Network


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

Hotspot


Dive into the research topics where Lisa R. King is active.

Publication


Featured researches published by Lisa R. King.


PLOS Medicine | 2009

Antigenic Fingerprinting of H5N1 Avian Influenza Using Convalescent Sera and Monoclonal Antibodies Reveals Potential Vaccine and Diagnostic Targets

Surender Khurana; Amorsolo L. Suguitan; Yonaira Rivera; Cameron P. Simmons; Antonio Lanzavecchia; Federica Sallusto; Jody Manischewitz; Lisa R. King; Kanta Subbarao; Hana Golding

Using whole-genome-fragment phage display libraries, Hana Golding and colleagues identify the viral epitopes recognized by serum antibodies in humans who have recovered from infection with H5N1 avian influenza.


Nature Medicine | 2005

Smallpox vaccine–induced antibodies are necessary and sufficient for protection against monkeypox virus

Yvette Edghill-Smith; Hana Golding; Jody Manischewitz; Lisa R. King; Dorothy E. Scott; Mike Bray; Aysegul Nalca; Jay W. Hooper; Chris A Whitehouse; Joern E. Schmitz; Keith A. Reimann; Genoveffa Franchini

Vaccination with live vaccinia virus affords long-lasting protection against variola virus, the agent of smallpox. Its mode of protection in humans, however, has not been clearly defined. Here we report that vaccinia-specific B-cell responses are essential for protection of macaques from monkeypox virus, a variola virus ortholog. Antibody-mediated depletion of B cells, but not CD4+ or CD8+ T cells, abrogated vaccine-induced protection from a lethal intravenous challenge with monkeypox virus. In addition, passive transfer of human vaccinia-neutralizing antibodies protected nonimmunized macaques from severe disease. Thus, vaccines able to induce long-lasting protective antibody responses may constitute realistic alternatives to the currently available smallpox vaccine (Dryvax).


Science Translational Medicine | 2010

Vaccines with MF59 Adjuvant Expand the Antibody Repertoire to Target Protective Sites of Pandemic Avian H5N1 Influenza Virus

Surender Khurana; Wanida Chearwae; Flora Castellino; Jody Manischewitz; Lisa R. King; Agnieszka Honorkiewicz; Michael T. Rock; Kathryn M. Edwards; Giuseppe Del Giudice; Rino Rappuoli; Hana Golding

