Skip to main content
Journal of Virology logoLink to Journal of Virology
. 2020 Jul 16;94(15):e00415-20. doi: 10.1128/JVI.00415-20

HIV RNA Rebound in Seminal Plasma after Antiretroviral Treatment Interruption

Sara Gianella a,✉,#, Antoine Chaillon a,#, Tae-Wook Chun b, Michael C Sneller b, Caroline Ignacio a, Milenka V Vargas-Meneses a, Gemma Caballero a, Ronald J Ellis a, Colin Kovacs c,d, Erika Benko c, Sanja Huibner d, Rupert Kaul d
Editor: Guido Silvestrie
PMCID: PMC7375368  PMID: 32434884

To cure HIV, we likely need to target the reservoirs in all anatomic compartments. Here, we used sophisticated statistical and phylogenetic methods to analyze blood and semen samples collected from 12 persons with HIV who began antiretroviral therapy (ART) during very early HIV infection and who interrupted their ART as part of a clinical trial. First, we found that HIV RNA rebound in semen occurred significantly later and reached lower levels than in blood. Second, we found that the virus in semen was genetically different in some participants compared to that in blood. Finally, we found increased HIV RNA molecular diversity in semen compared to that in blood in almost all study participants. These data suggest that the HIV RNA populations emerging from the genital compartment after ART interruption might not be the same as those emerging from blood plasma. Future research should evaluate whether the genital compartment might contribute to viral rebound in some people with HIV (PWH) interrupting ART.

KEYWORDS: ART interruption, seminal plasma

ABSTRACT

If strategies currently in development succeed in eradicating HIV reservoirs in peripheral blood and lymphoid tissues, residual sources of virus may remain in anatomic compartments. Paired blood and semen samples were collected from 12 individuals enrolled in a randomized, double-blind, placebo-controlled therapeutic vaccine clinical trial in people with HIV (PWH) who began antiretroviral therapy (ART) during acute or early infection (ClinicalTrials registration no. NCT01859325). After the week 56 visit (postintervention), all participants interrupted ART. At the first available time points after viral rebound, we sequenced HIV-1 env (C2-V3), gag (p24), and pol (reverse transcriptase) regions amplified from cell-free HIV RNA in blood and seminal plasma using the MiSeq Illumina platform. Comprehensive sequence and phylogenetic analyses were performed to evaluate viral population structure, compartmentalization, and viral diversity in blood and seminal plasma. Compared to that in blood, HIV RNA rebound in semen occurred significantly later (median of 66 versus 42 days post-ART interruption, P < 0.01) and reached lower levels (median 164 versus 16,090 copies/ml, P < 0.01). Three of five participants with available sequencing data presented compartmentalized viral rebound between blood and semen in one HIV coding region. Despite early ART initiation, HIV RNA molecular diversity was higher in semen than in blood in all three coding regions for most participants. Higher HIV RNA molecular diversity in the genital tract (compared to that in blood plasma) and evidence of compartmentalization illustrate the distinct evolutionary dynamics between these two compartments after ART interruption. Future research should evaluate whether the genital compartment might contribute to viral rebound in some PWH interrupting ART.

IMPORTANCE To cure HIV, we likely need to target the reservoirs in all anatomic compartments. Here, we used sophisticated statistical and phylogenetic methods to analyze blood and semen samples collected from 12 persons with HIV who began antiretroviral therapy (ART) during very early HIV infection and who interrupted their ART as part of a clinical trial. First, we found that HIV RNA rebound in semen occurred significantly later and reached lower levels than in blood. Second, we found that the virus in semen was genetically different in some participants compared to that in blood. Finally, we found increased HIV RNA molecular diversity in semen compared to that in blood in almost all study participants. These data suggest that the HIV RNA populations emerging from the genital compartment after ART interruption might not be the same as those emerging from blood plasma. Future research should evaluate whether the genital compartment might contribute to viral rebound in some people with HIV (PWH) interrupting ART.

INTRODUCTION

Antiretroviral therapy (ART) can suppress viral replication to undetectable levels in most people with HIV (1). However, ART cannot eradicate latently infected cells (2), and HIV RNA rebound occurs following treatment interruption (3), even when ART is initiated during the earliest phase of HIV infection in an attempt to limit the size of the HIV reservoir and improve immune reconstitution (4, 5). Rebounding virus might originate from a variety of sources, including the DNA compartment in peripheral blood mononuclear cells (PBMC), RNA or DNA compartments in lymphoid tissues, or possibly other anatomical compartments that harbor replication-competent HIV-infected cells or HIV particles (69). In most people with HIV (PWH), the establishment of viral reservoirs in multiple tissues and anatomic compartments occurs within the first few weeks of HIV infection, and this likely includes the genital tract (10, 11). The HIV population during the early stages of infection is typically homogenous (12), but compartmentalization can occur as a consequence of tissue-specific genetic differentiation and restricted viral migration between anatomic sites (1317). Compartmentalized viral evolution is frequently a consequence of discordant selective pressures and gives rise to tissue-adapted variants, which subsequently contribute to disease pathogenesis and viral transmission (18). Phylogenetic methods needed to quantify this restriction of gene flow between compartments are numerous and well developed (13). Our group and others have used similar methods to describe the presence of compartmentalized HIV RNA populations in the cerebrospinal fluid (CSF) supernatant, mostly using samples from ART-naive individuals (1921) and from chronically infected individuals following ART interruption (22).

While direct assessment of viral variants sampled from the genital tissue (e.g., testis and prostate) in living individuals is rarely feasible, HIV RNA collected from the seminal plasma can be an informative surrogate (23).

In this study, we leveraged a unique cohort of 12 participants enrolled in a randomized, double-blind, placebo-controlled clinical trial of HIV-MAG DNA vaccine prime and rVSVN4CT1gag booster vaccine in PWH who began ART during acute or early infection (ClinicalTrials registration no. NCT01859325) (24). Comprehensive sequence and phylogenetic analyses were performed to characterize the HIV RNA population rebounding in semen after interruption of ART compared to that in blood. This is important, since compartmentalized HIV reservoirs within the genital tract may be particularly difficult to target with viral eradication strategies, due to limited drug penetration, compartmentalization, tissue-specific viral adaptation, and the presence of unique cellular targets.

RESULTS

Participants, samples, and clinical laboratory tests.

Participants in the genital substudy (N = 12) were all cisgender men with HIV, with a median age of 42 years (range, 24 to 54 years), who had started therapy within a median of 25 days after the estimated date of infection and had been receiving ART for a median of 3.5 years (interquartile range [IQR], 2 to 13 years). Characteristics of the study participants are summarized in Table 1. Four study participants were in the placebo arm (ID1 to -4), while the other 8 were in the active vaccine arm (ID5 to -12).

