R, Mancham S, Grunhagen D, Menon AG, Lange JF, Burger P, Brandt A, Galjart B, Verhoef C, Kwekkeboom J, Bruno MJ. 2018. Blockade of LAG3 enhances responses of tumor-infiltrating T cells in mismatch repair-proficient liver metastases of colorectal cancer. Oncoimmunology 7:e1448332. doi: 10.1080/2162402X.2018.1448332. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Ma Q, Liu J, Wu G, Teng M, Wang S, Cui M, Li Y. 2018. Co-expression of LAG3 and TIM3 identifies a potent Treg population that suppresses macrophage functions in colorectal cancer patients. Clin Exp Pharmacol Physiol 45:1002. doi: 10.1111/1440-1681.12992. [DOI] [PubMed] [Google Scholar]
- 105.Vigano S, Banga R, Bellanger F, Pellaton C, Farina A, Comte D, Harari A, Perreau M. 2014. CD160-associated CD8 T-cell functional impairment is independent of PD-1 expression. PLoS Pathog 10:e1004380. doi: 10.1371/journal.ppat.1004380. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Ahmad F, Shankar EM, Yong YK, Tan HY, Ahrenstorf G, Jacobs R, Larsson M, Schmidt RE, Kamarulzaman A, Ansari AW. 2017. Negative checkpoint regulatory molecule 2B4 (CD244) upregulation is associated with invariant natural killer T cell alterations and human immunodeficiency virus disease progression. Front Immunol 8:338. doi: 10.3389/fimmu.2017.00338. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Yamamoto T, Price DA, Casazza JP, Ferrari G, Nason M, Chattopadhyay PK, Roederer M, Gostick E, Katsikis PD, Douek DC, Haubrich R, Petrovas C, Koup RA. 2011. Surface expression patterns of negative regulatory molecules identify determinants of virus-specific CD8+ T-cell exhaustion in HIV infection. Blood 117:4805–4815. doi: 10.1182/blood-2010-11-317297. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Chlewicki LK, Velikovsky CA, Balakrishnan V, Mariuzza RA, Kumar V. 2008. Molecular basis of the dual functions of 2B4 (CD244). J Immunol 180:8159–8167. doi: 10.4049/jimmunol.180.12.8159. [DOI] [PubMed] [Google Scholar]
- 109.Zhang Z, Xu X, Lu J, Zhang S, Gu L, Fu J, Jin L, Li H, Zhao M, Zhang J, Wu H, Su L, Fu YX, Wang FS. 2011. B and T lymphocyte attenuator down-regulation by HIV-1 depends on type I interferon and contributes to T-cell hyperactivation. J Infect Dis 203:1668–1678. doi: 10.1093/infdis/jir165. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Grabmeier-Pfistershammer K, Stecher C, Zettl M, Rosskopf S, Rieger A, Zlabinger GJ, Steinberger P. 2017. Antibodies targeting BTLA or TIM-3 enhance HIV-1 specific T cell responses in combination with PD-1 blockade. Clin Immunol 183:167–173. doi: 10.1016/j.clim.2017.09.002. [DOI] [PubMed] [Google Scholar]
- 111.Miselis NR, Linn D, Restaino C, Baral T, Xia J, Ueda R, Sawant A, Baker J, Raghunathan G, Wang X, Bowman E, Sukumar S. 2017. Antagonism of the co-inhibitory receptor BTLA enhances efficacy of anti-PD-1 treatment in murine syngeneic tumor models. J Cancer Res 77(Suppl 13):577. doi: 10.1158/1538-7445.AM2017-577. [DOI] [Google Scholar]
