ABSTRACT
Understanding the dynamics of early immune responses to HIV-1 infection, including the evolution of initial neutralizing and antibody-dependent cellular cytotoxicity (ADCC)-mediating antibodies, will inform HIV vaccine design. In this study, we assess the development of autologous neutralizing antibodies (ANAbs) against founder envelopes (Envs) from 18 participants with HIV-1 CRF01_AE acute infection. The timing of ANAb development directly associated with the magnitude of the longitudinal ANAb response. Participants that developed ANAbs within 6 months of infection had significantly higher ANAb responses at 1 year (50% inhibitory concentration [IC50] geometric mean titer [GMT] = 2,010 versus 184; P = 0.001) and 2 years (GMT = 3,479 versus 340; P = 0.015), compared to participants that developed ANAb responses after 6 months. Participants with later development of ANAb tended to develop an earlier, potent heterologous tier 1 (92TH023) neutralizing antibody (NAb) response (P = 0.049). CRF01_AE founder Env V1V2 loop lengths correlated indirectly with the timing (P = 0.002, r = −0.675) and directly with magnitude (P = 0.005, r = 0.635) of ANAb responses; Envs with longer V1V2 loop lengths elicited earlier and more potent ANAb responses. While ANAb responses did not associate with viral load, the viral load set point correlated directly with neutralization of the heterologous 92TH023 strain (P = 0.007, r = 0.638). In contrast, a striking inverse correlation was observed between viral load set point and peak ADCC against heterologous 92TH023 Env strain (P = 0.0005, r = −0.738). These data indicate that specific antibody functions can be differentially related to viral load set point and may affect HIV-1 pathogenesis. Exploiting Env properties, such as V1V2 length, could facilitate development of subtype-specific vaccines that elicit more effective immune responses and improved protection.
IMPORTANCE Development of an effective HIV-1 vaccine will be facilitated by better understanding the dynamics between the founder virus and the early humoral responses. Variations between subtypes may influence the evolution of immune responses and should be considered as we strive to understand these dynamics. In this study, autologous founder envelope neutralization and heterologous functional humoral responses were evaluated after acute infection by HIV-1 CRF01_AE, a subtype that has not been thoroughly characterized. The evolution of these humoral responses was assessed in relation to envelope characteristics, magnitude of elicited immune responses, and viral load. Understanding immune parameters in natural infection will improve our understanding of protective responses and aid in the development of immunogens that elicit protective functional antibodies. Advancing our knowledge of correlates of positive clinical outcomes should lead to the design of more efficacious vaccines.
KEYWORDS: HIV-1, autologous neutralization, CRF01_AE, V1V2, ADCC, viral load set point, neutralization breadth
INTRODUCTION
Immune responses in acute HIV-1 infection may affect disease features and progression. Understanding these events could contribute to improved vaccine design, as well as treatment approaches. Humoral immune responses to HIV-1 have been well characterized for subtype B, which is prominent in Europe, the Americas, and Oceania, and subtype C, which is prominent in Eastern and Southern Africa and India (1). However, understanding of other subtypes is not as well defined. Antibodies capable of neutralizing the founder subtype B and C viruses typically develop within months of infection (2, 3), while heterologous neutralizing antibodies (NAbs) appear after 1 or more years (4, 5). Of individuals who develop heterologous NAbs, only about 20 to 30% develop neutralization breadth (6, 7). While some studies have begun to focus on CRF01_AE infection and the developing immune responses (8, 9), a greater understanding of CRF01_AE founder Env properties and their elicited immune responses is needed to improve our understanding of humoral immunity in contemporary CRF01_AE infections.
CRF01_AE HIV-1, most prevalent in Asia, has Env features that appear to be distinct from other HIV-1 subtypes (1, 10, 11). The CRF01_AE Env is the least glycosylated among the subtypes, with an average of 28 potential N-linked glycosylation sites (12, 13). CRF01_AE Envs also contains an unusual histidine in the CD4-binding pocket at position 375, potentially leading to potentially greater sampling of the activated Env conformation (14). However, the incorporation of histidine at position 375 has not been shown to associate with Env antibody-dependent cellular cytotoxicity (ADCC) susceptibility, and these observations may vary by HIV-1 strain (15). This structural property has been implicated in the development of higher ADCC activity (16–19). Additionally, the neutralization sensitivities of CRF01_AE viruses are distinct from other subtypes (11). Further characterization is needed to understand how these unique properties of CRF01_AE HIV-1 Env affect the humoral immune response in natural infection and vaccination.
In the Thai RV144 vaccine trial, vaccination with CRF01_AE and subtype B antigens (canarypox vector ALVAC HIV [vCP1521] and AIDSVAX B/E gp120) lowered HIV acquisition by 31.2% (20). Analysis of this study showed a higher frequency of V2-specific antibodies in vaccinees (21), with an inverse correlation observed between V1V2-specific plasma IgG and infection risk (18, 22). Considering the potential protective role of V1V2-specific antibodies, a more in-depth evaluation of V1V2 antibody responses to CRF01_AE founder envelopes is warranted. V1V2 is a highly variable structure within Env (2, 23) with multiple N-linked glycans (24). This variable loop can shield neutralization sensitive epitopes within the Env trimeric spike (25–29) in a strain-specific manner (30, 31) and may facilitate immune evasion (29). Natural infection studies in subtype A and C have shown that V1V2-directed antibodies can contribute to autologous neutralization responses (32), with changes in the V1V2 loop resulting in neutralization resistance (32–35). Further understanding of V1V2-specific antibody responses in natural CRF01_AE HIV infection could inform modifications to vaccine antigens that may improve vaccine immunogenicity and elicitation of beneficial V1V2-specific antibodies.
The RV217 acute HIV-1 infection cohort is a prospective natural history study among high-risk participants from Thailand and East Africa (36). Healthy participants at increased risk for HIV acquisition were enrolled and initially monitored twice weekly. Upon detection of HIV-1 RNA from finger-stick blood samples, individuals were monitored frequently for 4 weeks and then at lengthening intervals for over 3 years. This study provided a detailed view of viral dynamics in acute infection. While other acute infection cohorts exist, these study samples are unique in their opportunity to evaluate the timing of acute CRF01_AE HIV-1 antibody development due to the close patient monitoring, defined seroconversion events, and frequency of sample collection after infection. The recently reported increase in CRF01_AE HIV prevalence, as well as the suggested faster clinical progression (37, 38), highlight the need for further investigation and continued vaccine development for this subtype.
In this study, we evaluated the longitudinal autologous and heterologous humoral immune responses to consensus founder Envs from 18 antiretroviral therapy (ART)-naive participants with CRF01_AE-infection. Here, we report specific distinctions among HIV-1 CRF01_AE founder Envs with respect to the magnitude and kinetics of antibodies that neutralize the autologous virus, NAb sensitivity, and ADCC activity and have investigated associations of these immune responses with viral load (VL) set point. Overall, HIV-1 CRF01_AE appears to take longer to induce autologous neutralizing antibodies (ANAbs) against the founder virus than has been previously reported for other subtypes (39). Participants with longer Env V1V2 lengths developed earlier autologous neutralizing antibodies, which reached higher magnitude over 3 years. A striking inverse correlation was seen between 92TH023 ADCC and VL set point, indicating plasma ADCC activity maybe be associated with improved viremic control. In four of these participants studied, the autologous NAb response preceded or coevolved with the autologous ADCC responses. Taken together, these data reveal insights into humoral responses that can inform HIV vaccine development for regions where CRF01_AE HIV-1 is prevalent.
RESULTS
Development of a Thai CRF01_AE founder Env pseudovirus panel.
Participants were selected for this study from the institutional review board (IRB)-approved, RV217 acute infection protocol executed in Thailand. Twelve male and six transgender female CRF01_AE HIV-infected participants were included; demographic data are available in Table S1. Founder env genes were cloned from plasma collected during the first week of diagnosed HIV infection. Of the 18 selected participants, 13 were infected with a single founder env, 4 participants were infected with 2 founder env genes, and 1 participant was infected with 5 founder env genes. env genes were cloned into expression vectors, and 26 founder HIV pseudoviruses (PSVs) were produced. All founder PSVs were determined to utilize the CCR5 coreceptor (data not shown), as typically seen in sexually transmitted HIV (40–42). Neutralization sensitivity of founder PSVs was evaluated using plasma from multisubtype plasma pools to determine neutralization tier phenotype, as previously established (43, 44). All founder PSVs were categorized as tier 2 (Fig. S1), corresponding to Env trimers that exist in a predominately closed conformation, except for the founder PSV from participant 217-B542219, which was categorized as tier 1b, corresponding to an intermediate Env conformation between closed and open (45).
Development of antibodies capable of neutralizing autologous and heterologous HIV.
Participant plasma samples collected longitudinally from preinfection up to 3 years postinfection were assessed for neutralization capacity against autologous founder and heterologous PSVs. Heterologous viruses included neutralization-sensitive (tier 1), CRF01_AE 92TH023, analyzed separately, and five tier 1 PSVs from subtypes A, B, and C and CRF01_AE that were used to generate a heterologous geometric mean titer (GMT). Plasma neutralization potency against autologous founder PSV, heterologous 92TH023 PSV, and heterologous PSV GMT at five time points is shown in Fig. 1A. Overall, plasma neutralization of the 92TH023 PSV (black dots) was observed earlier and at a higher magnitude over time than that observed for the autologous founder PSVs (Fig. 1A, red dots). Plasma neutralization of heterologous PSV GMT increased progressively over the 3-year period but was lower than both 92TH023 and autologous founder PSV neutralization in both timing and potency. Due to the frequency of plasma collections in the RV217 protocol, we were able to determine the timing of the development of autologous founder and heterologous 92TH023 PSV-neutralizing antibodies. Of the 18 participants, all had developed antibodies capable of neutralizing heterologous 92TH023 PSV by 6 months, yet only 9 participants were able to neutralize their autologous founder virus by 6 months (Fig. 1B). Antibodies capable of neutralizing heterologous 92TH023 PSV developed significantly earlier than antibodies capable of neutralizing the autologous founder PSV mean of 81 versus 234 days, respectively (Fig. 1B; P < 0.001).
FIG 1.
Longitudinal plasma neutralization of CRF01_AE founder viruses. (A) Plasma neutralization was evaluated using the autologous founder Env pseudovirus (PSV, red), heterologous tier 1 CRF01_AE 92TH023 PSV (black), and five tier 1 PSVs from subtypes A, B, and C and CRF01_AE that were used to generate a heterologous geometric mean titer (GMT, gray). Plasma neutralization was determined at multiple time points after infection; key time points are shown. (B) Time to detection of plasma neutralizing activity of the 92TH023 or autologous founder Env PSVs was determined. (C, D) The correlation between the magnitude of autologous founder Env PSV (C) or 92TH023 PSV (D), neutralization, and plasma neutralization breadth, determined using a panel of 35 diverse PSVs, was evaluated for plasma samples collected 1 year after infection. Significant differences between groups were determined by Mann-Whitney test and are indicated above the plots. *, P < 0.05; **, P < 0.005; ***, P < 0.0005. Correlations were evaluated by Spearman correlation analysis, and the trend lines are shown. Plasma with no neutralizing activity were given a 50% inhibitory dilution (ID50) value of 10.
To compare the development of autologous and heterologous neutralizing antibodies and the development of neutralization breadth in CRF01_AE infection, we compared the magnitude of autologous founder and 92TH023 PSV neutralization detected at 1 year with the neutralization breadth at 1 year, measured utilizing a panel of 35 diverse viral strains (7, 46). No association was observed between autologous founder (Fig. 1C) and heterologous 92TH023 (Fig. 1D) PSV neutralization and neutralization breadth at 1 year postinfection.
Timing and potency of autologous neutralizing antibody development.