TABLE 1.

Participants’ characteristics

Characteristics Value (N = 12)
Age (yrs) (median [range]) 42 (24–54)
Gender: cisgender male (n [%]) 12 (100)
Ethnicity: white (n [%]) 6 (50)
Viral clade B (n [%]) 9 (75)
Time from EDI to ART (days) (median [range]) 25 (3–73)
ART exposure (yrs) (median [range]) 3.5 (2–13)
CD4+ T cell counts/μl (at study entry) (median [range]) 470 (300–790)
CD4+ T cell counts/μl (at the time of ART interruption) (median [range]) 510 (390–1430)

HIV RNA dynamics in blood and semen.

Among the 12 participants included in the genital substudy, the median time from ART interruption to viral rebound (defined as first detectable viral load) in blood plasma was 42 days (range, 12 to 83 days) with a median peak HIV RNA load of 16,090 copies/ml (range, 160 to >50,000 copies/ml). The median time from ART interruption to viral rebound in semen was 66 days (range, 16 to 134 days), with a median peak viral load of 164 copies/ml (range, 70 to 3,500 copies/ml). Two participants (ID3 and ID9) experienced small blips in semen before viral rebound in blood. Interestingly, most study participants did not sustain HIV RNA rebound in semen (see Table 2).

TABLE 2.

Viral loads in blood and semen after interruption of antiretroviral therapy

Participant ID Intervention No. of wks HIV RNA (copies/ml)a
Semen HIV RNA Blood HIV RNA On ART
ID1 Placebo 0 NA <40 Yes
2 <40 <40 No
5 <40 <20 No
6 <40 2,532 No
7 309 15,955 No
10 <40 14,385 No
12 <40 6,212 No
14 <40 5,795 No
ID2 Placebo 0 <40 <40 Yes
2 <40 <40 No
4 <40 <40 No
12 832 103,410 No
ID3 Placebo 0 <40 <40 Yes
2 <40 <40 No
4 88 <40 No
6 <40 393 No
9 194 3,677 No
10 238 91 No
12 <40 68 No
14 <40 <40 No
18 <40 <40 No
ID4 Placebo 0 <40 <40 Yes
2 <40 <40 No
4 <40 2,230 No
6 234 173,502 No
10 125 503 No
12 <40 460 No
ID5 Vaccine 0 <40 <40 Yes
3 <40 <20 No
4 <40 1,527 No
5 794 83,710 No
6 3,475 5 145,797 No
10 <40 264 Yes
14 <40 <40 Yes
ID6 Vaccine 0 <40 <40 Yes
2 <40 144 No
4 <40 5,721 No
9 63 2,598 Yes
10 68 361 Yes
14 <40 <40 Yes
17 <40 <40 Yes
ID7 Vaccine 0 <40 <40 Yes
2 <40 <40 No
4 <40 <40 No
6 <40 <40 No
9 <40 80 No
11 <40 287 No
12 <40 2,391 No
14 <40 21,997 No
18 <40 10,450 Yes
ID8 Vaccine 0 <40 <40 Yes
2 <40 <40 No
4 <40 <40 No
9 <40 78 No
10 <40 112 No
14 <40 158 No
15 92 71 No
18 <40 <40 Yes
21 <40 <40 Yes
ID9 Vaccine 0 <40 <40 Yes
2 134 <40 No
4 <40 1,470 No
5 <40 310 No
10 <40 154 No
11 <40 654 No
14 <40 1,530 No
21 <40 105 No
24 <40 101 No
ID10 Vaccine 0 <40 <40 Yes
3 <40 <40 No
4 <40 <40 No
6 <40 3,283 No
11 <40 2,856 No
13 94 2,358 No
15 <40 2,938 No
17 <40 2,392 No
22 <40 <40 Yes
ID11 Vaccine 0 <40 <40 Yes
7 <40 <40 No
9 <40 548,703 No
11 52 806 No
14 <40 488 No
15 <40 389 No
17 <40 9,713 No
19 120 2,932 No
27 <40 222 Yes
ID12 Vaccine 0 <40 <40 Yes
2 <40 1,620 No
5 <40 16,224 No
a

Boldface font indicates time points with available sequencing data. NA, not available; ART, antiretroviral therapy; No. of wks, number of weeks postinterruption of ART.

HIV RNA population characteristics in blood and semen.

To characterize the cell-free HIV RNA population, we successfully sequenced partial env, gag, and/or pol from seminal and blood plasma in 5 participants (ID3 and ID4 in the placebo group and ID5, ID10, and ID11 in the vaccine group). For two participants, we also obtained sequences from one additional longitudinal time point (ID4 and ID5). In blood, we obtained an average of 156,672 reads (range, 69,870 to 798,639) and 9 haplotypes (range, 4 to 20) per sample. In semen, we obtained an average of 252,565 reads (range, 91,028 to 764,568) and 7 haplotypes (range, 1 to 26). Detailed characteristics of the number of reads/haplotypes for each sample are provided in Table 3.

TABLE 3.

Number of reads and haplotypes for each sample from 5 study participants with available sequencing data

Participant ID Intervention Compartment Region No. of readsa
No. of haplotypesa
TP1 TP2 TP1 TP2
ID3 Placebo Blood ENV 265,903 9
GAG 475,530 4
POL 197,201 28
Semen ENV 112,448 2
GAG NA NA
POL 764,568 1
ID4 Placebo Blood ENV 68,840 84,619 11 4
GAG 263,382 316,046 6 3
POL 92,861 103,091 9 7
Semen ENV 84,847 37,308 6 5
GAG 398,760 245,348 2 5
POL 75,182 43,460 23 26
ID5 Vaccine Blood ENV 146,747 181,407 6 4
GAG 427,147 371,492 3 3
POL NA NA NA NA
Semen ENV 31,103 73,285 8 7
GAG 270,242 362,771 3 4
POL NA NA NA NA
ID10 Vaccine Blood ENV 114,756 6
GAG 410,215 9
POL 141,114 10
Semen ENV NA NA
GAG 105,728 3
POL 566,952 15
ID11 Vaccine Blood ENV 121,687 9
GAG 344,835 6
POL 69,870 6
Semen ENV NA NA
Semen GAG 91,028 4
Semen POL NA NA
a

NA, not available (coverage too low); TP, time point.

