<< Back To Home

NEW THIS FRIDAY: THE SEARCH FOR AN HIV CURE

Thursday, 27th of June 2013 Print

 

·       THE SEARCH FOR AN HIV CURE: TACKLING LATENT INFECTION

 

The Lancet Infectious Diseases, Early Online Publication, 5 March 2013

Prof Stephen J Kent MD a d Corresponding AuthorEmail AddressJeanette C Reece MPH aJanka Petravic PhD bAlexey Martyushev BSc bMarit Kramski PhD aRobert De Rose PhD a, Prof David A Cooper DSc c, Prof Anthony D Kelleher PhD c, Prof Sean Emery PhD cPaul U Cameron PhD d e f, Prof Sharon R Lewin PhD d e f, Prof Miles P Davenport PhD b

Summary below; full text, with tables, is at http://www.thelancet.com/journals/laninf/article/PIIS1473-3099(13)70043-4/fulltext

Strategies to eliminate infectious HIV that persists despite present treatments and with the potential to cure HIV infection are of great interest. One patient seems to have been cured of HIV infection after receiving a bone marrow transplant with cells resistant to the virus, although this strategy is not viable for large numbers of infected people. Several clinical trials are underway in which drugs are being used to activate cells that harbour latent HIV. In a recent study, investigators showed that activation of latent HIV infection in patients on antiretroviral therapy could be achieved with a single dose of vorinostat, a licensed anticancer drug that inhibits histone deacetylase. Although far from a cure, such studies provide some guidance towards the logical next steps for research. Clinical studies that use a longer duration of drug dosing, alternative agents, combination approaches, gene therapy, and immune-modulation approaches are all underway.

Introduction

Combination antiretroviral therapy has had a pronounced effect on the course of HIV infection, turning it from an infection that is typically fatal within a period of years into a manageable chronic disease. However, from a practical, public health, and economic perspective, elimination of HIV-1 infection would be preferable to long-term suppression of viral replication with drugs. Cure will probably require reduction or elimination of the HIV-1 forms that persist in the presence of antiretroviral therapy. Several factors contribute to persistent HIV-1 infection, but a major reason is persistence of a replication-competent pool of virus in resting CD4 T cells.12

The pool of latently infected cells is established very early after the initial infection with HIV-1, usually before diagnosis and antiretroviral therapy in routine practice.3 Very early initiation of antiretroviral therapy has been associated with a reduction in the number of latently infected cells, but virus rebounds when treatment is stopped.4—7 HIV in latently infected cells can remain dormant for the life of the cell and is not substantially affected by intensification of antiretroviral therapy.8—10 Attempts to eliminate these cells are a key theme of eradication strategies,2 with calls to invest heavily in innovative approaches.11 A cure for HIV is usually defined as either a sterilising cure (by which in theory all latent HIV DNA is eliminated) or a functional cure (wherein latent HIV persists but viraemia is very low or absent without the use of antiretroviral therapy). We focus on the potential for a sterilising cure.

Antiretroviral therapy-based strategies

HIV-1 integrates into the chromosomal DNA of infected cells as part of its lifecycle. A small proportion of infected cells remain transcriptionally silent but with fully infectious virus.12 These cells are thought to restart active HIV-1 replication when antiretroviral therapy is withdrawn, even after many years of treatment. How these cells persist for many years is not fully understood. The presence of HIV-1 DNA in very long-lived cells (particularly memory CD4 T cells) seems to be the main reason that this latent pool of proviral HIV-1 does not decay or evolve.13—15 Some of these cells might homoeostatically proliferate to maintain the size of the viral reservoir.16 Alternatively, even very low-level replication could result in infection of new cells and replenishment of the reservoir despite antiretroviral therapy.8 However, the absence of viral evolution or the development of drug resistance argues against continued replication in patients on antiretroviral therapy.

Treatment intensification studies have not shown reduction in the size of the HIV-1 reservoir or prevented the recrudescence of viraemia after withdrawal of treatment.10 One study8 in 2010, however, showed that intensification of successful antiretroviral therapy with the integrase inhibitor raltegravir resulted in a subset of patients having increased unintegrated forms of HIV-1 DNA.8 Although controversial,17 these results imply that the integrase-inhibitor intensification might have slightly reduced replication. Complete blockage of new rounds of infection is likely to be an important starting point for future studies aimed at reducing the size of the latent reservoir.

The Berlin patient and bone marrow transplantation

One person is widely believed to have been cured of HIV-1 infection.18—20 Although anecdotal, the case is instructive and provides hope that other, safer strategies could be developed in the future. The patient, a 40-year-old HIV-positive man living in Berlin, developed acute myeloid leukaemia and received a bone-marrow transplant from a donor with a homozygous mutation (Δ32) in the gene encoding C-C chemokine receptor 5, CCR5, which renders the donor cells highly resistant to infection from most HIV-1 strains (figure 1). He stopped antiretroviral therapy and detectable HIV viraemia did not return. Intensive efforts to detect residual HIV-1 from several tissues generally resulted in undetectable or barely detectable HIV-1 DNA or RNA.18

 Click to toggle image size 

Click to toggle image size

Figure 1 Full-size image (48K) Download to PowerPoint

Timeline for treatment of the Berlin patient

The grey dotted line represents the limit of detection (one copy per mL) in tests used after transplantation. AML=acute myeloid leukaemia. cART=combined antiretroviral therapy.

According to a report21 from 2012, two HIV-positive patients with lymphoma who were on antiretroviral therapy received allogeneic bone marrow transplants (but from donors who were not resistant to HIV), and HIV-1 was no longer detectable in either patient. Although these patients have not interrupted their antiretroviral therapy and cure is unconfirmed, these additional cases raise the possibility that factors other than HIV- resistance conferred by the transplanted bone marrow could have cured the Berlin patient. Factors that could contribute to the elimination of HIV latency after bone marrow transplantation include graft-versus-host disease, the use of immunosuppressive drugs, and a possible effect of the hosts being heterozygous for the CCR5 mutation. Although bone marrow transplantation is not a method that could be implemented widely to cure HIV, investigation of the most relevant factors that reduce or eliminate HIV latency in this setting could lead to safer and more suitable alternatives.

