Abstract
Purpose of the review:
Despite decades of insights about how CD8+ T cells and NK cells contribute to natural control of infection, additional hurdles (mutational escape from cellular immunity, sequence diversity, and hard-to-access tissue reservoirs) will need to be overcome to develop a cure. In this review, we highlight recent findings of novel mechanisms of antiviral cellular immunity and discuss current strategies for therapeutic deisgn.
Recent findings:
Of note are the apparent converging roles of viral antigen-specific MHC-E-restricted CD8+ T cells and NK cells, IL-15 biologics to boost cytotoxicity, and broadly neutralizing antibodies in their native form or as anitbody fragments to neutralize virus and engage cellular immunity, respectively. Finally, renewed interest in myeloid cells as relevant viral reservoirs is an encouraging sign for designing inclusive therapeutic strategies.
Summary:
Several studies have shown promise in many pre-clinical models of disease, including SIV/SHIV infection in non-human primates and HIV infection in humanized mice. However, each model comes with its own limitations and may not fully predict human responses. We eagerly await the results of clinical trails assessing the efficacy of these strategies to achieve reductions in viral reservoirs, delay viral rebound, or ultimately elicit immune based control of infection without cART.
Keywords: Antigen-specific NK cells, CMV vaccine, HLA-E, IL-15, bNAbs, CAR T cells
INTRODUCTION
Defining the HIV reservoir – what can the immune system detect?
Understanding the nature of the reservoir is essential for designing strategies aimed at viral eradication to achieve immune-based control. The HIV reservoir is defined as any cell that can harbor virus, persist despite cART, and seed new infections. While CD4+ T cells constitute the majority of the HIV reservoir, HIV-infected macrophages and dendritic cells (DCs) are detected in tissues from virally suppressed PLWH (1–4)(* for 3 and 4), and macrophage-tropic viral sequences have been observed in PLWH following analytical treatment interruption (ATI) (5), emphasizing the importance of considering other cell subsets in addition to CD4+ T cells for HIV cure strategies. Latency (which has recently been described in human monocytes, the blood precursors for tissue-resident macrophages (6))* remains a major barrier for viral eradication. However, while there is no evidence for ongoing viral evolution during cART (7), residual viral RNA and protein expression during cART (3,4,8–10)(* for 8 and 10) may provide an opportunity for sustained immune detection and control of infection, even without the need for latency reversal. This review focuses on the challenges of eliminating productively infected cells and new strategies to control the virus.
CD8+ T cell-mediated control of infection
Many studies have shown the importance of CD8+ cytotoxic T lymphocytes (CTLs) in controlling HIV, initially by observing a drop in plasma viral load during acute stages of infection correlating with the expansion of HIV-specific CTLs (11,12). However, the selective pressure exerted by viral peptide-specific CTLs coupled with the downmodulation of MHC-I by HIV nef and vpu leads to the selection of cells infected with CTL-escaped virus, a major contributing factor to viral persistence (13). Additional evidence supporting the role of CTLs in controlling infection stems from individuals who naturally maintain a low (50–2000 HIV RNA copies/mL) or undetectable (defined by clinical assays <50 RNA copies/mL) viral load in the absence of cART, referred to as viremic controllers (VC) and elite controllers (EC), respectively. Numerous factors correlate with viral control in VC/ECs including protective HLA alleles (HLA-B*57 and HLA-B*27; reviewed in (11)), targeting of highly “networked” epitopes exhibiting little mutational tolerance (14), stable proliferative and cytolytic function of CTLs (15), and the ability of CTLs to respond to both lower concentrations of antigen, and a greater range of epitope variants (16). While loss of CD8+ T cell function in controllers has been shown to precede resurgence of viremia (17), no single factor likely determines initial protection, suggesting that control might require an optimal combination of both a highly functional immune response and targeting of “networked” epitopes. Complicating the role of CD8+ T cells in HIV control, it was recently observed that HIV-infected CD4+ T cells surviving co-culture with autologous CD8+ T cells show enrichment of genes associated with quiescence, suggesting that CD8+ T cells may play a role in promoting latency (18)*. With these challenges in mind, we will discuss strategies to improve CTL-mediated control of HIV.
NK cell contributions to control of acute and chronic infection
NK cells express a variety of germline-encoded activating and inhibitory receptors, whose ligands are differentially modulated on HIV-infected cells (19–25). Without the need for priming to elicit antiviral effects, they play a role in early stages of pathogenesis and potentially protection from viral acquisition. Cohort studies of highly exposed seronegative (HESN) individuals have identified particular NK cell phenotypes and enhanced effector functions associated with resistance to HIV acquisition (26–30). Furthermore, analysis of human samples prior to and then during acute HIV infection showed that increased NK cell activity was a correlate of future viremic control (31), and NK cell depletion in non-human primates (NHPs) enhanced acute viremia and inflammation (32), emphasizing the role of NK cells in control of early infection.
For chronic stages of viral infection, co-expression of KIR3DS1 and specific KIR3DL1 subtypes with HLA-B Bw4–80Ile is associated with lower plasma viremia and delayed progression to AIDS (33–35). For cART treated infection, Astorga-Gamaza et al recently showed that expression of the inhibitory receptor KLRG1 is associated with NK cell dysfunction, and that blocking KLRG1 enhances cellular function, promoting a reduction in intact HIV genomes (21)**. Humanized mouse models have yielded additional insights into the role of NK cells in chronic infection. Kim et al., showed that treatment of humanized mice with a latency reversal agent in combination with administration of human NK cells delayed the time to rebound following cART interruption (36). Furthemore, in elegant studies by Huot and colleagues, NK cells in african green monkeys (AGM), natural hosts for SIV that exhibit better viral control than rhesus macaques (RM), showed infiltration into the secondary lymphoid organs where they help control the infection in an MHC-E-restricted manner (37,38). Together, these studies emphasize the ability of NK cells to naturally control viral infection, which, in humans, may be enhanced through KLRG1 blockades and promoting NK cell infiltration into B cell follicles in the lymph nodes.