- 112.Pascutti MF, Geerman S, Slot E, van Gisbergen KP, Boon L, Arens R, van Lier RA, Wolkers MC, Nolte MA. 2015. Enhanced CD8 T cell responses through GITR-mediated costimulation resolve chronic viral infection. PLoS Pathog 11:e1004675. doi: 10.1371/journal.ppat.1004675. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Larsson M, Shankar EM, Che KF, Saeidi A, Ellegard R, Barathan M, Velu V, Kamarulzaman A. 2013. Molecular signatures of T-cell inhibition in HIV-1 infection. Retrovirology 10:31. doi: 10.1186/1742-4690-10-31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Whittall T, Peters B, Rahman D, Kingsley CI, Vaughan R, Lehner T. 2011. Immunogenic and tolerogenic signatures in human immunodeficiency virus (HIV)-infected controllers compared with progressors and a conversion strategy of virus control. Clin Exp Immunol 166:208–217. doi: 10.1111/j.1365-2249.2011.04463.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Boch J, Scholze H, Schornack S, Landgraf A, Hahn S, Kay S, Lahaye T, Nickstadt A, Bonas U. 2009. Breaking the code of DNA binding specificity of TAL-type III effectors. Science 326:1509–1512. doi: 10.1126/science.1178811. [DOI] [PubMed] [Google Scholar]
- 116.Moscou MJ, Bogdanove AJ. 2009. A simple cipher governs DNA recognition by TAL effectors. Science 326:1501. doi: 10.1126/science.1178817. [DOI] [PubMed] [Google Scholar]
- 117.Makarova KS, Zhang F, Koonin EV. 2017. SnapShot: class 2 CRISPR-Cas systems. Cell 168:328–328. doi: 10.1016/j.cell.2016.12.038. [DOI] [PubMed] [Google Scholar]
- 118.Wiedenheft B, Sternberg SH, Doudna JA. 2012. RNA-guided genetic silencing systems in bacteria and archaea. Nature 482:331–338. doi: 10.1038/nature10886. [DOI] [PubMed] [Google Scholar]
- 119.Allers K, Hutter G, Hofmann J, Loddenkemper C, Rieger K, Thiel E, Schneider T. 2011. Evidence for the cure of HIV infection by CCR5Delta32/Delta32 stem cell transplantation. Blood 117:2791–2799. doi: 10.1182/blood-2010-09-309591. [DOI] [PubMed] [Google Scholar]
- 120.Ruiz-Mateos E, Tarancon-Diez L, Alvarez-Rios AI, Dominguez-Molina B, Genebat M, Pulido I, Abad MA, Muñoz-Fernandez MA, Leal M. 2018. Association of heterozygous CCR5Delta32 deletion with survival in HIV-infection: a cohort study. Antiviral Res 150:15–19. doi: 10.1016/j.antiviral.2017.12.002. [DOI] [PubMed] [Google Scholar]
- 121.Tebas P, Stein D, Tang WW, Frank I, Wang SQ, Lee G, Spratt SK, Surosky RT, Giedlin MA, Nichol G, Holmes MC, Gregory PD, Ando DG, Kalos M, Collman RG, Binder-Scholl G, Plesa G, Hwang WT, Levine BL, June CH. 2014. Gene editing of CCR5 in autologous CD4 T cells of persons infected with HIV. N Engl J Med 370:901–910. doi: 10.1056/NEJMoa1300662. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Keele BF, Giorgi EE, Salazar-Gonzalez JF, Decker JM, Pham KT, Salazar MG, Sun C, Grayson T, Wang S, Li H, Wei X, Jiang C, Kirchherr JL, Gao F, Anderson JA, Ping LH, Swanstrom R, Tomaras GD, Blattner WA, Goepfert PA, Kilby JM, Saag MS, Delwart EL, Busch MP, Cohen MS, Montefiori DC, Haynes BF, Gaschen B, Athreya GS, Lee HY, Wood N, Seoighe C, Perelson AS, Bhattacharya T, Korber BT, Hahn BH, Shaw GM. 2008. Identification and characterization of transmitted and early founder virus envelopes in primary HIV-1 infection. Proc Natl Acad Sci U S A 105:7552–7557. doi: 10.1073/pnas.0802203105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Joseph SB, Swanstrom R. 2018. The evolution of HIV-1 entry phenotypes as a guide to changing target cells. J Leukoc Biol 103:421–431. doi: 10.1002/JLB.2RI0517-200R. [DOI] [PubMed] [Google Scholar]
