Host genomics of the HIV-1 reservoir size and its decay rate during suppressive antiretroviral treatment ======================================================================================================== * Christian W. Thorball * Alessandro Borghesi * Nadine Bachmann * Chantal von Siebenthal * Valentina Vongrad * Teja Turk * Kathrin Neumann * Niko Beerenwinkel * Jasmina Bogojeska * Volker Roth * Yik Lim Kok * Sonali Parbhoo * Mario Wieser * Jürg Böni * Matthieu Perreau * Thomas Klimkait * Sabine Yerly * Manuel Battegay * Andri Rauch * Patrick Schmid * Enos Bernasconi * Matthias Cavassini * Roger D. Kouyos * Huldrych F. Günthard * Karin J. Metzner * Jacques Fellay * Swiss HIV Cohort Study ## ABSTRACT **Introduction** A major hurdle to HIV-1 eradication is the establishment of a latent viral reservoir early after primary infection. Several factors are known to influence the HIV-1 reservoir size and decay rate on suppressive antiretroviral treatment (ART), but little is known about the role of human genetic variation. **Methods** We measured the reservoir size at three time points over a median of 5.4 years, and searched for associations between human genetic variation and two phenotypic readouts: the reservoir size at the first time point and its decay rate over the study period. We assessed the contribution of common genetic variants using genome-wide genotyping data from 797 patients with European ancestry enrolled in the Swiss HIV Cohort Study and searched for a potential impact of rare variants and exonic copy number variants using exome sequencing data generated in a subset of 194 study participants. **Results** Genome- and exome-wide analyses did not reveal any significant association with the size of the HIV-1 reservoir or its decay rate on suppressive ART. **Conclusions** Our results point to a limited influence of human genetics on the size of the HIV-1 reservoir and its long-term dynamics in successfully treated individuals. Keywords * HIV * genetics * ART * latent reservoir * GWAS * exome sequencing ## INTRODUCTION Combination antiretroviral treatment (ART) has turned the previously lethal infection by human immunodeficiency virus type 1 (HIV-1) into a chronic disease. Despite this significant achievement, HIV-1 as retrovirus, self-integrating its genome into the host chromosome, persists indefinitely in infected individuals during treatment [1–4], and life-long ART is required to control the infection. A major hurdle to HIV-1 eradication is the establishment, already during primary infection, of a latent viral reservoir of HIV-1 DNA persisting as provirus in resting memory CD4+ T cells [1,2,5–8]. At the molecular level, chromatin remodeling, epigenetic modifications, transcriptional interference, and availability of transcription factors have been considered as possible mechanisms contributing to HIV-1 latency [9]. The viral reservoir is measurable through different methods, including viral outgrowth assay and intracellular HIV-1 DNA quantification [10,11]. Currently, there is no consensus on the best HIV-1 reservoir biomarker. Total cell-associated HIV-1 DNA, easy to measure in different cell and tissue samples and applicable to large populations, has been shown to be a good proxy for the reservoir size [12]. Indeed, while HIV-1 DNA measurement is able to detect both integrated and nonintegrated viral genomes coding for intact or defective viruses [13], total HIV-1 DNA levels have been shown to correlate with viral outgrowth [14], and to predict the time to viral rebound at treatment interruption [15]. Moreover, the substantial loss of nonintegrated HIV-1 DNA genomes following ART initiation suggests that total HIV-1 DNA after prolonged suppression is largely accounted for by integrated viral genomes [16]. After an initial rapid decay following ART initiation, changes of the viral reservoir size over time display wide inter-individual variability. By limiting dilution culture assay, the half-life of the viral reservoir was first estimated to be 44 months (95% confidence interval 27.4-114.5) in individuals with undetectable viremia [4]. A more recent study showed a slow decline of total HIV-1 DNA with a half-life of 13 years after the first four years of suppressive ART [17]. Generally, different studies show a broad variability of the average decay rate, from 2.5 months to no measurable decay [18–26]. One study even reported an increase in the viral reservoir size in as much as 31% of patients in the 4-7 years following ART initiation [27], and recent data from our group confirm this observation, reporting an increase in the reservoir size in 26.8% of individuals in the 1.5-5.5 years after ART initiation [28]. Several factors are known to influence the decay rate of the viral reservoir: initiation of ART during acute HIV-1 infection substantially accelerates the decay rate, while viral blips and low-level viremia during ART slow it down, as shown in previous studies [22] and in recent data from our cohort [28]. Conversely, treatment intensification, i.e. treating with additional drugs, does not appear to influence the decay rate, suggesting that residual replication is not the main driver of the viral reservoir [29] or that it may happen in sanctuary sites. Human genetic variants have been shown to influence the outcome of various infections, including HIV. Previous genome-wide association studies (GWAS) addressed the role of common genetic polymorphisms in several HIV-related phenotypes, including plasma viral load (HIV-1 RNA) at set point, exceptional capacity to control viral replication, pace of CD4+ T lymphocyte decline, time to clinical AIDS, rapid progressor status or long-term non-progressor status (LTNP) [30–37], and, in one single study, the amount of intracellular HIV-1 DNA, measured at a single time point during the chronic phase of infection [38]. Rare genetic variants that are detectable through DNA sequencing technologies have been investigated far less. However, a large exome sequencing study did not reveal any convincing association of such variants with the natural history of HIV disease [39]. To date, no studies have addressed the role of human genetic variation in determining the initial viral reservoir size and the reservoir decay rate over time. In the current study, we searched for host genetic factors associated with the HIV-1 reservoir size and its long-term dynamics in a cohort of 797 HIV-1 positive individuals on suppressive ART for at least five years. ## METHODS ### Ethics statement Participants in the Swiss HIV Cohort Study (SHCS) consented to the cohort study and genetic analyses, as approved by the corresponding local Ethics Committees. ### Study participants The SHCS is an ongoing, nation-wide cohort study of HIV-positive individuals, including more than 70% of all persons living with HIV in Switzerland. Clinical and laboratory information has been prospectively recorded at follow-up visits every 3-6 months since 1988 [40]. The general enrolment criteria have been described previously [28]. Additionally, availability of genome-wide genotyping data from previous studies or of a DNA sample for genotyping was required for inclusion in this study (Figure 1). ![Figure 1.