Increasing evidence supports the existence of specific NK cell subsets with “memory-like” or “antigen-specific” properties. Memory NK cells were first identified in mice in response to haptens (39), and have since been identified in humans (40). In the context of HIV, TCF7-dependent NK cells exhibiting memory-like markers are increased in PLWH (41). Strikingly, NK cells from SIV and SHIV-infected RMs show specific lysis of Gag- and Env-pulsed DCs ex vivo, which was dependent on NKG2A/C expression (42). These memory NK cell responses could also be elicited through Ad26 HIV Env or SIV Gag vaccination (42). A potential mechanism of the apparent anitgen specificity is via a conserved Gag epitope, which, when presented on HLA-E, is thought to block the interaction between HLA-E and the inhibitory receptor, NKG2A, resulting in increased NK-mediated killing (43). Recently, an exciting study by Jost and colleagues observed the presence of HIV antigen-specific NK cells in PLWH, driven by the CD94/NKG2C receptor and viral peptides that stabilize HLA-E surface expression (44)**. Mechanisms of recognition and function were confirmed in assays with clonal memory-like NK cells, which showed high expression of KLRG1, NKG2C, and α4β7. Importantly, no HIV peptide-specific NK cell responses were observed in people without HIV, further supporting an antigen-specific memory mechanism.
HARNESSING IMMUNE CELLS FOR VIRAL CONTROL
To achieve undetectable viral loads in the absence of cART, therapeutics would need to induce sustained immune responses, since latently infected cells would continue to reactivate over time. While “shock-and-kill” approaches remain an attractive strategy to make meaningful reductions in the HIV reservoir, clinical trials have shown no delay in time to viral rebound following cART interruption with latency reversing agents (reviewed in (45)). Given the extensive amount of work to address latency reversal, we defer to a review by Rodari et al for a more comprehensive analysis of the current strategies around this approach (46). Below we will focus on methods to enhance the antiviral activity of CTLs and NK cells in the context of productively infected cells with the goal of achieving immune-mediated control of infection.
HLA-E: a common element for CTL and NK cell therapies
Insights from HIV controllers and CD8 depletion studies in NHPs (47,48) highlight the role of CD8+ T cells in controlling infection. However, determining which viral antigens to employ as immunogens remains a challenge for universal T cell vaccine design. While targeting mutation-resistant regions may overcome this hurdle (reviewed in (49) and (50)), an added level of complexity arises from HLA diversity across different populations. Targeting conserved viral epitopes presented on HLA-E may overcome this obstacle, as only two alleles are expressed. For more than 12 years, much attention has been paid to the rhesus CMV (strain 68–1) vaccine platform, which is unique in its ability to prime MHC-E-restricted T cell responses. The viral vector expresses SIV antigens, and, remarkably, provides NHPs an overall 59% protection from mucosal challenge with SIVmac239 (51–56). Protection is mediated by vaccine-induced MHC-E-restricted CTLs, which only develop if VL9 peptide in the viral protein, Rh67, is present in the vaccine vector (54), and if the vector’s myeloid tropism is maintained (53). Emphasizing that viral control is multifaceted, the development of MHC-E-restricted CTL responses in vaccinated NHPs is not enough to distinguish protected versus unprotected animals (55). Recent studies suggested that the change-from-baseline of an IL-15 response signature correlates with vaccine protection (55)*, but how this can be exploited to yield 100% vaccine efficacy is unknown. While HIV-specific HLA-E restricted CD8+ T cells have been observed in PLWH (57), and proof-of-principle studies show that these antiviral human responses can be primed in vitro (58), it’s unclear whether induction of these responses in humans will provide protection similar to what’s seen in NHPs. In an exciting step forward, VIR-1388 (HCMV-HIV vaccine) is currently being tested for safety and immunogenicity in a Phase 1 clinical trial in adults without HIV (NCT05854381). While initially considered a prophylactic, VIR-1388 may also possess therapeutic potential. If the vaccine were given during cART, could HLA-E-specific CTL responses prevent viral rebound during ATI?
Perhaps an optimal vaccine would stimulate both HLA-E-restricted CTL responses and the newly identified HIV antigen-specific NKG2C+ NK cells. Indeed, earlier work by Reeves et al showed that memory NK cell responses were observed in Ad26/SIV-vaccinated NHPs (42). With the recent study by Jost and colleagues confirming that HIV antigen-specific NK cells are found in PLWH, it is plausible that both CTL and NK cell responses could be targeted against more conserved (or potentially “networked”), viral peptides on HLA-E (44). Methods to enhance IL-15 signaling during the time of vaccination may be required to ensure proper development of effective HLA-E-restricted CTL responses and boost NK cell activity. The IL-15 superagonist, N-803, developed by Immunity Bio, has not only been shown to act as a latency reversal agent (47,59,60) but can also enhance activation of both T and NK cells in PLWH on cART (60)*. This could be supplemented with a STAT5 agonist, HODHBt, which synergizes with IL-15 to enhance activation of HIV-specific T cells (61)*. Separately, as the antigen-specific NK cells are KLRG1+ (44), blocking this pathway during chronic infection may be a method to further enhnace the expansion and antiviral activity of this NK cell population (21), but how this would affect the antigen-specific CTL populations is unknown. Finally, the evaluation of these strategies should consider whether HIV nef would adapt to elicit stronger HLA-E downregulation (as is seen with primary strains of HIV (62)) in response to CTL/NK selective pressure. This would make the case for also engaging NKG2A+ NK cell populations.
HIV-specific antibodies - neutralizing virus and engaging cellular immunity
One promising area for HIV therapeutics is in broadly neutralizing antibodies (bNAbs). In addition to their ability to neutralize the virus, preventing entry into new target cells, the Fc portion of the antibodies can engage in effector functions. While NK cells primarily function as innate immune cells, their expression of the Fc receptor FcRyIII (CD16) allows them to engage and kill antibody opsonized infected cells through antibody-dependent cellular cytotoxicity (ADCC) (reviewed in (63)). Of note, while ADCC enhances NK cell killing of HIV-infected CD4+ T cells (64–66), NK cell ADCC is less efficient towards infected macrophages in vitro, indicating that particular cell types may have mechanisms of limiting ADDC responses (65,67). In addition, Fc receptor expression on professional antigen-presenting cells allows them to capture and process antigen to stimulate T cells. Together, this emphasizes the potential for antibodies to engage mutliple immune compartments to help control infection.
