Network


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

Hotspot


Dive into the research topics where Ivelin S. Georgiev is active.

Publication


Featured researches published by Ivelin S. Georgiev.


Science | 2010

Structural basis for broad and potent neutralization of HIV-1 by antibody VRC01.

Tongqing Zhou; Ivelin S. Georgiev; Xueling Wu; Zhi-Yong Yang; Kaifan Dai; Andrés Finzi; Young Do Kwon; Johannes F. Scheid; Wei Shi; Ling Xu; Yongping Yang; Jiang Zhu; Michel C. Nussenzweig; Joseph Sodroski; Lawrence Shapiro; Gary J. Nabel; John R. Mascola; Peter D. Kwong

Designer Anti-HIV Developing a protective HIV vaccine remains a top global health priority. One strategy to identify potential vaccine candidates is to isolate broadly neutralizing antibodies from infected individuals and then attempt to elicit the same antibody response through vaccination (see the Perspective by Burton and Weiss). Wu et al. (p. 856, published online 8 July) now report the identification of three broadly neutralizing antibodies, isolated from an HIV-1–infected individual, that exhibited great breadth and potency of neutralization and were specific for the co-receptor CD4-binding site of the glycoprotein 120 (gp120), part of the viral Env spike. Zhou et al. (p. 811, published online 8 July) analyzed the crystal structure for one of these antibodies, VRC01, in complex with an HIV-1 gp120. VRC01 focuses its binding onto a conformationally invariant domain that is the site of initial CD4 attachment, which allows the antibody to overcome the glycan and conformational masking that diminishes the neutralization potency of most CD4-binding-site antibodies. The epitopes recognized by these antibodies suggest potential immunogens that can inform vaccine design. A human antibody achieves broad neutralization by binding the viral site of recognition for the primary host receptor, CD4. During HIV-1 infection, antibodies are generated against the region of the viral gp120 envelope glycoprotein that binds CD4, the primary receptor for HIV-1. Among these antibodies, VRC01 achieves broad neutralization of diverse viral strains. We determined the crystal structure of VRC01 in complex with a human immunodeficiency virus HIV-1 gp120 core. VRC01 partially mimics CD4 interaction with gp120. A shift from the CD4-defined orientation, however, focuses VRC01 onto the vulnerable site of initial CD4 attachment, allowing it to overcome the glycan and conformational masking that diminishes the neutralization potency of most CD4-binding-site antibodies. To achieve this recognition, VRC01 contacts gp120 mainly through immunoglobulin V-gene regions substantially altered from their genomic precursors. Partial receptor mimicry and extensive affinity maturation thus facilitate neutralization of HIV-1 by natural human antibodies.


Nature | 2011

Structure of HIV-1 gp120 V1/V2 domain with broadly neutralizing antibody PG9

Jason S. McLellan; Marie Pancera; Chris Carrico; Jason Gorman; Jean-Philippe Julien; Reza Khayat; Robert K. Louder; Robert Pejchal; Mallika Sastry; Kaifan Dai; Sijy O’Dell; Nikita Patel; Syed Shahzad-ul-Hussan; Yongping Yang; Baoshan Zhang; Tongqing Zhou; Jiang Zhu; Jeffrey C. Boyington; Gwo-Yu Chuang; Devan Diwanji; Ivelin S. Georgiev; Young Do Kwon; Doyung Lee; Mark K. Louder; Stephanie Moquin; Stephen D. Schmidt; Zhi-Yong Yang; Mattia Bonsignori; John A. Crump; Saidi Kapiga

Variable regions 1 and 2 (V1/V2) of human immunodeficiency virus-1 (HIV-1) gp120 envelope glycoprotein are critical for viral evasion of antibody neutralization, and are themselves protected by extraordinary sequence diversity and N-linked glycosylation. Human antibodies such as PG9 nonetheless engage V1/V2 and neutralize 80% of HIV-1 isolates. Here we report the structure of V1/V2 in complex with PG9. V1/V2 forms a four-stranded β-sheet domain, in which sequence diversity and glycosylation are largely segregated to strand-connecting loops. PG9 recognition involves electrostatic, sequence-independent and glycan interactions: the latter account for over half the interactive surface but are of sufficiently weak affinity to avoid autoreactivity. The structures of V1/V2-directed antibodies CH04 and PGT145 indicate that they share a common mode of glycan penetration by extended anionic loops. In addition to structurally defining V1/V2, the results thus identify a paradigm of antibody recognition for highly glycosylated antigens, which—with PG9—involves a site of vulnerability comprising just two glycans and a strand.


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.


Nature | 2012

Increased HIV-1 vaccine efficacy against viruses with genetic signatures in Env V2

Morgane Rolland; Paul T. Edlefsen; Brendan B. Larsen; Sodsai Tovanabutra; Eric Sanders-Buell; Tomer Hertz; Allan C. deCamp; Chris Carrico; Sergey Menis; Craig A. Magaret; Hasan Ahmed; Michal Juraska; Lennie Chen; Philip Konopa; Snehal Nariya; Julia N. Stoddard; Kim Wong; Haishuang Zhao; Wenjie Deng; Brandon Maust; Meera Bose; Shana Howell; A Bates; Michelle Lazzaro; Annemarie O'Sullivan; Esther Lei; Andrea Bradfield; Grace Ibitamuno; Vatcharain Assawadarachai; Robert J. O'Connell

The RV144 trial demonstrated 31% vaccine efficacy at preventing human immunodeficiency virus (HIV)-1 infection. Antibodies against the HIV-1 envelope variable loops 1 and 2 (Env V1 and V2) correlated inversely with infection risk. We proposed that vaccine-induced immune responses against V1/V2 would have a selective effect against, or sieve, HIV-1 breakthrough viruses. A total of 936 HIV-1 genome sequences from 44 vaccine and 66 placebo recipients were examined. We show that vaccine-induced immune responses were associated with two signatures in V2 at amino acid positions 169 and 181. Vaccine efficacy against viruses matching the vaccine at position 169 was 48% (confidence interval 18% to 66%; P = 0.0036), whereas vaccine efficacy against viruses mismatching the vaccine at position 181 was 78% (confidence interval 35% to 93%; P = 0.0028). Residue 169 is in a cationic glycosylated region recognized by broadly neutralizing and RV144-derived antibodies. The predicted distance between the two signature sites (21 ± 7 Å) and their match/mismatch dichotomy indicate that multiple factors may be involved in the protection observed in RV144. Genetic signatures of RV144 vaccination in V2 complement the finding of an association between high V1/V2-binding antibodies and reduced risk of HIV-1 acquisition, and provide evidence that vaccine-induced V2 responses plausibly had a role in the partial protection conferred by the RV144 regimen.


Immunity | 2013

Multidonor Analysis Reveals Structural Elements, Genetic Determinants, and Maturation Pathway for HIV-1 Neutralization by VRC01-Class Antibodies.

Tongqing Zhou; Jiang Zhu; Xueling Wu; Stephanie Moquin; Baoshan Zhang; Priyamvada Acharya; Ivelin S. Georgiev; Han R. Altae-Tran; Gwo-Yu Chuang; M. Gordon Joyce; Young Do Kwon; Nancy S. Longo; Mark K. Louder; Timothy S. Luongo; Krisha McKee; Chaim A. Schramm; Jeff Skinner; Yongping Yang; Zhongjia Yang; Z. F. Zhang; Anqi Zheng; Mattia Bonsignori; Barton F. Haynes; Johannes F. Scheid; Michel C. Nussenzweig; Melissa Simek; Dennis R. Burton; Wayne C. Koff; James C. Mullikin; Mark Connors

Antibodies of the VRC01 class neutralize HIV-1, arise in diverse HIV-1-infected donors, and are potential templates for an effective HIV-1 vaccine. However, the stochastic processes that generate repertoires in each individual of >10(12) antibodies make elicitation of specific antibodies uncertain. Here we determine the ontogeny of the VRC01 class by crystallography and next-generation sequencing. Despite antibody-sequence differences exceeding 50%, antibody-gp120 cocrystal structures reveal VRC01-class recognition to be remarkably similar. B cell transcripts indicate that VRC01-class antibodies require few specific genetic elements, suggesting that naive-B cells with VRC01-class features are generated regularly by recombination. Virtually all of these fail to mature, however, with only a few-likely one-ancestor B cell expanding to form a VRC01-class lineage in each donor. Developmental similarities in multiple donors thus reveal the generation of VRC01-class antibodies to be reproducible in principle, thereby providing a framework for attempts to elicit similar antibodies in the general population.


Proceedings of the National Academy of Sciences of the United States of America | 2009

Computational structure-based redesign of enzyme activity

Cheng-Yu Chen; Ivelin S. Georgiev; Amy C. Anderson; Bruce Randall Donald

We report a computational, structure-based redesign of the phenylalanine adenylation domain of the nonribosomal peptide synthetase enzyme gramicidin S synthetase A (GrsA-PheA) for a set of noncognate substrates for which the wild-type enzyme has little or virtually no specificity. Experimental validation of a set of top-ranked computationally predicted enzyme mutants shows significant improvement in the specificity for the target substrates. We further present enhancements to the methodology for computational enzyme redesign that are experimentally shown to result in significant additional improvements in the target substrate specificity. The mutant with the highest activity for a noncognate substrate exhibits 1/6 of the wild-type enzyme/wild-type substrate activity, further confirming the feasibility of our computational approach. Our results suggest that structure-based protein design can identify active mutants different from those selected by evolution.


Nature Structural & Molecular Biology | 2015

Crystal structure, conformational fixation and entry-related interactions of mature ligand-free HIV-1 Env

Young Do Kwon; Marie Pancera; Priyamvada Acharya; Ivelin S. Georgiev; Emma T. Crooks; Jason Gorman; M. Gordon Joyce; Xiaochu Ma; Sandeep Narpala; Cinque Soto; Daniel S. Terry; Yongping Yang; Tongqing Zhou; Goran Ahlsen; Robert T. Bailer; Michael Chambers; Gwo Yu Chuang; Nicole A. Doria-Rose; Aliaksandr Druz; Mark A. Hallen; Adam Harned; Tatsiana Kirys; Mark K. Louder; Sijy O'Dell; Gilad Ofek; Keiko Osawa; Madhu Prabhakaran; Mallika Sastry; Guillaume Stewart-Jones; Jonathan Stuckey

As the sole viral antigen on the HIV-1–virion surface, trimeric Env is a focus of vaccine efforts. Here we present the structure of the ligand-free HIV-1–Env trimer, fix its conformation and determine its receptor interactions. Epitope analyses revealed trimeric ligand-free Env to be structurally compatible with broadly neutralizing antibodies but not poorly neutralizing ones. We coupled these compatibility considerations with binding antigenicity to engineer conformationally fixed Envs, including a 201C 433C (DS) variant specifically recognized by broadly neutralizing antibodies. DS-Env retained nanomolar affinity for the CD4 receptor, with which it formed an asymmetric intermediate: a closed trimer bound by a single CD4 without the typical antigenic hallmarks of CD4 induction. Antigenicity-guided structural design can thus be used both to delineate mechanism and to fix conformation, with DS-Env trimers in virus-like-particle and soluble formats providing a new generation of vaccine antigens.


PLOS Pathogens | 2012

Early Low-Titer Neutralizing Antibodies Impede HIV-1 Replication and Select for Virus Escape

Katharine J. Bar; Chun-Yen Tsao; Shilpa S. Iyer; Julie M. Decker; Yongping Yang; Mattia Bonsignori; Xi Chen; Kwan-Ki Hwang; David C. Montefiori; Hua-Xin Liao; Peter Hraber; William Fischer; Hui Joyce Li; Shuyi Wang; Sarah Sterrett; Brandon F. Keele; Vitaly V. Ganusov; Alan S. Perelson; Bette T. Korber; Ivelin S. Georgiev; Jason S. McLellan; Jeffrey W. Pavlicek; Feng Gao; Barton F. Haynes; Beatrice H. Hahn; Peter D. Kwong; George M. Shaw

Single genome sequencing of early HIV-1 genomes provides a sensitive, dynamic assessment of virus evolution and insight into the earliest anti-viral immune responses in vivo. By using this approach, together with deep sequencing, site-directed mutagenesis, antibody adsorptions and virus-entry assays, we found evidence in three subjects of neutralizing antibody (Nab) responses as early as 2 weeks post-seroconversion, with Nab titers as low as 1∶20 to 1∶50 (IC50) selecting for virus escape. In each of the subjects, Nabs targeted different regions of the HIV-1 envelope (Env) in a strain-specific, conformationally sensitive manner. In subject CH40, virus escape was first mediated by mutations in the V1 region of the Env, followed by V3. HIV-1 specific monoclonal antibodies from this subject mapped to an immunodominant region at the base of V3 and exhibited neutralizing patterns indistinguishable from polyclonal antibody responses, indicating V1–V3 interactions within the Env trimer. In subject CH77, escape mutations mapped to the V2 region of Env, several of which selected for alterations of glycosylation. And in subject CH58, escape mutations mapped to the Env outer domain. In all three subjects, initial Nab recognition was followed by sequential rounds of virus escape and Nab elicitation, with Nab escape variants exhibiting variable costs to replication fitness. Although delayed in comparison with autologous CD8 T-cell responses, our findings show that Nabs appear earlier in HIV-1 infection than previously recognized, target diverse sites on HIV-1 Env, and impede virus replication at surprisingly low titers. The unexpected in vivo sensitivity of early transmitted/founder virus to Nabs raises the possibility that similarly low concentrations of vaccine-induced Nabs could impair virus acquisition in natural HIV-1 transmission, where the risk of infection is low and the number of viruses responsible for transmission and productive clinical infection is typically one.


Science | 2013

Delineating Antibody Recognition in Polyclonal Sera from Patterns of HIV-1 Isolate Neutralization

Ivelin S. Georgiev; Nicole A. Doria-Rose; Tongqing Zhou; Young Do Kwon; Ryan P. Staupe; Stephanie Moquin; Gwo-Yu Chuang; Mark K. Louder; Stephen D. Schmidt; Han R. Altae-Tran; Robert T. Bailer; Krisha McKee; Martha Nason; Sijy O’Dell; Gilad Ofek; Marie Pancera; Sanjay Srivatsan; Lawrence Shapiro; Mark Connors; Stephen A. Migueles; Lynn Morris; Yoshiaki Nishimura; Malcolm A. Martin; John R. Mascola; Peter D. Kwong

Building Better Vaccines In the past few years, several highly potent, broadly neutralizing antibodies (bNAbs) specific for the gp120 envelope protein of HIV-1 have been discovered. The goal of this work is to use this information to inform the design of vaccines that are able to induce such antibodies (see the Perspective by Crowe). However, because of extensive somatic hypermutation, the epitope bound by these antibodies often does not bind to the germline sequence. Jardine et al. (p. 711, published online 28 March; see the cover) used computational analysis and in vitro screening to design an immunogen that could bind to VRC01-class bNAbs and to their germline precursors. Georgiev et al. (p. 751) took advantage of the fact that only four sites on the HIV viral envelope protein seem to bind bNAbs, and sera that contain particular bNAbs show characteristic patterns of neutralization. An algorithm was developed that could successfully delineate the neutralization specificity of antibodies present in polyclonal sera from HIV-infected patients. An algorithm predicts the neutralization specificity of sera from HIV-infected individuals. [Also see Perspective by Crowe] Serum characterization and antibody isolation are transforming our understanding of the humoral immune response to viral infection. Here, we show that epitope specificities of HIV-1–neutralizing antibodies in serum can be elucidated from the serum pattern of neutralization against a diverse panel of HIV-1 isolates. We determined “neutralization fingerprints” for 30 neutralizing antibodies on a panel of 34 diverse HIV-1 strains and showed that similarity in neutralization fingerprint correlated with similarity in epitope. We used these fingerprints to delineate specificities of polyclonal sera from 24 HIV-1–infected donors and a chimeric siman-human immunodeficiency virus–infected macaque. Delineated specificities matched published specificities and were further confirmed by antibody isolation for two sera. Patterns of virus-isolate neutralization can thus afford a detailed epitope-specific understanding of neutralizing-antibody responses to viral infection.


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.

Collaboration


Dive into the Ivelin S. Georgiev's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

John R. Mascola

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

Yongping Yang

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

Tongqing Zhou

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

Aliaksandr Druz

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

M. Gordon Joyce

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Krisha McKee

National Institutes of Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge