Reservatório HIV

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 22

HHS Public Access

Author manuscript
Curr HIV/AIDS Rep. Author manuscript; available in PMC 2023 August 22.
Author Manuscript

Published in final edited form as:


Curr HIV/AIDS Rep. 2022 June ; 19(3): 194–206. doi:10.1007/s11904-022-00604-2.

HIV-1 Reservoir Persistence and Decay: Implications for Cure


Strategies
Edward F. Kreider1, Katharine J. Bar2
1Perelman School of Medicine, University of Pennsylvania, Stemmler Hall Room 130-150, 3450

Hamilton Walk, Philadelphia, PA 19104-6073, USA


2Perelman School of Medicine, University of Pennsylvania, 502D Johnson Pavilion, 3610
Author Manuscript

Hamilton Walk, Philadelphia, PA 19104-0673, USA

Abstract
Purpose of Review—Despite suppressive antiretroviral therapy (ART), a viral reservoir
persists in individuals living with HIV that can reignite systemic replication should treatment
be interrupted. Understanding how HIV-1 persists through effective ART is essential to develop
cure strategies to induce ART-free virus remission.

Recent Findings—The HIV-1 reservoir resides in a pool of CD4-expressing cells as a range of


viral species, a subset of which is genetically intact. Recent studies suggest that the reservoir on
ART is highly dynamic, with expansion and contraction of virus-infected cells over time. Overall,
the intact proviral reservoir declines faster than defective viruses, suggesting enhanced immune
Author Manuscript

clearance or cellular turnover. Upon treatment interruption, rebound viruses demonstrate escape
from adaptive and innate immune responses, implicating these selective pressures in restriction
of virus reactivation. Cure strategies employing immunotherapy are poised to test whether host
immune pressure can be augmented to enhance reservoir suppression or clearance. Alternatively,
genomic engineering approaches are being applied to directly eliminate intact viruses and shrink
the replication-competent virus pool.

Summary—New evidence suggests host immunity exerts selective pressure on reservoir viruses
and clears HIV-1 infected cells over years on ART. Efforts to build on the detectable, but
insufficient, reservoir clearance via empiric testing in clinical trials will inform our understanding
of mechanisms of viral persistence and the direction of future cure strategies.

Keywords
Author Manuscript

HIV-1 reservoirs; Persistence; Viral rebound; Cure

Introduction
The management of people living with HIV (PLWH) has evolved over the course of the
pandemic, transforming a near certain death sentence into a manageable chronic illness

Katharine J. Bar bark@pennmedicine.upenn.edu.


Conflict of Interest The authors declare no competing interests.
Kreider and Bar Page 2

with appropriate treatment [1, 2]. Despite medical advances that have markedly improved
Author Manuscript

disease outcomes, discontinuation of antiretroviral therapy (ART) reliably results in viral


recrudescence [3, 4] and, thus, PLWH must remain on therapy for the duration of their lives.
Lifelong treatment can be complicated by drug toxicities [5, 6], medication interactions
[7, 8], adherence issues [9], stigma [10], and ongoing costs to individuals and healthcare
systems [11]. Thus, interventions to induce ART-free virus remission are a priority.

The major barrier to such a cure is the viral reservoir—a collection of viral species that
persist through ART and is capable of productively infecting new cells to reestablish viremia
upon antiretroviral cessation. Accurate characterization of this viral pool is challenging,
but of vital importance to determining mechanisms of virus persistence and to evaluate
cure strategies. Here, we will provide an overview of our current understanding of
HIV-1 persistence, highlighting newly realized reservoir features that pose obstacles and
opportunities for cure interventions. Recent work by several groups reinforces the theme
Author Manuscript

that viral reservoirs are highly dynamic, but cells harboring rebound-competent viruses
appear to decrease in size slowly on suppressive ART. We will then highlight several cure
interventions that seek to enhance this viral clearance through immunomodulation or gene
therapy.

Persistent HIV-1 Reservoirs


Viral and Cellular Reservoirs—Primate immunodeficiency viruses, including HIV-1,
mediate both active and latent infection and are capable of infecting multiple CD4-
expressing cell types. Further complicating reservoir characterization, HIV-infected cells
reside within lymphoid tissues, blood, viscera, and immune-privileged sites [12]. Reservoir
viruses can endure long-term ART as integrated DNA proviruses in cells, or intact virions
within tissue networks, that are long-lived or self-renewing. At treatment interruption,
Author Manuscript

a subset of the reservoir that is both replication- and rebound-competent evades host
restriction and rekindles systemic virus replication. Years of study in both PLWH and
animal models of persistence have shed light on the heterogeneous cellular and viral species
responsible for reservoir maintenance.

The most thoroughly characterized component of the reservoir are transcriptionally silent
DNA proviruses integrated within memory CD4+ T cells [13, 14]. Ex vivo stimulation
of latently infected memory T cells yields infectious virus, implicating this pool as a
substantial contributor to the reservoir [13, 15]. Interrogations of this long-lived proviral
reservoir indicate that it may be enriched in certain cellular subsets, e.g., central and
stem cell memory CD4 + T cells, which persist through low-level cycling or exhibit self-
renewal without activation [16, 17]. While less numerous, effector memory CD4+ T cells
Author Manuscript

have higher frequencies of intact and inducible provirus [18–20]; thus, they may serve as
important sources of reactivating virus. Furthermore, CD4+ T cells expressing exhaustion
markers, including the immune checkpoint receptors programmed death protein 1 (PD-1),
cytotoxic lymphocyte-associated protein 4 (CTLA-4), and others, are key reservoirs as they
are enriched for HIV and/or SIV infection [21, 22]. Determining the key features and
relative contributions of the CD4 + T cell reservoir based on tissue location, function, and
differentiation status remains an active area of study (reviewed in [23]). Apart from T

Curr HIV/AIDS Rep. Author manuscript; available in PMC 2023 August 22.
Kreider and Bar Page 3

cells, latently infected tissue macrophages have also been proposed as components of the
Author Manuscript

viral reservoir, as these cells can be productively infected and can durably persist [24, 25].
Replication-competent virus has been retrieved from tissue macrophages in the male genital
tract [26], and from the brain and circulation in specific animal models [27–29]; however,
the scarcity of tissue macrophages within largely inaccessible tissues challenges verification
of these findings.

In addition to quiescent HIV-1 proviruses, cells with active viral transcription and translation
have been identified in PLWH on suppressive ART and implicated in both viral persistence
and immunopathogenesis. Cell-associated viral RNA (CA-RNA), which reflects proviral
transcription, extrachromosomal circular viral DNA, which is inherently nonself-renewing,
and HIV-1 proteins, reflecting ongoing viral translation, remain detectable in PLWH during
periods of suppressed viremia [30–32]. In theory, expression of viral RNA and protein
offer opportunities for immune recognition and clearance of infected cells in the absence
Author Manuscript

of virus replication. Indeed, comprehensive single-cell characterization approaches suggest


that actively transcribing and translating reservoirs are better targeted for clearance by host
immune pressures [33••]. Some expanded HIV + T cell clones, however, persist for years
despite being transcriptionally active [33••], suggesting cellular persistence and proliferation
outcompete immune clearance in the majority of PLWH. These HIV-1 viral products within
the blood and lymphoid tissues reflect that activity of the reservoir and correlate with
time to viral rebound [4, 34]. The relative contributions of transcriptionally silent DNA
proviruses, actively transcribing infected cells, or virions in other forms (e.g., trapped within
the follicular dendritic cell network) to the pool of rebound-competent viruses remain an
important topic of study [35•].

Characterization of the other extreme of the viral reservoir, deeply latent proviruses, has
Author Manuscript

also informed our understanding of virus persistence. Studies of the proviral landscape
of elite controllers (ECs), a rare subset of PLWH who maintain viral suppression in the
absence of ART [36], demonstrate that ECs in general have less frequent, but detectable,
replication-competent proviruses [37, 38]. Importantly, the proviruses in ECs are enriched in
regions of heterochromatin and exhibit long distances to the nearest transcription start site
[39••], suggesting a lower likelihood of reactivation in vivo. These deeply latent viruses may
also be enriched in some PLWH on long-term suppressive ART and appear less responsive
to activation and latency reversal [33••]. The enrichment of deeply latent viruses in ECs and
after long-term ART supports the hypothesis that effective anti-HIV-1 immune responses can
clear more transcriptionally active proviruses [40••, 41] and has spurred efforts to augment
host immune responses to enhance reservoir clearance [36, 42]
Author Manuscript

The Challenge of Measuring a Highly Defective Proviral Reservoir


Attempts to accurately measure heterogeneous cellular reservoirs are further hampered by
the extent to which provirus is defective. Proviral DNA genomes are rendered replication-
incompetent by large deletions, incomplete viral coding sequences, and apolipoprotein
B mRNA editing enzyme catalytic polypeptide-like 3G (APOBEC3G) hypermutation [3,
4, 43–45, 46••, 47]. Though unable to reactivate, some of these defective genomes are
capable of viral protein expression, and thus serve as potential sources of pathogenic

Curr HIV/AIDS Rep. Author manuscript; available in PMC 2023 August 22.
Kreider and Bar Page 4

inflammation [48, 49]. In many cohorts, defective proviruses comprise more than 95%
Author Manuscript

of all integrated viral genomes [40••, 50], complicating measurements of total versus
functional virus. Indeed, for years, the field lacked reservoir measurement assays that
were both feasible and accurate. Long considered the gold standard, the quantitative viral
outgrowth assay (QVOA) is now understood to provide a minimum estimate of replication-
competent virus using ex vivo stimulation of longlived CD4+ T cells [51, 52], as repetitive
stimulations yield increasing numbers of virus-producing cells [13, 53••]. Furthermore,
QVOA is resource-intensive and requires many input cells. Thus, many investigators have
relied on assays measuring total viral DNA, which overestimate the replication-competent
reservoir and do not consistently correlate with QVOA measures, but are high-throughput,
inexpensive, efficient across virus subtypes, and feasible with fewer cells [54•, 55•, 56,
57]. Assays measuring inducible provirus, like the Tat/Rev-Induced Limited Dilution Assay
(TILDA), which uses limiting dilution to measure the frequency of cells producing multiply-
Author Manuscript

spliced HIV RNA after stimulation, and Envelope Detection by Induced Transcription-based
Sequencing (EDITS), which uses sequencing to detect cells producing both mature env
RNA and functional HIV proteins after stimulation, provide a middle ground for stringency
and feasibility, but often correlate poorly with QVOA [52, 58–61]. Thus, methodologic
limitations have hindered accurate assessment of reservoir size in humans, particularly in the
restricted samples available in clinical trials.