- 124.Kamp C. 2009. Understanding the HIV coreceptor switch from a dynamical perspective. BMC Evol Biol 9:274. doi: 10.1186/1471-2148-9-274. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Li GH, Anderson C, Jaeger L, Do T, Major EO, Nath A. 2015. Cell-to-cell contact facilitates HIV transmission from lymphocytes to astrocytes via CXCR4. AIDS 29:755–766. doi: 10.1097/QAD.0000000000000605. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Ebina H, Misawa N, Kanemura Y, Koyanagi Y. 2013. Harnessing the CRISPR/Cas9 system to disrupt latent HIV-1 provirus. Sci Rep 3:2510. doi: 10.1038/srep02510. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Hu W, Kaminski R, Yang F, Zhang Y, Cosentino L, Li F, Luo B, Alvarez-Carbonell D, Garcia-Mesa Y, Karn J, Mo X, Khalili K. 2014. RNA-directed gene editing specifically eradicates latent and prevents new HIV-1 infection. Proc Natl Acad Sci U S A 111:11461–11466. doi: 10.1073/pnas.1405186111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Lebbink RJ, de Jong DC, Wolters F, Kruse EM, van Ham PM, Wiertz EJ, Nijhuis M. 2017. A combinational CRISPR/Cas9 gene-editing approach can halt HIV replication and prevent viral escape. Sci Rep 7:41968. doi: 10.1038/srep41968. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Liao HK, Gu Y, Diaz A, Marlett J, Takahashi Y, Li M, Suzuki K, Xu R, Hishida T, Chang CJ, Esteban CR, Young J, Izpisua Belmonte JC. 2015. Use of the CRISPR/Cas9 system as an intracellular defense against HIV-1 infection in human cells. Nat Commun 6:6413. doi: 10.1038/ncomms7413. [DOI] [PubMed] [Google Scholar]
- 130.Wang G, Zhao N, Berkhout B, Das AT. 2016. A combinatorial CRISPR-Cas9 attack on HIV-1 DNA extinguishes all infectious provirus in infected T cell cultures. Cell Rep 17:2819–2826. doi: 10.1016/j.celrep.2016.11.057. [DOI] [PubMed] [Google Scholar]
- 131.Wang G, Zhao N, Berkhout B, Das AT. 2016. CRISPR-Cas9 can inhibit HIV-1 replication but NHEJ repair facilitates virus escape. Mol Ther 24:522–526. doi: 10.1038/mt.2016.24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Yin C, Zhang T, Li F, Yang F, Putatunda R, Young WB, Khalili K, Hu W, Zhang Y. 2016. Functional screening of guide RNAs targeting the regulatory and structural HIV-1 viral genome for a cure of AIDS. AIDS 30:1163–1174. doi: 10.1097/QAD.0000000000001079. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Yoder KE, Bundschuh R. 2016. Host double strand break repair generates HIV-1 strains resistant to CRISPR/Cas9. Sci Rep 6:29530. doi: 10.1038/srep29530. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Zhu W, Lei R, Le Duff Y, Li J, Guo F, Wainberg MA, Liang C. 2015. The CRISPR/Cas9 system inactivates latent HIV-1 proviral DNA. Retrovirology 12:22. doi: 10.1186/s12977-015-0150-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Kaminski R, Chen Y, Fischer T, Tedaldi E, Napoli A, Zhang Y, Karn J, Hu W, Khalili K. 2016. Elimination of HIV-1 genomes from human T-lymphoid cells by CRISPR/Cas9 gene editing. Sci Rep 6:22555. doi: 10.1038/srep22555. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Dash PK, Kaminski R, Bella R, Su H, Mathews S, Ahooyi TM, Chen C, Mancuso P, Sariyer R, Ferrante P, Donadoni M, Robinson JA, Sillman B, Lin Z, Hilaire JR, Banoub M, Elango M, Gautam N, Mosley RL, Poluektova LY, McMillan J, Bade AN, Gorantla S, Sariyer IK, Burdo TH, Young WB, Amini S, Gordon J, Jacobson JM, Edagwa B, Khalili K, Gendelman HE. 2019. Sequential LASER ART and CRISPR treatments Eliminate hIV-1 in a subset of infected humanized mice. Nat Commun 10:2753. doi: 10.1038/s41467-019-10366-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Yin C, Zhang T, Qu X, Zhang Y, Putatunda R, Xiao X, Li F, Xiao W, Zhao H, Dai S, Qin X, Mo X, Young WB, Khalili K, Hu W. 2017. In vivo excision of HIV-1 provirus by saCas9 and multiplex single-guide RNAs in animal models. Mol Ther 25:1168–1186. doi: 10.1016/j.ymthe.2017.03.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Chew WL. 2018. Immunity to CRISPR Cas9 and Cas12a therapeutics. Wiley Interdiscip Rev Syst Biol Med 10:e1408. doi: 10.1002/wsbm.1408. [DOI] [PubMed] [Google Scholar]
- 139.Kaminski R, Chen Y, Salkind J, Bella R, Young WB, Ferrante P, Karn J, Malcolm T, Hu W, Khalili K. 2016. Negative feedback regulation of HIV-1 by gene editing strategy. Sci Rep 6:31527. doi: 10.1038/srep31527. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Garber ME, Wei P, Jones KA. 1998. HIV-1 Tat interacts with cyclin T1 to direct the P-TEFb CTD kinase complex to TAR RNA. Cold Spring Harbor Symp Quant Biol 63:371–380. doi: 10.1101/sqb.1998.63.371. [DOI] [PubMed] [Google Scholar]
- 141.Berkhout B, Silverman RH, Jeang KT. 1989. Tat trans-activates the human immunodeficiency virus through a nascent RNA target. Cell 59:273–282. doi: 10.1016/0092-8674(89)90289-4. [DOI] [PubMed] [Google Scholar]
- 142.Dingwall C, Ernberg I, Gait MJ, Green SM, Heaphy S, Karn J, Lowe AD, Singh M, Skinner MA, Valerio R. 1989. Human immunodeficiency virus 1 tat protein binds trans-activation-responsive region (TAR) RNA in vitro. Proc Natl Acad Sci U S A 86:6925–6929. doi: 10.1073/pnas.86.18.6925. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Jones KA. 1989. HIV trans-activation and transcription control mechanisms. New Biol 1:127–135. [PubMed] [Google Scholar]
- 144.Robinson R. 2007. A simple feedback resistor switch keeps latent HIV from awakening. PLoS Biol 5:e25. doi: 10.1371/journal.pbio.0050025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Rayne F, Debaisieux S, Yezid H, Lin YL, Mettling C, Konate K, Chazal N, Arold ST, Pugniere M, Sanchez F, Bonhoure A, Briant L, Loret E, Roy C, Beaumelle B. 2010. Phosphatidylinositol-(4,5)-bisphosphate enables efficient secretion of HIV-1 Tat by infected T-cells. EMBO J 29:1348–1362. doi: 10.1038/emboj.2010.32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Eugenin EA, King JE, Nath A, Calderon TM, Zukin RS, Bennett MV, Berman JW. 2007. HIV-tat induces formation of an LRP-PSD-95- NMDAR-nNOS complex that promotes apoptosis in neurons and astrocytes. Proc Natl Acad Sci U S A 104:3438–3443. doi: 10.1073/pnas.0611699104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Li W, Li G, Steiner J, Nath A. 2009. Role of Tat protein in HIV neuropathogenesis. Neurotox Res 16:205–220. doi: 10.1007/s12640-009-9047-8. [DOI] [PubMed] [Google Scholar]
- 148.Magnuson DS, Knudsen BE, Geiger JD, Brownstone RM, Nath A. 1995. Human immunodeficiency virus type 1 tat activates non-N-methyl-d-aspartate excitatory amino acid receptors and causes neurotoxicity. Ann Neurol 37:373–380. doi: 10.1002/ana.410370314. [DOI] [PubMed] [Google Scholar]
