Emily M. Plummer
La Jolla Institute for Allergy and Immunology
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Emily M. Plummer.
Antiviral Research | 2013
Stuart T. Perry; Michael D. Buck; Emily M. Plummer; Raju Penmasta; Hitesh Batra; Eric Stavale; Kelly L. Warfield; Raymond A. Dwek; Terry D. Butters; Dominic S. Alonzi; Steven M. Lada; Kevin King; Brennan Klose; Urban Ramstedt; Sujan Shresta
The aim of the present study was to evaluate the ability of the iminosugar drug UV-4 to provide in vivo protection from lethal dengue virus (DENV) challenge. This study utilized a well-described model of dengue hemorrhagic fever/dengue shock syndrome (DHF/DSS)-like lethal disease in AG129 mice lacking the type I and II interferon receptors. Herein, we present UV-4 as a potent iminosugar for controlling DENV infection and disease in this mouse model. Specifically, administration of UV-4 reduced mortality, as well as viremia and viral RNA in key tissues, and cytokine storm. In addition, UV-4 treatment can be delayed, and it does not alter the anti-DENV antibody response. These results have set the foundation for development of UV-4 as a DENV-specific antiviral in phase I human clinical trials.
Journal of Virology | 2012
Tyler R. Prestwood; Monica M. May; Emily M. Plummer; Malika M. Morar; Lauren E. Yauch; Sujan Shresta
ABSTRACT Human postmortem studies of natural dengue virus (DENV) infection have reported systemically distributed viral antigen. Although it is widely accepted that DENV infects mononuclear phagocytes, the sequence in which specific tissues and cell types are targeted remains uncharacterized. We previously reported that mice lacking alpha/beta and gamma interferon receptors permit high levels of DENV replication and show signs of systemic disease (T. R. Prestwood et al., J. Virol. 82:8411–8421, 2008). Here we demonstrate that within 6 h, DENV traffics to and replicates in both CD169+ and SIGN-R1+ macrophages of the splenic marginal zone or draining lymph node, respectively, following intravenous or intrafootpad inoculation. Subsequently, high levels of replication are detected in F4/80+ splenic red pulp macrophages and in the bone marrow, lymph nodes, and Peyers patches. Intravenously inoculated mice begin to succumb to dengue disease 72 h after infection, at which time viral replication occurs systemically, except in lymphoid tissues. In particular, high levels of replication occur in CD68+ macrophages of the kidneys, heart, thymus, and gastrointestinal tract. Over the course of infection, proportionately large quantities of DENV traffic to the liver and spleen. However, late during infection, viral trafficking to the spleen decreases, while trafficking to the liver, thymus, and kidneys increases. The present study demonstrates that macrophage populations, initially in the spleen and other lymphoid tissues and later in nonlymphoid tissues, are major targets of DENV infection in vivo.
Scientific Reports | 2015
Seleeke Flingai; Emily M. Plummer; Ami Patel; Sujan Shresta; Janess Mendoza; Kate E. Broderick; Niranjan Y. Sardesai; Kar Muthumani; David B. Weiner
Dengue virus (DENV) is the most important mosquito-borne viral infection in humans. In recent years, the number of cases and outbreaks has dramatically increased worldwide. While vaccines are being developed, none are currently available that provide balanced protection against all DENV serotypes. Advances in human antibody isolation have uncovered DENV neutralizing antibodies (nAbs) that are capable of preventing infection from multiple serotypes. Yet delivering monoclonal antibodies using conventional methods is impractical due to high costs. Engineering novel methods of delivering monoclonal antibodies could tip the scale in the fight against DENV. Here we demonstrate that simple intramuscular delivery by electroporation of synthetic DNA plasmids engineered to express modified human nAbs against multiple DENV serotypes confers protection against DENV disease and prevents antibody-dependent enhancement (ADE) of disease in mice. This synthetic nucleic acid antibody prophylaxis/immunotherapy approach may have important applications in the fight against infectious disease.
Journal of Virology | 2015
Emily M. Plummer; Michael D. Buck; Marisa Sanchez; Jason Greenbaum; Julia Turner; Rajvir Grewal; Brennan Klose; Aruna Sampath; Kelly L. Warfield; Bjoern Peters; Urban Ramstedt; Sujan Shresta
ABSTRACT The host-targeted antiviral drug UV-4B reduces viral replication and promotes survival in a mouse model of experimental dengue virus (DENV) infection. UV-4B is an iminosugar that inhibits the α-glucosidase family of enzymes and subsequently the folding of glycosylated proteins, both viral and host. Here, we utilized next-generation sequencing to investigate evolution of a flavivirus under selective pressure by a host-targeted antiviral in vivo. In viral populations recovered from UV-4B-treated mice, there was a significant increase in the number of single-nucleotide polymorphisms (SNPs) and the ratio of nonsynonymous to synonymous SNPs compared to findings in viral populations from vehicle-treated mice. The strongest evidence of positive selection was in the glycosylated membrane protein, thereby providing in vivo validation of the mechanism of action of an iminosugar. In addition, mutations in glycosylated proteins were present only in drug-treated mice after a single passage. However, the bulk of the other mutations were present in both populations, indicating nonspecific selective pressure. Together with the continued control of viremia by UV-4B, these findings are consistent with the previously predicted high genetic barrier to escape mutations in host-targeted antivirals. IMPORTANCE Although hundreds of millions of people are infected with DENV every year, there is currently no approved vaccine or antiviral therapy. UV-4B has demonstrated antiviral activity against DENV and is expected to enter clinical trials soon. Therefore, it is important to understand the mechanisms of DENV resistance to UV-4B. Host-targeted antivirals are thought to have a higher genetic barrier to escape mutants than directly acting antivirals, yet there are very few published studies of viral evolution under host-targeted antivirals. No study to date has described flavivirus evolution in vivo under selective pressure by a host-based antiviral drug. We present the first in vivo study of the sequential progression of viral evolution under selective pressure by a host-targeted antiviral compound. This study bolsters support for the clinical development of UV-4B as an antiviral drug against DENV, and it provides a framework to compare how treatment with other host-targeted antiflaviviral drugs in humans and different animal models influence viral genetic diversity.
Antiviral Research | 2016
Kelly L. Warfield; Emily M. Plummer; Andrew C. Sayce; Dominic S. Alonzi; William W. Tang; Beatrice E. Tyrrell; Michelle L. Hill; Alessandro T. Caputo; Sarah S. Killingbeck; P. Robert Beatty; Eva Harris; Ren Iwaki; Kyoko Kinami; Daisuke Ide; J. L. Kiappes; Atsushi Kato; Michael D. Buck; Kevin King; William E. Eddy; Mansoora Khaliq; Aruna Sampath; Anthony M. Treston; Raymond A. Dwek; Sven Enterlein; Joanna L. Miller; Nicole Zitzmann; Urban Ramstedt; Sujan Shresta
The antiviral activity of UV-4 was previously demonstrated against dengue virus serotype 2 (DENV2) in multiple mouse models. Herein, step-wise minimal effective dose and therapeutic window of efficacy studies of UV-4B (UV-4 hydrochloride salt) were conducted in an antibody-dependent enhancement (ADE) mouse model of severe DENV2 infection in AG129 mice lacking types I and II interferon receptors. Significant survival benefit was demonstrated with 10–20 mg/kg of UV-4B administered thrice daily (TID) for seven days with initiation of treatment up to 48 h after infection. UV-4B also reduced infectious virus production in in vitro antiviral activity assays against all four DENV serotypes, including clinical isolates. A set of purified enzyme, in vitro, and in vivo studies demonstrated that inhibition of endoplasmic reticulum (ER) α-glucosidases and not the glycosphingolipid pathway appears to be responsible for the antiviral activity of UV-4B against DENV. Along with a comprehensive safety package, these and previously published data provided support for an Investigational New Drug (IND) filing and Phases 1 and 2 clinical trials for UV-4B with an indication of acute dengue disease.
Viruses | 2015
Kelly L. Warfield; Emily M. Plummer; Dominic S. Alonzi; Gary W. Wolfe; Aruna Sampath; Tam Nguyen; Terry D. Butters; Sven Enterlein; Eric Stavale; Sujan Shresta; Urban Ramstedt
Iminosugars are capable of targeting the life cycles of multiple viruses by blocking host endoplasmic reticulum α-glucosidase enzymes that are required for competent replication of a variety of enveloped, glycosylated viruses. Iminosugars as a class are approved for use in humans with diseases such as diabetes and Gaucher’s disease, providing evidence for safety of this class of compounds. The in vitro antiviral activity of iminosugars has been described in several publications with a subset of these demonstrating in vivo activity against flaviviruses, herpesviruses, retroviruses and filoviruses. Although there is compelling non-clinical in vivo evidence of antiviral efficacy, the efficacy of iminosugars as antivirals has yet to be demonstrated in humans. In the current study, we report a novel iminosugar, UV-12, which has efficacy against dengue and influenza in mouse models. UV-12 exhibits drug-like properties including oral bioavailability and good safety profile in mice and guinea pigs. UV-12 is an example of an iminosugar with activity against multiple virus families that should be investigated in further safety and efficacy studies and demonstrates potential value of this drug class as antiviral therapeutics.