Aiming for a more feasible measurement of functional viruses, two groups independently
developed novel approaches to estimate the number of proviruses with intact sequences. The
intact proviral DNA assay (IPDA) [53••] and quadruplex quantitative PCR (Q4PCR) [62]
employ three and four quantitative PCR probes at two and four genomic loci, respectively,
for a single template quantitative PCR reaction to distinguish provirus with select defects
versus those without (of note, Q4PCR adds a subsequent near-full-length sequencing step).
Author Manuscript

Notably, IPDA and Q4PCR values correlate with replication-competent measurements using
QVOA [50, 53••, 63]. Modeling of longitudinal reservoir decay kinetics by IPDA across
cohorts suggests intact sequences wane more rapidly than the minimal decline seen in
defective sequences [40••, 46••]. This pattern of intact proviral decay across cohorts further
supports evidence that cells harboring intact sequences are preferentially lost, through
immune clearance mechanisms or apoptosis of the infected cell. The high-throughput
nature and reduced input cell requirements of these quantitative approaches also make
them attractive assays for research and clinical trials [55•], while issues of efficiency and
validation across diverse HIV-1 subtypes remain to be addressed [40••, 64, 65]. Moving
forward, these estimates of the intact provirus pool are being widely employed as endpoints
in clinical trials and applied across clinical cohorts [66].
Author Manuscript

The Dynamics of Reservoir Persistence


Following ART initiation, viral and cellular reservoirs are not stagnant. While the intact
reservoir on ART declines slowly, viral persistence is characterized by co-occurring
expansion and contraction of infected CD4+ T cell populations. Building on the
identification of expanded T cell clones bearing identical proviruses at shared integration
sites [67, 68], we now infer that many proviruses sampled by QVOA and total or integrated
HIV-1 DNA assays are derived from identical T cell clones, which can undergo expansion

Curr HIV/AIDS Rep. Author manuscript; available in PMC 2023 August 22.
Kreider and Bar Page 5

either prior to or after HIV-1 infection [69], be maintained independent of viral production,
Author Manuscript

and drive the low-level viremia observed on suppressive ART [70•]. Informed by improved
sequencing and novel single-cell analysis techniques, we now understand that the relative
frequencies of the infected T cell clones shift substantially over time [3, 43, 44, 47, 53••,
67, 71–74] via several processes, including antigen-specific T cell proliferation [69, 75],
homeostatic T cell maintenance [16, 76, 77], and proto-oncogene integration effects [67, 72].
Thus, HIV-1 persistence in CD4+ T cells is a complex process driven by mechanisms closely
linked to intrinsic T cell immunity, which poses challenges to the development of safe and
parsimonious interventions to reduce clonal reservoirs.

Reservoir evaluations at key timepoints of infection, including immediately following


transmission and at ART initiation, have further shed light on reservoir dynamics. Studies
of early ART initiation in human and nonhuman primate (NHP) models have conclusively
shown that a longlived reservoir can be established within days of HIV-1 or SIV acquisition
Author Manuscript

[78–80]. Reservoir size can be markedly, but not totally, reduced with early ART initiation
[41, 81], and these reservoirs decay slowly as they become increasingly clonal over years
of suppressive ART [14, 15, 43, 82]. Recent reports from several groups have shown that
the replication-competent reservoir has a marked enrichment for viruses that are genetically
and phenotypically similar to those circulating at the time of ART initiation [83••, 84••,
85]. The bias towards later viruses suggests that antiretrovirals may be a stabilizing force
on HIV-1-infected cell survival and persistence, which is supported by studies modeling
NHP infection showing slower turnover of cells infected at ART initiation [86]. If much
of the persistent reservoir is seeded at the time of ART initiation, this juncture serves as
a window of opportunity to attempt reservoir reduction—an approach that is now being
tested in several ongoing and in-development clinical trials. Alternatively, the dearth of
early circulating proviruses may indicate preferential immune clearance of archival wild-
Author Manuscript

type viruses, resulting in the persistence of proviral escape variants [87]. Reassuringly,
ART-suppressed PLWH have been shown to retain effective HIV-specific CD8+ T cell
responses without widespread escape in sampled proviruses [87, 88]. The exact contribution
of cytolytic T cells to reservoir reduction has been challenging to measure, but recent
evaluation of acutely infected individuals suggests that greater magnitude and quality of
the cellular immune response correlates with smaller reservoir size [89]. The potential
for immune surveillance to drive preferential loss of archival viruses supports the use of
immunotherapy to enhance the potency and breadth of adaptive responses as eradication
strategies.

The Rebound-Competent Reservoir


Phenotypic studies of rebound viruses have identified selective pressures that shape
Author Manuscript

the rebound-competent reservoir and highlight processes that can be exploited for cure
interventions. Analytic treatment interruption (ATI) results in viral rebound in nearly 95%
of ART-suppressed PLWH at 12 weeks post-ART cessation [4] with multiple genetically
distinct virus populations reactivating nearly simultaneously [3, 84••, 90–92]. Notably,
these rebound viruses are both genetically and phenotypically distinct from the replication-
competent viruses sampled from the same participants’ blood on ART [3, 90, 91, 93]. One
salient distinction between rebound and reservoir viruses is their relative sensitivity to type

Curr HIV/AIDS Rep. Author manuscript; available in PMC 2023 August 22.
Kreider and Bar Page 6

I interferons [84••]. The unique resistance to interferons seen in rebound viruses suggests
Author Manuscript

that innate immunity restricts which reservoir variants can reignite systemic replication.
Accordingly, intense sampling in the period between treatment interruption and detection
of rebound viremia has revealed that plasmacytoid dendritic cell frequency and soluble
inflammatory markers both spike prior to peripheral viral RNA detection [94]. Together,
these results highlight the innate pressure within the tissues as viruses reactivate locally.
Similarly, adaptive immune responses restrict rebound, with breakthrough viruses showing
resistance to contemporaneously circulating autologous neutralizing antibodies [3, 95].
Because replicating HIV-1 is a sensitive rheostat of the cumulative selective pressures it
is experiencing, further comparison of rebound virus phenotype to that of the remainder of
the reservoir may elucidate immune responses that can be leveraged into cure strategies.

Interventions to Clear or Suppress HIV-1 Reservoirs


Author Manuscript

Broadly Neutralizing Antibody Immunotherapy—Over the last decade, the field has
collectively isolated numerous broadly neutralizing antibodies (bnAbs) capable of inhibiting
genetically diverse HIV-1 strains, a subset of which have been tested in human trials. Passive
immunization with bnAbs can protect from transmission of neutralization-sensitive viruses
[96, 97]; thus, their elicitation has been a major focus in HIV prevention efforts [98]. In
PLWH, bnAbs have been shown to decrease viremia and maintain viral suppression in
those with sensitive viral reservoirs [90, 91, 99–105], demonstrating bnAbs’ ability to act as
suppressive ART. The promise of bnAbs for HIV cure efforts, however, lies in their potential
for effector function–mediated clearance of infected cells and immunomodulation, activities
that have not yet been conclusively demonstrated in human trials.

In both human and NHP studies, antibody effector function has been associated with
improved prevention, decreased viral load, and limited immunopathogenesis (reviewed in
Author Manuscript

[106]), but bnAbs’ capacity to recognize and enhance clearance of HIV-infected cells is less
well demonstrated. Modeling studies of bnAb administration in viremic hosts demonstrate
accelerated decline in viremia as compared to small molecule ART, suggesting that bnAbs
mediate additional activities beyond cell-free virus neutralization [107]. NHP studies of
bnAbs with or without latency reversal agents have also suggested clearance of infected
cells, but these studies may not represent PLWH as they rely on extremely early ART
initiation following infection with viruses prone to spontaneous control [108, 109].

A “vaccinal effect,” or enhancement of adaptive immune responses that persist after


antibody levels wane, is well characterized in oncology [110, 111], and is implicated, but
less well substantiated with anti-HIV bnAbs [103, 112–114]. Preclinical studies have shown
possible immunomodulation from bnAbs administered with early ART. In SHIV-infected
Author Manuscript

NHPs, passive immunization can result in improved T cell functionality, decreased cellular
exhaustion, and more potent autologous nAb responses [109, 115, 116]. Perhaps most
intriguingly, early bnAb immunotherapy in SHIV-infected NHPs has been associated with
durable virus control [109, 117, 118]. Prolonged virus suppression has also been seen post-
bnAb administration in small human studies [99], but the bnAb-specific contribution and
mechanism of action in human and NHP post-intervention control is yet to be determined.

Curr HIV/AIDS Rep. Author manuscript; available in PMC 2023 August 22.
Kreider and Bar Page 7

Despite excitement, concerns surrounding bnAb use remain. Most daunting, viral escape
Author Manuscript

variants have been described for every class of antibody and there exists no reliable way
to screen the entire viral archive for resistance. The field continues to develop more broad
and potent agents and combination therapy to attempt to overcome these challenges. Thus,
despite these caveats, bnAb immunotherapy holds promise and is being tested in multiple
clinical trials of combination cure strategies.

Latency Reversal and Immune Enhancement


Multiple strategies seek to enhance host immune responses to induce more effective
clearance and suppression of the HIV-1 reservoir in a broad range of PLWH. Given the
heterogeneity of the reservoir, combination strategies are likely needed, including latent
virus reactivation, suppression of virus replication, and improved cellular clearance. Many
promising agents have demonstrated efficacy in oncology, where the life-threatening nature
Author Manuscript

of cancer allows tolerance of drug-related toxicities. Paramount to the repurposing of these


immunomodulatory agents to HIV cure is demonstrating safety in otherwise healthy PLWH.