- 149.Couret J, Chang TL. 2016. Reactive oxygen species in HIV infection. EC Microbiol 3:597–604. [PMC free article] [PubMed] [Google Scholar]
- 150.Ambrosino C, Ruocco MR, Chen X, Mallardo M, Baudi F, Trematerra S, Quinto I, Venuta S, Scala G. 1997. HIV-1 Tat induces the expression of the interleukin-6 (IL6) gene by binding to the IL6 leader RNA and by interacting with CAAT enhancer-binding protein beta (NF-IL6) transcription factors. J Biol Chem 272:14883–14892. doi: 10.1074/jbc.272.23.14883. [DOI] [PubMed] [Google Scholar]
- 151.Buscemi L, Ramonet D, Geiger JD. 2007. Human immunodeficiency virus type-1 protein Tat induces tumor necrosis factor-alpha-mediated neurotoxicity. Neurobiol Dis 26:661–670. doi: 10.1016/j.nbd.2007.03.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Chen P, Mayne M, Power C, Nath A. 1997. The Tat protein of HIV-1 induces tumor necrosis factor-alpha production. Implications for HIV-1-associated neurological diseases. J Biol Chem 272:22385–22388. doi: 10.1074/jbc.272.36.22385. [DOI] [PubMed] [Google Scholar]
- 153.Johnson TP, Patel K, Johnson KR, Maric D, Calabresi PA, Hasbun R, Nath A. 2013. Induction of IL-17 and nonclassical T-cell activation by HIV-Tat protein. Proc Natl Acad Sci U S A 110:13588–13593. doi: 10.1073/pnas.1308673110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Nath A, Conant K, Chen P, Scott C, Major EO. 1999. Transient exposure to HIV-1 Tat protein results in cytokine production in macrophages and astrocytes. A hit and run phenomenon. J Biol Chem 274:17098–17102. doi: 10.1074/jbc.274.24.17098. [DOI] [PubMed] [Google Scholar]
- 155.Zidovetzki R, Wang JL, Chen P, Jeyaseelan R, Hofman F. 1998. Human immunodeficiency virus Tat protein induces interleukin 6 mRNA expression in human brain endothelial cells via protein kinase C- and cAMP-dependent protein kinase pathways. AIDS Res Hum Retroviruses 14:825–833. doi: 10.1089/aid.1998.14.825. [DOI] [PubMed] [Google Scholar]
- 156.Cao Y, Lei X, Ribeiro RM, Perelson AS, Liang J. 2018. Probabilistic control of HIV latency and transactivation by the Tat gene circuit. Proc Natl Acad Sci U S A 115:12453–12458. doi: 10.1073/pnas.1811195115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Mousseau G, Valente S. 2012. Strategies to block HIV transcription: focus on small molecule Tat inhibitors. Biology (Basel) 1:668–697. doi: 10.3390/biology1030668. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Mousseau G, Clementz MA, Bakeman WN, Nagarsheth N, Cameron M, Shi J, Baran P, Fromentin R, Chomont N, Valente ST. 2012. An analog of the natural steroidal alkaloid cortistatin A potently suppresses Tat-dependent HIV transcription. Cell Host Microbe 12:97–108. doi: 10.1016/j.chom.2012.05.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Kessing CF, Nixon CC, Li C, Tsai P, Takata H, Mousseau G, Ho PT, Honeycutt JB, Fallahi M, Trautmann L, Garcia JV, Valente ST. 2017. In vivo suppression of HIV rebound by didehydro-cortistatin A, a “block-and-lock” strategy for HIV-1 treatment. Cell Rep 21:600–611. doi: 10.1016/j.celrep.2017.09.080. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Mousseau G, Kessing CF, Fromentin R, Trautmann L, Chomont N, Valente ST. 2015. The Tat inhibitor didehydro-cortistatin A prevents HIV-1 reactivation from latency. mBio 6:e00465. doi: 10.1128/mBio.00465-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Mediouni S, Kessing CF, Jablonski JA, Thenin-Houssier S, Clementz M, Kovach MD, Mousseau G, de Vera IMS, Li C, Kojetin DJ, Evans DT, Valente ST. 2019. The Tat inhibitor didehydro-cortistatin A suppresses SIV replication and reactivation. FASEB J 33:8280–8293. doi: 10.1096/fj.201801165R. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Chomont N, El-Far M, Ancuta P, Trautmann L, Procopio FA, Yassine-Diab B, Boucher G, Boulassel M-R, Ghattas G, Brenchley JM, Schacker TW, Hill BJ, Douek DC, Routy J-P, Haddad EK, Sékaly R-P. 2009. HIV reservoir size and persistence are driven by T cell survival and homeostatic proliferation. Nat Med 15:893–900. doi: 10.1038/nm.1972. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Finzi D, Hermankova M, Pierson T, Carruth LM, Buck C, Chaisson RE, Quinn TC, Chadwick K, Margolick J, Brookmeyer R, Gallant J, Markowitz M, Ho DD, Richman DD, Siliciano RF. 1997. Identification of a reservoir for HIV-1 in patients on highly active antiretroviral therapy. Science 278:1295–1300. doi: 10.1126/science.278.5341.1295. [DOI] [PubMed] [Google Scholar]
- 164.Mediouni S, Jablonski J, Paris JJ, Clementz MA, Thenin-Houssier S, McLaughlin JP, Valente ST. 2015. Didehydro-cortistatin A inhibits HIV-1 Tat mediated neuroinflammation and prevents potentiation of cocaine reward in Tat transgenic mice. Curr HIV Res 13:64–79. doi: 10.2174/1570162X13666150121111548. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Li W, Huang Y, Reid R, Steiner J, Malpica-Llanos T, Darden TA, Shankar SK, Mahadevan A, Satishchandra P, Nath A. 2008. NMDA receptor activation by HIV-Tat protein is clade dependent. J Neurosci 28:12190–12198. doi: 10.1523/JNEUROSCI.3019-08.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Mousseau G, Aneja R, Clementz MA, Mediouni S, Lima NS, Haregot A, Kessing CF, Jablonski JA, Thenin-Houssier S, Nagarsheth N, Trautmann L, Valente ST. 2019. Resistance to the Tat inhibitor didehydro-cortistatin A is mediated by heightened basal HIV-1 transcription. mBio 10:e01750-18. doi: 10.1128/mBio.01750-18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Gautier VW, Gu L, O'Donoghue N, Pennington S, Sheehy N, Hall WW. 2009. In vitro nuclear interactome of the HIV-1 Tat protein. Retrovirology 6:47. doi: 10.1186/1742-4690-6-S2-P39. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Balachandran A, Wong R, Stoilov P, Pan S, Blencowe B, Cheung P, Harrigan PR, Cochrane A. 2017. Identification of small molecule modulators of HIV-1 Tat and Rev protein accumulation. Retrovirology 14:7. doi: 10.1186/s12977-017-0330-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169.Levine BL, Humeau LM, Boyer J, MacGregor RR, Rebello T, Lu X, Binder GK, Slepushkin V, Lemiale F, Mascola JR, Bushman FD, Dropulic B, June CH. 2006. Gene transfer in humans using a conditionally replicating lentiviral vector. Proc Natl Acad Sci U S A 103:17372–17377. doi: 10.1073/pnas.0608138103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.McGarrity GJ, Hoyah G, Winemiller A, Andre K, Stein D, Blick G, Greenberg RN, Kinder C, Zolopa A, Binder-Scholl G, Tebas P, June CH, Humeau LM, Rebello T. 2013. Patient monitoring and follow-up in lentiviral clinical trials. J Gene Med 15:78–82. doi: 10.1002/jgm.2691. [DOI] [PubMed] [Google Scholar]
- 171.Tebas P, Stein D, Binder-Scholl G, Mukherjee R, Brady T, Rebello T, Humeau L, Kalos M, Papasavvas E, Montaner LJ, Schullery D, Shaheen F, Brennan AL, Zheng Z, Cotte J, Slepushkin V, Veloso E, Mackley A, Hwang WT, Aberra F, Zhan J, Boyer J, Collman RG, Bushman FD, Levine BL, June CH. 2013. Antiviral effects of autologous CD4 T cells genetically modified with a conditionally replicating lentiviral vector expressing long antisense to HIV. Blood 121:1524–1533. doi: 10.1182/blood-2012-07-447250. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.VandenDriessche T, Chuah MK, Chiang L, Chang HK, Ensoli B, Morgan RA. 1995. Inhibition of clinical human immunodeficiency virus (HIV) type 1 isolates in primary CD4+ T lymphocytes by retroviral vectors expressing anti-HIV genes. J Virol 69:4045–4052. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Morgan RA, Walker R. 1996. Gene therapy for AIDS using retroviral mediated gene transfer to deliver HIV-1 antisense TAR and transdominant Rev protein genes to syngeneic lymphocytes in HIV-1 infected identical twins. Hum Gene Ther 7:1281–1306. doi: 10.1089/hum.1996.7.10-1281. [DOI] [PubMed] [Google Scholar]
- 174.Davis BM, Humeau L, Dropulic B. 2004. In vivo selection for human and murine hematopoietic cells transduced with a therapeutic MGMT lentiviral vector that inhibits HIV replication. Mol Ther 9:160–172. doi: 10.1016/j.ymthe.2003.11.003. [DOI] [PubMed] [Google Scholar]
- 175.Humeau LM, Binder GK, Lu X, Slepushkin V, Merling R, Echeagaray P, Pereira M, Slepushkina T, Barnett S, Dropulic LK, Carroll R, Levine BL, June CH, Dropulic B. 2004. Efficient lentiviral vector-mediated control of HIV-1 replication in CD4 lymphocytes from diverse HIV+ infected patients grouped according to CD4 count and viral load. Mol Ther 9:902–913. doi: 10.1016/j.ymthe.2004.03.005. [DOI] [PubMed] [Google Scholar]
- 176.Lu X, Humeau L, Slepushkin V, Binder G, Yu Q, Slepushkina T, Chen Z, Merling R, Davis B, Chang YN, Dropulic B. 2004. Safe two-plasmid production for the first clinical lentivirus vector that achieves >99% transduction in primary cells using a one-step protocol. J Gene Med 6:963–973. doi: 10.1002/jgm.593. [DOI] [PubMed] [Google Scholar]
- 177.Lu X, Yu Q, Binder GK, Chen Z, Slepushkina T, Rossi J, Dropulic B. 2004. Antisense-mediated inhibition of human immunodeficiency virus (HIV) replication by use of an HIV type 1-based vector results in severely attenuated mutants incapable of developing resistance. J Virol 78:7079–7088. doi: 10.1128/JVI.78.13.7079-7088.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178.MacGregor RR. 2001. Clinical protocol. A phase 1 open-label clinical trial of the safety and tolerability of single escalating doses of autologous CD4 T cells transduced with VRX496 in HIV-positive subjects. Hum Gene Ther 12:2028–2029. [PubMed] [Google Scholar]
- 179.Jayasuriya H, Lingham RB, Graham P, Quamina D, Herranz L, Genilloud O, Gagliardi M, Danzeisen R, Tomassini JE, Zink DL, Guan Z, Singh SB. 2002. Durhamycin A, a potent inhibitor of HIV Tat transactivation. J Nat Prod 65:1091–1095. doi: 10.1021/np010642f. [DOI] [PubMed] [Google Scholar]
- 180.Cecchetti V, Parolin C, Moro S, Pecere T, Filipponi E, Calistri A, Tabarrini O, Gatto B, Palumbo M, Fravolini A, Palu G. 2000. 6-Aminoquinolones as new potential anti-HIV agents. J Med Chem 43:3799–3802. doi: 10.1021/jm9903390. [DOI] [PubMed] [Google Scholar]
- 181.Parolin C, Gatto B, Del Vecchio C, Pecere T, Tramontano E, Cecchetti V, Fravolini A, Masiero S, Palumbo M, Palu G. 2003. New anti-human immunodeficiency virus type 1 6-aminoquinolones: mechanism of action. Antimicrob Agents Chemother 47:889–896. doi: 10.1128/aac.47.3.889-896.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182.Richter S, Parolin C, Gatto B, Del Vecchio C, Brocca-Cofano E, Fravolini A, Palu G, Palumbo M. 2004. Inhibition of human immunodeficiency virus type 1 tat-trans-activation-responsive region interaction by an antiviral quinolone derivative. Antimicrob Agents Chemother 48:1895–1899. doi: 10.1128/aac.48.5.1895-1899.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183.Tabarrini O, Stevens M, Cecchetti V, Sabatini S, Dell'Uomo M, Manfroni G, Palumbo M, Pannecouque C, De Clercq E, Fravolini A. 2004. Structure modifications of 6-aminoquinolones with potent anti-HIV activity. J Med Chem 47:5567–5578. doi: 10.1021/jm049721p. [DOI] [PubMed] [Google Scholar]
- 184.Sarli V, Giannis A. 2007. Selective inhibition of CBP/p300 HAT. Chem Biol 14:605–606. doi: 10.1016/j.chembiol.2007.06.001. [DOI] [PubMed] [Google Scholar]
- 185.Tabarrini O, Massari S, Sancineto L, Daelemans D, Sabatini S, Manfroni G, Cecchetti V, Pannecouque C. 2011. Structural investigation of the naphthyridone scaffold: identification of a 1,6-naphthyridone derivative with potent and selective anti-HIV activity. Chemmedchem 6:1249–1257. doi: 10.1002/cmdc.201100073. [DOI] [PubMed] [Google Scholar]
- 186.Massari S, Daelemans D, Barreca ML, Knezevich A, Sabatini S, Cecchetti V, Marcello A, Pannecouque C, Tabarrini O. 2010. A 1,8-naphthyridone derivative targets the HIV-1 Tat-mediated transcription and potently inhibits the HIV-1 replication. J Med Chem 53:641–648. doi: 10.1021/jm901211d. [DOI] [PubMed] [Google Scholar]
- 187.Turpin JA, Buckheit RW Jr, Derse D, Hollingshead M, Williamson K, Palamone C, Osterling MC, Hill SA, Graham L, Schaeffer CA, Bu M, Huang M, Cholody WM, Michejda CJ, Rice WG. 1998. Inhibition of acute-, latent-, and chronic-phase human immunodeficiency virus type 1 (HIV-1) replication by a bistriazoloacridone analog that selectively inhibits HIV-1 transcription. Antimicrob Agents Chemother 42:487–494. doi: 10.1128/AAC.42.3.487. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188.Litovchick A, Lapidot A, Eisenstein M, Kalinkovich A, Borkow G. 2001. Neomycin B-arginine conjugate, a novel HIV-1 Tat antagonist: synthesis and anti-HIV activities. Biochemistry 40:15612–15623. doi: 10.1021/bi0108655. [DOI] [PubMed] [Google Scholar]
- 189.Wong R, Balachandran A, Mao AY, Dobson W, Gray-Owen S, Cochrane A. 2011. Differential effect of CLK SR Kinases on HIV-1 gene expression: potential novel targets for therapy. Retrovirology 8:47. doi: 10.1186/1742-4690-8-47. [DOI] [PMC free article] [PubMed] [Google Scholar]