Strategies to purge latently infected cells

Several interventions in patients who are on stable, long-term antiretroviral therapy have been studied for their effect on the latent viral reservoir. Interleukin 2 activates CD4 T cells harbouring proviral HIV-1 DNA that, in theory, could be cleared with potent combination antiretroviral therapy.22 Interleukin-2 therapy showed promise in reducing the latent HIV reservoir in small studies,22 but in larger clinical trials did not reduce the latent virus reservoir.23 The anti-T-cell antibody muromonab-CD3 (also known as OKT3) also causes direct T-cell activation, but like interleukin 2 did not reduce HIV latency or improve clinical outcomes. Both agents were associated with substantial toxic effects.2425 Interleukin 7 can reactivate HIV-1 in vitro,26 although clinical trials of the molecule in patients on antiretroviral therapy have not shown any reduction in the size of the latent HIV-1 reservoir.27 Several groups are studying the ability of other agents (particularly histone deacetylase inhibitors) to activate latent HIV-1 transcription from resting CD4 T cells, and clinical trials are in progress to assess these agents.28

Identification of appropriate strategies for viral reactivation has proceeded in two ways. First, new drug discovery has been aimed at identification of agents that through minimum signalling can activate virus expression from latently infected cells without cellular or immune activation. This approach depends on appropriate models for latency. The gold standard for cellular target remains cells that were latently infected in vivo. However, this approach is not always practical and screening assays that use resting primary CD4 T cells infected in vitro have also been used.2930 Latently infected cell lines, although useful for defining the mechanisms that control latency,31 are oligoclonal and poorly mimic the variation in viral integration sites seen in vivo and in primary cell models.32 Until comparisons of in-vivo and in-vitro effects are available through clinical trials, the relative usefulness of drug screening assays for inhibition of HIV-1 latency will remain unclear. Despite these limitations, candidate agents have been identified, including those that act on NF-κB pathways of DNA transcription (eg, prostratin), epigenetic modifiers (eg, histone deacetylase inhibitors), inhibitors of DNA methylation, and agents (eg, disulfiram) that target the protein kinase AKT, which is involved in cell proliferation.33 A large array of small molecules that inhibit HIV-1 latency in at least some in-vitro models is now available.34

The second strategy is to move rapidly to clinical trials with existing, licensed therapeutic drugs that have the ability to reactivate the virus. Patients whose infections are suppressed with antiretroviral therapy are currently being enrolled in early clinical trials for treatment with viral activators. Preliminary results of one such study are promising,35 and further research should be done to define the appropriate doses and durations needed to achieve effective clearance of the latent reservoir. The histone deacetylase inhibitor valproic acid was one of the first drugs used, but it is a weak inhibitor of histone deacetylase at doses achievable in the clinic.36 Valproic acid showed promise in an initial investigation37 in which it was used together with intensified antiretroviral therapy. However, subsequent studies showed that valproic acid had no effect on the reservoir, emphasising that the relation between in-vitro and in-vivo activity of latency-activating agents is as yet unclear (table).35,37—41 Potentially more potent histone deacetylase inhibitors, such as vorinostat, have recently been assessed in clinical trials in the USA with a single dose35 and in Australia with a regimen of 400 mg per day (the licensed daily dose of the drug when used for the treatment of cutaneous T-cell lymphoma) for 14 days (NCT01365065). The histone deacetylase inhibitors panobinostat and romidepsin are both more potent than vorinostat in the activation of latent HIV-1 in vitro, and a clinical trial of panobinostat has started in Denmark (NCT01680094).

Click to open table 

TableTable imageOpens in a new browser window

Clinical studies of drugs to reduce viral latency by activating the latent virus

Inhibitors of DNA methylation such as decitabine are used in cancer chemotherapy and can modify HIV-1 expression,31 but are not yet in clinical trials for HIV-1. Disulfiram, a drug long used to treat alcoholism because of its inhibition of acetaldehyde dehydrogenase, might inhibit DNA methyltransferase and activate latent HIV.42 It is currently being assessed in a clinical trial with a regimen of 500 mg per day for 14 days (NCT01286259). Recent in-vitro work has shown that disulfiram activates latent HIV-1 via activation of the protein kinase AKT.33 Prostratin activates the NF-κB pathway and latent HIV-1 in vitro, but because of its serious toxic effects cannot be assessed in clinical trials. Development of activators of protein kinase C such as bryostatin or bryostatin analogues could be an alternative approach.43 Synergistic effects between histone deacetylase inhibitors, methylation inhibitors, histone methyltransferase inhibitors, activators of NF-κB, and interleukin 7 have all been shown in vitro,3044 but such combinations remain unexplored in clinical studies so far.

Gene therapy approaches

The curing of the Berlin patient suggests that clearance of HIV-1 can be achieved through repopulation with CCR5-deficient haemopoietic cells, even in the presence of what seemed to be small amounts of virus using CXCR4 before bone marrow transplantation.18 This finding raises important questions about the relative contribution of the total body irradiation used during transplantation compared with the contribution of low CCR5 expression per se. The 2012 report21 of the clearance of HIV-1 DNA after allogeneic stem cell transplantation for lymphoma (in the absence of CCR5 mutations in the donor) raises the possibility that transplantation, potentially enhanced by graft-versus-host disease, could contribute to clearance of latently infected cells.

Two separate but related approaches for gene therapy are also being explored (figure 2). The first uses zinc-finger nucleases transfected into either autologous CD34-positive stem cells or expanded populations of CD4 T cells that are reinfused back into the patient.45 The zinc finger targets CCR5 and permanently modifies it or truncates it within the genome of the transduced cells, thereby providing a pool of cells permanently resistant to HIV-1 infection (figure 2). CCR5 depletion with zinc-finger nucleases that target CCR5 effectively reduces HIV-1 in mouse models.46 An initial clinical trial showed a possible reduction in viral rebound after infusion of CCR5-deficient cells and structured treatment interruption in six patients.47 Reduction of CCR5 expression was associated with control of viraemia—one patient who had the most substantial CCR5 depletion had undetectable plasma viraemia by week 12. Although an important first step, larger studies with more robust reductions in setpoint viral load in patients who are off antiretroviral therapy are needed. A second approach is to knock down translation of CCR5 by use of a short interfering RNA (siRNA) that is retrovirally transduced into autologous CD34-positive or CD4 T cells (figure 2); this approach has also shown promise in preclinical investigation.48 Phase 1 trials of this method are expected to start in 2013.

