Abstract
Purpose of review
Highly active antiretroviral treatment has dramatically improved prognosis for people living with HIV by preventing AIDS-related morbidity and mortality through profound suppression of viral replication. However, a long-lived viral reservoir persists in latently infected cells that harbor replication-competent HIV genomes. If therapy is discontinued, latently infected memory cells inevitably reactivate and produce infectious virus, resulting in viral rebound. The reservoir is the biggest obstacle to a cure of HIV.
Recent findings
This review summarizes significant advances of the past year in the development of cellular and gene therapies for HIV cure. In particular, we highlight work done on suppression or disruption of HIV co-receptors, vectored delivery of antibodies and antibody-like molecules, T cell therapies, and HIV genome disruption.
Summary
A number of recent advancements in cellular and gene therapies have emerged at the forefront of HIV cure research, potentially having broad implications for the future of HIV treatment.
Keywords: HIV cure, gene therapy, CCR5, vector-delivered antibody, CAR T cell
Introduction
Developing an HIV cure remains a major challenge due to a long-lived viral reservoir, which persists despite effective antiretroviral therapy (ART). This reservoir is composed of infected cells harboring transcriptionally silent but replication competent provirus. Resting CD4+ T cells with memory phenotype, including central, effector, transitional, and stem memory T cells, are the best-characterized cellular reservoirs of HIV-1 infection, though other cell types may contribute [1,2]. Latently infected cells can reactivate and seed viral rebound if ART is discontinued. The reservoir decays with a half-life of 44 months, implying an estimated 73 years of treatment would be required for complete eradication under stable ART [3,4]. As a result, HIV-infected individuals face life-long antiretroviral therapy, and efforts toward an HIV cure must address the viral reservoir.
While a sterilizing cure would achieve complete elimination of virus from the body, perhaps a more readily attainable outcome is a functional cure, in which drug-free control of HIV is achieved through significant reservoir reduction or inactivation. Mathematical modeling suggests that greater than a four log reduction of the reservoir could prevent viral rebound following ART disruption [5]. Similarly, a three-log reduction could delay viral rebound for nearly 1 year. Early treatment initiation trials in both adults and infants provide strong evidence that reducing the size of the latent reservoir can delay virus rebound for months or even years after treatment cessation [6-8].
The single clearly-documented instance of HIV cure to date occurred via a cell and gene therapy strategy. Timothy Ray Brown, the “Berlin patient,” received an allogeneic stem-cell transplant for acute myelogenous leukemia from a donor homozygous for a naturally occurring 32 bp deletion in the gene encoding CCR5 CCR5Δ32, [9,10], an immune cell receptor critical for cellular entry by most types of HIV. The 32bp deletion in CCR5 confers cellular resistance to HIV infection. Ultrasensitive assays showed that the “Berlin patient” achieved at least a 7500-fold, or nearly four log, reservoir reduction, and he has remained without detectable virus for nine years after transplant. This case has inspired a field of ever growing HIV cure research, and here we highlight approaches that rely on cell and gene therapy for HIV cure (Figure).
Figure 1. Cell and gene therapy strategies for HIV cure.
A) Latently infected cells can reactivate and produce virus that reseeds the reservoir. B) Cell and gene therapy strategies that block viral production, kill infected cells, or protect cells from infection.
Suppression or disruption of HIV co-receptors
The cure of Timothy Ray Brown has generally been assumed to relate to the HIV-resistant phenotype of the cells he received [11] [12]. Subsequent studies have sought to repeat the cure, such as an HIV-positive patient with lymphoma who received transplantation from CCR5Δ32-homozygous cord blood cells [13]. However, widespread implementation of transplantation is unfeasible given the low prevalence of the resistant phenotype, HLA-matching requirements, and the inherent risk and expense of transplantation. Thus, many HIV cure efforts aim to mimic the resistant phenotype by using gene suppression or gene disruption to eliminate the CCR5 receptor in a patient’s own cells. Ideally, the resulting population of resistant immune cells would expand in the presence of HIV, as resistant cells avoid virus-induced cytotoxicity, enabling them to repopulate the immune system and improve immune function.
Strategies to eliminate CCR5 include RNA interference to knock down expression of CCR5 [14] and gene disruption to lethally mutate the gene encoding CCR5 [15,16]. Endonucleases such as Zinc finger nucleases (ZFNs), Tal-effector nucleases (TALENS), meganucleases, and CRISPR/Cas9 can be targeted to cleave specific DNA sequences, resulting in error prone DNA repair via non homologous end joining and ultimately mutations that eliminate functional protein. Recent studies reflect substantial effort to optimize delivery and biallelic disruption of CCR5 in vitro. CCR5 disruption by the CRISPR/Cas9 system has been a widely pursued strategy. Studies in the past year have demonstrated high efficiency of CCR5 gene disruption using both lentiviral delivery to CD4+ T cell lines [17] and chimeric adenoviral vector delivery to primary T cells [18]. Gene disruption by other endonucleases has also shown promise, as a CCR5-targeting TALEN achieved over 50% knockout in primary T cells when delivered by mRNA electroporation [19].
The first human trial of CCR5 disruption demonstrated the safety and promise of this approach. Patients were treated with ZFN-modified autologous CD4 T cells, and 4 patients completed a 12-week structured treatment interruption during which viral load initially rebounded but then showed sustained decline [20]. The modified CD4+T cells persisted at a significantly higher rate than unmodified cells and were detectable during long term follow up, demonstrating potential selective advantage and long-term persistence of modified cells. One participant’s HIV decreased to undetectable levels; this patient was identified to be heterozygous for CCR5Δ32, hinting that profound viral suppression in the absence of ART can be achieved with a sufficiently large pool of resistant CD4+ cells.
While modifying a patient’s CD4+ T cells may boost immune function, gene editing of hematopoietic stem cells (HSC) is of particular interest for HIV cure because autologous transplant and engraftment of edited HSCs would provide a lifelong source of HIV resistant cells. Just over a year ago, CRISPR/Cas9 was first used to efficiently disrupt CCR5 in human hematopoietic stem and progenitor cells [21]. In this study, nucleofection was used to deliver CRISPR/Cas9 and guide RNA, but recently adenovirus and adeno-associated virus vectors have proven to be particularly successful delivery strategies for HSC gene editing [22-24]. Induced pluripotent stem cells (iPSCs) are under investigation as well, with the idea that modified iPSCs could differentiate into hematopoietic cells. Ye et al and Kang et al both demonstrated efficient bilallelic disruption of CCR5 in iPSCs and additionally showed efficient differentiation of modified cells [25,26]. Along the same lines, mesenchymal stem cells have been gene edited and converted to CD34+ progenitor cells [27]. Studies in humanized mice suggest that modified hematopoietic stem and progenitor cells can engraft and support multilineage differentiation [28,29].