We next differentiated participants by the timing to development of autologous founder PSV-neutralizing antibodies by utilizing the 6-month time point, at which time 9 participants had developed autologous neutralizing antibodies and 9 participants had not. Participants with early development of autologous neutralizing antibodies (<6 months, shown in red in Fig. 2) developed higher magnitude responses over time, compared to participants with later development of autologous neutralizing antibodies (>6 months; shown in gray in Fig. 2A). Participants with early autologous founder PSV neutralization developed neutralization capacity at a median of 150 days with a median peak plasma neutralization 50% inhibitory dilution (ID50) of 5,798. Plasma neutralization of 4 of these 9 participants exceeded an ID50 of 10,000. Participant 217-B542219, with the founder PSV tier 1b neutralization phenotype, had the most potent autologous neutralizing antibody response. Comparatively, participants with late autologous founder PSV neutralization developed neutralization capacity at a median of 264 days with a median peak neutralization ID50 of 860. Differences in the potency of autologous neutralization between these groups were significant at 6 months, 1 year, and 2 years (Fig. 2B; P < 0.0001, 0.001, and 0.015, respectively); fewer participants had plasma samples available at the 3-year time point due to anti-retroviral therapy (ART) initiation. A significant inverse correlation was observed between the time to development of autologous neutralizing antibodies and the potency of the autologous neutralizing antibodies measured at 1 year (Fig. 2C; P = 0.0001, r = −0.738). These data demonstrate that early development of antibodies capable of neutralizing the autologous founder virus results in more potent autologous neutralizing antibody responses over the course of infection.
FIG 2.
Timing and magnitude of plasma neutralization against the autologous founder Env pseudovirus (PSV). (A) Plasma neutralization of the autologous founder Env PSV was evaluated at multiple time points prior to and throughout infection for each participant; individual responses are shown as lines with evaluated time points marked with circles. Individuals that developed autologous neutralizing antibodies (ANAb) in the first 6 months (early ANAb) are shown in red; individuals that developed autologous neutralizing antibodies after more than 6 months (late ANAb) are shown in gray. Locally estimated scatterplot smoothing (LOESS) regression analysis and ribbons displaying 95% confidence interval was performed through R version 3.5.1. (B) The magnitude of autologous neutralization at key time points is shown for individuals that developed early (red) or late (gray) ANAb. (C) The correlation between the time to development of autologous neutralizing antibodies and their magnitude measured 1 year after infection was evaluated. Significant differences between groups were determined by Mann-Whitney test and are indicated above the plots. *, P < 0.05; **, P < 0.005; ***, P < 0.0005. Correlations were evaluated by Spearman correlation analysis, and the trend line is shown. Plasma samples with no neutralizing activity were given a 50% inhibitory dilution (ID50) value of 10.
We further evaluated the impact of multiple founder env genes on the timing to development of neutralizing antibodies. No significant difference was observed between the 5 participants with multiple founder env genes (median = 253 days, range = 150 to 337 days) and the 13 participants with single founder env genes (median = 175 days, range = 68 to 690 days; P = 0.503; data not shown). For participants with multiple founder env genes, the env with the earliest detectable plasma neutralization was included in Fig. 1 and 2 and all subsequent analyses.
Neutralization sensitivity of CRF01_AE founder Env PSVs.
The neutralization sensitivity of the CRF01_AE founder PSVs to neutralizing monoclonal antibodies (MAbs) was then evaluated to identify potential Env features associated with autologous neutralizing antibody development. We utilized 20 neutralizing MAbs targeting the V1V2, V3, CD4-binding site (CD4bs), or the membrane proximal external region (MPER) of HIV-1 Env. The MAb geometric mean IC50 values are summarized in Table S2. Overall, the CRF01_AE founder Env PSVs were resistant to neutralization by b12 (CD4 bs), 2G12 (gp120 glycan), 447-52D (V3), and PGT151 (gp120-gp41 interface), and more than half of the PSVs were resistant to neutralization by sCD4 (CD4bs), PGT121 (V3), and Z13e1 (MPER).
Participants that developed early autologous founder-neutralizing antibodies had Envs that trended toward higher sensitivity to neutralizing antibodies targeting the V1V2 loop, compared to participants that developed autologous neutralizing antibodies later in infection (Fig. 3A; P = 0.063). No significant differences were observed between groups in their founder Env PSV sensitivity to neutralizing antibodies targeting the CD4bs, V3, or MPER domains (Fig. 3A). Individual V1V2-specific antibodies included in this analysis are shown in Fig. 3B. While the differences between groups are not significant, and the data within each group are highly distributed, there is a trend toward greater V1V2 neutralization sensitivity for the founder Envs from individuals that developed early autologous neutralizing antibodies (Fig. 3B).
FIG 3.
Founder Env features associated with autologous neutralizing antibody development. Neutralization sensitivity of founder Env PSVs was determined using a panel of monoclonal antibodies (MAbs) shown in Table S2. A founder Env pseudoviruses (PSVs) neutralization sensitivity score was calculated as the geometric mean of the 50% inhibitory concentration (IC50) values of MAbs targeting specific Env domains, either the CD4bs, V1V2, V3, or the membrane proximal external region (MPER). (A, B) Differences in founder Env PSV neutralization sensitivity by Env domain scores (A) or individual V1V2 MAbs (B) were evaluated between participants that developed early (red) or late (gray) autologous neutralizing antibody (ANAb) responses. (C) Founder Env V1V2 length was determined and compared between participants that developed early or late ANAb responses. (D, E) The correlation between founder Env V1V2 length and timing of ANAb development (D) or magnitude of the ANAb response 1 year after infection (E) was evaluated. For panels D and E, the colors of the symbols correspond to the categorization of participants by timing of ANAb detection: early (red) or late (gray). The sizes of the symbols correspond to the sensitivity of the founder Env PSV to neutralizing MAbs targeting the V1V2 Env domain. Significant differences between groups were determined by Mann-Whitney test and are indicated above the plots. *, P < 0.05. Correlations were evaluated by Spearman correlation analysis, and the trend lines are shown.
Founder Env V1V2 length associated with development of autologous neutralizing antibodies.
The V1V2 loop length of the founder Envs was determined for comparison with the timing and magnitude of the elicited autologous neutralizing antibody response. Participants that developed early autologous neutralizing antibodies has significantly longer founder Env V1V2 loop lengths compared to participants that developed autologous neutralizing antibodies later in infection (Fig. 3C; P = 0.012). Founder Env V1V2 loop length also correlated indirectly with the timing of autologous neutralizing antibody development (Fig. 3D; P = 0.002, r = −0.675) and directly with the magnitude of autologous founder Env PSV neutralization measured at 1 year (Fig. 3E; P = 0.005, r = 0.635). Founder Envs with longer V1V2 loop lengths were generally more sensitive to neutralization by V1V2-specific neutralizing antibodies; as shown by the symbol size proportional to V1V2 IC50 magnitude in Fig. 3D and E. However, the correlation between V1V2 length and Env V1V2 neutralization sensitivity was not significant (P = 0.6754, r = −0.1060).
These data demonstrate that founder CRF01_AE Envs with longer V1V2 loops elicit more rapid and potent autologous neutralizing antibody responses. Founder Env V1V2 charge and the number of potential N-linked glycosylation sites did not correlate with the timing or magnitude of autologous neutralizing antibody responses (data not shown).
Founder Env PSV V1V2 and V3 neutralization sensitivity associated with development of epitope-specific binding antibodies.
Participant plasma binding antibody responses to V1V2- and V3-specific antigens were evaluated to determine the relationship between autologous founder Env neutralization sensitivity and elicitation of epitope-specific antibody responses. Binding antibody data were available for a subset of participants from an analysis utilizing A244 gp70_V1V2 scaffold protein (gp70_V1V2) and 92TH023 cyclic V3 peptide (cV3) antigens (7). Founder Env PSV V1V2 or V3 neutralization sensitivity directly correlated with the timing to development of V1V2-specific (Fig. S2A) or V3-specific (Fig. S2B) binding antibody responses and indirectly correlated with the magnitude of the V1V2-specific (Fig. S2C) or V3-specific (Fig. S2D) binding antibody responses at 1 year, respectively. These correlations were significant for V3 responses, while the same trend was observed for V1V2 responses. These data indicate that increased neutralization sensitivity of founder CRF01_AE Env V1V2 and V3 domains tends to be associated with elicitation of more rapid and potent V1V2- and V3-specific binding antibody responses. However, no significant relationship was observed between V1V2- or V3-specific binding antibody responses and development of autologous neutralizing antibodies (data not shown). In addition, V1V2 length did not correlate with the timing or magnitude of the V1V2-binding antibody responses (data not shown).
Development of ADCC activity in HIV-1 CRF01_AE infection.
Given the potential protective role of ADCC-mediating Abs in the Thai RV144 vaccine efficacy trial (19) and numerous studies demonstrating the protective effect of ADCC activity (47–49), we further evaluated development of ADCC-mediating antibodies in participant plasma. ADCC activity was measured for all participants using target cells infected with the heterologous CRF01_AE 92TH023 infectious molecular clone (IMC). Most participants (13 of 18) developed antibodies capable of mediating ADCC against the 92TH023 IMC within 6 months (data not shown). ADCC activity increased progressively over time; however, the timing and magnitude of ADCC activity did not correlate with the timing and magnitude of the neutralizing antibody activity (data not shown). Additionally, IMCs were constructed using founder Envs from four participants and were used to measure autologous plasma ADCC against those participants’ autologous founder Env.
Of the four selected participants, three developed early autologous neutralizing antibodies (217-B548973, 217-B542284, and 217-B542219), and one developed autologous neutralizing antibodies later in infection (217-B542691). Plasma ADCC against the 92TH023 and autologous Env IMCs were evaluated longitudinally for the four participants, as shown in Fig. 4. All three participants with early ANAbs (217-B548973, 217-B542219, and 217-B542284) developed antibodies capable of mediating ADCC against the IMC with the autologous Env prior to developing ADCC activity against the heterologous 92TH023 IMC (Fig. 4A to C). The participant with late ANAb development (217-B542691), showed an early, potent heterologous neutralizing antibody response to 92TH023 (Fig. 4D).
FIG 4.
Development of neutralizing and non-neutralizing plasma antibodies. Longitudinal plasma antibody-dependent cellular cytotoxicity (ADCC) activity was evaluated using cell targets infected with the autologous founder Env or heterologous 92TH023 Env infectious molecular clone (IMC). Development of plasma neutralization and ADCC activity are shown for four participants, including three individuals that developed early autologous neutralizing antibody (ANAb): 217-B548973 (A), 217-B542284 (B), and 217-B542219 (C), and one individual that developed late ANAb: 217-B542691 (D). Plasma neutralization is shown with circle symbols; plasma ADCC is shown with square symbols. Activity against the autologous founder Env strain is shown with red symbols; activity against the heterologous 92TH023 Env strain is shown with black symbols.
Because of this interesting observation regarding neutralization noted within these longitudinal data, we then assessed the full longitudinal neutralization profile of autologous founder Env PSV versus heterologous 92TH023 PSV neutralization for all 18 participants. Of the nine participants that developed autologous neutralizing antibodies later in infection, eight developed antibodies capable of neutralizing 92TH023 PSV earlier and more potently (by at least one log) compared to their autologous PSV neutralization (data not shown). However, this was only observed for three of the nine participants that developed early autologous neutralizing antibodies. This indicates a significant difference between the groups (Fisher’s exact test P = 0.049) in terms of the evolution of functional humoral responses to autologous versus tier 1 heterologous virus.
Heterologous neutralization and ADCC responses associated with viral load set point.
