Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
PROTEIN-ANTIVIRAL COMPOUND CONJUGATES
Document Type and Number:
WIPO Patent Application WO/2023/009754
Kind Code:
A2
Abstract:
Provided herein are compounds, compositions, and methods for the treatment of diseases and disorders associated with influenza, including VX-787 and derivatives thereof, and protein (e.g., antibody) drug conjugates thereof.

Inventors:
NITTOLI THOMAS (US)
Application Number:
PCT/US2022/038723
Publication Date:
February 02, 2023
Filing Date:
July 28, 2022
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
REGENERON PHARMA (US)
International Classes:
A61K47/68; A61P31/16; C07D471/04; C07K16/10
Domestic Patent References:
WO2016100807A22016-06-23
WO2005089808A22005-09-29
WO2013055993A12013-04-18
WO2013055990A12013-04-18
WO2013053873A12013-04-18
WO2013053872A12013-04-18
WO2011130598A12011-10-20
WO2008122039A22008-10-09
WO2013068874A12013-05-16
WO2012166559A12012-12-06
WO2012005982A22012-01-12
WO2014065661A12014-05-01
WO2012059882A22012-05-10
Foreign References:
US20160176953A12016-06-23
NL192003B1996-08-01
US6596541B22003-07-22
US20160176953A12016-06-23
US5208020A1993-05-04
US20100129314A12010-05-27
US5714586A1998-02-03
US20130101546A12013-04-25
US58559212A
US20070258987A12007-11-08
US7750116B12010-07-06
US20080305497A12008-12-11
Other References:
"GenBank", Database accession no. ACP44150.1
VERT MSCHWACH GENGEL RCOUDANE J, J CONTROL RELEASE, vol. 53, no. 1-3, 1998, pages 85 - 92
JAIN R A, BIOMATERIALS, vol. 21, no. 23, 2000, pages 2475 - 2490
UHRICH K ECANNIZZARO S MLANGER R SSHAKESHEFF K M, CHEMICAL REVIEWS, vol. 99, no. 11, 1999, pages 3181 - 3198
PARK T G, BIOMATERIALS, vol. 16, no. 15, 1995, pages 1123 - 1130
PADLAN ET AL., FASEB J, vol. 9, 1995, pages 133 - 139
VAJDOS ET AL., J MOL BIOL, vol. 320, 2002, pages 415 - 428
NUCL. ACIDS RES., vol. 20, 1992, pages 6287 - 6295
ANGAL ET AL., MOLECULAR IMMUNOLOGY, vol. 30, 1993, pages 105
ABDICHE, Y.N. ET AL., ANALYTICAL BIOCHEMISTRY, vol. 377, no. 2, 2008, pages 209 - 217
PEARSON, METHODS MOL. BIOL., vol. 24, 1994, pages 307 - 331
GONNET ET AL., SCIENCE, vol. 256, 1992, pages 1443 - 45
ALTSCHUL ET AL., J. MOL. BIOL., vol. 215, 1990, pages 403 - 410
NUCLEIC ACIDS RES., vol. 25, 1997, pages 3389 - 3402
LLOYD, THE ART, SCIENCE AND TECHNOLOGY OF PHARMACEUTICAL COMPOUNDING, 1999
SUI ET AL., NATURE STRUCT, AND MOL. BIOL., 22 February 2009 (2009-02-22), pages 1 - 9
AUSUBEL ET AL., CURRENTPROTOC. MOL. BIOL.
HOLLANDER ET AL., BIOCONJUGATE CHEM., vol. 19, 2008, pages 358 - 361
HOFER ET AL., PROC. NATL. ACAD. SCI., USA, vol. 105, 2008, pages 12451 - 12456
CARRICO ET AL., NAT. CHEM. BIOL., vol. 3, 2007, pages 321 - 322
AGARWAL ET AL., PROC. NATL. ACAD. SCI., USA, vol. 110, 2013, pages 46 - 51
RABUKA ET AL., NAT. PROTOCOLS, vol. 10, 2012, pages 1052 - 1067
RYAN ET AL., FOOD & AGRICULTURE IMMUNOL., vol. 13, 2001, pages 127 - 130
JEGER ET AL., ANGEW CHEM INT ED ENGL., vol. 49, 2010, pages 9995 - 9997
DENNLER ET AL., BIOCONJUGATE CHEM, vol. 25, 2014, pages 569 - 578
DENNLER ET AL., BIOCONJUGATE CHEM., vol. 25, 2014, pages 569 - 578
"Antibody-Drug Conjugates and Immunotoxins", 2013, SPRINGER VERLAG
"Antibody-Drug Conjugates", 2015, SPRINGER INTERNATIONAL PUBLISHING
J. MED. CHEM., vol. 57, 2014, pages 6668
DORONINA ET AL., NATURE BIOTECHNOLOGY, vol. 21, 2003, pages 778
ACSMEDICINAL CHEMISTRY LETTERS, vol. 8, 2017, pages 261 - 265
EUROPEAN JOURNAL OF MEDICINAL CHEMISTRY, vol. 162, 2019, pages 249 - 265
TETRAHEDRON, vol. 74, 2018, pages 1951 - 1956
ACS MED. CHEM. LETT., vol. 8, 2017, pages 261 - 265
JOURNAL OF MEDICINAL CHEMISTRY, vol. 57, no. 15, 2014, pages 6668 - 6678
Attorney, Agent or Firm:
PATHAK, Rahul (US)
Download PDF:
Claims:
WHAT IS CLAIMED IS:

1. A drug conjugate according to the following formula: wherein,

BA is a binding agent;

R1 is F and R2 is H; or R1 and R2 cyclize to form a fused five-membered heteroaryl ring, optionally substituted with methyl;

R3 is H or HO-CH2-;

Cy is bridged 5 or 6 membered cycloalkyl, wherein the bridge is methylene or ethylene;

Q is -O- or -0-NH-; wherein when R3 is H, then Q is -0-NH-;

L is a linker; and k is an integer from one to thirty; or a pharmaceutically acceptable salt thereof.

2. The drug conjugate of claim 1 according to formula 101.

3. The drug conjugate of claim 2, wherein R3 is HO-CH2-

4. The drug conjugate of claim 2, wherein R3 is H and Q is -0-NH-.

5. The drug conjugate of claim 1 according to formula 201.

6. The drug conjugate of any of the previous claims, wherein R1 is F and R2 is H.

7. The drug conjugate of any of the previous claims, wherein Cy is bridged 6-membered cycloalkyl.

8. The drug conjugate of any of the previous claims according to the following formula:

or a pharmaceutically acceptable salt thereof.

9. The drug conjugate of any of the previous claims according to the following formula: or a pharmaceutically acceptable salt thereof.

10. The drug conjugate of any of the previous claims according to the following formula : or a pharmaceutically acceptable salt thereof.

11. The drug conjugate of any of the previous claims according to the following formula :

(105) (205) ; or a pharmaceutically acceptable salt thereof.

12. The drug conjugate of any of the previous claims according to the following formula : or a pharmaceutically acceptable salt thereof.

13. The drug conjugate of any of the previous claims according to the following formula : or a pharmaceutically acceptable salt thereof.

14. The drug conjugate of any of the previous claims according to the following formula : or a pharmaceutically acceptable salt thereof.

15. The drug conjugate of any of the previous claims according to the following formula : or a pharmaceutically acceptable salt thereof.

16. The drug conjugate of any of the previous claims according to the following formula :

(112) 212 or a pharmaceutically acceptable salt thereof.

17. The drug conjugate of any of the previous claims according to the following formula :

(122) (222).

18. The drug conjugate of any of the previous claims wherein L is: wherein: each AA is an amino acid residue; and n is an integer from zero to ten.

19. The drug conjugate of claim 18, wherein the SP1 spacer is: wherein:

X is absent or -N(H)-;

RG' a reactive group residue following reaction of a reactive group RG with a binding agent, for instance -NH-, -CONH-, a maleimide residue, a click residue, or a Diels- Alder residue; n is an integer from zero to ten; and b is, independently, an integer from 1 to 92.

20. The drug conjugate of claim 18 or 19, wherein the SP2 spacer, is selected from the group consisting of -NH-jp-CeTUj-CTh-, -NH-f/j-GPUj-CPhOGC))-, -NH-(/>G,H4)-

21. The drug conjugate of any of claims 18-20, is wherein (AA)nis valine-citrulline, citrulline-valine, valine-alanine, alanine-valine, valine-glycine, glycine-valine, glutamate-valine-citrulline, glutamine-valine-citrulline, or glycine-glycine- phenylalanine-glycine.

22. The drug conjugate of any of claims 18-21, wherein (AA)„-SP2 is selected from the group consisting of: valine-citrulline-PABC, citrulline-valine-PABC, glutamate- valine-citrulline-PABC, glutamine-valine-citrulline-PABC, glycine-glycine- phenylalanine-glycine-N(H)-CH2-, valine-alanine-PABC, valine-citrulline-NH-(/?- C6H4)-CH2-, \ aline-citrulline-NH-(/2-G,H4)-CH(CH4)0-. valine-alanine-NH-(/> C6H4)-CH2-, or valine-alanine-NH-G-GPBj-CfhOGO)-. The compound of claim any of the previous claims, selected from the group consisting of WO 2023/009754

or a pharmaceutically acceptable salt thereof; wherein

BA is an antibody or an antigen binding fragment thereof; and k is an integer from one to thirty.

24. The drug conjugate of any of the previous claims, wherein BA is an antibody or an antigen-binding fragment thereof.

25. The drug conjugate of any of the previous claims, wherein BA is an anti -influenza antibody or an antigen binding fragment thereof.

26. The drug conjugate of any of the previous claims, wherein BA is an anti hemagglutinin antibody or an antigen binding fragment thereof.

27. The antibody-drug conjugate or compound of any of the preceding claims, wherein BA is an anti-influenza A Group 1 antibody or an antigen binding fragment thereof.

28. The antibody-drug conjugate or compound of any of the preceding claims, wherein BA is an anti-influenza HI antibody or an antigen binding fragment thereof.

29. The antibody-drug conjugate or compound of any of the preceding claims, wherein BA is an anti-influenza A Group 2 antibody or an antigen binding fragment thereof.

30. The antibody-drug conjugate or compound of any of the preceding claims, wherein BA is an anti-influenza H3 antibody or an antigen binding fragment thereof.

31. The antibody-drug conjugate or compound of any of the preceding claims, wherein BA is an anti-influenza B antibody or an antigen binding fragment thereof.

32. The antibody-drug conjugate of any preceding claim, wherein the antibody-drug conjugate binds to and/or inhibits polymerase basic protein 2 (PB2) and/or polymerase basic protein 1 (PB1).

33. The drug conjugate or compound of any of the preceding claims, wherein BA is an antibody or antigen-binding fragment thereof comprising at least one glutamine residue for conjugation.

34. The conjugate or compound of any of the preceding claims, wherein BA is an antibody or antigen-binding fragment thereof comprising at least two, three, or four glutamine residues for conjugation.

35. The conjugate or compound of any of the preceding claims, wherein BA is an antibody or antigen-binding fragment thereof, wherein conjugation is at two Q295 residues in the EU numbering system; and k is 2.

36. The conjugate or compound of any of the preceding claims, wherein BA is an antibody or antigen-binding fragment thereof, wherein conjugation is at two Q295 residues and at two N297Q residues in the EU numbering system; and k is 4.

37. The drug conjugate of any of the preceding claims, wherein BA is an antibody or antigen-binding fragment thereof comprising an antibody heavy chain, wherein conjugation is at the C-terminus of the heavy chain; and k is 2.

38. The drug conjugate or compound of claim 29, wherein conjugation is via a glutamine at the C-terminus of the antibody heavy chain.

39. The drug conjugate or compound of claim 29, wherein conjugation is via a glutamine in a LLQGA sequence at the C-terminus of the antibody heavy chain.

40. The antibody-drug conjugate of any preceding claim, wherein the antibody or antigen binding fragment thereof comprises three LCDRs of an LCVR comprising the amino acid sequence set forth in SEQ ID NO: 26; and three HCDRs of an HCVR comprising the amino acid sequence set forth in SEQ ID NO: 18.

41. The antibody-drug conjugate of any preceding claim, wherein the antibody or antigen binding fragment thereof comprises a LCVR further comprising an amino acid sequence set forth in SEQ ID NO: 26; and a HCVR further comprising an amino acid sequence set forth in SEQ ID NO: 18.

42. The antibody-drug conjugate of any preceding claim, wherein the antibody comprises a. a HCDR1 that comprises an amino acid sequence set forth in SEQ ID NO: 20; b. a HCDR2 that comprises an amino acid sequence set forth in SEQ ID NO: 22; c. a HCDR3 that comprises an amino acid sequence set forth in SEQ ID NO: 24; d. a LCDR1 that comprises an amino acid sequence set forth in SEQ ID NO: 28; e. a LCDR2 that comprises an amino acid sequence set forth in SEQ ID NO: 30; and f. a LCDR3 that comprises an amino acid sequence set forth in SEQ ID NO: 32.

43. The drug conjugate or compound of any of the preceding claims, wherein BA is mAh 11729.

44. The antibody-drug conjugate of any of the previous claims having a structure selected from:

wherein indicates linkage to BA.

45. A pharmaceutical composition comprising the antibody-drug conjugate or compound of any preceding claim, and a pharmaceutically acceptable excipient, carrier, or diluent.

46. The pharmaceutical composition of claim 45, wherein the pharmaceutical composition is formulated for an administration selected from the group consisting of: oral, intravenous, intraperitoneal, inhalation, and intranasal.

47. A method for treatment, prophylaxis, reduction, or inhibition of a disease, disorder, or condition associated with an infection in a subject, comprising administering to the subject an effective amount of an antibody-drug conjugate, compound, or pharmaceutical composition of any preceding claim.

48. The method of claim 47, wherein the infection is a viral infection.

49. The method of claim 47, wherein the infection is influenza virus infection.

50. The method of claim 47, wherein the infection is influenza A virus infection.

51. The method of claim 47, wherein the infection is influenza B virus.

52. The method of claim 47, wherein the infection is influenza A virus infection and influenza B virus infection.

53. The method of any one of claims 47-52, wherein side effects of the compound when administered to the subject are reduced when compared to administration of the unconjugated antiviral compound to a comparable subject.

54. A method for treatment, prophylaxis, reduction, or inhibition of an influenza infection in a subject comprising administering to the subject an effective amount of an antibody-drug conjugate, compound, or pharmaceutical composition of any preceding claim.

55. The method of claim 54, wherein the influenza infection is caused by influenza A virus infection.

56. The method of claim 54, wherein the influenza infection is caused by an influenza A Group 1 virus.

57. The method of claim 54, wherein the influenza infection is caused by an influenza A HI virus.

58. The method of claim 54, wherein the influenza infection is caused by an influenza A Group 2 virus.

59. The method of claim 54, wherein the influenza infection is caused by an influenza A H3 virus.

60. The method of claim 54, wherein the influenza infection is caused by an unknown or undetermined influenza virus.

61. The method of claim 54, wherein the influenza infection is caused by influenza B virus infection.

62. The method of claim 54, wherein the influenza infection is caused by influenza A virus infection and influenza B virus infection.

63. The method of any one of claims 54-62, wherein the antibody-drug conjugate, compound, or pharmaceutical composition is administered in combination with a supplementary therapeutic agent.

64. The method of claim 63, wherein the supplementary therapeutic is selected from the group consisting of: an anti-viral drug, an anti-inflammatory drug, an antibody that binds specifically to influenza HA, a vaccine for influenza, a dietary supplement, and a palliative therapy to treat an influenza infection.

65. The method of claim 63, wherein the anti-inflammatory drug is selected from the group consisting of corticosteroids and non-steroidal anti-inflammatory drugs.

66. The method of claim 63, wherein the dietary supplement is an anti-oxidant.

67. The method of any one of claims 63-66, wherein the supplementary therapeutic agent is administered via a different route of administration as the antibody-drug conjugate, compound, or pharmaceutical composition.

68. The method of any one of claims 63-67, wherein the supplementary therapeutic agent is administered orally.

69. The method of any one of claims 63-68, wherein the anti-viral drug is oseltamivir.

70. The method of claim 69, wherein the oseltamivir is administered prior to administration of the antibody-drug conjugate, compound, or pharmaceutical composition.

71. The method of claim 69, wherein the oseltamivir is administered concurrently with the antibody-drug conjugate, compound, or pharmaceutical composition.

72. The method of claim 67, wherein the oseltamivir is administered after administration of the antibody-drug conjugate, compound, or pharmaceutical composition.

73. A linker-antiviral compound having the following structure

(404) (405) or a pharmaceutically acceptable salt thereof, wherein L is a linker;

RG is a reactive moiety, for instance -Nth, NHS ester, maleimide residue, azide, alkyne, strained alkyne, diene, or dienophile;

R1 is F and R2 is H; or R1 and R2 cyclize to form a fused five-membered heteroaryl ring, optionally substituted with methyl;

R3 is H or HO-CH2-;

Q is -O- or -0-NH-; wherein when R3 is H, then Q is -0-NH-; and

Cy is bridged 5 or 6 membered cycloalkyl, wherein the bridge is methylene or ethylene.

74. The linker-payload of claim 73 wherein L is: wherein: each AA is an amino acid residue; and n is an integer from zero to ten.

75. The linker-payload of claim 73 or74, wherein the SP1 spacer is: wherein: n is an integer from zero to ten; and b is, independently, an integer from 1 to 92.

76. The linker-payload of any of claims 73-75, wherein the SP2 spacer, is selected from the group consisting of: -NH-G-CeTLO-CTh-, -NH-l/j-CXfhl-CfbOCXO)-. -NH-(/>

77. The linker-payload of any of claims 73-76, wherein (AA)nis selected from the group consisting of: valine-citrulline, citrulline-valine, valine-alanine, alanine-valine, valine- glycine, glycine-valine, glutamate-valine-citrulline, glutamine-valine-citrulline, and glycine-glycine-phenylalanine-glycine.

78. The linker-payload of any of claims 73-77, wherein (AA)n-SP2 is selected from the group consisting of: valine-citrulline-PABC, citrulline-valine-PABC, glutamate- valine-citrulline-PABC, glutamine-valine-citrulline-PABC, glycine-glycine- phenylalanine-glycine-N(H)-CH2-, valine-alanine-PABC, valine-ci trull ine-NH-(/> C6H4)-CH2-, valine-ci trull ine-NH-(/>G,H4)-CfI(CtT,)0-. valine-alanine-NH-(/> C6H4)-CH2-, and Yaline-alamne-NH-(/?-G,H4)-CfhOGC))-

80. An antibody-drug conjugate comprising an antibody, or an antigen binding fragment thereof, where the antibody or antigen binding fragment thereof is conjugated to a compound of any of claims 73-79.

81. A method of preparing an antibody-drug conjugate comprising contacting a binding agent with a linker-antiviral compound of any of claims 73-80.

82. A compound according to the following formula:

(301) (311) wherein,

R1 is F and R2 is H; or R1 and R2 cyclize to form a fused five-membered heteroaryl ring, optionally substituted with methyl, for example as -C=CH-S- or as -C=CH-NMe-;

R3 is H or HO-CH2-;

Cy is bridged 5 or 6 membered cycloalkyl, wherein the bridge is methylene or ethylene; Q is -O- or -0-NH-; wherein when R3 is H, then Q is -0-NH-; or a pharmaceutically acceptable salt thereof.

83. The compound of claim 82, wherein R3 is HO-CH2-

84. The compound of claim 82, wherein R3 is H and Q is -0-NH-.

85. The compound of any of claims 82-84, wherein R1 is F and R2 is H.

86. The compound of any of claims 82-85, wherein Cy is bridged 6-membered cycloalkyl.

87. The compound of any of claims 82-85, wherein Cy is

88. The compound of any of claims 82-85, wherein Cy is and Q is -0-.

89. The compound of any of claims 82-85 according to the following formula:

or a pharmaceutically acceptable salt thereof.

90. The compound of any of claims 82-85 according to the following formula: or a pharmaceutically acceptable salt thereof.

91. The compound of any of claims 82-85 according to the following formula: or a pharmaceutically acceptable salt thereof.

92. A compound selected from the group consisting of:

and pharmaceutically acceptable salts thereof.

93. A pharmaceutical composition comprising the compound of any of claims 82-92 and one or more pharmaceutically acceptable carriers, excipients, or diluents.

94. A method of treatment according to any of the previous claims comprising the step of administering to a subject in need thereof the compound of any of claims 82-92 or the pharmaceutical composition of claim 91.

95. The compound or composition of any of the previous claims use in treatment.

96. The compound or composition of any of the previous claims for use in treatment of an influenza infection in a subject in need thereof.

97. Use of the compound or composition of any of the previous claims for manufacture of a medicament.

98. Use of the compound or composition of any of the previous claims for manufacture of a medicament for the treatment of an influenza infection in a subject in need thereof.

Description:
PROTEIN-ANTIVIRAL COMPOUND CONJUGATES

GOVERNMENT LICENSE RIGHTS

[001] This invention was made with Government support under Agreement HHS0100201700020C, awarded by the U.S. Department of Health and Human Services. The Government has certain rights in the invention.

CROSS REFERENCE TO RELATED APPLICATIONS

[002] The present application claims the benefit of US provisional application no. 63/226,713, filed July 28, 2021, the content of which is hereby incorporated by reference in its entirety.

FIELD

[003] Provided herein are antiviral compounds and protein conjugates thereof, and methods for treating a variety of diseases, disorders, and conditions including administering the antiviral compounds, and protein conjugates thereof.

BACKGROUND

[004] Influenza is a highly contagious disease, which has a long history characterized by waves of pandemics, epidemics, resurgences, and outbreaks. In spite of annual vaccination efforts, influenza infections result in substantial morbidity and mortality.

[005] Influenza viruses consist of three main types, A, B, and C. Influenza A viruses can be classified into subtypes based on allelic variations in antigenic regions of two genes that encode the surface glycoproteins, hemagglutinin (HA) and neuraminidase (NA), which are required for viral attachment and entry into the host cell.

[006] Hemagglutinin is a trimeric glycoprotein that contains two structural domains, a globular head domain that consists of the receptor-binding site (that is subject to frequent antigenic drift) and the stem region (more conserved among various strains of influenza virus). The HA protein is synthesized as a precursor (HA0), which undergoes proteolytic processing to produce two subunits (HA1 and HA2), which associate with one another to form the stem/globular head structure. The HA1 peptide is responsible for the attachment of virus to the cell surface. The HA2 peptide forms a stem-like structure that mediates the fusion of viral and cell membranes in endosomes, allowing the release of the ribonucleoprotein complex into the cytoplasm.

[007] Currently, there are eighteen subtypes defined by their hemagglutinin proteins (HI - H18). The eighteen HAs can be classified into two groups. Group 1 consists of HI, H2, H5, H6, H8, H9, HI 1, H12, H13, H16, H17, and H18 subtypes, and group 2 includes H3, H4, H7, H10, H14, and H15 subtypes.

[008] Despite decades of research, there are no marketed antibodies or antibody-drug conjugates (ADCs) that broadly neutralize or inhibit influenza A virus infection or attenuate disease caused by influenza A virus. Therefore, there is a need to identify new antibodies and ADCs that neutralize multiple subtypes of influenza A virus and can be used as medicaments for prevention or therapy of influenza A infection.

SUMMARY

[009] Provided herein are compounds useful, for example, in antiviral treatments. In certain embodiments, the compounds include VX-787 and derivatives thereof. In one embodiment, provided is an antibody-drug conjugate including an anti-influenza antibody or antigen-binding fragment thereof conjugated to a payload (e.g., an antiviral compound), linker-payload (e.g., linker-antiviral compound), and/or compound as described herein.

[0010] In certain embodiments, provided are compounds having the following structure

(101) (201) wherein L is a linker; BA is a binding agent; R 1 is F and R 2 is H; or R 1 and R 2 cyclize to form a fused five-membered heteroaryl ring, optionally substituted with methyl; R 3 is H or HO-CH2- ; Cy is bridged 5 or 6 membered cycloalkyl, wherein the bridge is methylene or ethylene;

Q is -O- or -0-NH-; wherein when R 3 is H, then Q is -0-NH-; and k is an integer from one to thirty.

[0011] In certain embodiments, provided are linker-payloads (e.g., linker-antiviral compounds) having the following structure:

(404) (405) or a pharmaceutically acceptable salt thereof, wherein L is a linker; R 1 is F and R 2 is H; or R 1 and R 2 cyclize to form a fused five-membered heteroaryl ring, optionally substituted with methyl; R 3 is H or HO-CH2-; Cy is bridged 5 or 6 membered cycloalkyl, wherein the bridge is methylene or ethylene; Q is -O- or -0-NH-; and RG is a reactive moiety.

[0012] In certain embodiments, provided are compounds having the following structure wherein R 1 is F and R 2 is H; or R 1 and R 2 cyclize to form a fused five-membered heteroaryl ring, optionally substituted with methyl; R 3 is H or HO-CH2-; Cy is bridged 5 or 6 membered cycloalkyl, wherein the bridge is methylene or ethylene; and Q is -O- or -0-NH-; wherein when R 3 is H, then Q is -0-NH-.

[0013] In certain embodiments, provided are compounds having the following structure wherein R 1 is F and R 2 is H; or R 1 and R 2 cyclize to form a fused five-membered heteroaryl ring, optionally substituted with methyl; R 3 is H or HO-CH2-; Cy is bridged 5 or 6 membered cycloalkyl, wherein the bridge is methylene or ethylene; and Q is -O- or -0-NH-; wherein when R 3 is H, then Q is -0-NH-.

[0014] In another embodiment, set forth herein are methods for making the payloads or compounds, linker-payloads, or antibody-drug conjugates, and compositions described herein.

[0015] In another embodiment, provided herein are methods for the treatment, prophylaxis, reduction, or inhibition of a disease, disorder, or condition associated with an infection, as described herein, in a subject including administering to the subject an effective amount of a payload (e.g., antiviral compound), linker-payload (e.g., linker-antiviral compound), antibody- drug conjugate, or pharmaceutical composition as described herein. In certain embodiments, provided are the compounds or compositions described herein for use in treatment. In certain embodiments, provided the compounds or compositions described herein for use in treatment of an influenza infection in a subject in need thereof. In certain embodiments, provided are uses of a compound or composition described herein for manufacture of a medicament. In certain embodiments, provided are uses of a compound or composition described herein for manufacture of a medicament for the treatment of an influenza infection in a subject in need thereof. DESCRIPTION OF EXEMPLARY EMBODIMENTS [0016] Provided herein are compounds, compositions, and methods useful for treating, for example, influenza infections in a subject.

[0017] Although any methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present invention, the preferred methods and materials are now described. All patents, applications and non-patent publications mentioned in this specification are incorporated herein by reference in their entireties.

Definitions

[0018] When referring to the compounds provided herein, the following terms have the following meanings unless indicated otherwise. Unless defined otherwise, all technical and scientific terms used herein have the same meaning as is commonly understood by one of ordinary skill in the art. In the event that there is a plurality of definitions for a term provided herein, these Definitions prevail unless stated otherwise.

[0019] The phrase “influenza hemagglutinin,” also called “influenza HA” is a trimeric glycoprotein found on the surface of influenza virions, which mediates viral attachment (via HA1 binding to a-2,3- and a-2,6-sialic acids) and entry (through conformational change) into host cells. The HA is comprised of two structural domains: a globular head domain containing the receptor binding site (subject to high frequency of antigenic mutations) and the stem region (more conserved among various strains of influenza virus). The influenza HA is synthesized as a precursor (HA0) that undergoes proteolytic processing to produce two subunits (HA1 and HA2) which associate with one another to form the stem/globular head structure. The viral HA is the most variable antigen on the virus (eighteen subtypes can be classified into two groups), but the stem (HA2) is highly conserved within each group.

[0020] The amino acid sequence of full-length Influenza HA is exemplified by the amino acid sequence of influenza isolate HI N1 A/Califomia/04/2009 provided in GenBank as accession number FJ966082.1. The phrase “influenza-HA” also includes protein variants of influenza HA isolated from different influenza isolates, e.g., GQ149237.1, NC_002017, KM972981.1, etc. The phrase “influenza-HA” also includes recombinant influenza HA or a fragment thereof. The phrase also encompasses influenza HA or a fragment thereof coupled to, for example, histidine tag, mouse or human Fc, or a signal sequence.

[0021] The phrase “influenza infection,” as used herein, also characterized as “flu” refers to the severe acute respiratory illness caused by influenza virus. The phrase includes respiratory tract infection and the symptoms that include high fever, headache, general aches and pains, fatigue and weakness, in some instances extreme exhaustion, stuffy nose, sneezing, sore throat, chest discomfort, cough, shortness of breath, bronchitis, pneumonia, and death in severe cases.

[0022] As used herein, “alkyl” refers to a monovalent and saturated hydrocarbon radical moiety. Alkyl is optionally substituted and can be linear, branched, or cyclic, i.e., cycloalkyl. Alkyl includes, but is not limited to, those radicals having 1-20 carbon atoms, i.e., Ci-20 alkyl; 1-12 carbon atoms, i.e., Ci-12 alkyl; 1-8 carbon atoms, i.e., Ci- 8 alkyl; 1-6 carbon atoms, i.e., Ci- 6 alkyl; and 1-3 carbon atoms, i.e., C1-3 alkyl. Examples of alkyl moieties include, but are not limited to, methyl, ethyl, «-propyl, /-propyl, «-butyl, s-butyl, /-butyl, /-butyl, a pentyl moiety, a hexyl moiety, cyclopropyl, cyclobutyl, cyclopentyl, and cyclohexyl. A pentyl moiety includes, but is not limited to, «-pentyl and /-pentyl. A hexyl moiety includes, but is not limited to, «-hexyl.

[0023] As used herein, “alkylene” refers to a divalent alkyl group. Unless specified otherwise, alkylene includes, but is not limited to, 1-20 carbon atoms. The alkylene group is optionally substituted as described herein for alkyl. In some embodiments, alkylene is unsubstituted.

[0024] Designation of an amino acid or amino acid residue without specifying its stereochemistry is intended to encompass the L- form of the amino acid, the D- form of the amino acid, or a racemic mixture thereof.

[0025] As used herein, “haloalkyl” refers to alkyl, as defined above, wherein the alkyl includes at least one substituent selected from a halogen, for example, fluorine (F), chlorine (Cl), bromine (Br), or iodine (I). Examples of haloalkyl include, but are not limited to, -CF3, -CH2CF3, -CCI2F, and -CCI3.

[0026] As used herein, “alkenyl” refers to a monovalent hydrocarbon radical moiety containing at least two carbon atoms and one or more non-aromatic carbon-carbon double bonds. Alkenyl is optionally substituted and can be linear, branched, or cyclic. Alkenyl includes, but is not limited to, those radicals having 2-20 carbon atoms, i.e., C2-20 alkenyl; 2- 12 carbon atoms, i.e., C2-12 alkenyl; 2-8 carbon atoms, i.e., C2-8 alkenyl; 2-6 carbon atoms, i.e., C 2-6 alkenyl; and 2-4 carbon atoms, i.e., C2-4 alkenyl. Examples of alkenyl moieties include, but are not limited to, vinyl, propenyl, butenyl, and cyclohexenyl.

[0027] As used herein, “alkynyl” refers to a monovalent hydrocarbon radical moiety containing at least two carbon atoms and one or more carbon-carbon triple bonds. Alkynyl is optionally substituted and can be linear, branched, or cyclic. Alkynyl includes, but is not limited to, those radicals having 2-20 carbon atoms, i.e., C2-20 alkynyl; 2-12 carbon atoms, i.e., C2-12 alkynyl; 2-8 carbon atoms, i.e., C2-8 alkynyl; 2-6 carbon atoms, i.e., C2-6 alkynyl; and 2-4 carbon atoms, i.e., C2-4 alkynyl. Examples of alkynyl moieties include, but are not limited to ethynyl, propynyl, and butynyl.

[0028] As used herein, “alkoxy” refers to a monovalent and saturated hydrocarbon radical moiety wherein the hydrocarbon includes a single bond to an oxygen atom and wherein the radical is localized on the oxygen atom, e.g., CH3CH2-O for ethoxy. Alkoxy substituents bond to the compound which they substitute through this oxygen atom of the alkoxy substituent. Alkoxy is optionally substituted and can be linear, branched, or cyclic, i.e., cycloalkoxy. Alkoxy includes, but is not limited to, those having 1-20 carbon atoms, i.e., Ci-20 alkoxy; 1-12 carbon atoms, i.e., Ci-12 alkoxy; 1-8 carbon atoms, i.e., Ci- 8 alkoxy; 1-6 carbon atoms, i.e., Ci- 6 alkoxy; and 1-3 carbon atoms, i.e., C1-3 alkoxy. Examples of alkoxy moieties include, but are not limited to, methoxy, ethoxy, «-propoxy. /-propoxy, «-butoxy. .v-butoxy. t-butoxy, /-butoxy. a pentoxy moiety, a hexoxy moiety, cyclopropoxy, cyclobutoxy, cyclopentoxy, and cyclohexoxy.

[0029] As used herein, “haloalkoxy” refers to alkoxy, as defined above, wherein the alkoxy includes at least one substituent selected from a halogen, e.g., F, Cl, Br, or I.

[0030] As used herein, “aryl” refers to a monovalent moiety that is a radical of an aromatic compound wherein the ring atoms are carbon atoms. Aryl is optionally substituted and can be monocyclic or polycyclic, e.g., bi cyclic or tricyclic. Examples of aryl moieties include, but are not limited to, those having 6 to 20 ring carbon atoms, i.e., Ce- 20 aryl; 6 to 15 ring carbon atoms, i.e., C6-i5aryl, and 6 to 10 ring carbon atoms, i.e., Ce-io aryl. Examples of aryl moieties include, but are limited to, phenyl, naphthyl, fluorenyl, azulenyl, anthryl, phenanthryl, and pyrenyl.

[0031] As used herein, “arylalkyl” refers to a monovalent moiety that is a radical of an alkyl compound, wherein the alkyl compound is substituted with an aromatic substituent, i.e., the aromatic compound includes a single bond to an alkyl group and wherein the radical is localized on the alkyl group. An arylalkyl group bonds to the illustrated chemical structure via H 2 the alkyl group. An arylalkyl can be represented by the structure, e.g., B , wherein B is an aromatic moiety, e.g., aryl or phenyl. Arylalkyl is optionally substituted, i.e., the aryl group and/or the alkyl group, can be substituted as disclosed herein. Examples of arylalkyl include, but are not limited to, benzyl.

[0032] As used herein, “alkylaryl” refers to a monovalent moiety that is a radical of an aryl compound, wherein the aryl compound is substituted with an alkyl substituent, i.e., the aryl compound includes a single bond to an alkyl group and wherein the radical is localized on the aryl group. An alkylaryl group bonds to the illustrated chemical structure via the aryl group.

An alkylaryl can be represented by the structure, e.g., , , , . or wherein B is an aromatic moiety, e.g., phenyl. Alkylaryl is optionally substituted, i.e., the aryl group and/or the alkyl group, can be substituted as disclosed herein. Examples of alkylaryl include, but are not limited to, toluyl.

[0033] As used herein, “aryloxy” refers to a monovalent moiety that is a radical of an aromatic compound wherein the ring atoms are carbon atoms and wherein the ring is substituted with an oxygen radical, i.e., the aromatic compound includes a single bond to an oxygen atom and wherein the radical is localized on the oxygen atom, e.g., for phenoxy. Aryloxy substituents bond to the compound which they substitute through this oxygen atom. Aryloxy is optionally substituted. Aryloxy includes, but is not limited to, those radicals having 6 to 20 ring carbon atoms, i.e., Ce-20 aryloxy; 6 to 15 ring carbon atoms, i.e.,

Ce-15 aryloxy, and 6 to 10 ring carbon atoms, i.e., Ce-io aryloxy. Examples of aryloxy moieties include, but are not limited to phenoxy, naphthoxy, and anthroxy.

[0034] As used herein, “arylene” refers to a divalent moiety of an aromatic compound wherein the ring atoms are only carbon atoms. Arylene is optionally substituted and can be monocyclic or polycyclic, e.g., bicyclic or tricyclic. Examples of arylene moieties include, but are not limited to those having 6 to 20 ring carbon atoms, i.e., Ce-20 arylene; 6 to 15 ring carbon atoms, i.e., Ce-15 arylene, and 6 to 10 ring carbon atoms, i.e., Ce-io arylene.

[0035] As used herein, “heteroalkyl” refers to an alkyl in which one or more carbon atoms are replaced by heteroatoms. As used herein, “heteroalkenyl” refers to an alkenyl in which one or more carbon atoms are replaced by heteroatoms. As used herein, “heteroalkynyl” refers to an alkynyl in which one or more carbon atoms are replaced by heteroatoms. Suitable heteroatoms include, but are not limited to, nitrogen, oxygen, and sulfur atoms. Heteroalkyl, heteroalkenyl, and heteroalkynyl are optionally substituted. Examples of heteroalkyl moieties include, but are not limited to, aminoalkyl, sulfonylalkyl, and sulfmylalkyl. Examples of heteroalkyl moieties also include, but are not limited to, methylamino, methylsulfonyl, and methylsulfmyl.

[0036] As used herein, “heteroaryl” refers to a monovalent moiety that is a radical of an aromatic compound wherein the ring atoms contain carbon atoms and at least one oxygen, sulfur, nitrogen, or phosphorus atom. Examples of heteroaryl moieties include, but are not limited to those having 5 to 20 ring atoms; 5 to 15 ring atoms; and 5 to 10 ring atoms. Heteroaryl is optionally substituted.

[0037] As used herein, “heteroarylene” refers to a divalent heteroaryl in which one or more ring atoms of the aromatic ring are replaced with an oxygen, sulfur, nitrogen, or phosphorus atom. Heteroarylene is optionally substituted.

[0038] As used herein, “heterocycloalky 1” refers to a cycloalkyl in which one or more carbon atoms are replaced by heteroatoms. Suitable heteroatoms include, but are not limited to, nitrogen, oxygen, and sulfur atoms. Heterocycloalkyl is optionally substituted. Examples of heterocycloalkyl moieties include, but are not limited to, morpholinyl, piperidinyl, tetrahydropyranyl, pyrrolidinyl, imidazolidinyl, oxazolidinyl, thiazolidinyl, dioxolanyl, dithiolanyl, oxanyl, orthianyl.

[0039] As used herein, “Lewis acid” refers to a molecule or ion that accepts an electron lone pair. The Lewis acids used in the methods described herein are those other than protons. Lewis acids include, but are not limited to, non-metal acids, metal acids, hard Lewis acids, and soft Lewis acids. Lewis acids include, but are not limited to, Lewis acids of aluminum, boron, iron, tin, titanium, magnesium, copper, antimony, phosphorus, silver, ytterbium, scandium, nickel, and zinc. Illustrative Lewis acids include, but are not limited to, AlBn, A1CT. BCT. boron trichloride methyl sulfide, BF3, boron trifluoride methyl etherate, boron trifluoride methyl sulfide, boron trifluoride tetrahydrofuran, dicyclohexylboron trifluoromethanesulfonate, iron (III) bromide, iron (III) chloride, tin (IV) chloride, titanium (IV) chloride, titanium (IV) isopropoxide, Cu(OTf)2, CuCh. CuBr2, zinc chloride, alkylaluminum halides (R n AlX3- n , wherein R is hydrocarbyl), Zn(OTf)2, ZnCh, Yb(OTf)3, Sc(OTfb, MgBr 2 , NiCh, Sn(OTf) 2 , Ni(OTf) 2 , and Mg(OTf) 2 .

[0040] As used herein, V-containing heterocycloalkyl,” refers to a cycloalkyl in which one or more carbon atoms are replaced by heteroatoms and wherein at least one replacing heteroatom is a nitrogen atom. Suitable heteroatoms in addition to nitrogen, include, but are not limited to, oxygen and sulfur atoms. A-containing heterocycloalkyl is optionally substituted. Examples of A-containing heterocycloalkyl moieties include, but are not limited to, morpholinyl, piperidinyl, pyrrolidinyl, imidazolidinyl, oxazolidinyl, or thiazolidinyl.

[0041] As used herein, “optionally substituted,” when used to describe a radical moiety, for example, optionally substituted alkyl, means that such moiety is optionally bonded to one or more substituents. Examples of such substituents include, but are not limited to, halo, cyano, nitro, amino, hydroxyl, optionally substituted haloalkyl, aminoalkyl, hydroxyalkyl, azido, epoxy, optionally substituted heteroaryl, optionally substituted heterocycloalkyl, are, independently at each occurrence, a hydrogen atom, alkyl, alkenyl, alkynyl, aryl, alkylaryl, arylalkyl, heteroalkyl, heteroaryl, or heterocycloalkyl, or R A and R B together with the atoms to which they are bonded, form a saturated or unsaturated carbocyclic ring, wherein the ring is optionally substituted, and wherein one or more ring atoms is optionally replaced with a heteroatom. In certain embodiments, when a radical moiety is optionally substituted with an optionally substituted heteroaryl, optionally substituted heterocycloalkyl, or optionally substituted saturated or unsaturated carbocyclic ring, the substituents on the optionally substituted heteroaryl, optionally substituted heterocycloalkyl, or optionally substituted saturated or unsaturated carbocyclic ring, if they are substituted, are not substituted with substituents which are further optionally substituted with additional substituents. In some embodiments, when a group described herein is optionally substituted, the substituent bonded to the group is unsubstituted unless otherwise specified.

[0042] Unless otherwise stated, structures depicted herein also are meant to include all isomeric (e.g., enantiomeric, diastereomeric, and geometric (or conformational)) forms of the structure; for example, the ( R )- and (S)- configurations for each asymmetric center, (Z)- and (E)- double bond isomers, and (Z)- and (E)- conformational isomers. Therefore, single stereochemical isomers as well as enantiomeric, diastereomeric, and geometric (or conformational) mixtures of the present compounds are within the scope of the description. Alternatively, as used herein, “enantiomeric excess (ee)” refers to a dimensionless mol ratio describing the purity of chiral substances that contain, for example, a single stereogenic center. For instance, an enantiomeric excess of zero would indicate a racemic (e.g., 50:50 mixture of enantiomers, or no excess of one enantiomer over the other). By way of further example, an enantiomeric excess of ninety -nine would indicate a nearly stereopure enantiomeric compound (i.e., large excess of one enantiomer over the other). The percentage enantiomeric excess, % ee = (|(//)-compound|-|CY)-compound|)/(|(//)-compound|+|CY)-compo und|) x 100, where the de compound > (///-compound; or % ee = ([(ri)-compound]-[(i?)-compound])/([(ri - compound l+l (///-compound | ) x 100, where the (///-compound > (///-compound. Moreover, as used herein, “diastereomeric excess (de)” refers to a dimensionless mol ratio describing the purity of chiral substances that contain more than one stereogenic center. For example, a diastereomeric excess of zero would indicate an equimolar mixture of diastereoisomers. By way of further example, diastereomeric excess of ninety-nine would indicate a nearly stereopure diastereomeric compound (i.e., large excess of one diastereomer over the other). Diastereomeric excess may be calculated via a similar method to ee. As would be appreciated by a person of skill, de is usually reported as percent de (% de). % de may be calculated in a similar manner to % ee.

[0043] As used herein, “binding agent” refers to any molecule, e.g., protein, antibody, or fragment thereof, capable of binding with specificity to a given binding partner, e.g., antigen.

[0044] As used herein, “linker” refers to a divalent, trivalent, or multivalent moiety that covalently links, or is capable of covalently linking (e.g., via a reactive group), the binding agent to one or more compounds described herein, for instance, payload or antiviral compounds and enhancement agents.

[0045] As used herein, “amide synthesis conditions” refers to reaction conditions suitable to effect the formation of an amide, e.g., by the reaction of a carboxylic acid, activated carboxylic acid, or acyl halide with an amine. In some examples, amide synthesis conditions refers to reaction conditions suitable to effect the formation of an amide bond between a carboxylic acid and an amine. In some of these examples, the carboxylic acid is first converted to an activated carboxylic acid before the activated carboxylic acid reacts with an amine to form an amide. Suitable conditions to effect the formation of an amide include, but are not limited to, those utilizing reagents to effect the reaction between a carboxylic acid and an amine, including, but not limited to, dicyclohexylcarbodiimide (DCC), diisopropylcarbodiimide (DIC), (benzotriazol-1 -yloxy)tris(dimethylamino)phosphonium hexafluorophosphate (BOP), (benzotriazol-1 -yloxy/tripyrrolidinophosphonium hexafluorophosphate (PyBOP), (7-azabenzotriazol-l-yloxy)tripyrrolidinophosphonium hexafluorophosphate (PyAOP), bromotripyrrolidinophosphonium hexafluorophosphate (PyBrOP), 0-(benzotriazol-l-yl)-/V,/V,/V , ,/V’ -tetramethyluronium hexafluorophosphate (HBTU), 0-(benzotriazol-l-yl)- V, V, V’, V’-tetramethyluronium tetrafluoroborate (TBTU),

1-[Bis(dimethylamino)methylene]-lH-l,2,3-triazolo[4,5-b]p yridinium 3-oxide hexafluorophosphate (HATU), N-ethoxycarbonyl-2-ethoxy-l,2-dihydroquinoline (EEDQ), N-ethyl-N'-(3-dimethylaminopropyl)carbodiimide (EDC),

2-chloro-l,3-dimethylimidazolidinium hexafluorophosphate (CIP),

2-chloro-4,6-dimethoxy-l,3,5-triazine (CDMT), carbonyldiimidazole (CDI), and l-Cyano-2- ethoxy-2-oxoethylidenaminooxy)dimethylamino-morpholino-carbe nium hexafluorophosphate (COMU). In some examples, a carboxylic acid is first converted to an activated carboxylic ester before treating the activated carboxylic ester with an amine to form an amide bond. In certain embodiments, the carboxylic acid is treated with a reagent. The reagent activates the carboxylic acid by deprotonating the carboxylic acid and then forms a product complex with the deprotonated carboxylic acid as a result of nucleophilic attack by the deprotonated carboxylic acid onto the protonated reagent. The activated carboxylic esters of certain carboxylic acids are subsequently more susceptible to nucleophilic attack by an amine than the carboxylic acid is before it is activated. This results in amide bond formation. As such, the carboxylic acid is described as activated. Exemplary reagents include DCC and DIC.

[0046] As used herein, the term “residue” refers to the chemical moiety within a compound that remains after a chemical reaction. For example, the term “amino acid residue” or - alkyl amino acid residue” refers to the product of an amide coupling or peptide coupling of an amino acid or a /V-alkyl amino acid to a suitable coupling partner; wherein, for example, a water molecule is expelled after the amide or peptide coupling of the amino acid or the /V-alkylamino acid, resulting in the product having the amino acid residue or /V-alkyl amino acid residue incorporated therein.

[0047] As used herein, “constitutional isomers” refers to compounds that have the same molecular formula, but different chemical structures resulting from the way the atoms are arranged. Exemplary constitutional isomers include «-propyl and isopropyl; «-butyl, .sec-butyl and /er/-butyl; and «-pentyl, isopentyl, and neopentyl, and the like.

[0048] Certain groups, moieties, substituents, and atoms are depicted with a wiggly line that intersects a bond or bonds to indicate the atom through which the groups, moieties, substituents, atoms are bonded. For example, a phenyl group that is substituted with an isopropyl group depicted as: has the following structure: . As used herein, illustrations showing substituents bonded to a cyclic group (e.g., aromatic, heteroaromatic, fused ring, and saturated or unsaturated cycloalkyl or heterocycloalkyl) through a bond between ring atoms are meant to indicate, unless specified otherwise, that the cyclic group may be substituted with that substituent at any ring position in the cyclic group or on any ring in the fused ring group, according to techniques set forth herein or which are known in the field to which the instant disclosure pertains. For example, the group, wherein subscript q is an integer from 0 to 4 and in which the positions of substituent R 1 are described genetically, i.e., not directly attached to any vertex of the bond line structure, i.e., specific ring carbon atom, includes the following, non-limiting examples of groups in which the substituent R 1 is bonded to a specific ring carbon atom:

[0049] As used herein, the phrase “reactive linker,” or the abbreviation “RL” refers to a monovalent group that includes a reactive group (“RG”) and spacer group (“SP”), depicted for example as , wherein RG is the reactive group and SP is the spacer group. As described herein, a reactive linker may include more than one reactive group and more than one spacer group. The spacer group is any divalent moiety that bridges the reactive group to another group, such as a payload (e.g., antiviral compound). The reactive linkers (RLs), together with the payloads (e.g., antiviral compounds) to which they are bonded, provide intermediates (“linker-payloads” (LPs) (e.g., linker-antiviral compounds)) useful as synthetic precursors for the preparation of the antibody conjugates described herein. As used herein, payloads can be antiviral compounds, and linker-payloads incorporating those antiviral compounds can be referred to as “linker-antiviral compounds.” The reactive linker includes a reactive group, which is a functional group or moiety that is capable of reacting with a reactive portion of another group, for instance, an antibody, modified antibody, or antigen binding fragment thereof. The moiety resulting from the reaction of the reactive group with the antibody, modified antibody, or antigen binding fragment thereof, together with the linking group, include the “binding agent linker” (“BL”) portion of the conjugate, described herein. In certain embodiments, the “reactive group” is a functional group or moiety (e.g., maleimide or A-hydroxysuccinimide (NHS) ester) that reacts with a cysteine or lysine residue of an antibody or antigen-binding fragment thereof. In some examples, the reactive group is a functional group, e.g., , which reacts with a cysteine residue on an antibody or antigen-binding fragment thereof, to form a carbon-sulfur bond thereto, e.g., , wherein Ab refers to an antibody or antigen-binding fragment thereof and S refers to the S atom on a cysteine residue through which the functional group bonds to the Ab. In some examples, the reactive group is a functional group, e.g., O , which reacts with a lysine residue on an antibody or antigen-binding fragment thereof, to form an amide bond thereto, e.g., , wherein Ab refers to an antibody or antigen-binding fragment thereof and NH refers to the NH atom on a lysine side chain residue through which the functional group bonds to the Ab. In some examples, the reactive group is a functional group, e.g., -NH2, which reacts with a lysine residue on an antibody or antigen-binding fragment thereof, to form an amino bond thereto, e.g., -NH-, wherein Ab refers to an antibody or antigen-binding fragment thereof and NH refers to the NH atom on a lysine side chain residue through which the functional group bonds to the Ab. In certain embodiments, the reaction is enzyme catalyzed. In certain embodiments, the reaction is catalyzed by transglutaminase.

[0050] As used herein, the phrase “biodegradable moiety” refers to a moiety that degrades in vivo to non-toxic, biocompatible components which can be cleared from the body by ordinary biological processes. In some embodiments, a biodegradable moiety completely or substantially degrades in vivo over the course of about 90 days or less, about 60 days or less, or about 30 days or less, where the extent of degradation is based on percent mass loss of the biodegradable moiety, and wherein complete degradation corresponds to 100% mass loss. Exemplary biodegradable moieties include, without limitation, aliphatic polyesters such as poly(s-caprolactone) (PCL), poly (3 -hydroxy butyrate) (PHB), poly(glycolic acid) (PGA), poly (lactic acid) (PLA) and its copolymers with glycolic acid (i.e., poly(D,L-lactide-coglycolide) (PLGA) (Vert M, Schwach G, Engel R and Coudane J (1998) J Control Release 53(1 -3): 85-92; Jain R A (2000) Biomaterials 21(23):2475-2490; Uhrich K E, Cannizzaro S M, Langer R S and Shakesheff K M (1999) Chemical Reviews 99(11): 3181-3198; and Park T G (1995) Biomaterials 16(15): 1123-1130, each of which are incorporated herein by reference in their entirety).

[0051] As used herein, the phrase “binding agent linker,” or “BL” refers to any divalent, trivalent, or multi-valent group or moiety that links, connects, or bonds a binding agent (e.g., an antibody or an antigen-binding fragment thereof) with a payload compound set forth herein (e.g., VX-787 and derivatives thereof) and, optionally, with one or more side chain compounds. Generally, suitable binding agent linkers for the antibody conjugates described herein are those that are sufficiently stable to exploit the circulating half-life of the antibody conjugates and, at the same time, capable of releasing its payload after antigen-mediated internalization of the conjugate. Linkers can be cleavable or non-cleavable. Cleavable linkers are linkers that are cleaved by intracellular metabolism following internalization, e.g., cleavage via hydrolysis, reduction, or enzymatic reaction. Non-cleavable linkers are linkers that release an attached payload via lysosomal degradation of the antibody following internalization. Suitable linkers include, but are not limited to, acid-labile linkers, hydrolytically-labile linkers, enzymatically cleavable linkers, reduction labile linkers, self-immolative linkers, and non-cleavable linkers. Suitable linkers also include, but are not limited to, those that are or comprise peptides, glucuronides, succinimide-thioethers, polyethylene glycol (PEG) units, hydrazones, mal- caproyl units, dipeptide units, vabne-citrulbne units, and para-aminobenzyloxycarbonyl (PABC), para-aminobenzyl (PAB) units. In some embodiments, the binding agent linker (BL) includes a moiety that is formed by the reaction of the reactive group (RG) of a reactive linker (RL) and reactive portion of the binding agent, e.g., antibody, modified antibody, or antigen binding fragment thereof.

1

[0052] In some examples, the BL includes the following moiety: , wherein is the bond to the cysteine of the antibody or antigen-binding fragment thereof. In some examples,

1 O the BL includes the following moiety: M * . wherein i « is the bond to the lysine of the antibody or antigen-binding fragment thereof.

[0053] In some embodiments, the binding agent is an antibody or an antigen-binding fragment thereof. The antibody can be in any form known to those of skill in the art.

[0054] The term “antibody,” as used herein, refers to any antigen-binding molecule or molecular complex comprising at least one complementarity determining region (CDR) that specifically binds to or interacts with a particular antigen. The term “antibody” includes immunoglobulin molecules comprising four polypeptide chains, two heavy (H) chains and two light (L) chains inter-connected by disulfide bonds (i.e., full antibody molecules), as well as multimers thereof (e.g., IgM) or antigen-binding fragments thereof. Each heavy chain comprises a heavy chain variable region (abbreviated herein as HCVR or VH) and a heavy chain constant region. The heavy chain constant region comprises three domains, CHI, CH2, and CH3. Each light chain comprises a light chain variable region (abbreviated herein as LCVR or VL) and a light chain constant region. The light chain constant region comprises one domain (CiT). The VH and VL regions can be further subdivided into regions of hypervariability, termed complementarity determining regions (CDRs), interspersed with regions that are more conserved, termed framework regions (FR). Each VH and VL is composed of three CDRs and four FRs, arranged from amino-terminus to carboxy -terminus in the following order: FR1, CDR1, FR2, CDR2, FR3, CDR3, FR4. In different embodiments disclosed herein, the FRs of the antibodies (or antigen-binding portion thereof) suitable for the compounds herein may be identical to the human germline sequences, or may be naturally or artificially modified. An amino acid consensus sequence may be defined based on a side-by-side analysis of two or more CDRs. The term “antibody,” as used herein, also includes antigen-binding fragments of full antibody molecules. The terms “antigen-binding domain” or “antigen-binding portion” of an antibody, “antigen-binding fragment” of an antibody, and the like, as used herein, include any naturally occurring, enzymatically obtainable, synthetic, or genetically engineered polypeptide or glycoprotein that specifically binds an antigen to form a complex. In certain embodiments, the term “antigen-binding fragment” refers to a polypeptide fragment of a multi-specific antigen-binding molecule. The terms “antigen-binding fragment” of an antibody, or “antibody fragment,” as used herein, refers to one or more fragments of an antibody that retain the ability to bind to an antigen such as influenza HA. Antigen-binding fragments of an antibody may be derived, e.g., from full antibody molecules using any suitable, standard technique(s) such as proteolytic digestion or recombinant genetic engineering technique(s) involving the manipulation and expression of DNA encoding antibody variable and optionally constant domains. Such DNA is known and/or is readily available from, e.g., commercial sources, DNA libraries (including, e.g., phage-antibody libraries), or can be synthesized. The DNA may be sequenced and manipulated chemically or by using molecular biology techniques, for example, to arrange one or more variable and/or constant domains into a suitable configuration, or to introduce codons, create cysteine residues, modify, add or delete amino acids, etc. Non- limiting examples of antigen-binding fragments include: (i) Fab fragments; (ii) F(ab')2 fragments; (iii) Fd fragments; (iv) Fv fragments; (v) single-chain Fv (scFv) molecules; (vi) dAb fragments; and (vii) minimal recognition units consisting of the amino acid residues that mimic the hypervariable region of an antibody (e.g. , a fragment containing a CDR, or an isolated CDR such as a CDR3 peptide), or a constrained FR3-CDR3-FR4 peptide. Other engineered molecules, such as domain-specific antibodies, single domain antibodies, domain-deleted antibodies, chimeric antibodies, CDR-grafted antibodies, diabodies, triabodies, tetrabodies, minibodies, nanobodies ( e.g monovalent nanobodies, bivalent nanobodies, etc.), small modular immunopharmaceuticals (SMIPs), and shark variable IgNAR domains, are also encompassed within the expression “antigen-binding fragment,” as used herein. An antigen binding fragment of an antibody will typically comprise at least one variable domain. The variable domain may be of any size or amino acid composition and will generally comprise at least one CDR which is adjacent to or in frame with one or more framework sequences. In antigen-binding fragments having a VH domain associated with a VL domain, the VH and VL domains may be situated relative to one another in any suitable arrangement. For example, the variable region may be dimeric and contain VH-VH, VH-VL, or VL-VL dimers. Alternatively, the antigen-binding fragment of an antibody may contain a monomeric VH or VL domain. In certain embodiments, an antigen-binding fragment of an antibody may contain at least one variable domain covalently linked to at least one constant domain. Non-limiting, exemplary configurations of variable and constant domains that may be found within an antigen-binding fragment of an antibody of this disclosure include: (i) VH-CH1; (ii) VH-CH2; (iii) VH-CH3; (iv) VH-CH1-CH2; (V) VH-CH1-CH2-CH3; (vi) VH-CH2-CH3; (vii) VH-CL; (viii) VL-CH1; (ix) VL- CH2; (X) VL-CH3; (xi) VL-CH1-CH2; (xii) VL-CH1-CH2-CH3; (xiii) VL-CH2-CH3; and (xiv) VL- CL. In any configuration of variable and constant domains, including any of the exemplary configurations listed above, the variable and constant domains may be either directly linked to one another or may be linked by a full or partial hinge or linker region. A hinge region may consist of at least 2 (e.g., 5, 10, 15, 20, 40, 60, or more) amino acids which result in a flexible or semi-flexible linkage between adjacent variable and/or constant domains in a single polypeptide molecule. Moreover, an antigen-binding fragment of an antibody herein may comprise a homo-dimer or hetero-dimer (or other multimer) of any of the variable and constant domain configurations listed above in non-covalent association with one another and/or with one or more monomeric VH or VL domain (e.g., by disulfide bond(s)). As with full antibody molecules, antigen-binding fragments may be monospecific or multispecific (e.g., bispecific). A multispecific antigen-binding fragment of an antibody will typically comprise at least two different variable domains, wherein each variable domain is capable of specifically binding to a separate antigen or to a different epitope on the same antigen. Any multispecific antibody format, including the exemplary bispecific antibody formats disclosed herein, may be adapted for use in the context of an antigen-binding fragment of an antibody of the present disclosure using routine techniques available in the art. In certain embodiments described herein, antibodies described herein are human antibodies.

[0055] Substitution of one or more CDR residues or omission of one or more CDRs is also possible. Antibodies have been described in the scientific literature in which one or two CDRs can be dispensed with for binding. Padlan et al. (1995 FASEB J. 9:133-139) analyzed the contact regions between antibodies and their antigens, based on published crystal structures, and concluded that only about one fifth to one third of CDR residues actually contact the antigen. Padlan et al. also found many antibodies in which one or two CDRs had no amino acids in contact with an antigen ( see also, Vajdos et al. 2002 J Mol Biol 320:415-428).

[0056] CDR residues not contacting antigen can be identified based on previous studies (for example, residues H60-H65 in CDRH2 are often not required), from regions of Rabat CDRs lying outside Chothia CDRs, by molecular modeling and/or empirically. If a CDR or residue(s) thereof is omitted, it is usually substituted with an amino acid occupying the corresponding position in another human antibody sequence or a consensus of such sequences. Positions for substitution within CDRs and amino acids to substitute can also be selected empirically. Empirical substitutions can be conservative or non-conservative substitutions.

[0057] The fully human anti-influenza-HA monoclonal antibodies disclosed herein may comprise one or more amino acid substitutions, insertions, and/or deletions in the framework and/or CDR regions of the heavy and light chain variable domains as compared to the corresponding germline sequences. Such mutations can be readily ascertained by comparing the amino acid sequences disclosed herein to germline sequences available from, for example, public antibody sequence databases. This disclosure includes antibodies, and antigen- binding fragments thereof, which are derived from any of the amino acid sequences disclosed herein, wherein one or more amino acids within one or more framework and/or CDR regions are mutated to the corresponding residue(s) of the germline sequence from which the antibody was derived, or to the corresponding residue(s) of another human germline sequence, or to a conservative amino acid substitution of the corresponding germline residue(s) (such sequence changes are referred to herein collectively as “germline mutations”). A person of ordinary skill in the art, starting with the heavy and light chain variable region sequences disclosed herein, can easily produce numerous antibodies and antigen-binding fragments which comprise one or more individual germline mutations or combinations thereof. In certain embodiments, all of the framework and/or CDR residues within the VH and/or VL domains are mutated back to the residues found in the original germline sequence from which the antibody was derived. In other embodiments, only certain residues are mutated back to the original germline sequence, e.g., only the mutated residues found within the first 8 amino acids of FR1 or within the last 8 amino acids of FR4, or only the mutated residues found within CDR1, CDR2, or CDR3. In other embodiments, one or more of the framework and/or CDR residue(s) are mutated to the corresponding residue(s) of a different germline sequence (i.e., a germline sequence that is different from the germline sequence from which the antibody was originally derived). Furthermore, the antibodies of this disclosure may contain any combination of two or more germline mutations within the framework and/or CDR regions, e.g., wherein certain individual residues are mutated to the corresponding residue of a particular germline sequence while certain other residues that differ from the original germline sequence are maintained or are mutated to the corresponding residue of a different germline sequence. Once obtained, antibodies and antigen-binding fragments that contain one or more germline mutations can be easily tested for one or more desired properties such as, improved binding specificity, increased binding affinity, improved or enhanced antagonistic or agonistic biological properties (as the case may be), reduced immunogenicity, etc. Antibodies and antigen-binding fragments obtained in this general manner are encompassed within this disclosure.

[0058] This disclosure also includes fully human anti-influenza-HA monoclonal antibodies comprising variants of any of the HCVR, LCVR, and/or CDR amino acid sequences disclosed herein having one or more conservative substitutions. For example, this disclosure includes anti-influenza-HA antibodies having HCVR, LCVR, and/or CDR amino acid sequences with, e.g., 10 or fewer, 8 or fewer, 6 or fewer, 4 or fewer, etc. conservative amino acid substitutions relative to any of the HCVR, LCVR, and/or CDR amino acid sequences disclosed herein.

[0059] The term “human antibody,” as used herein, is intended to include antibodies having variable and constant regions derived from human germline immunoglobulin sequences. The human mAbs of this disclosure may include amino acid residues not encoded by human germline immunoglobulin sequences (e.g., mutations introduced by random or site-specific mutagenesis in vitro or by somatic mutation in vivo), for example, in the CDRs and in particular CDR3. However, the term “human antibody,” as used herein, is not intended to include mAbs in which CDR sequences derived from the germline of another mammalian species (e.g., mouse) have been grafted onto human FR sequences. The term includes antibodies recombinantly produced in a non-human mammal, or in cells of a non-human mammal. The term is not intended to include antibodies isolated from or generated in a human subject. The term does not include naturally occurring antibodies that normally exist without modification or human intervention/manipulation, in a naturally occurring, unmodified living organism.

[0060] The term “recombinant,” as used herein, refers to antibodies or antigen-binding fragments thereof, created, expressed, isolated, or obtained by technologies or methods known in the art as recombinant DNA technology which include, e.g., DNA splicing and transgenic expression. The term refers to antibodies expressed in a non-human mammal (including transgenic non-human mammals (e.g., transgenic mice), or a cell (e.g., CHO cells) expression system, or isolated from a recombinant combinatorial human antibody library. The phrase “recombinant human antibody,” as used herein, is intended to include all human antibodies that are prepared, expressed, created, or isolated by recombinant means, such as antibodies expressed using a recombinant expression vector transfected into a host cell, antibodies isolated from a recombinant, combinatorial human antibody library, antibodies isolated from an animal (e.g., a mouse) that is transgenic for human immunoglobulin genes (see e.g., Taylor et al. (1992) Nucl. Acids Res. 20:6287-6295) or antibodies prepared, expressed, created, or isolated by any other means that involves splicing of human immunoglobulin gene sequences to other DNA sequences. Such recombinant human antibodies have variable and constant regions derived from human germline immunoglobulin sequences. In certain embodiments, however, such recombinant human antibodies are subjected to in vitro mutagenesis (or, when an animal transgenic for human Ig sequences is used, in vivo somatic mutagenesis) and thus the amino acid sequences of the VH and VL regions of the recombinant antibodies are sequences that, while derived from and related to human germline VH and VL sequences, may not naturally exist within the human antibody germline repertoire in vivo. Human antibodies can exist in two forms that are associated with hinge heterogeneity. In one form, an immunoglobulin molecule comprises a stable four chain construct of approximately 150-160 kDa in which the dimers are held together by an interchain heavy chain disulfide bond. In a second form, the dimers are not linked via inter-chain disulfide bonds and a molecule of about 75-80 kDa is formed composed of a covalently coupled light and heavy chain (half-antibody). These forms have been extremely difficult to separate, even after affinity purification. The frequency of appearance of the second form in various intact IgG isotypes is due to, but not limited to, structural differences associated with the hinge region isotype of the antibody. A single amino acid substitution in the hinge region of the human IgG4 hinge can significantly reduce the appearance of the second form (Angal et al. (1993) Molecular Immunology 30: 105) to levels typically observed using a human IgGl hinge. The instant disclosure encompasses antibodies having one or more mutations in the hinge, C H 2, or C H 3 region which may be desirable, for example, in production, to improve the yield of the desired antibody form.

[0061] An “isolated antibody,” as used herein, is intended to refer to an antibody that is substantially free of other antibodies (Abs) having different antigenic specificities (e.g., an isolated antibody that specifically binds influenza-HA, or a fragment thereof, is substantially free of Abs that specifically bind antigens other than influenza-HA). The antibodies described herein may be isolated antibodies. An “isolated antibody,” as used herein, further refers to an antibody that has been identified and separated and/or recovered from at least one component of its natural environment. For example, an antibody that has been separated or removed from at least one component of an organism, or from a tissue or cell in which the antibody naturally exists or is naturally produced, is an “isolated antibody” for purposes of the instant disclosure. An isolated antibody also includes an antibody in situ within a recombinant cell. Isolated antibodies are antibodies that have been subjected to at least one purification or isolation step. According to certain embodiments, an isolated antibody may be substantially free of other cellular material and/or chemicals. The antibodies used herein can comprise one or more amino acid substitutions, insertions, and/or deletions in the framework and/or CDR regions of the heavy and light chain variable domains as compared to the corresponding germline sequences from which the antibodies were derived. Such mutations can be readily ascertained by comparing the amino acid sequences disclosed herein to germline sequences available from, for example, public antibody sequence databases. The present disclosure includes antibodies, and antigen-binding fragments thereof, which are derived from any of the amino acid sequences disclosed herein, wherein one or more amino acids within one or more framework and/or CDR regions are mutated to the corresponding residue(s) of the germline sequence from which the antibody was derived, or to the corresponding residue(s) of another human germline sequence, or to a conservative amino acid substitution of the corresponding germline residue(s) (such sequence changes are referred to herein collectively as “germline mutations”). A person of ordinary skill in the art, starting with the heavy and light chain variable region sequences disclosed herein, can easily produce numerous antibodies and antigen-binding fragments which comprise one or more individual germline mutations or combinations thereof.

[0062] A “blocking antibody,” “neutralizing antibody,” or “antagonist antibody,” as used herein is intended to refer to an antibody whose binding to an antigen results in inhibition of at least one biological activity associated with the antigen. For example, an antibody or antibody- drug conjugate of this disclosure may prevent or block influenza attachment to, or entry into, a host cell. In addition, a “neutralizing antibody” is one that can neutralize, i.e., prevent, inhibit, reduce, impede, or interfere with, the ability of a pathogen to initiate and/or perpetuate an infection in a host. Such an antibody or antibody-drug conjugate, when possessing neutralizing ability via binding to influenza HA, can be referred to as an “antibody that neutralizes influenza-HA activity.” The terms “neutralizing antibody” and “an antibody that neutralizes” or “antibodies that neutralize” are used interchangeably herein. These antibodies can be used, alone or in combination, as prophylactic or therapeutic agents with other anti-viral agents upon appropriate formulation, or in association with active vaccination, or as a diagnostic tool. As used herein, an “anti-influenza antibody” can refer to an antibody whose binding to an antigen (e.g., HA) results in inhibition of at least one biological activity associated with influenza virus.

[0063] The term “epitope” refers to an antigenic determinant that interacts with a specific antigen-binding site in the variable region of an antibody molecule known as a paratope. A single antigen may have more than one epitope. Thus, different antibodies may bind to different areas on an antigen and may have different biological effects. The term “epitope” also refers to a site on an antigen to which B and/or T cells respond. It also refers to a region of an antigen that is bound by an antibody. B-cell epitopes can be formed both from contiguous amino acids or noncontiguous amino acids juxtaposed by tertiary folding of a protein. Epitopes formed from contiguous amino acids are typically retained on exposure to denaturing solvents, whereas epitopes formed by tertiary folding are typically lost on treatment with denaturing solvents. An epitope typically includes at least 3, and more usually, at least 5 or 8-10 amino acids in a unique spatial conformation. Epitopes may be defined as structural or functional. Functional epitopes are generally a subset of the structural epitopes and have those residues that directly contribute to the affinity of the interaction. Epitopes may also be conformational, that is, composed of non-linear amino acids. In certain embodiments, epitopes may include determinants that are chemically active surface groupings of molecules such as amino acids, sugar side chains, phosphoryl groups, or sulfonyl groups, and, in certain embodiments, may have specific three- dimensional structural characteristics, and/or specific charge characteristics.

[0064] The term “surface plasmon resonance,” refers to an optical phenomenon that allows for the analysis of real-time biomolecular interactions by detection of alterations in protein concentrations within a biosensor matrix, for example using the BIACORE™ system (Pharmacia Biosensor AB, Uppsala, Sweden and Piscataway, N.J).

[0065] Bio-layer interferometry is a label-free technology for measuring biomolecular interactions. It is an optical analytical technique that analyzes the interference pattern of white light reflected from two surfaces: a layer of immobilized protein on the biosensor tip, and an internal reference layer. Any change in the number of molecules bound to the biosensor tip causes a shift in the interference pattern that can be measured in real-time (Abdiche, Y.N., et al. Analytical Biochemistry, (2008), 377(2), 209-217). In certain embodiments, a “real-time bio-layer interferometer based biosensor (Octet HTX assay)” was used to assess the binding characteristics of certain of the anti -influenza HA antibodies.

[0066] The term “K D ,” as used herein, is intended to refer to the equilibrium dissociation constant of a particular antibody-antigen interaction.

[0067] The phrase “cross-competes,” as used herein, means an antibody or antigen-binding fragment thereof that binds to an antigen and inhibits or blocks the binding of another antibody or antigen-binding fragment thereof. The phrase also includes competition between two antibodies in both orientations, i.e., a first antibody that binds and blocks binding of second antibody and vice-versa. In certain embodiments, the first antibody and second antibody may bind to the same epitope. Alternatively, the first and second antibodies may bind to different, but overlapping epitopes such that binding of one inhibits or blocks the binding of the second antibody, e.g., by steric hindrance. Cross-competition between antibodies may be measured by methods known in the art, for example, by a real-time, label -free bio-layer interferometry assay. Cross-competition between two antibodies may be expressed as the binding of the second antibody that is less than the background signal due to self-self binding (wherein first and second antibodies are the same antibody). Cross-competition between two antibodies may be expressed, for example, as % binding of the second antibody that is less than the baseline self self background binding (wherein first and second antibodies are the same antibody).

[0068] The term “substantial identity” or “substantially identical,” when referring to a nucleic acid or fragment thereof, indicates that, when optimally aligned with appropriate nucleotide insertions or deletions with another nucleic acid (or its complementary strand), there is nucleotide sequence identity in at least about 90%, and more preferably at least about 95%, 96%, 97%, 98%, or 99% of the nucleotide bases, as measured by any well-known algorithm of sequence identity, such as FASTA, BLAST, or GAP, as discussed in WO 2016/100807 or US 2016/0176953 Al, each of which are incorporated herein by reference in their entirety. A nucleic acid molecule having substantial identity to a reference nucleic acid molecule may, in certain instances, encode a polypeptide having the same or substantially similar amino acid sequence as the polypeptide encoded by the reference nucleic acid molecule. [0069] As applied to polypeptides, the phrase “substantial similarity” or “substantially similar” means that two peptide sequences, when optimally aligned, such as by the programs GAP or BESTFIT using default gap weights, share at least 90% sequence identity, even more preferably at least 95%, 98%, or 99% sequence identity. Preferably, residue positions, which are not identical, differ by conservative amino acid substitutions. A “conservative amino acid substitution” is one in which an amino acid residue is substituted by another amino acid residue having a side chain (R group) with similar chemical properties (e.g., charge or hydrophobicity). In general, a conservative amino acid substitution will not substantially change the functional properties of a protein. In cases where two or more amino acid sequences differ from each other by conservative substitutions, the percent or degree of similarity may be adjusted upwards to correct for the conservative nature of the substitution. Means for making this adjustment are well known to those of skill in the art. (See, e.g., Pearson (1994) Methods Mol. Biol. 24: 307- 331 ). Examples of groups of amino acids that have side chains with similar chemical properties include 1) aliphatic side chains: glycine, alanine, valine, leucine and isoleucine; 2) aliphatic- hydroxyl side chains: serine and threonine; 3) amide-containing side chains: asparagine and glutamine; 4) aromatic side chains: phenylalanine, tyrosine, and tryptophan; 5) basic side chains: lysine, arginine, and histidine; 6) acidic side chains: aspartate and glutamate; and 7) sulfur-containing side chains: cysteine and methionine. Preferred conservative amino acids substitution groups are: valine-leucine-isoleucine, phenylalanine-tyrosine, lysine-arginine, alanine-valine, glutamate-aspartate, and asparagine-glutamine. Alternatively, a conservative replacement is any change having a positive value in the PAM250 log-likelihood matrix disclosed in Gonnet et al. (1992) Science 256: 1443 45. A “moderately conservative” replacement is any change having a nonnegative value in the PAM250 log-likelihood matrix.

[0070] Sequence similarity for polypeptides is typically measured using sequence analysis software. Protein analysis software matches similar sequences using measures of similarity assigned to various substitutions, deletions, and other modifications, including conservative amino acid substitutions. For instance, GCG software contains programs such as GAP and BESTFIT which can be used with default parameters to determine sequence homology or sequence identity between closely related polypeptides, such as homologous polypeptides from different species of organisms or between a wild type protein and a mutant thereof. See, e.g., GCG Version 6.1. Polypeptide sequences also can be compared using FASTA with default or recommended parameters; a program in GCG Version 6.1. FASTA (e.g., FASTA2 and FASTA3) provides alignments and percent sequence identity of the regions of the best overlap between the query and search sequences (Pearson (2000) supra). Sequences also can be compared using the Smith-Waterman homology search algorithm using an affine gap search with a gap open penalty of 12 and a gap extension penalty of 2, and BLOSUM matrix of 62. Another preferred algorithm when comparing a sequence of the invention to a database containing a large number of sequences from different organisms is the computer program BLAST, especially BLASTP or TBLASTN, using default parameters. See, e.g., Altschul etal. (1990) J. Mol. Biol. 215: 403-410 and (1997) Nucleic Acids Res. 25:3389-3402.

[0071] The phrase “therapeutically effective amount” refers to an amount that produces the desired effect for which it is administered. The exact amount will depend on the purpose of the treatment, and will be ascertainable by one skilled in the art using known techniques (see, for example, Lloyd (1999) The Art, Science and Technology of Pharmaceutical Compounding).

[0072] As used herein, the term “subject” refers to an animal, preferably a mammal, more preferably a human, in need of amelioration, prevention, and/or treatment of a disease or disorder such as a viral infection. The subject may have an influenza infection or is predisposed to developing an influenza virus infection. Subjects “predisposed to developing an influenza virus infection,” or subjects “who may be at elevated risk for contracting an influenza virus infection,” are those subjects with compromised immune systems because of autoimmune disease, those persons receiving immunosuppressive therapy (e.g., following organ transplant), those persons afflicted with human immunodeficiency syndrome (HIV) or acquired immune deficiency syndrome (AIDS), certain forms of anemia that deplete or destroy white blood cells, those persons receiving radiation or chemotherapy, or those persons afflicted with an inflammatory disorder. Additionally, subject of extreme young or old age are at increased risk. Any person who comes into physical contact or close physical proximity with an infected individual has an increased risk of developing an Influenza virus infection. Moreover, a subject is at risk of contracting an influenza infection due to proximity to an outbreak of the disease, e.g., subject resides in a densely-populated city or in close proximity to subjects having confirmed or suspected infections of Influenza virus, or choice of employment, e.g., hospital worker, pharmaceutical researcher, traveler to infected area, or frequent flier.

[0073] As used herein, the terms “treat,” “treating,” or “treatment” refer to the reduction or amelioration of the severity of at least one symptom or indication of influenza infection due to the administration of a therapeutic agent such as a disclosed antibody to a subject in need thereof. The terms include inhibition of progression of disease or of worsening of infection. The terms also include positive prognosis of disease, i.e., the subject may be free of infection or may have reduced or no viral titers upon administration of a therapeutic agent such as a disclosed antibody or antibody-drug conjugate. The therapeutic agent may be administered at a therapeutic dose to the subject.

[0074] The terms “prevent,” “preventing,” or “prevention” refer to inhibition of manifestation of influenza infection or any symptoms or indications of influenza infection upon administration of a disclosed antibody or antibody-drug conjugate. The term includes prevention of the spread of infection in a subject exposed to the virus or at risk of having influenza infection.

[0075] As used herein a “protective effect” may be demonstrated by any standard procedure known in the art to determine whether an agent such as an anti-viral agent, or an antibody such as an anti-influenza-HA antibody, or an antibody-drug conjugate disclosed herein can demonstrate any one or more of the following: e.g., an increase in survival after exposure to an infectious agent, a decrease in viral load, or amelioration of at least one symptom associated with the infectious agent.

[0076] As used herein, the phrases “antiviral drug” “anti-viral,” “antiviral compound,” and “anti-viral compound” apply to an anti-infective drug or therapy used to treat, prevent, or ameliorate a viral infection (e.g., influenza infection) in a subject. The term “anti-viral drug” (or its synonyms “antiviral drug,” “anti-viral compound,” and “antiviral compound”) includes, but is not limited to, TAMIFLU® (Oseltamivir), RELENZA® (Zanamivir), ribavirin, or interferon-alpha2b. Anti-viral drugs include influenza inhibitors. As used herein, an “influenza inhibitor” refers to a drug used to inhibit influenza virus infection, and includes, but is not limited to, oseltamivir. As used herein, a polymerase inhibitor can refer to an inhibitor of a nucleic acid polymerase, such as influenza polymerase. An exemplary polymerase inhibitor is VX-787. Without wishing to be bound by any particular theory, influenza inhibitors can function by targeting the influenza virus itself or by targeting a host cell that may be targeted by an influenza virus. For example, an influenza inhibitor that targets a host cell may inhibit translation in the cell, thereby reducing viral replication.

[0077] The phrase “specifically binds,” or “binds specifically to,” or the like, means that an antibody or antigen-binding fragment thereof forms a complex with an antigen that is relatively stable under physiologic conditions. Specific binding can be characterized by an equilibrium dissociation constant of at least about lxlO 8 M or less (e.g., a smaller KD denotes tighter binding). Methods for determining whether two molecules specifically bind are well known in the art and include, for example, equilibrium dialysis, surface plasmon resonance, and the like. As described herein, antibodies have been identified by real-time, label free biolayer interferometry assay on an Octet® HTX biosensor, which bind specifically to influenza- HA. Moreover, multi-specific antibodies that bind to one domain in influenza-HA and one or more additional antigens or a bi-specific that binds to two different regions of influenza-HA are nonetheless considered antibodies that “specifically bind”, as used herein. In addition to neutralizing antibodies, antibodies that bind specifically to HA, but are non-neutralizing, also can be used within the scope of the present disclosure to generate antibody-drug conjugates. Such antibodies may function, for example, to deliver a payload to influenza-infected cells.

[0078] The term “high affinity” antibody refers to those mAbs having a binding affinity to influenza-HA, expressed as K D , of at least 10 '8 M; preferably 10 '9 M; more preferably 10 '10 M, even more preferably 10 '11 M, even more preferably 10 '12 M, as measured by real-time, label free bio-layer interferometry assay, e.g., an Octet® HTX biosensor, or by surface plasmon resonance, e.g., BIACORE™, or by solution-affinity ELISA.

[0079] The phrase or term “slow off rate,” “K off ,” or “k d ” refers to an antibody that dissociates from influenza-HA, with a rate constant of lxl 0 '3 s '1 or less, preferably lxl O '4 s "1 or less, as determined by real-time, label free bio-layer interferometry assay, e.g., an Octet® HTX biosensor, or by surface plasmon resonance, e.g., BIACORE™.

[0080] The phrase “antigen-binding domain” or “antigen-binding portion” of an antibody, “antigen-binding fragment” of an antibody, and the like, as used herein, include any naturally occurring, enzymatically obtainable, synthetic, or genetically engineered polypeptide or glycoprotein that specifically binds an antigen to form a complex.

[0081] In specific embodiments, antibody or antibody fragments of this disclosure may be conjugated to a moiety such a ligand or a therapeutic moiety (“antibody-drug conjugate” or “immunoconjugate”), such as an anti-viral drug, a linker-payload including an anti-viral drug, a second anti-influenza antibody, or any other therapeutic moiety useful for treating an infection caused by influenza-HA.

[0082] As used herein, “sequentially administering” means that each dose of the compound is administered to the subject at a different point in time, e.g., on different days separated by a predetermined interval (e.g., hours, days, weeks, or months).

[0083] The phrases “initial dose,” “secondary doses,” and “tertiary doses,” refer to the temporal sequence of administration of the compounds described herein. Thus, the “initial dose” is the dose which is administered at the beginning of the treatment regimen (also referred to as the “baseline dose”); the “secondary doses” are the doses which are administered after the initial dose; and the “tertiary doses” are the doses which are administered after the secondary doses. The initial, secondary, and tertiary doses can all include the same amount the compound described herein, but generally can differ from one another in terms of frequency of administration.

[0084] The phrase “the immediately preceding dose,” as used herein, means, in a sequence of multiple administrations, the dose of the compound which is administered to a patient prior to the administration of the very next dose in the sequence with no intervening doses.

[0085] As used herein, the term “payload” refers to a small molecule active ingredient (e.g., an antiviral compound), optionally conjugated to an antibody or antigen-binding fragment thereof, directly or via a linker, that provides a desired biological effect (e.g., inhibiting influenza virus infection or replication). A payload can be less than or equal to 2,000 Da, less than or equal to 1,500 Da, or less than or equal to 900 Da.

Compounds or Payloads

[0086] Provided herein are antiviral compounds or payloads. Without being bound by any particular theory of operation, the antiviral compounds include VX-787 and derivatives thereof. In certain embodiments, the antiviral compounds can be delivered to cells as part of a conjugate. In certain embodiments, the antiviral compounds are capable of carrying out any activity of VX-787 and each of their derivatives at or in a target, for instance, a target cell. Certain antiviral compounds can have one or more additional activities.

[0087] In certain embodiments, set forth herein is a compound having the structure of Formula 301: or a pharmaceutically acceptable salt thereof. In Formula 301, R 1 is F and R 2 is H; or R 1 and R 2 cyclize to form a fused five-membered heteroaryl ring, optionally substituted with methyl; R 3 is H or HO-CH2-; Cy is bridged 5 or 6 membered cycloalkyl, wherein the bridge is methylene or ethylene; and Q is -O- or -0-NH-; wherein when R 3 is H, then Q is -0-NH-. In certain embodiments, R 3 is HO-CH2-. In certain embodiments, R 3 is H and Q is -0-NH-. In certain embodiments, R 1 is F and R 2 is H. In certain embodiments, R 1 and R 2 cyclize as - C=CH-S-. In certain embodiments, R 1 and R 2 cyclize as -C=CH-NMe-. In certain embodiments, Cy is bridged 6-membered cycloalkyl. In certain embodiments, Cy is . In certain embodiments, Cy is . In certain embodiments, R 3 is H; Q is

-0-NH-; R 1 is F; and R 2 is H. In certain embodiments, R 3 is H; Q is -0-NH-; R 1 is F; R 2 is cyclize

[0088] In certain embodiments, set forth herein is a compound having the structure of Formula 302: or a pharmaceutically acceptable salt thereof. In Formula 302, R 1 is F and R 2 is H; or R 1 and R 2 cyclize to form a fused five-membered heteroaryl ring, optionally substituted with methyl; R 3 is H or HO-CH2-; and Q is -O- or -0-NH-; wherein when R 3 is H, then Q is -0-NH-. In certain embodiments, R 3 is HO-CH2-. In certain embodiments, R 3 is H and Q is -0-NH-. In certain embodiments, R 1 is F and R 2 is H. In certain embodiments, R 1 and R 2 cyclize as - C=CH-S-. In certain embodiments, R 1 and R 2 cyclize as -C=CH-NMe-. In certain embodiments, R 3 is H; Q is -0-NH-; R 1 is F; and R 2 is H. In certain embodiments, R 3 is H;

Q is -0-NH-; and R 1 and R 2 cyclize as -C=CH-S-. In certain embodiments, R 3 is H; Q is -O- NH-; and R 1 and R 2 cyclize as -C=CH-NMe-.

[0089] In certain embodiments, set forth herein is a compound having the structure of Formula 303: or a pharmaceutically acceptable salt thereof. In Formula 303, R 1 is F and R 2 is H; or R 1 and R 2 cyclize to form a fused five-membered heteroaryl ring, optionally substituted with methyl; and Q is -0-NH-. In certain embodiments, R 1 is F and R 2 is H. In certain embodiments, Q is -0-NH-; R 1 is F; and R 2 is H.

[0090] In certain embodiments, set forth herein is a compound having the structure of Formula 304: or a pharmaceutically acceptable salt thereof. In Formula 304, R 1 is F and R 2 is H; or R 1 and R 2 cyclize to form a fused five-membered heteroaryl ring, optionally substituted with methyl; and Q is -0-NH-. In certain embodiments, R 1 is F and R 2 is H. In certain embodiments, Q is -0-NH-; R 1 is F; and R 2 is H.

[0091] In certain embodiments, set forth herein is a compound having the structure of Formula 305: or a pharmaceutically acceptable salt thereof. In Formula 305, R 1 is F and R 2 is H; or R 1 and R 2 cyclize to form a fused five-membered heteroaryl ring, optionally substituted with methyl. In certain embodiments, R 1 is F and R 2 is H. In certain embodiments, R 1 and R 2 cyclize as - C=CH-S-. In certain embodiments, R 1 and R 2 cyclize as -C=CH-NMe-.

[0092] In certain embodiments, set forth herein is a compound having the structure of Formula 306: or a pharmaceutically acceptable salt thereof. In Formula 306, R 1 is F and R 2 is H; or R 1 and R 2 cyclize to form a fused five-membered heteroaryl ring, optionally substituted with methyl. In certain embodiments, R 1 is F and R 2 is H. In certain embodiments, R 1 and R 2 cyclize as - C=CH-S-. In certain embodiments, R 1 and R 2 cyclize as -C=CH-NMe-.

[0093] In certain embodiments, set forth herein is a compound having the structure of Formula 311: or a pharmaceutically acceptable salt thereof. In Formula 311, R 1 is F and R 2 is H; or R 1 and R 2 cyclize to form a fused five-membered heteroaryl ring, optionally substituted with methyl; R 3 is H or HO-CH2-; Cy is bridged 5 or 6 membered cycloalkyl, wherein the bridge is methylene or ethylene; and Q is -O- or -0-NH-. In certain embodiments, when R 3 is H, then Q is -0-NH-. In certain embodiments, R 3 is HO-CH2-. In certain embodiments, R 3 is H and Q is -0-NH-. In certain embodiments, R 1 is F and R 2 is H. In certain embodiments, R 1 and R 2 cyclize as -C=CH-S-. In certain embodiments, R 1 and R 2 cyclize as -C=CH-NMe-. In certain embodiments, Cy is bridged 6-membered cycloalkyl. In certain embodiments, Cy is . In certain embodiments, Cy is . In certain embodiments, R 3 is H; Q is

-0-NH-; R 1 is F; and R 2 is H. In certain embodiments, R 3 is H; Q is -0-NH-; R 1 is F; R 2 is cyclize

[0094] In certain embodiments, set forth herein is a compound having the structure of Formula 312: or a pharmaceutically acceptable salt thereof. In Formula 312, R 1 is F and R 2 is H; or R 1 and R 2 cyclize to form a fused five-membered heteroaryl ring, optionally substituted with methyl; R 3 is H or HO-CH2-; and Q is -O- or -0-NH-. In certain embodiments, when R 3 is H, then Q is -0-NH-. In certain embodiments, R 3 is HO-CH2-. In certain embodiments, R 3 is H and Q is -0-NH-. In certain embodiments, R 1 is F and R 2 is H. In certain embodiments, R 1 and R 2 cyclize as -C=CH-S-. In certain embodiments, R 1 and R 2 cyclize as -C=CH-NMe-. In certain embodiments, R 3 is H; Q is -0-NH-; R 1 is F; and R 2 is H. In certain embodiments, R 3 is H;

Q is -0-NH-; and R 1 and R 2 cyclize as -C=CH-S-. In certain embodiments, R 3 is H; Q is -O- NH-; and R 1 and R 2 cyclize as -C=CH-NMe-.

[0095] In certain embodiments, set forth herein is a compound having the structure of Formula 313: or a pharmaceutically acceptable salt thereof. In Formula 313, R 1 is F and R 2 is H; or R 1 and R 2 cyclize to form a fused five-membered heteroaryl ring, optionally substituted with methyl; and Q is -0-NH-. In certain embodiments, R 1 is F and R 2 is H. In certain embodiments, Q is -0-NH-; R 1 is F; and R 2 is H.

