ATTENUATING VIRAL MUTATIONS IN PROTEIN GENES

Information

  • Patent Application
  • 20220354942
  • Publication Number
    20220354942
  • Date Filed
    May 05, 2022
    2 years ago
  • Date Published
    November 10, 2022
    a year ago
Abstract
Aspects of the disclosure relate to attenuated flaviviruses for use in vaccines and immunogenic compositions. Embodiments include methods for treating or preventing one or more conditions, for example flavivirus infection, using an attenuated flavivirus. In some embodiments, the disclosed methods and compositions involve one or more attenuated flaviviruses that are capable of inducing a protective immune response.
Description
SEQUENCE LISTING

A sequence listing required by 37 CFR 1.821-1.825 is being submitted electronically with this application. The sequence listing is incorporated herein by reference. The sequence listing that is contained in the file named “UTMBP0400US” which is 329.1 KB (as measured in Microsoft Windows®) and was created on Mar. 28, 2021 and modified on May 1, 2022.


FIELD OF THE INVENTION

The invention relates to compositions that elicit an immunological response against flavivirus infections and the clinical manifestations thereof, useful for the prevention and/or treatment of flavivirus infections in human and animal subjects.


BACKGROUND

West Nile virus (WNV) is a mosquito-borne flavivirus that is endemic in many parts of the world [1]. WNV was introduced into the United States (US) in 1999 and shortly thereafter spread throughout all of North America [1,2]. WNV cases are asymptomatic or manifest as WNV fever with symptoms such as fatigue, myalgia, arthralgia, or rash [3]. While most people recover completely from WNV fever, WNV is also capable of causing neurological disease, which may result in meningitis, encephalitis, acute flaccid paralysis, or other long-lasting neurological sequelae [3]. Approximately one of every 150 WNV neuroinvasive disease (WNND) cases are fatal [3]. Between 2002-2019, annual WNND reports to the US CDC ranged between 486-2946 with 43-284 deaths annually [4]. Since flaviviruses, such as WNV and yellow fever virus (YFV), are arthropod-borne and cannot be eradicated, there is a need for methods and compositions to protect humans from flaviviruses, for example, WNV and YFV.


SUMMARY

Aspects of the present disclosure address needs in the art by providing methods and compositions for treating and preventing flavivirus infection in subjects, human and animal. Certain embodiments are directed to methods for inducing an immune response in a subject comprising administering an effective amount of an immunogenic composition comprising an attenuated flavivirus and a pharmaceutically acceptable carrier or diluent to the subject. Also provided herein, in some aspects, are methods of immunizing a subject against a flavivirus infection comprising administering an effective amount of a vaccine composition comprising an attenuated flavivirus and a pharmaceutically acceptable carrier or diluent. In some embodiments, the disclosed methods comprise providing the immunogenic composition or vaccine composition to a subject who does not have, but is at risk of developing, infection by a flavivirus. In certain aspects a subject is at risk of infection by flavivirus by being in a geographic area in which a flavivirus is present. In some embodiments, the disclosed methods comprise providing the immunogenic composition to a subject who is infected by a flavivirus, i.e., therapeutically administering the immunogenic composition.


Embodiments include compositions comprising an attenuated flavivirus. In certain aspects, an attenuated flavivirus comprises one or more mutations in a nucleic acid construct encoding the attenuated flavivirus. Embodiments also include nucleic acid molecules encoding for all or part of the genome of an attenuated flavivirus. Embodiments include recombinant, transformed, or modified cells, vectors, and/or expression cassettes comprising such nucleic acid molecules. In certain aspects the flavivirus is West Nile Virus, Yellow Fever Virus, or Dengue virus (e.g., Dengue virus-4, accession MW793460.1, the nucleotide sequence and encoded protein sequence is incorporated herein by reference as of the filing date of this application).


In some embodiments, the compositions contemplated herein can comprise 1, 2, 3, 4, 5, or more of the following components: an attenuated flavivirus, a nucleic acid, a vector, a cell, a polypeptide, an oligonucleotide, a capsid (C) protein, a membrane (M) protein, an envelope (E) protein, or one or more non-structural (NS) proteins (NS1, NS2A, NS2B, NS3, NS4A, NS4B, and NS5). Any one or more of these components may be excluded from the disclosed compositions.


Embodiments of the disclosure include methods and compositions for treating or preventing a flavivirus infection in a subject, methods for diagnosing a flavivirus infection in a subject, methods for prognosing a flavivirus infection in a subject, and methods for identifying a subject having or at risk of having a flavivirus infection as a candidate for an attenuated flavivirus prophylactic or therapy. Methods of the disclosure can include 1, 2, 3, 4, 5, 6, or more of the following steps: providing an attenuated flavivirus prophylactic or therapy to a subject, providing a second antiviral therapy to a subject, providing both an attenuated flavivirus prophylactic or therapy and a second antiviral to a subject, providing an alternative therapy to a subject, determining a subject to have a flavivirus infection, providing two or more types of antiviral therapy to a subject, and identifying a subject as being a candidate for an attenuated flavivirus prophylactic or therapy. Certain embodiments of the disclosure may exclude one or more of the preceding elements and/or steps.


Embodiments also include, inter alia, methods of generating an attenuated flavivirus prophylactic or therapy, methods of producing an attenuated flavivirus prophylactic or therapy, methods of expressing an attenuated flavivirus, methods of detecting flavivirus infection, methods of treating one or more conditions, methods of purifying an attenuated flavivirus, and methods of treating flavivirus infection. The steps and embodiments discussed in this disclosure are contemplated as part of any of these methods. In some embodiments, the methods contemplated herein can comprise or exclude 1, 2, 3, 4, 5, or more of the following steps: providing an attenuated flavivirus, providing a nucleic acid to a cell, subjecting a cell to conditions sufficient to express a nucleic acid, providing an additional therapeutic, expressing a vector in a cell, and providing a pharmaceutical composition to a subject. Any one or more of these steps may be excluded from the disclosed methods.


Embodiments of the present disclosure include an attenuated flavivirus as represented by a West Nile Virus (WNV) wherein a nucleic acid construct encoding the genome of the attenuated flavivirus comprises one or more mutations corresponding to a mutation in the transmembrane domain of non-structural protein 4B (NS4B) of the flavivirus, wherein the one or more mutations result in a substitution of wild-type amino acid 54 present in a transmembrane domain of NS4B. NS4B protein is encoded, for example, by nucleotides 6916 to 7680 of SEQ ID NO:1. NS4B protein can also be identified as amino acid 2274 to 2519 of the polyprotein sequence of SEQ ID NO:2. NS4B region and NS4B protein can be readily identified in other flavivirus by nucleic acid and protein alignment. SEQ ID NO:19 provides the amino acid sequence of wild-type or non-attenuated WNV NS4B protein. In particular aspects, an attenuated flavivirus comprises a mutation in NS4B protein that corresponds to proline 54 of SEQ ID NO:19. In some embodiments, the substitution of wild-type amino acid 54 comprises a substitution of a proline residue with a nonpolar amino acid. In some embodiments, the nonpolar amino acid comprises alanine residue (P54A) or glycine residue (P54G).


In some embodiments, the nucleic acid construct encoding the genome of the attenuated WNV further comprises one or more mutations corresponding to one or more glycosylation sites of non-structural protein 1 (NS1) of the flavivirus. In some embodiments, the one or more mutations corresponding to one or more glycosylation sites of NS1 of the flavivirus result in a substitution of wild-type amino acids 130-132 of NS1. In some embodiments, the substitution of wild-type amino acids 130 and 131 comprises a substitution of an asparagine residue with a polar amino acid. In some embodiments, the polar amino acid comprises a glutamine residue. In some embodiments, the substitution of amino acid 132 comprises a substitution of a threonine residue with a nonpolar amino acid. In some embodiments, the nonpolar amino acid comprises an alanine residue. In some embodiments, the one or more mutations corresponding to one or more glycosylation sites of NS1 of the flavivirus further comprise a substitution of wild-type amino acid 175 of NS1. In some embodiments, the one or more mutations corresponding to one or more glycosylation sites of NS1 of the flavivirus further comprise a substitution of wild-type amino acid 207 of NS1. In some embodiments, the substitution of wild-type amino acid 175 or wild-type amino acid 207 comprises a substitution of an asparagine residue with a nonpolar amino acid. In some embodiments, the nonpolar amino acid comprises an alanine residue.


Embodiments of the present disclosure also include, inter alia, an attenuated flavivirus comprising a flavivirus wherein a nucleic acid construct encoding the genome of the attenuated flavivirus comprises one or more mutations corresponding to a transmembrane domain of non-structural protein 4B (NS4B) of the flavivirus, wherein the one or more mutations comprise a substitution of a wild-type amino acid homologous to wild-type amino acid 54 present in a transmembrane domain of WNV NS4B. In some embodiments, the flavivirus is selected from the group consisting of WNV, Japanese encephalitis virus, St. Louis encephalitis virus, tickborne encephalitis virus, dengue fever virus, Zika virus, and yellow fever virus (YFV) because a proline is found at the equivalent residue in all mosquito- and tick-borne flaviviruses sequenced to date where WNV is residue 54. For example, in some embodiments, the flavivirus is YFV. In some embodiments, the YFV wild-type amino acid homologous to wild-type amino acid 54 of the transmembrane domain of WNV NS4B is amino acid 52, and wherein substitution of amino acid 52 comprises a substitution of a proline residue with a nonpolar amino acid. In some embodiments, the nonpolar amino acid comprises alanine residue or glycine residue. In some embodiments, the nucleic acid construct encoding the genome of the attenuated flavivirus further comprises one or more mutations corresponding to one or more glycosylation sites of non-structural protein 1 (NS1) of the flavivirus.


In some embodiments, the one or more mutations of the attenuated flaviviruses decrease the neurotropism of the flavivirus, relative to a corresponding flavivirus lacking the mutation. In some embodiments, the one or more mutations decrease the viscerotropism of the flavivirus, relative to a corresponding flavivirus lacking the mutation. In some embodiments, the one or more mutations decrease cytokine response to the flavivirus, relative to a corresponding flavivirus lacking the mutation. In some embodiments, reversion of the one or more mutations to the wild-type amino acid is inhibited when the virus is grown in a host. In some embodiments, the virus induces an immune response when administered to or infecting a human or an animal host.


In some aspects, the disclosure relates to an immunogenic composition comprising an attenuated flavivirus described herein and a pharmaceutically acceptable carrier or diluent. In some embodiments, the immunogenic composition induces an immune response in a subject that is equivalent to an immune response induced by a corresponding wild type virus.


In some aspects, the disclosure relates to a method of decreasing viscerotropism of a flavivirus in a subject comprising administering an effective amount of the immunogenic composition to the subject. In some aspects, the disclosure relates to a method of decreasing neurotropism of a flavivirus in a subject comprising administering an effective amount of the immunogenic composition to the subject. In some aspects, the disclosure relates to a method of inducing an immune response in a subject comprising administering an effective amount of the immunogenic composition to the subject. In some embodiments, the subject does not have, but is at risk of developing, infection by a flavivirus. In some embodiments, the subject is infected by a flavivirus.


In some embodiments, the flavivirus is selected from the group of mosquito-borne and tick-borne flaviviruses. In certain aspects, the flavivirus is selected from the group consisting of WNV, Japanese encephalitis virus, St. Louis encephalitis virus, tickborne encephalitis virus, dengue fever virus, and YFV. In some embodiments, the flavivirus is WNV. In some embodiments, the flavivirus is YFV. In some embodiments, an immune response to at least one of WNV, Japanese encephalitis virus, St. Louis encephalitis virus, tickborne encephalitis virus, dengue fever virus, and YFV is induced in the subject. In some embodiments, the immune response induced in the subject is to WNV. In some embodiments, the immune response induced in the subject is to YFV.


In some aspects, the disclosure relates to a vaccine composition comprising an attenuated flavivirus described herein and a pharmaceutically acceptable carrier or diluent. In some aspects, the disclosure relates to a method of immunizing a subject against a flavivirus infection comprising administering an effective amount of the vaccine composition to the subject. In some embodiments, the subject does not have, but is at risk of developing, infection by a flavivirus. In some embodiments, the flavivirus is selected from the group of mosquito-borne and tick-borne flaviviruses. In certain aspects, the flavivirus is selected from the group consisting of WNV, Japanese encephalitis virus, St. Louis encephalitis virus, tickborne encephalitis virus, dengue fever virus, and YFV. In some embodiments, the flavivirus is WNV. In some embodiments, the flavivirus is YFV.


In some embodiments, the subjects disclosed herein are non-human primates, humans, horses, or birds. In some embodiments, the compositions are administered to the subject subcutaneously, intramuscularly, intranasally, orally, topically, transdermally, parenterally, gastrointestinally, transbronchially or transalveolarly. In some embodiments, the compositions are administered to the subject as a single dose or in multiple doses. In some embodiments, the compositions are administered to the subject as a single composition followed by a boost of the same or different composition.


In some aspects, the disclosure relates to a method of producing a vaccine against an attenuated flavivirus comprising WNV, the method comprising introducing into a nucleic acid construct encoding the genome of the attenuated WNV one or more mutations corresponding to a transmembrane domain of NS4B of the flavivirus, wherein the one or more mutations attenuates the flavivirus, relative to a corresponding flavivirus lacking the mutation, and further wherein the one or more mutations result in a substitution of wild-type amino acid 54 of the transmembrane domain of NS4B. In some embodiments, the substitution of wild-type amino acid 54 comprises a substitution of a proline residue with a nonpolar amino acid. In some embodiments, the nonpolar amino acid comprises alanine residue or glycine residue. In some embodiments, the method further comprises introducing into the nucleic acid construct encoding the genome of the attenuated WNV one or more additional mutations corresponding to one or more glycosylation sites of non-structural protein 1 (NS1) of the flavivirus. In some embodiments, the one or more mutations corresponding to one or more glycosylation sites of NS1 of the flavivirus result in a substitution of wild-type amino acids 130-132 of NS1. In some embodiments, the substitution of wild-type amino acids 130 and 131 comprises a substitution of an asparagine residue with a polar amino acid. In some embodiments, the polar amino acid comprises a glutamine residue. In some embodiments, the substitution of amino acid 132 comprises a substitution of a threonine residue with a nonpolar amino acid. In some embodiments, the nonpolar amino acid comprises an alanine residue.


In some aspects, the disclosure relates to a method of producing a vaccine against a flavivirus, the method comprising introducing into a nucleic acid construct encoding the genome of the attenuated flavivirus one or more mutations corresponding to a transmembrane domain of NS4B of the flavivirus, wherein the one or more mutations attenuates the flavivirus, relative to a corresponding flavivirus lacking the mutation, and further wherein the one or more mutations comprise a substitution of a wild-type amino acid homologous to wild-type amino acid 54 of the transmembrane domain of WNV NS4B. In some embodiments, the flavivirus is selected from the group consisting of WNV, Japanese encephalitis virus, St. Louis encephalitis virus, tickborne encephalitis virus, dengue fever virus, and YFV. In some embodiments, the flavivirus is YFV. In some embodiments, the YFV wild-type amino acid homologous to wild-type amino acid 54 of the transmembrane domain of WNV NS4B comprises amino acid 52, and wherein substitution of amino acid 52 comprises a substitution of a proline residue with a nonpolar amino acid. In some embodiments, the method further comprises introducing into the nucleic acid construct encoding the genome of the attenuated flavivirus one or more additional mutations corresponding to one or more glycosylation sites of non-structural protein 1 (NS1) of the flavivirus.


In some aspects, the disclosure relates to a method of manufacturing an attenuated flavivirus disclosed herein, the method comprising introducing a nucleic acid construct encoding the genome of the attenuated flavivirus into cells and isolating flavivirus produced in the cells from the cells or the supernatant thereof. In some embodiments, the cells are Vero cells. In some embodiments, the cells are cultured in serum free medium.


Other embodiments of the disclosure are discussed throughout this application. Any embodiment discussed with respect to one aspect of the disclosure applies to other aspects of the disclosure as well and vice versa. Each embodiment described herein is understood to be embodiments of the disclosure that are applicable to all aspects of the disclosure. It is contemplated that any embodiment discussed herein can be implemented with respect to any method or composition of the disclosure, and vice versa. Furthermore, compositions and kits of the disclosure can be used to achieve methods of the disclosure.


The use of the word “a” or “an” when used in conjunction with the term “comprising” in the claims and/or the specification may mean “one,” but it is also consistent with the meaning of “one or more,” “at least one,” and “one or more than one.”


Throughout this application, the term “about” is used to indicate that a value includes the standard deviation of error for the device or method being employed to determine the value.


The use of the term “or” in the claims is used to mean “and/or” unless explicitly indicated to refer to alternatives only or the alternatives are mutually exclusive, although the disclosure supports a definition that refers to only alternatives and “and/or.”


As used in this specification and claim(s), the words “comprising” (and any form of comprising, such as “comprise” and “comprises”), “having” (and any form of having, such as “have” and “has”), “including” (and any form of including, such as “includes” and “include”) or “containing” (and any form of containing, such as “contains” and “contain”) are inclusive or open-ended and do not exclude additional, unrecited elements or method steps.


As used herein, the terms “comprises,” “comprising,” “includes,” “including,” “has,” “having,” “contains”, “containing,” “characterized by” or any other variation thereof, are intended to encompass a non-exclusive inclusion, subject to any limitation explicitly indicated otherwise, of the recited components. For example, a chemical composition and/or method that “comprises” a list of elements (e.g., components or features or steps) is not necessarily limited to only those elements (or components or features or steps), but may include other elements (or components or features or steps) not expressly listed or inherent to the chemical composition and/or method.


As used herein, the transitional phrases “consists of” and “consisting of” exclude any element, step, or component not specified. For example, “consists of” or “consisting of” used in a claim would limit the claim to the components, materials or steps specifically recited in the claim except for impurities ordinarily associated therewith (i.e., impurities within a given component). When the phrase “consists of” or “consisting of” appears in a clause of the body of a claim, rather than immediately following the preamble, the phrase “consists of” or “consisting of” limits only the elements (or components or steps) set forth in that clause; other elements (or components) are not excluded from the claim as a whole.


As used herein, the transitional phrases “consists essentially of” and “consisting essentially of” are used to define a chemical composition and/or method that includes materials, steps, features, components, or elements, in addition to those literally disclosed, provided that these additional materials, steps, features, components, or elements do not materially affect the basic and novel characteristic(s) of the disclosure. The term “consisting essentially of” occupies a middle ground between “comprising” and “consisting of”.


Any method in the context of a therapeutic, diagnostic, or physiologic purpose or effect may also be described in “use” claim language such as “Use of” any compound, composition, or agent discussed herein for achieving or implementing a described therapeutic, diagnostic, or physiologic purpose or effect.


Other objects, features and advantages of the present disclosure will become apparent from the following detailed description. It should be understood, however, that the detailed description and the specific examples, while indicating specific embodiments of the disclosure, are given by way of illustration only, since various changes and modifications within the spirit and scope of the disclosure will become apparent to those skilled in the art from this detailed description.





DESCRIPTION OF THE DRAWINGS

The following drawings form part of the present specification and are included to further demonstrate certain aspects of the present disclosure. The disclosure may be better understood by reference to one or more of these drawings in combination with the detailed description of the specification embodiments presented herein.



FIG. 1. NS4B residue P54 is conserved amongst multiple flaviviruses. The highlighted residue is the specific amino acid characterized by mutagenesis and correspond to WNV NS4B residue P54. The alignment was generated using Clustal Omega and virus abbreviations and Genbank accession numbers are as follows: WNV: West Nile virus (AAF20092.2); KUNV: Kunjin virus (BAA00176.1); JEV: Japanese encephalitis virus (ABQ52691.1); SLEV: Saint Louis encephalitis virus (ACT31738.1); ZIKV: zika virus (AMR39836.1); DENV-1: dengue virus 1 (AIU47321.1); DENV-2: dengue virus 2 (AAC59275.1); DENV-3: dengue virus 3 (ALS05358.1); DENV-4: dengue virus 4 (ALB78116.1); YFV: YFV (AHB63685.1); POWV: powassan virus (NP_620099.1); TBEV: tick-borne encephalitis virus (AAA86870.1); LGTV: langat virus (ACH42698.1); OFFV: Omsk hemorrhagic fever virus (NP_878909.1).



FIG. 2. Location of P54 amino acid residue within the predicted structure of NS4B. Each alpha helix is labeled as α1-α9 and locations are based on NMR studies of DENV NS4B. Residue P54 was investigated in the present disclosure. Residue P54 is predicted to be within α2, which is the first transmembrane domain.



FIGS. 3A-3B. Multiplication kinetics are similar for NY99ic and WNV NS4B mutants. Vero cells (FIG. 3A) and A549 cells (FIG. 3B) were infected with a multiplicity of infection of 0.1 of each virus. Two biological replicates were infected and two samples from each flask were titrated at the time points indicated.



FIG. 4A-4C Attenuated WNV NS4B mutants exhibited two different patterns of cytokine induction. Cytokine levels in NS4B mutant infected cells were compared to NY99ic infected cells using a Kruskal-Wallis ANOVA with Dunn's post-hoc correction. A WNV NS4B-P38G attenuated mutant that was previously characterized was included as an attenuated control. Six replicates of each mutant and 12 replicates of NY99ic and mock were measured, except for IFN-α and IFN-β for which six replicates were tested for all viruses and controls. *p <0.05, **p <0.01, ***p <0.001, ****p <0.0001. FIG. 4A illustrating IL-1ra, IL-2, IL-6, IL-7, IL-4, IL-5, CXCL8 (IL-8), and IL-9; FIG. 4B illustrating IL-17, Eotaxin, GM-CSF, IFN-γ, FGF, G-CSF, CXCL 10 (IP-10), and CCL2 (MCP-1); and FIG. 4C illustrating CCL3 (MIP 1α), CCL4 (MIP 1(3), VEGF, IFN-α2, CCL5 (RANTES), TNF-α, and IFN-β.



FIG. 5. Each NS4B-P54 mutant has a unique SNV profile. For Vero cell P1 virus stocks, the frequency of each SNV is indicated by an (x), and the grey bars in the background display the total number of SNVs detected.



FIG. 6. WNV NS4B-P54 mutants had unique SNV frequency and genomic distribution. Each graph displays the SNVs ≥1% frequency that were detected in the Vero cell P1 virus stocks of each virus.



FIG. 7. The NS4B-P54A mutant had SNVs clustered in the NS4B protein, including one that encoded reversion. SNVs ≥1% frequency in the NS4B protein of the NS4B-P54A mutant are mapped to the corresponding genomic position. The NS4B amino acid substitutions are indicated above each line.



FIGS. 8A-8C. The WNV P54G mutant had reduced NS4B accumulation and NS1-NS4B colocalization in infected cells one day post infection. Vero cells were infected with a MOI of 0.1 of NY99ic, the NS4B-P54G mutant, a WNV NS1 glycosylation site mutant, or PBS as a mock infection, and fluorescence microscopy was performed one day post infection (FIG. 8A). Mean fluorescence intensity (FIG. 8B) and Pearson's correlation coefficient (FIG. 8C) were calculated on individual infected cells (n=18). Statistical difference in mean fluorescence intensity and colocalization were measured using a Kruskal-Wallis test with multiple comparisons to compare both mutants to NY99ic. * p <0.05, ** p <0.01, *** p <0.001.



FIGS. 9A-9C. Attenuated WNV NS4B-P54G mutant exhibited lower levels of both NS4B and NS1 two days post infection. Vero cells were infected with a MOI of 0.1 of NY99ic, the NS4B-P54G mutant, a WNV NS1 glycosylation site mutant, or PBS as a mock infection, and fluorescence microscopy was performed two days post infection (FIG. 9A). Mean fluorescence intensity (FIG. 9B) and Pearson's correlation coefficient (FIG. 9B) were calculated on individual infected cells (n=19). Statistical difference in mean fluorescence intensity and colocalization were measured using a Kruskal-Wallis test with multiple comparisons to compare both mutants to NY99ic. * p <0.05, ** p <0.01.



FIGS. 10A-10F. YFV NS4B-P52G mutation resulted in reduced quasispecies diversity compared to the parental Asibi strain. Shannon entropy (FIGS. 10A-10C) and SNV frequency (FIGS. 10D-10F) were utilized as two independent measurements of genomic diversity in the P0 unpassaged stocks of 17Dic, Asibi ic, and Asibi ic NS4B-P52G. YFV NS4B-P52G is homologous to WNV NS4B-P54G.





DESCRIPTION

The present disclosure is based, at least in part, on the discovery that flavivirus infections can be associated with neuroinvasion, neurotropism, and/or viscerotropism, and administration of an immunogenic or vaccine composition comprising an attenuated flavivirus disclosed herein has been surprising and unexpectedly found to prevent or decrease neuroinvasion. Further, administering an immunogenic or vaccine composition comprising an attenuated flavivirus disclosed herein can surprisingly prevent or decrease cytokine induction in infected cells.


The attenuated flaviviruses disclosed herein provide live, attenuated viruses useful as immunogens or vaccines. Live vaccines confer the most potent and durable, protective immune responses against disease caused by viral infections. In the case of flaviviruses, the development of a successful vaccine requires that the virulence properties are modified, for example, by mutation, so that the immunogenic composition or vaccine virus has reduced neurotropism and viscerotropism for humans or animals.


As disclosed herein, administration of a therapeutically effective amount of an immunogenic composition or vaccine comprising an attenuated flavivirus can prevent or decrease neuroinvasion, neurotropism, and/or viscerotropism. Accordingly, in some embodiments, disclosed are methods and compositions for treating or preventing flavivirus infection in a subject or inhibiting or decreasing neuroinvasion, neurotropism, and/or viscerotropism comprising administering a therapeutically effective amount of an immunogenic or vaccine composition comprising an attenuated flavivirus. In some embodiments, the flavivirus is selected from the group consisting of WNV, Japanese encephalitis virus, St. Louis encephalitis virus, tickborne encephalitis virus, dengue fever virus, and YFV. In some embodiments, the flavivirus is WNV. In some embodiments, the flavivirus is YFV. In some embodiments, the subject does not have, but is at risk of developing, infection by a flavivirus. In some embodiments, the subject has symptoms of a flavivirus infection. In some embodiments, the subject is infected by a flavivirus.


The following discussion is directed to various embodiments of the invention. The term “invention” is not intended to refer to any particular embodiment or otherwise limit the scope of the disclosure. Although one or more of these embodiments may be preferred, the embodiments disclosed should not be interpreted, or otherwise used, as limiting the scope of the disclosure, including the claims. In addition, one skilled in the art will understand that the following description has broad application, and the discussion of any embodiment is meant only to be exemplary of that embodiment, and not intended to intimate that the scope of the disclosure, including the claims, is limited to that embodiment. All publications mentioned herein are incorporated by reference for the purpose of describing and disclosing methodologies and materials that might be used in connection with the present disclosure.


I. FLAVIVIRUSES

Flaviviruses are small, enveloped, positive-strand RNA viruses that are generally transmitted by infected arthropods, such as mosquitoes and ticks. The Flavivirus genus of the Flaviviridae family includes approximately 70 viruses, mostly arboviruses, many of which, such as West Nile virus (WNV), yellow fever virus (YFV), dengue (DEN), Japanese encephalitis (JE), and tick-borne encephalitis (TBE) viruses, are major human pathogens (reviewed in Burke and Monath, Fields Virology, 4th Ed.: 1043-1126, 2001). For example, Japanese encephalitis is the leading cause of viral encephalitis in Asia, where 30,000 to 50,000 new cases are reported each year. As another example, since the first cases were diagnosed in the New York area in 1999, WNV has continued to spread rapidly across North America. The risks of this virus migrating into South America, as well as an epidemic in underdeveloped countries, are extremely high. Effective methods for preventing infection by these viruses are needed, with vaccination being the most cost-effective measure. In certain embodiments insect-specific flaviviruses (i.e., those that multiply in insects only and not vertebrate cells) can be specifically excluded.


A. West Nile Virus


WNV is a member of the Japanese encephalitis virus (JE) serocomplex of mosquito-borne flaviviruses that includes St. Louis encephalitis, JE, and Murray Valley encephalitis viruses (Calisher et al. 1989 J Gen Virol 70:27-43; Burke and Monath, 2001 in: Fields Virology, eds. Knipe and Howley, Lippincott Williams and Wilkins, Philadelphia, 4-th ed., pp. 1043-1125). Like other members of the JE antigenic complex, WNV is maintained in a natural cycle that involves mosquito vectors and birds, while humans and equines are usually incidental hosts. For many years WNV has been recognized as one of the most widely distributed flaviviruses with a geographic range including Africa, Australia, Europe, the Middle East and West Asia (Burke and Monath, 2001 in: Fields Virology, eds. Knipe, D. M. & Howley, P. M. Lippincott Williams and Wilkins, Philadelphia, 4-th ed., pp. 1043-1125; Hayes, 1989 in: The Arboviruses: Epidemiology and Ecology, ed. Monath Boca Raton, Fla. CRC Press, Volume V, pp. 59-88). During 1999 WNV first established itself in the USA in the Northeast and Mid-Atlantic States and more recently this virus extended its range to include the Southeastern and Western States (Anderson, J. F. et al. 1999 Science 286:2331-2333; Lanciotti, R. S. et al. 1999 Science 286:2333-2337; Campbell, G. L. et al. 2002 Lancet 2:519-529). The natural hosts for WNV are birds and mosquitoes. Over 300 different species of bird have been shown to be infected with the virus.


WNV has been demonstrated in a large number of mosquito species, but the most significant for viral transmission are Culex species that feed on birds, including C. pipiens, C. restuans, C. salinarius, C. quinquefasciatus, C. nigripalpus, C. erraticus and C. tarsalis. Experimental infection has also been demonstrated with soft tick vectors.


WNV has a broad host range, and is also known to be able to infect at least 30 mammalian species, including humans, some non-human primates, horses, dogs and cats. Some infected humans and horses experience disease but dogs and cats rarely show symptoms. Reptiles and amphibians can also be infected, including some species of crocodiles, alligators, snakes, lizards, and frogs. Mammals are considered incidental or dead-end hosts for the virus: they do not usually develop a high enough level of virus in the blood (viremia) to infect another mosquito feeding on them and carry on the transmission cycle.


According to the Center for Disease Control, infection with WNV is seasonal in temperate zones. Climates that are temperate, such as those in the United States and Europe, see peak season from July to October. Peak season changes depending on geographic region and warmer and humid climates can see longer peak seasons. All ages are equally likely to be infected but there is a higher amount of death and neuroinvasive WNV in people 60-89 years old. People of older age are more likely to have adverse effects of being infected.


In humans, WNV can cause a disease known as West Nile fever. According to the U.S. Centers for Disease Control and Prevention, approximately 80% of infected people have few or no symptoms, around 20% of people develop mild symptoms (such as fever, headache, vomiting, or a rash), and less than 1% of people develop severe symptoms (such as encephalitis, meningitis, or acute flaccid paralysis). WNV meningitis is clinically indistinguishable from viral meningitis due to other etiologies and typically presents with fever, headache, and nuchal rigidity. WNV encephalitis is a more severe clinical syndrome that usually manifests with fever and altered mental status, seizures, focal neurologic deficits, or movement disorders such as tremor or parkinsonism. WNV acute flaccid paralysis is usually clinically and pathologically identical to poliovirus-associated poliomyelitis, with damage of anterior horn cells, and may progress to respiratory paralysis requiring mechanical ventilation. WNV poliomyelitis often presents as isolated limb paresis or paralysis and can occur without fever or apparent viral prodrome. WNV-associated Guillain-Barré syndrome and radiculopathy have also been reported and can be distinguished from WNV poliomyelitis by clinical manifestations and electrophysiologic testing. Rarely, cardiac dysrhythmias, myocarditis, rhabdomyolysis, optic neuritis, uveitis, chorioretinitis, orchitis, pancreatitis, and hepatitis have been described in subjects with WNV disease.


In endemic regions, most human WNV infections are asymptomatic or cause mild illness with symptoms of low-grade fever, headache, body aches, rash, myalgia, and polyarthropathy. However, human epidemics with severe disease have been reported in Israel, France, Romania, and Russia. In acute severe illness, the virus can cause hepatitis, meningitis and encephalitis leading to paralysis, and coma resulting in death. The neuropathologic lesions are similar to those of JE, with diffuse CNS inflammation and neuronal degeneration. Virus is also found in the spleen, liver, lymph nodes, and lungs of infected individuals. During the 1999 outbreak of WNV in the USA, more than 60 people became ill and 7 died, while during 2002, morbidity was 3873 cases and there were 246 deaths (CDC Report: West Nile Update Current case Count, Jan. 2, 2003). Because of the recent and unexpected spread of WNV from the Northeast to the Southeast and the West of the USA, this virus is considered a significant emerging disease threat that has embedded itself over a considerable region of the country.


Laboratory diagnosis is generally accomplished by testing of serum or cerebrospinal fluid (CSF) to detect WNV-specific IgM antibodies. Immunoassays for WNV-specific IgM are available commercially and through state public health laboratories. WNV-specific IgM antibodies are usually detectable 3 to 8 days after onset of illness and persist for 30 to 90 days, but longer persistence has been documented. Therefore, positive IgM antibodies occasionally may reflect a past infection. If serum is collected within 8 days of illness onset, the absence of detectable virus-specific IgM does not rule out the diagnosis of WNV infection, and the test may need to be repeated on a later sample. The presence of WNV-specific IgM in blood or CSF provides good evidence of recent infection but may also result from cross-reactive antibodies after infection with other flaviviruses or from non-specific reactivity. According to product inserts for commercially available WNV IgM assays, all positive results obtained with these assays should be confirmed by neutralizing antibody testing of acute- and convalescent-phase serum specimens at a state public health laboratory or CDC. WNV IgG antibodies generally are detected shortly after IgM antibodies and persist for many years following a symptomatic or asymptomatic infection. Therefore, the presence of IgG antibodies alone is only evidence of previous infection and clinically compatible cases with the presence of IgG, but not IgM, should be evaluated for other etiologic agents. Plaque-reduction neutralization tests (PRNTs) performed in reference laboratories, including some state public health laboratories and CDC, can help determine the specific infecting flavivirus. PRNTs can also confirm acute infection by demonstrating a fourfold or greater change in WNV-specific neutralizing antibody titer between acute- and convalescent-phase serum samples collected 2 to 3 weeks apart. Viral cultures and tests to detect viral RNA (e.g., reverse transcriptase-polymerase chain reaction [RT-PCR]) can be performed on serum, CSF, and tissue specimens that are collected early in the course of illness and, if results are positive, can confirm an infection. Immunohistochemistry (IHC) can detect WNV antigen in formalin-fixed tissue. Negative results of these tests do not rule out WNV infection. Viral culture, RT-PCR, and IHC can be requested through state public health laboratories or CDC.


There is no specific treatment for WNV disease; clinical management is supportive. Subjects with severe meningeal symptoms often require pain control for headaches and antiemetic therapy and rehydration for associated nausea and vomiting. Subjects with encephalitis require close monitoring for the development of elevated intracranial pressure and seizures. Subjects with encephalitis or poliomyelitis should be monitored for inability to protect their airway. Acute neuromuscular respiratory failure may develop rapidly and prolonged ventilatory support may be required. Currently, no WNV vaccines are licensed for use in humans. In the absence of a vaccine, prevention of WNV disease depends on community-level mosquito control programs to reduce vector densities, personal protective measures to decrease exposure to infected mosquitoes, and screening of blood and organ donors. Personal protective measures include use of mosquito repellents, wearing long-sleeved shirts and long pants, and limiting outdoor exposure from dusk to dawn. Using air conditioning, installing window and door screens, and reducing peridomestic mosquito breeding sites, can further decrease the risk for WNV exposure.


B. Yellow Fever Virus (YFV)


Yellow fever is caused by YFV and is spread by the bite of an infected female mosquito. It infects only humans, other primates, and several types of mosquitoes. Yellow fever begins after an incubation period of three to six days. Most cases only cause a mild infection with fever, headache, chills, back pain, fatigue, loss of appetite, muscle pain, nausea, and vomiting. In these cases, the infection lasts only three to four days. In 15% of cases, people enter a second, toxic phase of the disease with recurring fever, this time accompanied by jaundice due to liver damage, as well as abdominal pain. Bleeding in the mouth, nose, the eyes, and the gastrointestinal tract cause vomit containing blood, hence the Spanish name for yellow fever, vómito negro (“black vomit”). There may also be kidney failure, hiccups, and delirium. Among those who develop jaundice, the fatality rate is 20 to 50%, while the overall fatality rate is about 3 to 7.5%. Severe cases may have a mortality greater than 50%.


Yellow fever belongs to the group of hemorrhagic fevers. After transmission from a mosquito, the viruses replicate in the lymph nodes and infect cells. The viruses infect, amongst others, monocytes, macrophages, Schwann cells, and dendritic cells. From there, they reach the liver and infect hepatocytes (probably indirectly via Kupffer cells), which leads to eosinophilic degradation of these cells and to the release of cytokines. Apoptotic masses known as Councilman bodies appear in the cytoplasm of hepatocytes. Fatality may occur when cytokine storm, shock, and multiple organ failure follow.


The viruses attach to the cell surfaces via specific receptors and are taken up by an endosomal vesicle. Receptor binding, as well as membrane fusion, are catalyzed by the protein E, which changes its conformation at low pH, causing a rearrangement of the 90 homodimers to 60 homotrimers. Inside the endosome, the decreased pH induces the fusion of the endosomal membrane with the virus envelope. The capsid enters the cytosol, decays, and releases the genome. After entering the host cell, the viral genome is replicated in the rough endoplasmic reticulum (ER) and in the so-called vesicle packets. At first, an immature form of the virus particle is produced inside the ER, whose M-protein is not yet cleaved to its mature form, so is denoted as precursor M (prM) and forms a complex with protein E. The immature particles are processed in the Golgi apparatus by the host protein furin, which cleaves prM to M. This releases E from the complex, which can now take its place in the mature, infectious virion.


Yellow fever virus is mainly transmitted through the bite of different Aedes species of mosquitoes in Africa and Hemagoggus and Sabethes species in South America. When YFV is found in urban areas, Aedes aegypti can be the mosquito vector. Like other arboviruses, which are transmitted by mosquitoes, YFV is taken up by a female mosquito when it ingests the blood of an infected human or another primate. Viruses reach the stomach of the mosquito, and if the virus concentration is high enough, the virions can infect epithelial cells and replicate there. From there, they reach the haemocoel (the blood system of mosquitoes) and from there the salivary glands. When the mosquito next sucks blood, it injects its saliva into the wound, and the virus reaches the bloodstream of the bitten person.


Three epidemiologically different infectious cycles occur in which the virus is transmitted from mosquitoes to humans or other primates. In the “urban cycle”, only the YFV mosquito Ae. aegypti is involved. It is well adapted to urban areas, and can also transmit other diseases, including Zika fever, dengue fever, and chikungunya. Besides the urban cycle, both in Africa and South America, a sylvatic cycle (forest or jungle cycle) is present, where Aedes africanus (in Africa) or mosquitoes of the genus Haemagogus and Sabethes (in South America) serve as vectors. In the jungle, the mosquitoes infect mainly nonhuman primates; the disease is mostly asymptomatic in African primates. People who become infected in the jungle can carry the virus to urban areas, where Ae. aegypti acts as a vector. Because of this sylvatic cycle, YFV cannot be eradicated except by eradicating the mosquitoes that serve as vectors. In Africa, a third infectious cycle known as “savannah cycle” or intermediate cycle, occurs between the jungle and urban cycles. Different mosquitoes of the genus Aedes are involved. Concern exists about YFV spreading to southeast Asia, where its vector A. aegypti already occurs.


Yellow fever virus has been the cause of epidemics in certain jungle locations of sub-Saharan Africa, as well as in some parts of South America. Although many YFV infections are mild, the disease can also cause severe, life-threatening illness. The initial or acute phase of the disease state is normally characterized by high fever, chills, headache, backache, muscle ache, loss of appetite, nausea, and vomiting. After three to four days, these symptoms disappear. In some subjects, symptoms then reappear, as the disease enters its so-called toxic phase. During this phase, high fever reappears and can lead to shock, bleeding (e.g., bleeding from the mouth, nose, eyes, and/or stomach), kidney failure, and liver failure. Indeed, liver failure causes jaundice, which is yellowing of the skin and the whites of the eyes, and thus gives “YFV” its name. About half of the subjects who enter the toxic phase die within 10 to 14 days. However, persons that recover from YFV have lifelong immunity against reinfection. The number of people infected with YFV over the last two decades has been increasing, with there now being about 200,000 YFV cases, and about 30,000 associated deaths, each year. The re-emergence of YFV thus presents a serious public health concern.


Yellow fever is most frequently a clinical diagnosis, based on symptomatology and travel history. Mild cases of the disease can only be confirmed virologically. Since mild cases of YFV can also contribute significantly to regional outbreaks, every suspected case of YFV (involving symptoms of fever, pain, nausea, and vomiting 6-10 days after leaving the affected area) is treated seriously. If YFV is suspected, the virus cannot be confirmed until 6-10 days following the illness. In a differential diagnosis, infections with YFV must be distinguished from other feverish illnesses such as malaria. Other viral hemorrhagic fevers, such as Ebola virus, Lassa virus, Marburg virus, and Junin virus, must be excluded as the cause.


As with other Flavivirus infections, no cure is known for YFV. Hospitalization is advisable and intensive care may be necessary because of rapid deterioration in some cases. Certain acute treatment methods lack efficacy: passive immunization after the emergence of symptoms is probably without effect; ribavirin and other antiviral drugs, as well as treatment with interferons, are ineffective in YFV subjects. Symptomatic treatment includes rehydration and pain relief with drugs such as paracetamol (acetaminophen).


Vaccination is recommended for those traveling to affected areas, because non-native people tend to develop more severe illness when infected. Protection begins by the 10th day after vaccine administration in 95% of people, and had been reported to last for at least 10 years. The World Health Organization (WHO) now states that a single dose of vaccine is sufficient to confer lifelong immunity against YFV disease. The attenuated live vaccine stem 17D was developed in 1937 by Max Theiler.


II. FLAVIVIRUS BIOLOGY

Flaviviruses share several common aspects: common size (40-65 nm), symmetry (enveloped, icosahedral nucleocapsid), nucleic acid (positive-sense, single-stranded RNA around 10,000-11,000 bases), and appearance in the electron microscope.


Most of these viruses are primarily transmitted by the bite from an infected arthropod (mosquito or tick), and hence are classified as arboviruses. Human infections with most of these arboviruses are incidental, as humans are unable to replicate the virus to high enough titers to reinfect the arthropods needed to continue the virus lifecycle—humans are then a dead end host. The exceptions to this are the YFV, dengue, and zika viruses. These three viruses still require mosquito vectors, but are well-enough adapted to humans as to not necessarily depend upon animal hosts (although they continue to have important animal transmission routes, as well). Other virus transmission routes for arboviruses include handling infected animal carcasses, blood transfusion, sex, child birth and consumption of unpasteurized milk products. Transmission from nonhuman vertebrates to humans without an intermediate vector arthropod however mostly occurs with low probability.


Entry into the host cell is achieved by attachment of the viral envelope protein E to host receptors, which mediates clathrin-mediated endocytosis. Replication follows the positive stranded RNA virus replication model. Flaviviruses have a (+) sense RNA genome and replicate in the cytoplasm of the host cells. The genome mimics the cellular mRNA molecule in all aspects except for the absence of the poly-adenylated (poly-A) tail. This feature allows the virus to exploit cellular apparatus to synthesize both structural and non-structural proteins, during replication. The cellular ribosome is crucial to the replication of the flavivirus, as it translates the RNA, in a similar fashion to cellular mRNA, resulting in the synthesis of a single polyprotein.


In general, the genome encodes a 5′ untranslated region (5′ UTR); a coding region encoding the three viral structural proteins; seven non-structural proteins, designated NS1, NS2A, NS2B, NS3, NS4A, NS4B, NS5; and a 3′ untranslated region (3′ UTR). The viral structural proteins include the capsid (C), premembrane/membrane (prM) and envelope (E) proteins.


The flavivirus particle contains a nucleocapsid composed of viral RNA and capsid protein C. The nucleocapsid is surrounded by an envelope containing the envelope glycoprotein E (50-60 kDa) and a small membrane protein M (7-8 kDa). Translation of the genomic RNA results in a single polyprotein precursor that is cleaved by cellular and viral proteases into viral proteins, in the order: C, prM/M, E, NS 1, NS2A, NS2B, NS3, NS4A, 2K, NS4B, and NS5, where C through E are the structural components of the virion and NS 1 through NS5 are non-structural proteins required for replication (Lindenbach and Rice, Fields Virology, 4th Ed.:991-1041, 2001). The prM protein C25 kDa) is the intracellular precursor for M. Immature virions containing prM are produced by budding into the lumen of the endoplasmic reticulum (ER) and are transported to the cell surface through the exocytosis pathway. Cleavage of prM occurs shortly prior to particle release in post-Golgi vesicles. Mature extracellular virus contains predominantly M protein, although a small fraction of uncleaved prM can also be present.