The relationships between the participant’s clinical feature of viral load (VL) set point and their plasma antibody neutralization and ADCC functions were then analyzed to better understand the potential role of these immune responses in HIV disease. No correlation was observed between the autologous neutralizing antibody activity measured at 1 year and the VL set point (Fig. 5A). However, neutralization of the neutralization-sensitive (tier 1), heterologous CRF01_AE 92TH023 PSV strain measured at 1 year correlated directly with the VL set point (Fig. 5B; P = 0.007, r = 0.638). Importantly, a striking inverse correlation was also observed between the VL set point and ADCC activity measured at 1 year using the 92TH023 IMC (Fig. 5C; P = 0.011, r = −0.606). The peak of ADCC activity measured over 3 years showed an even stronger inverse correlation with VL set point (data not shown; P = 0.0005, r = −0.736). Plasma 92TH023 neutralization and ADCC activity measured at 1 year correlated inversely, but the trend was not significant (data not shown, P = 0.0835, r = −0.4329). We then compared plasma 92TH023 neutralization and ADCC activity to the plasma IgG subclass distribution for nine individuals for which these data were previously generated (Fig. S3). Neutralization activity correlated directly with CRF01_AE gp140-reactive plasma IgG1 (P = 0.006, and r = 0.850) and IgG4 (P = 0.040, r = 0.705), but not with IgG2 or IgG3. ADCC activity did not significantly correlate with CRF01_AE gp140-reactive plasma IgG1, IgG2, IgG3 or IgG4 (P value range from 0.119 to 0.904; data not shown).
FIG 5.
Relationship between humoral immune responses and viral load. The correlation was evaluated between participant’s set point viral load (VL). (A to C) Magnitude of autologous founder Env pseudovirus (PSV) neutralization (A), heterologous 92TH023 PSV neutralization (B), or heterologous 92TH023 IMC antibody-dependent cellular cytotoxicity (ADCC) activity (C) measured 1 year after infection. (D) Humoral immune responses were categorized as greater than (white) or less than (gray) the median value, and the set point viral load between groups was compared. Correlations were evaluated by Spearman correlation analysis, and the trend lines are shown.
Data endpoints were categorized as values above (white circles) or below (gray circles) the median response for each antibody function, and the VL set points were compared between the two groups with high or low functional antibody responses (Fig. 5D). Participants with ADCC activity above the median had lower VL set points. Higher VL set points tended to be observed in participants with higher levels of 92TH023 neutralization, while no difference was observed for autologous neutralizing antibody responses (Fig. 5D). Thus, participants with immune systems capable of mounting more potent ADCC responses tended to show better control of VL, while higher tier 1 NAbs may be an indicator of the increased antigenic exposure with higher VL.
DISCUSSION
The RV217 cohort provides a unique opportunity to evaluate the development of humoral responses during acute CRF01_AE infection. Utilizing samples from this study, we wanted to determine whether features of the CRF01_AE founder Env could drive the development of functional antibodies, specifically focusing on earlier and higher magnitude neutralizing and ADCC-mediating antibody responses. We found that RV217 participants who developed ANAb responses within 6 months of infection also developed higher magnitude peak ANAb responses. These early responders maintained higher magnitude ANAbs longitudinally for up to 3 years, indicating that earlier responses lead to a greater overall potency of the ANAb response. An increase in antigen diversity during exposure through multiple infections did not result in earlier or greater ANAb titers. We observed a longer time to development of CRF01_AE founder ANAb responses (median of 31 weeks) compared to what has previously been reported for subtypes A (8 weeks) and C (14 weeks) (2, 3, 24, 50). Unfortunately, cross-subtype comparisons were not performed in this study to elucidate differences in humoral immune development against other subtypes. Underlying factors that can influence this outcome are currently being evaluated (7). Further studies are needed to understand the impact of early humoral responses on the evolution of the founder Env and development of neutralization breadth.
In this study, neutralization sensitivity of the CRF01_AE Env to V2 broadly neutralizing monoclonal antibodies (bNAbs) correlated with the development of earlier ANAb responses and V1V2-binding antibodies. A study evaluating Chinese CRF01_AE Envs (8) observed similar neutralization profiles as the Thai CRF01_AE founder Envs described here, including high sensitivity to some V2 bNAbs. The development of V1V2-specific antibodies has been implicated in mediating the first functional ANAb responses in subtype C (32). Analysis of the RV217 founder Env sequences revealed a significant relationship between longer V1V2 lengths and earlier development of ANAb responses, which was not previously observed for subtype C (33). Studies in subtype C infections have shown that shorter V1V2 loops lead to the development of neutralization breadth (34) and longer V1V2 loop lengths in subtype B coincided with increased neutralization resistance (29, 51). Also, in contrast to subtype C, we did not see a relationship between the number of V1V2 potential N-linked glycosylation sites (PNGSs) and ANAb titers (33). Many V2 bNAbs are dependent on glycosylations (52–54) and exert their effects by recognizing the quaternary structure of the V1V2 domains on the trimeric spike (52, 54–56); therefore, these subtype differences in neutralization could potentially be due to differences in Env glycosylation patterns between HIV subtypes. Our results indicate relationships between founder Env V1V2 epitope exposure and neutralization sensitivity during the development of ANAb responses that appear to be specific to CRF01_AE HIV.
In this study, neutralization of the heterologous 92TH023 HIV strain was observed prior to the detection of autologous neutralization of the founder Env, contrary to what has been previously reported for other subtypes (50). Prior vaccine studies have also shown that neutralization of tier 1 viruses, like 92TH023, is predominantly mediated by strain-specific, linear anti-V3 responses (31). In natural infection, these early V3-specific antibodies are not typically neutralizing due to V3 occlusion by V1V2 (57, 58). 92TH023 neutralization was detected as early as 2 months postinfection, and interestingly, individuals that developed earlier 92TH023 neutralization tended to develop later ANAb responses. This suggests a rapid development of decoy antibodies that may distract the immune system from development of more effective antibody responses over time (31, 57, 59–61). Similarly, V3 antibodies have been shown to develop early in subtype C infection, prior to the appearance of ANAb responses (median of 5 weeks versus 19 weeks), but they were not shown to contribute to ANAb responses (32, 33). We observed a correlation between founder Env sensitivity to V3-specific bNAbs and development of V3-binding antibodies, again indicating a relationship between founder Env epitope exposure and the specificity of antibodies elicited during natural CRF01_AE infection. Previous studies have indicated that the immune response to HIV-1 is driven by characteristics of the founder Env (62, 63), further demonstrating the importance of characterizing founder viruses. Additionally, a positive relationship between VL set point and neutralization of 92TH023 from 6 months through 3 years was observed, indicating that a higher antigenic load may lead to a greater magnitude of heterologous neutralization, as has been previously described (4, 5, 64–66).
HIV-1 VL set point has been used to evaluate the progression of HIV-1 disease (67–72) and vaccination efficacy during clinical trials (73) and during treatment-interruption studies (74, 75). Higher VL has been associated with increased mortality (71), while low VL or viremic control is a hallmark of those individuals designated long-term nonprogressors and elite controllers (76). The RV217 acute infection protocol is unique in the inclusion of high-risk participants prior to infection and in a sampling of participant plasma rapidly from very early time points, allowing for the determination of VL peak, nadir, and set point (36). With the availability of these data, we could begin to evaluate relationships between ANAb, heterologous virus neutralization, Env characteristics, and ADCC activity in association with VL set point. We observed an inverse correlation between lower VL set point and higher heterologous ADCC responses at 1 year and with peak ADCC activity observed over the course of infection; 92TH023 heterologous ADCC appears to be associated with viral control.
Interestingly, plasma neutralization of this same viral strain correlated directly with plasma VL. Individuals with higher viral loads have previously been shown to have higher plasma IgG and neutralizing antibody titers (4, 65). We similarly observed a significant positive correlation between plasma VL, neutralization titers, and Env-reactive IgG1. However, IgG levels or subclass did not associate with plasma ADCC activity, indicating that in part, different antibody populations may be mediating these two different plasma antibody functions. This discordance between plasma ADCC and neutralization has been previously demonstrated (15, 77, 78); this relationship may be affected by plasma potency, HIV-1 subtype, and target viral strain. Neutralizing antibodies have been shown to mediate ADCC activity (79), and ADCC-mediating non-neutralizing antibodies exist. Differences in plasma antibody ADCC and neutralizing activity may be observed due to differences in target Env expression between the two assays systems or through targeting of non-Env proteins in the ADCC assay. Additionally, while various ADCC assay formats have been shown to correlate, differences exist in the antigen availability for antibody recognition, as well as other variables (80). We used this particular ADCC assay format as we previously determined that it correlated with other relevant disease and vaccine outcomes (18, 81). However, use of another ADCC assay format could influence the trend observed in this study.
Previous studies have also shown inverse correlations between ADCC activity and VL (82–89). Another study evaluated ADCC activity during primary CRF01_AE HIV infection within a Chinese cohort and detected ADCC activity as early as 52 days postinfection, also noting an inverse correlation between ADCC activity and VL set point (9). ADCC was a potential correlate of protection from acquisition identified in the RV144 clinical trial (18). Studies in vaccinated rhesus macaques also demonstrate protection by ADCC-mediating antibodies and correlation with reduced acute viremia (90). ADCC-mediating antibodies have also been shown to inversely correlate with mother-to-child transmission among subtype A-infected women with high plasma VL (91). A larger collection of autologous founder Env proteins from the RV217 acute infection cohort is currently being evaluated for ADCC using protein-coated targets to delve deeper into the relationship between autologous ADCC and the development of neutralization and ADCC breadth.
The modest 31.2% efficacy of the RV144 vaccine trial highlights the importance of increasing vaccine immunogenicity and efficacy (18, 20). Due to significant genetic diversification of circulating HIV-1 strains observed over the last three decades (92, 93), there is a need to focus vaccine design on more contemporaneous Envs, such as these RV217 founder Envs, which were isolated between 2009 and 2015 (36). Additionally, the emergence of novel CRF01_AE recombinants (8, 94–96) and drug-resistant strains (97–100) has been reported. CRF01_AE infections appear to be rapidly expanding in certain countries and are now the dominant cause of new infections in China, Indonesia, Vietnam, and the Philippines (94, 101–105). Results from this study demonstrate that the humoral immune response to CRF01_AE HIV-1 infection is influenced by Env features, which may, in turn, affect viral load set point. While ANAb was directly related to V1V2 length and sensitivity to V2 MAbs, ANAb unfortunately showed no correlation with VL set point. With the production of autologous envelope proteins and more autologous IMC from the RV217 acute infection cohort, it will be important to study the relationships between autologous ADCC, V1V2 properties, and viral load control. Design of subtype-specific Env vaccine antigens with well defined features that can be related to viral control may then be used to elicit favorable humoral immune responses via vaccination to provide improved protection against HIV infection.
MATERIALS AND METHODS
Ethics statement.
For study RV217/WRAIR#1373 (FWA00000373 and IRB00000794) and study RV229/WRAIR#1386 (source of HIV-negative leukopaks for peripheral blood mononuclear cells [PBMCs] used as effector cells in ADCC assays), all participants were adults and provided written consent. All studies were reviewed and approved by the human ethics and safety committees in each country, as well as by the IRB of Walter Reed Army Institute of Research (Silver Spring, MD, USA), in compliance with all relevant federal guidelines and institutional policies. The human experimentation guidelines of the U.S. Army Surgeon General’s Human Subjects Research Review Board, the ethics review committee of the Ministry of Public Health of Thailand, and the institutional review boards of the Royal Thai Army Medical Department and of Mahidol University (FWA00001813 and IRB00001439) were followed in the conduct of this clinical research.
Production of cohort env clones.
Genotyping and single genome amplification were performed as previously described (106, 107) to determine env sequence and subtype. Founder env sequences were isolated and analyzed from plasma samples taken within 1 week after HIV detection. Viral RNA was isolated using the QIAamp viral RNA minikit (Qiagen, Hilden, Germany) and was converted to cDNA through nested reverse transcription (RT)-PCR using the SuperScript III first-strand synthesis system (Invitrogen, Waltham, MA, USA). The env genes were amplified by nested PCR and then were sequenced using an ABI 3730xl DNA analyzer (Applied Biosystems, Waltham, MA, USA). Genotyping of HIV-1 viruses were performed as described previously by quantitative PCR amplification (92, 108). env genes were amplified with Platinum Taq polymerase (Invitrogen, Waltham, MA, USA); the PCR products were qualified through gel electrophoresis, extracted, and column-purified using the Macherey-Nagel nucleic acid purification kit (Clontech, Kyoto, Japan). The purified PCR products were then ligated into the pcDNA 3.1/V5-His TOPO TA (Invitrogen, Waltham, MA, USA) using the manufacturer’s recommendations. The ligation products were transformed into STBL4 cells (Invitrogen, Waltham, MA, USA), DNA clones with the correct insert were purified using the PureYield plasmid miniprep kit (Promega, Madison, WI, USA), and infectivity was screened using TZM-bl cells. The sequences were verified using an ABI 3730xl DNA sequencer (Applied Biosystems, Waltham, MA, USA). env genes with minimal mutations were mutated back to the consensus using QuikChange XL site-directed mutagenesis kit (Agilent, Santa Clara, CA, USA), or this was performed commercially (GeneWiz, Chelmsford, MA, USA). Two env genes, 217-B544996.F1 and 217-B547460.F1, were commercially synthesized and ligated into the pcDNA 3.1 vector (GenScript, Piscataway, NJ, USA).
Virus stock production and titration.
Viruses were produced by transfection of HEK 293T cells. For PSV production, env DNA (8 μg) and pSG3Δenv (24 μg) were combined with X-tremeGENE 9 DNA transfection reagent (Sigma-Aldrich, St. Louis, MO, USA) at a 1:3 ratio as per the manufacturer’s recommendations, in serum-free Dulbecco’s modified Eagle’s medium (DMEM). For chimeric infectious molecular clone (CHIMC) production, 15 μg of plasmid DNA were combined with FuGENE 6 (Promega, Madison, WI, USA) at a 1:4 ratio as per manufacturer recommendations in serum-free DMEM. The transfection mix was incubated with the cells for 6 h at 37°C, and then the medium was replaced. Virus supernatants were harvested 72 h after transfection, filtered through a 0.22-μm PES membrane (Millipore, Burlington, MA, USA), and stored in 3-mL aliquots at −80°C. Viruses were titrated on TZM-bl cells in an endpoint dilution assay. Fourfold serial dilutions of PSV were added to 96-well black-walled plates (Corning, Corning, NY, USA) in a final volume of 25 μL/well. An additional 25 μL of supplemented DMEM was added to each well. TZM-bl cells were added to plates in 50 μL of supplemented DMEM with 40 μg/mL of DEAE-dextran (Sigma, St. Louis, MO, USA). Virus and cells were incubated together for 48 h at 37°C. Infectivity was measured by luminescence after adding 100 μL of the Bright-Glo luciferase assay system (Promega, Madison, WI, USA) to each well; luminescence was quantified using the Envision luminometer (Perkin Elmer, Waltham, MA, USA).
GHOST cell assays.
The GHOST cell infection assay was used to determine coreceptor usage of viral stocks. Parental, CXCR4-expressing, or CCR5-expressing GHOST cells were cultured in 24-well plates. The cells were infected with undiluted PSV in the presence of 20 μg/mL of Polybrene infection reagent (Millipore-Sigma, Burlington, MA, USA) for 4 h. The cells were washed, cultured for 2 days, and then harvested and fixed with 2% paraformaldehyde. The percentage of infected cells expressing green fluorescent protein (GFP) was measured by flow cytometry using a LSRII cytometer and FACSDIVA software (Becton, Dickinson, Franklin Lakes, NJ, USA). Postgating analysis was conducted with FlowJo software (FlowJo, LLC, Ashland, OR, USA). PSVs were designated using the CCR5 or CXCR4 receptor if the percentage of GFP-positive cells for either the CCR5- or CXCR4- cell lines was 5-fold greater than the percentage of GFP-positive cells in the parental line. Assay controls included an uninfected negative-control, murine leukemia virus (MuLV) as a positive control for all lines and BaL.01 (CCR5-tropic) and MN.3 (CXCR4-tropic) PSVs as specific cell line controls.
Antibodies, proteins, and plasma.
The following reagents were obtained through the National Institutes of Health (NIH) AIDS Reagent Program, Division of AIDS, NIAID, NIH: soluble CD4 recombinant protein (sCD4) from Progenics; VRC01 from John Mascola; 3BNC117 from Michel C. Nussenzweig; 10E8 from Mark Connors; PGT121, PGT126, and PGT145 from IAVI; and HIV-IG from NABI and NHLBI. B12, 2G12, PG9, PG16, 2F5, 447-52D, 4E10, and Z13e1 were purchased from Polymun. PGDM1400, PGT130, and PGT151 were graciously provided by Dennis Burton and Devin Sok at IAVI. VRC42.01 was generously provided by John Mascola and Nicole Doria-Rose at the Vaccine Research Center, NIH. PGT128 was made in-house by Vincent Dussupt. Anti-HIV-1 gp120 MAbs 697-30D was generously provided by Susan Zolla-Pazner, and anti-RSV was purchased from MedImmune (Frederick, MD, USA). Recombinant proteins (AE A244 gp70V1V2) utilized for surface plasmon resonance (SPR; Biacore, Uppsala, Sweden) were obtained from Bart Haynes and Hua-Xin Liao (Duke Protein Production Facility). RV217 study samples, viral load, and cell count data were collected as described by Robb et al. (36). Plasma pools A, B, C, D, CRF01_AE, CRF02_AG, and 21.2B (subtype B) and uninfected normal human sera were generated as described by Brown et al. (12).
Virus neutralization.
Virus neutralization assays were performed to test plasma and MAb neutralization of PSVs. Plasma samples were prepared by heat inactivation at 56°C for 45 min, followed by 5-min centrifugation at 2,000 rpm to remove particulates. Plasma samples were used at a starting dilution of 1:20 in supplemented DMEM; MAbs were used at a starting concentration of 25 μg/mL in supplemented DMEM. Viruses were diluted to a concentration to yield luminescence at least three times above the cell-only controls, as determined by the PSV titration. Neutralization reagents were serially diluted 1:4 in independent duplicates per plate and incubated with equal volumes of virus for 1 h at 37°C before adding TZM-bl cells with 40 μg/mL of DEAE-dextran (Sigma, St. Louis, MO, USA). The plates were incubated at 37°C for 48 h before reading the luminescence. Nonspecific neutralization by plasma was evaluated using MuLV env DNA pseudotyped with the pSG3Δenv backbone DNA. Uninfected normal human serum was also tested against all the PSVs as a negative control.
Antibody-dependent cellular cytotoxicity studies.
ADCC experiments were performed using CEM.NKR-CCR5+LUC+ target cells, as described previously (109). The 217-B548973, 217-B542284, 217-B542691, and 217-B542219 env genes were cloned into the CRF01_AE 40061. LucR backbone as described previously (110). The 40061.LucR_TH023Env CHIMC was then optimized for infection of CEM.NKR-CCR5+LUC+ target cells with 5 μg/mL DEAE-dextran (Sigma-Aldrich, St. Louis, MO, USA). 40061.LucR_217-B548973.F1env, 40061.LucR_217-B542284.F1env, 40061.LucR_217-B542691.F1env, and 40061.LucR_217-B542219.F1env CHIMC infections were optimized with 10 μg/mL DEAE-dextran for autologous ADCC studies. Renilla luciferase activity was read using ViviRen live cell substrate (Promega, Madison, WI, USA), and infection was optimized to yield luminescence three times above the cell-only controls, with cell viability of >60%. ADCC activity was tested using an HIV+ plasma pool and HIVIG as positive controls and HIV-negative donor plasma as a negative control. The test plasma samples were serially diluted 8-fold with a 1:100 starting dilution in a 96-well white-walled half-area plate (Corning, Corning, NY, USA). Infected target cells and effector peripheral blood mononuclear cells (PBMCs) were mixed at a 1:30 ratio (0.2 million cells/mL and 6.0 million cells/mL, respectively) and added to plates for a total volume of 50 μL/well. The plates were incubated for 30 min at room temperature with shaking at 300 rpm, centrifuged at 300 rpm for 1 min, and incubated for an additional 5.5 h at 37°C before reading live cell luminescence with ViviRen. Healthy donor PBMCs from the RV229 protocol were used in all subsequent ADCC experiments with RV217 plasma. ADCC activity (percentage of specific killing) was calculated as the decrease in infected cell luminescence at 1:800 dilution of plasma for all time points. Background levels of ADCC were evaluated using autologous preinfection plasma samples at the same dilution and were used to correct the infected plasma ADCC response. Responses of ≥2% were considered positive (after background subtraction of nonspecific killing in the presence of preinfection plasma).
Data and statistical analyses.
Neutralization data were analyzed using LabKey software and fit to a five-parameter equation, using an average of assay duplicates. The data reported are also averages of values derived from two independent experiments. Plasma samples that failed to neutralize virus were assigned a reciprocal ID50 of 10 for statistical purposes. ADCC data are represented as percentage of specific killing, calculated by the reduction of luminescence of infected cells with respect to the untreated infected cell control; the data are the averages of duplicates and represent the average of the percentage of specific killing. Statistical analyses were performed using GraphPad Prism version 7.0a as well as R version 3.5.1 and utilized data from 1 founder Env per participant.
ACKNOWLEDGMENTS
We gratefully acknowledge the RV217 study participants for donating their plasma samples to this study. We also thank the protocol staff and the personnel at Armed Forces Research Institute of Medical Sciences, Bangkok, Thailand, who collected plasma samples and supported this study. We are thankful for the technical support provided by Elizabeth Martinez, Christopher Jewell, Brendan Mann and Daniel Silas. We also acknowledge the generosity of David Montefiori at Duke University for providing the MN.3 gp160 expression vector and Nicole Doria-Rose and John Mascola at the Vaccine Research Center, National Institutes of Health, for providing the VRC42.01 MAb; Susan Zolla-Pazner for providing 697-30D; and Dennis Burton and Devin Sok for contribution of PGDM1400, PGT130, and PGT151. We are also grateful to Whitney Edwards and Linda Sharp at Duke University for support with the ADCC assay and data analysis.
This work was supported by cooperative agreement No. W81XWH-18-2-0040 between the Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., and the U.S. Department of Defense. H.V.T. was partially supported by Swiss National Science Foundation grant No. P3SMP3_148406/1. The views expressed are those of the authors and should not be construed to represent the positions of the U.S. Army or the Department of Defense. The investigators have adhered to the policies for protection of human subjects as prescribed in AR 70–25.
Conceptualization, S.K.G., L.W., and V.R.P.; Performing experiments, S.K.G., B.M.B., M.Z., and S.M.; Contributing new reagents/analytic tools, E.S.-B., A.L.C., M.B., G.F., and S.T.; Contributing resources and data, E.S.-B., H.V.T., A.L.C., M.B., G.D., S.S., S.K., S.N., L.-A.E., M.R., and S.J.K.; Contributing to provision of clinical samples, S.S., S.K., S.N., L.-A.E., N.L.M., and M.L.R.; Providing financial oversight and guidance, N.L.M. and J.A.A.; Data analysis, S.K.G. and L.W.; Writing, S.K.G., L.W., and V.R.P.
We declare no conflict of interest.
Footnotes
Supplemental material is available online only.
Contributor Information
Victoria R. Polonis, Email: vpolonis@hivresearch.org.
Guido Silvestri, Emory University.
REFERENCES
- 1.Bbosa N, Kaleebu P, Ssemwanga D. 2019. HIV subtype diversity worldwide. Curr Opin HIV AIDS 14:153–160. 10.1097/COH.0000000000000534. [DOI] [PubMed] [Google Scholar]
- 2.Liao HX, Lynch R, Zhou T, Gao F, Alam SM, Boyd SD, Fire AZ, Roskin KM, Schramm CA, Zhang Z, Zhu J, Shapiro L, Program NCS, Mullikin JC, Gnanakaran S, Hraber P, Wiehe K, Kelsoe G, Yang G, Xia SM, Montefiori DC, Parks R, Lloyd KE, Scearce RM, Soderberg KA, Cohen M, Kamanga G, Louder MK, Tran LM, Chen Y, Cai F, Chen S, Moquin S, Du X, Joyce MG, Srivatsan S, Zhang B, Zheng A, Shaw GM, Hahn BH, Kepler TB, Korber BT, Kwong PD, Mascola JR, Haynes BF, NISC Comparative Sequencing Program . 2013. Co-evolution of a broadly neutralizing HIV-1 antibody and founder virus. Nature 496:469–476. 10.1038/nature12053. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Murphy MK, Yue L, Pan R, Boliar S, Sethi A, Tian J, Pfafferot K, Karita E, Allen SA, Cormier E, Goepfert PA, Borrow P, Robinson JE, Gnanakaran S, Hunter E, Kong XP, Derdeyn CA. 2013. Viral escape from neutralizing antibodies in early subtype A HIV-1 infection drives an increase in autologous neutralization breadth. PLoS Pathog 9:e1003173. 10.1371/journal.ppat.1003173. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Gray ES, Madiga MC, Hermanus T, Moore PL, Wibmer CK, Tumba NL, Werner L, Mlisana K, Sibeko S, Williamson C, Abdool KS, Morris L, Team CS, CAPRISA002 Study Team . 2011. The neutralization breadth of HIV-1 develops incrementally over four years and is associated with CD4+ T cell decline and high viral load during acute infection. J Virol 85:4828–4840. 10.1128/JVI.00198-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Mikell I, Sather DN, Kalams SA, Altfeld M, Alter G, Stamatatos L. 2011. Characteristics of the earliest cross-neutralizing antibody response to HIV-1. PLoS Pathog 7:e1001251. 10.1371/journal.ppat.1001251. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Hraber P, Seaman MS, Bailer RT, Mascola JR, Montefiori DC, Korber BT. 2014. Prevalence of broadly neutralizing antibody responses during chronic HIV-1 infection. AIDS 28:163–169. 10.1097/QAD.0000000000000106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Townsley SM, Donofrio GC, Jian N, Leggat DJ, Dussupt V, Mendez-Rivera L, Eller LA, Cofer L, Choe M, Ehrenberg PK, Geretz A, Gift S, Grande R, Lee A, Peterson C, Piechowiak MB, Slike BM, Tran U, Joyce MG, Georgiev IS, Rolland M, Thomas R, Tovanabutra S, Doria-Rose NA, Polonis VR, Mascola JR, McDermott AB, Michael NL, Robb ML, Krebs SJ. 2021. B cell engagement with HIV-1 founder virus envelope predicts development of broadly neutralizing antibodies. Cell Host Microbe 29:564–578.e9. 10.1016/j.chom.2021.01.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Wang H, Yuan T, Li T, Li Y, Qian F, Zhu C, Liang S, Hoffmann D, Dittmer U, Sun B, Yang R. 2018. Evaluation of susceptibility of HIV-1 CRF01_AE variants to neutralization by a panel of broadly neutralizing antibodies. Arch Virol 163:3303–3315. 10.1007/s00705-018-4011-7. [DOI] [PubMed] [Google Scholar]
- 9.Chen X, Lin M, Qian S, Zhang Z, Fu Y, Xu J, Han X, Ding H, Dong T, Shang H, Jiang Y. 2018. The early antibody-dependent cell-mediated cytotoxicity response is associated with lower viral set point in individuals with primary HIV infection. Front Immunol 9:2322. 10.3389/fimmu.2018.02322. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Schader SM, Colby-Germinario SP, Quashie PK, Oliveira M, Ibanescu RI, Moisi D, Mespléde T, Wainberg MA. 2012. HIV gp120 H375 is unique to HIV-1 subtype CRF01_AE and confers strong resistance to the entry inhibitor BMS-599793, a candidate microbicide drug. Antimicrob Agents Chemother 56:4257–4267. 10.1128/AAC.00639-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Bricault CA, Yusim K, Seaman MS, Yoon H, Theiler J, Giorgi EE, Wagh K, Theiler M, Hraber P, Macke JP, Kreider EF, Learn GH, Hahn BH, Scheid JF, Kovacs JM, Shields JL, Lavine CL, Ghantous F, Rist M, Bayne MG, Neubauer GH, McMahan K, Peng H, Chéneau C, Jones JJ, Zeng J, Ochsenbauer C, Nkolola JP, Stephenson KE, Chen B, Gnanakaran S, Bonsignori M, Williams LD, Haynes BF, Doria-Rose N, Mascola JR, Montefiori DC, Barouch DH, Korber B. 2019. HIV-1 neutralizing antibody signatures and application to epitope-targeted vaccine design. Cell Host Microbe 25:59–72.e8. 10.1016/j.chom.2018.12.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Brown BK, Darden JM, Tovanabutra S, Oblander T, Frost J, Sanders-Buell E, de Souza MS, Birx DL, McCutchan FE, Polonis VR. 2005. Biologic and genetic characterization of a panel of 60 human immunodeficiency virus type 1 isolates, representing clades A, B, C, D, CRF01_AE, and CRF02_AG, for the development and assessment of candidate vaccines. J Virol 79:6089–6101. 10.1128/JVI.79.10.6089-6101.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Utachee P, Jinnopat P, Isarangkura-Na-Ayuthaya P, de Silva UC, Nakamura S, Siripanyaphinyo U, Wichukchinda N, Tokunaga K, Yasunaga T, Sawanpanyalert P, Ikuta K, Auwanit W, Kameoka M. 2009. Genotypic characterization of CRF01_AE env genes derived from human immunodeficiency virus type 1-infected patients residing in central Thailand. AIDS Res Hum Retroviruses 25:229–236. 10.1089/aid.2008.0232. [DOI] [PubMed] [Google Scholar]
- 14.Zoubchenok D, Veillette M, Prevost J, Sanders-Buell E, Wagh K, Korber B, Chenine AL, Finzi A. 2017. Histidine 375 modulates CD4 binding in HIV-1 CRF01_AE envelope glycoproteins. J Virol 91:e02151-16. 10.1128/JVI.02151-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Mielke D, Stanfield-Oakley S, Borate B, Fisher LH, Faircloth K, Tuyishime M, Greene K, Gao H, Williamson C, Morris L, Ochsenbauer C, Tomaras G, Haynes BF, Montefiori D, Pollara J, deCamp AC, Ferrari G. 2022. Selection of HIV envelope strains for standardized assessments of vaccine-elicited antibody-dependent cellular cytotoxicity-mediating antibodies. J Virol 96:e0164321. 10.1128/JVI.01643-21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Prevost J, Zoubchenok D, Richard J, Veillette M, Pacheco B, Coutu M, Brassard N, Parsons MS, Ruxrungtham K, Bunupuradah T, Tovanabutra S, Hwang KK, Moody MA, Haynes BF, Bonsignori M, Sodroski J, Kaufmann DE, Shaw GM, Chenine AL, Finzi A. 2017. Influence of the envelope gp120 Phe 43 cavity on HIV-1 sensitivity to antibody-dependent cell-mediated cytotoxicity responses. J Virol 91:e02452-16. 10.1128/JVI.02452-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Veillette M, Coutu M, Richard J, Batraville LA, Dagher O, Bernard N, Tremblay C, Kaufmann DE, Roger M, Finzi A. 2015. The HIV-1 gp120 CD4-bound conformation is preferentially targeted by antibody-dependent cellular cytotoxicity-mediating antibodies in sera from HIV-1-infected individuals. J Virol 89:545–551. 10.1128/JVI.02868-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Haynes BF, Gilbert PB, McElrath MJ, Zolla-Pazner S, Tomaras GD, Alam SM, Evans DT, Montefiori DC, Karnasuta C, Sutthent R, Liao HX, DeVico AL, Lewis GK, Williams C, Pinter A, Fong Y, Janes H, DeCamp A, Huang Y, Rao M, Billings E, Karasavvas N, Robb ML, Ngauy V, de Souza MS, Paris R, Ferrari G, Bailer RT, Soderberg KA, Andrews C, Berman PW, Frahm N, De Rosa SC, Alpert MD, Yates NL, Shen X, Koup RA, Pitisuttithum P, Kaewkungwal J, Nitayaphan S, Rerks-Ngarm S, Michael NL, Kim JH. 2012. Immune-correlates analysis of an HIV-1 vaccine efficacy trial. N Engl J Med 366:1275–1286. 10.1056/NEJMoa1113425. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Bonsignori M, Pollara J, Moody MA, Alpert MD, Chen X, Hwang KK, Gilbert PB, Huang Y, Gurley TC, Kozink DM, Marshall DJ, Whitesides JF, Tsao CY, Kaewkungwal J, Nitayaphan S, Pitisuttithum P, Rerks-Ngarm S, Kim JH, Michael NL, Tomaras GD, Montefiori DC, Lewis GK, DeVico A, Evans DT, Ferrari G, Liao HX, Haynes BF. 2012. Antibody-dependent cellular cytotoxicity-mediating antibodies from an HIV-1 vaccine efficacy trial target multiple epitopes and preferentially use the VH1 gene family. J Virol 86:11521–11532. 10.1128/JVI.01023-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Rerks-Ngarm S, Pitisuttithum P, Nitayaphan S, Kaewkungwal J, Chiu J, Paris R, Premsri N, Namwat C, de Souza M, Adams E, Benenson M, Gurunathan S, Tartaglia J, McNeil JG, Francis DP, Stablein D, Birx DL, Chunsuttiwat S, Khamboonruang C, Thongcharoen P, Robb ML, Michael NL, Kunasol P, Kim JH, Investigators M-T . 2009. Vaccination with ALVAC and AIDSVAX to prevent HIV-1 infection in Thailand. N Engl J Med 361:2209–2220. 10.1056/NEJMoa0908492. [DOI] [PubMed] [Google Scholar]
- 21.Karasavvas N, Billings E, Rao M, Williams C, Zolla-Pazner S, Bailer RT, Koup RA, Madnote S, Arworn D, Shen X, Tomaras GD, Currier JR, Jiang M, Magaret C, Andrews C, Gottardo R, Gilbert P, Cardozo TJ, Rerks-Ngarm S, Nitayaphan S, Pitisuttithum P, Kaewkungwal J, Paris R, Greene K, Gao H, Gurunathan S, Tartaglia J, Sinangil F, Korber BT, Montefiori DC, Mascola JR, Robb ML, Haynes BF, Ngauy V, Michael NL, Kim JH, de Souza MS, MOPH TAVEG Collaboration . 2012. The Thai phase III HIV type 1 vaccine trial (RV144) regimen induces antibodies that target conserved regions within the V2 loop of gp120. AIDS Res Hum Retroviruses 28:1444–1457. 10.1089/aid.2012.0103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Zolla-Pazner S, deCamp A, Gilbert PB, Williams C, Yates NL, Williams WT, Howington R, Fong Y, Morris DE, Soderberg KA, Irene C, Reichman C, Pinter A, Parks R, Pitisuttithum P, Kaewkungwal J, Rerks-Ngarm S, Nitayaphan S, Andrews C, O’Connell RJ, Yang ZY, Nabel GJ, Kim JH, Michael NL, Montefiori DC, Liao HX, Haynes BF, Tomaras GD. 2014. Vaccine-induced IgG antibodies to V1V2 regions of multiple HIV-1 subtypes correlate with decreased risk of HIV-1 infection. PLoS One 9:e87572. 10.1371/journal.pone.0087572. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Wood N, Bhattacharya T, Keele BF, Giorgi E, Liu M, Gaschen B, Daniels M, Ferrari G, Haynes BF, McMichael A, Shaw GM, Hahn BH, Korber B, Seoighe C. 2009. HIV evolution in early infection: selection pressures, patterns of insertion and deletion, and the impact of APOBEC. PLoS Pathog 5:e1000414. 10.1371/journal.ppat.1000414. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Wei X, Decker JM, Wang S, Hui H, Kappes JC, Wu X, Salazar-Gonzalez JF, Salazar MG, Kilby JM, Saag MS, Komarova NL, Nowak MA, Hahn BH, Kwong PD, Shaw GM. 2003. Antibody neutralization and escape by HIV-1. Nature 422:307–312. 10.1038/nature01470. [DOI] [PubMed] [Google Scholar]
- 25.Julien JP, Cupo A, Sok D, Stanfield RL, Lyumkis D, Deller MC, Klasse PJ, Burton DR, Sanders RW, Moore JP, Ward AB, Wilson IA. 2013. Crystal structure of a soluble cleaved HIV-1 envelope trimer. Science 342:1477–1483. 10.1126/science.1245625. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Kwon YD, Finzi A, Wu X, Dogo-Isonagie C, Lee LK, Moore LR, Schmidt SD, Stuckey J, Yang Y, Zhou T, Zhu J, Vicic DA, Debnath AK, Shapiro L, Bewley CA, Mascola JR, Sodroski JG, Kwong PD. 2012. Unliganded HIV-1 gp120 core structures assume the CD4-bound conformation with regulation by quaternary interactions and variable loops. Proc Natl Acad Sci USA 109:5663–5668. 10.1073/pnas.1112391109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Ly A, Stamatatos L. 2000. V2 loop glycosylation of the human immunodeficiency virus type 1 SF162 envelope facilitates interaction of this protein with CD4 and CCR5 receptors and protects the virus from neutralization by anti-V3 loop and anti-CD4 binding site antibodies. J Virol 74:6769–6776. 10.1128/jvi.74.15.6769-6776.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Lyumkis D, Julien JP, de Val N, Cupo A, Potter CS, Klasse PJ, Burton DR, Sanders RW, Moore JP, Carragher B, Wilson IA, Ward AB. 2013. Cryo-EM structure of a fully glycosylated soluble cleaved HIV-1 envelope trimer. Science 342:1484–1490. 10.1126/science.1245627. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.van Gils MJ, Bunnik EM, Boeser-Nunnink BD, Burger JA, Terlouw-Klein M, Verwer N, Schuitemaker H. 2011. Longer V1V2 region with increased number of potential N-linked glycosylation sites in the HIV-1 envelope glycoprotein protects against HIV-specific neutralizing antibodies. J Virol 85:6986–6995. 10.1128/JVI.00268-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Pinter A, Honnen WJ, He Y, Gorny MK, Zolla-Pazner S, Kayman SC. 2004. The V1/V2 domain of gp120 is a global regulator of the sensitivity of primary human immunodeficiency virus type 1 isolates to neutralization by antibodies commonly induced upon infection. J Virol 78:5205–5215. 10.1128/jvi.78.10.5205-5215.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Sanders RW, van Gils MJ, Derking R, Sok D, Ketas TJ, Burger JA, Ozorowski G, Cupo A, Simonich C, Goo L, Arendt H, Kim HJ, Lee JH, Pugach P, Williams M, Debnath G, Moldt B, van Breemen MJ, Isik G, Medina-Ramirez M, Back JW, Koff WC, Julien JP, Rakasz EG, Seaman MS, Guttman M, Lee KK, Klasse PJ, LaBranche C, Schief WR, Wilson IA, Overbaugh J, Burton DR, Ward AB, Montefiori DC, Dean H, Moore JP. 2015. HIV-1 vaccines: HIV-1 neutralizing antibodies induced by native-like envelope trimers. Science 349:aac4223. 10.1126/science.aac4223. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Moore PL, Gray ES, Choge IA, Ranchobe N, Mlisana K, Abdool KS, Williamson C, Morris L, Team CS. 2008. The C3-V4 region is a major target of autologous neutralizing antibodies in human immunodeficiency virus type 1 subtype C infection. J Virol 82:1860–1869. 10.1128/JVI.02187-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Gray ES, Moore PL, Choge IA, Decker JM, Bibollet-Ruche F, Li H, Leseka N, Treurnicht F, Mlisana K, Shaw GM, Karim SS, Williamson C, Morris L, Team CS, CAPRISA 002 Study Team . 2007. Neutralizing antibody responses in acute human immunodeficiency virus type 1 subtype C infection. J Virol 81:6187–6196. 10.1128/JVI.00239-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Rademeyer C, Moore PL, Taylor N, Martin DP, Choge IA, Gray ES, Sheppard HW, Gray C, Morris L, Williamson C, Team H, HIVNET 028 Study Team . 2007. Genetic characteristics of HIV-1 subtype C envelopes inducing cross-neutralizing antibodies. Virology 368:172–181. 10.1016/j.virol.2007.06.013. [DOI] [PubMed] [Google Scholar]
- 35.Sagar M, Wu X, Lee S, Overbaugh J. 2006. Human immunodeficiency virus type 1 V1-V2 envelope loop sequences expand and add glycosylation sites over the course of infection, and these modifications affect antibody neutralization sensitivity. J Virol 80:9586–9598. 10.1128/JVI.00141-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Robb ML, Eller LA, Kibuuka H, Rono K, Maganga L, Nitayaphan S, Kroon E, Sawe FK, Sinei S, Sriplienchan S, Jagodzinski LL, Malia J, Manak M, de Souza MS, Tovanabutra S, Sanders-Buell E, Rolland M, Dorsey-Spitz J, Eller MA, Milazzo M, Li Q, Lewandowski A, Wu H, Swann E, O’Connell RJ, Peel S, Dawson P, Kim JH, Michael NL, Team RVS, RV 217 Study Team . 2016. Prospective study of acute HIV-1 infection in adults in East Africa and Thailand. N Engl J Med 374:2120–2130. 10.1056/NEJMoa1508952. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Chu M, Zhang W, Zhang X, Jiang W, Huan X, Meng X, Zhu B, Yang Y, Tao Y, Tian T, Lu Y, Jiang L, Zhang L, Zhuang X. 2017. HIV-1 CRF01_AE strain is associated with faster HIV/AIDS progression in Jiangsu Province. Sci Rep 7:1570. 10.1038/s41598-017-01858-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Indriati DW, Witaningrum AM, Yunifiar MQ, Khairunisa SQ, Ueda S, Kotaki T, Nasronudin N, Kameoka M. 2020. The dominance of CRF01_AE and the emergence of drug resistance mutations among antiretroviral therapy-experienced, HIV-1-infected individuals in Medan, Indonesia. Acta Med Indones 52:366–374. [PubMed] [Google Scholar]
- 39.Moore PL, Gray ES, Morris L. 2009. Specificity of the autologous neutralizing antibody response. Curr Opin HIV AIDS 4:358–363. 10.1097/COH.0b013e32832ea7e8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Zhu T, Mo H, Wang N, Nam DS, Cao Y, Koup RA, Ho DD. 1993. Genotypic and phenotypic characterization of HIV-1 patients with primary infection. Science 261:1179–1181. 10.1126/science.8356453. [DOI] [PubMed] [Google Scholar]
- 41.Van't Wout AB, Kootstra NA, Mulder-Kampinga GA, Albrecht-van Lent N, Scherpbier HJ, Veenstra J, Boer K, Coutinho RA, Miedema F, Schuitemaker H. 1994. Macrophage-tropic variants initiate human immunodeficiency virus type 1 infection after sexual, parenteral, and vertical transmission. J Clin Invest 94:2060–2067. 10.1172/JCI117560. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.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 USA 105:7552–7557. 10.1073/pnas.0802203105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Seaman MS, Janes H, Hawkins N, Grandpre LE, Devoy C, Giri A, Coffey RT, Harris L, Wood B, Daniels MG, Bhattacharya T, Lapedes A, Polonis VR, McCutchan FE, Gilbert PB, Self SG, Korber BT, Montefiori DC, Mascola JR. 2010. Tiered categorization of a diverse panel of HIV-1 Env pseudoviruses for assessment of neutralizing antibodies. J Virol 84:1439–1452. 10.1128/JVI.02108-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Brown BK, Wieczorek L, Sanders-Buell E, Rosa Borges A, Robb ML, Birx DL, Michael NL, McCutchan FE, Polonis VR. 2008. Cross-clade neutralization patterns among HIV-1 strains from the six major clades of the pandemic evaluated and compared in two different models. Virology 375:529–538. 10.1016/j.virol.2008.02.022. [DOI] [PubMed] [Google Scholar]
- 45.Montefiori DC, Roederer M, Morris L, Seaman MS. 2018. Neutralization tiers of HIV-1. Curr Opin HIV AIDS 13:128–136. 10.1097/COH.0000000000000442. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Georgiev IS, Doria-Rose NA, Zhou T, Kwon YD, Staupe RP, Moquin S, Chuang GY, Louder MK, Schmidt SD, Altae-Tran HR, Bailer RT, McKee K, Nason M, O'Dell S, Ofek G, Pancera M, Srivatsan S, Shapiro L, Connors M, Migueles SA, Morris L, Nishimura Y, Martin MA, Mascola JR, Kwong PD. 2013. Delineating antibody recognition in polyclonal sera from patterns of HIV-1 isolate neutralization. Science 340:751–756. 10.1126/science.1233989. [DOI] [PubMed] [Google Scholar]
- 47.Suryawanshi P, Bagul R, Shete A, Thakar M. 2021. Anti-HIV-1 ADCC and HIV-1 Env can be partners in reducing latent HIV reservoir. Front Immunol 12:663919. 10.3389/fimmu.2021.663919. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Mielke D, Bandawe G, Zheng J, Jones J, Abrahams MR, Bekker V, Ochsenbauer C, Garrett N, Abdool Karim S, Moore PL, Morris L, Montefiori D, Anthony C, Ferrari G, Williamson C. 2021. ADCC-mediating non-neutralizing antibodies can exert immune pressure in early HIV-1 infection. PLoS Pathog 17:e1010046. 10.1371/journal.ppat.1010046. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Talathi S, Bagul R, Ghate M, Kulkarni S, Thakar M. 2020. Higher baseline ADCC responses in chronic nonprogressive HIV infection are associated with reduced HIV burden in later course of disease. Viral Immunol 33:77–85. 10.1089/vim.2019.0137. [DOI] [PubMed] [Google Scholar]
- 50.Moog C, Fleury HJ, Pellegrin I, Kirn A, Aubertin AM. 1997. Autologous and heterologous neutralizing antibody responses following initial seroconversion in human immunodeficiency virus type 1-infected individuals. J Virol 71:3734–3741. 10.1128/JVI.71.5.3734-3741.1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.van den Kerkhof TL, Feenstra KA, Euler Z, van Gils MJ, Rijsdijk LW, Boeser-Nunnink BD, Heringa J, Schuitemaker H, Sanders RW. 2013. HIV-1 envelope glycoprotein signatures that correlate with the development of cross-reactive neutralizing activity. Retrovirology 10:102. 10.1186/1742-4690-10-102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Walker LM, Huber M, Doores KJ, Falkowska E, Pejchal R, Julien JP, Wang SK, Ramos A, Chan-Hui PY, Moyle M, Mitcham JL, Hammond PW, Olsen OA, Phung P, Fling S, Wong CH, Phogat S, Wrin T, Simek MD, Protocol GPI, Koff WC, Wilson IA, Burton DR, Poignard P. 2011. Broad neutralization coverage of HIV by multiple highly potent antibodies. Nature 477:466–470. 10.1038/nature10373. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Sok D, van Gils MJ, Pauthner M, Julien JP, Saye-Francisco KL, Hsueh J, Briney B, Lee JH, Le KM, Lee PS, Hua Y, Seaman MS, Moore JP, Ward AB, Wilson IA, Sanders RW, Burton DR. 2014. Recombinant HIV envelope trimer selects for quaternary-dependent antibodies targeting the trimer apex. Proc Natl Acad Sci USA 111:17624–17629. 10.1073/pnas.1415789111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Walker LM, Phogat SK, Chan-Hui PY, Wagner D, Phung P, Goss JL, Wrin T, Simek MD, Fling S, Mitcham JL, Lehrman JK, Priddy FH, Olsen OA, Frey SM, Hammond PW, Protocol GPI, Kaminsky S, Zamb T, Moyle M, Koff WC, Poignard P, Burton DR, Protocol G Principal Investigators . 2009. Broad and potent neutralizing antibodies from an African donor reveal a new HIV-1 vaccine target. Science 326:285–289. 10.1126/science.1178746. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Doores KJ, Burton DR. 2010. Variable loop glycan dependency of the broad and potent HIV-1-neutralizing antibodies PG9 and PG16. J Virol 84:10510–10521. 10.1128/JVI.00552-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Julien JP, Lee JH, Cupo A, Murin CD, Derking R, Hoffenberg S, Caulfield MJ, King CR, Marozsan AJ, Klasse PJ, Sanders RW, Moore JP, Wilson IA, Ward AB. 2013. Asymmetric recognition of the HIV-1 trimer by broadly neutralizing antibody PG9. Proc Natl Acad Sci USA 110:4351–4356. 10.1073/pnas.1217537110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Davis KL, Gray ES, Moore PL, Decker JM, Salomon A, Montefiori DC, Graham BS, Keefer MC, Pinter A, Morris L, Hahn BH, Shaw GM. 2009. High titer HIV-1 V3-specific antibodies with broad reactivity but low neutralizing potency in acute infection and following vaccination. Virology 387:414–426. 10.1016/j.virol.2009.02.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Krachmarov CP, Honnen WJ, Kayman SC, Gorny MK, Zolla-Pazner S, Pinter A. 2006. Factors determining the breadth and potency of neutralization by V3-specific human monoclonal antibodies derived from subjects infected with clade A or clade B strains of human immunodeficiency virus type 1. J Virol 80:7127–7135. 10.1128/JVI.02619-05. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.de Taeye SW, de la Pena AT, Vecchione A, Scutigliani E, Sliepen K, Burger JA, van der Woude P, Schorcht A, Schermer EE, van Gils MJ, LaBranche CC, Montefiori DC, Wilson IA, Moore JP, Ward AB, Sanders RW. 2018. Stabilization of the gp120 V3 loop through hydrophobic interactions reduces the immunodominant V3-directed non-neutralizing response to HIV-1 envelope trimers. J Biol Chem 293:1688–1701. 10.1074/jbc.RA117.000709. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.de Taeye SW, Ozorowski G, Torrents de la Pena A, Guttman M, Julien JP, van den Kerkhof TL, Burger JA, Pritchard LK, Pugach P, Yasmeen A, Crampton J, Hu J, Bontjer I, Torres JL, Arendt H, DeStefano J, Koff WC, Schuitemaker H, Eggink D, Berkhout B, Dean H, LaBranche C, Crotty S, Crispin M, Montefiori DC, Klasse PJ, Lee KK, Moore JP, Wilson IA, Ward AB, Sanders RW. 2015. Immunogenicity of stabilized HIV-1 envelope trimers with reduced exposure of non-neutralizing epitopes. Cell 163:1702–1715. 10.1016/j.cell.2015.11.056. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.McGuire AT, Dreyer AM, Carbonetti S, Lippy A, Glenn J, Scheid JF, Mouquet H, Stamatatos L. 2014. HIV antibodies: antigen modification regulates competition of broad and narrow neutralizing HIV antibodies. Science 346:1380–1383. 10.1126/science.1259206. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Hessell AJ, Malherbe DC, Pissani F, McBurney S, Krebs SJ, Gomes M, Pandey S, Sutton WF, Burwitz BJ, Gray M, Robins H, Park BS, Sacha JB, LaBranche CC, Fuller DH, Montefiori DC, Stamatatos L, Sather DN, Haigwood NL. 2016. Achieving potent autologous neutralizing antibody responses against Tier 2 HIV-1 viruses by strategic selection of envelope immunogens. J Immunol 196:3064–3078. 10.4049/jimmunol.1500527. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Kouyos RD, Rusert P, Kadelka C, Huber M, Marzel A, Ebner H, Schanz M, Liechti T, Friedrich N, Braun DL, Scherrer AU, Weber J, Uhr T, Baumann NS, Leemann C, Kuster H, Chave JP, Cavassini M, Bernasconi E, Hoffmann M, Calmy A, Battegay M, Rauch A, Yerly S, Aubert V, Klimkait T, Boni J, Metzner KJ, Gunthard HF, Trkola A, Swiss H, Swiss HIV Cohort Study . 2018. Tracing HIV-1 strains that imprint broadly neutralizing antibody responses. Nature 561:406–410. 10.1038/s41586-018-0517-0. [DOI] [PubMed] [Google Scholar]
- 64.Piantadosi A, Panteleeff D, Blish CA, Baeten JM, Jaoko W, McClelland RS, Overbaugh J. 2009. Breadth of neutralizing antibody response to human immunodeficiency virus type 1 is affected by factors early in infection but does not influence disease progression. J Virol 83:10269–10274. 10.1128/JVI.01149-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Sather DN, Armann J, Ching LK, Mavrantoni A, Sellhorn G, Caldwell Z, Yu X, Wood B, Self S, Kalams S, Stamatatos L. 2009. Factors associated with the development of cross-reactive neutralizing antibodies during human immunodeficiency virus type 1 infection. J Virol 83:757–769. 10.1128/JVI.02036-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.van Gils MJ, Euler Z, Schweighardt B, Wrin T, Schuitemaker H. 2009. Prevalence of cross-reactive HIV-1-neutralizing activity in HIV-1-infected patients with rapid or slow disease progression. AIDS 23:2405–2414. 10.1097/QAD.0b013e32833243e7. [DOI] [PubMed] [Google Scholar]
- 67.Ho DD. 1996. Viral counts count in HIV infection. Science 272:1124–1125. 10.1126/science.272.5265.1124. [DOI] [PubMed] [Google Scholar]
- 68.Mellors JW, Munoz A, Giorgi JV, Margolick JB, Tassoni CJ, Gupta P, Kingsley LA, Todd JA, Saah AJ, Detels R, Phair JP, Rinaldo CR, Jr. 1997. Plasma viral load and CD4+ lymphocytes as prognostic markers of HIV-1 infection. Ann Intern Med 126:946–954. 10.7326/0003-4819-126-12-199706150-00003. [DOI] [PubMed] [Google Scholar]
- 69.Mellors JW, Rinaldo CR, Jr, Gupta P, White RM, Todd JA, Kingsley LA. 1996. Prognosis in HIV-1 infection predicted by the quantity of virus in plasma. Science 272:1167–1170. 10.1126/science.272.5265.1167. [DOI] [PubMed] [Google Scholar]
- 70.Blattner WA, Oursler KA, Cleghorn F, Charurat M, Sill A, Bartholomew C, Jack N, O’Brien T, Edwards J, Tomaras G, Weinhold K, Greenberg M. 2004. Rapid clearance of virus after acute HIV-1 infection: correlates of risk of AIDS. J Infect Dis 189:1793–1801. 10.1086/386306. [DOI] [PubMed] [Google Scholar]
- 71.Lavreys L, Baeten JM, Chohan V, McClelland RS, Hassan WM, Richardson BA, Mandaliya K, Ndinya-Achola JO, Overbaugh J. 2006. Higher set point plasma viral load and more-severe acute HIV type 1 (HIV-1) illness predict mortality among high-risk HIV-1-infected African women. Clin Infect Dis 42:1333–1339. 10.1086/503258. [DOI] [PubMed] [Google Scholar]
- 72.Okulicz JF, Marconi VC, Landrum ML, Wegner S, Weintrob A, Ganesan A, Hale B, Crum-Cianflone N, Delmar J, Barthel V, Quinnan G, Agan BK, Dolan MJ, Infectious Disease Clinical Research Program HIV Working Group . 2009. Clinical outcomes of elite controllers, viremic controllers, and long-term nonprogressors in the US Department of Defense HIV natural history study. J Infect Dis 200:1714–1723. 10.1086/646609. [DOI] [PubMed] [Google Scholar]
- 73.Buchbinder SP, Mehrotra DV, Duerr A, Fitzgerald DW, Mogg R, Li D, Gilbert PB, Lama JR, Marmor M, Del Rio C, McElrath MJ, Casimiro DR, Gottesdiener KM, Chodakewitz JA, Corey L, Robertson MN, Step Study Protocol Team . 2008. Efficacy assessment of a cell-mediated immunity HIV-1 vaccine (the Step Study): a double-blind, randomised, placebo-controlled, test-of-concept trial. Lancet 372:1881–1893. 10.1016/S0140-6736(08)61591-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Angel JB, Routy JP, Graziani GM, Tremblay CL. 2015. The effect of therapeutic HIV vaccination with ALVAC-HIV with or without remune on the size of the viral reservoir (A CTN 173 Substudy). J Acquir Immune Defic Syndr 70:122–128. 10.1097/QAI.0000000000000734. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Treasure GC, Aga E, Bosch RJ, Mellors JW, Kuritzkes DR, Para M, Gandhi RT, Li JZ. 2016. Brief report: relationship among viral load outcomes in HIV treatment interruption trials. J Acquir Immune Defic Syndr 72:310–313. 10.1097/QAI.0000000000000964. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Grabar S, Selinger-Leneman H, Abgrall S, Pialoux G, Weiss L, Costagliola D. 2009. Prevalence and comparative characteristics of long-term nonprogressors and HIV controller patients in the French Hospital Database on HIV. AIDS 23:1163–1169. 10.1097/QAD.0b013e32832b44c8. [DOI] [PubMed] [Google Scholar]
- 77.Smalls-Mantey A, Doria-Rose N, Klein R, Patamawenu A, Migueles SA, Ko SY, Hallahan CW, Wong H, Liu B, You L, Scheid J, Kappes JC, Ochsenbauer C, Nabel GJ, Mascola JR, Connors M. 2012. Antibody-dependent cellular cytotoxicity against primary HIV-infected CD4+ T cells is directly associated with the magnitude of surface IgG binding. J Virol 86:8672–8680. 10.1128/JVI.00287-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Borggren M, Jensen SS, Heyndrickx L, Palm AA, Gerstoft J, Kronborg G, Hønge BL, Jespersen S, da Silva ZJ, Karlsson I, Fomsgaard A. 2016. Neutralizing antibody response and antibody-dependent cellular cytotoxicity in HIV-1-infected individuals from Guinea-Bissau and Denmark. AIDS Res Hum Retroviruses 32:434–442. 10.1089/aid.2015.0118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Bruel T, Guivel-Benhassine F, Amraoui S, Malbec M, Richard L, Bourdic K, Donahue DA, Lorin V, Casartelli N, Noël N, Lambotte O, Mouquet H, Schwartz O. 2016. Elimination of HIV-1-infected cells by broadly neutralizing antibodies. Nat Commun 7:10844. 10.1038/ncomms10844. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Lewis GK, Ackerman ME, Scarlatti G, Moog C, Robert-Guroff M, Kent SJ, Overbaugh J, Reeves RK, Ferrari G, Thyagarajan B. 2019. Knowns and unknowns of assaying antibody-dependent cell-mediated cytotoxicity against HIV-1. Front Immunol 10:1025. 10.3389/fimmu.2019.01025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Barrows BM, Krebs SJ, Jian N, Zemil M, Slike BM, Dussupt V, Tran U, Mendez-Rivera L, Chang D, O’Sullivan AM, Mann B, Sanders-Buell E, Shubin Z, Creegan M, Paquin-Proulx D, Ehrenberg P, Laurence-Chenine A, Srithanaviboonchai K, Thomas R, Eller MA, Ferrari G, Robb M, Rao V, Tovanabutra S, Polonis VR, Wieczorek L. 2022. Fc receptor engagement of HIV-1 Env-specific antibodies in mothers and infants predicts reduced vertical transmission. Front Immunol 13:1051501. 10.3389/fimmu.2022.1051501. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Ahmad R, Sindhu ST, Toma E, Morisset R, Vincelette J, Menezes J, Ahmad A. 2001. Evidence for a correlation between antibody-dependent cellular cytotoxicity-mediating anti-HIV-1 antibodies and prognostic predictors of HIV infection. J Clin Immunol 21:227–233. 10.1023/A:1011087132180. [DOI] [PubMed] [Google Scholar]
- 83.Battle-Miller K, Eby CA, Landay AL, Cohen MH, Sha BE, Baum LL. 2002. Antibody-dependent cell-mediated cytotoxicity in cervical lavage fluids of human immunodeficiency virus type 1–infected women. J Infect Dis 185:439–447. 10.1086/338828. [DOI] [PubMed] [Google Scholar]
- 84.Forthal DN, Landucci G, Daar ES. 2001. Antibody from patients with acute human immunodeficiency virus (HIV) infection inhibits primary strains of HIV type 1 in the presence of natural-killer effector cells. J Virol 75:6953–6961. 10.1128/JVI.75.15.6953-6961.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Forthal DN, Landucci G, Keenan B. 2001. Relationship between antibody-dependent cellular cytotoxicity, plasma HIV type 1 RNA, and CD4+ lymphocyte count. AIDS Res Hum Retroviruses 17:553–561. 10.1089/08892220151126661. [DOI] [PubMed] [Google Scholar]
- 86.Jia M, Li D, He X, Zhao Y, Peng H, Ma P, Hong K, Liang H, Shao Y. 2013. Impaired natural killer cell-induced antibody-dependent cell-mediated cytotoxicity is associated with human immunodeficiency virus-1 disease progression. Clin Exp Immunol 171:107–116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Johansson SE, Rollman E, Chung AW, Center RJ, Hejdeman B, Stratov I, Hinkula J, Wahren B, Karre K, Kent SJ, Berg L. 2011. NK cell function and antibodies mediating ADCC in HIV-1-infected viremic and controller patients. Viral Immunol 24:359–368. 10.1089/vim.2011.0025. [DOI] [PubMed] [Google Scholar]
- 88.Kulkarni A, Kurle S, Shete A, Ghate M, Godbole S, Madhavi V, Kent SJ, Paranjape R, Thakar M. 2017. Indian long-term non-progressors show broad ADCC responses with preferential recognition of V3 region of envelope and a region from Tat protein. Front Immunol 8:5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Lambotte O, Ferrari G, Moog C, Yates NL, Liao HX, Parks RJ, Hicks CB, Owzar K, Tomaras GD, Montefiori DC, Haynes BF, Delfraissy JF. 2009. Heterogeneous neutralizing antibody and antibody-dependent cell cytotoxicity responses in HIV-1 elite controllers. AIDS 23:897–906. 10.1097/QAD.0b013e328329f97d. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Gomez-Roman VR, Patterson LJ, Venzon D, Liewehr D, Aldrich K, Florese R, Robert-Guroff M. 2005. Vaccine-elicited antibodies mediate antibody-dependent cellular cytotoxicity correlated with significantly reduced acute viremia in rhesus macaques challenged with SIVmac251. J Immunol 174:2185–2189. 10.4049/jimmunol.174.4.2185. [DOI] [PubMed] [Google Scholar]
- 91.Mabuka J, Nduati R, Odem-Davis K, Peterson D, Overbaugh J. 2012. HIV-specific antibodies capable of ADCC are common in breastmilk and are associated with reduced risk of transmission in women with high viral loads. PLoS Pathog 8:e1002739. 10.1371/journal.ppat.1002739. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Kijak GH, Tovanabutra S, Rerks-Ngarm S, Nitayaphan S, Eamsila C, Kunasol P, Khamboonruang C, Thongcharoen P, Namwat C, Premsri N, Benenson M, Morgan P, Bose M, Sanders-Buell E, Paris R, Robb ML, Birx DL, De Souza MS, McCutchan FE, Michael NL, Kim JH. 2013. Molecular evolution of the HIV-1 Thai epidemic between the time of RV144 immunogen selection to the execution of the vaccine efficacy trial. J Virol 87:7265–7281. 10.1128/JVI.03070-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Tovanabutra S, Beyrer C, Sakkhachornphop S, Razak MH, Ramos GL, Vongchak T, Rungruengthanakit K, Saokhieo P, Tejafong K, Kim B, De Souza M, Robb ML, Birx DL, Jittiwutikarn J, Suriyanon V, Celentano DD, McCutchan FE. 2004. The changing molecular epidemiology of HIV type 1 among northern Thai drug users, 1999 to 2002. AIDS Res Hum Retroviruses 20:465–475. 10.1089/088922204323087705. [DOI] [PubMed] [Google Scholar]
- 94.Chang D, Sanders-Buell E, Bose M, O’Sullivan AM, Pham P, Kroon E, Colby DJ, Sirijatuphat R, Billings E, Pinyakorn S, Chomchey N, Rutvisuttinunt W, Kijak G, de Souza M, Excler JL, Phanuphak P, Phanuphak N, O’Connell RJ, Kim JH, Robb ML, Michael NL, Ananworanich J, Tovanabutra S, RV254/SEARCH 010 Study Group . 2018. Molecular epidemiology of a primarily MSM acute HIV-1 cohort in Bangkok, Thailand and connections within networks of transmission in Asia. J Int AIDS Soc 21:e25204. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Sahbandar IN, Takahashi K, Djoerban Z, Firmansyah I, Naganawa S, Motomura K, Sato H, Kitamura K, Pohan HT, Sato S. 2009. Current HIV type 1 molecular epidemiology profile and identification of unique recombinant forms in Jakarta, Indonesia. AIDS Res Hum Retroviruses 25:637–646. 10.1089/aid.2008.0266. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.SahBandar IN, Takahashi K, Motomura K, Djoerban Z, Firmansyah I, Kitamura K, Sato H, Pohan HT, Sato S. 2011. The indonesian variants of CRF33_01B: near-full length sequence analysis. AIDS Res Hum Retroviruses 27:97–102. 10.1089/aid.2010.0163. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Budayanti NS, Merati TP, Bela B, Mahardika GN. 2019. Molecular antiretroviral resistance markers of human immunodeficiency virus-1 of CRF01_AE subtype in Bali, Indonesia. Curr HIV Res 16:374–382. 10.2174/1570162X17666190204101154. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Dong K, Ye L, Leng Y, Liang S, Feng L, Yang H, Su L, Li Y, Baloch S, He F, Yuan D, Pei X. 2019. Prevalence of HIV-1 drug resistance among patients with antiretroviral therapy failure in Sichuan, China, 2010–2016. Tohoku J Exp Med 247:1–12. 10.1620/tjem.247.1. [DOI] [PubMed] [Google Scholar]
- 99.Iemwimangsa N, Pasomsub E, Sukasem C, Chantratita W. 2017. Surveillance of HIV-1 drug-resistance mutations in Thailand from 1999 to 2014. Southeast Asian J Trop Med Public Health 48:271–281. [PubMed] [Google Scholar]
- 100.Ueda S, Witaningrum AM, Khairunisa SQ, Kotaki T, Nasronudin Kameoka M. 2018. Genetic diversity and drug resistance of HIV-1 circulating in North Sulawesi, Indonesia. AIDS Res Hum Retroviruses 35:407–413. [DOI] [PubMed] [Google Scholar]
- 101.Chen Y, Hora B, DeMarco T, Berba R, Register H, Hood S, Carter M, Stone M, Pappas A, Sanchez AM, Busch M, Denny TN, Gao F. 2019. Increased predominance of HIV-1 CRF01_AE and its recombinants in the Philippines. J Gen Virol 100:511–522. 10.1099/jgv.0.001198. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Feng Y, He X, Hsi JH, Li F, Li X, Wang Q, Ruan Y, Xing H, Lam TT, Pybus OG, Takebe Y, Shao Y. 2013. The rapidly expanding CRF01_AE epidemic in China is driven by multiple lineages of HIV-1 viruses introduced in the 1990s. AIDS 27:1793–1802. 10.1097/QAD.0b013e328360db2d. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Hemelaar J, Gouws E, Ghys PD, Osmanov S, WHO-UNAIDS Network for HIV Isolation and Characterisation . 2011. Global trends in molecular epidemiology of HIV-1 during 2000–2007. AIDS 25:679–689. 10.1097/QAD.0b013e328342ff93. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Merati TP, Ryan CE, Spelmen T, Wirawan DN, Bakta IM, Otto B, Oelrichs RB, Crowe SM. 2012. CRF01_AE dominates the HIV-1 epidemic in Indonesia. Sex Health 9:414–421. 10.1071/SH11121. [DOI] [PubMed] [Google Scholar]
- 105.Taylor BS, Sobieszczyk ME, McCutchan FE, Hammer SM. 2008. The challenge of HIV-1 subtype diversity. N Engl J Med 358:1590–1602. 10.1056/NEJMra0706737. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Kijak GH, Sanders-Buell E, Chenine AL, Eller MA, Goonetilleke N, Thomas R, Leviyang S, Harbolick EA, Bose M, Pham P, Oropeza C, Poltavee K, O’Sullivan AM, Billings E, Merbah M, Costanzo MC, Warren JA, Slike B, Li H, Peachman KK, Fischer W, Gao F, Cicala C, Arthos J, Eller LA, O’Connell RJ, Sinei S, Maganga L, Kibuuka H, Nitayaphan S, Rao M, Marovich MA, Krebs SJ, Rolland M, Korber BT, Shaw GM, Michael NL, Robb ML, Tovanabutra S, Kim JH. 2017. Rare HIV-1 transmitted/founder lineages identified by deep viral sequencing contribute to rapid shifts in dominant quasispecies during acute and early infection. PLoS Pathog 13:e1006510. 10.1371/journal.ppat.1006510. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Salazar-Gonzalez JF, Salazar MG, Keele BF, Learn GH, Giorgi EE, Li H, Decker JM, Wang S, Baalwa J, Kraus MH, Parrish NF, Shaw KS, Guffey MB, Bar KJ, Davis KL, Ochsenbauer-Jambor C, Kappes JC, Saag MS, Cohen MS, Mulenga J, Derdeyn CA, Allen S, Hunter E, Markowitz M, Hraber P, Perelson AS, Bhattacharya T, Haynes BF, Korber BT, Hahn BH, Shaw GM. 2009. Genetic identity, biological phenotype, and evolutionary pathways of transmitted/founder viruses in acute and early HIV-1 infection. J Exp Med 206:1273–1289. 10.1084/jem.20090378. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Tovanabutra S, Sanders EJ, Graham SM, Mwangome M, Peshu N, McClelland RS, Muhaari A, Crossler J, Price MA, Gilmour J, Michael NL, McCutchan FM. 2010. Evaluation of HIV type 1 strains in men having sex with men and in female sex workers in Mombasa, Kenya. AIDS Res Hum Retroviruses 26:123–131. 10.1089/aid.2009.0115. [DOI] [PubMed] [Google Scholar]
- 109.Joachim A, Nilsson C, Aboud S, Bakari M, Lyamuya EF, Robb ML, Marovich MA, Earl P, Moss B, Ochsenbauer C, Wahren B, Mhalu F, Sandstrom E, Biberfeld G, Ferrari G, Polonis VR. 2015. Potent functional antibody responses elicited by HIV-I DNA priming and boosting with heterologous HIV-1 recombinant MVA in healthy Tanzanian adults. PLoS One 10:e0118486. 10.1371/journal.pone.0118486. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Chenine AL, Merbah M, Wieczorek L, Molnar S, Mann B, Lee J, O’Sullivan AM, Bose M, Sanders-Buell E, Kijak GH, Herrera C, McLinden R, O’Connell RJ, Michael NL, Robb ML, Kim JH, Polonis VR, Tovanabutra S. 2018. Neutralization sensitivity of a novel HIV-1 CRF01_AE panel of infectious molecular clones. J Acquir Immune Defic Syndr 78:348–355. 10.1097/QAI.0000000000001675. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Tables S1 and S2 and Fig. S1 to S3. Download jvi.01635-22-s0001.pdf, PDF file, 0.4 MB (376.1KB, pdf)





