Abstract
Purpose of review
Despite the success of antiretroviral therapy in suppressing HIV, life-long therapy is required to avoid HIV reactivation from long-lived viral reservoirs. Currently, there is intense interest in searching for therapeutic interventions that can purge the viral reservoir to achieve complete remission in HIV patients off antiretroviral therapy. The evaluation of such interventions relies on our ability to accurately and precisely measure the true size of the viral reservoir. In this review, we assess the most commonly used HIV reservoir assays, as a clear understanding of the strengths and weaknesses of each is vital for the accurate interpretation of results and for the development of improved assays.
Recent findings
The quantification of intracellular or plasma HIV RNA or DNA levels remains the most commonly used tests for the characterization of the viral reservoir. While cost-effective and high-throughput, these assays are not able to differentiate between replication-competent or defective fractions, or quantify the number of infected cells. Viral outgrowth assays provide a lower bound for the fraction of cells that can produce infectious virus, but these assays are laborious, expensive and substantially underestimate the potential reservoir of replication-competent provirus. Newer assays are now available that seek to overcome some of these problems, including full-length proviral sequencing, inducible HIV RNA assays, ultrasensitive p24 assays and murine adoptive transfer techniques.
Summary
The development and evaluation of strategies for HIV remission relies upon our ability to accurately and precisely quantify the size of the remaining viral reservoir. At this time, all current HIV reservoir assays have drawbacks such that combinations of assays are generally needed to gain a more comprehensive view of the viral reservoir. The development of novel, rapid, high-throughput assays that can sensitively quantify the levels of the replication-competent HIV reservoir is still needed.
Keywords: HIV, reservoir, assays, QVOA, replication-competent, cure
Introduction
While antiretroviral therapy (ART) has had a dramatic impact on the HIV epidemic, it is not curative and life-long ART is needed to prevent HIV reactivation from long-lived viral reservoirs [1,2]. One of the highest priorities for the HIV field is the search for therapeutic interventions that can eliminate or control the HIV reservoir, with the hope of replicating the sustained ART-free HIV remission seen in HIV elite controllers (ECs) or post-treatment controllers (PTCs) [3–5]. However, the optimal method of measuring the HIV reservoir size remains controversial, as all current assays have strengths and weaknesses. This is reflected in the diversity of reservoir assays evaluated as part of previously completed (Table 1) and current clinical trials (Table 2) [3,6–8]. Overall, there are several categories of HIV reservoir assays. These assays include 1) intracellular HIV DNA, 2) cell-associated HIV RNA, 3) ultrasensitive plasma viremia, 4) viral outgrowth assays, 5) inducible HIV RNA, 6) protein-based assays and 7) murine adoptive transfer assay. In this review, we will summarize the most commonly used HIV reservoir assays, providing details on their interpretation, advantages and limitations.
Table 1.
Study | LRA | Total DNA | Integrated DNA | 2-LTR circles | CA-RNA | Ultrasensitive Plasma RNA | QVOA | TILDA |
---|---|---|---|---|---|---|---|---|
Gutiérrez, AIDS 2016 [51] | Bryostatin | + | + | |||||
Spivak, CID 2014 [79] | Disulfiram | + | + | |||||
Mothe, JAC 2015 [80] | Disulfiram in combination | + | + | |||||
Rasmussen, Lancet 2014 [8] | Panobinostat | + | + | + | + | + | + | |
Søgaard, PLoS Path 2015 [6] | Romidepsin | + | + | + | + | + | + | |
Leth, Lancet HIV 2016 [60] | Romidepsin in combination | + | + | + | + | + | ||
Lehrman, Lancet 2005 [41] | Valproic acid | + | + | + | ||||
Siliciano, JID 2007 [81] | Valproic acid | + | ||||||
Archin, AIDS 2008 [82] | Valproic acid | + | + | |||||
Sagot-Lerolle, AIDS 2008 [83] | Valproic acid | + | + | + | ||||
Routy, HIV Med 2012 [84] | Valproic acid | + | ||||||
Archin, Nature 2012 [3] | Vorinostat | + | + | |||||
Archin, JID 2014 [85] | Vorinostat | + | + | + | + | |||
Elliott, PLoS Path 2014 [86] | Vorinostat | + | + | + | + | + |
QVOA, quantitative viral outgrowth assay; CA-RNA, cell-associated HIV RNA; TILDA, tat/rev inducible limiting dilution assay; PKC, protein kinase C; JQ1, a bromodomain inhibitor.
Table 2.
ClinicalTrials.gov Identifier | Trial Name | Primary outcomes measured |
---|---|---|
NCT02092116 | REDUC | QVOA, total HIV DNA, integrated DNA |
NCT02707900 | VOR VAX | QVOA |
NCT02707692 | Perturbing of HIV reservoir with immune stimulation | CA-RNA |
NCT02336074 | RIVER | Total HIV DNA |
NCT02628340 | RESEACHRON | Total HIV DNA |
NCT02191098 | ALT-803 | TILDA |
NCT01933594 | A5315 | SCA and CA-RNA |
NCT02028403 | A5326 | Gag-specific CD8+ T cells and changes in SCA |
NCT02411539 | A5342 | CA-RNA/DNA ratio |
QVOA, quantitative viral outgrowth assay; CA-RNA, cell-associated HIV RNA; TILDA, tat/rev inducible limiting dilution assay; SCA, single-copy assay.
1. HIV DNA assays
Total HIV-1 DNA includes all forms of HIV-1 DNA: circular unintegrated, linear unintegrated and linear integrated. The relative abundance of each species has been reported to be in the following descending order: non-integrated linear DNA > integrated proviral DNA > non-integrated circular DNA [9,10]. Specifically in non-suppressed patients, non-integrated forms make up the vast majority of HIV DNA in the nucleus, which is approximately 100-fold more frequent than the integrated proviral DNA form [11]. Separate assays have been developed to quantify each species.
Quantitative real-time PCR (qPCR) or droplet digital PCR (ddPCR) for conserved viral regions can be performed to determine levels of HIV nucleic acid, as will be discussed below. qPCR monitors the progression of amplification in each cycle through the use of fluorescent probes, and quantification is performed by measuring the threshold cycle (Ct) at which fluorescence is higher than a certain threshold. While a standard curve is necessary for quantification of copy number by qPCR, it provides a wide dynamic range. Other caveats include susceptibility to primer/probe sequence mismatches that lead to inaccurate quantification and sensitivity challenges in detecting low copy numbers. The former can be tackled using patient-matched primers [12] or calculating the patient-specific mismatch-related quantification errors (MRQE) by comparing the amplification of patient sample with that of a control template without mismatches [13].
Another option is using the more recently described ddPCR, which measures only endpoint fluorescence after all thermal cycles are completed to provide absolute quantification for HIV DNA. Through a microfluidic system, the aqueous PCR reaction mixture is emulsified in a thermostable oil to form droplets, allowing a large number of microscopic reactions to be performed at the same time, at a level where some droplets will have no template and others may have 1 or more template copies [14,15]. After the reaction is complete, the samples are loaded into a droplet reader and each droplet is streamed in a single file past an optical detector to detect the amplitude of fluorescence from each droplet. The readout is a count of the number of positive droplets formed per sample well. Based on Poisson distribution, the data can be used to calculate the starting copy number of the target DNA template in the original sample. The dynamic range of ddPCR is determined by the number of replicates (droplets) formed in the reaction.
A recognized advantage of ddPCR over qPCR is avoiding the need for standard curves since ddPCR gives an absolute quantification. Furthermore, ddPCR may result in improved precision [14,16] and is more robust towards target sequence variation, which is especially important when working with genetically-diverse clinical samples [14]. On the other hand, ddPCR has a more limited dynamic range, demonstrates a higher risk of false positive wells and is more time consuming due to the additional time needed for droplet generation and droplet reading after the PCR is complete [14,16]. In addition, the ddPCR system and reagents are generally more costly than that for qPCR.
