COMPOSITIONS AND METHODS FOR TREATING AND PREVENTING INFLUENZA

Information

  • Patent Application
  • 20200231657
  • Publication Number
    20200231657
  • Date Filed
    November 05, 2019
    4 years ago
  • Date Published
    July 23, 2020
    3 years ago
Abstract
This disclosure relates to peptide agents, e.g., antibodies and antigen-binding fragments thereof, that bind hemagglutinin protein of influenza viruses, and methods of their use.
Description
SEQUENCE LISTING

The instant application contains a Sequence Listing which has been filed electronically in ASCII format and is hereby incorporated by reference in its entirety. Said ASCII copy, created on Nov. 2, 2016, is named P2029-700910_SL.txt and is 186,427 bytes in size.


BACKGROUND

Influenza is an infectious disease caused by RNA viruses of the family Orthomyxoviridae (the influenza viruses). Influenza viruses are classified based on core protein into three genera A, B and C that are further divided into subtypes determined by the viral envelope glycoproteins haemagglutinin (HA) and neuraminidase (NA). Influenza A viruses infect a range of mammalian and avian species, whereas type B and C infections are largely restricted to humans. Only types A and B cause human disease of any concern.


High mutation rates and frequent genetic reassortments of the influenza viruses contribute to great variability of the HA and NA antigens. Minor point mutations causing small changes (“antigenic drift”) occur relatively often. Antigenic drift enables the virus to evade immune recognition, resulting in repeated influenza outbreaks during interpandemic years. Major changes in the HA antigen (“antigenic shift”) are caused by reassortment of genetic material from different influenza A subtypes. Antigenic shifts resulting in new pandemic strains are rare events, occurring through reassortment between animal and human subtypes, for example in co-infected pigs.


Influenza A spreads around the world in seasonal epidemics, resulting in the deaths of between 250,000 and 500,000 people every year, and up to millions in some pandemic years. On average 41,400 people died each year in the United States between 1979 and 2001 from influenza.


SUMMARY

The disclosure is based, at least in part, on the discovery of human anti-HA antibodies comprising functional and structural properties disclosed herein, e.g., antibodies that bind a conserved region or epitope on influenza virus, and uses thereof.


Accordingly, the disclosure features binding agents, e.g., antibody molecules, or preparations, or isolated preparations thereof, that bind hemagglutinin (HA) from influenza viruses. In an embodiment, a binding agent, e.g., an antibody molecule, is broad spectrum, and binds more than one HA, e.g., an HA from one or both of Group 1 or Group 2 strains of influenza A viruses and/or one or more strains of influenza B viruses. Therefore, in some embodiments, a binding agent, e.g., an antibody molecule, featured in the disclosure can treat or prevent infection by a Group 1 influenza virus and a Group 2 influenza virus. In other embodiments, a binding agent, e.g., an antibody molecule, featured in the disclosure can treat or prevent infection by an influenza A virus and/or an influenza B virus. The binding agents, e.g., antibody molecules, share sufficient structural similarity with antibodies or variable regions disclosed herein such that they possess functional attributes of the antibodies disclosed herein. In some embodiments, the structural similarity can be in terms of three dimensional structure, or linear amino acid sequence, or both.


In an aspect, the disclosure features a method of treating a subject, e.g., a subject having influenza or at risk for influenza, the method comprising administering, or causing to be administered, to the subject an amount of an anti-HA antibody molecule described herein, e.g., Ab 044 (also known as VIS410 herein), of between 2 and 30 mg/kg, thereby treating the subject.


In an embodiment, the subject is treated for influenza, or a disorder associated with influenza.


In an embodiment, the treatment comprises preventing the subject from influenza, or a disorder associated with influenza.


In an embodiment, the amount of the antibody molecule is between 5 and 25 mg/kg, between 10 and 20 mg/kg, between 12 and 18 mg/kg, between 14 and 16 mg/kg, between 13 and 18 mg/kg, between 8 and 16 mg/kg, between 13 and 16 mg/kg, between 11 and 16 mg/kg, between 10 and 15 mg/kg, between 11 and 15 mg/kg, between 8 and 12 mg/kg, or between 10 and 12 mg/kg.


In an embodiment, the amount of the antibody molecule is between 14.5 and 30 mg/kg, between 14.5 and 25 mg/kg, between 14.5 and 20 mg/kg, between 14.5 and 18 mg/kg, between 14.5 and 16 mg/kg; or between 14.5 and 15.5 mg/kg.


In an embodiment, the amount of the antibody molecule is between 15 and 30 mg/kg, between 15 and 25 mg/kg, between 15 and 20 mg/kg, between 15 and 18 mg/kg, between 15 and 16 mg/kg, or between 15 and 15.5 mg/kg.


In an embodiment, the amount of the antibody molecule is between 9 and 14 mg/kg, between 9 and 13 mg/kg, between 9 and 12 mg/kg, between 9 and 11 mg/kg, between 9 and 10 mg/kg, between 10 and 14 mg/kg, between 11 and 14 mg/kg, between 12 and 14 mg/kg, between 13 and 14 mg/kg, between 10 and 13 mg/kg, between 11 and 12 mg/kg, between 10 and 12 mg/kg, or between 10 and 11 mg/kg.


In an embodiment, the amount of the antibody molecule is 10 mg/kg, 11 mg/kg, 12 mg/kg, 13 mg/kg, 14 mg/kg, or 15 mg/kg. In an embodiment, the amount of the antibody molecule is 15 mg/kg. In an embodiment, the amount of the antibody is 10 mg/kg.


In an embodiment, the subject is administered a single dose of the antibody molecule. In an embodiment, the subject is administered a flat dose of the antibody molecule. In an embodiment, the amount of the antibody molecule administered is between 500 mg and 3000 mg, e.g., between 1000 mg and 3000 mg, between 1500 mg and 3000 mg, between 2000 mg and 3000 mg, between 1800 mg and 2500 mg, between 2500 mg and 3000 mg, between 500 mg and 2500 mg, between 500 mg and 2000 mg, between 500 mg and 1500 mg, between 500 mg and 1000 mg, between 1000 mg and 2500 mg, between 1500 mg and 2000 mg, or between 2000 mg and 2500 mg, e.g., 1500 mg, 1600 mg, 1700 mg, 1800 mg, 1900 mg, 2000 mg, 2100 mg, 2200 mg, 2300 mg, 2400 mg, or 2500 mg.


In an embodiment, the antibody molecule is administered intravenously. In an embodiment, the antibody molecule is administered intravenously over a period of 1-3 hours, e.g., 1-2 hours or 2-3 hours, e.g., 2 hours. In an embodiment, the subject is administered intravenously at a flat dose (e.g., a single flat dose) between 2000 mg and 2500 mg, e.g., between 2200 mg and 2400 mg, e.g., 2300 mg.


In an embodiment, the subject is infected, or is at risk of being infected, with an influenza virus chose from an H1N1 virus, an H3N2 virus, an H7N9 virus, or a combination thereof.


In an embodiment, the antibody molecule does not cause an antibody dependent enhancement (ADE) in the subject, e.g., as determined by a method described herein. In an embodiment, the antibody molecule is administered in an amount that does not cause an ADE in the subject, e.g., as determined by a method described herein.


In an embodiment, the antibody molecule does not cause viral resistance, e.g., as determined by a method described herein. In an embodiment, the antibody molecule is administered in an amount that does not cause viral resistance, e.g., as determined by a method described herein.


In an embodiment, the method further comprises detecting an anti-drug antibody (ADA) (e.g., an antibody that binds to or inhibits the anti-HA antibody molecule described herein) in a sample from the subject. In an embodiment, the antibody molecule is administered to a subject who has not developed, or has not been detected for having, an ADA to the antibody molecule.


In an embodiment, treating comprises preventing infection (e.g., influenza virus infection). In an embodiment, the influenza is a seasonal influenza.


In an embodiment, the method comprises administering the antibody molecule prior to the date, e.g., a day or range of days, of an epidemic peak of influenza or a disorder associated with influenza, e.g., wherein the date of the epidemic peak is an expected date for the epidemic peak determined prior to the occurrence of the epidemic peak.


In an embodiment, the epidemic peak is in a region that includes: the place (e.g., street address) where the subject lives; or the city, province or state, in which the subject lives.


In an embodiment, the antibody molecule is administered to a subject 1 to 15 weeks prior to the date of an epidemic peak; 2 to 10 weeks prior to the date of an epidemic peak; 3 to 8 weeks prior to the date of an epidemic peak; or 4 to 6 weeks prior to the date of an epidemic peak. In an embodiment, the antibody molecule is administered to a subject 4 to 8 weeks prior to the date of an epidemic peak.


In an embodiment, the subject is between 0 and 15 years of age; between 16 and 49 years of age; between 50 and 64 years of age; or 65 years of age or above. In another embodiment, the subject is at least 30, 40, 50, 60, or 65 years of age.


In an embodiment, the subject resides in a single family residence; a residence, e.g., single family residence, with at least 1 or 2 persons at least 65 years old; an institution, e.g., a retirement facility, assisted living facility, a hospital, nursing home; or an institution in which more than 2, 3, 5, 10, 20 or 30 unrelated people, e.g., people at least 65 years of age, reside.


In an embodiment, administering comprises an intravenous infusion. In an embodiment, administering includes a single intravenous infusion. In an embodiment, administering includes an intravenous infusion over at least 20, 30, 40, 50, 60, 90, or 120 minutes.


In an embodiment, the amount of the antibody molecule administered is between 10 and 15 mg/kg; the subject is over 65 years of age; and the antibody molecule is administered to the subject 1 to 15 weeks (e.g., 4 to 8 weeks) prior to the expected date of an epidemic peak in a region where the subject resides.


In an embodiment, the amount of the antibody molecule administered is between 14.5 and 15.5 mg/kg; the subject is over 65 years of age; and the antibody molecule is administered to the subject 1 to 15 weeks (e.g., 4 to 8 weeks) prior to the expected date of an epidemic peak in a region where the subject resides.


In an embodiment, the antibody molecule comprises:


(a) a heavy chain immunoglobulin variable region segment comprising: a CDR1 comprising the sequence S-Y-A-M-H (SEQ ID NO:68); a CDR2 comprising the sequence V-V-S-Y-D-G-N-Y-K-Y-Y-A-D-S-V-Q-G (SEQ ID NO:69); and a CDR3 comprising the sequence D-S-R-L-R-S-L-L-Y-F-E-W-L-S-Q-G-Y-F-N-P (SEQ ID NO:70); and


(b) a light chain immunoglobulin variable region segment comprising: a CDR1 comprising the sequence Q-S-I-T-F-D-Y-K-N-Y-L-A (SEQ ID NO: 145); a CDR2 comprising the sequence W-G-S-Y-L-E-S (SEQ ID NO:72); and a CDR3 comprising the sequence Q-Q-H-Y-R-T-P-P-S (SEQ ID NO:73).


In an embodiment, the antibody molecule comprises a heavy chain immunoglobulin variable region segment that comprises SEQ ID NO: 25. In an embodiment, the antibody molecule comprises a light chain immunoglobulin variable region segment that comprises SEQ ID NO: 52. In an embodiment, the antibody molecule comprises: a heavy chain immunoglobulin variable region segment that comprises SEQ ID NO: 25 and a light chain immunoglobulin variable region segment that comprises SEQ ID NO: 52. In an embodiment, the antibody molecule comprises a tetramer of: two heavy chain immunoglobulin variable region segments, each comprising SEQ ID NO: 25 and two light chain immunoglobulin variable region segments, each comprising SEQ ID NO: 52.


In an embodiment, the antibody molecule comprises a full length antibody. In an embodiment, the antibody molecule comprises a humanized antibody molecule. In an embodiment, the antibody molecule comprises two heavy claim variable regions and two light chain variable regions. In an embodiment, the antibody molecule is an IgG antibody. In an embodiment, the antibody molecule is a single chain antibody (scFv), a F(ab′)2 fragment, a Fab fragment, or an Fd fragment.


In an embodiment, the method further comprises administering to the subject a second therapeutic agent, e.g., for influenza, or a disorder or symptom associated with influenza.


In an aspect, the disclosure features a method of protecting a population of subjects, e.g., from influenza or a disorder associated with influenza, comprising administering, and/or causing to be administered, an anti-HA antibody molecule described herein, e.g., Ab 044, to at least 2%, at least 4%, at least 6%, at least 8%, or at least 10% of the subjects in the population, thereby protecting the population.


In an embodiment, protection comprises, decreasing, in the population, one or more (e.g., two, three or all) of: the number of hospital admissions, e.g. of influenza infected individuals; the number incidents of influenza infection; the attack rate; or the number of deaths, e.g. of influenza infected individuals.


In an embodiment, the antibody molecule is administered to at least 2%, but not more than 5 or 10% of the subjects in the population. In another embodiment, the antibody molecule is administered to at least 4%, but nor more than 8 or 15% of the subjects of the population.


In an embodiment, the method decreases, in the population, one or more (e.g., two, three or all) of: the number of hospital admissions, e.g. of influenza infected individuals; the number incidents of influenza infection; the attack rate; or the number of deaths, e.g. of influenza infected individuals.


In an embodiment, (a) the percentage decrease in the number of hospital admissions, incidents of influenza infection, attack rate, or deaths, for the population, is greater than (b) the percentage of subjects in the population receiving the anti-HA antibody molecule. In an embodiment, (a) is at least 2, 3, 4, or 5 times greater than (b).


In an embodiment, the population is all the subjects present in a predefined area. In an embodiment, the population is all the subjects having a predefined characteristic, e.g., being at least 65 years of age, present in a predefined area. In an embodiment, the predefined area is or comprises a city, state, province or other political geographic area. In an embodiment, the predefined area is or comprises an area having a predefined number of subjects. In an embodiment, the predefined area is or comprises an area within a preselected distance of a preselected place or landmark.


In an embodiment, the method comprises administering, and/or causing to be administered, an amount of the antibody molecule of between 2 and 30 mg/kg,


In an embodiment, the amount of the amount of the antibody molecule is between 5 and 25 mg/kg, between 10 and 20 mg/kg, between 12 and 18 mg/kg, between 14 and 16 mg/kg, between 13 and 18 mg/kg, between 8 and 16 mg/kg, between 11 and 16 mg/kg, between 13 and 16 mg/kg, between 10 and 15 mg/kg, between 11 and 15 mg/kg, between 8 and 12 mg/kg, or between 10 and 12 mg/kg.


In an embodiment, the amount of the antibody molecule is between 14.5 and 30 mg/kg, between 14.5 and 25 mg/kg, between 14.5 and 20 mg/kg, between 14.5 and 18 mg/kg, between 14.5 and 16 mg/kg; or between 14.5 and 15.5 mg/kg.


In an embodiment, the amount of the antibody molecule is between 15 and 30 mg/kg, between 15 and 25 mg/kg, between 15 and 20 mg/kg, between 15 and 18 mg/kg, between 15 and 16 mg/kg, or between 15 and 15.5 mg/kg.


In an embodiment, the amount of the antibody molecule is between 9 and 14 mg/kg, between 9 and 13 mg/kg, between 9 and 12 mg/kg, between 9 and 11 mg/kg, between 9 and 10 mg/kg, between 10 and 14 mg/kg, between 11 and 14 mg/kg, between 12 and 14 mg/kg, between 13 and 14 mg/kg, between 10 and 13 mg/kg, between 11 and 12 mg/kg, between 10 and 12 mg/kg, or between 10 and 11 mg/kg.


In an embodiment, the amount of the antibody molecule is 10 mg/kg, 11 mg/kg, 12 mg/kg, 13 mg/kg, 14 mg/kg, or 15 mg/kg. In an embodiment, the amount of the antibody molecule is 15 mg/kg. In an embodiment, the amount of the antibody is 10 mg/kg.


In an embodiment, the subject is at risk for influenza, e.g., seasonal influenza.


In an embodiment, the method comprises administering the antibody molecule prior to the date, e.g., a day or range of days, of an epidemic peak of influenza or a disorder associated with influenza, e.g., wherein the date of the epidemic peak is an expected date for the epidemic peak determined prior to the occurrence of the epidemic peak.


In an embodiment, the epidemic peak is in a region that includes: the place (e.g., street address) where the subject lives; or the city, province or state, in which the subject lives.


In an embodiment, the antibody molecule is administered, and/or causing to be administered, to a subject 1 to 15 weeks prior to the date of an epidemic peak; 2 to 10 weeks prior to the date of an epidemic peak; 3 to 8 weeks prior to the date of an epidemic peak; or 4 to 6 weeks prior to the date of an epidemic peak. In an embodiment, the antibody molecule is administered to a subject 4 to 8 weeks prior to the date of an epidemic peak.


In an embodiment, the subject is between 0 and 15 years of age; between 16 and 49 years of age; between 50 and 64 years of age; or 65 years of age or above. In another embodiment, the subject is at least 30, 40, 50, 55, 60, or 65 years of age. In an embodiment, the average age of the subjects in the population is at least 30, 40, 50, 55, 60, or 65.


In an embodiment, the subject resides in a single family residence; a residence, e.g., a single family residence, with at least 1 or 2 persons at least 65 years old; an institution, e.g., a retirement facility, assisted living facility, a hospital, nursing home; or an institution in which more than 2, 3, 5, 10, 20 or 30 unrelated people, e.g., people at least 65 years of age, reside.


In an embodiment, administering comprises an intravenous infusion. In an embodiment, administering includes a single intravenous infusion. In an embodiment, administering includes an intravenous infusion over at least 20, 30, 40, 50, 60, 90, or 120 minutes.


In an embodiment, the amount of the antibody molecule administered is between 10 and 15 mg/kg; the subject is over 65 years of age; and the antibody molecule is administered to the subject 1 to 15 weeks (e.g., 4 to 8 weeks) prior to the expected date of an epidemic peak in a region where the subject resides.


In an embodiment, the amount of the antibody molecule administered is between 14.5 and 15.5 mg/kg; the subject is over 65 years of age; and the antibody molecule is administered to the subject 1 to 15 weeks (e.g., 4 to 8 weeks) prior to the expected date of an epidemic peak in a region where the subject resides.


In an embodiment, the antibody molecule comprises:


(a) a heavy chain immunoglobulin variable region segment comprising: a CDR1 comprising the sequence S-Y-A-M-H (SEQ ID NO:68); a CDR2 comprising the sequence V-V-S-Y-D-G-N-Y-K-Y-Y-A-D-S-V-Q-G (SEQ ID NO:69); and a CDR3 comprising the sequence D-S-R-L-R-S-L-L-Y-F-E-W-L-S-Q-G-Y-F-N-P (SEQ ID NO:70); and


(b) a light chain immunoglobulin variable region segment comprising: a CDR1 comprising the sequence Q-S-I-T-F-D-Y-K-N-Y-L-A (SEQ ID NO: 145); a CDR2 comprising the sequence W-G-S-Y-L-E-S (SEQ ID NO:72); and a CDR3 comprising the sequence Q-Q-H-Y-R-T-P-P-S (SEQ ID NO:73).


In an embodiment, the antibody molecule comprises a heavy chain immunoglobulin variable region segment that comprises SEQ ID NO: 25. In an embodiment, the antibody molecule comprises a light chain immunoglobulin variable region segment that comprises SEQ ID NO: 52. In an embodiment, the antibody molecule comprises: a heavy chain immunoglobulin variable region segment that comprises SEQ ID NO: 25 and a light chain immunoglobulin variable region segment that comprises SEQ ID NO: 52. In an embodiment, the antibody molecule comprises a tetramer of: two heavy chain immunoglobulin variable region segments, each comprising SEQ ID NO: 25 and two light chain immunoglobulin variable region segments, each comprising SEQ ID NO: 52.


In an embodiment, the antibody molecule comprises a full length antibody. In an embodiment, the antibody molecule comprises a humanized antibody molecule. In an embodiment, the antibody molecule comprises two heavy claim variable regions and two light chain variable regions. In an embodiment, the antibody molecule is an IgG antibody. In an embodiment, the antibody molecule is a single chain antibody (scFv), a F(ab′)2 fragment, a Fab fragment, or an Fd fragment.


In an embodiment, the method further comprises administering to the subject a second therapeutic agent, e.g., for influenza, or a disorder or symptom associated with influenza.


In an aspect, the disclosure features an anti-HA antibody molecule described herein, e.g., Ab 044, of between 2 and 30 mg/kg, for use in a method of treating a subject, e.g., a subject having influenza or at risk for influenza. In an embodiment, the subject is treated for influenza or a disorder associated with influenza. In an embodiment, the treatment comprises preventing the subject from influenza or a disorder associated with influenza.


In an embodiment, the method comprises administering the anti-HA antibody to the subject in an amount between 10 and 15 mg/kg 1 to 15 weeks prior to the expected date of an epidemic peak of influenza (or a disorder associated with influenza) in a region where the subject resides. In another embodiment, the method comprises administering to the subject an anti-HA antibody molecule in an amount between 11 and 16 mg/kg.


In another aspect, the disclosure features an anti-HA antibody molecule described herein, e.g., Ab 044, for use in a method of protecting a population of subjects, e.g., from influenza or a disorder associated with influenza, wherein the anti-HA antibody molecule is used in at least 2%, at least 4%, at least 6%, at least 8%, or at least 10% of the subjects in the population.


In one aspect, the disclosure features an anti-hemagglutinin (anti-HA) binding agent, e.g., a specific binding agent, e.g., an antibody molecule, or preparation, or isolated preparation thereof, comprising one or more or all of the following properties:


(a) it fails to produce any escape mutants as determined by the failure of a viral titer to recover following at least 10, 9, 8, 7, 6, or 5 rounds of serial infections in cell culture with a mixture of the antibody molecule and an influenza A virus, e.g., a Group 1 strain, e.g., an H1N1 strain, e.g., A/South Carolina/1/1918, A/Puerto Rico/08/1934, or A/California/04/2009, or an H5N1 strain, e.g., A/Indonesia/5/2005 or A/Vietnam/1203/2004;


(b) it produces fewer escape mutants than does a reference anti-HA antibody molecule, e.g., Ab 67-11, FI6, FI28, C179, F10, CR9114, or CR6261, e.g., when tested by the method described in (a);


(c) it prevents infection by at least 1, 2, 3, 4 or 5 influenza subtypes of Group 1, and by at least 1, 2, 3, 4 or 5 influenza subtypes of Group 2;


(d) it inhibits fusogenic activity of the targeted HA;


(e) it treats or prevents infection by a Group 1 virus, such as where the virus is an H1, H5, or H9 virus; and it treats or prevents infection by a Group 2 virus, such as where the virus is an H3 or H7 virus;


(f) it treats or prevents infection by influenza A strains H1N1 and H3N2;


(g) it is effective for prevention or treatment of infection, e.g., in humans or mice, with H1N1 and H3N2 when administered at 50 mg/kg, 25 mg/kg, 10 mg/kg, 6 mg/kg, 5 mg/kg, 4 mg/kg, 3 mg/kg, 2 mg/kg, or 1 mg/kg;


(h) it treats or prevents infection by influenza A H5N1 strains;


(i) it is effective for prevention or treatment of infection, e.g., in humans or mice, with H5N1 when administered at 50 mg/kg, 25 mg/kg, 10 mg/kg, 6 mg/kg, 5 mg/kg, 4 mg/kg, 3 mg/kg, 2 mg/kg, or 1 mg/kg;


(j) the concentration of antibody molecule required for 50% neutralization of influenza A virus is less than 10 μg/mL;


(k) it treats or prevents infection by an influenza B virus, e.g., B/Wisconsin/1/2010;


(l) it is effective for prevention or treatment of infection, e.g., in humans or mice, with an influenza B virus, e.g., B/Wisconsin/1/2010, when administered at 10 mg/kg, 6 mg/kg, 4 mg/kg, 3 mg/kg, 2 mg/kg, or 1 mg/kg;


(m) the concentration of antibody molecule required for 50% neutralization of influenza B virus, e.g., B/Wisconsin/1/2010, virus is less than 10 μg/mL;


(n) it prevents or minimizes secondary infection (e.g., secondary bacterial infection) or effects thereof on a subject;


(o) it is effective for preventing or minimizing secondary infection (e.g., secondary bacterial infection) or effects thereof on a subject when administered at 50 mg/kg, 25 mg/kg, 10 mg/kg, 6 mg/kg, 5 mg/kg, 4 mg/kg, 3 mg/kg, 2 mg/kg, or 1 mg/kg;


(p) it binds an epitope which comprises or consists of the hemagglutinin trimer interface; and


(q) it binds an epitope other than that bound by a reference anti-HA antibody molecule, e.g., Ab 67-11, F16, F128, C179, F10, CR9114, or CR6261, e.g., as determined by structural analysis, e.g., by X-ray crystallography or NMR spectroscopy; or


(r) in an embodiment it binds to an epitope, e.g., it has an epitope that overlaps with or is the same as, of an antibody disclosed herein, e.g., as determined by mutational analysis or crystal structure analysis.


In one embodiment, the binding agent, e.g., an anti-HA antibody molecule, has one or more of the following characteristics: the anti-HA antibody molecule prevents infection by at least 1, 2, 3, 4 or 5 influenza subtypes of Group 1, and by at least 1, 2, 3, 4 or 5 influenza subtypes of Group 2; the concentration of the anti-HA antibody molecule required for 50% neutralization of influenza A virus is less than 10 μg/mL; or the anti-HA antibody molecule binds an epitope that comprises or consists of the hemagglutinin trimer interface.


In one embodiment, the binding agent, e.g., an anti-HA antibody molecule, featured in the disclosure treats or prevents infection by a Group 1 virus, such as where the virus is an H1, H2, H5, H6, H8, H9, H12, H11, H13, H16, or H17 virus; and treats or prevents infection by a Group 2 virus, such as where the virus is an H3, H4, H7, H10 or H15 virus. In one embodiment, the binding agent, e.g., an anti-HA antibody molecule, featured in the disclosure prevents infection by at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or 11 influenza subtypes of Group 1, and by at least 1, 2, 3, 4, 5 or 6 influenza subtypes of Group 2. In one embodiment, the binding agent, e.g., an anti-HA antibody molecule, featured in the disclosure treats or prevents infection by one or more of H1N1, H2N2, H5N1, and H9N2, and also treats or prevents infection by one or more of H3N2 and H7N7. In an embodiment, a binding agent, e.g., antibody molecule, binds, and in some embodiments, neutralizes: at least one strain from the Group 1 H1, e.g., H1a or H1b, cluster and at least one strain from the Group 2 H3 or H7 cluster. In an embodiment, a binding agent, e.g., antibody molecule, binds, and in some embodiments, neutralizes: at least one strain from the Group 1 H1, e.g., H1a or H1b, cluster and at least one influenza B strain, e.g., B/Wisconsin/1/2010. In an embodiment, a binding agent, e.g., antibody molecule, binds, and in certain embodiments, neutralizes: at least one strain from the Group 2 H3 or H7 cluster and at least one influenza B strain, e.g., B/Wisconsin/I/2010. In an embodiment, a binding agent, e.g., antibody molecule, binds, and in certain embodiments, neutralizes: at least one strain from the Group 1 H1, e.g., H1 a or H1b, cluster, at least one strain from the Group 2 H3 or H7 cluster, and at least one influenza B strain, e.g., B/Wisconsin/1/2010. In one embodiment, the binding agent, e.g., an anti-HA antibody molecule, featured in the disclosure treats or prevents infection by one or more of influenza B viruses, e.g., B/Wisconsin/1/2010.


In one embodiment, the anti-HA antibody molecule is not an anti-HA antibody molecule previously described in the art. For example, the anti-HA antibody molecule is other than one or more or all of Ab 67-11 (U.S. Provisional Application No. 61/645,453), FI6 (FI6, as used herein, refers to any specifically disclosed F16 sequence in U.S. Application Publication No. 2010/0080813, U.S. Application Publication No. 2011/0274702, International Publication No. WO2013/011347, or Corti et al., Science 333:850-856, 2011, published online Jul. 28, 2011; FIGS. 12A to 12C of International Publication No. WO2013/170139 or U.S. Application Publication No. 2013/0302349), FI28 (U.S. Application Publication No. 2010/0080813), C179 (Okuno et al., J. Virol. 67:2552-1558, 1993), F10 (Sui et al., Nat. Struct. Mol. Biol. 16:265, 2009), CR9114 (Dreyfus et al., Science. 2012; 337(6100): 1343-1348; published online Aug. 9, 2012), or CR6261 (Ekiert et al., Science 324:246-251, 2009; published online Feb. 26, 2009).


In one embodiment, the binding agent, e.g., an anti-HA antibody molecule, neutralizes infection with H1N1 and H3N2 in vitro. In another embodiment, binding agent, e.g., an anti-HA antibody molecule, neutralizes infection with H1N1 and H3N2 in vivo. In one embodiment, the binding agent, e.g., an anti-HA antibody molecule, neutralizes infection with H5N1 in vitro. In another embodiment, binding agent, e.g., an anti-HA antibody molecule, neutralizes infection with H5N1 in vivo. In one embodiment, the binding agent, e.g., an anti-HA antibody molecule, neutralizes infection with an influenza B virus, e.g., B/Wisconsin/1/2010, in vitro. In another embodiment, the binding agent, e.g., an anti-HA antibody molecule neutralizes infection with an influenza B virus, e.g., B/Wisconsin/1/2010, in vivo.


In another embodiment, the concentration of the binding agent, e.g., an anti-HA antibody molecule, required for 50% neutralization of influenza A virus is 10 μg/mL or less, such as 9 μg/mL or less, 8 μg/mL or less, 7 μg/mL or less, 6 μg/mL or less, or 5 μg/mL or less. In another embodiment, the concentration of the binding agent, e.g., an anti-HA antibody molecule, required for 60% neutralization of influenza A virus, 50% neutralization of influenza A virus, or 40% neutralization of influenza A virus is 10 μg/mL or less, such as 9 μg/mL or less, 8 μg/mL or less, 7 μg/mL or less, 6 μg/mL or less, or 5 μg/mL or less.


In yet another embodiment, the binding agent, e.g., an anti-HA antibody molecule, is effective for prevention or treatment of infection, e.g., in humans or mice, with H1N1 and H3N2, such as when administered at 50 mg/kg, 25 mg/kg, 10 mg/kg, 6.0 mg/kg, 5.0 mg/kg, 4.0 mg/kg, 3.0 mg/kg, 2.0 mg/kg, 1.0 mg/kg or less. In still another embodiment, the binding agent, e.g., the anti-HA antibody molecule, is effective for prevention or treatment of infection, e.g., in humans or mice, with H5N1, such as when administered at 50 mg/kg, 25 mg/kg, 10 mg/kg, 6.0 mg/kg, 5.0 mg/kg, 4.0 mg/kg, 3.0 mg/kg, 2.0 mg/kg, 1.0 mg/kg or less.


In another embodiment, a binding agent, e.g., an anti-HA antibody molecule, is effective for the treatment or prevention of a Group 1 virus, where the Group 1 virus is H1, H5, or H9, and in another embodiment, the binding agent, e.g., an anti-HA antibody molecule, is effective for the treatment or prevention of a Group 2 virus, where the Group 2 virus is H3 or H7. In another embodiment, the concentration of the binding agent, e.g., an anti-HA antibody molecule, required for 50% neutralization of influenza B virus, e.g., B/Wisconsin/1/2010, is 10 μg/mL or less, such as 9 μg/mL or less, 8 μg/mL or less, 7 μg/mL or less, 6 μg/mL or less, or 5 μg/mL or less. In another embodiment, the concentration of the binding agent, e.g., an anti-HA antibody molecule, required for 60% neutralization of influenza B virus, e.g., B/Wisconsin/1/2010, 50% neutralization of influenza B virus, e.g., B/Wisconsin/1/2010, or 40% neutralization of influenza B virus, e.g., B/Wisconsin/1/2010, is 10 μg/mL or less, such as 9 μg/mL or less, 8 μg/mL or less, 7 μg/mL or less, 6 μg/mL or less, or 5 μg/mL or less.


In another embodiment, the binding agent, e.g., an anti-HA antibody molecule, is a full length tetrameric antibody, a single chain antibody (scFv), a F(ab′)2 fragment, a Fab fragment, or an Fd fragment. In another embodiment, the heavy chain of the antibody molecule is a γ1 heavy chain, and in yet another embodiment, the light chain of the antibody molecule is a κ light chain or a λ light chain. In yet another embodiment, the anti-HA antibody molecule featured in the disclosure is an IgG1 antibody.


In an embodiment, the antibody molecule binds an epitope that has one, two, three, four, five, or all of, the following properties a)-f): a) it includes one, two, or all of, H3 HA1 residues N38, 1278, and D291; b) it includes H3 HA2 residue N12; c) it does not include one, two or all of, H3 HA1 residues Q327, T328, and R329; d) it does not include one, two, three, four, or all of, H3 HA2 residues G1, L2, F3, G4, and D46; e) it includes one, two, or all of, H3 HA1 residues T318, R321, and V323; or f) it includes 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, or all of, H3 HA2 residues A7, E11, I18, D19, G20, W21, L38, K39, T41, Q42, A43, I45, I48, N49, L52, N53, I56, and E57.


In an embodiment, the antibody molecule has properties: a) and b). In an embodiment, the antibody molecule has properties: c) and d). In an embodiment, the antibody molecule has properties: a); and c) or d). In an embodiment, the antibody molecule has properties: b); and c) or d). In an embodiment, the antibody molecule has properties: c); and a) or b). In an embodiment, the antibody molecule has properties: d); and a) or b). In an embodiment, the antibody molecule has properties: a), b), c) and d). In an embodiment, the antibody molecule has properties: a), b), c), d), e), and f).


In an embodiment, the antibody molecule has a KD for H3 of equal to or less than 10−6, wherein said KD is increased by at least 2, 5, 10, or 100 fold, by a mutation or mutations in any of: a) H3 HA1 residues N38, 1278, or D291; b) H3 HA2 residue N12; c) H3 HA1 residues T318, R321, or V323; or d) H3 HA2 residues A7, E11, I18, D19, G20, W21, L38, K39, T41, Q42, A43, I45, I48, N49, L52, N53, 156, or E57. In an embodiment, the antibody molecule has a KD for H3 of equal to or less than 10−6, wherein said KD is increased by no more than 2, or 5 fold, by a mutation or mutations in any of: c) H3 HA1 residues Q327, T328, or R329; or d) H3 HA2 residues G1, L2, F3, G4, or D46.


In an embodiment, the antibody molecule binds an epitope that has one, two, three, four, five, or all of, the following properties aa)-ff): aa) it includes one, two, or all of, H1 HA1 residues H31, N279, and S292; bb) it includes H1 HA2 residue G12; cc) it does not include one or both of H1 HA1 residues Q328 and S329; dd) it does not include one, two, three, four, or all of, H1 HA2 residues G1, L2, F3, G4, and D46; ee) it includes one, two, or all of, H1 HA1 residues T319, R322, and 1324 are bound by both Ab 044 and F16; or ff) it includes 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, or all of, H1 HA2 residues A7, E11, I18, D19, G20, W21, Q38, K39, T41, Q42, N43, I45, I48, T49, V52, N53, 156, and E57. In an embodiment, the antibody molecule has properties: aa) and bb). In an embodiment, the antibody molecule has properties: cc) and dd). In an embodiment, the antibody molecule has properties: aa); and cc) or dd). In an embodiment, the antibody molecule has properties: bb); and cc) or dd). In an embodiment, the antibody molecule has properties: cc); and aa) or bb). In an embodiment, the antibody molecule has properties: dd); and aa) or bb). In an embodiment, the antibody molecule has properties: aa), bb), cc) and dd). In an embodiment, the antibody molecule has properties: aa), bb), cc), dd), ee), and ff).


In an embodiment, the antibody molecule has a KD for H1 of equal to or less than 10−6, wherein said KD is increased by at least 2, 5, 10, or 100 fold, by a mutation or mutations in any of: aa) H1 HA1 residues H31, N279, and S292; bb) H1 HA2 residue G12; cc) H1 HA1 residues T319, R322, and 1324; or dd) H1 HA2 residues A7, E11, I18, D19, G20, W21, Q38, K39, T41, Q42, N43, I45, I48, T49, V52, N53, 156, and E57. In an embodiment, the antibody molecule has a KD for H1 of equal to or less than 10−6, wherein said KD is increased by no more than 2, or 5 fold, by a mutation or mutations in any of: cc) H1 HA1 residues Q328 and S329; or dd) H1 HA2 residues G1, L2, F3, G4, and D46. In an embodiment, the antibody molecule has one, two, three or all of the following properties: a) and aa); b) and bb); c) and cc); or d) and dd). In an embodiment, the molecule has properties c), cc), d), and dd).


In an embodiment, the binding agent, e.g., a specific binding agent, e.g., an antibody molecule, comprises one or both of: a heavy chain variable region comprising at least, or more than, 60, 65, 70, 75, 80, 85, 87, 90, 95, 98 or 99 percent homology with a heavy chain variable region from Table 3, Table 4A, or Table 4B, or FIG. 2, FIG. 13 or FIG. 17 of International Publication No. WO2013/170139 or U.S. Application Publication No. 2013/0302349; and a light chain variable region comprising at least, or more than, 60, 65, 70, 75, 80, 85, 87, 90, 95, 98 or 99 percent homology with light chain variable region from Table 3, Table 4A, or Table 4B, or FIG. 3, FIG. 14 or FIG. 17 International Publication No. WO2013/170139 or U.S. Application Publication No. 2013/0302349.


In an embodiment, the antibody molecule comprises a heavy chain variable region 25 (SEQ ID NO: 25), or a structurally or functionally related variable heavy chain region as described herein. In an embodiment, the antibody molecule comprises a light chain variable region 52 (SEQ ID NO: 52), 155 (SEQ ID NO: 155), or 45 (SEQ ID NO: 45), or a structurally or functionally related variable light chain region as described herein. In an embodiment, the antibody molecule comprises: a heavy chain variable region 25 (SEQ ID NO: 25), or a structurally or functionally related variable heavy chain region as described herein; and a light chain variable region 52 (SEQ ID NO: 52), 155 (SEQ ID NO: 155), or 45 (SEQ ID NO: 45), or a structurally or functionally related variable light chain region as described herein.


In an embodiment, the antibody molecule comprises a heavy chain variable region comprising one, two, or all of CDR1, CDR2, and CDR3, from heavy chain variable region 25 (SEQ ID NO: 25), or a structurally or functionally related variable heavy chain region as described herein. In an embodiment, the antibody molecule comprises a light chain variable region comprising one, two, or all of CDR1, CDR2, and CDR3, from light chain variable region 52 (SEQ ID NO: 52), 155 (SEQ ID NO: 155), or 45 (SEQ ID NO: 45), or a structurally or functionally related sequence as described herein. In an embodiment, the antibody molecule comprises: a heavy chain variable region comprising one, two, or all of CDR1, CDR2, and CDR3, from heavy chain variable region 25 (SEQ ID NO: 25), or a structurally or functionally related variable heavy chain region as described herein; and a light chain variable region comprising one, two, or all of CDR1, CDR2, and CDR3, from light chain variable region 52 (SEQ ID NO: 52), 155 (SEQ ID NO: 155), or 45 (SEQ ID NO: 45), or a structurally or functionally related variable light chain region as described herein.


In an embodiment, the antibody molecule comprises a heavy chain variable region from FIG. 2 or FIG. 13 of International Publication No. WO2013/170139 or U.S. Application Publication No. 2013/0302349 or a structurally or functionally related variable heavy chain region as described herein. In an embodiment, the antibody molecule comprises a light chain variable region from FIG. 3 or FIG. 14 of International Publication No. WO2013/170139 or U.S. Application Publication No. 2013/0302349, or a structurally or functionally related variable light chain region as described herein. In an embodiment, the antibody molecule comprises one, two, or all of, a CDR1, CDR2, and CDR3 from a heavy chain variable region from FIG. 2 or FIG. 13 International Publication No. WO2013/170139 or U.S. Application Publication No. 2013/0302349, or a structurally or functionally related sequence as described herein. In an embodiment, the antibody molecule comprises one, two, or all of, a CDR1, CDR2, and CDR3 from a light chain variable region from FIG. 3 or FIG. 14 International Publication No. WO2013/170139 or U.S. Application Publication No. 2013/0302349, or a structurally or functionally related sequence as described herein. In an embodiment, the antibody molecule comprises one, two or all of, HC CDR1, HC CDR2, and HC CDR3 and one, two or all of, LC CDR1, LC CDR2, and LC CDR3 from an antibody disclosed in Table 3, or a structurally or functionally related sequence as described herein.


In another embodiment, the antibody molecule comprises the light chain LC45 (SEQ ID NO: 45). In yet another embodiment, the antibody comprises the light chain LC45, and the heavy chain HC25 (SEQ ID NO: 25) or 24 (SEQ ID NO: 24). In one embodiment, the antibody molecule comprises the light chain Ab032 (SEQ ID NO: 45) and the heavy chain 25 (SEQ ID NO: 25). In yet another embodiment, the antibody molecule comprises light chain LC52 (SEQ ID NO: 52) and heavy chain HC25 (SEQ ID NO: 25).


In an embodiment, the antibody molecule comprises one or both of: a) one or more framework regions (FRs) from heavy chain disclosed herein. E.g., the antibody molecule comprises one or more or all of FR1, FR2, FR3, or FR4, or FR sequences that differ individually, or collectively, by no more than 1, 2, 3, 4, of 5 amino acid residues, e.g., conservative residues, from a heavy chain disclosed herein; and b) one or more framework regions (FRs) from light chain disclosed herein. E.g., the antibody molecule comprises one or more or all of FR1, FR2, FR3, or FR4, or FR sequences that differ individually, or collectively, by no more than 1, 2, 3, 4, of 5 amino acid residues, e.g., conservative residues, from light chain disclosed herein.


In one aspect, an anti-HA antibody molecule featured in the disclosure, or preparation, or isolated preparation thereof, comprises: (a) a heavy chain immunoglobulin variable domain comprising a sequence at least 60, 70, 80, 85, 87, 90, 95, 97, 98, or 99, e.g., 90%, homologous, to a heavy chain consensus sequence provided herein, e.g., the heavy chain consensus sequence provided in FIG. 2 or FIG. 13 of International Publication No. WO2013/170139 or U.S. Application Publication No. 2013/0302349, e.g., the heavy chain consensus sequence provided in FIG. 2 of International Publication No. WO2013/170139 or U.S. Application Publication No. 2013/0302349, SEQ ID NO: 161; and (b) a light chain immunoglobulin variable domain comprising a sequence at least 60, 70, 80, 85, 87, 90, 95, 97, 98, or 99, e.g., 95%, homologous, to a light chain consensus sequence provided herein, e.g., the light chain consensus sequence provided in FIG. 3 or FIG. 14 of International Publication No. WO2013/170139 or U.S. Application Publication No. 2013/0302349, e.g., the light chain consensus sequence provided in FIG. 3 of International Publication No. WO2013/170139 or U.S. Application Publication No. 2013/0302349, SEQ ID NO: 62.


For example, in one embodiment, the anti-HA antibody molecule featured in the disclosure comprises one or both of: (a) a heavy chain immunoglobulin variable domain comprising the sequence of SEQ ID NO: 161, or a sequence at least 87% identical to SEQ ID NO: 161; and (b) a light chain immunoglobulin variable domain comprising the sequence SEQ ID NO: 62, or a sequence at least 95% identical to SEQ ID NO: 62.


In another embodiment the antibody molecule comprises: (a) a heavy chain immunoglobulin variable domain comprising the sequence of SEQ ID NO: 161, or a sequence at least 87% identical to SEQ ID NO: 161; and (b) a light chain immunoglobulin variable domain comprising the sequence SEQ ID NO:62, or a sequence at least 95% identical to SEQ ID NO: 62, wherein said antibody molecule: (i) fails to produce any escape mutants as determined by the failure of a viral titer to recover following at least 10, 9, 8, 7, 6, or 5 rounds of serial infections in cell culture with a mixture of the antibody molecule and an influenza virus (e.g., an influenza A virus, e.g., a Group 1 strain, e.g., an H1N1 strain, e.g., A/South Carolina/1/1918, A/Puerto Rico/08/1934, or A/California/04/2009, or an H5N1 strain, e.g., A/Indonesia/5/2005 or A/Vietnam/1203/2004, or an influenza B virus, e.g., B/Wisconsin/1/2010); and (ii) produces fewer escape mutants than does a reference anti-HA antibody molecule, e.g., Ab 67-11, FI6, FI28, C179, F10, CR9114, or CR6261, such as when tested by the method described in (i).


In an embodiment, the disclosure features an antibody molecule comprising one or both of: (a) a heavy chain immunoglobulin variable region comprising the sequence of SEQ ID NO: 161, or a sequence that differs from SEQ ID NO:161 by not more than 1, 2, 3, 4, 5, 6, 8, 10, 11, 12, 13, 14, 15 or 16, e.g., by no more than 2, 3, 4, or 5 amino acids, e.g., conservative amino acids; and (b) a light chain immunoglobulin variable domain comprising the sequence SEQ ID NO:62, or a sequence that differs from SEQ ID NO:62 that differs by no more than 1, 2, 3, 4 or 5 amino acids, e.g., conservative amino acids.


In one embodiment, the 1, 2, 3, 4, 5, 6, 8, 10, 11, 12, 13, 14, 15 or 16 amino acid differences, e.g., conservative amino acid differences, in the heavy chain immunoglobulin variable region are in the FR regions of the heavy chain immunoglobulin variable domain. In another embodiment, the 1, 2, 3, 4 or 5 amino acid differences, e.g., conservative amino acid differences, in the light chain immunoglobulin variable domain are in the FR regions of the light chain immunoglobulin variable domain. In one embodiment, the amino acid differences in the heavy chain immunoglobulin variable region, or in the light chain immunoglobulin variable region, are conservative amino acid changes.


In an embodiment, the binding agent, e.g., an antibody molecule, binds to an epitope, e.g., it has an epitope that overlaps with or is the same as, of an antibody disclosed herein, e.g., as determined by mutational analysis or crystal structure analysis.


In an embodiment, the antibody molecule comprises one or both of: a) one or more framework regions (FRs) from heavy chain consensus sequence disclosed herein. e.g., the antibody molecule comprises one or more or all of FR1, FR2, FR3, or FR4, or sequences that differ individually, or collectively, by no more than 1, 2, 3, 4, of 5 amino acid residues, e.g., conservative residues, from heavy chain consensus sequence disclosed herein; and b) one or more framework regions (FRs) from light chain consensus sequence disclosed herein. e.g., the antibody molecule comprises one or more or all of FR1, FR2, FR3, or FR4, or sequences that differ individually, or collectively, by no more than 1, 2, 3, 4, of 5 amino acid residues, e.g., conservative residues, from light chain consensus disclosed herein. In an embodiment, the binding agent, e.g., an antibody molecule, specifically binds the HA antigen.


In another aspect, the disclosure features, a binding agent, e.g., an antibody molecule, or preparation, or isolated preparation thereof, comprising a structural or functional property of Ab 044.


In an embodiment, the antibody molecule competes with a reference antibody molecule, e.g., an antibody molecule described herein, for binding to a substrate, e.g., an HA. The reference antibody molecule can be: a) an antibody molecule comprising: i) a heavy chain immunoglobulin variable region segment comprising: a CDR1 comprising the sequence S-Y-A-M-H (SEQ ID NO:68); a CDR2 comprising the sequence V-V-S-Y-D-G-N-Y-K-Y-Y-A-D-S-V-Q-G (SEQ ID NO:69); and a CDR3 comprising the sequence D-S-R-L-R-S-L-L-Y-F-E-W-L-S-Q-G-Y-F-N-P (SEQ ID NO:70); and ii) a light chain variable region segment comprising: a CDR1 comprising the sequence Q-S-I-T-F-D-Y-K-N-Y-L-A (SEQ ID NO: 145); a CDR2 comprising the sequence W-G-S-Y-L-E-S (SEQ ID NO:72); and a CDR3 comprising the sequence Q-Q-H-Y-R-T-P-P-S (SEQ ID NO:73); b) an antibody molecule comprises one or both of: (i) a heavy chain immunoglobulin variable region segment comprising SEQ ID NO: 25; and (ii) a light chain variable region segment comprising SEQ ID NO:52; or c) Ab 044.


The HA can be from a Group 1 strain, e.g., an H1N1 strain, e.g., A/South Carolina/1/1918, A/Puerto Rico/08/1934, or A/California/04/2009, or an H5N1 strain, e.g., A/Indonesia/5/2005 or A/Vietnam/1203/2004. Competition between the antibody molecule and a reference antibody molecule can be determined by evaluating the ability of one of the antibody molecules or the reference antibody molecule to decrease binding of the other to a substrate, e.g., HA, e.g., HA1 or HA5, e.g. from an H1N1 strain, e.g., A/South Carolina/1/1918, A/Puerto Rico/08/1934, or A/California/04/2009, or an H5N1 strain, e.g., A/Indonesia/5/2005 or A/Vietnam/1203/2004. Reduction of the ability to bind can be evaluated by methods in the art. Reduction of the ability to bind can be evaluated, e.g., by one or more of: a) Biacore analysis; b) ELISA assay; and c) flow cytometry.


The antibody molecule can compete with the reference antibody such that binding of the reference antibody is decreased by 50% or more. In an embodiment, the antibody molecule binds to the same epitope, or a portion thereof, which the reference antibody molecule binds. In an embodiment, the antibody molecule does not bind to the same epitope, or a portion thereof, which the reference antibody molecule binds.


In an embodiment, the antibody molecule binds to the same epitope, or a portion thereof, on HA, as does a reference antibody molecule, e.g. an antibody molecule disclosed herein. The reference antibody molecule can be: a) an antibody molecule comprising: i) a heavy chain immunoglobulin variable region segment comprising: a CDR1 comprising the sequence S-Y-A-M-H (SEQ ID NO:68); a CDR2 comprising the sequence V-V-S-Y-D-G-N-Y-K-Y-Y-A-D-S-V-Q-G (SEQ ID NO:69); and a CDR3 comprising the sequence D-S-R-L-R-S-L-L-Y-F-E-W-L-S-Q-G-Y-F-N-P (SEQ ID NO:70); and ii) a light chain variable region segment comprising: a CDR1 comprising the sequence Q-S-I-T-F-D-Y-K-N-Y-L-A (SEQ ID NO: 145); a CDR2 comprising the sequence W-G-S-Y-L-E-S (SEQ ID NO:72); and a CDR3 comprising the sequence Q-Q-H-Y-R-T-P-P-S (SEQ ID NO:73); b) an antibody molecule comprises one or both of: (i) a heavy chain immunoglobulin variable region segment comprising SEQ ID NO: 25; and (ii) a light chain variable region segment comprising SEQ ID NO:52; or c) Ab 044.


The HA can be HA1 or HA5, e.g., from an H1N1 strain, e.g., A/South Carolina/1/1918, A/Puerto Rico/08/1934, or A/California/04/2009, or an H5N1 strain, e.g., A/Indonesia/5/2005 or A/Vietnam/1203/2004Binding to the same epitope, or a portion thereof, can be shown by one or more of: a) mutational analysis, e.g., binding to HA, or binding affinity for HA, is decreased or abolished if a residue is mutated; b) analysis, e.g., comparison, of the crystal structure of the antibody molecule and HA and the crystal structure of a reference antibody and HA, e.g., to determine the touch points of each; c) competition of the two antibodies for binding to HA, e.g., HA1 or HA5, from, e.g., an H1N1 strain, e.g., A/South Carolina/1/1918, A/Puerto Rico/08/1934, or A/California/04/2009, or an H5N1 strain, e.g., A/Indonesia/5/2005 or A/Vietnam/1203/2004; and d) (c) and one or both of (a) and (b).


Competition between the antibody molecule and a reference antibody molecule can be determined by evaluating the ability of one of the antibody molecule or the reference antibody molecule to decrease binding of the other to a substrate, e.g., HA, e.g., HA1 or HA5, from, e.g., an H1N1 strain, e.g., A/South Carolina/1/1918, A/Puerto Rico/08/1934, or A/California/04/2009, or an H5N1 strain, e.g., A/Indonesia/5/2005 or A/Vietnam/1203/2004. Reduction of the ability to bind can be evaluated by methods in the art. Reduction of the ability to bind can be evaluated, e.g., by one or more of: a) Biacore analysis; b) ELISA assay; or c) flow cytometry.


The antibody molecule can compete with the reference antibody such that binding of the reference antibody is decreased by 50% or more. In an embodiment, the binding agent, e.g., an antibody molecule, comprises one or both of: a heavy chain variable region comprising at least 60, 70, 80, 85, 90, 95, 98 or 99 percent homology with SEQ ID NO: 25; and a light chain variable region comprising at least 60, 70, 80, 85, 90, 95, 98 or 99 percent homology with SEQ ID NO: 52.


In an embodiment, the binding agent, e.g., an antibody molecule, comprises one or both of: a heavy chain variable region comprising at least 60, 70, 80, 85, 90, 95, 98 or 99 percent homology with SEQ ID NO: 25; and a light chain variable region comprising at least 60, 70, 80, 85, 90, 95, 98 or 99 percent homology with SEQ ID NO: 52, wherein, each HC CDR differs by no more than 1, 2, 3, 4 or 5 amino acids, e.g., 1 or 2, e.g., conservative amino acids, from the corresponding CDR of SEQ ID NO: 25 and each LC CDR differs by no more than 1, 2, 3, 4 or 5 amino acids, e.g., 1 or 2, e.g., conservative amino acids, from the corresponding CDR of SEQ ID NO: 52.


In an embodiment, the binding agent, e.g., an antibody molecule, comprises one or both of: a heavy chain variable region comprising at least 60, 70, 80, 85, 90, 95, 98 or 99 percent homology with SEQ ID NO: 25; and a light chain variable region comprising at least 60, 70, 80, 85, 90, 95, 98 or 99 percent homology with SEQ ID NO: 52, wherein the antibody molecule comprises 1, 2, 3, 4, 5, or all of: (i) a HC CDR1 comprising: S at the 1st position and A at the 3rd position in HC CDR1; (ii) a HC CDR2 comprising one or both, e.g., one of: V at the 2nd position; or N at the 7th position and Q at the 16th position in HC CDR2; (iii) a HC CDR3 comprising: R at the 3rd position (and optionally, L at the 3rd position); (iv) a LC CDR1 comprising one or both of, e.g., one of: I at the 3rd position; or D at the 6th position in LC CDR1; (v) a LC CDR2 comprising one, two, or three of, e.g., one of: G at the 2nd position; Y at the 4th position; or L at the 5th position in LC CDR2; (vi) a LC CDR3 comprising: S at the 9th position in LC CDR3.


In an embodiment, the binding agent, e.g., an antibody molecule, comprises: (a) a heavy chain immunoglobulin variable region segment comprising SEQ ID NO:25 (or a sequence that differs by no more than 1, 2, 3, 4 or 5 amino acids, e.g., conservative amino acids, therefrom); and (b) a light chain variable region segment comprising SEQ ID NO:52 (or a sequence that differs by no more than 1, 2, 3, 4 or 5 amino acids, e.g., conservative amino acids, therefrom).


In an embodiment, the binding agent, e.g., an antibody molecule, comprises one or both of: (a) a heavy chain immunoglobulin variable region segment comprising: a CDR1 comprising the sequence S-Y-A-M-H (SEQ ID NO:68) (or a sequence that differs by no more than, 1, 2, 3, 4, or 5, e.g., 1 or 2 amino acids, e.g., conservative amino acids, therefrom); a CDR2 comprising the sequence V-V-S-Y-D-G-N-Y-K-Y-Y-A-D-S-V-Q-G (SEQ ID NO:69) (or a sequence that differs by no more than, 1, 2, 3, 4, or 5, e.g., 1 or 2 amino acids, e.g., conservative amino acids, therefrom); a CDR3 comprising the sequence D-S-R-L-R-S-L-L-Y-F-E-W-L-S-Q-G-Y-F-N-P (SEQ ID NO:70) (or a sequence that differs by no more than, 1, 2, 3, 4, or 5, e.g., 1 or 2 amino acids, e.g., conservative amino acids, therefrom); and (b) a light chain variable region segment comprising: a CDR1 comprising the sequence: Q-S-I-T-F-D-Y-K-N-Y-L-A (SEQ ID NO: 145) (or a sequence that differs by no more than, 1, 2, 3, 4, or 5, e.g., 1 or 2 amino acids, e.g., conservative amino acids, therefrom); a CDR2 comprising the sequence W-G-S-Y-L-E-S (SEQ ID NO:72) (or a sequence that differs by no more than, 1, 2, 3, 4, or 5, e.g., 1 or 2 amino acids, e.g., conservative amino acids, therefrom); a CDR3 comprising the sequence Q-Q-H-Y-R-T-P-P-S (SEQ ID NO:73) (or a sequence that differs by no more than, 1, 2, 3, 4, or 5, e.g., 1 or 2 amino acids, e.g., conservative amino acids, therefrom).


In an embodiment, the binding agent, e.g., an antibody molecule, comprises one or both of: a) LC CDR1-3, that collectively, differ from the AB 044 LC CDR1-3 by no more than, 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10, e.g., 1, 2, 3, or 4, amino acids, e.g., conservative amino acids; and b) HC CDR1-3, that collectively, differ from the AB 044 HC CDR1-3 by no more than, 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10, e.g., 1, 2, 3, or 4, amino acids, e.g., conservative amino acids.


In one embodiment, the antibody molecule comprises one or both of: (a) a heavy chain immunoglobulin variable region segment comprising SEQ ID NO: 25; and (b) a light chain variable region segment comprising SEQ ID NO: 52.


In an embodiment, the binding agent is an antibody molecule comprising one or both of: (a) a heavy chain immunoglobulin variable region segment comprising: a CDR1 comprising the sequence S-Y-A-M-H (SEQ ID NO:68) (or a sequence that differs by no more than, 1, 2, or 3, e.g., 1 or 2, amino acids, e.g., conservative amino acids, there from, optionally provided that at least 1 or 2 of the highlighted residue are not changed, e.g., both S and A are not changed); a CDR2 comprising the sequence V-V-S-Y-D-G-N-Y-K-Y-Y-A-D-S-V-Q-G (SEQ ID NO:69) (or a sequence that differs by no more than, 1, 2, 3, 4, or 5, e.g., 1 or 2 amino acids, e.g., conservative amino acids, therefrom, optionally provided that at least 1, 2, or 3 of the highlighted residues are not changed, e.g., V or both N and Q or all three of V, N, and Q are not changed); a CDR3 comprising the sequence D-S-R-L-R-S-L-L-Y-F-E-W-L-S-Q-G-Y-F-N-P (SEQ ID NO:70) (or a sequence that differs by no more than, 1, 2, 3, 4, or 5, e.g., 1 or 2 amino acids, e.g., conservative amino acids, therefrom, optionally provided that R is not changed); and (b) a light chain variable region segment comprising: a CDR1 comprising the sequence: Q-S-I-T-F-D-Y-K-N-Y-L-A (SEQ ID NO: 145) (or a sequence that differs by no more than, 1, 2, 3, 4, or 5, e.g., 1 or 2, amino acids, e.g., conservative amino acids, therefrom, optionally provided that at least 1 or 2 of the highlighted residues are not changed, e.g., I or D is not changed); a CDR2 comprising the sequence W-G-S-Y-L-E-S (SEQ ID NO:72) (or a sequence that differs by no more than, 1, 2, 3, 4, or 5, e.g., 1 or 2, amino acids, e.g., conservative amino acids, therefrom, optionally provided that at least 1, 2 or 3 of the highlighted residues are not changed, e.g., 1, 2 or all of G, Y, and L are not changed); a CDR3 comprising the sequence Q-Q-H-Y-R-T-P-P-S (SEQ ID NO:73) (or a sequence that differs by no more than, 1, 2, 3, 4, or 5, e.g., 1 or 2, amino acids, e.g., conservative amino acids, therefrom, optionally provided that at least 1 or both of the highlighted residues are not changed, e.g., S is not changed). In an embodiment a CDR of the light or heavy chain includes one of the highlighted residues, or one of the highlighted combinations of residues, for that CDR, (i.e., while other residues in that CDR might be changed, the highlighted residue or combination of residues, are not changed). E.g., in an embodiment, V or both N and Q, for heavy chain CDR2 are not changed.


In an embodiment, a CDR of the light and a CDR of the heavy chain each includes one of the highlighted residues, or one of the highlighted combinations of residues, for that CDR. In an embodiment each of two CDRs in the antibody molecule includes one of the highlighted residues, or one of the highlighted combinations of residues, for that CDR. In some embodiments, both are in the light chain. In some embodiments, both are in the heavy chain. In an embodiment each of the three CDRs in the heavy chain includes one of the highlighted residues, or one of the highlighted combinations of residues, for that CDR. In an embodiment each of the three CDRs in the light chain includes one of the highlighted residues, or one of the highlighted combinations of residues, for that CDR. In an embodiment each of the six CDRs in the heavy and light chain includes one of the highlighted residues, or one of the highlighted combinations of residues, for that CDR.


In one embodiment, the binding agent is an antibody molecule that comprises one or more or all of the following properties: (a) both S and A in HC CDR1 are unchanged; (b) V or both N and Q or all three of V, N, and Q in HC CDR2 are unchanged; (c) R in HC CDR3 is unchanged; (d) One or both of I and D in LC CDR1 are unchanged. (e) 1, 2 or 3 of G, Y and L in LC CDR2 are unchanged; or (f) S in LC CDR3 is unchanged. In an embodiment, the antibody molecule comprises 1, 2, 3, 4, 5, or all 6 properties selected from (a) to (f). In an embodiment, the antibody molecule comprises a heavy chain having a one or more properties selected from (a), (b), and (c) and a light chain having one or more properties selected from (d), (e), and (f).


In one embodiment, the binding agent, e.g., an antibody molecule, comprises one or both of: (a) a heavy chain immunoglobulin variable region segment comprising: a CDR1 comprising the sequence S-Y-A-M-H (SEQ ID NO:68); a CDR2 comprising the sequence V-V-S-Y-D-G-N-Y-K-Y-Y-A-D-S-V-Q-G (SEQ ID NO:69); a CDR3 comprising the sequence D-S-R-L-R-S-L-L-Y-F-E-W-L-S-Q-G-Y-F-N-P (SEQ ID NO:70); and (b) a light chain variable region segment comprising: a CDR1 comprising the sequence Q-S-I-T-F-D-Y-K-N-Y-L-A (SEQ ID NO: 145); a CDR2 comprising the sequence W-G-S-Y-L-E-S (SEQ ID NO:72); and a CDR3 comprising the sequence Q-Q-H-Y-R-T-P—P-S (SEQ ID NO:73).


In some embodiments, the antibody molecule comprises one or more or all of the following properties: (i) it fails to produce any escape mutants as determined by the failure of a viral titer to recover following at least 10, 9, 8, 7, 6, or 5 rounds of serial infections in cell culture with a mixture of the antibody molecule and an influenza virus (e.g., an influenza A virus, e.g., a Group 1 strain, e.g., an H1N1 strain, e.g., A/South Carolina/1/1918, A/Puerto Rico/08/1934, or A/California/04/2009, or an H5N1 strain, e.g., A/Indonesia/5/2005 or A/Vietnam/1203/2004, or an influenza B virus, e.g., B/Wisconsin/1/2010); and (ii) it produces fewer escape mutants than does a reference anti-HA antibody molecule, such as Ab 67-11, F16, FI28, C179, F10, CR9114, or CR6261, such as when tested by the method described in (i).


In an embodiment, the antibody molecule comprises one or both of: a) one or more framework regions (FRs) from SEQ ID NO: 25 e.g., the antibody molecule comprises one or more or all of FR1, FR2, FR3, or FR4, or sequences that differ individually, or collectively, by no more than 1, 2, 3, 4, of 5 amino acid residues, e.g., conservative residues, from SEQ ID NO: 25; and b) one or more framework regions (FRs) from SEQ ID NO: 52. E.g., the antibody molecule comprises one or more or all of FR1, FR2, FR3, or FR4, or sequences that differ individually, or collectively, by no more than 1, 2, 3, 4, of 5 amino acid residues, e.g., conservative residues, from SEQ ID NO: 52.


