Sie sind auf Seite 1von 6

Available online at www.sciencedirect.

com

ScienceDirect

Which therapeutic strategy will achieve a cure for HIV-1?


Anthony R Cillo and John W Mellors

Strategies to achieve a cure for HIV-1 infection can be Main body


broadly classified into three categories: eradication cure Specific strategies to achieve a cure for HIV-1
(elimination of all viral reservoirs), functional cure (immune Eradication cure
control without reservoir eradication), or a hybrid cure The solitary example of an eradication cure to date is
(reservoir reduction with improved immune control). The Timothy Ray Brown, the Berlin patient [1], whose HIV-
many HIV-1 cure strategies being investigated include infected immune system was replaced, through allogene-
modification of host cells to resist HIV-1, engineered T cells ic hematopoietic stem cell transplantation, with one that
to eliminate HIV-infected cells, broadly HIV-1 neutralizing was resistant to HIV-1 as a result of a homozygous
monoclonal antibodies, and therapeutic vaccination, but the CCR5D32 gene mutation of the donor’s cells. Even in
‘kick and kill’ strategy to expose latent HIV-1 with latency the case of near complete replacement of the recipient’s
reversing agents (LRAs) and kill the exposed cells through immune system, viral eradication is not guaranteed; the
immune effector functions is currently the most actively Boston patients who underwent allogeneic hematopoietic
pursued. It is unknown, however, whether LRAs can deplete stem cell transplant with wild-type CCR5 donors experi-
viral reservoirs in vivo or whether current LRAs are enced viral rebound despite persistence of <0.001% of
sufficiently safe for clinical use. host cells in peripheral blood [2,3]. In addition, alloge-
neic transplantation has high morbidity and mortality, and
is thus not suitable for those without life-threatening
Address hematologic malignancies. As a consequence, most strat-
Division of Infectious Diseases, School of Medicine, University of egies to achieve a cure of HIV-1 infection have focused on
Pittsburgh, Pittsburgh, PA, USA achieving either a functional cure or a hybrid cure, as
illustrated below.
Corresponding author: Mellors, John W (jwm1@pitt.edu)

Functional cure
Current Opinion in Virology 2016, 18:14–19 One strategy to achieve a functional cure is the editing of
This review comes from a themed issue on Antiviral strategies the human genome in either autologous CD4+ T cells [4]
Edited by Raymond F Schinazi and Erik de Clercq
or autologous hematopoietic stem cells [5,6] by homing
endonucleases to knock out the gene for CCR5, thereby
For a complete overview see the Issue and the Editorial
blocking the ability of HIV-1 to enter via the main
Available online 15th March 2016 coreceptor. Tebas et al. have advanced this concept to
http://dx.doi.org/10.1016/j.coviro.2016.02.001 clinical trials, using cyclophosphamide conditioning to
1879-6257/Published by Elsevier B.V. achieve successful engraftment of zinc-finger modified
autologous CD4+ T cells [4]. Questions still remain
regarding the efficacy of the CCR5 knockout approach,
namely the proportion of cells that need to be modified to
prevent viral rebound and whether CXCR4-tropic virus
emerges over time. A recent alternative approach has
Introduction been to modify autologous cells to express CRISPR/
Despite the success of antiretroviral therapy (ART) in Cas9 to disrupt the provirus rather than a host gene
fully suppressing viral replication, HIV-1 remains an [7], although this approach is complicated by proviral
incurable viral infection. Recently, significant resources sequence diversity. Others have attempted to prevent
have been invested in developing and evaluating poten- the spread of infection in a macaque model by adding an
tially curative strategies for HIV-1 infection. A cure for expression cassette for a fusion inhibitor (mC46) in au-
HIV-1, defined as control of viral replication in the tologous cells, preventing virus entry [8], but it is un-
absence of ART, could theoretically be achieved through known whether enough target cells can be modified and
several varied approaches and goals: an eradication cure whether HIV-1 can evolve resistance to this inhibitor over
(elimination of all viral reservoirs); a functional cure time.
(control of viral replication without reservoir eradication),
or a hybrid cure (reduction of the reservoir and boosting of Another strategy to achieve a functional cure is the
immune responses) (Figure 1). Here, we discuss specific introduction of designer immune responses, one of which
therapeutic approaches aimed at achieving a cure of is chimeric antigen receptor (CAR) expressing effector
HIV-1 (summarized in Table 1), with a focus on the cytotoxic T lymphocytes targeting HIV-1 epitopes on
‘kick and kill’ strategy. MHC-I or cell-surface HIV-1 envelope. CAR T cells