Incorporating additional anti-viral cassettes such as fusion inhibitors is an emerging theme that may further support sustained reservoir reduction. Multiple studies are already pursuing combinatorial approaches. Sather et al used a delete-and-replace strategy to efficiently knock out CCR5 and introduce the antiviral CD46 fusion inhibitor in mobilized CD34+ cells [24]. Two recent studies in humanized mice have used a similar approach. In the first, hematopoietic stem cells were modified to express the C46 peptide fusion inhibitor as well as a short hairpin (sh)RNA suppressing CCR5 expression [28]. In the second, CD4+ T cells were modified to express C46 and a modulator of CCR5 expression, the P2-CCL5 intrakine [30]. Both studies showed protection of CD4+ T cells and significantly reduced viral loads in animals receiving modified cells. Expression of other antiviral transgenes, such as the restriction factor TRIM5alpha [31] or RNA interference targeting HIV genome [32,33] [34] are also pursued.
Several factors jeopardize the success of co-receptor knockout strategies. A recent modeling study predicts that knocking out CCR5 for HIV cure will not decrease viral load long term unless combined with other strategies such as an incorporated suicide gene [35]. The emergence of CXCR4-tropic variants is another concern. CRISPRs have also been used to knock out CXCR4 in primary T cells, resulting in no hindrance to cell growth [36]. However, unlike CCR5, CXCR4 is essential for immune cell development [37,38]. In rare cases CCR5Δ32 individuals can be infected with X4 virus, though disease progression appears to be slower than in CCR5 WT individuals [39,40]. Only a small number of X4 variants were found by deep sequencing in the Berlin patient, suggesting that very low levels of X4 variants may not promote receptor switching [41]. However, re-infection of CCR5-lacking cells can clearly happen if a significant number of X4 variants are present, as has already occurred in a CCR5Δ32 transplant recipient [42]. Despite potential hurdles, several clinical trials evaluating the safety and efficacy of CCR5 disruption are underway including NCT01252641, NCT01044654 and NCT02500849.
Vectored delivery of antibodies and antibody-like molecules
Antibodies play a crucial role in controlling and preventing viral infections. A promising gene therapy approach for HIV cure is persistent in vivo expression of anti-HIV antibodies or antibody-like immunoadhesins. Passive transfer of broadly neutralizing antibodies (bNAbs) can protect primates from lentiviral challenge and significantly reduce viremia [43-45]. Eliciting bNAbs through vaccination has proved challenging, driving the use of vectored strategies which circumvent reliance on the host immune system. In these strategies, a viral or nonviral vector is used to deliver transgenes encoding an antibody with the goal of attaining long-term in vivo expression. Recombinant adeno-associated virus (rAAV) has been extensively studied in these efforts due to its efficient expression of transgene and nonpathogenicity. Such a system could provide a sterilizing barrier to HIV acquisition, and might also be used as a therapeutic vaccine in HIV cure efforts. Antibodies can mediate cellular killing via antibody-dependent cellular cytotoxicity (ADCC) and antibody-dependent cellular phagocytosis (ADCP), and thus could facilitate clearance of rare, reactivating T cells exhibiting surface Env expression.
Vector-expressed bNAbs can protect humanized mice from HIV-1 challenge [46,47], and in primates, an rAAV delivered immunoadhesin conferred long-lived albeit partial protection from SIV challenge [48]. Primate protection from SIV challenge has recently been reported with AAV vector expressing the simianized form of the VRC07 bNAb [49]. Recent work has tackled secondary challenges, such as testing harder-to-neutralize SIV strains and strategies to avoid anti-antibody responses, a major obstacle in developing effective vectored antibody therapies [50]. Fuchs et al recently showed that use of a fully simianized, authentic IgG structure does not ameliorate the immunogenicity of a foreign antibody as hoped [51]. However, protection from SIVmac239 was still observed. Rhesus macaques injected with rAAV encoding the monoclonal antibody 5L7 exhibited delayed peak viremia and lower viral loads at peak viremia and set point. Importantly, 5L7 was unable to neutralize the challenge virus SIVmac239, highlighting the important role of non-neutralizing antibodies in controlling infection. Non-neutralizing antibodies can mediate ADCC and ADCP by recruiting effector cells like NK-cells and macrophages to promote lysis or phagocytosis of infected cells. Thorough investigation of these antibody roles is especially important in light of the RV144 vaccine trial, wherein ADCC activity was correlated with lower acquisition of infection [52].
In an exciting approach that may address the issues of anti-HIV breadth and immunogenicity, Gardner et al used an engineered immunoadhesin form of CD4 fused to a CCR5 mimetic sulfopeptide, which they termed eCD4-Ig [53]. This immunoadhesin exhibits exceptional breadth and potency of neutralization compared to the best isolated bNAbs. It binds conserved regions of Env, and neutralizes neutralization-resistant tier 2 and tier 3 HIV-1 as well as HIV-2 and SIVmac. Further, eCD4-Ig mediated 30-40 times more killing by NK cells than IgG b12, a known strong mediator of ADCC. Expression of eCD4-Ig by rAAV protected macaques from multiple, increasing SHIV-AD8 challenges, and importantly, eCD4-Ig was not immunogenic and was detectable at protective concentrations in sera for 40 weeks.