In NHPs, treatment with bNAbs resulted in significant delays to viral rebound following ATI (68). In humans, the bNAb 3BNC117 is effective at delaying time to viral rebound during ATI as a monotherapy (69,70), with efficacy further enhanced when used in combination with another bNAb, 10–1074 (71). Ultimately, rebound was correlated with a loss of circulating antibody (71). The observed efficacy may be due to enhanced HIV-specific CTL activity (72,73)(* for 73). While vaccinal effects, where bNAbs form immune complexes and stimulate antigen processing and presentation by antigen-presenting cells (74), may play a role in control, to date the precise mechanism by which cellular immunity is enhanced is not known.
Combining bNAb treatment with other therapies to achieve more durable responses is an active area of research. One such approach is stimulation of the innate immune system via TLR agonism, which could enhance NK cell activity (75,76). This approach has shown promise. Lefitolimod, a TLR9 agonist, caused increased NK cell activation and HIV transcription in PLWH on cART (77). However the recent TITAN trial showed no additional benefit from treatment with lefitolimod in addition to 3BNC117 and 10–1074 (78)*. Additional studies are in process using the TLR7 agonist Vesatolimod (GS9620), which has shown slightly delayed time to viral rebound following ATI as a monotherapy (79), and is under investigation as a combination with a T cell vaccine (80). Finally, additional clinical trials are underway to assess whether N-803, to enhance CTL and NK cell activity, could contribute to delayed viral rebound in combination with bNAbs during ATI (NCT05245292 and NCT04340596).
HIV-specific T cell products and biologics
Taking lessons from the cancer field, several groups have focused on development of HIV-specific chimeric antigen receptor (CAR) T cells. Maldini et al developed HIV-resistant CAR T cells that have both the proliferative benefits of the 4–1BB CAR T and the enhanced effector function of the CD28 CAR. Adoptive transfer of these CAR T cells resulted in a slower decline in CD4+ T cells during acute infection in humanized mice, and when given in combination with cART, accelerated the decline in HIV RNA and decreased the frequencies of Gag+ cells in the tissues, including Gag+ CD14+ macrophages (81). This supports an earlier report that HIV-infected macrophages are susceptible to HIV-specific CAR T cells (65). Recently, duoCAR T cells, which are created using autologous T cells transduced with two CAR constructs (mD1.22-CAR and m36.4-CAR), eliminated HIV-infected splenic PBMCs of humanized mice, along with infected CD4+ T cells and monocytes in vitro (82)**. These are currently being tested in a Phase I/IIa clinical trial in PLWH (NCT04648046). Another interesting CAR T cell concept is CMV and HIV dual-specific CAR T cells, which have been tested in conjunction with a CMV vaccine in humanized mice; these endogenous CMV-specific T cells, made to express an anti-gp120 CAR, have been shown to expand in vivo and reduce viral load during cART and after cART interruption (83). Finally, promoting CAR T cell infiltration into the B cell follicle via CXCR5 overexpression may enhance delay to viral rebound following cART cessation (84). Overall, CAR T cells with multiple specificities that can persist in vivo, expand in response to a controlled stimulus, and infiltrate viral sancturaies in the B cell follicle may prove useful in reducing the HIV reservoir.
Also an inspiration from the cancer field (85–88), various bispecifics are under investigation to enhance clearance of HIV-infected cells. Bispecific killer cell engagers (BiKEs) (89) and bispecific T cell engagers (BiTEs) (90) show pre-clinical promise in enhancing directed cytotoxicity of NK cells and T cells, respectively, towards HIV-infected targets. Trispecific killer cell engagers (TriKEs), which engage CD3 and CD28 of the effector cell, along with the CD4 binding site N6, will potentially prove to be an improved platform (91)*.
CONCLUSION
Achieving sustained viral control following cART cessation will likely require engagement of multiple immune cell types. Decades of research characterizing PLWH who naturally control infection, mechanistic studies elucidating mechanisms of antiviral cellular immunity, and lessons from the cancer immunotherapy field have yielded inspiration for current strategies to control viral infection in the absence of cART. It’s clear that emerging themes in the cure field include HLA-E coordination of CTL and NK cell responses, IL-15 enhancement of CTL and NK cell function, and therapies that take advantage of bNAb breadth and cellular immunity. Understanding the nature of the HIV reservoir will be essential for optimizing these strategies for maximal antiviral activity. Indeed, over 10 years of research has shown that infected myeloid cells react differently than T cells to cytolytic immune cells, thus, the strategies discussed in this review would greatly benefit from assessments of antiviral activity towards these different cellular reservoirs.
KEY POINTS.
Rhesus CMV SIV vaccine-induced CD8+ T cells and memory NKG2C+ NK cells that recognize viral anitgens on MHC-E could be exploited to control infection.
IL-15 superagonist, N-803, is a potent booster of cellular immunity and latency reversal, the efficacy of which could be enhanced by small molesules that target STAT5 activtaion.
Inspired by the success of cancer immunotherapies, HIV broadly neutralizing antibodies and antibody fragments to engage cellular immunity show promise in pre-clinical models.
HIV-infected macrophages and dendritic cells have been observed in several tissues and should be considered as part of the resevroir that needs to be targeted for immune-based control of infection.
ACKNOWLEDGEMENTS
Financial support for PG, AS, and KLC provided by the NIH DP2 AI154438 and UM1 AI164565 awards. In addition, PG is supported by the T32 AI007349 award. The authors have no conflicts of interest to declare.