 Click to toggle image size 

Click to toggle image size

Figure 2 Full-size image (104K) Download to PowerPoint

Gene therapy approaches to reduce HIV latency and cure HIV

HIV usually enters cells by use of the CCR5 and CD4 coreceptors (A). Zinc fingers nucleases (ZFN) expressed by recombinant adenoviruses can block CCR5 gene expression, rendering the cell devoid of CCR5 and resistant to HIV. Alternatively, short hairpin RNAs (shRNAs) expressed by lentivirus vectors can degrade CCR5 RNA, also rendering the cell devoid of CCR5 and resistant to HIV, through expression of a short interfering RNA (siRNA) that binds to the CCR5 messenger RNA (mRNA). Transcription of HIV is favoured by an open chromatin structure in the nucleus, shown by widely spaced histones (B, left). siRNA can bind to the RNA-induced transcriptional silencing (RITS) complex, which can result in chromatin compaction and silencing of HIV transcription (right), potentially leading to a permanent latent state for HIV in the cell. AGO1=argonaute RISC catalytic component 1. HDAC1=histone deacetylase 1. H3K9me=trimethylation of histone H3 at lysine 9.

An alternative gene therapy-based approach is to enforce latency so that viral rebound does not occur if antiretroviral therapy is stopped. This outcome could be achieved by targeting the HIV-1 promoter with siRNAs or short hairpin RNAs that induce transcriptional gene silencing of the virus via induction of stable epigenetic changes in the integrated viral genome, particularly in the viral promoter or 5 long terminal repeat (figure 2).49 Sustained expression of these constructs in immune cells might allow a functional cure through the induction of long-term latency of the virus, resistant to reactivation by inflammatory, proliferative, and homoeostatic stimuli. Although this approach is encouraging in vitro, it is many years away from reaching the clinic.

Immune modulation and immune effector mechanisms

Immunity is also likely to have an important role in controlling HIV latency. Several studies have examined therapeutic vaccines for their ability to control HIV in the absence of antiretroviral therapy. Most studies into therapeutic vaccines have focused on cytotoxic T-lymphocyte responses by attempting to boost and broaden HIV-1-specific CD4 and CD8 T-cell responses, but with little success to date.50—57 In the context of virus reactivation by drugs such as histone deacetylase inhibitors, antiretroviral therapy blocks the infection of new cells after reactivation. However, little is known about the fate of the reactivated cells. Reactivated latently infected CD4 T cells might die as a result of viral cytopathic effect or elimination by the host immune responses, but without active elimination of the cell, a risk remains that the reactivated virus-expressing cells will return to latency.

Resting CD4 T cells latently infected with HIV-1 in vitro do not die after virus reactivation by the histone deacetylase inhibitor vorinostat,5859 which suggests that reactivation alone will not purge the viral latent reservoir. However, Shan and colleagues58 have shown that when HIV-specific cytotoxic T lymphocytes were first stimulated in vitro, they efficiently killed latently infected cells reactivated by the histone deacetylase inhibitor. Although data are limited to one in-vitro study, this finding adds credence to the idea of using therapeutic vaccines to activate cytotoxic T lymphocyte responses together with agents that reactivate latently infected cells. The induction of potent cytotoxic T lymphocyte responses in HIV-infected patients might, however, be difficult with present HIV vaccine strategies. The improvement of therapeutic vaccines for the induction of such responses is one possible approach that could be pursued.6061 For example, dendritic-cell-based vaccines have shown promise in this respect, although these can only be given in highly specialised centres.62 Conjugation of vaccines to anti-dendritic-cell antibodies to target the induction of potent cytotoxic T-lymphocyte responses is a simpler approach that showed promise in preclinical studies63 and is being investigated in a clinical trial (NCT01127464). The efficacy of live cytomegalovirus vector vaccines for the control of simian immunodeficiency virus (SIV) infection in macaques suggests a possible role for similar cytomegalovirus vectors as therapeutic vaccines for HIV-1 in future studies.64 Unfortunately, some of the most effective HIV-specific cytotoxic T lymphocytes are restricted by fairly uncommon HLA class I alleles (eg, HLA-B*27, HLA-B*57) and many existing responses in patients will have already forced viral escape early in the infection.65

An alternative, non-MHC-restricted immune response that might recognise and eliminate reactivated latently infected cells is antibody-dependent cellular cytotoxicity (ADCC). HIV-specific ADCC antibodies mediate the killing of infected cells by binding to viral antigens on the surface of infected cells and engaging innate immune cells such as natural killer cells or monocytes through their Fc receptors.66 However, little is known about the effect of long-term antiretroviral therapy on HIV-specific ADCC. HIV-specific ADCC antibody responses are often high in patients with very slowly progressing HIV infection.6768 Natural killer cell effectors are, however, decreased in number and can become dysfunctional in patients with progressive HIV disease, although results of recent studies suggest that function of natural killer cells is largely preserved, particularly in patients on effective antiretroviral therapy.69—77 We speculate that HIV-specific ADCC antibodies could potentially be induced to help to clear reactivated latently infected cells by use of protein therapeutic vaccines combined with potent adjuvants.

Another approach is to target cell-surface molecules that are highly expressed on cells that harbour latent HIV. The programmed cell death receptor PD-1 (also known as PDCD1) is highly expressed on so-called exhausted CD4 T cells that are resistant to activation and harbour latent HIV infection.78—80 The use of antibodies to inhibit PD-1 and its ligand has recently shown substantial activity in cancer trials8182 and some promise for the reduction of immune activation in macaques infected with SIV.83 Although such agents will have toxic effects, they could prove useful adjuncts to latency-eradication strategies.

Immune activation and latent HIV

Active HIV-1 infection is associated with increased immune activation. Antiretroviral therapy reduces immune activation, but not to normal. The residual immune activation is associated with residual long-term morbidity, despite antiretroviral therapy.84 Immune activation despite antiretroviral therapy might have a role in the promotion of low-level HIV replication and reseeding of the latent HIV-1 reservoir, although this idea remains controversial. The investigators of the study of raltegravir intensification of antiretroviral therapy noted that the increase in non-integrated HIV-1 forms after the intervention (which implies continued low-level replication) was largely confined to patients with increased immune activation despite antiretroviral therapy.8 This finding lends support to the idea that immune activation might drive continued viral replication in patients on antiretroviral therapy.