One outstanding issue is the long-term safety of AAV delivered transgenes. The ability to turn off transgene expression in the case of adverse events is desirable but not yet feasible. Numerous clinic trials have shown no adverse events following transduction of AAV, though potential for transgene or vector integration to drive cancer remains. AAV2 sequences have been found integrated in hepatocarcinomas, particularly in cancer driving genes [54]. This may give pause to AAV vectored gene therapy research, but the implications of this finding are contested [55,56]. Additionally, widespread preexisting immunity to AAV serotypes 1 and 2 might limit the target patient population for AAV vectored gene therapies, although the use of rare capsids or recombinant capsids may circumvent this problem [57]
CAR T cell therapy
Use of engineered T cells is gaining ground as an alternative strategy in HIV cure research. Here, T cells are engineered to specifically target HIV, and then reinfused into a patient. This concept has been pursued as a potential HIV treatment for decades, but initial attempts failed to provide durable benefit [58]. However, recent major advancements have been made using adoptive T cell therapy to target and kill cancer cells, rekindling interest in using T cell therapies for an HIV cure. In these treatments, a patient is infused with genetically modified T cells expressing a chimeric antigen receptor (CAR). A CAR is a hybrid surface receptor composed of two domains: a target-specific antigen recognition domain from a B cell receptor fused to an intracellular T cell activation domain. Essential to the recent success of these therapies is the inclusion of costimulatory domains, such as CD28 and CD137 [59-61], as these ensure robust T cell activation and expansion after infusion. CAR T cell technology has been most effectively employed in the treatment of hematologic malignancies, particularly B cell malignancies that use CARs targeted toward the pan B cell receptor CD19 [62,63]. Dramatic remissions of aggressive malignancies like B cell acute lymphoblastic leukemia have highlighted the power of immunotherapies [64,65].
The excitement surrounding the robust anti-tumor efficacy of the recent CAR trials has reignited interest in employing CAR T cell therapy for HIV cure. Rather than a tumor-specific antigen, the CAR targets the HIV gp120 envelope glycoprotein on the surface of infected cells, for example by using CD4 or the antigen binding domain of a broadly neutralizing antibody. Robust persistence of HIV-specific CAR T cells has previously been demonstrated, in some cases up to 11 years [66]. Additionally, CAR T cells can be combined with CCR5 knockout [24], which is not only of interest for possible synergistic therapy but also because CD8+ T cells expressing CD4-like molecules are vulnerable to HIV infection [67]. Recently, Zhen et al created human hematopoietic stem progenitor cells expressing a CD4ζ CAR. The cells also expressed short hairpin RNAs to interfere with CCR5 and the HIV LTR. These HSCs were able to differentiate into multiple lineages of functional immune cells. When genetically modified cells were transplanted into humanized mice, they suppressed HIV replication in vivo, and mice with greater expansion of the CAR T cells had near complete suppression of viremia [68].
Targeting the HIV genome directly
A final, promising approach to reservoir reduction is direct disruption of the HIV genome by engineered endonucleases. Strategies include the lethal mutation of essential viral genes or excision of proviral DNA by targeting the LTRs flanking the HIV provirus. Unlike other proposed cure strategies, provirus editing does not require reactivation of latently infected cells to eliminate the reservoir; instead it is a direct strategy to eliminate viable viral protein expression. Despite the appeal, there remain substantial hurdles to viability in clinical application. The major limiting factors are a lack of means to effectively deliver therapeutic enzymes to the reservoir in vivo, and the high efficiency of delivery and editing required to meaningfully reduce the reservoir. Additional concerns such as the potential immunogenicity of gene editing enzymes, toxicity due to off-target cleavage, and the potential development of treatment resistance will need to be addressed. Indeed, one study identified a mutational insertion in a highly conserved region of pol that provided resistance to subsequent endonuclease cleavage but did not lethally mutate the virus [69]. In another study, virus escaped CRISPR/Cas9 targeting of several conserved regions of the HIV genome [70]. These results highlight the need for careful choice of target and suggest that simultaneous targeting of multiple essential genes may be necessary.
Despite these obstacles, HIV genome editing remains actively pursued. Various gene-editing platforms including ZFNs, CRISPRs, meganucleases, and TALENS have shown the potential applicability of this strategy in vitro [71-76]. Additionally, new technologies like droplet digital PCR are improving the ease and accuracy of mutation detection [77]. In particular, the efficiency, simplicity, and specificity of gene disruption using the CRISPR/Cas9 system has driven recent interest in HIV genome targeting. [72,74,76,78,79]. CRISPR/Cas9 can operate with very high specificity, demonstrated by highly specific HIV excision using CRISPR/Cas9 guides designed to avoid off-target cutting [73]. As with many gene therapy strategies, efficient delivery of these enzymes to the appropriate target cells remains a challenge, though progress is being made. For instance, a group recently engineered a measles virus hemagglutinin to target CD4 [80]. Lentivirus pseudotyped with this molecule could genetically modify 2% of resting CD4+ cells in vivo.
Conclusions
The therapies discussed above may be even more effective in combination, for instance using CCR5 knockout along with CAR T cells or a therapeutic vaccine. CAR T cell or vectorized antibody therapies would likely best be combined with latency-reversing agents, to maximize HIV antigen expression on reservoir cells. While initial studies of latency reversing agents showed limited induction of viral RNA expression in vivo [81], new reagents like GS-9620, a TLR-7 agonist developed by Gilead, seem very promising and may prove useful with CAR T cell therapies [82]. Furthermore, since HIV-specific CD4+ and CD8+T cells can lose their effector functions and proliferative capacity in a phenomenon known as immune exhaustion [83] mediated by programed death 1 (PD-1), blockade of the PD-1 pathway may restore impaired anti-HIV immune responses [84], [85]. This may be of particular importance for cell-based therapies such as anti-HIV CAR T cells.
HIV infects over 36 million people worldwide, causing immense global disease and financial burdens. Many exciting and creative gene therapy strategies have emerged at the forefront of HIV cure research. CCR5 knockout and knockdown are the most clinically developed strategies today, but technical advances in cell manipulation will be required before they can be implemented on a global scale. We hope to witness soon successful proof of concept experiments and additional patients cured of HIV, along with progress and continued innovation on more widely implementable curative strategies.
Key points.
1) To date, CCR5 knockout or knockdown is the most progressed gene therapy strategy for HIV cure with a clinical trial in humans showing safety of therapy, persistence of gene modified cells, and promising clinical benefit.
2) The immunoadhesin eCD4-Ig is an ideal candidate to deliver as a therapeutic vaccine, showing exceptionally broad and potent neutralization of HIV isolates and exhibiting robust ADCC activity.
3) CAR T cells have demonstrated efficacy in attacking aggressive blood cancers and are a promising tool to target and kill HIV infected cells.
Acknowledgements
We thank Daniel Stone and Daniel Strongin for critical reading of the manuscript.