REFERENCES
- 1.Ganor Y, Real F, Sennepin A, Dutertre CA, Prevedel L, Xu L, et al. HIV-1 reservoirs in urethral macrophages of patients under suppressive antiretroviral therapy. Nat Microbiol. 2019. Apr;4(4):633–44. [DOI] [PubMed] [Google Scholar]
- 2.Ko A, Kang G, Hattler JB, Galadima HI, Zhang J, Li Q, et al. Macrophages but not Astrocytes Harbor HIV DNA in the Brains of HIV-1-Infected Aviremic Individuals on Suppressive Antiretroviral Therapy. J Neuroimmune Pharmacol. 2019. Mar;14(1):110–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Real F, Zhu A, Huang B, Belmellat A, Sennepin A, Vogl T, et al. S100A8-mediated metabolic adaptation controls HIV-1 persistence in macrophages in vivo. Nat Commun. 2022. Oct 11;13(1):5956. *This study characterizes HIV-infected macrophages in the genital mucosa, showing that the cells express HIV RNA and protein and that viral production can be induced by the alarmin S100A8, supporting the narrative that macrophages are physiologically relevant tissue reservoirs.
- 4. Banga R, Procopio FA, Lana E, Gladkov GT, Roseto I, Parsons EM, et al. Lymph node dendritic cells harbor inducible replication-competent HIV despite years of suppressive ART. Cell Host Microbe. 2023. Oct 11;31(10):1714–1731.e9. *Banga and colleagues show that lymph node DCs from PLWH on cART harbor replication-competent HIV, suggesting that in addition to macrophages, DCs should be considered as targets in cure strategies. It also emphasizes that DCs vs macrophages may represent distinct tissue reservoirs.
- 5.Andrade VM, Mavian C, Babic D, Cordeiro T, Sharkey M, Barrios L, et al. A minor population of macrophage-tropic HIV-1 variants is identified in recrudescing viremia following analytic treatment interruption. Proc Natl Acad Sci U S A. 2020. May 5;117(18):9981–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Veenhuis RT, Abreu CM, Costa PAG, Ferreira EA, Ratliff J, Pohlenz L, et al. Monocyte-derived macrophages contain persistent latent HIV reservoirs. Nat Microbiol. 2023. May;8(5):833–44. *Using a myeloid-adapted IPDA and QVOA, the authors show that replication-competent HIV can be derived from peripheral blood monocytes in PLWH on cART. Being the macrophage precursors, infected monocytes cells could seed tissue reservoirs, however, it is unclear how long-lived these cells are.
- 7.Vancoillie L, Hebberecht L, Dauwe K, Demecheleer E, Dinakis S, Vaneechoutte D, et al. Longitudinal sequencing of HIV-1 infected patients with low-level viremia for years while on ART shows no indications for genetic evolution of the virus. Virology. 2017. Oct 1;510:185–93. [DOI] [PubMed] [Google Scholar]
- 8. Beckford-Vera DR, Flavell RR, Seo Y, Martinez-Ortiz E, Aslam M, Thanh C, et al. First-in-human immunoPET imaging of HIV-1 infection using 89Zr-labeled VRC01 broadly neutralizing antibody. Nat Commun. 2022. Mar 9;13(1):1219. *The first-in-human ImmunoPET study where radiolabeled VRC01 was used to detect tissue reservoirs via a PET/MR scan. On cART, compared to people not living with HIV, virus was specifically detected in the gut/colon, the bone marrow, and nasal turbinate, supporting the idea of active viral protein production in tissues under cART.
- 9.Wiegand A, Spindler J, Hong FF, Shao W, Cyktor JC, Cillo AR, et al. Single-cell analysis of HIV-1 transcriptional activity reveals expression of proviruses in expanded clones during ART. Proceedings of the National Academy of Sciences. 2017. May 2;114(18):E3659–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Dubé M, Tastet O, Dufour C, Sannier G, Brassard N, Delgado GG, et al. Spontaneous HIV expression during suppressive ART is associated with the magnitude and function of HIV-specific CD4+ and CD8+ T cells. Cell Host & Microbe. 2023. Sep;31(9):1507–1522.e5. *In the context of cART, spontaneous expression of viral RNA and protein stimulate T cell responses, supporting the idea of active viral protein production in tissues under cART.
- 11.Collins DR, Gaiha GD, Walker BD. CD8(+) T cells in HIV control, cure and prevention. Nat Rev Immunol. 2020. Aug;20(8):471–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Walker BD, Chakrabarti S, Moss B, Paradis TJ, Flynn T, Durno AG, et al. HIV-specific cytotoxic T lymphocytes in seropositive individuals. Nature. 1987. Jul;328(6128):345–8. [DOI] [PubMed] [Google Scholar]
- 13.Goulder PJR, Watkins DI. HIV and SIV CTL escape: implications for vaccine design. Nat Rev Immunol. 2004. Aug;4(8):630–40. [DOI] [PubMed] [Google Scholar]
- 14.Gaiha GD, Rossin EJ, Urbach J, Landeros C, Collins DR, Nwonu C, et al. Structural topology defines protective CD8+ T cell epitopes in the HIV proteome. Science (New York, NY). 2019. May 5;364(6439):480. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Migueles SA, Laborico AC, Shupert WL, Sabbaghian MS, Rabin R, Hallahan CW, et al. HIV-specific CD8+ T cell proliferation is coupled to perforin expression and is maintained in nonprogressors. Nat Immunol. 2002. Nov;3(11):1061–8. [DOI] [PubMed] [Google Scholar]
- 16.Mothe B, Llano A, Ibarrondo J, Zamarreño J, Schiaulini M, Miranda C, et al. CTL Responses of High Functional Avidity and Broad Variant Cross-Reactivity Are Associated with HIV Control. PLOS ONE. 2012. Jan 4;7(1):e29717. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Collins DR, Urbach JM, Racenet ZJ, Arshad U, Power KA, Newman RM, et al. Functional impairment of HIV-specific CD8(+) T cells precedes aborted spontaneous control of viremia. Immunity. 2021. Oct 12;54(10):2372–2384 e7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Mutascio S, Mota T, Franchitti L, Sharma AA, Willemse A, Bergstresser SN, et al. CD8+ T cells promote HIV latency by remodeling CD4+ T cell metabolism to enhance their survival, quiescence, and stemness. Immunity. 2023;56(5):1132–47. *Activated CD8+ T cells from HIV naïve donors promote cellular quiescence and latency in a co-culture with autologous HIV-infected memory CD4+ T cells, suggesting non-cytolytic mechanisms of HIV control. While this finding is novel, it may pose an additional challenge for targeting the infected cell reservoirs for elimination.