A long-standing goal of the cure field is the development of safe and effective latency
reversal agents, or LRAs, to specifically induce latently infected cells to reactivate, thus
exposing them to immune clearance. The first-generation LRAs to reach clinical trials were
largely histone deacetylase inhibitors (HDACi). These agents demonstrated safety and in
vitro activity but failed to induce consistent on-ART viremia or reductions in reservoir
measures [119–122]. Recent clinical trials of HDACi in combination with therapeutic
vaccines [123] and bnAbs [124], failed to meet their primary outcomes of decreases
in reservoir measures or time to rebound, respectively. Newer LRAs, including second
mitochondrial inhibitor of caspase (SMAC) mimetics, which target noncanonical NF-kB
signaling, and antibodies that transiently deplete CD8+ T cells [125–127], have shown
Author Manuscript

more robust and reliable latency reversal across animal models [128, 129], and are
poised for human studies. Other agents aim to induce both latency reversal and enhanced
immune function; furthest in clinical development are IL-15 superagonists, Toll-like receptor
agonists, and immune checkpoint inhibitors.

The IL-15 superagonist N-803 is a fusion protein of mutant IL-15 and a dimeric IL-15
receptor αSu/Fc developed for enhanced pharmacokinetics and tissue distribution [130,
131]. In vitro use has shown latency reversal within primary cell models of HIV-1 latency
[132]. NHP data suggest enhanced cellular immune activity and migration to key tissue
sites [127, 133], but limited on-ART viremia when used independently. Notably, when
coupled with depletion of CD8+ T cells, LRA activity was markedly increased [126,
127]. N-803 has shown promise in several phase 1 cancer trials [134–136], and the first-
Author Manuscript

in-PLWH study demonstrated a reasonable safety profile, with injection site reactions and
lymphadenopathy as the primary toxicities [137]. In exploratory analyses, investigators
observed early increases in HIV + CD4 T cells as measured by IPDA and EDITS, followed
by decreases in the size of the inducible reservoir as measured by EDITS. Building on
this demonstration of tolerability and possible biologic activity, multiple phase 1 studies are
currently testing N-803 in combination with early ART initiation, bnAbs, and therapeutic
vaccination.

Curr HIV/AIDS Rep. Author manuscript; available in PMC 2023 August 22.
Kreider and Bar Page 8

Agents targeting the Toll-like receptors (TLRs), which are pathogen pattern recognition
Author Manuscript

receptors central to innate antiviral immunity, have also demonstrated different degrees
of LRA activity in preclinical studies. In S(H)IV-infected rhesus macaques (RM), TLR-7
agonists have demonstrated transient on-ART viremia and delays in rebound in some, but
not all studies [138, 139]. Similar variability was shown across studies of early ART-treated
S(H)IV-infected RM, where TLR-7 agonists, in combination with vaccination or bnAbs,
failed to induce on-ART viremia, but led to substantial delays in time to rebound [108, 140].
Extension of this strategy to a more robust infection model showed more modest effects
[141]. In a recent phase I trial in PLWH who controlled viremia to low levels prior to ART, a
TLR-7 agonist showed enhanced interferon responses and modest decreases in the reservoir
as measured by IPDA [142]. As with IL-15 superagonists, TLR agonists in combination
with other agents are currently being tested in humans, trials that will shed light on whether
this form of innate immune engagement can enhance reservoir clearance.
Author Manuscript

Another key focus of immunotherapy are checkpoint inhibitors (CPI), because HIV
reservoirs are known to be enriched in cells expressing immune checkpoint receptors and
CPIs have shown remarkable immunomodulation and improved outcomes in oncology [143,
144]. In NHP models of HIV-1 persistence, several CPIs have shown efficacy. Studies
testing anti-PD-1 and anti-CTLA-4 in SIV-infected RM demonstrate marked increases in
cellular cycling and differentiation and consistent LRA activity [145]. These CPIs have
been tested in pilot studies of people living with HIV and cancer, showing promising
reservoir reductions in these unique patient populations [146••]. In healthy PLWH, however,
phase 1 trials of ant-PD-1 or anti-PDL1 have been stopped due to immune-mediated
toxicities, highlighting concerns that the therapeutic index for many CPI approaches may
not be suitable for otherwise healthy PLWH. Thus, the potential of CPIs may depend on
development of safer interventions that balance immune activity with toxicities.
Author Manuscript

Cellular and Gene Therapy


Novel approaches in cellular and gene therapy have advanced in multiple fields to the
point of clinical testing or clinical efficacy. One promising approach adopted from oncology
is engineered chimeric antigen receptor T (CAR T) cells, which have shown remarkable
success in the treatment of hematologic malignancies, including cancer remission over 10
years post-treatment [147]. CAR T cells have long been studied as retroviral eradication
strategies given their ability to recognize and kill infected cells, but HIV-1 poses distinct
challenges compared to antigen-rich hematologic malignancies. Lentiviral-specific CAR T
cells potently kill cells infected with diverse HIV-1 isolates [148•, 149, 150], with newer
generations capable of peripheral viral clearance in humanized mouse models of infection
[149, 151]. Unfortunately, the same efficacy has been more challenging in NHP models.
Author Manuscript

While issues of engraftment may be overcome by vaccination with the CAR’s antigen
[152] or direct modification of hematopoietic stem cells to induce CAR expression in all
progeny [153, 154], anti-CAR humoral responses [155] and inability to mediate SHIV/SIV
containment in NHPs [156] remain major hurdles. Following technological improvements
in strain cross-reactivity and co-stimulatory signals, HIV-1 CARs have recently reentered
human clinical testing [157]. While multiple constructs are currently being evaluated [158],
a recent trial employing a bnAb-based CAR reported the preliminary result of declines in

Curr HIV/AIDS Rep. Author manuscript; available in PMC 2023 August 22.
Kreider and Bar Page 9

CA-RNA and intact proviral DNA coincident with CAR T cell infusion, as well as modestly
Author Manuscript

delayed time to rebound compared to matched historical controls [159].

Genomic engineering is also being employed to directly inactivate proviruses, bypassing


the need for host immune effectors. One promising approach is the expression of HIV-1-
specific recombinases. Because the proviral sequence is flanked by identical long terminal
repeats, or LTRs, recombinases can mediate near-full-length provirus excision. Although
early iterations of designer recombinases recognized a small subset of isolates [160],
subsequent efforts yielded Brec1, a Cre recombinase that recognizes a 34-nucleotide span
around the polyA stem loop present in 90% of subtype A, B, and C isolates [161]. While
Brec1 demonstrates in vitro recombination activity and mediates clearance of viral RNA in
a humanized mouse model [161, 162], some concerns about nonspecific Brec1 substrate
activity have been reported [163]. This approach, while elegant, is less flexible than
others given the difficulty in adjusting the recombinase recognition sequence and would
Author Manuscript

be rendered useless by anti-drug adaptive immune responses.

Alternatively, proviral inactivation has been achieved using the readily adaptable clustered
regularly interspaced short palindromic repeats/CRISPR associated protein 9 (CRISPR/
cas9) nuclease system for lentiviral genomic editing [164]. CRISPR/cas9 approaches have
mediated viral restriction in vitro, ex vivo, and in NHP models [165, 166•, 167], by targeting
cas9 endonuclease activity with guide RNAs specific to conserved HIV sequence motifs,
thereby damaging or excising HIV proviral genes. Rapid viral mutational escape has been
seen with the use of single guide RNAs [168–172]; however, viral escape can be overcome
by using multiple guides, resulting in greater in vitro editing efficiency [173, 174]. More
than half of published strategies target the LTR, which offers benefit over coding sequence
targeting; as LTR function depends on nucleic acid sequence, the LTR is highly conserved,
Author Manuscript

and each provirus is flanked by two LTRs, offering double the targets and possible proviral
excision. Despite theoretical benefits, indels are more frequent within LTRs as compared
to coding sequences (unpublished observation, E. Kreider, W. Fischer, B. Korber, and
G. Shaw). Unlike other engineered nucleases, panels of guide RNAs targeting conserved
regions can be rapidly generated that offer broad inter-subtype cross-reactivity [175–178].
These findings support the notion that in vivo editing of the proviral pool is a tenable
approach to reservoir clearance. Whether CRISPR/cas9 delivery, efficiency, and specificity
can overcome the obstacles of proviral DNA reservoir eradication in humans remains to be
seen.

Conclusions
The rebound-competent HIV-1 reservoir that persists through suppressive ART remains the
Author Manuscript

ultimate barrier to drug-free virus remission. Recent work has highlighted the multiple
processes that shape viral persistence, including the spectrum of cell types, locations,
and activation states comprising the reservoir; the highly defective nature of the proviral
population; and the expansion and contraction of host cells that themselves play important
immunologic roles. These findings reinforce the consistent theme that intact proviruses
decline over time, which supports ongoing development of cure strategies that build on
these reservoir clearing processes. In the coming years, deep interrogation of preclinical

Curr HIV/AIDS Rep. Author manuscript; available in PMC 2023 August 22.
Kreider and Bar Page 10

animal studies [179–181] and human clinical trials that seek to accelerate reservoir decay
Author Manuscript

or eradicate DNA proviruses will help identify promising cure interventions and further our
understanding of reservoir persistence (Fig. 1).

Acknowledgments
Funding EFK is supported by T32 AI118684–05 and BEAT-HIV Delaney Collaboratory UM1 AI164570; KJB is
supported by the Penn CFAR P30 AI 045008; P01 AI131338; R01 AI62646; R01 MH128155; UM1 AI068636; and
BEAT-HIV Delaney Collaboratory UM1 AI164570.

Human and Animal Rights and Informed Consent All reported studies/ experiments with human or animal
subjects performed by the authors have been previously published and complied with all applicable ethical
standards (including the Helsinki declaration and its amendments, institutional/national research committee
standard, and international/ national/institutional guidelines).