In one embodiment, the antibody molecule comprises: (a) a heavy chain immunoglobulin variable region segment that further comprises one or more or all of: an FR1 comprising the sequence Q-V-Q-L-L-E-T-G-G-G-L-V-K-P-G-Q-S-L-K-L-S-C-A-A-S-G-F-T-F-T (SEQ ID NO:74) (or a sequence that differs by no more than, 1, 2, 3, 4, or 5, e.g., 1 or 2, amino acids, e.g., conservative amino acids, therefrom, optionally provided that T is not changed); an FR2 comprising the sequence W-V-R-Q-P-P-G-K-G-L-E-W-V-A (SEQ ID NO:75) (or a sequence that differs by no more than, 1, 2, 3, 4, or 5, e.g., 1 or 2, amino acids, e.g., conservative amino acids, therefrom, optionally provided that W is not changed, or that if changed, is other than R); an FR3 comprising the sequence R-F-T-I-S-R-D-N-S-K-N-T-L-Y-L-Q-M-N-S-L-R-A-E-D-T-A-V-Y-Y-C-A-K (SEQ ID NO:76) (or a sequence that differs by no more than, 1, 2, 3, 4, or 5, e.g., 1 or 2, amino acids, e.g., conservative amino acids, therefrom, optionally provided that one, two or three of I, R, or L is not changed, or that if I is changed it is other than G, if R is changed it is other than P. or if L is changed it is other than A); and an FR4 comprising the sequence W-G-Q-G-T-T-L-T-V-S-S (SEQ ID NO:77) (or a sequence that differs by no more than, 1, 2, 3, 4, or 5, e.g., 1 or 2, amino acids, e.g., conservative amino acids, therefrom) or W-G-Q-G-T-T-V-T-V-S-S (SEQ ID NO:171) (or a sequence that differs by no more than, 1, 2, 3, 4, or 5, e.g., 1 or 2, amino acids, e.g., conservative amino acids, therefrom); and (b) a light chain immunoglobulin variable region segment comprising one or more or all of: an FR1 comprising the sequence D-I-Q-M-T-Q-S-P-S-S-L-S-A-S-V-G-D-R-V-T-I-T-C-R-S-S (SEQ ID NO:78) (or a sequence that differs by no more than, 1, 2, 3, 4, or 5, e.g., 1 or 2, amino acids, e.g., conservative amino acids, therefrom, optionally provided that R is not changed); an FR2 comprising the sequence W-Y-Q-Q-K-P-G-K-A-P-K-L-L-I-Y (SEQ ID NO:79) (or a sequence that differs by no more than, 1, 2, 3, 4, or 5, e.g., 1 or 2 amino acids, e.g., conservative amino acids, therefrom); an FR3 comprising the sequence G-V-P-S-R-F-S-G-S-G-S-G-T-D-F-T-L-T-I-S-S-L-Q-P-E-D-F-A-T-Y-Y-C (SEQ ID NO:80) (or a sequence that differs by no more than, 1, 2, 3, 4, or 5, e.g., 1 or 2 amino acids, e.g., conservative amino acids, therefrom, optionally provided that C is not changed, or if changed, is other than P); and an FR4 comprising the sequence F-G-Q-G-T-K-V-E-I-K (SEQ ID NO:81) (or a sequence that differs by no more than, 1, 2, 3, 4, or 5, e.g., 1 or 2 amino acids, e.g., conservative amino acids, therefrom). In an embodiment a FR of the light or heavy chain includes one of the highlighted residues, or one of the highlighted combinations of residues, for that FR, (i.e., while other residues in that FR might be changed, the highlighted residue or combination of residues, are not changed). E.g., in an embodiment, one, two or three of I, R, or L for heavy chain FR3 is not changed.


In an embodiment, a FR of the light and a FR of the heavy chain each includes one of the highlighted residues, or one of the highlighted combinations of residues, for that FR. In an embodiment each of two FRs in the antibody molecule includes one of the highlighted residues, or one of the highlighted combinations of residues, for that FR. In some embodiments, both are in the light chain. In some embodiments, both are in the heavy chain. In an embodiment each of FR2 and FR3 in the heavy chain includes one of the highlighted residues, or one of the highlighted combinations of residues, for that FR. In an embodiment each of FR1 and FR2 in the heavy and light chain includes one of the highlighted residues for that FR. In an embodiment all of the highlighted residues in heavy chain FR1-4 are unchanged. In an embodiment all of the highlighted residues in light chain FR1-4 are unchanged. In an embodiment all of the highlighted residues in both heavy and light chain FR1-4 are unchanged. In an embodiment, sequence of FR1 of the heavy chain variable region segment is Q-V-Q-L-L-E-T-G-G-G-L-V-K-P-G-Q-S-L-K-L-S-C-A-A-S-G-F-T-F-T (SEQ ID NO: 74). In an embodiment, sequence of FR1 of the heavy chain variable region segment is E-V-Q-L-L-E-S-G-G-G-L-V-K-P-G-Q-S-L-K-L-S-C-A-A-S-G-F-T-F-T (SEQ ID NO: 173).


In another embodiment, the binding agent, e.g., an antibody molecule, comprises one or more or all of the following properties: (a) it fails to produce any escape mutants as determined by the failure of a viral titer to recover following at least 10, 9, 8, 7, 6, or 5 rounds of serial infections in cell culture with a mixture of the antibody molecule and an influenza virus (e.g., an influenza A virus, e.g., a Group 1 strain, e.g., an H1N1 strain, e.g., A/South Carolina/1/1918, A/Puerto Rico/08/1934, or A/California/04/2009, or an H5N1 strain, e.g., A/Indonesia/5/2005 or A/Vietnam/1203/2004, or an influenza B virus, e.g., B/Wisconsin/1/2010); (b) it produces fewer escape mutants than does a reference anti-HA antibody molecule, e.g., Ab 67-11, FI6, FI28, C179, or CR6261, e.g., when tested by the method described in (a); (c) it binds with high affinity to a hemagglutinin (HA) of at least 1, 2, 3, 4 or 5 influenza subtypes of Group 1 and at least 1, 2, 3, 4 or 5 influenza subtypes of Group 2; (d) it treats or prevents infection by at least 1, 2, 3, 4 or 5 influenza subtypes of Group 1, and by at least 1, 2, 3, 4 or 5 influenza subtypes of Group 2; (e) it inhibits fusogenic activity of the targeted HA; (f) it treats or prevents infection by a Group 1 virus, wherein the virus is an H1, H5, or H9 virus; and treats or prevents infection by a Group 2 virus, wherein the virus is an H3 or H7 virus; (g) it treats or prevents infection by influenza A strains H1N1 and H3N2; (h) it is effective for prevention or treatment of infection, e.g., in humans or mice, with H1N1 and H3N2 when administered at 50 mg/kg, 25 mg/kg, 10 mg/kg, 6 mg/kg, 5 mg/kg, 4 mg/kg, 3 mg/kg, 2 mg/kg or 1 mg/kg; (i) it treats or prevents infection by influenza A strains H5N1; (j) it is effective for prevention or treatment of infection, e.g., in humans or mice, with H5N1 when administered at 50 mg/kg, 25 mg/kg, 10 mg/kg, 6 mg/kg, 5 mg/kg, 4 mg/kg, 3 mg/kg, 2 mg/kg or 1 mg/kg; (k) it binds with high affinity to a hemagglutinin (HA) of an influenza B virus, e.g., B/Wisconsin/1/2010; (l) it treats or prevents infection by an influenza B virus, e.g., B/Wisconsin/1/2010; (m) it is effective for prevention or treatment of infection, e.g., in humans or mice, with an influenza B virus, e.g., B/Wisconsin/1/2010 when administered at 50 mg/kg, 25 mg/kg, 10 mg/kg, 6 mg/kg, 5 mg/kg, 4 mg/kg, 3 mg/kg, 2 mg/kg or 1 mg/kg; (n) the concentration of antibody molecule required for 50% neutralization of influenza A virus is less than 10 μg/mL; (o) the concentration of antibody molecule required for 50% neutralization of influenza B virus, e.g., B/Wisconsin/1/2010, is less than 10 μg/mL; (p) it prevents or minimizes secondary infection (e.g., secondary bacterial infection) or effects thereof on a subject; (q) it is effective for preventing or minimizing secondary infection (e.g., secondary bacterial infection) or effects thereof on a subject when administered at 50 mg/kg, 25 mg/kg, 10 mg/kg, 6 mg/kg, 5 mg/kg, 4 mg/kg, 3 mg/kg, 2 mg/kg or 1 mg/kg; (r) it binds an epitope which comprises or consists of the hemagglutinin trimer interface; and (s) it binds an epitope other than that bound by a reference anti-HA antibody molecule, e.g., Ab 67-11, F16, F128, C179, F10, CR9114, or CR6261, e.g., when tested by a method disclosed herein, e.g., by competition in an ELISA assay.


In an embodiment, the binding agent, e.g., an antibody molecule, specifically binds the HA antigen. In an embodiment, the antibody molecule binds an epitope that has one, two, three, four, five, or all of, the following properties a)-f): a) it includes one, two, or all of, H3 HA1 residues N38, 1278, and D291; b) it includes H3 HA2 residue N12; c) it does not include one, two or all of, H3 HA1 residues Q327, T328, and R329; d) it does not include one, two, three, four, or all of, H3 HA2 residues G1, L2, F3, G4, and D46; e) it includes one, two, or all of, H3 HA1 residues T318, R321, and V323; or f) it includes 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, or all of, H3 HA2 residues A7, E11, I18, D19, G20, W21, L38, K39, T41, Q42, A43, I45, I48, N49, L52, N53, I56, and E57.


In an embodiment, the antibody molecule has properties: a) and b). In an embodiment, the antibody molecule has properties: c) and d). In an embodiment, the antibody molecule has properties: a); and c) or d). In an embodiment, the antibody molecule has properties: b); and c) or d). In an embodiment, the antibody molecule has properties: c); and a) or b). In an embodiment, the antibody molecule has properties: d); and a) or b). In an embodiment, the antibody molecule has properties: a), b), c) and d). In an embodiment, the antibody molecule has properties: a), b), c), d), e), and f).


In an embodiment, the antibody molecule has a KD for H3 of equal to or less than 10−6, wherein said KD is increased by at least 2, 5, 10, or 100 fold, by a mutation or mutations in any of: a) H3 HA1 residues N38, 1278, or D291; b) H3 HA2 residue N12; c) H3 HA1 residues T318, R321, or V323; or d) H3 HA2 residues A7, E11, I18, D19, G20, W21, L38, K39, T41, Q42, A43, I45, I48, N49, L52, N53, 156, or E57. In an embodiment, the antibody molecule has a KD for H3 of equal to or less than 10−6, wherein said KD is increased by no more than 2, or 5 fold, by a mutation or mutations in any of: c) H3 HA1 residues Q327, T328, or R329; or d) H3 HA2 residues G1, L2, F3, G4, or D46.


In an embodiment, the antibody molecule binds an epitope that has one, two, three, four, five, or all of, the following properties aa)-ff): aa) it includes one, two, or all of, H1 HA1 residues H31, N279, and S292; bb) it includes H1 HA2 residue G12; cc) it does not include one or both of H1 HA1 residues Q328 and S329; dd) it does not include one, two, three, four, or all of, H1 HA2 residues G1, L2, F3, G4, and D46; ee) it includes one, two, or all of, H1 HA1 residues T319, R322, and 1324 are bound by both Ab 044 and FI6; or ff) it includes 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, or all of, H1 HA2 residues A7, E11, I18, D19, G20, W21, Q38, K39, T41, Q42, N43, I45, I48, T49, V52, N53, 156, and E57.


In an embodiment, the antibody molecule has properties: aa) and bb). In an embodiment, the antibody molecule has properties: cc) and dd). In an embodiment, the antibody molecule has properties: aa); and cc) or dd). In an embodiment, the antibody molecule has properties: bb); and cc) or dd). In an embodiment, the antibody molecule has properties: cc); and aa) or bb). In an embodiment, the antibody molecule has properties: dd); and aa) or bb). In an embodiment, the antibody molecule has properties: aa), bb), cc) and dd). In an embodiment, the antibody molecule has properties: aa), bb), cc), dd), ee), and ff).


In an embodiment, the antibody molecule has a KD for H1 of equal to or less than 10−6, wherein said KD is increased by at least 2, 5, 10, or 100 fold, by a mutation or mutations in any of: aa) H1 HA1 residues H31, N279, and S292; bb) H1 HA2 residue G12; cc) H1 HA1 residues T319, R322, and 1324; or dd) H1 HA2 residues A7, E11, I18, D19, G20, W21, Q38, K39, T41, Q42, N43, I45, I48, T49, V52, N53, 156, and E57. In an embodiment, the antibody molecule has a KD for H1 of equal to or less than 10−6, wherein said KD is increased by no more than 2, or 5 fold, by a mutation or mutations in any of: cc) H1 HA1 residues Q328 and S329; or dd) H1 HA2 residues G1, L2, F3, G4, and D46.


In an embodiment, the antibody molecule has one, two, three or all of the following properties: a) and aa); b) and bb); c) and cc); d) and dd). In an embodiment, the molecule has properties c), cc), d), and dd).


In another aspect, the disclosure features, a binding agent, e.g., an antibody molecule, or preparation, or isolated preparation thereof, comprising a structural or functional property of Ab 069.


In an embodiment, the antibody molecule competes with a reference antibody molecule, e.g., an antibody molecule described herein, for binding to a substrate, e.g., an HA. The reference antibody molecule can be: a) an antibody molecule comprising: i) a heavy chain immunoglobulin variable region segment comprising: a CDR1 comprising the sequence S-Y-A-M-H (SEQ ID NO:68); a CDR2 comprising the sequence V-V-S-Y-D-G-N-Y-K-Y-Y-A-D-S-V-Q-G (SEQ ID NO:69); and a CDR3 comprising the sequence D-S-R-L-R-S-L-L-Y-F-E-W-L-S-Q-G-Y-F-N-P (SEQ ID NO:70); and ii) a light chain variable region segment comprising: a CDR1 comprising the sequence Q-S-I-T-F-E-Y-K-N-Y-L-A (SEQ ID NO: 172); a CDR2 comprising the sequence W-G-S-Y-L-E-S (SEQ ID NO:72); and a CDR3 comprising the sequence Q-Q-H-Y-R-T-P-P-S (SEQ ID NO:73); b) an antibody molecule comprises one or both of: (i) a heavy chain immunoglobulin variable region segment comprising SEQ ID NO: 25; and (ii) a light chain variable region segment comprising SEQ ID NO:155; or c) Ab 069.


The HA can be HA1 or HA5, e.g. from an H1N1 strain, e.g., A/South Carolina/1/1918, A/Puerto Rico/08/1934, or A/California/04/2009, or an H5N1 strain, e.g., A/Indonesia/5/2005 or A/Vietnam/1203/2004. Competition between the antibody molecule and a reference antibody molecule can be determined by evaluating the ability of one of the antibody molecule or the reference antibody molecule to decrease binding of the other to a substrate, e.g., HA, e.g., HA1 or HA5, e.g. from an H1N1 strain, e.g., A/South Carolina/1/1918, A/Puerto Rico/08/1934, or A/California/04/2009, or an H5N1 strain, e.g., A/Indonesia/5/2005 or A/Vietnam/1203/2004. Reduction of the ability to bind can be evaluated by methods in the art. Reduction of the ability to bind can be evaluated, e.g., by one or more of: a) Biacore analysis; b) ELISA assay; c) flow cytometry.


The antibody molecule can compete with the reference antibody such that binding of the reference antibody is decreased by 50% or more. In an embodiment, the antibody molecule binds to the same epitope, or a portion thereof, which the reference antibody molecule binds. In an embodiment, the antibody molecule does not bind to the same epitope, or a portion thereof, which the reference antibody molecule binds. In an embodiment, the antibody molecule binds to the same epitope, or a portion thereof, on HA, as does a reference antibody molecule, e.g. an antibody molecule disclosed herein. The reference antibody molecule can be: a) an antibody molecule comprising: i) a heavy chain immunoglobulin variable region segment comprising: a CDR1 comprising the sequence S-Y-A-M-H (SEQ ID NO:68); a CDR2 comprising the sequence V-V-S-Y-D-G-N-Y-K-Y-Y-A-D-S-V-Q-G (SEQ ID NO:69); and a CDR3 comprising the sequence D-S-R-L-R-S-L-L-Y-F-E-W-L-S-Q-G-Y-F-N-P (SEQ ID NO:70); and ii) a light chain variable region segment comprising: a CDR1 comprising the sequence Q-S-I-T-F-E-Y-K-N-Y-L-A (SEQ ID NO: 172); a CDR2 comprising the sequence W-G-S-Y-L-E-S (SEQ ID NO:72); and a CDR3 comprising the sequence Q-Q-H-Y-R-T-P—P-S (SEQ ID NO:73); b) an antibody molecule comprises one or both of: (i) a heavy chain immunoglobulin variable region segment comprising SEQ ID NO: 25; and (ii) a light chain variable region segment comprising SEQ ID NO: 155; or c) Ab 069.


The HA can be HA1 or HA5, e.g. from an H1N1 strain, e.g., A/South Carolina/1/1918, A/Puerto Rico/08/1934, or A/California/04/2009, or an H5N1 strain, e.g., A/Indonesia/5/2005 or A/Vietnam/1203/2004. Binding to the same epitope, or a portion thereof, can be shown by one or more of: a) mutational analysis, e.g., binding or lack thereof to mutant HA, e.g., if a residue is mutated; b) analysis, e.g., comparison, of the crystal structure of the antibody molecule and HA and the crystal structure of a reference antibody and HA, e.g., to determine the touch points of each; c) competition of the two antibodies for binding to HA, e.g., HA1 or HA5, from, e.g., an H1N1 strain, e.g., A/South Carolina/1/1918, A/Puerto Rico/08/1934, or A/California/04/2009, or an H5N1 strain, e.g., A/Indonesia/5/2005 or A/Vietnam/1203/2004; or d) (c) and one or both of (a) and (b);


Competition between the antibody molecule and a reference antibody molecule can be determined by evaluating the ability of one of the antibody molecule or the reference antibody molecule to decrease binding of the other to a substrate, e.g., HA, e.g., HA1 or HA5, e.g. from an H1N1 strain, e.g., A/South Carolina/1/1918, A/Puerto Rico/08/1934, or A/California/04/2009, or an H5N1 strain, e.g., A/Indonesia/5/2005 or A/Vietnam/1203/2004. Reduction of the ability to bind can be evaluated by methods in the art. Reduction of the ability to bind can be evaluated, e.g., by one or more of: a) Biacore analysis; b) ELISA assay; c) flow cytometry. The antibody molecule can compete with the reference antibody such that binding of the reference antibody is decreased by 50% or more.


In an embodiment, the binding agent, e.g., an antibody molecule, comprises one or both of: a heavy chain variable region comprising at least 60, 70, 80, 85, 90, 95, 98 or 99 percent homology with SEQ ID NO: 25; and a light chain variable region comprising at least 60, 70, 80, 85, 90, 95, 98 or 99 percent homology with SEQ ID NO: 155. In an embodiment, the binding agent, e.g., an antibody molecule, comprises one or both of: a heavy chain variable region comprising at least 60, 70, 80, 85, 90, 95, 98 or 99 percent homology with SEQ ID NO: 25; and a light chain variable region comprising at least 60, 70, 80, 85, 90, 95, 98 or 99 percent homology with SEQ ID NO: 155, wherein each HC CDR differs by no more than 1, 2, 3, 4 or 5 amino acids, e.g., 1 or 2, e.g., conservative amino acids, from the corresponding CDR of SEQ ID NO: 25 and each LC CDR differs by no more than 1, 2, 3, 4 or 5 amino acids, e.g., 1 or 2, e.g., conservative amino acids, from the corresponding CDR of SEQ ID NO: 155. In an embodiment, the binding agent, e.g., an antibody molecule, comprises one or both of: a heavy chain variable region comprising at least 60, 70, 80, 85, 90, 95, 98 or 99 percent homology with SEQ ID NO: 25; and a light chain variable region comprising at least 60, 70, 80, 85, 90, 95, 98 or 99 percent homology with SEQ ID NO: 155, wherein the antibody molecule comprises 1, 2, 3, 4, 5, or all of: (i) a HC CDR1 comprising: S at the 1st position and A at the 3rd position in HC CDR1; (ii) a HC CDR2 comprising one or both, e.g., one of: V at the 2nd position; or N at the 7th position and Q at the 16th position in HC CDR2; (iii) a HC CDR3 comprising: R at the 3rd position (and optionally, L at the 3rd position); (iv) a LC CDR1 comprising one or both of, e.g., one of:; I at the 3rd position; or E at the 6th position in LC CDR1; (v) a LC CDR2 comprising one, two or three of, e.g., one of: G at the 2nd position; Y at the 4th position; or L at the 5th position in LC CDR2; (vi) a LC CDR3 comprising: S at the 9th position in LC CDR3. In an embodiment, the binding agent, e.g., an antibody molecule, comprises one or both of: (a) a heavy chain immunoglobulin variable region segment comprising SEQ ID NO:25 (or a sequence that differs by no more than 1, 2, 3, 4 or 5 amino acids, e.g., conservative amino acids, therefrom); and (b) a light chain variable region segment comprising SEQ ID NO:155 (or a sequence that differs by no more than 1, 2, 3, 4 or 5 amino acids, e.g., conservative amino acids, therefrom). In one embodiment, the antibody molecule comprises one or both of: (a) a heavy chain immunoglobulin variable region segment comprising SEQ ID NO: 25; and (b) a light chain variable region segment comprising SEQ ID NO: 155.


In an embodiment, the binding agent, e.g., an antibody molecule, comprises one or both of: (a) a heavy chain immunoglobulin variable region segment comprising: a CDR1 comprising the sequence S-Y-A-M-H (SEQ ID NO:68) (or a sequence that differs by no more than, 1, 2, 3, 4, or 5, e.g., 1 or 2 amino acids, e.g., conservative amino acids, therefrom); a CDR2 comprising the sequence V-V-S-Y-D-G-N-Y-K-Y-Y-A-D-S-V-Q-G (SEQ ID NO:69) (or a sequence that differs by no more than, 1, 2, 3, 4, or 5, e.g., 1 or 2 amino acids, e.g., conservative amino acids, therefrom); a CDR3 comprising the sequence D-S-R-L-R-S-L-L-Y-F-E-W-L-S-Q-G-Y-F-N-P (SEQ ID NO:70) (or a sequence that differs by no more than, 1, 2, 3, 4, or 5, e.g., 1 or 2 amino acids, e.g., conservative amino acids, therefrom); and (b) a light chain variable region segment comprising: a CDR1 comprising the sequence: Q-S-I-T-F-E-Y-K-N-Y-L-A (SEQ ID NO: 172) or a sequence that differs by no more than, 1, 2, 3, 4, or 5, e.g., 1 or 2 amino acids, e.g., conservative amino acids, therefrom); a CDR2 comprising the sequence W-G-S-Y-L-E-S (SEQ ID NO:72) (or a sequence that differs by no more than, 1, 2, 3, 4, or 5, e.g., 1 or 2 amino acids, e.g., conservative amino acids, therefrom); a CDR3 comprising the sequence Q-Q-H-Y-R-T-P-P-S (SEQ ID NO:73) (or a sequence that differs by no more than, 1, 2, 3, 4, or 5, e.g., 1 or 2 amino acids, e.g., conservative amino acids, therefrom).


In an embodiment, the binding agent, e.g., an antibody molecule, comprises one or both of: a) LC CDR1-3, that collectively, differ from the AB 069 LC CDR1-3 by no more than, 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10, e.g., 1, 2, 3, or 4, amino acids, e.g., conservative amino acids; and b) HC CDR1-3, that collectively, differ from the AB 069 HC CDR1-3 by no more than, 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10, e.g., 1, 2, 3, or 4, amino acids, e.g., conservative amino acids.


In an embodiment, the binding agent is an antibody molecule comprising one or both of: (a) a heavy chain immunoglobulin variable region segment comprising: a CDR1 comprising the sequence S-Y-A-M-H (SEQ ID NO:68) (or a sequence that differs by no more than, 1, 2, or 3, e.g., 1 or 2 amino acids, e.g., conservative amino acids, therefrom, optionally provided that at least 1 or 2 of the highlighted residues are not changed, e.g., both S and A are not changed); a CDR2 comprising the sequence V-V-S-Y-D-G-N-Y-K-Y-Y-A-D-S-V-Q-G (SEQ ID NO:69) (or a sequence that differs by no more than, 1, 2, 3, 4, or 5, e.g., 1 or 2 amino acids, e.g., conservative amino acids, therefrom, optionally provided that at least 1, 2, or 3 of the highlighted residues are not changed, e.g., V or both N and Q or all three of V, N, and Q are not changed); a CDR3 comprising the sequence D-S-R-L-R-S-L-L-Y-F-E-W-L-S-Q-G-Y-F-N-P (SEQ ID NO:70) (or a sequence that differs by no more than, 1, 2, 3, 4, or 5, e.g., 1 or 2 amino acids, e.g., conservative amino acids, therefrom optionally provided that, R is not changed); and (b) a light chain variable region segment comprising: a CDR1 comprising the sequence: Q-S-I-T-F-E-Y-K-N-Y-L-A (SEQ ID NO: 172) or a sequence that differs by no more than, 1, 2, 3, 4, or 5, e.g., 1 or 2 amino acids, e.g., conservative amino acids, therefrom, optionally provided that at least 1 or 2 of the highlighted residues are not changed, e.g., I or E is not changed); a CDR2 comprising the sequence W-G-S-Y-L-E-S (SEQ ID NO:72) (or a sequence that differs by no more than, 1, 2, 3, 4, or 5, e.g., 1 or 2 amino acids, e.g., conservative amino acids, therefrom, optionally provided that at least 1, 2, or 3 of the highlighted residues are not changed, e.g., 1, 2 or all of G, Y, and L are not changed); a CDR3 comprising the sequence Q-Q-H-Y-R-T-P-P-S (SEQ ID NO:73) (or a sequence that differs by no more than, 1, 2, 3, 4, or 5, e.g., 1 or 2 amino acids, e.g., conservative amino acids, therefrom, optionally provided that, at least one or both of the highlighted residues are not changed, e.g., S is not changed).


In an embodiment, a CDR of the light or heavy chain includes one of the highlighted residues, or one of the highlighted combinations of residues, for that CDR, (i.e., while other residues in that CDR might be changed, the highlighted residue or combination of residues, are not changed). In an embodiment a CDR of the light and a CDR of the heavy chain each includes one of the highlighted residues, or one of the highlighted combinations of residues, for that CDR. In an embodiment, each of two CDRs in the antibody molecule includes one of the highlighted residues, or one of the highlighted combinations of residues, for that CDR. In some embodiments, both are in the light chain. In some embodiments, both are in the heavy chain. In an embodiment, each of the three CDRs in the heavy chain includes one of the highlighted residues, or one of the highlighted combinations of residues, for that CDR. In an embodiment, each of the three CDRs in the light chain includes one of the highlighted residues, or one of the highlighted combinations of residues, for that CDR. In an embodiment, each of the six CDRs in the heavy and light chain includes one of the highlighted residues, or one of the highlighted combinations of residues, for that CDR.


In one embodiment, the binding agent is an antibody molecule that comprises one or more or all of the following properties: (a) both S and A in HC CDR1 are unchanged; (b) V or both N and Q or all three of V, N, and Q in HC CDR2 are unchanged; (c) R in HC CDR3 is unchanged; (d) one or both of I and E in LC CDR1 are unchanged; (e) 1, 2 or 3 of G, Y and L in LC CDR2 are unchanged; (f) S in LC CDR3 is unchanged. In an embodiment, the antibody molecule comprises 1, 2, 3, 4, 5, or all 6 properties selected from (a) to (f). In an embodiment, the antibody molecule comprises a heavy chain having a one or more properties selected from (a), (b), and (c) and a light chain having one or more properties selected from (d), (e), and (f). In one embodiment, the antibody molecule comprises one or both of: (a) a heavy chain immunoglobulin variable region segment comprising: a CDR1 comprising the sequence S-Y-A-M-H (SEQ ID NO: 68); a CDR2 comprising the sequence V-V-S-Y-D-G-N-Y-K-Y-Y-A-D-S-V-Q-G (SEQ ID NO: 69); a CDR3 comprising the sequence D-S-R-L-R-S-L-L-Y-F-E-W-L-S-Q-G-Y-F-N-P (SEQ ID NO:70); and (b) a light chain variable region segment comprising: a CDR1 comprising the sequence Q-S-I-T-F-E-Y-K-N-Y-L-A (SEQ ID NO: 172); a CDR2 comprising the sequence W-G-S-Y-L-E-S (SEQ ID NO: 72); and a CDR3 comprising the sequence Q-Q-H-Y-R-T-P-P-S (SEQ ID NO: 73).


In some embodiments, the antibody molecule comprises one or more or all of the following properties: (i) it fails to produce any escape mutants as determined by the failure of a viral titer to recover following at least 10, 9, 8, 7, 6, or 5 rounds of serial infections in cell culture with a mixture of the antibody molecule and an influenza virus (e.g., an influenza A virus, e.g., a Group 1 strain, e.g., an H1N1 strain, e.g., A/South Carolina/1/1918, A/Puerto Rico/08/1934, or A/California/04/2009, or an H5N1 strain, e.g., A/Indonesia/5/2005 or A/Vietnam/1203/2004, or an influenza B virus, e.g., B/Wisconsin/1/2010); and (ii) it produces fewer escape mutants than does a reference anti-HA antibody molecule, such as Ab 67-11, F16, FI28, C179, F10, CR9114, or CR6261, such as when tested by the method described in (i).


In an embodiment, the antibody molecule comprises one or both of: a) one or more framework regions (FRs) from SEQ ID NO: 25, e.g., the antibody molecule comprises one or more or all of FR1, FR2, FR3, or FR4, or sequences that differ individually, or collectively, by no more than 1, 2, 3, 4, of 5 amino acid residues, e.g., conservative residues, from SEQ ID NO: 25; and b) one or more framework regions (FRs) from SEQ ID NO: 155, e.g., the antibody molecule comprises one or more or all of FR1, FR2, FR3, or FR4, or sequences that differ individually, or collectively, by no more than 1, 2, 3, 4, of 5 amino acid residues, e.g., conservative residues, from SEQ ID NO: 155.


In one embodiment, the antibody molecule comprises: (a) a heavy chain immunoglobulin variable region segment that further comprises one or more or all of: an FR1 comprising the sequence Q-V-Q-L-L-E-T-G-G-G-L-V-K-P-G-Q-S-L-K-L-S-C-A-A-S-G-F-T-F-T (SEQ ID NO:74) (or a sequence that differs by no more than, 1, 2, 3, 4, or 5, e.g., 1 or 2, amino acids, e.g., conservative amino acids, therefrom, optionally provided that T is not changed); an FR2 comprising the sequence W-V-R-Q-P-P-G-K-G-L-E-W-V-A (SEQ ID NO:75) (or a sequence that differs by no more than, 1, 2, 3, 4, or 5, e.g., 1 or 2, amino acids, e.g., conservative amino acids, therefrom, optionally provided that W is not changed, or that if changed, is other than R); an FR3 comprising the sequence R-F-T-I-S-R-D-N-S-K-N-T-L-Y-L-Q-M-N-S-L-R-A-E-D-T-A-V-Y-Y-C-A-K (SEQ ID NO:76) (or a sequence that differs by no more than, 1, 2, 3, 4, or 5, e.g., 1 or 2, amino acids, e.g., conservative amino acids, therefrom, optionally provided that one, two or three of I, R, or L is not changed, or that if I is changed it is other than G, if R is changed it is other than P. or if L is changed it is other than A); and (b) the light chain immunoglobulin variable region segment comprises one or more or all of: an FR1 comprising the sequence D-I-Q-M-T-Q-S-P-S-S-L-S-A-S-V-G-D-R-V-T-I-T-C-R-S-S (SEQ ID NO:78) (or a sequence that differs by no more than, 1, 2, 3, 4, or 5, e.g., 1 or 2 amino acids, e.g., conservative amino acids, therefrom, optionally provided that R is not changed); an FR2 comprising the sequence W-Y-Q-Q-K-P-G-K-A-P-K-L-L-I-Y (SEQ ID NO:79) (or a sequence that differs by no more than, 1, 2, 3, 4, or 5, e.g., 1 or 2 amino acids, e.g., conservative amino acids, therefrom); an FR3 comprising the sequence G-V-P-S-R-F-S-G-S-G-S-G-T-D-F-T-L-T-I-S-S-L-Q-P-E-D-F-A-T-Y-Y-C (SEQ ID NO:80) (or a sequence that differs by no more than, 1, 2, 3, 4, or 5, e.g., 1 or 2 amino acids, e.g., conservative amino acids, therefrom, optionally provided that C is not changed, or if changed, is other than P); and an FR4 comprising the sequence F-G-Q-G-T-K-V-E-I-K (SEQ ID NO:81) (or a sequence that differs by no more than, 1, 2, 3, 4, or 5, e.g., 1 or 2 amino acids, e.g., conservative amino acids, therefrom). In an embodiment a FR of the light or heavy chain includes one of the highlighted residues, or one of the highlighted combinations of residues, for that FR, (i.e., while other residues in that FR might be changed, the highlighted residue or combination of residues, are not changed). E.g., in an embodiment, one, two or three of I, R, or L for heavy chain FR3 is not changed.


In an embodiment, a FR of the light and a FR of the heavy chain each includes one of the highlighted residues, or one of the highlighted combinations of residues, for that FR. In an embodiment each of two FRs in the antibody molecule includes one of the highlighted residues, or one of the highlighted combinations of residues, for that FR. In some embodiments, both are in the light chain. In some embodiments, both are in the heavy chain. In an embodiment each of FR2 and FR3 in the heavy chain includes one of the highlighted residues, or one of the highlighted combinations of residues, for that FR. In an embodiment each of FR1 and FR2 in the heavy and light chain includes one of the highlighted residues for that FR. In an embodiment all of the highlighted residues in heavy chain FR1-4 are unchanged. In an embodiment all of the highlighted residues in light chain FR1-4 are unchanged. In an embodiment all of the highlighted residues in both heavy and light chain FR1-4 are unchanged.


In another embodiment, the binding agent, e.g., an antibody molecule, comprises one or more or all of the following properties: (a) it fails to produce any escape mutants as determined by the failure of a viral titer to recover following at least 10, 9, 8, 7, 6, or 5 rounds of serial infections in cell culture with a mixture of the antibody molecule and an influenza virus (e.g., an influenza A virus, e.g., a Group 1 strain, e.g., an H1N1 strain, e.g., A/South Carolina/1/1918, A/Puerto Rico/08/1934, or A/California/04/2009, or an H5N1 strain, e.g., A/Indonesia/5/2005 or A/Vietnam/1203/2004, or an influenza B virus, e.g., B/Wisconsin/1/2010); (b) it produces fewer escape mutants than does a reference anti-HA antibody molecule, e.g., Ab 67-11, FI6, FI28, C179, or CR6261, e.g., when tested by the method described in (a); (c) it binds with high affinity to a hemagglutinin (HA) of at least 1, 2, 3, 4 or 5 influenza subtypes of Group 1 and at least 1, 2, 3, 4 or 5 influenza subtypes of Group 2; (d) it treats or prevents infection by at least 1, 2, 3, 4 or 5 influenza subtypes of Group 1, and by at least 1, 2, 3, 4 or 5 influenza subtypes of Group 2; (e) it inhibits fusogenic activity of the targeted HA; (f) it treats or prevents infection by a Group 1 virus, wherein the virus is an H1, H5, or H9 virus; and treats or prevents infection by a Group 2 virus, wherein the virus is an H3 or H7 virus; (g) it treats or prevents infection by influenza A strains H1N1 and H3N2; (h) it is effective for prevention or treatment of infection, e.g., in humans or mice, with H1N1 and H3N2 when administered at 50 mg/kg, 25 mg/kg, 10 mg/kg, 6 mg/kg, 5 mg/kg, 4 mg/kg, 3 mg/kg, 2 mg/kg or 1 mg/kg; (i) it treats or prevents infection by influenza A strains H5N1; (j) it is effective for prevention or treatment of infection, e.g., in humans or mice, with H5N1 when administered at 50 mg/kg, 25 mg/kg, 10 mg/kg, 6 mg/kg, 5 mg/kg, 4 mg/kg, 3 mg/kg, 2 mg/kg or 1 mg/kg; (k) it binds with high affinity to a hemagglutinin (HA) of an influenza B virus, e.g., B/Wisconsin/1/2010; (l) it treats or prevents infection by an influenza B virus, e.g., B/Wisconsin/1/2010; (m) it is effective for prevention or treatment of infection, e.g., in humans or mice, with an influenza B virus, e.g., B/Wisconsin/1/2010 when administered at 50 mg/kg, 25 mg/kg, 10 mg/kg, 6 mg/kg, 5 mg/kg, 4 mg/kg, 3 mg/kg, 2 mg/kg or 1 mg/kg; (n) the concentration of antibody molecule required for 50% neutralization of influenza A virus is less than 10 μg/mL; (o) the concentration of antibody molecule required for 50% neutralization of influenza B virus, e.g., B/Wisconsin/1/2010, is less than 10 μg/mL; (p) it prevents or minimizes secondary infection (e.g., secondary bacterial infection) or effects thereof on a subject; (q) it is effective for preventing or minimizing secondary infection (e.g., secondary bacterial infection) or effects thereof on a subject when administered at 50 mg/kg, 25 mg/kg, 10 mg/kg, 6 mg/kg, 5 mg/kg, 4 mg/kg, 3 mg/kg, 2 mg/kg or 1 mg/kg; (r) it binds an epitope which comprises or consists of the hemagglutinin trimer interface; and (s) it binds an epitope other than that bound by a reference anti-HA antibody molecule, e.g., Ab 67-11, F16, F128, C179, F10, CR9114, or CR6261, e.g., when tested by a method disclosed herein, e.g., by competition in an ELISA assay.


In an embodiment, the binding agent, e.g., an antibody molecule, specifically binds the HA antigen. In an embodiment, the antibody molecule binds an epitope that has one, two, three, four, five, or all of, the following properties a)-f): a) it includes one, two, or all of, H3 HA1 residues N38, 1278, and D291; b) it includes H3 HA2 residue N12; c) it does not include one, two or all of, H3 HA1 residues Q327, T328, and R329; d) it does not include one, two, three, four, or all of, H3 HA2 residues G1, L2, F3, G4, and D46; e) it includes one, two, or all of, H3 HA1 residues T318, R321, and V323; or f) it includes 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, or all of, H3 HA2 residues A7, E11, I18, D19, G20, W21, L38, K39, T41, Q42, A43, I45, I48, N49, L52, N53, I56, and E57.


In an embodiment, the antibody molecule has properties: a) and b). In an embodiment, the antibody molecule has properties: c) and d). In an embodiment, the antibody molecule has properties: a); and c) or d). In an embodiment, the antibody molecule has properties: b); and c) or d). In an embodiment, the antibody molecule has properties: c); and a) or b). In an embodiment, the antibody molecule has properties: d); and a) or b). In an embodiment, the antibody molecule has properties: a), b), c) and d). In an embodiment, the antibody molecule has properties: a), b), c), d), e), and f).


In an embodiment, the antibody molecule has a KD for H3 of equal to or less than 10−6, wherein said KD is increased by at least 2, 5, 10, or 100 fold, by a mutation or mutations in any of: a) H3 HA1 residues N38, 1278, or D291; b) H3 HA2 residue N12; c) H3 HA1 residues T318, R321, or V323; or d) H3 HA2 residues A7, E11, I18, D19, G20, W21, L38, K39, T41, Q42, A43, I45, I48, N49, L52, N53, 156, or E57. In an embodiment, the antibody molecule has a KD for H3 of equal to or less than 10−6, wherein said KD is increased by no more than 2, or 5 fold, by a mutation or mutations in any of: c) H3 HA1 residues Q327, T328, or R329; or d) H3 HA2 residues G1, L2, F3, G4, or D46.


In an embodiment, the antibody molecule binds an epitope that has one, two, three, four, five, or all of, the following properties aa)-ff): aa) it includes one, two, or all of, H1 HA1 residues H31, N279, and S292; bb) it includes H1 HA2 residue G12; cc) it does not include one or both of H1 HA1 residues Q328 and S329; dd) it does not include one, two, three, four, or all of, H1 HA2 residues G1, L2, F3, G4, and D46; ee) it includes one, two, or all of, H1 HA1 residues T319, R322, and 1324 are bound by both Ab 044 and F16; or ff) it includes 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, or all of, H1 HA2 residues A7, E11, I18, D19, G20, W21, Q38, K39, T41, Q42, N43, I45, I48, T49, V52, N53, 156, and E57. In an embodiment, the antibody molecule has properties: aa) and bb). In an embodiment, the antibody molecule has properties: cc) and dd). In an embodiment, the antibody molecule has properties: aa); and cc) or dd). In an embodiment, the antibody molecule has properties: bb); and cc) or dd). In an embodiment, the antibody molecule has properties: cc); and aa) or bb). In an embodiment, the antibody molecule has properties: dd); and aa) or bb). In an embodiment, the antibody molecule has properties: aa), bb), cc) and dd). In an embodiment, the antibody molecule has properties: aa), bb), cc), dd), ee), and ff).


In an embodiment, the antibody molecule has a KD for H1 of equal to or less than 10−6, wherein said KD is increased by at least 2, 5, 10, or 100 fold, by a mutation or mutations in any of: aa) H1 HA1 residues H31, N279, and S292; bb) H1 HA2 residue G12; cc) H1 HA1 residues T319, R322, and 1324; or dd) H1 HA2 residues A7, E11, I18, D19, G20, W21, Q38, K39, T41, Q42, N43, I45, I48, T49, V52, N53, 156, and E57. In an embodiment, the antibody molecule has a KD for H1 of equal to or less than 10−6, wherein said KD is increased by no more than 2, or 5 fold, by a mutation or mutations in any of: cc) H1 HA1 residues Q328 and S329; or dd) H1 HA2 residues G1, L2, F3, G4, and D46;


In an embodiment, the antibody molecule has one, two, three or all of the following properties: a) and aa); b) and bb); c) and cc); d) and dd). In an embodiment, the molecule has properties c), cc), d), and dd). In an embodiment, the molecule has properties c), cc), d), and dd).


In another aspect, the disclosure features, a binding agent, e.g., an antibody molecule, or preparation, or isolated preparation thereof, comprising a structural or functional property of Ab 032.


In an embodiment, the antibody molecule competes with a reference antibody molecule, e.g., an antibody molecule described herein, for binding to a substrate, e.g., an HA. The reference antibody molecule can be: a) an antibody molecule comprising: i) a heavy chain immunoglobulin variable region segment comprising: a CDR1 comprising the sequence S-Y-A-M-H (SEQ ID NO:68); a CDR2 comprising the sequence V-V-S-Y-D-G-N-Y-K-Y-Y-A-D-S-V-Q-G (SEQ ID NO:69); and a CDR3 comprising the sequence D-S-R-L-R-S-L-L-Y-F-E-W-L-S-Q-G-Y-F-N-P (SEQ ID NO:70); and ii) a light chain variable region segment comprising: a CDR1 comprising the sequence Q-S-I-T-F-N-Y-K-N-Y-L-A (SEQ ID NO: 71); a CDR2 comprising the sequence W-G-S-Y-L-E-S (SEQ ID NO: 72); and a CDR3 comprising the sequence Q-Q-H-Y-R-T-P-P-S (SEQ ID NO:73); b) an antibody molecule comprises one or both of: (i) a heavy chain immunoglobulin variable region segment comprising SEQ ID NO: 25; and (ii) a light chain variable region segment comprising SEQ ID NO: 45; or c) Ab 032.


The HA can be HA1 or HA5, e.g. from an H1N1 strain, e.g., A/South Carolina/1/1918, A/Puerto Rico/08/1934, or A/California/04/2009, or an H5N1 strain, e.g., A/Indonesia/5/2005 or A/Vietnam/1203/2004. Competition between the antibody molecule and a reference antibody molecule can be determined by evaluating the ability of one of the antibody molecule or the reference antibody molecule to decrease binding of the other to a substrate, e.g., HA, e.g., HA1 or HA5, e.g. from an H1N1 strain, e.g., A/South Carolina/1/1918, A/Puerto Rico/08/1934, or A/California/04/2009, or an H5N1 strain, e.g., A/Indonesia/5/2005 or A/Vietnam/1203/2004. Reduction of the ability to bind can be evaluated by methods in the art. Reduction of the ability to bind can be evaluated, e.g., by one or more of: a) Biacore analysis; b) ELISA assay; and c) flow cytometry.


The antibody molecule can compete with the reference antibody such that binding of the reference antibody is decreased by 50% or more. In an embodiment, the antibody molecule binds to the same epitope, or a portion thereof, which the reference antibody molecule binds. In an embodiment, the antibody molecule does not bind to the same epitope, or a portion thereof, which the reference antibody molecule binds. In an embodiment, the antibody molecule binds to the same epitope, or a portion thereof, on HA, as does a reference antibody molecule, e.g. an antibody molecule disclosed herein. The reference antibody molecule can be: a) an antibody molecule comprising: i) a heavy chain immunoglobulin variable region segment comprising a CDR1 comprising the sequence S-Y-A-M-H (SEQ ID NO:68); a CDR2 comprising the sequence V-V-S-Y-D-G-N-Y-K-Y-Y-A-D-S-V-Q-G (SEQ ID NO:69); and a CDR3 comprising the sequence D-S-R-L-R-S-L-L-Y-F-E-W-L-S-Q-G-Y-F-N-P (SEQ ID NO:70); and ii) a light chain variable region segment comprising: a CDR1 comprising the sequence Q-S-I-T-F-N-Y-K-N-Y-L-A (SEQ ID NO: 71); a CDR2 comprising the sequence W-G-S-Y-L-E-S (SEQ ID NO:72); and a CDR3 comprising the sequence Q-Q-H-Y-R-T-P—P-S (SEQ ID NO:73); b) an antibody molecule comprises one or both of: (i) a heavy chain immunoglobulin variable region segment comprising SEQ ID NO: 25; and (ii) a light chain variable region segment comprising SEQ ID NO:45; or c) Ab 32.


The HA can be HA1 or HA5, e.g. from an H1N1 strain, e.g., A/South Carolina/1/1918, A/Puerto Rico/08/1934, or A/California/04/2009, or an H5N1 strain, e.g., A/Indonesia/5/2005 or A/Vietnam/1203/2004. Binding to the same epitope, or a portion thereof, can be shown by one or more of: a) mutational analysis, e.g., binding to HA, or binding affinity for HA, is decreased or abolished if a residue is mutated; b) analysis, e.g., comparison, of the crystal structure of the antibody molecule and HA and the crystal structure of a reference antibody and HA, e.g., to determine the touch points of each; c) competition of the two antibodies for binding to HA, e.g., HA1 or HA5, from, e.g., an H1N1 strain, e.g., A/South Carolina/1/1918, A/Puerto Rico/08/1934, or A/California/04/2009, or an H5N1 strain, e.g., A/Indonesia/5/2005 or A/Vietnam/1203/2004; and d) (c) and one or both of (a) and (b).


Competition between the antibody molecule and a reference antibody molecule can be determined by evaluating the ability of one of the antibody molecule or the reference antibody molecule to decrease binding of the other to a substrate, e.g., HA, e.g., HA1 or HA5, from, e.g., an H1N1 strain, e.g., A/South Carolina/1/1918, A/Puerto Rico/08/1934, or A/California/04/2009, or an H5N1 strain, e.g., A/Indonesia/5/2005 or A/Vietnam/1203/2004. Reduction of the ability to bind can be evaluated by methods in the art. Reduction of the ability to bind can be evaluated, e.g., by one or more of: a) Biacore analysis; b) ELISA assay; and c) flow cytometry. The antibody molecule can compete with the reference antibody such that binding of the reference antibody is decreased by 50% or more.


In an embodiment, the binding agent, e.g., an antibody molecule, comprises one or both of: a heavy chain variable region comprising least 60, 70, 80, 85, 90, 95, 98 or 99 percent homology with SEQ ID NO: 25; and a light chain variable region comprising least 60, 70, 80, 85, 90, 95, 98 or 99 percent homology with SEQ ID NO: 45. In an embodiment, the binding agent, e.g., an antibody molecule, comprises one or both of: a heavy chain variable region comprising least 60, 70, 80, 85, 90, 95, 98 or 99 percent homology with SEQ ID NO: 25; and a light chain variable region comprising least 60, 70, 80, 85, 90, 95, 98 or 99 percent homology with SEQ ID NO: 45, wherein each HC CDR differs by no more than 1, 2, 3, 4 or 5 amino acids, e.g., 1 or 2, e.g., conservative amino acids, from the corresponding CDR of SEQ ID NO: 25 and each LC CDR differs by no more than 1, 2, 3, 4 or 5 amino acids, e.g., 1 or 2, e.g., conservative amino acids, from the corresponding CDR of SEQ ID NO: 45.


In an embodiment, the binding agent, e.g., an antibody molecule, comprises one or both of: a heavy chain variable region comprising least 60, 70, 80, 85, 90, 95, 98 or 99 percent homology with SEQ ID NO: 25; and a light chain variable region comprising least 60, 70, 80, 85, 90, 95, 98 or 99 percent homology with SEQ ID NO: 45, wherein the antibody molecule comprises 1, 2, 3, 4, 5, or all of: (i) a HC CDR1 comprising: S at the 1st position and A at the 3rd position in HC CDR1; (ii) a HC CDR2 comprising one or both, e.g., one of: V at the 2nd position; or N at the 7th position and Q at the 16th position in HC CDR2; (iii) a HC CDR3 comprising: R at the 3rd position (and optionally, L at the 3rd position); (iv) a LC CDR1 comprising: I at the 3rd position; (v) a LC CDR2 comprising one, two, or three of, e.g., one of: G at the 2nd position; Y at the 4th position; or L at the 5th position in LC CDR2; (vi) a LC CDR3 comprising: S at the 9th position in LC CDR3; In an embodiment, the binding agent, e.g., an antibody molecule, comprises one or both of: (a) a heavy chain immunoglobulin variable region segment comprising SEQ ID NO:25 (or a sequence that differs by no more than 1, 2, 3, 4 or 5 amino acids, e.g., conservative amino acids, therefrom); and (b) a light chain variable region segment comprising SEQ ID NO: 155 (or a sequence that differs by no more than 1, 2, 3, 4 or 5 amino acids, e.g., conservative amino acids, therefrom).


In one embodiment, the antibody molecule comprises one or both of: (a) a heavy chain immunoglobulin variable region segment comprising SEQ ID NO: 25; and (b) a light chain variable region segment comprising SEQ ID NO:155. In an embodiment, the binding agent, e.g., an antibody molecule, comprises one or both of: (a) a heavy chain immunoglobulin variable region segment comprising a CDR1 comprising the sequence S-Y-A-M-H (SEQ ID NO:68) (or a sequence that differs by no more than, 1, 2, 3, 4, or 5, e.g., 1 or 2 amino acids, e.g., conservative amino acids, therefrom); a CDR2 comprising the sequence V-V-S-Y-D-G-N-Y-K-Y-Y-A-D-S-V-Q-G (SEQ ID NO:69) (or a sequence that differs by no more than, 1, 2, 3, 4, or 5, e.g., 1 or 2 amino acids, e.g., conservative amino acids, therefrom); a CDR3 comprising the sequence D-S-R-L-R-S-L-L-Y-F-E-W-L-S-Q-G-Y-F-N-P (SEQ ID NO:70) (or a sequence that differs by no more than, 1, 2, 3, 4, or 5, e.g., 1 or 2 amino acids, e.g., conservative amino acids, therefrom); and (b) a light chain variable region segment comprising a CDR1 comprising the sequence: Q-S-I-T-F N-Y-K-N-Y-L-A (SEQ ID NO:71) (or a sequence that differs by no more than, 1, 2, 3, 4, or 5, e.g., 1 or 2 amino acids, e.g., conservative amino acids, therefrom); a CDR2 comprising the sequence W-G-S-Y-L-E-S (SEQ ID NO:72) (or a sequence that differs by no more than, 1, 2, 3, 4, or 5, e.g., 1 or 2 amino acids, e.g., conservative amino acids, therefrom); a CDR3 comprising the sequence Q-Q-H-Y-R-T-P-P-S(SEQ ID NO:73) (or a sequence that differs by no more than, 1, 2, 3, 4, or 5, e.g., 1 or 2 amino acids, e.g., conservative amino acids, therefrom). In an embodiment, the binding agent, e.g., an antibody molecule, comprises one or both of: a) LC CDR1-3, that collectively, differ from the AB 032 LC CDR1-3 by no more than, 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10, e.g., 1, 2, 3, or 4, amino acids, e.g., conservative amino acids; and b) HC CDR1-3, that collectively, differ from the AB 032 HC CDR1-3 by no more than, 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10, e.g., 1, 2, 3, or 4, amino acids, e.g., conservative amino acids. In an embodiment, the binding agent is an antibody molecule comprising one or both of: (a) a heavy chain immunoglobulin variable region segment comprising a CDR1 comprising the sequence S-Y-A-M-H (SEQ ID NO:68) (or a sequence that differs by no more than, 1, 2, or 3, e.g., 1 or 2 amino acids, e.g., conservative amino acids, therefrom, optionally provided that at least 1 or 2 of the highlighted residues are not changed, e.g., both S and A are not changed); a CDR2 comprising the sequence V-V-S-Y-D-G-N-Y-K-Y-Y-A-D-S-V-Q-G (SEQ ID NO:69) (or a sequence that differs by no more than, 1, 2, 3, 4, or 5, e.g., 1 or 2 amino acids, e.g., conservative amino acids, therefrom, provided that, e.g., at least 1, 2, or 3 of the highlighted residues are not changed, e.g., V or both N and Q or all three of V, N, and Q are not changed); a CDR3 comprising the sequence D-S-R-L-R-S-L-L-Y-F-E-W-L-S-Q-G-Y-F-N-P (SEQ ID NO:70) (or a sequence that differs by no more than, 1, 2, 3, 4, or 5, e.g., 1 or 2 amino acids, e.g., conservative amino acids, therefrom, optionally provided that R is not changed); and (b) a light chain variable region segment comprising a CDR1 comprising the sequence: Q-S-I-T-F-N-Y-K-N-Y-L-A (SEQ ID NO: 71) or a sequence that differs by no more than, 1, 2, 3, 4, or 5, e.g., 1 or 2 amino acids, e.g., conservative amino acids, therefrom, optionally provided that at least 1 or 2 of the highlighted residues are not changed, e.g., I is not changed); a CDR2 comprising the sequence W-G-S-Y-L-E-S (SEQ ID NO:72) (or a sequence that differs by no more than, 1, 2, 3, 4, or 5, e.g., 1 or 2 amino acids, e.g., conservative amino acids, therefrom, optionally provided that at least 1, 2, or 3 of the highlighted residues are not changed, e.g., 1, 2 or all of G, Y, and L are not changed); a CDR3 comprising the sequence Q-Q-H-Y-R-T-P-P-S (SEQ ID NO:73) (or a sequence that differs by no more than, 1, 2, 3, 4, or 5, e.g., 1 or 2 amino acids, e.g., conservative amino acids, therefrom, optionally provided that at least one or both of the highlighted residues are not changed, e.g., S is not changed). In an embodiment a CDR of the light or heavy chain includes one of the highlighted residues, or one of the highlighted combinations of residues, for that CDR, (i.e., while other residues in that CDR might be changed, the highlighted residue or combination of residues, are not changed).


In an embodiment, a CDR of the light and a CDR of the heavy chain each includes one of the highlighted residues, or one of the highlighted combinations of residues, for that CDR. In an embodiment each of two CDRs in the antibody molecule includes one of the highlighted residues, or one of the highlighted combinations of residues, for that CDR. In some embodiments, both are in the light chain. In some embodiments, both are in the heavy chain. In an embodiment, each of the three CDRs in the heavy chain includes one of the highlighted residues, or one of the highlighted combinations of residues, for that CDR. In an embodiment, each of the three CDRs in the light chain includes one of the highlighted residues, or one of the highlighted combinations of residues, for that CDR. In an embodiment each of the six CDRs in the heavy and light chain includes one of the highlighted residues, or one of the highlighted combinations of residues, for that CDR.


In one embodiment, the binding agent is an antibody molecule that comprises one or more or all of the following properties: (a) both S and A in HC CDR1 are unchanged. (b) V or both N and Q or all three of V, N, and Q in HC CDR2 are unchanged. (c) R in HC CDR3 is unchanged. (d) I in LC CDR1 is unchanged. (e) 1, 2 or 3 of G, Y and L in LC CDR2 are unchanged; (f) S in LC CDR3 is unchanged. In an embodiment, the antibody molecule comprises 1, 2, 3, 4, 5, or all 6 properties selected from (a) to (f). In an embodiment, the antibody molecule comprises a heavy chain having a one or more properties selected from (a), (b), and (c) and a light chain having one or more properties selected from (d), (e), and (f).


In one embodiment, the antibody molecule comprises one or both of: (a) a heavy chain immunoglobulin variable region segment comprising: a CDR1 comprising the sequence S-Y-A-M-H (SEQ ID NO:68); a CDR2 comprising the sequence V-V-S-Y-D-G-N-Y-K-Y-Y-A-D-S-V-Q-G (SEQ ID NO:69); a CDR3 comprising the sequence D-S-R-L-R-S-L-L-Y-F-E-W-L-S-Q-G-Y-F-N-P (SEQ ID NO:70); and (b) a light chain variable region segment comprising a CDR1 comprising the sequence Q-S-I-T-F-N-Y-K-N-Y-L-A (SEQ ID NO: 71); a CDR2 comprising the sequence W-G-S-Y-L-E-S (SEQ ID NO:72); and a CDR3 comprising the sequence Q-Q-H-Y-R-T-P-P-S (SEQ ID NO:73).


In some embodiments, the antibody molecule comprises one or more or all of the following properties: (i) it fails to produce any escape mutants as determined by the failure of a viral titer to recover following at least 10, 9, 8, 7, 6, or 5 rounds of serial infections in cell culture with a mixture of the antibody molecule and an influenza virus (e.g., an influenza A virus, e.g., a Group 1 strain, e.g., an H1N1 strain, e.g., A/South Carolina/1/1918, A/Puerto Rico/08/1934, or A/California/04/2009, or an H5N1 strain, e.g., A/Indonesia/5/2005 or A/Vietnam/1203/2004, or an influenza B virus, e.g., B/Wisconsin/1/2010); and (ii) it produces fewer escape mutants than does a reference anti-HA antibody molecule, such as Ab 67-11, F16, FI28, C179, F10, CR9114, or CR6261, such as when tested by the method described in (i).


In an embodiment, the antibody molecule comprises one or both of: a) one or more framework regions (FRs) from SEQ ID NO: 25, e.g., the antibody molecule comprises one or more or all of FR1, FR2, FR3, or FR4, or sequences that differ individually, or collectively, by no more than 1, 2, 3, 4, of 5 amino acid residues, e.g., conservative residues, from SEQ ID NO: 25; and b) one or more framework regions (FRs) from SEQ ID NO: 45, e.g., the antibody molecule comprises one or more or all of FR1, FR2, FR3, or FR4, or sequences that differ individually, or collectively, by no more than 1, 2, 3, 4, of 5 amino acid residues, e.g., conservative residues, from SEQ ID NO: 45.


In one embodiment, the antibody molecule comprises: (a) a heavy chain immunoglobulin variable region segment that further comprises one or more or all of: an FR1 comprising the sequence Q-V-Q-L-L-E-T-G-G-G-L-V-K-P-G-Q-S-L-K-L-S-C-A-A-S-G-F-T-F-T (SEQ ID NO:74) (or a sequence that differs by no more than, 1, 2, 3, 4, or 5, e.g., 1 or 2, amino acids, e.g., conservative amino acids, therefrom, optionally provided that T is not changed); an FR2 comprising the sequence W-V-R-Q-P-P-G-K-G-L-E-W-V-A (SEQ ID NO:75) (or a sequence that differs by no more than, 1, 2, 3, 4, or 5, e.g., 1 or 2, amino acids, e.g., conservative amino acids, therefrom, optionally provided that W is not changed, or that if changed, is other than R); an FR3 comprising the sequence R-F-T-I-S-R-D-N-S-K-N-T-L-Y-L-Q-M-N-S-L-R-A-E-D-T-A-V-Y-Y-C-A-K (SEQ ID NO:76) (or a sequence that differs by no more than, 1, 2, 3, 4, or 5, e.g., 1 or 2, amino acids, e.g., conservative amino acids, therefrom, optionally provided that one, two or three of I, R, or L is not changed, or that if I is changed it is other than G, if R is changed it is other than P. or if L is changed it is other than A); and an FR4 comprising the sequence W-G-Q-G-T-T-L-T-V-S-S (SEQ ID NO:77) (or a sequence that differs by no more than, 1, 2, 3, 4, or 5, e.g., 1 or 2 amino acids, e.g., conservative amino acids, therefrom) or W-G-Q-G-T-T-V-T-V-S-S (SEQ ID NO:171) (or a sequence that differs by no more than, 1, 2, 3, 4, or 5, e.g., 1 or 2 amino acids, e.g., conservative amino acids, therefrom); and (b) the light chain immunoglobulin variable region segment comprises one or more or all of: an FR1 comprising the sequence D-I-Q-M-T-Q-S-P-S-S-L-S-A-S-V-G-D-R-V-T-I-T-C-R-S-S (SEQ ID NO:78) (or a sequence that differs by no more than, 1, 2, 3, 4, or 5, e.g., 1 or 2 amino acids, e.g., conservative amino acids, therefrom, optionally provided that R is not changed); an FR2 comprising the sequence W-Y-Q-Q-K-P-G-K-A-P-K-L-L-I-Y (SEQ ID NO:79) (or a sequence that differs by no more than, 1, 2, 3, 4, or 5, e.g., 1 or 2 amino acids, e.g., conservative amino acids, therefrom); an FR3 comprising the sequence G-V-P-S-R-F-S-G-S-G-S-G-T-D-F-T-L-T-I-S-S-L-Q-P-E-D-F-A-T-Y-Y-C (SEQ ID NO:80) (or a sequence that differs by no more than, 1, 2, 3, 4, or 5, e.g., 1 or 2 amino acids, e.g., conservative amino acids, therefrom, optionally provided that C is not changed, or if changed, is other than P); and an FR4 comprising the sequence F-G-Q-G-T-K-V-E-I-K (SEQ ID NO:81) (or a sequence that differs by no more than, 1, 2, 3, 4, or 5, e.g., 1 or 2 amino acids, e.g., conservative amino acids, therefrom). In an embodiment a FR of the light or heavy chain includes one of the highlighted residues, or one of the highlighted combinations of residues, for that FR, (i.e., while other residues in that FR might be changed, the highlighted residue or combination of residues, are not changed). E.g., in an embodiment, one, two or three of I, R, or L for heavy chain FR3 is not changed.


In an embodiment, a FR of the light and a FR of the heavy chain each includes one of the highlighted residues, or one of the highlighted combinations of residues, for that FR. In an embodiment, each of two FRs in the antibody molecule includes one of the highlighted residues, or one of the highlighted combinations of residues, for that FR. In some embodiments, both are in the light chain. In some embodiments, both are in the heavy chain. In an embodiment each of FR2 and FR3 in the heavy chain includes one of the highlighted residues, or one of the highlighted combinations of residues, for that FR. In an embodiment, each of FR1 and FR2 in the heavy and light chain includes one of the highlighted residues for that FR. In an embodiment, all of the highlighted residues in heavy chain FR1-4 are unchanged. In an embodiment, all of the highlighted residues in light chain FR1-4 are unchanged. In an embodiment all of the highlighted residues in both heavy and light chain FR1-4 are unchanged.


