3,161
Views
3
CrossRef citations to date
0
Altmetric
Editorial

Curing HIV by ‘kick and kill’: from theory to practice?

&
Pages 383-386 | Received 18 Jan 2019, Accepted 08 May 2019, Published online: 17 May 2019

1. Introduction

Although antiretroviral therapy (ART) suppresses viremia in HIV infected individuals, it is not a cure, and a treatment-resistant reservoir of latently infected cells remains. If ART is stopped viremia returns, often rapidly. Lifetime ART, if available, involves side effects, cost, and stigma. This has led to a great interest in curing HIV with diverse experimental approaches being considered ranging from therapeutic vaccination to gene therapy to bone marrow transplants with defective CCR5 genes.

One approach for achieving HIV cure has been termed ‘kick and kill’ (or ‘shock and kill’). Here, the reservoir is stimulated to reverse latency, and the now targetable cells are killed. Many of the concepts behind latency reversal were first considered over 25 years ago [Citation1,Citation2], including the role of chromatin acetylation and methylation. However, it has only been in the last few years that these ideas have been tested clinically. Attractive in principle, the concept of ‘kick and kill’ has not been proven. Here, we explore the rationale, ethics, and history of this approach, evidence for its effectiveness to date, and whether it is still a viable strategy or should be abandoned.

2. A ‘kick’ is not enough

Following the recognition of a latent HIV reservoir in resting CD4 T cells, approaches were devised to ‘flush out’ these residual proviruses. Based on the assumption that HIV replication might induce cell death, it was presumed that activating latently infected cells would lead to their destruction [Citation3].

This prompted the first studies exploring IL-2 to activate T cells to both stimulate HIV production and enhance CD4 T cell recovery through increased cell division. Chun et al. [Citation4] showed a decrease in IUPM (‘Infectious Units Per Million’ cells – a technique to measure replication competent provirus) with IL-2 and ART. However, there was no change in HIV DNA levels, possibly impacted by a dominance of replication-incompetent proviral sequences [Citation5,Citation6]. This was, nevertheless, early evidence that activating the HIV reservoir could be successful. However, subsequent studies with IL-2 showed no impact on viral rebound kinetics on stopping ART [Citation7]. Later studies combined OKT3 (a mouse anti-CD3 monoclonal antibody) with IL-2 to enhance T cell activation [Citation8]. While effective at activating T cells and inducing HIV expression, there was no effect on HIV DNA or IUPM [Citation8] suggesting that stimulation alone would not suffice.

Critically, there were significant safety concerns with these latter studies. Safety remains a crucial factor in kick and kill studies. The majority of participants will be well on antiretroviral therapy, with few side effects and effectively normal life expectancies. The decision, therefore, to introduce an experimental agent with potential potent side effects cannot be taken lightly. In addition, there is an increasing feeling that a treatment interruption (TI) is the only meaningful end-point in a clinical trial aimed at achieving remission, and this also raises ethical questions, particularly in regards to risks of onward transmission in viraemic participants. Deciding on the level of risk that the HIV community (patients, clinicians, and researchers) is willing to accept will be critical in moving the HIV cure agenda forward [Citation9,Citation10]. Because of these concerns, subsequent studies incorporated less potent activation strategies focusing on cytokines including IL-10 [Citation11], IFN-γ [Citation12] and IL-7 [Citation13], none of which reduced the reservoir (and some appeared to increase it [Citation14]).

3. Introducing the ‘kill’

Initial ‘kill’ approaches involved boosting immunity using treatment interruption (TI). It was hypothesized that multiple interruptions might act as a form of ‘self-vaccination’ to boost T cell responses. However, this approach only served to demonstrate that natural host HIV-specific immunity was inadequate [Citation15], and that other approaches would be required to effectively target newly-awakened latently infected cells. The history of vaccines in HIV is chequered. There is no effective preventative vaccine and little evidence that therapeutic vaccination is effective at reducing the HIV reservoir in humans [Citation16,Citation17], although there have been promising studies in monkeys [Citation18,Citation19].

