SUMMARY
HIV-1 DNA persists in infected cells despite combined antiretroviral therapy (cART), forming viral reservoirs. Recent trials of strategies targeting latent HIV reservoirs have rekindled hopes of curing HIV infection, and reliable markers are thus needed to evaluate viral reservoirs. Total HIV DNA quantification is simple, standardized, sensitive, and reproducible. Total HIV DNA load influences the course of the infection and is therefore clinically relevant. In particular, it is predictive of progression to AIDS and death, independently of HIV RNA load and the CD4 cell count. Baseline total HIV DNA load is predictive of the response to cART. It declines during cART but remains quantifiable, at a level that reflects both the history of infection (HIV RNA zenith, CD4 cell count nadir) and treatment efficacy (residual viremia, cumulative viremia, immune restoration, immune cell activation). Total HIV DNA load in blood is also predictive of the presence and severity of some HIV-1-associated end-organ disorders. It can be useful to guide individual treatment, notably, therapeutic de-escalation. Although it does not distinguish between replication-competent and -defective latent viruses, the total HIV DNA load in blood, tissues, and cells provides insights into HIV pathogenesis, probably because all viral forms participate in host cell activation and HIV pathogenesis. Total HIV DNA is thus a biomarker of HIV reservoirs, which can be defined as all infected cells and tissues containing all forms of HIV persistence that participate in pathogenesis. This participation may occur through the production of new virions, creating new cycles of infection and disseminating infected cells; maintenance or amplification of reservoirs by homeostatic cell proliferation; and viral transcription and synthesis of viral proteins without new virion production. These proteins can induce immune activation, thus participating in the vicious circle of HIV pathogenesis.
INTRODUCTION
HIV DNA persists in infected cells during combined antiretroviral therapy (cART), allowing the virus to reemerge from the reservoir if treatment is discontinued (1–3). The virus cannot currently be eradicated from the body, and treatment thus has to be maintained indefinitely. Recent clinical studies have rekindled hopes that HIV infection might be cured, notably by targeting viral reservoirs (4–7). An accurate, clinically relevant marker of HIV reservoirs is therefore needed (8).
Several markers have been proposed to quantify cell-associated HIV reservoirs (6) but there is no consensus method (6, 9–12). Intracellular HIV RNA load indicates the degree of ongoing HIV replication, while coculture assay of resting infected cells indicates their capacity to produce replication-competent virions. In contrast, total cell-associated HIV DNA is a global biomarker that includes integrated and nonintegrated viral genomes coding for both competent and defective viruses.
Total cellular HIV DNA is the focus of this review. It is easy to measure in whole blood, cell pellets, or tissues. Here, we examine the relevance of HIV DNA to HIV pathogenesis and persistence during both the natural and on-treatment course of infection, as well as its implications for tailored therapy and for trials of new approaches targeting HIV reservoirs.
PLACE OF TOTAL HIV DNA AMONG MARKERS USED TO EVALUATE HIV RESERVOIRS
There are many discussions and controversies concerning the best biomarker of HIV reservoirs. Two comprehensive studies have compared a panel of HIV reservoir biomarkers (9, 11), but such studies are limited by the fact that large amounts of blood are needed to test all markers in a given patient. The different approaches may be used differently according to the issue in question, such as the overall level of HIV infection in the body, viral persistence, reservoir activity, or the role of the HIV reservoir in maintaining immune activation. Clearly, no single marker can answer all these questions, but each can provide part of the answer. Markers used to quantify and monitor the HIV reservoir provide complementary information (6, 9).
Blankson et al. proposed to restrict the term “HIV reservoirs” to the cells or tissues where HIV persists in latent form but can reactivate in the form of replication-competent virus (13). This restricts the definition to resting infected cells. An alternative, broader definition of HIV reservoirs can also be proposed: all infected cells and tissues containing all forms of HIV persistence that can participate in HIV pathogenesis. This participation may occur through the production of new virions creating new cycles of infection and disseminating infected cells; maintenance or amplification of reservoirs by homeostatic cell proliferation; and viral transcription and synthesis of viral proteins without new virion production. These proteins can induce immune activation, thus participating in the vicious circle of HIV pathogenesis (14) (Fig. 1). Different biomarkers are relevant to each of these definitions.
FIG 1.
Several forms of HIV DNA compose the total HIV DNA and participate in HIV pathogenesis. The integrated form, the provirus, is the more persistent form and permits production of virions when quiescent infected cells are reactivated. Virions can infect new cells and propagate infection and the HIV reservoir. The provirus form persists in all cells during cell proliferation. Episomal forms with 1-LTR or 2-LTR persist and are diluted in some daughter cells during cell proliferation. Linear unintegrated HIV DNA is the more labile form and is essentially present when the virus is replicating. Defective provirus, with a deletion, nonsense mutation, or hypermutation, cannot produce new virions but can produce transcripts and viral proteins which could activate the immune system and participate in HIV pathogenesis.
Quantitative coculture assays measure the ability of infected cells (purified CD4+ T cells or peripheral blood mononuclear cells [PBMC]) to produce infectious virions upon stimulation (3, 15, 16, 17, 18). It is based on the limiting dilution method, and the results are expressed as infectious units per million cells (IUPM) (6, 9). This viral outgrowth assay requires the purification of PBMC or resting CD4 T cells via use of magnetic beads or flow cytometry, as well as cell activation to induce virus production during several weeks in a secure laboratory and also quantification of cells able to release infectious particles (10, 15). It requires a large volume of blood (120 to 180 ml) or leukapheresis to obtain the necessary large number of viable cells. It is difficult to apply for frequent serial measurements in clinical studies (9, 15) because it is labor-intensive and expensive. Its reproducibility has been estimated in one laboratory in terms of the coefficient of variation (0.95) and the confidence interval (CI) for one measurement (±0.7 log), indicating it is unreliable for detecting small differences in the size of the HIV reservoir (9, 19, 20). Reproducibility across laboratories has not yet been evaluated. Its limitations are illustrated by the case of the “Mississippi child,” a perinatally HIV-1-infected infant who experienced a viremic relapse after entering remission; quantitative coculture remained negative (in a cumulative total of 64 million resting CD4 T cells) throughout the period of virologic remission, whereas HIV DNA was detectable in PBMC and CD4 T cells (positive signal, <4 copies/106 cells). Her subsequent viral rebound 27.6 months after cART discontinuation proved the persistence of infectious virus somewhere in the “Mississippi child” in blood or unsampled tissues (21). Finally, despite its limits, this assay is certainly useful for some research purposes, such as estimation of blood reservoir productive activity.
Another marker is intracellular HIV RNA load, which measures HIV transcription in infected cells or viral entry. Cell-associated HIV RNA rapidly declined after cART initiation, mimicking cell-free HIV RNA decay, before reaching a plateau (22). This plateau is explained by the fact that cART does not stop HIV transcription. The level of this plateau does not correlate directly with HIV replication, because of insufficient transcription or nuclear retention of viral RNA in resting cells, or transcription of defective viruses (22). Cell-associated HIV RNA might be very useful for cure research, because it quantifies a dynamic process of transcription and reflects the activity of the HIV reservoir (22). It can be helpful for evaluating residual, transcriptionally active infected cells in patients on treatment. Several assays have been developed to quantify this intracellular HIV RNA (22), and their reproducibility across multiple labs needs to be evaluated.
Total HIV DNA, as its name suggests, includes both integrated and unintegrated HIV DNA (that includes episomal 1-long terminal repeat [1-LTR], 2-LTR, and linear HIV DNA) and reflects the global level of the total reservoir (Fig. 1). Each of these separate forms is difficult to quantify in the absence of a standardized, reproducible assay (6). Total HIV DNA assay does not distinguish between replication-competent and -defective viral genomes (9, 17). These forms of HIV DNA can coexist in infected cells. The neosynthesized double-strand HIV DNA is linear. This form is the prevalent form of reverse-transcribed genomes in infected cells and constitutes a preintegrated form of latency (3, 23–25). It is labile if it does not integrate into the host genome (26, 27). HIV DNA can also be circularized in episomal forms with one or two LTRs. Forms with one LTR are due to homologous recombination, whereas forms with two LTRs result from ligation of the viral genome (28). In quiescent CD4 T cells, episomal forms represent less than 10% of unintegrated HIV DNA (3). Two-LTR forms represented 0.1% to 0.6% of integrated forms in cell lines and peripheral blood mononuclear cells infected in vitro (29). Data on 2-LTR forms are conflicting. Some authors consider them labile and to be present only during recent infection (30, 31), but in vitro studies that eliminated confounding factors, such as infected cell death, and in vivo studies have demonstrated that they are stable (32–39). The number of 1-LTR and 2-LTR HIV DNA molecules decreases by dilution during cell division or cell death (32, 34, 40) (Fig. 1). These forms can be renewed by viral replication (30, 41). 2-LTR forms have been quantified during trials evaluating intensification with integrase inhibitors upon the hypothesis that these antiretrovirals enhance 2-LTR HIV DNA if residual replication occurs during cART (42). These unintegrated forms constitute an inducible viral reservoir (43) and can participate in HIV transcription, in the replication cycle (when they are complemented by integrated forms), and in the integration and synthesis of new virions (44–53). Integrated HIV DNA is the most stable form and constitutes the postintegrated form of latency (16, 54). It persists during cell proliferation (Fig. 1). This provirus has an estimated frequency of 1 copy per infected blood cell (55). It contributes to viral rebound after cART interruption. A significant percentage of infected resting CD4 T cells harbors proviruses that are either defective because of hypermutation or deletion or transcriptionally silenced (17, 56). Some studies have suggested that the frequency of resting HIV-infected cells, estimated by total HIV DNA quantification among resting CD4 T cells, is considerably higher than the frequency of infected cells capable of viral outgrowth (differences above 2 logs) (9). Some of these transcriptionally silent cells may be inducible. These data suggest that a significant proportion of the total HIV DNA measured by PCR is not being made into HIV RNA, protein, and viral particles. Cells harboring defective or silent proviruses do not therefore contribute to viremia (17, 56) but could participate in pathogenesis (Fig. 1). Total HIV DNA load correlates with the frequency of cells containing replication-competent virus (r = 0.73, P = 0.0009) (57). Eriksson et al. also reported a correlation between total HIV DNA load in resting CD4+ T cells (but not in PBMC) and the quantity of replication-competent virus (r = 0.45, P = 0.08) (9). Similar results were recently reported by Kiselinova et al. (11). More recently, Noel et al. also reported that the low level of total HIV DNA in cells from long-term nonprogressors (LTNPs) correlated with the low efficiency of virus production after activation ex vivo (58).
By comparison with other markers, total HIV DNA has the advantage of easy quantification by standardized, sensitive, real-time PCR (59–68), a method suitable for analyzing large numbers of samples with accuracy. It requires a relatively small amount of blood and can therefore be used for HIV diagnosis in young children, for example. It can be quantified in blood and other body fluids, is unaffected by freeze-thawing, and is the method most widely used to quantify the HIV reservoir in tissue biopsy specimens. It produces reproducible results with small errors (59, 69). The interassay reproducibility of real-time PCR quantification of total HIV DNA has been evaluated in a laboratory by using two controls, one high and one moderate, during more than a 9-month period. Means, coefficients of variation, and 95% confidence intervals (CIs) were, respectively, 3.30 log copies/106 leukocytes, 0.03, and ±0.22 log for the high-level sample and 2.40 log copies/106 leukocytes, 0.07, and ±0.35 log for the moderate-level sample. Figure 2 shows the high reproducibility of this assay. Reproducibility has also been evaluated for patient samples, with good performance (59). These values are lower than those reported with the viral outgrowth assay (9, 19, 20). A kit is commercially available (Biocentric, Bandol, France) that was developed with the support of the French HIV/AIDS Research Agency (ANRS) to facilitate comparisons of results from different laboratories. For example, HIV DNA load is an inclusion criterion in an ongoing cART de-escalation trial (TRULIGHT; NCT02302547 at ClinicalTrials.gov), in which it is quantified with the same assay after an interlaboratory quality-control procedure. Interlaboratory comparisons have had good results (69). More recently, an assay that dispenses with the extraction step has been proposed (70). Recent technical progress has permitted the development of HIV DNA quantification by digital droplet PCR (ddPCR), which does not require an external quantification standard (71–77). However, some false-positive signals have been observed with ddPCR, which can be a problem when this method is used for diagnosis (76). Moreover, ddPCR cannot help with quantification of low signals, for instance in HIV controllers. In fact, quantification of low signals follows the Poisson distribution, necessitating study of many cells and several replicates, whatever the technology (real-time PCR or ddPCR).
FIG 2.
Reproducibility of total HIV DNA quantification in two positive controls by real-time PCR (59). For this experiment, two different pools of blood cells (in EDTA) from HIV-infected patients were prepared. Aliquots were frozen at −80°C. One aliquot of each control (high-level positive control [A] and moderate-level positive control [B]) was tested in all assays performed during a >9-month period to verify the assays reproducibility over time. Results presented here are for 69 quantifications performed by different technicians and with different thermocyclers and with four lots of Biocentric reagents, for the high-level positive control (A) and moderate-level positive control (B). Means (red lines), standard deviations, coefficients of variation, and 95% confidence intervals were, respectively, 3.30 log copies/106 leukocytes, 0.11 log, 0.03, and ±0.22 log for the high-level positive-control sample and 2.40 log copies/106 leukocytes, 0.18 log, 0.07, and ±0.35 log for the moderate-level positive-control sample.
It is very unlikely that a single marker will alone provide sufficient information on viral reservoir status (8). However, the fact that HIV DNA shows a certain degree of correlation with other markers, including viral outgrowth assay results, indicates that HIV DNA may be usefully included in biomarker panels, whatever the question in hand. Total HIV DNA load is the most widely studied marker of the HIV reservoir, having been measured in large patient cohorts and trials at different stages of the infection.
SPECTRUM OF TOTAL HIV DNA LEVELS IN BLOOD
A broad range of total HIV DNA levels has been observed during the natural and therapeutic courses of HIV infection (Fig. 3).
FIG 3.
Spectrum of total HIV-1 DNA levels in PBMC during HIV infection. The natural history data are from HIV DNA quantified for 552 adults at the time of the primary infection (PRIMO cohort, ANRS) (81), for 271 patients who had seroconverted 6 to 24 months previously (SEROCO cohort, ANRS) (84), and for 121 perinatally infected children (median age, 6 years), of whom 46.6% and 20.3% were at CDC stage B and C, respectively (ANRS 1244/1278) (94). The data set for “during antiretroviral therapy” is for HIV DNA quantified during antiretroviral therapy initiated during the primary infection and continued for 2 years (90 patients; OPTIPRIM trial, ANRS) (109) or for a median of 3.6 years, with HIV RNA levels of <50 copies/ml for a median of 3.1 years (n = 35) (123). HIV DNA was quantified during antiretroviral therapy initiated early during the chronic phase in 116 adults (CD4 cell count, >350/mm3; plasma HIV RNA, <4.7 log copies/ml), with antiretroviral therapy for a median of 5.3 years (SALTO trial, ANRS) (220) and in 272 adults treated later (median CD4 cell nadir, 228/mm3; plasma HIV RNA, 5.3 log copies/ml) for a median of 7.3 years, with HIV RNA levels at <50 copies/ml for a median of 3.9 years (123). HIV DNA was quantified in 44 adults with advanced therapeutic failure and AIDS (CD4 count, ≤200/mm3; HIV RNA level, >4 log; genotypic score showing two or fewer active drugs) (ETOILE trial, ANRS) (197). For the HIV control data set, HIV DNA were quantified for patients who controlled the infection naturally (plasma HIV RNA undetectable for >10 years in the absence of antiretroviral treatment [15 patients]) (HIV controllers cohort, ANRS) (89) or after treatment interruption (14 patients) (VISCONTI study, ANRS) (92). The same standardized assay was used in the same laboratory. A broad range of HIV DNA levels was found. Medians and interquartile ranges are indicated.
Total HIV DNA in Blood during Untreated Infection
Cellular HIV DNA can be detected very early after infection. Ananworanich et al. reported that patients studied a few days after infection had a median total blood HIV DNA load of 0.9 log10 copies/106 PBMC at Fiebig stage I (at which only HIV RNA is detectable; 8 patients) and 2.7 log10 copies/106 PBMC at Fiebig stage III (detection of HIV RNA and HIV-specific antibodies is possible by enzyme-linked immunosorbent assay [ELISA] but not by Western blotting; 15 patients) (P = 0.01) (78). This was recently confirmed in a larger study (79). In the ANRS PRIMO cohort, the median HIV DNA load was 3.30 log10 copies/106 PBMC (range, <1.84 to 4.93) at an estimated median of 47 days after infection; it was significantly higher in patients with more acute infection (with only 0 or 1 HIV-specific antibody) than in patients included a bit later (with ≥2 antibodies), and also in patients who had a symptomatic primary infection (80). During the 6 months following infection, the median total HIV DNA level in blood was 2.8 to 3.2 log10 copies/106 PBMC among patients in the French PRIMO cohort (Fig. 3) and the Ivorian PRIMOCI cohort (674 and 200 patients, respectively) (80–83).
The total blood HIV DNA level was 2.86 log10 copies/106 PBMC during the 6 to 24 months following infection in 271 patients included in the ANRS SEROCO cohort (Fig. 3) (84).
During the chronic phase, the median HIV DNA level was 2.65 log10 copies/106 PBMC in 130 patients (85) and was quite stable over time (85–87).
Patients who naturally control HIV infection (long-term nonprogressors and elite controllers) have very low and stable total HIV DNA levels, with median values of 2.3 and 1.5 log10 copies/106 PBMC reported in, respectively, 66 and 15 patients (88–90). Similarly, posttreatment controllers in the VISCONTI cohort, for whom treatment was initiated early during the primary infection and long-term viral control after treatment interruption was maintained, had low total HIV DNA levels (median, 1.71 log10 copies/106 PBMC; 14 patients) (91, 92) (Fig. 3).
Very few data are available for untreated AIDS patients. HIV DNA load expressed per 106 CD4 T cells increased in one study during infection, reflecting the increasing proportion of infected cells among the depleted CD4 T cell subset (24 patients) (87).
Infected teenagers included in the ANRS IMMIP study had HIV DNA levels similar to those found in adults (93). Eighty-one Ivorian children, studied mainly at stage B or C of HIV infection, had a median of 3.4 log10 copies/106 PBMC (94) (Fig. 3).
Thus, a broad range of total HIV DNA levels is observed among patients. Moreover, HIV DNA levels vary widely during the natural course of infection, especially during the first few weeks.
Decline in Total HIV DNA during cART
The decline in total blood HIV DNA in patients adherent to cART has been extensively studied in both adults and children (57, 77, 78, 82, 95–109). This decline varies among patients. It correlates directly with pretherapeutic HIV DNA levels (97, 102) and HIV RNA levels (97), as well as with the baseline CD4 cell count (102), the CD4 cell increment (97, 100), and the chance of achieving HIV RNA loads of <2.5 copies/ml (107). Pretherapeutic HIV RNA load was also predictive of the decline in HIV DNA load in children, independently of pretherapeutic HIV DNA load (110). HIV DNA load before cART was also predictive of HIV DNA load 2 years after cART initiation and of the time to aviremia in children (111). A sharp decrease (≥0.5 log) in HIV DNA levels was significantly correlated with an improvement in the CD4 T cell count (112). A recent study of children showed lower HIV DNA levels after cART initiation when the plasma HIV RNA level was rapidly controlled (77).
HIV DNA decay is also dependent on the amount of cell-associated HIV RNA in blood and lymph nodes; residual viremia correlates with slower clearance of HIV-1-infected cells, possibly owing to infection of new cells (97). Another, larger study confirmed the association between HIV DNA levels during cART and markers of the dynamics of HIV reservoirs (residual HIV RNA, cell-associated HIV RNA, and 2-LTR HIV DNA) (113). However, another study failed to show a correlation between the HIV DNA slope or level on the one hand and, on the other hand, either residual viremia or the percentage of activated CD8+ T cells (averaged over years 1 to 4 after cART initiation) (108). This point therefore needs further investigation.
The rapid initial HIV DNA decay is mostly due to the decay of linear unintegrated DNA (114). Integrated forms decay slowly (115). 2-LTR forms decline more than integrated forms, and the decay of 2-LTR forms and integrated forms is slower in activated CD38 CD4 T cells than in nonactivated cells (115). In some patients on cART, total HIV DNA can be almost exclusively composed of integrated HIV DNA (104, 114, 116, 117), whereas other studies suggest that some nonintegrated HIV DNA forms can also persist (115, 118) and that unintegrated HIV DNA forms remain more frequent than integrated forms in quiescent CD4 T cells and monocytes after several months of cART (119, 120, 121). Heterogeneity among patients could explain these discrepancies.
