Rapid Biphasic Decay of Intact and Defective HIV DNA Reservoir During Acute Treated HIV Disease =============================================================================================== * Alton Barbehenn * Lei Shi * Junzhe Shao * Rebecca Hoh * Heather M. Hartig * Vivian Pae * Sannidhi Sarvadhavabhatla * Sophia Donaire * Caroline Sheikhzadeh * Jeffrey Milush * Gregory M. Laird * Mignot Mathias * Kristen Ritter * Michael Peluso * Jeffrey Martin * Frederick Hecht * Christopher Pilcher * Stephanie E. Cohen * Susan Buchbinder * Diane Havlir * Monica Gandhi * Timothy J. Henrich * Hiroyu Hatano * Jingshen Wang * Steven G. Deeks * Sulggi A. Lee ## ABSTRACT Antiretroviral therapy (ART) is not a cure. Upon ART cessation, virus rapidly rebounds from latently-infected cells (“the HIV reservoir”). The reservoir is largely stabilized at the time of ART initiation and then decays slowly. Here, leveraging >500 longitudinal samples from 67 people with HIV (PWH) treated during acute infection, we developed a novel mathematical model to predict reservoir decay using the intact proviral DNA assay (IPDA) from peripheral CD4+ T cells. Nonlinear generalized additive models adjusted for initial CD4+ T count, pre-ART viral load, and timing of ART initiation demonstrated rapid biphasic decay of intact DNA (week 0-5: t1/2 ∼0.71 months; week 5-24: t1/2 ∼3.9 months) that extended out to 1 year of ART, with similar trends for defective DNA. Predicted reservoir decay were faster for participants individuals with earlier timing of ART initiation, higher initial CD4+ T cell count, and lower pre-ART viral load. These estimates are ∼5-fold faster than prior reservoir decay estimates among chronic-treated PWH. Thus, these data add to our limited understanding of host viral control at the earliest stages of HIV reservoir stabilization, potentially informing future HIV cure efforts aimed at diverse, global population of PWH initiating ART at varying stages of disease. Keywords * acute HIV * antiretroviral therapy (ART) * HIV reservoir decay * pre-exposure prophylaxis (PrEP) ## INTRODUCTION Despite over four decades of research, we do not yet have an HIV cure. While antiretroviral therapy (ART) is able to suppress virus to undetectable levels, virus rapidly rebounds from latently-infected cells (“the HIV reservoir”) within weeks of ART interruption.1–8 Thus, a major goal is to eradicate and/or accelerate the decay of the reservoir in order to achieve clinical remission. However, HIV cure trials to date have largely failed to demonstrate a clinically meaningful reduction in the size of the HIV reservoir and/or lead to sustained ART-free remission.9–12 The majority of these trials have been conducted among people with HIV (PWH) treated during chronic infection and were administered therapeutic interventions long after the reservoir was established (i.e., several years after initiating ART)13–18. Recent combination therapy trials (e.g., broadly neutralizing antibodies given with ART) have yielded more promising results, and a few participants have demonstrated evidence of extended ART-free viral control19–21, but the mechanisms by which these small number of participants exhibit enhanced viral control remain unclear. One possibility is that since PWH treated during chronic HIV often have larger reservoirs22–29 and exhausted/dysfunctional immune responses30–32 (due to prolonged periods of untreated HIV infection), that demonstrating measurable HIV remission from these small trials may have been more challenging. Indeed, individuals who initiate ART “earlier” (within 6 months after infection) may be more likely to become “post-treatment controllers” (PTCs), rare PWH who, for unclear reasons, are able to control virus without ART following a brief of initial ART suppression.33 Thus, different host factors, such as timing of ART initiation in relation to chronicity of HIV infection, initial CD4+ T cell count, or pre-ART HIV viral load, may have profound impact on estimating HIV reservoir decay rates, and yet there are limited data modeling decay rates while accounting for these factors. Such information would be immensely useful in future HIV cure trials to compare expected reservoir decay rates in comparison to curative interventions and allow generalizability of findings to a more global population of PWH (treated during acute or chronic infection). While there have now been a handful of studies modeling how quickly the HIV reservoir decays during prolonged ART (studies spanning nearly 20 years on ART)34–37, there have been no studies modeling HIV reservoir decay rates after acute treated HIV infection. Here, leveraging ∼500 longitudinal blood measurements of the HIV reservoir, we developed a novel mathematical model of reservoir decay among 67 participants from the UCSF Treat Acute HIV cohort initiating ART within 100 days of the estimated date of HIV infection.38 These data demonstrate for the first time that the reservoir decays significantly faster in acute compared to chronic treated PWH initiating ART.3 Our partially parameterized mathematical model flexibly estimated the rate of change of the HIV reservoir to include participant-level random effects (to account for heterogeneity in decay rates) as well as key clinical covariates (initial CD4+ T count, pre-ART viral load, and days since estimated date of HIV infection). We fit various mono-, bi-, and triphasic decay curves for both HIV “intact” (infected cells harboring intact viral sequences able to produce infectious virions) and “defective” (the majority of the HIV reservoir but incapable of producing infectious virions) DNA. For both intact and defective DNA, we observed clear biphasic decay patterns with an initial rapid first phase of decay (which was nearly identical to previous estimates in chronic treated PWH initiating ART3) followed by a second slower phase of decay (∼5-fold faster that previously reported in chronic PWH initiating ART). Furthermore, these decay rates were significantly faster among individuals with known clinical factors associated with enhanced host viral control: higher initial CD4+ T cell count, earlier initiation of ART, and lower pre-ART viral loads.22,23,34,39,40 These data are the first to our knowledge to describe a predictive mathematical model to describe HIV reservoir decay rates during acute treated HIV. ## RESULTS ### Characteristics of study participants A total of 67 adults (83% of those screened) with a new diagnosis of acute HIV (<100 days between the estimated date of detected HIV infection to the baseline study visit/ART initiation) were included in the UCSF Treat Acute HIV cohort (**Table 1, Supplementary Figure 1**) 41. All 67 participants completed follow-up in the study for the full 24 weeks. A large proportion (65.7%) of participants co-enrolled in our longitudinal UCSF SCOPE HIV cohort and remained in study for additional follow-up, with study visits approximately every 3-4 months thereafter. Follow-up for the current analysis included longitudinal sampling for a median of 0.81 years. The estimated dates of detected infection (EDDI) were calculated to determine recency of infection using the Infection Dating Tool ([https://tools.incidence-estimation.org/idt/](https://tools.incidence-estimation.org/idt/)) 42 (**Supplementary Figure 2**). These estimates were then used to estimate acute HIV Fiebig stages 43,44. Consistent with our San Francisco-based study population, participants were mostly male (97%) and reflected local and national racial/ethnic trends of higher incident acute HIV in these populations 45. Baseline study visit HIV-1 antigen/antibody (Architect) and HIV-1 antibody (Geenius) testing demonstrated 27% and 28% false negative/indeterminate rates, respectively, consistent with our San Francisco Department of Public Health (SFDPH) reported estimates for new acute HIV diagnoses 46 (**Supplementary Figure 4**). Genotype data (Monogram) were available for a subset of 57 participants; 77% had wild-type HIV, 9% had M184V/I mutations (all were reported among participants citing prior and/or current PrEP use), and 14% had evidence of possible partner-transmitted resistance mutations (based on referral of newly diagnosed partners within our cohort and/or SFDPH partner tracing 46). View this table: [Table 1.](http://medrxiv.org/content/early/2024/03/28/2024.03.27.24304867/T1) Table 1. Baseline Characteristics of the UCSF Treat Acute Study Population Medians with interquartile ranges and frequencies with percentages are shown. Our cohort also reflected a high proportion of self-reported prior PrEP use (42% reporting ever use while 20% reported PrEP use in the past 10 days), reflecting San Francisco’s early and widespread adoption of PrEP 46. All PrEP reported in this study was oral PrEP with tenofovir disoproxil fumarate/emtricitabine (TDF/FTC), as this was the only form clinically available during the study period. Among individuals reporting overlapping PrEP use within 10 days from their estimated date of HIV acquisition (using self-reported PrEP use and the infection dating tool as described in the Methods 42), six participants were deemed to have probable HIV acquisition while on PrEP (median baseline plasma log10HIV RNA = 2.2 copies/mL), which was approximately ∼3 log10 lower than those not reporting PrEP overlap (**Table 1**). One participant was estimated to have acquired HIV in the setting of therapeutic PrEP concentrations (confirmed by plasma and hair ART concentrations), as described in our previous publication 47. ### Rapid biphasic decay of intact and defective DNA We estimated the rate of change of HIV intact and defective DNA as log10copies per 106 peripheral CD4+ T cells (quantified using the IPDA), as well as the influence of clinical covariates on the rate of HIV DNA change. A median of 2 x 106 CD4+ T cells per sample (median viability of 97%) was recovered from peripheral CD4+ T cells isolated by negative immunomagnetic selection. Duration of ART was used as the primary predictor to allow for time-varying estimation of changes within each person across the sampling timepoints. The rate of change was estimated for the primary outcome of HIV intact DNA, as well as the secondary outcome of defective DNA (3’ defective, 5’ defective, and total combined defective provirus). We initially modeled HIV intact and defective DNA