References
Author Manuscript

Papers of particular interest, published recently, have been highlighted as: • Of importance

•• Of major importance

1. Marcus JL, Leyden WA, Alexeeff SE, Anderson AN, Hechter RC, Hu H, et al. Comparison
of overall and comorbidity-free life expectancy between insured adults with and without HIV
infection, 2000–2016. Jama Netw Open. 2020;3:e207954.
2. Rodger AJ, Lodwick R, Schechter M, Deeks S, Amin J, Gilson R, et al. Mortality in well controlled
HIV in the continuous antiretroviral therapy arms of the SMART and ESPRIT trials compared with
the general population. AIDS. 2013;27:973–9. [PubMed: 23698063]
3. Salantes DB, Zheng Y, Mampe F, Srivastava T, Beg S, Lai J, et al. HIV-1 latent reservoir size
and diversity are stable following brief treatment interruption. J Clin Invest. 2018;128:3102–15.
[PubMed: 29911997]
4. Li JZ, Etemad B, Ahmed H, Aga E, Bosch RJ, Mellors JW, et al. The size of the expressed
HIV reservoir predicts timing of viral rebound after treatment interruption. AIDS. 2016;30:343–53.
Author Manuscript

[PubMed: 26588174]
5. Wood BR, Huhn GD. Excess weight gain with integrase inhibitors and tenofovir alafenamide: what
is the mechanism and does it matter? Open Forum Infect Dis. 2021;8:ofab542.
6. Grant PM, Cotter AG. Tenofovir and bone health. Curr Opin Hiv Aids. 2016;11:326–32. [PubMed:
26859637]
7. Vivithanaporn P, Kongratanapasert T, Suriyapakorn B, Songkunlertchai P, Mongkonariyawong P,
Limpikirati PK, et al. Potential drug-drug interactions of antiretrovirals and antimicrobials detected
by three databases. Sci Rep-uk. 2021;11:6089.
8. Stolbach A, Paziana K, Heverling H, Pham P. A review of the toxicity of HIV medications II:
interactions with drugs and complementary and alternative medicine products. J Medical Toxicol.
2015;11:326–41.
9. Iacob SA, Iacob DG, Jugulete G. Improving the adherence to antiretroviral therapy, a difficult but
essential task for a successful HIV treatment—clinical points of view and practical considerations.
Front Pharmacol. 2017;8:831. [PubMed: 29218008]
Author Manuscript

10. Sullivan PS, Johnson AS, Pembleton ES, Stephenson R, Justice AC, Althoff KN, et al.
Epidemiology of HIV in the USA: epidemic burden, inequities, contexts, and responses. Lancet.
2021;397:1095–106. [PubMed: 33617774]
11. Shrestha RK, Schommer JC, Taitel MS, Garza OW, Camp NM, Akinbosoye OE, et al. Costs and
cost-effectiveness of the patient-centered HIV care model: a collaboration between community-
based pharmacists and primary medical providers. Jaids J Acquir Immune Defic Syndromes.
2020;85:e48–54.

Curr HIV/AIDS Rep. Author manuscript; available in PMC 2023 August 22.
Kreider and Bar Page 11

12. Chaillon A, Gianella S, Dellicour S, Rawlings SA, Schlub TE, Oliveira MFD, et al. HIV
persists throughout deep tissues with repopulation from multiple anatomical sources. J Clin Invest.
Author Manuscript

2020;130:1699–712. [PubMed: 31910162]


13. Ho Y-C, Shan L, Hosmane NN, Wang J, Laskey SB, Rosenbloom DIS, et al. Replication-
competent noninduced proviruses in the latent reservoir increase barrier to HIV-1 cure. Cell.
2013;155:540–51. [PubMed: 24243014]
14. Crooks AM, Bateson R, Cope AB, Dahl NP, Griggs MK, Kuruc JD, et al. Precise Quantitation of
the latent HIV-1 reservoir: implications for eradication strategies. J Infect Dis. 2015;212:1361–5.
[PubMed: 25877550]
15. Siliciano JD, Kajdas J, Finzi D, Quinn TC, Chadwick K, Margolick JB, et al. Long-term follow-up
studies confirm the stability of the latent reservoir for HIV-1 in resting CD4+ T cells. Nat Med.
2003;9:727–8. [PubMed: 12754504]
16. Chomont N, El-Far M, Ancuta P, Trautmann L, Procopio FA, Yassine-Diab B, et al. HIV
reservoir size and persistence are driven by T cell survival and homeostatic proliferation. Nat
Med. 2009;15:893–900. [PubMed: 19543283]
17. Buzon MJ, Sun H, Li C, Shaw A, Seiss K, Ouyang Z, et al. HIV-1 persistence in CD4+ T cells with
Author Manuscript

stem cell-like properties. Nat Med. 2014;20:139–42. [PubMed: 24412925]


18. Duette G, Hiener B, Morgan H, Mazur FG, Mathivanan V, Horsburgh BA, et al. The HIV-1
proviral landscape reveals that Nef contributes to HIV-1 persistence in effector memory CD4+ T
cells. J Clin Invest. 2022;132(7):e154422. 10.1172/JCI154422.
19. Pardons M, Baxter AE, Massanella M, Pagliuzza A, Fromentin R, Dufour C, et al. Single-
cell characterization and quantification of translation-competent viral reservoirs in treated and
untreated HIV infection. Plos Pathog. 2019;15:e1007619.
20. Grau-Expósito J, Serra-Peinado C, Miguel L, Navarro J, Curran A, Burgos J, et al. A novel
single-cell FISH-flow assay identifies effector memory CD4(+) T cells as a major niche for HIV-1
transcription in HIV-infected patients. MBio. 2017;8:e00876-e917.
21. Fromentin R, Bakeman W, Lawani MB, Khoury G, Hartogensis W, DaFonseca S, et al. CD4+ T
cells expressing PD-1, TIGIT and LAG-3 contribute to HIV persistence during ART. Plos Pathog.
2016;12:e1005761.
22. McGary CS, Deleage C, Harper J, Micci L, Ribeiro SP, Paganini S, et al. CTLA-4+PD-1−
memory CD4+ T cells critically contribute to viral persistence in antiretroviral therapy-suppressed,
Author Manuscript

SIV-infected rhesus macaques. Immunity. 2017;47:776–788.e5. [PubMed: 29045906]


23. Fromentin R, Chomont N. HIV persistence in subsets of CD4+ T cells: 50 shades of reservoirs.
Semin Immunol. 2020;51:101438.
24. Castellano P, Prevedel L, Eugenin EA. HIV-infected macrophages and microglia that survive acute
infection become viral reservoirs by a mechanism involving Bim. Sci Rep-uk. 2017;7:12866.
25. Hashimoto D, Chow A, Noizat C, Teo P, Beasley MB, Leboeuf M, et al. Tissue-resident
macrophages self-maintain locally throughout adult life with minimal contribution from
circulating monocytes. Immunity. 2013;38:792–804. [PubMed: 23601688]
26. Ganor Y, Real F, Sennepin A, Dutertre C-A, Prevedel L, Xu L, et al. HIV-1 reservoirs in urethral
macrophages of patients under suppressive antiretroviral therapy. Nat Microbiol. 2019;4:633–44.
[PubMed: 30718846]
27. Honeycutt JB, Thayer WO, Baker CE, Ribeiro RM, Lada SM, Cao Y, et al. HIV persistence
in tissue macrophages of humanized myeloid only mice during antiretroviral therapy. Nat Med.
2017;23:638–43. [PubMed: 28414330]
Author Manuscript

28. Abreu C, Shirk EN, Queen SE, Beck SE, Mangus LM, Pate KAM, et al. Brain macrophages harbor
latent, infectious simian immunodeficiency virus. Aids Lond Engl. 2019;33(Suppl 2):S181–8.
29. Wong ME, Jaworowski A, Hearps AC. The HIV reservoir in monocytes and macrophages. Front
Immunol. 2019;10:1435. [PubMed: 31297114]
30. TR study group, Colby DJ, Trautmann, Pinyakorn S, Leyre L, Pagliuzza A, et al. Rapid HIV RNA
rebound after antiretroviral treatment interruption in persons durably suppressed in Fiebig I acute
HIV infection. Nat Med. 2018;24:923–6. [PubMed: 29892063]

Curr HIV/AIDS Rep. Author manuscript; available in PMC 2023 August 22.
Kreider and Bar Page 12

31. Estes JD, Kityo C, Ssali F, Swainson L, Makamdop KN, Prete GQD, et al. Defining total-body
AIDS-virus burden with implications for curative strategies. Nat Med. 2017;23:1271–6. [PubMed:
Author Manuscript

28967921]
32. Buzón MJ, Massanella M, Llibre JM, Esteve A, Dahl V, Puertas MC, et al. HIV-1 replication and
immune dynamics are affected by raltegravir intensification of HAART-suppressed subjects. Nat
Med. 2010;16:460–5. [PubMed: 20228817]
33. Einkauf KB, Osborn MR, Gao C, Sun W, Sun X, Lian X, et al. Parallel analysis of transcription,
integration, and sequence of single HIV-1 proviruses. Cell. 2022;185:266–282.e15. [PubMed:
35026153] •• Novel single-cell approach to study the reservoir, highlighting the decay of actively
transcribing reservoir and expansion of certain T cell clones.
34. Pasternak AO, Grijsen ML, Wit FW, Bakker M, Jurriaans S, Prins JM, et al. Cell-associated HIV-1
RNA predicts viral rebound and disease progression after discontinuation of temporary early ART.
JCI Insight. 2020;5:e134196. 10.1172/jci.insight.134196.
35. Cohn LB, Chomont N, Deeks SG. The biology of the HIV-1 latent reservoir and implications
for cure strategies. Cell Host Microbe. 2020;27:519–30. [PubMed: 32272077] • A recent,
comprehensive review of the HIV-1 reservoir.
Author Manuscript

36. Li JZ, Blankson JN. How elite controllers and posttreatment controllers inform our search for an
HIV-1 cure. J Clin Invest. 2021;131:e149414. 10.1172/JCI149414.
37. Woldemeskel BA, Kwaa AK, Blankson JN. Viral reservoirs in elite controllers of HIV-1 infection:
implications for HIV cure strategies. Ebiomedicine. 2020;62:103118.
38. Turk G, Seiger K, Lian X, Sun W, Parsons EM, Gao C, et al. A Possible Sterilizing Cure
of HIV-1 Infection Without Stem Cell Transplantation. Ann Intern Med. 2022;175(1):95–100.
10.7326/L21-0297. [PubMed: 34781719]
39. Jiang C, Lian X, Gao C, Sun X, Einkauf KB, Chevalier JM, et al. Distinct viral reservoirs in
individuals with spontaneous control of HIV-1. Nature. 2020;585:261–7. [PubMed: 32848246] ••
Single-cell analyses of elite controllers identifying deeply latent proviruses, supporting effective
immune clearance of HIV-1 reservoir.
40. Gandhi RT, Cyktor JC, Bosch RJ, Mar H, Laird GM, Martin A, et al. Selective decay of intact
HIV-1 proviral DNA on antiretroviral therapy. J Infect Dis. 2020;223:225–33. •• One of recent
studies applying IPDA to show decline in intact reservoir over time on ART.
41. Leyre L, Kroon E, Vandergeeten C, Sacdalan C, Colby DJ, Buranapraditkun S, et al. Abundant
Author Manuscript