In another embodiment, the binding agent, e.g., an antibody molecule, comprises one or more or all of the following properties: (a) it fails to produce any escape mutants as determined by the failure of a viral titer to recover following at least 10, 9, 8, 7, 6, or 5 rounds of serial infections in cell culture with a mixture of the antibody molecule and an influenza virus (e.g., an influenza A virus, e.g., a Group 1 strain, e.g., an H1N1 strain, e.g., A/South Carolina/1/1918, A/Puerto Rico/08/1934, or A/California/04/2009, or an H5N1 strain, e.g., A/Indonesia/5/2005 or A/Vietnam/1203/2004, or an influenza B virus, e.g., B/Wisconsin/1/2010); (b) it produces fewer escape mutants than does a reference anti-HA antibody molecule, e.g., Ab 67-11, FI6, FI28, C179, or CR6261, e.g., when tested by the method described in (a); (c) it binds with high affinity to a hemagglutinin (HA) of at least 1, 2, 3, 4 or 5 influenza subtypes of Group 1 and at least 1, 2, 3, 4 or 5 influenza subtypes of Group 2; (d) it treats or prevents infection by at least 1, 2, 3, 4 or 5 influenza subtypes of Group 1, and by at least 1, 2, 3, 4 or 5 influenza subtypes of Group 2; (e) it inhibits fusogenic activity of the targeted HA; (f) it treats or prevents infection by a Group 1 virus, wherein the virus is an H1, H5, or H9 virus; and treats or prevents infection by a Group 2 virus, wherein the virus is an H3 or H7 virus; (g) it treats or prevents infection by influenza A strains H1N1 and H3N2; (h) it is effective for prevention or treatment of infection, e.g., in humans or mice, with H1N1 and H3N2 when administered at 50 mg/kg, 25 mg/kg, 10 mg/kg, 6 mg/kg, 5 mg/kg, 4 mg/kg, 3 mg/kg, 2 mg/kg or 1 mg/kg; (i) it treats or prevents infection by influenza A strains H5N1; (j) it is effective for prevention or treatment of infection, e.g., in humans or mice, with H5N1 when administered at 50 mg/kg, 25 mg/kg, 10 mg/kg, 6 mg/kg, 5 mg/kg, 4 mg/kg, 3 mg/kg, 2 mg/kg or 1 mg/kg; (k) it binds with high affinity to a hemagglutinin (HA) of an influenza B virus, e.g., B/Wisconsin/1/2010; (l) it treats or prevents infection by an influenza B virus, e.g., B/Wisconsin/1/2010; (m) it is effective for prevention or treatment of infection, e.g., in humans or mice, with an influenza B virus, e.g., B/Wisconsin/1/2010 when administered at 50 mg/kg, 25 mg/kg, 10 mg/kg, 6 mg/kg, 5 mg/kg, 4 mg/kg, 3 mg/kg, 2 mg/kg or 1 mg/kg; (n) the concentration of antibody molecule required for 50% neutralization of influenza A virus is less than 10 μg/mL; (o) the concentration of antibody molecule required for 50% neutralization of influenza B virus, e.g., B/Wisconsin/1/2010, is less than 10 μg/mL; (p) it prevents or minimizes secondary infection (e.g., secondary bacterial infection) or effects thereof on a subject; (q) it is effective for preventing or minimizing secondary infection (e.g., secondary bacterial infection) or effects thereof on a subject when administered at 50 mg/kg, 25 mg/kg, 10 mg/kg, 6 mg/kg, 5 mg/kg, 4 mg/kg, 3 mg/kg, 2 mg/kg or 1 mg/kg; (r) it binds an epitope which comprises or consists of the hemagglutinin trimer interface; and (s) it binds an epitope other than that bound by a reference anti-HA antibody molecule, e.g., Ab 67-11, F16, F128, C179, F10, CR9114, or CR6261, e.g., when tested by a method disclosed herein, e.g., by competition in an ELISA assay.


In an embodiment, the binding agent, e.g., an antibody molecule, specifically binds the HA antigen. In an embodiment, the antibody molecule binds an epitope that has one, two, three, four, five, or all of, the following properties a)-f): a) it includes one, two, or all of, H3 HA1 residues N38, 1278, and D291; b) it includes H3 HA2 residue N12; c) it does not include one, two or all of, H3 HA1 residues Q327, T328, and R329; d) it does not include one, two, three, four, or all of, H3 HA2 residues G1, L2, F3, G4, and D46; e) it includes one, two, or all of, H3 HA1 residues T318, R321, and V323; or f) it includes 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, or all of, H3 HA2 residues A7, E11, I18, D19, G20, W21, L38, K39, T41, Q42, A43, I45, I48, N49, L52, N53, I56, and E57. In an embodiment, the antibody molecule has properties: a) and b). In an embodiment, the antibody molecule has properties: c) and d). In an embodiment, the antibody molecule has properties: a; and c or d. In an embodiment, the antibody molecule has properties: b); and c) or d). In an embodiment, the antibody molecule has properties: c); and a) or b). In an embodiment, the antibody molecule has properties: d); and a) or b). In an embodiment, the antibody molecule has properties: a), b), c) and d). In an embodiment, the antibody molecule has properties: a), b), c), d), e), and f). In an embodiment, the antibody molecule has a KD for H3 of equal to or less than 10−6, wherein said KD is increased by at least 2, 5, 10, or 100 fold, by a mutation or mutations in any of: a) H3 HA1 residues N38, 1278, or D291; b) H3 HA2 residue N12; c) H3 HA1 residues T318, R321, or V323; or d) H3 HA2 residues A7, E11, I18, D19, G20, W21, L38, K39, T41, Q42, A43, I45, I48, N49, L52, N53, 156, or E57. In an embodiment, the antibody molecule has a KD for H3 of equal to or less than 10−6, wherein said KD is increased by no more than 2, or 5 fold, by a mutation or mutations in any of: c) H3 HA1 residues Q327, T328, or R329; or d) H3 HA2 residues G1, L2, F3, G4, or D46.


In an embodiment, the antibody molecule binds an epitope that has one, two, three, four, five, or all of, the following properties aa)-ff): aa) it includes one, two, or all of, H1 HA1 residues H31, N279, and S292; bb) it includes H1 HA2 residue G12; cc) it does not include one or both of H1 HA1 residues Q328 and S329; dd) it does not include one, two, three, four, or all of, H1 HA2 residues G1, L2, F3, G4, and D46; ee) it includes one, two, or all of, H1 HA1 residues T319, R322, and 1324 are bound by both Ab 044 and FI6; or ff) it includes 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, or all of, H1 HA2 residues A7, E11, I18, D19, G20, W21, Q38, K39, T41, Q42, N43, I45, I48, T49, V52, N53, 156, and E57.


In an embodiment, the antibody molecule has properties: aa) and bb). In an embodiment, the antibody molecule has properties: cc) and dd). In an embodiment, the antibody molecule has properties: aa); and cc) or dd). In an embodiment, the antibody molecule has properties: bb); and c) or dd). In an embodiment, the antibody molecule has properties: cc); and aa) or bb). In an embodiment, the antibody molecule has properties: dd); and aa) or bb). In an embodiment, the antibody molecule has properties: aa), bb), cc) and dd). In an embodiment, the antibody molecule has properties: aa), bb), cc), dd), ee), and ff).


In an embodiment, the antibody molecule has a KD for H1 of equal to or less than 10−6, wherein said KD is increased by at least 2, 5, 10, or 100 fold, by a mutation or mutations in any of: aa) H1 HA1 residues H31, N279, and S292; bb) H1 HA2 residue G12; cc) H1 HA1 residues T319, R322, and 1324; or dd) H1 HA2 residues A7, E11, I18, D19, G20, W21, Q38, K39, T41, Q42, N43, I45, I48, T49, V52, N53, 156, and E57. In an embodiment, the antibody molecule has a KD for H1 of equal to or less than 10−6, wherein said KD is increased by no more than 2, or 5 fold, by a mutation or mutations in any of: cc) H1 HA1 residues Q328 and S329; or dd) H1 HA2 residues G1, L2, F3, G4, and D46; In an embodiment, the antibody molecule has one, two, three or all of the following properties: a) and aa); b) and bb); c) and cc); d) and dd). In an embodiment, the molecule has properties c), cc), d), and dd).


In another aspect, the disclosure features, a binding agent, e.g., an antibody molecule, or preparation, or isolated preparation thereof, comprising a structural or functional property of Ab 031. In an embodiment, the antibody molecule competes with a reference antibody molecule, e.g., an antibody molecule described herein, for binding to a substrate, e.g., an HA. The reference antibody molecule can be:


a) an antibody molecule comprising: i) a heavy chain immunoglobulin variable region segment comprising a CDR1 comprising the sequence S-Y-A-M-H (SEQ ID NO:68); a CDR2 comprising the sequence V-V-S-Y-D-G-N-Y-K-Y-Y-A-D-S-V-Q-G (SEQ ID NO:69); and a CDR3 comprising the sequence D-S-R-L-R-S-L-L-Y-F-E-W-L-S-Q-G-Y-F-N-P (SEQ ID NO:70); and ii) a light chain variable region segment comprising: a CDR1 comprising the sequence Q-S-I-T-F-N-Y-K—N-Y-L-A (SEQ ID NO:71); a CDR2 comprising the sequence W-G-S-Y-L-E-S (SEQ ID NO:72); and a CDR3 comprising the sequence Q-Q-H-Y-R-T-P-P-S (SEQ ID NO:73); b) an antibody molecule comprises one or both of: (i) a heavy chain immunoglobulin variable region segment comprising SEQ ID NO: 24; and (ii) a light chain variable region segment comprising SEQ ID NO:45; or c) Ab 031. The HA can be HA1 or HA5, e.g. from an H1N1 strain, e.g., A/South Carolina/1/1918, A/Puerto Rico/08/1934, or A/California/04/2009, or an H5N1 strain, e.g., A/Indonesia/5/2005 or A/Vietnam/1203/2004. Competition between the antibody molecule and a reference antibody molecule can be determined by evaluating the ability of one of the antibody molecule or the reference antibody molecule to decrease binding of the other to a substrate, e.g., HA, e.g., HA1 or HA5, e.g. from an H1N1 strain, e.g., A/South Carolina/1/1918, A/Puerto Rico/08/1934, or A/California/04/2009, or an H5N1 strain, e.g., A/Indonesia/5/2005 or A/Vietnam/1203/2004. Reduction of the ability to bind can be evaluated by methods in the art. Reduction of the ability to bind can be evaluated, e.g., by one or more of: a) Biacore analysis; b) ELISA assay; and c) flow cytometry. The antibody molecule can compete with the reference antibody such that binding of the reference antibody is decreased by 50% or more.


In an embodiment, the antibody molecule binds to the same epitope, or a portion thereof, which the reference antibody molecule binds. In an embodiment, the antibody molecule does not bind to the same epitope, or a portion thereof, which the reference antibody molecule binds. In an embodiment, the antibody molecule binds to the same epitope, or a portion thereof, on HA, as does a reference antibody molecule, e.g. an antibody molecule disclosed herein. The reference antibody molecule can be: a) an antibody molecule comprising: i) a heavy chain immunoglobulin variable region segment comprising a CDR1 comprising the sequence S-Y-A-M-H (SEQ ID NO:68); a CDR2 comprising the sequence V-V-S-Y-D-G-N-Y-K-Y-Y-A-D-S-V-Q-G (SEQ ID NO:69); and a CDR3 comprising the sequence D-S-R-L-R-S-L-L-Y-F-E-W-L-S-Q-G-Y-F-N-P (SEQ ID NO:70); and ii) a light chain variable region segment comprising: a CDR1 comprising the sequence Q-S-I-T-F—N—Y-K-N-Y-L-A (SEQ ID NO:71); a CDR2 comprising the sequence W-G-S-Y-L-E-S (SEQ ID NO:72); and a CDR3 comprising the sequence Q-Q-H-Y-R-T-P-P-S (SEQ ID NO:73); b) an antibody molecule comprises one or both of: (i) a heavy chain immunoglobulin variable region segment comprising SEQ ID NO: 24; and (ii) a light chain variable region segment comprising SEQ ID NO:45; or c) Ab 031. The HA can be HA1 or HA5, e.g. from an H1N1 strain, e.g., A/South Carolina/1/1918, A/Puerto Rico/08/1934, or A/California/04/2009, or an H5N1 strain, e.g., A/Indonesia/5/2005 or A/Vietnam/1203/2004. Binding to the same epitope, or a portion thereof, can be shown by one or more of: a) mutational analysis, e.g., binding to HA, or binding affinity for HA, is decreased or abolished if a residue is mutated; b) analysis, e.g., comparison, of the crystal structure of the antibody molecule and HA and the crystal structure of a reference antibody and HA, e.g., to determine the touch points of each; c) competition of the two antibodies for binding to HA, e.g., HA1 or HA5, from, e.g., an H1N1 strain, e.g., A/South Carolina/1/1918, A/Puerto Rico/08/1934, or A/California/04/2009, or an H5N1 strain, e.g., A/Indonesia/5/2005 or A/Vietnam/1203/2004; d) (c) and one or both of (a) and (b).


Competition between the antibody molecule and a reference antibody molecule can be determined by evaluating the ability of one of the antibody molecule or the reference antibody molecule to decrease binding of the other to a substrate, e.g., HA, e.g., HA1 or HA5, from, e.g., an H1N1 strain, e.g., A/South Carolina/1/1918, A/Puerto Rico/08/1934, or A/California/04/2009, or an H5N1 strain, e.g., A/Indonesia/5/2005 or A/Vietnam/1203/2004. Reduction of the ability to bind can be evaluated by methods in the art. Reduction of the ability to bind can be evaluated, e.g., by one or more of: a) Biacore analysis; b) ELISA assay; and c) flow cytometry. The antibody molecule can compete with the reference antibody such that binding of the reference antibody is decreased by 50% or more. In an embodiment, the binding agent, e.g., an antibody molecule, comprises one or both of: a heavy chain variable region comprising least 60, 70, 80, 85, 90, 95, 98 or 99 percent homology with SEQ ID NO: 24; and a light chain variable region comprising least 60, 70, 80, 85, 90, 95, 98 or 99 percent homology with SEQ ID NO: 45. In an embodiment, the binding agent, e.g., an antibody molecule, comprises one or both of: a heavy chain variable region comprising least 60, 70, 80, 85, 90, 95, 98 or 99 percent homology with SEQ ID NO: 24; and a light chain variable region comprising least 60, 70, 80, 85, 90, 95, 98 or 99 percent homology with SEQ ID NO: 45, wherein, optionally, each HC CDR differs by no more than 1, 2, 3, 4 or 5 amino acids, e.g., 1 or 2, e.g., conservative amino acids, from the corresponding CDR of SEQ ID NO: 24 and each LC CDR differs by no more than 1, 2, 3, 4 or 5 amino acids, e.g., 1 or 2, e.g., conservative amino acids, from the corresponding CDR of SEQ ID NO: 45.


In an embodiment, the binding agent, e.g., an antibody molecule, comprises one or both of: a heavy chain variable region comprising least 60, 70, 80, 85, 90, 95, 98 or 99 percent homology with SEQ ID NO: 25; and a light chain variable region comprising least 60, 70, 80, 85, 90, 95, 98 or 99 percent homology with SEQ ID NO: 45, wherein the antibody molecule comprises 1, 2, 3, 4, 5, or all of: (i) a HC CDR1 comprising: S at the 1st position and A at the 3rd position in HC CDR1; (ii) a HC CDR2 comprising one or both, e.g., one of: V at the 2nd position; or N at the 7th position and Q at the 16th position in HC CDR2; (iii) a HC CDR3 comprising: R at the 3rd position (and optionally, L at the 3rd position); (iv) a LC CDR1 comprising: I at the 3rd position; (v) a LC CDR2 comprising one, two, or three of, e.g., one of: G at the 2nd position; Y at the 4th position; or L at the 5th position in LC CDR2; (vi) a LC CDR3 comprising: S at the 9th position in LC CDR3.


In an embodiment, the binding agent comprises an antibody molecule comprising: (a) a heavy chain immunoglobulin variable region segment comprising SEQ ID NO:24 (or a sequence that differs by no more than 1, 2, 3, 4 or 5 amino acids, e.g., conservative amino acids, therefrom); and (b) a light chain variable region segment comprising SEQ ID NO:45 (or a sequence that differs by no more than 1, 2, 3, 4 or 5 amino acids, e.g., conservative amino acids, therefrom). In one embodiment, the antibody molecule comprises one or both of: (a) a heavy chain immunoglobulin variable region segment comprising SEQ ID NO: 24; and (b) a light chain variable region segment comprising SEQ ID NO:45. In an embodiment, the binding agent, e.g., an antibody molecule, comprises one or both of: (a) a heavy chain immunoglobulin variable region segment comprising a CDR1 comprising the sequence S-Y-A-M-H (SEQ ID NO:68) (or a sequence that differs by no more than, 1, 2, 3, 4, or 5, e.g., 1 or 2 amino acids, e.g., conservative amino acids, therefrom); a CDR2 comprising the sequence V-V-S-Y-D-G-N-Y-K-Y-Y-A-D-S-V-Q-G (SEQ ID NO:69) (or a sequence that differs by no more than, 1, 2, 3, 4, or 5, e.g., 1 or 2 amino acids, e.g., conservative amino acids, therefrom); and a CDR3 comprising the sequence D-S-R-L-R-S-L-L-Y-F-E-W-L-S-Q-G-Y-F-N-P (SEQ ID NO:70) (or a sequence that differs by no more than, 1, 2, 3, 4, or 5, e.g., 1 or 2 amino acids, e.g., conservative amino acids, therefrom); and (b) a light chain variable region segment comprising a CDR1 comprising the sequence Q-S-I-T-F-N-Y-K-N-Y-L-A (SEQ ID NO:71) (or a sequence that differs by no more than, 1, 2, 3, 4, or 5, e.g., 1 or 2 amino acids, e.g., conservative amino acids, therefrom); a CDR2 comprising the sequence W-G-S-Y-L-E-S (SEQ ID NO: 72) (or a sequence that differs by no more than, 1, 2, 3, 4, or 5, e.g., 1 or 2 amino acids, e.g., conservative amino acids, therefrom); and a CDR3 comprising the sequence Q-Q-H-Y-R-T-P-P-S (SEQ ID NO:73) (or a sequence that differs by no more than, 1, 2, 3, 4, or 5, e.g., 1 or 2 amino acids, e.g., conservative amino acids, therefrom).


In an embodiment, the binding agent, e.g., an antibody molecule, comprises one or both of: a) LC CDR1-3, that collectively, differ from the AB 031 LC CDR1-3 by no more than, 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10, e.g., 1, 2, 3, or 4, amino acids, e.g., conservative amino acids; and b) HC CDR1-3, that collectively, differ from the AB 031 HC CDR1-3 by no more than, 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10, e.g., 1, 2, 3, or 4, amino acids, e.g., conservative amino acids. In an embodiment, the binding agent, e.g., an antibody molecule, comprises one or both of: (a) a heavy chain immunoglobulin variable region segment comprising a CDR1 comprising the sequence S-Y-A-M-H (SEQ ID NO:68) (or a sequence that differs by no more than, 1, 2, or 3, e.g., 1 or 2 amino acids, e.g., conservative amino acids, therefrom, optionally provided that at least 1 or 2 of the highlighted residues are not changed, e.g., both S and A are not changed); a CDR2 comprising the sequence V-V-S-Y-D-G-N-Y-K-Y-Y-A-D-S-V-Q-G (SEQ ID NO:69) (or a sequence that differs by no more than, 1, 2, 3, 4, or 5, e.g., 1 or 2 amino acids, e.g., conservative amino acids, therefrom, provided that, e.g., at least 1, 2, or 3 of the highlighted residues are not changed, e.g., V or both N and Q or all three of V, N, and Q are not changed); a CDR3 comprising the sequence D-S-R-L-R-S-L-L-Y-F-E-W-L-S-Q-G-Y-F-N-P (SEQ ID NO:70) (or a sequence that differs by no more than, 1, 2, 3, 4, or 5, e.g., 1 or 2 amino acids, e.g., conservative amino acids, therefrom optionally provided that, e.g., R is not changed); and (b) a light chain variable region segment comprising a CDR1 comprising the sequence Q-S-I-T-F-N-Y-K—N-Y-L-A (SEQ ID NO: 71) or a sequence that differs by no more than, 1, 2, 3, 4, or 5, e.g., 1 or 2 amino acids, e.g., conservative amino acids, therefrom, optionally provided that at least 1 or 2 of the highlighted residues are not changed, e.g., I is not changed); a CDR2 comprising the sequence W-G-S-Y-L-E-S (SEQ ID NO:72) (or a sequence that differs by no more than, 1, 2, 3, 4, or 5, e.g., 1 or 2 amino acids, e.g., conservative amino acids, therefrom, optionally provided that at least 1, 2, or 3 of the highlighted residues are not changed, e.g., 1, 2 or all of G, Y, and L are not changed); a CDR3 comprising the sequence Q-Q-H-Y-R-T-P-P-S (SEQ ID NO:73) (or a sequence that differs by no more than, 1, 2, 3, 4, or 5, e.g., 1 or 2 amino acids, e.g., conservative amino acids, therefrom, optionally provided that at least one or both of the highlighted residues are not changed, e.g., S is not changed). In an embodiment a CDR of the light or heavy chain includes one of the highlighted residues, or one of the highlighted combinations of residues, for that CDR, (i.e., while other residues in that CDR might be changed, the highlighted residue or combination of residues, are not changed).


In an embodiment a CDR of the light and a CDR of the heavy chain each includes one of the highlighted residues, or one of the highlighted combinations of residues, for that CDR. In an embodiment each of two CDRs in the antibody molecule includes one of the highlighted residues, or one of the highlighted combinations of residues, for that CDR. In some embodiments, both are in the light chain. In some embodiments, both are in the heavy chain. In an embodiment each of the three CDRs in the heavy chain includes one of the highlighted residues, or one of the highlighted combinations of residues, for that CDR. In an embodiment each of the three CDRs in the light chain includes one of the highlighted residues, or one of the highlighted combinations of residues, for that CDR. In an embodiment each of the six CDRs in the heavy and light chain includes one of the highlighted residues, or one of the highlighted combinations of residues, for that CDR.


In one embodiment, the binding agent is an antibody molecule that comprises one or more or all of the following properties: (a) both S and A in HC CDR1 are unchanged; (b) V or both N and Q or all three of V, N, and Q in HC CDR2 are unchanged; (c) R in HC CDR3 is unchanged; (d) I in LC CDR1 is unchanged; (e) 1, 2 or 3 of G, Y, and L in LC CDR2 are unchanged; (f) S in LC CDR3 is unchanged. In an embodiment, the antibody molecule comprises 1, 2, 3, 4, 5, or all 6 properties selected from (a) to (f). In an embodiment, the antibody molecule comprises a heavy chain having a one or more properties selected from (a), (b), and (c) and a light chain having one or more properties selected from (d), (e), and (f).


In the embodiment, the antibody molecule comprises one or both of: (a) a heavy chain immunoglobulin variable region segment comprising: a CDR1 comprising the sequence S-Y-A-M-H (SEQ ID NO:68); a CDR2 comprising the sequence V-V-S-Y-D-G-N-Y-K-Y-Y-A-D-S-V-Q-G (SEQ ID NO:69); and a CDR3 comprising the sequence D-S-R-L-R-S-L-L-Y-F-E-W-L-S-Q-G-Y-F-N-P (SEQ ID NO:70); and (b) a light chain variable region segment comprising a CDR1 comprising the sequence Q-S-I-T-F-N-Y-K-N-Y-L-A (SEQ ID NO:71); a CDR2 comprising the sequence W-G-S-Y-L-E-S (SEQ ID NO:72); and a CDR3 comprising the sequence Q-Q-H-Y-R-T-P-P-S (SEQ ID NO:73). In some embodiments, the antibody molecule comprises one or more or all of the following properties: (i) it fails to produce any escape mutants as determined by the failure of a viral titer to recover following at least 10, 9, 8, 7, 6, or 5 rounds of serial infections in cell culture with a mixture of the antibody molecule and an influenza virus (e.g., an influenza A virus, e.g., a Group 1 strain, e.g., an H1N1 strain, e.g., A/South Carolina/1/1918, A/Puerto Rico/08/1934, or A/California/04/2009, or an H5N1 strain, e.g., A/Indonesia/5/2005 or A/Vietnam/1203/2004, or an influenza B virus, e.g., B/Wisconsin/1/2010); and (ii) it produces fewer escape mutants than does a reference anti-HA antibody molecule, e.g., Ab 67-11, FI6, FI28, C179, F10, CR9114, or CR6261, e.g., when tested by the method described in (i).


In an embodiment, the antibody molecule comprises one or both of: a) one or more framework regions (FRs) from SEQ ID NO: 24, e.g., the antibody molecule comprises one or more or all of FR1, FR2, FR3, or FR4, or sequences that differ individually, or collectively, by no more than 1, 2, 3, 4, of 5 amino acid residues, e.g., conservative residues, from SEQ ID NO: 24; and b) one or more framework regions (FRs) from SEQ ID NO: 45, e.g., the antibody molecule comprises one or more or all of FR1, FR2, FR3, or FR4, or sequences that differ individually, or collectively, by no more than 1, 2, 3, 4, of 5 amino acid residues, e.g., conservative residues, from SEQ ID NO: 45.


In one embodiment, the antibody molecule comprises: (a) a heavy chain immunoglobulin variable region segment that further comprises one or more or all of: an FR1 comprising the sequence E-V-Q-L-L-E-S-G-G-G-L-V-K-P-G-Q-S-L-K-L-S-C-A-A-S-G-F-T-F-T (SEQ ID NO:82) (or a sequence that differs by no more than, 1, 2, 3, 4, or 5, e.g., 1 or 2 amino acids, e.g., conservative amino acids, therefrom, optionally provided that T is not changed); an FR2 comprising the sequence W-V-R-Q-P-P-G-K-G-L-E-W-V-A (SEQ ID NO:75) (or a sequence that differs by no more than, 1, 2, 3, 4, or 5, e.g., 1 or 2 amino acids, e.g., conservative amino acids, therefrom, optionally provided that W is not changed, or that if changed, is other than R); an FR3 comprising the sequence R-F-T-I-S-R-D-N-S-K-N-T-L-Y-L-Q-M-N-S-L-R-A-E-D-T-A-V-Y-Y-C-A-K (SEQ ID NO:76) (or a sequence that differs by no more than, 1, 2, 3, 4, or 5, e.g., 1 or 2 amino acids, e.g., conservative amino acids, therefrom, optionally provided that one, two or three of I, R, or L is not changed, or that if I is changed it is other than G, if R is changed it is other than P. or if L is changed it is other than A); and an FR4 comprising the sequence W-G-Q-G-T-T-L-T-V-S-S (SEQ ID NO:77) (or a sequence that differs by no more than, 1, 2, 3, 4, or 5, e.g., 1 or 2 amino acids, e.g., conservative amino acids, therefrom) or W-G-Q-G-T-T-V-T-V-S-S (SEQ ID NO:171) (or a sequence that differs by no more than, 1, 2, 3, 4, or 5, e.g., 1 or 2 amino acids, e.g., conservative amino acids, therefrom); and (a) a light chain immunoglobulin variable region segment further comprises one or more or all of: an FR1 comprising the sequence D-I-Q-M-T-Q-S-P-S-S-L-S-A-S-V-G-D-R-V-T-I-T-C-R-S-S (SEQ ID NO:78) (or a sequence that differs by no more than, 1, 2, 3, 4, or 5, e.g., 1 or 2 amino acids, e.g., conservative amino acids, therefrom, optionally provided that R is not changed); an FR2 comprising the sequence W-Y-Q-Q-K-P-G-K-A-P-K-L-L-I-Y (SEQ ID NO:79) (or a sequence that differs by no more than, 1, 2, 3, 4, or 5, e.g., 1 or 2 amino acids, e.g., conservative amino acids, therefrom); an FR3 comprising the sequence G-V-P-S-R-F-S-G-S-G-S-G-T-D-F-T-L-T-I-S-S-L-Q-P-E-D-F-A-T-Y-Y-C (SEQ ID NO:80) (or a sequence that differs by no more than, 1, 2, 3, 4, or 5, e.g., 1 or 2 amino acids, e.g., conservative amino acids, therefrom, optionally provided that C is not changed, or if changed, is other than P); and an FR4 comprising the sequence F-G-Q-G-T-K-V-E-I-K (SEQ ID NO:81) (or a sequence that differs by no more than, 1, 2, 3, 4, or 5, e.g., 1 or 2 amino acids, e.g., conservative amino acids, therefrom). In an embodiment a FR of the light or heavy chain includes one of the highlighted residues, or one of the highlighted combinations of residues, for that FR, (i.e., while other residues in that FR might be changed, the highlighted residue or combination of residues, are not changed). E.g., in an embodiment, one, two or three of I, R, or L for heavy chain FR3 is not changed. In an embodiment a FR of the light and a FR of the heavy chain each includes one of the highlighted residues, or one of the highlighted combinations of residues, for that FR.


In an embodiment each of two FRs in the antibody molecule includes one of the highlighted residues, or one of the highlighted combinations of residues, for that FR. In some embodiments, both are in the light chain. In some embodiments, both are in the heavy chain. In an embodiment, each of FR2 and FR3 in the heavy chain includes one of the highlighted residues, or one of the highlighted combinations of residues, for that FR. In an embodiment, each of FR1 and FR2 in the heavy and light chain includes one of the highlighted residues for that FR. In an embodiment, all of the highlighted residues in heavy chain FR1-4 are unchanged. In an embodiment, all of the highlighted residues in light chain FR1-4 are unchanged. In an embodiment, all of the highlighted residues in both heavy and light chain FR1-4 are unchanged.


In one embodiment, the antibody molecule comprises: (a) the heavy chain immunoglobulin variable region segment comprises one or more or all of an FR1 comprising the sequence E-V-Q-L-L-E-S-G-G-G-L-V-K-P-G-Q-S-L-K-L-S-C-A-A-S-G-F-T-F-T (SEQ ID NO:82); an FR2 comprising the sequence W-V-R-Q-P-P-G-K-G-L-E-W-V-A (SEQ ID NO:75); an FR3 comprising the sequence R-F-T-I-S-R-D-N-S-K-N-T-L-Y-L-Q-M-N-S-L-R-A-E-D-T-A-V-Y-Y-C-A-K (SEQ ID NO:76); and an FR4 comprising the sequence W-G-Q-G-T-T-L-T-V-S-S (SEQ ID NO:77) or W-G-Q-G-T-T-V-T-V—S-S (SEQ ID NO:171); and (b) the light chain immunoglobulin variable region segment comprising one or more or all of an FR1 comprising the sequence D-I-Q-M-T-Q-S-P-S-S-L-S-A-S-V-G-D-R-V-T-I-T-C-R-S-S (SEQ ID NO:78); an FR2 comprising the sequence W-Y-Q-Q-K-P-G-K-A-P-K-L-L-I-Y (SEQ ID NO:79); an FR3 comprising the sequence G-V-P-S-R-F-S-G-S-G-S-G-T-D-F-T-L-T-I-S-S-L-Q-P-E-D-F-A-T-Y-Y-C(SEQ ID NO:80); and an FR4 comprising the sequence F-G-Q-G-T-K-V-E-I-K (SEQ ID NO:81).


In another embodiment, the antibody molecule comprises one or more or all of the following properties: (a) it fails to produce any escape mutants as determined by the failure of a viral titer to recover following at least 10, 9, 8, 7, 6, or 5 rounds of serial infections in cell culture with a mixture of the antibody molecule and an influenza virus (e.g., an influenza A virus, e.g., a Group 1 strain, e.g., an H1N1 strain, e.g., A/South Carolina/1/1918, A/Puerto Rico/08/1934, or A/California/04/2009, or an H5N1 strain, e.g., A/Indonesia/5/2005 or A/Vietnam/1203/2004, or an influenza B virus, e.g., B/Wisconsin/1/2010); (b) it produces fewer escape mutants than does a reference anti-HA antibody molecule, e.g., Ab 67-11, FI6, FI28, C179, F10, CR9114, or CR6261, e.g., when tested by the method described in (a); (c) it binds with high affinity to a hemagglutinin (HA) of at least 1, 2, 3, 4 or 5 influenza subtypes of Group 1 and at least 1, 2, 3, 4 or 5 influenza subtypes of Group 2; (d) it treats or prevents infection by at least 1, 2, 3, 4 or 5 influenza subtypes of Group 1, and by at least 1, 2, 3, 4 or 5 influenza subtypes of Group 2; (e) it inhibits fusogenic activity of the targeted HA; (f) it treats or prevents infection by a Group 1 virus, wherein the virus is an H1, H5, or H9 virus; and treats or prevents infection by a Group 2 virus, wherein the virus is an H3 or H7 virus; (g) it treats or prevents infection by influenza A strains H1N1 and H3N2; (h) it is effective for prevention or treatment of infection, e.g., in humans or mice, with H1N1 and H3N2 when administered at 50 mg/kg, 25 mg/kg, 10 mg/kg, 6 mg/kg, 5 mg/kg, 4 mg/kg, 3 mg/kg, 2 mg/kg, or 1 mg/kg; (i) it treats or prevents infection by influenza A strains H5N1; (j) it is effective for prevention or treatment of infection, e.g., in humans or mice, with H5N1 when administered at 50 mg/kg, 25 mg/kg, 10 mg/kg, 6 mg/kg, 5 mg/kg, 4 mg/kg, 3 mg/kg, 2 mg/kg, or 1 mg/kg; (k) it binds with high affinity to a hemagglutinin (HA) of an influenza B virus, e.g., B/Wisconsin/1/2010; (l) it treats or prevents infection by an influenza B virus, e.g., B/Wisconsin/1/2010; (m) it is effective for prevention or treatment of infection, e.g., in humans or mice, with an influenza B virus, e.g., B/Wisconsin/1/2010 when administered at 50 mg/kg, 25 mg/kg, 10 mg/kg, 6 mg/kg, 5 mg/kg, 4 mg/kg, 3 mg/kg, 2 mg/kg, or 1 mg/kg; (n) the concentration of antibody molecule required for 50% neutralization of influenza A virus is less than 10 μg/mL; (o) the concentration of antibody molecule required for 50% neutralization of influenza B virus, e.g., B/Wisconsin/1/2010, is less than 10 μg/mL; (p) it prevents or minimizes secondary infection (e.g., secondary bacterial infection) or effects thereof on a subject; (q) it is effective for preventing or minimizing secondary infection (e.g., secondary bacterial infection) or effects thereof on a subject when administered at 50 mg/kg, 25 mg/kg, 10 mg/kg, 6 mg/kg, 5 mg/kg, 4 mg/kg, 3 mg/kg, 2 mg/kg, or 1 mg/kg; (r) it binds an epitope which comprises or consists of the hemagglutinin trimer interface; and (s) it binds an epitope other than that bound by a reference anti-HA antibody molecule, e.g., Ab 67-11, FI6, FI28, C179, F10, CR9114, or CR6261, e.g., when tested by a method disclosed herein, e.g., by competition in an ELISA assay.


In another aspect, the disclosure features an antibody molecule comprising: (a) a heavy chain immunoglobulin variable region segment comprising SEQ ID NO:24 (or a sequence that differs by no more than 1, 2, 3, 4 or 5 amino acids, e.g., conservative amino acids, therefrom); and (b) a light chain variable region segment comprising SEQ ID NO:45 (or a sequence that differs by no more than 1, 2, 3, 4 or 5 amino acids, e.g., conservative amino acids, therefrom). In some embodiments, the antibody molecule comprises one or more or all of the following properties: (i) it fails to produce any escape mutants as determined by the failure of a viral titer to recover following at least 10, 9, 8, 7, 6, or 5 rounds of serial infections in cell culture with a mixture of the antibody molecule and an influenza a virus, e.g., a Group 1 strain, e.g., an H1N1 strain, e.g., A/South Carolina/1/1918, A/Puerto Rico/08/1934, or A/California/04/2009, or an H5N1 strain, e.g., A/Indonesia/5/2005 or A/Vietnam/1203/2004; and (ii) it produces fewer escape mutants than does a reference anti-HA antibody molecule, such as Ab 67-11, F16, FI28, C179, F10, CR9114, or CR6261, such as when tested by the method described in (i).


In an embodiment, the binding agent, e.g., an antibody molecule, specifically binds the HA antigen. In an embodiment, the antibody molecule binds an epitope that has one, two, three, four, five, or all of, the following properties a)-f): a) it includes one, two, or all of, H3 HA1 residues N38, 1278, and D291; b) it includes H3 HA2 residue N12; c) it does not include one, two or all of, H3 HA1 residues Q327, T328, and R329; d) it does not include one, two, three, four, or all of, H3 HA2 residues G1, L2, F3, G4, and D46; e) it includes one, two, or all of, H3 HA1 residues T318, R321, and V323; or f) it includes 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, or all of, H3 HA2 residues A7, E11, I18, D19, G20, W21, L38, K39, T41, Q42, A43, I45, I48, N49, L52, N53, I56, and E57. In an embodiment, the antibody molecule has properties: a) and b). antibody molecule has properties: c) and d). In an embodiment, the antibody molecule has properties: a); and c) or d). In an embodiment, the antibody molecule has properties: b); and c) or d). In an embodiment, the antibody molecule has properties: c); and a) or b). In an embodiment, the antibody molecule has properties: d); and a) or b).


In an embodiment, the antibody molecule has properties: a), b), c) and d). In an embodiment, the antibody molecule has properties: a), b), c), d), e), and f). In an embodiment, the antibody molecule has a KD for H3 of equal to or less than 10−6, wherein said KD is increased by at least 2, 5, 10, or 100 fold, by a mutation or mutations in any of: a) H3 HA1 residues N38, 1278, or D291; b) H3 HA2 residue N12; c) H3 HA1 residues T318, R321, or V323; or d) H3 HA2 residues A7, E11, I18, D19, G20, W21, L38, K39, T41, Q42, A43, I45, I48, N49, L52, N53, 156, or E57. In an embodiment, the antibody molecule has a KD for H3 of equal to or less than 10−6, wherein said KD is increased by no more than 2, or 5 fold, by a mutation or mutations in any of: c) H3 HA1 residues Q327, T328, or R329; or d) H3 HA2 residues G1, L2, F3, G4, or D46.


In an embodiment, the antibody molecule binds an epitope that has one, two, three, four, five, or all of, the following properties aa)-ff): aa) it includes one, two, or all of, H1 HA1 residues H31, N279, and S292; bb) it includes H1 HA2 residue G12; cc) it does not include one or both of H1 HA1 residues Q328 and S329; dd) it does not include one, two, three, four, or all of, H1 HA2 residues G1, L2, F3, G4, and D46; ee) it includes one, two, or all of, H1 HA1 residues T319, R322, and 1324 are bound by both Ab 044 and F16; or ff) it includes 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, or all of, H1 HA2 residues A7, E11, I18, D19, G20, W21, Q38, K39, T41, Q42, N43, I45, I48, T49, V52, N53, 156, and E57. In an embodiment, the antibody molecule has properties: aa) and bb). In an embodiment, the antibody molecule has properties: cc; and dd. In an embodiment, the antibody molecule has properties: aa); and cc) or dd). In an embodiment, the antibody molecule has properties: bb); and cc) or dd). In an embodiment, the antibody molecule has properties: cc); and aa) or bb). In an embodiment, the antibody molecule has properties: dd); and aa) or bb). In an embodiment, the antibody molecule has properties: aa), bb), cc) and dd). In an embodiment, the antibody molecule has properties: aa), bb), cc), dd), ee), and ff). In an embodiment, the antibody molecule has a KD for H1 of equal to or less than 10−6, wherein said KD is increased by at least 2, 5, 10, or 100 fold, by a mutation or mutations in any of: aa) H1 HA1 residues H31, N279, and S292; bb) H1 HA2 residue G12; cc) H1 HA1 residues T319, R322, and 1324; or dd) H1 HA2 residues A7, E11, I18, D19, G20, W21, Q38, K39, T41, Q42, N43, I45, I48, T49, V52, N53, 156, and E57. In an embodiment, the antibody molecule has a KD for H1 of equal to or less than 10−6, wherein said KD is increased by no more than 2, or 5 fold, by a mutation or mutations in any of: cc) H1 HA1 residues Q328 and S329; or dd) H1 HA2 residues G1, L2, F3, G4, and D46; In an embodiment, the antibody molecule has one, two, three or all of the following properties: a) and aa); b) and bb); c) and cc); d) and dd). In an embodiment, the molecule has properties c), cc), d), and dd).


In another aspect, the disclosure features, a binding agent, e.g., an antibody molecule, or preparation, or isolated preparation thereof, comprising a structural or functional property of one or both a heavy chain variable region and a light chain variable region disclosed herein.


In an embodiment, the antibody molecule competes with a reference antibody molecule, e.g., an antibody molecule described herein, for binding to a substrate, e.g., an HA. The reference antibody molecule can be: a) an antibody molecule comprising the heavy and light CDRs from: a heavy chain variable region from Table 3, Table 4A, or Table 4B, or FIG. 2, FIG. 13, or FIG. 17, of International Publication No. WO2013/170139 or U.S. Application Publication No. 2013/0302349; and a light chain variable region from Table 3, Table 4A, or Table 4B, or FIG. 3, FIG. 14, or FIG. 17, of International Publication No. WO2013/170139 or U.S. Application Publication No. 2013/0302349; b) an antibody molecule that comprises: (i) a heavy chain immunoglobulin variable region segment from Table 3, Table 4A, or Table 4B, or FIG. 2, FIG. 13, or FIG. 17, of International Publication No. WO2013/170139 or U.S. Application Publication No. 2013/0302349; and (ii) a light chain variable region segment from Table 3, Table 4A, or Table 4B, or FIG. 3, FIG. 14, or FIG. 17, of International Publication No. WO2013/170139 or U.S. Application Publication No. 2013/0302349; or c) an antibody disclosed herein.


The HA can be HA1 or HA5, e.g. from an H1N1 strain, e.g., A/South Carolina/1/1918, A/Puerto Rico/08/1934, or A/California/04/2009, or an H5N1 strain, e.g., A/Indonesia/5/2005 or A/Vietnam/1203/2004. Competition between the antibody molecule and a reference antibody molecule can be determined by evaluating the ability of one of the antibody molecule or the reference antibody molecule to decrease binding of the other to a substrate, e.g., HA, e.g., HA1 or HA5, e.g. from an H1N1 strain, e.g., A/South Carolina/1/1918, A/Puerto Rico/08/1934, or A/California/04/2009, or an H5N1 strain, e.g., A/Indonesia/5/2005 or A/Vietnam/1203/2004. Reduction of the ability to bind can be evaluated by methods in the art. Reduction of the ability to bind can be evaluated, e.g., by one or more of: a) Biacore analysis; b) ELISA assay; and c) flow cytometry. The antibody molecule can compete with the reference antibody such that binding of the reference antibody is decreased by 50% or more. In an embodiment, the antibody molecule binds to the same epitope, or a portion thereof, which the reference antibody molecule binds. In an embodiment, the antibody molecule does not bind to the same epitope, or a portion thereof, which the reference antibody molecule binds.


In an embodiment, the antibody molecule binds to the same epitope, or a portion thereof, on HA, as does a reference antibody molecule, e.g. an antibody molecule disclosed herein. The reference antibody molecule can be: a) an antibody molecule comprising the heavy and light CDRs from: a heavy chain variable region from Table 3, Table 4A, or Table 4B, or FIG. 2, FIG. 13, or FIG. 17, of International Publication No. WO2013/170139 or U.S. Application Publication No. 2013/0302349; and a light chain variable region from Table 3, Table 4A, or Table 4B, or FIG. 3, FIG. 14, or FIG. 17, of International Publication No. WO2013/170139 or U.S. Application Publication No. 2013/0302349; b) an antibody molecule that comprises: (i) a heavy chain immunoglobulin variable region segment from Table 3, Table 4A, or Table 4B, FIG. 2, FIG. 13, or FIG. 17, of International Publication No. WO2013/170139 or U.S. Application Publication No. 2013/0302349; and (ii) a light chain variable region segment from Table 3, Table 4A, or Table 4B, FIG. 3, FIG. 14, or FIG. 17, of International Publication No. WO2013/170139 or U.S. Application Publication No. 2013/0302349; or c) an antibody disclosed herein.


The HA can be HA1 or HA5, e.g. from an H1N1 strain, e.g., A/South Carolina/1/1918, A/Puerto Rico/08/1934, or A/California/04/2009, or an H5N1 strain, e.g., A/Indonesia/5/2005 or A/Vietnam/1203/2004. Binding to the same epitope, or a portion thereof, can be shown by one or more of: a) mutational analysis, e.g., binding to HA, or binding affinity for HA, is decreased or abolished if a residue is mutated; b) analysis, e.g., comparison, of the crystal structure of the antibody molecule and HA and the crystal structure of a reference antibody and HA, e.g., to determine the touch points of each; c) competition of the two antibodies for binding to HA, e.g., HA1 or HA5, e.g. from an H1N1 strain, e.g., A/South Carolina/1/1918, A/Puerto Rico/08/1934, or A/California/04/2009, or an H5N1 strain, e.g., A/Indonesia/5/2005 or A/Vietnam/1203/2004; and d) (c) and one or both of (a) and (b).


Competition between the antibody molecule and a reference antibody molecule can be determined by evaluating the ability of one of the antibody molecule or the reference antibody molecule to decrease binding of the other to a substrate, e.g., HA, e.g., HA1 or HA5, from, e.g., an H1N1 strain, e.g., A/South Carolina/1/1918, A/Puerto Rico/08/1934, or A/California/04/2009, or an H5N1 strain, e.g., A/Indonesia/5/2005 or A/Vietnam/1203/2004. Reduction of the ability to bind can be evaluated by methods in the art. Reduction of the ability to bind can be evaluated, e.g., by one or more of: a) Biacore analysis; b) ELISA assay; and c) flow cytometry. The antibody molecule can compete with the reference antibody such that binding of the reference antibody is decreased by 50% or more; d) competition of the two antibodies for binding to HA, e.g., HA1 or HA5, from, e.g., an H1N1 strain, e.g., A/South Carolina/1/1918, A/Puerto Rico/08/1934, or A/California/04/2009, or an H5N1 strain, e.g., A/Indonesia/5/2005 or A/Vietnam/1203/2004; and e) (c) and one or both of (a) and (b).


Competition between the antibody molecule and a reference antibody molecule can be determined by evaluating the ability of one of the antibody molecule or the reference antibody molecule to decrease binding of the other to a substrate, e.g., HA, e.g., HA1 or HA5, from, e.g., an H1N1 strain, e.g., A/South Carolina/1/1918, A/Puerto Rico/08/1934, or A/California/04/2009, or an H5N1 strain, e.g., A/Indonesia/5/2005 or A/Vietnam/1203/2004. Reduction of the ability to bind can be evaluated by methods in the art.


In an embodiment, the binding agent, e.g., an antibody molecule, comprises one or both of: a heavy chain variable region comprising least 60, 70, 80, 85, 90, 95, 98 or 99 percent homology with a reference heavy chain from Table 3, Table 4A, or Table 4B, or FIG. 2, FIG. 13 or FIG. 17, of International Publication No. WO2013/170139 or U.S. Application Publication No. 2013/0302349; and a light chain variable region comprising least 60, 70, 80, 85, 90, 95, 98 or 99 percent homology with reference light chain from Table 3, Table 4A, or Table 4B, or FIG. 3, FIG. 14 or FIG. 17, of International Publication No. WO2013/170139 or U.S. Application Publication No. 2013/0302349, wherein, optionally, each HC CDR differs by no more than 1, 2, 3, 4 or 5 amino acids, e.g., 1 or 2, e.g., conservative amino acids, from the corresponding HC CDR from its reference heavy chain and each LC CDR differs by no more than 1, 2, 3, 4 or 5 amino acids, e.g., 1 or 2, e.g., conservative amino acids, from the corresponding CDR in its reference light chain. In an embodiment, the binding agent, e.g., an antibody molecule, comprises: a heavy chain variable region comprising least 60, 70, 80, 85, 90, 95, 98 or 99 percent homology with a heavy chain from Table 3 and a light chain variable region comprising least 60, 70, 80, 85, 90, 95, 98 or 99 percent homology with the corresponding light chain from Table 3. In an embodiment, the binding agent, e.g., an antibody molecule, comprises: a heavy chain variable region comprising least 60, 70, 80, 85, 90, 95, 98 or 99 percent homology with a heavy chain from Table 4A and a light chain variable region comprising least 60, 70, 80, 85, 90, 95, 98 or 99 percent homology with the corresponding light chain from Table 4A. In an embodiment, the binding agent, e.g., an antibody molecule, comprises: a heavy chain variable region comprising least 60, 70, 80, 85, 90, 95, 98 or 99 percent homology with a heavy chain from Table 4B and a light chain variable region comprising least 60, 70, 80, 85, 90, 95, 98 or 99 percent homology with the corresponding light chain from Table 4B.


In an embodiment, the binding agent, e.g., an antibody molecule, comprises one or both of: a heavy chain variable region from Table 3, Table 4A, or Table 4B, or FIG. 2, FIG. 13, or FIG. 17, of International Publication No. WO2013/170139 or U.S. Application Publication No. 2013/0302349; and a light chain variable region from Table 3, Table 4A, or Table 4B, or FIG. 3, FIG. 14, or FIG. 17, of International Publication No. WO2013/170139 or U.S. Application Publication No. 2013/0302349. In an embodiment, the binding agent, e.g., an antibody molecule, comprises: a heavy chain variable region from Table 3 and the corresponding light chain from Table 3; a heavy chain from Table 4A and the corresponding light chain from Table 4A; or a heavy chain from Table 4B and the corresponding light chain from Table 4B.


In an embodiment, the binding agent, e.g., an antibody molecule, comprises one or both of: (a) a heavy chain immunoglobulin variable region segment comprising a CDR1, a CDR2 and a CDR3 from a heavy chain sequence of Table 3, Table 4A, or Table 4B, or FIG. 2, FIG. 13, or FIG. 17, of International Publication No. WO2013/170139 or U.S. Application Publication No. 2013/0302349 (or CDRs that, individually or collectively, differ therefrom by no more than, 1, 2, 3, 4, or 5, e.g., 1 or 2 amino acids, e.g., conservative amino acids)); and (b) a light chain immunoglobulin variable region segment comprising a CDR1, a CDR2 and a CDR3 from a light chain sequence of Table 3, Table 4A, or Table 4B, or FIG. 3, FIG. 14, or FIG. 17, of International Publication No. WO2013/170139 or U.S. Application Publication No. 2013/0302349 (or CDRs that, individually or collectively, differ therefrom by no more than, 1, 2, 3, 4, or 5, e.g., 1 or 2 amino acids, e.g., conservative amino acids).


In an embodiment, the binding agent, e.g., an antibody molecule, comprises one or both of: CDRs from a heavy chain of Table 3 and the light chain CDRs from the corresponding light chain from Table 3. In an embodiment, the binding agent, e.g., an antibody molecule, comprises one or both of: CDRs from a heavy chain of Table 4A and the light chain CDRs from the corresponding light chain from Table 4A. In an embodiment, the binding agent, e.g., an antibody molecule, comprises one or both of: CDRs from a heavy chain of Table 4B and the light chain CDRs from the corresponding light chain from Table 4B.


In some embodiments, the binding agent, e.g., an antibody molecule, comprises one or more or all of the following properties: (i) it fails to produce any escape mutants as determined by the failure of a viral titer to recover following at least 10, 9, 8, 7, 6, or 5 rounds of serial infections in cell culture with a mixture of the antibody molecule and an influenza virus (e.g., an influenza A virus, e.g., a Group 1 strain, e.g., an H1N1 strain, e.g., A/South Carolina/1/1918, A/Puerto Rico/08/1934, or A/California/04/2009, or an H5N1 strain, e.g., A/Indonesia/5/2005 or A/Vietnam/1203/2004, or an influenza B virus, e.g., B/Wisconsin/1/2010); (ii) it produces fewer escape mutants than does a reference anti-HA antibody molecule, e.g., Ab 67-11, FI6, FI28, C179, F10, CR9114, or CR6261, e.g., when tested by the method described in (i); and (iii) it is other than Ab 67-11 and FI6.


In one embodiment, the antibody molecule comprises one or both of: (a) a heavy chain immunoglobulin variable region segment comprising a CDR1, a CDR2; and a CDR3 from a heavy chain sequence of FIG. 2, FIG. 13, or FIG. 17, of International Publication No. WO2013/170139 or U.S. Application Publication No. 2013/0302349; and (b) a light chain immunoglobulin variable region segment comprising a CDR1, a CDR2 and a CDR3 from a light chain sequence of FIG. 3, FIG. 14, or FIG. 17, of International Publication No. WO2013/170139 or U.S. Application Publication No. 2013/0302349. In one embodiment, the antibody molecule comprises: (a) a heavy chain immunoglobulin variable region segment from FIG. 2 or FIG. 17; and (b) a light chain immunoglobulin variable region segment from FIG. 3 or FIG. 17.


In one embodiment, the heavy chain immunoglobulin variable region further comprises an Isoleucine-Aspartate (Ile-Asp) dipeptide at the N-terminus. In another embodiment, the light chain immunoglobulin variable region further comprises an Ile-Asp dipeptide at the N-terminus. In yet another embodiment, both the heavy chain immunoglobulin variable region and the light chain immunoglobulin variable region or an antibody featured in the disclosure further comprises an Ile-Asp dipeptide at the N-terminus. In other embodiment the Ile-Asp dipeptide is absent from one or both the heavy and light chain.


In one embodiment, the binding agent, e.g., an antibody molecule, further comprises one or more or all of the following: (a) it treats or prevents infection by at least 1, 2, 3, 4 or 5 influenza subtypes of Group 1, and by at least 1, 2, 3, 4 or 5 influenza subtypes of Group 2; (b) it inhibits fusogenic activity of the targeted HA; (c) it treats or prevents infection by a Group 1 virus, wherein the virus is an H1, H5, or H9 virus; and treats or prevents infection by a Group 2 virus, wherein the virus is an H3 or H7 virus; (d) it treats or prevents infection by influenza A strains H1N1 and H3N2; (e) it is effective for prevention or treatment of infection, e.g., in humans or mice, with H1N1 and H3N2 when administered at 50 mg/kg, 25 mg/kg, 10 mg/kg, 6 mg/kg, 5 mg/kg, 4 mg/kg, 3 mg/kg, 2 mg/kg, or 1 mg/kg; (f) it treats or prevents infection by influenza A strains H5N1; (g) it is effective for prevention or treatment of infection, e.g., in humans or mice, with H5N1 when administered at 50 mg/kg, 25 mg/kg, 10 mg/kg, 6 mg/kg, 5 mg/kg, 4 mg/kg, 3 mg/kg, 2 mg/kg, or 1 mg/kg; (h) it binds with high affinity to a hemagglutinin (HA) of an influenza B virus, e.g., B/Wisconsin/1/2010; (i) it treats or prevents infection by an influenza B virus, e.g., B/Wisconsin/1/2010; (j) it is effective for prevention or treatment of infection, e.g., in humans or mice, with an influenza B virus, e.g., B/Wisconsin/1/2010 when administered at 50 mg/kg, 25 mg/kg, 10 mg/kg, 6 mg/kg, 5 mg/kg, 4 mg/kg, 3 mg/kg, 2 mg/kg, or 1 mg/kg; (k) the concentration of antibody molecule required for 50% neutralization of influenza A virus is less than 10 μg/mL; (l) the concentration of antibody molecule required for 50% neutralization of influenza B virus, e.g., B/Wisconsin/1/2010, is less than 10 μg/mL; (m) it prevents or minimizes secondary infection (e.g., secondary bacterial infection) or effects thereof on a subject; (n) it is effective for preventing or minimizing secondary infection (e.g., secondary bacterial infection) or effects thereof on a subject when administered at 50 mg/kg, 25 mg/kg, 10 mg/kg, 6 mg/kg, 5 mg/kg, 4 mg/kg, 3 mg/kg, 2 mg/kg, or 1 mg/kg; (o) it binds an epitope which comprises or consists of the hemagglutinin trimer interface; and (p) it binds an epitope other than that bound by a reference anti-HA antibody molecule, e.g., Ab 67-11, F16, F128, C179, F10, CR9114, or CR6261, e.g., when tested by a method disclosed herein, e.g., by competition in an ELISA assay.


In an embodiment, the antibody molecule comprises one or both of: a) one or more framework regions (FRs) from heavy chain disclosed herein, e.g., the antibody molecule comprises one or more or all of FR1, FR2, FR3, or FR4, or sequences that differ individually, or collectively, by no more than 1, 2, 3, 4, of 5 amino acid residues, e.g., conservative residues, from heavy chain disclosed herein; and b) one or more framework regions (FRs) from light chain disclosed herein, e.g., the antibody molecule comprises one or more or all of FR1, FR2, FR3, or FR4, or sequences that differ individually, or collectively, by no more than 1, 2, 3, 4, of 5 amino acid residues, e.g., conservative residues, from light chain disclosed herein.


In an embodiment, the binding agent, e.g., an antibody molecule, specifically binds the HA antigen. In an embodiment, the antibody molecule binds an epitope that has one, two, three, four, five, or all of, the following properties a)-f): a) it includes one, two, or all of, H3 HA1 residues N38, 1278, and D291; b) it includes H3 HA2 residue N12; c) it does not include one, two or all of, H3 HA1 residues Q327, T328, and R329; d) it does not include one, two, three, four, or all of, H3 HA2 residues G1, L2, F3, G4, and D46; e) it includes one, two, or all of, H3 HA1 residues T318, R321, and V323; or f) it includes 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, or all of, H3 HA2 residues A7, E11, I18, D19, G20, W21, L38, K39, T41, Q42, A43, I45, I48, N49, L52, N53, I56, and E57. In an embodiment, the antibody molecule has properties: a) and b). In an embodiment, the antibody molecule has properties: c) and d). In an embodiment, the antibody molecule has properties: a); and c) or d). In an embodiment, the antibody molecule has properties: b); and c) or d). In an embodiment, the antibody molecule has properties: c); and a) or b). In an embodiment, the antibody molecule has properties: d); and a) or b). In an embodiment, the antibody molecule has properties: a), b), c) and d). In an embodiment, the antibody molecule has properties: a), b), c), d), e), and f). In an embodiment, the antibody molecule has a KD for H3 of equal to or less than 10−6, wherein said KD is increased by at least 2, 5, 10, or 100 fold, by a mutation or mutations in any of: a) H3 HA1 residues N38, 1278, or D291; b) H3 HA2 residue N12; c) H3 HA1 residues T318, R321, or V323; or d) H3 HA2 residues A7, E11, I18, D19, G20, W21, L38, K39, T41, Q42, A43, I45, I48, N49, L52, N53, 156, or E57. In an embodiment, the antibody molecule has a KD for H3 of equal to or less than 10−6, wherein said KD is increased by no more than 2, or 5 fold, by a mutation or mutations in any of: c) H3 HA1 residues Q327, T328, or R329; or d) H3 HA2 residues G1, L2, F3, G4, or D46.


In an embodiment, the antibody molecule binds an epitope that has one, two, three, four, five, or all of, the following properties aa)-ff): aa) it includes one, two, or all of, H1 HA1 residues H31, N279, and S292; bb) it includes H1 HA2 residue G12; cc) it does not include one or both of H1 HA1 residues Q328 and S329; dd) it does not include one, two, three, four, or all of, H1 HA2 residues G1, L2, F3, G4, and D46; ee) it includes one, two, or all of, H1 HA1 residues T319, R322, and 1324 are bound by both Ab 044 and F16; or ff) it includes 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, or all of, H1 HA2 residues A7, E11, I18, D19, G20, W21, Q38, K39, T41, Q42, N43, I45, I48, T49, V52, N53, 156, and E57. In an embodiment, the antibody molecule has properties: aa) and bb). In an embodiment, the antibody molecule has properties: cc) and dd). In an embodiment, the antibody molecule has properties: aa); and cc) or dd). In an embodiment, the antibody molecule has properties: bb); and cc) or dd). In an embodiment, the antibody molecule has properties: cc); and aa) or bb). In an embodiment, the antibody molecule has properties: dd); and aa) or bb). In an embodiment, the antibody molecule has properties: aa), bb), cc) and dd). In an embodiment, the antibody molecule has properties: aa), bb), cc), dd), ee), and ff). In an embodiment, the antibody molecule has a KD for H1 of equal to or less than 10−6, wherein said KD is increased by at least 2, 5, 10, or 100 fold, by a mutation or mutations in any of: aa) H1 HA1 residues H31, N279, and S292; bb) H1 HA2 residue G12; cc) H1 HA1 residues T319, R322, and 1324; or dd) H1 HA2 residues A7, E11, I18, D19, G20, W21, Q38, K39, T41, Q42, N43, I45, I48, T49, V52, N53, 156, and E57. In an embodiment, the antibody molecule has a KD for H1 of equal to or less than 10−6, wherein said KD is increased by no more than 2, or 5 fold, by a mutation or mutations in any of: cc) H1 HA1 residues Q328 and S329; or dd) H1 HA2 residues G1, L2, F3, G4, and D46; In an embodiment, the antibody molecule has one, two, three or all of the following properties: a) and aa); b) and bb); c) and cc); d) and dd). In an embodiment, the molecule has properties c), cc), d), and dd).


In one aspect, the disclosure features an anti-hemagglutinin (anti-HA) binding agent, e.g., antibody molecule, or preparation, or isolated preparation thereof, comprising: (a) a heavy chain immunoglobulin variable region segment comprising one or more or all of a CDR1 comprising the sequence G-F-T-F-[S/T]-[S/T]-Y-[A/G]-M-H (SEQ ID NO: 1), or a sequence that differs from SEQ ID NO:1 by no more than 1 or 2 residues; a CDR2 comprising the sequence V-[I/V/L]-S-[Y/F]-D-G-[S/N]-[Y/N]-[K/R]-Y-Y-A-D-S-V-Q-G (SEQ ID NO:2) or a sequence that differs from SEQ ID NO:2 by no more than 1 or 2 residues; and a CDR3 comprising the sequence D-[S/T]-[R/K/Q]-L-R-[S/T]-L-L-Y-F-E-W-L-S-[Q/S]-G-[Y/L/V]-[F/L]-[N/D]-[P/Y] (SEQ ID NO:3), or a sequence that differs from SEQ ID NO:3 by no more than 1 or 2 residues; and (b) a light chain variable region segment comprising one or more or all of a CDR1 comprising the sequence [K/R]-S-S-Q-[S/T]-[V/L/I]-[T/S]-[Y/F/W]-[N/S/D]-Y-K-N-Y-L-A (SEQ ID NO:4) or a sequence that differs from SEQ ID NO:4 by no more than 1 or 2 residues, or comprising the sequence [K/R]-S-S-Q-[S/T]-[V/L/I]-[T/S]-[Y/F/W]-[N/S/D/Q/R/E]-Y-K-N-Y-L-A (SEQ ID NO: 170) or a sequence that differs from SEQ ID NO: 170 by no more than 1 or 2 residues or [K/R]-S-S-Q-[S/T]-[V/L/I]-[T/S]-[Y/F/W]-[N/S/D/E]-Y-K-N-Y-L-A (SEQ ID NO:4) or a sequence that differs from SEQ ID NO: 170 by no more than 1 or 2 residues; a CDR2 comprising the sequence W-[A/G]-S-[T/A/Y/H/K/D]-[R/L]-E-[S/T] (SEQ ID NO:5) or a sequence that differs from SEQ ID NO:5 by no more than 1 or 2 residues; a CDR3 comprising the sequence Q-Q-[Y/H]-Y-R-T-P-P-[T/S] (SEQ ID NO:6) or a sequence that differs from SEQ ID NO:6 by no more than 1 or 2 residues;


optionally, provided that, if the light chain variable region segment comprises: a CDR 1 comprising the sequence K-S-S-Q-S-V-T-Y-N-Y-K-N-Y-L-A (SEQ ID NO:83); a CDR2 comprising the sequence W-A-S-T-R-E-S (SEQ ID NO:84); and a CDR3 comprising the sequence Q-Q-Y-Y-R-T-P-P-T (SEQ ID NO:85); then the heavy chain variable region segment comprises one or more of the following: (a) CDRs other than the following: a CDR1 comprising the sequence S-Y-G-M-H (SEQ ID NO:86); a CDR2 comprising the sequence V-I-S-Y-D-G-S-Y-K-Y-Y-A-D-S-V-Q-G (SEQ ID NO:87); or a CDR3 comprising the sequence D-S-E-L-R-S-L-L-Y-F-E-W-L-S-Q-G-Y-F-N-P (SEQ ID NO:88); or (b) FRs other than the following: an FR1 other than E-V-Q-L-L-E-S-G-G-G-L-V-K-P-G-Q-S-L-K-L-S-C-A-A-S-G-F-T-F-T (SEQ ID NO:82); an FR2 other than W-V-R-Q-P-P-G-K-G-L-E-W-V-A (SEQ ID NO:75); an FR3 other than R-F-T-I-S-R-D-N-S-K-N-T-L-Y-L-Q-M-N-S-L-R-A-E-D-T-A-V-Y-Y-C-A-K (SEQ ID NO:76); or an FR4 other than W-G-A-G-T-T-L-T-V-S-S (SEQ ID NO:89); (c) a CDR1 where the amino residue at position 5 of SEQ ID NO: 1 is an S, the amino acid residue at position 6 of SEQ ID NO: 1 is a T, or the amino acid residue at position 8 of SEQ ID NO:1 is an A; (d) a CDR2 wherein the amino residue at position 2 of SEQ ID NO:2 is a V or an L, the amino acid at position 4 is an F, the amino acid at position 7 is an N, the amino acid at position 8 is a Y, or the amino acid at position 9 is a R; (e) a CDR3 wherein the amino residue at position 2 of SEQ ID NO:3 is a T, the amino acid residue at position 3 of SEQ ID NO:3 is an R, a K, or a Q, the amino acid residue at position 6 of SEQ ID NO:3 is a T, the amino acid residue at position 15 of SEQ ID NO:3 is an S, the amino acid residue at position 17 of SEQ ID NO:3 is an L, or a V, the amino acid residue at position 18 of SEQ ID NO:3 is an L, the amino acid residue at position 19 of SEQ ID NO:3 is a D, or the amino acid residue at position 20 of SEQ ID NO:3 is a Y; (f) an FR1 wherein the amino residue at position 11 of SEQ ID NO:7 is a Q, or the amino acid residue at position 7 of SEQ ID NO:7 is a T; (g) an FR4 wherein the amino residue at position 3 of SEQ ID NO: 10 is a Q, the amino acid residue at position 5 of SEQ ID NO: 10 is an A; the amino acid residue at position 6 of SEQ ID NO: 10 is an M, or the amino acid residue at position 7 of SEQ ID NO: 10 is a V; or (h) it produces fewer escape mutants than does a reference anti-HA antibody molecule, e.g., Ab 67-11, F16, F128, C179, F10, CR9114, or CR6261, e.g., when tested by a method disclosed herein, and also provided that, if the heavy chain immunoglobulin variable region segment comprises: a CDR1 comprising the sequence S-Y-G-M-H (SEQ ID NO:86); a CDR2 comprising the sequence V-I-S-Y-D-G-S-Y-K-Y-Y-A-D-S-V-Q-G (SEQ ID NO:87); and a CDR3 comprising the sequence D-S-E-L-R-S-L-L-Y-F-E-W-L-S-Q-G-Y-F-N-P (SEQ ID NO:88), then the light chain variable region segment comprises one of more of the following: (a) CDRs other than the following: CDR1 KSSQSVTYNYKNYLA (SEQ ID NO:83); CDR2 WASTRES (SEQ ID NO:84); or CDR3 QQYYRTPPT (SEQ ID NO:85); (b) FRs other than the following: FR1 comprising the sequence EIVMTQSPDSLAVSLGERATINC (SEQ ID NO:90); FR2 comprising the sequence WYQQKPGQPPKLLIY (SEQ ID NO:91); FR3 comprising the sequence GVPDRFSGSGSGTDFTLTISSLQAEDVAVYYC (SEQ ID NO:92); or FR4 comprising the sequence FGGGTKLDIK (SEQ ID NO:93); (c) a CDR1 wherein the amino residue at position 1 of SEQ ID NO:4 is an R, the amino residue at position 5 of SEQ ID NO:4 is a T, the amino residue at position 6 of SEQ ID NO:4 is an L or an I, the amino residue at position 7 of SEQ ID NO:4 is an S, the amino residue at position 8 of SEQ ID NO:4 is an F or a W, or the amino residue at position 9 of SEQ ID NO:4 is an S or a D; (d) a CDR2 wherein the amino residue at position 2 of SEQ ID NO:5 is a G, the amino residue at position 4 of SEQ ID NO:5 is an A, a Y, an H, a K, or a D, the amino residue at position 5 of SEQ ID NO:5 is an L, the amino residue at position 7 of SEQ ID NO:5 is a T; (e) a CDR3 wherein the amino residue at position 3 of SEQ ID NO:6 is an H; the amino acid residue at position 9 of SEQ ID NO:6 is an S; (f) an FR1 wherein the amino residue at position 1 of SEQ ID NO:11 is a D; the amino residue at position 3 of SEQ ID NO:11 is a Q, the amino residue at position 9 of SEQ ID NO:11 is an S, the amino residue at position 10 of SEQ ID NO:11 is a T, the amino residue at position 11 of SEQ ID NO:11 is a V, the amino residue at position 12 of SEQ ID NO:11 is an S, the amino residue at position 13 of SEQ ID NO:11 is an A, the amino residue at position 14 of SEQ ID NO:11 is a T, the amino residue at position 15 of SEQ ID NO:11 is a V or an R, the amino residue at position 17 of SEQ ID NO:11 is a D, the amino residue at position 20 of SEQ ID NO:11 is an S, the amino residue at position 22 of SEQ ID NO:11 is a T, a Q, a D, or an R; (g) an FR2 wherein the amino residue at position 8 of SEQ ID NO: 12 is a K; or the amino residue at position 9 of SEQ ID NO: 12 is an A; (h) an FR3 wherein the amino residue at position 4 of SEQ ID NO: 13 is an E or an S; the amino residue at position 24 of SEQ ID NO: 13 is a P, the amino residue at position 27 of SEQ ID NO: 13 is an F, a K, or a D, the amino residue at position 29 of SEQ ID NO: 13 is a T; (i) an FR4 wherein the amino residue at position 3 of SEQ ID NO: 14 is a Q, a T, an S, or an N, the amino residue at position 7 of SEQ ID NO: 14 is a V, or the amino residue at position 8 of SEQ ID NO: 14 is an E; or (j) it produces fewer escape mutants than does a reference anti-HA antibody molecule, e.g., Ab 67-11, F16, F128, C179, F10, CR9114, or CR6261, e.g., when tested by a method disclosed herein; and further provided that if the light chain variable region segment comprises: a CDR 1 comprising the sequence K-S-S-Q-S-V-T-F-N-Y-K-N-Y-L-A (SEQ ID NO: 146); a CDR2 comprising the sequence W-A-S-A-R-E-S (SEQ ID NO: 147); and a CDR3 comprising the sequence Q-Q-H-Y-R-T-P-P-T (SEQ ID NO: 148); then the heavy chain variable region segment comprises one or more of the following: CDRs other than the CDR's described at FIG. 12 of International Publication No. WO2013/170139 or U.S. Application Publication No. 2013/0302349; or FRs other than the FRs described at FIG. 12 of International Publication No. WO2013/170139 or U.S. Application Publication No. 2013/0302349.


In one embodiment, the heavy chain CDR sequences, collectively, differ from the recited sequences by no more than 5, 4, 3, 2 or 1 amino acid residues; and the light chain CDR sequences, collectively, differ from the recited sequences by no more than 5, 4, 3, 2 or 1 amino acid residues.


Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Although methods and materials similar or equivalent to those described herein can be used in the practice or testing of the invention, suitable methods and materials are described below. All publications, patent applications, patents, and other references mentioned herein are incorporated by reference in their entirety. In case of conflict, the present specification, including definitions, will control. In addition, the materials, methods, and examples are illustrative only and not intended to be limiting.


The details of one or more embodiments featured in the disclosure are set forth in the accompanying drawings and the description below. Other features, objects, and advantages featured in the disclosure will be apparent from the description and drawings, and from the claims.





BRIEF DESCRIPTION OF THE DRAWINGS


FIGS. 1A-1B depict the pharmacokinetic profiles of VIS410 in serum (FIG. 1A) and nasopharyngeal (FIG. 1B) samples. Mean concentrations along with the corresponding standard deviation at each time point were plotted on a logarithmic scale for each dose level. Cohort dose levels are as follows Cohort 1: 2 mg/kg; Cohort 2: 5 mg/kg; Cohort 3: 15 mg/kg; Cohort 4: 30 mg/kg; Cohort 5: 50 mg/kg.



FIGS. 2A-2C depict the microsimulation results of VIS410 prophylactic use. Changes in attack rate (FIG. 2A), overall hospitalization rate (FIG. 2B), and hospitalization rate in individuals older than 65 years of age (FIG. 2C) as a function of the population-level prophylaxis coverage. The boxplots aggregate outcomes over time of administration and transmission setting, as these epidemiological variables might in some cases be difficult to predict. The boxplots for 0% coverage summarize 750 individual simulations, while the boxplots for 2% to 6% coverage summarize 3,750 simulations each. The yellow boxplots show results for VIS410 administration to the elderly only, while the white boxplots show general population VIS410 administration. The whiskers show the full range of outcomes, and the median value is shown next to the median line of each boxplot. With one exception, all pairwise comparisons between different coverage levels, when keeping the group administration method fixed (“all” or “elderly only”), show a statistically significant difference by the Mann-Whitney test (p=0-002); the one exception is in FIG. 2A when comparing no coverage to 2% coverage and distribution to the elderly only (significant at p=0-05). Note that in FIG. 2B when comparing elderly versus general population distribution, the Mann-Whitney p-values are p=0-21 (2% coverage), p=0-05 (4% coverage), and p=0-005 (6% coverage).



FIG. 3 depicts the results of VIS410 prophylactic use stratified by date of administration and transmission setting. Median baseline attack rate (MBAR) is used to separate the simulations into those that have low (<10%), medium (10%-16%), or high (>16%) median attack rates when coverage is zero. In these simulations, VIS410 was administered to the elderly only and coverage was set to 6%. Each boxplot corresponds to 250 simulations. The whiskers show the full range of outcomes, and the median value is shown next to the median line of each boxplot. The gray line denotes the baseline median for each scenario when coverage is zero.



FIG. 4 depicts the protective levels conferred by VIS410 as a function of time (half-life of 13 days).



FIG. 5 depicts the prevalence curves (375 simulations) of seasonal influenza for varying transmission scenarios. Filled colors show the middle 90% ranges of simulation outputs classified into three transmission intensities: attack rate >16% (red/severe), attack rate between 10% and 16% (blue/moderate), attack rate<10% (green/mild).



FIG. 6 depicts the cumulative prevalence curves (375 simulations) of seasonal influenza for varying transmission scenarios. Filled colors show the middle 90% ranges of simulation outputs classified into three transmission intensities: attack rate>16% (red/severe), attack rate between 10% and 16% (blue/moderate), attack rate<10% (green/mild).



FIG. 7 depicts the hospitalization curves (375 simulations) of seasonal influenza for varying transmission scenarios. Vertical axis shows total number of hospitalized patients, of all ages, in a population of one million individuals. Filled colors show the middle 90% ranges of simulation outputs classified into three transmission intensities: attack rate>16% (red/severe), attack rate between 10% and 16% (blue/moderate), attack rate<10% (green/mild).



FIG. 8 depicts the sequence logo of predicted VIS410 epitope positions for H1N1 (top) and H3N2 (bottom) for influenza A strains collected from 2012 through 2015.



FIG. 9 depicts the effect of VIS410 administration on elderly hospitalization events for a low transmission (top panel) and high transmission (bottom panel) epidemic. The effect of VIS410 administration was modeled when prophylaxis was initiated 6 weeks (left), 4 weeks (center), and 2 weeks (right) prior to the peak of the epidemic.



FIG. 10A depicts the weight loss of animals infected with H1N1 PR8 and untreated or treated with ribavirin or a prophylactic dose of VIS410 (0.6, 2.5, or 10 mg/kg).



FIG. 10B depicts the Kaplan-Meier survival curves for animals infected with H1N1 PR8 and untreated or treated with ribavirin or a prophylactic dose of VIS410 (0.6, 2.5, or 10 mg/kg).



FIGS. 11A-11B depict the mean (+SD) serum (FIG. 11A) and nasopharyngeal (FIG. 11B) VIS410 concentration versus time profiles (log-linear scale).



FIG. 12A depicts the median viral shedding versus time profiles of VIS410 compared to Placebo as measured by qRT-PCR in mITT population.



FIG. 12B depicts the median 50% tissue culture infective dose (TCID50) time profiles of VIS410 compared to Placebo as measured by a cell based assay in mITT population.



FIG. 13 depicts the time to resolution of upper respiratory tract symptom score in mITT population.



FIG. 14A depicts the result of phenotypic resistance testing using ViroSpot™ assay.



FIG. 14B depicts the result of phenotypic resistance testing based on IC50.



FIG. 15 depicts the in vivo ADE study design.



FIGS. 16A-16B depict the protection of CD-1 mice from influenza A virus-induced morbidity by VIS410 in a dose dependent manner as compared to irrelevant human IgG1.



FIG. 17 depicts the average lung viral load on Days 1 and 14 pi in CD-1 mice treated with different doses of VIS410 and irrelevant human IgG1.



FIG. 18 depicts the tolerance of VIS410 to existing sequence variation in its epitope.



FIG. 19 depicts the evolutionary trajectory of VIS410 epitope.





Additional figures include FIGS. 1-27 of International Publication No. WO2013/170139 and U.S. Application Publication No. 2013/0302349, the contents of which are incorporated by reference in their entirety.


DETAILED DESCRIPTION

The disclosure is based, at least in part, on the design and synthesis of antibody molecules that can bind an epitope that is conserved across multiple hemagglutinin subtypes of influenza viruses (e.g., influenza A and influenza B viruses). For example, the antibody molecules described herein are useful as broad spectrum therapy against disease caused by at least one influenza A strain belonging to Group 1 and one influenza A strain belonging to Group 2 to neutralize infectivity of viruses belonging to both Group 1 and Group 2 (at least one subtype of each).


The antibody molecules were designed by a rational structure-based approach to target a region on the virus that is not fully accessible to the human immune system and, therefore, not amenable to antibody selection through more classical screening approaches. This rational-based approach to the design and development of broad-spectrum antibody molecules allows for the development of more efficacious vaccines for pandemic and seasonal influenza. This approach also allows for the advance preparation of pandemic vaccines so that they are ready to be employed against specific virus subtypes (e.g., avian virus subtypes) that may mutate to become human-adapted and highly transmissible. Vaccines (e.g., seasonal vaccines) that utilize the antibody molecules described herein can generate a more potent immune response without the use of adjuvants and provide broad protection against viral strain variation.


The antibody molecules described herein can be used, e.g., in methods of protecting a population of subjects from influenza. For example, the protection can include decreasing, in the population, the number of hospital admissions, e.g. of influenza infected individuals; the number incidents of influenza infection; the attack rate; or the number of deaths, e.g. of influenza infected individuals. In certain embodiments, the antibody molecules described herein can be used effectively and safely as either a single-dose therapeutic or prophylactic for influenza. Without wishing to be bound by theory, it is believed that in an embodiment, including the antibody molecules described herein in prophylaxis among the public health interventions for influenza (e.g., seasonal influenza) can result in beneficial effects, for example, lowering attack rates and reducing hospitalizations in high risk individuals.


Definitions

As used herein, the term “antibody molecule” refers to a polypeptide that comprises sufficient sequence from an immunoglobulin heavy chain variable region and/or sufficient sequence from an immunoglobulin light chain variable region, to provide antigen specific binding. It comprises full length antibodies as well as fragments thereof, e.g., Fab fragments, that support antigen binding. Typically an antibody molecule will comprise heavy chain CDR1, CDR2, and CDR3 and light chain CDR1, CDR2, and CDR3 sequence. Antibody molecules include human, humanized, CDR-grafted antibodies and antigen binding fragments thereof. In some embodiments, an antibody molecule comprises a protein that comprises at least one immunoglobulin variable region segment, e.g., an amino acid sequence that provides an immunoglobulin variable domain or immunoglobulin variable domain sequence.


The VH or VL chain of the antibody molecule can further include all or part of a heavy or light chain constant region, to thereby form a heavy or light immunoglobulin chain, respectively. In one embodiment, the antibody molecule is a tetramer of two heavy immunoglobulin chains and two light immunoglobulin chains.


An antibody molecule can comprise one or both of a heavy (or light) chain immunoglobulin variable region segment. As used herein, the term “heavy (or light) chain immunoglobulin variable region segment,” refers to an entire heavy (or light) chain immunoglobulin variable region, or a fragment thereof, that is capable of binding antigen. The ability of a heavy or light chain segment to bind antigen is measured with the segment paired with a light or heavy chain, respectively. In some embodiment, a heavy or light chain segment that is less than a full length variable region will, when paired with the appropriate chain, bind with an affinity that is at least 20, 30, 40, 50, 60, 70, 80, 90, or 95% of what is seen when the full length chain is paired with a light chain or heavy chain, respectively.


An immunoglobulin variable region segment may differ from a reference or consensus sequence. As used herein, to “differ,” means that a residue in the reference sequence or consensus sequence is replaced with either a different residue or an absent or inserted residue.


An antibody molecule can comprise a heavy (H) chain variable region (abbreviated herein as VH), and a light (L) chain variable region (abbreviated herein as VL). In another example, an antibody comprises two heavy (H) chain variable regions and two light (L) chain variable regions or antibody binding fragments thereof. The light chains of the immunoglobulin may be of type kappa or lambda. In one embodiment, the antibody molecule is glycosylated. An antibody molecule can be functional for antibody dependent cytotoxicity and/or complement-mediated cytotoxicity, or may be non-functional for one or both of these activities. An antibody molecule can be an intact antibody or an antigen-binding fragment thereof.


Antibody molecules include “antigen-binding fragments” of a full length antibody, e.g., one or more fragments of a full-length antibody that retain the ability to specifically bind to an HA target of interest. Examples of binding fragments encompassed within the term “antigen-binding fragment” of a full length antibody include (i) a Fab fragment, a monovalent fragment consisting of the VL, VH, CL and CH1 domains; (ii) a F(ab′) or F(ab′)2 fragment, a bivalent fragment including two Fab fragments linked by a disulfide bridge at the hinge region; (iii) an Fd fragment consisting of the VH and CH1 domains; (iv) an Fv fragment consisting of the VL and VH domains of a single arm of an antibody, (v) a dAb fragment (Ward et al., (1989) Nature 341:544-546), which consists of a VH domain; and (vi) an isolated complementarity determining region (CDR) that retains functionality. Furthermore, although the two domains of the Fv fragment, VL and VH, are coded for by separate genes, they can be joined, using recombinant methods, by a synthetic linker that enables them to be made as a single protein chain in which the VL and VH regions pair to form monovalent molecules known as single chain Fv (scFv). See e.g., Bird et al. (1988) Science 242:423-426; and Huston et al. (1988) Proc. Natl. Acad. Sci. USA 85:5879-5883. Antibody molecules include diabodies.


As used herein, an antibody refers to a polypeptide, e.g., a tetrameric or single chain polypeptide, comprising the structural and functional characteristics, particularly the antigen binding characteristics, of an immunoglobulin. Typically, a human antibody comprises two identical light chains and two identical heavy chains. Each chain comprises a variable region.


The variable heavy (VH) and variable light (VL) regions can be further subdivided into regions of hypervariability, termed “complementarity determining regions” (“CDR”), interspersed with regions that are more conserved, termed “framework regions” (FR). Human antibodies have three VH CDRs and three VL CDRs, separated by framework regions FR1-FR4. The extent of the FRs and CDRs has been precisely defined (see, Kabat, E. A., et al. (1991) Sequences of Proteins of Immunological Interest, Fifth Edition, U.S. Department of Health and Human Services, NIH Publication No. 91-3242; and Chothia, C. et al. (1987) J. Mol. Biol. 196:901-917). Kabat definitions are used herein. Each VH and VL is typically composed of three CDRs and four FRs, arranged from amino-terminus to carboxyl-terminus in the following order: FR1, CDR1, FR2, CDR2, FR3, CDR3, FR4.


The heavy and light immunoglobulin chains can be connected by disulfide bonds. The heavy chain constant region typically comprises three constant domains, CH1, CH2 and CH3. The light chain constant region typically comprises a CL domain. The variable region of the heavy and light chains contains a binding domain that interacts with an antigen. The constant regions of the antibodies typically mediate the binding of the antibody to host tissues or factors, including various cells of the immune system (e.g., effector cells) and the first component (Clq) of the classical complement system.


The term “immunoglobulin” comprises various broad classes of polypeptides that can be distinguished biochemically. Those skilled in the art will appreciate that heavy chains are classified as gamma, mu, alpha, delta, or epsilon (γ, μ, α, δ, ε) with some subclasses among them (e.g., γ1-γ4). It is the nature of this chain that determines the “class” of the antibody as IgG, IgM, IgA IgD, or IgE, respectively. The immunoglobulin subclasses (isotypes) e.g., IgG1, IgG2, IgG3, IgG4, IgA1, etc. are well characterized and are known to confer functional specialization. Modified versions of each of these classes and isotypes are readily discernable to the skilled artisan in view of the instant disclosure and, accordingly, are within the scope of the instant disclosure. All immunoglobulin classes are clearly within the scope of the present disclosure. Light chains are classified as either kappa or lambda (κ, λ). Each heavy chain class may be bound with either a kappa or lambda light chain.


Suitable antibodies include, but are not limited to, monoclonal, monospecific, polyclonal, polyspecific, human antibodies, primatized antibodies, chimeric antibodies, bi-specific antibodies, humanized antibodies, conjugated antibodies (i.e., antibodies conjugated or fused to other proteins, radiolabels, cytotoxins), Small Modular ImmunoPharmaceuticals (“SMIPs™”), single chain antibodies, cameloid antibodies, and antibody fragments.


In some embodiments, an antibody is a humanized antibody. A humanized antibody refers to an immunoglobulin comprising a human framework region and one or more CDR's from a non-human, e.g., mouse or rat, immunoglobulin. The immunoglobulin providing the CDR's is often referred to as the “donor” and the human immunoglobulin providing the framework often called the “acceptor,” though in some embodiments, no source or no process limitation is implied. Typically a humanized antibody comprises a humanized light chain and a humanized heavy chain immunoglobulin.


An “immunoglobulin domain” refers to a domain from the variable or constant domain of immunoglobulin molecules. Immunoglobulin domains typically contain two β-sheets formed of about seven β-strands, and a conserved disulphide bond (see, e.g., A. F. Williams and A. N. Barclay (1988) Ann. Rev. Immunol. 6:381-405).


As used herein, an “immunoglobulin variable domain sequence” refers to an amino acid sequence that can form the structure of an immunoglobulin variable domain. For example, the sequence may include all or part of the amino acid sequence of a naturally-occurring variable domain. For example, the sequence may omit one, two or more N- or C-terminal amino acids, internal amino acids, may include one or more insertions or additional terminal amino acids, or may include other alterations. In one embodiment, a polypeptide that comprises an immunoglobulin variable domain sequence can associate with another immunoglobulin variable domain sequence to form a target binding structure (or “antigen binding site”), e.g., a structure that interacts with the target antigen.


As used herein, the term antibodies comprises intact monoclonal antibodies, polyclonal antibodies, single domain antibodies (e.g., shark single domain antibodies (e.g., IgNAR or fragments thereof)), multispecific antibodies (e.g., bi-specific antibodies) formed from at least two intact antibodies, and antibody fragments so long as they exhibit the desired biological activity. Antibodies for use herein may be of any type (e.g., IgA, IgD, IgE, IgG, or IgM).


The antibody or antibody molecule can be derived from a mammal, e.g., a rodent, e.g., a mouse or rat, horse, pig, or goat. In embodiments, an antibody or antibody molecule is produced using a recombinant cell. In some embodiments an antibody or antibody molecule is a chimeric antibody, for example, from mouse, rat, horse, pig, or other species, bearing human constant and/or variable regions domains.


A binding agent, as used herein, is an agent that bind, e.g., specifically binds, a target antigen, e.g., HA. Binding agents of the invention share sufficient structural relationship with anti-HA antibody molecules disclosed herein to support specific binding to HA, and in some embodiments, other functional properties of an anti-HA antibody molecule disclosed herein. In some embodiments, a binding agent will exhibit a binding affinity at of at least 10, 20, 30, 40, 50, 60, 70, 80, or 90% of an antibody molecule disclosed herein, e.g., an antibody molecule with which it shares, significant structural homology, e.g., CDR sequences. Binding agents can be naturally occurring, e.g., as are some antibodies, or synthetic. In an embodiment a binding agents is a polypeptide, e.g., an antibody molecule, e.g., an antibody. While some binding agents are antibody molecules, other molecules, e.g., other polypeptides, can also function as binding agents. Polypeptide binding agents can be monomeric or multimeric, e.g., dimeric, trimeric, or tetrameric and can be stabilized by intra- or interchain bonds, e.g., disulfide bonds. They can contain natural or non-naturally occurring amino acid residues. In some embodiments, binding agents are antibody molecules, or other polypeptides, that present one or more CDRs of antibody molecules disclosed herein or that otherwise mimic the structure of an antibody molecule disclosed herein. Binding agents can also comprise aptamers, nucleic acids or other molecular entities. A binding agent can be developed in a variety of ways, e.g., by immunization, by rational design, screening of random structures, or a combination of those or other approaches. Typically a binding agent will act by making contact with substantially the same epitope as an antibody molecule disclosed herein, e.g., an antibody molecule with which it shares, significant structural homology, e.g., CDR sequences. A binding agent can interact with amino acids, saccharides, or combinations thereof. Polypeptides other than antibodies can be used as a scaffold to present sequence, e.g., one or more, or a complete set of heavy chain and/or light chain CDRs, disclosed herein. Exemplary scaffolds include adnectin, zinc finger DNA-binding proteins. protein A, lipoclins, ankryin consensus repeat domain, thioredoxin, anticalins, centyrin, avimer domains, ubiquitin, peptidomimetics, stapled peptides, cystine-knot miniproteins, and IgNARs. In some embodiments, a binding agent is or comprises a nucleic acid, e.g., DNA, RNA or mixtures thereof. In some embodiments, a binding agent, e.g., a nucleic acid, shows secondary, tertiary, or quaternary structure. In some embodiments a binding agent, e.g., a nucleic acid, forms a structure that mimics the structure of an antibody molecule disclosed herein.


A broad spectrum binding agent, e.g., antibody molecule, as used herein, binds, a plurality of different HA molecules, and optionally neutralizes viruses comprising the different HA molecules. In an embodiment, it binds a first HA and binds a second HA from influenza A Group 1, and optionally neutralizes viruses comprising the first or second HA molecules. In an embodiment, it binds a first HA from an influenza A Group 1 virus, and binds a second HA from an influenza A Group 2 virus, and optionally neutralizes viruses comprising the different HA molecules. In an embodiment, it binds a first HA from an influenza A Group 1 or 2 virus and binds a HA from an influenza B virus, and optionally neutralizes viruses comprising the different HA molecules. In an embodiment, it binds, and in embodiments neutralizes, at least two different clades or clusters of virus, e.g., from different Groups. In some embodiments, it binds, and in some embodiments neutralizes, all or substantially all strains of Group 1 an/or Group 2 disclosed herein. In an embodiment, a binding agent, e.g., antibody molecule, binds, and in some embodiments, neutralizes: at least one strain from the Group 1 H1, e.g., H1a or H1b, cluster and at least one strain from the Group 2 H3 or H7 cluster. In an embodiment, a binding agent, e.g., antibody molecule, binds, and in some embodiments, neutralizes: at least one strain from the Group 1 H1, e.g., H1a or H1b, cluster and at least one influenza B strain. In an embodiment, a binding agent, e.g., antibody molecule, binds, and in some embodiments, neutralizes: at least one strain from the Group 2 H3 or H7 cluster and at least one influenza B strain. In an embodiment, a binding agent, e.g., antibody molecule, binds, and in some embodiments, neutralizes: at least one strain from the Group 1 H1, e.g., H1a or H1b, cluster, at least one strain from the Group 2 H3 or H7 cluster, and at least one influenza B strain. In some embodiments, binding agent, e.g., antibody molecule, binds, and optionally neutralizes or mediate infection of particular hosts, e.g., avian, camel, canine, cat, civet, equine, human, mouse, swine, tiger, or other mammal or bird.


The term “combination therapy”, as used herein, refers to administration of a plurality of agents, e.g., wherein at least one binding agent, e.g., antibody molecule, disclosed herein is administered to a subject, e.g., a human subject. The introduction of the agents into the subject can be at different times. In some embodiments, the agents are administered in overlapping regimens, or such that the subject is simultaneously exposed to both agents, or such that the response of the subject is better than would be seen with either agent administered alone.


As used herein, an “escape mutant” is a mutated influenza strain that is resistant to neutralization by an anti-HA antibody molecule described herein. In some embodiments, an escape mutant is resistant to neutralization with a binding agent, e.g., antibody molecule, but its parent strain is neutralized by the binding agent, e.g., antibody molecule. Resistance can be tested by various methods, including, but not limited to, genotypic testing (e.g., Sanger sequencing/nested PCR-baseline and last qPCR sample (Ct<32)), and phenotypic testing (e.g., plaque reduction on primary sample, e.g., ViroSpot™ assay (e.g., virus titration−last post-baseline≥2 Log10 TCID50/mL) or IC50 single passage sample (e.g., antibody titration−last post-baseline≥1 Log10 TCID50/mL).


As used herein, “pandemic influenza” refers to a new viral strain that arises due to human adaptation of an influenza strain by mutation or by emergence of a strain by reassortment of different strains of influenza A. The resulting pandemic strain is significantly different from previous strains and most people will have little or no pre-existing immunity. Symptoms and complications may be more severe and more frequent than those typical of seasonal influenza. Examples of past pandemic flu viruses include, e.g., the 2009 H1N1 ‘swine flu,’ the 1957-58 H2N2 ‘Asian flu’ and the 1968 H3N2 influenza strains.


The terms “purified” and “isolated” as used herein in the context of an antibody molecule, e.g., an antibody, or generally a polypeptide, obtained from a natural source, refers to a molecule which is substantially free of contaminating materials from the natural source, e.g., cellular materials from the natural source, e.g., cell debris, membranes, organelles, the bulk of the nucleic acids, or proteins, present in cells. Thus, a polypeptide, e.g., an antibody molecule, that is isolated includes preparations of a polypeptide having less than about 30%, 20%, 10%, 5%, 2%, or 1% (by dry weight) of cellular materials and/or contaminating materials. The terms “purified” and “isolated” when used in the context of a chemically synthesized species, e.g., an antibody molecule, refers to the species which is substantially free of chemical precursors or other chemicals which are involved in the syntheses of the molecule.


A preparation of binding agents, e.g., antibody molecules, as used herein, comprises a plurality of molecules of a binding agent, e.g., antibody molecule, described herein. In some embodiments, the binding agent, e.g., antibody molecule, makes up at least 60, 70, 80, 90, 95, 98, 99, 99.5 or 99.9%, of the preparation, or of the active ingredients of the preparation, by weight or number. In some embodiments, that binding agent is an antibody molecule which makes up at least 60, 70, 80, 90, 95, 98, 99, 99.5 or 99.9%, of the preparation, or of the active ingredients, or polypeptide ingredients, or antibody molecules, of the preparation, by weight or number. In some embodiments, the binding agent is an antibody molecule and the preparation contains no more than 30, 20, 10, 5, 2, 1, or 0.5%, by weight or number, of a contaminant, e.g., a reactant, solvent, precursor or other species, from the source, or used in the preparation, of the antibody molecule, e.g., a species from a cell, reaction mixture, or other system used to produce the antibody molecule.


As used herein, the term “prevent infection” means that a subject (e.g., a human) is less likely to be infected by influenza if the subject receives the antibody prior to (e.g., 1 day, 2 days, 1 week, 2 weeks, 3 weeks, or 1 month of more) before being exposed to influenza.


As used herein, “seasonal influenza” is a strain that is identical or closely related to strains that have been circulating in the human population in recent years and therefore most people are at least partially immune to it. Such a strain is not likely to cause severe disease. Symptoms can include fever, cough, runny nose, and muscle pain, and in rare cases, death can result from complications, such as pneumonia. Outbreaks follow predictable seasonal patterns, annually, and usually in fall and winter and in temperate climates. Infection due to seasonal influenza is commonly referred to as the flu.


As used herein, specific binding, means that a binding agent, e.g., an antibody molecule, binds its antigen with a KD of equal to or less than 10−5. In some embodiments, the antibody binds it's antigen with a KD of equal to or less than 10−6, 10−7, 10−8, 10−9, 10−10, 10−11, or 10−12.


As used herein, the term “therapeutically effective amount” refers to an amount of a therapeutic agent, e.g., a binding agent, e.g., an antibody molecule, which results in a positive outcome for the subject. In some embodiments, it can be statistically correlated with therapeutic effect or benefit, e.g., the lessening or prevention of a manifestation of an effect or a symptom, when administered to a population of subjects. In some embodiments, it is an amount that also provides a preselected, or reasonable, benefit/risk ratio. In some embodiments, it is an amount effective to reduce the incidence and/or severity of and/or to delay onset of one or more features, symptoms, or characteristics of a disease, disorder, or condition. A therapeutically effective amount is can be administered in a dosing regimen that may comprise one or multiple unit doses.


As used herein, the term “treat infection” means that a subject (e.g., a human) who has been infected with an influenza and experiences symptoms of the influenza (e.g., the flu), will in some embodiments, suffer less severe symptoms and/or will recover faster when the antibody molecule is administered than if the antibody is never administered. In some embodiments, when an infection is treated, an assay to detect virus in the subject will detect less virus after effective treatment for the infection. For example, a diagnostic assay using an antibody molecule, such as an antibody molecule described herein, will detect less or no virus in a biological sample of a patient after administration of an antibody molecule for the effective treatment of the viral infection. Other assays, such as PCR (e.g., qPCR) can also be used to monitor treatment in a patient, to detect the presence, e.g., decreased presence (or absence) after treatment of viral infection in the patient. Treatment can, e.g., partially or completely alleviate, ameliorate, relive, inhibit, reduce the severity of, and/or reduces incidence and optionally, delay onset of, one or more manifestations of the effects or symptoms, features, and/or causes of a particular disease, disorder, and/or condition (e.g., influenza). In some embodiments, treatment is of a subject who does not exhibit signs of the relevant disease, disorder and/or condition and/or of a subject who exhibits only early signs of the disease, disorder, and/or condition. In some embodiments, treatment is of a subject who exhibits one or more established signs of the relevant disease, disorder and/or condition. In some embodiments, treatment is of a subject diagnosed as suffering from influenza.


Calculations of “homology” or “sequence identity” or “identity” between two sequences (the terms are used interchangeably herein) can be performed as follows. The sequences are aligned for optimal comparison purposes (e.g., gaps can be introduced in one or both of a first and a second amino acid or nucleic acid sequence for optimal alignment and non-homologous sequences can be disregarded for comparison purposes). The optimal alignment is determined as the best score using the GAP program in the GCG software package with a Blossum 62 scoring matrix with a gap penalty of 12, a gap extend penalty of 4, and a frameshift gap penalty of 5. The amino acid residues or nucleotides at corresponding amino acid positions or nucleotide positions are then compared. When a position in the first sequence is occupied by the same amino acid residue or nucleotide as the corresponding position in the second sequence, then the molecules are identical at that position (as used herein amino acid or nucleic acid “identity” is equivalent to amino acid or nucleic acid “homology”). The percent identity between the two sequences is a function of the number of identical positions shared by the sequences.


Hemagglutinin (HA) Polypeptides and Influenza

Influenza viruses are negative sense, single-stranded, segmented RNA envelope viruses. Two glycoproteins, a hemagglutinin (HA) polypeptide and a neuraminidase (NA) polypeptide, are displayed on the outer surface of the viral envelope. There are several Influenza A subtypes, labeled according to an H number (for the type of hemagglutinin) and an N number (for the type of neuraminidase). There are 17 different H antigens (H1 to H17) and nine different N antigens (N1 to N9). Influenza strains are identified by a nomenclature based on the number of the strain's HA polypeptide and NA polypeptide subtypes, for example, H1N1, H1N2, H1N3, H1N4, H1N5, and the like.


HA is the major viral surface glycoprotein that mediates binding and entry of the virus into host cells and is a primary target of neutralizing antibody responses. HA is a trimer of three identical monomers. Each monomer is synthesized as a precursor, HA0, that is proteolytically processed into two disulfide-bonded polypeptide chains, HA1 and HA2. The ectodomain of this protein has (i) a globular head domain possessing receptor binding activity and major antigenic determinants, (ii) a hinge region, and (iii) a stem region where a sequence critical for fusion, the fusion peptide, is located. The viral replication cycle is initiated when the virion attaches via its surface hemagglutinin proteins to sialylated glycan receptors on the host cell and enters the cell by endocytosis. The acidic environment in the endosome induces conformational changes in HA that expose the fusion peptide hidden within the stem region of the trimer. The exposed fusion peptide mediates the fusion of the viral and target cell membranes resulting in the release of the viral ribonucleoprotein into the cell cytoplasm.


Influenza A hemagglutinin subtypes have been divided into two main groups and four smaller clades, and these are further divided into clusters. Group 1 influenza A strains are divided into 3 clades: (i) H8, H9 and H12 (“the H9 cluster”); (ii) H1, H2, H5, H6 and H17 (“the H1a cluster”); and (iii) H11, H13 and H16 (“the H1b cluster”). Group 2 strains are divided into 2 clades: (i) H3, H4 and H14 (“the H3 cluster”); and (ii) H7, H10 and H15 (“the H7 cluster”). The H1b and the H1a clusters are classified together as the H1 cluster. The different HA subtypes do not necessarily share strong amino acid sequence identity, but their overall 3D structures are similar.


Of the 17 HA polypeptide subtypes, only 3 (H1, H2 and H3) have adapted for human infection. These subtypes have in common an ability to bind alpha 2,6 sialylated glycans. In contrast, their avian counterparts preferentially bind to alpha 2,3 sialylated glycans. HA polypeptides that have adapted to infect humans (e.g., of HA polypeptides from the pandemic H1N1 (1918) and H3N2 (1967-68) influenza subtypes) have been characterized by an ability to preferentially bind to α2,6 sialylated glycans in comparison with their avian progenitors that preferentially bind to α2,3 sialylated glycans (see, e.g., Skehel & Wiley, Annu Rev Biochem, 69:531, 2000; Rogers, & Paulson, Virology, 127:361, 1983; Rogers et al., Nature, 304:76, 1983; Sauter et al., Biochemistry, 31:9609, 1992


Further, HA polypeptides that mediate infection of humans preferentially bind to umbrella topology glycans over cone topology glycans (see, e.g., U.S. 2011/0201547). Without wishing to be bound by any particular theory, it has been proposed that the ability to infect human hosts correlates less with binding to glycans of a particular linkage, and more with binding to glycans of a particular topology, even though cone-topology glycans may be α2,6 sialylated glycans. In has been demonstrated that HA polypeptides that mediate infection of humans bind to umbrella topology glycans, often showing preference for umbrella topology glycans over cone topology glycans (See, for example, U.S. Application Publication Nos. 2009/0269342, 2010/0061990, 2009/0081193, and 2008/0241918, and International Publication No. WO2008/073161).


Mature HA polypeptides include three domains, (i) a globular domain (a.k.a., the head domain) consists mainly of the HA1 peptide and contains the receptor (sialylated glycoproteins)-binding region, (ii) a stalk domain (HA1 and HA2) where the membrane fusion peptide resides, and (iii) a transmembrane domain (HA2) that anchors hemagglutinin to the viral envelope. A set of amino acids in the interface of the HA1 and HA2 peptides is highly conserved across all influenza subtypes. The HA1/HA2 membrane proximal region (MPER), including a canonical alpha-helix, is also highly conserved across influenza subtypes.


HA polypeptides interact with the surface of cells by binding to a glycoprotein receptor, known as the HA receptor. Binding of an HA polypeptide to an HA receptor is predominantly mediated by N-linked glycans on the HA receptors. HA polypeptides on the surface of flu virus particles recognize sialylated glycans that are associated with HA receptors on the surface of the cellular host. Following replication of viral proteins and genome by the cellular machinery, new viral particles bud from the host to infect neighboring cells.


Currently, vaccines are administered to subjects, e.g., humans, to prevent the flu, e.g., to prevent infection or to minimize the effects of an infection with influenza virus. Traditional vaccines contain a cocktail of antigens from various strains of influenza and are administered to humans to prevent the human from getting infected with the virus. HA is the main target of influenza A-neutralizing antibodies, and HA undergoes continuous evolution driven by the selective pressure of the antibody response, which is primarily directed against the membrane-distal receptor-binding subdomain of the HA polypeptide. The subject, however, is protected only from strains that are identical to, or closely related to, the strains from which the antigens in the cocktail were derived. The human is still most vulnerable to infection by other strains of the flu that were not included in the cocktail. One of the advantages of the antibodies provided herein is their ability to bind an epitope of HA that is conserved across multiple strains of influenza A, and in some embodiments, influenza B. Thus, administration of an anti-HA antibody described herein will be more effective to protect an individual from infection from a broader spectrum of influenza (e.g., influenza A and, in some embodiments, influenza B) and conditions associate thereof (e.g., secondary infections, e.g., secondary bacterial infections). Further, the antibodies are effective in treating a subject after infection has occurred.


Anti-HA Antibody Molecules

Binding agents, and in particular, the antibody molecules described herein, can bind to influenza A viruses from both Group 1 and Group 2, and in some embodiments also bind influenza B viruses. For example, the antibody molecules described herein can bind to an HA polypeptide on at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or 11 strains from Group 1, and can also bind to an HA polypeptide on at least 1, 2, 3, 4, 5, or 6 strains from Group 2. In another example, the antibody molecules described herein can bind to an HA polypeptide on an influenza strain from at least 1, 2 or 3 clades from Group 1, and can also bind to an HA polypeptide on an influenza strain from one or both clades of Group 2. The antibody molecules described herein inhibit cell entry and thus targeting an early step in the infection process.


The binding agents, and in particular, the antibody molecules featured in the disclosure, can be effective to treat or prevent infection by seasonal or pandemic influenza strains. The binding agents, and in particular the antibody molecules described herein, can be characterized by their ability to prevent or treat a Group 1 or a Group 2 strain of influenza A viruses or, in some embodiments, a strain of influenza B viruses. The binding agents, and in particular the antibody molecules featured in the disclosure, are effective to prevent or treat infection by one or more strains of Group 1, one or more strains of Group 2, and also one or more strains of influenza B viruses. In an embodiment, the binding agent is used to treat or prevent an influenza virus infection caused by an influenza virus chose from an H1N1 virus, an H3N2 virus, an H7N9 virus, or a combination thereof.


The binding agents, and in particular the antibody molecules can be effective to treat the infection when administered the same day as the subject is exposed, or when administered, e.g., 1 day, 2 days, 3 days, 4 days or later after infection, or upon a first symptom experienced by the patient. In an embodiment, the antibody molecule does not cause an antibody dependent enhancement (ADE) in the subject, e.g., as determined by a method described herein. In an embodiment, the antibody molecule does not cause viral resistance, e.g., as determined by a method described herein.


Strains


The antibody molecules described herein are effective to treat one or more influenza strains of Group 1, one or more influenza strains of Group 2, and also one or more influenza B strains, and specific isolates within these strains. Certain antibody molecules may be more effective for treatment of certain isolates than other isolates. Exemplary influenza strains and isolates are described in the below Table 1. Affinity can also be in reference to a particular isolate of a given Group 1 or Group 2 strain for influenza A viruses or a strain for influenza B viruses. Exemplary isolates are as provided in the above Table 1. Other exemplary influenza virus strains and isolates are also described herein, e.g., in FIG. 18.









TABLE 1







Exemplary Influenza Strains and Isolates










Type
Group
HA type
Isolate





A
1
H1N1
A/PR/8/34 (aka PR-8)





A/Solomon Islands/03/06





A/Solomon Islands/20/1999





A/California/07/2009





A/New Caledonia/20/99





A/Bangkok/10/83





A/Yamagata/120/86





A/Osaka/930/88





A/Suita/1/89





A/California/04/2009


A
1
H2N2
A/Okuda/57





A/Adachi/2/57





A/Kumamoto/1/65





A/Kaizuka/2/65





A/Izumi/5/65





A/Chicken/PA/2004


A
1
H5N1
A/Vietnam/1203/04





A/Duck/Singapore/3/97





A/Duck/MN/1525/81


A
1
H9N2
A/Hong Kong/1073/2004





A/Swine/Hong Kong/9/98





A/Guinea fowl/HK/WF10/99


A
1
H16N3
A/black headed gull/Mongolia/1756/2006


A
2
H3N2
X-31





A/Victoria/3/75





A/Wyoming/03/2003





A/Wisconsin/67/2005





A/Brisbane/10/2007





A/California/7/2004





A/New York/55/2004





A/Moscow/10/1999





A/Aichi/2/68





A/Beijing/32/92/X-117





A/Fukuoka/C29/85





A/Sichuan/2/87





A/Ibaraki/1/90





A/Suita/1/90





A/Perth/16/2009





A/Uruguay/716/2007





A/Fujian/411/2003





A/Panama/2007/99





A/Shangdong/09/93


A
2
H7N7
A/Netherlands/219/2003


B


B/Wisconsin/1/2010









Mechanisms of Inhibition


While not being limited by a specific mechanism, HA specific antibodies can inhibit infection by numerous methods, such as by blocking viral attachment to sialic acid residues on surface proteins on host cells, by interfering with the structural transition of HA that triggers fusion activity in the endosome, or by simultaneously inhibiting attachment and virus-cell fusion. In some embodiments, antibody molecules featured herein bind an epitope at the HA trimer interface. Structural changes at the trimer interface are important for fusion of the viral membrane and the endocytic membrane, and the antibody molecules described herein interfere with this critical step of infection. Assays to measure fusogenic activity of HA are known in the art. For example, one fusion assay measures syncytia formation, which occurs in cell-cell fusion events. Cells that express and display an influenza viral strain HA can be used in the assay. Membrane-anchored hemagglutinin in these cells is induced to convert to the fusion conformation by a brief (e.g., 3 minute) exposure to low pH (e.g., pH 5). A 2-3-hour incubation period follows to allow the cells to recover and fuse to form syncytia. A nuclear stain can be used to aid in the visualization of these fusion products, and their count is used as a gauge of fusion activity. A candidate anti-HA antibody can be added either before or after the low pH treatment to determine at which stage of the fusion process the antibody interferes.


Another type of fusion assay monitors content mixing. To measure content mixing, host cells (e.g., erythrocytes) are loaded with a dye (e.g., Lucifer yellow) to determine whether the contents of HA-bound host cells could be delivered to HA-expressing cells after exposure to fusion-inducing conditions (e.g., low pH, such as pH less than 6 or pH less than 5). If the dye fails to mix with the contents of the host cells, then the conclusion can be made that fusion is inhibited. See, e.g., Kemble et al., J. Virol. 66:4940-4950, 1992. In another example, a fusion assay is performed by monitoring lipid mixing. The lipid mixing assay can be performed by labeling host cells (e.g., erythrocytes) with a fluorescent dye (e.g., R18 (octadecylrhodamine)) or dye pairs (e.g., CPT-PC/DABS-PC) (for fluorescence resonance energy transfer), exposing the host cells and HA-expressing cells to fusion-inducing conditions, and assaying for fluorescence dequenching (FDQ). Lipid mixing leads to dilution of the label into the viral envelope and a consequent dequenching. A lag in dequenching or the absence of dequenching is indicative of membrane fusion inhibition. See, e.g., Kemble et al., J. Virol. 66:4940-4950, 1992; and Carr et al., Proc. Natl. Acad. Sci. 94:14306-14313, 1997.


Escape Mutants


In some embodiments, influenza strains will rarely if ever produce escape mutants when contacted with the featured antibody molecules. Escape mutants can be identified by methods known in the art. For example, an antibody featured in the disclosure will not produce an escape mutant when the cells are infected with the virus under prolonged or repeated exposure to anti-HA antibodies featured in the disclosure.


One exemplary method includes infection of cells (e.g. MDCK cells) with a fixed amount of influenza A viral particles in the presence of the antibody at a concentration known to attenuate infection rates by 50%. Viral progeny collected after each passaging is used to infect a fresh cell culture in the presence of the same or greater concentration of the antibody. After multiple cycles of infection, e.g., after 15 cycles, 12 cycles, 11 cycles, 10 cycles, 9 cycles, 8 cycles, 7 cycles, 6 cycles, or 5 cycles, of infection under these conditions, the HA nucleotide sequence extracted from 20 viral plaque picks is evaluated for enrichment for mutations that renders the viral isolate resistant to neutralization by the antibody (an escape mutant). If no mutants with reduced sensitivity to the antibody are detected after the multiple rounds of selection, e.g., after 11 rounds, 10 rounds, or 9 rounds of selection, the antibody is determined to be resistant to escape mutations (see, e.g., Throsby et al. (2008) PLoS One, volume 3, e3942).


In another example, an assay that measures minimum inhibitory concentration (MIC) of the neutralizing antibody can be used to identify escape mutants. The MIC of an antibody molecule is the lowest concentration of an antibody molecule that can be mixed with virus to prevent infection of cell culture with influenza. If escape mutants arise within a viral population, then the MIC of a particular antibody will be observed to increase with increased rounds of propagation under the antibody selective pressure, as the proportion of the viral particles that carry the resistance mutation within the population increased. Influenza escape mutants rarely if ever evolve in response to an anti-HA antibody molecule described herein, and therefore the MIC will stay the same over time.


Another assay suitable for monitoring for the development of escape mutants is a Cytopathic Effect (CPE) assay. A CPE assay monitors the ability of an antibody to neutralize (i.e., prevent infection by) an influenza strain. A CPE assay provides the minimal concentration of antibody required in cell culture to neutralize the virus. If escape mutants arise, than the CPE of a particular antibody will increase over time, as the antibody becomes less effective at neutralizing the virus. Viral strains rarely if ever produce escape mutants in response to an anti-HA antibody molecule described herein, and therefore the CPE will stay essentially the same over time.


Quantitative polymerase chain reaction (qPCR) can also be used to monitor for the development of escape mutants. qPCR is useful to monitor the ability of an antibody to neutralize (i.e., prevent infection by) an influenza strain. If an antibody effectively neutralizes a virus, then qPCR performed on cell culture samples will not detect presence of viral genomic nucleic acid. If escape mutants arise, than over time, qPCR will amplify more and more viral genomic nucleic acid. Escape mutants rarely if ever develop in response to an anti-HA antibody molecule described herein, and therefore qPCR will rarely if ever detect viral genomic nucleic acid, even after the passage of time.


Binding and Affinity


In some embodiments, the binding agents, particularly antibody molecules, featured herein bind to two or more of the following: at least one HA polypeptide from a Group 1 influenza strain (e.g., an H1, H2, H5, H6, H8, H9 H12, H11, H13, H16 or H17 polypeptide); at least one HA polypeptide from a Group 2 influenza strain (e.g., an H3, H4, H14, H7, H10, or H15 polypeptide); and at least one HA polypeptide from an influenza B strain. In an embodiment, a binding agent, e.g., an antibody molecule, has a KD for an HA from a Group 1 influenza strain (e.g., an H1, H2, H5, H6, H8, H9 H12, H11, H13, H16 or H17 polypeptide) of equal to or less than 10−6, 10−7, 10−8, 10−9, 10−10, 10−11, or 10−12. In an embodiment, a binding agent, e.g., an antibody molecule, has a KD for an HA from a Group 2 influenza strain (e.g., an H3, H4, H14, H7, H10, or H15 polypeptide) of equal to or less than 10−6, 10−7, 10−8, 10−9, 10−10, 10−11, or 10−12. In an embodiment, a binding agent, e.g., an antibody molecule, has a KD for an influenza B HA of equal to or less than 10−6, 10−7, 10−8, 10−9, 10−10, 10−11, or 10−12. In an embodiment, a binding agent, e.g., an antibody molecule, has: a) a first KD (representing an affinity for an HA from a Group 1 influenza strain, e.g., an H1, H2, H5, H6, H8, H9 H12, H11, H13, H16 or H17 polypeptide); and b) a second KD (representing an affinity for an HA from a Group 2 influenza strain, e.g., an H3, H4, H14, H7, H10, or H15 polypeptide), wherein the first and second KD are one or both of: both equal to or less than 10−8; and within 10 or 100 fold of each other;


In an embodiment, a binding agent, e.g., an antibody molecule, has a) a first KD (representing an affinity for an H1, e.g., the H1 from an H1N1 strain, e.g., A/South Carolina/1/1918, A/Puerto Rico/08/1934, or A/California/04/2009, or an H5N1 strain, e.g., A/Indonesia/5/2005 or A/Vietnam/1203/2004); and b) a second KD (representing an affinity for an H3 polypeptide, e.g., the H3 from an H3N2 strain, e.g., A/Brisbane/59/2007), wherein the first and second KD are one or both of: both equal to or less than 10−8; and within 10 or 100 fold of each other. In an embodiment, a binding agent, e.g., an antibody molecule, has: a) a first KD (representing an affinity for an H1, e.g., the H1 from an H1N1 strain, e.g., A/South Carolina/I/1918, A/Puerto Rico/08/1934, or A/California/04/2009, or an H5N1 strain, e.g., A/Indonesia/5/2005 or A/Vietnam/1203/2004); and b) a second KD (representing an affinity for an H3 polypeptide, e.g., the H3 from an H3N2 strain, e.g., A/Brisbane/59/2007), wherein the first and second KD are one or both of: both equal to or less than 10−8; and within 10 or 100 fold of each other.


In an embodiment, a binding agent, e.g., an antibody molecule, has: a) a first KD (representing an affinity for an HA from a Group 1 influenza strain, e.g., an H1, H2, H5, H6, H8, H9 H12, H11, H13, H16 or H17 polypeptide and/or an affinity for an HA from a Group 2 influenza strain, e.g., an H3, H4, H14, H7, H10, or H15 polypeptide); and b) a second KD (representing an affinity for an influenza B HA, e.g., from B/Wisconsin/1/2010); wherein the first and second KD are one or both of: both equal to or less than 10−8; and within 10 or 100 fold of each other. In an embodiment, a binding agent, e.g., an antibody molecule, has: a) a first KD (representing an affinity for an HA from a Group 1 influenza strain, e.g., an H1, e.g., the H1 from an H1N1 strain, e.g., A/South Carolina/I/1918, A/Puerto Rico/08/1934, or A/California/04/2009, or an H5N1 strain, e.g., A/Indonesia/5/2005 or A/Vietnam/1203/2004, and/or an affinity for an HA from a Group 2 influenza strain, e.g., an H3 polypeptide, from an H3N2 strain, e.g., from A/Brisbane/59/2007); and b) a second KD (an affinity for an influenza B HA); wherein the first and second KD are: one or both of: both equal to or less than 10−8; and within 10 or 100 fold of each other.


In one embodiment, the antibody molecule binds to at least one HA polypeptide from a Group 1 influenza strain with a higher affinity than a reference anti-HA antibody, and to at least one HA polypeptide from a Group 2 influenza strain with a higher affinity than a reference anti-HA antibody. In another embodiment, the antibody molecule binds to at least one HA polypeptide from an influenza A strain with a higher affinity than a reference anti-HA antibody, and to at least one HA polypeptide from an influenza B strain with a higher affinity than a reference anti-HA antibody. Exemplary reference HA antibodies include Ab 67-11 (U.S. Provisional application No. 61/645,453, filed on the same date as the present application), FI6 (F16, as used herein, refers to any specifically disclosed FI6 sequence in U.S. Application Publication No. 2010/0080813, US Application Publication No. 2011/0274702, International Publication No. WO2013/011347 or Corti et al., Science 333:850-856, 2011, published online Jul. 28, 2011; FIGS. 12A to 12C of International Publication No. WO2013/170139 or U.S. Application Publication No. 2013/0302349), F128 (U.S. Application Publication No. 2010/0080813), and C179 (Okuno et al., J. Virol. 67:2552-1558, 1993), F10 (Sui et al., Nat. Struct. Mol. Biol. 16:265, 2009), CR9114 (Dreyfus et al., Science. 2012; 337(6100):1343-1348; published online Aug. 9, 2012), and CR6261 (Ekiert et al., Science 324:246-251, 2009; published online Feb. 26, 2009).


Affinity, or relative affinity or aviditiy, can be measured by methods known in the art, such as by ELISA assay (Enzyme Linked Immunosorbent Assay), Surface Plasmon Resonance (SPR, e.g., by a Biacore™ Assay), or KinExA® assay (Sapidyne, Inc.). Relative binding affinity is expressed herein according to ELISA assay. As used herein, an anti-HA antibody that binds with “high affinity” to a Group 1 HA, to a Group 2 HA, and to an influenza B HA, can bind a Group 1 HA with a Kd less than or equal to 200 pM, e.g., less than or equal to 100 pM, as measured by ELISA, can bind a Group 2 HA with a Kd less than or equal to 200 pM, e.g., less than or equal to 100 pM, as measured by ELISA, and can bind an influenza B HA with a Kd less than or equal to 200 pM, e.g., less than or equal to 100 pM, as measured by ELISA.


Exemplary Anti-HA Antibody Molecules


Provided herein are antibodies that have one or more CDR sequences and one or more framework (FR) sequences as shown in Table 2.









TABLE 2







Heavy and Light Chain CDR and 


FR Sequences for Anti-HA Antibodies









CDR/FR

SEQ


Region
Amino Acid Sequence
ID NO:












HC CDR1
[S/T]Y[A/G]MH
1





HC CDR2
V[I/V/L]S[Y/F]DG[S/N][Y/N]
2



[K/R]YYADSVQG






HC CDR3
D[S/T][R/K/Q]LR[S/T]LLYFEWLS
3



[Q/S]G[Y/L/V][F/L][N/D][P/Y]






LC CDR1
Q[S/T][V/L/I][T/S][Y/F/W]
4



[N/S/D]YKNYLA






LC CDR1
Q[S/T][V/L/I][T/S][Y/F/W]
170



[N/S/D/Q/R/E]YKNYLA






LC CDR2
W[A/G]S[T/A/Y/H/K/D]
5



[R/L]E[S/T]






LC CDR3
QQ[Y/H]YRTPP[T/S]
6





HC FR1
[E/Q]VQLLE[S/T]GGGLVKPGQSLKL
7



SCAASGFTF[S/T]






HC FR2
WVRQPPGKGLEWVA
8





HC FR3
RFTISRDNSKNTLYLQMNSLRAEDTAVY
9



YCAK






HC FR4
WG[A/Q]G[T/A][T/M][L/V]TVSS
10





LC FR1
[E/D]I[V/Q]MTQSP[D/S][S/T]
11



[L/V][A/S][V/A][S/T][L/V/R]




G[E/D]R[A/V][T/S]I




[N/T/Q/D/R/]C[K/R]SS






LC FR2
WYQQKPG[Q/K][P/A]PKLLIY
12





LC FR3
GVP[D/E/S]RFSGSGSGTDFTLTISS
13



LQ[A/P]ED[V/F/K/D]A[V/T]YYC






LC FR4
FG[G/Q/T/S/N]GTK[L/V][D/E]IK
14









In one embodiment, the anti-HA antibody comprises a heavy chain and/or a light chain as defined in Table 3 below. The amino acid sequences of the variable heavy and light chains of Table 3 are provided in FIGS. 2 and 3, respectively, or in FIG. 17, of International Publication No. WO2013/170139 or U.S. Application Publication No. 2013/0302349.









TABLE 3







Heavy and Light Chain Amino Acid Sequence


Designations for Anti-HA Antibodies













Antibody
HC
SEQ ID NO:
LC
SEQ ID NO:
















1.
Ab A18
15
15
28
28


2.
Ab 014
16
16
29
29


3.
Ab 028
16
16
30
30


4.
Ab 001
17
17
31
31


5.
Ab 002
18
18
31
31


6.
Ab 003
19
19
31
31


7.
Ab 009
17
17
32
32


8.
Ab 010
18
18
32
32


9.
Ab 011
19
19
32
32


10.
Ab 017
17
17
33
33


11.
Ab B18
18
18
33
33


12.
Ab 019
19
19
33
33


13.
Ab 025
17
17
34
34


14.
Ab 026
18
18
34
34


15.
Ab 027
19
19
34
34


16.
Ab 086
20
20
34
34


17.
Ab 154
21
21
29
29


18.
Ab 155
21
21
30
30


19.
Ab 157
22
22
29
29


20.
Ab 159
22
22
35
35


21.
Ab 160
17
17
36
36


22.
Ab 186
17
17
37
37


23.
Ab 187
17
17
38
38


24.
Ab 188
17
17
39
39


25.
Ab 189
17
17
40
40


26.
Ab 190
17
17
41
41


27.
Ab 191
17
17
42
42


28.
Ab 192
17
17
43
43


29.
Ab 193
17
17
44
44


30.
Ab 194
19
19
37
37


31.
Ab 195
19
19
38
38


32.
Ab 196
19
19
39
39


33.
Ab 197
19
19
40
40


34.
Ab 198
19
19
41
41


35.
Ab 199
19
19
42
42


36.
Ab 200
19
19
43
43


37.
Ab 202
17
17
45
45


38.
Ab 203
18
18
45
45


39.
Ab 204
19
19
45
45


40.
Ab 210
23
23
45
45


41.
Ab 211
17
17
46
46


42.
Ab 212
18
18
46
46


43.
Ab 213
19
19
46
46


44.
Ab 219
23
23
46
46


45.
Ab A001
24
24
47
47


46.
Ab A002
24
24
48
48


47.
Ab A003
24
24
49
49


48.
Ab 004
25
25
47
47


49.
Ab 005
25
25
48
48


50.
Ab 006
25
25
49
49


51.
Ab 007
26
26
47
47


52.
Ab 008
26
26
48
48


53.
Ab A009
26
26
49
49


54.
Ab A010
24
24
50
50


55.
Ab A011
24
24
51
51


56.
Ab 012
25
25
50
50


57.
Ab 013
25
25
51
51


58.
Ab A14
26
26
50
50


59.
Ab 015
26
26
51
51


60.
Ab 016
27
27
47
47


61.
Ab A017
27
27
48
48


62.
Ab C18
27
27
49
49


63.
Ab A019
27
27
50
50


64.
Ab 031
24
24
45
45


65.
Ab 032
25
25
45
45


66.
Ab 033
26
26
45
45


67.
Ab 034
27
27
45
45


68.
Ab 037
24
24
46
46


69.
Ab 038
25
25
46
46


70.
Ab 039
26
26
46
46


71.
Ab 040
27
27
46
46


72.
Ab 043
25
25
60
60


73.
Ab 044
25
25
52
52


74.
Ab 045
25
25
57
57


75.
Ab 046
25
25
59
59


76.
Ab 047
25
25
55
55


77.
Ab 048
25
25
58
58


78.
Ab 049
25
25
54
54


79.
Ab 050
25
25
56
56


80.
Ab 051
25
25
53
53


81.
Ab 052
25
25
61
61


82.
Ab 067
25
25
153
153


83.
Ab 068
25
25
154
154


84.
Ab 069
25
25
155
155


85.
Ab 070
25
25
156
156


86.
Ab 071
162
162
52
52


87.
Ab 072
163
163
52
52


88.
Ab 073
25
25
165
165


89.
Ab 074
25
25
166
166


90.
Ab 075
25
25
167
167


91.
Ab 076
25
25
168
168


92.
Ab 077
25
25
169
169


93.
Ab 078
164
164
52
52


94.
Ab 079
164
164
155
155


95.
Ab 080
164
164
166
166


96.
Ab 081
164
164
169
169









In one embodiment, the anti-HA antibody comprises a heavy chain as defined in Table 4A below, and/or a light chain as defined in Table 4A below.









TABLE 4A







Heavy and Light Chain Amino Acid Sequence Designations












HC
SEQ ID NO:
LC
SEQ ID NO:
















15
15
28
28



16
16
29
29



17
17
30
30



18
18
35
35



19
19
31
31



21
21
32
32



22
22
33
33



20
20
34
34



23
23
36
36



24
24
45
45



25
25
46
46



26
26
37
37



27
27
38
38



Hc consensus
161
39
39



(HC161)



162
162
40
40



163
163
41
41



164
164
42
42





43
43





44
44





47
47





48
48





49
49





50
50





51
51





52
52





53
53





54
54





55
55





56
56





57
57





58
58





59
59





60
60





61
61





153
153





154
154





155
155





156
156





LC
62





consensus





(LC62)





165
165





166
166





167
167





168
168





169
169










In one embodiment, an antibody featured in the disclosure comprises a heavy chain sequence as defined in Table 4A and a light chain sequence as defined in Table 4A.


In one embodiment, an antibody featured in the disclosure comprises a heavy chain sequence as defined herein, e.g., in Table 4A, where a dipeptide is fused to the N-terminus. Typically, the dipeptide is isoleucine-aspartic acid (Ile-Asp). In another embodiment, an antibody featured in the disclosure comprises a light chain sequence as defined herein, e.g., in Table 4A, where a dipeptide is fused to the N-terminus. Typically, the dipeptide is Ile-Asp. In yet another embodiment, an antibody featured in the disclosure comprises a heavy chain comprising an N-terminal Ile-Asp dipeptide and a light chain comprising an Ile-Asp dipeptide. In the propeptide sequence of the heavy chain or light chain polypeptide, the Ile-Asp dipeptide occurs between the signal sequence and FR1. Heavy chain and light chain variable sequences comprising an Ile-Asp dipeptide at the N-terminus are identified in Table 4B.









TABLE 4B







Heavy and Light Chain Amino Acid Sequence Designations, where


the Sequence Includes an N-terminal Ile-Asp Dipeptide










HC
SEQ ID NO:
LC
SEQ ID NO:













15-ID
96
28-ID
110


16-ID
97
29-ID
111


17-ID
98
30-ID
112


18-ID
99
35-ID
113


19-ID
100
31-ID
114


21-ID
101
32-ID
115


22-ID
102
33-ID
116


20-ID
103
34-ID
117


23-ID
104
36-ID
118


24-ID
105
45-ID
119


25-ID
106
46-ID
120


26-ID
107
37-ID
121


27-ID
108
38-ID
122


Hc consensus ID
109
39-ID
123


(161-ID) 




40-ID
124




41-ID
125




42-ID
126




43-ID
127




44-ID
128




47-ID
129




48-ID
130




49-ID
131




50-ID
132




51-ID
133




52-ID
134




53-ID
135




54-ID
136




55-ID
137




56-ID
138




57-ID
139




58-ID
140




59-ID
141




60-ID
142




61ID
143




153-ID 
157




154-ID 
158




155-ID 
159




156-ID 
160




LC consensus ID
144




(62-ID)









In another embodiment, an antibody featured in the disclosure is other than an antibody known in the art. For example, the antibody is not Ab 67-11 (U.S. Provisional application No. 61/645,453) F16 (FI6, as used herein, refers to any specifically disclosed FI6 sequence in U.S. Application Publication No. 2010/0080813, US Application Publication No. 2011/0274702, International Publication No. WO2013/011347 or Corti et al., Science 333:850-856, 2011, published online Jul. 28, 2011; FIGS. 12A to 12C of International Publication No. WO2013/170139 or U.S. Application Publication No. 2013/0302349), FI28 (U.S. Application Publication No. 2010/0080813), and C179 (Okuno et al., J. Virol. 67:2552-1558, 1993), F10 (Sui et al., Nat. Struct. Mol. Biol. 16:265, 2009), CR9114 (Dreyfus et al., Science. 2012; 337(6100):1343-1348; published online Aug. 9, 2012), and CR6261 (Ekiert et al., Science 324:246-251, 2009; published online Feb. 26, 2009). In one embodiment, an antibody featured in the disclosure is other than Ab 67-11 (U.S. Provisional application No. 61/645,453, filed on the same date as the present application).


Variants


In an embodiment, an antibody molecule, e.g., an antibody featured in the disclosure has a variable heavy chain immunoglobulin domain that is at least 85%, 87%, 88%, 89%, 90%, 92%, 94%, 95%, 96%, 97%, 98%, or 99% homologous, or at least 85%, 87%, 88%, 89%, 90%, 92%, 94%, 95%, 96%, 97%, 98%, or 99% identical, to a heavy chain disclosed herein, e.g., from Table 3, Table 4A, or Table 4B, or FIG. 2, FIG. 13 or FIG. 17, of International Publication No. WO2013/170139 or U.S. Application Publication No. 2013/0302349, e.g. consensus sequence of SEQ ID NO: 161, and has a variable light chain immunoglobulin domain that is at least 85%, 87%, 88%, 89%, 90%, 92%, 94%, 95%, 96%, 97%, 98%, or 99% homologous, or at least 85%, 87%, 88%, 89%, 90%, 92%, 94%, 95%, 96%, 97%, 98%, or 99% identical, to a light chain disclosed herein, e.g., from Table 3, Table 4A, or Table 4B, or FIG. 3, FIG. 14 or FIG. 17, of International Publication No. WO2013/170139 or U.S. Application Publication No. 2013/0302349, e.g., the consensus sequence of SEQ ID NO: 62. The consensus sequences were determined through the analysis of biochemical and biophysical properties of several hundred computationally designed VH/VL combinations. The consensus sequences represent the amino acid sequences in which each amino acid is the one that occurs most frequently at that site when multiple sequences comprising desirable biochemical and biophysical data are aligned.


An exemplary anti-HA binding antibody has one or more CDRs, e.g., all three HC CDRs and/or all three LC CDRs of a particular antibody disclosed herein, or CDRs that are, in sum, at least 85%, 87%, 88%, 89%, 90%, 92%, 94%, 95%, 96%, 97%, 98%, or 99% homologous, or at least 85%, 87%, 88%, 89%, 90%, 92%, 94%, 95%, 96%, 97%, 98%, or 99% identical, to such an antibody. In one embodiment, the H1 and H2 hypervariable loops have the same canonical structure as those of an antibody described herein. In one embodiment, the L1 and L2 hypervariable loops have the same canonical structure as those of an antibody described herein.


In one embodiment, the amino acid sequence of the HC and/or LC variable domain sequence is at least 85%, 87%, 88%, 89%, 90%, 92%, 94%, 95%, 96%, 97%, 98%, or 99% homologous, or at least 85%, 87%, 88%, 89%, 90%, 92%, 94%, 95%, 96%, 97%, 98%, or 99% identical, to the amino acid sequence of the HC and/or LC variable domain of an antibody described herein. The amino acid sequence of the HC and/or LC variable domain sequence can differ by at least one amino acid, but no more than ten, eight, six, five, four, three, or two amino acids from the corresponding sequence of an antibody described herein. For example, the differences may be primarily or entirely in the framework regions.


In certain embodiments, the amino acid differences are conservative amino acid differences (e.g., conservative amino acid substitutions). A “conservative” amino acid substitution is one in which the amino acid residue is replaced with an amino acid residue comprising a similar side chain. Families of amino acid residues comprising similar side chains have been defined in the art. These families include, e.g., amino acids with basic side chains (e.g., lysine, arginine, histidine), acidic side chains (e.g., aspartic acid, glutamic acid), uncharged polar side chains (e.g., glycine, asparagine, glutamine, serine, threonine, tyrosine, cysteine), nonpolar side chains (e.g., alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, tryptophan), beta-branched side chains (e.g., threonine, valine, isoleucine) and aromatic side chains (e.g., tyrosine, phenylalanine, tryptophan, histidine).


The amino acid sequences of the HC and LC variable domain sequences can be encoded by a nucleic acid sequence that hybridizes under high stringency conditions to a nucleic acid sequence described herein or one that encodes a variable domain or an amino acid sequence described herein. In one embodiment, the amino acid sequences of one or more framework regions (e.g., FR1, FR2, FR3, and/or FR4) of the HC and/or LC variable domain are at least 85%, 87%, 88%, 89%, 90%, 92%, 94%, 95%, 96%, 97%, 98%, or 99% homologous, or at least 85%, 87%, 88%, 89%, 90%, 92%, 94%, 95%, 96%, 97%, 98%, or 99% identical, to corresponding framework regions of the HC and LC variable domains of an antibody described herein. In one embodiment, one or more heavy or light chain framework regions (e.g., HC FR1, FR2, and FR3) are at least 85%, 87%, 88%, 89%, 90%, 92%, 94%, 95%, 96%, 97%, 98%, or 99% homologous, or at least 85%, 87%, 88%, 89%, 90%, 92%, 94%, 95%, 96%, 97%, 98%, or 99% identical, to the sequence of corresponding framework regions from a human germline antibody.


Validation of Epitopes


In one embodiment, the antibodies featured in the disclosure are useful for validating a vaccine based on a particular epitope. For example, an epitope that is the target of an antibody featured in the disclosure can be assessed by computation methods to identify a peptide framework suitable for supporting the epitope conformation, such as to stabilize an epitope that is transient or minimally accessible in nature. Computational abstraction of the epitope and framework properties allows automated screening of databases to identify candidate acceptor peptide scaffolds. The acceptor scaffold can have a particular tertiary structure that includes, for example, one or more of a beta sheet, a beta sandwich, a loop, or an alpha or beta helix. The candidate epitope-scaffold antigens can be assayed in vitro, such as to identify binding properties with an antibody featured in the disclosure, e.g., binding affinity or structure analysis of the epitope-scaffold/antibody complex, or in vitro neutralization. The ability of the epitope-scaffold to generate an immune response (e.g., to generate antibodies) can be tested by administering the epitope-scaffold to an animal (e.g., in a mammal, such as a rat, a mouse, a guinea pig, or a rabbit), and then testing sera for the presence of anti-epitope-scaffold antibodies, e.g., by ELISA assay. The ability of the epitope-scaffold to elicit protection against infection by an influenza A Group 1 or Group 2 strain, or by both types of influenza strains, or an influenza B strain, can be assessed in vivo, such as in an animal (e.g., in a mammal). Thus, an antibody featured in the disclosure can provide validation that the epitope is functionally important and that targeting the epitope will provide protection from infection with a Group 1 or Group 2 influenza strain, or both types of strains, or an influenza B strain.


Production of Antibody Molecules


The nucleic acids (e.g., the genes) encoding an antibody molecule generated by a method described herein can be sequenced, and all or part of the nucleic acids can be cloned into a vector that expresses all or part of the nucleic acids. For example, the nucleic acids can include a fragment of the gene encoding the antibody, such as a single chain antibody (scFv), a F(ab′)2 fragment, a Fab fragment, or an Fd fragment. The disclosure also provides host cells comprising the nucleic acids encoding an antibody or fragment thereof as described herein. The host cells can be, for example, prokaryotic or eukaryotic cells, e.g., mammalian cells, or yeast cells, e.g., Pichia (see, e.g., Powers et al. (2001) J. Immunol. Methods 251:123-35), Hanseula, or Saccharomyces.


Antibody molecules, particularly full length antibody molecules, e.g., IgGs, can be produced in mammalian cells. Exemplary mammalian host cells for recombinant expression include Chinese Hamster Ovary (CHO) cells (including dhfr CHO cells, described in Urlaub and Chasin (1980) Proc. Natl. Acad. Sci. USA 77:4216-4220, used with a DHFR selectable marker, e.g., as described in Kaufman and Sharp (1982) Mol. Biol. 159:601-621), lymphocytic cell lines, e.g., NS0 myeloma cells and SP2 cells, COS cells, K562, and a cell from a transgenic animal, e.g., a transgenic mammal. For example, the cell is a mammary epithelial cell. In addition to the nucleic acid sequence encoding the immunoglobulin domain, the recombinant expression vectors may carry additional nucleic acid sequences, such as sequences that regulate replication of the vector in host cells (e.g., origins of replication) and selectable marker genes. The selectable marker gene facilitates selection of host cells into which the vector has been introduced (see e.g., U.S. Pat. Nos. 4,399,216; 4,634,665; and 5,179,017). Exemplary selectable marker genes include the dihydrofolate reductase (DHFR) gene (for use in dhfr host cells with methotrexate selection/amplification) and the neo gene (for G418 selection).


In an exemplary system for recombinant expression of an antibody molecule (e.g., a full length antibody or an antigen-binding portion thereof), a recombinant expression vector encoding both the antibody heavy chain and the antibody light chain is introduced into dhfr− CHO cells by calcium phosphate-mediated transfection. Within the recombinant expression vector, the antibody heavy and light chain genes are each operatively linked to enhancer/promoter regulatory elements (e.g., derived from SV40, CMV, adenovirus and the like, such as a CMV enhancer/AdMLP promoter regulatory element or an SV40 enhancer/AdMLP promoter regulatory element) to drive high levels of transcription of the genes. The recombinant expression vector also carries a DHFR gene, which allows for selection of CHO cells that have been transfected with the vector using methotrexate selection/amplification. The selected transformant host cells are cultured to allow for expression of the antibody heavy and light chains and intact antibody molecule is recovered from the culture medium. Standard molecular biology techniques are used to prepare the recombinant expression vector, to transfect the host cells, to select for transformants, to culture the host cells, and to recover the antibody from the culture medium. For example, some antibodies can be isolated by affinity chromatography with a Protein A or Protein G. For example, purified antibodies can be concentrated to about 100 mg/mL to about 200 mg/mL using protein concentration techniques that are known in the art.


Antibody molecules can also be produced by a transgenic animal. For example, U.S. Pat. No. 5,849,992 describes a method for expressing an antibody molecule in the mammary gland of a transgenic mammal. A transgene is constructed that includes a milk-specific promoter and nucleic acid sequences encoding the antibody molecule of interest, e.g., an antibody described herein, and a signal sequence for secretion. The milk produced by females of such transgenic mammals includes, secreted therein, the antibody of interest, e.g., an antibody described herein. The antibody molecule can be purified from the milk, or for some applications, used directly. Antibody molecules can also be expressed in vivo, following administration of a vector containing nucleic acids encoding the antibody heavy chain and the antibody light chain. Vector mediated gene-transfer is then used to engineer secretion of the anti-HA antibody into circulation. For example, an anti-HA antibody heavy chain and an anti-HA antibody light chain as described herein are cloned into an adeno-associated virus (AAV)-based vector, and each of the anti-HA antibody heavy chain and the anti-HA antibody light chain are under control of a promoter, such as a cytomegalovirus (CMV) promoter. Administration of the vector to a subject, such as to a patient, e.g., a human patient, such as by intramuscular injection, results in expression of an anti-HA antibody, and secretion into the circulation.


Modifications of Binding Agents


Binding, agents, e.g., antibody molecules can be modified to have numerous properties, e.g., to have altered, e.g., extended half-life, to be associated with, e.g., covalently bound to detectable moieties, e.g., labels, to be associated with, e.g., covalently bound to toxins, or to have other properties, e.g., altered immune functions. Antibody molecules may include modifications, e.g., modifications that alter Fc function, e.g., to decrease or remove interaction with an Fc receptor or with C1q, or both. In one example, the human IgG1 constant region can be mutated at one or more residues.


For some antibody molecules that include an Fc domain, the antibody production system may be designed to synthesize antibody molecules in which the Fc region is glycosylated. The Fc domain can be produced in a mammalian expression system that appropriately glycosylates the residue corresponding to asparagine 297. The Fc domain can also include other eukaryotic post-translational modifications. Other suitable Fc domain modifications include those described in WO2004/029207. For example, the Fc domain can be an XmAb® Fc (Xencor, Monrovia, Calif.). The Fc domain, or a fragment thereof, can have a substitution in an Fcγ Receptor (FcγR) binding region, such as the domains and fragments described in WO05/063815. In some embodiments, the Fc domain, or a fragment thereof, has a substitution in a neonatal Fc Receptor (FcRn) binding region, such as the domains and fragments described in WO05047327. In other embodiments, the Fc domain is a single chain, or fragment thereof, or modified version thereof, such as those described in WO2008143954. Other suitable Fc modifications are known and described in the art.


Antibody molecules can be modified, e.g., with a moiety that improves its stabilization and/or retention in circulation, e.g., in blood, serum, lymph, bronchoalveolar lavage, or other tissues, e.g., by at least 1.5, 2, 5, 10, or 50 fold. For example, an antibody molecule generated by a method described herein can be associated with a polymer, e.g., a substantially non-antigenic polymer, such as a polyalkylene oxide or a polyethylene oxide. Suitable polymers will vary substantially by weight. Polymers comprising molecular number average weights ranging from about 200 to about 35,000 daltons (or about 1,000 to about 15,000, and 2,000 to about 12,500) can be used.


For example, an antibody molecule generated by a method described herein can be conjugated to a water soluble polymer, e.g., a hydrophilic polyvinyl polymer, e.g. polyvinylalcohol or polyvinylpyrrolidone. A non-limiting list of such polymers include polyalkylene oxide homopolymers such as polyethylene glycol (PEG) or polypropylene glycols, polyoxyethylenated polyols, copolymers thereof and block copolymers thereof, provided that the water solubility of the block copolymers is maintained. Additional useful polymers include polyoxyalkylenes such as polyoxyethylene, polyoxypropylene, and block copolymers of polyoxyethylene and polyoxypropylene (Pluronics); polymethacrylates; carbomers; branched or unbranched polysaccharides that comprise the saccharide monomers D-mannose, D- and L-galactose, fucose, fructose, D-xylose, L-arabinose, D-glucuronic acid, sialic acid, D-galacturonic acid, D-mannuronic acid (e.g. polymannuronic acid, or alginic acid), D-glucosamine, D-galactosamine, D-glucose and neuraminic acid including homopolysaccharides and heteropolysaccharides such as lactose, amylopectin, starch, hydroxyethyl starch, amylose, dextrane sulfate, dextran, dextrins, glycogen, or the polysaccharide subunit of acid mucopolysaccharides, e.g. hyaluronic acid; polymers of sugar alcohols such as polysorbitol and polymannitol; heparin or heparan.


Binding agents, e.g., antibody molecules, as disclosed herein, can by conjugated to another entity or moiety (e.g., to a cytotoxic or cytostatic moiety, a label or detectable moiety, or a therapeutic moiety). Exemplary moieties include: a cytotoxic or cytostatic agent, e.g., a therapeutic agent, a drug, a compound emitting radiation, molecules of plant, fungal, or bacterial origin, or a biological protein (e.g., a protein toxin) or particle (e.g., a recombinant viral particle, e.g., via a viral coat protein), a detectable agent; a pharmaceutical agent, and/or a protein or peptide that can mediate association of the antibody or antibody portion with another molecule (such as a streptavidin core region or a polyhistidine tag). A binding agent, e.g., an antibody molecule, as disclosed herein, can be functionally linked by any suitable method (e.g., chemical coupling, genetic fusion, covalent binding, noncovalent association or otherwise) to one or more other molecular entities.


Binding agents, e.g., antibody molecules, disclosed herein can be conjugated with a detectable moiety, e.g., a label or imaging agent. Such moieties can include enzymes (e.g., horseradish peroxidase, beta-galactosidase, luciferase, alkaline phosphatase, acetylcholinesterase, glucose oxidase and the like), radiolabels (e.g., 3H, 14C, 15N, 35S, 90Y, 99Tc, 111In, 125I, 131I and the like), haptens, fluorescent labels (e.g., FITC, rhodamine, lanthanide phosphors, fluorescein, fluorescein isothiocyanate, rhodamine, 5-dimethylamine-1-napthalenesulfonyl chloride, phycoerythrin and the like), phosphorescent molecules, chemiluminescent molecules, chromophores, luminescent molecules, photoaffinity molecules, colored particles or affinity ligands, such as biotin, predetermined polypeptide epitopes recognized by a secondary reporter (e.g., leucine zipper pair sequences, or binding sites for secondary antibodies, metal binding domains, epitope tags). In some embodiments, a moiety, e.g., a detectable moiety, e.g., a label, is attached by spacer arms of various lengths to reduce potential steric hindrance.


In some embodiments, a binding agent, e.g., antibody molecule, disclosed herein, is derivatized with a detectable enzyme and is detected by adding additional reagents that the enzyme uses to produce a detectable reaction product. For example, when the detectable agent horseradish peroxidase is present, the addition of hydrogen peroxide and diaminobenzidine leads to a colored reaction product, which is detectable. A binding agent, e.g., antibody molecule, disclosed herein, ay also be derivatized with a prosthetic group (e.g., streptavidin/biotin and avidin/biotin). For example, an antibody may be derivatized with biotin, and detected through indirect measurement of avidin or streptavidin binding.


In some embodiments, the moiety comprises paramagnetic ions and NMR-detectable substances, among others. For example, in some embodiments, a paramagnetic ion is one or more of chromium (III), manganese (II), iron (III), iron (II), cobalt (II), nickel (II), copper (II), neodymium (III), samarium (III), ytterbium (III), gadolinium (III), vanadium (II), terbium (III), dysprosium (III), holmium (III), erbium (III), lanthanum (III), gold (III), lead (II), and/or bismuth (III). Binding agents, e.g., antibody molecules, as disclosed herein, can be modified to be associated with, e.g., conjugated to, a therapeutic agent, e.g., an agent comprising anti-viral activity, anti-inflammatory activity, or cytotoxic activity, etc. In some embodiments, therapeutic agents can treat symptoms or causes of influenza infection (e.g., for example, anti-viral, pain-relief, antiinflammatory, immunomodulatory, sleep-inducing activities, etc).


Treatment Methods and Administration

The binding agents, e.g., antibody molecules, featured in the disclosure, can be used to treat a subject, e.g., a subject, e.g., a human subject, infected with, or at risk for becoming infected with, an influenza virus.


Any human is candidate to receive an antibody molecule featured in the disclosure for treatment or prevention of an infection by an influenza virus. Humans at high risk of infection, such as immunocompromised individuals, and humans who are at high risk of exposure to influenza virus are particularly suited to receive treatment with the antibody molecule. Immunocompromised individuals include the elderly (65 years and older) and children (e.g., 6 months to 18 years old), and people with chronic medical conditions. People at high risk of exposure include heath care workers, teachers and emergency responders (e.g., firefighters, policemen).


The antibody molecules described herein can also be used to prevent or reduce (e.g., minimize) secondary infection (e.g., secondary bacterial infection) or a risk of comprising secondary infection associated with influenza, or any effects (e.g., symptoms or complications) thereof on a subject. Opportunistic secondary bacterial infections (e.g., secondary bacterial pneumonia, e.g., primarily with Streptococcus pneumonia) contribute significantly to the overall morbidity and mortality associated with seasonal and pandemic influenza infections. The antibody molecules described herein can be used to prevent or reduce (e.g., minimize) the complications from secondary, opportunistic infections (e.g., bacterial infections) in a subject.


An antibody molecule can be administered to a subject, e.g., a human subject, by a variety of methods. For many applications, the route of administration is one of: intravenous injection or infusion, subcutaneous injection, or intramuscular injection. An antibody molecule can be administered as a fixed dose, or in a mg/kg dose. The antibody molecule can be administered intravenously (IV) or subcutaneously (SC). For example, the antibody molecule can be administered at a fixed unit dose of between about 50-600 mg IV, e.g., every 4 weeks, or between about 50-100 mg SC (e.g., 75 mg), e.g., at least once a week (e.g., twice a week). In one embodiment, the antibody molecule is administered IV at a fixed unit dose of 50 mg, 60 mg, 80 mg, 100 mg, 120 mg, 130 mg, 140 mg, 150 mg, 160 mg, 180 mg, 200 mg, 300 mg, 400 mg, 500 mg, or 600 mg or more. Administration of the IV dose can be once or twice or three times or more per week, or once every two, three, four, or five weeks, or less frequently.


An anti-HA antibody molecule featured in the disclosure can also be administered intravenously, such as a fixed unit dose between 500 mg and 3000 mg, e.g., between 1000 mg and 3000 mg, between 1500 mg and 3000 mg, between 2000 mg and 3000 mg, between 1800 mg and 2500 mg, between 2500 mg and 3000 mg, between 500 mg and 2500 mg, between 500 mg and 2000 mg, between 500 mg and 1500 mg, between 500 mg and 1000 mg, between 1000 mg and 2500 mg, between 1500 mg and 2000 mg, or between 2000 mg and 2500 mg, e.g., 1500 mg, 1600 mg, 1700 mg, 1800 mg, 1900 mg, 2000 mg, 2100 mg, 2200 mg, 2300 mg, 2400 mg, or 2500 mg. In an embodiment, the antibody molecule is administered intravenously over a period of 1-3 hours, e.g., 1-2 hours or 2 to 3 hours, e.g., 2 hours. In an embodiment, the antibody molecule is administered as a single dose. In one embodiment, the antibody molecule is administered SC at a fixed unit dose of 50 mg, 60 mg, 70 mg, 75 mg, 80 mg, 100 mg, or 120 mg or more. Administration of the SC dose can be once or twice or three times or more per week, or once every two, three, four, or five weeks, or less frequently. An anti-HA antibody molecule featured in the disclosure can also be administered by inhalation, such as by intranasal or by oral inhalation, such as at a fixed unit dose of 50 mg, 60 mg, 80 mg, 100 mg, 120 mg, 130 mg, 140 mg, 150 mg, 160 mg, 180 mg, 200 mg, 300 mg, 400 mg, 500 mg, 600 mg, 700 mg, 800 mg, 900 mg, 1000 mg, 1100 mg, 1200 mg, 1300 mg, 1400 mg, 1500 mg, 1600 mg, 1700 mg, 1800 mg, 1900 mg, 2000 mg, 2100 mg, 2200 mg, 2300 mg, 2400 mg, 2500 mg, or more.


In an embodiment, the antibody molecule is administered in an amount that does not cause an ADE in the subject, e.g., as determined by a method described herein. In an embodiment, the antibody molecule is administered in an amount that does not cause viral resistance, e.g., as determined by a method described herein. In one embodiment, an anti-HA antibody is administered to a subject via vector-mediated gene transfer, such as through the delivery of a vector encoding the heavy chain and the light chain of an anti-HA antibody, and the antibody is expressed from the heavy chain and light chain genes in the body. For example, nucleic acids encoding a heavy chain and a light chain can be cloned in a AAV vector, such as a self-complementary AAV vector, the scAAV vector administered to a human by injection, such as by IM injection, and the antibody is expressed and secreted into the circulation of the human.


An antibody molecule can also be administered in a bolus at a dose of between about 1 and 50 mg/kg, e.g., between about 1 and 10 mg/kg, between about 1 and 25 mg/kg or about 25 and 50 mg/kg, e.g., about 50 mg/kg, 25 mg/kg, 10 mg/kg, 6.0 mg/kg, 5.0 mg/kg, 4.0 mg/kg, 3.0 mg/kg, 2.0 mg/kg, 1.0 mg/kg, or less. Modified dose ranges include a dose that is less than about 3000 mg/subject, about 1500 mg/subject, about 1000 mg/subject, about 600 mg/subject, about 500 mg/subject, about 400 mg/subject, about 300 mg/subject, about 250 mg/subject, about 200 mg/subject, or about 150 mg/subject, typically for administration every fourth week or once a month. The antibody molecule can be administered, for example, every three to five weeks, e.g., every fourth week, or monthly.


Dosing can be adjusted according to a patient's rate of clearance of a prior administration of the antibody. For example, a patient may not be administered a second or follow-on dose before the level of antibodies in the patient's system has dropped below a pre-determined level. In one embodiment, a sample from a patient (e.g., plasma, serum, blood, urine, or cerebrospinal fluid (CSF)) is assayed for the presence of antibodies, and if the level of antibodies is above a pre-determined level, the patient will not be administered a second or follow-on dose. If the level of antibodies in the patient's system is below a pre-determined level, then the patient is administered a second or follow-on dose. A patient whose antibody levels are determined to be too high (above the pre-determined level) can be tested again after one or two or three days, or a week, and if the level of antibody in the patient samples has dropped below the pre-determined level, the patient may be administered a second or follow-on dose of antibody.


In certain embodiments, the antibody may be prepared with a carrier that will protect the drug against rapid release, such as a controlled release formulation, including implants, and microencapsulated delivery systems. Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid. Many methods for the preparation of such formulations are patented or generally known. See, e.g., Controlled Drug Delivery (Drugs and the Pharmaceutical Sciences), Second Edition, J. Robinson and V. H. L. Lee, eds., Marcel Dekker, Inc., New York, 1987.


Pharmaceutical compositions can be administered with a medical device. For example, pharmaceutical compositions can be administered with a needleless hypodermic injection device, such as the devices disclosed in U.S. Pat. Nos. 5,399,163; 5,383,851; 5,312,335; 5,064,413; 4,941,880; 4,790,824; or 4,596,556. Examples of well-known implants and modules are discussed in, e.g., U.S. Pat. No. 4,487,603, which discloses an implantable micro-infusion pump for dispensing medication at a controlled rate; U.S. Pat. No. 4,486,194, which discloses a therapeutic device for administering medicaments through the skin; U.S. Pat. No. 4,447,233, which discloses a medication infusion pump for delivering medication at a precise infusion rate; U.S. Pat. No. 4,447,224, which discloses a variable flow implantable infusion apparatus for continuous drug delivery; U.S. Pat. No. 4,439,196, which discloses an osmotic drug delivery system comprising multi-chamber compartments; and U.S. Pat. No. 4,475,196, which discloses an osmotic drug delivery system. Of course, many other such implants, delivery systems, and modules are also known. In some embodiments, the binding agent, e.g., an antibody molecule, is administered buccally, orally, or by nasal delivery, e.g., as a liquid, spray, or aerosol, e.g., by topical application, e.g., by a liquid or drops, or by inhalation.


An antibody molecule described herein can be administered with one or more additional therapeutic agents, e.g., a second drug, for treatment of a viral infection, or a symptom of the infection. The antibody molecule and the one or more second or additional agents can be formulated together, in the same formulation, or they can be in separate formulations, and administered to a patient simultaneously or sequentially, in either order.


Dosage regimens are adjusted to provide the desired response, such as a therapeutic response or a combinatorial therapeutic effect. Generally, any combination of doses (either separate or co-formulated) of an antibody molecule and a second or additional agent can be used in order to provide a subject with both agents in bioavailable quantities. Dosage unit form or “fixed dose” as used herein refers to physically discrete units suited as unitary dosages for the subjects to be treated; each unit contains a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier and optionally in association with another agent.


A pharmaceutical composition may include a “therapeutically effective amount” of an agent described herein. In some embodiments, where the antibody molecule is administered in combination with a second or additional agent, such effective amounts can be determined based on the combinatorial effect of the administered first and second or additional agent. A therapeutically effective amount of an agent may also vary according to factors such as the disease state, age, sex, and weight of the individual, and the ability of the compound to elicit a desired response in the individual, such as amelioration of at least one infection parameter, or amelioration of at least one symptom of the infection, such as chills, fever, sore throat, muscle pain, headache, coughing, weakness, fatigue and general discomfort. A therapeutically effective amount is also one in which any toxic or detrimental effects of the composition are outweighed by the therapeutically beneficial effects.


In an embodiment, administration of a binding agent, e.g., antibody molecule, provided, e.g., as a pharmaceutical preparation, is by one of the following routes: oral, intravenous, intramuscular, intra-arterial, subcutaneous, intraventricular, transdermal, interdermal, rectal, intravaginal, intraperitoneal, topical (as by liquids, powders, ointments, creams, sprays, or drops), mucosal, nasal, buccal, enteral, sublingual; intratracheal instillation, bronchial instillation, and/or inhalation; and/or as an oral spray, nasal spray, and/or aerosol. In an embodiment, the method described herein further comprises determining the presence or absence of an anti-drug antibody (ADA) in the subject. In an embodiment, the subject is selected for administration of an antibody molecule described herein on the basis of the absence of an ADA in the subject. ADA can be detected, e.g., by ELISA, in a sample from the subject.


Combination Treatments and Exemplary Second or Additional Agents

Binding agents, e.g., antibody molecules, provided e.g., as pharmaceutical compositions, can be administered either alone or in combination with one or more other therapy, e.g., the administration of a second or additional therapeutic agent.


In some embodiments, the combination can result in a lower dose of the antibody molecule or of the other therapy being needed, which, in some embodiments, can reduce side effects. In some embodiments, the combination can result in enhanced delivery or efficacy of one or both agents. The agents or therapies can be administered at the same time (e.g., as a single formulation that is administered to a patient or as two separate formulations administered concurrently) or sequentially in any order. Such second or additional agents include vaccines, anti-viral agents, and/or additional antibodies. In typical embodiments the second or additional agent is not co-formulated with the binding agent, e.g., antibody molecule, though in others it is. In some embodiments, the binding agent, e.g., antibody molecule, and the second or additional agent are administered such that one or more of the following is achieved: therapeutic levels, or therapeutic effects, of one overlap the other; detectable levels of both are present at the same time; or the therapeutic effect is greater than what would be seen in the absence of either the binding agent, e.g., antibody molecule, or the second or additional agent. In some embodiments, each agent will be administered at a dose and on a time schedule determined for that agent.


The second or additional agent can be, for example, for treatment or prevention of influenza. For example, the binding agents, e.g., antibody molecules, e.g., therapeutic antibodies, provided herein can be administered in combination with a vaccine, e.g., a vaccine described herein or a mixture (a.k.a. a cocktail) of influenza peptides to stimulate the patient's immune system to prevent infection with particular strains of influenza A. In other examples, the second or additional agent is an anti-viral agent (e.g., an anti-NA or anti-M2 agent), a pain reliever, an anti-inflammatory, an antibiotic, a steroidal agent, a second therapeutic antibody molecule (e.g., an anti-HA antibody), an adjuvant, a protease or glycosidase (e.g., sialidase), etc.


Exemplary anti-viral agents include, e.g., vaccines, neuraminidase inhibitors or nucleoside analogs. Exemplary anti-viral agents can include, e.g., zidovudine, gangcyclovir, vidarabine, idoxuridine, trifluridine, foscarnet, acyclovir, ribavirin, amantadine, remantidine, saquinavir, indinavir, ritonavir, alpha-interferons and other interferons, a neuraminidase inhibitor (e.g., zanamivir (Relenza®), oseltamivir (Tamiflu®), laninamivir, peramivir), rimantadine. Exemplary second antibody molecules include, for exampleAb 67-11 (U.S. Provisional application No. 61/645,453, FI6 (U.S. Application Publication No. 2010/0080813), FI28 (U.S. Application Publication No. 2010/0080813), C179 (Okuno et al., J. Virol. 67:2552-8, 1993), F10 (Sui et al., Nat. Struct. Mol. Biol. 16:265, 2009), CR9114 (Dreyfus et al., Science 337:1343, 2012), or CR6261 (Ekiert et al., Science 324:246, 2009). Thus, Ab 044 can be used in combination of any of those antibodies. In other embodiments, two or more binding agents, e.g., antibody molecules disclosed herein, can be administered in combination, e.g., Ab 044 can be administered in combination with Ab 032. In the case of combinations, two agents can be administered as part of the same dosage unit or administered separately. Other exemplary agents useful for treating the symptoms associated with influenza infection are acetaminophen, ibuprofen, aspirin, and naproxen.


In one embodiment, the antibody molecule and the second or additional agent are provided as a co-formulation, and the co-formulation is administered to the subject. It is further possible, e.g., at least 24 hours before or after administering the co-formulation, to administer separately one dose of the antibody formulation and then one dose of a formulation containing a second or additional agent. In another implementation, the antibody molecule and the second or additional agent are provided as separate formulations, and the step of administering includes sequentially administering the antibody molecule and the second or additional agent. The sequential administrations can be provided on the same day (e.g., within one hour of one another or at least 3, 6, or 12 hours apart) or on different days.


In some embodiments, the antibody molecule and the second or additional agent are each administered as a plurality of doses separated in time. The antibody molecule and the second or additional agent are generally each administered according to a regimen. The regimen for one or both may have a regular periodicity. The regimen for the antibody molecule can have a different periodicity from the regimen for the second or additional agent, e.g., one can be administered more frequently than the other. In one implementation, one of the antibody molecule and the second or additional agent is administered once weekly and the other once monthly. In another implementation, one of the antibody molecule and the second or additional agent is administered continuously, e.g., over a period of more than 30 minutes but less than 1, 2, 4, or 12 hours, and the other is administered as a bolus. In some embodiments, sequential administrations are administered. The time between administration of the one agent and another agent can be minutes, hours, days, or weeks. The use of an antibody molecule described herein can also be used to reduce the dosage of another therapy, e.g., to reduce the side-effects associated with another agent that is being administered. Accordingly, a combination can include administering a second or additional agent at a dosage at least 10, 20, 30, or 50% lower than would be used in the absence of the antibody molecule. The antibody molecule and the second or additional agent can be administered by any appropriate method, e.g., subcutaneously, intramuscularly, or intravenously.


In some embodiments, each of the antibody molecule and the second or additional agent is administered at the same dose as each is prescribed for monotherapy. In other embodiments, the antibody molecule is administered at a dosage that is equal to or less than an amount required for efficacy if administered alone. Likewise, the second or additional agent can be administered at a dosage that is equal to or less than an amount required for efficacy if administered alone. In some cases, the formulations described herein, e.g., formulations containing an antibody molecule featured in the disclosure, include one or more second or additional agents, or are administered in combination with a formulation containing one or more second or additional agents. In an embodiment a binding agent, e.g., antibody molecule, provided, e.g., as a pharmaceutical preparation, is administered by inhalation or aerosol delivery of a plurality of particles, e.g., particles comprising a mean particle size of 4, 5, 6, 7, 8, 9, 10, 11, 12, or 13 microns.


Pharmaceutical Compositions

The binding agents, e.g., antibody molecules, featured in the disclosure can be formulated as pharmaceutical compositions, such as for the treatment or prevention of influenza.


Typically, a pharmaceutical composition includes a pharmaceutically acceptable carrier. As used herein, “pharmaceutically acceptable carrier” includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like that are physiologically compatible.


A “pharmaceutically acceptable salt” refers to a salt that retains the desired biological activity of the parent compound and does not impart any undesired toxicological effects (see e.g., Berge, S. M., et al. (1977) J. Pharm. Sci. 66:1-19). Examples of such salts include acid addition salts and base addition salts. Acid addition salts include those derived from nontoxic inorganic acids, such as hydrochloric, nitric, phosphoric, sulfuric, hydrobromic, hydroiodic, and the like, as well as from nontoxic organic acids such as aliphatic mono- and dicarboxylic acids, phenyl-substituted alkanoic acids, hydroxy alkanoic acids, aromatic acids, aliphatic and aromatic sulfonic acids and the like. Base addition salts include those derived from alkaline earth metals, such as sodium, potassium, magnesium, calcium and the like, as well as from nontoxic organic amines, such as N,N′-dibenzylethylenediamine, N-methylglucamine, chloroprocaine, choline, diethanolamine, ethylenediamine, procaine and the like.


The compositions comprising antibody molecules can be formulated according to methods known in the art. Pharmaceutical formulation is a well-established art, and is further described in Gennaro (ed.), Remington: The Science and Practice of Pharmacy, 20th ed., Lippincott, Williams & Wilkins (2000) (ISBN: 0683306472); Ansel et al., Pharmaceutical Dosage Forms and Drug Delivery Systems, 7th Ed., Lippincott Williams & Wilkins Publishers (1999) (ISBN: 0683305727); and Kibbe (ed.), Handbook of Pharmaceutical Excipients American Pharmaceutical Association, 3rd ed. (2000) (ISBN: 091733096X).


Pharmaceutical compositions may be in a variety of forms. These include, for example, liquid, semi-solid and solid dosage forms, such as liquid solutions (e.g., injectable and infusible solutions), dispersions or suspensions, tablets, pills, powders, liposomes and suppositories. The form can depend on the intended mode of administration and therapeutic application. Typically, compositions for the agents described herein are in the form of injectable or infusible solutions. Such compositions can be administered by a parenteral mode (e.g., intravenous, subcutaneous, intraperitoneal, or intramuscular injection). The phrases “parenteral administration” and “administered parenterally” as used herein mean modes of administration other than enteral and topical administration, usually by injection, and include, without limitation, intravenous, intramuscular (IM), intraarterial, intrathecal, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intraarticular, subcapsular, subarachnoid, intraspinal, epidural and by intrasternal injection or by infusion.


Pharmaceutical compositions may be provided in a sterile injectable form (e.g., a form that is suitable for subcutaneous injection or intravenous infusion). In some embodiments, pharmaceutical compositions are provided in a liquid dosage form that is suitable for injection or topical application. In some embodiments, pharmaceutical compositions are provided as in dry form, e.g., as powders (e.g. lyophilized and/or sterilized preparations). The Pharmaceutical composition can be provided under conditions that enhance stability, e.g., under nitrogen or under vacuum. Dry material can be reconstituted with an aqueous diluent (e.g., water, buffer, salt solution, etc.) prior to injection.


In one embodiment, the pharmaceutical composition containing an anti-HA antibody is administered intranasally. In another embodiment, the pharmaceutical composition containing an anti-HA antibody is administered by inhalation, such as by oral or by nasal inhalation. In some embodiments, the pharmaceutical composition is suitable for buccal, oral or nasal delivery, e.g., as a liquid, spray, or aerosol, e.g., by topical application, e.g., by a liquid or drops, or by inhalation). In some embodiments, a pharmaceutical preparation comprises a plurality of particles, suitable, e.g., for inhaled or aerosol delivery. In some embodiments, the mean particle size of 4, 5, 6, 7, 8, 9, 10, 11, 12, or 13 microns. In some embodiments, a pharmaceutical preparation is formulated as a dry powder, suitable, e.g., for inhaled or aerosol delivery. In some embodiments, a pharmaceutical preparation is formulated as a wet powder, through inclusion of a wetting agent, e.g., water, saline, or other liquid of physiological pH. In some embodiments, a pharmaceutical preparation is provided as drops, suitable, e.g., for delivery to the nasal or buccal cavity. In some embodiments, the pharmaceutical composition is disposed in a delivery device, e.g., a syringe, a dropper or dropper bottle, an inhaler, or a metered dose device, e.g., an inhaler.


In one embodiment, a pharmaceutical composition contains a vector, such as an adenovirus-associated virus (AAV)-based vector, that encodes a heavy chain of an anti-HA antibody molecule, and a light chain of an anti-HA antibody molecule featured in the disclosure. The composition containing the vector can be administered to a subject, such as a patient, such as by injection, e.g., IM injection. Genes encoding the anti-HA antibody under control of, for example, cytomegalovirus (CMV) promoters, are expressed in the body, and the recombinant anti-HA antibody molecule is introduced into the circulation. See, e.g., Balazs et al., Nature 30:481:81-84, 2011.


Pharmaceutical compositions typically should be sterile and stable under the conditions of manufacture and storage. A pharmaceutical composition can also be tested to insure it meets regulatory and industry standards for administration. The composition can be formulated as a solution, microemulsion, dispersion, liposome, or other ordered structure suitable to high drug concentration. Sterile injectable solutions can be prepared by incorporating an agent described herein in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, followed by filtered sterilization. Generally, dispersions are prepared by incorporating an agent described herein into a sterile vehicle that contains a basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile powders for the preparation of sterile injectable solutions, typical methods of preparation are vacuum drying and freeze-drying that yields a powder of an agent described herein plus any additional desired ingredient from a previously sterile-filtered solution thereof. The proper fluidity of a solution can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants. Prolonged absorption of injectable compositions can be brought about by including in the composition an agent that delays absorption, for example, monostearate salts and gelatin.


A pharmaceutical composition may be provided, prepared, packaged, and/or sold in bulk, as a single unit dose, and/or as a plurality of single unit doses. Typically a bulk preparation will contain at least 2, 5, 10, 20, 50, or 100 unit doses. A unit dose is typically the amount introduced into the patient in a single administration. In some embodiments, only a portion of a unit dose is introduced. In some embodiments, a small multiple, e.g., as much as 1.5, 2, 3, 5, or 10 times a unit dose is administered. The amount of the active ingredient is generally equal to a dose which would be administered to a subject and/or a convenient fraction of such a dose such as, for example, one-half or one-third of such a dose.


Immunogens and Vaccines

Antibodies of the invention have elucidated epitopes that are useful for inducing immunity to, and in some embodiments, provide protection from, one or more, e.g., at least two, influenza strains. These epitopes are referred to herein as “broad range immunogens.” As used herein, the term “broad range vaccine” refers to a preparation comprising a broad range immunogen, or a nucleic acid encoding a broad range immunogen, that can induce formation of antibodies or immunity against the broad range immunogen or an organism, e.g., an influenza virus. Additional immunogens and vaccines, and uses thereof, are described in International Publication No. WO2013/170139 or U.S. Application Publication No. 2013/0302349, the contents of which are hereby incorporated by reference in their entirety.


EPitope

HAs exist in nature as homotrimers of proteolytically processed mature subunits. Each subunit of the trimer is synthesized as a precursor. A precursor molecule is proteolytically processed into two disulfide bonded polypeptide chains to form a mature HA polypeptide. The mature HA polypeptide includes two domains: (1) a core HA-1 domain that extends from the base of the molecule through the fibrous stem to the membrane distal head region that contains the glycan receptor binding domain, returning to fibrous region ending in the cleavage site, and (2) HA-2 domain that includes the stem region and the transmembrane domain of HA. HA-1 includes a glycan binding site. The glycan binding site may be responsible for mediating binding of HA to the HA-receptor. The HA-2 domain acts to present the HA-1 domain. The HA trimer can be stabilized by polar and non-polar interactions between the three long HA alpha-helices of the stem of HA monomers.


HA sequences from all influenza subtypes share a set of amino acids in the interface of the HA-1 and HA-2 domains that are well conserved. The HA-1/HA-2 interface membrane proximal epitope region (MPER) that includes the canonical a-helix and residues in its vicinity are also conserved across a broad spectrum of subtypes. (Ekiert et al., Science., 324(5924):246, 2009; Sui et al., Nat Struct Mol Biol. 16(3):265, 2009).


Ab 044 has high affinity for HA's from Group 1 and Group 2. It binds a conformational epitope that is broadly conserved across a plurality of influenza strains. Numerous amino acid residues distributed along the linear sequences of HA from different strains/subtypes contribute the Ab 044 conformational epitope. The interaction of Ab 044 with H3 was analyzed by docking studies and residues bound by (or not bound by) Ab 044 were identified. The Fv of Ab 044 was docked against HA of group I and II strains using ZDOCK. The structure of the HA antigen was modeled using the SWISS MODEL homology modeling server keeping the solved crystal structure of H1N1 as the template. ZDOCK uses shape complementarity along with desolvation and electrostatic energy terms (‘ZRANK’) to rank docked poses. To ensure the docked poses do not deviate significantly from the native complex, mapped epitope and paratope residues by alanine scanning are forced to be included in the binding interface.


For comparison studies, amino acids that bind (or do not bind) F16 were taken from published US patent application US 2011/0274702 A1, Neutralizing Anti-Influenza A Virus Antibodies and Uses Thereof, filed Jul. 18, 2011.


ZDOCK is a Fast Fourier Transform based protein docking program. It was developed by Zhiping Weng at the University of Massachusetts Medical School. In ZDOCK, two PDB files are input and the output is the predicted structure of their complex. The program searches all possible binding modes in the translational and rotational space between the two proteins and evaluates each by an energy scoring function. The protein's structure is converted to a digital signal and a Fast Fourier Transform technique used to reduce computational time. ZDOCK is discussed in Pierce B G, Hourai Y, Weng Z. (2011) Accelerating Protein Docking in ZDOCK Using an Advanced 3D Convolution Library. PLoS One 6(9): e24657, Pierce B, Tong W, Weng Z. (2005) M-ZDOCK: A Grid-based Approach for Cn Symmetric Multimer Docking. Bioinformatics 21(8): 1472-1476; Mintseris J, Pierce B, Wiehe K, Anderson R, Chen R, Weng Z. (2007) Integrating Statistical Pair Potentials into Protein Complex Prediction. Proteins 69(3): 511-520; and Chen R, Li L, Weng Z. (2003) ZDOCK: An Initial-stage Protein Docking Algorithm. Proteins 52(1): 80-7.


SWISS-MODEL is a fully automated protein structure homology-modeling server. It is accessible via the ExPASy web server, or from the program DeepView (Swiss Pdb-Viewer). Swiss-Model is discussed in Arnold K., Bordoli L., Kopp J., and Schwede T. (2006). The SWISS-MODEL Workspace: A web-based environment for protein structure homology modelling. Bioinformatics, 22, 195-201; Kiefer F, Arnold K, Kiinzli M, Bordoli L, Schwede T (2009). The SWISS-MODEL Repository and associated resources. Nucleic Acids Research. 37, D387-D392; and Peitsch, M. C. (1995) Protein modeling by E-mail Bio/Technology 13: 658-660.


H3 residues that bind Ab 044 and H3 residues that bind F16 are discussed below.


H3 HA1


The amino acid sequence of H3 HA1 is provided below, as SEQ ID NO: 173. Residues N38, 1278, and D291 shown in dashed boxes, are bound by Ab 044 but not by FI6; Residues Q327, T328, and R329 shown in dotted boxes, are bound by FI6 but not by Ab 044; residues T318, R321, and V323 shown in solid boxes, are bound by both Ab 044 and FI6.










(SEQ ID NO: 173)





embedded image





GIDCTLIDAL LGDPHCDVFQ NETWDLFVER SKAFSNCYPY DVPDYASLRS LVASSGTLEF





ITEGFTWTGV TQNGGSNACK RGPGSGFFSR LNWLTKSGST YPVLNVTMPN NDNFDKLYIW





GIHHPSTNQE QTSLYVQASG RVTVSTRRSQ QTIIPNIGSR PWVRGLSSRI SIYWTIVKPG







embedded image






embedded image








H3 HA2


The amino acid sequence of H3 HA21 is provided below, as SEQ ID NO: 174 Residue N12 shown in a dash box, is bound by Ab 044 but not by FI6; Residues G1, L2, F3, G4, and D46 shown in dotted boxes, are bound by FI6 but not by Ab 044; residues A7, E11, I18, D19, G20, W21, L38, K39, T41, Q42, A43, I45, I48, N49, L52, N53, 156, and E57, shown in solid boxes, are bound by both Ab 044 and FI6.










(SEQ ID NO: 174)





embedded image





EKFHQIEKEF SEVEGRIQDL EKYVEDTKID LWSYNAELLV ALENQHTIDL TDSEMNKLFE





KTRRQLRENA EEMGNGCFKI YHKCDNACIE SIRNGTYDHD VYRDEALNNR FQIKG






H1 residues that bind Ab 044 and H1 residues that bind FI6 are discussed below.


H1 HA1 The amino acid sequence of H1 HA1 is provided below, as SEQ ID NO: 181. Residues H31, N279, and S292 shown in dashed boxes, are bound by Ab 044 but not by FI6. Residues Q328 and S329 shown in dotted boxes, are bound by FI6 but not by Ab 044. Residues T319, R322, and 1324 shown in solid boxes, are bound by both Ab 044 and FI6.











(SEQ ID NO: 181)





embedded image





EDSHNGKLCK LKGIAPLQLG KCNIAGWLLG NPECDLLLTA






SSWSYIVETS NSENGTCYPG DFIDYEELRE QLSSVSSFEK






FEIFPKTSSW PNHETTKGVT AACSYAGASS FYRNLLWLTK






KGSSYPKLSK SYVNNKGKEV LVLWGVHHPP TGTDQQSLYQ






NADAYVSVGS SKYNRRFTPE IAARPKVRDQ AGRMNYYWTL






LEPGDTITFE ATGNLIAPWY AFALNRGSGS GIITSDAPVH








embedded image







embedded image








H1 HA2


The amino acid sequence of H1 HA2 is provided below, as SEQ ID NO: 182. Residues G12 shown in a dashed box, is bound by Ab 044 but not by FI6. Residues G1, L2, F3, G4, and D46 shown in dotted boxes, are bound by F16 but not by Ab 044. Residues A7, E11, I18, D19, G20, W21, Q38, K39, T41, Q42, N43, I45, I48, T49, V52, N53, 156, and E57 shown in solid boxes, are bound by both Ab 044 and F16.











(SEQ ID NO: 182)





embedded image







embedded image





LNKKVDDGFL DIWTYNAELL VLLENERTLD FHDSNVRNLY 






EKVKSQLKNN AKEIGNGCFE FYHKCDDACM ESVRNGTYDY






PKYSEESKLN REEIDGVKLE SMGVYQILAI YSTVASSLVL






LVSLGAISFW MCSNGSLQCR ICI






A three dimensional representation of H3 HA with the amino acids residues that are predicted to be part of Ab 044 epitope but not part of F16's epitope highlighted (i.e., the highlighted amino acids are unique to Ab 044's epitope) is depicted in FIG. 26 of International Publication No. WO2013/170139 or U.S. Application Publication No. 2013/0302349. A three dimensional representation of H3 HA with the amino acid residues that are part of F16's epitope but not predicted to be part of Ab 044's epitope highlighted is depicted in FIG. 27 of International Publication No. WO2013/170139 or U.S. Application Publication No. 2013/0302349.


Diagnostic Methods

The methods described herein can further include a diagnostic step as described herein. The binding agents, e.g., antibody molecules, provided herein are useful for identifying the presence of influenza in a biological sample, e.g., a patient sample, such as a fluid sample, e.g., a blood, serum, saliva, mucous, or urine sample, or a tissue sample, such as a biopsy. In one embodiment, a patient sample is contacted with a binding agent, e.g., an antibody molecule, featured in the disclosure, and binding is detected. Binding can be detected with a number of formats and means of detection, e.g., with an antigen capture assay, such as an ELISA assay or Western blot, or an immunohistochemistry assay. In some embodiments, the binding agent, e.g., an antibody molecule, is provided, e.g., coupled to an insoluble matrix, e.g., a bead or other substrate, and a detection molecule used to detect binding of HA.


Binding of binding agent, e.g., antibody molecule, to HA, can be detected with a reagent comprising a detectable moiety, e.g., a reagent, e.g., an antibody, which binds the binding agent, e.g., antibody molecule. In some embodiments, the binding agent, e.g., antibody molecule, has a detectable moiety. Suitable detectable moieties include enzymes (e.g., horseradish peroxidase, beta-galactosidase, luciferase, alkaline phosphatase, acetylcholinesterase, glucose oxidase and the like), radiolabels (e.g., 3H, 14C, 15N, 35S, 90Y, 99Tc, 111In, 125I, 131I), haptens, fluorescent labels (e.g., FITC, rhodamine, lanthanide phosphors, fluorescein, fluorescein isothiocyanate, rhodamine, 5-dimethylamine-1-napthalenesulfonyl chloride, phycoerythrin and the like), phosphorescent molecules, chemiluminescent molecules, chromophores, luminescent molecules, photoaffinity molecules, colored particles or affinity ligands, such as biotin, predetermined polypeptide epitopes recognized by a secondary reporter (e.g., leucine zipper pair sequences, or binding sites for secondary antibodies, metal binding domains, epitope tags). In some embodiments, labels are attached by spacer arms of various lengths to reduce potential steric hindrance.


In some embodiments, a human is tested for presence of influenza virus be a method described herein, and if the test is positive, a binding agents, e.g., antibody molecules, e.g., an antibody, provided herein, is administered. The binding agents, e.g., antibody molecules, e.g., an antibody, provided herein can be used for cytology assays, such as to identify an HA in a cell. The assay can be a colorimetric assay. A biological sample from a normal (non-infected) individual is used as a control. The diagnostic assay can be performed in vitro. The diagnostic assay can also be performed to determine infection of cells in culture, e.g., of mammalian cells in culture. The antibody molecules can be used in in vitro assays.


Because the antibody molecules featured herein bind a broad spectrum of HA subtypes, the diagnostic assays featured in the disclosure can detect the presence of influenza virus in patients infected with a variety of distinct strains of influenza. A patient sample can be further tested with subtype specific antibodies, or other assays (e.g., RFLP (Restriction Fragment Length Polymorphism), PCR (Polymerase Chain Reaction), RT-PCR (Reverse Transcription coupled to Polymerase Chain Reaction), Northern blot, Southern blot or DNA sequencing) to further determine the particular strain of virus. In one embodiment, a patient determined to be infected with influenza A can be further administered an antibody molecule featured in the disclosure, to treat the infection. Also provided are solid substrates, e.g., beads, dipsticks, arrays, and the like, on which is disposed a binding agent, e.g., antibody molecule.


Kits

A binding agent, e.g., an antibody molecule, disclosed herein, e.g., generated by the methods described herein, can be provided in a kit, e.g., for use in a method described herein. The kit can include one or more other components, e.g., containers, buffers or other diluents, delivery devices, and the like.


In one embodiment, the kit includes materials for administering an antibody molecule to a subject, such as for treatment or prevention of infection by influenza viruses. For example, the kit can include one or more or all of: (a) a container that contains a composition that includes an antibody molecule, optionally (b) a container that contains a composition that includes a second therapeutic agent, and optionally (c) informational material. In another embodiment, the kit includes materials for using an antibody molecule in a diagnostic assay, such as for detection of HA in a biological sample. For example, the kit can include one or more or all of: (a) a container that contains a composition that includes an antibody molecule, optionally (b) a container that contains a reagents, e.g., labeled with a detectable moiety, to detect the antibody, e.g., for use in an ELISA or immunohistochemistry assay, and optionally (c) informational material. In other embodiments, the kit comprises a binding agent, e.g., antibody molecule, comprising a detectable moiety.


In an embodiment, the kit comprises a solid substrate, e.g., bead, dipstick, array, and the like, on which is disposed a binding agent, e.g., antibody molecule. The informational material can be descriptive, instructional, marketing or other material that relates to the methods described herein and/or the use of the agents for therapeutic benefit, or for a diagnostic assay. The informational material of the kits is not limited in its form. In one embodiment, the informational material can include information about production of the antibody, concentration, date of expiration, batch or production site information, and so forth. In one embodiment, the informational material relates to methods of administering the antibody, e.g., in a suitable dose, dosage form, or mode of administration (e.g., a dose, dosage form, or mode of administration described herein), to treat a subject who has an infection, e.g., viral infection or secondary infection (e.g., secondary bacterial infection). In another embodiment, the informational material relates to methods for using the antibody molecule for a diagnostic assay, e.g., to detect the presence of influenza viruses in a biological sample. The information can be provided in a variety of formats, including printed text, computer readable material, video recording, or audio recording, or information that provides a link or address to substantive material. In addition to the agent, the composition in the kit can include other ingredients, such as a solvent or buffer, a stabilizer, or a preservative. The agent can be provided in any form, e.g., a liquid, dried or lyophilized form, and substantially pure and/or sterile. When the agents are provided in a liquid solution, the liquid solution typically is an aqueous solution. When the agents are provided as a dried form, reconstitution generally is by the addition of a suitable solvent. The solvent, e.g., sterile water or buffer, can optionally be provided in the kit.


The kit can include one or more containers for the composition or compositions containing the agents. In some embodiments, the kit contains separate containers, dividers or compartments for the composition and informational material. For example, the composition can be contained in a bottle, vial, or syringe, and the informational material can be contained in a plastic sleeve or packet. In other embodiments, the separate elements of the kit are contained within a single, undivided container. For example, the composition is contained in a bottle, vial or syringe that has attached thereto the informational material in the form of a label. In some embodiments, the kit includes a plurality (e.g., a pack) of individual containers, each containing one or more unit dosage forms (e.g., a dosage form described herein) of the agents. The containers can include a combination unit dosage, e.g., a unit that includes both the antibody molecule and the second or additional agent, such as in a desired ratio. For example, the kit can include a plurality of syringes, ampoules, foil packets, blister packs, or medical devices each containing, for example, a single combination unit dose. The containers of the kits can be air tight, waterproof (e.g., impermeable to changes in moisture or evaporation), and/or light-tight.


The kit optionally includes a device suitable for administering the composition, e.g., a syringe or device for delivering particles or aerosols, e.g., an inhaler, a spray device, or a dropper or other suitable delivery device. The device can be provided pre-loaded with one or both of the agents or can be empty but suitable for loading. The invention is further illustrated by the following examples, which should not be construed as further limiting.


Other Embodiments

The antibody molecule described herein can be encoded by a nucleic acid molecule, e.g., an isolated nucleic acid molecule. In an embodiment, the nucleic acid molecule comprises a nucleotide sequence that encodes a heavy chain immunoglobulin variable region segment featured in the disclosure. In another embodiment, the nucleic acid molecule comprises a nucleotide sequence encoding a light chain immunoglobulin variable region segment featured in the disclosure. In yet another aspect, the nucleic acid molecule comprises a nucleotide sequence that encodes a heavy chain immunoglobulin variable region segment featured in the disclosure and a light chain immunoglobulin variable region segment featured in the disclosure. In an embodiment, the nucleic acid molecule is present in a vector, e.g., a recombinant vector (e.g., an expression vector). In an embodiment, the vector comprises a nucleic acid molecule that comprises a nucleotide sequence that encodes a heavy chain immunoglobulin variable region segment featured in the disclosure, a nucleotide sequence that encodes a light chain immunoglobulin variable region segment featured in the disclosure, or both. In one embodiment, the nucleic acid molecule in the recombinant vector includes a nucleotide sequence encoding (a) a heavy chain immunoglobulin variable region segment comprising the amino acid sequence of: S-Y-A-M-H (SEQ ID NO:68) in CDR1; V-V-S-Y-D-G-N-Y-K-Y-Y-A-D-S-V-Q-G (SEQ ID NO:69) in CDR2; and D-S-R-L-R-S-L-L-Y-F-E-W-L-S-Q-G-Y-F-N-P (SEQ ID NO:70) in CDR3; and (b) a light chain immunoglobulin variable region segment comprising the amino acid sequence of: Q-S-I-T-F-D-Y-K-N-Y-L-A (SEQ ID NO:145) in CDR1; W-G-S-Y-L-E-S (SEQ ID NO:72) in CDR2; and Q-Q-H-Y-R-T-P-P-S (SEQ ID NO:73) in CDR3.


In an embodiment, the antibody molecule described herein is produced from a cell containing a recombinant vector featured in the disclosure, such as a recombinant vector comprising a nucleic acid sequence that encodes a heavy chain immunoglobulin variable region, or a recombinant vector comprising a nucleic acid sequence that encodes a light chain immunoglobulin variable region. In one embodiment, the cell contains a recombinant vector comprising a nucleic acid sequence that encodes a heavy chain immunoglobulin variable region, and a recombinant vector comprising a nucleic acid sequence that encodes a light chain immunoglobulin variable region. In yet another embodiment, the cell contains a recombinant vector comprising a nucleic acid sequence that encodes a heavy chain immunoglobulin variable region, and a nucleic acid sequence that encodes a light chain immunoglobulin variable region. In an embodiment, the antibody molecule is produced, e.g., by providing a host cell comprising a nucleic acid sequence expressing a heavy chain segment and a nucleic acid sequence expressing a light chain segment, and expressing the nucleic acids in the host cell. In one embodiment, the nucleic acid sequence expressing the heavy chain segment and the nucleic acid sequence expressing the light chain segment are on the same recombinant expression vector. In another embodiment, the nucleic acid sequence expressing the heavy chain segment and the nucleic acid sequence expressing the light chain segment are on separate recombinant expression vectors.


In an embodiment, a pharmaceutical composition containing an antibody molecule featured in the disclosure, and a pharmaceutically acceptable carrier, is used in a method described herein.


In an embodiment, the method described herein treats or prevents an infection with an influenza virus (e.g., an influenza A virus, e.g., a Group 1 strain, e.g., an H1N1 strain, e.g., A/South Carolina/1/1918, A/Puerto Rico/08/1934, or A/California/04/2009, or an H5N1 strain, e.g., A/Indonesia/5/2005 or A/Vietnam/1203/2004, or an influenza B virus, e.g., B/Wisconsin/1/2010), in a subject, e.g., a human subject, that comprises: administering a binding agent, e.g., an antibody molecule, featured in the disclosure to a subject, e.g., human subject, in need thereof. In one embodiment, the influenza A virus is an H1, H5, H9, H3 or H7 strain, such as an H1N1 strain, an H3N2 strain, or an H5N1 strain of influenza A virus. In an embodiment, the administration results in, or correlates with, one or more of a reduction in the incidence or severity of a symptom or manifestation of an influenza infection, or the delay or onset of a symptom or manifestation of an influenza infection. In an embodiment, the administration results in, or correlates with, one or more of a reduction in the incidence or severity of a symptom or manifestation of a secondary infection, or the delay or onset of a symptom or manifestation of a secondary infection. In some embodiments, the subject, e.g., a human subject, has been administered, or the method comprises, administering, or recommending the administration of, a second or additional therapy.


In some embodiments, the antibody molecule is administered in combination with a second or additional agent or therapy. In some embodiments, the second or additional therapy comprises administration of a vaccine or an anti-viral therapy, e.g., an anti-NA or an anti-M2 therapy. In an embodiment, the second or additional therapy comprises an administration of a vaccine, e.g., a vaccine described herein or a mixture (a.k.a. a cocktail) of influenza peptides to stimulate the patient's immune system to prevent infection with particular strains of influenza A. In an embodiment, the second or additional agent comprises administering an anti-viral agent, a pain reliever, an anti-inflammatory, an antibiotic, a steroidal agent, a second therapeutic antibody molecule (e.g., an anti-HA antibody), an adjuvant, a protease or glycosidase (e.g., sialidase). In an embodiment, the second or additional agent comprises, acyclovir, ribavirin, amantadine, remantidine, a neuraminidase inhibitor (e.g., zanamivir (Relenza®), oseltamivir (Tamiflu®), laninamivir, peramivir), or rimantadine.


In an embodiment, the second or additional agent comprises a second antibody molecule, e.g., Ab 67-11 (U.S. Provisional application No. 61/645,453, FI6 (U.S. Application Publication No. 2010/0080813), FI28 (U.S. Application Publication No. 2010/0080813), C179 (Okuno et al., J. Virol. 67:2552-8, 1993), F10 (Sui et al., Nat. Struct. Mol. Biol. 16:265, 2009), CR9114 (Dreyfus et al., Science 337:1343, 2012), or CR6261 (see, e.g., Ekiert et al., Science 324:246, 2009). Thus, Ab 044 can be used in combination of any of those antibodies. In an embodiment, the second or additional agent comprises a second or additional binding agent, e.g., antibody molecule, e.g., an anti-HA antibody, e.g., an anti-HA antibody disclosed herein. E.g., two or more of Ab 044, Ab 069, Ab 032, and Ab 031 can be administered. E.g., Ab 044 can be administered in combination with Ab 069 or Ab 032. In the case of combinations, two agents can be administered as part of the same dosage unit or administered separately. Other exemplary agents useful for treating the symptoms associated with influenza infection are acetaminophen, ibuprofen, aspirin, and naproxen.


In an embodiment, the binding agent, e.g., an antibody molecule, is administered to a human subject suffering from or susceptible to an influenza infection. In an embodiment, the binding agent, e.g., an antibody molecule, is administered prior to known exposure to influenza, or to particular influenza subtypes or strains. In an embodiment, the binding agent, e.g., an antibody molecule, is administered prior to manifestation of effects or symptoms of influenza infection, or to one or more particular effects manifestation of effects or symptoms of influenza infection. In an embodiment, the binding agent, e.g., an antibody molecule, is administered after known exposure to influenza, or to particular influenza subtypes or strains. In an embodiment, the binding agent, e.g., an antibody molecule, is administered after manifestation of effects or symptoms of influenza infection, or after observation of one or more particular effects manifestation of effects or symptoms of influenza infection. In an embodiment, the binding agent, e.g., an antibody molecule, is administered in response to, or to treat or prevent, a manifestation of an effect or a symptom of influenza infection, e.g., inflammation, fever, nausea, weight loss, loss of appetite, rapid breathing, increase heart rate, high blood pressure, body aches, muscle pain, eye pain, fatigue, malaise, dry cough, runny nose, and/or sore throat.


In an embodiment, the method further comprises, testing the human subject for the influenza virus, e.g., with a method disclosed herein. In some embodiments, the administration is responsive to a positive test for influenza.


In an embodiment, the method described herein treats a subject, e.g., a human subject, an infected with an influenza virus (e.g., an influenza A virus, e.g., a Group 1 strain, e.g., an H1N1 strain, e.g., A/South Carolina/1/1918, A/Puerto Rico/08/1934, or A/California/04/2009, or an H5N1 strain, e.g., A/Indonesia/5/2005 or A/Vietnam/1203/2004, or an influenza B virus, e.g., B/Wisconsin/1/2010) by administering a binding agent, e.g., an antibody molecule, featured in the disclosure. For example, the influenza A virus is an H1, H5, H9, H3 or H7 strain, such as an H1N1 strain, an H3N2 strain, or an H5N1 strain of influenza A virus. In one embodiment, a binding agent, e.g., an anti-HA antibody, described herein is administered instead of a vaccine for prevention of influenza. In another embodiment, the binding agent, e.g., anti-HA antibody molecule, is administered in combination with (simultaneously or sequentially with) a vaccine for prevention of the flu.


In an embodiment, the method further comprises detecting influenza (e.g., influenza A or influenza B) virions in a biological sample, such as by contacting the sample with a binding agent, e.g., an antibody molecule, featured in the disclosure, and then detecting the binding of the antibody molecule to the sample. In one embodiment, the method of detecting the influenza virus (e.g., influenza A or influenza B virus) is performed in vitro.


In an embodiment, the method further includes: (a) providing a sample from a patient; (b) contacting the sample with a binding agent, e.g., an antibody molecule, featured in the disclosure, and (c) determining whether the binding agent, e.g., an antibody molecule, featured in the disclosure binds a polypeptide in the sample, where if the binding agent, e.g., an antibody molecule, binds a polypeptide in the sample, then the patient is determined to be infected with an influenza virus (e.g., an influenza A virus, e.g., a Group 1 strain, e.g., an H1N1 strain, e.g., A/South Carolina/1/1918, A/Puerto Rico/08/1934, or A/California/04/2009, or an H5N1 strain, e.g., A/Indonesia/5/2005 or A/Vietnam/1203/2004, or an influenza B virus, e.g., e.g., B/Wisconsin/1/2010). In one embodiment, the patient is determined to be infected with an influenza virus (e.g., an influenza A virus, e.g., a Group 1 strain, e.g., an H1N1 strain, e.g., A/South Carolina/1/1918, A/Puerto Rico/08/1934, or A/California/04/2009, or an H5N1 strain, e.g., A/Indonesia/5/2005 or A/Vietnam/1203/2004, or an influenza B virus, e.g., B/Wisconsin/1/2010), and the patient is further administered a binding agent, e.g., an antibody molecule, disclosed herein, e.g., the binding agent, e.g., an antibody molecule, with which the test was performed.


In an embodiment, the method further includes inducing immunity to one or more influenza strains, or preventing, delaying or reducing infection with an influenza strain, or symptom thereof, in a vertebrate, e.g., a human. The method comprises administering to the vertebrate, e.g., a human, a broad range vaccine, or broad range immunogen, described herein.


In an embodiment, the broad range vaccine, or broad range immunogen, induces an immune response against, or confers protection against, one or more influenza strains. In an embodiment, the broad range vaccine, or broad range immunogen, induces an immune response against, or confers protection against, two influenza strains. In an embodiment, the broad range vaccine, or broad range immunogen, induces an immune response against, or confers protection against, two Group 1 influenza strains. In an embodiment, the broad range vaccine induces, or broad range immunogen, an immune response against, or confers protection against, at least one Group 1 strain, and a second strain from Group 1, Group 2 or an influenza B strain. In one embodiment, the influenza A virus is an H1, H5, H9, H3 or H7 strain, such as an H1N1 strain, an H3N2 strain, or an H5N1 strain of influenza A virus.


In an embodiment, the administration results in, or correlates with, one or more of: a reduction in the chance of an infection, a reduction in the incidence or severity of a symptom or manifestation of an influenza infection, or the delay or onset of a symptom or manifestation of an influenza infection. In an embodiment, the administration results in, or correlates with, one or more of: a reduction in the incidence or severity of a symptom or manifestation of a secondary infection, or the delay or onset of a symptom or manifestation of a secondary infection.


In some embodiments, the subject, e.g., a human subject, has been administered, or the method comprises, administering, or recommending the administration of, a second or additional therapy. In some embodiments, the broad range vaccine is administered in combination with a second or additional agent or therapy. In some embodiments, the second or additional agent comprises administration of another vaccine or another anti-viral therapy, e.g., an anti-NA or an anti-M2 therapy. In an embodiment, the second or additional agent comprises administration of a vaccine comprising a mixture (a.k.a. a cocktail) of influenza peptides to stimulate the patient's immune system to prevent infection with particular strains of influenza A. In an embodiment, the second or additional agent comprises administering an anti-viral agent, a pain reliever, an anti-inflammatory, an antibiotic, a steroidal agent, a second therapeutic antibody molecule (e.g., an anti-HA antibody), an adjuvant, a protease or glycosidase (e.g., sialidase). In an embodiment, the second or additional agent comprises, acyclovir, ribavirin, amantadine, remantidine, a neuraminidase inhibitor (e.g., zanamivir (Relenza®), oseltamivir (Tamiflu®), laninamivir, peramivir), or rimantadine. In an embodiment, the second or additional agent comprises an antibody molecule, e.g., Ab 67-11 (U.S. Provisional application No. 61/645,453, FI6 (U.S. Application Publication No. 2010/0080813), FI28 (U.S. Application Publication No. 2010/0080813), C179 (Okuno et al., J. Virol. 67:2552-8, 1993), F10 (Sui et al., Nat. Struct. Mol. Biol. 16:265, 2009), CR9114 (Dreyfus et al., Science 337:1343, 2012), or CR6261 (Ekiert et al., Science 324:246, 2009). In an embodiment, the second or additional agent comprises an antibody molecule disclosed herein, e.g., an antibody molecule selected from Ab-044, Ab 069, Ab 032, and Ab 031 antibody molecules. In the case of combinations, two agents can be administered as part of the same dosage unit or administered separately. Other exemplary second or additional agents useful for treating the symptoms associated with influenza infection are acetaminophen, ibuprofen, aspirin, and naproxen.


In an embodiment, the method further comprises, testing the human subject for the influenza virus, e.g., with a method disclosed herein. In some embodiments, the administration is responsive to a positive test for influenza. In an embodiment, the method further comprises reducing the severity of influenza in a population. The method includes administering a broad range vaccine, or broad range immunogen, to sufficient individuals in the population to prevent or decrease the chance of influenza virus transmission to another individual in the population.


Anti-HA antibody molecules described herein are also disclosed in International Publication No. WO2013/170139, U.S. Pat. Nos. 8,877,200, 9,096,657, and U.S. Patent Application Publication No. US 2013/0302349. The contents of the aforesaid publications are incorporated by reference in their entirety.









TABLE 4C







Nucleic acid and amino acid sequences











SEQ






ID NO.
Lab no.
Source
Comment
Sequence














1
n.a.
Table 2
Consensus AA sequence of HC 
[S/T]Y[A/G]MH





CDR1






2
n.a.
Table 2
Consensus AA sequence of HC 
V[I/V/L]S[Y/F]DG[S/N][Y/N][K/R]YYADSVQG





CDR2






3
n.a.
Table 2
Consensus AA sequence of HC 
D[S/T][R/K/Q]LR[S/T]LLYFEWLS[Q/S]G[Y/L/V][F/L][N/D][P/Y]





CDR3






4
n.a.
Table 2
Consensus AA sequence of LC 
Q[S/T][V/L/I][T/S][Y/F/W][N/S/D]YKNYLA





CDR1






170
n.a.
Table 2
Consensus AA sequence of LC 
Q[S/T][V/L/I][T/S][Y/F/W][N/S/D/Q/R/E]YKNYLA





CDR1






5
n.a.
Table 2
Consensus AA sequence of LC 
W[A/G]S[T/A/Y/H/K/D][R/L]E[S/T]





CDR2






6
n.a.
Table 2
Consensus AA sequence of LC 
QQ[Y/H]YRTPP[T/S]





CDR3






7
n.a.
Table 2
Consensus AA sequence of HC FR1
[E/Q]VQLLE[S/T]GGGLVKPGQSLKLSCAASGFTF[S/T]





8
n.a.
Table 2
Consensus AA sequence of HC FR2
WVRQPPGKGLEWVA





9
n.a.
Table 2
Consensus AA sequence of HC FR3
RFTISRDNSKNTLYLQMNSLRAEDTAVYYCAK





10
n.a.
Table 2
Consensus AA sequence of HC FR4
WG[A/Q]G[T/A][T/M][L/V]TVSS





11
n.a.
Table 2
Consensus AA sequence of LC FR1
[E/D]I[V/Q]MTQSP[D/S][S/T][L/V][A/S][V/A][S/T][L/V/R]G[E/D]R[A/V]





12
n.a.
Table 2
Consensus AA sequence of LC FR2
WYQQKPG[Q/K][P/A]PKLLIY





13
n.a.
Table 2
Consensus AA sequence of LC FR3
GVP[D/E/S]RFSGSGSGTDFTLTISSLQ[A/P]ED[V/F/K/D]A[V/T]YYC





14
n.a.
Table 2
Consensus AA sequence of LC FR4
FG[G/Q/T/S/N]GTK[L/V][D/E]IK





15
15
Table 3,
AA sequence of HC VR  of Ab 
EVQLLESGGGLVKPGQSLKLSCAASGFTFTSYGMHWVRQPPGKGLEWVAVISYDGSYKYYADSVQGRFTISRDNSKNTLYLQMNSL



VH15
Table 4A,
A18; entire HC domain is in 
RAEDTAVYYCAKDSRLRSLLYFEWLSQGYFNPWGAGTTLTVSS




FIG. 2
FIG. 1; ID version is in FIG. 






13; NT sequence is in Example 1






28
28
Table 3,
AA sequence of LC VR of Ab 
EIVMTQSPDSLAVSLGERATINCKSSQSVTYNYKNYLAWYQQKPGQPPKLLIYWASTRESGVPDRFSGSGSGTDFTLTISSLQAED



VL28
Table 4A
A18; entire LC domain is in 
VAVYYCQQYYRTPPTFGGGTKLDIK




FIG. 3
FIG. 1; ID version is in FIG. 






14; NT sequence is in Example 1






16
16
Table 3
AA sequence of HC VR of Abs 
EVQLLESGGGLVKPGQSLKLSCAASGFTFSSYGMHWVRQPPGKGLEWVAVVSYDGSNKYYADSVQGRFTISRDNSKNTLYLQMNSL



VH16
Table 4A
014, 028; ID version is in 
RAEDTAVYYCAKDTKLRSLLYFEWLSSGLLDYWGQGAMVTVSS




FIG. 2
FIG. 13; NT sequence is in 






Example 1






29
29
Table 3
AA sequence of LC VR of Abs 014,
EIVMTQSPDSLAVSLGERATINCKSSQSVTFSYKNYLAWYQQKPGQPPKLLIYWASTRESGVPDRFSGSGSGTDFTLTISSLQAED



VL29
Table 4A
154, 157; ID version is in FIG.
VAVYYCQQYYRTPPTFGGGTKLDIK




FIG. 3
14; NT sequence is in Example 1






30
30
Table 3
AA sequence of LC VR of Abs 028,
EIVMTQSPDSLAVSLGERATINCKSSQSVTFDYKNYLAWYQQKPGQPPKLLIYWASTRESGVPDRFSGSGSGTDFTLTISSLQAED



VL30
Table 4A
155; ID version is in FIG. 14; 
VAVYYCQQYYRTPPTFGGGTKLDIK




FIG. 3
NT sequence is in Example 1






17
17
Table 3
AA sequence of HC VR of Abs 001,
EVQLLESGGGLVKPGQSLKLSCAASGFTFTSYGMHWVRQPPGKGLEWVAVVSYDGNYKYYADSVQGRFTISRDNSKNTLYLQMNSL



VH17
Table 4A
009, 017, 025, 160, 186, 187, 
RAEDTAVYYCAKDSRLRSLLYFEWLSQGYFNPWGAGTTLTVSS




FIG. 2
188, 189, 190, 191, 192, 193, 






202, 211; ID version is in FIG.






13;






31
31
Table 3
AA sequence of LC VR of Abs 001,
EIVMTQSPDSLAVSLGERATINCKSSQTVTFNYKNYLAWYQQKPGQPPKLLIYWASTRESGVPDRFSGSGSGTDFTLTISSLQAED



VL31
Table 4A
002, 003; ID version is in FIG.
VAVYYCQQHYRTPPSFGGGTKLDIK




FIG. 3
14;






18
18
Table 3
AA sequence of HC VR of Abs 002,
EVQLLESGGGLVKPGQSLKLSCAASGFTFTSYGMHWVRQPPGKGLEWVAVLSYDGNYKYYADSVQGRFTISRDNSKNTLYLQMNSL



VH18
Table 4A
010, B18, 026, 203, 212; ID 
RAEDTAVYYCAKDSRLRSLLYFEWLSQGYFNPWGAGTTLTVSS




FIG. 2
version is in FIG. 13;






19
19
Table 3
AA sequence of HC VR of Abs 003,
EVQLLESGGGLVKPGQSLKLSCAASGFTFTTYAMHWVRQPPGKGLEWVAVLSYDGNYKYYADSVQGRFTISRDNSKNTLYLQMNSL



VH19
Table 4A
011, 019, 027, 194, 195, 196, 
RAEDTAVYYCAKDSRLRSLLYFEWLSQGYFNPWGAGTTLTVSS




FIG. 2
197, 198, 199, 200, 204, 213; 






ID version is in FIG. 13;






32
32
Table 3
AA sequence of LC VR of Abs 009,
EIVMTQSPDSLAVSLGERATINCKSSQTLSFNYKNYLAWYQQKPGQPPKLLIYWASTRESGVPDRFSGSGSGTDFTLTISSLQAED



VL32
Table 4A
010, 011; ID version is in FIG.
VAVYYCQQHYRTPPSFGGGTKLDIK




FIG. 3
14;






33
33
Table 3
AA sequence of LC VR of Abs 017,
EIVMTQSPDSLAVSLGERATINCKSSQTVTFNYKNYLAWYQQKPGQPPKLLIYFASTRESGVPDRFSGSGSGTDFTLTISSLQAED



VL33
Table 4A
B18, 019; ID version is in FIG.
VAVYYCQQHYRTPPSFGGGTKLDIK




FIG. 3
14;






34
34
Table 3
AA sequence of LC VR of Abs 025,
EIVMTQSPDSLAVSLGERATINCKSSQTLSFNYKNYLAWYQQKPGQPPKLLIYFASTRESGVPDRFSGSGSGTDFTLTISSLQAED



VL34
Table 4A
026, 027, 086; ID version is in
VAVYYCQQHYRTPPSFGGGTKLDIK




FIG. 3
FIG. 14;






20
20
Table 3
AA sequence of HC VR of Ab 086;
EVQLLESGGGLVKPGQSLKLSCAASGFTFTTYAMHWVRQPPGKGLEWVAVVSFDGNNRYYADSVQGRFTISRDNSKNTLYLQMNSL



VH20
Table 4A
ID version is in FIG. 13;
RAEDTAVYYCAKDSQLRSLLYFEWLSSGVLDYWGQGAMVTVSS




FIG. 2







21
21
Table 3
AA sequence of HC VR of Abs 154,
EVQLLESGGGLVKPGQSLKLSCAASGFTFSSYGMHWVRQPPGKGLEWVAVVSYDGNNKYYADSVQGRFTISRDNSKNTLYLQMNSL



VH21
Table 4A
155; ID version is in FIG. 13;
RAEDTAVYYCAKDSKLRSLLYFEWLSSGLLDYWGQGAMVTVSS




FIG. 2







22
22
Table 3
AA sequence of HC VR of Abs 157,
EVQLLESGGGLVKPGQSLKLSCAASGFTFTTYAMHWVRQPPGKGLEWVAVVSYDGNNKYYADSVQGRFTISRDNSKNTLYLQMNSL



VH22
Table 4A
159; ID version is in FIG. 13;
RAEDTAVYYCAKDSKLRSLLYFEWLSSGLLDYWGQGAMVTVSS




FIG. 2







35
35
Table 3
AA sequence of LC VR of Ab 159;
EIVMTQSPDSLAVSLGERATINCKSSQSVTWSYKNYLAWYQQKPGQPPKLLIYWASTRESGVPDRFSGSGSGTDFTLTISSLQAED



VL35
Table 4A
ID version is in FIG. 14;
VAVYYCQQYYRTPPTFGGGTKLDIK




FIG. 3







36
36
Table 3
AA sequence of LC VR of Ab 160;
EIVMSQSPDTLAVTLGERASINCKSSQTVTFNYKNYLAWYQQKPGQPPKVLIYWASARETGVPERFSGSGSGTDFTLTISSLQAED



VL36
Table 4A
ID version is in FIG. 14;
VAVYYCQQHYRTPPSFGQGTKLEIK




FIG. 3







37
37
Table 3
AA sequence of LC VR of Abs 186,
EIVMTQSPDSLAVSLGERATINCKSSQTVTFNYKNYLAWYQQKPGQPPKLLIYWASTRESGVPDRFSGSGSGTDFTLTISSLQAED



VL37
Table 4A
194; ID version is in FIG. 14;
VAVYYCQQHYRTPPSFGTGTKLDIK





38
38
Table 3
AA sequence of LC VR of Abs 187,
EIVMTQSPDSLAVSLGERATINCKSSQTVTFNYKNYLAWYQQKPGQPPKLLIYWASTRESGVPDRFSGSGSGTDFTLTISSLQAED



VL38
Table 4A
195; ID version is in FIG. 14;
VAVYYCQQHYRTPPSFGSGTKLDIK




FIG. 3







39
39
Table 3
AA sequence of LC VR of Abs 188,
EIVMTQSPDSLAVSLGERATINCKSSQTVTFNYKNYLAWYQQKPGQPPKLLIYWASTRESGVPDRFSGSGSGTDFTLTISSLQAED



VL39
Table 4A
196; ID version is in FIG. 14;
VAVYYCQQHYRTPPSFGQGTKLDIK




FIG. 3







40
40
Table 3
AA sequence of LC VR of Abs 189,
EIVMTQSPDSLAVSLGERATINCKSSQTVTFNYKNYLAWYQQKPGQPPKLLIYWASTRESGVPDRFSGSGSGTDFTLTISSLQAED



VL40
Table 4A
197; ID version is in FIG. 14;
VAVYYCQQHYRTPPSFGNGTKLDIK




FIG. 3







41
41
Table 3
AA sequence of LC VR of Abs 190,
EIVMTQSPDSLAVSLGERATINCKSSQTLSFNYKNYLAWYQQKPGQPPKLLIYWASTRESGVPDRFSGSGSGTDFTLTISSLQAED



VL41
Table 4A
198; ID version is in FIG. 14;
VAVYYCQQHYRTPPSFGTGTKLDIK




FIG. 3







42
42
Table 3
AA sequence of LC VR of Abs 191,
EIVMTQSPDSLAVSLGERATINCKSSQTLSFNYKNYLAWYQQKPGQPPKLLIYWASTRESGVPDRFSGSGSGTDFTLTISSLQAED



VL42
Table 4A
199; ID version is in FIG. 14;
VAVYYCQQHYRTPPSFGSGTKLDIK





43
43
Table 3
AA sequence of LC VR of Abs 192,
EIVMTQSPDSLAVSLGERATINCKSSQTLSFNYKNYLAWYQQKPGQPPKLLIYWASTRESGVPDRFSGSGSGTDFTLTISSLQAED



VL43
Table 4A
200; ID version is in FIG. 14;
VAVYYCQQHYRTPPSFGQGTKLDIK




FIG. 3







44
44
Table 3
AA sequence of LC VR of Abs 193;
EIVMTQSPDSLAVSLGERATINCKSSQTLSFNYKNYLAWYQQKPGQPPKLLIYWASTRESGVPDRFSGSGSGTDFTLTISSLQAED



VL44
Table 4A
ID version is in FIG. 14;
VAVYYCQQHYRTPPSFGNGTKLDIK




FIG. 3







45
45
Table 3
AA sequence of LC VR of Abs 202,
DIQMTQSPSSLSASVGDRVTITCRSSQSITFNYKNYLAWYQQKPGKAPKLLIYWGSYLESGVPSRFSGSGSGTDFTLTISSLQPED



VL45
Table 4A
203, 204, 210, 031, 032, 033, 
FATYYCQQHYRTPPSFGQGTKVEIK




FIG. 3
034; ID version is in FIG. 14;






NT sequence is in Example 1






46
46
Table 3
AA sequence of LC VR of Abs 211,
DIQMTQSPSSLSASVGDRVTITCRSSQSITFNYKNYLGWYQQKPGKAPKLLIYWGSYLESGVPSRFSGSGSGTDFTLTISSLQPED



VL46
Table 4A
212, 213, 219, 037, 038, 039, 
FATYYCQQHYRTPPSFGQGTKVEIK




FIG. 3
040; ID version is in FIG. 14;






23
23
Table 3
AA sequence of HC VR of Abs 210,
EVQLLESGGGLVKPGQSLKLSCAASGFTFTSYGMHWVRQPPGKGLEWVAVVSYDGNYKYYADSVQGRFTISRDNSKNTLYLQMNSL



VH23
Table 4A
219; ID version is in FIG. 13;
RAEDTAVYYCAKDSKLRSLLYFEWLSQGYFNPWGAGTTLTVSS




FIG. 2







24
24
Table 3
AA sequence of HC VR of Abs 
EVQLLESGGGLVKPGQSLKLSCAASGFTFTSYAMHWVRQPPGKGLEWVAVVSYDGNYKYYADSVQGRFTISRDNSKNTLYLQMNSL



VH24
Table 4A
A001, A002, A003, A010, A011, 
RAEDTAVYYCAKDSRLRSLLYFEWLSQGYFNPWGQGTTLTVSS




FIG. 2
031, 037; ID version is in FIG.






13; NT sequence is in Example 1






47
47
Table 3
AA sequence of LC VR of Abs 
DIVMTQSPDTLAVTLGERATIQCKSSQTVTFNYKNYLAWYQQKPGQPPKLLIYWASTRESGVPDRFSGSGSGTDFTLTITSLQAED



VL47
Table 4A
A001, 004, 007, 016; ID version
VAVYYCQQHYRTPPSFGQGTKLDIK




FIG. 3
is in FIG. 14;






48
48
Table 3
AA sequence of LC VR of Abs 
DIVMTQSPDTVAVTVGERATINCKSSQTVTFNYKNYLAWYQQKPGQPPKLLIYWASTRESGVPDRFSGSGSGTDFTLTISSLQAED



VL48
Table 4A
002, 005, 008, A017; ID version
VAVYYCQQHYRTPPSFGQGTKLDIK




FIG. 3
is in FIG. 14;






25
25
Table 3
AA sequence of HC VR of Abs 004,
QVQLLETGGGLVKPGQSLKLSCAASGFTFTSYAMHWVRQPPGKGLEWVAVVSYDGNYKYYADSVQGRFTISRDNSKNTLYLQMNSL



VH25
Table 4A
005, 006, 012, 013, 032, 038, 
RAEDTAVYYCAKDSRLRSLLYFEWLSQGYFNPWGQGTTLTVSS




FIG. 2
043, 044, 045, 046, 047, 048, 






049, 050, 051, 052, 067, 068, 






069, 070, 073, 074, 075, 076, 






077; ID version is in FIG. 13;






NT sequence is in Example 1






49
49
Table 3
AA sequence of LC VR of Abs 
DIVMTQSPDTVAVTLGERATIDCKSSQTVTFNYKNYLAWYQQKPGQPPKLLIYWASTRESGVPDRFSGSGSGTDFTLTISSLQAED



VL49
Table 4A
A003, 006, A009, C18; ID 
VAVYYCQQHYRTPPSFGQGTKLDIK




FIG. 3
version is in FIG. 14;






26
26
Table 3
AA sequence of HC VR of Abs 007,
EVQLLESGGGLVKPGQSLKLSCAASGFTFTSYAMHWVRQPPGKGLEWVAVVSYDGNYKYYADSVQGRFTISRDNSKNTLYLQMNSL



VH26
Table 4A
008, A009, A14, 015, 033, 039; 
RAEDTAVYYCAKDSQLRTLLYFEWLSQGYFNPWGQGTTLTVSS




FIG. 2
ID version is in FIG. 13;






50
50
Table 3
AA sequence of LC VR of Abs 
DIVMTQSPDTLAVTVGERATIRCKSSQTVTFNYKNYLAWYQQKPGQPPKLLIYWASTRESGVPDRFSGSGSGTDFTLTISSLQAED



VL50
Table 4A
A010 012, A14, A019; ID version
VAVYYCQQHYRTPPSFGQGTKLDIK




FIG. 3
is in FIG. 14;






51
51
Table 3
AA sequence of LC VR of Ab A011,
DIVMTQSPDTLAVSRGERATIDCKSSQTVTFNYKNYLAWYQQKPGQPPKLLIYWASTRESGVPDRFSGSGSGTDFTLTISSLQAED



VL51
Table 4A
013, 015; ID version is in FIG.
EAVYYCQQHYRTPPSFGQGTKLDIK




FIG. 3
14;






27
27
Table 3
AA sequence of HC VR of Abs 016,
EVQLLESGGGLVKPGQSLKLSCAASGFTFTSYAMHWVRQPPGKGLEWVAVVSYDGNYKYYADSVQGRFTISRDNSKNTLYLQMNSL



VH27
Table 4A
A017, C18, A019, 034,040; ID 
RAEDTAVYYCAKDSRLRTLLYFEWLSQGYFDPWGQGTTLTVSS




FIG. 2
version is in FIG. 13;






60
60
Table 3
AA sequence of LC VR of Ab 043;
DIQMTQSPSSLSASVGDRVTITCRSSQSITFNYKNYLAWYQQKPGKAPKLLIYWGSYLESGVPSRFSGSGSGTDFTLTISSLQPED



VL60
Table 4A
ID version is in FIG. 14;
FATYYCQQYYRTPPSFGQGTKVEIK




FIG. 3







52
52
Table 3
AA sequence of LC VR of Abs 044,
DIQMTQSPSSLSASVGDRVTITCRSSQSITFDYKNYLAWYQQKPGKAPKLLIYWGSYLESGVPSRFSGSGSGTDFTLTISSLQPED



VL52
Table 4A
071, 072, 078; ID version is in
FATYYCQQHYRTPPSFGQGTKVEIK




FIG. 3
FIG. 14; NT sequence is in 






Example 1






57
57
Table 3
AA sequence of LC VR of Ab 045;
DIQMTQSPSSLSASVGDRVTITCRSSQSITFNYKNYLAWYQQKPGKAPKLLIYWGSYLESGVPSRFSGSGSGTDFTLTISSLQPED



VL57
Table 4A
ID version is in FIG. 14;
VATYYCQQHYRTPPSFGQGTKVEIK




FIG. 3







59
59
Table 3
AA sequence of LC VR of Ab 046;
DIQMTQSPSSLSASVGDRVTITCRSSQSITFNYKNYLAWYQQKPGKAPKLLIYWGSYLESGVPSRFSGSGSGTDFTLTISSLQPED



VL59
Table 4A
ID version is in FIG. 14;
DATYYCQQHYRTPPSFGQGTKVEIK




FIG. 3







55
55
Table 3
AA sequence of LC VR of Ab 047;
DIQMTQSPSSLSASVGDRVTITCRSSQSITFNYKNYLAWYQQKPGKAPKLLIYWGSKLESGVPSRFSGSGSGTDFTLTISSLQPED



VL55
Table 4A
ID version is in FIG. 14;
FATYYCQQHYRTPPSFGQGTKVEIK




FIG. 3







58
58
Table 3
AA sequence of LC VR of Ab 048;
DIQMTQSPSSLSASVGDRVTITCRSSQSITFNYKNYLAWYQQKPGKAPKLLIYWGSYLESGVPSRFSGSGSGTDFTLTISSLQPED



VL58
Table 4A
ID version is in FIG. 14;
KATYYCQQHYRTPPSFGQGTKVEIK




FIG. 3







54
54
Table 3
AA sequence of LC VR of Ab 049;
DIQMTQSPSSLSASVGDRVTITCRSSQSITFNYKNYLAWYQQKPGKAPKLLIYWGSHLESGVPSRFSGSGSGTDFTLTISSLQPED



VL54
Table 4A
ID version is in FIG. 14;
FATYYCQQHYRTPPSFGQGTKVEIK




FIG. 3







56
56
Table 3
AA sequence of LC VR of Ab 050;
DIQMTQSPSSLSASVGDRVTITCRSSQSITFNYKNYLAWYQQKPGKAPKLLIYWGSDLESGVPSRFSGSGSGTDFTLTISSLQPED



VL56
Table 4A
ID version is in FIG. 14;
FATYYCQQHYRTPPSFGQGTKVEIK





53
53
Table 3
AA sequence of LC VR of Ab 051;
DIQMTQSPSSLSASVGDRVTITCRSSQSITFNYKNYLAWYQQKPGKAPKLLIYWGSTLESGVPSRFSGSGSGTDFTLTISSLQPED



VL53
Table 4A
ID version is in FIG. 14;
FATYYCQQHYRTPPSFGQGTKVEIK




FIG. 3







61
61
Table 3
AA sequence of LC VR of Ab 052;
DIQMTQSPSSLSASVGDRVTITCRSSQSITFNYKNYLAWYQQKPGKAPKLLIYWGSTRESGVPSRFSGSGSGTDFTLTISSLQPED



VL61
Table 4A
ID version is in FIG. 14;
FATYYCQQHYRTPPSFGQGTKVEIK




FIG. 3







153
153
Table 3
AA sequence of LC VR of Ab 067;
DIQMTQSPSSLSASVGDRVTITCRSSQSITFQYKNYLAWYQQKPGKAPKLLIYWGSYLESGVPSRFSGSGSGTDFTLTISSLQPED



VL153
Table 4A
ID version is in FIG. 14;
FATYYCQQHYRTPPSFGQGTKVEIK




FIG. 3







154
154
Table 3
AA sequence of LC VR of Ab 068;
DIQMTQSPSSLSASVGDRVTITCRSSQSITFRYKNYLAWYQQKPGKAPKLLIYWGSYLESGVPSRFSGSGSGTDFTLTISSLQPED



VL154
Table 4A
ID version is in FIG. 14;
FATYYCQQHYRTPPSFGQGTKVEIK




FIG. 3







155
155
Table 3
AA sequence of LC VR of Abs 
DIQMTQSPSSLSASVGDRVTITCRSSQSITFEYKNYLAWYQQKPGKAPKLLIYWGSYLESGVPSRFSGSGSGTDFTLTISSLQPED



VL155
Table 4A
069, 079; ID version is in 
FATYYCQQHYRTPPSFGQGTKVEIK





FIG. 14;






156
156
Table 3
AA sequence of LC VR of Ab 070;
DIQMTQSPSSLSASVGDRVTITCRSSQSITFDYKNYLAWYQQKPGKAPKLLIYWGSTRESGVPSRFSGSGSGTDFTLTISSLQPED



VL156
Table 4A
ID version is in FIG. 14;
FATYYCQQHYRTPPSFGQGTKVEIK




FIG. 3







162
162
Table 3
AA sequence of HC VR of Ab 071
EVQLLESGGGLVKPGQSLKLSCAASGFSFSTYAMHWVRQPPGKGLEWVAVVSYDGNYKYYADTVQGRFTISRDNSKNTLYLQMNSL



VL162
Table 4A

RAEDTAVYYCAKDSRLRSLLYFEWLSQGYFNPWGQGTTLTVSS




FIG. 17







163
163
Table 3
AA sequence of HC VR of Ab 072
EVQLLESGGGLRKPGQSLKLSCAASGFSFSTYAMHWVRQPPGKGLEWVAVVSYDGNYKYYADSVQGRFTISRDNSKNTLYLQMNSL



VL163
Table 4A

RAEDTAVYYCAKDSRLRSLLYFEWLSQGYFNPWGQGTTLTVSS




FIG. 17







165
165
Table 3
AA sequence of LC VR of Ab 073
DIQMTQSPSSLSASVGDRVTITCRSSQSITWNYKNYLAWYQQKPGKAPKLLIYWGSYLESGVPSRFSGSGSGTDFTLTISSLQPED



VL165
Table 4A

FATYYCQQHYRTPPSFGQGTKVEIK




FIG. 17







166
166
Table 3
AA sequence of LC VR of Abs 
DIQMTQSPSSLSASVGDRVTITCRSSQSITWDYKNYLAWYQQKPGKAPKLLIYWGSYLESGVPSRFSGSGSGTDFTLTISSLQPED



VL166
Table 4A
074, 080
FATYYCQQHYRTPPSFGQGTKVEIK




FIG. 17




167
167
Table 3
AA sequence of LC VR of Ab 075
DIQMTQSPSSLSASVGDRVTITCRSSQSITWQYKNYLAWYQQKPGKAPKLLIYWGSYLESGVPSRFSGSGSGTDFTLTISSLQPED



VL167
Table 4A

FATYYCQQHYRTPPSFGQGTKVEIK




FIG. 17







168
168
Table 3
AA sequence of LC VR of Ab 076
DIQMTQSPSSLSASVGDRVTITCRSSQSITWRYKNYLAWYQQKPGKAPKLLIYWGSYLESGVPSRFSGSGSGTDFTLTISSLQPED



VL168
Table 4A

FATYYCQQHYRTPPSFGQGTKVEIK




FIG. 17







169
169
Table 3
AA sequence of LC VR of Abs 
DIQMTQSPSSLSASVGDRVTITCRSSQSITWEYKNYLAWYQQKPGKAPKLLIYWGSYLESGVPSRFSGSGSGTDFTLTISSLQPED



VL169
Table 4A
077, 081
FATYYCQQHYRTPPSFGQGTKVEIK




FIG. 17







164
164
Table 3
AA sequence of HC VR of Abs 
QVQLLETGGGLVKPGQSLKLSCAASGFTFTSYAMHWVRQPPGKGLEWVAVVSYDGNYKYYADSVQGRFTISRDNSKNTLYLQMNSL



VL164
Table 4A
078, 079, 080, 081
RAEDTAVYYCAKDSRLRSLLYFEWLSQGYFNPWGQGTTVTVSS




FIG. 17







161
HC161
Table 4A
AA sequence of HC VR consensus;
EVQLLESGGGLVKPGQSLKLSCAASGFTFSSYGMHWVRQPPGKGLEWVAVVSYDGSNKYYADSVQGRFTISRDNSKNTLYLQMNSL




FIG. 2
ID version is in FIG. 13;
RAEDTAVYYCAKDSKLRSLLYFEWLSSGLLDYWGQGAMVTVSS





62
LC62
Table 4A
AA sequence of LC VR consensus;
DIQMTQSPSSLSASVGDRVTITCRSSQSITFNYKNYLAWYQQKPGKAPKLLIYWGSYLESGVPSRFSGSGSGTDFTLTISSLQPED




FIG. 3
ID version is in FIG. 14;
FATYYCQQHYRTPPSFGQGTKVEIK





96
 15-ID
Table 4B
AA sequence of HC VR of Ab A18;
IDEVQLLESGGGLVKPGQSLKLSCAASGFTFTSYGMHWVRQPPGKGLEWVAVISYDGSYKYYADSVQGRFTISRDNSKNTLYLQMN




FIG. 13
non-ID version is in FIG. 2;
SLRAEDTAVYYCAKDSRLRSLLYFEWLSQGYFNPWGAGTTLTVSS





110
 28-ID
Table 4B
AA sequence of LC VR of Ab A18;
IDEIVMTQSPDSLAVSLGERATINCKSSQSVTYNYKNYLAWYQQKPGQPPKLLIYWASTRESGVPDRFSGSGSGTDFTLTISSLQA




FIG. 14
non-ID version is in FIG. 3
EDVAVYYCQQYYRTPPTFGGGTKLDIK





97
 16-ID
Table 4B
AA sequence of HC VR of Abs 014,
IDEVQLLESGGGLVKPGQSLKLSCAASGFTFSSYGMHWVRQPPGKGLEWVAVVSYDGSNKYYADSVQGRFTISRDNSKNTLYLQMN




FIG. 13
028; non-ID version is in FIG. 
SLRAEDTAVYYCAKDTKLRSLLYFEWLSSGLLDYWGQGAMVTVSS





2;






111
 29-ID
Table 4B
AA sequence of LC VR of Abs 014,
IDEIVMTQSPDSLAVSLGERATINCKSSQSVTFSYKNYLAWYQQKPGQPPKLLIYWASTRESGVPDRFSGSGSGTDFTLTISSLQA




FIG. 14
154, 157; non-ID version is in
EDVAVYYCQQYYRTPPTFGGGTKLDIK





FIG. 3;






98
 17-ID
Table 4B
AA sequence of HC VR of Ab 001,
IDEVQLLESGGGLVKPGQSLKLSCAASGFTFTSYGMHWVRQPPGKGLEWVAVVSYDGNYKYYADSVQGRFTISRDNSKNTLYLQMN




FIG. 13
009, 017, 025, 160, 186, 187, 
SLRAEDTAVYYCAKDSRLRSLLYFEWLSQGYFNPWGAGTTLTVSS





188, 189, 190, 191, 192, 193, 






202, 211; non-ID version is in 






FIG. 2;






112
 30-ID
Table 4B
AA sequence of LC VR of Abs 028,
IDEIVMTQSPDSLAVSLGERATINCKSSQSVTFDYKNYLAWYQQKPGQPPKLLIYWASTRESGVPDRFSGSGSGTDFTLTISSLQA




FIG. 14
155; non-ID version is in FIG. 
EDVAVYYCQQYYRTPPTFGGGTKLDIK





3;






99
 18-ID
Table 4B
AA sequence of HC VR of Abs 002,
IDEVQLLESGGGLVKPGQSLKLSCAASGFTFTSYGMHWVRQPPGKGLEWVAVLSYDGNYKYYADSVQGRFTISRDNSKNTLYLQMN




FIG. 13
010, B18, 026, 203, 212; non-ID
SLRAEDTAVYYCAKDSRLRSLLYFEWLSQGYFNPWGAGTTLTVSS





version is in FIG. 2;






113
 35-ID
Table 4B
AA sequence of LC VR of Ab 159; 
IDEIVMTQSPDSLAVSLGERATINCKSSQSVTWSYKNYLAWYQQKPGQPPKLLIYWASTRESGVPDRFSGSGSGTDFTLTISSLQA




FIG. 14
non-ID version is in FIG. 3;
EDVAVYYCQQYYRTPPTFGGGTKLDIK





100
 19-ID
Table 4B
AA sequence of HC VR of Abs 003,
IDEVQLLESGGGLVKPGQSLKLSCAASGFTFTTYAMHWVRQPPGKGLEWVAVLSYDGNYKYYADSVQGRFTISRDNSKNTLYLQMN




FIG. 13
011, 019, 027, 194, 195, 196, 
SLRAEDTAVYYCAKDSRLRSLLYFEWLSQGYFNPWGAGTTLTVSS





197, 198, 199, 200, 204, 213; 






non-ID version is in FIG. 2;






114
 31-ID
Table 4B
AA sequence of LC VR of Abs 001,
IDEIVMTQSPDSLAVSLGERATINCKSSQTVTFNYKNYLAWYQQKPGQPPKLLIYWASTRESGVPDRFSGSGSGTDFTLTISSLQA




FIG. 14
002, 003; non-ID version is in
EDVAVYYCQQHYRTPPSFGGGTKLDIK





FIG. 3;






101
 21-ID
Table 4B
AA sequence of HC VR of Abs 154,
IDEVQLLESGGGLVKPGQSLKLSCAASGFTFSSYGMHWVRQPPGKGLEWVAVVSYDGNNKYYADSVQGRFTISRDNSKNTLYLQMN




FIG. 13
155; non-ID version is in FIG. 
SLRAEDTAVYYCAKDSKLRSLLYFEWLSSGLLDYWGQGAMVTVSS





2;






115
 32-ID
Table 4B
AA sequence of LC VR of Abs 009,
IDEIVMTQSPDSLAVSLGERATINCKSSQTLSFNYKNYLAWYQQKPGQPPKLLIYWASTRESGVPDRFSGSGSGTDFTLTISSLQA




FIG. 14
010, 011; non-ID version is in
EDVAVYYCQQHYRTPPSFGGGTKLDIK





FIG. 3;






102
 22-ID
Table 4B
AA sequence of HC VR of Abs 157,
IDEVQLLESGGGLVKPGQSLKLSCAASGFTFTTYAMHWVRQPPGKGLEWVAVVSYDGNNKYYADSVQGRFTISRDNSKNTLYLQMN




FIG. 13
159; non-ID version is in FIG. 
SLRAEDTAVYYCAKDSKLRSLLYFEWLSSGLLDYWGQGAMVTVSS





2;






116
 33-ID
Table 4B
AA sequence of LC VR of Abs 017,
IDEIVMTQSPDSLAVSLGERATINCKSSQTVTFNYKNYLAWYQQKPGQPPKLLIYFASTRESGVPDRFSGSGSGTDFTLTISSLQA




FIG. 14
B18, 019; non-ID version is in 
EDVAVYYCQQHYRTPPSFGGGTKLDIK





FIG. 3;






103
 20-ID
Table 4B
AA sequence of HC VR of Ab 086; 
IDEVQLLESGGGLVKPGQSLKLSCAASGFTFTTYAMHWVRQPPGKGLEWVAVVSFDGNNRYYADSVQGRFTISRDNSKNTLYLQMN




FIG. 13
non-ID version is in FIG. 2;
SLRAEDTAVYYCAKDSQLRSLLYFEWLSSGVLDYWGQGAMVTVSS





117
 34-ID
Table 4B
AA sequence of LC VR of Abs 
IDEIVMTQSPDSLAVSLGERATINCKSSQTLSFNYKNYLAWYQQKPGQPPKLLIYFASTRESGVPDRFSGSGSGTDFTLTISSLQA




FIG. 14
025, 026, 027, 086; non-ID 
EDVAVYYCQQHYRTPPSFGGGTKLDIK





version is in FIG. 3;






104
 23-ID
Table 4B
AA sequence of HC VR of Abs 
IDEVQLLESGGGLVKPGQSLKLSCAASGFTFTSYGMHWVRQPPGKGLEWVAVVSYDGNYKYYADSVQGRFTISRDNSKNTLYLQMN




FIG. 13
210, 219; non-ID version is in 
SLRAEDTAVYYCAKDSKLRSLLYFEWLSQGYFNPWGAGTTLTVSS





FIG. 2;






118
 36-ID
Table 4B
AA sequence of LC VR of Ab 160;
IDEIVMSQSPDTLAVTLGERASINCKSSQTVTFNYKNYLAWYQQKPGQPPKVLIYWASARETGVPERFSGSGSGTDFTLTISSLQA




FIG. 14
non-ID version is in FIG. 3;
EDVAVYYCQQHYRTPPSFGQGTKLEIK





105
 24-ID
Table 4B
AA sequence of HC VR of Abs 
IDEVQLLESGGGLVKPGQSLKLSCAASGFTFTSYAMHWVRQPPGKGLEWVAVVSYDGNYKYYADSVQGRFTISRDNSKNTLYLQMN




FIG. 13
A001, A002, A003, A010, A011, 
SLRAEDTAVYYCAKDSRLRSLLYFEWLSQGYFNPWGQGTTLTVSS





031, 037; non-ID version is in 






FIG. 2;






119
 45-ID
Table 4B
AA sequence of LC VR of Abs 
IDDIQMTQSPSSLSASVGDRVTITCRSSQSITFNYKNYLAWYQQKPGKAPKLLIYWGSYLESGVPSRFSGSGSGTDFTLTISSLQP




FIG. 14
202, 203, 204, 210, 031, 032, 
EDFATYYCQQHYRTPPSFGQGTKVEIK





033, 034; non-ID version is in






FIG. 3;






106
 25-ID
Table 4B
AA sequence of HC VR of Abs 
IDQVQLLETGGGLVKPGQSLKLSCAASGFTFTSYAMHWVRQPPGKGLEWVAVVSYDGNYKYYADSVQGRFTISRDNSKNTLYLQMN




FIG. 13
004, 005, 006, 012, 013, 032, 
SLRAEDTAVYYCAKDSRLRSLLYFEWLSQGYFNPWGQGTTLTVSS





038, 043, 044, 045, 046, 047, 






048, 049, 050, 051, 052, 067, 






068, 069, 070, 073, 074, 075, 






076, 077; non-ID version is in






FIG. 2;






120
 46-ID
Table 4B
AA sequence of LC VR of Abs 
IDDIQMTQSPSSLSASVGDRVTITCRSSQSITFNYKNYLGWYQQKPGKAPKLLIYWGSYLESGVPSRFSGSGSGTDFTLTISSLQP




FIG. 14
211, 212, 213, 219, 037, 038,  
EDFATYYCQQHYRTPPSFGQGTKVEIK





039, 040; non-ID version is in






FIG. 3;






107
 26-ID
Table 4B
AA sequence of HC VR of Abs 
IDEVQLLESGGGLVKPGQSLKLSCAASGFTFTSYAMHWVRQPPGKGLEWVAVVSYDGNYKYYADSVQGRFTISRDNSKNTLYLQMN




FIG. 13
007, 008, A009, A14, 015, 033,
SLRAEDTAVYYCAKDSQLRTLLYFEWLSQGYFNPWGQGTTLTVSS





039; non-ID version is in FIG.






2;






121
 37-ID
Table 4B
AA sequence of LC VR of Abs 
IDEIVMTQSPDSLAVSLGERATINCKSSQTVTFNYKNYLAWYQQKPGQPPKLLIYWASTRESGVPDRFSGSGSGTDFTLTISSLQA




FIG. 14
186, 194; non-ID version is in
EDVAVYYCQQHYRTPPSFGTGTKLDIK





FIG. 3;






108
 27-ID
Table 4B
AA sequence of HC VR of Abs 
IDEVQLLESGGGLVKPGQSLKLSCAASGFTFTSYAMHWVRQPPGKGLEWVAVVSYDGNYKYYADSVQGRFTISRDNSKNTLYLQMN




FIG. 13
016, A017, C18, A019, 034, 040;
SLRAEDTAVYYCAKDSRLRTLLYFEWLSQGYFDPWGQGTTLTVSS





non-ID version is in FIG. 2;






122
 38-ID
Table 4B
AA sequence of LC VR of Abs 
IDEIVMTQSPDSLAVSLGERATINCKSSQTVTFNYKNYLAWYQQKPGQPPKLLIYWASTRESGVPDRFSGSGSGTDFTLTISSLQA




FIG. 14
187, 195; non-ID version is in
EDVAVYYCQQHYRTPPSFGSGTKLDIK





FIG. 3;






109
161-ID
Table 4B
AA sequence of HC VR consensus 
IDEVQLLESGGGLVKPGQSLKLSCAASGFTFSSYGMHWVRQPPGKGLEWVAVVSYDGSNKYYADSVQGRFTISRDNSKNTLYLQMN




FIG. 13
ID; non-ID version is in FIG. 
SLRAEDTAVYYCAKDSKLRSLLYFEWLSSGLLDYWGQGAMVTVSS





2;






123
 39-ID
Table 4B
AA sequence of LC VR of Abs 
IDEIVMTQSPDSLAVSLGERATINCKSSQTVTFNYKNYLAWYQQKPGQPPKLLIYWASTRESGVPDRFSGSGSGTDFTLTISSLQA




FIG. 14
188, 196; non-ID version is in
EDVAVYYCQQHYRTPPSFGQGTKLDIK





FIG. 3;






124
 40-ID
Table 4B
AA sequence of LC VR of Abs 
IDEIVMTQSPDSLAVSLGERATINCKSSQTVTFNYKNYLAWYQQKPGQPPKLLIYWASTRESGVPDRFSGSGSGTDFTLTISSLQA




FIG. 14
189, 197; non-ID version is in
EDVAVYYCQQHYRTPPSFGNGTKLDIK





FIG. 3;






125
 41-ID
Table 4B
AA sequence of LC VR of Abs 
IDEIVMTQSPDSLAVSLGERATINCKSSQTLSFNYKNYLAWYQQKPGQPPKLLIYWASTRESGVPDRFSGSGSGTDFTLTISSLQA




FIG. 14
190, 198; non-ID version is in
EDVAVYYCQQHYRTPPSFGTGTKLDIK





FIG. 3;






126
 42-ID
Table 4B
AA sequence of LC VR of Abs 
IDEIVMTQSPDSLAVSLGERATINCKSSQTLSFNYKNYLAWYQQKPGQPPKLLIYWASTRESGVPDRFSGSGSGTDFTLTISSLQA




FIG. 14
191, 199; non-ID version is in
EDVAVYYCQQHYRTPPSFGSGTKLDIK





FIG. 3;






127
 43-ID
Table 4B
AA sequence of LC VR of Abs 
IDEIVMTQSPDSLAVSLGERATINCKSSQTLSFNYKNYLAWYQQKPGQPPKLLIYWASTRESGVPDRFSGSGSGTDFTLTISSLQA




FIG. 14
192, 200; non-ID version is in
EDVAVYYCQQHYRTPPSFGQGTKLDIK





FIG. 3;






128
 44-ID
Table 4B
AA sequence of LC VR of Abs 
IDEIVMTQSPDSLAVSLGERATINCKSSQTLSFNYKNYLAWYQQKPGQPPKLLIYWASTRESGVPDRFSGSGSGTDFTLTISSLQA




FIG. 14
193; non-ID version is in FIG.
EDVAVYYCQQHYRTPPSFGNGTKLDIK





3;






129
 47-ID
Table 4B
AA sequence of LC VR of Abs 
IDDIVMTQSPDTLAVTLGERATIQCKSSQTVTFNYKNYLAWYQQKPGQPPKLLIYWASTRESGVPDRFSGSGSGTDFTLTITSLQA




FIG. 14
A001, 004, 007, 016
EDVAVYYCQQHYRTPPSFGQGTKLDIK





130
 48-ID
Table 4B
AA sequence of LC VR of Abs 
IDDIVMTQSPDTVAVTVGERATINCKSSQTVTFNYKNYLAWYQQKPGQPPKLLIYWASTRESGVPDRFSGSGSGTDFTLTISSLQA




FIG. 14
002, 005, 008, A017; non-ID 
EDVAVYYCQQHYRTPPSFGQGTKLDIK





version is in FIG. 3;






131
 49-ID
Table 4B
AA sequence of LC VR of Abs 
IDDIVMTQSPDTVAVTLGERATIDCKSSQTVTFNYKNYLAWYQQKPGQPPKLLIYWASTRESGVPDRFSGSGSGTDFTLTISSLQA




FIG. 14
A003, 006, A009, C18; non-ID 
EDVAVYYCQQHYRTPPSFGQGTKLDIK





version is in FIG. 3;






132
 50-ID
Table 4B
AA sequence of LC VR of Abs 
IDDIVMTQSPDTLAVTVGERATIRCKSSQTVTFNYKNYLAWYQQKPGQPPKLLIYWASTRESGVPDRFSGSGSGTDFTLTISSLQA




FIG. 14
A010 012, A14, A019; non-ID 
EDVAVYYCQQHYRTPPSFGQGTKLDIK





version is in FIG. 3;






133
 51-ID
Table 4B
AA sequence of LC VR of Ab 
IDDIVMTQSPDTLAVSRGERATIDCKSSQTVTFNYKNYLAWYQQKPGQPPKLLIYWASTRESGVPDRFSGSGSGTDFTLTISSLQA




FIG. 14
A011, 013, 015; non-ID version
EDEAVYYCQQHYRTPPSFGQGTKLDIK





is in FIG. 3;






134
 52-ID
Table 4B
AA sequence of LC VR of Abs 
IDDIQMTQSPSSLSASVGDRVTITCRSSQSITFDYKNYLAWYQQKPGKAPKLLIYWGSYLESGVPSRFSGSGSGTDFTLTISSLQP




FIG. 14
044, 071, 072, 078; non-ID 
EDFATYYCQQHYRTPPSFGQGTKVEIK





version is in FIG. 3;






135
 53-ID
Table 4B
AA sequence of LC VR of Ab 051;
IDDIQMTQSPSSLSASVGDRVTITCRSSQSITFNYKNYLAWYQQKPGKAPKLLIYWGSTLESGVPSRFSGSGSGTDFTLTISSLQP




FIG. 14
non-ID version is in FIG. 3;
EDFATYYCQQHYRTPPSFGQGTKVEIK





136
 54-ID
Table 4B
AA sequence of LC VR of Ab 049;
IDDIQMTQSPSSLSASVGDRVTITCRSSQSITFNYKNYLAWYQQKPGKAPKLLIYWGSHLESGVPSRFSGSGSGTDFTLTISSLQP




FIG. 14
non-ID version is in FIG. 3;
EDFATYYCQQHYRTPPSFGQGTKVEIK





137
 55-ID
Table 4B
AA sequence of LC VR of Ab 047;
IDDIQMTQSPSSLSASVGDRVTITCRSSQSITFNYKNYLAWYQQKPGKAPKLLIYWGSKLESGVPSRFSGSGSGTDFTLTISSLQP




FIG. 14
non-ID version is in FIG. 3;
EDFATYYCQQHYRTPPSFGQGTKVEIK





138
 56-ID
Table 4B
AA sequence of LC VR of Ab 050;
IDDIQMTQSPSSLSASVGDRVTITCRSSQSITFNYKNYLAWYQQKPGKAPKLLIYWGSDLESGVPSRFSGSGSGTDFTLTISSLQP




FIG. 14
non-ID version is in FIG. 3;
EDFATYYCQQHYRTPPSFGQGTKVEIK





139
 57-ID
Table 4B
AA sequence of LC VR of Ab 045;
IDDIQMTQSPSSLSASVGDRVTITCRSSQSITFNYKNYLAWYQQKPGKAPKLLIYWGSYLESGVPSRFSGSGSGTDFTLTISSLQP




FIG. 14
non-ID version is in FIG. 3;
EDVATYYCQQHYRTPPSFGQGTKVEIK





140
 58-ID
Table 4B
AA sequence of LC VR of Ab 048;
IDDIQMTQSPSSLSASVGDRVTITCRSSQSITFNYKNYLAWYQQKPGKAPKLLIYWGSYLESGVPSRFSGSGSGTDFTLTISSLQP




FIG. 14
non-ID version is in FIG. 3;
EDKATYYCQQHYRTPPSFGQGTKVEIK





141
 59-ID
Table 4B
AA sequence of LC VR of Ab 046;
IDDIQMTQSPSSLSASVGDRVTITCRSSQSITFNYKNYLAWYQQKPGKAPKLLIYWGSYLESGVPSRFSGSGSGTDFTLTISSLQP




FIG. 14
non-ID version is in FIG. 3;
EDDATYYCQQHYRTPPSFGQGTKVEIK





142
 60-ID
Table 4B
AA sequence of LC VR of Ab 043;
IDDIQMTQSPSSLSASVGDRVTITCRSSQSITFNYKNYLAWYQQKPGKAPKLLIYWGSYLESGVPSRFSGSGSGTDFTLTISSLQP




FIG. 14
non-ID version is in FIG. 3;
EDFATYYCQQYYRTPPSFGQGTKVEIK





143
 61-ID
Table 4B
AA sequence of LC VR of Ab 052;
IDDIQMTQSPSSLSASVGDRVTITCRSSQSITFNYKNYLAWYQQKPGKAPKLLIYWGSTRESGVPSRFSGSGSGTDFTLTISSLQP




FIG. 14
non-ID version is in FIG. 3;
EDFATYYCQQHYRTPPSFGQGTKVEIK





157
153-ID
Table 4B
AA sequence of LC VR of Ab 067;
IDDIQMTQSPSSLSASVGDRVTITCRSSQSITFQYKNYLAWYQQKPGKAPKLLIYWGSYLESGVPSRFSGSGSGTDFTLTISSLQP




FIG. 14
non-ID version is in FIG. 3;
EDFATYYCQQHYRTPPSFGQGTKVEIK





158
154-ID
Table 4B
AA sequence of LC VR of Ab 068;
IDDIQMTQSPSSLSASVGDRVTITCRSSQSITFRYKNYLAWYQQKPGKAPKLLIYWGSYLESGVPSRFSGSGSGTDFTLTISSLQP




FIG. 14
non-ID version is in FIG. 3;
EDFATYYCQQHYRTPPSFGQGTKVEIK





159
155-ID
Table 4B
AA sequence of LC VR of Abs 
IDDIQMTQSPSSLSASVGDRVTITCRSSQSITFEYKNYLAWYQQKPGKAPKLLIYWGSYLESGVPSRFSGSGSGTDFTLTISSLQP




FIG. 14
069, 079; non-ID version is in
EDFATYYCQQHYRTPPSFGQGTKVEIK





FIG. 3;






160
156-ID
Table 4B
AA sequence of LC VR of Ab 070;
IDDIQMTQSPSSLSASVGDRVTITCRSSQSITFDYKNYLAWYQQKPGKAPKLLIYWGSTRESGVPSRFSGSGSGTDFTLTISSLQP




FIG. 14
non-ID version is in FIG. 3;
EDFATYYCQQHYRTPPSFGQGTKVEIK





144
 62-ID
Table 4B
AA sequence of LC VR consensus
IDDIQMTQSPSSLSASVGDRVTITCRSSQSITFNYKNYLAWYQQKPGKAPKLLIYWGSYLESGVPSRFSGSGSGTDFTLTISSLQP




FIG. 14
ID; non-ID version is in FIG. 
EDFATYYCQQHYRTPPSFGQGTKVEIK





3;






63
VH16
Example 1
NT sequence of HC VR of Abs 
GAGGTACAGCTCCTCGAATCGGGAGGGGGACTGGTCAAACCCGGTCAATCGCTCAAACTCTCGTGTGCAGCGTCAGGTTTTACGTT





014, 028
CAGCTCATATGGGATGCACTGGGTCCGCCAGCCTCCGGGAAAGGGACTGGAGTGGGTGGCAGTCGTGTCGTATGACGGGAGCAATA






AGTACTACGCCGATTCAGTGCAAGGTCGGTTTACCATTTCGAGGGATAACAGCAAGAACACGCTCTACTTGCAGATGAACTCACTT






AGAGCGGAAGATACGGCTGTGTACTATTGCGCCAAAGACACAAAGCTGCGATCCCTGTTGTACTTCGAATGGTTGTCCTCGGGCTT






GCTTGACTATTGGGGGCAGGGCGCCATGGTCACAGTATCCAGCGCGTCGACTAAGGGGCCC





64
VL29
Example 1
NT sequence of LC VR of Abs 
GAGATCGTGATGACGCAGAGCCCCGATAGCCTCGCTGTCTCATTGGGGGAACGGGCCACGATTAACTGCAAATCCTCACAGTCGGT





014, 154, 157
GACTTTCAGCTATAAGAATTACCTGGCATGGTATCAGCAGAAGCCGGGTCAACCCCCAAAACTGTTGATCTACTGGGCCTCCACAC






GCGAGTCGGGAGTCCCGGACCGATTTTCGGGTTCAGGGTCCGGCACTGACTTTACCCTCACAATTTCATCGCTTCAAGCGGAGGAT






GTAGCAGTGTACTATTGTCAGCAGTATTACAGAACACCTCCCACCTTCGGAGGGGGAACGAAACTTGACATCAAGGGATCC





65
VL30
Example 1
NT sequence of LC VR of Abs 
NT: GAGATCGTGATGACGCAGAGCCCCGATAGCCTCGCTGTCTCATTGGGGGAACGGGCCACGATTAACTGCAAATCCTCACAGT





028, 155
CGGTGACTTTCGACTATAAGAATTACCTGGCATGGTATCAGCAGAAGCCGGGTCAACCCCCAAAACTGTTGATCTACTGGGCCTCC






ACACGCGAGTCGGGAGTCCCGGACCGATTTTCGGGTTCAGGGTCCGGCACTGACTTTACCCTCACAATTTCATCGCTTCAAGCGGA






GGATGTAGCAGTGTACTATTGTCAGCAGTATTACAGAACACCTCCCACCTTCGGAGGGGGAACGAAACTTGACATCAAGGGATCC





66
VH15
Example 1
NT sequence of HC VR of Ab A18
GAAGTGCAACTCCTCGAGTCAGGAGGAGGTTTGGTGAAACCGGGTCAGTCCTTGAAACTGAGCTGTGCAGCAAGCGGGTTCACGTT






TACGTCGTACGGCATGCACTGGGTACGGCAGCCTCCCGGGAAGGGACTTGAATGGGTCGCCGTCATCTCATACGACGGGTCGTACA






AATACTATGCGGATAGCGTGCAAGGTCGCTTCACAATTTCCCGGGACAATTCGAAGAATACACTGTATCTTCAGATGAACTCGCTC






AGGGCTGAGGACACGGCGGTCTATTACTGCGCGAAGGATTCGCGACTCAGATCCCTTTTGTACTTTGAGTGGCTGTCGCAGGGGTA






TTTCAACCCATGGGGAGCCGGAACCACTTTGACCGTATCAAGCGCGTCAACAAAGGGGCCC





67
VL28
Example 1
NT sequence of LC VR of Ab A18
GAAATTGTAATGACGCAGAGCCCTGATAGCCTTGCCGTGTCCCTGGGTGAGAGGGCGACAATCAATTGTAAGTCATCACAGTCGGT






CACGTACAACTACAAGAACTACCTGGCGTGGTATCAACAGAAACCCGGGCAGCCGCCCAAATTGCTCATCTATTGGGCTTCGACAC






GGGAGTCGGGTGTGCCAGACCGCTTCTCCGGGTCAGGATCGGGAACTGACTTCACGTTGACTATTTCGTCCCTCCAGGCAGAAGAT






GTAGCCGTCTACTATTGCCAACAGTATTACAGAACGCCGCCTACATTTGGAGGCGGGACCAAACTTGACATCAAGGGATCCGTGGC






CGCCCCCAGCGTCTTCATCTTCCCGCCCAGCGACGAGCAGCTGAAGTCGGGCACGGCCAGCGTGGTGTGCCTCCTGAACAACTTCT






ACCCCCGCGAGGCGAAGGTCCAGTGGAAGGTGGACAACGCCCTGCAGAGCGGGAACAGCCAGGAGAGCGTGACCGAGCAGGACTCG






AAGGACAGCACCTACAGCCTCAGCAGCACCCTGACGCTGAGCAAGGCCGACTACGAGAAGCACAAGGTCTACGCCTGCGAGGTGAC






CCACCAGGGGCTCTCGAGCCCCGTGACCAAGAGCTTCAACCGGGGCGAGTG





149
VL52
Example 1
NT sequence of EC VR of Abs 
GACATTCAGATGACTCAGTCGCCTTCGTCATTGTCCGCCTCCGTGGGTGATAGGGTCACGATCACGTGCCGGAGCAGCCAGTCCAT





044, 071, 072, 078
CACCTTCAATTACAAAAACTATTTGGCATGGTATCAACAGAAACCCGGAAAGGCGCCGAAGCTCCTGATCTACTGGGGTTCATATC






TTGAGTCGGGGGTGCCGTCGAGATTTTCGGGCAGCGGATCAGGGACGGATTTCACGCTGACCATTTCGTCACTCCAGCCCGAGGAC






TTTGCGACATATTACTGTCAACAGCACTACAGGACACCCCCATCTTTCGGACAGGGGACTAAAGTAGAAATCAAGGGATCCGTGGC






CGCCCCCAGCGTCTTCATCTTCCCGCCCAGCGACGAGCAGCTGAAGTCGGGCACGGCCAGCGTGGTGTGCCTCCTGAACAACTTCT






ACCCCCGCGAGGCGAAGGTCCAGTGGAAGGTGGACAACGCCCTGCAGAGCGGGAACAGCCAGGAGAGCGTGACCGAGCAGGACTCG






AAGGACAGCACCTACAGCCTCAGCAGCACCCTGACGCTGAGCAAGGCCGACTACGAGAAGCACAAGGTCTACGCCTGCGAGGTGAC






CCACCAGGGGCTCTCGAGCCCCGTGACCAAGAGCTTCAACCGGGGCGAGTGCTGA





150
VL45
Example 1
NT sequence of LC VR of Abs 
GACATTCAGATGACTCAGTCGCCTTCGTCATTGTCCGCCTCCGTGGGTGATAGGGTCACGATCACGTGCCGGAGCAGCCAGTCCAT





202, 203, 204, 210, 031, 032, 
CACCTTCAATTACAAAAACTATTTGGCATGGTATCAACAGAAACCCGGAAAGGCGCCGAAGCTCCTGATCTACTGGGGTTCATATC





033, 034
TTGAGTCGGGGGTGCCGTCGAGATTTTCGGGCAGCGGATCAGGGACGGATTTCACGCTGACCATTTCGTCACTCCAGCCCGAGGAC






TTTGCGACATATTACTGTCAACAGCACTACAGGACACCCCCATCTTTCGGACAGGGGACTAAAGTAGAAATCAAGGGATCCGTGGC






CGCCCCCAGCGTCTTCATCTTCCCGCCCAGCGACGAGCAGCTGAAGTCGGGCACGGCCAGCGTGGTGTGCCTCCTGAACAACTTCT






ACCCCCGCGAGGCGAAGGTCCAGTGGAAGGTGGACAACGCCCTGCAGAGCGGGAACAGCCAGGAGAGCGTGACCGAGCAGGACTCG






AAGGACAGCACCTACAGCCTCAGCAGCACCCTGACGCTGAGCAAGGCCGACTACGAGAAGCACAAGGTCTACGCCTGCGAGGTGAC






CCACCAGGGGCTCTCGAGCCCCGTGACCAAGAGCTTCAACCGGGGCGAGTGCTGAGAATTC





151
VH25
Example 1
NT sequence of HC VR of Abs 
CAGGTACAATTGCTTGAGACAGGTGGAGGACTCGTGAAGCCAGGTCAGTCATTGAAACTGAGCTGTGCCGCATCCGGGTTCACATT





004, 005, 006, 012, 013, 032,
CACTTCCTACGCGATGCACTGGGTCCGCCAGCCTCCCGGAAAGGGACTTGAGTGGGTCGCTGTGGTATCGTATGATGGGAATTACA





038, 043, 044, 045, 046, 047,
AATACTATGCAGACTCCGTGCAAGGCCGGTTTACGATTAGCAGGGACAACTCGAAGAATACCCTTTACCTCCAAATGAACTCGCTC





048, 049, 050, 051, 052, 067,
CGAGCGGAGGACACGGCGGTGTATTACTGCGCGAAGGATTCACGGTTGAGATCGCTGCTCTATTTTGAATGGTTGTCACAGGGGTA





068, 069, 070, 073, 074, 075,
CTTCAACCCGTGGGGTCAGGGAACAACACTGACCGTCAGCTCAGCCTCGACTAAAGGGCCCAGCGTGTTCCCGCTGGCCCCCAGCA





076, 077
GCAAGAGCACCAGCGGCGGGACCGCCGCCCTGGGCTGCCTCGTCAAGGACTACTTCCCCGAGCCCGTGACCGTGTCGTGGAACAGC






GGCGCGCTGACGAGCGGGGTCCACACCTTCCCGGCCGTGCTGCAGAGCAGCGGCCTCTACTCGCTGAGCAGCGTGGTCACCGTGCC






CAGCAGCAGCCTGGGGACCCAGACGTACATCTGCAACGTGAACCACAAGCCCTCGAACACCAAGGTCGACAAGAAGGTGGAGCCCC






CGAAGAGCTGCGACAAAACTCACACATGCCCACCGTGCCCAGGTACTGAACTCCTGGGGGGACCGTCAGTCTTCCTCTTCCCCCCA






AAACCCAAGGACACCCTCATGATCTCCCGGACCCCTGAGGTCACATGCGTGGTGGTGGACGTGAGCCACGAAGACCCTGAGGTCAA






GTTCAACTGGTACGTGGACGGCGTGGAGGTGCATAATGCCAAGACAAAGCCGCGGGAGGAGCAGTACAACAGCACGTACCGTGTGG






TCAGCGTCCTCACCGTCCTGCACCAGGACTGGCTGAATGGCAAGGAGTACAAGTGCAAGGTCTCCAACAAAGCCCTCCCAGCCCCC






ATCGAGAAAACCATCTCCAAAGCCAAAGGTGAGCCCCGAGAACCACAGGTGTACACCCTGCCCCCATCCCGGGATGAGCTGACCAA






GAACCAGGTCAGCCTGACCTGCCTGGTCAAAGGCTTCTATCCCAGCGACATCGCCGTGGAGTGGGAGAGCAATGGGCAGCCGGAGA






ACAACTACAAGACCACGCCTCCCGTGCTGGACTCCGACGGCTCCTTCTTCCTCTACAGCAAGCTCACCGTGGACAAGAGCAGGTGG






CAGCAGGGGAACGTCTTCTCATGCTCCGTGATGCATGAGGCTCTGCACAACCACTACACGCAGAAGAGCCTCTCCCTGTCTCCGGG






TAAATGA





152
VH24
Example 1
NT sequence of HC VR of Abs 
GAAGTACAATTGCTTGAGTCGGGTGGAGGACTCGTGAAGCCAGGTCAGTCATTGAAACTGAGCTGTGCCGCATCCGGGTTCACATT





A001, A002, A003, A010, A011, 
CACTTCCTACGCGATGCACTGGGTCCGCCAGCCTCCCGGAAAGGGACTTGAGTGGGTCGCTGTGGTATCGTATGATGGGAATTACA





031, 037
AATACTATGCAGACTCCGTGCAAGGCCGGTTTACGATTAGCAGGGACAACTCGAAGAATACCCTTTACCTCCAAATGAACTCGCTC






CGAGCGGAGGACACGGCGGTGTATTACTGCGCGAAGGATTCACGGTTGAGATCGCTGCTCTATTTTGAATGGTTGTCACAGGGGTA






CTTCAACCCGTGGGGTCAGGGAACAACACTGACCGTCAGCTCAGCCTCGACTAAAGGGCCCAGCGTGTTCCCGCTGGCCCCCAGCA






GCAAGAGCACCAGCGGCGGGACCGCCGCCCTGGGCTGCCTCGTCAAGGACTACTTCCCCGAGCCCGTGACCGTGTCGTGGAACAGC






GGCGCGCTGACGAGCGGGGTCCACACCTTCCCGGCCGTGCTGCAGAGCAGCGGCCTCTACTCGCTGAGCAGCGTGGTCACCGTGCC






CAGCAGCAGCCTGGGGACCCAGACGTACATCTGCAACGTGAACCACAAGCCCTCGAACACCAAGGTCGACAAGAAGGTGGAGCCCC






CGAAGAGCTGCGACGGTACCCACACATGCCCACCGTGCCCAGGTACTGAACTCCTGGGGGGACCGTCAGTCTTCCTCTTCCCCCCA






AAACCCAAGGACACCCTCATGATCTCCCGGACCCCTGAGGTCACATGCGTGGTGGTGGACGTGAGCCACGAAGACCCTGAGGTCAA






GTTCAACTGGTACGTGGACGGCGTGGAGGTGCATAATGCCAAGACAAAGCCGCGGGAGGAGCAGTACAACAGCACGTACCGTGTGG






TCAGCGTCCTCACCGTCCTGCACCAGGACTGGCTGAATGGCAAGGAGTACAAGTGCAAGGTCTCCAACAAAGCCCTCCCAGCCCCC






ATCGAGAAAACCATCTCCAAAGCCAAAGGTGAGCCCCGAGAACCACAGGTGTACACCCTGCCCCCATCCCGGGATGAGCTGACCAA






GAACCAGGTCAGCCTGACCTGCCTGGTCAAAGGCTTCTATCCCAGCGACATCGCCGTGGAGTGGGAGAGCAATGGGCAGCCGGAGA






ACAACTACAAGACCACGCCTCCCGTGCTGGACTCCGACGGCTCCTTCTTCCTCTACAGCAAGCTCACCGTGGACAAGAGCAGGTGG






CAGCAGGGGAACGTCTTCTCATGCTCCGTGATGCATGAGGCTCTGCACAACCACTACACGCAGAAGAGCCTCTCCCTGTCTCCGGG






TAAATGA





94
15
FIG. 1
AA sequence of HC of Ab A18
EVQLLESGGGLVKPGQSLKLSCAASGFTFTSYGMHWVRQPPGKGLEWVAVISYDGSYKYYADSVQGRFTISRDNSKNTLYLQMNSL






RAEDTAVYYCAKDSRLRSLLYFEWLSQGYFNPWGAGTTLTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNS






GALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKVEPPKSCDKTHTCPPCPGTELLGGPSVFLFPP






KPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAP






IEKTISKAKGEPREPQVYTLPPSRDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRW






QQGNVFSCSVMHEALHNHYTQKSLSLSPGK





95
28
FIG. 1
AA sequence of LC of Ab A18
EIVMTQSPDSLAVSLGERATINCKSSQSVTYNYKNYLAWYQQKPGQPPKLLIYWASTRESGVPDRFSGSGSGTDFTLTISSLQAED






VAVYYCQQYYRTPPTFGGGTKLDIKGSVAAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREAKVQWKVDNALQSGNSQESVTEQDS






KDSTYSLSSTLTLSKADYEKHKVYACEVTHQGLSSPVTKSFNRGE





145
n.a.
see text
AA sequence of LC CDR1 of Ab 
QSITFDYKNYLA





044






146
n.a.
see text
AA sequence of LC CDR1 of FI6
KSSQSVTFNYKNYLA





VK






147
n.a.
see text
AA sequence of LC CDR2 of FI6
WASARES





VK






148
n.a.
see text
AA sequence of LC CDR3 of FI6
QQHYRTPPT





VK






68
n.a.
see text
AA sequence of HC CDR1 of Abs 
SYAMH





044, 069, 032, 031






69
n.a.
see text
AA sequence of HC CDR2 of Abs 
VVSYDGNYKYYADSVQG





044, 069, 032, 031






70
n.a.
see text
AA sequence of HC CDR3 of Abs 
DSRLRSLLYFEWLSQGYFNP





044, 069, 032, 031






71
n.a.
see text
AA sequence of LC CDR1 of Abs 
QSITFNYKNYLA





032, 031






72
n.a.
see text
AA sequence of LC CDR2 of Abs 
WGSYLES





044, 069, 032, 031






73
n.a.
see text
AA sequence of LC CDR3 of Abs 
QQHYRTPPS





044, 069, 032, 031






74
n.a.
see text
AA sequence of HC FR1 of Ab 069
QVQLLETGGGLVKPGQSLKLSCAASGFTFT





75
n.a.
see text
AA sequence of HC FR2 of Ab 069
WVRQPPGKGLEWVA





76
n.a.
see text
AA sequence of HC FR3 of Ab 069
RFTISRDNSKNTLYLQMNSLRAEDTAVYYCAK





77
n.a.
see text
AA sequence of HC FR4 of Ab 069
WGQGTTLTVSS





78
n.a.
see text
AA sequence of LC FR1 of Ab 069
DIQMTQSPSSLSASVGDRVTITCRSS





79
n.a.
see text
AA sequence of LC FR2 of Ab 069
WYQQKPGKAPKLLIY





80
n.a.
see text
AA sequence of LC FR3 of Ab 069
GVPSRFSGSGSGTDFTLTISSLQPEDFATYYC





81
n.a.
see text
AA sequence of LC FR4 of Ab 069
FGQGTKVEIK





82
n.a.
see text
AA sequence of HC FR1 of Ab 031
EVQLLESGGGLVKPGQSLKLSCAASGFTFT





83
n.a.
see text
AA sequence of LC CDR1 of Ab 
KSSQSVTYNYKNYLA





A18 et al.






84
n.a.
see text
AA sequence of LC CDR2 of Ab 
WASTRES





A18 et al.






85
n.a.
see text
AA sequence of LC CDR3 of Ab 
QQYYRTPPT





A18 et al.






86
n.a.
see text
AA sequence of HC CDR1 of Ab 
SYGMH





A18 et al.






87
n.a.
see text
AA sequence of HC CDR2 of Ab 
VISYDGSYKYYADSVQG





A18 et al.






88
n.a.
see text
AA sequence of an HC CDR3
DSELRSLLYFEWLSQGYFNP





89
n.a.
see text
AA sequence of HC FR4 of Ab A18 
WGAGTTLTVSS





et al.






90
n.a.
see text
AA sequence of LC FR1 of Ab A18 
EIVMTQSPDSLAVSLGERATINC





et al.






91
n.a.
see text
AA sequence of LC FR2 of Ab A18
WYQQKPGQPPKLLIY





et al.






92
n.a.
see text
AA sequence of LC FR3 of Ab A18
GVPDRFSGSGSGTDFTLTISSLQAEDVAVYYC





et al.






93
n.a.
see text
AA sequence of LC FR4 of Ab A18
FGGGTKLDIK





et al.






171
n.a.
see text
AA sequence of HC FR4 of Ab 078
WGQGTTVTVSS





et al






172
n.a.
see text
AA sequence of LC CDR1 of Ab 
QSITFEYKNYLA





069






173
n.a.
see text
AA sequence of H3 HA1
QDLPGNDNSTATLCLGHHAVPNGTLVKTITDDQIEVTNATELVQSSSTGKICNNPHRILDGIDCTLIDALLGDPHCDVFQNETWDL






FVERSKAFSNCYPYDVPDYASLRSLVASSGTLEFITEGFTWTGVTQNGGSNACKRGPGSGFFSRLNWLTKSGSTYPVLNVTMPNND






NFDKLYIWGIHHPSTNQEQTSLYVQASGRVTVSTRRSQQTIIPNIGSRPWVRGLSSRISIYWTIVKPGDVLVINSNGNLIAPRGYF






KMRTGKSSIMRSDAPIDTCISECITPNGSIPNDKPFQNVNKITYGACPKYVKQNTLKLATGMRNVPEKQTR





174
n.a.
see text
AA sequence of H3 HA2
GLFGAIAGFIENGWEGMIDGWYGFRHQNSEGTGQAADLKSTQAAIDQINGKLNRVIEKTNEKFHQIEKEFSEVEGRIQDLEKYVED






TKIDLWSYNAELLVALENQHTIDLTDSEMNKLFEKTRRQLRENAEEMGNGCFKIYHKCDNACIESIRNGTYDHDVYRDEALNNRFQ






IKG





175
n.a.
FIG. 12
AA sequence of HC VR of FI6
QVQLVQSGGGVVQPGRSLRLSCVASGFTFSTYAMHWVRQAPGRGLEWVAVISYDGNYKYYADSVKGRFSISRDNSNNTLHLEMNTL






RTEDTALYYCAKDSQLRSLLYFEWLSQGYFDPWGQGTLVTVTS





176
n.a.
FIG. 12
AA sequence of HC VR of FI370
QVQLVQSGGGVVPPGRSLRLSCAASGFTFSTYGMHWVRQAPGKGLEWVAVISYDGNYKYYADSVRGRFTISRDNSKNTLNLDMNSL






RTEDTALYYCAKDSQLRSLLYFDWLSQGYFDHWGQGTLVTVSS





177
n.a.
FIG. 12
AA sequence of HC VR of FI6 
QVQLVESGGGVVQPGRSLRLSCAASGFTFSSYGMHWVRQAPGKGLEWVAVISYDGSNKYYADSVKGRFTISRDNSKNTLYLQMNSL





variant 1
RAEDTAVYYCAKDSQLRSLLYFDWLSQGYFDYWGQGTLVTVSS





178
n.a.
FIG. 12
AA sequence of HC VR of FI6 
QVQLVESGGGVVQPGRSLRLSCAASGFTFSTYAMHWVRQAPGKGLEWVAVISYDANYKYYADSVKGRFTISRDNSKNTLYLQMNSL





variant 3
RAEDTAVYYCAKDSQLRSLLYFEWLSQGYFDYWGQGTLVTVSS





179
n.a.
FIG. 12
AA sequence of HC VR of FI6/370
QVQLVQSGGGVVQPGRSLRLSCAASGFTFSTYGMHWVRQAPGKGLEWVAVISYDGNYKYYADSVKGRFTISRDNSKNTLYLEMNSL






RTEDTALYYCAKDSQLRSLLYFDWLSQGYFDHWGQGTLVTVSS





180
n.a.
FIG. 12
AA sequence of kappa LC VR of 
DIQMTSQPDSLAVSLGARATINCKSSQSVTFNYKNYLAWYQQKPGQPPKVLIYWASARESGVPDRFSGSGSGTDFTLTISSLQAED





FI6
VAVYYCQQHYRTPPTFGQGTKVEIK





181

See text
AA sequence of H1 HA1
TNADTICIGYHANNSTDTVDTVLEKNVTVTHSVNLLEDSHNGKLCKLKGIAPLQLGKCNIAGWLLGNPECDLLLTASSWSYIVETS






NSENGTCYPGDFIDYEELREQLSSVSSFEKFEIFPKTSSWPNHETTKGVTAACSYAGASSFYRNLLWLTKKGSSYPKLSKSYVNNK






GKEVLVLWGVHHPPTGTDQQSLYQNADAYVSVGSSKYNRRFTPEIAARPKVRDQAGRMNYYWTLLEPGDTITFEATGNLIAPWYAF






ALNRGSGSGIITSDAPVHDCNTKCQTPHGAINSSLPFQNIHPVTIGECPKYVRSTKLRMATGLRNIPSIQS





182

See text
AA sequence of H1 HA2
GLFGAIAGFIEGGWTGMIDGWYGYHHQNEQGSGYAADQKSTQNAIDGITNKVNSVIEKMNTQFTAVGKEFNNLERRIENLNKKVDD






GFLDIWTYNAELLVLLENERTLDFHDSNVRNLYEKVKSQLKNNAKEIGNGCFEFYHKCDDACMESVRNGTYDYPKYSEESKLNREE






IDGVKLESMGVYQILAIYSTVASSLVLLVSLGAISFWMCSNGSLQCRICI









EXAMPLES
Example 1: Safety and Upper Respiratory Pharmacokinetics of the Hemagglutinin Antibody VIS410 Support Treatment and Prophylaxis Based on Population Modeling of Seasonal Influenza A Outbreaks
Summary

Background.


Seasonal influenza is a major public health concern in vulnerable populations. Monoclonal antibody therapies represent a modality for treatment and prophylaxis given their safe profile and broad neutralizing ability.


Methods.


Using a single-ascending dose study (n=41) at dose levels from 2 mg/kg-50 mg/kg, the safety and pharmacokinetics (e.g., serum and upper respiratory pharmacokinetics) of a broadly-neutralizing antibody (VIS410, also known as Ab 044 herein) against influenza A were characterized (ClinicalTrials.gov identifier NCT02045472). The primary endpoints were safety and tolerability of VIS410 compared to placebo. An epidemic microsimulation model was developed for testing the ability of VIS410 to mitigate attack rates and severe disease in at risk-populations.


Findings.


VIS410 was found to be generally safe and well-tolerated at all dose levels, from 2-50 mg/kg. Overall, 27 of 41 subjects (65.9%) reported a total of 67 treatment emergent adverse events (TEAEs). TEAEs were reported by 20 of 30 subjects (66.7%) who received VIS410 and by 7 of 11 subjects (63.6%) who received placebo. 14 of 16 TEAEs related to study drug were considered mild (Grade 1) and 2 were moderate (Grade 2). Two subjects (1 subject who received 30 mg/kg VIS410 and 1 subject who received placebo) experienced serious AEs (grade 3 or 4 TEAEs) that were not related to study drug. VIS410 exposure was approximately dose-proportional with a mean half-life of 12.9 days. Mean VIS410 Cmax levels in the upper respiratory tract were 20.0 and 25.3 μg/ml at the 30 mg/kg and 50 mg/kg doses, respectively, with corresponding serum Cmax levels of 980.5 and 1316 μg/mL. Using these pharmacokinetic data, the microsimulation model showed that median attack rate reductions ranged from 8.6% (interquartile range (IQR): 4.7%-11.0%) for 2% coverage to 22.6% (IQR: 12.7-30.0%) for 6% coverage. The overall benefits to the elderly, a vulnerable subgroup, are largest when VIS410 is distributed exclusively to elderly individuals, resulting in reductions in hospitalization rates between 11.4% (IQR: 8.2%-13.3%) for 2% coverage and 30.9% (IQR: 24.8%-35.1%) for 6% coverage among those more than 65 years of age.


Interpretation.


VIS410 was generally safe and well tolerated and had good relative exposure in both serum and upper respiratory tract, supporting its use as either a single-dose therapeutic or prophylactic for influenza A. Including VIS410 prophylaxis among the public health interventions for seasonal influenza can be used to lower attack rates and substantially reduce hospitalizations in individuals over the age of 65.


To summarize, in this Example, the safety, tolerability, and pharmacokinetics of a broadly neutralizing, stalk-binding monoclonal antibody (VIS410) against Influenza A were investigated in a Phase 1 clinical trial. Based on these results and preclinical data, a mathematical modeling approach was used to investigate whether VIS410 could be used prophylactically to lessen the burden of a seasonal influenza epidemic and to protect at-risk groups from associated complications.


VIS410 is a broadly neutralizing monoclonal antibody that was engineered to bind a conserved region on the influenza A hemagglutinin protein that is used by the virus to bind and enter infected cells. VIS410 has a direct mechanism of action, inhibiting HA-mediated cell fusion, neutralizing the virus and preventing cell infection. For a drug to be an effective prophylactic against seasonal influenza it should have a strong neutralizing effect against both group 1 viruses, such as H1N1, and group 2 viruses, such as H3N2, target an epitope that is conserved and widely shared among influenza subtypes, have a PK/PD profile that affords sufficient protection for a typical flu season, and have a good safety profile. The pre-clinical and phase 1 clinical data demonstrates that VIS410 possesses these properties. It was shown that VIS410 is safe and well tolerated in a phase 1 clinical trial in healthy adult volunteers who were given a single infusion of the drug. Measurements of the drug levels of VIS410 in the upper respiratory tract demonstrated that protective levels were achieved at the site of influenza infection. Given the phase 1 trial results, Epidemic modeling analyses indicate that for a sufficiently potent and long-lasting antibody, such as VIS410, that prophylactic administration to 4-6% of the population, focused on high risk individuals (e.g., elderly individuals), would be sufficient to lower (e.g., substantially suppress overall) hospitalizations related to severe influenza. Seasonal influenza A infection results in significant morbidity and mortality especially in high risk groups such as the elderly. Notably, given the current state-of-the-art in the production of antibodies, it is possible to rapidly ensure availability of adequate supply of monoclonal antibody to protect such a population during an influenza season in a much shorter time scale than that for production of a vaccine, incorporating novel strains (generally >6 months).


Introduction

Severe influenza occurs each winter especially in high-risk groups such as young children, older adults, patients with pulmonary conditions, inflammatory conditions, malignancies, and pregnant women (Newton et al. The American journal of managed care 2000; 6(5 Suppl): S265-75; Schanzer et al. Vaccine 2008; 26(36): 4697-703). Despite available therapy with neuraminidase inhibitors, including oseltamivir, zanamivir, and peramivir; 10%-44% of hospitalized patients require intensive care and 25%-50% of these patients die. In the United States, it is estimated that as many as to 400,000 patients are hospitalized with influenza each year, with as many as 50,000 deaths per year (Centers for Disease Control U.; Hamborsky et al., editors. Epidemiology and Prevention of Vaccine-Preventable Diseases. 13th ed. Washington, D.C.: Public Health Foundation; 2015). Furthermore, as evidenced by pandemic influenza A infections such as the 2009 “swine flu” pandemic, newly emerging influenza subtypes represent a considerable threat to global public health as they have the potential to cause significant morbidity and mortality.


The majority of the severe disease burden during seasonal influenza is experienced by individuals over the age of 65, who are susceptible to a number of complications following infection with influenza virus (Reed et al. PloS one 2015; 10(3): e0118369; Thompson et al. Jama 2004; 292(11): 1333-40). Currently available public health interventions have not significantly mitigated disease burden for the elderly. Vaccination with trivalent or tetravalent killed influenza has historically had lower measured efficacy in elderly individuals compared to adults and children (Darvishian et al. The Lancet Infectious diseases 2014; 14(12): 1228-39; Breteler Vaccine 2013; 31(45): 5168-77; Osterholm The Lancet Infectious diseases 2012; 12(1): 36-44). Prophylaxis or early treatment with neuraminidase inhibitors are the current de facto standard of care however, some controversy exists as to whether a direct link can be established between early oseltamivir treatment and lower hospitalization rates (Jefferson et al. Bmj 2014; 348: g2545). Based on these shortfalls in care, there is a need to develop countermeasures to reduce or mitigate the effects of influenza in the elderly and other susceptible populations.


The benefits of broadly neutralizing antibodies are that they can protect elderly individuals from influenza infection regardless of immune response and potentially provide a reliable option when considering the vaccine mismatches that occur against influenza every three to five years. Using an antibody engineering approach, a broadly neutralizing antibody (VIS410) that targets a unique, conserved epitope on influenza hemagglutinin and binds to and neutralizes influenza A virus across group 1 and group 2 subtypes was developed. In vitro, VIS410 has been shown to neutralize groups 1 and 2 influenza strains; over 40 different virus strains have been tested to date, with EC50 values ranging from 0.1-˜60 μg/mL and representing broad temporal/geographical, subtype, and epitope diversity (Tharakaraman et al. Proc Natl Acad Sci USA. 2015; 112(35):10890-5; Baranovich et al. Antimicrob Agents Chemother. 2016; pii: AAC.02457-15). Additionally, in vivo studies in mouse models demonstrated that VIS410 administered as a prophylactic or therapeutic protects mice challenged with lethal doses of influenza A, including A/Puerto Rico/8/1934 [H1N1], A/California/04/2009 [H1N1], A/Victoria/3/1975 [H3N2], and A/Vietnam/1203/2004 [H5N1]. VIS410 also demonstrated protection against newly emerging pathogenic H7N9 strains, A/Anhui/1/2013 and oseltamivir-resistant A/Shanghai/1/2013 in a lethal BALB/c mouse model (Baranovich et al. Antimicrob Agents Chemother. 2016; pii: AAC.02457-15). VIS410 is being developed as a single dose treatment for hospitalized patients with influenza A is currently in phase 2 studies.


Described herein are the safety and pharmacokinetics of VIS410 in the serum and the upper respiratory tract, the primary target organ of infection of influenza A. Furthermore, this information was utilized to model the application of a broadly neutralizing antibody, such as VIS410, during an influenza outbreak to mitigate severe disease, especially for at risk-populations. Evidence has been provided that VIS410 is generally safe and well-tolerated in healthy subjects with protective levels of antibody achieved in the upper respiratory tract, and that it has a pharmacokinetic/pharmacodynamic (PK/PD) profile that may allow it to be used as a prophylactic during or prior to a period of high influenza activity. Taken together, these data support the development of a broadly neutralizing monoclonal antibody as a strategy for reducing the severity of seasonal influenza.


Methods

Production of Antibody.


VIS410 was produced under current Good Manufacturing Practice (cGMP) at Gallus Biopharmaceuticals (Princeton, N.J.) in a CHO cell line. After production at a 200 L scale, VIS410 was purified by protein A and ion exchange polishing steps. Testing of bulk drug substance indicated that the material was >99% monomer, containing <0.1 μg/mg residual DNA and <0.1 ng/mg of host cell proteins. VIS410 materials were formulated at 25 mg/mL in in 40 mM Citrate-Sodium Phosphate, 150 mM NaCl, pH 6.0, containing 0.025% Tween-80.


Phase I Clinical Trial.


A Phase 1, double-blind, placebo-controlled, single ascending dose-escalation study was completed in healthy adult subjects (ClinicalTrials.gov identifier NCT02045472). This study was conducted according to the International Conference on Harmonisation harmonised tripartite guideline E6(R1): Good Clinical Practice. Institutional Research Board approval for the study was obtained in writing before the study began. The primary endpoint for the study was the safety and tolerability of VIS410 compared to placebo and the secondary endpoint was the serum pharmacokinetics of a single dose of VIS410. Eligible subjects were admitted to the clinic for dose administration and were discharged 24-hours post-infusion. Overall, 30 subjects were dosed with VIS410 and 11 subjects were dosed with a placebo control infusion. Nine subjects were dosed in the first cohort (Cohort 1); 6 subjects received VIS410 (2 mg/kg) and 3 subjects received placebo (sodium chloride 0.9%). Eight subjects were dosed in the subsequent cohorts (Cohorts 2 through 5) and were randomly assigned in a 6:2 ratio to receive either VIS410 or placebo. The detailed phase 1 protocol is also presented herein.


Briefly, in the first cohort (Cohort 1) the first four sentinel subjects were randomly assigned to receive either VIS410 (2 mg/kg; n=2) or placebo (n=2) and received study drug at least 48 hours before the remaining subjects in the cohort were dosed. After the investigator had assessed that the infusions were well tolerated, the remaining subjects in the cohort were dosed concurrently (VIS410 n=4 and placebo n=1). In each subsequent cohort, the first 3 subjects were randomly assigned to receive either VIS410 (n=2) or placebo (n=1) and received study drug at least 48 hours before the remaining subjects in the cohort were dosed. After the investigator had assessed that the infusions were well tolerated, the remaining members of the cohort (VIS410 n=4 and placebo n=1) were dosed concurrently. Dose escalation to the next dosing level occurred after the Safety Monitoring Committee (SMC) comprised of the investigator, an independent medical monitor, and the sponsor reviewed the safety data through Day 7 after the infusion.


Assessment of safety by the SMC was determined from vital sign measurements; physical examinations; hematology, chemistry, and urinalysis laboratory testing; 12-lead triplicate electrocardiograms (ECGs); use of concomitant medications; and review of adverse events (AEs). Blood samples for pharmacokinetic (PK) analysis and for assessment of antidrug antibodies (ADA) to VIS410 were obtained before and after the infusion during the 120-day study period (Days 1, 2, 3, 7, 14±1, 28±3, 56±7, and 120±7). Nasopharyngeal (NP) swabs, to assess upper respiratory VIS410 concentrations, were collected before and after the infusion from subjects in the 15, 30, and 50 mg/kg cohorts (Days 1, 3 and 7).


Pharmacokinetic and Antidrug Antibody Assays.


Blood samples were collected at the time points described herein. Serum was aliquoted and stored at −20° C. to −80° C. All samples were tested for IgG antibody concentrations using an enzyme-linked immunosorbent assay. Nasopharyngeal swabs (one from each nostril) for the analysis of the local concentration of VIS410 were taken using COPAN flocked swabs from subjects in the 15, 30, and 50 mg/kg cohorts at the time points described herein. The swabs from each nostril were combined in 1 transport tube containing 3 mL COPAN Universal Transport Medium and stored at −70° C. Samples were tested for VIS410 by an immunoassay. The samples for ADA analysis were collected in serum separator tubes. Serum was aliquoted and stored at −20° C. to −80° C. The enzyme-linked immunosorbent assay was performed. Descriptive statistics were used to summarize data between groups. Statistical comparisons of the frequency of adverse events for placebo versus VIS410 receiving subjects used Fisher's exact test. All statistical analyses were conducted using SAS® software Version 9.3 (SAS Institute, Inc, Cary, N.C.), and the PK analysis was conducted using Phoenix WinNonlin® Version 6.2.1 (Pharsight Corporation, St Louis, Mo.).


Individual-Based Population Model.


An individual-based microsimulation was developed based on a previously developed model, and is similar in structure and design to an array of microsimulation models that have been developed over the past decade (Boni et al. Philosophical transactions of the Royal Society of London Series B, Biological sciences 2013; 368(1614): 20120207; Ferguson et al. Nature 2005; 437(7056): 209-14; Germann et al. Proceedings of the National Academy of Sciences of the United States of America 2006; 103(15): 5935-40; Longini et al. Science 2005; 309(5737): 1083-7). Individual-based microsimulation methods were used to test the population-level effects of deploying VIS410 during a typical winter influenza epidemic and to perform sensitivity analyses on key unknown parameters.


Briefly, the model simulates an age-structured population of one million individuals living in a city with 100 pre-defined neighborhoods or locations. Daily work commutes, random within-city travel, household structure, pre-existing immunity, and age-based social contacts are included in the model. Influenza infection and potential hospitalization are modeled by randomly infecting individuals by location or household, in proportion to the current level of infections and contacts in that location or household. Infection, seasonality, contact structure, hospitalization, and the clinical course and epidemiology of influenza in the model were validated using characteristics of past influenza epidemics of influenza A, as described herein.


In the microsimulation, VIS410 was deployed as a population-wide prophylaxis strategy. A small percentage of individuals received VIS410 prophylaxis (between 0% and 6%) in the early stages of the epidemic, and two modes of distribution were included: randomly to all individuals or randomly to only elderly individuals (>65 years old). The distribution time was varied between eight weeks prior to the epidemic peak and the date of the modelled epidemic peak. VIS410 levels in individuals were modeled using an exponential decay function, with a half-life of 13 days. VIS410 was modeled to be administered at a level that was 8-fold over a minimally protective dose of approximately 1-2 mg/kg based on preclinical estimates, corresponding to over 3 half-lives of protection. Levels of VIS410 over the protective threshold confer a 90% reduction in the probability of being infected, with the protection decreasing exponentially as a function of VIS410 levels below the minimally protective threshold. Levels of VIS410 below 0.1-fold of the protective threshold were considered to be non-protective. This behavior is shown in FIG. 4.


Results


VIS410 is an engineered human IgG1 antibody that targets a unique, conserved conformational epitope on the stem of Influenza A virus HA protein. Previous studies have identified that VIS410 has broad reactivity against both group 1 and group 2 influenza A. A bioinformatics analysis of over 26,500 H1N1 and H3N2 HA sequences demonstrates the conserved nature of the targeted epitope within H1 and H3 subgroups (Tharakaraman et al. Proc Natl Acad Sci USA. 2015; 112(35):10890-5). FIG. 8 shows the observed residue composition of this epitope based on a sequence analysis of currently circulating strains (collected since 2012), supporting the pre-clinical in vitro and in vivo analyses which indicates that VIS410 is effective against a broad panel of seasonal H1 and H3 influenza viruses as well as H7N9 virus.


A Phase 1, placebo-controlled, single ascending dose study of VIS410 in healthy volunteers was initiated at a single site in North America. Five cohorts were dosed with levels ranging from 2 to 50 mg/kg (Table 5). A total of 41 subjects were enrolled in the phase 1 study. Overall, 36 subjects (87.8%) completed the study and 5 subjects (12.2%) discontinued early. All 41 subjects (100.0%) who received study drug (VIS410 or placebo) were included in the safety analysis set. All 30 subjects (100.0%) who received a dose of VIS410 and had at least one evaluable PK parameter were included in the PK analysis set. Five subjects (12.2%) withdrew consent (4 of 30 subjects [13.3%] who received VIS410 and 1 of 11 subject [9.1%] who received placebo). For 1 subject (Subject 402; 30 mg/kg VIS410), the investigator was unblinded to the subject's treatment due to serious adverse events (SAEs) of leukopenia and herpes simplex esophagitis. This SAE was ultimately found to be unrelated to the study drug as a primary herpes simplex virus type 1 infection was confirmed based on analysis of pre and post event serology.









TABLE 5







Summary of Subject Disposition











VIS410


















2
5
15
30
50






mg/kg
mg/kg
mg/kg
mg/kg
mg/kg
Total
Placebo
Overall



(n = 6)
(n = 6)
(n = 6)
(n = 6)
(n = 6)
(N = 30)
(N = 11)
(N = 41)





Total number of










subjects, No. (%)










Completed
6
4
5
5
6
26
10
36



(100.0)
(66.7)
(83.3)
(83.3)
(100.0)
(86.7)
(90.9)
(87.8)


Discontinued
0
2
1
1
0
4
1
5




(33.3)
(16.7)
(16.7)

(13.3)
(9.1)
(12.2)


Primary reason for










discontinuation,










No. (%)










Subject withdrew
0
2
1
1
0
4
1
5


consent

(33.3)
(16.7)
(16.7)

(13.3)
(9.1)
(12.2)


Study Population, No.










(%)










Safety analysis seta
6
6
6
6
6
30
11
41



(100.0)
(100.0)
(100.0)
(100.0)
(100.0)
(100.0)
(100.0)
(100.0)


Pharmacokinetic
6
6
6
6
6
30
0
30


analysis setb
(100.0)
(100.0)
(100.0)
(100.0)
(100.0)
(100.0)

(73.2)





Note: Percentages were based on the number of subjects within each group and overall.



aThe safety analysis set included all subjects who received a dose of VIS410 or placebo.




bThe pharmacokinetic analysis set included all subjects who received a dose of VIS410 and had at least 1 evaluable pharmacokinetic parameter.







Safety Results.


Overall, 27 of 41 subjects (65.9%) reported a total of 67 treatment emergent adverse events (TEAEs). TEAEs were reported by 20 of 30 subjects (66.7%) who received VIS410 and by 7 of 11 subjects (63.6%) who received placebo. 18 of 41 subjects (43.9%) overall (16 of 30 subjects [53.3%] who received VIS410 and 2 of 11 subjects [18.2%] who received placebo; p>0.05) experienced TEAEs related to study drug. 14 of 16 TEAEs related to study drug were considered mild (Grade 1) and 2 were moderate (Grade 2).


Overall, the highest percentage of subjects that reported TEAEs were classified as nervous system disorders (11 subjects; 26.8%) followed by gastrointestinal (GI) disorders and infections and infestations (10 subjects; 24.4% each). The percentage of subjects reporting nervous system disorders was similar following administration of VIS410 (7 of 30 subjects; 23.3%) compared with placebo (4 of 11 subjects; 36.4%) and did not reach statistical significance; no notable differences were observed across VIS410 dose levels. Gastrointestinal disorders were reported by subjects who received VIS410 only (10 of 30 subjects; 33.3% for VIS410 receiving subjects, compared to 0% for placebo, p<0.05). The percentage of subjects reporting GI disorders was highest in the 50 mg/kg VIS410 cohort (5 of 6 subjects; 83.3%). The percentage of subjects reporting infections and infestations was similar following administration of VIS410 (6 of 30 subjects; 20.0%) compared with placebo (4 of 11 subjects; 36.4%); no notable differences were observed across VIS410 dose levels (See Tables 6 and 7).









TABLE 6







Summary of Gastrointestinal Adverse Events











VIS410


















2
5
15
30
50





Adverse
mg/kg
mg/kg
mg/kg
mg/kg
mg/kg
Total
Placebo
Overall


Event
(N = 6)
(N = 6)
(N = 6)
(N = 6)
(N = 6)
(N = 30)
(N = 11)
(N = 41)


















Diarrhea
0
2
1
2
5
10
0
10




(33.3%)
(16.7%)
(33.3%)
(83.3%)
(33.3%)

(24.4%)


Nausea
0
0
0
2
2
4
0
4






(33.3%)
(33.3%)
(13.3%)

(9.8%)


Vomiting
0
0
0
0
2
2
0
2







(33.3%)
(6.7%)

(4.9%)
















TABLE 7







Summary of Gastrointestinal Adverse Events by Severity











VIS410


















2
5
15
30
50





Adverse
mg/kg
mg/kg
mg/kg
mg/kg
mg/kg
Total
Placebo
Overall


Event
(N = 6)
(N = 6)
(N = 6)
(N = 6)
(N = 6)
(N = 30)
(N = 11)
(N = 41)





Diarrhea
0
2
1
2
5
10
0
10




(33.3%)
(16.7%)
(33.3%)
(83.3%)
(33.3%)

(24.4%)


Grade 1
0
2
1
2
5
10
0
10




(33.3%)
(16.7%)
(33.3%)
(83.3%)
(33.3%)

(24.4%)


Grade 2
0
0
0
0
0
 0
0
 0


Grade 3
0
0
0
0
0
 0
0
 0


Grade 4
0
0
0
0
0
 0
0
 0


Nausea
0
0
0
2
2
 4
0
 4






(33.3%)
(33.3%)
(13.3%)

(9.8%)


Grade 1
0
0
0
2
1
 3
0
 3






(33.3%)
(16.7%)
(10.0%)

(7.3%)


Grade 2
0
0
0
0
1
 1
0
 1







(16.7%)
(3.3%)

(2.4%)


Grade 3
0
0
0
0
0
 0
0
 0


Grade 4
0
0
0
0
0
 0
0
 0


Vomiting
0
0
0
0
2
 2
0
 2







(33.3%)
(6.7%)

(4.9%)


Grade 1
0
0
0
0
1
 1
0
 1







(16.7%)
(3.3%)

(2.4%)


Grade 2
0
0
0
0
1
 1
0
 1







(16.7%)
(3.3%)

(2.4%)


Grade 3
0
0
0
0
0
 0
0
 0


Grade 4
0
0
0
0
0
 0
0
 0





*Grade 1 = mild;


Grade 2 + moderate;


Grade 3 = Severe;


Grade 4 = Life-threatening






Additional summary safety data from the Phase I study is shown in Tables 8 and 9.









TABLE 8







Subjects with Confirmed ADA and TEAE















ADA



Onset Time



VIS410
Titer



from Start


Subject #
Cohort
(Day)
TEAE
Severity*
Relationship
of Infusion

















204
2 mg/kg
10
Headache
Grade 2
Related
7
hrs




(Day 120)



50
mins





Headache
Grade 1
Not related
50
days


205
2 mg/kg
10
None








(Day 14)
reported








40









(Day 120)







208
2 mg/kg
40
Diarrhea
Grade 1
Related
11
hrs




(Day 120)



21
mins


305
15 mg/kg
10
None








(Day 120)
reported









*Grade 1 = mild;


Grade 2 + moderate;


Grade 3 = severe;


Grade 4 = serious adverse event






Subjects who developed clinically significant upper respiratory infections had viral testing of their nasopharyngeal swabs by the site investigator for the duration of the study (Day 120). None were found to have influenza although the 30 mg/kg and 50 mg/kg cohorts were dosed from December 2014 to January 2015 where, based on state reported epidemiology, there were high relative incidences of influenza A and other respiratory virus infections. A summary table of infections and infestations lists the relevant infections and dose groups (See Table 9). The most frequently reported TEAEs overall were diarrhea reported by 10 of 41 subjects (24.4%) and headache reported by 8 of 41 subjects (19.5%). The most frequently reported drug-related TEAE overall was diarrhea (9 subjects; 22.0%) that generally occurred following infusion and spontaneously resolved within 24 hours. Most GI TEAEs were mild (Grade 1) with the exception of 2 subjects in the 50 mg/kg dose that had moderate (Grade 2) grading of their symptoms.









TABLE 9







Summary of Infections and Infestations











VIS410


















2
5
15
30
50
Total





mg/kg
mg/kg
mg/kg
mg/kg
mg/kg
VIS410
Placebo
Overall


Adverse Event
(N = 6)
(N = 6)
(N = 6)
(N = 6)
(N = 6)
(N = 30)
(N = 11)
(N = 41)





All Infections
0
0
1
3
2
6
4
10


and


(16.7%)
(50.0%)
(33.3%)
(20.0%)
(36.4%)
(24.4%)


Infestations










Upper
0
0
0
1
1
2
2
 4


respiratory tract



(16.7%)
(16.7%)
(6.7%)
(18.2%)
(9.8%)


infection










Appendicitis
0
0
0
0
0
0
1
 1









(9.1%)
(2.4%)


Corona virus
0
0
0
1
0
1
0
 1


infection



(16.7%)

(3.3%)

(2.4%)


Gastroenteritis
0
0
1
0
0
1
0
 1





(16.7%)


(3.3%)

(2.4%)


Herpes simplex
0
0
0
1
0
1
0
 1


esophagitis



(16.7%)

(3.3%)

(2.4%)


Herpes zoster
0
0
0
0
0
0
1
 1









(9.1%)
(2.4%)


Rhinitis
0
0
0
0
1
1
0
 1







(16.7%)
(3.3%)

(2.4%)


Rhinovirus
0
0
0
0
0
0
1
 1


Infection






(9.1%)
(2.4%)









There were no subjects with SAEs that were related to study drug. Approximately 3 weeks following infusion, a subject that received 30 mg/kg of VIS410 developed a primary HSV-1 infection with associated esophagitis and transient leukopenia. Based on serological data, the HSV-1 infection was confirmed to be primary and given that the clinical sequelae of HSV-1 were consistent with the clinical manifestations, the SAE was considered to be unrelated to the study drug by the site investigator. This subject resolved their leukopenia spontaneously and resolved their esophagitis following treatment with valacyclovir administered by their treating physician. No subjects discontinued from the study due to a TEAE, and all TEAEs resolved by the end of the study. Overall, mean clinical laboratory results, vital sign measurements, and ECG values observed after dosing were similar to baseline levels. Mean changes from baseline were also similar across VIS410 dose levels, and no apparent treatment- or dose-related trends were observed.


Pharmacokinetic Results.


Mean AUC0-t, AUC0-∞, and Cmax for VIS410 increased approximately proportional to dose (FIGS. 1A-1B and Table 10). Across the dose cohorts, mean t1/2 values of VIS410 ranged between 250.72 to 376.38 hours and median Tmax values ranged between 1.92 and 3.50 hours. The mean clearance values of VIS410 ranged from 11.41 to 14.07 mL/hr and mean volume of distribution values ranged from 4,914.4 to 6,189.8 mL, across all of the doses tested. A dose proportional increase in the Cmax for VIS410 was observed, which ranged from 58.6 μg/mL at a 2 mg/kg dose to 1316 μg/mL at a 50 mg/kg dose. Additionally, PK samples were examined for the presence of anti-drug antibodies (ADA). Of note, no preexisting ADAs were detected before administration of VIS410. It was found that four subjects (three at 5 mg/ml and one at 15 mg/ml dose levels) developed low titer ADA, 120 days after administration of VIS410. Notably, the presence of ADA did not alter drug PK as exclusion of ADA-positive subject data from the analysis did not substantially affect calculated PK parameters, including half-life and drug exposure.









TABLE 10







Mean (CV) Serum Pharmacokinetic Parameters of VIS410









VIS410












Parameter (unit)
2 mg/kg (N = 6)
5 mg/kg (N = 6)
15 mg/kg (N = 6)
30 mg/kg (N = 6)
50 mg/kg (N = 6)




















AUC0-t
10828
(11)
28026
(50)
90332
(33)
163914
(41)
322070
(16)


(hr · μg/mL)


AUC0-∞
11074
(12)
36086
(25)
100410
(20)
190921
(10)
323451
(16)


(hr · μg/mL)


Cmax (μg/mL)
58.6
(16.8)
180.5
(29.6)
446.1
(13.6)
980.5
(16.7)
1316.0
(14.3)


Tmax (hr)a
3.00
(1.92, 3.00)
3.50
(1.92, 4.00)
3.00
(1.92, 3.08)
2.46
(1.92, 4.00)
1.92
(1.92, 3.00)


t1/2 (hr)b
250. 7
(11.6)
293.1
(38.5)
341.2
(17.1)
288.6
(15.3)
376.4
(16.0)


CL (mL/hr)
14.1
(10.4)
12.6
(28.6)
12.9
(40.8)
11.7
(19.7)
11.4
(6.4)


Vd (mL)
5089
(16)
4914
(21)
6027
(22)
4779
(15)
6190
(17)





Abbreviations: AUC, area under the curve; Cmax, maximal concentration of VIS410; Tmax, time at which maximal concentration is achieved; t1/2, half-life; CL, clearance; Vd, volume of distribution; CV, coefficient of variation; hr, hours; Kel, terminal elimination rate constant.


Note:


Kel-associated pharmacokinetic parameters for Subject 202 (5 mg/kg VIS410) and Subject 306 (15 mg/kg VIS410) were set to missing due to >20% extrapolation of AUC0-∞.



aFor Tmax, the median (minimum, maximum) values are presented.




bVIS410 serum half-life (12.9 days), was calculated by averaging the mean t1/2 of all cohorts.







Nasopharyngeal (NP) swabs were collected for the 15, 30, and 50 mg/kg VIS410 treatment groups but not for the 2 and 5 mg/kg groups. Following a single IV infusion of VIS410 over 120 minutes, across the dose cohorts tested, NP VIS410 concentrations appeared to increase with each increasing dose level, in a similar manner to serum Cmax levels (Table 11 and FIG. 1A-1B). Mean NP VIS410 concentrations reached peak levels within 24 hours after dosing for all the dose cohorts tested and remained measurable throughout the collection period. Mean NP VIS410 concentrations for the ADA-negative subset were comparable to the PK analysis set demonstrating that the ADA status did not influence VIS410 NP concentrations.









TABLE 11







VIS410 Nasopharyngeal Pharmacokinetic Cmax Statistics













Dose

Mean Cmax ± SD



Cohort
(mg/kg)
n
(μg/mL)
















3
15
6
7.6 ± 5.2



4
30
6
20.0 ± 16.3



5
50
6
25.3 ± 10.4







Cmax—Maximum observed nasal concentration.






Modelling of Population-Level Benefits.


Using the measured half-life and biodistribution information as well as information on protective levels in animals, it was modeled if a population-level prophylaxis strategy with VIS410 would be able to reduce influenza burden during a single influenza season. The microsimulation results indicated that prophylaxis of even a small percentage of the population can have a substantial impact on the outcome of the epidemic as measured by the reduction in both attack rates and hospitalization rates for the elderly. Simulations were carried out for a range of temperate-zone influenza epidemic scenarios corresponding to attack rates between 4.8% and 27%. Attack rates and hospitalization rates for the 3 coverage levels explored in this study are shown in Table 12. For a prophylactic dose of 8-fold over a protective threshold of 1-2 mg/kg as estimated from preclinical prophylactic experiments and with administration initiated 0-8 weeks prior to the epidemic peak, median reductions in attack rates from 50 simulations were 8.6% (IQR: 4.7%-11.0%) for 2% coverage, 16.1% (IQR: 8.1%-20.9%) for 4% coverage, and 22.6% (IQR: 12.7%-30.0%) for 6% coverage (FIG. 2A). The associated reductions in hospitalization of the elderly were 8.8% (IQR: 4.9%-11.6%), 16.5% (IQR: 8.8%-21.9%) and 22.9% (IQR: 13.0%-30.6%), respectively, for the three coverage scenarios (FIG. 2C).









TABLE 12







Results of Microsimulation Measurements













Coverage













Metric
Age
Admin
Untreated
2% (IQR)
4% (IQR)
6% (IQR)





Attack Rate
 0-5
all
  9.1 (6.7-12.7)
  8.4 (6.0-11.7)
  7.8 (5.4-10.8)
  7.2 (5.0-10.1)


(%)

elderly

  8.9 (6.4-12.3)
  8.6 (6.2-12.0)
  8.4 (6.0-11.7)



 6-15
all
 15.6 (11.6-21.8)
 14.5 (10.4-20.1)
 13.6 (9.4-18.6)
 12.6 (8.7-17.5)




elderly

 15.3 (11.2-21.1)
 14.9 (10.7-20.7)
 14.7 (10.5-20.2)



16-25
all
 13.1 (9.5-18.3)
 12.1 (8.6-16.8)
 11.3 (7.8-15.5)
 10.4 (7.2-14.6)




elderly

 12.8 (9.2-17.6)
 12.4 (8.9-17.3)
 12.2 (8.6-16.9)



26-34
all
 11.4 (8.2-15.9)
 10.5 (7.4-14.6)
  9.8 (6.7-13.5)
  9.0 (6.2-12.7)




elderly

 11.1 (8.0-15.4)
 10.8 (7.7-15.0)
 10.5 (7.5-14.6)



35-49
all
 18.6 (13.5-25.5)
 17.1 (12.3-23.5)
 16.0 (11.1-21.8)
 14.8 (10.3-20.5)




elderly

 18.1 (13.1-24.7)
 17.5 (12.6-24.2)
 17.2 (12.3-23.6)



50-64
all
 12.3 (9.0-17.1)
 11.4 (8.2-15.7)
 10.7 (7.4-14.5)
  9.8 (6.8-13.6)




elderly

 12.1 (8.7-16.5)
 11.7 (8.4-16.1)
 11.4 (8.1-15.6)



>65
all
  7.2 (5.240.0)
 6.6 (4.7-9.1)
 6.2 (4.3-8.4)
 5.7 (4.0-7.8)




elderly

 6.5 (4.7-8.8)
 5.8 (4.1-7.8)
 5.2 (3.7-6.9)



All
all
 13.1 (9.5-18.2)
 12.1 (8.6-16.7)
 11.3 (7.8-15.4)
 10.4 (7.2-14.5)



Ages
elderly

 12.7 (9.2-17.4)
 12.3 (8.8-17.0)
 11.9 (8.5-16.5)


Hospitalization
 0-5
all
 72.1 (52.1-102.8)
 67.5 (46.0-95.1)
 62.9 (42.9-87.4)
 58.3 (38.3-81.3)


Rate (per

elderly

 70.6 (49.1-98.2)
 70.6 (47.5-96.6)
 67.5 (47.5-95.1)


100K)
 6-15
all
 5.3 (2.6-7.9)
 4.4 (2.6-7.0)
 4.4 (2.6-6.2)
 4.4 (2.6-6.2)




elderly

 5.3 (3.5-7.0)
 4.4 (2.6-7.0)
 4.4 (2.6-7.0)



16-25
all
 27.0 (19.0-37.2)
 24.8 (16.8-35.0)
 22.6 (15.3-32.1)
 21.2 (13.9-29.9)




elderly

 26.3 (18.2-36.5)
 25.5 (17.5-35.7)
 24.8 (16.8-35.0)



26-34
all
 23.5 (16.1-31.6)
 21.3 (14.4-29.3)
 19.5 (13.2-27.6)
 17.8 (12.1-25.3)




elderly

 22.4 (15.5-31.0)
 21.8 (14.9-30.4)
 21.3 (14.4-29.3)



35-49
all
 31.5 (22.3-43.1)
 29.1 (20.3-39.7)
 27.1 (18.5-37.3)
 24.7 (16.9-34.9)




elderly

 31.0 (21.8-42.1)
 30.0 (20.8-41.2)
 29.5 (20.3-40.2)



50-64
all
 55.0 (38.7-74.7)
 51.1 (35.4-69.6)
 47.2 (32.0-64.6)
 43.2 (29.8-60.1)




elderly

 53.3 (37.6-73.0)
 51.7 (36.5-70.7)
 50.5 (35.4-69.1)



>65
all
272.4 (198.3-370.6)
249.2 (178.1-339.5)
230.8 (161.3-315.5)
212.5 (148.6-296.3)




elderly

243.6 (174.1-331.5)
217.7 (154.2-294.7)
194.1 (137.4-262.0)



All
all
 63.5 (46.1-86.7)
 58.5 (41.4-79.4)
 54.2 (37.5-74.0)
 49.7 (34.5-69.1)



Ages
elderly

 59.0 (42.3-80.6)
 55.1 (39.1-75.3)
 51.8 (36.6-69.7)









In addition to investigating coverage levels, it was assessed whether administration of VIS410 prophylaxis to the elderly would be an improvement over general population administration. In the microsimulations, general-population administration results in larger reductions in attack rate than administration to the elderly alone, partially because of the nature of social contacts by which individuals are more likely to associate with those in their same age group. However, prophylaxis of elderly populations was associated with a larger reduction in elderly hospitalizations than distribution to the population at large. The median reductions in >65 years old hospitalizations were 11.4% (IQR: 8.2%-13.3%) for 2% coverage, 21.6% (IQR: 17.4%-24.9%) for 4% coverage, and 30.9% (IQR: 24.8%-35.1%) for 6% coverage when VIS410 was administered to the elderly only. Hospitalization rate in the elderly is an important outcome measure as this age group makes up the majority of influenza hospitalizations and is particularly vulnerable to severe outcomes. Hospitalization rates across all age groups differed by a small amount (±4%) when comparing general-population prophylaxis to prophylaxis of the elderly only (FIG. 2B). The impact of VIS410 prophylaxis on seasonal influenza epidemics shown in FIGS. 2A-2B represents an aggregation across a number of simulation variables (including severity of the epidemic, and date of administration of VIS410 relative to the date of peak activity). When the analysis is restricted to a single epidemic scenario, the effect of VIS410 on the severity of the epidemic is much more pronounced (FIG. 9).


An additional critical parameter that had a large influence on attack rates and hospitalizations was the timing of VIS410 deployment (FIG. 3). For an influenza season of moderate intensity, in the model described herein, administration of VIS410 four to eight weeks prior to peak prevalence resulted in a reduction of hospitalizations of 34.3% (IQR: 31.9%-36.6%) for 6% coverage, but the impact on hospitalizations was more marginal when administered zero to two weeks prior to the peak, with the reduction of hospitalizations at 13.9% (IQR: 12.1%-15.4%). The absolute case reduction of a prophylaxis strategy is very sensitive to the individual protective period assumed for an administered dose of VIS410, which is longer than 40 days in the model (FIG. 4). If prophylaxis is distributed too late, the majority of individuals will have already been infected, but if given too early, the prophylactic effects of VIS410 administration would wane before the major part of the epidemic wave passes through the population. Administration just prior to the peak is not optimal for population prophylaxis. At this period, approximately 30-40% of the season's infections have already occurred, and the opportunity is lost to protect individuals who become infected during the early and slow phase of the epidemic.


Influenza A remains a major public health threat based on seasonal infections and the potential for pandemic infection. Monoclonal antibody therapies like VIS410 that target broadly neutralizing epitopes represent a powerful class of therapies with multiple mechanisms of anti-viral activity, including direct neutralization of either viral attachment or viral fusion, and Fc-mediated activity including complement deposition and recruitment of cells of the innate immune system that enable the destruction of virus-infected cells (Longini et al. Science 2005; 309(5737): 1083-7; Brandenburg et al. PLoS one 2013; 8(12): e80034). Additionally, as demonstrated here, the relatively safe profile of an antibody therapy enables dosing at high levels through bolus administration compared to many small molecule therapies.


VIS410 was initially developed for the treatment of hospitalized patients with influenza A for at least the following reasons. (1) There are no approved treatments for hospitalized influenza patients, representing a large unmet need. (2) Administration of polyclonal antisera has demonstrated the ability to reduce morbidity and mortality in this population (Hung et al. Chest 2013; 144(2): 464-73). (3) The data from VIS410 in several preclinical models have demonstrated the ability to rapidly reduce viral titers by greater than one log10 and reduce ARDS in lethal models of H7N9 (Tharakaraman et al. Proc Natl Acad Sci USA. 2015; 112(35):10890-5). (4) Pre-clinical data in the ferret suggest that VIS410 can also prevent aerosol transmission of influenza (H1N1) despite its short half-life in this animal model (Lakdawala et al. Therapy or prophylaxis with an HA-stem antibody (VIS410) limits respiratory droplet transmission of influenza viruses in the ferret model. Options for the Control of Influenza VIII. Cape Town, South Africa: International Society for Influenza and Other Respiratory Virus Diseases; 2013). (5) As demonstrated in this Phase 1 study, the relatively safe profile of an antibody therapy enables dosing at high levels through bolus administration that can potentially enable a more rapid drop in viral loads compared to many small molecule therapies.


In this study, it was investigated whether VIS410 could be a useful therapy and/or prophylactic countermeasure. To this end, it was demonstrated here that VIS410 is generally safe and well tolerated, even at the relatively high dose levels of 30 mg/kg and 50 mg/kg. The most common AE related to study drug was loose stool or diarrhea (10° F. 40 subjects; 24.4%). Most subjects had minor and transient loose stool that resolved spontaneously. Two of the six subjects at the highest dose of 50 mg/ml had moderate diarrhea with associated nausea and vomiting that resolved within 6 hours. None of the subjects with diarrhea had any clinically significant issues such as hypotension and there were no associated laboratory abnormalities. The time of onset and transient nature of these AEs suggest that they may be related to an infusion reaction and options such as slowing infusion or pretreatment can be explored in future development to further mitigate these AEs.


There is precedence for infusion reaction related GI events as previously observed in both IVIG therapy and other monoclonal antibodies and appears to be related to mast cell activation that correlates to the period around the Cmax phase of the initial infusion. Serum PK was approximately dose proportional, and nasal PK of the target organ (nasopharyngeal/upper respiratory tract) demonstrated a partitioning compared to serum of 1:53. ADA was observed at very low levels in 4/30 subjects treated with VIS410. The presence of ADA in response to treatment with IgG1 monoclonal antibodies such as VIS410 is not unique and has been observed in marketed human monoclonal antibodies such as adalimumab.36 If ADAs were to have a clinically-relevant impact on efficacy, it would be expected to observe a change in the PK of VIS410 as a result of ADA appearance. This was not observed in this study. While this was a small study in healthy volunteers, and ADAs will continue to be monitored thru the development program, these observations would suggest that an impact of ADAs on acute treatment or a one-time prophylaxis of influenza is unlikely. However, it may be likely that the 13% (4 of 30) of subjects who produced ADAs, upon re-exposure to VIS410, would elicit a similar immune response and produce ADAs again. Because the time-course to elicit the ADA response upon re-exposure and clinical significance of this hypothetical concern is unknown, it would be difficult to speculate on the potential impact that re-administration may have for VIS410 therapy either as a treatment or prophylactic modality.


Given the phase 1 trial results, another question was addressed, that is, whether VIS410 could be successfully deployed in the event of an epidemic outbreak to improve public health outcomes. Given VIS410's half-life, it was predicted that its distribution to the primary site of influenza A infection (nasopharynx), and its potency, that limited, directed use of the agent would reduce the total burden of disease. It is noted that universal prophylaxis is unlikely to be practical or feasible. To test the hypothesis of the ability of VIS410 to reduce influenza disease burden, a micro-simulation of seasonal influenza that is in good general agreement with observed attack rates was developed. In multiple scenarios, administration of a broadly neutralizing antibody like VIS410 at an estimated dose of 8-16 mg/kg to the at-risk elderly, for example in nursing homes and within the hospital, prior to an influenza outbreak reduces the frequency of serious influenza. This effect can be achieved even with administration of VIS410 at a relatively low coverage (between 2% and 6%), having a measurable impact on mitigating hospitalization events in an influenza outbreak.


Sensitivity analysis of the models indicates that timing of administration may be a crucial component of the decision-making process for the deployment of VIS410 as a prophylaxis. The analysis suggests that between four to eight weeks prior to an epidemic peak is the optimal timing for deployment, and this is also dependent on the dose given which determines the length of an individual's protective period. As recently developed climate-based models have made wintertime influenza peak forecasting possible with a four to six week lead time, it can in fact be possible to have accurate enough influenza prediction to begin the early roll-out of a prophylaxis (Shaman et al. Nature communications 2013; 4: 2837; Shaman et al. Proceedings of the National Academy of Sciences of the United States of America 2012; 109(50): 20425-30). Another factor may be determining whether an influenza season will be short or long, and the forecasting exercises would need to be re-run with this exact scenario in mind: timed deployment of a population-level prophylactic whose aim is to stem transmission and reduce hospitalizations in the elderly.


Desirable outcomes for influenza public health interventions include reductions in attack rates and hospitalizations across all age groups. For hospitalization reductions in particular, it is usually not possible to prioritize one age group over another, and for this reason there is a long unresolved question in influenza about the age-targeting of public health interventions (e.g., targeting high-contact or high-vulnerability individuals for intervention). Targeting high-contact individuals may have a larger impact on mitigating the epidemic as a whole, including larger attack-rate reductions in high-vulnerability individuals (FIG. 2A). On the other hand, targeting high-vulnerability individuals has a more direct and measurable impact on the individuals that receive prophylaxis (FIG. 2C), and it may make it easier to argue for higher coverage levels if it can be clearly seen that protection is highly efficacious on an individual level.


The general indirect benefits seen in this population modeling exercise are seen in all population-level analyses of public health interventions for infectious disease. Precise outcomes from the population exercise may be affected by, for example, geographic, demographic, and contact structure of the population in question; individual variation in the protective period; interaction between VIS410 therapy and acquisition/loss of influenza-specific immunity; and the sometimes unpredictable shape of influenza epidemics. For long-term effects of VIS410 as a public health strategy and the relationship with immunity, it is noted that VIS410 targets a non-immunodominant epitope. As such, in animals, there is no measurable difference in the strength of the native immunological response between infected, untreated animals, and infected VIS410-treated animals. In both cases, re-challenge with the same virus results in no infection due to a memory response. For short-term effects (single epidemic season), the general prophylaxis principles described in this analysis can be robust to different characteristics of temperate-zone influenza epidemics, and individual cities or states can perform analyses and make decisions that are specific to their populations and their past experience with influenza.


In summary, based on the results presented here, it was found that the safety and pharmacokinetic profile of VIS410 allow for not only treating influenza on an individual level but also as a public health strategy to mitigate the effects of seasonal or pandemic influenza based on its ability to reduce the overall burden of disease when strategically administered in a vulnerable population.


Model Description

An individual-based epidemic microsimulation was developed in C++. The simulation was based on a previously developed model (Boni et al. (2013) Phil Trans R Soc L B 368: 20120207) and is similar in structure and design to an array of microsimulation models that have been developed over the past decade (Ferguson et al. (2005) Nature 437: 209-214; Germann et al. (2006) Proc Natl Acad Sci USA 103: 5935-5940; Longini and Koopman (1982) Biometrics 38: 115-126). The simulation has a 6-hourly time step and asynchronous updating which is implemented with a special scheduler class that keeps track of which individuals need updating at every time step. The population is structured into households, and locations (neighborhoods). Individuals spend 12 hours a day in their household potentially infecting household contacts (nighttime), and 12 hours a day (daytime) in a pre-assigned location potentially infecting others in the general population who are in the same location; general-population daytime contact rates are age-specific (Mossong et al. (2008) PLoS Med 5: e74). Individuals also engage in travel to random locations with a probability of 0.001 per timestep. The model has 1,000 locations and one million individuals.


Individuals in the model can pass through any of eight clinical states: susceptible to infection, exposed to influenza virus, latently infected (i.e. infectious but with mild or no symptoms), infected with symptoms, severe influenza, hospitalized with severe influenza, recovered and immune, and deceased. Individuals can be assigned to one of seven age groups: 0-5, 6-15, 16-25, 26-34, 35-49, 50-64, and >65 years of age.


During the daytime time steps, a location-specific force of infection (FOI) is computed using the population contact structure and the age structure of the infected individuals in that location. The FOI is multiplied by a scaling parameter (β) and a Poisson random number (mean=β·FOI) of new individuals-to-be-infected (challenged by virus) is generated at each location at each daytime time step; each individual is then selected for infection or non-infection according to their immunity and protection by VIS410 (see below). The parameter β is used to calibrate the attack rates and epidemic shape/duration in the model (see below). In order to simulate the introduction of infected individuals from other populations, 10 random susceptible individuals (0.001%) were infected each day.


A seasonal forcing function for β was implemented using the positive part of a cosine function to ensure that simulated epidemics peaked at times consistent with observed epidemics. Although influenza epidemics can peak at any point between December and March in northern temperate countries, it was not necessary to include all of this variation, as the important feature for VIS410 administration will be how close to the epidemic peak the therapy is distributed and used.


Susceptible individuals in the model carry some level of natural partial immunity to influenza which is based on their age and likelihood of past infection/vaccination. Natural or partial immunity is modeled simply on a scale of zero to one that describes an individual's relative immunity to infection if she/he encounters an infected contact. In other words, a completely naïve individual would have an immunity of 0.0 and would meet infectious individuals and contract an influenza infection proportional to some rate β (general scaling parameter for transmission, from above). An individual that is partially immune with an immunity level of 0.67, for example, would be three times (1/(1−0.67)) less likely to become infected under the same pattern of contacts as a completely naïve individual; this individual would only have a 33% chance of becoming infected if he were challenged by virus. The mean immune levels for different age classes are shown in the table below. These are modeled as normal distributions with standard deviations set to 0.05. For the younger age classes a fraction of individuals are assumed to be completely naïve (immunity=0.0); age-specific rates for being considered completely naïve are listed in Table 13. These assumptions are based on an average 15% annual attack rate (Keitel et al. (1997) Vaccine 15: 1114-1122; Edwards et al. (1994) J Infect Dis 169: 68-76; Neuzil et al. (2002) J Infect Dis 185: 147-152) and vaccination rates in the US and Europe which are in the 15% to 50% range depending on country, season, and age group (Report MW (2012) Influenza Vaccination Coverage Among Health-Care Personnel—2011-12 Influenza Season, United States. 61: 2008-2011; Blank et al. (2008) BMC Public Health 8: 272).









TABLE 13







Age-based immunity levels used in the microsimulations









Age
Mean Immunity-
Fraction


Group
Level
Naïve












0-5
0.50
0.3000


 6-15
0.50
0.2019


16-25
0.40
0.0398


26-34
0.40
0.0100


35-49
0.40
0.0100


50-64
0.30
0.0100


>65
0.20
0.0100









For each individual, the level of VIS410 antibody circulating in that person's blood was tracked. A therapeutic dose is set to (1-2 mg/kg) and this blood-concentration decays exponentially with a half-life of 13 days. If an individual is selected for infection in a particular timestep, that individual will be refractory to infection (i.e., protected by VIS410) with probability equal to—





(Protective EfficacyVIS410)*(blood concentration)/(blood therapeutic concentration)


In these simulations, the protective efficacy of VIS410 was set to 0.9, providing a maximum of 90% reduction in the probability of being infected when VIS410 levels are equal to or above the prophylactic dose (1-2 mg/ml). Individuals can receive 4-fold or 8-fold the prophylactic dose at the time of prophylaxis. A concentration below 0.1-fold the prophylactic dose is set to zero in the simulation. This behavior is shown in FIG. 4.


Age-specific hospitalization and mortality (given hospitalization) rates are taken from previous studies (section below). Age and household size distributions were set for the US population (US_Census_Bureau (2014) Age Demographic and Housing Estimates.; Statista.com (n.d.) Distribution of Households in the US by Household Size. Accessed 6 Nov. 2015.).


Strategy Definitions


VIS410 prophylaxis strategies were modeled by considering the fraction of the population that would receive prophylaxis (the coverage f), the time before the epidemic peak at which the treatment is distributed (between 0 to 8 weeks before the peak), and whether the treatment was distributed to individuals of all ages or only individuals over the age of 65. VIS410 deployment for a given coverage level was spread equally over a 14 day period after the desired date of administration. Coverage levels of 2%, 4%, and 6% of the total population size were explored.


Model Validation

Infection Duration


Mean infection duration was set to a 1 day latent and infectious period and 2.75 days (with a standard deviation of 0.75 days) of a symptomatic and infectious period, for non-severe patients based on the known course of influenza infections (Carrat et al. (2008) Am J Epidemiol 167: 775-785; Hien et al. (2010) PLoS Med 7: e1000277). Although some of these studies show viremia out to day 7, these data also show a reduction in symptoms after days 3 or 4, and a difference in whether influenza can be molecularly confirmed or virologically confirmed in the late stages of infection. For individuals progressing to severe influenza, the duration of the severe stage of disease was set to 3.0 days. A fraction (5%) of individuals progressed to severity, thus the mean duration of a non-hospitalized influenza infection was 3.90 in the simulation.


Household Infection Rates


During the nighttime time steps, household infections occur according to previously inferred probabilities of household infection (Philip et al. (1961) Am J Hyg 73: 123-137; Longini et al. (1988) Am J Epidemiol 128: 845-859; Longini and Koopman (1982) Biometrics 38: 115-126; Cowling et al. Ann Intern Med 151: 437-446; Cowling et al. (2010) N Engl J Med 362: 2175-2184; Papenburg et al. (2010) Clin Infect Dis 51: 1033-1041; Petrie et al. (2013) PLoS One 8: e75339; Suess et al. (2012) BMC Infect Dis 12: 26; Klick et al. (2011) Epidemiology 22: 793-796). Note that the results in these studies vary substantially depending on whether the strain was pandemic or seasonal, prior immunity in household contacts, oseltamivir use in the study, and whether and at what time point influenza was molecularly/virologically confirmed. A probability of infection (given an infected contact in a household) of 0.0216 per individual in a six-hour time step was chosen; this corresponds to an average 13% household attack rate for the duration of an infection for families with 4 or 5 members.


Epidemics Duration and Attack Rate


The epidemic duration and attack rate are affected by β (the transmission scaling parameter), migration between locations, immunity levels, and household contact rates. Household infection rates were calibrated separately so that the expected household attack rates were achieved. Between-location movement rates affect the duration of the epidemic. Individuals moved on a daily basis to workplaces, schools, or other daily locations that were pre-determined. This movement rate was set so that 1 in 250 individuals had random travel patterns assigned per day and calibrated so that the size and duration of the epidemic correspond to US influenza epidemic patterns.


Influenza attack rates in the US range from 5% to 25% (Longini et al. (1988) Am J Epidemiol 128: 845-859; Keitel et al. (1997) Vaccine 15: 1114-1122; Edwards et al. (1994) J Infect Dis 169: 68-76; Neuzil et al. (2002) J Infect Dis 185: 147-152; Monto et al. (1985) Am J Epidemiol 121: 811-822). These vary by age group and can be as high as 30% for children, depending on the influenza season. As most serological studies on seasonal influenza do not break individuals out into narrow age bands (with exceptions (Monto et al. (1985) Am J Epidemiol 121: 811-822; Hayward et al. (2014) Lancet Resp Med 2600: 16-19), it is difficult to know much about age-specific attack rates except that (1) children generally have higher attack rates than adults, and (2) elderly groups tend to have lower attack rates, but this needs to evaluated in light of the fact that elderly individuals can have low serological responses to influenza infection.


The duration of an influenza-like illness (ILI) season can be calibrated using CDC's ILINet, which shows weekly ILI incidence, for the past 11 years in 10 different regions in the US. Median duration is 15 weeks, and inter-quartile range is 10-20 weeks. These are epidemics of “influenza-like illness” which is defined syndromically, so the true influenza season may be slightly shorter (this depends on the season, subtype, and other circulating viruses). To compute these ILI durations, region-specific baselines were computed from the ILI trends (bottom two terciles of the data), and ILI rates that were two standard deviations above the baseline were considered as high ILI activity and used to define the ILI season.


15 different epidemic scenarios were defined based on the transmission parameter β (five different values) and on host immunity levels (three different values). The five β values considered are: 0.36, 0.38, 0.40, 0.425, and 0.45. And, three different scenarios of “relative levels of pre-existing immunity”: 1.0 (a baseline or reference value), 0.94, and 1.06, were considered.


The levels of pre-existing immunity differ in the age groups according to Table 14.









TABLE 14







Age-Based Pre-Existing Immunity Levels


Used in the Microsimulation












Ages 0-15
Ages 16-49
Ages 50-64
Ages ≥65















Mean Immunity in
0.470
0.376
0.282
0.188


Scenario 1


Mean Immunity in
0.5
0.4
0.3
0.2


Scenario 2


Mean Immunity in
0.530
0.424
0.318
0.212


Scenario 3









Here, the value represents the reduction in susceptibility to infection if a person comes into sufficient contact with an infectious individual. The percentage naïve does not change for the different scenarios.


The age specific and overall attack rates for the fifteen different transmission scenarios are shown in Table 15.









TABLE 15







Median Attack Rates by Age Groups










Relative
Median Attack Rates (%)













β
immunity
Ages 0-15
Ages 16-49
Ages 50-64
Ages >= 65
All Ages
















0.36
0.94
15.87
17.40
14.49
8.26
15.45



1.00
10.03
11.04
9.25
5.31
9.84



1.06
5.92
6.43
5.46
3.24
5.77


0.38
0.94
17.93
19.79
16.47
9.49
17.56



1.00
11.51
12.63
10.62
6.11
11.24



1.06
7.10
7.72
6.54
3.84
6.91


0.40
0.94
19.83
21.71
18.22
10.57
19.35



1.00
13.23
14.60
12.24
7.12
13.01



1.06
8.12
8.98
7.56
4.41
8.00


0.425
0.94
22.82
25.16
21.17
12.34
22.45



1.00
15.63
17.24
14.50
8.48
15.34



1.06
9.95
10.99
9.35
5.44
9.83


0.45
0.94
25.58
28.37
23.96
14.06
25.31



1.00
18.61
20.44
17.25
10.10
18.22



1.06
11.99
13.26
11.36
6.56
11.88









These fifteen scenarios were chosen (i.e., calibrated) in order to achieve attack rates in the 5% to 25% range and epidemic durations in agreement with CDC data on influenza-like illness incidence in the United States. Typical prevalence rates, attack rates, hospitalization rates, and epidemic durations are shown in FIGS. 5-7.


Age-Specific Hospitalization and Mortality


The model construction requires us to assign the probability of an influenza infection becoming severe, the probability of a severe influenza infection being hospitalized, and the probability of a hospitalized influenza patient dying. The probability of progressing from non-severe symptomatic influenza to severe influenza is poorly defined, as typically severity is measured in hospitalized patients but not in the general patient pool. In the calibrations below, this fraction was set to 5%; in other words, 5% of influenza infections progress from “normal influenza” to “severe, but not necessarily hospitalized or not yet hospitalized, influenza.” From these 5% the hospitalization and mortality rates can be calibrated as these data are collected at national levels in the US and most other countries. Using US data these rates/probabilities were set to values shown Table 16.









TABLE 16







Age-Based Hospitalization and Mortality


Rates Used in the Microsimulation









Age Group
Hospitalization Prob
Mortality Prob












0-5
0.1600
0.010


 6-15
0.0065
0.020


16-25
0.0410
0.025


26-34
0.0400
0.033


35-49
0.0340
0.050


50-64
0.0880
0.066


>65
0.7500
0.066









Here, the hospitalization probability in the second column is the probability of becoming hospitalized if one has a severe influenza infection. The mortality probability is the probability of a hospitalized patient dying as a result of complications resulting from influenza infection.


Hospitalization rates were taken from previous studies of influenza associated hospitalization in the US (Thompson et al. (2004) J Am Med Assoc 292: 1333-1340; Bhat et al. (2005) N Engl J Med: 2559-2567; Dawood et al. (2010) J Pediatr 157: 808-814; Jhung et al. (2011) Clin Infect Dis 52 Suppl 1: S13-S26; Zhou et al. (2012) Clin Infect Dis 54: 1427-1436). Depending on the hospital classifications used and the severity of the influenza season, annual influenza-attributed hospitalization rates in the United States fall between 20 and 120 per 100,000 individuals. For individuals over the age of 65, this rate falls between 200 and 400 per 100,000.


For the fifteen transmission scenarios chosen for the analysis, general-population hospitalization rates fall between 28 and 123 per 100,000. For the over 65 age group, these rates are between 200 and 370 per 100,000 individuals.


Model calibration was performed for influenza A, where possible. A low transmission season in the model can also be considered one that is predominantly influenza B. For a season that is mixed or approximately equally split between influenza A and influenza B, the reductions in attack rate and hospitalization resulting from VIS410 prophylaxis would be lower.


Sensitivity Analysis

To perform a basic sensitivity analysis to various epidemiological scenarios, the transmission scenario (the transmission parameter 3 and the pre-existing population immunity), the coverage level (2%, 4%, 6%), the date of administration of VIS410 prophylaxis (0, 2, 4, 6, and 8 weeks prior to the peak), the dose given (4-fold or 8-fold), and whether VIS410 was administered to all age groups or targeted only to the elderly, were varied. This represents 900 scenarios. Performing 50 stochastic runs for each scenario yields 45,000 simulation outputs.


The Pearson partial correlation coefficients between a simulation output and a simulation input (parameter), holding the other parameters constant, are shown in Table 17.









TABLE 17







Partial Correlation Coefficients
















VIS410


VIS410




Pre-
Administration


administration,




existing
group (0 = all
Dose (4-
Population
days prior to



β
immunity
ages, 1 = elderly)
or 8-fold)
Coverage
epidemic peak
















Attack Rate (all
0.912
−0.967
0.411
−0.089
−0.384
−0.419


ages)








Hospitalization
0.889
−0.955
0.077
−0.115
−0.450
−0.408


Rate (all ages)








Hospitalization
0.859
−0.938
−0.208
−0.134
−0.488
−0.394


Rate (>65 only)















The signs and magnitudes in the first two columns are as expected. Increasing the transmission parameter or pre-existing population immunity has substantial effects on overall attack rates and hospitalizations.


As the administration parameter was “increased” from 0 (all ages) to 1 (elderly only), a clear positive effect was seen on the attack rate, and a small positive effect on the total hospitalization numbers. The reason the hospitalization effect is small is that the increase in attack rate comes in the least likely groups to be hospitalized, children and young adults. As the administration parameter is “increased” from 0 to 1, hospitalizations in the elderly decline (PCC=−0.208) as expected as this group benefits from direct protection due to VIS410 prophylaxis. This effect is seen in the three panels of FIGS. 2A-2C.


As expected, increasing the dose or increasing coverage reduces attack rates and hospitalizations (fourth and fifth columns).


The final column shows the partial correlation between administration time and the attack rate/hospitalization outcomes. However, FIG. 3 already shows that this relationship is non-monotonic. Hence, the negative correlations should not be taken to mean that earlier administration will always be associated with a reduction in attack rate or hospitalizations.


Epitope Analysis of Currently Circulating Strains of Influenza A

To assess the diversity of the amino acids comprising the predicted epitope of hemagglutinin (HA) targeted by VIS410, all HA sequences from H1N1 and H3N2 collected from Jan. 1, 2012 through Jun. 30, 2015 were analyzed. Sequences were obtained from the EpiFlu database provided by the Global Initiative on Sharing Avian Influenza Data (GISAID). 5,782 H1N1 sequences and 10,210 H3N2 sequences were obtained. A multiple sequence alignment (MSA) for both H1N1 and H3N2 was created using HMMalign version 3.1b1 against a curated sequence profile of HA. Using this MSA, the amino acid variation at positions predicted to be in the epitope were computed. Sequence variation at these positions is shown in the sequence logo plots shown in FIG. 8, illustrating that the 25 positions comprising the predicted epitope are highly conserved.


Detailed Phase I Study Design

VIS410 was administered as an IV infusion over at least 120 minutes. Randomization was used to reduce selection bias, double-blinding was employed to reduce potential bias during data collection and evaluation of safety variables, and the placebo group served as the control group. The dose-escalation design and the safety evaluation period within cohorts and between successive cohorts allowed for incremental safety review. In this study, subjects received the following doses: 2, 5, 15, 30, and 50 mg/kg. These doses were consistent with serum concentrations of VIS410 that were achieved during in vivo studies and demonstrated protection from challenge with various influenza A strains and prevention of influenza spreading via respiratory droplets. See Table 18 for the schedule of events.


Institutional research board approval was obtained prior to start of the study. Informed consent and Health Insurance Portability and Accountability Act (HIPAA) authorization were obtained at the screening visit. The screening visit was conducted within 30 days of study product infusion (Days −30 to −1).


Inclusion Criteria


For inclusion in the study, each subject was required to meet all of the following criteria at screening:

  • 1. After the nature of the study was fully explained, the subject read, understood, and signed the IRB-approved ICF which provided written informed consent and HIPAA authorization.
  • 2. Aged ≥18 and ≤55 years.
  • 3. Had a body mass index ≥18 and ≤33 kg/m2, inclusive, and weighed ≤90 kg for subjects in Cohort 5 receiving a 50-mg/kg dose.
  • 4. Was in general good health without history of any of the conditions listed in the exclusion criteria.
  • 5. Nonsmoker for at least 6 months.
  • 6. A woman agreed not to become pregnant from the time of study enrollment until at least 3 months after the completion of the monoclonal antibody infusion. If a woman was sexually active and had no history of hysterectomy or tubal ligation, she agreed to use hormonal birth control, barrier method of birth control with spermicidal gel, or an intrauterine device and continued using approved contraception for at least 3 months after the completion of the VIS410 infusion. These birth control measures were not applicable for postmenopausal women, defined as either having amenorrhea for ≥12 months or follicle-stimulating hormone >40 MIU/mL.
  • 7. Sexually active male subjects agreed to use a barrier method of birth control with spermicidal gel during the course of the study and continued using a barrier method of birth control for at least 3 months after the completion of the VIS410 infusion.
  • 8. Subject had screening laboratory values that met the following criteria:
    • a. White blood cells: 3000 to 12 000/mm3
    • b. Platelets: >150 000/mm3
    • c. Hemoglobin: >13 g/dL (male) or >12 g/dL (female)
    • d. Creatinine: ≤1.40 mg/dL
    • e. Blood urea nitrogen: ≤25 mg/dL
    • f. Aspartate aminotransferase: ≤50 IU/L
    • g. Alanine aminotransferase: ≤67 IU/L
    • h. Alkaline phosphatase: ≤150 IU/L
    • i. Bilirubin: ≤1.4 mg/dL
    • j. Glucose (fasting): <115 mg/dL
    • k. Drug and alcohol screen: Negative


Exclusion Criteria


Any of the following was regarded as a criterion for exclusion of a subject from the study:

  • 1. Previously received an antibody or biologic therapy, whether licensed or investigational (e.g., immunoglobulin products, monoclonal antibodies, or antibody fragments).
  • 2. History of any of the following illnesses or conditions: cancer, heart disease, diabetes mellitus, respiratory condition (such as asthma requiring daily medication), autoimmune disorder, blood dyscrasias, or psychiatric disorder that precluded compliance with protocol.
  • 3. Any chronic condition that required daily prescription or over-the-counter medicine, except for vitamins and birth control products.
  • 4. Abused drugs or alcohol within the previous 12 months.
  • 5. History of a previous severe allergic reaction with generalized urticaria, angioedema, or anaphylaxis.
  • 6. Physical finding on an examination considered clinically significant, such as murmur (other than functional), hepatosplenomegaly, lymphadenopathy, or focal neurological deficit.
  • 7. Blood pressure >160/100 or <90/50 on 2 separate readings.
  • 8. Urinalysis results determined to be clinically significant per investigator discretion.
  • 9. Positive serology for human immunodeficiency virus antibody, hepatitis C virus antibody, or hepatitis B surface antigen.
  • 10. Positive urine pregnancy test during screening or within 24 hours of monoclonal antibody administration or an unwillingness to undergo pregnancy testing for female subjects.
  • 11. Positive drug or alcohol testing at screening or within 24 hours of monoclonal antibody administration.
  • 12. Breastfeeding.
  • 13. Received another investigational study agent within 30 days or 5 half-lives, whichever was longer, before administration of the study product.
  • 14. Received any live virus or bacterial vaccinations within 3 months prior to screening or was expected to receive any live virus or bacterial vaccinations during the study.
  • 15. Received inactivated influenza vaccines within 2 weeks of Day 0 or was expected to receive an inactivated influenza virus during the study.
  • 16. Any other condition that, in the opinion of the investigator, would have jeopardized the safety or rights of the subject participating in the study, or made it unlikely the subject could have completed the protocol.









TABLE 18







Schedule of Events









Procedure
Screening
Study Time After Infusion (days)

















Time Point (Day)
−30 to −1
0a
1
2
3
7
14
28
56
120


Study Visit
1
2
3
4
5
6
7
8
9
10





Informed consent/HIPAA
X











authorization












Inclusion/exclusion criteria
X











Demographics and medical history
X











Serum for hepatitis panel and HIV
X











Drug and alcohol toxicology screenb
X
X










Pregnancy test
X
X










Vital signsc
X
X
X
X
X
X
X
X
X



Electrocardiogramd
X
X










Physical examinatione
X
X
X

X

X

X



Clinic admissionf

X










PK samplingg

X
X
X
X
X
X
X
X
X


ADA samplingh

X




X

X
X


Study infusion

X










Hematologyi
X
X
X

X
X
X
X




Serum chemistry/liver function testsi
X
X
X

X
X
X
X




Urinalysisi
X
X
X

X
X
X
X




Concomitant medications
X
X
X
X
X
X
X
X
X
X


Adverse eventsj

X
X
X
X
X
X
X
X
X


Nasopharyngeal swabsk

X
X

X
X





Abbreviations:


ADA, antidrug antibodies;


HIPAA, Health Insurance Portability and Accountability Act;


HIV, human immunodeficiency virus;


PK, pharmacokinetic.



aThe first 2 subjects of each cohort may have been admitted at Day −1 to facilitate pre-infusion procedures, although these subjects received study drug or placebo on Day 0. The pre-infusion activities for these subjects may have occurred on Day −1 or Day 0.




bDrug and alcohol toxicology testing was performed within 24 hours of starting monoclonal antibody infusion.




cVital sign measurements were recorded at screening and on Day 0 at the start of study infusion (±5 minutes), during the infusion at 15-minute intervals for 60 minutes, then every 30 minutes until the end of infusion. After the end of infusion, vital signs were obtained at 30, 45, 60, and 90 minutes, and 2, 3, 6, 8, 14, and 20 hours until the subject was discharged from the clinic.




dOn Day 0, there were 2 triplicate 12-lead electrocardiograms performed: one before infusion and one at the end of infusion (±10 minutes).




ePhysical examinations were performed at the screening visit, on Day 0 before the infusion, upon discharge from the clinic on Day 1 after the infusion, and on Days 3, 14 ± 1, and 56 ± 7 after the infusion, for applicable subjects. Targeted physical examinations may have been performed at other study visits as needed for adverse event assessment.




fEach subject was admitted to the clinic for his/her designated monoclonal antibody infusion. Discharge from the clinic unit did not occur before the 24-hour post-infusion study procedures were performed.




gOn the day of study infusion (Day 0, Visit 2), serum samples for PK analysis were collected before the start of infusion; at the end of the infusion; and at 60 minutes, and 2, 8, and 24 hours after the end of infusion. Subjects had PK samples drawn on Days 2, 3, 7, 14 ± 1,28 ± 3, 56 ± 7 and 120 ± 7 after the infusion.



hSubjects had serum samples collected for ADA analysis before the infusion and on Days 14 ± 1, 56 ± 7, and 120 ± 7 after the infusion.



iOn the day of study infusion (Day 0, Visit 2), blood samples for hematology and serum chemistry/liver function tests, and urinalysis samples were collected before the start of the study infusion.




jOn the day of study infusion (Day 0, Visit 2), adverse events were assessed from the start of the infusion until discharge from the clinic at scheduled time points per the protocol, and as needed.




kNasopharyngeal swabs (one from each nostril) were collected from subjects in the 15, 30, and 50 mg/kg cohorts on Day 0 before the start of infusion, at 8 and 24 hours after the end of infusion, and on Days 3 and 7 after the infusion.







Example 2: Preclinical Animal Data Supporting Prophylactic Dose Level
Methodology

An A/Puerto Rico/8/1934(H1N1) lethal mouse models was employed. Animals were administered antibody IP in a volume of 200 μL as prophylaxis one day prior to infection. Then, mice were anaesthetized under isoflurane and challenged i.n. with 50 pL viral suspension (˜100 pfu). Weight and appearance of the animals were recorded daily. Animals were euthanized upon loss of considerable weight (>20%) in conjunction with high body score indicating illness. Lungs were harvested from a subgroup of animals on day four post-infection for the determination of viral load by plaque assay. In addition, lungs on day eight were submitted for histological examination.


Results

The study was completed as follows (Table 19).









TABLE 19







Experimental Design











Agent
Dose (mg/kg)
Administration







PBS (Vehicle)





Ribavirin
75 (3
−24 hours, +24 hours, +48




doses)
hours



VIS410
10
24 h prior to infection



VIS410
2.5
24 h prior to infection



VIS410
0.6
24 h prior to infection










Visual Cues

Animals were monitored for signs of illness (ruffled fur, hunching) daily. Untreated mice that were challenged with H1N1 appeared sick three days post-infection and were euthanized on day seven, as expected. Mice that were challenged with H1N1 and treated with three doses of ribavirin exhibited negligible signs of illness and recovered fully (FIGS. 10A-10B). Mice that were treated with VIS410 one day prior to challenge at 2.5 mg/kg or 10 mg/kg exhibited no sign of illness. Mice that were treated with VIS410 at 0.6 mg/kg one day prior exhibited some signs of illness, with 60% of animals surviving.


Viral Load

The lung viral loads four days after H1N1 infection were assessed in a single plaque assay (Table 20). Comparisons were made between treatment groups to assess the significance of the reductions in lung viral load. Significance (p<0.05) was determined Mann Whitney U test. The lung viral load in all treatment arms was significantly different from that in the untreated group.









TABLE 20







Lung Viral Load in Mice Four Days


after Challenge with H1N1 PR8











Group
Dose (mg/kg)
Lung Viral Load















Untreated

6.03



Ribavirin
75(×3)
4.38



VIS410
10
4.45



VIS410
2.5
4.08



VIS410
0.6
5.38










Example 3: Evaluation of Efficacy and Emergence of Resistance to an Anti-HA Antibody Molecule in a Human Challenge Model of Infection with a p2009 H1N1 Virus

In this study, the efficacy and emergence of resistance to an exemplary anti-HA antibody molecule (i.e., VIS410) was evaluated.


Methods

The efficacy and emergence of viral resistance to VIS410 were evaluated in a Phase 2a human challenge study in healthy volunteers with an H1N1 strain isolated during the 2009 pandemic (p2009 HINI). Eighteen subjects received a single intravenous 2300 mg dose of VIS410 24 hours after viral inoculation. Influenza virus replication from nasopharyngeal swab specimens was measured by tissue culture infectious dose 50 (TCID50) assay and quantitative PCR methods. Emergence of resistance was assessed by using both phenotypic and genotypic approaches to characterize influenza viruses in nasopharyngeal swab specimens from subjects. Briefly, phenotypic resistance was assessed by culturing virus in the presence or absence of pre-defined concentrations of VIS410 and detecting virus outgrowth by nucleoprotein ELISA and viral foci immunostaining. Genotypic resistance was assessed by performing nested Sanger population sequencing of the full-length HA gene and next generation sequencing to detect potential minority species.


Results

VIS410 demonstrated potent antiviral activity at 2300 mg with a 91% and 76% reduction in median viral load AUC compared to placebo for TCID50 and qPCR, respectively. There was no emergence of resistance following administration of a single 2300 mg dose of VIS410. Influenza viruses cultured from nasopharyngeal swab specimens were uniformly sensitive to VIS410 neutralization in vitro. Additionally, Sanger and next generation sequencing did not reveal any deleterious adaptations that would affect VIS410 binding/function.


Thus, a single dose IV administration of VIS410 at 2300 mg provided potent antiviral activity and no viral resistance exhibited in this study.


Example 4: Pharmacokinetics of an Anti-HA Antibody Molecule in a Human Challenge Model of Infection with p2009 H1N1 Virus

In this study, the pharmacokinetics of an exemplary anti-HA antibody molecule (i.e., VIS410) was evaluated.


Methods

Serum and nasal pharmacokinetics (PK) of VIS410 have been characterized in a Phase 2a human challenge study in healthy volunteers, with an HINI strain isolated during the 2009 pandemic (p2009 H1N1). This randomized, placebo-controlled, double-blind study evaluated the PK of a single 2 h IV infusion of VIS410 (2300 mg) administered 24 h after inoculation. Blood samples for PK analysis and for assessment of antidrug antibodies (ADA) to VIS410 were collected before and up to 84 days post infusion in all VIS410 subjects (n=18). Nasopharyngeal samples were collected for nasal PK up to Day 10 to assess VIS410 concentration at the site of infection. VIS410 concentrations were analyzed using a validated ELISA method. Standard non-compartmental methods were used to estimate PK parameters in serum and nasal mucosa.


Study Design

This study was a randomized, double-blind, placebo controlled, Phase 2a human challenge study. Data presented are from the interim analysis of the 31 healthy volunteers. Subjects were inoculated intranasally (Day 1) with approximately 106 tissue culture infective dose (TCID50) of influenza A (H1N1) strain isolated during the 2009 pandemic. One day (24 hours) after inoculation (Day 2), subjects received a single intravenous administration of 2300 mg of VIS410 (n=18) or placebo (0.9% sodium chloride) (n=13).


Serial blood samples for determination of VIS410 serum concentration were collected. Serum VIS410 concentrations were determined using a validated ELISA with a lower limit of quantification of 50 ng/mL. Serial nasopharyngeal swabs for determination of VIS410 nasal concentration and viral shedding were collected. Nasal mucosa VIS410 concentrations were determined using a validated ELISA with a lower limit of quantification of 0.50 ng/mL.


Nasal and serum PK parameters were determined using standard non-compartmental methods using Phoenix WinNonlin software. Serum PK samples were collected pre-dose and at serial times relative to the end of infusion. Nasal PK and viral samples were collected Day −2 (prior to inoculation), pre-dose and at serial times relative to the end of infusion. Non-parametric Mann Whitney U test was used to assess the difference between treatment groups in the area under the viral load time curve (AUC) for H1N1 based on qRT-PCR and TCID50 from nasopharyngeal swabs.


Results

All 18 VIS410 subjects were included in the PK analysis. Of the 31 randomized and treated subjects, 20 (7 placebo and 13 VIS410) were included in the analysis of viral shedding. Seven subjects (4 placebo and 3 VIS410) were excluded due to baseline HAI titer of >10 to the challenge virus and 4 subjects (2 placebo and 2 VIS410) were excluded for not being infected post inoculation. Mean serum and nasal PK parameters are presented in Table 21. The mean serum and nasal concentration versus time profiles are presented in FIGS. 11A-11B, respectively. Statistically significant reduction in viral AUC and peak viral load was observed with a single dose of VIS410 2300 mg vs. placebo (Table 22). All data are presented as mean (CV %).









TABLE 21







Mean Serum and Nasopharyngeal VIS410 PK Parameters




















Cmax
Tmax
AUC0-last
Tlast
AUC0-∞
AUC%extrap
Vd
CL

















Treatment
(μg/mL)
(day)
(day*μg/mL)
(day)
(day*μg/mL)
(%)
(mL)
(mL/day)




















Serum
VIS410
N
18
18
18
18
18
18
18
18



2300
Mean
792
0.260
6,900
55.2
7,150
3.49
5530
334



mg
CV %
32.3
94.1
19.5
5.87
19.4
42.9
25.8
20.0


Nasal
VIS410
N
18
18
18
18







2300
Mean
28.4
3.73
81.6
8.04







mg
CV %
112
72.8
92.2
0.507
















TABLE 22







Interim Analysis of Antiviral Effect of VIS410


2300 mg Compared to Placebo (mITT population)












Placebo
VIS410




Viral Measure
(N = 7)
(N = 13)
Reduction
p Value














Median Viral AUC
552
47.1
91%
0.019


TCID50 (log10X hours)


Median Viral AUC
1033
232
76%
0.024


qPCR (log10X hours)


Median Peak Viral Load
5.00
2.75
2.3
0.009


TCID50 (log10)


Median Peak Viral Load
7.14
5.61
1.5
0.043


qPCR (log10)









Based on preliminary data following a 2300 mg dose (n=18) the serum Cmax was 792 (32%) μg/mL, AUC0-last 7150 (19%) μg*d/mL, clearance 334 (20%) mL/d, and a long half-life of approximately 12 days. Nasopharyngeal concentrations of VIS410 exceeded the in vitro EC50 (0.3-11 μg/mL) of the majority of influenza strains tested within 6 hours of dosing (mean [CV %] concentration after 6 h was 5.6 [140%] μg/mL; Cmax for nasopharyngeal concentrations was 28.4 [112%] μg/mL and remained elevated through Day 8 [9.7 (120%) μg/mL]). VIS410 also demonstrated potent antiviral activity at 2300 mg with a 2.3 and 1.5 log10 reduction in median peak viral load compared to placebo for TCID50 and PCR, respectively. None of the subjects were tested positive for ADA.


Serum PK in this study is consistent with those of a human IgG1 antibody. The observed half-life of approximately 12 days supports a single dose administration of VIS410 in patients with influenza A infection. Nasopharyngeal concentration versus time profiles were highly variable and a clear elimination phase was not evident in the majority of profiles. A single VIS410 dose of 2300 mg resulted in a statistically significant reduction in both viral AUC and peak viral load compared to placebo. Thus, a single dose IV administration of VIS410 at 2300 mg provides potent antiviral activity, which is consistent with the observed high and sustained systemic and nasopharyngeal exposures in relation to the in vitro EC50.


Example 5: Safety and Efficacy of the Monoclonal Antibody VIS410 in a Human Volunteer Challenge Model of Infection with an H1N1 Influenza A Virus
Methods

The efficacy of VIS410 was tested in a Phase 2a human challenge study with an H1N1 strain isolated during the 2009 pandemic. This randomized, placebo-controlled, double-blind study was designed to assess the efficacy and safety of VIS410 in healthy human volunteers challenged with influenza A. Twenty-four hours after viral inoculation, subjects were randomized to receive either VIS410 or placebo and monitored for viral shedding by nasopharyngeal swabs, clinical symptoms and pharmacokinetics. A total of 31 subjects were randomized, all of whom either received VIS410 as an intravenous infusion at a dose of 2300 mg or placebo.


Study Design

This study was a randomized, double-blind, placebo controlled, Phase 2a human challenge study. The primary objectives were to assess the safety and tolerability of VIS410 and the effect of VIS410 on the area under the curve of viral shedding over time (viral AUC).


Healthy adult volunteers (N=31) were inoculated intranasally (Day 1) with approximately 106 tissue culture infective dose (TCID) of influenza A (H1N1) strain isolated during the 2009 pandemic. One day (24 hours) after inoculation (Day 2), subjects received a single intravenous administration of 2300 mg of VIS410 (n=18) or placebo (0.9% sodium chloride) (n=13). Nasopharyngeal swabs for determination of viral shedding were collected on Day −1 (prior to inoculation), pre-dose and at 0, 6, 12, 24, 30, 36, 48, 54, 60, 72, 78, 84, 96, 102, 108, 120, 126, 132, 144, 150, 156, 168, 174, 180, 192, 198, and 204 hours relative to the end of infusion.


The quantity of influenza virus from nasopharyngeal swab specimens was measured by tissue culture infectious dose 50 (TCID50) assay and quantitative RT-PCR (qRT-PCR) methods. A symptom score card was used to record the incidence, severity and duration of signs and symptoms of influenza-like illness through Day 10.


The modified intent-to-treat (mITT) population used in the PD analysis was defined as all randomized subjects who received study drug who met the inclusion criterion of seronegativity by hemagglutinin inhibition assay (HAI) (≤10) on Day 1 and were infected, defined by either seroconversion (≥4×HAI titers from baseline) or measurable viral load (2 consecutive qRT-PCR time points above the level of quantification). Standard non-compartmental approaches using Phoenix WinNonlin (Pharsight Corporation, Princeton, N.J., USA; Version 6.3) were used to calculate peak viral load (VL) and VL AUC. Non-parametric Mann Whitney U test was used to assess the difference between treatment groups in the area under the viral load time curve (AUC) for H1N1 based on qRT-PCR and TCID50 from nasopharyngeal swabs


Results

All 31 subjects received study drug and were included in the safety analysis. Of the 31 randomized and treated subjects, 20 (7 placebo and 13 VIS410) were included in the mITT PD analysis of viral shedding. Seven subjects (4 placebo and 3 VIS410) were excluded due to baseline HAI titer of >10 to the challenge virus and 4 subjects (2 placebo and 2 VIS410) were excluded for not being infected post inoculation.


A robust H1N1 infection model was achieved with median peak viral load of >4.5 log 10 by TCID50 and >7 log 10 by qPCR. VIS410 was generally safe and well tolerated with adverse events reported in 76.9% and 94.4% of the subjects in the placebo and VIS410 treatment arm, respectively.


There were no drug-related discontinuations, serious adverse events, or deaths in the study. Gastrointestinal disorders were the most commonly reported events in the VIS410 arm (88.9%) vs. placebo (15.4%). Abdominal pain and loose stool were the most commonly reported GI events (61.1% and 50%, respectively in the VIS410 arm). Use of a pretreatment regimen containing a histamine blocker (diphenhydramine 50 mg PO) reduced the severity of the GI events with majority (54.5%) being mild


Statistically significant reduction in viral AUC and peak viral load was observed with a single dose of VIS410 2300 mg vs. placebo (Table 22)


The median duration of viral shedding measured by qRT-PCR was 5.29 days (mean=4.92 days) for VIS410 2300 mg and 7.78 days (mean=6.52 days) for placebo, while the median time to resolution of viral shedding was 5.21 days (mean=5.23 days) and 8.24 (mean=7.37 days) for VIS410 2300 mg and placebo, respectively (FIG. 12A). The TCID50 versus time profiles of VIS410 compared to Placebo as measured by a cell based assay are shown in FIG. 12B.


Upper respiratory symptoms resolved a median of 2 days faster in the VIS410 treatment group versus placebo (FIG. 13).


VIS410 was generally safe and well tolerated with a pre-treatment regimen that included over-the-counter oral anti-histamines and NSAIDs. There were no drug-related discontinuations, serious adverse events, or deaths reported in this study. The overall area-under-the-curve (AUC) of viral shedding for the VIS410 treated subjects was 91% (p=0.019) lower than the placebo group, as measured by the cell based assay TCID50, and 76% (p=0.024) lower than the placebo group, as measured by viral RNA quantitation (qPCR). Peak viral levels for the VIS410 treatment groups were 2.2 logs (p=0.009) lower than placebo, as measured by the cell based assay TCID50, and 1.5 logs (p=0.043) lower, as measured by qPCR. Furthermore, subject-reported upper respiratory symptoms resolved a median of 2 days faster in the VIS410 treatment group versus placebo.


Phenotypic resistance was tested using ViroSpot™ assay and based on IC50 values. No phenotypic variants were identified by ViroSpot™ assay (27 samples assessed). Phenotypic assessment of IC50 revealed similar median of Placebo vs. VIS410 (25 samples). The results are shown in FIGS. 14A-14B. No resistant variants were identified.


The study achieved its primary endpoint of reducing the viral shedding area-under-the-curve in the VIS410 treatment group and showed a trend towards a shorter duration and lesser magnitude of upper-respiratory symptoms. VIS410 was generally safe and well tolerated with a pre-treatment regimen that included over-the-counter oral anti-histamines. A single VIS410 dose of 2300 mg resulted in a statistically significant reduction in both viral AUC and peak viral load compared to placebo. VIS410 showed a trend towards reduction in duration of viral shedding and the duration and severity of upper respiratory symptoms compared to placebo.


Example 6: Evaluation of Antibody Dependent Enhancement (ADE) in Preclinical Models of Influenza A Virus Infection Treated with an Anti-HA Antibody Molecule

Antibody dependent enhancement (ADE) is the phenomena where non-neutralizing concentrations of antibodies can bind to virus particles and enhance disease by mediating entry into Fc-bearing cells, such as macrophages, consequently increasing virus tropism and pathogenesis. Data describing ADE during influenza infection in vivo is limited to preclinical and retrospective clinical vaccine studies, where polyclonal immune responses were found to elicit increased inflammation and pathology during heterologous influenza virus infection. With the advent of broadly neutralizing anti-viral monoclonal antibody therapies there is an increased need to understand the proposed therapeutic mechanisms and unintentional immunologic and virologic impact of these antibodies on disease progression. In this study, ADE potential of an exemplary anti-HA antibody molecule (i.e., VIS410) was evaluated in an in vivo efficacy model.


Methods

VIS410 was evaluated in 6-8 week old female CD-1 mice challenged with a lethal dose 25 (LD25) of mouse-adapted A/Puerto Rico/8/34 (H1N1) or A/Victoria/3/75 (H3N2). Four hours after virus inoculation, doses of VIS410 (0.02, 0.2, 2, 20 mg/kg) and irrelevant human IgG1 antibody (0.02, 20 mg/kg) were administered intravenously. This dose range represented both protective and sub-neutralizing levels of VIS410, which could be compared for efficacy or enhancement to the corresponding doses of control antibody. Groups of mice were either harvested at the peak of infection or monitored for 14 days for weight-loss, clinical score and survival, all mice were evaluated for lung viral load and pathology.


Study Design

The study design is illustrated in FIG. 15.


Results

VIS410 treatment in mice demonstrated a dose dependent protection from weight-loss, clinical signs, and mortality during infection with H1N1 and H3N2 influenza A viruses. As shown in FIGS. 16A-16B, VIS410 protected CD-1 mice from influenza A virus-induced morbidity in a dose dependent manner as compared to irrelevant human IgG1. FIG. 14 shows the average lung viral load on Days 1 and 14. As shown in FIG. 17, lung viral loads were equivalent between 0.02 mg/kg VIS410 and placebo treated animals on Day 1 post infection (pi) (H1N1 6.1±0.4 vs. 6.3±0.6 TCID50/g; H3N2 3.9±1.8 vs. 5.1±0.8 TCID50/g, respectively), with all animals that survived to Day 14 pi successfully resolving infection.


Immunohistochemistry and pathology also correlated with dose, with animals receiving the higher doses of VIS410 displaying less viral antigen staining and decreased inflammation while animals treated with 0.02 mg/kg VIS410 or placebo had the greatest viral antigen staining at Day 1 pi and highest pathology scores at Day 14 pi.









TABLE 23





Day 1 IHC and Day 14 pi Lung Pathology in H1N1 Influenza Infected CD-1 Mice Treated with


Different Doses of VIS410 and Irrelevant Human IgG1


















VIS410
Placebo













Immunohistochemistry
20 mg/kg
2 mg/kg
0.2 mg/kg
0.02 mg/kg
20 mg/kg
0.02 mg/kg





Trachea/Primary
NP

+/−
++
NP



Bronchus (IHC)
NP

+
NP

NP




NP
NP
NP
+
++



+/−
NP
+


+/−



+/−
NP

+/−
+
++


Bronchioles (IHC)



+/−

++














+/−


++















+/−
+











IHC Immunohistochemistry for influenza A Virus Nucleoprotein (NP) Demonstrating Virus Replication


NP Not Present, Therefor Not Evaluated


IHC Score:


− All negative


+/− 1-Few Nuclei Stain Faintly Positive for IAV-NP or “Suspect Positive”


+ Few Nuclei Stain Positive for IAV-NP


++ Several Nuclei Stain Positive for IAV-NP


+++ Many Nuclei Stain Positive for IAV-NP













VIS410
Placebo













Pathology
20 mg/kg
2 mg/kg
0.2 mg/kg
0.02 mg/kg
20 mg/kg
0.02 mg/kg





Extend of Alveolitis
0.07 ± 0.26b
0.67 ± 0.49a
1.36 ± 0.84
1.92 ± 1.04
1.50 ± 0.76
1.80 ± 0.92


(Score 0-3)








Severity of Alveolitis
0.13 ± 0.52b
1.13 ± 0.99
2.07 ± 1.14
2.00 ± 0.89
2.21 ± 0.89
2.00 ± 0.67


(Score 0-3)








Alveolar Edema
  0 ± 0a
  0 ± 0
57.1 ± 51.4
61.5 ± 50.6
50.0 ± 51.9
50.0 ± 52.7


(% Positive Slides)








Alveolar Hemorrhage
  0 ± 0
  0 ± 0
  0 ± 0
 7.7 ± 27.7
 7.1 ± 26.7
  0 ± 0


(% Positive Slides)








Presence of Type II
 6.7 ± 258b
66.7 ± 48.8
78.6 ± 42.6
84.6 ± 37.6
85.7 ± 36.3
80.0 ± 42.2


Pneumocyte








Hyperplasia








(% Positive Slides)








Severity of Bronchiolitis
0.07 ± 0.26a
0.47 ± 0.52
0.78 ± 0.58
0.77 ± 0.44
0.71 ± 0.47
0.70 ± 0.48


(Score 0-3)








Extent of Lymphocytic
0.53 ± 0.64
0.33 ± 0.62
1.00 ± 0.55
0.92 ± 0.86
0.93 ± 0.73
1.00 ± 0.67


Cuffing








(Score 0-3)








Severity of Tracheitis/
  0 ± 0
  0 ± 0
  0 ± 0
  0 ± 0
  0 ± 0
  0 ± 0


Bronchitis








(Score 0-3)










Data Represent Mean ± Std Dev



aP < 0.05 vs. Irrelvant Human IgG1 Groups One-Way ANOVA (Kruskal-Wallis test) and Dunn's Post Hoc Test




bP < 0.005 vs. Irrelvant Human IgG1 Groups

















TABLE 24





Day 1 IHC and Day 14 pi Lung Pathology in H3N2 Influenza Infected CD-1 Mice Treated with


Different Doses of VIS410 and Irrelevant Human IgG1


















VIS410
Placebo













Immunohistochemistry
20 mg/kg
2 mg/kg
0.2 mg/kg
0.02 mg/kg
20 mg/kg
0.02 mg/kg





Trachea/Primary
+



++



Bronchus (IHC)
NP
NP
++

++
++




NP
+
++
NP
++




++
++

+








++
++


Bronchioles (IHC)














+





































IHC Immunohistochemistry for influenza A Virus Nucleoprotein (NP) Demonstrating Virus Replication


NP Not Present, Therefor Not Evaluated


IHC Score:


− All negative


+/− 1-Few Nuclei Stain Faintly Positive for IAV-NP or “Suspect Positive”


+ Few Nuclei Stain Positive for IAV-NP


++ Several Nuclei Stain Positive for IAV-NP


+++ Many Nuclei Stain Positive for IAV-NP













VIS410
Placebo













Pathology
20 mg/kg
2 mg/kg
0.2 mg/kg
0.02 mg/kg
20 mg/kg
0.02 mg/kg





Extend of Alveolitis
0.14 ± 0.36b
0.60 ± 0.63
1.07 ± 0.59
1.71 ± 0.91
1.29 ± 0.61
1.57 ± 0.94


(Score 0-3)








Severity of Alveolitis
0.14 ± 0.36b
0.73 ± 0.80
1.60 ± 0.98
2.14 ± 1.03
1.64 ± 0.74
1.64 ± 0.93


(Score 0-3)








Alveolar Edema
  0 ± 0
 6.7 ± 25.8
 6.7 ± 25.8
35.7 ± 49.7
21.4 ± 42.6
28.6 ± 46.9


(% Positive Slides)








Alveolar Hemorrhage
  0 ± 0
  0 ± 0
  0 ± 0
  0 ± 0
  0 ± 0
 7.1 ± 26.7


(% Positive Slides)








Presence of Type II
  0 ± 0a
20.0 ± 41.4a
53.3 ± 51.6
78.6 ± 42.6
78.6 ± 42.6
57.1 ± 51.4


Pneumocyte








Hyperplasia








(% Positive Slides)








Severity of Bronchiolitis
  0 ± 0b
0.20 ± 0.41
0.53 ± 0.52
0.64 ± 0.63
0.71 ± 0.61
0.50 ± 0.65


(Score 0-3)








Extent of Lymphocytic
0.71 ± 0.73
0.67 ± 0.62
0.93 ± 0.70
1.07 ± 0.73
0.78 ± 0.78
0.93 ± 0.73


Cuffing








(Score 0-3)








Severity of Tracheitis/
0.07 ± 0.27
  0 ± 0
 0.7 ± 0.26
0.14 ± 0.36
  0 ± 0
0.08 ± 0.28


Bronchitis








(Score 0-3)
















Data Represent Mean ± Std Dev


aP < 0.05 vs. Irrelvant Human IgG1 Groups One-Way ANOVA (Kruskal-Wallis test) and Dunn's Post Hoc Test


bP < 0.005 vs. Irrelvant Human IgG1 Groups









Thus, in a sub-lethal mouse model of influenza A virus infection, VIS410 was protective at the highest doses (e.g., 2 and 20 mg/kg) while at suboptimal (e.g., sub-therapeutic) doses VIS410 neither protected nor elicited ADE, e.g., as measured by morbidity, mortality, virology and pathology assessments.


Example 7: Anti-Influenza Antibody VIS410 Targets a Broadly Conserved Epitope on Hemagglutinin

Given the rapid evolution of HA, a sequence analysis of historical and currently circulating influenza strains was performed to monitor the conservation and evolution of VIS410 epitope residues, and the impact of these observed polymorphisms on binding and neutralization was assessed.


Methods


The VIS410 epitope was predicted using experimental data and in silico antibody docking methods. Sequences of influenza HA from various subtypes were collected from GenBank and the Global Initiative on Sharing Avian Influenza Data (GISAID). A bioinformatics analysis was performed to analyze the composition and evolution of amino acids found at VIS410 epitope positions in HA. ELISA was used to assay VIS410 for binding to HAs that differ in epitope amino acids, and virus neutralization assays were used to assess VIS410's ability to neutralize influenza viruses with epitope variation.


Results


VIS410 binds to an epitope that is highly conserved in group 1 and group 2 HAs and an analysis of over 44,000 sequences shows that the natural variability in these residues is limited within each group. Polymorphisms at epitope positions that occur at >1% were identified and interrogated in the context of existing strains harboring these mutations. VIS410 neutralized influenza virus strains that together covered >97% of the observed positional variability at each epitope position in H1 strains and >93% of the positional variability at each epitope position in H3 strains. Furthermore, when combined with ELISA binding data, VIS410 was empirically shown to bind to epitopes with amino acid content found in >99% of HA sequences.


Specifically, to assess VIS410's breadth of binding and tolerance to sequence variation, a panel of seasonal influenza strains were selected with diverse geographic origin and spanning 4 decades. Strains exhibiting polymorphisms at the VIS410 epitope were identified based on a sequence analysis of available HA sequences, and were included in the panel. Strains were included that contain sequence diversity at epitope positions where the most frequently amino acid was observed at <95% (orange columns below). VIS410 successfully neutralized this diverse panel of strains in a cell-based microneutralization assay. The results are shown in FIG. 18.


H1N1 sequences were analyzed for isolates collected from 2005 through 2016 (obtained from EpiFlu). In order to monitor the trajectory of the sequence diversity, the sequence entropy for epitope residues as well as all surface residues were calculated over this time period. The mean sequence entropy is shown using a heatmap (FIG. 19). VIS410 epitope residues show lower sequence entropy (higher conservation) than non-epitope surface residues. Of note in this analysis, even during the 2009 H1N1 pandemic, the VIS410 epitope showed little sequence variability.


Thus, VIS410 displays broad binding and neutralization and is tolerant to observed polymorphisms in its epitope including newly emerging mutations found in currently circulating strains.


INCORPORATION BY REFERENCE

All publications, patents, and patent applications mentioned herein are hereby incorporated by reference in their entirety as if each individual publication, patent or patent application was specifically and individually indicated to be incorporated by reference. In case of conflict, the present application, including any definitions herein, will control.


EQUIVALENTS

Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, many equivalents to the specific embodiments of the invention described herein. Such equivalents are intended to be encompassed by the following claims.

Claims
  • 1.-22. (canceled)
  • 23. A method of treating a human subject, the method comprising administering to the subject an amount of an anti-HA antibody molecule of between 11 and 16 mg/kg, wherein the anti-HA antibody molecule comprises: (a) a heavy chain immunoglobulin variable region segment comprising: a CDR1 comprising the sequence of SEQ ID NO:68; a CDR2 comprising the sequence of SEQ ID NO:69; and a CDR3 comprising the sequence of SEQ ID NO:70; and (b) a light chain immunoglobulin variable region segment comprising: a CDR1 comprising the sequence of SEQ ID NO: 145; a CDR2 comprising the sequence of SEQ ID NO:72; and a CDR3 comprising the sequence of SEQ ID NO:73,thereby treating the subject.
  • 24. The method of claim 23, wherein the subject is infected, or is at risk of being infected, with an influenza virus.
  • 25. The method of claim 23, wherein the antibody molecule is administered to prevent the subject from influenza, or a disorder associated with influenza.
  • 26. The method of claim 23, wherein the influenza virus is an H1N1 virus, an H3N2 virus, an H7N9 virus, or a combination thereof.
  • 27. The method of claim 23, wherein the antibody molecule is administered at a dose of between 12 and 14 mg/kg.
  • 28. The method of claim 23, wherein the antibody molecule is administered at a dose of between 11 and 14 mg/kg.
  • 29. The method of claim 23, wherein the antibody molecule is administered at a dose of between 11 and 12 mg/kg.
  • 30. The method of claim 23, wherein the subject is 65 years of age or above.
  • 31. The method of claim 23, wherein the antibody molecule is administered 1 to 15 weeks prior to the date of an epidemic peak of influenza in a region where the subject resides.
  • 32. The method of claim 23, wherein the antibody molecule is administered 2 to 10 weeks prior to the date of an epidemic peak of influenza in a region where the subject resides.
  • 33. The method of claim 23, wherein the antibody molecule is administered 4 to 8 weeks prior to the date of an epidemic peak of influenza in a region where the subject resides.
  • 34. The method of claim 23, wherein the subject resides in a single-family residence, an assisted living facility, a hospital, nursing home, or an institution in which more than 2 unrelated people reside.
  • 35. The method of claim 23, wherein administering comprises a single intravenous infusion.
  • 36. The method of claim 23, further comprising administering to the subject a second therapeutic agent for influenza, or a disorder or symptom associated with influenza.
  • 37. The method of claim 23, wherein said antibody molecule comprises a heavy chain immunoglobulin variable region segment that comprises SEQ ID NO: 25.
  • 38. The method of claim 23, wherein said antibody molecule comprises a light chain immunoglobulin variable region segment that comprises SEQ ID NO: 52.
  • 39. The method of claim 23, wherein said antibody molecule comprises a heavy chain immunoglobulin variable region segment that comprises SEQ ID NO: 25 and a light chain immunoglobulin variable region segment that comprises SEQ ID NO: 52.
  • 40. The method of claim 23, wherein said antibody molecule is an IgG antibody.
  • 41. A method of treating a human subject, the method comprising administering to the subject an amount of an anti-HA antibody molecule of between 11 and 16 mg/kg, wherein the subject is infected, or is at risk of being infected, with an influenza virus, and is 65 years of age or above; andwherein the anti-HA antibody molecule comprises: (a) a heavy chain immunoglobulin variable region segment comprising: a CDR1 comprising the sequence of SEQ ID NO:68; a CDR2 comprising the sequence of SEQ ID NO:69; and a CDR3 comprising the sequence of SEQ ID NO:70; and (b) a light chain immunoglobulin variable region segment comprising: a CDR1 comprising the sequence of SEQ ID NO: 145; a CDR2 comprising the sequence of SEQ ID NO:72; and a CDR3 comprising the sequence of SEQ ID NO:73,thereby treating the subject.
  • 42. A method of preventing a human subject from becoming infected with influenza virus, the method comprising administering to the subject an amount of an anti-HA antibody molecule of between 11 and 16 mg/kg, wherein the anti-HA antibody molecule is administered 1 to 15 weeks prior to the date of an epidemic peak of influenza in a region that includes the city, province or state, in which the subject lives; andwherein the anti-HA antibody molecule comprises: (a) a heavy chain immunoglobulin variable region segment comprising: a CDR1 comprising the sequence of SEQ ID NO:68; a CDR2 comprising the sequence of SEQ ID NO:69; and a CDR3 comprising the sequence of SEQ ID NO:70; and (b) a light chain immunoglobulin variable region segment comprising: a CDR1 comprising the sequence of SEQ ID NO: 145; a CDR2 comprising the sequence of SEQ ID NO:72; and a CDR3 comprising the sequence of SEQ ID NO:73,thereby preventing the subject from becoming infected with influenza virus.
CROSS REFERENCE TO RELATED APPLICATIONS

This application is a continuation of U.S. patent application Ser. No. 15/349,235, filed Nov. 11, 2016, which claims the benefit of U.S. Provisional Application No. 62/255,262, filed Nov. 13, 2015, U.S. Provisional Application No. 62/299,141, filed Feb. 24, 2016, and U.S. Provisional Application No. 62/315,977, filed Mar. 31, 2016. The contents of the aforementioned applications are hereby incorporated by reference in their entirety.

Provisional Applications (3)
Number Date Country
62315977 Mar 2016 US
62299141 Feb 2016 US
62255262 Nov 2015 US
Continuations (1)
Number Date Country
Parent 15349235 Nov 2016 US
Child 16674710 US