Time from HIV-1 Infection to Treatment Initiation Influences Total HIV DNA Decay and Levels during cART
cART reduces the HIV DNA load more markedly when initiated during primary HIV infection (PHI) than during chronic HIV infection (CHI) in adults and children (Fig. 3) (57, 77, 82, 101, 105, 122, 123).
In a longitudinal study of 307 patients, the decline in HIV DNA was analyzed with a nonlinear mixed-effects model that included >1,100 HIV DNA data points. The model showed two phases of decay. During the first 2 years, a rapid decline in HIV DNA was observed both in patients treated since PHI and in patients first treated during CHI, with similar HIV DNA half-lives (113 days and 146 days, respectively). The second decay phase was much slower, and the half-life was significantly shorter in the PHI group (25 years) than in the CHI group (377 years; P < 0.001). At the end of the study, after a median duration of viral suppression of 4 years, HIV DNA levels were significantly lower in the PHI group than in the CHI group (median, 2.15 versus 2.84 log copies/106 PBMC; P < 0.0001) (Fig. 3) (123). Logistic regression analysis showed that starting cART during PHI (odds ratio [OR], 16; 95% CI, 3.5 to 72.3) and a low pretherapeutic HIV DNA level (<3.3 log; OR, 4.8; 95% CI, 1.2 to 19.3) were independent predictors of reaching “optimal viro-immunological responder status” (i.e., HIV DNA of <2.3 log copies/106 PBMC, associated with normalization of absolute/relative CD4 T cell counts and the CD4/CD8 ratio) (123). Recently, a mixed-effects model of the ANRS PRIMO cohort data set, using >1,300 HIV DNA values from 327 patients, demonstrated that the timing of cART initiation during PHI could influence the first slope of HIV DNA decline: the earlier cART was initiated after infection, the faster HIV DNA levels fell during the first 8 months on treatment (−0.171, −0.131, and −0.0068 log10 copies/106 PBMC per month when cART was initiated 15 days, 1 month, and 3 months after infection, respectively; P < 0.0001) (124). Ultimately, the earliness of cART initiation impacts HIV DNA levels observed after 5 years of effective treatment (1.62 and 2.24 log10 copies/106 PBMC, respectively, when cART is initiated 15 days and 3 months after infection; P = 0.0006) (124).
Buzon et al. reported a statistical correlation between the time from HIV infection to treatment initiation and the total HIV DNA level after 10 years of continuous treatment in a cohort of adults first treated early in the infection (57). The lowest HIV DNA levels were observed in patients who had low baseline levels and who received early treatment (107). In children, the HIV DNA level was markedly lower when viral control was achieved before age 1 year than between the ages of 1 and 5 years or after age 5 years (73). This difference is linked to the duration of uncontrolled viral replication between initial infection and effective treatment. The lower level of HIV DNA in children treated early versus late correlates with a lower level of replication-competent virus (75).
Because of its impact on the HIV DNA level, antiretroviral treatment of newborns within the first hours after infection has consequences for diagnosis during the first months of life, a period when maternal antibodies interfere with serological tests. Antiretroviral prophylaxis of mother-to-child HIV transmission both during pregnancy and in newborns can mask the primary infection in newborns. In the French perinatal cohort, HIV DNA levels, like viremia, were lower at age 1 month among infected infants receiving multidrug prophylaxis (levels were sometimes near the limit of quantification), stressing the need for highly sensitive assays to diagnose neonates born to HIV-infected mothers (59, 125). Under these conditions, total HIV DNA can represent an early diagnostic marker in infants (59, 126). HIV DNA can also be quantified in dried blood spots by real-time PCR, which can be helpful for this diagnosis in resource-limited countries (127).
Thus, the time from HIV-1 infection to treatment initiation strongly impacts the initial rate of decay of total HIV DNA and the level achieved after several years of cART.
Total HIV DNA levels show a broad range of values (Fig. 3) during the natural course of infection, during antiretroviral therapy (initiated during either PHI or CHI), and also during therapeutic failures or AIDS. Patients who control the infection naturally or after treatment interruption have very low HIV DNA levels (37, 61, 88, 89, 91, 92, 128).
TOTAL HIV DNA LEVELS IN TISSUES AND FLUIDS REFLECT THE SPREAD OF INFECTION THROUGH THE BODY
Measurement of total HIV DNA in tissues and fluids provides interesting information on the pathogenesis of HIV infection, as it indicates the spread of the virus through the body (Fig. 4).
FIG 4.
Anatomical HIV reservoirs. Reservoir cells are highly disseminated.
Lymph Nodes
The lymph nodes are a major reservoir site, with a large pool of target cells, a high level of activation, and a high level of replication, inducing infection of new cells (Fig. 4). Lymph nodes thus play an important role in the dynamics of HIV reservoirs, in an intricate relation with CD4 T cell dynamics. Follicular helper CD4 T cells (TFH) play an important role in the lymph node HIV reservoir and are infected during their differentiation (129). A recent study compared viral DNA levels in different cell subsets from lymph nodes of simian controllers and progressors. It was found that TFH, one of the most numerous subsets of effector T cells, harbored high levels of viral DNA, similar to those observed in non-TFH from progressors but significantly higher than those observed in non-TFH from controllers (130). This was linked to preferential production of replication-competent virus in B cell follicles of lymph nodes and spleens from controllers, where TFH are localized (130). These B cell follicles were anatomically protected from specific CD8 T cell responses in controllers, explaining the difference in viral DNA levels between TFH and non-TFH cells (130). Moreover, in monkeys receiving cART, infected TFH were more strongly involved in residual replication than other T cells: more simian immunodeficiency virus (SIV) RNA was produced by TFH for a given SIV DNA level (130). Lymph nodes thus constitute an obstacle to HIV remission, as they are not readily accessible by specific immune responses (130) or antiretrovirals (131). Phylogenetic analyses have shown viral evolution in lymphoid tissue during cART; this reflects ongoing replication that replenishes the HIV reservoir (132). When cART is interrupted, many cells expressing different viral RNA variants are found, contributing to viral rebound (133). These new data demonstrate that lymph node cells harboring viral DNA permit productive infection even in the presence of suppressive cART. The very rare TFH in blood also contribute to the HIV reservoir in patients receiving cART (134).
Gut-Associated Lymphoid Tissue
Gut-associated lymphoid tissue (GALT), which harbors 60% of the body's lymphocytes, plays an essential role in the pathogenesis of HIV infection, through Th17 cell depletion, bacterial translocation, and local host cell activation (facilitating HIV replication); this has been reported both in humans and in simian models (135–157) (Fig. 4). Gastrointestinal CD4 T cells have been shown to harbor, on average, 13 times more HIV DNA than blood CD4 T cells during acute and early-stage HIV infection (151, 158–160). Non-CD4 T cells harbored less HIV DNA than CD4 T cells in the gut, but the infection level of non-T leukocytes was higher in GALT than in blood (161). Myeloid cells also harbor HIV DNA in GALT (162). A recent study of patients with acute infection showed that HIV DNA in the gut was significantly lower at Fiebig stage I than at Fiebig stages II to IV (79). The HIV DNA load in GALT declines after cART initiation (78) but does not disappear (150, 158, 163, 164), and HIV DNA levels vary at different gut sites (151, 158–161). Memory effector cells harbored the most HIV DNA in the ileum and rectum (161). HIV DNA in the rectum remained higher in cART-treated patients than in HIV controllers (165). Total HIV DNA in GALT correlates with total HIV DNA load in blood at different stages of the infection, both without and during treatment (139, 158). This finding could partly explain the relevance of HIV DNA to HIV pathogenesis, even when measured only in blood, like the CD4 T cell count and HIV RNA load, two routine follow-up markers.
Other Tissues and Fluids
The central nervous system constitutes a viral reservoir, and circulating monocytes play a particular role in transferring the infection to the brain (166). HIV DNA is also detected in astrocytes (167). cART can often control local replication, but cells harboring viral DNA persist (168, 169).
HIV DNA is quantifiable in semen in nonsperm cells (170, 171). It can persist in the genital tract of women on long-term effective cART (HIV RNA, <50 copies/ml at >6 months) and has been linked to current residual viremia (OR, 3.4; 95% CI, 1.1 to 10.9) and to a history of AIDS-defining illness (OR, 11; 95% CI, 2 to 61) (172). The presence of HIV in genital secretions should be taken into account when estimating the residual risk of transmission.
HIV DNA is also useful for detecting infection of kidney grafts in HIV-infected recipients who had undetectable plasma HIV RNA at the time of transplantation (173). HIV was found to infect the kidney allograft in 68% of cases and might influence graft survival. Detection of HIV DNA and HIV RNA in a patient's urine is a noninvasive way of monitoring kidney graft infection (173, 174).
In the context of stem cell transplantation for AIDS-related lymphoma, total HIV DNA load in the autograft predicts the posttransplant HIV peripheral reservoir size in patients on continuous highly active antiretroviral treatment (175).
Recently, adipose tissue, and particularly memory CD4 T cells in this tissue, has been identified as a potentially important HIV reservoir, with the detection of total viral DNA in untreated macaques but also in patients on effective treatment (176, 177). This reservoir is inducible by ex vivo activation.
In the future, measurement of total HIV DNA in different tissues and fluids may help to evaluate therapeutic strategies designed to eradicate viral reservoirs.
TOTAL HIV DNA: A CLINICALLY RELEVANT MARKER
Total HIV DNA Load Is Predictive of the Natural Disease Course, Independently of HIV RNA Load and the CD4 T Cell Count
Total HIV DNA load in PBMC varies widely among untreated HIV-infected adults and children (82, 85, 102, 110, 178, 179), even during PHI, with an interquartile range of 2.7 to 3.5 log10 copies/106 PBMC (Fig. 3) (81). The range of HIV DNA levels was smaller than the range of HIV RNA levels, but nonetheless identified several patient subgroups in large cohorts; in particular, LTNPs and elite controllers had significantly lower HIV DNA levels than progressors (61, 88–90) (Fig. 3). Likewise, symptomatic patients had significantly higher HIV DNA loads than asymptomatic patients during CHI (179). This heterogeneity of HIV DNA loads has noteworthy implications for the course of infection.
Total HIV DNA load in the first 6 months following seroconversion is predictive of immunologic progression, independently of HIV RNA load and the CD4 T cell count (81, 106, 180). In the ANRS PRIMO cohort, patients who progressed rapidly had a median total HIV DNA load of 3.3 log, compared to 3.0 log in other patients (81). Similar results were obtained in the first 19 or 24 months after seroconversion (83, 84), with higher HIV DNA levels having major independent prognostic value for progression to clinical AIDS, <200 CD4 cells/mm3, and death. Patients who had not progressed more than 5 years after seroconversion had significantly lower HIV DNA load in the months following seroconversion than patients who did not control the infection (2.02 log versus 2.90 log) (181). LTNPs with HIV DNA loads of >1.85 log at seroconversion and a large increase in HIV DNA over time had an increased risk of losing their LTNP status (182). These data show that even small differences in HIV DNA levels influence the course of HIV infection.
The predictive value of HIV DNA load for progression to AIDS and death was confirmed during CHI, independently of age at seroconversion, the CD4 cell count, and HIV RNA load (85).
A meta-analysis of six studies with a total of 1,074 participants indicated that total HIV DNA was a strong predictive marker of AIDS (relative risk [RR], 3.01, 95% CI, 1.88 to 4.82) and of all-cause mortality (RR, 3.49; 95% CI, 2.06 to 5.89) (183). It was a significantly better predictor for progression to AIDS than was HIV RNA (ratio of RRs, 1.47; 95% CI, 1.05 to 2.07) and for a combined endpoint of AIDS and death (ratio of RRs, 1.51; 95% CI, 1.11 to 2.05) (183).
In summary, HIV DNA levels vary among patients during the course of the infection and have high predictive value for disease progression (Fig. 3).
Pretherapeutic Total HIV DNA Levels in PBMC Are Predictive of Virologic, Immunologic, and Clinical Responses to Antiretroviral Therapy
HIV DNA load is predictive of the long-term success of cART. Indeed, patients with lower blood HIV DNA loads at cART initiation have better virologic and immunologic responses to treatment and longer survival (179).
Lower baseline HIV DNA levels were predictive of achieving an undetectable HIV RNA load on cART (103, 178, 184–186), while high HIV DNA load was associated with persistent residual HIV RNA below 50 copies/ml during cART, independent of baseline HIV RNA load (185). This was confirmed by Parisi et al., who showed that baseline HIV DNA load predicted the residual HIV RNA plasma level during effective cART (107).
Pretherapeutic HIV DNA load has been linked to immune cell activation status during cART, correlating positively with the number of Ki-67+ CD8 T cells after 6 months of treatment (187).
Baseline HIV DNA load was predictive of overall survival among patients with relapsing or refractory HIV-related lymphoma treated with high-dose chemotherapy followed by autologous stem cell transplantation (188).
Total HIV DNA Load during cART Is Informative of Patient Pretherapeutic and Therapeutic History
HIV DNA load during treatment correlated with blood HIV DNA load at cART initiation in adults and children (82, 103, 110). In adults, HIV DNA load at years 4, 7, and 10 of antiretroviral treatment correlated positively with the pretherapeutic HIV DNA level (108). Likewise, Ananworanich et al. showed that total blood HIV DNA at cART initiation predicted the HIV reservoir size at week 24 of treatment that was started during PHI (P < 0.001) (78). The plasma HIV RNA zenith and the CD4 cell nadir were predictive of HIV DNA load during cART in patients whose plasma HIV RNA remained below the detection limit for more than 3 years; HIV DNA load during cART correlated positively with pretherapeutic HIV RNA load and negatively with the CD4 cell nadir (189, 190, 191, 192). Total HIV DNA load, measured 6 weeks after cART initiation, was the strongest independent predictor of the pretherapeutic HIV RNA level (193). Thus, total HIV DNA load during cART reflects pretherapeutic characteristics of HIV infection, including HIV DNA load, the plasma HIV RNA zenith, and the CD4 cell nadir.
Multivariate analysis indicated that low HIV DNA load in patients with cART for a median of 25 months was significantly associated with prolonged HIV RNA suppression on cART (194). HIV DNA load during cART was independently associated with cumulative HIV RNA viremia over the previous 5 years (93, 192). HIV DNA load could therefore reflect therapeutic adherence over time. Furthermore, in patients on effective cART, total HIV DNA correlated with residual plasma viremia, measured with an ultrasensitive assay (115, 190, 191, 195). Total HIV DNA load, measured 6 weeks after cART initiation, was predictive of virologic outcome in asymptomatic, chronically HIV-1-infected persons (193). Median HIV DNA load was 2.20 log copies/106 cells (range, 0.70 to 2.80) in patients treated for at least 10 years without ART interruptions or immunomodulatory therapy and selected on the basis of undetectable HIV RNA throughout follow-up, with at least one yearly HIV RNA measurement, and a total of more than 20 measurements (196). Conversely, in patients with advanced therapeutic failure and AIDS, median HIV DNA load was 3.44 log copies/106 PBMC (197) (Fig. 3). cART-treated patients with X4-tropic viruses in HIV DNA, reflecting more advanced disease, had higher levels of HIV DNA than patients with R5-tropic viruses (198).
High HIV DNA load in PBMC is associated with intermittent HIV shedding in the semen of men who have sex with men and who have been on successful antiretroviral therapy for >6 months: after multivariable adjustments, total HIV DNA (OR of 2.6 and 95% confidence interval [CI] of 1.2 to 6.0 for >2.5 log10 copies/106 PBMC; P = 0.02) and cannabis use accompanying sexual intercourse (OR of 2.8, CI of 1.2 to 6.7; P = 0.02) were the two factors significantly associated with HIV RNA detection in seminal plasma (199).
Moreover, multivariate analysis showed that low total HIV DNA loads in patients on long-term suppressive antiretroviral treatment were clearly associated with concomitant residual plasma viremia of <1 copy/ml, a high CD4/CD8 cell ratio, and a high CD4 T cell percentage (P < 0.0001) (191). Several studies have shown that a lower HIV DNA load during cART is associated with better immune recovery (194, 200, 201). Similarly, a low HIV DNA load was found to be predictive of better immune restoration in children on cART (202) and was associated with CD4 cell count dynamics during cART combined with interleukin-2 (203). In patients with advanced therapeutic failure and AIDS, a larger viral reservoir was associated with poorer CD4 cell recovery during optimized background therapy (197). This was confirmed by Hatano et al., who showed that high HIV DNA levels during cART were associated with low CD4 cell counts (204).
Overall, total HIV DNA loads in blood, which are influenced by the timing of cART initiation, have been linked to multiple outcomes during cART: cumulative HIV RNA viremia, which reflects therapeutic failure; residual viremia; immune recovery.
Although it is beyond the scope of this review, several studies have investigated the value of analyzing genotypic resistance on HIV DNA, which could detect archived resistant quasispecies (205–207).
In conclusion, total HIV DNA in PBMC or blood can be used as a virologic marker in patients on long-term effective cART. As observed during the natural history of HIV infection, total HIV DNA reflects the dynamics of HIV infection and the HIV reservoir during cART. It is the most easily quantifiable virologic marker in patients on cART, and several studies have shown that it is related to the history of infection and to therapeutic efficacy.
Total HIV DNA in PBMC Is Predictive of the Presence and Severity of Some HIV-Associated Disorders
HIV DNA has an important role in the pathogenesis of neurologic disorders. Indeed, total HIV DNA load in PBMC is predictive of HIV-associated dementia and correlates with the severity of HIV-associated neurocognitive disorders (208, 209). Monocyte HIV DNA load at treatment initiation can predict cognitive performance at 48 weeks (210).
TOTAL HIV DNA CAN HELP GUIDE THERAPEUTIC STRATEGIES
Biological markers are needed to evaluate strategies such as de-escalation (to reduce drug exposure and toxicity) and structured treatment interruption. Several studies have shown that HIV DNA load can be useful in this setting.
De-escalation Therapy
Baseline HIV DNA was found to be predictive of the virologic response to treatment induction with (211) or a treatment switch to (212) ritonavir-boosted protease inhibitor monotherapy. The MONARK study showed that nonresponders to first-line ritonavir-boosted lopinavir (LPV/r) monotherapy had significantly higher baseline HIV DNA levels (3.16 log10 copies/106 PBMC) than responders (2.86 log10 copies/106 PBMC) (211). Multivariate analysis of the MONOI study data set reported that the HIV DNA level at the time of a switch to ritonavir-boosted darunavir (DRV/r) monotherapy predicted the risk of viral rebound at week 96 (odds ratio, 2.66; P = 0.04) in patients with plasma HIV RNA levels of <400 copies/ml for the previous 18 months and <50 copies/ml at screening, with no history of virologic failure on a protease inhibitor-containing regimen, a CD4 lymphocyte nadir of >50 cells/mm3, no history of HIV-related neurological disease, and no hepatitis B virus coinfection (212). This was confirmed in the MONET randomized study of the switch to DRV/r monotherapy in patients on stable cART for at least 6 months with plasma HIV RNA levels of <50 copies/ml and no history of virologic failure (213). Geretti et al. showed that baseline HIV-1 DNA levels were higher in patients who had at least one HIV RNA result of >50 copies/ml during 144 weeks of follow-up (P < 0.05) (213). With the risk of virologic failure being higher during DRV/r or LPV/r monotherapy than during triple therapy (214), boosted PI monotherapy should only be proposed to carefully selected patients, and baseline HIV DNA levels might prove helpful for choosing the best candidates (213). Recent French HIV/AIDS management guidelines state that HIV DNA levels of <2.3 log copies/106 PBMC are associated with the success of such strategies (215).
Trials of de-escalation strategies with protease inhibitor-sparing regimens have led to similar results. Low baseline HIV DNA levels (below 2.35 log copies/106 PBMC) were independently associated with a lower risk of virologic failure or viral blips when a protease inhibitor was replaced by a nucleoside or nonnucleoside reverse transcriptase inhibitor (216). Another study showed that patients undergoing de-escalation with a nucleoside reverse transcriptase inhibitor dual combination had sustained an HIV RNA load of <50 copies/ml if they had received early treatment (median CD4 nadir, 340/mm3), had a low HIV RNA zenith (median, 3.9 log/ml), and had a low HIV DNA level (median, 2.5 log copies/106 PBMC) (217).
De-escalation strategies should be avoided in patients with high HIV DNA levels, as mentioned in the recent French guidelines (215). The HIV DNA level is being used for inclusion in an ongoing randomized trial of a reductive antiretroviral strategy based on nucleoside reverse transcriptase inhibitor dual combination after triple therapy (TRULIGHT) (study NCT02302547 at ClinicalTrials.gov) for instance. This study is enrolling patients with HIV DNA levels below 2.7 log copies/106 PBMC. More work is needed to identify clinically relevant total HIV DNA cutoffs for use in de-escalation strategies.
Structured Treatment Interruption
The HIV DNA load in PBMC at structured treatment interruption was the only biomarker predictive of the time to plasma HIV RNA rebound after treatment interruption, notably in the SPARTAC trial (106, 218), and also of the viral setpoint in patients first treated during PHI (218) or during CHI in the SALTO and other trials (219, 220, 221). HIV DNA load was lower after cART cessation if patients were treated within 60 days after infection, and the viral setpoint was lower in patients who were treated early (222). HIV DNA was recently reported to be the only predictor of progression following treatment interruption (106). A high HIV DNA load in PBMC at cART interruption predicted a shorter time to antiretroviral resumption, independently of the CD4 nadir (220, 221).
The total HIV DNA level in blood at the time of structured treatment interruption also predicted CD8 and CD4 T cell activation status after 12 months off treatment (187). HIV DNA levels 12 months after interruption correlated strongly with the proportion of CD38-expressing CD8 and CD4 T cells (187).
Other Therapeutic Strategies
In a phase III trial of alpha interferon administration during structured cART interruption, the probability of resuming treatment was higher among patients in the interferon arm who had a low CD4 nadir and high baseline HIV-DNA load, suggesting that interferon is less beneficial in patients with a large HIV reservoir or that the effect of treatment differs because the initial pool of HIV-infected cells is larger (223).
The impacts of therapeutic vaccine strategies can be evaluated by studying viral reservoirs in terms of total HIV DNA. In the ACTG5197 trial, Li et al. reported that a therapeutic rAd5 HIV gag vaccine had no impact on HIV DNA levels and that a higher HIV DNA load was associated with viral rebound after treatment interruption (224).
HIV DNA load could therefore be useful for evaluating therapeutic strategies.
TOTAL HIV DNA CAN BE USED TO EVALUATE TREATMENT STRATEGIES, INCLUDING THOSE TARGETING HIV RESERVOIRS
Total HIV DNA measurement could help to evaluate innovative therapeutic strategies such as intensification, stem cell transplantation, chemotherapy, immune modulation, and antilatency agents.
Different strategies have been compared for their ability to reduce total HIV DNA levels. The OPTIPRIM randomized trial, in which total HIV DNA decay was the primary endpoint, recently showed that standard triple-drug therapy reduced total HIV DNA load in PBMC as effectively as a five-drug regimen when initiated during PHI (109). No decrease in total HIV DNA levels has been observed with intensification strategies using the integrase inhibitor raltegravir, for example, in patients treated during CHI with controlled plasma viremia (42, 225). Similar declines in total HIV DNA in blood were observed in patients for whom standard triple-drug therapy or LPV/r monotherapy were initiated in the MONARK trial (211).
Stem cell transplantation for hematologic disorders can impact HIV reservoirs. The “Berlin patient,” suffering from acute myeloid leukemia, received myeloablative conditioning, two sessions of total body irradiation, and two allogeneic stem cell transplants from a donor who was homozygous for the CCR5 Delta32 deletion. His long-term control of HIV was evaluated more than 8 years after treatment interruption. HIV DNA and cell-associated HIV RNA remained undetectable in PBMC, ileum, lymph nodes, and spinal fluid, but HIV DNA was detected at low levels in the rectum and low HIV RNA signals were detected in plasma (4, 226). These signals could correspond to false positives or to defective viruses; their functional significance is unclear, as the patient had no viral rebound (226). Unfortunately, this result has not yet been reproduced.
The impact of chemotherapy on HIV DNA load in lymphoma patients receiving cART has been evaluated in a small study of 9 patients and showed no beneficial effect (227). Some types of anticancer chemotherapy can have antilatency effects (228, 229). Further studies are needed to evaluate the effects of chemotherapy on HIV reservoirs.
The effects of immune modulators such as cytokines can also be appreciated through their impact on total HIV DNA levels. The effect of interleukin-7, which enhances T cell recovery in HIV-infected patients, was evaluated for the HIV reservoir: there were no changes in the frequency of infected cells among target cells (when total HIV DNA levels were expressed as log copies per 106 PBMC or 106 CD4 T cells) at week 12. In contrast, when expressed as the log10 copies per milliliter of blood, the HIV DNA load increased significantly in individuals treated with 30 μg/kg of body weight interleukin-7 (median change, +0.51 log10 copies/μl; P = 0.006, compared with day 0 level), reflecting the augmentation of the absolute number of infected cells in the body (230, 231). Interleukin-7 induced proliferation of CD4 T cells, including latently infected cells (232). These results indicate that interleukin-7 does not modify the proportion of infected cells but increases the total number of infected cells.
Regarding antilatency agents, no decrease in total HIV DNA was observed with valproic acid, a histone deacetylase inhibitor that activates viral transcription, when added to a cART regimen (233). Similar results were recently obtained with vorinostat, another histone deacetylase inhibitor (234). Those authors concluded that, even if vorinostat triggered a significant and sustained increase in HIV transcription from latent cells in most patients, additional interventions would be needed to obtain quantitative virus production and eventually clear latently infected cells (234).
STUDIES OF HIV DNA HELP TO UNDERSTAND HIV PATHOGENESIS AND PERSISTENCE
Total HIV DNA load correlates with immune responses. In LTNPs, total HIV DNA load correlates negatively with p24-specific CD4 Th1 cell proliferation, gamma interferon (IFN-γ) production, and IFN-γ-producing cell frequencies and positively with Gag-specific IFN-γ-producing CD8 T cell frequencies (88). In untreated children, lower HIV DNA levels were associated with higher HIV-specific CD8 T lymphocyte frequencies (235) and with less abundant T cell receptor excision circles (TREC), which reflect the circulating reserve of naive T lymphocytes (236). After cART initiation, a more rapid HIV DNA decline was observed in children with higher baseline TREC levels, indicating that the pool of naive T lymphocytes influences changes in the reservoir size in patients on cART (236).
HIV DNA load has also been linked to systemic immune activation. High HIV DNA levels in patients on cART were associated with higher frequencies of CD4 T cells expressing CD38, HLA-DR, CCR5, and/or PD-1, reflecting immune activation (204). This correlation between HIV burden and cell activation was also found in the sigmoid colon of patients on cART (237). The HIV DNA level in the sigmoid colon was also positively associated with bacterial translocation (quantified in a lipopolysaccharide assay) and with poor Th17 reconstitution, reflecting mucosal barrier damage (156), both of which are key elements in HIV pathogenesis (147, 154, 157). Cytomegalovirus replication in blood and semen, participating in immune activation, was correlated with higher levels of HIV DNA in PBMC of antiretroviral-naive patients (238). This was recently confirmed; in a multivariate analysis, shedding of cytomegalovirus DNA in semen, which is associated with increased activation and proliferation of T cells in blood, was predictive of higher blood HIV DNA load (239).
Quantification of total HIV DNA in blood CD4 T lymphocyte subsets sorted by flow cytometry according to their differentiation status provided information on the viral reservoir distribution. Central memory and transitional memory CD4 T cells have been identified as the main contributors to the HIV blood reservoir in chronic patients first treated during CHI (104). These long-lived memory cells with a high proliferative capacity contribute to the stability of the HIV reservoir. On the contrary, in patients with primary HIV infection, the contributions of T cell subsets to the reservoir are different. In the context of high-level activation, effector memory T cells are the main contributors to the blood reservoir, with central memory T cells being relatively protected (240, 241). Similarly, when treatment is started during PHI, long-lived central memory cells contribute little to the HIV reservoir in comparison to more highly differentiated and shorter-lived T cells (242). cART initiated during PHI diminished the level of infection of each CD4 T cell subset but did not affect the contribution of each subset; the pattern observed at the time of PHI was similar to that seen after 2 years of early cART (240, 242). It was also similar to the pattern observed in LTNPs harboring the protective human leukocyte antigen allele B27 or B57 (243) and in posttreatment controllers (92).
The observation that the HIV reservoir resides mainly in short-lived CD4 T cells when treatment is initiated during PHI could explain the sharper reduction in total HIV DNA in patients treated during primary infection than in patients treated during CHI. Short-lived CD4 T cells have a low proliferative capacity but are able to give rise to infected daughter cells or to produce replication-competent virus that infects new cells and contributes to HIV persistence. Detection of viral clusters in effector memory CD4 T cells in blood and lymph nodes suggests that the proliferation of these cells helps to maintain the viral reservoir (244). The small fraction of central memory CD4 T cells participating in the HIV reservoir during the primary infection, or after cART initiated in PHI, have a long half-life and high proliferative capacity. They also contribute to the stability of the small reservoir in patients treated during the acute phase of infection. Moreover, central memory T cells play a major role in immune responses, and their protection against HIV infection by very early cART initiation could partly explain the clinical, immunologic, and virologic benefits of early treatment.
To conclude, early cART reduces the contribution of long-lived central memory CD4 T cells to the total HIV reservoir (240, 242), at similar levels to those observed in patients who naturally control HIV without treatment (243). Early cART has a greater impact on HIV reservoirs than later cART. However, a certain amount of total HIV DNA persists despite cART (16), even if treatment is initiated within the first 6 months after infection and is continued for >10 years. Latently infected long-lived memory CD4 T cells (central memory and T memory stem cells) persist in most early-treated individuals (57). Additional interventions will thus be required to eliminate all cells capable of producing replication-competent virus, but treatment initiation during primary infection may be the critical first step to contain HIV reservoirs (92, 245).
DISCUSSION AND CONCLUSIONS
Many studies have shown the clinical relevance of total HIV DNA load. This marker has been used most extensively in studies of HIV reservoir dynamics in both treated and untreated patients. It provides complementary information to standard markers (CD4 T cell count and HIV RNA level) and has predictive value in many settings and at different stages of HIV infection. When measured during cART, it can provide information on the pretherapeutic history (pretherapeutic HIV DNA load, plasma HIV RNA zenith, CD4 T cell nadir) and can predict several treatment outcomes, such as residual viremia, immune recovery, and cell activation status. Finally, it can help to evaluate HIV infection in tissues and body fluids and the impact of new antiretrovirals on tissue reservoirs.
Total HIV DNA load can also help with the choice of therapeutic strategies such as de-escalation, as recently acknowledged in the French guidelines (215).
Total HIV DNA levels, together with other markers, can help to identify candidates for intervention studies aimed at inducing drug-free remission (246). HIV DNA load will not be the only marker used for this purpose, as some individuals with low HIV DNA levels are unable to control the infection after treatment interruption. As each marker of HIV reservoirs provides different information, HIV DNA could be used in combination with other markers. It would also be informative to study immunologic markers of activation, inflammation, and/or T cell exhaustion. Total HIV DNA load could thus be useful for identifying patients who might respond to such cure strategies, among patients with a relatively competent immune system. Changes in reservoir size can be easily monitored by total HIV DNA assay, for example, in large clinical trials of strategies designed to eradicate HIV or to induce a functional cure. Other methods to measure changes in HIV reservoirs during such trials should be included, such as the Tat/Rev-induced limiting dilution assay (TILDA) and/or measurement of cell-associated HIV RNA, in addition to HIV DNA measurements, as some of the observed changes in HIV DNA levels could be due partly to depletion of HIV DNA forms not capable of reinitiating infection. The total HIV DNA level also helps increase understanding the pathogenesis of HIV infection, as shown by studies of CD4 T cell subsets.
One limitation of total HIV DNA as a reservoir marker is that it includes defective and more labile, unintegrated forms. However, unintegrated genomes or defective viruses might also contribute to HIV pathogenesis, by increasing the antigenic load responsible for T cell activation and exhaustion (247, 248). Several studies have shown that unintegrated HIV DNA can participate in HIV transcription and in the synthesis of viral proteins and infectious virus (44-53, 249). A recent study pointed out the role of 1-LTR forms among these unintegrated HIV DNA species (250). These synthesis activities of viral RNA and proteins are at lower levels than those produced by integrated HIV DNA forms, but they are sufficient to induce T cell activation (46). Recently, Siliciano's group showed that defective HIV proviruses can be transcribed during latency reversal (251). Replication can be blocked after this transcription, without production of viral proteins, because of insufficient transcription levels and/or mislocalization of viral RNA (22). Blockade can also occur later. O'Doherty's group recently found that infected resting CD4 cells express low levels of viral protein due to nascent LTR-driven transcription, without releasing infectious viruses, raising the possibility that reservoirs, including defective proviruses, may express HIV proteins and thus be visible to the immune system (252, 253). In untreated patients with abundant unintegrated HIV DNA, total HIV DNA loads in blood cells correlate strongly with the level of CD8 and CD4 T cell activation (187). In antiretroviral-treated adults, total HIV DNA in resting CD4 T cells is strongly associated with CD4 and CD8 T cell activation, whereas there is no association between cell activation and integrated DNA or IUPM coculture results (254). Cell-associated HIV RNA load, which reflects the level of viral transcription, including abortive transcription, also correlates with immune activation in untreated patients, patients on cART, and natural controllers (204, 255, 256, 257). In patients on cART with undetectable viremia, HIV transcript loads correlate negatively with the CD4 T cell count (257) and positively with lymphoproliferative responses to HIV p24 antigen (258). Activation stimulates transcription of persistent virus (257). These data suggest that defective proviruses might also produce antigens even if they do not produce replication-competent viruses. They could thereby play a role in HIV pathogenesis. Overall, the activity of the viral reservoir contributes to antigenic stimulation of the immune system (22). Several studies have examined the mechanisms of correlation between HIV DNA or cell-associated HIV RNA and activation and HIV pathogenesis. These mechanisms involve pattern recognition receptors: membrane-bound Toll-like receptor (TLR), cytosolic sensors like DNA-dependent activators of IFN regulatory factors, or triggered intrinsic cell defenses. HIV DNA would be particularly more likely to contribute to immune activation if it was transcribed, and especially if it was translated into viral proteins that could then be processed into peptides and presented. For example, cell-associated viral RNA interacts with TLR-7 to stimulate plasmacytoid dendritic cells, leading to α-IFN production that enhances antigen presentation (259). More recently, it was reported that incomplete HIV DNA accumulates in the cytosol before integration and activates apoptotic and inflammatory mechanisms (260, 261). These mechanisms contribute to HIV pathogenesis in infected cells unable to produce new virions (260, 261). Interferon gamma-inducible protein 16 (IFI16) serves as a link to unintegrated double-strand HIV DNA and acts as a sensor, resulting in pyroptosis and CD4 T cell depletion (261, 262). IFI16 expression correlates with CD4 T cell activation (263). HIV DNA can also interact with other sensors prior to integration (264, 265). Links between HIV DNA and activation during effective cART are less clear, but the higher the HIV DNA load, the more likely it is to partially reactivate and produce viral RNA that could contribute to inflammation. It could also induce cell proliferation by integration in specific genes (266), and these genes are likely to produce cytokines and increase activation. Finally, HIV proteins like the capsid can interact with sensors, inducing maturation of dendritic cells and innate and T immune activation, for example (267, 268). HIV DNA, HIV RNA, and proteins constitute pathogen-associated molecular patterns (PAMP) recognized by innate immune sensors and could impact activation, inflammation, and pathogenesis. This supports the utility of quantifying all forms of HIV DNA, including defective and silent forms that can be transcribed or translated without producing infectious virus. This may explain the clinical relevance of total HIV DNA.
In conclusion, the results discussed in this review show that total HIV DNA has clinical relevance as a marker of HIV reservoirs and that its level influences the course of the infection, even if it does not distinguish between replication-competent and -defective viral forms. Total HIV DNA measurement is the simplest and most sensitive, reproducible, and standardized approach for HIV reservoir measurement and can be performed routinely in clinical practice. It can be useful, together with HIV RNA load and CD4 T cell count, for adapting treatments to specific patient subgroups. It is also a promising additional biomarker for monitoring the efficacy of antiretroviral treatments and novel strategies aiming at reducing or eliminating HIV reservoirs.
ACKNOWLEDGMENTS
We thank Marine Fillion, Ludivine David, and Elise Gardiennet, who participated in all protocols and trials in the laboratory and performed total HIV DNA quantifications. We thank Asier Saez-Cirion for helpful discussions. We thank David Young for linguistic assistance.
We thank ANRS for supporting our research on HIV reservoirs.
We declare no conflict of interest with this publication.
Biographies
Véronique Avettand-Fènoël, Pharm.D., Ph.D., received her degrees at Paris Descartes University, France. She holds a Ph.D. in Virology and is currently Associate Professor in the Department of Clinical Microbiology with specialization in virology at Necker Hospital in Paris, France. Her daily work is mainly focused on virologic management of people living with HIV. She teaches at the School of Medicine, Paris-Descartes University. Her research has focused on HIV reservoirs for more than 10 years, with particular interest in the different subsets of CD4 T cells and in reservoirs in tissues such as the rectum, kidney grafts, and adipose tissue. She is involved in collaborative studies of patients at the time of primary infection, as well as long-term nonprogressors and posttreatment controllers. She is also virologic investigator in several clinical trials of early antiretroviral treatment and deescalation strategies on cell-associated HIV DNA. She takes part in the coordinated Action on HIV Reservoirs in the ANRS.
Laurent Hocqueloux, M.D., earned his medical diploma in Paris and graduated as a practitioner in Internal Medicine and Infectiology (Paris Descartes University). He is working as a physician in the Infectious Diseases Department of Orléans–La Source Regional Hospital (France). He is also a member of the AC32 Reservoir Study Group of ANRS. He has been involved in HIV/AIDS patient care for more than 15 years. His work focuses on the impact of an early antiretroviral therapy on both qualitative and quantitative aspects of viral reservoirs and immune restoration. Dr. Hocqueloux's team was the first to describe a group of HIV posttreatment controllers. He is currently the principal investigator of two studies in the field of antiretroviral de-escalation strategies. His work has provided data on the advantages of reducing viral reservoirs measured in terms of total cell-associated HIV DNA.
Jade Ghosn, M.D., Ph.D., received his medical education at Necker School of Medecine, Paris Descartes University, Paris, France. His M.D. is in Infectious Diseases, and his Ph.D. is in Virology. He is currently Associate Professor in the Department of Therapeutics and Infectiology at Hotel-Dieu University Hospital in Paris, France; his daily work is mainly focused on the clinical management of people living with HIV. He teaches at the Necker School of Medicine, Paris Descartes University. His main research interests include the establishment of HIV DNA reservoirs during primary infection, the impact of antiretroviral treatment on intracellular HIV DNA according to the timing of its initiation, and the relationship between intracellular HIV DNA in peripheral blood and HIV RNA/HIV DNA in the male genital tract. He has been working in the field of HIV since 2001.
Antoine Cheret, M.D., PhD., received his medical training at the University of Lille. He is specialized in Infectious Diseases and currently works at the Department of Internal Medicine, Kremlin-Bicêtre Hospital AP-HP, University of Paris-Sud. He holds a Ph.D. in Infectious Diseases from Paris-Descartes University. As a clinician, he has focused on the management of HIV-infected patients since 2003. His research work seeks to understand HIV reservoirs, particularly during primary HIV infection, and noninfectious comorbidities in HIV-infected patients. His particular field of expertise is the pathogenesis of HIV infection and the response to antiretroviral treatment.
Pierre Frange, M.D., Ph.D., is a graduate of Paris Sud University of Medicine (France). He trained in pediatrics with Assistance Publique-Hôpitaux de Paris (AP-HP). He also holds a Ph.D. in virology from Paris Descartes University (France). He joined the Pediatric Oncology Unit at the Curie Institute (Paris, France; 2005 to 2006), the Pediatric Immunology and Hematology Unit at Necker Hospital (Paris, France; from 2006 to 2013), and the Microbiology Laboratory at Necker Hospital (since 2013). He has experience in managing HIV-infected children and adolescents and children born to HIV-infected women. As a member of the Steering Committee of the ANRS French Perinatal Cohort, he is principal investigator and coinvestigator in several clinical trials in pediatric infectious diseases. His current research focuses on HIV reservoirs in perinatally infected children/adolescents, treatment of HIV-infected pregnant women and children, and primary HIV infection in children and adults.