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2019/12/09/19013763/F1.medium.gif) [Figure 1.](http://medrxiv.org/content/early/2019/12/09/19013763/F1) Figure 1. Patient selection flowchart. Specific inclusion and exclusion criteria are listed for each selection step. ART (antiretroviral therapy); PBMCs (peripheral blood mononuclear cells); PI (protease inhibitor); PCA (principal component analysis). ### Quantification of total HIV-1 DNA The collection of longitudinal cryopreserved peripheral blood mononuclear cells (PBMCs) from eligible participants and the quantification of total HIV-1 DNA by droplet digital PCR has been described previously along with the calculation of the reservoir decay rate [28]. Briefly, this study utilized total HIV-1 DNA quantifications from three time points at a median of ∼1.5 years, ∼3.5 years, and ∼5.4 years after initiation of ART. ### Genotyping and genome-wide association analyses Genome-wide genotyping data were obtained from previous GWAS that used various microarrays, including the HumanCore-12, HumanHap550, Human610, Human1M and Infinium CoreExome-24 BeadChips (Illumina Inc., San Diego, CA, USA), or generated from DNA extracted from peripheral blood mononuclear cells using the HumanOmniExpress-24 BeadChip (Illumina Inc., San Diego, CA, USA). Genotypes from each genotyping array were filtered and imputed separately, with variants first flipped to the correct strand with BCFTOOLS (v1.8) according to the human GRCh37 reference genome and filtered based on a less than 20% deviation from the 1000 genomes phase 3 EUR reference panel. Genotypes were phased, and missing genotypes were imputed with EAGLE2 [41] and PBWT [42] respectively, using the 1000 Genomes Project Phase 3 reference panel on the Sanger Imputation Service [43]. Study participants were filtered based on European ancestry as determined by principal component analysis (PCA) using EIGENSTRAT (v6.1.4) [44] and the HapMap project [45] as reference populations (Figure S1A). Imputed variants were filtered by minor allele frequency (MAF) < 5%, missingness > 10%, deviation from Hardy-Weinberg equilibrium (PHWE < 1e-6) and imputation quality score (INFO < 0.8). The remaining genotypes were then combined using PLINK (v1.90b5) [46] prior to analyses. To carry out the GWASs, genome-wide genotypes were tested for association with each of the two study phenotypes (reservoir size or reservoir decay rate) in two separate genome-wide association analyses. Statistical significance was set to the standard genome-wide significance threshold of P < 5e-8 to correct for multiple testing. The associations were computed using linear mixed models with genetic relationship matrixes calculated between pairs of individuals according to the leave-one-chromosome-out method as implemented in GCTA mlma-loco (v1.91.4beta) [47,48], only including age and sex as covariates, to avoid masking of true associations by confounders. To further assess the contribution of variables previously shown to be associated with either reservoir size or decay rate, we ran multiple genome-wide association analyses, each including age, sex, and one single covariate, for each of the two study phenotypes. Finally, we conducted a GWAS including all the covariates except those showing mutual correlations. These covariates included time on ART, time to viral suppression, infection stage (acute or chronic), HIV-1 RNA pre-ART, last CD4+ T cell count pre-ART, HIV-1 subtype, transmission group, and occurrence of blips or low-level viremia during treatment. Classical HLA alleles at the four-digit level and variable amino acids within HLA proteins were imputed using SNP2HLA (v1.03) with the T1DGC reference panel consisting of 5,225 individuals of European ancestry [49]. Association analyses with the imputed HLA alleles and multi-allelic amino acids was performed using linear regressions in PLINK and multivariate omnibus tests, respectively. For all HLA analyses age, sex and the first principal component was included as covariates. Genotypes at specific loci, i.e. the CCR5Δ32 deletion (rs333) and the HLA-B*57:01 allele, known to influence the setpoint viral load (spVL) [50,51], available from genome-wide genotyping data, were tested for association with the reservoir size and its decay rate in 797 patients. High quality genotyping information on the CCR5Δ32 deletion was available for most individuals (N = 687), while all had available HLA information. ### Exome sequencing and analysis All coding exons were captured using either the Illumina Truseq 65 Mb enrichment kit (Illumina Inc., San Diego, CA, USA) or the IDT xGen Exome Research Panel v1.0 (Integrated DNA Technologies Inc., Coralville, IA, USA) and sequenced on the Illumina HiSeq4000. Sequence reads were aligned to the human reference genome (GRCh37) including decoys with BWA-MEM (v0.7.10) [52]. PCR duplicates were flagged using Picard tools (v2.18.14) and variant calling performed using GATK (v3.7) [53]. To ensure a high-quality variant set across capture kit batches, all samples were merged and variants filtered based on sequencing depth (DP ≥ 20) and genotype quality (GQ ≥ 30) using BCFTOOLS (v1.8). Furthermore, individual genotypes were set as missing in cases of low depth (DP < 10) or low quality (GQ < 20). The effect of the included variants was annotated with SnpEff (v4.3T) [54]. For single variant association analysis, the VCF file was converted to PLINK format using BCFTOOLS and PLINK. Only variants with a MAF above 5%, missingness per variant below 5% and absence of severe deviation from Hardy-Weinberg equilibrium (PHWE > 1e-6) were retained for the subsequent association analyses using PLINK. Sex, age and the first principal component were included as covariates. Only individuals of European descent were retained for the analyses, as determined by PCA (Figure S1B). The combined effect of rare protein-altering variants (MAF < 5%), defined as either missense, stop-gain, frameshift, essential splice variant or an indel by SnpEff, on the reservoir size and decay rate was analyzed using optimal sequence kernel association tests (SKAT-O) [55]. For the decay rate, individuals were split into two groups due to the non-normal distribution; one exhibiting a very high decay over time (< −0.03 −log10(DNA)) and another with a stable reservoir size (≥ −0.03 and ≤ 0.03 −log10(DNA)). For this case-control analysis we used the SKATbinary function with linear weighted variants as implemented in the SKAT R package. In both cases, the analyses were adjusted for age, sex, and the first principal component. Classical HLA class I and II alleles at the four-digit level were imputed from the exome sequencing data using HLA*LA [56]. All reads mapping to the MHC region or marked as unmapped were extracted using Samtools (v1.8) and used as input into HLA*LA. For association analyses, the 4-digit HLA alleles were extracted and analyzed using PyHLA [57] assuming an additive model, a minimum frequency of 5% and including age, sex and the first principal component as covariates. ### Copy number variation Copy number variations (CNVs) were called from exome sequencing data using CLAMMS [58]. CNVs were called for all samples in batches according to the exome capture kit used. Within batches, samples were normalized based on coverage and potential intra-batch effects adjusted for through the use of recommended mapping metrics extracted with Picard tools (v2.18.14). After CNV calling, samples with the number of CNVs two times above the median were excluded (N = 2). CNV association analyses were performed for duplications and deletions separately for common CNVs (frequency > 5 %) with PLINK adjusting for age and sex. Potential rare CNVs (frequency < 5%) impacting immune related genes were examined by overlapping called CNVs with curated immune-related genes from Immport [59] which were also listed as protein coding in GENCODE (v25). ### Statistical analyses All statistical analyses were performed using the R statistical software (v3.5.2), unless otherwise specified. ## RESULTS ### Host genetic determinants of the reservoir size and long-term dynamics To investigate the effects of host genetic variation on the size of the HIV-1 reservoir 1.5 years after ART initiation and its long-term dynamics under ART over a median duration of 5.4 years, we performed a GWAS, including 797 well-characterized HIV-1 positive individuals. All study participants were enrolled in the SHCS and were of European ancestry with longitudinal total HIV-1 DNA measurements available (Table 1). The median HIV-1 reservoir size was 2.76 (IQR: 2.48-3.03) log10 total HIV-1 DNA copies/1 million genomic equivalents measured ∼1.5 years after initiation of ART (Figure