Title of Invention

"AN ISOLATED MUTANT FLAVIVIRUS E-GLYCOPROTEIN POLYPEPTIDE, NUCLEIC ACID MOLECULE ENCODING SAID POLYPEPTIDE AND ITS VIRUS-LIKE PARTICLE"

Abstract An isolated mutant flavivirus E-glycoprotein polypeptide that exhibits measurably reduced antibody cross-reactivity as compared to the corresponding wild-type flavivirus E-glycoprotein, said mutant polypeptide comprising at least one amino acid substitution at position 104, 106, or 107 as compared to said corresponding wild-type flavivirus E-glycoprotein polypeptide.
Full Text PRIORITY CLAIM
This application claims the benefit of U.S. Provisional Patent Application No. 60/591,898 filed July 27, 2004, which is incorporated herein by reference in its entirety.
STATEMENT OF GOVERNMENT SUPPORT
This invention was made by the Centers for Disease Control and Prevention, an agency of the United States Government. Therefore, the U.S. Government has certain rights in this invention.
FIELD
This disclosure relates to a structure-based rational mutagenesis method for identifying flavivirus envelope (E)-glycoprotein cross-reactive epitopes. The disclosure further relates to flavivirus E-glycoprotein cross-reactive epitopes and mutants thereof having reduced or ablated cross-reactivity. Flavivirus cross-reactive E-glycoprotein epitopes with reduced or ablated cross-reactivity are useful in the diagnosis, inhibition and treatment of diseases caused by flaviviruses.
BACKGROUND
The Flaviviridae are a diverse family of enveloped viruses infecting both arthropods and vertebrates. Flaviviruses have a positive-sense single-stranded RNA genome 10.7 kb in length, transcribed into a single polyprotein precursor encoding three structural proteins, capsid, premembrane (prM), envelope (E), and seven non-structural proteins (Lindenbach & Rice, Flaviviridae: the viruses and their replication. In Fields Virology, 4th ed., Knipe and Howley. Eds., Philadelphia, Lippincott Williams & Wilkins, pp. 991-1041, 2001; Rice et al, Science 229:726-33, 1985). The flavivirus E-glycoprotein is the primary antigen, inducing protective immunity; it is essential for membrane fusion, and mediates binding to cellular receptors (Allison et al., J. Virol. 75:4268-75, 2001; Crill & Roehrig, J. Virol. 75:7769-73, 2001; Rey et al, Nature 375:291-98, 1995). Flavivirus E-glycoprotein therefore directly affects host range, tissue tropism, and the virulence of these viruses.
The flavivirus E-glycoprotein contains three structural and functional domains. Domain I (DI) is an 8-stranded (3-barrel containing two large insertion loops that form the elongated finger-like domain II (DII) (Rey et al, Nature 375:291-98, 1995). DII is involved in stabilizing the E-glycoprotein dimer and contains the internal fusion peptide (Allison et al., J. Virol. 75:4268-75, 2001). Domain III (Dili) forms a ten-stranded P-barrel with an immunoglobulin-like fold and contains the cellular receptor-binding motifs (Crill & Roehrig, J. Virol. 75:7769-73, 2001; Modis et al, PNAS 100:6986-91, 2003). DI and Dili contain predominately type-specific and subcomplex-reactive epitopes, whereas DII contains the major flavivirus group- and subgroup-cross-reactive
epitopes, which are sensitive to reduction and denaturation and are formed from discontinuous amino
acid sequences (Mandl et al, J. Virol. 63:564-71, 1989; Key et al, Nature 375:291-98, 1995;
Roehrig et al., Virology 246:317-28, 1998).
Members of the Flaviviridae family that infect humans frequently cause severe morbidity
and mortality, and epidemics of flaviviruses continue to be a major public health concern worldwide.
More than two billion people are at risk of being infected with members of the genus Flavivirus which
includes at least 70 distinct virus species (Burke & Monath, Flaviviruses. In Fields Virology, 4th ed.,
Knipe and Howley. Eds., Philadelphia, Lippincott Williams & Wilkins, pp. 1043-1125, 2001; Kuno et
al.,J. Virol. 72:73-83, 1998; Solomon & Mallewa, J. Infect. 42:104-15, 2001). The medically
important flaviviruses include yellow fever (YF) virus in Africa, Latin and South America; Japanese
encephalitis (JE) virus in Asia and Australia; West Nile (WN) virus in Africa, Central Europe, and
most recently in North America; tick-borne encephalitis (TBE) complex viruses in the temperate
regions of Europe, North America and Asia; and the four serotypes of dengue viruses (DEN-1, -2, -3,
and -4) in tropical and subtropical regions of the world (Lindenbach & Rice, Flaviviridae: the viruses
and their replication. In Fields Virology, 4th ed., Knipe and Howley. Eds., Philadelphia, Lippincott
Williams & Wilkins, pp. 991-1041, 2001).
Human infection by flaviviruses results in a humoral immune response involving virus
species-specific as well as flavivirus cross-reactive antibodies (Calisher et al.,J. Gen. Virol. 70:37-43,
1989; Tesh etal., Emerg. Inf. Dis. 8:245-51, 2002). The presence of flavivirus cross-reactive
antibodies in human sera produces two public health concerns upon secondary infection with a
heterologous flavivirus. Serodiagnosis of secondary flavivirus infections, especially in areas with
multiple co-circulating flaviviruses, can be particularly difficult due to the inability to differentiate
primary from secondary cross-reactive serum antibodies using currently available viral antigens.
Therefore, definitive epidemiological information either cannot be obtained or is delayed to the point
that effective control and prevention strategies may be delayed. Additionally, the presence of subneutralizing
levels of flavivirus cross-reactive serum antibodies may result in increasing the severity
of secondary flavivirus infections due to antibody-dependant enhancement (ADE), in particular,
following secondary dengue virus infection (Ferguson et al.,PNAS 96:790-94, 1999; Halstead, Rev.
Infect. Dis. 11:830-39, 1989; Takada & Kawaoka, Rev. Med. Virol. 13:387-98, 2003; Wallace et al.,
J. Gen Virol. 84:1723-28, 2003). Thus, there exists a need for a method for identifying and
characterizing flavivirus cross-reactive epitopes for improved flavivirus serodiagnosis and
development of flavivirus vaccines.
SUMMARY OF THE DISCLOSURE
Multiple flavivirus E-glycoprotein cross-reactive epitopes and mutant E-glycoprotein
polypeptides thereof exhibiting reduced or ablated cross-reactivity have been identified. In various
embodiments, these E-glycoprotein polypeptides with reduced or ablated cross-reactivity are capable
of eliciting effective type-specific immune responses against flaviviruses. In one example, the
identified cross-reactive epitopes incorporate the highly conserved Gly104, Glyioe, and Leuio? residues.
In another example, the identified cross-reactive epitope centers on the strictly conserved Trp23l
residue and its structurally related neighbors Glui26 and Thr226.
Also described herein are recombinant flavivirus E-glycoprotein constructs that can be used
directly or indirectly to stimulate flavivirus type-specific antibodies. These constructs are designed to
elicit T-cell, B-cell, or both T-cell and B-cell responses against flavivirus type-specific epitopes. The
constructs, when integrated into a vector, can be used as immunogens, can be used as DNA vaccines,
and can be used as sources of recombinant protein for stimulation of immune responses in subjects, as
well as for protein boosts to subjects who have received a nucleic acid construct previously. Also
provided are methods of identifying and characterizing flavivirus E-glycoprotein amino acid residues
incorporated into cross-reactive epitopes, using structure-based rational mutagenesis.
The foregoing and other features and advantages will become more apparent from the
following detailed description of several embodiments, which proceeds with reference to the
accompanying figures,
BRIEF DESCRIPTION OF THE FIGURES
Figure 1 is a diagrammatical representation of the quaternary structure of the DEN-2 virus
E-glycoprotein homodimer, top view, looking down towards the viral surface, showing the locations
of flavivirus cross-reactive epitope residues (space-filling representation). The structural and
functional domains I, II, and III are also shown.
Figure 2 is a series of diagrammatical representations of the structural locations of crossreactive
epitope residues for flavivirus cross-reactive monoclonal antibodies (mAbs) in the atomic
structure of the DEN-2 virus E-glycoprotein dimer, as well as a bar graph indicating fold reductions in
mAb reactivities assayed by indirect immuno-fluorescence assay (IFA) and/or antigen-capture ELISA
(Ag-ELISA) for mutations at these E-glycoprotein positions.
FIG. 2A is a diagrammatical representation of a portion of the atomic structure of the DEN-2
virus E-glycoprotein homodimer, showing the flavivirus group-reactive mAb 4G2 and 6B6C-1
epitope residues from the fusion peptide region of DII. The flavivirus fusion peptide comprises the
highly conserved E-glycoprotein residues 98-113, which form a surface exposed loop of hydrophobic
residues rich in glycine at the tip of DII (Key et at, Nature 375:291-98, 1995; Allison et al.,J. Virol.
75:4268-75, 2001). The view is looking downward toward the viral membrane surface at an angle of
approximately 45°, while looking in towards the fusion peptide region about 45° off of parallel to the
dimer's longitudinal axis. The molecular surfaces of DI and Dili from the alternate sub-unit are
depicted as space-filling Van der Waals surfaces to highlight the close fitting of the fusion peptide
into this region. Fusion peptide residues 100-108 are depicted as stick representations with the
participating amino acids labeled. Glycan moieties attached to Asn153 and Asn67 are labeled CHOI53
and CHO67, respectively.
FIG. 2B is a diagrammatical representation of a portion of the atomic structure of the DEN-2
virus E-glycoprotein homodimer, showing the flavivirus subgroup-reactive mAb 1B7-5 epitope
residues. The view and labeling are the same as in FIG. 2A. Identified residues are depicted as sticks
and labeled.
FIG. 2C is a bar graph showing fold decreases in mAb reactivities in Ag-ELISA for DEN-2
VLPs with substitutions at the listed residues. mAbs 4G2 and 6B6C-1 are flavivirus group-reactive
and 1B7-5 is flavivirus subgroup-reactive. Substitutions at G)04 and W23] produced plasmids that
were unable to secrete measurable VLP antigen into tissue culture media. Therefore, fold decreases
in mAb reactivities for these two constructs are from IFA. Wild-type plasmid did not produce an
endpoint nearly as far out in IFA as in Ag-ELISA (see Table 3), therefore the fold reductions for
substitutions at G)04 and W23i were not as great as for other constructs measured by Ag-ELISA, even
though substitutions at these two positions appeared to completely ablate mAb reactivity.
Figure 3 is a bar graph showing the percent of cross-reactive epitope residue substitutions
altering reactivities of mAbs of different cross-reactivities. The total number of SLEV and WNV
mAbs of each type is shown in the legend on the y-axis, and the number of substitutions altering these
mAbs is shown in the columns.
BRIEF DESCRIPTION OF THE APPENDICES
Appendix I contains Tables 1-13.
SEQUENCE LISTING
The nucleic and amino acid sequences listed in the accompanying sequence listing are
shown using standard letter abbreviations for nucleotide bases, and three letter code for amino acids,
as defined in 37 C.F.R. 1.822. Only one strand of each nucleic acid sequence is shown, but the
complementary strand is understood as included by any reference to the displayed strand. In the
accompanying sequence listing:
SEQ ID NOs: 1-12 show the nucleic acid sequences of mutagenic primers used to generate
the K64N mutation, T76M mutation, Q77R mutation, G104H mutation, G,06Q mutation, L107K mutation,
E,26A mutation, T226N mutation, W23|F mutation, W23)L mutation, H244R mutation, and K247R
mutation, respectively, in the pCB8D2-2J-2-9-l DEN-2 prM/E expression plasmid.
SEQ ID NOs: 13 and 14 show the nucleic and amino acid sequences of a recombinant DEN-
2 virus E-glycoprotein antigen.
SEQ ID NOs: 15 and 16 show the nucleic and amino acid sequences of a recombinant DEN-
2 virus E-glycoprotein antigen incorporating the G104H substitution.
SEQ ID NOs: 17 and 18 show the nucleic and amino acid sequences of a recombinant DEN-
2 virus E-glycoprotein antigen incorporating the Gjo6Q substitution.
SEQ ID NOs: 19 and 20 show the nucleic and amino acid sequences of a recombinant DEN-
2 virus E-glycoprotein antigen incorporating the Li0?K substitution.
SEQ ID NOs: 21 and 22 show the nucleic and amino acid sequences of a recombinant DEN-
2 virus E-glycoprotein antigen incorporating the E126A substitution.
SEQ ID NOs: 23 and 24 show the nucleic and amino acid sequences of a recombinant DEN-
2 virus E-glycoprotein antigen incorporating the T226N substitution.
SEQ ID NOs: 25 and 26 show the nucleic and amino acid sequences of a recombinant DEN-
2 virus E-glycoprotein antigen incorporating the W23iF substitution.
SEQ ID NOs: 27 and 28 show the nucleic and amino acid sequences of a recombinant DEN-
2 virus E-glycoprotein antigen incorporating the W23iL substitution.
SEQ ID NOs: 29 and 30 show the nucleic and amino acid sequences of a recombinant DEN-
2 virus E-glycoprotein antigen incorporating the double E)26A/ T226N substitution.
SEQ ID NOs: 31-79 show the nucleic acid sequences of mutagenic primers used to generate
site-specific mutations into the SLEV and WNV E genes.
SEQ ID NOs: 80 and 81 show the nucleic and amino acid sequences of a recombinant SLEV
virus E-glycoprotein antigen.
SEQ ID NOs: 82 and 83 show the nucleic and amino acid sequences of a recombinant SLEV
virus E-glycoprotein antigen incorporating the G|06Q substitution.
SEQ ID NOs: 84 and 85 show the nucleic and amino acid sequences of a recombinant WNV
virus E-glycoprotein antigen.
SEQ ID NOs: 86 and 87 show the nucleic and amino acid sequences of a recombinant WNV
virus E-glycoprotein antigen incorporating the GmV substitution.
DETAILED DESCRIPTION
AbbreviationsADE antibody-dependant enhancementAg-ELISA antigen-capture ELISAD domainDEN dengue
DENY dengue virus
E envelope
ELISA enzyme-linked immunoabsorbent assay
IFA indirect immuno-fluorescence assay
JE Japanese encephalitis
JEV Japanese encephalitis virus
mAb monoclonal antibody
MHIAF murine hyper-immune ascetic fluid
MVEV Murray Valley encephalitis virus
PCR polymerase chain reaction
prM premembrane
SLE St. Louis encephalitis
SLEV St. Louis encephalitis virus
TBE tick-borne encephalitis
VLP virus-like particle
WN West Nile
WNV West Nile virus
YF yellow fever
//. Terms
Unless otherwise noted, technical terms are used according to conventional usage.
Definitions of common terms in molecular biology may be found in Benjamin Lewin, Genes VII,
published by Oxford University Press, 2000 (ISBN 019879276X); Kendrew et al. (eds.), The
Encyclopedia of Molecular Biology, published by Blackwell Publishers, 1994 (ISBN 0632021829);
and Robert A. Meyers (ed.), Molecular Biology and Biotechnology: a Comprehensive Desk
Reference, published by Wiley, John & Sons, Inc., 1995 (ISBN 0471186341); and other similar
references.
As used herein, the singular terms "a," "an," and "the" include plural referents unless context
clearly indicates otherwise. Similarly, the word "or" is intended to include "and" unless the context
clearly indicates otherwise. Also, as used herein, the term "comprises" means "includes." Hence
"comprising A or B" means including A, B, or A and B. It is further to be understood that all base
sizes or amino acid sizes, and all molecular weight or molecular mass values, given for nucleic acids
or polypeptides are approximate, and are provided for description. Although methods and materials
similar or equivalent to those described herein can be used in the practice or testing of the present
invention, suitable methods and materials are described below. All publications, patent applications,
patents, and other references mentioned herein are incorporated by reference in their entirety. In case
of conflict, the present specification, including explanations of terms, will control. The materials,
methods, and examples are illustrative only and not intended to be limiting.
In order to facilitate review of the various embodiments of this disclosure, the following
explanations of specific terms are provided:
Animal: Living multi-cellular vertebrate organisms, a category that includes, for example,
mammals and birds. The term mammal includes both human and non-human mammals. Similarly,
the term "subject" includes both human and veterinary subjects, for example, humans, non-human
primates, dogs, cats, horses, and cows.
Antibody: A protein (or protein complex) that includes one or more polypeptides
substantially encoded by immunoglobulin genes or fragments of immunoglobulin genes. The
recognized immunoglobulin genes include the kappa, lambda, alpha, gamma, delta, epsilon, and mu
constant region genes, as well as the myriad immunoglobulin variable region genes. Light chains are
classified as either kappa or lambda. Heavy chains are classified as gamma, mu, alpha, delta, or
epsilon, which in turn define the immunoglobulin classes, IgG, IgM, IgA, IgD and IgE, respectively.
The basic immunoglobulin (antibody) structural unit is generally a tetramer. Each tetramer
is composed of two identical pairs of polypeptide chains, each pair having one "light" (about 25 kDa)
and one "heavy" (about 50-70 kDa) chain. The N-terminus of each chain defines a variable region of
about 100 to 110 or more amino acids primarily responsible for antigen recognition. The terms
"variable light chain" (VL) and "variable heavy chain" (VH) refer, respectively, to these light andheavy chains.
As used herein, the term "antibody" includes intact immunoglobulins as well as a number of
well-characterized fragments. For instance, Fabs, Fvs, and single-chain Fvs (SCFvs) that bind to
target protein (or epitope within a protein or fusion protein) would also be specific binding agents for
that protein (or epitope). These antibody fragments are as follows: (1) Fab, the fragment which
contains a monovalent antigen-binding fragment of an antibody molecule produced by digestion of
whole antibody with the enzyme papain to yield an intact light chain and a portion of one heavy chain;
(2) Fab', the fragment of an antibody molecule obtained by treating whole antibody with pepsin,
followed by reduction, to yield an intact light chain and a portion of the heavy chain; two Fab'
fragments are obtained per antibody molecule; (3) (Fab')2, the fragment of the antibody obtained by
treating whole antibody with the enzyme pepsin without subsequent reduction; (4) F(ab')2, a dimer of
two Fab' fragments held together by two disulfide bonds; (5) Fv, a genetically engineered fragment
containing the variable region of the light chain and the variable region of the heavy chain expressed
as two chains; and (6) single chain antibody, a genetically engineered molecule containing the
variable region of the light chain, the variable region of the heavy chain, linked by a suitable
polypeptide linker as a genetically fused single chain molecule. Methods of making these fragments
are routine (see, for example, Harlow and Lane, Using Antibodies: A Laboratory Manual, CSHL,
New York, 1999).
Antibodies for use in the methods and compositions of this disclosure can be monoclonal or
polyclonal. Merely by way of example, monoclonal antibodies can be prepared from murine
hybridomas according to the classical method of Kohler and Milstein (Nature 256:495-97, 1975) or
derivative methods thereof. Detailed procedures for monoclonal antibody production are described in
Harlow and Lane, Using Antibodies: A Laboratory Manual, CSHL, New York, 1999.
Antibody binding affinity: The strength of binding between a single antibody binding site
and a ligand (e.g., an antigen or epitope). The affinity of an antibody binding site X for a ligand Y is
represented by the dissociation constant (Kd), which is the concentration of Y that is required to
occupy half of the binding sites of X present in a solution. A smaller (Kd) indicates a stronger or
higher-affinity interaction between X and Y and a lower concentration of ligand is needed to occupy
the sites. In general, antibody binding affinity can be affected by the alteration, modification and/or
substitution of one or more amino acids in the epitope recognized by the antibody paratope.
In one example, antibody binding affinity is measured by end-point titration in an Ag-ELISA
assay. Antibody binding affinity is substantially lowered (or measurably reduced) by the modification
and/or substitution of one or more amino acids in the epitope recognized by the antibody paratope if
the end-point titer of a specific antibody for the modified/substituted epitope differs by at least 4-fold,
such as at least 10-fold, at least 100-fold or greater, as compared to the unaltered epitope.
Antigen: A compound, composition, or substance that can stimulate the production of
antibodies or a T-cell response in an animal, including compositions that are injected or absorbed into
an animal. An antigen reacts with the products of specific humoral or cellular immunity, including
those induced by heterologous immunogens. In one embodiment, an antigen is a flavivirus antigen.
cDNA (complementary DNA): A piece of DNA lacking internal, non-coding segments
(introns) and regulatory sequences that determine transcription. cDNA is synthesized in the
laboratory by reverse transcription from messenger RNA extracted from cells.
Epitope: An antigenic determinant. These are particular chemical groups, such as
contiguous or non-contiguous peptide sequences, on a molecule that are antigenic, that is, that elicit a
specific immune response. An antibody binds a particular antigenic epitope based on the three
dimensional structure of the antibody and the matching (or cognate) three dimensional structure of the
epitope.
A "cross-reactive epitope" is an epitope found in two or more antigens expressed by different
genes, and responsible for inducing cross-reactive antibodies. For example, a "flavivirus crossreactive
epitope" is a flavivirus epitope found in a peptide from two or more flaviviruses, and
responsible for inducing flavivirus cross-reactive antibodies.
A "substituted epitope" comprises at least one structural substitution in the epitope, such as a
substitution of one amino acid for another. In certain provided embodiments, amino acid substitutions
at probable or identified cross-reactive epitope residues are designed to reduce or ablate crossreactive
antibody recognition without substantially altering E-glycoprotein structural conformation oraffecting type-specific antibody binding sites, disrupting dimer interactions, or impairing particle
formation, maturation, or secretion.
Flavivirus cross-reactive antibody: An antibody that recognizes (that is, specifically binds
to) an epitope found on a peptide from more than one species of flavivirus. Flavivirus cross-reactive
antibodies are classified as either complex cross-reactive or group cross-reactive antibodies.
Complex cross-reactive antibodies recognize epitopes shared by all viruses within a complex, such as
the JE virus complex or the DEN virus complex. Group cross-reactive antibodies recognize epitopes
shared by all members of the genus Flavivirus.
Antibody cross-reactivity is further refined within the sub-complex and sub-group crossreactive
categories. Sub-complex cross-reactive antibodies recognize epitopes shared by most, but
not all, members of a particular flavivirus complex (e.g., DENV-1, -2, and -3, but not DENV-4),
while sub-group cross-reactive antibodies recognize epitopes shared by flaviviruses from several
complexes, but not all members of the flavivirus group (e.g., all members of the DEN virus and JE
virus complexes, but not all members of the tick-borne virus complex). Specific, non-limiting
examples of flavivirus cross-reactive antibodies include the group cross-reactive mAbs 4G2 and
6B6C-1, the sub-group cross-reactive mAb 1B7-5, and the sub-complex cross-reactive mAb 10A1D-
2.
Flavivirus E-glycoprotein: A structural envelope protein that mediates binding of
flavivirus virions to cellular receptors on host cells. The flavivirus E-glycoprotein is required for
membrane fusion, and is the primary antigen inducing protective immunity to flavivirus infection.
Flavivirus E-glycoprotein affects host range, tissue tropism and viral virulence. The flavivirus Eglycoprotein
contains three structural and functional domains, DI-DIII. In mature virus particles the
E-glycoprotein forms head to tail homodimers lying flat and forming a dense lattice on the viral
surface.
Flavivirus E-glycoprotein domain: A domain of a protein is a part of a protein that shares
common structural, physiochemical and/or functional features; for example hydrophobic, polar,
globular, helical domains or properties, for example a DNA binding domain, an ATP binding domain,
and the like. The flavivirus E-glycoprotein contains three recognized structural and functional
domains, DI-DIII. DI is an 8-stranded P-barrel containing two large insertion loops that form the
elongated finger-like DII. DII is involved in stabilizing the E-glycoprotein dimer and contains the
internal fusion peptide that mediates flaviviral entry into host cells via membrane fusion. Dili forms a
ten-stranded p-barrel with an immunoglobulin-like fold and contains the cellular receptor-binding
motifs. DI and Dili contain predominately type- and subtype-specific epitopes, whereas DII contains
the major flavivirus group and subgroup cross-reactive epitopes, which are sensitive to reduction and
denaturation and are therefore believed to be formed from discontinuous amino acid sequences.
Flavivirus type-specific antibody: An antibody that recognizes (that is, specifically binds
to) an epitope found on a peptide from only.one specific member of the flaviviruses. Specific, nonlimiting
examples of flavivirus type-specific antibodies include: DI mAb 9A4D-1, DII mAb 1A5D-1,
and Dili mAbs 3H5, 9A3D-8 and 9D12, which only recognize epitopes found in the DENV-2 Eglycoprotein.
Hybridization: Oligonucleotides and their analogs hybridize by hydrogen bonding, which
includes Watson-Crick, Hoogsteen or reversed Hoogsteen hydrogen bonding, between
complementary bases. Generally, nucleic acid consists of nitrogenous bases that are either
pyrimidines (cytosine (C), uracil (U), and thymine (T)) or purines (adenine (A) and guanine (G)).
These nitrogenous bases form hydrogen bonds between a pyrimidine and a purine, and the bonding of
the pyrimidine to the purine is referred to as "base pairing." More specifically, A will hydrogen bond
to T or U, and G will bond to C. "Complementary" refers to the base pairing that occurs between to
distinct nucleic acid sequences or two distinct regions of the same nucleic acid sequence.
"Specifically hybridizable" and "specifically complementary" are terms that indicate a
sufficient degree of complementarity such that stable and specific binding occurs between the
oligonucleotide (or its analog) and the DNA or UNA target. The oligonucleotide or oligonucleotide
analog need not be 100% complementary to its target sequence to be specifically hybridizable. An
oligonucleotide or analog is specifically hybridizable when binding of the oligonucleotide or analog
10to the target DNA or RNA molecule interferes with the normal Function of the target DNA or RNA,
and there is a sufficient degree of complementarity to avoid non-specific binding of the
oligonucleotide or analog to non-target sequences under conditions where specific binding is desired,
for example under physiological conditions in the case of in vivo assays or systems. Such binding is
referred to as specific hybridization.
Hybridization conditions resulting in particular degrees of stringency will vary depending
upon the nature of the hybridization method of choice and the composition and length of the
hybridizing nucleic acid sequences. Generally, the temperature of hybridization and the ionic strength
(especially the Na+ and/or Mg++ concentration) of the hybridization buffer will determine the
stringency of hybridization, though wash times also influence stringency. Calculations regarding
hybridization conditions required for attaining particular degrees of stringency are discussed by
Sambrook et al. (ed.), Molecular Cloning: A Laboratory Manual, 2nd ed., vol. 1-3, Cold Spring
Harbor Laboratory Press, Cold Spring Harbor, NY, 1989, chapters 9 and 11; and Ausubel et al. Short
Protocols in Molecular Biology, 4th ed., John Wiley & Sons, Inc., 1999.
For purposes of the present disclosure, "stringent conditions" encompass conditions under
which hybridization will only occur if there is less than 25% mismatch between the hybridization
molecule and the target sequence. "Stringent conditions" may be broken down into particular levels
of stringency for more precise definition. Thus, as used herein, "moderate stringency" conditions are
those under which molecules with more than 25% sequence mismatch will not hybridize; conditions
of "medium stringency" are those under which molecules with more than 15% mismatch will not
hybridize, and conditions of "high stringency" are those under which sequences with more than 10%
mismatch will not hybridize. Conditions of "very high stringency" are those under which sequences
with more than 6% mismatch will not hybridize.
"Specific hybridization" refers to the binding, duplexing, or hybridizing of a molecule only
or substantially only to a particular nucleotide sequence when that sequence is present in a complex
mixture (for example, total cellular DNA or RNA). Specific hybridization may also occur under
conditions of varying stringency.
Immune stimulatory composition: A term used herein to mean a composition useful for
stimulating or eliciting a specific immune response (or immunogenic response) in a vertebrate. The
immune stimulatory composition can be a protein antigen or a plasmid vector used to express a
protein antigen. In some embodiments, the immunogenic response is protective or provides
protective immunity, in that it enables the vertebrate animal to better resist infection with or disease
progression from the organism against which the immune stimulatory composition is directed.
Without wishing to be bound by a specific theory, it is believed that an immunogenic
response induced by an immune stimulatory composition may arise from the generation of an
antibody specific to one or more of the epitopes provided in the immune stimulatory composition.
Alternatively, the response may comprise a T-helper or cytotoxic cell-based response to one or more
of the epitopes provided in the immune stimulatory composition. All three of these responses may
originate from nai've or memory cells. One specific example of a type of immune stimulatory
composition is a vaccine.
In some embodiments, an "effective amount" or "immune-stimulatory amount" of an immune
stimulatory composition is an amount which, when administered to a subject, is sufficient to engender
a detectable immune response. Such a response may comprise, for instance, generation of an
antibody specific to one or more of the epitopes provided in the immune stimulatory composition.
Alternatively, the response may comprise a T-helper or CTL-based response to one or more of the
epitopes provided in the immune stimulatory composition. All three of these responses may originate
from nai've or memory cells. In other embodiments, a "protective effective amount" of an immune
stimulatory composition is an amount which, when administered to a subject, is sufficient to confer
protective immunity upon the subject.
Inhibiting or treating a disease: Inhibiting the full development of a disease or condition,
for example, in a subject who is at risk for a disease. Specific examples of diseases include dengue
fever, dengue hemorrhagic fever, yellow fever, Japanese encephalitis, tick-borne encephalitis, and
West Nile disease. "Treatment" refers to a therapeutic intervention that ameliorates a sign or
symptom of a disease or pathological condition after it has begun to develop. As used herein, the
term "ameliorating," with reference to a disease, pathological condition or symptom, refers to any
observable beneficial effect of the treatment. The beneficial effect can be evidenced, for example, by
a delayed onset of clinical symptoms of the disease in a susceptible subject, a reduction in severity of
some or all clinical symptoms of the disease, a slower progression of the disease, a reduction in the
number of relapses of the disease, an improvement in the overall health or well-being of the subject,
or by other parameters well known in the art that are specific to the particular disease.
Isolated: An "isolated" or "purified" biological component (such as a nucleic acid, peptide,
protein, protein complex, or particle) has been substantially separated, produced apart from, or
purified away from other biological components in the cell of the organism in which the component
naturally occurs, that is, other chromosomal and extrachromosomal DNA and RNA, and proteins.
Nucleic acids, peptides and proteins that have been "isolated" or "purified" thus include nucleic acids
and proteins purified by standard purification methods. The term also embraces nucleic acids,
peptides and proteins prepared by recombinant expression in a host cell, as well as chemically
synthesized nucleic acids or proteins. The term "isolated" or "purified" does not require absolute
purity; rather, it is intended as a relative term. Thus, for example, an isolated biological component is
one in which the biological component is more enriched than the biological component is in its
natural environment within a cell, or other production vessel. Preferably, a preparation is purified
such that the biological component represents at least 50%, such as at least 70%, at least 90%, at least
95%, or greater, of the total biological component content of the preparation.
Nucleic acid molecule: A polymeric form of nucleotides, which may include both sense and
anti-sense strands of RNA, cDNA, genomic DNA, and synthetic forms and mixed polymers of the
above. A nucleotide refers to a ribonucleotide, deoxynucleotide or a modified form of either type of
nucleotide. The term "nucleic acid molecule" as used herein is synonymous with "nucleic acid" and
"polynucleotide." A nucleic acid molecule is usually at least 10 bases in length, unless otherwise
specified. The term includes single- and double-stranded forms of DNA. A polynucleotide may
include either or both naturally occurring and modified nucleotides linked together by naturally
occurring and/or non-naturally occurring nucleotide linkages.
Oligonucleotide : A nucleic acid molecule generally comprising a length of 300 bases or
fewer. The term often refers to single-stranded deoxyribonucleotides, but it can refer as well to
single- or double-stranded ribonucleotides, RNA:DNA hybrids and double-stranded DNAs, among
others. The term "oligonucleotide" also includes oligonucleosides (that is, an oligonucleotide minus
the phosphate) and any other organic base polymer.
In some examples, oligonucleotides are about 10 to about 90 bases in length, for example,
12, 13, 14, 15, 16, 17, 18, 19 or 20 bases in length. Other oligonucleotides are about 25, about 30,
about 35, about 40, about 45, about 50, about 55, about 60 bases, about 65 bases, about 70 bases,
about 75 bases or about 80 bases in length. Oligonucleotides may be single-stranded, for example,
for use as probes or primers, or may be double-stranded, for example, for use in the construction of a
mutant gene. Oligonucleotides can be either sense or anti-sense oligonucleotides. An oligonucleotide
can be modified as discussed above in reference to nucleic acid molecules. Oligonucleotides can be
obtained from existing nucleic acid sources (for example, genomic or cDNA), but can also be
synthetic (for example, produced by laboratory or in vitro oligonucleotide synthesis).
Operably linked: A first nucleic acid sequence is operably linked with a second nucleic
acid sequence when the first nucleic acid sequence is placed in a functional relationship with the
second nucleic acid sequence. For instance, a promoter is operably linked to a coding sequence is the
promoter affects the transcription or expression of the coding sequence. Generally, operably linked
DNA sequences are contiguous and, where necessary to join two protein coding regions, in the same
reading frame. If introns are present, the operably linked DNA sequences may not be contiguous.
Paratope: That portion of an antibody that is responsible for its binding to an antigenic
determinant (epitope) on an antigen.
Polypeptide: A polymer in which the monomers are amino acid residues joined together
through amide bonds. When the amino acids are alpha-amino acids, either the L-optical isomer or the
D-optical isomer can be used, the L-isomers being preferred for many biological uses. The terms
"polypeptide" or "protein" as used herein are intended to encompass any amino acid molecule and
include modified amino acid molecules such as glycoproteins. The term "polypeptide" is specifically
intended to cover naturally occurring proteins, as well as those which are recombinantly or
synthetically produced.
Probes and primers: A probe comprises an isolated nucleic acid molecule attached to a
detectable label or other reporter molecule. Typical labels include radioactive isotopes, enzyme
substrates, co-factors, ligands, chemiluminescent or fluorescent agents, haptens, and enzymes.
Methods for labeling and guidance in the choice of labels appropriate for various purposes are
discussed, for example, in Sambrook et al. (ed.), Molecular Cloning: A Laboratory Manual, 2nd ed.,
vol. 1-3, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY, 1989 and Ausubel et al.
Short Protocols in Molecular Biology, 4th ed., John Wiley & Sons, Inc., 1999.
Primers are short nucleic acid molecules, for instance DNA oligonucleotides 6 nucleotides or
more in length, for example that hybridize to contiguous complementary nucleotides or a sequence to
be amplified. Longer DNA oligonucleotides may be about 10, 12, 15, 20, 25, 30, or 50 nucleotides or
more in length. Primers can be annealed to a complementary target DNA strand by nucleic acid
hybridization to form a hybrid between the primer and the target DNA strand, and then the primer
extended along the target DNA strand by a DNA polymerase enzyme. Primer pairs can be used for
amplification of a nucleic acid sequence, for example, by the polymerase chain reaction (PCR) or
other nucleic-acid amplification methods known in the art. Other examples of amplification include
strand displacement amplification, as disclosed in U.S. Patent No. 5,744,311; transcription-free
isothermal amplification, as disclosed in U.S. Patent No. 6,033,881; repair chain reaction
amplification, as disclosed in WO 90/01069; ligase chain reaction amplification, as disclosed in EPA-
320 308; gap filling ligase chain reaction amplification, as disclosed in 5,427,930; and NASBA™
RNA transcription-free amplification, as disclosed in U.S. Patent No. 6,025,134.
Methods for preparing and using nucleic acid probes and primers are described, for example,
in Sambrook et al. (ed.), Molecular Cloning: A Laboratory Manual, 2nd ed., vol. 1-3, Cold Spring
Harbor Laboratory Press, Cold Spring Harbor, NY, 1989; Ausubel et al. Short Protocols in
Molecular Biology, 4* ed., John Wiley & Sons, Inc., 1999; and Innis et al. PCR Protocols, A Guide
to Methods and Applications, Academic Press, Inc., San Diego, CA, 1990. Amplification primer
pairs can be derived from a known sequence, for example, by using computer programs intended for
that purpose such as Primer (Version 0.5, © 199J, Whitehead Institute for Biomedical Research,
Cambridge, MA). One of ordinary skill in the art will appreciate that the specificity of a particular
probe or primer increases with its length. Thus, in order to obtain greater specificity, probes and
primers can be selected that comprise at least 20, 25, 30, 35, 40, 45, 50 or more consecutive
nucleotides of a target nucleotide sequences.
Recombinant nucleic acid: A nucleic acid molecule that is not naturally occurring or has a
sequence that is made by an artificial combination of two otherwise separated segments of sequence.
This artificial combination is accomplished by chemical synthesis or, more commonly, by the
artificial manipulation of isolated segments of nucleic acids, e.g., by genetic engineering techniques
such as those described in Sambrook et al. (ed.), Molecular Cloning: A Laboratory Manual, 2nd ed.,
vol. 1-3, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY, 1989. The term
recombinant includes nucleic acids that have been altered solely by addition, substitution, or deletion
of a portion of a natural nucleic acid molecule.
Regulatory sequences or elements: These terms refer generally to a class of DNA
sequences that influence or control expression of genes. Included in the term are promoters,
enhancers, locus control regions (LCRs), insulators/boundary elements, silencers, matrix attachment
regions (MARs, also referred to as scaffold attachment regions), represser, transcriptional
terminators, origins of replication, centromeres, and meiotic recombination hotspots. Promoters are
sequences of DNA near the 5'-end of a gene that act as a binding site for DNA-dependent UNA
polymerase, and from which transcription is initiated. Enhancers are control elements that elevate the
level of transcription from a promoter, usually independently of the enhancer's orientation or distance
from the promoter. LCRs confer tissue-specific and temporally regulated expression to genes to
which they are linked. LCRs function independently of their position in relation to the gene, but are
copy-number dependent. It is believed that they function to open the nucleosome structure, so other
factors can bind to the DNA. LCRs may also affect replication timing and origin usage. Insulators
(also know as boundary elements) are DNA sequences that prevent the activation (or inactivation) of
transcription of a gene, by blocking effects of surrounding chromatin. Silencers and repressers are
control elements that suppress gene expression; they act on a gene independently of their orientation
or distance from the gene. MARs are sequences within DNA that bind to the nuclear scaffold; they
can affect transcription, possibly by separating chromosomes into regulatory domains. It is believedthat MARs mediate higher-order, looped structures within chromosomes. Transcriptional terminators
are regions within the gene vicinity where RNA Polymerase is released from the template. Origins of
replication are regions of the genome, during DNA synthesis or replication phases of cell division,
that begin the replication process of DNA. Meiotic recombination hotspots are regions of the genome
that recombine more frequently than average during meiosis.
Sequence identity: The similarity between two nucleic acid sequences, or two amino acid
sequences, is expressed in terms of the similarity between the sequences, otherwise referred to as
sequence identity. Sequence identity is frequently measured in terms of percentage identity (or
similarity or homology); the higher the percentage, the more similar the two sequences are.
Methods of alignment of sequences for comparison are well known in the art. Various
programs and alignment algorithms are described in: Smith and Waterman (Adv. Appl. Math., 2:482,
1981); Needleman and Wunsch (J. Mol. Bio!., 48:443, 1970); Pearson and Lipman (Proc. Natl. Acad.
Sci., 85:2444, 1988); Higgins and Sharp (Gene, 73:237-44, 1988); Higgins and Sharp (CABIOS,
5:151-53, 1989); Corpet et al. (Nuc. Acids Res., 16:10881-90, 1988); Huang et al. (Comp. Appls.
Biosci,, 8:155-65, 1992); and Pearson et al. (Meth. Mol. Biol, 24:307-31, 1994). Altschul et al.
(Nature Genet., 6:119-29, 1994) presents a detailed consideration of sequence alignment methods and
homology calculations.
The alignment tools ALIGN (Myers and Miller, CABIOS 4:11-17, 1989) or LFASTA
(Pearson and Lipman, 1988) may be used to perform sequence comparisons (Internet Program ©
1996, W. R. Pearson and the University of Virginia, "fasta20u63" version 2.0u63, release date
December 1996). ALIGN compares entire sequences against one another, while LFASTA compares
regions of local similarity. These alignment tools and their respective tutorials are available on the
Internet at the NCSA website. Alternatively, for comparisons of amino acid sequences of greater than
about 30 amino acids, the "Blast 2 sequences" function can be employed using the default
BLOSUM62 matrix set to default parameters, (gap existence cost of 11, and a per residue gap cost of
1). When aligning short peptides (fewer than around 30 amino acids), the alignment should be
performed using the "Blast 2 sequences" function, employing the PAM30 matrix set to default
parameters (open gap 9, extension gap 1 penalties). The BLAST sequence comparison system is
available, for instance, from the NCBI web site; see also Altschul et al.,J. Mol. Biol., 215:403-10,
1990; Gish. and States, Nature Genet., 3:266-72, 1993; Madden et al., Meth. Enzymol., 266:131-41,
1996; Altschul et al., Nucleic Acids Res., 25:3389-402, 1997; and Zhang and Madden, Genome Res.,
7:649-56, 1997.
Orthologs (equivalent to proteins of other species) of proteins are in some instances
characterized by possession of greater than 75% sequence identity counted over the full-length
alignment with the amino acid sequence of specific protein using ALIGN set to default parameters.
Proteins with even greater similarity to a reference sequence will show increasing percentage
identities when assessed by this method, such as at least 80%, at least 85%, at least 90%, at least 92%,
at least 95%, or at least 98% sequence identity. In addition, sequence identity can be compared over
the full length of one or both binding domains of the disclosed fusion proteins.
When significantly less than the entire sequence is being compared for sequence identity,
homologous sequences will typically possess at least 80% sequence identity over short windows of
10-20, and may possess sequence identities of at least 85%, at least 90%, at least 95%, or at least 99%
depending on their similarity to the reference sequence. Sequence identity over such short windows
can be determined using LFASTA; methods are described at the NCSA website. One of skill in the
art will appreciate that these sequence identity ranges are provided for guidance only; it is entirely
possible that strongly significant homologs could be obtained that fall outside of the ranges provided.
Similar homology concepts apply for nucleic acids as are described for protein. An alternative
indication that two nucleic acid molecules are closely related is that the two molecules hybridize to
each other under stringent conditions.
Nucleic acid sequences that do not show a high degree of identity may nevertheless encode
similar amino acid sequences, due to the degeneracy of the genetic code. It is understood that
changes in nucleic acid sequence can be made using this degeneracy to produce multiple nucleic acidsequences that each encode substantially the same protein.Specific binding agent: An agent that binds bstantially only to a defined target. Thus aprotein-specific binding agent binds substantially only the defined protein, or to a specific region
within the protein. As used herein, protein-specific binding agents include antibodies and other
agents that bind substantially to a specified polypeptide. The antibodies may be monoclonal or
polyclonal antibodies that are specific for the polypeptide, as well as immunologically effective
portions ("fragments") thereof.
The determination that a particular agent binds substantially only to a specific polypeptide
may readily be made by using or adapting routine procedures. Examples of suitable in vitro assays
which make use of the Western blotting procedure include IFA and Ag-ELISA, and are described in
many standard texts, including Harlow and Lane, Using Antibodies: A Laboratory Manual, CSHL,
New York, 1999.
Transformed: A "transformed" cell is a cell into which has been introduced a nucleic acid
molecule by molecular biology techniques. The term encompasses all techniques by which a nucleic
acid molecule might be introduced into such a cell, including transfection with viral vectors,
transformation with plasmid vectors, and introduction of naked DNA by electroporation, lipofection,
and particle gun acceleration.
Vector: A nucleic acid molecule as introduced into a host cell, thereby producing a
transformed host cell. A vector may include nucleic acid sequences that permit it to replicate in a host
cell, such as an origin of replication. A vector may also include one or more selectable marker genes
and other genetic elements known in the art.
///. Overview of Several Embodiments
Isolated mutant flavivirus polypeptides exhibiting measurably reduced antibody crossreactivity
(compared to corresponding wild-type polypeptides) are disclosed herein. In one
embodiment, the isolated flavivirus polypeptides are flavivirus E-glycoproteins that include an amino
acid sequence as shown in SEQ ID NO: 14, wherein at least one of the amino acids at position 104,
106, 107, 126, 226, or 231 is substituted (compared to corresponding wild-type E-glycoproteins).
Specific, non-limiting examples of the amino acid substitutions at positions 104, 106, 107, 126,226,
and 231 include: G104H (SEQ ID NO: 16), G106Q (SEQ ID NO: 18), L107K (SEQ ID NO: 20), E126A
(SEQ ID NO: 22), T226N (SEQ ID NO: 24), W23,F (SEQ ID NO: 26), and W231L (SEQ ID NO: 28).
Also disclosed are isolated nucleic acid molecules encoding the flavivirus polypeptides with at least
one amino acid substitution at position 104, 106, 107, 126, 226, or 231 of SEQ ID NO: 14.
Representative nucleic acid molecules are shown in SEQ ID NOs: 15, 17, 19, 21, 23, 25, and 27.
In another embodiment, the isolated flavivirus polypeptides are flavivirus E-glycoproteins
that include an amino acid sequence as shown in SEQ ID NO: 81, wherein at least one of the amino
acids at position 106 is substituted (compared to corresponding wild-type E-glycoproteins). Specific,
non-limiting examples of the amino acid substitutions at position 106 include: G^Q (SEQ ID NO:
83). Also disclosed are isolated nucleic acid molecules encoding the flavivirus polypeptides with at
least one amino acid substitution at position 106 of SEQ ID NO: 81. A representative nucleic acid
molecule is shown in SEQ ID NO: 82.
In yet another embodiment, the isolated flavivirus polypeptides are flavivirus Eglycoproteins
that include an amino acid sequence as shown in SEQ ID NO: 85, wherein at least one
of the amino acids at position 106 is substituted (compared to corresponding wild-type Eglycoproteins).
Specific, non-limiting examples of the amino acid substitutions at position 106
include: GioeV (SEQ ID NO: 87). Also disclosed are isolated nucleic acid molecules encoding the
flavivirus polypeptides with at least one amino acid substitution at position 106 of SEQ ID NO: 85. A
representative nucleic acid molecule is shown in SEQ ID NO: 86.
Pharmaceutical and immune stimulatory compositions are also disclosed that include one or
more flavivirus E-glycoprotein polypeptides exhibiting measurably reduced antibody cross-reactivity,
with at least one amino acid substitution at position 104, 106, 107, 126, 226, or 231 of SEQ ID NO:
14. Also disclosed are pharmaceutical and immune stimulatory compositions that include one or
more nucleic acid molecules encoding the flavivirus polypeptides with at least one amino acid
substitution at position 104, 106, 107, 126, 226, or 231 of SEQ ID NO: 14. Representative nucleic
acid molecules are shown in SEQ ID NOs: 15, 17, 19, 21, 23, 25, and 27.
Also disclosed are pharmaceutical and immune stimulatory compositions that include one or
more flavivirus E-glycoprotein polypeptides exhibiting measurably reduced antibody cross-reactivity,
with at least one amino acid substitution at position 106 of SEQ ID NO: 81 or SEQ ID NO: 85. Also
disclosed are pharmaceutical and immune stimulatory compositions that include one or more nucleic
acid molecules encoding the flavivirus polypeptides with at least one ammo acid substitution at
position 106 of SEQ ID NO: 81 or SEQ ID NO: 85. Representative nucleic acid molecules are shown
in SEQ ID NOs: 82 and 86.
In another embodiment, a method is provided for identifying and modifying a flavivirus
cross-reactive epitope. This method includes selecting a candidate cross-reactive epitope using a
structure-based design approach, and designing a substituted epitope including at least one amino acid
residue substitution compared to the candidate epitope. The candidate epitope is then contacted with
a specific binding agent under conditions whereby a candidate epitope/specific binding agent complex
can form. Likewise, the substituted epitope is contacted with the same specific binding agent under
the same conditions used for candidate epitope/specific binding agent complex formation. A
candidate epitope is identified as a flavivirus cross-reactive epitope when the substituted epitope has a
substantially lower binding affinity for the specific binding agent compared to the candidate epitope,
and wherein the flavivirus cross-reactive epitope binds to a specific binding agent that binds to at least
two flaviviruses. In specific, non-limiting examples, the at least two flaviviruses are selected from
dengue serotype 1 virus, dengue serotype 2 virus, dengue serotype 3 virus, dengue serotype 4 virus,
yellow fever virus, Japanese encephalitis virus, St. Louis encephalitis virus, and West Nile virus. In
yet another specific example of the provided method, the specific binding agent is a flavivirus crossreactive
antibody.
In a further specific example of the provided method, the structure-based design approach
includes identifying at least one conserved flavivirus amino acid between two or more flavivirus
groups or subgroups, and mapping the conserved flavivirus amino acid onto a structure of a flavivirus
E-glycoprotein. In another specific example, the conserved flavivirus amino acid exhibits two or
more of the following structural characteristics: it is located in DII of the E-glycoprotein, it is
conserved across the flaviviruses, it is on the outer or lateral surface of the E-glycoprotein dimer, it
has at least 35% surface accessibility potential, its side chain projection is accessible for antibody
paratopes, or it has a high p-factor.
18
In yet a further specific example of the provided method, the structure-based design
approach includes identifying at least one conserved flavivirus amino acid between two or more
flavivirus complexes or subcomplexes, and mapping the conserved flavivirus amino acid onto a
structure of a flavivirus E-glycoprotein. In still another specific example, the conserved flavivirus
amino acid exhibits two or more of the following structural characteristics: it has at least 35% surface
accessibility potential, it is on the outer or lateral surface of the E-glycoprotein dimer, it is conserved
across the flaviviruses, its side chain projection is accessible for antibody paratopes, or it has a high Pfactor.
In another embodiment, a method is provided for detecting a flavivirus antibody in a sample.
This method includes contacting the sample with the disclosed mutant flavivirus polypeptides under
conditions whereby a polypeptide/antibody complex can form, and detecting polypeptide/antibody
complex formation, thereby detecting a flavivirus antibody in a sample. Also disclosed are methods
of diagnosing a flavivirus infection in a subject. In one embodiment, the method includes contacting
a sample from the subject with the disclosed mutant flavivirus polypeptides under conditions whereby
a polypeptide/antibody complex can form, and detecting polypeptide/antibody complex formation,
thereby diagnosing a flavivirus infection in a subject.
Also disclosed is a flavivirus E-glycoprotein engineered to comprise at least one amino acid
residue substitution according to the methods described herein.
IV. Identifying Flavivirus Cross-Reactive Epitopes
The current disclosure provides methods for identifying flavivirus cross-reactive epitopes, as
well as distinguishing such epitopes from species-specific (or type-specific) epitopes.
In one embodiment, the method comprises a structure-based design approach, which
optionally includes one or more of the following requirements in order to identify cross-reactive
epitopes: 1) the epitope is located in DII of the E-glycoprotein, for example, amino acids 52-135 and
195-285 in the THE virus E-glycoprotein, 52-132 and 193-280 in the DEN-2 virus E-glycoprotein,
and conserved across the flaviviruses or multiple flaviviral species; 2) the epitope is on the outer or
lateral surface of the E-glycoprotein dimer; 3) the epitope has at least 35% surface accessibility
potential; 4) one or more side chain projections of amino acids within the epitope are accessible to
antibody paratopes; and 5) residues with high temperature (P) factors are favored.
In one embodiment, a structure-based design approach comprises a procedural algorithm
developed to localize epitopes responsible for inducing flavivirus cross-reactive antibodies. Strictlyconserved
flavivirus residues are initially identified. These residues are mapped, for example, onto a
2.0 A resolution E-glycoprotein structure for TBE virus (Rey et al., Nature 375:291-98, 1995), a high
resolution DEN-2 virus E-glycoprotein structure (Modis et al.,PNAS 100:6986-91, 2003), or other
similar structure. Optionally, strictly-conserved flavivirus residues are also mapped onto a computer
predicted homology model structure for the DEN-2 virus E-glycoprotein using, for example, the
Swiss-Pdb Viewer 3.7 structure analysis software (Guex et al., Electrophoresis 18:2714-23, 1997).
The following criteria (individually or in combination of two or more) are then employed in
certain embodiments to select probable flavivirus group or subgroup cross-reactive epitope residues:
1) an amino acid located in DII (for example, amino acids 52-135 and 195-285 in the TBE virus Eglycoprotein
(Rey et al, Nature 375:291-98, 1995); 52-132 and 193-280 in the DEN-2 virus Eglycoprotein
(Modis et al., PNAS 100:6986-91, 2003)), and conserved among more than one
flavivirus; 2) amino acids on the outer or lateral surface of the E-glycoprotein dimer; 3) amino acids
with at least 35% surface accessibility potential; 4) side chain projections accessible to antibody
paratopes; and 5) residues with high temperature (P) factors should be favored, as these residues tend
to be flexible and are able to conform to the antibody paratope, increasing the antibody-antigen (Ab-
Ag) affinity.
Similar criteria (individually or in combination of two or more) are employed in certain
embodiments to select probable flavivirus complex or subcomplex cross-reactive epitope residues.
The procedural algorithm for the identification of flavivirus complex and sub-complex cross-reactive
epitopes utilizes the following optimaliry criteria: 1) The identification and selection of amino acid
residues with >35% of their surface solvent accessible. These residues are identified from the
published atomic structure coordinates of the DENV-2 soluble ectodomain of the envelope
glycoprotein and homology models of SLEV and WNV derived from the DENV-2 structure (Modis
et al, Proc. Natl. Acad. Sci. USA 100:6986-91, 2003). In addition to examination of amino acid
residues in structural domain II, residues in domains I and III were examined, since published results
indicate that some complex and sub-complex cross-reactive epitopes are mapped onto domains I and
III in addition to domain II (Roehrig et al., Virology 246:317-28, 1998). 2) Amino acids on the outer
or lateral surface of the E-glycoprotein dimer, and accessible to antibody. 3) Amino acid
conservation across the flavivirus complex (based upon a structural alignment of the protein
sequences). Residues conserved across all member viruses of the same complex are favored. If
conserved within but not across the entire complex, then residues with shared identities between
WNV and SLEV are favored in the JEV complex, and residues with shared identities between
DENV-2 and two or more other viruses in the DENV complex are favored over those shared with
DENV-2 and only one other DENV complex virus. 4) Side chain projections exposed towards the
outer surface and accessible to antibody paratopes. 5) Residues with high temperature (P-) factors
should be favored, as these residues tend to be flexible and are able to conform to the antibody
paratope, increasing the antibody-antigen affinity. Amino acid residues with high temperature factors
are more commonly found in antigen epitopes than lower temperature factor residues. 6) Following
identification of potential individual flavivirus complex and sub-complex cross-reactive epitope
residues, all residues are mapped and highlighted on the same E-glycoprotein dimer structure
together. With this technique, groups of potential cross-reactive epitope residues forming clusters
(and hence probable epitopes) are readily identified. 7) Residues fitting all of these criteria and
occurring in structural clusters approximately 20 x 30 A2 (which is the average "footprint" of an
antibody Fab that interacts with an antigen epitope) are favored over residues that are more isolated
in the protein structure. 8) Within an identified structural cluster of potential epitope residues,
residues that more completely satisfy greater numbers of the optimality criteria are selected for the
first round of mutagenesis analysis.
A. Outer and/or Lateral Surface Amino Acids
In one embodiment, the outer and/or lateral surface of the E-glycoprotein dimer comprises
those residues which are exposed on the surface of the E-glycoprotein dimer in a way that they are
physically capable of interacting with a host-derived immunoglobulin antibody molecule. The
flavivirus virion contains a host cell-derived lipid bilayer, with E-glycoprotein dimers imbedded
within this lipid bilayer via their trans-membrane domains. The ectodomains of the E-glycoprotein
dimers lie on top of this bilayer, forming a dense lattice and essentially coating the virion in a protein
shell. Because of this structural organization, there are regions of the E-glycoprotein that, under
general assembled virion conditions, cannot physically interact with an immunoglobulin molecule,
and therefore are highly unlikely to form part of an antibody epitope. Such inaccessible regions
include the trans-membrane domains (because they are imbedded within the lipid bilayer and are
covered by the ectodomain) and more than two-thirds of the residues of the ectodomain itself, which
are either on the bottom surface of the dimer (and therefore packed between the lipid layer and the
ectodomain), or are packed into the interior of this globular protein rather than on its surface.
Because of these structural constraints, under normal conditions immunoglobulin molecules can only
interact with residues on the outer exposed surface of the E-glycoprotein dimer, and with a subset of
residues on the outer lateral surface. Because of the close packing of E-glycoprotein dimers into a
network across the surface of the virion, and the difficulty of a large immunoglobulin molecule
accessing these narrow spaces, it is believed that only some of the lateral surface residues are
available for immunoglobulin interaction. For these reasons, only residues located on the outer or
lateral surface of the E-glycoprotein are considered as participating in possible flavivirus crossreactive
epitopes. An inspection of the location of a residue (e.g., a residue conserved among more
than one flavivirus, such as Gly104, Gly]06, LeU|07, or Trp23i) in the E-glycoprotein dimer atomic
structure allows for a determination as to whether or not a residue is located on the outer or lateral
surface of the dimer.
B. Surface Accessibility Potential
In one embodiment, surface accessibility potential comprises that portion of the predicted
electron density surrounding any amino acid residue's side chain that is exposed on the surface of the
protein, and theoretically available to interact with another molecule. For any given "surface"
residue, its surface accessibility is affected by the local (and surrounding) secondary structure of the
alpha-carbon main chain, and the positions and types of immediately surrounding side-chain
projections. Thus, by definition, maximum accessibility would be for a residue X in the peptide
GGXGG in an extended conformation, as the glycine residues have no side chains and therefore
amino acid X's surface accessibility is not constrained by either the alpha-carbon backbone shape or
the surrounding residues' side chain projections (see, e.g., Li et al, Nature Struct. Bio. 10:482-88,
2003; and Faelber et al.,J. Mol. Biol. 313:83-97, 2001).
C. Accessible Side Chain Projections
In one embodiment, the side chain projection(s) accessible for antibody paratopes comprises
a qualitative assessment of how exposed and/or available a given amino acid's reactive side chain is
to interact with a hypothetical immunoglobulin molecule. The angle of projection of a side chain is
determined primarily by its position in the primary amino acid chain. However, upon folding of this
polypeptide chain, the side chain projections are additionally altered or affected by electrostatic and
other forces from surrounding residues. The accessibility of an amino acid's side chain projections to
be bound by antibody is a specific criterion that is inherent in an amino acid's "surface accessibility."
Hence, theoretical amino acid X could have 50% surface accessibility and yet its side-chain may still
be directed towards the interior of the protein and therefore be unlikely to interact energetically with
an immunoglobulin molecule (see, e.g., Li et al., Nature Struct. Bio. 10:482-88, 2003; Faelber et al.,
J. Mol. Biol. 313:83-97, 2001; and Eyal et al.,J. Comp. Chem. 25: 712-24, 2003).
D. High Temperature Factors
In one embodiment, a temperature or p-factor comprises a criterion which represents a
particular amino acid's potential flexibility within the protein. Any given atom within a protein
structure is defined by four parameters, the three x, y and z coordinates, defining its position in space,
and its (5- or temperature factor. For well defined, high-resolution crystal structures, p-values are
typically 40 A2 can be a signal that there is little confidence in
the assignment of these atoms within the protein (for example, if the protein is disordered and does
not consistently fold into the same structure). However, in well-defined atomic-level resolution
protein structures, high p-factors associated with particular atoms for individual amino acids are
typically interpreted as indicators of that residue or atom's potential flexibility. This criterion is
relevant to epitope determination, as shape complementarity of the molecular surfaces of both the
antibody paratope and the antigen epitope is know to be an important factor effecting antibody
avidity. Flexible residues, identified by their higher p-factors, are better able to make slight
positional adjustments, thereby improving shape complementarity and the energetics of the Ag-Ab
interaction. It has been demonstrated that epitope amino acids involved in antibody interactions are
more likely to have high p-factors than are amino acids from the same protein that do not interact
with antibodies (see, e.g., Mylvaganam et al.,J. Mol. Biol. 281:301-22, 1998).
Amino acid substitutions at probable cross-reactive epitope residues are modeled, selecting
substitutions that should reduce or ablate antibody recognition without altering E-glycoprotein
structural conformation, disrupting dimer interactions, or impairing particle formation, maturation, or
secretion. For this reason, cysteine residues otherwise satisfying the cross-reactive epitope criteria
are not recommended for mutagenesis because their involvement in disulphide bridging is believed to
be necessary for proper E-glycoprotein structure and function (Modis et al, PNAS 100:6986-91,
2003; Key et al., Nature 375:291-98, 1995). Stability calculations are performed for all possible
amino acid substitutions of candidate residues using, for example, the FOLD-X server (Guerois et ai,
J. Mol. Biol. 320:369-87, 2002; available on the internet) and the TBE virus E-glycoprotein pdb file
coordinates (Rey et al., Nature 375:291-98, 1995). By way of example, amino acid substitutions
modeled in the TBE virus E-glycoprotein with free energies of folding equal to or less than that of the
non-mutated wild-type E-glycoprotein are re-examined with the Swiss-PdbViewer software, to
identify those substitutions that minimized local structural disturbances while maintaining structurally
relevant biochemical interactions such as hydrogen bonding and/or charge interactions with
neighboring amino acids.
Optionally, upon the successful identification of cross-reactive epitope residues, the Eglycoprotein
structure can be further analyzed to identify additional residues forming cross-reactive
epitopes. By way of example, a "nearest neighbor" search is conducted of the surface of the Eglycoprotein
structure, looking for additional residues located within 10-15 A of the identified
residue. This distance is within the binding footprint of a single antibody paratope (Faebler et al.,J.
Mol. Biol. 313:83-97, 2001). In this second iteration of cross-reactive epitope residue identification,
the same five optimality criterion as above are used, with one change. The criterion of strict
conservation across the flaviviruses is relaxed to now include variable residues. In this way, residues
either conserved in their physiochemical nature and/or conserved only within a particular flavivirus
complex (such as the four DEN serotypes) or subgroup can be identified.
Also provided are methods for designing a substituted epitope comprising at least one amino
acid residue substitution compared to a wild-type candidate epitope; obtaining a first sample
comprising the candidate epitope; obtaining a second sample comprising the substituted epitope;
contacting the first sample with a specific binding agent; and contacting the second sample with the
specific binding agent, wherein the cross-reactive epitope is identified when the substituted epitope
has a substantially lower binding affinity for the specific binding agent compared to the candidate
epitope. Antibody binding affinities can be determined by many methods well known in the art, such
as end-point titration in an Ag-ELISA assay, competition binding in an ELISA assay, a solid-phase
radioimmunoassay, and the Biacore® surface plasmon resonance technique (Malmqvist, Biochem.
Soc. Trans. 27:335-40, 1999; and Drake et al.,Anal. Biochem. 328:35-43, 2004).
In some embodiments the specific binding agent is an antibody, for example, a polyclonal
antibody or a mAb. A specific, non-limiting example of a polyclonal antibody is polyclonal anti-
DEN-2 MHIAF. Specific, non-limiting examples of mAbs include 4G2 (ATCC No. HB-112), 6B6C-
1, 1B7-5, 10A1D-2, 1A5D-1, and 1B4C-2 (Roehrig et al, Virology 246:317-28, 1998).
V. Flavivirus Cross-Reactive Epitopes and Variants Thereof
The disclosure also provides an isolated polypeptide comprising at least one flavivirus
cross-reactive epitope residue, wherein the antibody cross-reactivity of the at least one flavivirus
cross-reactive epitope has been reduced or ablated. In one embodiment, one or more amino acid
substitutions of one or more flavivirus cross-reactive epitope residues causes the reduction or ablation
of antibody cross-reactivity. In another embodiment, the at least one flavivirus cross-reactive epitope
residue with reduced or ablated cross-reactivity has measurably lower binding affinity with one or
more flavivirus group-reactive mAbs, due to substitution of the flavivirus cross-reactive epitope
residue(s), but its binding with one or more DEN-2 virus type-specific mAbs is not affected.
Specific, non-limiting examples of an isolated polypeptide comprising at least one flavivirus
cross-reactive epitope residue with reduced or ablated cross-reactivity include, the amino acid
sequences shown in SEQ ID NO: 16 (G104H), SEQ ID NO: 18 (GmQ), SEQ ID NO: 20 (L,07K), SEQ
ID NO: 22 (E126A), SEQ ID NO: 24 (T226N), SEQ ID NO: 26 (W23iF), SEQ ID NO: 28 (W231L), SEQ
ID NO: 30 (E,26A/ T226N), SEQ ID NO: 83, and SEQ ID NO: 87.
Manipulation of the nucleotide sequence of a flavivirus cross-reactive epitope using standard
procedures, including for instance site-directed mutagenesis or PCR and M13 primer mutagenesis, can
be used to produce variants with reduced or ablated cross-reactivity. Details of these techniques are
provided in Sambrook et al. (ed.), Molecular Cloning: A Laboratory Manual, 2nd ed., vol. 1-3, Cold
Spring Harbor Laboratory Press, Cold Spring Harbor, NY, 1989. The simplest modifications involve
the substitution of one or more amino acids for amino acids having similar physiochemical and/or
structural properties. These so-called conservative substitutions are likely to have minimal impact on
the activity and/or structure of the resultant protein. Examples of conservative substitutions are shown
below.
Conservative substitutions generally maintain (a) the structure of the polypeptide backbone
in the area of the substitution, for example, as a sheet or helical conformation, (b) the charge or
hydrophobicity of the molecule at the target site, or (c) the bulk of the side chain. Substitutions that
should reduce or ablate antibody recognition without altering E-glycoprotein structural conformation,
disrupting dimer interactions, or impairing particle formation, maturation, or secretion include: Gly to
His, Gly to Gin, Leu to Lys, Glu to Ala, Thr to Asn, Trp to Phe, and Trp to Leu.
The substitutions which in general are expected to produce the greatest changes in protein
properties will be non-conservative, for instance changes in which (a) a hydrophilic residue, for
example, seryl or threonyl, is substituted for (or by) a hydrophobic residue, for example, leucyl,
isoleucyl, phenylalanyl, valyl or alanyl; (b) a cysteine or proline is substituted for (or by) any other
residue; (c) a residue having an electropositive side chain, for example, lysyl, arginyl, or histadyl, is
substituted for (or by) an electronegative residue, for example, glutamyl or aspartyl; or (d) a residue
having a bulky side chain, for example, phenylalanine, is substituted for (or by) one not having a side
chain, for example, glycine.
The disclosure also provides isolated nucleic acids that encode the described polypeptldes.
Nucleic acids of the invention thus include nucleic acids that encode: 1) polypeptides comprising at
least one flavivirus cross-reactive epitope with reduced or ablated cross-reactivity; and 2)
polypeptides that that are at least 95% identical to the polypeptides comprising at least one flavivirus
cross-reactive epitope with reduced or ablated cross-reactivity.
Recombinant nucleic acids may, for instance, contain all or part of a disclosed nucleic acid
operably linked to a regulatory sequence or element, such as a promoter, for instance, as part of a
clone designed to express a protein. Cloning and expression systems are commercially available for
such purposes and are well known in the art. The disclosure also provides cells or organisms
transformed with recombinant nucleic acid constructs that encode all or part of the described
polypeptides. Also disclosed are virus-like particles (VLPs) that include one or more of the described
flavivirus E-glycoprotein polypeptides.
VI. Specific Binding Agents
This disclosure provides specific binding agents that bind to polypeptides disclosed herein,
e.g., flavivirus E-glycoprotein polypeptides with reduced or ablated cross-reactivity. The binding
agent may be useful for identifying flavivirus cross-reactive epitopes, and for detecting and purifying
polypeptides comprising flavivirus cross-reactive epitopes. Examples of the binding agents are a
polyclonal or monoclonal antibody, and fragments thereof, that bind to polypeptides disclosed herein.
A specific, non-limiting example of a polyclonal antibody is polyclonal anti-DEN-2 MHIAF.
Specific, non-limiting examples of mAbs include 4G2, 6B6C-1, 1B7-5, 10A1D-2, 1A5D-1, and
1B4C-2.Monoclonal or polyclonal antibodies can be raised to recognize the polypeptides described
herein, or variants thereof. Optimally, antibodies raised against these polypeptides will specifically
detect the polypeptide with which the antibodies are generated. That is, antibodies raised against the
polypeptide will recognize and bind the polypeptide, and will not substantially recognize or bind to
other polypeptides or antigens. The determination that an antibody specifically binds to a target
polypeptide is made by any one of a number of standard immunoassay methods; for instance, the
Western blotting technique (Sambrook et al. (ed.), Molecular Cloning: A Laboratory Manual, 2nd ed.,
vol. 1-3, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY, 1989), Ag-ELISA and IFA.
Substantially pure flavivirus recombinant polypeptide antigens suitable for use as
immunogens can be isolated from the transformed cells described herein, using methods well known
in the art. Monoclonal or polyclonal antibodies to the antigens can then be prepared.
Monoclonal antibodies to the polypeptides can be prepared from murine hybridomas
according to the classic method of Kohler & Milstein (Nature 256:495-97, 1975), or a derivative
method thereof. Briefly, a mouse is repetitively inoculated with a few micrograms of the selected
protein immunogen (for example, a polypeptide comprising at least one flavivirus cross-reactive
epitope with reduced or ablated cross-reactivity, a portion of a polypeptide comprising at least one
flavivirus cross-reactive epitope with reduced or ablated cross-reactivity, or a synthetic peptide
comprising at least one flavivirus cross-reactive epitope with reduced or ablated cross-reactivity) over
a period of a few weeks. The mouse is then sacrificed, and the antibody-producing cells of the spleen
isolated. The spleen cells are fused by means of polyethylene glycol with mouse myeloma cells, and
the excess unfused cells destroyed by growth of the system on selective media comprising
aminopterin (HAT media). The successfully fused cells are diluted and aliquots of the dilution placed
in wells of a microtiter plate where growth of the culture is continued. Antibody-producing clones are
identified by detection of antibody in the supernatant fluid of the wells by immunoassay procedures,
such as ELISA, as originally described by Engvall (Meth. EnzymoL, 70:419-39, 1980), or a derivative
method thereof. Selected positive clones can be expanded and their monoclonal antibody product
harvested for use. Detailed procedures for monoclonal antibody production are described in Harlow
and Lane, Using Antibodies: A Laboratory Manual, CSHL, New York, 1999.
Polyclonal antiserum containing antibodies can be prepared by immunizing suitable animals
with a polypeptide comprising at least one flavivirus cross-reactive epitope with reduced or ablated
cross-reactivity, a portion of a polypeptide comprising at least one flavivirus cross-reactive epitope
with reduced or ablated cross-reactivity, or a synthetic peptide comprising at least one flavivirus
cross-reactive epitope with reduced or ablated cross-reactivity, which can be unmodified or modified,
to enhance immunogenicity.
Effective antibody production (whether monoclonal or polyclonal) is affected by many
factors related both to the antigen and the host species. For example, small molecules tend to be less
immunogenic than others and may require the use of carriers and adjuvant. Also, host animals vary in
response to site of inoculations and dose, with either inadequate or excessive doses of antigen
resulting in low liter antisera. Small doses (ng level) of antigen administered at multiple intradermal
sites appear to be most reliable. An effective immunization protocol for rabbits can be found in
Vaitukaitis et al. (J. Clin. Endocrinol. Metab., 33:988-91, 1971).
Booster injections can be given at regular intervals, and antiserum harvested when the
antibody titer thereof, as determined semi-quantitatively, for example, by double immunodiffusion in
agar against known concentrations of the antigen, begins to fall. See, for example, Ouchterlony et al.,
Handbook of Experimental Immunology, Wier, D. (ed.), Chapter 19, Blackwell, 1973. A plateau
concentration of antibody is usually in the range of 0.1 to 0.2 mg/ml of serum (about 12
26
Affinity of the antisera for the antigen is determined by preparing competitive binding curves, as
described, for example, by Fisher (Manual of Clinical Immunology, Ch. 42, 1980).
Antibody fragments may be used in place of whole antibodies and may be readily expressed
in prokaryotic host cells. Methods of making and using immunologically effective portions of
monoclonal antibodies, also referred to as "antibody fragments," are well known and include those
described in Better & Horowitz, Methods Enzymol. 178:476-96, 1989; Glockshuber et ai,
Biochemistry 29:1362-67, 1990; and U.S. Patent Nos. 5,648,237 (Expression of Functional Antibody
Fragments); 4,946,778 (Single Polypeptide Chain Binding Molecules); and 5,455,030
(Immunotherapy Using Single Chain Polypeptide Binding Molecules), and references cited therein.
Conditions whereby a polypeptide/binding agent complex can form, as well as assays for the
detection of the formation of a polypeptide/binding agent complex and quantitation of binding
affinities of the binding agent and polypeptide, are standard in the art. Such assays can include, but
are not limited to, Western blotting, immunoprecipitation, immunofluorescence,
immunocytochemistry, immunohistochemistry, fluorescence activated cell sorting (FACS),
fluorescence in situ hybridization (FISH), immunomagnetic assays, ELISA, ELISPOT (Coligan et at,
Current Protocols in Immunology, Wiley, NY, 1,995), agglutination assays, flocculation assays, cell
panning, etc., as are well known to one of skill in the art.
Binding agents of this disclosure can be bound to a substrate (for example, beads, tubes,
slides, plates, nitrocellulose sheets, etc.) or conjugated with a detectable moiety, or both bound and
conjugated. The detectable moieties contemplated for the present disclosure can include, but are not
limited to, an immunofluorescent moiety (for example, fluorescein, rhodamine), a radioactive moiety
(for example, 32P, I251,35S), an enzyme moiety (for example, horseradish peroxidase, alkaline
phosphatase), a colloidal gold moiety, and a biotin moiety. Such conjugation techniques are standard
in the art (for example, see Harlow and Lane, Using Antibodies: A Laboratory Manual, CSHL, New
York, 1999; Yang et al., Nature, 382:319-24, 1996).
VII. Detection ofFlavivirus Antibodies
The present disclosure further provides a method of detecting a flavivirus-reactive antibody
in a sample, comprising contacting the sample with a polypeptide or peptide of this disclosure under
condition whereby an antibody/polypeptide complex can form; and detecting formation of the
complex, thereby detecting flavivirus antibody in a sample.
The method of detecting flavivirus-reactive antibody in a sample can be performed, for
example, by contacting a fluid or tissue sample from a subject with a polypeptide of this disclosure
and detecting the binding of the polypeptide to the antibody. A fluid sample of this method can
comprise any biological fluid which could contain the antibody, such as cerebrospinal fluid, blood,
bile plasma, serum, saliva and urine. Other possible examples of body fluids include sputum, mucus
and the like.
Enzyme immunoassays such as IFA, ELISA and immunoblotting can be readily adapted to
accomplish the detection of flavivirus antibodies according to the methods of this disclosure. An
ELISA method effective for the detection of the antibodies can, for example, be as follows: 1) bind
the polypeptide to a substrate; 2) contact the bound polypeptide with a fluid or tissue sample
containing the antibody; 3) contact the above with a secondary antibody bound to a detectable moiety
which is reactive with the bound antibody (for example, horseradish peroxidase enzyme or alkaline
phosphatase enzyme); 4) contact the above with the substrate for the enzyme; 5) contact the above
with a color reagent; and 6) observe/measure color change or development.
Another immunologic technique that can be useful in the detection of flavivirus antibodies
uses mAbs for detection of antibodies specifically reactive with flavivirus polypeptides in a
competitive inhibition assay. Briefly, a sample is contacted with a polypeptide of this invention
which is bound to a substrate (for example, a 96-well plate). Excess sample is thoroughly washed
away. A labeled (for example, enzyme-linked, fluorescent, radioactive, etc.) mAb is then contacted
with any previously formed polypeptide-antibody complexes and the amount of mAb binding is
measured. The amount of inhibition of mAb binding is measured relative to a control (no antibody),
allowing for detection and measurement of antibody in the sample. The degree of mAb binding
inhibition can be a very specific assay for detecting a particular flavivirus variety or strain, when
based on mAb binding specificity for a particular variety or strain of flavivirus. mAbs can also be
used for direct detection of flavivirus in cells by, for example, IFA according to standard methods.
As a further example, a micro-agglutination test can be used to detect the presence of
flavivirus antibodies in a sample. Briefly, latex beads, red blood cells or other agglutinable particles
are coated with a polypeptide of this disclosure and mixed with a sample, such that antibodies in the
sample that are specifically reactive with the antigen crosslink with the antigen, causing agglutination.
The agglutinated polypeptide-antibody complexes form a precipitate, visible with the naked eye or
measurable by spectrophotometer.
In yet another example, a microsphere-based immunoassay can be used to detect the
presence of flavivirus antibodies in a sample. Briefly, microsphere beads are coated with a
polypeptide of this disclosure and mixed with a sample, such that antibodies in the sample that are
specifically reactive with the antigen bind the antigen. The bead-bound polypeptide-antibody
complexes are allowed to react with fluorescent-dye labeled anti-species antibody (such as FITClabeled
goat anti-human IgM), and are measured, using a microsphere reader (such as a Luminex
instrument).
The present disclosure further provides a method of diagnosing a flavivirus infection in a
subject, comprising contacting a sample from the subject with the polypeptide of this disclosure
under conditions whereby an antibody/polypeptide complex can form; and detecting
antibody/polypeptide complex formation, thereby diagnosing a flavivirus infection in a subject.
In examples of the diagnostic methods, the polypeptide of this disclosure can be bound to a
substrate and contacted with a fluid sample such as blood, serum, urine or saliva. This sample can be
taken directly from the patient or in a partially purified form. In this manner, antibodies specific for
the polypeptide (the primary antibody) will specifically react with the bound polypeptide. Thereafter,
a secondary antibody bound to, or labeled with, a detectable moiety can be added to enhance the
detection of the primary antibody. Generally, the secondary antibody will be selected for its ability to
react with multiple sites on the primary antibody. Thus, for example, several molecules of the
secondary antibodies can react with each primary antibody, making the primary antibody more
detectable.
The detectable moiety allows for visual detection of a precipitate or a color change, visual
detection by microscopy, or automated detection by spectrometry, radiometric measurement or the
like. Examples of detectable moieties include fluorescein, rhodamine, Cy5, and Cy3 (for
fluorescence microscopy and/or the microsphere-based immunoassay), horseradish peroxidase (for
either light or electron microscopy and biochemical detection), biotin-streptavidin (for light or
electron microscopy) and alkaline phosphatase (for biochemical detection by color change).
Vlll. Pharmaceutical and Immune Stimulatory Compositions and Uses Thereof
Pharmaceutical compositions including flavivirus nucleic acid sequences or flavivirus
polypeptides comprising at least one flavivirus cross-reactive epitope with reduced or ablated crossreactivity
are also encompassed by the present disclosure. These pharmaceutical compositions
include a therapeutically effective amount of one or more active compounds, such as flavivirus
polypeptides comprising at least one flavivirus cross-reactive epitope with reduced or ablated crossreactivity,
or one or more nucleic acid molecules encoding these polypeptides, in conjunction with a
pharmaceutically acceptable carrier. It is contemplated that in certain embodiments, flavivirus nucleic
acid sequences or flavivirus polypeptides comprising multiple flavivirus cross-reactive epitopes with
reduced or ablated cross-reactivity will be useful in preparing the pharmaceutical compositions of the
disclosure.
Disclosed herein are substances suitable for use as immune stimulatory compositions for the
inhibition or treatment of a flavivirus infection, for example, a dengue virus infection. In one
embodiment, an immune stimulatory composition contains a flavivirus polypeptide including at least
one flavivirus cross-reactive epitope with reduced or ablated cross-reactivity. In a further
embodiment, the immune stimulatory composition contains a nucleic acid vector that includes
flavivirus nucleic acid molecules described herein, or that includes a nucleic acid sequence encoding
at least one flavivirus cross-reactive epitope with reduced or ablated cross-reactivity. In a specific,
non-limiting example, a nucleic acid sequence encoding at least one flavivirus cross-reactive epitope
with reduced or ablated cross-reactivity is expressed in a transcriptional unit, such as those described
in published PCT Application Nos. PCT/US99/12298 and PCT/US02/10764 (both of which are
incorporated herein in their entirety).
The provided immune stimulatory flavivirus polypeptides, constructs or vectors encoding
such polypeptides, are combined with a pharmaceutically acceptable carrier or vehicle for
administration as an immune stimulatory composition to human or animal subjects. In a particular
embodiment, the immune stimulatory composition administered to a subject directs the synthesis of a
mutant flavivirus E-glycoprotein as described herein, and a cell within the body of the subject, after
incorporating the nucleic acid within it, secretes VLPs comprising the mutant E-glycoprotein with
reduced or ablated cross-reactivity. It is believed that such VLPs then serve as an in vivo immune
stimulatory composition, stimulating the immune system of the subject to generate protective
immunological responses. In some embodiments, more than one immune stimulatory flavivirus
polypeptide, construct or vector may be combined to form a single preparation.
The immunogenic formulations may be conveniently presented in unit dosage form and
prepared using conventional pharmaceutical techniques. Such techniques include the step of bringing
into association the active ingredient and the pharmaceutical carrier(s) or excipient(s). In general, the
formulations are prepared by uniformly and intimately bringing into association the active ingredient
with liquid carriers. Formulations suitable for parenteral administration include aqueous and nonaqueous
sterile injection solutions which may contain anti-oxidants, buffers, bacteriostats and solutes
which render the formulation isotonic with the blood of the intended recipient; and aqueous and nonaqueous
sterile suspensions which may include suspending agents and thickening agents. The
formulations may be presented in unit-dose or multi-dose containers, for example, sealed ampules and
vials, and may be stored in a freeze-dried (lyophilized) condition requiring only the addition of a
sterile liquid carrier, for example, water for injections, immediately prior to use. Extemporaneous
injection solutions and suspensions may be prepared from sterile powders, granules and tablets
commonly used by one of ordinary skill in the art.
In certain embodiments, unit dosage formulations are those containing a dose or unit, or an
appropriate fraction thereof, of the administered ingredient. It should be understood that in addition
to the ingredients particularly mentioned above, formulations encompassed herein may include other
agents commonly used by one of ordinary skill in the art.
The compositions provided herein, including those for use as immune stimulatory
compositions, may be administered through different routes, such as oral, including buccal and
sublingual, rectal, parenteral, aerosol, nasal, intramuscular, subcutaneous, intradermal, and topical.
They may be administered in different forms, including but not limited to solutions, emulsions and
suspensions, microspheres, particles, microparticles, nanoparticles, and liposomes.
The volume of administration will vary depending on the route of administration. By way of
example, intramuscular injections may range from about 0.1 ml to about 1.0 ml. Those of ordinary
skill in the art will know appropriate volumes for different routes of administration.
A relatively recent development in the field of immune stimulatory compounds (for example,
vaccines) is the direct injection of nucleic acid molecules encoding peptide antigens (broadly
described in Janeway & Travers, Immunobiology: The Immune System In Health and Disease, page
13.25, Garland Publishing, Inc., New York, 1997; and McDonnell & Askari, N. Engl. J. Med. 334:42-
45, 1996). Vectors that include nucleic acid molecules described herein, or that include a nucleic acid
sequence encoding a flavivirus polypeptide comprising at least one flavivirus cross-reactive epitope
with reduced or ablated cross-reactivity may be utilized in such DNA vaccination methods.
Thus, the term "immune stimulatory composition" as used herein also includes nucleic acid
vaccines in which a nucleic acid molecule encoding a flavivirus polypeptide comprising at least one
flavivirus cross-reactive epitope with reduced or ablated cross-reactivity is administered to a subject
in a pharmaceutical composition. For genetic immunization, suitable delivery methods known to
those skilled in the art include direct injection of plasmid DNA into muscles (Wolff et al, Hum. Mol.
Genet. 1:363, 1992), delivery of DNA complexed with specific protein carriers (Wu et al., J. Biol.
Chem. 264:16985, 1989), co-precipitation of DNA with calcium phosphate (Benvenisty and Reshef,
Proc. Natl. Acad. Sci. 83:9551, 1986), encapsulation of DNA in liposomes (Kaneda et al., Science
243:375, 1989), particle bombardment (Tang et al, Nature 356:152, 1992; Eisenbraun et al., DNA
Cell Biol. 12:791, 1993), and in vivo infection using cloned retroviral vectors (Seeger et al., Proc.
Natl. Acad. Sci. 81:5849, 1984). Similarly, nucleic acid vaccine preparations can be administered via
viral carrier.
The amount of immunostimulatory compound in each dose of an immune stimulatory
composition is selected as an amount that induces an immunostimulatory or immunoprotective
response without significant, adverse side effects. Such amount will vary depending upon which
specific immunogen is employed and how it is presented. Initial injections may range from about 1
ug to about 1 mg, with some embodiments having a range of about 10 ug to about 800 ug, and still
other embodiments a range of from about 25 ug to about 500 ug. Following an initial administration
of the immune stimulatory composition, subjects may receive one or several booster administrations,
adequately spaced. Booster administrations may range from about 1 ug to about 1 mg, with other
embodiments having a range of about 10 ug to about ,750 ug, and still others a range of about 50 ug to
about 500 ug. Periodic boosters at intervals of 1-5 years, for instance three years, may be desirable to
maintain the desired levels of protective immunity.
It is also contemplated that the provided immunostimulatory molecules and compositions can
be administered to a subject indirectly, by first stimulating a cell in vitro, which stimulated cell is
thereafter administered to the subject to elicit an immune response. Additionally, the pharmaceutical
or immune stimulatory compositions or methods of treatment may be administered in combination
with other therapeutic treatments.
IX. KitsAlso provided herein are kits useful in the detection and/or diagnosis of flaviviruses. An
example of an assay kit provided herein is a recombinant flavivirus polypeptide (or fragment thereof)
as an antigen and an enzyme-conjugated anti-human antibody as a second antibody. Examples of
such kits also can include one or more enzymatic substrates. Such kits can be used to test if a sample
from a subject contains antibodies against a flavivirus-specific protein. In such a kit, an appropriate
amount of a flavivirus polypeptide (or fragment thereof) is provided in one or more containers, or
eld on a substrate. A flavivirus polypeptide can be provided in an aqueous solution or as a freezedried
or lyophilized powder, for instance. The container(s) in which the flavivirus polypeptide(s) are
supplied can be any conventional container that is capable of holding the supplied form, for instance,
microfuge tubes, ampoules, or bottles.
The amount of each polypeptide supplied in the kit can be any appropriate amount, and can
depend on the market to which the product is directed. For instance, if the kit is adapted for research
or clinical use, the amount of each polypeptide provided would likely be an amount sufficient for
several assays. General guidelines for determining appropriate amounts can be found, for example, in
Ausubel et al. (eds.), Short Protocols in Molecular Biology, John Wiley and Sons, New York, NY,
1999 and Harlow and Lane, Using Antibodies: A Laboratory Manual, CSHL, New York, 1999.
The subject matter of the present disclosure is further illustrated by the following nonlimiting
Example 1
Identification of DII cross-reactive epitope residues
This example demonstrates the identification of flavivirus cross-reactive epitopes using a
structure-based rational mutagenesis method.
Cell culture, virus strain and recombinant plasmid
COS-1 cells (ATCC CRL 1650; Manassas, VA) were grown at 37°C with 5% CO2 on
Dulbecco's modified Eagle's minimal essential medium (DMEM, GIBCO, Grand Island, NY)
supplemented with 10% heat-inactivated fetal bovine serum (FBS, Hyclone Laboratories, Inc.,
Logan, UT), 110 mg/1 sodium pyruvate, 0.1 mM nonessential amino acids, 2 mM L-glutamine, 20
ml/1 7.5% NaHCO3, lOOU/ml penicillin, and 100 ug/ml streptomycin. CHO cells (ATCC CCL 61;
Manassas, VA) were grown under the same conditions as COS-1 cells with DMEM/F12 nutrient
mixture (GIBCO, Grand Island, NY).
Flavivirus plasmids capable of expressing extracellular VLPs composed of prM/M and Eglycoproteins
for JE, WN, SLE, and the four DEN virus serotypes have been constructed (Chang et
al, J. Virol. 74:4244-52, 2000; Chang et al, Virology 306:170-80, 2003; Davis et al, J. Virol.
75:4040-47, 2001). These VLPs, produced by recombinant plasmid-transformed eukaryotic cells,
contain the flavivirus prM/M and E-glycoproteins in their native viral conformations, and although
non-infectious, they maintain many of the same properties as mature virus particles including,
hemagglutination activity, membrane fusion, and the induction of protective immune responses in
animals (Chang et al.,J. Virol. 74:4244-52, 2000; Chang et al, Virology 306:170-80, 2003; Davis et
al, J. Virol 75:4040-47, 2001; Hunt, et al, J. Virol. Methods 97:133-49, 2001).
The recombinant expression plasmid pCB8D2-2J-2-9-l (the DEN-2 prM/E expression
plasmid, Chang et al, Virology 306:170-80, 2003) was used as the template DNA for both sitedirected
mutagenesis and for transient expression of DEN-2 recombinant antigen (see below). This
plasmid includes the human cytomegalovirus early gene promoter, Kozak sequence, JE virus signal
sequence, DEN-2 virus prM/M gene, DEN-2 virus chimeric E gene (with amino-terminal 80% from
DEN-2 virus and carboxy-terminal 20% from JE virus), and bovine growth hormone poly(A) signal.
The replacement of the terminal 20% of DEN-2 virus E gene sequences with JE virus E gene
sequences dramatically increases the secretion of extracellular VLPs into the culture medium without
altering the native DEN-2 virus E-glycoprotein conformation (Chang el al., Virology 306:170-80,
2003),
Procedural algorithm
To localize the epitopes responsible for inducing flavivirus cross-reactive antibodies, the
following procedural algorithm was developed: Strictly-conserved flavivirus residues were initially
identified. These residues were mapped onto the 2.0 A resolution E-glycoprotein structure for TBE
virus (Rey et al, Nature 375:291-98, 1995) and onto a computer predicted homology model structure
for the DEN-2 virus E-glycoprotein using the Swiss-Pdb Viewer 3.7 structure analysis software
(Guex et al., Electrophoresis 18:2714-23, 1997; available on the ExPASy Molecular Biology
Server). A brief review of high resolution structures for antigen-antibody complexes revealed that
10-20 residues typically are involved in making direct contacts between the antigen epitope and
antibody paratope. These contacts result in 20-30 residues that are "buried" by the typical antibody
footprint, measuring approximately 20 x 30 A. On average however, only 25% of the buried side
chains, or 4-6 residues, account for most of the mAb binding energy (Arevalo et al, Nature 356:859-
63, 1993; Bhrtet al., PNAS 91:1089-93, 1994; Davies & Cohen, PNAS 93:7-12, 1996; Faebler eta!.,
J. Mol Biol 313:83-97, 2001; Fleury et al, Nature St. Biol. 6:530-34, 1999; Li etal, Biochemistry
39:6296-6309, 2000; Lo et al.,J. Mol. Biol. 285:2177-98, 1999; and Mylvaganam et al.,J. Mol. Biol.
281:301-22, 1998).
The following criteria were developed to select probable flavivirus group cross-reactive
epitope residues: 1) an amino acid located in DII (for example, amino acids 52-135 and 195-285 in
the TBE virus E-glycoprotein (Rey et al., Nature 375:291-98, 1995); 52-132 and 193-280 in the
DEN-2 virus E-glycoprotein (Modis et al, PNAS 100:6986-91, 2003)) and conserved among more
than one flavivirus; 2) amino acids on the outer or lateral surface of the E-glycoprotein dimer; 3)
amino acids with at least 35% surface accessibility potential; 4) side chain projections accessible to
antibody paratopes; and 5) residues with high temperature (p1-) factors should be favored, as these
residues tend to be flexible and are able to conform to the antibody paratope, increasing the antibodyantigen
affinity.
Using this structure-based design approach, candidate flavivirus cross-reactive epitope
residues were narrowed down from a total of 53 conserved amino acids in DII (38 invariant and 15
almost completely conserved), to less than ten probable DII cross-reactive epitope residues. Amino
acid substitutions at these probable cross-reactive epitope residues were computer modeled, selecting
substitutions that should reduce or ablate antibody recognition without altering E-glycoprotein
33
structural conformation, disrupting dimer interactions, or impairing particle formation, maturation, or
secretion. For this reason, cysteine residues otherwise satisfying the cross-reactive epitope criteria
were not considered for mutagenesis because of their involvement in disulphide bridging necessary
for proper E-glycoprotein structure and function (Modis et al, PNAS 100:6986-91, 2003; Rey et al.,
Nature 375:291-98, 1995).
Stability calculations were performed for all possible amino acid substitutions of candidate
residues using the FOLD-X server (Guerois et al, J. Mol. Biol. 320:369-87, 2002; available on the
internet) and the TBE virus E-glycoprotein pdb file coordinates (Rey et al, Nature 375:291-98,
1995). Amino acid substitutions modeled in the TBE virus E-glycoprotein with free energies of
folding equal to or less than that of the non-mutated wild-type E-glycoprotein were re-examined with
the Swiss-PdbViewer software to identify those substitutions that minimized local structural
disturbances while maintaining structurally relevant biochemical interactions such as hydrogen
bonding and/or charge interactions with neighboring amino acids. Because the outer surface of
mature flavivirus particles are covered in a dense network of E and prM/M proteins, any
conformational changes in the E-glycoprotein are likely to induce concerted reorganization across the
surface of the virion (Kuhn et al, Cell 108:717-25, 2002; Modis et al, PNAS 100:6986-91, 2003). A
comparison of the a priori stability calculations based on the TBE virus E-glycoprotein structure with
a posteriori stability calculations from the DEN-2 virus atomic structure are shown in Table 2.
Site-directed mutagenesis
Site-specific mutations were introduced into the DEN-2 virus E gene using the Stratagene
Quick Change® multi site-directed mutagenesis kit (Stratagene, La Jolla, CA) and pCB8D2-2J-2-9-l
as DNA template following the manufacturer's recommended protocols. The sequences of the
mutagenic primers used for all constructs are listed in Table 1. Four or five colonies from each
mutagenic PCR transformation were selected and grown in 5 ml LB broth cultures, mini-prepped and
sequenced. Structural gene regions and regulatory elements of all purified plasmids were sequenced
entirely upon identification of the correct mutation. Automated DNA sequencing was performed
using a Beckman Coulter CEQ™ 8000 genetic analysis system (Beckman Coulter, Fullerton, CA)
and analyzed using Beckman Coulter CEQ™ 8000 (Beckman Coulter, Fullerton, CA) and
Lasergene® software (DNASTAR, Madison, WI).
Transient expression of DEN-2 virus recombinant antigens in COS-] or CHO cells
COS-1 and CHO cells were electroporated with pCB8D2-2J-2-9-l using the protocol
described by Chang et al. (J. Virol 74:4244-52, 2000). Electroporated cells were recovered in 50 ml
DMEM, seeded into 150 cm2 culture flasks for VLP expression and into 96 well tissue culture plates
(Costar® #3603; Corning, Inc., Corning, NY) for IFA, and incubated at 37°C with 5% CO2. Six to
eight hours following electroporation, the growth medium in the 150 cm2 culture flasks was replaced
with DMEM containing 2% FBS. Cells in 96 well plates for IFA were fixed 14-18 hours post
34
electroporation. Tissue-culture medium and cells were harvested 48 and 96 hours post
electroporation for antigen characterization.
Characterization of mutant pCB8D2-J2-2-9-l infected cells and secreted antigen
Fourteen to eighteen hours following electroporation, 96 well tissue culture plates containing
cells transformed with the mutated pCB8D2-2J-2-9-l clones were washed twice with phosphate
buffered saline (PBS), fixed with 3:1 acetone:PBS for 10 minutes and air dried. E-glycoproteinspecific
mAbs specific for each of the three E-glycoprotein domains were used to determine affinity
reductions in DII cross-reactive epitopes by indirect IFA as described by Chang et al. (J. Virol.
Tissue culture medium was harvested 48 hours and 96 hours following electroporation. Cell
debris was removed from tissue culture media by centrifugation for 30 minutes at 10,000 rpm. Ag-
ELISA was used to detect secreted antigen from the mutagenized pCB8D2-2J-2-9-l transformed
COS-1 cells. Secreted antigen was captured with polyclonal rabbit anti-DEN-2 sera (Roehrig et al,
Virology 246:317-28, 1998) at a 1:10,000 dilution. Murine hyper-immune ascetic fluid (MHIAF)
specific for DEN-2 virus was used at a 1:3000 dilution to detect captured antigen, and this MHIAF
was detected using horseradish peroxidase conjugated goat anti-mouse HIAF at a 1:5000 dilution.
Secreted antigen from tissue culture medium was concentrated by centrifugation overnight at 19,000
rpm, and resuspended in TNE buffer (50mM Tris, lOOmM NaCl, lOmM EDTA, pH 7.5) to 1/200*
the original volume. Concentrated antigen was analyzed with a panel of anti-DEN-2 mAbs in Ag-
ELISA to determine mAb end point reactivities of the mutated antigens following the protocol of
Roehrig et al. (Virology 246:317-28, 1998).
Affinity reductions in DII cross-reactive epitopes
Three anti-DEN-2 mAbs, 4G2, 6B6C-1 and 1B7-5, were used to determine affinity
reductions in DII cross-reactive epitopes. These three mAbs share several characteristics: they
recognize surface accessible epitopes in DII, they are flavivirus group- or subgroup-reactive, they are
reduction-denaturation sensitive, they block virus-mediated cell-membrane fusion, they neutralize
virus infectivity, and tryptic fragment mapping indicates that the binding domains of these three
mAbs are formed by two discontinuous DEN-2 virus E-glycoprotein peptide fragments, aal-120 and
158-400 (Aaskov et al.,Arch Virol, 105:209-21, 1989; Henchal et al, Am. J. Trap. Med. Hyg.
34:162-69, 1985; Megret etal, Virology 187:480-91, 1992; Roehrig et al., Virology 246:317-28,
1998). Prospective cross-reactive epitope residues were assessed by looking for decreases in the
reactivity of these three DII flavivirus cross-reactive mAbs for the mutant plasmid transfected cells
by IFA, and mutant VLPs in Ag-ELISA. Proper E-glycoprotein folding and structural conformation
was assessed with a panel of E-glycoprotein DEN virus complex-, subcomplex-, and type-specific
mAbs.
Four potential flavivirus cross-reactive epitope residues were initially focused on. Single
amino acid substitutions were introduced into the DEN-2 prM/E expression plasmid at the following
positions (of SEQ ID NO: 14): Gly,06 to Glu (G,06Q), Trp23i to Phe (W23|F), His244 to Arg (H244R),
and Lys247 to Arg (K247R) (Table 1). Substitutions at Gly,06 and Trp23, strongly interfered with the
binding of flavivirus cross-reactive mAbs (Table 3), However, substitutions at His244 and Lys247 did
not have a measurable effect on the binding of the cross reactive mAbs or of any other mAbs from
the panel.
Glyioe is located within the fusion peptide at the very tip of DII in the E-glycoprotein
monomer (Allison et al., J. Virol. 73:5605-12, 1999; FIGS. 1 and 2). As with the other fusion
peptide residues, Glyioe is strongly conserved across the flaviviruses, the one exception being Modoc
virus with alanine at this position (Table 4). Gly106 is located at the distal end of each E-monomer
along the upper and outer-lateral surface of the dimer. This residue has moderately high surface
accessibility, and its relatively high temperature (p-) factor suggests its potential flexibility. The
substitution of a large, bulky, polar glutamine for the glycine at this position was modeled. The
glutamine substitution fit well into the surrounding region, did not appear to disrupt the local
hydrogen bonding network, and produced acceptable stability calculations using the TBE virus Eglycoprotein
structure coordinates (Table 2).
Trp231 is located in a long intervening loop sequence between DII p-strands h and i (Modis
et al, PNAS 100:6986-91, 2003; FIG. 1). Trp231 lays in a trough on the upper and outer surface of
DII (FIG. 2). It is structurally close to the glycan on Asns7, and lies laterally exterior to the disulfide
bridge between Cys60 and Cys,21. The large hydrophobic side chain lays parallel to the dimer surface
within this trough. This residue is only moderately surface accessible yet its high temperature (P-)
factor and the lack of hydrogen bonding from surrounding residues to the side chain suggest its
potential flexibility. Although all of the substitutions that were modeled at Trp23) were predicted to
induce substantially high energetic costs from the stability analyses, the phenylalanine substitution
was the least costly substitution at this position (Table 2). The phenylalanine fit well into the
surrounding molecular region with limited disruption of the local hydrogen bonding network.
Binding of the GioeQ mutant to either of the two flavivirus group-reactive mAbs, 4G2 and
6B6C-1, was not detected (Table 3). DEN-2 type-specific mAbs 1A5D-1 and 1B4C-2 (DII and DI,
respectively) exhibited reduced affinities for G106Q transfected cells and for secreted VLPs. Dengue
complex-specific mAb 10A1D-2 also exhibited moderately reduced reactivity for the G106Q VLP
antigen (Table 3). However, the reactivity of the GioeQ mutant was unchanged from the reactivity of
the wild-type pCB8D2-2J-2-9-l antigen for polyclonal anti-DEN-2 MHIAF, as well as for the
remaining subcomplex- and type-specific mAbs: 9A4D-1 (DI), 4E5 (DII), and 3H5, 9A3D-8,
10A4D-2, 9D12, and 1A1D-2 (Dili) (Table 3).
The W231F substitution also abolished the binding of both flavivirus group-reactive mAbs,
4G2 and 6B6C-1, as well as that of flavivirus subgroup-reactive mAb 1B7-5 (Table 3). This
substitution additionally interfered with the binding of type-specific DI mAb 1B4C-2, but the binding
of the remaining subcomplex- and type-specific DI, DII and Dili mAbs and a polyclonal DEN-2
MHIAF were unchanged relative to the non-mutated wild-type plasmid (Table 3). In three separate
experiments, secretion of W23]F VLP antigen into the tissue culture medium from transiently
transfected COS-1 cells was not detected. Consequently, the effects of this substitution could only be
analyzed by IFA of plasmid transfected cells.
The H244R and K247R substitutions did not have an effect on the binding of any mAbs in
either IFA of transfected cells, or in Ag-ELISA of secreted VLP antigen.
Example 2
Identification of additional cross-reactive epitopes
through nearest neighbor search
This example demonstrates the identification of additional cross-reactive epitopes using a
"nearest neighbor" search.
Following the identification of cross-reactive epitope residues G\6 and W231 (DEN-2
numbering), the E-glycoprotein atomic structure was reexamined to search for additional flavivirus
cross-reactive epitope'residues. A "nearest neighbor" search was conducted of the surface of the Eglycoprotein
structure, looking for additional residues located within 10-15 A of the identified
residue. This distance is within the binding footprint of a single antibody paratope (Faebler et al.,J.
Mol. Biol. 313:83-97, 2001). In this second iteration of cross-reactive epitope residue identification
the same five optimality criterion as above were used, with one change. The criterion of strict
conservation across the flaviviruses was relaxed to now include variable residues. In this way,
residues either conserved in their physiochemical nature and/or conserved only within a particular
flavivirus complex (such as the four DEN virus serotypes) could be identified.
This nearest neighbor search yielded another seven potential cross-reactive epitope residues.
Amino acid substitutions at these positions were modeled into the TBE virus E-glycoprotein
structure as described above. Mutagenic PCR primers were then synthesized (Table 1) and used to
introduce mutations into the wild-type DEN-2 prM/E expression plasmid. Plasmids were transiently
transfected into CHO cells, and transfected cells and secreted VLP antigen were analyzed with the
anti-DEN-2 mAb panel (Table 3). The substitutions introduced at these positions (of SEQ ID NO:
14) were: Lys64 to Asn (K^N), Thr76 to Met (T76M), Gln77 to Arg (Q77R), Gly)04 to His (Gi04H),
Leu,o7 to Lys (L)07K), Ghii26 to Ala (Ei26A), and Thr226 to Asn (T226N) (Table 2). A single double
mutant combining substitutions at positions 126 and 226 (E126A/T226N) was also examined. Since the
initial W23]F substitution interfered with antigen secretion, the effects of an alternative substitution at
this position, Trp23i to Leu (W23iL), were also examined.
The G,04H, Li07K, and W23]L substitutions had the greatest effect on decreasing the
reactivities of DII cross-reactive mAbs. Gly104 is located on the upper surface of the dimer at the tip
of the tight loop structure which the fusion peptide adopts in the E-glycoprotein dimer (Modis et al.,
PNAS 100:6986-91, 2003; FIG. 2). The residue has moderately high surface accessibility and a
relatively high temperature (p-) factor. The replacement of this small aliphatic glycine was modeled
with a large polar histidine at this position. The histidine residue fits well into this pocket and was
predicted not to alter the hydrogen-bond network in the region. Moreover, because the tick-borne
flaviviruses have a histidine at this position (Table 4) it seemed probable that this substitution would
not disrupt the structure in this localized region or elsewhere within DII. In fact, a posteriori stability
calculations based upon the DEN-2 E atomic structure (Modis et al., PNAS 100:6986-91, 2003)
indicate that the G104H substitution is energetically favorable (Table 2).
The Gio4H substitution, like both substitutions examined at Trp23i, produced a plasmid that
was unable to secrete measurable VLP antigen into the tissue culture medium upon transfection of
either COS-1 or CHO cells. Consequently, the effects of G^H and W23iL substitutions were
analyzed solely by IFA of plasmid transfected cells, as described above for W23iF. The GWH
substitution ablated the reactivity of all three of the flavivirus cross-reactive mAbs, 4G2, 6B6C-1,
and 1B7-5. The type-specific DII mAb 1A5D-1 also showed strongly reduced reactivity for cells
transiently transcribed with this plasmid Table 3). W23iL showed a reduction in mAb reactivities
very similar to W23iF, knocking out any discernable recognition of all three cross-reactive mAbs
(Table 3). The reactivity of DI mAb 1B4C-2 was also reduced by this mutation, but there were no
discernable changes in the reactivities of the remaining subcomplex- and type-specific mAbs or the
anti-DEN-2 MHIAF for either the Gi04H or W231L plasmid constructs (Table 3).
The L107K substituted plasmid exhibited a pattern of reduced reactivities for flavivirus crossreactive
mAbs unlike any of the other substitutions. Leu)07 sits directly below GlyI06 on the outer
lateral surface of the E-protein dimer. This residue has relatively high surface accessibility and
temperature (p-) factor, and its hydrophobic side-chain is directed laterally away from the dimer.
This residue is also strongly conserved across the flaviviruses; the exceptions being the tick-borne
Powassan virus, JE virus strain SA-14-14-2, and DEN-2 virus strain PUO-280 (Table 4). All of these
viruses have a phenylalanine instead of a leucine at this position. A large basic lysine was substituted
for the leucine at this position. Modeling of this L107K substitution indicated that it too was unlikely
to alter the localized hydrogen bonding network. This observation and the low thermodynamic free
energy (ddG) stability calculation (Table 2) suggested that this substitution was unlikely to induce
localized or domain associated conformational changes.
Flavivirus group-reactive mAb 4G2 showed no discernable reactivity for this construct in
either IFA of plasmid transfected cells, or by Ag-ELISA of secreted VLP antigen. However, the
reactivities of the other two cross-reactive mAbs, 6B6C-1 and 1B7-5, were unchanged for this
construct relative to the non-mutated wild-type plasmid (Table 3). L^K plasmid-transfected cells
and secreted VLP antigen also showed moderately reduced reactivity for mAbs 1A5D-1, 10A1D-2
and 1B4C-2, while all other mAbs and the polyclonal MHIAF reactivities were not significantly
different than they were for the wild-type plasmid (Table 3).
Unlike Leu107, Glui26 appears to be incorporated into epitopes recognized by flavivirus
group-reactive mAb 6B6C-1 and subgroup-reactive mAb 1B7-5, but not in the epitope of flavivirus
38
group-reactive mAb 4G2. Glu126 is located 10-12A from Trp231 in the same trough on the upper and
outer surface of DII. The bulky side chain projects from the a-carbon backbone up into this trough
producing a moderately high surface accessibility and a high p-factor (Fig. 2). The replacement of
this large, negatively charged acidic glutamine was modeled with a small hydrophobic alanine at this
position. This substitution was predicted to induce a moderately high, but acceptable energetic cost
in the free energy stability analysis based on the TBE virus E-glycoprotein structure coordinates
(THE virus equals Lys,26, Table 2).
The Ei26A substitution reduced the reactivity of flavivirus group-reactive mAb 6B6C-1, and
moderately reduced the reactivity of subgroup-reactive mAb 1B7-5 (Table 3). However, mAb 6B6C-
1 exhibited reduced reactivity only by IFA of mutant plasmid transfected cells, and 1B7-5 only
showed reactivity reductions for this construct in Ag-ELISA (Table 3). Similarly, type-specific DII
mAb 1A5D-1 exhibited moderately reduced reactivity by Ag-ELISA, but there was no detectable
reduction in its reactivity by IFA (Table 3). The T226N substitution did not alter the reactivity of any
of the flavivirus group-reactive mAbs relative to the non-mutated wild-type plasmid, and the
E|26A/T226N double mutant generally showed a similar pattern of reduction of mAb reactivity as did
Ei26A alone. The two exceptions to this correlation were in the reactivities of mAbs 1B7-5 and
10A1D-2. E|26A/T226N exhibited the same moderate 87% reduction in Ag-ELISA reactivity for
flavivirus subgroup-reactive mAb 1B7-5 as did E^A. However, the double mutant also exhibited a
strong 97% reduction for this same mAb by IFA, which was not observed for either single mutant
(Table 3). DEN virus complex-specific mAb 10A1D-2 also exhibited moderate reactivity decreases
by IFA for this double mutant (Table 3).
K64N, T76M, and ChvR were all unchanged in their reactivities for the flavivirus crossreactive
mAbs. The T76M VLP antigen did however show reduced reactivity for DII type-specific
mAb 1A5D-1 and for DI mAb 1B4C-2 in Ag-ELISA (Table 3).
Example 3
Spatial characterization and organization of flavivirus group-reactive
epitope residues
This example describes the spatial characterization and organization of exemplary flavivirus
cross-reactive epitope residues.
The six residues (G104, G106, L\07, Ei26, T226, and W23i) identified as participating in the
flavivirus cross-reactive epitopes are spatially arranged on the DEN-2 virus E-glycoprotein surface in
two clusters (FIG. 1). The most prominent grouping of these residues is the clustering of three
residues from the highly conserved fusion peptide region of DII (Allison et al.,J. Virol. 75:4268-75,
2001). These residues, Glyio4, GlyI06, and Leu107, are almost completely conserved across the
flaviviruses (Table 4).
The cross-reactive mAbs most strongly affected by substitutions in this region were 4G2 and
6B6C-1. These two mAbs are considered to be quite similar; both are flavivirus group-reactive and
have been grouped into the Al epitope of the E-glycoprotein (Gentry et al.,Am, J. Trap. Med. Hyg.
31:548-55, 1982; Henchal etal.,Am. J. Trap. Med. Hyg. 34:162-69, 1985; Mandl etal, J. Virol
63:564-71, 1989; Roehrig et al, Virology 246:317-28, 1998). The data disclosed herein demonstrate
that although the epitopes of these two mAbs spatially overlap, they do not contain exactly the same
residues. Substitutions at GI04, G106, or L107 knock out the ability of mAb 4G2 to bind to the Eglycoprotein.
However, only substitutions at G104 and G|06 interfere with the binding ability of mAb
6B6C-1. LIO? is therefore not a component of the flavivirus group-reactive epitope recognized by
mAb6B6C-l.
The G104H substitution dramatically reduced the reactivities of all three of the flavivirus
cross-reactive mAbs for this construct (Table 3). Without being bound by theory, it is unlikely that a
glycine residue, with no side chain, would directly participate in the binding energetics of an
antibody-antigen (Ab-Ag) interaction. However, if a glycine residue is included in the buried surface
area of this antibody epitope, the introduction of a large bulky hydrophobic side chain is likely to
disrupt the Ab-Ag shape complementarity and hence increase the dissociation rate-constant (Kj) of
the Ab-Ag interaction (Li et al., Nature Struct. Biol. 10:482-88, 2003). Gi04H also reduced the
recognition of type-specific DII mAb 1A5D-1 (Table 3). The 1A5D-1 epitope is non-neutralizing,
reduction sensitive and moderately surface accessible (Roehrig et al., Virology 246:317-28, 1998).
All of the fusion peptide substitutions introduced into this region reduced the reactivity of 1A5D-1,
consistent with the interpretation that the buried surface area footprint of this mAb not only includes
DEN-2 virus serotype-specific residues, but also includes these strongly conserved residues as well.
A comparison of the DEN-2 atomic structure with flavivirus E-glycoprotein alignments identifies at
least two unique DEN-2, DII, surface accessible residues (Glu7l and Asn83), and a third residue
variable within DEN-2 but distinct from the other DEN virus serotypes (Thr8i). All of these residues
are within 10-22 A of Gly104, a distance well within the buried surface area of a typical Ab-Ag
interface (Lo et al.,J. Mol. Biol. 285:2177-98, 1999). Alternatively, less surface accessible typespecific
residues nearby could participate in mAb 1A5D-1 binding since this epitope itself is only
moderately surface accessible (Roehrig et al., Virology 246:317-28, 1998). Since this mAb is DEN-2
virus specific, these type-specific residues would be expected to provide the majority of the binding
energy for 1A5D-1.
The GioeQ substitution also knocked out the reactivities of both of the flavivirus groupreactive
mAbs, 4G2 and 6B6C-1, though it did not alter the binding of subgroup-reactive mAb 1B7-5
(Table 3, FIG. 2). Type-specific DII mAb 1A5D-1 again lost all measurable reactivity to the G106Q
construct, as did 1B4C-2. The 1A5D-1 epitope footprint appears to include conserved fusion peptide
residues in addition to DEN-2 serotype-specific residues as discussed herein. The reduced reactivity
of DI mAb 1B4C-2 for the GjoeQ construct is difficult to explain. Because of the lack of biological
activity of DI, epitope assignments to this domain can be problematic (Roehrig et al., Virology
246:317-28, 1998). Without being bound by theory, the involvement of Gly)06 as well as that of
Leu 107 are consistent with the possibility that either the previous DI assignment is incorrect, or that
40
the 1B4C-2 mAb footprint includes residues from both DI and DII. However, if 1B4C-2 recognizes
such an inter-domain epitope, this high affinity mAb would be expected to interfere with the Eglycoprotein
dimer to trimer reorganization associated with virus-mediated membrane fusion, which
it does not.
Leu,o7 is the third residue identified in the fusion peptide region of DII that is incorporated
into flavivirus cross-reactive epitopes. Unlike the substitutions at E-glycoprotein positions 104 and
106, the Lio?K substitution knocked out the reactivity of flavivirus group-reactive mAb 4G2, but it
did not alter the reactivity of the other flavivirus group-reactive mAb, 6B6C-1 (Table 3, FIG. 2).
Beyond this discrepancy, the reactivity patterns of the rest of the mAbs for this construct were similar
to that observed for the other fusion peptide substitutions. mAbs 1A5D-1, 10A1D-2, and 1B4C-2 all
showed little to no reactivity for the L^K. construct (Table 3).
Previous studies have examined the effects of mutagenesis in this fusion peptide region.
Pletnev et al. (J. Virol. 67:4956-63, 1993) performed mutagenesis to fusion peptide residues 104 and
107 in a chimeric infectious clone containing the TBE virus structural genes and DEN-4 virus nonstructural
genes. TBE virus has a histidine at position 104 as do all of the tick-borne flavi viruses.
Pletnev et al. constructed the opposite substitution that was constructed herein, H|04G, replacing the
tick-associated histidine with the mosquito-associated glycine, but they were unable to recover live
virus from this construct. They also constructed a double mutant H^G/ L^F from which they were
able to recover virus; however, they were unable to detect any effect of these mutations on mouse
neurovirulence. Allison et al. (J. Virol. 75:4268-75, 2001) also performed mutagenesis at Leu)07
examining the role of this residue in virus-mediated membrane fusion using TBE virus VLPs. They
replaced Leu107 with phenylalanine, threonine, or aspartic acid. They found that all of these
mutations reduced the rate of fusion. Moreover, consistent with the results presented herein, they
found that the L^D substitution appeared to completely abolish the binding of their DII flavivirus
group-reactive mAb A1.
The fourth residue identified as having a major effect on the flavivirus cross-reactive mAbs
was Trp23i, an invariant residue across the flaviviruses (Table 4). Both substitutions introduced at
Trp23i dramatically reduced the reactivity of all three of the flavivirus cross-reactive mAbs, 4G2,
6B6C-1, and 1B7-5. This residue is structurally distant from the fusion peptide region (FIGS. 1 and
2). It is somewhat surprising that substitutions at this residue affect the binding of mAbs also shown
to recognize the distant fusion peptide residues. With out being bound by theory, the strict
conservation of tryptophan (Table 4) and the predicted high energetic costs of substitutions at this
position (Table 2) suggest that this residue could be important for proper DI/DII conformational
structure and function. If this were the case, the loss of reactivity of mAbs recognizing fusion peptide
residues could occur from the induction of localized structural disturbances across DII occurring at a
distance from Trp23i. However, the Trp23i substitutions did not significantly affect the binding of
any of the remaining DII mAbs, 4E5, 1A5D-1, and 10A1D-2 (DI or DII); whereas mAb 1A5D-1
reactivity was reduced or ablated by all of the fusion peptide substitutions. mAb 4E5 does not
recognize native virus yet it blocks virus-mediated cell-membrane fusion, presumably by recognizing
an epitope that is exposed only during or after low-pH-catalyzed conformational changes (Roehrig et
al., Virology 246:317-28, 1998). Without being bound by theory, if substitutions at Trp23i induced
domain wide structural alterations, a loss of reactivity of mAb 1A5D-1 (and the possible exposure of
the non-native-accessible mAb 4E5 epitope, resulting in an increase, or at least a change in, the
reactivity of mAb 4E5 by IFA for these constructs), would be expected. Moreover, the reactivities of
polyclonal MHIAF and of all of the Dili mAbs were no different for these constructs than they were
for the non-mutated wild-type plasmid transfected cells (Table 3). Dili however, is reductiondenaruration
stable and folds into its native IgC like conformation even when this domain is
expressed alone without the remainder of the E-glycoprotein (Bhardwaj et al.,J. Virol. 75:4002-07,
2001).
Both W23iF and W231L plasmids, as well as the G)04H plasmid, failed to secrete measurable
VLP antigen into tissue culture media following transient transfection of COS-1 or CHO cells. The
inability of cells transfected with these plasmids to secrete VLP antigen into tissue-culture media
could result from the disruption of a variety of protein maturation processes. Without being bound
by theory, interference with particle maturation could occur via disruption of E-prM/M
intermolecular interactions, E-glycoprotein dimer interactions, or via the disruption of dimer
organization into the surface lattice covering mature particles. Although the two processes are