Adeline Melard received her master's research degree in Infectious Diseases (Microbiology, Virology, Immunology) from Denis Diderot University (France) after fundamental virology courses at the Pasteur Institute, Paris, France. She is currently an engineer with virology laboratory EA 7327 of René Descartes University (France). She has been working on HIV, and specifically on HIV reservoirs, for 9 years, with a strong background in molecular biology. She is involved in many ANRS trials and participates in the development of new assays for laboratory studies.
Jean-Paul Viard, M.D., Ph.D., is a graduate of the Paris Est-Créteil University School of Medicine and studied immunology at the Pasteur Institute. After a residency in medicine in the Paris university hospitals (1982 to 1989), he was assistant professor (1989 to 1993), and a full-time hospital practitioner (1994 to 2011) in clinical immunology and infectious diseases at the Necker Hospital. He is head of the Immunoinfectiology Unit at Paris Hôtel-Dieu Hospital and professor of medicine (therapeutics) at Paris Descartes University (since 2011). The remarkable progress of antiretroviral treatment has transformed HIV infection into a chronic condition, but pathogenic issues remain key in the search for a cure or control of the infection. Dr. Viard participated in original, even pioneering, work linking HIV reservoirs to the clinical setting. His interest in clinical care and clinical research in the field of HIV infection dates back to the early 1990s.
Christine Rouzioux, Pharm.D., Ph.D., is Professor of Virology and responsible for the Virology Laboratory at the Necker School of Medicine. She is the Director of the EA 7327 Research Team at Paris Descartes University, Paris, France. Her research on HIV reservoirs started in 2003. She first developed a quantitative real-time PCR assay for HIV diagnosis in babies born to infected mothers, then explored the role of HIV reservoirs in clinical settings, French cohort studies, and clinical trials. She is responsible for the ANRS (French Agency on Research on AIDS and Viral Hepatitis) Coordinated Action on HIV Reservoirs.
REFERENCES
- 1.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. 1999. Latent infection of CD4+ T cells provides a mechanism for lifelong persistence of HIV-1, even in patients on effective combination therapy. Nat Med 5:512–517. doi: 10.1038/8394. [DOI] [PubMed] [Google Scholar]
- 2.Wong JK, Hezareh M, Gunthard HF, Havlir DV, Ignacio CC, Spina CA, Richman DD. 1997. Recovery of replication-competent HIV despite prolonged suppression of plasma viremia. Science 278:1291–1295. doi: 10.1126/science.278.5341.1291. [DOI] [PubMed] [Google Scholar]
- 3.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. 1997. Quantification of latent tissue reservoirs and total body viral load in HIV-1 infection. Nature 387:183–188. doi: 10.1038/387183a0. [DOI] [PubMed] [Google Scholar]
- 4.Hutter G, Nowak D, Mossner M, Ganepola S, Mussig A, Allers K, Schneider T, Hofmann J, Kucherer C, Blau O, Blau IW, Hofmann WK, Thiel E. 2009. Long-term control of HIV by CCR5 delta32/delta32 stem-cell transplantation. N Engl J Med 360:692–698. doi: 10.1056/NEJMoa0802905. [DOI] [PubMed] [Google Scholar]
- 5.Trono D, Van Lint C, Rouzioux C, Verdin E, Barre-Sinoussi F, Chun TW, Chomont N. 2010. HIV persistence and the prospect of long-term drug-free remissions for HIV-infected individuals. Science 329:174–180. doi: 10.1126/science.1191047. [DOI] [PubMed] [Google Scholar]
- 6.Lewin SR, Rouzioux C. 2011. HIV cure and eradication: how will we get from the laboratory to effective clinical trials? AIDS 25:885–897. doi: 10.1097/QAD.0b013e3283467041. [DOI] [PubMed] [Google Scholar]
- 7.Richman DD, Margolis DM, Delaney M, Greene WC, Hazuda D, Pomerantz RJ. 2009. The challenge of finding a cure for HIV infection. Science 323:1304–1307. doi: 10.1126/science.1165706. [DOI] [PubMed] [Google Scholar]
- 8.Li JZ, Smith DM, Mellors JW. 2015. The need for treatment interruption studies and biomarker identification in the search for an HIV cure. AIDS 29:1429–1432. doi: 10.1097/QAD.0000000000000658. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Eriksson S, Graf E, Dahl V, Strain MC, Yukl S, Lysenko E, Bosch RJ, Lai J, Chioma S, Emad F, Abdel-Mohsen M, Hoh R, Hecht FM, Hunt P, Somsouk M, Wong J, Johnston R, Siliciano R, Richman D, O'Doherty U, Palmer S, Deeks S, Siliciano J. 2013. Comparative analysis of measures of viral reservoirs in HIV-1 eradication studies. PLoS Pathog 9:e1003174. doi: 10.1371/journal.ppat.1003174. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Massanella M, Richman DD. 2016. Measuring the latent reservoir in vivo. J Clin Invest 126:464–472. doi: 10.1172/JCI80567. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Kiselinova M, De Spiegelaere W, Buzon MJ, Malatinkova E, Lichterfeld M, Vandekerckhove L. 2016. Integrated and total HIV-1 DNA predict ex vivo viral outgrowth. PLoS Pathog 12:e1005472. doi: 10.1371/journal.ppat.1005472. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Banga R, Procopio FA, Perreau M. 2016. Current approaches to assess HIV-1 persistence. Curr Opin HIV AIDS 11:424–431. doi: 10.1097/COH.0000000000000282. [DOI] [PubMed] [Google Scholar]
- 13.Blankson JN, Persaud D, Siliciano RF. 2002. The challenge of viral reservoirs in HIV-1 infection. Annu Rev Med 53:557–593. doi: 10.1146/annurev.med.53.082901.104024. [DOI] [PubMed] [Google Scholar]
- 14.Douek DC. 2003. Disrupting T-cell homeostasis: how HIV-1 infection causes disease. AIDS Rev 5:172–177. [PubMed] [Google Scholar]
- 15.Siliciano JD, Siliciano RF. 2005. Enhanced culture assay for detection and quantitation of latently infected, resting CD4+ T-cells carrying replication-competent virus in HIV-1-infected individuals. Methods Mol Biol 304:3–15. doi: 10.1385/1-59259-907-9:003. [DOI] [PubMed] [Google Scholar]
- 16.Chun TW, Stuyver L, Mizell SB, Ehler LA, Mican JA, Baseler M, Lloyd AL, Nowak MA, Fauci AS. 1997. Presence of an inducible HIV-1 latent reservoir during highly active antiretroviral therapy. Proc Natl Acad Sci U S A 94:13193–13197. doi: 10.1073/pnas.94.24.13193. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Ho YC, Shan L, Hosmane NN, Wang J, Laskey SB, Rosenbloom DI, Lai J, Blankson JN, Siliciano JD, Siliciano RF. 2013. Replication-competent noninduced proviruses in the latent reservoir increase barrier to HIV-1 cure. Cell 155:540–551. doi: 10.1016/j.cell.2013.09.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Cillo AR, Sobolewski MD, Bosch RJ, Fyne E, Piatak M Jr, Coffin JM, Mellors JW. 2014. Quantification of HIV-1 latency reversal in resting CD4+ T cells from patients on suppressive antiretroviral therapy. Proc Natl Acad Sci U S A 111:7078–7083. doi: 10.1073/pnas.1402873111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Siliciano JD, Kajdas J, Finzi D, Quinn TC, Chadwick K, Margolick JB, Kovacs C, Gange SJ, Siliciano RF. 2003. Long-term follow-up studies confirm the stability of the latent reservoir for HIV-1 in resting CD4+ T cells. Nat Med 9:727–728. doi: 10.1038/nm880. [DOI] [PubMed] [Google Scholar]
- 20.Crooks AM, Bateson R, Cope AB, Dahl NP, Griggs MK, Kuruc JD, Gay CL, Eron JJ, Margolis DM, Bosch RJ, Archin NM. 2015. Precise quantitation of the latent HIV-1 reservoir: implications for eradication strategies. J Infect Dis 212:1361–1365. doi: 10.1093/infdis/jiv218. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Luzuriaga K, Gay H, Ziemniak C, Sanborn KB, Somasundaran M, Rainwater-Lovett K, Mellors JW, Rosenbloom D, Persaud D. 2015. Viremic relapse after HIV-1 remission in a perinatally infected child. N Engl J Med 372:786–788. doi: 10.1056/NEJMc1413931. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Pasternak AO, Lukashov VV, Berkhout B. 2013. Cell-associated HIV RNA: a dynamic biomarker of viral persistence. Retrovirology 10:41. doi: 10.1186/1742-4690-10-41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Bukrinsky MI, Stanwick TL, Dempsey MP, Stevenson M. 1991. Quiescent T lymphocytes as an inducible virus reservoir in HIV-1 infection. Science 254:423–427. doi: 10.1126/science.1925601. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Blankson JN, Finzi D, Pierson TC, Sabundayo BP, Chadwick K, Margolick JB, Quinn TC, Siliciano RF. 2000. Biphasic decay of latently infected CD4+ T cells in acute human immunodeficiency virus type 1 infection. J Infect Dis 182:1636–1642. doi: 10.1086/317615. [DOI] [PubMed] [Google Scholar]
- 25.Persaud D, Pierson T, Ruff C, Finzi D, Chadwick KR, Margolick JB, Ruff A, Hutton N, Ray S, Siliciano RF. 2000. A stable latent reservoir for HIV-1 in resting CD4(+) T lymphocytes in infected children. J Clin Invest 105:995–1003. doi: 10.1172/JCI9006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Pierson TC, Zhou Y, Kieffer TL, Ruff CT, Buck C, Siliciano RF. 2002. Molecular characterization of preintegration latency in human immunodeficiency virus type 1 infection. J Virol 76:8518–8531. doi: 10.1128/JVI.76.17.8518-8513.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Zhou Y, Zhang H, Siliciano JD, Siliciano RF. 2005. Kinetics of human immunodeficiency virus type 1 decay following entry into resting CD4+ T cells. J Virol 79:2199–2210. doi: 10.1128/JVI.79.4.2199-2210.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Kilzer JM, Stracker T, Beitzel B, Meek K, Weitzman M, Bushman FD. 2003. Roles of host cell factors in circularization of retroviral DNA. Virology 314:460–467. doi: 10.1016/S0042-6822(03)00455-0. [DOI] [PubMed] [Google Scholar]
- 29.Yamamoto N, Tanaka C, Wu Y, Chang MO, Inagaki Y, Saito Y, Naito T, Ogasawara H, Sekigawa I, Hayashida Y. 2006. Analysis of human immunodeficiency virus type 1 integration by using a specific, sensitive and quantitative assay based on real-time polymerase chain reaction. Virus Genes 32:105–113. doi: 10.1007/s11262-005-5851-2. [DOI] [PubMed] [Google Scholar]
- 30.Chavez HH, Tran TA, Dembele B, Nasreddine N, Lambotte O, Gubler B, le Nevot E, Delfraissy JF, Taoufik Y. 2007. Lack of evidence for prolonged double-long terminal repeat episomal HIV DNA stability in vivo. J Acquir Immune Defic Syndr 45:247–249. doi: 10.1097/QAI.0b013e3180415dc2. [DOI] [PubMed] [Google Scholar]
- 31.Sharkey ME, Teo I, Greenough T, Sharova N, Luzuriaga K, Sullivan JL, Bucy RP, Kostrikis LG, Haase A, Veryard C, Davaro RE, Cheeseman SH, Daly JS, Bova C, Ellison RT III, Mady B, Lai KK, Moyle G, Nelson M, Gazzard B, Shaunak S, Stevenson M. 2000. Persistence of episomal HIV-1 infection intermediates in patients on highly active anti-retroviral therapy. Nat Med 6:76–81. doi: 10.1038/71569. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Butler SL, Johnson EP, Bushman FD. 2002. Human immunodeficiency virus cDNA metabolism: notable stability of two-long terminal repeat circles. J Virol 76:3739–3747. doi: 10.1128/JVI.76.8.3739-3747.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Butler SL, Hansen MS, Bushman FD. 2001. A quantitative assay for HIV DNA integration in vivo. Nat Med 7:631–634. doi: 10.1038/87979. [DOI] [PubMed] [Google Scholar]
- 34.Pierson TC, Kieffer TL, Ruff CT, Buck C, Gange SJ, Siliciano RF. 2002. Intrinsic stability of episomal circles formed during human immunodeficiency virus type 1 replication. J Virol 76:4138–4144. doi: 10.1128/JVI.76.8.4138-4144.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Brussel A, Mathez D, Broche-Pierre S, Lancar R, Calvez T, Sonigo P, Leibowitch J. 2003. Longitudinal monitoring of 2-long terminal repeat circles in peripheral blood mononuclear cells from patients with chronic HIV-1 infection. AIDS 17:645–652. doi: 10.1097/00002030-200303280-00001. [DOI] [PubMed] [Google Scholar]
- 36.Buzon MJ, Seiss K, Weiss R, Brass AL, Rosenberg ES, Pereyra F, Yu XG, Lichterfeld M. 2011. Inhibition of HIV-1 integration in ex vivo infected CD4 T cells from elite controllers. J Virol 85:9646–9650. doi: 10.1128/JVI.05327-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Graf EH, Mexas AM, Yu JJ, Shaheen F, Liszewski MK, Di Mascio M, Migueles SA, Connors M, O'Doherty U. 2011. Elite suppressors harbor low levels of integrated HIV DNA and high levels of 2-LTR circular HIV DNA compared to HIV+ patients on and off HAART. PLoS Pathog 7:e1001300. doi: 10.1371/journal.ppat.1001300. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Munir S, Thierry S, Subra F, Deprez E, Delelis O. 2013. Quantitative analysis of the time-course of viral DNA forms during the HIV-1 life cycle. Retrovirology 10:87. doi: 10.1186/1742-4690-10-87. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Pace MJ, Graf EH, O'Doherty U. 2013. HIV 2-long terminal repeat circular DNA is stable in primary CD4+ T cells. Virology 441:18–21. doi: 10.1016/j.virol.2013.02.028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Bushman F. 2003. Measuring covert HIV replication during HAART: the abundance of 2-LTR circles is not a reliable marker. AIDS 17:749–750. doi: 10.1097/00002030-200303280-00014. [DOI] [PubMed] [Google Scholar]
- 41.Sharkey M, Triques K, Kuritzkes DR, Stevenson M. 2005. In vivo evidence for instability of episomal human immunodeficiency virus type 1 cDNA. J Virol 79:5203–5210. doi: 10.1128/JVI.79.8.5203-5210.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Buzon MJ, Massanella M, Llibre JM, Esteve A, Dahl V, Puertas MC, Gatell JM, Domingo P, Paredes R, Sharkey M, Palmer S, Stevenson M, Clotet B, Blanco J, Martinez-Picado J. 2010. HIV-1 replication and immune dynamics are affected by raltegravir intensification of HAART-suppressed subjects. Nat Med 16:460–465. doi: 10.1038/nm.2111. [DOI] [PubMed] [Google Scholar]
- 43.Petitjean G, Al Tabaa Y, Tuaillon E, Mettling C, Baillat V, Reynes J, Segondy M, Vendrell JP. 2007. Unintegrated HIV-1 provides an inducible and functional reservoir in untreated and highly active antiretroviral therapy-treated patients. Retrovirology 4:60. doi: 10.1186/1742-4690-4-60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Cara A, Cereseto A, Lori F, Reitz MS Jr. 1996. HIV-1 protein expression from synthetic circles of DNA mimicking the extrachromosomal forms of viral DNA. J Biol Chem 271:5393–5397. doi: 10.1074/jbc.271.10.5393. [DOI] [PubMed] [Google Scholar]
- 45.Nakajima N, Lu R, Engelman A. 2001. Human immunodeficiency virus type 1 replication in the absence of integrase-mediated DNA recombination: definition of permissive and nonpermissive T-cell lines. J Virol 75:7944–7955. doi: 10.1128/JVI.75.17.7944-7955.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Gillim-Ross L, Cara A, Klotman ME. 2005. Nef expressed from human immunodeficiency virus type 1 extrachromosomal DNA downregulates CD4 on primary CD4+ T lymphocytes: implications for integrase inhibitors. J Gen Virol 86:765–771. doi: 10.1099/vir.0.80570-0. [DOI] [PubMed] [Google Scholar]
- 47.Poon B, Chang MA, Chen IS. 2007. Vpr is required for efficient Nef expression from unintegrated human immunodeficiency virus type 1 DNA. J Virol 81:10515–10523. doi: 10.1128/JVI.00947-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Kelly J, Beddall MH, Yu D, Iyer SR, Marsh JW, Wu Y. 2008. Human macrophages support persistent transcription from unintegrated HIV-1 DNA. Virology 372:300–312. doi: 10.1016/j.virol.2007.11.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Gelderblom HC, Vatakis DN, Burke SA, Lawrie SD, Bristol GC, Levy DN. 2008. Viral complementation allows HIV-1 replication without integration. Retrovirology 5:60. doi: 10.1186/1742-4690-5-60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Kantor B, Ma H, Webster-Cyriaque J, Monahan PE, Kafri T. 2009. Epigenetic activation of unintegrated HIV-1 genomes by gut-associated short chain fatty acids and its implications for HIV infection. Proc Natl Acad Sci U S A 106:18786–18791. doi: 10.1073/pnas.0905859106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Sloan RD, Wainberg MA. 2011. The role of unintegrated DNA in HIV infection. Retrovirology 8:52. doi: 10.1186/1742-4690-8-52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Thierry S, Munir S, Thierry E, Subra F, Leh H, Zamborlini A, Saenz D, Levy DN, Lesbats P, Saib A, Parissi V, Poeschla E, Deprez E, Delelis O. 2015. Integrase inhibitor reversal dynamics indicate unintegrated HIV-1 dna initiate de novo integration. Retrovirology 12:24. doi: 10.1186/s12977-015-0153-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Chan CN, Trinite B, Lee CS, Mahajan S, Anand A, Wodarz D, Sabbaj S, Bansal A, Goepfert PA, Levy DN. 2016. HIV-1 latency and virus production from unintegrated genomes following direct infection of resting CD4 T cells. Retrovirology 13:1. doi: 10.1186/s12977-015-0234-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Chun TW, Finzi D, Margolick J, Chadwick K, Schwartz D, Siliciano RF. 1995. In vivo fate of HIV-1-infected T cells: quantitative analysis of the transition to stable latency. Nat Med 1:1284–1290. doi: 10.1038/nm1295-1284. [DOI] [PubMed] [Google Scholar]
