AU2013202434B2 - Dengue virus neutralizing antibodies and uses thereof - Google Patents

Dengue virus neutralizing antibodies and uses thereof Download PDF

Info

Publication number
AU2013202434B2
AU2013202434B2 AU2013202434A AU2013202434A AU2013202434B2 AU 2013202434 B2 AU2013202434 B2 AU 2013202434B2 AU 2013202434 A AU2013202434 A AU 2013202434A AU 2013202434 A AU2013202434 A AU 2013202434A AU 2013202434 B2 AU2013202434 B2 AU 2013202434B2
Authority
AU
Australia
Prior art keywords
seq
antibody
antibodies
variable region
acid sequence
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
AU2013202434A
Other versions
AU2013202434A1 (en
Inventor
Antonio Lanzavecchia
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Institute for Research in Biomedicine IRB
Original Assignee
Inst Res Biomedicine
Institute for Research in Biomedicine IRB
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from AU2009305113A external-priority patent/AU2009305113B2/en
Application filed by Inst Res Biomedicine, Institute for Research in Biomedicine IRB filed Critical Inst Res Biomedicine
Priority to AU2013202434A priority Critical patent/AU2013202434B2/en
Publication of AU2013202434A1 publication Critical patent/AU2013202434A1/en
Application granted granted Critical
Publication of AU2013202434B2 publication Critical patent/AU2013202434B2/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Abstract

The invention relates to antibodies and antigen binding fragments thereof and to cocktails of antibodies and antigen binding fragments that neutralize dengue virus infection without contributing to antibody-dependent enhancement of dengue virus infection. The invention also relates to inmortalized B cells that produce, and to epitopes that bind to, such antibodies and antigen binding fragments, In addition, the invention relates to the use of the antibodies, antigen binding fragments, and epitopes in screening methods as well as in the diagnosis and therapy of dengue virus infection.

Description

AUSTRALIA PATENTS ACT 1990 REGULATION 3,2 Name of Applicant- INSTITUTE FOR RESEARCH IN BIOMEDICINE Actual Inventor/s: Antonio Lanzavecchia Address for Service: E. K WELLINGTON & CO., Patent and Trade Mark Attomeys, 312 St. Kilda Road, Melboume, Southbank, Victoria, 3006. Invention Tide: "DENGUE VWRUS NEUTRALIZING ANTIBODIES AND USES THEREOF" Details of Associated Provisional Applications Nos: The following statement is a full description of this inwention including the best method of performing it known to us, This application is a 'divisional' application derived from Australian Patent Application No. 2009305113 (PCT/1B2009/007372: WO 2010/043977), claiming priority of US Application No. 61/104911, die entire text of which are hereby incorporated herein by reference. 5 BACKGROUND Dengue viruses (DENV) are human pathogens with a significant threat to world health. These viruses are estimated to cause several hundred thousand cases of dengue fever, dengue hemorhagic fever and dengue shock syndrome annualy, There are four closely related serotypes of dengue viruses, DEN- 1, DENV-2, DENV-3 and DENV-4, of the genus Fkaivirus. 10 The four viruses are spread from human to human through the bite of Aedes aegyp, a highly urbanized mosquito species that has successfully resisted all attempts at eradication and control Vaccination is considered to be the only efficient method of control of dengue. To this end, several tetravalent dengue candidate vaccines are in late stages of development A first infection with one lngue virus serotype induces a life-long protective immunity 15 to the homologous serotype Howev there o Cro-ro oagi different serotype Indeed, pm-existing immunity against one serotype is associated with increased risk for dengue infection and dengue hemorrhagic fever caused by a different serotype due to antibody-dependent enhancement (ADE) of infection, in AE, antibodies raised by prior dengue infection or passively transferred from mother form infectious immune complexes that 20 attach to Fc-receptor-beaing cells in the mononuclear phagocyelineage resuming in efficient infection. Accordingly, there is a need for materials and methods for preventing dengue virus infection without increasing the risk of antibody-dependent enhancement of infecuon. SUMMARY 25 The invention is based, in pat, on the discovery of antibodies and cocktails of antibodies that neutralize dengue virus infectionwhou contributing to antibody-dependent enhancement of dengue virus infection. Accordinglyin one aspect of the invention, the invention comprises a human antibody, an antibody variant, or an antigen binding fragment thereof, that neutralize a dengue virus, vherein the antibodyabody varianor antigen binding fragment does not 30 contribute to antibody -dependent enhancement of engue virus infecton Uone embodiment
-IA-
the invention comprises a human antibody, an antibody variant, or an antigen binding fragment thereof, that neutralize a dengue virus, wherein the antibody, antibody variant, or antigen binding fragment comprises a mutation in the Fe region, and wherein the mutation reduces binding of the antibody to an Fe rceptor 5 In another embodiment of the invention, the invention comprises a pharmaceutical composition comprising two or more human antibodies, or antigen binding fragments thereof The antibodies or antigen binding fragments neutralize dengue virus semutypes DENN, DENY 2, DENV-3, and DENV-4 by binding at least two distinct epitopes on each dengue virus sexutypei The antibodies of the phannaceutical composition do not contribute to antibody 10 dependent enhancement of dengue virus infection. In yet another embodiment, the invention comprises an antibody' or an antigen binding fragment thereof, comprising at least one complementarity determining region (CDR) sequence having the sequence of any one of SEQ lW NOs: 16, 7-22, 33-3s,49-54, 67-72, 83-88, 99, 100, 105110, 121-123, 124, 125, 135-139, 149. 153-158, 169-74 8-188, or 189, wherein the 15 antibody neutralizes dengue virus infection, In yet another embodiment, the invention comprises an antibody, or an antigen binding fragment thereof, wherein the antibody comprises a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 13 and a light chain variable region comprising the amino acid sequence of SEQ ID NO: 14; or a heavy chain variable region comprising the amino acid 20 sequence of SEQ ID NO: 29 and a light chain variable region comprising the amino acid sequence of SEQ ID NO: 30; or a heavy chain Tvariable region comprising the anino acid sequence of SEQ. ID NO: 45 and a light chain variable region comparing the amino acid sequence of SEQ ID NO: 46; or a heavy chan vanable region comprising the amino acid sequence of SEQ ID N): 61 and a light chain variable region comprising the amino acid 25 sequence of SEQ ID NC: 62; or a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 65 and a light chain variable region comprising the amino acid sequence of SEQ ID NO: 62; or a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 79 and a light chain variable region comprising the amino acid sequence of SEQ ID NO: 80; or a heavy chain variable region comprising the amino acid 30 sequence of SEQ ID NO: 95 and a light chain variable region comprising the amino acid sequence of SEQ ID NO 96; or a heavy chain variable region comprising the amino acid sequence of SEQ ID NCK)95 and a light chain variableregion comparing the amino acid sequence of SEQ ) NO: 103; or a heavy chain variable region comprising the amino acid 2 sequence of SEQ 1D NO 117 and a light chain variable region comprising the amino acid sequence of SEQ ID NO: 118; or a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: I 31 and a light chain variable region conprising the amino acid sequence of SEQ ID NO 132; or a heavy chain variable region comprising the amino acid 5 sequence of SEQ D NO: 145 and a light chain variable region comprising the amino acid sequence of SEQ ID NO: 146; or a heavy chain variable region comprising the amino acid sequence of SEQ 1f) NO: 151 and a light chain variable region comprising the amino acid sequence of SEQ ID NO; 146; or a heavy chain variable region comprising the amino acid sequence of SEQ ID NO; 165 and a light chain variable region comprising the amino acid 10 sequence of SEQ ID NO: 166; or a heavy chain variable region comprising the amino acid sequence of SEQ ID NO; 181 and a light chain variable region comprising the amino acid sequence of SEQ .19 NO: 182; or a heavy chain variable region comprising the amino acid sequence of SEQ I) NO 195 and a light chain variable region comprising the amino acid sequence of SEQ WD NO: 196, Whereia the antibody neutralizes engue virus infection 15 In a further embodiment, the invention comprises a recombinant anybody, antibody variant, or antigen binding fragment thereof that can neutralize a dengue viruS. The recombinant antibody, antibody variant, or antigen binding fragment does not contribute to antibody-dependent enhancenient of dengue virus infection. In another aspect, the invention comprises a nucleic acid molecule comprising a 20 polynueleotide encoding an antibody or antibody fragment of the invention that neutralizes dengue vius infection In yet another aspect, the invention comprises a cell.epressing an antibody of the invention n still another aspect, the invention comprises an isolated or purified immunogenic polypeptide comprising an epitope that binds to an antibody of the invention. 'The invention also comupries a pharm-ace-utical cmoioncomprising an antibody, an 25 antibody variant or an antigen binding fragment of the invention, a nucleic acid of the invention, or an immunogenic polypeptide of the invention and a pharmaceutically acceptable diluent or carrier and, optionally, an agent useful for extending the half life of the anybody or antigen binding fragment thereof. In another aspect of the invention, the invention provides a method of inhibiting or 30 preventing dengue virus infection or a dengue virus-related disease or a method of treating dengue virus infection or a dengue virus-related disease, The method comprises administering to a subject in need thereofa therapeutically effective amount of at least one antibody, anybody variant, antigen binding fragment, or a pharmaceutical composition of the invention. 3 In yet another aspect of the invention, the invention comprises a method of screening for polypeptides that can induce or reveal an inunune response against dengue virus, comprising screening polypeptide libraries using an antibody, an antibody fragment or variant of the invention. 5 In yet another aspect of the invention, the invention comprises a method of monitoring the quality of anti-dengue virus vaccines. The method comprises using an antibody, an antibody vaiant, or an antigen binding fragment thereof of the invention to check that the antigen of the vaccine contains the specific epitope in the correct conformation In a further aspect of the invention, the invention comprises a vaccine comprising an 10 epitope which specifically binds to an antibody, an antibody fragment or variant of the invention. Use of an antibody oftnhe invention, or an antigen binding fragment thereof, a nucleic acid of the invention, an imunuogenic polypeptide of the invention, or a pharmaceutical composition of the invention (i) in the manufacture of a medicament for the treatment of dengue virus infection. (ii) in a vaccine, or (iii) in diagnosis of dengue virus infection is also 15 contemplated to be within the scope of the invention. Further, use of an antibody of the invention, or an antigen binding fragment thereof, for monitoring the quality of antiAENV vaccines by checking that the antigen of said vaccine contains the specific epitope in the correct confonnation is also contemplated to be within the scope of the invention, In a further aspect, the invention comprises an epitope which specifically binds to an 20 antibody of any one of the invention, or an antigen binding fragment thereof, for use (i) in therapy, (ii) in the manufacture of a edicament fir treating dengue virus infection, (iii) as a vaccine, or (iv) in screening for ligands able to neutralise dengue virus infection. BRIEF DESCRIPTION OF FIGURES Figure 1. VERO cells and ,K562 cells were used in virus neutralization and enhancement 25 assays with each serotype of dengue virus using wild-type anti dengue virus antibodies Antibodies to dengue virus inhibi infecion of the target virus on VERO cells in a dose dependent manner. On 1K562 cells the antibodies lead to a dose dependent antibodydependent enhancement (ADE) of infection. Figure 2, Anti-dengue virus antibodies that have a CH2 L..4A and L5A substitution 30 (LALA variants) in the heavy chain neutralize target virus infection on VERO cells as did the unadified antibodies, However, the LALA variants completely abolished the antibody dependent enhancement of infection by the target virus on K562 cells. 4 DETAILED DESCRIPTION OF THE INVENTION The invention is based on the discovery of antibodies and cocktails of antibodies that neutralize dengue virus (DENV) infection without contributing to antibody-dependent enhancement (ADE) of dengue virus infection. In one aspect of the invention, the invention 5 comprises a human antibody, a variant antibody, or an antigen binding fragment thereof, that neutralizes a dengue virus without contributing to antibody-dependent enhancement of dengue virus infection. The antibodies or antibody fragmens can neutralize mote than one dengue virus serotype, for example, 2, 3 or all 4 dengue virus serorypes DENV-I, DENV-2, DEN V-3, and DENV-4. 10 The invention also comprises a pharmaceutical composition comprising, for example, an antibody cocktail that comprises two or more human antibodies, antibxy variants or antigen binding fragments thereof. The pharmaceutical compositions of the invention comprising a cocktail of human antibodies, anidbody fragments or variants neutralize all four dengue virs serotypes, i e. DENV-i, DENV-2, DENV-3, and DENV4, In one embodimentthe cocktail of 15 antibodies, antibody fragments or variants neutralize dengue virus by binding at least two distinct epitopes on each dengue virus serotype It is noted that the antibodies variants and fragments of the pharmaceutical composition do not contribute to antibody-dependent enhancement of dengue virus infection. in one embodiment, the cocktail comprises two antibodies, fragments or variants thereof. in another embodiment, the cocktail comprises three antibodies, fragments or variants 20 thereof, In yet another embodiment, the cocktail comprises more than 3 antibodies, e.g., 4, 5, 6k 7 or 8 antitxdies. As used herein, the terms "fragment," "antibody fragment," and "antigen binding pigment" are used interchangeably to refer to any fragment of an antibody of the invention that retains the antigen-binding activity of the antibody. Exemplary antibody fragments include but 25 are not limited to, Fab, Fab, F(ab') 2 , Fv, and scfv fragments. The items "nutation," and substitutionn" are used interchangeably to refer to a change in one or more nucleic acid or amino acid residues As used herein, the terms "variant," and "antibody variant" are used interchangeably to refer to any variant of an antibody of the. invention that retains the antigen-binding activity of the 30 antibodies The term variant includes antibodies that comprise mutations and/or substitttions. Exemplary antibody variants include. but are notlimited to, those that have an I. to A substitudon at position CR2 4. 5. or both. 5 Antibodies of the invention The invention provides antibodies that neutmlize dengue virus, but dnot contribute to ADE of dengue. virus infection A "neutralizing antibody" is one that can neutralire the ability of a pathogen to initiate and/or perpetuate an infection in a host The antibodies of the invention 5 am able to neutralize one or more dengue virus serotypes DENy DENV- DENNV and DENVW4 In one embodiment, the antibody of the invention neutralizes more thai one, e.g 2, 3, or all 4 dengue virs serotypes In another embodiment a pharmaceutica composition comprising two or more antibodies, antibody fragments or variants can neutralize al 4 dengue virus serotypes, In yet another embodiment, the pharmaceutical composition comprising two or 10 more antibodies, antibody fragments or variants neutralizes dengue virus infection by targeting two distinct epitopes on each dengue virus serotype, These antibodies, antigen binding fragment and variants can be used as prophylactic or therapeutic agents upon appropriate formulation, or as a diagnostic tool, as described herein. The. antibodies of the invention may be monoclonal, for example, human monoclonal 15 antibodies, or recombinant antibodies. The invention also provides fragments of the antibodies of the invention, particularly fragments that retain the antigen-binding activity of the antibodies Although the specificationincluding die caitns may in som places, refer explicitly to antibodyfragmentes),variants)andr derivatives)o antibodies, i derstood that the term antibody" or "antiody of the invention" includes all categories of andbodies, namely antibody 20 fragments) variant(s) and derivative(s) of antibodies, Without being bound to any theory, it is believed that anioydpnetenhancement of dengue virus infection is brought about by the binding of the Fe region of the antibody, in particular, the Fe region of the heavy chain of an IgG molecule, to an Fc receptor, eg., an Fey receptor on a host cell The invention, on the other hand, provides antibodies, including igO 25 molecules, that have reduced binding to the Fe receptors (FcR). In one embodiment the antibody of the invention comprises one or more mutations in the Fe region. The mutations may be any mtation that reduces binding of the antibody to an Fc receptor. in one embodiment, the FC region of an antibody of the invention comprises a substitution at positions CRl2 4, 5, or both. In general, the amino acid at positions 4 and 5 of C12 of the wild-type Igl and 1g03 is a 30 leucine ("). In one embodiment, the antibodies of the invention comprise an amino acid at position CH2 4 5, or both, that is not an L In another embodiment, the antibodies of the invention comprise an alanine ("A") at position CH2 4, or 5, or both An antibody comlprising a CH2 L4A and an L5A substitution is referred t herein as a "LALA" variant, 6 Alternatively, the invention provides antibody fragments that do not comprise an Fc region and thus do not bind to an FeR, Exemplary antibody fragments include, but are not limited to Fab, Fab, F(ab'k, Fv and scFv The sequences of the heavy chains and light chains of several exemplary antibodies of the 5 invention, each comprising three CDRs on the heavy chain and three CDRs on the light chain have been determined The position of the CDR amino acids are defined according to the IMC3T numbering system 2 3. The sequences of the CDRs, heavy chains, light chains as well as the sequences of the nucleic acid moeuleis encoding the CDRs, heavy chains, light chains of many exemplary antibodies of the invention are disclosed in the sequence listimg. Table 1 10 provides the SEQ ID NOs for the amino acid sequences of the six CDRs, the variable region of the heavy and light chains, respectively, of exemplary antibodies of the invention Table 2 provides the SEQ 11) NOs for the sequences of the nucleic acid molecules encoding the CDRs, heavy chains and light chains of exemplary antibodies of the invention Table 1. Amino Acid SEQ Is for Antibody CDRs, Heavy and tight Chains Heavy Chain Light Chain Atbd C------- Variable Region Variable Region HM -DV-1 1-6 13 14 HMB-DV2 17-22 29 30 HMBDV-3 33-38_ 45 _ 46 11MB-DV-4 49546, 62 HMB-DV-5 67-2 79 80 HMB-DV-6 83-88 95 96 H1MB-DV-7 83-85, 99, 53, 100 95 103 HMIB-DV-8 105-110 117 118 HMB-DV-9 121-123, 70 124, 125 131 -- 132 -HBT -1 10 135-139, 109 145 146 H1MB-DV-11 149, 136-139, 109 151 146 IMD-DV-12 53-158 165 166 HMB-DV-13 169-174 181 182 HMB-DV-14 185-188, 37, 189 195 196 15 Table 2. Nucleic Acid SEQ IDs fr Anibody CDRs Heavy and Light Chains C 1eavy Chain Light Chain Antibody CU~s Variable Regin Variable Region HMB-DV-1 7-12 15 16 11MB-D V-2 23-28 31 32 MB-DlV-3 39-44 47 48 lIMB-DV~4 55-60 63,66 64 1MB-DV-5 73-78 81 82 HMB-UV-6 89-94 97 98 1MB-D V-7 89-91, 101, 59, 102 97 104 HMT-DV8 11 -116 119 -120 -MB-DV-9 126-128, 76. 129, 130 133 134 HMB-DV10 1143, 115, 144 147 148 HMB-DV-11 150, 141-143, 115, 144 152 148 HMB-DV-12 159-164 167 168 H1MB-DV-13 175-180 ___ 83 '____ 184 HMB-DV-14 190-193, 43, 194 197 198 In one embodiment, the antibodies or antigen-binding fragments of the invention comprise one or more heavy or light chain CDRs of the exempry antibodies of the invention. In an exemplary embodiment, the antihtdies or antigenbinding fragments of the invention neutralize dengue virus infection and comprise at least one CDR sequence having the sequence 5 of anyone of SEQ 11) NOs: 1-6. 17-22,3338, 49-54 67-2, 83-88, 99, 1W, 105410, 121-123, 124, 125, 135-139, 149, 153-158, 169174, 185-188, or 189 In another embodiment, the antilAies, antibody variants or antigen binding fragments of the invention comprise a heavy chain comprising an adno acid sequence of one or more of SEQ ID NOs: 1-3, 1719, 33-35, 49-51, 67-69, 83-85, 105-107, 121-123 135-1.37, 149, 153-155, 169 10 171, or 185-187. In yet another enbodinient the antibodies, antibody variants or antigen binding fragments of the invention comprise a heavy chain CDR 1 selected from the group consisting of SEQID NOs: I, 17,33,49,67,83 105 121 15149, 153, 169 ,and 5; a heavy chain CDR2 selected fronthe group consisting of SEQ ID N0s: 2 1834, 50. 68, 84, 106, 122, 136. 154, 170, and 186 and a heavy chain CDR3 selected from the group consitng of SEQ U) NOs: 3, 15 19,35, 51, 69, 85, 107, 123, 137, 155 171, and 187 For example, the antibodies of the invention comprise a heavy chain comprising SEQ ID NC): 1 for (1, SEQ ID NO 2 for CDR12, SEQ I NO: 3 for CDRH13; SEQ 1D O 17 for CDRH1, SEQ I) NO: 18 for (DRH12 and SEQ I) NO: 19 for CDRH13; SEQ El) NC): 33 for C(DR 11, SEQ ID NO: 34 for CDR2 and SEQ 1D NO: 35 for CDR13; SEQ 11) NO: 49 for 20 CDR111, SEQ ID NO: 50 for CDRH22 and SEQ 11) NO: 31 fr RCDRH3;SEQ 11) NO: 67 for CDRH1, SEQ ID NC) 68 for CDRH-12 and SEQ D NO: 69 for CD2)RH3; SEQ ID NO: 83 for DR11, SEQ ID NO: 84 for CDRH12 and SEQ ID NO; 85 for DR13; SEQ ID NO: 105 for CDRN1, SEQ ID NO: 106 for ("DRH2P and SEQ ID NO: 107 for CDRT H13; SEQ 1D NO: 121 for CDRH1, SEQ fID NO: 122 for CDR12 and SEQ ID NC): 123 for CDRHI-3; SEQ ID NO: 135 for 25 CDR11, SEQ ID NO 136 for CDRH12 and SEQ 1D NO 137for CDRH03 SEQ I) NC: 149 for (DR 1H1, SEQ ID NO: 136 for CDR112 and SEQ) ID NO: 137 for CDR3 SEQ 11) NO 153 for CDRHI1, SEQ ID NO: 154 for CDRH2 and SEQ ID NO: 155 for CDRH-13; SEQ ID NO 169 for CDRH SEQ ID) NO: 170 for CDRH2 and SEQ ID NO: 171 for (DRH1 and SEQ ID NO: 185 for CDR111, SEQ ID NO: 186 for CDR12 and SEQ ID NO I87 for ODH1(13. 8 In yet another embodiment, the antibodies, anybody variants or antibody fragments of the invention comprise a light chain compnsing an amino acid sequence of one or more of SEQ ID NOs: 4-6, 20-22, 36-38, 52-54, 70-72, 86-88, 99, 100, 108-110, 124, 125, 138, 139, 156-158, 172474, 188, or 189, In a further embodiment, the anibodies, antibody variants or anibody 5 fragments of the invention comprise a light chain CUR1 selected from the group consisting of SEQ ID NOs: 4, 20, 36, 52, 70, 86, 99, 108, 138, 156, 172, and 188; a light chain CDR2 selected from the group consisting of SEQ ID NOs: 5,21,37, 53, 71,87, 109, 124, 157, and 173; and a light chain CDR3 selected from the group consisting of SEQ I) Nts: 6, 22, 38,54, 72, 88, 100, 110, 125, 139, 158, 174, and 189, 10 For example, the antbodies of the invention comprise a light chain comprising SEQ ID NO: 4 for CURLI, SEQ I D NO: 5 for CDRL2; SEQ 11 NO 6 for CDRI?; SEQ ID NO: 20 for CDRi, SEQ ID NO: 21 for CDRL2; SEQ ID NO: 22 fir CDRL3; SEQ ID No: 36 for CRU, SEQ ID NO: 37 for CURL2; SEQ ID NO: 38 for CURL3; SEQ ID NO: 52 for CDRIL SEQ 1D NO:53 for CDRL2 SEQ ID NO: 54 for CDRL; SEQ 1 NO: 70 for CURL1 SEQ NOD NC): 71 15 for CDRL2; SEQ ID) NO: 72 for CDRI?; SEQ D NO: 86 for CDR, SEQ ID NO: 87 for CURI2 SEQ ID NO: 88 for GURU; SEQ ID NO:99 for CRL, SEQ ID NO:53 for CDRL2; SEQ ID NO: 100 for CURL?; SEQ ID NO: 108 for CDRIl SEQ ID NO 109 for CDRI,2; SEQ ID NO: 110 for CDRL3 SEQ ID NO: 70 for CUR1 SEQ ID NO: 124 for CDRL2; SEQl ) NO: 125 for CDRL3; SEQ U) NO: 138 for CDRIL, SEQ ID NO: 109 for CURL2; SEQ ID NO 20 139 for CURL?: SEQ ID NO: 156 for CDRL, SEQ ID NO: 157 for CDRL2; SEQ ID NO: 158 for CDRL3; SEQ ID NO: 172 for CDRIA, SEQ fD NO: 173 for CURL2: SEQ ID NO: 174 for CDRL; and SEQ ID NO: 188 for CDRLl, SEQ If) NO 37 for CURE:2; SEQ ID NO: 189 for CDR1. In one embodiment, an antibody of the invention, or antigen binding fragment thereof, 25 comprises all of the CDRs of antibody HMB-DV-I as listed in Table 1, and neutralizres dengue virus infection in a human host. In another embodiment, an antibody of the invention, or antigen binding fragment thereof, comprises all of the CURs of antibody HMI-DV-2 as hsted in Table 1, and neutralizes dengue virus infection in a human host. in another embodiment, an antibody of the invenion, or antigen binding fragment thereof, comprises all of the CDRs of antibody HM 30 DVI as listed in Table 1, and neutralizes dengue virus infection in a human host In yet another embodiment, an antibody of the invention or antigen binding fragment thereof comprises all of the CDRs of antibody HMB-D-4 as listed in Table 1, and neutralizes dengue virus infection in a human host. In yet another embodiment, an antibody of the invention, or antigen binding fragment thereof, compises all of the CURsof antibody 1MB-D-5 as listed in Table 1, and 9 neutralizes dengue virus infection ia human host In yet another embodiment, an antibody of the invention, or antigen binding fragment thereof, comprises all of the CDRs of antibody IUMB DV-6 as listed in Table 1, and neutralizes dengue virus infection in a human host. In yet another embodient, an antibody of the invention, or antigen binding fragment thereof comprises all of 5 the CDRs of antibody lMB-I-7 as listed in Table 1and neutralizes dengue virus infection in a human host. In a further emXliment, an antibody of the invention, or antigen binding fragment thereof, comprises all of the CDRs of antibody HMBDV$ as listed in Table I, and neutralizes dengue virus infection in a human host In another embodiment, an antibody of the invention or 10 antigen binding fragment thereof, comprises all of the CDRs of antibody HM-DV-9 as listed in Table 1, and neutralizes dengue virus infection in a human host In still another embodiment an antibody of the invention, or antigen binding fragment thereof, comprises all of the CDRs of antibody HMB-DV-40 as listed in Table 1, and neutralizes dengue virus infection in a human host, In yet another embodAient, an antibody of the invention, or antigen binding fragment 15 thereof, comprises all of the CDRs of antibody IMB-DV-1 I as listed in Table 1, and neutralizes dengue virus infection in a human host. In yet another embodiment, an antibody of the invention, or antigen binding fragment thereof, comprises all of the CDRs of antibody HMB-DV 12 as listed in Table I, and neutralizes dengue virus infection in a human host; In yet another embodiment, an antibody of the invention, or antigen binding fragment thereof, comprises all of 20 the CURs of antibody IM-DV -13 as listed in Table 1, and neutralizes dengue virus infection in a human host In yet another embodiitent, an anybody of the invention, or antigen binding fragment thereof, comprises all of the CDR-;of antibody HMB-DlV44 as listed in Table 1,and neutralizes dengue virus infection in a hann host. In still another embodiment, the antibodies of the invention comprise a heavy chain with 25 anramino acid sequence that is at least 70%, at least 75%, atleast 80% at least 85%, at least 90%, at least95%, at least 98%, or at least 99% identical to those of SEQ ID NOs; 13 29, 45, 61. 65, 79, 95, 117, 131. 145, 151, 165, 181, or 195 In yet another embodiment, the antibodies of the invention comprise a light chain with an amino acid sequence that is at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, or at least 99% identical 30 to those of SEQ f) NOs: 14, 30, 46, 62, 80, 96, 103, 118, 132, 146, 166, 182, or 196. In a ftirthefr etbodiment, the antibodies, antibody variants or antibody fragments of the inventioncomprise a heavy chain variable region comprising the amino acid sequence of any one of SEQ ID NOs: 13 29. 45, 61, 65,79,95. 117, 131, 145, 15, 16,5 181, or 195, and a light chain 10 variable region comprising the amino acid sequence of any one of SEQ I) NOs: 14, 30 46, 62, , 969 103, 118, 132, 146, 166, 182, or 196 In yet another embodiment, the antibodies, antibody variants or antibody fragments of the invention neutralize dengue virus infection and comprise a heavy chain variable region comprising 5 the amino acid sequence of SEQ 1D NO' 13 and a light chain variable region comprising the amino acid sequence of SEQ ID NO: 14; or a heavy chain variable region comprising the amino acid sequence of SEQ 11) NO: 29 and a light chain variable region comprising the amno acid sequence of SEQ ID NO: 30; or a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 45 and a light chain variable region comprising the amino acid sequence of SEQ 1D NO: 46; or 10 a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 6i and a light chain variable region comprising the amino acid sequence of SEQ I) NO: 62; or a heavy chain variable region comprising the amnuo acid sequence of SEQ ID NO: 65 and a light chain variable region comprising the amino acid sequence of SEQ ID NO: 62; or a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 79 and a light chain variable region conipri sing 15 the amino acid sequence of SEQ ID NO: 80; or a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 95 and a light chain variable region comprising the amino acid sequence of SEQ ID NO: 96; or a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 95 and a light chain variable region comprising the amino acid sequence of SEQ ID NO: 103: or a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 117 20 and a light chain variable region comprising the amino acid sequence of SEQ ID NO: 118; or a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 1.31 and a light chain variable region comprising the amino acid sequence of SEQ ID NO: 132; or a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 145 and a light chain variable region comprising the amino acid sequence of SEQ Io NO: 146; or a heavy chain variable region 25 comprising the amino acid sequence of SEQ U) NO: 151 and a light chain variable region comprising the amino acid sequence of SEQ I) NO: 146; or a heavy chain variable region comprising the amino acid sequence of SEQ D NC: 165 and a light chain variable region comprising the amino acid sequence of SEQ ID NO: 166; or a heavy chain variable region comprising the amin acid sequence of SEQ ID NO: 181 and a light chain variable region 30 comprising the amino acid sequence of SEQ ID NC): 182; or a heavy chain variable region comprising the amino acid sequence of SEQ iD) NO: 195 and a light chain variable region comprising the amino acid sequence of SEQ ID NO: 196, Methods for chain replacement and for CDR grafting are well known in the art, Originally these methods were developed to humanize non-hman antibodies (generally mouse 35 antibodies) or to select human antibody counterparts having equivalent bioactivity to the non 11 human antibodies. These methods include replacement techniques where only one of the CDRs, for example, the CDR3s, of the non-human antibody are retained and the remainder of the V region, including the framework and the other two CDRs, for example, the CDRs I and 2, are individually replaced in steps performed sequentially (e.g. U.S. Patent Application No. 5 20030166871; Rader, et al. Proc Natt Acad Sci USA 95:8910-15, 1998; Steinberg, et a, J BioI Chem 275:36073-78, 2000; R ader, et al., Jio Chem 275:13668-76, 2000). In addition, methods of crating antibodies with the binding specificities of a reference antiboxdy for a target antigen are described in Patent Application No. WO05/069,970, The methods include transferring, from the refelce antibody to a recipient antibody or antibody 10 fragment, the minimal essential binding specificity of the reference antibody, Examples of regions that can be transferred include, but are not limited to, the transfer of a single CDR segment, for example a CDR3 segment, from the heavy and/or from the light chain, or a D segment, or a CDR3-R4 segment, or any CDR3-FR4 segment that comprises the minimal essential binding specificity determinant. Antibodies created using these methods retain the 15 binding specificity, and often affinity, of the reference antibody. The antibodies, antibody variants or antibody fragments of the invention include antiodies that comprise, inter alia, one or more CDRs, a heavy chain or a light chain of an exemplary antibody of the invention and retain their specificity and ability to neutralize dengue virus infection. 20 Exemplary antibodies of the invention include, but are not limited to, liMB -DVI, 1MB- DV2, 1MB- DV3, IfMB-DV4, HMBDV$, HMB DV6, 1IMB-D7, H1NMB-DV8, MB DV9, HMB-DV10, HMB-DVI 1, HIMB-DV12 HMB-IDVI3, and HMB-DV14. Variants of 1I1MB- )V4 consist ofu heavy chain variants having aminoacid sequence recited in SEQ ID NO; 61 and SEQ Il) NO: 65, and a light chain having the amino acid sequence 25 recited in SEQ 1D NO: 62. The nucleic acid sequences encoding the heavy chain Variants are recited in SEQ f) NO: 63 and SEQ ID NO: 66 The nucleic acid encoding the light chain is recited in SEQ ID NO: 64. Thus, antibodies comprising the HIMB-DV4 variant heavy chains (SEQ I D NOs: 61, 67) and light chain (SEQ I D NO: 62) are included within the scope of the invention. 30 As used herein, the temnm HMB -DV4" is used to refer to any and/or all variants of IMB -DV4, for example those wth heavy chains corresponding to SEQ 1) NOs: 61 and 65 and light chain corresponding to SEQ ID NO: 62. 12 In one embodiment, an antibody cocktail of the invention comprises two or More antibodies selected from the group consisting of IMI-fDV. fIMB-DV2, 11MB-DV3, 11MB DV4. HMBDV5, J1MB-DV6, HMB-DV7, HMB -D VS. lIMB -DV9, JMB-DV10 HMB-DViI, HIMB-DV12, lIMB-DV13, and HIMB-IDW14. In another embodimenta cocktail of the invention 5 comprises three antibodies selected from the group consisting of HM-DV1IMB-DV2. IMB DV3, HMBDV4, H1MB-DVS IIMB-DV6H MB-DV7 *MB-DV8 HMB-V9, HMB-DV10, HMB-DVIl HIMB-DV 12, HMB-D13S and HIMB-DVI4. In vet another emnbodiment, an antibody cocktail of the invention comprises more than three, for example, 4 5,6 7, or 8 antibodiessected from the group consisting of .HMR-DV I, lMB-DV2, 11MB-DV, HIB 10 DV4, HMB-DV5, iIMB-.DV6 HMfDB-DV71 .HM-DV8, IMB-DV9, HIMB-VO, 1IMB-DVI 1, IIMB-DV12, HMB-DV13, and IMB-DV14. In an exemplary embodiment, a cocktail of the invention comprises H1MB-DV$, RMB- DV6, and HMB-DV8 Theinvention further comprises an antibody, or fragment thereof that binds to an epitope capable of binding to an antibody of the invention The invention alo comprises an antibody or 15 an antibody fragment that competes with an antilyxl of the ivention. In another aspect, the invention also includes nucleic acid sequences encoding part or all of the light and heavy chains and CDRs of the antibodies of the present invention In one embodiment, nucleic acid sequences according to the invention include nucleic acid sequences having at least 70%. at least 75%, at least 80%, at least 85% at least 90%, at least 95%, at least 20 98% or at least 99% identity to the nucleic acid encoding a heavy or light chain of an antixxy of the invention. In another embodiment a nucleic acid sequence of the invention has the sequence of a nucleic acid encoding a heavy or light chain CDR of an antibody of the invention. For example, a nucleic acid sequence according to the invention comprises a sequence tat is at least 70%, at least 75%, at least 80%, at least 85%, at least 90%at least 95%, at least 98%,or at 25 least 99% identical to the nucleic acid sequences of SEQ ID Ns:7 -12, 23-28, 39-44, 5560, 73 78, 89-94, 101, 102, 111-116, 126-128, 129, 130, 140- 144, 150, 159-164, 175-180, and 190- 194. Due to the redundancy of the genetic code, variants of these sequences will exist that encode the same amino acid sequences, These variants are included within the scope of the invention, 30 Variant antibodies are also included within the scope of the invention 'Thus variants of the sequences recited in the application ae also included within the scope of the invention, Such variants include natural variants generated by somatic mutation invioduring the immune response or in viro upon culture of immortalized B cdl clones Alernatively, variants may arise 13 due to the degeneracy of the genetic code, as mentioned above or nay be produced due to errors in transcription or translation, Variants may also be introduced to modify the antibody effector function, for instance in the Fc region to reduce the binding of the antibody to an Fc receptor Further variants of the antibody sequences having improved affinity and/or potency may 5 be obtained using methods known in the art and are included within the scope of the invention. For example, amino acid substitutions may be used to obtain anibodies with further improved affinity. Alternatively, codon optimisatin of the nucleotide sequence may be used to improve the efficiency of translation in expression systems for the production of the antibody Further, polynucleotides comprising a sequence optimized for antibody specificity or neutralizing activity 10 by the application of a directed evolution method to any of the nucleic acid sequences of the invention are also within the scope of the invention. In one embodiment variant antibody sequences may share 70% or more (i.e. 75%, 80%, 85%, 90%, 95%, 97%, 98%, 99% or more) amino acid sequence identity with the sequences recited in the application, In some embodimens such sequence identity is calculated with regard 15 to the full length of the reference sequence (i.e. the sequence recited in the application). In some further embodiments, percentage identity, as referred to herein, is as determined using BLAST version 2.1.3 using the default parameters specified by the NCBI (the National Center for Biotechnology Information; http/wwwncbi nlm nihgov/)rBosum 62 matrix gap open penalty=i11 and gap extension penalty=1L. 20 Further included within the scope of the invention are vectors, for example expression vectors, comprising a nucleic acid sequence according to the invention. Cells transformed with such vectors are also included within the scope of the invention. Examples of such cells include but are not limited to, eukaryotic cells, e.g. yeast cells animal cells or plant cels; in one embodiment the cells are mammalian, e.g. human, ClO HEK293TM 5ERC6. NS, myckoma or 25 hybridoma cells. The invention also relates to monoclonal antibodies that bind to an epitope capable of binding an antibody of the invention In one embodiment, the invention includes a monoclonal antibody that binds to an epitope capable of binding a monocdonal antibody selected from the group consisting ofIMIVDV 1, HMB-D iHMEBDV3, IMBDV4, HMB~DV5, Hn13 V6, 30 HMBY7, HAMB-DVS. HMB-DV9, EIMB-DV 10, H3MB -DV I% HMB- DV 12, NB-DV13 and HIM- 14. Monoclonal and recombinant antibodies are particularly useful in identification and purification of the individual polypeptides or other antigens against which they are directed The 14 antbodies of the invention have additional utility in that they may be employed as reagents in inmanoassas, radioimmunoassays (RIA) or enzyme-linked inmmunosorbent assays (EULSA) In these applications, the antibodies can be labelled with an analyticallydetectable reagent such as a radioisotope, a fluorescent molecule or an enzyme, The antibodies may also be used for the 5 molecular identification and characterisation (epitope mapping) of antigens, Antibodies of the invention will typically be glycosylated. N-linked glycans attached to the C2 domain of a heavy chain, for instance, can influence C iq and FcR binding, with aglycosylated antibodies having lower affinity for these receptors. The glycan structure can also affect activity e.g. differences in complement-mediated cell death may be seen depending on the 10 number of galactose sugars (0, i or 2) at the terminus of a glycan's biantennary chain. An antibody's glycans preferably do not lead to a human immunogenic response after administration, Antibodies of the invention can be coupled to a drug for delivery to a treatmemn site or coupled to a detectable label to facilitate imaging of a site comprising cells of interest, such as 15 cells infected with dengue virus, Methods for coupling antibodies to drgs and detectable labels are well known in the art, as aen methods for imaging using detectable labels. Labelled antibodies may he employed in a wide variety of assays, employing a wide variety of labels, Detection of the formation of an antibody-antigen complex between an antibody of the invention and an epitope of interest (a DENV epitope) can be facilitated by attaching a detectable 20 substance to the antibody Suitable detection means include the use of labels such as radionuclides, enzymes, coenzymes, fluorescers, chemiluminescers, chrmmogens. enzyme substrates or co-factors, enzyme inhibitors prosthetic group complexes, free radicals, particles, dyesand the like. Examples of suiable enzymes include horseradish peroxidase, alkaline phosphatase, f-galactosidase, or acetyicholinesterase; examples of suitable prosthetic group 25 complexes include streptavidin/biotin and avidinibiotin examples of suitable fluorescent materials include uumbelliferone, fluorescein, fluorescein isothiocyanate, rhodimine dichlorotriazinylanine fluorescein, dansyl chloride or phycoerythrin;an example of a luminescent material is luminol- examples of bioluminescent materials include luciferase, luciferin, and aequorin; and examples of suitable radioactive material include 1 1, s o , 30 Such labeled reagents may be used in a variety of well-known assays, such as radioinmnoassays, enzyme immunoassays, eg ElIsA; fluorescent immunoassays, and the like. 15 An antibody according to the invention may be conjugated to a therapeutic moiety such as a cytotoxin a therapeutic agent, or a radioactuve metal ion or radioisotope. Examples of radioisotopes include, but are not limited to, 1-131, T 123, 1-125t Y-90, Re-188, Re-I8i, At-211, Cu-67, Bi-2 12, li2Bi2 Pdt - 09, Tc-99, In-I 11, and the. like Such antibody conjugates can be 5 used for modifying a given biological response: the drug moiety is not to be constied as limited to classical chemical therapeutic agents. For example, the drg moiety may be a protein or polypeptide possessing a desired biological activity Such proteins may include, for example, a toxin such as abrin, riciri A, pseudomonas exotoxin calicheanicin bacterial toxin, or diphtheria toxin. 10 Techniques for conjugating such therapeutic moiety to antibodies are well known. See, for example, Arnon el at ([985) "Monoclonal Antibodies for inmunotargeting of Drugs in Cancer Therapy," in Monoclonal Antibodies and Cancer The rapy, ed. Reisfeld et at. (Alan R. Liss, Inc.), pp, 243-256 edHeRsuom ei atL (1987) "Antbodies for Drug Delivery in Controled Drug Delive cd. Robinson et al (2 ed; Marcel Dekker, Inc,), pp. 623-653 'Thorpe 15 (1985)"Antibody Carriers of Cytotoxic Agents in Cancer Therapy: A Review," in Monodknal Antibodies '84 Biological and Clinical Applications, ed Pincher et g pp. 475-506 (Bditrice Kurtis, Milano, Italy, 1985) "Analysis, Results, and FUture Prospective of the Therapeutic Use of Radiolabeled Antibody in Cancer Therapy,"in Monoclona Anibodiesfor Cancer Detection and Therapy. ed. Baldwin et at (Academic Press, New York, 1985), pp. 303-316; and Thorpe el 20 at, (1982) onmunol Rev. 62:119-158, Alternatively, an antibody can be conjugated to a second antibody to form an antibody beteroconjugate as described in reference 4. In addition. Iinkcrs may be used between the labels and the antibodies of the invention [5]. Antibodies or, antigen-blading fragments thereof may be directly labelled with radioactive iodine, indium, yttrium, or other radioactive particle known in 25 the art [6]. Treatment may consist of a combination of treatment wit conjugated and non conjugated antibodies administered simultaneously or subsequently [7, 81 Antibodies of the invention may also be attached to a solid support. Additionally, antibodies of the invention, or functional antibody fragments thereof, can be chemically modified by covalent conjugation to a polymer to, for example, increase their 30 circulating half-I ife, for example. Examples of polymers, and methods to attach them to peptides, are shown in references 9-12 In some enbodiments the polymers may he selected from polyoxyethylated polyols and polyethylene glycol (PEG). PEG is soluble in water at oom temperature and has the general formula: R(Q-CI-CHL O-R where R can be hydrogen, or a 16 protective group such as an alkyl or alkanol group. In one embodiment the protective group may have between I and 8 carbons. In a further embodiment the protective group is methyl The symbol n is a positive integer, In one embodiment n is between I and 1,000. In another embodiment n is between 2 and 500. In one embodiment the PEG has an average molecular 5 weight between 0,000 and 40,X00 in a further embodiment the PEG has a molecular weight between 2,000 and 20,0(10 In yet a further embodiment the PEG has a molecular weight of between 3,000 and 12,0X.. In one embodiment PEG has at least one hydroxy group. In another embodiment the PEG has a terminal hydroxy group, In yet another embodiment it is the terminal hydroxy group which is activated to react with a free amino group on the inhibitor. However, it 10 will be understood that the type and amount of the reactive groups may be varied to achieve a covalendy conjugated PEG/antibody of the present invention. Antibodies of the invention can be modified by introducing random amino acid mutations into particular region of the CR2 or CH3 domain of the heavy chain in order to alter their binding affinity for FcRn and/or their serum half-life in comparison to the unmodified 15 antibodies. Examples of such modifications include, but are not limited to, substitutions of at least one amino acid from the heavy chain constant region selected from the group consisting of amino acid residues 250, 314, and 428, Water-soluble polyoxyethylated polyols are also useful in the present invention. They include polyoxyethylated sorbitol, polyoxyethylated glucose, polyoxyethylated glycerol (POG), 20 and the like. In one embodiment. POG is used. Without being bound by any theory, because the glycerol backbone of polyoxyethylated glycerol is the same backbone occurring naturally in, fr example, animals and humans in mono-, di-, triglycerides, this branching would not necessarily be seen as a foreign agent in the body. In some embodiments PO has a molecular weight in the same range as PEG The structure for POG is shown in reference 13, and a discussion of 25 POGiL-2 conjugates is found in reference 9. Another drug delivery system that can be used for creasing circulatory half life is the liposome. Methods of preparing liposo. me delivery systems are discussed in references 14, IS and 16. Other drug delivery systems are known in the art and are described in, for example, references 17 and 18. 30 Anubodies of the invention may be provided in purified form. Typically, the antibody will be present in a composition that is substantially free of other polypeptides e~g where less than 90% (by weight), usually less than 60% and more usually less than 50% of the composition is made up of other polypeptides 17 Antibodies of the invention may be imnunogenic in nonituan (or heterologous) hosts eig. in mice. in particular, the antibodies may have an idiotope that is inmunogenic in nonghuman hosts, but not in a human host. Antibodies of the invention for human use include those that cannot be easily isolated from hosts such as ice, goats, rabbits, rats, non-primate 5 mamnas. efc. and cannot generally be obtained by humanisation or from xeno--mice Antibodies of the invention can be of any isotype (e g. IgA, Igo, 1gM i. e an t, y or p heavy chain), but wil generally be IgG. Within the IgG isotype, antibodies may be gGI, 1gG2, IgG3 r IgG4 subclass. In one embodiment, the antibody is IgOrl Antibodies of the invention may have a w or a ) light chain. iO Included within the scope of the invention are DENV-neutralizing recombinant or engineered bispecific antibody molecules or antigenbinding fragments thereof: Such antibodies and fragments may comprise a first binding site for an epitope o a first Dengue vrusserotype and a second binding site for a second epiope on the same dengue virus serotype or on a different, for example, a second, third or fourth, dengue virus serotype. The variable domains of 15 the respective binding sizes can be formed as immunoglobulin isotypes of the invention or as heterodimeric Fab, Fab% F(ab') 2 , ScFv or diabodies that can be linked together via one or more peptide linkers. Production of antibodies Monoclonal antibodies according to the invention can be made by any method known in 20 the art, The general methodology for making monoclonal antibodies using hybridoma technology is well known [19, 20]. Preferably, the alternative EBV immortalisation method described in reference 21 is used, Using the method described in reference 21 B cells producing the antibody of the invention can be transformed with EB V in the presence of a polyclonal B cell activator. 25 Transformation with EBV is a standard technique and can easily be adapted to include polyclonal B cell activators. Additional~ ~ ~ ~ ~ ~ ~ ~ ~~~p siuatofcluagrwhaddfeetaonmyptionally, be added during the transformation step to further enhance he efficiency These stimulants may be cytokines such as fL-2 and IL-I In one aspect, I1-2 is added during the immortalisation step to 30 further improve the efficiency of immortaiisation but its use is not essential. The imoraiedBcels produced using these methods can then be cultured using methods known in the art and anibodies isolated thereafter. 18 The anihodies of the invention can also be made by culturingsingle plasma cells in microwell culture Plates using the method described in UK Patent Application 0819376.5. Further, from single plasma cell cultures, RNA can be extracted and single cell PCR can be perfoned using methods known in the art The VII and Vregions of the antibodies can be 5 amplified by RT-PCR, sequenced and cloned into an expression vector that is then transfected into i-K293T ells or other host cells. The cloning of nucleic acid in expression vectors, the transfection of host cells, the culture of the transfected host cells and the isolation of the produced antibody can be done using any methods known to one of skill in the an. Monoclonal antibodies may be further purified, if desired, using filtration, centrifugation 10 and various chromatographic methods such as HPLC or affinity chromatography. Techniques for purification of monoclonal antibodies, including techniques for producing pharmaceutical grade antibodies, am well known in the art. Fragments of the monoclonal antibodies of the invention can be obtained from the monoclonal antibodies by methods that include digestion with enzymes, such as pepsin or 15 papain, and/or by cleavage of disulfide bonds by chemical reduction. Alternatively, fragments of the monoclonal antibodies can be obtained by cloning and expression of part of the sequences of the heavy or light chains, Antibody fragments" may include Eab, Fab' .(ab't and Fv fragments The invention also encompasses single-chain Fv fragments (scFv) derived from the heavy and light chains of a monoclonal antibody of the invention e.g. the invention includes a 20 seUv comprising the CDRs from an antibody of the invention. Alsoincluded are heavy or ight chain monomers and dimers as well as single chain antibodies, eg. single chain FB in which the heavy and Elght chain variable domains are joined by a peptide linked. Standard techniques of molecular biology may be used to prepare DNA sequences coding for the antibodies or fragments or variants of the antibodies of the present invention. Desired 25 DNA sequences may be synthesised completely orin part usingoligonucleotde synthesis techniques. Site-directed mutagenesis and polymerase chain racton PCR}techniques may be used as appropriate, Any suitable hostellvector system may be used for expression of the DNA sequences encoding the antibody molecules of the present invention or fragments thereof Bacterial, for 30 example F coli, and other microbial systems may be usedin part, for expression of antibody fragments such as b and F(abF fragments and especially Pv fragments and single chain antibody fragments, for example, single chain Fys Eukaryotic mammalian, host cell expression systems may be. used for production of larger antibody molecues, including complete 19 antibody molecules. Suitable mammalian host cells include CHO, NEK293T. PERC6, NSO, myelona or hybridoma cells. 'The present invention also provides a process for the production of an antibody of the invention comprising culturing a host cell comprising a vector of the present invention under 5 conditions suitable for leading to expression of protein from DNA encoding the anybody of the present invention, and isolating the antibody molecule. The antibody molecule may comprise only a heavy or light chain polypeptide, in which case only a heavy chain or light chain polypeptide coding sequence needs to be used to transfect the host cells, For production of products comprising both heavy and light chains, the cell line 10 may be transfected with two vectors, a first vector encoding a light chain polypeptide and a second vector encoding a heavy chain polypeptide. AlterMatively, a single vector may be used, the vector including sequences encoding light chain and heavy chain polypeptides. Alternatively, antibodies according to the invention may be produced by i) expressing a nucleic acid sequence according to the invention in a cell, and ii) isolating the expressed 15 antibody product, Additionally, the method may include iii) purifying the antibody. Screening and isolation of 8 cells Transformed B cells may be screened for those producing antibodies of the desired antigen specificity, and individual B cell clones may then be produced from the positive cells The screening step may be carried out by ELiSA, by staining of issues or cells (including 20 infected or transfected cells), a neutralisation assay or one of a number of other methods known in the art for identifying desired antigen specifilty. 'the assay may select on the basis of simple antigen recognitionor may select on the additional basis of a desired function e.g to select neutralizing antibodies rather than just antigenlunding atibodics, to select antibodies that can change characteristics of targeted cells, such as their signalling cascades, their shape, their 25 growth rate, their capability of influencing other cells, their response to the influence by other cells or by other reagents or by a change in conditions their differentiation status, e The cloning step for separating individual clones from the mixture of positive cells may be carried out using inting dilution, micromanipulation, single cell deposition by cell sorting or another method known in the art. 3 The irmortalised B cell clones of the invention can be used in various ways g. as a source of monoclonal antibdiesas a sowce of nucleic acid (DNA or mRNA} encoding a monoclonal antibody of interest for research etc. 20 The invention provides a composition comprising immortalised B memory cells, wherein the cells produce antibodies that neutralize one or more dengue virus serotypes, and wherein the antibodies are produced at >5pg per cell per day. The invention also provides a composition comprising clones of an immortalised B memory cell, wherein the clones produce a monoclonal 5 antibody that neutralizes one or more dengue virus serotypes, and wherein the antibody is produced at ;5pg per cell per day, Exemplary immortalised B cell clone according to the invention include, but are not limited to, 1MB-DVI, HMBWDV2, HMB-DV3, HMB-DV4j H1MB-DV, I11MB- DV6 1MB DV7 H1MB-DV8 I- B-DV9, HMB-DVIGi0 MB-DM Il,, MB-DV12, HiMB-IV13, and 10 IIMB-DV14 Epftopes As mentioned above, the antibodies of the invention can be used to map the epitopes to which they bind. The epitopes recognised by the antibodies of the present invention may have a number of uses. 'he epitope and mimotopes thereof in purified or synthetic form can be used to 15 raise immune responses (ie. as a vaccine, orkfo the production of antibodies for other uses) or for screening patient serum for antibodies that immunoreact with the epitope or mimotopes thereof In one embodiment such an epitope or mimotope, or antigen comprising such an epitope or mimotope may be used as a vaccine for raising an immune response. Ihe antibodies and antigen binding fragments of the invention can also be used in a method of monitoring the 20 quality of vaccines. In particular the antibodies can be used to check that the an igen in a vaccine contains the specific epitope in the correct conformation The epitope may also be useful in screening for ligands that bind to said epitope. Such ligands, include but are not limited to antibodies including those from camel, sharks and other species, fragments of antibodies peptides phage display technology produs, aprtamer, 25 adnectins, synthetic compounds, or fragments of otherviral or cellar proteins, thatnay block the epitope and so prevent infection, Such ligands are encompassed within the scope of the invention. Recombinant expression The imnortalised B memory cells of the invemion may also be used as a source of 30 nucleic acid for the cloning of antibody genes for subseque recombinant expression. Expression from recombinant sources is more common for pharmaceutical purposes than 21 expression from B cells or hybridomas e.g. for reasons of stability, reproducibility, culture ease, tc. Thus the invention provides a method for preparing a recombinant cell, comprising the steps of: (i) obtaining one or more nuclei, acids (e.g. heavy and/or light chain genes) from the B 5 cell clone that encodes the antibody of interest; and (ii) inserting the nucleic acid into an expression host in orter to permit expression of the antibody of interest in that host. Similarly, the invention provides a method for preparing a recombinant cell, comprising the steps of: () sequencing nucleic acid(s) from the B cell clone that encodes the antibody of interest; and (ii) using the sequence information from step (i) to prepare nucleic acid(s) fbr 10 insertion into an expression host in order to permit expression of the antibody of interest in that host. The nucleic acid may, but need not, he manipulated between steps (i) and (ii) to introduce restriction sites, to change codon usage, to optimise transcription and/or translation regulatory sequences, and/or to modify effector function, The invention also provides a method of preparing a recombinant cell, comprising the 15 step of transforming a host cell with one or more nucleic acids that encode a monoclonal antibody of interest, wherein the nuclic acids are nucleic acids that were derived from an immortalised B cell clone of the invention, Thus the procedures for first preparing the nucleic acids) and then using it to transform a host cell can be performed at different times by different people in different places (egin different countries) 20 These recombinant cells of the invention can then be used tor expression and culture purposes. They are particularly useful for expression of antibodies for lairge-scale pharmaceautical production 'They can also be used as the active ingredient of a pharnaceutical composition. Any suitable culture techniques can be used, including but not limited to static cultuM, roller bottle culture, ascites fluid, hollowlfiber type bioreactor cartridge, modular 25 minifermnenterstirred tank, microcarrier culture, ceramic core perfusion. etc. Methods for obtaining and sequencing imimunoglobulin genes from B cells are well known in the art (eg., see reference 22y. The expression host is preferably a eukaryotic cell, including yeast and animal cells, particularly mammalian cells (eg, -10 cells, NSO cells, hunan cells such as PER;C6 [Crucell; 30 eference 23 ar HKB-1 I [Bayer; references 24 & 25] cells, myeloma cells 126 & 271, etc.), as wel as plant cel.Prefered expression hosts can glycosylate the antibody of the invention, particularly with carbohydrate structures that are not themselves immunogenicin hmans. In one embodiment the expression host may be able to grow in serum-free media In a further 22 einbodiment the expression host ray be able to grow in culture without the presence of animal-derived products. The expression hosi may be cultured to give a cell line. The invention provides a method for preparing one or more nucleic acid molecules (e.g. 5 heavy and light chain genes) that encode an antibody of interest, comprising the steps of: (i) preparing an immortalised B cell clone according to the invention: (ii) obtaining from the B cell clone nucleic acid that encodes the antibody of interest The invention also provides a method for obtaining a nucleic acid sequence that encodes antibody of interest, comprising the steps of: (i) preparing an immortalised B cell clone according to the invention; (ii) 10 sequencing nucleic acid from the B cell clone that encodes the antibody of interest. The invention also provides a method of preparing nucleic acid molecule(s) that encodes an antibody of interest, comprising the step of obtaining the nucleic acid from a B cell clone that was obtained from a transformed B cell of the invention. Thus the procedures for first obtaining the B cell clone and then preparing nucleic acids) from it can be performed at very different 15 times by different people in different places (e.g. in different countries). The invention provides a method for preparing an antibody (e g. for pharmaceutical use), comprising the steps of: (i) obtaining and/or sequencing one or more nucleic acids (eg. heavy and light chain genes); (ii) using the sequence information from step (i) to prepare nucleic acids) for insertion into an expression host in order to permit expression of the antibody of interest in 20 that host; (iii) culturing or sub-culuring the expression host under conditions where the antibody of interest is expressed and, optionally (iv) purifying the antibody of the interest. The nucleic acid can, but need not be, obtained and/or sequenced from a B cell clone expressing the antibody of interest; In one embodiment, the nucleic acid from step (i) may, optionally be modified so as to introduce desired substitutions in the amino acid sequence of the antibody. 25 The invention also provides a method of preparing an antibody comprising the steps of culturing or sub-culturing an expression host cell population under conditions where the antibody of interest is expressed and, optionally, purifying the antibody of the interest, wherein said expression h st cell population has been prepared by (i) providing nucleic acid s ) encoding an antibody of interest; (ii) inserting the nucleic acid(s) into an expression host that can express the 30 antibody of interest, and (iii) culturng or sub-culturing expression hosts comprising said inserted nucleic acids to produce said expression host cell population. 23 Pharmaceutical compositions The invention provides a pharmaceuticals composition containing the antibodies and/or antibody fragments of the invention and/or nudei acid encodina such antibodies and/or immortalised B cells that express such antibodies and/or the epiopes recognised by the 5 antibodies of the invention, A pharmaceutical composition may also contain a pharmaceutically acceptable carrier to allow administration. The carrier should not itself induce the production of antibodies harmful to the individual receiving the composition and should not be toxic Suitable carrers may be large, slowly metabolised macrotoleules such as proteins, polypeptides, liposomes. polysaccharides, polylacic acids, polyglycolic acids, polymeric amino acids. amino 10 acid copolymers and inactive virus particles, Pharmaceutically acceptable salts can be used, for example mineral acid salts, such as hydrochiorides, hydrobromides, phosphates and sulphats, or salts of organic acids such as acetates, propionates, malonates and benzoates. Pharmaceutically acceptable caiers in therapeutic compositions may additionally 15 contain. liquids such as water, saline, glycerol and ethanol, Additionally, auxiliary substances, such as wetting or emulsifying agents or pH buffering substances, may be parent in such compositions. Such carriers enable the pharmaceutical compositions to be formulated as tablets, pill, dragees, capsules, liquids, gels., syrups slurries and suspensions, for ingestion by the patient. 20 Within the scope of the invention, forns of administration may include those forms suitable for pareueral administration, e 8, by injection or infusion, for example by bolus injection or continuous infusion. Where the product is for injection or infusion, itmay take the form of a suspensions solution or emulsion in an oily or aqueous vehicle and it may contain fonrulatory agents, such as suspending presentive stabilizing and/or dispersing agents. 25 Alternatively, the antibody molecule may be in dry fam f-or reconstitution before use with an appropriate sterile liquid. Once formuIated, the compositions of the invention can be administered directly to the subject In one eribodiment the compositions are adapted for administration to human subjects. The pharmaceutical compositions of this invention may be administered by any number 30 of routes including, but not limited to, oral,intravenous, intramuscular intra-arteial, intramedullary, intraperitoneal intrathecal i traventricular transdermal transcutaneous, topical subcutarneous, ntranasal, enteral, subingualintravaginal or rectal routes. Hyposprays may also be used to administer the pharmaceutical compositions of the invention. Typically, the 24 therapeutic compositions may be 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 Direct delivery of the compositions will generally be accomplished injection, 5 subcutaneously, intraperitoneally, intravenously or intramuscularly, or deliver to the interstitial space of a tissue. Dosage treatment may be a single dose schedule or a multiple dose schedule Known antibody-based pharmaceuticals provide guidance relating to frequency of administration e~g whether a pharmaceutical should be delivered daily, weely, monthy, et, Frequency and dosage may also depend on the severity of symptoms. 10 Compositions of the invention may he prepared in various forms. For example, the compositions may be prepared as injectables, either as liquid solutions or suspensions Solid forms suitable for solution, or suspension, in liquid vehicles prior to injection can also be prepared (eag a lyophilised composition, like Synagisru and Hercepti*h{, for reconstitution with sterile water containing a preservative) The composition may be prepared for topical 15 administration as an ointmenttream or powder The composiion may be prepared for orM administration e.g as a tablet or capsule, as a spray, or as a syrup (optionally flavoured). The composition may be prepared for pulmonary admnnistration e.g, as an inhaler, using a fine powder or a spray. The composition may be prepared as a suppository or pessary. 'The composition may be preparedA for nasal, aural or ocular administration e.g. as drops . 'The 20 composition may be in kit form, designed such that a combined composition is reconstituted just prior to administration to a patient, For example. a lyophilsed antibody can be provided in kit form with sterile water or a sterile buffer. It will be appreciated that the active ingredient in the composition will be an antibody moleculean antibody fragment or variants and derivatives thereof As suchit wil be 25 susceptible to degradation in the gastrointestinal tract. Thus, if the composition is to be administered by a route using the gastrointestinal tract, the composition will need to contain agents which protect the antibody from degradation but which release the antibody once Iihas been absorbed- fr the gastrointestinal tract. A thorough discussion of pharmaceutically acceptable carriers s available in Gennaro 30 (2000) Remington: The Science and Practice of Phan2nacy0th edition, ISBN: 0683306472, Phannaceutical compositions of the invention generally have a pH between 5.5 and 8.5, in soni embodiments this may be between 6 and 8, and in further enbodiments about . The pH may be maintained by the use of a buffer. The composition may be sterile and/or pyrozen free, 25 The composition may be isotonic with respect to humans In one embodiment phannaceutical compositions of the invention are supplied in hemieticallygsealed containers Pharmaceutical compositions wil include a therapeutically effective amount of one or more antibodies of the invention and/or a polypeptide omprising an epitope that binds an 5 antibody of the invention ie. an amount that is sufficient to treat, ameliorate, or prevent a desired disease or condition, or to exhibit a detectable therpeuti effect Therapenudc effects also include reduction physical symptoms. The precise effete amount for any particular subject will depend upon their size and health, the nature and extant of the condition, and the therapeutics or combination of therapeutics selected for administration. The effective amount for 10 a given situation is determined by routine experimentation and is within thejudgment of a clinician, For purposes of the present invention, an effective dose will generally he from about 0.01mg/kgo to about or about 0.05 mg/kg to about 10 rmg/kg of the composition of the present invention in the individual to which it is administered Known antibodybased pharmaceuticals provide guidance in this respect e. I terceptin'I i s administered by 15 intravenous infusion of a 21 mg/mil solution, with an initial lading dose of 4mgkg body weight and a weekly maintenance dose of 2mg/kg body weight; RitwxanM is administered wee kly at 375mg/n; et:% In one embodiment pharmaceutical compositions can include more than one (eg 2,3, 4, 5, 6, 7, 8, etc.) antibody of the invention. In another embodiment the composition comprises two 20 ormore (e.g. 2, 3, 4, 5, etc.) antibodies, wherein the fit antibody is specific for a first DENV epitope, and the second antibody is specific forasecon DENV epitope In yet another embodiment, the pharmaceutical composition comprises three antibodies of the invention. In another embodiment~ the composition comprises two or more (e~g 2,3. 4, ete. antibodies, that together neutmlise more than one dengue virus serotype. In yet another embodiment, the two or 25 more antibodies of the invention together neutralise all four dengue virus serolypes, DENV4, DEN V2, DENV-3 and T)ENV4. In a further embodimenttwo or more antibodies of the invention together neutralize all four dengue virus serotypes by binding at least two distinct epitopes on each dengue virms serotvpe. Exemplary antibodies of the invention for use in a pharmaceutical composition that 30 neutralize a dengue virus without contributing to antibody-depe adent enhancement of dengue vims infection include, but are not limited to, HMW-DV 1, HMB-DV2 HMINB-DV3M, }MB-DV4, 1M8DV5, IMB DV6, HMB-DVY, PIMWDV8, IMB-DV9, HIMB- DV 10, HM -YVI i. HMB)V H2. IMB-DV 13, and HIBDV 1-4 26 In one embodiment, a pharmaceutical composition includes two exemplary antibodies of the invention, for example, HMB-DV3 and H1MB-DV7: IMB-DV3 and HME-DV; IMB-DV3 and HMB-DV 12; IIMB-DV3 and H1MB-D V14: h1MB-)V6 and HMB-DV7; HMB-DV6 and HMB-DV8. In another embodiment, a pharmaceutical composition includes three exemplary 5 antibodies of the invention, for example, HMB-DV2, HMB-DV3 and HMB-DV; HMB-DV2, HMB-DV6 and IMB-DVS; I{MB-DV2, -II DV8 and HMB-DV9; HMB-DV2, IMB-DV8 and HIMB-DV12; 1HMB-FV2, HMBDV8 and HMiB-DY14; HMBDV5 FMB-DY6 and HMB-DV8. Based on the teachings hereinone of skill in the an can determine other combinations of antibodies for use in a pharmaceutical composition .10 in one embodiment, the invention prvides a pharmaceutical composition comprising the antibody 1MB-DV1 or an antigen binding fragment thereof and a pharmaceutidally acceptable diluent or carrier. In another embodiment, the invention provides a pharmaceutical composidon comprising the antibody HM.B-DV2 or an antigen binding fragment theeof, and a pharmaceutically acceptable diluent or carrier. In another embodiment, the invention provides a 15 pharmaceutical composition comprising the antibody IMB-DV3 or an antigen binding fragment thereof, and a pharmaceuticaly acceptable dihient or carrier, In another embodiment, the invention provides a pharmaceutical composition comprising the antibody HMBDV4 or an antigen binding fragment thereof, anad a phrmcetialcet~able diluent or carrier, In yet, another embodiment, the invention provides a pharmaceutical composition comprising the 20 antibody HIMB -DV5 or an antigen binding fragment thereof, and a pharmaceutically acceptable diuent or carrier. In another embodiment, the invention provides a pharmaceutical composition comprising the antibody HIMB-DV6 or an antigen binding fragment thereof, and a phannaceutically acceptable diluent or carrier, In another embodimentthe invention provides a pharmaceutical composition comprisig the antibody HMB-DY7 or an antigen binding fragment 2$ thereof, and a pharmaceutically acceptable diluent or carrier In yet another embodiment, the invention provides a pharmaceutical composition comprising the antibody HMB-DV8 or an antgen binding fragment thereof and a pharmaceutically acceptable diluent or carriern Manother embodiment, the invention provides a pharmaceutical composition comprising the antibody H-MB3-DV9 or an antigen binding fragment 30 thereof, and a pharmaceutically acceptable diluent or carrier. in another embodiment, the invention provides a pharmaceutical composi tion comprising the antibody 1-IMB-D10 or an antigen binding fragment thereof and a pharmaceutically acceptable diluent or carrier- In vet another embxxAnent, the invention provides a pharmaceutical composition comprising the antibody 11MB-DVII or an antigen binding fragment thereof and a pharmaceutically acceptable 27 diluent or carrier, In another embodientt the invention provides a pharmaceutical comiposition comprising the antibody HMBADV 12 or an antigen binding fragment thereof and a pharmaceutically acceptable diluent or carder In another embodiment theinvention provides a pharmaceutical composition comprising the antibody HMIV1or an antigen binding $ fragment thereof, and a phamnaceutically acceptable diluent or carrier, In yet another embodiment, the invention provides a pharmaceutical composition comprising the antibody HMB -DV14 or an antigen binding fragment thereof, and a pharmaceutically acceptable diluent or carrier. Antibodies of the invention may be administered (either combined or separtely) with 10 other therapeutics etg. with chemotherapeutic compoundswith radiotherapy. etc. Preferred therpeutic compounds include anti-iral compounds. Such combination therapy provides an additive or synergistic improvement in therapeutic efficacy relative to the individual therapeutic agents when administered alone. The term "synergy" is used to describe a combined effect of two or more active agents that is greater than the sum of the individual effects of each respective 15 active agent, Thus where the combined effect of to vor more agents results in "synergistic inhibition" of an activity or process, itis intended that the inhibition of the activity or process is greater than the sum of the inhibitory effects of each respective active agent- The term synergistic therapeutic effect" refers to a therapeutic effect observed with a combination of two or more therapies wherein the therapeutic effect (as measured by any of a number of parameters) 20 is greater than the sum of the individual therapeutic effects observed with the respective individual therapies. in compositions of the invention that include antibodies of the invention, the antibodies may make up at least 50% by weight (eg 0%, 0% 75%, 80% 85%, 90%, 95%, 97%, 98%, 99% or more) of the total protein in the composition The antibodies are thus in purified form, 25 The invention provides a method of preparing a phannaceutical, comprising the steps of: (i) preparing an antibody of the invention; and (ii) admixing the purified antibody with one or more pharmaceutically acceptable carriers The invention also provides a method of preparing a pharmaceutical, comprising the step of admixing an antibody with one or more pharmaceuticallyracceptahie carriers, wherein the 30 antibody is a monoclonal antibody that was obtained from a transformed B cell of the invention Thus the procedures for first obtaining the monoclonal antibody and then preparing the pharmaceutical can be performed at very different times by different people in different places (e-g in different countries). 28 As an alternative to delivenag antibodies for therapeutic purposes, it is possible to deliver nucleic acid (typically DNA) that encodes the monoclonal antibody (or active fragment thereof) of interest to a subject, such that the nucleic acid can be expressed in the subject in situ to provide a desired therapeutic effect Suitable gene therapy and nucleic acid delivery vectors are 5 known in the art. Compositions of the invention may be immunogeoi conapositions and in some embodiments may be vaccine compositions comprising an antigen comprising a DENy epitope, Vaccines according the invention may either be prophylactic e. to prevent infection) or therapeutic (i.e. to treat infection) 10 Compositions may include an antimicrobial, particularly i packaged in a multiple dose format. Compositions may comprise detergent e.g, a Tween (polysorbate, such as Tween 80. Detergents are generally present at low levels e.g, <0,01%. Compositions may include sodium salts (e~g. sodium chloride) to give tonicity. A concentration of 10±2mg/ml NaCI is typical. Compositions may comprise a sugar alcohol (eg nannitol) or a disaccharide (eg 15 sucrose or trehalose) e g at around 15-30mg/mi (eg 25 ma/mI) particularly if they are to be lyophilised or if they include material which has been recontituted from lyophilised material, The pH of a composition for lyophilisation may be adjusted to around 6. prior to lyophilisation The compositions of the invention may also comprise one or more nnmunoregulatory agents. In one enbodiment, one or more of the imnunoregulatory agents includes) an adjuvant 20 Medical treatments and uses The antibodies, antigen binding fragments, derivatives and variants thereof, or the cocktails and pharmaceutical compositions of the invention can be used for the treatment of DENV infection, for the prevention of DENV infection or for the diagnosis of DENy infection. Methods of diagnosis may include contacting an antibody or an antibody fragment with a 25 sample. Such samples may be tissue samples taken from for example, salivary glands, lung, liver pancreas, kidney, ear, eye, placenta, alimentary tract, heart ovaries, pituitary, adrenals, thyroid, brain or skin. The methodsof diagnosis may aho include the detection of an antigantintibody complex. 'The invention therefore provides (i) an antibody an antibody frament, or variants and 30 derivatives thereof acconjing to the invention, (ii) an immortalized B cell clone according to the invention, (iii) an epitope capable of binding an antibody of the invention or (iv) a ligand, 9 preferably an antibody, capable, cif bi ding an epitope that binds, an antilbody or the inventi, for use in therapy. Also provided is a method of treating a subject comprising administering to that subject (i) an antibody, an antibody fragment, variants and derivatives thereof, or a pharmaceutical 5composiion according to the invention, or, a ligand, preferably an antibody, capable of binding an epitope that binds an antibody of the invention. The invention also provides the use of (i) an antibody, an antibody fragment, or variants and derivatives thereof according to the invention, (i) an immortalised B cell clone according to the invention, (iii) an epitope capable of binding an antibody of the invention, or (iv) a ligand, 10 preferably an antibody that binds to an epitope capable of binding an antibody of the invention, in the manufacture of a medicament for the prevenion or treatment of DENV infection, The invention provides a pharmaceutical composition for use as a medicament for the prevention or treatment of DENV infection, It also provides the use of an antibody of the invention and/or a protein comprising an epitope to which such an antibody binds in the 15 manufacture of a medicament for treatment of a patient andor diagnosis in a patient. It also provides a method for treating a subject, e.g., a human subject. The method comprises the step of administering to the subject a therapeutically effective dose of a composition of the invention. One way of checking efficacy otherapeutic treatment involves monitoring disease symptoms after administration of the composition of the invention.. Treatment can be a single dose 20 schedule or a multiple dose schedui. In one embodiment, an antibody, antibody fragment, antibody variant, epitope or pharnaceutical composition according to the invention is administered to a subject in need of such treatment Such a subject includes, but is not limited to, one who is particulady at risk of or susceptible to DENT infection. 25 Antibodies of the invention can be used in passive immunisation. Antibodies and fragments or variants thereof, or a nucleic acid encoding an antibody or an antibody fragment or variant as described in the present invention may also be used in a kit for the diagnosis of dengue virus ifection, E~pitopes capable of binding an antibody of the invention, a g. he nt onoClonal antibodies 30 HIMBWI. FIM DV2, HMB-DV3, IMBDV4, HMvBDV5, HMB-DV6 HMBDV7, HM DV8 HMB>DV9 HIMB-DV 10, HMAIPDVI , HMANDVi2, lMB- DV 13.and HIMB4DV14, may be used in a kit for monitoring the efficacy of vaccinati procedures by detecting the presence of protective anti-DENV antibodies. 30 Antibodies, antibody fraginents, or variants and derivatives thereof, as described in the present invention may also be used in a kit for monitornig vaccine manufacture with the desired immunogenicity The invention also provides a method of preparing a pharmaceutical composition, 5 comprising the step of admiring a monoclonal antibody with one or more phannaceutically acceptable carriers, wherein the monoclonal antibody is a monoclonal antibody that Was obtained from an expression host of the invention 'Thus the, procedures for first obtaining the monoclonal antibody (e.g expressing it and/or purifying it) and then adnixing it with the pharmaceutical carrier(s) can be performed at very different times by different people in different places (e.g in 10 different countries), Starting with a transformed B cell of the invention, various steps of culturing, sub-culturing, cloning, sub-cloning, sequencing, nucleic acid preparation etc' can be perfonned in order to perpetuate the antibody expressed by the transformed B cell, with optional optimnisation at each step, In a preferred embodimentdte above methods further comprise 15 techniques of optimisatio (.. affinity maturation or optimisation) applied to the nucleic acids encoding thentxdy. The invention encompasses all cells, nuclei acids vectors sequences, antibodies etc. used and prepared during such steps. In all these methods, the nucleic acid used in the expression host may be manipulated to insert, delete or amend certain nucleic acid sequences. Changes from such manipulation include, 20 but are not limited to, changes to introduce restriction sites, to amend codon usage, to add or optinise transcription and/or translation regulatory sequencesetc. It is also possible tco change the nucleic acid to altrthe encoded amino acids. For examplit may be useful to introduce one or more (e.g. 1, 2, 3z 4, 5. 6, 7. 8, 9 10, etc) amino acid substitutions deletions and/or insertions into the antibody's amino acid sequence. Such point mutations can modify effector functions 25 antigen-binding affinity, post-translational modifications, inmunogenicity etc, can introduce anino acids for the atachment of covalent groups (g abelsor can introduce tags (eg for purification purposes. Mutations can be introduced in specific sites or can be introduced at randomt followed by selection (e.g. molecular evolution), For instance, one nr more nucleic acids encoding any of the CDRregions, heavy chain variable regions or light chain variable 30 regions of antibodies of the invention can be randomly or directionally mutated to introduce different properties in the encoded amino acids. Such changes can be the result of an iterative process wherein initial changes are retained and new changes at other nucleotide positions are introduced. Moreover changes achieved in independent steps may be combined. Different 31.
properties introduced into the encoded amino acids may include, but are not limited to, enihanced affinity, General The tern "comprising" encompasses "including" as well as "consisting" e.g. a 5 composition "comprising" X may consist exclusively of X or may include something additional e.g. X + Y The word "substantially" does not exclude "completely" eg a composition which is "substantially free" from Y may be completely free from Y- Where necessary, the word "substantially" may be omitted from the definition of the invention, 10 'The term "about" in relation to a numerical value x means, for example, xtl0% The tern "disease" as used herein is intended to be generally synonymous, and is used interchangeably with, the termsdisorder" and"condition" (as in medical condition, in that all reflect an abnormal condition of the human or animal body or of one of its parts that impairs normal functioning, is typically manifested by distinguishing signs and symptonis, and causes 15 the human or animal to have a reduced duration or quality of life. As used herein, reference to "treatment" of a patient is intended to include prevention and prophylaxis as wells therapy. 'the term "patient" means all mammals including humans, Generally, the patient is a human EXAMPL ES 20 Exemplary embodiments of the present invention are provided in the following examples, The following examples are presented only by way of illustration and to assist one of ordinary skill in using the invention, The examples are not intended in any way to limit the scope of the invention. Example L Cloning of B cells and screening for idenficadion of Dengue virus specific Ak 25 Memiry B celh were isolated from the blood of DENV immune donors and immortalized using EBV and CpG as described in reference, Briefly, IgG t memory B cells were isolated using CD22 heads, followed by removal of Ig t . .IgfV igA* B cells using specific antibodies and cell sorting. The sorted cells (Ig') were immortalized with EBV in the presence of CpG 2006 and irradiated allogeneic mnonuclear cells. Replicate cultures each containing 30 30 50 memory B cells were set up in several 96 well U-bottom plates. After two weeks the culture supernatants were collected and tested for their capacity to stain C6/36 cells infected with DENY of serotypes 1, 2, 3 or 4 by immunofluorescence analysis and/or to hind to recombinant DENVI 4FE2 proteins by ELISA. Supernatants were tested for their capacity to neutralize DENV infection of either VERO cells or DC-SIGN-transfected Raji cells and to enhance infection of K-562 cells. B cell clones were isolated from positive polyclonal cultures as described previously 5 [281, TgG concentrations in the supematants of selected clones were determined using an IgG specific ELB$A. Example 2. Human mAbs from immortalized B cells recognize Dengue virus proteins and neutrdlie infection. For the viral neutralization and viral enhancement assay! titrated amounts of attenuated 10 DENV of serotypes 1, 2, 3 or 4 were mixed with an equal volume of culture supernatants, Viruses and muluplicity of infection (Mi) used were; rDENIA30.- (O031B186- lA+V2) MOI 0.04; rDEN2i4A30 (04 BV351-l.A-V2) M1 0.04; rDEN3/4&30 (DEN3#107C) MI 0.02; rDEN4A30 (06JBV591-V'3-+IAi+v2) MOI 0.04. After 1[hour incubation at room temperature thermixture was added to target cells (e.g. VERO cells DC-SIGN -Raji cell, or K562 cells) in 96 15 well flat bottom plates and incubated at 37*C for 72-96 hours. The cell were then stained with a mouset monoclonal antibody to Dengue virus 1-4 F proteins cone 42), followed by a fluorescein-labeled goat anti mouse Ig and analyzed byFAGS. The neutalzing tier is indicated as the concentration of antibody (ggfml) that gives a 50% reduction of DENV infection For identification of the target antigens recognized by the monoclonal antibodies yeasts 20 displaying Dengue virus F protein domains III or domain 1-11 were stained with the monoclonal antibodies followed by Cy54-abeled goat ani human. lU antibodies and analyzed by FACS. Western blotting experiments were performed using lysates Cf DENV-infected cells Table 3 shows that three different types of antbodies have been identified They include those that are specific for domain III (DIll) of F protein, those that are specific for domains 1-i. 25 (DI-fl) of E protein and those specific for prM. The antibodies show different degrees of cross reactivity with the 4 different DENV serotypes and neutralize those serotypes to which they bind 33 Table 3 Target Antigen Specificity of Neutralixing anti-Dengue Virus Antibodies Aengem Virus serotypes Antiody arge AntgenNeutralized HM.D - - E, 1I- 1,2,3 81MB-D) 12 E, DII 1,3 UMB-DV-3 prM 1,234 H- MB-DV-4 E, D1-1U 1,2,34 --- 1-f i ................. ..... --------.......... HMB-DV-5 E, DI-I 1,2,34 IIMB-DV-6 E, Ill L2,3 H1MB.DY-7 E___ D____U __L12,3 HMB-DV- S E 4 HMB-DV- 9 E. Dlii 2 I-LMWDV-1O 2 ~, HM- 0E DUI 1;121 4 HMB-DY-12 E 5 9191 HM$.DV-13 E D9ll 1,2,3 4 HMB-DV-4 E D-1- 2 Table 4 shows the results of vims neutralimton assays on VERO cells and DC-SIGN transfected Raji ols. Table 4, Neutrinzation of Dengue Vims (serotypes IN -DENV4) by Antibodies Neutralizatkm Antibody Cell type )ENXVI DENV2 D flENV3 DENV4 VERO 0013 0.77 0.014 >20 DC-SIGN- Raji 0.03 5.340 0.055 > 20 VERO 0 06 > 20 0)06 >20 HIMB-DV- 2. .____________ DC-SGN-Ra j 0.014 > 20 0.013 > 20 VERO o12 _ L615 012 0 0,070 DC-SIGN-Raji ND ND ND N*D --- -0 02 5 j..80 0 367 __________DC.SION-Raji 2.25 1 370 0.63 20 VEIRO .066 o.034 0J18 0.200 DCSIN-Raji 39 0,504 0,348 > 20 VERO C.00 o.00 j .0oOM > 20 DC- SIGN-Raji ,7 0A40 0332 > 20 VER) 0,016 00 0,020 > 20 DC-SIGN-Raji ND ND ND NID V1DY ERO >__ 20'20 > 20 0.006 HM BD---- ----- - - - -- --- DC-SION- Raji ND N ND ND) VERO 2 002 > 2 >20 DC-SiON Raji ND NI) N) ND VERO) > 20 00 > 20 52 If NO_____ifD)-SIGN-RaND ND ND N----- VEi M> N0 00 v62 -- -- ------ ----- DM i NR ND ND ND N) RVMB-DV.12 ERO0----- ---- ND ----- Q P3 ,, -I N D DC SiGN-Ra ND ND) ND ND HM-V-3 vr;RO 93 1.z3 > 20 RD> 20 00> 0 1IM BtDV 14 V.N ... >------ --- -- DC- SIGN-Rji j ND ND - ND NI) ND: not determine Example 3. Neutralizing recombinant anti-Dengue virus antibodies with mutations in the Fe region do not cause enhancement of virus infection on K562 cells, Antibody-dependent enhancement (ADE) of dengue virus infection has been described in the 5 literature. This property could limit the rapeutic effectiveness of anti-dengue virus antibodies for use in clinical situations. Therefore, mRNAs frotm the immortalized [ cell lines expressing antibodies HMB-DV-5, HMBDV-6 and HMB-lV-8 were isolated, eDNA was synthesized using oligo-di specific primers, variable regions of heavy and light chain were sequenced and cloned into an expression vector using specific primers. Vectors were transfected into host cells for recombinant 10 expression. In addition to recombinant production of the wild-type IgG1 antibodies, each of the heavy chains was mutated at amino acids 4 and 5 of CH2 domain by substituting an alanine in place of the natural leucine using site-directed mutagenesis thereby creating the LALA variant of each antibody, Both recombinant wild type and mutated antibodies were harvested from the expression cell lines and purified. Both wild-type IgGI anti-dengue virus antibody and the LALA variant bound 15 to the target protein in comparable marner (data not shown). Virus neutralization and enhancement was deermed as.above on VERO cells and K562 cels Each of the three antibodies has a defined molecular target a wel as serotype target (see Table 3). Figure 1 shows that the unmodified recombinant antibodies neutralize target virus infection of VEROC cells in a dose-dependent manner (DOTED .INE On K562 cells, a cell line that is not 20 efficiently infected by Dengue viruses, the unmodified antibodies show a enhancemenof viral infection at t are generally higher that those required for neutralization (SOD LINES). The experiment was repeated using the I-ALA variants of each antibody, Figure 2 shows that each of the LALA varians of the recombinant anti-dengue virus antibodies also neutralized the target virus on VER( cells (DOTTED LNES) in a dose-dependent manner. However, each of the 25 LALA antibodies did not show evidence of antibody-dependent enhancement of infection on K562 35 cells (SOID LINES). Note, the dose-response is flat on the K562 cells at the concentrations of antibodies used in this experiment and the line appears very close to the X-axis. All patents and publications infenred to herein am exprssly incorporated by reference in their entirety, 5 It should be noted that there are alternative ways of implementing the present invention and that various modifications can be made without departing from the scope and spirit of the invention. Accordmgly, the present embodiments ae to be considered s illustrative and not resrictive, anM the invention is not to he limited to the details given herein, hut may be modified within the scope and equivalents of the amppended claims. 36 RESERENCES (the contents of kich are hereby incorporated by reference) [1) Lefranc et al (2003) Dev Comp Immunol 27(1):55-77. [21 Lefranc el a0 (1997) Inmmunology Today, 18:509. [3] Lefranc (1999) The Immunologist, 7:132-136. [41] US 4,676,980 [51 US 4, 831,175 [6) US 5,59,3721 [71 W000/52031 [8] W00/52473 [91 US 4,766,106 [101 US 4.179337 [11] Us 4,495,25 [121 US 4,609,546 [13J Knauf at (1988J. Bio, Chenm 263:15064-15070 [141 Gabiron e at (1982) Cancuer Research 42:4734 115] Cafico (1981) Biochem Biophys Ara 649:129 [16] Szoka (1980)Ann Rev Bioph ys tng 9:467 [17) Poznansky ei at (1980) Drug Delivery Systems (R. Juliano, ed, Oxford, N.Y) pp. 253-15 (7181 Powaausky (1984) F/turn Revs 36:277 [19] Kohler, G, and Milstein, C_ 1975, Nature 256:495497, [20] Kozbar et al 1983, Immunokgy Tdy 4:72. 1121] W02004/076677 [22] Chapter 4 of Kuby Immunology (4th edition, 2000; ASIN: 0716733315 [23] Jones et at BiOtechnol Prog 2003,19(1): 163-8 [24] c oeholy 2001,37:23 [253 Cho et at Biotechno Prog 200,19:229-32 [261 US 5,807,715 [271 US 6,300, 104 [283 Traggiai et a. (2004) Nat Med 10(8):871-875 37

