TREIMM-1124; No. of Pages 13

Feature Review

Antibody B cell responses in HIV-1 infection Hugo Mouquet1,2 1 2

Laboratory of Humoral Response to Pathogens, Department of Immunology, Institut Pasteur, Paris, France CNRS-URA 1961, Paris, France

In rare cases, B cells can supply HIV-1-infected individuals with unconventional antibodies equipped to neutralize the wide diversity of viral variants. Innovations in single-cell cloning, high-throughput sequencing, and structural biology methods have enabled the capture and thorough characterization of these exceptionally potent broadly neutralizing antibodies (bNAbs). Here, I review the recent findings in humoral responses to HIV1, focusing on the interplay between naturally occurring bNAbs and the virus both at systemic and mucosal levels. In this context, I discuss how an improved understanding of bNAb generation may provide invaluable insight into the fundamental mechanisms governing adaptive B cell responses to viruses, and how this knowledge is currently contributing to the development of vaccine and therapeutic strategies against HIV-1. Antibodies against HIV-1 More than a century ago, Von Behring and Kitasato provided the first evidence on the role of the humoral response in host protection against infectious agents using serum therapy experiments against diphtheria and tetanus toxins [1]. ‘Briefly expressed, serum therapy works through anti-bodies’ explained Emil Von Behring (http://www.nobelprize.org). We now know that antibodies are valuable immunotherapeutics for a wide range of infectious diseases [2,3], that they can confer mother-to-child protective immunity [4], and that they present the best-known correlates of protection for many licensed vaccines [5]. Indeed, in addition to polyreactive (see Glossary) natural antibodies that act as the first line of defense against invading pathogens, humoral memory is composed of high-affinity antibodies that mediate long-lived immunity against infectious agents, for example, by providing protection against reinfection [5]. Thereby, the induction of high-affinity antibodies that inactivate or neutralize the culprit pathogen is a necessary component for most effective vaccines. Soon after the discovery of HIV-1 as the etiological agent causing acquired immunodeficiency syndrome (AIDS) [6], the existence of antibodies in patients’ sera capable of neutralizing HIV-1 strains in vitro was demonstrated [7,8]. To mediate viral neutralization, antibodies must recognize functional sites on the HIV-1 envelope spike, Corresponding author: Mouquet, H. ([email protected]). Keywords: HIV-1; antibodies; B cells; adaptive immunity; viruses. 1471-4906/ ß 2014 Elsevier Ltd. All rights reserved. http://dx.doi.org/10.1016/j.it.2014.08.007

gp160, which is a hetero-trimer composed of gp120 and gp41 protein subunits [9]. Even so, targeted epitopes on functional gp160 sites must be sufficiently conserved across most HIV-1 quasi-species to enable viral neutralization by one single cross-reacting neutralizing antibody. These conserved functional epitopes are called sites of vulnerability of the HIV-1 envelope protein. Since then, major efforts have been made to develop an efficient HIV-1 vaccine capable of inducing protective immunity, particularly by eliciting cross-neutralizing antibodies. So far, however, all attempts have failed despite the encouraging clinical vaccine trial carried out in Thailand known as the RV144 trial. This trial evaluated on 16 000 Thai subjects a vaccine regimen consisting of a nonreplicating avian pox Glossary Germinal centers: germinal centers are specialized microenvironments within secondary lymphoid organs that support the maturation, proliferation, and differentiation of B lymphocytes during T cell dependent immune responses. These microanatomical structures are divided into light and dark zones; in the dark zone, B cells expand and diversify their antigen receptors through somatic hypermutation (that potentially increases antibody affinity) and class-switching (that alters antibody effector functions), whereas in the light zone B cells are selected for binding to their cognate antigen. HIV-1 broadly neutralizing antibodies (bNAbs): HIV-1 bNAbs are antibodies capable of neutralizing in vitro numerous HIV-1 viral strains from diverse subgroups (or clades). bNAbs can be classified into two groups according to the time they have been isolated and their neutralizing activity. First-generation bNAbs (b12, 2G12, 4E10, and 2F5) were isolated in the 1990s, and exhibit limited neutralization potency and/or breadth. On the contrary, next-generation bNAbs identified from 2009 by single-cell cloning strategies have more potent and broad neutralization activity. Latent HIV-1 reservoirs: the latent reservoir of HIV-1 consists of a pool of resting memory CD4+ T cells that contain integrated DNA of replicationcompetent HIV-1 proviruses. HIV-1 reservoirs are long-lived, and mediate the persistence of infection by being immunologically silent and insensitive to antiretroviral therapy. Polyreactivity: antibody polyreactivity is the ability of an antibody, secreted or expressed at the surface of B cells as a BCR, to bind to various structurally unrelated ligands generally, with a low affinity. Sites of vulnerability: sites of vulnerability of HIV-1 are the epitopes targeted by bNAbs, which are well-conserved regions of the viral envelope glycoprotein among divergent HIV-1 quasi-species. Antibodies recognizing these sites either interfere with the receptor/co-receptor binding to HIV-1 gp120 or with viral fusion. T cell dependent and independent B cell responses: during humoral immune responses to protein antigens, a T cell dependent activation of B lymphocytes is required, and necessitates the cooperation of B cells with helper T cells for the production of high-affinity antibodies. Upon exposure to non-protein molecules such as polysaccharides and lipids, B cells can respond autonomously by producing antibodies without presenting antigen to CD4+ T cells (defined as T cell independent B cell response). Virological synapse: the virological synapse is a specialized site of cell-to-cell contact between cells infected with lymphotropic viruses (e.g., HIV-1 and human T cell leukemia virus type 1; HTLV-1) and uninfected T cells. Virological synapses play a crucial role in the pathogenesis of these viruses by facilitating the transfer of viral material from infected to uninfected T cells.

Trends in Immunology xx (2014) 1–13

1

TREIMM-1124; No. of Pages 13

Feature Review clade E gp120 vector (ALVAC)-prime and a clades B/E gp120 protein (AIDSVAX)-boost, and showed a modest protection rate of about 31% [10]. Remarkably, antibodies raised against the variable loop 2 of gp120 in vaccinees were found to be the main positive predictive factor for protection [11]. In spite of this, major obstacles still prevent vaccines from achieving sterilizing immunity against the diversity of worldwide circulating HIV-1 strains. Similarly, significant barriers disable the immune system to destroy the virus in infected patients such as certain molecular peculiarities of its envelope protein that prevent an effective antibody recognition and neutralization [12], the generation of viral escape mutants and the persistence of longlived reservoirs of HIV-1 [13]. Nonetheless, rare seropositive individuals are able to produce potent bNAbs capable of neutralizing most of the known HIV-1 stains [14]. In the past 5 years, scrutiny of the human antibody response using methods that directly clone antibodies from single antigen-specific B cells isolated from these patients, has led to important insights as to how the humoral system operates to naturally combat infection. Recent advances in the arena of HIV-1-specific antibodies, notably the discovery and characterization of extremely potent bNAbs have also added new momentum to the quest for effective therapeutic approaches to prevent HIV-1 infection [14]. Here, I discuss the current understanding of how the humoral arm of the immune system naturally tries to fight HIV-1 infection in light of recent progress on the molecular dissection of bNAbs, and their interplay with HIV-1. In an effort to ‘phenocopy’ bNAb-based humoral responses with an adequate vaccine, there has been a major push worldwide to better understand the diverse immunological aspects of bNAb biology, including their gene/structural features, the identification of their epitopes on the viral spike, their antiviral properties, and the development/ maturation of bNAb-expressing B cell lineages during infection. These discoveries are reviewed herein, and in this context, I also discuss the design of antibody-based therapies for AIDS treatment and vaccines against HIV-1. Systemic and mucosal antibody responses to HIV-1 The initial antibody response to HIV-1 can be detected as immune complexes as early as 1 week post-infection [15]. In the following days, circulating anti-gp41 antibodies are produced, followed by production of anti-gp120 antibodies a few weeks later, but none of these antibodies are capable of neutralizing the infecting viral strain (autologous virus) [15]. The first autologous neutralizing antibodies appear several months post-infection and although they are unable to neutralize heterologous viruses (diversified viruses isolated from other seropositive individuals) [16–20], they exert a selective pressure on the virus that subsequently, rapidly evolves by generating escape mutants [19–21]. The development of crossreactive neutralizing antibodies capable of neutralizing dozens of heterologous viruses generally takes 2–4 years after seroconversion [22], but in rare cases could arise earlier [23]. Approximately 20% of HIV-1-infected individuals develop high levels of crossreactive neutralizing antibodies that target different regions of HIV-1 envelope protein [17,24–28]. Among them, rare individuals called ‘Elite 2

Trends in Immunology xxx xxxx, Vol. xxx, No. x

neutralizers’ (representing 1% of seropositive individuals) develop bNAbs [29], which by definition are active against hundreds of different cell-free viral quasi-species from various HIV-1 subtypes. Unexpectedly, the development of bNAbs is not an inherent trait of adults, and infected infants can also mount a bNAb-like immune response against HIV-1, 20 months post-infection [30]. Serological profiling and mapping experiments in elite neutralizers revealed that serum neutralization frequently originates from a limited set of specificities [31–33]. Specifically, gp160-specific bNAbs produced are mostly IgG1, and most of them target known sites of vulnerability [22,24,27,32,33]. Serological neutralizing breadth and potency in HIV-infected patients can be accounted for by combinations of: (i) a multitude of additive/synergetic neutralizing antibodies targeting various epitopes [34]; (ii) clonal variants of a same specificity but with slightly different neutralization spectra [33,35,36]; and (iii) few potent bNAbs recognizing different epitopes and exhibiting complementary neutralizing activity [37,38]. Cell-to-cell transmission of HIV-1 is thought to be one of the major mechanisms contributing to viral spread in vivo [39]. Therefore, it would particularly be interesting to examine whether neutralizing serum antibodies from Elite neutralizers are also active against HIV-1 cell-to-cell spread in vitro. Due to viral escape, bNAbs do not suppress viral replication in infected persons [19,20], but they could contribute to sustain low-level viral loads in some individuals [40]. In this respect, and similar to observations made in infected rhesus monkeys [41,42], B cell depletion induced by rituximab immunotherapy in a HIV-1 seropositive individual with lymphoma led to a drop of neutralizing antibody titers and to a subsequent rebound of plasma viremia [43]. Moreover, HIV-specific IgG2 antibodies were shown to be positively associated with the lack of or slow progression towards AIDS and thus, could participate in keeping the virus in check in certain individuals [44,45]. Finally, HIV-1 antibodies may play a beneficial role by decreasing the risk of a subsequent infection (superinfection), and of vertical transmission in infected pregnant women [179]. Neutralizing antibodies act through their antigen-binding sites by interfering with receptor/co-receptor attachment or viral fusion with the target cells. Moreover, the Fc region of antibodies equips them with different effector functions permitting elimination of infected cells by innate immune cells engaged via the Fc receptors. Interestingly, antibodies responsible for several Fc-mediated mechanisms such as antibody-dependent cell-mediated cytotoxity (ADCC) and antibody-dependent cell-mediated viral inhibition (ADCVI) could reduce the risk of mother-to-child transmission of HIV-1 [46]. Furthermore, these antibodies have been associated with protection in individuals naturally controlling the infection (long-term non-progressors) [47,48] and in RV144 vaccinees [11]. These Fc-mediated properties are thought to be attributable to non- or weakly neutralizing antibodies, but whether serum crossreactive antibodies or bNAbs can also utilize these cellular effector functions remains only partially explored [49,50]. Antibodies have a well-documented role in protection of mucosal tissues against infection by bacteria, viruses, and