HIV-infected cells in blood and tissues are rapidly cleared upon ART initiation during acute HIV
infection. Sci Transl Med. 2020;12:eaav3491.
42. Hartana CA, Yu XG. Immunological effector mechanisms in HIV-1 elite controllers. Curr Opin
Hiv Aids. 2021;16:243–8. [PubMed: 34270465]
43. Cohn LB, Silva IT, Oliveira TY, Rosales RA, Parrish EH, Learn GH, et al. HIV-1 integration
landscape during latent and active infection. Cell. 2015;160:420–32. [PubMed: 25635456]
44. Bruner KM, Murray AJ, Pollack RA, Soliman MG, Laskey SB, Capoferri AA, et al. Defective
proviruses rapidly accumulate during acute HIV-1 infection. Nat Med. 2016;22:1043–9. [PubMed:
27500724]
45. Bender AM, Simonetti FR, Kumar MR, Fray EJ, Bruner KM, Timmons AE, et al. The landscape
of persistent viral genomes in ART-treated SIV, SHIV, and HIV-2 infections. Cell Host Microbe.
2019;26:73–85.e4. [PubMed: 31295427]
46. Peluso MJ, Bacchetti P, Ritter KD, Beg S, Lai J, Martin JN, et al. Differential decay of intact and
defective proviral DNA in HIV-1-infected individuals on suppressive antiretroviral therapy. JCI
Author Manuscript

Insight. 2020;5(4):e132997. 10.1172/jci.insiht.132997. •• One of recent studies applying IPDA to


show decline in intact reservoir over time on ART.
47. Wang Z, Gurule EE, Brennan TP, Gerold JM, Kwon KJ, Hosmane NN, et al. Expanded cellular
clones carrying replicationcompetent HIV-1 persist, wax, and wane. Proc National Acad Sci.
2018;115:201720665.
48. Imamichi H, Smith M, Adelsberger JW, Izumi T, Scrimieri F, Sherman BT, et al. Defective HIV-1
proviruses produce viral proteins. Proc National Acad Sci. 2020;117:3704–10.

Curr HIV/AIDS Rep. Author manuscript; available in PMC 2023 August 22.
Kreider and Bar Page 13

49. Pollack RA, Jones RB, Pertea M, Bruner KM, Martin AR, Thomas AS, et al. Defective HIV-1
proviruses are expressed and can be recognized by cytotoxic T lymphocytes, which shape the
Author Manuscript

proviral landscape. Cell Host Microbe. 2017;21:494506.e4.


50. Gaebler C, Falcinelli SD, Stoffel E, Read J, Murtagh R, Oliveira TY, et al. Sequence
Evaluation and Comparative Analysis of Novel Assays for Intact Proviral HIV-1 DNA. J Virol.
2021;95(6):e01986–20. 10.1128/JVI.01986-20.
51. Siliciano JD, Siliciano RF. Enhanced culture assay for detection and quantitation of latently
infected, resting CD4+ T-cells carrying replication-competent virus in HIV-1-infected individuals.
Methods Mol Biol Clifton N J. 2005;304:3–15.
52. Stone M, Rosenbloom D, Bacchetti P, Deng X, Dimapasoc M, Keating S, et al. Assessing
suitability of next-generation viral outgrowth assays as proxies for classic QVOA to measure
HIV-1 latent reservoir size. J Infect Dis. 2020;224:1209–18.
53. Bruner KM, Wang Z, Simonetti FR, Bender AM, Kwon KJ, Sengupta S, et al. A quantitative
approach for measuring the reservoir of latent HIV-1 proviruses. Nature. 2019;566:120–5.
[PubMed: 30700913] •• Development and validation of the intact proviral DNA assay.
54. Deeks SG, Archin N, Cannon P, Collins S, Jones RB, de Jong MAWP, et al. Research
Author Manuscript

priorities for an HIV cure: International AIDS Society Global Scientific Strategy 2021. Nat
Med. 2021;27:2085–98. [PubMed: 34848888] • A position piece put forth by leaders in the field
outlining the research priorities.
55. Abdel-Mohsen M, Richman D, Siliciano RF, Nussenzweig MC, Howell BJ, Martinez-Picado J, et
al. Recommendations for measuring HIV reservoir size in cure-directed clinical trials. Nat Med.
2020;26:1339–50. [PubMed: 32895573] • Comprehensive assessment of reservoir measurement
methods and recommendations for evaluation of clinical trials.
56. Spina CA, Anderson J, Archin NM, Bosque A, Chan J, Famiglietti M, et al. An in-depth
comparison of latent HIV-1 reactivation in multiple cell model systems and resting CD4+ T cells
from aviremic patients. Plos Pathog. 2013;9:e1003834.
57. Eriksson S, Graf EH, Dahl V, Strain MC, Yukl SA, Lysenko ES, et al. Comparative analysis of
measures of viral reservoirs in HIV-1 eradication studies. Plos Pathog. 2013;9:e1003174.
58. Procopio FA, Fromentin R, Kulpa DA, Brehm JH, Bebin A-G, Strain MC, et al. A novel assay to
measure the magnitude of the inducible viral reservoir in HIV-infected individuals. EBioMedicine.
2015;2:874–83. [PubMed: 26425694]
Author Manuscript

59. Lungu C, Banga R, Gruters RA, Procopio FA. Inducible HIV-1 reservoir quantification: clinical
relevance, applications and advancements of TILDA. Front Microbiol. 2021;12:686690.
60. Rosenbloom DIS, Bacchetti P, Stone M, Deng X, Bosch RJ, Richman DD, et al. Assessing
intra-lab precision and inter-lab repeatability of outgrowth assays of HIV-1 latent reservoir size.
Plos Comput Biol. 2019;15:e1006849.
61. Nguyen K, Das B, Dobrowolski C, Karn J. Multiple histone lysine methyltransferases are required
for the establishment and maintenance of HIV-1 latency. MBio. 2017;8:e00133–e217. [PubMed:
28246360]
62. Gaebler C, Lorenzi JCC, Oliveira TY, Nogueira L, Ramos V, Lu C-L, et al. Combination of
quadruplex qPCR and next-generation sequencing for qualitative and quantitative analysis of the
HIV-1 latent reservoir. J Exp Medicine. 2019;216:2253–64.
63. Falcinelli SD, Kilpatrick KW, Read J, Murtagh R, Allard B, Ghofrani S, et al. Longitudinal
dynamics of intact HIV proviral DNA and outgrowth virus frequencies in a cohort of individuals
receiving antiretroviral therapy. J Infect Dis. 2020;224:92–100.
Author Manuscript

64. Simonetti FR, White JA, Tumiotto C, Ritter KD, Cai M, Gandhi RT, et al. Intact proviral DNA
assay analysis of large cohorts of people with HIV provides a benchmark for the frequency and
composition of persistent proviral DNA. Proc National Acad Sci. 2020;117:18692–700.
65. Kinloch NN, Ren Y, Alberto WDC, Dong W, Khadka P, Huang SH, et al. HIV-1 diversity
considerations in the application of the Intact Proviral DNA Assay (IPDA). Nat Commun.
2021;12:165. [PubMed: 33420062]
66. Margolis DM. Measuring the haystack’s needles. J Infect Dis. 2020;223:184–6.

Curr HIV/AIDS Rep. Author manuscript; available in PMC 2023 August 22.
Kreider and Bar Page 14

67. Wagner TA, McLaughlin S, Garg K, Cheung CYK, Larsen BB, Styrchak S, et al. Proliferation
of cells with HIV integrated into cancer genes contributes to persistent infection. Science.
Author Manuscript

2014;345:570–3. [PubMed: 25011556]


68. Schröder ARW, Shinn P, Chen H, Berry C, Ecker JR, Bushman F. HIV-1 integration in the human
genome favors active genes and local hotspots. Cell. 2002;110:521–9. [PubMed: 12202041]
69. Simonetti FR, Zhang H, Soroosh GP, Duan J, Rhodehouse K, Hill AL, et al. Antigen-driven
clonal selection shapes the persistence of HIV-1-infected CD4+ T cells in vivo. J Clin Invest.
2021;131(3):e145254. 10.1172/JCI145254.
70. Halvas EK, Joseph KW, Brandt LD, Guo S, Sobolewski MD, Jacobs JL, et al. HIV-1 viremia not
suppressible by antiretroviral therapy can originate from large T cell clones producing infectious
virus. J Clin Invest. 2020;130:5847–57 [PubMed: 33016926] .• Linked low-level viremia on ART
to HIV-1 reservoir clone expansion.
71. Cohn LB, da Silva IT, Valieris R, Huang AS, Lorenzi JCC, Cohen YZ, et al. Clonal CD4+ T
cells in the HIV-1 latent reservoir display a distinct gene profile upon reactivation. Nat Med.
2018;24:604–9. [PubMed: 29686423]
72. Maldarelli F, Wu X, Su L, Simonetti FR, Shao W, Hill S, et al. Specific HIV integration sites are
Author Manuscript

linked to clonal expansion and persistence of infected cells. Science. 2014;345:179–83. [PubMed:
24968937]
73. Vibholm LK, Lorenzi JCC, Pai JA, Cohen YZ, Oliveira TY, Barton JP, et al. Characterization of
intact proviruses in blood and lymph node from HIV-infected individuals undergoing analytical
treatment interruption. J Virol. 2019;93:e01920–e2018. [PubMed: 30700598]
74. Lorenzi JCC, Cohen YZ, Cohn LB, Kreider EF, Barton JP, Learn GH, et al. Paired quantitative
and qualitative assessment of the replication-competent HIV-1 reservoir and comparison with
integrated proviral DNA. Proc National Acad Sci. 2016;113:E7908–16.
75. Mendoza P, Jackson JR, Oliveira TY, Gaebler C, Ramos V, Caskey M, et al. Antigen-responsive
CD4+ T cell clones contribute to the HIV-1 latent reservoir. J Exp Med. 2020;217:e20200051.
76. Bosque A, Famiglietti M, Weyrich AS, Goulston C, Planelles V. Homeostatic proliferation fails
to efficiently reactivate HIV-1 latently infected central memory CD4+ T cells. Plos Pathog.
2011;7:e1002288.
77. McManus WR, Bale MJ, Spindler J, Wiegand A, Musick A, Patro SC, et al. HIV-1 in lymph nodes
is maintained by cellular proliferation during antiretroviral therapy. J Clin Invest. 2019;129:4629–
Author Manuscript