- 19.Davis ZB, Sowrirajan B, Cogswell A, Ward JP, Planelles V, Barker E. CD155 on HIV-Infected Cells Is Not Modulated by HIV-1 Vpu and Nef but Synergizes with NKG2D Ligands to Trigger NK Cell Lysis of Autologous Primary HIV-Infected Cells. AIDS Res Hum Retroviruses. 2017. Feb;33(2):93–100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Cerboni C, Neri F, Casartelli N, Zingoni A, Cosman D, Rossi P, et al. Human immunodeficiency virus 1 Nef protein downmodulates the ligands of the activating receptor NKG2D and inhibits natural killer cell-mediated cytotoxicity. J Gen Virol. 2007. Jan;88(Pt 1):242–50. [DOI] [PubMed] [Google Scholar]
- 21. Astorga-Gamaza A, Perea D, Sanchez-Gaona N, Calvet-Mirabent M, Gallego-Cortés A, Grau-Expósito J, et al. KLRG1 expression on natural killer cells is associated with HIV persistence, and its targeting promotes the reduction of the viral reservoir. Cell Rep Med. 2023. Oct 17;4(10):101202. **Dysfunctional NK cells expressing the inhibitory receptor, KLRG1, are found in the periphery and lymph nodes in PLWH on cART, and are associated with higher viral transcription. Importantly, blocking KLRG1 using an antibody enhanced NK cell function, suggesting another method to promote antiviral cytolytic immunity using a new type of checkpoint blockade.
- 22.Shah AH, Sowrirajan B, Davis ZB, Ward JP, Campbell EM, Planelles V, et al. Degranulation of natural killer cells following interaction with HIV-1-infected cells is hindered by downmodulation of NTB-A by Vpu. Cell Host Microbe. 2010. Nov 18;8(5):397–409. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Matusali G, Potestà M, Santoni A, Cerboni C, Doria M. The Human Immunodeficiency Virus Type 1 Nef and Vpu Proteins Downregulate the Natural Killer Cell-Activating Ligand PVR. J Virol. 2012. Apr;86(8):4496–504. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Fittje P, Hœlzemer A, Garcia-Beltran WF, Vollmers S, Niehrs A, Hagemann K, et al. HIV-1 Nef-mediated downregulation of CD155 results in viral restriction by KIR2DL5+ NK cells. PLOS Pathogens. 2022. Jun 24;18(6):e1010572. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Richard J, Sindhu S, Pham TN, Belzile JP, Cohen EA. HIV-1 Vpr up-regulates expression of ligands for the activating NKG2D receptor and promotes NK cell-mediated killing. Blood. 2010. Feb 18;115(7):1354–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Boulet S, Kleyman M, Kim JY, Kamya P, Sharafi S, Simic N, et al. A combined genotype of KIR3DL1 high expressing alleles and HLA-B*57 is associated with a reduced risk of HIV infection. AIDS. 2008. Jul 31;22(12):1487. [DOI] [PubMed] [Google Scholar]
- 27.Montoya CJ, Velilla PA, Chougnet C, Landay AL, Rugeles MT. Increased IFN-γ production by NK and CD3+/CD56+ cells in sexually HIV-1-exposed but uninfected individuals. Clinical Immunology. 2006. Aug 1;120(2):138–46. [DOI] [PubMed] [Google Scholar]
- 28.Zhao NQ, Vendrame E, Ferreira AM, Seiler C, Ranganath T, Alary M, et al. Natural killer cell phenotype is altered in HIV-exposed seronegative women. PLoS One. 2020;15(9):e0238347. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Rincón DS, Flórez-Álvarez L, Taborda NA, Hernandez JC, Rugeles MT, Zapata-Builes W. NK cells from Men Who Have Sex with Men at high risk for HIV-1 infection exhibit higher effector capacity. Sci Rep. 2023. Oct 5;13(1):16766. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Scott-Algara D, Truong LX, Versmisse P, David A, Luong TT, Nguyen NV, et al. Cutting edge: increased NK cell activity in HIV-1-exposed but uninfected Vietnamese intravascular drug users. J Immunol. 2003. Dec 1;171(11):5663–7. [DOI] [PubMed] [Google Scholar]
- 31.Kazer SW, Aicher TP, Muema DM, Carroll SL, Ordovas-Montanes J, Miao VN, et al. Integrated single-cell analysis of multicellular immune dynamics during hyperacute HIV-1 infection. Nat Med. 2020. Apr;26(4):511–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Woolley G, Mosher M, Kroll K, Jones R, Hueber B, Sugawara S, et al. Natural Killer Cells Regulate Acute SIV Replication, Dissemination, and Inflammation, but Do Not Impact Independent Transmission Events. J Virol. 2023. Jan 31;97(1):e0151922. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Qi Y, Martin MP, Gao X, Jacobson L, Goedert JJ, Buchbinder S, et al. KIR/HLA Pleiotropism: Protection against Both HIV and Opportunistic Infections. PLOS Pathogens. 2006. Aug 18;2(8):e79. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Martin MP, Qi Y, Gao X, Yamada E, Martin JN, Pereyra F, et al. Innate partnership of HLA-B and KIR3DL1 subtypes against HIV-1. Nat Genet. 2007. Jun;39(6):733–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Martin MP, Gao X, Lee JH, Nelson GW, Detels R, Goedert JJ, et al. Epistatic interaction between KIR3DS1 and HLA-B delays the progression to AIDS. Nat Genet. 2002. Aug;31(4):429–34. [DOI] [PubMed] [Google Scholar]
- 36. Kim JT, Zhang TH, Carmona C, Lee B, Seet CS, Kostelny M, et al. Latency reversal plus natural killer cells diminish HIV reservoir in vivo. Nat Commun. 2022. Jan 10;13(1):121. *Allogenieic NK cells, when administered to HIV-infected humanized mice during cART interruption, delayed viral rebound, an effect that was enhanced in the presence of a latency reversing agent, supporting the idea of exploiting NK cell activity for cure strategies.