S2A). The median decay rate between 1.5-5.4 years after initiation of ART was −0.06 (IQR: −0.12-0.00) log10 total HIV-1 DNA copies/1 million genomic equivalents per year (Figure S2B). With our sample size we had 80% power to detect variants with a MAF of 10% explaining at least 5% of the variance in HIV-1 reservoir size or decay rate [60]. View this table: [Table 1.](http://medrxiv.org/content/early/2019/12/09/19013763/T1) Table 1. Patient characteristics First, we performed GWAS using age and sex as covariates. We did not observe any genome-wide significant variant (P < 5e-8) associated with either HIV-1 reservoir size or long-term dynamics (Figure 2, S3). However, as we have previously determined, multiple factors are associated with the HIV-1 reservoir size and its decay rate [28], some of which are correlated (Figure S4). Thus, we performed additional analyses iteratively including these factors to test whether they could mask genetic associations. We ran multiple GWAS each adjusting for age, sex, plus one of the associated covariates, as well as all of the covariates together. The addition of the covariates did not have any significant effect on the results nor the genome-wide inflation factor (lambda) (Table S1). ![Figure 2.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2019/12/09/19013763/F2.medium.gif) [Figure 2.](http://medrxiv.org/content/early/2019/12/09/19013763/F2) Figure 2. Association results with HIV-1 reservoir size. Manhattan plot with association p-values (-log10(P)) per genetic variant plotted by genomic position. Dashed line indicates the threshold for genome-wide significance (P = 5e-8). No variants were found to be genome-wide significant. Genetic variation in the HLA region has previously been associated with multiple HIV-related outcomes, including spVL [51]. To test whether specific HLA variants were associated with reservoir size or long-term dynamics, we imputed the HLA alleles and amino acids for all 797 individuals from the genotyping data. In line with the previous results, we did not observe any genome-wide significant associations with any HLA allele or amino acid. ### Impact of protein-coding and rare variants To assess the impact of rare variants as well as protein-coding variants missed by genotyping arrays on the HIV-1 reservoir size and long-term dynamics, we performed exome sequencing in 194 of the 797 study participants. Patients were selected at the two extremes of the observed reservoir decay rate: either very rapid, or absent (no change in reservoir size over ∼5.4 years), while individuals with increasing HIV-1 reservoir sizes were excluded (N=12). Thus, the long-term dynamics phenotype was binarized for subsequent analyses of the decay rate, while the HIV-1 reservoir size phenotype remained normally distributed (Figure S5). To ensure that no common variants, missed by the genotype chips, were associated with the HIV-1 reservoir size or long-term dynamics, we performed a GWAS for common variants using age and sex as covariates. As with the genotyping data, we observed no genome-wide significant variants for either phenotype (Figure S6). We then examined the potential role of rare variants (MAF < 5%) with a functional impact defined as either missense, frameshift, stop gained, splice acceptor or donor. Since HIV-1 primarily infects CD4+ T cells, we only included variants within genes expressed in these cells as determined by Gutierrez-Arcelus *et al*. [61]. The significance threshold after correcting for the number of tests performed was P = 1.21e-5. We did not observe any significant associations for either the HIV-1 reservoir or the decay rate. The *AMBRA1* gene showed the strongest association with HIV-1 reservoir size (P = 4.15e-5, not significant) (Figure S7). To confirm the lack of HLA association seen with the genotyping data, we imputed the HLA haplotypes from the exome data using HLA*LA. Again, we did not observe any significant HLA association with the study outcomes. ### Copy number variations To examine the role of large exonic CNVs not captured by standard genotyping and exome pipelines, we called CNVs from the mapped sequencing reads of the exome samples using the software CLAMMS. The contribution of common CNVs to HIV-1 reservoir size and long-term dynamics was analyzed by association analyses including age, sex and the first principal component as covariates. No significant association was observed after Bonferroni correction (Figure S8). We also searched for rare CNVs in curated immune-related genes from Immport [59] but did not discover any suggestive immune-related CNVs. ### Influence of HLA-B*57:01 and the CCR5Δ32 deletion on reservoir size and long-term dynamics We have previously shown that pre-ART RNA viral load levels are associated with the HIV-1 reservoir size and the occurrence of blips [28]. The HLA-B*57:01 allele and the CCR5Δ32 deletion are well known genetic variants influencing HIV-1 spVL [50,51], and could thus also be associated with the with the HIV-1 reservoir size or its decay rate. However, we did not observe any nominal association (all P > 0.05) with either reservoir size or its long-term dynamics for HLA-B*57:01 and CCR5Δ32 (Figure S9). ## DISCUSSION We used a combination of genomic technologies to assess the potential role of human genetic factors in determining both the HIV-1 reservoir size and its long-term dynamics in a well-characterized, population-based cohort. We studied 797 HIV-1-positive individuals of European origin under suppressive ART over a median of 5.4 years, for whom extensive clinical data are available, allowing detailed characterization and correction for potential confounders [28]. We measured the HIV-1 reservoir size at three time points and selected two phenotypes for our genomic study: the reservoir size at ∼1.5 years after ART initiation and the slope of the reservoir decay rate calculated over the three time points. Previous HIV host genetic studies mostly focused on phenotypes reflecting the natural history of HIV-1 infection, prior to ART initiation, including spontaneous viral control and disease progression [30–37]. A single study specifically tested for associations between common genetic variants and the amount of intracellular HIV-1 DNA, measured at a single time point during the chronic phase of infection prior to initiation of any antiretroviral therapy [38]. Here, in contrast, we longitudinally assessed samples collected from patients under suppressive ART to search for human genetic determinants of the long-term dynamics of the HIV-1 reservoir during treatment. We first conducted a GWAS on 797 individuals to test for association between common genetic variants and the phenotypes. Given the small proportion of non-European subjects in the initial study cohort, we only included patients of European ancestry to avoid any false positive associations or masking of true positive associations due to different allele frequencies in small proportions of individuals belonging to different subpopulations (Figure 1) [62]. Regardless of including or not independent covariates other than the standard ones (i.e., sex and age), no genetic variant reached the genome-wide significance threshold for association with any of the two phenotypes. This may reflect a small effect size of genetic variants on the HIV-1 reservoir size and decay rate. We acknowledge that a larger sample size and thus increased statistical power may allow detecting genetic variants with a smaller effect size associated with the phenotypes. However, it should be noted that this study is by far the largest today that has investigated the size and decay of the HIV-reservoir in well characterized and well suppressed HIV-positive individuals over a longer time period. Alternatively, the control of the HIV-1 reservoir size and its long-term dynamics may be under the control of viral or host factors other than the individual germline genetic background. A previous report from our