SciELO - Scientific Electronic Library Online

 
vol.31 issue4Perinatal screening and data processing with the SUMA(r) technology in CubaRational design of an antitumor peptide based on the 32-51 region of the Limulus anti-LPS factor protein obtained from a chemical library author indexsubject indexarticles search
Home Pagealphabetic serial listing  

My SciELO

Services on Demand

Journal

Article

Indicators

  • Have no cited articlesCited by SciELO

Related links

  • Have no similar articlesSimilars in SciELO

Share


Biotecnología Aplicada

On-line version ISSN 1027-2852

Biotecnol Apl vol.31 no.4 La Habana Oct.-Dec. 2014

 

REPORT

 

Remarks on the Conference HIV pathogenesis, virus versus host, Alberta, Canada, March 9-14, 2014

 

Observaciones sobre el congreso La patogénesis del VIH, virus contra hospedero, Alberta, Canadá, marzo 9-14, 2014

 

 

Anna Ramírez-Suárez, Taimí Paneque-Guerrero, Dionne Casillas-Casanova, Celia Fernández-Ortega

Departamento de Farmacéuticos, Dirección de Investigaciones Biomédicas, Centro de Ingeniería Genética y Biotecnología, CIGB. Ave. 31 entre 158 y 190, Cubanacán, Playa, CP 11600, La Habana, Cuba.

 

 


ABSTRACT

Combined anti-retroviral therapy has been really efficient in suppressing HIV replication, but it does not cure the infection. This is due to the permanency of integrated viral DNA in the infected cells. To achieve the HIV cure, the interaction between virus and its host needs to be extremely understood. Because of this, scientific community persists in dedicating efforts to profoundly comprehend the pathogenesis of this virus. The present work summaries important aspects discussed in the last meeting HIV-Pathogenesis, Virus vs. Host. It was held in Fairmont Banff Springs, Banff, Alberta, Canada, March 9-14. The conference debated the latest advances in the biology of HIV-1. Topics as virus entry into the cell, into the host, virus exit, virushost genetics and co-evolution, host-virus interactions and responses, reservoirs, latency, reactivation, HIV and central nervous system, animal models and HIV and the microbiome at the mucosa were updated and deeply discussed. In parallel HIV vaccines: Adaptive Immunity and Beyond meeting was celebrated. The event, part of the Keystone Symposia Global Health Series, was a singular opportunity to analyze the state of HIV research and the new challenges in the battle against HIV infection.

Keywords: HIV-1, pathogenesis, latency, reservoirs, cure, eradication.


RESUMEN

La terapia antirretroviral combinada ha sido muy eficiente en suprimir la replicación del VIH, pero no cura la infección. Esto se debe a la presencia de ADN viral integrado en las células infectadas. Para lograr la cura del VIH, debe entenderse profundamente la interacción entre el virus y su hospedero. Es por ello que la comunidad científica dedica esfuerzos a comprender en detalle la patogénesis de este virus. El presente trabajo resume aspectos relevantes discutidos en la Conferencia Patogénesis del VIH, Virus contra Hospedero, la cual se celebró en Fairmont Banff Springs, Banff, Alberta, Canadá, entre el 9 y el 14 de marzo del año 2014. En ella se discutieron los últimos avances en la biología del VIH como: la entrada del virus a la célula, entrada al hospedero, la salida del virus, genética virus-hospedero y co-evolución, interacciones virus-hospedero y respuestas, reservorios, latencia, reactivación, VIH y sistema nervioso central, modelos animales así como la microbioma en la mucosa. El evento se celebró de manera simultánea con la Conferencia Vacunas VIH: Más allá de la inmunidad adaptativa. Ambos escenarios constituyeron una oportunidad magnífica para analizar el estado de la investigación del VIH y los nuevos retos en la batalla contra la infección.

Palabras clave: VIH-1, patogénesis, latencia viral, reservorios virales, cura, erradicación.


 

 

INTRODUCTION

Combined antiretroviral therapy (c-ART) has significantly impacted in the reduction of morbidity and mortality, transforming the Acquired Immunodeficiency Syndrome (AIDS) from a deadly to a treatable chronic disease [1]. However, replication competent viruses persist in patients despite therapy, leading to viral rebound upon treatment interruption [2-5]. Moreover, lifelong therapy has adverse effects, develops virus resistance, does not completely restore the immune system of infected patients and constitutes a great economic challenge for less-developed countries with high incidence of the epidemic [6-9].