Current Opinion in Virology 2016, 18:14–19 www.sciencedirect.com


Curative strategies for HIV-1 Cillo and Mellors 15

Figure 1

Eradication Cure Eradication


Timothy Ray Cure
Brown (Berlin No functional HIV-1
Patient) remaining in the body

Hybrid Cure Functional Cure


Reduce Hybrid Functional Elite
reservoir size Cure Cure controllers
and diversity Reduced functional Control of HIV VISCONTI
with “kick” reservoirs & improve without ART or Post-ART
Enhance immune control deleterious controllers
immune without ART immunologic effects
Host cell
responses modification
with “kill”

Current Opinion in Virology

Broad definitions of a cure for HIV-1, with specific examples. An eradication cure would be one in which no functional HIV-1 remains in the body,
and is represented by allogeneic stem cell transplantation and complete replacement of the recipient’s HIV-infected immune system. Functional
cure, illustrated by elite controllers and post-treatment controllers (and also possibly successful host cell modification), is defined as control of
HIV-1 replication without ART, immune dysfunction (i.e. depletion of CD4+ T cells or persistent immune activation) or reservoir eradication. A
hybrid cure could be achieved by reduction in the size and diversity of the latent reservoir (as in an eradication cure), combined with enhanced
immune control of HIV-1.

have proven efficacious for B cell lymphoma by targeting Considerable effort has gone into the discovery and
CD19 [9], but at the cost of healthy B cells. Off-target characterization of bNAbs for HIV-1 envelope, but elicit-
specificity of engineered T cells may be a significant ing bNAb-like antibodies through therapeutic vaccina-
safety barrier in this approach, as fatal cardiac toxicity tion remains difficult because of the extensive affinity
occurred in myeloma and melanoma trials using affinity maturation required to achieve the often structurally
purified T cells [10,11]. Whether sufficient numbers abnormal bNAbs (reviewed by [12]). However, passive
of CAR modified T cells could be targeted to invariant infusion of a bNAb or a cocktail of bNAbs could prove to
HIV-1 epitopes and maintained in vivo are important be a successful approach to an ART-free functional cure.
challenges for this approach. The AIDS Clinical Trials Group (ACTG) study A5342 is

Table 1

Specific HIV-1 curative strategies under investigation

Overarching strategy Specific strategy Application of specific strategies


Eradication cure Allogeneic stem cell transplantation  Timothy Ray Brown (Berlin patient) [1]
 Boston patients [2,3]
Functional cure Genome editing  Knockout of CCR5 in CD4+ T cells or hematopoietic stem cells [4–6]
 Knockout of HIV-1 proviruses [7]
 Expression of a viral fusion inhibitor [8]
Designer immune responses  Chimeric antigen receptors on CD8+ T cells [9]
 Broadly neutralizing monoclonal antibodies [12]
Control of viral replication  Highly effective CD8+ T cell response to control viral replication,
similar to elite controllers [13–17]
 Post-treatment control of viral replication, similar to the VISCONTI
cohort controllers [18]
Hybrid cure Kick and kill  LRAs disrupt proviral latency, followed by death of infected cells
 Reduced reservoir size and diversity permits suppression of viral
replication with boosted immune responses

