Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
COMPOSITIONS AND METHODS FOR THE TREATMENT OF BACTERIAL INFECTIONS
Document Type and Number:
WIPO Patent Application WO/2018/128826
Kind Code:
A1
Abstract:
Compositions and methods for the treatment of bacterial infections include conjugates containing an Fc domain covalently linked to one or more monomers of cyclic heptapeptides or one or more dimers of cyclic heptapeptides. In particular, conjugates can be used in the treatment of bacterial infections caused by Gram-negative bacteria.

Inventors:
AKERS-RODRIGUEZ SUZANNE (US)
BALKOVEC JAMES (US)
BENSEN DANIEL (US)
BLIZZARD TIMOTHY (US)
BORCHARDT ALLEN (US)
BRADY THOMAS (US)
CHEN ZHI-YONG (US)
DO QUYEN-QUYEN (US)
FORTIER JOANNE (US)
JIANG WANLONG (US)
LAM THANH (US)
TARI LESLIE (US)
Application Number:
PCT/US2017/067750
Publication Date:
July 12, 2018
Filing Date:
December 20, 2017
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
CIDARA THERAPEUTICS INC (US)
International Classes:
A61K38/00; A61K38/04; A61K38/12; A61K38/14
Domestic Patent References:
WO2011024887A12011-03-03
Foreign References:
US20050255042A12005-11-17
US5593960A1997-01-14
Attorney, Agent or Firm:
BELLIVEAU, Michael, J. (US)
Download PDF:
Claims:
1. A conjugate described by formula (1):

wherein each M1 comprises a first cyclic heptapeptide comprising a linking nitrogen and each M2 comprises a second cyclic heptapeptide comprising a linking nitrogen;

each E comprises an Fc domain monomer;

L’1 in each M2-L’1-M1 is a linker covalently attached to a sulfur atom of a hinge cysteine in each E and to the linking nitrogen in each of M1 and M2;

T is an integer from 1, 2, 3, 4, or 5,

the two squiggly lines connected to the two Es indicate that each M2-L’1-M1 is covalently attached to a pair of sulfur atoms of two hinge cysteines in the two Es,

or a pharmaceutically acceptable salt thereof. 2. A conjugate described by formula (2):

wherein each M comprises a cyclic heptapeptide comprising a linking nitrogen;

each E comprises an Fc domain monomer;

L’1 in each L’1-M is a linker covalently attached to a sulfur atom in a hinge cysteine in E and to the linking nitrogen in M;

T is 1, 2, 3, 4, or 5,

the two squiggly lines connected to the two sulfur atoms indicate that each L’1-M is covalently attached to a pair of sulfur atoms of two hinge cysteines in the two Es,

or a pharmaceutically acceptable salt thereof. 3. The conjugate of claim 1 or 2, wherein each E comprises an Fc domain monomer having the sequence of any one of SEQ ID NOs: 1-31. 4. The conjugate of any one of claims 1-3, wherein each Fc domain monomer comprises the sequence of

MVRSDKTHTCPPCPPC*KC*PAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYV DGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQV YTLPPSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRW QQGNVFSCSVMHEALHNHYTQKSLSLSPGKHHHHHH (SEQ ID NO 10) Cys13, Cys16, or Cys18 of SEQ ID NO: 10. 6. The conjugate of claim 4, wherein the pair of sulfur atoms are the sulfur atoms of Cys10 and Cys13, Cys10 and Cys16, Cys 10 and Cys18, Cys13 and Cys 16, Cys13 and Cys 18, or Cys 16 and Cys 18 in the two Es. 7. The conjugate of any one of claims 1-6, wherein T is 1, 2, 3, or 4. . Th n f l im 1 wh r in h n h h r r :

9. The conjugate of claim 1, wherein the conjugate has the structure:

. 10. A population of conjugates of claim 1, wherein the average value of T is 1 to 5. ( )

wherein each M1 comprises a first cyclic heptapeptide comprising a linking nitrogen and each M2 comprises a second cyclic heptapeptide comprising a linking nitrogen;

E comprises an Fc domain monomer;

L’1 in each M2-L’1-M1 is a linker covalently attached to a sulfur atom of a hinge cysteine in E and to the linking nitrogen in each of M1 and M2;

T is 1, 2, 3, 4, or 5,

the squiggly line connected to the E indicates that each M2-L’1-M1 is covalently attached to a sulfur atom of a hinge cysteine in E,

or a pharmaceutically acceptable salt thereof. 12. The conjugate of claim 11, wherein each E comprises an Fc domain monomer having the sequence of any one of SEQ ID NOs: 1-31. 13. A conjugate described by formula (4):

wherein each M comprises a cyclic heptapeptide comprising a linking nitrogen;

E comprises an Fc domain monomer;

L’1 in each L’1-M is a linker covalently attached to a sulfur atom of a hinge cysteine in E and to the linking nitrogen in M;

T is 1, 2, 3, 4, or 5;

the squiggly line connected to E indicates that each L’1-M is covalently attached to the sulfur atom of the hinge cysteine in E,

or a pharmaceutically acceptable salt thereof. 14. The conjugate of claim 13, wherein each E comprises an Fc domain monomer having the sequence of any one of SEQ ID NOs: 1-31. 15. The conjugate of any one of claims 11-14, wherein E has the sequence of SEQ ID NO: 10. 16 The conjugate of claim 15 wherein the sulfur atom is in Cys10 Cys13 Cys16 or Cys18 of SEQ ID

17. The conjugate of any one of claims 11-14, wherein T is 1, 2, 3, or 4. 18. The conjugate of claim 11, wherein the conjugate has the structure:

. 19. The conjugate of claim 13, wherein the conjugate has the structure: . 20. A population of conjugates of claim 11 or 13, wherein the average value of T is 1 to 5. 21. A population of conjugates of claim 11 or 13, wherein the average value of T is 1 to 2. 22. A conjugate described by formula (3):

(3)

wherein each M1 comprises a first cyclic heptapeptide comprising a linking nitrogen and each M2 comprises a second cyclic heptapeptide comprising a linking nitrogen;

E comprises an Fc domain monomer;

L’1 in each M2- L’1-M1 is a linker covalently attached to a nitrogen atom of a surface exposed lysine in E and to the linking nitrogen in each of M1 and M2;

T is 1, 2, 3, 4, or 5,

the squiggly line connected to the E indicates that each M2-L’1-M1 is covalently attached to the nitrogen atom of a surface exposed lysine in E,

or a pharmaceutically acceptable salt thereof. of any one of SEQ ID NOs: 1-31. 24. A conjugate described by formula (4):

wherein each M comprises a cyclic heptapeptide comprising a linking nitrogen;

E comprises an Fc domain monomer;

L’1 in each L’1-M is a linker covalently attached to a nitrogen atom of a surface exposed lysine in E and to the linking nitrogen in M;

T is an integer from 1 to 11,

the squiggly line connected to E indicates that each L’1-M is covalently attached to the nitrogen atom of a surface exposed lysine in E,

or a pharmaceutically acceptable salt thereof. 25. The conjugate of claim 24, wherein each E comprises an Fc domain monomer having the sequence of any one of SEQ ID NOs: 1-31. 26. The conjugate of any one of claims 22-25, wherein E comprises the sequence of

MVRSDKTHTCPPCPPC*KC*PAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYV DGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQV YTLPPSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRW QQGNVFSCSVMHEALHNHYTQKSLSLSPGKHHHHHH (SEQ ID NO: 10). 27. The conjugate of claim 26, wherein each nitrogen atom is indendently in Lys35, Lys63, Lys77, Lys79, Lys106, Lys123, Lys129, Lys181, Lys203, Lys228, or Lys236 of SEQ ID NO: 10.

. 2 . Th n f l im 24 wh r in the conjugate has the structure

30. A population of conjugates of claim 11 or 24, wherein the average value of T is 1 to 5. 31. A population of conjugates of claim 11 or 24, wherein the average value of T is 1 to 2. 32. The conjugate of any one of claims 1-31, wherein the conjugate forms a homodimer comprising an Fc domain. 33. The conjugate of any one of claims 1-32, wherein E homodimerizes with another E to form an Fc domain. 34. A conjugate comprising

(i) a first cyclic heptapeptide;

(ii) a second cyclic heptapeptide;

(iii) an Fc domain monomer or an Fc domain; and

(iv) a linker covalently attached to the first cyclic heptapeptide, the second cyclic heptapeptide, and the Fc domain monomer or the Fc domain.

(D-Ia)

wherein each M1 comprises a first cyclic heptapeptide comprising a linking nitrogen and each M2 comprises a second cyclic heptapeptide comprising a linking nitrogen;

L’ in each M2-L’-M1 is a linker covalently attached to the Fc domain monomer and to the linking nitrogen in each of M1 and M2;

each E is an Fc domain monomer;

n is 1 or 2;

T is an integer from 1 to 20,

or a pharmaceutically acceptable salt thereof. 36. The conjugate of claim 35, wherein when n is 2, E dimerizes to form an Fc domain, and the conjugate is described by formula (D-Ib):

(D-Ib)

wherein J is an Fc domain; and

T is an integer from 2 to 20,

or a pharmaceutically acceptable salt thereof. 37. The conjugate of claim 35 or 36, wherein L’ in each M2-L’-M1 is described by formula (D-L):

(D-L)

wherein L is a remainder of L’;

A1 is a 1-5 amino acid peptide covalently attached to the linking nitrogen in each M1 or is absent; and

A2 is a 1-5 amino acid peptide covalently attached to the linking nitrogen in each M2 or is absent.

(D-II)

wherein L is a remainder of L’;

each of R1, R12, R’1, and R’12 is, independently, a lipophilic moiety, a polar moiety, or H;

each of R11, R13, R14, R’11, R’13, and R’14 is, independently, optionally substituted C1-C5 alkamino, a polar moiety, a positively charged moiety, or H;

each of R15 and R’15 is, independently, a lipophilic moiety or a polar moiety;

each of R2, R3, R4, R5, R6, R7, R8, R9, R10, R’2, R’3, R’4, R’5, R’6, R’7, R’8, R’9, and R’10 is, independently, a lipophilic moiety, a positively charged moiety, a polar moiety, H, optionally substituted C1-C5 alkamino, optionally substituted C1-C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5- C15 aryl, optionally substituted C1-C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl; or two R groups on the same or adjacent atoms join to form an optionally substituted 5-8 membered ring;

each of a’, b’, c’, a, b, and c is, independently, 0 or 1;

each of N1, N2, N3, N4, N’1, N’2, N’3, and N’4 is a nitrogen atom;

each of C1, C2, C3, C’1, C’2, and C’3 is a carbon atom,

or a pharmaceutically acceptable salt thereof. 39. The conjugate of claim 38, wherein the conjugate comprises at least one optionally substituted 5-8 membered ring formed by joining (i) R2, R3, and C1; (ii) R3, R4, N1, and C1; (iii) R5, R6, and C2; (iv) R6, R7, N2, and C2; (v) R8, R9, and C3; (vi) R9, R10, N3, and C3; (vii) R’2, R’3, and C’1; (viii) R’3, R’4, N’1, and C’1; (ix) R’5, R’6, and C’2; (x) R’6, R’7, N’2, and C’2; (xi) R’8, R’9, and C’3; or (xii) R’9, R’10, N’3, and C’3.

(D-III)

or a pharmaceutically acceptable salt thereof. 41. The conjugate of any one of claims 38-40, wherein

each of R1, R12, R’1, and R’12 is, independently, a lipophilic moiety;

each of R11, R13, R14, R’11, R’13, and R’14 is, independently, optionally substituted C1-C5 alkamino, a polar moiety, or a positively charged moiety; and/or

each of R15 and R’15 is, independently, a polar moiety. 42. The conjugate of any one of claims 35-41, wherein each of R1 and R12 is a lipophilic moiety. 43. The conjugate of any one of claims 35-42, wherein each of R’1 and R’12 is a lipophilic moiety. 44. The conjugate of claim 42 or 43, wherein each lipophilic moiety is, independently, optionally substituted C1-C20 alkyl, optionally substituted C5-C15 aryl, optionally substituted C6-C35 alkaryl, or optionally C5-C10 substituted heteroaryl. 45. The conjugate of any one of claims 42-44, wherein each lipophilic moiety is, independently, C1-C8 alkyl, methyl substituted C2-C4 alkyl, (C1-C10)alkylene(C6)aryl, phenyl substituted (C1- C10)alkylene(C6)aryl, or alkyl substituted C4-C9 heteroaryl. 46. The conjugate of any one of claims 42-45, wherein each lipophilic moiety is, independently, benzyl, isobutyl, sec-butyl, isopropyl, n-propyl, methyl, biphenylmethyl, n-octyl, or methyl substituted indolyl. 47. The conjugate of any one of claims 38-46, wherein each of R11, R13, R14, R’11, R’13, and R’14 is independently optionally substituted C1-C5 alkamino. 48. The conjugate of claim 47, wherein each of R11, R13, R14, R’11, R’13, and R’14 is CH2CH2NH2.

50. The conjugate of claim 49, wherein each polar moiety comprises a hydroxyl group, a carboxylic acid group, an ester group, or an amide group. 51. The conjugate of claim 49 or 50, wherein each polar moiety is hydroxyl substituted C1-C4 alkyl. 52. The conjugate of any one of claims 49-51, wherein each polar moiety is CHCH3OH. 53. The conjugate of claim 38, wherein the conjugate is described by formula (D-IV):

wherein each of R’1 and R1 is, independently, benzyl or CH2CH(CH3)2,

or a pharmaceutically acceptable salt thereof. 54. The conjugate of claim 53, wherein the conjugate is described by formula (D-IV-1):

wherein each of R’1 and R1 is, independently, benzyl or CH2CH(CH3)2,

or a pharmaceutically acceptable salt thereof.

(D-IV-2)

wherein each of R’1 and R1 is, independently, benzyl or CH2CH(CH3)2,

or a pharmaceutically acceptable salt thereof. 56. The conjugate of claim 53, wherein the conjugate is described by formula (D-IV-3):

(D-IV-3)

wherein each of R’1 and R1 is, independently, benzyl or CH2CH(CH3)2;

each of R’16 and R16 is, independently, a lipophilic moiety, optionally substituted C1-C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1-C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl; or a pharmaceutically acceptable salt thereof. 57. The conjugate of claim 56, each of R’16 and R16 is, independently, C1-C6 alkyl or benzyl.

(D-V)

wherein each of R’1 and R1 is, independently, benzyl or CH2CH(CH3)2;

each of R2, R6, R’2, and R’6 is, independently, a positively charged moiety, a polar moiety, optionally substituted C1-C5 alkamino, optionally substituted C1-C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1-C20 heteroalkyl, optionally substituted C3- C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl,

or a pharmaceutically acceptable salt thereof. 59. The con u ate of claim 58 wherein the con u ate is described b formula D-V-1 :

(D-V-1)

wherein each of R’1 and R1 is, independently, benzyl or CH2CH(CH3)2;

or a pharmaceutically acceptable salt thereof.

(D-V-2)

wherein each of R’1 and R1 is, independently, benzyl or CH2CH(CH3)2;

or a pharmaceutically acceptable salt thereof. 61. The conu ate of claim 58 wherein the con u ate is described b formula D-V-3 :

(D-V-3)

wherein each of R’1 and R1 is, independently, benzyl or CH2CH(CH3)2;

or a pharmaceutically acceptable salt thereof.

(D-V-4)

wherein each of R’1 and R1 is, independently, benzyl or CH2CH(CH3)2;

or a pharmaceutically acceptable salt thereof. 63. The conjugate of claim 61 or 62, wherein the conjugate is described by formula (D-V-5):

(D-V-5)

wherein each of R’1 and R1 is, independently, benzyl or CH2CH(CH3)2;

or a pharmaceutically acceptable salt thereof.

64. The conjugate of claim 58, wherein the conjugate is described by formula (D-V-6):

(D-V-6)

wherein each of R'1 and R1 is, independently, benzyl or CH2CH(CH3)2;

each of R'16 and R16 is, independently, a lipophilic moiety, optionally substituted C1 -C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1 -C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20

heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl; or a pharmaceutically acceptable salt thereof.

65. The conjugate of claim 64, wherein the conjugate is described by formula (D-V-7):

(D-V-7)

wherein each of R'1 and R1 is, independently, benzyl or CH2CH(CH3)2;

each of R'16 and R16 is, independently, a lipophilic moiety, optionally substituted C1 -C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1 -C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20

heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl; or a pharmaceutically acceptable salt thereof.

66. The n f l im wh r in h n i ri f rm l D-V- :

(D-V-8)

wherein each of R’1 and R1 is, independently, benzyl or CH2CH(CH3)2;

each of R’16 and R16 is, independently, a lipophilic moiety, optionally substituted C1-C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1-C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl; or a pharmaceutically acceptable salt thereof. 67. The conjugate of claim 66, wherein the conjugate is described by formula (D-V-9):

(D-V-9)

wherein each of R’1 and R1 is, independently, benzyl or CH2CH(CH3)2;

each of R’16 and R16 is, independently, a lipophilic moiety, optionally substituted C1-C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally

C C C C C C heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl; or a pharmaceutically acceptable salt thereof. 68. The conjugate of claim 66 or 67, wherein the conjugate is described by formula (D-V-10):

wherein each of R’1 and R1 is, independently, benzyl or CH2CH(CH3)2;

each of R’16 and R16 is, independently, a lipophilic moiety, optionally substituted C1-C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1-C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl; or a pharmaceutically acceptable salt thereof. 69. The conjugate of any one of claims 64-68, wherein each of R’16 and R16 is, independently, C1-C6 alkyl, benzyl, or phenethyl. 70. The conjugate of claim 38, wherein the conjugate is described by formula (D-VI):

each of R2, R6, R8, R’2, R’6, and R’8 is, independently, a positively charged moiety, a polar moiety, optionally substituted C1-C5 alkamino, optionally substituted C1-C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1-C20 heteroalkyl, optionally substituted C3- C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl,

or a pharmaceutically acceptable salt thereof. 71. The conjugate of claim 70, wherein the conjugate is described by formula (D-VI-1):

(D-VI-1)

wherein each of R’1 and R1 is, independently, benzyl or CH2CH(CH3)2;

or a pharmaceutically acceptable salt thereof. 72. The conjugate of claim 71, wherein the conjugate is described by formula (D-VI-2):

(D-VI-2)

wherein each of R’1 and R1 is, independently, benzyl or CH2CH(CH3)2;

or a pharmaceutically acceptable salt thereof

(D-VI-2a)

wherein each of R’1 and R1 is, independently, benzyl or CH2CH(CH3)2;

or a pharmaceutically acceptable salt thereof. 74. Th n f l im 72 wh r in h n i ri f rm l D-VI-2 :

wherein each of R’1 and R1 is, independently, benzyl or CH2CH(CH3)2;

or a pharmaceutically acceptable salt thereof.

wherein each E is an Fc domain monomer;

n is 1 or 2;

T is an integer from 1 to 20;

each of R’1 and R1 is, independently, benzyl or CH2CH(CH3)2,

or a pharmaceutically acceptable salt thereof. 76. Th n f l im 72 wh r in h n i ri h f rm l D-VI-2 :

wherein each of R’1 and R1 is, independently, benzyl or CH2CH(CH3)2;

or a pharmaceutically acceptable salt thereof.

wherein each of R’1 and R1 is, independently, benzyl or CH2CH(CH3)2; or a pharmaceutically acceptable salt thereof. 78. A conu ate described b formula D-VI-2d :

( )

wherein each E is an Fc domain monomer;

n is 1 or 2;

T is an integer from 1 to 20;

each of R’1 and R1 is, independently, benzyl or CH2CH(CH3)2, or a pharmaceutically acceptable salt thereof.

(D VI 2e)

wherein each of R’1 and R1 is, independently, benzyl or CH2CH(CH3)2;

or a pharmaceutically acceptable salt thereof. 80. The conjugate of claim 72, wherein the conjugate is described by formula (D-VI-2f):

( )

wherein each of R’1 and R1 is, independently, benzyl or CH2CH(CH3)2;

or a pharmaceutically acceptable salt thereof.

(D-VI-2f)

wherein each E is an Fc domain monomer;

n is 1 or 2;

T is an integer from 1 to 20;

each of R’1 and R1 is, independently, benzyl or CH2CH(CH3)2,

or a pharmaceutically acceptable salt thereof. 82. The conjugate of claim 70, wherein the conjugate is described by formula (D-VI-3):

(D-VI-3)

wherein each of R’1 and R1 is, independently, benzyl or CH2CH(CH3)2;

or a pharmaceutically acceptable salt thereof.

(D-VI-4)

wherein each of R’1 and R1 is, independently, benzyl or CH2CH(CH3)2;

or a pharmaceutically acceptable salt thereof. 84. The conjugate of claim 82 or 83, wherein the conjugate is described by formula (D-VI-5):

(D-VI-5)

wherein each of R’1 and R1 is, independently, benzyl or CH2CH(CH3)2;

or a pharmaceutically acceptable salt thereof.

(D-VI-6)

wherein each of R’1 and R1 is, independently, benzyl or CH2CH(CH3)2;

each of R’16 and R16 is, independently, a lipophilic moiety, optionally substituted C1-C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1-C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl; or a pharmaceutically acceptable salt thereof. 86. The conu ate of claim 76 wherein the con u ate is described b the formula D-VI-6a :

(D-VI-6a)

wherein each of R’1 and R1 is, independently, benzyl or CH2CH(CH3)2;

or a pharmaceutically acceptable salt thereof.

(D-VI-6b)

wherein each of R’1 and R1 is, independently, benzyl or CH2CH(CH3)2; or a pharmaceutically acceptable salt thereof. 88. A conjugate described by formula (D-VI-6b):

(D-VI-6b)

wherein each E is an Fc domain monomer;

n is 1 or 2;

T is an integer from 1 to 20;

each of R’1 and R1 is, independently, benzyl or CH2CH(CH3)2, or a pharmaceutically acceptable salt thereof.

wherein each of R’1 and R1 is, independently, benzyl or CH2CH(CH3)2;

or a pharmaceutically acceptable salt thereof. 90. The conjugate of claim 76, wherein the conjugate is described by formula (D-VI-6d):

wherein each of R’1 and R1 is, independently, benzyl or CH2CH(CH3)2;

or a pharmaceutically acceptable salt thereof.

(D-VI-6d)

wherein each E is an Fc domain monomer;

n is 1 or 2;

T is an integer from 1 to 20;

each of R’1 and R1 is, independently, benzyl or CH2CH(CH3)2,

or a pharmaceutically acceptable salt thereof. 92. The conjugate of claim 85, wherein the conjugate is described by formula (D-VI-7):

(D-VI-7)

wherein each of R’1 and R1 is, independently, benzyl or CH2CH(CH3)2;

each of R’16 and R16 is, independently, a lipophilic moiety, optionally substituted C1-C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1-C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl; or a pharmaceutically acceptable salt thereof. 93. The conjugate of claim 70, wherein the conjugate is described by formula (D-VI-8):

(D-VI-8)

wherein each of R’1 and R1 is, independently, benzyl or CH2CH(CH3)2;

each of R’16 and R16 is, independently, a lipophilic moiety, optionally substituted C1-C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1-C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl; or a pharmaceutically acceptable salt thereof. 94. Th n f l im wh r in h n i ri f rm l D-VI- :

(D-VI-9)

wherein each of R’1 and R1 is, independently, benzyl or CH2CH(CH3)2; optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1-C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl; or a pharmaceutically acceptable salt thereof. 95. The conjugate of claim 93 or 94, wherein the conjugate is described by formula (D-VI-10):

wherein each of R’1 and R1 is, independently, benzyl or CH2CH(CH3)2;

each of R’16 and R16 is, independently, a lipophilic moiety, optionally substituted C1-C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1-C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl; or a pharmaceutically acceptable salt thereof. 96. The conjugate of any one of claims 85-95, wherein each of R’16 and R16 is, independently, C1-C6 alkyl or benzyl.

(D-VII)

wherein each of R’1 and R1 is, independently, benzyl or CH2CH(CH3)2;

each of R2, R6, R8, R’2, and R’6 is, independently, a positively charged moiety, a polar moiety, optionally substituted C1-C5 alkamino, optionally substituted C1-C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1-C20 heteroalkyl, optionally substituted C3- C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl,

or a pharmaceutically acceptable salt thereof. 98. The con u ate of claim 38 wherein the con u ate is described b formula D-VIII :

(D-VIII)

wherein each of R’1 and R1 is, independently, benzyl or CH2CH(CH3)2;

each of R2, R6, R8, and R’2 is, independently, a positively charged moiety, a polar moiety, optionally substituted C1-C5 alkamino, optionally substituted C1-C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl,

or a pharmaceutically acceptable salt thereof. 99. The conjugate of claim 38, wherein the conjugate is described by formula (D-IX):

(D-IX)

wherein each of R’1 and R1 is, independently, benzyl or CH2CH(CH3)2;

each of R2, R6, and R’2 is, independently, a positively charged moiety, a polar moiety, optionally substituted C1-C5 alkamino, optionally substituted C1-C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1-C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl,

or a pharmaceutically acceptable salt thereof. 100. The conjugate of claim 99, wherein the conjugate is described by formula (D-IX-1):

(D-IX-1)

101. The conjugate of claim 100, wherein the conjugate is described by formula (D-IX-2):

(D-IX-2)

wherein each of R’1 and R1 is, independently, benzyl or CH2CH(CH3)2;

or a pharmaceutically acceptable salt thereof. 102. The conjugate of claim 99, wherein the conjugate is described by formula (D-IX-3):

(D-IX-3)

wherein each of R’1 and R1 is, independently, benzyl or CH2CH(CH3)2;

each of R’16 and R16 is, independently, a lipophilic moiety, optionally substituted C1-C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1-C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl; or a pharmaceutically acceptable salt thereof.

(D-IX-4)

wherein each of R’1 and R1 is, independently, benzyl or CH2CH(CH3)2;

each of R’16 and R16 is, independently, a lipophilic moiety, optionally substituted C1-C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1-C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20

heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl;

or a pharmaceutically acceptable salt thereof. 104. The conjugate of any one of claim 102 or 103, wherein each of R’16 and R16 is, independently, C1- C6 alkyl, benzyl, or phenethyl. 105. The conjugate of claim 38, wherein the conjugate is described by formula (D-X):

(D-X)

wherein each of R’1 and R1 is, independently, benzyl or CH2CH(CH3)2;

each of R2 and R’2 is, independently, a positively charged moiety, a polar moiety, optionally substituted C1 C5 alkamino optionally substituted C1 C20 alkyl optionally substituted C3 C20 cycloalkyl heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl,

or a pharmaceutically acceptable salt thereof. 106. The conjugate of claim 38, wherein the conjugate is described by formula (D-XI):

(D-XI)

wherein each of R’1 and R1 is, independently, benzyl or CH2CH(CH3)2;

each of R2 and R6 is, independently, a positively charged moiety, a polar moiety, optionally substituted C1-C5 alkamino, optionally substituted C1-C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1-C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl,

or a pharmaceutically acceptable salt thereof. 107. The conjugate of claim 106, wherein the conjugate is described by formula (D-XI-1):

wherein each of R’1 and R1 is, independently, benzyl or CH2CH(CH3)2;

or a pharmaceutically acceptable salt thereof. 108. The conjugate of claim 107, wherein the conjugate is described by formula (D-XI-2):

(D-XI-2)

wherein each of R’1 and R1 is, independently, benzyl or CH2CH(CH3)2;

or a pharmaceutically acceptable salt thereof. 109. The conjugate of claim 106, wherein the conjugate is described by formula (D-XI-3):

(D-XI-3)

wherein each of R’1 and R1 is, independently, benzyl or CH2CH(CH3)2;

R16 is a lipophilic moiety, optionally substituted C1-C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1-C20 heteroalkyl, optionally substituted C3- C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or

C C

110. The conjugate of claim 109, wherein the conjugate is described by formula (D-XI-4):

(D-XI-4)

wherein each of R’1 and R1 is, independently, benzyl or CH2CH(CH3)2;

R16 is a lipophilic moiety, optionally substituted C1-C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1-C20 heteroalkyl, optionally substituted C3- C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl;

or a pharmaceutically acceptable salt thereof. 111. The conjugate of any one of claim 109 or 110, wherein R16 is C1-C6 alkyl, benzyl, or phenethyl. 112. The conjugate of claim 35 or 36, wherein the conjugate is described by formula (D-XII):

each of R2 and R’2 is, independently, a positively charged moiety, a polar moiety, optionally substituted C1-C5 alkamino, optionally substituted C1-C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1-C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl;

R6, R7, N2, and C2 together form an optionally substituted 5-8 membered ring comprising optionally substituted C3-C7 heterocycloalkyl comprising an N heteroatom and additional 0-2

heteroatoms independently selected from N, O, and S, or optionally substituted C2-C7 heteroaryl comprising an N heteroatom and additional 0-2 heteroatoms independently selected from N, O, and S;

R’6, R’7, N’2, and C’2 together form an optionally substituted 5-8 membered ring comprising optionally substituted C3-C7 heterocycloalkyl comprising an N heteroatom and additional 0-2

heteroatoms independently selected from N, O, and S, or optionally substituted C2-C7 heteroaryl comprising an N heteroatom and additional 0-2 heteroatoms independently selected from N, O, and S; or a pharmaceutically acceptable salt thereof. 113. The conjugate of claim 112, wherein the conjugate is described by formula (D-XII-1):

(D-XII-1)

wherein each of R’1 and R1 is, independently, benzyl or CH2CH(CH3)2;

or a pharmaceutically acceptable salt thereof.

(D-XIII)

wherein each of R’1 and R1 is, independently, benzyl or CH2CH(CH3)2,

or a pharmaceutically acceptable salt thereof. 115. The conjugate of claim 35 or 36, wherein the conjugate is described by formula (D-XIV):

(D-XIV)

wherein each A1 is a 1-5 amino acid peptide covalently attached to the linking nitrogen in each M1;

each A2 is a 1-5 amino acid peptide covalently attached to the linking nitrogen in each M2, or is absent;

each of L and L1 is a remainder of L’;

or a pharmaceutically acceptable salt thereof. 116. The conjugate of any one of claims 58-114, wherein R2 is optionally substituted C1-C5 alkamino. 117. The conjugate of any one of claims 58-114, wherein R’2 is optionally substituted C1-C5 alkamino. 118. The conjugate of claim 116 or 117, wherein the optionally substituted C1-C5 alkamino is CH2NH2 or

120. The conjugate of any one of claims 58-114, wherein R’2 is a polar moiety. 121. The conjugate of claim 119 or 120, wherein the polar moiety comprises a hydroxyl group, a carboxylic acid group, an ester group, or an amide group. 122. The conjugate of any one of claims 119-121, wherein the polar moiety is hydroxyl substituted C1-C4 alkyl. 123. The conjugate of claim 117, wherein the polar moiety is CHCH3OH or CH2OH. 124. The conjugate of any one of claims 58-114, wherein R6 is a polar moiety. 125. The conjugate of any one of claims 58-114, wherein R’6 is a polar moiety. 126. The conjugate of claim 124 or 125, wherein the polar moiety comprises a hydroxyl group, a carboxylic acid group, an ester group, or an amide group. 127. The conjugate of any one of claims 124-126, wherein the polar moiety is hydroxyl substituted C1-C4 alkyl. 128. The conjugate of claim 127, wherein the polar moiety is CHCH3OH or CH2OH. 129. The conjugate of any one of claims 55-114, wherein R8 is optionally substituted C1-C5 alkamino. 130. The conjugate of any one of claims 55-114, wherein R’8 is optionally substituted C1-C5 alkamino. 131. The conjugate of claim 129 or 130, wherein the optionally substituted C1-C5 alkamino is CH2NH2 or

132. The conjugate of any one of claims 55-114, wherein R8 is optionally substituted C5-C15 aryl. 133. The conjugate of any one of claims 55-114, wherein R’8 is optionally substituted C5-C15 aryl. 134. The conjugate of claim 132 or 133, wherein the optionally substituted C5-C15 aryl is naphthyl. comprising C4-C5 heterocycloalkyl comprising an N heteroatom and additional 0 or 1 heteroatom independently selected from N, O, and S; and

wherein R’6, R’7, N’2, and C’2 together form a 5- or 6-membered ring comprising C4-C5 heterocycloalkyl comprising an N heteroatom and additional 0 or 1 heteroatom independently selected from N, O, and S. 136. The conjugate of any one of claims 35-135, wherein L’, L, or L1 comprises one or more optionally substituted C1-C20 alkylene, optionally substituted C1-C20 heteroalkylene, optionally substituted C2-C20 alkenylene, optionally substituted C2-C20 heteroalkenylene, optionally substituted C2-C20 alkynylene, optionally substituted C2-C20 heteroalkynylene, optionally substituted C3-C20 cycloalkylene, optionally substituted C3-C20 heterocycloalkylene, optionally substituted C4-C20 cycloalkenylene, optionally substituted C4-C20 heterocycloalkenylene, optionally substituted C8-C20 cycloalkynylene, optionally substituted C8-C20 heterocycloalkynylene, optionally substituted C5-C15 arylene, optionally substituted C2-C15 heteroarylene, O, S, NRi, P, carbonyl, thiocarbonyl, sulfonyl, phosphate, phosphoryl, or imino, wherein Ri is H, optionally substituted C1-C20 alkyl, optionally substituted C1-C20 heteroalkyl, optionally substituted C2-C20 alkenyl, optionally substituted C2-C20 heteroalkenyl, optionally substituted C2-C20 alkynyl, optionally substituted C2-C20 heteroalkynyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C5-C15 aryl, or optionally substituted C2-C15 heteroaryl. 137. The conjugate of claim 136, wherein the backbone of L’, L, or L1 consists of one or more optionally substituted C1-C20 alkylene, optionally substituted C1-C20 heteroalkylene, optionally substituted C2-C20 alkenylene, optionally substituted C2-C20 heteroalkenylene, optionally substituted C2-C20 alkynylene, optionally substituted C2-C20 heteroalkynylene, optionally substituted C3-C20 cycloalkylene, optionally substituted C3-C20 heterocycloalkylene, optionally substituted C4-C20 cycloalkenylene, optionally substituted C4-C20 heterocycloalkenylene, optionally substituted C8-C20 cycloalkynylene, optionally substituted C8-C20 heterocycloalkynylene, optionally substituted C5-C15 arylene, optionally substituted C2-C15 heteroarylene, O, S, NRi, P, carbonyl, thiocarbonyl, sulfonyl, phosphate, phosphoryl, or imino, wherein Ri is H, optionally substituted C1-C20 alkyl, optionally substituted C1-C20 heteroalkyl, optionally substituted C2-C20 alkenyl, optionally substituted C2-C20 heteroalkenyl, optionally substituted C2-C20 alkynyl, optionally substituted C2-C20 heteroalkynyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C5-C15 aryl, or optionally substituted C2-C15 heteroaryl.

138. The conjugate of claim 136 or 137, wherein L', L, or L1 is oxo substituted.

139. The conjugate of any one of claims 35-138, wherein the backbone of L', L, or L1 comprises no more than 250 atoms.

140. The conjugate of any one of claims 35-139, wherein L', L, or L1 is capable of forming an amide, a carbamate, a sulfonyl, or a urea linkage.

141 . The conjugate of any one of claims 37-135, wherein L or L1 is a bond.

142. The conjugate of any one of claims 35-141 , wherein each L is described by formula (D-L-l):

c

wherein LA is described by formula

optionally substituted C1 -C20 alkylene, optionally substituted C1 -C20 heteroalkylene, optionally substituted C2-C20 alkenylene, optionally substituted C2-C20 heteroalkenylene, optionally substituted C2-C20 alkynylene, optionally substituted C2-C20 heteroalkynylene, optionally substituted C3-C20 cycloalkylene, optionally substituted C3-C20 heterocycloalkylene, optionally substituted C4-C20 cycloalkenylene, optionally substituted C4-C20 heterocycloalkenylene, optionally substituted C8-C20 cycloalkynylene, optionally substituted C8-C20 heterocycloalkynylene, optionally substituted C5-C1 5 arylene, or optionally substituted C2-C15 heteroarylene;

each of and YC4 is, independently, O, S, NR',

P, carbonyl, thiocarbonyl, sulfonyl, phosphate, phosphoryl, or imino;

R' is H, optionally substituted C1 -C20 alkyl, optionally substituted C1 -C20 heteroalkyl, optionally substituted C2-C20 alkenyl, optionally substituted C2-C20 heteroalkenyl, optionally substituted C2-C20 alkynyl, optionally substituted C2-C20 heteroalkynyl, optionally substituted C3-C20 cycloalkyi, optionally C4-C20 heterocycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C5-C15 aryl, or optionally substituted C2-C15 heteroaryl; each of g1, h1, i1, j1, k1, l1, m1, n1, o1, g2, h2, i2, j2, k2, l2, m2, n2, o2, g3, h3, i3, j3, k3, l3, m3, n3, and o3 is, independently, 0 or 1;

Q is a nitrogen atom, optionally substituted C1-C20 alkylene, optionally substituted C1-C20 heteroalkylene, optionally substituted C2-C20 alkenylene, optionally substituted C2-C20

heteroalkenylene, optionally substituted C2-C20 alkynylene, optionally substituted C2-C20

heteroalkynylene, optionally substituted C3-C20 cycloalkylene, optionally substituted C3-C20 heterocycloalkylene, optionally substituted C4-C20 cycloalkenylene, optionally substituted C4-C20 heterocycloalkenylene, optionally substituted C8-C20 cycloalkynylene, optionally substituted C8-C20 heterocycloalkynylene, optionally substituted C5-C15 arylene, or optionally substituted C2-C15 heteroarylene. 143. The conjugate of claim 142, wherein L is

,

,

, ,

wherein R* is a bond or comprises one or more of optionally substituted C1-C20 alkylene, optionally substituted C1-C20 heteroalkylene, optionally substituted C2-C20 alkenylene, optionally substituted C2- C20 heteroalkenylene, optionally substituted C2-C20 alkynylene, optionally substituted C2-C20 heteroalkynylene, optionally substituted C3-C20 cycloalkylene, optionally substituted C3-C20 heterocycloalkylene, optionally substituted C4-C20 cycloalkenylene, optionally substituted C4-C20 heterocycloalkenylene, optionally substituted C8-C20 cycloalkynylene, optionally substituted C8-C20 heterocycloalkynylene, optionally substituted C5-C15 arylene, optionally substituted C2-C15 heteroarylene, O, S, NRi, P, carbonyl, thiocarbonyl, sulfonyl, phosphate, and imino, and

wherein Ri is H, optionally substituted C1-C20 alkyl, optionally substituted C1-C20 heteroalkyl, optionally substituted C2-C20 alkenyl, optionally substituted C2-C20 heteroalkenyl, optionally substituted C2 C20 lk l ti ll b tit t d C2 C20 h t lk l ti ll b tit t d C3 C20 l lk l C8-C20 heterocycloalkynyl, optionally substituted C5-C15 aryl, or optionally substituted C2-C15 heteroaryl. 144. The conjugate of any one of claims 35-143, wherein E has the sequence of any one of SEQ ID NOs: 1-31. 145. The conjugate of any one of claims 35-144, wherein when n is 2, E dimerizes to form an Fc domain. 146. A conjugat f f rm l D-XV :

(D-XV)

wherein:

each A is an independently selected amino acid;

each E is an Fc domain monomer;

each L is a linker that, when each m is 2, 3, 4, or 5, is bound to any of A and covalently attached to E;

each m is 0, 1, 2, 3, 4, or 5;

n is 1 or 2;

T is an integer from 1 to 20; and

Q1, Q2, Q3, Q4, Q5 and Q6 are each independently selected from the side chain of an amino acid; or a pharmaceutically acceptable salt thereof. 147. The conjugate of claim 146, wherein Q1, Q2, Q3, Q4, Q5 and Q6 are each independently selected from the side chain of a natural amino acid, or a pharmaceutically acceptable salt thereof. 148. The conjugate of claim 146, wherein at least one of Q1, Q2, Q3, Q4, Q5 and Q6 is selected from the side chain of a non-natural amino acid, or a pharmaceutically acceptable salt thereof. selected from the side chain of a non-natural amino acid, or a pharmaceutically acceptable salt thereof. 150. The conjugate of claim 146, wherein at least three of Q1, Q2, Q3, Q4, Q5 and Q6 are independently selected from the side chain of a non-natural amino acid, or a pharmaceutically acceptable salt thereof. 151. The conjugate of claim 146, wherein at least four of Q1, Q2, Q3, Q4, Q5 and Q6 are independently selected from the side chain of a non-natural amino acid, or a pharmaceutically acceptable salt thereof. 152. The conjugate of claim 146, wherein at least five of Q1, Q2, Q3, Q4, Q5 and Q6 are independently selected from the side chain of a non-natural amino acid, or a pharmaceutically acceptable salt thereof. 153. The conjugate of claim 146, wherein each of Q1, Q2, Q3, Q4, Q5 and Q6 is independently selected from the side chain of a non-natural amino acid, or a pharmaceutically acceptable salt thereof. 154. The conjugate of claim 146, wherein each Q1, Q2, Q3, Q4, Q5 and Q6 is independently selected from the side chain of serine, threonine, cysteine, glycine, proline, alanine, valine, isoleucine, leucine, methionine, phenylalanine, tyrosine, and tryptophan, or a pharmaceutically acceptable salt thereof. 155. The conjugate of claim 146, wherein each of Q1, Q2, Q3, Q4, Q5 and Q6 is independently selected from C1-C4 alkyl, C1-C2 hydroxyalkyl, C1-C5 alkamino, and C6-C35 alkaryl, or a pharmaceutically acceptable salt thereof. 156. The conjugate of claim 146, wherein each of Q1, Q2, Q3, Q4, Q5 and Q6 is independently selected from 2-methyl-1-propyl, 2-propyl, 1-hydroxyethyl, butyl, benzyl, hydroxymethyl, propyl, 2-butyl, methyl, and 2-aminoethyl, or a pharmaceutically acceptable salt thereof.

one of

or a pharmaceutically acceptable salt thereof. 158. The conjugate of any one of claims 146-157, wherein when n is 2, E dimerizes to form an Fc domain, or a pharmaceutically acceptable salt thereof.

,

(D-XVI)

wherein:

each A1 and A2 is an independently selected amino acid;

each L is a linker that, when each m is 1, 2, 3, 4, or 5, is bound to a nitrogen atom in any A1 and a nitrogen atom in any A2 and covalently linked to E;

each E is an Fc domain monomer;

each m is independently 0, 1, 2, 3, 4, or 5;

n is 1 or 2;

T is an integer from 1 to 20; and

Q1, Q2, Q3, Q4, Q5 and Q6 are each independently selected from the side chain of an amino acid; or a pharmaceutically acceptable salt thereof. 160. The conjugate of claim 159, wherein Q1, Q2, Q3, Q4, Q5 and Q6 are each independently selected from the side chain of a natural amino acid, or a pharmaceutically acceptable salt thereof. 161. The conjugate of claim 159, wherein at least one of Q1, Q2, Q3, Q4, Q5 and Q6 is selected from the side chain of a non-natural amino acid, or a pharmaceutically acceptable salt thereof. 162. The conjugate of claim 159, wherein at least two of Q1, Q2, Q3, Q4, Q5 and Q6 are independently selected from the side chain of a non-natural amino acid, or a pharmaceutically acceptable salt thereof. 163. The conjugate of claim 159, wherein at least three of Q1, Q2, Q3, Q4, Q5 and Q6 are independently selected from the side chain of a non-natural amino acid, or a pharmaceutically acceptable salt thereof. 164. The conjugate of claim 159, wherein at least four of Q1, Q2, Q3, Q4, Q5 and Q6 are independently selected from the side chain of a non-natural amino acid, or a pharmaceutically acceptable salt thereof. 165. The conjugate of claim 159, wherein at least five of Q1, Q2, Q3, Q4, Q5 and Q6 are independently

166. The conjugate of claim 159, wherein each of Q1, Q2, Q3, Q4, Q5 and Q6 is independently selected from the side chain of a non-natural amino acid, or a pharmaceutically acceptable salt thereof. 167. The conjugate of claim 159, wherein each Q1, Q2, Q3, Q4, Q5 and Q6 is independently selected from the side chain of serine, threonine, cysteine, glycine, proline, alanine, valine, isoleucine, leucine, methionine, phenylalanine, tyrosine, and tryptophan, or a pharmaceutically acceptable salt thereof. 168. The conjugate of claim 159, wherein each of Q1, Q2, Q3, Q4, Q5 and Q6 is independently selected from C1-C4 alkyl, C1-C2 hydroxyalkyl, C1-C5 alkamino, and C6-C35 alkaryl, or a pharmaceutically acceptable salt thereof. 169. The conjugate of claim 159, wherein each of Q1, Q2, Q3, Q4, Q5 and Q6 is independently selected from 2-methyl-1-propyl, 2-propyl, 1-hydroxyethyl, butyl, benzyl, hydroxymethyl, propyl, 2-butyl, methyl, and 2-aminoethyl, or a pharmaceutically acceptable salt thereof.

one of

or a pharmaceutically acceptable salt thereof. 171. The conjugate of any one of claims 159-170, wherein:

each A1 and A2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H- tetrazol-5-yl)alanine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4-diaminobutyric acid, 3- hydroxyproline, threonine, 2-amino-4-phenylbutyric acid, 3-(2-naphthyl)alanine, 2-piperazinecarboxylic acid, 2-aminooctanoic acid, serine, 2-aminohexanoic acid, 4-amino-4-piperidinyl carboxylic acid, methionine, methionine sulfoxide, methionine sulfone, S-methylcysteine, S-ethylcysteine, S- propylhomocysteine, cyclopropylalanine, 3-fluoroalanine, 2-amino-5-methylhexanoic acid, 2-amino-5- methylhex-4-enoic acid, alpha-t-butylglycine, and alpha-neopentylglycine; and

each m is independently selected from 1, 2, 3, 4, or 5;

or a pharmaceutically acceptable salt thereof. pharmaceutically acceptable salt thereof. 173. The conjugate of any one of claims 159-171, wherein the combination of Q1, Q2, Q3, Q4, Q5 and Q6 is selected from one of

or a pharmaceutically acceptable salt thereof. 174. The conjugate of claim 159, wherein:

each A1 and A2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H- tetrazol-5-yl)alanine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4-diaminobutyric acid, 3- hydroxyproline, threonine, 2-amino-4-phenylbutyric acid, 3-(2-naphthyl)alanine, 2-piperazinecarboxylic acid, 2-aminooctanoic acid, serine, 2-aminohexanoic acid, 4-amino-4-piperidinyl carboxylic acid, methionine, methionine sulfoxide, methionine sulfone, S-methylcysteine, S-ethylcysteine, S- propylhomocysteine, cyclopropylalanine, 3-fluoroalanine, 2-amino-5-methylhexanoic acid, 2-amino-5- methylhex-4-enoic acid, alpha-t-butylglycine, and alpha-neopentylglycine;

each m is independently 1, 2, 3 or 4;

the combination of Q1, Q2, Q3, Q4, Q5 and Q6 is selected from one of

or a pharmaceutically acceptable salt thereof. 175. The conjugate of any one of claims 159-174, wherein when n is 2, E dimerizes to form an Fc domain, or a pharmaceutically acceptable salt thereof.

,

(D-XVII)

wherein:

each A1 and A2 is an independently selected amino acid;

each X is absent or is -CH2CH2C(O)NH-;

each E is an Fc domain monomer;

each m is independently 0, 1, 2, 3, 4, or 5;

n is 1 or 2;

Q1, Q2, Q3, Q4, Q5 and Q6 are each independently selected from the side chain of an amino acid; each d is an integer from 0 to 10;

each e is an integer from 0 to 10; and

T is an integer from 1 to 20,

or a pharmaceutically acceptable salt thereof. 177. The conjugate of claim 176, wherein each of Q1, Q2, Q3, Q4, Q5 and Q6 is independently selected from C1-C4 alkyl, C1-C2 hydroxyalkyl, C1-C5 alkamino, and C6-C35 alkaryl, or a pharmaceutically acceptable salt thereof. 178. The conjugate of claim 177, wherein each of Q1, Q2, Q3, Q4, Q5 and Q6 is independently selected from 2-methyl-1-propyl, 2-propyl, 1-hydroxyethyl, butyl, benzyl, hydroxymethyl, propyl, 2-butyl, methyl, and 2-aminoethyl, or a pharmaceutically acceptable salt thereof. one of

180. The conjugate of claim 179, wherein the combination of Q1, Q2, Q3, Q4, Q5 and Q6 is selected from one of

or a pharmaceutically acceptable salt thereof. 181. The conjugate of any one of claims 176-180, wherein:

each A1 and A2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H- t t l5 l) l i 3 i l i i i 2 b li id 24di i b t i id 3 methionine, methionine sulfoxide, methionine sulfone, S-methylcysteine, S-ethylcysteine, S- propylhomocysteine, cyclopropylalanine, 3-fluoroalanine, 2-amino-5-methylhexanoic acid, 2-amino-5- methylhex-4-enoic acid, alpha-t-butylglycine, and alpha-neopentylglycine;

each m is 2 or 3;

each d is an integer from 1 to 10;

each e is an integer from 1 to 10;

or a pharmaceutically acceptable salt thereof. 182. The conjugate of claim 176, wherein:

each A1 and A2 is independently selected from 2,4-diaminobutyric acid and threonine;

each m is 2 or 3;

each d is an integer from 1 to 10; and

each e is an integer from 1 to 10;

or a pharmaceutically acceptable salt thereof. 183. The conjugate of claim 182, wherein each m is 2, or a pharmaceutically acceptable salt thereof. 184. The conjugate of claim 182, wherein each m is 3, or a pharmaceutically acceptable salt thereof. 185. The conjugate of claim 182, wherein:

each d is 10;

each e is 10; and

each X is absent;

or a pharmaceutically acceptable salt thereof. 186. The conjugate of claim 182, wherein:

each m is 2;

each d is 1;

each e is 1; and

each X is -CH2CH2C(O)NH-;

or a pharmaceutically acceptable salt thereof. 187. The conjugate of any one of claims 176-186, wherein when n is 2, E dimerizes to form an Fc domain, or a pharmaceutically acceptable salt thereof. ,

(D-XVIII)

wherein:

each A1 and A2 is an independently selected amino acid;

each E is an Fc domain monomer;

each m is independently 0, 1, 2, 3, 4, or 5;

n is 1 or 2;

Q1, Q2, Q3, Q4, Q5 and Q6 are each independently selected from the side chain of an amino acid; each d is an integer from 0 to 10;

each e is an integer from 0 to 10;

each f is an integer from 0 to 10;

each g is an integer from 0 to 10; and

T is an integer from 1 to 20,

or a pharmaceutically acceptable salt thereof. 189. The conjugate of claim 188, wherein each of Q1, Q2, Q3, Q4, Q5 and Q6 is independently selected from C1-C4 alkyl, C1-C2 hydroxyalkyl, C1-C5 alkamino, and C6-C35 alkaryl, or a pharmaceutically acceptable salt thereof. 190. The conjugate of claim 189, wherein each of Q1, Q2, Q3, Q4, Q5 and Q6 is independently selected from 2-methyl-1-propyl, 2-propyl, 1-hydroxyethyl, butyl, benzyl, hydroxymethyl, propyl, 2-butyl, methyl, and 2-aminoethyl, or a pharmaceutically acceptable salt thereof. one of

or a pharmaceutically acceptable salt thereof. 192. The conjugate of claim 191, wherein the combination of Q1, Q2, Q3, Q4, Q5 and Q6 is selected from one of

or a pharmaceutically acceptable salt thereof. each A1 and A2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H- tetrazol-5-yl)alanine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4-diaminobutyric acid, 3- hydroxyproline, threonine, 2-amino-4-phenylbutyric acid, 3-(2-naphthyl)alanine, 2-piperazinecarboxylic acid, 2-aminooctanoic acid, serine, 2-aminohexanoic acid, 4-amino-4-piperidinyl carboxylic acid, methionine, methionine sulfoxide, methionine sulfone, S-methylcysteine, S-ethylcysteine, S- propylhomocysteine, cyclopropylalanine, 3-fluoroalanine, 2-amino-5-methylhexanoic acid, 2-amino-5- methylhex-4-enoic acid, alpha-t-butylglycine, and alpha-neopentylglycine;

each m is 2;

each d is 1;

each e is 1;

each f is 3; and

each g is 1;

or a pharmaceutically acceptable salt thereof. 194. The conjugate of claim 188, wherein:

each A1 and A2 is independently selected from 2,4-diaminobutyric acid and threonine;

each m is 2;

each d is 1;

each e is 1;

each f is 3; and

each g is 1;

or a pharmaceutically acceptable salt thereof. 195. The conjugate of any one of claims 188-194, wherein when n is 2, E dimerizes to form an Fc domain, or a pharmaceutically acceptable salt thereof.