group had shown a correlation between viral blips during the first 1.5 years of suppressive ART and the HIV-1 reservoir size 1.5 years after ART initiation, and between viral blips after 1.5-5.4 years of suppressive ART or low-level viremia and a slower decay rate [28]. Importantly, viral blips are generally thought to reflect transient increases in viral replication, and probably occur under multifactorial influence from viral and host factors [63–70], with these latter possibly including, but not being limited to, germline genetic variation. The biological relations between viral reservoir, decay rate, viral blips, and the contribution of the individual genetic background still need full elucidation. Standard GWAS is designed to detect associations with common genetic variants (i.e., with a MAF of at least 0.05), with little power to investigate the role of rare variants. Thus, to further assess the contribution of rare variants in individuals at the extreme of the decay rate distribution, we used exome sequencing in a selected subset of 194 study participants with very high decay rate, or conversely, a stable reservoir size over time (Figure S5). Here again, our analyses did not detect significant associations with the phenotypes. Although not reaching statistical significance, a rare genetic variant with potential functional impact in *AMBRA1* had a p-value for association just below the corrected threshold. The expression of *AMBRA1*, a core component of the autophagy machinery, has previously been associated with long-term viral control in HIV-1 non-progressors [71]. Future studies may further elucidate whether genetic variation in *AMBRA1* may account for inter-individual differences in the long-term dynamics of the HIV-1 reservoir. Large deletions or duplications of genomic material may be implicated in human phenotypes, with CNVs impacting the exonic regions being more likely to have a functional role. Thus, we further investigated whether any common or rare CNV spanning exonic regions was associated with the phenotype. Again, no CNV was statistically associated with the phenotypes both in the exome-wide analyses and in analyses focused on immune-related genes. An inherent limitation of our exome-based association analyses was their inability to detect rare variants outside the coding or splice-site regions. The exonic regions account for approximately 1-2% of the whole human genome. Because many regulatory sequences are located in extra-genic sites, our analysis did not fully investigate the role of highly conserved, non-coding genetic regions in influencing the phenotypes linked to HIV-1 latency. Additionally, we focused on specific genetic variants, i.e., the HLA haplotypes and the CCR5Δ32 deletion, previously demonstrated to have a role in HIV-1 related phenotypes [30,51]. Indeed, previous studies unraveled a robust association between variation in the HLA region and the HIV-1 spVL [30]. Likewise, heterozygosity for the CCR5Δ32 deletion has been shown to influence spontaneous HIV-1 control [51]. Thus, we imputed HLA genotypes from genotyping and exome data, and studied the CCR5Δ32 deletion, without, however, detecting any significant associations with the phenotypes or the covariates (Figure S9). Specifically, we found no correlation between HLA genotypes and HIV-1 RNA plasma levels prior to ART initiation, apparently contrasting with the previous findings of an association between HLA-B*57:01 haplotype and spVL. This probably reflects historical changes in the therapeutic approach following a diagnosis of HIV-1 infection, given that ART is currently initiated soon after clinical diagnosis, before most patients reach a stable plateau of plasma viral load. In our study, the quantification of the reservoir size at different time points may have been influenced by factors as, for example, blips and low-level viremia, which may have reduced our ability to detect significant genetic effects. It is also possible that, in the future, novel methods to assess the viral reservoir will allow the detection of significant contributions of genetic factors [72]. So far, it remains unanswered whether the initial response to acute infection, the containment of ongoing replication, and the control of latently infected cells are under the influence of the same or different molecular networks. It needs to be noted that in previous work we have shown that host genetic factors as defined by GWAS did not explain the severity of symptoms during acute HIV-infection, although severity of symptoms correlated well with viral load and CD4 cell counts [73]. In conclusion, our study suggests that human individual germline genetic variation has little, if any, influence on the control of the HIV-1 viral reservoir size and its long-term dynamics. Complex, likely multifactorial biological processes govern HIV-1 viral persistence. Larger studies will possibly clarify the role of common or rare genetic variants explaining small proportions of the variability of the phenotypes related to viral latency. ## Data Availability The datasets generated during and/or analyzed during the current study are not publicly available due to privacy reasons, the sensitivities associated with HIV infections, and the representativeness of the dataset, but is available on request. ## Data availability The datasets generated during and/or analyzed during the current study are not publicly available due to privacy reasons, the sensitivities associated with HIV infections, and the representativeness of the dataset, but is available on request. ## Conflicts of interest statement H.F.G. has received unrestricted research grants from Gilead Sciences and Roche; fees for data and safety monitoring board membership from Merck; consulting/advisory board membership fees from Gilead Sciences, ViiV, Merck, Sandoz and Mepha. T.K. has received consulting/advisory board membership fees from Gilead Sciences and from ViiV Healthcare for work that has no connection to the work presented here. K.J.M. has received travel grants and honoraria from Gilead Sciences, Roche Diagnostics, GlaxoSmithKline, Merck Sharp & Dohme, Bristol-Myers Squibb, ViiV and Abbott; and the University of Zurich received research grants from Gilead Science, Roche, and Merck Sharp & Dohme for studies that Dr. Metzner serves as principal investigator, and advisory board honoraria from Gilead Sciences. A.R. reports support to his institution for advisory boards and/or travel grants from MSD, Gilead Sciences, Pfizer and Abbvie, and an investigator initiated trial (IIT) grant from Gilead Sciences. All remuneration went to his home institution and not to A.R. personally, and all remuneration was provided outside the submitted work. All other authors declare no competing financial interests. ## Author contributions N.B., J.B., V.R., R.D.K., H.F.G., K.J.M., and J.F. contributed to the conception and design of the study. C.v.S., V.V., K.N., Y.I.K., and K.J.M. contributed to the acquisition of data. A.B., C.W.T., N.B., T.T., S.P., M.W., R.D.K., and J.F. contributed to the analysis and interpretation of data. J.B., M.P., T.K., S.Y., M.B., A.R., P.S., E.B., M.C., H.F.G., and the members of the Swiss HIV Cohort Study (SHCS) conceived and managed the cohort, collected and contributed patient samples and clinical data. A.B., C.W.T., and J.F. contributed to the drafting the article. All authors read and approved the final manuscript. ## Acknowledgements We thank the patients for participating in the SHCS, the study nurses and physicians for excellent patient care, A. Scherrer, A. Traytel, and S. Wild for excellent data management and D. Perraudin and M. Amstutz for administrative assistance. This work was funded within the framework of the Swiss HIV Cohort Study (SNF grant# 33CS30_177499 to H.F.G.). The data were gathered by the Five Swiss University Hospitals, two Cantonal Hospitals, 15 affiliated hospitals and 36 private physicians (listed in [http://www.shcs.ch/180-health-care-providers](http://www.shcs.ch/180-health-care-providers)). The work was furthermore supported by the Systems.X grant # 51MRP0\_158328 (to N.B., J.B., J.F.,V.R., R.D.K., M.W., H.F.G. and K.J.M.), by SNF grant 324730B_179571 (to H.F.G.), SNF grant SNF 310030_141067/1 (to H.F.G. and K.J.M.), SNF grants no. PZ00P3-142411 and BSSGI0_155851 to R.D.K., the Yvonne-Jacob Foundation (to H.F.G.), the University of Zurich’s Clinical Research Priority Program viral infectious disease, ZPHI (to H.F.G) and the Vontobel Foundation (to H.F.G. and K.J.M.). M.W. is partially supported by the NCCR MARVEL, funded by the Swiss National Science Foundation. Members of the Swiss HIV Cohort Study: Anagnostopoulos A, Battegay M, Bernasconi E, Böni J, Braun DL, Bucher HC, Calmy A, Cavassini M, Ciuffi A, Dollenmaier G, Egger M, Elzi L, Fehr J, Fellay J, Furrer H, Fux CA, Günthard HF (President of the SHCS), Haerry D (deputy of “Positive Council”), Hasse B, Hirsch HH, Hoffmann M, Hösli I, Huber M, Kahlert CR (Chairman of the Mother & Child Substudy), Kaiser L, Keiser O, Klimkait T, Kouyos RD, Kovari H, Ledergerber B, Martinetti G, Martinez de Tejada B, Marzolini C, Metzner KJ, Müller N, Nicca D, Paioni P, Pantaleo G, Perreau M, Rauch A (Chairman of the Scientific Board), Rudin C, Scherrer AU (Head of Data Centre), Schmid P, Speck R, Stöckle M (Chairman of the Clinical and Laboratory Committee), Tarr P, Trkola A, Vernazza P, Wandeler G, Weber R, Yerly S. * Received December 2, 2019. * Revision received December 9, 2019. * Accepted December 9, 2019. * © 2019, Posted by Cold Spring Harbor Laboratory This pre-print is available under a Creative Commons License (Attribution-NonCommercial 4.0 International), CC BY-NC 4.0, as described at [http://creativecommons.org/licenses/by-nc/4.0/](http://creativecommons.org/licenses/by-nc/4.0/) ## REFERENCES 1. 1.Finzi D, Hermankova M, Pierson T, Carruth LM, Buck C, Chaisson RE, et al. Identification of a Reservoir for HIV-1 in Patients on Highly Active Antiretroviral Therapy. Science. 1997 Nov 14;278(5341):1295–300. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6Mzoic2NpIjtzOjU6InJlc2lkIjtzOjEzOiIyNzgvNTM0MS8xMjk1IjtzOjQ6ImF0b20iO3M6Mzk6Ii9tZWRyeGl2L2Vhcmx5LzIwMTkvMTIvMDkvMTkwMTM3NjMuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 2. 2.Wong JK, Hezareh M, Günthard HF, Havlir DV, Ignacio CC, Spina CA, et al. Recovery of Replication-Competent HIV Despite Prolonged Suppression of Plasma Viremia. Science. 1997 Nov 14;278(5341):1291–5. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6Mzoic2NpIjtzOjU6InJlc2lkIjtzOjEzOiIyNzgvNTM0MS8xMjkxIjtzOjQ6ImF0b20iO3M6Mzk6Ii9tZWRyeGl2L2Vhcmx5LzIwMTkvMTIvMDkvMTkwMTM3NjMuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 3. 3.Chun T-W, Stuyver L, Mizell SB, Ehler LA, Mican JAM, Baseler M, et al. Presence of an inducible HIV-1 latent reservoir during highly active antiretroviral therapy. Proc Natl Acad Sci. 1997 Nov 25;94(24):13193–7. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NDoicG5hcyI7czo1OiJyZXNpZCI7czoxMToiOTQvMjQvMTMxOTMiO3M6NDoiYXRvbSI7czozOToiL21lZHJ4aXYvZWFybHkvMjAxOS8xMi8wOS8xOTAxMzc2My5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 4. 4.Siliciano JD, Kajdas J, Finzi D, Quinn TC, Chadwick K, Margolick JB, et al. Long-term follow-up studies confirm the stability of the latent reservoir for HIV-1 in resting CD4+ T cells. Nat Med. 2003 Jun;9(6):727–8. [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%2F2019%2F12%2F09%2F19013763.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000183444100027&link_type=ISI) 5. 5.Chun TW, Carruth L, Finzi D, Shen X, DiGiuseppe JA, Taylor H, et al. Quantification of latent tissue reservoirs and total body viral load in HIV-1 infection. Nature. 1997 May 8;387(6629):183–8. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/387183a0&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=9144289&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2019%2F12%2F09%2F19013763.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=A1997WX94500055&link_type=ISI) 6. 6.Chun TW, Engel D, Berrey MM, Shea T, Corey L, Fauci AS. Early establishment of a pool of latently infected, resting CD4(+) T cells during primary HIV-1 infection. Proc Natl Acad Sci U S A. 1998 Jul 21;95(15):8869–73. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NDoicG5hcyI7czo1OiJyZXNpZCI7czoxMDoiOTUvMTUvODg2OSI7czo0OiJhdG9tIjtzOjM5OiIvbWVkcnhpdi9lYXJseS8yMDE5LzEyLzA5LzE5MDEzNzYzLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 7. 7.Smith MZ, Wightman F, Lewin SR. HIV reservoirs and strategies for eradication. Curr HIV/AIDS Rep. 2012 Mar;9(1):5–15. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1007/s11904-011-0108-2&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=22249405&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2019%2F12%2F09%2F19013763.atom) 8. 8.Siliciano RF, Greene WC. HIV latency. Cold Spring Harb Perspect Med. 2011 Sep;1(1):a007096. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTQ6ImNzaHBlcnNwZWN0bWVkIjtzOjU6InJlc2lkIjtzOjExOiIxLzEvYTAwNzA5NiI7czo0OiJhdG9tIjtzOjM5OiIvbWVkcnhpdi9lYXJseS8yMDE5LzEyLzA5LzE5MDEzNzYzLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 9. 9.Ruelas DS, Greene WC. An integrated overview of HIV-1 latency. Cell. 2013 Oct 24;155(3):519–29. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.cell.2013.09.044&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=24243012&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2019%2F12%2F09%2F19013763.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000326571800008&link_type=ISI) 10. 10.Han Y, Wind-Rotolo M, Yang H-C, Siliciano JD, Siliciano RF. Experimental approaches to the study of HIV-1 latency. Nat Rev Microbiol. 2007 Feb;5(2):95–106. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/nrmicro1580&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=17224919&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2019%2F12%2F09%2F19013763.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000243520400009&link_type=ISI) 11. 11.Hodel F, Patxot M, Snäkä T, Ciuffi A. HIV-1 latent reservoir: size matters. Future Virol. 2016 Dec;11(12):785–94. 12. 12.Avettand-Fènoël V, Hocqueloux L, Ghosn J, Cheret A, Frange P, Melard A, et al. Total HIV-1 DNA, a Marker of Viral Reservoir Dynamics with Clinical Implications. Clin Microbiol Rev. 2016 Oct;29(4):859–80. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiY21yIjtzOjU6InJlc2lkIjtzOjg6IjI5LzQvODU5IjtzOjQ6ImF0b20iO3M6Mzk6Ii9tZWRyeGl2L2Vhcmx5LzIwMTkvMTIvMDkvMTkwMTM3NjMuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 13. 13.Bruner KM, Wang Z, Simonetti FR, Bender AM, Kwon KJ, Sengupta S, et al. A quantitative approach for measuring the reservoir of latent HIV-1 proviruses. Nature. 2019;566(7742):120–5. [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%2F2019%2F12%2F09%2F19013763.atom) 14. 14.Kiselinova M, De Spiegelaere W, Buzon MJ, Malatinkova E, Lichterfeld M, Vandekerckhove L. Integrated and Total HIV-1 DNA Predict Ex Vivo Viral Outgrowth. PLoS Pathog. 2016 Mar;12(3):e1005472. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1371/journal.ppat.1005472&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=26938995&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2019%2F12%2F09%2F19013763.atom) 15. 15.Williams JP, Hurst J, Stöhr W, Robinson N, Brown H, Fisher M, et al. HIV-1 DNA predicts disease progression and post-treatment virological control. eLife. 2014 Sep 12;3:e03821. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.7554/eLife.03821&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=25217531&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2019%2F12%2F09%2F19013763.atom) 16. 16.Koelsch KK, Liu L, Haubrich R, May S, Havlir D, Günthard HF, et al. Dynamics of total, linear nonintegrated, and integrated HIV-1 DNA in vivo and in vitro. J Infect Dis. 2008 Feb 1;197(3):411–9. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1086/525283&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=18248304&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2019%2F12%2F09%2F19013763.