Financial Support and Sponsorship
This work was funded by NIH supported Martin Delaney Collaboratory grant U19 AI 096111 and in part by a developmental grant from the University of Washington Center for AIDS Research (CFAR), an NIH funded program under award number P30 AI 027757 which is supported by the following NIH Institutes and Centers (NIAID, NCI, NIMH, NIDA, NICHD, NHLBI, NIA, NIGMS and NIDDK).
Footnotes
Conflicts of Interest
None
References
- 1.Chomont N, El-Far M, Ancuta P, Trautmann L, Procopio FA, Yassine-Diab B, Boucher G, Boulassel MR, Ghattas G, Brenchley JM, et al. HIV reservoir size and persistence are driven by T cell survival and homeostatic proliferation. Nat Med. 2009;15:893–900. doi: 10.1038/nm.1972. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Soriano-Sarabia N, Archin NM, Bateson R, Dahl NP, Crooks AM, Kuruc JD, Garrido C, Margolis DM. Peripheral Vgamma9Vdelta2 T Cells Are a Novel Reservoir of Latent HIV Infection. PLoS Pathog. 2015;11:e1005201. doi: 10.1371/journal.ppat.1005201. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Finzi D, Blankson J, Siliciano JD, Margolick JB, Chadwick K, Pierson T, Smith K, Lisziewicz J, Lori F, Flexner C, 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. doi: 10.1038/8394. [DOI] [PubMed] [Google Scholar]
- 4.Siliciano JD, Kajdas J, Finzi D, Quinn TC, Chadwick K, Margolick JB, Kovacs C, Gange SJ, Siliciano RF. Long-term follow-up studies confirm the stability of the latent reservoir for HIV-1 in resting CD4+ T cells. Nat Med. 2003;9:727–728. doi: 10.1038/nm880. [DOI] [PubMed] [Google Scholar]
- 5.Hill AL, Rosenbloom DI, Fu F, Nowak MA, Siliciano RF. Predicting the outcomes of treatment to eradicate the latent reservoir for HIV-1. Proc Natl Acad Sci U S A. 2014;111:13475–13480. doi: 10.1073/pnas.1406663111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Hocqueloux L, Prazuck T, Avettand-Fenoel V, Lafeuillade A, Cardon B, Viard JP, Rouzioux C. Long-term immunovirologic control following antiretroviral therapy interruption in patients treated at the time of primary HIV-1 infection. AIDS. 2010;24:1598–1601. doi: 10.1097/qad.0b013e32833b61ba. [DOI] [PubMed] [Google Scholar]
- 7.Saez-Cirion A, Bacchus C, Hocqueloux L, Avettand-Fenoel V, Girault I, Lecuroux C, Potard V, Versmisse P, Melard A, Prazuck T, 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:e1003211. doi: 10.1371/journal.ppat.1003211. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Persaud D, Gay H, Ziemniak C, Chen YH, Piatak M, Jr., Chun TW, Strain M, Richman D, Luzuriaga K. Absence of detectable HIV-1 viremia after treatment cessation in an infant. N Engl J Med. 2013;369:1828–1835. doi: 10.1056/NEJMoa1302976. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Hutter G, Nowak D, Mossner M, Ganepola S, Mussig A, Allers K, Schneider T, Hofmann J, Kucherer C, Blau O, et al. Long-term control of HIV by CCR5 Delta32/Delta32 stem-cell transplantation. N Engl J Med. 2009;360:692–698. doi: 10.1056/NEJMoa0802905. [DOI] [PubMed] [Google Scholar]
- 10.Allers K, Hutter G, Hofmann J, Loddenkemper C, Rieger K, Thiel E, Schneider T. Evidence for the cure of HIV infection by CCR5Delta32/Delta32 stem cell transplantation. Blood. 2011;117:2791–2799. doi: 10.1182/blood-2010-09-309591. [DOI] [PubMed] [Google Scholar]
- 11.Mavigner M, Watkins B, Lawson B, Lee ST, Chahroudi A, Kean L, Silvestri G. Persistence of virus reservoirs in ART-treated SHIV-infected rhesus macaques after autologous hematopoietic stem cell transplant. PLoS Pathog. 2014;10:e1004406. doi: 10.1371/journal.ppat.1004406. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Henrich TJ, Hanhauser E, Marty FM, Sirignano MN, Keating S, Lee TH, Robles YP, Davis BT, Li JZ, Heisey A, et al. Antiretroviral-free HIV-1 remission and viral rebound after allogeneic stem cell transplantation: report of 2 cases. Ann Intern Med. 2014;161:319–327. doi: 10.7326/M14-1027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Duarte RF, Salgado M, Sanchez-Ortega I, Arnan M, Canals C, Domingo-Domenech E, Fernandez-de-Sevilla A, Gonzalez-Barca E, Moron-Lopez S, Nogues N, et al. CCR5 Delta32 homozygous cord blood allogeneic transplantation in a patient with HIV: a case report. Lancet HIV. 2015;2:e236–242. doi: 10.1016/S2352-3018(15)00083-1. [DOI] [PubMed] [Google Scholar]
- 14.Shimizu S, Yadav SS, An DS. Stable Delivery of CCR5-Directed shRNA into Human Primary Peripheral Blood Mononuclear Cells and Hematopoietic Stem/Progenitor Cells via a Lentiviral Vector. Methods Mol Biol. 2016;1364:235–248. doi: 10.1007/978-1-4939-3112-5_19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Perez EE, Wang J, Miller JC, Jouvenot Y, Kim KA, Liu O, Wang N, Lee G, Bartsevich VV, Lee YL, et al. Establishment of HIV-1 resistance in CD4+ T cells by genome editing using zinc-finger nucleases. Nat Biotechnol. 2008;26:808–816. doi: 10.1038/nbt1410. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Holt N, Wang J, Kim K, Friedman G, Wang X, Taupin V, Crooks GM, Kohn DB, Gregory PD, Holmes MC, 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. doi: 10.1038/nbt.1663. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Wang W, Ye C, Liu J, Zhang D, Kimata JT, Zhou P. CCR5 gene disruption via lentiviral vectors expressing Cas9 and single guided RNA renders cells resistant to HIV-1 infection. PLoS One. 2014;9:e115987. doi: 10.1371/journal.pone.0115987. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Li C, Guan X, Du T, Jin W, Wu B, Liu Y, Wang P, Hu B, Griffin GE, Shattock RJ, et al. Inhibition of HIV-1 infection of primary CD4+ T-cells by gene editing of CCR5 using adenovirus-delivered CRISPR/Cas9. J Gen Virol. 2015;96:2381–2393. doi: 10.1099/vir.0.000139. [DOI] [PubMed] [Google Scholar]