Drug treatments to reduce chronic inflammation are of interest for several diseases,85 and such treatments are being investigated for HIV-induced immune activation in several small studies. These studies have two goals: to improve long-term morbidity and to reduce the continuing low-level HIV-1 replication that occurs despite antiretroviral therapy, which would allow more efficient decay or clearance of latently infected cells. The gut-associated lymphoid tissues are damaged during HIV-1 infection and probably contribute to persistent immune activation. Drugs being investigated in clinical trials to reduce gut-damage associated immune activation and so potentially to reduce latent HIV-1 reservoirs include probiotics (NCT01439841),86 anti-lipopolysaccharide antibodies,87 the bowel anti-inflammatory agent mesalazine (NCT01090102), and the antibiotic rifaximin (NCT01466595). Research into treatment with agents such at valganciclovir to reduce herpesvirus-induced immune activation common in patients with HIV-1 is at the early proof-of-concept stage.88 Another approach being investigated in a clinical trial is the reduction of inflammation and thereby the HIV reservoir by use of the angiotensin-converting-enzyme inhibitor lisinopril (NCT01535235). Whether approaches to reduce immune activation in HIV-1 infection will reduce the latent reservoir of HIV DNA is unknown.

Timing of interventions to reduce HIV latency

The timing of when antiretroviral therapy is started after HIV-1 infection might affect the extent of latency during treatment. Patients with acute HIV-1 infection treated with antiretroviral drugs have fewer latently infected cells, and the extent of latent infection seems to be associated with the size of the viral load and duration of viraemia before antiretroviral therapy.7 Additionally, a small subset of patients given antiretroviral therapy during acute infection can control HIV well when treatment is stopped.47 Although such individuals are difficult to identify for recruitment to clinical trials, they could represent an opportunity to explore more effective interventions against latency.

A second question is when to give interventions to reduce latently infected cells relative to the timing of antiretroviral therapy. Studies are currently done in patients who are on long-term antiretroviral therapy. This approach has the advantage of minimising the risk that any HIV-1 activation could reseed the latent reservoir. On the other hand, this approach might mean that the latent pool is targeted at a time when it is more resistant to activation. Indeed, the notion of a stable population of latently infected CD4 T cells has historically been defined in the context of long-term antiretroviral therapy. However, some researchers have questioned whether such a stable reservoir of HIV-1 DNA is also present during active infection,89 because of the high immune activation. If the latent HIV-1 DNA pool is much more labile during active infection, then this possibility presents a potential avenue for intervention.

We recently studied the turnover of viral DNA in resting CD4 T cells in active SIV infection of macaques.90 When viral loads were low, the turnover of the putative latent reservoir was very slow (many years), which is consistent with the low turnover of virus in patients with HIV-1 on treatment. However, in animals with high viral loads, the turnover of SIV DNA within resting CD4 T cells was fast (days), which suggests that high immune activation during active infection might prevent the establishment of true latency. This idea is supported by findings from a study in patients after the initiation of antiretroviral therapy that suggested rapid exchange between productively and latently infected cell pools during both chronic and acute infection.91 Additionally, studies in HIV-infected patients show a delay between the appearance of mutations in plasma and their appearance in the proviral DNA, both for immune-adaptive mutations92 and drug-resistance mutations.93—95

These findings suggest a new strategy to address the reduction or elimination of the viral reservoir. Since many agents currently under investigation aim to purge the latent reservoir through activation of latent cells, targeting the reservoir when it is already turning over rapidly during active infection might be easier to achieve than when it is stable under long-term antiretroviral therapy. The possible effects of giving a latency-activating drug at different times relative to antiretroviral therapy can be investigated by use of a simple model of HIV infection (figure 3).9697 The model predicts that the short-term treatment with latency activators would have the strongest effect on latent HIV-1 DNA if given immediately before commencement of antiretroviral therapy, when it enhances the already existing high activation rate of latent cells caused by the high viral load. Of course, the precise mechanisms of action of a putative latency-activating drug are uncertain, as is whether these would work synergistically with immune activation (as is assumed here). Moreover, whether the reduction in latent HIV would be sufficient to reliably yield a functional cure in the absence of antiretroviral therapy remains unknown, but studies aimed at the incremental reduction of the reservoir size should ultimately answer such questions.

 Click to toggle image size

Click to toggle image size

Figure 3 Full-size image (45K) Download to PowerPoint

Possible effects of early or late treatment with latency activators

We used a mathematical model of viral replication to explore how the timing of administration of a latency-activating drug could potentially affect the latent reservoir (A). The model is based on the assumption that the drug triples the activation rate and is given for a short time (3 days), either immediately before initiation of or after long-term use of combined antiretroviral therapy (cART). If the latency activator is given after long-term use of cART (B) it has a small effect on number of latently infected cells, because the cells are mostly quiescent. If the same latency activator is given for 3 days immediately before the start of cART (C), it causes a large decrease in the number of latently infected cells, because the cells already have a high reactivation rate. The model is adapted from the standard model of HIV infection9697 such that a proportion of infected CD4 T cells (1-f) enters the latent pool (L). Latent cells do not produce virus or die, but can be activated to become productively infected cells at a rate proportional to the viral load (αV, where α is the activation constant). cART blocks new infections—the equivalent of setting susceptibility β to zero (on the assumption that the treatment is 100% effective). Parameters used in the figure: λ=10 cells/mL per day; f=0·95; dT=0·01 per day; α=1·25 × 10−7; β=8 × 10−8 mL/copy per day; p=800 copies/cell per day; δ=0·8 per day; c=20 per day.

The timing-of-cure approaches with respect to initiation of antiretroviral therapy could hold some promise, but they are not without their own risks. A very real potential exists for toxic effects from attempts to cure at the time of starting antiretroviral therapy, since cellular activation, the loss of a key subset of immune cells, and potentially further increases of HIV-1 replication could be transiently induced. Paradoxically, the results of our previous study90 and our model (figure 3) suggest that the patients who could benefit the most from such an intervention would be those with high levels of viral replication—ie, those who are likely to have a high turnover of HIV-1 DNA within resting CD4 T cells, making this reservoir more vulnerable.

Conclusions

Innovative strategies to reduce or eliminate latent HIV-1 infection are being pursued to reduce the global burden of HIV and costs and adverse effects associated with lifelong antiretroviral therapy. Several different strategies are in early stage clinical trials. Approaches that attempt to reduce latent HIV at the time that antiretroviral therapy is started should also be investigated.