The two most commonly used ddPCR platforms include systems from Biorad and RainDance. The Biorad platform represented the first commercially available ddPCR system and is the most widely adopted. The RainDance platform was more recently introduced into the field [17], but may be able to yield a significantly greater number of droplets. A rigorous comparison of the two ddPCR platforms for HIV detection is still needed.
Total HIV DNA
To determine levels of total HIV DNA, qPCR or ddPCR for conserved viral regions can be performed [18]. In both of these platforms, probes for HIV DNA have targeted HIV-1 gag [18,19], pol [14] or the LTR region [20–22]. When the three different targets were compared side-to-side, targeting the LTR region was shown to be superior and resulted in some cases in a log10-higher number of HIV copies [23]. To determine the copy number of HIV-1 DNA per cell, a parallel measurement of a control gene (such as CCR5 [24], β-globin [25] or albumin [18]) is frequently conducted to quantify the number of cells assayed during the HIV DNA measurement. Alternatively, a parallel blood sample, drawn simultaneously, can be stained for CD4 and run for flow cytometric analysis [19].
In patients on suppressive antiretroviral therapy, total HIV DNA level is reflective of the total HIV proviral reservoir size and may predict viral rebound timing after treatment interruption [26,27]. However, a PCR-based approach for measuring the viral reservoir leads to an over-estimation of the size of the replication-competent reservoir, as the vast majority of viral genomes quantified are not replication-competent. It has been estimated that measuring total HIV DNA may over-estimate the intact HIV reservoir by greater than 100-fold in those treated during chronic infection and by more than 10-fold in those treated during acute infection [28].
It is important to note that unintegrated viral DNA contributes to the total HIV DNA signal, while unintegrated DNA contributes very little to the viral reservoir due to its limited transcription potential [25,29]. This is an especially important distinction when monitoring reservoir size in viremic non-suppressed patients, who have an excess of unintegrated HIV DNA that confounds the interpretation of total DNA levels [30,31].
Circular unintegrated HIV DNA
Circular unintegrated forms include 1-LTR and 2-LTR circular HIV DNA. In non-suppressed patients, non-integrated DNA makes up the majority of total DNA [30]. However, in virologically suppressed patients, circular HIV DNA forms represent a minor population and do not give rise to infectious virus. The 2-LTR circles, for example, represent approximately 0.03-5% of total viral DNA [32]. Quantification of 1-LTR circles by qPCR has been technically challenging and unreliable because they lack unique sequence segments to distinguish them from proviral or linear unintegrated DNA [33]. The presence of 2-LTR circles can be assayed by using primers specific for the junction between the two LTR ends and performing either qPCR or ddPCR [14]. Historically, 2-LTR circles have been measured as a reflection of active viral replication, but this has been called into question [34].
Integrated HIV proviral DNA
Alu-gag PCR
There are situations where measurement of only integrated proviral HIV DNA is indicated. This is especially relevant for participants who are not virologically suppressed, where linear and circular nonintegrated forms may dominate the total DNA measurement. To quantify integrated DNA alone, a nested PCR, known as Alu-gag PCR, can be performed [35,36]. The first PCR utilizes a forward primer that is virus-specific, recognizing the U5 region of the LTR, while the reverse primer binds to Alu repeats. This results in specific amplification of integrated HIV DNA. Alu elements are abundant ~300 bp-interspersed repeat sequences, distributed at a frequency of 1 Alu element in every 2.5 kb of the human genome [37]. Since the integration of HIV is random within the human genome, Alu-gag pre-amplification generates a population of cellular-HIV junction DNA sequences of various lengths. After the initial amplification round, qPCR is used to quantify total copies of integrated provirus [38]. The caveats here are that only integrated forms of the virus that reside close to Alu repeats will be reliably amplified [39] and the high variability between sample replicates. To address this, repetitive sampling has been introduced [36,39,40].
Gel separation
Running DNA samples on a gel can be used to separate genomic (approximately 20 kb) high-molecular weight (HMW) DNA from episomal DNA. HMW DNA is then recovered from the gel and subjected to HIV-specific qPCR. This has been implemented in some studies [41] and authors have reported that this fractionation procedure gets rid of 97-99% of linear HIV-1 DNA and 99% of 2-LTR circles [30]. This assay is not commonly employed, but can also determine levels of all non-integrated HIV species, where HIV unintegrated DNA equals HIV DNA from total DNA minus integrated HIV DNA from HMW DNA.
Fluorescence In-Situ Hybridization (FISH)
FISH can be used to quantify number of integrated proviral DNA copies [42] and was successfully used on splenocytes from splenectomized HIV patients [43]. That study demonstrated that the majority of infected cells in HIV patients harbor more than one proviral copy, with a mean of 3-4 proviruses per infected cell. DNAscope, an optimizied in-situ hybridization platform that relies on the use of probes spanning the entire length of the viral DNA (vDNA), was demonstrated to detect latently infected cells in lymphoid tissue sections from macaques [44]. This might represent a sensitive tool to identify sites of latent viral reservoir at the tissue level.
Full-length Single Genome Sequencing (SGS)
Defective proviruses constitute the vast majority (93-98%) of proviral HIV DNA [28]. To better estimate the true size of intact proviral reservoir, methods to perform full-length single-genome proviral sequencing are becoming increasingly popular. Genomic DNA is extracted from patient cells and subjected to a limiting dilution PCR protocol using primers that span the two LTRs to amplify near-full length HIV proviral DNA from single templates [28,45,46]. Sanger or next-generation sequencing is used to sequence the amplified proviruses. These methods are labor- and cost-intensive, but can provide important details on the types of defective proviral genomes present, in addition to quantifying the number of intact proviruses. Results from these studies have demonstrated that qPCR/ddPCR quantification of HIV proviral DNA significantly overestimates the size of the intact HIV reservoir.
2. Cell-associated HIV RNA assays
Within HIV-infected cells, several forms of RNA exist: multiply spliced (ms), incompletely spliced (is) and unspliced (us) [47]. Initially, msRNA transcripts are generated, encoding for regulatory proteins, such as tat and rev. As the infection proceeds, there is a shift towards isRNA and usRNA, encoding for the full viral genome to be packaged, as well as structural and accessory viral proteins. Assays that measure unspliced HIV RNA have been the most commonly used platform for previous clinical studies, as illustrated for studies determining the effect of HIV latency-reversing agents (Table 1).
Total HIV RNA transcripts can be isolated and quantified by RT-qPCR or ddPCR, using primers and probes targeting the LTR region [20–22]. To assay usRNA species, probe and primers targeting viral gag, located downstream of the major 5′ splice donor site (D1), are used. To target the msRNA species, probe and primers encoding for tat and rev are used [48]. We have reported that levels of cell-associated (CA)-usRNA are predictive of the timing of viral rebound after treatment interruption [49].
HIV RNA copy numbers are normalized to cellular input, either by running a parallel qPCR for a control gene (such as β-actin [50] or CCR5 [51]) or estimated by total extracted RNA amounts, assuming that 1 ng RNA corresponds to approximately 1000 cells [51]. Some groups also report the average transcription per infected cell, calculated as the CA-HIV RNA/DNA ratio [21]. One major limitation of CA-RNA quantification methods is that defective CA-RNA species are also present, similar to HIV DNA [52] and thus RT-qPCR likely overestimates the number of intact HIV transcripts.