,

(D-XIX)

wherein:

each A1 and A2 is an independently selected amino acid;

each E is an Fc domain monomer;

each m is independently 0, 1, 2, 3, 4, or 5;

n is 1 or 2;

Q1, Q2, Q3, Q4, Q5 and Q6 are each independently selected from the side chain of an amino acid; each d is an integer from 0 to 10;

each e is an integer from 0 to 10;

each f is an integer from 0 to 10;

each g is an integer from 0 to 25; and

T is an integer from 1 to 20,

or a pharmaceutically acceptable salt thereof. 197. The conjugate of claim 196, wherein each of Q1, Q2, Q3, Q4, Q5 and Q6 is independently selected from C1-C4 alkyl, C1-C2 hydroxyalkyl, C1-C5 alkamino, and C6-C35 alkaryl, or a pharmaceutically acceptable salt thereof. 198. The conjugate of claim 197, wherein each of Q1, Q2, Q3, Q4, Q5 and Q6 is independently selected from 2-methyl-1-propyl, 2-propyl, 1-hydroxyethyl, butyl, benzyl, hydroxymethyl, propyl, 2-butyl, methyl, and 2-aminoethyl, or a pharmaceutically acceptable salt thereof. one of

or a pharmaceutically acceptable salt thereof. 200. The conjugate of claim 199, wherein the combination of Q1, Q2, Q3, Q4, Q5 and Q6 is selected from one of

or a pharmaceutically acceptable salt thereof. each A1 and A2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H- tetrazol-5-yl)alanine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4-diaminobutyric acid, 3- hydroxyproline, threonine, 2-amino-4-phenylbutyric acid, 3-(2-naphthyl)alanine, 2-piperazinecarboxylic acid, 2-aminooctanoic acid, serine, 2-aminohexanoic acid, 4-amino-4-piperidinyl carboxylic acid, methionine, methionine sulfoxide, methionine sulfone, S-methylcysteine, S-ethylcysteine, S- propylhomocysteine, cyclopropylalanine, 3-fluoroalanine, 2-amino-5-methylhexanoic acid, 2-amino-5- methylhex-4-enoic acid, alpha-t-butylglycine, and alpha-neopentylglycine;

each m is 2;

each d is 1;

each e is 1;

each f is 1; and

each g is 8 to 25;

or a pharmaceutically acceptable salt thereof. 202. The conjugate of claim 196, wherein:

each A1 and A2 is independently selected from 2,4-diaminobutyric acid and threonine;

each m is 2;

each d is 1;

each e is 1;

each f is 1; and

each g is 8 to 25;

or a pharmaceutically acceptable salt thereof. 203. The conjugate of any one of claims 196-202, wherein when n is 2, E dimerizes to form an Fc domain, or a pharmaceutically acceptable salt thereof.

,

(D-XX)

wherein:

each A1 and A2 is an independently selected amino acid;

each E is an Fc domain monomer;

each m is independently 0, 1, 2, 3, 4, or 5;

n is independently 1 or 2;

Q1, Q2, Q3, Q4, Q5 and Q6 are each independently selected from the side chain of an amino acid; each d is an integer from 0 to 15; and

T is an integer from 1 to 20,

or a pharmaceutically acceptable salt thereof. 205. The conjugate of claim 204, wherein each of Q1, Q2, Q3, Q4, Q5 and Q6 is independently selected from C1-C4 alkyl, C1-C2 hydroxyalkyl, C1-C5 alkamino, and C6-C35 alkaryl, or a pharmaceutically acceptable salt thereof. 206. The conjugate of claim 205, wherein each of Q1, Q2, Q3, Q4, Q5 and Q6 is independently selected from 2-methyl-1-propyl, 2-propyl, 1-hydroxyethyl, butyl, benzyl, hydroxymethyl, propyl, 2-butyl, methyl, and 2-aminoethyl, or a pharmaceutically acceptable salt thereof.

one of

or a pharmaceutically acceptable salt thereof. 208. The conjugate of any one of claim 202, wherein the combination of Q1, Q2, Q3, Q4, Q5 and Q6 is selected from one of

or a pharmaceutically acceptable salt thereof. each A1 and A2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H- tetrazol-5-yl)alanine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4-diaminobutyric acid, 3- hydroxyproline, threonine, 2-amino-4-phenylbutyric acid, 3-(2-naphthyl)alanine, 2-piperazinecarboxylic acid, 2-aminooctanoic acid, serine, 2-aminohexanoic acid, 4-amino-4-piperidinyl carboxylic acid, methionine, methionine sulfoxide, methionine sulfone, S-methylcysteine, S-ethylcysteine, S- propylhomocysteine, cyclopropylalanine, 3-fluoroalanine, 2-amino-5-methylhexanoic acid, 2-amino-5- methylhex-4-enoic acid, alpha-t-butylglycine, and alpha-neopentylglycine;

each m is 2; and

each d is 10;

or a pharmaceutically acceptable salt thereof. 210. The conjugate of claim 204, wherein:

each A1 and A2 is independently selected from 2,4-diaminobutyric acid and piperazine-2- carboxylic acid;

each m is 2; and

each d is 10;

or a pharmaceutically acceptable salt thereof. 211. The conjugate of any one of claims 204-210, wherein when n is 2, E dimerizes to form an Fc domain, or a pharmaceutically acceptable salt thereof.

(D-XXI)

wherein:

each A1 and A2 is an independently selected amino acid;

each E is an Fc domain monomer ;

each Y is , -C(O)CH2CH2-, -CH2-, or is absent;

each X is -C(O)CH2CH2- or is absent;

each m is independently 0, 1, 2, 3, 4, or 5;

n is 1 or 2;

Q1, Q2, Q3, Q4, Q5 and Q6 are each independently selected from the side chain of an amino acid; each d is an integer from 0 to 15;

each e is an integer from 1 to 10;

each f is an integer from 1 to 5;

each g is an integer from 1 to 5; and

T is an integer from 1 to 20,

or a pharmaceutically acceptable salt thereof. 213. The conjugate of claim 212, wherein each of Q1, Q2, Q3, Q4, Q5 and Q6 is independently selected from C1-C4 alkyl, C1-C2 hydroxyalkyl, C1-C5 alkamino, and C6-C35 alkaryl, or a pharmaceutically acceptable salt thereof. 214. The conjugate of claim 213, wherein each of Q1, Q2, Q3, Q4, Q5 and Q6 is independently selected from 2-methyl-1-propyl, 2-propyl, 1-hydroxyethyl, butyl, benzyl, hydroxymethyl, propyl, 2-butyl, methyl,

215. The conjugate of claim 214, wherein the combination of Q1, Q2, Q3, Q4, Q5 and Q6 is selected from one of

or a pharmaceutically acceptable salt thereof. 216. The conjugate of claim 215, wherein the combination of Q1, Q2, Q3, Q4, Q5 and Q6 is selected from one of

or a pharmaceutically acceptable salt thereof. each A1 and A2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H- tetrazol-5-yl)alanine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4-diaminobutyric acid, 3- hydroxyproline, threonine, 2-amino-4-phenylbutyric acid, 3-(2-naphthyl)alanine, 2-piperazinecarboxylic acid, 2-aminooctanoic acid, serine, 2-aminohexanoic acid, 4-amino-4-piperidinyl carboxylic acid, methionine, methionine sulfoxide, methionine sulfone, S-methylcysteine, S-ethylcysteine, S- propylhomocysteine, cyclopropylalanine, 3-fluoroalanine, 2-amino-5-methylhexanoic acid, 2-amino-5- methylhex-4-enoic acid, alpha-t-butylglycine, and alpha-neopentylglycine;

each m is 2, 3 or 4;

each d is 1;

each f is 1 or 2; and

each g is 1 or 2;

or a pharmaceutically acceptable salt thereof. 218. The conjugate of claim 214, wherein:

each A1 and A2 is independently selected from 2,4-diaminobutyric acid, 3-aminoalanine, 2- piperazinecarboxylic acid, 2-aminohexanoic acid, 2-aminooctanoic acid, methionine, and threonine; each m is 2, 3 or 4;

each d is 1;

each f is 1 or 2;

each g is 1 or 2;

or a pharmaceutically acceptable salt thereof. 219. The conjugate of any one of claims 214-216, wherein:

each A1 and A2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H- tetrazol-5-yl)alanine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4-diaminobutyric acid, 3- hydroxyproline, threonine, 2-amino-4-phenylbutyric acid, 3-(2-naphthyl)alanine, 2-piperazinecarboxylic acid, 2-aminooctanoic acid, serine, 2-aminohexanoic acid, 4-amino-4-piperidinyl carboxylic acid, methionine, methionine sulfoxide, methionine sulfone, S-methylcysteine, S-ethylcysteine, S- propylhomocysteine, cyclopropylalanine, 3-fluoroalanine, 2-amino-5-methylhexanoic acid, 2-amino-5- methylhex-4-enoic acid, alpha-t-butylglycine, and alpha-neopentylglycine;

each m is 3 or 4;

each d is 1; each g is 1;

or a pharmaceutically acceptable salt thereof. 220. The conjugate of claim 219, wherein

each A1 and A2 is independently selected from 2,4-diaminobutyric acid and threonine; and each m is 3;

or a pharmaceutically acceptable salt thereof. 221. The conjugate of any one of claims 214-216, wherein:

each A1 and A2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H- tetrazol-5-yl)alanine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4-diaminobutyric acid, 3- hydroxyproline, threonine, 2-amino-4-phenylbutyric acid, 3-(2-naphthyl)alanine, 2-piperazinecarboxylic acid, 2-aminooctanoic acid, serine, 2-aminohexanoic acid, 4-amino-4-piperidinyl carboxylic acid, methionine, methionine sulfoxide, methionine sulfone, S-methylcysteine, S-ethylcysteine, S- propylhomocysteine, cyclopropylalanine, 3-fluoroalanine, 2-amino-5-methylhexanoic acid, 2-amino-5- methylhex-4-enoic acid, alpha-t-butylglycine, and alpha-neopentylglycine;

each m is 2, 3 or 4;

each d is 1;

each Y is -C(O)CH2CH2-;

each f is 1 or 2; and

each g is 1 or 2;

or a pharmaceutically acceptable salt thereof. 222. The conjugate of claim 221, wherein each A1 and A2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H-tetrazol-5-yl)alanine, 2,4-diaminobutyric acid, 3-aminoalanine, 2- aminohexanoic acid, 2-piperazinecarboxylic acid, 2-aminooctanoic acid, methionine, methionine sulfoxide, methionine sulfone, S-methylcysteine, S-ethylcysteine, S-propylhomocysteine,

cyclopropylalanine, 3-fluoroalanine, 2-amino-5-methylhexanoic acid, 2-amino-5-methylhex-4-enoic acid, alpha-t-butylglycine, and alpha-neopentylglycine, or a pharmaceutically acceptable salt thereof. 223. The conjugate of claim 222, wherein each m is 4, each d is 1, each f is 1, and each g is 1, or a pharmaceutically acceptable salt thereof. 224. The conjugate of claim 222, wherein each m is 3, each f is 2, and each g is 1, or a pharmaceutically acceptable salt thereof. each A1 and A2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H- tetrazol-5-yl)alanine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4-diaminobutyric acid, 3- hydroxyproline, threonine, 2-amino-4-phenylbutyric acid, 3-(2-naphthyl)alanine, 2-piperazinecarboxylic acid, 2-aminooctanoic acid, serine, 2-aminohexanoic acid, 4-amino-4-piperidinyl carboxylic acid, methionine, methionine sulfoxide, methionine sulfone, S-methylcysteine, S-ethylcysteine, S- propylhomocysteine, cyclopropylalanine, 3-fluoroalanine, 2-amino-5-methylhexanoic acid, 2-amino-5- methylhex-4-enoic acid, alpha-t-butylglycine, and alpha-neopentylglycine;

each m is 2, 3 or 4;

each d is 1;

each Y is -CH2-;

each f is 1 or 2; and

each g is 1 or 2;

or a pharmaceutically acceptable salt thereof. 226. The conjugate of claim 225, wherein:

each A1 and A2 is independently selected from 2,4-diaminobutyric acid, 2-aminooctanoic acid and threonine;

each f is 1; and

each g is 1;

or a pharmaceutically acceptable salt thereof. 227. The conjugate of claim 225, wherein:

each A1 and A2 is independently selected from 2,4-diaminobutyric acid, 2-aminohexanoic acid, 2- aminooctanoic acid and threonine, or a pharmaceutically acceptable salt thereof;

each m is 4;

each f is 1; and

each g is 1;

or a pharmaceutically acceptable salt thereof. 228. The conjugate of claim 227, wherein:

each A1 and A2 is independently selected from 2,4-diaminobutyric acid, 2-aminohexanoic acid, 2- aminooctanoic acid and threonine, or a pharmaceutically acceptable salt thereof;

each m is 3;

each f is 1; and

each g is 1;

or a pharmaceutically acceptable salt thereof.

,

(D-XXII)

wherein:

each A1 and A2 is an independently selected amino acid;

each X is -C(O)CH2CH2CH2-Y- or -C(O)CH2CH2C(O)NH-;

each Y is heteroaryl;

each E is an Fc domain monomer;

each m is independently 0, 1, 2, 3, 4, or 5;

n is 1 or 2;

T is an integer from 1 to 20;

Q1, Q2, Q3, Q4, Q5 and Q6 are each independently selected from the side chain of an amino acid; and

each d is an integer from 0 to 15;

or a pharmaceutically acceptable salt thereof. 230. The conjugate of claim 229, wherein each of Q1, Q2, Q3, Q4, Q5 and Q6 is independently selected from C1-C4 alkyl, C1-C2 hydroxyalkyl, C1-C5 alkamino, and C6-C35 alkaryl, or a pharmaceutically acceptable salt thereof. 231. The conjugate of claim 230, wherein each of Q1, Q2, Q3, Q4, Q5 and Q6 is independently selected from 2-methyl-1-propyl, 2-propyl, 1-hydroxyethyl, butyl, benzyl, hydroxymethyl, propyl, 2-butyl, methyl, and 2-aminoethyl, or a pharmaceutically acceptable salt thereof. one of

233. The conjugate of any one of claim 232, wherein the combination of Q1, Q2, Q3, Q4, Q5 and Q6 is selected from one of

234. The conjugate of any one of claims 229-233, wherein:

each A1 and A2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H- tetrazol-5-yl)alanine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4-diaminobutyric acid, 3- hd li th i 2 i 4 h lb t i id 3(2 hth l) l i 2 i i b li propylhomocysteine, cyclopropylalanine, 3-fluoroalanine, 2-amino-5-methylhexanoic acid, 2-amino-5- methylhex-4-enoic acid, alpha-t-butylglycine, and alpha-neopentylglycine;

each m is 2 or 3;

each d is an integer from 1 to 3;

or a pharmaceutically acceptable salt thereof. 235. The conjugate of claim 234, wherein each A1 and A2 is independently selected from 2,4- diaminobutyric acid, 3-(2-naphthyl)alanine, and threonine, or a pharmaceutically acceptable salt thereof. 236. The conjugate of claim 235, wherein:

each X is -C(O)CH2CH2CH2-Y-;

each m is 3;

each d is 3; and

each Y is 1,4-triazololyl;

or a pharmaceutically acceptable salt thereof. 237. The conjugate of claim 235, wherein:

each X is -C(O)CH2CH2C(O)-;

each m is 2; and

each d is 1;

or a pharmaceutically acceptable salt thereof. 238. The conjugate of claim 236, wherein when n is 2, E dimerizes to form an Fc domain, or a pharmaceutically acceptable salt thereof. 239. The conjugate of any one of claims 229-237, wherein when n is 2, E dimerizes to form an Fc domain, or a pharmaceutically acceptable salt thereof.

,

(D-XXIII)

wherein:

each A1 and A2 is an independently selected amino acid;

each E is an Fc domain monomer;

each R is C1-C20 alkyl;

each m is independently 0, 1, 2, 3, 4, or 5;

n is 1 or 2;

Q1, Q2, Q3, Q4, Q5 and Q6 are each independently selected from the side chain of an amino acid; each d is an integer from 0 to 20;

each e is an integer from 0 to 20;

each f is an integer from 0 to 20; and

T is an integer from 1 to 20,

or a pharmaceutically acceptable salt thereof. 241. The conjugate of claim 240, wherein each of Q1, Q2, Q3, Q4, Q5 and Q6 is independently selected from C1-C4 alkyl, C1-C2 hydroxyalkyl, C1-C5 alkamino, and C6-C35 alkaryl, or a pharmaceutically acceptable salt thereof. 242. The conjugate of claim 226, wherein each of Q1, Q2, Q3, Q4, Q5 and Q6 is independently selected from 2-methyl-1-propyl, 2-propyl, 1-hydroxyethyl, butyl, benzyl, hydroxymethyl, propyl, 2-butyl, methyl, and 2-aminoethyl, or a pharmaceutically acceptable salt thereof. one of

or a pharmaceutically acceptable salt thereof. 244. The conjugate of claim 243,, wherein the combination of Q1, Q2, Q3, Q4, Q5 and Q6 is selected from one of

or a pharmaceutically acceptable salt thereof. 245. The conjugate of any one of claims 240-244, wherein:

h A1 d A2 i i d d tl l t d f l i i i i l t i 3(2H acid, 2-aminooctanoic acid, serine, 2-aminohexanoic acid, 4-amino-4-piperidinyl carboxylic acid, methionine, methionine sulfoxide, methionine sulfone, S-methylcysteine, S-ethylcysteine, S- propylhomocysteine, cyclopropylalanine, 3-fluoroalanine, 2-amino-5-methylhexanoic acid, 2-amino-5- methylhex-4-enoic acid, alpha-t-butylglycine, and alpha-neopentylglycine;

each m is 2 or 3;

each d is an integer from 1 to 3;

each e is an integer from 1 to 3;

each f is an integer from 1 to 3; and

each R is C1-C10 alkyl;

or a pharmaceutically acceptable salt thereof. 246. The conjugate of claim 240, wherein:

each A1 and A2 is independently selected from 2,4-diaminobutyric acid and threonine;

each m is 2;

each d is 1;

each e is 1;

each f is 1;

each R is C1-C10 alkyl; and

or a pharmaceutically acceptable salt thereof. 247. The conjugate of any one of claims 240-246, wherein when n is 2, E dimerizes to form an Fc domain, or a pharmaceutically acceptable salt thereof.

,

(D-XXIV)

wherein:

each A1 and A2 is an independently selected amino acid;

each E is an Fc domain monomer;

each R is C1-C20 alkyl;

each m is independently 0, 1, 2, 3, 4, or 5;

n is 1 or 2;

Q1, Q2, Q3, Q4, Q5 and Q6 are each independently selected from the side chain of an amino acid; each d is an integer from 1 to 20;

each e is an integer from 1 to 20;

each f is an integer from 1 to 20; and

T is an integer from 1 to 20,

or a pharmaceutically acceptable salt thereof. 249. The conjugate of claim 238, wherein each of Q1, Q2, Q3, Q4, Q5 and Q6 is independently selected from C1-C4 alkyl, C1-C2 hydroxyalkyl, C1-C5 alkamino, and C6-C35 alkaryl, or a pharmaceutically acceptable salt thereof. 250. The conjugate of claim 2, wherein each of Q1, Q2, Q3, Q4, Q5 and Q6 is independently selected from 2-methyl-1-propyl, 2-propyl, 1-hydroxyethyl, butyl, benzyl, hydroxymethyl, propyl, 2-butyl, methyl, and 2- aminoethyl, or a pharmaceutically acceptable salt thereof. one of

or a pharmaceutically acceptable salt thereof. 252. The conjugate of any one of claim 248-251, wherein the combination of Q1, Q2, Q3, Q4, Q5 and Q6 is selected from one of

or a pharmaceutically acceptable salt thereof. 253. The conjugate of any one of claims 248-252, wherein:

h A1 d A2 i i d d tl l t d f l i i i i l t i 3(2H acid, 2-aminooctanoic acid, serine, 2-aminohexanoic acid, 4-amino-4-piperidinyl carboxylic acid, methionine, methionine sulfoxide, methionine sulfone, S-methylcysteine, S-ethylcysteine, S- propylhomocysteine, cyclopropylalanine, 3-fluoroalanine, 2-amino-5-methylhexanoic acid, 2-amino-5- methylhex-4-enoic acid, alpha-t-butylglycine, and alpha-neopentylglycine;

each m is 2, 3, or 4;

each d is an integer from 1 to 3;

each e is an integer from 1 to 3;

each f is an integer from 1 to 3;

each R is C1-C10 alkyl; and

or a pharmaceutically acceptable salt thereof. 254. The conjugate of claim 248, wherein:

each A1 and A2 is independently selected from 2,4-diaminobutyric acid, 2-aminooctanoic acid, and threonine;

each m is 2, 3, or 4;

each d is 1;

each e is 1;

each f is 1; and

each R is C1-C10 alkyl;

or a pharmaceutically acceptable salt thereof. 255. The conjugate of claim 254, wherein each m is 2, or a pharmaceutically acceptable salt thereof. 256. The conjugate of claim 254, wherein each m is 3, or a pharmaceutically acceptable salt thereof. 257. The conjugate of claim 254, wherein each m is 4, or a pharmaceutically acceptable salt thereof. 258. The conjugate of any one of claims 248-257, wherein when n is 2, E dimerizes to form an Fc domain, or a pharmaceutically acceptable salt thereof.

,

(D-XXV)

wherein:

each A1 and A2 is an independently selected amino acid;

each E is an Fc domain monomer;

each Y is , -C(O)CH2CH2-, -CH2-, or is absent;

each e is an integer from 1 to 10;

each m is independently 0, 1, 2, 3, 4, or 5;

n is 1 or 2;

Q1, Q2, Q3, Q4, Q5 and Q6 are each independently selected from the side chain of an amino acid; each d is an integer from 0 to 15; and

T is an integer from 1 to 20,

or a pharmaceutically acceptable salt thereof. 260. The conjugate of claim 259, wherein each of Q1, Q2, Q3, Q4, Q5 and Q6 is independently selected from C1-C4 alkyl, C1-C2 hydroxyalkyl, C1-C5 alkamino, and C6-C35 alkaryl, or a pharmaceutically acceptable salt thereof. 261. The conjugate of claim 260, wherein each of Q1, Q2, Q3, Q4, Q5 and Q6 is independently selected from 2-methyl-1-propyl, 2-propyl, 1-hydroxyethyl, butyl, benzyl, hydroxymethyl, propyl, 2-butyl, methyl, and 2-aminoethyl, or a pharmaceutically acceptable salt thereof. one of

or a pharmaceutically acceptable salt thereof. 263. The conjugate of claim 262, wherein the combination of Q1, Q2, Q3, Q4, Q5 and Q6 is selected from one of

or a pharmaceutically acceptable salt thereof. 264. The conjugate of any one of claims 259-263, wherein each Y is–C(O)CH2CH2-, or a

h ti ll t bl lt th f each A1 and A2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H- tetrazol-5-yl)alanine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4-diaminobutyric acid, 3- hydroxyproline, threonine, 2-amino-4-phenylbutyric acid, 3-(2-naphthyl)alanine, 2-piperazinecarboxylic acid, 2-aminooctanoic acid, serine, 2-aminohexanoic acid, 4-amino-4-piperidinyl carboxylic acid, methionine, methionine sulfoxide, methionine sulfone, S-methylcysteine, S-ethylcysteine, S- propylhomocysteine, cyclopropylalanine, 3-fluoroalanine, 2-amino-5-methylhexanoic acid, 2-amino-5- methylhex-4-enoic acid, alpha-t-butylglycine, and alpha-neopentylglycine;

each m is 2, 3, or 4; and

each d is an integer from 1 to 3;

or a pharmaceutically acceptable salt thereof. 266. The conjugate of claim 265, wherein:

each A1 and A2 is independently selected from 2,4-diaminobutyric acid, 2-aminooctanoic acid, and threonine; and

each d is 1;

or a pharmaceutically acceptable salt thereof. 267. The conjugate of claim 266, wherein each m is 2, or a pharmaceutically acceptable salt thereof. 268. The conjugate of claim 266, wherein each m is 3, or a pharmaceutically acceptable salt thereof. 269. The conjugate of claim 266, wherein each m is 4, or a pharmaceutically acceptable salt thereof. 270. The conjugate of any one of claims 259-269, wherein when n is 2, E dimerizes to form an Fc domain, or a pharmaceutically acceptable salt thereof. 271. A conjugate of formula (D-XXVI):

(D-XXVI)

wherein:

each A1 and A2 is an independently selected amino acid; n is 1 or 2;

Q1, Q2, Q3, Q4, Q5 and Q6 are each independently selected from the side chain of an amino acid; each d is an integer from 0 to 15; and

T is an integer from 1 to 20,

or a pharmaceutically acceptable salt thereof. 272. The conjugate of claim 271, wherein each of Q1, Q2, Q3, Q4, Q5 and Q6 is independently selected from C1-C4 alkyl, C1-C2 hydroxyalkyl, C1-C5 alkamino, and C6-C35 alkaryl, or a pharmaceutically acceptable salt thereof. 273. The conjugate of claim 272, wherein each of Q1, Q2, Q3, Q4, Q5 and Q6 is independently selected from 2-methyl-1-propyl, 2-propyl, 1-hydroxyethyl, butyl, benzyl, hydroxymethyl, propyl, 2-butyl, methyl, and 2-aminoethyl, or a pharmaceutically acceptable salt thereof. 274. The conjugate of claim 273, wherein the combination of Q1, Q2, Q3, Q4, Q5 and Q6 is selected from one of

275. The conjugate of claim 274, wherein the combination of Q1, Q2, Q3, Q4, Q5 and Q6 is selected from one of

or a pharmaceutically acceptable salt thereof. 276. The conjugate of any one of claims 271-275, wherein:

each A1 and A2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H- tetrazol-5-yl)alanine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4-diaminobutyric acid, 3- hydroxyproline, threonine, 2-amino-4-phenylbutyric acid, 3-(2-naphthyl)alanine, 2-piperazinecarboxylic acid, 2-aminooctanoic acid, serine, 2-aminohexanoic acid, 4-amino-4-piperidinyl carboxylic acid, methionine, methionine sulfoxide, methionine sulfone, S-methylcysteine, S-ethylcysteine, S- propylhomocysteine, cyclopropylalanine, 3-fluoroalanine, 2-amino-5-methylhexanoic acid, 2-amino-5- methylhex-4-enoic acid, alpha-t-butylglycine, and alpha-neopentylglycine;

each m is 2, 3, or 4; and

each d is an integer from 1 to 3;

or a pharmaceutically acceptable salt thereof. 277. The conjugate of claim 276, wherein:

each A1 and A2 is independently selected from 2,4-diaminobutyric acid, 2-aminooctanoic acid, and threonine; and

each d is 1;

or a pharmaceutically acceptable salt thereof. 278. The conjugate of claim 277, wherein each m is 2, or a pharmaceutically acceptable salt thereof. 279. The conjugate of claim 277, wherein each m is 3, or a pharmaceutically acceptable salt thereof. 280. The conjugate of claim 277, wherein each m is 4, or a pharmaceutically acceptable salt thereof. 281. The conjugate of any one of claims 271-280, wherein when n is 2, E dimerizes to form an Fc domain, or a pharmaceutically acceptable salt thereof.

