Reviews:

Microbial Cell, Vol. 11, No. 1, pp. 207 - 220; doi: 10.15698/mic2024.07.826

Neutralizing the threat: harnessing broadly neutralizing antibodies against HIV-1 for treatment and prevention

Juan C Becerra1, Lauren Hitchcock1, Khoa Vu1 and Johannes S Gach1

Download PDF download pdf
Show/hide additional information

    1 Department of Medicine, Division of Infectious Diseases, University of California, CA, 92697, Irvine, Irvine, USA.

Keywords: HIV1, bnAbs, prevention, treatment, viral reservoir. Abbreviations:

ADCC - antibody-dependent cellular cytotoxicity,

ADCP - antibody-dependent cellular phagocytosis

ART - antiretroviral therapy

bnAbs - broadly neutralizing antibodies,

CDC - complement-dependent cytotoxicity,

CDCP - complement-dependent cellular phagocytosis,

HIV-1 - human immunodeficiency virus-1,

IgG - immunoglobulin G.
Received originally: 17/10/2023 Received in revised form: 06/05/2024
Accepted: 15/05/2024 Published: 03/07/2024

Correspondence:
Johannes S Gach, Department of Medicine, Division of Infectious Diseases, University of California, Irvine, Irvine, CA 92697, USA; jgach@uci.edu

Conflict of interest statement: The authors declare that no competing interest exists.
Please cite this article as: Juan C Becerra, Lauren Hitchcock, Khoa Vu, Johannes S Gach (2024). Neutralizing the threat: harnessing broadly neutralizing antibodies against HIV-1 for treatment and prevention. Microbial Cell 11: 207-220. doi: 10.15698/mic2024.07.826

Abstract

Broadly neutralizing antibodies (bnAbs) targeting the human immunodeficiency virus-1 (HIV-1) have played a crucial role in elucidating and characterizing neutralization-sensitive sites on the HIV-1 envelope spike and in informing vaccine development. Continual advancements in identifying more potent bnAbs, along with their capacity to trigger antibody-mediated effector functions, coupled with modifications to extend their half-life, position them as promising candidates for both HIV-1 treatment and prevention. While current  pharmacological interventions have made significant progress in managing HIV-1 infection and enhancing quality of life, no definitive cure or vaccines have been developed thus far. Standard treatments involve daily oral anti-retroviral therapy, which, despite its efficacy, can lead to notable long-term side effects. Recent clinical trial data have demonstrated encouraging therapeutic and preventive potential for bnAb therapies in both HIV-1-infected individuals and those without the infection. This review provides an overview of the advancements in HIV- 1-specific bnAbs and discusses the insights gathered from recent clinical trials regarding their application in treating and preventing HIV-1 infection.

INTRODUCTION

Antibody-mediated immunity is essential for the host defense system by eliminating disease-causing pathogens such as viruses or bacteria and preventing them from infecting human cells 1. Antibodies of the immunoglobulin G (IgG) isotype are the most abundant in humans and can recognize antigens with one or two Fab regions and interact with Fc gamma receptors (FcγRs) through the immunoglobulin Fc region 2, 3. IgG plays a vital role in the protection against human immunodeficiency virus-1 (HIV-1) infection, as demonstrated by numerous non-human primate studies 4, 5.

 

HIV-1 envelope spikes (Figure 1A), responsible for mediating the attachment and fusion between viral and host cell membranes 6, represent the primary targets of neutralizing immune responses in HIV-1-infected individuals 7, 8. However, HIV-1 has evolved evasive mechanisms by displaying major sequence variations on the envelope spike as well as a dense array of host-derived, immunologically “self”, N-linked glycans that efficiently shield the underlying protein from host antibody responses 9, 10. Moreover, HIV-1 virions typically exhibit a limited quantity of intact or functional envelope spikes on their surface, averaging seven to 14 spikes per viral particle 11, 12, 13. As a result, multivalent engagement with B cell receptors and, consequently, the B cell immune response against HIV-1 is limited 14, 15.

FIGURE 1: Examples of potential forms of HIV-1 envelope on the viral surface. (A) Intact envelope spikes consist of three non-covalently bound gp120/gp41 heterodimers embedded in the viral membrane via a transmembrane domain and a cytoplasmic tail. (B) Free monomeric gp120, released through envelope shedding. (C) Shaved gp41 stumps on the viral surface.


 

Furthermore, low spike numbers decrease the likelihood of bivalent antibody binding that might otherwise enhance neutralization 8, 16 or other antibody-mediated effector functions 17. In addition to the sparse number of functional envelope spikes, a high prevalence of non-functional forms of envelope, acting as decoys 18, 19, is present, which include soluble monomeric gp120 (Figure 1B) and fusion-incompetent gp41 stumps on the viral surface (Figure 1C). These non-functional forms of envelope succeed in diverting the immune responses toward immunodominant, non-neutralizing, epitopes rather than to the less accessible neutralizing epitopes 20. Accordingly, the first autologous neutralizing antibodies emerge relatively late following HIV-1 transmission (i.e., ~ twelve or more weeks) 21. These initial autologous strain-specific neutralizing antibodies are capable of blocking viral entry and preventing further host cell infection to some extent by (i) interfering with the engagement of CD4 receptors or co-receptors CCR5 and CXCR4, (ii) stabilizing the pre-fusion envelope spike to prevent membrane fusion, or (iii) accelerating envelope spike decay thereby applying selection pressure on cell-free HIV-1 virions 22.

 

Nevertheless, once HIV-1 enters and integrates into the host genome the error-prone viral reverse transcriptase, with a mutation rate of 5.4 x 10-5 per base per replication cycle 23, introduces random mutations into the envelope gene that can affect antibody-binding epitopes contributing neutralization-resistant variants that can continue infecting new cells 24. As a result, high mutation rates, combined with a short replication cycle and a tendency for recombination, induce quasispecies and generate viral diversity within an individual over the course of HIV-1 infection 25. A study that followed HIV-1 quasispecies generation in an infected couple revealed that, during the early stages, the distribution of virion varieties formed relatively tight phylogenetic clusters, becoming increasingly dispersed over time, with a corresponding increase in genetic diversity after one year 26. After three years, the viral sequences obtained from both partners gave rise to distinct phylogenetic clusters in the phylogenetic tree 26. Therefore, autologous neutralizing antibody responses are generally not associated with control of viremia in most HIV-1 infected individuals and lag behind viral escape variants 27.

 

While almost all infected individuals develop strain-specific neutralizing antibodies, specifically against autologous viruses, large clinical cohort studies revealed that approximately 10–30% of HIV-1 infections result in some level of serum neutralization breadth after several years of infection 28. In rare cases, around 1-2% of HIV-1-infected individuals, termed elite neutralizers, develop broadly neutralizing antibodies (bnAbs) 29, 30. The neutralization breadth of an antibody refers to its ability to recognize and neutralize different clades and strains of HIV-1 by targeting conserved sites on the functional envelope trimer 31. BnAbs can recognize and inactivate many different heterologous HIV-1 variants, while low-breadth autologous antibodies are more specific to the initially transmitted founder strains 31. The neutralization breadth of most bnAbs increases with the time of infection as they go through several rounds of somatic hypermutation 32. Nevertheless, the pathway to neutralization breadth also differs depending on the epitope being targeted 33.

BINDING EPITOPES OF HIV-1 bnAbs

Recent advances in stabilizing and expressing a nearly complete form of the HIV-1 trimer vastly refined our understanding of the metastability of the trimer and its structures allowing for the detailed characterization of bnAb epitopes 34, 35. Sites of vulnerability (Figure 2) to bnAbs include the CD4 binding site (CD4bs), the V1/V2 loops at the trimer apex, the V3 glycan high mannose supersite, the silent face of gp120, the gp120-gp41 interface, the gp41 fusion peptide, and the membrane-proximal external region (MPER) 35, 36, 37, 38. Examples of bnAbs and their respective targets are shown in Table 1. Epitopes on the high-mannose patch are of particular interest for vaccine design considering they are frequently targeted by various classes of bnAbs, which do not require multiple rounds of maturation 39. A recent study that investigated an HIV-1 V2 loop-directed bnAb lineage reported that extensive antibody somatic hypermutation does not necessarily result in increased breadth and that many mutations do not impact or negatively affect breadth 40. While the majority of bnAbs can interact with both trimeric and monomeric envelope spikes, some bnAbs require intact quaternary structures that are believed to form only on the functional trimer 41, 42. In contrast, non-neutralizing antibodies typically bind to monomeric envelope subunits 43.

FIGURE 2: Sites of vulnerability on an HIV-1 envelope spike. BnAbs can be directed against several epitopes on gp120 including the CD4bs, the V1/V2 apex, the V3 glycan supersite site, and the silent face as well as epitopes on gp41 such as the gp41-gp120 interface region, the fusion peptide and the MPER. Examples of bnAbs targeting these sites are shown in Table 1.


Table 1: Examples of broadly neutralizing HIV-1 envelope-specific antibodies and their respective binding sites.

Envelope Subunit

Epitope

Specific antibodies

gp120

V1/V2 apex

PG9 44; PG16 45; PGT141-145 46; CH01-04 47; PGDM1400 48; CAP256-VRC26.01-33 49; VRC38.01 50; PCT64 51

V3 glycan supersite

2G12 52; PGT121 46; PGT128 46; PGT135 46;10-1074 53; PCDN-33A 54; BG18 55; DH270.6 56

CD4bs

B12 57; VRC01 58; 3BNC117 59; 8ANC131 60; CH235.N6 61; PGV04 62; N49-P7 63; ACS101-103 64

Silent face

VRC-PG05 65; SF12 66

gp120/gp41 interface

8ANC195 67 ; 35022 68

gp41

Fusion peptide

VRC34.01 69; ACS202 70; PGT151 46

MPER

4E10 71; 2F5 71; 10E8 72; DH511 72

CD4bs = CD4 binding site, MPER = membrane proximal external region.

EFFECTOR FUNCTIONS OF HIV-1 bnAbs

Recent data indicates that the principal mode of protection conferred by bnAbs involves the direct neutralization of virions 73, 74. However, bnAbs also elicit innate effector mechanisms via Fc-mediated interactions with FcγRs present on diverse immune cell types including natural killer (NK) cells, macrophages, and neutrophils 75, 76. This engagement triggers processes such as antibody-dependent cellular cytotoxicity (ADCC) and antibody-dependent cellular phagocytosis (ADCP), both of which may substantially augment the overall protective efficacy 77, 78. Additionally, Fc-mediated complement-dependent effector functions, including complement-dependent cytotoxicity (CDC) or complement-dependent cellular phagocytosis (CDCP), have also been associated with the elimination of HIV-1 79. Both effector functions are usually initiated upon binding many molecules of IgG to a multivalent antigen or by high local antigen densities, which in turn allow the multimerization (i.e., hexamerization) of neighboring IgG-Fc domains 3, 79, 80. The hexameric structure forms an ideal platform for complement component 1q attachment and activation of the complement cascade that results in CDC or CDCP 81.

 

Clinical evidence of a protective role for Fc-mediated effector functions in HIV-1-infected individuals has been demonstrated in several studies 82, 83, 84, 85, 86, 87. For example, elevated levels of virus-specific ADCC in the patient serum/plasma have been correlated with slower disease progression 82, 83, 84. Fc-mediated effector functions also played an important role in controlling HIV-1 infection in elite controllers 85. Furthermore, ADCC was identified as an immune correlate of the modest protection observed in the RV144 vaccine trial 86, 87. Studies using passive immunization with bnAbs, and non-neutralizing antibodies in animal models, have provided valuable insights into the contribution of Fc-mediated effector functions to protective efficacy 88, 89, 90. While some experiments suggest a significant role for these functions, others report conflicting results. For instance, in non-human primate models, the protective effect of certain bnAbs was partially abrogated when Fc-receptor binding was eliminated, suggesting the involvement of Fc-mediated effector functions 91, 92. However, in other studies 73, 93, 94, the absence of evidence for a contribution of Fc-mediated effector function challenges this notion, raising questions about the variability in bnAb efficacy across different viral infection models.

 

Recent advances in mathematical modeling and HIV-1 dynamics have enabled quantification of the relative contribution of Fc-mediated effector functions to the antiviral activity of bnAbs, particularly in contexts where established infections need to be treated 84, 93. These models suggest that Fc-mediated effector functions can account for a substantial portion (around 20-45%) of bnAb activity. On the other hand, investigations into engineered bnAb variants with enhanced FcγR binding aimed at boosting ADCC activity have brought about mixed results 93, 95, calling attention to the complex role of effector functions in modulating therapeutic outcomes.