Another recently described technique is the PrimeFlow RNA Assay, which is based on fluorescence in-situ RNA hybridization using a set of 20–40 probes. Simultaneous use of PrimeFlow and p24 protein staining via flow cytometry was successfully reported in cell lines and in vitro infected cells [53]. The PrimeFlow assay is reported to detect different classes of HIV RNA transcripts with a limit of detection (LOD) of 10–100 infected cells within 106 total cells [54]. Furthermore, a recently published paper used this dual technique to detect and characterize infected CD4 T cells from the peripheral blood of both suppressed and non-suppressed patients [55]. The assay can also be used with chromogenic detection, allowing microscopic imaging of viral RNA (vRNA) positive cells, termed RNAscope. vRNA and vDNA double positive cells, as well as vDNA positive vRNA negative cells, were identified in lymphoid tissue sections from macaques using this technique [44]. A potential limitation of this assay is the requirement of large volumes of blood or difficult-to-sample tissue sections.
3. Ultrasensitive Plasma Residual Viremia assays
Single copy assay (SCA)
To characterize residual plasma viremia, several assays were developed to detect a single copy or less of HIV-1 RNA per ml of plasma. The SCA assay consists of an initial nucleic acid extraction through pelleting of virions by ultracentrifugation. To monitor this step of viral RNA recovery from the plasma and control for possible reverse transcriptase qPCR (RT-qPCR) inhibition, samples are spiked with an internal control standard, a known quantity of replication competent avian sarcoma (RCAS) virions. Following a proteinase K digestion of proteins, nucleic acids are precipitated. HIV-1 RNA is quantified via an RT-qPCR reaction using primers and probe targeting either the gag (gSCA) or integrase (iSCA) regions [56]. The gSCA was the original version of the SCA, but inefficient amplification of viral RNA in 15-30% of clinical plasma samples led to the need for initial confirmation of primer/probe efficiency for each participant sample [57]. The newer iSCA assay uses primers and probe targeting the more conserved integrase region and with significantly decreased rates of primer/probe sequence mismatches with patient samples [57]. For a more sensitive assay, a modification to iSCA, called mega-iSCA, was developed, which assays large volumes of plasma (20-35 ml) with a LOD down to <0.1 copies/ml.
The HIV Molecular and Monitoring Core gag assay (HMMCgag) is an assay developed at NCI Frederick and represents a variant of the SCA, using primers and probe targeting the beginning of the gag gene. The assay’s design relies on a hybrid of qPCR and digital PCR format to maximize the advantages and minimize the disadvantages of both assay formats. The assay is set up in multiple replicates and for samples where all reactions are positive for amplification; viral load is calculated based on the standard curve. But for samples for which some reactions are not positive for amplification, the viral load is instead calculated based on the Poisson distribution for the frequency of positive wells [58].
Transcription mediated amplification (TMA)
TMA is a qualitative nucleic acid amplification test, commonly used in blood donor screening [59], with a binary positive or negative result [6,8,51,60]. The assay is commercially available through Procleix Ultrio Plus and is widely used in Europe. First, viral RNA is captured by oligonucleotides containing a T7 promoter primer site, which is subsequently captured by magnetic particles to allow for the separation of viral RNA. This is followed by an isothermal transcription-mediated amplification step using reverse transcriptase and T7 RNA polymerase leading to the exponential production of RNA amplicons. Finally, the amplicons are detected via a fluorescent probe. When run in a single replicate, the assay has a sensitivity of 3.6 HIV copies/ml at a 50% limit of detection [61]. When the assay is performed in quadruplicate, the sensitivity drops down to less than 3.5 HIV copies/ml [62].
Modified Abbott qPCR assay for low-copy detection
The conventional Abbott qPCR assay is an automated platform to measure HIV-1 viral load in plasma samples using volumes up to 1 mL. The assay utilizes primers and probe targeting the integrase region of pol, with a unique probe design, being partially double-stranded and tolerating mismatches. Thus the assay can be used for quantifying HIV-1 group M (subtype A-H), group N and group O isolates [63]. The LOD for the conventional assay is 40 copies/ml. The modified version of this assay relies on concentrating virus from a high volume of plasma (up to 30 ml) by ultracentrifugation on an iodixanol density cushion. The concentrated pellet is resuspended and HIV-1 RNA is quantified using the Abbott HIV-1 qPCR assay with a reported LOD of less than 1 copy/ml [64].
4. Quantitative viral outgrowth assay (QVOA)
While nucleic acid-based measures of HIV reservoir size have some advantages, they are unable to quantify the size of the replication-competent reservoir. QVOA, or the Infectious Units Per Million (IUPM) assay, has historically been the gold standard for detection of the replication-competent HIV reservoir [65,66]. Using a limiting dilution culture format, QVOA measures the number of wells containing detectable HIV-derived p24 antigen released in the supernatant after resting cells are subjected to one round of stimulation [65,66]. Previous reports show that QVOA is relatively robust, and with sufficient cell numbers, HIV-1 can be recovered from the majority of HIV-1 infected participants on suppressive antiretroviral therapy [2,65,67,68]. QVOA was used to demonstrated the high stability and low decay rate of the latent virus reservoir in suppressed patients [68], showing generally less than 2-fold variation between longitudinal measurements [69].
The main advantage of this assay is that it detects only the replication-competent virus reservoir. However, QVOA underestimates the reservoir size because at any given time, only a subset of the replication-competent reservoir is activated. Studies have calculated that only 1% of cells harboring HIV provirus release infectious virions after being subjected to maximum in vitro activation [46]. A large proportion of those non-induced cells harbor defective copies of the integrated HIV provirus, but a subset represents a population of cells with intact virus, which are not activated in any single round of activation. Some reports demonstrated that QVOA did not strongly correlate with the frequency of cells harboring intact proviruses and likely underestimates the total replication-competent virus reservoir by approximately 25-fold [28,46]. Thus, QVOA should be thought of as the lower-bound estimate of the replication-competent reservoir. QVOA has not been useful in in vivo studies measuring effect of latency reversal agents (LRAs) on the size of virus reservoir, which highlights its limited sensitivity [6,8]. It has been suggested that a more than 6-fold difference between longitudinal QVOA measurements could be used to reliably detect a change in reservoir size with high confidence [69]. Other limitations of QVOA include the requirement of a large sample volume and that it is both time- and resource-intensive. Of note, integrated HIV DNA levels has been found to be significantly correlated with QVOA [70].
5. Inducible HIV RNA assays
The quantification of the replication-competent HIV reservoir is challenging due to the overwhelming proportion of infected cells that harbor replication-deficient proviruses and the low frequency of transcriptionally-active cells in those on chronic suppressive ART. The inducible HIV RNA assays seek to bridge the divide between the nucleic acid-based measurements of the HIV reservoir (CA-DNA or CA-RNA) and QVOA. They provide a more accurate reading of the inducible HIV reservoir than the CA-RNA assay alone while potentially providing the frequency of HIV-expressing cells in an assay that is far more rapid and scalable than QVOA. However, replication-defective proviruses may lead to RNA transcripts [52] and one limitation of all assays measuring HIV RNA levels is the inability to fully define the replication-competent fraction.
CA-RNA
This assay measures the copy number of cell-associated viral RNA after stimulation. Briefly, CD4 T cells are stimulated, followed by cell lysis and RNA extraction. Levels of usRNA and msRNA can be measured using RT-qPCR [71,72]. The extent of HIV-1 transcription after activation is based on levels of msRNA, like rev and tat. A limitation of this assay is the requirement of RNA extraction, where potential loss of viral RNA may occur. This was addressed in the newer TILDA assay, which will be discussed below.
Supernatant RNA
This assay relies on measuring the production of viral particles in culture supernatants of stimulated cells. CD4 T cells isolated from patients are stimulated (using PHA [73], CD3/CD28 beads [74] or latency reversal agents [74]), releasing infectious virions in the supernatant. At different time points post-stimulation, viral RNA is isolated from the cellular supernatant using a commercial RNA isolation kit and levels of HIV RNA are assayed using RT-qPCR. Similar to the inducible CA-RNA assay, the limitation of this assay is that it requires an RNA extraction step.