Claims (3)

  1. 4. The composition of any one of claims 1 to 3, wherein the antibodies, Or antigen binding fagmeits thereof are monoclonal antibodies or recombinant antibodies 5 The composition of any one of claims I to 4, herein the or an antigen finding figments hereo comprise at least one complementarity determining region (CDR) sequence having the sequence of any one of SEQ ID NOs: 16 1722 338,
  2. 49-54, 67T72, 830S. 1 19 1(A, 105-110. 121.123, 124. 12$, 13 5-139, 149. 1 S3-158, 1694174, 1854 88, or 189. 6. The composion of anyone of claims 1 to 4. wherein the antibodies, or an angien consisting o. SE 1i) NOs: 1, 17, 33,49, 67. 83, 105., 135. 149, 153, 169. and 185; a heavy chain CDR2 selected from the group consisting of SEQ ID NOs2 18. 34, 50, 6S. 84, 1 06K 122. 1 3, 154, 170. and 1 6;nd at heavy Chain CDR selected 38 troim the group consisung of SEQ ID NOs:3 1935,51, 69, 85 107 123 7, 1s5, 17 i and 187, i The composition of any one of claims 1 to 5 whereinethe antibdies, or an antigen binding fragments thereof, comprise a light chain CURl selected from the group consistng of SEQ ID NOs: 4, 20, 36. 52. 70, 6 99. 108, 38. 156, 172, and 188; a Hght chain CDR2 sekcted from the group conssting of SEQ 11) NOs: 5, 21 3753 71, 87, 109. 124J 17, .and 73; and a light chain CDR3 selected from the group conssting of SEQ iD N 6 22, 38 54, 72, 88 100, 110. 125, 139, 158, 174, and
  3. 189. 8. The composition of clain 6. whereinl the antibodiesran antigen binding fragments thereof comprise a heavy chain comprising SEQ ID NO: I for(CDRHtSEQ ID NO: 21-brTM1DR2, SEQ I) NO: 3 for CDRH3 SEQ ID NO: 17 for CDRiD SEQ ID NO 18 for CDRH2 and SEQ IU NO: 19 forCDR4H3: SEQ ID NO: 33 for CDRil SEQ ID NO: 34 for DRH2 and SEQ ID NO: 35 for DRI3B; SEQ [D NO: 49 for (DRII, SEQ ID NO: 50 for (DRF12 and SEQ ID NO: 51 forCDRH3; SEQ ID NO: 67 for CUiHI SEQ I) NO 68 for CUAR H and SELQ ID NO: 69 for (DRH3; SEQ ID NO: 83 for CIDRH1. SEQ ID NO: 84 for CDRE12 aid SEQ I) NO: 85 for CDRH3; SEQ ID NO15 for DlCD R SEQ ID NO: 106 for CDRH2 and SEQ ID NO: 107 for DRI SEQ I) NO: 127.1 for (DRll. S I) NO: 122 fo CDRI12 and SEQ ID NO: 123 for CDR3; SEQ ID NC) 135 for CDRHI, SEQ ID NO: 136 for CDRH2 and SEQ D- NO:1 37 for CPDR3; SEQ ID NO: 149 fOr CDRI6, SEQ ID NO 136 fr DRN2 and SEQ ID NO: 137 for CD13; SEQ ID NO: 153 (or CDPE, SEQ ID NO: 154 for CDRH2 and SEQ ID NC: 1 55 for(DRH3; SEQ ID NO: 169 for CDRI-H SEQ IDNO 170 for CDRii2 and SEQ ID NO:17 for mDRH;and SEQ ID NO: 135 for CDRHM SEQ ID NO: 186 ft CDR2 aind SEQ ID NO: 187 for 9. The composition oclam 7. wherein te anti bodies, or an antigen binding fragmems thertf eomprksc a ligh chainco g S EQ ID NO 4 fOr DRI SEQ IDI NO: 5 or CDR I'SEQ ID NO 6 or DR3 SEQ iD NO:2 r It CDRI STEQ ID N 21 39 for CDRE2; SEFQ ID NO: 22 3 CUR; SEQ if NO: 36 for CDR[l SEQ ID NO: 37 for CURIA; SEQ ID NO::8 for R:SEQ (D NO: 52 for CbL SEQ ID NO: 53 for DCR2; SEQ ID NO: 54 foWDIRII 3, SEQI D NO: 70 .r CDRI] SEQ ID NO: 71 forCDRL 2; SEQ 10 NO: 72 forDR3; SEQ 1) NO:86 for CURLE SEQTD NO: 8?for CDR 2SEQEQ ID NO: 88f CURLSSEQ IL NO: 99 for CDRL, SEQ ID) NO: 53 for CDR2 SEQ ID NO: 100fr CDRL3; SEQ ID NO: 108 for (RI, SEQ ID NO: 109 for CDRI 2 SEQ ID NO: 110 for CDRL3; SEQ ID NO: 70 for CDRI-, SEQ ID NO: 124 for CDR2; SEQ ID NO: 125 for CDRL3; SEDQ ID NO: 138 frURIA SEQ ID NO: 109 for CRL2; SEQ ID NO: 139 for CIDRL3. SEQ ID NO: 156 for CDREL SEQ ID NO: 157 for CDRL2: SEQ ID NO: 1 8 for CDRL3; SEQ If) NO: 172 fOr CURd, SEQ ID NO: 173 for (DRI2; SEQ ID NO: 174 for CDRL3: and SEQ U NO:188 for CDRLI, SEQ I NO37 for CURLI2 SEQ ID NO: 189 for CDRIS. 10. The composition of daim 8, whereti the antibodie, or an antigen binding fragments thereof comprise a light chain comprising SEQ UD NO: 4 for CDRL, SEQ ID NO: 5 for CDR 2 SEQ ID NO: 6 for CDRL; SEQ ID NO: 20 (r CUR I SEQ ID NO: 21 for CUR. ;E INO: 22 for CDRL3 SEQ ID NO: 36 for GURI.SEQ I) NO: 37 for CDR E2: SEQ I NO: 3(r CURlS; SEQ 1D NO:D for 52 xo RU. SEQ ID NO: 53 for CRL2, SEQ [D NO: 54 for CURL3; SEQID NO: 70 fDr CR1 ,SEQ ID NO: 71 for CDRL-2; SEQ I) NO: 72 fOr CURLS; SEQ ID NO:86 for CDRI. SEQ I) NO 87 for CDRL2; SEQ ID NC: 88 for CRLI SEQ ID NO 99 for CI) lDRSEQ IL NO:5 (r CRL2;SEQ I) NO100 for CUR:SEQ IL) NO: 108 forCDRLi, SEIMID NO: 10) for CURI2;SEQ ID N):O 10 fr CDRIL<V SE QID NO: 70 for CDRil, SEQ ID NO: 124for CURLS: SEQ 1D NO: 25 forCURS; SEQ ID NOi I 38 for CDRLE SEQ 1D NO: 109 for CURLS; SEQ HI)NO: 139 (hr CURL3; SEQ ID NO: 156 for CUR~I, SEQ Ii)NO: 157 for CURLS2; SEQIDU NO. 158 for CDRLS; SEQ ID) NO: 172 for CDREL SEQ ID NO: 1%for CURI2: SEQ ID NO: 14 for CURI ; and SEQ L No 188 for CURI SEQ ID NO: 37 rD CRL2: SEQ ID NO: 189 for CURi. 40 IL The compositon of any one of caims I to 4. wherein the antibodies, or an anigen binding fragments thereof comprise a hea) chai variable region having at least 70% sequence identity to any one of SEQ ID NOs: 139 45. 61. 659, 7995, 117, 131, 145, 151, 165, 8 1 or 195. 12 The composition of any one of claims I to 4. erein the antibodies, or an antigen binding frawents thereof comprise a eight ain variable region haing at least 70% sequence identiy to any one of SEQ ID NOs: 14, 30. 46, 62, SO 96, 103. 118, 132, 146, 166, 182, or 196, 13 The composition of any one of claims I to 4, wherein the antbodies, or an antigen binding frgments thereof, comprise a heavy chain variable region comprising the amino acid sequence of any one of SEQ ID NOs 13 29, 45, 61, 65, 79 95,117, 131, 145, 151, 165, I81, or 195, and a light chai variable region comprising the amino acid sequence of any one of SEQ ID Ns: 14, 30, 46, 62, 80,96, 103, 118, 132. 146 166, 182, or 196, 14t The composition of any one of claims 1 to 4 wheein the antibodies, or an antigen binding fragments thereof comprises a heavy chain variable region comprising the ami aci sequence of SEQ 11) NO; 13 alight chain variable region comprsin the amino acid sequence of SEQ ID NO: 14; or a heavy chain variable region comprising theamino acid sequence of SEQ D NO; 29 and a light chain variable region comprisingthe amino acid sequence of SEQ D NO: 30: or a heavy chatn variable reion comprising the amino acid sequence oEQ ID NO; 45 and a tight cin vaai egionomprising the amino acid sequenceof SEQ D NO: 46; or a heavy chan vaiale region comprising the amino acid sequel of SEQ ID NO: 61 and a ht chain variabe region comprising the anino acid sequence of SEQ DI) NO: 62 ra heav chain varwiabl rain conprisny sthe amino a"cid seune fSQI,. NA: 15 and a hlh chin varibl regon compriin the amino acid seqc~e of SE I D NO:~i 62; ori a heavy chain varaiabl region coi~risiJne the amino acidA sequence of' SFlQ I E NO:j 79) and a light chanK variable region cmpisn the am-ino ac id sequej~lnce ofSEQ ID NO: 80 or a heavy chain variable region comprising the amino acid 41 sequence ofSEQ ID NO: 5 a light chain variable region comprising the anino acid sequence oSQ NO: 9 or a heavychainariablregion comprising the anino acid sequence of SEQ ID NO: 95 and a light chain variable region comprising the amino acid sequence of' SEQ I) NO: 103 or a heavy chain variable region comprising he amino acidsequence of SEQ ID NO 117 and a light chain variable region comprising the amnno acid sequence of S EQ ID NC: 118: or a heavy chain variable region comprising the amino acidsequene of EQ ID NO: 131 and a light chain variable region comprisingthe amino acid sequence of SEQ ID NO: 132: or a heavy chain variable region comprising the amino acid sequence of SEQ I) NO: 145 and a light chain variable region comprising the amino acid sequence of SEQ ID NO: 146; or a heavy chai variable region. comprising the anmo acid sequenc-e of'SEQ' R1) NO: 151 and a light chain variable region comprising the amino acid sequence of SEQ ID NO: 146; or a heavy chain variableregion comprising the arino acid sequence of SQ iD NO: 165 and a light chain variable region comprising the amino acid sequence S EQID NO 166; or a heavy hain vriable region comprisi the amino acid sequence of SEQ ID NO: 181 and a light chain variable region compnsing the amino acid sequence of SEQ) N: 12: or a heavy chain varable region comprising the amio acid sequence of SEQ ID NO: 195 and a lighthain variable region comprising the amino acid sequence of SEQ it) N : 196 I5 The composition of any one of claims I to 4. wherein the antibodies are selected from the gmup consisting of H IDNl, HAMBIDV2, HMB-4)V3, HMB-DV4 lIMBDv5, IMBPV6, HIMB-DV7. HMB- V8 HMWDV9, HMIVDVIO, NBaDV 1, l 1MB Dv 12. iIMIVDV 13 and HMB-DV14, 16. The pharmaceutical composition of any one of' caims 1 to 15, further comrisinga pharmaceutically acceptable di mentor carrier and, optionalIy, an agent useful for extending the half life of the antibodies or antigen binding'agients thereof 17. A. method ofinhibiting or preventing, or method of treating. a dengue iu i fect ion or a denguevus-related disease comprisig the stepsof administering to a sub ect 42 in need thereof, a therapeutically or prophylactically effective amount of the composition of any one of claims I to 16. 18. A method of treatment of dengue virus infection or a dengue virus-related disease comprising the steps of identifying a patient in need of such treatment and administering to said patient a therapeutically effective amount of the pharmaceutical composition of any one of claims I to 16. 9 The method of claim 17 or 18, additionally comprising the administration of a further therapeutic agent. 20. The method of claim 19, wherein said further therapeutIc agent is an antiwiral agent 21. Use of the pharmaceutical composition of any one of claims I to 16 (i) in the manufacture of a medicament for the treatment of dengue virus infection, (ii) in a vaccine, or (iii) in diagnosis of dengue virus infection. KA4 431
AU2013202434A 2008-10-13 2013-04-04 Dengue virus neutralizing antibodies and uses thereof Active AU2013202434B2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU2013202434A AU2013202434B2 (en) 2008-10-13 2013-04-04 Dengue virus neutralizing antibodies and uses thereof

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US61/104,911 2008-10-13
AU2009305113A AU2009305113B2 (en) 2008-10-13 2009-10-13 Dengue virus neutralizing antibodies and uses thereof
AU2013202434A AU2013202434B2 (en) 2008-10-13 2013-04-04 Dengue virus neutralizing antibodies and uses thereof

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
AU2009305113A Division AU2009305113B2 (en) 2008-10-13 2009-10-13 Dengue virus neutralizing antibodies and uses thereof

Publications (2)

Publication Number Publication Date
AU2013202434A1 AU2013202434A1 (en) 2013-05-02
AU2013202434B2 true AU2013202434B2 (en) 2016-01-28

Family

ID=48366617

Family Applications (1)

Application Number Title Priority Date Filing Date
AU2013202434A Active AU2013202434B2 (en) 2008-10-13 2013-04-04 Dengue virus neutralizing antibodies and uses thereof

Country Status (1)

Country Link
AU (1) AU2013202434B2 (en)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004067567A2 (en) * 2003-01-31 2004-08-12 Novartis Ag Anti-dengue virus antibodies, compositions, methods and uses
WO2005056600A2 (en) * 2003-12-08 2005-06-23 The Government Of The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Monoclonal antibodies that bind or neutralize dengue virus

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004067567A2 (en) * 2003-01-31 2004-08-12 Novartis Ag Anti-dengue virus antibodies, compositions, methods and uses
WO2005056600A2 (en) * 2003-12-08 2005-06-23 The Government Of The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Monoclonal antibodies that bind or neutralize dengue virus

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
CHAMBERS T.J. et al., Journal of Virology, 2003, Vol. 77, pages 3655-3668 *
GONCALVEZ A. P. et al., PNAS, 2007, Vol. 104, pages 9422-9427 *
PUTTIKHUNT C. et al., Journal of Medical Virology, 2008, Vol. 80, pages 125-133 *

Also Published As

Publication number Publication date
AU2013202434A1 (en) 2013-05-02

Similar Documents

Publication Publication Date Title
CA2739551C (en) Dengue virus neutralizing antibodies and uses thereof
JP5710123B2 (en) Human cytomegalovirus neutralizing antibody and use thereof
KR101732056B1 (en) Neutralizing anti-influenza a virus antibodies and uses thereof
JP2021054859A (en) Antibody therapeutics that bind cd47
DK2734545T3 (en) NEUTRALIZING ANTI-INFLUENZA A VIRUS ANTIBODIES AND APPLICATIONS THEREOF
CN111333723B (en) Monoclonal antibody aiming at rabies virus G protein and application thereof
CN113773385A (en) Antibodies to bradykinin B1 receptor ligands
EP3220947B1 (en) Antibodies that potently neutralize rabies virus and other lyssaviruses and uses thereof
JP2014526886A (en) Antibodies cross-reactive with macrophage migration inhibitory factor (MIF) and D-dopachrome tomerase (D-DT)
CA3130805A1 (en) Antibodies binding to plasmodium circumsporozoite protein and uses thereof
AU2013202434B2 (en) Dengue virus neutralizing antibodies and uses thereof
EP4257195A2 (en) Anti-cfae antibodies and methods of use
CN116789827A (en) Methods of treating diseases using TIM-3 binding antibodies
CN114437215A (en) Anti-human CD38 antibody and preparation method and application thereof

Legal Events

Date Code Title Description
MK4 Application lapsed section 142(2)(d) - no continuation fee paid for the application
NA Applications received for extensions of time, section 223

Free format text: AN APPLICATION TO EXTEND THE TIME FROM 13 OCT 2013 TO 13 MAY 2014 IN WHICH TO PAY A CONTINUATION FEE HAS BEEN FILED .

NB Applications allowed - extensions of time section 223(2)

Free format text: THE TIME IN WHICH TO PAY A CONTINUATION FEE HAS BEEN EXTENDED TO 13 MAY 2014 .

FGA Letters patent sealed or granted (standard patent)