 

Therefore, having an understanding of the interplay between Fab-mediated neutralization and Fc-mediated effector functions, as well as the factors influencing their effectiveness, is crucial for optimizing antibody-based strategies for HIV-1 prevention and therapeutic intervention. One of the ways by which Fc-mediated effector functions confer protection against viral infections, thereby contributing to overall efficacy is by targeting infected cells for elimination as seen in a recent clinical trial 96. Cells infected with HIV-1 exhibit increased vulnerability to elimination compared to cell-free HIV-1 virions through ADCC and ADCP, owing to the diminished abundance of envelope spikes on the surface of these virions 97. The presence of fewer spikes makes cell-free HIV-1 virions less favorable targets for immune-mediated clearance, even in the presence of complement 98. Further research into the differential abilities of bnAbs to recruit effector functions in vivo will be essential for advancing antibody-based HIV-1 therapies.

CHALLENGES OF ELICITING bnAbs

HIV-1-specific bnAbs have been evaluated in various populations and clinical scenarios 99. Evidence that bnAbs play a key role in controlling new and pre-existing HIV-1 infections supports the idea that a vaccine capable of eliciting bnAbs could provide the immune system with the head start necessary to prevent HIV-1 infection 22. Despite recent advances in vaccine design, such as soluble envelope trimer engineering, eliciting bnAbs against HIV-1 remains particularly challenging 34 given that most targets displayed on the viral envelope spike require prolonged antibody maturation pathways to be generated and effective 100. Besides extensive somatic hypermutation, bnAbs feature other unusual characteristics such as long complementary determining regions (CDRs), polyreactivity, and a high prevalence of rare precursor genes 101. To address these challenges, novel strategies such as targeting germline precursors of bnAbs have been implemented for HIV-1 vaccine design 102, 103. A recent phase 1 clinical trial with a germline-targeting priming vaccine candidate (IAVI G001) reported the presence of VRC01-class bnAb precursors in 97% of the vaccine recipients 104. The key to the apparent success appears to be the targeting of germinal B-cells with a nanoparticle carrying 60 copies of an engineered antigen (eOD-GT8 60mer) resembling the inferred immunogenic precursor target of VRC01 recognized by immature B cells, and a subsequent inoculation with a more refined version of the antigen of interest. This guided immune response resulted in the production both VRC01-class B-cells precursors and memory B cells 104. These encouraging results establish a clinical proof of concept for germline-targeting vaccine priming and support the development of boosting regimens to further enhance bnAb maturation. Currently, two clinical trials (NCT05001373 and NCT05414786) are being conducted to assess the safety and immunogenicity of mRNA-based vaccines based on the eOD-GT8 60mer 105.

HIV-1 bnAbs FOR TREATMENT

Antiretroviral therapy (ART) has effectively lowered HIV-1-associated morbidity and mortality rates by suppressing viral replication 106 and demonstrated remarkable efficacy in prevention 107. However, ART does not eradicate established HIV-1 infections, thus highlighting the need for a life-long treatment to prevent HIV-1 reactivation from long-lived viral reservoirs 108, 109, 110. Most HIV-1-infected individuals who discontinue ART typically exhibit a viral rebound within weeks even including those who initiated ART early during acute infection with long-term suppression of plasma viremia 111. Due to barriers to universal ART uptake including stigma, adverse effects, emerging drug resistance and reluctant to adhere to lifelong ART medication, novel preventive and therapeutic interventions are highly desirable 112, 113. At a minimum, alternative intervention characteristics should include durable viral suppression below viral transmission levels, prevention of disease progression, low susceptibility to drug resistance, and being generally safe and reasonably tolerated 111.

 

To date, HIV-1-specific bnAbs are being thoroughly investigated as potential ART alternatives because they directly target specific viral epitopes and potentially harness host immune responses 114. While first-generation bnAbs were generally safe when administered in the context of ongoing viremia or during ART interruption, they had limited potency and limited antiviral activity after multiple high-dose infusions 115, 116. In contrast, second-generation bnAbs revealed enhanced potency requiring less frequent administrations to maintain serum concentrations at acceptable target levels 117. Multiple potent second-generation bnAbs against HIV-1 have been and are currently being assessed in clinical trials for HIV-1 treatment (Table 2). As of now, single, combination, and repeated administrations of these second-generation bnAbs have been generally well tolerated with rare adverse events 111.


Table 2: Examples of completed and active clinical trials using second-generation HIV-1bnAbs for the treatment of HIV-1.

CLINICAL TRIAL IDENTIFIER

BROADLY NEUTRALIZING ANTIBODIES

ENVELOPE BINDING SITE

COMPLETED

NCT02960581

PGT121

V3 glycan

NCT03205917

PGDM1400

PGT121

VRC07-523LS

V1/V2 loop

V3 glycan

CD4bs

NCT02018510

3BNC117

CD4bs

NCT02446847

3BNC117

CD4bs

NCT02588586

3BNC117

CD4bs

NCT02825797

3BNC117

10-1074

CD4bs

V3 glycan supersite

NCT03526848

3BNC117

10-1074

CD4bs

V3 glycan supersite

NCT02511990

10-1074

V3 loop

NCT03707977

VRC01LS

10-1074

CD4bs

V3 glycan supersite

NCT02664415

VRC01

CD4bs

NCT01950325

VRC01

CD4bs

NCT02840474

VRC01LS

VRC07-523LS

CD4bs

CD4bs

NCT03254277

3BNC117-LS

CD4bs

NCT04250636

3BNC117-LS

10-1074-LS

CD4bs

V3 glycan supersite

NCT03875209

10E8.4

MPER

NCT02579083

VRC01-N

CD4bs

NCT03015181

VRC01-523LS

CD4bs

NCT03837756

3BNC117

10-1074-LS

CD4bs

V3 glycan supersite

NCT03041012

3BNC117

CD4bs

ACTIVE

NCT04871113

GSK3810109A

CD4bs

NCT04319367

10-1074-LS

V3 glycan supersite

NCT05300035

10-1074-LS

V3 glycan supersite

NCT03374202

AAV-VRC07

CD4bs

NCT04357821

VRC07-523LS

10-1074

CD4bs

V3 glycan supersite

NCT04173819

3BNC117-LS-J

10-1074-LS-J

CD4bs

V3 glycan supersite

NCT04983030

PGDM1400

PGT121

VRC07-523LS

V1/V2 loop

V3 glycan

CD4bs

NCT05281510

VRC07-523LS

CAP256V2LS

CD4

V1/V2 loop

NCT05719441

VRC07-523LS

PGT121.414.LS

CD4bs

V3 glycan

Clinical data from prevention trials (Table 2) consistently demonstrate that bnAbs extend viral suppression, regardless of ART. However, their effectiveness depends on virus sensitivity and therapeutic antibody levels. In recent trials, a single infusion of bnAb 3BNC117 (NCT02018510) reduced HIV-1 viremia for up to 28 days, but with the emergence of resistant strains 59. BnAb VRC01 (NCT01950325) showed reduced viremia, though some participants had minimal response due to resistant virus 118. Single doses of bnAb 10-1074 (NCT02511990) and PGT121 (NCT02960581) led to the rapid emergence of resistant viruses, despite their potency 119, 120. In contrast, multiple bnAb doses, as shown in a phase 2a trial (NCT02446847), may prolong suppression, with notable delays in viral rebound compared to historical controls 121. These findings underscore the potential limitations of single-dose bnAb regimens and highlight the importance of exploring multiple dosing strategies to achieve sustained viral suppression in HIV-1-infected individuals.

 

Combining bnAbs targeting various HIV-1 neutralizing epitopes has been trialed to combat resistance. Recent phase 1 trials showed that pairing 3BNC117 and 10-1074 increased viral suppression with fewer escape mutants. One trial (NCT02825797) reported 15 weeks of suppression without mutants after three doses at weeks 0, 3, and 6 122. Another trial with an eight-dose regimen over 28 weeks achieved complete viral suppression for 43 weeks post-ART interruption or 15 weeks post-infusion, without escape mutants 123. However, due to HIV-1 diversity, this combination may only benefit approximately 50% of clade B-infected individuals sensitive to both antibodies 122. Testing a triple bnAb formulation (NCT03205917) consisting of PGDM1400, PGT121, and VRC07-523LS showed promising coverage; however, viral rebound occurred within a median of 20 days post-infusion for two (i.e., PGDM1400 and PGT121) out of the three antibodies 124, indicating the need for further refinement in antibody selection for sustained effectiveness.

 

Potential improvements in antibody selection may involve conducting genotypic and phenotypic analysis of plasma virus samples 125, akin to the Zurich Primary HIV Infection Study 126, to identify candidates responsive to specific bnAbs. Additionally, implementing computational studies capable of predicting viral rebound and delineating HIV-1 escape pathways from other bnAbs, for which therapy trials are lacking, could inform the design of optimal treatments 127, 128.

 

Furthermore, antibody Fc domain engineering methods to significantly improve the half-life of existing bnAbs have been successfully implemented in recent clinical trials 129, 122, 130. The half-life of circulating IgGs is regulated through Fc binding to the neonatal Fc receptors protecting them from lysosomal degradation 131, 132. To increase the half-life of an IgG molecule, a serine and leucine double mutation can be introduced at positions M428L and N434S in the Fc domain. The double mutation not only increases the serum half-life and the persistence in genital tissues but also retains neutralizing antibody levels for longer time133, 134. Such changes in LS-antibodies are expected to be translated into administering bnAbs treatments at 3–6-month intervals, which could be a promising alternative to ART regimens, especially in geographically isolated locations. In summary, although the clinical trials utilizing bnAbs discussed above have shown promise, further fine-tuning in the selection of bnAbs and their treatment schedules is necessary to optimize prevention effectiveness in both ART-treated and untreated individuals.

HIV-1 bnAbs AND VIRAL RESERVOIRS

The HIV-1 viral reservoir consists of latently infected cells such as resting CD4+ T cells and other long-lived cells that harbor integrated HIV-1 DNA within their genome, allowing the virus to persist in the body for extended periods 135. The presence of these reservoirs is a critical obstacle to viral eradication. The reservoirs are usually established within the first few days of infection and enable viral rebound after ART interruption despite generally efficient suppression of viral replication by ART 136. However, early ART initiation during acute infection may limit the size and diversity of HIV-1 reservoirs compared to late ART initiation during chronic infection 112, 136, 137.

 

In the search for an eradication strategy, various approaches for reducing and eliminating viral reservoirs have been investigated, including, but not limited, to the “shock and kill” strategy, which aims to reactivate dormant infected cells for immune clearance 138, 139; gene editing techniques like CRISPR/Cas9 to directly target and disrupt viral DNA 140, 141; immune-based therapies such as therapeutic vaccines and immunomodulatory agents to enhance immune response 142, 143, 144; development of novel antiretroviral drugs with enhanced potency 145, 146; and exploration of combination approaches integrating multiple strategies for more effective reservoir targeting 147, 148. An antibody-based strategy for eliminating viral reservoirs, employing bnAbs to target and eradicate HIV-infected cells, has also been explored 149. While earlier stage 1 clinical trials showed limited success 118, 150,151 recent preclinical studies and early clinical trials have demonstrated the potential to diminish viral reservoir and delay viral rebound 96, 152, 153 . For example, a recent phase 1b/2a study (NCT03041012) assessed whether early intervention with bnAb 3BNC117 followed by a histone deacetylase inhibitor (romidepsin) shortly after ART initiation could alter the course of HIV-1 infection. The authors found that early intervention with 3BNC117 at ART initiation enhanced elimination of plasma viruses and infected cells, boosted HIV-1-specific CD8+ immunity, and was associated with sustained ART-free virologic control among individuals with 3BNC117-sensitive virus 152. These findings lend evidence to interventions administered at the time of ART initiation as a strategy to limit HIV-1 persistence. Another recent phase 1b study examined the impact of multiple infusions of 3BNC117 and 10-1074 in individuals chronically infected with HIV on sustained viral suppression without ART and reservoir size when administered independently 96. Analysis of the reservoir after six months of therapy indicated alterations in the size and makeup of the intact proviral reservoir with no observed reduction in the defective reservoir.

 

Understanding the differences in viral reservoirs and susceptibility to bnAbs between individuals starting ART early versus those with chronic infection is essential for designing clinical trials and implementing HIV-1 cure strategies focused on reducing viral reservoirs 136. For example, children living with HIV-1 and receiving ART from birth are particularly suitable for bnAb treatment due to their limited viral resistance and smaller viral reservoir size 154 . Moreover, they offer an opportunity for ART-sparing strategies, avoiding long-term toxicities and adherence issues. A bnAb treatment study in Botswana (NCT03707977) achieved sustained HIV-1 suppression in children with favorable reservoir characteristics, including a smaller initial proviral reservoir and sensitivity to bnAb neutralization 129 . Despite these promising results, challenges remain, such as understanding the long-term effects of antibody infusions in children, the establishment of long-lived viral reservoirs, and their susceptibility to bnAbs. To tackle the latter, various combinations of bnAbs with potent latency-reversing agents or other immunomodulatory approaches, such as toll-like receptor agonists, cytokines, checkpoint blockade inhibitors, or therapeutic vaccines, are currently under investigation 99.

 

Although promising, there are lingering questions about the effectiveness and accessibility of bnAbs in tissues compared to the bloodstream, as well as concerns about viral escape mutations and the selection pressures influencing viral rebound in the presence of non-suppressive bnAb therapy. Nonetheless, therapies based on bnAbs offer a promising avenue for researching an HIV-1 cure, and are central to further exploration in current studies.

ROLE OF bnAbs FOR PREVENTION

Whether bnAbs can be used to prevent HIV-1 acquisition has been recently assessed in two randomized phase 2b efficacy clinical trials conducted in the Americas and Europe (HVTN 704/HPTN 085; NCT02716675) as well as in sub-Saharan Africa (HVTN 703/HPTN 081, NCT02568215) 155. Both proof of concept studies used the CD4bs-specific antibody VRC01 administered either at a low dose of 10 mg/kg or at a high dose of 30 mg/kg. Both trials showed that VRC01 was not able to prevent HIV-1 acquisition more efficiently than placebo. Out of 2699 participants in HVTN 704/HPTN 085, 32 infections occurred in the low dose group, 28 infections in the high dose group, and 38 infections in the placebo group. The estimated prevention efficacy of the pooled groups versus placebo was 26.5%.

 

Among the 1924 participants in HVTN 703/HPTN 081, 28 HIV-1 infections occurred in the low-dose group, 19 in the high-dose group, and 29 in the placebo group. Similarly, the estimated prevention efficacy was non-significant at 8.8%. Although VRC01 did not prevent overall HIV-1 acquisition more effectively than placebo a pooled data analysis looking at the incidence of infection with VRC01-sensitive isolates revealed an estimated prevention efficacy of over 75% suggesting that bnAb prophylaxis could be effective. Recently, a more potent and half-life enhanced bnAb (VRC07-523LS) was also tested in a phase 1 study (NCT03015181) and deemed safe for clinical use 130. It will be of interest to see whether this antibody reveals a better prevention efficacy compared to the previous trials.

 

More clinical studies are beginning to focus on bnAb combination regimens as an alternative to single regimens like VRC01 (Table 3). Currently, a double regimen including the two bnAbs CAP256V2LS and VRC07-523LS was tested for safety and pharmacokinetics. The phase 1 clinical trial conducted in South Africa with 42 HIV-negative participants (CAPRISA 012B) reported no serious adverse events or dose-limiting toxicities 117. Another phase 1 clinical trial is looking at the safety and pharmacokinetics of the bnAb combination 3BNC117 and 10-1074 in 25 HIV-1 negative participants for prophylactic use against HIV-1 (NCT02824536) 156.


Table 3: Examples of completed and active clinical trials using second-generation HIV-1bnAbs for the prevention of HIV-1.

CLINICAL TRIAL IDENTIFIER

BROADLY NEUTRALIZING ANTIBODIES 

ENVELOPE BINDING SITE 

COMPLETED

NCT02568215

VRC01

CD4bs

NCT02716675

VRC01

CD4bs

NCT02824536

3BNC117

10-1074

CD4bs,

V3 glycan supersite

ACTIVE

NCT04408963

CAP256V2LS

V1/V2 Loops

Aside from infusions, a recent phase 1 clinical study (NCT02579083) described the use of MB66, a multipurpose prevention technology vaginal film product, in combination with bnAb VRC01-N against HIV-1 and HSV8-N against HSV-1 and 2 157 . MB66 was well tolerated when administered over seven consecutive days. The study observed peak antibody levels in vaginal secretions one-hour post-film administration, which remained significantly elevated through 24 hours. Vaginal samples collected after 24 hours significantly neutralized both HIV-1 and HSV-2 ex vivo.

CONCLUSION

The administration of HIV-1 envelope-specific bnAbs in humans has shown both safety and efficacy in reducing viremia. Moreover, bnAbs maintain short-term viral suppression of antibody-sensitive HIV-1 variants in the absence of ART. While both single-dose and multi-dose regimens are viable, multi-dose regimens have demonstrated prolonged HIV-1 suppression. Notably, individuals infected with HIV-1 who exhibit high sensitivity to specific bnAbs experience superior and longer-lasting viral suppression compared to those with lower sensitivity. Although combinatorial therapy with multiple bnAbs has partially addressed sensitivity issues, genotyping and phenotyping of locally circulating viruses will be essential for optimizing therapy outcomes. Enhanced bnAb combination therapies with extended intervals between treatments could offer patients greater flexibility without the need for daily oral medications like ART.

 

In the context of passive HIV-1 vaccination, HIV-1 envelope-specific bnAbs have been tested in high-risk uninfected populations, yielding limited protection. Initial clinical trials using a single bnAb regimen, such as VRC01, failed to achieve statistically significant protection compared to unvaccinated controls. Therefore, further studies employing combinatorial antibody regimens and improved antibody candidates, particularly those with extended half-lives, are necessary to explore the prophylactic potential of HIV-1-specific bnAbs, especially in high-risk groups such as men and transgender individuals who have sex with men.

 

The persistence of viral reservoirs poses a challenge to HIV eradication efforts despite effective ART. Various strategies are being explored, including antibody-based approaches using bnAbs to target and eliminate HIV-infected cells with recent studies showing promise in reducing the viral reservoir and delaying viral rebound. However, challenges remain in understanding differences in viral reservoirs and bnAb susceptibility, tissue accessibility, viral escape mutations, and rebound under non-suppressive bnAb therapy. Despite these challenges, bnAb-based therapies offer hope for significant advances towards eliminating HIV-1 infections.

 

References

  1. Binley J M, Wrin T, Korber B, Zwick M B, Wang M, Chappey C, Stiegler G, Kunert R, Zolla-Pazner S, Katinger H, Petropoulos C J, Burton D R (2004). Comprehensive cross-clade neutralization analysis of a panel of anti-human immunodeficiency virus type 1 monoclonal antibodies. J Virol 78(23): 1323213252. doi:10.1128/JVI.78.23.13232-13252.2004
  2. Huang J, Kang B H, Pancera M, Lee J H, Tong T, Feng Y, Imamichi H, Georgiev I S, Chuang G Y, Druz A, Doria-Rose N A, Laub L, Sliepen K, Van Gils M J, De La Peña A T, Derking R, Klasse P J, Migueles S A, Bailer R T, Alam M, Pugach P, Haynes B F, Wyatt R T, Sanders R W, Binley J M, Ward A B, Mascola J R, Kwong P D, Connors M (2014). Broad and potent HIV-1 neutralization by a human antibody that binds the gp41-gp120 interface. Nature 515(7525): 138142. doi: 10.1038/nature13601
  3. Scharf L, Wang H, Gao H, Chen S, Mcdowall A W, Bjorkman P J (2015). Broadly Neutralizing Antibody 8ANC195 Recognizes Closed and Open States of HIV-1 Env. Cell 162(6): 13791390. doi:10.1016/j.cell.2015.08.035
  4. Williams L D, Ofek G, Schätzle S, Mcdaniel J R, Lu X, Nicely N I, Wu L, Lougheed C S, Bradley T, Louder M K, Mckee K, Bailer R T, O’Dell S, Georgiev I S, Seaman M S, Parks R J, Marshall D J, Anasti K, Yang G, Nie X, Tumba N L, Wiehe K, Wagh K, Korber B, Kepler T B, Alam S Munir, Morris L, Kamanga G, Cohen M S, Bonsignori M, et al.  (2017). Potent and broad HIV-neutralizing antibodies in memory B cells and plasma. Sci Immunol 2(7): 2200. doi: 10.1126/sciimmunol.aal2200
  5. Walker L M, Huber M, Doores K J, Falkowska E, Pejchal R, Julien J P, Wang S K, Ramos A, Chan-Hui P Y, Moyle M, Mitcham J L, Hammond P W, Olsen O A, Phung P, Fling S, Wong C H, Phogat S, Wrin T, Simek M D, Koff W C, Wilson I A, Burton D R, Poignard P, Investigators PGP (2011). Broad neutralization coverage of HIV by multiple highly potent antibodies. Nature 477(7365): 466470. doi: 10.1038/nature10373
  6. Van Gils M J, Van Den Kerkhof T L, Ozorowski G, Cottrell C A, Sok D, Pauthner M, Pallesen J, De Val N, Yasmeen A, De Taeye S W, Schorcht A, Gumbs S, Johanna I, Saye-Francisco K, Liang C H, Landais E, Nie X, Pritchard L K, Crispin M, Kelsoe G, Wilson I A, Schuitemaker H, Klasse P J, Moore J P, Burton D R, Ward A B, Sanders R W (2016). An HIV-1 antibody from an elite neutralizer implicates the fusion peptide as a site of vulnerability. Nat Microbiol 2: 16199. doi:10.1038/nmicrobiol.2016.199
  7. Kong R, Xu K, Zhou T, Acharya P, Lemmin T, Liu K, Ozorowski G, Soto C, Taft J D, Bailer R T, Cale E M, Chen L, Choi C W, Chuang G Y, Doria-Rose N A, Druz A, Georgiev I S, Gorman J, Huang J, Joyce M G, Louder M K, Ma X, Mckee K, O’Dell S, Pancera M, Yang Y, Blanchard S C, Mothes W, Burton D R, Koff W C, et al. (2016). Fusion peptide of HIV-1 as a site of vulnerability to neutralizing antibody. Science 352(6287): 828833. doi: 10.1126/science.aae0474
  8. Schoofs T, Barnes C O, Suh-Toma N, Golijanin J, Schommers P, Gruell H, West A P, Bach F, Lee Y E, Nogueira L, Georgiev I S, Bailer R T, Czartoski J, Mascola J R, Seaman M S, Mcelrath M J, Doria-Rose N A, Klein F, Nussenzweig M C, Bjorkman P J (2019). Broad and Potent Neutralizing Antibodies Recognize the Silent Face of the HIV Envelope. Immunity 50(6): 15131529. doi: 10.1016/j.immuni.2019.04.014
  9. Zhou T, Zheng A, Baxa U, Chuang G Y, Georgiev I S, Kong R, O’Dell S, Shahzad-Ul-Hussan S, Shen C H, Tsybovsky Y, Bailer R T, Gift S K, Louder M K, Mckee K, Rawi R, Stevenson C H, Stewart-Jones GBE, Taft J D, Waltari E, Yang Y, Zhang B, Shivatare S S, Shivatare V S, Lee C D, Wu C Y, Mullikin J C, Bewley C A, Burton D R, Polonis V R, Shapiro L, et al. (2018). A Neutralizing Antibody Recognizing Primarily N-Linked Glycan Targets the Silent Face of the HIV Envelope. Immunity 48(3): 500513. doi: 10.1016/j.immuni.2018.02.013
  10. Van Schooten J, Farokhi E, Schorcht A, Van Den Kerkhof TLGM, Gao H, Van Der Woude P, Burger J A, Meesters TGR, Bijl T, Ghalaiyini R, Turner H L, Dorning J, Van Schaik BDC, Van Kampen AHC, Labranche C C, Stanfield R L, Sok D, Montefiori D C, Burton D R, Seaman M S, Ozorowski G, Wilson I A, Sanders R W, Ward A B, Van Gils M J (2022). Identification of IOMA-class neutralizing antibodies targeting the CD4-binding site on the HIV-1 envelope glycoprotein. Nat Commun 13(1): 4515. doi: 10.1038/s41467-022-32208-0
  11. Sajadi M M, Dashti A, Tehrani Z Rikhtegaran, Tolbert W D, Seaman M S, Ouyang X, Gohain N, Pazgier M, Kim D, Cavet G, Yared J, Redfield R R, Lewis G K, Devico A (2018). Identification of Near-Pan-neutralizing Antibodies against HIV-1 by Deconvolution of Plasma Humoral Responses. Cell 173(7): 17831795. doi: 10.1016/j.cell.2018.03.061
  12. Falkowska E, Ramos A, Feng Y, Zhou T, Moquin S, Walker L M, Wu X, Seaman M S, Wrin T, Kwong P D, Wyatt R T, Mascola J R, Poignard P, Burton D R (2012). PGV04, an HIV-1 gp120 CD4 binding site antibody, is broad and potent in neutralization but does not induce conformational changes characteristic of CD4. J Virol 86(8): 43944403. doi: 10.1128/JVI.06973-11
  13. Bonsignori M, Zhou T, Sheng Z, Chen L, Gao F, Joyce M G, Ozorowski G, Chuang G Y, Schramm C A, Wiehe K, Alam S M, Bradley T, Gladden M A, Hwang K K, Iyengar S, Kumar A, Lu X, Luo K, Mangiapani M C, Parks R J, Song H, Acharya P, Bailer R T, Cao A, Druz, A, Georgiev I S, Kwon Y D, Louder M K, Zhang B, Zheng A, et al. (2016). Maturation Pathway from Germline to Broad HIV-1 Neutralizer of a CD4-Mimic Antibody. Cell 165(2): 449463. doi: 10.1016/j.cell.2016.02.022
  14. Zhou T, Lynch R M, Chen L, Acharya P, Wu X, Doria-Rose N A, Joyce M G, Lingwood D, Soto C, Bailer R T, Ernandes M J, Kong R, Longo N S, Louder M K, Mckee K, O’Dell S, Schmidt S D, Tran L, Yang Z, Druz A, Luongo T S, Moquin S, Srivatsan S, Yang Y, Zhang B, Zheng A, Pancera M, Kirys T, Georgiev I S, Gindin T, et al. (2015). Structural Repertoire of HIV-1-Neutralizing Antibodies Targeting the CD4 Supersite in 14 Donors. Cell 161(6): 12801292. doi: 10.1016/j.cell.2015.05.007
  15. Caskey M, Klein F, Lorenzi J C, Seaman M S, West A P, Buckley N, Kremer G, Nogueira L, Braunschweig M, Scheid J F, Horwitz J A, Shimeliovich I, Ben-Avraham S, Witmer-Pack M, Platten M, Lehmann C, Burke L A, Hawthorne T, Gorelick R J, Walker B D, Keler T, Gulick R M, Fatkenheuer G, Schlesinger S J, Nussenzweig M C (2016). Corrigendum: Viraemia suppressed in HIV-1-infected humans by broadly neutralizing antibody 3BNC117. Nature 535(7613): 580. doi: 10.1126/science.1187659
  16. Wu X, Yang Z Y, Li Y, Hogerkorp C M, Schief W R, Seaman M S, Zhou T, Schmidt S D, Wu L, Xu L, Longo N S, Mckee K, O’Dell S, Louder M K, Wycuff D L, Feng Y, Nason M, Doria-Rose N, Connors M, Kwong P D, Roederer M, Wyatt R T, Nabel G J, Mascola J R (2010). Rational design of envelope identifies broadly neutralizing human monoclonal antibodies to HIV-1. Science 329(5993): 856861. doi: 10.1126/science.1187659
  17. Zwick M B, Parren P W, Saphire E O, Church S, Wang M, Scott J K, Dawson P E, Wilson I A, Burton D R (2003). Molecular features of the broadly neutralizing immunoglobulin G1 b12 required for recognition of human immunodeficiency virus type 1 gp120. J Virol 77(10): 58635876. doi: 10.1128/jvi.77.10.5863-5876.2003
  18. Bonsignori M, Kreider E F, Fera D, Meyerhoff R R, Bradley T, Wiehe K, Alam S M, Aussedat B, Walkowicz W E, Hwang K K, Saunders K O, Zhang R, Gladden M A, Monroe A, Kumar A, Xia S M, Cooper M, Louder M K, Mckee K, Bailer R T, Pier B W, Jette C A, Kelsoe G, Williams W B,mMorris L, Kappes J, Wagh K, Kamanga G, Cohen M S, Hraber P T, et al. (2017). Staged induction of HIV-1 glycan-dependent broadly neutralizing antibodies. Sci Transl Med 9(381): 7514. doi: 10.1126/scitranslmed.aai7514
  19. Freund N T, Wang H, Scharf L, Nogueira L, Horwitz J A, Bar-On Y, Golijanin J, Sievers S A, Sok D, Cai H, Lorenzi J C Cesar, Halper-Stromberg A, Toth I, Piechocka-Trocha A, Gristick H B, Van Gils M J, Sanders R W, Wang L X, Seaman M S, Burton D R, Gazumyan A, Walker B D, West A P, Bjorkman P J, Nussenzweig M C (2017). Coexistence of potent HIV-1 broadly neutralizing antibodies and antibody-sensitive viruses in a viremic controller. Sci Transl Med 9(373): 2144. doi: 10.1126/scitranslmed.aal2144
  20. Macleod D T, Choi N M, Briney B, Garces F, Ver L S, Landais E, Murrell B, Wrin T, Kilembe W, Liang C H, Ramos A, Bian C B, Wickramasinghe L, Kong L, Eren K, Wu C Y, Wong C H, Kosakovsky Pond S L, Wilson I A, Burton D R, Poignard P, Network IPCITIAHR (2016). Early Antibody Lineage Diversification and Independent Limb Maturation Lead to Broad HIV-1 Neutralization Targeting the Env High-Mannose Patch. Immunity 44(5): 12151226. doi: 10.1016/j.immuni.2016.04.016
  21. Mouquet H, Scharf L, Euler Z, Liu Y, Eden C, Scheid J F, Halper-Stromberg A, Gnanapragasam P N, Spencer D I, Seaman M S, Schuitemaker H, Feizi T, Nussenzweig M C, Bjorkman P J (2012). Complex-type N-glycan recognition by potent broadly neutralizing HIV antibodies. Proc Natl Acad Sci U S A 109(47): 32683277. doi: 10.1073/pnas.1217207109
  22. Gach J S, Furtmüller P G, Quendler H, Messner P, Wagner R, Katinger H, Kunert R (2010). Proline is not uniquely capable of providing the pivot point for domain swapping in 2G12, a broadly neutralizing antibody against HIV-1. J Biol Chem 285(2): 11221127. doi: 10.1074/jbc.M109.058792
  23. Landais E, Murrell B, Briney B, Murrell S, Rantalainen K, Berndsen Z T, Ramos A, Wickramasinghe L, Smith M L, Eren K, De Val N, Wu M, Cappelletti A, Umotoy J, Lie Y, Wrin T, Algate P, Chan-Hui P Y, Karita E, Ward A B, Wilson I A, Burton D R, Smith D, Pond SLK, Poignard P, Investigators IPC, Network IAHR (2017). HIV Envelope Glycoform Heterogeneity and Localized Diversity Govern the Initiation and Maturation of a V2 Apex Broadly Neutralizing Antibody Lineage. Immunity 47(5): 9901003. doi: 10.1016/j.immuni.2017.11.002
  24. Cale E M, Gorman J, Radakovich N A, Crooks E T, Osawa K, Tong T, Li J, Nagarajan R, Ozorowski G, Ambrozak D R, Asokan M, Bailer R T, Bennici A K, Chen X, Doria-Rose N A, Druz A, Feng Y, Joyce M G, Louder M K, O’Dell S, Oliver C, Pancera M, Connors M, Hope T J, Kepler T B, Wyatt R T, Ward A B, Georgiev I S, Kwong P D, Mascola J R, et al. (2017). Virus-like Particles Identify an HIV V1V2 Apex-Binding Neutralizing Antibody that Lacks a Protruding Loop. Immunity 46(5): 777791. doi: 10.1016/j.immuni.2017.04.011
  25. Doria-Rose N A, Schramm C A, Gorman J, Moore P L, Bhiman J N, Dekosky B J, Ernandes M J, Georgiev I S, Kim H J, Pancera M, Staupe R P, Altae-Tran H R, Bailer R T, Crooks E T, Cupo A, Druz A, Garrett N J, Hoi K H, Kong R, Louder M K, Longo N S, Mckee K, Nonyane M, O’Dell S, Roark R S, Rudicell R S, Schmidt S D, Sheward D J, Soto C, Wibmer C K, et al. (2014). Developmental pathway for potent V1V2-directed HIV-neutralizing antibodies. Nature 509(7498): 5562. doi: 10.1038/nature13036
  26. Sok D, Van Gils M J, Pauthner M, Julien J P, Saye-Francisco K L, Hsueh J, Briney B, Lee J H, Le K M, Lee P S, Hua Y, Seaman M S, Moore J P, Ward A B, Wilson I A, Sanders R W, Burton D R (2014). Recombinant HIV envelope trimer selects for quaternary-dependent antibodies targeting the trimer apex. Proc Natl Acad Sci U S A 111(49): 1762417629. doi: 10.1073/pnas.1415789111
  27. Mclellan J S, Pancera M, Carrico C, Gorman J, Julien J P, Khayat R, Louder R, Pejchal R, Sastry M, Dai K, O’Dell S, Patel N, Shahzad-Ul-Hussan S, Yang Y, Zhang B, Zhou T, Zhu J, Boyington J C, Chuang G Y, Diwanji D, Georgiev I, Kwon Y D, Lee D, Louder M K, Moquin S, Schmidt S D, Yang Z Y, Bonsignori M, Crump J A, Kapiga S H, et al. (2011). Structure of HIV-1 gp120 V1/V2 domain with broadly neutralizing antibody PG9. Natur 480: 336343. doi: 10.1056/NEJMoa2031738
  28. Pejchal R, Walker L M, Stanfield R L, Phogat S K, Koff W C, Poignard P, Burton D R, Wilson I A (2010). Structure and function of broadly reactive antibody PG16 reveal an H3 subdomain that mediates potent neutralization of HIV-1. Proc Natl Acad Sci U S A 107(25): 1148311488. doi: 10.1073/pnas.1004600107
  29. Bonsignori M, Hwang K K, Chen X, Tsao C Y, Morris L, Gray E, Marshall D J, Crump J A, Kapiga S H, Sam N E, Sinangil F, Pancera M, Yongping Y, Zhang B, Zhu J, Kwong P D, O’Dell S, Mascola J R, Wu L, Nabel G J, Phogat S, Seaman M S, Whitesides J F, Moody M A, Kelsoe G, Yang X, Sodroski J, Shaw G M, Montefiori D C, Kepler T B, et al. (2011). Analysis of a clonal lineage of HIV-1 envelope V2/V3 conformational epitope-specific broadly neutralizing antibodies and their inferred unmutated common ancestors. J Virol 85: 999810009. doi: 10.1128/JVI.05045-11
  30. Twigg H L (2005). Humoral immune defense (antibodies): recent advances. Proc Am Thorac Soc 2(5): 417421. doi: 10.1513/pats.200508-089JS
  31. Burton D R (2002). Antibodies, viruses and vaccines. Nat Rev Immunol 2(9): 706713. doi: 10.1038/nri891
  32. De Taeye S W, Rispens T, Vidarsson G (2019). The Ligands for Human IgG and Their Effector Functions. Antibodies (Basel) 8(2): 30. doi: 10.3390/antib8020030
  33. Walsh S R, Seaman M S (2021). Broadly Neutralizing Antibodies for HIV-1 Prevention. Front Immunol 12: 712122. doi: 10.3389/fimmu.2021.712122
  34. Del Prete G Q, Lifson J D, Keele B F (2016). Nonhuman primate models for the evaluation of HIV-1 preventive vaccine strategies: model parameter considerations and consequences. Curr Opin HIV AIDS 11(6): 546554. doi: 10.1097/COH.0000000000000311
  35. Gach J S, Gorlani A, Dotsey E Y, Becerra J C, Anderson C T, Berzins B, Felgner P L, Forthal D N, Deeks S G, Wilkin T J, Casazza J P, Koup R A, Katlama C, Autran B, Murphy R L, Achenbach C J (2016). HIV-1-Specific Antibody Response and Function after DNA Prime and Recombinant Adenovirus 5 Boost HIV Vaccine in HIV-Infected Subjects. PLoS One 11(8): 160341. doi: 10.1371/journal.pone.0160341
  36. Beltran-Pavez C, Ferreira C B, Merino-Mansilla A, Fabra-Garcia A, Casadella M, Noguera-Julian M, Paredes R, Olvera A, Haro I, Brander C, Garcia F, Gatell J M, Yuste E, Sanchez-Merino V (2018). Guiding the humoral response against HIV-1 toward a MPER adjacent region by immunization with a VLP-formulated antibody-selected envelope variant. PLoS One 13(12): 208345. doi: 10.1371/journal.pone.0208345
  37. Galimidi R P, Klein J S, Politzer M S, Bai S, Seaman M S, Nussenzweig M C, West A P, Bjorkman P J (2015). Intra-spike crosslinking overcomes antibody evasion by HIV-1. Cell 160(3): 433446. doi: 10.1016/j.cell.2015.01.016
  38. Moyo T, Ferreira R C, Davids R, Sonday Z, Moore P L, Travers S A, Wood N T, Dorfman J R (2017). Chinks in the armor of the HIV-1 Envelope glycan shield: Implications for immune escape from anti-glycan broadly neutralizing antibodies. Virology 501: 1224. doi: 10.1016/j.virol.2016.10.026
  39. Crispin M, Doores K J (2015). Targeting host-derived glycans on enveloped viruses for antibody-based vaccine design. Curr Opin Virol 11: 6369. doi: 10.1016/j.coviro.2015.02.002
  40. Amitai A, Chakraborty A K, Kardar M (2018). The low spike density of HIV may have evolved because of the effects of T helper cell depletion on affinity maturation. PLoS Comput Biol 14(8): 1006408. doi: 10.1371/journal.pcbi.1006408
  41. Zhu P, Liu J, Bess J, Chertova E, Lifson J D, Grisé H, Ofek G A, Taylor K A, Roux K H (2006). Distribution and three-dimensional structure of AIDS virus envelope spikes. Nature 441(7095): 847852. doi: 10.1038/nature04817
  42. Stano A, Leaman D P, Kim A S, Zhang L, Autin L, Ingale J, Gift S K, Truong J, Wyatt R T, Olson A J, Zwick M B (2017). Dense Array of Spikes on HIV-1 Virion Particles. J Virol 91(14): 415432. doi: 10.1128/JVI.00415-17
  43. Schiller J, Chackerian B (2014). Why HIV virions have low numbers of envelope spikes: implications for vaccine development. PLoS Pathog 10(8): 1004254. doi: 10.1371/journal.ppat.1004254
  44. Ota T, Doyle-Cooper C, Cooper A B, Huber M, Falkowska E, Doores K J, Hangartner L, Le K, Sok D, Jardine J, Lifson J, Wu X, Mascola J R, Poignard P, Binley J M, Chakrabarti B K, Schief W R, Wyatt R T, Burton D R, Nemazee D (2012). Anti-HIV B Cell lines as candidate vaccine biosensors. J Immunol 189(10): 48164824. doi: 10.4049/jimmunol.1202165
  45. Klein J S, Bjorkman P J (2010). Few and far between: how HIV may be evading antibody avidity. PLoS Pathog 6(5): 1000908. doi: 10.1371/journal.ppat.1000908
  46. Burton D R (2023). Antiviral neutralizing antibodies: from in vitro to in vivo activity. Nat Rev Immunol 23(11): 720734. doi: 10.1038/s41577-023-00858-w
  47. Burton D R, Hangartner L (2016). Broadly Neutralizing Antibodies to HIV and Their Role in Vaccine Design. Annu Rev Immunol 34: 635659. doi: 10.1146/annurev-immunol-041015-055515
  48. Moore P L, Crooks E T, Porter L, Zhu P, Cayanan C S, Grise H, Corcoran P, Zwick M B, Franti M, Morris L, Roux K H, Burton D R, Binley J M (2006). Nature of nonfunctional envelope proteins on the surface of human immunodeficiency virus type 1. J Virol 80(5): 25152528. doi: 10.1128/JVI.80.5.2515-2528.2006
  49. Cook J D, Khondker A, Lee J E (2022). Conformational plasticity of the HIV-1 gp41 immunodominant region is recognized by multiple non-neutralizing antibodies. Commun Biol 5(1): 291. doi: 10.1128/JVI.80.5.2515-2528.2006
  50. Moore P L, Gray E S, Morris L (2009). Specificity of the autologous neutralizing antibody response. Curr Opin HIV AIDS 4(5): 358363. doi: 10.1128/JVI.80.5.2515-2528.2006
  51. Griffith S A, Mccoy L E (2021). To bnAb or Not to bnAb: Defining Broadly Neutralising Antibodies Against HIV-1. Front Immunol 12: 708227. doi: 10.3389/fimmu.2021.708227
  52. Gao F, Chen Y, Levy D N, Conway J A, Kepler T B, Hui H (2004). Unselected mutations in the human immunodeficiency virus type 1 genome are mostly nonsynonymous and often deleterious. J Virol 78(5): 24262433. doi: 10.1128/jvi.78.5.2426-2433.2004
  53. Bar K J, Tsao C Y, Iyer S S, Decker J M, Yang Y, Bonsignori M, Chen X, Hwang K K, Montefiori D C, Liao H X, Hraber P, Fischer W, Li H, Wang S, Sterrett S, Keele B F, Ganusov V V, Perelson A S, Korber B T, Georgiev I, Mclellan J S, Pavlicek J W, Gao F, Haynes B F, Hahn B H, Kwong P D, Shaw G M (2012). Early low-titer neutralizing antibodies impede HIV-1 replication and select for virus escape. PLoS Pathog 8(5): 1002721. doi: 10.1371/journal.ppat.1002721
  54. Charpentier C, Nora T, Tenaillon O, Clavel F, Hance A J (2006). Extensive recombination among human immunodeficiency virus type 1 quasispecies makes an important contribution to viral diversity in individual patients. J Virol 80(5): 24722482. doi: 10.1128/JVI.80.5.2472-2482.2006
  55. Yu F, Wen Y, Wang J, Gong Y, Feng K, Ye R, Jiang Y, Zhao Q, Pan P, Wu H, Duan S, Su B, Qiu M (2018). The Transmission and Evolution of HIV-1 Quasispecies within One Couple: a Follow-up Study based on Next-Generation Sequencing. Sci Rep 8(1): 1404. doi: 10.1038/s41598-018-19783-3
  56. Alter G, Moody M A (2010). The humoral response to HIV-1: new insights, renewed focus. J Infect Dis 202(2): 315322. doi: 10.1086/655654
  57. Mccoy L E, Mcknight Á (2017). Lessons learned from humoral responses of HIV patients. Curr Opin HIV AIDS 12(3): 195202. doi: 10.1097/COH.0000000000000361
  58. Walker L M, Simek M D, Priddy F, Gach J S, Wagner D, Zwick M B, Phogat S K, Poignard P, Burton D R (2010). A limited number of antibody specificities mediate broad and potent serum neutralization in selected HIV-1 infected individuals. PLoS Pathog 6(8): 1001028. doi: 10.1371/journal.ppat.1001028
  59. Landais E, Moore P L (2018). Development of broadly neutralizing antibodies in HIV-1 infected elite neutralizers. Retrovirology 15(1): 61. doi: 10.1186/s12977-018-0443-0
  60. Conti S, Karplus M (2019). Estimation of the breadth of CD4bs targeting HIV antibodies by molecular modeling and machine learning. PLoS Comput Biol 15(4): 1006954. doi: 10.1371/journal.pcbi.1006954
  61. Scheid J F, Mouquet H, Feldhahn N, Seaman M S, Velinzon K, Pietzsch J, Ott R G, Anthony R M, Zebroski H, Hurley A, Phogat A, Chakrabarti B, Li Y, Connors M, Pereyra F, Walker B D, Wardemann H, Ho D, Wyatt R T, Mascola J R, Ravetch J V, Nussenzweig M C (2009). Broad diversity of neutralizing antibodies isolated from memory B cells in HIV-infected individuals. Nature 458(7238): 636640. doi: 10.1038/nature07930
  62. Derdeyn C A, Moore P L, Morris L (2014). Development of broadly neutralizing antibodies from autologous neutralizing antibody responses in HIV infection. Curr Opin HIV AIDS 9(3): 210216. doi: 10.1097/COH.0000000000000057
  63. De Taeye S W, Moore J P, Sanders R W (2016). HIV-1 Envelope Trimer Design and Immunization Strategies To Induce Broadly Neutralizing Antibodies. Trends Immunol 37(3): 221232. doi: 10.1016/j.it.2016.01.007
  64. Reiss EIMM, Van Haaren M M, Van Schooten J, Claireaux MAF, Maisonnasse P, Antanasijevic A, Allen J D, Bontjer I, Torres J L, Lee W H, Ozorowski G, Vázquez Bernat N, Kaduk M, Aldon Y, Burger J A, Chawla H, Aartse A, Tolazzi M, Gao H, Mundsperger P, Crispin M, Montefiori D C, Karlsson Hedestam C G, Scarlatti G, Ward A B, Le Grand R, Shattock R, Dereuddre-Bosquet N, Sanders R W, Van Gils M J (2022). Fine-mapping the immunodominant antibody epitopes on consensus sequence-based HIV-1 envelope trimer vaccine candidates. NPJ Vaccines 7(1): 152. doi: 10.1038/s41541-022-00576-9
  65. Parker Miller E, Finkelstein M T, Erdman M C, Seth P C, Fera D (2021). A Structural Update of Neutralizing Epitopes on the HIV Envelope, a Moving Target. Viruses 13(9): 1774. doi: 10.3390/v13091774
  66. Ding C, Patel D, Ma Y, Mann Jfs, Wu J, Gao Y (2021). Employing Broadly Neutralizing Antibodies as a Human Immunodeficiency Virus Prophylactic & Therapeutic Application. Front Immunol 12: 697683. doi: 10.3389/fimmu.2021.697683
  67. Gach J S, Leaman D P, Zwick M B (2011). Targeting HIV-1 gp41 in close proximity to the membrane using antibody and other molecules. Curr Top Med Chem 11(24): 29973021. doi: 10.2174/156802611798808505
  68. Lorin V, Fernández I, Masse-Ranson G, Bouvin-Pley M, Molinos-Albert L M, Planchais C, Hieu T, Péhau-Arnaudet G, Hrebík D, Girelli-Zubani G, Fiquet O, Guivel-Benhassine F, Sanders R W, Walker B D, Schwartz O, Scheid J F, Dimitrov J D, Plevka P, Braibant M, Seaman M S, Bontems F, Santo J P Di, Rey F A, Mouquet H (2022). Epitope convergence of broadly HIV-1 neutralizing IgA and IgG antibody lineages in a viremic controller. J Exp Med 219(3): 20212045. doi: 10.1084/jem.20212045
  69. Sacks D, Bhiman J N, Wiehe K, Gorman J, Kwong P D, Morris L, Moore P (2019). Somatic hypermutation to counter a globally rare viral immunotype drove off-track antibodies in the CAP256-VRC26 HIV-1 V2-directed bNAb lineage. PLoS Pathog 15(9): 1008005. doi: 10.1371/journal.ppat.1008005
  70. Walker L M, Phogat S K, Chan-Hui P Y, Wagner D, Phung P, Goss J L, Wrin T, Simek M D, Fling S, Mitcham J L, Lehrman J K, Priddy F H, Olsen O A, Frey S M, Hammond P W, Kaminsky S, Zamb T, Moyle M, Koff W C, Poignard P, Burton D R (2009). Broad and potent neutralizing antibodies from an African donor reveal a new HIV-1 vaccine target. Investigators PGP 326(5950): 285289. doi: 10.1126/science.1178746
  71. Gorny M K, Stamatatos L, Volsky B, Revesz K, Williams C, Wang X H, Cohen S, Staudinger R, Zolla-Pazner S (2005). Identification of a new quaternary neutralizing epitope on human immunodeficiency virus type 1 virus particles. J Virol 79(8): 52325237. doi: 10.1128/JVI.79.8.5232-5237.2005
  72. Sanders R W, Derking R, Cupo A, Julien J P, Yasmeen A, De Val N, Kim H J, Blattner C, De La Peña A T, Korzun J, Golabek M, De Los Reyes K, Ketas T J, Van Gils M J, King C R, Wilson I A, Ward A B, Klasse P J, Moore J P (2013). A next-generation cleaved, soluble HIV-1 Env trimer, BG505 SOSIP.664 gp140, expresses multiple epitopes for broadly neutralizing but not non-neutralizing antibodies. PLoS Pathog 9(9): e1003618. doi: 10.1371/journal.ppat.1003618
  73. Hangartner L, Beauparlant D, Rakasz E, Nedellec R, Hozé N, Mckenney K, Martins M A, Seabright G E, Allen J D, Weiler A M, Friedrich T C, Regoes R R, Crispin M, Burton D R (2021). Effector function does not contribute to protection from virus challenge by a highly potent HIV broadly neutralizing antibody in nonhuman primates. Sci Transl Med 13(585): 3349. doi: 10.1126/scitranslmed.abe3349
  74. Stab V, Stahl-Hennig C, Ensser A, Richel E, Fraedrich K, Sauermann U, Tippler B, Klein F, Burton D R, Tenbusch M, Überla K (2023). HIV-1 neutralizing antibodies provide sterilizing immunity by blocking infection of the first cells. Cell Rep Med 4(10): 101201. doi: 10.1016/j.xcrm.2023.101201
  75. Lewis G K, Pazgier M, Evans D T, Ferrari G, Bournazos S, Parsons M S, Bernard N F, Finzi A (2017). Beyond Viral Neutralization. AIDS Res Hum Retroviruses 33(8): 760764. doi: 10.1089/AID.2016.0299
  76. Su B, Dispinseri S, Iannone V, Zhang T, Wu H, Carapito R, Bahram S, Scarlatti G, Moog C (2019). Update on Fc-Mediated Antibody Functions Against HIV-1 Beyond Neutralization. Front Immunol 10: 2968. doi: 10.3389/fimmu.2019.02968
  77. Danesh A, Ren Y, Jones R Brad (2020). Roles of fragment crystallizable-mediated effector functions in broadly neutralizing antibody activity against HIV. Curr Opin HIV AIDS 15(5): 316323. doi: 10.1097/COH.0000000000000644
  78. Lu L L, Suscovich T J, Fortune S M, Alter G (2018). Beyond binding: antibody effector functions in infectious diseases. Nat Rev Immunol 18(1): 4661. doi: 10.1038/nri.2017.106
  79. Dufloo J, Guivel-Benhassine F, Buchrieser J, Lorin V, Grzelak L, Dupouy E, Mestrallet G, Bourdic K, Lambotte O, Mouquet H, Bruel T, Schwartz O (2020). Anti-HIV-1 antibodies trigger non-lytic complement deposition on infected cells. EMBO Rep 21(2): 49351. doi: 10.15252/embr.201949351
  80. Ugurlar D, Howes S C, De Kreuk B J, Koning R I, De Jong R N, Beurskens F J, Schuurman J, Koster A J, Sharp T H, Parren Pwhi, Gros P (2018). Structures of C1-IgG1 provide insights into how danger pattern recognition activates complement. Science 359(6377): 794797. doi: 10.1126/science.aao4988
  81. Diebolder C A, Beurskens F J, De Jong R N, Koning R I, Strumane K, Lindorfer M A, Voorhorst M, Ugurlar D, Rosati S, Heck A J, Van De Winkel J G, Wilson I A, Koster A J, Taylor R P, Saphire E O, Burton D R, Schuurman J, Gros P, Parren P W (2014). Complement is activated by IgG hexamers assembled at the cell surface. Science 343(6176): 12601263. doi: 10.1126/science.1248943
  82. Wren L H, Chung A W, Isitman G, Kelleher A D, Parsons M S, Amin J, Cooper D A, Stratov I, Navis M, Kent S J, Investigators ASC (2013). Specific antibody-dependent cellular cytotoxicity responses associated with slow progression of HIV infection. Immunology 138(2): 116123. doi: 10.1111/imm.12016
  83. Chung A W, Navis M, Isitman G, Wren L, Silvers J, Amin J, Kent S J, Stratov I (2011). Activation of NK cells by ADCC antibodies and HIV disease progression. J Acquir Immune Defic Syndr 58(2): 127131. doi: 10.1097/QAI.0b013e31822c62b9
  84. Wang P, Gajjar M R, Yu J, Padte N N, Gettie A, Blanchard J L, Russell-Lodrigue K, Liao L E, Perelson A S, Huang Y, Ho D D (2020). Quantifying the contribution of Fc-mediated effector functions to the antiviral activity of anti-HIV-1 IgG1 antibodies in vivo. Proc Natl Acad Sci U S A 117(30): 1800218009. doi: 10.1073/pnas.2008190117
  85. Lambotte O, Ferrari G, Moog C, Yates N L, Liao H X, Parks R J, Hicks C B, Owzar K, Tomaras G D, Montefiori D C, Haynes B F, Delfraissy J F (2009). Heterogeneous neutralizing antibody and antibody-dependent cell cytotoxicity responses in HIV-1 elite controllers. Aids 23(8): 897906. doi: 10.1073/pnas.2008190117
  86. Haynes B F, Gilbert P B, Mcelrath M J, Zolla-Pazner S, Tomaras G D, Alam S M, Evans D T, Montefiori D C, Karnasuta C, Sutthent R, Liao H X, Devico A L, Lewis G K, Williams C, Pinter A, Fong Y, Janes H, Decamp A, Huang Y, Rao M, Billings E, Karasavvas N, Robb M L, Ngauy V, De Souza M S De, Paris R, Ferrari G, Bailer R T, Soderberg K A, Andrews C, et al. (2012). Immune-correlates analysis of an HIV-1 vaccine efficacy trial. N Engl J Med 366(14): 12751286. doi: 10.1056/NEJMoa1113425
  87. Gray G E, Huang Y, Grunenberg N, Laher F, Roux S, Andersen-Nissen E, De Rosa S C, Flach B, Randhawa A K, Jensen R, Swann E M, Bekker L G, Innes C, Lazarus E, Morris L, Mkhize N N, Ferrari G, Montefiori D C, Shen X, Sawant S, Yates N, Hural J, Isaacs A, Phogat S, Diazgranados C A, Lee C, Sinangil F, Michael N L, Robb M L, Kublin J G, et al. (2019). Immune correlates of the Thai RV144 HIV vaccine regimen in South Africa. Sci Transl Med 11(510): 1880. doi: 10.1126/scitranslmed.aax1880
  88. Horwitz J A, Bar-On Y, Lu C L, Fera D, Lockhart Aak, Lorenzi Jcc, Nogueira L, Golijanin J, Scheid J F, Seaman M S, Gazumyan A, Zolla-Pazner S, Nussenzweig M C (2017). Non-neutralizing Antibodies Alter the Course of HIV-1 Infection In Vivo. Cell 170(4): 637648. doi: 10.1126/scitranslmed.aax1880
  89. Bournazos S, Klein F, Pietzsch J, Seaman M S, Nussenzweig M C, Ravetch J V (2014). Broadly neutralizing anti-HIV-1 antibodies require Fc effector functions for in vivo activity. Cell 158(6): 12431253. doi: 10.1016/j.cell.2014.08.023
  90. Halper-Stromberg A, Lu C L, Klein F, Horwitz J A, Bournazos S, Nogueira L, Eisenreich T R, Liu C, Gazumyan A, Schaefer U, Furze R C, Seaman M S, Prinjha R, Tarakhovsky A, Ravetch J V, Nussenzweig M C (2014). Broadly neutralizing antibodies and viral inducers decrease rebound from HIV-1 latent reservoirs in humanized mice. Cell 158(5): 989999. doi: 10.1016/j.cell.2014.07.043
  91. Hessell A J, Hangartner L, Hunter M, Havenith C E, Beurskens F J, Bakker J M, Lanigan C M, Landucci G, Forthal D N, Parren P W, Marx P A, Burton D R (2007). Fc receptor but not complement binding is important in antibody protection against HIV. Nature 449(7158): 101104. doi: 10.1038/nature06106
  92. Hessell A J, Poignard P, Hunter M, Hangartner L, Tehrani D M, Bleeker W K, Parren P W, Marx P A, Burton D R (2009). Effective, low-titer antibody protection against low-dose repeated mucosal SHIV challenge in macaques. Nat Med 15(8): 951954. doi: 10.1038/nm.1974
  93. Asokan M, Dias J, Liu C, Maximova A, Ernste K, Pegu A, Mckee K, Shi W, Chen X, Almasri C, Promsote W, Ambrozak D R, Gama L, Hu J, Douek D C, Todd J P, Lifson J D, Fourati S, Sekaly R P, Crowley A R, Ackerman M E, Ko S H, Kilam D, Boritz E A, Liao L E, Best K, Perelson A S, Mascola J R, Koup R A (2020). Fc-mediated effector function contributes to the in vivo antiviral effect of an HIV neutralizing antibody. Proc Natl Acad Sci U S A 117(31): 1875418763. doi: 10.1073/pnas.2008236117
  94. Parsons M S, Lee W S, Kristensen A B, Amarasena T, Khoury G, Wheatley A K, Reynaldi A, Wines B D, Hogarth P M, Davenport M P, Kent S J (2019). Fc-dependent functions are redundant to efficacy of anti-HIV antibody PGT121 in macaques. J Clin Invest 129(1): 182191. doi: 10.1172/JCI122466
  95. Moldt B, Shibata-Koyama M, Rakasz E G, Schultz N, Kanda Y, Dunlop D C, Finstad S L, Jin C, Landucci G, Alper M D, Dugast A S, Parren P W, Nimmerjahn F, Evans D T, Alter G, Forthal D N, Schmitz J E, Iida S, Poignard P, Watkins D I, Hessell A J, Burton D R (2012). A nonfucosylated variant of the anti-HIV-1 monoclonal antibody b12 has enhanced FcγRIIIa-mediated antiviral activity in vitro but does not improve protection against mucosal SHIV challenge in macaques. J Virol 86(11): 61896196. doi: 10.1128/JVI.00491-12
  96. Gaebler C, Nogueira L, Stoffel E, Oliveira T Y, Breton G, Millard K G, Turroja M, Butler A, Ramos V, Seaman M S, Reeves J D, Petroupoulos C J, Shimeliovich I, Gazumyan A, Jiang C S, Jilg N, Scheid J F, Gandhi R, Walker B D, Sneller M C, Fauci A, Chun T W, Caskey M, Nussenzweig M C (2022). Prolonged viral suppression with anti-HIV-1 antibody therapy. Nature 606(7913): 368374. doi: 10.1038/s41586-022-04597-1
  97. Gach J S, Bouzin M, Wong M P, Chromikova V, Gorlani A, Yu K T, Sharma B, Gratton E, Forthal D N (2017). Human immunodeficiency virus type-1 (HIV-1) evades antibody-dependent phagocytosis. PLoS Pathog 13(12): 1006793. doi: 10.1371/journal.ppat.1006793
  98. Gach J S, Matsuno S Y, Mercado M, Hangartner L, Forthal D N (2022). Internalization of HIV-1 by Phagocytes Is Increased When Virions Are Opsonized with Multimeric Antibody in the Presence of Complement. J Virol 96(2): 168921. doi: 10.1128/JVI.01689-21
  99. Caskey M (2020). Broadly neutralizing antibodies for the treatment and prevention of HIV infection. Curr Opin HIV AIDS 15(1): 4955. doi: 10.1097/COH.0000000000000600
  100. Doria-Rose N A, Joyce M G (2015). Strategies to guide the antibody affinity maturation process. Curr Opin Virol 11: 137147. doi: 10.1016/j.coviro.2015.04.002
  101. Roskin K M, Jackson KIL, Lee J Y, Hoh R A, Joshi S A, Hwang K K, Bonsignori M, Pedroza-Pacheco I, Liao H X, Moody M A, Fire A Z, Borrow P, Haynes B F, Boyd S D (2020). Aberrant B cell repertoire selection associated with HIV neutralizing antibody breadth. Nat Immunol 21(2): 199209. doi: 10.1038/s41590-019-0581-0
  102. Caniels T G, Medina-Ramírez M, Zhang J, Sarkar A, Kumar S, Labranche A, Derking R, Allen J D, Snitselaar J L, Capella-Pujol J, Sánchez IDM, Yasmeen A, Diaz M, Aldon Y, Bijl TPL, Venkatayogi S, Martin Beem J S, Newman A, Jiang C, Lee W H, Pater M, Burger J A, Van Breemen M J, De Taeye S W, Rantalainen K, Labranche C, Saunders K O, Montefiori D, Ozorowski G, Ward A B, et al. (2023). Germline-targeting HIV-1 Env vaccination induces VRC01-class antibodies with rare insertions. Cell Rep Med 4(4): 101003. doi: 10.1016/j.xcrm.2023.101003
  103. Steichen J M, Lin Y C, Havenar-Daughton C, Pecetta S, Ozorowski G, Willis J R, Toy L, Sok D, Liguori A, Kratochvil S, Torres J L, Kalyuzhniy O, Melzi E, Kulp D W, Raemisch S, Hu X, Bernard S M, Georgeson E, Phelps N, Adachi Y, Kubitz M, Landais E, Umotoy J, Robinson A, Briney B, Wilson I A, Burton D R, Ward A B, Crotty S, Batista F D, et al. (2019). A generalized HIV vaccine design strategy for priming of broadly neutralizing antibody responses. Science 366(6470). doi: 10.1126/science.aax4380
  104. Leggat D J, Cohen K W, Willis J R, Fulp W J, Decamp A C, Kalyuzhniy O, Cottrell C A, Menis S, Finak G, Ballweber-Fleming L, Srikanth A, Plyler J R, Schiffner T, Liguori A, Rahaman F, Lombardo A, Philiponis V, Whaley R E, Seese A, Brand J, Ruppel A M, Hoyland W, Yates N L, Williams L D, Greene K, Gao H, Mahoney C R, Corcoran M M, Cagigi A, Taylor A, et al. (2022). Vaccination induces HIV broadly neutralizing antibody precursors in humans. Science 378(6623): 6502. doi: 10.1126/science.aax4380
  105. Matarazzo L, Bettencourt PJG (2023). mRNA vaccines: a new opportunity for malaria, tuberculosis and HIV. Front Immunol 14: 1172691. doi: 10.3389/fimmu.2023.1172691
  106. Zhao Y, Wu Z, Mcgoogan J M, Shi C X, Li A, Dou Z, Ma Y, Qin Q, Brookmeyer R, Detels R, Montaner JSG (2018). Immediate Antiretroviral Therapy Decreases Mortality Among Patients With High CD4 Counts in China: A Nationwide, Retrospective Cohort Study. Clin Infect Dis 66(5): 727734. doi: 10.1093/cid/cix878
  107. Gandhi R T, Bedimo R, Hoy J F, Landovitz R J, Smith D M, Eaton E F, Lehmann C, Springer S A, Sax P E, Thompson M A, Benson C A, Buchbinder S P, Del Rio C, Eron J J, Günthard H F, Molina J M, Jacobsen D M, Saag M S (2023). Antiretroviral Drugs for Treatment and Prevention of HIV Infection in Adults: 2022 Recommendations of the International Antiviral Society-USA Panel. JAMA 329(1): 6384. doi: 10.1001/jama.2022.22246
  108. Denton P W, Søgaard O S, Tolstrup  (1956). Impacts of HIV Cure Interventions on Viral Reservoirs in Tissues. Front Microbiol 10. doi: 10.3389/fmicb.2019.01956
  109. Chun T W, Stuyver L, Mizell S B, Ehler L A, Mican J A, Baseler M, Lloyd A L, Nowak M A, Fauci A S (1997). Presence of an inducible HIV-1 latent reservoir during highly active antiretroviral therapy. Proc Natl Acad Sci U S A 94(24): 1319313197. doi: 10.1073/pnas.94.24.13193
  110. Petitjean G, Al Tabaa Y, Tuaillon E, Mettling C, Baillat V, Reynes J, Segondy M, Vendrell J P (2007). Unintegrated HIV-1 provides an inducible and functional reservoir in untreated and highly active antiretroviral therapy-treated patients. Retrovirology 4: 60. doi: 10.1186/1742-4690-4-60
  111. Hsu D C, Mellors J W, Vasan S (2021). Can Broadly Neutralizing HIV-1 Antibodies Help Achieve an ART-Free Remission. Front Immunol 12: 710044. doi: 10.3389/fimmu.2021.710044
  112. Li J Z, Aga E, Bosch R J, Pilkinton M, Kroon E, Maclaren L, Keefer M, Fox L, Barr L, Acosta E, Ananworanich J, Coombs R, Mellors J W, Landay A L, Macatangay B, Deeks S, Gandhi R T, Smith D M (2022). Time to Viral Rebound After Interruption of Modern Antiretroviral Therapies. Clin Infect Dis 74(5): 865870. doi:10.1093/cid/ciab541
  113. Iacob S A, Iacob D G, Jugulete G (2017). Improving the Adherence to Antiretroviral Therapy, a Difficult but Essential Task for a Successful HIV Treatment-Clinical Points of View and Practical Considerations. Front Pharmacol 8: 831. doi: 10.3389/fphar.2017.00831
  114. Phelps M, Balazs A B (2021). Contribution to HIV Prevention and Treatment by Antibody-Mediated Effector Function and Advances in Broadly Neutralizing Antibody Delivery by Vectored Immunoprophylaxis. Front Immunol 12: 734304. doi: 10.3389/fimmu.2021.734304
  115. Trkola A, Kuster H, Rusert P, Joos B, Fischer M, Leemann C, Manrique A, Huber M, Rehr M, Oxenius A, Weber R, Stiegler G, Vcelar B, Katinger H, Aceto L, Gunthard H F (2005). Delay of HIV-1 rebound after cessation of antiretroviral therapy through passive transfer of human neutralizing antibodies. Nature Med 11(6): 615622. doi: 10.1038/nm1244
  116. Stiegler G, Armbruster C, Vcelar B, Stoiber H, Kunert R, Michael N L, Jagodzinski L L, Ammann C, Jäger W, Jacobson J, Vetter N, Katinger H (2002). Antiviral activity of the neutralizing antibodies 2F5 and 2G12 in asymptomatic HIV-1-infected humans: a phase I evaluation. AIDS 16(15): 20192025. doi: 10.1038/nm1244
  117. Mahomed S, Garrett N, Capparelli E V, Osman F, Mkhize N N, Harkoo I, Gengiah T N, Mansoor L E, Baxter C, Archary D, Yende-Zuma N, Samsunder N, Carlton K, Narpala S, Mcdermott A B, Doria-Rose N A, Moore P L, Morris L, Abdool Karim Q, Mascola J R (2023). Safety and pharmacokinetics of escalating doses of neutralising monoclonal antibody CAP256V2LS administered with and without VRC07-523LS in HIV-negative women in South Africa (CAPRISA 012B): a phase 1, dose-escalation, randomised controlled trial. Abdool Karim SS 10(4): 36. doi: 10.1016/S2352-3018(23)00003-6
  118. Lynch R M, Boritz E, Coates E E, Dezure A, Madden P, Costner P, Enama M E, Plummer S, Holman L, Hendel C S, Gordon I, Casazza J, Conan-Cibotti M, Migueles S A, Tressler R, Bailer R T, Mcdermott A, Narpala S, O’Dell S, Wolf G, Lifson J D, Freemire B A, Gorelick R J, Pandey J P, Mohan S, Chomont N, Fromentin R, Chun T W, Fauci A S, Schwartz R M, et al. (2015). Virologic effects of broadly neutralizing antibody VRC01 administration during chronic HIV-1 infection. Sci Transl Med 7(319): 319206. doi: 10.1126/scitranslmed.aad5752
  119. Caskey M, Schoofs T, Gruell H, Settler A, Karagounis T, Kreider E F, Murrell B, Pfeifer N, Nogueira L, Oliveira T Y, Learn G H, Cohen Y Z, Lehmann C, Gillor D, Shimeliovich I, Unson-O’brien C, Weiland D, Robles A, Kümmerle T, Wyen C, Levin R, Witmer-Pack M, Eren K, Ignacio C, Kiss S, West A P, Mouquet H, Zingman B S, Gulick R M, Keler T, et al. (2017). Antibody 10-1074 suppresses viremia in HIV-1-infected individuals. Nat Med 23(2): 185191. doi: 10.1038/nm.4268
  120. Stephenson K E, Julg B, Tan C S, Zash R, Walsh S R, Rolle C P, Monczor A N, Lupo S, Gelderblom H C, Ansel J L, Kanjilal D G, Maxfield L F, Nkolola J, Borducchi E N, Abbink P, Liu J, Peter L, Chandrashekar A, Nityanandam R, Lin Z, Setaro A, Sapiente J, Chen Z, Sunner L, Cassidy T, Bennett C, Sato A, Mayer B, Perelson A S, Decamp A, et al. (2021). Safety, pharmacokinetics and antiviral activity of PGT121, a broadly neutralizing monoclonal antibody against HIV-1: a randomized, placebo-controlled, phase 1 clinical trial. Nat Med 27(10): 17181724. doi: 10.1038/s41591-021-01509-0
  121. Scheid J F, Horwitz J A, Bar-On Y, Kreider E F, Lu C L, Lorenzi J C, Feldmann A, Braunschweig M, Nogueira L, Oliveira T, Shimeliovich I, Patel R, Burke L, Cohen Y Z, Hadrigan S, Settler A, Witmer-Pack M, West A P, Juelg B, Keler T, Hawthorne T, Zingman B, Gulick R M, Pfeifer N, Learn G H, Seaman M S, Bjorkman P J, Klein F, Schlesinger S J, Walker B D, et al. (2016). HIV-1 antibody 3BNC117 suppresses viral rebound in humans during treatment interruption. Nature 535(7613): 556560. doi: 10.1038/nature18929
  122. Mendoza P, Gruell H, Nogueira L, Pai J A, Butler A L, Millard K, Lehmann C, Suárez I, Oliveira T Y, Lorenzi JCC, Cohen Y Z, Wyen C, Kümmerle T, Karagounis T, Lu C L, Handl L, Unson-O’brien C, Patel R, Ruping C, Schlotz M, Witmer-Pack M, Shimeliovich I, Kremer G, Thomas E, Seaton K E, Horowitz J, West A P, Bjorkman P J, Tomaras G D, Gulick R M, et al. (2018). Combination therapy with anti-HIV-1 antibodies maintains viral suppression. Nature 561(7724): 479484. doi: 10.1038/s41586-018-0531-2
  123. Sneller M C, Blazkova J, Justement J S, Shi V, Kennedy B D, Gittens K, Tolstenko J, Mccormack G, Whitehead E J, Schneck R F, Proschan M A, Benko E, Kovacs C, Oguz C, Seaman M S, Caskey M, Nussenzweig M C, Fauci A S, Moir S, Chun T W (2022). Combination anti-HIV antibodies provide sustained virological suppression. Nature 606(7913): 375381. doi: 10.1038/s41586-018-0531-2
  124. Julg B, Stephenson K E, Wagh K, Tan S C, Zash R, Walsh S, Ansel J, Kanjilal D, Nkolola J, Walker-Sperling VEK, Ophel J, Yanosick K, Borducchi E N, Maxfield LAbbink P, Peter L, Yates N L, Wesley M S, Hassell T, Gelderblom H C, Decamp A, Mayer B T, Sato A, Gerber M W, Giorgi E E, Gama L, Koup R A, Mascola J R, Monczor A, Lupo S, et al. (2022). Safety and antiviral activity of triple combination broadly neutralizing monoclonal antibody therapy against HIV-1: a phase 1 clinical trial. Nat Med 28(6): 12881296. doi: 10.1038/s41591-022-01815-1
  125. Moldt B, Parvangada A, Martin R, Pace C, Balakrishnan M, Thomsen N D, Collins S, Kuster H, Braun D L, Günthard H F, Geleziunas R, Callebaut C (2021). Evaluation of Broadly Neutralizing Antibody Sensitivity by Genotyping and Phenotyping for Qualifying Participants to HIV Clinical Trials. J Acquir Immune Defic Syndr 88(1): 6169. doi: 10.1097/QAI.0000000000002722
  126. Freind M C, Tallón De Lara C Tallón, Kouyos R D, Wimmersberger D, Kuster H, Aceto L, Kovari H, Flepp M, Schibli A, Hampel B, Grube C, Braun D L, Günthard H F (2024). Cohort Profile: The Zurich Primary HIV Infection Study. Microorganisms 12(2): 302. doi: 10.3390/microorganisms12020302
  127. Lamont C, Otwinowski J, Vanshylla K, Gruell H, Klein F, Nourmohammad A (2022). Design of an optimal combination therapy with broadly neutralizing antibodies to suppress HIV-1. Elife 11: 76004. doi: 10.7554/eLife.76004
  128. Moraka N O, Choga W T, Pema M N, Chawawa M K, Gobe I, Mokomane M, Bareng O T, Bhebhe L, Kelentse N, Mulenga G, Pretorius Holme M, Mohammed T, Koofhethile C K, Makhema J M, Shapiro R, Lockman S, Moyo S, Gaseitsiwe S (2023). Predicted resistance to broadly neutralizing antibodies (bnAbs) and associated HIV-1 envelope characteristics among seroconverting adults in Botswana. Sci Rep 13(1): 18134. doi: 10.1038/s41598-023-44722-2
  129. Shapiro R L, Ajibola G, Maswabi K, Hughes M, Nelson B S, Niesar A, Pretorius Holme M, Powis K M, Sakoi M, Batlang O, Moyo S, Mohammed T, Maphorisa C, Bennett K, Hu Z, Giguel F, Reeves J D, Reeves M A, Gao C, Yu X, Ackerman M E, Mcdermott A, Cooper M, Caskey M, Gama L, Jean-Philippe P, Yin D E, Capparelli E V, Lockman S, Makhema J, et al. (2023). Broadly neutralizing antibody treatment maintained HIV suppression in children with favorable reservoir characteristics in Botswana. Sci Transl Med 15(703): 4. doi: 10.1128/JVI.00491-12
  130. Gaudinski M R, Houser K V, Doria-Rose N A, Chen G L, Rothwell RSS, Berkowitz N, Costner P, Holman L A, Gordon I J, Hendel C S, Kaltovich F, Conan-Cibotti M, Lorenzo M Gomez, Carter C, Sitar S, Carlton K, Gall J, Laurencot C, Lin B C, Bailer R T, Mcdermott A B, Ko S Y, Pegu A, Kwon Y D, Kwong P D, Namboodiri A M, Pandey J P, Schwartz R, Arnold F, Hu Z, et al. (2019). Safety and pharmacokinetics of broadly neutralising human monoclonal antibody VRC07-523LS in healthy adults: a phase 1 dose-escalation clinical trial. Lancet HIV 6(10): 667679. doi: 10.1016/S2352-3018(19)30181-X
  131. Akilesh S, Christianson G J, Roopenian D C, Shaw A S (2007). Neonatal FcR expression in bone marrow-derived cells functions to protect serum IgG from catabolism. J Immunol 179(7): 45804588. doi: 10.4049/jimmunol.179.7.4580
  132. Roopenian D C, Akilesh S (2007). FcRn: the neonatal Fc receptor comes of age. Nat Rev Immunol 7(9): 715725. doi: 10.1038/nri2155
  133. Zalevsky J, Chamberlain A K, Horton H M, Karki S, Leung I W, Sproule T J, Lazar G A, Roopenian D C, Desjarlais J R (2010). Enhanced antibody half-life improves in vivo activity. Nat Biotechnol 28(2): 157159. doi: 10.1038/nbt.1601
  134. Ko S Y, Pegu A, Rudicell R S, Yang Z Y, Joyce M G, Chen X, Wang K, Bao S, Kraemer T D, Rath T, Zeng M, Schmidt S D, Todd J P, Penzak S R, Saunders K O, Nason M C, Haase A T, Rao S S, Blumberg R S, Mascola J R, Nabel G J (2014). Enhanced neonatal Fc receptor function improves protection against primate SHIV infection. Nature 514(7524): 642645. doi: 10.1038/nature13612
  135. Chen J, Zhou T, Zhang Y, Luo S, Chen H, Chen D, Li C, Li W (2022). The reservoir of latent HIV. Front Cell Infect Microbiol 12: 945956. doi: 10.3389/fcimb.2022.945956
  136. Moldt B, Günthard H F, Workowski K A, Little S J, Eron J J, Overton E T, Lehmann C, Rokx C, Kozal M J, Gandhi R T, Braun D L, Parvangada A, Li J, Martin R, Selzer L, Cox S, Margot N, Liu H, Slamowitz D, Makadzange T, Collins S E, Geleziunas R, Callebaut C (2022). Evaluation of HIV-1 reservoir size and broadly neutralizing antibody susceptibility in acute antiretroviral therapy-treated individuals. AIDS 36(2): 205214. doi: 10.1097/QAD.0000000000003088
  137. Leyre L, Kroon E, Vandergeeten C, Sacdalan C, Colby D J, Buranapraditkun S, Schuetz A, Chomchey N, De Souza M, Bakeman W, Fromentin R, Pinyakorn S, Akapirat S, Trichavaroj R, Chottanapund S, Manasnayakorn S, Rerknimitr R, Wattanaboonyoungcharoen P, Kim J H, Tovanabutra S, Schacker T W, O’connell R, Valcour V G, Phanuphak P, Robb M L, Michael N, Trautmann L, Phanuphak N, Ananworanich J, Chomont N, et al. (2020). Abundant HIV-infected cells in blood and tissues are rapidly cleared upon ART initiation during acute HIV infection. Sci Transl Med 12(533): 3491. doi: 10.1126/scitranslmed.aav3491
  138. Kula-Pacurar A, Rodari A, Darcis G, Van Lint C (2021). Shocking HIV-1 with immunomodulatory latency reversing agents. Semin Immunol 51: 101478. doi: 10.1016/j.smim.2021.101478
  139. Abner E, Jordan A (2019). HIV “shock and kill” therapy: In need of revision. Antiviral Res 166: 1934. doi: 10.1016/j.antiviral.2019.03.008
  140. Dash P K, Chen C, Kaminski R, Su H, Mancuso P, Sillman B, Zhang C, Liao S, Sravanam S, Liu H, Waight E, Guo L, Mathews S, Sariyer R, Mosley R L, Poluektova L YCaocci M, Amini S, Gorantla S, Burdo T H, Edagwa B, Gendelman H E, Khalili K (2019). CRISPR editing of CCR5 and HIV-1 facilitates viral elimination in antiretroviral drug-suppressed virus-infected humanized mice. Proc Natl Acad Sci U S A 120: 2217887120. doi: 10.1073/pnas.2217887120
  141. Liu Y, Binda C S, Berkhout B, Das A T (2023). CRISPR-Cas attack of HIV-1 proviral DNA can cause unintended deletion of surrounding cellular DNA. J Virol 97(12): 133423. doi: 10.1128/jvi.01334-23
  142. Seddiki N, Picard F, Dupaty L, Lévy Y, Godot V (2020). The Potential of Immune Modulation in Therapeutic HIV-1 Vaccination. Vaccines 8(3): 419. doi: 10.3390/vaccines8030419
  143. Richel E, Wagner J T, Klessing S, Di Vincenzo R, Temchura V, Überla K (2023). Antigen-dependent modulation of immune responses to antigen-Fc fusion proteins by Fc-effector functions. Front Immunol 14: 1275193. doi: 10.3389/fimmu.2023.1275193
  144. Martinsen J T, Gunst J D, Højen J F, Tolstrup M, Søgaard O S  (2020). The Use of Toll-Like Receptor Agonists in HIV-1 Cure Strategies. Front Immunol 11: 1112. doi: 10.3389/fimmu.2020.01112
  145. Prener L, Baszczyňski O, Kaiser M M, Dračínský M, Stepan G, Lee Y J, Brumshtein B, Yu H, Jansa P, Lansdon E B, Janeba Z (2023). Design and Synthesis of Novel HIV-1 NNRTIs with Bicyclic Cores and with Improved Physicochemical Properties. J Med Chem 66(3): 17611777. doi: 10.1021/acs.jmedchem.2c01574
  146. Cambou M C, Landovitz R J (2020). Novel Antiretroviral Agents. Curr HIV/AIDS Rep 17(2): 118124. doi: 10.1007/s11904-020-00486-2
  147. Jean M J, Fiches G, Hayashi T, Zhu J (2019). Current Strategies for Elimination of HIV-1 Latent Reservoirs Using Chemical Compounds Targeting Host and Viral Factors. AIDS Res Hum Retroviruses 35(1): 124. doi: 10.1089/AID.2018.0153
  148. Ngo M H, Pankrac J, Ho RCY, Ndashimye E, Pawa R, Ceccacci R, Biru T, Olabode A S, Klein K, Li Y, Kovacs C, Assad R, Jacobson J M, Canaday D H, Tomusange S, Jamiru S, Anok A, Kityamuweesi T, Buule P, Galiwango R M, Reynolds S J, Quinn T C, Redd A D, Prodger J L, Mann JFS (2024). Effective and targeted latency reversal in CD4. Emerg Microbes Infect 13(1): 2327371. doi: 10.1080/22221751.2024.2327371
  149. Frattari G S, Caskey M, Søgaard O S (2023). Broadly neutralizing antibodies for HIV treatment and cure approaches. Curr Opin HIV AIDS 18(4): 157163. doi: 10.1097/COH.0000000000000802
  150. Cohen Y Z, Lorenzi JCC, Krassnig L, Barton J P, Burke L, Pai J, Lu C L, Mendoza P, Oliveira T Y, Sleckman C, Millard K, Butler A L, Dizon J P, Belblidia S A, Witmer-Pack M, Shimeliovich I, Gulick R M, Seaman M S, Jankovic M, Caskey M, Nussenzweig M C (2018). Relationship between latent and rebound viruses in a clinical trial of anti-HIV-1 antibody 3BNC117. J Exp Med 215(9): 23112324. doi: 10.1084/jem.20180936
  151. Gruell H, Gunst J D, Cohen Y Z, Pahus M H, Malin J J, Platten M, Millard K G, Tolstrup M, Jones R B, Conce Alberto W D, Lorenzi JCC, Oliveira T Y, Kümmerle T, Suárez I,Unson-O’brien C, Nogueira L, Olesen R, Østergaard L, Nielsen H, Lehmann C, Nussenzweig M C, Fätkenheuer G, Klein F, Caskey M, Søgaard O S (2022). Effect of 3BNC117 and romidepsin on the HIV-1 reservoir in people taking suppressive antiretroviral therapy (ROADMAP): a randomised, open-label, phase 2A trial. Lancet Microbe 3(3): 203214. doi: 10.1016/S2666-5247(21)00239-1
  152. Gunst J D, Pahus M H, Rosás-Umbert M, Lu I N, Benfield T, Nielsen H, Johansen I S, Mohey R, Østergaard L, Klastrup V, Khan M, Schleimann M H, Olesen R, Støvring H, Denton P W, Kinloch N N, Copertino D C, Ward A R, Alberto WDC, Nielsen S D, Puertas M C, Ramos V, Reeves J D, Petropoulos C J, Martinez-Picado J, Brumme Z L, Jones R B, Fox J, Tolstrup M, Nussenzweig M C, et al. (2022). Early intervention with 3BNC117 and romidepsin at antiretroviral treatment initiation in people with HIV-1: a phase 1b/2a, randomized trial. Nat Med 28(11): 24242435. doi: 10.1038/s41591-022-02023-7
  153. Rosás-Umbert M, Gunst J D, Pahus M H, Olesen R, Schleimann M, Denton P W, Ramos V, Ward A, Kinloch N N, Copertino D C, Escribà T, Llano A, Brumme Z L, Jones R Brad, Mothe B, Brander C, Fox J, Nussenzweig M C, Fidler S, Caskey M, Tolstrup M, Søgaard O S (2022). Administration of broadly neutralizing anti-HIV-1 antibodies at ART initiation maintains long-term CD8. Nat Commun 13(1): 6473. doi: 10.1038/s41467-022-34171-2
  154. Mavigner M, Chahroudi A (2023). The next thing” to treat children with HIV? Sci Transl Med 15(703): 293. doi: 10.1126/scitranslmed.adi0293
  155. Corey L, Gilbert P B, Juraska M, Montefiori D C, Morris L, Karuna S T, Edupuganti S, Mgodi N M, Decamp A C, Rudnicki E, Huang Y, Gonzales P, Cabello R, Orrell C, Lama J R, Laher F, Lazarus E M, Sanchez J, Frank I, Hinojosa J, Sobieszczyk M E, Marshall K E, Mukwekwerere P G, Makhema J, Baden L R, Mullins J I, Williamson C, Hural J, Mcelrath M J, Bentley C, et al. (2021). Two Randomized Trials of Neutralizing Antibodies to Prevent HIV-1 Acquisition. N Engl J Med 384(11): 10031014. doi: 10.1056/NEJMoa2031738
  156. Cohen Y Z, Butler A L, Millard K, Witmer-Pack M, Levin R, Unson-O’Brien C, Patel R, Shimeliovich I, Lorenzi JCC, Horowitz J, Walsh S R, Lin S, Weiner J A, Tse A, Sato A, Bennett C, Mayer B, Seaton K E, Yates N L, Baden L R, deCamp A C, Ackerman M E, Seaman M S, Tomaras G D, Nussenzweig M C, Caskey M (2019). Safety, pharmacokinetics, and immunogenicity of the combination of the broadly neutralizing anti-HIV-1 antibodies 3BNC117 and 10-1074 in healthy adults: A randomized, phase 1 study. PLoS One 14(8): e0219142. doi: 10.1371/journal.pone.0219142
  157. Politsch J A, Cu-Uvin S, Moench T R, Tashima K T, Marathe J G, Guthrie K M, Cabral H, Nyhuis T, Brennan M, Zeitlin L, Spiegel HML, Mayer K H, Whaley K J, Anderson D J (2021). Safety, acceptability, and pharmacokinetics of a monoclonal antibody-based vaginal multipurpose prevention film (MB66): A Phase I randomized trial. . PLoS Med 18(2): e1003495. doi: 10.1371/journal.pmed.1003495

 

ACKNOWLEDGMENTS

The authors thank Valerie Gach for crafting Figure 1 and Figure 2 with Procreate (Version 5.3.6).

COPYRIGHT

© 2024

Creative Commons License
Neutralizing the threat: harnessing broadly neutralizing antibodies against HIV-1 for treatment and prevention by Becerra et al. is licensed under a Creative Commons Attribution 4.0 International License.

By continuing to use the site, you agree to the use of cookies. more information

The cookie settings on this website are set to "allow cookies" to give you the best browsing experience possible. If you continue to use this website without changing your cookie settings or you click "Accept" below then you are consenting to this. Please refer to our "privacy statement" and our "terms of use" for further information.

Close