Network


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

Hotspot


Dive into the research topics where Paul E. Leon is active.

Publication


Featured researches published by Paul E. Leon.


Journal of Clinical Investigation | 2015

Preexisting human antibodies neutralize recently emerged H7N9 influenza strains

Carole J. Henry Dunand; Paul E. Leon; Kaval Kaur; Gene S. Tan; Nai-Ying Zheng; Sarah F. Andrews; Min Huang; Xinyan Qu; Yunping Huang; Marlene Salgado-Ferrer; Irvin Y. Ho; William Taylor; Rong Hai; Jens Wrammert; Rafi Ahmed; Adolfo García-Sastre; Peter Palese; Florian Krammer; Patrick C. Wilson

The emergence and seasonal persistence of pathogenic H7N9 influenza viruses in China have raised concerns about the pandemic potential of this strain, which, if realized, would have a substantial effect on global health and economies. H7N9 viruses are able to bind to human sialic acid receptors and are also able to develop resistance to neuraminidase inhibitors without a loss in fitness. It is not clear whether prior exposure to circulating human influenza viruses or influenza vaccination confers immunity to H7N9 strains. Here, we demonstrate that 3 of 83 H3 HA-reactive monoclonal antibodies generated by individuals that had previously undergone influenza A virus vaccination were able to neutralize H7N9 viruses and protect mice against homologous challenge. The H7N9-neutralizing antibodies bound to the HA stalk domain but exhibited a difference in their breadth of reactivity to different H7 influenza subtypes. Mapping viral escape mutations suggested that these antibodies bind at least two different epitopes on the stalk region. Together, these results indicate that these broadly neutralizing antibodies may contribute to the development of therapies against H7N9 strains and may also be effective against pathogenic H7 strains that emerge in the future.


Journal of Virology | 2014

Characterization of a broadly neutralizing monoclonal antibody that targets the fusion domain of group 2 influenza A virus hemagglutinin

Gene S. Tan; Peter S. Lee; Ryan M. B. Hoffman; Beryl Mazel-Sanchez; Florian Krammer; Paul E. Leon; Andrew B. Ward; Ian A. Wilson; Peter Palese

ABSTRACT Due to continuous changes to its antigenic regions, influenza viruses can evade immune detection and cause a significant amount of morbidity and mortality around the world. Influenza vaccinations can protect against disease but must be annually reformulated to match the current circulating strains. In the development of a broad-spectrum influenza vaccine, the elucidation of conserved epitopes is paramount. To this end, we designed an immunization strategy in mice to boost the humoral response against conserved regions of the hemagglutinin (HA) glycoprotein. Of note, generation and identification of broadly neutralizing antibodies that target group 2 HAs are rare and thus far have yielded only a few monoclonal antibodies (MAbs). Here, we demonstrate that mouse MAb 9H10 has broad and potent in vitro neutralizing activity against H3 and H10 group 2 influenza A subtypes. In the mouse model, MAb 9H10 protects mice against two divergent mouse-adapted H3N2 strains, in both pre- and postexposure administration regimens. In vitro and cell-free assays suggest that MAb 9H10 inhibits viral replication by blocking HA-dependent fusion of the viral and endosomal membranes early in the replication cycle and by disrupting viral particle egress in the late stage of infection. Interestingly, electron microscopy reconstructions of MAb 9H10 bound to the HA reveal that it binds a similar binding footprint to MAbs CR8020 and CR8043. IMPORTANCE The influenza hemagglutinin is the major antigenic target of the humoral immune response. However, due to continuous antigenic changes that occur on the surface of this glycoprotein, influenza viruses can escape the immune system and cause significant disease to the host. Toward the development of broad-spectrum therapeutics and vaccines against influenza virus, elucidation of conserved regions of influenza viruses is crucial. Thus, defining these types of epitopes through the generation and characterization of broadly neutralizing monoclonal antibodies (MAbs) can greatly assist others in highlighting conserved regions of hemagglutinin. Here, we demonstrate that MAb 9H10 that targets the hemagglutinin stalk has broadly neutralizing activity against group 2 influenza A viruses in vitro and in vivo.


PLOS Pathogens | 2016

Broadly-Reactive Neutralizing and Non-neutralizing Antibodies Directed against the H7 Influenza Virus Hemagglutinin Reveal Divergent Mechanisms of Protection

Gene S. Tan; Paul E. Leon; Randy A. Albrecht; Irina Margine; Ariana Hirsh; Justin Bahl; Florian Krammer

In the early spring of 2013, Chinese health authorities reported several cases of H7N9 influenza virus infections in humans. Since then the virus has established itself at the human-animal interface in Eastern China and continues to cause several hundred infections annually. In order to characterize the antibody response to the H7N9 virus we generated several mouse monoclonal antibodies against the hemagglutinin of the A/Shanghai/1/13 (H7N9) virus. Of particular note are two monoclonal antibodies, 1B2 and 1H5, that show broad reactivity to divergent H7 hemagglutinins. Monoclonal antibody 1B2 binds to viruses of the Eurasian and North American H7 lineages and monoclonal antibody 1H5 reacts broadly to virus isolates of the Eurasian lineage. Interestingly, 1B2 shows broad hemagglutination inhibiting and neutralizing activity, while 1H5 fails to inhibit hemagglutination and demonstrates no neutralizing activity in vitro. However, both monoclonal antibodies were highly protective in an in vivo passive transfer challenge model in mice, even at low doses. Experiments using mutant antibodies that lack the ability for Fc/Fc-receptor and Fc/complement interactions suggest that the protection provided by mAb 1H5 is, at least in part, mediated by the Fc-fragment of the mAb. These findings highlight that a protective response to a pathogen may not only be due to neutralizing antibodies, but can also be the result of highly efficacious non-neutralizing antibodies not readily detected by classical in vitro neutralization or hemagglutination inhibition assays. This is of interest because H7 influenza virus vaccines induce only low hemagglutination inhibiting antibody titers while eliciting robust antibody titers as measured by ELISA. Our data suggest that these binding but non-neutralizing antibodies contribute to protection in vivo.


Proceedings of the National Academy of Sciences of the United States of America | 2016

Optimal activation of Fc-mediated effector functions by influenza virus hemagglutinin antibodies requires two points of contact

Paul E. Leon; Wenqian He; Caitlin E. Mullarkey; Mark J. Bailey; Matthew S. Miller; Florian Krammer; Peter Palese; Gene S. Tan

Significance The mechanism of how antiviral antibodies induce Fc–FcγR effector functions remains to be fully elucidated. Although the ability to activate effector functions is attributed to antibody isotype, this does not fully address why identical isotypes have different capabilities to stimulate effector function. We show that antibodies that target the influenza virus hemagglutinin (HA) require a second intermolecular interaction to optimally activate effector cells. We demonstrate that the receptor-binding domain of the HA is required to bind to sialic acid expressed on the surface of effector cells to optimize effector cell activation. This finding provides a basic understanding of how an optimal antibody-dependent cell-mediated response against influenza virus is achieved and may allow for better vaccine design. Influenza virus strain-specific monoclonal antibodies (mAbs) provide protection independent of Fc gamma receptor (FcγR) engagement. In contrast, optimal in vivo protection achieved by broadly reactive mAbs requires Fc–FcγR engagement. Most strain-specific mAbs target the head domain of the viral hemagglutinin (HA), whereas broadly reactive mAbs typically recognize epitopes within the HA stalk. This observation has led to questions regarding the mechanism regulating the activation of Fc-dependent effector functions by broadly reactive antibodies. To dissect the molecular mechanism responsible for this dichotomy, we inserted the FLAG epitope into discrete locations on HAs. By characterizing the interactions of several FLAG-tagged HAs with a FLAG-specific antibody, we show that in addition to Fc–FcγR engagement mediated by the FLAG-specific antibody, a second intermolecular bridge between the receptor-binding region of the HA and sialic acid on effector cells is required for optimal activation. Inhibition of this second molecular bridge, through the use of an F(ab′)2 or the mutation of the sialic acid-binding site, renders the Fc–FcγR interaction unable to optimally activate effector cells. Our findings indicate that broadly reactive mAbs require two molecular contacts to possibly stabilize the immunologic synapse and potently induce antibody-dependent cell-mediated antiviral responses: (i) the interaction between the Fc of a mAb bound to HA with the FcγR of the effector cell and (ii) the interaction between the HA and its sialic acid receptor on the effector cell. This concept might be broadly applicable for protective antibody responses to viral pathogens that have suitable receptors on effector cells.


Nature Communications | 2017

Alveolar macrophages are critical for broadly-reactive antibody-mediated protection against influenza A virus in mice

Wenqian He; Chi-Jene Chen; Caitlin E. Mullarkey; Jennifer R. Hamilton; Christine K. Wong; Paul E. Leon; Melissa B. Uccellini; Veronika Chromikova; Carole Henry; Kevin W. Hoffman; Jean K. Lim; Patrick C. Wilson; Matthew S. Miller; Florian Krammer; Peter Palese; Gene S. Tan

The aim of candidate universal influenza vaccines is to provide broad protection against influenza A and B viruses. Studies have demonstrated that broadly reactive antibodies require Fc–Fc gamma receptor interactions for optimal protection; however, the innate effector cells responsible for mediating this protection remain largely unknown. Here, we examine the roles of alveolar macrophages, natural killer cells, and neutrophils in antibody-mediated protection. We demonstrate that alveolar macrophages play a dominant role in conferring protection provided by both broadly neutralizing and non-neutralizing antibodies in mice. Our data also reveal the potential mechanisms by which alveolar macrophages mediate protection in vivo, namely antibody-induced inflammation and antibody-dependent cellular phagocytosis. This study highlights the importance of innate effector cells in establishing a broad-spectrum antiviral state, as well as providing a better understanding of how multiple arms of the immune system cooperate to achieve an optimal antiviral response following influenza virus infection or immunization.Broadly reactive antibodies that recognize influenza A virus HA can be protective, but the mechanism is not completely understood. Here, He et al. show that the inflammatory response and phagocytosis mediated by the interaction between protective antibodies and macrophages are essential for protection.


Antimicrobial Agents and Chemotherapy | 2015

Direct Administration in the Respiratory Tract Improves Efficacy of Broadly Neutralizing Anti-Influenza Virus Monoclonal Antibodies

Victor H. Leyva-Grado; Gene S. Tan; Paul E. Leon; Mark A. Yondola; Peter Palese

ABSTRACT The emergence of influenza virus strains resistant to approved neuraminidase inhibitors and the time constrains after infection when these drugs can be effective constitute major drawbacks for this class of drugs. This highlights a critical need to discover new therapeutic agents that can be used for the treatment of influenza virus-infected patients. The use of broadly neutralizing anti-influenza monoclonal antibodies (MAbs) has been sought as an alternative immunotherapy against influenza infection. Here, we tested in mice previously characterized broadly neutralizing anti-hemagglutinin (HA) stalk MAbs prophylactically and therapeutically using different routes of administration. The efficacy of treatment against an influenza H1N1 pandemic virus challenge was compared between two systemic routes of administration, intraperitoneal (i.p.) and intravenous (i.v.), and two local routes, intranasal (i.n.) and aerosol (a.e.). The dose of MAb required for prophylactic protection was reduced by 10-fold in animals treated locally (i.n. or a.e.) compared with those treated systemically (i.p. or i.v.). Improved therapeutic protection was observed in animals treated i.n. on day 5 postinfection (60% survival) compared with those treated via the i.p. route (20% survival). An increase in therapeutic efficacy against other influenza virus subtypes (H5N1) was also observed when a local route of administration was used. Our findings demonstrate that local administration significantly decreases the amount of broadly neutralizing monoclonal antibody required for protection against influenza, which highlights the potential use of MAbs as a therapeutic agent for influenza-associated disease.


Journal of Visualized Experiments | 2018

A Method to Assess Fc-mediated Effector Functions Induced by Influenza Hemagglutinin Specific Antibodies

Mark J. Bailey; Felix Broecker; Paul E. Leon; Gene S. Tan

Antibodies play a crucial role in coupling the innate and adaptive immune responses against viral pathogens through their antigen binding domains and Fc-regions. Here, we describe how to measure the activation of Fc effector functions by monoclonal antibodies targeting the influenza virus hemagglutinin with the use of a genetically engineered Jurkat cell line expressing an activating type 1 Fc-FcγR. Using this method, the contribution of specific Fc-FcγR interactions conferred by immunoglobulins can be determined using an in vitro assay.


Journal of Visualized Experiments | 2017

Generation of Escape Variants of Neutralizing Influenza Virus Monoclonal Antibodies

Paul E. Leon; Teddy John Wohlbold; Wenqian He; Mark J. Bailey; Carole Henry; Patrick C. Wilson; Florian Krammer; Gene S. Tan

Influenza viruses exhibit a remarkable ability to adapt and evade the host immune response. One way is through antigenic changes that occur on the surface glycoproteins of the virus. The generation of escape variants is a powerful method in elucidating how viruses escape immune detection and in identifying critical residues required for antibody binding. Here, we describe a protocol on how to generate influenza A virus escape variants by utilizing human or murine monoclonal antibodies (mAbs) directed against the viral hemagglutinin (HA). With the use of our technique, we previously characterized critical residues required for the binding of antibodies targeting either the head or stalk of the novel avian H7N9 HA. The protocol can be easily adapted for other virus systems. Analyses of escape variants are important for modeling antigenic drift, determining single nucleotide polymorphisms (SNPs) conferring resistance and virus fitness, and in the designing of vaccines and/or therapeutics.


Cell Host & Microbe | 2016

Both Neutralizing and Non-Neutralizing Human H7N9 Influenza Vaccine-Induced Monoclonal Antibodies Confer Protection.

Carole J. Henry Dunand; Paul E. Leon; Min Huang; Angela Choi; Veronika Chromikova; Irvin Y. Ho; Gene S. Tan; John Cruz; Ariana Hirsh; Nai-Ying Zheng; Caitlin E. Mullarkey; Francis A. Ennis; Masanori Terajima; John J. Treanor; David J. Topham; Kanta Subbarao; Peter Palese; Florian Krammer; Patrick C. Wilson


Immunity | 2016

Targeting Viral Proteostasis Limits Influenza Virus, HIV, and Dengue Virus Infection

Nicholas S. Heaton; Natasha Moshkina; Romain Fenouil; Thomas J. Gardner; Sebastian Aguirre; Priya S. Shah; Nan Zhao; Lara Manganaro; Judd F. Hultquist; Justine Noel; David H. Sachs; Jennifer R. Hamilton; Paul E. Leon; Amit Chawdury; Shashank Tripathi; Camilla Melegari; Laura Campisi; Rong Hai; Giorgi Metreveli; Andrea V. Gamarnik; Adolfo García-Sastre; Benjamin D. Greenbaum; Viviana Simon; Ana Fernandez-Sesma; Nevan J. Krogan; Lubbertus C. F. Mulder; Harm van Bakel; Domenico Tortorella; Jack Taunton; Peter Palese

Collaboration


Dive into the Paul E. Leon's collaboration.

Top Co-Authors

Avatar

Gene S. Tan

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Peter Palese

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Florian Krammer

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Adolfo García-Sastre

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Caitlin E. Mullarkey

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Jennifer R. Hamilton

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Mark J. Bailey

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Rong Hai

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Wenqian He

Icahn School of Medicine at Mount Sinai

View shared research outputs
Researchain Logo
Decentralizing Knowledge