interdependent, these substitutions may not interfere with particle formation per se, but may directly
interfere with particle secretion itself. In fact, the IFA staining pattern of DEN-2 GKMH and of
W23|F/L transfected cells was highly punctate and localized within inclusion bodies. Similar IFA
staining patterns have.been observed with non-secreting constructs of dengue and other flaviviruses
(Chang et al, Virology 306:170-80, 2003). Studies with TBE virus VLPs have shown that
interactions between prM and E are involved in prM-mediated intracellular transport of prM-E
heterodimers (Allison et al.,J. Virol. 73:5605-12, 1999). The location of G\ym near the interiorlateral
edge of DII puts it very close to the E-dimer "hole" where the prM/M proteins are located in
the heterodimer (Kuhn et al., Cell 108:717-25, 2002; FIG. 1). Therefore, it seems likely that G^H
interferes with VLP secretion via disruption of the prM-E interactions necessary for intracellular
transport and secretion. The identity of this residue is positively correlated with arthropod vector,
The mosquito-born flaviviruses have a glycine at this position whereas the tick-borne flaviviruses
have a histidine. Interestingly, Pletnev et al. (J. Virol. 67:4956-63, 1993) introduced the reverse
substitution, H,04G, into the TBE virus E-glycoprotein in a TBE/DEN-4 chimeric infectious clone,
and they were unable to recover virus from this mutant. The inability of G^H transfected cells to
secrete VLP antigen similarly suggests that this too could be a lethal substitution in DEN-2 virus.
Taken together, these two results are consistent with the idea that vector-specific selection has
produced strong epistasis between this residue and other unidentified residue(s) elsewhere in the Eor
prM/M proteins.
Example 4
Identification of flavivirus complex and sub-complex cross-reactive
epitope residues
This example demonstrates the identification of flavivirus complex and sub-complex crossreactive
epitopes using a structure-based rational mutagenesis method.
Cell culture, virus strains and recombinant plasmids
CHO cells (ATCC CCL 61; Manassas, VA) were grown at 37°C with 5% CO2 on Dulbeco's
modified Eagle's minimal essential medium with F-12 nutrient mixture (D-MEM/F-12, GIBCO,
Grand Island, NY) supplemented with 10% heat-inactivated fetal bovine serum (FBS, Hyclone
Laboratories, Inc., Logan, UT), 110 mg/1 sodium pyruvate, 0.1 mM nonessential amino acids, 2 mM
L-glutamine, 2.438 g/L NaHCO3, 100 U/ml penicillin, and 100 ug/ml streptomycin.
The recombinant expression plasmids pCB8SJ2 and pCBWN were used as template DNAs
for both site-directed mutagenesis and for transient expression of St. Louis encephalitis virus (SLEV)
and West Nile virus (WNV) recombinant antigen (see below). The pCB8SJ2 plasmid includes the
human cytomegalovirus early gene promoter, Japanese encephalitis virus (JEV) signal sequence,
SLEV prM and E gene region (amino-terminal 80%), JEV carboxyl terminal 20%, and bovine growth
hormone poly(A) signal. The replacement of the terminal 20% of SLEV E with JEV E gene
sequences dramatically increases the secretion of extracellular VLPs into the culture medium without
altering the native SLEV E glycoprotein conformation (Purdy et al.,J. Clin. Micro. 42:4709-17,
2004). The pCBWN plasmid includes the human cytomegalovirus early gene promoter, JEV signal
sequence, WNV prM and E gene region in its entirety, and bovine growth hormone poly(A) signal
(Davis et al.,J. Viral. 75:4040-47, 2001).
Procedural algorithm
Following the identification and ablation of flavivirus group cross-reactive epitopes,
flavivirus complex and sub-complex cross-reactive epitopes have been identified. Two different
flavivirus complexes, the JEV complex and the DENV complex, were focused on. The DENV
complex consists of the four dengue serotypes, DENV-1, DENV-2, DENV-3, and DENV-4. The
large JEV complex includes JEV, WNV, Murray Valley encephalitis virus (MVEV), and SLEV.
The procedural algorithm for the identification of flavivirus complex and sub-complex crossreactive
epitopes utilizes the following optimality criteria: 1) The identification and selection of
amino acid residues with >35% of their surface solvent accessible. These residues are identified from
the published atomic structure coordinates of the DENV-2 soluble ectodomain of the envelope
glycoprotein and homology models of SLEV and WNV derived from the DENV-2 structure (Modis
et al, Proc. Natl. Acad. Sci. USA 100:6986-91, 2003). In addition to examination of amino acid
residues in structural domain II, residues in domains I and III were examined, since published results
indicate that some complex and sub-complex cross-reactive epitopes are mapped onto domains I and
III in addition to domain II (Roehrig et al., Virology 246:317-28, 1998). 2) Amino acids on the outer
or lateral surface of the E-glycoprotein dimer, and accessible to antibody. 3) Amino acid
conservation across the flavivirus complex (based upon a structural alignment of the protein
sequences). Residues conserved across all member viruses of the same complex are favored. If
conserved within but not across the entire complex, then residues with shared identities between
WNV and SLEV are favored in the JEV complex, and residues with shared identities between
DENV-2 and two or more other viruses in the DENY complex are favored over those shared with
DENV-2 and only one other DENY complex virus. 4) Side chain projections exposed towards the
outer surface and accessible to antibody paratopes. 5) Residues with high temperature (P-) factors
should be favored, as these residues tend to be flexible and are able to conform to the antibody
paratope, increasing the antibody-antigen affinity. Amino acid residues with high temperature factors
are more commonly found in antigen epitopes than lower temperature factor residues. 6) Following
identification of potential individual flavivirus complex and sub-complex cross-reactive epitope
residues, all residues are mapped and highlighted on the same E-glycoprotein dimer structure
together. With this technique, groups of potential cross-reactive epitope residues forming clusters
(and hence probable epitopes) are readily identified. 7) Residues fitting all of these criteria and
occurring in structural clusters approximately 20 x 30 A2 (which is the average "footprint" of an
antibody Fab that interacts with an antigen epitope) are favored over residues that are more isolated in
the protein structure. 8) Within an identified structural cluster of potential epitope residues, residues
that more completely satisfy greater numbers of the optimality criteria are selected for the first round
of mutagenesis analysis.
Site-directed mutagenesis
Site-specific mutations were introduced into the SLEY and WNV E genes using the
Stratagene Quick Change® multi site-directed mutagenesis kit (Stratagene, La Jolla, CA) and
pCB8SJ2 and pCBWN as DNA templates following the manufacturer's recommended protocols. The
sequences of the mutagenic primers used for all constructs are listed in Table 5. Four or five colonies
from each mutagenic PCR transformation were selected and grown in 5 ml LB broth cultures. DNA
was mini-prepped and sequenced from these cultures. Structural gene regions and regulatory
elements of all purified plasmids were sequenced entirely upon identification of the correct mutation.
Automated DNA sequencing was performed using a Beckman Coulter CEQ™ 8000 genetic analysis
system (Beckman Coulter, Fullerton, CA) and analyzed using Beckman Coulter CEQ™ 8000
(Beckman Coulter, Fullerton, CA) and Lasergene® software (DNASTAR, Madison, WI).
Transient expression of SLEV and WNV recombinant antigens by CHO cells
CHO cells were electroporated with pCB8SJ2 or pCBWN using the protocol described by
Chang et al (J. Virol, 74:4244-52, 2000). Electroporated cells were recovered in 50 ml DMEM,
seeded into 150 cm2 culture flasks for VLP expression and into 96-well tissue culture plates for IFA,
and incubated at 37°C with 5% C02. Cells in 96 well plates for IFA were fixed 14-24 hours post
electroporation. Tissue-culture medium and cells were harvested 48-72 hours post electroporation for
antigen characterization.
Characterization of mutant pCB8SJ2 andpCBWN infected cells and secreted antigen
Fourteen to twenty four hours following electroporation, 96-well tissue culture plates
(Costar® #3603 Coming, Inc., Corning, NY) containing cells transformed with the mutated pCB8SJ2
or pCBWN clones were washed twice with PBS, fixed with 3:1 acetone:PBS (v:v) for 10 minutes and
air dried. E-glycoprotein-specific mAbs recognizing each of the three E-glycoprotein domains (Table
6) were used to determine affinity reductions in cross-reactive epitopes by IFA as described by Chang
et al. (J. Virol. 74:4244-52, 2000).
Tissue culture medium was harvested 48-72 hours following electroporation. Cell debris
was removed from tissue culture media by centrifugation for 30 minutes at 10,000 rpm. Ag-ELISA
was used to detect secreted antigen from the mutagenized pCB8SJ2 and pCBWN transformed CHO
cells. Secreted antigen was captured with polyclonal rabbit anti-SLEV and rabbit anti-pCBWN sera
at 1:30,000 and 1:50,000 dilutions, respectively. MHIAF specific for SLEV and WNV was used at a
1:15,000 dilution to detect captured antigen, and this MHIAF was detected using horseradish
peroxidase conjugated goat anti-mouse HIAF at a 1:5000 dilution.
Secreted antigen was concentrated from positive tissue culture medium by centrifugation
overnight at 19,000 rpm, and resuspended in TN buffer (50 mM Tris, 100 mM NaCl, pH 7.5) to
1/100* the original volume. Alternatively, some antigens were concentrated using Millipore's
Amicon® Ultra PL-100 (Millipore, Billerica, MA) centrifugal filter devices. Concentrated antigen
was analyzed with a panel of anti-flavivirus mAbs in Ag-ELISA to determine mAb end point
reactivities of the mutated antigens, following the protocol of Roehrig et al. (Virology 246:317-28,
1998). This Ag-ELISA protocol is the same as that used herein to detect secreted antigen, with the
exception of using the specified mAbs (Table 6) instead of polyclonal MHIAF.
Antigenic characterization and MAb screening of potential cross-reactive epitope residue mutants
Using the structure-based design approach described above, candidate flavivirus complex
and sub-complex cross-reactive epitope residues were narrowed down to 34 in DENV-2 and 31 each
in WNV and SLEV. From these residues and with reiterative application of the optimality criteria
described herein 17 DENV-2, 13 WNV, and 11 SLEV residues were chosen as most likely to be
incorporated into complex and sub-complex cross-reactive epitopes (highlighted in Tables 7-9).
Amino acid substitutions were modeled at these probable cross-reactive epitope residues, selecting
substitutions that should potentially disrupt or ablate antibody recognition without altering Eglycoprotein
structural conformation, disrupting dimer interactions, or impairing particle formation,
maturation, or secretion. Stability calculations were performed for all possible amino acid
substitutions of candidate residues using the PoPMuSiC server, (available on the Universite Libre de
Bruxelles' web site) and the DENV-2 E-glycoprotein pdb file coordinates (Modis et al,, Proc. Natl.
Acad. Sci. 100:6986-91, 2003) or homology model coordinates for WNV and SLEV. Amino acid
substitutions modeled in the E-glycoprotein structures with free energies of folding equal to or less
than that of the non-mutated wild-type E-glycoprotein were re-examined with the Swiss-Pdb Viewer
software (available on the Swiss Institute of Bioinformatics' web site) to identify those substitutions
that minimized local structural disturbances while maintaining structurally relevant biochemical
interactions such as hydrogen bonding and/or charge interactions with neighboring amino acids.
Substitutions at 11 of 16 potential cross-reactive epitope residues selected for mutagenesis in
pCB8SJ2 altered the reactivities of all 14 of the anti-SLE mAbs, relative to wild-type pCB8SJ2
(Table 10). Eight of the 14 MAbs were flavivirus group- or subgroup-cross-reactive (see Table 6).
Substitutions at nine of the 16 residues analyzed altered the reactivity of all eight of the flavivirus
group- or subgroup-cross-reactive mAbs. Substitutions at four of 16 potential cross-reactive epitope
residues altered all three of the JEV complex- and subcomplex-cross reactive mAbs. Only one
substitution however, affected type-specific mAb reactivities (Fig. 3). The effect of this substitution
(GioeQ) on type-specific mAb reactivities was to actually increase the reactivity of these mAbs
relative to that of the wild-type unaltered pCB8SJ2. Without being bound by a single theory, such
increase in the reactivity of type-specific antibodies is believed to be beneficial for the development
of type-specific flavivirus antigens.
Substitutions at 14 of 17 residues selected for mutagenesis in pCBWN altered the reactivities
of all 10 of the anti-WNV mAbs, relative to wild-type pCBWN (Table 11). Six of the 10 anti-WNV
mAbs were flavivirus group- or subgroup-cross-reactive, two were JEV complex cross-reactive and
two were WNV type-specific (see Table 6). Nine of the 17 substitutions examined altered the
reactivities of all six group- and subgroup-cross-reactive mAbs; 12 of these 17 substitutions affected
the reactivities of both of the JEV complex cross-reactive mAbs. The GioeV substitution in pCBWN
was the only substitution to alter type-specific mAb reactivities, and, as with pCB8SJ2, this
substitution actually increased the reactivity of the type-specific mAbs (Fig. 3).
The outcome that many of these substitutions altered mAb reactivities (Tables 10 and 11;
Fig. 3) illustrates not only the efficiency of the described algorithms for identifying cross-reactive
epitope residues, but also that these cross-reactive epitopes can be altered to ablate or appreciably
interfere with the ability of an antibody to recognize these modified antigens. For example, 82% and
69% of the potential cross-reactive epitope residue substitutions examined in pCBWN and pCB8SJ2,
respectively, affected all of the cross-reactive antibodies reactive to these two viruses from the
antibody panel (see Fig. 3). The high percentage of residues, selected a priori, affecting mAb
reactivities illustrates the accuracy of the cross-reactive epitope residue selection algorithms.
The mAb characterization of potential cross-reactive epitope residue mutants illustrates the
importance of the E-protein fusion peptide region as a potently cross-reactive antigenic determinant.
As described herein, substitutions at fusion peptide residues GKM, G106, and LIQ? strongly affected
many of the mAb reactivities for DENV-2, SLEV and WNV (see Tables 10 and 11). Without being
bound by a single theory, Gioe appears to be the most important cross-reactive antigenic determinant
of these residues. Substitutions at G,06 altered the reactivities of 7 of 10 cross-reactive mAbs
recognizing SLEV, and 7 of 8 cross-reactive mAbs recognizing WNV (see Tables 10 and 11).
Substitutions at fusion peptide residue G104 also affected the reactivities of many mAbs for each of
these viruses. However, all substitutions examined at this position produced plasmids that were
unable to efficiently secrete VLP antigen upon transient transformation into eukaryotic cells. This
observation was true for all three flaviviruses examined: DENV-2, SLEV and WNV.
Substitutions at fusion peptide residue Gjoe had a variety of effects on mAb reactivities for
both pCBWN and pCB8SJ2. The majority of the substitutions at this residue reduced or ablated a
mAb's ability to recognize the antigen. This occurred with cross-reactive mAbs 4G2, 6B6C-1, 4A1B-
9, and 2B5B-3 in Gi06V-pCBWN and with 4G2 and 2B5B-3 for G106Q-pCB8SJ2 (see Tables 10 and
11), indicating that the substituted residue is a part of the antigenic epitope recognized by these
antibodies.
Example 5
Human IgM MAC-ELISA Serology
This example demonstrates the representative nature of a murine antibody response as a
model of human antibody response to substitutions in the flavivirus cross-reactive epitopes.
Human sera
Well-characterized serum specimens were assembled from the Diagnostic and Reference
Laboratory, Arbovirus Diseases Branch, Division of Vector-Borne Infectious Diseases, US Centers
for Disease Control and Prevention. A serum panel (see Table 12) was assembled from patients
infected in the US between 1999 and 2004 with either WNV («=6) or SLEV («=10), as determined by
the standard 90% plaque-reduction neutralization (PRNT) assay. SLEV is endemic to North
America, whereas WNV was first introduced into North America in 1999 and has spread epidemically
since that time.
The flavivirus responsible for the most recent infection was determined as that with the
highest neutralizing antibody titer, which had to be at least four-fold greater than that for any other
virus tested. Because of the high level of cross-reactivity between the SLEV and WNV viruses, it is
often difficult to determine the infecting virus by ELISA, thus requiring the PRNT. SLEV infected
sera with measurably high levels of cross-reactivity for WNV were purposefully selected in order to
maximize the ability to asses for improved discrepancy (specificity) of the pCBWN-GioeV versus the
pCBWN wild-type antigen. SLEV infected patient sera were split into two groups based upon
previously determined (Diagnostic and Reference Laboratory) positive to negative (P/N) ratios for
SLEV and for WNV. 'Equivocal' SLEV sera («=5) were those that were clear SLEV infections from
the PRNT data, yet had MAC-ELISA P/N ratios that were not statistically different between SLEV
and WNV. Three of these equivocal SLEV samples were negative (P/N was presumptive positive (P/N >2.0 and 5.0) for both
viruses. 'Misleading' SLEV sera («=5) were SLEV positive in the PRNT, yet had MAC-ELISA P/N
ratios that were not only positive for both viruses, but were actually greater for WNV than for SLEV.
Definitive 'positive' WNV infected patient sera («=6) were selected based on MAC-ELISA results
from the Diagnostic and Reference Laboratory collection for use as positive control sera to assess the
accuracy of the pCBWN-G,o6V plasmid derived antigen.
IgM ELISA protocols
IgM ELlSAs were performed following the protocols of Purdy et al. (J. Clin. Micro.
42:4709-17, 2004) and Holmes et al. (J. Clin. Micro. 43:3227-36, 2005). Briefly, the inner 60 wells
of Immulon II HB flat-bottomed 96-well plates (Dynatech Industries Inc., Chantilly, VA) were coated
overnight at 4°C in a humidified chamber with 75 ul of goat anti-human IgM (Kierkegaard & Perry
Laboratories, Gaithersburg, MD) diluted at 1:2000 in coating buffer (0.015 M sodium carbonate,
0.035 M sodium bicarbonate, pH 9.6). Wells were blocked with 300 ul of InBlock blocking buffer
(Inbios, Seattle, WA, L/N FA1032) for 60 minutes at 37°C in a humidified chamber. 50 ul of sera
were added to each well and incubated again for 60 minutes at 37°C in a humidified chamber. Human
test sera were diluted 1:400 in sample dilution buffer (Inbios, L/N FA 1055). Positive control sera
were diluted 1:3000 for SLEV and 1:800 for WNV. Positive and negative control VLP antigens were
tested on all patient sera in triplicate by diluting appropriately in sample dilution buffer and adding
50 ul to appropriate wells for incubation overnight at 4°C in a humidified chamber. Captured
antigens were detected with 50 ul/well of polyclonal rabbit anti-pCBWN diluted 1:1000 in sample
dilution buffer and incubated for 60 m at 37°C in a humidified chamber. Rabbit sera was detected
with horseradish peroxidase conjugated goat anti-rabbit sera diluted 1:8000 in IgM conjugate dilution
buffer (Inbios, L/N FA 1056) and incubated for 60 m at 37°C in a humidified chamber. Bound
conjugate was detected with 75 ul of 3,3'5,5'-tetramethylbenzidine (Neogen Corp, Lexington, KY)
substrate, incubated at RT for 10 min, stopped with 50 ul of 2N H2SO4, and then read at A450 using an
ELx405HT Bio-Kinetics microplate reader (Bio-Tek Instruments Inc., Winooski, VT).
IgM test validation and interpretation
Test validation and P/N values were determined according to the procedure of Martin et al.
(J. Clin. Micro. 38:1823-26, 2000), using internal positive and negative serum controls included in
each 96-well plate. Positive (P) values for each specimen were determined as the average A45o for the
patient serum sample incubated with positive VLP antigen. Negative (N) values were determined for
each plate as the average A4so for the normal human serum control incubated with positive VLP
antigen.
Human serology
To determine how representative the murine antibody response (mAb data) is as a model of
the human antibody response (serological data) to the viral substitution antigens described herein,
serological assays were performed with single substitution, prototype type-specific antigens. As the
mAb screening results indicated that fusion peptide residue 106 was incorporated into multiple crossreactive
epitopes for both WNV and SLEV, this substitution was selected to conduct MAC-ELISA
serum tests.
The prototype type-specific GioeV-WNV Ag dramatically outperformed the wild-type (wt)-
WNV Ag when tested on 10 difficult to discern 'equivocal' or positively 'misleading' SLEV-infected
patient sera (Table 12). Six of 10 of these SLEV infected sera were correctly diagnosed as WNVnegative
by MAC-ELISA (P/N >2.0 WNV Ag, only four sera were correctly scored as WNV negative, one was equivocal, and five were
misdiagnosed as WNV positive with this unmodified Ag. When antigens were directly compared on
each individual serum sample, the G105V-WNV Ag produced lower P/N ratios than did the wt-WNV
Ag in nine of 10 cases on these SLEV infected sera, indicating that the G105V-WNV Ag exhibits
improved specificity and reduced cross-reactivity relative to the wt-WNV Ag.
The prototype type-specific GioeV-WNV Ag also outperformed the unaltered wt-WNV Ag in
MAC-ELISA sensitivity tests on positive WNV infected human sera (Table 12). Five of six WNV
infected patient sera had positive P/N ratios when tested with the GioeV-WNV Ag, whereas four were
positive with the wt-WNV Ag. The single WNV positive serum sample that tested negative with the
wt-Ag and equivocal with the GioaV Ag had the lowest neutralizing tilers of the WNV sera in the
PRNT (see Table 12), indicative of a weak antibody titer.
In addition to improved accuracy with the GioeV-WNV Ag, it was also more sensitive than
was the wt-WNV Ag. In 5 of the 6 WNV infected sera, the MAC-ELISA P/N ratios were higher with
the G]06V- than with the wt-WNV Ag (Table 12). Higher P/N ratios are expected from an improved
type-specific Ag relative to the cross-reactive wt Ag when tested on sera infected with the same virus.
The prototype type-specific GjoeV-WNV Ag exhibited improved specificity, accuracy, and
sensitivity relative to the unmodified wt-WNV Ag. The GioeV-WNV Ag was more specific and
accurate for WNV diagnosis than was the wt Ag, correctly diagnosing more WNV infected sera as
positive and fewer SLEV infected sera as negative, than did the wt-WNV Ag. The GioeV-WNV Ag
was also more sensitive at detecting WNV antibody in WNV infected serum than was the wt-WNV
Ag. The positive signal indicating the presence of WNV antibody (P/N ratios) was greater for GioeVWNV
Ag than it was for the wt-Ag when testing WNV infected sera, and less than that of the wt-Ag
when testing non-WNV infected sera.
Example 6
Murine Immunization
This example demonstrates the ability of prototypical type-specific flavivirus mutant
compositions to generate type-specific neutralizing antibody responses in mice.
Mouse vaccination
Groups of six female outbred ICR mice were used in this study. Mice were immunized by
injection with pCB8D2-2J-2-9-l, pCB8D2-2J-2-9-l-G106Q, pCBWN, pCBWN-G,o6V, pCB8SJ2, or
pCB8SJ2-G,o6Q expression plasmids as described herein. Each mouse was injected with 100 ug of
Picogreen® fluorometer quantified plasmid DNA in PBS pH 7.5, at a concentration of 1 ug/ul. Mice
were immunized with 50 ug of plasmid DNA injected intramuscularly into each thigh on weeks 0 and
3. Mice were bled on week six following initial vaccination.
Plaque reduction neutralization assays
Six week post-vaccination serum specimens were tested for the presence of type-specific
neutralizing (Nt) antibody (Ab) by plaque reduction neutralization test (PRNT). PRNT was
performed with freshly confluent Vero cell monolayers as described by Chang et al. (J. Virol.
74:4244-52, 2000) using DENV-2 (16681), WNV (NY-99), and SLEV (MSI-7) viruses.
Neutralizing antibody responses
Mice were immunized with wild-type and G!06 substituted plasmids for WNV, SLEV, and
DENV-2 to determine if there were differences between the wild-type and G]06 prototype typespecific
antigens for type-specific Nt Ab liter, cross-reactive Nt Ab titer, and protection from virus
challenge. The type-specific Nt Ab titer results are shown in Table 13. There was little difference in
the 75% PRNT titer between wt and G106 substituted plasmids for all three viruses. The 75% Nt Ab
titer was greater than or equal to 1:128 for almost all of the mice immunized with both the DENV-2
and both the WNV DNA vaccines. One mouse immunized with the wt DENV-2 DNA vaccine had a
75% PRNT titer of 1:64, and two mice immunized with the pCBWN-Gio6V DNA vaccine had 75%
PRNT liters of 1:64 and 1:16.
These results demonstrale lhat for all three flaviviruses tesled, mere was little to no
detectable difference in type-specific neutralizing antibody liter between the prototype type-specific
G)06 mutant vaccines and their wt counterparts. These results also illustrate lhat the methods
described herein for ablating cross-reaclive epitope residues can be used to generate type-specific
flavivirus prM/E expression plasmids for use as DNA vaccines that still maintain potent type-specific
neutralizing immunogenicily.
Example 7
Reduction of Cross-reactive Immunogenicity of Type-specific Genetic Vaccines
This example provides methods by which prototypical type-specific flavivirus mutant
compositions can be used lo generate a reduced cross-reactive neutralizing antibody response relalive
to the unaltered wild-lype composilions.
Mouse vaccination and plaque reduction neutralization assays
Female outbred ICR mice (such as the mice in Example 6) can be used in this study.
Twelve-week post vaccination serum samples from immunized mice will be tested for cross-reactive
(heterologous) Nt antibody response by PRNT. Unlike the type-specific PRNTs performed in
Example 6, the cross-reactive PRNTs will be performed by examining Nt of immunized mouse sera
not only for the type-specific virus used for immunization, but also for Nt of the seven other medically
important flaviviruses. Thus, all 12-week mouse sera will be tested for neutralization against eight
different flaviviruses: all four dengue serocomplex viruses, DENV-1 (16007), DENV-2 (16681),
DENV-3 (H87), and DENV-4 (H241); three JEV serocomplex viruses, JEV (SA14-14-2), WNV
(NY-99) and SLEV (MSI-7); and the single medically important member of the yellow fever virus
serocomplex, YFV (17D).
Predicted antibody response
Without being bound by theory, similar type-specific Nt Ab titers between the prototype
type-specific Gioe mutant vaccines and their wt counterparts are expected. Thus, both pCBWN and
pCBWN-GioeV vaccinated mouse sera are predicted to have similar Nt Ab titers against WNV, and
pCB8D2-2J-2-9-l and pCB8D2-2J-2-9-l-G106Q will have similar Nt Ab titers against DENV-2.
However, when these same sera are tested for Nt against the heterologous flaviviruses, it is expected
that significantly lower PRNT titers for prototype type-specific G)06 mutant vaccinated mouse sera
will be observed than for the counterpart wt vaccinated mouse sera. For example, mice immunized
with pCBWN and pCBWN-G,06V will both have similar PRNT titers against WNV, whereas,
pCBWN-G106V immunized mice will have significantly lower PRNT titer against SLEV, JEV, YF,
and the four dengue serotype viruses, than wild-type pCBWN immunized mice.
Example 8
Combining Multiple Cross-reactive Epitope Substitutions into Single Plasmid Constructs
This example provides methods by which individual substitutions affecting different
flavivirus cross-reactive epitopes can be combined into a single construct.
Individual substitutions affecting different flavivirus cross-reactive epitopes (such as those
disclosed herein) can be combined into a single construct based, for example, on mAb screening
results disclosed herein (see, Tables 3, 10 and 11), as well as additional mAb screening studies. For
example, a mutagenesis primer has been designed for SLEV to introduce both the GioeQ and L^K
substitutions into a single pCB8SJ2 plasmid (see, Table 5). This double mutation plasmid has been
constructed, and its sequence confirmed.
Cells can be transformed with this double mutated plasmid (or another plasmid containing a
sequence encoding an E glycoprotein having a combination of two or more mutated amino acids),
and the antigen characterized. In SLEV, the G106Q substitution alone alters the reactivities of many
mAbs recognizing distinct cross-reactive epitopes (Table 10). However, this substitution alone has
no significant effect on the flavivirus group cross-reactive epitope recognized by MAb T-23-1. The
Lio7K substitution does knock out the ability of mAb T-23-1 to recognize the flavivirus cross-reactive
epitope. Without being bound by theory, this suggests that L107 is incorporated in the cross-reactive
epitope recognized by mAb T-23-1, while doe is not.
Because of the generally additive effects observed when combining these substitutions into
single constructs (see, Tables 10 and 11), it is expected that GioeQ/LioyK. antigen will combine the
different effects observed from mAb screening of the individual mutants into a single, multiple
substituted mutant. Upon transfection into mammalian cells, such a multiple mutant plasmid can be
used to produce improved type-specific antigens. When utilized as genetic vaccines, these plasmids
are expected to exhibit further reductions in cross-reactive immunogenicity while still inducing a
potent type-specific immune response.
Example 9
Immune Stimulatory Compositions for the Inhibition or Treatment of a Flavivirus Infection
This example provides methods for administering substances suitable for use as immune
stimulatory compositions for the inhibition or treatment of a flavivirus infection.
An immune stimulatory composition containing a therapeutically effective amount of a
flavivirus polypeptide that includes at least one flavivirus cross-reactive epitope with reduced or
ablated cross-reactivity (particularly in an E glycoprotein) can be administered to a subject at risk for,
or exposed, to a flavivirus (e.g., a dengue virus, West Nile virus, etc.). Alternatively, an immune
stimulatory composition containing a therapeutically effective amount of a nucleic acid vector that
includes flavivirus nucleic acid molecules described herein, or that includes a nucleic acid sequence
encoding at least one flavivirus cross-reactive epitope with reduced or ablated cross-reactivity
(particularly in an E glycoprotein), can be administered to a subject at risk for, or exposed to a
flavivirus.
Dosages and routes of administration for the immune stimulatory composition can be readily
determined by one of ordinary skill in the art. Therapeutically effective amounts of an immune
stimulatory composition can be determined, in one example, by in vitro assays or animal studies.
When in vitro or animal assays are used, a dosage is administered to provide a target tissue
concentration similar to that which has been shown to be effective in the in vitro or animal assays.
While this disclosure has been described with an emphasis on preferred embodiments, it will
be apparent to those of ordinary skill in the art that variations and equivalents of the preferred
embodiments may be used and it is intended that the disclosure may be practiced otherwise than as
specifically described herein. Accordingly, this disclosure includes all modifications encompassed
within the spirit and scope of the disclosure as defined by the claims below.
APPENDIX I
TABLES
Table 1. Nucleotide sequence of primers used for mutagenesis. The mismatched nucleotides causing
the desired substitutions are underlined.
Sequence
5'-TGTTGTTGTGTTGGTTAGGTTTGCCTCTATACAG-3'
5'-TGGGTTCCCCTTGCATTGGGCAGCGAGATTCTGTTGTTG-3'
5 ' -TTC ATTTAGGCTGGGTTCCCCTCGTGTTGGGC AG-3 '
5 '-CCCTTTCCAAATAGTCCAC AGTG ATTTCCCCATCCTCTGTCTACC-3 '
5 ' -GCCTCCCTTTCCAAATAGTTG ACATCC ATTTCCCCA-3 '
5 '-GGTCACAATGCCTCCCTTTCCAAATTTTCCACATCCATTTCCCC-3 '
5 '-AGTTTTCTGGTTGCACAACTTTTCCTGCCATGTTCTTTTTGC-3 '
5 '-GTATCCAATTTGACCCTTGATTGTCCGCTCCGGGCAACC-3 '
5 '-GTCTCTTTCTGTATGAAATTTGACCCTTGTGTGTC-3 '
5 '-AATGTCTCTTTCTGTATCAGATTTGACCCTTGTGTGTCCGCTCC-3 '
5 '-TCCTGTTTCTTCGCACGGGG ATTTTTGAAAGTGACC-3 '
5 ' -AC AAC AACATCCTGTCGCTTCGCATGGGG ATTTTTG-3 '