42. [PubMed: 31361603]


78. Henrich TJ, Hatano H, Bacon O, Hogan LE, Rutishauser R, Hill A, et al. HIV-1 persistence
following extremely early initiation of antiretroviral therapy (ART) during acute HIV-1 infection:
an observational study. Plos Med. 2017;14:e1002417.
79. Whitney JB, Hill AL, Sanisetty S, Penaloza-MacMaster P, Liu J, Shetty M, et al. Rapid seeding
of the viral reservoir prior to SIV viraemia in rhesus monkeys. Nature. 2014;512:74–7. [PubMed:
25042999]
80. Whitney JB, Lim S-Y, Osuna CE, Kublin JL, Chen E, Yoon G, et al. Prevention of SIVmac251
reservoir seeding in rhesus monkeys by early antiretroviral therapy. Nat Commun. 2018;9:5429.
[PubMed: 30575753]
81. Buzon MJ, Martin-Gayo E, Pereyra F, Ouyang Z, Sun H, Li JZ, et al. Long-term antiretroviral
treatment initiated at primary HIV-1 infection affects the size, composition, and decay kinetics of
the reservoir of HIV-1-infected CD4 T cells. J Virol. 2014;88:10056–65. [PubMed: 24965451]
82. Bachmann N, von Siebenthal C, Vongrad V, Turk T, Neumann K, Beerenwinkel N, et al.
Author Manuscript

Determinants of HIV-1 reservoir size and long-term dynamics during suppressive ART. Nat
Commun. 2019;10:3193. [PubMed: 31324762]
83. Abrahams M-R, Joseph SB, Garrett N, Tyers L, Moeser M, Archin N, et al. The replication-
competent HIV-1 latent reservoir is primarily established near the time of therapy initiation. Sci
Transl Med. 2019;11:eaaw5589. •• Demonstrated enrichment of viruses circulating at time of ART
initiation in replication-competent reservoir.
84. Gondim MVP, Sherrill-Mix S, Bibollet-Ruche F, Russell RM, Trimboli S, Smith AG, et al.
Heightened resistance to host type 1 interferons characterizes HIV-1 at transmission and after

Curr HIV/AIDS Rep. Author manuscript; available in PMC 2023 August 22.
Kreider and Bar Page 15

antiretroviral therapy interruption. Sci Transl Med. 2021;13:eabd8179. •• Described substantial


IFN I-resistance of HIV-1 at rebound, suggesting innate immune pressure at this juncture.
Author Manuscript

85. Brodin J, Zanini F, Thebo L, Lanz C, Bratt G, Neher RA, et al. Establishment and stability of the
latent HIV-1 DNA reservoir. Elife. 2016;5:e18889.
86. Reece J, Petravic J, Balamurali M, Loh L, Gooneratne S, Rose RD, et al. An “escape clock” for
estimating the turnover of SIV DNA in resting CD4+ T cells. Plos Pathog. 2012;8:e1002615.
87. Deng K, Pertea M, Rongvaux A, Wang L, Durand CM, Ghiaur G, et al. Broad CTL response is
required to clear latent HIV-1 due to dominance of escape mutations. Nature. 2015;517:381–5.
[PubMed: 25561180]
88. Warren JA, Zhou S, Xu Y, Moeser MJ, MacMillan DR, Council O, et al. The HIV-1 latent reservoir
is largely sensitive to circulating T cells. Elife. 2020;9:e57246.
89. Takata H, Buranapraditkun S, Kessing C, Fletcher JLK, Muir R, Tardif V, et al. Delayed
differentiation of potent effector CD8+ T cells reducing viremia and reservoir seeding in acute
HIV infection. Sci Transl Med. 2017;9:eaag1809.
90. Scheid JF, Horwitz JA, Bar-On Y, Kreider EF, Lu C-L, Lorenzi JCC, et al. HIV-1
antibody 3BNC117 suppresses viral rebound in humans during treatment interruption. Nature.
Author Manuscript

2016;535:556–60. [PubMed: 27338952]


91. Bar KJ, Sneller MC, Harrison LJ, Justement JS, Overton ET, Petrone ME, et al. Effect of
HIV antibody VRC01 on viral rebound after treatment interruption. New Engl J Medicine.
2016;375:2037–50.
92. Rothenberger MK, Keele BF, Wietgrefe SW, Fletcher CV, Beilman GJ, Chipman JG, et al. Large
number of rebounding/founder HIV variants emerge from multifocal infection in lymphatic tissues
after treatment interruption. Proc National Acad Sci. 2015;112:E1126–34.
93. Scheerder M- AD, Vrancken B, Dellicour S, Schlub T, Lee E, Shao W, et al. HIV Rebound is
predominantly fueled by genetically identical viral expansions from diverse reservoirs. Cell Host
Microbe. 2019;26:347–358.e7. [PubMed: 31471273]
94. Mitchell JL, Takata H, Muir R, Colby DJ, Kroon E, Crowell TA, et al. Plasmacytoid dendritic cells
sense HIV replication before detectable viremia following treatment interruption. J Clin Invest.
2020;130:2845–58. [PubMed: 32017709]
95. Bertagnolli LN, Varriale J, Sweet S, Brockhurst J, Simonetti FR, White J, et al. Autologous IgG
Author Manuscript

antibodies block outgrowth of a substantial but variable fraction of viruses in the latent reservoir
for HIV-1. Proc National Acad Sci. 2020;117:32066–77.
96. Pegu A, Borate B, Huang Y, Pauthner MG, Hessell AJ, Julg B, et al. A meta-analysis of passive
immunization studies shows that serum-neutralizing antibody titer associates with protection
against SHIV challenge. Cell Host Microbe. 2019;26:336–346. e3. [PubMed: 31513771]
97. Corey L, Gilbert PB, Juraska M, Montefiori DC, Morris L, Karuna ST, et al. Two randomized
trials of neutralizing antibodies to prevent HIV-1 acquisition. New Engl J Med. 2021;384:1003–14.
[PubMed: 33730454]
98. Haynes BF, Shaw GM, Korber B, Kelsoe G, Sodroski J, Hahn BH, et al. HIV-host interactions:
implications for vaccine design. Cell Host Microbe. 2016;19:292–303. [PubMed: 26922989]
99. 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;561:479–84. [PubMed:
30258136]
100. Cale EM, Bai H, Bose M, Messina MA, Colby DJ, Sanders-Buell E, et al. Neutralizing antibody
VRC01 failed to select for HIV-1 mutations upon viral rebound. J Clin Invest. 2020;130:3299–
Author Manuscript

304. [PubMed: 32182219]


101. Caskey M, Klein F, Nussenzweig MC. Broadly neutralizing anti-HIV-1 monoclonal antibodies in
the clinic. Nat Med. 2019;25:547–53. [PubMed: 30936546]
102. Caskey M, Schoofs T, Gruell H, Settler A, Karagounis T, Kreider EF, et al. Antibody 10–
1074 suppresses viremia in HIV1-infected individuals. Nat Med. 2017;23:185–91. [PubMed:
28092665]
103. Schoofs T, Klein F, Braunschweig M, Kreider EF, Feldmann A, Nogueira L, et al. HIV-1 therapy
with monoclonal antibody 3BNC117 elicits host immune responses against HIV-1. Sci New York
N Y. 2016;352:997–1001.

Curr HIV/AIDS Rep. Author manuscript; available in PMC 2023 August 22.
Kreider and Bar Page 16

104. 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;522:487–91.
Author Manuscript

[PubMed: 25855300]
105. Lynch RM, Boritz E, Coates EE, DeZure A, Madden P, Costner P, et al. Virologic effects of
broadly neutralizing antibody VRC01 administration during chronic HIV-1 infection. Sci Transl
Med. 2015;7:319ra206–319ra206.
106. Crowley AR, Ackerman ME. Mind the gap: how interspecies variability in IgG and its
receptors may complicate comparisons of human and non-human primate effector function. Front
Immunol. 2019;10:697. [PubMed: 31024542]
107. Lu C-L, Murakowski DK, Bournazos S, Schoofs T, Sarkar D, Halper-Stromberg A, et al.
Enhanced clearance of HIV-1– infected cells by broadly neutralizing antibodies against HIV-1 in
vivo. Science. 2016;352:1001–4. [PubMed: 27199430]
108. Borducchi EN, Liu J, Nkolola JP, Cadena AM, Yu W-H, Fischinger S, et al. Antibody and TLR7
agonist delay viral rebound in SHIV-infected monkeys. Nature. 2018;563:360–4. [PubMed:
30283138]
109. Barouch DH, Whitney JB, Moldt B, Klein F, Oliveira TY, Liu J, et al. Therapeutic efficacy
Author Manuscript

of potent neutralizing HIV-1-specific monoclonal antibodies in SHIV-infected rhesus monkeys.


Nature. 2013;503:224–8. [PubMed: 24172905]
110. Cartron G, Watier H, Golay J, Solal-Celigny P. From the bench to the bedside: ways to improve
rituximab efficacy. Blood. 2004;104:2635–42. [PubMed: 15226177]
111. DiLillo DJ, Ravetch JV. Differential Fc-receptor engagement drives an anti-tumor vaccinal effect.
Cell. 2015;161:1035–45. [PubMed: 25976835]
112. Hua CK, Ackerman ME. Increasing the clinical potential and applications of anti-HIV antibodies.
Front Immunol. 2017;8:1655. [PubMed: 29234320]
113. Lambour J, Naranjo-Gomez M, Piechaczyk M, Pelegrin M. Converting monoclonal antibody-
based immunotherapies from passive to active: bringing immune complexes into play. Emerg
Microbes Infec. 2016;5:1–9.
114. Villinger F, Mayne AE, Bostik P, Mori K, Jensen PE, Ahmed R, et al. Evidence for antibody-
mediated enhancement of simian immunodeficiency virus (SIV) gag antigen processing and cross
presentation in SIV-infected rhesus macaques. J Virol. 2003;77:10–24. [PubMed: 12477806]
Author Manuscript

115. Bolton DL, Pegu A, Wang K, McGinnis K, Nason M, Foulds K, et al. Human immunodeficiency
virus type 1 monoclonal antibodies suppress acute simian-human immunodeficiency virus
viremia and limit seeding of cell-associated viral reservoirs. J Virol. 2016;90:1321–32. [PubMed:
26581981]
116. Haigwood NL, Montefiori DC, Sutton WF, McClure J, Watson AJ, Voss G, et al.
Passive immunotherapy in simian immunodeficiency virus-infected macaques accelerates the
development of neutralizing antibodies. J Virol. 2004;78:5983–95. [PubMed: 15140996]
117. Nishimura Y, Gautam R, Chun T-W, Sadjadpour R, Foulds KE, Shingai M, et al. Early
antibody therapy can induce longlasting immunity to SHIV. Nature. 2017;543:559–63. [PubMed:
28289286]
118. Shingai M, Donau OK, Plishka RJ, Buckler-White A, Mascola JR, Nabel GJ, et al. Passive
transfer of modest titers of potent and broadly neutralizing anti-HIV monoclonal antibodies block
SHIV infection in macaques. J Exp Med. 2014;211:2061–74. [PubMed: 25155019]
119. Elliott JH, Wightman F, Solomon A, Ghneim K, Ahlers J, Cameron MJ, et al. Activation of HIV
transcription with short-course vorinostat in HIV-infected patients on suppressive antiretroviral
Author Manuscript

therapy. Plos Pathog. 2014;10:e1004473.


120. Elliott JH, McMahon JH, Chang CC, Lee SA, Hartogensis W, Bumpus N, et al. Short-term
administration of disulfiram for reversal of latent HIV infection: a phase 2 dose-escalation study.
Lancet HIV. 2015;2:e520–9. [PubMed: 26614966]
121. Rasmussen TA, Tolstrup M, Brinkmann CR, Olesen R, Erikstrup C, Solomon A, et al.
Panobinostat, a histone deacetylase inhibitor, for latent-virus reactivation in HIV-infected patients
on suppressive antiretroviral therapy: a phase 1/2, single group, clinical trial. Lancet HIV.
2014;1:e13–21. [PubMed: 26423811]

Curr HIV/AIDS Rep. Author manuscript; available in PMC 2023 August 22.
Kreider and Bar Page 17

122. Søgaard OS, Graversen ME, Leth S, Olesen R, Brinkmann CR, Nissen SK, et al. The
depsipeptide romidepsin reverses HIV-1 latency in vivo. Plos Pathog. 2015;11:e1005142.
Author Manuscript

123. Fidler S, Stöhr W, Pace M, Dorrell L, Lever A, Pett S, et al. Antiretroviral therapy alone
versus antiretroviral therapy with a kick and kill approach, on measures of the HIV reservoir in
participants with recent HIV infection (the RIVER trial): a phase 2, randomised trial. Lancet.
2020;395:888–98. [PubMed: 32085823]
124. Gruell H, Gunst JD, Cohen YZ, Pahus MH, Malin JJ, Platten M, et al. Effect of 3BNC117
and romidepsin on the HIV-1 reservoir in people taking suppressive antiretroviral therapy
(ROADMAP): a randomised, open-label, phase 2A trial. Lancet Microbe. 2022;3:E203–14.
[PubMed: 35544074]
125. Mavigner M, Liao LE, Brooks AD, Ke R, Mattingly C, Schoof N, et al. CD8
lymphocyte depletion enhances the latency reversal activity of the SMAC mimetic AZD5582
in ART-suppressed SIV-infected rhesus macaques. J Virol. 2021;95(8):e01429–20. 10.1128/
JVI.01429-20.
126. 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
Author Manuscript

SimianHuman Immunodeficiency Virus-Infected, Long-Term ARTTreated Rhesus Macaques. J


Virol. 2020;94(19):e00755–20. 10.1128/JVI.00755-20.
127. 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;578:154–9. [PubMed: 31969705]
128. Singh V, Dashti A, Mavigner M, Chahroudi A. Latency reversal 2.0: giving the immune system a
seat at the table. Curr Hiv-aids Rep. 2021;18:117–27. [PubMed: 33433817]
129. Bricker KM, Chahroudi A, Mavigner M. New latency reversing agents for HIV-1 cure: insights
from nonhuman primate models. Viruses. 2021;13:1560. [PubMed: 34452425]
130. Xu W, Jones M, Liu B, Zhu X, Johnson CB, Edwards AC, et al. Efficacy and mechanism-of-
action of a novel superagonist interleukin-15: interleukin-15 receptor αSu/Fc fusion complex
in syngeneic murine models of multiple myeloma. Cancer Res. 2013;73:3075–86. [PubMed:
23644531]
131. Harwood O, O’Connor S. Therapeutic potential of IL-15 and N-803 in HIV/SIV infection.
Viruses. 2021;13:1750. [PubMed: 34578331]
Author Manuscript

132. Jones RB, Mueller S, O’Connor R, Rimpel K, Sloan DD, Karel D, et al. A subset of
latency-reversing agents expose HIV-infected resting CD4+ T-cells to recognition by cytotoxic
T-lymphocytes. Plos Pathog. 2016;12:e1005545.
133. Webb GM, Molden J, Busman-Sahay K, Abdulhaqq S, Wu HL, Weber WC, et al. The human
IL-15 superagonist N-803 promotes migration of virus-specific CD8+ T and NK cells to B cell
follicles but does not reverse latency in ART-suppressed, SHIV-infected macaques. Plos Pathog.
2020;16:e1008339.
134. Conlon KC, Lugli E, Welles HC, Rosenberg SA, Fojo AT, Morris JC, et al. Redistribution,
hyperproliferation, activation of natural killer cells and CD8 T cells, and cytokine production
during first-in-human clinical trial of recombinant human interleukin-15 in patients with cancer. J
Clin Oncol. 2015;33:74–82. [PubMed: 25403209]
135. Romee R, Cooley S, Berrien-Elliott MM, Westervelt P, Verneris MR, Wagner JE, et al. First-in-
human phase 1 clinical study of the IL-15 superagonist complex ALT-803 to treat relapse after
transplantation. Blood. 2018;131:2515–27. [PubMed: 29463563]
Author Manuscript

136. Rosario M, Liu B, Kong L, Collins LI, Schneider SE, Chen X, et al. The IL-15-based
ALT-803 complex enhances FcγRIIIatriggered NK cell responses and in vivo clearance of B
cell lymphomas. Clin Cancer Res. 2016;22:596–608. [PubMed: 26423796]
137. Miller JS, Davis ZB, Helgeson E, Reilly C, Thorkelson A, Anderson J, Lima NS, Jorstad S, Hart
GT, Lee JH, Safrit JT, Wong H, Cooley S, Gharu L, Chung H, Soon-Shiong P, Dobrowolski
C, Fletcher CV, Karn J, Douek DC, Schacker TW. Safety and virologic impact of the IL-15
superagonist N-803 in people living with HIV: a phase 1 trial. Nat Med. 2022;28(2):392–400.
10.1038/s41591-021-01651-9. [PubMed: 35102335]

Curr HIV/AIDS Rep. Author manuscript; available in PMC 2023 August 22.
Kreider and Bar Page 18

138. Lim SY, Osuna CE, Hraber PT, Hesselgesser J, Gerold JM, Barnes TL, et al. TLR7 agonists
induce transient viremia and reduce the viral reservoir in SIV-infected rhesus macaques on
Author Manuscript

antiretroviral therapy. Sci Transl Med. 2018;10(439):eaao4521. 10.1126/scitranslmed.aao4521.


139. Prete GQD, Alvord WG, Li Y, Deleage C, Nag M, Oswald K, et al. TLR7 agonist administration
to SIV-infected macaques receiving early initiated cART does not induce plasma viremia. JCI
Insight. 2019;4:e127717.
140. Borducchi EN, Cabral C, Stephenson KE, Liu J, Abbink P, Ng’ang’a D, et al. Ad26/MVA
therapeutic vaccination with TLR7 stimulation in SIV-infected rhesus monkeys. Nature.
2016;540:284–7. [PubMed: 27841870]
141. Hsu DC, Schuetz A, Imerbsin R, Silsorn D, Pegu A, Inthawong D, et al. TLR7 agonist, N6-
LS and PGT121 delayed viral rebound in SHIV-infected macaques after antiretroviral therapy
interruption. Plos Pathog. 2021;17:e1009339.
142. 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. Sci Transl Med. 2021;13:eabg3071.
143. Sharpe AH. Introduction to checkpoint inhibitors and cancer immunotherapy. Immunol Rev.
Author Manuscript

2017;276:5–8. [PubMed: 28258698]


144. Haslam A, Prasad V. Estimation of the percentage of US patients with cancer who are eligible for
and respond to checkpoint inhibitor immunotherapy drugs. Jama Netw Open. 2019;2:e192535.
145. Harper J, Gordon S, Chan CN, Wang H, Lindemuth E, Galardi C, et al. CTLA-4 and PD-1
dual blockade induces SIV reactivation without control of rebound after antiretroviral therapy
interruption. Nat Med. 2020;26:519–28. [PubMed: 32284611]
146. Rasmussen TA, Rajdev L, Rhodes A, Dantanarayana A, Tennakoon S, Chea S, et al. Impact of
Anti-PD-1 and Anti-CTLA-4 on the Human Immunodeficiency Virus (HIV) reservoir in people
living with HIV with cancer on antiretroviral therapy: the AIDS malignancy consortium 095
Study. Clin Infect Dis. 2021;73(7):e1973–e1981. 10.1093/cid/ciaa1530. [PubMed: 33677480] ••
Clinical trial of people living with HIV and cancer receiving checkpoint inhibitors.
147. Melenhorst JJ, Chen GM, Wang M, Porter DL, Chen C, Collins MA, et al. Decade-long
leukaemia remissions with persistence of CD4+ CAR T cells. Nature. 2022;602(7897):503–9.
10.1038/s41586-021-04390-6. [PubMed: 35110735]
148. Herzig E, Kim KC, Packard TA, Vardi N, Schwarzer R, Gramatica A, et al. Attacking latent HIV
Author Manuscript

with convertible CAR-T cells, a highly adaptable killing platform. Cell. 2019;179:880894.e10.
• Technologic advancement to generate modular CARs capable of incorporating a variety of
antigen-binding domains.
149. Anthony-Gonda K, Bardhi A, Ray A, Flerin N, Li M, Chen W, et al. Multispecific anti-HIV
duoCAR-T cells display broad in vitro antiviral activity and potent in vivo elimination of HIV-
infected cells in a humanized mouse model. Sci Transl Med. 2019;11:eaav5685.
150. Leibman RS, Richardson MW, Ellebrecht CT, Maldini CR, Glover JA, Secreto AJ, et al.
Supraphysiologic control over HIV-1 replication mediated by CD8 T cells expressing a
reengineered CD4-based chimeric antigen receptor. Plos Pathog. 2017;13:e1006613.
151. 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;26:1776–87. [PubMed: 32868878]
152. Rust BJ, Kean LS, Colonna L, Brandenstein KE, Poole NH, Obenza W, et al. Robust expansion
of HIV CAR T cells following antigen boosting in ART-suppressed nonhuman primates. Blood.
Author Manuscript

2020;136:1722–34. [PubMed: 32614969]


153. Zhen A, Peterson CW, Carrillo MA, Reddy SS, Youn CS, Lam BB, et al. Long-term persistence
and function of hematopoietic stem cell-derived chimeric antigen receptor T cells in a nonhuman
primate model of HIV/AIDS. Plos Pathog. 2017;13:e1006753.
154. Barber-Axthelm IM, Barber-Axthelm V, Sze KY, Zhen A, Suryawanshi GW, Chen ISY,
et al. Stem cell–derived CAR T cells traffic to HIV reservoirs in macaques. JCI Insight.
2021;6:e141502.

Curr HIV/AIDS Rep. Author manuscript; available in PMC 2023 August 22.
Kreider and Bar Page 19

155. Haeseleer F, Fukazawa Y, Park H, Varco-Merth B, Rust BJ, Smedley JV, et al. Immune
inactivation of anti-simian immunodeficiency virus chimeric antigen receptor T cells in rhesus
Author Manuscript

macaques. Mol Ther Methods Clin Dev. 2021;22:304–19. [PubMed: 34485613]


156. Iwamoto N, Patel B, Song K, Mason R, Bolivar-Wagers S, Bergamaschi C, et al. Evaluation of
chimeric antigen receptor T cell therapy in non-human primates infected with SHIV or SIV. Plos
One. 2021;16:e0248973.
157. Peterson CW. HIV-specific CAR T cells return to the clinic. J Clin Invest. 2021;131(19):e153204.
10.1172/JCI153204.
158. Mu W, Carrillo MA, Kitchen SG. Engineering CAR T cells to target the HIV reservoir. Front Cell
Infect Microbiol. 2020;10:410. [PubMed: 32903563]
159. Liu B, Zhang W, Xia B, Jing S, Du Y, Zou F, et al. Broadly neutralizing antibody-derived
CAR T cells reduce viral reservoir in individuals infected with HIV-1. J Clin Invest.
2021;131(19):e150211. 10.1172/JCI150211.
160. Sarkar I, Hauber I, Hauber J, Buchholz F. HIV-1 proviral DNA excision using an evolved
recombinase. Science. 2007;316:1912–5. [PubMed: 17600219]
161. Karpinski J, Hauber I, Chemnitz J, Schäfer C, Paszkowski-Rogacz M, Chakraborty D, et al.
Author Manuscript

Directed evolution of a recombinase that excises the provirus of most HIV-1 primary isolates
with high specificity. Nat Biotechnol. 2016;34:401–9. [PubMed: 26900663]
162. Hauber I, Hofmann-Sieber H, Chemnitz J, Dubrau D, Chusainow J, Stucka R, et al. Highly
significant antiviral activity of HIV-1 LTR-specific Tre-recombinase in humanized mice. Plos
Pathog. 2013;9:e1003587.
163. Bessen JL, Afeyan LK, Dančík V, Koblan LW, Thompson DB, Leichner C, et al. High-resolution
specificity profiling and off-target prediction for site-specific DNA recombinases. Nat Commun.
2019;10:1937. [PubMed: 31028261]
164. Xiao Q, Guo D, Chen S. Application of CRISPR/Cas9-based gene editing in HIV-1/AIDS
therapy. Front Cell Infect Microbiol. 2019;9:69. [PubMed: 30968001]
165. Dash PK, Kaminski R, Bella R, Su H, Mathews S, Ahooyi TM, et al. Sequential LASER ART
and CRISPR treatments eliminate HIV-1 in a subset of infected humanized mice. Nat Commun.
2019;10:2753. [PubMed: 31266936]
166. Mancuso P, Chen C, Kaminski R, Gordon J, Liao S, Robinson JA, et al. CRISPR based editing of
Author Manuscript

SIV proviral DNA in ART treated non-human primates. Nat Commun. 2020;11:6065. [PubMed:
33247091] • Proof-of-concept that proviral editing with CRISPR/cas9 is possible in infected
primates.
167. Bella R, Kaminski R, Mancuso P, Young W-B, Chen C, Sariyer R, et al. Removal of HIV
DNA by CRISPR from patient blood engrafts in humanized mice. Mol Ther - Nucleic Acids.
2018;12:275–82. [PubMed: 30195766]
168. Ueda S, Ebina H, Kanemura Y, Misawa N, Koyanagi Y. AntiHIV-1 potency of the CRISPR/
Cas9 system insufficient to fully inhibit viral replication. Microbiol Immunol. 2016;60:483–96.
[PubMed: 27278725]
169. Wang G, Zhao N, Berkhout B, Das AT. CRISPR-Cas9 can inhibit HIV-1 replication but NHEJ
repair facilitates virus escape. Mol Ther. 2016;24:522–6. [PubMed: 26796669]
170. Mefferd AL, Bogerd HP, Irwan ID, Cullen BR. Insights into the mechanisms underlying
the inactivation of HIV-1 proviruses by CRISPR/Cas. Virology. 2018;520:116–26. [PubMed:
29857168]
171. Wang Z, Wang W, Cui YC, Pan Q, Zhu W, Gendron P, et al. HIV-1 employs multiple mechanisms
Author Manuscript

to resist Cas9/single guide RNA targeting the viral primer binding site. J Virol. 2018;92:e01135–
e1218. [PubMed: 30068653]
172. Wang Z, Pan Q, Gendron P, Zhu W, Guo F, Cen S, et al. CRISPR/Cas9-derived mutations
both inhibit HIV-1 replication and accelerate viral escape. Cell Rep. 2016;15:481–9. [PubMed:
27068471]
173. Smith LM, Ladner JT, Hodara VL, Parodi LM, Harris RA, Callery JE, et al. Multiplexed simian
immunodeficiency virusspecific paired RNA-guided Cas9 nickases inactivate proviral DNA. J
Virol. 2021;95(23):e0088221. 10.1128/JVI.00882-21.

Curr HIV/AIDS Rep. Author manuscript; available in PMC 2023 August 22.
Kreider and Bar Page 20

174. Wang G, Zhao N, Berkhout B, Das AT. A combinatorial CRISPR-Cas9 attack on HIV-1 DNA
extinguishes all infectious provirus in infected T cell cultures. Cell Rep. 2016;17:2819–26.
Author Manuscript

[PubMed: 27974196]
175. Chung C-H, Allen AG, Atkins A, Link RW, Nonnemacher MR, Dampier W, et al. Computational
design of gRNAs targeting genetic variants across HIV-1 subtypes for CRISPR-mediated
antiviral therapy. Front Cell Infect Microbiol. 2021;11:593077.
176. Dampier W, Sullivan NT, Chung C-H, Mell JC, Nonnemacher MR, Wigdahl B. Designing
broad-spectrum anti-HIV-1 gRNAs to target patient-derived variants. Sci Rep-uk. 2017;7:14413.
177. Sullivan NT, Dampier W, Chung C-H, Allen AG, Atkins A, Pirrone V, et al. Novel gRNA
design pipeline to develop broadspectrum CRISPR/Cas9 gRNAs for safe targeting of the HIV-1
quasispecies in patients. Sci Rep-uk. 2019;9:17088.
178. Herskovitz J, Hasan M, Patel M, Blomberg WR, Cohen JD, Machhi J, et al. CRISPR-Cas9
mediated exonic disruption for HIV-1 elimination. Ebiomedicine. 2021;73:103678.
179. Li H, Wang S, Lee FH, Roark RS, Murphy AI, Smith J, et al. New SHIVs and improved design
strategy for modeling HIV-1 transmission, immunopathogenesis, prevention and cure. J Virol.
2021;95(11):e00071–21. 10.1128/JVI.00071-21. [PubMed: 33658341]
Author Manuscript

180. Bauer AM, Ziani W, Lindemuth E, Kuri-Cervantes L, Li H, Lee FH, et al. Novel transmitted/
founder Simian-Human immunodeficiency viruses for human immunodeficiency virus latency
and cure research. J Virol. 2020;94(8):e01659–19. 10.1128/JVI.01659-19 [PubMed: 31969435]
181. Ziani W, Bauer A, Lu H, Wang X, Wu X, Bar KJ, et al. Immune responses and viral persistence
in Simian/Human immunodeficiency virus SHIV.C.CH848-Infected Rhesus Macaques. J Virol.
2021;95(9):e02198–20. 10.1128/JVI.02198-20. [PubMed: 33568508]
Author Manuscript
Author Manuscript

Curr HIV/AIDS Rep. Author manuscript; available in PMC 2023 August 22.
Kreider and Bar Page 21
Author Manuscript
Author Manuscript
Author Manuscript

Fig. 1.
Author Manuscript

Dynamics of the HIV-1 reservoir following ART initiation. A Schematization of three


measures of the proviral reservoir size plotted over time. “Total proviral DNA” corresponds
to quantitative measurements of any proviral genomes, which are generally stable overtime.
Methods that can differentiate defective from intact proviruses like IPDA and Q4PCR have
shown relative stability of rthe defective pool, but a gradual decline in the intact proviral
reservoir (red line). B Multiple forces that impact the size of the intact (filled color circles)
and defective (white circles) proviral reservoir have been identified and/or postulated. Forces

Curr HIV/AIDS Rep. Author manuscript; available in PMC 2023 August 22.
Kreider and Bar Page 22

that expand population size are listed on top and forces thought to cause decay of the intact
Author Manuscript

reservoir shown on bottom. C With time on suppressive ART, cells harboring intact proviral
populations decrease in total number and become more clonal, as indicated by the expansion
of certain clones (e.g., red clone) and loss of others (e.g., yellow clone). Figure created with
BioRender
Author Manuscript
Author Manuscript
Author Manuscript

Curr HIV/AIDS Rep. Author manuscript; available in PMC 2023 August 22.

You might also like