- 55.Josefsson L, King MS, Makitalo B, Brannstrom J, Shao W, Maldarelli F, Kearney MF, Hu WS, Chen J, Gaines H, Mellors JW, Albert J, Coffin JM, Palmer SE. 2011. Majority of CD4+ T cells from peripheral blood of HIV-1-infected individuals contain only one HIV DNA molecule. Proc Natl Acad Sci U S A 108:11199–11204. doi: 10.1073/pnas.1107729108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Sanchez G, Xu X, Chermann JC, Hirsch I. 1997. Accumulation of defective viral genomes in peripheral blood mononuclear cells of human immunodeficiency virus type 1-infected individuals. J Virol 71:2233–2240. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Buzon MJ, Martin-Gayo E, Pereyra F, Ouyang Z, Sun H, Li JZ, Piovoso M, Shaw A, Dalmau J, Zangger N, Martinez-Picado J, Zurakowski R, Yu XG, Telenti A, Walker BD, Rosenberg ES, Lichterfeld M. 2014. Long-term antiretroviral treatment initiated at primary HIV-1 infection affects the size, composition, and decay kinetics of the reservoir of HIV-1-infected CD4 T cells. J Virol 88:10056–10065. doi: 10.1128/JVI.01046-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Noel N, Pena R, David A, Avettand-Fenoel V, Erkizia I, Jimenez E, Lecuroux C, Rouzioux C, Boufassa F, Pancino G, Venet A, Van Lint C, Martinez-Picado J, Lambotte O, Saez-Cirion A, Prado JG. 2016. Long-term spontaneous control of HIV-1 relates to low frequency of infected cells and inefficient viral reactivation. J Virol 90:6148–6158. doi: 10.1128/JVI.00419-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Avettand-Fenoel V, Chaix ML, Blanche S, Burgard M, Floch C, Toure K, Allemon MC, Warszawski J, Rouzioux C. 2009. LTR real-time PCR for HIV-1 DNA quantitation in blood cells for early diagnosis in infants born to seropositive mothers treated in HAART area (ANRS CO 01). J Med Virol 81:217–223. doi: 10.1002/jmv.21390. [DOI] [PubMed] [Google Scholar]
- 60.Izopet J, Tamalet C, Pasquier C, Sandres K, Marchou B, Massip P, Puel J. 1998. Quantification of HIV-1 proviral DNA by a standardized colorimetric PCR-based assay. J Med Virol 54:54–59. doi:. [DOI] [PubMed] [Google Scholar]
- 61.Christopherson C, Kidane Y, Conway B, Krowka J, Sheppard H, Kwok S. 2000. PCR-based assay to quantify human immunodeficiency virus type 1 DNA in peripheral blood mononuclear cells. J Clin Microbiol 38:630–634. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Desire N, Dehee A, Schneider V, Jacomet C, Goujon C, Girard PM, Rozenbaum W, Nicolas JC. 2001. Quantification of human immunodeficiency virus type 1 proviral load by a TaqMan real-time PCR assay. J Clin Microbiol 39:1303–1310. doi: 10.1128/JCM.39.4.1303-1310.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Eriksson LE, Leitner T, Wahren B, Bostrom AC, Falk KI. 2003. A multiplex real-time PCR for quantification of HIV-1 DNA and the human albumin gene in CD4+ cells. APMIS 111:625–633. doi: 10.1034/j.1600-0463.2003.1110605.x. [DOI] [PubMed] [Google Scholar]
- 64.Lillo FB, Grasso MA, Lodini S, Bellotti MG, Colucci G. 2004. Few modifications of the Cobas Amplicor HIV Monitor 1.5 test allow reliable quantitation of HIV-1 proviral load in peripheral blood mononuclear cells. J Virol Methods 120:201–205. doi: 10.1016/j.jviromet.2004.05.008. [DOI] [PubMed] [Google Scholar]
- 65.Casabianca A, Gori C, Orlandi C, Forbici F, Federico Perno C, Magnani M. 2007. Fast and sensitive quantitative detection of HIV DNA in whole blood leucocytes by SYBR green I real-time PCR assay. Mol Cell Probes 21:368–376. doi: 10.1016/j.mcp.2007.05.005. [DOI] [PubMed] [Google Scholar]
- 66.Casabianca A, Orlandi C, Canovari B, Scotti M, Acetoso M, Valentini M, Petrelli E, Magnani M. 2014. A real time PCR platform for the simultaneous quantification of total and extrachromosomal HIV DNA forms in blood of HIV-1 infected patients. PLoS One 9:e111919. doi: 10.1371/journal.pone.0111919. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Beloukas A, Paraskevis D, Haida C, Sypsa V, Hatzakis A. 2009. Development and assessment of a multiplex real-time PCR assay for the quantification of HIV-1 DNA. J Clin Microbiol 47:2194–2199. doi: 10.1128/JCM.01264-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Hong F, Aga E, Cillo AR, Yates AL, Besson G, Fyne E, Koontz DL, Jennings C, Zheng L, Mellors JW. 2016. Novel assays for measurement of total cell-associated HIV-1 DNA and RNA. J Clin Microbiol 54:902–911. doi: 10.1128/JCM.02904-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.De Rossi A, Zanchetta M, Vitone F, Antonelli G, Bagnarelli P, Buonaguro L, Capobianchi MR, Clementi M, Abbate I, Canducci F, Monachetti A, Riva E, Rozera G, Scagnolari C, Tagliamonte M, Re MC. 2010. Quantitative HIV-1 proviral DNA detection: a multicentre analysis. New Microbiol 33:293–302. [PubMed] [Google Scholar]
- 70.Vandergeeten C, Fromentin R, Merlini E, Bramah-Lawani M, DaFonseca S, Bakeman W, McNulty A, Ramgopal M, Michael N, Kim JH, Ananworanich J, Chomont N. 2014. Cross-clade ultrasensitive PCR-based assays to measure HIV persistence in large cohort studies. J Virol 88:12385–12396. doi: 10.1128/JVI.00609-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Henrich TJ, Gallien S, Li JZ, Pereyra F, Kuritzkes DR. 2012. Low-level detection and quantitation of cellular HIV-1 DNA and 2-LTR circles using droplet digital PCR. J Virol Methods 186:68–72. doi: 10.1016/j.jviromet.2012.08.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Strain MC, Lada SM, Luong T, Rought SE, Gianella S, Terry VH, Spina CA, Woelk CH, Richman DD. 2013. Highly precise measurement of HIV DNA by droplet digital PCR. PLoS One 8:e55943. doi: 10.1371/journal.pone.0055943. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Persaud D, Patel K, Karalius B, Rainwater-Lovett K, Ziemniak C, Ellis A, Chen YH, Richman D, Siberry GK, Van Dyke RB, Burchett S, Seage GR III, Luzuriaga K. 2014. Influence of age at virologic control on peripheral blood human immunodeficiency virus reservoir size and serostatus in perinatally infected adolescents. JAMA Pediatr 168:1138–1146. doi: 10.1001/jamapediatrics.2014.1560. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Jones M, Williams J, Gartner K, Phillips R, Hurst J, Frater J. 2014. Low copy target detection by Droplet Digital PCR through application of a novel open access bioinformatic pipeline, definetherain. J Virol Methods 202:46–53. doi: 10.1016/j.jviromet.2014.02.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Luzuriaga K, Tabak B, Garber M, Chen YH, Ziemniak C, McManus MM, Murray D, Strain MC, Richman DD, Chun TW, Cunningham CK, Persaud D. 2014. HIV type 1 (HIV-1) proviral reservoirs decay continuously under sustained virologic control in HIV-1-infected children who received early treatment. J Infect Dis 210:1529–1538. doi: 10.1093/infdis/jiu297. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Bosman KJ, Nijhuis M, van Ham PM, Wensing AM, Vervisch K, Vandekerckhove L, De Spiegelaere W. 2015. Comparison of digital PCR platforms and semi-nested qPCR as a tool to determine the size of the HIV reservoir. Sci Rep 5:13811. doi: 10.1038/srep13811. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Martinez-Bonet M, Puertas MC, Fortuny C, Ouchi D, Mellado MJ, Rojo P, Noguera-Julian A, Munoz-Fernandez MA, Martinez-Picado J. 2015. Establishment and replenishment of the viral reservoir in perinatally HIV-1-infected children initiating very early antiretroviral therapy. Clin Infect Dis 61:1169–1178. doi: 10.1093/cid/civ456. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Ananworanich J, Schuetz A, Vandergeeten C, Sereti I, de Souza M, Rerknimitr R, Dewar R, Marovich M, van Griensven F, Sekaly R, Pinyakorn S, Phanuphak N, Trichavaroj R, Rutvisuttinunt W, Chomchey N, Paris R, Peel S, Valcour V, Maldarelli F, Chomont N, Michael N, Phanuphak P, Kim JH, RV254/SEARCH 010 Study Group. 2012. Impact of multi-targeted antiretroviral treatment on gut T cell depletion and HIV reservoir seeding during acute HIV infection. PLoS One 7:e33948. doi: 10.1371/journal.pone.0033948. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Ananworanich J, Sacdalan CP, Pinyakorn S, Chomont N, de Souza M, Luekasemsuk T, Schuetz A, Krebs SJ, Dewar R, Jagodzinski L, Ubolyam S, Trichavaroj R, Tovanabutra S, Spudich S, Valcour V, Sereti I, Michael N, Robb M, Phanuphak P, Kim JH, Phanuphak N. 2016. Virological and immunological characteristics of HIV-infected individuals at the earliest stage of infection. J Virus Erad 2:43–48. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Ghosn J, Deveau C, Chaix ML, Goujard C, Galimand J, Zitoun Y, Allegre T, Delfraissy JF, Meyer L, Rouzioux C. 2010. Despite being highly diverse, immunovirological status strongly correlates with clinical symptoms during primary HIV-1 infection: a cross-sectional study based on 674 patients enrolled in the ANRS CO 06 PRIMO cohort. J Antimicrob Chemother 65:741–748. doi: 10.1093/jac/dkq035. [DOI] [PubMed] [Google Scholar]
- 81.Goujard C, Bonarek M, Meyer L, Bonnet F, Chaix ML, Deveau C, Sinet M, Galimand J, Delfraissy JF, Venet A, Rouzioux C, Morlat P. 2006. CD4 cell count and HIV DNA level are independent predictors of disease progression after primary HIV type 1 infection in untreated patients. Clin Infect Dis 42:709–715. doi: 10.1086/500213. [DOI] [PubMed] [Google Scholar]
- 82.Ngo-Giang-Huong N, Deveau C, Da Silva I, Pellegrin I, Venet A, Harzic M, Sinet M, Delfraissy JF, Meyer L, Goujard C, Rouzioux C. 2001. Proviral HIV-1 DNA in subjects followed since primary HIV-1 infection who suppress plasma viral load after one year of highly active antiretroviral therapy. AIDS 15:665–673. doi: 10.1097/00002030-200104130-00001. [DOI] [PubMed] [Google Scholar]
- 83.Minga AK, Anglaret X, d' Aquin Toni T, Chaix ML, Dohoun L, Abo Y, Coulibaly A, Duvignac J, Gabillard D, Rouet F, Rouzioux C. 2008. HIV-1 DNA in peripheral blood mononuclear cells is strongly associated with HIV-1 disease progression in recently infected West African adults. J Acquir Immune Defic Syndr 48:350–354. doi: 10.1097/QAI.0b013e3181775e55. [DOI] [PubMed] [Google Scholar]
- 84.Rouzioux C, Hubert JB, Burgard M, Deveau C, Goujard C, Bary M, Sereni D, Viard JP, Delfraissy JF, Meyer L. 2005. Early levels of HIV-1 DNA in peripheral blood mononuclear cells are predictive of disease progression independently of HIV-1 RNA levels and CD4+ T cell counts. J Infect Dis 192:46–55. doi: 10.1086/430610. [DOI] [PubMed] [Google Scholar]
- 85.Kostrikis LG, Touloumi G, Karanicolas R, Pantazis N, Anastassopoulou C, Karafoulidou A, Goedert JJ, Hatzakis A. 2002. Quantitation of human immunodeficiency virus type 1 DNA forms with the second template switch in peripheral blood cells predicts disease progression independently of plasma RNA load. J Virol 76:10099–10108. doi: 10.1128/JVI.76.20.10099-10108.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Persaud D, Zhou Y, Siliciano JM, Siliciano RF. 2003. Latency in human immunodeficiency virus type 1 infection: no easy answers. J Virol 77:1659–1665. doi: 10.1128/JVI.77.3.1659-1665.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Cone RW, Gowland P, Opravil M, Grob P, Ledergerber B. 1998. Levels of HIV-infected peripheral blood cells remain stable throughout the natural history of HIV-1 infection. Swiss HIV Cohort Study. AIDS 12:2253–2260. [DOI] [PubMed] [Google Scholar]
- 88.Martinez V, Costagliola D, Bonduelle O, N′go N, Schnuriger A, Theodorou I, Clauvel JP, Sicard D, Agut H, Debre P, Rouzioux C, Autran B. 2005. Combination of HIV-1-specific CD4 Th1 cell responses and IgG2 antibodies is the best predictor for persistence of long-term nonprogression. J Infect Dis 191:2053–2063. doi: 10.1086/430320. [DOI] [PubMed] [Google Scholar]
- 89.Lambotte O, Boufassa F, Madec Y, Nguyen A, Goujard C, Meyer L, Rouzioux C, Venet A, Delfraissy JF. 2005. HIV controllers: a homogeneous group of HIV-1-infected patients with spontaneous control of viral replication. Clin Infect Dis 41:1053–1056. doi: 10.1086/433188. [DOI] [PubMed] [Google Scholar]
- 90.Sajadi MM, Heredia A, Le N, Constantine NT, Redfield RR. 2007. HIV-1 natural viral suppressors: control of viral replication in the absence of therapy. AIDS 21:517–519. doi: 10.1097/QAD.0b013e328013d9eb. [DOI] [PubMed] [Google Scholar]
- 91.Hocqueloux L, Prazuck T, Avettand-Fenoel V, Lafeuillade A, Cardon B, Viard JP, Rouzioux C. 2010. Long-term immunovirologic control following antiretroviral therapy interruption in patients treated at the time of primary HIV-1 infection. AIDS 24:1598–1601. doi: 10.1097/QAD.0b013e32833b61ba. [DOI] [PubMed] [Google Scholar]
- 92.Saez-Cirion 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, Boufassa F, Lambotte O, Goujard C, Meyer L, Costagliola D, Venet A, Pancino G, Autran B, Rouzioux C, ANRS VISCONTI Study Group. 2013. Post-treatment HIV-1 controllers with a long-term virological remission after the interruption of early initiated antiretroviral therapy. PLoS Pathog 9:e1003211. doi: 10.1371/journal.ppat.1003211. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Avettand-Fenoel V, Blanche S, Le Chenadec J, Scott-Algara D, Dollfus C, Viard JP, Bouallag N, Benmebarek Y, Riviere Y, Warszawski J, Rouzioux C, Buseyne F. 2012. Relationships between HIV disease history and blood HIV-1 DNA load in perinatally infected adolescents and young adults: the ANRS-EP38-IMMIP study. J Infect Dis 205:1520–1528. doi: 10.1093/infdis/jis233. [DOI] [PubMed] [Google Scholar]
- 94.Boulle C, Rouet F, Fassinou P, Msellati P, Debeaudrap P, Chaix ML, Rouzioux C, Avettand-Fenoel V. 2014. HIV-1 DNA concentrations and evolution among African HIV-1-infected children under antiretroviral treatment (ANRS 1244/1278). J Antimicrob Chemother 69:3047–3050. doi: 10.1093/jac/dku274. [DOI] [PubMed] [Google Scholar]
- 95.Izopet J, Salama G, Pasquier C, Sandres K, Marchou B, Massip P, Puel J. 1998. Decay of HIV-1 DNA in patients receiving suppressive antiretroviral therapy. J Acquir Immune Defic Syndr Hum Retrovirol 19:478–483. doi: 10.1097/00042560-199812150-00006. [DOI] [PubMed] [Google Scholar]
- 96.Garrigue I, Pellegrin I, Hoen B, Dumon B, Harzic M, Schrive MH, Sereni D, Fleury H. 2000. Cell-associated HIV-1-DNA quantitation after highly active antiretroviral therapy-treated primary infection in patients with persistently undetectable plasma HIV-1 RNA. AIDS 14:2851–2855. doi: 10.1097/00002030-200012220-00006. [DOI] [PubMed] [Google Scholar]
- 97.Yerly S, Perneger TV, Vora S, Hirschel B, Perrin L. 2000. Decay of cell-associated HIV-1 DNA correlates with residual replication in patients treated during acute HIV-1 infection. AIDS 14:2805–2812. doi: 10.1097/00002030-200012220-00001. [DOI] [PubMed] [Google Scholar]
- 98.Karlsson AC, Birk M, Lindback S, Gaines H, Mittler JE, Sonnerborg A. 2001. Initiation of therapy during primary HIV type 1 infection results in a continuous decay of proviral DNA and a highly restricted viral evolution. AIDS Res Hum Retroviruses 17:409–416. doi: 10.1089/088922201750102463. [DOI] [PubMed] [Google Scholar]
- 99.De Rossi A, Walker AS, De Forni D, Gibb DM. 2002. Biphasic decay of cell-associated HIV-1 DNA in HIV-1-infected children on antiretroviral therapy. AIDS 16:1961–1963. doi: 10.1097/00002030-200209270-00015. [DOI] [PubMed] [Google Scholar]
- 100.Viard JP, Burgard M, Hubert JB, Aaron L, Rabian C, Pertuiset N, Lourenco M, Rothschild C, Rouzioux C. 2004. Impact of 5 years of maximally successful highly active antiretroviral therapy on CD4 cell count and HIV-1 DNA level. AIDS 18:45–49. doi: 10.1097/00002030-200401020-00005. [DOI] [PubMed] [Google Scholar]
- 101.Strain MC, Little SJ, Daar ES, Havlir DV, Gunthard HF, Lam RY, Daly OA, Nguyen J, Ignacio CC, Spina CA, Richman DD, Wong JK. 2005. Effect of treatment, during primary infection, on establishment and clearance of cellular reservoirs of HIV-1. J Infect Dis 191:1410–1418. doi: 10.1086/428777. [DOI] [PubMed] [Google Scholar]
- 102.Morand-Joubert L, Marcellin F, Launay O, Guiramand-Hugon S, Gerard L, Yeni P, Aboulker JP. 2005. Contribution of cellular HIV-1 DNA quantification to the efficacy analysis of antiretroviral therapy: a randomized comparison of 2 regimens, including 3 drugs from 2 or 3 classes (TRIANON, ANRS 081). J Acquir Immune Defic Syndr 38:268–276. [PubMed] [Google Scholar]
- 103.Hoen B, Cooper DA, Lampe FC, Perrin L, Clumeck N, Phillips AN, Goh LE, Lindback S, Sereni D, Gazzard B, Montaner J, Stellbrink HJ, Lazzarin A, Ponscarme D, Staszewski S, Mathiesen L, Smith D, Finlayson R, Weber R, Wegmann L, Janossy G, Kinloch-de Loes S. 2007. Predictors of virological outcome and safety in primary HIV type 1-infected patients initiating quadruple antiretroviral therapy: QUEST GW PROB3005. Clin Infect Dis 45:381–390. doi: 10.1086/519428. [DOI] [PubMed] [Google Scholar]
- 104.Chomont N, El-Far M, Ancuta P, Trautmann L, Procopio FA, Yassine-Diab B, Boucher G, Boulassel MR, Ghattas G, Brenchley JM, Schacker TW, Hill BJ, Douek DC, Routy JP, Haddad EK, Sekaly RP. 2009. HIV reservoir size and persistence are driven by T cell survival and homeostatic proliferation. Nat Med 15:893–900. doi: 10.1038/nm.1972. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Jain V, Hartogensis W, Bacchetti P, Hunt PW, Hatano H, Sinclair E, Epling L, Lee TH, Busch MP, McCune JM, Pilcher CD, Hecht FM, Deeks SG. 2013. Antiretroviral therapy initiated within 6 months of HIV infection is associated with lower T-cell activation and smaller HIV reservoir size. J Infect Dis 208:1202–1211. doi: 10.1093/infdis/jit311. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Williams JP, Hurst J, Stohr 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. 2014. HIV-1 DNA predicts disease progression and post-treatment virological control. eLife 3:e03821. doi: 10.7554/eLife.03821. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Parisi SG, Andreis S, Mengoli C, Scaggiante R, Ferretto R, Manfrin V, Cruciani M, Giobbia M, Boldrin C, Basso M, Andreoni M, Palu G, Sarmati L. 2012. Baseline cellular HIV DNA load predicts HIV DNA decline and residual HIV plasma levels during effective antiretroviral therapy. J Clin Microbiol 50:258–263. doi: 10.1128/JCM.06022-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Besson GJ, Lalama CM, Bosch RJ, Gandhi RT, Bedison MA, Aga E, Riddler SA, McMahon DK, Hong F, Mellors JW. 2014. HIV-1 DNA decay dynamics in blood during more than a decade of suppressive antiretroviral therapy. Clin Infect Dis 59:1312–1321. doi: 10.1093/cid/ciu585. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Cheret A, Nembot G, Melard A, Lascoux C, Slama L, Miailhes P, Yeni P, Abel S, Avettand-Fenoel V, Venet A, Chaix ML, Molina JM, Katlama C, Goujard C, Tamalet C, Raffi F, Lafeuillade A, Reynes J, Ravaux I, Hoen B, Delfraissy JF, Meyer L, Rouzioux C. 2015. Intensive five-drug antiretroviral therapy regimen versus standard triple-drug therapy during primary HIV-1 infection (OPTIPRIM-ANRS 147): a randomised, open-label, phase 3 trial. Lancet Infect Dis 15:387–396. doi: 10.1016/S1473-3099(15)70021-6. [DOI] [PubMed] [Google Scholar]