Journal of Immunological Methods | 2014
Emily M. Plummer; Sujan Shresta
Dengue virus (DENV) has substantial global impact, with an estimated 390million people infected each year. In spite of this, there is currently no approved DENV-specific vaccine or antiviral. One reason for this is the difficulty involved with development of an adequate animal model. While non-human primates support viral replication, they do not exhibit signs of clinical disease. A mouse model is an ideal alternative; however, wild-type mice are resistant to DENV-induced disease. Infection of interferon receptor-deficient mice results in disease that recapitulates key features of severe dengue disease in humans. For the development of vaccines, interferon receptor-deficient mice provide a stringent model for testing vaccine-induced immune components from vaccinated wild-type mice.
Cell Reports | 2017
Aaron F. Carlin; Emily M. Plummer; Edward A. Vizcarra; Nicholas Sheets; Yunichel Joo; William W. Tang; Jeremy Day; Jay Greenbaum; Christopher K. Glass; Michael S. Diamond; Sujan Shresta
Interferon-regulatory factors (IRFs) are a family of transcription factors (TFs) that translate viral recognition into antiviral responses, including type I interferon (IFN) production. Dengue virus (DENV) and other clinically important flaviviruses are suppressed by type I IFN. While mice lacking the type I IFN receptor (Ifnar1-/-) succumb to DENV infection, we found that mice deficient in three transcription factors controlling type I IFN production (Irf3-/-Irf5-/-Irf7-/- triple knockout [TKO]) survive DENV challenge. DENV infection of TKO mice resulted in minimal type I IFN production but a robust type II IFN (IFN-γ) response. Using loss-of-function approaches for various molecules, we demonstrate that the IRF-3-, IRF-5-, IRF-7-independent pathway predominantly utilizes IFN-γ and, to a lesser degree, type I IFNs. This pathway signals via IRF-1 to stimulate interleukin-12 (IL-12) production and IFN-γ response. These results reveal a key antiviral role for IRF-1 by activating both type I and II IFN responses during DENV infection.
Methods of Molecular Biology | 2014
Emily M. Plummer; Sujan Shresta
Validation of a mouse model of dengue virus (DENV) infection relies on verification of viremia and productive replication in mouse tissues following infection. Here, we describe a quantitative assay for determining viral RNA levels in mouse serum and tissues. For the purpose of confirming DENV replication, we outline a fluorescence immunohistochemistry (FIHC) protocol for staining a nonstructural protein of DENV.
Molecular Therapy | 2016
Seleeke Flingai; Emily M. Plummer; Ami Patel; Janess Mendoza; Yang Wang; Keith Riemann; Mark S. Klempner; Kate E. Broderick; Niranjan Y. Sardesai; Kar Muthumani; David B. Weiner
The development of vaccines against arthropod-borne infectious diseases has been wrought with difficulties. Recent advances in human antibody isolation have uncovered neutralizing monoclonal antibodies (mAbs) that are capable of providing protection against pathogen challenge in various animal models. Yet generating and delivering biologically-relevant levels of such antibodies using conventional monoclonal antibody methodology is impractical, often requiring huge expenses and repeated administrations for clinical benefit. Creating new methods of delivering monoclonal antibodies could drastically tip the scale in the fight against a number of devastating pathogens.Here, we describe an approach to delivering neutralizing mAbs in vivo using DNA plasmid-mediated antibody gene transfer. This approach, which we term DNA mAb (DMAb) delivery, generates biologically relevant levels of mAbs after a single intramuscular injection of antibody-encoding DNA followed by in vivo electroporation (EP). First, we demonstrate the ability of DMAb technology to deliver cross-reactive neutralizing antibodies against DENV into the host circulation. Since this approach allows for genetic tailoring of the exact features of the desired antibody, we incorporated Fc region modifications to a naturally occurring human anti-DENV neutralizing antibody to enhance antibody function in vivo. We show that intramuscular delivery in mice of pDVSF-3 LALA, which encodes a human anti-DENV1-3 IgG1 neutralizing antibody modified with a mutation that abrogates FcγR binding, produces anti-DENV antisera capable of binding and neutralizing DENV1-3. Importantly, mice receiving pDVSF-3 LALA, but not the unmodified pDVSF-3 WT, were protected from both virus-only disease and antibody-enhanced lethal disease.Using a similar, targeted genetic approach to antibody modifications, we also show that DMAbs encoding antibodies against Borrelia burgdorferi (the causative agent of Lyme disease) can undergo extensive amino acid modifications that substantially increase in vivo mAb production levels compared to wild-type DMAb sequences. These data illustrate a subset of the functional optimizations made possible with the DMAb platform.This work was supported by grants funded to DBW through the National Institutes of Health, the DARPA-PROTECT award, and Inovio Pharmaceuticals Inc.
Collaboration
Dive into the Emily M. Plummer's collaboration.
United States Army Medical Research Institute of Infectious Diseases
View shared research outputs