Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Rui Kong is active.

Publication


Featured researches published by Rui Kong.


Nature | 2014

Developmental pathway for potent V1V2-directed HIV-neutralizing antibodies

Nicole A. Doria-Rose; Chaim A. Schramm; Jason Gorman; Penny L. Moore; Jinal N. Bhiman; Brandon J. DeKosky; Michael J. Ernandes; Ivelin S. Georgiev; Helen J. Kim; Marie Pancera; Ryan P. Staupe; Han R. Altae-Tran; Robert T. Bailer; Ema T. Crooks; Albert Cupo; Aliaksandr Druz; Nigel Garrett; Kam Hon Hoi; Rui Kong; Mark K. Louder; Nancy S. Longo; Krisha McKee; Molati Nonyane; Sijy O’Dell; Ryan S. Roark; Rebecca S. Rudicell; Stephen D. Schmidt; Daniel J. Sheward; Cinque Soto; Constantinos Kurt Wibmer

Antibodies capable of neutralizing HIV-1 often target variable regions 1 and 2 (V1V2) of the HIV-1 envelope, but the mechanism of their elicitation has been unclear. Here we define the developmental pathway by which such antibodies are generated and acquire the requisite molecular characteristics for neutralization. Twelve somatically related neutralizing antibodies (CAP256-VRC26.01–12) were isolated from donor CAP256 (from the Centre for the AIDS Programme of Research in South Africa (CAPRISA)); each antibody contained the protruding tyrosine-sulphated, anionic antigen-binding loop (complementarity-determining region (CDR) H3) characteristic of this category of antibodies. Their unmutated ancestor emerged between weeks 30–38 post-infection with a 35-residue CDR H3, and neutralized the virus that superinfected this individual 15 weeks after initial infection. Improved neutralization breadth and potency occurred by week 59 with modest affinity maturation, and was preceded by extensive diversification of the virus population. HIV-1 V1V2-directed neutralizing antibodies can thus develop relatively rapidly through initial selection of B cells with a long CDR H3, and limited subsequent somatic hypermutation. These data provide important insights relevant to HIV-1 vaccine development.


Cell | 2015

Structural Repertoire of HIV-1-Neutralizing Antibodies Targeting the CD4 Supersite in 14 Donors.

Tongqing Zhou; Rebecca M. Lynch; Lei Chen; Priyamvada Acharya; Xueling Wu; Nicole A. Doria-Rose; M. Gordon Joyce; Daniel Lingwood; Cinque Soto; Robert T. Bailer; Michael J. Ernandes; Rui Kong; Nancy S. Longo; Mark K. Louder; Krisha McKee; Sijy O’Dell; Stephen D. Schmidt; Lillian Tran; Zhongjia Yang; Aliaksandr Druz; Timothy S. Luongo; Stephanie Moquin; Sanjay Srivatsan; Yongping Yang; Baoshan Zhang; Anqi Zheng; Marie Pancera; Tatsiana Kirys; Ivelin S. Georgiev; Tatyana Gindin

The site on the HIV-1 gp120 glycoprotein that binds the CD4 receptor is recognized by broadly reactive antibodies, several of which neutralize over 90% of HIV-1 strains. To understand how antibodies achieve such neutralization, we isolated CD4-binding-site (CD4bs) antibodies and analyzed 16 co-crystal structures -8 determined here- of CD4bs antibodies from 14 donors. The 16 antibodies segregated by recognition mode and developmental ontogeny into two types: CDR H3-dominated and VH-gene-restricted. Both could achieve greater than 80% neutralization breadth, and both could develop in the same donor. Although paratope chemistries differed, all 16 gp120-CD4bs antibody complexes showed geometric similarity, with antibody-neutralization breadth correlating with antibody-angle of approach relative to the most effective antibody of each type. The repertoire for effective recognition of the CD4 supersite thus comprises antibodies with distinct paratopes arrayed about two optimal geometric orientations, one achieved by CDR H3 ontogenies and the other achieved by VH-gene-restricted ontogenies.