Search strategy and selection criteria

We searched PubMed and ClinicalTrials.gov for reports about cures for HIV and about HIV latency to identify clinically applicable approaches. We used the search terms “HIV latency”, “HIV cure”, and “latent HIV” and reviewed articles published in English between Jan 1, 1990 and Dec 1, 2012.

Contributors

All authors contributed equally.

Conflicts of interest

SRL has received honoraria and speaking fees from Gilead, Merck, and ViiV Healthcare. She is a principal investigator of investigator-initiated studies sponsored by Gilead, Merck, and Janssen. SE receives research grant support from Abbott, Merck, Pfizer, and ViiV Healthcare. ADK has received an investigator grant for work on a clinical trial and payment for attendance at an international conference from Merck. All other authors declare that they have no conflicts of interest.

References

1 Siliciano JD, Siliciano RF. The latent reservoir for HIV-1 in resting CD4+ T cells: a barrier to cure. Curr Opin HIV AIDS 2006; 1: 121-128. PubMed

2 Chun TW, Fauci AS. HIV reservoirs: pathogenesis and obstacles to viral eradication and cure. AIDS 2012; 26: 1261-1268. CrossRef | PubMed

3 Finzi D, Blankson J, Siliciano JD, et al. Latent infection of CD4+ T cells provides a mechanism for lifelong persistence of HIV-1, even in patients on effective combination therapy. Nat Med 1999; 5: 512-517. CrossRef | PubMed

4 Schmid A, Gianella S, von Wyl V, et al. Profound depletion of HIV-1 transcription in patients initiating antiretroviral therapy during acute infection. PLoS One 2010; 5: e13310. CrossRef | PubMed

5 Ananworanich J, Schuetz A, Vandergeeten C, et al. Impact of multi-targeted antiretroviral treatment on gut T cell depletion and HIV reservoir seeding during acute HIV infection. PLoS One 2012; 7: e33948. CrossRef | PubMed

6 Archin NM, Vaidya NK, Kuruc JD, et al. Immediate antiviral therapy appears to restrict resting CD4+ cell HIV-1 infection without accelerating the decay of latent infection. Proc Natl Acad Sci USA 2012; 109: 9523-9528. PubMed

7 Koelsch KK, Boesecke C, McBride K, et al. Impact of treatment with raltegravir during primary or chronic HIV infection on RNA decay characteristics and the HIV viral reservoir. AIDS 2011; 25: 2069-2078. CrossRef | PubMed

8 Buzon MJ, Massanella M, Llibre JM, et al. HIV-1 replication and immune dynamics are affected by raltegravir intensification of HAART-suppressed subjects. Nat Med 2010; 16: 460-465. CrossRef | PubMed

9 Dinoso JB, Kim SY, Wiegand AM, et al. Treatment intensification does not reduce residual HIV-1 viremia in patients on highly active antiretroviral therapy. Proc Natl Acad Sci USA 2009; 106: 9403-9408. PubMed

10 Gandhi RT, Bosch RJ, Aga E, et al. No evidence for decay of the latent reservoir in HIV-1-infected patients receiving intensive enfuvirtide-containing antiretroviral therapy. J Infect Dis 2010; 201: 293-296. CrossRef | PubMed

11 Richman DD, Margolis DM, Delaney M, Greene WC, Hazuda D, Pomerantz RJ. The challenge of finding a cure for HIV infection. Science 2009; 323: 1304-1307. CrossRef | PubMed

12 Siliciano RF, Greene WC. HIV latency. Cold Spring Harb Perspect Med 2012; 1: a007096. PubMed

13 Chun TW, Carruth L, Finzi D, et al. Quantification of latent tissue reservoirs and total body viral load in HIV-1 infection. Nature 1997; 387: 183-188. CrossRef | PubMed

14 Wong JK, Hezareh M, Günthard HF, et al. Recovery of replication-competent HIV despite prolonged suppression of plasma viremia. Science 1997; 278: 1291-1295. CrossRef | PubMed

15 Finzi D, Hermankova M, Pierson T, et al. Identification of a reservoir for HIV-1 in patients on highly active antiretroviral therapy. Science 1997; 278: 1295-1300. CrossRef | PubMed

16 Chomont N, El-Far M, Ancuta P, et al. HIV reservoir size and persistence are driven by T cell survival and homeostatic proliferation. Nat Med 2009; 15: 893-900. CrossRef | PubMed

17 Chege D, Kovacs C, la Porte C, et al. Effect of raltegravir intensification on HIV proviral DNA in the blood and gut mucosa of men on long-term therapy: a randomized controlled trial. AIDS 2012; 26: 167-174. CrossRef | PubMed

18 Allers K, Hütter G, Hofmann J, et al. Evidence for the cure of HIV infection by CCR5Delta32/Delta32 stem cell transplantation. Blood 2011; 117: 2791-2799. CrossRef | PubMed

19 Hütter G, Nowak D, Mossner M, et al. Long-term control of HIV by CCR5 Delta32/Delta32 stem-cell transplantation. N Engl J Med 2009; 360: 692-698. CrossRef | PubMed

20 Hütter G, Thiel E. Allogeneic transplantation of CCR5-deficient progenitor cells in a patient with HIV infection: an update after 3 years and the search for patient no 2. AIDS 2011; 25: 273-274. CrossRef | PubMed

21 Henrich TJ, Sciaranghella G, Li JZ, et al. Long-term reduction in peripheral blood HIV-1 reservoirs following reduced-intensity conditioning allogeneic stem cell transplantation in two HIV-positive individuals. 19th International AIDS Conference; Washington, DC; July 22—27, 2012. THAA0101.