Tat/rev Induced Limiting Dilution Assay (TILDA)
TILDA is an assay that measures the frequency of cells with inducible HIV msRNA [71,75]. Briefly, about 1 million cells CD4 T cells are stimulated with PMA and ionomycin for 12 hours, a time-point chosen based on kinetic studies demonstrating maximal RNA production of tat and rev. After distribution in a limiting dilution format directly in the reaction buffer, samples are subjected to RT-qPCR for tat and rev transcripts. Tat/rev were observed to be frequently missing in defective proviruses with internal deletions. Thus this assay is attractive as it is less likely to be measuring defective RNA species [28]. Using the maximum likelihood method, the frequency of cells producing HIV msRNA post-stimulation can be calculated based on the number of positive wells [76]. Results from TILDA correlated with those obtained through measuring integrated viral DNA. The TILDA assay is less sample-intensive than QVOA and does not involve an RNA extraction step or amplification of virus replication, avoiding the need for a prolonged culture time [76]. However, it is important to note that not all cells producing msRNA are releasing intact infectious virions. Compared to nucleic acid-quantification assays, this technique is more resource-intensive.
6. Protein-based assays
There are few high-throughput and sensitive assays for detecting HIV protein production. Investigators at Merck recently reported the development of an ultra-sensitive immunoassay to quantify p24 and the uncleaved p55 levels in cell lysates and in media from cultured patient cells. It is reported that the assay can detect protein levels down to 14 fg/ml with a dynamic range of >4 log10, but does require the use of specialized equipment (Quanterix Simoa technology) [77].
7. Murine viral outgrowth assay (MVOA)
The murine viral outgrowth assay (MVOA) is a binary end-point assay that uses a mouse model to determine whether patient-derived cells harbor infectious virus [78]. In this assay, either whole PBMCs or sorted CD4 T cells are injected into NOD/Prkdcscid/gamma-chain knockout (NSG) mice. Some mice are further subjected to CD8 T cell depletion or subjected to T cell stimulation via injection of an anti-CD3 antibody. Over time, HIV RNA from the plasma of xenografted mice is isolated and quantified by RT-qPCR. The assay was successful in recovering virus from patient cells, including an elite controller, who had negative QVOA results [78]. It can be used to survey a large number of patient cells, requiring 1 mouse per 10-50 million CD4 T cells. However it suffers from drawbacks related to the inherent heterogeneity of human cell engraftment in the murine host and the lack of a quantitative readout.
Conclusion
The development and evaluation of HIV curative strategies relies upon our ability to accurately and precisely quantify the size of the remaining HIV reservoir. At this time, all current HIV reservoir assays have drawbacks such that combinations of assays are generally needed to gain a more comprehensive view of the HIV reservoir. Techniques that quantify levels of HIV cell-associated DNA are high-throughput, but significantly over-estimate the size of the intact, or true viral reservoir. While the QVOA assay has historically been considered the gold-standard for measuring the size of the replication-competent reservoir, this assay is challenging to perform and is useful only for determining the lower bound for the size of the replication-competent reservoir. Quantifying the number of intact proviruses by sequencing appears to provide the best current estimate of the HIV reservoir’s potential true size, but this assay is still relatively new, and is both labor-intensive and expensive, calling into question its scalability in large clinical studies. Newer assays for the single-cell measurement of HIV-expressing cells and the high-throughput quantification of HIV protein levels represent promising technologies, but still require additional validation. The development of a rapid, high-throughput assay that can sensitively quantify the levels of the replication-competent HIV reservoir remains the holy grail of HIV reservoir assays and would accelerate the journey to our ultimate goal of finding an effective HIV curative strategy.
Acknowledgments
Conflict of Interest
JL has received research funding and consulted for Gilead and Merck and was funded in part by a grant from NIH/NIAID (125109).
Footnotes
Compliance with Ethics Guidelines
There were no human or animal experiments performed for the purpose of this review.
References
Papers of particular interest have been highlighted as**
- 1.Chun TW, Fauci aS. Latent reservoirs of HIV: obstacles to the eradication of virus. Proc Natl Acad Sci U S A. 1999;96:10958–61. doi: 10.1073/pnas.96.20.10958. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Finzi D, Blankson J, Siliciano JD, Margolick JB, Chadwick K, Pierson T, Smith K, Lisziewicz J, Lori F, Flexner C, Quinn TC, Chaisson RE, Rosenberg E, Walker B, Gange S, Gallant J, Siliciano RF. 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;5:512–7. doi: 10.1038/8394. [DOI] [PubMed] [Google Scholar]
- 3.Archin NM, Liberty AL, Kashuba AD, Choudhary SK, Kuruc JD, Crooks AM, Parker DC, Anderson EM, Kearney MF, Strain MC, Richman DD, Hudgens MG, Bosch RJ, Coffin JM, Eron JJ, Hazuda DJ, Margolis DM. Administration of vorinostat disrupts HIV-1 latency in patients on antiretroviral therapy. Nature. 2012;487:482–5. doi: 10.1038/nature11286. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Buckheit RW, Salgado M, Martins KO, Blankson JN. The implications of viral reservoirs on the elite control of HIV-1 infection. Cell Mol Life Sci. 2013;70:1009–19. doi: 10.1007/s00018-012-1101-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Sáez-Cirión A, Bacchus C, Hocqueloux L, Avettand-Fenoel V, Girault I, Lecuroux C, Potard V, Versmisse P, Melard A, Prazuck T, Descours B, Guergnon J, Viard J-P, Boufassa F, Lambotte O, Goujard C, Meyer L, Costagliola D, Venet A, Pancino G, Autran B, Rouzioux C, ANRS VISCONTI Study Group 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]
- 6.Søgaard OS, Graversen ME, Leth S, Olesen R, Brinkmann CR, Nissen SK, Kjaer AS, Schleimann MH, Denton PW, Hey-Cunningham WJ, Koelsch KK, Pantaleo G, Krogsgaard K, Sommerfelt M, Fromentin R, Chomont N, Rasmussen TA, Østergaard L, Tolstrup M. The Depsipeptide Romidepsin Reverses HIV-1 Latency In Vivo. PLoS Pathog. 2015;11:e1005142. doi: 10.1371/journal.ppat.1005142. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Delagrèverie HM, Delaugerre C, Lewin SR, Deeks SG, Li JZ. Ongoing Clinical Trials of Human Immunodeficiency Virus Latency-Reversing and Immunomodulatory Agents. Open Forum Infect Dis. 2016;3:ofw189. doi: 10.1093/ofid/ofw189. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Rasmussen TA, Tolstrup M, Brinkmann CR, Olesen R, Erikstrup C, Solomon A, Winckelmann A, Palmer S, Dinarello C, Buzon M, Lichterfeld M, Lewin SR, Ostergaard L, Søgaard OS. Panobinostat, a histone deacetylase inhibitor, for latent virus reactivation in HIV-infected patients on suppressive antiretroviral therapy: A phase 1/2, single group, clinical trial. Lancet HIV. 2014;1:e13–21. doi: 10.1016/S2352-3018(14)70014-1. [DOI] [PubMed] [Google Scholar]
- 9.Vandegraaff N, Kumar R, Burrell CJ. Kinetics of Human Immunodeficiency Virus Type 1 (HIV) DNA Integration in Acutely Infected Cells as Determined Using a Novel Assay for Detection of Integrated HIV DNA. J Virol. 2001;1:11253–60. doi: 10.1128/JVI.75.22.11253-11260.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Butler SL, Hansen MS, Bushman FD. A quantitative assay for HIV DNA integration in vivo. Nat Med. 2001;7:631–4. doi: 10.1038/87979. [DOI] [PubMed] [Google Scholar]
- 11.Chun TW, Carruth L, Finzi D, Shen X, DiGiuseppe JA, Taylor H, Hermankova M, Chadwick K, Margolick J, Quinn TC, Kuo YH, Brookmeyer R, Zeiger MA, Barditch-Crovo P, Siliciano RF. Quantification of latent tissue reservoirs and total body viral load in HIV-1 infection. Nature. 1997;387:183–8. doi: 10.1038/387183a0. [DOI] [PubMed] [Google Scholar]
- 12.Kaiser P, Joos B, Niederöst B, Weber R, Günthard HF, Fischer M. Productive human immunodeficiency virus type 1 infection in peripheral blood predominantly takes place in CD4/CD8 double-negative T lymphocytes. J Virol. 2007;81:9693–706. doi: 10.1128/JVI.00492-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Pasternak AO, Jurriaans S, Bakker M, Prins JM, Berkhout B, Lukashov VV. Cellular levels of HIV unspliced RNA from patients on combination antiretroviral therapy with undetectable plasma viremia predict the therapy outcome. PLoS One. 2009;4:e8490. doi: 10.1371/journal.pone.0008490. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Strain MC, Lada SM, Luong T, Rought SE, Gianella S, Terry VH, Spina CA, Woelk CH, Richman DD. Highly precise measurement of HIV DNA by droplet digital PCR. PLoS One. 2013;8:e55943. doi: 10.1371/journal.pone.0055943. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Hindson BJ, Ness KD, Masquelier DA, Belgrader P, Heredia NJ, Makarewicz AJ, Bright IJ, Lucero MY, Hiddessen AL, Legler TC, Kitano TK, Hodel MR, Petersen JF, Wyatt PW, Steenblock ER, Shah PH, Bousse LJ, Troup CB, Mellen JC, Wittmann DK, Erndt NG, Cauley TH, Koehler RT, So AP, Dube S, Rose KA, Montesclaros L, Wang S, Stumbo DP, Hodges SP, Romine S, Milanovich FP, White HE, Regan JF, Karlin-Neumann GA, Hindson CM, Saxonov S, Colston BW. High-throughput droplet digital PCR system for absolute quantitation of DNA copy number. Anal Chem. 2011;83:8604–10. doi: 10.1021/ac202028g. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Bosman KJ, Nijhuis M, van Ham PM, Wensing AMJ, Vervisch K, Vandekerckhove L, De Spiegelaere W. Comparison of digital PCR platforms and semi-nested qPCR as a tool to determine the size of the HIV reservoir. Sci Rep. 2015;5:13811. doi: 10.1038/srep13811. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Abdel-Mohsen M, Chavez L, Tandon R, Chew GM, Deng X, Danesh A, Keating S, Lanteri M, Samuels ML, Hoh R, Sacha JB, Norris PJ, Niki T, Shikuma CM, Hirashima M, Deeks SG, Ndhlovu LC, Pillai SK. Human Galectin-9 Is a Potent Mediator of HIV Transcription and Reactivation. PLoS Pathog. 2016;12:e1005677. doi: 10.1371/journal.ppat.1005677. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Klatt NR, Bosinger SE, Peck M, Richert-Spuhler LE, Heigele A, Gile JP, Patel N, Taaffe J, Julg B, Camerini D, Torti C, Martin JN, Deeks SG, Sinclair E, Hecht FM, Lederman MM, Paiardini M, Kirchhoff F, Brenchley JM, Hunt PW, Silvestri G. Limited HIV infection of central memory and stem cell memory CD4+ T cells is associated with lack of progression in viremic individuals. PLoS Pathog. 2014;10:e1004345. doi: 10.1371/journal.ppat.1004345. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Nottet HSLM, van Dijk SJ, Fanoy EB, Goedegebuure IW, de Jong D, Vrisekoop N, van Baarle D, Boltz V, Palmer S, Borleffs JCC, Boucher CaB. HIV-1 can persist in aged memory CD4+ T lymphocytes with minimal signs of evolution after 8.3 years of effective highly active antiretroviral therapy. J Acquir Immune Defic Syndr. 2009;50:345–53. doi: 10.1097/QAI.0b013e318197eb04. [DOI] [PubMed] [Google Scholar]
- 20.Yukl SA, Sinclair E, Somsouk M, Hunt PW, Epling L, Killian M, Girling V, Li P, Havlir DV, Deeks SG, Wong JK, Hatano H. A comparison of methods for measuring rectal HIV levels suggests that HIV DNA resides in cells other than CD4+ T cells, including myeloid cells. AIDS. 2014;28:439–42. doi: 10.1097/QAD.0000000000000166. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Yukl SA, Shergill AK, Ho T, Killian M, Girling V, Epling L, Li P, Wong LK, Crouch P, Deeks SG, Havlir DV, McQuaid K, Sinclair E, Wong JK. The distribution of HIV DNA and RNA in cell subsets differs in gut and blood of HIV-positive patients on ART: implications for viral persistence. J Infect Dis. 2013;208:1212–20. doi: 10.1093/infdis/jit308. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Hatano H, Somsouk M, Sinclair E, Harvill K, Gilman L, Cohen M, Hoh R, Hunt PW, Martin JN, Wong JK, Deeks SG, Yukl SA. Comparison of HIV DNA and RNA in gut-associated lymphoid tissue of HIV-infected controllers and noncontrollers. AIDS. 2013;27:2255–60. doi: 10.1097/QAD.0b013e328362692f. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Rozera G, Abbate I, Bruselles A, Bartolini B, D’Offizi G, Nicastri E, Tommasi C, Capobianchi MR. Comparison of real-time PCR methods for measurement of HIV-1 proviral DNA. J Virol Methods. 2010;164:135–8. doi: 10.1016/j.jviromet.2009.11.031. [DOI] [PubMed] [Google Scholar]
- 24.Malnati MS, Scarlatti G, Gatto F, Salvatori F, Cassina G, Rutigliano T, Volpi R, Lusso P. A universal real-time PCR assay for the quantification of group-M HIV-1 proviral load. Nat Protoc. 2008;3:1240–8. doi: 10.1038/nprot.2008.108. [DOI] [PubMed] [Google Scholar]
- 25.Brussel A, Sonigo P. Evidence for Gene Expression by Unintegrated Human Immunodeficiency Virus Type 1 DNA Species. J Virol. 2004;78:11263–71. doi: 10.1128/JVI.78.20.11263-11271.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Yerly S, Gunthard HF, Fagard C, Joos B, Perneger TV, Hirschel B, Perrin L. Proviral HIV-DNA predicts viral rebound and viral setpoint after structured treatment interruptions. Aids. 2004;18:1951–3. doi: 10.1097/00002030-200409240-00011. [DOI] [PubMed] [Google Scholar]
- 27.Williams JP, Hurst J, Stöhr W, Robinson N, Brown H, Fisher M, Kinloch S, Cooper D, Schechter M, Tambussi G, Fidler S, Carrington M, Babiker A, Weber J, Koelsch KK, Kelleher AD, Phillips RE, Frater J. HIV-1 DNA predicts disease progression and post-treatment virological control. Elife. 2014;3:e03821. doi: 10.7554/eLife.03821. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Bruner KM, Murray AJ, Pollack RA, Soliman MG, Laskey SB, Capoferri AA, Lai J, Strain MC, Lada SM, Hoh R, Ho Y-C, Richman DD, Deeks SG, Siliciano JD, Siliciano RF. Defective proviruses rapidly accumulate during acute HIV-1 infection. Nat Med. 2016;22:1043–9. doi: 10.1038/nm.4156. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Wu Y, Marsh JW. Gene transcription in HIV infection. Microbes Infect. 2003;5:1023–7. doi: 10.1016/s1286-4579(03)00187-4. [DOI] [PubMed] [Google Scholar]
- 30.Koelsch KK, Liu L, Haubrich R, May S, Havlir D, Günthard HF, Ignacio CC, Campos-Soto P, Little SJ, Shafer R, Robbins GK, D’Aquila RT, Kawano Y, Young K, Dao P, Spina CA, Richman DD, Wong JK. Dynamics of total, linear nonintegrated, and integrated HIV-1 DNA in vivo and in vitro. J Infect Dis. 2008;197:411–9. doi: 10.1086/525283. [DOI] [PubMed] [Google Scholar]
- 31.Besson GJ, Lalama CM, Bosch RJ, Gandhi RT, Bedison MA, Aga E, Riddler SA, McMahon DK, Hong F, Mellors JW. HIV-1 DNA decay dynamics in blood during more than a decade of suppressive antiretroviral therapy. Clin Infect Dis. 2014;59:1312–21. doi: 10.1093/cid/ciu585. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Iyer SR, Yu D, Biancotto A, Margolis LB, Wu Y. Measurement of human immunodeficiency virus type 1 preintegration transcription by using Rev-dependent Rev-CEM cells reveals a sizable transcribing DNA population comparable to that from proviral templates. J Virol. 2009;83:8662–73. doi: 10.1128/JVI.00874-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Yoder KE, Fishel R. PCR-based detection is unable to consistently distinguish HIV 1LTR circles. J Virol Methods. 2006;138:201–6. doi: 10.1016/j.jviromet.2006.07.022. [DOI] [PubMed] [Google Scholar]
- 34.Pace MJ, Graf EH, O’Doherty U. HIV 2-long terminal repeat circular DNA is stable in primary CD4+T Cells. Virology. 2013;441:18–21. doi: 10.1016/j.virol.2013.02.028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Chomont N, El-Far M, Ancuta P, Trautmann L, Procopio FA, Yassine-Diab B, Boucher G, Boulassel M-R, Ghattas G, Brenchley JM, Schacker TW, Hill BJ, Douek DC, Routy J-P, Haddad EK, Sékaly R-P. 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]
- 36.De Spiegelaere W, Malatinkova E, Lynch L, Van Nieuwerburgh F, Messiaen P, O’Doherty U, Vandekerckhove L. Quantification of integrated HIV DNA by repetitive-sampling Alu-HIV PCR on the basis of poisson statistics. Clin Chem. 2014;60:886–95. doi: 10.1373/clinchem.2013.219378. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Grover D, Mukerji M, Bhatnagar P, Kannan K, Samir K, Brahmachari SK. Alu repeat analysis in the complete human genome: Trends and variations with respect to genomic composition. Bioinformatics. 2004;20:813–7. doi: 10.1093/bioinformatics/bth005. [DOI] [PubMed] [Google Scholar]
- 38.Mbisa JL, Delviks-Frankenberry KA, Thomas JA, Gorelick RJ, Pathak VK. Real-time PCR analysis of HIV-1 replication post-entry events. Methods Mol Biol. 2009;485:55–72. doi: 10.1007/978-1-59745-170-3_5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Agosto LM, Yu JJ, Dai J, Kaletsky R, Monie D, O’Doherty U. HIV-1 integrates into resting CD4+ T cells even at low inoculums as demonstrated with an improved assay for HIV-1 integration. Virology. 2007;368:60–72. doi: 10.1016/j.virol.2007.06.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Liszewski MK, Yu JJ, O’Doherty U. Detecting HIV-1 integration by repetitive-sampling Alu-gag PCR. Methods. 2009;47:254–60. doi: 10.1016/j.ymeth.2009.01.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Lehrman G, Hogue IB, Palmer S, Jennings C, Spina CA, Wiegand A, Landay AL, Coombs RW, Richman DD, Mellors JW, Coffin JM, Bosch RJ, Margolis DM. Depletion of latent HIV-1 infection in vivo: a proof-of-concept study. Lancet. 2005;366:549–55. doi: 10.1016/S0140-6736(05)67098-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Suspène R, Meyerhans A. Quantification of unintegrated HIV-1 DNA at the single cell level in vivo. PLoS One. 2012;7:e36246. doi: 10.1371/journal.pone.0036246. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Jung A, Maier R, Vartanian J-P, Bocharov G, Jung V, Fischer U, Meese E, Wain-Hobson S, Meyerhans A. Recombination: Multiply infected spleen cells in HIV patients. Nature. 2002;418:144. doi: 10.1038/418144a. [DOI] [PubMed] [Google Scholar]
- 44.Deleage C, Wietgrefe SW, Del Prete G, Morcock DR, Hao XP, Piatak M, Bess J, Anderson JL, Perkey KE, Reilly C, McCune JM, Haase AT, Lifson JD, Schacker TW, Estes JD. Defining HIV and SIV Reservoirs in Lymphoid Tissues. Pathog Immun. 2016;1:68–106. doi: 10.20411/pai.v1i1.100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Li B, Gladden AD, Altfeld M, Kaldor JM, Cooper DA, Kelleher AD, Allen TM. Rapid reversion of sequence polymorphisms dominates early human immunodeficiency virus type 1 evolution. J Virol. 2007;81:193–201. doi: 10.1128/JVI.01231-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Ho Y-C, Shan L, Hosmane NN, Wang J, Laskey SB, Rosenbloom DIS, Lai J, Blankson JN, Siliciano JD, Siliciano RF. Replication-Competent Noninduced Proviruses in the Latent Reservoir Increase Barrier to HIV-1 Cure. Cell. 2013;155:540–51. doi: 10.1016/j.cell.2013.09.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Purcell DF, Martin MA. Alternative splicing of human immunodeficiency virus type 1 mRNA modulates viral protein expression, replication, and infectivity. J Virol. 1993;67:6365–78. doi: 10.1128/jvi.67.11.6365-6378.1993. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Pasternak AO, Adema KW, Bakker M, Jurriaans S, Berkhout B, Cornelissen M, Lukashov VV. Highly sensitive methods based on seminested real-time reverse transcription-PCR for quantitation of human immunodeficiency virus type 1 unspliced and multiply spliced RNA and proviral DNA. J Clin Microbiol. 2008;46:2206–11. doi: 10.1128/JCM.00055-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Li JZ, Etemad B, Ahmed H, Aga E, Bosch RJ, Mellors JW, Kuritzkes DR, Lederman MM, Para M, Gandhi RT. The size of the expressed HIV reservoir predicts timing of viral rebound after treatment interruption. AIDS. 2016;30:343–53. doi: 10.1097/QAD.0000000000000953. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Ledderose C, Heyn J, Limbeck E, Kreth S. Selection of reliable reference genes for quantitative real-time PCR in human T cells and neutrophils. BMC Res Notes. 2011;4:427. doi: 10.1186/1756-0500-4-427. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Gutiérrez C, Serrano-Villar S, Madrid-Elena N, Pérez-Elías MJ, Martín ME, Barbas C, Ruipérez J, Muñoz E, Muñoz-Fernández MA, Castor T, Moreno S. Bryostatin-1 for latent virus reactivation in HIV-infected patients on antiretroviral therapy. AIDS. 2016;30:1385–92. doi: 10.1097/QAD.0000000000001064. [DOI] [PubMed] [Google Scholar]