Cell | 2016

Maturation Pathway from Germline to Broad HIV-1 Neutralizer of a CD4-Mimic Antibody

Mattia Bonsignori; Tongqing Zhou; Zizhang Sheng; Lei Chen; Feng Gao; M. Gordon Joyce; Gabriel Ozorowski; Gwo-Yu Chuang; Chaim A. Schramm; Kevin Wiehe; S. Munir Alam; Todd Bradley; Morgan A. Gladden; Kwan-Ki Hwang; Sheelah Iyengar; Amit Kumar; Xiaozhi Lu; Kan Luo; Michael C. Mangiapani; Robert Parks; Hongshuo Song; Priyamvada Acharya; Robert T. Bailer; Allen Cao; Aliaksandr Druz; Ivelin S. Georgiev; Young Do Kwon; Mark K. Louder; Baoshan Zhang; Anqi Zheng

Antibodies with ontogenies from VH1-2 or VH1-46-germline genes dominate the broadly neutralizing response against the CD4-binding site (CD4bs) on HIV-1. Here, we define with longitudinal sampling from time-of-infection the development of a VH1-46-derived antibody lineage that matured to neutralize 90% of HIV-1 isolates. Structures of lineage antibodies CH235 (week 41 from time-of-infection, 18% breadth), CH235.9 (week 152, 77%), and CH235.12 (week 323, 90%) demonstrated the maturing epitope to focus on the conformationally invariant portion of the CD4bs. Similarities between CH235 lineage and five unrelated CD4bs lineages in epitope focusing, length-of-time to develop breadth, and extraordinary level of somatic hypermutation suggested commonalities in maturation among all CD4bs antibodies. Fortunately, the required CH235-lineage hypermutation appeared substantially guided by the intrinsic mutability of the VH1-46 gene, which closely resembled VH1-2. We integrated our CH235-lineage findings with a second broadly neutralizing lineage and HIV-1 co-evolution to suggest a vaccination strategy for inducing both lineages.


Science | 2016

Fusion peptide of HIV-1 as a site of vulnerability to neutralizing antibody

Rui Kong; Ke Xu; Tongqing Zhou; Priyamvada Acharya; Thomas Lemmin; Liu K; Gabriel Ozorowski; Cinque Soto; Justin D. Taft; Robert T. Bailer; Evan M. Cale; Lei Chen; Choi Cw; Gwo-Yu Chuang; Nicole A. Doria-Rose; Aliaksandr Druz; Ivelin S. Georgiev; Jason Gorman; Jian-Dong Huang; Michael Gordon Joyce; Mark K. Louder; Xiaochu Ma; Krisha McKee; Sijy O'Dell; Marie Pancera; Yili Yang; Scott C. Blanchard; Walther Mothes; Dennis R. Burton; Wayne C. Koff

An antibody to block viral fusion A small fraction of HIV-1–infected individuals develop broad and potent antibodies that bind the HIV-1 envelope protein (Env). These antibodies recognize a limited set of conserved epitopes on Env, such as Envs host receptor-binding site. Kong et al. now report a neutralizing antibody isolated from an HIV-1–infected individual that binds to the fusion peptide of Env. This is unexpected because viruses often try to mask such key components of their cell entry machinery from antibody attack. Crystal structures of the antibody bound to the fusion peptide and to Env itself define the epitope, provide insight into the specific mechanism of antibody binding, and may inform HIV-1 vaccine design. Science, this issue p. 828 A neutralizing antibody against HIV-1 unexpectedly targets a key component of the virus’ cell entry machinery. The HIV-1 fusion peptide, comprising 15 to 20 hydrophobic residues at the N terminus of the Env-gp41 subunit, is a critical component of the virus-cell entry machinery. Here, we report the identification of a neutralizing antibody, N123-VRC34.01, which targets the fusion peptide and blocks viral entry by inhibiting conformational changes in gp120 and gp41 subunits of Env required for entry. Crystal structures of N123-VRC34.01 liganded to the fusion peptide, and to the full Env trimer, revealed an epitope consisting of the N-terminal eight residues of the gp41 fusion peptide and glycan N88 of gp120, and molecular dynamics showed that the N-terminal portion of the fusion peptide can be solvent-exposed. These results reveal the fusion peptide to be a neutralizing antibody epitope and thus a target for vaccine design.


Journal of Virology | 2015

Improving neutralization potency and breadth by combining broadly reactive HIV-1 antibodies targeting major neutralization epitopes.

Rui Kong; Mark K. Louder; Kshitij Wagh; Robert T. Bailer; Allan C. deCamp; Kelli M. Greene; Hongmei Gao; Justin D. Taft; Anna Gazumyan; Cassie Liu; Michel C. Nussenzweig; Bette T. Korber; David C. Montefiori; John R. Mascola

ABSTRACT The isolation of broadly neutralizing HIV-1 monoclonal antibodies (MAbs) to distinct epitopes on the viral envelope glycoprotein (Env) provides the potential to use combinations of MAbs for prevention and treatment of HIV-1 infection. Since many of these MAbs have been isolated in the last few years, the potency and breadth of MAb combinations have not been well characterized. In two parallel experiments, we examined the in vitro neutralizing activities of double-, triple-, and quadruple-MAb combinations targeting four distinct epitopes, including the CD4-binding site, the V1V2-glycan region, the V3-glycan supersite, and the gp41 membrane-proximal external region (MPER), using a panel of 125 Env-pseudotyped viruses. All MAb combinations showed substantially improved neutralization breadth compared to the corresponding single MAbs, while the neutralization potency of individual MAbs was maintained. At a 50% inhibitory concentration (IC50) cutoff of 1 μg/ml per antibody, double-MAb combinations neutralized 89 to 98% of viruses, and triple combinations neutralized 98 to 100%. Overall, the improvement of neutralization breadth was closely predicted by an additive-effect model and explained by complementary neutralization profiles of antibodies recognizing distinct epitopes. Subtle but consistent favorable interactions were observed in some MAb combinations, whereas less favorable interactions were observed on a small subset of viruses that are highly sensitive to V3-glycan MAbs. These data demonstrate favorable in vitro combinations of broadly neutralizing HIV-1 MAbs and suggest that such combinations could have utility for HIV-1 prevention and treatment. IMPORTANCE Over the last 5 years, numerous broadly reactive HIV-1-neutralizing MAbs have been isolated from B cells of HIV-1-infected donors. Each of these MAbs binds to one of the major vulnerable sites (epitopes) on the surface of the viral envelope glycoprotein. Since antibodies to distinct viral epitopes could theoretically act together to provide greater potency and breadth of virus neutralization, we tested physical mixtures of double, triple, and quadruple combinations of neutralizing MAbs targeting four major epitopes on HIV-1 Env. When tested together, antibody combinations showed substantially improved neutralization breadth compared to single MAbs. This improvement could be explained by the complementary neutralization profiles of individual MAbs. We further demonstrated that each antibody maintained its full neutralization potency when used in combination with other MAbs. These data provide a rationale for clinical use of antibody-based combinations for HIV-1 prevention and therapy.


Proceedings of the National Academy of Sciences of the United States of America | 2016

Envelope residue 375 substitutions in simian-human immunodeficiency viruses enhance CD4 binding and replication in rhesus macaques.

Hui Li; Shuyi Wang; Rui Kong; Wenge Ding; Fang Hua Lee; Zahra F. Parker; Eunlim Kim; Gerald H. Learn; Paul Hahn; Ben Policicchio; Egidio Brocca-Cofano; Claire Deleage; Xingpei Hao; Gwo Yu Chuang; Jason Gorman; Matthew R. Gardner; Mark G. Lewis; Theodora Hatziioannou; Sampa Santra; Cristian Apetrei; Ivona Pandrea; S. Munir Alam; Hua-Xin Liao; Xiaoying Shen; Georgia D. Tomaras; Michael Farzan; Elena Chertova; Brandon F. Keele; Jacob D. Estes; Jeffrey D. Lifson

Significance Simian–human immunodeficiency viruses (SHIVs) are an invaluable tool for assessing HIV-1 vaccines, developing therapeutic “cure” strategies, and understanding viral immunopathogenesis. However, only limited success has been achieved in creating SHIVs that incorporate HIV-1 envelopes (Envs) that retain the antigenic features of clinically relevant viruses. Here we focus on a critical residue of the CD4-binding region, Env375, which is under strong positive selection across the broad range of primate lentiviruses. We find that genotypic variation of residue 375 allows for the creation of pathogenic SHIVs that retain the antigenicity, tier 2 neutralization sensitivity, and persistence properties characteristic of primary HIV-1 strains. Taken together, our findings suggest a new paradigm for SHIV design and modeling with important applications to HIV-1 vaccine, cure, and pathogenesis research. Most simian–human immunodeficiency viruses (SHIVs) bearing envelope (Env) glycoproteins from primary HIV-1 strains fail to infect rhesus macaques (RMs). We hypothesized that inefficient Env binding to rhesus CD4 (rhCD4) limits virus entry and replication and could be enhanced by substituting naturally occurring simian immunodeficiency virus Env residues at position 375, which resides at a critical location in the CD4-binding pocket and is under strong positive evolutionary pressure across the broad spectrum of primate lentiviruses. SHIVs containing primary or transmitted/founder HIV-1 subtype A, B, C, or D Envs with genotypic variants at residue 375 were constructed and analyzed in vitro and in vivo. Bulky hydrophobic or basic amino acids substituted for serine-375 enhanced Env affinity for rhCD4, virus entry into cells bearing rhCD4, and virus replication in primary rhCD4 T cells without appreciably affecting antigenicity or antibody-mediated neutralization sensitivity. Twenty-four RMs inoculated with subtype A, B, C, or D SHIVs all became productively infected with different Env375 variants—S, M, Y, H, W, or F—that were differentially selected in different Env backbones. Notably, SHIVs replicated persistently at titers comparable to HIV-1 in humans and elicited autologous neutralizing antibody responses typical of HIV-1. Seven animals succumbed to AIDS. These findings identify Env–rhCD4 binding as a critical determinant for productive SHIV infection in RMs and validate a novel and generalizable strategy for constructing SHIVs with Env glycoproteins of interest, including those that in humans elicit broadly neutralizing antibodies or bind particular Ig germ-line B-cell receptors.


PLOS Pathogens | 2016

Optimal Combinations of Broadly Neutralizing Antibodies for Prevention and Treatment of HIV-1 Clade C Infection

Kshitij Wagh; Tanmoy Bhattacharya; Carolyn Williamson; Alex. Robles; Madeleine. Bayne; Jetta Garrity; Michael. Rist; Cecilia Rademeyer; Hyejin Yoon; Alan S. Lapedes; Hongmei Gao; Kelli M. Greene; Mark K. Louder; Rui Kong; Salim Safurdeen. Abdool Karim; Dennis R. Burton; Dan H. Barouch; Michel C. Nussenzweig; John R. Mascola; Lynn Morris; David C. Montefiori; Bette T. Korber; Michael S. Seaman

The identification of a new generation of potent broadly neutralizing HIV-1 antibodies (bnAbs) has generated substantial interest in their potential use for the prevention and/or treatment of HIV-1 infection. While combinations of bnAbs targeting distinct epitopes on the viral envelope (Env) will likely be required to overcome the extraordinary diversity of HIV-1, a key outstanding question is which bnAbs, and how many, will be needed to achieve optimal clinical benefit. We assessed the neutralizing activity of 15 bnAbs targeting four distinct epitopes of Env, including the CD4-binding site (CD4bs), the V1/V2-glycan region, the V3-glycan region, and the gp41 membrane proximal external region (MPER), against a panel of 200 acute/early clade C HIV-1 Env pseudoviruses. A mathematical model was developed that predicted neutralization by a subset of experimentally evaluated bnAb combinations with high accuracy. Using this model, we performed a comprehensive and systematic comparison of the predicted neutralizing activity of over 1,600 possible double, triple, and quadruple bnAb combinations. The most promising bnAb combinations were identified based not only on breadth and potency of neutralization, but also other relevant measures, such as the extent of complete neutralization and instantaneous inhibitory potential (IIP). By this set of criteria, triple and quadruple combinations of bnAbs were identified that were significantly more effective than the best double combinations, and further improved the probability of having multiple bnAbs simultaneously active against a given virus, a requirement that may be critical for countering escape in vivo. These results provide a rationale for advancing bnAb combinations with the best in vitro predictors of success into clinical trials for both the prevention and treatment of HIV-1 infection.