‘Kick and Kill’ suggests an alternative role for vaccination; one might discriminate between a vaccine-induced immune response targeting latently infected cells versus one targeting cells actively producing virus, i.e. the difference between asking the immune system to target the reservoir (‘cure’) vs cells actively producing virions. These could require very different mechanisms, and there is a case to be made for the term ‘reservoir’ vaccine, which may require different immune system components beyond CD8 T cells [Citation20].

4. Moving forward in the era of effective ART

With increasingly effective ART, newer anti-latency drugs were sought to drive out HIV expression. These included histone deacetylase inhibitors (HDACi), histone methyltransferase inhibitors, DNA methyltransferase inhibitors, bromodomain inhibitors, PKC agonists, and PI3K/AKT inhibitors as well as – more recently – SMAC mimetics and agonists for TLR7 or TLR9 (reviewed elsewhere [Citation21]).

HDACi have been most widely tested, and clinical trials are ongoing. Early results from in vivo studies [Citation22,Citation23] showed increased HIV RNA transcription but did not translate into a decrease in reservoir size. Ex vivo, HDACi-treated cells are not more susceptible to T cell killing, although if exposed to heterologous highly effective T cells from ‘elite controllers’ there was evidence of increased cytotoxicity [Citation24], arguably consistent with increased viral antigen presentation following HDACi administration.

Enhancing CD8 T cell function was the primary focus of kill efforts, which have now expanded to including other components of cellular immunity, such as natural killer (NK) cells, particularly due to advances in engineering broadly neutralizing antibodies (bNAbs). Infusion of bNAbs reduces viremia, delays viral rebound after ART interruption and enhances infected cell clearance [Citation25]. The proposed role of bNAbs in enhancing NK and T cell function via possible combinations of antibody-dependent cellular cytotoxicity (ADCC), immune complex formation and dendritic cell engagement (the ‘vaccinal effect’) as well as their potential to elicit further effective antibody responses suggest that an optimized ‘kill’ will require multiple arms of the immune system [Citation26,Citation27].

5. Clinical trials and HIV cure

Only a few clinical studies have tested ‘kick and kill’ in vivo, with most employing T cell vaccines with HDACi. In the BCNO2-Romi trial, 15 participants received MVA.HIVconsv vaccination [Citation28] with the HDACi romidepsin. At the time of writing, the results were not published, but at CROI 2017 it was reported that of 11 participants selected to stop ART, four remained off therapy (restart criteria >2000 copies/ml) for 7, 12, 14, and 22 weeks [Citation29]. Follow-up of these individuals will be fascinating, but as short periods of apparent remission can be seen after ART alone, the data are hard to interpret without placebo controls [Citation30]. One study using a different T cell vaccine (Vacc4x with rhuGM-CSF) with romidepsin reported a reduction in both HIV DNA and IUPM, and was the first to demonstrate a potential impact on the reservoir, although again without controls [Citation31]. The first randomized controlled trial using an HDACi with a T cell vaccine was the RIVER trial where patients received a ChAdV63.HIVconsv vaccine with MVA.HIVconsv boost followed by vorinostat [Citation32]. Despite increased histone acetylation and improved T cell function, there was no evidence of a reduced HIV reservoir as measured by HIV DNA (total or integrated) or viral outgrowth [Citation32]. RIVER raises the key question of whether it showed no effect due to the futility of ‘kick and kill’ as a concept, or if the chosen drugs and vaccines were not potent enough.

6. Moving forward

Why have we not seen convincing outcomes from ‘kick and kill’ studies? Several findings may provide insight. A large fraction of integrated HIV DNA is replication competent but not activated in a single stimulation round ex vivo, even with potent stimuli [Citation5]. If all latency cannot be reversed under optimized ex vivo conditions, the likelihood of a safe clinical intervention achieving this is minimal. It may be that the ‘kick’ (and possibly the ‘kill’) need to be given on multiple occasions over many months or years. Current clinical trial designs do not test this and – if we are looking for the cumulative summation of multiple small impacts – may not be sufficiently powered for proof of principle. If the kick is effective and antigen is presented, the immune system might still not provide adequate ‘kill’ due to immune ‘distractions’ caused by replication incompetent viruses producing non cross-reactive viral proteins [Citation33], due to persistent and non-reversible functional T cell exhaustion [Citation34], or to innate resistance of the reservoir to CTL [Citation20]. One of the key barriers to effective immunity – the development of viral immune escape – should be less of an issue for a ‘reservoir’ vaccine, as the absence of circulating viremia on ART should remove from the equation error-prone viral replication and the opportunity to adapt to immune-induced selection pressure. Nevertheless, even if CTL were effective at eliminating reactivated cells, whether these effectors are able to effectively reach the necessary anatomical sites (e.g. germinal centers) in order to reduce the HIV reservoir remains unclear [Citation35]. Thus, if it exists, the right ‘kill’ is likely to be in the form of a combination of multiple immune enhancing approaches, including possible engineered agents such as CAR T cells (reviewed in [Citation21,Citation25]).

7. Conclusions

Progress in achieving HIV remission through ‘kick and kill’ has been limited. Whether this is due to the futility of the concept or the ineffectiveness of currently available agents remains to be seen. However, the premise remains theoretically attractive, and it remains the most scalable approach to reducing the HIV reservoir.

Declaration of interest

The authors have no relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript. This includes employment, consultancies, honoraria, stock ownership or options, expert testimony, grants or patents received or pending, or royalties.

Reviewer disclosures

Peer reviewers on this manuscript have no relevant financial or other relationships to disclose.

Additional information

Funding

This paper was funded by the Medical Research Council UK (Grant Number MR/L006588/1 and MR/P011233/1).