- 52.Kearney MF, Wiegand A, Shao W, Coffin JM, Mellors JW, Lederman M, Gandhi RT, Keele BF, Li JZ. Origin of Rebound Plasma HIV Includes Cells with Identical Proviruses That Are Transcriptionally Active before Stopping of Antiretroviral Therapy. J Virol. 2016;90:1369–76. doi: 10.1128/JVI.02139-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Martrus G, Niehrs A, Cornelis R, Rechtien A, García-Beltran W, Lütgehetmann M, Hoffmann C, Altfeld M. Kinetics of HIV-1 Latency Reversal Quantified on the Single-Cell Level Using a Novel Flow-Based Technique. J Virol. 2016;90:9018–28. doi: 10.1128/JVI.01448-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Romerio F, Zapata J. Detection and enrichment to near purity of rare HIV-1 infected cells by PrimeFlow RNA. J Virus Erad. 2015;(Suppl 1):1–18. Oral Presentation 3.2. [Google Scholar]
- 55.Baxter AE, Niessl J, Fromentin R, Richard J, Porichis F, Charlebois R, Massanella M, Brassard N, Alsahafi N, Delgado G-G, Routy J-P, Walker BD, Finzi A, Chomont N, Kaufmann DE. Single-Cell Characterization of Viral Translation-Competent Reservoirs in HIV-Infected Individuals. Cell Host Microbe. 2016;20:368–80. doi: 10.1016/j.chom.2016.07.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Palmer S, Wiegand AP, Maldarelli F, Mican JM, Polis M, Dewar RL, Planta A, Liu S, Metcalf Ja, John W, Coffin JM, Bazmi H, Mellors JW. New Real-Time Reverse Transcriptase-Initiated PCR Assay with Single-Copy Sensitivity for Human Immunodeficiency Virus Type 1 RNA in Plasma New Real-Time Reverse Transcriptase-Initiated PCR Assay with Single-Copy Sensitivity for Human Immunodeficiency Viru. J Clin Microbiol. 2003;41:4561–4536. doi: 10.1128/JCM.41.10.4531-4536.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Cillo AR, Vagratian D, Bedison MA, Anderson EM, Kearney MF, Fyne E, Koontz D, Coffin JM, Piatak M, Mellors JW. Improved single-copy assays for quantification of persistent HIV-1 viremia in patients on suppressive antiretroviral therapy. J Clin Microbiol. 2014;52:3944–51. doi: 10.1128/JCM.02060-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Somsouk M, Dunham RM, Cohen M, Albright R, Abdel-Mohsen M, Liegler T, Lifson J, Piatak M, Gorelick R, Huang Y, Wu Y, Hsue PY, Martin JN, Deeks SG, McCune JM, Hunt PW. The immunologic effects of mesalamine in treated HIV-infected individuals with incomplete CD4+ T cell recovery: A randomized crossover trial. PLoS One. 2014;9:1–19. doi: 10.1371/journal.pone.0116306. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Stramer SL, Krysztof DE, Brodsky JP, Fickett TA, Reynolds B, Dodd RY, Kleinman SH. Comparative analysis of triplex nucleic acid test assays in United States blood donors. Transfusion. 2013;53:2525–37. doi: 10.1111/trf.12178. [DOI] [PubMed] [Google Scholar]
- 60.Leth S, Schleimann MH, Nissen SK, Højen JF, Olesen R, Graversen ME, Jørgensen S, Kjær AS, Denton PW, Mørk A, Sommerfelt MA, Krogsgaard K, Østergaard L, Rasmussen TA, Tolstrup M, Søgaard OS. Combined effect of Vacc-4×, 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;3018:1–10. doi: 10.1016/S2352-3018(16)30055-8. [DOI] [PubMed] [Google Scholar]
- 61.Busch MP, Glynn SA, Wright DJ, Hirschkorn D, Laycock ME, McAuley J, Tu Y, Giachetti C, Gallarda J, Heitman J, Kleinman SH. Relative sensitivities of licensed nucleic acid amplification tests for detection of viremia in early human immunodeficiency virus and hepatitis C virus infection. Transfusion. 2005;45:1853–63. doi: 10.1111/j.1537-2995.2005.00649.x. [DOI] [PubMed] [Google Scholar]
- 62.Hatano H, Delwart EL, Norris PJ, Lee T-H, Dunn-Williams J, Hunt PW, Hoh R, Stramer SL, Linnen JM, McCune JM, Martin JN, Busch MP, Deeks SG. Evidence for persistent low-level viremia in individuals who control human immunodeficiency virus in the absence of antiretroviral therapy. J Virol. 2009;83:329–35. doi: 10.1128/JVI.01763-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Tang N, Huang S, Salituro J, Mak WB, Cloherty G, Johanson J, Li YH, Schneider G, Robinson J, Hackett J, Swanson P, Abravaya K. A RealTime HIV-1 viral load assay for automated quantitation of HIV-1 RNA in genetically diverse group M subtypes A-H, group O and group N samples. J Virol Methods. 2007;146:236–45. doi: 10.1016/j.jviromet.2007.07.003. [DOI] [PubMed] [Google Scholar]
- 64.Yukl SA, Li P, Fujimoto K, Lampiris H, Lu CM, Hare CB, Deeks SG, Liegler T, Pandori M, Havlir DV, Wong JK. Modification of the Abbott RealTime assay for detection of HIV-1 plasma RNA viral loads less than one copy per milliliter. J Virol Methods. 2011;175:261–5. doi: 10.1016/j.jviromet.2011.04.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Finzi D, Hermankova M, Pierson T, Carruth LM, Buck C, Chaisson RE, Quinn TC, Chadwick K, Margolick J, Brookmeyer R, Gallant J, Markowitz M, Ho DD, Richman DD, Siliciano RF. Identification of a reservoir for HIV-1 in patients on highly active antiretroviral therapy. Science. 1997;278:1295–300. doi: 10.1126/science.278.5341.1295. [DOI] [PubMed] [Google Scholar]
- 66.Siliciano JD, Siliciano RF. Enhanced Culture Assay for Detection and Quantitation of Latently Infected, Resting Virus in HIV-1-Infected Individuals. Methods Mol Biol. 2005;304:3–15. doi: 10.1385/1-59259-907-9:003. [DOI] [PubMed] [Google Scholar]
- 67.Chun TW, Carruth L, Finzi D, Shen X, DiGiuseppe JA, Taylor H, Hermankova M, Chadwick K, Margolick J, Quinn TC, Kuo YH, Brookmeyer R, Zeiger MA, Barditch-Crovo P, Siliciano RF. Quantification of latent tissue reservoirs and total body viral load in HIV-1 infection. Nature. 1997;387:183–8. doi: 10.1038/387183a0. [DOI] [PubMed] [Google Scholar]
- 68.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;9:727–8. doi: 10.1038/nm880. [DOI] [PubMed] [Google Scholar]
- 69.Crooks AM, Bateson R, Cope AB, Dahl NP, Griggs MK, Kuruc JD, Gay CL, Eron JJ, Margolis DM, Bosch RJ, Archin NM. Precise quantitation of the latent HIV-1 reservoir: Implications for eradication strategies. J Infect Dis. 2015;212:1361–5. doi: 10.1093/infdis/jiv218. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Eriksson S, Graf EH, Dahl V, Strain MC, Yukl SA, Lysenko ES, Bosch RJ, Lai J, Chioma S, Emad F, Abdel-Mohsen M, Hoh R, Hecht F, Hunt P, Somsouk M, Wong J, Johnston R, Siliciano RF, Richman DD, O’Doherty U, Palmer S, Deeks SG, Siliciano JD. Comparative analysis of measures of viral reservoirs in HIV-1 eradication studies. PLoS Pathog. 2013;9:e1003174. doi: 10.1371/journal.ppat.1003174. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Hermankova M, Siliciano JD, Zhou Y, Monie D, Chadwick K, Margolick JB, Quinn TC, Siliciano RF. Analysis of human immunodeficiency virus type 1 gene expression in latently infected resting CD4+ T lymphocytes in vivo. J Virol. 2003;77:7383–92. doi: 10.1128/JVI.77.13.7383-7392.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Wei DG, Chiang V, Fyne E, Balakrishnan M, Barnes T, Graupe M, Hesselgesser J, Irrinki A, Murry JP, Stepan G, Stray KM, Tsai A, Yu H, Spindler J, Kearney M, Spina CA, McMahon D, Lalezari J, Sloan D, Mellors J, Geleziunas R, Cihlar T. Histone deacetylase inhibitor romidepsin induces HIV expression in CD4 T cells from patients on suppressive antiretroviral therapy at concentrations achieved by clinical dosing. PLoS Pathog. 2014;10:e1004071. doi: 10.1371/journal.ppat.1004071. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Laird GM, Eisele EE, Rabi SA, Lai J, Chioma S, Blankson JN, Siliciano JD, Siliciano RF. Rapid quantification of the latent reservoir for HIV-1 using a viral outgrowth assay. PLoS Pathog. 2013;9:e1003398. doi: 10.1371/journal.ppat.1003398. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Cillo AR, Sobolewski MD, Bosch RJ, Fyne E, Piatak M, Coffin JM, Mellors JW. Quantification of HIV-1 latency reversal in resting CD4+ T cells from patients on suppressive antiretroviral therapy. Proc Natl Acad Sci U S A. 2014;111:7078–83. doi: 10.1073/pnas.1402873111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Vesanen M, Markowitz M, Cao Y, Ho DD, Saksela K. Human immunodeficiency virus type-1 mRNA splicing pattern in infected persons is determined by the proportion of newly infected cells. Virology. 1997;236:104–9. doi: 10.1006/viro.1997.8718. [DOI] [PubMed] [Google Scholar]
- 76.Procopio FA, Fromentin R, Kulpa DA, Brehm JH, Bebin AG, Strain MC, Richman DD, O’Doherty U, Palmer S, Hecht FM, Hoh R, Barnard RJO, Miller MD, Hazuda DJ, Deeks SG, Sékaly RP, Chomont N. A Novel Assay to Measure the Magnitude of the Inducible Viral Reservoir in HIV-infected Individuals. EBioMedicine. 2015;2:874–83. doi: 10.1016/j.ebiom.2015.06.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Howell B, Wu G, Swanson M, Lu M, Graham G, Strizki J, Wolkenberg S, Barnard R, Blair W, Hazuda D. Developing and applying ultrasensitive p24 protein immunoassay for HIV lateny. J Virus Erad. 2015;(Suppl 1):1–18. Oral Presentation 3.1. [Google Scholar]
- 78.Metcalf Pate KA, Pohlmeyer CW, Walker-Sperling VE, Foote JB, Najarro KM, Cryer CG, Salgado M, Gama L, Engle EL, Shirk EN, Queen SE, Chioma S, Vermillion MS, Bullock B, Li M, Lyons CE, Adams RJ, Zink MC, Clements JE, Mankowski JL, Blankson JN. A murine viral outgrowth assay to detect residual HIV type 1 in patients with undetectable viral loads. J Infect Dis. 2015;212:1387–96. doi: 10.1093/infdis/jiv230. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Spivak AM, Andrade A, Eisele E, Hoh R, Bacchetti P, Bumpus NN, Emad F, Buckheit R, Ccance-Katz EF, Lai J, Kennedy M, Chander G, Siliciano RF, Siliciano JD, Deeks SG. A pilot study assessing the safety and latency-reversing activity of disulfiram in HIV-1-infected adults on antiretroviral therapy. Clin Infect Dis. 2014;58:883–90. doi: 10.1093/cid/cit813. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Mothe B, Climent N, Plana M, Rosàs M, Jiménez JL, Muñoz-Fernández MÁ, Puertas MC, Carrillo J, Gonzalez N, León A, Pich J, Arnaiz JA, Gatell JM, Clotet B, Blanco J, Alcamí J, Martinez-Picado J, Alvarez-Fernández C, Sánchez-Palomino S, Guardo AC, Peña J, Benito JM, Rallón N, Perdiguero B, García-Arriaza J, Esteban M, de Quirós JCLB, Brander C, García F, Cobarsi P, Alvarez C, Sánchez S, Leal L, Torres B, Lucero C, Muñoz-Fernández MA, Gómez CE, Cepeda V, Sánchez-Sorzano CO, Jiménez L. Safety and immunogenicity of a modified vaccinia Ankara-based HIV-1 vaccine (MVA-B) in HIV-1-infected patients alone or in combination with a drug to reactivate latent HIV-1. J Antimicrob Chemother. 2015;70:1833–42. doi: 10.1093/jac/dkv046. [DOI] [PubMed] [Google Scholar]
- 81.Siliciano JD, Lai J, Callender M, Pitt E, Zhang H, Margolick JB, Gallant JE, Cofrancesco J, Moore RD, Gange SJ, Siliciano RF. Stability of the latent reservoir for HIV-1 in patients receiving valproic acid. J Infect Dis. 2007;195:833–6. doi: 10.1086/511823. [DOI] [PubMed] [Google Scholar]
- 82.Archin NM, Eron JJ, Palmer S, Hartmann-Duff A, Martinson JA, Wiegand A, Bandarenko N, Schmitz JL, Bosch RJ, Landay AL, Coffin JM, Margolis DM. Valproic acid without intensified antiviral therapy has limited impact on persistent HIV infection of resting CD4+ T cells. AIDS. 2008;22:1131–5. doi: 10.1097/QAD.0b013e3282fd6df4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Sagot-Lerolle N, Lamine A, Chaix M-L, Boufassa F, Aboulker J-P, Costagliola D, Goujard C, Pallier C, Delfraissy J-F, Lambotte O. Prolonged valproic acid treatment does not reduce the size of latent HIV reservoir. AIDS. 2008;22:1125–9. doi: 10.1097/QAD.0b013e3282fd6ddc. [DOI] [PubMed] [Google Scholar]
- 84.Routy JP, Tremblay CL, Angel JB, Trottier B, Rouleau D, Baril JG, Harris M, Trottier S, Singer J, Chomont N, Sékaly RP, Boulassel MR. Valproic acid in association with highly active antiretroviral therapy for reducing systemic HIV-1 reservoirs: Results from a multicentre randomized clinical study. HIV Med. 2012;13:291–6. doi: 10.1111/j.1468-1293.2011.00975.x. [DOI] [PubMed] [Google Scholar]
- 85.Archin NM, Bateson R, Tripathy MK, Crooks AM, Yang K-H, Dahl NP, Kearney MF, Anderson EM, Coffin JM, Strain MC, Richman DD, Robertson KR, Kashuba AD, Bosch RJ, Hazuda DJ, Kuruc JD, Eron JJ, Margolis DM. HIV-1 expression within resting CD4+ T cells after multiple doses of vorinostat. J Infect Dis. 2014;210:728–35. doi: 10.1093/infdis/jiu155. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Elliott JH, Wightman F, Solomon A, Ghneim K, Ahlers J, Cameron MJ, Smith MZ, Spelman T, McMahon J, Velayudham P, Brown G, Roney J, Watson J, Prince MH, Hoy JF, Chomont N, Fromentin R, Procopio FA, Zeidan J, Palmer S, Odevall L, Johnstone RW, Martin BP, Sinclair E, Deeks SG, Hazuda DJ, Cameron PU, Sékaly RP, Lewin SR. Activation of HIV Transcription with Short-Course Vorinostat in HIV-Infected Patients on Suppressive Antiretroviral Therapy. PLoS Pathog. 2014;10:1–19. doi: 10.1371/journal.ppat.1004473. [DOI] [PMC free article] [PubMed] [Google Scholar]