Network


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

Hotspot


Dive into the research topics where William R. Schief is active.

Publication


Featured researches published by William R. Schief.


Science | 2010

Rational Design of Envelope Identifies Broadly Neutralizing Human Monoclonal Antibodies to HIV-1

Xueling Wu; Zhi-Yong Yang; Yuxing Li; Carl-Magnus Hogerkorp; William R. Schief; Michael S. Seaman; Tongqing Zhou; Stephen D. Schmidt; Lan Wu; Ling Xu; Nancy S. Longo; Krisha McKee; Sijy O’Dell; Mark K. Louder; Diane Wycuff; Yu Feng; Martha Nason; Nicole A. Doria-Rose; Mark Connors; Peter D. Kwong; Mario Roederer; Richard T. Wyatt; Gary J. Nabel; John R. Mascola

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. Cross-reactive neutralizing antibodies (NAbs) are found in the sera of many HIV-1–infected individuals, but the virologic basis of their neutralization remains poorly understood. We used knowledge of HIV-1 envelope structure to develop antigenically resurfaced glycoproteins specific for the structurally conserved site of initial CD4 receptor binding. These probes were used to identify sera with NAbs to the CD4-binding site (CD4bs) and to isolate individual B cells from such an HIV-1–infected donor. By expressing immunoglobulin genes from individual cells, we identified three monoclonal antibodies, including a pair of somatic variants that neutralized over 90% of circulating HIV-1 isolates. Exceptionally broad HIV-1 neutralization can be achieved with individual antibodies targeted to the functionally conserved CD4bs of glycoprotein 120, an important insight for future HIV-1 vaccine design.


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.


Science | 2011

A potent and broad neutralizing antibody recognizes and penetrates the HIV glycan shield.

Robert Pejchal; Katie J. Doores; Laura M. Walker; Reza Khayat; Po-Ssu Huang; Sheng-Kai Wang; Robyn L. Stanfield; Jean-Philippe Julien; Alejandra Ramos; Matthew Crispin; Rafael S. Depetris; Umesh Katpally; Andre J. Marozsan; Albert Cupo; Sebastien Maloveste; Yan Liu; Ryan McBride; Yukishige Ito; Rogier W. Sanders; Cassandra Ogohara; James C. Paulson; Ten Feizi; Christopher N. Scanlan; Chi-Huey Wong; John P. Moore; William C. Olson; Andrew B. Ward; Pascal Poignard; William R. Schief; Dennis R. Burton

An HIV antibody achieves potency and breadth by binding simultaneously to two conserved glycans on the viral envelope protein. The HIV envelope (Env) protein gp120 is protected from antibody recognition by a dense glycan shield. However, several of the recently identified PGT broadly neutralizing antibodies appear to interact directly with the HIV glycan coat. Crystal structures of antigen-binding fragments (Fabs) PGT 127 and 128 with Man9 at 1.65 and 1.29 angstrom resolution, respectively, and glycan binding data delineate a specific high mannose-binding site. Fab PGT 128 complexed with a fully glycosylated gp120 outer domain at 3.25 angstroms reveals that the antibody penetrates the glycan shield and recognizes two conserved glycans as well as a short β-strand segment of the gp120 V3 loop, accounting for its high binding affinity and broad specificify. Furthermore, our data suggest that the high neutralization potency of PGT 127 and 128 immunoglobulin Gs may be mediated by cross-linking Env trimers on the viral surface.


Science | 2013

Rational HIV Immunogen Design to Target Specific Germline B Cell Receptors

Joseph G. Jardine; Jean-Philippe Julien; Sergey Menis; Takayuki Ota; Oleksandr Kalyuzhniy; Andrew T. McGuire; Devin Sok; Po-Ssu Huang; Skye MacPherson; Meaghan Jones; Travis Nieusma; John C. Mathison; David Baker; Andrew B. Ward; Dennis R. Burton; Leonidas Stamatatos; David Nemazee; Ian A. Wilson; William R. Schief

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. Structural knowledge of broadly neutralizing antibodies against HIV-1 guides the design of an immunogen to elicit them. Vaccine development to induce broadly neutralizing antibodies (bNAbs) against HIV-1 is a global health priority. Potent VRC01-class bNAbs against the CD4 binding site of HIV gp120 have been isolated from HIV-1–infected individuals; however, such bNAbs have not been induced by vaccination. Wild-type gp120 proteins lack detectable affinity for predicted germline precursors of VRC01-class bNAbs, making them poor immunogens to prime a VRC01-class response. We employed computation-guided, in vitro screening to engineer a germline-targeting gp120 outer domain immunogen that binds to multiple VRC01-class bNAbs and germline precursors, and elucidated germline binding crystallographically. When multimerized on nanoparticles, this immunogen (eOD-GT6) activates germline and mature VRC01-class B cells. Thus, eOD-GT6 nanoparticles have promise as a vaccine prime. In principle, germline-targeting strategies could be applied to other epitopes and pathogens.


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.


Cell Host & Microbe | 2012

A Blueprint for HIV Vaccine Discovery

Dennis R. Burton; Rafi Ahmed; Dan H. Barouch; Salvatore T. Butera; Shane Crotty; Adam Godzik; Daniel E. Kaufmann; M. Juliana McElrath; Michel C. Nussenzweig; Bali Pulendran; Chris Scanlan; William R. Schief; Guido Silvestri; Hendrik Streeck; Bruce D. Walker; Laura M. Walker; Andrew B. Ward; Ian A. Wilson; Richard T. Wyatt

Despite numerous attempts over many years to develop an HIV vaccine based on classical strategies, none has convincingly succeeded to date. A number of approaches are being pursued in the field, including building upon possible efficacy indicated by the recent RV144 clinical trial, which combined two HIV vaccines. Here, we argue for an approach based, in part, on understanding the HIV envelope spike and its interaction with broadly neutralizing antibodies (bnAbs) at the molecular level and using this understanding to design immunogens as possible vaccines. BnAbs can protect against virus challenge in animal models, and many such antibodies have been isolated recently. We further propose that studies focused on how best to provide T cell help to B cells that produce bnAbs are crucial for optimal immunization strategies. The synthesis of rational immunogen design and immunization strategies, together with iterative improvements, offers great promise for advancing toward an HIV vaccine.


Proceedings of the National Academy of Sciences of the United States of America | 2010

Structure of HIV-1 gp120 with gp41-interactive region reveals layered envelope architecture and basis of conformational mobility.

Marie Pancera; Shahzad Majeed; Yih-En Andrew Ban; Lei Chen; Chih-chin Huang; Leopold Kong; Young Do Kwon; Jonathan Stuckey; Tongqing Zhou; James E. Robinson; William R. Schief; Joseph Sodroski; Richard T. Wyatt; Peter D. Kwong

The viral spike of HIV-1 is composed of three gp120 envelope glycoproteins attached noncovalently to three gp41 transmembrane molecules. Viral entry is initiated by binding to the CD4 receptor on the cell surface, which induces large conformational changes in gp120. These changes not only provide a model for receptor-triggered entry, but affect spike sensitivity to drug- and antibody-mediated neutralization. Although some of the details of the CD4-induced conformational change have been visualized by crystal structures and cryoelectron tomograms, the critical gp41-interactive region of gp120 was missing from previous atomic-level characterizations. Here we determine the crystal structure of an HIV-1 gp120 core with intact gp41-interactive region in its CD4-bound state, compare this structure to unliganded and antibody-bound forms to identify structurally invariant and plastic components, and use ligand-oriented cryoelectron tomograms to define component mobility in the viral spike context. Newly defined gp120 elements proximal to the gp41 interface complete a 7-stranded β-sandwich, which appeared invariant in conformation. Loop excursions emanating from the sandwich form three topologically separate—and structurally plastic—layers, topped off by the highly glycosylated gp120 outer domain. Crystal structures, cryoelectron tomograms, and interlayer chemistry were consistent with a mechanism in which the layers act as a shape-changing spacer, facilitating movement between outer domain and gp41-associated β-sandwich and providing for conformational diversity used in immune evasion. A “layered” gp120 architecture thus allows movement among alternative glycoprotein conformations required for virus entry and immune evasion, whereas a β-sandwich clamp maintains gp120–gp41 interaction and regulates gp41 transitions.


Proceedings of the National Academy of Sciences of the United States of America | 2010

Elicitation of structure-specific antibodies by epitope scaffolds

Gilad Ofek; F. Javier Guenaga; William R. Schief; Jeff Skinner; David Baker; Richard T. Wyatt; Peter D. Kwong

Elicitation of antibodies against targets that are immunorecessive, cryptic, or transient in their native context has been a challenge for vaccine design. Here we demonstrate the elicitation of structure-specific antibodies against the HIV-1 gp41 epitope of the broadly neutralizing antibody 2F5. This conformationally flexible region of gp41 assumes mostly helical conformations but adopts a kinked, extended structure when bound by antibody 2F5. Computational techniques were employed to transplant the 2F5 epitope into select acceptor scaffolds. The resultant “2F5-epitope scaffolds” possessed nanomolar affinity for antibody 2F5 and a range of epitope flexibilities and antigenic specificities. Crystallographic characterization of the epitope scaffold with highest affinity and antigenic discrimination confirmed good to near perfect attainment of the target conformation for the gp41 molecular graft in free and 2F5-bound states, respectively. Animals immunized with 2F5-epitope scaffolds showed levels of graft-specific immune responses that correlated with graft flexibility (p < 0.04), while antibody responses against the graft—as dissected residue-by-residue with alanine substitutions—resembled more closely those of 2F5 than sera elicited with flexible or cyclized peptides, a resemblance heightened by heterologous prime-boost. Lastly, crystal structures of a gp41 peptide in complex with monoclonal antibodies elicited by the 2F5-epitope scaffolds revealed that the elicited antibodies induce gp41 to assume its 2F5-recognized shape. Epitope scaffolds thus provide a means to elicit antibodies that recognize a predetermined target shape and sequence, even if that shape is transient in nature, and a means by which to dissect factors influencing such elicitation.


Nature | 2014

Proof of principle for epitope-focused vaccine design

Bruno E. Correia; John T. Bates; Rebecca Loomis; Gretchen Baneyx; Christopher Carrico; Joseph G. Jardine; Peter B. Rupert; Colin Correnti; Oleksandr Kalyuzhniy; Vinayak Vittal; Mary J. Connell; Eric Stevens; Alexandria Schroeter; Man Chen; Skye MacPherson; Andreia M. Serra; Yumiko Adachi; Margaret A. Holmes; Yuxing Li; Rachel E. Klevit; Barney S. Graham; Richard T. Wyatt; David Baker; Roland K. Strong; James E. Crowe; Philip R. Johnson; William R. Schief

Vaccines prevent infectious disease largely by inducing protective neutralizing antibodies against vulnerable epitopes. Several major pathogens have resisted traditional vaccine development, although vulnerable epitopes targeted by neutralizing antibodies have been identified for several such cases. Hence, new vaccine design methods to induce epitope-specific neutralizing antibodies are needed. Here we show, with a neutralization epitope from respiratory syncytial virus, that computational protein design can generate small, thermally and conformationally stable protein scaffolds that accurately mimic the viral epitope structure and induce potent neutralizing antibodies. These scaffolds represent promising leads for the research and development of a human respiratory syncytial virus vaccine needed to protect infants, young children and the elderly. More generally, the results provide proof of principle for epitope-focused and scaffold-based vaccine design, and encourage the evaluation and further development of these strategies for a variety of other vaccine targets, including antigenically highly variable pathogens such as human immunodeficiency virus and influenza.


Journal of Experimental Medicine | 2013

Engineering HIV envelope protein to activate germline B cell receptors of broadly neutralizing anti-CD4 binding site antibodies.

Andrew T. McGuire; Sam Hoot; Anita M. Dreyer; Adriana Lippy; Andrew B. Stuart; Kristen W. Cohen; Joseph G. Jardine; Sergey Menis; Johannes F. Scheid; Anthony P. West; William R. Schief; Leonidas Stamatatos

Eliminating key glycosylation sites on HIV envelope (Env) restores binding of the germline versions of known broadly neutralizing anti-Env antibodies.

Collaboration


Dive into the William R. Schief's collaboration.

Top Co-Authors

Avatar

Sergey Menis

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar

Daniel W. Kulp

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar

Dennis R. Burton

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Richard T. Wyatt

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar

Devin Sok

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar

Ian A. Wilson

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar

Peter D. Kwong

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Yumiko Adachi

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar

David Baker

University of Washington

View shared research outputs
Researchain Logo
Decentralizing Knowledge