Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2020 Jan 10.
Published in final edited form as: BioDrugs. 2012 Jun 1;26(3):143–162. doi: 10.2165/11631400-000000000-00000

Candidate Antibody-Based Therapeutics Against HIV-1

Rui Gong 1, Weizao Chen 1, Dimiter S Dimitrov 1
PMCID: PMC6953404  NIHMSID: NIHMS1065951  PMID: 22462520

Abstract

Antibody-based therapeutics have been successfully used for the treatment of various diseases and as research tools. Several well characterized, broadly neutralizing monoclonal antibodies (bnmAbs) targeting HIV-1 envelope glycoproteins or related host cell surface proteins show sterilizing protection of animals, but they are not effective when used for therapy of an established infection in humans. Recently, a number of novel bnmAbs, engineered antibody domains (eAds), and multifunctional fusion proteins have been reported which exhibit exceptionally potent and broad neutralizing activity against a wide range of HIV-1 isolates from diverse genetic subtypes. eAds could be more effective in vivo than conventional full-size antibodies generated by the human immune system. Because of their small size (12~15kD), they can better access sterically restricted epitopes and penetrate densely packed tissue where HIV-1 replicates than the larger full-size antibodies. HIV-1 possesses a number of mechanisms to escape neutralization by full-size antibodies but could be less likely to develop resistance to eAds. Here, we review the in vitro and in vivo antiviral efficacies of existing HIV-1 bnmAbs, summarize the development of eAds and multispecific fusion proteins as novel types of HIV-1 inhibitors, and discuss possible strategies to generate more potent antibody-based candidate therapeutics against HIV-1, including some that could be used to eradicate the virus.

1. Introduction

More than 60 million people have been infected by HIV-1 and more than 25 million people have died of HIV-1-related diseases. Currently, a variety of treatments are available. A highly active antiretroviral therapy (HAART) or ‘cocktail therapy’, that includes two nucleoside/nucleotide reverse-transcriptase inhibitors (NRTIs/NtRTIs) plus a non-nucleoside reverse-transcriptase inhibitor (NNRTI) or a protease inhibitor (PI) or another NRTI, is a highly efficient method in reducing the number of HIV-1 particles in the bloodstream to undetectable levels and providing clinical benefits.[13] However, HAART can lead to serious adverse effects[4,5] and the emergence of drug-resistant mutants.[6,7] Several entry inhibitors were developed, including the HIV-1 entry inhibitor T20 (Enfuvirtide, Fuzeon) and the C-C chemokine receptor type 5 (CCR5) antagonist maraviroc (Selzentry®, or Celsentri® outside the US). Very recently, raltegravir (Isentress®), which represents a new class of intergrase inhibitors, was approved by the US FDA for clinical use. Still, primary resistance has already been observed with the use of these new drugs.[8]

A major problem for these therapeutics is that long-term use can lead to drug-related toxicities. In addition, there is a continuous need for new drugs to cope with potential drug resistance. Monoclonal antibodies (mAbs) are, in general, safer than small molecule drugs and have been successful in the prevention and/or treatment of various diseases including cancers, immune disorders, and infectious diseases. To date, more than 30 mAbs have been approved in the US and EU, and hundreds of mAbs have been evaluated in clinical trials in the past decade.[9] However, none of the approved mAbs is for the treatment of HIV-1-infected patients. Palivizumab (Synagis®), which targets the respiratory syncytial virus (RSV), is the only mAb approved for clinical use against any infectious disease, and it is for prevention, not for therapy.

One of the difficulties in developing mAbs as HIV-1 therapeutics is the extreme variability of the virus and the rapid emergence of resistant mutants.[10] This requires that antibodies exhibit sufficient level of breadth and potently neutralize genetically diverse HIV-1 isolates. Despite the disappointing results from several broadly neutralizing mAbs (bnmAbs) tested in human clinical trials, the fact that passive immunization is protective as prophylaxis in animals and the recent identification of novel, more potent bnmAbs indicate some potential for antibody-based therapeutics against HIV-1.

2. Targets

HIV-1 entry is initiated by binding of its envelope glycoprotein (Env) gp120 to receptor CD4 on the target cell surface. The binding induces extensive conformational changes in gp120, resulting in the formation of the coreceptor-binding site and enabling binding of gp120 to a coreceptor, either CCR5 or C-X-C chemokine receptor type 4 (CXCR4). Substantial structural rearrangements then occur in the Env gp41 which eventually causes virus-cell fusion and injection of the viral genomic DNA into target cells.

2.1. HIV-1 Envelope Glycoproteins (Envs)

The Env is a typical type I viral envelope protein. It is synthesized as a precursor glycoprotein, gp160, which is proteolytically cleaved into a surface subunit, gp120 (for receptor and coreceptor binding), and a transmembrane subunit, gp41 (for virus-cell fusion). The two subunits exist as a trimeric state on the viral spike and interact with each other non-covalently (figure 1).

Fig. 1.

Fig. 1.

Entry of HIV-1 into CD4+ cell. After binding of gp120 to CD4 (pdb: 1WIP,[11,12] rendered by PyMOL), serial conformational change occurs including: (i) exposure of co-receptor binding site and subsequent binding of gp120 to co-receptor; (ii) exposure of fusion peptide; (iii) formation of fusion core on gp41 through interaction between NHR and CHR; (iv) re-arrangement of MEPR region, etc. These events lead to exposure of the new epitope on the Env which could be targeted by broadly neutralizing antibodies. CHR = C-terminal heptad repeat; CoR = coreceptor; D1–D4 = domain 1-domain 4; Env = envelope glycoprotein; FP = fusion peptide; MPER = membrane proximal external region; NHR = N-terminal heptad repeat.

Gp120 is highly glycosylated and consists of five conserved domains, C1–C5, and five variable loops, V1–V5.[13,14] The receptor CD4-binding site (CD4bs) on gp120 is a conserved pocket-like structure flanked by some of the loops (figure 2). Although it is recessed to a certain extent from antibody access, broad and potent HIV-1 neutralization by sera from some HIV-1-infected patients is mediated to a significant extent by CD4bs antibodies. Another highly conserved structure on gp120, the coreceptor binding site (CoRbs), is exposed and/or formed after virus attachment to host cells via CD4 binding and, therefore, antibodies targeting the CoRbs are called CD4-induced (CD4i) antibodies (figure 2). This structure is also highly immunogenic. However, because the CoRbs is oriented closely toward the cellular membrane, which leaves a very small space in between, full-size CD4i antibodies generated by the immune system are generally less potent and broad compared with the CD4bs antibodies. Among the loop structures, V3 (figure 2) is particularly functionally important. First, it recognizes a coreceptor.[17] Second, it may determine the overall sensitivity of the virus to neutralization through interaction with other elements on the viral spike.[18,19] It is highly immunogenic and, therefore, represents one of the major targets of neutralizing responses elicited by infection or immunization with HIV-1 Envs.[20,21] However, the antibodies targeting V3 might exhibit limited breadth of neutralization or may be isolate-specific due to high variability of the loop sequences and structures. Although non-immunogenic, the ‘glycan shield’ on gp120 can, unusually, elicit immune responses in a small number of HIV-1-infected subjects (figure 2), which potently and broadly neutralize primary and T-cell line-adapted HIV-1 isolates and inhibit syncytium formation in cell lines.[22,23] It has been found that gp120 in trimeric states adopts some quaternary conformations that are primarily located in the conserved regions of the V2 and V3 loops of gp120.[24,25] MAbs isolated against these epitopes exhibit a high level of neutralizing activities and breadth comparable to or higher than those of the bnmAbs to monomeric gp120. This finding suggests additional conserved regions on the native Env trimer may exist and remain to be defined. Antibodies to these regions could better affect virus entry, have more potential as therapeutics, and are highly valuable for vaccine immunogen design.

Fig. 2.

Fig. 2.

Neutralizing epitopes on HIV-1 Env gp160. The structure (pdb: 1G9M[15,16]) of a gp120 core from the HXBc2 laboratory-adapted isolate in complex with a 2 domain (D1 and D2) fragment of CD4 and Fab 17b targeting CD4i epitope was rendered by PyMOL. The positions of V1/V2 and V3 are also indicated. CD4bs = CD4 binding site; CHR = C-terminal heptad repeat; CoRbs (CD4i) = coreceptor binding site (CD4 induced epitope); Env = envelope glycoprotein; MPER = membrane proximal external region; NHR = N-terminal heptad repeat.

Gp41 (figure 1) is directly involved in viral-cell membrane fusion and, therefore, is an important target for antibody intervention. It is composed of a fusion peptide (FP) in the N terminus followed by the polar region, the N-terminal heptad repeat (NHR), the C-terminal heptad repeat (CHR), the immunodominant region and the membrane proximal external region (MPER).[26,27] The FP is highly hydrophobic and is hidden before gp120 binds to CD4.[28] Previous studies showed that an FP variant, in which the highly conserved residues are replaced by their D-enantiomers, could inhibit HIV-1-mediated cell-cell fusion.[29] Moreover, a synthetic peptide targeting the FP is capable of inhibiting a wide variety of HIV-1 strains.[30] After exposure of gp41, the NHR and the CHR can interact with each other and form a six-stranded α-helical bundle consisting of an inner triple stranded coiled-coil buttressed by three C-helices, which is the fusion core structure of gp41 and crucial for membrane fusion[31,32] (figure 1). These two structures are ‘hot regions’ because peptides from the CHR (e.g. C34[33,34]) could inhibit the viral entry efficiently through interaction with the NHR. Between the CHR and the transmembrane domain is the MPER, a conserved tryptophan-rich region containing about 20 residues (figure 2). Mutational and biophysical analysis has provided compelling evidence to support the involvement of the MPER in HIV-1 fusion.[35,36] Therefore, antibodies against these regions could have the potential of inhibiting the entry of HIV-1 into host cells.

2.2. Receptor Molecules

In contrast to the Env, host receptor molecules, which are important for viral entry, are conserved. The HIV-1 primary receptor CD4 is a type I integral membrane glycoprotein (58 kDa) that is expressed mainly on the surface of thymocytes, T-helper lymphocytes, and cells of the macrophage/monocyte lineage.[3739] CD4 is required for shaping the T-cell repertoire during thymic development, is necessary for appropriate activation of mature T cells,[40] and helps B cells induce antibody responses.[41] CD4 consists of an extracellular region of 370 amino acid residues organized in four immunoglobulin-like domains (D1–D4).[42,43] The crystal structure of D1–D4 (Protein Data Bank [pdb]: 1WIP[11,12]) was rendered by PyMOL (The PyMOL Molecular Graphics System, Version 1.2, Schrödinger, LLC) (figure 1). The first domain, D1, directly binds to gp120.[44,45] It has been found that CD4 also contains a binding site for protein disulfide isomerase (PDI) and forms a PDI-CD4-gp120 complex, causing major conformational changes in gp120 to activate the fusogenic potential of HIV-1 Env.[4654] Gp120-CD4 interaction induces conformational changes not only in gp120 but also in CD4. Therefore, interference with CD4 conformational changes by antibodies interacting with native CD4 but not with gp120-complexed CD4 represents a novel neutralization principle in addition to directly blocking the gp120-binding surface.

HIV-1 coreceptors (CoRs) [figure 1], CCR5[5560] and CXCR4,[61] are also required for viral entry and some of the HIV-1 isolates can infect coreceptor-expressing cells in a CD4-independent manner, which makes coreceptors an attractive target for drug development.[6265] In addition, unlike CD4, CCR5 may not be functionally important for host cells. A minority of people (about 1/1000 Northern Europeans) are born with CCR5 deletion mutation, which endows them with inherent resistance to HIV-1 infection.[66,67] Therefore, blockade and depletion of the coreceptor may not result in adverse effects such as immunosuppression caused by some of the CD4 antibodies. The receptor and coreceptor are self-antigens and do not elicit antibody responses in humans. However, antibodies against these molecules can be produced by immunization of animals followed by humanization to reduce immunogenicity. Such antibodies can also be selected from libraries where heavy and light chains of naturally occurring human antibodies are randomly assembled.

3. Full-Size Monoclonal Antibodies

3.1. Targeting Envs

The mAb b12 (see table I for some representatives of anti-HIV-1 antibodies and antibody-based proteins) is the first reported representative of the bnmAbs that targets the CD4bs on gp120 as a competitive inhibitor of CD4 binding. It was selected from a phage-displayed antibody library constructed from the bone marrow of an HIV-1-infected donor.[68,69] B12 efficiently neutralizes a wide range of HIV-1 isolates from different clades in vitro[70,71] and can protect macaques against vaginal challenge with a simian-human immunodeficiency virus (SHIV) that uses CCR5 as a coreceptor.[7274] It was also found that b12 could mediate strong antibody-dependent cellular cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC) of HIV-1-infected cells in vitro.[75] However, ADCC is probably the major effector function for protection against HIV-1 infection in vivo.[76] The four other mAbs: F105,[77] b13,[78] m14,[79] and m18,[80] which also interact with the CD4bs, have only modest neutralizing activities. Structure analysis reveals that slight differences in recognition result in substantial differences in F105 and b13-bound conformations relative to b12-bound gp120.[81,82] Modeling and binding experiments showed these conformations are poorly compatible with the viral spike. These findings indicate that the accuracy of CD4bs recognition is crucial for neutralization of HIV-1. About 20 years after the discovery of b12, two mAbs targeting the CD4bs, VRC01 and VRC02, were recently identified by isolation of single mAb-producing B cells against an antigenically resurfaced HIV-1 gp120 specific for the structurally conserved site of initial CD4 receptor binding.[83] Notably, VRC01 and VRC02 can neutralize over 90% of circulating HIV-1 isolates with high potency in vitro. Although VRC01 partially mimics the binding of CD4 to gp120, a shift from the CD4-defined orientation focuses VRC01 onto the vulnerable site of initial CD4 attachment, allowing it to overcome the glycan and conformational masking that diminishes the neutralization potency of most CD4bs antibodies.[84] Interestingly, a single substitution in VRC01 heavy-chain complementarity determining region 2 (CDR2) could increase contact with the gp120 bridging sheet and improve breadth and potency, critical properties for potential clinical use.[85] The characterization of 576 new HIV-1 antibodies from four unrelated individuals showed these new antibodies shared a consensus sequence of 68 immunoglobulin heavy-chain amino acids and arose independently from two related heavy-chain genes.[86] The crystal structure of one of these antibodies and VRC01 shared conserved contacts to the HIV-1 Env.[86]

Table I.

Some representatives of antibodies and antibody-like fusion proteins against HIV-1

Epitope Name Year Type Animal/virus Clinical trial In vivo efficacy Comment
CD4bs b12 1991 Human lgG1 Macaque/CCR5-tropic SHIV (162P4) No 4/4 animals were protected from vaginal infection at dose of 25mg/kg through intravenous injection; 9/12 animals were protected from vaginal infection at dose of 5 mg/kg through vaginal administration First reported bnmAb targeting CD4bs, solved crystal structure
VRC01 VRC02 2010 Human lgG1 NA No NA Can neutralize over 90% of circulating HIV-1 isolates with high potency in vitro, solved crystal structure of VRC01
PRO 542 (CD4-lgG2) 1995 IgG-like fusion protein Human-PBL-SCID mouse/HIV-1 Phase II Well tolerated at 25 mg/kg in 12 HIV-infected individuals, mediated an 80% response rate and statistically significant ≈ 0.5 log10 mean reductions in viral load for 4–6 wk posttreatment in advanced disease (HIV-1 RNA >100 000copies/mL; CD4 lymphocytes <200 cells/mm3) Fusion of CD4 D1 and D2 domains to lgG2, longer half-life (3 days) than sCD4 (45 min), does not appear to bind to Fc receptors
CoRbs (CD4i) 17b 1996 Human Fab/lgG1 NA No NA Binds to CD4i epitope that overlap with the CoRbs, solved crystal structure of gp120 core/two-domain soluble CD4/Fab 17b
412d 2001 Human lgG1 NA No NA A critical sulfotyrosine on CCR5 and on 412d induces similar structural rearrangements in gp120
X5 2002 Human Fab/lgG1 NA No NA Shows the existence of conserved receptor-inducible gp120 epitopes that can serve as targets
m36 2008 Human lgG1 V domain Humanized NOD/SCID/γcnull mouse/HIV-1 No 1 mg m36.4, an affinity-matured version of m36, provided sterilizing protection of 4/6 animals against intrasplenic challenge with high-titer HIV-1 First reported V domain-based eAd that targets a sterically restricted CD4i epitope and potently neutralizes genetically diverse HIV-1 isolates in vitro
m1a1 2009 Human lgG1 C domain No No NA First reported C domain-based eAd that recognizes a highly conserved CD4i epitope overlapping with that of m36 CD4i epitope, modest neutralization activity in vitro
V3 Cβi 1988 Humanized mouse IgG Chimpanzee/HIV-1 (lllb) No Protected a chimpanzee at dose of 36 mg/kg from HIV-1 infection during 52-week observation period First characterized bnmAb targeting V3 of gp120
hNM01 2004 Humanized mouse lgG1 NA Phase I Dose: day 1: 0.2mg/kg, day 15:1 mg/kg, day 29:5 mg/kg, and day 43: 5 mg/kg; 3/4 patients had a decrease in their plasma viral load measurements; no significant change in CD4 counts Neutralizes the virus by the activation of complement and the subsequent disruption of the viral envelope after binding to V3
KD-247 2006 Humanized mouse lgG1 Cynomolgus/SHIV (C2/1) Phase I Passive immunization at a single dose of 45 mg/kg 24 h prior to viral challenge completely protected animals from viral challenge; phase I study is ongoing Efficiently neutralizes CXCR4- and CCR5-tropic primary HIV-1 clade B and clade B’ and suppress the ex vivo generation of primary HIV-1 quasispecies in peripheral blood mononuclear cell cultures from HIV-1 - infected individuals
HGN194 2010 Human lgG1 Macaque/SHIV (CCR5-tropic, Clade C) No After high-dose mucosal challenge, all treated animals with dose of 50 mg/kg were completely protected Identified by using appropriate screening methods, neutralizes all Tier-1 and a proportion of Tier-2 pseudoviruses tested, irrespective of clade
Quarter-nary structure PG9 PG16 2009 Human lgG1 NA No NA Selected from a high-throughput neutralization screen of antibody-containing culture supernatants of about 30 000 activated memory B cells from a clade A-infected African donor, solved crystal structure of V1/V2 in complex with PG9
PG9 PG16 2009 Human lgG1 NA No NA Selected from a high-throughput neutralization screen of antibody-containing culture supernatants of about 30 000 activated memory B cells from a clade A-infected African donor, solved crystal structure of V1/V2 in complex with PG9
Carbohydrate PGT128 2011 Human lgG1 NA No NA Cyrstal structure of complex of Fab PGT 128/fully glycosylated gp120 outer domain reveals that the antibody penetrates the glycan shield and recognizes two conserved glycans as well as a short β-strand segment of the gp120 V3 loop, accounting for its high binding affinity and broad specificity
2G12 1996 Human lgG1 Macaque/SHIV (89.6PD and vpu+) Phase II Administered 1 g 2G12, 1 g 4E10 and 1,3g 2F5 (combination) sequentially in each infusion; 2/8 chronically and 4/6 acutely infected individuals showed evidence of a pronounced delay in viral rebound during Adopts a multivalent binding surface by using a unique domain-exchanged structure, in which the heavy chain variable domains of two antibody molecules are swapped
MPER 2F5 1993 Human lgG1 Macaque/SHIV (89.6PD and vpu+) Phase II See description in 2G12 Spurs interest in its structural characterization: these two bnmAbs induces large conformational changes in the MPER relative to the membrane, phase 11 clinical trial is ongoing
4E10 2001 Human lgG1 NA Phase II
CD4 TNX-355 (ibalizumab, hu5A8) 1992 Humanized lgG4 Macaque/S IV Phase II Treatment of arm A: 10 mg/kg, weekly, 10 doses; arm B: a single loading dose of 10 mg/kg on day 1, followed by 5 maintenance doses of 6 mg/kg biweekly and arm C: 25 mg/kg, biweekly, 5 doses resulted in substantial reductions in HIV-1 RNA levels (0.5 to 1,7log10) in 20 of 22 subjects Targets the D2 domain of human and rhesus CD4; phase II clinical trial is ongoing
CD4-BFFI 2009 IgG-like fusion protein NA No NA Combines the anti-CD4 mAb 6314 with a fusion inhibitor similar to T-651; in vivo pharmacokinetic studies demonstrate that CD4-BFFI is stable in monkey blood, and a dose of 10 mg/kg maintains serum concentrations greater than 2000-fold over the IC90 value for 7 days post-dosing; higher antiviral potency compared with the fusion inhibitor T-651 or the anti-CD4 mAb 6314
CCR5 PRO 140 1999 Humanized lgG4 NA Phase II Subcutaneous PRO 140 demonstrated potent and prolonged antiretroviral activity. Mean log10 reductions in HIV-1 RNA level were 0.23, 0.99, 1.37, and 1.65 for the placebo, 162 mg weekly, 324 mg biweekly, and 324 mg weekly dose groups, respectively Exhibits potent and broad-spectrum inhibition of primary HIV-1 R5 isolates from different clades but does not affect CCR5 activity; phase II clinical trial is ongoing
CCR5–2320 2011 IgG-like fusion protein NA No NA Constructed by connection of scFvs with IgGs, in contrast to monospecific CCR5 antibodies; bispecific antibody derivatives block two alternative docking sites of CCR5-tropic HIV strains on the CCR5 coreceptor; consequently, these molecules show significantly increased antiviral activity compared with the parent antibodies

bnmAbs = broadly neutralizing monoclonal antibodies; CCR5 = C-C chemokine receptor type 5; CD4bs = receptor CD4-binding site; CD4i = CD4-induced; CoRbs = coreceptor binding site; CXCR4 = C-X-C chemokine receptor type 4; eAd = engineered antibody domain; IC90 = inhibitory concentration at which 90% of bacteria are inhibited; MPER = membrane proximal external region; NA = not applicable; NOD = non-obese diabetic; PBL = peripheral blood lymphocyte; sCD4 = soluble CD4; scFvs = single-chain variable fragments; SCID = severe combined immunodeficiency; SHIV = simian-human immunodeficiency virus; SIV = simian immunodeficiency virus.

17b and 48d are human mAbs isolated from different HIV-1-infected individuals.[87] They bind to CD4i epitopes that overlap with the CoRbs.[88] The crystal structure of gp120 core/two-domain soluble CD4 (sCD4)/antigen-binding fragment (Fab) 17b shows a cavity-laden CD4-gp120 interface and a conserved binding site for the coreceptor, and provides information for conformational changes of gp120 upon CD4 binding, the nature of CD4i epitopes, and possible mechanisms of immune evasion (figure 2).[15,89] CD4i antibodies 412d and E51 are obtained from an HIV-1-infected individual by selection of the potent enzyme-linked immunosorbent assay (ELISA) response of his serum to the HIV-1 gp120[90] that functionally emulates CCR5. The tyrosine sulfation on 412d and E51 can contribute to the potency and diversity of the human humoral response.[91,92] Although the conformations of tyrosine-sulfated regions of CCR5 (α-helix) and 412d (extended-loop) are dramatically different, a critical sulfotyrosine on CCR5 and on 412d induces similar structural rearrangements in gp120.[93] A sulfated peptide derived from the heavy-chain complementarity determining region 3 (CDR3) of E51 can efficiently bind gp120 in the absence of CD4 and neutralizes HIV-1 more effectively than peptides based on the CCR5 amino terminus.[94] X5 is a CD4i antibody selected from a phage-displayed library from a seropositive donor with a relatively high broadly neutralizing serum antibody titer, which neutralizes a number of virus isolates as Fab but has more limited and weaker neutralizing activity as IgG1.[95,96] These results provide proof-of-concept that the receptor-inducible gp120 epitopes elicit potent broadly cross-reactive neutralizing antibody responses in HIV-1-infected patients and could be potentially useful for development of vaccines and inhibitors. They also show that the size of the CD4i antibodies is inversely correlated with their ability to neutralize viruses, suggesting that further decreasing the antibody molecular size may be beneficial.

Cβ1, a humanized IgG chimeric mAb[97] originating from a murine IgG 0.5β,[98] is the first characterized mAb targeting V3 of gp120. In an animal study, it protected chimpanzee from HIV-1 infection during a 52-week observation period, providing direct evidence that anti-V3 loop antibodies can prevent an HIV-1 infection in the absence of other virus-specific immune responses.[97] After the discovery of Cβ1, other anti-V3 antibodies were also found. hNM01 is a humanized monoclonal antibody based on murine mAb mNM01 that neutralizes the virus by the activation of complement and the subsequent disruption of the viral envelope after binding to V3.[99,100] The results of a phase I clinical trial were somewhat encouraging but further evaluation with higher doses in larger numbers of patients is required.[101,102] KD-247,[103,104] a newer anti-V3 antibody, is currently being evaluated in a phase I clinical trial now.[105] Although the ability of anti-V3 mAbs to neutralize a significant proportion of HIV-1 isolates from different subtypes (clades) has remained controversial due to its high variability, more anti-V3 mAbs have been identified which display cross-clade neutralizing activity.[106,107] The evaluation in an animal study reveals that a significant proportion ofviruses can be neutralized by the newly identified anti-V3 mAbs such as HGN194.[108] These results further confirm that the anti-V3 mAbs can prevent infection from different clades, implying that V3 is still a useful target.

2G12, a carbohydrate-targeted bnmAb, was selected from 33 hybridomas producing human mAbs against HIV-1 which were established by cell fusion or EBV transformation.[23,109112] Unlike the conventional antibodies, 2G12 adopts a multivalent binding surface by using a unique domain-exchanged structure, in which the heavy-chain variable domains (VHs) of two antibody molecules are swapped. This extraordinary configuration provides two conventional antigen-combining sites and a third potential noncanonical binding site at the novel VH/VH interface for multivalent interactions with a conserved cluster of oligomannose type sugars on the surface of gp120.[113,114] 2G12 alone and in combination with other antibodies (antibody cocktail) exhibits neutralizing activity in animal experiments[115117] and clinical trials.[118120] Escape mutant analysis showed that the activity of 2G12 was crucial for the in vivo effect of the neutralizing antibody cocktail.[121] Interestingly, 2G12 could form dimers that are more potent than monomeric 2G12 in neutralization of various strains of HIV-1 in vitro.[122] The 2G12 dimer could elicit ADCC at a lower concentration compared with monomeric 2G12.[123] Dimeric 2G12, therefore, could be a potent prophylactic reagent against HIV-1 in vivo and be used as a part of an antibody cocktail to prevent HIV-1 infection.[124] Recently, a panel of new bnmAbs have been identified that are almost 10-fold more potent than VRC01, PG9, and PG16 (see below for PG9 and PG16), and 100-fold more potent than the conventional mAbs.[125] The crystal structure of one of the antibodies, PGT 128, complexed with a fully glycosylated gp120 outer domain at 3.25 Å reveals that the antibody penetrates the glycan shield and recognizes two conserved glycans as well as a short β-strand segment of the V3 loop, accounting for its high binding affinity and broad specificity.[126] However, their neutralizing breadth is relatively narrower than that of VRC01 when compared at concentrations that produce 50% inhibition (IC50s) of between 1–50 μg/mL. These data raise a general concern that further increasing antibody potency against certain isolates may result in a substantial decrease in neutralization breadth due to the high variability of the virus. It is therefore likely that an ideal potency and breadth could never be reached by using a single bnmAb.

PG9 and PG16 are two bnmAbs targeting conserved regions of the variable loops of the gp120 subunit preferentially expressed on trimeric Envs. They were selected from a high-throughput neutralization screen of antibody-containing culture supernatants of about 30 000 activated memory B cells from a clade A-infected African donor.[127] The epitopes defined by PG9 and PG16 are associated with the conserved regions of V1, V2, and V3 loops. They overlap with the CD4bs and possibly with the CoRbs, are sensitive to a loss of the glycan at N332 and an I165A substitution in gp120, but are distinct from that recognized by 2G12.[128,129] The crystal structure of V1/V2 in complex with PG9 has been solved, which is helpful for identification of conserved features that enable recognition of gp120.[130] Very recently, plasma bnmAbs specific for epitopes that include carbohydrates and are critically dependent on the glycan at position 332 of Env gp120 were found in CCR5-tropic SHIV-infected macaque.[131] These quaternary structure-specific antibodies appear to target antigenic variants of the same epitope, with neutralization breadth determined by the prevalence of recognized variants among circulating isolates, which provides important and novel information that will assist in improving the engineering of Env-based immunogens.[132134]

As described above, the formation of the fusion core in gp41 is one of the key steps in viral entry. The gp41 NHR structure is relatively conserved and has been successfully targeted by small molecule inhibitors. A mAb targeting the hydrophobic pocket situated in the NHR groove, designated D5, was identified that inhibited the infection of diverse HIV-1 clinical isolates, although relatively weakly.[135] m44, a mAb presumably targeting the CHR of gp41, can neutralize most of the 22 HIV-1 primary isolates from different clades as tested in assays based on infection of peripheral blood mononuclear cells (PBMCs) by replication-competent viruses.[136] Recently, a panel of anti-gp41 antibodies were also isolated from memory B cells of HIV-1-infected patients, which targeted different epitopes in gp41 and could neutralize tier-2 viruses.[137] These findings indicate that the conserved conformational epitopes on gp41 could be useful in the design of vaccine immunogens and as a target for therapeutics.

The identification of three bnmAbs targeting the MPER of gp41 (2F5,[138] Z13, and 4E10[139]) has implicated this region as a highly promising vaccine target and has, therefore, spurred interest in its structural characterization.[140146] In general, there are two distinct models of the binding patterns and disruption of HIV-1 MPER fusogenic functions by these antibodies. 2F5, like 4E10, induces large conformational changes in the MPER relative to the membrane. The difference is that 4E10 straddles the hinge and extracts residues W672 and F673 while 2F5 lifts up N-terminal residues to the hinge region by exposing L669 and W670 in MPER. In contrast, Z13e1,[147] a matured version of Z13, does not cause significant change in membrane orientation or conformation, but rather immobilizes the MPER hinge through extensive rigidifying surface contacts.[144] The difference in binding mechanism also leads to a difference in neutralization activities.[139] The animal studies showed that a triple combination of 2G12/2F5/4E10 could protect neonatal rhesus macaques from infection and no virus was detected in the plasma, PBMCs, or lymph nodes for more than 1 year.[148] Therefore, the same combination was also chosen for human clinical trials. In a phase I evaluation, seven individuals showed no drug-related adverse effects.[120,149] During the phase II clinical trial, chronically and acutely infected individuals were recruited.[121] These subjects were on antiretroviral treatment (ART) before antibody administration. After administration of these antibodies, the viral rebound was delayed in four of six acutely infected patients whereas only two of eight chronically infected patients showed a profound delay of rebound, suggesting that these bnmAbs could be more effective in early infection. Further evaluation of these three antibodies in a passive immunization trial indicates that the in vivo activities are likely due to direct neutralization or Fc receptor-mediated mechanisms, such as phagocytosis and ADCC, or both, but not due to CDC,[150] which is consistent with the previous study.[76] Recently, a new MPER-targeted mAb, m66.6, was reported that binds to essentially the same epitope as 2F5.[151] Interestingly, both the heavy and light chains of m66.6 are from different germlines and less mutated than 2F5, suggesting that neutralizing responses against the 2F5/m66.6 epitope can be elicited through different maturation pathways.

3.2. Receptor Molecules

Host receptor molecules are conserved and, therefore, antibodies targeting them could be more efficient than those against the Envs to overcome the high mutation frequency used by HIV-1 to escape from the human immune system surveillance.

A humanized mouse mAb, TNX-355 (ibalizumab), was developed against the D2 domain of human and rhesus CD4. TNX-355 in an IgG4 format inhibited HIV and simian immunodeficiency virus (SIV) replication in vitro and was safely administered to rhesus monkeys without depleting CD4+ T cells.[152156] A phase I human clinical trial with TNX-355 demonstrated that the antibody was well tolerated. A single dose of TNX-355 conferred a significant decrease in viral load and increase in CD4+ T cell counts in HIV-1-infected subjects.[157,158] TNX-355 is currently being developed by TaiMed Biologics, through licensing with Genetech, Inc, and a phase II trial is ongoing.[159]

The HIV-1 coreceptors, CCR5 or CXCR4, are also attractive targets for development of bnmAbs. A humanized mouse anti-CCR5 mAb, PRO 140, and its murine progenitor, PA14, exhibits potent and broad-spectrum inhibition of primary HIV-1 R5 isolates from different clades but does not affect CCR5 activity.[160,161] A phase I human clinical trial showed that PRO 140 given intravenously was generally well tolerated and exhibited potent, rapid, prolonged and dose-dependent antiviral activity in the subjects with early-stage HIV-1 infection.[162,163] Two phase II studies of the safety and pharmacokinetics of PRO 140 given intravenously and subcutaneously performed by Progenics Pharmaceuticals, Inc. are ongoing.[164,165]

4. Engineered Antibody Domains (eAds)

HIV-1 uses strategies to escape the neutralizing immune responses, one of which is steric occlusion of their highly conserved functionally important structures on Envs. We therefore have hypothesized that antibody fragments of smaller size, especially engineered antibody domains (eAds) [11–15 kDa], could be more effective than full-size antibodies.[166] Not only is the overall size of eAds much smaller than that of full-size antibodies but also their paratopes are concentrated over a smaller area so that eAds provide the capability of interacting with novel epitopes that are inaccessible to conventional antibodies or antibody fragments with paired light-chain variable domains (VLs) and VHs. eAds could also be more resistant to HIV-1 escape than full-size antibodies because they may bind to the epitopes which are more conserved as required for the maintenance of substantial binding to receptor or coreceptor. Moreover, eAds may have better binding kinetics (e.g. increased association rate) while recognizing and anchoring the physically obstructed epitopes on HIV-1 Envs.

In the following sections, we discuss the progress of the development of two types of eAds against HIV-1 including antibody variable domain-based (V domain-based) and antibody constant domain-based (C domain-based) eAds.

4.1. V Domain-Based eAds

About twenty years ago, an isolated mouse single VH was shown to maintain the binding activity to lysozymes, indicating that such domains are functional.[167] Later, it was found that a unique kind of antibody composed only of heavy chains is naturally formed in camelids (camels and llamas) [designated HCAbs] and cartilaginous fishes (wobbegong and nurse sharks) [designated Ig-NAR],[168170] suggesting their variable regions (referred to as VHHs in camelids and V-NAR in cartilaginous fish) could also recognize antigens as single domain fragments.[171] Therefore, a direction is the development of binders based on VHs or VLs from full-size antibodies. We term them ‘variable domain-based engineered antibody domains (V domain-based eAds)’.

m36 is the first reported V domain-based eAd based on a human antibody VH (~15 kDa) that targets a sterically restricted CD4i epitope and potently neutralizes genetically diverse HIV-1 isolates in vitro.[166] The structure of m36 was modeled by SWISS-MODEL[172174] and rendered by PyMOL (figure 3). In a humanized non-obese diabetic (NOD)/severe combined immunodeficiency (SCID)/γcnull mouse model,[176] m36.4, an affinity-matured version of m36, provided sterilizing protection in four of six animals against intrasplenic challenge with high-titer HIV-1 (>1000 TCID50s [50% median tissue culture infectious dose]) while extensive infection was detected in all four control animals.[177]

Fig. 3.

Fig. 3.

Models of engineered antibody domains based on V or C domains. m36 was selected from a library based on the heavy chain variable domain (VH) of a human IgG (pdb: 1HZH,[81,175] a model) while m1a1 was indentified from a library based on the second constant domain (CH2) scaffold.

Recently, several VHHs selected from libraries constructed from immunized camelids by HIV-1 Envs have been characterized as potent HIV-1 inhibitors by targeting the CD4bs or CXCR4.[178181] In addition, a novel eAd was identified from a llama immunized with a recombinant form of Nef, an HIV-1 nonstructural protein found in the cytoplasm of infected cells and in association with cellular membranes.[182] It binds to Nef with a high affinity and can inhibit critical biologic activities of Nef both in vitro and in vivo. This anti-Nef eAd may represent an efficient tool to elucidate the molecular functions of Nef in the virus life cycle and could help to develop new strategies for the control of AIDS.

4.2. C Domain-Based eAds

The second domain of the IgG, IgA, and IgD heavy chain constant regions, CH2, is unique in that it exhibits very weak carbohydrate-mediated interchain protein-protein interactions in contrast to the extensive interchain interactions that occur between the other domains. The expression of murine CH2 in bacteria, which do not support glycosylation, results in a monomeric protein.[183] It has been hypothesized that isolated CH2 (from IgG, IgA, and IgD) and CH3 (equivalent to CH2 in other antibody isotypes including IgE and IgM) could be used as a promising scaffold for eAd library construction due to their solubility and stability.[184] Moreover, they could offer additional favorable properties compared with V domain-based scaffolds because they contain binding sites or portions of binding sites for Fc receptors such as neonatal Fc receptor (FcRn) and Fc gamma receptors (FcγR), and complement C1q,[185190] which extend the half-life and mediate stability and effector functions in vivo. In line with this hypothesis is the finding that the half-life of human CH2 (about 70 hours) in rabbits is much longer than that of human CH3 and Fab (about 15 hours), and that human CH2 might activate the complement cascade.[191,192] We term the binders based on CH2 scaffold ‘constant domain-based engineered antibody domains (C domain-based eAds)’.

A previous study reported the construction of a large (5 × 1010 members) phage-displayed antibody library by using the human CH2 as a scaffold and mutating all residues in its two loops (BC and FG) to four residues (Y, A, D, or S). The library was panned with a gp120-sCD4 complex, and several binders against HIV-1 were selected.[193] The highest-affinity binder, m1a1, specifically recognizes a highly conserved CD4i epitope overlapping with that of m36 and can neutralize seven of nine HIV-1 isolates from different clades.[193] The structure of m1a1 was also modeled by SWISS-MODEL and rendered by PyMOL (figure 3).

However, the native CH2 domain has significantly lower thermal stability compared with other small scaffolds such as the 10th type III domain of human fibronectin (FN3),[183,194,195] and has a high probability of instability when engineered for binding to antigens and enhanced effector functions. We have recently generated a shortened human CH2 variant, m01s, by introducing an additional disulfide bond between CH2 strands A and G, and removing seven residues from the N-terminus, which showed not only increased stability but also enhanced binding to human FcRn.[196] A library was constructed by using m01s as a scaffold, from which a binder was identified that bound to HIV-1 MPER and could neutralize a panel of HIV-1 isolates (Gong et al., unpublished data). Because m01s binds to FcRn in a pH-dependent manner,[196] binders selected from this library might have a longer half-life in vivo compared with antibody variable domain-based binders.

Although some success in development of eAds as candidate therapeutics has already been achieved, there are still some issues that need to be addressed before eAds can be suitable for in vivo use. The camelid VHH and shark V-NAR domains are in general soluble and are stable in vitro.[197] However, for in vivo administration, humanization (or deimmunization) may be crucial to reduce immunogenicity. Compared with VHHs and V-NARs, other VH domains such as human VH domains may be relatively unstable and tend to aggregate, and their unfolding is normally irreversible because of the lack of VL partners. Therefore, stabilization is required to make these domains aggregation-resistant and unfolding-reversible.[198200] It has been found that V domain-based antibodies had significantly different reaction kinetics compared with the fragments consisting of paired heavy and light chain domains, which likely leads to lower affinity to their targets.[201] Accordingly, further maturation may be needed to achieve high affinity. In addition, the half-lives of V domain-based eAds in circulation are short and they do not have biological effector functions, as has been described for other antibody fragments, including Fabs and single-chain variable fragments (scFvs). Some methods,[202] including introduction of serum albumin binding peptide,[203,204] conjugation of polyethylene glycol (PEG),[205] and fusion with anti-serum albumin V domain-based eAd,[206] were developed to extend the half-life, but potential risks might exist. The C domain-based eAds need to be further characterized for their biophysical and biological properties.

5. Increasing Potency

HIV-1 uses multiple strategies to escape from the host immune system surveillance. These include rapid generation of viral variants, high variability of Envs, steric occlusion of conserved neutralizing epitopes on Envs that limits access of bnmAbs, mimicry of human self-antigens by conserved neutralizing epitopes on Envs that results in polyspecificity and autoreactivity of broadly neutralizing antibodies, and encoding of immunogenic conserved non-neutralizing or infection-enhancement eptiopes on Envs. Although a number of bnmAbs have been identified, some of which show potent and broad neutralizing activity against HIV-1 in vitro and in animal models, we are still far away from the use of these antibodies in the treatment of HIV-infected patients. Improvement of the efficacy of the existing bnmAbs and identification of novel more potent antibodies are still urgent tasks.

5.1. Design of Antigens and Novel Targets

Some (10–25%) HIV-1-infected individuals develop broadly neutralizing sera, from which bnmAbs have been successfully isolated by using recombinantly expressed Envs. However, the structural differences between the isolated Env and the native Env on the viral spike may result in an unpredictable loss of antibodies that preferentially recognize the native Env structures when the isolated Env is used. The design of soluble versions of trimeric Env that display structural and functional properties similar to those observed on intact viruses is highly desirable.[207] Therefore, efforts have been made to preserve or re-construct functional trimeric Env, and immunization with this resulted in improved elicitation of cross-reactive neutralizing antibodies against primary isolates. These trimeric Env could be useful for selection of novel potent neutralizing antibodies. HIV-1 entry requires extensive conformational changes in both gp120 and gp41, leading to the formation of highly conserved structures. These structures, however, are either sterically hidden or only transiently exposed during entry. Therefore, stabilized fusion intermediates could be valuable antigens for antibody selection. Previous studies have demonstrated efficient isolation of X5 and m36 against the CD4i epitopes on gp120 by using gp120-sCD4 complexes. Immediately prior to fusion, gp41 adopts a prehairpin intermediate state in which the N- (N-trimer) and C-terminal (C-trimer) trimers of gp41 are exposed. Neutralization of HIV-1 by IgGs 2F5 and 4E10 could be potentiated by the addition of a peptide that holds gp41 in this state. Both the N- and C-trimers have been successfully targeted by peptide inhibitors, suggesting that they could also be targeted by antibodies. A recent study opened a new avenue for design of antigens for selection of antibodies against specific epitopes of interest.[83] HIV-1 Env is a large protein recognized by several types of antibodies including isolate-specific non-neutralizing antibodies, some of which compete with cross-reactive neutralizing antibodies in binding to Envs. Therefore, the use of original Env for selection may not be efficient. Based on this concept, a modified gp120, RSC3, was designed that specifically reacts with antibodies directed against the CD4bs. The use of RSC3 led to the discovery of exceptionally potent and broad antibodies, VRC01 and VRC02.[83,84] This approach can be extended to select novel bnmAbs against other highly conserved, functionally important epitopes. In addition, further characterization of core epitopes on Env that is conserved and indispensable for viral infectivity could also be helpful for design of antigens.[208]

Besides the widely used antigens (i.e. Env, receptor and coreceptor) for HIV-1 bnmAb selection, other new interesting targets have been or are being identified with the increase in our knowledge of HIV-1. As mentioned above, inhibition of the activity of PDI on the surface of CD4+ cells can prevent the activation of gp41 and HIV-1 entry into target cells. PDI catalyzes the formation of a disulfide bond between the cysteines in gp120 after formation of PDI-CD4-gp120 complex. Therefore, PDI and PDI-CD4-gp120 complex are potentially useful targets for antibodies that could block the enzymatic activity of PDI or its interactions with CD4-gp120 or the subsequent fusion process. Such antibodies have been reported that could potently inhibit HIV-1 infection.[47,50,53,209]

5.2. Libraries

Highly diverse antibody libraries have become important sources for selection of antibodies with high affinity and novel properties. Combinatorial strategies provide efficient ways of creating antibody libraries containing a large number of individual clones. These strategies include the reassembly of naturally occurring genes encoding the heavy and light chains from either immune or nonimmune B cell sources, and introduction of synthetic diversity to either the framework regions or the CDRs of the variable domains of antibodies. Phage display and other display technologies (e.g. yeast display and ribosome display) have already been used to construct large antibody libraries (up to 1011 or higher), from which various useful antibodies could be selected through several rounds of biopanning and screening. Compared with the traditional method based on hybridoma, these library methodologies are much more efficient. However, natural antibody repertoires are subject to change; those harvested at a specific time point for library construction cannot cover all theoretical genetic diversity. Moreover, antibodies from such libraries may not represent the native antibodies because the heavy and light chains are randomly combined. Although the preservation of cognate pairing between heavy and light chains may not be critical for therapeutic antibodies, it is relevant to studying antibody maturation pathways. These leave scope for antibody libraries based on the conventional designs to be improved.

The advent of the high-throughput next-generation 454 sequencing technology has allowed direct observation of millions of diverse antibody variants that are generated by the immune systems at different time points and have survived positive, negative, and antigen-driven selection. Recently, large numbers of antibodies from humans have been sequenced and analyzed using this method.[210216] Based on a collection of antibody sequences from the literature, large libraries could therefore be designed and directly synthesized through a novel gene synthesis technology that enables not only precise determination of diversity but also positional control of amino acid composition and incorporation frequencies.[217] The obtained library could reach a diversity beyond that of the donor-derived natural repertoire due to recombination.

The single-cell PCR methodology allows for construction of libraries with original pairs of VH and VL. This methodology is particularly useful in exploration of the natural human antibody responses. Although progress has been made,[218] some technical difficulties remain to be solved.

5.3. Screening Methods

The selection of a large number of anti-HIV-1 bnmAbs from HIV-1 envelope-binding memory B cells from HIV-1 infected patients which show different types of broad activity exhibited by b12, 2F5, 4E10, or 2G12 indicates the possibility of finding novel bnmAbs by direct B-cell screening.[219] The successful identification of PG9 and PG16 by functional high-throughput screening of B cells sets a good example for development of new approaches to isolate more potent broadly neutralization antibodies.[127] This approach is particularly useful to identify broadly neutralizing antibodies that bind poorly to recombinant forms of Env. With this functional screening method, many new bnmAbs have been recently identified which show almost 10-fold higher potency than the previously described VRC01, PG9, and PG16, and are 100-fold more potent than the prototype HIV-1 bnmAbs such as b12.[220] The discovery of VRC01 and VRC02 by using an antigenically resurfaced HIV-1 gp120, in which the neutralizing surface of CD4bs was preserved while other antigenic regions were eliminated, is another good example for efficient isolation of antibodies targeting a specific surface area of Env.

All existing bnmAbs are isolated from HIV-1-infected individuals, whose sera exhibit broadly neutralizing activity. Due to the limitations of the current screening methods, however, the most potent and broadest bnmAbs in the patients may not be selected. Recent studies show that all HIV-1 bnmAbs are heavily mutated compared with their corresponding germlines and antibodies with high degrees of somatic hypermutation (SHM) generally exhibit higher potency and cross-reactivity than their less mutated intermediates.[151,215,221,222] Therefore, the large-scale sequencing of potentially a whole antibody repertoire offers another novel approach to select possibly more potent antibodies based on their sequence similarities and levels of SHM compared with the known bnmAbs selected from the same patients via conventional methods.[216]

5.4. Multifunctional Antibody-Based Fusion Proteins

It has been proposed that a combination of multiple entry inhibitors with the same or different mechanisms of action would increase the antiviral potency and durability to resistance, and provide a broader coverage of the virus. PRO 542 (CD4-IgG2) is a fusion protein comprising human IgG2 in which the variable fragment (Fv) portions of both heavy and light chains have been replaced by the D1 and D2 domains of human CD4, which can protect the human-peripheral blood lymphocyte (PBL)-SCID mouse model[223] from infection by primary isolates of HIV-1 in vivo.[224,225] It has also been shown that PRO 542 and T-20 are potently synergistic in blocking virus-cell and cell-cell fusion.[226] In a clinical trial, both single and multiple doses of PRO 542 were well tolerated.[227] Statistically significant acute reductions in HIV-1 RNA levels were observed across all study patients including children and adults, and greater antiviral effects were observed in the cohort of patients with more advanced HIV-1 disease.[228,229] Further evaluation of PRO 542 was performed by Progenics Pharmaceuticals, Inc ().[230] Fusion of two-domain sCD4 to 17b exhibited very broad and potent activity superior to that of b12, 2F5, 4E10, and 2G12 in neutralization against a large panel of isolates.[231] Combinations of other CD4i mAbs (e.g. E51 and m36) with sCD4 and antibody Fc also resulted in increased neutralizing activities.[232,233] The fully functional soluble stable single-domain sCD4 was also generated, which could be used to decrease the molecular size of large fusion proteins.[234] A novel, bifunctional HIV-1 entry inhibitor by combining the anti-CD4 mAb 6314 with a fusion inhibitor similar to T-651 (anti-CD4 mAb based bi-functional fusion inhibitor, CD4-BFFI) was described that showed higher antiviral potency compared with the fusion inhibitor T-651 or the anti-CD4 mAb 6314 used independently.[235,236] Importantly, CD4-BFFI can inhibit all the tested HIV-1 strains among which many strains are only partially inhibited by 6314. This CD4-BFFI also retains antiviral potency against virus strains resistant to two fusion inhibitors, a CCR5 antagonist and an anti-CCR5 mAb. Besides the fusion of antibodies with other non-Ig molecules, the bispecific antibodies through connection of scFvs with IgGs targeting two alternative docking sites of CCR5-tropic HIV strains on CCR5 showed 18- to 57-fold increased antiviral activities compared with the parental antibodies.[237] In addition, one prototypic tetravalent CCR5 antibody, CCR5–2320, had antiviral activity against virus strains resistant to the single parental antibodies.[237] These results indicate that bispecific antibodies with tetra-valence produced by physical linkage of two CCR5 antibodies targeting different epitopes on CCR5 have enhanced antiviral potency against wild-type and CCR5 antibody-resistant HIV-1 strains.

In addition to direct fusion of two independent antibodies, some new approaches have been developed to facilitate the assembly of bi-functional antibodies without significantly increasing antibody molecular size. For example, based on the knobs-into-holes technology that enables heterodimerization of the heavy chains, correct association of the light chains and their cognate heavy chains is achieved by exchange of heavy-chain and light-chain domains within Fab of one half of the bifunctional antibody.[238,239] Through this method, the bispecific anti–HIV-1 antibodies (BiAbs) that can bind bivalently by virtue of one scFv arm that binds to gp120 and a second arm to the gp41 subunit of gp160 were engineered by heteroligation, which showed the bivalent could enhance neutralization compared with the parental antibodies.[240,241]

6. Conclusions and Perspectives

The continued spread of HIV-1 worldwide and, in particular, in sub-Saharan Africa, where an estimated 22 million people are currently living with HIV-1/AIDS, underscores the urgent need for preventative or therapeutic antibodies. However, despite nearly 25 years of intense international research, an efficient vaccine or antibody is not yet available for clinical use. Although antibodies with broadly neutralizing activities can confer sterilizing protection against infection in animal models, some of them have failed for the therapy of HIV-1 infection in humans, and the clinical trials of others are still ongoing. None have entered phase III trials (summary in table I). This is in contrast to the clinical benefits provided by the currently approved therapeutic antibodies, such as those against cancer and immune disorders. There are no such antibodies against HIV-1, most likely because they are not capable of circumventing the escape mechanisms used by the virus, among other reasons. Thus, further studies are needed that could provide valuable insights into the interplay between the virus and the human immune system and how we could provide clinical benefits for HIV-1-infected humans by using antibodies. Finally, antibodies undoubtedly could play a critical role for reaching the ultimate goal – eradication of the virus.

Acknowledgments

We thank members of our group for useful discussions. This article was supported by the Intramural Research Program of the NIH, NCI, CCR, and the IATAP of the NIH.

References

  • 1.Ho DD. Time to hit HIV, early and hard. N Engl J Med 1995; 333: 450–1 [DOI] [PubMed] [Google Scholar]
  • 2.Harrington M, Carpenter CC. Hit HIV-1 hard, but only when necessary. Lancet 2000; 355: 2147–52 [DOI] [PubMed] [Google Scholar]
  • 3.Jochmans D Novel HIV-1 reverse transcriptase inhibitors. Virus Res 2008; 134: 171–85 [DOI] [PubMed] [Google Scholar]
  • 4.Saag MS. Treatment of HIV infection: the antiretroviral nucleoside analogues. Nucleoside analogues: adverse effects. Hosp Pract (Off Ed) 1992; 27 Suppl. 2: 26–36 [DOI] [PubMed] [Google Scholar]
  • 5.McDonald CK, Kuritzkes DR. Human immunodeficiency virus type 1 protease inhibitors. Arch Intern Med 1997; 157: 951–9 [PubMed] [Google Scholar]
  • 6.Richman DD. Resistance of clinical isolates of human immunodeficiency virus to antiretroviral agents. Antimicrob Agents Chemother 1993; 37: 1207–13 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Condra JH, Schleif WA, Blahy OM, et al. In vivo emergence of HIV-1 variants resistant to multiple protease inhibitors. Nature 1995; 374: 569–71 [DOI] [PubMed] [Google Scholar]
  • 8.Baldwin CE, Sanders RW, Deng Y, et al. Emergence of a drug-dependent human immunodeficiency virus type 1 variant during therapy with the T20 fusion inhibitor. J Virol 2004; 78: 12428–37 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Dimitrov DS, Marks JD. Therapeutic antibodies: current state and future trends – is a paradigm change coming soon? Methods Mol Biol 2009; 525: 1–27, xiii [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Wei X, Decker JM, Wang S, et al. Antibody neutralization and escape by HIV-1. Nature 2003; 422: 307–12 [DOI] [PubMed] [Google Scholar]
  • 11.Wu H, Kwong PD, Hendrickson WA. Dimeric association and segmental variability in the structure of human CD4. Nature 1997; 387: 527–30 [DOI] [PubMed] [Google Scholar]
  • 12.NCBI. Structure Of T-cell surface glycoprotein Cd4, monoclinic crystal form [online]. Available from URL: http://www.ncbi.nlm.nih.gov/Structure/mmdb/mmdbsrv.cgi?uid=6042 [Accessed 2012 Mar 09]
  • 13.Leonard CK, Spellman MW, Riddle L, et al. Assignment of intrachain disulfide bonds and characterization of potential glycosylation sites of the type 1 recombinant human immunodeficiency virus envelope glycoprotein (gp120) expressed in Chinese hamster ovary cells. J Biol Chem 1990; 265: 10373–82 [PubMed] [Google Scholar]
  • 14.Douglas NW, Munro GH, Daniels RS. HIV/SIV glycoproteins: structure-function relationships. J Mol Biol 1997; 273: 122–49 [DOI] [PubMed] [Google Scholar]
  • 15.Kwong PD, Wyatt R, Majeed S, et al. Structures of HIV-1 gp120 envelope glycoproteins from laboratory-adapted and primary isolates. Structure 2000; 8: 1329–39 [DOI] [PubMed] [Google Scholar]
  • 16.NCBI. HIV-1 Hxbc2 Gp120 envelope glycoprotein complexed with CD4 and induced neutralizing antibody 17b [online]. Available from URL: http://www.ncbi.nlm.nih.gov/Structure/mmdb/mmdbsrv.cgi?uid=14983 [Accessed 2012 Mar 23]
  • 17.Hwang SS, Boyle TJ, Lyerly HK, et al. Identification of the envelope V3 loop as the primary determinant of cell tropism in HIV-1. Science 1991; 253: 71–4 [DOI] [PubMed] [Google Scholar]
  • 18.Hwang SS, Boyle TJ, Lyerly HK, et al. Identification of envelope V3 loop as the major determinant of CD4 neutralization sensitivity of HIV-1. Science 1992; 257: 535–7 [DOI] [PubMed] [Google Scholar]
  • 19.Sullivan N, Sun Y, Binley J, et al. Determinants of human immunodeficiency virus type 1 envelope glycoprotein activation by soluble CD4 and monoclonal antibodies. J Virol 1998; 72: 6332–8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Javaherian K, Langlois AJ, LaRosa GJ, et al. Broadly neutralizing antibodies elicited by the hypervariable neutralizing determinant of HIV-1. Science 1990; 250: 1590–3 [DOI] [PubMed] [Google Scholar]
  • 21.Gao F, Weaver EA, Lu Z, et al. Antigenicity and immunogenicity of a synthetic human immunodeficiency virus type 1 group m consensus envelope glycoprotein. J Virol 2005; 79: 1154–63 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Hansen JE, Clausen H, Hu SL, et al. An O-linked carbohydrate neutralization epitope of HIV-1 gp 120 is expressed by HIV-1 env gene recombinant vaccinia virus. Arch Virol 1992; 126: 11–20 [DOI] [PubMed] [Google Scholar]
  • 23.Trkola A, Purtscher M, Muster T, et al. Human monoclonal antibody 2G12 defines a distinctive neutralization epitope on the gp120 glycoprotein of human immunodeficiency virus type 1. J Virol 1996; 70: 1100–8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Zhu P, Liu J, Bess J Jr, et al. Distribution and three-dimensional structure of AIDS virus envelope spikes. Nature 2006; 441: 847–52 [DOI] [PubMed] [Google Scholar]
  • 25.Liu J, Bartesaghi A, Borgnia MJ, et al. Molecular architecture of native HIV-1 gp120 trimers. Nature 2008; 455: 109–13 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Earl PL, Broder CC, Doms RW, et al. Epitope map of human immunodeficiency virus type 1 gp41 derived from 47 monoclonal antibodies produced by immunization with oligomeric envelope protein. J Virol 1997; 71: 2674–84 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Kowalski M, Potz J, Basiripour L, et al. Functional regions of the envelope glycoprotein of human immunodeficiency virus type 1. Science 1987; 237: 1351–5 [DOI] [PubMed] [Google Scholar]
  • 28.Gallaher WR. Detection of a fusion peptide sequence in the transmembrane protein of human immunodeficiency virus. Cell 1987; 50: 327–8 [DOI] [PubMed] [Google Scholar]
  • 29.Gerber D, Pritsker M, Gunther-Ausborn S, et al. Inhibition of HIV-1 envelope glycoprotein-mediated cell fusion by a DL-amino acid-containing fusion peptide: possible recognition of the fusion complex. J Biol Chem 2004; 279: 48224–30 [DOI] [PubMed] [Google Scholar]
  • 30.Munch J, Standker L, Adermann K, et al. Discovery and optimization of a natural HIV-1 entry inhibitor targeting the gp41 fusion peptide. Cell 2007; 129: 263–75 [DOI] [PubMed] [Google Scholar]
  • 31.Chan DC, Fass D, Berger JM, et al. Core structure of gp41 from the HIV envelope glycoprotein. Cell 1997; 89: 263–73 [DOI] [PubMed] [Google Scholar]
  • 32.Weissenhorn W, Dessen A, Harrison SC, et al. Atomic structure of the ectodomain from HIV-1 gp41. Nature 1997; 387: 426–30 [DOI] [PubMed] [Google Scholar]
  • 33.Lu M, Kim PS. A trimeric structural subdomain of the HIV-1 transmembrane glycoprotein. J Biomol Struct Dyn 1997; 15: 465–71 [DOI] [PubMed] [Google Scholar]
  • 34.Chan DC, Chutkowski CT, Kim PS. Evidence that a prominent cavity in the coiled coil of HIV type 1 gp41 is an attractive drug target. Proc Natl Acad Sci U S A 1998; 95: 15613–7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Salzwedel K, West JT, Hunter E. A conserved tryptophan-rich motif in the membrane-proximal region of the human immunodeficiency virus type 1 gp41 ectodomain is important for Env-mediated fusion and virus infectivity. J Virol 1999; 73: 2469–80 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Suarez T, Gallaher WR, Agirre A, et al. Membrane interface-interacting sequences within the ectodomain of the human immunodeficiency virus type 1 envelope glycoprotein: putative role during viral fusion. J Virol 2000; 74: 8038–47 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Klatzmann D, Champagne E, Chamaret S, et al. T-lymphocyte T4 molecule behaves as the receptor for human retrovirus LAV. Nature 1984; 312: 767–8 [DOI] [PubMed] [Google Scholar]
  • 38.McDougal JS, Kennedy MS, Sligh JM, et al. Binding of HTLV-III/LAV to T4+ T cells by a complex of the 110K viral protein and the T4 molecule. Science 1986; 231: 382–5 [DOI] [PubMed] [Google Scholar]
  • 39.Dalgleish AG, Beverley PC, Clapham PR, et al. The CD4 (T4) antigen is an essential component of the receptor for the AIDS retrovirus. Nature 1984; 312: 763–7 [DOI] [PubMed] [Google Scholar]
  • 40.Robey E, Axel R. CD4: collaborator in immune recognition and HIV infection. Cell 1990; 60: 697–700 [DOI] [PubMed] [Google Scholar]
  • 41.Raff MC. Role of thymus-derived lymphocytes in the secondary humoral immune response in mice. Nature 1970; 226: 1257–8 [DOI] [PubMed] [Google Scholar]
  • 42.Clark SJ, Jefferies WA, Barclay AN, et al. Peptide and nucleotide sequences of rat CD4 (W3/25) antigen: evidence for derivation from a structure with four immunoglobulin-related domains. Proc Natl Acad Sci U S A 1987; 84: 1649–53 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Maddon PJ, Molineaux SM, Maddon DE, et al. Structure and expression of the human and mouse T4 genes. Proc Natl Acad Sci U S A 1987; 84: 9155–9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Arthos J, Deen KC, Chaikin MA, et al. Identification of the residues in human CD4 critical for the binding of HIV. Cell 1989; 57: 469–81 [DOI] [PubMed] [Google Scholar]
  • 45.Ryu SE, Kwong PD, Truneh A, et al. Crystal structure of an HIV-binding recombinant fragment of human CD4. Nature 1990; 348: 419–26 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Lara HH, Ixtepan-Turrent L, Garza-Trevino EN, et al. Antiviral propierties of 5,5’-dithiobis-2-nitrobenzoic acid and bacitracin against T-tropic human immunodeficiency virus type 1. Virol J 2011; 8: 137. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Ryser HJ, Levy EM, Mandel R, et al. Inhibition of human immunodeficiency virus infection by agents that interfere with thiol-disulfide interchange upon virus-receptor interaction. Proc Natl Acad Sci U S A 1994; 91: 4559–63 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Gallina A, Hanley TM, Mandel R, et al. Inhibitors of protein-disulfide isomerase prevent cleavage of disulfide bonds in receptor-bound glycoprotein 120 and prevent HIV-1 entry. J Biol Chem 2002; 277: 50579–88 [DOI] [PubMed] [Google Scholar]
  • 49.Barbouche R, Miquelis R, Jones IM, et al. Protein-disulfide isomerase-mediated reduction of two disulfide bonds of HIV envelope glycoprotein 120 occurs post-CXCR4 binding and is required for fusion. J Biol Chem 2003; 278: 3131–6 [DOI] [PubMed] [Google Scholar]
  • 50.Markovic I, Stantchev TS, Fields KH, et al. Thiol/disulfide exchange is a prerequisite for CXCR4-tropic HIV-1 envelope-mediated T-cell fusion during viral entry. Blood 2004; 103: 1586–94 [DOI] [PubMed] [Google Scholar]
  • 51.Gowthaman U, Jayakanthan M, Sundar D. Molecular docking studies of dithionitrobenzoic acid and its related compounds to protein disulfide isomerase: computational screening of inhibitors to HIV-1 entry. BMC Bioinformatics 2008; 9 Suppl 12: S14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Wang Z, Zhou Z, Guo ZY, et al. Snapshot of the interaction between HIV envelope glycoprotein 120 and protein disulfide isomerase. Acta Biochim Biophys Sin (Shanghai) 2010; 42: 358–62 [DOI] [PubMed] [Google Scholar]
  • 53.Fenouillet E, Barbouche R, Courageot J, et al. The catalytic activity of protein disulfide isomerase is involved in human immunodeficiency virus envelope-mediated membrane fusion after CD4 cell binding. J Infect Dis 2001; 183: 744–52 [DOI] [PubMed] [Google Scholar]
  • 54.Bi S, Hong PW, Lee B, et al. Galectin-9 binding to cell surface protein disulfide isomerase regulates the redox environment to enhance T-cell migration and HIV entry. Proc Natl Acad Sci U S A 2011; 108: 10650–5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Samson M, Labbe O, Mollereau C, et al. Molecular cloning and functional expression of a new human CC-chemokine receptor gene. Biochemistry 1996; 35: 3362–7 [DOI] [PubMed] [Google Scholar]
  • 56.Deng H, Liu R, Ellmeier W, et al. Identification of a major co-receptor for primary isolates of HIV-1. Nature 1996; 381: 661–6 [DOI] [PubMed] [Google Scholar]
  • 57.Dragic T, Litwin V, Allaway GP, et al. HIV-1 entry into CD4+ cells is mediated by the chemokine receptor CC-CKR-5. Nature 1996; 381: 667–73 [DOI] [PubMed] [Google Scholar]
  • 58.Choe H, Farzan M, Sun Y, et al. The beta-chemokine receptors CCR3 and CCR5 facilitate infection by primary HIV-1 isolates. Cell 1996; 85: 1135–48 [DOI] [PubMed] [Google Scholar]
  • 59.Doranz BJ, Rucker J, Yi Y, et al. A dual-tropic primary HIV-1 isolate that uses fusin and the beta-chemokine receptors CKR-5, CKR-3, and CKR-2b as fusion cofactors. Cell 1996; 85: 1149–58 [DOI] [PubMed] [Google Scholar]
  • 60.Alkhatib G, Combadiere C, Broder CC, et al. CC CKR5: a RANTES, MIP-1alpha, MIP-1beta receptor as a fusion cofactor for macrophage-tropic HIV-1. Science 1996; 272: 1955–8 [DOI] [PubMed] [Google Scholar]
  • 61.Feng Y, Broder CC, Kennedy PE, et al. HIV-1 entry cofactor: functional cDNA cloning of a seven-transmembrane, G protein-coupled receptor. Science 1996; 272: 872–7 [DOI] [PubMed] [Google Scholar]
  • 62.Reeves JD, Hibbitts S, Simmons G, et al. Primary human immunodeficiency virus type 2 (HIV-2) isolates infect CD4-negative cells via CCR5 and CXCR4: comparison with HIV-1 and simian immunodeficiency virus and relevance to cell tropism in vivo. J Virol 1999; 73: 7795–804 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Zerhouni B, Nelson JA, Saha K. Isolation of CD4-independent primary human immunodeficiency virus type 1 isolates that are syncytium inducing and acutely cytopathic for CD8+ lymphocytes. J Virol 2004; 78: 1243–55 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Zerhouni B, Nelson JA, Saha K. CXCR4-dependent infection of CD8+, but not CD4+, lymphocytes by a primary human immunodeficiency virus type 1 isolate. J Virol 2004; 78: 12288–96 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Decker JM, Bibollet-Ruche F, Wei X, et al. Antigenic conservation and immunogenicity of the HIV coreceptor binding site. J Exp Med 2005; 201: 1407–19 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Stephens JC, Reich DE, Goldstein DB, et al. Dating the origin of the CCR5-Delta32 AIDS-resistance allele by the coalescence ofhaplotypes. Am J Hum Genet 1998; 62: 1507–15 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Libert F, Cochaux P, Beckman G, et al. The deltaccr5 mutation conferring protection against HIV-1 in Caucasian populations has a single and recent origin in Northeastern Europe. Hum Mol Genet 1998; 7: 399–406 [DOI] [PubMed] [Google Scholar]
  • 68.Burton DR, Barbas CF 3rd, Persson MA, et al. A large array of human monoclonal antibodies to type 1 human immunodeficiency virus from combinatorial libraries of asymptomatic seropositive individuals. Proc Natl Acad Sci U S A. 1991; 88: 10134–7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Roben P, Moore JP, Thali M, et al. Recognition properties of a panel of human recombinant Fab fragments to the CD4 binding site of gp120 that show differing abilities to neutralize human immunodeficiency virus type 1. J Virol 1994; 68: 4821–8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Burton DR, Pyati J, Koduri R, et al. Efficient neutralization of primary isolates of HIV-1 by a recombinant human monoclonal antibody. Science 1994; 266: 1024–7 [DOI] [PubMed] [Google Scholar]
  • 71.Binley JM, Wrin T, Korber B, et al. Comprehensive cross-clade neutralization analysis of a panel of anti-human immunodeficiency virus type 1 monoclonal antibodies. J Virol 2004; 78: 13232–52 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Parren PW, Marx PA, Hessell AJ, et al. Antibody protects macaques against vaginal challenge with a pathogenic R5 simian/human immunodeficiency virus at serum levels giving complete neutralization in vitro. J Virol 2001; 75: 8340–7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Veazey RS, Shattock RJ, Pope M, et al. Prevention of virus transmission to macaque monkeys by a vaginally applied monoclonal antibody to HIV-1 gp120. Nat Med 2003; 9: 343–6 [DOI] [PubMed] [Google Scholar]
  • 74.Hessell AJ, Poignard P, Hunter M, et al. Effective, low-titer antibody protection against low-dose repeated mucosal SHIV challenge in macaques. Nat Med 2009; 15: 951–4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Hezareh M, Hessell AJ, Jensen RC, et al. Effector function activities of a panel of mutants of a broadly neutralizing antibody against human immunodeficiency virus type 1. J Virol 2001; 75: 12161–8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Hessell AJ, Hangartner L, Hunter M, et al. Fc receptor but not complement binding is important in antibody protection against HIV. Nature 2007; 449: 101–4 [DOI] [PubMed] [Google Scholar]
  • 77.Posner MR, Hideshima T, Cannon T, et al. An IgG human monoclonal antibody that reacts with HIV-1/GP120, inhibits virus binding to cells, and neutralizes infection. J Immunol 1991; 146: 4325–32 [PubMed] [Google Scholar]
  • 78.Barbas CF 3rd, Collet TA, Amberg W, et al. Molecular profile of an antibody response to HIV-1 as probed by combinatorial libraries. J Mol Biol 1993; 230: 812–23 [DOI] [PubMed] [Google Scholar]
  • 79.Zhang MY, Xiao X, Sidorov IA, et al. Identification and characterization of a new cross-reactive human immunodeficiency virus type 1-neutralizing human monoclonal antibody. J Virol 2004; 78: 9233–42 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Zhang MY, Shu Y, Phogat S, et al. Broadly cross-reactive HIV neutralizing human monoclonal antibody Fab selected by sequential antigen panning ofa phage display library. J Immunol Methods 2003; 283: 17–25 [DOI] [PubMed] [Google Scholar]
  • 81.Saphire EO, Parren PW, Pantophlet R, et al. Crystal structure of a neutralizing human IGG against HIV-1: a template for vaccine design. Science 2001; 293: 1155–9 [DOI] [PubMed] [Google Scholar]
  • 82.Chen L, Kwon YD, Zhou T, et al. Structural basis of immune evasion at the site of CD4 attachment on HIV-1 gp120. Science 2009; 326: 1123–7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Wu X, Yang ZY, Li Y, et al. Rational design of envelope identifies broadly neutralizing human monoclonal antibodies to HIV-1. Science 2010; 329: 856–61 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Zhou T, Georgiev I, Wu X, et al. Structural basis for broad and potent neutralization ofHIV-1 by antibody VRC01. Science 2010; 329: 811–7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Diskin R, Scheid JF, Marcovecchio PM, et al. Increasing the potency and breadth of an HIV antibody by using structure-based rational design. Science 2011; 334: 1289–93 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Scheid JF, Mouquet H, Ueberheide B, et al. Sequence and structural convergence of broad and potent HIV antibodies that mimic CD4 binding. Science 2011; 333: 1633–7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Thali M, Moore JP, Furman C, et al. Characterization of conserved human immunodeficiency virus type 1 gp120 neutralization epitopes exposed upon gp120-CD4 binding. J Virol 1993; 67: 3978–88 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Wu L, Gerard NP, Wyatt R, et al. CD4-induced interaction of primary HIV-1 gp120 glycoproteins with the chemokine receptor CCR-5. Nature 1996; 384: 179–83 [DOI] [PubMed] [Google Scholar]
  • 89.Kwong PD, Wyatt R, Robinson J, et al. Structure of an HIV gp120 envelope glycoprotein in complex with the CD4 receptor and a neutralizing human antibody. Nature 1998; 393: 648–59 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Montefiori DC, Hill TS, Vo HT, et al. Neutralizing antibodies associated with viremia control in a subset of individuals after treatment of acute human immunodeficiency virus type 1 infection. J Virol 2001; 75: 10200–7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Choe H, Li W, Wright PL, et al. Tyrosine sulfation of human antibodies contributes to recognition of the CCR5 binding region of HIV-1 gp120. Cell 2003; 114: 161–70 [DOI] [PubMed] [Google Scholar]
  • 92.Xiang SH, Farzan M, Si Z, et al. Functional mimicry of a human immunodeficiency virus type 1 coreceptor by a neutralizing monoclonal antibody. J Virol 2005; 79: 6068–77 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Huang CC, Lam SN, Acharya P, et al. Structures of the CCR5 N terminus and of a tyrosine-sulfated antibody with HIV-1 gp120 and CD4. Science 2007; 317: 1930–4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Dorfman T, Moore MJ, Guth AC, et al. A tyrosine-sulfated peptide derived from the heavy-chain CDR3 region of an HIV-1-neutralizing antibody binds gp120 and inhibits HIV-1 infection. J Biol Chem 2006; 281: 28529–35 [DOI] [PubMed] [Google Scholar]
  • 95.Moulard M, Phogat SK, Shu Y, et al. Broadly cross-reactive HIV-1-neutralizing human monoclonal Fab selected for binding to gp120-CD4-CCR5 complexes. Proc Natl Acad Sci U S A 2002; 99: 6913–8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Labrijn AF, Poignard P, Raja A, et al. Access of antibody molecules to the conserved coreceptor binding site on glycoprotein gp120 is sterically restricted on primary human immunodeficiency virus type 1. J Virol 2003; 77: 10557–65 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Emini EA, Schleif WA, Nunberg JH, et al. Prevention of HIV-1 infection in chimpanzees by gp120 V3 domain-specific monoclonal antibody. Nature 1992; 355: 728–30 [DOI] [PubMed] [Google Scholar]
  • 98.Matsushita S, Robert-Guroff M, Rusche J, et al. Characterization of a human immunodeficiency virus neutralizing monoclonal antibody and mapping of the neutralizing epitope. J Virol 1988; 62: 2107–14 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Nakamura M, Sasaki H, Terada M, et al. Complement-dependent virolysis of HIV-1 with monoclonal antibody NM-01. AIDS Res Hum Retroviruses 1993; 9: 619–26 [DOI] [PubMed] [Google Scholar]
  • 100.Nakamura M, Terada M, Sasaki H, et al. Virolysis and in vitro neutralization of HIV-1 by humanized monoclonal antibody hNM-01. Hybridoma 2000; 19: 427–34 [DOI] [PubMed] [Google Scholar]
  • 101.Dezube BJ, Doweiko JP, Proper JA, et al. Monoclonal antibody hNM01 in HIV-infected patients: a phase I study. J Clin Virol 2004; 31 Suppl. 1: S45–7 [DOI] [PubMed] [Google Scholar]
  • 102.Immune Network Ltd and ViroLogic, Inc. Immune Network enters research evaluation agreement with ViroLogic to evaluate anti-HIV antibody hNM01 [media release]. 2001. Jul 18 [online]. Available from URL: http://www.immunenetwork.com/sys/current/newsArchive.php?Id=153
  • 103.Eda Y, Takizawa M, Murakami T, et al. Sequential immunization with V3 peptides from primary human immunodeficiency virus type 1 produces cross-neutralizing antibodies against primary isolates with a matching narrow-neutralization sequence motif. J Virol 2006; 80: 5552–62 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Eda Y, Murakami T, Ami Y, et al. Anti-V3 humanized antibody KD-247 effectively suppresses ex vivo generation of human immunodeficiency virus type 1 and affords sterile protection of monkeys against a heterologous simian/human immunodeficiency virus infection. J Virol 2006; 80: 5563–70 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.The Chemo-Sero-Therapeutic Research Institute. A Study of Anti-HIV Monoclonal Antibody KD-247 [ClinicalTrials.gov identifier NCT00917813]. US National Institutes of Health, ClinicalTrials.gov [online]. Available from URL: http://clinicaltrials.gov [Accessed 2012 Feb 20]
  • 106.Corti D, Langedijk JP, Hinz A, et al. Analysis of memory B cell responses and isolation of novel monoclonal antibodies with neutralizing breadth from HIV-1-infected individuals. PLoS ONE 2010; 5: e8805. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Hioe CE, Wrin T, Seaman MS, et al. Anti-V3 monoclonal antibodies display broad neutralizing activities against multiple HIV-1 subtypes. PLoS ONE 2010; 5: e10254. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Watkins JD, Siddappa NB, Lakhashe SK, et al. An anti-HIV-1 V3 loop antibody fully protects cross-clade and elicits T-cell immunity in macaques mucosally challenged with an R5 clade C SHIV. PLoS ONE 2011; 6: e18207. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Buchacher A, Predl R, Strutzenberger K, et al. Generation of human monoclonal antibodies against HIV-1 proteins; electrofusion and Epstein-Barr virus transformation for peripheral blood lymphocyte immortalization. AIDS Res Hum Retroviruses 1994; 10: 359–69 [DOI] [PubMed] [Google Scholar]
  • 110.Sanders RW, Venturi M, Schiffner L, et al. The mannose-dependent epitope for neutralizing antibody 2G12 on human immunodeficiency virus type 1 glycoprotein gp120. J Virol 2002; 76: 7293–305 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Scanlan CN, Pantophlet R, Wormald MR, et al. The broadly neutralizing anti-human immunodeficiency virus type 1 antibody 2G12 recognizes a cluster of alpha1–>2 mannose residues on the outer face of gp120. J Virol 2002; 76: 7306–21 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Trkola A, Pomales AB, Yuan H, et al. Cross-clade neutralization of primary isolates of human immunodeficiency virus type 1 by human monoclonal antibodies and tetrameric CD4-IgG. J Virol 1995; 69: 6609–17 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Calarese DA, Scanlan CN, Zwick MB, et al. Antibody domain exchange is an immunological solution to carbohydrate cluster recognition. Science 2003; 300: 2065–71 [DOI] [PubMed] [Google Scholar]
  • 114.Doores KJ, Fulton Z, Huber M, et al. Antibody 2G12 recognizes di-mannose equivalently in domain- and nondomain-exchanged forms but only binds the HIV-1 glycan shield if domain exchanged. J Virol 2010; 84: 10690–9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Mascola JR, Lewis MG, Stiegler G, et al. Protection of Macaques against pathogenic simian/human immunodeficiency virus 89.6PD by passive transfer of neutralizing antibodies. J Virol 1999; 73: 4009–18 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Baba TW, Liska V, Hofmann-Lehmann R, et al. Human neutralizing monoclonal antibodies of the IgG1 subtype protect against mucosal simian-human immunodeficiency virus infection. Nat Med 2000; 6: 200–6 [DOI] [PubMed] [Google Scholar]
  • 117.Mascola JR, Stiegler G, VanCott TC, et al. Protection of macaques against vaginal transmission of a pathogenic HIV-1/SIV chimeric virus by passive infusion of neutralizing antibodies. Nat Med 2000; 6: 207–10 [DOI] [PubMed] [Google Scholar]
  • 118.Armbruster C, Stiegler GM, Vcelar BA, et al. A phase I trial with two human monoclonal antibodies (hMAb 2F5,2G12) against HIV-1. AIDS 2002; 16:227–33 [DOI] [PubMed] [Google Scholar]
  • 119.Stiegler G, Armbruster C, Vcelar B, et al. Antiviral activity of the neutralizing antibodies 2F5 and 2G12 in asymptomatic HIV-1-infected humans: a phase I evaluation. AIDS 2002; 16: 2019–25 [DOI] [PubMed] [Google Scholar]
  • 120.Armbruster C, Stiegler GM, Vcelar BA, et al. Passive immunization with the anti-HIV-1 human monoclonal antibody (hMAb) 4E10 and the hMAb combination 4E10/2F5/2G12. J Antimicrob Chemother 2004; 54: 915–20 [DOI] [PubMed] [Google Scholar]
  • 121.Trkola A, Kuster H, Rusert P, et al. Delay of HIV-1 rebound after cessation of antiretroviral therapy through passive transfer of human neutralizing antibodies. Nat Med 2005; 11: 615–22 [DOI] [PubMed] [Google Scholar]
  • 122.West AP Jr, Galimidi RP, Foglesong CP, et al. Design and expression of a dimeric form of human immunodeficiency virus type 1 antibody 2G12 with increased neutralization potency. J Virol 2009; 83: 98–104 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Klein JS, Webster A, Gnanapragasam PN, et al. A dimeric form of the HIV-1 antibody 2G12 elicits potent antibody-dependent cellular cytotoxicity. AIDS 2010; 24: 1633–40 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Luo XM, Lei MY, Feidi RA, et al. Dimeric 2G12 as a potent protection against HIV-1. PLoS Pathog 2010; 6: e1001225. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Walker LM, Huber M, Doores KJ, et al. Broad neutralization coverage of HIV by multiple highly potent antibodies. Nature 2011; 477: 466–70 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Pejchal R, Doores KJ, Walker LM, et al. A potent and broad neutralizing antibody recognizes and penetrates the HIV glycan shield. Science 2011; 334: 1097–103 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Walker LM, Phogat SK, Chan-Hui PY, et al. Broad and potent neutralizing antibodies from an African donor reveal a new HIV-1 vaccine target. Science 2009; 326: 285–9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Walker LM, Simek MD, Priddy F, et al. A limited number of antibody specificities mediate broad and potent serum neutralization in selected HIV-1 infected individuals. PLoS Pathog 2010; 6: e1001028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Pancera M, McLellan JS, Wu X, et al. Crystal structure of PG16 and chimeric dissection with somatically related PG9: structure-function analysis of two quaternary-specific antibodies that effectively neutralize HIV-1. J Virol 2010; 84: 8098–110 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.McLellan JS, Pancera M, Carrico C, et al. Structure of HIV-1 gp120 V1/V2 domain with broadly neutralizing antibody PG9. Nature 2011; 480: 336–43 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Walker LM, Sok D, Nishimura Y, et al. Rapid development of glycan-specific, broad, and potent anti-HIV-1 gp120 neutralizing antibodies in an R5 SIV/HIV chimeric virus infected macaque. Proc Natl Acad Sci U S A 2011; 108: 20125–9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Wu X, Changela A, O’Dell S, et al. Immunotypes of a quaternary site of HIV-1 vulnerability and their recognition by antibodies. J Virol 2011; 85: 4578–85 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Davenport TM, Friend D, Ellingson K, et al. Binding interactions between soluble HIV envelope glycoproteins and quaternary-structure-specific monoclonal antibodies PG9 and PG16. J Virol 2011; 85: 7095–107 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Euler Z, Bunnik EM, Burger JA, et al. Activity of broadly neutralizing antibodies, including PG9, PG16, and VRC01, against recently transmitted subtype B HIV-1 variants from early and late in the epidemic. J Virol 2011; 85: 7236–45 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Miller MD, Geleziunas R, Bianchi E, et al. A human monoclonal antibody neutralizes diverse HIV-1 isolates by binding a critical gp41 epitope. Proc Natl Acad Sci U S A 2005; 102: 14759–64 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Zhang MY, Vu BK, Choudhary A, et al. Cross-reactive human immunodeficiency virus type 1-neutralizing human monoclonal antibody that recognizes a novel conformational epitope on gp41 and lacks reactivity against self-antigens. J Virol 2008; 82: 6869–79 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Pietzsch J, Scheid JF, Mouquet H, et al. Anti-gp41 antibodies cloned from HIV-infected patients with broadly neutralizing serologic activity. J Virol 2010; 84: 5032–42 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Muster T, Steindl F, Purtscher M, et al. A conserved neutralizing epitope on gp41 of human immunodeficiency virus type 1. J Virol 1993; 67: 6642–7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Zwick MB, Labrijn AF, Wang M, et al. Broadly neutralizing antibodies targeted to the membrane-proximal external region of human immunodeficiency virus type 1 glycoprotein gp41. J Virol 2001; 75: 10892–905 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Parker CE, Deterding LJ, Hager-Braun C, et al. Fine definition of the epitope on the gp41 glycoprotein of human immunodeficiency virus type 1 for the neutralizing monoclonal antibody 2F5. J Virol 2001; 75: 10906–11 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Cardoso RM, Zwick MB, Stanfield RL, et al. Broadly neutralizing anti-HIV antibody 4E10 recognizes a helical conformation of a highly conserved fusion-associated motif in gp41. Immunity 2005; 22: 163–73 [DOI] [PubMed] [Google Scholar]
  • 142.Sun ZY, Oh KJ, Kim M, et al. HIV-1 broadly neutralizing antibody extracts its epitope from a kinked gp41 ectodomain region on the viral membrane. Immunity 2008; 28: 52–63 [DOI] [PubMed] [Google Scholar]
  • 143.Fiebig U, Schmolke M, Eschricht M, et al. Mode of interaction between the HIV-1-neutralizing monoclonal antibody 2F5 and its epitope. AIDS 2009; 23: 887–95 [DOI] [PubMed] [Google Scholar]
  • 144.Song L, Sun ZY, Coleman KE, et al. Broadly neutralizing anti-HIV-1 antibodies disrupt a hinge-related function of gp41 at the membrane interface. Proc Natl Acad Sci U S A 2009; 106: 9057–62 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Ofek G, Tang M, Sambor A, et al. Structure and mechanistic analysis of the anti-human immunodeficiency virus type 1 antibody 2F5 in complex with its gp41 epitope. J Virol 2004; 78: 10724–37 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Pejchal R, Gach JS, Brunel FM, et al. A conformational switch in human immunodeficiency virus gp41 revealed by the structures of overlapping epitopes recognized by neutralizing antibodies. J Virol 2009; 83: 8451–62 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Nelson JD, Brunel FM, Jensen R, et al. An affinity-enhanced neutralizing antibody against the membrane-proximal external region of human immunodeficiency virus type 1 gp41 recognizes an epitope between those of 2F5 and 4E10. J Virol 2007; 81: 4033–43 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Ferrantelli F, Rasmussen RA, Buckley KA, et al. Complete protection of neonatal rhesus macaques against oral exposure to pathogenic simian-human immunodeficiency virus by human anti-HIV monoclonal antibodies. J Infect Dis 2004; 189: 2167–73 [DOI] [PubMed] [Google Scholar]
  • 149.Joos B, Trkola A, Kuster H, et al. Long-term multiple-dose pharmacokinetics of human monoclonal antibodies (MAbs) against human immunodeficiency virus type 1 envelope gp120 (MAb 2G12) and gp41 (MAbs 4E10 and 2F5). Antimicrob Agents Chemother 2006; 50: 1773–9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Huber M, von Wyl V, Ammann CG, et al. Potent human immunodeficiency virus-neutralizing and complement lysis activities of antibodies are not obligatorily linked. J Virol 2008; 82: 3834–42 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Zhu Z, Qin HR, Chen W, et al. Cross-reactive HIV-1-neutralizing human monoclonal antibodies identified from a patient with 2F5-like antibodies. J Virol 2011; 85: 11401–8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Burkly LC, Olson D, Shapiro R, et al. Inhibition of HIV infection by a novel CD4 domain 2-specific monoclonal antibody. Dissecting the basis for its inhibitory effect on HIV-induced cell fusion. J Immunol 1992; 149: 1779–87 [PubMed] [Google Scholar]
  • 153.Moore JP, Sattentau QJ, Klasse PJ, et al. A monoclonal antibody to CD4 domain 2 blocks soluble CD4-induced conformational changes in the envelope glycoproteins of human immunodeficiency virus type 1 (HIV-1) and HIV-1 infection of CD4+ cells. J Virol 1992; 66: 4784–93 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Reimann KA, Burkly LC, Burrus B, et al. In vivo administration to rhesus monkeys of a CD4-specific monoclonal antibody capable of blocking AIDS virus replication. AIDS Res Hum Retroviruses 1993; 9: 199–207 [DOI] [PubMed] [Google Scholar]
  • 155.Reimann KA, Lin W, Bixler S, et al. A humanized form of a CD4-specific monoclonal antibody exhibits decreased antigenicity and prolonged plasma half-life in rhesus monkeys while retaining its unique biological and antiviral properties. AIDS Res Hum Retroviruses 1997; 13: 933–43 [DOI] [PubMed] [Google Scholar]
  • 156.Reimann KA, Khunkhun R, Lin W, et al. A humanized, nondepleting anti-CD4 antibody that blocks virus entry inhibits virus replication in rhesus monkeys chronically infected with simian immunodeficiency virus. AIDS Res Hum Retroviruses 2002; 18: 747–55 [DOI] [PubMed] [Google Scholar]
  • 157.Kuritzkes DR, Jacobson J, Powderly WG, et al. Antiretroviral activity of the anti-CD4 monoclonal antibody TNX-355 in patients infected with HIV type 1. J Infect Dis 2004; 189: 286–91 [DOI] [PubMed] [Google Scholar]
  • 158.Jacobson JM, Kuritzkes DR, Godofsky E, et al. Safety, pharmacokinetics, and antiretroviral activity of multiple doses of ibalizumab (formerly TNX-355), an anti-CD4 monoclonal antibody, in human immunodeficiency virus type 1-infected adults. Antimicrob Agents Chemother 2009; 53: 450–7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Tannox. TNX-355 with optimized background therapy (OBT) in treatment-experienced subjects with HIV-1 [ClinicalTrials.gov identifier NCT00089700]. US National Institutes of Health, ClinicalTrials.gov [online]. Available from URL: http://clinicaltrials.gov [Accessed 2012 Feb 20]
  • 160.Olson WC, Rabut GE, Nagashima KA, et al. Differential inhibition of human immunodeficiency virus type 1 fusion, gp120 binding, and CC-chemokine activity by monoclonal antibodies to CCR5. J Virol 1999; 73: 4145–55 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Trkola A, Ketas TJ, Nagashima KA, et al. Potent, broad-spectrum inhibition of human immunodeficiency virus type 1 by the CCR5 monoclonal antibody PRO 140. J Virol 2001; 75: 579–88 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Jacobson JM, Saag MS, Thompson MA, et al. Antiviral activity of single-dose PRO 140, a CCR5 monoclonal antibody, in HIV-infected adults. J Infect Dis 2008; 198: 1345–52 [DOI] [PubMed] [Google Scholar]
  • 163.Jacobson JM, Thompson MA, Lalezari JP, et al. Anti-HIV-1 activity of weekly or biweekly treatment with subcutaneous PRO 140, a CCR5 monoclonal antibody. J Infect Dis 2010; 201: 1481–7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.AIDSinfo drug database. PRO 140 [online]. Available from URL: http://www.aidsinfo.nih.gov/DrugsNew/DrugDetailT.aspx?int_id=423 [Accessed 2012 Feb 20]
  • 165.Progenics Pharmaceuticals, Inc. Study of PRO 140 by subcutaneous administration in adult subjects with HIV-1 infection [ClinicalTrials.gov identifier NCT00642707]. US National Institutes of Health, ClinicalTrials.gov [online]. Available from URL: http://clinicaltrials.gov [Accessed 2012 Feb 20]
  • 166.Chen W, Zhu Z, Feng Y, et al. Human domain antibodies to conserved sterically restricted regions on gp120 as exceptionally potent cross-reactive HIV-1 neutralizers. Proc Natl Acad Sci U S A 2008; 105: 17121–6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Ward ES, Gussow D, Griffiths AD, et al. Binding activities of a repertoire of single immunoglobulin variable domains secreted from Escherichia coli. Nature 1989; 341: 544–6 [DOI] [PubMed] [Google Scholar]
  • 168.Hamers-Casterman C, Atarhouch T, Muyldermans S, et al. Naturally occurring antibodies devoid of light chains. Nature 1993; 363: 446–8 [DOI] [PubMed] [Google Scholar]
  • 169.Muyldermans S, Atarhouch T, Saldanha J, et al. Sequence and structure of VH domain from naturally occurring camel heavy chain immunoglobulins lacking light chains. Protein Eng 1994; 7: 1129–35 [DOI] [PubMed] [Google Scholar]
  • 170.Greenberg AS, Avila D, Hughes M, et al. A new antigen receptor gene family that undergoes rearrangement and extensive somatic diversification in sharks. Nature 1995; 374: 168–73 [DOI] [PubMed] [Google Scholar]
  • 171.Muyldermans S, Cambillau C, Wyns L. Recognition of antigens by single-domain antibody fragments: the superfluous luxury of paired domains. Trends Biochem Sci 2001; 26: 230–5 [DOI] [PubMed] [Google Scholar]
  • 172.Peitsch MC. Protein modeling by e-mail. Nat Biotechnol 1995; 13: 658–60 [Google Scholar]
  • 173.Arnold K, Bordoli L, Kopp J, et al. The SWISS-MODEL workspace: a web-based environment for protein structure homology modelling. Bioinformatics 2006; 22: 195–201 [DOI] [PubMed] [Google Scholar]
  • 174.Kiefer F, Arnold K, Kunzli M, et al. The SWISS-MODEL Repository and associated resources. Nucleic Acids Res 2009; 37: D387–92 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.NCBI. Crystal structure of the intact human Igg B12 with broad and potent activity against primary Hiv-1 isolates: a template for HIV vaccine design [online]. Available from URL: http://www.ncbi.nlm.nih.gov/Structure/mmdb/mmdbsrv.cgi?uid=16748 [Accessed 2012 Mar 23] [Google Scholar]
  • 176.Ito M, Hiramatsu H, Kobayashi K, et al. NOD/SCID/gamma(c)(null) mouse: an excellent recipient mouse model for engraftment of human cells. Blood 2002; 100: 3175–82 [DOI] [PubMed] [Google Scholar]
  • 177.Cong Zhang J Zheng, W Chen, et al. Proactive immunity: eAd inhibit in vivo HIV infection in humanized mice and can be expressed by lentiviral vectors to reprogram cells to secrete broadly neutralizing anti-HIV antibodies [poster abstract no. 379]. 18th Conference on Retroviruses and Opportunistic Infections [online]. Available from URL: http://www.retroconference.org/2011/Abstracts/41951.htm [Accessed 2012 Feb 15]
  • 178.Forsman A, Beirnaert E, Aasa-Chapman MM, et al. Llama antibody fragments with cross-subtype human immunodeficiency virus type 1 (HIV-1)-neutralizing properties and high affinity for HIV-1 gp120. J Virol 2008; 82: 12069–81 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Koh WW, Steffensen S, Gonzalez-Pajuelo M, et al. Generation of a family-specific phage library of llama single chain antibody fragments that neutralize HIV-1. J Biol Chem 2010; 285: 19116–24 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.Jahnichen S, Blanchetot C, Maussang D, et al. CXCR4 nanobodies (VHH-based single variable domains) potently inhibit chemotaxis and HIV-1 replication and mobilize stem cells. Proc Natl Acad Sci U S A 2010; 107: 20565–70 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Gorlani A, Brouwers J, McConville C, et al. Llama antibody fragments have good potential for application as HIV type 1 topical microbicides. AIDS Res Hum Retroviruses 2012. February; 28 (2): 198–205 [DOI] [PubMed] [Google Scholar]
  • 182.Bouchet J, Basmaciogullari SE, Chrobak P, et al. Inhibition of the Nef regulatory protein of HIV-1 by a single-domain antibody. Blood 2011; 117: 3559–68 [DOI] [PubMed] [Google Scholar]
  • 183.Feige MJ, Walter S, Buchner J. Folding mechanism of the CH2 antibody domain. J Mol Biol 2004; 344: 107–18 [DOI] [PubMed] [Google Scholar]
  • 184.Dimitrov DS. Engineered CH2 domains (nanoantibodies). MAbs 2009; 1: 26–8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.Raghavan M, Bjorkman PJ. Fc receptors and their interactions with immunoglobulins. Annu Rev Cell Dev Biol 1996; 12: 181–220 [DOI] [PubMed] [Google Scholar]
  • 186.Sondermann P, Huber R, Oosthuizen V, et al. The 3.2-A crystal structure of the human IgG1 Fc fragment-Fc gammaRIII complex. Nature 2000; 406: 267–73 [DOI] [PubMed] [Google Scholar]
  • 187.Shields RL, Namenuk AK, Hong K, et al. High resolution mapping of the binding site on human IgG1 for Fc gamma RI, Fc gamma RII, Fc gamma RIII, and FcRn and design of IgG1 variants with improved binding to the Fc gamma R. J Biol Chem 2001; 276: 6591–604 [DOI] [PubMed] [Google Scholar]
  • 188.Roopenian DC, Akilesh S. FcRn: the neonatal Fc receptor comes of age. Nat Rev Immunol 2007; 7: 715–25 [DOI] [PubMed] [Google Scholar]
  • 189.Duncan AR, Winter G. The binding site for C1q on IgG. Nature 1988; 332: 738–40 [DOI] [PubMed] [Google Scholar]
  • 190.Idusogie EE, Presta LG, Gazzano-Santoro H, et al. Mapping of the C1q binding site on rituxan, a chimeric antibody with a human IgG1 Fc. J Immunol 2000; 164: 4178–84 [DOI] [PubMed] [Google Scholar]
  • 191.Yasmeen D, Ellerson JR, Dorrington KJ, et al. The structure and function of immunoglobulin domains. IV. The distribution of some effector functions among the Cgamma2 and Cgamma3 homology regions of human immunoglobulin G1. J Immunol 1976; 116: 518–26 [PubMed] [Google Scholar]
  • 192.Ellerson JR, Yasmeen D, Painter RH, et al. A fragment corresponding to the C(H)2 region of immunoglobulin G (IgG) with complement fixing activity. FEBS Lett 1972; 24: 318–22 [DOI] [PubMed] [Google Scholar]
  • 193.Xiao X, Feng Y, Vu BK, et al. A large library based on a novel (CH2) scaffold: identification of HIV-1 inhibitors. Biochem Biophys Res Commun 2009; 387: 387–92 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194.Gong R, Vu BK, Feng Y, et al. Engineered human antibody constant domains with increased stability. J Biol Chem 2009; 284: 14203–10 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195.Hackel BJ, Kapila A, Wittrup KD. Picomolar affinity fibronectin domains engineered utilizing loop length diversity, recursive mutagenesis, and loop shuffling. J Mol Biol 2008; 381: 1238–52 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196.Gong R, Wang Y, Feng Y, et al. Shortened engineered human antibody CH2 domains: increased stability and binding to the human neonatal Fc receptor. J Biol Chem 2011; 286: 27288–93 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197.Dumoulin M, Conrath K, Van Meirhaeghe A, et al. Single-domain antibody fragments with high conformational stability. Protein Sci 2002; 11: 500–15 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198.Jespers L, Schon O, Famm K, et al. Aggregation-resistant domain antibodies selected on phage by heat denaturation. Nat Biotechnol 2004; 22: 1161–5 [DOI] [PubMed] [Google Scholar]
  • 199.Arbabi-Ghahroudi M, MacKenzie R, Tanha J. Selection of non-aggregating VH binders from synthetic VH phage-display libraries. Methods Mol Biol 2009; 525: 187–216, xiii [DOI] [PubMed] [Google Scholar]
  • 200.Famm K, Hansen L, Christ D, et al. Thermodynamically stable aggregation-resistant antibody domains through directed evolution. J Mol Biol 2008; 376: 926–31 [DOI] [PubMed] [Google Scholar]
  • 201.Borrebaeck CA, Malmborg AC, Furebring C, et al. Kinetic analysis of recombinant antibody-antigen interactions: relation between structural domains and antigen binding. Biotechnology 1992; 10: 697–8 [DOI] [PubMed] [Google Scholar]
  • 202.Kontermann RE. Strategies to extend plasma half-lives of recombinant antibodies. BioDrugs 2009; 23: 93–109 [DOI] [PubMed] [Google Scholar]
  • 203.Dennis MS, Zhang M, Meng YG, et al. Albumin binding as a general strategy for improving the pharmacokinetics of proteins. J Biol Chem 2002; 277: 35035–43 [DOI] [PubMed] [Google Scholar]
  • 204.Andersen JT, Pehrson R, Tolmachev V, et al. Extending half-life by indirect targeting of the neonatal Fc receptor (FcRn) using a minimal albumin binding domain. J Biol Chem 2011; 286: 5234–41 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 205.Krinner EM, Hepp J, Hoffmann P, et al. A highly stable polyethylene glycol-conjugated human single-chain antibody neutralizing granulocyte-macrophage colony stimulating factor at low nanomolar concentration. Protein Eng Des Sel 2006; 19: 461–70 [DOI] [PubMed] [Google Scholar]
  • 206.Holt LJ, Basran A, Jones K, et al. Anti-serum albumin domain antibodies for extending the half-lives of short lived drugs. Protein Eng Des Sel 2008; 21: 283–8 [DOI] [PubMed] [Google Scholar]
  • 207.Harris A, Borgnia MJ, Shi D, et al. Trimeric HIV-1 glycoprotein gp140 immunogens and native HIV-1 envelope glycoproteins display the same closed and open quaternary molecular architectures. Proc Natl Acad Sci U S A 2011; 108: 11440–5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 208.Pietzsch J, Scheid JF, Mouquet H, et al. Human anti-HIV-neutralizing antibodies frequently target a conserved epitope essential for viral fitness. J Exp Med 2010; 207: 1995–2002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 209.Ryser HJ, Fluckiger R. Progress in targeting HIV-1 entry. Drug Discov Today. 2005; 10: 1085–94 [DOI] [PubMed] [Google Scholar]
  • 210.Glanville J, Zhai W, Berka J, et al. Precise determination of the diversity of a combinatorial antibody library gives insight into the human immunoglobulin repertoire. Proc Natl Acad Sci U S A 2009; 106: 20216–21 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 211.Boyd SD, Marshall EL, Merker JD, et al. Measurement and clinical monitoring of human lymphocyte clonality by massively parallel VDJ pyrosequencing. Sci Transl Med 2009; 1: 12ra23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 212.Boyd SD, Gaeta BA, Jackson KJ, et al. Individual variation in the germline Ig gene repertoire inferred from variable region gene rearrangements. J Immunol 2010; 184: 6986–92 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 213.Ravn U, Gueneau F, Baerlocher L, et al. By-passing in vitro screening: next generation sequencing technologies applied to antibody display and in silico candidate selection. Nucleic Acids Res 2010; 38: e193. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 214.Jackson KJ, Boyd S, Gaeta BA, et al. Benchmarking the performance of human antibody gene alignment utilities using a 454 sequence dataset. Bioinformatics 2010; 26: 3129–30 [DOI] [PubMed] [Google Scholar]
  • 215.Wu X, Zhou T, Zhu J, et al. Focused evolution of HIV-1 neutralizing antibodies revealed by structures and deep sequencing. Science 2011; 333: 1593–602 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 216.Dimitrov DS. Therapeutic antibodies, vaccines and antibodyomes. MAbs 2010; 2: 347–56 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 217.Zhai W, Glanville J, Fuhrmann M, et al. Synthetic antibodies designed on natural sequence landscapes. J Mol Biol 2011; 412: 55–71 [DOI] [PubMed] [Google Scholar]
  • 218.Tiller T, Meffre E, Yurasov S, et al. Efficient generation of monoclonal antibodies from single human B cells by single cell RT-PCR and expression vector cloning. J Immunol Methods 2008; 329: 112–24 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 219.Scheid JF, Mouquet H, Feldhahn N, et al. Broad diversity of neutralizing antibodies isolated from memory B cells in HIV-infected individuals. Nature 2009; 458: 636–40 [DOI] [PubMed] [Google Scholar]
  • 220.Walker LM, Huber M, Doores KJ, et al. Broad neutralization coverage of HIV by multiple highly potent antibodies. Nature 2011; 477 (7365): 466–70 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 221.Xiao X, Chen W, Feng Y, et al. Germline-like predecessors of broadly neutralizing antibodies lack measurable binding to HIV-1 envelope glycoproteins: implications for evasion of immune responses and design of vaccine immunogens. Biochem Biophys Res Commun 2009; 390: 404–9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 222.Xiao X, Chen W, Feng Y, et al. Maturation pathways of cross-reactive HIV-1 neutralizing antibodies. Viruses 2009; 1: 802–17 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 223.Mosier DE, Gulizia RJ, Baird SM, et al. Human immunodeficiency virus infection of human-PBL-SCID mice. Science 1991; 251: 791–4 [DOI] [PubMed] [Google Scholar]
  • 224.Allaway GP, Davis-Bruno KL, Beaudry GA, et al. Expression and characterization of CD4-IgG2, a novel heterotetramer that neutralizes primary HIV type 1 isolates. AIDS Res Hum Retroviruses 1995; 11: 533–9 [DOI] [PubMed] [Google Scholar]
  • 225.Gauduin MC, Allaway GP, Olson WC, et al. CD4-immunoglobulin G2 protects Hu-PBL-SCID mice against challenge by primary human immunodeficiency virus type 1 isolates. J Virol 1998; 72: 3475–8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 226.Nagashima KA, Thompson DA, Rosenfield SI, et al. Human immunodeficiency virus type 1 entry inhibitors PRO 542 and T-20 are potently synergistic in blocking virus-cell and cell-cell fusion. J Infect Dis 2001; 183: 1121–5 [DOI] [PubMed] [Google Scholar]
  • 227.Jacobson JM, Lowy I, Fletcher CV, et al. Single-dose safety, pharmacology, and antiviral activity of the human immunodeficiency virus (HIV) type 1 entry inhibitor PRO 542 in HIV-infected adults. J Infect Dis 2000; 182: 326–9 [DOI] [PubMed] [Google Scholar]
  • 228.Shearer WT, Israel RJ, Starr S, et al. Recombinant CD4-IgG2 in human immunodeficiency virus type 1-infected children: phase 1/2 study. The Pediatric AIDS Clinical Trials Group Protocol 351 Study Team. J Infect Dis 2000; 182: 1774–9 [DOI] [PubMed] [Google Scholar]
  • 229.Jacobson JM, Israel RJ, Lowy I, et al. Treatment of advanced human immunodeficiency virus type 1 disease with the viral entry inhibitor PRO 542. Antimicrob Agents Chemother 2004; 48: 423–9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 230.Mukhtar M, Parveen Z, Pomerantz RJ. Technology evaluation: PRO-542, Progenics Pharmaceuticals inc. Curr Opin Mol Ther 2000; 2: 697–702 [PubMed] [Google Scholar]
  • 231.Lagenaur LA, Villarroel VA, Bundoc V, et al. sCD4–17b bifunctional protein: extremely broad and potent neutralization of HIV-1 Env pseudotyped viruses from genetically diverse primary isolates. Retrovirology 2010; 7: 11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 232.West AP Jr, Galimidi RP, Foglesong CP, et al. Evaluation of CD4-CD4i antibody architectures yields potent, broadly cross-reactive anti-human immunodeficiency virus reagents. J Virol 2010; 84: 261–9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 233.Chen W, Xiao X, Wang Y, et al. Bifunctional fusion proteins of the human engineered antibody domain m36 with human soluble CD4 are potent inhibitors of diverse HIV-1 isolates. Antiviral Res 2010; 88: 107–15 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 234.Chen W, Feng Y, Gong R, et al. Engineered single human CD4 domains as potent HIV-1 inhibitors and components of vaccine immunogens. J Virol 2011; 85: 9395–405 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 235.Ji C, Kopetzki E, Jekle A, et al. CD4-anchoring HIV-1 fusion inhibitor with enhanced potency and in vivo stability. J Biol Chem 2009; 284: 5175–85 [DOI] [PubMed] [Google Scholar]
  • 236.Jekle A, Chow E, Kopetzki E, et al. CD4-BFFI: a novel, bifunctional HIV-1 entry inhibitor with high and broad antiviral potency. Antiviral Res 2009; 83: 257–66 [DOI] [PubMed] [Google Scholar]
  • 237.Schanzer J, Jekle A, Nezu J, et al. Development of tetravalent, bispecific CCR5 antibodies with antiviral activity against CCR5 monoclonal antibody-resistant HIV-1 strains. Antimicrob Agents Chemother 2011; 55: 2369–78 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 238.Ridgway JB, Presta LG, Carter P. ‘Knobs-into-holes’ engineering of antibody CH3 domains for heavy chain heterodimerization. Protein Eng 1996; 9: 617–21 [DOI] [PubMed] [Google Scholar]
  • 239.Schaefer W, Regula JT, Bahner M, et al. Immunoglobulin domain crossover as a generic approach for the production of bispecific IgG antibodies. Proc Natl Acad Sci U S A 2011; 108: 11187–92 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 240.Mouquet H, Scheid JF, Zoller MJ, et al. Polyreactivity increases the apparent affinity of anti-HIV antibodies by heteroligation. Nature 2010; 467: 591–5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 241.Mouquet H, Warncke M, Scheid JF, et al. Enhanced HIV-1 neutralization by antibody heteroligation. Proc Natl Acad Sci U S A 2012; 109 (3): 875–80 [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES