Elsevier

Antiviral Research

Volume 131, July 2016, Pages 92-99
Antiviral Research

A humanized monoclonal antibody neutralizes yellow fever virus strain 17D-204 in vitro but does not protect a mouse model from disease

https://doi.org/10.1016/j.antiviral.2016.04.013Get rights and content

Highlights

  • Murine-human chimeric MAb 864-cIgG was developed for therapy of severe adverse effects (SAEs) from YFV vaccination.

  • MAb 864-cIgG bound YFV vaccine strain 17D-204 in ELISA and neutralized 17D-204 infectivity for cell culture.

  • MAb 864-cIgG had no protective capacity in the IFN α/β and γ receptor-deficient AG129 mouse model of 17D-204 infection.

  • MAb 864-cIgG therapy appears to have no potential for therapy of SAEs associated with YFV vaccination.

Abstract

The yellow fever virus (YFV) vaccine 17D-204 is considered safe and effective, yet rare severe adverse events (SAEs), some resulting in death, have been documented following vaccination. Individuals exhibiting post-vaccinal SAEs are ideal candidates for antiviral monoclonal antibody (MAb) therapy; the time until appearance of clinical signs post-exposure is usually short and patients are quickly hospitalized. We previously developed a murine-human chimeric monoclonal antibody (cMAb), 2C9-cIgG, reactive with both virulent YFV and 17D-204, and demonstrated its ability to prevent and treat YF disease in both AG129 mouse and hamster models of infection. To counteract possible selection of 17D-204 variants that escape neutralization by treatment with a single MAb (2C9-cIgG), we developed a second cMAb, 864-cIgG, for use in combination with 2C9-cIgG in post-vaccinal therapy. MAb 864-cIgG recognizes/neutralizes only YFV 17D-204 vaccine substrain and binds to domain III (DIII) of the viral envelope protein, which is different from the YFV type-specific binding site of 2C9-cIgG in DII. Although it neutralized 17D-204 in vitro, administration of 864-cIgG had no protective capacity in the interferon receptor-deficient AG129 mouse model of 17D-204 infection. The data presented here show that although DIII-specific 864-cIgG neutralizes virus infectivity in vitro, it does not have the ability to abrogate disease in vivo. Therefore, combination of 864-cIgG with 2C9-cIgG for treatment of YF vaccination SAEs does not appear to provide an improvement on 2C9-cIgG therapy alone.

Introduction

Despite the availability of a vaccine considered generally safe and very effective, yellow fever (YF) remains an important mosquito-borne disease in tropical regions of South America and Africa. It is estimated that there are 180,000 human infections and 78,000 deaths annually due to YF (Garske et al., 2014). Yellow fever virus (YFV) is the prototype flavivirus and has a positive sense, single-stranded RNA genome that encodes three structural proteins (C, prM/M, and E) and seven nonstructural proteins (Chambers et al., 1990, Hahn et al., 1987, Rice et al., 1985). The virion is composed of a nucleocapsid surrounded by an envelope derived from infected cell membranes that contains the E and M proteins. The E or envelope glycoprotein is the major flaviviral surface protein, and is arranged as a scaffold of 90 homodimers on the mature virion surface (Kaufmann et al., 2010, Kuhn et al., 2002, Zhang et al., 2013a). The E protein is a class II fusion protein that contains the major determinants for cell attachment, cell entry of virions, and elicitation of virus-neutralizing antibodies (Crill and Roehrig, 2001, Heinz, 1986, Heinz and Allison, 2003, Roehrig, 2003, Stiasny et al., 2007). The crystal structure of the E protein has been solved for several flaviviruses, although not for YFV (Luca et al., 2012, Luca et al., 2013, Modis et al., 2003, Modis et al., 2005, Nybakken et al., 2006, Rey et al., 1995). The flaviviral E protein can be divided into three structural domains: DI, DII, and DIII. Epitopes in DII and DIII elicit virus-neutralizing monoclonal antibodies (MAbs) and anti-DIII MAbs usually have more potent in vitro neutralization titers than anti-DII MAbs (Roehrig, 2003). Certain anti-E MAbs possess antiviral prophylactic and therapeutic activity in animal models of flavivirus infection (Brandriss et al., 1986, Engle and Diamond, 2003, Gould et al., 1986, Hawkes et al., 1988, Johnson and Roehrig, 1999, Julander et al., 2014, Kimura-Kuroda and Yasui, 1988, Mathews and Roehrig, 1984, Thibodeaux et al., 2012b), and the humanized anti-West Nile virus (WNV) EDIII MAb MGAWN1 has demonstrated efficacy in animal models and undergone Phase I clinical trials to demonstrate safety in humans (Beigel et al., 2010). We recently developed a YFVtype-specific chimeric murine-human MAb, 2C9-cIgG, and demonstrated its prophylactic and therapeutic activity in two animal models of infection (Julander et al., 2014, Thibodeaux et al., 2012b). MAb 2C9-cIgG reacts with both virulent and vaccine YFV, binding to an epitope in DII of the E protein (Lobigs et al., 1987). Interferon receptor-deficient AG129 mice were protected from or successfully treated after challenge with 17D-204 when the cMAb was inoculated 24 h prior to or 24 h after viral infection (Thibodeaux et al., 2012b). MAb 2C9-cIgG was more effective in an immunocompetent hamster model challenged with virulent YFV Jimenez strain (Julander et al., 2014). Hamsters were protected from disease when 2C9-cIgG was administered 24 h before and up to 72 h post-infection (PI).

Yellow fever vaccination with live-attenuated 17D-204 is generally considered safe and effective; however, rare severe adverse events (SAEs), some resulting in death, have been documented following vaccination, particularly in individuals with innate immunity defects or >60 years of age (Wkly Epidemiol Rec, CDC, 2001, Martin et al., 2001, Monath, 2010, Vasconcelos et al., 2001). SAEs are not due to mutations in the vaccine virus but rather to as yet undetermined host-specified factors (Monath, 2010). There are no specific therapies for YFV infection (Julander, 2013, Monath, 2008). Because the timing of viral exposure is known for vacinees, individuals experiencing post-vaccinal SAEs are likely candidates for anti-YF antibody therapy. One theoretical limitation of single MAb therapy for flaviruses is the high mutation rate of flaviviral ssRNA genomes, which could result in generation of MAb escape mutants (Ryman et al., 1998). Neutralization escape variants of WNV have been selected both in vitro and in vivo following single dose MAb treatment (Zhang et al., 2009, Zhang et al., 2010). To reduce this possibility, cocktails of MAbs reactive with different E protein epitopes might be more effective for therapy.

We report here the generation of a second YFV-reactive cMAb, 864-cIgG, and its in vitro and in vivo activity. The parent murine MAb 864 (m864) was isolated following immunization of mice with 17D-204 (Buckley and Gould, 1985, Cammack and Gould, 1986, Gould et al., 1985, Gould et al., 1986). MAb 864 is substrain specific and reacts only with YFV 17D-204 vaccine, neutralizes virus infectivity, and has been shown to protect mice from virus challenge when administered to 3–4 week-old immunocompetent mice as mouse ascitic fluid 24 h before YF-17D challenge via the intracerebral route (Cammack and Gould, 1986, Gould et al., 1986). Unlike mMAb 2C9, mMAb 864 identifies a neutralization epitope in DIII of the E protein (Ryman et al., 1998); thus we predicted that combined therapy using 864-cIgG and 2C9-cIgG should increase therapeutic efficacy compared to 2C9-cIgG alone.

Section snippets

Cells and viruses

The previously characterized murine hybridoma line, m864, was obtained from the CDC-Division of Vector-Borne Diseases (DVBD), Fort Collins, CO, and was cultured in Dulbecco's modified minimum essential medium (DMEM) with 15% fetal calf serum (FCS). Ag8.653 and Vero cells were cultured in the same medium as the hybridoma, supplemented with 10% FCS (DMEM-10).

YFV 17D-204 was obtained from the CDC-DVBD, Fort Collins, CO. Its passage history is unknown. Purified virus was prepared as previously

Locations of m864 and m2C9 binding sites on the YFV E protein

The YFV E protein amino acids (AAs) shown to be critical for binding of m864 and m2C9 by isolation and genome sequencing of neutralization escape mutants are shown in a ribbon structure of the E protein in Fig. 1. Mutations resulting in substitutions in 17D-204 E protein AAs N71, D72, and possibly M125 in DII (shown in yellow) were shown to disrupt binding of 2C9 (Lobigs et al., 1987). As expected for a YFV type-specific epitope, these residues are identical in the parental YFV Asibi strain and

Discussion

Despite the existence of an effective YF vaccine, there are instances in which prophylactic/therapeutic MAbs for YFV might be needed. First, a large number of virulent YFV infections and deaths occur annually among unvaccinated individuals in endemic areas. Effective prophylactic or therapeutic MAbs could be used for treatment of unvaccinated, at-risk individuals in the event of an outbreak. Our previously-developed anti-YFV MAb 2C9-cIgG was shown in animal models to mitigate disease due to

Acknowledgements

This work was funded by NIH/NIAID grant U54AI-065357 to the Rocky Mountain Regional Center of Excellence in Biodefense and Emerging Infectious Disease Research. We thank Brad Biggerstaff, Rebecca Clark, and Barb Andre for help with statistical analyses.

References (69)

  • M. Lobigs et al.

    Location of a neutralization determinant in the E protein of yellow fever virus (17D vaccine strain)

    Virology

    (1987)
  • M. Martin et al.

    Fever and multisystem organ failure associated with 17D-204 yellow fever vaccination: a report of four cases

    Lancet

    (2001)
  • T.P. Monath

    Treatment of yellow fever

    Antivir. Res.

    (2008)
  • T.C. Pierson et al.

    Structural insights into the mechanisms of antibody-mediated neutralization of flavivirus infection: implications for vaccine development

    Cell Host Microbe

    (2008)
  • T.C. Pierson et al.

    Flaviviruses: braking the entering

    Curr. Opin. Virol.

    (2013)
  • J.T. Roehrig

    Antigenic structure of flavivirus proteins

    Adv. Virus Res.

    (2003)
  • J.T. Roehrig et al.

    Antigenic analysis of the surface glycoproteins of a Venezuelan equine encephalomyelitis virus (TC-83) using monoclonal antibodies

    Virology

    (1982)
  • K.D. Ryman et al.

    Mutation in a 17D-204 vaccine substrain-specific envelope protein epitope alters the pathogenesis of yellow fever virus in mice

    Virology

    (1998)
  • A. Sabchareon et al.

    Protective efficacy of the recombinant, live-attenuated, CYD tetravalent dengue vaccine in Thai schoolchildren: a randomised, controlled phase 2b trial

    Lancet

    (2012)
  • B.A. Thibodeaux et al.

    A small animal peripheral challenge model of yellow fever using interferon-receptor deficient mice and the 17D-204 vaccine strain

    Vaccine

    (2012)
  • B.A. Thibodeaux et al.

    A humanized IgG but not IgM antibody is effective in prophylaxis and therapy of yellow fever infection in an AG129/17D-204 peripheral challenge mouse model

    Antivir. Res.

    (2012)
  • P.F.C. Vasconcelos et al.

    Serious adverse events associated with yellow fever 17DD vaccine in Brazil: a report of two cases

    Lancet

    (2001)
  • K.L. Williams et al.

    Antibodies targeting dengue virus envelope domain III are not required for serotype-specific protection or prevention of enhancement in vivo

    Virology

    (2012)
  • S. Zhang et al.

    Role of BC loop residues in structure, function and antigenicity of the West Nile virus envelope protein receptor-binding domain III

    Virology

    (2010)
  • S.K. Austin et al.

    Structural basis of differential neutralization of DENV-1 genotypes by an antibody that recognizes a cryptic epitope

    PLoS Pathog.

    (2012)
  • J.H. Beigel et al.

    Safety and pharmacokinetics of single intravenous dose of MGAWN1, a novel monoclonal antibody to West Nile virus

    Antimicrob. Agents Chemother.

    (2010)
  • M.W. Brandriss et al.

    Lethal 17D yellow fever encephalitis in mice. I. Passive protection by monoclonal antibodies to the envelope proteins of 17D yellow fever and dengue 2 viruses

    J. General Virol.

    (1986)
  • A. Buckley et al.

    Neutralization of yellow fever virus studied using monoclonal and polyclonal antibodies

    J. General Virol.

    (1985)
  • CDC

    Fever, jaundice, and multiple organ system failure associated with 17D-derived yellow fever vaccination, 1996-2001

    MMWR. Morb. Mortal. Wkly. Rep.

    (2001)
  • T.J. Chambers et al.

    Flavivirus genome organization, expression, and replication

    Annu. Rev. Microbiol.

    (1990)
  • W.A. Collier et al.

    Changed virulence of the yellow fever virus vaccine strain 17D by a single mouse passage

    Trop. Geogr. Med.

    (1959)
  • W.D. Crill et al.

    Monoclonal antibodies that bind to domain III of dengue virus E glycoprotein are the most efficient blockers of virus adsorption to vero cells

    J. Virol.

    (2001)
  • K.A. Dowd et al.

    A dynamic landscape for antibody binding modulates antibody-mediated neutralization of West Nile virus

    PLoS Pathog.

    (2011)
  • M.J. Engle et al.

    Antibody prophylaxis and therapy against West Nile virus infection in wild-type and immunodeficient mice

    J. Virol.

    (2003)
  • Cited by (0)

    View full text