,

(D-XXVII)

wherein:

each A1 and A2 is an independently selected amino acid;

each E is an Fc domain monomer;

each Y is , -C(O)CH2CH2-, -CH2-, or is absent;

each m is independently 0, 1, 2, 3, 4, or 5;

n is 1 or 2;

Q1, Q2, Q3, Q4, Q5 and Q6 are each independently selected from the side chain of an amino acid; each d is an integer from 0 to 15;

each e is an integer from 1 to 3; and

T is an integer from 1 to 20,

or a pharmaceutically acceptable salt thereof. 283. The conjugate of claim 281, wherein each of Q1, Q2, Q3, Q4, Q5 and Q6 is independently selected from C1-C4 alkyl, C1-C2 hydroxyalkyl, C1-C5 alkamino, and C6-C35 alkaryl, or a pharmaceutically acceptable salt thereof. 284. The conjugate of claim 283, wherein each of Q1, Q2, Q3, Q4, Q5 and Q6 is independently selected from 2-methyl-1-propyl, 2-propyl, 1-hydroxyethyl, butyl, benzyl, hydroxymethyl, propyl, 2-butyl, methyl, and 2-aminoethyl, or a pharmaceutically acceptable salt thereof. one of

or a pharmaceutically acceptable salt thereof. 286. The conjugate of claim 285, wherein the combination of Q1, Q2, Q3, Q4, Q5 and Q6 is selected from one of

or a pharmaceutically acceptable salt thereof. 287. The conjugate of any one of claims 282-286, wherein each Y is -C(O)CH2CH2-, or a

h ti ll t bl lt th f each A1 and A2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H- tetrazol-5-yl)alanine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4-diaminobutyric acid, 3- hydroxyproline, threonine, 2-amino-4-phenylbutyric acid, 3-(2-naphthyl)alanine, 2-piperazinecarboxylic acid, 2-aminooctanoic acid, serine, 2-aminohexanoic acid, 4-amino-4-piperidinyl carboxylic acid, methionine, methionine sulfoxide, methionine sulfone, S-methylcysteine, S-ethylcysteine, S- propylhomocysteine, cyclopropylalanine, 3-fluoroalanine, 2-amino-5-methylhexanoic acid, 2-amino-5- methylhex-4-enoic acid, alpha-t-butylglycine, and alpha-neopentylglycine;

each m is 2, 3, or 4;

each d is an integer from 1 to 3;

or a pharmaceutically acceptable salt thereof. 289. The conjugate of claim 288, wherein:

each A1 and A2 is independently selected from 2,4-diaminobutyric acid, 2-aminooctanoic acid, and threonine; and

each d is 1; and

each e is 1 or 2;

or a pharmaceutically acceptable salt thereof. 290. The conjugate of claim 289, wherein each m is 2, or a pharmaceutically acceptable salt thereof. 291. The conjugate of claim 289, wherein each m is 3, or a pharmaceutically acceptable salt thereof. 292. The conjugate of claim 289, wherein each m is 4, or a pharmaceutically acceptable salt thereof. 293. The conjugate of any one of claims 282-292, wherein when n is 2, E dimerizes to form an Fc domain, or a pharmaceutically acceptable salt thereof.

,

(D-XXVIII)

wherein:

each A1 and A2 is an independently selected amino acid;

each E is an Fc domain monomer;

each X is independently selected from

, -C(O)CH2CH2C(O)-, -CH2CH2NHC(O)CH2CH2-, -C(O)-, and -CH2-, or is absent;

each m is independently 0, 1, 2, 3, 4, or 5;

n is 1 or 2;

Q1, Q2, Q3, Q4, Q5 and Q6 are each independently selected from the side chain of an amino acid; each d is an integer from 1 to 15;

each g is an integer from 1 to 15;

each e is independently from 1 to 15; and

T is an integer from 1 to 20,

or a pharmaceutically acceptable salt thereof.

(D-XXVIII-1)

or a pharmaceutically acceptable salt thereof. 296. The conjugate of claim 294, wherein the conjugate is of the formula (D-XXVIII-2):

(D-XXVIII-2)

or a pharmaceutically acceptable salt thereof. 297. The conjugate of any one of claims 294-296, wherein each of Q1, Q2, Q3, Q4, Q5 and Q6 is independently selected from C1-C4 alkyl, C1-C2 hydroxyalkyl, C1-C5 alkamino, and C6-C35 alkaryl, or a pharmaceutically acceptable salt thereof. 298. The conjugate of claim 297, wherein each of Q1, Q2, Q3, Q4, Q5 and Q6 is independently selected from 2-methyl-1-propyl, 2-propyl, 1-hydroxyethyl, butyl, benzyl, hydroxymethyl, propyl, 2-butyl, methyl, and 2-aminoethyl, or a pharmaceutically acceptable salt thereof.

299. The conjugate of claim 298, wherein the combination of Q1, Q2, Q3, Q4, Q5 and Q6 is selected from one of

or a pharmaceutically acceptable salt thereof. one of

or a pharmaceutically acceptable salt thereof. 301. The conjugate of any one of claims 294-300, wherein:

each A1 and A2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H- tetrazol-5-yl)alanine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4-diaminobutyric acid, 3- hydroxyproline, threonine, 2-amino-4-phenylbutyric acid, 3-(2-naphthyl)alanine, 2-piperazinecarboxylic acid, 2-aminooctanoic acid, serine, 2-aminohexanoic acid, 4-amino-4-piperidinyl carboxylic acid, methionine, methionine sulfoxide, methionine sulfone, S-methylcysteine, S-ethylcysteine, S- propylhomocysteine, cyclopropylalanine, 3-fluoroalanine, 2-amino-5-methylhexanoic acid, 2-amino-5- methylhex-4-enoic acid, alpha-t-butylglycine, and alpha-neopentylglycine;

each m is 2, 3, or 4;

each d is an integer from 1 to 5;

each e is 1;

each X is -C(O)CH2CH2C(O)-;

or a pharmaceutically acceptable salt thereof. 302. The conjugate of claim 301, wherein:

each A1 and A2 is independently selected from 2,4-diaminobutyric acid, 2-aminohexanoic acid, and threonine; and

each d is 1;

or a pharmaceutically acceptable salt thereof. 303. The conjugate of claim 302, wherein each m is 2, or a pharmaceutically acceptable salt thereof. 304. The conjugate of claim 302, wherein each m is 3, or a pharmaceutically acceptable salt thereof. 305. The conjugate of claim 302, wherein each m is 4, or a pharmaceutically acceptable salt thereof. 306. The conjugate of any one of claims 294-305, wherein when n is 2, E dimerizes to form an Fc domain, or a pharmaceutically acceptable salt thereof.

wherein each M comprises a cyclic heptapeptide comprising a linking nitrogen atom;

each E is an Fc domain monomer;

L’ in each L’-M is a linker, wherein L’ is covalently attached to the linking nitrogen atom in M and to E;

n is 1 or 2; and

T is an integer from1 to 20;

or a pharmaceutically acceptable salt thereof. 308. The conjugate of claim 307, wherein when n is 2, E dimerizes to form an Fc domain, and the conjugate is described by formula (M-Ib):

wherein J is an Fc domain; and

T is an integer from 2 to 20,

or a pharmaceutically acceptable salt thereof. 309. The conjugate of claim 307 or 308, wherein L’ in each L’-M is described by formula (M-L’):

wherein L is a remainder of L’; and

A is 1 to 5 amino acids, wherein at least one A is covalently attached to the linking nitrogen atom in M, or a pharmaceutically acceptable salt thereof.

(M-II)

wherein each L is a remainder of L’ and is covalently attached to or may take the place of R2, R3, R4, R5, R6, R7, R8, R9, R10, or R16;

each of R1 and R12 is, independently, a lipophilic moiety, a polar moiety, or H;

each of R11, R13, and R14 is, independently, optionally substituted C1-C5 alkamino, a polar moiety, a positively charged moiety, or H;

each R15 is a lipophilic moiety or a polar moiety;

each of R2, R3, R4, R5, R6, R7, R8, R9, and R10 is, independently, a lipophilic moiety, a positively charged moiety, a polar moiety, H, optionally substituted C1-C5 alkamino, optionally substituted C1-C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1-C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20

heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl; or two R groups on the same or adjacent atoms join to form an optionally substituted 5-8 membered ring;

each R16 is H, optionally substituted C1-C5 alkamino, optionally substituted C1-C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1-C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20

heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl, or is absent; and

each of a, b, and c is, independently, 0 or 1;

or a pharmaceutically acceptable salt thereof. 311. The conjugate of claim 310, wherein the conjugate comprises at least one optionally substituted 5-8 membered ring formed by joining (i) R2, R3, and C1; (ii) R3, R4, N1, and C1; (iii) R5, R6, and C2; (iv) R6, R7,

312. The con f n n f l im 7- 11 wh r in h n i ri f rmula (M-III):

or a pharmaceutically acceptable salt thereof. 313. The conjugate of any one of claims 310-312, wherein

each of R1 and R12 is a lipophilic moiety;

each of R11, R13, and R14 is, independently, optionally substituted C1-C5 alkamino, a polar moiety, or a positively charged moiety; and

each R15 is a polar moiety;

or a pharmaceutically acceptable salt thereof. 314. The conjugate of claim 313, wherein each lipophilic moiety is independently selected from optionally substituted C1-C20 alkyl, optionally substituted C5-C15 aryl, optionally substituted C6-C35 alkaryl, or optionally C5-C10 substituted heteroaryl, or a pharmaceutically acceptable salt thereof. 315. The conjugate of claim 313 or 314, wherein each lipophilic moiety is, independently, C1-C8 alkyl, methyl substituted C2-C4 alkyl, (C1-C10)alkylene(C6)aryl, phenyl substituted (C1-C10)alkylene(C6)aryl, or alkyl substituted C4-C9 heteroaryl, or a pharmaceutically acceptable salt thereof. 316. The conjugate of claim 314 or 315, wherein each lipophilic moiety is, independently, benzyl, isobutyl, sec-butyl, isopropyl, n-propyl, methyl, biphenylmethyl, n-octyl, or methyl substituted indolyl, or a pharmaceutically acceptable salt thereof. 317. The conjugate of any one of claims 310-316, wherein each of R11, R13, and R14 is independently optionally substituted C1-C5 alkamino, or a pharmaceutically acceptable salt thereof. acceptable salt thereof. 319. The conjugate of any one of claims 310-318, wherein each R15 is a polar moiety, or a pharmaceutically acceptable salt thereof. 320. The conjugate of claim 319, wherein the polar moiety comprises C1-C4 hydroxyalkyl, a carboxylic acid group, an ester group, or an amide group, or a pharmaceutically acceptable salt thereof. 321. The conjugate of claim 319 or 320, wherein the polar moiety is C1-C4 hydroxyalkyl, or a pharmaceutically acceptable salt thereof. 322. The conjugate of any one of claims 320 or 321, wherein the polar moiety is CHCH3OH, or a pharmaceutically acceptable salt thereof. 323. The conjugate of claim 310 wherein the con u ate is described b formula M-IV :

wherein each R1 is benzyl or CH2CH(CH3)2,

or a pharmaceutically acceptable salt thereof.

,

(M-IV-1)

or a pharmaceutically acceptable salt thereof. 325. The con f l im 24 wh r in h n i ri f rm l M-IV-2:

,

(M-IV-2)

or a pharmaceutically acceptable salt thereof.

,

(M-V)

wherein each R1 is benzyl or CH2CH(CH3)2; and

each R2 is a positively charged moiety, a polar moiety, optionally substituted C1-C5 alkamino, optionally substituted C1-C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4- C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1-C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl;

or a pharmaceutically acceptable salt thereof. 327. The conjugate of claim 326 wherein the con u ate is described b formula (M-V-1):

,

(M-V-1)

or a pharmaceutically acceptable salt thereof.

,

(M-VI)

wherein each R1 is benzyl or CH2CH(CH3)2;

each R2 is optionally substituted C1-C20 alkylene, optionally substituted C1-C20 heteroalkylene, optionally substituted C2-C20 alkenylene, optionally substituted C2-C20 heteroalkenylene, optionally substituted C2-C20 alkynylene, optionally substituted C2-C20 heteroalkynylene, optionally substituted C3-C20 cycloalkylene, optionally substituted C3-C20 heterocycloalkylene, optionally substituted C4-C20 cycloalkenylene, optionally substituted C4-C20 heterocycloalkenylene, optionally substituted C8-C20 cycloalkynylene, optionally substituted C8-C20 heterocycloalkynylene, optionally substituted C5-C15 arylene, or optionally substituted C2-C15 heteroarylene; and

each R16 is H, optionally substituted C1-C5 alkamino, optionally substituted C1-C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1-C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20

heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl;

or a pharmaceutically acceptable salt thereof. 329. The conjugate of claim 328, wh r in h n i ri formula (M-VI-1):

(M-VI-1)

wherein each R16 is H, optionally substituted C1-C5 alkamino, optionally substituted C1-C20 alkyl, heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl;

or a pharmaceutically acceptable salt thereof. 330. The conjugate of claim 310 wherein the con u ate is described b formula M-VII):

,

(M-VII)

wherein each R1 is benzyl or CH2CH(CH3)2;

each R2 is optionally substituted C1-C20 alkylene, optionally substituted C1-C20 heteroalkylene, optionally substituted C2-C20 alkenylene, optionally substituted C2-C20 heteroalkenylene, optionally substituted C2-C20 alkynylene, optionally substituted C2-C20 heteroalkynylene, optionally substituted C3-C20 cycloalkylene, optionally substituted C3-C20 heterocycloalkylene, optionally substituted C4-C20 cycloalkenylene, optionally substituted C4-C20 heterocycloalkenylene, optionally substituted C8-C20 cycloalkynylene, optionally substituted C8-C20 heterocycloalkynylene, optionally substituted C5-C15 arylene, or optionally substituted C2-C15 heteroarylene;

each R6 is a positively charged moiety, a polar moiety, optionally substituted C1-C5 alkamino, optionally substituted C1-C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4- C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1-C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl; and

each R16 is H, optionally substituted C1-C5 alkamino, optionally substituted C1-C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl; or a pharmaceutically acceptable salt thereof. 331. The conjugate of claim 330, wherein the conjugate is described by formula (M-VII-1):

(M-VII-1)

or a pharmaceutically acceptable salt thereof. 332. The conjugate of claim 331 wherein the con u ate is described b formula (M-VII-2):

(M-VII-2)

wherein each R16 is H, optionally substituted C1-C20 alkyl, or optionally substituted C1-C20 heteroalkyl, or a pharmaceutically acceptable salt thereof.

,

(M-VII-3)

or a pharmaceutically acceptable salt thereof. 334. The conjugate of claim 1 wh r in h n i ri f rm la (M-VIII):

,

(M-VIII)

wherein each R1 is benzyl or CH2CH(CH3)2; and

each R2 is optionally substituted C1-C20 alkylene, optionally substituted C1-C20 heteroalkylene, optionally substituted C2-C20 alkenylene, optionally substituted C2-C20 heteroalkenylene, optionally substituted C2-C20 alkynylene, optionally substituted C2-C20 heteroalkynylene, optionally substituted C3-C20 cycloalkylene, optionally substituted C3-C20 heterocycloalkylene, optionally substituted C4-C20 cycloalkenylene, optionally substituted C4-C20 heterocycloalkenylene, optionally substituted C8-C20 cycloalkynylene, optionally substituted C8-C20 heterocycloalkynylene, optionally substituted C5-C15 arylene, or optionally substituted C2-C15 heteroarylene;

each R6, R7, N2, and C2 together form an optionally substituted 5-8 membered ring comprising optionally substituted C3-C7 heterocycloalkyl comprising a nitrogen atom and additional 0-2 heteroatoms nitrogen atom and additional 0-2 heteroatoms independently selected from N, O, and S; or a pharmaceutically acceptable salt thereof. 335. The conjugate of claim 334 wherein the con u ate is described b formula (M-VIII-1):

,

(M-VIII-1)

or a pharmaceutically acceptable salt thereof. 336. The conjugate of claim 335 wherein the con u ate is described b formula (M-VIII-2):

,

(M-VIII-2)

or a pharmaceutically acceptable salt thereof.

(M-VIII-3)

or a pharmaceutically acceptable salt thereof. 338. The conjugate of claim 335 formula (M-VIII-4):

,

(M-VIII-4)

or a pharmaceutically acceptable salt thereof. 339. The conjugate of claim 338, wherein the conjugate is described by formula (M-VIII-5):

,

(M-VIII-5)

or a pharmaceutically acceptable salt thereof.

,

(M-IX)

wherein each R1 is benzyl or CH2CH(CH3)2;

each R2 is optionally substituted C1-C20 alkylene, optionally substituted C1-C20 heteroalkylene, optionally substituted C2-C20 alkenylene, optionally substituted C2-C20 heteroalkenylene, optionally substituted C2-C20 alkynylene, optionally substituted C2-C20 heteroalkynylene, optionally substituted C3-C20 cycloalkylene, optionally substituted C3-C20 heterocycloalkylene, optionally substituted C4-C20 cycloalkenylene, optionally substituted C4-C20 heterocycloalkenylene, optionally substituted C8-C20 cycloalkynylene, optionally substituted C8-C20 heterocycloalkynylene, optionally substituted C5-C15 arylene, or optionally substituted C2-C15 heteroarylene;

each of R6 and R8 is, independently, a positively charged moiety, a polar moiety, optionally substituted C1-C5 alkamino, optionally substituted C1-C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1-C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl; and

each R16 is H, optionally substituted C1-C5 alkamino, optionally substituted C1-C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1-C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20

heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl;

or a pharmaceutically acceptable salt thereof.

,

(M-IX-1)

or a pharmaceutically acceptable salt thereof. 342. The conjugate of claim 341, wherein the conjugate is described by formula (M-IX-2):

(M-IX-2)

wherein each R16 is H, optionally substituted C1-C20 alkyl, or optionally substituted C1-C20 heteroalkyl;

or a pharmaceutically acceptable salt thereof.

(M-IX-3)

wherein each R16 is H, optionally substituted C1-C20 alkyl, or optionally substituted C1-C20 heteroalkyl;

or a pharmaceutically acceptable salt thereof. 344. The conjugate of claim 310, wherein the conjugate is described by formula (M-X):

,

(M-X)

wherein each R1 is benzyl or CH2CH(CH3)2;

each R2 is optionally substituted C1-C20 alkylene, optionally substituted C1-C20 heteroalkylene, optionally substituted C2-C20 alkenylene, optionally substituted C2-C20 heteroalkenylene, optionally substituted C2-C20 alkynylene, optionally substituted C2-C20 heteroalkynylene, optionally substituted C3-C20 cycloalkylene, optionally substituted C3-C20 heterocycloalkylene, optionally substituted C4-C20 cycloalkenylene, optionally substituted C4-C20 heterocycloalkenylene, optionally substituted C8-C20 cycloalkynylene, optionally substituted C8-C20 heterocycloalkynylene, optionally substituted C5-C15 arylene, or optionally substituted C2-C15 heteroarylene;

each R6, R7, N2, and C2 together form an optionally substituted 5-8 membered ring comprising optionally substituted C3 C7 heterocycloalkyl comprising a nitrogen atom and additional 0 2 heteroatoms nitrogen atom and additional 0-2 heteroatoms independently selected from N, O, and S; each R8 is a positively charged moiety, a polar moiety, optionally substituted C1-C5 alkamino, optionally substituted C1-C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4- C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1-C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl;

each R16 is H, optionally substituted C1-C5 alkamino, optionally substituted C1-C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1-C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20

heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl,

or a pharmaceutically acceptable salt thereof. 345. The conjugate of claim 344, wherein the conjugate is described by formula (M-X-1):

,

(M-X-1)

or a pharmaceutically acceptable salt thereof.

,

(M-XI)

wherein each R1 is benzyl or CH2CH(CH3)2; and

each of R2 and R6 is, independently, a positively charged moiety, a polar moiety, optionally substituted C1-C5 alkamino, optionally substituted C1-C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1-C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl,

or a pharmaceutically acceptable salt thereof. 347. The conjugate of claim 346, wherein the conjugate is described by formula (M-XI-1):

,

(M-XI-1)

or a pharmaceutically acceptable salt thereof.

,

(M-XI-2)

or a pharmaceutically acceptable salt thereof. 349. The conjugate f l im 1 wh r in h n i ri f rm l M-XII):

(M-XII)

wherein each R1 is benzyl or CH2CH(CH3)2;

each R2 is a positively charged moiety, a polar moiety, optionally substituted C1-C5 alkamino, optionally substituted C1-C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4- C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1-C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl; and

each R6, R7, N2, and C2 together form an optionally substituted 5-8 membered ring comprising nitrogen atom and additional 0-2 heteroatoms independently selected from N, O, and S; or a pharmaceutically acceptable salt thereof. 350. The conjugate of claim 310, wherein the conjugate is described by formula (M-XIII):

(M-XIII)

wherein each R1 is benzyl or CH2CH(CH3)2;

each R2 is a positively charged moiety, a polar moiety, optionally substituted C1-C5 alkamino, optionally substituted C1-C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4- C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1-C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl;

each R6 is optionally substituted C1-C20 alkylene, optionally substituted C1-C20 heteroalkylene, optionally substituted C2-C20 alkenylene, optionally substituted C2-C20 heteroalkenylene, optionally substituted C2-C20 alkynylene, optionally substituted C2-C20 heteroalkynylene, optionally substituted C3-C20 cycloalkylene, optionally substituted C3-C20 heterocycloalkylene, optionally substituted C4-C20 cycloalkenylene, optionally substituted C4-C20 heterocycloalkenylene, optionally substituted C8-C20 cycloalkynylene, optionally substituted C8-C20 heterocycloalkynylene, optionally substituted C5-C15 arylene, or optionally substituted C2-C15 heteroarylene; and

each R16 is H, optionally substituted C1-C5 alkamino, optionally substituted C1-C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1-C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20

heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl;

or a pharmaceutically acceptable salt thereof.

,

(M-XIII-1)

wherein each R16 is H or optionally substituted C6-C35 heteroalkaryl;

or a pharmaceutically acceptable salt thereof. 352. The conjugate of claim 310, wherein the conjugate is described by formula (M-XIV):

,

(M-XIV)

wherein each R1 is benzyl or CH2CH(CH3)2;

each R2 is a positively charged moiety, a polar moiety, optionally substituted C1-C5 alkamino, optionally substituted C1-C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4- C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1-C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl;

each R6, R7, N2, and C2 together form an optionally substituted 5-8 membered ring comprising optionally substituted C3-C7 heterocycloalkyl comprising a nitrogen atom and additional 0-2 heteroatoms nitrogen atom and additional 0-2 heteroatoms independently selected from N, O, and S; and each R16 is H, optionally substituted C1-C5 alkamino, optionally substituted C1-C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1-C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl; or a pharmaceutically acceptable salt thereof. 353. The conjugate of claim 352, wherein the conjugate is described by formula (M-XIV-1):

,

(M-XIV-1)

wherein each R16 is H or optionally substituted C6-C35 heteroalkaryl;

or a pharmaceutically acceptable salt thereof. 354. The conjugate of claim 310, wherein the conjugate is described by formula (M-XV):

, substituted C1-C5 alkamino, optionally substituted C1-C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1-C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl;

or a pharmaceutically acceptable salt thereof. 355. The conjugate of claim 354, wherein the conjugate is described by formula (M-XV-1):

,

(M-XV-1)

or a pharmaceutically acceptable salt thereof. 356. The conju ate of claim 355 wherein the con u ate is described b formula M-XV-2):

(M-XV-2)

or a pharmaceutically acceptable salt thereof.