In the last years, evidences of a cure known as “functional cure” have been presented by the scientific community. Its scope, comprising the reduction of the reservoirs, bringing about a drug-free control of infection [10, 11] has been observed in a rare population of individuals known as Elite controllers who naturally control HIV-1 infection without c-ART. Later, a virus control was also observed in the VISCONTI cohort where 14 HIV patients following discontinuation of c-ART maintained long lasting control of viremia [12]. Hopes about a new case of HIV functional cure (the “Mississippi baby”) were arisen; signs of viral replication were not seen for two years in a child born from an HIV-infected mother not in therapy that received antiretroviral drugs within the first 30 h postpartum and was treated for the first 15 months before therapy interruption [13]. But a recent report informed on the viral rebound in the child [14].

Up to now, three different strategies have been considered to achieve the functional cure: 1) initiation of c-ART during the very early primary stage of acute HIV-1 infection [13]; 2) selective depletion of discrete CD4+ T cell subsets carrying integrated the HIV-1 DNA without viral reactivation [15]; and 3) the socalled “shock/kick and kill” strategy consisting in inducing, through drugs, transcription of quiescent, replication-competent HIV-1 provirus (the shock/ kick phase) in the presence of c-ART (to block viral spread), making virus reactivating cells susceptible to immune clearance, cytopathic effects and/or the effects of ad hoc therapeutics (the kill phase) [16].

This last strategy has rise considerable enthusiasm, with histone diacetilase inhibitors (HDACis) as the most advanced intervention as HIV-1 antilatency agents in the clinical testing. Results of a pilot single-dose trial in few aviremic patients on c-ART with the suberoylanilidehydroxamic acid, SAHA (vorinostat) have shown that this drug can disrupt latent infection in a detectable proportion of the reservoir of latent resting CD4+ T cells [17]. Nevertheless, the wide effectiveness of the treatment at clearing the viral reservoir is still unknown. Besides, SAHA is a mutagen in vitro in standard bacterial assays and, consequently, its use in humans has been accepted only for short-term exposure [18]. Novel HDACis are also being studied in order to reduce toxicity and increase specificity. These include givinostat, belinostat [19], panobinostat [20] oxamflatin [21], NCH-51 [22], romidepsin [23] and droxinostat [24]. Histone methyltransferase inhibitors, chaetocin inhibitors [25, 26], inducers of protein kinase C and NF-κB/NF-AT pathways and P-TEFb stimulators have also been considered [27]. Disulfiram, a zinc-chelating agent approved by the Food and Drug Administration to treat alcoholism, has been recently reported to turn on HIV-1 transcription without global T cell activation or cytokine release in a primary cell model of latent HIV infection [28]. Nevertheless, initial results of an ongoing single-arm pilot clinical trial (NCT01286259) did not provide clear evidence of any positive effect, showing a rapid and modest increase in plasma HIV-1 RNA in all participants, but a very modest reduction in the size of the latent reservoir [29]. Two classes of quinoline derivatives from quinoline-8-ol, a bivalent cationchelator, adducts of 5-ch5-chloroquinolin-8-ol, and quinolin-8-yl carbamates, induce HIV-1 expression without cell activation [30].

Several immunologic strategies have been proposed in combination with reactivating compounds, to enhance the clearance of reactivated latently-infected cells and to improve host immune responses. These strategies include therapeutic vaccines, neutralizing monoclonal antibodies and immune modulating drugs.

The fact that potent CD8+ T lymphocytes can control HIV-1 by selectively killing virus-producing cells in Elite controllers [31] has led to a renewed interest in therapeutic vaccines, but this time with the goal of increasing virus-specific immune responses, either accelerating the decay of the reservoir during c-ART or improving the control of viral rebound after interruption of c-ART. Such interventions include CMV vectors; Ad26 prime, modified vaccinia Ankara (MVA) boost regimens; and lymph node-targeted amphiphilic peptide vaccines [27, 32].

A new generation of monoclonal antibodies (mAbs) with improved potency and breadth has also encouraged new therapeutic approaches. Particularly, two studies in rhesus monkeys demonstrated that infusión of mAbs cocktails or individual mAbs in chronically SHIV-infected rhesus monkeys resulted in substantial, although transient, suppression of viremia [33, 34], based on previous studies in humanized mice [35].

The blockade of immune checkpoint molecules PD-1, CTLA-4, LAG-3, TIM-3, TIGIT, and 2B4 may also prove effectiveness, with clinical trials of PD-1 inhibitors in treated HIV-1 disease been initiated [32].

Another approach, the use of cytokines, is being analyzed to stimulate HIV-1 replication and to interfere with mechanisms responsible for HIV-1 latency in individuals on c-ART [36-38]. Currently, the ERAIMMUNE 01 clinical trial is ongoing to test IL-7 in combination with an anti-HIV-1 vaccine and raltegravir plus maraviroc, to inhibit proliferation or reseeding of the HIV latent reservoir [27].