References

  • Verdin E, Paras P Jr., Van Lint C. Chromatin disruption in the promoter of human immunodeficiency virus type 1 during transcriptional activation. Embo J. 1993 Aug;12(8):3249–3259. PubMed PMID: 8344262; PubMed Central PMCID: PMCPMC413592
  • Van Lint C, Emiliani S, Ott M, et al. Transcriptional activation and chromatin remodeling of the HIV-1 promoter in response to histone acetylation. Embo J. 1996 Mar 1;15(5):1112–1120. PubMed PMID: 8605881; PubMed Central PMCID: PMCPMC450009
  • Green DR, Scott DW. Activation-induced apoptosis in lymphocytes. Curr Opin Immunol. 1994 6;Jun(3):476–487. PubMed PMID: 7917117.
  • 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 Jun;5(6):651–655. PubMed PMID: 10371503.
  • Yc Ho, Shan L, Hosmane NN, et al. Replication-competent noninduced proviruses in the latent reservoir increase barrier to HIV-1 cure. Cell. 2013 Oct 24;155(3):540–551. PubMed PMID: 24243014; PubMed Central PMCID: PMCPMC3896327.
  • Bruner KM, Wang Z, Simonetti FR, et al. A quantitative approach for measuring the reservoir of latent HIV-1 proviruses. Nature. 2019 Feb;566(7742):120–125. PubMed PMID: 30700913; PubMed Central PMCID: PMCPMC6447073.
  • Davey RT Jr., Bhat N, Yoder C, et al. HIV-1 and T cell dynamics after interruption of highly active antiretroviral therapy (HAART) in patients with a history of sustained viral suppression. Proc Natl Acad Sci U S A. 1999 Dec 21;96(26):15109–15114. PubMed PMID: 10611346; PubMed Central PMCID: PMCPMC24781
  • 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 Dec 3;13(17):2405–2410. PubMed PMID: 10597782.
  • Julg B, Dee L, Ananworanich J, et al. Recommendations for analytical antiretroviral treatment interruptions in HIV research trials-report of a consensus meeting. Lancet HIV. 2019 Apr 6;(4):e259–e268. PubMed PMID: 30885693. DOI:10.1016/S2352-3018(19)30052-9.
  • Henderson GE, Peay HL, Kroon E, et al. Ethics of treatment interruption trials in HIV cure research: addressing the conundrum of risk/benefit assessment. J Med Ethics. 2018 Apr;44(4):270–276. PubMed PMID: 29127137; PubMed Central PMCID: PMCPMC5869463.
  • Angel JB, Jacobson MA, Skolnik PR, et al. A multicenter, randomized, double-blind, placebo-controlled trial of recombinant human interleukin-10 in HIV-infected subjects. Aids. 2000 Nov 10;14(16):2503–2508. PubMed PMID: 11101061
  • Lafeuillade A, Poggi C, Chadapaud S, et al. Pilot study of a combination of highly active antiretroviral therapy and cytokines to induce HIV-1 remission. J Acquir Immune Defic Syndr. 2001 Jan 1;26(1):44–55. PubMed PMID: 11176268
  • Vandergeeten C, Fromentin R, DaFonseca S, et al. Interleukin-7 promotes HIV persistence during antiretroviral therapy. Blood. 2013 May 23;121(21):4321–4329. PubMed PMID: 23589672; PubMed Central PMCID: PMCPMC3663425.
  • Katlama C, Lambert-Niclot S, Assoumou L, et al. Treatment intensification followed by interleukin-7 reactivates HIV without reducing total HIV DNA: a randomized trial. Aids. 2016 Jan;30(2):221–230. PubMed PMID: 26684819.
  • Garcia F, Plana M, Ortiz GM, et al. The virological and immunological consequences of structured treatment interruptions in chronic HIV-1 infection. Aids. 2001 Jun 15;15(9):F29–40. PubMed PMID: 11416735
  • Achenbach CJ, Assoumou L, Deeks SG, et al. Effect of therapeutic intensification followed by HIV DNA prime and rAd5 boost vaccination on HIV-specific immunity and HIV reservoir (EraMune 02): a multicentre randomised clinical trial. Lancet HIV. 2015 Mar;2(3):e82–91. PubMed PMID: 26424549; PubMed Central PMCID: PMCPMC6322849.
  • Sneller MC, Justement JS, Gittens KR, et al. A randomized controlled safety/efficacy trial of therapeutic vaccination in HIV-infected individuals who initiated antiretroviral therapy early in infection. Sci Transl Med. 2017 Dec 6;9(419). PubMed PMID: 29212716. DOI:10.1126/scitranslmed.aan8848.
  • Borducchi EN, Cabral C, Stephenson KE, et al. Ad26/MVA therapeutic vaccination with TLR7 stimulation in SIV-infected rhesus monkeys. Nature. 2016 Dec 8;540(7632):284–287. PubMed PMID: 27841870; PubMed Central PMCID: PMCPMC5145754.
  • Hansen SG, Ford JC, Lewis MS, et al. Profound early control of highly pathogenic SIV by an effector memory T-cell vaccine. Nature. 2011 May 26;473(7348):523–527. PubMed PMID: 21562493; PubMed Central PMCID: PMCPMC3102768.
  • Huang SH, Ren Y, Thomas AS, et al. Latent HIV reservoirs exhibit inherent resistance to elimination by CD8+ T cells. J Clin Invest. 2018 Feb 1;128(2):876–889. PubMed PMID: 29355843; PubMed Central PMCID: PMCPMC5785246.
  • Kim Y, Anderson JL, Lewin SR. Getting the “Kill” into “Shock and Kill”: strategies to eliminate latent HIV. Cell Host Microbe. 2018 Jan 10;23(1):14–26. . PubMed PMID: 29324227; PubMed Central PMCID: PMCPMC5990418.
  • Archin NM, Liberty AL, Kashuba AD, et al. Administration of vorinostat disrupts HIV-1 latency in patients on antiretroviral therapy. Nature. 2012 Jul 25;487(7408):482–485. PubMed PMID: 22837004; PubMed Central PMCID: PMCPMC3704185.
  • Elliott JH, Wightman F, Solomon A, et al. Activation of HIV transcription with short-course vorinostat in HIV-infected patients on suppressive antiretroviral therapy. PLoS Pathog. 2014 Oct;10(10):e1004473. PubMed PMID: 25393648; PubMed Central PMCID: PMCPMC4231123.
  • 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 Mar 23;36(3):491–501. PubMed PMID: 22406268; PubMed Central PMCID: PMCPMC3501645.
  • Sengupta S, Rf S. Targeting the latent reservoir for HIV-1. Immunity. 2018 May 15;48(5):872–895. . PubMed PMID: 29768175; PubMed Central PMCID: PMCPMC6196732.
  • Florez-Alvarez L, Hernandez JC, Zapata W. NK Cells in HIV-1 infection: from basic science to vaccine strategies. Front Immunol. 2018;9:2290. . PubMed PMID: 30386329; PubMed Central PMCID: PMCPMC6199347
  • Wen YM, Mu L, Shi Y. Immunoregulatory functions of immune complexes in vaccine and therapy. EMBO Mol Med. 2016 Oct;8(10):1120–1133. . PubMed PMID: 27572622; PubMed Central PMCID: PMCPMC5048363
  • Letourneau S, Im EJ, Mashishi T, et al. Design and pre-clinical evaluation of a universal HIV-1 vaccine. PLoS One. 2007 Oct 3;2(10):e984. PubMed PMID: 17912361; PubMed Central PMCID: PMCPMC1991584.
  • Mothe B, Molto J, Manzardo C, et al. Viral control induced by HIVconv vaccines & romidepsin in early treated individuals. Abstract 119LB. Conference on Retroviruses and Opportunistic Infections; 13–16 Feb 2017; Seattle, Washington.
  • Martin GE, Gossez M, Williams JP, et al. Post-treatment control or treated controllers? Viral remission in treated and untreated primary HIV infection. Aids. 2017 Feb 20;31(4):477–484. PubMed PMID: 28060012; PubMed Central PMCID: PMCPMC5278888.
  • Leth S, Schleimann MH, Nissen SK, et al. Combined effect of Vacc-4x, recombinant human granulocyte macrophage colony-stimulating factor vaccination, and romidepsin on the HIV-1 reservoir (REDUC): a single-arm, phase 1B/2A trial. Lancet HIV. 2016 Oct;3(10):e463–72. PubMed PMID: 27658863.
  • Fidler S, Stohr W, Pace M et al. A randomised controlled trial comparing the impact of antiretroviral therapy (ART) with a ‘Kick-and-Kill’ approach to ART alone on HIV reservoirs in individuals with primary HIV infection (PHI); RIVER trial. Abstract TUAA0202LB; 2nd International AIDS Conference 23–27 July 2018, Amsterdam, the Netherlands.
  • Pollack RA, Jones RB, Pertea M, et al. Defective HIV-1 proviruses are expressed and can be recognized by cytotoxic T lymphocytes, which shape the proviral landscape. Cell Host Microbe. 2017 Apr 12;21(4):494–506 e4. PubMed PMID: 28407485; PubMed Central PMCID: PMCPMC5433942.
  • Sen DR, Kaminski J, Barnitz RA, et al. The epigenetic landscape of T cell exhaustion. Science. 2016 Dec 2;354(6316):1165–1169. PubMed PMID: 27789799; PubMed Central PMCID: PMCPMC5497589.
  • Connick E, Mattila T, Folkvord JM, et al. CTL fail to accumulate at sites of HIV-1 replication in lymphoid tissue. J Immunol. 2007 Jun 1;178(11):6975–6983. PubMed PMID: 17513747.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.