- 19.Mock U, Machowicz R, Hauber I, Horn S, Abramowski P, Berdien B, Hauber J, Fehse B. mRNA transfection of a novel TAL effector nuclease (TALEN) facilitates efficient knockout of HIV co-receptor CCR5. Nucleic Acids Res. 2015;43:5560–5571. doi: 10.1093/nar/gkv469. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Tebas P, Stein D, Tang WW, Frank I, Wang SQ, Lee G, Spratt SK, Surosky RT, Giedlin MA, Nichol G, et al. Gene editing of CCR5 in autologous CD4 T cells of persons infected with HIV. N Engl J Med. 2014;370:901–910. doi: 10.1056/NEJMoa1300662. [20. **Tebas 2014This paper describes the first human clinical trial testing reinfusion of CD4+ T cells genetically modified to knock out CCR5.] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Mandal PK, Ferreira LM, Collins R, Meissner TB, Boutwell CL, Friesen M, Vrbanac V, Garrison BS, Stortchevoi A, Bryder D, et al. Efficient ablation of genes in human hematopoietic stem and effector cells using CRISPR/Cas9. Cell Stem Cell. 2014;15:643–652. doi: 10.1016/j.stem.2014.10.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Wang J, DeClercq JJ, Hayward SB, Li PW, Shivak DA, Gregory PD, Lee G, Holmes MC. Highly efficient homology-driven genome editing in human T cells by combining zinc-finger nuclease mRNA and AAV6 donor delivery. Nucleic Acids Res. 2015 doi: 10.1093/nar/gkv1121. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Saydaminova K, Ye X, Wang H, Richter M, Ho M, Chen H, Xu N, Kim JS, Papapetrou E, Holmes MC, et al. Efficient genome editing in hematopoietic stem cells with helper-dependent Ad5/35 vectors expressing site-specific endonucleases under microRNA regulation. Mol Ther Methods Clin Dev. 2015;1:14057. doi: 10.1038/mtm.2014.57. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Sather BD, Romano Ibarra GS, Sommer K, Curinga G, Hale M, Khan IF, Singh S, Song Y, Gwiazda K, Sahni J, et al. Efficient modification of CCR5 in primary human hematopoietic cells using a megaTAL nuclease and AAV donor template. Sci Transl Med. 2015;7:307ra156. doi: 10.1126/scitranslmed.aac5530. [24. *Sather 2015This report demonstrates gene editing of CD34+ cells to disrupt CCR5 and knock in antiviral genes.] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Ye L, Wang J, Beyer AI, Teque F, Cradick TJ, Qi Z, Chang JC, Bao G, Muench MO, Yu J, et al. Seamless modification of wild-type induced pluripotent stem cells to the natural CCR5Delta32 mutation confers resistance to HIV infection. Proc Natl Acad Sci U S A. 2014;111:9591–9596. doi: 10.1073/pnas.1407473111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Kang H, Minder P, Park MA, Mesquitta WT, Torbett BE, Slukvin II. CCR5 Disruption in Induced Pluripotent Stem Cells Using CRISPR/Cas9 Provides Selective Resistance of Immune Cells to CCR5-tropic HIV-1 Virus. Mol Ther Nucleic Acids. 2015;4:e268. doi: 10.1038/mtna.2015.42. [DOI] [PubMed] [Google Scholar]
- 27.Manotham K, Chattong S, Setpakdee A. Generation of CCR5-defective CD34 cells from ZFN-driven stop codon-integrated mesenchymal stem cell clones. J Biomed Sci. 2015;22:25. doi: 10.1186/s12929-015-0130-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Burke BP, Levin BR, Zhang J, Sahakyan A, Boyer J, Carroll MV, Colon JC, Keech N, Rezek V, Bristol G, et al. Engineering Cellular Resistance to HIV-1 Infection In Vivo Using a Dual Therapeutic Lentiviral Vector. Mol Ther Nucleic Acids. 2015;4:e236. doi: 10.1038/mtna.2015.10. [28. *Burke 2015This paper showed CD34+ genetically modified to express the fusion inhibitor C46 and to downregulate CCR5 by expressing shRNA. These cells engrafted in mice and resulted in reduced viral load.] [DOI] [PubMed] [Google Scholar]
- 29.Myburgh R, Ivic S, Pepper MS, Gers-Huber G, Li D, Audige A, Rochat MA, Jaquet V, Regenass S, Manz MG, et al. Lentivector Knockdown of CCR5 in Hematopoietic Stem and Progenitor Cells Confers Functional and Persistent HIV-1 Resistance in Humanized Mice. J Virol. 2015;89:6761–6772. doi: 10.1128/JVI.00277-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Petit NY, Baillou C, Burlion A, Dorgham K, Levacher B, Amiel C, Schneider V, Lemoine FM, Gorochov G, Marodon G. Gene transfer of two entry inhibitors protects CD4 T cell from HIV-1 infection in humanized mice. Gene Ther. 2015 doi: 10.1038/gt.2015.101. [30. *Petit 2015This paper demonstrates CD4+ cells modified to express two separate entry inhibitors decreased viremia in humanized mice.] [DOI] [PubMed] [Google Scholar]
- 31.Jung U, Urak K, Veillette M, Nepveu-Traversy ME, Pham QT, Hamel S, Rossi JJ, Berthoux L. Preclinical Assessment of Mutant Human TRIM5alpha as an Anti-HIV-1 Transgene. Hum Gene Ther. 2015;26:664–679. doi: 10.1089/hum.2015.059. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Scarborough RJ, Adams KL, Daher A, Gatignol A. Effective inhibition of HIV-1 production by short hairpin RNAs and small interfering RNAs targeting a highly conserved site in HIV-1 Gag RNA is optimized by evaluating alternative length formats. Antimicrob Agents Chemother. 2015;59:5297–5305. doi: 10.1128/AAC.00949-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Herrera-Carrillo E, Berkhout B. Gene therapy strategies to block HIV-1 replication by RNA interference. Adv Exp Med Biol. 2015;848:71–95. doi: 10.1007/978-1-4939-2432-5_4. [DOI] [PubMed] [Google Scholar]
- 34.Cheng Q, Dong L, Zhang F, Yuan P, Li Z, Sun Y, Yin J, Peng B, He X, Liu W. Short communication: Efficiently inhibiting HIV-1 replication by a prototype foamy virus vector expressing novel H1 promoter-driven short hairpin RNAs. AIDS Res Hum Retroviruses. 2015;31:183–188. doi: 10.1089/AID.2014.0217. [DOI] [PubMed] [Google Scholar]
- 35.Pandit A, de Boer RJ. HIV-1 CCR5 gene therapy will fail unless it is combined with a suicide gene. Sci Rep. 2015;5:18088. doi: 10.1038/srep18088. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Hou P, Chen S, Wang S, Yu X, Chen Y, Jiang M, Zhuang K, Ho W, Hou W, Huang J, et al. Genome editing of CXCR4 by CRISPR/cas9 confers cells resistant to HIV-1 infection. Sci Rep. 2015;5:15577. doi: 10.1038/srep15577. [36. *Hou 2015This paper demonstrates efficient disruption of the coreceptor CXCR4 using CRISPR/Cas9 and shows that division and propagation of primary CD4+ T cells is not affected.] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Dar A, Kollet O, Lapidot T. Mutual, reciprocal SDF-1/CXCR4 interactions between hematopoietic and bone marrow stromal cells regulate human stem cell migration and development in NOD/SCID chimeric mice. Exp Hematol. 2006;34:967–975. doi: 10.1016/j.exphem.2006.04.002. [DOI] [PubMed] [Google Scholar]
- 38.Nagasawa T, Nakajima T, Tachibana K, Iizasa H, Bleul CC, Yoshie O, Matsushima K, Yoshida N, Springer TA, Kishimoto T. Molecular cloning and characterization of a murine pre-B-cell growth-stimulating factor/stromal cell-derived factor 1 receptor, a murine homolog of the human immunodeficiency virus 1 entry coreceptor fusin. Proc Natl Acad Sci U S A. 1996;93:14726–14729. doi: 10.1073/pnas.93.25.14726. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Le AQ, Taylor J, Dong W, McCloskey R, Woods C, Danroth R, Hayashi K, Milloy MJ, Poon AF, Brumme ZL. Differential evolution of a CXCR4-using HIV-1 strain in CCR5wt/wt and CCR532/32 hosts revealed by longitudinal deep sequencing and phylogenetic reconstruction. Sci Rep. 2015;5:17607. doi: 10.1038/srep17607. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Henrich TJ, Hanhauser E, Harrison LJ, Palmer CD, Romero-Tejeda M, Jost S, Bosch RJ, Kuritzkes DR. CCR5-Delta32 Heterozygosity, HIV-1 Reservoir Size, and Lymphocyte Activation in Individuals Receiving Long-term Suppressive Antiretroviral Therapy. J Infect Dis. 2015 doi: 10.1093/infdis/jiv504. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Symons J, Vandekerckhove L, Hutter G, Wensing AM, van Ham PM, Deeks SG, Nijhuis M. Dependence on the CCR5 coreceptor for viral replication explains the lack of rebound of CXCR4-predicted HIV variants in the Berlin patient. Clin Infect Dis. 2014;59:596–600. doi: 10.1093/cid/ciu284. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Kordelas L, Verheyen J, Beelen DW, Horn PA, Heinold A, Kaiser R, Trenschel R, Schadendorf D, Dittmer U, Esser S, et al. Shift of HIV tropism in stem-cell transplantation with CCR5 Delta32 mutation. N Engl J Med. 2014;371:880–882. doi: 10.1056/NEJMc1405805. [DOI] [PubMed] [Google Scholar]
- 43.Barouch DH, Whitney JB, Moldt B, Klein F, Oliveira TY, Liu J, Stephenson KE, Chang HW, Shekhar K, Gupta S, et al. Therapeutic efficacy of potent neutralizing HIV-1-specific monoclonal antibodies in SHIV-infected rhesus monkeys. Nature. 2013;503:224–228. doi: 10.1038/nature12744. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Shingai M, Nishimura Y, Klein F, Mouquet H, Donau OK, Plishka R, Buckler-White A, Seaman M, Piatak M, Jr., Lifson JD, et al. Antibody-mediated immunotherapy of macaques chronically infected with SHIV suppresses viraemia. Nature. 2013;503:277–280. doi: 10.1038/nature12746. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Shingai M, Donau OK, Plishka RJ, Buckler-White A, Mascola JR, Nabel GJ, Nason MC, Montefiori D, Moldt B, Poignard P, et al. Passive transfer of modest titers of potent and broadly neutralizing anti-HIV monoclonal antibodies block SHIV infection in macaques. J Exp Med. 2014;211:2061–2074. doi: 10.1084/jem.20132494. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Balazs AB, Chen J, Hong CM, Rao DS, Yang L, Baltimore D. Antibody-based protection against HIV infection by vectored immunoprophylaxis. Nature. 2012;481:81–84. doi: 10.1038/nature10660. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Lewis AD, Chen R, Montefiori DC, Johnson PR, Clark KR. Generation of neutralizing activity against human immunodeficiency virus type 1 in serum by antibody gene transfer. J Virol. 2002;76:8769–8775. doi: 10.1128/JVI.76.17.8769-8775.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Johnson PR, Schnepp BC, Zhang J, Connell MJ, Greene SM, Yuste E, Desrosiers RC, Clark KR. Vector-mediated gene transfer engenders long-lived neutralizing activity and protection against SIV infection in monkeys. Nat Med. 2009;15:901–906. doi: 10.1038/nm.1967. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Saunders KO, Wang L, Joyce MG, Yang ZY, Balazs AB, Cheng C, Ko SY, Kong WP, Rudicell RS, Georgiev IS, et al. Broadly Neutralizing Human Immunodeficiency Virus Type 1 Antibody Gene Transfer Protects Nonhuman Primates from Mucosal Simian-Human Immunodeficiency Virus Infection. J Virol. 2015;89:8334–8345. doi: 10.1128/JVI.00908-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Martinez-Navio JM, Fuchs SP, Pedreno-Lopez S, Rakasz EG, Gao G, Desrosiers RC. Host Anti-antibody Responses Following Adeno-associated Virus-mediated Delivery of Antibodies Against HIV and SIV in Rhesus Monkeys. Mol Ther. 2015 doi: 10.1038/mt.2015.191. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Fuchs SP, Martinez-Navio JM, Piatak M, Jr., Lifson JD, Gao G, Desrosiers RC. AAV-Delivered Antibody Mediates Significant Protective Effects against SIVmac239 Challenge in the Absence of Neutralizing Activity. PLoS Pathog. 2015;11:e1005090. doi: 10.1371/journal.ppat.1005090. [51. **Fuchs 2015This paper shows that even when antibodies display no neutralizing activity toward a given SIV, they can still mediate protective effects, likely through ADCC or ADCP.] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Haynes BF, Gilbert PB, McElrath MJ, Zolla-Pazner S, Tomaras GD, Alam SM, Evans DT, Montefiori DC, Karnasuta C, Sutthent R, et al. Immune-correlates analysis of an HIV-1 vaccine efficacy trial. N Engl J Med. 2012;366:1275–1286. doi: 10.1056/NEJMoa1113425. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Gardner MR, Kattenhorn LM, Kondur HR, von Schaewen M, Dorfman T, Chiang JJ, Haworth KG, Decker JM, Alpert MD, Bailey CC, et al. AAV-expressed eCD4-Ig provides durable protection from multiple SHIV challenges. Nature. 2015;519:87–91. doi: 10.1038/nature14264. [53. **Gardner 2015This paper shows that eCD4-Ig is more broad and potent than known neutralizing antibodies as well as a potent mediator of ADCC. AAV expressed eCD4-Ig completely protected macaques from SIV challenge.] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Nault JC, Datta S, Imbeaud S, Franconi A, Mallet M, Couchy G, Letouze E, Pilati C, Verret B, Blanc JF, et al. Recurrent AAV2-related insertional mutagenesis in human hepatocellular carcinomas. Nat Genet. 2015;47:1187–1193. doi: 10.1038/ng.3389. [DOI] [PubMed] [Google Scholar]
- 55.Berns KI, Byrne BJ, Flotte TR, Gao G, Hauswirth WW, Herzog RW, Muzyczka N, VandenDriessche T, Xiao X, Zolotukhin S, et al. Adeno-Associated Virus Type 2 and Hepatocellular Carcinoma? Hum Gene Ther. 2015;26:779–781. doi: 10.1089/hum.2015.29014.kib. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Buning H, Schmidt M. Adeno-associated vector toxicity - To be or not ot be. Molecular Therapy. 2015;23:1673–1674. doi: 10.1038/mt.2015.182. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Asokan A, Schaffer DV, Samulski RJ. The AAV vector toolkit: poised at the clinical crossroads. Mol Ther. 2012;20:699–708. doi: 10.1038/mt.2011.287. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Leibman RS, Riley JL. Engineering T Cells to Functionally Cure HIV-1 Infection. Mol Ther. 2015;23:1149–1159. doi: 10.1038/mt.2015.70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Savoldo B, Ramos CA, Liu E, Mims MP, Keating MJ, Carrum G, Kamble RT, Bollard CM, Gee AP, Mei Z, et al. CD28 costimulation improves expansion and persistence of chimeric antigen receptor-modified T cells in lymphoma patients. J Clin Invest. 2011;121:1822–1826. doi: 10.1172/JCI46110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Porter DL, Levine BL, Kalos M, Bagg A, June CH. Chimeric antigen receptor-modified T cells in chronic lymphoid leukemia. N Engl J Med. 2011;365:725–733. doi: 10.1056/NEJMoa1103849. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Maus MV, Grupp SA, Porter DL, June CH. Antibody-modified T cells: CARs take the front seat for hematologic malignancies. Blood. 2014;123:2625–2635. doi: 10.1182/blood-2013-11-492231. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Kochenderfer JN, Yu Z, Frasheri D, Restifo NP, Rosenberg SA. Adoptive transfer of syngeneic T cells transduced with a chimeric antigen receptor that recognizes murine CD19 can eradicate lymphoma and normal B cells. Blood. 2010;116:3875–3886. doi: 10.1182/blood-2010-01-265041. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Kochenderfer JN, Dudley ME, Feldman SA, Wilson WH, Spaner DE, Maric I, Stetler-Stevenson M, Phan GQ, Hughes MS, Sherry RM, et al. B-cell depletion and remissions of malignancy along with cytokine-associated toxicity in a clinical trial of anti-CD19 chimeric-antigen-receptor-transduced T cells. Blood. 2012;119:2709–2720. doi: 10.1182/blood-2011-10-384388. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Brentjens RJ, Davila ML, Riviere I, Park J, Wang X, Cowell LG, Bartido S, Stefanski J, Taylor C, Olszewska M, et al. CD19-targeted T cells rapidly induce molecular remissions in adults with chemotherapy-refractory acute lymphoblastic leukemia. Sci Transl Med. 2013;5:177ra138. doi: 10.1126/scitranslmed.3005930. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Grupp SA, Kalos M, Barrett D, Aplenc R, Porter DL, Rheingold SR, Teachey DT, Chew A, Hauck B, Wright JF, et al. Chimeric antigen receptor-modified T cells for acute lymphoid leukemia. N Engl J Med. 2013;368:1509–1518. doi: 10.1056/NEJMoa1215134. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Scholler J, Brady TL, Binder-Scholl G, Hwang WT, Plesa G, Hege KM, Vogel AN, Kalos M, Riley JL, Deeks SG, et al. Decade-long safety and function of retroviral-modified chimeric antigen receptor T cells. Sci Transl Med. 2012;4:132ra153. doi: 10.1126/scitranslmed.3003761. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Kamata M, Kim PY, Ng HL, Ringpis GE, Kranz E, Chan J, O'Connor S, Yang OO, Chen IS. Ectopic expression of anti-HIV-1 shRNAs protects CD8(+) T cells modified with CD4zeta CAR from HIV-1 infection and alleviates impairment of cell proliferation. Biochem Biophys Res Commun. 2015;463:216–221. doi: 10.1016/j.bbrc.2015.05.