www.sciencedirect.com Current Opinion in Virology 2016, 18:14–19


16 Antiviral strategies

currently being undertaken to test whether a bNAb can Box 1 Key points
reduce the size of the reservoir in vivo. Complicating
factors with non-autologous bNAbs are the generation of  HIV-1 curative strategies can be broadly classified into eradication
anti-bNAb antibodies, and the pre-existence of HIV-1 cure, functional cure, or hybrid cure.
envelope with escaped epitopes to the bNAbs, which  An eradication cure is least likely to be achieved because of the
persistence of long-lived, CD4+ T cells with intact proviruses
could limit long-term efficacy. throughout the human body.
 Control of HIV-1 in the absence of reservoir reduction (functional
Modulation of T cell mediated immunity could also cure) will be more difficult to achieve compared with strategies that
control viral replication in the absence of ART. Rare both reduce the reservoir and enhance HIV-1 specific immune
effector functions (hybrid cure).
elite controllers are capable of controlling viral replication
 Many specific strategies are under investigation for their ability to
in the absence of ART, often due to favorable HLA achieve a cure of HIV-1, with kick and kill being the most
alleles and highly effective CD8+ T cell responses intensively pursued approach to date.
[13–15]. The vast majority of people infected with  Clinical trials of latency reversing agents have generally shown
HIV-1 do not control viral replication, and would require increases in cellular HIV-1 RNA from baseline, but their effect on
HIV-1 protein expression, virion production and the latent inducible
therapeutic vaccination to elicit the proper cytotoxic reservoir are undefined.
T cell (CTL) responses for control of viral replication.  Reduction in the size and diversity of the viral reservoir, in
This may be possible in the context of a highly effective, conjunction with modulation of immune responses is an under-
patrolling CTL response capable of preventing systemic developed but vital area of functional HIV-1 cure research.
viral spread. Hansen et al. demonstrated that a rhesus
CMV-vectored vaccine could control viremia following
challenge and eliminate virally-infected cells [16,17].
Though promising, the utility of this approach in the ing HDACi treatment [20]. This gene dysregulation may
context of an established latent infection in humans is contribute to the impaired CTL function observed ex vivo
currently uncertain. Another example of immune control following treatment with HDACi [22]. Trials of the more
is exhibited by a limited number of individuals in the potent HDACi panobinostat and romidepsin have also
VISCONTI cohort, who have shown durable control of been performed, and have shown upregulation of cellular
viremia following early initiation of ART in the absence HIV-1 RNA [23,24]. In contrast to studies of vorinostat,
of favorable elite controller alleles [18]. Immunologic modest increases in plasma viremia were detected by
correlates of control in this cohort have yet to be defined. either transcription-mediated amplification (TMA) or the
Roche COBAS AmpliPrep/TaqMan platform following
Hybrid cure dosing with panobinostat or romidepsin. Though not an
A strategy that aims to both reduce the viral reservoir and HDACi, disulfiram has also been shown to modestly
enhance immune control of the residual reservoir is increase plasma viremia and cellular unspliced HIV-1
referred to as the ‘kick and kill’ or ‘shock and kill’ RNA levels [25,26].
strategy. In this approach, reservoir reduction is achieved
by pharmacologically reactivating latent proviruses (kick) Trials of LRAs to date should be interpreted with caution,
with latency reversing agents (LRAs), followed by sub- in part because assays used to measure virion HIV-1 RNA
sequent death of the infected cell through either immune in plasma (TMA and AmpliPrep/TaqMan platforms)
clearance or cytopathic effects of HIV-1 protein expres- cannot readily distinguish virion-associated viral RNA
sion. The kick component has been tested with numerous from HIV-1 DNA in plasma resulting from cellular cyto-
candidate LRAs in proof of principle clinical trials. As toxicity from the HDACi. Another important caveat is
such, the remainder of this review will focus on the results that cellular HIV-1 RNA expression following HDACi
of clinical trials of LRAs to date, the preclinical assess- treatment could be a result of readthrough transcripts,
ment of new LRAs, and perspectives on the future of this which can be mistaken for increases in functional
approach. unspliced HIV-1 RNA [27]. Finally, it will be important
in future studies to assess whether increases in cellular
Clinical trials of HIV-1 latency reversing agents HIV-1 transcription are related to increases in translation
The best-studied putative LRAs to date have been of HIV-1 proteins, rendering reactivated cells visible to
histone deacetylase inhibitors (HDACi), with vorinostat, the immune system.
panobinostat and romidepsin having been investigated
clinically (Table 2). Trials of vorinostat have shown Ex vivo assessments of new latency reversing agents
upregulation of cellular HIV-1 RNA expression Results from clinical trials have generated optimism that
[19,20], though not consistently after repeated dosing the kick and kill strategy can be used to reverse latency.
[21], and without concomitant increases in plasma viremia However, HDACi only antagonize one known mecha-
[19,20]. HDACi may also have longer term effects on nism of proviral latency, namely nucleosome structure.
gene expression in vivo than previously thought, as gene Mechanisms of proviral latency are complex and multi-
profiling revealed protracted gene dysregulation follow- factorial, and have been reviewed elsewhere (reviewed in

Current Opinion in Virology 2016, 18:14–19 www.sciencedirect.com


Curative strategies for HIV-1 Cillo and Mellors 17

Table 2

Recent clinical trials of latency reversing agents

Clinical trial, NCT identifier First author, location(s) of trial Primary endpoint Results of the trial
Single-dose vorinostat, Archin et al. [19], University of North Changes in unspliced cellular HIV-1 Mean 4.8-fold increase in cellular
NCT01319383 Carolina Chapel Hill RNA in resting CD4+ T cells HIV-1 RNA over pre-vorinostat
Pilot study of disulfiram, Spivak, Andrade et al. [25], Changes in plasma HIV-1 RNA No significant change in viremia
NCT01286259 University of Utah/Johns Hopkins levels during disulfiram, but 1.88-fold
University increase observed post-treatment
Short-course vorinostat, Archin et al. [21], University of North Changes in unspliced cellular HIV-1 No significant increase in cellular
NCT01319383 Carolina Chapel Hill RNA in resting CD4+ T cells HIV-1 RNA over time
Short-course vorinostat, Elliott et al. [20], Alfred Hospital and Changes in unspliced cellular HIV-1 Median peak increase of 7.4-fold
NCT01365065 Monash University/Burnet Institute/ RNA in total CD4+ T cells cellular HIV-1 RNA from pre-
University of Melbourne vorinostat; protracted gene
dysregulation observed
Short-course panobinostat, Rasmussen et al. [23], Aarhus Changes in unspliced cellular HIV-1 Median maximum increase
NCT01680094 University RNA in total CD4+ T cells of 3.5-fold from pre-panobinostat;
no change in time to rebound off
ART
Multi-dose romidepsin, Søgaard et al. [24], Aarhus Changes in unspliced cellular HIV-1 2.4–5.0-fold increase in cellular
NCT02092116 University RNA in total CD4+ T cells HIV-1 RNA from pre-romidepsin
Short-course disulfiram, Elliott et al. [26], Monash University Changes in unspliced cellular HIV-1 Increases in unspliced HIV-1 RNA
NCT01944371 and Alfred Hospital/University of RNA in total CD4+ T cells observed in all groups during and
California San Francisco post disulfiram treatment, ranging
from 1.6- to 2.5-fold

[28]). Activation of NF-kB through agonism of protein appears to increase cellular HIV-1 mRNA transcription
kinase C (PKC) with bryostatin-1 or ingenol derivatives, at or near the level of synergy [40]. Several studies have
which permits binding of this transcription factor to the also shown that PKC agonists in conjunction with BRD4
HIV-1 long terminal repeat [29], has recently been tested inhibitors increase cellular HIV-1 mRNA transcription in
in ex vivo systems by several groups. The PKC agonist at least a supra-additive manner [40,41,42]. These
bryostatin-1 has been investigated in vivo for cancer studies suggest that HIV-1 transcription can be modulat-
[30,31], and is currently in a Phase II trial for Alzheimer’s ed by LRAs targeting HDAC, PKC or BRD4. However,
disease (NCT02431468). A trial in Spain in which bryos- the relative potency of LRAs compared with full T cell
tatin was given to HIV-1 patients is currently closed, and activation at the level of individual proviruses [44] should
results are pending (BRYOLAT, NCT02269605). be investigated for promising LRAs. An important unan-
swered question is whether these compounds alone or in
Another aspect of proviral latency that is amenable to combination will be safe enough for use in HIV-positive
therapeutic intervention is the availability of P-TEFb individuals who are healthy on suppressive ART.
(consisting of Cyclin T1 and CDK9, reviewed by [32]) to Compounds with narrow therapeutic indices in preclini-
interact with the viral protein trans-activator of transcrip- cal animal studies should not be pursued in human
tion (Tat) [33]. Following release of P-TEFb from the studies.
suppressive 7SK snRNA complex [34], Cyclin T1 and
CDK9 can interact with bromodomain and extra terminal One current knowledge gap that exists in the study of
domain 4 (BRD4) as part of the super-elongation com- LRAs is whether they have caused a decrease in the size
plex; however, this limits availability for P-TEFb for of the inducible reservoir ex vivo. To achieve a hybrid
interaction with Tat [35]. Bromodomain inhibitors cure, it will likely be necessary to reduce both the size and
function by preventing the interaction of P-TEFb with diversity of the reservoir with LRAs; assays will be
BRD4, allowing Tat to bind P-TEFb and facilitating required to determine if LRAs in preclinical development
HIV-1 transcriptional elongation [36]. BRD4 inhibitors can reduce the size of the reservoir, and the degree of
have been developed for the treatment of cancer [37,38], additional immunologic control bNAbs or CTL responses
and are in early phase trials for hematologic malignancies that will be required to prevent viral escape after cessa-
[39]. tion of ART. An approach to a functional cure consisting
of a reduction in the size and diversity of the reservoir,
Recent ex vivo studies have focused on combinations of followed by boosting of immune responses to conserved
PKC inhibitors, or BRD4 inhibitors, with HDACi. These viral epitopes could be thought of as a ‘kick, kill and
studies have revealed that PKC agonists alone are gener- contain’ strategy. Careful studies of the change in reser-
ally the most potent LRAs [27,40,41,42], likely due to voir size and investigation of control by immunologic
partial T cell activation through the PKC pathway [43]. mechanisms will inform whether a kick, kill and contain
HDACi in combination with bryostatin-1 or ingenol strategy can be used to achieve a cure of HIV-1.

www.sciencedirect.com Current Opinion in Virology 2016, 18:14–19


18 Antiviral strategies

Conclusion 8. Younan PM, Polacino P, Kowalski JP, Peterson CW, Maurice NJ,
Williams NP, Ho O, Trobridge GD, Von Laer D, Prlic M et al.:
Many strategies to achieve an HIV-1 cure are being Positive selection of mC46-expressing CD4+ T cells and
studied (Box 1). Early proof of principle clinical trials maintenance of virus specific immunity in a primate AIDS
model. Blood 2013, 122:179-187.
of LRAs have shown increased levels of HIV-1 RNA
9. Porter Dl, Levin BL, Kalos M, Bagg A, June CH: Chimeric antigen
transcription, and ex vivo assessments of new LRAs tar- receptor-modified T cells in chronic lymphoid leukemia. N Engl
geting different aspect of proviral latency are underway. J Med 2011, 365:725-733.
Open questions are whether reservoir reduction is 10. Cameron BJ, Gerry AB, Dukes J, Harper JV, Kannan V, Bianchi FC,
achieved with LRAs and whether LRAs or LRA combi- Grand F, Brewer JE, Gupta M, Plesa G et al.: Identification of a
titin-derived HLA-A1-presented peptide as a cross-reactive
nations have favorable toxicity profiles. Improving the kill target for engineered MAGE A3-directed T cells. Sci Transl Med
aspect of the kick and kill strategy by enhancing immune 2013 http://dx.doi.org/10.1126/scitranslmed.3006034.
effectors is a critical component of the ‘kick, kill and 11. Linette GP, Stadtmauer EA, Maus MV, Rapoport AP, Levin BL,
contain’ strategy. Many promising strategies for an HIV-1 Emer L, Litzky L, Bagg A, Carreno BM, Cimino PJ et al.:
Cardiovascular toxicity and titin cross-reactivity of affinity-
cure are in their infancy, but it is unknown whether any enhanced T cells in myeloma and melanoma. Blood 2013,
strategy can achieve an ART-free remission of HIV-1. 122:863-871.
Nevertheless, such a goal should be pursued rigorously 12. Mascola JR, Haynes BF: HIV-1 neutralizing antibodies:
with vigilance for potential toxicities of the approaches understanding nature’s pathways. Immunol Rev 2013, 254:
225-244.
taken.
13. Betts MR, Nascon MC, West SM, De Roas SC, Migueles SA,
Abraham J, Lederman MM, Benito JM, Goepfert PA, Conners M
Conflict of interest statement et al.: HIV nonprogressors preferentially maintain highly
functional HIV-specific CD8+ T cells. Blood 2006, 107:
ARC has no conflicts to disclose. JWM is a consultant to 4781-4789.
Gilead Sciences, has received grant funding from Gilead
14. Migueles SA, Sabbaghian MS, Shupert WL, Bettinotti MP,
Sciences, and owns shares of Co-Crystal, Inc. Marincola FM, Martino L, Hallahan CW, Selig SM, Schwartz D,
Sullivan J, Connors M: HLA B*5701 is highly associated with
restriction of virus replication in a subgroup of HIV-infected
References and recommended reading long term nonprogressors. Proc Natl Acad Sci U S A 2000,
Papers of particular interest, published within the period of review, 97:2709-2714.
have been highlighted as:
15. Horton H, Frank I, Baydo R, Jalbert E, Penn J, Wilson S,
 of special interest McNevin JP, McSweyn MD, Lee D, Huang Y, De Roas SC et al.:
 of outstanding interest Preservation of T cell proliferation restricted by protective
HLA alleles is critical for immune control of HIV-1 infection.
J Immunol 2006, 177:7406-7414.
1. Hütter G, Nowak D, Mossner M, Ganepola S, Müssig A, Allers K,
Schneider T, Hofmann J, Kütcherer C, Blau O et al.: Long-term 16. Hansen SG, Ford JC, Lewis MS, Ventura AB, Hughes CM,
control of HIV by CCR5 Delta32/Delta32 stem-cell Coyne-Johnson L, Whizin N, Oswald K, Shoemaker R, Swanson T
transplantation. N Engl J Med 2009, 360:692-698. et al.: Profound early control of highly pathogenic SIV by an
effector memory T-cell vaccine. Nature 2011, 473:527.
2. Henrich TJ, Hu Z, Li JZ, Sciaranghella G, Busch MP, Keating SM,
Gallien S, Lin NH, Giguel FF, Lavoie L et al.: Long-term reduction 17. Hansen SG, Piatak M Jr, Ventura AB, Hughes CM, Gilbride RM,
in peripheral blood HIV type 1 reservoirs following reduced- Ford JC, Oswalk K, Shoemaker R, Li Y, Lewis MS et al.: Immune
intensity conditioning allogeneic stem cell transplantation. clearance of highly pathogenic SIV infection. Nature 2013,
J Infect Dis 2013, 207:1694-1702. 502:100-104.
3. Henrich TJ, Hanhauser E, Marty FM, Sirignano MN, Keating S, 18. Sáez-Cirión A, Bacchus C, Hocqueloux L, Avettand-Fenoel V,
 Lee TH, Robles YP, Davis BT, Li JZ, Heisey A et al.: Antiretroviral- Girault I, Lecuroux C, Potard V, Versmisse P, Melard A, Prazuck T
free HIV-1 remission and viral rebound after allogeneic stem et al.: Post-treatment HIV-1 controllers with a long-term
cell transplantation: report of 2 cases. Ann Intern Med 2014, virological remission after the interruption of early initiated
161:319-327. antiretroviral therapy ANRS VISCONTI Study. PLoS Pathog
This study demonstrated that profound reductions in the reservoir 2013 http://dx.doi.org/10.1371/journal.ppat.1003211.
through near-complete replacement of the host immune system by
allogeneic stem cell transplantation did not result in permanent ART-free 19. Archin NM, Liberty Al, Kashuba AD, Choudhary SK, Kuruc JD,
remission of HIV-1 infection. Crooks AM, Anderson EM, Kearney MF et al.: Administration of
vorinostat disrupts HIV-1 latency in patients on antiretroviral
4. Tebas P, Stein D, Tang WW, Frank I, Wang SQ, Lee G, Spratt SK, therapy. Nature 2012, 487:482-485.
Surosky RT, Giedlin MA et al.: Gene editing of CCR5 in
autologous CD4 T cells of persons infected with HIV. N Engl J 20. Elliot JM, Wightman F, Soloman A, Ghneim K, Ahlers J,
Med 2014, 370:901-910.  Cameron MJ, Smith MZ, Spelman T, McMahon J, Velayudham P
et al.: Activation of HIV transcription with short-course
5. Holt N, Wang J, Kenneth K, Friedman G, Wang X, Taupin V, vorinostat in HIV-infected patients on suppressive
Crooks GM, Kohn DB, Gregory PD, Holmes MC, Cannon PM: Zinc antiretroviral therapy. PLoS Pathog 2014 http://dx.doi.org/
finger nuclease-mediated CCR5 knockout hematopoietic 10.1371/journal.ppat.1004473.
stem cell transplantation controls HIV-1 in vivo. Nat Biotechnol In this in vivo study, short-course vorinostat treatment resulted in pro-
2010, 28:839-847. tracted increases in cellular HIV-1 RNA transcription. Additionally, the
authors also found persistent gene dysregulation after administration of
6. Li L, Krymskaya L, Wang J, Henley J, Rao A, Cao FL, Tran CA, vorinostat.
Torres-Coronado M, Gardner A, Gonzales N et al.: Genomic
editing of the HIV-1 coreceptor CCR5 in adult hematopoietic 21. Archin NM, Bateson R, Tripathy MK, Crooks AM, Yang KH,
stem and progenitor cells using zinc finger nucleases. Mol Dahi NP, Kearney MF, Anderson EM, Coffin JM, Strain MC et al.:
Ther 2013, 21:1259-1269. HIV-1 expression within resting CD4+ T cells after multiple
doses of vorinostat. J Infect Dis 2014, 210:728-735.
7. Liao HK, Gu Y, Diza A, Marlett J, Takahashi Y, Li M, Suzuki K, Xu R,
Hishida T, Chang CJ et al.: Use of the CRISPR/Cas9 system as 22. Jones RB, O’Connor R, Mueller S, Foley M, Szeto GL, Karel D,
an intracellular defense against HIV-1 infected in human cells. Lichterfeld M, Kovacs C, Ostrowski MA, Trocha A et al.: Histone
Nat Commun 2015 http://dx.doi.org/10.1038/ncomms7413. deacetylase inhibitors impair the elimination of HIV-infected

Current Opinion in Virology 2016, 18:14–19 www.sciencedirect.com


Curative strategies for HIV-1 Cillo and Mellors 19

cells by cytotoxic T-lymphocytes. PLoS Pathog 2014 http:// for HIV Tat transcriptional activation in vivo and in vitro. Genes
dx.doi.org/10.1737/journal.ppat.1994287. Dev 1997, 11:2633-2644.
23. Rasmussen TA, Tolstrup M, Brinkmann CR, Olesen R, Erikstrup C, 34. Yang Z, Zhu Q, Luo K, Zhou Q: The 7SK small nuclear RNA
Solomon A, Winckelmann A, Palmer S, Dinarello C, Buzon M et al.: inhibits the CDK9/cyclin T1 kinase to control transcription.
Panobinostat, a histone deacetylase inhibitors, for latent-virus Nature 2001, 414:317-322.
reactivation in HIV-infected patients on suppressive
antiretroviral therapy: a phase 1/2, single group, clinical trial. 35. Bisgrove DA, Mahmoudi, Henklein P, Verdin E: Conserved
Lancet HIV 2014 http://dx.doi.org/10.1016/S2352-3018(14)70014- P-TEFb-interacting domain of BRD4 inhibits HIV transcription.
1. Proc Natl Acad Sci U S A 2007, 104:13690-13695.
24. Søgaard OS, Graversen ME, Leth S, Olesen R, Brinkmann CR, 36. Zhu J, Gaiha GD, John SP, Pertel T, Chin Cr, Gao G, Qu H,
Nissen SK, Kjaer AS, Schleimann MH, Denton PW, Walker BD, Elledge SJ, Brass AL: Reactivation of latent HIV-1 by
Hey-Cunningham WJ et al.: The depsipeptide romidepsin inhibition of BRD4. Cell Rep 2012, 2:807-816.
reverses HIV-1 latency in vivo. PLoS Pathog 2015 http://
dx.doi.org/10.1371/journal.ppat.1005142. 37. Filippakopoulos P, Qi J, Picaud S, Shen Y, Smith WB, Fedorov O,
Morse EM, Keates T, Hickman TT, Felletar I et al.: Selective
25. Spivak Am, Andrade A, Eisele E, Hoh R, Bacchetti P, Bumpus NN, inhibition of BET bromodomains. Nature 2010, 468:
Emad F, Buckheit R 3rd, McCance-Katz EF, Lai J et al.: A pilot 1067-1073.
study assessing the safety and latency-reversing activity of
disulfiram in HIV-1-infected adults on antiretroviral therapy. 38. Puissant A, Frumm SM, Alexe G, Bassil CF, Qi J, Chanthery YH,
Clin Infect Dis 2014, 58:883-890. Nekritz EA, Zeid R, Gustafson WC, Greninger F et al.:
Targeting MYCN in neuroblastoma by BET bromodomain
26. Elliott JH, McMahon JH, Chang CC, Lee SA, Hartogensis W, inhibition. Cancer Discov 2013 http://dx.doi.org/10.1158/2159-
 Bumpus N, Savic R, Roney J, Hoh R, Solomon A et al.: Short-term 8290.CD-12-0418.
administration of disulfiram for reversal of latent HIV infection:
a phase 2 dose-escalation study. Lancet HIV 2015 http:// 39. Odore E, Lokiec F, Cvitkovic E, Bekradda M, Herait P, Bourdel F,
dx.doi.org/10.1016/S2352-3018(15)00226-X. Kahatt C, Raffoux E, Stathis A, Thieblemont C et al.: Phase I
This is the first in vivo study to demonstrate increases in cellular unspliced population pharmacokinetic assessment of the oral
HIV-1 RNA transcripts using a latency reversing agent that does not bromodomain inhibitor OTX015 in patients with haematologic
inhibit histone deacetylation. malignancies. Clin Pharmacokinet 2015. [Epub ahead of print].
27. Bullen CK, Laird GM, Durand CM, Siliciano JD, Siliciano RF: New 40. Laird GM, Bullen CK, Rosenbloom DI, Martin AR, Hill AL,
 ex vivo approaches distinguish effective and ineffective single  Durand CM, Siliciano JD, Siliciano RF: Ex vivo analysis identifies
agents for reversing HIV-1 latency in vivo. Nat Med 2014, effective HIV-1 latency-reversing drug combinations. J Clin
20:425-429. Invest 2015, 125:1901-1912.
Using resting CD4+ T cells isolated from patients on suppressive anti- This study utilized resting CD4+ T cells isolated from patients on sup-
retroviral therapy, this study demonstrated latency reversal with PKC pressive antiretroviral therapy to show that combinations of latency
agonists and also showed that histone deacetylase inhibitors may lead to reversing agents synergistically activate transcription of HIV-1 RNA.
readthrough transcription of HIV-1 RNA.
41. Darcis G, Kula A, Bouchat S, Fujinaga, Corazza F, Ait-Ammar A,
28. Van Lint C, Bouchat S, Marcell A: HIV-1 transcription and Delacort N, Melard A, Kabeya K, Vanhulle C et al.: An in-depth
latency: an update. Retrovirology 2013 http://dx.doi.org/10.1186/ comparison of latency-reversing agent combinations in
1742-4690-10-67. various in vitro and ex vivo HIV-1 latency models identified
bryostatin-1+JQ1 and ingenol-B+JQ1 to potently reactivate
29. Williams SA, Chen LF, Kwon H, Fenard D, Bisgrove D, Verdin E,
viral gene expression. PLoS Pathog 2015 http://dx.doi.org/
Greene WC: Prostratin antagonizes HIV latency by activation
10.1371/journal.ppat.1005063.
NF-kappaB. J Biol Chem 2004, 279:42008-42017.
30. Prendiville J, Crowther D, Thatcher N, Woll PJ, Fox BW, 42. Jiang G, Mendes EA, Kaiser P, Wong DP, Tang Y, Cai I, Fenton A,
McGown A, Testa N, Stern P, McDermott R, Potter M et al.: A Melcher GP, Hildreth JE, Thompson GR et al.: Synergistic
phase I study of intravenous bryostatin 1 in patients with reactivation of latent HIV expression by ingenol-3-angelate,
advanced cancer. Br J Cancer 1993, 68:418-424. PEP005, target NF-kB signaling in combination with JQ1
induced p-TEFb activation. PLoS Pathog 2015 http://dx.doi.org/
31. Zonder JA, Shields AF, Zalupski M, Chaplen R, Heilbrun LK, 10.1371/journal.ppat.1005066.
Arlauskas P, Philip PA: A phase II trial of bryostatin 1 in the
treatment of metastatic colorectal cancer. Clin Cancer Res 43. Valge VE, Wong JG, Datlof BM, Sinskey AJ, Rao A: Protein kinase
2001, 7:38-42. C is required for responses to T cell receptor ligands by not to
interleukin-2 in T cells. Cell 1988, 55:101-112.
32. Price DH: P-TEFb, a cyclin-dependent kinase controlling
elongation by RNA polymerase II. Mol Cell Biol 2000, 20: 44. Cillo AR, Sobolewski MD, Bosch RJ, Fyne E, Piatak M Jr,
2629-2634. Coffin JM, Mellors JW: Quantification of HIV-1 latency reversal
in resting CD4+ T cells from patients on suppressive
33. Mancebo HS, Lee G, Flygare J, Tomassini J, Luu P, Zhu Y, Peng J, antiretroviral therapy. Proc Natl Acad Sci U S A 2014, 111:
Blau C, Hazuda D, Price D, Flores O: P-TEFb kinase is required 7078-7083.

www.sciencedirect.com Current Opinion in Virology 2016, 18:14–19

Das könnte Ihnen auch gefallen