[0096] In certain embodiments, set forth herein is a compound having the structure of Formula 314: or a pharmaceutically acceptable salt thereof. In Formula 314, R 1 is F and R 2 is H; or R 1 and R 2 cyclize to form a fused five-membered heteroaryl ring, optionally substituted with methyl; and Q is -0-NH-. In certain embodiments, R 1 is F and R 2 is H. In certain embodiments, Q is -0-NH-; R 1 is F; and R 2 is H.

[0097] In certain embodiments, set forth herein is a compound having the structure of Formula 315: or a pharmaceutically acceptable salt thereof. In Formula 315, R 1 is F and R 2 is H; or R 1 and R 2 cyclize to form a fused five-membered heteroaryl ring, optionally substituted with methyl. In certain embodiments, R 1 is F and R 2 is H. In certain embodiments, R 1 and R 2 cyclize as - C=CH-S-. In certain embodiments, R 1 and R 2 cyclize as -C=CH-NMe-.

[0098] In certain embodiments, set forth herein is a compound having the structure of Formula 316: or a pharmaceutically acceptable salt thereof. In Formula 306, R 1 is F and R 2 is H; or R 1 and R 2 cyclize to form a fused five-membered heteroaryl ring, optionally substituted with methyl. In certain embodiments, R 1 is F and R 2 is H. In certain embodiments, R 1 and R 2 cyclize as - C=CH-S-. In certain embodiments, R 1 and R 2 cyclize as -C=CH-NMe-.

[0099] In certain embodiments, set forth herein is a compound having the structure of Formula 321: or a pharmaceutically acceptable salt thereof. In Formula 321, R 1 is F and R 2 is H; or R 1 and R 2 cyclize to form a fused five-membered heteroaryl ring, optionally substituted with methyl; R 3 is H or HO-CH2-; Cy is bridged 5 or 6 membered cycloalkyl, wherein the bridge is methylene or ethylene; and Q is -O- or -0-NH-. In certain embodiments, R 3 is HO-CH2- and Q is -O-. In certain embodiments, R 3 is H and Q is -O-. In certain embodiments, R 1 is F and R 2 is H. In certain embodiments, R 1 and R 2 cyclize as -C=CH-S-. In certain embodiments, R 1 and R 2 cyclize as -C=CH-NMe-. In certain embodiments, R 3 is H; Q is -O-; R 1 is F; and R 2 is H. In certain embodiments, R 3 is H; Q is -O-; R 1 is F; and R 2 is H. In certain embodiments,

R 3 is H; Q is -O-; and R 1 and R 2 cyclize as -C=CH-S-. In certain embodiments, R 3 is H; Q is -O-; and R 1 and R 2 cyclize as -C=CH-NMe-.

[00100] In certain embodiments, set forth herein is a compound having the structure of Formula 322: or a pharmaceutically acceptable salt thereof. In Formula 322, R 1 is F and R 2 is H; or R 1 and R 2 cyclize to form a fused five-membered heteroaryl ring, optionally substituted with methyl; and Q is -0-NH-. In certain embodiments, R 1 is F and R 2 is H. In certain embodiments, Q is -0-NH-; R 1 is F; and R 2 is H. In certain embodiments, Q is -0-; R 1 is F; and R 2 is H.

[00101] In certain embodiments, set forth herein is a compound having the structure of Formula 323: or a pharmaceutically acceptable salt thereof. In Formula 323, R 1 is F and R 2 is H; or R 1 and R 2 cyclize to form a fused five-membered heteroaryl ring, optionally substituted with methyl. In certain embodiments, R 1 is F and R 2 is H. In certain embodiments, R 1 and R 2 cyclize as - C=CH-S-. In certain embodiments, R 1 and R 2 cyclize as -C=CH-NMe-. In certain embodiments, Q is -0-NH-; R 1 is F; and R 2 is H. In certain embodiments, Q is -0-; R 1 is F; and R 2 is H.

[00102] In certain embodiments, provided herein is a compound selected from the group consisting of:

20c

117

and pharmaceutically acceptable salts thereof.

Binding agents

[00103] Suitable binding agents for any of the conjugates provided in the instant disclosure include, but are not limited to, antibodies, viral receptors, or any other cell binding or peptide binding molecules or substances. The full-length amino acid sequence of an exemplary Influenza HA is shown in GenBank as accession number ACP44150.1.

[00104] Suitable binding agents include antibodies (e.g., fully human antibodies) and antigen-binding fragments thereof that specifically bind to influenza virus proteins, such as the surface proteins hemagglutinin (HA), neuraminidase (NA), and Matrix-2 (M2). In some embodiments, these binding agents modulate the interaction of influenza virus with host cells. In some embodiments, the antibodies or antigen-binding fragments thereof bind to mature hemagglutinin. In some embodiments, the antibodies or antigen-binding fragments thereof bind to an HA0 hemagglutinin precursor protein. The anti-influenza HA antibodies may bind to the influenza virus HA with high affinity. In certain embodiments, the antibodies herein are blocking antibodies wherein the antibodies may bind to influenza HA and block the attachment to and/or entry of the virus into host cells. In some embodiments, the blocking antibodies herein may block the binding of influenza virus to cells and as such may inhibit or neutralize viral infectivity of host cells. In some embodiments, the blocking antibodies may be useful for treating a subj ect suffering from an influenza virus infection. The antibodies when administered to a subject in need thereof may reduce the infection by a virus such as influenza in the subject. They may be used to decrease viral loads in a subject. They may be used alone or as adjunct therapy with other therapeutic moieties or modalities known in the art for treating a viral infection. In certain embodiments, these antibodies may bind to an epitope in the stem region of the viral HA, the head region of the viral HA, or both. Furthermore, the identified antibodies can be used prophylactically (before infection) to protect a mammal from infection, or can be used therapeutically (after infection is established) to ameliorate a previously established infection, or to ameliorate at least one symptom associated with the infection.

[00105] In certain embodiments, the antibodies are obtained from mice immunized with a primary immunogen, such as a full length influenza HA or with a recombinant form of influenza HA or fragments thereof followed by immunization with a secondary immunogen, or with an immunogenically active fragment of influenza HA. In certain embodiments, the antibodies are obtained from mice immunized with an influenza vaccine composition followed by booster immunization with one or more recombinantly produced HA peptides. In certain embodiments, the antibodies are obtained from humans. In certain embodiments, the antibodies are obtained from mammals (e.g., non-human mammals). In certain embodiments, the antibodies are obtained from non-human primates.

[00106] The immunogen may be a biologically active and/or immunogenic fragment of influenza HA or DNA encoding the active fragment thereof. The fragment may be derived from the stem region of the HA protein. (See, Sui el al. Nature Struct, and Mol. Biol. Published online 22 Feb. 2009; Pages 1 -9), the head region of the HA protein, or a combination thereof.

[00107] The peptides may be modified to include addition or substitution of certain residues for tagging or for purposes of conjugation to carrier molecules, such as, keyhole limpet hemocyanin (KLH). For example, a cysteine may be added at either the N-terminal or C- terminal end of a peptide, or a linker sequence may be added to prepare the peptide for conjugation to, for example, KLH for immunization.

[00108] Certain anti -influenza antibodies, anti-influenza-HA antibodies, or ADCs herein have antiviral activity, such as being able to bind to and neutralize the activity of influenza- HA, as determined by in vitro or in vivo assays. Certain anti-influenza antibodies, anti- influenza-HA antibodies, or ADCs herein are able to bind to HA but do not have neutralizing activity, as determined by in vitro or in vivo assays. The ability of the antibodies or ADCs herein to bind to and neutralize the activity of influenza-HA and thus the attachment and/or entry of the virus into a host cell followed by the ensuing viral infection, may be measured using any standard method known to those skilled in the art, including binding assays, or activity assays, as described herein.

[00109] The antibodies or ADCs specific for influenza-HA may contain no additional labels or moieties, or they may contain an N-terminal or C-terminal label or moiety. In one embodiment, the label or moiety is biotin. In a binding assay, the location of a label (if any) may determine the orientation of the peptide relative to the surface upon which the peptide is bound. For example, if a surface is coated with avidin, a peptide containing an N-terminal biotin will be oriented such that the C-terminal portion of the peptide will be distal to the surface. In one embodiment, the label may be a radionuclide, a fluorescent dye, or a MRI- detectable label. In certain embodiments, such labeled antibodies may be used in diagnostic assays including imaging assays.

[00110] In certain embodiments, the antibody comprises a light chain. In certain embodiments, the light chain is a kappa light chain. In certain embodiments, the light chain is a lambda light chain. In certain embodiments, the antibody comprises a heavy chain. In some embodiments, the heavy chain is an IgA. In some embodiments, the heavy chain is an IgD. In some embodiments, the heavy chain is an IgE. In some embodiments, the heavy chain is an IgG. In some embodiments, the heavy chain is an IgM. In some embodiments, the heavy chain is an IgGl . In some embodiments, the heavy chain is an IgG2. In some embodiments, the heavy chain is an IgG3. In some embodiments, the heavy chain is an IgG4. In some embodiments, the heavy chain is an IgAl. In some embodiments, the heavy chain is an IgA2.

[00111] In some embodiments, the antibody is an antibody fragment. In some embodiments, the antibody fragment is an Fv fragment. In some embodiments, the antibody fragment is a Fab fragment. In some embodiments, the antibody fragment is a F(ab')2 fragment. In some embodiments, the antibody fragment is a Fab' fragment. In some embodiments, the antibody fragment is an scFv (sFv) fragment. In some embodiments, the antibody fragment is an scFv- Fc fragment.

[00112] In some embodiments, the antibody is a monoclonal antibody. In some embodiments, the antibody is a polyclonal antibody. In some embodiments, the antibody is a bispecific antibody including a first antigen-binding domain, and a second antigen-binding domain.

[00113] In some embodiments, the antibody is a chimeric antibody. In some embodiments, the antibody is a humanized antibody. In some embodiments, the antibody is a human antibody.

[00114] In certain embodiments, the antibody comprises a glutamine residue at one or more heavy chain positions numbered 295 in the EU numbering system. In the present disclosure, this position is referred to as glutamine 295, or as Gln295, or as Q295. Those of skill will recognize that this is a conserved glutamine residue in the wild type sequence of many antibodies. Identification of residue Q295 can be accomplished readily with standard sequence alignment tools, including those described herein. In other useful embodiments, the antibody can be engineered to comprise a glutamine residue. In certain embodiments, the antibody comprises one or more N297Q mutations. Techniques for modifying an antibody sequence to include a glutamine residue are within the skill of those in the art (see, e.g., Ausubel el al. Current Protoc. Mol. Biol.). In one embodiment, the antibody includes an antibody heavy chain and further includes a peptide tag at the C-terminus of the antibody heavy chain. In one embodiment, the antibody includes an antibody heavy chain and further includes a peptide tag, e.g., transglutaminase recognition sequence or pentapeptide tag, at the C-terminus of the antibody heavy chain, wherein the peptide tag is the pentapeptide sequence LLQGA.

Preparation of Human Antibodies

[00115] Methods for generating human antibodies in transgenic mice are known in the art. Any such known methods can be used in the context of this disclosure to make human antibodies that specifically bind to Influenza-HA. An immunogen comprising any one of the following can be used to generate antibodies to Influenza HA. In certain embodiments, the antibodies herein are obtained from mice immunized with a full length, native influenza HA (See, e.g., GenBank accession number FJ966082.1), or with a live attenuated or inactivated virus, or with DNA encoding the protein or fragment thereof. Alternatively, the influenza-HA protein or a fragment thereof may be produced using standard biochemical techniques and modified and used as immunogen. In one embodiment, the immunogen is a recombinantly produced influenza-HA protein or fragment thereof. In certain embodiments herein, the immunogen may be an influenza virus vaccine. In certain embodiments, one or more booster injections may be administered. In certain embodiments, the booster injections may comprise one or more influenza virus strains, or hemagglutinins derived from these strains, e.g., see Protein Sciences HI A/New Caledonia/20/1999, H5 A/lndonesia/05/2005, H3 A/Victoria/361/2011, H7 A/Netherlands/219/2003, or H9 A/Hong Kong/1073/1988, or the influenza B virus strains B/Victoria/2/87, B/Nanchang/3451/93, B/Singapore/11/1994, B/Florida/4/2006, or B/Yamagata/ 16/88. In certain embodiments, the booster injections may contain a 1 : 1 mixture of the influenza strains, or a 1 : 1 mixture of the hemagglutinins derived from the strains. In certain embodiments, the immunogen may be a recombinant Influenza HA peptide expressed in E. coli or in any other eukaryotic or mammalian cells such as Chinese hamster ovary (CHO) cells or influenza virus itself. [00116] Using VELOCIMMUNE® technology (see, e.g., US 6,596,541, Regeneron Pharmaceuticals, VELOCIMMUNE®) or any other known method for generating monoclonal antibodies, high affinity chimeric antibodies to influenza-HA are initially isolated having a human variable region and a mouse constant region. The VELOCIMMUNE® technology involves generation of a transgenic mouse having a genome comprising human heavy and light chain variable regions operably linked to endogenous mouse constant region loci such that the mouse produces an antibody comprising a human variable region and a mouse constant region in response to antigenic stimulation. The DNA encoding the variable regions of the heavy and light chains of the antibody are isolated and operably linked to DNA encoding the human heavy and light chain constant regions. The DNA is then expressed in a cell capable of expressing the fully human antibody.

[00117] Generally, a VELOCIMMUNE® mouse is challenged with the antigen of interest, and lymphatic cells (such as B-cells) are recovered from the mice that express antibodies. The lymphatic cells may be fused with a myeloma cell line to prepare immortal hybridoma cell lines, and such hybridoma cell lines are screened and selected to identify hybridoma cell lines that produce antibodies specific to the antigen of interest. DNA encoding the variable regions of the heavy chain and light chain may be isolated and linked to desirable isotypic constant regions of the heavy chain and light chain. Such an antibody protein may be produced in a cell, such as a CHO cell. Alternatively, DNA encoding the antigen-specific chimeric antibodies or the variable domains of the light and heavy chains may be isolated directly from antigen- specific lymphocytes.

[00118] Initially, high affinity chimeric antibodies are isolated having a human variable region and a mouse constant region. As described in WO 2016/100807 or US 2016/0176953 Al, each of which are incorporated by reference in their entirety, the antibodies are characterized and selected for desirable characteristics, including affinity, selectivity, epitope, etc. The mouse constant regions are replaced with a desired human constant region to generate the fully human antibody herein, for example, wild-type or modified IgGl or IgG4. While the constant region selected may vary according to specific use, high affinity antigen-binding and target specificity characteristics reside in the variable region. Bioequivalents

[00119] The anti-influenza-HA antibodies and antibody fragments herein encompass proteins having amino acid sequences that vary from those of the described antibodies, but that retain the ability to bind Influenza HA. Such variant antibodies and antibody fragments comprise one or more additions, deletions, or substitutions of amino acids when compared to parent sequence, but exhibit biological activity that is essentially equivalent to that of the described antibodies. Likewise, the antibody-encoding DNA sequences of the present disclosure encompass sequences that comprise one or more additions, deletions, or substitutions of nucleotides when compared to the disclosed sequence, but that encode an antibody or antibody fragment that is essentially bioequivalent to an antibody or antibody fragment herein. Other bioequivalent anti-influenza-HA antibodies and antibody fragments are as described in WO 2016/100807 or US 2016/0176953 Al, each of which are incorporated by reference in their entirety.

Biological Characteristics of the Antibodies

[00120] In general, the antibodies herein function by binding to Influenza HA. For example, provided herein are antibodies and antigen-binding fragments of antibodies that bind Influenza HA (e.g., at 25 °C or at 37 °C) with a K D of less than 10 nM, as measured by real-time bio layer interferometer based biosensor (Octet HTX assay), or by surface plasmon resonance. In certain embodiments, the antibodies or antigen-binding fragments thereof bind influenza-HA with a KD of less than about 5 nM, less than about 2 nM, less than about 1 nM, less than about 500 pM, less than 250 pM, or less than 100 pM, as measured by surface plasmon resonance, e.g., using the assay format as described in WO 2016/100807 or US 2016/0176953 Al, each of which are incorporated by reference in their entirety, or a substantially similar assay.

[00121] Non-limiting, exemplary in vitro assays for measuring binding activity are illustrated in Example 3 of WO 2016/100807 or US 2016/0176953 Al, each of which is incorporated herein by reference in their entirety. In WO 2016/100807 or US 2016/0176953 Al Example 3, the binding affinity and dissociation constants of anti-influenza-HA antibodies for influenza-HA were determined by real-time bio-layer interferometer based biosensor (Octet HTX assay). In Examples 4 and 5 of WO 2016/100807 or US 2016/0176953 Al, neutralization assays were used to determine infectivity of diverse group 1 strains of influenza virus. In Example 6 of WO 2016/100807 or US 2016/0176953 Al, certain antibodies were shown to mediate complement dependent cytotoxicity (CDC) of virus-infected cells in vitro. Examples 7 and 10 of WO 2016/100807 or US 2016/0176953 A1 demonstrate that certain antibodies of the disclosure are capable of neutralizing an influenza A infection in vivo when administered either prophylactically or therapeutically.

[00122] Also provided herein are antibodies and antigen-binding fragments thereof that bind Influenza HA with a dissociative half-life (t½) of greater than about 100 minutes as measured by surface plasmon resonance at 25 °C, e.g., using an assay format as defined in WO 2016/100807 or US 2016/0176953 Al, each of which are incorporated herein by reference in their entirety, or a substantially similar assay. In certain embodiments, the antibodies or antigen-binding fragments herein bind Influenza HA with a t½ of greater than about 200 minutes, greater than about 300 minutes, greater than about 400 minutes, greater than about 500 minutes, greater than about 600 minutes, greater than about 700 minutes, greater than about 800 minutes, greater than about 900 minutes, or greater than about 1000 minutes as measured by surface plasmon resonance at 25 °C, e.g., using an assay format as defined in WO 2016/100807 or US 2016/0176953 Al, each of which is incorporated herein by reference in their entirety (e.g., mAb-capture or antigen-capture format), or a substantially similar assay. In one embodiment, the antibodies and antigen-binding fragments herein bind Influenza HA with a dissociative half-life (t½) of greater than 300 minutes. In one embodiment, an antibody herein provides for about a 1.5 to 2-fold increase in dissociative half-life as compared to a comparator antibody designated Control I mAh, when tested in monkeys and mice.

[00123] Also provided herein are antibodies or antigen-binding fragments thereof that neutralize the infectivity of influenza virus for its host cells. In some embodiments, the antibodies exhibit a neutralization potency against various representative group 1 influenza viruses (H1N1 A/Puerto Rico/08/1934; H5N1 A/Vietnam/1203/2004; H1N1 A Califomia/07/2009; H1N1 A/Wisconsin/1933; H1N1 A/Brisbane/59/1997, H9N2 A Hong Kong/33982/2009, H13N6 a/gull/Maryland/704/1977 and H16N3

A/shorebird/Delaware/172/2006) with an IC50 ranging from about 1.6 nM to about 130 nM in a microneutralization assay, e.g., as shown in Examples 4 and 5 of WO 2016/100807 or US 2016/0176953 Al, each of which is incorporated herein by reference in their entirety, or a substantially similar assay. In one embodiment, the antibodies or antigen-binding fragments thereof that neutralize the infectivity of influenza virus for its host cells do so with an IC50 of less than 130 nM. [00124] Also provided herein are antibodies or antigen-binding fragments thereof that mediate complement dependent cytotoxicity of infected cells, with an EC 50 ranging from about 20 nM to about 66 nM (see example 6 in WO 2016/100807 or US 2016/0176953 Al, each of which is incorporated herein by reference in their entirety). In one embodiment, the antibodies or antigen-binding fragments thereof mediate complement-dependent cytotoxicity of infected cells, with an EC 50 less than 66 nM.

[00125] Described herein are anti-influenza-A HA antibodies that demonstrate an increase in protection, or neutralization of influenza A infection in vivo, as compared to a control antibody. Certain antibodies show neutralization when administered either prophylactically (prior to infection) or therapeutically (after infection); see example 7 in WO 2016/100807 or US 2016/0176953 Al, each of which is incorporated herein by reference in their entirety.

[00126] In one embodiment, provided herein is an isolated recombinant antibody or antigen binding fragment thereof that binds specifically to Influenza HA, wherein the antibody or fragment thereof exhibits two or more of the following characteristics: (a) is a fully human monoclonal antibody; (b) binds to influenza HA with a dissociation constant (KD) of less than 10 9 M, as measured in a surface plasmon resonance assay; (c) demonstrates a dissociative half- life (t½) ranging from about 370 minutes to greater than 1000 minutes; (d) demonstrates neutralization of group 1 influenza A viruses selected from H1N1, H5N1, H9N2, H13N6, and H16N3, with an IC50 ranging from about 1.6 nM to about 130 nM; (e) demonstrates complement mediated lysis of influenza virus infected cells with an EC50 of about 20 nM to about 66 nM; or (1) demonstrates protection, as measured by increased survival in an animal model of influenza virus infection when administered either before or after virus challenge.

[00127] The antibodies herein may possess two or more of the aforementioned biological characteristics, or any combinations thereof. Other biological characteristics of the antibodies herein will be evident to a person of ordinary skill in the art from a review of the present disclosure including the working Examples herein.

Heavy and Light Chain Variable Region Amino Acid and Nucleotide Sequences

[00128] In some embodiments, the antibody, or antigen-binding fragment thereof, conjugated to the linker-payload or payload can be an antibody that targets Influenza HA. Exemplary Influenza HA antibodies can be found, for example, in WO 2016/100807 or US 2016/0176953 Al, each of which are incorporated herein by reference in their entirety. In some embodiments, an Influenza HA antibody comprises a heavy chain complementarity determining region (HCDR)-1 comprising SEQ ID NO: 20; an HCDR2 comprising SEQ ID NO: 22; an HCDR3 comprising SEQ ID NO: 24; a light chain complementarity determining region (LCDR)-1 comprising SEQ ID NO: 28; an LCDR2 comprising SEQ ID NO: 30; and an LCDR3 comprising SEQ ID NO: 32. In some embodiments, an Influenza HA antibody comprises a heavy chain variable region (HCVR) comprising SEQ ID NO: 18 and a light chain variable region (LCVR) comprising SEQ ID NO: 26. In any of the foregoing embodiments, the Influenza HA antibody can be prepared by site-directed mutagenesis to insert a glutamine residue at a site without resulting in disabled antibody function or binding. For example, in any of the foregoing embodiments, the Influenza HA antibody can comprise an Asn297Gln (N297Q) mutation. Such antibodies having an N297Q mutation can also contain one or more additional naturally occurring glutamine residues in their variable regions, which can be accessible to transglutaminase and therefore capable of conjugation to a payload or a linker- payload. In one embodiment, the antibody includes a HCVR and further includes a peptide tag at the C-terminus of the HCVR. In one embodiment, the antibody includes a HCVR and further includes a peptide tag at the C-terminus of the HCVR, wherein the peptide tag is the pentapeptide sequence LLQGA. In one embodiment, the antibody includes two HCVRs and further includes a peptide tag at the C-terminus of each HCVR. In one embodiment, the antibody includes two HCVRs and further includes a peptide tag at the C-terminus of the HCVRs, wherein the peptide tag is the pentapeptide sequence LLQGA.

[00129] Table 1 sets forth the amino acid sequence identifiers of the heavy and light chain variable regions and CDRs of selected anti-influenza HA antibodies. The corresponding nucleic acid sequence identifiers are set forth in Table 2.

Table 1: Amino Acid Sequence Identifiers

* mAb contains one or more mutations in the constant region

Table 2: Nucleic Acid Sequence Identifiers

* mAb contains one or more mutations in the constant region.

[00130] The binding agent linkers can be bonded to the binding agent, e.g., antibody or antigen-binding molecule, through an attachment at a particular amino acid within the antibody or antigen-binding molecule. Exemplary amino acid attachments that can be used in the context of this embodiment of the disclosure include, e.g., lysine (see, e.g., US 5,208,020; US 2010/0129314; Hollander et ciL, Bioconjugate Chem., 2008, 19:358-361; WO 2005/089808; US 5,714,586; US 2013/0101546; and US 2012/0585592), cysteine (see, e.g., US 2007/0258987; WO 2013/055993; WO 2013/055990; WO 2013/053873; WO 2013/053872; WO 2011/130598; US 2013/0101546; and US 7,750,116), selenocysteine (see, e.g, WO 2008/122039; and Hofer et al, Proc. Natl. Acad. ScL, USA, 2008, 705:12451-12456), formyl glycine (see, e.g., Carrico et al.,Nat. Chem. Biol., 2007, 3:321-322; Agarwal etal., Proc. Natl. Acad. Sci., USA, 2013, 770:46-51, and Rabuka et al., Nat. Protocols, 2012, 70:1052-1067), non-natural amino acids (see, e.g., WO 2013/068874, and WO 2012/166559), and acidic amino acids (see, e.g. , WO 2012/05982). Linkers can also be conjugated to an antigen-binding protein via attachment to carbohydrates (see, e.g, US 2008/0305497, WO 2014/065661, Ryan et al., Food cS Agriculture Immunol., 2001, 73:127-130, and Jeger et al.,Angew Chem Int Ed Engl., 2010, 49\ 9995-9997).

[00131] In some examples, the binding agent is an antibody or antigen binding molecule, and the antibody is bonded to the linker through a lysine residue. In some embodiments, the antibody or antigen binding molecule is bonded to the linker through a cysteine residue, lysine residue, or glutamine residue. In certain embodiments, the antibody or antigen binding molecule is bonded to the linker through a cysteine residue. In certain embodiments, a linker maleimide moiety bonds to an antibody cysteine residue. In certain embodiments, the antibody or antigen binding molecule is bonded to the linker through a lysine residue. In certain embodiments, a linker N-hydroxysuccinimide moiety bonds to an antibody lysine residue to form an amide linkage.

[00132] . In certain embodiments, the antibody or antigen binding molecule is bonded to the linker through a glutamine residue (see, e.g., Jeger et al, Angew Chem Int Ed Engl., 2010, 49\ 9995-9997 and Dennler et al, Bioconjugate Chem. 2014, 25: 569-578). Antibodies comprising glutamine residues can be isolated from natural sources or engineered to comprise one or more glutamine residues. In certain embodiments, antibodies or antigen binding molecules are engineered by mutations, for example insertions or deletions to facilitate reaction via transglutaminase. In certain embodiments, antibodies or antigen binding molecules are engineered to remove one or more glycosylation sites. In certain embodiments, antibodies or antigen binding molecules are engineered to add one or more glutamine residues. In certain embodiments, glutamine residues are added within a TGase recognition tag, as described herein. Techniques for engineering glutamine residues into an antibody polypeptide chain (glutaminyl-modified antibodies or antigen binding molecules) are within the skill of the practitioners in the art. In certain embodiments, the antibody is aglycosylated.

[00133] In certain embodiments, the antibody or a glutaminyl-modified antibody or antigen binding molecule comprises at least one glutamine residue in at least one polypeptide chain sequence. In certain embodiments, the antibody or a glutaminyl-modified antibody or antigen binding molecule comprises two heavy chain polypeptides, each with one Gln295 or Q295 residue. In further embodiments, the antibody or a glutaminyl-modified antibody or antigen binding molecule comprises one or more glutamine residues at a site other than a heavy chain 295. Included herein are antibodies of this section bearing N297Q mutation(s) described herein. In certain embodiments, a glutamine residue is added at the heavy chain C-terminus.

[00134] In certain embodiments, the glutamine is polypeptide engineered with a glutamine- containing tag (e.g., glutamine-containing peptide tags, Q-tags or TGase recognition tag). The term “TGase recognition tag” or “Q-Tag” refers to a sequence of amino acids comprising a glutamine residue that when incorporated into (e.g. appended to) a polypeptide sequence, under suitable conditions, is recognized by a transglutaminase (“TGase”) and leads to cross-linking by the TGase through a reaction between an amino acid side chain within the sequence of amino acids and a reactive group. The recognition tag may be a peptide sequence that is not naturally present in the polypeptide. In certain embodiments, the TGase recognition tag comprises at least one glutamine. In certain embodiments, the TGase recognition tag comprises an amino acid sequence XXQX, wherein X is any amino acid (e.g., conventional amino acid Leu, Ala, Gly, Ser, Val, Phe, Tyr, His, Arg, Asn, Glu, Asp, Cys, Gin, He, Met, Pro, Thr, Lys, or Trp or nonconventional amino acid). In certain embodiments, the TGase recognition tag comprises an amino acid sequence selected from the group consisting of LLQGG, LLQG, LSLSQG, GGGLLQGG, GLLQG, LLQ, GSPLAQSHGG, GLLQGGG, GLLQGG, GLLQ, LLQLLQGA, LLQGA, LLQYQGA, LLQGSG, LLQYQG, LLQLLQG, SLLQG, LLQLQ, LLQLLQ, and LLQGR. See for example, WO2012059882, the entire contents of which are incorporated herein.

[00135] In one embodiment, the antibody or antigen binding molecule includes an antibody heavy chain and further includes a TGase recognition tag at the C-terminus of the antibody heavy chain. In one embodiment, the antibody or antigen binding molecule includes an antibody heavy chain and further includes a TGase recognition tag at the C-terminus of the antibody heavy chain, wherein the TGase recognition tag is the pentapeptide sequence LLQGA. In one embodiment, the antibody or antigen binding molecule includes two antibody heavy chains and further includes a TGase recognition tag at the C-terminus of each antibody heavy chain. In one embodiment, the antibody or antigen binding molecule includes two antibody heavy chains and further includes a TGase recognition tag at the C-terminus of each antibody heavy chain, wherein the TGase recognition tag is the pentapeptide sequence LLQGA. [00136] Antibodies, or antigen binding molecules, can also be modified at one or more glutamine residues via transglutaminase (see, e.g., Jeger et al.,Angew Chemlnt Ed Engl., 2010, 49: 9995-9997 and Dennler et al, Bioconjugate Chem. 2014, 25: 569-578). For example, in the presence of transglutaminase, one or more glutamine residues of an antibody can be coupled to a primary amine compound to provide a moiety capable of reacting with a reactive group on a linker-payload. In certain embodiments, the primary amine compound provides a diene or dienophile. In certain embodiments, the primary amine compound provides a diene or dienophile, and the linker-payload provides a complementary dienophile or diene, respectively, for conjugation via a Diels-Alder reaction. In certain embodiments, the primary amine compound provides an azido group. In certain embodiments, the primary amine compound provides an azido group and the linker-payload provides a complementary alkyne, for conjugation via a click reaction.

Linkers

[00137] In certain embodiments, the linker L portion of the conjugates described herein is a moiety, for instance a divalent moiety, that covalently links a binding agent to a payload compound described herein. In other instances, the linker L is a trivalent or multivalent moiety that covalently links a binding agent to a payload compound described herein. Suitable linkers may be found, for example, in Antibody-Drug Conjugates and Immunotoxins Phillips, G. L., Ed.; Springer Verlag: New York, 2013; Antibody-Drug Conjugates, Ducry, L., Ed.; Humana Press, 2013; Antibody-Drug Conjugates, Wang, I, Shen, W.-C., and Zaro, J. L., Eds.; Springer International Publishing, 2015, the contents of each incorporated herein in their entirety by reference. In certain embodiments, the linker L portion of the linker-payloads described herein is a moiety covalently linked to a payload compound described herein, capable of divalently and covalently linking a binding agent to a payload compound described herein. In other instances, the linker L portion of the linker-payloads described herein is a moiety covalently linked to a payload compound described herein, capable of covalently linking, as a trivalent or multivalent moiety, a binding agent to a payload compound described herein. Payload compounds include compounds of Formulas 301-306, 15, 20a, 2b, and 20c above, and their residues following bonding to or incorporation of linker L are linker-payloads. The linker- payloads can be further bonded to binding agents such as antibodies or antigen binding fragments thereof to form antibody-drug conjugates. Those of skill in the art will recognize that certain functional groups of payload moieties are convenient for linking to linkers and/or binding agents. For example, in certain embodiments, the linker is absent and payloads are directly bonded to binding agents. In another embodiment, payloads include carboxylic acids and binding agents include lysines, where each carboxylic acid and lysine participate in amide bond formation to bind payload residues directly to binding agent residues. Payload functional groups further include carboxylic acids (e.g., in the form of esters upon linking to L, as in VX- 787 and derivatives thereof), hydroxamic acids, and ring nitrogens.

[00138] In certain embodiments, the linkers are stable in physiological conditions. In certain embodiments, the linkers are cleavable, for instance, able to release at least the payload portion in the presence of an enzyme or at a particular pH range or value. In some embodiments, a linker comprises an enzyme-cleavable moiety. Illustrative enzyme-cleavable moieties include, but are not limited to, peptide bonds, ester linkages, hydrazones, and disulfide linkages. In some embodiments, the linker comprises a cathepsin-cleavable linker.

[00139] In some embodiments, the linker comprises a non-cleavable moiety. In some embodiments, the non-cleavable linker is derived from residue thereof. In some embodiments, the non-cleavable linker-payload residue is or a regioisomer thereof. In some embodiments, the non- cleavable linker is derived from or a residue thereof. In some Payload embodiments, the non-cleavable linker-payload residue is ° , or a regioisomer thereof. In one embodiment, the linker is maleimide cyclohexane carboxylate or

4-(iV-maleimidomethyl)cyclohexanecarboxylic acid (MCC). In the structures, - s - indicates a bond to a binding agent. In the structures, in some examples, -f 5 - indicates an amide bond which results from the reaction of, for example, one or more binding agent glutamines with one or more linkers or linker-payloads having amine functionality. In the structures, in some examples, indicates a Diels-Alder residue which results from the reaction of, for example, a binding agent having an diene or dienophile functionality and a linker-payload having a complementary dienophile or diene functionality, respectively. In the structures, in some examples, indicates a click chemistry residue which results from the reaction of, for example, a binding agent having an azide or alkyne functionality and a linker-payload having a complementary alkyne or azide functionality. In the structures, in other examples, indicates a divalent sulfide which results from the reaction of, for example, one or more binding agent cysteines with one or more linkers or linker-payloads having maleimide functionality via

Michael addition reactions. In the structures, in other examples, indicates an amide bond which results from the reaction of, for example, one or more binding agent lysines with one or more linkers or linker-payloads having activated or unactivated carboxyl functionality, as would be appreciated by a person of skill in the art. In one embodiment, indicates an amide bond which results from the reaction of, for example, one or more binding agent lysines with one or more linkers or linker-payloads having activated carboxyl functionality, as would be appreciated by a person of skill in the art.

[00140] In some embodiments, suitable linkers include, but are not limited to, those that are chemically bonded to two cysteine residues of a single binding agent, e.g., antibody. Such linkers can serve to mimic the antibody’s disulfide bonds that are disrupted as a result of the conjugation process.

[00141] In some embodiments, the linker comprises one or more amino acids. Suitable amino acids include natural, non-natural, standard, non-standard, proteinogenic, non-proteinogenic, and L- or D- a-amino acids. In some embodiments, the linker comprises alanine, valine, glycine, leucine, isoleucine, methionine, tryptophan, phenylalanine, proline, serine, threonine, cysteine, tyrosine, asparagine, glutamine, aspartic acid, glutamic acid, lysine, arginine, histidine, or citrulline, a derivative thereof, or any combination thereof (e.g., dipeptides, tripeptides, oligopeptides, polypeptides, and the like). In certain embodiments, one or more side chains of the amino acids are linked to a side chain group, described below. In some embodiments, the linker is a peptide comprising or consisting of the amino acids valine and citrulline (e.g., divalent -Val-Cit- or divalent -VCit-). In some embodiments, the linker is a peptide comprising or consisting of the amino acids alanine and alanine, or divalent — AA— . In some embodiments, the linker is a peptide comprising or consisting of the amino acids glutamic acid and alanine, or -EA-. In some embodiments, the linker is a peptide comprising or consisting of the amino acids glutamic acid and glycine, or -EG-. In some embodiments, the linker is a peptide comprising or consisting of the amino acids glycine and glycine, or -GG-. In some embodiments, the linker is a peptide comprising or consisting of the amino acids glutamine, valine, and citrulline, or-Q-V-Cit- or-QVCit- In some embodiments, the linker is a peptide comprising or consisting of the amino acids glutamic acid, valine, and citrulline, or -E-V-Cit- or -EVCit- In some embodiments, the linker is a peptide comprising or consisting of the amino acids -GGGGS-. In some embodiments, the linker is a peptide comprising or consisting of the amino acids -GGGGG-. In some embodiments, the linker is a peptide comprising or consisting of the amino acids -GGGGK-. In some embodiments, the linker is a peptide comprising or consisting of the amino acids -GFGG-. In some embodiments, the linker is a peptide comprising or consisting of the amino acids -GGFG-. In some embodiments, the linker is a peptide comprising or consisting of the amino acids lysine, valine, and citrulline, or -KVCit- In some embodiments, the linker is a peptide comprising or consisting of the amino acids -KVA-. In some embodiments, the linker is a peptide comprising or consisting of the amino acids -VA-. In any of the embodiments in this paragraph, and throughout this disclosure, the standard three-letter or one-letter amino acid designations are used, as would be appreciated by a person of skill in the art. Exemplary single-letter amino acid designations include, G for glycine, K for lysine, S for serine, V for valine, A for alanine, and F for phenylalanine.

[00142] In some embodiments, the linker comprises a self-immolative group. The self-immolative group can be any such group known to those of skill. In particular embodiments, the self-immolative group is />aminoben/yl (PAB), or a derivative thereof. Useful derivatives include /?-aminoben/ylo\y carbonyl (PABC). Those of skill will recognize that a self-immolative group is capable of carrying out a chemical reaction which releases the remaining atoms of a linker from a payload.

[00143] In some embodiments, the linker is:

A P

|-SP 1 -(AA)— SP 2 - - wherein:

SP 1 is a spacer; SP 2 is a spacer; is one or more bonds to the binding agent;

-f 5 - is one or more bonds to the payload; each AA is an amino acid residue; and n is an integer from zero to ten.

[00144] The SP 1 spacer is a moiety that connects the (AA)„ moiety or residue to the binding agent (BA) or to a reactive group residue which is bonded to BA. Suitable SP 1 spacers include, but are not limited to, those comprising alkylene or polyether, or both. The ends of the spacers, for example, the portion of the spacer bonded to the BA or an AA, can be moieties derived from reactive moieties that are used for purposes of coupling the antibody or an AA to the spacer during chemical synthesis of the conjugate. In certain embodiments, n is 0, 1, 2, 3, or 4 (i.e., when n is 0, AA is absent). In particular embodiments, n is 2. In particular embodiments, n is 3. In particular embodiments, n is 4. In certain embodiments, SP 1 is absent. In certain embodiments, SP 2 is absent. In certain embodiments, (AA) n is absent.

[00145] In some embodiments, the SP 1 spacer comprises an alkylene. In some embodiments, the SP 1 spacer comprises a C5-7 alkylene. In some embodiments, the SP 1 spacer comprises a polyether. In some embodiments, the SP 1 spacer comprises a polymer of ethylene oxide such as polyethylene glycol (PEG). Polymeric units of polyethylene glycol are commonly represented as -(OCEhCEy p- , where p could be an integer from one to one hundred. For example, -(OCEECEh^- can also be represented as -OCH2CH2-OCH2CH2- or PEG 2 . In certain embodiments, the polyethylene glycol is PEGi. In certain embodiments, the polyethylene glycol is PEG 2 . In certain embodiments, the polyethylene glycol is PEG 3 . In certain embodiments, the polyethylene glycol is PEG 4 . In certain embodiments, the polyethylene glycol is PEG 5 . In certain embodiments, the polyethylene glycol is PEG 6 . In certain embodiments, the polyethylene glycol is PEG 7 . In certain embodiments, the polyethylene glycol is PEGs. In certain embodiments, the polyethylene glycol is PEG9. In certain embodiments, the polyethylene glycol is PEG10. In certain embodiments, the polyethylene glycol is PEGn. In certain embodiments, the polyethylene glycol is PEG 12 . In certain embodiments, the polyethylene glycol is PEG 13 . In certain embodiments, the polyethylene glycol is PEG14. In certain embodiments, the polyethylene glycol is PEG15. In certain embodiments, the polyethylene glycol is PEG 16 . In certain embodiments, the polyethylene glycol is PEG 17 . In certain embodiments, the polyethylene glycol is PEGis. In certain embodiments, the polyethylene glycol is PEG 19 . In certain embodiments, the polyethylene glycol is PEG20. In certain embodiments, the polyethylene glycol is PEG21. In certain embodiments, the polyethylene glycol is PEG22. In certain embodiments, the polyethylene glycol is PEG23. In certain embodiments, the polyethylene glycol is PEG24. In certain embodiments, the polyethylene glycol is PEG25. In certain embodiments, the polyethylene glycol is PEG26. In certain embodiments, the polyethylene glycol is PEG27. In certain embodiments, the polyethylene glycol is PEG28. In certain embodiments, the polyethylene glycol is PEG29. In certain embodiments, the polyethylene glycol is PEG30. In certain embodiments, the polyethylene glycol is PEG31. In certain embodiments, the polyethylene glycol is PEG32. In certain embodiments, the polyethylene glycol is PEG33. In certain embodiments, the polyethylene glycol is PEG34. In certain embodiments, the polyethylene glycol is PEG35. In certain embodiments, the polyethylene glycol is PEG36. In certain embodiments, the polyethylene glycol is PEG37. In certain embodiments, the polyethylene glycol is PEG38. In certain embodiments, the polyethylene glycol is PEG39. In certain embodiments, the polyethylene glycol is PEG40. In certain embodiments, the polyethylene glycol is PEG41. In certain embodiments, the polyethylene glycol is PEG42. In certain embodiments, the polyethylene glycol is PEG43. In certain embodiments, the polyethylene glycol is PEG44. In certain embodiments, the polyethylene glycol is PEG45. In certain embodiments, the polyethylene glycol is PEG46. In certain embodiments, the polyethylene glycol is PEG47. In certain embodiments, the polyethylene glycol is PEG48. In certain embodiments, the polyethylene glycol is PEG49. In certain embodiments, the polyethylene glycol is PEG50. In certain embodiments, the polyethylene glycol is PEG51. In certain embodiments, the polyethylene glycol is PEG52. In certain embodiments, the polyethylene glycol is PEG53. In certain embodiments, the polyethylene glycol is PEG54. In certain embodiments, the polyethylene glycol is PEG55. In certain embodiments, the polyethylene glycol is PEG56. In certain embodiments, the polyethylene glycol is PEG57. In certain embodiments, the polyethylene glycol is PEG58. In certain embodiments, the polyethylene glycol is PEG59. In certain embodiments, the polyethylene glycol is PEG60. In certain embodiments, the polyethylene glycol is PEG61. In certain embodiments, the polyethylene glycol is PEG62. In certain embodiments, the polyethylene glycol is PEG63. In certain embodiments, the polyethylene glycol is PEG64. In certain embodiments, the polyethylene glycol is PEG65. In certain embodiments, the polyethylene glycol is PEG66. In certain embodiments, the polyethylene glycol is PEG67. In certain embodiments, the polyethylene glycol is PEG 68 . In certain embodiments, the polyethylene glycol is PEG69. In certain embodiments, the polyethylene glycol is PEG70. In certain embodiments, the polyethylene glycol is PEG71. In certain embodiments, the polyethylene glycol is PEG72. In certain embodiments, the polyethylene glycol is PEG 73 . In certain embodiments, the polyethylene glycol is PEG 74 . In certain embodiments, the polyethylene glycol is PEG 75 . In certain embodiments, the polyethylene glycol is PEG 76 . In certain embodiments, the polyethylene glycol is PEG 77 . In certain embodiments, the polyethylene glycol is PEG 78 . In certain embodiments, the polyethylene glycol is PEG 79 . In certain embodiments, the polyethylene glycol is PEGso. In certain embodiments, the polyethylene glycol is PEGsi. In certain embodiments, the polyethylene glycol is PEGs 2 . In certain embodiments, the polyethylene glycol is PEGs 3 . In certain embodiments, the polyethylene glycol is PEGs 4 . In certain embodiments, the polyethylene glycol is PEGss. In certain embodiments, the polyethylene glycol is PEGs6. In certain embodiments, the polyethylene glycol is PEGs?. In certain embodiments, the polyethylene glycol is PEGss. In certain embodiments, the polyethylene glycol is PEGsy. In certain embodiments, the polyethylene glycol is PEG 90 . In certain embodiments, the polyethylene glycol is PEG 91 . In certain embodiments, the polyethylene glycol is PEG92.

[00146] In some embodiments, the SP 1 spacer is: wherein:

X is absent or -N(H)-;

RG' is a reactive group residue following reaction of a reactive group RG with a binding agent; is a bond to the binding agent; is a bond to (AA)„; n is an integer from zero to ten; and b is, independently, an integer from 1 to 92.

[00147] The reactive group RG can be any reactive group known to those of skill in the art to be capable of forming one or more bonds to the binding agent. The reactive group RG is a moiety comprising a portion in its structure that is capable of reacting with the binding agent ( e.g ., reacting with an antibody at its glutamine, cysteine, or lysine residues, or at a diene or dienophile moiety or an azide moiety, for example, a PEG-N 3 functionalized antibody at one or more glutamine residues; or at an amino moiety, for example, a PEG-NFh functionalized antibody at one or more glutamine residues) to form antibody-drug conjugates described herein. Following conjugation to the binding agent, the reactive group becomes the reactive group residue (RG'). Illustrative reactive groups include, but are not limited to, those that comprise amino, haloacetyl, isothiocyanate, succinimide, N-hydroxysuccinimide, or maleimide portions that are capable of reacting with the binding agent.