- 37.Huot N, Rascle P, Petitdemange C, Contreras V, Stürzel CM, Baquero E, et al. SIV-induced terminally differentiated adaptive NK cells in lymph nodes associated with enhanced MHC-E restricted activity. Nat Commun. 2021. Feb 24;12:1282. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Huot N, Rascle P, Petitdemange C, Contreras V, Palgen JL, Stahl-Hennig C, et al. Non-human Primate Determinants of Natural Killer Cells in Tissues at Steady-State and During Simian Immunodeficiency Virus Infection. Frontiers in Immunology [Internet]. 2020. [cited 2023 Oct 20];11. Available from: https://www.frontiersin.org/articles/10.3389/fimmu.2020.02134 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.O’Leary JG, Goodarzi M, Drayton DL, von Andrian UH. T cell- and B cell-independent adaptive immunity mediated by natural killer cells. Nat Immunol. 2006. May;7(5):507–16. [DOI] [PubMed] [Google Scholar]
- 40.Foley B, Cooley S, Verneris MR, Curtsinger J, Luo X, Waller EK, et al. Human CMV-induced Memory-like NKG2C+ NK cells are Transplantable and Expand in vivo in Response to Recipient CMV Antigen. J Immunol. 2012. Nov 15;189(10):5082–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Wang Y, Lifshitz L, Gellatly K, Vinton CL, Busman-Sahay K, McCauley S, et al. HIV-1-induced cytokines deplete homeostatic innate lymphoid cells and expand TCF7-dependent memory NK cells. Nat Immunol. 2020. Mar;21(3):274–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Reeves RK, Li H, Jost S, Blass E, Li H, Schafer JL, et al. Antigen-specific NK cell memory in rhesus macaques. Nat Immunol. 2015. Sep;16(9):927–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Davis ZB, Cogswell A, Scott H, Mertsching A, Boucau J, Wambua D, et al. A Conserved HIV-1-Derived Peptide Presented by HLA-E Renders Infected T-cells Highly Susceptible to Attack by NKG2A/CD94-Bearing Natural Killer Cells. PLoS Pathog. 2016. Feb 1;12(2):e1005421. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Jost S, Lucar O, Lee E, Yoder T, Kroll K, Sugawara S, et al. Antigen-specific memory NK cell responses against HIV and influenza use the NKG2/ HLA-E axis. Science Immunology. 2023. Dec 8;8(90):eadi3974. **Supporting the momentum building around the idea of memory NK cells, this paper elegantly shows with NK cell cloning techniques that HIV antigen-specific NK cells that are NKG2C+ and KLRG1+ are found in PLWH, but not HIV naïve individuals. Given the specificity of antigen presented on HLA-E, this works suggests that these NK cells may be able to be induced upon vaccination.
- 45.Debrabander Q, Hensley KS, Psomas CK, Bramer W, Mahmoudi T, van Welzen BJ, et al. The efficacy and tolerability of latency-reversing agents in reactivating the HIV-1 reservoir in clinical studies: a systematic review. Journal of Virus Eradication. 2023. Sep 1;9(3):100342. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Rodari A, Darcis G, Van Lint CM. The Current Status of Latency Reversing Agents for HIV-1 Remission. Annu Rev Virol. 2021. Sep 29;8(1):491–514. [DOI] [PubMed] [Google Scholar]
- 47.McBrien JB, Mavigner M, Franchitti L, Smith SA, White E, Tharp GK, et al. Robust and persistent reactivation of SIV and HIV by N-803 and depletion of CD8+ cells. Nature. 2020. Feb;578(7793):154–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Chowdhury A, Hayes TL, Bosinger SE, Lawson BO, Vanderford T, Schmitz JE, et al. Differential Impact of In Vivo CD8+ T Lymphocyte Depletion in Controller versus Progressor Simian Immunodeficiency Virus-Infected Macaques. Journal of Virology. 2015. Aug 3;89(17):8677–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Kaseke C, Tano-Menka R, Senjobe F, Gaiha GD. The Emerging Role for CTL Epitope Specificity in HIV Cure Efforts. J Infect Dis. 2021. Feb 15;223(12 Suppl 2):32–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Korber B, Fischer W. T cell-based strategies for HIV-1 vaccines. Hum Vaccin Immunother. 2019. Oct 25;16(3):713–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Hansen SG, Marshall EE, Malouli D, Ventura AB, Hughes CM, Ainslie E, et al. A live-attenuated RhCMV/SIV vaccine shows long-term efficacy against heterologous SIV challenge. Sci Transl Med. 2019. Jul 17;11(501):eaaw2607. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Hansen SG, Ford JC, Lewis MS, Ventura AB, Hughes CM, Coyne-Johnson L, et al. Profound early control of highly pathogenic SIV by an effector memory T-cell vaccine. Nature. 2011. May;473(7348):523–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Hansen SG, Hancock MH, Malouli D, Marshall EE, Hughes CM, Randall KT, et al. Myeloid cell tropism enables MHC-E-restricted CD8+ T cell priming and vaccine efficacy by the RhCMV/SIV vaccine. Sci Immunol. 2022. Jun 24;7(72):eabn9301. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Verweij MC, Hansen SG, Iyer R, John N, Malouli D, Morrow D, et al. Modulation of MHC-E transport by viral decoy ligands is required for RhCMV/SIV vaccine efficacy. Science. 2021. Apr 30;372(6541):eabe9233. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Barrenäs F, Hansen SG, Law L, Driscoll C, Green RR, Smith E, et al. Interleukin-15 response signature predicts RhCMV/SIV vaccine efficacy. PLOS Pathogens. 2021. Jul 6;17(7):e1009278. *While previous reports characterizing the 68–1 rhesus CMV vector vaccinated non-human primates show that induced MHC-E-restricted CD8+ T cell responses are required for protection for SIV challenge, this study identified an IL-15 signaling signature, induced upon vaccination, that correlated with protection. This adds to several other studies characterizing the correlates and mechanisms of protection from SIV challenge for rhesus CMV vaccinated animals, an important step for translating this work to the clinic.