,

(M-XVI)

wherein each R1 is benzyl or CH2CH(CH3)2;

each of R2 and R8 is, independently, a positively charged moiety, a polar moiety, optionally substituted C1-C5 alkamino, optionally substituted C1-C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1-C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl; and

each R6, R7, N2, and C2 together form an optionally substituted 5-8 membered ring comprising optionally substituted C3-C7 heterocycloalkyl comprising a nitrogen atom and additional 0-2 heteroatoms independently selected from N, O, and S, or optionally substituted C2-C7 heteroaryl comprising a nitrogen atom and additional 0-2 heteroatoms independently selected from N, O, and S;

or a pharmaceutically acceptable salt thereof. 358. The conjugate of claim 310 wherein the con u ate is described b formula M-XVII):

wherein each R1 is benzyl or CH2CH(CH3)2;

each of R2 and R6 is, independently, a positively charged moiety, a polar moiety, optionally substituted C1-C5 alkamino, optionally substituted C1-C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1-C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl;

each R8 is optionally substituted C1-C20 alkylene, optionally substituted C1-C20 heteroalkylene, optionally substituted C3-C20 cycloalkylene, optionally substituted C3-C20 heterocycloalkylene, optionally substituted C4-C20 heterocycloalkenylene, optionally substituted C8-C20 heterocycloalkynylene, optionally substituted C5-C15 arylene, or optionally substituted C2-C15 heteroarylene; and

each R16 is H, optionally substituted C1-C5 alkamino, optionally substituted C1-C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1-C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20

heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl,

or a pharmaceutically acceptable salt thereof. 359. The conjugate of claim 358 wherein the con u ate is described b formula M-XVII-1):

,

(M-XVII-1)

or a pharmaceutically acceptable salt thereof.

(M-XVII-2)

or a pharmaceutically acceptable salt thereof. 361. The conjugat f l im wh r in h n i ri f rm l M-XVII-3):

(M-XVII-3)

or a pharmaceutically acceptable salt thereof.

,

(M-XVIII)

wherein each R1 is benzyl or CH2CH(CH3)2;

each R2 is a positively charged moiety, a polar moiety, optionally substituted C1-C5 alkamino, optionally substituted C1-C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4- C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1-C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl;

each R6, R7, N2, and C2 together form an optionally substituted 5-8 membered ring comprising optionally substituted C3-C7 heterocycloalkyl comprising a nitrogen atom and additional 0-2 heteroatoms independently selected from N, O, and S, or optionally substituted C2-C7 heteroaryl comprising a nitrogen atom and additional 0-2 heteroatoms independently selected from N, O, and S;

each R8 is optionally substituted C1-C20 alkylene, optionally substituted C1-C20 heteroalkylene, optionally substituted C2-C20 alkenylene, optionally substituted C2-C20 heteroalkenylene, optionally substituted C2-C20 alkynylene, optionally substituted C2-C20 heteroalkynylene, optionally substituted C3-C20 cycloalkylene, optionally substituted C3-C20 heterocycloalkylene, optionally substituted C4-C20 cycloalkenylene, optionally substituted C4-C20 heterocycloalkenylene, optionally substituted C8-C20 cycloalkynylene, optionally substituted C8-C20 heterocycloalkynylene, optionally substituted C5-C15 arylene, or optionally substituted C2-C15 heteroarylene; and

each R16 is H, optionally substituted C1-C5 alkamino, optionally substituted C1-C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1-C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20

heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15

363. The conjugate of any one of claims 326, 346, 349, 350, 352, 354, 357-359, and 362, wherein each R2 is optionally substituted C1-C5 alkamino or a polar moiety, or a pharmaceutically acceptable salt thereof. 364. The conjugate of claim 363, wherein the optionally substituted C1-C5 alkamino is CH2NH2 or CH2CH2NH2, or a pharmaceutically acceptable salt thereof. 365. The conjugate of any one of claims 330, 331, 340, 341, 346, 354, 357, and 358, wherein each R6 is a polar moiety, or a pharmaceutically acceptable salt thereof. 366. The conjugate of claim 365, wherein the polar moiety comprises C1-C4 hydroxylalkyl group, a carboxylic acid group, an ester group, or an amide group, or a pharmaceutically acceptable salt thereof. 367. The conjugate of claim 365 or 366, wherein the polar moiety is C1-C4 hydroxyalkyl, or a

pharmaceutically acceptable salt thereof. 368. The conjugate of claim 357, wherein the polar moiety is CHCH3OH or CH2OH, or a pharmaceutically acceptable salt thereof. 369. The conjugate of any one of claims 340, 341, 344, 345, 354, and 357, wherein each R8 is optionally substituted C1-C5 alkamino or optionally substituted C5-C15 aryl, or a pharmaceutically acceptable salt thereof. 370. The conjugate of claim 369, wherein each R8 is CH2NH2 or CH2CH2NH2, or a pharmaceutically acceptable salt thereof. 371. The conjugate of claim 369, wherein each R8 is naphthyl, or a pharmaceutically acceptable salt thereof. 372. The conjugate of any one of claims 334, 335, 344, 345, 349, 352, 357, and 362, wherein each R6, R7, N2, and C2 together form a 5- or 6-membered ring comprising C4-C5 heterocycloalkyl comprising a nitrogen atom and additional 0 or 1 heteroatom independently selected from N, O, and S, or a pharmaceutically acceptable salt thereof. 373. The conjugate of any one of claims 307-372, wherein when n is 2, E dimerizes to form an Fc domain. each L’ or L comprises one or more optionally substituted C1-C20 alkylene, optionally substituted C1-C20 heteroalkylene, optionally substituted C2-C20 alkenylene, optionally substituted C2-C20 heteroalkenylene, optionally substituted C2-C20 alkynylene, optionally substituted C2-C20 heteroalkynylene, optionally substituted C3-C20 cycloalkylene, optionally substituted C3-C20 heterocycloalkylene, optionally substituted C4-C20 cycloalkenylene, optionally substituted C4-C20 heterocycloalkenylene, optionally substituted C8-C20 cycloalkynylene, optionally substituted C8-C20 heterocycloalkynylene, optionally substituted C5-C15 arylene, optionally substituted C2-C15 heteroarylene, O, S, NRi, P, carbonyl, thiocarbonyl, sulfonyl, phosphate, phosphoryl, or imino; and

Ri is H, optionally substituted C1-C20 alkyl, optionally substituted C1-C20 heteroalkyl, optionally substituted C2-C20 alkenyl, optionally substituted C2-C20 heteroalkenyl, optionally substituted C2-C20 alkynyl, optionally substituted C2-C20 heteroalkynyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C4- C20 heterocycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C5-C15 aryl, or optionally substituted C2-C15 heteroaryl;

or a pharmaceutically acceptable salt thereof. 375. The conjugate of claims 374, wherein:

the backbone of each L’ or L consists of one or more optionally substituted C1-C20 alkylene, optionally substituted C1-C20 heteroalkylene, optionally substituted C2-C20 alkenylene, optionally substituted C2-C20 heteroalkenylene, optionally substituted C2-C20 alkynylene, optionally substituted C2- C20 heteroalkynylene, optionally substituted C3-C20 cycloalkylene, optionally substituted C3-C20 heterocycloalkylene, optionally substituted C4-C20 cycloalkenylene, optionally substituted C4-C20 heterocycloalkenylene, optionally substituted C8-C20 cycloalkynylene, optionally substituted C8-C20 heterocycloalkynylene, optionally substituted C5-C15 arylene, optionally substituted C2-C15 heteroarylene, O, S, NRi, P, carbonyl, thiocarbonyl, sulfonyl, phosphate, phosphoryl, or imino; and

Ri is H, optionally substituted C1-C20 alkyl, optionally substituted C1-C20 heteroalkyl, optionally substituted C2-C20 alkenyl, optionally substituted C2-C20 heteroalkenyl, optionally substituted C2-C20 alkynyl, optionally substituted C2-C20 heteroalkynyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C4- C20 heterocycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C5-C15 aryl, or optionally substituted C2-C15 heteroaryl;

or a pharmaceutically acceptable salt thereof. 376. The conjugate of 374 or 375, wherein each L’ or L is oxo substituted, or a pharmaceutically acceptable salt thereof. 377. The conjugate of any one of claims 307-376, wherein the backbone of each L’ or L comprises no more than 120 atoms, or a pharmaceutically acceptable salt thereof. carbamate, a sulfonyl, or a urea linkage, or a pharmaceutically acceptable salt thereof. 379. The conjugate of any one of claims 309-378, wherein each L is a bond, or a pharmaceutically acceptable salt thereof. 380. The conjugate of any one of claims 309-379, wherein each L is described by formula (M-L-1):

wherein:

J1 is a bond attached to A or M if A is absent;

J2 is a bond attached to E;

each of Q1, Q2, Q3, Q4, and Q5 is, independently, optionally substituted C1-C20 alkylene, optionally substituted C1-C20 heteroalkylene, optionally substituted C2-C20 alkenylene, optionally substituted C2-C20 heteroalkenylene, optionally substituted C2-C20 alkynylene, optionally substituted C2-C20 heteroalkynylene, optionally substituted C3-C20 cycloalkylene, optionally substituted C3-C20 heterocycloalkylene, optionally substituted C4-C20 cycloalkenylene, optionally substituted C4-C20 heterocycloalkenylene, optionally substituted C8-C20 cycloalkynylene, optionally substituted C8-C20 heterocycloalkynylene, optionally substituted C5-C15 arylene, or optionally substituted C2-C15 heteroarylene;

each of T1, T2, T3, T4 is, independently, O, S, NRi, P, carbonyl, thiocarbonyl, sulfonyl, phosphate, phosphoryl, or imino;

Ri is H, optionally substituted C1-C20 alkyl, optionally substituted C1-C20 heteroalkyl, optionally substituted C2-C20 alkenyl, optionally substituted C2-C20 heteroalkenyl, optionally substituted C2-C20 alkynyl, optionally substituted C2-C20 heteroalkynyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C5-C15 aryl, or optionally substituted C2-C15 heteroaryl; and each of g, h, i, j, k, l, m, n, and o is, independently, 0 or 1;

or a pharmaceutically acceptable salt thereof.

wherein each of d and e is, independently, an integer from 1 to 26; or a pharmaceutically acceptable salt thereof.

,

(M-XIX)

wherein:

each A is an independently selected amino acid;

each L is a linker that, when each m is 2, 3, 4, or 5, is bound to any of A;

each E is an Fc domain monomer;

each m is 0, 1, 2, 3, 4, or 5;

n is 1 or 2;

Tis an integer from 1 to 20; and

Q1, Q2, and Q3 are each independently selected from the side chain of an amino acid;

or a pharmaceutically acceptable salt thereof. 383. The conjugate of claim 382, wherein Q1, Q2, and Q3 are each independently selected from the side chain of a natural amino acid, or a pharmaceutically acceptable salt thereof. 384. The conjugate of claim 382, wherein at least one of Q1, Q2, and Q3 is selected from the side chain of a non-natural amino acid, or a pharmaceutically acceptable salt thereof. 385. The conjugate of claim 382, wherein at least two of Q1, Q2, and Q3 are independently selected from the side chain of a non-natural amino acid, or a pharmaceutically acceptable salt thereof. 386. The conjugate of claim 382, wherein each of Q1, Q2, and Q3 is independently selected from the side chain of a non-natural amino acid, or a pharmaceutically acceptable salt thereof. 387. The conjugate of claim 382, wherein each of Q1, Q2, and Q3 is independently selected from the side chain of serine, threonine, cysteine, glycine, proline, alanine, valine, isoleucine, leucine, methionine, phenylalanine, tyrosine, and tryptophan, or a pharmaceutically acceptable salt thereof. alkyl, C1-C2 hydroxyalkyl, C1-C5 alkamino, and C6-C35 alkaryl, or a pharmaceutically acceptable salt thereof. 389. The conjugate of claim 388, wherein each of Q1, Q2, and Q3 is independently selected from 2- methyl-1-propyl, 2-propyl, 1-hydroxyethyl, butyl, benzyl, hydroxymethyl, propyl, 2-butyl, methyl, and 2- aminoethyl, or a pharmaceutically acceptable salt thereof. 390. The conjugate of claim 389, wherein the combination of Q1, Q2, and Q3 is selected from:

or a pharmaceutically acceptable salt thereof. 391. The conjugate of any one of claims 382-390, wherein each m is 1, 2 or 3, or a pharmaceutically acceptable salt thereof. 392. The conjugate of any one of claims 382-390, wherein:

each m is 1, 2, 3, or 4;

393. The conjugate of any one of claims 382-390, wherein:

each A is independently selected from glycine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4- diaminobutyric acid, 3-hydroxyproline, threonine and 2-amino-4-phenylbutyric acid; and

each m is 1, 2, 3, 4, or 5;

or a pharmaceutically acceptable salt thereof. 394. The conjugate of claim 393, wherein each m is 1, 2, or 3, or a pharmaceutically acceptable salt thereof. 395. The conjugate of any one of claims 382-390, wherein:

each A is independently selected from glycine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4- diaminobutyric acid, 3-hydroxyproline, threonine and 2-amino-4-phenylbutyric acid; and

each m is 1, 2, 3, 4, or 5;

or a pharmaceutically acceptable salt thereof. 396. The conjugate of claim 395, wherein:

each m is 1, 2, or 3;

or a pharmaceutically acceptable salt thereof. 397. The conjugate of any one of claims 307-396, wherein when n is 2, E dimerizes to form an Fc domain,

or a pharmaceutically acceptable salt thereof.

,

(M-XX)

wherein:

each A is an independently selected amino acid;

each E is an Fc domain monomer;

each m is 0, 1, 2, 3, 4, or 5;

n is 1 or 2;

Q1, Q2, and Q3 are each independently selected from the side chain of an amino acid;

each d is 0 to 20; and

T is an integer from 1 to 20;

or a pharmaceutically acceptable salt thereof. 399. The conjugate of claim 398, wherein Q1, Q2, and Q3 are each independently selected from the side chain of a natural amino acid, or a pharmaceutically acceptable salt thereof. 400. The conjugate of claim 398, wherein at least one of Q1, Q2, and Q3 is selected from the side chain of a non-natural amino acid, or a pharmaceutically acceptable salt thereof. 401. The conjugate of claim 398, wherein at least two of Q1, Q2, and Q3 are independently selected from the side chain of a non-natural amino acid, or a pharmaceutically acceptable salt thereof. 402. The conjugate of claim 398, wherein each of Q1, Q2, and Q3 is independently selected from the side chain of a non-natural amino acid, or a pharmaceutically acceptable salt thereof. 403. The conjugate of claim 398, wherein each of Q1, Q2, and Q3 is independently selected from the side chain of serine, threonine, cysteine, glycine, proline, alanine, valine, isoleucine, leucine, methionine, phenylalanine, tyrosine, and tryptophan, or a pharmaceutically acceptable salt thereof. alkyl, C1-C2 hydroxyalkyl, C1-C5 alkamino, and C6-C35 alkaryl, or a pharmaceutically acceptable salt thereof. 405. The conjugate of claim 404, wherein each of Q1, Q2, and Q3 is independently selected from 2- methyl-1-propyl, 2-propyl, 1-hydroxyethyl, butyl, benzyl, hydroxymethyl, propyl, 2-butyl, methyl, and 2- aminoethyl, or a pharmaceutically acceptable salt thereof. 406. The conjugate of claim 405, wherein the combination of Q1, Q2, and Q3 is selected from:

407. The conjugate of any one of claims 398-406, wherein each m is 1, 2 or 3, or a pharmaceutically acceptable salt thereof. 408. The conjugate of any one of claims 398-406, wherein:

each m is 1, 2, 3, or 4;

or a pharmaceutically acceptable salt thereof. each A is independently selected from glycine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4- diaminobutyric acid, 3-hydroxyproline, threonine, 3-(2-naphthyl)alanine, and 2-amino-4-phenylbutyric acid; and

each m is 1, 2, 3, 4, or 5;

or a pharmaceutically acceptable salt thereof. 410. The conjugate of claim 409, wherein each m is 1, 2, or 3, or a pharmaceutically acceptable salt thereof. 411. The conjugate of any one of claims 398-406, wherein:

each A is independently selected from glycine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4- diaminobutyric acid, 3-hydroxyproline, threonine, 3-(2-naphthyl)alanine, and 2-amino-4-phenylbutyric acid;

each m is 1, 2, 3, 4, or 5;

or a pharmaceutically acceptable salt thereof. 412. The conjugate of claim 411, wherein:

each m is 1, 2, or 3;

or a pharmaceutically acceptable salt thereof. 413. The conjugate of any one of claims 398-412, wherein each d is 0 to 10, or a pharmaceutically acceptable salt thereof. 414. The conjugate of claim 413, wherein each d is 0 to 5, or a pharmaceutically acceptable salt thereof. 415. The conjugate of claim 414, where each d is 0 or 1, or a pharmaceutically acceptable salt thereof. 416. The conjugate of claim 398, wherein:

each A is independently selected from glycine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4- diaminobutyric acid, 3-hydroxyproline, threonine, 3-(2-naphthyl)alanine, and 2-amino-4-phenylbutyric acid, and 2-aminopentanoic acid;

each m is 1, 2, 3, or 4;

Q1, Q2, and Q3 are each independently selected from 2-methyl-1-propyl, 2-propyl, 1-hydroxyethyl, butyl, benzyl, hydroxymethyl, propyl, 2-butyl, methyl, and 2,4-diaminobutyric acid; and each d is 0 to 10;

or a pharmaceutically acceptable salt thereof. 417. The conjugate of claim 416, wherein Q1, Q2, and Q3 are each independently selected from 2-methyl-

418. The conjugate of claim 416 or 417, wherein:

each A is independently selected from glycine, 2,4-diaminobutyric acid, 3-hydroxyproline, 3-(2- naphthyl)alanine, and threonine, and 2-aminopentanoic acid;

each m is 2 to 4;

each d is 0 or 1;

or a pharmaceutically acceptable salt thereof. 419. The conjugate of any one of claims 307-418, wherein when n is 2, E dimerizes to form an Fc domain,

or a pharmaceutically acceptable salt thereof. 420. A con u ate of formula M-XXI :

,

(M-XXI)

wherein:

each A is an independently selected amino acid;

each E is an Fc domain monomer;

each m is 0, 1, 2, 3, 4, or 5;

n is 1 or 2;

Q1, Q2, and Q3 are each independently selected from the side chain of an amino acid;

each d is an integer from 1 to 26;

each e is an integer from 1 to 26; and

T is an integer from 1 to 20;

or a pharmaceutically acceptable salt thereof. 421. The conjugate of claim 420, wherein Q1, Q2, and Q3 are each independently selected from the side chain of a natural amino acid, or a pharmaceutically acceptable salt thereof. 422. The conjugate of claim 420, wherein at least one of Q1, Q2, and Q3 is selected from the side chain of

423. The conjugate of claim 420, wherein at least two of Q1, Q2, and Q3 are independently selected from the side chain of a non-natural amino acid, or a pharmaceutically acceptable salt thereof. 424. The conjugate of claim 420, wherein each of Q1, Q2, and Q3 is independently selected from the side chain of a non-natural amino acid, or a pharmaceutically acceptable salt thereof. 425. The conjugate of claim 420, wherein each of Q1, Q2, and Q3 is independently selected from the side chain of serine, threonine, cysteine, glycine, proline, alanine, valine, isoleucine, leucine, methionine, phenylalanine, tyrosine, and tryptophan, or a pharmaceutically acceptable salt thereof. 426. The conjugate of claim 420, wherein each of Q1, Q2, and Q3 is independently selected from C1-C4 alkyl, C1-C2 hydroxyalkyl, C1-C5 alkamino, and C6-C35 alkaryl, or a pharmaceutically acceptable salt thereof. 427. The conjugate of claim 426, wherein each of Q1, Q2, and Q3 is independently selected from 2- methyl-1-propyl, 2-propyl, 1-hydroxyethyl, butyl, benzyl, hydroxymethyl, propyl, 2-butyl, methyl, and 2- aminoethyl, or a pharmaceutically acceptable salt thereof.

or a pharmaceutically acceptable salt thereof. 429. The conjugate of any one of claims 420-428, wherein each m is 1, 2 or 3, or a pharmaceutically acceptable salt thereof. 430. The conjugate of any one of claims 420-428, wherein:

each m is 1, 2, 3, or 4;

or a pharmaceutically acceptable salt thereof. 431. The conjugate of any one of claims 420-428, wherein:

each A is independently selected from glycine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4- diaminobutyric acid, 3-hydroxyproline, threonine and 2-amino-4-phenylbutyric acid; and

each m is 1, 2, 3, 4, or 5;

or a pharmaceutically acceptable salt thereof. thereof. 433. The conjugate of any one of claims 420-428, wherein:

each A is independently selected from glycine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4- diaminobutyric acid, 3-hydroxyproline, threonine and 2-amino-4-phenylbutyric acid; and

each m is 1, 2, 3, 4, or 5;

or a pharmaceutically acceptable salt thereof. 434. The conjugate of claim 433, wherein:

each m is 1, 2, or 3;

or a pharmaceutically acceptable salt thereof. 435. The conjugate of any one of claims 420-434, wherein each d is an integer from 1 to 10, and each e is an integer from 1 to 10, or a pharmaceutically acceptable salt thereof. 436. The conjugate of claim 435, wherein each d is an integer from 1 to 5, and each e is an integer from 1 to 6, or a pharmaceutically acceptable salt thereof. 437. The conjugate of claim 435, wherein each d is 1, and each e is an integer from 1 to 6, or a pharmaceutically acceptable salt thereof. 438. The conjugate of claim 436, where each d is 1, and each e is 6, or a pharmaceutically acceptable salt thereof. 439. The conjugate of claim 420, wherein:

each A is independently selected from glycine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4- diaminobutyric acid, 3-hydroxyproline, threonine and 2-amino-4-phenylbutyric acid;

each m is 1, 2 or 3;

Q1, Q2, and Q3 are each independently selected from 2-methyl-1-propyl, 2-propyl, 1-hydroxyethyl, butyl, benzyl, hydroxymethyl, propyl, 2-butyl, methyl, and 2,4-diaminobutyric acid;

each d is an integer from 1 to 10; and

each e is an integer from 1 to10;

or a pharmaceutically acceptable salt thereof. 440. The conjugate of claim 420, wherein Q1, Q2, and Q3 are each independently selected from 2-methyl- 1-propyl and 1-hydroxyethyl, or a pharmaceutically acceptable salt thereof. each A is independently selected from 2,4-diaminobutyric acid and threonine;

each m is 2;

each d is 1; and

each e is 6;

or a pharmaceutically acceptable salt thereof. 442. The conjugate of any one of claims 307-441, wherein when n is 2, E dimerizes to form an Fc domain, or a pharmaceutically acceptable salt thereof. 44 . A n f f rm l M-XXII :

,

(M-XXII)

wherein:

each A is an independently selected amino acid;

each X is -CH2- or -C(O)-;

each Z is -C(O)NH- , -NHC(O)-,-CH2NH-, or O;

each E is an Fc domain monomer;

each m is 0, 1, 2, 3, 4, or 5;

n is 1 or 2;

Q1, Q2, and Q3 are each independently selected from the side chain of an amino acid;

each d is an integer from 1 to 26;

each e is an integer from 1 to 26; and

T is an integer from 1 to 20;

or a pharmaceutically acceptable salt thereof. 444. The conjugate of claim 443, wherein Q1, Q2, and Q3 are each independently selected from the side chain of a natural amino acid, or a pharmaceutically acceptable salt thereof. 445. The conjugate of claim 443, wherein at least one of Q1, Q2, and Q3 is selected from the side chain of

446. The conjugate of claim 443, wherein at least two of Q1, Q2, and Q3 are independently selected from the side chain of a non-natural amino acid, or a pharmaceutically acceptable salt thereof. 447. The conjugate of claim 443, wherein each of Q1, Q2, and Q3 is independently selected from the side chain of a non-natural amino acid, or a pharmaceutically acceptable salt thereof. 448. The conjugate of claim 443, wherein each of Q1, Q2, and Q3 is independently selected from the side chain of serine, threonine, cysteine, glycine, proline, alanine, valine, isoleucine, leucine, methionine, phenylalanine, tyrosine, and tryptophan, or a pharmaceutically acceptable salt thereof. 449. The conjugate of claim 443, wherein each of Q1, Q2, and Q3 is independently selected from C1-C4 alkyl, C1-C2 hydroxyalkyl, C1-C5 alkamino, and C6-C35 alkaryl, or a pharmaceutically acceptable salt thereof. 450. The conjugate of claim 449, wherein each of Q1, Q2, and Q3 is independently selected from 2- methyl-1-propyl, 2-propyl, 1-hydroxyethyl, butyl, benzyl, hydroxymethyl, propyl, 2-butyl, methyl, and 2- aminoethyl, or a pharmaceutically acceptable salt thereof.

or a pharmaceutically acceptable salt thereof. 452. The conjugate of any one of claims 443-451, wherein each m is 1, 2 or 3, or a pharmaceutically acceptable salt thereof. 453. The conjugate of any one of claims 443-451, wherein:

each m is 1, 2, 3, or 4;

or a pharmaceutically acceptable salt thereof. 454. The conjugate of any one of claims 443-451, wherein:

each A is independently selected from glycine, 2-aminoalanine, 3-aminoalanine, piperazine-2- carboxylic acid, 2,4-diaminobutyric acid, 3-hydroxyproline, threonine and 2-amino-4-phenylbutyric acid; and

each m is 1, 2, 3, 4, or 5;

or a pharmaceutically acceptable salt thereof. thereof. 456. The conjugate of any one of claims 443-451, wherein:

each A is independently selected from glycine, 2-aminoalanine, 3-aminoalanine, piperazine-2- carboxylic acid, 2,4-diaminobutyric acid, 3-hydroxyproline, threonine and 2-amino-4-phenylbutyric acid; each m is 1, 2, 3, 4, or 5;

or a pharmaceutically acceptable salt thereof. 457. The conjugate of claim 456, wherein:

each m is 1, 2, or 3;

or a pharmaceutically acceptable salt thereof. 458. The conjugate of claim 443, where each d is 1, and each e is 6 to 24, or a pharmaceutically acceptable salt thereof. 459. The conjugate of claim 443, wherein:

each A is independently selected from glycine, 2-aminoalanine, 3-aminoalanine, piperazine-2- carboxylic acid, 2,4-diaminobutyric acid, 3-hydroxyproline, threonine and 2-amino-4-phenylbutyric acid; each m is 1, 2 or 3;

Q1, Q2, and Q3 are each independently selected from 2-methyl-1-propyl, 2-propyl, 1-hydroxyethyl, butyl, benzyl, hydroxymethyl, propyl, 2-butyl, methyl, and 2,4-diaminobutyric acid;

each d is an integer from 1 to 10; and

each e is an integer from 1 to10;

or a pharmaceutically acceptable salt thereof. 460. The conjugate of claim 443, wherein Q1, Q2, and Q3 are each independently selected from benzyl, 2-methyl-1-propyl and 1-hydroxyethyl, or a pharmaceutically acceptable salt thereof. 461. The conjugate of claim 443, wherein:

each A is independently selected from 2-aminoalanine, 2,4-diaminobutyric acid, 3-hydroxyproline, piperazine-2-carboxylic acid, and threonine;

each Z is, -CH2NH-, -C(O)NH- or -NHC(O)-;

each m is 2 or 3;

each d is 1; and

each e is 4 to 26;

or a pharmaceutically acceptable salt thereof. each A is independently selected from glycine, 2-aminoalanine, 2,4-diaminobutyric acid, 3- hydroxyproline, and threonine;

each m is 2;

each d is 1; and

each e is 4 to 26;

or a pharmaceutically acceptable salt thereof. 463. The conjugate of any one of claims 307-462, wherein when n is 2, E dimerizes to form an Fc domain,

or a pharmaceutically acceptable salt thereof. 464. A con u ate of formula M-XXIV :

,

(M-XXIV)

wherein:

each A is an independently selected amino acid;

each E is an Fc domain monomer;

each m is 0, 1, 2, 3, 4, or 5;

n is 1 or 2;

Q1, Q2, and Q3 are each independently selected from the side chain of an amino acid; and each d is an integer from 1 to 26;

each e is an integer from 1 to 26; and

T is an integer from 1 to 20;

or a pharmaceutically acceptable salt thereof. 465. The conjugate of claim 464, wherein Q1, Q2, and Q3 are each independently selected from the side chain of a natural amino acid, or a pharmaceutically acceptable salt thereof. 466. The conjugate of claim 464, wherein at least one of Q1, Q2, and Q3 is selected from the side chain of t l i id h ti ll t bl lt th f the side chain of a non-natural amino acid, or a pharmaceutically acceptable salt thereof. 468. The conjugate of claim 464, wherein each of Q1, Q2, and Q3 is independently selected from the side chain of a non-natural amino acid, or a pharmaceutically acceptable salt thereof. 469. The conjugate of claim 464, wherein each of Q1, Q2, and Q3 is independently selected from the side chain of serine, threonine, cysteine, glycine, proline, alanine, valine, isoleucine, leucine, methionine, phenylalanine, tyrosine, and tryptophan, or a pharmaceutically acceptable salt thereof. 470. The conjugate of claim 464, wherein each of Q1, Q2, and Q3 is independently selected from C1-C4 alkyl, C1-C2 hydroxyalkyl, C1-C5 alkamino, and C6-C35 alkaryl, or a pharmaceutically acceptable salt thereof. 471. The conjugate of claim 470, wherein each of Q1, Q2, and Q3 is independently selected from 2- methyl-1-propyl, 2-propyl, 1-hydroxyethyl, butyl, benzyl, hydroxymethyl, propyl, 2-butyl, methyl, and 2- aminoethyl, or a pharmaceutically acceptable salt thereof.