Table 2. Stability free energy (ddG) calculations for putative domain II cross-reactive epitope
substitutions based upon the published pdb coordinates for the DEN-2 virus (Modis et al, PNAS
100:6986-91, 2003) and the TBE virus (Rey et al, Nature 375:291-98, 1995) E-glycoprotein
na: not applicable (these constructs did not secrete VLP antigen and thus could not be examined by Ag-ELISA); nd: not determined.
55
Table 4. Amino acid sequence variability for proposed cross-reactive epitope residues in domain II of the flavivirus E protein.
Japanese Encephalitis
Murray Valley encephalitis
West Nile
St. Louis encephalitis

DENV-2 strains containing variable amino acid sequences at these positions are indicated below with their GenBank accession numbers (all incorporated
by reference as of the date of filing of this application).
64R:AF359579; 77L:M24449, X15434, X15214; 107F: M24446
126K:L10053, D00346, M29095, AF204178, M24450, M24451, AF410348, AF410361, AF410362, AF410365, AF204177, D10514
Table 5. Nucleotide sequence of primers used for mutagenesis. The mismatched nucleotides causing
the desired substitutions are underlined.

CTCCCTTTTCCAAACAGACCACAGTGGTTACCCCATCCGC
CTCCCTTTTCCAAACAGACCACAGTTGTTACCCCATCCGC
CCCTTTTCCAAACAGACCACAGTCGTTACCCCATCCGC
CTCCCTTTTCCAAACAGACCACACTTGTTACCCCATCCGC
CTCCCTTTTCCAAACAGCTGACATCCGTTACCCCATCCGC
CTCCCTTTTCCAAACAGCTTACATCCGTTACCCCATCCGC
TCCCTTTTCCAAACAGTACACATCCGTTACCCCATCCGC
CTTTTCCAAACAGATCACATCCGTTACCCCATCCGC
CTCCCTTTTCCAAAGAAACCACATCCGTTACCCCATCCGC
AATGCTCCCTTTTCCAAAGAACTGACATCCGTTACCCCATCCGC
CGGGCTTATGGTGAATTGAGCCGCTTGGTTTTTTCC
TTCCATACTCGCCCATGTTGGCAATAAAGGACGGTG
GTAACTGTTCCATACTCGGACATGTTGGCCGTAAAGG
GTAACTGTTCCATAGTTGCCCATGTTGGCCGTAAAGG
CTCTGTTGCGCCAATCGTTTGTGGCAGGGCTCGTC
TTCTCTGTTGCGGAAATCAGTTGTGGCAGGGCTCGTC
TACTACAGTTTGCTTGGTGGCACGCGGTTCCTC
TGATTGCAAGGTTAGGGTTGATCCGCTAACAGTGGC
CGTTCCCTTGATTTTGACGTAGTCAAGCTTAGCTCTGC
ACACATGCCATATGCCGTTCCCTTGATTTTGACC
CAGGGTCCGTTGCTTCCATCATACTGCAGTTCCAC
CGATCATGACCTTGTTGTTCGATCCCCCTGTGC
TTCGATCATGACCTTGTTGAACGCTCCCCCTGTGC
TTTGCCAAATAGTCCGCAGTTGTTGCCCCAGCCCC
TTGCCAAATAGTCCGCAGTCGTTGCCCCAGC
CCTTTGCCAAATAGTCCGCACTTGTTGCCCCAGCCCC
TTGCCAAATAGTCCGCATGCGTTGCCCCAGC
TTTGCCAAATAGGACGCAGCCGTTGCCCCAGCC
TTTGCCAAATAGCCTGCAGCCGTTGCCCCAGCC
CCTTTGCCAAATAGGTAGCAGCCGTTGCCCCAGCCCC
TTTGCCAAATAGAGCGCAGCCGTTGCCCCAGCC
TTCCTTTGCCAAAGTATCCGCAGCCGTTGCCCCAGCC
CCTTTGCCAAAGAATCCGCAGCCGTTGCCCCAGC
CCTTTGCCAAAATGTCCGCAGCCGTTGCCCCAGC
CCTTTGCCAAATCTTCCGCAGCCGTTGCCCCAGC
CTTGGTAGAGCAGGCAAATACGGCGCATGTGTC
CCAACCTGTGTGGAGTAGTCTCCGTGCGAC
CCAACCTGTGTGGAGCCGTTTCCGTGCGACTC
CTGAGTGGCTCCAACATCTGTGGAGTAGTTTCCGTGCG
AGGAGTGATGCTGAAGTACCCTGCCTGAGTGG
CCAAGCTTTAGTACGTATGAAGGCGCCGCAGGAG
CCTCTCCATAGCCTCCAAGCTTTAGTGTGTATGAAGG
AACGTCTCTCTGTTCCTGAACACAGTACTTCCAGCAC
CCGACGTCAACTTGACAGTGTTGTCTGAAAATTCCACAGG
GTTCCCTTCAACTGCAAGTTTTCCATCTTCACTCTACAC

ACAGACGCCATAGTTTGTTCCCTTCAACTGCAATTTTTCC
CCATCCGTGCCGTTGTACTGCAATTCCAACACCACAG
GGACCTTAGCGTTGACCGTGGCCACTGAAAC
ACCTTAGCGCTGGCCGTGGCCACTGAAAC
Table 6. E-glycoprotein-specific mAbs recognizing each of the three E-glycoprotein domains
sub-grp: DEN+JE comp
sub-grp: DEN+JE comp
sub-grp. JE comp + YF
sub-grp: DEN-2,3 ,4 + JE
sub-grp: DEN-2, JE, SLE
sub-grp: DEN- 1, 2, JE, WN*
sub-grp: DEN-1,2,3,4 + SLE
JE comp.
JE comp.
sub-comp: DEN-2,3
sub-comp: DEN-1,2,3
sub-comp: SLE + JE
sub-comp: DEN-2 + JEV
sub-comp: DEN-1,2,3
top outer edge
top outer edge SDM in D2
out-mid-lat. SDM in D2
out low lat mid SDM in D216681 -yes
up-mid-top below CHO-153
up-mid-top near CHO 153
up-in-top near CHO & prM
up-mid-top near prM
up-mid-top high.exp.
out-lat mid DI/II region
DI/II border out-mid
DI/II border up-out edge
DI/III up-out edge
DI/III up-out edge
in-mid-up
in-mid-up no SDM in D2
up-out-middle (SDM in D2)
up-out-mid SDM previous
up-out-middle
out-up lat DI/III face
out-up lat DI/III face, above D290
mid-out-end DI/III SDMD2?~yes
mid-out-end DI/III flavis=S/T YES
mid-out-end DI/III
out-up DI-DII interface
out-up DI-DII interface
up-out-top "end" Dili
up-out-top "end" Dili
tip-top-mid Dili
tip-top-mid Dili
tip-top-mid Dili
"RGD" loop up-out-lat
"ROD" loop up-out-lat
"RGD" loop up-out-lat
* not identified as > 35% SA in this particular structure/model
B-f: p-factor (temperature factor) a qualitative assessment of the scale (5-60A2).
SC?: is the amino acid side chain accessible and available for antibody binding
Ep?: might this amino acid be incorporated into an antigen epitope?
DVc: DENY 1-4 complex;
Jec: JE complex (medically important clade =JE, MVE, WN, SLE)
SDM: site-directed mutagenesis
Table 8. Potential JEV complex- and sub-complex cross-reactive epitope residues from WNV, with residues
chosen for mutagenesis highlighted
D2# SLE# WN# Pom. B-f. Location? SC? Conservation ED?
top inner edge near N67 D2
out-lateral-low-mid.
outer lateral middle
out-up-lat. loop near FP
out-mid-lat.
out-up-lat
up-mid-top near N67 in D2
up-mid-top
out mid lat DI/II border
up-mid-top below CHO-153
up-mid-top CHO on D2-153
up-mid-top near CHO 153 yes
up-in-top near CHO & prM
up-mid-top near prM
up-mid-top high.exp.
DI/II border out-mid
DI/II border up-out-top edge
DI/III border outer edge yes
DI/III up-out edge
DI/III up-out edge
up-out-mid. SDM?
prM hole low but above P243
prM hole low, above 243/244
up-top-in good aa JEc YES
out-up lat DI/III face
mid-out-end DI/III
mid-out-end DI/III
up-out-top "end" Dili
tip-top-mid Dili YES
tip-top-mid Dili YES
"ROD" loop up-out-lat yes
"ROD" loop up-out-lat ~y/na
-yes WN, SLE=L ?yes
yes "all" mosq.+ticks ?
no variable, Q=DVc ?
yes variable type? Yes
-yes JEc=A(SLE=S) yes
-no? variable ?possible
yes JEc=K, SDM in WN YES
yes var. yes
-yes var. L=WN,SLE ?/YES
yes H=DVc; yes if
no CHO
.yes "allmosq"=N YES
Y=JEc :SDMinWN YES
yes Q=Jec, SDMinWN YES
yes DVc=TJEc=I/V YES
yes JEc=R, except JE=K yes
yes P=mosq. (D2,3=S) yes?
yes DVc=E, JEc=T yes
var. JEc=K (SLE=N) yes?
yes DVc=var, JEc=G yes
yes "all"mosq=neg D2=G yes
yes DVc, JEc= T yes?
yes all flavis no?
yes "all" flavis no?
JEc=S, SDMWN YES
-yes flavis=K yes?
-yes DVc=M JEc=T ?pos?
YES flavis=S/T YES
YES JEc=T ex JE=S YES
JEc=A SDMinWN YES
JEc=N SDMiaWN YES
JEc=R YES
mosq=G or E YES
* not identified as > 35% SA in this particular structure/model
B-f: p-factor (temperature factor) a qualitative assessment of the scale (5-60A2).
SC?: is the amino acid side chain accessible and available for antibody binding
Ep?: might this amino acid be incorporated into an antigen epitope?
DVc: DENV1-4 complex;
Jec: JE complex (medically important clade =JE, MVE, WN, SLE)
SDM: site-directed mutagenesis
Table 9. Potential JEV complex- and sub-complex cross-reactive epitope residues from SLEV, with residues
chosen for mutagenesis highlighted
D2# SLE# WN# Pom. B-f. Location? SC? Conservation Ep?
T68* L68 L68 DII low top inner edge near N67 D2
T76 T76 T76 DII low out-lateral-low-mid.
Q77 T77* M77 DII med outer lateral middle
N83 T83 D83 DII med out-up-lat. loop near FP
Q86 S86 A86 DII high out-mid-lat.
K88 P88 P88 DII high out-up-lat
K122 K122 SI 22 DII med+ up-mid-top
Q131 L131 L131 DII/DI high out low lat mid
H149* S149 V149 DI high up-mid-top below CHO-153
N153* N154 N154 DI med+ up-mid-top CHO on D2-153
NA* Y155 Y155 DI na up-mid-top near CHO 153 yes
D154 Q158 Q158 DI high up-in-top near CHO & prM
T155* 1159* V159 DI high up-mid-top near prM
E161 R166 R166 DI high up-mid-top high.exp.
SI69 PI74 PI74 DI med+ DI/II border out-mid
E172 T177 T177 DI high DI/II border up-out-top edge
El74 N179 K179 DI high DI/III border outer edge
T176 GI81 G181 DI med DI/III up-out edge
G177* El82 El82 DI med DI/III up-out edge
T226 T231 T231 DII higli up-out-mid. SDM prev.
H244* H246 H246 DII med prM hole low but above P243
K247 K249 K249 DII med prM hole low, above 243/244
S274 S276 S276 DII/I high up-top-in good aa JEc YES
K291 K294 K294 DI med+ out-up lat DI/III face
M297 T300 T300 DIII/I high mid-out-end DI/III
S298 T301 T301 DIII/I med+ mid-out-end DI/III
E327* T330 T330 Dili high up-out-top "end" Dili
K361 A367 A367 Dili high tip-top-mid Dili
D362 N368 N368 Dili med tip-top-mid Dili
V382* R388 R388 Dili low "ROD" loop up-out-lat
E383 G389* G389 Dili high "ROD" loop up-out-lat
-yes WN,SLE=L ?yes
yes "all" mosq. + ticks ?
no variable, Q=DVc ?
yes variable type? Yes
-yes JEc=A(SLE=S) yes
-no? variable ?possible
yes var. yes
-yes var. L=WN,SLE ?/no
yes H=DVc; yes if
no CHO
yes "all mosq"=N YES
Y=JEc SDM SLE YES
yes Q=Jec, SDM SLE YES
yes DVc=TJEc=I/V YES
yes JEc=R, except JE=K yes
yes P=mosq. (D2,3=S) yes?
yes DVc=E,JEc=T YES
yes var. JEc=K (SLE=N) yes?
yes DVc=var, JEc=G yes
yes "all"inosq=neg D2=G yes
yes DVc,JEc=T
all flavis
"all" flavis
SDM SLE
flavis=K
DVc=M JEc=T
flavis=S/T
JEc=T ex JE=S
YES JEc=A, SDM in SLE YES
YES JEc=N, SDM in SLE YES
-yes JEc=R YES
~y/na mosq=G or E YES
as > 35% SA in this particular structure/model
B-f: p-factor (temperature factor) a qualitative assessment of the scale (5-60A2).
SC?: is the amino acid side chain accessible and available for antibody binding
Ep?: might this amino acid be incorporated into an antigen epitope?
DVc: DENV1-4 complex;
Jec: JE complex (medically important clade =JE, MVE, WN, SLE)
SDM: site-directed mutagenesis
Table 10. Inverse Iog10 end-point tilers of anli-SLEV mAbs determined by Ihe AG-ELISA for antigens expressed by wild-type pCB8SJ2 and cross-reactive reduced
mutated conslrucls
Mabs: MHIAF 4G2 T-23-1 T-23-2 6B6C-1 2B6B-2 4A1B-9 1B7-S 2BSB-3 T-16 6B4A-10 1B5D-1
supr supr JE JEV +
6B5A-2 4A4C-4 IB2C-5
Shaded block: Significantly altered endpoints relative to pCB8SJ2 derived wild-type VLP antigens. Most substitutions reduced mAb reactivity, however, some mAbs
reactivity increased.
Table 11. Inverse logic end-point titers of anti-WNV mAbs determined by the AG-ELISA for antigens expressed by wild-type pCBWN and cross-reactive reduced
mutated constructs
Mutants
Shaded block: Significantly altered endpoints relative to pCB8SJ2 derived wild-type VLP antigens. Most substitutions reduced mAb reactivity, however, some mAbs
reactivity increased.
Table 12. Comparative detection of human IgM antibody by MAC-ELISA with wild type (wt-) and
G106V- prototype type-specific antigens.
Serum Specimen Description
Infecting Virii
' Sera were assigned to one of three classes; positive, equivocal, or misleading as described in
materials and methods. Assignments were based upon previously determined P/N ratios3 reported
by the Diagnostics and Reference Laboratory, Arbovirus Diseases Branch, Division of Vector-
Borne Diseases, US Centers for Disease Control and Prevention.
2 PRNT90, Plaque reduction neutralization test; tilers represent inverse 90% plaque reduction
endpoints as reported by the Diagnostics and Reference Laboratory, ADD, DVBID, CDC.
3 Values represent ratios calculated as described in Materials and Methods. Positive ratios >3.0 are
shown in bold
4 Ratios reported by the Diagnostics and Reference Laboratory, ADB, DVBID, CDC.
5 Ratios determined in this study comparing wild-type (wt-) WNV Ag. with prototype cross-reactivity
reduced G106V-WNV Ag.
68
Table 13. Type-specific neutralizing antibody tilers as determined by PRNT
Plasmid DNA used for
immunization1