- 56.Picker LJ, Lifson JD, Gale M, Hansen SG, Früh K. Programming cytomegalovirus as an HIV vaccine. Trends in Immunology. 2023. Apr 1;44(4):287–304. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Bansal A, Gehre MN, Qin K, Sterrett S, Ali A, Dang Y, et al. HLA-E–restricted HIV-1–specific CD8+ T cell responses in natural infection. J Clin Invest [Internet]. 2021. Aug 16 [cited 2023 Nov 18];131(16). Available from: https://www.jci.org/articles/view/148979 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Yang H, Rei M, Brackenridge S, Brenna E, Sun H, Abdulhaqq S, et al. HLA-E-restricted, Gag-specific CD8+ T cells can suppress HIV-1 infection, offering vaccine opportunities. Sci Immunol. 2021. Mar 25;6(57):eabg1703. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.McBrien JB, Wong AKH, White E, Carnathan DG, Lee JH, Safrit JT, et al. Combination of CD8β Depletion and Interleukin-15 Superagonist N-803 Induces Virus Reactivation in Simian-Human Immunodeficiency Virus-Infected, Long-Term ART-Treated Rhesus Macaques. Journal of Virology. 2020. Sep 15;94(19): 10.1128/jvi.00755-20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Miller JS, Davis ZB, Helgeson E, Reilly C, Thorkelson A, Anderson J, et al. Safety and virologic impact of the IL-15 superagonist N-803 in people living with HIV: a phase 1 trial. Nat Med. 2022. Feb;28(2):392–400. *This phase I clinical trail of N-803 showed increased NK cell and CD8+ T cell activation and proliferation following administration, in addition to an increase in active HIV transcription in PBMCs, confirming that in vivo, like what is observed in vitro, the IL-15 superagonist acts as both a method to enhance cellular immunity and reverse latency
- 61. Copertino DC, Holmberg CS, Weiler J, Ward AR, Howard JN, Levinger C, et al. The latency-reversing agent HODHBt synergizes with IL-15 to enhance cytotoxic function of HIV-specific T cells. JCI Insight. 2023. Sep 22;8(18):e169028. *HODHBt treatment of PBMCs from PLWH enhances the activity of IL-15, resulting in enhanced cytotoxic activity of HIV-specific CD8+ T cells. Such boosting of effector function, in addition to the previously reported enhancement of latency reversing activity, suggests that in HODHBt or derivatives could enhance the therapeutic efficacy of IL-15 or N-803.
- 62.van Stigt Thans T, Akko JI, Niehrs A, Garcia-Beltran WF, Richert L, Stürzel CM, et al. Primary HIV-1 Strains Use Nef To Downmodulate HLA-E Surface Expression. J Virol. 2019. Oct 15;93(20):e00719–19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Bernard NF, Kant S, Kiani Z, Tremblay C, Dupuy FP. Natural Killer Cells in Antibody Independent and Antibody Dependent HIV Control. Frontiers in Immunology [Internet]. 2022. [cited 2023 Nov 23];13. Available from: https://www.frontiersin.org/articles/10.3389/fimmu.2022.879124 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Bruel T, Guivel-Benhassine F, Amraoui S, Malbec M, Richard L, Bourdic K, et al. Elimination of HIV-1-infected cells by broadly neutralizing antibodies. Nat Commun. 2016. Mar 3;7(1):10844. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Clayton KL, Mylvaganam G, Villasmil-Ocando A, Stuart H, Maus MV, Rashidian M, et al. HIV-infected macrophages resist efficient NK cell-mediated killing while preserving inflammatory cytokine responses. Cell Host Microbe. 2021. Mar 10;29(3):435–447.e9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Ward J, Bonaparte M, Sacks J, Guterman J, Fogli M, Mavilio D, et al. HIV modulates the expression of ligands important in triggering natural killer cell cytotoxic responses on infected primary T-cell blasts. Blood. 2007. Aug 15;110(4):1207–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Laumaea A, Marchitto L, Ding S, Beaudoin-Bussières G, Prévost J, Gasser R, et al. Small CD4 mimetics sensitize HIV-1-infected macrophages to antibody-dependent cellular cytotoxicity. Cell Reports. 2023. Jan;42(1):111983. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Shingai M, Nishimura Y, Klein F, Mouquet H, Donau OK, Plishka R, et al. Antibody-mediated immunotherapy of macaques chronically infected with SHIV suppresses viraemia. Nature. 2013. Nov;503(7475):277–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Scheid JF, Horwitz JA, Bar-On Y, Kreider EF, Lu CL, Lorenzi JCC, et al. HIV-1 antibody 3BNC117 suppresses viral rebound in humans during treatment interruption. Nature. 2016. Jul 28;535(7613):556–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Caskey M, Klein F, Lorenzi JCC, Seaman MS, West AP, Buckley N, et al. Viraemia suppressed in HIV-1-infected humans by broadly neutralizing antibody 3BNC117. Nature. 2015. Jun;522(7557):487–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Mendoza P, Gruell H, Nogueira L, Pai JA, Butler AL, Millard K, et al. Combination therapy with anti-HIV-1 antibodies maintains viral suppression. Nature. 2018. Sep;561(7724):479–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. Rosás-Umbert M, Gunst JD, Pahus MH, Olesen R, Schleimann M, Denton PW, et al. Administration of broadly neutralizing anti-HIV-1 antibodies at ART initiation maintains long-term CD8+ T cell immunity. Nat Commun. 2022. Oct 29;13(1):6473. *In the eCLEAR study, at the time of cART initiation, in trial participants that exhibited endogenous virus sensitivity to 3BNC117, the administration of this antibody prolonged HIV-specific CD8+ T cell activity for up to 1 year and correlated with an increased time to viral rebound. Thus, beyond its viral neutralizing effects, antibodies can be used to enhance cellular immunity in PLWH.
- 73.Niessl J, Baxter AE, Mendoza P, Jankovic M, Cohen YZ, Butler AL, et al. Combination anti-HIV-1 antibody therapy is associated with increased virus-specific T cell immunity. Nat Med. 2020;26(2):222–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Bournazos S, Ravetch JV. Fcγ Receptor Pathways during Active and Passive Immunization. Immunol Rev. 2015. Nov;268(1):88–103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Offersen R, Nissen SK, Rasmussen TA, Østergaard L, Denton PW, Søgaard OS, et al. A Novel Toll-Like Receptor 9 Agonist, MGN1703, Enhances HIV-1 Transcription and NK Cell-Mediated Inhibition of HIV-1-Infected Autologous CD4+ T Cells. J Virol. 2016. Apr 14;90(9):4441–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Noh JY, Yoon SR, Kim TD, Choi I, Jung H. Toll-Like Receptors in Natural Killer Cells and Their Application for Immunotherapy. J Immunol Res. 2020. Jan 4;2020:2045860. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Vibholm L, Schleimann MH, Højen JF, Benfield T, Offersen R, Rasmussen K, et al. Short-Course Toll-Like Receptor 9 Agonist Treatment Impacts Innate Immunity and Plasma Viremia in Individuals With Human Immunodeficiency Virus Infection. Clin Infect Dis. 2017. Jun 15;64(12):1686–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. Gunst JD, Højen JF, Pahus MH, Rosás-Umbert M, Stiksrud B, McMahon JH, et al. Impact of a TLR9 agonist and broadly neutralizing antibodies on HIV-1 persistence: the randomized phase 2a TITAN trial. Nat Med. 2023. Oct;29(10):2547–58. *In the TITAN trial, while lefitolimod had no effect on viral rebound, treatment of participants with 3BNC117 and 10–1074 resulted in delayed time to viral rebound, with the results of the bNAb treatment agreeing with prior studies. Why the TLR9 agonist, which has been shown to increase CD16+ NK cells in PLWH, failed to enhance delay to viral rebound will require further study.
- 79.SenGupta D, Brinson C, DeJesus E, Mills A, Shalit P, Guo S, et al. The TLR7 agonist vesatolimod induced a modest delay in viral rebound in HIV controllers after cessation of antiretroviral therapy. Science Translational Medicine. 2021. Jun 23;13(599):eabg3071. [DOI] [PubMed] [Google Scholar]
- 80.Bailón L, Llano A, Cedeño S, Escribà T, Rosás-Umbert M, Parera M, et al. Safety, immunogenicity and effect on viral rebound of HTI vaccines in early treated HIV-1 infection: a randomized, placebo-controlled phase 1 trial. Nat Med. 2022. Dec;28(12):2611–21. [DOI] [PubMed] [Google Scholar]
- 81.Maldini CR, Claiborne DT, Okawa K, Chen T, Dopkin DL, Shan X, et al. Dual CD4-based CAR T cells with distinct costimulatory domains mitigate HIV pathogenesis in vivo. Nat Med. 2020. Nov;26(11):1776–87. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82. Anthony-Gonda K, Ray A, Su H, Wang Y, Xiong Y, Lee D, et al. In vivo killing of primary HIV-infected cells by peripheral-injected early memory-enriched anti-HIV duoCAR T cells. JCI Insight. 2022. Nov 8;7(21):e161698. **CAR T cells with dual specificity for the CD4 binding site and co-receptor binding site of the HIV env exhibited antiviral function in vivo and were able to kill HIV-infected monocytes, supporting earlier work that CAR T cells can kill HIV-infected macrophages. This CAR construct is currently being tested in the clinic.
- 83.Guan M, Lim L, Holguin L, Han T, Vyas V, Urak R, et al. Pre-clinical data supporting immunotherapy for HIV using CMV-HIV-specific CAR T cells with CMV vaccine. Molecular Therapy - Methods & Clinical Development. 2022. Jun 9;25:344–59. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Pampusch MS, Abdelaal HM, Cartwright EK, Molden JS, Davey BC, Sauve JD, et al. CAR/CXCR5-T cell immunotherapy is safe and potentially efficacious in promoting sustained remission of SIV infection. PLoS Pathog. 2022. Feb;18(2):e1009831. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Pawlowski KD, Duffy JT, Tiwari A, Zannikou M, Balyasnikova IV. Bi-Specific Killer Cell Engager Enhances NK Cell Activity against Interleukin-13 Receptor Alpha-2 Positive Gliomas. Cells. 2023. Jun 25;12(13):1716. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Goebeler ME, Bargou RC. T cell-engaging therapies - BiTEs and beyond. Nat Rev Clin Oncol. 2020. Jul;17(7):418–34. [DOI] [PubMed] [Google Scholar]
- 87.Nikkhoi SK, Li G, Eleya S, Yang G, Vandavasi VG, Hatefi A. Bispecific killer cell engager with high affinity and specificity toward CD16a on NK cells for cancer immunotherapy. Front Immunol. 2022;13:1039969. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Zah E, Nam E, Bhuvan V, Tran U, Ji BY, Gosliner SB, et al. Systematically optimized BCMA/CS1 bispecific CAR-T cells robustly control heterogeneous multiple myeloma. Nat Commun. 2020. May 8;11(1):2283. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Li W, Wu Y, Kong D, Yang H, Wang Y, Shao J, et al. One-domain CD4 Fused to Human Anti-CD16 Antibody Domain Mediates Effective Killing of HIV-1-Infected Cells. Sci Rep. 2017. Aug 22;7:9130. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Sengupta S, Board NL, Wu F, Moskovljevic M, Douglass J, Zhang J, et al. TCR-mimic bispecific antibodies to target the HIV-1 reservoir. Proceedings of the National Academy of Sciences. 2022. Apr 12;119(15):e2123406119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91. Promsote W, Xu L, Hataye J, Fabozzi G, March K, Almasri CG, et al. Trispecific antibody targeting HIV-1 and T cells activates and eliminates latently-infected cells in HIV/SHIV infections. Nat Commun. 2023. Jun 22;14(1):3719. * A trispecific antibody targeting the HIV Env (N6), CD3 and CD28 was able to activate latently infected cells and enhance CD8+ T cell elimination of these infected cells. While the antibody was well tolerated in macaques, additional follow up studies are needed to determine whether an effect on reservoir size or viral rebound will be observed.