- 110.Saitoh A, Hsia K, Fenton T, Powell CA, Christopherson C, Fletcher CV, Starr SE, Spector SA. 2002. Persistence of human immunodeficiency virus (HIV) type 1 DNA in peripheral blood despite prolonged suppression of plasma HIV-1 RNA in children. J Infect Dis 185:1409–1416. doi: 10.1086/340614. [DOI] [PubMed] [Google Scholar]
- 111.Uprety P, Chadwick EG, Rainwater-Lovett K, Ziemniak C, Luzuriaga K, Capparelli EV, Yenokyan G, Persaud D. 2015. Cell-associated HIV-1 DNA and RNA decay dynamics during early combination antiretroviral therapy in HIV-1-infected infants. Clin Infect Dis 61:1862–1870. doi: 10.1093/cid/civ688. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Vitone F, Gibellini D, Schiavone P, Re MC. 2005. Quantitative DNA proviral detection in HIV-1 patients treated with antiretroviral therapy. J Clin Virol 33:194–200. doi: 10.1016/j.jcv.2004.11.003. [DOI] [PubMed] [Google Scholar]
- 113.Kiselinova M, Geretti AM, Malatinkova E, Vervisch K, Beloukas A, Messiaen P, Bonczkowski P, Trypsteen W, Callens S, Verhofstede C, De Spiegelaere W, Vandekerckhove L. 2015. HIV-1 RNA and HIV-1 DNA persistence during suppressive ART with PI-based or nevirapine-based regimens. J Antimicrob Chemother 70:3311–3316. doi: 10.1093/jac/dkv250. [DOI] [PubMed] [Google Scholar]
- 114.Koelsch KK, Liu L, Haubrich R, May S, Havlir D, Gunthard 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. 2008. Dynamics of total, linear nonintegrated, and integrated HIV-1 DNA in vivo and in vitro. J Infect Dis 197:411–419. doi: 10.1086/525283. [DOI] [PubMed] [Google Scholar]
- 115.Murray JM, Zaunders JJ, McBride KL, Xu Y, Bailey M, Suzuki K, Cooper DA, Emery S, Kelleher AD, Koelsch KK. 2014. HIV DNA subspecies persist in both activated and resting memory CD4+ T cells during antiretroviral therapy. J Virol 88:3516–3526. doi: 10.1128/JVI.03331-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Furtado MR, Callaway DS, Phair JP, Kunstman KJ, Stanton JL, Macken CA, Perelson AS, Wolinsky SM. 1999. Persistence of HIV-1 transcription in peripheral-blood mononuclear cells in patients receiving potent antiretroviral therapy. N Engl J Med 340:1614–1622. doi: 10.1056/NEJM199905273402102. [DOI] [PubMed] [Google Scholar]
- 117.Ibanez A, Puig T, Elias J, Clotet B, Ruiz L, Martinez MA. 1999. Quantification of integrated and total HIV-1 DNA after long-term highly active antiretroviral therapy in HIV-1-infected patients. AIDS 13:1045–1049. doi: 10.1097/00002030-199906180-00007. [DOI] [PubMed] [Google Scholar]
- 118.Koelsch KK, Boesecke C, McBride K, Gelgor L, Fahey P, Natarajan V, Baker D, Bloch M, Murray JM, Zaunders J, Emery S, Cooper DA, Kelleher AD. 2011. Impact of treatment with raltegravir during primary or chronic HIV infection on RNA decay characteristics and the HIV viral reservoir. AIDS 25:2069–2078. doi: 10.1097/QAD.0b013e32834b9658. [DOI] [PubMed] [Google Scholar]
- 119.Chun TW, Engel D, Berrey MM, Shea T, Corey L, Fauci AS. 1998. Early establishment of a pool of latently infected, resting CD4(+) T cells during primary HIV-1 infection. Proc Natl Acad Sci U S A 95:8869–8873. doi: 10.1073/pnas.95.15.8869. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Calcaterra S, Cappiello G, Di Caro A, Garbuglia AR, Benedetto A. 2001. Comparative analysis of total and integrated HIV-1 DNA in peripheral CD4 lymphocytes and monocytes after long treatment with HAART. J Infect 43:239–245. doi: 10.1053/jinf.2001.0875. [DOI] [PubMed] [Google Scholar]
- 121.Yu JJ, Wu TL, Liszewski MK, Dai J, Swiggard WJ, Baytop C, Frank I, Levine BL, Yang W, Theodosopoulos T, O'Doherty U. 2008. A more precise HIV integration assay designed to detect small differences finds lower levels of integrated DNA in HAART treated patients. Virology 379:78–86. doi: 10.1016/j.virol.2008.05.030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Pires A, Hardy G, Gazzard B, Gotch F, Imami N. 2004. Initiation of antiretroviral therapy during recent HIV-1 infection results in lower residual viral reservoirs. J Acquir Immune Defic Syndr 36:783–790. doi: 10.1097/00126334-200407010-00004. [DOI] [PubMed] [Google Scholar]
- 123.Hocqueloux L, Avettand-Fenoel V, Jacquot S, Prazuck T, Legac E, Melard A, Niang M, Mille C, Le Moal G, Viard JP, Rouzioux C. 2013. Long-term antiretroviral therapy initiated during primary HIV-1 infection is key to achieving both low HIV reservoirs and normal T cell counts. J Antimicrob Chemother 68:1169–1178. doi: 10.1093/jac/dks533. [DOI] [PubMed] [Google Scholar]
- 124.Laanani M, Ghosn J, Essat A, Melard A, Seng R, Gousset M, Panjo H, Mortier E, Girard PM, Goujard C, Meyer L, Rouzioux C. 2015. Impact of the timing of initiation of antiretroviral therapy during primary HIV-1 infection on the decay of cell-associated HIV-DNA. Clin Infect Dis 60:1715–1721. doi: 10.1093/cid/civ171. [DOI] [PubMed] [Google Scholar]
- 125.Burgard M, Blanche S, Jasseron C, Descamps P, Allemon MC, Ciraru-Vigneron N, Floch C, Heller-Roussin B, Lachassinne E, Mazy F, Warszawski J, Rouzioux C. 2012. Performance of HIV-1 DNA or HIV-1 RNA tests for early diagnosis of perinatal HIV-1 infection during anti-retroviral prophylaxis. J Pediatr 160:60–6.e61. doi: 10.1016/j.jpeds.2011.06.053. [DOI] [PubMed] [Google Scholar]
- 126.Laure F, Courgnaud V, Rouzioux C, Blanche S, Veber F, Burgard M, Jacomet C, Griscelli C, Brechot C. 1988. Detection of HIV1 DNA in infants and children by means of the polymerase chain reaction. Lancet ii:538–541. [DOI] [PubMed] [Google Scholar]
- 127.Mitchell C, Dross S, Beck IA, Micek MA, Frenkel LM. 2014. Low concentrations of HIV-1 DNA at birth delays diagnosis, complicating identification of infants for antiretroviral therapy to potentially prevent the establishment of viral reservoirs. Clin Infect Dis 58:1190–1193. doi: 10.1093/cid/ciu068. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Cortes FH, Passaes CP, Bello G, Teixeira SL, Vorsatz C, Babic D, Sharkey M, Grinsztejn B, Veloso V, Stevenson M, Morgado MG. 2015. HIV controllers with different viral load cutoff levels have distinct virologic and immunologic profiles. J Acquir Immune Defic Syndr 68:377–385. doi: 10.1097/QAI.0000000000000500. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Xu H, Wang X, Malam N, Aye PP, Alvarez X, Lackner AA, Veazey RS. 2015. Persistent simian immunodeficiency virus infection drives differentiation, aberrant accumulation, and latent infection of germinal center follicular T helper cells. J Virol 90:1578–1587. doi: 10.1128/JVI.02471-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Fukazawa Y, Lum R, Okoye AA, Park H, Matsuda K, Bae JY, Hagen SI, Shoemaker R, Deleage C, Lucero C, Morcock D, Swanson T, Legasse AW, Axthelm MK, Hesselgesser J, Geleziunas R, Hirsch VM, Edlefsen PT, Piatak M Jr, Estes JD, Lifson JD, Picker LJ. 2015. B cell follicle sanctuary permits persistent productive simian immunodeficiency virus infection in elite controllers. Nat Med 21:132–139. doi: 10.1038/nm.3781. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Fletcher CV, Staskus K, Wietgrefe SW, Rothenberger M, Reilly C, Chipman JG, Beilman GJ, Khoruts A, Thorkelson A, Schmidt TE, Anderson J, Perkey K, Stevenson M, Perelson AS, Douek DC, Haase AT, Schacker TW. 2014. Persistent HIV-1 replication is associated with lower antiretroviral drug concentrations in lymphatic tissues. Proc Natl Acad Sci U S A 111:2307–2312. doi: 10.1073/pnas.1318249111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Lorenzo-Redondo R, Fryer HR, Bedford T, Kim EY, Archer J, Kosakovsky Pond SL, Chung YS, Penugonda S, Chipman JG, Fletcher CV, Schacker TW, Malim MH, Rambaut A, Haase AT, McLean AR, Wolinsky SM. 2016. Persistent HIV-1 replication maintains the tissue reservoir during therapy. Nature 530:51–56. doi: 10.1038/nature16933. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Rothenberger MK, Keele BF, Wietgrefe SW, Fletcher CV, Beilman GJ, Chipman JG, Khoruts A, Estes JD, Anderson J, Callisto SP, Schmidt TE, Thorkelson A, Reilly C, Perkey K, Reimann TG, Utay NS, Nganou Makamdop K, Stevenson M, Douek DC, Haase AT, Schacker TW. 2015. Large number of rebounding/founder HIV variants emerge from multifocal infection in lymphatic tissues after treatment interruption. Proc Natl Acad Sci U S A 112:E1126–E1134. doi: 10.1073/pnas.1414926112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Pallikkuth S, Sharkey M, Babic DZ, Gupta S, Stone GW, Fischl MA, Stevenson M, Pahwa S. 2015. Peripheral T follicular helper cells are the major HIV reservoir within central memory CD4 T cells in peripheral blood from chronically HIV-infected individuals on combination antiretroviral therapy. J Virol 90:2718–2728. doi: 10.1128/JVI.02883-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Clayton F, Snow G, Reka S, Kotler DP. 1997. Selective depletion of rectal lamina propria rather than lymphoid aggregate CD4 lymphocytes in HIV infection. Clin Exp Immunol 107:288–292. doi: 10.1111/j.1365-2249.1997.236-ce1111.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Veazey RS, DeMaria M, Chalifoux LV, Shvetz DE, Pauley DR, Knight HL, Rosenzweig M, Johnson RP, Desrosiers RC, Lackner AA. 1998. Gastrointestinal tract as a major site of CD4+ T cell depletion and viral replication in SIV infection. Science 280:427–431. doi: 10.1126/science.280.5362.427. [DOI] [PubMed] [Google Scholar]
- 137.Veazey RS, Marx PA, Lackner AA. 2001. The mucosal immune system: primary target for HIV infection and AIDS. Trends Immunol 22:626–633. doi: 10.1016/S1471-4906(01)02039-7. [DOI] [PubMed] [Google Scholar]
- 138.Poles MA, Elliott J, Taing P, Anton PA, Chen IS. 2001. A preponderance of CCR5(+) CXCR4(+) mononuclear cells enhances gastrointestinal mucosal susceptibility to human immunodeficiency virus type 1 infection. J Virol 75:8390–8399. doi: 10.1128/JVI.75.18.8390-8399.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Anton PA, Mitsuyasu RT, Deeks SG, Scadden DT, Wagner B, Huang C, Macken C, Richman DD, Christopherson C, Borellini F, Lazar R, Hege KM. 2003. Multiple measures of HIV burden in blood and tissue are correlated with each other but not with clinical parameters in aviremic subjects. AIDS 17:53–63. doi: 10.1097/00002030-200301030-00008. [DOI] [PubMed] [Google Scholar]
- 140.Guadalupe M, Reay E, Sankaran S, Prindiville T, Flamm J, McNeil A, Dandekar S. 2003. Severe CD4+ T-cell depletion in gut lymphoid tissue during primary human immunodeficiency virus type 1 infection and substantial delay in restoration following highly active antiretroviral therapy. J Virol 77:11708–11717. doi: 10.1128/JVI.77.21.11708-11717.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Brenchley JM, Schacker TW, Ruff LE, Price DA, Taylor JH, Beilman GJ, Nguyen PL, Khoruts A, Larson M, Haase AT, Douek DC. 2004. CD4+ T cell depletion during all stages of HIV disease occurs predominantly in the gastrointestinal tract. J Exp Med 200:749–759. doi: 10.1084/jem.20040874. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Veazey RS, Lackner AA. 2004. Getting to the guts of HIV pathogenesis. J Exp Med 200:697–700. doi: 10.1084/jem.20041464. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Veazey RS, Lackner AA. 2005. HIV swiftly guts the immune system. Nat Med 11:469–470. doi: 10.1038/nm0505-469. [DOI] [PubMed] [Google Scholar]
- 144.Mattapallil JJ, Douek DC, Hill B, Nishimura Y, Martin M, Roederer M. 2005. Massive infection and loss of memory CD4+ T cells in multiple tissues during acute SIV infection. Nature 434:1093–1097. doi: 10.1038/nature03501. [DOI] [PubMed] [Google Scholar]
- 145.Li Q, Duan L, Estes JD, Ma ZM, Rourke T, Wang Y, Reilly C, Carlis J, Miller CJ, Haase AT. 2005. Peak SIV replication in resting memory CD4+ T cells depletes gut lamina propria CD4+ T cells. Nature 434:1148–1152. doi: 10.1038/nature03513. [DOI] [PubMed] [Google Scholar]
- 146.Mehandru S, Tenner-Racz K, Racz P, Markowitz M. 2005. The gastrointestinal tract is critical to the pathogenesis of acute HIV-1 infection. J Allergy Clin Immunol 116:419–422. doi: 10.1016/j.jaci.2005.05.040. [DOI] [PubMed] [Google Scholar]
- 147.Brenchley JM, Price DA, Schacker TW, Asher TE, Silvestri G, Rao S, Kazzaz Z, Bornstein E, Lambotte O, Altmann D, Blazar BR, Rodriguez B, Teixeira-Johnson L, Landay A, Martin JN, Hecht FM, Picker LJ, Lederman MM, Deeks SG, Douek DC. 2006. Microbial translocation is a cause of systemic immune activation in chronic HIV infection. Nat Med 12:1365–1371. doi: 10.1038/nm1511. [DOI] [PubMed] [Google Scholar]
- 148.Brenchley JM, Price DA, Douek DC. 2006. HIV disease: fallout from a mucosal catastrophe? Nat Immunol 7:235–239. doi: 10.1038/ni1316. [DOI] [PubMed] [Google Scholar]
- 149.Dandekar S. 2007. Pathogenesis of HIV in the gastrointestinal tract. Curr HIV/AIDS Rep 4:10–15. doi: 10.1007/s11904-007-0002-0. [DOI] [PubMed] [Google Scholar]
- 150.Belmonte L, Olmos M, Fanin A, Parodi C, Bare P, Concetti H, Perez H, de Bracco MM, Cahn P. 2007. The intestinal mucosa as a reservoir of HIV-1 infection after successful HAART. AIDS 21:2106–2108. doi: 10.1097/QAD.0b013e3282efb74b. [DOI] [PubMed] [Google Scholar]
- 151.Mehandru S, Poles MA, Tenner-Racz K, Manuelli V, Jean-Pierre P, Lopez P, Shet A, Low A, Mohri H, Boden D, Racz P, Markowitz M. 2007. Mechanisms of gastrointestinal CD4+ T-cell depletion during acute and early human immunodeficiency virus type 1 infection. J Virol 81:599–612. doi: 10.1128/JVI.01739-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Nilsson J, Kinloch-de-Loes S, Granath A, Sonnerborg A, Goh LE, Andersson J. 2007. Early immune activation in gut-associated and peripheral lymphoid tissue during acute HIV infection. AIDS 21:565–574. doi: 10.1097/QAD.0b013e3280117204. [DOI] [PubMed] [Google Scholar]
- 153.Verhoeven D, Sankaran S, Dandekar S. 2007. Simian immunodeficiency virus infection induces severe loss of intestinal central memory T cells which impairs CD4+ T-cell restoration during antiretroviral therapy. J Med Primatol 36:219–227. doi: 10.1111/j.1600-0684.2007.00239.x. [DOI] [PubMed] [Google Scholar]
- 154.Brenchley JM, Paiardini M, Knox KS, Asher AI, Cervasi B, Asher TE, Scheinberg P, Price DA, Hage CA, Kholi LM, Khoruts A, Frank I, Else J, Schacker T, Silvestri G, Douek DC. 2008. Differential Th17 CD4 T-cell depletion in pathogenic and nonpathogenic lentiviral infections. Blood 112:2826–2835. doi: 10.1182/blood-2008-05-159301. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Yukl S, Wong JK. 2008. Blood and guts and HIV: preferential HIV persistence in GI mucosa. J Infect Dis 197:640–642. doi: 10.1086/527325. [DOI] [PubMed] [Google Scholar]
- 156.Chege D, Sheth PM, Kain T, Kim CJ, Kovacs C, Loutfy M, Halpenny R, Kandel G, Chun TW, Ostrowski M, Kaul R. 2011. Sigmoid Th17 populations, the HIV latent reservoir, and microbial translocation in men on long-term antiretroviral therapy. AIDS 25:741–749. doi: 10.1097/QAD.0b013e328344cefb. [DOI] [PubMed] [Google Scholar]
- 157.Douek DC, Roederer M, Koup RA. 2009. Emerging concepts in the immunopathogenesis of AIDS. Annu Rev Med 60:471–484. doi: 10.1146/annurev.med.60.041807.123549. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Avettand-Fenoel V, Prazuck T, Hocqueloux L, Melard A, Michau C, Kerdraon R, Agoute E, Rouzioux C. 2008. HIV-DNA in rectal cells is well correlated with HIV-DNA in blood in different groups of patients, including long-term non-progressors. AIDS 22:1880–1882. doi: 10.1097/QAD.0b013e32830fbdbc. [DOI] [PubMed] [Google Scholar]
- 159.Chun TW, Nickle DC, Justement JS, Meyers JH, Roby G, Hallahan CW, Kottilil S, Moir S, Mican JM, Mullins JI, Ward DJ, Kovacs JA, Mannon PJ, Fauci AS. 2008. Persistence of HIV in gut-associated lymphoid tissue despite long-term antiretroviral therapy. J Infect Dis 197:714–720. doi: 10.1086/527324. [DOI] [PubMed] [Google Scholar]
- 160.Yukl SA, Gianella S, Sinclair E, Epling L, Li Q, Duan L, Choi ALM, Girling V, Ho T, Li P, Fujimoto K, Lampiris H, Hare CB, Pandori M, Haase AT, Günthard HF, Shergill AK, McQuaid K, Havlir DV, Wong JK. 2010. Differences in HIV burden and immune activation within the gut of HIV-positive patients receiving suppressive antiretroviral therapy. J Infect Dis 202:1553–1561. doi: 10.1086/656722. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.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. 2013. 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 208:1212–1220. doi: 10.1093/infdis/jit308. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Yukl SA, Sinclair E, Somsouk M, Hunt PW, Epling L, Killian M, Girling V, Li P, Havlir DV, Deeks SG, Wong JK, Hatano H. 2014. 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 28:439–442. doi: 10.1097/QAD.0000000000000166. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Lampinen TM, Critchlow CW, Kuypers JM, Hurt CS, Nelson PJ, Hawes SE, Coombs RW, Holmes KK, Kiviat NB. 2000. Association of antiretroviral therapy with detection of HIV-1 RNA and DNA in the anorectal mucosa of homosexual men. AIDS 14:F69–F75. doi: 10.1097/00002030-200003310-00001. [DOI] [PubMed] [Google Scholar]
- 164.Anton PA, Poles MA, Elliott J, Mao SH, McGowan I, Lenz HJ, Chen IS. 2001. Sensitive and reproducible quantitation of mucosal HIV-1 RNA and DNA viral burden in patients with detectable and undetectable plasma viral HIV-1 RNA using endoscopic biopsies. J Virol Methods 95:65–79. doi: 10.1016/S0166-0934(01)00295-6. [DOI] [PubMed] [Google Scholar]
- 165.Hatano H, Somsouk M, Sinclair E, Harvill K, Gilman L, Cohen M, Hoh R, Hunt PW, Martin JN, Wong JK, Deeks SG, Yukl SA. 2013. Comparison of HIV DNA and RNA in gut-associated lymphoid tissue of HIV-infected controllers and noncontrollers. AIDS 27:2255–2260. doi: 10.1097/QAD.0b013e328362692f. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Valcour VG, Shiramizu BT, Shikuma CM. 2010. HIV DNA in circulating monocytes as a mechanism to dementia and other HIV complications. J Leukoc Biol 87:621–626. doi: 10.1189/jib.0809571. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Churchill MJ, Wesselingh SL, Cowley D, Pardo CA, McArthur JC, Brew BJ, Gorry PR. 2009. Extensive astrocyte infection is prominent in human immunodeficiency virus-associated dementia. Ann Neurol 66:253–258. doi: 10.1002/ana.21697. [DOI] [PubMed] [Google Scholar]
- 168.Zhao L, Galligan DC, Lamers SL, Yu S, Shagrun L, Salemi M, McGrath MS. 2009. High level HIV-1 DNA concentrations in brain tissues differentiate patients with post-HAART AIDS dementia complex or cardiovascular disease from those with AIDS. Sci China C Life Sci 52:651–656. doi: 10.1007/s11427-009-0085-5. [DOI] [PubMed] [Google Scholar]
- 169.Zink MC, Brice AK, Kelly KM, Queen SE, Gama L, Li M, Adams RJ, Bartizal C, Varrone J, Rabi SA, Graham DR, Tarwater PM, Mankowski JL, Clements JE. 2010. Simian immunodeficiency virus-infected macaques treated with highly active antiretroviral therapy have reduced central nervous system viral replication and inflammation but persistence of viral DNA. J Infect Dis 202:161–170. doi: 10.1086/653213. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Ball JK, Curran R, Irving WL, Dearden AA. 1999. HIV-1 in semen: determination of proviral and viral titres compared to blood, and quantification of semen leukocyte populations. J Med Virol 59:356–363. doi:. [DOI] [PubMed] [Google Scholar]
- 171.Ghosn J, Viard JP, Katlama C, de Almeida M, Tubiana R, Letourneur F, Aaron L, Goujard C, Salmon D, Leruez-Ville M, Rouzioux C, Chaix ML. 2004. Evidence of genotypic resistance diversity of archived and circulating viral strains in blood and semen of pre-treated HIV-infected men. AIDS 18:447–457. doi: 10.1097/00002030-200402200-00011. [DOI] [PubMed] [Google Scholar]
- 172.Prazuck T, Chaillon A, Avettand-Fenoel V, Caplan AL, Sayang C, Guigon A, Niang M, Barin F, Rouzioux C, Hocqueloux L. 2013. HIV-DNA in the genital tract of women on long-term effective therapy is associated to residual viremia and previous AIDS-defining illnesses. PLoS One 8:e69686. doi: 10.1371/journal.pone.0069686. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Canaud G, Dejucq-Rainsford N, Avettand-Fenoel V, Viard JP, Anglicheau D, Bienaime F, Muorah M, Galmiche L, Gribouval O, Noel LH, Satie AP, Martinez F, Sberro-Soussan R, Scemla A, Gubler MC, Friedlander G, Antignac C, Timsit MO, Onetti Muda A, Terzi F, Rouzioux C, Legendre C. 2014. The kidney as a reservoir for HIV-1 after renal transplantation. J Am Soc Nephrol 25:407–419. doi: 10.1681/ASN.2013050564. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174.Canaud G, Avettand-Fenoel V, Legendre C. 2015. HIV-positive-to-HIV-positive kidney transplantation. N Engl J Med 372:2069. doi: 10.1056/NEJMc1503288. [DOI] [PubMed] [Google Scholar]
- 175.Zanussi S, Bortolin MT, Pratesi C, Tedeschi R, Basaglia G, Abbruzzese L, Mazzucato M, Spina M, Vaccher E, Tirelli U, Rupolo M, Michieli M, Di Mascio M, De Paoli P. 2015. Autograft HIV-DNA load predicts HIV-1 peripheral reservoir after stem cell transplantation for AIDS-related lymphoma patients. AIDS Res Hum Retroviruses 31:150–159. doi: 10.1089/aid.2014.0157. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Couturier J, Suliburk JW, Brown JM, Luke DJ, Agarwal N, Yu X, Nguyen C, Iyer D, Kozinetz CA, Overbeek PA, Metzker ML, Balasubramanyam A, Lewis DE. 2015. Human adipose tissue as a reservoir for memory CD4+ T cells and HIV. AIDS 29:667–674. doi: 10.1097/QAD.0000000000000599. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.Damouche A, Lazure T, Avettand-Fenoel V, Huot N, Dejucq-Rainsford N, Satie AP, Melard A, David L, Gommet C, Ghosn J, Noel N, Pourcher G, Martinez V, Benoist S, Bereziat V, Cosma A, Favier B, Vaslin B, Rouzioux C, Capeau J, Muller-Trutwin M, Dereuddre-Bosquet N, Le Grand R, Lambotte O, Bourgeois C. 2015. Adipose tissue is a neglected viral reservoir and an inflammatory site during chronic HIV and SIV infection. PLoS Pathog 11:e1005153. doi: 10.1371/journal.ppat.1005153. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178.Hatzakis AE, Touloumi G, Pantazis N, Anastassopoulou CG, Katsarou O, Karafoulidou A, Goedert JJ, Kostrikis LG. 2004. Cellular HIV-1 DNA load predicts HIV-RNA rebound and the outcome of highly active antiretroviral therapy. AIDS 18:2261–2267. doi: 10.1097/00002030-200411190-00006. [DOI] [PubMed] [Google Scholar]
- 179.Tierney C, Lathey JL, Christopherson C, Bettendorf DM, D'Aquila RT, Hammer SM, Katzenstein DA. 2003. Prognostic value of baseline human immunodeficiency virus type 1 DNA measurement for disease progression in patients receiving nucleoside therapy. J Infect Dis 187:144–148. doi: 10.1086/345870. [DOI] [PubMed] [Google Scholar]
- 180.Katzenstein TL, Oliveri RS, Benfield T, Eugen-Olsen J, Nielsen C, Gerstoft J. 2002. Cell-associated HIV DNA measured early during infection has prognostic value independent of serum HIV RNA measured concomitantly. Scand J Infect Dis 34:529–533. doi: 10.1080/00365540110080845. [DOI] [PubMed] [Google Scholar]
- 181.Madec Y, Boufassa F, Rouzioux C, Delfraissy JF, Meyer L. 2005. Undetectable viremia without antiretroviral therapy in patients with HIV seroconversion: an uncommon phenomenon? Clin Infect Dis 40:1350–1354. doi: 10.1086/429318. [DOI] [PubMed] [Google Scholar]
- 182.Madec Y, Boufassa F, Avettand-Fenoel V, Hendou S, Melard A, Boucherit S, Surzyn J, Meyer L, Rouzioux C. 2009. Early control of HIV-1 infection in long-term nonprogressors followed since diagnosis in the ANRS SEROCO/HEMOCO cohort. J Acquir Immune Defic Syndr 50:19–26. doi: 10.1097/QAI.0b013e31818ce709. [DOI] [PubMed] [Google Scholar]
- 183.Tsiara CG, Nikolopoulos GK, Bagos PG, Goujard C, Katzenstein TL, Minga AK, Rouzioux C, Hatzakis A. 2012. Impact of HIV type 1 DNA levels on spontaneous disease progression: a meta-analysis. AIDS Res Hum Retroviruses 28:366–373. doi: 10.1089/aid.2011.0032. [DOI] [PubMed] [Google Scholar]
- 184.McDermott JL, Giri AA, Martini I, Bono M, Giacomini M, Campelli A, Tagliaferro L, Cara A, Varnier OE. 1999. Level of human immunodeficiency virus DNA in peripheral blood mononuclear cells correlates with efficacy of antiretroviral therapy. J Clin Microbiol 37:2361–2365. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185.Havlir DV, Strain MC, Clerici M, Ignacio C, Trabattoni D, Ferrante P, Wong JK. 2003. Productive infection maintains a dynamic steady state of residual viremia in human immunodeficiency virus type 1-infected persons treated with suppressive antiretroviral therapy for five years. J Virol 77:11212–11219. doi: 10.1128/JVI.77.20.11212-11219.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186.Havlir DV, Koelsch KK, Strain MC, Margot N, Lu B, Ignacio CC, Miller MD, Wong JK. 2005. Predictors of residual viremia in HIV-infected patients successfully treated with efavirenz and lamivudine plus either tenofovir or stavudine. J Infect Dis 191:1164–1168. doi: 10.1086/428588. [DOI] [PubMed] [Google Scholar]
- 187.Weiss L, Chevalier MF, Assoumou L, Didier C, Girard PM, Piketty C, Costagliola D, Rouzioux C. 2014. T-cell activation positively correlates with cell-associated HIV-DNA level in viremic patients with primary or chronic HIV-1 infection. AIDS 28:1683–1687. doi: 10.1097/QAD.0000000000000319. [DOI] [PubMed] [Google Scholar]
- 188.Bortolin MT, Zanussi S, Talamini R, Simonelli C, Pratesi C, Tedeschi R, Abbruzzese L, Manuele R, Rupolo M, Tirelli U, De Paoli P. 2010. Predictive value of HIV type 1 DNA levels on overall survival in HIV-related lymphoma patients treated with high-dose chemotherapy (HDC) plus autologous stem cell transplantation (ASCT). AIDS Res Hum Retroviruses 26:245–251. doi: 10.1089/aid.2009.0081. [DOI] [PubMed] [Google Scholar]
- 189.Burgard M, Boufassa F, Viard JP, Garrigue I, Ruffault A, Izopet J, Vabret A, Descamps D, Colson P, Seigneurin JM, Rouzioux C. 2009. Factors influencing peripheral blood mononuclear cell-associated HIV-1 DNA level after long-term suppressive antiretroviral therapy in 236 patients. AIDS 23:2165–2171. doi: 10.1097/QAD.0b013e32833032d4. [DOI] [PubMed] [Google Scholar]
- 190.Lambert-Niclot S, Flandre P, Valantin MA, Soulie C, Fourati S, Wirden M, Sayon S, Pakianather S, Bocket L, Masquelier B, Dos Santos G, Katlama C, Calvez V, Marcelin AG. 2012. Similar evolution of cellular HIV-1 DNA level in darunavir/ritonavir monotherapy versus triple therapy in MONOI-ANRS136 trial over 96 weeks. PLoS One 7:e41390. doi: 10.1371/journal.pone.0041390. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191.Fourati S, Flandre P, Calin R, Carcelain G, Soulie C, Lambert-Niclot S, Maiga A, Ait-Arkoub Z, Tubiana R, Valantin MA, Autran B, Katlama C, Calvez V, Marcelin AG. 2014. Factors associated with a low HIV reservoir in patients with prolonged suppressive antiretroviral therapy. J Antimicrob Chemother 69:753–756. doi: 10.1093/jac.dkt428. [DOI] [PubMed] [Google Scholar]
- 192.Cuzin L, Pugliese P, Saune K, Allavena C, Ghosn J, Cottalorda J, Rodallec A, Chaix ML, Fafi-Kremer S, Soulie C, Ouka M, Charpentier C, Bocket L, Mirand A, Guiguet M. 2015. Levels of intracellular HIV-DNA in patients with suppressive antiretroviral therapy. AIDS 29:1665–1671. doi: 10.1097/QAD.0000000000000723. [DOI] [PubMed] [Google Scholar]
- 193.Rodriguez-Sainz C, Ramos R, Valor L, Lopez F, Santamaria B, Hernandez DC, Cruz JS, Navarro J, Modrego J, Alecsandru D, Fernandez-Cruz E. 2010. Prognostic value of peripheral blood mononuclear cell-associated HIV-1 DNA for virological outcome in asymptomatic HIV-1 chronic infection. J Clin Virol 48:168–172. doi: 10.1016/j.jcv.2010.03.020. [DOI] [PubMed] [Google Scholar]
- 194.Sarmati L, Parisi SG, Nicastri E, d'Ettorre G, Palmisano L, Andreotti M, Andreoni C, Giuliano M, Gatti F, Boldrin C, Palu G, Vullo V, Vella S, Andreoni M. 2005. Association between cellular human immunodeficiency virus DNA level and immunological parameters in patients with undetectable plasma viremia level during highly active antiretroviral therapy. J Clin Microbiol 43:6183–6185. doi: 10.1128/JCM.43.12.6183-6185.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 195.Chun TW, Murray D, Justement JS, Hallahan CW, Moir S, Kovacs C, Fauci AS. 2011. Relationship between residual plasma viremia and the size of HIV proviral DNA reservoirs in infected individuals receiving effective antiretroviral therapy. J Infect Dis 204:135–138. doi: 10.1093/infdis/jir208. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 196.Guihot A, Tubiana R, Breton G, Marcelin AG, Samri A, Assoumou L, Goncalves E, Bricaire F, Costagliola D, Calvez V, Rouzioux C, Autran B, Katlama C, Carcelain G. 2010. Immune and virological benefits of 10 years of permanent viral control with antiretroviral therapy. AIDS 24:614–617. doi: 10.1097/QAD.0b013e32833556f3. [DOI] [PubMed] [Google Scholar]
- 197.Avettand-Fenoel V, Bouteloup V, Melard A, Fagard C, Chaix ML, Leclercq P, Chene G, Viard JP, Rouzioux C. 2010. Higher HIV-1 DNA associated with lower gains in CD4 cell count among patients with advanced therapeutic failure receiving optimized treatment (ANRS 123-ETOILE). J Antimicrob Chemother 65:2212–2214. doi: 10.1093/jac/dkq282. [DOI] [PubMed] [Google Scholar]
- 198.Soulie C, Marcelin AG, Ghosn J, Amellal B, Assoumou L, Lambert S, Duvivier C, Costagliola D, Katlama C, Calvez V. 2007. HIV-1 X4/R5 co-receptor in viral reservoir during suppressive HAART. AIDS 21:2243–2245. doi: 10.1097/QAD.0b013e3282f0e3d0. [DOI] [PubMed] [Google Scholar]
- 199.Ghosn J, Leruez-Ville M, Blanche J, Delobelle A, Beaudoux C, Mascard L, Lecuyer H, Canestri A, Landman R, Zucman D, Ponscarme D, Rami A, Viard JP, Spire B, Rouzioux C, Costagliola D, Suzan-Monti M. 2014. HIV-1 DNA levels in peripheral blood mononuclear cells and cannabis use are associated with intermittent HIV shedding in semen of men who have sex with men on successful antiretroviral regimens. Clin Infect Dis 58:1763–1770. doi: 10.1093/cid/ciu187. [DOI] [PubMed] [Google Scholar]
- 200.Chun TW, Justement JS, Pandya P, Hallahan CW, McLaughlin M, Liu S, Ehler LA, Kovacs C, Fauci AS. 2002. Relationship between the size of the human immunodeficiency virus type 1 (HIV-1) reservoir in peripheral blood CD4+ T cells and CD4+:CD8+ T cell ratios in aviremic HIV-1-infected individuals receiving long-term highly active antiretroviral therapy. J Infect Dis 185:1672–1676. doi: 10.1086/340521. [DOI] [PubMed] [Google Scholar]
- 201.Ostrowski SR, Katzenstein TL, Thim PT, Pedersen BK, Gerstoft J, Ullum H. 2005. Low-level viremia and proviral DNA impede immune reconstitution in HIV-1-infected patients receiving highly active antiretroviral therapy. J Infect Dis 191:348–357. doi: 10.1086/427340. [DOI] [PubMed] [Google Scholar]
- 202.Ometto L, De Forni D, Patiri F, Trouplin V, Mammano F, Giacomet V, Giaquinto C, Douek D, Koup R, De Rossi A. 2002. Immune reconstitution in HIV-1-infected children on antiretroviral therapy: role of thymic output and viral fitness. AIDS 16:839–849. doi: 10.1097/00002030-200204120-00003. [DOI] [PubMed] [Google Scholar]
- 203.Levy Y, Thiebaut R, Gougeon ML, Molina JM, Weiss L, Girard PM, Venet A, Morlat P, Poirier B, Lascaux AS, Boucherie C, Sereni D, Rouzioux C, Viard JP, Lane C, Delfraissy JF, Sereti I, Chene G. 2012. Effect of intermittent interleukin-2 therapy on CD4+ T-cell counts following antiretroviral cessation in patients with HIV. AIDS 26:711–720. doi: 10.1097/QAD.0b013e3283519214. [DOI] [PubMed] [Google Scholar]
- 204.Hatano H, Jain V, Hunt PW, Lee TH, Sinclair E, Do TD, Hoh R, Martin JN, McCune JM, Hecht F, Busch MP, Deeks SG. 2013. Cell-based measures of viral persistence are associated with immune activation and programmed cell death protein 1 (PD-1)-expressing CD4+ T cells. J Infect Dis 208:50–56. doi: 10.1093/infdis/jis630. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 205.Gantner P, Morand-Joubert L, Raffi F, Sueur C, Fagard C, Lascoux-Combe C, Salmon D, Amiel C, Lambert-Niclot S, Fofana D, Viard JP, Fafi-Kremer S, Rouzioux C, Avettand-Fenoel V, Ghosn J. 2016. Drug resistance and tropism as markers of the dynamics of HIV-1 DNA quasispecies in blood cells of heavily pretreated patients who achieved sustained virological suppression. J Antimicrob Chemother 71:751–761. doi: 10.1093/jac/dkv395. [DOI] [PubMed] [Google Scholar]
- 206.Delaugerre C, Braun J, Charreau I, Delarue S, Nere ML, de Castro N, May T, Marchou B, Simon F, Molina JM, Aboulker JP. 2012. Comparison of resistance mutation patterns in historical plasma HIV RNA genotypes with those in current proviral HIV DNA genotypes among extensively treated patients with suppressed replication. HIV Med 13:517–525. doi: 10.1111/j.1468-1293.2012.01002.x. [DOI] [PubMed] [Google Scholar]
- 207.Wirden M, Soulie C, Valantin MA, Fourati S, Simon A, Lambert-Niclot S, Bonmarchand M, Clavel-Osorio C, Marcelin AG, Katlama C, Calvez V. 2011. Historical HIV-RNA resistance test results are more informative than proviral DNA genotyping in cases of suppressed or residual viraemia. J Antimicrob Chemother 66:709–712. doi: 10.1093/jac/dkq544. [DOI] [PubMed] [Google Scholar]
- 208.Shiramizu B, Paul R, Williams A, Shikuma C, Watters M, Grove J, Valcour V. 2007. HIV proviral DNA associated with decreased neuropsychological function. J Neuropsychiatry Clin Neurosci 19:157–163. doi: 10.1176/jnp.2007.19.2.157. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 209.Shiramizu B, Williams AE, Shikuma C, Valcour V. 2009. Amount of HIV DNA in peripheral blood mononuclear cells is proportional to the severity of HIV-1-associated neurocognitive disorders. J Neuropsychiatry Clin Neurosci 21:68–74. doi: 10.1176/jnp.2009.21.1.68. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 210.Valcour VG, Shiramizu BT, Sithinamsuwan P, Nidhinandana S, Ratto-Kim S, Ananworanich J, Siangphoe U, Kim JH, de Souza M, Degruttola V, Paul RH, Shikuma CM. 2009. HIV DNA and cognition in a Thai longitudinal HAART initiation cohort: the SEARCH 001 Cohort Study. Neurology 72:992–998. doi: 10.1212/01.wnl.0000344404.12759.83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 211.Avettand-Fenoel V, Flandre P, Chaix ML, Ghosn J, Delaugerre C, Raffi F, Ngovan P, Cohen-Codar I, Delfraissy JF, Rouzioux C. 2010. Impact of 48 week lopinavir/ritonavir monotherapy on blood cell-associated HIV-1-DNA in the MONARK trial. J Antimicrob Chemother 65:1005–1007. doi: 10.1093/jac/dkq084. [DOI] [PubMed] [Google Scholar]
- 212.Lambert-Niclot S, Flandre P, Valantin MA, Peytavin G, Duvivier C, Haim-Boukobza S, Algarte-Genin M, Yazdanpanah Y, Girard PM, Katlama C, Calvez V, Marcelin AG. 2011. Factors associated with virological failure in HIV-1-infected patients receiving darunavir/ritonavir monotherapy. J Infect Dis 204:1211–1216. doi: 10.1093/infdis/jir518. [DOI] [PubMed] [Google Scholar]
- 213.Geretti AM, Arribas JR, Lathouwers E, Foster GM, Yakoob R, Kinloch S, Hill A, van Delft Y, Moecklinghoff C. 2013. Dynamics of cellular HIV-1 DNA levels over 144 weeks of darunavir/ritonavir monotherapy versus triple therapy in the MONET trial. HIV Clin Trials 14:45–50. doi: 10.1310/hct1401-45. [DOI] [PubMed] [Google Scholar]
- 214.Perez-Valero I, Arribas JR. 2011. Protease inhibitor monotherapy. Curr Opin Infect Dis 24:7–11. doi: 10.1097/QCO.0b013e3283422cdf. [DOI] [PubMed] [Google Scholar]
- 215.Morlat P, Groupe des Experts Prise en Charge Médicale des Personnes Infectées par le VIH . 2015. Prise en charge médicale des personnes vivant avec le VIH. Actualisation 2015 du rapport 2013 CNS/ANRS, Paris, France: http://cns.sante.fr/wp-content/uploads/2015/10/experts-vih_actualisation2015.pdf. [Google Scholar]
- 216.Sarmati L, Parisi SG, Nicastri E, d'Ettorre G, Andreoni C, Dori L, Gatti F, Montano M, Buonomini AR, Boldrin C, Palu G, Vullo V, Andreoni M. 2007. Cellular HIV-1 DNA quantitation in patients during simplification therapy with protease inhibitor-sparing regimens. J Med Virol 79:880–886. doi: 10.1002/jmv.20914. [DOI] [PubMed] [Google Scholar]
- 217.Prazuck T, Zucman D, Avettand-Fenoel V, Ducasse E, Bornarel D, Mille C, Rouzioux C, Hocqueloux L. 2013. Long-term HIV-1 virologic control in patients on a dual NRTI regimen. HIV Clin Trials 14:120–126. doi: 10.1310/hct1403-120. [DOI] [PubMed] [Google Scholar]
- 218.Lafeuillade A, Poggi C, Hittinger G, Counillon E, Emilie D. 2003. Predictors of plasma human immunodeficiency virus type 1 RNA control after discontinuation of highly active antiretroviral therapy initiated at acute infection combined with structured treatment interruptions and immune-based therapies. J Infect Dis 188:1426–1432. doi: 10.1086/379251. [DOI] [PubMed] [Google Scholar]
- 219.Yerly S, Gunthard HF, Fagard C, Joos B, Perneger TV, Hirschel B, Perrin L. 2004. Proviral HIV-DNA predicts viral rebound and viral setpoint after structured treatment interruptions. AIDS 18:1951–1953. doi: 10.1097/00002030-200409240-00011. [DOI] [PubMed] [Google Scholar]
- 220.Piketty C, Weiss L, Assoumou L, Burgard M, Melard A, Ragnaud JM, Bentata M, Girard PM, Rouzioux C, Costagliola D. 2010. A high HIV DNA level in PBMCs at antiretroviral treatment interruption predicts a shorter time to treatment resumption, independently of the CD4 nadir. J Med Virol 82:1819–1828. doi: 10.1002/jmv.21907. [DOI] [PubMed] [Google Scholar]
- 221.Assoumou L, Weiss L, Piketty C, Burgard M, Melard A, Girard PM, Rouzioux C, Costagliola D. 2015. A low HIV-DNA level in peripheral blood mononuclear cells at antiretroviral treatment interruption predicts a higher probability of maintaining viral control. AIDS 29:2003–2007. doi: 10.1097/QAD.0000000000000734. [DOI] [PubMed] [Google Scholar]
- 222.Gianella S, von Wyl V, Fischer M, Niederoest B, Battegay M, Bernasconi E, Cavassini M, Rauch A, Hirschel B, Vernazza P, Weber R, Joos B, Gunthard HF. 2011. Effect of early antiretroviral therapy during primary HIV-1 infection on cell-associated HIV-1 DNA and plasma HIV-1 RNA. Antivir Ther 16:535–545. doi: 10.3851/IMP1776. [DOI] [PubMed] [Google Scholar]
- 223.Boue F, Reynes J, Rouzioux C, Emilie D, Souala F, Tubiana R, Goujard C, Lancar R, Costagliola D. 2011. Alpha interferon administration during structured interruptions of combination antiretroviral therapy in patients with chronic HIV-1 infection: INTERVAC ANRS 105 trial. AIDS 25:115–118. doi: 10.1097/QAD.0b013e328340a1e7. [DOI] [PubMed] [Google Scholar]
- 224.Li JZ, Heisey A, Ahmed H, Wang H, Zheng L, Carrington M, Wrin T, Schooley RT, Lederman MM, Kuritzkes DR. 2014. Relationship of HIV reservoir characteristics with immune status and viral rebound kinetics in an HIV therapeutic vaccine study. AIDS 28:2649–2657. doi: 10.1097/QAD.0000000000000478. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 225.Delaugerre C, Charreau I, Braun J, Nere ML, de Castro N, Yeni P, Ghosn J, Aboulker JP, Molina JM, Simon F. 2010. Time course of total HIV-1 DNA and 2-long-terminal repeat circles in patients with controlled plasma viremia switching to a raltegravir-containing regimen. AIDS 24:2391–2395. doi: 10.1097/QAD.0b013e32833d214c. [DOI] [PubMed] [Google Scholar]
- 226.Yukl SA, Boritz E, Busch M, Bentsen C, Chun TW, Douek D, Eisele E, Haase A, Ho YC, Hutter G, Justement JS, Keating S, Lee TH, Li P, Murray D, Palmer S, Pilcher C, Pillai S, Price RW, Rothenberger M, Schacker T, Siliciano J, Siliciano R, Sinclair E, Strain M, Wong J, Richman D, Deeks SG. 2013. Challenges in detecting HIV persistence during potentially curative interventions: a study of the Berlin patient. PLoS Pathog 9:e1003347. doi: 10.1371/journal.ppat.1003347. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 227.Cillo AR, Krishnan S, McMahon DK, Mitsuyasu RT, Para MF, Mellors JW. 2014. Impact of chemotherapy for HIV-1 related lymphoma on residual viremia and cellular HIV-1 DNA in patients on suppressive antiretroviral therapy. PLoS One 9:e92118. doi: 10.1371/journal.pone.0092118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 228.Bouchat S, Delacourt N, Kula A, Darcis G, Van Driessche B, Corazza F, Gatot JS, Melard A, Vanhulle C, Kabeya K, Pardons M, Avettand-Fenoel V, Clumeck N, De Wit S, Rohr O, Rouzioux C, Van Lint C. 2015. Sequential treatment with 5-aza-2′-deoxycytidine and deacetylase inhibitors reactivates HIV-1. EMBO Mol Med 8:117–138. doi: 10.15252/emmm.201505557. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 229.Darcis G, Kula A, Bouchat S, Fujinaga K, Corazza F, Ait-Ammar A, Delacourt N, Melard A, Kabeya K, Vanhulle C, Van Driessche B, Gatot JS, Cherrier T, Pianowski LF, Gama L, Schwartz C, Vila J, Burny A, Clumeck N, Moutschen M, De Wit S, Peterlin BM, Rouzioux C, Rohr O, Van Lint C. 2015. An in-depth comparison of latency-reversing agent combinations in various in vitro and ex vivo HIV-1 latency models identified bryostatin-1 + JQ1 and Ingenol-B + JQ1 to potently reactivate viral gene expression. PLoS Pathog 11:e1005063. doi: 10.1371/journal.ppat.1005063. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 230.Levy Y, Lacabaratz C, Weiss L, Viard JP, Goujard C, Lelievre JD, Boue F, Molina JM, Rouzioux C, Avettand-Fenoel V, Croughs T, Beq S, Thiebaut R, Chene G, Morre M, Delfraissy JF. 2009. Enhanced T cell recovery in HIV-1-infected adults through IL-7 treatment. J Clin Invest 119:997–1007. doi: 10.1172/JCI38052. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 231.Levy Y, Sereti I, Tambussi G, Routy JP, Lelievre JD, Delfraissy JF, Molina JM, Fischl M, Goujard C, Rodriguez B, Rouzioux C, Avettand-Fenoel V, Croughs T, Beq S, Morre M, Poulin JF, Sekaly RP, Thiebaut R, Lederman MM. 2012. Effects of recombinant human interleukin 7 on T-cell recovery and thymic output in HIV-infected patients receiving antiretroviral therapy: results of a phase I/IIa randomized, placebo-controlled, multicenter study. Clin Infect Dis 55:291–300. doi: 10.1093/cid/cis383. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 232.Vandergeeten C, Fromentin R, DaFonseca S, Lawani MB, Sereti I, Lederman MM, Ramgopal M, Routy JP, Sekaly RP, Chomont N. 2013. Interleukin-7 promotes HIV persistence during antiretroviral therapy. Blood 121:4321–4329. doi: 10.1182/blood-2012-11-465625. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 233.Sagot-Lerolle N, Lamine A, Chaix ML, Boufassa F, Aboulker JP, Costagliola D, Goujard C, Pallier C, Delfraissy JF, Lambotte O. 2008. Prolonged valproic acid treatment does not reduce the size of latent HIV reservoir. AIDS 22:1125–1129. doi: 10.1097/QAD.0b013e3282fd6ddc. [DOI] [PubMed] [Google Scholar]
- 234.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, Sekaly RP, Lewin SR. 2014. Activation of HIV transcription with short-course vorinostat in HIV-infected patients on suppressive antiretroviral therapy. PLoS Pathog 10:e1004473. doi: 10.1371/journal.ppat.1004473. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 235.Scott-Algara D, Rouzioux C, Blanche S, Burgard M, Didier C, Riviere Y, Buseyne F. 2010. In untreated HIV-1-infected children, PBMC-associated HIV DNA levels and cell-free HIV RNA levels are correlated to distinct T-lymphocyte populations. J Acquir Immune Defic Syndr 53:553–563. doi: 10.1097/QAI.0b013e3181cf060r. [DOI] [PubMed] [Google Scholar]
- 236.De Rossi A, Walker AS, De Forni D, Klein N, Gibb DM. 2005. Relationship between changes in thymic emigrants and cell-associated HIV-1 DNA in HIV-1-infected children initiating antiretroviral therapy. Antivir Ther 10:63–71. [PubMed] [Google Scholar]
- 237.Sheth PM, Chege D, Shin LY, Huibner S, Yue FY, Loutfy M, Halpenny R, Persad D, Kovacs C, Chun TW, Kandel G, Ostrowski M, Kaul R. 2008. Immune reconstitution in the sigmoid colon after long-term HIV therapy. Mucosal Immunol 1:382–388. doi: 10.1038/mi.2008.23. [DOI] [PubMed] [Google Scholar]
- 238.Gianella S, Anderson CM, Vargas MV, Richman DD, Little SJ, Morris SR, Smith DM. 2013. Cytomegalovirus DNA in semen and blood is associated with higher levels of proviral HIV DNA. J Infect Dis 207:898–902. doi: 10.1093/infdis/jis777. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 239.Gianella S, Massanella M, Richman DD, Little SJ, Spina CA, Vargas MV, Lada SM, Daar ES, Dube MP, Haubrich RH, Morris SR, Smith DM. 2014. Cytomegalovirus replication in semen is associated with higher levels of proviral HIV DNA and CD4+ T cell activation during antiretroviral treatment. J Virol 88:7818–7827. doi: 10.1128/JVI.00831-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 240.Bacchus C, Cheret A, Avettand-Fenoel V, Nembot G, Melard A, Blanc C, Lascoux-Combe C, Slama L, Allegre T, Allavena C, Yazdanpanah Y, Duvivier C, Katlama C, Goujard C, Seksik BC, Leplatois A, Molina JM, Meyer L, Autran B, Rouzioux C. 2013. A single HIV-1 cluster and a skewed immune homeostasis drive the early spread of HIV among resting CD4+ cell subsets within one month post-infection. PLoS One 8:e64219. doi: 10.1371/journal.pone.0064219. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 241.Ganesan A, Chattopadhyay PK, Brodie TM, Qin J, Gu W, Mascola JR, Michael NL, Follmann DA, Roederer M. 2010. Immunologic and virologic events in early HIV infection predict subsequent rate of progression. J Infect Dis 201:272–284. doi: 10.1086/649430. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 242.Cheret A, Bacchus-Souffan C, Avettand-Fenoel V, Melard A, Nembot G, Blanc C, Samri A, Saez-Cirion A, Hocqueloux L, Lascoux-Combe C, Allavena C, Goujard C, Valantin MA, Leplatois A, Meyer L, Rouzioux C, Autran B. 2015. Combined ART started during acute HIV infection protects central memory CD4+ T cells and can induce remission. J Antimicrob Chemother 70:2108–2120. doi: 10.1093/jac/dkv084. [DOI] [PubMed] [Google Scholar]
- 243.Descours B, Avettand-Fenoel V, Blanc C, Samri A, Melard A, Supervie V, Theodorou I, Carcelain G, Rouzioux C, Autran B. 2012. Immune responses driven by protective human leukocyte antigen alleles from long-term nonprogressors are associated with low HIV reservoir in central memory CD4 T cells. Clin Infect Dis 54:1495–1503. doi: 10.1093/cid/cis188. [DOI] [PubMed] [Google Scholar]
- 244.von Stockenstrom S, Odevall L, Lee E, Sinclair E, Bacchetti P, Killian M, Epling L, Shao W, Hoh R, Ho T, Faria NR, Lemey P, Albert J, Hunt P, Loeb L, Pilcher C, Poole L, Hatano H, Somsouk M, Douek D, Boritz E, Deeks SG, Hecht FM, Palmer S. 2015. Longitudinal genetic characterization reveals that cell proliferation maintains a persistent HIV type 1 DNA pool during effective HIV therapy. J Infect Dis 212:596–607. doi: 10.1093/infdis/jiv092. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 245.Ananworanich J, Dube K, Chomont N. 2015. How does the timing of antiretroviral therapy initiation in acute infection affect HIV reservoirs? Curr Opin HIV AIDS 10:18–28. doi: 10.1097/COH.0000000000000122. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 246.Ghosn J, Delaugerre C. 2015. Can we avoid treatment interruption studies in the search for an HIV cure? AIDS 29:1575–1577. doi: 10.1097/QAD.0000000000000763. [DOI] [PubMed] [Google Scholar]
- 247.Stevenson M, Haggerty S, Lamonica CA, Meier CM, Welch SK, Wasiak AJ. 1990. Integration is not necessary for expression of human immunodeficiency virus type 1 protein products. J Virol 64:2421–2425. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 248.Appay V, Sauce D. 2008. Immune activation and inflammation in HIV-1 infection: causes and consequences. J Pathol 214:231–241. doi: 10.1002/path.2276. [DOI] [PubMed] [Google Scholar]
- 249.Shimura K, Miyazato P, Oishi S, Fujii N, Matsuoka M. 2015. Impact of HIV-1 infection pathways on susceptibility to antiviral drugs and on virus spread. Virology 484:364–376. doi: 10.1016/j.virol.2015.06.029. [DOI] [PubMed] [Google Scholar]
- 250.Thierry S, Thierry E, Subra F, Deprez E, Leh H, Bury-Mone S, Delelis O. 2016. Opposite transcriptional regulation of integrated vs unintegrated HIV genomes by the NF-κB pathway. Sci Rep 6:25678. doi: 10.1038/srep25678. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 251.Ho Y-C, Pollack R, Yong P, Siliciano RF. 2015. Defective HIV-1 proviruses can be transcribed upon activation, abstr 392. Conf Retroviruses Opportunistic Infect 2015, Seattle, WA. [Google Scholar]
- 252.Pace MJ, Graf EH, Agosto LM, Mexas AM, Male F, Brady T, Bushman FD, O'Doherty U. 2012. Directly infected resting CD4+T cells can produce HIV Gag without spreading infection in a model of HIV latency. PLoS Pathog 8:e1002818. doi: 10.1371/journal.ppat.1002818. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 253.DeMaster LK, Liu X, VanBelzen DJ, Trinite B, Zheng L, Agosto LM, Migueles SA, Connors M, Sambucetti L, Levy DN, Pasternak AO, O'Doherty U. 2016. A subset of CD4/CD8 double-negative T cells expresses HIV proteins in patients on antiretroviral therapy. J Virol 90:2165–2179. doi: 10.1128/JVI.01913-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 254.Cockerham LR, Siliciano JD, Sinclair E, O'Doherty U, Palmer S, Yukl SA, Strain MC, Chomont N, Hecht FM, Siliciano RF, Richman DD, Deeks SG. 2014. CD4+ and CD8+ T cell activation are associated with HIV DNA in resting CD4+ T cells. PLoS One 9:e110731. doi: 10.1371/journal.pone.0110731. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 255.Soares RS, Tendeiro R, Foxall RB, Baptista AP, Cavaleiro R, Gomes P, Camacho R, Valadas E, Doroana M, Lucas M, Antunes F, Victorino RM, Sousa AE. 2011. Cell-associated viral burden provides evidence of ongoing viral replication in aviremic HIV-2-infected patients. J Virol 85:2429–2438. doi: 10.1128/JVI.01921-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 256.Hunt PW, Hatano H, Sinclair E, Lee TH, Busch MP, Martin JN, McCune JM, Deeks SG. 2011. HIV-specific CD4+ T cells may contribute to viral persistence in HIV controllers. Clin Infect Dis 52:681–687. doi: 10.1093/cid/ciq202. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 257.Ishizaka A, Sato H, Nakamura H, Koga M, Kikuchi T, Hosoya N, Koibuchi T, Nomoto A, Kawana-Tachikawa A, Mizutani T. 2016. Short intracellular HIV-1 transcripts as biomarkers of residual immune activation in patients on antiretroviral therapy. J Virol 90:5665–5674. doi: 10.1128/JVI.03158-415. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 258.Patterson BK, McCallister S, Schutz M, Siegel JN, Shults K, Flener Z, Landay A. 2001. Persistence of intracellular HIV-1 mRNA correlates with HIV-1-specific immune responses in infected subjects on stable HAART. AIDS 15:1635–1641. doi: 10.1097/00002030-200109070-00005. [DOI] [PubMed] [Google Scholar]
- 259.Beignon AS, McKenna K, Skoberne M, Manches O, DaSilva I, Kavanagh DG, Larsson M, Gorelick RJ, Lifson JD, Bhardwaj N. 2005. Endocytosis of HIV-1 activates plasmacytoid dendritic cells via Toll-like receptor-viral RNA interactions. J Clin Invest 115:3265–3275. doi: 10.1172/JCI26032. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 260.Doitsh G, Cavrois M, Lassen KG, Zepeda O, Yang Z, Santiago ML, Hebbeler AM, Greene WC. 2010. Abortive HIV infection mediates CD4 T cell depletion and inflammation in human lymphoid tissue. Cell 143:789–801. doi: 10.1016/j.cell.2010.11.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 261.Monroe KM, Yang Z, Johnson JR, Geng X, Doitsh G, Krogan NJ, Greene WC. 2014. IFI16 DNA sensor is required for death of lymphoid CD4 T cells abortively infected with HIV. Science 343:428–432. doi: 10.1126/science.1243640. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 262.Jakobsen MR, Bak RO, Andersen A, Berg RK, Jensen SB, Tengchuan J, Laustsen A, Hansen K, Ostergaard L, Fitzgerald KA, Xiao TS, Mikkelsen JG, Mogensen TH, Paludan SR. 2013. IFI16 senses DNA forms of the lentiviral replication cycle and controls HIV-1 replication. Proc Natl Acad Sci U S A 110:E4571–E4580. doi: 10.1073/pnas.1311669110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 263.Nissen SK, Hojen JF, Andersen KL, Kofod-Olsen E, Berg RK, Paludan SR, Ostergaard L, Jakobsen MR, Tolstrup M, Mogensen TH. 2014. Innate DNA sensing is impaired in HIV patients and IFI16 expression correlates with chronic immune activation. Clin Exp Immunol 177:295–309. doi: 10.1111/cei.12317. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 264.Gao D, Wu J, Wu YT, Du F, Aroh C, Yan N, Sun L, Chen ZJ. 2013. Cyclic GMP-AMP synthase is an innate immune sensor of HIV and other retroviruses. Science 341:903–906. doi: 10.1126/science.1240933. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 265.Landau NR. 2014. The innate immune response to HIV-1: to sense or not to sense. DNA Cell Biol 33:271–274. doi: 10.1089/dna.2014.2378. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 266.Wagner TA, McLaughlin S, Garg K, Cheung CY, Larsen BB, Styrchak S, Huang HC, Edlefsen PT, Mullins JI, Frenkel LM. 2014. HIV latency. Proliferation of cells with HIV integrated into cancer genes contributes to persistent infection. Science 345:570–573. doi: 10.1126/science.1256304. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 267.Manel N, Hogstad B, Wang Y, Levy DE, Unutmaz D, Littman DR. 2010. A cryptic sensor for HIV-1 activates antiviral innate immunity in dendritic cells. Nature 467:214–217. doi: 10.1038/nature09337. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 268.Henrick BM, Yao XD, Rosenthal KL. 2015. HIV-1 structural proteins serve as PAMPs for TLR2 heterodimers significantly increasing infection and innate immune activation. Front Immunol 6:426. doi: 10.3389/fimmu.2015.00426. [DOI] [PMC free article] [PubMed] [Google Scholar]