Although not statistically significant, HIV RNA in semen had a higher entropy than the HIV RNA population in blood (env, P = 0.25; gag, P = 0.07; pol, P = 0.08). Specifically, the mean and 0.95 confidence intervals (CI95%) of entropy in seminal plasma were 0.0505 ± 0.0253 (env), 0.0335 ± 0.0155 (gag), and 0.0515 ± 0.0215 (pol) compared to 0.0396 ± 0.0304 (env), 0.0231 ± 0.0061 (gag), and 0.0375 ± 0.0078 (pol) in blood (Fig. 1 and Table 4).

FIG 1.

FIG 1

Molecular diversity measures in blood and semen. Viral diversity was assessed by measuring the Shannon entropy index from all cleaned reads mapped to HIV partial gag, pol, and env regions. Comparison between entropy measures across participants was performed using a pairwise Wilcoxon adjusted test using the conservative Bonferroni correction method implemented in R package stats.

TABLE 4.

Characteristics of each sampled compartment (Shannon entropy) from 5 study participants with available sequencing data

Participant ID Intervention Time Point Compartment Region Entropy
Mean (IQR) Maximum Median
ID3 Placebo TP1 Blood ENV 0.0194 (0–0.0050) 0.5623 0
GAG 0.0205 (0–0.0238) 0.1220 0.0207
POL 0.0278 (0–0.0397) 0.4276 0.0189
Semen ENV 0.0882 (0–0.0448) 1.0968 0
GAG NA NA NA
POL 0.0128 (0–0.0194) 0.4362 0
ID4 Placebo TP1 Blood ENV 0.0382 (0–0.0216) 0.7181 0
GAG 0.0198 (0–0.0252) 0.2060 0.0211
POL 0.0469 (0–0.0457) 0.5586 0.0213
Semen ENV 0.0384 (0–0.0298) 0.8426 0
GAG 0.0273 (0–0.0390) 0.1899 0.0215
POL 0.0732 (0–0.0584) 0.7959 0.0251
TP2 Blood ENV 0.0361 (0–0.0247) 0.6365 0
GAG 0.0187 (0–0.0250) 0.0922 0.0213
POL 0.0374 (0–0.0436) 0.4714 0.0200
Semen ENV 0.0568 (0–0.0441) 0.9433 0
GAG 0.0320 (0–0.0449) 0.2293 0.0223
POL 0.0781 (0–0.0532) 1.1047 0.0238
ID5 Vaccine TP1 Blood ENV 0.0347 (0–0.0255) 0.7298 0
GAG 0.0229 (0–0.0383) 0.1246 0.0213
POL NA NA NA
Semen ENV 0.0630 (0–0.0436) 1.0397 0
GAG 0.0278 (0–0.0412) 0.1713 0.0206
POL NA NA NA
TP2 Blood ENV 0.0175 (0–0.0203) 0.7851 0
GAG 0.0205 (0–0.0341) 0.1068 0.0208
POL NA NA NA
Semen ENV 0.0377 (0–0.0256) 0.6365 0
GAG 0.0336 (0–0.0528) 0.2255 0.0235
POL NA NA NA
ID10 Vaccine TP1 Blood ENV 0.0199 (0–0.0234) 0.3368 0
GAG 0.0379 (0–0.0414) 0.5690 0.0210
POL 0.0398 (0–0.0444) 0.4888 0.0205
Semen ENV NA NA NA
POL 0.0191 (0–0.0237) 0.2021 0
GAG 0.0293 (0–0.0196) 0.5462 0
ID11 Vaccine TP1 Blood ENV 0.1113 (0–0.0460) 1.1438 0
GAG 0.0218 (0–0.0375) 0.2331 0.0211
POL 0.0490 (0–0.0553) 0.4313 0.0234
Semen ENV NA NA NA
GAG 0.0217 (0–0.0386) 0.0965 0.0212
POL NA NA NA

The mean diversity was not significantly different between the first and the second sampled time points (when available). When looking at individual participants, all but one participant (ID11) with available sequence data presented higher HIV RNA diversity in semen in at least one coding region (Table 5).

TABLE 5.

Wilcoxon pairwise comparisons of Shannon entropy between compartmentsa

Participant ID Intervention Time point Region Sample 1 Sample 2 Adjusted P value
ID3 Placebo TP1 ENV Semen Blood <0.01
POL Semen Blood <0.01
ID4 Placebo TP1 ENV Semen Blood <0.01
GAG Semen Blood 0.016
POL Semen Blood 0.067
TP2 ENV Semen Blood <0.01
GAG Semen Blood <0.01
POL Semen Blood 0.02
ID5 Vaccine TP1 ENV Semen Blood 0.044
GAG Semen Blood 0.621
TP2 ENV Semen Blood <0.01
GAG Semen Blood <0.01
ID10 Vaccine TP1 GAG Semen Blood
POL Semen Blood <0.01
ID11 Vaccine TP1 GAG Semen Blood 0.632
a

Comparison between entropy measures across compartments was performed using a pairwise Wilcoxon adjusted test using the conservative Bonferroni correction method implemented in R package stats.

Phylogenies and population structure of the rebounding HIV RNA populations.

All sequences clustered by study participant where combined in a single phylogeny, which shows that there was no interparticipant contamination (Fig. 2).

FIG 2.

FIG 2

(A to C) Approximate maximum likelihood phylogeny, including all data, from 5 study participants with available sequencing data. All sequences clustered by study participant, which confirm the lack of interparticipant contamination. Participants ID3 and ID4 were assigned to the placebo group, while ID5, ID10, and ID11 were assigned to the vaccine active group.

Using a conservative combination of tree-based (Simmonds association index and Slatkin-Maddison tests) and distance-based (fixation index [FST] test) analyses and after adjusting for the haplotype frequencies, we found evidence of significant viral compartmentalization in at least two of the three measures for 3 of 5 participants in one coding region (ID4 pol, ID5 gag, and ID10 pol, P < 0.05) (Fig. 3). Phylogenetic trees illustrating different HIV RNA populations in blood and semen are shown in Fig. 4. Tree topologies showed intermingled sequences from blood and seminal plasma, confirming the lack of viral compartmentalization for most participants and regions. Phylogenetic trees for pol D4 and pol D10 confirm the presence of a monophyletic clade originating from semen, but a definitive evaluation is limited due to the short length of the sequences and the overall limited diversity in our data set.

FIG 3.

FIG 3

Genetic compartmentalization results for each participant and each coding region. Compartmentalization was assessed using one distance-based and two tree-based approaches: (i) the fixation index FST test (55), (ii) Simmonds association index (AI) (52), and (iii) the Slatkin-Maddison (SM) test (54). Blue denotes compartmentalization. “Consensus” denotes the final result: light blue indicates significant results in at least 2 of 3 tests, dark blue indicates significant results in all tests. *, compartmentalization was declared only if the results of at least 2/3 tests using both reads and haplotypes agreed. (Left) Results when comparing all reads from blood and semen. (Right) Results with reads collapsed to haplotypes. See Materials and Methods for details. Of note, no compartmentalization analysis was performed for ID3 in the pol region because only one haplotype was obtained.

FIG 4.

FIG 4

Phylogenetic trees illustrating different HIV RNA populations in blood and semen. Topologies showed intermingled sequences from blood and seminal plasma, confirming the lack of viral compartmentalization for most participants and regions. Phylogenetic trees for pol D4 and pol D10 confirm the presence of monophyletic clade originating from semen, but a definitive evaluation is limited due to the short length of the sequences and the overall limited diversity in our data set. Participants ID3 and ID4 were assigned to the placebo group, while ID5, ID10, and ID11 were assigned to the vaccine active group.

DISCUSSION

A successful HIV eradication strategy must target all known mechanisms of viral persistence (25). One such mechanism is the establishment of genetically distinct proviral populations in different tissues and anatomical compartments (14), including the genital tract and the central nervous system. In this study, we prospectively collected serial semen samples during structured ART interruption from a unique cohort of 12 PWH enrolled in a randomized, double-blind, placebo-controlled clinical trial of a therapeutic HIV vaccine (24). All study participants began ART during acute or early HIV infection and with sustained viral suppression thereafter (24). The results of the parent trial were previously reported (24): compared to placebo, the vaccination had no significant effect on the kinetics or magnitude of viral rebound after interruption of ART and no impact on the size of the HIV reservoir in the CD4+ T cell compartment. Notably, almost one-third of participants in the placebo arm exhibited sustained suppression of viremia (<400 copies/ml) after treatment interruption, which was consistent with that recently reported in a retrospective analysis of transient viral control in a subgroup of participants in the Short Pulse AntiRetroviral Therapy at Acute Seroconversion (SPARTAC) trial (26). Consistently, the magnitude of HIV RNA rebound in seminal plasma was small, and none of the participants presented a sustained rebound. Overall, the median time from ART interruption to viral rebound in semen was significantly longer than in blood (66 versus 42 days, respectively), although two participants experienced small blips in semen before viral rebound in blood (see Table 2).

Furthermore, in our study of PWH who started ART during the earliest phase of infection (median of 24 days after the estimated date of infection), we found that 3 of 5 participants with available sequencing data presented a compartmentalized HIV RNA rebound within the genital compartment after ART interruption, even when sampled a few days/week after viral rebound (Fig. 3). This pattern implies that at least some of the rebounding virus within the seminal plasma might originate from a genital source and might represent a barrier to sterilizing cure. This is in line with a recent publication from Ganor et al. (9), which demonstrated using penile tissues from PWH under suppressive combination ART, that urethral macrophages contain integrated HIV-1 DNA, RNA, proteins, and intact virions in virus-containing compartment-like structures, whereas viral components remain undetectable in urethral T cells. Urethral HIV-1 reservoirs might be established during early sexual transmission, resulting in rapid infection of resident macrophages, which subsequently become latent and form inducible reservoirs that can be reactivated with lipopolysaccharide (LPS), as recently reported (27).

Importantly, the phylogenies for different viral genes sampled from the same individuals did not show identical dynamics. This observation could result from an independent rebound in the genital tract, for which the archived viruses in blood and semen had homogenized env but different rt genes, e.g., due to a past recombination event. Previous studies have suggested complex intrahost recombination dynamics (28, 29) and demonstrated how selection can affect HIV sequences in different compartments (30, 31), indicating that such eventualities may be common. These findings are comparable to a previous report of rebound in CSF, where unique rebounding species were found (22).

A recent French study of a therapeutic HIV vaccine found a robust early rebound in semen following ART interruption (32), with no evidence of genetic compartmentalization in 10 participants focusing on partial C2/V3 env (454 Roche sequencing platform). While the env gene has the greatest amount of molecular diversity and evolution of all coding regions (3336), here we sequenced 3 partial coding regions (env, gag, and pol) and found evidence of significant viral compartmentalization in pol (n = 2) and gag (n = 1). Therefore, focusing on one partial coding region (env) may have incorrectly inferred that some viral populations were not compartmentalized, when they may be compartmentalized when exploring other genome regions. We cannot also exclude that these differences are due to differences in the study population (chronic versus acutely treated individuals), rebound dynamics, the type of vaccine administered, and/or the sequencing technology (454 versus Illumina MiSeq). Sequencing the near-full-length proviral genome (8, 3739) would increase the sensitivity of these analyses, but these approaches require a high template input, which may limit the ability to explore reservoirs such as the genital tract.

Interestingly, we found higher genetic diversity in semen (as measured by Shannon entropy) in all coding regions compared to that in blood. It is possible that a considerable amount of divergence happened before viral suppression, but this is unlikely in our cohort of people who started ART very early during infection. As an alternative, there might be some degree of viral evolution in the genital tract during ART which requires further confirmation.

This study has several limitations. First, the study only included 12 individuals, and from these, we were only able to obtain semen sequencing data for 5, thereby limiting the power of subsequent statistical analysis. However, despite this small sample size, our study delivers a detailed characterization of semen virus in this unique prospective cohort with intensive clinical, laboratory, and informatics analyses. Second, all our participants started ART early in infection, allowing little baseline genetic diversity to compare rebounding virus populations. Furthermore, the sampling times for blood and semen were days to weeks after rebound, meaning virus replication could account for several nucleotides of difference between the initial rebounding virus and the sampled virus. On the same line, the short length of our amplicons also limited the total diversity among each rebounding virus population within a participant. Thus, theoretically, sequencing error and virus evolution or random change from rebound to sampling could account for all the diversity seen within each gene sequence comparison.

Also, low template input into the sequencing reaction and sequencing errors could cause biases or skewing in the sampling of the original viral population and negatively impact our ability to perform accurate analyses on these samples.

Template input was particularly low in some (but not all) seminal plasma samples, which likely explains the high rate of sequencing failures and negatively impacts our capacity to find unique clades within the seminal compartment. Other possible causes of sequencing failures might be primer mismatches and possibly PCR inhibition in semen.

Despite these limitations, our study suggests that the HIV RNA population sometimes rebounds independently within the genital tract and that the HIV DNA reservoirs in anatomic compartments might present additional obstacles to eradication and need to be actively targeted to achieve a complete cure. Our data suggest that failing to collect appropriate specimens (such as semen or cerebrospinal fluid) during structured ART interruption in the setting of cure trials might overlook some important events emanating from the anatomic compartments, which could be contributing to overall rebound.

MATERIALS AND METHODS

Ethics statement.

The parent therapeutic vaccine clinical protocol (ClinicalTrials.gov identifier NCT01859325) was approved by the Institutional Review Board of the National Institutes of Allergy and Infectious Diseases (NIAID) and previously published (24). Semen samples for the genital tract substudy were collected under a separate protocol approved by the Human Research Protections Program at the University of Toronto. All adult subjects provided written informed consent.

Participants, samples, and clinical laboratory tests.

Between September 2013 and February 2015, a total of 31 PWH enrolled in a randomized, double-blind, placebo-controlled clinical trial of HIV-MAG DNA vaccine prime and rVSVN4CT1gag booster vaccine (NCT01859325) (24). After the week 56 visit postintervention, all participants underwent treatment interruption to determine if vaccine administration resulted in an improved immune control of viral replication. Study participants were subsequently followed through week 96 postintervention for safety and efficacy parameters.

A subset of 12 participants agreed to enroll in a genital tract substudy. Semen was collected at baseline (before ART interruption) and approximatively every 2 weeks for a median of 18 weeks (range, 12 to 27 weeks) post-ART interruption. Semen was processed as previously described (4043) by masturbation into a sterile container containing 10 ml of RPMI with penicillin-streptomycin following 48 h of abstinence, and was processed within 2 h. Semen was centrifuged at 850 × g for 10 min, and semen plasma HIV RNA viral load assayed using the Abbott RealTime HIV-1 assay (Abbott Molecular Diagnostics; limit of detection, 40 copies/ml). Due to occasional spillage of RPMI during semen collection and/or sample transport, correction for semen dilution assumed a semen volume of 2 ml, as previously validated.

All study participants were asked to practice condom-protected sex for the duration of the study, and sexually transmitted infection (STI) screening (syphilis serology and urine molecular diagnostics for Neisseria gonorrhoeae and Chlamydia trachomatis) was performed at study screening, at baseline, and at each semen donation visit. No STIs were diagnosed in any participant for the duration of the study.

HIV RNA extraction and next-generation sequencing from blood plasma and seminal supernatant.

Nucleic acid was concentrated from 1 ml of blood plasma by high-speed centrifugation. Subsequently, RNA was isolated according to the manufacturer’s instructions (QIAamp viral RNA minikit; Qiagen, Hilden, Germany), and cDNA was produced (RETROscript kit; Applied Biosystems/Ambion, Austin, TX). For semen, viral population was concentrated from 2 to 6 ml of supernatant in order to maximize template input in each MiSeq reaction mixture (depending on viral load and sample availability). Three coding regions—gag p24 (HXB2 coordinates 1366 to 1619), pol RT (2708 to 3242), and env C2-V3 (6928 to 7344)—were amplified by PCR with region-specific primers, as previously described (44).

Sequence processing and bioinformatics analysis.

(i) Sequencing. The Illumina MiSeq instrument and MiSeq reagent kit V3 600-cycle paired-end sequencing kits (MS-102-2003/MS-102-3003) were used to sequence the DNA libraries. The median number of reads per amplicon was 320,452 (IQR, 198,053 to 1,494,661).

(ii) Read mapping and filtering. The reads were analyzed using a custom pipeline adapted from Zanini et al. (45). Briefly, (i) reads were first mapped onto the HIV-1 reference HxB2, (ii) mapped reads were classified into partial gag, pol, and env regions (ambiguous reads were discarded) and trimmed for PHRED quality above or equal to 30, (iii) a consensus sequence was computed for each 3 regions in each sample from a subset of the reads, using a chain of overlapping local multiple sequence alignments, (iv) reads were remapped against their own consensus, (v) reads were trimmed for mapping errors at the edges (small indels), (vi) filtered reads were mapped a third time against a patient-specific consensus sequence from the initial time point (day 0), and (vii) reads were refiltered and checked again for cross-contamination.

(iii) Recombination screening. We screened all sets of representative reads for evidence of recombination using GARD (46).

(iv) Diversity. Viral diversity was assessed by measuring the Shannon entropy index from all cleaned reads mapped to HIV partial gag, pol, and env regions (47). Comparison between entropy measures across participants was performed using a pairwise Wilcoxon adjusted test using the conservative Bonferroni correction method implemented in R package stats (48, 49).

(v) Phylogenetic analysis. HIV haplotypes above a minimal frequency threshold of 0.01 were extracted from reads covering the gag, pol, and env regions and were used to construct maximum likelihood (ML) phylogenies using IQtree (50).

(vi) Population structure. Compartmentalization was assessed using two tree-based and one distance-based approach:

  • (1)

    To quantify the overall spatial structure of the phylogenies, we first measured the phylogenetic association in the location trait data (i.e., semen or blood) (51, 52). Using the posterior set of trees, we calculated the Simmonds association index (AI) (52) for each discrete “location trait” (i.e., compartment) using BaTS v1.0 (53). To provide statistical evidence for genetic segregation of the HIV populations, the reported P value was inferred as the proportion of trees from the null distribution equal to, or more extreme than, the median posterior estimate of the statistic from the posterior set of trees.

  • (2)

    The tree-based Slatkin-Maddison (SM) test (54): trees were considered compartmentalized with the SM test if 10,000 permutations of the SM test yielded a P value of <0.05.

  • (3)

    The fixation index (55) defined as FST=1πIπD, where πI is the estimate of mean pairwise intracompartment genetic distance (TN93) (56) and πD is its intercompartment counterpart (22).

These methods were repeated after collapsing identical reads into haplotypes to consider the haplotype relative abundance. HIV populations from each of the 3 HIV coding regions were considered compartmentalized if at least 2 of 3 methods showed evidence of compartmentalization consistently before and after collapsing reads into haplotypes.

Data availability.

All read files are available in the NCBI Sequence Read Archive under accession numbers SAMN14848556 to SAMN14848588.

ACKNOWLEDGMENTS

This work was supported by the Department of Veterans Affairs and grants from the National Institutes of Health: AI131385, AI118422, AI036214, MH062512, MH107345, AI134295, and HD094646. The funders had no role in study design, data collection and analysis, the decision to publish, or preparation of the manuscript.

We have no conflicts of interest.

REFERENCES

  • 1.Palella FJ Jr, Baker RK, Moorman AC, Chmiel JS, Wood KC, Brooks JT, Holmberg SD, HIV Outpatient Study Investigators. 2006. Mortality in the highly active antiretroviral therapy era: changing causes of death and disease in the HIV outpatient study. J Acquir Immune Defic Syndr 43:27–34. doi: 10.1097/01.qai.0000233310.90484.16. [DOI] [PubMed] [Google Scholar]
  • 2.Sengupta S, Siliciano RF. 2018. Targeting the latent reservoir for HIV-1. Immunity 48:872–895. doi: 10.1016/j.immuni.2018.04.030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Hoen B, Fournier I, Lacabaratz C, Burgard M, Charreau I, Chaix ML, Molina JM, Livrozet JM, Venet A, Raffi F, Aboulker JP, Rouzioux C, PRIMSTOP Study Group. 2005. Structured treatment interruptions in primary HIV-1 infection: the ANRS 100 PRIMSTOP trial. J Acquir Immune Defic Syndr 40:307–316. doi: 10.1097/01.qai.0000182628.66713.31. [DOI] [PubMed] [Google Scholar]
  • 4.Hocqueloux L, Prazuck T, Avettand-Fenoel V, Lafeuillade A, Cardon B, Viard JP, Rouzioux C. 2010. Long-term immunovirologic control following antiretroviral therapy interruption in patients treated at the time of primary HIV-1 infection. AIDS 24:1598–1601. doi: 10.1097/qad.0b013e32833b61ba. [DOI] [PubMed] [Google Scholar]
  • 5.Saez-Cirion A, Bacchus C, Hocqueloux L, Avettand-Fenoel V, Girault I, Lecuroux C, Potard V, Versmisse P, Melard A, Prazuck T, Descours B, Guergnon J, Viard JP, Boufassa F, Lambotte O, Goujard C, Meyer L, Costagliola D, Venet A, Pancino G, Autran B, Rouzioux C, ANRS VISCONTI Study Group. 2013. Post-treatment HIV-1 controllers with a long-term virological remission after the interruption of early initiated antiretroviral therapy ANRS VISCONTI study. PLoS Pathog 9:e1003211. doi: 10.1371/journal.ppat.1003211. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Margolis DM. 2014. How Might We Cure HIV? Curr Infect Dis Rep 16:392. doi: 10.1007/s11908-014-0392-2. [DOI] [PubMed] [Google Scholar]
  • 7.Gray LR, Roche M, Flynn JK, Wesselingh SL, Gorry PR, Churchill MJ. 2014. Is the central nervous system a reservoir of HIV-1? Curr Opin HIV AIDS 9:552–558. doi: 10.1097/COH.0000000000000108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Chaillon A, Gianella S, Dellicour S, Rawlings SA, Schlub TE, De Oliveira MF, Ignacio C, Porrachia M, Vrancken B, Smith DM. 2020. HIV persists throughout deep tissues with repopulation from multiple anatomical sources. J Clin Invest 130:1699–1712. doi: 10.1172/JCI134815. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Ganor Y, Real F, Sennepin A, Dutertre C-A, Prevedel L, Xu L, Tudor D, Charmeteau B, Couedel-Courteille A, Marion S, Zenak A-R, Jourdain J-P, Zhou Z, Schmitt A, Capron C, Eugenin EA, Cheynier R, Revol M, Cristofari S, Hosmalin A, Bomsel M. 2019. HIV-1 reservoirs in urethral macrophages of patients under suppressive antiretroviral therapy. Nat Microbiol 4:633–644. doi: 10.1038/s41564-018-0335-z. [DOI] [PubMed] [Google Scholar]
  • 10.Chun TW, Engel D, Berrey MM, Shea T, Corey L, Fauci AS. 1998. Early establishment of a pool of latently infected, resting CD4+ T cells during primary HIV-1 infection. Proc Natl Acad Sci U S A 95:8869–8873. doi: 10.1073/pnas.95.15.8869. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Murray AJ, Kwon KJ, Farber DL, Siliciano RF. 2016. The latent reservoir for HIV-1: how immunologic memory and clonal expansion contribute to HIV-1 persistence. J Immunol 197:407–417. doi: 10.4049/jimmunol.1600343. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Keele BF, Giorgi EE, Salazar-Gonzalez JF, Decker JM, Pham KT, Salazar MG, Sun C, Grayson T, Wang S, Li H, Wei X, Jiang C, Kirchherr JL, Gao F, Anderson JA, Ping LH, Swanstrom R, Tomaras GD, Blattner WA, Goepfert PA, Kilby JM, Saag MS, Delwart EL, Busch MP, Cohen MS, Montefiori DC, Haynes BF, Gaschen B, Athreya GS, Lee HY, Wood N, Seoighe C, Perelson AS, Bhattacharya T, Korber BT, Hahn BH, Shaw GM. 2008. 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. doi: 10.1073/pnas.0802203105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Zarate S, Pond SL, Shapshak P, Frost SD. 2007. Comparative study of methods for detecting sequence compartmentalization in human immunodeficiency virus type 1. J Virol 81:6643–6651. doi: 10.1128/JVI.02268-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Svicher V, Ceccherini-Silberstein F, Antinori A, Aquaro S, Perno CF. 2014. Understanding HIV compartments and reservoirs. Curr HIV/AIDS Rep 11:186–194. doi: 10.1007/s11904-014-0207-y. [DOI] [PubMed] [Google Scholar]
  • 15.Blackard JT. 2012. HIV compartmentalization: a review on a clinically important phenomenon. Curr HIV Res 10:133–142. doi: 10.2174/157016212799937245. [DOI] [PubMed] [Google Scholar]
  • 16.Kariuki SM, Selhorst P, Anthony C, Matten D, Abrahams M-R, Martin DP, Ariën KK, Rebe K, Williamson C, Dorfman JR. 8 April 2020. Compartmentalization and clonal amplification of HIV-1 in the male genital tract characterized using next-generation sequencing. J Virol doi: 10.1128/JVI.00229-20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Oliveira MF, Chaillon A, Nakazawa M, Vargas M, Letendre SL, Strain MC, Ellis RJ, Morris S, Little SJ, Smith DM, Gianella S. 2017. Early antiretroviral therapy is associated with lower HIV DNA molecular diversity and lower inflammation in cerebrospinal fluid but does not prevent the establishment of compartmentalized HIV DNA populations. PLoS Pathog 13:e1006112. doi: 10.1371/journal.ppat.1006112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Pillai SK, Good B, Pond SK, Wong JK, Strain MC, Richman DD, Smith DM. 2005. Semen-specific genetic characteristics of human immunodeficiency virus type 1 env. J Virol 79:1734–1742. doi: 10.1128/JVI.79.3.1734-1742.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Harrington PR, Schnell G, Letendre SL, Ritola K, Robertson K, Hall C, Burch CL, Jabara CB, Moore DT, Ellis RJ, Price RW, Swanstrom R. 2009. Cross-sectional characterization of HIV-1 env compartmentalization in cerebrospinal fluid over the full disease course. AIDS 23:907–915. doi: 10.1097/QAD.0b013e3283299129. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Smith DM, Zarate S, Shao H, Pillai SK, Letendre SL, Wong JK, Richman DD, Frost SD, Ellis RJ, HNRC Group. 2009. Pleocytosis is associated with disruption of HIV compartmentalization between blood and cerebral spinal fluid viral populations. Virology 385:204–208. doi: 10.1016/j.virol.2008.11.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Choi JY, Hightower GK, Wong JK, Heaton R, Woods S, Grant I, Marcotte TD, Ellis RJ, Letendre SL, Collier AC, Marra CM, Clifford DB, Gelman BB, McArthur JC, Morgello S, Simpson DM, McCutchan JA, Richman DD, Smith DM, Charter Group. 2012. Genetic features of cerebrospinal fluid-derived subtype B HIV-1 Tat. J Neurovirol 18:81–90. doi: 10.1007/s13365-011-0059-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Gianella S, Kosakovsky Pond SL, Oliveira MF, Scheffler K, Strain MC, De la Torre A, Letendre S, Smith DM, Ellis RJ. 2016. Compartmentalized HIV rebound in the central nervous system after interruption of antiretroviral therapy. Virus Evol 2:vew020. doi: 10.1093/ve/vew020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Chaillon A, Smith DM, Vanpouille C, Lisco A, Jordan P, Caballero G, Vargas M, Gianella S, Mehta SR. 2017. HIV trafficking between blood and semen during early untreated HIV infection. J Acquir Immune Defic Syndr 74:95–102. doi: 10.1097/QAI.0000000000001156. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Sneller MC, Justement JS, Gittens KR, Petrone ME, Clarridge KE, Proschan MA, Kwan R, Shi V, Blazkova J, Refsland EW, Morris DE, Cohen KW, McElrath MJ, Xu R, Egan MA, Eldridge JH, Benko E, Kovacs C, Moir S, Chun T-W, Fauci AS. 2017. A randomized controlled safety/efficacy trial of therapeutic vaccination in HIV-infected individuals who initiated antiretroviral therapy early in infection. Sci Transl Med 9:eaan8848. doi: 10.1126/scitranslmed.aan8848. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Richman DD, Margolis DM, Delaney M, Greene WC, Hazuda D, Pomerantz RJ. 2009. The challenge of finding a cure for HIV infection. Science 323:1304–1307. doi: 10.1126/science.1165706. [DOI] [PubMed] [Google Scholar]
  • 26.Martin GE, Gossez M, Williams JP, Stöhr W, Meyerowitz J, Leitman EM, Goulder P, Porter K, Fidler S, Frater J. 2017. Post-treatment control or treated controllers? Viral remission in treated and untreated primary HIV infection. AIDS 31:477–484. doi: 10.1097/QAD.0000000000001382. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Ganor Y, Zhou Z, Bodo J, Tudor D, Leibowitch J, Mathez D, Schmitt A, Vacher-Lavenu M, Revol M, Bomsel M. 2013. The adult penile urethra is a novel entry site for HIV-1 that preferentially targets resident urethral macrophages. Mucosal Immunol 6:776–786. doi: 10.1038/mi.2012.116. [DOI] [PubMed] [Google Scholar]
  • 28.Brown RJ, Peters PJ, Caron C, Gonzalez-Perez MP, Stones L, Ankghuambom C, Pondei K, McClure CP, Alemnji G, Taylor S, Sharp PM, Clapham PR, Ball JK. 2011. Intercompartmental recombination of HIV-1 contributes to env intrahost diversity and modulates viral tropism and sensitivity to entry inhibitors. J Virol 85:6024–6037. doi: 10.1128/JVI.00131-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Miller RL, Ponte R, Jones BR, Kinloch NN, Omondi FH, Jenabian M-A, Dupuy FP, Fromentin R, Brassard P, Mehraj V, Chomont N, Poon AFY, Joy JB, Brumme ZL, Routy J-P. 2019. HIV diversity and genetic compartmentalization in blood and testes during suppressive antiretroviral therapy. J Virol 93:e00755-19. doi: 10.1128/JVI.00755-19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Hightower GK, Letendre SL, Cherner M, Gibson SA, Ellis RJ, Wolfson TJ, Gamst AC, Ignacio CC, Heaton RK, Grant I, Richman DD, Smith DM, HNRC Group. 2009. Select resistance-associated mutations in blood are associated with lower CSF viral loads and better neuropsychological performance. Virology 394:243–248. doi: 10.1016/j.virol.2009.08.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Neher RA, Leitner T. 2010. Recombination rate and selection strength in HIV intra-patient evolution. PLoS Comput Biol 6:e1000660. doi: 10.1371/journal.pcbi.1000660. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Palich R, Ghosn J, Chaillon A, Boilet V, Nere M-L, Chaix M-L, Delobel P, Molina J-M, Lucht F, Bouchaud O. 2019. Viral rebound in semen after antiretroviral treatment interruption in an HIV therapeutic vaccine double-blind trial. AIDS 33:279–284. doi: 10.1097/QAD.0000000000002058. [DOI] [PubMed] [Google Scholar]
  • 33.Travers SAA, O'Connell MJ, McCormack GP, McInerney JO. 2005. Evidence for heterogeneous selective pressures in the evolution of the env gene in different human immunodeficiency virus type 1 subtypes. J Virol 79:1836–1841. doi: 10.1128/JVI.79.3.1836-1841.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Seibert SA, Howell CY, Hughes MK, Hughes AL. 1995. Natural selection on the gag, pol, and env genes of human immunodeficiency virus 1 (HIV-1). Mol Biol Evol 12:803–813. doi: 10.1093/oxfordjournals.molbev.a040257. [DOI] [PubMed] [Google Scholar]
  • 35.Leitner T, Kumar S, Albert J. 1997. Tempo and mode of nucleotide substitutions in gag and env gene fragments in human immunodeficiency virus type 1 populations with a known transmission history. J Virol 71:4761–4770. doi: 10.1128/JVI.71.6.4761-4770.1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Vrancken B, Alavian SM, Aminy A, Amini-Bavil-Olyaee S, Pourkarim MR. 2018. Why comprehensive datasets matter when inferring epidemic links or subgenotyping. Infect Genet Evol 65:350–351. doi: 10.1016/j.meegid.2018.08.012. [DOI] [PubMed] [Google Scholar]
  • 37.Hiener B, Horsburgh BA, Eden JS, Barton K, Schlub TE, Lee E, von Stockenstrom S, Odevall L, Milush JM, Liegler T, Sinclair E, Hoh R, Boritz EA, Douek D, Fromentin R, Chomont N, Deeks SG, Hecht FM, Palmer S. 2017. Identification of genetically intact HIV-1 proviruses in specific CD4+ T cells from effectively treated participants. Cell Rep 21:813–822. doi: 10.1016/j.celrep.2017.09.081. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Bruner KM, Murray AJ, Pollack RA, Soliman MG, Laskey SB, Capoferri AA, Lai J, Strain MC, Lada SM, Hoh R, Ho Y-C, Richman DD, Deeks SG, Siliciano JD, Siliciano RF. 2016. Defective proviruses rapidly accumulate during acute HIV-1 infection. Nat Med 22:1043–1049. doi: 10.1038/nm.4156. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Lee GQ, Orlova-Fink N, Einkauf K, Chowdhury FZ, Sun X, Harrington S, Kuo H-H, Hua S, Chen H-R, Ouyang Z, Reddy K, Dong K, Ndung'u T, Walker BD, Rosenberg ES, Yu XG, Lichterfeld M. 2017. Clonal expansion of genome-intact HIV-1 in functionally polarized Th1 CD4+ T cells. J Clin Invest 127:2689–2696. doi: 10.1172/JCI93289. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Kaul R, Pettengell C, Sheth PM, Sunderji S, Biringer A, MacDonald K, Walmsley S, Rebbapragada A. 2008. The genital tract immune milieu: an important determinant of HIV susceptibility and secondary transmission. J Reprod Immunol 77:32–40. doi: 10.1016/j.jri.2007.02.002. [DOI] [PubMed] [Google Scholar]
  • 41.Sheth PM, Danesh A, Sheung A, Rebbapragada A, Shahabi K, Kovacs C, Halpenny R, Tilley D, Mazzulli T, MacDonald K, Kelvin D, Kaul R. 2006. Disproportionately high semen shedding of HIV is associated with compartmentalized cytomegalovirus reactivation. J Infect Dis 193:45–48. doi: 10.1086/498576. [DOI] [PubMed] [Google Scholar]
  • 42.Sheth PM, Kovacs C, Kemal KS, Jones RB, Raboud JM, Pilon R, la Porte C, Ostrowski M, Loutfy M, Burger H, Weiser B, Kaul R, Toronto Mucosal Immunology Group. 2009. Persistent HIV RNA shedding in semen despite effective antiretroviral therapy. AIDS 23:2050–2054. doi: 10.1097/QAD.0b013e3283303e04. [DOI] [PubMed] [Google Scholar]
  • 43.Sheth PM, Shahabi K, Rebbapragada A, Kovacs C, Dimayuga R, Chackalakkal S, MacDonald K, Mazzulli T, Kaul R. 2004. HIV viral shedding in semen: lack of correlation with systemic virus-specific CD8 responses. AIDS 18:2202–2205. doi: 10.1097/00002030-200411050-00015. [DOI] [PubMed] [Google Scholar]
  • 44.Gianella S, Delport W, Pacold ME, Young JA, Choi JY, Little SJ, Richman DD, Pond SLK, Smith DM. 2011. Detection of minority resistance during early HIV-1 infection: natural variation and spurious detection rather than transmission and evolution of multiple viral variant. J Virol 85:8359–8367. doi: 10.1128/JVI.02582-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Zanini F, Brodin J, Thebo L, Lanz C, Bratt G, Albert J, Neher RA. 2015. Population genomics of intrapatient HIV-1 evolution. Elife 4:e11282. doi: 10.7554/eLife.11282. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Pond SL, Frost SD. 2005. A genetic algorithm approach to detecting lineage-specific variation in selection pressure. Mol Biol Evol 22:478–485. doi: 10.1093/molbev/msi031. [DOI] [PubMed] [Google Scholar]
  • 47.Shannon CE. 1997. The mathematical theory of communication. 1963. MD Comput 14:306–317. [PubMed] [Google Scholar]
  • 48.Hommel G. 1988. A stagewise rejective multiple test procedure based on a modified Bonferroni test. Biometrika 75:383–386. doi: 10.1093/biomet/75.2.383. [DOI] [Google Scholar]
  • 49.Hochberg Y. 1988. A sharper Bonferroni procedure for multiple tests of significance. Biometrika 75:800–802. doi: 10.1093/biomet/75.4.800. [DOI] [Google Scholar]
  • 50.Schmidt HA, Minh BQ, von Haeseler A, Nguyen L-T. 2015. IQ-TREE: a fast and effective stochastic algorithm for estimating maximum-likelihood phylogenies. Mol Biol Evol 32:268–274. doi: 10.1093/molbev/msu300. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Lemey P, Rambaut A, Drummond AJ, Suchard MA. 2009. Bayesian phylogeography finds its roots. PLoS Comput Biol 5:e1000520. doi: 10.1371/journal.pcbi.1000520. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Wang TH, Donaldson YK, Brettle RP, Bell JE, Simmonds P. 2001. Identification of shared populations of human immunodeficiency virus type 1 infecting microglia and tissue macrophages outside the central nervous system. J Virol 75:11686–11699. doi: 10.1128/JVI.75.23.11686-11699.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Parker J, Rambaut A, Pybus OG. 2008. Correlating viral phenotypes with phylogeny: accounting for phylogenetic uncertainty. Infect Genet Evol 8:239–246. doi: 10.1016/j.meegid.2007.08.001. [DOI] [PubMed] [Google Scholar]
  • 54.Slatkin M, Maddison WP. 1989. A cladistic measure of gene flow inferred from the phylogenies of alleles. Genetics 123:603–613. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Hudson RR. 1992. Gene trees, species trees and the segregation of ancestral alleles. Genetics 131:509–513. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Tamura K, Nei M. 1993. Estimation of the number of nucleotide substitutions in the control region of mitochondrial DNA in humans and chimpanzees. Mol Biol Evol 10:512–526. doi: 10.1093/oxfordjournals.molbev.a040023. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

All read files are available in the NCBI Sequence Read Archive under accession numbers SAMN14848556 to SAMN14848588.


Articles from Journal of Virology are provided here courtesy of American Society for Microbiology (ASM)

RESOURCES