22 Chun TW, Engel D, Mizell SB, et al. Effect of interleukin-2 on the pool of latently infected, resting CD4+ T cells in HIV-1-infected patients receiving highly active anti-retroviral therapy. Nat Med 1999; 5: 651-655. CrossRef | PubMed

23 Stellbrink HJ, van Lunzen J, Westby M, et al. Effects of interleukin-2 plus highly active antiretroviral therapy on HIV-1 replication and proviral DNA (COSMIC trial). AIDS 2002; 16: 1479-1487. CrossRef | PubMed

24 Abrams D, Levy Y, Losso MH, et al. Interleukin-2 therapy in patients with HIV infection. N Engl J Med 2009; 361: 1548-1559. CrossRef | PubMed

25 Prins JM, Jurriaans S, van Praag RM, et al. Immuno-activation with anti-CD3 and recombinant human IL-2 in HIV-1-infected patients on potent antiretroviral therapy. AIDS 1999; 13: 2405-2410. CrossRef | PubMed

26 Wang FX, Xu Y, Sullivan J, et al. IL-7 is a potent and proviral strain-specific inducer of latent HIV-1 cellular reservoirs of infected individuals on virally suppressive HAART. J Clin Invest 2005; 115: 128-137. CrossRef | PubMed

27 Imamichi H, Degray G, Asmuth DM, et al. HIV-1 viruses detected during episodic blips following interleukin-7 administration are similar to the viruses present before and after interleukin-7 therapy. AIDS 2011; 25: 159-164. CrossRef | PubMed

28 Wightman F, Ellenberg P, Churchill M, Lewin SR. HDAC inhibitors in HIV. Immunol Cell Biol 2012; 90: 47-54. CrossRef | PubMed

29 Bosque A, Planelles V. Induction of HIV-1 latency and reactivation in primary memory CD4+ T cells. Blood 2009; 113: 58-65. CrossRef | PubMed

30 Saleh S, Wightman F, Ramanayake S, et al. Expression and reactivation of HIV in a chemokine induced model of HIV latency in primary resting CD4+ T cells. Retrovirology 2011: B80.

31 Fernandez G, Zeichner SL. Cell line-dependent variability in HIV activation employing DNMT inhibitors. Virol J 2010; 7: 266. CrossRef | PubMed

32 Gallastegui E, Marshall B, Vidal D, et al. Combination of biological screening in a cellular model of viral latency and virtual screening identifies novel compounds that reactivate HIV-1. J Virol 2012; 86: 3795-3808. CrossRef | PubMed

33 Doyon G, Zerbato J, Mellors JW, Sluis-Cremer N. Disulfiram reactivates latent HIV-1 expression through depletion of the phosphatase and tensin homolog. AIDS 2013; 27: F7-11. CrossRef | PubMed

34 Xing S, Siliciano RF. Targeting HIV latency: pharmacologic strategies toward eradication. Drug Discov Today 201210.1016/j.drudis.2012.12.008. published online Dec 25. PubMed

35 Archin NM, Liberty AL, Kashuba AD, et al. Administration of vorinostat disrupts HIV-1 latency in patients on antiretroviral therapy. Nature 2012; 487: 482-485. CrossRef | PubMed

36 Shehu-Xhilaga M, Rhodes D, Wightman F, et al. The novel histone deacetylase inhibitors metacept-1 and metacept-3 potently increase HIV-1 transcription in latently infected cells. AIDS 2009; 23: 2047-2050. CrossRef | PubMed

37 Lehrman G, Hogue IB, Palmer S, et al. Depletion of latent HIV-1 infection in vivo: a proof-of-concept study. Lancet 2005; 366: 549-555. Summary | Full Text | PDF(97KB) | CrossRef | PubMed

38 Siliciano JD, Lai J, Callender M, et al. Stability of the latent reservoir for HIV-1 in patients receiving valproic acid. J Infect Dis 2007; 195: 833-836. CrossRef | PubMed

39 Sagot-Lerolle N, Lamine A, Chaix ML, et al. Prolonged valproic acid treatment does not reduce the size of latent HIV reservoir. AIDS 2008; 22: 1125-1129. CrossRef | PubMed

40 Archin NM, Cheema M, Parker D, et al. Antiretroviral intensification and valproic acid lack sustained effect on residual HIV-1 viremia or resting CD4+ cell infection. PLoS One 2010; 5: e9390. CrossRef | PubMed

41 Routy JP, Tremblay CL, Angel JB, et al. Valproic acid in association with highly active antiretroviral therapy for reducing systemic HIV-1 reservoirs: results from a multicentre randomized clinical study. HIV Med 2012; 13: 291-296. CrossRef | PubMed

42 Xing S, Bullen CK, Shroff NS, et al. Disulfiram reactivates latent HIV-1 in a Bcl-2-transduced primary CD4+ T cell model without inducing global T cell activation. J Virol 2011; 85: 6060-6064. CrossRef | PubMed

43 DeChristopher BA, Loy BA, Marsden MD, Schrier AJ, Zack JA, Wender PA. Designed, synthetically accessible bryostatin analogues potently induce activation of latent HIV reservoirs in vitro. Nat Chem 2012; 4: 705-710. PubMed

44 Bouchat S, Gatot JS, Kabeya K, et al. Histone methyltransferase inhibitors induce HIV-1 recovery in resting CD4(+) T cells from HIV-1-infected HAART-treated patients. AIDS 2012; 26: 1473-1482. CrossRef | PubMed

45 Perez EE, Wang J, Miller JC, et al. Establishment of HIV-1 resistance in CD4+ T cells by genome editing using zinc-finger nucleases. Nat Biotechnol 2008; 26: 808-816. CrossRef | PubMed

46 Holt N, Wang J, Kim K, et al. Human hematopoietic stem/progenitor cells modified by zinc-finger nucleases targeted to CCR5 control HIV-1 in vivo. Nat Biotechnol 2010; 28: 839-847. CrossRef | PubMed

47 Positive results with autologous transplant of ZFN-modified CD4 T-cells: a step toward a practical, functional cure for HIV. AIDS Patient Care STDS 2011; 25: 693-695. CrossRef | PubMed

48 Shimizu S, Hong P, Arumugam B, et al. A highly efficient short hairpin RNA potently down-regulates CCR5 expression in systemic lymphoid organs in the hu-BLT mouse model. Blood 2010; 115: 1534-1544. CrossRef | PubMed

49 Suzuki K, Juelich T, Lim H, et al. Closed chromatin architecture is induced by an RNA duplex targeting the HIV-1 promoter region. J Biol Chem 2008; 283: 23353-23363. CrossRef | PubMed

50 Papagno L, Alter G, Assoumou L, et al. Comprehensive analysis of virus-specific T-cells provides clues for the failure of therapeutic immunization with ALVAC-HIV vaccine. AIDS 2011; 25: 27-36. CrossRef | PubMed

51 Autran B, Costagliola D, Murphy R, Katlama C. Evaluating therapeutic vaccines in patients infected with HIV. Expert Rev Vaccines 2004; 3 (suppl 4): S169-S177. CrossRef | PubMed