Once translated, cleavage of the polyprotein by a combination of viral and host proteases releases mature polypeptide products. Nevertheless, cellular post-translational modification is dependent on the presence of a poly-A tail; therefore this process is not host-dependent. Instead, the polyprotein contains an autocatalytic feature which automatically releases the first peptide, a virus specific enzyme. This enzyme is then able to cleave the remaining polyprotein into the individual products. One of the products cleaved is a polymerase, responsible for the synthesis of a (−) sense RNA molecule. Consequently, this molecule acts as the template for the synthesis of the genomic progeny RNA.


The genomic RNA is modified at the 5′ end of positive-strand genomic RNA with a cap-1 structure (me7-GpppA-me2). Cellular RNA cap structures are formed via the action of an RNA triphosphatase, with guanylyltransferase, N7-methyltransferase and 2′-O methyltransferase. The virus encodes these activities in its non-structural proteins.


Most of the non-structural proteins associate to form the replicase complex, which catalyzes RNA accumulation in close association with modified cytoplasmic membranes. NS3 and its cofactor NS2B is the main viral protease. NS2B contributes to the structure of NS3, anchors the enzyme to the membrane, is required for the majority of NS3-mediated processing events, and can modulate its helicase activity. NS3, together with the NS2B cofactor, mediates cleavage of the flavivirus C protein from its membrane anchor, and is therefore essential for maturation of a major virion component. Processing of C on the cytosolic side of the ER membrane by the flaviviral protease is essential for subsequent cleavage of C from the envelope protein, prM, by the host enzyme signal peptidase in the ER lumen. This regulated sequence of cleavages ensures that processing of prM from the polyprotein is delayed, and budding is stalled, until genomic RNA substrates accumulate through replication; the secretion of the flavivirus glycoproteins, E and prM, as immunogenic subviral particles is thereby minimized. NS3 also has helicase and nucleoside triphosphatase (NTPase) activities, which are essential for replication. The NS3 protein encodes a RNA triphosphatase within its helicase domain. It uses the helicase ATP hydrolysis site to remove the γ-phosphate from the 5′ end of the RNA. The N-terminal domain of the non-structural protein 5 (NS5) has both the N7-methyltransferase and guanylyltransferase activities necessary for forming mature RNA cap structures. RNA binding affinity is reduced by the presence of ATP or GTP and enhanced by S-adenosyl methionine. This protein also encodes a 2′-O methyltransferase.


NS2A is also an essential component of the replicase; it colocalizes with replication complexes and has an essential role in the process of RNA accumulation. Purified NS2A binds RNA, leading to the hypothesis that this integral membrane protein might shuttle genomic substrates out of membrane bound replication complexes to the sites of packaging. NS5 constitutes the viral RNA-dependent RNA polymerases and methyltransferase activities, which are important for RNA synthesis and genome capping respectively. NS1, a secreted protein unique to flaviviruses, plays a role early in replication. It is an essential gene that generates a highly conserved ca. 48 kDa glycoprotein that is localized to the lumen of the ER by a signal sequence located at the C-terminus of the structural envelope protein E. NS1 is a highly conserved protein consisting of 352 amino acids with an approximate molecular weight of 40 to 50 kDa, depending on its glycosylation status. The majority of the Flavivirus genus members have two N-linked glycosylation sites at asparagine 130 and asparagine 207, including all four DEN serotypes, JE, and ZIK viruses; YF has glycosylation sites at positions 130 and 208. A few members, such as WN, St. Louis encephalitis, and Murray Valley encephalitis (MVE) viruses, have a third glycosylation site found at amino acid position 175. Interestingly, Entebbe bat virus (ENTV) has four potential N-linked glycosylation sites in NS1, including the two commonly found in all flaviviruses as well as at residues 106 and 326. TBEV appears to have three putative N-linked glycosylation sites at residues 85, 207, and 223. The amino acids at each glycosylation motif are characterized by N-X-T/S.


Flavivirus NS4A and NS4B proteins also have essential roles in RNA accumulation. The flavivirus NS4B is a predominantly hydrophobic transmembrane protein that localizes in the perinuclear region of virus-infected cells and traverses the ER membrane [7]. Detailed nuclear magnetic resonance (NMR) studies of dengue virus 2 (DENV-2) and DENV-3 NS4B structures indicate that this protein has five transmembrane domains as well as several regions that reside within the ER lumen and a series of amino acids that reside in the cytoplasm [8,9]. Although structural studies have not been reported for WNV NS4B, it is predicted that the structure is conserved amongst members of the flavivirus genus. NS4B is known to exist as both a monomer and dimer [10], and it interacts with other viral proteins including NS1, NS2B, NS3, and NS4A at various times during the replication cycle [11-14]. During replication, NS4B is found closely associated to all NS proteins and to double-stranded RNA (dsRNA), indicating it plays a role in the replication complex [15]. Along with a function in viral replication, several roles of NS4B in host innate immune antagonism have been identified. For instance, DENY, YFV (YFV), and WNV NS4B are able to antagonize type-1 interferon signaling by inhibiting STAT1 and STAT2 phosphorylation and thus inhibiting downstream induction of interferon-stimulated genes (ISGs) [16,17]. Studies of the four serotypes of DENV have found that NS4B can inhibit host RNA interference (RNAi) by inhibiting Dicer activity [18], but it is not yet known if WNV NS4B functions similarly.


NS4B has proven to be capable of harboring a number of amino acids whose mutation can have varying impacts on virus viability, virulence, and attenuation. Specifically, there have been reports of NS4B mutations that confer an attenuated phenotype identified in DENV-2 and DENV-4 NS4B [19-21], as well as YFV [22]. Additionally, the YFV 17D and the Japanese encephalitis virus (JEV) SA14-14-2 live, attenuated vaccines have two and one NS4B mutation(s), respectively, that may be important to vaccine attenuation [23,24]. In terms of WNV, the inventors have previously identified several NS4B mutations that confer attenuation in mice including P38G, C102S, and E249G, although compensatory mutations typically appear to contribute to the attenuated phenotype [25-27].


The 5′ UTR of flaviviruses are highly structured, has a length of approximately 100 nucleotides and harbors two conserved RNA secondary structures which are vital for the viral life cycle. The first structural element is termed 5′ SLA and comprises three stems (S1, S2, S3) folded as Y-shaped-like stem structure, and a side structure domain (SSD). Its overall length is around 70 nucleotides. Disruption experiments of S1 and S2 led to a stop of viral replication. Further, SLA is the promoter for RNA synthesis and interacts with the viral protein NS5 during circularization of the viral genome. After recruitment of NS5, the two loop regions of S3 (TL) and SSD (SSL) are considered to interact with NS5 to promote polymerase activity. Despite the diversity of SSD, its stable structure is essential for infectivity. The second element is termed 5′ SLB and contains the translation initiation codon at the top region of the stem loop. It further contains the 5′UAR (upstream AUG region), which is essential for the circularization of the genome. The 5′UAR interacts with the 3′UAR, which is located at the 3′ UTR of the genome to form a long-range RNA-RNA interaction. During replication, the 5′ UTR interacts with the 3′ UTR of the genome to initiate synthesis of new viral replicates and viral protein translation. In direct adjacency to the 5′ UTR lies the capsid-coding hairpin region (cHP) structure, which is essential for the viral replication. The cHP actually lies in the ORF of the viral genome and is followed by the 5′CS (conserved sequence), which forms another long-range RNA-RNA interaction with the 3′ UTR (3′CS). The cHP aids in the start codon recognition and viral replication. Studies show that the function of cHP is sequence-independent but structure-dependent.


The 3′ UTR ranges between 400 and 700 nucleotides in length. Its RNA secondary structure is known to be necessary for the viral replication during infection. The 3′ UTR of flavivirus—and sometimes even a small part of the 3′ end of the coding region—is also called subgenomic flavivirus RNA (sfRNA). SfRNA is produced by incomplete degradation of the viral genome by the host cell (via XRN1). Local RNA secondary structures (xrRNA elements) in the 3′ UTR and long-range RNA-RNA interactions between 5′ UTR and 3′ UTR of flaviviruses stall XRN1 and cause the undigested fragment of the genome, which has been shown to play a role in pathways comprising both host defense and viral infection.


In contrast to the structurally conserved 5′ UTR of flaviviruses, individual structural elements differ between different viruses, which is associated with the host-adaptation. Flaviviruses are therefore classified into four different groups: Mosquito-borne flaviviruses (MBFV), tick-borne flaviviruses (TBFV), insect-specific flaviviruses (ISFV) and those with no known vector (NKV). Across all groups, three RNA secondary structure elements are conserved within the 3′ UTR: the dumbbell element (DB), cis-acting replication element (CRE) and the exoribonuclease-resistant RNA elements (xrRNA). Further, unique elements have been observed for specific groups as well.


Dumbbell (DB) elements are important for viral RNA synthesis. These two conserved secondary structures are also known as pseudo-repeat elements. They were originally identified within the genome of Dengue virus and are found adjacent to each other within the 3′UTR. These DB elements have a secondary structure consisting of three helices and they play a role in ensuring efficient translation. Via the formation of additional pseudoknots, the loop regions of DB pairs with a complementary motif further downstream of the respective DB element. The DB elements also expose conserved sequences (CS) and repeated conserved sequences (RCS). Deletion of DB1 has a small but significant reduction in translation but deletion of DB2 has little effect. Deleting both DB1 and DB2 reduced translation efficiency of the viral genome to 25%. Further, the DB elements also play a role in viral translation, as deletion of both elements reduced viral translation levels.


The cis-acting replication element (CRE) structure is structurally conserved among known flaviviruses. It consists of a small hairpin (sHP) and a larger structural element (3′ SL). Mutations of sHP are shown to be lethal for Dengue virus in mosquito cells. CRE is highly involved in the 5′-3′ UTR interaction of flaviviruses. Regions of sHP are interacting with the SLB element and the cHP in the 5′ UTR, whereas the 3′ SL harbors a sequence that can interact with SLB, to further stabilize this long-range RNA-RNA interaction.


The exoribonuclease-resistant RNA elements (xrRNA) are described throughout all groups of flaviviruses. Usually, each virus harbors two xrRNAs, xrRNA1 and xrRNA2, in the beginning of the 3′ UTR. The formation of these stem-loops, especially xrRNA1, is vital to ensure resistance against XRN1 activity. The Y-shaped stem-loop is also termed SL II and SL IV, respectively. In order to function as xrRNA, the sequence downstream is needed as well, since the upper loop region forms a pseudoknot (PK) with the single-stranded region directly downstream to its respective hairpin. In some species, the region downstream also forms a small hairpin. In such cases, the PK interactions takes place between the two loop regions. Conserved formation of these structures were observed in mammalian cells but not in mosquito cells, suggesting this region has varying functions in different hosts. In plant-viruses, xrRNA elements have been observed as well, showing some similarities to flaviviral xrRNAs. However, plant-virus xrRNA and flaviviral xrRNA are distinguishable by their underlying three-dimensional folds.


The E protein is the main functional and antigenic surface component of the virion. The molecular structure of the ectodomain of E, which forms a homodimer on the surface of mature viral particles at neutral pH, has been resolved by cryoelectron microscopy (Rey et al., Nature 375:291-298, 1995) and fitted into the electron density map of viral particles (Kuhn et al., Cell 108:717-725, 2002). During infection, the E protein functions as a class II fusion protein (Modis et al., Nature 427:313-319, 2004). Following virus binding to a cellular receptor and internalization, the acidic pH in the resulting endosomes triggers dissociation of the dimers such that the previously hidden hydrophobic fusion loop of each monomer is exposed outwardly. Concurrently, the loops insert into the cell (endosome) membrane and monomers rearrange into elongated trimers. Further refolding of the trimers brings the cell and viral membranes into close proximity and forces them to fuse, releasing the contents of the viral particle into the cytoplasm. Previous studies showed that some substitutions in the E protein of DEN and JE, which are selected during serial passages in mouse brain and in cultured monkey kidney and mosquito cells, have been localized in particular regions of the 3D structure of the protein, and were reported to be associated with changes in the fusion function of the viruses. The studies showed that the fusion pH threshold for some attenuated vaccines decreased by 0.6 to 1 pH unit by comparison with the corresponding parental virus isolate. Some changes in six residues in the DEN3 protein E (residues 54, 191, 202, 266, 268, and 277) map to the region in domain II. This region is proposed as a focus for the low-pH mediated conformational change required for the surface exposure of the conserved hydrophobic cd fusion loop (Lee et al., Virology 232:281-290, 1997).


There is no evidence that the small (mature) M protein plays a role in the events leading to virus internalization from the endosome or has any other appreciable function, while its intracellular precursor, prM, is known to be important for morphogenesis and transport of progeny viral particles. The prM protein also facilitates proper folding of E (Lorenz et al., J. Virol. 76:5480-5491, 2002) and functions to protect the E protein dimer from premature conformational rearrangement during passage of new particles towards the cell surface through acidic secretory compartments (Guirakhoo et al., J. Gen. Virol. 72:1323-1329, 1991; Guirakhoo et al., Virology 191:921-931, 1992).


Flavivirus genomic RNA replication occurs on rough endoplasmic reticulum membranes in membranous compartments. New viral particles are subsequently assembled. This occurs during the budding process which is also responsible for the accumulation of the envelope and cell lysis.


Flaviviruses, including YFV and WNV, have two principal biological properties responsible for their induction of disease states in humans and animals. The first of these two properties is neurotropism, which is the propensity of the virus to invade and infect nervous tissue of the host. Neurotropic flavivirus infection can result in inflammation of and injury to the brain and spinal cord (i.e., encephalitis), impaired consciousness, paralysis, and convulsions. The second of these biological properties of flaviviruses is viscerotropism, which is the propensity of the virus to invade and infect vital visceral organs, including the liver, kidney, and heart.


Neurotropic flavivirus infection begins with neuroinvasion, which is the ability of viruses to enter nervous tissue and cause neurological alterations. Once inoculated in the dermis, these viruses spread to infect target cells such as dendritic cells or monocytes/macrophages or enter directly into the lymph nodes, muscles, liver, spleen or nervous system via nerve endings (Chambers & Diamond, 2003; McMinn, 1997). In some cases during infection with these viruses, the blood-brain barrier (BBB) is disturbed as a result of cytokines and chemokines, such as such as tumour necrosis factor-alpha (TNF-alpha), or enzymes, such as matrix metalloproteinase (MMP), that favor the entry of WNV and JEV into nervous tissue by increasing permeability of the endothelium and permitting the entry of viruses into the cerebral parenchyma (Chambers & Diamond 2003; Chaturvedi et al., 1991). Additionally during infection, endothelial cells are activated and overexpress cellular adhesion molecules that favour the transmigration of immune cells into the cerebral parenchyma, such as E-selectin, VCAM-1 and ICAM-1 (Shen et al., 1997; Verna et al., 2009).


The ability of some viruses to infect and replicate in neurons is called neurotropism and is determined by viral and cellular factors. Mostly virus determinants are associated with envelope glycoprotein gene mutations that favor interactions between the virus and molecules on the neuron surface. These interactions promote the fusion of the virus with the plasma membrane and can also trigger endocytosis or transcytosis of the virus. In flaviviruses, the envelope protein (E) is the principal component of the virion surface. It participates in the recognition and subsequent binding to the receptor and the fusion of the virus with the cell membranes (Lindenbach et al., 2007). This protein is formed by three beta-barrel domains known as domains I, II and III, and these last two are responsible for interacting with putative receptor molecules (Pastorino et al., 2010). The molecules that have been reported as possible receptors for flavivirus in different cell populations include ICAM-3 (Jindadamrongwech & Smith, 2004), CD209 (DC-SIGN) (Tassaneetrithep et al., 2003), DC-SIGNR (Davis et al., 2006), integrins (Chu & Ng, 2004), the mannose receptor (Miller et al., 2008), HSP70 and HSP90 (Das et al., 2009; Reyes del Valle et al., 2005), the laminin receptor (Tio et al., 2005) and heparin sulphate (HS) (Germi et al., 2002) among others (Barba-Spaeth et al., 2005; Upanan et al., 2008).


Flaviviruses also have some propensity to infect visceral organs. Viscerotropic flavivirus infection can result in inflammation and injury of the liver (hepatitis), kidney (nephritis), and cardiac muscle (myocarditis), leading to failure or dysfunction of these organs.


Neurotropism and viscerotropism appear to be distinct and separate properties of flaviviruses. Some flaviviruses are primarily neurotropic (such as WNV), others are primarily viscerotropic (e.g., YFV and dengue virus), and still others exhibit both properties (such as Kyasanur Forest disease virus). However, both neurotropism and viscerotropism are present to some degree in all flaviviruses. Within a host, an interaction between viscerotropism and neurotropism is likely to occur, because infection of viscera occurs before invasion of the central nervous system. Thus, neurotropism depends on the ability of the virus to replicate in extraneural organs (viscera). This extraneural replication produces viremia, which in turn is responsible for invasion of the brain and spinal cord. Therefore, while the viscerotropism of these viruses may not necessarily cause dysfunction of vital visceral organs, replication of virus in these organs can cause viremia and thus contribute to invasion of the central nervous system. Thus, in addition to decreasing risk of damage to visceral organs, decreasing the viscerotropism of these viruses by mutagenesis can reduce their abilities to invade the brain and cause encephalitis and other conditions.


III. RECOMBINANT FLAVIVIRUSES

The present disclosure provides recombinant attenuated flaviviruses that can be used in therapeutic methods, such as methods of inducing an immune response and vaccination methods. Central to the flaviviruses of the disclosure are the presence of attenuating mutations in the genomes of the viruses. These mutations can attenuate the viruses by, for example, decreasing the viscerotropism and/or neurotropism of the viruses. The mutations can be present in regions of the flavivirus genome including the 3′ untranslated region (3′ UTR), capsid sequences, envelope sequences, and/or non-structural sequences. Thus, in some embodiments, mutagenesis can be used to produce attenuated flavivirus that can reduce disease in a subject. Each of these types of mutations, which can be combined with each other and/or other attenuating mutations, are described herein.


Considering that small changes to NS4B can have a substantial impact on the phenotype of flaviviruses, mutation of NS4B may be important for live, attenuated flavivirus vaccine development. In some embodiments, the attenuated flavivirus comprises a flavivirus wherein a nucleic acid construct encoding the genome of the attenuated flavivirus comprises one or more mutations corresponding to a transmembrane domain of non-structural protein 4B (NS4B) of the flavivirus.


The NMR structure of DENV NS4B [8,9] can be used to determine the putative location of amino acid residues of interest, which allows for mutations in different domains of NS4B to be evaluated. Several novel amino acid mutations in NS4B can be made for their ability to alter the phenotype of flaviviruses. Proline 54 occupies the first transmembrane domain of NS4B in WNV and is a candidate for attenuating mutations to reduce neuroinvasion. Thus, in some embodiments, the one or more mutations comprise a substitution of wild-type amino acid proline 54 of the transmembrane domain of NS4B in WNV. In some embodiments, the substitution of wild-type amino acid 54 comprises a substitution of a proline residue with a nonpolar amino acid. In some embodiments, the nonpolar amino acid comprises alanine residue or glycine residue. In some embodiments, the substitution of wild-type amino acid 54 comprises a substitution of a proline residue with a glycine residue. In some embodiments, reversion of the P54G mutation to the wild-type amino acid is inhibited when the virus is grown in a host. Thus, in some embodiments, P54G is a stable mutation. Two nucleotide changes are required to revert glycine to proline, which may reduce reversion to the wild-type amino acid.


P54 in WNV is homologous to P52 in YFV. Thus, in some embodiments, the one or more mutations comprise a substitution of a wild-type amino acid homologous to wild-type amino acid 54 of the transmembrane domain of WNV NS4B. In some embodiments, the YFV wild-type amino acid homologous to wild-type amino acid 54 of the transmembrane domain of WNV NS4B comprises amino acid 52, and wherein substitution of amino acid 52 comprises a substitution of a proline residue with a nonpolar amino acid. In some embodiments, the nonpolar amino acid comprises alanine residue or glycine residue. In some embodiments, the substitution of wild-type amino acid 52 comprises a substitution of a proline residue with a glycine residue. In some embodiments, reversion of the P52G mutation to the wild-type amino acid is inhibited when the virus is grown in a host. Thus, in some embodiments, P52G is a stable mutation.


Since proline residues can be key to protein structure, specifically in alpha helices [9,39,40], it is possible that mutation of P54 or P52 alters the structure and thus the function of NS4B. For example, immunostaining of virus-infected cells demonstrated that cells infected with a P54G mutant do not accumulate NS4B as rapidly as NY99ic-infected cells even though NS1 staining was comparable. Furthermore, a reduction in colocalization of NS1 and NS4B in cells infected with the P54G mutant could indicate that there is less interaction between these two viral proteins, which may inhibit the function of the replication complex. Another consideration is that mutation in NS4B could alter dimer formation. The regions involved in NS4B dimerization include amino acids 129-165 in the cytoplasmic loop and amino acids 166-248 in the C-terminal region, whereas the region including residue 54 has very little impact on dimerization [10].


In some embodiments, the substitution of wild-type amino acid 54 in WNV or amino acid 52 in YFV of the transmembrane domain of NS4B results in reduced induction of cytokines and chemokines as compared to mock-infected cells. In some embodiments, the substitution of wild-type amino acid 54 in WNV or amino acid 52 in YFV of the transmembrane domain of NS4B results in reduced induction of IFN-β as compared to mock-infected cells. In many viral infections, cytokine storm is associated with severe pathology, thus, low cytokine induction by P54 WNV mutants or P52 YFV mutants could reduce disease severity. Previous studies demonstrated that a P38G (+NS3-N480H/NS4B-T1161) WNV mutant induced stronger pro-inflammatory cytokines than NY99 in dendritic cells, THP-1 cells, THP macrophages, and C57Bl/6 mice [35,36], indicating that induction of a robust immune response may contribute to the mechanism of attenuation for the WNV P38G mutant. Thus, the mechanism of attenuation and protection of a P38G as compared to a P54 WNV or P52 YFV mutant is likely to be different.