Journal of Virology | 2012

Broad and Potent Neutralizing Antibody Responses Elicited in Natural HIV-2 Infection

Rui Kong; Hui Li; Frederic Bibollet-Ruche; Julie M. Decker; Natalie N. Zheng; Geoffrey S. Gottlieb; Nancy B. Kiviat; Papa Salif Sow; Ivelin S. Georgiev; Beatrice H. Hahn; Peter D. Kwong; James E. Robinson; George M. Shaw

ABSTRACT Compared with human immunodeficiency virus type 1 (HIV-1), little is known about the susceptibility of HIV-2 to antibody neutralization. We characterized the potency and breadth of neutralizing antibody (NAb) responses in 64 subjects chronically infected with HIV-2 against three primary HIV-2 strains: HIV-27312A, HIV-2ST, and HIV-2UC1. Surprisingly, we observed in a single-cycle JC53bl-13/TZM-bl virus entry assay median reciprocal 50% inhibitory concentration (IC50) NAb titers of 1.7 × 105, 2.8 × 104, and 3.3 × 104, respectively. A subset of 5 patient plasma samples tested against a larger panel of 17 HIV-2 strains where the extracellular gp160 domain was substituted into the HIV-27312A proviral backbone showed potent neutralization of all but 4 viruses. The specificity of antibody neutralization was confirmed using IgG purified from patient plasma, HIV-2 Envs cloned by single-genome amplification, viruses grown in human CD4+ T cells and tested for neutralization sensitivity on human CD4+ T target cells, and, as negative controls, env-minus viruses pseudotyped with HIV-1, vesicular stomatitis virus, or murine leukemia virus Env glycoproteins. Human monoclonal antibodies (MAbs) specific for HIV-2 V3 (6.10F), V4 (1.7A), CD4 binding site (CD4bs; 6.10B), CD4 induced (CD4i; 1.4H), and membrane-proximal external region (MPER; 4E10) epitopes potently neutralized the majority of 32 HIV-2 strains bearing Envs from 13 subjects. Patient antibodies competed with V3, V4, and CD4bs MAbs for binding to monomeric HIV-2 gp120 at titers that correlated significantly with NAb titers. HIV-2 MPER antibodies did not contribute to neutralization breadth or potency. These findings indicate that HIV-2 Env is highly immunogenic in natural infection, that high-titer broadly neutralizing antibodies are commonly elicited, and that unlike HIV-1, native HIV-2 Env trimers expose multiple broadly cross-reactive epitopes readily accessible to NAbs.


Journal of Virology | 2012

Epitope Mapping of Broadly Neutralizing HIV-2 Human Monoclonal Antibodies

Rui Kong; Hui Li; Ivelin S. Georgiev; Anita Changela; Frederic Bibollet-Ruche; Julie M. Decker; Sarah Rowland-Jones; Assan Jaye; Yongjun Guan; George K. Lewis; Johannes P. M. Langedijk; Beatrice H. Hahn; Peter D. Kwong; James E. Robinson; George M. Shaw

ABSTRACT Recent studies have shown that natural infection by HIV-2 leads to the elicitation of high titers of broadly neutralizing antibodies (NAbs) against primary HIV-2 strains (T. I. de Silva, et al., J. Virol. 86:930–946, 2012; R. Kong, et al., J. Virol. 86:947–960, 2012; G. Ozkaya Sahin, et al., J. Virol. 86:961–971, 2012). Here, we describe the envelope (Env) binding and neutralization properties of 15 anti-HIV-2 human monoclonal antibodies (MAbs), 14 of which were newly generated from 9 chronically infected subjects. All 15 MAbs bound specifically to HIV-2 gp120 monomers and neutralized heterologous primary virus strains HIV-27312A and HIV-2ST. Ten of 15 MAbs neutralized a third heterologous primary virus strain, HIV-2UC1. The median 50% inhibitory concentrations (IC50s) for these MAbs were surprisingly low, ranging from 0.007 to 0.028 μg/ml. Competitive Env binding studies revealed three MAb competition groups: CG-I, CG-II, and CG-III. Using peptide scanning, site-directed mutagenesis, chimeric Env constructions, and single-cycle virus neutralization assays, we mapped the epitope of CG-I antibodies to a linear region in variable loop 3 (V3), the epitope of CG-II antibodies to a conformational region centered on the carboxy terminus of V4, and the epitope(s) of CG-III antibodies to conformational regions associated with CD4- and coreceptor-binding sites. HIV-2 Env is thus highly immunogenic in vivo and elicits antibodies having diverse epitope specificities, high potency, and wide breadth. In contrast to the HIV-1 Env trimer, which is generally well shielded from antibody binding and neutralization, HIV-2 is surprisingly vulnerable to broadly reactive NAbs. The availability of 15 human MAbs targeting diverse HIV-2 Env epitopes can facilitate comparative studies of HIV/SIV Env structure, function, antigenicity, and immunogenicity.


PLOS Pathogens | 2017

Mapping Polyclonal HIV-1 Antibody Responses via Next-Generation Neutralization Fingerprinting.

Nicole A. Doria-Rose; Han R. Altae-Tran; Ryan S. Roark; Stephen D. Schmidt; Matthew S. Sutton; Mark K. Louder; Gwo-Yu Chuang; Robert T. Bailer; Valerie Cortez; Rui Kong; Krisha McKee; Sijy O'Dell; Felicia. Wang; Salim Safurdeen. Abdool Karim; James M. Binley; Mark Connors; Barton F. Haynes; Malcolm A. Martin; David C. Montefiori; Lynn Morris; Julie Overbaugh; Peter D. Kwong; John R. Mascola; Ivelin S. Georgiev

Computational neutralization fingerprinting, NFP, is an efficient and accurate method for predicting the epitope specificities of polyclonal antibody responses to HIV-1 infection. Here, we present next-generation NFP algorithms that substantially improve prediction accuracy for individual donors and enable serologic analysis for entire cohorts. Specifically, we developed algorithms for: (a) selection of optimized virus neutralization panels for NFP analysis, (b) estimation of NFP prediction confidence for each serum sample, and (c) identification of sera with potentially novel epitope specificities. At the individual donor level, the next-generation NFP algorithms particularly improved the ability to detect multiple epitope specificities in a sample, as confirmed both for computationally simulated polyclonal sera and for samples from HIV-infected donors. Specifically, the next-generation NFP algorithms detected multiple specificities in twice as many samples of simulated sera. Further, unlike the first-generation NFP, the new algorithms were able to detect both of the previously confirmed antibody specificities, VRC01-like and PG9-like, in donor CHAVI 0219. At the cohort level, analysis of ~150 broadly neutralizing HIV-infected donor samples suggested a potential connection between clade of infection and types of elicited epitope specificities. Most notably, while 10E8-like antibodies were observed in infections from different clades, an enrichment of such antibodies was predicted for clade B samples. Ultimately, such large-scale analyses of antibody responses to HIV-1 infection can help guide the design of epitope-specific vaccines that are tailored to take into account the prevalence of infecting clades within a specific geographic region. Overall, the next-generation NFP technology will be an important tool for the analysis of broadly neutralizing polyclonal antibody responses against HIV-1.

Collaboration


Dive into the Rui Kong's collaboration.

Top Co-Authors

Avatar

Ivelin S. Georgiev

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Mark K. Louder

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Robert T. Bailer

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Tongqing Zhou

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Aliaksandr Druz

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Gwo-Yu Chuang

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Krisha McKee

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Nicole A. Doria-Rose

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Baoshan Zhang

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Choi Cw

National Institutes of Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge