Network


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

Hotspot


Dive into the research topics where Karen L. Saye-Francisco is active.

Publication


Featured researches published by Karen L. Saye-Francisco.


Proceedings of the National Academy of Sciences of the United States of America | 2014

Recombinant HIV envelope trimer selects for quaternary-dependent antibodies targeting the trimer apex

Devin Sok; Marit J. van Gils; Matthias Pauthner; Jean-Philippe Julien; Karen L. Saye-Francisco; Jessica Hsueh; Bryan Briney; Jeong Hyun Lee; Khoa Le; Peter S. Lee; Yuanzi Hua; Michael S. Seaman; John P. Moore; Andrew B. Ward; Ian A. Wilson; Rogier W. Sanders; Dennis R. Burton

Significance Despite the high antigenic diversity of the HIV envelope trimer (Env), broadly neutralizing antibodies (bnAbs) have identified conserved regions that serve as targets for vaccine design. One of these regions is located at the apex of Env and is expressed fully only in the context of the correctly folded trimer. This work describes the isolation of bnAbs that target this region using a recombinant native-like Env trimer as an affinity reagent to sort specific antibody-producing cells. Characterization of these antibodies reveals a highly diverse antibody response against the trimer apex and provides molecular information that will be useful in the design of immunogens to elicit bnAbs to this region of Env. Broadly neutralizing antibodies (bnAbs) targeting the trimer apex of HIV envelope are favored candidates for vaccine design and immunotherapy because of their great neutralization breadth and potency. However, methods of isolating bnAbs against this site have been limited by the quaternary nature of the epitope region. Here we report the use of a recombinant HIV envelope trimer, BG505 SOSIP.664 gp140, as an affinity reagent to isolate quaternary-dependent bnAbs from the peripheral blood mononuclear cells of a chronically infected donor. The newly isolated bnAbs, named “PGDM1400–1412,” show a wide range of neutralization breadth and potency. One of these variants, PGDM1400, is exceptionally broad and potent with cross-clade neutralization coverage of 83% at a median IC50 of 0.003 µg/mL. Overall, our results highlight the utility of BG505 SOSIP.664 gp140 as a tool for the isolation of quaternary-dependent antibodies and reveal a mosaic of antibody responses against the trimer apex within a clonal family.


Science Translational Medicine | 2014

Promiscuous Glycan Site Recognition by Antibodies to the High-Mannose Patch of gp120 Broadens Neutralization of HIV

Devin Sok; Katie J. Doores; Bryan Briney; Khoa Le; Karen L. Saye-Francisco; Alejandra Ramos; Daniel W. Kulp; Jean-Philippe Julien; Sergey Menis; Lalinda Wickramasinghe; Michael S. Seaman; William R. Schief; Ian A. Wilson; Pascal Poignard; Dennis R. Burton

HIV broadly neutralizing monoclonal antibodies targeting the high-mannose patch of Env can use alternate glycan sites for neutralization. Neutralizing Antibodies with a Sweet Tooth Sugar can be quite tempting—as anyone who’s seen a kid rip into birthday cake can attest. Yet, antibodies can also have a sweet tooth, targeting glycan modifications on the surface of proteins. Indeed, some antibodies that neutralize multiple HIV strains—broadly neutralizing monoclonal antibodies (bnmAbs)—target a high-mannose patch on the HIV protein Env. Although this high-mannose patch is centered around the glycan at position 332 (N332), it has remained unclear if the N332 glycan is absolutely required for neutralization and, if not, why not. Sok et al. found that these mannose patch–targeting antibodies can bind alternate glycans in the absence of N332, which helps to explain their ability to neutralize many strains of HIV. Specifically, some bnmAbs can bind to the N334 site when that replaces the N332 site and some can form more interactions with other glycans, particularly complex-type glycans on variable loops, if the N332 sugar is absent. These data also suggest that mannose patch–targeting bnmAbs can work in combination to neutralize a wider range of different strains than single bnmAbs. The promiscuity of glycan binding by these sugar-loving antibodies is important to consider for both vaccine and therapeutic antibody development. Broadly neutralizing monoclonal antibodies (bnmAbs) that target the high-mannose patch centered around the glycan at position 332 on HIV Env are promising vaccine leads and therapeutic candidates because they effectively protect against mucosal SHIV challenge and strongly suppress SHIV viremia in established infection in macaque models. However, these antibodies demonstrate varying degrees of dependency on the N332 glycan site, and the origins of their neutralization breadth are not always obvious. By measuring neutralization on an extended range of glycan site viral variants, we found that some bnmAbs can use alternate N-linked glycans in the absence of the N332 glycan site and therefore neutralize a substantial number of viruses lacking the site. Furthermore, many of the antibodies can neutralize viruses in which the N332 glycan site is shifted to the 334 position. Finally, we found that a combination of three antibody families that target the high-mannose patch can lead to 99% neutralization coverage of a large panel of viruses containing the N332/N334 glycan site and up to 66% coverage for viruses that lack the N332/N334 glycan site. The results indicate that a diverse response against the high-mannose patch may provide near-equivalent coverage as a combination of bnmAbs targeting multiple epitopes. Additionally, the ability of some bnmAbs to use other N-linked glycan sites can help counter neutralization escape mediated by shifting of glycosylation sites. Overall, this work highlights the importance of promiscuous glycan binding properties in bnmAbs to the high-mannose patch for optimal antiviral activity in either protective or therapeutic modalities.


Cell | 2016

Tailored Immunogens Direct Affinity Maturation toward HIV Neutralizing Antibodies.

Bryan Briney; Devin Sok; Joseph G. Jardine; Daniel W. Kulp; Patrick Skog; Sergey Menis; Ronald Jacak; Oleksandr Kalyuzhniy; Natalia de Val; Fabian Sesterhenn; Khoa Le; Alejandra Ramos; Meaghan Jones; Karen L. Saye-Francisco; Tanya R. Blane; Skye Spencer; Erik Georgeson; Xiaozhen Hu; Gabriel Ozorowski; Yumiko Adachi; Michael Kubitz; Anita Sarkar; Ian A. Wilson; Andrew B. Ward; David Nemazee; Dennis R. Burton; William R. Schief

Summary Induction of broadly neutralizing antibodies (bnAbs) is a primary goal of HIV vaccine development. VRC01-class bnAbs are important vaccine leads because their precursor B cells targeted by an engineered priming immunogen are relatively common among humans. This priming immunogen has demonstrated the ability to initiate a bnAb response in animal models, but recall and maturation toward bnAb development has not been shown. Here, we report the development of boosting immunogens designed to guide the genetic and functional maturation of previously primed VRC01-class precursors. Boosting a transgenic mouse model expressing germline VRC01 heavy chains produced broad neutralization of near-native isolates (N276A) and weak neutralization of fully native HIV. Functional and genetic characteristics indicate that the boosted mAbs are consistent with partially mature VRC01-class antibodies and place them on a maturation trajectory that leads toward mature VRC01-class bnAbs. The results show how reductionist sequential immunization can guide maturation of HIV bnAb responses.


PLOS Pathogens | 2016

Minimally Mutated HIV-1 Broadly Neutralizing Antibodies to Guide Reductionist Vaccine Design.

Joseph G. Jardine; Devin Sok; Jean-Philippe Julien; Bryan Briney; Anita Sarkar; Chi Hui Liang; Erin A. Scherer; Carole J. Henry Dunand; Yumiko Adachi; Devan Diwanji; Jessica Hsueh; Meaghan Jones; Oleksandr Kalyuzhniy; Michael Kubitz; Skye Spencer; Matthias Pauthner; Karen L. Saye-Francisco; Fabian Sesterhenn; Patrick C. Wilson; Denise M. Galloway; Robyn L. Stanfield; Ian A. Wilson; Dennis R. Burton; William R. Schief

An optimal HIV vaccine should induce broadly neutralizing antibodies (bnAbs) that neutralize diverse viral strains and subtypes. However, potent bnAbs develop in only a small fraction of HIV-infected individuals, all contain rare features such as extensive mutation, insertions, deletions, and/or long complementarity-determining regions, and some are polyreactive, casting doubt on whether bnAbs to HIV can be reliably induced by vaccination. We engineered two potent VRC01-class bnAbs that minimized rare features. According to a quantitative features frequency analysis, the set of features for one of these minimally mutated bnAbs compared favorably with all 68 HIV bnAbs analyzed and was similar to antibodies elicited by common vaccines. This same minimally mutated bnAb lacked polyreactivity in four different assays. We then divided the minimal mutations into spatial clusters and dissected the epitope components interacting with those clusters, by mutational and crystallographic analyses coupled with neutralization assays. Finally, by synthesizing available data, we developed a working-concept boosting strategy to select the mutation clusters in a logical order following a germline-targeting prime. We have thus developed potent HIV bnAbs that may be more tractable vaccine goals compared to existing bnAbs, and we have proposed a strategy to elicit them. This reductionist approach to vaccine design, guided by antibody and antigen structure, could be applied to design candidate vaccines for other HIV bnAbs or protective Abs against other pathogens.


Immunity | 2016

A Prominent Site of Antibody Vulnerability on HIV Envelope Incorporates a Motif Associated with CCR5 Binding and Its Camouflaging Glycans.

Devin Sok; Matthias Pauthner; Bryan Briney; Jeong Hyun Lee; Karen L. Saye-Francisco; Jessica Hsueh; Alejandra Ramos; Khoa Le; Meaghan Jones; Joseph G. Jardine; Raiza Bastidas; Anita Sarkar; Chi-Hui Liang; Sachin S. Shivatare; Chung-Yi Wu; William R. Schief; Chi-Huey Wong; Ian A. Wilson; Andrew B. Ward; Jiang Zhu; Pascal Poignard; Dennis R. Burton

The dense patch of high-mannose-type glycans surrounding the N332 glycan on the HIV envelope glycoprotein (Env) is targeted by multiple broadly neutralizing antibodies (bnAbs). This region is relatively conserved, implying functional importance, the origins of which are not well understood. Here we describe the isolation of new bnAbs targeting this region. Examination of these and previously described antibodies to Env revealed that four different bnAb families targeted the (324)GDIR(327) peptide stretch at the base of the gp120 V3 loop and its nearby glycans. We found that this peptide stretch constitutes part of the CCR5 co-receptor binding site, with the high-mannose patch glycans serving to camouflage it from most antibodies. GDIR-glycan bnAbs, in contrast, bound both (324)GDIR(327) peptide residues and high-mannose patch glycans, which enabled broad reactivity against diverse HIV isolates. Thus, as for the CD4 binding site, bnAb effectiveness relies on circumventing the defenses of a critical functional region on Env.


Nature microbiology | 2017

An HIV-1 antibody from an elite neutralizer implicates the fusion peptide as a site of vulnerability

Marit J. van Gils; Tom L. G. M. van den Kerkhof; Gabriel Ozorowski; Christopher A. Cottrell; Devin Sok; Matthias Pauthner; Jesper Pallesen; Natalia de Val; Anila Yasmeen; Steven W. de Taeye; Anna Schorcht; Stephanie Gumbs; Inez Johanna; Karen L. Saye-Francisco; Chi-Hui Liang; Elise Landais; Xiaoyan Nie; Laura K. Pritchard; Max Crispin; Garnett Kelsoe; Ian A. Wilson; Hanneke Schuitemaker; Per Johan Klasse; John P. Moore; Dennis R. Burton; Andrew B. Ward; Rogier W. Sanders

The induction by vaccination of broadly neutralizing antibodies (bNAbs) capable of neutralizing various HIV-1 viral strains is challenging, but understanding how a subset of HIV-infected individuals develops bNAbs may guide immunization strategies. Here, we describe the isolation and characterization of the bNAb ACS202 from an elite neutralizer that recognizes a new, trimer-specific and cleavage-dependent epitope at the gp120–gp41 interface of the envelope glycoprotein (Env), involving the glycan N88 and the gp41 fusion peptide. In addition, an Env trimer, AMC011 SOSIP.v4.2, based on early virus isolates from the same elite neutralizer, was constructed, and its structure by cryo-electron microscopy at 6.2 Å resolution reveals a closed, pre-fusion conformation similar to that of the BG505 SOSIP.664 trimer. The availability of a native-like Env trimer and a bNAb from the same elite neutralizer provides the opportunity to design vaccination strategies aimed at generating similar bNAbs against a key functional site on HIV-1.


Nature | 2017

Rapid elicitation of broadly neutralizing antibodies to HIV by immunization in cows

Devin Sok; Khoa Le; Melissa Vadnais; Karen L. Saye-Francisco; Joseph G. Jardine; Jonathan L. Torres; Zachary T. Berndsen; Leopold Kong; Robyn L. Stanfield; Jennifer Ruiz; Alejandra Ramos; Chi-Hui Liang; Patricia L. Chen; Michael F. Criscitiello; Waithaka Mwangi; Ian A. Wilson; Andrew B. Ward; Vaughn V. Smider; Dennis R. Burton

No immunogen to date has reliably elicited broadly neutralizing antibodies to HIV in humans or animal models. Advances in the design of immunogens that antigenically mimic the HIV envelope glycoprotein (Env), such as the soluble cleaved trimer BG505 SOSIP, have improved the elicitation of potent isolate-specific antibody responses in rabbits and macaques, but so far failed to induce broadly neutralizing antibodies. One possible reason for this failure is that the relevant antibody repertoires are poorly suited to target the conserved epitope regions on Env, which are somewhat occluded relative to the exposed variable epitopes. Here, to test this hypothesis, we immunized four cows with BG505 SOSIP. The antibody repertoire of cows contains long third heavy chain complementary determining regions (HCDR3) with an ultralong subset that can reach more than 70 amino acids in length. Remarkably, BG505 SOSIP immunization resulted in rapid elicitation of broad and potent serum antibody responses in all four cows. Longitudinal serum analysis for one cow showed the development of neutralization breadth (20%, n = 117 cross-clade isolates) in 42 days and 96% breadth (n = 117) at 381 days. A monoclonal antibody isolated from this cow harboured an ultralong HCDR3 of 60 amino acids and neutralized 72% of cross-clade isolates (n = 117) with a potent median IC50 of 0.028 μg ml−1. Breadth was elicited with a single trimer immunogen and did not require additional envelope diversity. Immunization of cows may provide an avenue to rapidly generate antibody prophylactics and therapeutics to address disease agents that have evolved to avoid human antibody responses.


PLOS Pathogens | 2017

Lipid interactions and angle of approach to the HIV-1 viral membrane of broadly neutralizing antibody 10E8: Insights for vaccine and therapeutic design.

Adriana Irimia; Andreia M. Serra; Anita Sarkar; Ronald Jacak; Oleksandr Kalyuzhniy; Devin Sok; Karen L. Saye-Francisco; Torben Schiffner; Ryan Tingle; Michael Kubitz; Yumiko Adachi; Robyn L. Stanfield; Marc C. Deller; Dennis R. Burton; William R. Schief; Ian A. Wilson

Among broadly neutralizing antibodies to HIV, 10E8 exhibits greater neutralizing breadth than most. Consequently, this antibody is the focus of prophylactic/therapeutic development. The 10E8 epitope has been identified as the conserved membrane proximal external region (MPER) of gp41 subunit of the envelope (Env) viral glycoprotein and is a major vaccine target. However, the MPER is proximal to the viral membrane and may be laterally inserted into the membrane in the Env prefusion form. Nevertheless, 10E8 has not been reported to have significant lipid-binding reactivity. Here we report x-ray structures of lipid complexes with 10E8 and a scaffolded MPER construct and mutagenesis studies that provide evidence that the 10E8 epitope is composed of both MPER and lipid. 10E8 engages lipids through a specific lipid head group interaction site and a basic and polar surface on the light chain. In the model that we constructed, the MPER would then be essentially perpendicular to the virion membrane during 10E8 neutralization of HIV-1. As the viral membrane likely also plays a role in selecting for the germline antibody as well as size and residue composition of MPER antibody complementarity determining regions, the identification of lipid interaction sites and the MPER orientation with regard to the viral membrane surface during 10E8 engagement can be of great utility for immunogen and therapeutic design.


Journal of Virology | 2013

Antibody Conjugation Approach Enhances Breadth and Potency of Neutralization of Anti-HIV-1 Antibodies and CD4-IgG

Julia Gavrilyuk; Hitoshi Ban; Hisatoshi Uehara; Shannon J. Sirk; Karen L. Saye-Francisco; Angelica Cuevas; Elise Zablowsky; Avinash Oza; Michael S. Seaman; Dennis R. Burton; Carlos F. Barbas

ABSTRACT Broadly neutralizing antibodies PG9 and PG16 effectively neutralize 70 to 80% of circulating HIV-1 isolates. In this study, the neutralization abilities of PG9 and PG16 were further enhanced by bioconjugation with aplaviroc, a small-molecule inhibitor of virus entry into host cells. A novel air-stable diazonium hexafluorophosphate reagent that allows for rapid, tyrosine-selective functionalization of proteins and antibodies under mild conditions was used to prepare a series of aplaviroc-conjugated antibodies, including b12, 2G12, PG9, PG16, and CD4-IgG. The conjugated antibodies blocked HIV-1 entry through two mechanisms: by binding to the virus itself and by blocking the CCR5 receptor on host cells. Chemical modification did not significantly alter the potency of the parent antibodies against nonresistant HIV-1 strains. Conjugation did not alter the pharmacokinetics of a model IgG in blood. The PG9-aplaviroc conjugate was tested against a panel of 117 HIV-1 strains and was found to neutralize 100% of the viruses. PG9-aplaviroc conjugate IC50s were lower than those of PG9 in neutralization studies of 36 of the 117 HIV-1 strains. These results support this new approach to bispecific antibodies and offer a potential new strategy for combining HIV-1 therapies.


Methods | 2014

Simplifying the synthesis of SIgA: combination of dIgA and rhSC using affinity chromatography.

Brian Moldt; Karen L. Saye-Francisco; Niccole Schultz; Dennis R. Burton; Ann J. Hessell

The mucosal epithelia together with adaptive immune responses, such as local production and secretion of dimeric and polymeric immunoglobulin A (IgA), are a crucial part of the first line of defense against invading pathogens. IgA is primarily secreted as SIgA and plays multiple roles in mucosal defense. The study of SIgA-mediated protection is an important area of research in mucosal immunity but an easy, fast and reproducible method to generate pathogen-specific SIgA in vitro has not been available. We report here a new method to produce SIgA by co-purification of dimeric IgA, containing J chain, and recombinant human SC expressed in CHO cells. We previously reported the generation, production and characterization of the human recombinant monoclonal antibody IgA2 b12. This antibody, derived from the variable regions of the neutralizing anti-HIV-1 mAb IgG1 b12, blocked viral attachment and uptake by epithelial cells in vitro. We used a cloned CHO cell line that expresses monomeric, dimeric and polymeric species of IgA2 b12 for large-scale production of dIgA2 b12. Subsequently, we generated a CHO cell line to express recombinant human secretory component (rhSC). Here, we combined dIgA2 b12 and CHO-expressed rhSC via column chromatography to produce SIgA2 b12 that remains fully intact upon elution with 0.1M citric acid, pH 3.0. We have performed biochemical analysis of the synthesized SIgA to confirm the species is of the expected size and retains the functional properties previously described for IgA2 b12. We show that SIgA2 b12 binds to the HIV-1 gp120 glycoprotein with similar apparent affinity to that of monomeric and dimeric forms of IgA2 b12 and neutralizes HIV-1 isolates with similar potency. An average yield of 6 mg of SIgA2 b12 was achieved from the combination of 20mg of purified dIgA2 b12 and 2L of rhSC-containing CHO cell supernatant. We conclude that synthesized production of stable SIgA can be generated by co-purification. This process introduces a simplified means of generating a variety of pathogen-specific SIgA antibodies for research and clinical applications.

Collaboration


Dive into the Karen L. Saye-Francisco's collaboration.

Top Co-Authors

Avatar

Dennis R. Burton

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

Andrew B. Ward

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar

William R. Schief

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar

Anita Sarkar

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar

Bryan Briney

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar

Joseph G. Jardine

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar

Khoa Le

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar

Matthias Pauthner

Scripps Research Institute

View shared research outputs
Researchain Logo
Decentralizing Knowledge