TREIMM-1124; No. of Pages 13

Feature Review

Trends in Immunology xxx xxxx, Vol. xxx, No. x

protozoa, as well as protection from damage by pathogenassociated toxins; IgA in particular is noteworthy given its prevalence in mucosal tissues [51]. Although mucosal IgAs may constitute an important component for preventing HIV entry at mucosal sites, little is known about the mucosal antibody response to HIV-1 and whether mucosal IgAs could efficiently protect against the infection. At mucosal surfaces, anti-gp41 IgA molecules are transiently produced at a high level during the first 2 weeks of infection, likely by short-lived plasma cells in a T-independent manner [52]. Due to the massive and rapid depletion of gut CD4+ T cells and various types of damage to gut-associated lymphoid tissues during the early stage of infection, that is, germinal center loss [53,54], the T cell dependent induction of long-lived plasma cells producing high-affinity antiviral IgA antibodies is probably impaired [55]. Nevertheless, HIV-infected individuals but also HIV-exposed individuals that are persistently seronegative can develop mucosal HIV-specific IgAs capable of blocking viral transcytosis (translocation throughout epithelial cells) [56–58]. In addition, some mucosa-derived IgA monoclonal antibodies to gp160 have been shown to neutralize HIV-1, by inhibiting its transcytosis and its intracellular replication in epithelial cells [59,60]. Lastly, Bomsel et al. showed that gp41vaccinated macaques were protected against repeated vaginal challenges of high simian–human immunodeficiency virus (SHIV) doses and that all of them developed not only gp41-specific vaginal IgGs with neutralizing and/ or ADCC activities, but also vaginal IgAs that block HIV-1 transcytosis [61]. Bearing in mind that AIDS is primarily a sexually transmitted disease, and that too few studies have been undertaken to investigate mucosal immunity against HIV-1, unraveling the mucosal antibody response in infected individuals is crucial in trying to design strategies to block mucosal transmission of the virus. Thus, HIV-1 infection induces systemic and mucosal B cell responses composed of antibodies with neutralizing and/or antiviral effector properties. However, these conventional antibodies have a limited neutralization efficacy against diversifying HIV-1 strains, and bNAbs more rarely and slowly develop in infected individuals. Indeed, due to both the various strategies used by the virus to escape immune pressure and to the detrimental effects of the infection on the adaptive B cell immunity, the antibody response to HIV-1 is inherently weakened.

memory B lymphocytes (i.e., resembling tissue-resident memory B cells). Importantly, it has recently been shown that gp160-specific B cell clones reside within abnormal B cell subsets, including exhausted tissue-like memory B cells and activated memory B cells, which predominate in HIV-infected individuals and are associated with higher levels of viremia [62]. This constitutes another pathophysiological example of how B-cell immune dysfunctions may contribute to an inadequate humoral response to HIV-1. These alterations arise from systemic bystander effects and possibly from direct interactions of B cells with the viral proteins particularly, gp120 and Nef (reviewed in [63]). Jelicic et al. showed that the direct interaction of HIV-1 gp120 with integrin a4b7 expressed on human B cells in vitro induces repressive signaling events including the production of the immunosuppressive cytokine transforming growth factor (TGF)-b1, induced expression of the inhibitory receptor FcRL4 and decreased expression of CD80 on B cells [64]. This virus-dependent mechanism leads to an inhibition of B cell activation and proliferation as typically observed during the infection, and is potentially a major obstacle hampering the humoral immune response against HIV-1 [64]. Finally, HIV-1 infection negatively impacts on follicular helper T (Tfh) cells, which are key players in establishing humoral responses (see below), and Tfh cell deregulation may undeniably diminish B cell immunity against the virus (reviewed recently in [65]). In addition to these deleterious effects rendering B cell response less effective, and apart from the rapid mutation of variable regions of the HIV-1 envelope protein resulting in an enormous diversity of circulating HIV-1 strains [19,20], the virus – so called Nature’s Master of Disguise [66] – uses a multitude of strategies to avoid or divert antibody B cell recognition (Figure 1). Studies of HIV-specific monoclonal antibodies revealed the peculiar nature of HIV-1 gp160 as the origin of several escape mechanisms including: (i) carbohydrate shielding and shifting [67,68]; (ii) conformation masking [70]; (iii) steric occlusion [71]; (iv) transient epitope exposure [72]; (v) nonfunctional envelope spikes such as gp120–gp41 monomers, gp41 stumps or uncleaved gp160 precursors, which may divert the immune response from functional targets [73,74]; and (vi) low density of functional HIV gp160 on the viral surface [75,76] (Figure 1). Despite such hurdles, bNAbs targeting conserved functional sites on gp120 and gp41 can develop in rare infected individuals.

Multiple viral strategies for disrupting and evading the immune response HIV-1 infection induces major perturbations in B cell development, physiology, and function that likely interfere with the establishment of a normal antiviral humoral response. Deregulation of B cell differentiation and function during HIV-1 infection is now well documented and consists of a variety of abnormalities, which are also associated with pathological changes in lymphoid tissues supporting B cell immune response (follicular hyperplasia and germinal centers alterations) such as: (i) B cell hyperactivity (e.g., hypergammaglobulinemia and polyclonal B cell activation); (ii) increased frequency of peripheral blood immature/transitional B cells; (iii) increased plasmablast differentiation; and (iv) exhaustion of blood tissue-like

Features of HIV-1 neutralizing antibodies In the early 1990s, first-generation HIV-1 bNAbs were isolated using Epstein–Barr virus transformation and phage display methods [77–82]. Their extensive molecular characterization improved our knowledge of the host immune response to HIV-1 by identifying important sites of vulnerability, antibody structure-to-function relations, and numerous viral escape mechanisms as described above [83]. Nevertheless, the uncommon nature of the antibodies (unnatural IgH and IgL pairing for b12, self-reactivity for 2F5/4E10, and an atypical antigen-combining site shaped by swapped VH domains for 2G12), their limited neutralization breadth and/or potency, and the inability of vaccines to induce their development created doubt about the possibility of an antibody-based vaccine. 3

TREIMM-1124; No. of Pages 13

Feature Review

Angenic variability

>

15

nm

Low density surface expression of Env

Trends in Immunology xxx xxxx, Vol. xxx, No. x

Nonfunconal Env

Steric occlusion

Glycan shielding

Transient epitope exposure

Conformaons masking TRENDS in Immunology

Figure 1. Envelope-dependent defense mechanisms of HIV-1. The HIV-1 Env or HIV-1 spike is the antigenic target of neutralizing antibodies. As well as from the Env antigenic variability resulting from the low-fidelity reverse transcription of the viral genetic material, several architectural and structural features of the HIV-1 spike limit or block the access of antibodies to conserved neutralizing epitopes: (i) Many hostderived glycans cover the surface of the spike (making up 50% of its mass) to form a ‘glycan armor’ protecting the virus from antibody recognition; (ii) some epitopes are only momentarily exposed to antibodies such as the ‘pre-fusion state’ form of gp41 to which membrane proximal external region-specific broadly neutralizing antibodies bind, and the co-receptor binding site (or CD4-induced site, CD4i) that opens following a conformational change induced by the interaction of gp120 with a CD4 molecule; (iii) similarly, conformational masking of the CD4 binding site (CD4bs) prevents antibody-mediated neutralization; (iv) the large size of immunoglobulins can sterically hinder the antibody binding to some epitopes (such as the CD4i), which can be accessed by Fab fragments alone; (v) most antibodies generated during HIV-1 infection target epitopes on nonfunctional spikes (noncleaved Env precursors, gp120 monomers, and gp41 trunks), which are not expressed by mature functional spikes; and (vi) the small number of gp160 glycoproteins (15 per virion), which are randomly distributed at the HIV surface, likely impair the ability of the antibody to bridge two Env (bivalent antibody binding), therefore reducing avidity effects. Abbreviation: Env, surface envelope glycoprotein.

In the past 5 years, research into the origin and nature of bNAbs was re-energized by the use of methodologies for producing antigen-specific antibodies from human B cells either by B cell capture [34], originally developed a decade ago as a means to study B cell tolerance and its breakage in autoimmune diseases [69,84], or by screening of cultured B cells [85,86]. The engineering of novel recombinant and soluble HIV-1 envelope proteins used as baits to identify and capture gp160-specific B cells by flow cytometry [87– 89], in combination with a meticulous selection of HIVinfected donors, has enabled single cell antibody cloning techniques to successfully generate hundreds of HIV-specific antibodies and among them, dozens of new extremely potent next-generation bNAbs (Figure 2) [14]. Until recently, mapping and structural definition of the HIV-1 bNAbs epitopes has allowed the identification of several sites of vulnerability on the HIV-1 envelope spike (Figure 2), comprising four major targets: the CD4 binding site (CD4bs), N-glycan-associated epitopes on the V1/V2 loops and the V3 loop, and the membrane proximal external region (MPER) on gp41 (Figure 2) [90]. Using similar technical approaches, the latest research efforts have led to the discovery of two other sites of vulnerability, targeted by 8ANC185 and PGT151-PGT158 bNAbs [91–93]. Both sites involve N-glycan-associated epitopes in the gp120/gp41 bridging regions [91–93] (Figure 2); thus, the 4

glycan-dependent gp120–gp41 interface is a novel ‘supersite’ of vulnerability that should be considered for vaccine immunogens design. Importantly, scrutiny of the humoral response in patients with broad serum neutralizing activity using single B cell capture and cloning approaches first revealed the diversity of the gp160-specific IgG class-switched B cell memory compartment in terms of clonal expansion, targeted epitopes, and neutralization capacity [34,94]. Gene repertoire of anti-HIV-1 memory antibodies showed abnormalities compared to that of healthy controls, in particular a preferential use of heavy-chain variable region 1 (VH1) and immunoglobulin light chain kappa (Igk), and evidence of high rates of somatic hypermutation [34,94]. Increased hypermutation level is also an important gene signature of HIV-1 bNAbs, which carry more than 40% amino acid substitutions in their heavy chains, as well as nucleotide insertions and/or deletions (termed ‘indels’) in heavy and light chain genes that are rare in other antibodies (Figure 3A) [14]. Many bNAbs also express unusually long CDRH3 loops necessary to reach a cryptic region of their epitope, by penetrating the glycan shield or by extracting it from the lipid membrane (Figure 3A) [83,90]. Somatic mutations, spanning both the complementarity determining region (CDR) and the relatively mutation resistant framework regions, increase the affinity of anti-HIV antibodies to their ligand, and are essential for viral neutralization [36,95,96]. Surprisingly, common VH gene segments (VH1–2 and VH1–46) encode next generation anti-CD4bs bNAbs of the VRC01 class originating from different individuals and despite extensive hypermutations (Figure 3B), VRC01-class bNAbs share a large consensus IgH sequence and conserve structural features for contacting the HIV-1 spike [36,97]. Regardless of their neutralization activity, a large fraction of gp160-specific antibodies are polyreactive, reacting with a number of host and other non-HIV antigens [98]. Anti-gp41 bNAbs, 2F5 and 4E10, were the first described to be polyspecific and reactive against several autoantigens, for example, membrane phospholipids and human larynx carcinoma HEp-2 cells [99,180]. In this regard, anti-gp41 antibodies are more frequently poly- and self-reactive than gp120-specific antibodies [94,96,100–102]. Other potent bNAbs were also shown to be weakly poly- or self-reactive such as antibodies to the CD4bs (45-46, 12A12, CH98 and CH103/CH104/CH106) and the glycan-associated V1/2 loops (CH01-CH04) [36,40,86,103]. Nevertheless, the majority of the potent natural HIV-1 bNAbs are not polyreactive and/or autoreactive [98,104]. Antibody polyreactivity could originate either from the preferential selection of mature naı¨ve B cells expressing polyreactive receptors (6% of naı¨ve B cells in healthy humans [69]), which would yield this reactivity unaltered throughout affinity maturation of anti-HIV B cells, or alternatively, from the extensive affinity maturation itself as a byproduct of the accumulated hypermutations, as shown for instance in the CH103 B cell lineage [103,105]. Hence, understanding both the mechanisms linking acquisition of bNAb activity with poly/autoreactivity, and whether polyreactivity is a prerequisite for antibody neutralization are important matters that must be addressed.

TREIMM-1124; No. of Pages 13

Feature Review

Trends in Immunology xxx xxxx, Vol. xxx, No. x

N-glycans V1/V2 loops PG9/PG16, CH01-CH04, CAP256-VRC26 PGT141-PGT145

N156 CD4 binding site

N160 N-glycans V3 loops

b12, VRC01-VRC03/NIH45-46, VRC06b*, VRC23, HJ16, 1NC9, 8ANC134, 12A12, 3BNC60/3BNC117, VRC-PG04, VRC-CH30 - VRC-CH34, CH98, CH103-CH106

N332

PGT121-PGT123/ 10-1074, PGT125-PGT128, PGT135-PGT137, VRC24 CH01-CH04

N301

N276 N262

Unknown 3BNC176

N234

N448

Clustered N-glycans 2G12

N-glycans gp120/gp41 bridging region

N637

8ANC195, PGT151-PGT158

MPER 2F5, 4E10, Z13 m66.6, 10E8 TRENDS in Immunology

Figure 2. Sites of vulnerability on HIV-1 envelope glycoprotein. The HIV-1 envelope crystal structure [171] (PDB ID: 4NCO) was fit onto the cryoelectron microscopy reconstruction of an unliganded membrane-bound HIV-1 Env trimer ([172] (EMDB: 5019 and 5021). Env model that was used was fully glycosylated using predicted glycoforms (except for gp41, gp120 V2 and V4 loops, whose glycans were not modeled because of the disorder of those protein regions in the crystal structure or uncertainty in registry assignment). The figure was generated using UCSF Chimera [173]. N160-dependent V1/V2 loops (orange), N332-dependent V3 loop (purple), CD4 binding-site (pink) and glycans-dependent gp120/gp41 bridging region epitopes (blue and red) are highlighted as defined by PG9 [174,175], PGT128 [176], VRC01 [177], 8ANC195 [93] and PGT151 [91] binding. The MPER of gp41, for which only limited structural information is available in the context of the HIV-1 Env trimer, is also a site of vulnerability and is highlighted in yellow. bNAbs specifically mapped to each site of vulnerability are indicated in boxes. *VRC06b epitope is overlapping both the CD4bs and the CD4i. Abbreviations: bNAbs, broadly neutralizing antibodies; Env, surface envelope glycoprotein; MPER, membrane proximal external region.

Thus, HIV-1 bNAbs share common molecular features required for overcoming the various gp160-dependent mechanisms protecting the sites of vulnerability on the virion. Two main conserved signatures stand out: intensive somatic gene modifications (high rates of hypermutation and gene indels) and polyreactivity; both of which may constitute roadblocks for the immune system to generate bNAbs. Development of next generation HIV-1 bNAbs Genetic and biological host factors that determine or influence the generation of HIV-1 bNAbs are not fully understood, and are an area of active investigation. From an immunological standpoint, genetic predisposition to the natural control of HIV-1 (HIV-1 controller phenotype) is principally linked to certain HLA class I alleles predicted to enable efficient effector CD8+ T cell responses [106]. Whether specific genetic traits are associated with the development of bNAbs in Elite neutralizers remains an

open question. Conversely, it has been proposed that certain characteristics of the immune system could inherently limit the development of HIV-1 bNAbs explaining per se the rarity of such a phenomenon. At the gene level, the scarcity of some specific V(D)J rearrangements encoding germline precursors of HIV-1 bNAbs in the human immunoglobulin gene repertoire, such as those creating exceptionally long CDRH3s, could represent a restricting factor [107]. By contrast, the great number of DNA modifications: somatic hypermutations, and indels that accumulates in bNAbs-encoding genes through multiple rounds of affinity maturation is undoubtedly a major difficulty to overcome. While the affinity maturation process leading to the generation and selection of bNAb-expressing B cells is not well understood yet, chronic stimulation by extended antigen exposure appears to be mandatory, and this is rather associated with the progression of the disease than with its control (as in HIV-1 controllers) [26,108]. However, it is important to note that viral replication and evolution also 5

TREIMM-1124; No. of Pages 13

Feature Review

Trends in Immunology xxx xxxx, Vol. xxx, No. x

HIV-1 bNAbs

(A)

Increased VL mutaons

(B)

Long CDRH3

CD4bs bNAbs

Inseron N58 R71

W50

Deleon

VH

VL

VH

VL

VL

VH 1-2 GL

Short CDRL3

Increased VH mutaons

VH TRENDS in Immunology

Figure 3. Immunoglobulin gene and structural features of bNAbs. Crystal structure of 10-1074 Fab [35] (PDB ID: 4FQC) is used to display some of the molecular features of HIV-1 bNAbs (A). Most bNAbs exhibit unusual numbers of somatic hypermutations in heavy and light chain-coding genes not only in the complementary determining region but also in the more mutation-resistant framework regions [95]. Hypermutation-induced amino acid substitutions (IgH, red spheres; IgL blue spheres), frequently accompanied with nucleotide insertions and/or deletions, allow adequate shaping of the antigen-binding site to fit complex neutralizing epitopes expressed on gp120 and gp41 glycoprotein subunits. On average, the CDRH3 length of IgG memory B cell antibodies from uninfected healthy individuals is about 14 amino acids. In order to access hidden portions of their epitopes, many bNAbs have extra-long CDRH3 loops (generally beyond 20 residues, and up to 35 amino acids for CAP256-VRC26), which appear to be the product of germline VDJ rearrangements. Crystal structure of 3BNC60 [36] (PDB ID: 3RPI) is used to display common molecular features of CD4bs bNAbs of the VRC01 class (B). Many CD4bs bNAbs, isolated from different donors, originate from the VH1-2 gene segment (VH1-2 GL). Despite extensive hypermutations, these antibodies share a large consensus IgH sequence, including three germline residues (blue spheres: W50, N58, and R71), therefore conserving structural features necessary for contacting the HIV spike. In addition, the CDRL3 length of CD4bs bNAbs is usually short (5 residues) to avoid steric clashes with gp120. The figure was generated using MacPyMOL (The PyMOL Molecular Graphics System 2011) [178]. Abbreviations: bNAbs, broadly neutralizing antibodies; VH, heavy chain variable domain; VL, light chain variable domain.

occurs in HIV-1 controllers [109], and that some of these individuals also fit the Elite neutralizer category [34]. In addition to the persistence of stimulating antigen, viral diversification within sites of vulnerability is a crucial event driving the evolution of B cells from the precursors originally recruited against transmitted-founder viruses to the affinity maturated clones expressing bNAbs. This was elegantly shown for anti-CD4bs antibody, CH103 [103], and more recently for anti-V1/V2 loops antibody clonal lineage, CAP256-VRC26, which was initiated by the interaction of its B cell precursor with a superinfecting virus [110]. Neutralizing antibodies and HIV-1 are thus coevolving in a recurring ordered manner; first, the virus evades antibody recognition by mutating the targeted epitope, and then, the immune system fights back by extending the maturation of the B cell lineage to give birth to new clonal antibody variants with increased neutralization breadth, which are able to neutralize the escape mutant viruses [68,103,110]. Importantly, this process could be more complex, potentially involving additional B cell lineages that would cooperatively intervene by driving the early steps of the viral evolution necessary to induce the bNAbs lineages, as recently described for CH103 antibody [111]. What favors the priming, maturation, and selection of B cell clones harboring bNAbs specificities in the context of the B cell competition in germinal centers (GCs) is not known, but it certainly implicates an effective access to antigen and to survival signals provided by Tfh cells [112]. Indeed, by interacting with GC B cells through specific surface receptors/ligands [such as MHC class II-peptideTCR, CD40–CD40L, inducible co-stimulatory molecule (ICOS)–ICOSL, and signaling lymphocytic activation molecule (SLAM) receptors], and by stimulating them with interleukin (IL)-21, Tfh cells play a crucial role in humoral responses by orchestrating B cell maturation and 6

differentiation in GCs [112]. Moreover, the fact that activated Tfh cells can move freely between different GCs and join ongoing GC reactions to provide help to B cells, may ensure an efficient humoral response against pathogens diversifying their antigenic epitopes such as HIV-1 [113]. Recent works truly provide compelling evidence supporting the role of Tfh cells in the maturation and selection of bNAb-expressing B cells [65,114]. In this regard, Crotty and colleagues identified in humans a population of circulating memory Tfh cells (with a CD4+PD1+CXCR3 CXCR5+ phenotype), robustly supplying help to B cells, and which positively correlates with the presence of bNAbs in elite neutralizers [115]. Finally, the recent study by Gitlin et al. shed light on how B cell clonal selection is regulated in the GC. The authors showed that high-affinity GC B cells are selected to proliferate the greatest number of times in the GC dark zone, with each round of cell division leading to greater diversification via somatic mutation [116]. Hence, this feed-forward loop may provide a mechanistic explanation for the preferential selection and maturation of high affinity HIV-1 bNAb-expressing B cell clones that exhibit high levels of hypermutation. Human HIV-1 bNAbs usually belong to highly expanded clonal lineages (or clonotypes), and represent the most terminally differentiated members, suggesting that their elicitation during the infection is a progressive and slow process [117]. Recent advances in the development of highthroughput DNA sequencing technologies now permit investigations on antibody gene repertoires (also called antibodyomes) at an unprecedented level [118]. Next-generation high-throughput sequencing of Ig genes (Ig-seq), along with computational and structural biology tools, constitutes a promising approach for a better understanding of the humoral response to HIV-1. For instance, studies using 454-pyrosequencing and structural biology methods

TREIMM-1124; No. of Pages 13

Feature Review have provided precious information about the affinity maturation pathways giving rise to VRC01-like bNAbs [97,119]. Similar Ig-seq approaches are now being used to reconstitute the ‘family trees’ of other bNAbs such as the PGTs (PGT121-PGT123 and PGT135-137) and anti-gp41 10E8 [120–122]. Importantly, by facilitating the mapping of HIV-1 bNAb clonotypes, the Ig-seq approaches may be a crucial step in establishing vaccine schemes that could instruct B cells to follow the tortuous but inescapable maturation pathways (a strategy called ‘B-cell lineage immunogen design’) [123]. The priming of suitable B cell progenitors and their clonal progeny is a pivotal event in this scenario. Hence, efforts are currently being made to rationally design HIV-1 envelope-derived immunogens that could efficiently activate bNAb B cell precursors, especially for attractive candidates such as VRC01-like antibodies that derive from germline-encoded B cell receptors (BCRs) with the same VH gene family (VH1) [124,125]. It is still unclear whether during infection, the envelope expressed by transmitted/founder viruses correspond to the stimulating antigens of bNAb B cell precursors [103], or if as suggested by some authors, B cells selected against unrelated antigens are subsequently crossreacting with envelope proteins leading to their recruitment in the immune response to HIV-1 [123]. Such a scenario may occur at mucosal sites where B cells primed by commensal bacteria could then crossreact with HIV-1 as a result of a molecular mimetism between microbial antigens and gp41 protein [126]. Polyreactivity is a frequent property of gp160-specific antibodies [98], and since most polyreactive B cells are removed from the B cell repertoire during development [69,127], it has been proposed that physiological tolerance might prevent the genesis of HIV-1 bNAbs [128,129]. However, so far this concept has only been validated for 2F5 and 4E10 [130–132], and it remains to be verified if such a rule could also apply to the other polyreactive bNAbs. On the contrary, it is essential to highlight that somatic hypermutations can create polyreactive BCRs, which are commonly expressed by several B cell populations in healthy humans such as IgG+ memory B lymphocytes [98,133]. Furthermore, although the magnitude and spectrum of polyreactivity for HIV-1 bNAbs still need to be accurately defined, only a minority of them appears to be as highly poly- and self-reactive as the archetypes 2F5 and 4E10, which when knocked in on mouse B cells in their affinity maturated form failed to pass central and peripheral tolerance checkpoints [104,130–132]. In this model, it has been suggested that the molecular-mimicry-based crossreactivity of 2F5 and 4E10 with autoantigens (kynureninase enzyme and RNA splice factor 3B, respectively) is responsible for the tolerization of 2F5- and 4E10-expressing B cells [134]. Interestingly, this developmental blockade could be bypassed using an appropriate immunogen/adjuvants combination, allowing the production of 2F5 antibodies through the activation/expansion of residual anergic 2F5-expressing B cells [135]. Are B cell precursors expressing germline BCRs of potent next-generation bNAbs also counterselected in such models? Can HIV-1 infection interfere with normal tolerance mechanisms allowing the selection of ordinarily ‘banned’ clones? More answers are required to

Trends in Immunology xxx xxxx, Vol. xxx, No. x

adjudicate on this particular matter. Of note, if immune tolerance mechanisms truly interfere with bNAb development, one can hypothesize that tolerance breakage occurring during certain autoimmune diseases may facilitate the acquisition of self-reactivity associated with bNAb activity. In support of this, a recent study has shown that an HIV-1-infected individual who subsequently developed a systemic lupus erythematous autoimmune disease could develop polyreactive anti-CD4bs bNAbs exhibiting crossreactivity to human autoantigens including double-stranded DNA [40]. Last but not least, polyreactive binding exhibited by HIV-1 antibodies could be functionally important to increase their avidity against virions displaying low-density spikes at their surfaces [96,136]. Therefore, moderate polyreactivity might be beneficial for HIV-1 antibodies to combat the infection without representing a substantial obstacle to the development of most HIV-1 bNAbs. Taken together, the data indicates that multiple host and viral obstacles hamper the generation of HIV-1 bNAbs, potentially accounting for the rarity of this event in infected humans. B cell deregulation and viral defense mechanisms are major impediments that weaken the humoral immune response to HIV-1. Difficulties for the natural development of bNAbs also lie in their unusual molecular characteristics, which are ‘forged’ for counteracting the protecting mechanisms used by HIV-1 to escape from antibody pressure. Without being counterselected due to potentially harmful polyreactivity levels, highly mutated anti-HIV-1 antibodies need to be positively selected during the immune response especially against escape variants, and this requires the pivotal help of Tfh cells, which are themselves profoundly altered by the infection. Considering that in the course of the infection, the virus is escaping and diversifying at a high rate, many successive B cell checkpoints must to be overcome in order for bNAbs to develop. Antiviral activity of broadly HIV-1 neutralizing antibodies and therapeutic potential The antiviral activity of HIV-1 antibodies is typically measured in vitro using cell-free pseudovirus particles and reporter cell lines, such as the HeLa-derived TzMbl cell [137]. In these assays, which measure the inhibition of free virus binding to cellular receptors and/or of viral fusion (Figure 4), first-generation bNAbs can achieve neutralization breadth but only several orders of magnitude less than the recently isolated bNAbs, which can neutralize up to 95% of HIV-1 strains at low concentration [14]. Over the course of the epidemic, circulating HIV-1 strains progressively became more resistant to neutralization by antigp120 but not anti-gp41 antibodies; nevertheless, contemporary HIV-1 variants are still sensitive to neutralization by most first- and next-generation anti-gp160 bNAbs [35,138–140]. HIV-1 antibodies can also access viruses present at the virological synapse [141], but only a subset of potent HIV-1 bNAbs are capable of inhibiting most of the steps implicated in the lymphocyte-to-lymphocyte spread of HIV-1 including the formation of virological synapses and the transfer of viral material from infected cells to the targets 7

TREIMM-1124; No. of Pages 13

Feature Review

Cell-free viral neutralizaon

Trends in Immunology xxx xxxx, Vol. xxx, No. x

Inhibion of cell-to -cell viral spread

Fc-dependent anviral acvity

Inhibion of viral transcytosis1 Intracellular viral neutralizaon2 1

2

Infected cell

CD4

CCR5

Infected cell

Target cell

Epithelial cells

Fc receptor Perforin, granzymes

Target cell

Effector cell Plasma cell

TRENDS in Immunology

Figure 4. Antiviral activity of HIV-1 bNAbs. For decades, the major antiviral property considered for anti-gp160 antibodies was HIV neutralization in in vitro assays. In these systems, cell-free viral neutralization relies on the inhibition by antibodies of receptor/co-receptor attachment or viral fusion with the target cells. However, only a subset of potent HIV-1 bNAbs determined by this assay can efficiently block one of the most important modes of viral spread in vivo, which occurs through cell-to-cell transmission. Moreover, as already shown for some anti-gp160 antibodies, bNAbs may facilitate the clearance of infected cells by innate immune cells through Fc-dependent mechanisms such as antibody-dependent cell-mediated cytotoxicity. In this case, the antigen-binding site of the IgG molecule attaches cell-surface Env, and its Fc fragment is bound by Fc receptors expressed on effectors cells such as natural killer cells, which lyse the infected cells via the secretion of proteolytic enzymes, granzymes, and perforin. bNabs could also have a crucial role for the protection against HIV-1 infection at mucosal sites. Anti-HIV antibodies could inhibit the passage of HIV-1 from the lumen to the basolateral pole (HIV-1 transcytosis) (1) but also, neutralize virions inside epithelial cells (2) and in the lamina propria where they are produced by plasma cells. Abbreviations: bNAbs, broadly neutralizing antibodies; CCR5, chemokine CC receptor 5.

