Network


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

Hotspot


Dive into the research topics where Yaroslav Tsybovsky is active.

Publication


Featured researches published by Yaroslav Tsybovsky.


Science | 2016

Rapid development of a DNA vaccine for Zika virus

Kimberly A. Dowd; Sung-Youl Ko; Kaitlyn M. Morabito; Eun Sung Yang; Rebecca S. Pelc; Christina R. DeMaso; Leda R. Castilho; Peter Abbink; Michael Boyd; Ramya Nityanandam; David N. Gordon; John R. Gallagher; Xuejun Chen; John-Paul Todd; Yaroslav Tsybovsky; Audray K. Harris; Yan-Jang S. Huang; Stephen Higgs; Dana L. Vanlandingham; Hanne Andersen; Mark G. Lewis; Rafael De La Barrera; Kenneth H. Eckels; Richard G. Jarman; Martha Nason; Dan H. Barouch; Mario Roederer; Wing-Pui Kong; John R. Mascola; Theodore C. Pierson

A DNA vaccine candidate for Zika The ongoing Zika epidemic in the Americas and the Caribbean urgently needs a protective vaccine. Two DNA vaccines composed of the genes that encode the structural premembrane and envelope proteins of Zika virus have been tested in monkeys. Dowd et al. show that two doses of vaccine given intramuscularly completely protected 17 of 18 animals against Zika virus challenge. A single low dose of vaccine was not protective but did reduce viral loads. Protection correlated with serum antibody neutralizing activity. Phase I clinical trials testing these vaccines are already ongoing. Science, this issue p. 237 DNA-vaccine–induced neutralizing antibodies largely protect monkeys after experimental challenge by virus infection. Zika virus (ZIKV) was identified as a cause of congenital disease during the explosive outbreak in the Americas and Caribbean that began in 2015. Because of the ongoing fetal risk from endemic disease and travel-related exposures, a vaccine to prevent viremia in women of childbearing age and their partners is imperative. We found that vaccination with DNA expressing the premembrane and envelope proteins of ZIKV was immunogenic in mice and nonhuman primates, and protection against viremia after ZIKV challenge correlated with serum neutralizing activity. These data not only indicate that DNA vaccination could be a successful approach to protect against ZIKV infection, but also suggest a protective threshold of vaccine-induced neutralizing activity that prevents viremia after acute infection.


Nature Medicine | 2016

Molecular-level analysis of the serum antibody repertoire in young adults before and after seasonal influenza vaccination

Jiwon Lee; Daniel R. Boutz; Veronika Chromikova; M. Gordon Joyce; Christopher Vollmers; Kwanyee Leung; Andrew P. Horton; Brandon J. DeKosky; Chang-Han Lee; Jason J. Lavinder; Ellen M. Murrin; Constantine Chrysostomou; Kam Hon Hoi; Yaroslav Tsybovsky; Paul V. Thomas; Aliaksandr Druz; Baoshan Zhang; Yi Zhang; Lingshu Wang; Wing-Pui Kong; Daechan Park; Lyubov Popova; Cornelia L. Dekker; Mark M. Davis; Chalise E. Carter; Ted M. Ross; Andrew D. Ellington; Patrick C. Wilson; Edward M. Marcotte; John R. Mascola

Molecular understanding of serological immunity to influenza has been confounded by the complexity of the polyclonal antibody response in humans. Here we used high-resolution proteomics analysis of immunoglobulin (referred to as Ig-seq) coupled with high-throughput sequencing of transcripts encoding B cell receptors (BCR-seq) to quantitatively determine the antibody repertoire at the individual clonotype level in the sera of young adults before and after vaccination with trivalent seasonal influenza vaccine. The serum repertoire comprised between 40 and 147 clonotypes that were specific to each of the three monovalent components of the trivalent influenza vaccine, with boosted pre-existing clonotypes accounting for ∼60% of the response. An unexpectedly high fraction of serum antibodies recognized both the H1 and H3 monovalent vaccines. Recombinant versions of these H1 + H3 cross-reactive antibodies showed broad binding to hemagglutinins (HAs) from previously circulating virus strains; several of these antibodies, which were prevalent in the serum of multiple donors, recognized the same conserved epitope in the HA head domain. Although the HA-head-specific H1 + H3 antibodies did not show neutralization activity in vitro, they protected mice against infection with the H1N1 and H3N2 virus strains when administered before or after challenge. Collectively, our data reveal unanticipated insights regarding the serological response to influenza vaccination and raise questions about the added benefits of using a quadrivalent vaccine instead of a trivalent vaccine.


Cell | 2016

Vaccine-Induced Antibodies that Neutralize Group 1 and Group 2 Influenza A Viruses.

M. Gordon Joyce; Adam K. Wheatley; Paul V. Thomas; Gwo-Yu Chuang; Cinque Soto; Robert T. Bailer; Aliaksandr Druz; Ivelin S. Georgiev; Rebecca A. Gillespie; Masaru Kanekiyo; Wing-Pui Kong; Kwanyee Leung; Sandeep N. Narpala; Madhu Prabhakaran; Eun Sung Yang; Baoshan Zhang; Yi Zhang; Mangaiarkarasi Asokan; Jeffrey C. Boyington; Tatsiana Bylund; Sam Darko; Christopher R. Lees; Amy Ransier; Chen-Hsiang Shen; Lingshu Wang; James R. R. Whittle; Xueling Wu; Hadi M. Yassine; Celia Santos; Yumiko Matsuoka

Antibodies capable of neutralizing divergent influenza A viruses could form the basis of a universal vaccine. Here, from subjects enrolled in an H5N1 DNA/MIV-prime-boost influenza vaccine trial, we sorted hemagglutinin cross-reactive memory B cells and identified three antibody classes, each capable of neutralizing diverse subtypes of group 1 and group 2 influenza A viruses. Co-crystal structures with hemagglutinin revealed that each class utilized characteristic germline genes and convergent sequence motifs to recognize overlapping epitopes in the hemagglutinin stem. All six analyzed subjects had sequences from at least one multidonor class, and-in half the subjects-multidonor-class sequences were recovered from >40% of cross-reactive B cells. By contrast, these multidonor-class sequences were rare in published antibody datasets. Vaccination with a divergent hemagglutinin can thus increase the frequency of B cells encoding broad influenza A-neutralizing antibodies. We propose the sequence signature-quantified prevalence of these B cells as a metric to guide universal influenza A immunization strategies.


Nature Structural & Molecular Biology | 2016

Iterative structure-based improvement of a fusion-glycoprotein vaccine against RSV

M. Gordon Joyce; Baoshan Zhang; Li Ou; Man Chen; Gwo-Yu Chuang; Aliaksandr Druz; Wing-Pui Kong; Yen-Ting Lai; Emily Rundlet; Yaroslav Tsybovsky; Yongping Yang; Ivelin S. Georgiev; Christopher R. Lees; Marie Pancera; Mallika Sastry; Cinque Soto; Guillaume Stewart-Jones; Paul V. Thomas; Joseph G. Van Galen; Ulrich Baxa; Kelly K. Lee; John R. Mascola; Barney S. Graham; Peter D. Kwong

Structure-based design of vaccines has been a long-sought goal, especially the iterative optimization used so successfully with structure-based design of drugs. We previously developed a 1st-generation vaccine antigen called DS-Cav1, comprising a pre-fusion-stabilized form of the fusion (F) glycoprotein, which elicited high titers of protective responses against respiratory syncytial virus (RSV) in mice and macaques. Here we report the improvement of DS-Cav1 through iterative cycles of structure-based design that significantly increased the titer of RSV-protective responses. The resultant 2nd-generation “DS2”-stabilized immunogens have F subunits genetically linked, fusion peptide deleted, and interprotomer movements stabilized by an additional disulfide bond. These DS2-immunogens are promising vaccine candidates with superior attributes, such as the absence of a requirement for furin cleavage and increased antigenic stability to heat inactivation. The iterative structure-based improvement described here may have utility in the optimization of other vaccine antigens.


Nature microbiology | 2016

Platelet-derived growth factor-α receptor is the cellular receptor for human cytomegalovirus gHgLgO trimer.

Anna Kabanova; Jessica Marcandalli; Tongqing Zhou; Siro Bianchi; Ulrich Baxa; Yaroslav Tsybovsky; Daniele Lilleri; Chiara Silacci-Fregni; Mathilde Foglierini; Blanca Fernandez-Rodriguez; Aliaksandr Druz; Baoshan Zhang; Roger Geiger; Massimiliano Pagani; Federica Sallusto; Peter D. Kwong; Davide Corti; Antonio Lanzavecchia; Laurent Perez

Human cytomegalovirus encodes at least 25 membrane glycoproteins that are found in the viral envelope1. While gB represents the fusion protein, two glycoprotein complexes control the tropism of the virus: the gHgLgO trimer is involved in the infection of fibroblasts, and the gHgLpUL128L pentamer is required for infection of endothelial, epithelial and myeloid cells2–5. Two reports suggested that gB binds to ErbB1 and PDGFRα (refs 6,7); however, these results do not explain the tropism of the virus and were recently challenged8,9. Here, we provide a 19 Å reconstruction for the gHgLgO trimer and show that it binds with high affinity through the gO subunit to PDGFRα, which is expressed on fibroblasts but not on epithelial cells. We also provide evidence that the trimer is essential for viral entry in both fibroblasts and epithelial cells. Furthermore, we identify the pentamer, which is essential for infection of epithelial cells, as a trigger for the ErbB pathway. These findings help explain the broad tropism of human cytomegalovirus and indicate that PDGFRα and the viral gO subunit could be targeted by novel anti-viral therapies.


Journal of Virology | 2017

Structure-Based Design of a Soluble Prefusion-Closed HIV-1 Env Trimer with Reduced CD4 Affinity and Improved Immunogenicity.

Gwo-Yu Chuang; Hui Geng; Marie Pancera; Kai Xu; Cheng Cheng; Priyamvada Acharya; Michael Chambers; Aliaksandr Druz; Yaroslav Tsybovsky; Timothy G. Wanninger; Yongping Yang; Nicole A. Doria-Rose; Ivelin S. Georgiev; Jason Gorman; M. Gordon Joyce; Sijy O'Dell; Tongqing Zhou; Adrian B. McDermott; John R. Mascola; Peter D. Kwong

ABSTRACT The HIV-1 envelope (Env) trimer is a target for vaccine design as well as a conformational machine that facilitates virus entry by transitioning between prefusion-closed, CD4-bound, and coreceptor-bound conformations by transitioning into a postfusion state. Vaccine designers have sought to restrict the conformation of the HIV-1 Env trimer to its prefusion-closed state as this state is recognized by most broadly neutralizing, but not nonneutralizing, antibodies. We previously identified a disulfide bond, I201C-A433C (DS), which stabilizes Env in the vaccine-desired prefusion-closed state. When placed into the context of BG505 SOSIP.664, a soluble Env trimer mimic developed by Sanders, Moore, and colleagues, the engineered DS-SOSIP trimer showed reduced conformational triggering by CD4. Here, we further stabilize DS-SOSIP through a combination of structure-based design and 96-well-based expression and antigenic assessment. From 103 designs, we identified one, named DS-SOSIP.4mut, with four additional mutations at the interface of potentially mobile domains of the prefusion-closed structure. We also determined the crystal structures of DS-SOSIP.4mut at 4.1-Å resolution and of an additional DS-SOSIP.6mut variant at 4.3-Å resolution, and these confirmed the formation of engineered disulfide bonds. Notably, DS-SOSIP.4mut elicited a higher ratio of tier 2 autologous titers versus tier 1 V3-sensitive titers than BG505 SOSIP.664. DS-SOSIP.4mut also showed reduced recognition of CD4 and increased thermostability. The improved antigenicity, thermostability, and immunogenicity of DS-SOSIP.4mut suggest utility as an immunogen or a serologic probe; moreover, the specific four alterations identified here, M154, M300, M302, and L320 (4mut), can also be transferred to other HIV-1 Env trimers of interest to improve their properties. IMPORTANCE One approach to elicit broadly neutralizing antibodies against HIV-1 is to stabilize the structurally flexible HIV-1 envelope (Env) trimer in a conformation that displays predominantly broadly neutralizing epitopes and few to no nonneutralizing epitopes. The prefusion-closed conformation of HIV-1 Env has been identified as one such preferred conformation, and a current leading vaccine candidate is the BG505 DS-SOSIP variant, comprising two disulfides and an Ile-to-Pro mutation of Env from strain BG505. Here, we introduced additional mutations to further stabilize BG505 DS-SOSIP in the vaccine-preferred prefusion-closed conformation. In guinea pigs, our best mutant, DS-SOSIP.4mut, elicited a significantly higher ratio of autologous versus V3-directed neutralizing antibody responses than the SOSIP-stabilized form. We also observed an improvement in thermostability and a reduction in CD4 affinity. With improved antigenicity, stability, and immunogenicity, DS-SOSIP.4mut-stabilized trimers may have utility as HIV-1 immunogens or in other antigen-specific contexts, such as with B-cell probes.


Science immunology | 2017

Preferential induction of cross-group influenza A hemagglutinin stem–specific memory B cells after H7N9 immunization in humans

Sarah F. Andrews; M. Gordon Joyce; Michael Chambers; Rebecca A. Gillespie; Masaru Kanekiyo; Kwanyee Leung; Eun Sung Yang; Yaroslav Tsybovsky; Adam K. Wheatley; Michelle C. Crank; Jeffrey C. Boyington; Madhu Prabhakaran; Sandeep Narpala; Xuejun Chen; Robert T. Bailer; Grace L. Chen; Emily E. Coates; Peter D. Kwong; Richard A. Koup; John R. Mascola; Barney S. Graham; Julie E. Ledgerwood; Adrian B. McDermott

Broadly protective antibodies are preferentially induced upon vaccination with a group 2 immunogen. Stemming the tide of influenza A universal flu vaccine would prevent the need for yearly flu shots, but successful development has been hampered by the diversity and adaptability of the influenza virus. Andrews et al. compared B cell responses to the relatively conserved stem region of the influenza cell surface molecule hemagglutinin (HA) in humans vaccinated with either group 2 H7N9 or group 1 H5N1. They found that the stem-targeted memory B cells after H7N9 vaccination recognized both group 1 and group 2 influenza subtypes, whereas H5N1 vaccination induced responses primarily to group 1 subtypes. These data suggest that a group 2 stem immunogen, administered in the proper conditions, would have a higher likelihood of eliciting cross-group protection. Antigenic drift and shift of influenza strains underscore the need for broadly protective influenza vaccines. One strategy is to design immunogens that elicit B cell responses against conserved epitopes on the hemagglutinin (HA) stem. To better understand the elicitation of HA stem–targeted B cells to group 1 and group 2 influenza subtypes, we compared the memory B cell response to group 2 H7N9 and group 1 H5N1 vaccines in humans. Upon H7N9 vaccination, almost half of the HA stem–specific response recognized the group 1 and group 2 subtypes, whereas the response to H5N1 was largely group 1–specific. Immunoglobulin repertoire analysis of HA-specific B cells indicated that the H7N9 and H5N1 vaccines induced genetically similar cross-group HA stem–binding B cells, albeit at a much higher frequency upon H7N9 vaccination. These data suggest that a group 2–based stem immunogen could prove more effective than a group 1 immunogen at eliciting broad cross-group protection in humans.


PLOS ONE | 2016

Structure-Based Design of Head-Only Fusion Glycoprotein Immunogens for Respiratory Syncytial Virus

Jeffrey C. Boyington; M. Gordon Joyce; Mallika Sastry; Guillaume Stewart-Jones; Man Chen; Wing-Pui Kong; Joan O. Ngwuta; Paul V. Thomas; Yaroslav Tsybovsky; Yongping Yang; Baoshan Zhang; Lei Chen; Aliaksandr Druz; Ivelin S. Georgiev; Kiyoon Ko; Tongqing Zhou; John R. Mascola; Barney S. Graham; Peter D. Kwong

Respiratory syncytial virus (RSV) is a significant cause of severe respiratory illness worldwide, particularly in infants, young children, and the elderly. Although no licensed vaccine is currently available, an engineered version of the metastable RSV fusion (F) surface glycoprotein—stabilized in the pre-fusion (pre-F) conformation by “DS-Cav1” mutations—elicits high titer RSV-neutralizing responses. Moreover, pre-F-specific antibodies, often against the neutralization-sensitive antigenic site Ø in the membrane-distal head region of trimeric F glycoprotein, comprise a substantial portion of the human response to natural RSV infection. To focus the vaccine-elicited response to antigenic site Ø, we designed a series of RSV F immunogens that comprised the membrane-distal head of the F glycoprotein in its pre-F conformation. These “head-only” immunogens formed monomers, dimers, and trimers. Antigenic analysis revealed that a majority of the 70 engineered head-only immunogens displayed reactivity to site Ø-targeting antibodies, which was similar to that of the parent RSV F DS-Cav1 trimers, often with increased thermostability. We evaluated four of these head-only immunogens in detail, probing their recognition by antibodies, their physical stability, structure, and immunogenicity. When tested in naïve mice, a head-only trimer, half the size of the parent RSV F trimer, induced RSV titers, which were statistically comparable to those induced by DS-Cav1. When used to boost DS-Cav1-primed mice, two head-only RSV F immunogens, a dimer and a trimer, boosted RSV-neutralizing titers to levels that were comparable to those boosted by DS-Cav1, although with higher site Ø-directed responses. Our results provide proof-of-concept for the ability of the smaller head-only RSV F immunogens to focus the vaccine-elicited response to antigenic site Ø. Decent primary immunogenicity, enhanced physical stability, potential ease of manufacture, and potent immunogenicity upon boosting suggest these head-only RSV F immunogens, engineered to retain the pre-fusion conformation, may have advantages as candidate RSV vaccines.


Immunity | 2018

A Neutralizing Antibody Recognizing Primarily N-Linked Glycan Targets the Silent Face of the HIV Envelope

Tongqing Zhou; Anqi Zheng; Ulrich Baxa; Gwo-Yu Chuang; Ivelin S. Georgiev; Rui Kong; Sijy O’Dell; Syed Shahzad-ul-Hussan; Chen-Hsiang Shen; Yaroslav Tsybovsky; Robert T. Bailer; Syna K. Gift; Mark K. Louder; Krisha McKee; Reda Rawi; Catherine H. Stevenson; Guillaume Stewart-Jones; Justin D. Taft; Eric Waltari; Yongping Yang; Baoshan Zhang; Sachin S. Shivatare; Vidya S. Shivatare; Chang-Chun D. Lee; Chung-Yi Wu; Betty Benjamin; Robert W. Blakesley; Gerry Bouffard; Shelise Brooks; Holly Coleman

&NA; Virtually the entire surface of the HIV‐1‐envelope trimer is recognized by neutralizing antibodies, except for a highly glycosylated region at the center of the “silent face” on the gp120 subunit. From an HIV‐1‐infected donor, #74, we identified antibody VRC‐PG05, which neutralized 27% of HIV‐1 strains. The crystal structure of the antigen‐binding fragment of VRC‐PG05 in complex with gp120 revealed an epitope comprised primarily of N‐linked glycans from N262, N295, and N448 at the silent face center. Somatic hypermutation occurred preferentially at antibody residues that interacted with these glycans, suggesting somatic development of glycan recognition. Resistance to VRC‐PG05 in donor #74 involved shifting of glycan‐N448 to N446 or mutation of glycan‐proximal residue E293. HIV‐1 neutralization can thus be achieved at the silent face center by glycan‐recognizing antibody; along with other known epitopes, the VRC‐PG05 epitope completes coverage by neutralizing antibody of all major exposed regions of the prefusion closed trimer. Graphical Abstract Figure. No caption available. HighlightsIdentified and defined crystal structure of antibody VRC‐PG05 in complex with gp120VRC‐PG05 epitope is at the center of the glycosylated silent face of HIV‐1 gp120VRC‐PG05 utilizes both glycopeptide and glycan‐cluster mechanisms of recognitionVRC‐PG05 completes neutralizing antibody coverage of the prefusion‐closed Env trimer &NA; The center of the “silent face” on the HIV‐1 envelope is shielded by glycans and has been devoid of antibody recognition. Zhou et al. identify the antibody VRC‐PG05, which binds a glycan‐dominated epitope at the silent face center and completes antibody recognition of all major exposed regions of the envelope trimer.


npj Vaccines | 2017

Protection of calves by a prefusion-stabilized bovine RSV F vaccine

Baoshan Zhang; Lei Chen; Chiara Silacci; Michelle Thom; Jeffrey C. Boyington; Aliaksandr Druz; M. Gordon Joyce; Efrain Guzman; Wing Pui Kong; Yen-Ting Lai; Guillaume Stewart-Jones; Yaroslav Tsybovsky; Yongping Yang; Tongqing Zhou; Ulrich Baxa; John R. Mascola; Davide Corti; Antonio Lanzavecchia; Geraldine Taylor; Peter D. Kwong

Bovine respiratory syncytial virus, a major cause of respiratory disease in calves, is closely related to human RSV, a leading cause of respiratory disease in infants. Recently, promising human RSV-vaccine candidates have been engineered that stabilize the metastable fusion (F) glycoprotein in its prefusion state; however, the absence of a relevant animal model for human RSV has complicated assessment of these vaccine candidates. Here, we use a combination of structure-based design, antigenic characterization, and X-ray crystallography to translate human RSV F stabilization into the bovine context. A “DS2” version of bovine respiratory syncytial virus F with subunits covalently fused, fusion peptide removed, and pre-fusion conformation stabilized by cavity-filling mutations and intra- and inter-protomer disulfides was recognized by pre-fusion-specific antibodies, AM14, D25, and MPE8, and elicited bovine respiratory syncytial virus-neutralizing titers in calves >100-fold higher than those elicited by post-fusion F. When challenged with a heterologous bovine respiratory syncytial virus, virus was not detected in nasal secretions nor in respiratory tract samples of DS2-immunized calves; by contrast bovine respiratory syncytial virus was detected in all post-fusion- and placebo-immunized calves. Our results demonstrate proof-of-concept that DS2-stabilized RSV F immunogens can induce highly protective immunity from RSV in a native host with implications for the efficacy of prefusion-stabilized F vaccines in humans and for the prevention of bovine respiratory syncytial virus in calves.Respiratory disease: A bovine model for respiratory syncytial virus vaccinesResearchers have produced a vaccine that protects against bovine respiratory syncytial virus (bRSV) in calves, with implications for humans. An international team comprising Geraldine Taylor, The Pirbright Institute, UK, Davide Corti and Antonio Lanzavecchia, Institute for Research in Biomedicine, Switzerland, and Peter Kwong, Vaccine Research Center, NIAID, NIH, United States, and their teams constructed the subunit vaccine from an engineered bRSVfusion (F) glycoprotein that protected challenged calves by generating a highly protective immune response. This approach allays some of the dangers of whole-virus vaccines. The results warrant further investigation, as current bRSV vaccines have significant downsides.Moreover, as the engineered bRSV F glycoprotein is structurally and reactively similar to the prefusion-stabilized human RSV (hRSV) F glycoprotein, the findings highlight potential benefits of similar vaccines in humans, as no licensed hRSV vaccine is currently available.

Collaboration


Dive into the Yaroslav Tsybovsky's collaboration.

Top Co-Authors

Avatar

Aliaksandr Druz

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

John R. Mascola

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Ivelin S. Georgiev

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

Baoshan Zhang

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
Top Co-Authors

Avatar

Tongqing Zhou

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
Researchain Logo
Decentralizing Knowledge