Network


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

Hotspot


Dive into the research topics where Maryna C. Eichelberger is active.

Publication


Featured researches published by Maryna C. Eichelberger.


Clinical Infectious Diseases | 2010

Rapid Selection of Oseltamivir-and Peramivir-Resistant Pandemic H1N1 Virus during Therapy in 2 Immunocompromised Hosts

Matthew J. Memoli; Rachel J. Hrabal; Arash Hassantoufighi; Maryna C. Eichelberger; Jeffery K. Taubenberger

Pandemic 2009 H1N1 virus isolates containing the neuraminidase inhibitor resistance mutation H275Y have been reported. We describe rapid selection for the H275Y resistance mutation during therapy in 2 immunocompromised individuals at 9 and 14 days of therapy, as well as the first described case of clinically significant resistance to peramivir.


Proceedings of the National Academy of Sciences of the United States of America | 2011

Discordant antigenic drift of neuraminidase and hemagglutinin in H1N1 and H3N2 influenza viruses

Matthew R. Sandbulte; Kim B. Westgeest; Jin Gao; Xiyan Xu; Alexander Klimov; Colin A. Russell; David F. Burke; Derek J. Smith; Ron A. M. Fouchier; Maryna C. Eichelberger

Seasonal epidemics caused by influenza virus are driven by antigenic changes (drift) in viral surface glycoproteins that allow evasion from preexisting humoral immunity. Antigenic drift is a feature of not only the hemagglutinin (HA), but also of neuraminidase (NA). We have evaluated the antigenic evolution of each protein in H1N1 and H3N2 viruses used in vaccine formulations during the last 15 y by analysis of HA and NA inhibition titers and antigenic cartography. As previously shown for HA, genetic changes in NA did not always lead to an antigenic change. The noncontinuous pattern of NA drift did not correspond closely with HA drift in either subtype. Although NA drift was demonstrated using ferret sera, we show that these changes also impact recognition by NA-inhibiting antibodies in human sera. Remarkably, a single point mutation in the NA of A/Brisbane/59/2007 was primarily responsible for the lack of inhibition by polyclonal antibodies specific for earlier strains. These data underscore the importance of NA inhibition testing to define antigenic drift when there are sequence changes in NA.


Vaccine | 2008

FDA/NIH/WHO public workshop on immune correlates of protection against influenza A viruses in support of pandemic vaccine development, Bethesda, Maryland, US, December 10-11, 2007.

Maryna C. Eichelberger; Hana Golding; Maureen A. Hess; Jerry P. Weir; Kanta Subbarao; Catherine J. Luke; Martin Friede; David Wood

The goals of the workshop were to identify gaps in our knowledge and abilities to address the unique challenges encountered in the development of vaccines intended to protect against pandemic influenza and to facilitate implementation of a global research agenda to improve efficacy assessment of pandemic influenza vaccines. This workshop included discussions on: (i) current knowledge regarding immune correlates of protection against seasonal influenza; (ii) human immune responses to avian influenza infection and vaccines for novel influenza viruses; (iii) limitations of currently available assays to evaluate vaccine immunogenicity; and (iv) potential insights from animal models for correlates of protection against avian influenza.


Influenza and Other Respiratory Viruses | 2009

A miniaturized assay for influenza neuraminidase‐inhibiting antibodies utilizing reverse genetics‐derived antigens

Matthew Sandbulte; Jin Gao; Timothy M. Straight; Maryna C. Eichelberger

Background  Antibodies to neuraminidase (NA) contribute to protection during influenza virus infection, but NA inhibition (NI) titers are not routinely analyzed in vaccine trials. One reason is the cumbersome nature of the conventional thiobarbituric acid (TBA) NI assay, which uses chemical methods to quantify free sialic acid following incubation of NA with substrate in the presence of serum. In addition, the assay is complicated by the need to use virus of a hemagglutinin (HA) subtype novel to the host to detect NA‐specific antibodies only.


Journal of Virology | 2013

Molecular Basis for Broad Neuraminidase Immunity: Conserved Epitopes in Seasonal and Pandemic H1N1 as well as H5N1 Influenza Viruses

Hongquan Wan; Jin Gao; Kemin Xu; Hongjun Chen; Laura Couzens; Katie Rivers; Judy D. Easterbrook; Kevin Yang; Lei Zhong; Mohsen Rajabi; Jianqiang Ye; Ishrat Sultana; Xiu-Feng Wan; X. Liu; Daniel R. Perez; Jeffery K. Taubenberger; Maryna C. Eichelberger

ABSTRACT Influenza A viruses, including H1N1 and H5N1 subtypes, pose a serious threat to public health. Neuraminidase (NA)-related immunity contributes to protection against influenza virus infection. Antibodies to the N1 subtype provide protection against homologous and heterologous H1N1 as well as H5N1 virus challenge. Since neither the strain-specific nor conserved epitopes of N1 have been identified, we generated a panel of mouse monoclonal antibodies (MAbs) that exhibit different reactivity spectra with H1N1 and H5N1 viruses and used these MAbs to map N1 antigenic domains. We identified 12 amino acids essential for MAb binding to the NA of a recent seasonal H1N1 virus, A/Brisbane/59/2007. Of these, residues 248, 249, 250, 341, and 343 are recognized by strain-specific group A MAbs, while residues 273, 338, and 339 are within conserved epitope(s), which allows cross-reactive group B MAbs to bind the NAs of seasonal H1N1 and the 1918 and 2009 pandemic (09pdm) H1N1 as well as H5N1 viruses. A single dose of group B MAbs administered prophylactically fully protected mice against lethal challenge with seasonal and 09pdm H1N1 viruses and resulted in significant protection against the highly pathogenic wild-type H5N1 virus. Another three N1 residues (at positions 396, 397, and 456) are essential for binding of cross-reactive group E MAbs, which differ from group B MAbs in that they do not bind 09pdm H1N1 viruses. The identification of conserved N1 epitopes reveals the molecular basis for NA-mediated immunity between H1N1 and H5N1 viruses and demonstrates the potential for developing broadly protective NA-specific antibody treatments for influenza.


Journal of Virological Methods | 2014

An optimized enzyme-linked lectin assay to measure influenza A virus neuraminidase inhibition antibody titers in human sera.

Laura Couzens; Jin Gao; Kim B. Westgeest; Matthew Sandbulte; Vladimir Y. Lugovtsev; Ron A. M. Fouchier; Maryna C. Eichelberger

Antibodies to neuraminidase (NA), the second most abundant surface protein on influenza virus, contribute toward protection against influenza. The traditional thiobarbituric acid (TBA) method to quantify NA inhibiting antibodies is cumbersome and not suitable for routine serology. An enzyme-linked lectin assay (ELLA) described by Lambre et al. (1990) is a practical alternative method for measuring NA inhibition (NI) titers. This report describes optimization of the ELLA for measuring NI titers in human sera against influenza A viruses, using H6N1 and H6N2 viruses as antigens. The optimized ELLA is subtype-specific and reproducible. While the titers measured by ELLA are somewhat greater than those measured by a miniaturized TBA method, seroconversion rates are the same, suggesting similarity in assay sensitivity under these optimized conditions. The ELLA described in this report provides a practical format for routine evaluation of human antibody responses to NA.


Vaccine | 2010

A practical influenza neutralization assay to simultaneously quantify hemagglutinin and neuraminidase-inhibiting antibody responses.

Arash Hassantoufighi; Henry Zhang; Matthew Sandbulte; Jin Gao; Jody Manischewitz; Lisa King; Hana Golding; Timothy Straight; Maryna C. Eichelberger

Influenza vaccine immunogenicity is commonly assessed by determining hemagglutination inhibition (HAI) titers in serum samples. HAI titers have been used to predict vaccine efficacy, but this often fails when live attenuated vaccines are evaluated, because it does not encompass all immune mediators of protection. Although antibodies that inhibit viral neuraminidase (NA) also contribute to protection against disease, there is currently no routine assessment of NA inhibition titers. A serological method with the capacity to measure functional inhibition of both HA and NA would be valuable. We developed a high-throughput virus neutralization assay that uses viral NA activity to quantify influenza replication (the AVINA assay), and showed its capacity to identify antivirals with a broad range of target specificities. In this report we demonstrate that antibodies with specificity for either HA or NA are detected in this assay, whereas a commonly used virus neutralization assay only detects those with HA-specificity. We also compared human responses to seasonal influenza vaccines measured by HAI, micro-neutralization, NA inhibition and AVINA assays. The response rates to both trivalent inactivated and live attenuated vaccines were greater when measured by the AVINA than the other assays, reflecting the dual antigen reactivity and increased sensitivity of the assay. The potential of this single assay to predict protection against influenza-induced tachypnea was demonstrated in vaccinated cotton rats. The AVINA assay is therefore a practical, comprehensive method to determine influenza vaccine immunogenicity and potential efficacy.


Virology | 2012

Protection against a lethal H5N1 influenza challenge by intranasal immunization with virus-like particles containing 2009 pandemic H1N1 neuraminidase in mice.

Judith D. Easterbrook; Louis M. Schwartzman; Jin Gao; John C. Kash; David M. Morens; Laura Couzens; Hongquan Wan; Maryna C. Eichelberger; Jeffery K. Taubenberger

Highly pathogenic H5N1 influenza shares the same neuraminidase (NA) subtype with the 2009 pandemic (H1N1pdm09), and cross-reactive NA immunity might protect against or mitigate lethal H5N1 infection. In this study, mice were either infected with a sublethal dose of H1N1pdm09 or were vaccinated and boosted with virus-like particles (VLP) consisting of the NA and matrix proteins, standardized by NA activity and administered intranasally, and were then challenged with a lethal dose of HPAI H5N1 virus. Mice previously infected with H1N1pdm09 survived H5N1 challenge with no detectable virus or respiratory tract pathology on day 4. Mice immunized with H5N1 or H1N1pdm09 NA VLPs were also fully protected from death, with a 100-fold and 10-fold reduction in infectious virus, respectively, and reduced pathology in the lungs. Human influenza vaccines that elicit not only HA, but also NA immunity may provide enhanced protection against the emergence of seasonal and pandemic viruses.


Journal of Proteome Research | 2013

Comparative Glycomics Analysis of Influenza Hemagglutinin (H5N1) Produced in Vaccine Relevant Cell Platforms

Yanming An; Joseph A. Rininger; Donald L. Jarvis; Xianghong Jing; Zhiping Ye; Jared J. Aumiller; Maryna C. Eichelberger; John F. Cipollo

Hemagglutinin (HA) is the major antigen in influenza vaccines, and glycosylation is known to influence its antigenicity. Embryonated hen eggs are traditionally used for influenza vaccine production, but vaccines produced in mammalian and insect cells were recently licensed. This raises the concern that vaccines produced with different cell systems might not be equivalent due to differences in their glycosylation patterns. Thus, we developed an analytical method to monitor vaccine glycosylation through a combination of nanoLC/MS(E) and quantitative MALDI-TOF MS permethylation profiling. We then used this method to examine glycosylation of HAs from two different influenza H5N1 strains produced in five different platforms, including hen eggs, three different insect cell lines (High Five, expresSF+ and glycoengineered expresSF+), and a human cell line (HEK293). Our results demonstrated that (1) sequon utilization is not necessarily equivalent in different cell types, (2) there are quantitative and qualitative differences in the overall N-glycosylation patterns and structures produced by different cell types, (3) ∼20% of the N-glycans on the HAs produced by High Five cells are core α1,3-fucosylated structures, which may be allergenic in humans, and (4) our method can be used to monitor differences in glycosylation during the cellular glycoengineering stages of vaccine development.


The Journal of Infectious Diseases | 2010

Rapid Selection of a Transmissible Multidrug-Resistant Influenza A/H3N2 Virus in an Immunocompromised Host

Matthew J. Memoli; Rachel J. Hrabal; Arash Hassantoufighi; Brett W. Jagger; Zong-Mei Sheng; Maryna C. Eichelberger; Jeffery K. Taubenberger

BACKGROUND The overall impact of influenza virus infection in immunocompromised patients is largely unknown. Antigenic drift and genetic variations during prolonged influenza infection have been demonstrated. In this report we describe a multidrug-resistant H3N2 influenza virus isolated from an immunocompromised patient after 5 days of therapy. METHODS Multiple nasal wash samples were collected from an infected patient, and viral isolates were characterized. Sensitivity to antiviral agents was evaluated. Fitness and transmissibility were assessed in ferrets and tissue culture. RESULTS An in-frame 4-amino acid deletion emerged in the neuraminidase (NA) gene of an H3N2 virus after 5 days of oseltamivir therapy. No other changes in the NA or hemagglutinin genes were noted. Drug sensitivity assays revealed resistance to oseltamivir (>10-fold increase in 50% inhibitory concentration [IC(50)]) and reduction in sensitivity to zanamivir (3-7-fold increase in IC(50) or 50% effective concentration). No change in fitness or transmissibility was observed. CONCLUSIONS An in-frame NA gene deletion was rapidly selected for in an immunocompromised patient, resulting in decreased sensitivity of the isolate to available NA inhibitors without a change in fitness or transmissibility. This finding has implications for our understanding of the emergence of antiviral resistance and treatment of patients with influenza A infection, especially those who are immunocompromised.

Collaboration


Dive into the Maryna C. Eichelberger's collaboration.

Top Co-Authors

Avatar

Jin Gao

Center for Biologics Evaluation and Research

View shared research outputs
Top Co-Authors

Avatar

Laura Couzens

Center for Biologics Evaluation and Research

View shared research outputs
Top Co-Authors

Avatar

Hongquan Wan

Center for Biologics Evaluation and Research

View shared research outputs
Top Co-Authors

Avatar

Ishrat Sultana

Center for Biologics Evaluation and Research

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Michail A. Alterman

Center for Biologics Evaluation and Research

View shared research outputs
Top Co-Authors

Avatar

Arash Hassantoufighi

Center for Biologics Evaluation and Research

View shared research outputs
Top Co-Authors

Avatar

Melkamu Getie-Kebtie

Center for Biologics Evaluation and Research

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Lianlian Jiang

Center for Biologics Evaluation and Research

View shared research outputs
Researchain Logo
Decentralizing Knowledge