(Figure 4) [142]. These antibodies can also block viral transmission from infected lymphocytes to plasmacytoid dendritic cells, and compromise innate sensing of HIV-1 [142]. Similarly, viral transfer from infected immature dendritic cells to T cells can be blocked by HIV-1 bNAbs in an Fc-dependent manner, more efficiently by anti-gp120 than anti-gp41 antibodies, but not by non-neutralizing IgG [144]. Finally, anti-gp120 but not anti-gp41 bNAbs can inhibit the transfer of viruses from infected macrophages to T cells [143]. Overall, antibody specificity and effector function are key components to an efficient block of the HIV-1 cell-to-cell spread. Therefore, since viral spread from infected immune cells (T cells, dendritic cells, and macrophages) to uninfected CD4+ T cells is probably the dominant mode of HIV-1 propagation in vivo, next generation anti-gp120 bNAbs may represent the most desirable candidates for therapeutic interventions. Mucosal transmission of HIV-1 can be blocked in vitro and in humanized mice by HIV-1 bNAbs with modest neutralization capacity especially when expressed as monomeric or dimeric IgA [145–148] (Figure 4). Additionally, when first-generation bNAbs are injected as IgG in nonhuman primates challenged rectally or vaginally with SHIV, they protect against infection [149–154]. Whether bNAbs are effective in these in vivo models due to their ability to directly interfere with viral transcytosis and/or to decrease the infectivity of transcytosed viruses is not known. In this regard, the effect of HIV-1 IgG antibodies on HIV-1 transcytosis through epithelial cells in vitro is still disputed [155,156]. Thus, the inhibitory activity of next-generation bNAbs (as IgG and IgAs) against HIV-1 8

transcytosis remains to be evaluated. Finally, understanding the precise mechanisms allowing the transport of intravenously injected IgG antibodies to mucosal surfaces, and whether it implicates the neonatal Fc receptor (FcRn) [156,157], are key points to be addressed. Numerous studies have established that prophylactic passive transfer of first-generation bNAbs (2G12, b12, 2F5, and 4E10) to monkeys protects them against SHIV infection [49,149–151,154,158–162]. However, antibody immunoprophylaxis in humanized mice and in macaques using next-generation bNAbs has proven to be more efficient, suppressing viremia for a prolonged period of time and requiring much lower amounts of passively transferred antibody than for first-generation bNAbs [163–167]. For instance, passive administration of PGT121 as prophylactic treatment in rhesus macaques vaginally challenged with SHIV induced complete protection at serum concentrations at least two logs lower than the ones preconized for first-generation bNAbs [165]. More importantly, single bNAbs or a cocktail of two used as therapeutics in chronically infected macaques can suppress viral loads to undetectable levels, without the emergence of resistant viral strains [166,167]. Remarkably, antibody treatment also leads to a decrease in cell-associated HIV DNA, and improves HIV-specific T cell immunity [166,167], possibly suggesting a vaccine-like effect of bNAb immunotherapy [160]. It remains to be determined whether passive administration of HIV-1 bNAbs could purge latently infected cells undergoing reactivation. A part of the answer was revealed in a recent study by Halper-Stromberg et al. in which a combination of three potent bNAbs (3BNC117, PG16, and

TREIMM-1124; No. of Pages 13

Feature Review 10-1074; bNAbs tri-mix) could obstruct the establishment of the latent reservoir in humanized mice when administered early after infection [50]. For established infections, bNAbs in the presence of a combination of inducers of viral transcription was shown to interfere with the maintenance of the HIV-1 reservoir, as measured by viremia rebound following antibody decay [50]. Strikingly, bNAbs displaying efficient and long lasting therapeutic or prophylactic properties in infected humanized mice and non-human primates are those that effectively interfere with cell-tocell spread of HIV-1 in vitro [142,163]. Similarly, antibodies unable to inhibit intercellular viral spread failed to control viremia in these animal models. Therefore, the ability of potent bNAbs to block cell-contact-mediated viral transmission could possibly explain why these antibodies efficiently protect in vivo from infection. Finally, other mechanisms such as Fc-dependent effector function may play a critical role for in vivo protection as formerly shown for b12 [49,149], and more recently for a potent bNAbs trimix (3BNC117, PG16, and 10-1074) used in infected humanized mice [50]; this possibility must be further explored in the case of potent bNAbs. Antibody-mediated immunotherapy against HIV-1 was tested a decade ago in infected humans using first-generation bNAbs (2G12, 4E10, and 2F5). Antibodies passively administered alone or in combination before antiretroviral therapy (ART) cessation failed to control viral rebound due to the rapid emergence of HIV-1 escape variants [168,169]. Despite the discouraging outcome of early clinical trials, the use of potent next-generation human HIV-1 bNAbs offers new therapeutic opportunities. Indeed, considering the promising results obtained in infected animal models, after treatment with a single antibody or cocktail of antibody molecules with or without ART treatment, next generation bNAbs (i.e., anti-CD4bs VRC01/45-46 and 3BNC117, and anti-glycans/V3 loop PGT121 and 101074) should be evaluated in new clinical trials. The effectiveness of passive immunization with bNAbs could be tested in various clinical circumstances including mother-to-child HIV transmission [170] or in cohorts of seropositive patients not tolerating or resistant to ART [14]. Concluding remarks Humoral immune responses are fundamental to the host protection against pathogens, and rely on the extraordinary diversity of antibody molecules that ensure the recognition of a theoretically infinite number of foreign antigens. Since the beginning of the AIDS epidemic 30 years ago, intensive research efforts have been engaged to dissect the interplay between HIV-1 and neutralizing antibodies. However, only recently did the discovery and characterization at a molecular level of broadly neutralizing HIV-1 antibodies radically modify our conception of antiviral humoral immunity, bringing hopes for the elaboration of an effective vaccine as well as immunotherapeutic strategies. Groundbreaking findings were made possible by significant technical advances and importantly, by interdisciplinary and collaborative research enterprises. Studies have revealed unparalleled insights on the genetic diversity, structural features, and antiviral properties of HIV-1 bNAbs, and we are now better appreciating the

Trends in Immunology xxx xxxx, Vol. xxx, No. x

Box 1. Outstanding questions  Are B cell defects commonly found during infection also characteristic of Elite neutralizers?  Are tolerance checkpoints effective in HIV-1 infection, and do they prevent the development of polyreactive anti-HIV-1 antibodies?  Does the polyreactivity of some bNAbs play a role in their neutralization activity?  What are the stimulating antigens initiating HIV-1 B cell responses, especially B cell lineages of bNAbs?  Does B cell competition in germinal centers interfere with the development of bNAbs clonal lineages?  Can crossreactive neutralizing antibodies naturally be produced at mucosal sites, and can they protect against mucosal transmission?  Can potent bNAbs isolated at systemic level efficiently protect mucosa from infection? What are the mechanisms involved in mucosal immunity in vivo; does it implicate antibody binding to neonatal Fc receptor (FcRn)?  Can new-generation bNAbs exhibit in vitro and in vivo Fcdependent antiviral properties such as ADCC?  Can new generation bNAbs affect the HIV-1 latent reservoirs?  Can the immunotherapy-mediated protection achieved in monkeys be reproduced in humans?  Can the ‘B cell lineage immunogen design’ vaccine strategy lead to bNAbs development in animal models and humans?

processes involved in B cell selection, maturation, and dynamics enabling their elicitation in rare individuals. Many immune evasion mechanisms used by the virus to ‘trick’ antibodies have been elucidated, and research on bNAbs has uncovered several functionally conserved neutralizing sites, identified as the Achilles heels on the virus. Evolutionary pathways of HIV-1-specific B cell clones chasing constantly mutating viral targets are starting to be elucidated, and might guide future vaccine-design strategies. Considering that these bNAbs molecules are extremely potent prophylactic and therapeutic reagents in non-human primates models, they also offer unique opportunities as potential treatment alternatives to antiviral drugs alone in humans, and their efficacy should be evaluated in clinical trials (Box 1). Acknowledgments We are grateful to Jean-Philippe Julien and Ian A. Wilson (The Scripps Research Institute) for providing us the structural model used in Figure 2, and to Caroline Eden (Icahn School of Medicine at Mount Sinai) for helpful comments and manuscript editing. This work was supported by the European Research Council (ERC) – Seventh Framework Program (ERC-2013-StG 337146). H.M. was supported by the G5 Institut Pasteur Program and the Milieu Inte´rieur Program (ANR-10LABX-69-01).

References 1 Silverstein, A.M. (2009) A History of Immunology, Academic Press 2 Berry, J.D. and Gaudet, R.G. (2011) Antibodies in infectious diseases: polyclonals, monoclonals and niche biotechnology. N. Biotechnol. 28, 489–501 3 Marasco, W.A. and Sui, J. (2007) The growth and potential of human antiviral monoclonal antibody therapeutics. Nat. Biotechnol. 25, 1421–1434 4 Zinkernagel, R.M. (2001) Maternal antibodies, childhood infections, and autoimmune diseases. N. Engl. J. Med. 345, 1331–1335 5 Plotkin, S.A. (2008) Vaccines: correlates of vaccine-induced immunity. Clin. Infect. Dis. 47, 401–409 6 Barre-Sinoussi, F. et al. (1983) Isolation of a T-lymphotropic retrovirus from a patient at risk for acquired immune deficiency syndrome (AIDS). Science 220, 868–871 9

TREIMM-1124; No. of Pages 13

Feature Review 7 Robert-Guroff, M. et al. (1985) HTLV-III-neutralizing antibodies in patients with AIDS and AIDS-related complex. Nature 316, 72–74 8 Weiss, R.A. et al. (1985) Neutralization of human T-lymphotropic virus type III by sera of AIDS and AIDS-risk patients. Nature 316, 69–72 9 Zolla-Pazner, S. (2004) Identifying epitopes of HIV-1 that induce protective antibodies. Nat. Rev. Immunol. 4, 199–210 10 Rerks-Ngarm, S. et al. (2009) Vaccination with ALVAC and AIDSVAX to prevent HIV-1 infection in Thailand. N. Engl. J. Med. 361, 2209– 2220 11 Haynes, B.F. et al. (2012) Immune-correlates analysis of an HIV-1 vaccine efficacy trial. N. Engl. J. Med. 366, 1275–1286 12 Haynes, B.F. et al. (2011) B cell responses to HIV-1 infection and vaccination: pathways to preventing infection. Trends Mol. Med. 17, 108–116 13 International AIDS Society Scientific Working Group on HIV Cure et al. (2012) Towards an HIV cure: a global scientific strategy. Nat. Rev. Immunol. 12, 607–614 14 Klein, F. et al. (2013) Antibodies in HIV-1 vaccine development and therapy. Science 341, 1199–1204 15 Tomaras, G.D. et al. (2008) Initial B-cell responses to transmitted human immunodeficiency virus type 1: virion-binding immunoglobulin M (IgM) and IgG antibodies followed by plasma anti-gp41 antibodies with ineffective control of initial viremia. J. Virol. 82, 12449–12463 16 Deeks, S.G. et al. (2006) Neutralizing antibody responses against autologous and heterologous viruses in acute versus chronic human immunodeficiency virus (HIV) infection: evidence for a constraint on the ability of HIV to completely evade neutralizing antibody responses. J. Virol. 80, 6155–6164 17 Gray, E.S. et al. (2007) Neutralizing antibody responses in acute human immunodeficiency virus type 1 subtype C infection. J. Virol. 81, 6187–6196 18 Moog, C. et al. (1997) Autologous and heterologous neutralizing antibody responses following initial seroconversion in human immunodeficiency virus type 1-infected individuals. J. Virol. 71, 3734–3741 19 Richman, D.D. et al. (2003) Rapid evolution of the neutralizing antibody response to HIV type 1 infection. Proc. Natl. Acad. Sci. U.S.A. 100, 4144–4149 20 Wei, X. et al. (2003) Antibody neutralization and escape by HIV-1. Nature 422, 307–312 21 Frost, S.D. et al. (2005) Neutralizing antibody responses drive the evolution of human immunodeficiency virus type 1 envelope during recent HIV infection. Proc. Natl. Acad. Sci. U.S.A. 102, 18514–18519 22 Gray, E.S. et al. (2011) The neutralization breadth of HIV-1 develops incrementally over four years and is associated with CD4+ T cell decline and high viral load during acute infection. J. Virol. 85, 4828– 4840 23 Mikell, I. et al. (2011) Characteristics of the earliest cross-neutralizing antibody response to HIV-1. PLoS Pathog. 7, e1001251 24 Binley, J.M. et al. (2008) Profiling the specificity of neutralizing antibodies in a large panel of plasmas from patients chronically infected with human immunodeficiency virus type 1 subtypes B and C. J. Virol. 82, 11651–11668 25 Dhillon, A.K. et al. (2007) Dissecting the neutralizing antibody specificities of broadly neutralizing sera from human immunodeficiency virus type 1-infected donors. J. Virol. 81, 6548– 6562 26 Doria-Rose, N.A. et al. (2010) Breadth of human immunodeficiency virus-specific neutralizing activity in sera: clustering analysis and association with clinical variables. J. Virol. 84, 1631–1636 27 Li, Y. et al. (2009) Analysis of neutralization specificities in polyclonal sera derived from human immunodeficiency virus type 1-infected individuals. J. Virol. 83, 1045–1059 28 Hraber, P. et al. (2014) Prevalence of broadly neutralizing antibody responses during chronic HIV-1 infection. Aids 28, 163–169 29 Simek, M.D. et al. (2009) Human immunodeficiency virus type 1 elite neutralizers: individuals with broad and potent neutralizing activity identified by using a high-throughput neutralization assay together with an analytical selection algorithm. J. Virol. 83, 7337–7348 30 Goo, L. et al. (2014) Early development of broadly neutralizing antibodies in HIV-1-infected infants. Nat. Med. 20, 655–658 10

Trends in Immunology xxx xxxx, Vol. xxx, No. x

31 Georgiev, I.S. et al. (2013) Delineating antibody recognition in polyclonal sera from patterns of HIV-1 isolate neutralization. Science 340, 751–756 32 Tomaras, G.D. et al. (2011) Polyclonal B cell responses to conserved neutralization epitopes in a subset of HIV-1-infected individuals. J. Virol. 85, 11502–11519 33 Walker, L.M. et al. (2010) A limited number of antibody specificities mediate broad and potent serum neutralization in selected HIV-1 infected individuals. PLoS Pathog. 6, e1001028 34 Scheid, J.F. et al. (2009) Broad diversity of neutralizing antibodies isolated from memory B cells in HIV-infected individuals. Nature 458, 636–640 35 Mouquet, H. et al. (2012) Complex-type N-glycan recognition by potent broadly neutralizing HIV antibodies. Proc. Natl. Acad. Sci. U.S.A. 109, E3268–E3277 36 Scheid, J.F. et al. (2011) Sequence and structural convergence of broad and potent HIV antibodies that mimic CD4 binding. Science 333, 1633–1637 37 Bonsignori, M. et al. (2012) Two distinct broadly neutralizing antibody specificities of different clonal lineages in a single HIV-1-infected donor: implications for vaccine design. J. Virol. 86, 4688–4692 38 Klein, F. et al. (2012) Broad neutralization by a combination of antibodies recognizing the CD4 binding site and a new conformational epitope on the HIV-1 envelope protein. J. Exp. Med. 209, 1469–1479 39 Sattentau, Q. (2008) Avoiding the void: cell-to-cell spread of human viruses. Nat. Rev. Microbiol. 6, 815–826 40 Bonsignori, M. et al. (2014) An autoreactive antibody from an SLE/ HIV-1 individual broadly neutralizes HIV-1. J. Clin. Invest. 124, 1835–1843 41 Miller, C.J. et al. (2007) Antiviral antibodies are necessary for control of simian immunodeficiency virus replication. J. Virol. 81, 5024–5035 42 Schmitz, J.E. et al. (2003) Effect of humoral immune responses on controlling viremia during primary infection of rhesus monkeys with simian immunodeficiency virus. J. Virol. 77, 2165–2173 43 Huang, K.H. et al. (2010) B-cell depletion reveals a role for antibodies in the control of chronic HIV-1 infection. Nat. Commun. 1, 102 44 Martinez, V. et al. (2005) Combination of HIV-1-specific CD4 Th1 cell responses and IgG2 antibodies is the best predictor for persistence of long-term nonprogression. J. Infect. Dis. 191, 2053–2063 45 Ngo-Giang-Huong, N. et al. (2001) HIV type 1-specific IgG2 antibodies: markers of helper T cell type 1 response and prognostic marker of long-term nonprogression. AIDS Res. Hum. Retroviruses 17, 1435–1446 46 Mabuka, J. et al. (2012) HIV-specific antibodies capable of ADCC are common in breastmilk and are associated with reduced risk of transmission in women with high viral loads. PLoS Pathog. 8, e1002739 47 Ackerman, M.E. et al. (2013) Natural variation in Fc glycosylation of HIV-specific antibodies impacts antiviral activity. J. Clin. Invest. 123, 2183–2192 48 Baum, L.L. et al. (1996) HIV-1 gp120-specific antibody-dependent cellmediated cytotoxicity correlates with rate of disease progression. J. Immunol. 157, 2168–2173 49 Hessell, A.J. et al. (2007) Fc receptor but not complement binding is important in antibody protection against HIV. Nature 449, 101–104 50 Halper-Stromberg, A. et al. (2014) Broadly neutralizing antibodies and viral inducers decrease rebound from HIV-1 latent reservoirs in humanized mice. Cell 158, 989–999 51 Kaetzel, C.S. (2007) Mucosal Immune Defense: Immunoglobulin A, Springer 52 Yates, N.L. et al. (2013) HIV-1 gp41 envelope IgA is frequently elicited after transmission but has an initial short response half-life. Mucosal Immunol. 6, 692–703 53 Levesque, M.C. et al. (2009) Polyclonal B cell differentiation and loss of gastrointestinal tract germinal centers in the earliest stages of HIV1 infection. PLoS Med. 6, e1000107 54 Mehandru, S. et al. (2004) Primary HIV-1 infection is associated with preferential depletion of CD4+ T lymphocytes from effector sites in the gastrointestinal tract. J. Exp. Med. 200, 761–770 55 Chaoul, N. et al. (2012) Default in plasma and intestinal IgA responses during acute infection by simian immunodeficiency virus. Retrovirology 9, 43

TREIMM-1124; No. of Pages 13

Feature Review 56 Alfsen, A. et al. (2001) Secretory IgA specific for a conserved epitope on gp41 envelope glycoprotein inhibits epithelial transcytosis of HIV-1. J. Immunol. 166, 6257–6265 57 Bomsel, M. et al. (1998) Intracellular neutralization of HIV transcytosis across tight epithelial barriers by anti-HIV envelope protein dIgA or IgM. Immunity 9, 277–287 58 Devito, C. et al. (2000) Mucosal and plasma IgA from HIV-1-exposed uninfected individuals inhibit HIV-1 transcytosis across human epithelial cells. J. Immunol. 165, 5170–5176 59 Friedman, J. et al. (2012) Isolation of HIV-1-neutralizing mucosal monoclonal antibodies from human colostrum. PLoS ONE 7, e37648 60 Tudor, D. et al. (2009) HIV-1 gp41-specific monoclonal mucosal IgAs derived from highly exposed but IgG-seronegative individuals block HIV-1 epithelial transcytosis and neutralize CD4(+) cell infection: an IgA gene and functional analysis. Mucosal Immunol. 2, 412–426 61 Bomsel, M. et al. (2011) Immunization with HIV-1 gp41 subunit virosomes induces mucosal antibodies protecting nonhuman primates against vaginal SHIV challenges. Immunity 34, 269–280 62 Kardava, L. et al. (2014) Abnormal B cell memory subsets dominate HIV-specific responses in infected individuals. J. Clin. Invest. 124, 3252–3262 63 Moir, S. and Fauci, A.S. (2013) Insights into B cells and HIV-specific B-cell responses in HIV-infected individuals. Immunol. Rev. 254, 207– 224 64 Jelicic, K. et al. (2013) The HIV-1 envelope protein gp120 impairs B cell proliferation by inducing TGF-beta1 production and FcRL4 expression. Nat. Immunol. 14, 1256–1265 65 Pissani, F. and Streeck, H. (2014) Emerging concepts on T follicular helper cell dynamics in HIV infection. Trends Immunol. 35, 278–286 66 Mascola, J.R. and Montefiori, D.C. (2003) HIV-1: nature’s master of disguise. Nat. Med. 9, 393–394 67 Binley, J.M. et al. (2010) Role of complex carbohydrates in human immunodeficiency virus type 1 infection and resistance to antibody neutralization. J. Virol. 84, 5637–5655 68 Moore, P.L. et al. (2012) Evolution of an HIV glycan-dependent broadly neutralizing antibody epitope through immune escape. Nat. Med. 18, 1688–1692 69 Wardemann, H. et al. (2003) Predominant autoantibody production by early human B cell precursors. Science 301, 1374–1377 70 Kwong, P.D. et al. (2002) HIV-1 evades antibody-mediated neutralization through conformational masking of receptor-binding sites. Nature 420, 678–682 71 Labrijn, A.F. et al. (2003) 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. 77, 10557–10565 72 Frey, G. et al. (2008) A fusion-intermediate state of HIV-1 gp41 targeted by broadly neutralizing antibodies. Proc. Natl. Acad. Sci. U.S.A. 105, 3739–3744 73 Poignard, P. et al. (2003) Heterogeneity of envelope molecules expressed on primary human immunodeficiency virus type 1 particles as probed by the binding of neutralizing and nonneutralizing antibodies. J. Virol. 77, 353–365 74 Moore, P.L. et al. (2006) Nature of nonfunctional envelope proteins on the surface of human immunodeficiency virus type 1. J. Virol. 80, 2515–2528 75 Zhu, P. et al. (2006) Distribution and three-dimensional structure of AIDS virus envelope spikes. Nature 441, 847–852 76 Klein, J.S. et al. (2009) Examination of the contributions of size and avidity to the neutralization mechanisms of the anti-HIV antibodies b12 and 4E10. Proc. Natl. Acad. Sci. U.S.A. 106, 7385–7390 77 Gorny, M.K. et al. (1993) Repertoire of neutralizing human monoclonal antibodies specific for the V3 domain of HIV-1 gp120. J. Immunol. 150, 635–643 78 Thali, M. et al. (1993) Characterization of conserved human immunodeficiency virus type 1 gp120 neutralization epitopes exposed upon gp120-CD4 binding. J. Virol. 67, 3978–3988 79 Burton, D.R. et al. (1991) 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. 88, 10134–10137

Trends in Immunology xxx xxxx, Vol. xxx, No. x

80 Trkola, A. et al. (1996) Human monoclonal antibody 2G12 defines a distinctive neutralization epitope on the gp120 glycoprotein of human immunodeficiency virus type 1. J. Virol. 70, 1100–1108 81 Buchacher, A. et al. (1994) Generation of human monoclonal antibodies against HIV-1 proteins; electrofusion and Epstein-Barr virus transformation for peripheral blood lymphocyte immortalization. AIDS Res. Hum. Retroviruses 10, 359–369 82 Muster, T. et al. (1993) A conserved neutralizing epitope on gp41 of human immunodeficiency virus type 1. J. Virol. 67, 6642–6647 83 West, A.P., Jr et al. (2014) Structural insights on the role of antibodies in HIV-1 Vaccine and therapy. Cell 156, 633–648 84 Meffre, E. and Wardemann, H. (2008) B-cell tolerance checkpoints in health and autoimmunity. Curr. Opin. Immunol. 20, 632–638 85 Walker, L.M. et al. (2009) Broad and potent neutralizing antibodies from an African donor reveal a new HIV-1 vaccine target. Science 326, 285–289 86 Bonsignori, M. et al. (2011) Analysis of a clonal lineage of HIV-1 envelope V2/V3 conformational epitope-specific broadly neutralizing antibodies and their inferred unmutated common ancestors. J. Virol. 85, 9998–10009 87 Scheid, J.F. et al. (2009) A method for identification of HIV gp140 binding memory B cells in human blood. J. Immunol. Methods 343, 65–67 88 Wu, X. et al. (2010) Rational design of envelope identifies broadly neutralizing human monoclonal antibodies to HIV-1. Science 329, 856–861 89 Yang, X. et al. (2000) Characterization of stable, soluble trimers containing complete ectodomains of human immunodeficiency virus type 1 envelope glycoproteins. J. Virol. 74, 5716–5725 90 Julien, J.P. et al. (2012) Structural insights into key sites of vulnerability on HIV-1 Env and influenza HA. Immunol. Rev. 250, 180–198 91 Blattner, C. et al. (2014) Structural delineation of a quaternary, cleavage-dependent epitope at the gp41-gp120 interface on intact HIV-1 Env trimers. Immunity 40, 669–680 92 Falkowska, E. et al. (2014) Broadly neutralizing HIV antibodies define a glycan-dependent epitope on the prefusion conformation of gp41 on cleaved envelope trimers. Immunity 40, 657–668 93 Scharf, L. et al. (2014) Antibody 8ANC195 reveals a site of broad vulnerability on the HIV-1 envelope spike. Cell Rep. 7, 785–795 94 Mouquet, H. et al. (2011) Memory B cell antibodies to HIV-1 gp140 cloned from individuals infected with clade A and B viruses. PLoS ONE 6, e24078 95 Klein, F. et al. (2013) Somatic mutations of the immunoglobulin framework are generally required for broad and potent HIV-1 neutralization. Cell 153, 126–138 96 Mouquet, H. et al. (2010) Polyreactivity increases the apparent affinity of anti-HIV antibodies by heteroligation. Nature 467, 591–595 97 Zhou, T. et al. (2013) Multidonor analysis reveals structural elements, genetic determinants, and maturation pathway for HIV-1 neutralization by VRC01-class antibodies. Immunity 39, 245–258 98 Mouquet, H. and Nussenzweig, M.C. (2012) Polyreactive antibodies in adaptive immune responses to viruses. Cell. Mol. Life Sci. 69, 1435– 1445 99 Alam, S.M. et al. (2007) The role of antibody polyspecificity and lipid reactivity in binding of broadly neutralizing anti-HIV-1 envelope human monoclonal antibodies 2F5 and 4E10 to glycoprotein 41 membrane proximal envelope epitopes. J. Immunol. 178, 4424– 4435 100 Dennison, S.M. et al. (2011) Nonneutralizing HIV-1 gp41 envelope cluster II human monoclonal antibodies show polyreactivity for binding to phospholipids and protein autoantigens. J. Virol. 85, 1340–1347 101 Morris, L. et al. (2011) Isolation of a human anti-HIV gp41 membrane proximal region neutralizing antibody by antigen-specific single B cell sorting. PLoS ONE 6, e23532 102 Zhu, Z. et al. (2011) Cross-reactive HIV-1-neutralizing human monoclonal antibodies identified from a patient with 2F5-like antibodies. J. Virol. 85, 11401–11408 103 Liao, H.X. et al. (2013) Co-evolution of a broadly neutralizing HIV-1 antibody and founder virus. Nature 496, 469–476

11

TREIMM-1124; No. of Pages 13

Feature Review 104 Verkoczy, L. and Diaz, M. (2014) Autoreactivity in HIV-1 broadly neutralizing antibodies: implications for their function and induction by vaccination. Curr. Opin. HIV AIDS 9, 224–234 105 Liao, H.X. et al. (2011) Initial antibodies binding to HIV-1 gp41 in acutely infected subjects are polyreactive and highly mutated. J. Exp. Med. 208, 2237–2249 106 Walker, B.D. and Yu, X.G. (2013) Unravelling the mechanisms of durable control of HIV-1. Nat. Rev. Immunol. 13, 487–498 107 Zhang, Y. et al. (2013) The potential of the human immune system to develop broadly neutralizing HIV-1 antibodies: implications for vaccine development. Aids 27, 2529–2539 108 Doria-Rose, N.A. et al. (2009) Frequency and phenotype of human immunodeficiency virus envelope-specific B cells from patients with broadly cross-neutralizing antibodies. J. Virol. 83, 188–199 109 O’Connell, K.A. et al. (2010) Control of HIV-1 in elite suppressors despite ongoing replication and evolution in plasma virus. J. Virol. 84, 7018–7028 110 Doria-Rose, N.A. et al. (2014) Developmental pathway for potent V1V2-directed HIV-neutralizing antibodies. Nature 509, 55–62 111 Gao, F. et al. (2014) Cooperation of B cell lineages in induction of HIV1-broadly neutralizing antibodies. Cell 158, 481–491 112 Victora, G.D. and Nussenzweig, M.C. (2012) Germinal centers. Annu. Rev. Immunol. 30, 429–457 113 Shulman, Z. et al. (2013) T follicular helper cell dynamics in germinal centers. Science 341, 673–677 114 Streeck, H. et al. (2013) Harnessing CD4(+) T cell responses in HIV vaccine development. Nat. Med. 19, 143–149 115 Locci, M. et al. (2013) Human circulating PD-(+)1CXCR3(-)CXCR5(+) memory Tfh cells are highly functional and correlate with broadly neutralizing HIV antibody responses. Immunity 39, 758–769 116 Gitlin, A.D. et al. (2014) Clonal selection in the germinal centre by regulated proliferation and hypermutation. Nature 509, 637–640 117 Mouquet, H. and Nussenzweig, M.C. (2013) HIV: roadmaps to a vaccine. Nature 496, 441–442 118 Georgiou, G. et al. (2014) The promise and challenge of highthroughput sequencing of the antibody repertoire. Nat. Biotechnol. 32, 158–168 119 Wu, X. et al. (2011) Focused evolution of HIV-1 neutralizing antibodies revealed by structures and deep sequencing. Science 333, 1593–1602 120 Sok, D. et al. (2013) The effects of somatic hypermutation on neutralization and binding in the PGT121 family of broadly neutralizing HIV antibodies. PLoS Pathog. 9, e1003754 121 Zhu, J. et al. (2012) Somatic populations of PGT135-137 HIV-1neutralizing antibodies identified by 454 pyrosequencing and bioinformatics. Front. Microbiol. 3, 315 122 Zhu, J. et al. (2013) Mining the antibodyome for HIV-1-neutralizing antibodies with next-generation sequencing and phylogenetic pairing of heavy/light chains. Proc. Natl. Acad. Sci. U.S.A. 110, 6470–6475 123 Haynes, B.F. et al. (2012) B-cell-lineage immunogen design in vaccine development with HIV-1 as a case study. Nat. Biotechnol. 30, 423–433 124 McGuire, A.T. et al. (2013) Engineering HIV envelope protein to activate germline B cell receptors of broadly neutralizing anti-CD4 binding site antibodies. J. Exp. Med. 210, 655–663 125 Jardine, J. et al. (2013) Rational HIV immunogen design to target specific germline B cell receptors. Science 340, 711–716 126 Trama, A.M. et al. (2014) HIV-1 envelope gp41 antibodies can originate from terminal ileum B cells that share cross-reactivity with commensal bacteria. Cell Host Microbe 16, 215–226 127 Wardemann, H. and Nussenzweig, M.C. (2007) B-cell self-tolerance in humans. Adv. Immunol. 95, 83–110 128 Haynes, B.F. et al. (2005) Antibody polyspecificity and neutralization of HIV-1: a hypothesis. Hum. Antibodies 14, 59–67 129 Verkoczy, L. et al. (2011) Role of immune mechanisms in induction of HIV-1 broadly neutralizing antibodies. Curr. Opin. Immunol. 23, 383–390 130 Chen, Y. et al. (2013) Common tolerance mechanisms, but distinct cross-reactivities associated with gp41 and lipids, limit production of HIV-1 broad neutralizing antibodies 2F5 and 4E10. J. Immunol. 191, 1260–1275 131 Verkoczy, L. et al. (2011) Rescue of HIV-1 broad neutralizing antibody-expressing B Cells in 2F5 VH  VL knockin mice reveals multiple tolerance controls. J. Immunol. 187, 3785–3797 12

Trends in Immunology xxx xxxx, Vol. xxx, No. x

132 Verkoczy, L. et al. (2010) Autoreactivity in an HIV-1 broadly reactive neutralizing antibody variable region heavy chain induces immunologic tolerance. Proc. Natl. Acad. Sci. U.S.A. 107, 181–186 133 Tiller, T. et al. (2007) Autoreactivity in human IgG+ memory B cells. Immunity 26, 205–213 134 Yang, G. et al. (2013) Identification of autoantigens recognized by the 2F5 and 4E10 broadly neutralizing HIV-1 antibodies. J. Exp. Med. 210, 241–256 135 Verkoczy, L. et al. (2013) Induction of HIV-1 broad neutralizing antibodies in 2F5 knock-in mice: selection against membrane proximal external region-associated autoreactivity limits Tdependent responses. J. Immunol. 191, 2538–2550 136 Mouquet, H. et al. (2012) Enhanced HIV-1 neutralization by antibody heteroligation. Proc. Natl. Acad. Sci. U.S.A. 109, 875–880 137 Heyndrickx, L. et al. (2012) International network for comparison of HIV neutralization assays: the NeutNet report II. PLoS ONE 7, e36438 138 Bouvin-Pley, M. et al. (2013) Evidence for a continuous drift of the HIV-1 species towards higher resistance to neutralizing antibodies over the course of the epidemic. PLoS Pathog. 9, e1003477 139 Bunnik, E.M. et al. (2010) Adaptation of HIV-1 envelope gp120 to humoral immunity at a population level. Nat. Med. 16, 995–997 140 Euler, Z. et al. (2011) 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. 85, 7236–7245 141 Martin, N. et al. (2010) Virological synapse-mediated spread of human immunodeficiency virus type 1 between T cells is sensitive to entry inhibition. J. Virol. 84, 3516–3527 142 Malbec, M. et al. (2013) Broadly neutralizing antibodies that inhibit HIV-1 cell to cell transmission. J. Exp. Med. 210, 2813–2821 143 Duncan, C.J. et al. (2013) High multiplicity HIV-1 infection and neutralizing antibody evasion mediated by the macrophage-T cell virological synapse. J. Virol. 88, 2025–2034 144 Su, B. et al. (2012) Neutralizing antibodies inhibit HIV-1 transfer from primary dendritic cells to autologous CD4 T lymphocytes. Blood 120, 3708–3717 145 Hur, E.M. et al. (2012) Inhibitory effect of HIV-specific neutralizing IgA on mucosal transmission of HIV in humanized mice. Blood 120, 4571–4582 146 Mantis, N.J. et al. (2007) Inhibition of HIV-1 infectivity and epithelial cell transfer by human monoclonal IgG and IgA antibodies carrying the b12 V region. J. Immunol. 179, 3144–3152 147 Shen, R. et al. (2010) GP41-specific antibody blocks cell-free HIV type 1 transcytosis through human rectal mucosa and model colonic epithelium. J. Immunol. 184, 3648–3655 148 Watkins, J.D. et al. (2013) Anti-HIV IgA isotypes: differential virion capture and inhibition of transcytosis are linked to prevention of mucosal R5 SHIV transmission. Aids 27, F13–F20 149 Hessell, A.J. et al. (2009) Effective, low-titer antibody protection against low-dose repeated mucosal SHIV challenge in macaques. Nat. Med. 15, 951–954 150 Hessell, A.J. et al. (2009) Broadly neutralizing human anti-HIV antibody 2G12 is effective in protection against mucosal SHIV challenge even at low serum neutralizing titers. PLoS Pathog. 5, e1000433 151 Hessell, A.J. et al. (2010) Broadly neutralizing monoclonal antibodies 2F5 and 4E10 directed against the human immunodeficiency virus type 1 gp41 membrane-proximal external region protect against mucosal challenge by simian-human immunodeficiency virus SHIVBa-L. J. Virol. 84, 1302–1313 152 Moog, C. et al. (2014) Protective effect of vaginal application of neutralizing and nonneutralizing inhibitory antibodies against vaginal SHIV challenge in macaques. Mucosal Immunol. 7, 46–56 153 Watkins, J.D. et al. (2011) 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 6, e18207 154 Mascola, J.R. et al. (2000) Protection of macaques against vaginal transmission of a pathogenic HIV-1/SIV chimeric virus by passive infusion of neutralizing antibodies. Nat. Med. 6, 207–210 155 Chomont, N. et al. (2008) Neutralizing monoclonal antibodies to human immunodeficiency virus type 1 do not inhibit viral transcytosis through mucosal epithelial cells. Virology 370, 246–254

TREIMM-1124; No. of Pages 13

Feature Review 156 Gupta, S. et al. (2013) The Neonatal Fc receptor (FcRn) enhances human immunodeficiency virus type 1 (HIV-1) transcytosis across epithelial cells. PLoS Pathog. 9, e1003776 157 Ko, S.Y. et al. (2014) Enhanced neonatal Fc receptor function improves protection against primate SHIV infection. Nature http:// dx.doi.org/10.1038/nature13612 158 Baba, T.W. et al. (2000) Human neutralizing monoclonal antibodies of the IgG1 subtype protect against mucosal simian-human immunodeficiency virus infection. Nat. Med. 6, 200–206 159 Mascola, J.R. et al. (1999) Protection of macaques against pathogenic simian/human immunodeficiency virus 89.6PD by passive transfer of neutralizing antibodies. J. Virol. 73, 4009–4018 160 Ng, C.T. et al. (2010) Passive neutralizing antibody controls SHIV viremia and enhances B cell responses in infant macaques. Nat. Med. 16, 1117–1119 161 Parren, P.W. et al. (2001) Antibody protects macaques against vaginal challenge with a pathogenic R5 simian/human immunodeficiency virus at serum levels giving complete neutralization in vitro. J. Virol. 75, 8340–8347 162 Shibata, R. et al. (1999) Neutralizing antibody directed against the HIV-1 envelope glycoprotein can completely block HIV-1/SIV chimeric virus infections of macaque monkeys. Nat. Med. 5, 204–210 163 Horwitz, J.A. et al. (2013) HIV-1 suppression and durable control by combining single broadly neutralizing antibodies and antiretroviral drugs in humanized mice. Proc. Natl. Acad. Sci. U.S.A. 110, 16538– 16543 164 Klein, F. et al. (2012) HIV therapy by a combination of broadly neutralizing antibodies in humanized mice. Nature 492, 118–122 165 Moldt, B. et al. (2012) Highly potent HIV-specific antibody neutralization in vitro translates into effective protection against mucosal SHIV challenge in vivo. Proc. Natl. Acad. Sci. U.S.A. 109, 18921–18925 166 Shingai, M. et al. (2013) Antibody-mediated immunotherapy of macaques chronically infected with SHIV suppresses viraemia. Nature 503, 277–280

Trends in Immunology xxx xxxx, Vol. xxx, No. x

167 Barouch, D.H. et al. (2013) Therapeutic efficacy of potent neutralizing HIV-1-specific monoclonal antibodies in SHIV-infected rhesus monkeys. Nature 503, 224–228 168 Mehandru, S. et al. (2007) Adjunctive passive immunotherapy in human immunodeficiency virus type 1-infected individuals treated with antiviral therapy during acute and early infection. J. Virol. 81, 11016–11031 169 Trkola, A. et al. (2005) Delay of HIV-1 rebound after cessation of antiretroviral therapy through passive transfer of human neutralizing antibodies. Nat. Med. 11, 615–622 170 Voronin, Y. et al. (2014) HIV monoclonal antibodies: a new opportunity to further reduce mother-to-child HIV transmission. PLoS Med. 11, e1001616 171 Julien, J.P. et al. (2013) Crystal structure of a soluble cleaved HIV-1 envelope trimer. Science 342, 1477–1483 172 Liu, J. et al. (2008) Molecular architecture of native HIV-1 gp120 trimers. Nature 455, 109–113 173 Pettersen, E.F. et al. (2004) UCSF Chimera – a visualization system for exploratory research and analysis. J. Comput. Chem. 25, 1605– 1612 174 Julien, J.P. et al. (2013) Asymmetric recognition of the HIV-1 trimer by broadly neutralizing antibody PG9. Proc. Natl. Acad. Sci. U.S.A. 110, 4351–4356 175 McLellan, J.S. et al. (2011) Structure of HIV-1 gp120 V1/V2 domain with broadly neutralizing antibody PG9. Nature 480, 336–343 176 Pejchal, R. et al. (2011) A potent and broad neutralizing antibody recognizes and penetrates the HIV glycan shield. Science 334, 1097–1103 177 Zhou, T. et al. (2010) Structural basis for broad and potent neutralization of HIV-1 by antibody VRC01. Science 329, 811–817 178 Schro¨dinger, L. (2011) The PyMOL Molecular Graphics System. (1.2r3pre edn), The PyMOL Molecular Graphics System 179 Zolla-Pazner, S. (2014) A critical question for HIV vaccine development: which antibodies to induce? Science 345, 167–168 180 Haynes, B.F. et al. (2005) Cardiolipin polyspecific autoreactivity in two broadly neutralizing HIV-1 antibodies. Science 308, 1906–1908

13

Antibody B cell responses in HIV-1 infection.

In rare cases, B cells can supply HIV-1-infected individuals with unconventional antibodies equipped to neutralize the wide diversity of viral variant...
2MB Sizes 0 Downloads 7 Views