'Mice were immunized intramuscularly with lOOug of plasmid DNA on weeks 0 and 3.
2PRNT plaque reduction neutralization tesl, 75% neulralizalion endpoint tilers on mouse
sera collected 6 weeks post vaccination.





We Claim:
1. An isolated mutant flavivirus E-glycoprotein polypeptide that exhibits measurably reduced antibody cross-reactivity as compared to the corresponding wild-type flavivirus E-glycoprotein, said mutant polypeptide comprising at least one amino acid substitution at position 104, 106, or 107 as compared to said corresponding wild-type flavivirus E-glycoprotein polypeptide.
2. The isolated mutant flavivirus E-glycoprotein polypeptide as claimed in claim 1, wherein the amino acid substitution is selected from the group consisting of G104H, G104N, G106Q, G106V, L107K, L107Y, and a combination of two or more thereof.
3. The isolated mutant flavivirus E-glycoprotein polypeptide as claimed in claim 1, comprising an amino acid sequence as shown in SEQ ID NO: 14, having at least one amino acid substitution at position 104, 106, or 107 compared to a wild-type flavivirus E-glycoprotein polypeptide.
4. The isolated mutant flavivirus E-glycoprotein polypeptide as claimed in claim 2, wherein the amino acid substitution is selected from the group consisting of:
G104H; G106Q; L107K; and a combination of two or more thereof.
5. An isolated nucleic acid molecule encoding a polypeptide as claimed in claim 1.
6. A recombinant nucleic acid molecule, comprising a regulatory sequence operably linked to the nucleic acid molecule as claimed in claim 5.
7. A virus-like particle, comprising the polypeptide as claimed in claim 1.

Documents:

1310-DELNP-2007-Abstract-(02-05-2012).pdf

1310-delnp-2007-Abstract-(10-08-2011).pdf

1310-delnp-2007-abstract.pdf

1310-DELNP-2007-Claims-(02-05-2012).pdf

1310-delnp-2007-Claims-(10-08-2011).pdf

1310-delnp-2007-claims.pdf

1310-DELNP-2007-Correspondence Others-(02-05-2012).pdf

1310-delnp-2007-Correspondence Others-(10-08-2011).pdf

1310-delnp-2007-correspondence-ohers-1.pdf

1310-DELNP-2007-Correspondence-Others.pdf

1310-delnp-2007-Description (Complete)-(10-08-2011).pdf

1310-delnp-2007-description (complete).pdf

1310-delnp-2007-Drawings-(10-08-2011).pdf

1310-delnp-2007-drawings.pdf

1310-DELNP-2007-Form-1-(02-05-2012).pdf

1310-delnp-2007-Form-1-(10-08-2011).pdf

1310-DELNP-2007-Form-1.pdf

1310-delnp-2007-form-18.pdf

1310-DELNP-2007-Form-2-(02-05-2012).pdf

1310-delnp-2007-Form-2-(10-08-2011).pdf

1310-delnp-2007-form-2.pdf

1310-DELNP-2007-Form-3-(02-05-2012).pdf

1310-delnp-2007-Form-3-(10-08-2011).pdf

1310-DELNP-2007-Form-3.pdf

1310-delnp-2007-form-5.pdf

1310-delnp-2007-GPA-(10-08-2011).pdf

1310-DELNP-2007-GPA.pdf

1310-delnp-2007-pct-210.pdf

1310-delnp-2007-pct-220.pdf

1310-DELNP-2007-PCT-237.pdf

1310-delnp-2007-pct-304.pdf

1310-delnp-2007-pct-326.pdf

1310-delnp-2007-pct-373.pdf

1310-delnp-2007-Petition-137-(10-08-2011).pdf


Patent Number 253277
Indian Patent Application Number 1310/DELNP/2007
PG Journal Number 28/2012
Publication Date 13-Jul-2012
Grant Date 10-Jul-2012
Date of Filing 19-Feb-2007
Name of Patentee THE GOVERNMENT OF THE UNITED STATES OF AMERICA AS REPRESENTED BY THE SECRETARY OF THE DEPARTMENT OF HEALTH AND HUMAN SERVICES
Applicant Address TECHNOLOGY TRANSFER OFFICE, 4770 BUFORD HIGHWAY (K79) ATLANTA, GA 30341, USA
Inventors:
# Inventor's Name Inventor's Address
1 GWONGJEN J. CHANG 4237 BEAVER CREEK DRIVE, FT. COLLINS, CO 80526, USA
2 WAYNE D. CRILL 1121 AKIN AVENUE, FORT COLLINS, CO 80521, USA
PCT International Classification Number C12N 15/10, A61K 39/12
PCT International Application Number PCT/US2005/026672
PCT International Filing date 2005-07-27
PCT Conventions:
# PCT Application Number Date of Convention Priority Country
1 60/591,898 2004-07-27 U.S.A.