52 Autran B, Kinloch-de Loes S, Katlama C. Therapeutic immunization in HIV infection. Curr Opin HIV AIDS 2006; 1: 323-329. CrossRef | PubMed

53 Tubiana R, Carcelain G, Vray M, et al. Therapeutic immunization with a human immunodeficiency virus (HIV) type 1-recombinant canarypox vaccine in chronically HIV-infected patients: The Vacciter Study (ANRS 094). Vaccine 2005; 23: 4292-4301. CrossRef | PubMed

54 Kinloch-de Loes S, Hoen B, Smith DE, et al. Impact of therapeutic immunization on HIV-1 viremia after discontinuation of antiretroviral therapy initiated during acute infection. J Infect Dis 2005; 192: 607-617. CrossRef | PubMed

55 Fuller DH, Rajakumar P, Che JW, et al. Therapeutic DNA vaccine induces broad T cell responses in the gut and sustained protection from viral rebound and AIDS in SIV-infected rhesus macaques. PLoS One 2011; 7: e33715. CrossRef | PubMed

56 Emery S, Kelleher AD, Workman C, et al. Influence of IFNγ co-expression on the safety and antiviral efficacy of recombinant fowlpox virus HIV therapeutic vaccines following interruption of antiretroviral therapy. Hum Vaccin 2007; 3: 260-267. PubMed

57 García F, León A, Gatell JM, Plana M, Gallart T. Therapeutic vaccines against HIV infection. Hum Vaccin Immunother 2012; 8: 569-581. PubMed

58 Shan L, Deng K, Shroff NS, et al. Stimulation of HIV-1-specific cytolytic T lymphocytes facilitates elimination of latent viral reservoir after virus reactivation. Immunity 2012; 36: 491-501. CrossRef | PubMed

59 Migueles SA, Connors M. Small molecules and big killers: the challenge of eliminating the latent HIV reservoir. Immunity 2012; 36: 320-321. CrossRef | PubMed

60 Garcia F, Leon A, Gatell JM, Plana M, Gallart T. Therapeutic vaccines against HIV infection. Hum Vaccin Immunother 2012; 8: 569-581. PubMed

61 De Rose R, Fernandez CS, Smith MZ, et al. Control of viremia and prevention of AIDS following immunotherapy of SIV-infected macaques with peptide-pulsed blood. PLoS Pathog 2008; 4: e1000055. CrossRef | PubMed

62 Garcia F, Climent N, Assoumou L, et al. A therapeutic dendritic cell-based vaccine for HIV-1 infection. J Infect Dis 2011; 203: 473-478. CrossRef | PubMed

63 Idoyaga J, Lubkin A, Fiorese C, et al. Comparable T helper 1 (Th1) and CD8 T-cell immunity by targeting HIV gag p24 to CD8 dendritic cells within antibodies to Langerin, DEC205, and Clec9A. Proc Natl Acad Sci USA 2011; 108: 2384-2389. PubMed

64 Hansen SG, Ford JC, Lewis MS, et al. Profound early control of highly pathogenic SIV by an effector memory T-cell vaccine. Nature 2011; 473: 523-527. CrossRef | PubMed

65 Goulder PJ, Watkins DI. HIV and SIV CTL escape: implications for vaccine design. Nat Rev Immunol 2004; 4: 630-640. PubMed

66 Chung A, Rollman E, Johansson S, Kent SJ, Stratov I. The utility of ADCC responses in HIV infection. Curr HIV Res 2008; 6: 515-519. CrossRef | PubMed

67 Forthal DN, Landucci G, Daar ES. Antibody from patients with acute human immunodeficiency virus (HIV) infection inhibits primary strains of HIV type 1 in the presence of natural-killer effector cells. J Virol 2001; 75: 6953-6961. CrossRef | PubMed

68 Chung AW, Navis M, Isitman G, et al. Activation of NK cells by ADCC antibodies and HIV disease progression. J Acquir Immune Defic Syndr 2011; 58: 127-131. CrossRef | PubMed

69 Bruunsgaard H, Pedersen C, Skinhøj P, Pedersen BK. Clinical progression of HIV infection: role of NK cells. Scand J Immunol 1997; 46: 91-95. CrossRef | PubMed

70 De Maria A, Fogli M, Costa P. The impaired NK cell cytolytic function in viremic HIV-1 infection is associated with a reduced surface expression of natural cytotoxicity receptors (NKp46, NKp30 and NKp44). Eur J Immunol 2003; 33: 2410-2418. CrossRef | PubMed

71 Geertsma MF, Stevenhagen A, van Dam EM, Nibbering PH. Expression of zeta molecules is decreased in NK cells from HIV-infected patients. FEMS Immunol Med Microbiol 1999; 26: 249-257. CrossRef | PubMed

72 Hu PF, Hultin LE, Hultin P, et al. Natural killer cell immunodeficiency in HIV disease is manifest by profoundly decreased numbers of CD16+CD56+ cells and expansion of a population of CD16dimCD56- cells with low lytic activity. J Acquir Immune Defic Syndr Hum Retrovirol 1995; 10: 331-340. PubMed

73 Ljunggren K, Karlson A, Fenyö EM, Jondal M. Natural and antibody-dependent cytotoxicity in different clinical stages of human immunodeficiency virus type 1 infection. Clin Exp Immunol 1989; 75: 184-189. PubMed

74 Mavilio D, Benjamin J, Daucher M, et al. Natural killer cells in HIV-1 infection: dichotomous effects of viremia on inhibitory and activating receptors and their functional correlates. Proc Natl Acad Sci USA 2003; 100: 15011-15016. PubMed

75 Sirianni MC, Mezzaromau I, Aiutiu F, Moretta A. Analysis of the cytolytic activity mediated by natural killer cells from acquired immunodeficiency syndrome patients in response to phytohemagglutinin or anti-CD16 monoclonal antibody. Eur J Immunol 1994; 24: 1874-1878. CrossRef | PubMed

76 Björkström NK, Ljunggren H-G, Sandberg JK. CD56 negative NK cells: origin, function, and role in chronic viral disease. Trends Immunol 2010; 31: 401-406. CrossRef | PubMed

77 Johansson SE, Rollman E, Chung AW, et al. NK cell function and antibodies mediating ADCC in HIV-1-infected viremic and controller patients. Viral Immunol 2011; 24: 359-368. CrossRef | PubMed

78 Day CL, Kaufmann DE, Kiepiela P, et al. PD-1 expression on HIV-specific T cells is associated with T-cell exhaustion and disease progression. Nature 2006; 443: 350-354. CrossRef | PubMed

79 Trautmann L, Janbazian L, Chomont N, et al. Upregulation of PD-1 expression on HIV-specific CD8+ T cells leads to reversible immune dysfunction. Nat Med 2006; 12: 1198-1202. CrossRef | PubMed

80 Hatano H, Jain V, Hunt PW, et al. Cell-based measures of viral persistence are associated with immune activation and programmed cell death protein 1 (PD-1)-expressing CD4+ T cells. J Infect Dis 201210.1093/infdis/jis630. published online Oct 22. PubMed

81 Brahmer JR, Tykodi SS, Chow LQ, et al. Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N Engl J Med 2012; 366: 2455-2465. CrossRef | PubMed

82 Topalian SL, Hodi FS, Brahmer JR, et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med 2012; 366: 2443-2454. CrossRef | PubMed

83 Dyavar Shetty R, Velu V, Titanji K, et al. PD-1 blockade during chronic SIV infection reduces hyperimmune activation and microbial translocation in rhesus macaques. J Clin Invest 2012; 122: 1712-1716. CrossRef | PubMed

84 El-Sadr WM, Lundgren J, Neaton JD, et al. CD4+ count-guided interruption of antiretroviral treatment. N Engl J Med 2006; 355: 2283-2296. CrossRef | PubMed

85 Tabas I, Glass CK. Anti-inflammatory therapy in chronic disease: challenges and opportunities. Science 2013; 339: 166-172. CrossRef | PubMed

86 Klatt NR, Harris LD, Vinton CL, et al. Compromised gastrointestinal integrity in pigtail macaques is associated with increased microbial translocation, immune activation, and IL-17 production in the absence of SIV infection. Mucosal Immunol 2010; 3: 387-398. CrossRef | PubMed

87 Byakwaga H, Kelly M, Purcell DF, et al. Intensification of antiretroviral therapy with raltegravir or addition of hyperimmune bovine colostrum in HIV-infected patients with suboptimal CD4+ T-cell response: a randomized controlled trial. J Infect Dis 2011; 204: 1532-1540. CrossRef | PubMed

88 Hunt PW, Martin JN, Sinclair E, et al. Valganciclovir reduces T cell activation in HIV-infected individuals with incomplete CD4+ T cell recovery on antiretroviral therapy. J Infect Dis 2011; 203: 1474-1483. CrossRef | PubMed

89 Deeks SG. HIV: how to escape treatment. Nature 2011; 477: 36-37. CrossRef | PubMed

90 Reece J, Petravic J, Balamurali M, et al. An “escape clock” for estimating the turnover of SIV DNA in resting CD4+ T cells. PLoS Pathog 2012; 8: e1002615. CrossRef | PubMed

91 Murray JM, McBride K, Boesecke C, et al. Integrated HIV DNA accumulates prior to treatment while episomal HIV DNA records ongoing transmission afterwards. AIDS 2012; 26: 543-550. CrossRef | PubMed

92 Zhang L, Rowe L, He T, et al. Compartmentalization of surface envelope glycoprotein of human immunodeficiency virus type 1 during acute and chronic infection. J Virol 2002; 76: 9465-9473. CrossRef | PubMed

93 Strain MC, Gunthard HF, Havlir DV, et al. Heterogeneous clearance rates of long-lived lymphocytes infected with HIV: intrinsic stability predicts lifelong persistence. Proc Natl Acad Sci USA 2003; 100: 4819-4824. PubMed

94 Persaud D, Palumbo P, Ziemniak C, et al. Early archiving and predominance of nonnucleoside reverse transcriptase inhibitor-resistant HIV-1 among recently infected infants born in the United States. J Infect Dis 2007; 195: 1402-1410. CrossRef | PubMed

95 Poveda E, Briz V, Paraskevis D, Barreiro P, Hatzakis A, Soriano V. Dynamics of drug-resistant HIV-1 in plasma and peripheral blood cells in patients during and after enfuvirtide therapy. AIDS Res Hum Retroviruses 2007; 23: 1078-1082. CrossRef | PubMed

96 Nowak MA, May R. Virus dynamics: mathematical principles of immunology and virology. Oxford: Oxford University Press, 2000.

97 Perelson AS. Modelling viral and immune system dynamics. Nat Rev Immunol 2002; 2: 28-36. PubMed

a Department of Microbiology and Immunology, University of Melbourne, VIC, Australia

b Centre for Vascular Research, University of New South Wales, Sydney, NSW, Australia

c Kirby Institute, University of New South Wales, Sydney, NSW, Australia

d Alfred Hospital, Melbourne, VIC, Australia

e Monash University, Melbourne, VIC, Australia

f Burnet Institute, Melbourne, VIC, Australia

Corresponding Author InformationCorrespondence to: Prof S J Kent, Department of Microbiology and Immunology, University of Melbourne, VIC 3010, Australia

-- 
To subscribe or unsubscribe from these Child Survival Updates, pls contact kidsurvival@gmail.com. If you unsubscribe, indicate from which E mail address you are receving these updates.
 
When subscribing, write from your most permanent E-mail address, not always that of your current employer.
 
Do not subscribe on behalf of friends or colleagues; forward updates to them for their decision.

Those wishing to read only malaria updates should subscribe at
kidsurvivalmalaria@gmail.com
 

Those wishing to read only vaccination updates should subscribe at kidsurvivalvaccination@gmail.com
 
READER COMMENTS
 
If you have a comment you want posted, send to rdavis@africamail.com
WEBPAGE
 
These updates are also available at www.childsurvival.net  
-- 
To subscribe or unsubscribe from these Child Survival Updates, pls contact kidsurvival@gmail.com. If you unsubscribe, indicate from which E mail address you are receving these updates.
 
When subscribing, write from your most permanent E-mail address, not always that of your current employer.
 
Do not subscribe on behalf of friends or colleagues; forward updates to them for their decision.

Those wishing to read only malaria updates should subscribe at
kidsurvivalmalaria@gmail.com
 

Those wishing to read only vaccination updates should subscribe at kidsurvivalvaccination@gmail.com
 
READER COMMENTS
 
If you have a comment you want posted, send to rdavis@africamail.com
WEBPAGE
 
These updates are also available at www.childsurvival.net  

41177467