[00148] The SP 2 spacer, when present, is a moiety that connects the (AA)„ moiety to the payload. Suitable spacers include, but are not limited to, those described above as SP 1 spacers. Further suitable SP 2 spacers include, but are not limited to, those comprising alkylene or poly ether, or both. The ends of the SP 2 spacers, for example, the portion of the spacer directly bonded to the payload or an AA, can be moieties derived from reactive moieties that are used for purposes of coupling the payload or AA to the SP 2 spacer during the chemical synthesis of the conjugate. In some examples, the ends of the SP 2 spacers, for example, the portion of the SP 2 spacer directly bonded to the payload or an AA, can be residues of reactive moieties that are used for purposes of coupling the payload or an AA to the spacer during the chemical synthesis of the conjugate.

[00149] In some embodiments, the SP 2 spacer, when present, is selected from the group consisting of -NH-(/>G,H 4 )-ClT-. -NH-(/>G,H 4 )-CH 2 0C(0)-. NH-(/>G,FI 4 )-CH(CtT)0-. an amino acid, a dipeptide, a tripeptide, an oligopeptide any combinations thereof. In certain p embodiments, each * is a bond to the payload, and each 5 is a bond to (AA)„ or absent if n = 0.

[00150] In the above formulae, each (AA)„ is an amino acid or, optionally, a /?-aminobenzyloxy carbonyl residue (PABC). n can be 0; if so, (AA)„ is absent. If PABC is present, preferably only one PABC is present. Preferably, the PABC residue, if present, is bonded to a terminal AA in the (AA) n group, proximal to the payload . Suitable amino acids for each AA include natural, non-natural, standard, non-standard, proteinogenic, non- proteinogenic, and L- or D- a-amino acids. In some embodiments, the AA comprises alanine, valine, leucine, isoleucine, methionine, tryptophan, phenylalanine, proline, serine, threonine, cysteine, tyrosine, asparagine, glutamine, aspartic acid, glutamic acid, lysine, arginine, histidine, or citrulline, a derivative thereof, or any combinations thereof (e.g., dipeptides, tripeptides, and oligopeptides, and the like). In certain embodiments, one or more side chains of the amino acids is linked to a side chain group, described below. In some embodiments, n is two. In some embodiments, the (AA) n is valine-citrulline. In some embodiments, (AA) n is citrulline-valine. In some embodiments, (AA) n is valine-alanine. In some embodiments, (AA) n is alanine-valine. In some embodiments, (AA) n is valine-glycine. In some embodiments, (AA) n is glycine-valine. In some embodiments, (AA) n is glutamate-valine-citrulline. In some embodiments, (AA) n is glutamine-valine-citrulline. In some embodiments, (AA) n is glycine- glycine-phenylalanine-glycine In some embodiments, the (AA) n is valine-citrulline-PABC. In some embodiments, (AA) n is citrulline-valine-PABC. In some embodiments, n is three. In some embodiments, (AA) n is glutamate-valine-citrulline. In some embodiments, (AA) n is glutamine-valine-citrulline. In some embodiments, (AA) n is lysine-valine-alanine. In some embodiments, (AA) n is lysine-valine-citrulline. In some embodiments, n is four. In some embodiments, (AA) n is glutamate-valine-citrulline-PABC. In some embodiments, (AA) n is glutamine-valine-citrulline-PABC.

[00151] In certain embodiments, (AA) n -SP 2 is valine-citrulline-PABC. In certain embodiments, (AA) n -SP 2 is citrulline-valine-PABC. In certain embodiments, (AA) n -SP 2 is glutamate-valine-citrulline-PABC. In certain embodiments, (AA) n -SP 2 is glutamine-valine- citrulline-PABC. In certain embodiments, (AA) n -SP 2 is glycine-glycine-phenylalanine- glycine-N(H)-CH2-. In certain embodiments, (AA) n -SP 2 is valine-alanine-PABC. In certain embodiments, (AA) n -SP 2 is valine-ci trull ine-NH-(/>Ci,H4)-Cfh- In certain embodiments, (AA) n -SP 2 is valine-ci trull ine-NH-(/>Cr,H 4 )-CH(CH 3 )0- In certain embodiments, (AA) n -SP 2 is valine-alanine-NH-(/>Cr,H4)-Cfh-. In certain embodiments, (AA) n -SP 2 is valine-alanine- NH-(P-C 6 H 4 )-CH 2 0C(0)-.

[00152] Those of skill will recognize PABC as a residue of />-aminobenzyloxy carbonyl with the following structure:

The PABC residue has been shown to facilitate cleavage of certain linkers in vitro and in vivo. For example, in certain embodiments, upon cleavage of PABC, the carboxylate or carboxylic acid moiety (i.e., O or O respectively) remains intact with the remainder of p the antiviral compound or payload. In certain embodiments, each « is a bond to the remainder of the antiviral compound (e.g., the payload). Those of skill will recognize PAB as a divalent

H residue of />ami nobenzyl (i.e., -NH-(/>G,H4)-CH2- or In certain embodiments, the PAB residue has been shown to facilitate cleavage of certain linkers in vitro

H and in vivo. For example, in certain embodiments, upon cleavage of v p the alkoxide or hydroxyl moiety (i.e respectively) remains intact with the remainder of the antiviral compound (e.g., the payload). In certain embodiments, each is a bond to the remainder of the antiviral compound or payload.

Linker -Payloads

[00153] In certain embodiments, linker-payloads include any specific compound or payload embraced by any one or more of Formulas 301-306, 15, 20a, 20b, and 20c above, bonded to a linker, wherein the linker(s) described herein include a moiety that is reactive with an antibody or antigen binding fragment thereof described herein. In particular embodiments, the linker is bonded to the carboxyl, hydroxamic acid, or ring nitrogen in any one or more of Formulas 301- 306, 15, 20a, 20b, and 20c above.

[00154] In certain embodiments, set forth herein is a compound having the structure of Formula 401: or a pharmaceutically acceptable salt thereof. In Formula 401, L is a linker; RG is a reactive moiety; R 1 is F and R 2 is H; or R 1 and R 2 cyclize to form a fused five-membered heteroaryl ring, optionally substituted with methyl; R 3 is H or HO-CH2-; Cy is bridged 5 or 6 membered cycloalkyl, wherein the bridge is methylene or ethylene; and Q is -O- or -0-NH-. In certain embodiments, when R 3 is H, then Q is -0-NH-. In certain embodiments, L is any linker described herein. In certain embodiments, RG is any reactive group described herein. In certain embodiments, R 3 is H and Q is -0-NH-. In certain embodiments, R 1 is F and R 2 is H. In certain embodiments, R 1 and R 2 cyclize as -C=CH-S-. In certain embodiments, R 1 and R 2 cyclize as -C=CH-NMe-. In certain embodiments, Cy is . In certain embodiments,

RG is selected from the group consisting of -NFf, maleimide, NHS ester, alkyne, strained alkyne, diene, and dienophile. In certain embodiments, RG is -NFf.

[00155] In certain embodiments, set forth herein is a compound having the structure of Formula 402: or a pharmaceutically acceptable salt thereof. In Formula 402, L is a linker; RG is a reactive moiety; R 1 is F and R 2 is H; or R 1 and R 2 cyclize to form a fused five-membered heteroaryl ring, optionally substituted with methyl; R 3 is H or HO-CH2-; and Cy is bridged 5 or 6 membered cycloalkyl, wherein the bridge is methylene or ethylene. In certain embodiments, L is any linker described herein. In certain embodiments, RG is any reactive group described herein. In certain embodiments, R 1 and R 2 cyclize as -C=CH-S-. In certain embodiments, R 1 and R 2 cyclize as -C=CH-NMe-. In certain embodiments, R 1 is F and R 2 is H. In certain embodiments, R 1 is F and R 2 is H and Cy is . In certain embodiments, RG is selected from the group consisting of -NFh, maleimide, NHS ester, alkyne, strained alkyne, diene, and dienophile. In certain embodiments, RG is -Nth.

[00156] In certain embodiments, set forth herein is a compound having the structure of Formula 403: or a pharmaceutically acceptable salt thereof. In Formula 403, L is a linker; RG is a reactive moiety; R 1 is F and R 2 is H; or R 1 and R 2 cyclize to form a fused five-membered heteroaryl ring, optionally substituted with methyl; and Cy is bridged 5 or 6 membered cycloalkyl, wherein the bridge is methylene or ethylene. In certain embodiments, RG is selected from the group consisting of -NFh, maleimide, NHS ester, alkyne, strained alkyne, diene, and dienophile. In certain embodiments, RG is -NH2.

[00157] In some embodiments, the linker L is: wherein:

SP 1 is a spacer;

SP 2 is a spacer; is one or more bonds to the binding agent;

5 is one or more bonds to the payload; each AA is an amino acid residue; and n is an integer from zero to ten. [00158] The SP 1 spacer is described above. In some embodiments, the SP 1 spacer is: wherein:

X is absent or -N(H)-; n is an integer from zero to ten; and b is, independently, an integer from 1 to 92.

[00159] The SP 2 spacer is described above. In some embodiments, the SP 2 spacer, when present, is selected from the group consisting of -NH-(/>G,H4)-Cfb-. -NH-f/j-G.tB)- CTkOCXO)-, NH-(/>G,H 4 )-CH(Cfh)0-. an amino acid, a dipeptide, a tripeptide, an oligopeptide , and any combinations thereof. In certain embodiments, each is a bond to the payload, and each is a bond to (AA)„ or absent if n = 0.

[00160] In the above formulae, each (AA)„ is an amino acid or, optionally, a /?-aminobenzyloxy carbonyl residue (PABC). n can be 0; if so, (AA)„ is absent. If PABC is present, preferably only one PABC is present. Preferably, the PABC residue, if present, is bonded to a terminal AA in the (AA) n group, proximal to the payload . Suitable amino acids for each AA include natural, non-natural, standard, non-standard, proteinogenic, non- proteinogenic, and L- or D- a-amino acids. In some embodiments, the AA comprises alanine, valine, leucine, isoleucine, methionine, tryptophan, phenylalanine, proline, serine, threonine, cysteine, tyrosine, asparagine, glutamine, aspartic acid, glutamic acid, lysine, arginine, histidine, or citrulline, a derivative thereof, or any combinations thereof (e.g., dipeptides, tripeptides, and oligopeptides, and the like). In certain embodiments, one or more side chains of the amino acids is linked to a side chain group, described below. In some embodiments, n is two. In some embodiments, the (AA)„ is valine-citrulline. In some embodiments, (AA)„ is citrulline-valine. In some embodiments, (AA) n is valine-alanine. In some embodiments, (AA) n is alanine-valine. In some embodiments, (AA) n is valine-glycine. In some embodiments, (AA) n is glycine-valine. In some embodiments, (AA) n is glutamate-valine-citrulline. In some embodiments, (AA) n is glutamine-valine-citrulline. In some embodiments, (AA) n is glycine- glycine-phenylalanine-glycine In some embodiments, the (AA) n is valine-citrulline-PABC. In some embodiments, (AA) n is citrulline-valine-PABC. In some embodiments, n is three. In some embodiments, (AA) n is glutamate-valine-citrulline. In some embodiments, (AA) n is glutamine-valine-citrulline. In some embodiments, (AA) n is lysine-valine-alanine. In some embodiments, (AA) n is lysine-valine-citrulline. In some embodiments, n is four. In some embodiments, (AA) n is glutamate-valine-citrulline-PABC. In some embodiments, (AA) n is glutamine-valine-citrulline-PABC.

[00161] In certain embodiments, provided herein are compounds (viz., linker-payloads) selected from the group consisting of:

Conjugates/Antibody Drug Conjugates (ADCs)

[00162] Provided herein are human anti-influenza-HA monoclonal antibodies conj ugated to a therapeutic moiety, such as a toxoid or an antiviral drug, to treat influenza virus infection (i.e., an ADC). The antibody may be linked to the therapeutic agent at any location along the antibody so long as the antibody is able to bind its target. In one embodiment, the therapeutic agent may be a second different antibody to Influenza-HA or ADC thereof. In certain embodiments, the antibody may be conjugated to an agent specific for a virally infected cell. The type of therapeutic moiety that may be conjugated to the anti-influenza-HA antibody and will take into account the condition to be treated and the desired therapeutic effect to be achieved. In certain embodiments, provided herein are antibodies, or an antigen binding fragment thereof, wherein the antibody is conjugated to one or more compounds of Formulae I and/or II as described herein. In one embodiment, the anti-influenza antibody or antigen binding fragment thereof is conjugated to a payload via a linker, each as described in any of their respective embodiments disclosed herein.

[00163] In one embodiment, the antibody-drug conjugate has the following structure

BA — L-P k wherein BA is an anti-influenza binding agent; L is a linker as described herein; and P is an antiviral compound or payload as described herein. In certain embodiments, embodiments, BA is Ab, an anti-influenza antibody or an antigen binding fragment thereof. In one embodiment, Ab is an anti-influenza antibody or an antigen binding fragment thereof; and P is an influenza inhibitor. In one embodiment, Ab is an anti-influenza antibody or an antigen binding fragment thereof; and P is a polymerase inhibitor. In one embodiment, Ab is an anti-influenza antibody or an antigen binding fragment thereof; and P is VX-787, a derivative thereof, or a residue thereof. In one embodiment, Ab is an anti -hemagglutinin antibody or an antigen binding fragment thereof; and P is an antiviral compound. In one embodiment, Ab is an anti-influenza antibody or an antigen binding fragment thereof; and P is an antiviral compound. In one embodiment, Ab is an anti -hemagglutinin antibody or an antigen binding fragment thereof; and P is an influenza inhibitor. In one embodiment, Ab is an anti -hemagglutinin antibody or an antigen binding fragment thereof; and P is a polymerase inhibitor. In any embodiment in this paragraph, Ab is an anti-influenza antibody or an antigen binding fragment thereof or an anti hemagglutinin antibody or an antigen binding fragment thereof wherein the antibody is conjugated to a compound according to 301, 401, 402, or 403, as described above. In one embodiment, Ab is an anti -hemagglutinin antibody or an antigen binding fragment thereof; and P is VX-787, a derivative thereof, or a residue thereof. In any of the embodiments in this paragraph, k is an integer from one to thirty.

[00164] In certain embodiments, provided herein are ADCs where the antibody or antigen binding fragment thereof is conjugated to a linker-payload compound of any of the following formulas as described herein:

or a pharmaceutically acceptable salt thereof. In Formula 101, BA is a binding agent as described herein; L is a linker as described herein; R 1 is F and R 2 is H; or R 1 and R 2 cyclize to form a fused five-membered heteroaryl ring, optionally substituted with methyl; R 3 is H or HO-CH2-; Cy is bridged 5 or 6 membered cycloalkyl, wherein the bridge is methylene or ethylene; and Q is -O- or -0-NH-; wherein when R 3 is H, then Q is -0-NH-. In certain embodiments, R 3 is HO-CH2-. In certain embodiments, R 3 is H and Q is -0-NH-. In certain embodiments, R 1 is F and R 2 is H. In certain embodiments, R 1 and R 2 cyclize as -C=CH-S-. In certain embodiments, R 1 and R 2 cyclize as -C=CH-NMe-. In certain embodiments, Cy is bridged 6-membered cycloalkyl. In certain embodiments, Cy is . In certain embodiments, Cy is . In certain embodiments, R 3 is H; Q is -0-NH-; R 1 is F; and R 2 is H. In certain embodiments, R 3 is H; Q is -0-NH-; R 1 is F; R 2 is H; and Cy is . In certain embodiments, R 3 is H; Q is -0-NH-; R 1 and R 2 cyclize as -C=CH-S - C=CH-S-; and Cy is H 'q — J . In certain embodiments, R 3 is H; Q is -0-; R 1 and R 2 cyclize

[00166] In certain embodiments, provided herein are ADCs according to Formula 201: or a pharmaceutically acceptable salt thereof. In Formula 201, BA is a binding agent as described herein; L is a linker as described herein; R 1 is F and R 2 is H; or R 1 and R 2 cyclize to form a fused five-membered heteroaryl ring, optionally substituted with methyl; R 3 is H or HO-CH2-; Cy is bridged 5 or 6 membered cycloalkyl, wherein the bridge is methylene or ethylene. In certain embodiments, R 3 is HO-CH2-. In certain embodiments, R 3 is H. In certain embodiments, R 1 is F and R 2 is H. In certain embodiments, Cy is bridged 6-membered cycloalkyl. In certain embodiments, In certain embodiments, Cy is . In certain embodiments, R 3 is H; R 1 is F; and R 2 is H. In certain embodiments,

[00167] In certain embodiments, provided herein are ADCs according to Formula 102: or a pharmaceutically acceptable salt thereof. In Formula 102, BA is a binding agent as described herein; L is a linker as described herein; R 1 is F and R 2 is H; or R 1 and R 2 cyclize to form a fused five-membered heteroaryl ring, optionally substituted with methyl; R 3 is H or HO-CH2-; and Q is -O- or -0-NH-; wherein when R 3 is H, then Q is -0-NH-. In certain embodiments, R 3 is HO-CH2-. In certain embodiments, R 3 is H and Q is -0-NH-. In certain embodiments, R 1 is F and R 2 is H. In certain embodiments, R 1 and R 2 cyclize as -C=CH-S-. In certain embodiments, R 1 and R 2 cyclize as -C=CH-NMe-. In certain embodiments, R 3 is H; Q is -0-NH-; R 1 is F; and R 2 is H. In certain embodiments, R 3 is H; Q is -0-; R 1 is F; and R 2 is H. In certain embodiments, R 3 is H; Q is -0-NH-; and R 1 and R 2 cyclize as -C=CH-S-. In certain embodiments, R 3 is H; Q is -0-NH-; and R 1 and R 2 cyclize as -C=CH-NMe-.

[00168] In certain embodiments, provided herein are ADCs according to Formula 202: or a pharmaceutically acceptable salt thereof. In Formula 202, BA is a binding agent as described herein; L is a linker as described herein; R 1 is F and R 2 is H; or R 1 and R 2 cyclize to form a fused five-membered heteroaryl ring, optionally substituted with methyl; and R 3 is H or HO-CH2-. In certain embodiments, R 3 is HO-CH2-. In certain embodiments, R 3 is H. In certain embodiments, R 1 is F and R 2 is H. In certain embodiments, R 3 is H; R 1 is F; and R 2 is H. In certain embodiments, R 1 and R 2 cyclize as -C=CH-S-. In certain embodiments, R 1 and R 2 cyclize as -C=CH-NMe-. [00169] In certain embodiments, provided herein are ADCs according to Formula 112: or a pharmaceutically acceptable salt thereof. In Formula 112, BA is a binding agent as described herein; L is a linker as described herein; R 1 is F and R 2 is H; or R 1 and R 2 cyclize to form a fused five-membered heteroaryl ring, optionally substituted with methyl; and Q is - O- or -0-NH-. In certain embodiments, Q is -0-. In certain embodiments, Q is -0-NH-. In certain embodiments, R 1 is F and R 2 is H. In certain embodiments, R 1 and R 2 cyclize as - C=CH-S-. In certain embodiments, R 1 and R 2 cyclize as -C=CH-NMe-. In certain embodiments, Q is -0-; R 1 is F; and R 2 is H. In certain embodiments, Q is -0-NH-; R 1 is F; and R 2 is H. In certain embodiments, Q is -0-NH-; and R 1 and R 2 cyclize as -C=CH-S-. In certain embodiments, Q is -0-NH-; and R 1 and R 2 cyclize as -C=CH-NMe-.

[00170] In certain embodiments, provided herein are ADCs according to Formula 202: or a pharmaceutically acceptable salt thereof. In Formula 212, BA is a binding agent as described herein; L is a linker as described herein; R 1 is F and R 2 is H; or R 1 and R 2 cyclize to form a fused five-membered heteroaryl ring, optionally substituted with methyl. In certain embodiments, R 1 is F and R 2 is H. In certain embodiments, R 1 and R 2 cyclize as -C=CH-S-. In certain embodiments, R 1 and R 2 cyclize as -C=CH-NMe-.

[00171] In certain embodiments, provided herein are ADCs according to Formula 102: or a pharmaceutically acceptable salt thereof. In Formula 122, BA is a binding agent as described herein; L is a linker as described herein; R 1 is F and R 2 is H; or R 1 and R 2 cyclize to form a fused five-membered heteroaryl ring, optionally substituted with methyl; R 3 is H or HO-CH2-; and Q is -O- or -0-NH-; wherein when R 3 is H, then Q is -0-NH-. In certain embodiments, R 3 is HO-CH2-. In certain embodiments, R 3 is H and Q is -0-NH-. In certain embodiments, R 1 is F and R 2 is H. In certain embodiments, R 1 and R 2 cyclize as -C=CH-S-. In certain embodiments, R 1 and R 2 cyclize as -C=CH-NMe-. In certain embodiments, R 3 is H; Q is -0-NH-; R 1 is F; and R 2 is H. In certain embodiments, R 3 is H; Q is -0-NH-; R 1 is F; and R 2 is H. In certain embodiments, R 3 is H; Q is -0-NH-; and R 1 and R 2 cyclize as - C=CH-S-. In certain embodiments, R 3 is H; Q is -0-NH-; and R 1 and R 2 cyclize as -C=CH- NMe-.

[00172] In certain embodiments, provided herein are ADCs according to Formula 202: or a pharmaceutically acceptable salt thereof. In Formula 222, BA is a binding agent as described herein; L is a linker as described herein; R 1 is F and R 2 is H; or R 1 and R 2 cyclize to form a fused five-membered heteroaryl ring, optionally substituted with methyl; and R 3 is H or HO-CH2-. In certain embodiments, R 1 and R 2 cyclize as -C=CH-S-. In certain embodiments, R 1 and R 2 cyclize as -C=CH-NMe-. In certain embodiments, R 3 is HO-CH2-. In certain embodiments, R 3 is H. In certain embodiments, R 1 is F and R 2 is H. In certain embodiments, R 3 is H; R 1 is F; and R 2 is H.

[00173] In certain embodiments, provided herein are ADCs according to Formula 103: or a pharmaceutically acceptable salt thereof. In Formula 103, BA is a binding agent as described herein; L is a linker as described herein; R 1 is F and R 2 is H; or R 1 and R 2 cyclize to form a fused five-membered heteroaryl ring, optionally substituted with methyl; R 3 is H or HO-CH2-; and Q is -O- or -0-NH-; wherein when R 3 is H, then Q is -0-NH-. In certain embodiments, R 3 is HO-CH2-. In certain embodiments, R 3 is H and Q is -0-NH-. In certain embodiments, R 1 is F and R 2 is H. In certain embodiments, R 1 and R 2 cyclize as -C=CH-S-. In certain embodiments, R 1 and R 2 cyclize as -C=CH-NMe-. In certain embodiments, R 3 is H; Q is -0-NH-; R 1 is F; and R 2 is H. In certain embodiments, R 3 is H; Q is -0-NH-; R 1 is F; and R 2 is H. In certain embodiments, R 3 is H; Q is -0-NH-; and R 1 and R 2 cyclize as - C=CH-S-. In certain embodiments, R 3 is H; Q is -0-NH-; and R 1 and R 2 cyclize as -C=CH- NMe-.

[00174] In certain embodiments, provided herein are ADCs according to Formula 203: or a pharmaceutically acceptable salt thereof. In Formula 203, BA is a binding agent as described herein; L is a linker as described herein; R 1 is F and R 2 is H; or R 1 and R 2 cyclize to form a fused five-membered heteroaryl ring, optionally substituted with methyl; and R 3 is H or HO-CH2-. In certain embodiments, R 3 is HO-CH2-. In certain embodiments, R 3 is H. In certain embodiments, R 1 is F and R 2 is H. In certain embodiments, R 3 is H; R 1 is F; and R 2 is H.

[00175] In certain embodiments, provided herein are ADCs according to Formula 103: or a pharmaceutically acceptable salt thereof. In Formula 104, BA is a binding agent as described herein; L is a linker as described herein; R 1 is F and R 2 is H; or R 1 and R 2 cyclize to form a fused five-membered heteroaryl ring, optionally substituted with methyl; and Q is - O- or -0-NH-. In certain embodiments, R 1 is F and R 2 is H. In certain embodiments, R 1 and R 2 cyclize as -C=CH-S-. In certain embodiments, R 1 and R 2 cyclize as -C=CH-NMe-. In certain embodiments, Q is -0-NH-; R 1 is F; and R 2 is H. In certain embodiments, Q is -O- NH-; and R 1 and R 2 cyclize as -C=CH-S-. In certain embodiments, Q is -0-NH-; and R 1 and R 2 cyclize as -C=CH-NMe-.

[00176] In certain embodiments, provided herein are ADCs according to Formula 204: or a pharmaceutically acceptable salt thereof. In Formula 204, BA is a binding agent as described herein; L is a linker as described herein; and R 1 is F and R 2 is H, or R 1 and R 2 cyclize to form a fused five-membered heteroaryl ring, optionally substituted with methyl. In certain embodiments, R 1 is F and R 2 is H.

[00177] In certain embodiments, provided herein are ADCs according to Formula 105: or a pharmaceutically acceptable salt thereof. In Formula 105, BA is a binding agent as described herein; L is a linker as described herein.

[00178] In certain embodiments, provided herein are ADCs according to Formula 205: or a pharmaceutically acceptable salt thereof. In Formula 205, BA is a binding agent as described herein; and L is a linker as described herein.

[00179] In certain embodiments, provided herein are ADCs according to Formula 106: or a pharmaceutically acceptable salt thereof. In Formula 106, BA is a binding agent as described herein; L is a linker as described herein; and R 1 is F and R 2 is H; or R 1 and R 2 cyclize to form a fused five-membered heteroaryl ring, optionally substituted with methyl. In certain embodiments, R 1 is F and R 2 is H. In certain embodiments, R 1 and R 2 cyclize as - C=CH-S-. In certain embodiments, R 1 and R 2 cyclize as -C=CH-NMe-.

[00180] In certain embodiments, provided herein are ADCs according to Formula 206: or a pharmaceutically acceptable salt thereof. In Formula 206, BA is a binding agent as described herein; L is a linker as described herein; and R 1 is F and R 2 is H, or R 1 and R 2 cyclize to form a fused five-membered heteroaryl ring, optionally substituted with methyl. In certain embodiments, R 1 is F and R 2 is H.

[00181] In certain embodiments, provided herein are ADCs according to Formula 107: or a pharmaceutically acceptable salt thereof. In Formula 107, BA is a binding agent as described herein; L is a linker as described herein.

[00182] In certain embodiments, provided herein are ADCs according to Formula 207: or a pharmaceutically acceptable salt thereof. In Formula 207, BA is a binding agent as described herein; and L is a linker as described herein.

[00183] In certain embodiments, provided herein are ADCs according to Formula 108: or a pharmaceutically acceptable salt thereof. In Formula 105, BA is a binding agent as described herein; L is a linker as described herein.

[00184] In certain embodiments, provided herein are ADCs according to Formula 208: or a pharmaceutically acceptable salt thereof. In Formula 208, BA is a binding agent as described herein; and L is a linker as described herein.

[00185] In certain embodiments, provided herein are ADCs according to Formula 110:

or a pharmaceutically acceptable salt thereof. In Formula 110, BA is a binding agent as described herein; L is a linker as described herein; R 1 is F and R 2 is H; or R 1 and R 2 cyclize to form a fused five-membered heteroaryl ring, optionally substituted with methyl; and Q is - O- or -0-NH-. In certain embodiments, R 1 is F and R 2 is H. In certain embodiments, R 1 and R 2 cyclize as -C=CH-S-. In certain embodiments, R 1 and R 2 cyclize as -C=CH-NMe-.

[00186] In certain embodiments, provided herein are ADC compounds having the following structure:

wherein indicates linkage to BA.

[00187] In certain embodiments of Formulas 101-108, k is 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10. In certain embodiments, k is a range from 1-2, 1-3, 2-3, 2-4, 3-4, or 1-4. In certain embodiments, compounds conjugated to — L — BA as above include one or more compounds of Formulas 301-306, 15, 20a, 20b, and 20c, as described above, wherein BA is a binding agent; L is a linker; and k is 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10. In certain embodiments, where compounds conjugated to — L — BA as above include one or more compounds of Formulas 301-306, 15, 20a, 20b, and 20c, as described above, wherein BA is a binding agent and L is a linker, k is a range from 1-2, 1-3, 2-3, 2-4, 3-4, or 1-4.

[00188] In one embodiment, provided herein are ADC compounds selected from the group consisting of

WO 2023/009754

or a pharmaceutically acceptable salt thereof, wherein BA is an antibody or an antigen binding fragment thereof; and k is an integer from one to thirty. In certain embodiments, k is 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10. In certain embodiments, kis a range from 1-2, 1-3, 1-4, 1-5, 1-6, 1-7, 1-8, 1-9, 1-10, 2-3, 2-4, 2-5, 2-6, 2-7, 2-8, 2-9, 2-10, 3-4, 3-5, 3-6. 3-7. 3-8, 3-9, 3-10, 4-5, 4-6, 4- 7, 4-8, 4-9, 4-10, 5-6, 5-7, 5-8, 5-9, 5-10, 6-7, 6-8, 6-9, 6-10, 7-8, 7-9, 7-10, 8-9, 8-10, or 9-10. In any of the embodiments in this paragraph, BA is contemplated to include one or more glutamine residues for conjugation to a payload and/or linker-payload, when k is greater than one. For example, the above ADC delineations contemplate a drug:antibody ratio (DAR) of one or more where one or more glutamine residues of BA accommodate a payload and/or linker-payload (e.g., when k is > 1). The bonds from BA to -NH- indicate bonds from the linker-payload (L-P) to a transglutaminated glutamine residue of BA, respectively. Therefore, the nitrogen from a transglutaminated glutamine residue of BA, are within the brackets as drawn to show that BA can be conjugated with more than one payload and/or linker-payload (e.g., BA having a DAR > 1). In one embodiment, BA is an antibody or an antigen-binding fragment thereof as described herein.

[00189] In certain ADC embodiments described herein, BA is a transglutaminase-modified antibody or antigen-binding fragment thereof. In one embodiment, BA is a transglutaminase- modified antibody or antigen-binding fragment thereof comprising at least one glutamine residue used for conjugation. In one embodiment, BA is a transglutaminase-modified antibody or antigen-binding fragment thereof comprising at least two glutamine residues used for conjugation. In one embodiment, BA is a transglutaminase-modified antibody or antigen binding fragment thereof comprising at least three glutamine residues used for conjugation. In one embodiment, BA is a transglutaminase-modified antibody or antigen-binding fragment thereof comprising at least four glutamine residues used for conjugation. In one embodiment, BA is a transglutaminase-modified antibody or antigen-binding fragment thereof comprising at least one glutamine residue available for conjugation. In one embodiment, BA is a transglutaminase-modified antibody or antigen-binding fragment thereof comprising at least two glutamine residues available for conjugation. In one embodiment, BA is a transglutaminase-modified antibody or antigen-binding fragment thereof comprising at least three glutamine residues available for conjugation. In one embodiment, BA is a transglutaminase-modified antibody or antigen-binding fragment thereof comprising at least four glutamine residues available for conjugation. In one embodiment, BA is a transglutaminase-modified antibody or antigen-binding fragment thereof, wherein conjugation is at two Q295 residues; and k is 2. In one embodiment, BA is a transglutaminase-modified antibody or antigen-binding fragment thereof, wherein conjugation is at two Q295 residues in the EU numbering system; and k is 2. In one embodiment, BA is a transglutaminase-modified antibody or antigen-binding fragment thereof comprising an antibody heavy chain, wherein conjugation is at the C-terminus of the heavy chain; and k is 2. In one embodiment, BA is a transglutaminase-modified antibody or antigen-binding fragment thereof comprising an antibody heavy chain, wherein conjugation is via a glutamine. In one embodiment, BA is a transglutaminase-modified antibody or antigen-binding fragment thereof comprising an antibody heavy chain, wherein conjugation is via a glutamine; and k is 2. In one embodiment, BA is a transglutaminase-modified antibody or antigen-binding fragment thereof comprising an antibody heavy chain, wherein conjugation is via the glutamine in a LLQGA sequence at the C-terminus of the antibody heavy chain. In one embodiment, BA is a transglutaminase- modified antibody or antigen-binding fragment thereof comprising an antibody heavy chain, wherein conjugation is via the glutamine in a LLQGA sequence at the C-terminus of the antibody heavy chain; and k is 2. In one embodiment, BA is a transglutaminase-modified antibody or antigen-binding fragment thereof, wherein conjugation is at two Q295 residues and two N297Q residues; and k is 4. In one embodiment, BA is a transglutaminase-modified antibody or antigen-binding fragment thereof, wherein conjugation is at two Q295 residues in the EU numbering system and two N297Q residues; and k is 4. In one embodiment, BA is mAbll729, as described herein.

[00190] In one embodiment, BA is a transglutaminase-modified antibody or antigen-binding fragment thereof comprising an antibody heavy chain, wherein conjugation is at the C-terminus of the heavy chain; and DAR is a) about 2.0; b) is greater than 0 to about 12.0; c) is about 0.5 to about 8.0; d) is about 0.5 to about 6.0; e) is about 1.0 to about 4.0; f) is about 1.0 or about 2.0; g) is about 1.0; or h) is about 2.0. In one embodiment, BA is a transglutaminase-modified antibody or antigen-binding fragment thereof comprising an antibody heavy chain, wherein conjugation is via a glutamine. In one embodiment, BA is a transglutaminase-modified antibody or antigen-binding fragment thereof comprising an antibody heavy chain, wherein conjugation is via a glutamine; and DAR is a) about 2.0; b) is greater than 0 to about 12.0; c) is about 0.5 to about 8.0; d) is about 0.5 to about 6.0; e) is about 1.0 to about 4.0; f) is about 1.0 or about 2.0; g) is about 1.0; or h) is about 2.0. In one embodiment, BA is a transglutaminase-modified antibody or antigen-binding fragment thereof comprising an antibody heavy chain, wherein conjugation is via the glutamine in a LLQGA sequence at the C-terminus of the antibody heavy chain. In one embodiment, BA is a transglutaminase- modified antibody or antigen-binding fragment thereof comprising an antibody heavy chain, wherein conjugation is via the glutamine in a LLQGA sequence at the C-terminus of the antibody heavy chain; and DAR is a) about 2.0; b) is greater than 0 to about 12.0; c) is about 0.5 to about 8.0; d) is about 0.5 to about 6.0; e) is about 1.0 to about 4.0; f) is about 1.0 or about 2.0; g) is about 1.0; or h) is about 2.0. In one embodiment, BA is mAbll729, as described herein.

[00191] In certain ADC embodiments described herein, BA is an anti-influenza antibody or antigen-binding fragment thereof. In one embodiment, BA is an anti -influenza A antibody or an antigen binding fragment thereof. In one embodiment, BA is an anti-influenza A Group 1 antibody or an antigen binding fragment thereof. In one embodiment, BA is an anti-influenza HI antibody or an antigen binding fragment thereof. In one embodiment, BA is an antiinfluenza A Group 2 antibody or an antigen binding fragment thereof. In one embodiment, BA is an anti-influenza H3 antibody or an antigen binding fragment thereof. In one embodiment, BA is an anti-influenza B antibody or an antigen binding fragment thereof. In one embodiment, an ADC includes an anti -influenza antibody or antigen-binding fragment thereof conjugated to a payload via a linker, wherein the antibody-drug conjugate binds to and/or inhibits polymerase basic protein 2 (PB2), and/or polymerase basic protein 1 (PB1). In one embodiment, an ADC includes an anti -influenza antibody or antigen-binding fragment thereof conjugated to a payload via a linker, wherein the antibody-drug conjugate binds to and/or inhibits polymerase basic protein 2 (PB2) (VX-787) with an affinity of at least 4.0xl0 '9 M, at least 3.5xl0 '9 M, or at least 3.0xl0 '9 M as measured by ELISA. In one embodiment, an ADC includes an antiinfluenza antibody or antigen-binding fragment thereof conjugated to a payload via a linker, wherein the antibody-drug conjugate binds to and/or inhibits polymerase basic protein 1 (PB1) with an affinity of at least 4.0xl0 '9 M, at least 3.5xl0 '9 M, or at least 3.0xl0 '9 M as measured by ELISA. In one embodiment, an ADC includes an anti-influenza antibody or antigen-binding fragment thereof conjugated to a payload via a linker, wherein the antibody-drug conjugate binds to and/or inhibits polymerase basic protein 2 (PB2) (VX-787) with an IC50 of at least 2.5xl0 '9 M, at least 2.0xl0 '9 M, or at least 1.5xl0 '9 M as measured by ImmunoSpot® analysis. In one embodiment, an ADC includes an anti -influenza antibody or antigen-binding fragment thereof conjugated to a payload via a linker, wherein the antibody-drug conjugate binds to and/or inhibits polymerase basic protein 1 (PB1) with an IC50 of at least 2.5xl0 '9 M, at least 2.0xl0 '9 M, or at least 1.5xl0 '9 M as measured by ImmunoSpot® analysis. Methods of Preparing Compounds or Payloads, and Linker -Payloads

[00192] The compounds provided herein can be prepared, isolated, or obtained by any method apparent to those of skill in the art. Exemplary methods of preparation are described in detail in the Examples below. In certain embodiments, compounds provided herein are commercially available or can generally be prepared according to Schemes A-C:

Scheme A. Exemplary Preparation Scheme

VX-787 and derivatives

[00193] In the above Exemplary Preparation Scheme A (see, J. Med. Chem. 2014, 57, 6668), R 1 is described in the context of formulas 101-108, 301-306, and 401-403. In Scheme A, following Diels-Alder cycloaddition with maleic anhydride and 1,3-cyclohexadiene, endo-Al can be stirred under basic conditions to provide epimerized trans-A2. Curtius rearrangement and trapping with benzyl alcohol provides A3. Hydrogenation provides A4. Treatment with 2,4-dichloropyrimidines and chiral separation provides A6 via intermediate A5. Suzuki coupling with substituted azaindole boronate esters, followed by deprotection, yields compounds of formulas 301-306, including VX-787 and derivatives thereof.

[00194] The linker-payloads described herein can generally be synthesized by a series of coupling steps as shown in Scheme C:

Scheme C. Exemplary Preparation Scheme where n = 1-92 [00195] In the above Exemplary Preparation Scheme C, each R is as described in the context herein. In Scheme C, VX-787 and derivatives thereof are treated with Linkers bearing a leaving group (LG) to provide linker-payloads according to formulas 401-403 (e.g., linker-(VX-787)).

[00196] The conj ugates described herein can be synthesized by coupling the linker-payloads described herein with a binding agent, for example, an antibody described herein under standard conjugation conditions (see, e.g., Doronina etal. Nature Biotechnology 2003, 21, 778, which is incorporated herein by reference in its entirety). When the binding agent is an antibody, the antibody may be coupled to a linker-payload via one or more glutamine, cysteine, or lysine residues of the antibody. Linker-payloads can be coupled to glutamine residues, for example, by treating the antibody with a linker-payload containing a suitable reactive moiety, for example, an amino group in the presence of a transglutaminase enzyme under suitable reaction conditions (see, e.g., Examples herein). Linker-payloads can be coupled to cysteine residues, for example, by subjecting the antibody to a reducing agent, for example, dithiotheritol, to cleave the disulfide bonds of the antibody, purifying the reduced antibody, for example, by gel filtration, and subsequently treating the antibody with a linker-payload containing a suitable reactive moiety, for example, a maleimido group. Suitable solvents include, but are not limited to water, DMA, DMF, and DMSO. Linker-payloads containing a reactive group, for example, an activated ester or acid halide group, can be coupled to lysine residues of the antibody. Suitable solvents include, but are not limited to water, DMA, DMF, and DMSO. Conjugates can be purified using known protein techniques, including, for example, size exclusion chromatography, dialysis, and ultrafiltration/diafiltration.

[00197] Binding agents, for example antibodies, can be conjugated via Diels-Alder chemistry reactions. In some embodiments of Diels-Alder reactions, the linker-payload includes a reactive group, for example, a dienophile that is capable of undergoing Diels-Alder reaction with a diene. In some embodiments of Diels-Alder reactions, the linker-payload includes a reactive group, for example, a diene that is capable of undergoing Diels-Alder reaction with a dienophile. Binding agents, for example antibodies, can also be conjugated via click chemistry reactions. In some embodiments of said click chemistry reactions, the linker- payload includes a reactive group, for example, an alkyne that is capable of undergoing a regioisomeric 1,3-cycloaddition reaction with an azide. Such suitable reactive groups are described above.

[00198] In certain embodiments, the antibody is functionalized with, for example, diene- polyethylene glycol groups or azido-polyethylene glycol groups. In certain embodiments, such functionalized antibody is derived by treating an antibody having at least one glutamine residue, for example, a C-terminal TGase recognition tag, with a primary amine compound in the presence of the enzyme transglutaminase. In certain embodiments, such functionalized antibody is derived by treating an antibody having at least one glutamine residue, for example, heavy chain Gln295, with a primary amine compound in the presence of the enzyme transglutaminase. In certain embodiments, such functionalized antibody is derived by treating an antibody having at least one glutamine residue, for example, heavy chain Gln297, with a primary amine compound in the presence of the enzyme transglutaminase. Such antibodies include Asn297Gln (N297Q) mutants. In certain embodiments, such functionalized antibody is derived by treating an antibody having at least two glutamine residues, for example, heavy chain Gln295 and heavy chain Gln297, with a primary amine compound in the presence of the enzyme transglutaminase. Such antibodies include Asn297Gln (N297Q) mutants. In certain embodiments, the antibody has two heavy chains as described in this paragraph for a total of two or a total of four glutamine residues.

[00199] In one embodiment, the functionalized antibody or antigen binding molecule includes an antibody heavy chain and further includes a peptide tag at the C-terminus of the antibody heavy chain. In one embodiment, the functionalized antibody or antigen binding molecule includes an antibody heavy chain and further includes a peptide tag at the C-terminus of the antibody heavy chain, wherein the peptide tag is the pentapeptide sequence LLQGA. In embodiment, the functionalized antibody or antigen binding molecule includes two antibody heavy chains and further includes a peptide tag at the C-terminus of each antibody heavy chain. In one embodiment, the functionalized antibody or antigen binding molecule includes two antibody heavy chains and further includes a peptide tag at the C-terminus of each antibody heavy chain, wherein the peptide tag is the pentapeptide sequence LLQGA.

[00200] In certain embodiments, the antibody comprises two glutamine residues, one in each heavy chain. In particular embodiments, the antibody comprises a Q295 residue in each heavy chain. In further embodiments, the antibody comprises one, two, three, four, five, six, seven, eight, or more glutamine residues. These glutamine residues can be in heavy chains, light chains, or in both heavy chains and light chains. These glutamine residues can be wild-type residues, or engineered residues. The antibodies can be prepared according to standard techniques.

[00201] Those of skill will recognize that antibodies are often glycosylated at residue N297, near residue Q295 in a heavy chain sequence. Glycosylation at residue N297 can interfere with a transglutaminase at residue Q295 (Dennler et at, supra). Accordingly, in advantageous embodiments, the antibody is not glycosylated. In certain embodiments, the antibody is deglycoslated or aglycosylated. In particular embodiments, an antibody heavy chain has an N297 mutation. Alternatively stated, the antibody is mutated to no longer have an asparagine residue at position 297. In particular embodiments, an antibody heavy chain has an N297Q mutation. Such an antibody can be prepared by site-directed mutagenesis to remove or disable a glycosylation sequence or by site-directed mutagenesis to insert a glutamine residue at a site apart from any interfering glycosylation site or any other interfering structure. Such an antibody also can be isolated from natural or artificial sources.

[00202] The transglutaminase can be any transglutaminase deemed suitable by those of skill in the art. In certain embodiments, the transglutaminase is an enzyme that catalyzes the formation of an isopeptide bond between a free amino group on the linker-payload compound and the acyl group on the side chain of a glutamine residue. Transglutaminase is also known as protein-glutamine-y-glutamyl transferase. In particular embodiments, the transglutaminase is classified as EC 2.3.2.13. The transglutaminase can be from any source deemed suitable. In certain embodiments, the transglutaminase is microbial. Useful transglutaminases have been isolated from Streptomyces mobaraense, Streptomyces cinnamoneum, Streptomyces griseo- carneum, Streptomyces lavendulae, and Bacillus subtilis. Non-microbial transglutaminases, including mammalian transglutaminases, can also be used, e.g., a non-microbial transglutaminase in combination with a cofactor. In certain embodiments, the transglutaminase can be produced by any technique or obtained from any source deemed suitable by the practitioner of skill. In particular embodiments, the transglutaminase is obtained from a commercial source.

Pharmaceutical Compositions and Methods of Treatment

[00203] Provided herein are methods of treating and preventing diseases, conditions, or disorders comprising administering a therapeutically or prophylactically effective amount or one or more of the compounds disclosed herein, for example, one or more of the compounds of a formula provided herein. Diseases, disorders, and/or conditions include, but are not limited to, those associated with viral infections as described herein.

[00204] The compounds described herein can be administered alone or together with one or more additional (supplementary) therapeutic agents. The one or more additional therapeutic agents can be administered just prior to, concurrent with, or shortly after the administration of the compounds described herein. The present disclosure also includes pharmaceutical compositions comprising any of the compounds described herein in combination with one or more additional therapeutic agents, and methods of treatment comprising administering such combinations to subjects in need thereof.

[00205] Suitable additional therapeutic agents include, but are not limited to: an anti-viral drug such as a second antiviral compound or payload, an autoimmune therapeutic agent, a hormone, a biologic, or a monoclonal antibody. In some embodiments, the supplementary therapeutic agent can be selected from the group consisting of: an anti-viral drug, an anti inflammatory drug (e.g., a corticosteroid or non-steroidal anti-inflammatory drug), an antibody that binds specifically to influenza HA, a vaccine for influenza, a dietary supplement (e.g., and anti-oxidant), and a palliative therapy to treat an influenza infection. In one embodiment, the anti-inflammatory drug is selected from the group consisting of corticosteroids and non steroidal anti-inflammatory drugs. In one embodiment, the dietary supplement is an anti oxidant. Suitable therapeutic agents also include, but are not limited to, any pharmaceutically acceptable salts or derivatives of an antiviral compound or payload set forth herein. In some embodiments, the supplementary therapeutic agent is administered via a different route of administration as an antibody-drug conjugate, compound, or pharmaceutical composition described herein. For example, a supplementary therapeutic agent can be administered orally. An exemplary anti-viral drug to be administered as a supplementary therapeutic agent is oseltamivir. In some embodiments, the oseltamivir is administered prior to administration of the antibody-drug conjugate, compound, or pharmaceutical composition. In some embodiments, the oseltamivir is administered concurrently with the antibody-drug conjugate, compound, or pharmaceutical composition. In some embodiments, the oseltamivir is administered after administration of the antibody-drug conjugate, compound, or pharmaceutical composition. In some embodiments, the anti-viral drug is an anti-influenza A drug or an anti -influenza B drug (e.g., an antibody or antigen-binding portion thereof), such as an antibody that binds specifically to influenza A HA or an antibody that binds specifically to influenza B HA.

[00206] In some embodiments of the methods described herein, multiple doses of a compound described herein (or a pharmaceutical composition comprising a combination of a compound described herein and any of the additional therapeutic agents mentioned herein) may be administered to a subject over a defined time course. The methods according to this embodiment of the disclosure comprise sequentially administering to a subject multiple doses of a compound described herein. The present disclosure includes methods which comprise sequentially administering to the patient a single initial dose of a compound described herein, followed by one or more secondary doses of the compound, and optionally followed by one or more tertiary doses of the compound. Exemplary doses of a compound of the present disclosure include, but are not limited to, 50 mg/kg, 49 mg/kg, 48 mg/kg, 47 mg/kg, 46 mg/kg, 45 mg/kg, 44 mg/kg, 43 mg/kg, 42 mg/kg, 41 mg/kg, 40 mg/kg, 39 mg/kg, 38 mg/kg, 37 mg/kg, 36 mg/kg, 35 mg/kg, 34 mg/kg, 33 mg/kg, 32 mg/kg, 31 mg/kg, 30 mg/kg, 29 mg/kg, 28 mg/kg, 27 mg/kg, 26 mg/kg, 25 mg/kg, 24 mg/kg, 23 mg/kg, 22 mg/kg, 21 mg/kg, 20 mg/kg, 19 mg/kg, 18 mg/kg, 17 mg/kg, 16 mg/kg, 15 mg/kg, 14 mg/kg, 13 mg/kg, 12 mg/kg, 11 mg/kg, 10 mg/kg, 9 mg/kg, 8 mg/kg, 7 mg/kg, 6 mg/kg, 5 mg/kg, 4 mg/kg, 3 mg/kg, 2 mg/kg, 1 mg/kg, 0.9 mg/kg, 0.8 mg/kg, 0.7 mg/kg, 0.6 mg/kg, 0.5 mg/kg, 0.4 mg/kg, 0.3 mg/kg, 0.2 mg/kg, 0.1 mg/kg, and 0.05 mg/kg.

[00207] In certain embodiments, the amount of the compound included in the initial, secondary, and/or tertiary doses varies from one another (e.g., adjusted up or down as appropriate) during the course of treatment. In certain embodiments, two or more (e.g., 2, 3, 4, or 5) doses are administered at the beginning of the treatment regimen as “loading doses” followed by subsequent doses that are administered on a less frequent basis (e.g., “maintenance doses”).

[00208] In certain exemplary embodiments of the present disclosure, each secondary and/or tertiary dose is administered 1 to 26 (e.g., 1, 1 ½, 2, 2 ½, 3, 3 ½, 4, 4 ½, 5, 5 ½, 6, 6 ½, 7, 7 ½, 8, 8½, 9, 9½, 10, 10½, 11, 11½, 12, 12½, 13, 13½, 14, 14½, 15, 15½, 16, 16½, 17, 17½, 18, 18½, 19, 19½, 20, 20½, 21, 21 ½, 22, 22 ½, 23, 23½, 24, 24½, 25, 25½, 26, 26½, or more) weeks after the immediately preceding dose.

[00209] The methods according to this embodiment of the disclosure may comprise administering to a patient any number of secondary and/or tertiary doses of the compound. For example, in certain embodiments, only a single secondary dose is administered to the patient. In other embodiments, two or more (e.g., 2, 3, 4, 5, 6, 7, 8, or more) secondary doses are administered to the patient. Likewise, in certain embodiments, only a single tertiary dose is administered to the patient. In other embodiments, two or more (e.g., 2, 3, 4, 5, 6, 7, 8, or more) tertiary doses are administered to the patient. The administration regimen may be carried out indefinitely over the lifetime of a particular subject, or until such treatment is no longer therapeutically needed or advantageous. [00210] In embodiments involving multiple secondary doses, each secondary dose may be administered at the same frequency as the other secondary doses. For example, each secondary dose may be administered to the patient 1 to 2 weeks or 1 to 2 months after the immediately preceding dose. Similarly, in embodiments involving multiple tertiary doses, each tertiary dose may be administered at the same frequency as the other tertiary doses. For example, each tertiary dose may be administered to the patient 2 to 12 weeks after the immediately preceding dose. In certain embodiments of the disclosure, the frequency at which the secondary and/or tertiary doses are administered to a patient can vary over the course of the treatment regimen. The frequency of administration may also be adjusted during the course of treatment by a physician depending on the needs of the individual patient following clinical examination.

[00211] The present disclosure includes administration regimens in which 2 to 6 loading doses are administered to a patient at a first frequency (e.g., once a week, once every two weeks, once every three weeks, once a month, once every two months, etc.), followed by administration of two or more maintenance doses to the patient on a less frequent basis. For example, according to this embodiment of the disclosure, if the loading doses are administered at a frequency of once a month, then the maintenance doses may be administered to the patient once every six weeks, once every two months, once every three months, etc.

[00212] The present disclosure includes pharmaceutical compositions of the compounds and/or conjugates described herein, e.g., antibody-drug conjugates of the compounds Formulae 101-403, e.g., compositions comprising a compound described herein, a salt, stereoisomer, regioisomer, polymorph thereof, and a pharmaceutically acceptable carrier, diluent, and/or excipient. Examples of suitable carriers, diluents and excipients include, but are not limited to, buffers for maintenance of proper composition pH (e.g., citrate buffers, succinate buffers, acetate buffers, phosphate buffers, lactate buffers, oxalate buffers, and the like), carrier proteins (e.g., human serum albumin), saline, polyols (e.g., trehalose, sucrose, xylitol, sorbitol, and the like), surfactants (e.g., polysorbate 20, polysorbate 80, polyoxolate, and the like), antimicrobials, and antioxidants.

[00213] In certain embodiments, the compounds or payloads, linker-payloads, ADCs, or compositions thereof may be provided via alternate routes of administration. In certain embodiments, the route of administration for the composition(s) is selected from the group of subcutaneous, intradermal, intramuscular, oral, intravenous, intraperitoneal, inhalation, and intranasal. In one embodiment, the route of administration for the composition(s) is oral. In one embodiment, the route of administration for the composition(s) is intravenous. In one embodiment, the route of administration for the composition(s) is intraperitoneal. In one embodiment, the route of administration for the composition(s) is inhalation. In one embodiment, the route of administration for the composition(s) is intranasal.

[00214] In some examples, set forth herein methods for treatment, prophylaxis, reduction, or inhibition of a disease, disorder, or condition associated with an infection in a subject comprising administering to the subject an effective amount or a therapeutically effective amount of a compound of Formulae 101-403, a linker-payload described herein, and/or an ADC described herein, combinations thereof, or a pharmaceutical composition thereof. In some embodiments, the infection is a viral infection. In some embodiments, the infection is influenza virus infection. In some embodiments, the infection is influenza A virus infection. In some embodiments, the infection is influenza B virus. In some embodiments, the infection is influenza A virus infection and influenza B virus infection. In certain embodiments, side effects associated with administration of an unconjugated payload to the subject are reduced when compared to administration of the conjugated payload or ADC to a comparable subject.

[00215] The compounds disclosed herein can also be used for treatment, prophylaxis, reduction, or inhibition of an influenza infection in a subject comprising administering to the subject an effective amount of an antibody-drug conjugate, compound, or pharmaceutical composition described herein. In some embodiments, the influenza infection is caused by influenza A virus infection. In some embodiments, the influenza infection is caused by influenza A Group 1 virus. In some embodiments, the influenza infection is caused by influenza A HI virus. In some embodiments, the influenza infection is caused by influenza A Group 2 virus. In some embodiments, the influenza infection is caused by influenza A H3 virus. In some embodiments, the influenza infection is caused by an unknown or undetermined influenza virus. In some embodiments, the influenza infection is caused by influenza B virus infection. In some embodiments, the influenza infection is caused by influenza A virus infection and influenza B virus infection. In some embodiments, the influenza infection is caused by influenza A virus infection, influenza A Group 1 infection, influenza A HI infection, influenza A Group 2 infection, influenza A H3 infection, an unknown or undetermined influenza virus, influenza B virus infection, or any combination thereof. EXAMPLES

[00216] Provided herein are VX-787 derivatives, protein conjugates thereof, and methods for treating diseases, disorders, and conditions including administering VX-787 derivatives and conjugates thereof.

Example 1: Linker-payload synthesis

[00217] Payloads, linker-payloads, and conjugates were synthesized as indicated below. All the solvents used were used as is and purchased either from Sigma Aldrich or Fisher Scientific. 'H spectra were recorded on Varian Inova 300 MHz and 500 MHz NMR instruments. The chemical shifts (d) were reported in ppm with respect to the NMR solvents used for analysis and were reported as s - singlet, d - doublet, t - triplet, q - quartet, dd - doublet of doublet, dt - doublet of triplet, dq - doublet of quartet, and m - multiple! Coupling constants (./) were reported in hertz (Hz). Chromatographic purities were determined on an Agilent 1100, 1260 Infinity with 6130 Quadrupole LC/MS, or 1200 Series LC/MS systems using Chromolith ® FastGradient RP-18e analytical columns (50 c 2 mm, Merck KGaA, P/N 1.52007.0001) and the following analytical HPLC method: injection volume 2-10 mL; flow rate 1 mL/min; 5-95% acetonitrile in water over 4 min; Agilent diode array detector at l = 254 nm; room temperature. Low resolution mass spectrometry was performed on Agilent systems using electrospray ionization sources and analyzed with either single quadrupole or ion trap mass detectors.

1.1 Scheme 1

[00218] Compound 11 was prepared following the literature procedure from ACS Medicinal

Chemistry Letters 2017, 8, 261-265.

[00219] Compound 13: To a solution of compound 11 (50 mg, 0.096 mmol) and compound 12 (25 mg, 0.08 mmol) in 2-methyl THF (1 mL) and water (0.2 mL), was added K3PO4 (3 mg, 0.0144 mmol). The mixture was purged with argon for 10 mins, then X-phos (4.5 mg, 0.0096 mmol) and Pd 2 (dba) 3 (1.8 mg, 0.002 mmol) were added, and the reaction was heated to 110 °C for 9 h. The reaction was diluted with ethyl acetate (2 mL) and filtered through a plug of a celite, then was concentrated. The residue was purified on a 24 g silica gold column using hexanes/ethyl acetate. Pure fractions were combined and concentrated to obtain compound 13 as an off-white solid (10 mg, 20%). MS (ESI, pos.): calc’d for C33H35F2N5O6S, 667.2; found 668.2 (M+H).

[00220] Compound 15: To a solution of compound 13 (10 mg, 0.015 mmol) in MeOH (2 mL), was added TFA (1.5 mL) and the mixture was stirred at room temperature for 4 h. Volatiles were removed in vacuo to obtain compound 14, which was dissolved in acetonitrile (1 mL). To the resulting solution was added 4 M HC1 in dioxane (22.5 pL, 0.09 mmol) and the mixture was heated to 70 °C for 30 min. Volatiles were again removed under reduced pressure and the residue was dissolved in 1:1 THF/MeOH (1 mL). To this solution was added 2 N aqueous NaOH (0.2 mL) and the reaction was heated to 30 °C for 4 h. The reaction was acidified to pH 4-5 with 2 N HC1. The aqueous layer was discarded, and the organic layer was concentrated to dryness then purified on 30 g C18 Aq column using 5-95% MeCN/H 2 0 (both having 0.05% AcOH). Pure fractions were combined, frozen and lyophilized to obtain compound 15 (3.5 mg, 54%) as a fluffy off-white solid. MS (ESI, pos.): calc’d for C21H21F2N5O3, 429.2; found 430.1 (M+H). ¾ NMR (500 MHz; DMSO-de): d 12.15 (s, 1H), 8.51 (dd, J= 10.1, 2.9 Hz, 1H), 8.19-8.17 (m, 2H), 7.55-7.54 (m, 1H), 5.05 (q, J= 15.5 Hz, 2H), 4.72-4.70 (m, 1H), 2.76 (s, 1H), 2.00 (d, J= 0.9 Hz, 1H), 1.90-1.86 (m, 1H), 1.81-1.78 (m, 1H), 1.78-1.73 (m, 2H), 1.59-1.46 (m, 3H), 1.43-1.33 (m, 2H).

1.2 Scheme 2 [00221] Compounds 19b and 19c were prepared by following the procedures described in European Journal of Medicinal Chemistry, 2019, 162, 249-265. Hydroxylamine hydrochloride was free based prior to use by KOH in MeOH.

[00222] General procedure to prepare hydroxamic acid derivatives from the corresponding carboxylic acids: Compound 20a: To a mixture of VX-787 (19a, 10 mg, 0.025 mmol) in anhydrous THF at 0 °C, N-methylmorpholine (NMM) (8 pL, 0.075 mmol) and ethyl chloroformate (3 pL, 0.03 mmol) were added. After stirring at 0 °C for 10 min freshly prepared hydroxylamine solution in MeOH (20 pL, large excess) was added. The reaction was stirred at 0 °C for 5 min then was warmed to the ambient temperature and volatiles were removed under reduced pressure. The residue was purified on a 15.5 g C18 Aq column using 5-95% MeCNTHO (both with 0.05% HO Ac). Pure fractions were combined, frozen and lyophilized to obtain compound 20a (6.2 mg, 60%) as a fluffy white solid. MS (ESI, pos.): calc’d for C20H20F2N6O2, 414.2; found 415.2 (M+H). ¾ NMR (500 MHz; DMSO-de): d 12.22 (s, 1H), 10.40 (s, 1H), 8.66 (s, 1H), 8.54 (dd, J= 9.8, 2.8 Hz, 1H), 8.26 (s, 1H), 8.19 (s, 1H), 8.12 (d, J = 4.0 Hz, 1H), 7.47 (d, J= 7.3 Hz, 1H), 4.82 (t, J= 7.1 Hz, 1H), 1.88-1.68 (m, 7H), 1.60-1.54 (m, 1H), 1.48-1.43 (m, 1H), 1.35 (t, J= 11.6 Hz, 2H).

[00223] Compound 20b was prepared from compound 19b using the same general procedure. Yield = 28% as a fluffy off-white solid. MS (ESI, pos.): calc’d for C22H21FN6O2S, 452.1; found 453.2 (M+H). ¾ NMR (500 MHz; DMSO-de): d 12.24 (s, 1H), 10.44 (s, 1H), 8.69-8.66 (m, 2H), 8.32 (d, .7= 1.7 Hz, 1H), 8.26 (d, .7= 1.4 Hz, 1H), 7.71 (d, .7= 6.0 Hz, 1H), 7.54 (d, J= 7.2 Hz, 1H), 7.41 (d, .7= 5.9 Hz, 1H), 5.01 (t, .7= 6.9 Hz, 1H), 1.96-1.73 (m, 7H), 1.60-1.59 (m, 1H), 1.51 (d, J= 6.0 Hz, 1H), 1.41-1.37 (m, 2H).

[00224] Compound 20c was prepared from compound 19c using the same general procedure. Yield = 44% as a pale yellow solid. MS (ESI, pos.): calc’d for C23H24FN7O2, 449.20; found 450.15 (M+H). ¾ NMR (500 MHz; CD3OD): d 8.82 (dd,/ = 9.6, 2.6 Hz, 1H), 8.82 (dd, / = 9.6, 2.6 Hz, 1H), 8.33 (s, 1H), 8.14 (s, 1H), 6.97 (d,/ = 3.1 Hz, 1H), 6.60 (d,/ = 3.0 Hz, 1H), 4.99-4.97 (m, 1H), 3.84 (s, 3H), 2.45 (d,/ = 6.5 Hz, 1H), 2.05-1.78 (m, 6H), 1.72-1.61 (m, 2H), 1.56-1.45 (m, 2H). 1.3 Scheme 3

[00225] Compound 22: To a solution of Fmoc-PEG-8-Val-Cit-PAB-OH (21, 120 mg, 0.117 mmol) in anhydrous DCM (5 mL), was added thionyl chloride (10 pL, 0.128 mmol) and the reaction was stirred for 20 min. The reaction was concentrated, dissolved in DMSO (1 mL), and purified on Teledyne ISCO using a 30 g Cl 8 Aq column with 5-95% MeCN/FLO (both having 0.05% AcOH). Pure fractions were combined, frozen and lyophilized to obtain compound 22 (90 mg, 74%) as a fluffy off-white solid. MS (ESI, pos.): calc’d for C52H75CIN6O14, 1042.5; found 1043.4 (M+H).

[00226] Compound 23: To a solution of compound 20a (8 mg, 0.019 mmol) and compound 22 (20 mg, 0.019 mmol) in anhydrous acetonitrile (4 mL), were added sodium iodide (5.8 mg, 0.038 mmol) and potassium carbonate (5.2 mg, 0.038 mmol). The reaction was heated to 50 °C for 4 h then cooled to room temperature and concentrated under reduced pressure. The residue was purified on a Teledyne ISCO 50 g Cl 8 Aq column with gradient elution of 5-95% MeCN/ThO (both having 0.05% AcOH). Pure fractions were combined, frozen and lyophilized to obtain compound 23 (10 mg, 37%) as a fluffy off-white solid. MS (ESI, pos.): calc’d for C22H94F2N12O16, 1420.7; found 1421.6 (M+H).

[00227] Compound 24: To a solution of compound 23 (10 mg, 0.007 mmol) in DMF (0.6 mL) was added a 5% piperidine solution in DMF (0.3 mL) and the reaction was stirred for 45 min purified on a Gemini 30 x 150 mm column using gradient elution 5-95% MeCN/FEO (both having 0.05% AcOH). Pure fractions were combined, frozen and lyophilized to obtain compound 24 (5.3 mg, 63%) as a fluffy off-white solid. MS (ESI, pos.): calc’d for C57H84F2N12O14, 1198.6; found 1199.6 (M+H). ¾ NMR (500 MHz; DMSO-de): d 10.98 (s, 1H), 9.97 (s, 1H), 8.53 (dd, J= 9.8, 2.8 Hz, 1H), 8.27-8.23 (m, 2H), 8.12 (dd, J= 15.1, 5.7 Hz, 2H), 7.85 (d, J= 8.6 Hz, 1H), 7.55 (d, J= 8.4 Hz, 2H), 7.46 (d, J= 7.5 Hz, 1H), 7.22 (d, J = 8.4 Hz, 2H), 5.97 (t, J = 5.7 Hz, 1H), 5.39 (s, 2H), 4.82 (t, J= 7.0 Hz, 1H), 4.65 (q, J= 8.4 Hz, 2H), 4.36 (q, J= 6.8 Hz, 1H), 4.22 (dd, J= 8.7, 6.8 Hz, 1H), 3.61-3.44 (m, 32H), 3.06-2.86 (m, 6H), 1.96-1.90 (m, 4H), 1.87-1.77 (m, 3H), 1.72-1.67 (m, 4H), 1.58-1.57 (m, 3H), 1.47-1.34 (m, 5H), 0.83 (dd, J= 16.1, 6.8 Hz, 6H).

1.4 Scheme 4

[00228] Compound 27: To a solution of Fmoc-Cap-NHS (25, 91 mg, 0.2 mmol) and Gly- Gly-Phe-OH (26, 56 mg, 0.2 mmol) in DMF (1 mL), was added DIEA (70 pL, 0.4 mmol). After stirring for 30 min the reaction mixture was injected onto a Teledyne ISCO 50 g C18 Aq column and eluted with 5-95% MeCN/FhO gradient elution (both having 0.05% AcOH). Pure fractions were combined, frozen and lyophilized to obtain compound 27 (93 mg, 76%) as a fluffy off-white solid. MS (ESI, pos.): calc’d for C34H38N4O7, 614.3; found 615.3 (M+H).

1.5 Scheme 5

[00229] (2-(((allyloxy)carbonyl)amino)acetamido)methyl acetate (30) was prepared following the procedure from Tetrahedron 2018, 74, 1951-1956.

[00230] Compound 29: To a solution of compound 20a (4.2 mg, 0.01 mmol) and (2- (((allyloxy)carbonyl)amino)acetamido)methyl acetate (30, 2.3 mg, 0.01 mmol) in anhydrous THF (0.5 mL) at 0 °C, was added /-BuOK (10 pL, 0.01 mmol, 1 M solution in THF) and the reaction was stirred at 0 °C for 30 min. The reaction was quenched with aqueous saturated ammonium chloride solution (0.5 mL). Volatiles were removed in vacuo and the residue was purified on a Teledyne ISCO 30 g C18 Aq column using gradient elution 5-95% MeCNTUO (both having 0.05% AcOH). Pure fractions containing the desired product were combined, frozen and lyophilized to obtain compound 29 (3 mg, 51%) as a fluffy off-white solid. MS (ESI, pos.): calc’d for C27H30F2N8O5, 584.2; found 585.2 (M+H).

[00231] Compound 31: To a solution of compound 29 (3 mg, 0.005 mmol) in THF (1 mL), were added Pd(PPh3)4 (0.6 mg, 0.0005 mmol) and phenylsilane (1 pL, 0.0076 mmol). The reaction was stirred for 20 min at room temperature then filtered through a pad of celite. The filtrate was concentrated and the resulting compound 30 was combined with Fmoc-Cap-Gly- Gly-Phe-OH (27, 4 mg, 0.0055 mmol), HATU (3 mg, 0.0076 mmol) and HOAt (0.7 mg, 0.005 mmol) in THF (1 mL) and DMF (0.2 mL). To this mixture was added DIEA (3 pL, 0.015 mmol) and the resulting solution was stirred for 1 h at the room temperature. Volatiles were removed under reduced pressure and the residue was purified on a Teledyne ISCO 30 g Cl 8 Aq column using gradient elution 5-95% MeCN/thO (both having 0.05% AcOH). Pure fractions were combined, frozen and lyophilized to obtain compound 31 (2 mg, 37% over two steps) as a fluffy off-white solid. MS (ESI, pos.): calc’d for C57H62F2N12O9, 1096.5; found

1097.4 (M+H).

[00232] Compound 32: To a solution of compound 31 (6 mg, 0.005 mmol) in DMF (0.6 mL), was added a 5% piperidine solution in DMF (0.3 mL). The reaction was stirred for 45 min at room temperature then injected onto a Gemini 30 xl50 mm column and eluted with 5- 95% MeCN/FhO (both having 0.05% AcOH). Pure fractions were combined, frozen and lyophilized to obtain compound 32 (2.6 mg, 54%) as a fluffy off-white solid. MS (ESI, pos.): calc’d for C42H52F2N12O7, 874.4; found 875.4 (M+H). 'HNMR (500 MHz; DMSO-d 6 ): d 8.53- 8.50 (m, 1H), 8.26 (s, 1H), 8.19 (d, J = 7.0 Hz, 1H), 8.11 (d, J = 4.0 Hz, 1H), 7.25-7.20 (m, 4H), 7.18-7.15 (m, 1H), 4.75-4.71 (m, 2H), 4.50-4.48 (m, 1H), 3.74 (dt, J= 17.9, 9.5 Hz, 3H), 3.68-3.63 (m, 2H), 3.09-2.94 (m, 3H), 279-2.74 (m, 1H), 2.12 (t, J= 8.9 Hz, 2H), 1.92 (d, J =

5.4 Hz, 1H), 1.83-1.70 (m, 9H), 1.57 (t , J= 10.5 Hz, 1H), 1.48-1.45 (m, 3H), 1.34 (t, J= 8.8 Hz, 4H), 1.24 (dd, J= 14.2, 8.2 Hz, 2H).

1.6 Scheme 6

[00233] Compound 35: To a solution of Fmoc-Val-Cit-OH (33, 497 mg, 1.0 mmol) and 1- (4-aminophenyl)ethan-l-ol (34, 274 mg, 2.0 mmol) in DCM (4.5 mL) and MeOH (2 mL), was added EEDQ (495 mg, 2.0 mmol) and the mixture was stirred for 1 h at room temperature. The reaction became a gum, to which was added DCM (4.5 mL) and MeOH (2 mL). The resulting mixture was stirred overnight then solvents were removed under reduced pressure. The residue was washed sequentially with diethyl ether (5 mL), ethyl acetate (5 mL) and diethyl ether (5 mL), then dried on high vacuum to obtain compound 35 (585 mg, 95%). MS (ESI, pos.): calc’d for C 34 H 41 N 5 O 6 , 615.3; found 616.3 (M+H).

[00234] Compound 36: To a solution of compound 35 (150 mg, 0.244 mmol) in DMF (1 mL), was added 5% piperidine solution in DMF (1 mL). The reaction was stirred for 45 mins at the room temperature then injected onto a Teledyne ISCO 50 g Cl 8 Aq column and eluted with 5-95% MeCN/FLO (both having 0.05% AcOH). Pure fractions were combined, frozen and lyophilized to obtain compound 36 (103 mg, 94%) as its acetic acid salt. MS (ESI, pos.): calc’d for C19H31N5O4, 393.2; found 394.3.

[00235] Compound 37: To a solution of compound 36 (45 mg, 0.1 mmol) and Fmoc-amido PEG8-NHS ester (91 mg, 0.12 mmol) in DMF was added DIEA (21 pL, 0.12 mmol) and the reaction was stirred for 40 min at room temperature. The mixture was injected onto a Teledyne ISCO 50 g C18 Aq column and eluted with 5-95% MeCN/FLO (both having 0.05% AcOH). Pure fractions were combined, frozen and lyophilized to obtain compound 37 (103 mg, 99%). MS (ESI, pos.): calc’d for CssHvsNeOis, 1038.6; found 1039.5 (M+H).

[00236] Compound 38: To a DCM (4 mL) solution of compound 37 (25 mg, 0.024 mmol) were added VX-787 (19a, 11.5 mg, 0.028 mmol), EDCI (7.0 mg, 0.036 mmol) andDMAP (1.2 mg, 0.009 mmol), and the reaction was stirred overnight. The reaction was concentrated and injected onto a Teledyne ISCO 50 g C18 Aq column and eluted with 5-95% MeCN/H 2 0 (both having 0.05% AcOH). Pure fractions were combined, frozen and lyophilized to obtain compound 38 (9.8 mg, 29%). MS (ESI, pos.): calc’d for C73H95F2N11O16, 1419.7; found 1420.7 (M+H).

[00237] Compound 39: To a solution of compound 38 (9.8 mg, 0.007 mmol) in DMF (0.8 mL) was added a 5% solution of piperidine in DMF (0.5 mL) and the reaction was stirred for 40 min at room temperature. The product was purified on a Gemini 30 x 150 mm column using gradient elution 5-95% MeCNTLO (both having 0.05% AcOH). Pure fractions were combined, frozen and lyophilized to obtain a diastereomeric mixture of compound 39 (2.6 mg, 54%) as a fluffy off-white solid. MS (ESI, pos.): calc’d for C58H85F2N11O14, 1197.6; found 1198.6 (M+H). 'HNMR (500 MHz; DMSO-d 6 ): d 9.95 (d, J= 9.9 Hz, 1H), 8.49-8.46 (m, 1H), 8.26-8.25 (m, 1H), 8.18-8.12 (m, 3H), 7.89 (d, J= 8.6 Hz, 1H), 7.61 (t, J = 7.7 Hz, 1H), 7.48 (dd , J = 12.1, 8.7 Hz, 2H), 7.20 (t, J = 9.3 Hz, 1H), 6.02 (dt, J= 1.2, 0.6 Hz, 1H), 5.77-5.73 (m, 1H), 5.41 (s, 2H), 4.75 (q, J= 6.8 Hz, 1H), 4.36-4.31 (m, 1H), 4.20 (m, 1H), 3.57 (m, 3H),

3.48 (m, 3 OH), 2.63 (t, J= 5.8 Hz, 4H), 2.37 (m, 4H), 1.97-1.89 (m, 4H), 1.72 (m, 6H), 1.59-

1.48 (m, 4H), 1.35 (dd, J= 25.5, 6.5 Hz, 6H), 0.83 (ddd, J= 14.4, 6.7, 3.5 Hz, 6H).

1.7 Scheme 7

[00238] Compound 41: To a suspension of Alloc-Val-Ala-PAB-OH (40, 50 mg, 0.132 mmol) in anhydrous DCM (1.5 mL) under argon was added thionyl chloride (85 pL, 1.17 mmol). The resulting suspension was stirred at room temperature for 3.5 h then was concentrated in vacuo. Anhydrous DCM (1 mL) was added to the residue and the mixture was concentrated in vacuo again to afford compound 41 as a white solid, which was used without purification. MS (ESI, pos.): calc’d for C19H26CIN3O4, 395.2; found 396.2 (M+H).

1.8 Scheme 8

[00239] Compound 42: A suspension of compound 19a (30 mg, 0.075 mmol) in anhydrous MeOH (0.5 mL) was treated with cone. H2SO4 (2 pL) and stirred for 2 days, at which time the reaction was not complete. Additional MeOH (0.3 mL) and H2SO4 (10 pL) were added and the reaction was heated at 50 °C for 2 days, at which time the reaction had become a solution and LCMS indicated complete consumption of starting acid. The reaction was cooled to room temperature and partitioned between ethyl acetate (10 mL) and dilute aqueous NaHC0 3 (5 mL). The aqueous later was extracted with two 5-mL portions of ethyl acetate. The combined organic layers were washed with brine (5 mL), then were dried over Na2SC>4, filtered, and concentrated to afford compound 42 (31 mg, quant.) as a pale-yellow solid, which was used without purification. MS (ESI, pos.): calc’d for C 21 H 21 F 2 N 5 O 2 , 413.2; found 414.5 (M+H).

[00240] Compound 43: To a mixture of compound 41 (4 mg, 0.0109 mmol) and compound 42 (5 mg, 0.0121 mmol) in anhydrous DMA (0.1 mL) were added powdered K2CO3 (5 mg, 0.0362 mmol) and Nal (2 mg, 0.0133 mmol). The resulting yellow mixture was stirred at room temperature overnight then was chromatographed on a 5.5 g C18Aq column, eluting with 5- 100% MeCN in H2O (both with 0.05% HOAc). Fractions containing pure product were combined and lyophilized to afford compound 43 (4 mg, 48%) as a white fluffy solid. MS calc’d for C 4 oH 46 F 2 N 8 0 6 , 772.4; found 773.4 (M+H).

[00241] Compound 45: To a solution of compound 43 (14 mg, 0.0181 mmol) in anhydrous DCM (1.5 mL) under Argon was added Pd(Ph3P)4 (7 mg, 0.00606 mmol) and PhSflE (4.5 pL, 0.0362 mmol). After stirring at room temperature for 30 min the reaction was concentrated in vacuo. The residue was dissolved in anhydrous DMA (200 pL) and a solution of Fmoc-amido- PEG8-NHS ester (20 mg, 0.063 mmol) in DMA (400 pL) was added. To the resulting solution was added DIEA and the reaction was stirred at room temperature for 1 h then chromatographed on a 5.5 g C18Aq column, eluting with MeCN in H2O (both with 0.05% HOAc). Pure fractions were combined and lyophilized to afford compound 45 (11 mg, 47% for 2 steps) as a white fluffy solid. MS calc’d for C70H89F2N9O15, 1333.6; found 1334.6 (M+H).

[00242] Compound 46: A mixture of compound 45 (13 mg, 0.00974 mmol) in a 0.3 M solution of NaOH in 80% MeOH-H20 (0.65 mL) was stirred at room temperature overnight. The reaction was brought to pH 6 with 0.2 N HC1. Volatiles were removed under reduced pressure and the residue was dissolved in DMSO and purified by chromatography on a 5.5 g Cl 8Aq column, eluting with 10-60% MeCN in H2O (with 0.05% HOAc in both). Pure fractions were combined and lyophilized to afford compound 46 (4 mg, 38%) as a white fluffy solid. MS (ESI, pos.): calc’d for C54H77F2N9O13, 1097.6; found 1098.5 (M+H). ¾ NMR (500 MHz, DMSO-de) d 9.91 (s, 1H), 8.53 (dd, J = 9.5, 2.5 Hz, 1H), 8.32-8.30 (m, 1H), 8.30 (s, 1H), 8.19 (br d, J= 6.5 Hz, 1H), 8.08 (d, .7= 3.5 Hz, 1H), 7.88 (d, J= 8.5 Hz, 1H), 7.53 (d, J= 8.5 Hz, 2H), 7.41 (d, J= 8.5 Hz, 1H), 7.24 (d, J= 8.5 Hz, 2H), 5.49 (d, J= 15 Hz, 1H), 4.43 (d, J= 15 Hz, 1H), 4.72-4.68 (m, 1H), 4.38-4.30 (m, 1H), 4.22-4.11 (m, 2H), 3.58-3.44 (m, 28H, partially obscured by residual H 2 0), 2.68 (t, J = 5.5 Hz, 2H), 2.61-2.57 (m, 2H), 2.46-2.33 (m, 2H) 2.02- 1.86 (m, 4H), 1.80-1.74 (m, 1H), 1.73-1.64 (m, 2H), 1.60-1.51 (m, 2H), 1.50-1.42 (m, 1H), 1.41-1.29 (m, 3H), 1.26 (d, J= 7 Hz, 3H), 1.24-1.19 (m, 1H), 0.84 (d, J= 7 Hz, 3H), 0.80 (d, J = 7 Hz, 3H).

1.9 Scheme 9

[00243] Compound 48: To a mixture of amino-PEG8 acid (47, 53 mg, 0.120 mmol) and NaHCCb (20 mg, 0.328 mmol) in 1:1 THF / H2O (0.60 mL) was added a solution of Alloc- NHS ester (32 mg, 0.161 mmol) in THF (0.20 mL). The mixture was stirred at room temperature overnight then diluted with ethyl acetate (5 mL) and saturated aqueous NaHCCh solution (2 mL). The layers were separated, and the aqueous layer was extracted with three 3- mL portions of ethyl acetate. The combined organic layers were dried over Na 2 SC> 4 , filtered, and concentrated in vacuo to afford compound 48 as a colorless oil, which was used in the next step without purification. MS (ESI, pos.): calc’d for C23H43NO12, 525.3; found 526.3 (M+H).

[00244] Compound 49: To a solution of 48 (0.12 mmol) and N-hydroxysuccinimide (16 mg, 0.14 mmol) in anhydrous DCM (1 mL) was added EDC-HCI (34 mg, 0.177 mmol). The resulting solution was stirred at room temperature for 24 h, at which time LCMS indicated some acid remained. Additional /V-hydroxysuccinimide (5 mg, 0.043 mmol) and EDC-HCI (4 mg, 0.021 mmol) were added. After stirring at room temperature for 1 h, the reaction was concentrated in vacuo. The residue was chromatographed on a 15.5 g C18Aq column, eluting with 5-100% MeCN in thO (both with 0.05% HO Ac). Fractions containing the desired product were combined and lyophilized to afford compound 49 (55 mg, 73% over 2 steps). MS (ESI, pos.): calc’d for C27H46N2O14, 622.3; found 623.3 (M+H).

[00245] Compound 50: An 8 mL glass vial was charged with Val-Ala-PAB-OH (10 mg, 0.0303 mmol). A solution of compound 49 (25 mg, 0.0401) in anhydrous DMA (0.50 mL) and DIEA (7.5 pL, 0.043 mmol) was added. The reaction was stirred at room temperature for 2.25 h then was loaded onto a 15.5 g C18Aq column and eluted with 0-100% MeCN in H2O (both with 0.05% HO Ac). Fractions containing the desired product were lyophilized to provide compound 50 (18 mg, 75%) as a fluffy white solid. MS (ESI, pos.): calc’d for C38H64N4O14, 800.4; found 801.3 (M+H).

[00246] Compound 51: To a solution of 50 (14 mg, 0.0175 mmol) in anhydrous DMF (175 pL) was added DIEA (4.6 pL, 0.0264 mmol) then bis-PNP carbonate (10.6 mg, 0.0348 mmol). The bright yellow solution was stirred at room temperature for 2.5 h. The reaction was diluted with ethyl acetate (5 mL) and washed with saturated aqueous KHSO4 solution (3 mL). The aqueous layer was extracted with two 3-mL portions of ethyl acetate. The combined organic layers were washed with 1 : 1 H 2 0/brine (3 mL) then with brine (3 mL), then were dried over Na2SC>4, filtered, and concentrated in vacuo to afford a colorless oil. Chromatography on 4 g S1O2, eluting with ethyl acetate in hexanes (0-100%) afforded compound 51 (13 mg, 76%). MS (ESI, pos.): calc’d for C45H67N5O18, 965.5; found 966.3 (M+H).

[00247] Compound 52: To a mixture of VX-787 (19a, 50 mg, 0.125 mmol) in allyl alcohol (1.5 mL) was added cone. H2SO4 (25 pL, 0.45 mmol). The resulting suspension was heated at 65 °C for 6.5 h then left stirring at room temperature overnight. The resulting solution was concentrated in vacuo to -0.5 mL and treated with saturated aqueous NaHCCh solution to bring to pH 7-8. The mixture was concentrated in vacuo to yield a gummy solid. Chromatography on a 50 g C18Aq column, eluting with 0-100% MeCN in H2O (both with 0.05% HO Ac) and lyophilization of product-containing fractions afforded compound 52 (41 mg, 75%) as ayellow solid. MS (ESI, pos.): calc’d for C23H23F2N5O2, 439.18; found 440.1 (M+H).

[00248] Compound 53: An 8 mL vial was charged with compound 51 (13 mg, 0.0134 mmol) and compound 52 (6.5 mg, 0.0148 mmol). To the vial was added anhydrous DMA (150 pL), then DMAP (2 mg, 0.0164 mmol). The bright yellow solution was stirred at room temperature for 1.5 h then chromatographed on a 15.5 g C18Aq column, eluting with 5-100% MeCN in H2O (both with 0.05% HOAc). Product-containing fractions were combined and lyophilized to afford compound 53 (12 mg, 71%) as a fluffy white solid. MS (ESI, pos.): calc’d for C62H85F2N9O17, 1265; found 1266.6 (M+H).

[00249] Compound 54: To a solution of compound 53 (9.4 mg, 0.0074 mmol) in dry DCM (0.74 mL) under argon was added Pd(Ph3P)4 (4.1 mg, 0.0036 mmol) and PhSiEE (2.8 pL, 0.022 mmol). After stirring at room temperature for 30 min, the reaction was concentrated. Purification on a Gemini 30 x 150 mm prep HPLC column, eluting with 5-95% MeCN in H2O (each containing 10 mM NEBOAc) afforded 2 mg (23%) of compound 54 as an acetate salt. MS (ESI, pos.): calc’d for C55H77F2N9O15, 1141.6; found 1142.5 (M+H). ¾ NMR (500 MHz, CD3OD) d 8.98 (dd, J= 9.7, 2.9 Hz, 1H), 8.56 (s, 1H), 8.36 (d, J = Hz, 1H), 8.02 (d, J= 3.4 Hz, 1H), 7.70 (d, J= 8.3 Hz, 2H), 7.56 (d, J= 8.3 Hz, 2H), 5.53 (d, J= 11.2 Hz, 1H), 5.49 (d, J= 12.2 Hz, 1H), 4.48 (q, J= 7.3 Hz, 1H), 4.21 (d, J= 6.3 Hz, 1H), 3.72-3.77 (m, 2H), 3.69- 3.57 (m, 3 OH), 3.07 (t, J= 4.88 Hz, 2H), 2.56-2.58 (m, 2H), 2.50 (d, J = 6.3 Hz, 1H), 2.12-2.16 (m, 1H), 2.12 (br s, 1H). 2.03 (br s, 1H), 1.9 (s, 5H) 1.74-1.84 (m, 3H), 1.56-1.64 (m, 2H), 1.40-1.48 (m, 2H) overlapping with 1.45 (d, J= 6.8 Hz, 3H), 1.00 (d, J= 6.4 Hz, 3H), 0.98 (d, .7= 6.8 Hz, 3H).

1.10 Scheme 10

[00250] Compound 55 was prepared following the methods described in ACS Med. Chem. Lett. 2017, 8, 261-265.

[00251] Compound 57: To a 0 °C suspension of Compound 55 (13 mg, 0.032 mmol), O- [tert-butyl(dimethyl)silyl]hydroxylamine (TBSONH2, 8.3 mg, 0.056 mmol), and DMAP (0.90 mg, 0.0074 mmol) in DCM (650 pL) was added EDC-HC1 (7.4 mg, 0.039 mmol). The resulting white suspension was warmed to room temperature and stirred overnight. The reaction was not complete at that time. Added 3 mg more TBSONH2 and 2 mg more EDC-HC1 to the reaction and continued stirring at room temperature for 1 h 20 min, at which time LCMS indicated the reaction was complete. The reaction was concentrated under vacuum. To the resulting mixture of Compound 56 and Compound 57 was added MeCN (300 pL), water (300 pL). and acetic acid (60 pL). The reaction was stirred at room temperature until LCMS indicated complete conversion of the intermediate Compound 56 to the desired hydroxamic acid (57). The reaction was concentrated under vacuum to remove the MeCN. The resulting slurry was dissolved in DMF and loaded onto a 15.5 g C18Aq ISCO RediSep column. Gradient elution with 5-40% MeCN in ThO with 0.05% HOAc in both and lyophilization of pure fractions provided Compound 57 (5.0 mg, 36%) as a white flocculent solid. MS (ESI, pos.) calc’d for C19H19F2N7O2, 415.16; found, 416.2 (M+H). Ή NMR (500 MHz; CD3OD) indicated a mixture of three rotamers or tautomers in a ratio of 92/6/2 at 25 °C: d 8.68 (dd, J = 2.1, 8.4, 1H), 8.52 (br s, 1H), 8.11 (d, J= 3.8 Hz, 0.94H), 8.08 (d, J= 3.9, 0.06H ), 5.13 (d, J= 5.8, 0.06H), 5.05 (d, J= 6.9 Hz, 0.92H), 4.90 (d, J = 8.1, 0.02H), 2.76 (d, J= 7.8, 0.06H) 2.54 (d, J= 6.8 Hz, 0.94H), 2.05—1.94 (m, 3H), 1.92—1.81 (m, 3H), 1.74—1.67 (m, 1H), 1.66—1.60 (m, 1H), 1.56—1.48 (m, 2H).

1.11 Scheme 11

[00252] Compound 58 was prepared following the methods described in European Journal o Medicinal Chemistry 2019, 162, 249-265.

[00253] Compound 59 was prepared as described in ACS Med. Chem. Lett. 2017, 8, 261-265.

[00254] Compound 60: A 10-mL microwave vial equipped with a magnetic stirbar was charged with Compound 58 (22 mg, 0.0625 mmol), Compound 59 (50 mg, 0.0989 mmol), tris(dibenzylideneacetone)dipalladium(0) (3.0 mg, 0.00328 mmol), and X-Phos (3.0 mg, 0.00629 mmol). The vial was capped with a crimp top and purged with argon for 5 min. 2- Methyl-THF (1 mL, purged with argon for 30 min) was added, then a solution of tripotassium phosphate (40 mg, 0.188 mmol) in water (0.2 mL, purged with argon for 30 min) was added and stirring was initiated. The argon balloon was removed, and the vial was placed in a pre heated 100 °C oil bath. The reaction was heated at that temperature for 3 h then let cool to room temperature. LCMS indicated complete consumption of starting material (58). The aqueous layer was removed and Na2SC>4 was added to the remaining organic solution. The solution was filtered through celite, washing with ethyl acetate. The filtrate was concentrated under vacuum. The product was chromatographed on 4 g SiCh Gold ISCO Redisep column, eluting with 5- 40% EtO Ac-hex, holding at 40% until the product eluted. Fractions containing product were combined and concentrated to afford Compound 60 (50 mg, 0.0720 mmol, quant.) as a yellow foam / glassy solid. The product was used in the next step without further purification. MS (ESI, pos.): calc’d for C41H35FN6O2S, 694.25; found, 695.60 (M+H).

[00255] Compound 61: To a solution of Compound 60 (15 mg, 0.0216 mmol) in methanol (300 pL) were added 50% aq hydroxylamine solution (300 pL, 4.54 mmol) and sodium cyanide (1 mg, 0.0204 mmol). To the resulting suspension was added THF (300 pL). The resulting clear solution was stirred at room temperature for 4 days then concentrated to remove the organics. The resulting suspension was dissolved in DMF and loaded onto a 15.5 g C18Aq ISCO column. Eluted with 50-90% MeCN in H2O with 0.05% HOAc as modifier. Pure fractions were combined and lyophilized, affording Compound 61 (10.7 mg, 71%) as a white solid. MS (ESI, pos.): calc’d for C40H34FN7O2S, 695.25; found 696.20 (M+H).

[00256] Compound 62: To a solution of Compound 61 (10 mg, 0.0144 mmol) in DCM (400 pL) were added triethylsilane (23 pL, 0.144 mmol) and trifluoroacetic acid (22 pL, 0.287 mmol). The resulting yellow solution was stirred at room temperature for 90 min then concentrated under vacuum. To the residue was added 2 mL DCM and the solution was concentrated again. Repeated the DCM treatment two more times, resulting in a white solid. The product was purified by chromatography on a 15.5 g C18Aq ISCO RediSep column, eluting with 10-45% MeCN in H2O with 0.05% HOAc in both. Held at 45% MeCN until the product finished eluting. Fractions containing pure product were combined and lyophilized. The resulting solid was suspended in MeCN containing a little H2O, frozen, and lyophilized again to afford 5.0 mg (76%) of Compound 62 as a white fluffy solid. Purity >99% by UPLC. MS (ESI, pos) calc’d for C21H20FN7O2S, 453.14; found 454.81 (M+H). ¾ NMR (300 MHz, CD3OD) indicated a mixture of three rotomers or tautomers in a ratio of 45/40/15 at 25 °C: d 8.92—8.80 (m, 1H), 8.58—8.52 (m, 1H), 7.73 (d, J= 6.0, 0.55H), 7.68 (d, J= 6.0, 0.45H) 7.47 (d, J= 6.4, 0.45H), 7.45 (d, J= 6.0, 0.40H), 7.43 (d, J= 6.4, 0.15H), 5.27 (br d, J= 5.9, 0.40H), 5.17 (br d, J= 7.3, 0.15H), 5.11 (br d, J= 6.7, 0.45H), 2.83 (br d, J = 7.8, 0.55H), 2.63 (br d, J = 6.4, 0.45h), 2.29—1.97 (m, 4H), 1.97—1.86 (m, 2H), 1.85—1.40 (m, 4H). 1.12 Scheme 12

[00257] Compound 64: Fmoc-Glu(OAllyl)-OH 63 (3.5 g, 8.548 mmol) was combined with HATU (3.25 g, 8.548 mmol) andHOBt (1.16 g, 8.548 mmol) in DMF (37 mL). To this mixture was added DIEA (3 mL, 17.097 mmol). After stirring the resulting solution for 5 min at room temperature, Val-Ala-PAB-OH (3 g, 10.258 mmol) was added, and the resulting solution was stirred at room temperature for 2 hours. Reaction mixture was diluted with EtOAc, washed with cold DI water followed by brine. Organic layer was dried over MgS0 4 , filtered and concentrated. The residue viscous oil was sonicated with Et 2 0 to precipitate out the product. The mixture was stirred overnight and filtered to afford compound 64 as a pale-yellow solid. MS (ESI, pos.): calc’d for C38H44N4O8, 684.32; found 685.35 (M+H).

[00258] Compound 65: In a 20 mL vial, compound 64 (266 mg, 0.388 mmol) was dissolved in 5% piperidine solution in DMF (2 mL). The reaction was stirred at room temperature for 75 mins, and purified on a 50 g C18 Aq column, eluting with 0-30% MeCNTUO (both having 0.05% AcOH). Pure fractions were combined, frozen, and lyophilized to afford compound 65 (145 mg, 81% yield) as a fluffy off-white solid. MS (ESI, pos.): calc’d for C23H34N4O6, 462.25; found 463.2 (M+H).

[00259] Compound 66: DIEA (63 pL, 0.361 mmol) was added to a solution of compound 65 (145 mg, 0.313 mmol) and Alloc-amido-PEG8-NHS-ester 49 (150 mg, 0.241 mmol) in anhydrous DMA (5 mL), and the resulting solution was stirred for 1 hour at room temperature. The reaction was purified on a 50 g C18 Aq column and eluted with 0-80% MeCN/H 2 0 (both having 0.05% AcOH). Pure fractions were combined, frozen, and lyophilized to obtain compound 66 (130 mg, 56% yield) as a fluffy off-white solid. MS (ESI, pos.): calc’d for C46H75N5O17, 969.52; found 970.69 (M+H).

[00260] Compound 67: To a solution of compound 66 (130 mg, 0.134 mmol) in DCM (2.5 mL) was added thionyl chloride (29 pL, 0.402 mmol). The resulting solution was stirred at room temperature for 2 hours. The crude reaction was concentrated and purified on a 4 g S1O2 gold column eluting with 0-20% MeOH in DCM to afford compound 67 (76 mg, 57% yield) as colorless gel. MS (ESI, pos.): calc’d for C46H74CIN5O16, 987.48; found 989.46 (M+H).

[00261] Compound 68: VX-787 Allyl ester 52 (30 mg, 0.068 mmol), compound 67 (74.2 mg, 0.075 mmol), K2CO3 (28.3 mg, 0.205 mmol) and sodium iodide (8.9 mg, 0.075 mmol) were combined in DMA (1.5 mL). The resulting solution was stirred at 50 °C for 4 hours. Crude reaction was loaded onto a 15.5 g C18 Aq column and eluted with 0-95% MeCNTHO (both having 0.05% AcOH). Pure fractions were combined, frozen, and lyophilized to afford compound 68 (60 mg, 63% yield) as a fluffy off-white solid. MS (ESI, pos.): calc’d for C69H96F2N10O18, 1390.69; found 1391.5 (M+H).

[00262] Compound 69: To a solution of compound 68 (60 mg, 0.043 mmol) in anhydrous DCM (4 mL) under argon was added Pd(Ph3P)4 (24.9 mg, 0.022 mmol), PhSflE (16 pL, 0.129 mmol). The resulting solution was stirred at room temperature for 1 hour. The crude reaction was loaded onto a 15.5 g C18 Aq column and eluted with 0-60% MeCNTHO (both having 0.05% AcOH). Pure fractions were combined, frozen, and lyophilized to obtain the title compound697 (25 mg, 47% yield). Purification was repeated on the lyophilized solid on a Gemini 30 x 150 mm prep HPLC column, eluting with 0-60% MeCNTHO (both having 0.05% AcOH) to afford compound 69 (11.2 mg, 21%) as a fluffy off-white solid. MS (ESI, pos.): calc’d for C59H84F2N10O16, 1226.60; found 1227.5 (M+H). X H-NMR (500 MHz; DMSO-de): d 9.98 (s, 1H), 8.51 (dd, J= 9.5, 3.0 Hz, 1H), 8.39 (d, J= 6.5 Hz, 1H), 8.33 (d, J= 1.0 Hz, 1H), 8.30 (s, 1H), 8.20 (d, .7= 6.5 Hz, 1H), 8.11 (d, J = 4.0 Hz, 1H), 7.75 (d, J= 8.5 Hz, 1H), 7.55 (d, J= 8.5 Hz, 2H), 7.50 (d, J= 7.0 Hz, 1H), 7.26 (d, J= 8.5 Hz, 2H), 5.48 (s, 2H), 4.69 (t , J = 5.5 Hz, 1H), 4.35-4.32 (m, 1H), 4.24 (q, J= 6.5 Hz, 1H), 4.14 (dd, J= 8.0, 6.5 Hz, 1H), 3.62- 3.54 (m, 6H), 3.48-3.45 (m, 30H), 2.77 (t, J= 5.5 Hz, 2H), 2.74-2.73 (m, 1H), 2.42-2.35 (m, 1H), 2.34-2.28 (m, 1H), 2.18-2.07 (m, 2H), 2.00 (s, 1H), 1.91-1.88 (m, 1H), 1.84-1.69 (m, 4H), 1.62-1.32 (m, 5H), 1.28 (d, J= 7.0 Hz, 3H), 0.82 (dd, J= 19.0, 6.5 Hz, 6H). 1.13 Scheme 13

[00263] Compound 70: Using the same method as for compound 35 on a 100 mg scale, compound 70 was prepared from Fmoc-Val-Cit-OH 33 and 1 -(4-aminophenyl)-2,2,2- trifluoroethan-l-ol 34a. 105 mg (78% yield) of product was isolated. MS (ESI, pos.): calc’d for C34H38F3N5O6, 669.28; found 670.38 (M+H).

[00264] Compound 71: Using the same method as for compound 36 on a 100 mg scale, compound 71 was prepared from compound 70. 55 mg (82% yield) of product was isolated. MS (ESI, pos.): calc’d for C19H28F3N5O4, 447.21; found 448.24 (M+H).

[00265] Compound 72: Using the same method as 37 on a 125 mg scale, compound 72 was prepared from compound 71. 159 mg (89% yield) of product was isolated. MS (ESI, pos.): calc’d for C53H75F3N6O15, 1092.52; found 1093.63 (M+H).

[00266] Compound 73: To a DCM solution of compound 72 (151 mg, 0.138 mmol), VX- 787 19a (50 mg, 0.125 mmol) and DMAP (3.1 mg, 0.025 mmol) were added. 1 M solution of DCC in DCM (0.125 mL, 0.188 mmol) was added dropwise and the reaction was stirred at room temperature overnight. The reaction was concentrated and purified on a 100 g C18 Aq column eluting with 5-95% MeCNTUO (both having 0.05% AcOH as a modifier). Pure fractions were combined, frozen and lyophilized to obtain compound 73 (102 mg, 50%) as off- white fluffy solid. The isolated product was further purified on a Teledyne ISCO with Gemini 30x150 mm column eluting with 0-90% MeCNTUO (both having 0.05% AcOH as a modifier). Pure fractions were combined, frozen and lyophilized to get two separate diastereomeric pure compounds 73a (25 mg, 25%, first eluting product) and 73b (30 mg, 29%, later eluting product) as off-white fluffy solid. MS (ESI, pos.): calc’d for C 73 H 92 F 5 N 11 O 16 , 1473.66; found 1474.7 (M+H).

[00267] Compound 74a: Using the same method as 39 on a 25 mg scale, compound 74a was prepared from compound 73a. 16.0 mg (75% yield) of product was isolated. MS (ESI, pos.): calc’d for CsstfcFsNnOw, 1251.60; found 1252.5 (M+H). ¾-NMR (500 MHz; DMSO-rfy): d 10.09 (s, 1H), 8.48 (dd, J= 10.0 Hz, 3.0 Hz, 1H), 8.26-8.24 (m, 1H), 8.18-8.17 (m, 2H), 8.14- 8.13 (m, 1H), 7.86 (d, J= 8.0 Hz, 1H), 7.70 (d, J= 7.5 Hz, 1H), 7.59 (d, J= 8.5 Hz, 2H), 7.38 (d, J = 8.5 Hz, 2H), 6.34 (q, J = 7.0 Hz, 1H), 6.00-5.96 (m, 1H), 5.40 (s, 2H), 4.82-4.78 (m, 1H), 4.38-4.34 (m, 2H), 4.22 (t , J= 7.0 Hz, 1H), 3.61-3.57 (m, 4H), 3.50-3.47 (m, 30H), 3.37- 3.35 (m, 2H), 3.15 (d, J= 7.5 Hz, 1H), 3.04-2.92 (m, 2H), 2.41-2.34 (m, 1H), 2.10-2.07 (m, 1H), 1.99-1.94 (m, 1H), 1.94-1.90 (m, 1H), 1.86-1.72 (m, 4H), 1.72-1.50 (m, 3H), 1.49-1.25 (m, 5H), 0.85 (dd, J= 15.5, 6.5 Hz, 6H).

[00268] Compound 74b: Using the same method as 39 on a 30 mg scale, compound 74b was prepared from compound 73b. 17.3 mg (68% yield) of product was isolated. MS (ESI, pos.): calc’d for C58H82F5N11O14, 1251.60; found 1252.5 (M+H). ¾-NMR (500 MHz; DMSO- d 6 ): d 10.06 (s, 1H), 8.44 (dd, J= 10.0 Hz, 2.5 Hz, 1H), 8.26-8.24 (m, 1H), 8.18-8.12 (m, 3H), 7.86 (d, J = 8.0 Hz, 1H), 7.67 (d, J= 6.5 Hz, 1H), 7.54 (d, J= 8.5 Hz, 2H), 7.38 (d, J= 8.5 Hz, 2H), 6.40 (q, J= 7.0 Hz, 1H), 6.00-5.97 (m, 1H), 5.40 (s, 2H), 4.76-4.73 (m, 1H), 4.37-4.33 (m, 2H), 4.22 (t, J= 7.0 Hz, 1H), 3.62-3.55 (m, 4H), 3.53-3.46 (m, 30H), 3.37-3.35 (m, 2H), 3.16 (d, J= 7.5 Hz, 1H), 3.04-2.91 (m, 2H), 2.39-2.37 (m, 1H), 2.12-2.07 (m, 1H), 1.99-1.93 (m, 1H), 1.87-1.78 (m, 5H), 1.71-1.63 (m, 3H), 1.61-1.33 (m, 5H), 0.83 (dd, J= 14.5, 6.5 Hz, 6H).

1.14 Scheme 14

[00269] Compound 75: Using the same method as 35 on a 500 mg scale, compound 75 was prepared from Fmoc-Val-Cit-OH 33 and l-(4-aminophenyl)propan-l-ol 34b. 489 mg (77% yield) of product was isolated as a pale-yellow solid. MS (ESI, pos.): calc’d for C35H43N5O6, 629.32; found 630.23 (M+H).

[00270] Compound 76: Using the same method as 36 on a 400 mg scale, compound 76 was prepared from compound 75. 147 mg (52% yield) of product was isolated as off-white fluffy solid. MS (ESI, pos.): calc’d for C20H33N5O4, 407.25; found 408.20 (M+H).

[00271] Compound 77: Using the same method as 37 on a 211 mg scale, compound 77 was prepared from compound 76. 217 mg (74% yield) of product was isolated as off-white fluffy solid. MS (ESI, pos.): calc’d for C 54 H 8 oN 6 Oi 5 , 1052.57; found 1053.74 (M+H).

[00272] Compound 78: Using the same method as 73 on a 75 mg scale, compound 78 was prepared from compound 77. 112.5 mg (38% yield) of product was isolated as a mixture of diastereomers. MS (ESI, pos.): calc’d for C74H97F2N11O16, 1433.71; found 1435.56 (M+H).

[00273] Compound 79: Using the same method as 39 on a 25 mg scale, compound 79 was prepared from compound 78. 16.0 mg (75% yield) of product was isolated. MS (ESI, pos.): calc’d for C59H87F2N11O14, 1211.64; found 1212.6 (M+H). ^-NMR (500 MHz; DMSC s): d 9.95 (s, 1H), 8.49 (dd, J = 9.5 Hz, 2.5 Hz, 1H), 8.33-8.24 (m, 1H), 8.21-8.15 (m, 1H), 8.14-8.09 (m, 1H), 7.86 (d, J = 8.0 Hz, 1H), 7.65-7.59 (m, 1H), 7.47 (d, J = 8.0 Hz, 2H), 7.19 (d, J = 8.0 Hz, 2H), 7.15 (d, = 8.5 Hz, 1H), 6.02-5.96 (m, 1H), 5.59-5.51 (m, 1H), 5.44 (d, J= 10.0 Hz, 1H), 5.39 (s, 2H), 4.84-4.76 (m, 1H), 4.37-4.31 (m, 2H), 4.21 (t , J= 7.0 Hz, 1H), 3.61-3.56 (m, 4H), 3.52-3.45 (m, 30H), 3.01-2.90 (m, 2H), 2.65-2.60 (m, 2H), 2.41-2.34 (m, 2H), 2.09-1.88 (m, 4H), 1.85-1.71 (m, 4H), 1.56- 1.21 (m, 8H), 0.83 (dd, J= 14.5, 7.0 Hz, 6H), 0.73 (t, J = 7.5 Hz, 2H), 0.68 (t ,J= 7.5 Hz, 1H).

1.15 Scheme 15

[00274] General procedure to prepare protected hydroxamic acid derivatives from the corresponding carboxylic acids: Compound 80a: A room temperature 1 M solution of DCC in dichloromethane (250 pL, 0.376 mmol) was added dropwise to a suspension of VX-787 (100 mg, 0.250 mmol), 0-(/er/-butyldimethylsilyl)hydroxylamine (TBSONH2, 44.3 mg, 0.300 mmol) and DMAP (3 mg, 0.025 mmol) in anhydrous dichloromethane (8 mL) at room temperature. The reaction mixture was stirred at room temperature for 2 hours. Volatiles were removed in vacuo and ethyl acetate was added. The solution was cooled to 0 °C and the precipitate was filtered. The filtrate was concentrated and purified on a 50 g C18 Aq column eluting with 0 to 100% MeCN/TkO (both having 0.05% AcOH). Pure fractions were combined, frozen and lyophilized to obtain compound 80 (62 mg, 39%) as fluffy off-white solid. MS (ESI, pos.): calc’d for C26H34F2N6O2S1, 528.25; found 529.80 (M+H).

[00275] Compound 80b: Using the same general procedure as 80a on a 100 mg scale, compound 80b was prepared from <9-(tetrahydro-2//-pyran-2-yl (hydroxyl amine (THPONH2). Il l mg (89% yield) of product was isolated as off-white fluffy solid. MS (ESI, pos.): calc’d for C25H28F2N6O3, 498.22; found 499.75 (M+H). 1.16 Scheme 16

[00276] Compound 82: To a solution of compound 81 (91.5 mg, 0.097 mmol) in DCM (1.9 mL) was added thionyl chloride (21 pL, 0.292 mmol). The reaction mixture was stirred at room temperature for 1 hour and was then concentrated in vacuo. Anhydrous DCM (2 mL) was added to the residue and the mixture was concentrated in vacuo again to afford compound 82 (92 mg, 99% yield) as a pale-yellow gel, which was used in the next step without purification. MS (ESI, pos.): calc’d for C49H69CIN4O13, 956.45; found 958.38 (M+H).

[00277] Compound 83: To a solution of 80a (30 mg, 0.057 mmol) in DMF (1.1 mL), KOH (3.8 mg, 0.068 mmol) was added. After stirring the resulting solution at 0 °C for 30 mins, compound 82 (59.8 mg, 0.062 mmol) was added. The reaction mixture was stirred at room temperature for 90 mins until the alkylated compound was completely deprotected according to LCMS. The crude reaction was purified on a 50 g Cl 8 Aq column and eluted with 0-65% MeCN/ThO (both having 0.05% AcOH). Pure fractions were combined, frozen, and lyophilized to obtain compound 83 as a fluffy off-white solid. Purification was repeated on the lyophilized solid on a Gemini 30 x 150 mm prep HPLC column, eluting with 0-50% MeCN/ThO (both having 0.05% AcOH) to afford compound 83 (5.9 mg, 9%) as a colorless gel. MS (ESI, pos.): calc’d for C54H78F2N10O13, 1112.57; found 1113.5 (M+H). Tl-NMR ^OO MHz; DMSO-de): d 9.98-9.97 (m, 1H), 8.83-8.81 (m, 1H), 8.57-8.53 (m, 1H), 8.52-8.49 (m, 1H), 8.26-8.25 (m, 1H), 8.18 (s, 1H), 8.08 (d, J= 4.0 Hz, 1H), 8.07-8.01 (m, 1H), 7.67-7.63 (m, 1H), 7.54-7.52 (m, 2H), 7.11-7.10 (m, 2H), 4.89-4.81 (m, 1H), 4.70-4.63 (m, 1H), 4.59- 4.53 (m, 1H), 4.31-4.27 (m, 1H), 4.17-4.10 (m, 1H), 4.03-3.99 (m, 1H), 3.65-3.55 (m, 6 H), 3.53-3.34 (m, 33H), 2.63-2.62 (m, 2H), 2.35-2.32 (m, 2H), 2.03-1.92 (m, 3H), 1.48-1.22 (m, 14H), 0.85-0.82 (m, 6H). 1.17 Scheme 17

[00278] Compound 85: To a solution of Fmoc-Glu(0¾u)-OH 84 (1 g, 2.35 mmol) and N- hydroxysuccinimide (324.6 mg, 2.82 mmol) in anhydrous DCM (20 mL) was added EDC-HC1 (675.8 mg, 0.177 mmol). The resulting solution was stirred at room temperature for 19 hours, at which time LCMS indicated some acid remained. Additional N-hydroxysuccinimide (324.6 mg, 2.82 mmol) and EDC-HC1 (675.8 mg, 0.177 mmol) were added. After stirring at room temperature for 3 hours, the reaction was concentrated in vacuo. The residue was chromatographed on a 40 g SiC gold column, eluting with 0-10% MeOH in DCM. Fractions containing the desired product were combined and concentrated to afford compound 85 (1.01 g, 82%) as off-white solid. MS (ESI, pos.): calc’d for C28H30N2O8, 522.2; found 545.30 (M+Na).

[00279] Compound 86 and 87: To a solution of compound 85 (1 g, 1.914 mmol) and Val- Ala-PAB-OH (673.7 mg, 2.296 mmol) in anhydrous DMF (8.3 mL), DIEA (0.67 mL, 3.827 mmol) was added. The reaction was stirred at room temperature for 20 hours and purified on a 100 g Cl 8 Aq column eluting with 0-80% MeCN in H2O (both with 0.05% HO Ac). Fractions containing the desired products were lyophilized to provide compound 86 (262.5 mg, 20%) and compound 87 (441 mg, 48%) as a fluffy white solid. Compound 86: MS (ESI, pos.): calc’d for C39H48N4O8, 700.35; found 701.53 (M+H). Compound 87: MS (ESI, pos.): calc’d for C24H38N4O6, 478.28; found 479.43 (M+H).

[00280] Compound 88: DIEA (50 pL, 0.288 mmol) was added to a solution of compound 87 (119.4 mg, 0.249 mmol) and Fmoc-amido-PEG8-NHS-ester (146 mg, 0.192 mmol) in anhydrous DMA (4 mL), and the resulting solution was stirred at room temperature for 1 hour. The crude reaction was purified on a 150 g C18 Aq column and eluted with 0-80% MeCN/H20 (both having 0.05% AcOH). Pure fractions were combined, frozen, and lyophilized to obtain compound 88 (149 mg, 72% yield) as a fluffy off-white solid. MS (ESI, pos.): calc’d for CssHssNsOn, 1123.59; found 1124.69 (M+H).

[00281] Compound 89: To a solution of compound 88 (149 mg, 0.133 mmol) in DCM (2.7 mL) was added thionyl chloride (29 pL, 0.398 mmol). The reaction mixture was stirred at room temperature for 24 hours and was then concentrated in vacuo. Anhydrous DCM (2 mL) was added to the residue and the mixture was concentrated in vacuo again to afford compound 89 (143 mg, 94% yield) as a colorless gel, which was used in the next step without purification. MS (ESI, pos.): calc’d for C58H84CIN5O16, 1141.56; found 1142.66 (M+H).

[00282] Compound 90: To a solution of 80b (18 mg, 0.036 mmol) inDMF (0.72 mL), KOH (6.1 mg, 0.108 mmol) was added. After stirring the resulting solution for 30 mins, compound

89 (45 mg, 0.040 mmol) was added. The reaction mixture was stirred at room temperature for

90 mins until the alkylated product was observed as the major product according to LCMS. Piperidine (9 pL) was then added and the reaction mixture was stirred at room temperature for 30 mins. The crude reaction was purified on a 5.5 g C18 Aq column and eluted with 0-80% MeCNTEO (both having 0.05% TFA). Pure fractions were combined, frozen, and lyophilized to obtain compound 90 (25 mg, 50% yield) as a fluffy off-white solid. MS (ESI, pos.): calc’d for C68H101F2N11O17, 1381.73; found 1382.85 (M+H).

[00283] Compound 91: To a solution of 90 (25 mg, 0.018 mmol) in MeCN (0.181 mL) and H2O (0.181 mL), TFA (0.138 mL, 1.809 mmol) was added, and the resulting solution was stirred at room temperature for 30 mins. The crude reaction was purified on a 5.5 g C18 Aq column and eluted with 0-80% MeCNTHO (both having 0.05% TFA). Pure fractions were combined, frozen, and lyophilized to obtain compound 91 as a fluffy off-white solid. Purification was repeated twice on the lyophilized solid on a Gemini 30 x 150 mm prep HPLC column, eluting with 0-60% MeCN/H 2 0 (both having 0.05% TFA) to afford compound 91 (5.3 mg, 24% yield) as a fluffy off-white solid. MS (ESI, pos.): calc’d for C59H85F2N11O16, 1241.61; found 1242.6 (M+H). ¾-NMR (500 MHz; DMSO-de): d 9.89 (br, 1H), 8.58-8.52 (m, 1H), 8.28-8.24 (m, 1H), 8.17-8.10 (m, 1H), 7.86-7.84 (m, 1H), 7.76-7.65 (m, 2H), 7.55-7.53 (m, 1H), 7.50-7.48 (m, 1H), 7.25-7.22 (m, 1H), 7.15-7.13 (m, 1H), 6.55-6.46 (m, 1H), 4.86-4.81 (m, 1H), 4.69-4.64 (m, 1H), 4.41-4.35 (m, 1H), 4.22-4.18 (m, 1H), 3.60-3.39 (m, 35H), 2.99- 2.96 (m, 2H), 2.44-2.33 (m, 1H), 2.00-1.31 (m, 13H), 1.29-1.26 (m, 3H), 0.88-0.83 (m, 6H).

1.18 Scheme 18

[00284] Compound 92 was prepared by adapting methods described in ACS Med. Chem. Lett. 2017, 8, 261-265.

[00285] Compound 93: A dry 4 mL vial was charged with methyl Compound 92 (20 mg, 0.048 mmol), AllocNHPEG8-Val-Ala-PAB-Cl (41, 48 mg, 0.059 mmol), potassium carbonate (20 mg, 0.145 mmol), and sodium iodide (4.0 mg, 0.027 mmol). Anhydrous DMA (0.5 mL) was added. The resulting salmon-colored mixture was stirred at room temperature for 60 h. The reaction mixture was loaded directly onto a 50 g C18Aq ISCO column. Eluted with 10- 100% MeCN in EhO with 0.05% HO Ac modifier. The cleanest product-containing fractions were combined and lyophilized to afford 40 mg of a mixture of N-alkylated regioisomers. The isomers were separated by chromatography on a Gemini 30 x 150 mm prep HPLC column, eluting with 30-55% MeCN in ¾0 with 10 mM MEOAc as modifier. Pure fractions of the later-eluting peak (major isomer) were combined and lyophilized to afford Compound 93 (11 mg, 19%) as an off-white fluffy solid. MS (ESI, pos.): calc’d for C58H82F2N10O15, 1196.59; found, 1197.5 (M+H). [00286] Compound 94: To a solution Compound 93 (10 mg, 0.00835 mmol) in methanol (110 pL) were added 50% aqueous hydroxylamine solution (110 pL. 1.67 mmol) and sodium cyanide (1 mg, 0.0204 mmol). The resulting slightly cloudy solution was stirred at room temperature overnight. The MeOH was removed under vacuum. The remaining aqueous solution was loaded onto a 5.5 g C18Aq column, rinsing the vial with DMF. Eluted with 10- 60% MeCN in EbO with 0.05% HOAc as modifier. Fractions that were >95% pure by LCMS were combined and lyophilized to afford Compound 94 (8.8 mg, 88%) as an off-white fluffy solid. MS (ESI, pos.): calc’d for C57H81F2N11O15, 1197.59; found, 1198.5 (M+H).

[00287] Compound 95: A vial containing Compound 94 (5.0 mg, 0.00417 mmol) was purged with argon. A separate vial containing tetrakis(triphenylphosphin)-)palladium(0) (5.0 mg, 0.00433 mmol) was purged with argon. To this vial was added 200 pL of a 37/2/1 solution of CHCb/HOAc/NMM that had been de-oxygenated with a stream of argon for 10 min. The mixture was stirred to dissolve the catalyst. The catalyst solution was added by syringe to the vial containing the substrate under argon. The catalyst vial was rinsed with an additional 100 pL of the solvent and the rinse was added to the reaction. Stirred at room temperature for 2 h, at which time LCMS indicated complete consumption of starting material. The reaction was concentrated under vacuum then purified by chromatography on a 5.5 g Cl 8Aq ISCO RediSep column, eluting with 5-40% MeCN in H2O with 0.05% HOAc in both. Held at 40% MeCN until the product and Ph3PO had completely eluted. Product-containing fractions were lyophilized to afford 6 mg of a yellow solid that contained Ph3PO as an impurity. The product was purified on a 30 x 150 mm Gemini prep HPLC column, eluting with 10-95% MeCN in H2O with 0.05% HOAc in both. Product-containing fractions were lyophilized to afford Compound 95 (1.6 mg, 34%) as an off-white solid. MS (ESI, pos.): calc’d for C53H77F2N11O13, 1113.57; found 1114.6 (M+H).

1.19 Scheme 19

[00288] Compound 97 was prepared following the same method as described for Compound 60. From 46 mg of Compound 96 was obtained 95 mg (quant.) of Compound 97 as a yellow solid. MS (ESI, pos.): calc’d for C42H38FN7O2, 691.31; found 692.30 (M+H).

[00289] Compound 98 was prepared following the same procedure as for Compound 61. From 20 mg of Compound 97 was obtained 12.3 mg (61%) of Compound 98 as a yellow solid. MS (ESI, po.):: calc’d for C41H37FN8O2, 692.30; found, 693.40 (M+H).

[00290] Compound 99 was prepared following the same procedure as for Compound 62. From 11 mg of Compound 98 was obtained 2 mg (28%) of Compound 99 as a pale yellow fluffy solid. MS (ESI, po.)) calc’d for C22H23FN8O, 450.19; found 451.50 (M+H).

Scheme 20

[00291] Compound 101 was prepared following the same procedure as for Compound 61. From 100 mg of Compound 100 was obtained 75 mg (75%) of Compound 101 as a white solid. MS (ESI, pos.): calc’d for C38H33F2N7O2, 657.27: Found, 658.47 (M+H), 680.42 (M+Na).

[00292] (2-((((9//-Fluoren-9-yl)metho\y)carbonyl)amino)acetamido)met hyl acetate (102) was prepared following the procedure from Tetrahedron 2018, 74, 1951-1956.

[00293] Compound 103: To a solution of compound 102 (24.6 mg, 0.067 mmol) and compound 101 (22 mg, 0.033 mmol) in anhydrous DCM (2.8 mL) was added pyridinium p- toluenesulfonate (PPTS, 16.8 mg, 0.067 mmol). The resulting solution was stirred at 40 °C for 16 hours, at which time LCMS indicated some hydroxamic acid remained. Additional compound 102 (24.6 mg, 0.067 mmol) and PPTS (16.8 mg, 0.067 mmol) were added. After stirring at 40 °C for 24 hours, the reaction was concentrated in vacuo. The residue was chromatographed on a 5.5 g C18 Aq column eluting with 0-100% MeCN/FhO (both having 10 mM NFEOAc). Pure fractions were combined, frozen, and lyophilized to obtain compound 103 (22 mg, 68% yield) as a fluffy off-white solid. MS (ESI, pos.): calc’d for C56H49F2N9O5, 965.38; found 966.86 (M+H). [00294] Compound 104: In a 20 mL vial, compound 103 (22 mg, 0.023 mmol) was dissolved in 5% piperidine solution in DMF (0.12 mL), and the resulting solution was stirred at room temperature for 1 hour. The reaction was purified on a 5.5 g Cl 8 Aq column eluting with 0- 100% MeCN/LbO (both having 10 mM NLLOAc). Pure fractions were combined, frozen, and lyophilized to afford compound 104 (15 mg, 89% yield) as a fluffy off-white solid. MS (ESI, pos.): calc’d for C 41 H 39 F 2 N 9 O 3 , 743.82; found 744.65 (M+H).

[00295] Compound 105: Compound 104 (15 mg, 0.020 mmol) was combined with Fmoc- Cap-Gly-Gly-Phe-OH 27 (12.4 mg, 0.020 mmol), HATU (7.7 mg, 0.020 mmol) and HO At (2.7 mg, 8.548 mmol) in DMF (0.67 mL). To this mixture was added DIEA (7 pL, 0.040 mmol), and the resulting solution was stirred at room temperature for 3 hours. The reaction was purified on a 5.5 g C18 Aq column eluting with 0-100% MeCNTHO (both having 10 mM MEOAc). Pure fractions were combined, frozen, and lyophilized to afford compound 105 (15 mg, 56% yield) as a fluffy off-white solid. MS (ESI, pos.): calc’d for C75H75F2N13O9, 1339.58; found 1341.28 (M+H).

[00296] Compound 106: To a solution of compound 105 (15 mg, 0.0111 mmol) in DCM (1 mL) were added triethylsilane (17.9 pL, 0.112 mmol) and trifluoroacetic acid (22 pL, 0.224 mmol). The resulting solution was stirred at room temperature for 20 hours and then concentrated in vacuo. The reaction was purified on a 5.5 g Cl 8 Aq column eluting with 0- 95% MeCNTLO (both having 10 mM MEOAc). Pure fractions were combined, frozen, and lyophilized to afford compound 106 (9 mg, 73% yield) as a fluffy off-white solid. MS (ESI, pos.): calc’d for C56H61F2N13O9, 1097.47; found 1099.24 (M+H).

[00297] Compound 107: In a 20 mL vial, compound 106 (9 mg, 0.008 mmol) was dissolved in 5% piperidine solution in DMF (0.82 mL), and the resulting solution was stirred at room temperature for 2 hours. The reaction was purified on a 5.5 g C18 Aq column eluting with 0- 50% MeCNTLO (both having 10 mM MEOAc). Pure fractions were combined, frozen, and lyophilized to afford compound 107 (4 mg, 56% yield) as an off-white solid. MS (ESI, pos.): calc’d for C41H51F2N13O7, 875.40; found 877.07 (M+H). [00298] Scheme 21

[00299] Compound 110: To a solution of methyl 4-aminobicyclo[2.1.1]hexane-l- carboxylate hydrochloride (108, 19.1 mg, 0.1 mmol) and 2,4-dichloro-5-fluoropyrimidine (109, 20 mg, 0.12 mmol) in DCM (2 mL), DIEA (38 pL, 0.22 mmol) was added. The reaction was heated to 32 °C for 16 h then volatiles were removed under reduced pressure. The residue was purified on 4 g silica gold column using hexanes / ethyl acetate to obtain the compound 110 (22 mg, 77%) as an off-white solid. MS (ESI, pos.): calc’d for C12H13CIFN3O2, 285.1; found 286.1 (M+H).

[00300] Compound 111 was prepared by adapting the literature procedure described in Journal of Medicinal Chemistry (2014), 57(15), 6668-6678.

[00301] Compound 112: A mixture of compound 110 (22 mg, 0.077 mmol), compound 111 (38.5 mg, 0.092 mmol) and K3PO4 (2.9 mg, 0.014 mmol) in 2-methyl THF (1 mL) and water (0.2 mL) was purged with argon for 10 mins. X-Phos (4.4 mg, 0.009 mmol) and Pd2(dba)3 (1.8 mg, 0.002 mmol) were added and the resulting mixture was heated to 110 °C for 3 h. The reaction was cooled to room temperature, diluted with ethyl acetate (5 mL), and filtered through a pad of celite. The filter cake was rinsed with ethyl acetate (3 mL), and combined filtrate was concentrated. Purification on 12 g silica gold column using hexanes / ethyl acetate afforded compound 112 (30 mg, 72%) as a yellowish solid. MS (ESI, pos.): calc’d for C26H23F2N5O4S, 539.1; found 540.1 (M+H).

[00302] Compound 113: To a solution of compound 112 (30 mg, 0.056 mmol) in acetonitrile (0.5 mL), 4 M HCI in dioxane (83 pL, 0.344 mmol) was added. After heating to 70 °C for 18 h, the reaction was cooled to room temperature and the volatiles were removed in vacuo. The residue was dissolved in 1 : 1 THF/MeOH (1 mL) and 2 N aqueous sodium hydroxide (167 pL. 0.344 mmol) was added. After heating to 30 °C for 2 h, the reaction was cooled to room temperature, acidified to pH 4-5 by adding 1 N HC1, and diluted with 2-methyl THF (5 mL). Brine (1 mL) was added, and the aqueous layer was discarded. The organic layer was concentrated, and the residue was purified on 30 g Cl 8 Aq column using 5-95% MeCNTHO (both having 0.05% AcOH). Pure fractions were combined and lyophilized to obtain compound 113 (11.2 mg, 54%) as an off-white fluffy solid. MS (ESI, pos.): calc’d for C18H15F2N5O2, 371.1; found 372.1 M+H). ¾ NMR (500 MHz; DMSO-de): d 12.26 (d, J= 2.2 Hz, 2H), 8.43 (dd, .7= 9.8, 2.9 Hz, 1H), 8.27 (dd, .7 = 2.8, 1.4 Hz, 1H), 8.19 (d, J= 3.8 Hz, 1H), 8.12 (d, J = 2.8 Hz, 1H), 8.07 (s, 1H), 2.20 (s, 2H), 2.10-2.06 (m, 2H), 1.94-1.91 (m, 4H).

1.20 Scheme 22

116 117

[00303] Compound 115: To a solution of methyl 4-aminobicyclo[2.1.1]hexane-l- carboxylate hydrochloride (108, 19.1 mg, 0.1 mmol) and 2,4-dichlorothieno[2,3-d]pyrimidine (114, 28.6 mg, 0.13 mmol) in DMF (1 mL), potassium carbonate (42 mg, 0.3 mmol) was added and the mixture was heated to 65 °C for 1.5 h. The reaction was cooled to the room temperature, diluted with water (10 mL) and saturated aqueous ammonium chloride solution (5 mL) and extracted with ethyl acetate (3 x 5 mL). The combined organics were dried over anhydrous sodium sulfate and concentrated in vacuo. The residue was purified on 4 g silica gold column using hexanes / ethyl acetate to obtain the title compound 115 (27.6 mg, 85%) as an off-white solid. MS (ESI, pos.): calc’d for C14H14CIFN3O2S, 323.0; found 324.1 (M+H).

[00304] Compound 116: A mixture of compound 115 (27.5 mg, 0.085 mmol), compound 111 (42.4 mg, 0.102 mmol) and K3PO4 (3.3 mg, 0.015 mmol) in 2-methyl THF (1 mL) and water (0.2 mL) was purged with argon for 10 min. X-Phos (4.9 mg, 0.0102 mmol) and Pd 2 (dba) 3 (2.0 mg, 0.0021 mmol) were added and the resulting mixture was heated to 110 °C for 6 h. The reaction was cooled to room temperature and was diluted with ethyl acetate (5 mL) and filtered through a pad of celite. The filter cake was rinsed with ethyl acetate (3 mL). The filtrate was concentrated and purified on 4 g silica gold column using hexanes / ethyl acetate to obtain the title compound 116 (20 mg, 45%) as a yellowish solid. MS (ESI, pos.): calc’d for C28H24FN5O4S2, 577.1; found 578.1 (M+H).

[00305] Compound 117: To a solution of compound 116 (20 mg, 0.035 mmol) in acetonitrile (0.5 mL), was added 4 M HCI in dioxane (53 pL, 0.21 mmol). The mixture was heated to 70 °C for 8 h then cooled to room temperature and the volatiles were removed in vacuo. The residue was re-dissolved in 1:1 THF/MeOH (1 mL) and 2 N aqueous sodium hydroxide (105 pL, 0.21 mmol) was added. The mixture was heated to 30 °C for 1 h. The reaction was cooled to room temperature, acidified to pH 4-5 by adding 1 N HCI and diluted with 2-methyl THF (5 mL). Brine (1 mL) was added, and the aqueous layer was discarded. The organic layer was concentrated, and the residue was purified on a 30 x 150 mm Gemini column using 5-95% MeCNTBO (both having 0.05% AcOH). Pure fractions were combined and lyophilized to obtain the title compound 117 (5 mg, 35%) as a fluffy off-white solid. MS (ESI, pos.): calc’d for C20H16FN5O2S, 409.1; found 410.1 (M+H). 1H NMR (500 MHz; DMSO-de): d 12.29 (s, 1H), 8.55 (dd, J= 9.7, 2.9 Hz, 1H), 8.28 (dd, J= 2.8, 1.4 Hz, 1H), 8.23-8.22 (m, 2H), 7.63 (d, J= 6.0 Hz, 1H), 7.44 (d, J= 6.0 Hz, 1H), 2.24 (s, 2H), 2.14 (dd, J = 8.8, 4.3 Hz, 2H), 1.95- 1.92 (m, 4H).

1.21 Scheme 23 [00306] Compound 119 was prepared using the same general procedure as used for compound 13 Yield = 30%. MS (ESI, pos.): calc’d for C35H36FN5O6S2, 705.2; found 706.2 (M+H).

[00307] Compound 120 was prepared following the same general procedure as used for compound 15 Yield = 38% over 3 steps. MS (ESI, pos.): calc’d for C23H22FN5O3S, 467.1; found 468.1 (M+H). ¾-NMR (500 MHz; DMSO-de): d 12.14 (s, 1H), 8.62 (dd, J= 10.0, 2.9 Hz, 1H), 8.20 (dd, .7 = 2.8, 1.3 Hz, 1H), 7.69 (d, J= 6.0 Hz, 1H), 7.63 (d, .7= 7.1 Hz, 1H), 7.45 (d,J= 5.9 Hz, 1H), 5.18 (q, J= 13.4 Hz, 2H), 4.86 (t, J= 7.0 Hz, 1H), 2.81 (d,J= 7.0 Hz, 1H), 2.05 (d, J= 0.7 Hz, 1H), 1.98 (t, J= 1.5 Hz, 1H), 1.90-1.86 (m, 1H), 1.77 (d, J= 9.0 Hz, 2H), 1.62-1.40 (m, 6H).

Example 2: Anti-hemagglutinin non-cytotoxic antibody drug conjugate synthesis

[00308] Anti-hemagglutinin non-cytotoxic antibody drug conjugates were synthesized as described below. [00309] Anti -hemagglutinin (anti -HA) monoclonal antibody mAh 11729 was mutated to introduce a consensus LLQGA pentapeptide sequence at the C-terminus of the heavy chain. A non-HA binding mAh (derived from an immunological antigen having no relation to infectious diseases) containing the same consensus sequence at the C-terminus of the heavy chain was used as a non-binding isotype control. The mutation allowed the antibodies to be enzymatically conjugated to a maximum loading of 2 on the heavy chains (one on each heavy chain).

[00310] Antibodies with a conjugation site at the C-terminus of the heavy chain site were conjugated at 1 mg/mL in PBS pH 7.4. Linker payload compounds were added in a 10-40 fold molar excess over antibody and the enzymatic reaction was initiated by addition of 14 units of bacterial transglutaminase (Zedira, T1001) per mg antibody and incubated at 37 °C for 16 hours. The conjugates were purified by Protein A chromatography (Pierce Protein A Columns, ThermoScientific, product no. 20356). The conjugates were analyzed by ESI-MS for the determination of the payload: antibody ratio (DAR) using a Waters Acquity UPLC. The chromatographic separation was achieved on a C4 column (2.1 X 50 mm ACQUITY UPLC BEH protein C4, 1.7 um, 300 A) in a 10 min gradient (minute: percentage of mobile phase B; 0:10%, 1:10%, 5:90%, 7:90%, 7.2:10%, 10:10%). The mobile phase A was 0.1% formic acid in water, and mobile phase B was 0.1% formic acid in acetonitrile. The flow rate was set at 0.3 mL/min. The detector TOF scan was set from m/z 500-4500 with major parameters as listed (Capillary voltage 3.0 kV; Sampling Cone 80V; Source Offset at 100V; Source temperatures 150 °C; Desolvation temperature 450 °C; Cone gas 0 L/hr; Desolvation gas 800 L/hr). The spectra were deconvoluted with MaxEnt function within MassLynx software. Size-exclusion HPLC established that all conjugates were >92% monomeric (Table 1). This procedure produced a mAbll729-HC-Cterm-linker payload and Isotype Control -HC-Cterm-linker payload conjugates with drug to antibody ratios listed in the following table:

Example 2: Anti-hemagglutinin ADC Activity

[00311] mAb 11729 is a monoclonal antibody that binds the stem domain of group 1 influenza HA molecules, and displays antiviral activity against H1N1 in vitro. VX-787 and derivatives of this molecule have been conjugated to mAb 11729. These conjugates, as well as free payloads, were assayed for anti-influenza activity.

[00312] To test antiviral efficacy, mAb 11729 and ADCs mAb 11729-HC-Cterm-24, mAb 11729-HC-Cterm-32, mAb 11729-HC-Cterm-39, mAb 11729-HC-Cterm-46, mAb 11729-HC- Cterm-69, mAb 11729-HC-Cterm-74b, mAb 11729-HC-Cterm-79, were assayed for their ability to suppress the infection of cells by influenza virus.

[00313] MDCK London cells (IRR) were seeded at 20,000 cells/well in 100 pL of growth media (DMEM containing 1 % sodium pyruvate, 10% Fetal Bovine Serum and 0.5% Gentamicin; Life Technologies) in a 96-well plate. The cells were incubated at 37 °C and 5% C02 for 24 hours. The following day, all antibodies were diluted to a starting concentration of 3440 nM in Trypsin infection media (DMEM containing 1 % sodium pyruvate, 0.21 % Low IgG BSA solution, 1 mg/mL Trypsin TPCK-Treated and 0.5% Gentamicin; Sigma, Life Technologies) and titrated 1:3 to a final concentration of 0.02 nM. Free payloads were diluted in the same infection media to a starting concentration of 6880 nM and titrated 1:3 to a final concentration of 0.04 nM. H1N1 A/Puerto Rico/08/1934 influenza virus that was engineered to express GFP in cells that it infects (“H1N1 A/Puerto Rico/08/1934-GFP”) was diluted to an MOI of one in Trypsin infection media (Life Technologies) and mixed 1:1 with diluted antibody, ADC, or payload. Growth Media was removed from seeded 96-well plates and virus- antibody, virus-ADC, or virus-small molecule mixture was added onto cells at 100 pL per well. Plates were lightly tapped and returned to 37 °C 5% CO2 for 20 hours. Subsequently, plates were washed with PBS and overlayed with 50 pL of PBS. Plates were read immediately for GFP signal on a Molecular Devices Spectramax i3x plate reader.

[00314] VX-787 derivatives 15, 20a, 20b, and 20c displayed antiviral activity. The anti -HA antibody conjugated to the VX-787 linker-payloads 24, 32, 39, and 46 displayed enhanced anti viral potency against influenza A infection compared to the unconjugated antibody. Antiviral activity of VX-787 ADCs:

Antiviral activity of VX-787 free payloads:

SEQUENCE LISTING

<110> Regeneron Pharmaceuticals, Inc. Lisa Purcell Ngambo Jonathan Viau William Olson

<120> Human Antibodies to Influenza Hemagglutinin

<130> 10119US01 <140> US 14/974,361 <141> 2015-12-18

<150> 62/094752 <151> 2014-12-19

<150> 62/152122 <151> 2015-04-24

<160> 6328

<170> FastSEQ for Windows Version 4.0

<210> 1 <211> 375 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic

<400> 1 caggtgcagc tggtgcagtc tggggctgag gtgaagaagc ctgggtcctc ggtgaaggtc 60 tcctgcaagg cttctggagg caccttcagc aactatgcta tcagctgggt gcgacaggcc 120 cctggacaag ggcttgagag gatgggaggg atcatcccta tctttggtac agcaaactac 180 gcacagaagt tccagggcag agtcacgatt accacggacg aatccacgag cacagcctac 240 atggagctgg gcagcctgag atctgaggac acggccgtgt attactgtgc gagagggaag 300 gatttctatg gttcggggag ttattataac ctctttgact actggggcca gggaaccctg 360 gtcaccgtct cctca 375

<210> 2 <211> 125 <212> PRT

<213> Artificial Sequence

<220>

<223> synthetic

<400> 2

Gin Val Gin Leu Val Gin Ser Gly Ala Glu Val Lys Lys Pro Gly Ser 1 5 10 15

Ser Val Lys Val Ser Cys Lys Ala Ser Gly Gly Thr Phe Ser Asn Tyr 20 25 30

Ala lie Ser Trp Val Arg Gin Ala Pro Gly Gin Gly Leu Glu Arg Met 35 40 45

Gly Gly lie lie Pro lie Phe Gly Thr Ala Asn Tyr Ala Gin Lys Phe 50 55 60

Gin Gly Arg Val Thr lie Thr Thr Asp Glu Ser Thr Ser Thr Ala Tyr 65 70 75 80 Met Glu Leu Gly Ser Leu Arg Ser Glu Asp Thr Ala Val Tyr Tyr Cys 85 90 95

Ala Arg Gly Lys Asp Phe Tyr Gly Ser Gly Ser Tyr Tyr Asn Leu Phe 100 105 110

Asp Tyr Trp Gly Gin Gly Thr Leu Val Thr Val Ser Ser 115 120 125

<210> 3 <211> 24 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 3 ggaggcacct tcagcaacta tgct 24

<210> 4 <211> 8 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic <400> 4

Gly Gly Thr Phe Ser Asn Tyr Ala 1 5

<210> 5 <211> 24 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 5 atcatcccta tctttggtac agca 24

<210> 6 <211> 8 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic <400> 6 lie lie Pro lie Phe Gly Thr Ala 1 5

<210> 7 <211> 54 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic

<400> 7 gcgagaggga aggatttcta tggttcgggg agttattata acctctttga ctac 54

<210> 8 <211> 18 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic <400> 8

Ala Arg Gly Lys Asp Phe Tyr Gly Ser Gly Ser Tyr Tyr Asn Leu Phe 1 5 10 15

Asp Tyr

<210> 9 <211> 321 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic

<400> 9 gacatccaga tgacccagtc tccttccacc ctgtctgcat ctgtaggaga catagtcacc 60 atcacttgcc gggccagtca gagtattagt agctggttgg cctggtatca gcagaaacca 120 gggaaagccc ctaagctcct gatctataag gcgtctagtt tacaaagtgg ggtcccatca 180 aggttcagcg gcagtggatc tgggacagaa ttcactctca ccatcagcag cctgcagcct 240 gatgattttg caacttttta ctgccaacac tataatactt attcgtggac gttcggccaa 300 gggaccaagg tggaaatcaa a 321

<210> 10 <211> 107 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic

<400> 10 Asp lie Gin Met Thr Gin Ser Pro Ser Thr Leu Ser Ala Ser Val Gly 1 5 10 15

Asp lie Val Thr lie Thr Cys Arg Ala Ser Gin Ser lie Ser Ser Trp 20 25 30

Leu Ala Trp Tyr Gin Gin Lys Pro Gly Lys Ala Pro Lys Leu Leu lie 35 40 45

Tyr Lys Ala Ser Ser Leu Gin Ser Gly Val Pro Ser Arg Phe Ser Gly 50 55 60

Ser Gly Ser Gly Thr Glu Phe Thr Leu Thr lie Ser Ser Leu Gin Pro 65 70 75 80

Asp Asp Phe Ala Thr Phe Tyr Cys Gin His Tyr Asn Thr Tyr Ser Trp 85 90 95

Thr Phe Gly Gin Gly Thr Lys Val Glu lie Lys 100 105 <210> 11 <211> 18 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 11 cagagtatta gtagctgg 18

<210> 12 <211> 6 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic <400> 12

Gin Ser lie Ser Ser Trp 1 5

<210> 13 <211> 9 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 13 aaggcgtct 9

<210> 14 <211> 3 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic

<400> 14 Lys Ala Ser 1

<210> 15 <211> 27 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 15 caacactata atacttattc gtggacg 27

<210> 16 <211> 9 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic <400> 16

Gin His Tyr Asn Thr Tyr Ser Trp Thr 1 5

<210> 17 <211> 360 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic

<400> 17 caggtgcagc tggtgcagtc tggggctgag gtgaagaagt ctgggtcctc ggtgaaggtc 60 tcctgcaagg cttctggagg caccttcagc agctatgcta ttagctgggt gcgccaggcc 120 cctggacaag ggcttgagtg gatgggaggg atcatcccta tttttggtac accaagctat 180 gcacagaagt tccaggacag agtcacgatt accacggacg aatccacgag cacagtttac 240 atggagctga gcagcctgag atctgaagac acggccgtgt attactgtgc gagacagcag 300 cccgtctacc agtacaatat ggacgtctgg ggccaaggga ccacggtcac cgtctcctca 360

<210> 18 <211> 120 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic <400> 18

Gin Val Gin Leu Val Gin Ser Gly Ala Glu Val Lys Lys Ser Gly Ser 1 5 10 15

Ser Val Lys Val Ser Cys Lys Ala Ser Gly Gly Thr Phe Ser Ser Tyr 20 25 30

Ala lie Ser Trp Val Arg Gin Ala Pro Gly Gin Gly Leu Glu Trp Met 35 40 45

Gly Gly lie lie Pro lie Phe Gly Thr Pro Ser Tyr Ala Gin Lys Phe 50 55 60

Gin Asp Arg Val Thr lie Thr Thr Asp Glu Ser Thr Ser Thr Val Tyr 65 70 75 80

Met Glu Leu Ser Ser Leu Arg Ser Glu Asp Thr Ala Val Tyr Tyr Cys 85 90 95

Ala Arg Gin Gin Pro Val Tyr Gin Tyr Asn Met Asp Val Trp Gly Gin 100 105 110

Gly Thr Thr Val Thr Val Ser Ser 115 120

<210> 19 <211> 24 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 19 ggaggcacct tcagcagcta tgct 24

<210> 20 <211> 8 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic <400> 20

Gly Gly Thr Phe Ser Ser Tyr Ala 1 5

<210> 21 <211> 24 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 21 atcatcccta tttttggtac acca 24

<210> 22 <211> 8 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic <400> 22 lie lie Pro lie Phe Gly Thr Pro 1 5

<210> 23 <211> 39 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 23 gcgagacagc agcccgtcta ccagtacaat atggacgtc 39

<210> 24 <211> 13 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic <400> 24

Ala Arg Gin Gin Pro Val Tyr Gin Tyr Asn Met Asp Val 1 5 10

<210> 25 <211> 321 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic

<400> 25 gacatccaga tgacccagtc tccatcctcc ctgtctgcat ctgtaggaga cagagtcacc 60 atcacttgcc gggcaagtca gggcattaga aataatttag gctggtatca gcagaaacca 120 ctgaaagccc ctaagcgcct gatctatgct gcgtccagtt tgcaaagtgg ggtcccatca 180 aggttcagcg gcagtggatc tgggacagaa ttcactctca caatcagcag cctgcagcct 240 gaagattttg caacttatta ctgtctacaa tataataatt acccgtggac gttcggccaa 300 gggaccaagg tggaaatcaa a 321

<210> 26 <211> 107 <212> PRT <213> Artificial Sequence

<220>

<223> synthetic

<400> 26 Asp lie Gin Met Thr Gin Ser Pro Ser Ser Leu Ser Ala Ser Val Gly 1 5 10 15

Asp Arg Val Thr lie Thr Cys Arg Ala Ser Gin Gly lie Arg Asn Asn 20 25 30

Leu Gly Trp Tyr Gin Gin Lys Pro Leu Lys Ala Pro Lys Arg Leu lie 35 40 45

Tyr Ala Ala Ser Ser Leu Gin Ser Gly Val Pro Ser Arg Phe Ser Gly 50 55 60

Ser Gly Ser Gly Thr Glu Phe Thr Leu Thr lie Ser Ser Leu Gin Pro 65 70 75 80

Glu Asp Phe Ala Thr Tyr Tyr Cys Leu Gin Tyr Asn Asn Tyr Pro Trp 85 90 95

Thr Phe Gly Gin Gly Thr Lys Val Glu lie Lys 100 105

<210> 27 <211> 18 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 27 cagggcatta gaaataat 18

<210> 28 <211> 6 <212> PRT <213> Artificial Sequence <220>

<223> synthetic <400> 28

Gin Gly lie Arg Asn Asn 1 5

<210> 29 <211> 9 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 29 gctgcgtcc 9

<210> 30 <211> 3 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic

<400> 30 Ala Ala Ser 1

<210> 31 <211> 27 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 31 ctacaatata ataattaccc gtggacg 27

<210> 32 <211> 9 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic <400> 32

Leu Gin Tyr Asn Asn Tyr Pro Trp Thr 1 5

<210> 33 <211> 366 <212> DNA <213> Artificial Sequence <220>

<223> synthetic

<400> 33 caggtgcagc tggtgcagtc tggggctgaa gtgaagaagc ctgggtcctc ggtgaaggtc 60 tcctgcaagg cttctggagg caccttcaac agctatgcta tcagctgggt gcgacaggcc 120 cctggacaag ggcttgagtg gatgggaggc atcatcccta tctttgctac aacaaacttc 180 gcacagaagt tccagggcag agtcacgatt acctcggacg aatccacgaa cacagcctac 240 atggagctga ccagcctgag atctgaggac acggccgcgt attactgtgc aagggggggt 300 tggtataact ggcagtacgt ggggtttgac tcctggggcc aagggaccac ggtcaccgtc 360 tcctca 366

<210> 34 <211> 122 <212> PRT <213> Artificial Sequence

<220>

<223> synthetic

<400> 34 Gin Val Gin Leu Val Gin Ser Gly Ala Glu Val Lys Lys Pro Gly Ser 1 5 10 15

Ser Val Lys Val Ser Cys Lys Ala Ser Gly Gly Thr Phe Asn Ser Tyr 20 25 30

Ala lie Ser Trp Val Arg Gin Ala Pro Gly Gin Gly Leu Glu Trp Met 35 40 45

Gly Gly lie lie Pro lie Phe Ala Thr Thr Asn Phe Ala Gin Lys Phe 50 55 60

Gin Gly Arg Val Thr lie Thr Ser Asp Glu Ser Thr Asn Thr Ala Tyr 65 70 75 80

Met Glu Leu Thr Ser Leu Arg Ser Glu Asp Thr Ala Ala Tyr Tyr Cys 85 90 95

Ala Arg Gly Gly Trp Tyr Asn Trp Gin Tyr Val Gly Phe Asp Ser Trp 100 105 110

Gly Gin Gly Thr Thr Val Thr Val Ser Ser 115 120

<210> 35 <211> 24 <212> DNA <213> Artificial Sequence

<220>

<223> synthetic <400> 35 ggaggcacct tcaacagcta tgct 24

<210> 36 <211> 8 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic <400> 36 Gly Gly Thr Phe Asn Ser Tyr Ala 1 5

<210> 37 <211> 24 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 37 atcatcccta tctttgctac aaca 24

<210> 38 <211> 8 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic <400> 38 lie lie Pro lie Phe Ala Thr Thr 1 5

<210> 39 <211> 45 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 39 gcaagggggg gttggtataa ctggcagtac gtggggtttg actcc 45

<210> 40 <211> 15 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic <400> 40

Ala Arg Gly Gly Trp Tyr Asn Trp Gin Tyr Val Gly Phe Asp Ser 1 5 10 15

<210> 41 <211> 321 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 41 gacatccaga tgacccagtc tccttccacc ctgtctgcat ctgtaggaga cagagtcgcc 60 atcacttgcc gggccagtca gagtattagt acctggttgg cctggtatca gcagaaacca 120 gggaaagccc ctaagctcct gatctataag gcgtctagtt tagaaaatgg ggtcccttca 180 aggttcagcg gcagtggatc tgggacagat ttcactctca ccatcagcag cctgcaggct 240 gatgattttg caacttatta ctgccaaaag tataatagta attcgtggac gttcggccaa 300 gggaccaagg tggatatcaa a 321

<210> 42 <211> 107 <212> PRT

<213> Artificial Sequence

<220>

<223> synthetic

<400> 42 Asp lie Gin Met Thr Gin Ser Pro Ser Thr Leu Ser Ala Ser Val Gly 1 5 10 15

Asp Arg Val Ala lie Thr Cys Arg Ala Ser Gin Ser lie Ser Thr Trp 20 25 30

Leu Ala Trp Tyr Gin Gin Lys Pro Gly Lys Ala Pro Lys Leu Leu lie 35 40 45

Tyr Lys Ala Ser Ser Leu Glu Asn Gly Val Pro Ser Arg Phe Ser Gly 50 55 60

Ser Gly Ser Gly Thr Asp Phe Thr Leu Thr lie Ser Ser Leu Gin Ala 65 70 75 80

Asp Asp Phe Ala Thr Tyr Tyr Cys Gin Lys Tyr Asn Ser Asn Ser Trp 85 90 95

Thr Phe Gly Gin Gly Thr Lys Val Asp lie Lys 100 105

<210> 43 <211> 18 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 43 cagagtatta gtacctgg 18

<210> 44 <211> 6 <212> PRT

<213> Artificial Sequence

<220>

<223> synthetic <400> 44

Gin Ser lie Ser Thr Trp 1 5

<210> 45 <211> 9 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 45 aaggcgtct 9

<210> 46 <211> 3 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic

<400> 46 Lys Ala Ser 1

<210> 47 <211> 27 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 47 caaaagtata atagtaattc gtggacg 27

<210> 48 <211> 9 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic <400> 48

Gin Lys Tyr Asn Ser Asn Ser Trp Thr 1 5

<210> 49 <211> 360 <212> DNA

<213> Artificial Sequence

<220>

<223> synthetic

<400> 49 caggtccacc tggtgcagtc tgggccagag gtgaagaagc ctgggtcctc ggtgaaggtc 60 tcctgcaagg cttctggagt caccttcatc agtcatgcta tcagctgggt gcgacaggcc 120 cctggacaag ggcttgaatg ggtgggagga atcatcgcta tctttggtac aacaaactac 180 gcacagaagt tccagggcag agtcacggtt acaacggaca aatccacgaa cacagtctac 240 atggaattga gcagactgag atctgaggac acggccattt attactgtgc gcgaggtgag 300 acctactacg agggaaactt tgacttctgg ggccagggaa ccctggtcac cgtctcctca 360

<210> 50 <211> 120 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic <400> 50

Gin Val His Leu Val Gin Ser Gly Pro Glu Val Lys Lys Pro Gly Ser 1 5 10 15

Ser Val Lys Val Ser Cys Lys Ala Ser Gly Val Thr Phe lie Ser His 20 25 30

Ala lie Ser Trp Val Arg Gin Ala Pro Gly Gin Gly Leu Glu Trp Val 35 40 45

Gly Gly lie lie Ala lie Phe Gly Thr Thr Asn Tyr Ala Gin Lys Phe 50 55 60

Gin Gly Arg Val Thr Val Thr Thr Asp Lys Ser Thr Asn Thr Val Tyr 65 70 75 80

Met Glu Leu Ser Arg Leu Arg Ser Glu Asp Thr Ala lie Tyr Tyr Cys 85 90 95

Ala Arg Gly Glu Thr Tyr Tyr Glu Gly Asn Phe Asp Phe Trp Gly Gin 100 105 110

Gly Thr Leu Val Thr Val Ser Ser 115 120

<210> 51 <211> 24 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 51 ggagtcacct tcatcagtca tgct 24

<210> 52 <211> 8 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic

<400> 52

Gly Val Thr Phe lie Ser His Ala 1 5

<210> 53 <211> 24 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 53 atcatcgcta tctttggtac aaca 24 <210> 54 <211> 8 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic <400> 54 lie lie Ala lie Phe Gly Thr Thr 1 5

<210> 55 <211> 39 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 55 gcgcgaggtg agacctacta cgagggaaac tttgacttc 39

<210> 56 <211> 13 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic <400> 56

Ala Arg Gly Glu Thr Tyr Tyr Glu Gly Asn Phe Asp Phe 1 5 10

<210> 57 <211> 324 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic

<400> 57 gtcatccaga tgacccagtc tccatcctcc ctgtctgcat ctgtaggaga cagagtcacc 60 atcacttgcc gggccagtca gagcgttagg agtaatttaa attggtatca gcagacacca 120 gggaaagccc ctaggctcct gatctatgct gcatccagtt tgcaaagtgg ggtcccatca 180 aggttcagtg gcagtggatc tgggacagat ttcactctca ccatcagcag tctgcaacct 240 gaagattttg caacttatta ctgtcaacag agttacagta cccctccgat caccttcggc 300 caagggacac gactggagat taaa 324

<210> 58 <211> 108 <212> PRT <213> Artificial Sequence

<220>

<223> synthetic <400> 58 Val lie Gin Met Thr Gin Ser Pro Ser Ser Leu Ser Ala Ser Val Gly 1 5 10 15

Asp Arg Val Thr lie Thr Cys Arg Ala Ser Gin Ser Val Arg Ser Asn 20 25 30

Leu Asn Trp Tyr Gin Gin Thr Pro Gly Lys Ala Pro Arg Leu Leu lie 35 40 45

Tyr Ala Ala Ser Ser Leu Gin Ser Gly Val Pro Ser Arg Phe Ser Gly 50 55 60

Ser Gly Ser Gly Thr Asp Phe Thr Leu Thr lie Ser Ser Leu Gin Pro 65 70 75 80

Glu Asp Phe Ala Thr Tyr Tyr Cys Gin Gin Ser Tyr Ser Thr Pro Pro 85 90 95 lie Thr Phe Gly Gin Gly Thr Arg Leu Glu lie Lys 100 105

<210> 59 <211> 18 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 59 cagagcgtta ggagtaat 18

<210> 60 <211> 6 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic <400> 60

Gin Ser Val Arg Ser Asn 1 5

<210> 61 <211> 9 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 61 gctgcatcc 9

<210> 62 <211> 3 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic <400> 62 Ala Ala Ser 1

<210> 63 <211> 30 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 63 caacagagtt acagtacccc tccgatcacc 30

<210> 64 <211> 10 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic <400> 64

Gin Gin Ser Tyr Ser Thr Pro Pro lie Thr 1 5 10

<210> 65 <211> 324 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic

<400> 65 gacatccaga tgacccagtc tccatcctcc ctgtctgcat ctgtaggaga cagagtcacc 60 atcacttgcc gggcaagtca gagcattagc agctatttaa attggtatca gcagaaacca 120 gggaaagccc ctaagctcct gatctatgct gcatccagtt tgcaaagtgg ggtcccgtca 180 aggttcagtg gcagtggatc tgggacagat ttcactctca ccatcagcag tctgcaacct 240 gaagattttg caacttacta ctgtcaacag agttacagta cccctccgat caccttcggc 300 caagggacac gactggagat taaa 324

<210> 66 <211> 108 <212> PRT

<213> Artificial Sequence

<220>

<223> synthetic

<400> 66

Asp lie Gin Met Thr Gin Ser Pro Ser Ser Leu Ser Ala Ser Val Gly 1 5 10 15

Asp Arg Val Thr lie Thr Cys Arg Ala Ser Gin Ser lie Ser Ser Tyr 20 25 30

Leu Asn Trp Tyr Gin Gin Lys Pro Gly Lys Ala Pro Lys Leu Leu lie 35 40 45

Tyr Ala Ala Ser Ser Leu Gin Ser Gly Val Pro Ser Arg Phe Ser Gly 50 55 60 Ser Gly Ser Gly Thr Asp Phe Thr Leu Thr lie Ser Ser Leu Gin Pro 65 70 75 80

Glu Asp Phe Ala Thr Tyr Tyr Cys Gin Gin Ser Tyr Ser Thr Pro Pro 85 90 95 lie Thr Phe Gly Gin Gly Thr Arg Leu Glu lie Lys 100 105

<210> 67 <211> 18 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 67 cagagcatta gcagctat 18

<210> 68 <211> 6 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic <400> 68

Gin Ser lie Ser Ser Tyr 1 5

<210> 69 <211> 9 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 69 gctgcatcc 9

<210> 70 <211> 3 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic

<400> 70 Ala Ala Ser 1

<210> 71 <211> 30 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 71 caacagagtt acagtacccc tccgatcacc 30

<210> 72 <211> 10 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic <400> 72

Gin Gin Ser Tyr Ser Thr Pro Pro lie Thr 1 5 10

<210> 73 <211> 369 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic

<400> 73 caggtgcagc tggtgcagtc tggagcagag gtgaggaagc ctgggtcctc ggtgaaggtc 60 tcctgcaagg cttctggagg aaccttcacc ggccatgcta tcagctgggt gcgacaggcc 120 cctggacaag gccttgagtg gatgggaggg gtcgtcccta tctttggttc agcaaactac 180 gcacagaagt tccagggcag agtcacgatg accatggacg aatccacgag tacagcctac 240 atggacctga gcagcctgag atctgaggac acggccgttt attattgtgt gagagatccg 300 ggcaactcgg gatactacta ctacggtatg gacgtctggg gccaagggac cacggtcacc 360 gtctcctca 369

<210> 74 <211> 123 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic

<400> 74 Gin Val Gin Leu Val Gin Ser Gly Ala Glu Val Arg Lys Pro Gly Ser 1 5 10 15

Ser Val Lys Val Ser Cys Lys Ala Ser Gly Gly Thr Phe Thr Gly His 20 25 30

Ala lie Ser Trp Val Arg Gin Ala Pro Gly Gin Gly Leu Glu Trp Met 35 40 45

Gly Gly Val Val Pro lie Phe Gly Ser Ala Asn Tyr Ala Gin Lys Phe 50 55 60

Gin Gly Arg Val Thr Met Thr Met Asp Glu Ser Thr Ser Thr Ala Tyr 65 70 75 80

Met Asp Leu Ser Ser Leu Arg Ser Glu Asp Thr Ala Val Tyr Tyr Cys 85 90 95

Val Arg Asp Pro Gly Asn Ser Gly Tyr Tyr Tyr Tyr Gly Met Asp Val 100 105 110

Trp Gly Gin Gly Thr Thr Val Thr Val Ser Ser 115 120 <210> 75 <211> 24 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 75 ggaggaacct tcaccggcca tgct 24

<210> 76 <211> 8 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic <400> 76

Gly Gly Thr Phe Thr Gly His Ala 1 5

<210> 77 <211> 24 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 77 gtcgtcccta tctttggttc agca 24

<210> 78 <211> 8 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic <400> 78

Val Val Pro lie Phe Gly Ser Ala 1 5

<210> 79 <211> 48 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 79 gtgagagatc cgggcaactc gggatactac tactacggta tggacgtc 48 <210> 80 <211> 16 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic <400> 80

Val Arg Asp Pro Gly Asn Ser Gly Tyr Tyr Tyr Tyr Gly Met Asp Val 1 5 10 15

<210> 81 <211> 324 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic

<400> 81 gacatccaga tgacccagtc tccatcctcc ctgtctgcat ctgtaggaga cagagtcacc 60 atcacttgcc gggcaagtca gagcattagc agctatttaa attggtttca gcagaaacca 120 gggaaagccc ctaagctcct gatctatact gcatccagtt tgcaaagtgg ggtcccatca 180 aggttcagtg gcagtggatc tgggacagat ttcactctca ccatcagcag tctgcaacct 240 gaagattttg caacttacta ctgtcaacag agttacagtt cccctccgat caccttcggc 300 caagggacac gactggagat taaa 324

<210> 82 <211> 108 <212> PRT <213> Artificial Sequence

<220>

<223> synthetic

<400> 82 Asp lie Gin Met Thr Gin Ser Pro Ser Ser Leu Ser Ala Ser Val Gly 1 5 10 15

Asp Arg Val Thr lie Thr Cys Arg Ala Ser Gin Ser lie Ser Ser Tyr 20 25 30

Leu Asn Trp Phe Gin Gin Lys Pro Gly Lys Ala Pro Lys Leu Leu lie 35 40 45

Tyr Thr Ala Ser Ser Leu Gin Ser Gly Val Pro Ser Arg Phe Ser Gly 50 55 60

Ser Gly Ser Gly Thr Asp Phe Thr Leu Thr lie Ser Ser Leu Gin Pro 65 70 75 80

Glu Asp Phe Ala Thr Tyr Tyr Cys Gin Gin Ser Tyr Ser Ser Pro Pro 85 90 95 lie Thr Phe Gly Gin Gly Thr Arg Leu Glu lie Lys 100 105

<210> 83 <211> 18 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 83 cagagcatta gcagctat 18

<210> 84 <211> 6 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic <400> 84

Gin Ser lie Ser Ser Tyr 1 5

<210> 85 <211> 9 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 85 actgcatcc 9

<210> 86 <211> 3 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic

<400> 86 Thr Ala Ser 1

<210> 87 <211> 30 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 87 caacagagtt acagttcccc tccgatcacc 30

<210> 88 <211> 10 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic <400> 88 Gin Gin Ser Tyr Ser Ser Pro Pro lie Thr 1 5 10

<210> 89 <211> 369 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic

<400> 89 caggtccacc tggtgcagtc tggggctgag gtgaagaagc ctgggtcgtc ggtgaaggtc 60 tcctgcaagg cttctggagg caccttcagc acctatgcta tcacctgggt gcgacaggcc 120 cctggacaag ggcttgagtg gataggaggg atcagccgtt tctttggttc agcaaactac 180 gcacagaagt ttcagggcag agtcacaatt accacggacg aatccacgaa cacagcctac 240 atggaactaa gcagcctgag atctgaggac acggccgtat attattgtgc gagagatcct 300 ggaaacacgg gctattattt ttacggtatg gacgtctggg gccaagggac cacggtcacc 360 gtctcctca 369

<210> 90 <211> 123 <212> PRT <213> Artificial Sequence

<220>

<223> synthetic

<400> 90 Gin Val His Leu Val Gin Ser Gly Ala Glu Val Lys Lys Pro Gly Ser 1 5 10 15

Ser Val Lys Val Ser Cys Lys Ala Ser Gly Gly Thr Phe Ser Thr Tyr

20 25 30

Ala lie Thr Trp Val Arg Gin Ala Pro Gly Gin Gly Leu Glu Trp lie 35 40 45

Gly Gly lie Ser Arg Phe Phe Gly Ser Ala Asn Tyr Ala Gin Lys Phe 50 55 60

Gin Gly Arg Val Thr lie Thr Thr Asp Glu Ser Thr Asn Thr Ala Tyr 65 70 75 80

Met Glu Leu Ser Ser Leu Arg Ser Glu Asp Thr Ala Val Tyr Tyr Cys 85 90 95

Ala Arg Asp Pro Gly Asn Thr Gly Tyr Tyr Phe Tyr Gly Met Asp Val

100 105 110

Trp Gly Gin Gly Thr Thr Val Thr Val Ser Ser 115 120

<210> 91 <211> 24 <212> DNA <213> Artificial Sequence

<220>

<223> synthetic <400> 91 ggaggcacct tcagcaccta tgct 24

<210> 92 <211> 8 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic <400> 92

Gly Gly Thr Phe Ser Thr Tyr Ala 1 5

<210> 93 <211> 24 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 93 atcagccgtt tctttggttc agca 24

<210> 94 <211> 8 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic <400> 94 lie Ser Arg Phe Phe Gly Ser Ala 1 5

<210> 95 <211> 48 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 95 gcgagagatc ctggaaacac gggctattat ttttacggta tggacgtc 48

<210> 96 <211> 16 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic <400> 96

Ala Arg Asp Pro Gly Asn Thr Gly Tyr Tyr Phe Tyr Gly Met Asp Val 1 5 10 15

<210> 97 <211> 324 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic

<400> 97 gacatccaga tgacccagtc tccatcctcc ctgtctgcat ctgtaggaga cagagtcacc 60 atcacttgcc gggcaagtca gagcattagc agctatttaa attggtatca gcagaaacca 120 gggaaagccc ctaagctcct gatctatggt gcatccagtt tgcaaagtgg ggtcccatca 180 aggttcagtg gcagtggatc tgggacagat ttcactctca ccatcagcag tctgcaacct 240 gaagattttg caacttacta ctgtcaacag agtaagagta attctccgat caccttcggc 300 caagggacac gactggagat taaa 324

<210> 98 <211> 108 <212> PRT <213> Artificial Sequence

<220>

<223> synthetic

<400> 98 Asp lie Gin Met Thr Gin Ser Pro Ser Ser Leu Ser Ala Ser Val Gly 1 5 10 15

Asp Arg Val Thr lie Thr Cys Arg Ala Ser Gin Ser lie Ser Ser Tyr 20 25 30

Leu Asn Trp Tyr Gin Gin Lys Pro Gly Lys Ala Pro Lys Leu Leu lie 35 40 45

Tyr Gly Ala Ser Ser Leu Gin Ser Gly Val Pro Ser Arg Phe Ser Gly 50 55 60

Ser Gly Ser Gly Thr Asp Phe Thr Leu Thr lie Ser Ser Leu Gin Pro 65 70 75 80

Glu Asp Phe Ala Thr Tyr Tyr Cys Gin Gin Ser Lys Ser Asn Ser Pro 85 90 95 lie Thr Phe Gly Gin Gly Thr Arg Leu Glu lie Lys 100 105

<210> 99 <211> 18 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 99 cagagcatta gcagctat 18

<210> 100 <211> 6 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic <400> 100

Gin Ser lie Ser Ser Tyr 1 5 <210> 101 <211> 9 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 101 ggtgcatcc 9

<210> 102 <211> 3 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic

<400> 102 Gly Ala Ser 1

<210> 103 <211> 30 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 103 caacagagta agagtaattc tccgatcacc 30

<210> 104 <211> 10 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic <400> 104

Gin Gin Ser Lys Ser Asn Ser Pro lie Thr 1 5 10

<210> 105 <211> 369 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 105 caggtccagc tggtgcagtc tggggctgag gtgaagaagc ctgggtcctc ggtgaaggtc 60 tcctgcaagg cttctggagg caccttcatc acctatgcta tcagctgggt gcgacaggcc 120 cctggacaag ggcttgagtg gatgggaggt gtcatcccta tctttggtac tccaaggtac 180 gcacagaagt tccagggcag agtcacgatt accacggacg attccacgac cacagcctac 240 atggagctga gcagcctgag atctgacgac acggccgtgt attattgtgc gacaaggagc 300 agctcgtctc cctattatta ctacggtatg gacgtctggg gccaagggac cacggtcacc 360 gtctcctca 369

<210> 106 <211> 123 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic

<400> 106 Gin Val Gin Leu Val Gin Ser Gly Ala Glu Val Lys Lys Pro Gly Ser 1 5 10 15

Ser Val Lys Val Ser Cys Lys Ala Ser Gly Gly Thr Phe lie Thr Tyr 20 25 30

Ala lie Ser Trp Val Arg Gin Ala Pro Gly Gin Gly Leu Glu Trp Met 35 40 45

Gly Gly Val lie Pro lie Phe Gly Thr Pro Arg Tyr Ala Gin Lys Phe 50 55 60

Gin Gly Arg Val Thr lie Thr Thr Asp Asp Ser Thr Thr Thr Ala Tyr 65 70 75 80

Met Glu Leu Ser Ser Leu Arg Ser Asp Asp Thr Ala Val Tyr Tyr Cys 85 90 95

Ala Thr Arg Ser Ser Ser Ser Pro Tyr Tyr Tyr Tyr Gly Met Asp Val 100 105 110

Trp Gly Gin Gly Thr Thr Val Thr Val Ser Ser 115 120

<210> 107 <211> 24 <212> DNA <213> Artificial Sequence

<220>

<223> synthetic <400> 107 ggaggcacct tcatcaccta tgct 24

<210> 108 <211> 8 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic

<400> 108

Gly Gly Thr Phe lie Thr Tyr Ala 1 5

<210> 109 <211> 24 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 109 gtcatcccta tctttggtac tcca 24

<210> 110 <211> 8 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic <400> 110

Val lie Pro lie Phe Gly Thr Pro 1 5

<210> 111 <211> 48 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 111 gcgacaagga gcagctcgtc tccctattat tactacggta tggacgtc 48

<210> 112 <211> 16 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic <400> 112

Ala Thr Arg Ser Ser Ser Ser Pro Tyr Tyr Tyr Tyr Gly Met Asp Val 1 5 10 15

<210> 113 <211> 336 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic

<400> 113 gatattgtga tgacccagac tccactctcc tcacctgtca cccttggaca gccggcctcc 60 atctcctgca ggtctagtca aagcctcgta cacagtgatg gaaacaccta tttgagttgg 120 cttcagcaga ggccaggcca gcctccaaga ctcctaattt ataagatttc taaccggttc 180 tctggggtcc cagacagatt cagtggcagt ggggcaggga cagatttcac actgaaaatc 240 agcagggtgg aagctgagga tgtcgggatt tattactgca ttcaagctac acaatttccg 300 tacacttttg gccaggggac caagctggag atcaaa 336

<210> 114 <211> 112 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic

<400> 114 Asp lie Val Met Thr Gin Thr Pro Leu Ser Ser Pro Val Thr Leu Gly 1 5 10 15

Gin Pro Ala Ser lie Ser Cys Arg Ser Ser Gin Ser Leu Val His Ser 20 25 30

Asp Gly Asn Thr Tyr Leu Ser Trp Leu Gin Gin Arg Pro Gly Gin Pro 35 40 45

Pro Arg Leu Leu lie Tyr Lys lie Ser Asn Arg Phe Ser Gly Val Pro 50 55 60

Asp Arg Phe Ser Gly Ser Gly Ala Gly Thr Asp Phe Thr Leu Lys lie 65 70 75 80

Ser Arg Val Glu Ala Glu Asp Val Gly lie Tyr Tyr Cys lie Gin Ala 85 90 95 Thr Gin Phe Pro Tyr Thr Phe Gly Gin Gly Thr Lys Leu Glu lie Lys 100 105 110

<210> 115 <211> 33 <212> DNA <213> Artificial Sequence

<220>

<223> synthetic <400> 115 caaagcctcg tacacagtga tggaaacacc tat 33

<210> 116 <211> 11 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic <400> 116

Gin Ser Leu Val His Ser Asp Gly Asn Thr Tyr 1 5 10

<210> 117 <211> 9 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 117 aagatttct 9

<210> 118 <211> 3 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic

<400> 118 Lys lie Ser 1

<210> 119 <211> 27 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 119 attcaagcta cacaatttcc gtacact 27

<210> 120 <211> 9 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic <400> 120 lie Gin Ala Thr Gin Phe Pro Tyr Thr 1 5

<210> 121 <211> 372 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic

<400> 121 caggtgcagc tggtgcagtc tggggctgag gtgaagaagc ctggggcctc agtgacggtc 60 tcctgcaagg cttctggata caccttcacc aattatgata tcaactgggt gcgacaggcc 120 actggacaag gacttgagtg gttgggatgg atgagcccta acagtggtaa caaaggctat 180 gcacagaagt tccagggcag agtctccatg actttgaata ccgccttaag cacagcctac 240 atggaactga gcagcctgag atctgaggac acggccgtgt attactgtgc gagatggtat 300 tgtagtgatg ccagttgcta tcccgatgct tttgatatct ggggccaagg gacaatggtc 360 accgtctctt ca 372

<210> 122 <211> 124 <212> PRT <213> Artificial Sequence

<220>

<223> synthetic <400> 122 Gin Val Gin Leu Val Gin Ser Gly Ala Glu Val Lys Lys Pro Gly Ala 1 5 10 15

Ser Val Thr Val Ser Cys Lys Ala Ser Gly Tyr Thr Phe Thr Asn Tyr 20 25 30

Asp lie Asn Trp Val Arg Gin Ala Thr Gly Gin Gly Leu Glu Trp Leu 35 40 45

Gly Trp Met Ser Pro Asn Ser Gly Asn Lys Gly Tyr Ala Gin Lys Phe 50 55 60

Gin Gly Arg Val Ser Met Thr Leu Asn Thr Ala Leu Ser Thr Ala Tyr 65 70 75 80

Met Glu Leu Ser Ser Leu Arg Ser Glu Asp Thr Ala Val Tyr Tyr Cys 85 90 95

Ala Arg Trp Tyr Cys Ser Asp Ala Ser Cys Tyr Pro Asp Ala Phe Asp 100 105 110 lie Trp Gly Gin Gly Thr Met Val Thr Val Ser Ser 115 120

<210> 123 <211> 24 <212> DNA <213> Artificial Sequence

<220>

<223> synthetic <400> 123 ggatacacct tcaccaatta tgat 24

<210> 124 <211> 8 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic <400> 124

Gly Tyr Thr Phe Thr Asn Tyr Asp 1 5

<210> 125 <211> 24 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 125 atgagcccta acagtggtaa caaa 24

<210> 126 <211> 8 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic <400> 126

Met Ser Pro Asn Ser Gly Asn Lys 1 5

<210> 127 <211> 51 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 127 gcgagatggt attgtagtga tgccagttgc tatcccgatg cttttgatat c 51

<210> 128 <211> 17 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic <400> 128

Ala Arg Trp Tyr Cys Ser Asp Ala Ser Cys Tyr Pro Asp Ala Phe Asp 1 5 10 15

He

<210> 129 <211> 321 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic

<400> 129 gacatccaga tgacccagtc tccatcttcc gtgtctgcat ctgtaggaga cagagtcacc 60 atcacttgtc gggcgagtca ggatttttcc gactggttag cctggtatca gcagaaacct 120 gggaaagccc ctgagctcct gatctatgct acatccagtt tgcatactgg ggtcccatca 180 aggttcagcg gcagtggatc tgggacagat ttcactctca ccatcagcac cctgcagcct 240 gaagattttg ctacttacta ttgtctacag gctaacaatt tcccgctcac tttcggcgga 300 gggaccaagg tggagatcaa a 321

<210> 130 <211> 107 <212> PRT <213> Artificial Sequence

<220>

<223> synthetic <400> 130

Asp lie Gin Met Thr Gin Ser Pro Ser Ser Val Ser Ala Ser Val Gly 1 5 10 15

Asp Arg Val Thr He Thr Cys Arg Ala Ser Gin Asp Phe Ser Asp Trp 20 25 30

Leu Ala Trp Tyr Gin Gin Lys Pro Gly Lys Ala Pro Glu Leu Leu He 35 40 45

Tyr Ala Thr Ser Ser Leu His Thr Gly Val Pro Ser Arg Phe Ser Gly 50 55 60

Ser Gly Ser Gly Thr Asp Phe Thr Leu Thr lie Ser Thr Leu Gin Pro 65 70 75 80

Glu Asp Phe Ala Thr Tyr Tyr Cys Leu Gin Ala Asn Asn Phe Pro Leu 85 90 95

Thr Phe Gly Gly Gly Thr Lys Val Glu lie Lys 100 105

<210> 131 <211> 18 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 131 caggattttt ccgactgg 18

<210> 132 <211> 6 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic

<400> 132

Gin Asp Phe Ser Asp Trp 1 5

<210> 133 <211> 9 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 133 gctacatcc 9

<210> 134 <211> 3 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic

<400> 134 Ala Thr Ser 1

<210> 135 <211> 27 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 135 ctacaggcta acaatttccc gctcact 27

<210> 136 <211> 9 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic <400> 136

Leu Gin Ala Asn Asn Phe Pro Leu Thr 1 5

<210> 137 <211> 366 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic

<400> 137 caggtgcagc tggtggagtc tgggggaggc gtggcccagc ctgggaggtc cctgagactc 60 tcctgtgcag cctctggaat caccttcaat aactatggca tgcactgggt ccgccaggct 120 ccaggcaagg ggctggagtg ggtggcattt atttcagatg aaggaagaaa taaacactat 180 ggagactccg tgaagggccg attcaccatc gacagagaca attccaagaa cacactgtat 240 ctgcaaatga atagcctgag agctgaggac acggctgtat attactgtgc gaaattgggg 300 gataatagga atcaccacta cggtttggac gtctggggcc aagggaccac ggtcaccgtc 360 tcctca 366

<210> 138 <211> 122 <212> PRT <213> Artificial Sequence

<220>

<223> synthetic <400> 138

Gin Val Gin Leu Val Glu Ser Gly Gly Gly Val Ala Gin Pro Gly Arg 1 5 10 15

Ser Leu Arg Leu Ser Cys Ala Ala Ser Gly lie Thr Phe Asn Asn Tyr 20 25 30

Gly Met His Trp Val Arg Gin Ala Pro Gly Lys Gly Leu Glu Trp Val 35 40 45

Ala Phe lie Ser Asp Glu Gly Arg Asn Lys His Tyr Gly Asp Ser Val 50 55 60

Lys Gly Arg Phe Thr lie Asp Arg Asp Asn Ser Lys Asn Thr Leu Tyr 65 70 75 80

Leu Gin Met Asn Ser Leu Arg Ala Glu Asp Thr Ala Val Tyr Tyr Cys 85 90 95

Ala Lys Leu Gly Asp Asn Arg Asn His His Tyr Gly Leu Asp Val Trp 100 105 110

Gly Gin Gly Thr Thr Val Thr Val Ser Ser 115 120

<210> 139 <211> 24 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 139 ggaatcacct tcaataacta tggc 24

<210> 140 <211> 8 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic <400> 140

Gly lie Thr Phe Asn Asn Tyr Gly 1 5

<210> 141 <211> 24 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 141 atttcagatg aaggaagaaa taaa 24

<210> 142 <211> 8 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic <400> 142 lie Ser Asp Glu Gly Arg Asn Lys 1 5

<210> 143 <211> 45 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 143 gcgaaattgg gggataatag gaatcaccac tacggtttgg acgtc 45

<210> 144 <211> 15 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic <400> 144

Ala Lys Leu Gly Asp Asn Arg Asn His His Tyr Gly Leu Asp Val 1 5 10 15

<210> 145 <211> 321 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic

<400> 145 gacatccagt tgacccagtc tccatccttc ctgtctgcat ctgtaggaga cagagtcacc 60 atcacttgct gggccagtca gggcattacc aattatttag cctggtatca gcaaaaacca 120 gggaaagccc ctagcctcct gatctatgct gcatccactt tgcaaagtgg ggtcccatca 180 aggttcagcg gcagtggaca tgggacagaa ttcactctca cagtcagcag cctgcagcct 240 gaagattttg taacttatta ctgtcaacag tttaatagtt acccgatcac cttcggccaa 300 gggacacgac tggagattaa a 321

<210> 146 <211> 107 <212> PRT <213> Artificial Sequence

<220>

<223> synthetic <400> 146

Asp lie Gin Leu Thr Gin Ser Pro Ser Phe Leu Ser Ala Ser Val Gly 1 5 10 15

Asp Arg Val Thr lie Thr Cys Trp Ala Ser Gin Gly lie Thr Asn Tyr 20 25 30

Leu Ala Trp Tyr Gin Gin Lys Pro Gly Lys Ala Pro Ser Leu Leu lie 35 40 45

Tyr Ala Ala Ser Thr Leu Gin Ser Gly Val Pro Ser Arg Phe Ser Gly 50 55 60

Ser Gly His Gly Thr Glu Phe Thr Leu Thr Val Ser Ser Leu Gin Pro 65 70 75 80

Glu Asp Phe Val Thr Tyr Tyr Cys Gin Gin Phe Asn Ser Tyr Pro lie 85 90 95

Thr Phe Gly Gin Gly Thr Arg Leu Glu lie Lys 100 105

<210> 147 <211> 18 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 147 cagggcatta ccaattat 18

<210> 148 <211> 6 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic <400> 148

Gin Gly lie Thr Asn Tyr 1 5

<210> 149 <211> 9 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 149 gctgcatcc 9

<210> 150 <211> 3 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic

<400> 150 Ala Ala Ser 1

<210> 151 <211> 27 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 151 caacagttta atagttaccc gatcacc 27

<210> 152 <211> 9 <212> PRT

<213> Artificial Sequence <220> <223> synthetic

<400> 152

Gin Gin Phe Asn Ser Tyr Pro lie Thr 1 5

<210> 153 <211> 366 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic

<400> 153 caggtccagc tggtgcagtc tggggctgag gtgaagaagc ctgggtcctc ggtgaaggtc 60 tcctgtaagg cttctggagg caccttcagc aactatgata tcagctgggt gcgacaggcc 120 cctggacaag gacttgagtg gatgggaggg atcatcccca tcattggtac agcaaactac 180 gcacagaagt tccagggcag agtcacgatt acaacggacg aatccacgag cacagcctac 240 atggagctga gcagcctgag atctgaggac acggccgtgt attactgtgc gagagatccg 300 ggtatagcag tggctggttc gagctttgac tactggggcc agggaaccct ggtcaccgtc 360 tcctca 366

<210> 154 <211> 122 <212> PRT

<213> Artificial Sequence

<220>

<223> synthetic <400> 154

Gin Val Gin Leu Val Gin Ser Gly Ala Glu Val Lys Lys Pro Gly Ser 1 5 10 15

Ser Val Lys Val Ser Cys Lys Ala Ser Gly Gly Thr Phe Ser Asn Tyr 20 25 30

Asp lie Ser Trp Val Arg Gin Ala Pro Gly Gin Gly Leu Glu Trp Met 35 40 45

Gly Gly lie lie Pro lie lie Gly Thr Ala Asn Tyr Ala Gin Lys Phe 50 55 60

Gin Gly Arg Val Thr lie Thr Thr Asp Glu Ser Thr Ser Thr Ala Tyr 65 70 75 80

Met Glu Leu Ser Ser Leu Arg Ser Glu Asp Thr Ala Val Tyr Tyr Cys 85 90 95

Ala Arg Asp Pro Gly lie Ala Val Ala Gly Ser Ser Phe Asp Tyr Trp 100 105 110

Gly Gin Gly Thr Leu Val Thr Val Ser Ser 115 120

<210> 155 <211> 24 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 155 ggaggcacct tcagcaacta tgat 24 <210> 156 <211> 8 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic <400> 156

Gly Gly Thr Phe Ser Asn Tyr Asp 1 5

<210> 157 <211> 24 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 157 atcatcccca tcattggtac agca 24

<210> 158 <211> 8 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic <400> 158 lie lie Pro lie lie Gly Thr Ala 1 5

<210> 159 <211> 45 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 159 gcgagagatc cgggtatagc agtggctggt tcgagctttg actac 45

<210> 160 <211> 15 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic <400> 160

Ala Arg Asp Pro Gly lie Ala Val Ala Gly Ser Ser Phe Asp Tyr 1 5 10 15 <210> 161 <211> 321 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic

<400> 161 gccatccaga tgacccagtc tccatcctcc ctgtctgcat ctgttggaga cagagtcacc 60 atcacttgcc gggcaagtca gggcattaga catgatttag gctggtatca gcagaaacca 120 gggaaagccc ctaagctcct gatctatgct gcatccaatt tacaaagtgg ggtcccatca 180 aggttcagcg gcagtggatc tggcacagat ttcactctca ccatcagcag cctgcagcct 240 gaagattttg caacttatta ctgtctacaa gattacaatt accctcggac gttcggccaa 300 gggaccaagg tggaaatcaa a 321

<210> 162 <211> 107 <212> PRT <213> Artificial Sequence

<220>

<223> synthetic <400> 162

Ala lie Gin Met Thr Gin Ser Pro Ser Ser Leu Ser Ala Ser Val Gly 1 5 10 15

Asp Arg Val Thr lie Thr Cys Arg Ala Ser Gin Gly lie Arg His Asp 20 25 30

Leu Gly Trp Tyr Gin Gin Lys Pro Gly Lys Ala Pro Lys Leu Leu lie 35 40 45

Tyr Ala Ala Ser Asn Leu Gin Ser Gly Val Pro Ser Arg Phe Ser Gly 50 55 60

Ser Gly Ser Gly Thr Asp Phe Thr Leu Thr lie Ser Ser Leu Gin Pro 65 70 75 80

Glu Asp Phe Ala Thr Tyr Tyr Cys Leu Gin Asp Tyr Asn Tyr Pro Arg 85 90 95

Thr Phe Gly Gin Gly Thr Lys Val Glu lie Lys 100 105

<210> 163 <211> 18 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 163 cagggcatta gacatgat 18

<210> 164 <211> 6 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic <400> 164

Gin Gly lie Arg His Asp 1 5

<210> 165 <211> 9 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 165 gctgcatcc 9

<210> 166 <211> 3 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic

<400> 166 Ala Ala Ser 1

<210> 167 <211> 27 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 167 ctacaagatt acaattaccc tcggacg 27

<210> 168 <211> 9 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic <400> 168

Leu Gin Asp Tyr Asn Tyr Pro Arg Thr 1 5

<210> 169 <211> 348 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 169 caggtgcagc tggtgcagtc tggggctgag gtgaggacgc ctgggtcctc ggtgaaggtc 60 tcctgcaagg cttctggagg caccttcagc agctatgctg tcacctgggt gcgacaggcc 120 cctggacaag ggcttgagtg gatgggaggg ctcatccctt tctttggtcc agcaaactac 180 gcacagagat tccagggcag agtctcgatt accacggacg aatccacgaa catagcctac 240 ttggagctga gcagcctgag atccgaggac tcggccgttt attactgtgc gagagggcgg 300 aacttcggtg actactgggg ccagggaacc ctggtcaccg tctcctca 348

<210> 170 <211> 116 <212> PRT

<213> Artificial Sequence

<220>

<223> synthetic <400> 170

Gin Val Gin Leu Val Gin Ser Gly Ala Glu Val Arg Thr Pro Gly Ser 1 5 10 15

Ser Val Lys Val Ser Cys Lys Ala Ser Gly Gly Thr Phe Ser Ser Tyr 20 25 30

Ala Val Thr Trp Val Arg Gin Ala Pro Gly Gin Gly Leu Glu Trp Met 35 40 45

Gly Gly Leu lie Pro Phe Phe Gly Pro Ala Asn Tyr Ala Gin Arg Phe 50 55 60

Gin Gly Arg Val Ser lie Thr Thr Asp Glu Ser Thr Asn lie Ala Tyr 65 70 75 80

Leu Glu Leu Ser Ser Leu Arg Ser Glu Asp Ser Ala Val Tyr Tyr Cys 85 90 95

Ala Arg Gly Arg Asn Phe Gly Asp Tyr Trp Gly Gin Gly Thr Leu Val 100 105 110

Thr Val Ser Ser 115

<210> 171 <211> 24 <212> DNA

<213> Artificial Sequence

<220>

<223> synthetic

<400> 171 ggaggcacct tcagcagcta tgct 24

<210> 172 <211> 8 <212> PRT

<213> Artificial Sequence

<220>

<223> synthetic

<400> 172

Gly Gly Thr Phe Ser Ser Tyr Ala 1 5

<210> 173 <211> 24 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 173 ctcatccctt tctttggtcc agca 24

<210> 174 <211> 8 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic <400> 174

Leu lie Pro Phe Phe Gly Pro Ala 1 5

<210> 175 <211> 27 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 175 gcgagagggc ggaacttcgg tgactac 27

<210> 176 <211> 9 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic <400> 176

Ala Arg Gly Arg Asn Phe Gly Asp Tyr 1 5

<210> 177 <211> 321 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic

<400> 177 gacatccaga tgacccagtc tccatcctca ctgtctgcat ctataggaga cagagtcacc 60 atcacttgtc gggcgagtca ggacattagc aattatttag cctggtttca gcagaaacca 120 gggaaagtcc ctaagtccct gatctatgct gcatccaatt tgcaaagtgg ggtcccatca 180 aagttcagcg gcagtggatc tgggacagat ttcactctca ccatcagtac cctgcagcct 240 gaagattttg caacttatta ctgccaacag tatagtagtt atccattcac tttcggccct 300 gggaccaaag tggatatcaa a 321 <210> 178 <211> 107 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic <400> 178

Asp lie Gin Met Thr Gin Ser Pro Ser Ser Leu Ser Ala Ser lie Gly 1 5 10 15

Asp Arg Val Thr lie Thr Cys Arg Ala Ser Gin Asp lie Ser Asn Tyr 20 25 30

Leu Ala Trp Phe Gin Gin Lys Pro Gly Lys Val Pro Lys Ser Leu lie 35 40 45

Tyr Ala Ala Ser Asn Leu Gin Ser Gly Val Pro Ser Lys Phe Ser Gly 50 55 60

Ser Gly Ser Gly Thr Asp Phe Thr Leu Thr lie Ser Thr Leu Gin Pro 65 70 75 80

Glu Asp Phe Ala Thr Tyr Tyr Cys Gin Gin Tyr Ser Ser Tyr Pro Phe 85 90 95

Thr Phe Gly Pro Gly Thr Lys Val Asp lie Lys 100 105

<210> 179 <211> 18 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 179 caggacatta gcaattat 18

<210> 180 <211> 6 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic <400> 180

Gin Asp lie Ser Asn Tyr 1 5

<210> 181 <211> 9 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 181 gctgcatcc 9 <210> 182 <211> 3 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic

<400> 182 Ala Ala Ser 1

<210> 183 <211> 27 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 183 caacagtata gtagttatcc attcact 27

<210> 184 <211> 9 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic <400> 184

Gin Gin Tyr Ser Ser Tyr Pro Phe Thr 1 5

<210> 185 <211> 351 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic

<400> 185 caggtgcagc tggtgcagtc tggggctgag gtgaagaagc ctgggtcctc ggtgaaggtc 60 tcctgcaagg cttctggagg caccttcagc agctatgcta tcagctgggt gcgacaggcc 120 cctggacaag ggcttgagtg gatgggaggg atcatcccta tctttggtac aacaaactac 180 gcacagaagt tccagggcag agtcacgatt accacggacg aatccacgag cacagcctac 240 atggagctga gcagcctgag atctgaggac acggccgtgt attactgtgc gagaaggcac 300 ggtatatcct ttgactactg gggccaggga accctggtca ccgtctcctc a 351

<210> 186 <211> 117 <212> PRT <213> Artificial Sequence

<220>

<223> synthetic <400> 186 Gin Val Gin Leu Val Gin Ser Gly Ala Glu Val Lys Lys Pro Gly Ser 1 5 10 15

Ser Val Lys Val Ser Cys Lys Ala Ser Gly Gly Thr Phe Ser Ser Tyr 20 25 30

Ala lie Ser Trp Val Arg Gin Ala Pro Gly Gin Gly Leu Glu Trp Met 35 40 45

Gly Gly lie lie Pro lie Phe Gly Thr Thr Asn Tyr Ala Gin Lys Phe 50 55 60

Gin Gly Arg Val Thr lie Thr Thr Asp Glu Ser Thr Ser Thr Ala Tyr 65 70 75 80

Met Glu Leu Ser Ser Leu Arg Ser Glu Asp Thr Ala Val Tyr Tyr Cys 85 90 95

Ala Arg Arg His Gly lie Ser Phe Asp Tyr Trp Gly Gin Gly Thr Leu 100 105 110

Val Thr Val Ser Ser 115

<210> 187 <211> 24 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 187 ggaggcacct tcagcagcta tgct 24

<210> 188 <211> 8 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic <400> 188

Gly Gly Thr Phe Ser Ser Tyr Ala 1 5

<210> 189 <211> 24 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 189 atcatcccta tctttggtac aaca 24

<210> 190 <211> 8 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic <400> 190 lie lie Pro lie Phe Gly Thr Thr 1 5

<210> 191 <211> 30 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 191 gcgagaaggc acggtatatc ctttgactac 30

<210> 192 <211> 10 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic <400> 192

Ala Arg Arg His Gly lie Ser Phe Asp Tyr 1 5 10

<210> 193 <211> 321 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic

<400> 193 gacatccaga tgacccagtc tccatcctcc ctgtctgcat ctgtaggaga cagagtcacc 60 gtcacttgcc gggcaagtca gggcattaga aatgatttgg cctggtatca gcagaaacca 120 gggaaagccc ctaagcgcct gatctatgct gcatccagtt tgcaaagtgg ggtcccatca 180 aggttcagcg gcagtggatc tgggacagaa ttcactctca caatcagcag cctgcagcct 240 gaagattttg caacttatta ctgtctacag cataatagtt atccgtggac gttcggccta 300 gggaccaagg tggaaatcaa a 321

<210> 194 <211> 107 <212> PRT <213> Artificial Sequence

<220>

<223> synthetic <400> 194

Asp lie Gin Met Thr Gin Ser Pro Ser Ser Leu Ser Ala Ser Val Gly 1 5 10 15

Asp Arg Val Thr Val Thr Cys Arg Ala Ser Gin Gly lie Arg Asn Asp 20 25 30

Leu Ala Trp Tyr Gin Gin Lys Pro Gly Lys Ala Pro Lys Arg Leu lie 35 40 45 Tyr Ala Ala Ser Ser Leu Gin Ser Gly Val Pro Ser Arg Phe Ser Gly 50 55 60

Ser Gly Ser Gly Thr Glu Phe Thr Leu Thr lie Ser Ser Leu Gin Pro 65 70 75 80

Glu Asp Phe Ala Thr Tyr Tyr Cys Leu Gin His Asn Ser Tyr Pro Trp 85 90 95

Thr Phe Gly Leu Gly Thr Lys Val Glu lie Lys 100 105

<210> 195 <211> 18 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 195 cagggcatta gaaatgat 18

<210> 196 <211> 6 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic

<400> 196

Gin Gly lie Arg Asn Asp 1 5

<210> 197 <211> 9 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 197 gctgcatcc 9

<210> 198 <211> 3 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic

<400> 198 Ala Ala Ser 1

<210> 199 <211> 27 <212> DNA <213> Artificial Sequence

<220>

<223> synthetic <400> 199 ctacagcata atagttatcc gtggacg 27

<210> 200 <211> 9 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic <400> 200

Leu Gin His Asn Ser Tyr Pro Trp Thr 1 5

<210> 201 <211> 366 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic

<400> 201 caggtgcagc tggtgcagtc tggggctgag gtgaagaagc ctgggtcctc ggtgaaggtc 60 tcctgcaagg cttctggagg caccttcagc agctatacta tcagctgggt tcgacaggcc 120 cctggacaag ggcttgagtg gatgggaggg atcagcccta tctttggtac agcaaactac 180 gcacagaagt tccagggcag agtcacgatt accacggacg aatccacgag cacagcctac 240 atggagctga gcagcctgag atctgaggac acggccgtgt attactgtgc gagagagtcc 300 ccgtataact ggaaccaaaa atacttccag tattggggcc agggcaccct ggtcaccgtc 360 tcctca 366

<210> 202 <211> 122 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic

<400> 202 Gin Val Gin Leu Val Gin Ser Gly Ala Glu Val Lys Lys Pro Gly Ser 1 5 10 15

Ser Val Lys Val Ser Cys Lys Ala Ser Gly Gly Thr Phe Ser Ser Tyr 20 25 30

Thr lie Ser Trp Val Arg Gin Ala Pro Gly Gin Gly Leu Glu Trp Met 35 40 45

Gly Gly lie Ser Pro lie Phe Gly Thr Ala Asn Tyr Ala Gin Lys Phe 50 55 60

Gin Gly Arg Val Thr lie Thr Thr Asp Glu Ser Thr Ser Thr Ala Tyr 65 70 75 80

Met Glu Leu Ser Ser Leu Arg Ser Glu Asp Thr Ala Val Tyr Tyr Cys 85 90 95

Ala Arg Glu Ser Pro Tyr Asn Trp Asn Gin Lys Tyr Phe Gin Tyr Trp 100 105 110 Gly Gin Gly Thr Leu Val Thr Val Ser Ser 115 120

<210> 203 <211> 24 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 203 ggaggcacct tcagcagcta tact 24

<210> 204 <211> 8 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic <400> 204

Gly Gly Thr Phe Ser Ser Tyr Thr 1 5

<210> 205 <211> 24 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 205 atcagcccta tctttggtac agca 24

<210> 206 <211> 8 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic <400> 206 lie Ser Pro lie Phe Gly Thr Ala 1 5

<210> 207 <211> 45 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 207 gcgagagagt ccccgtataa ctggaaccaa aaatacttcc agtat 45

<210> 208 <211> 15 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic <400> 208

Ala Arg Glu Ser Pro Tyr Asn Trp Asn Gin Lys Tyr Phe Gin Tyr 1 5 10 15

<210> 209 <211> 321 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic

<400> 209 gacatccaga tgacccagtc tccttccacc ctgtctgcat ctgtaggaga cagagtcacc 60 atcacttgcc gggccagtca gagtattagt agctggttgg cctggtatca gcagaaacca 120 gggaaagccc ctaagctcct gatctataag gcgtctagtt tagaaagtgg ggtcccatca 180 aggttcagcg gcagtggatc tgggacagaa ttcactctca ccatcagcag cctgcagcct 240 gatgattttg caacttatta ctgccaacag tataatagtt attcgtacac ttttggccag 300 gggaccaagc tggagatcaa a 321

<210> 210 <211> 107 <212> PRT <213> Artificial Sequence

<220>

<223> synthetic

<400> 210 Asp lie Gin Met Thr Gin Ser Pro Ser Thr Leu Ser Ala Ser Val Gly 1 5 10 15

Asp Arg Val Thr lie Thr Cys Arg Ala Ser Gin Ser lie Ser Ser Trp 20 25 30

Leu Ala Trp Tyr Gin Gin Lys Pro Gly Lys Ala Pro Lys Leu Leu lie 35 40 45

Tyr Lys Ala Ser Ser Leu Glu Ser Gly Val Pro Ser Arg Phe Ser Gly 50 55 60

Ser Gly Ser Gly Thr Glu Phe Thr Leu Thr lie Ser Ser Leu Gin Pro 65 70 75 80

Asp Asp Phe Ala Thr Tyr Tyr Cys Gin Gin Tyr Asn Ser Tyr Ser Tyr 85 90 95

Thr Phe Gly Gin Gly Thr Lys Leu Glu lie Lys 100 105

<210> 211 <211> 18 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 211 cagagtatta gtagctgg 18

<210> 212 <211> 6 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic <400> 212

Gin Ser lie Ser Ser Trp 1 5

<210> 213 <211> 9 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 213 aaggcgtct 9

<210> 214 <211> 3 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic

<400> 214 Lys Ala Ser 1

<210> 215 <211> 27 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 215 caacagtata atagttattc gtacact 27

<210> 216 <211> 9 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic <400> 216

Gin Gin Tyr Asn Ser Tyr Ser Tyr Thr 1 5

<210> 217 <211> 363 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic

<400> 217 caggtccagc tggtgcagtc tgggtctgag gtgaagaage ctgggtcctc ggtgaaggtc 60 tcctgcaagg cgtctggagg gaccctcagc ctctatgctg tcagctgggt gcgacaggcc 120 cctggacagg ggcttgagtg gatggggggg atcatcccta tctttggtac aacaaaatac 180 gcacaggagt tccagggcag agtcacgttt tccacggacg agtccacgag cacagcctac 240 atggagctga acagcctgcg atetgaggae acggccgtgt attactgtgc gagacaatgg 300 actatgacta gaaccttgga ttttgacatc tggggccagg gaaccctggt caccgtctcc 360 tea 363

<210> 218 <211> 121 <212> PRT

<213> Artificial Sequence

<220>

<223> synthetic

<400> 218 Gin Val Gin Leu Val Gin Ser Gly Ser Glu Val Lys Lys Pro Gly Ser 1 5 10 15

Ser Val Lys Val Ser Cys Lys Ala Ser Gly Gly Thr Leu Ser Leu Tyr 20 25 30

Ala Val Ser Trp Val Arg Gin Ala Pro Gly Gin Gly Leu Glu Trp Met 35 40 45

Gly Gly lie lie Pro lie Phe Gly Thr Thr Lys Tyr Ala Gin Glu Phe 50 55 60

Gin Gly Arg Val Thr Phe Ser Thr Asp Glu Ser Thr Ser Thr Ala Tyr 65 70 75 80

Met Glu Leu Asn Ser Leu Arg Ser Glu Asp Thr Ala Val Tyr Tyr Cys 85 90 95 Ala Arg Gin Trp Thr Met Thr Arg Thr Leu Asp Phe Asp lie Trp Gly

100 105 110

Gin Gly Thr Leu Val Thr Val Ser Ser 115 120

<210> 219 <211> 24 <212> DNA

<213> Artificial Sequence

<220>

<223> synthetic

<400> 219 ggagggaccc tcagcctcta tget 24 <210> 220 <211> 8 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic <400> 220

Gly Gly Thr Leu Ser Leu Tyr Ala 1 5

<210> 221 <211> 24 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 221 atcatcccta tctttggtac aaca 24

<210> 222 <211> 8 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic <400> 222 lie lie Pro lie Phe Gly Thr Thr 1 5

<210> 223 <211> 42 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 223 gcgagacaat ggactatgac tagaaccttg gattttgaca tc 42

<210> 224 <211> 14 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic <400> 224

Ala Arg Gin Trp Thr Met Thr Arg Thr Leu Asp Phe Asp lie 1 5 10 <210> 225 <211> 321 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic

<400> 225 gacatccaga tgacccagtc tccatcctcc ctgtctgcat ctgtaggaga cagagtcacc 60 atcacttgcc ggacaagtca gggcattaga aatgatttag gctggtatca gcagaaaccg 120 gggaaagccc ctaagcgcct gatctatggt gcatccagtt tgcaaagtgg ggtcccatca 180 agattcagcg gcagtggatc tgggacagaa ttcactctca caatcagcag cctgcagcct 240 gaagattttg caagttatta ctgtctacaa cataataatt atccgtggac gttcggccaa 300 gggaccaagg tggaaatcaa a 321

<210> 226 <211> 107 <212> PRT <213> Artificial Sequence

<220>

<223> synthetic

<400> 226 Asp lie Gin Met Thr Gin Ser Pro Ser Ser Leu Ser Ala Ser Val Gly 1 5 10 15

Asp Arg Val Thr lie Thr Cys Arg Thr Ser Gin Gly lie Arg Asn Asp 20 25 30

Leu Gly Trp Tyr Gin Gin Lys Pro Gly Lys Ala Pro Lys Arg Leu lie 35 40 45

Tyr Gly Ala Ser Ser Leu Gin Ser Gly Val Pro Ser Arg Phe Ser Gly 50 55 60

Ser Gly Ser Gly Thr Glu Phe Thr Leu Thr lie Ser Ser Leu Gin Pro 65 70 75 80

Glu Asp Phe Ala Ser Tyr Tyr Cys Leu Gin His Asn Asn Tyr Pro Trp 85 90 95

Thr Phe Gly Gin Gly Thr Lys Val Glu lie Lys 100 105

<210> 227 <211> 18 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 227 cagggcatta gaaatgat 18

<210> 228 <211> 6 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic <400> 228 Gin Gly lie Arg Asn Asp 1 5

<210> 229 <211> 9 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 229 ggtgcatcc 9

<210> 230 <211> 3 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic

<400> 230 Gly Ala Ser 1

<210> 231 <211> 27 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 231 ctacaacata ataattatcc gtggacg 27

<210> 232 <211> 9 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic <400> 232

Leu Gin His Asn Asn Tyr Pro Trp Thr 1 5

<210> 233 <211> 369 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 233 caggtgcagc tggtggagtc tgggggaggc gtggtccagc ctgggaggtc cctgagactc 60 tcctgtgtag cctccggatt caccttcagt gtctatggca tgaactgggt ccgccaggct 120 ccaggcaagg gtctggactg ggtggcagtc atatcaaatg atggaagtta taaatactat 180 gcggactccg tgaagggccg attcaccatc tccagagaca attccaagaa cacggtgtat 240 ctgcaaatga acagcctgag aggtgaggac acggctattt attactgtgc gaaaaggaat 300 gaatgggagc tagaggaata ctacggtatg gacgtctggg gccaagggac cacggtcacc 360 gtctcctca 369

<210> 234 <211> 123 <212> PRT

<213> Artificial Sequence

<220>

<223> synthetic <400> 234

Gin Val Gin Leu Val Glu Ser Gly Gly Gly Val Val Gin Pro Gly Arg 1 5 10 15

Ser Leu Arg Leu Ser Cys Val Ala Ser Gly Phe Thr Phe Ser Val Tyr 20 25 30

Gly Met Asn Trp Val Arg Gin Ala Pro Gly Lys Gly Leu Asp Trp Val 35 40 45

Ala Val lie Ser Asn Asp Gly Ser Tyr Lys Tyr Tyr Ala Asp Ser Val 50 55 60

Lys Gly Arg Phe Thr lie Ser Arg Asp Asn Ser Lys Asn Thr Val Tyr 65 70 75 80

Leu Gin Met Asn Ser Leu Arg Gly Glu Asp Thr Ala lie Tyr Tyr Cys 85 90 95

Ala Lys Arg Asn Glu Trp Glu Leu Glu Glu Tyr Tyr Gly Met Asp Val 100 105 110

Trp Gly Gin Gly Thr Thr Val Thr Val Ser Ser 115 120

<210> 235 <211> 24 <212> DNA

<213> Artificial Sequence

<220>

<223> synthetic

<400> 235 ggattcacct tcagtgtcta tggc 24

<210> 236 <211> 8 <212> PRT

<213> Artificial Sequence

<220>

<223> synthetic

<400> 236

Gly Phe Thr Phe Ser Val Tyr Gly 1 5

<210> 237 <211> 24 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 237 atatcaaatg atggaagtta taaa 24

<210> 238 <211> 8 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic

<400> 238 lie Ser Asn Asp Gly Ser Tyr Lys 1 5

<210> 239 <211> 48 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 239 gcgaaaagga atgaatggga gctagaggaa tactacggta tggacgtc 48

<210> 240 <211> 16 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic <400> 240

Ala Lys Arg Asn Glu Trp Glu Leu Glu Glu Tyr Tyr Gly Met Asp Val 1 5 10 15

<210> 241 <211> 363 <212> DNA

<213> Artificial Sequence

<220>

<223> synthetic

<400> 241 gaggtgcagc tggtggagtc tgggggagga ttggtacagt ctggagggtc cctgagactc 60 tcctgtgtag cctctggatt caccttcaat aattatgaga tgaattgggt ccgccaggct 120 ccagggaagg ggctggagtg gatttcatac attgatatta atggtggaag taccatctac 180 tatgcagact ctgtgaaggg ccgattcacc atctccagag acaatgccaa gaagtcactg 240 tatctgcaaa tgaacagcct gagagccgag gacacggcta tttactactg tgcgagcgcc 300 tttggttcgg gtggttttct ttttgactat tggggccagg gaaccctggt cactgtctcc 360 tea 363

<210> 242 <211> 121 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic

<400> 242 Glu Val Gin Leu Val Glu Ser Gly Gly Gly Leu Val Gin Ser Gly Gly 1 5 10 15

Ser Leu Arg Leu Ser Cys Val Ala Ser Gly Phe Thr Phe Asn Asn Tyr 20 25 30

Glu Met Asn Trp Val Arg Gin Ala Pro Gly Lys Gly Leu Glu Trp lie 35 40 45

Ser Tyr lie Asp lie Asn Gly Gly Ser Thr lie Tyr Tyr Ala Asp Ser 50 55 60

Val Lys Gly Arg Phe Thr lie Ser Arg Asp Asn Ala Lys Lys Ser Leu 65 70 75 80

Tyr Leu Gin Met Asn Ser Leu Arg Ala Glu Asp Thr Ala lie Tyr Tyr 85 90 95

Cys Ala Ser Ala Phe Gly Ser Gly Gly Phe Leu Phe Asp Tyr Trp Gly 100 105 110

Gin Gly Thr Leu Val Thr Val Ser Ser 115 120

<210> 243 <211> 24 <212> DNA <213> Artificial Sequence

<220>

<223> synthetic <400> 243 ggattcacct tcaataatta tgag 24

<210> 244 <211> 8 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic <400> 244

Gly Phe Thr Phe Asn Asn Tyr Glu 1 5

<210> 245 <211> 27 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 245 attgatatta atggtggaag taccatc 27

<210> 246 <211> 9 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic <400> 246 lie Asp lie Asn Gly Gly Ser Thr lie 1 5

<210> 247 <211> 39 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 247 gcgagcgcct ttggttcggg tggttttctt tttgactat 39

<210> 248 <211> 13 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic <400> 248

Ala Ser Ala Phe Gly Ser Gly Gly Phe Leu Phe Asp Tyr 1 5 10

<210> 249 <211> 366 <212> DNA

<213> Artificial Sequence

<220>

<223> synthetic

<400> 249 caggtgcagc tggtggagtc tgggggaggc gtggtccagc ctgggatgtc cctgagactc 60 tcctgtatag cgtctggatt caccttcagt gactatgtca tacactgggt ccgccaggct 120 ccaggcaagg ggctggagtg ggtggcaatt atatggtatg atggaagtaa taaatactat 180 gcagactccg tgaagggccg attcaccatc tccagagaca attccaagaa cacgctgtat 240 cttgaaatga atagactgag agtcgaggac acggctgtgt tttactgtgc gaggggtctg 300 gggatcgaag attacaatta cggtatggac gtctggggcc aagggaccac ggtcaccgtc 360 tcctca 366

<210> 250 <211> 122 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic

<400> 250 Gin Val Gin Leu Val Glu Ser Gly Gly Gly Val Val Gin Pro Gly Met 1 5 10 15

Ser Leu Arg Leu Ser Cys lie Ala Ser Gly Phe Thr Phe Ser Asp Tyr 20 25 30

Val lie His Trp Val Arg Gin Ala Pro Gly Lys Gly Leu Glu Trp Val 35 40 45

Ala lie lie Trp Tyr Asp Gly Ser Asn Lys Tyr Tyr Ala Asp Ser Val 50 55 60

Lys Gly Arg Phe Thr lie Ser Arg Asp Asn Ser Lys Asn Thr Leu Tyr 65 70 75 80

Leu Glu Met Asn Arg Leu Arg Val Glu Asp Thr Ala Val Phe Tyr Cys 85 90 95

Ala Arg Gly Leu Gly lie Glu Asp Tyr Asn Tyr Gly Met Asp Val Trp 100 105 110

Gly Gin Gly Thr Thr Val Thr Val Ser Ser 115 120

<210> 251 <211> 24 <212> DNA <213> Artificial Sequence

<220>

<223> synthetic <400> 251 ggattcacct tcagtgacta tgtc 24

<210> 252 <211> 8 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic <400> 252

Gly Phe Thr Phe Ser Asp Tyr Val 1 5

<210> 253 <211> 24 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 253 atatggtatg atggaagtaa taaa 24

<210> 254 <211> 8 <212> PRT <213> Artificial Sequence

<220>

<223> synthetic <400> 254 lie Trp Tyr Asp Gly Ser Asn Lys 1 5

<210> 255 <211> 45 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 255 gcgaggggtc tggggatcga agattacaat tacggtatgg acgtc 45

<210> 256 <211> 15 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic <400> 256

Ala Arg Gly Leu Gly lie Glu Asp Tyr Asn Tyr Gly Met Asp Val 1 5 10 15

<210> 257 <211> 357 <212> DNA

<213> Artificial Sequence

<220>

<223> synthetic

<400> 257 gaggtgcagc tggtgcagtc tggagcagag gtgaaaaagc ccggggagtc tctgaagatc 60 tcctgtaagg gttctggata cagctttacc agctactgga tcggctgggt gcgccagatg 120 cccgggaaag gcctggagtg gatggggatc atctatcctg gtgactctga taccagatac 180 agcccgtcct tccaaggcca ggtcaccatc tcagccgaca agtccatcag caccgcctac 240 ctgcagtgga gcagcctgaa ggcctcggac accgccatgt attactgtgc gagacactgg 300 accgacgagg atgcttttga tatctggggc caagggacaa tggtcaccgt ctcttca 357

<210> 258 <211> 119 <212> PRT

<213> Artificial Sequence

<220>

<223> synthetic

<400> 258

Glu Val Gin Leu Val Gin Ser Gly Ala Glu Val Lys Lys Pro Gly Glu 1 5 10 15 Ser Leu Lys lie Ser Cys Lys Gly Ser Gly Tyr Ser Phe Thr Ser Tyr 20 25 30

Trp lie Gly Trp Val Arg Gin Met Pro Gly Lys Gly Leu Glu Trp Met 35 40 45

Gly lie lie Tyr Pro Gly Asp Ser Asp Thr Arg Tyr Ser Pro Ser Phe 50 55 60

Gin Gly Gin Val Thr lie Ser Ala Asp Lys Ser lie Ser Thr Ala Tyr 65 70 75 80

Leu Gin Trp Ser Ser Leu Lys Ala Ser Asp Thr Ala Met Tyr Tyr Cys 85 90 95

Ala Arg His Trp Thr Asp Glu Asp Ala Phe Asp lie Trp Gly Gin Gly 100 105 110

Thr Met Val Thr Val Ser Ser 115

<210> 259 <211> 24 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 259 ggatacagct ttaccagcta ctgg 24

<210> 260 <211> 8 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic

<400> 260

Gly Tyr Ser Phe Thr Ser Tyr Trp 1 5

<210> 261 <211> 24 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 261 atctatcctg gtgactctga tacc 24

<210> 262 <211> 8 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic <400> 262 lie Tyr Pro Gly Asp Ser Asp Thr 1 5

<210> 263 <211> 36 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 263 gcgagacact ggaccgacga ggatgctttt gatatc 36

<210> 264 <211> 12 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic <400> 264

Ala Arg His Trp Thr Asp Glu Asp Ala Phe Asp lie 1 5 10

<210> 265 <211> 369 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic

<400> 265 caggtccagc tggtgcagtc tgggactgag gtgaagaagc ctgggtcttc ggtgaaggtc 60 tcctgcaagg cttctggagg caccttcggc agttatccta tcagctgggt gcgacaggcc 120 cctggacaag ggcttgagtg gatgggaggg atcatcccta tctttggtac agcaaactcc 180 gcacagaagt tccagggcag agtcacgatt accacggacg aacccacgag cacagccttc 240 atggagctga gcagcctgag atctgaggac acggccgtgt attactgtgc gagaccgtat 300 tacgatattt tgactgacta ctacggtatg gacgtctggg gtcaagggac cacggtcacc 360 gtctcctca 369

<210> 266 <211> 123 <212> PRT

<213> Artificial Sequence

<220>

<223> synthetic <400> 266

Gin Val Gin Leu Val Gin Ser Gly Thr Glu Val Lys Lys Pro Gly Ser 1 5 10 15

Ser Val Lys Val Ser Cys Lys Ala Ser Gly Gly Thr Phe Gly Ser Tyr 20 25 30

Pro lie Ser Trp Val Arg Gin Ala Pro Gly Gin Gly Leu Glu Trp Met 35 40 45

Gly Gly lie lie Pro lie Phe Gly Thr Ala Asn Ser Ala Gin Lys Phe 50 55 60 Gin Gly Arg Val Thr lie Thr Thr Asp Glu Pro Thr Ser Thr Ala Phe

65 70 75 80

Met Glu Leu Ser Ser Leu Arg Ser Glu Asp Thr Ala Val Tyr Tyr Cys

85 90 95

Ala Arg Pro Tyr Tyr Asp lie Leu Thr Asp Tyr Tyr Gly Met Asp Val 100 105 110

Trp Gly Gin Gly Thr Thr Val Thr Val Ser Ser 115 120

<210> 267 <211> 24 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 267 ggaggcacct tcggcagtta tcct 24

<210> 268 <211> 8 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic

<400> 268

Gly Gly Thr Phe Gly Ser Tyr Pro 1 5

<210> 269 <211> 24 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 269 atcatcccta tctttggtac agca 24

<210> 270 <211> 8 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic <400> 270 lie lie Pro lie Phe Gly Thr Ala 1 5

<210> 271 <211> 48 <212> DNA <213> Artificial Sequence

<220>

<223> synthetic <400> 271 gcgagaccgt attacgatat tttgactgac tactacggta tggacgtc 48

<210> 272 <211> 16 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic <400> 272

Ala Arg Pro Tyr Tyr Asp lie Leu Thr Asp Tyr Tyr Gly Met Asp Val 1 5 10 15

<210> 273 <211> 354 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic

<400> 273 caggtccagc tggtgcagtc tggggctgag gtgaagaagc ctgggtcctc ggtgaaggtc 60 tcctgcaagg cttctggagg caccttcagc agctatgcta tcagctgggt gcgacaggcc 120 cctggacaag ggcttgagtg gatgggaggg atcatcccta tctttggtac agcaaactac 180 gcacagaagt tccagggcag agtcacgatt accacggacg aatccacgag cacagcctac 240 atggagctga gcagtctgag atctgaggac acggccgtgt attactgtgc gacagaaggg 300 tacaactgga actatgacta ttggggccag ggaaccctgg tcaccgtctc ctca 354

<210> 274 <211> 118 <212> PRT

<213> Artificial Sequence

<220>

<223> synthetic <400> 274

Gin Val Gin Leu Val Gin Ser Gly Ala Glu Val Lys Lys Pro Gly Ser 1 5 10 15

Ser Val Lys Val Ser Cys Lys Ala Ser Gly Gly Thr Phe Ser Ser Tyr 20 25 30

Ala lie Ser Trp Val Arg Gin Ala Pro Gly Gin Gly Leu Glu Trp Met 35 40 45

Gly Gly lie lie Pro lie Phe Gly Thr Ala Asn Tyr Ala Gin Lys Phe 50 55 60

Gin Gly Arg Val Thr lie Thr Thr Asp Glu Ser Thr Ser Thr Ala Tyr 65 70 75 80

Met Glu Leu Ser Ser Leu Arg Ser Glu Asp Thr Ala Val Tyr Tyr Cys 85 90 95

Ala Thr Glu Gly Tyr Asn Trp Asn Tyr Asp Tyr Trp Gly Gin Gly Thr 100 105 110

Leu Val Thr Val Ser Ser 115

<210> 275 <211> 24 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 275 ggaggcacct tcagcagcta tgct 24

<210> 276 <211> 8 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic <400> 276

Gly Gly Thr Phe Ser Ser Tyr Ala 1 5

<210> 277 <211> 24 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 277 atcatcccta tctttggtac agca 24

<210> 278 <211> 8 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic <400> 278 lie lie Pro lie Phe Gly Thr Ala 1 5

<210> 279 <211> 33 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 279 gcgacagaag ggtacaactg gaactatgac tat 33 <210> 280 <211> 11 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic <400> 280

Ala Thr Glu Gly Tyr Asn Trp Asn Tyr Asp Tyr 1 5 10

<210> 281 <211> 363 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic

<400> 281 caggtccagc tggtgcagtc tggggctgag gtgaagaage ctgggtcctc ggtgaaggtc 60 tcctgcaagg cttctggagg caccttcagc agctatgtta tcacctgggt gcgacaggtc 120 cctggacaag ggcttgagtg gatgggaggg atcatcccta tctttggtac agcaaactac 180 gcacagaagt tccagggcag agteaegatt accacggacg aatccacgag cacagcctac 240 atggacctga gcagcctgag atetgaggae acggcctttt attattgtgc gagaaggagg 300 tctaactggg gatctcatgc ttttgatatc tggggccaag ggacaatggt caccgtctct 360 tea 363

<210> 282 <211> 121 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic <400> 282

Gin Val Gin Leu Val Gin Ser Gly Ala Glu Val Lys Lys Pro Gly Ser 1 5 10 15

Ser Val Lys Val Ser Cys Lys Ala Ser Gly Gly Thr Phe Ser Ser Tyr 20 25 30

Val lie Thr Trp Val Arg Gin Val Pro Gly Gin Gly Leu Glu Trp Met 35 40 45

Gly Gly lie lie Pro lie Phe Gly Thr Ala Asn Tyr Ala Gin Lys Phe 50 55 60

Gin Gly Arg Val Thr lie Thr Thr Asp Glu Ser Thr Ser Thr Ala Tyr 65 70 75 80

Met Asp Leu Ser Ser Leu Arg Ser Glu Asp Thr Ala Phe Tyr Tyr Cys 85 90 95

Ala Arg Arg Arg Ser Asn Trp Gly Ser His Ala Phe Asp lie Trp Gly 100 105 110

Gin Gly Thr Met Val Thr Val Ser Ser 115 120

<210> 283 <211> 24 <212> DNA <213> Artificial Sequence <220>

<223> synthetic <400> 283 ggaggcacct tcagcagcta tgtt 24

<210> 284 <211> 8 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic <400> 284

Gly Gly Thr Phe Ser Ser Tyr Val 1 5

<210> 285 <211> 24 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 285 atcatcccta tctttggtac agca 24

<210> 286 <211> 8 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic <400> 286 lie lie Pro lie Phe Gly Thr Ala 1 5

<210> 287 <211> 42 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 287 gcgagaagga ggtctaactg gggatctcat gcttttgata tc 42

<210> 288 <211> 14 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic <400> 288

Ala Arg Arg Arg Ser Asn Trp Gly Ser His Ala Phe Asp lie 1 5 10

<210> 289 <211> 387 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic

<400> 289 gaagtgcagc tggtggagtc tgggggaggc ttggtacagc ctggcaggtc cctgagactc 60 tcctgtgcag cctctggaat cacctttgat gattatgcca tgtactgggt ccggcaagct 120 ccagggaagg gcctggagtg ggtctcaggt attagttgga atagtggtag cataggctat 180 gcggactctg tgaagggccg attcaccatc tccagagaca acgccaagaa ctccctgtat 240 ctgcaaatga acagtctgag agttgaggac acggccttgt attattgtgc aaaagataag 300 gggtattacg atattttgac tggagattac tactactact acggtatgga cgtctggggc 360 caagggacca cggtcaccgt ctcctca 387

<210> 290 <211> 129 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic <400> 290

Glu Val Gin Leu Val Glu Ser Gly Gly Gly Leu Val Gin Pro Gly Arg 1 5 10 15

Ser Leu Arg Leu Ser Cys Ala Ala Ser Gly lie Thr Phe Asp Asp Tyr 20 25 30

Ala Met Tyr Trp Val Arg Gin Ala Pro Gly Lys Gly Leu Glu Trp Val 35 40 45

Ser Gly lie Ser Trp Asn Ser Gly Ser lie Gly Tyr Ala Asp Ser Val 50 55 60

Lys Gly Arg Phe Thr lie Ser Arg Asp Asn Ala Lys Asn Ser Leu Tyr

65 70 75 80

Leu Gin Met Asn Ser Leu Arg Val Glu Asp Thr Ala Leu Tyr Tyr Cys

85 90 95

Ala Lys Asp Lys Gly Tyr Tyr Asp lie Leu Thr Gly Asp Tyr Tyr Tyr 100 105 110

Tyr Tyr Gly Met Asp Val Trp Gly Gin Gly Thr Thr Val Thr Val Ser 115 120 125

Ser

<210> 291 <211> 24 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 291 ggaatcacct ttgatgatta tgcc 24

<210> 292 <211> 8 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic <400> 292

Gly lie Thr Phe Asp Asp Tyr Ala 1 5

<210> 293 <211> 24 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 293 attagttgga atagtggtag cata 24

<210> 294 <211> 8 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic <400> 294 lie Ser Trp Asn Ser Gly Ser lie 1 5

<210> 295 <211> 66 <212> DNA

<213> Artificial Sequence <220>

<223> synthetic <400> 295 gcaaaagata aggggtatta cgatattttg actggagatt actactacta ctacggtatg 60 gacgtc 66

<210> 296 <211> 22 <212> PRT

<213> Artificial Sequence <220>

<223> synthetic mAb5385

HCVR

QVQLVQSGAEVKKPGASVTVSCQVSGYTLTSYGLSWVRQAPGQGLEWVGWINTYDGQ TKYVK

KFQGRVTMTTHTGTNTAYMEMKSLRSDDTAVYYCARVEGVRGVMGFHYYPMDVWGQG TMVTV

SS

HCDR1

GYTLTSYG

HCDR2

INTYDGQT

HCDR3

ARVEGVRGVMGFHYYPMDV

LCVR

QSVLTQPPSVSGAPGQRVTISCTGSSSNIGAGYAVHWYQQLPGTAPKLLISGNSNRP SGVPD RFSGSKSGTSASLAITGLQAEDEADYYCQSYDSSLSGSVFGGGTKLTVL

LCDR1

SSNIGAGYA

LCDR2

GNS

LCDR3

QSYDSSLSGSV