The mutations of the present disclosure can also be used to complement or improve the attenuation of flavivirus strains that already include one or more other attenuating mutations. For example, the mutations described herein may be combined with an attenuated virus strain developed by the ablation of the glycosylation sites in the envelope (E) and non-structural 1 (NS1) proteins. West Nile virus (WNV), like all members of the Japanese encephalitis (JE) serogroup except JE virus, contains three N-linked glycosylation (N-X-S/T) sites in the NS1 protein at asparagine residues NS1(130), NS1(175) and NS1(207). This E(154S)/NS1(130A/175A/207A) strain showed modest reduction in multiplication kinetics in cell culture and small plaque phenotype compared to the parental NY99 strain yet displayed greater than a 200,000-fold attenuation for mouse neuroinvasiveness compared to the parental strain. Mice infected with 1000PFU of E(154S)/NS1(130A/175A/207A) showed undetectable viremia at either two or three days post infection; nonetheless, high titer neutralizing antibodies were detected in mice inoculated with low doses of this virus and protected against lethal challenge with a 50% protective dose of 50PFU. See Whiteman et al., Vaccine. 2010, 28(4):1075-83.


Further attenuation may be achieved by mutating the asparagine to serine or glutamine in addition to mutating other residues in the NS1(130-132) glycosylation motif and combining these mutations with those described herein to decrease neuroinvasiveness and neurovirulence. NS1(130-132QQA/175A/207A), the most attenuated mutant virus, showed modest changes in infectivity titers versus the parental strain, was not temperature sensitive, and did not show reversion in mice. Mutant virus was completely attenuated for neuroinvasiveness after intraperitoneal inoculation with >1,000,000 PFU, and mice were protected against lethal challenge. Overall, changing the asparagine of the NS1(130) glycosylation motif to a serine or glutamine attenuated WNV further than the asparagine to alanine substitution. Further, mutating all three of the amino acids of the NS1(130-132) glycosylation motif (NTT-QQA) along with NS1(175) and NS1(207) asparagine to alanine mutations gave the most stable and attenuated strain. See Whiteman et al., Vaccine. 2011, 29(52):9702-10.


Thus, in some embodiments, the nucleic acid construct encoding the genome of the attenuated flaviviruses disclosed herein further comprises one or more mutations corresponding to one or more glycosylation sites of non-structural protein 1 (NS1) of the flavivirus. In some embodiments, the one or more mutations corresponding to one or more glycosylation sites of NS1 of the flavivirus comprise a substitution of wild-type amino acids 130-132 of WNV NS1. In some embodiments, the substitution of wild-type amino acids 130 and 131 comprises a substitution of an asparagine residue with a polar amino acid. In some embodiments, the polar amino acid comprises a glutamine residue. In some embodiments, the substitution of amino acid 132 comprises a substitution of a threonine residue with a nonpolar amino acid. In some embodiments, the nonpolar amino acid comprises an alanine residue. In some embodiments, the one or more mutations corresponding to one or more glycosylation sites of NS1 of the flavivirus further comprise a substitution of wild-type amino acid 175 of WNV NS1. In some embodiments, the one or more mutations corresponding to one or more glycosylation sites of NS1 of the flavivirus further comprise a substitution of wild-type amino acid 207 of WNV NS1. In some embodiments, the substitution of wild-type amino acid 175 or wild-type amino acid 207 comprises a substitution of an asparagine residue with a nonpolar amino acid. In some embodiments, the nonpolar amino acid comprises an alanine residue. Homologous mutations to one or more glycosylation sites of YFV NS1 are also contemplated.


Other mutations which may be combined with the mutations disclosed herein include NS4B-P38G, NS4B-C102S, and/or E249G in WNV and homologous mutations in YFV. These mutations are described in detail in Wicker et al., Virology. 2006, 349(2):245-53; Wicker et al., Virology. 2012, 426(1): 22-33; and Davis et al., Virology. 2004, 330(1):342-50.


It is also possible to combine mutations or deletions specified in NS4B with one or more mutations or deletions in the 3′UTR, the capsid gene, in the prM gene, or in the E gene at sites known to attenuate flaviviruses. Thus, optionally, in some embodiments, the mutations of NS4B can be included in a strain with one or more additional attenuating mutations, such as 3′UTR mutations, attenuating mutations in the hinge region of the envelope protein of the virus, amino acids in the membrane protein (for example, the membrane helix portion of the membrane protein), or any of the capsid or envelope protein mutations described herein.


For example, 3′UTR mutations may comprise short, attenuating deletions of for example, less than 30 nucleotides (e.g., 1, 2, 3, etc., and up to 29 (e.g., 2-25, 3-20, 4-15, 5-10, or 6-8 nucleotides in length)) or substitutions in the 3′UTR. In some examples, the short 3′UTR deletions or substitutions are designed to destabilize the secondary structure of one or more predicted stem structures in the 3′UTR.


Capsid mutations may comprise short deletions (e.g., deletions of 1, 2, 3, or 4 amino acids) in the capsid protein. Examples of such mutations include viable deletions affecting Helix I of the protein. Other short mutations in this region can be tested for viability and attenuation, and are also included. Capsid protein sequences of other flaviviruses have been published, e.g., for TBE, WNV, Kunjin, JE, and dengue viruses (e.g., Pletnev et al., Virology 174:250-263, 1990).


Membrane mutations may comprise, for example, mutations in the membrane helix portion of the membrane protein (e.g., in any one or more amino acids corresponding to amino acids 40-75 of WNV, for example, amino acid 66). As a specific example, in the case of a WNV membrane protein, the membrane protein amino acid 66 (leucine in wild type WNV) can be replaced with another amino acid, such as proline. In addition to proline, other hydrophobic amino acids, such as isoleucine, methionine, or valine, or small amino acids, such as alanine or glycine, can substitute the wildtype amino acid at position 66 of the membrane protein. As other examples, amino acids at positions 60, 61, 62, 63, and/or 64 of WNV (or corresponding positions in other flaviviruses) can be substituted, alone or in combination with each other, a mutation at position 66, and/or another mutation(s). Examples of substitutions at these positions include: arginine to glycine at position 60, Valine to alanine at position 61, Valine to glutamic acid or methionine at position 62, phenylalanine to serine at position 63, and Valine to isoleucine at position 64.


Envelope mutations may comprise hinge region mutations (e.g., substitutions at positions corresponding to amino acids 48-61, 127, 131, 170, 173, 200, 299, 305, 380, and 196-283 of YFV and substitutions in residues lining the hydrophobic pocket of the domain II (Hurrelbrink et al., Adv. Virus Res. 60:1-42, 2003: Modis et al., PNAS 100:6986-91, 2003) including residues 52 and 200 in the case of YFV, amino acid 279 of Japanese encephalitis, and amino acids 204, 252, 253, 257, 258, and 261 of dengue 1 virus (see, e.g., WO 03/103571)), as well as substitutions in amino acids 107, 138, 176, 177, 244, 264, 280, 316, and 440 of the WNV. The envelope (E) gene contains functional domains within which amino acid changes may affect function and thereby reduce virulence, as described by Hurrelbrink and McMinn (Adv. Virus Dis. 60:1-42, 2003). The polypeptide chain of the envelope protein folds into three distinct domains: a central domain (domain I), a dimerization domain (domain II), and an immunoglobulin-like module domain (domain III). The hinge region is present between domains I and II and, upon exposure to acidic pH, undergoes a conformational change (hence the designation “hinge”) that results in the formation of envelope protein trimers that are involved in the fusion of viral and endosomal membranes, after virus uptake by receptor-mediated endocytosis. Prior to the conformational change, the proteins are present in the form of dimers. The functional regions of the E protein in which mutations may be inserted that, together with NS4B deletions/mutations, may result in an appropriately attenuated vaccine include: (a) the putative receptor binding region on the external Surface of domain III, (b) the molecular hinge region between domains I and II, which determines the acid-dependent conformational changes of the E protein in the endosome and reduce the efficiency of virus internalization; (c) the interface of prM/M and E proteins, a region of the E protein that interfaces with prM/M following the rearrangement from dimer to trimer after exposure to low pH in the endosome; (d) the tip of the fusion domain of domain II, which is involved in fusion to the membrane of the endosome during internalization events; and (e) the stem-anchor region, which is also functionally is involved in conformational changes of the E protein during acid-induced fusion events.


Further, the viruses of the invention can also include any other mutations that may or may not be attenuating, but are otherwise beneficial for the vaccine (e.g., for vaccine manufacturing), for example, nucleotide changes in the UTRS or amino acid changes in structure or non-structural proteins that can spontaneously accumulate during virus propagation and be desirable.


Flaviviruses that can be subject to the mutations of the invention include mosquito-borne flaviviruses, such as West Nile (New York 1999 strain); YFV (the wildtype Asibi strain; the YF17D vaccine strain (Smithbum et al., World Health Org., p. 238, 1956; Freestone, in Plotkin et al. (eds.), Vaccines, 2nd edition, W. B. Saunders, Philadelphia, 1995); YF17DD (GenBank Accession No. U 17066); YF17D-213 (GenBank Accession No. U17067) (dos Santos et al., Virus Res. 35:35-41, 1995); YF17D-204 France (X15067, X15062); YF17D-204, 234 US (Rice et al., Science 229:726-733, 1985; Rice et al., New Biologist 1:285-296, 1989; C 03700, K 02749); and YFV strains described by Galler et al., Vaccine 16 (9/10):1024-28, 1998); Japanese encephalitis (e.g., SA14-14-2), dengue (serotypes 1-4), Murray Valley encephalitis, St. Louis encephalitis, Kunjin, Rocio encephalitis, and Ilheus viruses; tick-borne flaviviruses, such as Central European encephalitis, Siberian encephalitis, Russian Spring-Summer encephalitis, Kyasanur Forest Disease, Alkhurma, Omsk Hemorrhagic fever, Louping ill, Powassan, Negishi, Absettarov, Hansalova, Apoi, and Hypr viruses; as well as viruses from the Hepacivirus genus (e.g., Hepatitis C virus).


A. Polypeptides


In that the compositions of the present disclosure are particularly suitable for use in treating or preventing flavivirus infection, preferred proteins are contemplated. Preferred proteins include flaviviral proteins, for example, the polyprotein precursor that is cleaved by cellular and viral proteases into viral proteins, in the order: C, prM/M, E, NS 1, NS2A, NS2B, NS3, NS4A, 2K, NS4B, and NS5, where C through E are the structural components of the virion and NS1 through NS5 are non-structural proteins required for replication.


As used herein, a “protein”, “peptide”, or “polypeptide” refers to a molecule comprising at least three amino acid residues. As used herein, the term “wild-type” refers to the endogenous version of a molecule that occurs naturally in an organism. In some embodiments, wild-type versions of a protein or peptide are employed, however, in many embodiments of the disclosure, a modified protein or peptide is employed. The terms described above may be used interchangeably. A “modified protein” or “modified polypeptide” or a “variant” or a “mutant” refers to a protein or peptide whose chemical structure, particularly its amino acid sequence, is altered with respect to the wild-type protein or peptide. In some embodiments, a modified/variant/mutant protein or peptide has at least one modified activity or function (recognizing that proteins or polypeptides may have multiple activities or functions). It is specifically contemplated that a modified/variant protein or peptide may be altered with respect to one activity or function yet retain a wild-type activity or function in other respects.


Recombinant DNA technology may be employed wherein a nucleotide sequence that encodes a peptide or polypeptide is inserted into an expression vector, transformed or transfected into an appropriate host cell and cultivated under conditions suitable for expression. In particular embodiments, there are isolated nucleic acid segments and recombinant vectors incorporating nucleic acid sequences that encode a peptide. The term “recombinant” may be used in conjunction with a peptide or the name of a specific peptide, and this generally refers to a peptide produced from a nucleic acid molecule that has been manipulated in vitro or that is a replication product of such a molecule.


In certain embodiments a protein may be purified. Generally, “purified” will refer to a specific or protein, polypeptide, or peptide composition that has been subjected to fractionation to remove various other proteins, polypeptides, or peptides, and which composition substantially retains its activity, as may be assessed, for example, by the protein assays, as would be known to one of ordinary skill in the art for the specific or desired protein, polypeptide, or peptide.


B. Polynucleotides


Some aspects are directed to a nucleic acid construct encoding the genome of an attenuated flavivirus disclosed herein, or a complement thereof. Some aspects are directed to a nucleic acid construct encoding for a polypeptide or fragment thereof that corresponds to an attenuated flavivirus. In certain embodiments, nucleic acid sequences can exist in a variety of instances such as: isolated segments and recombinant vectors of incorporated sequences or recombinant polynucleotides encoding the genome of an attenuated flavivirus, or a fragment, derivative, mutein, or variant thereof, polynucleotides sufficient for use as hybridization probes, polymerase chain reaction (PCR) primers or sequencing primers for identifying, analyzing, mutating or amplifying a polynucleotide encoding a polypeptide, antisense oligonucleotides for inhibiting expression of a polynucleotide, and complementary sequences of the foregoing described herein. The nucleic acids can be single-stranded or double-stranded and can comprise RNA and/or DNA nucleotides and artificial variants thereof (e.g., peptide nucleic acids).


The term “polynucleotide” refers to a nucleic acid molecule that either is recombinant or has been isolated from total genomic nucleic acid. Included within the term “polynucleotide” are oligonucleotides (nucleic acids 100 residues or less in length), recombinant vectors, including, for example, plasmids, cosmids, phage, viruses, and the like. Polynucleotides include, in certain aspects, regulatory sequences, isolated substantially away from their naturally occurring genes or protein encoding sequences. Polynucleotides may be single-stranded (coding or antisense) or double-stranded, and may be RNA, DNA (genomic, cDNA or synthetic), analogs thereof, or a combination thereof. Additional coding or non-coding sequences may, but need not, be present within a polynucleotide.


In this respect, the term “gene,” “polynucleotide,” or “nucleic acid” is used to refer to a nucleic acid that encodes a protein, polypeptide, or peptide (including any sequences required for proper transcription, post-translational modification, or localization). As will be understood by those in the art, this term encompasses genomic sequences, expression cassettes, cDNA sequences, and smaller engineered nucleic acid segments that express, or may be adapted to express, proteins, polypeptides, domains, peptides, fusion proteins, and mutants. A nucleic acid encoding all or part of a polypeptide may contain a contiguous nucleic acid sequence encoding all or a portion of such a polypeptide. It also is contemplated that a particular polypeptide may be encoded by nucleic acids containing variations having slightly different nucleic acid sequences but, nonetheless, encode the same or substantially similar protein.


In certain embodiments, there are polynucleotide variants having substantial identity to the sequences disclosed herein; those comprising at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% or higher sequence identity, including all values and ranges there between, compared to a polynucleotide sequence provided herein (SEQ ID NO:1, 3, 5, and 7) using the methods described herein (e.g., BLAST analysis using standard parameters). In certain aspects, the isolated polynucleotide will comprise a nucleotide sequence encoding a polypeptide that has at least 80%, at least 85%, at least 90%, or at least 95%, or at least 99% and above, identity to an amino acid sequence described herein, over the entire length of the sequence; or a nucleotide sequence complementary to said isolated polynucleotide. In certain aspects, the polynucleotide will encode a flavivirus polyprotein with a NS4B having a substitution at proline 54.


The nucleic acid segments, regardless of the length of the coding sequence itself, may be combined with other nucleic acid sequences, such as promoters, polyadenylation signals, additional restriction enzyme sites, multiple cloning sites, other coding segments, and the like, such that their overall length may vary considerably. The nucleic acids can be any length. They can be, for example, 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 75, 100, 125, 175, 200, 250, 300, 350, 400, 450, 500, 750, 1000, 1500, 3000, 5000 or more nucleotides in length, and/or can comprise one or more additional sequences, for example, regulatory sequences, and/or be a part of a larger nucleic acid, for example, a vector. It is therefore contemplated that a nucleic acid fragment of almost any length may be employed, with the total length preferably being limited by the ease of preparation and use in the intended recombinant nucleic acid protocol. In some cases, a nucleic acid sequence may encode a polypeptide sequence with additional heterologous coding sequences, for example to allow for purification of the polypeptide, transport, secretion, post-translational modification, or for therapeutic benefits such as targeting or efficacy. As discussed above, a tag or other heterologous polypeptide may be added to the modified polypeptide-encoding sequence, wherein “heterologous” refers to a polypeptide that is not the same as the modified polypeptide.


C. Modified Polynucleotides and Peptides


Mutations can be made in the viruses of the disclosure using standard methods, including, but not limited to, site-directed mutagenesis, direct synthesis, deletion, or other method using techniques known to those skilled in the art. It is understood by those skilled in the art that the virulence screening assays, as described herein and as are well known in the art, can be used to distinguish between virulent and avirulent structures. Nucleic acid sequences of any construct disclosed herein and/or nucleic acid molecules encoding the flavivirus proteins may be synthesized using any known nucleic acid synthesis techniques and inserted into an appropriate vector. Avirulent, immunogenic viruses of embodiments herein can therefore be produced using recombinant engineering techniques known to those skilled in the art.


Thus, in some aspects, the present disclosure is directed to a method of producing a recombinant attenuated flavivirus. In some embodiments, the method produces a recombinant attenuated flavivirus that can be used as a vaccine to treat or prevent flavivirus infection. In some embodiments, the method comprises introducing into a nucleic acid construct encoding the genome of the attenuated flavivirus one or more mutations corresponding to a transmembrane domain of non-structural protein 4B (NS4B) of the flavivirus. In some embodiments, the one or more mutations attenuates the flavivirus, relative to a corresponding flavivirus lacking the mutation, and further.


In some embodiments, the one or more mutations comprise a substitution of wild-type amino acid 54 of the transmembrane domain of WNV NS4B. In some embodiments, the one or more mutations comprise a substitution of wild-type amino acid 52 of the transmembrane domain of YFV NS4B. In some embodiments, the substitution of wild-type amino acid 54 of WNV or wild-type amino acid 52 of YFV comprises a substitution of a proline residue with a nonpolar amino acid. In some embodiments, the nonpolar amino acid comprises alanine residue or glycine residue. In some embodiments, the method further comprises introducing into the nucleic acid construct encoding the genome of the attenuated flavivirus one or more additional mutations corresponding to one or more glycosylation sites of non-structural protein 1 (NS1) of the flavivirus. In some embodiments, the one or more mutations corresponding to one or more glycosylation sites of NS1 of the flavivirus comprise a substitution of wild-type amino acids 130-132 of WNV NS1. In some embodiments, the substitution of wild-type amino acids 130 and 131 comprises a substitution of an asparagine residue with a polar amino acid. In some embodiments, the polar amino acid comprises a glutamine residue. In some embodiments, the substitution of amino acid 132 comprises a substitution of a threonine residue with a nonpolar amino acid. In some embodiments, the nonpolar amino acid comprises an alanine residue. In some embodiments, the one or more mutations corresponding to one or more glycosylation sites of NS1 of the flavivirus further comprise a substitution of wild-type amino acid 175 of NS1. In some embodiments, the one or more mutations corresponding to one or more glycosylation sites of NS1 of the flavivirus further comprise a substitution of wild-type amino acid 207 of NS1. In some embodiments, the substitution of wild-type amino acid 175 or wild-type amino acid 207 comprises a substitution of an asparagine residue with a nonpolar amino acid. In some embodiments, the nonpolar amino acid comprises an alanine residue. Homologous mutations to one or more glycosylation sites of YFV NS1 are also contemplated.


The mutations described above are deletions and substitutions, but other types of mutations, such as insertions, can be used in the invention as well. In addition, as is noted above, the mutations can be present singly or in the context of one or more additional mutations. Further, in addition to the specific amino acids noted above, the substitutions can be made with other amino acids, such as amino acids that would result in a conservative change from those noted above. Conservative substitutions typically include substitutions within the following groups: glycine, alanine, valine, isoleucine, and leucine; aspartic acid, glutamic acid, asparagine, and glutamine; serine and threonine; lysine and arginine; and phenylalanine and tyrosine. Further, both conservative and non-conservative changes can be selected for analysis of their attenuating effect(s) based on computer-predicted (using protein structure modeling software) changes they cause in the E protein X-ray structure.


As modifications and/or changes may be made in the structure of the polynucleotides and/or proteins according to the present disclosure, while obtaining attenuated flaviviruses and/or molecules having similar or improved characteristics, such biologically functional equivalents are also encompassed within the present invention.


The biological functional equivalent may comprise a polynucleotide that has been engineered to contain distinct sequences while at the same time retaining the capacity to encode the “wild-type” or standard protein or peptide or “variant” protein or peptide. This can be accomplished to the degeneracy of the genetic code, i.e., the presence of multiple codons, which encode for the same amino acids. The term “functionally equivalent codon” is used herein to refer to codons that encode the same amino acid, such as the six different codons for arginine. Also considered are “neutral substitutions” or “neutral mutations” which refers to a change in the codon or codons that encode biologically equivalent amino acids. In one example, one of skill in the art may wish to introduce a restriction enzyme recognition sequence into a polynucleotide while not disturbing the ability of that polynucleotide to encode a protein.


In terms of functional equivalents, it is well understood by the skilled artisan that, inherent in the definition of a “biologically functional equivalent” protein and/or polynucleotide, is the concept that there is a limit to the number of changes that may be made within a defined portion of the molecule while retaining a molecule with an acceptable level of equivalent biological activity. Biologically functional equivalents are thus defined herein as those proteins (and polynucleotides) in selected amino acids (or codons) which may be substituted.


In general, the shorter the length of the molecule, the fewer changes that can be made within the molecule while retaining function. Longer domains may have an intermediate number of changes. The full-length protein will have the most tolerance for a larger number of changes. However, it must be appreciated that certain molecules or domains that are highly dependent upon their structure may tolerate little or no modification and retain a biologically viable virus that is infectious to cells.


In one example, a polynucleotide may be (and encode) a biological functional equivalent with more significant changes. Certain amino acids may be substituted for other amino acids in a protein structure without appreciable loss of interactive binding capacity with structures such as, for example, antigen-binding regions of antibodies, binding sites on substrate molecules, receptors, and such like.


Substitutional variants typically contain the exchange of one amino acid for another at one or more sites within the protein, and may be designed to modulate one or more properties of the polypeptide, with or without the loss of other functions or properties. Substitutions may be conservative, that is, one amino acid is replaced with one of similar shape and charge. Conservative substitutions are well known in the art and include, for example, the changes of: alanine to serine; arginine to lysine; asparagine to glutamine or histidine; aspartate to glutamate; cysteine to serine; glutamine to asparagine; glutamate to aspartate; glycine to proline; histidine to asparagine or glutamine; isoleucine to leucine or valine; leucine to valine or isoleucine; lysine to arginine; methionine to leucine or isoleucine; phenylalanine to tyrosine, leucine or methionine; serine to threonine; threonine to serine; tryptophan to tyrosine; tyrosine to tryptophan or phenylalanine; and valine to isoleucine or leucine. So-called “conservative” changes do not disrupt the biological activity of the protein, as the structural change is not one that impinges of the protein's ability to carry out its designed function. It is thus contemplated by the inventors that various changes may be made in the sequence of genes and proteins disclosed herein, while still fulfilling the goals of the present invention. Alternatively, substitutions may be non-conservative such that a function or activity of the polypeptide is affected. Non-conservative changes typically involve substituting a residue with one that is chemically dissimilar, such as a polar or charged amino acid for a nonpolar or uncharged amino acid, and vice versa. Non-conservative substitutions may involve the exchange of a member of one of the amino acid classes for a member from another class.


In other embodiments, alteration of the function of a polypeptide is intended by introducing one or more substitutions. For example, certain amino acids may be substituted for other amino acids in a protein structure without appreciable loss of interactive binding capacity. Structures such as, for example, an enzymatic catalytic domain or interaction components may have amino acid substituted to maintain such function. Since it is the interactive capacity and nature of a protein that defines that protein's biological functional activity, certain amino acid substitutions can be made in a protein sequence, and in its underlying DNA coding sequence, and nevertheless produce a protein with like properties. It is thus contemplated by the inventors that various changes may be made in the DNA sequences of genes without appreciable loss of their biological utility or activity.


Deletion variants typically lack one or more residues of the native or wild type protein. Individual residues can be deleted or a number of contiguous amino acids can be deleted. A stop codon may be introduced (by substitution or insertion) into an encoding nucleic acid sequence to generate a truncated protein. For example, it is contemplated that peptides may be mutated by truncation, or deletion of a number of contiguous amino acids, rendering them shorter than their corresponding wild-type form.


Insertional mutants typically involve the addition of amino acid residues at a non-terminal point in the polypeptide. This may include the insertion of one or more amino acid residues. Terminal additions may also be generated and can include fusion proteins which are multimers or concatemers of one or more peptides or polypeptides described or referenced herein. For example, it is contemplated that peptides might be altered by fusing or conjugating a heterologous protein or polypeptide sequence with a particular function (e.g., for targeting or localization, for enhanced activity, for purification purposes, etc.).


Additionally, the polypeptides of the disclosure may be chemically modified. Glycosylation of the polypeptides can be altered, for example, by modifying one or more sites of glycosylation within the polypeptide sequence to increase the affinity of the polypeptide for antigen (U.S. Pat. Nos. 5,714,350 and 6,350,861).


It also will be understood that amino acid and nucleic acid sequences may include additional residues, such as additional N- or C-terminal amino acids, or 5′ or 3′ sequences, respectively, and yet still be essentially as set forth in one of the sequences disclosed herein, so long as the sequence meets the criteria set forth above, including the maintenance of biological protein activity where protein expression is concerned. The addition of terminal sequences particularly applies to nucleic acid sequences that may, for example, include various non-coding sequences flanking either of the 5′ or 3′ portions of the coding region.


Amino acid sequence variants of the disclosure can be substitutional, insertional, or deletion variants, for example. It is contemplated that a region or fragment of a polypeptide of the disclosure may have an amino acid sequence that has, has at least or has at most 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100, 101, 102, 103, 104, 105, 106, 107, 108, 109, 110, 111, 112, 113, 114, 115, 116, 117, 118, 119, 120, 121, 122, 123, 124, 125, 126, 127, 128, 129, 130, 131, 132, 133, 134, 135, 136, 137, 138, 139, 140, 141, 142, 143, 144, 145, 146, 147, 148, 149, 150, 151, 152, 153, 154, 155, 156, 157, 158, 159, 160, 161, 162, 163, 164, 165, 166, 167, 168, 169, 170, 171, 172, 173, 174, 175, 176, 177, 178, 179, 180, 181, 182, 183, 184, 185, 186, 187, 188, 189, 190, 191, 192, 193, 194, 195, 196, 197, 198, 199, 200 or more amino acid substitutions, contiguous amino acid additions, or contiguous amino acid deletions with respect to any of SEQ ID NOs:2, 4, 6, 8, or 19. Alternatively, a region or fragment of a polypeptide of the disclosure may have an amino acid sequence that comprises or consists of an amino acid sequence that is, is at least, or is at most 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100% (or any range derivable therein) identical to any of SEQ ID NOs:2, 4, 6, 8, or 19. In certain aspects the amino acid sequence will include a mutation corresponding to a WNV NS4B protein having a substitution of proline 54.


Moreover, in some embodiments, a region or fragment comprises an amino acid region of 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100, 101, 102, 103, 104, 105, 106, 107, 108, 109, 110, 111, 112, 113, 114, 115, 116, 117, 118, 119, 120, 121, 122, 123, 124, 125, 126, 127, 128, 129, 130, 131, 132, 133, 134, 135, 136, 137, 138, 139, 140, 141, 142, 143, 144, 145, 146, 147, 148, 149, 150, 151, 152, 153, 154, 155, 156, 157, 158, 159, 160, 161, 162, 163, 164, 165, 166, 167, 168, 169, 170, 171, 172, 173, 174, 175, 176, 177, 178, 179, 180, 181, 182, 183, 184, 185, 186, 187, 188, 189, 190, 191, 192, 193, 194, 195, 196, 197, 198, 199, 200, 201, 202, 203, 204, 205, 206, 207, 208, 209, 210, 211, 212, 213, 214, 215, 216, 217, 218, 219, 220, 221, 222, 223, 224, 225, 226, 227, 228, 229, 230, 231, 232, 233, 234, 235, 236, 237, 238, 239, 240, 241, 242, 243, 244, 245, 246, 247, 248, 249, 250, 251, 252, 253, 254, 255, 256, 257, 258, 259, 260, 261, 262, 263, 264, 265, 266, 267, 268, 269, 270, 271, 272, 273, 274, 275, 276, 277, 278, 279, 280, 281, 282, 283, 284, 285, 286, 287, 288, 289, 290, 291, 292, 293, 294, 295, 296, 297, 298, 299, 300, 301, 302, 303, 304, 305, 306, 307, 308, 309, 310, 311, 312, 313, 314, 315, 316, 317, 318, 319, 320, 321, 322, 323, 324, 325, 326, 327, 328, 329, 330, 331, 332, 333, 334, 335, 336, 337, 338, 339, 340, 341, 342, 343, 344, 345, 346, 347, 348, 349, 350, 351, 352, 353, 354, 355, 356, 357, 358, 359, 360, 361, 362, 363, 364, 365, 366, 367, 368, 369, 370, 371, 372, 373, 374, 375, 376, 377, 378, 379, 380, 381, 382, 383, 384, 385, 386, 387, 388, 389, 390, 391, 392, 393, 394, 395, 396, 397, 398, 399, 400, 401, 402, 403, 404, 405, 406, 407, 408, 409, 410, 411, 412, 413, 414, 415, 416, 417, 418, 419, 420, 421, 422, 423, 424, 425, 426, 427, 428, 429, 430, 431, 432, 433, 434, 435, 436, 437, 438, 439, 440, 441, 442, 443, 444, 445, 446, 447, 448, 449, 450, 451, 452, 453, 454, 455, 456, 457, 458, 459, 460, 461, 462, 463, 464, 465, 466, 467, 468, 469, 470, 471, 472, 473, 474, 475, 476, 477, 478, 479, 480, 481, 482, 483, 484, 485, 486, 487, 488, 489, 490, 491, 492, 493, 494, 495, 496, 497, 498, 499, 500 or more contiguous amino acids starting at position 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100, 101, 102, 103, 104, 105, 106, 107, 108, 109, 110, 111, 112, 113, 114, 115, 116, 117, 118, 119, 120, 121, 122, 123, 124, 125, 126, 127, 128, 129, 130, 131, 132, 133, 134, 135, 136, 137, 138, 139, 140, 141, 142, 143, 144, 145, 146, 147, 148, 149, 150, 151, 152, 153, 154, 155, 156, 157, 158, 159, 160, 161, 162, 163, 164, 165, 166, 167, 168, 169, 170, 171, 172, 173, 174, 175, 176, 177, 178, 179, 180, 181, 182, 183, 184, 185, 186, 187, 188, 189, 190, 191, 192, 193, 194, 195, 196, 197, 198, 199, 200, 201, 202, 203, 204, 205, 206, 207, 208, 209, 210, 211, 212, 213, 214, 215, 216, 217, 218, 219, 220, 221, 222, 223, 224, 225, 226, 227, 228, 229, 230, 231, 232, 233, 234, 235, 236, 237, 238, 239, 240, 241, 242, 243, 244, 245, 246, 247, 248, 249, 250, 251, 252, 253, 254, 255, 256, 257, 258, 259, 260, 261, 262, 263, 264, 265, 266, 267, 268, 269, 270, 271, 272, 273, 274, 275, 276, 277, 278, 279, 280, 281, 282, 283, 284, 285, 286, 287, 288, 289, 290, 291, 292, 293, 294, 295, 296, 297, 298, 299, 300, 301, 302, 303, 304, 305, 306, 307, 308, 309, 310, 311, 312, 313, 314, 315, 316, 317, 318, 319, 320, 321, 322, 323, 324, 325, 326, 327, 328, 329, 330, 331, 332, 333, 334, 335, 336, 337, 338, 339, 340, 341, 342, 343, 344, 345, 346, 347, 348, 349, 350, 351, 352, 353, 354, 355, 356, 357, 358, 359, 360, 361, 362, 363, 364, 365, 366, 367, 368, 369, 370, 371, 372, 373, 374, 375, 376, 377, 378, 379, 380, 381, 382, 383, 384, 385, 386, 387, 388, 389, 390, 391, 392, 393, 394, 395, 396, 397, 398, 399, 400, 401, 402, 403, 404, 405, 406, 407, 408, 409, 410, 411, 412, 413, 414, 415, 416, 417, 418, 419, 420, 421, 422, 423, 424, 425, 426, 427, 428, 429, 430, 431, 432, 433, 434, 435, 436, 437, 438, 439, 440, 441, 442, 443, 444, 445, 446, 447, 448, 449, 450, 451, 452, 453, 454, 455, 456, 457, 458, 459, 460, 461, 462, 463, 464, 465, 466, 467, 468, 469, 470, 471, 472, 473, 474, 475, 476, 477, 478, 479, 480, 481, 482, 483, 484, 485, 486, 487, 488, 489, 490, 491, 492, 493, 494, 495, 496, 497, 498, 499, 500 in any of SEQ ID NOs:2, 4, 6, or 8 (where position 1 is at the N-terminus of the SEQ ID NO). The polypeptides of the disclosure may include 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, or 50 or more variant amino acids or be at least 60%, 61%, 62%, 63%, 64%, 65%, 66%, 67%, 68%, 69%, 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% similar, identical, or homologous with at least, or at most 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100, 101, 102, 103, 104, 105, 106, 107, 108, 109, 110, 111, 112, 113, 114, 115, 116, 117, 118, 119, 120, 121, 122, 123, 124, 125, 126, 127, 128, 129, 130, 131, 132, 133, 134, 135, 136, 137, 138, 139, 140, 141, 142, 143, 144, 145, 146, 147, 148, 149, 150, 151, 152, 153, 154, 155, 156, 157, 158, 159, 160, 161, 162, 163, 164, 165, 166, 167, 168, 169, 170, 171, 172, 173, 174, 175, 176, 177, 178, 179, 180, 181, 182, 183, 184, 185, 186, 187, 188, 189, 190, 191, 192, 193, 194, 195, 196, 197, 198, 199, 200, 201, 202, 203, 204, 205, 206, 207, 208, 209, 210, 211, 212, 213, 214, 215, 216, 217, 218, 219, 220, 221, 222, 223, 224, 225, 226, 227, 228, 229, 230, 231, 232, 233, 234, 235, 236, 237, 238, 239, 240, 241, 242, 243, 244, 245, 246, 247, 248, 249, 250, 300, 400, 500, 550, 600, or more contiguous amino acids, or any range derivable therein, of any of SEQ ID NOs:2, 4, 6, or 8.


The polypeptides of the disclosure may include at least, at most, or exactly 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100, 101, 102, 103, 104, 105, 106, 107, 108, 109, 110, 111, 112, 113, 114, 115, 116, 117, 118, 119, 120, 121, 122, 123, 124, 125, 126, 127, 128, 129, 130, 131, 132, 133, 134, 135, 136, 137, 138, 139, 140, 141, 142, 143, 144, 145, 146, 147, 148, 149, 150, 151, 152, 153, 154, 155, 156, 157, 158, 159, 160, 161, 162, 163, 164, 165, 166, 167, 168, 169, 170, 171, 172, 173, 174, 175, 176, 177, 178, 179, 180, 181, 182, 183, 184, 185, 186, 187, 188, 189, 190, 191, 192, 193, 194, 195, 196, 197, 198, 199, 200, 201, 202, 203, 204, 205, 206, 207, 208, 209, 210, 211, 212, 213, 214, 215, 216, 217, 218, 219, 220, 221, 222, 223, 224, 225, 226, 227, 228, 229, 230, 231, 232, 233, 234, 235, 236, 237, 238, 239, 240, 241, 242, 243, 244, 245, 246, 247, 248, 249, 250, 251, 252, 253, 254, 255, 256, 257, 258, 259, 260, 261, 262, 263, 264, 265, 266, 267, 268, 269, 270, 271, 272, 273, 274, 275, 276, 277, 278, 279, 280, 281, 282, 283, 284, 285, 286, 287, 288, 289, 290, 291, 292, 293, 294, 295, 296, 297, 298, 299, 300, 301, 302, 303, 304, 305, 306, 307, 308, 309, 310, 311, 312, 313, 314, 315, 316, 317, 318, 319, 320, 321, 322, 323, 324, 325, 326, 327, 328, 329, 330, 331, 332, 333, 334, 335, 336, 337, 338, 339, 340, 341, 342, 343, 344, 345, 346, 347, 348, 349, 350, 351, 352, 353, 354, 355, 356, 357, 358, 359, 360, 361, 362, 363, 364, 365, 366, 367, 368, 369, 370, 371, 372, 373, 374, 375, 376, 377, 378, 379, 380, 381, 382, 383, 384, 385, 386, 387, 388, 389, 390, 391, 392, 393, 394, 395, 396, 397, 398, 399, 400, 401, 402, 403, 404, 405, 406, 407, 408, 409, 410, 411, 412, 413, 414, 415, 416, 417, 418, 419, 420, 421, 422, 423, 424, 425, 426, 427, 428, 429, 430, 431, 432, 433, 434, 435, 436, 437, 438, 439, 440, 441, 442, 443, 444, 445, 446, 447, 448, 449, 450, 451, 452, 453, 454, 455, 456, 457, 458, 459, 460, 461, 462, 463, 464, 465, 466, 467, 468, 469, 470, 471, 472, 473, 474, 475, 476, 477, 478, 479, 480, 481, 482, 483, 484, 485, 486, 487, 488, 489, 490, 491, 492, 493, 494, 495, 496, 497, 498, 499, 500, 501, 502, 503, 504, 505, 506, 507, 508, 509, 510, 511, 512, 513, 514, 515, 516, 517, 518, 519, 520, 521, 522, 523, 524, 525, 526, 527, 528, 529, 530, 531, 532, 533, 534, 535, 536, 537, 538, 539, 540, 541, 542, 543, 544, 545, 546, 547, 548, 549, 550, 551, 552, 553, 554, 555, 556, 557, 558, 559, 560, 561, 562, 563, 564, 565, 566, 567, 568, 569, 570, 571, 572, 573, 574, 575, 576, 577, 578, 579, 580, 581, 582, 583, 584, 585, 586, 587, 588, 589, 590, 591, 592, 593, 594, 595, 596, 597, 598, 599, 600, 601, 602, 603, 604, 605, 606, 607, 608, 609, 610, 611, 612, 613, 614, or 615 substitutions (or any range derivable therein).


The substitution may be at amino acid position 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100, 101, 102, 103, 104, 105, 106, 107, 108, 109, 110, 111, 112, 113, 114, 115, 116, 117, 118, 119, 120, 121, 122, 123, 124, 125, 126, 127, 128, 129, 130, 131, 132, 133, 134, 135, 136, 137, 138, 139, 140, 141, 142, 143, 144, 145, 146, 147, 148, 149, 150, 151, 152, 153, 154, 155, 156, 157, 158, 159, 160, 161, 162, 163, 164, 165, 166, 167, 168, 169, 170, 171, 172, 173, 174, 175, 176, 177, 178, 179, 180, 181, 182, 183, 184, 185, 186, 187, 188, 189, 190, 191, 192, 193, 194, 195, 196, 197, 198, 199, 200, 201, 202, 203, 204, 205, 206, 207, 208, 209, 210, 211, 212, 213, 214, 215, 216, 217, 218, 219, 220, 221, 222, 223, 224, 225, 226, 227, 228, 229, 230, 231, 232, 233, 234, 235, 236, 237, 238, 239, 240, 241, 242, 243, 244, 245, 246, 247, 248, 249, 250, 251, 252, 253, 254, 255, 256, 257, 258, 259, 260, 261, 262, 263, 264, 265, 266, 267, 268, 269, 270, 271, 272, 273, 274, 275, 276, 277, 278, 279, 280, 281, 282, 283, 284, 285, 286, 287, 288, 289, 290, 291, 292, 293, 294, 295, 296, 297, 298, 299, 300, 301, 302, 303, 304, 305, 306, 307, 308, 309, 310, 311, 312, 313, 314, 315, 316, 317, 318, 319, 320, 321, 322, 323, 324, 325, 326, 327, 328, 329, 330, 331, 332, 333, 334, 335, 336, 337, 338, 339, 340, 341, 342, 343, 344, 345, 346, 347, 348, 349, 350, 351, 352, 353, 354, 355, 356, 357, 358, 359, 360, 361, 362, 363, 364, 365, 366, 367, 368, 369, 370, 371, 372, 373, 374, 375, 376, 377, 378, 379, 380, 381, 382, 383, 384, 385, 386, 387, 388, 389, 390, 391, 392, 393, 394, 395, 396, 397, 398, 399, 400, 401, 402, 403, 404, 405, 406, 407, 408, 409, 410, 411, 412, 413, 414, 415, 416, 417, 418, 419, 420, 421, 422, 423, 424, 425, 426, 427, 428, 429, 430, 431, 432, 433, 434, 435, 436, 437, 438, 439, 440, 441, 442, 443, 444, 445, 446, 447, 448, 449, 450, 451, 452, 453, 454, 455, 456, 457, 458, 459, 460, 461, 462, 463, 464, 465, 466, 467, 468, 469, 470, 471, 472, 473, 474, 475, 476, 477, 478, 479, 480, 481, 482, 483, 484, 485, 486, 487, 488, 489, 490, 491, 492, 493, 494, 495, 496, 497, 498, 499, 500, 501, 502, 503, 504, 505, 506, 507, 508, 509, 510, 511, 512, 513, 514, 515, 516, 517, 518, 519, 520, 521, 522, 523, 524, 525, 526, 527, 528, 529, 530, 531, 532, 533, 534, 535, 536, 537, 538, 539, 540, 541, 542, 543, 544, 545, 546, 547, 548, 549, 550, 551, 552, 553, 554, 555, 556, 557, 558, 559, 560, 561, 562, 563, 564, 565, 566, 567, 568, 569, 570, 571, 572, 573, 574, 575, 576, 577, 578, 579, 580, 581, 582, 583, 584, 585, 586, 587, 588, 589, 590, 591, 592, 593, 594, 595, 596, 597, 598, 599, 600, 601, 602, 603, 604, 605, 606, 607, 608, 609, 610, 611, 612, 613, 614, or 615 of any of SEQ ID NOs:2, 4, 6, or 8 (or any derivable range therein).


Amino acid substitutions are generally based on the relative similarity of the amino acid side-chain substituents, for example, their hydrophobicity, hydrophilicity, charge, size, and/or the like. An analysis of the size, shape and/or type of the amino acid side-chain substituents reveals that arginine, lysine and/or histidine are all positively charged residues; that alanine, glycine and/or serine are all a similar size; and/or that phenylalanine, tryptophan and/or tyrosine all have a generally similar shape. Therefore, based upon these considerations, arginine, lysine and/or histidine; alanine, glycine and/or serine; and/or phenylalanine, tryptophan and/or tyrosine; are defined herein as biologically functional equivalents.


In making such changes to produce biologically functional equivalents, the hydropathic index of amino acids may be considered. The hydropathy profile of a protein is calculated by assigning each amino acid a numerical value (“hydropathy index”) and then repetitively averaging these values along the peptide chain. Each amino acid has been assigned a value based on its hydrophobicity and charge characteristics. They are: isoleucine (+4.5); valine (+4.2); leucine (+3.8); phenylalanine (+2.8); cysteine/cysteine (+2.5); methionine (+1.9); alanine (+1.8); glycine (−0.4); threonine (−0.7); serine (−0.8); tryptophan (−0.9); tyrosine (−1.3); proline (1.6); histidine (−3.2); glutamate (−3.5); glutamine (−3.5); aspartate (−3.5); asparagine (−3.5); lysine (−3.9); and arginine (−4.5). The importance of the hydropathic amino acid index in conferring interactive biologic function on a protein is generally understood in the art (Kyte and Doolittle, 1982). It is accepted that the relative hydropathic character of the amino acid contributes to the secondary structure of the resultant protein, which in turn defines the interaction of the protein with other molecules, for example, enzymes, substrates, receptors, DNA, antibodies, antigens, and the like. It is also known that certain amino acids may be substituted for other amino acids having a similar hydropathy index or score, and still retain a similar biological activity. In making changes based upon the hydropathy index, in certain embodiments, the substitution of amino acids whose hydropathy indices are within ±2 is included. In some aspects of the disclosure, those that are within ±1 are included, and in other aspects of the disclosure, those within ±0.5 are included.


It also is understood in the art that the substitution of like amino acids can be made effectively on the basis of hydrophilicity. U.S. Pat. No. 4,554,101, incorporated herein by reference, states that the greatest local average hydrophilicity of a protein, as governed by the hydrophilicity of its adjacent amino acids, correlates with a biological property of the protein. In certain embodiments, the greatest local average hydrophilicity of a protein, as governed by the hydrophilicity of its adjacent amino acids, correlates with its immunogenicity and antigen binding, that is, as a biological property of the protein. The following hydrophilicity values have been assigned to these amino acid residues: arginine (+3.0); lysine (+3.0); aspartate (+3.0±1); glutamate (+3.0±1); serine (+0.3); asparagine (+0.2); glutamine (+0.2); glycine (0); threonine (−0.4); proline (−0.5±1); alanine (−0.5); histidine (−0.5); cysteine (−1.0); methionine (−1.3); valine (−1.5); leucine (−1.8); isoleucine (−1.8); tyrosine (−2.3); phenylalanine (−2.5); and tryptophan (−3.4). In making changes based upon similar hydrophilicity values, in certain embodiments, the substitution of amino acids whose hydrophilicity values are within ±2 are included, in other embodiments, those which are within ±1 are included, and in still other embodiments, those within ±0.5 are included. In some instances, one may also identify epitopes from primary amino acid sequences based on hydrophilicity. These regions are also referred to as “epitopic core regions.” It is understood that an amino acid can be substituted for another having a similar hydrophilicity value and still produce a biologically equivalent and immunologically equivalent protein.


Additionally, one skilled in the art can review structure-function studies identifying residues in similar polypeptides or proteins that are important for activity or structure. In view of such a comparison, one can predict the importance of amino acid residues in a protein that correspond to amino acid residues important for activity or structure in similar proteins. One skilled in the art may opt for chemically similar amino acid substitutions for such predicted important amino acid residues.


One skilled in the art can also analyze the three-dimensional structure and amino acid sequence in relation to that structure in similar proteins or polypeptides. In view of such information, one skilled in the art may predict the alignment of amino acid residues of an antibody with respect to its three-dimensional structure. One skilled in the art may choose not to make changes to amino acid residues predicted to be on the surface of the protein, since such residues may be involved in important interactions with other molecules. Moreover, one skilled in the art may generate test variants containing a single amino acid substitution at each desired amino acid residue. These variants can then be screened using standard assays for binding and/or activity, thus yielding information gathered from such routine experiments, which may allow one skilled in the art to determine the amino acid positions where further substitutions should be avoided either alone or in combination with other mutations. Various tools available to determine secondary structure can be found on the World Wide Web at expasy.org/proteomics/protein structure.


In some embodiments of the disclosure, amino acid substitutions are made that: (1) reduce susceptibility to proteolysis, (2) reduce susceptibility to oxidation, (3) alter binding affinity for forming protein complexes, (4) alter ligand or antigen binding affinities, and/or (5) confer or modify other physicochemical or functional properties on such polypeptides. For example, single or multiple amino acid substitutions (in certain embodiments, conservative amino acid substitutions) may be made in the naturally occurring sequence. Substitutions can be made in that portion of the antibody that lies outside the domain(s) forming intermolecular contacts. In such embodiments, conservative amino acid substitutions can be used that do not substantially change the structural characteristics of the protein or polypeptide (e.g., one or more replacement amino acids that do not disrupt the secondary structure that characterizes the native antibody).


As outlined above, amino acid substitutions generally are based on the relative similarity of the amino acid side-chain substituents, for example, their hydrophobicity, hydrophilicity, charge, size, and the like. Exemplary substitutions that take into consideration the various foregoing characteristics are well known and include: arginine and lysine; glutamate and aspartate; serine and threonine; glutamine and asparagine; and valine, leucine and isoleucine.


D. Methods of Making Recombinant Flaviviruses


The viruses of the present invention can be made using standard methods in the art. For example, an RNA molecule corresponding to the genome of a virus can be introduced into primary cells, chick embryos, or diploid cell lines, from which (or the supernatants of which) progeny virus can then be purified. Another method that can be used to produce the viruses employs heteroploid cells, such as Vero cells (Yasumura et al., Nihon Rinsho 21, 1201-1215, 1963). In this method, a nucleic acid molecule (e.g., an RNA molecule) corresponding to the genome of a virus is introduced into the heteroploid cells, virus is harvested from the medium in which the cells have been cultured, harvested virus is treated with a nuclease (e.g., an endonuclease that degrades both DNA and RNA, such as Benzonase™; U.S. Pat. No. 5,173,418), the nuclease-treated virus is concentrated (e.g., by use of ultrafiltration using a filter having a molecular weight cut-off of, e.g., 500 kDa), and the concentrated virus is formulated for the purposes of vaccination. Details of this method are provided in WO 03/060088 A2, which is incorporated herein by reference.


In some embodiments, a method of manufacturing an attenuated flavivirus disclosed herein comprises introducing a nucleic acid construct encoding the genome of the attenuated flavivirus into cells and isolating flavivirus produced in the cells from the cells or the supernatant thereof. In some embodiments, the cells are Vero cells. In some embodiments, the cells are cultured in serum free medium.


IV. FLAVIVIRUS INFECTION PREVENTION AND TREATMENT

Aspects of the present disclosure are directed to compositions and methods of using such compositions to treat or prevent flavivirus infection in a subject. In some embodiments, the flavivirus is selected from the group consisting of WNV, Japanese encephalitis virus, St. Louis encephalitis virus, tickborne encephalitis virus, dengue fever virus, and YFV. In some embodiments, the flavivirus is WNV. In some embodiments, the flavivirus is YFV.


In some embodiments, the attenuated flaviviruses or polynucleotides that encode the genome of the attenuated flaviviruses comprise the nucleic acid sequence of SEQ ID NOs:1, 3, 5, or 7.


In some aspects, the disclosure is directed to an immunogenic composition for inducing an immune response in a subject comprising an attenuated flavivirus disclosed herein and a pharmaceutically acceptable carrier or diluent. In some embodiments, the attenuated flavivirus induces the same immune response in the subject as the corresponding wild type virus. In some aspects, the disclosure is directed to a method of inducing an immune response in a subject comprising administering an effective amount of an immunogenic composition for inducing an immune response in a subject comprising an attenuated flavivirus disclosed herein and a pharmaceutically acceptable carrier or diluent to the subject. In some embodiments, an immune response to at least one of WNV, Japanese encephalitis virus, St. Louis encephalitis virus, tickborne encephalitis virus, dengue fever virus, and YFV is induced in the subject. In some embodiments, the immune response induced in the subject is to WNV. In some embodiments, the immune response induced in the subject is to YFV.


In some aspects, the disclosure is directed to a vaccine composition comprising an attenuated flavivirus disclosed herein and a pharmaceutically acceptable carrier or diluent. In some aspects, the disclosure is directed to a method of immunizing a subject against a flavivirus infection comprising administering an effective amount of a vaccine composition comprising an attenuated flavivirus disclosed herein to the subject.


In certain embodiments, the disclosed methods further comprise treating a subject who does not have, but is at risk of developing, infection by a flavivirus. In certain embodiments, the disclosed methods comprise treating a subject who is infected by a flavivirus. In certain embodiments, the disclosed methods further comprise treating a subject who has been diagnosed as having symptoms of a flavivirus infection. In certain embodiments, the disclosed methods further comprise treating a subject who has been identified as being at risk of having a flavivirus infection. A subject may be diagnosed with or as having symptoms of or may be identified as being at risk of having a flavivirus infection using tests and diagnostic methods known in the art and described herein.


In some embodiments, the methods further comprise determining a subject is in need of treatment comprising a therapeutically effective amount of an immunogenic or vaccine composition comprising an attenuated flavivirus disclosed herein. In some embodiments, the methods further comprise providing to a subject a treatment comprising a therapeutically effective amount of an immunogenic or vaccine composition comprising an attenuated flavivirus disclosed herein when it is determined that the subject is in need thereof. In some embodiments, determining a subject is in need of a treatment comprising a therapeutically effective amount of an immunogenic or vaccine composition comprising an attenuated flavivirus disclosed herein comprises diagnosing the subject with a flavivirus infection. In some embodiments, determining a subject is in need of a treatment comprising a therapeutically effective amount of an immunogenic or vaccine composition comprising an attenuated flavivirus disclosed herein comprises diagnosing the subject as having symptoms of a flavivirus infection. In some embodiments, determining a subject is in need of a treatment comprising a therapeutically effective amount of an immunogenic or vaccine composition comprising an attenuated flavivirus disclosed herein comprises identifying the subject as being at risk of having a flavivirus infection.


In some embodiments, the disclosed methods comprise administering to a subject suffering from a flavivirus infection a therapeutically effective amount of an immunogenic or vaccine composition comprising an attenuated flavivirus disclosed herein. As disclosed herein, flavivirus infections can be associated with neuroinvasion, neurotropism, and/or viscerotropism, and administration of an immunogenic or vaccine composition comprising an attenuated flavivirus disclosed herein has been surprising and unexpectedly found to prevent or decease neuroinvasion. Further, administering an immunogenic or vaccine composition comprising an attenuated flavivirus disclosed herein can surprisingly prevent or decrease cytokine induction in infected cells. Thus, in some embodiments, the one or more mutations decrease cytokine response to the flavivirus, relative to a corresponding flavivirus lacking the mutation. Accordingly, some embodiments are directed to compositions and corresponding methods for treating a subject suffering from flavivirus infection with a therapeutically effective amount of an immunogenic or vaccine composition comprising an attenuated flavivirus disclosed herein. In some embodiments, the immunogenic or vaccine composition comprising an attenuated flavivirus induces an immune response in a host.


The attenuated flaviviruses disclosed herein also demonstrate surprising genetic stability. Thus, in some embodiments, one or more mutations in a nucleic acid construct encoding the genome of the attenuated flavivirus inhibits reversion of the one or more mutations to the wild-type amino acid is inhibited when the virus is grown in a host.


In further embodiments, disclosed is an attenuated flavivirus, wherein one or more mutations in a nucleic acid construct encoding the genome of the attenuated flavivirus decrease the viscerotropism of the flavivirus, relative to a corresponding flavivirus lacking the mutation. Also disclosed is a method for decreasing viscerotropism of the flavivirus in vivo, comprising contacting at least one cell of a host with a therapeutically effective amount of an immunogenic or vaccine composition comprising an attenuated flavivirus disclosed herein. In further embodiments, disclosed is an attenuated flavivirus, wherein one or more mutations in a nucleic acid construct encoding the genome of the attenuated flavivirus decrease the neurotropism of the flavivirus, relative to a corresponding flavivirus lacking the mutation. Also disclosed is a method for decreasing neurotropism of the flavivirus in vivo, comprising contacting at least one cell of a host with a therapeutically effective amount of an immunogenic or vaccine composition comprising an attenuated flavivirus disclosed herein. In some embodiments, the flavivirus infection is one which is characterized by neuroinvasion, neurotropism, and/or viscerotropism.


The subject may have a condition that has as a symptom and/or a neuroinvasion, neurotropism, and/or viscerotropism, for example. Embodiments of the disclosure include treatment or prevention of any medical condition in which modulation of neuroinvasion, neurotropism, and/or viscerotropism by flaviviruses would be beneficial. In specific embodiments, an individual is provided a therapeutically effective amount of an immunogenic or vaccine composition comprising an attenuated flavivirus for attenuation of neuroinvasion, neurotropism, and/or viscerotropism in an individual or a delay or reversal in neuroinvasion, neurotropism, and/or viscerotropism in an individual. In specific embodiments, the medical condition treated or prevented with an immunogenic or vaccine composition comprising an attenuated flavivirus comprises flavivirus infections which can lead to neuroinvasion, neurotropism, and/or viscerotropism. In particular embodiments, neuroinvasion, neurotropism, and/or viscerotropism is not treated with compositions the disclosure. In some cases, an immunogenic or vaccine composition comprising an attenuated flavivirus treats or prevents the medical condition in the individual by ameliorating, inhibiting, delaying, or reversing neuroinvasion, neurotropism, and/or viscerotropism, for example.


Although in some cases the immunogenic or vaccine composition comprising an attenuated flavivirus is provided as a sole therapy for the individual, in other cases the individual is provided one or more additional therapies for treating or preventing flavivirus infection, neuroinvasion, neurotropism, and/or viscerotropism. The one or more additional therapies may be of any kind, but in specific cases the one or more additional therapies are one or more additional anti-viral therapeutics, for example, anti-flaviviral medications. In some embodiments, the one or more additional therapies are one or more additional therapeutics to treat symptoms of flaviviral infection.


V. FLAVIVIRUS IMMUNOGENIC COMPOSITIONS AND VACCINES

The flaviviruses disclosed herein provide live, attenuated viruses useful as immunogens or vaccines. In one embodiment, the flaviviruses exhibit high immunogenicity while at the same time not producing dangerous pathogenic or lethal effects. As used herein, an immunogen or vaccine is said to prevent or attenuate a disease if administration of the immunogen or vaccine to an individual results either in the total or partial immunity of the individual to the disease, or in the total or partial attenuation (i.e., suppression) of symptoms or conditions associated with the disease. Accordingly, the invention relates to a method for raising a protective immune response in a human subject, the method comprising administering a therapeutically effective amount of an immunogenic composition or vaccine as described anywhere throughout the specification to the subject. Also provided herein is a method of providing immune protection in humans against flavivirus infection comprising administering an effective amount of the attenuated flavivirus compositions of the disclosure to the subject, thereby providing protection from flavivirus infection. The disclosure also relates to a method for raising a protective immune response in a subject, the method comprising administering a therapeutically effective amount of an immunogenic composition comprising an attenuated flavivirus. Other aspects of this invention also describe the use of a composition as described above or throughout the specification for the manufacture of a medicament for the treatment or prevention of flavivirus infection or disease caused thereby.


The viruses of this disclosure can comprise the structural and non-structural genes of a WNV or a YFV, for example. The strategy described herein of using a genetic background that contains the viral genomes, and, by mutation, the properties of attenuation, has led to the development of live, attenuated flavivirus vaccine candidates of desired immunogenicity. Thus, vaccine candidates for control of flavivirus pathogens, for example, WNV or YFV, can be designed.


Viruses described herein are typically grown using techniques known in the art. Virus plaque or focus forming unit (FFU) titrations are then performed and plaques or FFU are counted in order to assess the viability, titer and phenotypic characteristics of the virus grown in cell culture. Wild type viruses are mutagenized to derive attenuated candidate starting materials.


Immunogenic compositions or vaccines may either be prophylactic (to prevent infection) or therapeutic (to treat disease after infection). The viruses of the invention can be administered as primary prophylactic agents in those at risk of infection, or can be used as secondary agents for treating infected subjects. Because the viruses are attenuated, they are particularly well suited for administration to “at risk individuals” such as the elderly, children, or HIV infected persons. The immunogenic compositions or vaccines can also be used in veterinary contexts, e.g., in the vaccination of horses against WNV infection, or in the vaccination of birds (e.g., valuable, endangered, or domestic birds, such as flamingos, bald eagles, and geese, respectively). Further, the immunogenic compositions or vaccines can include a virus, such as a virus, including a particular mutation, in a mixture with viruses lacking such mutations.


Such immunogenic compositions or vaccines comprise antigen or antigens, usually in combination with “pharmaceutically acceptable carriers” or “acceptable carriers”, as may be used interchangeably as will be clear from the context, which include any carrier that does not itself induce the production of antibodies harmful to the individual receiving the composition.


A. Formulation


Formulation of the viruses of the invention can be carried out using methods that are standard in the art. Embodiments provide for administration of compositions to subjects in a biologically compatible form suitable for pharmaceutical administration in vivo. By “biologically compatible form suitable for administration in vivo,” it is meant a form of the active agent (e.g. pharmaceutical protein, peptide, or gene etc. of the embodiments) to be administered in which any toxic effects are outweighed by the therapeutic effects of the active agent. Numerous pharmaceutically acceptable solutions for use in vaccine preparation are well known and can readily be adapted for use in the present invention by those of skill in this art (see, e.g., Remington's Pharmaceutical Sciences (18th edition), ed. A. Gennaro, 1990, Mack Publishing Co., Easton, Pa.). In two specific examples, the viruses are formulated in Minimum Essential Medium Earle's Salt (MEME) containing 7.5% lactose and 2.5% human serum albumin or MEME containing 10% sorbitol. However, the viruses can simply be diluted in a physiologically acceptable solution, such as sterile saline or sterile buffered saline. In another example, the viruses can be administered and formulated, for example, in the same manner as the YFV 17D vaccine, e.g., as a clarified suspension of infected chicken embryo tissue, or a fluid harvested from cell cultures infected with a chimeric virus.


The viruses described herein are individually or jointly combined with a pharmaceutically acceptable carrier or vehicle for administration as an immunogen or vaccine to humans or animals. The terms “pharmaceutically acceptable carrier” or “pharmaceutically acceptable vehicle” are used herein to mean any composition or compound including, but not limited to, water or saline, a gel, salve, solvent, diluent, fluid ointment base, liposome, micelle, giant micelle, and the like, which is suitable for use in contact with living animal or human tissue without causing adverse physiological responses, and which does not interact with the other components of the composition in a deleterious manner.


Suitable carriers are typically large, slowly metabolized macromolecules such as proteins, polysaccharides, polylactic acids, polyglycolic acids, polymeric amino acids, amino acid copolymers, lipid aggregates (such as oil droplets or liposomes), and inactive virus particles. Such carriers are well known to those of ordinary skill in the art. Additionally, these carriers may function as immunostimulating agents (“adjuvants”). Furthermore, the antigen may be conjugated to a toxoid, such as a toxoid from diphtheria, tetanus, cholera, H. pylori, etc. Adjuvants to enhance effectiveness of the composition include, but are not limited to: (1) aluminum salts (alum), such as aluminum hydroxide, aluminum phosphate, aluminum sulfate, etc; (2) oil-in-water emulsion formulations (with or without other specific immunostimulating agents such as muramyl peptides (see below) or bacterial cell wall components), such as for example (a) MF59 (PCT Publ. No. WO 90/14837), containing 5% Squalene, 0.5% Tween 80, and 0.5% Span 85 (optionally containing various amounts of MTP-PE (see below), although not required) formulated into submicron particles using a microfluidizer such as Model 110Y microfluidizer (Microfluidics, Newton, Mass.), (b) SAF, containing 10% Squalane, 0.4% Tween 80, 5% pluronic-blocked polymer L 121, and thr-MDP (see below) either microfluidized into a submicron emulsion or vortexed to generate a larger particle size emulsion, and (c) Ribi TM adjuvant system (RAS), (Ribi Immunochem, Hamilton, Mont.) containing 2% Squalene, 0.2% Tween 80, and one or more bacterial cell wall components from the group consisting of monophosphorolipid A (MPL), trehalose dimycolate (TDM), and cell wall skeleton (CWS), preferably MPL+CWS (Detox™); (3) saponin adjuvants, such as Stimulon TM (Cambridge Bioscience, Worcester, Mass.) may be used or particles generated therefrom such as ISCOMs (immunostimulating complexes); (4) Complete Freunds Adjuvant (CFA) and Incomplete Freunds Adjuvant (IFA); (5) cytokines, such as interleukins (IL-1, IL-2, etc.), macrophage colony stimulating factor (M-CSF), tumor necrosis factor (TNF), etc; and (6) other substances that act as immunostimulating agents to enhance the effectiveness of the composition. Alum and MF59 are preferred. As mentioned above, muramyl peptides include, but are not limited to, N-acetyl-muramyl-L-threonyl-D-isoglutamine (thr-MDP), N-acetyl-nornuramyl-L-alanyl-D-isoglutamine (nor-MDP), N-acetylmuramyl-L-alanyl-D-isoglutaminyl-L-alanine-2-(1′-2′-dipalmitoyl-sn-glycero-3-huydroxyphosphoryloxy)-ethylamine (MTPPE), etc.


The immunogenic compositions (e.g., the antigen, pharmaceutically acceptable carrier, and adjuvant) typically will contain diluents, such as water, saline, glycerol, ethanol, etc. Additionally, auxiliary substances, such as wetting or emulsifying agents, pH buffering substances, and the like, may be present in such vehicles. Typically, the immunogenic compositions are prepared as injectables, either as liquid solutions or suspensions; solid forms suitable for solution in, or suspension in, liquid vehicles prior to injection may also be prepared. The preparation also may be emulsified or encapsulated in liposomes for enhanced adjuvant effect, as discussed above under pharmaceutically acceptable carriers. Immunogenic compositions of the present disclosure elicit formation of antibodies with high binding specificity to a composition of a flavivirus.


Such immunogenic compositions used as vaccines comprise an immunologically effective amount of the antigenic polypeptides, as well as any other of the above-mentioned components, as needed. By “immunologically effective amount”, or “therapeutically effective amount” as may be used interchangeably and as will be clear from the context, it is meant that the administration of that amount to an individual or host subject animal, either in a single dose or as part of a series, is effective for treatment or prevention or the amount effective at various dosages and for periods of time necessary to achieve the desired result. For example, the active ingredients of the compositions of the invention are present in a therapeutically effective amount if the administration of the composition to a subject results in a detectable change in the physiology of the recipient subject, such as the induction of a neutralizing antibody against the attenuated flavivirus. This amount varies depending upon the health and physical condition of the individual or host subject animal to be treated, the taxonomic group of individual to be treated (e.g., nonhuman primate, primate, etc.), the capacity of the individual's immune system to synthesize antibodies, the degree of protection desired, the formulation of the vaccine, the treating doctor's assessment of the medical situation, and other relevant factors. For example, effective amounts of the compositions can vary from 0.01-500 μg per product per dose, 1-100 μg per product per dose, or 5-50 μg per product per dose. Dosage regima may be adjusted to provide the optimum therapeutic response. It is expected that the amount will fall in a relatively broad range that can be determined through routine trials.


The immunogenic compositions are conventionally administered parenterally, e.g., by injection, either subcutaneously or intramuscularly. Additional formulations suitable for other modes of administration include oral and pulmonary formulations, suppositories, and transdermal applications. Dosage treatment may be a single dose schedule or a multiple dose schedule. The vaccine may be administered in conjunction with other immunoregulatory agents.


The immunogenic or vaccine formulations may be conveniently presented in viral plaque forming unit (PFU) unit or focus forming unit (FFU) dosage form and prepared by 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 non-aqueous 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 non-aqueous 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 ampoules and vials, and may be stored in a freeze-dried (lyophilized) condition requiring only the addition of the 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.


Pharmaceutical compositions suitable for injectable use may be administered by means known in the art. For example, sterile aqueous solutions (where water soluble) or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersion may be used. In all cases, the composition can be sterile and can be fluid to the extent that easy syringability exists. It might be stable under the conditions of manufacture and storage and may be preserved against the contaminating action of microorganisms such as bacteria and fungi. The pharmaceutically acceptable carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyetheylene glycol, and the like), and suitable mixtures thereof. The proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants. Prevention of microorganisms can be achieved by heating, exposing the agent to detergent, irradiation or adding various antibacterial or antifungal agents.


Sterile injectable solutions can be prepared by incorporating active compound in the required amount with one or a combination of ingredients enumerated above, as required. Aqueous compositions can include an effective amount of a therapeutic compound, peptide, epitopic core region, stimulator, inhibitor, and the like, dissolved or dispersed in a pharmaceutically acceptable carrier or aqueous medium. Compounds and biological materials disclosed herein can be purified by means known in the art.


In another embodiment, nasal solutions or sprays, aerosols or inhalants may be used to deliver the compound of interest. Additional formulations that are suitable for other modes of administration include suppositories and pessaries. A rectal pessary or suppository may also be used. In general, for suppositories, traditional binders and carriers may include, for example, polyalkylene glycols or triglycerides; such suppositories may be formed from mixtures containing the active ingredient in the range of 0.5% to 10%, for example, 1% 2%.


Upon formulation, solutions will be administered in a manner compatible with the dosage formulation and in such amount as is therapeutically effective. The formulations are easily administered in a variety of dosage forms, such as the type of injectable solutions described above. It is contemplated that slow release capsules, timed-release microparticles, and the like can also be employed. These particular aqueous solutions are especially suitable for intravenous, intramuscular, subcutaneous and intraperitoneal administration.


The composition may be stored at temperatures of from about −100° C. to about 4° C. The composition may also be stored in a lyophilized state at different temperatures including room temperature. The composition may be sterilized through conventional means known to one of ordinary skill in the art. Such means include, but are not limited to, filtration. The composition may also be combined with bacteriostatic agents to inhibit bacterial growth.


B. Administration


The vaccines of the invention can be administered using methods that are well known in the art, and appropriate amounts of the vaccines to be administered can readily be determined by those of skill in the art. What is determined to be an appropriate amount of virus to administer can be determined by consideration of factors such as, e.g., the size and general health of the subject to whom the virus is to be administered. For example, the viruses of the invention can be formulated as sterile aqueous solutions containing between 102 and 108, e.g., 103 to 10′, infectious units (e.g., plaque-forming units or tissue culture infectious doses) in a dose volume of 0.1 to 1.0 ml, to be administered. In addition, because flaviviruses may be capable of infecting the human host via mucosal routes, such as the oral route (Gresikova et al., “Tick-borne Encephalitis,” In The Arboviruses, Ecology and Epidemiology, Monath (ed.), CRC Press, Boca Raton, Fla., 1988, Volume IV, 177-203), the viruses can be administered by mucosal routes as well. Preferred 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, the formulations of the present invention may include other agents commonly used by one of ordinary skill in the art.


The immunogenic or vaccine composition may be administered through different routes, such as oral or parenteral, including, but not limited to, buccal and sublingual, rectal, aerosol, nasal, intramuscular, subcutaneous, intradermal, and topical. The composition may be administered in different forms, including, but not limited to, solutions, emulsions and suspensions, microspheres, particles, microparticles, nanoparticles and liposomes.


Further, the vaccines of the invention can be administered in a single dose or, optionally, administration can involve the use of a priming dose followed by a booster dose that is administered, as determined to be appropriate by those of skill in the art. In some embodiments, if multiple immunizations are given, they are given one to two months apart. In some embodiments, a subject is immunized at 0, 1, and 2 months. In some embodiments, a subject is immunized at 0, 1, and 3 months. In some embodiments, a subject is immunized at 0, 1, and 6 months.


In some embodiments, 1 to about 6 doses may be required per immunization schedule. Initial doses may range from about 100 to about 100,000 PFU or FFU, about 500 to about 20,000 PFU or FFU, about 1000 to about 12,000 PFU or FFU and about 1000 to about 4000 PFU or FFU. Booster injections may range in dosage from about 100 to about 20,000 PFU or FFU, about 500 to about 15,000, about 500 to about 10,000 PFU or FFU, and about 1000 to about 5000 PFU or FFU. For example, the volume of administration will vary depending on the route of administration. Intramuscular injections may range in volume from about 0.1 ml to 1.0 ml. In certain embodiments, a dual dose on day 0 may be administered in single anatomical or multiple anatomical locations in order to induce an immune response with reduced interference or different lymph nodes.


Optionally, adjuvants that are known to those skilled in the art can be used in the administration of the viruses of the invention. Such adjuvants include, but are not limited to, the following: polymers, co-polymers such as polyoxyethylene-polyoxypropylene copolymers, including block co-polymers, polymer p 1005, Freund's complete adjuvant (for animals), Freund's incomplete adjuvant; sorbitan monooleate, squalene, CRL-8300 adjuvant, alum, QS 21, muramyl dipeptide, CpG oligonucleotide motifs and combinations of CpG oligonucleotide motifs, trehalose, bacterial extracts, including mycobacterial extracts, detoxified endotoxins, membrane lipids, or combinations thereof. Further adjuvants that can be used to enhance the immunogenicity of the viruses include, for example, liposomal formulations, synthetic adjuvants, such as (e.g., QS21), muramyl dipeptide, monophosphoryl lipid A, polyphosphazine, CpG oligonucleotides, or other molecules that appear to work by activating Toll-like Receptor (TLR) molecules on the surface of cells or on nuclear membranes within cells. Although these adjuvants are typically used to enhance immune responses to inactivated vaccines, they can also be used with live vaccines. Both agonists of TLRs or antagonists may be useful in the case of live vaccines. In the case of a virus delivered via a mucosal route, for example, orally, mucosal adjuvants such as the heat-labile toxin of E. coli (LT) or mutant derivations of LT can be used as adjuvants. In addition, genes encoding cytokines that have adjuvant activities can be inserted into the viruses. Thus, genes encoding cytokines, such as GM-CSF, IL-2, IL-12, IL-13, or IL-5, can be inserted together with foreign antigen genes to produce a vaccine that results in enhanced immune responses, or to modulate immunity directed more specifically towards cellular, humoral, or mucosal responses.


VI. KITS

Other embodiments concern kits of use with the methods (e.g. methods of application or administration of a vaccine or immunogenic composition) and compositions described herein. Some embodiments concern kits having vaccine or immunogenic compositions of use to prevent or treat subjects having, exposed or suspected of being exposed to one or more flaviviruses. In certain embodiments, a kit may contain one or more than one formulation of attenuated vaccines at predetermined ratios. Kits can be portable, for example, able to be transported and used in remote areas such as military installations or remote villages. Other kits may be of use in a health facility to treat a subject having been exposed to one or more flaviviruses or suspected of being at risk of exposure to flaviviruses.


Kits can also include a suitable container, for example, vials, tubes, mini- or microfuge tubes, test tube, flask, bottle, syringe or other container. Where an additional component or agent is provided, the kit can contain one or more additional containers into which this agent or component may be placed. Kits herein will also typically include a means for containing the agent, composition and any other reagent containers in close confinement for commercial sale. Such containers may include injection or blow-molded plastic containers into which the desired vials are retained. Optionally, one or more additional agents such as immunogenic agents or other anti-viral agents, anti-fungal or antibacterial agents may be needed for compositions described, for example, for compositions of use as a vaccine against one or more additional microorganisms.


VII. EXAMPLES

The following examples as well as the figures are included to demonstrate preferred embodiments of the disclosure. It should be appreciated by those of skill in the art that the techniques disclosed in the examples or figures represent techniques discovered by the inventors to function well in the practice of the disclosure, and thus can be considered to constitute preferred modes for its practice. However, those of skill in the art should, in light of the present disclosure, appreciate that many changes can be made in the specific embodiments which are disclosed and still obtain a like or similar result without departing from the spirit and scope of the disclosure.


Example 1— Rescue of New West Nile Virus (WNV) NS4B Mutants

The inventors were interested in mutating residues that are shared between different mosquito-borne flaviviruses in order to correlate results obtained from WNV to other flaviviruses. Accordingly, residue P54 was targeted for mutation based on its homology in multiple flaviviruses (FIG. 1). Besides conservation in the flavivirus genus, the specified amino acid residue was selected due to its putative location in the NS4B protein, thus providing a means of investigation of a specific region of NS4B for attenuating mutations. Specifically, P54 is in the first transmembrane domain of NS4B (FIG. 2).


The genotypes of NS4B-P54 mutants are shown in Table 1. In total, two new NS4B mutants were generated including P54A and P54G. The genomic consensus sequence of each virus was verified using next-generation sequencing (NGS), and neither of the new mutants had any compensatory consensus mutations upon rescue. After a single passage in Vero cells, neither of the P54 mutants had additional consensus sequence mutations. All viruses had similar infectivity titers, with NY99ic producing large plaques while the NS4B-P54 mutants produced medium-sized plaques (Table 1).


All viruses were rescued from transfection and passaged once in Vero cells for subsequent studies. Infectivity titers are listed as log 10 PFU/mL. Temperature sensitivity significance was measured using a one-way ANOVA with Bonferroni's multiple comparisons. ***p 0.001.









TABLE 1







Consensus genotypes, plaque phenotypes, temperature sensitivity, and mouse attenuation of NS4B mutants



















Vero





Survived





Additional
cell



Δ titer
Survival
10,000

High dose


NS4B
consensus
passage
Plaque
37° C.
41° C.
41° vs
500 PFU
PFU
High dose
survival


mutant
mutations
number
morphology
titer
titer
37° C.
i.p. (%)
challenge
(PFU/mouse)
(%)




















NY99ic

1
large
8.2
8.1
0.1
0/10 (0)  
n.d.
n.d.
n.d.


P54A

1
medium
8.3
7.3
***1.0
 5/5 (100)
5/5
3.5 × 105
 5/5 (100)











5.4 × 107
 5/5 (100)


P54G

1
medium
8.2
8.0
0.2
 5/5 (100)
5/5
5.2 × 105
 5/5 (100)











1.5 × 107
5/10 (50) 





Infectivity titers are listed as log10 PFU/mL. Temperature sensivity significance was measured using a one-way ANOVA with Bonferroni’s multiple


comparisons. Abbreviations: n.d. = not done, ***p < 0.001.






Example 2— Multiplication Kinetics of WNV Mutants in Vero and A549 Cells

Multiplication kinetics of the NS4B-P54 mutants were compared to that of parental NY99ic using a MOI of 0.1 in both Vero (Type I Interferon [IFN-I] deficient) and A549 cells (IFN-I competent). In both cell lines, the mutants had similar multiplication kinetics to those of NY99ic (FIG. 3). The P54A mutant had the most significantly reduced kinetics compared to NY99ic. Specifically, in Vero cells the P54A mutant grew to titers >100-fold lower than those of NY99ic at early time points, but by two dpi, the mutant and NY99ic exhibited similar infectivity titers (FIG. 3A). In A549 cells, the P54A mutant multiplied to titers >10-fold below those of NY99ic at all time points between 1-4 dpi (FIG. 3B). Despite the observed differences from NY99ic, both NS4B-P54 mutants were able to multiply in each cell line to relatively high titers >6 log10 PFU/mL.


Example 3— Temperature Sensitivity of WNV Mutants

To investigate temperature sensitive (TS) phenotypes, infectivity titers were compared at both 37° C. and 41° C. While the P54G mutant was similar to NY99ic in that it did not exhibit significant reduction of infectivity titer at 41° C., P54A was significantly TS (p <0.0001) (Table 1). Specifically, the P54A mutant had a 10-fold reduction in titer at 41° C. compared to 37° C.


Example 4— Mouse Neuroinvasive Phenotype of WNV Mutants

To investigate attenuation of mouse neuroinvasion, experiments were undertaken in outbred mice as they are a highly susceptible model for WNV infection. First, each of the new mutants was tested using i.p. inoculation of 500 PFU, and then were tested at a high dose by the i.p. route where the virus was administered undiluted (undiluted doses listed in Table 1).


All (5/5) mice inoculated with 500 PFU of either P54A or P54G mutants survived infection (Table 1). Furthermore, all surviving mice were challenged with 10,000 PFU of NY99ic at 36 dpi, and all challenged mice survived (Table 1); therefore, mutation of P54A and P54G conferred an attenuated phenotype and also induced protective immunity.


To investigate the attenuated phenotype further, groups of mice were inoculated with undiluted virus. P54A was tested at both 350,000 and 54 million PFU doses, and P54G was tested at both 520,000 and 15 million PFU doses. Both the P54A and P54G mutants caused no lethality (5/5 mice) following inoculation of >300,000 PFU (Table 1). All (5/5) mice survived a 54 million PFU dose of the P54A mutant, while 50% (5/10) of mice survived a 15 million PFU dose of the P54G mutant (Table 1).


Example 5— Cytokine Induction of WNV Mutants in A549 Cells

Using a BioPlex Pro 27-plex human cytokine assay and a custom IFN-α/IFN-β assay, 29 cytokines were measured in A549 cell culture supernatant at 36 hpi. Cells were infected with both new mutants, the NS4B-P38G (+NS3-N480H/NS4B-T1161) mutant as an attenuated control, NY99ic, or PBS as a mock infection. Two cytokines (IL-10 and IL-15) were not detected at all, and four cytokines (IL-1β, IL-12 p70, IL-13, and PDGF-bb) were either detected at very low levels (<1 pg/mL) or were only sporadically measured in fewer than half of the replicates, and therefore, these six cytokines were not analyzed for significance.


Several patterns were detected in the cytokine profiles of the NS4B mutants. Cells infected with the P38G (+N53-N480H/NS4B-T1161) mutant produced higher pro-inflammatory cytokine and chemokine responses than NY99ic-infected cells (FIG. 4), which was consistent with previous studies of cytokine induction from this mutant in immune cells and a mouse model [35,36]. In comparison, both attenuated P54A and P54G mutants induced relatively low cytokine responses that were typically similar to those in mock-infected cells and lower than those in NY99ic-infected cells (FIG. 4). The only cytokine for which all attenuated mutants had a similar trend that was distinct from the virulent viruses was IFN-β, where P38 and P54 attenuated mutants decreased production compared to NY99ic, albeit the P38G mutant was the only one for which the difference was statistically significant (FIG. 4).


Example 6— Stability of WNV NS4B Mutant Genotypes

To investigate the stability of each NS4B mutation, SNVs were analyzed in the cell culture stocks of each virus. Viruses that were passaged once in Vero cells (P1) were analyzed since these were the stocks utilized in the mouse studies. Each of these stocks had similar depths of coverage of sequencing reads with the average coverage ranging from 7217-7796, therefore, stocks were analyzed without down sampling. The total number and frequency of SNVs identified varied between the different viruses, and there was no obvious pattern to correlate SNV number or frequency with attenuation (FIG. 5). Although SNVs as low as 0.1% frequency were detected, analysis was concentrated on SNVs ≥1% to focus on the variants of the greatest significance. Comparison of the SNVs of the new NS4B mutants demonstrated that there were no SNVs that were shared amongst all of the mutants and none that appeared associated with attenuated or virulent phenotypes of the mutants (Table 2).


Most SNVs detected in the P54G mutant quasispecies were approximately 1% frequency with only 10% (3/30) that were >5% frequency in the population, and none were detected in the NS4B gene (FIG. 6, Table 2). In comparison, of 20 SNVs ≥1% frequency in the P54A mutant quasispecies, 50% (10/20) had a frequency >10% and 45% (9/20) were clustered in the NS4B gene (FIG. 6, Table 2). The nine nucleotide changes in NS4B encoded seven amino acid substitutions, including one that exhibited reversion of NS4B-A54P in 13.0% of the population (FIG. 7).









TABLE 2







NS4B-P54A mutation reverted during cell culture passage,


but NS4B-P54G mutation did not












Mutant
P0
P1
P5







P54A
18.0%
13.0%
69.3%



P54G
<0.1%
<0.1%
<0.1%







The frequency of reversion to the wild-type proline residue of NS4B-P54 mutants were measured in Vero cell P0, P1, and P5 virus stocks. The limit of detection was 0.1%.






To further investigate the stability of the P54A mutation, Vero cell P0 and P5 stocks were sequenced and variants were analyzed. The P0 stock of the P54A mutant (the virus rescued directly after transfection) had slightly lower average sequencing coverage than the other mutants analyzed (6165 compared to >7216), but regardless, the P0 stock had a similar genotype to the P1 stock. Specifically, no extra coding consensus sequence changes were identified throughout the genome and of 20 SNVs >1% frequency, one encoded reversion in 18.0% of the population (Table 3). In comparison, the P5 stock had 16 SNVs >1% frequency and had undergone consensus sequence reversion and harbored both NS4B-A54P and NS4B-D90G consensus genome mutations (Table 3). It is notable that during serial passage of the P54A mutant, the viral titer did not significantly change, but the plaque phenotype changed from medium-sized (P0 and P1) to mixed medium and large sized plaques (P5). Based on the instability of the P54A mutation, P0 and P5 stocks of the P54G mutant were also analyzed. The P0 stock had an average coverage depth of only 1729, which was significantly lower sequencing coverage depth than the other viruses analyzed (the P54G mutant P1 and P5 stocks had average coverage of 7655 and 7605 reads, respectively). Therefore, the SNV results of this stock may be biased since there were fewer viral reads. With this limitation in mind, the P0 stock of the P54G mutant was similar to the P1 stock in that it harbored 34 SNVs >1% frequency, none of which were in NS4B. The P5 stock of the P54G mutant had 18 SNVs >1% frequency, and although six were in NS4B, there was no evidence of reversion of the P54G mutation and there were no additional consensus sequence changes (Table 4). Consistent with the sequencing results, the viral titer and the medium-size plaque phenotype remained constant during five serial passages of the P54G mutant.









TABLE 3







WNV NS4B mutants exhibited distinct single nucleotide variants















Nucleotide
Major
Minor
Viral Protein
Major
Minor
Frequency



Position
Nucleotide
Nucleotide
Position
Residue
Residue
(%)

















NY99ic
2135
A
G
E-390
E
G
1.9


P54A
904
A
U
prM-147
S
S
7.3



1435
A
G
E-157
T
A
10.7



6347
A
G
NS3-579
E
G
23.2



7000
A
C
NS4B-29
M
L
6.9



7075
G
C
NS4B-54
*A
*p
13.0



7077
C
A
NS4B-54


12.7



7184
A
G
NS4B-90
D
G
35.7



7189
G
A
NS4B-92
G
R
29.5



7223
G
A
NS4B-103
*W
*Y
14.6



7224
G
U
NS4B-103


14.3



7420
A
C
NS4B-169
M
L
5.9



7627
C
G
NS4B-238
L
V
7.1



8973
A
C
NS5-431
P
P
10.6



8973
A
U
NS5-431
P
P
6.9



9956
U
G
NS5-759
L
R
9.6



10342
A
C
NS5-888
M
L
8.0



10440
A
G
3′ UTR


23.6



10464
A
G
3′ UTR


2.5



10582
G
U
3′ UTR


1.4



10814
A
U
3′ UTR


1.9


P54G
117
G
A
C-7
G
G
1.2



139
A
G
C-15
N
D
5.0



155
G
A
C-20
G
E
7.3



225
A
G
C-43
P
P
1.1



320
A
G
C-75
Q
R
1.7



330
G
A
C-78
M
I
1.4



488
G
A
prM-8
*G
*E
2.0



489
G
A
prM-8


1.3



507
A
G
prM-14
V
V
1.1



681
A
G
prM-72
S
S
1.9



728
G
A
prM-88
R
K
3.6



767
A
G
prM-101
E
G
1.2



831
A
G
prM-122
V
V
2.1



920
G
A
prM-152
R
K
1.5



2346
A
G
E-460
I
M
1.1



2624
A
G
NS1-52
E
G
1.1



2788
A
G
NS1-107
T
A
1.8



3440
A
G
NS1-324
Q
R
6.8



3561
C
U
NS2A-12
G
G
3.0



4113
A
G
NS2A-196
K
K
4.4



4871
A
C
NS3-87
H
P
1.6



4918
G
A
NS3-103
G
S
1.9



5187
G
C
NS3-192
L
L
1.9



5502
A
U
NS3-297
A
A
1.7



5580
A
G
NS3-323
S
S
1.6



6357
C
U
NS3-582
V
V
1.2



6717
A
G
NS4A-83
G
G
4.4



8179
A
G
NS5-167
A
G
1.1



9681
A
G
NS5-667
G
G
1.2



10888
U
A
3′ UTR


1.2





All SNVs ≥1% frequency in the P1 cell culture stock of each virus are listed.


*Two nucleotide changes occurred simultaneously, viewed on Tablet software













TABLE 4







WNV NS4B-P54G mutant harbored 18 single nucleotide variants


≥1% frequency after five Vero cell passages.













Nucleo-
Major
Minor
Viral





tide
Nucleo-
Nucleo-
Protein
Major
Minor
Frequency


Position
tide
tide
Position
Residue
Residue
(%)
















139
A
G
C-15
N
D
3.3


155
G
A
C-20
G
E
2.4


728
G
A
prM-88
R
K
2.4


2743
A
G
NS1-92
K
E
1.6


3440
A
G
NS1-324
Q
R
46.8


3561
C
U
NS2A-12
G
G
2.3


4113
A
G
NS2A-196
K
K
2.6


4918
G
A
NS3-103
G
S
19.1


5166
G
A
NS3-185
R
R
1.0


5187
G
C
NS3-192
L
L
18.7


6717
A
G
NS4A-83
G
G
1.7


7081
C
A
NS4B-56
L
I
16.5


7091
U
G
NS4B-59
L
W
43.0


7123
A
G
NS4B-70
T
A
2.1


7183
G
A
NS4B-90
D
N
2.1


7193
U
C
NS4B-93
V
A
1.2


7274
G
U
NS4B-120
C
F
1.6


10888
U
A
3′ UTR


1.2









The frequency of reversion to the wild-type proline residue of NS4B-P54 mutants were measured in Vero cell P0, P1, and P5 virus stocks. The limit of detection was 0.1%.


Even though the P54A mutation did not rapidly revert in mouse experiments, the SNVs identified in the P1 cell culture stock exhibited instability of the genotype with many high frequency SNVs, especially in the NS4B gene. One of these, NS4B-D90G, was selected for during cell culture passage and encoded a consensus sequence change by P5. NS4B-90 is on the ER lumen side of the protein, and while the role of this residue during WNV infection is not known, it is possible that the loss of negative charge at this position helped to compensate for changes to NS4B induced by the P54A mutation. However, NS4B-D90G mutation was not evident in the quasispecies of the P54G mutant through P5.


Example 7— Immunostaining of the Attenuated WNV NS4B-P54G Mutant

Since the P54G mutant was strongly attenuated in mice and had a stable genotype after cell culture passage, confocal microscopy was used to further investigate the mechanism of attenuation for this single site mutant. Virus-infected Vero cells were stained for both NS4B and NS1 since it is known that these two viral proteins interact [11]. Furthermore, an attenuated NS1 glycosylation site mutant (hereafter termed NS1mut) that was used for NS1 immunostaining in previous studies [34] was used as an attenuated control virus. One day post infection, the P54G mutant had fewer infected cells than NY99ic (FIG. 8A), and this was expected based on the differences in infectivity titers (FIG. 3A). Cells infected with the P54G mutant, however, had significantly lower levels of NS4B (p=0.0002), but had similar staining of NS1 compared to NY99ic (FIG. 8B). Consistent with previous studies, the attenuated WNV NS1mut had lower levels of NS1 (p=0.006), but the staining of NS4B was similar to that in NY99ic infected cells (FIG. 8B). Pearson's correlation coefficient (PCC) was also calculated to compare colocalization of NS1 and NS4B in infected cells. For PCC, a value of 0 indicates no colocalization, whereas a value of 1 indicates perfect colocalization. At one dpi, all infected cells exhibited some degree of colocalization of NS1-NS4B, but both the NS4B-P54G mutant and the NS1mut had significantly less colocalization compared to NY99ic (p=0.004 and p=0.01, respectively) (FIG. 8C).


At two dpi, the P54G mutant still exhibited lower levels of NS4B compared to NY99ic (p=0.03), however, there were also significantly lower levels of NS1 staining (p=0.03) (FIG. 9A and FIG. 9B). In comparison, the NS1mut attenuated control had similar levels of NS4B as NY99ic and there was stronger NS1 staining compared to NY99ic (p=0.02) (FIG. 9A and FIG. 9B), which was in agreement with previous studies demonstrating that this mutant had a block of NS1 transport out of the endoplasmic reticulum [34]. At 48 hpi, NY99ic and the NS1mut had strong colocalization of NS1-NS4B, but the P54G mutant still exhibited a significant reduction in colocalization (p=0.009) (FIG. 9C).


Example 8— Genomic Diversity of a YFV Asibi NS4B-P52G Mutant

Since the WNV NS4B-P54 residue is highly conserved in the Flavivirus genus, the inventors hypothesized that the homologous mutation will also attenuate other pathogenic Flaviviruses. Therefore, the inventors investigated whether or not a YFV NS4B-P52G mutant (the equivalent residue to WNV NS4B-P54G) would demonstrate an attenuated genotype. A YFV NS4B-P52G mutant was generated based on site-directed mutagenesis of an infectious clone of the wild-type (WT) Asibi strain. The mutant genome was sequenced and had the correct mutation and no compensatory mutations. Genetic diversity (Shannon entropy and SNV frequency) of the YFV Asibi NS4B-P52G mutant was compared to the parent WT Asibi strain and an infectious clone (ic) of the live attenuated 17D vaccine strain, derived from WT strain Asibi. Previous studies demonstrated that YFV 17D had low genetic diversity compared to the virulent WT Asibi strain, which had higher levels of diversity more typical of an RNA virus [29,37]. Furthermore, the homogeneity of the 17D population is thought to contribute to viral attenuation by reducing the likelihood of reversion to virulence [38]. The coverage of the Asibi ic and YFV 17Dic sequences were down-sampled to resemble the depth of coverage of the NS4B-P52G mutant. The down-sampled coverage of sequencing reads for the Asibi ic, 17Dic, and NS4B-P52G mutant was 1426, 1436, and 1433, respectively. As expected, the Asibi ic exhibited multiple peaks of high entropy across the genome and 12 SNVs >1% frequency were detected (FIG. 10). In contrast, the 17Dic had fewer peaks of high entropy and had no detectable SNVs >1% frequency (FIG. 10). For both measurements of diversity, the YFV NS4B-P52G mutant more closely resembled the 17D ic. Specifically, the Shannon entropy of the mutant had few peaks of high entropy and had only two detectable SNVs >1% frequency (FIG. 10), supporting that the NS4B-P52G mutation induced changes to the YFV quasispecies that are characteristic of the attenuated YFV 17D vaccine strain [29,37,38].


Interestingly, for WNV the NS4B-P54G mutation generated an attenuated phenotype without reduction of genetic diversity compared to WT WNV, whereas, for YFV both the NS4B-P52G and 17D attenuated mutants were associated with loss of genetic diversity suggesting that attenuation of a neurotropic (brain) flavivirus is different than that of a viscerotropic (liver) flavivirus.


Example 9— Exemplary Methods

Cell Culture. Vero African Green Monkey kidney cells and A549 human alveolar epithelial cells were grown at 37° C. with 5% CO2 in minimum essential media (MEM-Gibco) supplemented with 100 U/mL penicillin, 100 ug/mL streptomycin, 2 mM L-glutamine, 0.1 mM non-essential amino acids, and 8% fetal bovine serum (FBS).


Reverse Genetics. An infectious clone based on WNV strain NY99-flamingo382 was utilized as previously described [28]. Asibi and 17D YFV infectious clones were prepared as previously described [29]. Mutagenesis primers were designed for mutation of WNV NS4B residues P54A, P54G, as well as YFV NS4B P52G. Primer sequences are listed in Table 5.









TABLE 5







NS4B mutagenesis primers











NS4B





Mutation
Primer 1
Primer 2






WNV-P54A
acaacagcgg
gatcaaatgc



(SEQ ID
tcctcactgc
tttagcaggg



NO: 9
cctgctaaag
cagtgaggac



and 10)
catttgatc
cgctgttgt






WNV-P54G
Tgctttagca
tgtgacaaca



(SEQ ID
gtccagtgag
gcggtcctca



NO: 11
gaccgctgtt
ctggactgct



and 12)
gtcaca
aaagca






YFV-P52G
gatccagtgg
cattgttaca



(SEQ ID
tgcaacattc
atgctctctg



NO: 17
cagagagcat
gaatgttgca



and
tgtaacaatg
ccactggatc



18)









Mutagenesis was completed using the Agilent QuikChange II XL Site-Directed Mutagenesis kit (Cat. No. 200521) according to the manufacturer's protocol. After mutagenesis, plasmids were transformed into MC1061 competent E. Coli cells and grown in 200 mL Luria broth (LB) with 100 ug/mL ampicillin. After growth for 14-16 hours, bacteria were pelleted and suspended in glucose-tris-EDTA buffer. Cells were lysed using 0.2 M NaOH/1% SDS, and lysis was neutralized using 3 M KOAc. After isopropanol precipitation, the plasmid was treated with RNAse A for 60 minutes, then purified using phenol:chloroform:isoamyl alcohol. The purified plasmid was ethanol precipitated, then desalted and concentrated using the QiaQuick PCR purification kit (Qiagen). Preparation, purification, and in vitro transcription were carried out for each clone as previously described [28]. Viruses were rescued in Vero cells between 4-6 days post transfection. WNV clones were passaged once in Vero cells to generate the stocks utilized for the experiments described below and P0 unpassaged YFV clones were utilized for genomic analysis described below.


Infectivity Titration. Infectivity titers were measured using plaque titration in 6-well cell culture dishes that were 90% confluent with Vero cells. Cells were washed with PBS and then ten-fold serial dilutions (10−1-10′) of each virus were incubated on the cell monolayers for 30 minutes with rocking every 5 minutes. Following incubation, cells were overlaid with 4 mL of media containing 1% agarose and 1% MEM supplemented with 2% FBS, plates were incubated at 37° C. for two days (or at 41° C. for temperature sensitivity assays), and then and additional 2 mL of overlay containing 2% neutral red (Sigma) was added to each well. Plates were monitored for two days as plaques formed, and infectivity titers were calculated based upon the reciprocal dilution of plaque-containing wells.


Multiplication kinetics. Multiplication kinetics were undertaken using Vero and A549 cells at a multiplicity of infection (MOI) of 0.1. Infections were completed in duplicate flasks and virus was allowed to adsorb for 30 minutes at room temperature. Flasks were washed once with PBS, and then supplemented with MEM maintenance media containing 2% FBS. Cells were incubated at 37° C. for 4 days, and at 0, 24, 36, 48, 72, and either 84/96 hours post infection, two aliquots were collected from each flask for titration. Aliquots were centrifuged at 1,500 RPM 5 minutes to pellet cell debris, and then supernatants were stored at −80° C. until plaque titration.


Mouse virulence. Groups of outbred 4-week-old Swiss Webster mice (Taconic Farms, Germantown, N.Y.) were used to investigate virus neuroinvasion. All viruses were inoculated via the intraperitoneal route (i.p.) into groups of five mice using an inoculum of 500 PFU, and mice were monitored for 36 days for signs of neurological disease. At 36 days post infection (dpi) surviving mice were challenged with an i.p. inoculum of 104 PFU of NY99ic (≥1000 LD50) and then monitored for 28 days post challenge. For viruses that were attenuated with the 500 PFU dose, studies were repeated using a high inoculum ranging from 520,000-39,000,000 PFU (dependent upon the infectivity titer).


Cytokine quantification. Cytokines were measured in cell culture supernatant of A549 cells at 36 hpi following infection of the cells with a MOI of 0.1. Supernatants were collected from cells infected with each of the NS4B mutants as well as NY99ic and mock infected cells. Samples were centrifuged at 1,500 RPM for 5 minutes to remove cell debris, and then stored at −80° C. until use. Supernatants were gamma irradiated to remove infectivity and then processed for multiplex assay according to the manufacturer's protocol using a Bio-Plex Pro Human Cytokine 27-plex Assay (Cat. No. M500KCAFOY) and a Bio-Plex custom IFN-α/IFN-β assay. A Kruskal-Wallis ANOVA with Dunn's correction was utilized to compare each sample to NY99ic-infected cells.


Sequencing Analysis. RNA was extracted from Vero cell culture supernatant using the QiaAmp Viral RNA Kit (Qiagen). Paired-end reads were sequenced on the Illumina NextSeq 550 platform and Trimmomatic [30] was utilized to remove adapters and any sequences with a quality score below 30. The trimmed reads were aligned to the appropriate reference sequence (WNV NY99ic, YFV 17Dic, or YFV Asibi ic) using Bowtie2 with the very sensitive local parameter. All reads were sorted based on genome position and coordinate position using SAMtools, and PCR duplicates were marked and removed using Picard Tools (Broad Institute) with the optical duplicate pixel distance set to 0. Depth of coverage was measured with SAMtools. For the YFV 17Dic and Asibi ic, depth of coverage was randomly down-sampled using Picard Tools. LoFreq was utilized to measure single nucleotide variants in the viral RNA populations [31]. Individual sequencing contigs were visualized in Tablet software (James Hutton Institute) [32]. The diversity indices for the YFV clones were calculated using the R package deepSNV (v. 1.32.0) to calculate nucleotide frequency at each position in the genome and Shannon entropy was calculated as previously described[33].


Fluorescence Microscopy. Vero cells were infected with a MOI of 0.1 of NY99ic, the NS4B-P54G mutant, a WNV NS1 glycosylation site mutant [34], or PBS as a mock infection. After 24 or 48 hpi, cells were seeded onto Teflon-coated microscope slides (Polysciences Cat. No. 18357-1). Cells were placed in the 37° C. incubator for approximately five hours to adhere and then they were fixed with a 1:1 acetone:methanol solution. Slides were stored at −20° C. until immunostaining. Prior to staining, slides were incubated in 5% normal goat serum/3% bovine serum albumin blocking buffer for 1 hour at room temperature (r.t.). Slides were washed twice with tris-buffered saline (TBS) then incubated with primary antibody for two hours at r.t. DENV NS4B monoclonal antibody 44-4-7 (kindly provided by Dr. Pei-Yong Shi) and a WNV NS1 polyclonal antibody (Genetex Cat. No. 132053) were diluted 1:500 to generate working stocks. Slides were washed twice with TBS prior to incubation with secondary antibodies (Invitrogen Cat. No. A-11001 and A-11011) for 1 hour at r.t. After washing three times with TBS, slides were incubated with 7 μg/mL DAPI for 5 minutes, and then washed again four times with TBS. Slides were mounted with Vectashield (Vector Laboratories Cat. No. H-1000) and stored at 4° C. overnight. Slides were imaged with a Zeiss LSM 880 confocal microscope using a 1.4 numerical aperture 63× oil immersion lens. Using ImageJ software, background fluorescence was subtracted uniformly from each image and mean fluorescence intensities and Pearson's correlation coefficients were calculated by drawing regions of interest around infected cells.


Statistical Analysis. ANOVA, Kruskal-Wallis tests, and calculations of standard deviation were all completed in GraphPad Prism v. 8.0.


Example 10— Yellow Fever Virus and Dengue Virus-4

Yellow fever virus (YFV) Asibi NS4B-P52G and Dengue virus-4 (DENV-4) NS4B-P47G Mutants. Since the WNV NS4B-P54 residue is highly conserved in the Flavivirus genus, the inventors contemplate that the homologous mutation will also attenuate other pathogenic flaviviruses. Demonstration of proof of principle in other flaviviruses include YFV as it genetically very different to WNV causing viscerotropic rather than neurotropic disease, and DENV-4 because it causes a febrile illness and is genetically different to both YFV and WNV.


It is shown that YFV NS4B-P52G mutant (the equivalent residue to WNV NS4B-P54G) was generated by site-directed mutagenesis of the WT Asibi strain (Asibi ic). Next Generation Sequencing (NGS) showed the mutant genome had the correct mutation and no compensatory mutations. Genetic diversity (Shannon entropy and SNV frequency) of Asibi NS4B-P52G was compared to the parent WT Asibi ic and to an ic of the 17D live attenuated vaccine strain (17D ic). YFV 17D has low genetic diversity compared to Asibi. Furthermore, the homogeneity of the 17D viral RNA population is thought to contribute to viral attenuation by reducing the likelihood of reversion to virulence. The coverage of the Asibi ic and 17Dic sequences were down-sampled to resemble the depth of coverage of the NS4B-P52G mutant. The down-sampled coverage of sequencing reads for the Asibi ic, 17Dic, and NS4B-P52G were 1426, 1436, and 1433, respectively. The Asibi ic exhibited multiple peaks of high entropy across the genome and 12 single nucleotide variants (SNVs)>1% frequency were detected. In contrast, 17Dic had fewer peaks of high entropy and had no SNVs >1% frequency. For both measurements of diversity, NS4B-P52G more closely resembled 17D ic. Specifically, the Shannon entropy of the mutant had few peaks of high entropy and had only 2 detectable SNVs >1% frequency, supporting that the NS4B-P52G mutation induced changes in genetic diversity that are characteristic of the 17D LAV, possibly via affecting the function of the RC.


YFV NS4B-P52G does not form plaques but does give cytopathic effect when used to infect monkey kidney Vero cells so it is possible to titrate the virus by an assay called 50% Tissue Culture infectious dose (TCID50).


A DENV-4 NS4B-P47G mutant has been generated (the equivalent residue to WNV NS4B-P54G) by site-directed mutagenesis of an infectious clone of the wild-type DENV-4 strain 1036. The virus gave cytopathic effect post transfection.


REFERENCES



  • 1. Chancey et al., Biomed Res. Int. 2015, 1-20.

  • 2. Mann et al., Int. J. Environ. Res. Public Health 2013, 10, 5111-5129.

  • 3. CDC West Nile Virus-Symptoms, Diagnosis, & Treatment. Available online: URL www.cdc.gov/westnile/symptoms/index.html (accessed on Jul. 5, 2019).

  • 4. CDC West Nile Virus-Statistics and Maps Available online: URL www.cdc.gov/westnile/statsmaps/index.html (accessed on Jul. 5, 2019).

  • 5. Klema et al., Viruses 2015, 7, 4640-4656.

  • 6. Brinton, Viruses 2013, 6, 13-53.

  • 7. Zmurko et al., Rev. Med. Virol. 2015, 25, 205-223.

  • 8. Li et al., BBA—Biomembr. 2015, 1848, 3150-3157.

  • 9. Li et al., Angew. Chemie—Int. Ed. 2016, 55, 12068-12072.

  • 10. Zou et al., J. Virol. 2014, 88, 3379-91.

  • 11. Youn et al., J. Virol. 2012, 86, 7360-71.

  • 12. Yu et al., Virology 2013, 446, 365-377.

  • 13. Zou et al., J. Virology 2015, 89, 3471-3483.

  • 14. Zou et al., J. Virol. 2015, 89, 3455-3470.

  • 15. Kaufusi et al., PLoS One 2014, 9, e84040.

  • 16. Munoz-Jordan et al., J. Virol. 2005, 79, 8004-8013.

  • 17. Wang et al., J. Virol. 2005, 79, 1934-1942.

  • 18. Kakumani et al., J. Virol. 2013, 87, 8870-8883.

  • 19. Grant et al., J. Virol. 2011, 85, 7775-7787.

  • 20. Pletnev et al., Proc. Natl. Acad. Sci. 2002, 99, 3036-3041.

  • 21. Orozco et al., J. Gen. Virol. 2012, 93, 2152-2157.

  • 22. Dunster et al., Virology 1999, 318, 309-318.

  • 23. Hahn et al., Proc. Natl. Acad. Sci. U.S.A. 1987, 84, 2019-23.

  • 24. Gromowski et al., J. Virol. 2015, 89, 6328-6337.

  • 25. Wicker et al., Virology 2006, 349, 245-253.

  • 26. Wicker et al., Virology 2012, 426, 22-33.

  • 27. Davis et al., Virology 2004, 330, 342-350.

  • 28. Beasley et al., J. Virol. 2005, 79, 8339-8347.

  • 29. Collins et al., MBio 2018, 9, 1-13.

  • 30. Bolger et al., Bioinformatics 2014, 30, 2114-2120.

  • 31. Wilm et al., Nucleic Acids Res. 2012, 40, 11189-11201.

  • 32. Milne et al., Brief Bioinform. 2012, 14, 193-202.

  • 33. Nishijima et al., PLoS One 2012, 7, 1-10.

  • 34. Whiteman et al., J. Virol. 2015, 89, 1474-1478.

  • 35. Welte et al., Vaccine 2011, 29, 4853-4861.

  • 36. Xie et al., Vaccine 2015, 33,869-878.

  • 37. Beck et al., J. Infect. Dis. 2014, 209, 334-344.

  • 38. Davis et al., MBio 2019, 10, 1-14.

  • 39. Kim et al., Protein Sci. 1999, 8, 1492-1499.

  • 40. Kumeta et al., J. Cell Sci. 2017, 131.

  • 41. Xie et al., Virology 2014, 450-451, 250-257.


Claims
  • 1. An attenuated flavivirus genome comprising a NS4B gene segment encoding a mutant NS4B protein having an amino acid substitution corresponding to an amino acid substitution at proline 54 of the West Nile Virus (WNV) NS4B protein of, the amino acid sequence of the WNV NS4B protein corresponding to the amino acids of SEQ ID NO:19.
  • 2. The attenuated flavivirus genome of claim 1, wherein the substitution at proline 54 of the NS4B protein is a nonpolar amino acid.
  • 3. The attenuated flavivirus genome of claim 2, wherein the nonpolar amino acid is an alanine residue (P54A) or glycine residue (P54G).
  • 4. The attenuated flavivirus genome of claim 1, further comprising one or more mutations in the gene segment encoding non-structural protein 1 (NS1), corresponding to amino acids 792 to 1143 of SEQ ID NO:2, the one or more mutations corresponding to the disruption of one or more glycosylation sites of the NS1 protein.
  • 5. The attenuated flavivirus genome of claim 4, wherein the one or more mutations corresponding to one or more glycosylation sites of the NS1 protein is a substitution of one or more of wild-type amino acids asparagine 130, asparagine 131, or threoninine 132 of NS1.
  • 6. The attenuated flavivirus genome of claim 5, wherein the substitution of wild-type amino acids asparagine 130, asparagine 131, or asparagine 130 and asparagine 131 is a substitution of an asparagine residue with a polar amino acid.
  • 7. The attenuated flavivirus genome of claim 6, wherein the polar amino acid is a glutamine residue.
  • 8. The attenuated flavivirus genome of claim 5, wherein the substitution of amino acid threonine 132 is a substitution of the threonine residue with a nonpolar amino acid.
  • 9. The attenuated flavivirus genome of claim 8, wherein the nonpolar amino acid is an alanine residue.
  • 10. The attenuated flavivirus genome of claim 5, wherein the one or more mutations corresponding to one or more glycosylation sites of NS1 of the flavivirus further comprise a substitution of wild-type amino acid 175 of NS1, a substitution of wild-type amino acid 207 of NS1, or a substitution of wild-type amino acid 175 and amino acid 207 of NS1.
  • 11. (canceled)
  • 12. The attenuated flavivirus genome of claim 10, wherein the substitution of wild-type amino acid asparagine 175 wild-type amino acid asparagine 207, or wild-type amino acid asparagine 175 and wild-type amino acid asparagine 207 is a substitution of an asparagine residue with a nonpolar amino acid.
  • 13. The attenuated flavivirus genome of claim 12, wherein the nonpolar amino acid is an alanine residue.
  • 14.-18. (canceled)
  • 19. A nucleic acid construct encoding the genome of the attenuated flavivirus of claim 1.
  • 20. The nucleic acid construct of claim 19, wherein the flavivirus is selected from the group consisting of West Nile virus, Japanese encephalitis virus, St. Louis encephalitis virus, tickborne encephalitis virus, Zika virus, dengue fever virus, and YFV.
  • 21. The nucleic acid construct of claim 19, wherein the flavivirus is yellow fever virus (YFV).
  • 22.-23. (canceled)
  • 24. The nucleic acid construct of claim 19, wherein the nucleic acid encoding the genome of the attenuated flavivirus further comprises one or more mutations corresponding to one or more glycosylation sites of non-structural protein 1 (NS1) of the flavivirus.
  • 25.-29. (canceled)
  • 30. An immunogenic composition comprising the attenuated flavivirus encoded by claim 1 and a pharmaceutically acceptable carrier or diluent.
  • 31. (canceled)
  • 32. A method of inducing an immune response in a subject comprising administering an effective amount of the composition of claim 30 to the subject.
  • 33.-38. (canceled)
  • 39. The method of claim 32, wherein the subject is a non-human primate, a human, a horse, or a bird.
  • 40.-42. (canceled)
  • 43. A vaccine composition comprising the attenuated flavivirus genome of claim 1 or an attenuated flavivirus encoded by the attenuated genome of claim 1 and a pharmaceutically acceptable carrier or diluent.
  • 44.-75. (canceled)
RELATED APPLICATIONS

This application is a U.S. Utility application claiming priority to U.S. Provisional Application 63/186,160 filed May 9, 2021, which is incorporated herein by reference in its entirety.

STATEMENT REGARDING FEDERALLY FUNDED RESEARCH

This invention was made with government support under T32 A1007526, R01 A1099123, and AI127744 awarded by the National Institutes of Health. The government has certain rights in the invention.

Provisional Applications (1)
Number Date Country
63186160 May 2021 US