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000253131600013&link_type=ISI) 17. 17.Gandhi RT, McMahon DK, Bosch RJ, Lalama CM, Cyktor JC, Macatangay BJ, et al. Levels of HIV-1 persistence on antiretroviral therapy are not associated with markers of inflammation or activation. PLOS Pathog. 2017 Apr 20;13(4):e1006285. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1371/journal.ppat.1006285&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=28426825&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2019%2F12%2F09%2F19013763.atom) 18. 18.Zanchetta M, Walker S, Burighel N, Bellanova D, Rampon O, Giaquinto C, et al. Long-term decay of the HIV-1 reservoir in HIV-1-infected children treated with highly active antiretroviral therapy. J Infect Dis. 2006 Jun 15;193(12):1718–27. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1086/504264&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=16703516&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2019%2F12%2F09%2F19013763.atom) 19. 19.Chun T-W, Justement JS, Moir S, Hallahan CW, Maenza J, Mullins JI, et al. Decay of the HIV reservoir in patients receiving antiretroviral therapy for extended periods: implications for eradication of virus. J Infect Dis. 2007 Jun 15;195(12):1762–4. [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%2F2019%2F12%2F09%2F19013763.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000246986900006&link_type=ISI) 20. 20.Strain MC, Günthard HF, Havlir DV, Ignacio CC, Smith DM, Leigh-Brown AJ, et al. Heterogeneous clearance rates of long-lived lymphocytes infected with HIV: intrinsic stability predicts lifelong persistence. Proc Natl Acad Sci U S A. 2003 Apr 15;100(8):4819–24. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NDoicG5hcyI7czo1OiJyZXNpZCI7czoxMDoiMTAwLzgvNDgxOSI7czo0OiJhdG9tIjtzOjM5OiIvbWVkcnhpdi9lYXJseS8yMDE5LzEyLzA5LzE5MDEzNzYzLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 21. 21.Izopet J, Salama G, Pasquier C, Sandres K, Marchou B, Massip P, et al. Decay of HIV-1 DNA in patients receiving suppressive antiretroviral therapy. J Acquir Immune Defic Syndr Hum Retrovirology Off Publ Int Retrovirology Assoc. 1998 Dec 15;19(5):478–83. 22. 22.Ramratnam B, Mittler JE, Zhang L, Boden D, Hurley A, Fang F, et al. The decay of the latent reservoir of replication-competent HIV-1 is inversely correlated with the extent of residual viral replication during prolonged anti-retroviral therapy. Nat Med. 2000 Jan;6(1):82–5. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/71577&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=10613829&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2019%2F12%2F09%2F19013763.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000084583300040&link_type=ISI) 23. 23.van Rij RP, van Praag RME, Prins JM, Rientsma R, Jurriaans S, Lange JMA, et al. Persistence of viral HLA-DR-CD4 T-cell reservoir during prolonged treatment of HIV-1 infection with a five-drug regimen. Antivir Ther. 2002 Mar;7(1):37–41. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=12008786&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2019%2F12%2F09%2F19013763.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000175079500005&link_type=ISI) 24. 24.Pires A, Hardy G, Gazzard B, Gotch F, Imami N. Initiation of antiretroviral therapy during recent HIV-1 infection results in lower residual viral reservoirs. J Acquir Immune Defic Syndr 1999. 2004 Jul 1;36(3):783–90. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1097/00126334-200407010-00004&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=15213561&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2019%2F12%2F09%2F19013763.atom) 25. 25.Strain MC, Little SJ, Daar ES, Havlir DV, Gunthard HF, Lam RY, et al. Effect of treatment, during primary infection, on establishment and clearance of cellular reservoirs of HIV-1. J Infect Dis. 2005 May 1;191(9):1410–8. [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%2F2019%2F12%2F09%2F19013763.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000228128800006&link_type=ISI) 26. 26.Fischer M, Joos B, Niederöst B, Kaiser P, Hafner R, von Wyl V, et al. Biphasic decay kinetics suggest progressive slowing in turnover of latently HIV-1 infected cells during antiretroviral therapy. Retrovirology. 2008 Nov 26;5:107. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1186/1742-4690-5-107&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=19036147&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2019%2F12%2F09%2F19013763.atom) 27. 27.Besson GJ, Lalama CM, Bosch RJ, Gandhi RT, Bedison MA, Aga E, et al. HIV-1 DNA decay dynamics in blood during more than a decade of suppressive antiretroviral therapy. Clin Infect Dis Off Publ Infect Dis Soc Am. 2014 Nov;59(9):1312–21. 28. 28.Bachmann N, Siebenthal C von, Vongrad V, Turk T, Neumann K, Beerenwinkel N, et al. Determinants of HIV-1 reservoir size and long-term dynamics during suppressive ART. Nat Commun. 2019 Jul 19;10(1):1–11. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41467-019-09078-0&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=30602773&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2019%2F12%2F09%2F19013763.atom) 29. 29.International AIDS Society Scientific Working Group on HIV Cure, Deeks SG, Autran B, Berkhout B, Benkirane M, Cairns S, et al. Towards an HIV cure: a global scientific strategy. Nat Rev Immunol. 2012 20;12(8):607–14. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/nri3262&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=22814509&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2019%2F12%2F09%2F19013763.atom) 30. 30.Fellay J, Shianna KV, Ge D, Colombo S, Ledergerber B, Weale M, et al. A whole-genome association study of major determinants for host control of HIV-1. Science. 2007 Aug 17;317(5840):944–7. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6Mzoic2NpIjtzOjU6InJlc2lkIjtzOjEyOiIzMTcvNTg0MC85NDQiO3M6NDoiYXRvbSI7czozOToiL21lZHJ4aXYvZWFybHkvMjAxOS8xMi8wOS8xOTAxMzc2My5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 31. 31.Fellay J, Ge D, Shianna KV, Colombo S, Ledergerber B, Cirulli ET, et al. Common genetic variation and the control of HIV-1 in humans. PLoS Genet. 2009 Dec;5(12):e1000791. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1371/journal.pgen.1000791&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=20041166&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2019%2F12%2F09%2F19013763.atom) 32. 32.Pelak K, Goldstein DB, Walley NM, Fellay J, Ge D, Shianna KV, et al. Host determinants of HIV-1 control in African Americans. J Infect Dis. 2010 Apr 15;201(8):1141–9. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1086/651382&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=20205591&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2019%2F12%2F09%2F19013763.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000275493400006&link_type=ISI) 33. 33.International HIV Controllers Study, Pereyra F, Jia X, McLaren PJ, Telenti A, de Bakker PIW, et al. The major genetic determinants of HIV-1 control affect HLA class I peptide presentation. Science. 2010 Dec 10;330(6010):1551–7. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6Mzoic2NpIjtzOjU6InJlc2lkIjtzOjEzOiIzMzAvNjAxMC8xNTUxIjtzOjQ6ImF0b20iO3M6Mzk6Ii9tZWRyeGl2L2Vhcmx5LzIwMTkvMTIvMDkvMTkwMTM3NjMuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 34. 34.Herbeck JT, Gottlieb GS, Winkler CA, Nelson GW, An P, Maust BS, et al. Multistage genomewide association study identifies a locus at 1q41 associated with rate of HIV-1 disease progression to clinical AIDS. J Infect Dis. 2010 Feb 15;201(4):618–26. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1086/649842&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=20064070&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2019%2F12%2F09%2F19013763.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000273843900019&link_type=ISI) 35. 