using a linear effect of time on ART (which assumes a constant rate of change regardless of the duration of viral suppression). However, we observed evidence of nonlinearity, and thus, instead of fitting a single constant rate of decline, we fit nonlinear generalized additive models to estimate the appropriate nonparametric relationship for each predictor term. Clinical covariates of age, gender, pre-ART CD4+ T cell count, pre-ART viral load, and timing of ART initiation (number of weeks since estimated date of infection) were tested for inclusion in these models. Final models included terms for initial CD4+ T cell count, pre-ART viral load, and timing of ART initiation. **Figure 4** illustrates that of the three clinical covariates, only timing of ART initiation changes the shape of the reservoir decay, whereas initial CD4+ T cell count and pre-ART viral load account for patient-level variation. Additionally, we added individual participant-level random effects into regression models to account for heterogeneity in outcome measures within individuals (see **Statistical Methods**). Given the small number of participants reporting PrEP use within 10 days of HIV acquisition (**Table 1**), inclusion of this parameter as a covariate in the model and sensitivity analyses excluding these individuals did not change results, and thus was not included as a covariate in final models. In addition, participants reporting potential PrEP overlap did not clearly fall in the lower range of reservoir measurements, but rather, were dispersed among the range of HIV intact and defective DNA values (**Supplementary Figure 5**). ![Figure 1.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2024/03/28/2024.03.27.24304867/F1.medium.gif) [Figure 1.](http://medrxiv.org/content/early/2024/03/28/2024.03.27.24304867/F1) Figure 1. Fitted models match observed HIV reservoir patterns for HIV intact and defective DNA The decay patterns for each participant are shown as thin grey lines, while the decay pattern for the model-predicted average participant is shown as the thick black lines. Biphasic decay patters were observed for HIV intact (left panel) and total combined (3’ plus 5’) defective (right panel) (a). For defective HIV DNA, the biphasic decay pattern was observed more strongly for 5’ but also seen with 3’ defective DNA (b). Average predicted participant predictions were made by taking the mean of *Ei* (estimated time between HIV infection and ART initiation), *ci* (initial CD4+ T cell count), and *vi* (log10 pre-ART plasma viral load) across participants from our final model. ![Figure 2.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2024/03/28/2024.03.27.24304867/F2.medium.gif) [Figure 2.](http://medrxiv.org/content/early/2024/03/28/2024.03.27.24304867/F2) Figure 2. An inflection point (*τ = 5* weeks) was chosen from a grid from 0 weeks to 24 weeks, by half-weeks Results are shown for both intact (a) and defective (b) HIV DNA decay models, respectively. The biphasic decay model’s inflection point was tuned by estimating the *τ* that minimized prediction error measured by leave-one-out mean absolute error (MAE, upper panels) and leave-one-out mean squared error (MSE, lower panels). The best *τ* for each loss (red point) is shown in relation to e final model’s selected inflection point of *τ = 5* weeks as reference (vertical dashed line). ![Figure 3.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2024/03/28/2024.03.27.24304867/F3.medium.gif) [Figure 3.](http://medrxiv.org/content/early/2024/03/28/2024.03.27.24304867/F3) Figure 3. Lower pre-ART viral load and higher initial CD4+ T counts are associated with lower HIV DNA reservoirs in fitted spline models Fitted spline models with corresponding 95% confidence intervals for HIV intact (a) and defective (b) DNA plotted against log10 pre-ART viral load (left panel) and iitial CD4+ T cell count (right panel). The fitted spline models are plotted in red with corresponding 95% confidence intervals denoted by blue dashed lines. Lower intact HIV DNA reservoirs (a) were sociated with lower pre-ART viral load and higher initial CD4+ T counts. For example, a participant with an initial CD4+ T cell count of 900 cells/mm3 was predicted to have ∼10 times smaller HIV intact reservoir than a participant with an initial CD4+ T cell count of 300 cells/mm3. Similar patterns were observed for HIV defective DNA, but the fitted splines were less linear (b). ![Figure 4.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2024/03/28/2024.03.27.24304867/F4.medium.gif) [Figure 4.](http://medrxiv.org/content/early/2024/03/28/2024.03.27.24304867/F4) Figure 4. Clinical factors influence HIV intact and defective DNA decay rates Faster decay rates were observed among individuals with earlier ART initiation (a), higher CD4+ T cell count (b), and lower pre-ART viral load (c). Left and right panels show decay rates for HIV intact and defective DNA, respectively. Individual participant decay rates shown in thin black lines, and cohort medians shown by bold black solid or dashed lines. Therefore, we then fit various mono-, bi-, and triphasic decay curves for both HIV intact and defective DNA using semiparametric generalized additive models that introduced up to two inflection points, tau (*τ*), delineating distinct exponential decay phases (see **Statistical Methods**). We then estimated the decay model’s inflection points *τ* by minimizing prediction errors as measured by leave-one-out mean absolute deviation (MAD) and leave-one-out mean squared error (MSE) approaches (**Figure 2, Supplementary Figure 7**). For HIV intact DNA, we found that the biphasic model best fit our data, while for defective DNA, the biphasic and triphasic models were relatively equivalent in performance (**Supplementary Figure 6**). A biphasic model outperformed a monophasic decay model for both intact and defective DNA. These analyses demonstrated that *τ* = 5 weeks closely fit our data using this biphasic model. To ensure that the selected *τ* = 5 was not influenced by potential outlier data, we performed three different sensitivity analyses excluding (1) individuals reporting prior PrEP use within 10 days of estimated date of HIV infection, (2) participants with plasma viral load “blips” (defined as a one-time viral load >1000 copies/mL or two consecutive viral loads >100 copies/mL between weeks 0-24), and (3) participants with sudden increases in HIV intact DNA (defined as >50% increase between two consecutive measurements of HIV intact DNA during weeks 0-24). These sensitivity analyses demonstrated that *τ* = 5 is a reasonable choice for the mode l’s inflection point and that the model estimates were unchanged after excluding these potential outliers. Based on our final selected multivariate model, we observed a rapid biphasic decay of both intact and defective DNA during acute treated HIV infection, with an initial steep decay until approximately 5 weeks of ART, followed by a slower decay (**Figure 1a**). For example, for HIV intact DNA, we estimated that the first phase of reservoir decay had an estimated half-life (t1/2) ∼0.70 (95% confidence interval [CI] = 0.59, 0.82) months in unadjusted models and a t1/2 ∼0.71 (95%CI = 0.60, 0.82) months in models adjusted for clinical covariates. This was followed by a slower second decay phase after ∼5 weeks of ART with a t1/2 ∼3.61 (95%CI = 2.81, 4.42) months in unadjusted models and a t1/2 ∼3.86 (95%CI = 2.99, 4.72) months in models adjusted for clinical covariates. Similar first phase (weeks 0-5) decay rates were observed for HIV defective DNA in unadjusted (t1/2 ∼0.34, 95% CI = 0.29, 0.38) and adjusted (t1/2 ∼0.34, 95% CI = 0.29, 0.39) models but estimates were less stable for the second phase of defective DNA decay due to considerable variability in these data (**Figure 1, Supplementary Tables 1-2**). Thus, we did not observe discernable differences in decay rates for HIV intact versus defective DNA in the first five weeks after ART initiation, in contrast to prior reservoir decay studies among chronic treated PWH during nearly 20 years of ART34–36,48, although we did see a discernable differences in decay rates for HIV intact versus defective DNA from weeks 5-24 that seems to continue out to a year (**Supplementary Figure 7**). For all models, fitted values closely predicted observed values for both intact and defective DNA (**Supplementary Figure 10**). We further validated our models using our clinical data to determine whether our model captured expected clinical relationships. Our model tells us that for every additional week ART initiation is delayed, the half-life for the first phase of HIV intact reservoir decay increases by ∼0.02 months; similarly, the defective reservoir’s half-life increases by ∼0.01 months. Importantly, our model tells us that the timing of ART initiation has a larger impact on the second phase of HIV reservoir decay. During weeks 5-24, our model predicts an increase in intact reservoir decay half-life of ∼0.27 months for every week delay in ART initiation and no significant change to the defective HIV reservoir (**Supplementary Table 3**). Concretely, this means that a patient who initiates ART 30 days post infection has an average intact HIV reservoir decay half-life of ∼2.5 months while a patient who initiates ART 100 days post infection has an average intact HIV reservoir decay half-life of ∼6.3 months; as suggested in **Supplementary Figure 7**, this may continue out to a year on ART. Clinical validation of the initial CD4+ T cell count and pre-ART viral load terms is more straightforward because they are only used to account for participant-level variation, not changes in half-life. Our model tells us that both lower pre-ART viral load and higher initial CD4+ T counts were associated with lower intact and defective HIV DNA reservoirs (**Supplementary Table 4**). Finally, using bootstrapping methods (random sampling of the clinical covariates to compute model predictions for 300 new participants) we estimated HIV DNA intact and defective counts and then averaged the model predictions within strata (tertiles) of each clinical parameter (e.g., average HIV intact DNA decay rate for the first, second, and third tertiles of pre-ART viral loads (**Figure 5**). These analyses clearly demonstrated that our model predicts lower HIV intact and defective DNA reservoirs for individuals with higher initial CD4+ T cell count and lower pre-ART viral load, and the HIV intact and defective DNA reservoirs decayed more rapidly for earlier timing of ART initiation. ![](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2024/03/28/2024.03.27.24304867/F5/graphic-11.medium.gif) [](http://medrxiv.org/content/early/2024/03/28/2024.03.27.24304867/F5/graphic-11) ![](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2024/03/28/2024.03.27.24304867/F5/graphic-12.medium.gif) [](http://medrxiv.org/content/early/2024/03/28/2024.03.27.24304867/F5/graphic-12) Figure 5. Model predictions demonstrate the impact of clinical factors on HIV DNA decay rates Average model predictions were made by bootstrapping 300 random participants and predicting their HIV DNA counts, the model predictions were then averaged within each stratum (tertiles of clinical predictors of HIV reservoir size: timing of ART initiation, initial CD4+ T cell count, and pre-ART viral load). The strata for each key clinical parameter demonstrated that slight changes in these clinical parameters (e.g., by tertiles) significantly influence reservoir decay rates for both intact and defective DNA. Predicted reservoir size 95% confidence limits were narrower for the first phase of decay (weeks 0-5) relative to the second phase of decay (weeks 5-24). ## DISCUSSION The HIV reservoir is established within days after initial viral infection49–51, and although ART can suppress virus to undetectable levels in the blood, persistently cells are a source of viral rebound when therapy is interrupted. A major goal of HIV cure strategies is to induce ART-free remission52. Thus, clearly defining complex reservoir decay dynamics at the time of HIV reservoir establishment – including how key host factors such as the timing of ART initiation, initial CD4+ T cell count, and pre-ART viral load influence these decay rates – will be important in interpreting HIV cure trial results. Potential promising therapeutic interventions may be optimally studied in specific patient populations, but results from such studies can still inform future therapies for all PWH. For example, chronic treated PWH may benefit from the same curative interventions as acute treated PWH but might require adjunctive therapies to reverse cell exhaustion53–55. Here, using a unique cohort of individuals followed longitudinally since acute HIV infection and immediate ART initiation, we observed a clear biphasic decay of the reservoir during the first year of therapy. Decay rates were accelerated among individuals with clinical factors associated with enhanced viral control (higher initial CD4+ T cell count, lower pre-ART viral load, and earlier timing of ART initiation)22,23,34,39,40 and were faster than prior reports among chronic treated PWH initiating ART3. Our model allowed flexible estimation of HIV intact and defective decay rates while adjusting for clinical covariates, in contrast prior studies which mostly fit fully parameterized, mixed effects models3,48,56,57. For example, with HIV intact DNA, the observed initial rapid phase was estimated to have a half-life (t1/2) of 0.71 months until approximately 5 weeks of ART. This was then followed by a slower second phase of decay with a half-life of 3.86 months. Strikingly, our estimated decay rate of HIV intact DNA during the first phase of decay is nearly identical to estimates of HIV intact DNA among chronic treated PWH initiating ART in a prior study (t1/2 = 0.43 months)3. However, our estimates for the second phase of decay (t1/2 = 3.9 months) were ∼5-fold faster than reported their study of chronic treated PWH, where the second phase of decay was estimated to be much longer (t1/2 = 19 months)3. This initial rapid decay phase is thought to reflect death of productively infected cells in the setting of rapid antigen reduction with ART initiation4 and thus similarly rapid decay rates immediately after ART initiation may be similar for PWH who start therapy during acute, as well as chronic infection. Meanwhile, the second phase of decay is thought to reflect a “contraction phase” when activated cells shift to a more memory phenotype58 and therefore, the larger differences that we observed in our cohort compared to earlier findings in chronic-treated PWH may indicate a unique phase during which intervention may considerably influence HIV reservoir decay rates with earlier ART initiation. PWH treated during acute infection have been shown to have less exhausted/relatively intact host immune responses due to earlier removal of antigen and reduction of subsequent inflammation and immune activation. 59 For these reasons, the study of acute treated PWH provides a unique opportunity to identify the optimal timing, potential “windows of time” during which intervention with immunomodulatory agents may have a greater impact on reducing the reservoir size. Most (but not all) studies have shown that those who initiate ART during acute and early infection also have smaller reservoirs than PWH who initiated ART during chronic infection 22–26,28,50,51,60. Given the limited data demonstrating reservoir decay rates during acute and early infection 26,50,61,62, none of which included statistical modeling incorporating key clinical parameters such as timing of ART initiation, initial CD4+ T cell count, and pre-ART viral load, our data add considerable insight to our current understanding of the timing and modification of early HIV reservoir establishment and stabilization. Nonetheless, there are a wealth of other invaluable HIV cohorts, such as the RV254/SEARCH 010 study, that if these data are compiled across studies, may further inform our understanding of how quickly the reservoir decays, and how factors such as ART initiation during acute infection may alter those trajectories. Indeed, in the RV254/SEARCH 010 study, the presented HIV reservoir data (their study measured integrated and unintegrated HIV DNA) plotted longitudinally, seem to indicate that similar decay patterns might be observed in their cohort as well. 50 Acute treated PWH in the study demonstrated rapid decay of both integrated and unintegrated HIV DNA while among chronic treated PWH in their study, only a modest decline in integrated HIV DNA was observed. (The latter may be an artifact of the previously noted observation that among chronic PWH, the majority of integrated HIV DNA tend to be latent or defective, rather than productively infected, as in acute PWH). 50 While there have been limited studies describing HIV reservoir decay rates in acute treated PWH, there now been several studies describing HIV reservoir decay rates during chronic treated HIV (followed as long as ∼20 years of ART). These studies have now demonstrated that cells harboring “intact” provirus (i.e., infected cells capable of producing infectious virus) decay more quickly than defective provirus (infected cells that make up the majority of the reservoir and contain deletions and/or mutated viral sequences63,64) during long-term ART34–37. While our data are unable to yet describe decay rates past 1 year of ART, our study offers for the first time a detailed estimation of HIV reservoir decay rates in the first few weeks to months of ART initiation during acute HIV. While we did not observe discernable differences between HIV intact and defective decay rates as previously described among chronic PWH, 34–36,48 we did find that HIV defective DNA was much more “noisy” than intact DNA (as shown in **Figure 2**), and thus the lack of statistical significance in the second phase of decay reported here may be further investigated in future larger studies. Indeed, the putative mechanism underlying the differential decay rates of HIV intact and defective reservoir during long-term ART is thought to be due to selective immune clearance over time; cells harboring inducible “replication-competent” virus may be more readily detected by host immune surveillance and cleared.34–36,48,65,66 It is unclear if these same factors are at play as early as 5 weeks after initiating ART during acute HIV infection. Studies estimating HIV reservoir decay rates in chronic PWH have shown that the HIV intact reservoir decays with a t1/2 of ∼44 months. 67,68 Subsequent studies studying individuals as far out as ∼20 years of ART suggest that this t1/2 further slows to ∼224-276 months after about 7 years of ART.34–36,48 Interestingly, these long-term follow-up studies have found that during this late phase of reservoir decay, some individuals appear to demonstrate a plateau, or even a slight increase, in their HIV intact reservoir size. The reasons for this are unclear but some have argued that this may be due to a “tipping point” at which the host immune response has cleared the majority of the intact reservoir in the body and is then counterbalanced by ongoing cell proliferation. 48 While we were able to perform an in-depth statistical modeling study, leveraging unique acute treated participant samples and a large number of longitudinal HIV reservoir measurements, our study has several limitations that deserve mention. While the peripheral HIV reservoir largely reflect infected-cell populations originating from tissues reservoir, 49,50,69,70 we only included peripheral CD4+ T cell HIV reservoir measures in our model. In addition, while we used the widely utilized IPDA (a high throughput assay that can be performed on this large number of samples), alternative assays, such as near-full length proviral sequencing or QVOA, would have been informative to further interpret our findings (if these were able to be performed at the scale required for this large number of samples). However, HIV intact DNA measured using the IPDA has been shown to closely reflect trends resulting from these other assays, even considering the known enrichment of integrated forms of HIV DNA observed in acute PWH.50 As with all molecular assays for HIV, certain polymorphisms at primer or probe binding sites can impact IPDA assay performance. In this study, we observed IPDA signal failure for 6 of 67 participants (8.9%) – a rate consistent recent reports on very large cohorts of PWH from North America and Europe where subtype B predominates (6-7%) 34,71. While the IPDA does not perfectly discriminate intact from defective proviruses, the cross sectional and longitudinal correlations between IPDA and QVOA measurements of the HIV reservoir strongly support the use of the higher throughput IPDA in large scale studies of proviral decay dynamics. We did not include parameters for potential influences from changes in clonal landscape over time as our study did not include HIV integration analyses. However, the clonal landscape at the time of acute HIV is extremely diverse, and we hypothesize that this effect is more likely to have higher impact after longer duration of ART suppression. Future models should include these parameters to formally test this hypothesis. Finally, there are few highly characterized acute HIV cohorts to date, and each study possesses unique host and viral characteristics making direct cross-cohort comparisons challenging. These cohort-specific factors include host genetics, socio-behavioral demographics (our study included mostly men who have sex with men), clinical practices and public policy guidelines for ART initiation, and viral subtype (mostly subtype B in our study). Our HIV reservoir decay model should be further validated in these global populations, including individuals initiating treatment during chronic HIV, which represents the majority of people currently living with HIV. Overall, these data encourage future collaborative work to determine decay dynamics across global populations so that future cure strategies may be effective across clades and in people with larger reservoirs, exhausted immune responses who initiated ART during chronic HIV. ## METHODS ### Study participants Individuals with newly diagnosed acute HIV (<100 days from estimated date of infection) were enrolled in the UCSF Treat Acute HIV cohort between December 1, 2015 to November 30, 2020 and co-enrolled in the UCSF SCOPE HIV cohort, an ongoing longitudinal study of over 2,500 PWH to characterize the natural history of HIV disease. Eligible participants were consented, provided same-day ART initiation with tenofovir/emtricitabine (TDF/FTC, then TAF/FTC once available in 2016) + dolutegravir (DTG) and linked to clinical care 46. Individuals reporting concomitant PrEP use (<100 days from any potential exposure to HIV by history and/or clinical test results) were also started on darunavir+ritonavir (DRV/r) as a fourth drug, which was continued until confirmation of baseline HIV genotyping test results (Monogram Biosciences, South San Francisco, CA, U.S.A.). Additional ART changes necessary for clinical care (e.g., laboratory abnormalities, drug-drug interactions, and/or participant preference) were honored and adjusted during the period of study. Participants signed a release of information which allowed clinical data extraction to determine prior HIV negative test results from the SFDPH, as well as additional HIV test results. Study participants were seen for monthly study visits for the first 24 weeks and then every 3-4 months thereafter. Inclusion criteria for the study were prior HIV negative testing within the last 90 days, laboratory-confirmed HIV-1 infection by antibody/antigen and/or plasma HIV RNA assay, and willingness to be participate in the study for at least 24 weeks. Participants with severe renal or hepatic impairment, concurrent treatment with immunomodulatory drugs, or exposure to any immunomodulatory drugs in the preceding 90 days prior to study entry, pregnant or breastfeeding women, or participants unwilling to agree to the use a double-barrier method of contraception throughout the study period, were excluded. For each study participant, using the Infection Dating Tool ([https://tools.incidence-estimation.org/idt/](https://tools.incidence-estimation.org/idt/)) 42, the estimated date of detected HIV infection (EDDI), along with a “confidence interval” for early probable (EP-EDDI) and late probable (LP-EDDI) date was calculated using each participant’s prior clinical HIV test results, as well the study baseline HIV test results. At each visit, detailed interviews included questions regarding current medications, medication adherence, intercurrent illnesses, and hospitalizations were performed. In addition, peripheral blood sampling at each visit was performed to measure plasma HIV RNA, CD4+ T cell count, and clinical labs (complete blood count, metabolic panel), as well as blood for storage - PBMCs and plasma. For a subset of individuals who reported potential PrEP overlap with HIV EDDI, participants were consented and co-enrolled in the SeroPrEP study and dried blood spot and hair were collected to confirm PrEP use. Finally, some individuals were invited to participate in optional sub-studies, which included leukapheresis and lymphoid tissue sampling (rectal and ileal biopsies, lymph node fine needle aspirate) after 12 months of ART suppression (data not included here in this paper). All participants provided written informed consent, and the institutional review board of UCSF approved the research. ### HIV reservoir quantification There is currently no “gold standard” for measuring the HIV reservoir. Moreover, the HIV reservoir largely consists of “defective” virus that harbors mutations prohibiting the production of infectious virus 63,64. The frequencies of HIV intact and defective (3’ and 5’) DNA were quantified using the intact proviral DNA assay (PDA), as previously described 72. Briefly, CD4+ T cells were isolated from cryopreserved PBMCs (EasySep Human CD4+ T cell Enrichment Kit, Stemcell Technologies), with cell count, viability, and purity assessed by flow cytometry both before and after selection. Negatively selected CD4+ T cells were recovered for the samples, and genomic DNA was extracted using the QIAamp DNA Mini Kit (Qiagen). DNA concentration and quality were determined by fluorometry (Qubit dsDNA BR Assay Kit, Thermo Fisher Scientific) and ultraviolet-visible (UV/VIS) spectrophotometry (QIAxpert, Qiagen). The frequency of “intact” HIV provirus (i.e., estimating the potentially replication-competent reservoir) was determined using two multiplex digital droplet polymerase chain reaction (ddPCR) assays performed in parallel: (1) the HIV-1 Proviral Discrimination reaction which distinguishes intact from defective provirus via two strategically placed amplicons in HIV psi and RRE regions as well as a hypermutation discrimination probe, and (2) the Copy Reference/Shearing reaction, which quantifies DNA shearing and input diploid cell equivalents using the human *RPP30* gene72. All ddPCR reactions were assembled via automated liquid handles to maximize reproducibility and analyzed using the BioRad QX200 AutoDG Digital Droplet PCR system (BioRad). Up to 700 ng of genomic DNA were analyzed per reaction, and final input DNA concentrations were dependent upon the concentration of recovered DNA. Samples were batch processed and analyzed, including negative controls from uninfected donors and J-Lat full-length clone 6.3 (E. Verdin, Gladstone Institutes and UCSF, San Francisco, CA, USA) cells as positive controls, as previously described. Across the >500 IPDA measurements performed, we interrogated a median of 4.8 x 105 CD4+ T cell genomes per assay and observed a median DNA shearing index (DSI) of 0.40. ### Statistical Methods We developed a novel semiparametric biphasic decay model to estimate the rate of change of HIV DNA over time in log10 copies per 106 CD4+ T cells: ![Formula][1] In equation (1), *iit* represents either the intact or defective HIV DNA counts, of the *i*-th participant at *i*-th visit. The number of weeks since ART initiation is denoted *Tit*. The model additionally controls for baseline clinical information defined as the initial CD4+ T cell count, *Ci*, pre-ART viral load, *Vi*, and the estimated time between HIV infection and ART initiation, *Ei*. The delay in ART initiation is centered to have a mean of zero for later analysis; this offset is approximately 60 days in our cohort. Participant-level random effects are denoted µ*i*. Building on existing models, 3,48 we parameterize the decay as a continuous, linear spline with a single knot at ![Graphic][2]. Using this parameterization, we can directly interpret *β*1 as the decay rate before *τ* and *β*2 as the decay rate after *τ*; we use the same parameterization for ![Graphic][3] but with different *β* values. We use a cubic spline with 20 regularly spaced knots for both *f*3(*Ci*) and *f*4(*Vi*). Once *τ* is fixed, model fitting is performed using least-squares regression. When testing our model against monophasic and triphasic models, we used either linear terms or linear splines with two knots for *f*1 and *f*2 respectively. The estimation of the inflection point, τ, is done by minimizing the model’s mean absolute prediction error. We fixed a grid of candidate *τ* values from 0 weeks to 25 weeks, by half-week. The mean absolute prediction error was estimated using leave-one-out cross-validation calculated as ![Formula][4] where ![Graphic][5] denotes the predicted HIV DNA counts for patient *ι* at time *t* using the model fit with every patient except patient *i*. This loss metric was also used to estimate the two knots in the triphasic model. Akaike information criteria (AIC) was used to compare the fits of the mono-, bi-, and triphasic models. Half-life estimates and their confidence intervals are computed using the multivariate delta method. During the first decay phase, we are interested in estimating the baseline half-life in months and the adjustment to the half-life in months due to delay in ART initiation. The half-life (in months) before *τ* is calculated as ![Formula][6] where the second equality is the degree-one Taylor series about *Ei* = 0; this is the reason we center *Ei* in our pre-processing. Thus we are interested in estimating the base-line half-life ![Graphic][7] and the adjustment to half-life for every week delay in ART ![Graphic][8]. Appealing the asymptotic normality of our parameter estimates and the multivariate delta method, we get the following asymptotic distribution for our half-life estimates: ![Formula][9] ![Formula][10] where Σ is the covariance between *β*1 and *β*2, and *J* is the Jacobian matrix of *g*(*β*1,*β*2). We plug-in ![Graphic][11], along with ![Graphic][12], and ![Graphic][13], from our model to estimate the asymptotic confidence intervals for base-line half-life and adjustment to half-life for delay in ART initiation. Percent decay per week before *τ* is calculated using the transformation *h*(**β**1) = −100, (2*β*1 − 1); the negative sign accounts for the directionality of the decay. These calculations are the same for estimates of the half-life after *τ*. ## Supporting information Supplemental Tables and Figures [[supplements/304867_file02.docx]](pending:yes) ## Data Availability All data produced in the present study are available upon reasonable request to the authors. ## CONFLICTS The authors do not have a commercial or other association that might pose a conflict of interest. ## FUNDING This work was supported in part by the National Institutes of Health: K23GM112526 (SAL), the DARE Collaboratory (UM1AI164560; SGD), the AccelevirDx HIV Reservoir Testing Resource (U24AI143502; GML), and NIH/NIAID R01A141003 (TJH). This work was also supported by the amfAR Research Consortium on HIV Eradication a.k.a. ARCHE (108072-50-RGRL; SGD), the Bill & Melinda Gates Foundation (INV-002703; SGD), and investigator-initiated research grants from ViiV Healthcare (A126326; SAL) and Gilead Sciences (IN-US-236-1354; SAL). The content of this publication does not necessarily reflect the views or policies of the Department of Health and Human Services, the San Francisco Department of Health, nor does mention of trade names, commercial products, or organizations imply endorsement by the U.S. Government. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript. ## Previous presentation Preliminary data were presented in July 2023, as a poster presentation at the International AIDS Society (IAS) conference in Brisbane, Australia. ## Reprints Reprint requests can be directed to Dr. Sulggi Lee, the corresponding author (contact information above). **Supplementary Figure 1. The UCSF Treat Acute HIV cohort study participants**. A total of 67 participants met inclusion criteria for acute HIV, defined as <100 days since the estimated date of detected HIV infection (EDDI) using the Infection Dating Tool ([https://tools.incidence-estimation.org/idt/](https://tools.incidence-estimation.org/idt/)). The numbers of study participants by Fiebig stages (I-V) of HIV recency are also shown ([https://doi.org:10.1097/01.aids.0000076308.76477.b8](https://doi.org:10.1097/01.aids.0000076308.76477.b8)). PrEP = Pre-exposure prophylaxis with tenofovir disoproxil fumarate/emtricitabine (TDF/FTC). **Supplementary Figure 2. Calculation of estimated dates of detected HIV infection**. The estimated dates of detected HIV infection (EDDI), along with a “confidence interval” for early probable (EP-EDDI) and late probable (LP-EDDI) dates, were calculated using participants’ clinical test results as well as baseline study visit confirmatory assay results. **Supplementary Figure 3. Fiebig and race/ethnicity distribution within the UCSF Treat Acute HIV cohort.** The distribution of study participants in the UCSF Treat Acute HIV cohort, by timing of ART initiation (a) and self-reported race/ethnicity (b). A total of 67 participants met inclusion criteria for acute HIV, defined as <100 days since the estimated date of detected HIV infection (EDDI) using the Infection Dating Tool ([https://tools.incidence-estimation.org/idt/](https://tools.incidence-estimation.org/idt/)). The proportions of study participants by Fiebig stages (I-V) are also shown in (a). **Supplementary Figure 4. HIV-1/2 test results for study participants**. The proportion of study participants with either negative and/or indeterminate test results for HIV-1/2 p24 antigen/antibody assay (Architect) (a) and HIV-1/2 differentiation (Geenius) antibody assay (b) at baseline study visit were consistent with rates with our San Francisco Department of Public Health rates (27% and 28%, respectively). **Supplementary Figure 5. Observed HIV intact and defective reservoir decays highlighting participants reporting a history of PrEP use**. Most participants did not report PrEP overlap near the time of HIV diagnosis (grey lines). A subset of participants reported PrEP overlap within 10 days of HIV diagnosis; N=6 participants acquired HIV while already taking PrEP (yellow lines) and N=8 participants were prescribed PrEP but were found to already have acquired HIV (blue lines). **Supplementary Figure 6. Semiparametric monophasic, biphasic, and triphasic generalized additive model fit comparison suggest biphasic model has the most parsimonious fit**. Infection points were selected by minimizing the leave-one-out prediction mean absolute error (MAE) for each model and both HIV DNA assays. Tuning results for the triphasic model show that the best model (emphasized in red) either has two knots near each other or one knot near zero (a) suggesting that it is similar in shape to a biphasic model; leave-one-out prediction mean squared error (MSE) is proved for extra support. The Akaike information criteria (AIC) value and 95% confidence interval was then used to compare monophasic, biphasic, and triphasic models for both assays (b). AIC estimates and confidence intervals were found by fitting models on 500 bootstrap resampled data sets. For both HIV intact and defective DNA, the biphasic and triphasic models had relatively equivalent performance. **Supplementary Figure 7. Modeling using the full first year of data suggests reservoir decay trends continue past the first 24 weeks on ART**. 65.7% of the total study participants continued in the study beyond 24 weeks. Observed HIV reservoir patterns are shown as thin grey lines, while the decay pattern for the model-predicted average participant is shown as the thick black lines (a). Average predicted participant predictions were made by taking the mean of *Ei* (estimated time between HIV infection and ART initiation), *ci* (initial CD4+ T cell count), and *vi* (log10 pre-ART plasma viral load) across participants. Model inflection point tuning is shown for both intact (left panels) and defective (right panels) HIV DNA decay models, respectively (b). The biphasic decay model’s inflection point was tuned by estimating the *τ* that minimized prediction error measured by leave-one-out mean absolute deviation (MAE, upper panels) and leave-one-out mean squared error (MSE, lower panels). A grid of *τ* values from weeks 0 to 52, by half-weeks, was considered as potential inflection points. The best *τ* for each loss (red point) is shown in relation to the final model’s selected inflection point of *τ* = 5 weeks as reference (vertical dashed line). **Supplementary Figure 8. Inflection point sensitivity analyses demonstrate some variability when excluding populations of potential outliers.** To test whether the final model inflection point selection of *τ = 5* weeks was influenced by potential outlier data we performed *τ* estimation on three clinically interesting sub-populations. Separate models were fit that excluded (a) participants reporting prior PrEP use (<10 days overlap between last PrEP use and estimated date of detected HIV infection), (b) participants with plasma viral load “blips” (defined as a one-time viral load >1000 copies/mL or two consecutive viral loads >100 copies/mL between weeks 0-24), and (c) participants with sudden increases in HIV intact DNA (defined as >50% increase between two consecutive measurements of HIV intact DNA during weeks 0-24). A regular grid of possible *τ* was used (0-26 weeks by half-week) and the leave-one-out cross-validation (LOOCV) mean absolute prediction error (MAE) was computed for each candidate τ. The model attaining the best prediction error is denoted with a red point and our selected inflection point (τ *= 5*) is shown with a dashed vertical line. Refer to **Supplementary Table 2** to get the sample size for each sensitivity analysis and to **Supplementary Figure 9 to see which patients are excluded from each sensitivity analysis.** **Supplementary Figure 9. Model estimate sensitivity analyses demonstrated that results were overall unchanged after excluding populations of potential outliers**. The final model (*τ* = 5 weeks) was fit on three clinically interesting sub-populations to assess if the influence of potential outlier data. Separate models were fit that excluded (a) participants reporting prior PrEP use (<10 days overlap between last PrEP use and estimated date of detected HIV infection), (b) participants with plasma viral load “blips” (defined as a one-time viral load >1000 copies/mL or two consecutive viral loads >100 copies/mL between weeks 0-24), and (c) participants with sudden increases in HIV intact DNA (defined as >50% increase between two consecutive measurements of HIV intact DNA during weeks 0-24). Models were fit using the cohort data (grey lines), but not the potential outlier data (red lines). The resulting predict average participant HIV reservoir decay patterns are shown as thick black lines. Refer to **Supplementary Table 2** to get the sample sizes and half-life estimates for each sensitivity analysis. **Supplementary Figure 10. Predicted versus observed plots show good model performance for both HIV intact and defective DNA**. Validation for the final models for intact and defective HIV DNA decay was initially performed by looking at the plots of predicted vs observed HIV DNA counts. These plots show that both models produce relatively unbiased estimates across the observed range of HIV DNA counts and that the residual variance in the defective reservoir is much higher than the intact reservoir. A dashed, red line at y=x is added to help interpret the model fits. **Supplementary Table 1. Model estimates of HIV intact and defective DNA decay rates from baseline models**. Baseline models include random intercepts for each participant but are otherwise unadjusted for clinical covariates. Model estimates are provided for discrete periods of ART suppression after initial treatment initiation: weeks 0 up to *τ* = 5 and for weeks after *τ.* **Supplementary Table 2. Model estimates of HIV intact and defective DNA decay rates from models adjusted for initial CD4+ T cell count, pre-ART HIV RNA, and timing of ART initiation**. Models also include a random intercept for each participant. Model estimates are provided for discrete periods of ART suppression after initial treatment initiation: weeks 0 up to *τ* = 5 and for weeks after *τ*. **Supplementary Table 3. Changes in HIV decay rates of intact and defective DNA associated with timing of ART initiation**. Faster decay rates in both intact and defective HIV DNA during phase 1 are associated with lower delay in ART initiation. Intact, but not defective, HIV DNA have faster predicted decay rates associated with lower delay in ART initiation. **Supplementary Table 4. Changes in the intact and defective HIV DNA reservoir size associated with initial CD4+ T cell count and pre-ART viral load**. Table values are log10 adjustments to the viral reservoir size at each clinical parameter. Higher initial CD4+ T cell count and lower pre-ART viral load are associated with smaller viral reservoirs. Additional estimates can be taken from **Figure 3**. ## Acknowledgements The authors wish to acknowledge the participation of all the study participants who contributed to this work as well as the clinical research staff of the UCSF Treat Acute HIV and SCOPE cohorts who made this research possible. All funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript. All authors provided critical feedback in finalizing the report. HH, SGD, and SAL conceived and designed the study. HH, SGD, and SAL obtained funding to support the clinical enrollment of study participants, and SAL and SGD obtained funding to support characterization of the HIV reservoir. SEC, SB, DH, and MG facilitated coordination with San Francisco Department of Public Health and Ward 86 clinical services to link patients into care and provided critical feedback on the clinical management of acute HIV. SAL, RH, SGD, TJH, HMH, SS, SD, and VP coordinated the collection, management, and quality control processes for the clinical data, and SAL, SGD, JM, FH, and CP provided biospecimens. JM and TJH performed biospecimen processing, GML, MM, and KR performed the HIV reservoir assays. LS developed the initial decay models, under the guidance of SAL and JW, and AB and SAL further modified these models under the guidance of JW, LS, and JS. GML, MM, KR, LS, JS, JW, and SAL analyzed the HIV reservoir data. SAL, AB, LS, and JW wrote the report. SS, SD, VP, CS, LS, JS, JW, and SAL finalized data visualization for the manuscript. All authors provided critical feedback in finalizing the manuscript. ## Footnotes * # Co-Lead Authors: Alton Barbehenn and Lei Shi * Received March 27, 2024. * Revision received March 27, 2024. * Accepted March 28, 2024. * © 2024, Posted by Cold Spring Harbor Laboratory This pre-print is available under a Creative Commons License (Attribution-NonCommercial-NoDerivs 4.0 International), CC BY-NC-ND 4.0, as described at [http://creativecommons.org/licenses/by-nc-nd/4.0/](http://creativecommons.org/licenses/by-nc-nd/4.0/) ## REFERENCES 1. 1.Chun, T. W. et al. Rebound of plasma viremia following cessation of antiretroviral therapy despite profoundly low levels of HIV reservoir: implications for eradication. AIDS 24, 2803–2808 (2010). doi:10.1097/QAD.0b013e328340a239 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1097/QAD.0b013e328340a239&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=20962613&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F03%2F28%2F2024.03.27.24304867.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000284057300006&link_type=ISI) 2. 2.Hocqueloux, L. et al. Long-term immunovirologic control following antiretroviral therapy interruption in patients treated at the time of primary HIV-1 infection. AIDS 24, 1598–1601 (2010). doi:10.1097/qad.0b013e32833b61ba [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1097/QAD.0b013e32833b61ba&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=20549847&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F03%2F28%2F2024.03.27.24304867.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000278636400027&link_type=ISI) 3. 3.White, J. A. et al. Complex decay dynamics of HIV virions, intact and defective proviruses, and 2LTR circles following initiation of antiretroviral therapy. Proc Natl Acad Sci U S A 119 (2022). doi:10.1073/pnas.2120326119 [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NDoicG5hcyI7czo1OiJyZXNpZCI7czoxNzoiMTE5LzYvZTIxMjAzMjYxMTkiO3M6NDoiYXRvbSI7czo1MDoiL21lZHJ4aXYvZWFybHkvMjAyNC8wMy8yOC8yMDI0LjAzLjI3LjI0MzA0ODY3LmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 4. 4.Perelson, A. S. et al. Decay characteristics of HIV-1-infected compartments during combination therapy. Nature 387, 188–191 (1997). doi:10.1038/387188a0 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/387188a0&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=9144290&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F03%2F28%2F2024.03.27.24304867.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=A1997WX94500056&link_type=ISI) 5. 5.Kwon, K. J. & Siliciano, R. F. HIV persistence: clonal expansion of cells in the latent reservoir. J Clin Invest 127, 2536–2538 (2017). doi:10.1172/JCI95329 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1172/JCI95329&link_type=DOI) 6. 6.Finzi, D. & Siliciano, R. F. Viral dynamics in HIV-1 infection. Cell 93, 665–671 (1998). doi:10.1016/s0092-8674(00)81427-0 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/S0092-8674(00)81427-0&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=9630210&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F03%2F28%2F2024.03.27.24304867.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000073956700002&link_type=ISI) 7. 7.Chomont, N. et al. HIV reservoir size and persistence are driven by T cell survival and homeostatic proliferation. Nature medicine 15, 893–900 (2009). doi:10.1038/nm.1972 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/nm.1972&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=19543283&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F03%2F28%2F2024.03.27.24304867.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000268770400033&link_type=ISI) 8. 8.Yukl, S. A. et al. Differences in HIV burden and immune activation within the gut of HIV-positive patients receiving suppressive antiretroviral therapy. The Journal of infectious diseases 202, 1553–1561 (2010). doi:10.1086/656722 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1086/656722&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=20939732&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F03%2F28%2F2024.03.27.24304867.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000283057000014&link_type=ISI) 9. 9.Archin, N. M. et al. Administration of vorinostat disrupts HIV-1 latency in patients on antiretroviral therapy. Nature 487, 482–485 (2012). doi:10.1038/nature11286 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/nature11286&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=22837004&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F03%2F28%2F2024.03.27.24304867.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000306815300038&link_type=ISI) 10. 10.Elliott, J. H. et al. Short-term administration of disulfiram for reversal of latent HIV infection: a phase 2 dose-escalation study. Lancet HIV 2, e520–529 (2015). doi:10.1016/S2352-3018(15)00226-X [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/S2352-3018(15)00226-X&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=26614966&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F03%2F28%2F2024.03.27.24304867.atom) 11. 11.Rasmussen, T. A. et al. Panobinostat, a histone deacetylase inhibitor, for latent-virus reactivation in HIV-infected patients on suppressive antiretroviral therapy: a phase 1/2, single group, clinical trial. Lancet HIV 1, e13–21 (2014). doi:10.1016/S2352-3018(14)70014-1 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/S2352-3018(14)70014-1&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=26423811&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F03%2F28%2F2024.03.27.24304867.atom) 12. 12.Elliott, J. H. et al. Activation of HIV transcription with short-course vorinostat in HIV-infected patients on suppressive antiretroviral therapy. PLoS pathogens 10, e1004473 (2014). doi:10.1371/journal.ppat.1004473 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1371/journal.ppat.1004473&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=25393648&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F03%2F28%2F2024.03.27.24304867.atom) 13. 13.Gay, C. L. et al. Assessing the impact of AGS-004, a dendritic cell-based immunotherapy, and vorinostat on persistent HIV-1 Infection. Sci Rep 10, 5134 (2020). doi:10.1038/s41598-020-61878-3 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41598-020-61878-3&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=32198428&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F03%2F28%2F2024.03.27.24304867.atom) 14. 14.Fidler, S. et al. Antiretroviral therapy alone versus antiretroviral therapy with a kick and kill approach, on measures of the HIV reservoir in participants with recent HIV infection (the RIVER trial): a phase 2, randomised trial. Lancet 395, 888–898 (2020). doi:10.1016/S0140-6736(19)32990-3 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/S0140-6736(19)32990-3&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=32085823&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F03%2F28%2F2024.03.27.24304867.atom) 15. 15.Gutierrez, C. et al. Bryostatin-1 for latent virus reactivation in HIV-infected patients on antiretroviral therapy. AIDS 30, 1385–1392 (2016). doi:10.1097/QAD.0000000000001064 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1097/QAD.0000000000001064&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=26891037&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F03%2F28%2F2024.03.27.24304867.atom) 16. 16.Vibholm, L. et al. Short-Course Toll-Like Receptor 9 Agonist Treatment Impacts Innate Immunity and Plasma Viremia in Individuals With Human Immunodeficiency Virus Infection. Clin Infect Dis 64, 1686–1695 (2017). doi:10.1093/cid/cix201 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/cid/cix201&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=28329286&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F03%2F28%2F2024.03.27.24304867.atom) 17. 17.Riddler, S. A. et al. Vesatolimod, a Toll-like Receptor 7 Agonist, Induces Immune Activation in Virally Suppressed Adults Living With Human Immunodeficiency Virus-1. Clin Infect Dis 72, e815–e824 (2021). doi:10.1093/cid/ciaa1534 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/cid/ciaa1534&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=33043969&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F03%2F28%2F2024.03.27.24304867.atom) 18. 18.Elliott, J. H. et al. Short-term administration of disulfiram for reversal of latent HIV infection: a phase 2 dose-escalation study. Lancet HIV 2, e520–529 (2015). doi:10.1016/S2352-3018(15)00226-X [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/S2352-3018(15)00226-X&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=26614966&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F03%2F28%2F2024.03.27.24304867.atom) 19. 19.Bar, K. J. et al. Effect of HIV Antibody VRC01 on Viral Rebound after Treatment Interruption. N Engl J Med 375, 2037–2050 (2016). doi:10.1056/NEJMoa1608243 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1056/NEJMoa1608243&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=27959728&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F03%2F28%2F2024.03.27.24304867.atom) 20. 20.Gunst, J. D. et al. Early intervention with 3BNC117 and romidepsin at antiretroviral treatment initiation in people with HIV-1: a phase 1b/2a, randomized trial. Nat Med 28, 2424–2435 (2022). doi:10.1038/s41591-022-02023-7 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41591-022-02023-7&link_type=DOI) 21. 21.Gunst, J. D. et al. in Conference on Retroviruses and Opportunistic Infections. 22. 22.Archin, N. M. et al. Immediate antiviral therapy appears to restrict resting CD4+ cell HIV-1 infection without accelerating the decay of latent infection. Proc Natl Acad Sci U S A 109, 9523–9528 (2012). doi:10.1073/pnas.1120248109 [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NDoicG5hcyI7czo1OiJyZXNpZCI7czoxMToiMTA5LzI0Lzk1MjMiO3M6NDoiYXRvbSI7czo1MDoiL21lZHJ4aXYvZWFybHkvMjAyNC8wMy8yOC8yMDI0LjAzLjI3LjI0MzA0ODY3LmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 23. 23.Buzon, M. J. et al. 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 (2014). doi:10.1128/JVI.01046-14 [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoianZpIjtzOjU6InJlc2lkIjtzOjExOiI4OC8xNy8xMDA1NiI7czo0OiJhdG9tIjtzOjUwOiIvbWVkcnhpdi9lYXJseS8yMDI0LzAzLzI4LzIwMjQuMDMuMjcuMjQzMDQ4NjcuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 24. 24.Chun, T. W. et al. 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 (1998). doi:10.1073/pnas.95.15.8869 [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NDoicG5hcyI7czo1OiJyZXNpZCI7czoxMDoiOTUvMTUvODg2OSI7czo0OiJhdG9tIjtzOjUwOiIvbWVkcnhpdi9lYXJseS8yMDI0LzAzLzI4LzIwMjQuMDMuMjcuMjQzMDQ4NjcuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 25. 25.Strain, M. C. et al. Effect of treatment, during primary infection, on establishment and clearance of cellular reservoirs of HIV-1. J Infect Dis 191, 1410–1418 (2005). doi:10.1086/428777 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1086/428777&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=15809898&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F03%2F28%2F2024.03.27.24304867.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000228128800006&link_type=ISI) 26. 26.Hocqueloux, L., Saez-Cirion, A. & Rouzioux, C. Immunovirologic control 24 months after interruption of antiretroviral therapy initiated close to HIV seroconversion. JAMA Intern Med 173, 475–476 (2013). doi:10.1001/jamainternmed.2013.2176 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1001/jamainternmed.2013.2176&link_type=DOI) 27. 27.Ananworanich, J. et al. Virological and immunological characteristics of HIV-infected individuals at the earliest stage of infection. J Virus Erad 2, 43–48 (2016). 28. 28.Ananworanich, J. et al. Impact of multi-targeted antiretroviral treatment on gut T cell depletion and HIV reservoir seeding during acute HIV infection. PLoS One 7, e33948 (2012). doi:10.1371/journal.pone.0033948 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1371/journal.pone.0033948&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=22479485&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F03%2F28%2F2024.03.27.24304867.atom) 29. 29.Deleage, C. et al. Impact of early cART in the gut during acute HIV infection. JCI Insight 1 (2016). doi:10.1172/jci.insight.87065 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1172/jci.insight.87065&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=27446990&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F03%2F28%2F2024.03.27.24304867.atom) 30. 30.Takata, H. et al. Long-term antiretroviral therapy initiated in acute HIV infection prevents residual dysfunction of HIV-specific CD8(+) T cells. EBioMedicine 84, 104253 (2022). doi:10.1016/j.ebiom.2022.104253 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.ebiom.2022.104253&link_type=DOI) 31. 31.Oxenius, A. et al. Early highly active antiretroviral therapy for acute HIV-1 infection preserves immune function of CD8+ and CD4+ T lymphocytes. Proc Natl Acad Sci U S A 97, 3382–3387 (2000). doi:10.1073/pnas.97.7.3382 [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NDoicG5hcyI7czo1OiJyZXNpZCI7czo5OiI5Ny83LzMzODIiO3M6NDoiYXRvbSI7czo1MDoiL21lZHJ4aXYvZWFybHkvMjAyNC8wMy8yOC8yMDI0LjAzLjI3LjI0MzA0ODY3LmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 32. 32.Streeck, H. et al. Immunological and virological impact of highly active antiretroviral therapy initiated during acute HIV-1 infection. J Infect Dis 194, 734–739 (2006). doi:10.1086/503811 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1086/503811&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=16941338&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F03%2F28%2F2024.03.27.24304867.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000240316600003&link_type=ISI) 33. 33.Namazi, G. et al. The Control of HIV After Antiretroviral Medication Pause (CHAMP) Study: Posttreatment Controllers Identified From 14 Clinical Studies. J Infect Dis 218, 1954–1963 (2018). doi:10.1093/infdis/jiy479 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/infdis/jiy479&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=30085241&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F03%2F28%2F2024.03.27.24304867.atom) 34. 34.Peluso, M. J. et al. Differential decay of intact and defective proviral DNA in HIV-1-infected individuals on suppressive antiretroviral therapy. JCI Insight 5 (2020). doi:10.1172/jci.insight.132997 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1172/jci.insight.132997&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=32045386&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F03%2F28%2F2024.03.27.24304867.atom) 35. 35.Gandhi, R. T. et al. Selective Decay of Intact HIV-1 Proviral DNA on Antiretroviral Therapy. J Infect Dis 223, 225–233 (2021). doi:10.1093/infdis/jiaa532 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/infdis/jiaa532&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=32823274&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F03%2F28%2F2024.03.27.24304867.atom) 36. 36.Gandhi, R. T. et al. Varied Patterns of Decay of Intact Human Immunodeficiency Virus Type 1 Proviruses Over 2 Decades of Antiretroviral Therapy. J Infect Dis 227, 1376–1380 (2023). doi:10.1093/infdis/jiad039 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/infdis/jiad039&link_type=DOI) 37. 37.Antar, A. A. et al. Longitudinal study reveals HIV-1-infected CD4+ T cell dynamics during long-term antiretroviral therapy. J Clin Invest 130, 3543–3559 (2020). doi:10.1172/JCI135953 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1172/JCI135953&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=32191639&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F03%2F28%2F2024.03.27.24304867.atom) 38. 38.Shi, L. et al. in International AIDS Society Conference on HIV Science. 39. 39.Ananworanich, J. et al. HIV DNA Set Point is Rapidly Established in Acute HIV Infection and Dramatically Reduced by Early ART. EBioMedicine 11, 68–72 (2016). doi:10.1016/j.ebiom.2016.07.024 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.ebiom.2016.07.024&link_type=DOI) 40. 40.Crowell, T. A. et al. Virologic failure is uncommon after treatment initiation during acute HIV infection. AIDS 30, 1943–1950 (2016). doi:10.1097/QAD.0000000000001148 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1097/QAD.0000000000001148&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F03%2F28%2F2024.03.27.24304867.atom) 41. 41.Lee SA, H. T., Gandhi M, Coffey S, Harting H, Hoh R, Peluso MJ, Siegel D, Crouch P, Scott H, Cohen SD, Sachdev D, Bacon O, Busch M, Pilcher C, Buchbinder S, Havlir DV, Deeks SG. in International AIDS Society Conference. 42. 42.Grebe, E. et al. Interpreting HIV diagnostic histories into infection time estimates: analytical framework and online tool. BMC Infect Dis 19, 894 (2019). doi:10.1186/s12879-019-4543-9 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1186/s12879-019-4543-9&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=31655566&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F03%2F28%2F2024.03.27.24304867.atom) 43. 43.Fiebig, E. W. et al. Dynamics of HIV viremia and antibody seroconversion in plasma donors: implications for diagnosis and staging of primary HIV infection. AIDS 17, 1871–1879 (2003). doi:10.1097/01.aids.0000076308.76477.b8 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1097/00002030-200309050-00005&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=12960819&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F03%2F28%2F2024.03.27.24304867.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000185067700002&link_type=ISI) 44. 44.Keele, B. F. et al. Identification and characterization of transmitted and early founder virus envelopes in primary HIV-1 infection. Proc Natl Acad Sci U S A 105, 7552–7557 (2008). doi:10.1073/pnas.0802203105 [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NDoicG5hcyI7czo1OiJyZXNpZCI7czoxMToiMTA1LzIxLzc1NTIiO3M6NDoiYXRvbSI7czo1MDoiL21lZHJ4aXYvZWFybHkvMjAyNC8wMy8yOC8yMDI0LjAzLjI3LjI0MzA0ODY3LmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 45. 45.Prevention., C. f. D. C. a. HIV Surveillance Report. [http://www.cdc.gov/hiv/library/reports/hiv-surveillance.html](http://www.cdc.gov/hiv/library/reports/hiv-surveillance.html). Published May 2022. Accessed Nov 2, 2023., (2020). 46. 46.Buchbinder, S. P. & Havlir, D. V. Getting to Zero San Francisco: A Collective Impact Approach. J Acquir Immune Defic Syndr 82 Suppl 3, S176–S182 (2019). doi:10.1097/QAI.0000000000002200 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1097/QAI.0000000000002200&link_type=DOI) 47. 47.Cohen, S. E. et al. Acquisition of tenofovir-susceptible, emtricitabine-resistant HIV despite high adherence to daily pre-exposure prophylaxis: a case report. Lancet HIV (2018). doi:10.1016/S2352-3018(18)30288-1 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/S2352-3018(18)30288-1&link_type=DOI) 48. 48.McMyn, N. F. et al. The latent reservoir of inducible, infectious HIV-1 does not decrease despite decades of antiretroviral therapy. J Clin Invest 133 (2023). doi:10.1172/JCI171554 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1172/JCI171554&link_type=DOI) 49. 49.Whitney, J. B. et al. Rapid seeding of the viral reservoir prior to SIV viraemia in rhesus monkeys. Nature 512, 74–77 (2014). doi:10.1038/nature13594 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/nature13594&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=25042999&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F03%2F28%2F2024.03.27.24304867.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000339908000034&link_type=ISI) 50. 50.Leyre, L. et al. Abundant HIV-infected cells in blood and tissues are rapidly cleared upon ART initiation during acute HIV infection. Sci Transl Med 12 (2020). doi:10.1126/scitranslmed.aav3491 [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTE6InNjaXRyYW5zbWVkIjtzOjU6InJlc2lkIjtzOjE1OiIxMi81MzMvZWFhdjM0OTEiO3M6NDoiYXRvbSI7czo1MDoiL21lZHJ4aXYvZWFybHkvMjAyNC8wMy8yOC8yMDI0LjAzLjI3LjI0MzA0ODY3LmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 51. 51.Ananworanich, J. et al. Virological and immunological characteristics of HIV-infected individuals at the earliest stage of infection. J Virus Erad 2, 43–48 (2016). 52. 52.Deeks, S. G. et al. Research priorities for an HIV cure: International AIDS Society Global Scientific Strategy 2021. Nat Med 27, 2085–2098 (2021). doi:10.1038/s41591-021-01590-5 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41591-021-01590-5&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=34848888&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F03%2F28%2F2024.03.27.24304867.atom) 53. 53.Gay, C. L. et al. Clinical Trial of the Anti-PD-L1 Antibody BMS-936559 in HIV-1 Infected Participants on Suppressive Antiretroviral Therapy. J Infect Dis 215, 1725–1733 (2017). doi:10.1093/infdis/jix191 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/infdis/jix191&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F03%2F28%2F2024.03.27.24304867.atom) 54. 54.Gay, C. L. et al. Suspected Immune-Related Adverse Events With an Anti-PD-1 Inhibitor in Otherwise Healthy People With HIV. J Acquir Immune Defic Syndr 87, e234–e236 (2021). doi:10.1097/QAI.0000000000002716 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1097/QAI.0000000000002716&link_type=DOI) 55. 55.Colston, E. et al. An open-label, multiple ascending dose study of the anti-CTLA-4 antibody ipilimumab in viremic HIV patients. PLoS One 13, e0198158 (2018). doi:10.1371/journal.pone.0198158 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1371/journal.pone.0198158&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F03%2F28%2F2024.03.27.24304867.atom) 56. 56.Reeves, D. B. et al. Impact of misclassified defective proviruses on HIV reservoir measurements. Nat Commun 14, 4186 (2023). doi:10.1038/s41467-023-39837-z [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41467-023-39837-z&link_type=DOI) 57. 57.Reeves, D. B. et al. A majority of HIV persistence during antiretroviral therapy is due to infected cell proliferation. Nat Commun 9, 4811 (2018). doi:10.1038/s41467-018-06843-5 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41467-018-06843-5&link_type=DOI) 58. 58.Shan, L. et al. Transcriptional Reprogramming during Effector-to-Memory Transition Renders CD4(+) T Cells Permissive for Latent HIV-1 Infection. Immunity 47, 766–775 e763 (2017). doi:10.1016/j.immuni.2017.09.014 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.immuni.2017.09.014&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=29045905&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F03%2F28%2F2024.03.27.24304867.atom) 59. 59.Jain, V. et al. 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 (2013). doi:10.1093/infdis/jit311 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/infdis/jit311&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=23852127&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F03%2F28%2F2024.03.27.24304867.atom) 60. 60.Henrich, T. J. et al. HIV-1 persistence following extremely early initiation of antiretroviral therapy (ART) during acute HIV-1 infection: An observational study. PLoS Med 14, e1002417 (2017). doi:10.1371/journal.pmed.1002417 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1371/journal.pmed.1002417&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=29112956&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F03%2F28%2F2024.03.27.24304867.atom) 61. 61.Chun, T. W. et al. Decay of the HIV reservoir in patients receiving antiretroviral therapy for extended periods: implications for eradication of virus. J Infect Dis 195, 1762–1764 (2007). doi:10.1086/518250 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1086/518250&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=17492591&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F03%2F28%2F2024.03.27.24304867.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000246986900006&link_type=ISI) 62. 62.Jain, V., Hartogensis, W., Bacchetti, P., Hunt, P.W., Hatano, H., Sinclair, E., Epling, L., Lee, T-H., Busch, M., McCune, J.M., Pilcher, C.D., Hecht, F.M., and Deeks, S.G. Antiretroviral therapy initiated within 6 months of HIV infection is associated with lower T-cell activation and smaller HIV reservoir size. Journal of Infectious Diseases In press (2013). 63. 63.Ho, Y. C. et al. Replication-competent noninduced proviruses in the latent reservoir increase barrier to HIV-1 cure. Cell 155, 540–551 (2013). doi:10.1016/j.cell.2013.09.020 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.cell.2013.09.020&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=24243014&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F03%2F28%2F2024.03.27.24304867.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000326571800010&link_type=ISI) 64. 64.Bruner, K. M. et al. Defective proviruses rapidly accumulate during acute HIV-1 infection. Nat Med 22, 1043–1049 (2016). doi:10.1038/nm.4156 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/nm.4156&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=27500724&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F03%2F28%2F2024.03.27.24304867.atom) 65. 65.Pinzone, M. R. et al. Longitudinal HIV sequencing reveals reservoir expression leading to decay which is obscured by clonal expansion. Nat Commun 10, 728 (2019). doi:10.1038/s41467-019-08431-7 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41467-019-08431-7&link_type=DOI) 66. 66.Gondim, M. V. P. et al. Heightened resistance to host type 1 interferons characterizes HIV-1 at transmission and after antiretroviral therapy interruption. Sci Transl Med 13 (2021). doi:10.1126/scitranslmed.abd8179 [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTE6InNjaXRyYW5zbWVkIjtzOjU6InJlc2lkIjtzOjE1OiIxMy81NzYvZWFiZDgxNzkiO3M6NDoiYXRvbSI7czo1MDoiL21lZHJ4aXYvZWFybHkvMjAyNC8wMy8yOC8yMDI0LjAzLjI3LjI0MzA0ODY3LmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 67. 67.Finzi, D. et al. Identification of a reservoir for HIV-1 in patients on highly active antiretroviral therapy. Science 278, 1295–1300 (1997). doi:10.1126/science.278.5341.1295 [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6Mzoic2NpIjtzOjU6InJlc2lkIjtzOjEzOiIyNzgvNTM0MS8xMjk1IjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjQvMDMvMjgvMjAyNC4wMy4yNy4yNDMwNDg2Ny5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 68. 68.Siliciano, J. D. et al. Long-term follow-up studies confirm the stability of the latent reservoir for HIV-1 in resting CD4+ T cells. Nature medicine 9, 727–728 (2003). doi:10.1038/nm880 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/nm880&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=12754504&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F03%2F28%2F2024.03.27.24304867.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000183444100027&link_type=ISI) 69. 69.Kumar, M. R. et al. Biphasic decay of intact SHIV genomes following initiation of antiretroviral therapy complicates analysis of interventions targeting the reservoir. Proc Natl Acad Sci U S A 120, e2313209120 (2023). doi:10.1073/pnas.2313209120 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1073/pnas.2313209120&link_type=DOI) 70. 70.Martin, A. R. et al. Similar Frequency and Inducibility of Intact Human Immunodeficiency Virus-1 Proviruses in Blood and Lymph Nodes. J Infect Dis 224, 258–268 (2021). doi:10.1093/infdis/jiaa736 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/infdis/jiaa736&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=33269401&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F03%2F28%2F2024.03.27.24304867.atom) 71. 71.Simonetti, F. R. et al. Intact proviral DNA assay analysis of large cohorts of people with HIV provides a benchmark for the frequency and composition of persistent proviral DNA. Proc Natl Acad Sci U S A 117, 18692–18700 (2020). doi:10.1073/pnas.2006816117 [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NDoicG5hcyI7czo1OiJyZXNpZCI7czoxMjoiMTE3LzMxLzE4NjkyIjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjQvMDMvMjgvMjAyNC4wMy4yNy4yNDMwNDg2Ny5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 72. 72.Bruner, K. M. et al. A quantitative approach for measuring the reservoir of latent HIV-1 proviruses. Nature 566, 120–125 (2019). doi:10.1038/s41586-019-0898-8 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41586-019-0898-8&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=30700913&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2024%2F03%2F28%2F2024.03.27.24304867.atom) [1]: /embed/graphic-1.gif [2]: /embed/inline-graphic-1.gif [3]: /embed/inline-graphic-2.gif [4]: /embed/graphic-2.gif [5]: /embed/inline-graphic-3.gif [6]: /embed/graphic-3.gif [7]: /embed/inline-graphic-4.gif [8]: /embed/inline-graphic-5.gif [9]: /embed/graphic-4.gif [10]: /embed/graphic-5.gif [11]: /embed/inline-graphic-6.gif [12]: /embed/inline-graphic-7.gif [13]: /embed/inline-graphic-8.gif