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Zhen A, Kamata M, Rezek V, Rick J, Levin B, Kasparian S, Chen IS, Yang OO, Zack JA, Kitchen SG. HIV-specific Immunity Derived From Chimeric Antigen Receptor-engineered Stem Cells. Mol Ther. 2015;23:1358–1367. doi: 10.1038/mt.2015.102. [68. *Zhen 2015This paper shows CAR T cell therapy can result in decrease of HIV viremia in an animal model.] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.De Silva Feelixge HS, Stone D, Pietz HL, Roychoudhury P, Greninger AL, Schiffer JT, Aubert M, Jerome KR. Detection of treatment-resistant infectious HIV after genome-directed antiviral endonuclease therapy. Antiviral Res. 2015 doi: 10.1016/j.antiviral.2015.12.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Wang G, Zhao N, Berkhout B, Das AT. CRISPR-Cas9 can inhibit HIV-1 replication but NHEJ repair facilitates virus escape. Mol Ther. 2016 doi: 10.1038/mt.2016.24. [70. *Wang 2016This paper shows that while HIV genomes can be mutated by CRISPR/Cas9, the virus rapidly escapes targeting.] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Qu X, Wang P, Ding D, Li L, Wang H, Ma L, Zhou X, Liu S, Lin S, Wang X, et al. Zinc-finger-nucleases mediate specific and efficient excision of HIV-1 proviral DNA from infected and latently infected human T cells. Nucleic Acids Res. 2013;41:7771–7782. doi: 10.1093/nar/gkt571. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Ebina H, Misawa N, Kanemura Y, Koyanagi Y. Harnessing the CRISPR/Cas9 system to disrupt latent HIV-1 provirus. Sci Rep. 2013;3:2510. doi: 10.1038/srep02510. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Hu W, Kaminski R, Yang F, Zhang Y, Cosentino L, Li F, Luo B, Alvarez-Carbonell D, Garcia-Mesa Y, Karn J, et al. RNA-directed gene editing specifically eradicates latent and prevents new HIV-1 infection. Proc Natl Acad Sci U S A. 2014;111:11461–11466. doi: 10.1073/pnas.1405186111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Zhu W, Lei R, Le Duff Y, Li J, Guo F, Wainberg MA, Liang C. The CRISPR/Cas9 system inactivates latent HIV-1 proviral DNA. Retrovirology. 2015;12:22. doi: 10.1186/s12977-015-0150-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Ebina H, Kanemura Y, Misawa N, Sakuma T, Kobayashi T, Yamamoto T, Koyanagi Y. A high excision potential of TALENs for integrated DNA of HIV-based lentiviral vector. PLoS One. 2015;10:e0120047. doi: 10.1371/journal.pone.0120047. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Liao HK, Gu Y, Diaz A, Marlett J, Takahashi Y, Li M, Suzuki K, Xu R, Hishida T, Chang CJ, et al. Use of the CRISPR/Cas9 system as an intracellular defense against HIV-1 infection in human cells. Nat Commun. 2015;6:6413. doi: 10.1038/ncomms7413. [76. *Liao 2015Researchers created induced pluripotent stem cells that express anti-HIV targeted CRISPR/Cas9] [DOI] [PubMed] [Google Scholar]
- 77.R Hall Sedlak, Niyonzima Nixon, De Silva Feelixge Harshana S., Roychoudhury Pavitra, Greninger Alexander L., Weber Nicholas D., Boissel Sandrine, Scharenberg Andrew M., Cheng Anqi, Magaret Amalia, Bumgarner Roger, Stone Daniel, Jerome Keith R. Digital detection of endonuclease mediated gene disruption in the HIV provirus. Scientific Reports. 2016;6 doi: 10.1038/srep20064. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Schumann K, Lin S, Boyer E, Simeonov DR, Subramaniam M, Gate RE, Haliburton GE, Ye CJ, Bluestone JA, Doudna JA, et al. Generation of knock-in primary human T cells using Cas9 ribonucleoproteins. Proc Natl Acad Sci U S A. 2015;112:10437–10442. doi: 10.1073/pnas.1512503112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Dampier W, Nonnemacher MR, Sullivan NT, Jacobson JM, Wigdahl B. HIV Excision Utilizing CRISPR/Cas9 Technology: Attacking the Proviral Quasispecies in Reservoirs to Achieve a Cure. MOJ Immunol. 2014;1 doi: 10.15406/moji.2014.01.00022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Zhou Q, Uhlig KM, Muth A, Kimpel J, Levy C, Munch RC, Seifried J, Pfeiffer A, Trkola A, Coulibaly C, et al. Exclusive Transduction of Human CD4+ T Cells upon Systemic Delivery of CD4-Targeted Lentiviral Vectors. J Immunol. 2015;195:2493–2501. doi: 10.4049/jimmunol.1500956. [DOI] [PubMed] [Google Scholar]
- 81.Archin NM, Liberty AL, Kashuba AD, Choudhary SK, Kuruc JD, Crooks AM, Parker DC, Anderson EM, Kearney MF, Strain MC, et al. Administration of vorinostat disrupts HIV-1 latency in patients on antiretroviral therapy. Nature. 2012;487:482–485. doi: 10.1038/nature11286. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Gane EJ, Lim YS, Gordon SC, Visvanathan K, Sicard E, Fedorak RN, Roberts S, Massetto B, Ye Z, Pflanz S, et al. The oral toll-like receptor-7 agonist GS-9620 in patients with chronic hepatitis B virus infection. J Hepatol. 2015;63:320–328. doi: 10.1016/j.jhep.2015.02.037. [DOI] [PubMed] [Google Scholar]
- 83.Khaitan A, Unutmaz D. Revisiting immune exhaustion during HIV infection. Curr HIV/AIDS Rep. 2011;8:4–11. doi: 10.1007/s11904-010-0066-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Peligero C, Argilaguet J, Guerri-Fernandez R, Torres B, Ligero C, Colomer P, Plana M, Knobel H, Garcia F, Meyerhans A. PD-L1 Blockade Differentially Impacts Regulatory T Cells from HIV-Infected Individuals Depending on Plasma Viremia. PLoS Pathog. 2015;11:e1005270. doi: 10.1371/journal.ppat.1005270. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Akhmetzyanova I, Drabczyk M, Neff CP, Gibbert K, Dietze KK, Werner T, Liu J, Chen L, Lang KS, Palmer BE, et al. PD-L1 Expression on Retrovirus-Infected Cells Mediates Immune Escape from CD8+ T Cell Killing. PLoS Pathog. 2015;11:e1005224. doi: 10.1371/journal.ppat.1005224. [DOI] [PMC free article] [PubMed] [Google Scholar]