35.Le Clerc S, Limou S, Coulonges C, Carpentier W, Dina C, Taing L, et al. Genomewide association study of a rapid progression cohort identifies new susceptibility alleles for AIDS (ANRS Genomewide Association Study 03). J Infect Dis. 2009 Oct 15;200(8):1194–201. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1086/605892&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=19754311&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2019%2F12%2F09%2F19013763.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000270089400003&link_type=ISI) 36. 36.Limou S, Le Clerc S, Coulonges C, Carpentier W, Dina C, Delaneau O, et al. Genomewide association study of an AIDS-nonprogression cohort emphasizes the role played by HLA genes (ANRS Genomewide Association Study 02). J Infect Dis. 2009 Feb 1;199(3):419–26. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1086/596067&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=19115949&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2019%2F12%2F09%2F19013763.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000262326600018&link_type=ISI) 37. 37.Limou S, Coulonges C, Herbeck JT, van Manen D, An P, Le Clerc S, et al. Multiple-cohort genetic association study reveals CXCR6 as a new chemokine receptor involved in long-term nonprogression to AIDS. J Infect Dis. 2010 Sep 15;202(6):908–15. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1086/655782&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=20704485&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2019%2F12%2F09%2F19013763.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000281091200013&link_type=ISI) 38. 38.Dalmasso C, Carpentier W, Meyer L, Rouzioux C, Goujard C, Chaix M-L, et al. Distinct genetic loci control plasma HIV-RNA and cellular HIV-DNA levels in HIV-1 infection: the ANRS Genome Wide Association 01 study. PloS One. 2008;3(12):e3907. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1371/journal.pone.0003907&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=19107206&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2019%2F12%2F09%2F19013763.atom) 39. 39.McLaren PJ, Pulit SL, Gurdasani D, Bartha I, Shea PR, Pomilla C, et al. Evaluating the Impact of Functional Genetic Variation on HIV-1 Control. J Infect Dis. 2017 27;216(9):1063–9. 40. 40.Schoeni-Affolter F, Ledergerber B, Rickenbach M, Rudin C, Günthard HF, Telenti A, et al. Cohort Profile: The Swiss HIV Cohort Study. Int J Epidemiol. 2010 Oct 1;39(5):1179–89. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/ije/dyp321&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=19948780&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2019%2F12%2F09%2F19013763.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000283932000011&link_type=ISI) 41. 41.Loh P-R, Danecek P, Palamara PF, Fuchsberger C, Reshef YA, Finucane HK, et al. Reference-based phasing using the Haplotype Reference Consortium panel. Nat Genet. 2016 Nov;48(11):1443–8. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/ng.3679&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=27694958&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2019%2F12%2F09%2F19013763.atom) 42. 42.Durbin R. Efficient haplotype matching and storage using the positional Burrows– Wheeler transform (PBWT). Bioinformatics. 2014 May 1;30(9):1266–72. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/bioinformatics/btu014&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=24413527&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2019%2F12%2F09%2F19013763.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000336095100011&link_type=ISI) 43. 43.McCarthy S, Das S, Kretzschmar W, Delaneau O, Wood AR, Teumer A, et al. A reference panel of 64,976 haplotypes for genotype imputation. Nat Genet [Internet]. 2016 Aug 22 [cited 2016 Aug 31];advance online publication. Available from: [http://www.nature.com/ng/journal/vaop/ncurrent/full/ng.3643.html](http://www.nature.com/ng/journal/vaop/ncurrent/full/ng.3643.html) 44. 44.Price AL, Patterson NJ, Plenge RM, Weinblatt ME, Shadick NA, Reich D. Principal components analysis corrects for stratification in genome-wide association studies. Nat Genet. 2006 Aug;38(8):904–9. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/ng1847&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=16862161&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2019%2F12%2F09%2F19013763.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000239325700019&link_type=ISI) 45. 45.The International HapMap 3 Consortium. Integrating common and rare genetic variation in diverse human populations. Nature. 2010 Sep;467(7311):52–8. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/nature09298&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=20811451&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2019%2F12%2F09%2F19013763.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000281461200033&link_type=ISI) 46. 46.Chang CC, Chow CC, Tellier LC, Vattikuti S, Purcell SM, Lee JJ. Second-generation PLINK: rising to the challenge of larger and richer datasets. GigaScience. 2015 Dec 1;4(1):1–16. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1186/2047-217X-4-1&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=25838885&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2019%2F12%2F09%2F19013763.atom) 47. 47.Yang J, Lee SH, Goddard ME, Visscher PM. GCTA: A Tool for Genome-wide Complex Trait Analysis. Am J Hum Genet. 2011 Jan 7;88(1):76–82. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.ajhg.2010.11.011&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=21167468&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2019%2F12%2F09%2F19013763.atom) 48. 48.Yang J, Zaitlen NA, Goddard ME, Visscher PM, Price AL. Advantages and pitfalls in the application of mixed-model association methods. Nat Genet. 2014 Feb;46(2):100–6. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/ng.2876&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=24473328&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2019%2F12%2F09%2F19013763.atom) 49. 49.Jia X, Han B, Onengut-Gumuscu S, Chen W-M, Concannon PJ, Rich SS, et al. Imputing Amino Acid Polymorphisms in Human Leukocyte Antigens. PLOS ONE. 2013 Jun 6;8(6):e64683. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1371/journal.pone.0064683&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=23762245&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2019%2F12%2F09%2F19013763.atom) 50. 50.Fellay J, Shianna KV, Ge D, Colombo S, Ledergerber B, Weale M, et al. A Whole-Genome Association Study of Major Determinants for Host Control of HIV-1. Science. 2007 Aug 17;317(5840):944–7. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6Mzoic2NpIjtzOjU6InJlc2lkIjtzOjEyOiIzMTcvNTg0MC85NDQiO3M6NDoiYXRvbSI7czozOToiL21lZHJ4aXYvZWFybHkvMjAxOS8xMi8wOS8xOTAxMzc2My5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 51. 51.McLaren PJ, Coulonges C, Bartha I, Lenz TL, Deutsch AJ, Bashirova A, et al. Polymorphisms of large effect explain the majority of the host genetic contribution to variation of HIV-1 virus load. Proc Natl Acad Sci. 2015 Nov 24;112(47):14658–63. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NDoicG5hcyI7czo1OiJyZXNpZCI7czoxMjoiMTEyLzQ3LzE0NjU4IjtzOjQ6ImF0b20iO3M6Mzk6Ii9tZWRyeGl2L2Vhcmx5LzIwMTkvMTIvMDkvMTkwMTM3NjMuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 52. 52.Li H, Durbin R. Fast and accurate short read alignment with Burrows-Wheeler transform. Bioinforma Oxf Engl. 2009 Jul 15;25(14):1754–60. 53. 53.McKenna A, Hanna M, Banks E, Sivachenko A, Cibulskis K, Kernytsky A, et al. The Genome Analysis Toolkit: A MapReduce framework for analyzing next-generation DNA sequencing data. Genome Res. 2010 Sep 1;20(9):1297–303. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NjoiZ2Vub21lIjtzOjU6InJlc2lkIjtzOjk6IjIwLzkvMTI5NyI7czo0OiJhdG9tIjtzOjM5OiIvbWVkcnhpdi9lYXJseS8yMDE5LzEyLzA5LzE5MDEzNzYzLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 54. 54.Cingolani P, Platts A, Wang LL, Coon M, Nguyen T, Wang L, et al. A program for annotating and predicting the effects of single nucleotide polymorphisms, SnpEff: SNPs in the genome of Drosophila melanogaster strain w1118; iso-2; iso-3. Fly (Austin). 2012 Jun;6(2):80–92. 55. 55.Lee S, Emond MJ, Bamshad MJ, Barnes KC, Rieder MJ, Nickerson DA, et al. Optimal unified approach for rare-variant association testing with application to small-sample case-control whole-exome sequencing studies. Am J Hum Genet. 2012 Aug 10;91(2):224–37. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.ajhg.2012.06.007&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=22863193&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2019%2F12%2F09%2F19013763.atom) 56. 56.Dilthey AT, Gourraud P-A, Mentzer AJ, Cereb N, Iqbal Z, McVean G. High-Accuracy HLA Type Inference from Whole-Genome Sequencing Data Using Population Reference Graphs. PLOS Comput Biol. 2016 Oct 28;12(10):e1005151. 57. 57.Fan Y, Song Y-Q. PyHLA: tests for the association between HLA alleles and diseases. BMC Bioinformatics. 2017 Feb 6;18(1):90. 58. 58.Packer JS, Maxwell EK, O’Dushlaine C, Lopez AE, Dewey FE, Chernomorsky R, et al. CLAMMS: a scalable algorithm for calling common and rare copy number variants from exome sequencing data. Bioinformatics. 2016 Jan 1;32(1):133–5. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/bioinformatics/btv547&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=26382196&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2019%2F12%2F09%2F19013763.atom) 59. 59.Bhattacharya S, Dunn P, Thomas CG, Smith B, Schaefer H, Chen J, et al. ImmPort, toward repurposing of open access immunological assay data for translational and clinical research. Sci Data. 2018 27;5:180015. 60. 60.Purcell S, Cherny SS, Sham PC. Genetic Power Calculator: design of linkage and association genetic mapping studies of complex traits. Bioinformatics. 2003 Jan 1;19(1):149–50. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/bioinformatics/19.1.149&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=12499305&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2019%2F12%2F09%2F19013763.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000180463900021&link_type=ISI) 61. 61.Gutierrez-Arcelus M, Teslovich N, Mola AR, Polidoro RB, Nathan A, Kim H, et al. Lymphocyte innateness defined by transcriptional states reflects a balance between proliferation and effector functions. Nat Commun. 2019 Feb 8;10(1):1–15. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41467-019-09078-0&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=30602773&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2019%2F12%2F09%2F19013763.atom) 62. 62.Marees AT, de Kluiver H, Stringer S, Vorspan F, Curis E, Marie-Claire C, et al. A tutorial on conducting genome-wide association studies: Quality control and statistical analysis. Int J Methods Psychiatr Res. 2018;27(2):e1608. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1002/mpr.1608&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=29484742&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2019%2F12%2F09%2F19013763.atom) 63. 63.Jones LE, Perelson AS. Transient viremia, plasma viral load, and reservoir replenishment in HIV-infected patients on antiretroviral therapy. J Acquir Immune Defic Syndr 1999. 2007 Aug 15;45(5):483–93. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1097/QAI.0b013e3180654836&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=17496565&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2019%2F12%2F09%2F19013763.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000248621200001&link_type=ISI) 64. 64.Fletcher CV, Staskus K, Wietgrefe SW, Rothenberger M, Reilly C, Chipman JG, et al. Persistent HIV-1 replication is associated with lower antiretroviral drug concentrations in lymphatic tissues. Proc Natl Acad Sci U S A. 2014 Feb 11;111(6):2307–12. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NDoicG5hcyI7czo1OiJyZXNpZCI7czoxMDoiMTExLzYvMjMwNyI7czo0OiJhdG9tIjtzOjM5OiIvbWVkcnhpdi9lYXJseS8yMDE5LzEyLzA5LzE5MDEzNzYzLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 65. 65.Lorenzo-Redondo R, Fryer HR, Bedford T, Kim E-Y, Archer J, Pond SLK, et al. Persistent HIV-1 replication maintains the tissue reservoir during therapy. Nature. 2016 Feb 4;530(7588):51–6. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/nature16933&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=26814962&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2019%2F12%2F09%2F19013763.atom) 66. 66.Podsadecki TJ, Vrijens BC, Tousset EP, Rode RA, Hanna GJ. Decreased adherence to antiretroviral therapy observed prior to transient human immunodeficiency virus type 1 viremia. J Infect Dis. 2007 Dec 15;196(12):1773–8. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1086/523704&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=18190257&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2019%2F12%2F09%2F19013763.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000251922600010&link_type=ISI) 67. 67.Young J, Rickenbach M, Calmy A, Bernasconi E, Staehelin C, Schmid P, et al. Transient detectable viremia and the risk of viral rebound in patients from the Swiss HIV Cohort Study. BMC Infect Dis. 2015 Sep 21;15:382. 68. 68.Simonetti FR, Sobolewski MD, Fyne E, Shao W, Spindler J, Hattori J, et al. Clonally expanded CD4+ T cells can produce infectious HIV-1 in vivo. Proc Natl Acad Sci U S A. 2016 Feb 16;113(7):1883–8. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NDoicG5hcyI7czo1OiJyZXNpZCI7czoxMDoiMTEzLzcvMTg4MyI7czo0OiJhdG9tIjtzOjM5OiIvbWVkcnhpdi9lYXJseS8yMDE5LzEyLzA5LzE5MDEzNzYzLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 69. 69.Wang Z, Gurule EE, Brennan TP, Gerold JM, Kwon KJ, Hosmane NN, et al. Expanded cellular clones carrying replication-competent HIV-1 persist, wax, and wane. Proc Natl Acad Sci U S A. 2018 13;115(11):E2575–84. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NDoicG5hcyI7czo1OiJyZXNpZCI7czoxMjoiMTE1LzExL0UyNTc1IjtzOjQ6ImF0b20iO3M6Mzk6Ii9tZWRyeGl2L2Vhcmx5LzIwMTkvMTIvMDkvMTkwMTM3NjMuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 70. 70.Lee GQ, Orlova-Fink N, Einkauf K, Chowdhury FZ, Sun X, Harrington S, et al. Clonal expansion of genome-intact HIV-1 in functionally polarized Th1 CD4+ T cells. J Clin Invest. 2017 Jun 30;127(7):2689–96. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1172/JCI93289&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=28628034&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2019%2F12%2F09%2F19013763.atom) 71. 71.Nardacci R, Amendola A, Ciccosanti F, Corazzari M, Esposito V, Vlassi C, et al. Autophagy plays an important role in the containment of HIV-1 in nonprogressor-infected patients. Autophagy. 2014 Jul;10(7):1167–78. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.4161/auto.28678&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=24813622&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2019%2F12%2F09%2F19013763.atom) 72. 72.Gaebler C, Lorenzi JCC, Oliveira TY, Nogueira L, Ramos V, Lu C-L, et al. Combination of quadruplex qPCR and next-generation sequencing for qualitative and quantitative analysis of the HIV-1 latent reservoir. J Exp Med. 2019 Oct 7;216(10):2253–64. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiamVtIjtzOjU6InJlc2lkIjtzOjExOiIyMTYvMTAvMjI1MyI7czo0OiJhdG9tIjtzOjM5OiIvbWVkcnhpdi9lYXJseS8yMDE5LzEyLzA5LzE5MDEzNzYzLmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 73. 73.Braun DL, Kouyos R, Oberle C, Grube C, Joos B, Fellay J, et al. A novel Acute Retroviral Syndrome Severity Score predicts the key surrogate markers for HIV-1 disease progression. PloS One. 2014;9(12):e114111. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1371/journal.pone.0114111&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=25490090&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2019%2F12%2F09%2F19013763.atom)