An oil-based adjuvant improves the efficacy of an H5N1 vaccine by inducing antibodies against additional sites on influenza surface proteins. Revving Up a Flu Vaccine By preventing incalculable illnesses, vaccines are one of medicine’s great triumphs. Part of the credit goes to the adjuvants, substances included in vaccine preparations that boost the immune response but have no effect on their own. These agents—as various as aluminum salts and lipids—are thought to activate the innate immune system, the generalized protective response most animals show to pathogens. But the precise mechanisms by which these agents augment the immune response continue to elude scientists. Khurana and colleagues have now closely compared the immune response to an avian influenza vaccine without adjuvant to that induced with an oil-in-water adjuvant. Mixing the adjuvant with the vaccine induced antibodies that recognized a wider variety of flu antigens, including some known to inactivate the virus. The adjuvant examined by these authors was MF59—an emulsion of squalene, a natural hydrocarbon, and several fatty acids—which is used to boost influenza virus vaccines marketed in Europe. Flu vaccines that include MF59 engender more neutralizing antibodies than those that do not. To better understand the characteristics of these additional antibodies to flu proteins, the authors analyzed sera from subjects in two clinical trials of vaccines against the avian flu H5N1, with and without MF59. As expected, MF59 increased the amount of antibody against H5N1 flu, but the diversity of the new antibodies was also amplified. The authors looked specifically for antibodies directed at regions of the hemagglutinin protein and at neuraminidase, through which the virus binds to and infects its host’s cells. [The H5N1 name of the influenza strain refers to the exact type of hemagglutinin (H) and neuraminidase (N) proteins carried by the virus.] MF59 caused production of more antibodies directed against the hemagglutinin region that binds host cells, rather than the region that anchors the protein in the membrane, and many more regions within these areas were targeted. Only vaccines with MF59 generated antibodies against long sequences and against the native conformation of the host cell binding region. These particular antibodies are important because they are the ones that successfully block infection—the ultimate goal of any vaccine. They were also able to bind to and neutralize hemagglutinin from other strains of H5N1 flu—from Indonesia and China—much more effectively than were antibodies induced by vaccines without adjuvant; this finding showed that MF59-containing vaccines also have the desirable feature of extending protection beyond the particular flu strain used for vaccination. This study does not reveal exactly how the wider immune repertoire induced by MF59 is generated. But it does begin to define precisely the nature of the adjuvant-induced antibodies to influenza and to explain why they are more effective than those made in response to vaccines without adjuvant. MF59 is the adjuvant used in some vaccines against the current pandemic H1N1 flu, and the conclusions from this study are likely to apply to those vaccines as well. Vaccines against influenza viruses with pandemic potential, including H5N1, are under development. Because of a lack of preexisting immunity to these viruses, adjuvants (immune potentiators or enhancers) are needed to improve immune responses, to conserve scarce vaccine, and for cross-protection against strains that have drifted evolutionarily from the original. Aluminum-based adjuvants do not improve vaccine immunogenicity for influenza subunit vaccines, whereas oil-in-water adjuvants are effective, especially with H5N1-inactivated vaccines. We used whole-genome-fragment phage display libraries followed by surface plasmon resonance (SPR) technologies to elucidate the effect of different adjuvants on the antibody repertoire against H5N1 vaccine in humans. The oil-in-water adjuvant MF59 induced epitope spreading from HA2 to HA1 in hemagglutinin (HA) and neuraminidase relative to unadjuvanted or aluminum-adjuvanted vaccines. Moreover, we observed an increase by a factor of 20 in the frequency of HA1-to-HA2–specific phage clones in sera after MF59-adjuvanted vaccine administration and a factor of 2 to 3 increase in the avidity of antibodies binding to properly folded HA1(28–319), as measured by SPR. The adjuvant-dependent increase in binding to conformational HA1 epitopes correlated with broadening of cross-clade neutralization and predicted improved in vivo protection. Thus, MF59 adjuvant improves the immune response to a H5N1 vaccine by inducing qualitative and quantitative expansion of the antibody repertoires with protective potential.


Journal of Virology | 2002

Dissection of Human Immunodeficiency Virus Type 1 Entry with Neutralizing Antibodies to gp41 Fusion Intermediates

Hana Golding; Marina Zaitseva; Eve de Rosny; Lisa R. King; Jody Manischewitz; Igor A. Sidorov; Miroslaw K. Gorny; Susan Zolla-Pazner; Dimiter S. Dimitrov; Carol D. Weiss

ABSTRACT Human immunodeficiency virus type 1 (HIV-1) entry requires conformational changes in the transmembrane subunit (gp41) of the envelope glycoprotein (Env) involving transient fusion intermediates that contain exposed coiled-coil (prehairpin) and six-helix bundle structures. We investigated the HIV-1 entry mechanism and the potential of antibodies targeting fusion intermediates to block Env-mediated membrane fusion. Suboptimal temperature (31.5°C) was used to prolong fusion intermediates as monitored by confocal microscopy. After transfer to 37°C, these fusion intermediates progressed to syncytium formation with enhanced kinetics compared with effector-target (E/T) cell mixtures that were incubated only at 37°C. gp41 peptides DP-178, DP-107, and IQN17 blocked fusion more efficiently (5- to 10-fold-lower 50% inhibitory dose values) when added to E/T cells at the suboptimal temperature prior to transfer to 37°C. Rabbit antibodies against peptides modeling the N-heptad repeat or the six-helix bundle of gp41 blocked fusion and viral infection at 37°C only if preincubated with E/T cells at the suboptimal temperature. Similar fusion inhibition was observed with human six-helix bundle-specific monoclonal antibodies. Our data demonstrate that antibodies targeting gp41 fusion intermediates are able to bind to gp41 and arrest fusion. They also indicate that six-helix bundles can form prior to fusion and that the lag time before fusion occurs may include the time needed to accumulate preformed six-helix bundles at the fusion site.


Science Translational Medicine | 2013

Vaccine-Induced Anti-HA2 Antibodies Promote Virus Fusion and Enhance Influenza Virus Respiratory Disease

Surender Khurana; Crystal L. Loving; Jody Manischewitz; Lisa R. King; Phillip C. Gauger; Jamie N. Henningson; Amy L. Vincent; Hana Golding

Heterologous influenza A vaccination–induced antibodies correlated with vaccine-associated enhanced respiratory disease. The (Mis)match Game Even the most beneficial things—like vaccines—sometimes have a downside. Learning what causes the downside is critical for avoiding it. In the case of viral vaccines, there have been some reports of rare vaccine-induced disease enhancement—for example, vaccine-associated enhanced respiratory disease (VAERD) for influenza. Khurana et al. now report that mismatched strains of the same subtype of influenza may lead to VAERD in pigs. The authors vaccinated pigs with whole inactivated H1N2 influenza virus. These pigs had enhanced pneumonia and disease after infection with another strain—pH1N1. Looking more closely, the authors found that the immune sera from the H1N2-vaccinated pigs contained high titers of cross-reactive hemagglutinin antibodies. These antibodies actually enhanced pH1N1 infection in cell culture by promoting virus membrane fusion activity, and this enhanced fusion correlated with lung pathology. This mechanism of VAERD should be considered when devising strategies to devise a universal flu vaccine. Vaccine-induced disease enhancement has been described in connection with several viral vaccines in animal models and in humans. We investigated a swine model to evaluate mismatched influenza vaccine-associated enhanced respiratory disease (VAERD) after pH1N1 infection. Vaccinating pigs with whole inactivated H1N2 (human-like) virus vaccine (WIV-H1N2) resulted in enhanced pneumonia and disease after pH1N1 infection. WIV-H1N2 immune sera contained high titers of cross-reactive anti-pH1N1 hemagglutinin (HA) antibodies that bound exclusively to the HA2 domain but not to the HA1 globular head. No hemagglutination inhibition titers against pH1N1 (challenge virus) were measured. Epitope mapping using phage display library identified the immunodominant epitope recognized by WIV-H1N2 immune sera as amino acids 32 to 77 of pH1N1-HA2 domain, close to the fusion peptide. These cross-reactive anti-HA2 antibodies enhanced pH1N1 infection of Madin-Darby canine kidney cells by promoting virus membrane fusion activity. The enhanced fusion activity correlated with lung pathology in pigs. This study suggests a role for fusion-enhancing anti-HA2 antibodies in VAERD, in the absence of receptor-blocking virus-neutralizing antibodies. These findings should be considered during the evaluation of universal influenza vaccines designed to elicit HA2 stem-targeting antibodies.


Journal of Virology | 2009

Electroporation of Synthetic DNA Antigens Offers Protection in Nonhuman Primates Challenged with Highly Pathogenic Avian Influenza Virus

Dominick J. Laddy; Jian Yan; Amir S. Khan; Hanne Andersen; Amanda Cohn; Jack Greenhouse; Mark G. Lewis; Jody Manischewitz; Lisa R. King; Hana Golding; Ruxandra Draghia-Akli; David B. Weiner

ABSTRACT Avian influenza highlights the need for novel vaccination techniques that would allow for the rapid design and production of safe and effective vaccines. An ideal platform would be capable of inducing both protective antibodies and potent cellular immune responses. These potential advantages of DNA vaccines remain unrealized due to a lack of efficacy in large animal studies and in human trials. Questions remain regarding the potential utility of cellular immune responses against influenza virus in primates. In this study, by construct optimization and in vivo electroporation of synthetic DNA-encoded antigens, we observed the induction of cross-reactive cellular and humoral immune responses individually capable of providing protection from influenza virus infection in the rhesus macaque. These studies advance the DNA vaccine field and provide a novel, more tolerable vaccine with broad immunogenicity to avian influenza virus. This approach appears important for further investigation, including studies with humans.


Journal of Immunology | 2006

Subunit Recombinant Vaccine Protects against Monkeypox

Jean-Michel Heraud; Yvette Edghill-Smith; Victor I. Ayala; Irene Kalisz; Janie Parrino; V. S. Kalyanaraman; Jody Manischewitz; Lisa R. King; Anna Hryniewicz; Christopher J. Trindade; Meredith Hassett; Wen-Po Tsai; David Venzon; Aysegul Nalca; Monica Vaccari; Peter Silvera; Mike Bray; Barney S. Graham; Hana Golding; Jay W. Hooper; Genoveffa Franchini