The ultime goal, the sterilizing cure, has been also considered. It is aimed to eradicate the latent virus reservoirs. This intervention has been only successfully achieved in the “Berlin patient”, a man receving an allogenic hematopoietic stem cells transplant (HSCT) from a homozygous CCR5Δ32 donor for acute myelogeneous leukemia and who appeared to be free of both cancer and HIV-1 for over 5 years, without c-ART [39-41]. A similar case comprised two HIV-1 patients in 2010, known as the “Boston patients” who received HSCT for treatment of refractory lymphoma and being also covered with c-ART for two or more years after intervention [42], but unfortunately, viremia rebound within two weeks post-interruption. Phylogenetic analysis demonstrat-ed that viral rebound was caused by the presence of one or few latent proviruses, being consistent with the persistence of a minimal reservoir of infected cells despite HSCT [43].

Bone marrow transplant was also considered, but its associated risks have limited its applicability in HIV patients. Related attempts comprise transforming the patient’s cells by generating a CCR5 deletion cells resistant to infection or the direct deletion of the virus from infected cells, followed by an autologous transplant. In fact, HIV co-receptors and pro-viral sequences can be targeted through the use of DNA-editing enzymes [44], such as: zinc-finger nucleases (ZNFs) [45], transcription activator-like effector nucleases and homing endonucleases [44, 46, 47].

One completed [48] and two ongoing (NCT01044654, NCT01252641) [49, 50] clinical trial explore the use of ZFNs to manipulate CCR5 expression in CD4+ T cells and precursor cells [45, 51]. They bring evidences on the safety and efficacy of CCR5-ZFN-treated autologous cells in patients with chronic aviremic HIV infection under c-ART. Promising results were obtained, with increased CD4+ T cell counts, reduced viral load, improved ability of cells to localize to anatomical reservoirs, and decreased reservoir size. Moreover, engineered cells were detected to persist in the body, their populations declining at slower rates than non-engineered cells after c-ART interruption [27]. Nevertheless, these cells do not prevent the infection by CXCR4-tropic viruses and may drive selection for either X4-specific or dual-tropic HIV-1 viruses [52, 53]. Recent preclinical studies in cell lines and humanized mice with ZFNs targeting CXCR4 in CCR5Δ32 CD4+ T cells indicate protection from both R5- and X4-tropic HIV viruses [54, 55]. Otherwise, the physiological impact of lacking both receptors needs to be characterized. Although all these interventions seem in general to be safe, their efficacy in humans has yet to be proven [27].

Many of these issues, together with other relevant aspects on the wide spectrum of the virus-host interaction were discussed in the HIV Pathogenesis, virus versus host meeting, as part of the Keystone Symposia Global Health Series, held at Fairmont Banff Springs, in Banff, Alberta, Canada, on March 9 to 14, 2014.

 

THE HIV PATHOGENESIS, VIRUS VERSUS HOST MEETING

The meeting focused on the latest advances in basic and translational HIV research with emphasis on mechanistic models. Key aspects of HIV biology from infection to AIDS were presented. The program included: 1) Virus entry/exit into/from the cell; 2) Entry into the host; 3) Virus-host genetics and co-evolution; 4) Virus-host interactions and responses; 5) Pathogenesis; 6) Reservoirs, latency and reactivation; and 7) HIV and the microbiome at the mucosa. Scientific environment was enriched with the simultaneous meeting on “HIV Vaccines: Adaptive Immunity and Beyond”, which shared opening and closing keynote addresses and two plenary sessions with this meeting.

The organizers presented a Pre-meeting Workshop for the Global Health Travel Award winners. Prominent lectures on hot topics were delivered by leaders in the HIV field: Dr. Stephen DeRosa from the University of Washington, USA, talking on the HIV im-munity; Dr. Penny Moore from the National Institute for Communicable Diseases, South Africa, humoral HIV immunity; Dr. Michael Emerman from the Fred Hutchinson Cancer Research Center, USA, on HIV restriction factors; and Dr. David Margolis from the University of North Carolina, Chapel Hill, USA, on the HIV cure perspectives.

During the congress, Dr. Victor Garcia Martinez, Professor of Medicine at the Center for AIDS Research, USA, dictated an outstanding conference about the contribution of humanized bone marrow/ liver/thymus (BLT) mice to the elucidation of the molecular basis of vaginal HIV transmission and prevention. BLT mice develop a functional human immune system and offer a rapid, reliable and reproducible experimental system for HIV research. Very relevant to HIV transmission is the extensive reconstitution of the female reproductive tract of BLT mice with human hematopoietic cells. Dr. Garcia Martinez and his group have evaluated the vaginal transmission of highly relevant transmitted/founder viruses, the role of semen on HIV transmission, the effect of semen on the efficacy of topical microbicides and the role of cell-associated virus on vaginal transmission. The results have been crucial in providing novel insight into mucosal HIV transmission and the effectiveness of different prevention strategies.

An excellent lecture on the host and HIV exit was delivered by Dr. Fadila Bouamr from the National Institute of Health, USA. Her group investigated how viruses interact with hosts to promote their replication. The nucleocapsid (NC) domain of the HIV-1 structural protein Gag was found to accomplish a surprinsingly complex set of functions during the HIV replication cycle. In addition to the role of this protein in genome integration, Gag-Gag multimerization, and reverse transcription, Dr. Fadila and colleagues reported the first evidence of NC involvement in HIV-1 budding and exit. NC mutant virions remained tethered to the surface of CD4+ T cells, indicating the failure to recruit or utilize members of the host machinery that catalizes membrane scission and virus exit. Alix Bro 1, a member of the endosomal sorting complexes re-quired for transport (ESCRT) binds NC, establishing a direct role for NC in mediating interactions with ESCRT necessary for virus release, with the involvement of RNA for molecule recruitment.

Dr. Janice Clements from the Johns Hopkins University School of Medicine, USA, spoke about the macrophages contribution to the latent viral reservoir in tissues of c-ART-suppressed SIV-infected macaques. HIV latency has been well established in resting CD4+ T cells and their longevity provides a lifelong reservoir in HIV infected individuals on c-ART. It is known that HIV and SIV infect CD4+ T cells, monocytes, macrophages and microglia. Nevertheless, viral latency has not been well studied in tissues of HIV and SIV infected individuals and viral assays are needed for quantitating infected and latent monocytes and macrophages. Clements and colleagues used an SIV macaque model suppressed by c-ART to less than 10 copies per milliliter of plasma. Monocytes and macrophages from SIV-infected macaques, either suppressed or not, were quantified. The study demonstrated that monocytes and macrophages support SIV infection and the suppression of SIV by c-ART leads to low level virus replication in tissues. SIV was reactivated from monocytes and tissue macrophages using the Quantitative Viral Outgrowth Assay. This demonstrates that macrophages lineage cells can provide a latent reservoir for SIV.

Results about APOBEC3 genes family (A3), an interferon-stimulated genes family, were showed by Dr. Richard T. D’Aquila from Northwestern University Feinberg School of Medicine, USA. He and his group posit that early in the infection, high A3 levels/ activities are one of the innate defenses lost during acute infection in non-controllers and preserved by the anti-HIV CTL activity in controllers. They tried to unravel how to favor A3 control of Vif-positive HIV-1 virus by boosting cellular A3s. Dr. D’Aquila presented lead compounds unrelated to interferons that induce some A3s, and block early steps of Vif-positive HIV replication in vitro. They also considered that knowing the A3F virion core localization mechanisms may contribute to enhance anti-HIV activity of other virion-packaged A3s.

Dr. Andrea Cimarelli from the International Center for Infectology Research, USA, explored the complexity of infection of myeloid cells by primate lentiviruses during the early phases of infection. Myeloid cells support viral replication at low levels due to a major restriction in reverse transcription. An important factor of this restriction is SAMHD1, a triphosphohydrolase that limits dNTPs. Dr. Cimarelli suggested that the slow accumulation of viral DNA (vDNA) as orchestrated by SAMHD1 leads to the exposure of vDNA to the protracted action of cytoplasmic effectors. Evidences indicate that one such factor is the cytidine deaminase APOBEC3A which restricts lentiviral in-fection by decreasing vDNA accumulation in myeloid cells. On the other hand, based on experiments with Vpx (a protein that degrades SAMHD1) and HIV-2/ SIVSM and HIV-1 viruses, it seems like the low levels of replication in myeloid cells may be a common goal for primate lentiviruses that has been resolved through different evolutionary strategies. This and other results suggest that another restriction may differentially target primate lentiviruses in myeloid cells.

Regarding the natural resistance to HIV infection, Dr. Reuben S. Harris from the University of Minnesota, USA, spoke on the role of select APOBEC3H haplotypes. They demonstrated that naturally occurring variants of APOBEC3H are stably expressed and are able to encapsidate and restrict HIV replication in primary CD4+ T cells. Weak Vif alleles comprise approximately 25 % of HIV subtype B isolates, suggesting a dynamic relationship between the evolution of Vif to antagonize APOBEC3H in populations in which some individuals express an active APOBEC3H.

HIV eradication approaches were also adressed by Dr. David M. Margolis. Most eradication strategies elicit proviral expression with the assumption that c-ART will prevent new infections while the immune system, possibly with the help of other therapeutics, will clear infected cells. There are other strategies on the use of HIV-specific recombinases to destroy proviral DNA, or HIV-dependent suicide genes to selectively kill HIV-infected cells. Several strategies are being evaluated as transcriptional activators, such as: HDACis, Disulfiram, PD-1 inhibition and vaccination. Dr. Margolis talked about the complex and multimodal nature of HIV eradication approaches, with emphasis on the results of clinical trials with vorinostat, a HDACis currently approved for oncologic clinical use which activates HIV transcription in cell-line models of latent HIV and in ex vivo cultured cells. Vorinostat, administered as a 400-mg single dose, induces the expression of the full-length HIV RNA in latently-infected resting CD4+ T cells. After 14 doses, ninety percent of participants had a significant increase in HIV production from the former latently-infected cells. These results prove that this drug can bring HIV out of hiding in resting T cells.

A very interesting lecture about prospects of HIV eradication was delivered by Dr. Robert Siliciano from the Johns Hopkins University School of Medicine of Baltimore, USA. The availability of assays that accurately measure the size of the latent reservoir in resting CD4+ T cells is crucial from the therapeutic perspective, with latency reversing agents (LRAs) as key triggers of reservoir activation. Dr. Siliciano discussed a recent work indicating that PCR-based assays overestimate the size of the reservoir, while the Quantitative Viral Outgrowth Assay may underestimate the reservoir by as much as 60-fold due to the failure to induce replication competent proviruses by a single round of activation. Recent studies with patients’ cells in the presence of LRAs were also analyzed [56]. Remarkably, the most currently used LRAs do not reverse latency in patients’ cells, but they are effective in in vitro models of latency. This finding suggests that LRAs proven to be effective in patients’ cells must be of critical priority.

Dr. Nicholas Chomont, from the Vaccine and Gene Therapy Institute of Florida, USA, talked about what could be learned about HIV persistence during infection from CD4+ T cell homeostasis. Chomont and colleagues identified a small group of specific checkpoint blockers (including PD1) that are associated with immune dysfunction, the size of the viral reservoir, and which may induce latency in CD4+ T cells. On the other hand, it has been observed that the gamma-c cytokine IL-7 plays a crucial role in the maintenance of the viral reservoir by promoting the homeostatic proliferation of latently-infected cells. All these indicate that HIV persistence during c-ART is ensured by immunological mechanisms that are responsible for the homeostasis of the memory CD4+ T cell pool. Dr. Chomont proposed that interfering with the regulators of T cell homeostasis may disrupt HIV latency in virally-suppressed subjects under suppressive c-ART. It would also pave the way for the development of novel strategies to cure the HIV infection.

In another talk, novel means for eliminating latent HIV reservoirs were presented by Dr. Jerome A. Zack from the University of California, Los Angeles, USA. His group designed and synthesized new latency activating compounds that function via the protein kinase C (PKC) signalling pathway. They we able to produce a naturally occurring nanoparticle called “vault”, by which can latency activating compounds can be more selectively introduced into the target cell type. The novel molecules were evaluated in cell line models for HIV latency, together with several primary cell assays and latently infected cells from c-ART treated patients. All the tested compounds potently activated HIV from latency, in many cases more strongly than the natural molecules. Particularly, vault nanoparticles carrying bryostatin 1 were able to activate HIV from latency and showed to be active in CD4+ splenocytes following its intravenous administration in immunocompetent mice. Dr. Zack considered that novel PKC activators and nanoparticle delivery methods may impact in activation-elimination strategies to eradicate latent HIV reservoirs.

 

CONCLUDING REMARKS

HIV eradication is still an outstanding challenge for the scientific community [27]. ART has achieved to control viral replication and significantly decreased the AIDS-related morbidity and mortality, but without curing the infection [57]. An integral understanding of the virus-host interaction remains as a major prerequisite for the rational improvement of therapeutic strategies [58]. Two major strategies are envisaged to eradicate HIV infection: a functional cure and a sterilizing cure, both aimed to diminish and eliminate the viral reservoirs, respectively [10, 11]. The finding of reliable, fast and economic methods that can reproducibly evaluate the real effectiveness of treatments under investigation in different laboratories, still requires especial and sustained scientific efforts [27].

 

ACKNOWLEDGEMENTS

The authors thanks to the Global Health Travel Award for making possible the participation at the HIV Pathogenesis, Virus vs. Host Conference. They also acknowledge to the Scholarship coordinator from Keystone Symposia, Jeff Lehman, by his favorable and professional support.

 

REFERENCES

1. Katlama C, Deeks SG, Autran B, Martinez-Picado J, van Lunzen J, Rouzioux C, et al. Barriers to a cure for HIV: new ways to target and eradicate HIV-1 reservoirs. Lancet. 2013;381(9883):2109-17.

2. Finzi D, Blankson J, Siliciano JD, Margolick JB, Chadwick K, Pierson T, 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(5):512-7.

3. Siliciano JD, Kajdas J, Finzi D, Quinn TC, Chadwick K, Margolick JB, et al. Long-term follow-up studies confirm the stability of the latent reservoir for HIV-1 in resting CD4+ T cells. Nat Med. 2003;9(6):727-8.

4. Finzi D, Hermankova M, Pierson T, Carruth LM, Buck C, Chaisson RE, et al. Identification of a reservoir for HIV-1 in patients on highly active antiretroviral therapy. Science. 1997;278(5341):1295-300.

5. Blankson J, Persaud D, Siliciano RF. Latent reservoirs for HIV-1. Curr Opin Infect Dis. 1999;12(1):5-11.

6. Maartens G, Celum C, Lewin SR. HIV infection: epidemiology, pathogenesis, treatment, and prevention. Lancet. 2014;384(9939):258-71.

7. Este JA, Cihlar T. Current status and challenges of antiretroviral research and therapy. Antiviral Res. 2010;85(1):25-33.

8. Hecht R, Stover J, Bollinger L, Muhib F, Case K, de Ferranti D. Financing of HIV/AIDS programme scale-up in low-income and middle-income countries, 2009-31. Lancet. 2010;376(9748):1254-60.

9. Hill AM, Cho M, Mrus JM. The costs of full suppression of plasma HIV RNA in highly antiretroviral-experienced patients. AIDS Rev. 2011;13(1):41-8.

10. International, Aids Society Scientific Working Group on H. I. V. Cure, Deeks SG, Autran B, Berkhout B, Benkirane M, Cairns S, et al. Towards an HIV cure: a global scientific strategy. Nat Rev Immunol. 2012;12(8):607-14.

11. Trono D, Van Lint C, Rouzioux C, Verdin E, Barre-Sinoussi F, Chun TW, et al. HIV persistence and the prospect of long-term drug-free remissions for HIV-infected individuals. Science. 2010;329(5988):174-80.

12. Saez-Cirion A, Bacchus C, Hocqueloux L, Avettand-Fenoel V, Girault I, Lecuroux C, et al. Post-treatment HIV-1 controllers with a long-term virological remission after the interruption of early initiated antiretroviral therapy ANRS VISCONTI Study. PLoS Pathog. 2013;9(3):e1003211.

13. Persaud D, Gay H, Ziemniak C, Chen YH, Piatak M, Jr., Chun TW, et al. Absence of detectable HIV-1 viremia after treatment cessation in an infant. N Engl J Med. 2013;369(19):1828-35.

14. National Institutes of Health (NIH). “Mississippi baby” now has detectable HIV, researchers find. 2014 [cited 2014 Dec 17]. Available from: http://www.niaid.nih.gov/news/newsreleases/2014/pages/mississippibabyhiv.aspx.

15. Lewis MG, DaFonseca S, Chomont N, Palamara AT, Tardugno M, Mai A, et al. Gold drug auranofin restricts the viral reservoir in the monkey AIDS model and induces containment of viral load following ART suspension. Aids. 2011;25(11): 1347-56.

16. Hamer DH. Can HIV be Cured? Mechanisms of HIV persistence and strategies to combat it. Curr HIV Res. 2004;2(2):99-111.

17. Archin NM, Liberty AL, Kashuba AD, Choudhary SK, Kuruc JD, Crooks AM, et al. Administration of vorinostat disrupts HIV-1 latency in patients on antiretroviral therapy. Nature. 2012;487(7408):482-5.

18. Margolis DM. Histone deacetylase inhibitors and HIV latency. Curr Opin HIV AIDS. 2011;6(1):25-9.

19. Matalon S, Palmer BE, Nold MF, Furlan A, Kassu A, Fossati G, et al. The histone deacetylase inhibitor ITF2357 decreases surface CXCR4 and CCR5 expression on CD4(+) T-cells and monocytes and is superior to valproic acid for latent HIV-1 expression in vitro. J Acquir Immune Defic Syndr. 2010;54(1):1-9.

20. Rasmussen TA, Schmeltz Sogaard O, Brinkmann C, Wightman F, Lewin SR, Melchjorsen J, et al. Comparison of HDAC inhibitors in clinical development: effect on HIV production in latently infected cells and T-cell activation. Hum Vaccin Immunother. 2013;9(5):993-1001.

21. Yin H, Zhang Y, Zhou X, Zhu H. Histonedeacetylase inhibitor Oxamflatin increase HIV-1 transcription by inducing histone modification in latently infected cells. Mol Biol Rep. 2011;38(8):5071-8.

22. Victoriano AF, Imai K, Togami H, Ueno T, Asamitsu K, Suzuki T, et al. Novel histone deacetylase inhibitor NCH-51 activates latent HIV-1 gene expression. FEBS Lett. 2011;585(7):1103-11.

23. Furumai R, Matsuyama A, Kobashi N, Lee KH, Nishiyama M, Nakajima H, et al. FK228 (depsipeptide) as a natural prodrug that inhibits class I histone deacetylases. Cancer Res. 2002;62(17):4916-21.

24. Huber K, Doyon G, Plaks J, Fyne E, Mellors JW, Sluis-Cremer N. Inhibitors of histone deacetylases: correlation between isoform specificity and reactivation of HIV type 1 (HIV-1) from latently infected cells. J Biol Chem. 2011;286(25):22211-8.

25. Imai K, Togami H, Okamoto T. Involvement of histone H3 lysine 9 (H3K9) methyltransferase G9a in the maintenance of HIV-1 latency and its reactivation by BIX01294. J Biol Chem. 2010;285(22):16538-45.

26. Bernhard W, Barreto K, Saunders A, Dahabieh MS, Johnson P, Sadowski I. The Suv39H1 methyltransferase inhibitor chaetocin causes induction of integrated HIV-1 without producing a T cell response. FEBS Lett. 2011;585(22):3549-54.

27. Battistini A, Sgarbanti M. HIV-1 latency: an update of molecular mechanisms and therapeutic strategies. Viruses. 2014;6(4):1715-58.

28. Xing S, Bullen CK, Shroff NS, Shan L, Yang HC, Manucci JL, 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(12):6060-4.

29. Spivak AA, Hoh R, Bacchetti P, Eisele E, Buckheit R, Lai J, et al. Safety and feasibility of using disulfiram to enhance HIV transcription among long-term ARV-treated adults: Preliminary results from a pilot study. Proceedings of the 19th Conference on Retroviruses and Opportunistic Infections; 2012 March 5-8; Seattle, USA. 2012.

30. Xing S, Bhat S, Shroff NS, Zhang H, Lopez JA, Margolick JB, et al. Novel structurally related compounds reactivate latent HIV-1 in a bcl-2-transduced primary CD4+ T cell model without inducing global T cell activation. J Antimicrob Chemother. 2012;67(2):398-403.

31. Migueles SA, Osborne CM, Royce C, Compton AA, Joshi RP, Weeks KA, et al. Lytic granule loading of CD8+ T cells is required for HIV-infected cell elimination associated with immune control. Immunity. 2008;29(6):1009-21.

32. Barouch DH, Deeks SG. Immunologic strategies for HIV-1 remission and eradication. Science. 2014;345(6193):169-74.

33. Barouch DH, Whitney JB, Moldt B, Klein F, Oliveira TY, Liu J, et al. Therapeutic efficacy of potent neutralizing HIV-1-specific monoclonal antibodies in SHIV-infected rhesus monkeys. Nature. 2013;503(7475):224-8.

34. Shingai M, Nishimura Y, Klein F, Mouquet H, Donau OK, Plishka R, et al. Antibody-mediated immunotherapy of macaques chronically infected with SHIV suppresses viraemia. Nature. 2013;503(7475):277-80.

35. Klein F, Halper-Stromberg A, Horwitz JA, Gruell H, Scheid JF, Bournazos S, et al. HIV therapy by a combination of broadly neutralizing antibodies in humanized mice. Nature. 2012;492(7427):118-22.

36. Chun TW, Engel D, Mizell SB, Ehler LA, Fauci AS. Induction of HIV-1 replication in latently infected CD4+ T cells using a combination of cytokines. J Exp Med. 1998;188(1):83-91.

37. Chun TW, Engel D, Mizell SB, Hallahan CW, Fischette M, Park S, 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(6):651-5.

38. Vandergeeten C, Fromentin R, Chomont N. The role of cytokines in the establishment, persistence and eradication of the HIV reservoir. Cytokine Growth Factor Rev. 2012;23(4-5):143-9.

39. Hutter G, Nowak D, Mossner M, Ganepola S, Mussig A, Allers K, et al. Long-term control of HIV by CCR5 Delta32/ Delta32 stem-cell transplantation. N Engl J Med. 2009;360(7):692-8.

40. Allers K, Hutter G, Hofmann J, Loddenkemper C, Rieger K, Thiel E, et al. Evidence for the cure of HIV infection by CCR5Delta32/Delta32 stem cell transplantation. Blood. 2011;117(10):2791-9.

41. Yukl SA, Boritz E, Busch M, Bentsen C, Chun TW, Douek D, et al. Challenges in detecting HIV persistence during potentially curative interventions: a study of the Berlin patient. PLoS Pathog. 2013;9(5):e1003347.

42. Henrich TJ, Hu Z, Li JZ, Sciaranghella G, Busch MP, Keating SM, et al. Long-term reduction in peripheral blood HIV type 1 reservoirs following reduced-intensity conditioning allogeneic stem cell transplantation. J Infect Dis. 2013;207(11):1694-702.

43. Henrich TJ, Hanhauser E, Marty FM, Sirignano MN, Keating S, Lee TH, et al. Antiretroviral-free HIV-1 remission and viral rebound after allogeneic stem cell transplantation: report of 2 cases. Ann Intern Med. 2014;161(5):319-27.

44. Buchholz F, Hauber J. Engineered DNA modifying enzymes: components of a future strategy to cure HIV/AIDS. Antiviral Res. 2013;97(2):211-7.

45. Perez EE, Wang J, Miller JC, Jouvenot Y, Kim KA, Liu O, et al. Establishment of HIV- 1 resistance in CD4+ T cells by genome editing using zinc-finger nucleases. Nat Biotechnol. 2008;26(7):808-16.

46. Mussolino C, Morbitzer R, Lutge F, Dannemann N, Lahaye T, Cathomen T. A novel TALE nuclease scaffold enables high genome editing activity in combination with low toxicity. Nucleic Acids Res. 2011;39(21):9283-93.

47. Schiffer JT, Aubert M, Weber ND, Mintzer E, Stone D, Jerome KR. Targeted DNA mutagenesis for the cure of chronic viral infections. J Virol. 2012;86(17):8920-36.

48. Tebas P, Stein D, Tang WW, Frank I, Wang SQ, Lee G, et al. Gene editing of CCR5 in autologous CD4 T cells of persons infected with HIV. N Engl J Med. 2014;370(10):901-10.

49. Lalezari JMR, Wang S, Lee G, Giedlin M, Tang W, Spratt K, et al. A single infusion of zinc finger nuclease CCR5 modified autologous CD4 T cells (SB-728-T) increases CD4 counts and leads to decrease in HIV proviral load in an aviremic HIV-infected subject. Proceedings of the 19th Conference on Retroviruses and Opportunistic Infections; 2012 March 5-8; Seattle, USA. 2012.

50. June CTP, Stein D, Mitsuyasu R, Lalezari J, Wang S, Lee G, et al. Introduction of acquired CCR5 deficiency with zinc finger nuclease-modified autologous CD4 T cells (SB-728-T) correlates with increases in CD4 count and effects on viral load in HIV-infected subjects. Proceedings of the 19th Conference on Retroviruses and Opportunistic Infections; 2012 March 5-8; Seattle, USA. 2012.

51. Holt N, Wang J, Kim K, Friedman G, Wang X, Taupin V, et al. Human hematopoietic stem/progenitor cells modified by zinc-finger nucleases targeted to CCR5 control HIV-1 in vivo. Nat Biotechnol. 2010;28(8):839-47.

52. Westby M, Lewis M, Whitcomb J, Youle M, Pozniak AL, James IT, et al. Emergence of CXCR4-using human immunodeficiency virus type 1 (HIV-1) variants in a minority of HIV-1-infected patients following treatment with the CCR5 antagonist maraviroc is from a pretreatment CXCR4-using virus reservoir. J Virol. 2006;80(10):4909-20.

53. Nedellec R, Coetzer M, Lederman MM, Offord RE, Hartley O, Mosier DE. Resistance to the CCR5 inhibitor 5P12-RANTES requires a difficult evolution from CCR5 to CXCR4 coreceptor use. PLoS One. 2011;6(7):e22020.

54. Wilen CB, Wang J, Tilton JC, Miller JC, Kim KA, Rebar EJ, et al. Engineering HIV-resistant human CD4+ T cells with CXCR4-specific zinc-finger nucleases. PLoS Pathog. 2011; 7(4):e1002020.

55. Didigu CA, Wilen CB, Wang J, Duong J, Secreto AJ, Danet-Desnoyers GA, et al. Simultaneous zinc-finger nuclease editing of the HIV coreceptors ccr5 and cxcr4 protects CD4+ T cells from HIV-1 infection. Blood. 2014;123(1):61-9.

56. Bruner KM, Hosmane NN, Siliciano RF. Towards an HIV-1 cure: measuring the latent reservoir. Trends Microbiol. 2015;23(4):192-203.

57. De Crignis E, Mahmoudi T. HIV eradication: combinatorial approaches to activate latent viruses. Viruses. 2014;6(11):4581-608.

58. Rubbert A, Behrens G, Ostrowski M. Pathogenesis of HIV-1 Infection. In: Hoffmann C, Rockstroh JK, editors. HIV 2011. 2011. Hamburg: Medizin Fokus. Available from: http://www.hivbook.com.

 

 

Received in October, 2014.
Accepted in December, 2014.

 

 

Anna Ramírez-Suárez. Departamento de Farmacéuticos, Dirección de Investigaciones Biomédicas, Centro de Ingeniería Genética y Biotecnología, CIGB. Ave. 31 entre 158 y 190, Cubanacán, Playa, CP 11600, La Habana, Cuba. E-mail: anna.ramirez@cigb.edu.cu.

Creative Commons License All the contents of this journal, except where otherwise noted, is licensed under a Creative Commons Attribution License