The smallpox vaccine Dryvax, a live vaccinia virus (VACV), protects against smallpox and monkeypox, but is contraindicated in immunocompromised individuals. Because Abs to VACV mediate protection, a live virus vaccine could be substituted by a safe subunit protein-based vaccine able to induce a protective Ab response. We immunized rhesus macaques with plasmid DNA encoding the monkeypox orthologs of the VACV L1R, A27L, A33R, and B5R proteins by the intradermal and i.m. routes, either alone or in combination with the equivalent recombinant proteins produced in Escherichia coli. Animals that received only DNA failed to produce high titer Abs, developed innumerable skin lesions after challenge, and died in a manner similar to placebo controls. By contrast, the animals vaccinated with proteins developed moderate to severe disease (20–155 skin lesions) but survived. Importantly, those immunized with DNA and boosted with proteins had mild disease with 15 or fewer lesions that resolved within days. DNA/protein immunization elicited Th responses and binding Ab titers to all four proteins that correlated negatively with the total lesion number. The sera of the immunized macaques recognized a limited number of linear B cell epitopes that are highly conserved among orthopoxviruses. Their identification may guide future efforts to develop simpler, safer, and more effective vaccines for monkeypox and smallpox.


Journal of Virology | 2011

H5N1 Virus-Like Particle Vaccine Elicits Cross-Reactive Neutralizing Antibodies That Preferentially Bind to the Oligomeric Form of Influenza Virus Hemagglutinin in Humans

Surender Khurana; Jian Wu; Nitin Verma; Swati Verma; Ramadevi Raghunandan; Jody Manischewitz; Lisa R. King; Eloi Kpamegan; Steven Pincus; Gale Smith; Gregory M. Glenn; Hana Golding

ABSTRACT Transmission of pathogenic avian influenza viruses (AIV) from wild birds to domestic poultry and humans is continuing in multiple countries around the world. In preparation for a potential AIV pandemic, multiple vaccine candidates are under development. In the case of H5N1 AIV, a clear shift in transmission from clade 1 to clade 2 viruses occurred in recent years. The virus-like particle (VLP) represents an economical approach to pandemic vaccine development. In the current study, we evaluated the humoral immune response in humans vaccinated with H5N1 A/Indonesia/05/2005 (clade 2.1) VLP vaccine manufactured in Sf9 insect cells. The VLPs were comprised of the influenza virus hemagglutinin (HA), neuraminidase (NA), and matrix 1 (M1) proteins. In an FDA-approved phase I/II human clinical study, two doses of H5N1 VLPs at 15, 45, or 90 μg HA/dose resulted in seroconversion and production of functional antibodies. Moreover, cross-reactivity against other clade 2 subtypes was demonstrated using virus neutralization assays. H5N1 whole-genome fragment phage display libraries (GFPDL) were used to elucidate the antibody epitope repertoire in postvaccination human sera. Diverse epitopes in HA1/HA2 and NA were recognized by postvaccination sera from the two high-dose groups, including large segments spanning the HA1 receptor binding domain. Importantly, the vaccine elicited sera that preferentially bound to an oligomeric form of recombinant HA1 compared with monomeric HA1. The oligomeric/monomeric HA1 binding ratios of the sera correlated with the virus neutralizing titers. Additionally, the two high-dose VLP vaccine groups generated NA-inhibiting antibodies that were associated with binding to a C-terminal epitope close to the sialic acid binding site. These findings represent the first report describing the quality of the antibody responses in humans following AIV VLP immunization and support further development of such vaccines against emerging influenza virus strains.


PLOS ONE | 2010

Properly Folded Bacterially Expressed H1N1 Hemagglutinin Globular Head and Ectodomain Vaccines Protect Ferrets against H1N1 Pandemic Influenza Virus

Surender Khurana; Swati Verma; Nitin Verma; Corey J. Crevar; Donald M. Carter; Jody Manischewitz; Lisa R. King; Ted M. Ross; Hana Golding

Background In the face of impending influenza pandemic, a rapid vaccine production and mass vaccination is the most effective approach to prevent the large scale mortality and morbidity that was associated with the 1918 “Spanish Flu”. The traditional process of influenza vaccine production in eggs is time consuming and may not meet the demands of rapid global vaccination required to curtail influenza pandemic. Methodology/Principal Findings Recombinant technology can be used to express the hemagglutinin (HA) of the emerging new influenza strain in a variety of systems including mammalian, insect, and bacterial cells. In this study, two forms of HA proteins derived from the currently circulating novel H1N1 A/California/07/2009 virus, HA1 (1–330) and HA (1–480), were expressed and purified from E. coli under controlled redox refolding conditions that favoured proper protein folding. However, only the recombinant HA1 (1–330) protein formed oligomers, including functional trimers that bound receptor and caused agglutination of human red blood cells. These proteins were used to vaccinate ferrets prior to challenge with the A/California/07/2009 virus. Both proteins induced neutralizing antibodies, and reduced viral loads in nasal washes. However, the HA1 (1–330) protein that had higher content of multimeric forms provided better protection from fever and weight loss at a lower vaccine dose compared with HA (1–480). Protein yield for the HA1 (1–330) ranged around 40 mg/Liter, while the HA (1–480) yield was 0.4–0.8 mg/Liter. Conclusions/Significance This is the first study that describes production in bacterial system of properly folded functional globular HA1 domain trimers, lacking the HA2 transmembrane protein, that elicit potent neutralizing antibody responses following vaccination and protect ferrets from in vivo challenge. The combination of bacterial expression system with established quality control methods could provide a mechanism for rapid large scale production of influenza vaccines in the face of influenza pandemic threat.


Journal of Virology | 2001

Immunoglobulin G3 from Polyclonal Human Immunodeficiency Virus (HIV) Immune Globulin Is More Potent than Other Subclasses in Neutralizing HIV Type 1

Orit Scharf; Hana Golding; Lisa R. King; Nancy Eller; Doug Frazier; Basil Golding; Dorothy E. Scott

ABSTRACT Passive antibody prophylaxis against human immunodeficiency virus type 1 (HIV-1) has been accomplished in primates, suggesting that this strategy may prove useful in humans. While antibody specificity is crucial for neutralization, other antibody characteristics, such as subclass, have not been explored. Our objective was to compare the efficiencies of immunoglobulin G (IgG) subclasses from polyclonal human HIV immune globulin (HIVIG) in the neutralization of HIV-1 strains differing in coreceptor tropism. IgG1, IgG2, and IgG3 were enriched from HIVIG by using protein A-Sepharose. All three subclasses bound major HIV-1 proteins, as shown by Western blot assay and enzyme-linked immunosorbent assay. In HIV-1 fusion assays using X4, R5, or X4R5 envelope-expressing effector cells, IgG3 more efficiently blocked fusion. In neutralization assays with cell-free viruses using X4 (LAI, IIIB), R5 (BaL), and X4R5 (DH123), a similar hierarchy of neutralization was found: IgG3 > IgG1 > IgG2. IgG3 has a longer, more flexible hinge region than the other subclasses. To test whether this is important, IgG1 and IgG3 were digested with pepsin to generate F(ab′)2 fragments or with papain to generate Fab fragments. IgG3 F(ab′)2 fragments were still more efficient in neutralization than F(ab′)2 of IgG1. However, Fab fragments of IgG3 and IgG1 demonstrated equivalent neutralization capacities and the IgG3 advantage was lost. These results suggest that the IgG3 hinge region confers enhanced HIV-neutralizing ability. Enrichment and stabilization of IgG3 may therefore lead to improved HIVIG preparations. The results of this study have implications for the improvement of passive immunization with polyclonal or monoclonal antibodies and suggest that HIV-1 vaccines which induce high-titer IgG3 responses could be advantageous.

Collaboration


Dive into the Lisa R. King's collaboration.

Top Co-Authors

Avatar

Hana Golding

Center for Biologics Evaluation and Research

View shared research outputs
Top Co-Authors

Avatar

Jody Manischewitz

Food and Drug Administration

View shared research outputs
Top Co-Authors

Avatar

Surender Khurana

Center for Biologics Evaluation and Research

View shared research outputs
Top Co-Authors

Avatar

Marina Zaitseva

Center for Biologics Evaluation and Research

View shared research outputs
Top Co-Authors

Avatar

Clement A. Meseda

Center for Biologics Evaluation and Research

View shared research outputs
Top Co-Authors

Avatar

Jerry P. Weir

Center for Biologics Evaluation and Research

View shared research outputs
Top Co-Authors

Avatar

John Scott

Center for Biologics Evaluation and Research

View shared research outputs
Top Co-Authors

Avatar

Michael Merchlinsky

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Swati Verma

Center for Biologics Evaluation and Research

View shared research outputs
Top Co-Authors

Avatar

Elizabeth M. Coyle

Center for Biologics Evaluation and Research

View shared research outputs
Researchain Logo
Decentralizing Knowledge