473. The conjugate of any one of claims 464-472, wherein each m is 1, 2 or 3, or a pharmaceutically acceptable salt thereof. 474. The conjugate of any one of claims 464-472, wherein:

each m is 1, 2, 3, or 4;

or a pharmaceutically acceptable salt thereof. 475. The conjugate of any one of claims 464-474, wherein:

each A is independently selected from glycine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4- diaminobutyric acid, 3-hydroxyproline, threonine and 2-amino-4-phenylbutyric acid; and

each m is 1, 2, 3, 4, or 5;

or a pharmaceutically acceptable salt thereof. 476. The conjugate of claim 475, wherein each m is 1, 2, or 3, or a pharmaceutically acceptable salt

477. The conjugate of any one of claims 464-472, wherein:

each A is independently selected from glycine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4- diaminobutyric acid, 3-hydroxyproline, threonine and 2-amino-4-phenylbutyric acid;

each m is 1, 2, or 3;

or a pharmaceutically acceptable salt thereof. 478. The conjugate of any of claims 464-477, wherein each d is an integer from 1 to 10, and each e is an integer from 1 to 10, or a pharmaceutically acceptable salt thereof. 479. The conjugate of claim 478, where each d is an integer from 1 to 5, and each e is an integer from 1 to 5, or a pharmaceutically acceptable salt thereof. 480. The conjugate of claim 479, wherein each d is 1, and each e is 4, or a pharmaceutically acceptable salt thereof. 481. The conjugate of claim 464, wherein:

each A is independently selected from glycine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4- diaminobutyric acid, 3-hydroxyproline, threonine and 2-amino-4-phenylbutyric acid;

each m is 1, 2 or 3;

Q1, Q2, and Q3 are each independently selected from 2-methyl-1-propyl, 2-propyl, 1-hydroxyethyl, butyl, benzyl, hydroxymethyl, propyl, 2-butyl, methyl, and 2,4-diaminobutyric acid;

each d is an integer from 1 to 10; and

each e is an integer from 1 to 10;

or a pharmaceutically acceptable salt thereof. 482. The conjugate of claim 481, wherein Q1, Q2, and Q3 are each independently selected from benzyl, 2-methyl-1-propyl and 1-hydroxyethyl, or a pharmaceutically acceptable salt thereof. 483. The conjugate of claim 482, wherein:

each A is independently selected from 2,4-diaminobutyric acid and threonine;

each m is 2;

each d is 1; and

each e is 4;

or a pharmaceutically acceptable salt thereof. 484. The conjugate of any one of claims 307-483, wherein when n is 2, E dimerizes to form an Fc domain, or a pharmaceutically acceptable salt thereof.

wherein:

each A is an independently selected amino acid;

each E is an Fc domain monomer;

each m is 0, 1, 2, 3, 4, or 5;

n is 1 or 2;

Q1, Q2, and Q3 are each independently selected from the side chain of an amino acid;

each d is an integer from 1 to 26;

each e is an integer from 1 to 26;

T is an integer from 1 to 20;

or a pharmaceutically acceptable salt thereof. 486. The conjugate of claim 485, wherein Q1, Q2, and Q3 are each independently selected from the side chain of a natural amino acid, or a pharmaceutically acceptable salt thereof. 487. The conjugate of claim 485, wherein at least one of Q1, Q2, and Q3 is selected from the side chain of a non-natural amino acid, or a pharmaceutically acceptable salt thereof. 488. The conjugate of claim 485, wherein at least two of Q1, Q2, and Q3 are independently selected from the side chain of a non-natural amino acid, or a pharmaceutically acceptable salt thereof. 489. The conjugate of claim 485, wherein each of Q1, Q2, and Q3 is independently selected from the side chain of a non-natural amino acid, or a pharmaceutically acceptable salt thereof. 490. The conjugate of claim 485, wherein each of Q1, Q2, and Q3 is independently selected from the side chain of serine, threonine, cysteine, glycine, proline, alanine, valine, isoleucine, leucine, methionine, phenylalanine, tyrosine, and tryptophan, or a pharmaceutically acceptable salt thereof. alkyl, C1-C2 hydroxyalkyl, C1-C5 alkamino, and C6-C35 alkaryl, or a pharmaceutically acceptable salt thereof. 492. The conjugate of claim 491, wherein each of Q1, Q2, and Q3 is independently selected from 2- methyl-1-propyl, 2-propyl, 1-hydroxyethyl, butyl, benzyl, hydroxymethyl, propyl, 2-butyl, methyl, and 2- aminoethyl, or a pharmaceutically acceptable salt thereof. 493. The conjugate of claim 485, wherein the combination of Q1, Q2, and Q3 is selected from:

or a pharmaceutically acceptable salt thereof. 494. The conjugate of any one of claims 485-493, wherein each m is 1, 2 or 3, or a pharmaceutically acceptable salt thereof. 495. The conjugate of any one of claims 485-494, wherein:

each m is 1, 2, 3, or 4;

496. The conjugate of any one of claims 485-495, wherein:

each A is independently selected from glycine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4- diaminobutyric acid, 3-hydroxyproline, threonine, 2-aminooctanoic acid, and 2-amino-4-phenylbutyric acid; and

each m is 1, 2, 3, 4, or 5;

or a pharmaceutically acceptable salt thereof. 497. The conjugate of claim 496, wherein each m is 1, 2, or 3, or a pharmaceutically acceptable salt thereof. 498. The conjugate of any one of claims 485-497, wherein:

each A is independently selected from glycine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4- diaminobutyric acid, 3-hydroxyproline, threonine, 2-aminooctanoic acid, and 2-amino-4-phenylbutyric acid;

each m is 1, 2, 3, or 4;

or a pharmaceutically acceptable salt thereof. 499. The conjugate of any of claims 485-498, wherein each d is an integer from 1 to 10, and each e is an integer from 1 to 10, or a pharmaceutically acceptable salt thereof. 500. The conjugate of claim 499, where each d is an integer from 1 to 5, and each e is an integer from 1 to 5, or a pharmaceutically acceptable salt thereof. 501. The conjugate of claim 500, wherein each m is 4, each d is 1, and each e is 4, or a pharmaceutically acceptable salt thereof. 502. The conjugate of claim 485, wherein:

each A is independently selected from glycine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4- diaminobutyric acid, 3-hydroxyproline, threonine, 2-aminooctanoic acid, and 2-amino-4-phenylbutyric acid;

each m is 1, 2, 3, or 4;

Q1, Q2, and Q3 are each independently selected from 2-methyl-1-propyl, 2-propyl, 1-hydroxyethyl, butyl, benzyl, hydroxymethyl, propyl, 2-butyl, methyl, and 2,4-diaminobutyric acid;

each d is 1; and

each e is an integer from 1 to 5;

or a pharmaceutically acceptable salt thereof. 503. The conjugate of claim 502, wherein Q1, Q2, and Q3 are each independently selected from benzyl,

504. The conjugate of claim 503, wherein:

each A is independently selected from 2,4-diaminobutyric acid, 2-aminooctanoic acid, and threonine;

each m is 4;

each d is 1; and

each e is 4;

or a pharmaceutically acceptable salt thereof. 505. The conjugate of any one of claims 307-504, wherein when n is 2, E dimerizes to form an Fc domain,

or a pharmaceutically acceptable salt thereof. 506. A conju ate of formula M-XXVI :

,

(M-XXVI)

wherein:

each A is an independently selected amino acid;

each X is heteroaryl or heterocyclyl;

each E is an Fc domain monomer;

each m is 0, 1, 2, 3, 4, or 5;

n is 1 or 2;

Q1, Q2, and Q3 are each independently selected from the side chain of an amino acid;

each d is an integer from 1 to 26;

each e is an integer from 1 to 26; and

T is an integer from 1 to 20;

or a pharmaceutically acceptable salt thereof. 507. The conjugate of claim 506, wherein Q1, Q2, and Q3 are each independently selected from the side chain of a natural amino acid, or a pharmaceutically acceptable salt thereof. a non-natural amino acid, or a pharmaceutically acceptable salt thereof. 509. The conjugate of claim 506, wherein at least two of Q1, Q2, and Q3 are independently selected from the side chain of a non-natural amino acid, or a pharmaceutically acceptable salt thereof. 510. The conjugate of claim 506, wherein each of Q1, Q2, and Q3 is independently selected from the side chain of a non-natural amino acid, or a pharmaceutically acceptable salt thereof. 511. The conjugate of claim 506, wherein each of Q1, Q2, and Q3 is independently selected from the side chain of serine, threonine, cysteine, glycine, proline, alanine, valine, isoleucine, leucine, methionine, phenylalanine, tyrosine, and tryptophan, or a pharmaceutically acceptable salt thereof. 512. The conjugate of claim 506, wherein each of Q1, Q2, and Q3 is independently selected from C1-C4 alkyl, C1-C2 hydroxyalkyl, C1-C5 alkamino, and C6-C35 alkaryl, or a pharmaceutically acceptable salt thereof. 513. The conjugate of claim 506, wherein each of Q1, Q2, and Q3 is independently selected from 2- methyl-1-propyl, 2-propyl, 1-hydroxyethyl, butyl, benzyl, hydroxymethyl, propyl, 2-butyl, methyl, and 2- aminoethyl, or a pharmaceutically acceptable salt thereof.

or a pharmaceutically acceptable salt thereof. 515. The conjugate of any one of claims 506-514, wherein each m is 1, 2 or 3, or a pharmaceutically acceptable salt thereof. 516. The conjugate of any one of claims 506-515, wherein:

each m is 1, 2, 3, or 4;

or a pharmaceutically acceptable salt thereof. 517. The conjugate of any one of claims 506-516, wherein:

each A is independently selected from glycine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4- diaminobutyric acid, 3-hydroxyproline, threonine, 2-aminooctanoic acid, 3-(2-naphthyl)alanine, and 2- amino-4-phenylbutyric acid; and

each m is 1, 2, 3, 4, or 5;

or a pharmaceutically acceptable salt thereof. thereof. 519. The conjugate of any one of claims 506-518, wherein:

each A is independently selected from glycine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4- diaminobutyric acid, 3-hydroxyproline, threonine, 2-aminooctanoic acid, 3-(2-naphthyl)alanine, and 2- amino-4-phenylbutyric acid;

each m is 1, 2, 3, or 4;

or a pharmaceutically acceptable salt thereof. 520. The conjugate of any of claims 506-519, wherein each d is an integer from 1 to 10, and each e is an integer from 1 to 10, or a pharmaceutically acceptable salt thereof. 521. The conjugate of claim 520, where each d is an integer from 1 to 5, and each e is an integer from 1 to 5, or a pharmaceutically acceptable salt thereof. 522. The conjugate of claim 521, wherein each m is 4, each d is 1, and each e is 4, or a pharmaceutically acceptable salt thereof. 523. The conjugate of claim 506, wherein:

each A is independently selected from glycine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4- diaminobutyric acid, 3-hydroxyproline, threonine, 2-aminooctanoic acid, 3-(2-naphthyl)alanine, and 2- amino-4-phenylbutyric acid;

each X is heteroaryl;

each m is 1, 2, 3, or 4;

Q1, Q2, and Q3 are each independently selected from 2-methyl-1-propyl, 2-propyl, 1-hydroxyethyl, butyl, benzyl, hydroxymethyl, propyl, 2-butyl, methyl, and 2,4-diaminobutyric acid;

each d is an integer from 1 to 5; and

each e is an integer from 1 to 5;

or a pharmaceutically acceptable salt thereof. 524. The conjugate of claim 523, wherein:

each A is independently selected from 2,4-diaminobutyric acid, threonine, 2-aminooctanoic acid, and 2-amino-4-phenylbutyric acid;

each m is 2, 3, or 4; and

each e is an integer from 1 to 3;

or a pharmaceutically acceptable salt thereof. 525. The conjugate of claim 524, wherein each X is triazolyl, or a pharmaceutically acceptable salt

526. The conjugate of claim 525, wherein each X is 1,4-triazolyl, or a pharmaceutically acceptable salt thereof. 527. The conjugate of claim 524, wherein Q1, Q2, and Q3 are each independently selected from benzyl, 2-methyl-1-propyl and 1-hydroxyethyl, or a pharmaceutically acceptable salt thereof. 528. The conjugate of claim 527, wherein:

each A is independently selected from 2,4-diaminobutyric acid, 2-aminooctanoic acid, and threonine;

each m is 4;

each d is 1; and

each e is 3;

or a pharmaceutically acceptable salt thereof. 529. The conjugate of any one of claims 307-528, wherein when n is 2, E dimerizes to form an Fc domain,

or a pharmaceutically acceptable salt thereof. 530. The conjugate of any one of claims 307-529, wherein a concentration of the conjugate, or a pharmaceutically acceptable salt thereof, that activates an immune cell is less than or equal to 10,000 nM. 531. The conjugate of claim 530, wherein a concentration of the conjugate, or a pharmaceutically acceptable salt thereof, that activates an immune cell is less than or equal to equal to 1,000 nM. 532. The conjugate of claim 531, wherein a concentration of the conjugate, or a pharmaceutically acceptable salt thereof, that activates an immune cell is less than or equal to equal to 100 nM. 533. A pharmaceutical composition comprising a conjugate of any of claims 1-532, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient. 534. The pharmaceutical composition of claim 533, further comprising an antibacterial agent. 535. The pharmaceutical composition of claim 534, wherein the antibacterial agent is selected from the group consisting of linezolid, tedizolid, posizolid, radezolid, retapamulin, valnemulin, tiamulin, azamulin, lefamulin, plazomicin, amikacin, gentamicin, gamithromycin, kanamycin, neomycin, netilmicin, tobramycin, paromomycin, streptomycin, spectinomycin, geldanamycin, herbimycin, rifaximin, loracarbef, ertapenem doripenem imipenem/cilastatin meropenem cefadroxil cefazolin cefalotin cefalexin ceftobiprole, teicoplanin, vancomycin, telavancin, dalbavancin, oritavancin, clindamycin, lincomycin, daptomycin, solithromycin, azithromycin, clarithromycin, dirithromycin, erythromycin, roxithromycin, troleandomycin, telithromycin, spiramycin, aztreonam, furazolidone, nitrofurantoin, amoxicillin, ampicillin, azlocillin, carbenicillin, cloxacillin, dicloxacillin, flucloxacillin, mezlocillin, methicillin, nafcillin, oxacillin, penicillin g, penicillin v, piperacillin, penicillin g, temocillin, ticarcillin, amoxicillin clavulanate,

ampicillin/sulbactam, piperacillin/tazobactam, ticarcillin/clavulanate, bacitracin, ciprofloxacin, enoxacin, gatifloxacin, gemifloxacin, levofloxacin, lomefloxacin, moxifloxacin, nalidixic acid, norfloxacin, ofloxacin, trovafloxacin, grepafloxacin, sparfloxacin, temafloxacin, mafenide, sulfacetamide, sulfadiazine, silver sulfadiazine, sulfadimethoxine, sulfamethizole, sulfamethoxazole, sulfanilimide, sulfasalazine, sulfisoxazole, trimethoprim-sulfamethoxazole (tmp-smx), sulfonamidochrysoidine, eravacycline, demeclocycline, doxycycline, minocycline, oxytetracycline, tetracycline, clofazimine, dapsone, capreomycin, cycloserine, ethambutol(bs), ethionamide, isoniazid, pyrazinamide, rifampicin, rifabutin, rifapentine, streptomycin, arsphenamine, chloramphenicol, fosfomycin, fusidic acid, metronidazole, mupirocin, platensimycin, quinupristin/dalfopristin, thiamphenicol, tigecycline, tinidazole, and

trimethoprim, prodrugs thereof, and pharmaceutically acceptable salts thereof. 536. The pharmaceutical composition of claim 535, wherein a prodrug of tedizolid is tedizolid phosphate. 537. The pharmaceutical composition of claim 535, wherein the antibacterial agent is tedizolid, azithromycin, meropenem, amikacin, levofloxacin, rifampicin, linezolid, erythromycin, or solithromycin. 538. The pharmaceutical composition of claim 537, wherein the antibacterial agent is tedizolid, azithromycin, meropenem, amikacin, or levofloxacin. 539. A method of protecting against or treating a bacterial infection in a subject, said method comprising administering to said subject a conjugate of any one of claims 1-531. 540. The method of claim 539, further comprising administering to the subject an antibacterial agent. 541. A method of protecting against or treating a bacterial infection in a subject, said method comprising administering to said subject (1) a conjugate of any one of claims 1-532 and (2) an antibacterial agent. 542. A method of inducing immune cell activation of the immune response in a subject having a bacterial infection, said method comprising administering to said subject a conjugate of any one of claims 1-532. 543. The method of claim 542, further comprising administering to the subject an antibacterial agent. infection, said method comprising administering to said subject (1) a conjugate of any one of claims 1-532 and (2) an antibacterial agent. 545. The method of claim 540 or 5416, wherein the conjugate and the antibacterial agent are administered substantially simultaneously. 546. The method of claim 540 or 541, wherein the conjugate and the antibacterial agent are administered separately. 547. The method of claim 546, wherein the conjugate is administered first, followed by administering of the antibacterial agent alone. 548. The method of claim 546, wherein the antibacterial agent is administered first, followed by administering of the conjugate alone.

549. The method of claim 540 or 541, wherein the conjugate and the antibacterial agent are administered substantially simultaneously, followed by administering of the conjugate or the antibacterial agent alone. 550. The method of claim 540 or 541, wherein the conjugate or the antibacterial agent is administered first, followed by administering of the conjugate and the antibacterial agent substantially simultaneously. 551. The method of claim 540 or 541, wherein administering the conjugate and the antibacterial agent together lowers the MIC of each of the conjugate and the antibacterial agent relative to the MIC of each of the conjugate and the antibacterial agent when each is used alone. 552. The method of any one of claims 539-551, wherein the conjugate and/or the antibacterial agent is administered intramuscularly, intravenously, intradermally, intraarterially, intraperitoneally, intralesionally, intracranially, intraarticularly, intraprostatically, intrapleurally, intratracheally, intranasally, intravitreally, intravaginally, intrarectally, topically, intratumorally, peritoneally, subcutaneously, subconjunctival, intravesicularlly, mucosally, intrapericardially, intraumbilically, intraocularally, orally, locally, by inhalation, by injection, or by infusion. 553. A method of preventing, stabilizing, or inhibiting the growth of bacteria, or killing bacteria, comprising contacting the bacteria or a site susceptible to bacterial growth with a conjugate of any of claims 1-532. 554. The method of claim 553, further comprising contacting the bacteria or the site susceptible to bacterial growth with an antibacterial agent. contacting the bacteria or a site susceptible to bacterial growth with (1) a conjugate of any of claims 1-532 and (2) an antibacterial agent. 556. The method of any one of claims 539-554, wherein the antibacterial agent is selected from the group consisting of linezolid, tedizolid, posizolid, radezolid, retapamulin, valnemulin, tiamulin, azamulin, lefamulin, plazomicin, amikacin, gentamicin, gamithromycin, kanamycin, neomycin, netilmicin, tobramycin, paromomycin, streptomycin, spectinomycin, geldanamycin, herbimycin, rifaximin, loracarbef, ertapenem, doripenem, imipenem/cilastatin, meropenem, cefadroxil, cefazolin, cefalotin, cefalexin, cefaclor, cefamandole, cefoxitin, cefprozil, cefuroxime, cefixime, cefdinir, cefditoren, cefoperazone, cefotaxime, cefpodoxime, ceftazidime, ceftibuten, ceftizoxime, ceftriaxone, cefepime, ceftaroline fosamil, ceftobiprole, teicoplanin, vancomycin, telavancin, dalbavancin, oritavancin, clindamycin, lincomycin, daptomycin, solithromycin, azithromycin, clarithromycin, dirithromycin, erythromycin, roxithromycin, troleandomycin, telithromycin, spiramycin, aztreonam, furazolidone, nitrofurantoin, amoxicillin, ampicillin, azlocillin, carbenicillin, cloxacillin, dicloxacillin, flucloxacillin, mezlocillin, methicillin, nafcillin, oxacillin, penicillin g, penicillin v, piperacillin, penicillin g, temocillin, ticarcillin, amoxicillin clavulanate,

ampicillin/sulbactam, piperacillin/tazobactam, ticarcillin/clavulanate, bacitracin, ciprofloxacin, enoxacin, gatifloxacin, gemifloxacin, levofloxacin, lomefloxacin, moxifloxacin, nalidixic acid, norfloxacin, ofloxacin, trovafloxacin, grepafloxacin, sparfloxacin, temafloxacin, mafenide, sulfacetamide, sulfadiazine, silver sulfadiazine, sulfadimethoxine, sulfamethizole, sulfamethoxazole, sulfanilimide, sulfasalazine, sulfisoxazole, trimethoprim-sulfamethoxazole (tmp-smx), sulfonamidochrysoidine, eravacycline, demeclocycline, doxycycline, minocycline, oxytetracycline, tetracycline, clofazimine, dapsone, capreomycin, cycloserine, ethambutol(bs), ethionamide, isoniazid, pyrazinamide, rifampicin, rifabutin, rifapentine, streptomycin, arsphenamine, chloramphenicol, fosfomycin, fusidic acid, metronidazole, mupirocin, platensimycin, quinupristin/dalfopristin, thiamphenicol, tigecycline, tinidazole, and

trimethoprim, prodrugs thereof, and pharmaceutically acceptable salts thereof. 557. The method of claim 556, wherein a prodrug of tedizolid is tedizolid phosphate. 558. The method of claim 556, wherein the antibacterial agent is tedizolid, azithromycin, meropenem, amikacin, levofloxacin, rifampicin, linezolid, erythromycin, or solithromycin. 559. The method of claim 558, wherein the antibacterial agent is tedizolid, azithromycin, meropenem, amikacin, or levofloxacin. 560. The method of any one of claims 539-559, wherein the bacterial infection is caused by Gram- negative bacteria. 561. The method of any one of claims 539-560, wherein the bacterial infection is caused by a resistant

562. The method of claim 561, wherein the resistant strain of bacteria possesses the mcr-1 gene, the mcr-2 gene, and/or a chromosomal mutation conferring polymyxin resistance. 563. The method of claim 562, wherein the resistant strain of bacteria possesses the mcr-1 gene. 564. The method of claim 562, wherein the resistant strain of bacteria possesses the mcr-2 gene. 565. The method of claim 562, wherein the resistant strain of bacteria possesses a chromosomal mutation conferring polymyxin resistance. 566. The method of any one of claims 539-565, wherein the resistant strain of bacteria is a resistant strain of E. coli. 567. A method of preventing lipopolysaccharides (LPS) in Gram-negative bacteria from activating an immune system in a subject, comprising administering to the subject a conjugate of any one of claims 1- 532. 568. The method of claim 567, wherein the method prevents LPS from activating a macrophage. 569. The method of claim 567 or 568, wherein the method prevents LPS-induced nitric oxide production from a macrophage.

Description:
COMPOSITIONS AND METHODS FOR THE TREATMENT OF BA Background

The need for novel antibacterial treatments for bacterial infections is significant and especially critical in the medical field. Antibacterial resistance is a serious global healthcare threat. Polymyxins are a class of antibiotics that exhibit potent antibacterial activities against Gram-negative bacteria. However, the use of polymyxins as an antibiotic has been limited due to the associated toxicity and adverse effects (e.g., nephrotoxicity). Moreover, mcr-1, a plasmid-borne gene conferring bacterial resistance to polymyxins, has a high potential for dissemination and further threatens the efficacy of this class of antibiotics.

Because of the shortcomings of existing antibacterial treatments, combined with the emergence of multidrug-resistant Gram-negative bacteria, there is a need in the art for improved antibacterial therapies having greater efficacy, bioavailability, and reduced toxicity. Summary

The disclosure relates to conjugates, compositions, and methods for inhibiting bacterial growth (e.g., Gram-negative bacterial growth) and for the treatment of bacterial infections (e.g., Gram-negative bacterial infections). In particular, such conjugates contain monomers or dimers of cyclic heptapeptides conjugated to Fc domains. The monomers or dimers of cyclic heptapeptides in the conjugates bind to lipopolysaccharides (LPS) in the cell membrane of Gram-negative bacteria to disrupt and permeabilize the cell membrane, leading to cell death and/or sensitization of the Gram-negtaive bacteria to other antibiotics, and the Fc domains in the conjugates bind to FcγRs (e.g., FcRn, FcγRI, FcγRIIa, FcγRIIc, FcγRIIIa, and FcγRIIIb) on immune cells, e.g., neutrophils, to activate phagocytosis and effector functions, such as antibody-dependent cell-mediated cytotoxicity (ADCC), thus leading to the engulfment and destruction of bacterial cells by immune cells and further enhancing the antibacterial activity of the conjugates.

In one aspect, the disclosure features n escribed by formula (1):

(1)

wherein each M1 includes a first cyclic heptapeptide including a linking nitrogen and each M2 includes a second cyclic heptapeptide including a linking nitrogen; each E includes an Fc domain monomer (e.g., an Fc domain monomer having the sequence of any one of SEQ ID NOs: 1-14, 1-29, or 1-31); L’ 1 in each M2-L’ 1 -M1 is a linker covalently attached to a sulfur atom of a hinge cysteine in each E and to the linking nitrogen in each of M1 and M2; T is an integer from 1, 2, 3, 4, or 5, the two squiggly lines connected to the two Es indicate that each M2-L’ 1 -M1 is covalently attached to a pair of sulfur atoms of two hinge cysteines in the two Es, or a pharmaceutically acceptable salt thereof. When T is greater than 1 (e.g., T is 2, 3, 4, or 5), each M2-L’ 1 -M1 may be independently selected (e.g., indep

the M2-L’ 1 -M1 structures described herein).

In another aspect, the invention features a con u ate described by formula (2):

wherein each M includes a cyclic heptapeptide including a linking nitrogen; each E includes an Fc domain monomer (e.g., an Fc domain monomer having the sequence of any one of SEQ ID NOs: 1-14, 1-29, or 1-31); L’ 1 in each L’ 1 -M is a linker covalently attached to a sulfur atom in a hinge cysteine in E and to the linking nitrogen in M; T is 1, 2, 3, 4, or 5, the two squiggly lines connected to the two sulfur atoms indicate that each L’ 1 -M is covalently attached to a pair of sulfur atoms of two hinge cysteines in the two Es, or a pharmaceutically acceptable salt thereof.

In some embodiments, each E includes an Fc domain monomer having the sequence of any one of SEQ ID Nos: 1-14 or 1-31.

In some embodiments of these aspects, each E comprises the sequence

MVRSDKTHTCPPCPPC*KC*PAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDV SHEDPEVKFNWYV DGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKA KGQPREPQV YTLPPSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYS KLTVDKSRW QQGNVFSCSVMHEALHNHYTQKSLSLSPGKHHHHHH (SEQ ID NO: 10).

In some embodiments, at least one of the pair of sulfur atoms is the sulfur atom corresponding to (e.g., the sulfur atom of) a hinge cysteine of SEQ ID NO: 10, i.e., Cys10, Cys13, Cys16, or Cys18 of SEQ ID NO: 10. In some embodiments, the pair of sulfur atoms are the sulfur atoms corresponding to (e.g., the sulfur atoms of) Cys10 and Cys13 in SEQ ID NO: 10, Cys10 and Cys16 in SEQ ID NO: 10, Cys 30 and Cys18 in SEQ ID NO: 10, Cys13 and Cys 36 in SEQ ID NO: 10, Cys13 and Cys 38 in SEQ ID NO: 10, and/or Cys 36 and Cys 38 in SEQ ID NO: 10.

In some embodiments, when T is 2, the pair of sulfur atoms are (e.g., the sulfur atoms corresponding to) Cys10 and Cys13 in SEQ ID NO: 10 or Cys 36 and Cys 38 in SEQ ID NO: 10.

In some embodiments, the pair of sulfur atoms include one sulfur atom of a cysteine from each E, i.e., L ’1 -M along with the sulfur atoms to which it is attached forms a bridge between two Fc domains (e.g., two Fc domains comprising the sequence of SEQ ID NO: 10). In some embodiments, the pair of sulfur atoms are the sulfur atom corresponding to (e.g., the sulfur atom of) Cys10 of SEQ ID NO: 10 from one E and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys10 of SEQ ID NO: 10 from another E. In some embodiments, the pair of sulfur atoms are the sulfur atom corresponding to (e.g., the sulfur atom of) Cys13 of SEQ ID NO: 10 from one E and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys13 of SEQ ID NO: 10 from another E. In some embodiments, the pair of sulfur atoms are the sulfur atom corresponding to (e.g., the sulfur atom of) Cys16 of SEQ ID NO: 10 from one E and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys16 of SEQ ID NO: 10 from another E. In some embodiments, the pair of sulfur atoms are the sulfur atom corresponding to (e.g., the sulfur atom of) Cys18 of SEQ ID NO: 10 from one E and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys18 of SEQ ID NO: 10 from another E.

In some embodiments, when T is 2, the pairs of sulfur atoms are the sulfur atom corresponding to (e.g., the sulfur atom of) Cys10 of SEQ ID NO: 10 from one E and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys10 of SEQ ID NO: 10 from another E and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys13 of SEQ ID NO: 10 from one E and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys13 of SEQ ID NO: 10 from another E. In some embodiments, when T is 2, the pairs of sulfur atoms are the sulfur atom corresponding to (e.g., the sulfur atom of) Cys10 of SEQ ID NO: 10 from one E and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys10 of SEQ ID NO: 10 from another E and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys16 of SEQ ID NO: 1 0 from one E and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys16 of SEQ ID NO: 10 from another E. In some embodiments, when T is 2, the pairs of sulfur atoms are the sulfur atom

corresponding to (e.g., the sulfur atom of) Cys10 of SEQ ID NO: 10 from one E and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys10 of SEQ ID NO: 10 from another E and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys18 of SEQ ID NO: 10 from one E and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys18 of SEQ ID NO: 10 from another E.

In some embodiments, when T is 2, the pairs of sulfur atoms are the sulfur atom corresponding to (e.g., the sulfur atom of) Cys13 of SEQ ID NO: 10 from one E and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys13 of SEQ ID NO: 10 from another E and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys16 of SEQ ID NO: 10 from one E and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys16 of SEQ ID NO: 10 from another E. In some embodiments, when T is 2, the pairs of sulfur atoms are the sulfur atom corresponding to (e.g., the sulfur atom of) Cys13 of SEQ ID NO: 10 from one E and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys13 of SEQ ID NO: 10 from another E and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys18 of SEQ ID NO: 1 0 from one E and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys18 of SEQ ID NO: 10 from another E.

In some embodiments, when T is 2, the pairs of sulfur atoms are the sulfur atom corresponding to (e.g., the sulfur atom of) Cys16 of SEQ ID NO: 10 from one E and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys16 of SEQ ID NO: 10 from another E and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys18 of SEQ ID NO: 10 from one E and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys18 of SEQ ID NO: 10 from another E.

In some embodiments, when T is 3, the pairs of sulfur atoms are the sulfur atom corresponding to (e.g., the sulfur atom of) Cys10 of SEQ ID NO: 10 from one E and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys10 of SEQ ID NO: 10 from another E; the sulfur atom corresponding to (e.g., the sulfur atom of) Cys13 of SEQ ID NO: 10 from one E and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys13 of SEQ ID NO: 10 from another E; and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys16 of SEQ ID NO: 10 from one E and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys1 6 of SEQ ID NO: 1 0 from another E. In some embodiments, when T is 3, the pairs of sulfur atoms are the sulfur atom corresponding to (e.g., the sulfur atom of) Cys10

and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys10 of SEQ ID NO: 10 from another E; the sulfur atom corresponding to (e.g., the sulfur atom of) Cys13 of SEQ ID NO: 10 from one E and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys13 of SEQ ID NO: 10 from another E; and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys18 of SEQ ID NO: 10 from one E and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys18 of SEQ ID NO: 10 from another E. In some embodiments, when T is 3, the pairs of sulfur atoms are the sulfur atom corresponding to (e.g., the sulfur atom of) Cys10 of SEQ ID NO: 10 from one E and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys10 of SEQ ID NO: 10 from another E; the sulfur atom corresponding to (e.g., the sulfur atom of) Cys18 of SEQ ID NO: 10 from one E and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys18 of SEQ ID NO: 10 from another E; and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys16 of SEQ ID NO: 10 from one E and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys16 of SEQ ID NO: 10 from another E. In some embodiments, when T is 3, the pairs of sulfur atoms are the sulfur atom corresponding to (e.g., the sulfur atom of) Cys13 of SEQ ID NO: 10 from one E and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys13 of SEQ ID NO: 10 from another E; the sulfur atom corresponding to (e.g., the sulfur atom of) Cys18 of SEQ ID NO: 10 from one E and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys18 of SEQ ID NO: 10 from another E; and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys16 of SEQ ID NO: 10 from one E and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys16 of SEQ ID NO: 10 from another E.

In some embodiments, when T is 3, the pairs of sulfur atoms are the sulfur atom corresponding to (e.g., the sulfur atom of) Cys10 of SEQ ID NO: 10 from one E and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys10 of SEQ ID NO: 10 from another E; the sulfur atom corresponding to (e.g., the sulfur atom of) Cys13 of SEQ ID NO: 10 from one E and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys13 of SEQ ID NO: 10 from another E; the sulfur atom corresponding to (e.g., the sulfur atom of) Cys16 of SEQ ID NO: 10 from one E and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys16 of SEQ ID NO: 10 from another E; and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys18 of SEQ ID NO: 10 from one E and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys18 of SEQ ID NO: 10 from another E.

In some embodiments, the conjugate has the structure:

wherein each of a, b, c, and d is, independently, 0 or 1 and wherein when a, b, c, or d is 0, the two sulfur atoms form a disulfide bond.

In some embodiments, a is 1 and b, c, and d are 0. In some embodiments, a and b are 1 and c and d are 0. In some embodiments, a and c are 1 and b and d are 0. In some embodiments, a and d are 1 and b and c are 0. In some embodiments, a, b, and c are 1 and d is 0. In some embodiments, a, b, and d are 1 and c is 0. In some embodiments, a, c, and d are 1 and b is 0. In some embodiments, b and c are 1 and a and d are 0. In some embodiments, b and d are 1 and a and c are 0. In some

embodiments, b, c, and d are 1 and a is 0. In some embodiments, c and d are 1 and a and b are 0. In some embodiments, a, b, c, and d are 1.

In some embodiments of these aspects, each E comprises the sequence

MVRSDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEV KFNWYVDGVEVH NAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPRE PQVYTLPPSR EEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKS RWQQGNVFS CSVMHEALHNHYTQKSLSLSPGK (SEQ ID NO: 4).

In some embodiments, at least one of the pair of sulfur atoms is the sulfur atom corresponding to (e.g., the sulfur atom of) a hinge cysteine of SEQ ID NO: 4, i.e., Cys10 and/or Cys13. In some embodiments, the pair of sulfur atoms are the sulfur atoms corresponding to (e.g., the sulfur atoms of) Cys10 and Cys13 in SEQ ID NO: 4.

In some embodiments, the pair of sulfur atoms include one sulfur atom of a cysteine from each E, i.e., L ’1 -M along with the sulfur atoms to which it is attached forms a bridge between two Fc domains (e.g., two Fc domains comprising the sequence of SEQ ID NO: 4). In some embodiments, the pair of sulfur atoms are the sulfur atom corresponding to (e.g., the sulfur atom of) Cys10 of SEQ ID NO: 4 from one E and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys10 of SEQ ID NO: 4 from another E. In some embodiments, the pair of sulfur atoms are the sulfur atom corresponding to (e.g., the sulfur atom of) Cys13 of SEQ ID NO: 4 from one E and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys13 of SEQ ID NO: 4 from another E. In some embodiments, when T is 2, the pairs of sulfur atoms are the sulfur atom corresponding to (e.g., the sulfur atom of) Cys10 of SEQ ID NO: 4 from one E and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys10 of SEQ ID NO: 4 from another E and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys13 of SEQ ID NO: 4 fro

corresponding to (e.g., the sulfur atom of) Cys13 of SEQ ID NO: 4 from another E.

In some embodiments, the conjugate has the structure:

wherein each of a and b is, independently, 0 or 1 and wherein when a or b is 0, the two sulfur atoms form a disulfide bond. In some embodiments, a is 1 and b is 0. In some embodiments, a is 0 and b is 1. In some embodiments, a and b are 1.

In m m im n h M-L ’1 h h r r :

.

I n some embodiments, each M-L’1 has the structure:

.

In some embodiments of these aspects, each E comprises the sequence

MVRSDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEV KFNWYVDGVEVH NAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPRE PQVYTLPPSR EEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSF

CSVMHEALHNHYTQKSLSLSPGKHHHHHH (SEQ ID NO: 8).

In some embodiments, at least one of the pair of sulfur atoms is the sulfur atom corresponding to (e.g., the sulfur atom of) a hinge cysteine of SEQ ID NO: 8, i.e., Cys10 and/or Cys13. In some embodiments, the pair of sulfur atoms are the sulfur atoms corresponding to (e.g., the sulfur atoms of) Cys10 and Cys13 in SEQ ID NO: 8.

In some embodiments, the pair of sulfur atoms include one sulfur atom of a cysteine from each E, i.e., L ’1 -M along with the sulfur atoms to which it is attached forms a bridge between two Fc domains (e.g., two Fc domains comprising the sequence of SEQ ID NO: 8). In some embodiments, the pair of sulfur atoms are the sulfur atom corresponding to (e.g., the sulfur atom of) Cys10 of SEQ ID NO: 8 from one E and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys10 of SEQ ID NO: 8 from another E. In some embodiments, the pair of sulfur atoms are the sulfur atom corresponding to (e.g., the sulfur atom of) Cys13 of SEQ ID NO: 8 from one E and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys13 of SEQ ID NO: 8 from another E. In some embodiments, when T is 2, the pairs of sulfur atoms are the sulfur atom corresponding to (e.g., the sulfur atom of) Cys10 of SEQ ID NO: 8 from one E and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys10 of SEQ ID NO: 8 from another E and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys13 of SEQ ID NO: 8 from one E and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys13 of SEQ ID NO: 8 from another E.

In some embodiments, the conjugate has the structure:

wherein each of a and b is, independently, 0 or 1 and wherein when a or b is 0, the two sulfur atoms form a disulfide bond. In some embodiments, a is 1 and b is 0. In some embodiments, a is 0 and b is 1. In some embodiments, a and b are 1. In some embodiments of these aspects, the conjugate has the stru

or

.

cts, the conjugate has the structure:

In another aspect, the invention also features a population of conjugates described in the previous two aspects, in which the average value of T is 1 to 5.

In another aspect, the invention features a conjugate described by formula (3):

( )

wherein each M1 includes a first cyclic heptapeptide including a linking nitrogen and each M2 includes a second cyclic heptapeptide including a linking nitrogen; E includes an Fc domain monomer (e.g., an Fc domain monomer having the sequence of any one of SEQ ID NOs: 1-14, 1-29, or 1-31); L’ 1 in each M 2-L’1-M1 is a linker covalently attached to a sulfur atom of a hinge cysteine in E and to the linking nitrogen in each of M1 and M2; T is 1, 2, 3, 4, or 5, the squiggly line connected to the E indicates that each M2-L’ 1 -M1 is covalently attached to a sulfur atom of a hinge cysteine in E, or a pharmaceutically acceptable salt thereof. When T is greater than 1 (e.g., T is 2, 3, 4, or 5), each M2-L’ 1 -M1 may be independently selected (e.g., independently selected from any of the M2-L’ 1 -M1 structures described herein). In some embodiments, each E includes an Fc domain monomer having the sequence of any one of SEQ ID NOs: 1 -14, 1 -29, or 1 -31 .

In some embodiments of these aspects, each E comprises the sequence

MVRSDKTHTCPPCPPC*KC*PAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHE DPEVKFNWYV DGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKA KGQPREPQV YTLPPSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYS KLTVDKSRW

In some embodiments, at least one of the pair of sulfur atoms is the sulfur atom corresponding to (e.g., the sulfur atom of) a hinge cysteine of SEQ ID NO: 10, i.e., Cys1 0, Cys13, Cys16, or Cys18 of SEQ ID NO: 1 0. In some embodiments, the pair of sulfur atoms are the sulfur atoms corresponding to (e.g., the sulfur atoms of) Cys1 0 and Cys13 in SEQ ID NO: 10, Cys10 and Cys16 in SEQ ID NO: 10, Cys 30 and Cys18 in SEQ ID NO: 10, Cys13 and Cys 36 in SEQ ID NO: 10, Cys13 and Cys 38 in SEQ ID NO: 10, and/or Cys 36 and Cys 38 in SEQ ID NO: 10.

In some embodiments, when T is 2, the pair of sulfur atoms are the sulfur atoms corresponding to (e.g., the sulfur atoms of) Cys10 and Cys13 in SEQ ID NO: 10 and Cys 36 and Cys 38 in SEQ ID NO: 10.

In some embodiments, the pair of sulfur atoms include one sulfur atom of a cysteine from each E, i.e., L '1 -M along with the sulfur atoms to which it is attached forms a bridge between two Fc domains (e.g., two Fc domains comprising the sequence of SEQ ID NO: 10). In some embodiments, the pair of sulfur atoms are the sulfur atom corresponding to (e.g., the sulfur atom of) Cys10 of SEQ ID NO: 10 from one E and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys1 0 of SEQ ID NO: 10 from another E. In some embodiments, the pair of sulfur atoms are the sulfur atom corresponding to (e.g., the sulfur atom of) Cys13 of SEQ ID NO: 1 0 from one E and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys13 of SEQ ID NO: 10 from another E. In some embodiments, the pair of sulfur atoms are the sulfur atom corresponding to (e.g., the sulfur atom of) Cys16 of SEQ ID NO: 10 from one E and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys16 of SEQ ID NO: 10 from another E. In some

embodiments, the pair of sulfur atoms are the sulfur atom corresponding to (e.g., the sulfur atom of) Cys18 of SEQ ID NO: 10 from one E and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys18 of SEQ ID NO: 10 from another E.

In some embodiments, when T is 2, the pairs of sulfur atoms are the sulfur atom corresponding to (e.g., the sulfur atom of) Cys10 of SEQ ID NO: 10 from one E and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys10 of SEQ ID NO: 10 from another E and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys13 of SEQ ID NO: 10 from one E and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys13 of SEQ ID NO: 10 from another E. In some embodiments, when T is 2, the pairs of sulfur atoms are the sulfur atom corresponding to (e.g., the sulfur atom of) Cys10 of SEQ ID NO: 10 from one E and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys10 of SEQ ID NO: 10 from another E and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys16 of SEQ ID NO: 1 0 from one E and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys16 of SEQ ID NO: 10 from another E. In some embodiments, when T is 2, the pairs of sulfur atoms are the sulfur atom

corresponding to (e.g., the sulfur atom of) Cys10 of SEQ ID NO: 10 from one E and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys10 of SEQ ID NO: 10 from another E and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys18 of SEQ ID NO: 10 from one E and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys18 of SEQ ID NO: 10 from another E.

In some embodiments, when T is 2, the pairs of sulfur atoms are the sulfur atom corresponding to (e.g., the sulfur atom of) Cys13 of SEQ ID NO: 10 from one E and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys13 of SEQ ID NO: 10 from another E and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys16 of SEQ ID NO: 10 from one E and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys16 of SEQ ID NO: 10 from another E. In some embodiments, when T is 2, the pairs of sulfur atoms are the sulfur atom corresponding to (e.g., the sulfur atom of) Cys13 of SEQ ID NO: 10 from one E and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys13 of SEQ ID NO: 10 from another E and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys18 of SEQ ID NO: 1 0 from one E and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys18 of SEQ ID NO: 10 from another E.

In some embodiments, when T is 2, the pairs of sulfur atoms are the sulfur atom corresponding to (e.g., the sulfur atom of) Cys16 of SEQ ID NO: 10 from one E and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys16 of SEQ ID NO: 10 from another E and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys18 of SEQ ID NO: 10 from one E and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys18 of SEQ ID NO: 10 from another E.

In some embodiments, when T is 3, the pairs of sulfur atoms are the sulfur atom corresponding to (e.g., the sulfur atom of) Cys10 of SEQ ID NO: 10 from one E and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys10 of SEQ ID NO: 10 from another E; the sulfur atom corresponding to (e.g., the sulfur atom of) Cys13 of SEQ ID NO: 10 from one E and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys13 of SEQ ID NO: 10 from another E; and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys16 of SEQ ID NO: 10 from one E and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys1 6 of SEQ ID NO: 1 0 from another E. In some embodiments, when T is 3, the pairs of sulfur atoms are the sulfur atom corresponding to (e.g., the sulfur atom of) Cys10 of SEQ ID NO: 10 from one E and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys1 0 of SEQ ID NO: 10 from another E; the sulfur atom corresponding to (e.g., the sulfur atom of) Cys13 of SEQ ID NO: 1 0 from one E and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys13 of SEQ ID NO: 1 0 from another E; and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys18 of SEQ ID NO: 1 0 from one E and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys18 of SEQ ID NO: 10 from another E. In some embodiments, when T is 3, the pairs of sulfur atoms are the sulfur atom corresponding to (e.g., the sulfur atom of) Cys10 of SEQ ID NO: 10 from one E and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys1 0 of SEQ ID NO: 1 0 from another E; the sulfur atom corresponding to (e.g., the sulfur atom of) Cys18 of SEQ ID NO: 10 from one E and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys18 of SEQ ID NO: 10 from another E; and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys16 of SEQ ID NO: 10 from one E and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys16 of

SEQ ID NO: 10 from another E. In some embodiments, when T is 3, the pairs of sulfur atoms are the sulfur atom corresponding to (e.g., the sulfur atom of) Cys13 of SEQ ID NO: 1 0 from one E and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys13 of SEQ ID NO: 10 fr

corresponding to (e.g., the sulfur atom of) Cys18 of SEQ ID NO: 10 from one E and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys18 of SEQ ID NO: 10 from another E; and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys16 of SEQ ID NO: 10 from one E and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys16 of SEQ ID NO: 10 from another E.

In some embodiments, when T is 3, the pairs of sulfur atoms are the sulfur atom corresponding to (e.g., the sulfur atom of) Cys10 of SEQ ID NO: 10 from one E and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys10 of SEQ ID NO: 10 from another E; the sulfur atom corresponding to (e.g., the sulfur atom of) Cys13 of SEQ ID NO: 10 from one E and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys13 of SEQ ID NO: 10 from another E; the sulfur atom corresponding to (e.g., the sulfur atom of) Cys16 of SEQ ID NO: 10 from one E and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys16 of SEQ ID NO: 10 from another E; and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys18 of SEQ ID NO: 10 from one E and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys18 of SEQ ID NO: 10 from another E.

In some embodiments, the conjugate has the structure:

wherein each of a, b, c, and d is, independently, 0 or 1 and wherein when a, b, c, or d is 0, the two sulfur atoms form a disulfide bond.

In some embodiments, each M-L 1 -M has the structure:

In m m im n h M-L 1 -M h h r r :

.

In some embodiments, a is 1 and b, c, and d are 0. In some embodiments, a and b are 1 and c and d are 0. In some embodiments, a and c are 1 and b and d are 0. In some embodiments, a and d are 1 and b and c are 0. In some embodiments, a, b, and c are 1 and d is 0. In some embodiments, a, b, and d are 1 and c is 0. In some embodiments, a, c, and d are 1 and b is 0. In some embodiments, b and c are 1 and a and d are 0. In some embodiments, b and d are 1 and a and c are 0. In some

embodiments, b, c, and d are 1 and a is 0. In some embodiments, c and d are 1 and a and b are 0. In some embodiments, a, b, c, and d are 1.

I n some embodiments of these aspects, each E comprises the sequence

MVRSDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEV KFNWYVDGVEVH NAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPRE PQVYTLPPSR EEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKS RWQQGNVFS CSVMHEALHNHYTQKSLSLSPGK (SEQ ID NO: 4).

In some embodiments, at least one of the pair of sulfur atoms is the sulfur atom corresponding to (e.g., the sulfur atom of) a hinge cysteine of SEQ ID NO: 4, i.e., Cys10 and/or Cys13. In some embodiments, the pair of sulfur atoms are the sulfur atoms corresponding to (e.g., the sulfur atoms of) Cys10 and Cys13 in SEQ ID NO: 4.

In some embodiments, the pair of sulfur atoms include one sulfur atom of a cysteine from each E, i.e., L ’1 -M along with the sulfur atoms to which it is attached forms a bridge between two Fc domains (e.g., two Fc domains comprising the sequence of SEQ ID NO: 4). In some embodiments, the pair of sulfur atoms are the sulfur atom corresponding to (e.g., the sulfur atom of) Cys10 of SEQ ID NO: 4 from one E and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys10 of SEQ ID NO: 4 from another E. In some embodiments, the pair of sulfur atoms are the sulfur atom corresponding to (e.g., the sulfur atom of) Cys13 of SEQ ID NO: 4 from one E and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys13 of SEQ ID NO: 4 from another E. In some embodiments, when T is 2, the pairs of sulfur atoms are the sulfur atom corresponding to (e.g., the sulfur atom of) Cys10 of SEQ ID NO: 4 from one E and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys10 of SEQ ID NO: 4 from another E and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys13 of SEQ ID NO: 4 fro

corresponding to (e.g., the sulfur atom of) Cys13 of SEQ ID NO: 4 from another E.

In some embodiments the con u ate has the structure:

wherein each of a and b is, independently, 0 or 1 and wherein when a or b is 0, the two sulfur atoms form a disulfide bond. In some embodiments, a is 1 and b is 0. In some embodiments, a is 0 and b is 1. In some embodiments, a and b are 1.

In some embodiments of these aspects, each E comprises the sequence

MVRSDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEV KFNWYVDGVEVH NAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPRE PQVYTLPPSR EEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKS RWQQGNVFS CSVMHEALHNHYTQKSLSLSPGKHHHHHH (SEQ ID NO: 8).

In some embodiments, at least one of the pair of sulfur atoms is the sulfur atom corresponding to (e.g., the sulfur atom of) a hinge cysteine of SEQ ID NO: 8, i.e., Cys10 and/or Cys13. In some embodiments, the pair of sulfur atoms are the sulfur atoms corresponding to (e.g., the sulfur atoms of) Cys10 and Cys13 in SEQ ID NO: 8.

In some embodiments, the pair of sulfur atoms include one sulfur atom of a cysteine from each E, i.e., L ’1 -M along with the sulfur atoms to which it is attached forms a bridge between two Fc domains (e.g., two Fc domains comprising the sequence of SEQ ID NO: 8). In some embodiments, the pair of sulfur atoms are the sulfur atom corresponding to (e.g., the sulfur atom of) Cys10 of SEQ ID NO: 8 from one E and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys10 of SEQ ID NO: 8 from another E. In some embodiments, the pair of sulfur atoms are the sulfur atom corresponding to (e.g., the sulfur atom of) Cys13 of SEQ ID NO: 8 from one E and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys13 of SEQ ID NO: 8 from another E. In some embodiments, when T is 2, the pairs of sulfur atoms are the sulfur atom corresponding to (e.g., the sulfur atom of) Cys10 of SEQ ID NO: 8 from one E and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys10 of SEQ ID NO: 8 from another E and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys13 of SEQ ID NO: 8 from one E and the sulfur atom corresponding to (e.g., the sulfur atom of) Cys13 of SEQ ID NO: 8 from another E. In some embodiments, the conjugate has the structure:

wherein each of a and b is, independently, 0 or 1 and wherein when a or b is 0, the two sulfur atoms form a disulfide bond. In some embodiments, a is 1 and b is 0. In some embodiments, a is 0 and b is 1. In some embodiments, a and b are 1.

In m m im n h M-L 1 -M h h r r :

.

In another aspect, the invention features a conjugate described by formula (4):

(4)

wherein each M includes a cyclic heptapeptide including a linking nitrogen; E includes an Fc domain monomer (e.g., an Fc domain monomer having the sequence of any one of SEQ ID NOs: 1-14, 1-29, or 1-31); L’ 1 in each L’ 1 -M is a linker covalently attached to a sulfur atom of a hinge cysteine in E and to the linking nitrogen in M; T is 1, 2, 3, 4, or 5; the squiggly line connected to E indicates that each L’ 1 -M is covalently attached to the sulfur atom corresponding to (e.g., the sulfur atom of) the hinge cysteine in E, or a pharmaceutically acceptable salt thereof.

In some embodiments, each E includes an Fc domain monomer having the sequence of any one of SEQ ID NOs: 1-14, 1-29, or 1-31.

In some embodiments of these aspects, each E comprises the sequence

M VRSDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPE VKFNWYVDGVEVH NAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPRE PQVYTLPPSR EEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSF

CSVMHEALHNHYTQKSLSLSPGK (SEQ ID NO: 4).

In some embodiments, at least one of the sulfur atoms is the sulfur atom corresponding to (e.g., the sulfur atom of) a hinge cysteine of SEQ ID NO: 4, i.e., Cys10 and/or Cys13. In some embodiments, the sulfur atoms is the sulfur atom corresponding to (e.g, the sulfur atom of) Cys10 in SEQ ID NO: 4. In some embodiments, the sulfur atom corresponding to (e.g, the sulfur atom of) Cys10 in SEQ ID NO: 4.

In some embodiments, the conjugate has the structure:

wherein each of a and b is, independently, 0 or 1 and wherein when a or b is 0, the sulfur atoms is a thiol. In some embodiments, a is 1 and b is 0. In some embodiments, a is 0 and b is 1. In some embodiments, a and b are 1.

In some embodiments, the conjugate has the structure:

.

In m m im n h nugate has the structure:

.

In another aspect, the invention features a population of conjugates described in the previous two aspects, wherein the average value of T is 1 to 5. In some embodiments, the average value of T is 1 to 2.

In another aspect, the invention features a conjugate described by formula (3):

(3) wherein each M1 includes a first cyclic heptapeptide including a linking nitro

second cyclic heptapeptide including a linking nitrogen; E includes an Fc domain monomer (e.g., an Fc domain monomer having the sequence of any one of SEQ ID NOs: 1-14, 1-29, or 1-31); L’ 1 in each M2- L’ 1 -M1 is a linker covalently attached to a nitrogen atom of a surface exposed lysine in E and to the linking nitrogen in each of M1 and M2; T is 1, 2, 3, 4, or 5, the squiggly line connected to the E indicates that each M2-L’ 1 -M1 is covalently attached to the nitrogen atom of a surface exposed lysine in E, or a pharmaceutically acceptable salt thereof.

In some embodiments, each E includes an Fc domain monomer having the sequence of any one of SEQ ID NOs: 1-14, 1-29, or 1-31.

In some embodiments of these aspects, each E comprises the sequence

MVRSDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEV KFNWYVDGVEVH NAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPRE PQVYTLPPSR EEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKS RWQQGNVFS CSVMHEALHNHYTQKSLSLSPGK (SEQ ID NO: 4).

In some embodiments, at least one of the sulfur atoms is the sulfur atom of a hinge cysteine of SEQ ID NO: 4, i.e., Cys10 and/or Cys13. In some embodiments, the sulfur atom is sulfur atom comrresponding to (e.g., the sulfur atom of) Cys10 in SEQ ID NO: 4. In some embodiments, the sulfur atom is the sulfur atom corresponding to (e.g., the sulfur atom of) Cys10 in SEQ ID NO: 4.

In some embodiments, the conjugate has the structure:

wherein each of a and b is, independently, 0 or 1 and wherein when a or b is 0, the sulfur atoms is a thiol. In some embodiments, a is 1 and b is 0. In some embodiments, a is 0 and b is 1. In some embodiments, a and b are 1.

In m m im n M-L ’1 -M h h r r :

. In another aspect, the invention features, a conjugate described by formula (4):

wherein each M includes a cyclic heptapeptide including a linking nitrogen; E includes an Fc domain monomer (e.g., an Fc domain monomer having the sequence of any one of SEQ ID NOs: 1-14, 1-29, or 1-31); L’ 1 in each L’ 1 -M is a linker covalently attached to a nitrogen atom of a surface exposed lysine in E and to the linking nitrogen in M; T is an integer from 1 to 11, the squiggly line connected to E indicates that each L’ 1 -M is covalently attached to the nitrogen atom of a surface exposed lysine in E, or a pharmaceutically acceptable salt thereof.

In some embodiments, each E includes an Fc domain monomer having the sequence of any one of SEQ ID NOs: 1-14, 1-29, or 1-31.

In some embodiments, E includes the sequence of

M VRSDKTHTCPPCPPC*KC*PAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVD VSHEDPEVKFNWYV DGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKA KGQPREPQV YTLPPSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYS KLTVDKSRW QQGNVFSCSVMHEALHNHYTQKSLSLSPGKHHHHHH (SEQ ID NO: 10)

In some embodiments of the previous two aspects, the nitrogen atom is the nitrogen of a surface exposed lysine, e.g., the nitrogen atom corresponding to (e.g, the nitrogen atom of) Lys35, Lys63, Lys77, Lys79, Lys106, Lys123, Lys129, Lys181, Lys203, Lys228, or Lys236 of SEQ ID NO: 10. In some embodiments, the nitrogen atom is the nitrogen atom corresponding to (e.g

Lys79, Lys108, Lys230, and/or Lys238 of SEQ ID NO:10.

In some embodiments, the conjugate has the structure:

wherein each of a, b, c, d, and e is, independently, 0 or 1 and wherein when a, b, c, d, or e is 0, the two nitrogen atom is NH2. In some embodiments, a is 1 and b, c, d, and e are 0. In some embodiments, b is 1 and a, c, d, and e are 0. In some embodiments, c is 1 and a, b, d, and e are 0. In some embodiments, d is 1 and a, b, c, and e are 0. In some embodiments, e is 1 and a, b, c, and d are 0. In some embodiments, a and b are 1 and c, d, and e are 0. In some embodiments, a and c are 1 and b, d, and e are 0. In some embodiments, a and d are 1 and b, c, and e are 0. In some embodiments, a and e are 1 and b, c, and d are 0. In some embodiments, b and c are 1 and a, d, and e are 0. In some embodiments, b and d are 1 and a, c, and e are 0. In some embodiments, b and e are 1 and a, c, and d are 0. In some embodiments, c and d are 1 and a, b, and e are 0. In some embodiments, c and e are 1 and a, b, and d are 0. In some embodiments, d and e are 1 and a, b, and c are 0. In some embodiments, a, b, and c are 1 and d and e are 0. In some embodiments, a, b, and d are 1 and c and e are 0. In some embodiments, a, b, and e are 1 and c and d are 0. In some embodiments, a, c, and d are 1 and b and e are 0. In some embodiments, a, c, and e are 1 and b and d are 0. In some embodiments, a, d, and e are 1 and b and c are 0. In some embodiments, b, c, and d are 1 and a and e are 0. In some embodiments, b, d, and e are 1 and a and c are 0. In some embodiments, c, d, and e are 1 and a and b are 0.

In some embodiments, the conjugate has the structure

. In m m im n h njugate has the structure

In another aspect, the invention features a population of conjugates described in the previous two aspects, wherein the average value of T is 1 to 5. In some embodiments, the average value of T is 1 to 2.

In some embodiments of the conjugates described herein, the conjugate forms a homodimer including an Fc domain.

In some embodiments of the conjugates described herein, E homodimerizes with another E to form an Fc domain.

In another aspect, the invention features a conjugate including (i) a first cyclic heptapeptide; (ii) a second cyclic heptapeptide; (iii) an Fc domain monomer or an Fc domain; and (iv) a linker covalently attached to the first cyclic heptapeptide, the second cyclic heptapeptide, and the Fc domain monomer or the Fc domain.

In another aspect, the inventi n f r n ri ed by formula (D-Ia):

( )

wherein each M1 includes a first cyclic heptapeptide including a linking nitrogen and each M2 includes a second cyclic heptapeptide including a linking nitrogen; L’ in each M2-L’-M1 is a linker covalently attached to the Fc domain monomer and to the linking nitrogen in each of M1 and M2; each E is an Fc domain monomer; n is 1 or 2; T is an integer from 1 to 20, or a pharmaceutically acceptable salt thereof. When T is greater than 1 (e.g., T is 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20), each M2-L’- M1 may be independently selected (e.g., independently selected from any of the M2-L’-M1 structures described herein).

In some embodiments, E includes the sequence of

MVRSDKTHTCPPCPPC*KC*PAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDV SHEDPEVKFNWYV DGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKA KGQPREPQV YTLPPSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYS KLTVDKSRW QQGNVFSCSVMHEALHNHYTQKSLSLSPGKHHHHHH (SEQ ID NO: 10)

In some embodiments of the previous two aspects, the nitrogen atom is the nitrogen of a surface exposed lysine, e.g., the nitrogen atom corresponding to (e.g., the nitrogen atom of) Lys35, Lys63, Lys77, Lys79, Lys106, Lys123, Lys129, Lys181, Lys203, Lys228, or Lys236 of SEQ ID NO: 10. In some embodiments, the nitrogen atom is the nitrogen atom corresponding to (e.g

Lys65, Lys79, Lys108, Lys230, and/or Lys238 of SEQ ID NO:10.

In some embodiments, the conjugate has the structure:

wherein each of a, b, c, d, and e is, independently, 0 or 1 and wherein when a, b, c, d, or e is 0, the two nitrogen atom is NH2. In some embodiments, a is 1 and b, c, d, and e are 0. In some

embodiments, b is 1 and a, c, d, and e are 0. In some embodiments, c is 1 and a, b, d, and e are 0. In some embodiments, d is 1 and a, b, c, and e are 0. In some embodiments, e is 1 and a, b, c, and d are 0. In some embodiments, a and b are 1 and c, d, and e are 0. In some embodiments, a and c are 1 and b, d, and e are 0. In some embodiments, a and d are 1 and b, c, and e are 0. In some embodiments, a and e are 1 and b, c, and d are 0. In some embodiments, b and c are 1 and a, d, and e are 0. In some embodiments, b and d are 1 and a, c, and e are 0. In some embodiments, b and e are 1 and a, c, and d are 0. In some embodiments, c and d are 1 and a, b, and e are 0. In some embodiments, c and e are 1 and a, b, and d are 0. In some embodiments, d and e are 1 and a, b, and c are 0. In some embodiments, a, b, and c are 1 and d and e are 0. In some embodiments, a, b, and d are 1 and c and e are 0. In some embodiments, a, b, and e are 1 and c and d are 0. In some embodiments, a, c, and d are 1 and b and e are 0. In some embodiments, a, c, and e are 1 and b and d are 0. In some embodiments, a, d, and e are 1 and b and c are 0. In some embodiments, b, c, and d are 1 and a and e are 0. In some embodiments, b, d, and e are 1 and a and c are 0. In some embodiments, c, d, and e are 1 and a and b are 0.

In s m m im n L ’1 -M h h r r :

. In some embodiments of this aspect, when n is 2, E dimerizes to form an Fc domain, and the conjugate is described by formula (D-Ib):

(D-Ib)

wherein J is an Fc domain; and T is an integer from 2 to 20, or a pharmaceutically acceptable salt thereof.

In some embodiments of this aspect, L’ in each M2-L’-M1 is described by formula (D-L):

(D-L)

wherein L is a remainder of L’; A1 is a 1-5 amino acid peptide covalently attached to the linking nitrogen in each M1 or is absent; and A2 is a 1-5 amino acid peptide covalently attached to the linking nitrogen in each M2 or is absent. In some embodiments of this aspect, the conjugate is described by

(D-II)

wherein L is a remainder of L’; each of R 1 , R 12 , R’ 1 , and R’ 12 is, independently, a lipophilic moiety, a polar moiety, or H; each of R 11 , R 13 , R 14 , R’ 11 , R’ 13 , and R’ 14 is, independently, optionally substituted C1-C5 alkamino, a polar moiety, a positively charged moiety, or H; each of R 15 and R’ 15 is, independently, a lipophilic moiety or a polar moiety; each of R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , R 8 , R 9 , R 10 , R’ 2 , R’ 3 , R’ 4 , R’ 5 , R’ 6 , R’ 7 , R’ 8 , R’ 9 , and R’ 10 is, independently, a lipophilic moiety, a positively charged moiety, a polar moiety, H, optionally substituted C1-C5 alkamino, optionally substituted C1-C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1-C20 heteroalkyl, optionally substituted C3- C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl; or two R groups on the same or adjacent atoms join to form an optionally substituted 5-8 membered ring; each of a’, b’, c’, a, b, and c is, independently, 0 or 1; each of N 1 , N 2 , N 3 , N 4 , N’ 1 , N’ 2 , N’ 3 , and N’ 4 is a nitrogen atom; each of C 1 , C 2 , C 3 , C’ 1 , C’ 2 , and C’ 3 is a carbon atom, or a pharmaceutically acceptable salt thereof.

In some embodiments, the conjugate includes at least one optionally substituted 5-8 membered ring formed by joining (i) R 2 , R 3 , and C 1 ; (ii) R 3 , R 4 , N 1 , and C 1 ; (iii) R 5 , R 6 , and C 2 ; (iv) R 6 , R 7 , N 2 , and C 2 ; (v) R 8 , R 9 , and C 3 ; (vi) R 9 , R 10 , N 3 , and C 3 ; (vii) R’ 2 , R’ 3 , and C’ 1 ; (viii) R’ 3 , R’ 4 , N’ 1 , and C’ 1 ; (ix) R’ 5 , R’ 6 , and C’ 2 ; (x) R’ 6 , R’ 7 , N’ 2 , and C’ 2 ; (xi) R’ 8 , R’ 9 , and C’ 3 ; or (xii) R’ 9 , R’ 10 , N’ 3 , and C’ 3 .

In some embodiments, the conjugate is described by formula (D-III

(D-III)

or a pharmaceutically acceptable salt thereof.

In some embodiments, each of R 1 , R 12 , R’ 1 , and R’ 12 is, independently, a lipophilic moiety; each of R 11 , R 13 , R 14 , R’ 11 , R’ 13 , and R’ 14 is, independently, optionally substituted C1-C5 alkamino, a polar moiety, or a positively charged moiety; and/or each of R 15 and R’ 15 is, independently, a polar moiety.

In some embodiments, each of R 1 and R 12 is a lipophilic moiety. In some embodiments, each of R’ 1 and R’ 12 is a lipophilic moiety. In some embodiments, each lipophilic moiety is, independently, optionally substituted C1-C20 alkyl, optionally substituted C5-C15 aryl, optionally substituted C6-C35 alkaryl, or optionally C5-C10 substituted heteroaryl. In some embodiments, each lipophilic moiety is, independently, C1-C8 alkyl, methyl substituted C2-C4 alkyl, (C1-C10)alkylene(C6)aryl, phenyl substituted (C1-C10)alkylene(C6)aryl, or alkyl substituted C4-C9 heteroaryl. In some embodiments, each lipophilic moiety is, independently, benzyl, isobutyl, sec-butyl, isopropyl, n-propyl, methyl, biphenylmethyl, n-octyl, or methyl substituted indolyl.

In some embodiments, each of R 11 , R 13 , R 14 , R’ 11 , R’ 13 , and R’ 14 is independently optionally substituted C1-C5 alkamino (e.g., CH2CH2NH2).

In some embodiments, each of R 15 and R’ 15 is a polar moiety. In some embodiments, each polar moiety includes a hydroxyl group, a carboxylic acid group, an ester group, or an amide group. In some embodiments, each polar moiety is hydroxyl substituted C1-C4 alkyl. In some embodiments, each polar moiety is CHCH3OH.

In some embodiments of this aspect, the conjugate is described by

(D-IV)

wherein each of R’ 1 and R 1 is, independently, benzyl or CH2CH(CH3)2, or a pharmaceutically acceptable salt thereof.

In some embodiments, the conjugate is described by formula (D-IV-1):

(D-IV-1)

wherein each of R’ 1 and R 1 is, independently, benzyl or CH2CH(CH3)2, or a pharmaceutically acceptable salt thereof.

In some embodiments, the conjugate is described by formula (D-IV-2):

(D-IV-2) wherein each of R’ 1 and R 1 is, independently, benzyl or CH 2 CH(CH 3 ) 2 , or a

salt thereof.

In some embodiments, the conjugate is described by formula (D-IV-3):

(D-IV-3)

wherein each of R’ 1 and R 1 is, independently, benzyl or CH2CH(CH3)2; each of R’ 16 and R 16 is, independently, a lipophilic moiety, optionally substituted C1-C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1-C20 heteroalkyl, optionally substituted C3- C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl, or a pharmaceutically acceptable salt thereof.

In some embodiments, each of R’ 16 and R 16 is, independently, C1-C6 alkyl or benzyl.

In some embodiments the con u ate is described b formula D-V :

(D-V)

wherein each of R’ 1 and R 1 is, independently, benzyl or CH2CH(CH3)2; each of R 2 , R 6 , R’ 2 , and R’ 6 is, independently, a positively charged moiety, a polar moiety, optionally substituted C1-C5 alkamino, optionally substituted C1-C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4- C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1-C20 heteroalkyl, optionally substituted C3-C20 he

substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl, or a pharmaceutically acceptable salt thereof.

I n some embodiments, the conjugate is described by formula (D-V-1):

(D-V-1)

wherein each of R’ 1 and R 1 is, independently, benzyl or CH2CH(CH3)2, or a pharmaceutically acceptable salt thereof.

I n some embodiments, the conjugate is described by formula (D-V-2):

(D-V-2)

wherein each of R’ 1 and R 1 is, independently, benzyl or CH2CH(CH3)2, or a pharmaceutically acceptable salt thereof.

In some embodiments, the conjugate is described by formula (D-V-3):

(D-V-3)

wherein each of R’ 1 and R 1 is, independently, benzyl or CH2CH(CH3)2, or a pharmaceutically acceptable salt thereof.

In some embodiments, the conjugate is described by formula (D-V-4):

(D-V-4)

wherein each of R’ 1 and R 1 is, independently, benzyl or CH2CH(CH3)2, or a pharmaceutically acceptable salt thereof.

In some embodiments, the conjugate is described by formula (D-V-5):

(D-V-5)

wherein each of R’ 1 and R 1 is, independently, benzyl or CH2CH(CH3)2, or a pharmaceutically acceptable salt thereof.

In some embodiments, the conjugate is described by formula (D-V-6):

(D-V-6) wherein each of R’ 1 and R 1 is, independently, benzyl or CH 2 CH(CH 3 ) 2 ; each

independently, a lipophilic moiety, optionally substituted C1-C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1-C20 heteroalkyl, optionally substituted C3- C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl, or a pharmaceutically acceptable salt thereof.

In some embodiments, the conjugate is described by formula (D-V-7):

wherein each of R’ 1 and R 1 is, independently, benzyl or CH2CH(CH3)2; each of R’ 16 and R 16 is, independently, a lipophilic moiety, optionally substituted C1-C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1-C20 heteroalkyl, optionally substituted C3- C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl, or a pharmaceutically acceptable salt thereof.

In m m im n h n i ri f rm l D-V- :

(D-V-8)

wherein each of R’ 1 and R 1 is, independently, benzyl or CH2CH(CH3)2; each of R’ 16 and R 16 is, independently, a lipophilic moiety, optionally substituted C1-C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, o ptionally substituted C5-C15 aryl, optionally substituted C1-C20 heteroalkyl, optionally substituted C3- C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl, or a pharmaceutically acceptable salt thereof.

In some embodiments, the conjugate is described by formula (D-V-9):

wherein each of R’ 1 and R 1 is, independently, benzyl or CH2CH(CH3)2; each of R’ 16 and R 16 is, independently, a lipophilic moiety, optionally substituted C1-C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, o ptionally substituted C5-C15 aryl, optionally substituted C1-C20 heteroalkyl, optionally substituted C3- C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl, or a pharmaceutically acceptable salt thereof.

In some embodiments, the conjugate is described by formula (D-V-10):

20 (D-V-10)

wherein each of R’ 1 and R 1 is, independently, benzyl or CH2CH(CH3)2; each of R’ 16 and R 16 is, independently, a lipophilic moiety, optionally substituted C1-C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1-C20 heteroalkyl, optionally substituted C3- C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl, or a pharmaceutically acceptable salt thereof.

In some embodiments, each of R’ 16 and R 16 is, independently, C1-C6 alkyl or benzyl.

In some embodiments of this as ect the conu ate is described b formula D-VI :

(D-VI)

wherein each of R’ 1 and R 1 is, independently, benzyl or CH2CH(CH3)2; each of R 2 , R 6 , R 8 , R’ 2 , R’ 6 , and R’ 8 is, independently, a positively charged moiety, a polar moiety, optionally substituted C1-C5 alkamino, optionally substituted C1-C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4- C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1-C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl, or a pharmaceutically acceptable salt thereof.

In m m im n h n i ri f rm l D-VI-1 :

(D-VI-1) wherein each of R’ 1 and R 1 is, independently, benzyl or CH 2 CH(CH 3 ) 2 ; or a

salt thereof.

In some embodiments, the conjugate is described by formula (D-VI-2):

wherein each of R’ 1 and R 1 is, independently, benzyl or CH2CH(CH3)2; or a pharmaceutically acceptable salt thereof.

In some embodiments the con u ate is described b formula D-VI-2a :

wherein each of R’ 1 and R 1 is, independently, benzyl or CH2CH(CH3)2; or a pharmaceutically acceptable salt thereof.

In some embodiments, the conjugate is described by formula (D-VI-2b):

(D-VI-2b)

wherein each of R’ 1 and R 1 is, independently, benzyl or CH2CH(CH3)2; or a pharmaceutically acceptable salt thereof.

In some embodiments, the conjugate is described by formula (D-VI-2c):

wherein each of R’ 1 and R 1 is, independently, benzyl or CH2CH(CH3)2; or a pharmaceutically acceptable salt thereof.

In some embodiments, the conjugate is described by formula (D-VI-2d):

wherein each of R’ 1 and R 1 is, independently, benzyl or CH2CH(CH3)2; or a pharmaceutically acceptable salt thereof.

In some embodiments, the conjugate is described by formula (D-VI-2e):

wherein each of R’ 1 and R 1 is, independently, benzyl or CH2CH(CH3)2; or a pharmaceutically acceptable salt thereof.

In some embodiments, the conjugate is described by formula (D-VI-2f):

(D-VI-2f)

wherein each of R’ 1 and R 1 is, independently, benzyl or CH2CH(CH3)2; or a pharmaceutically acceptable salt thereof.

In some embodiments, the conjugate is described by formula (D-VI-3):

wherein each of R’ 1 and R 1 is, independently, benzyl or CH2CH(CH3)2; or a pharmaceutically acceptable salt thereof.

In some embodiments, the conjugate is described by formula (D-VI-4):

(D-VI-4)

wherein each of R’ 1 and R 1 is, independently, benzyl or CH2CH(CH3)2; or a pharmaceutically acceptable salt thereof.

In m m im n h n i ri f rm l D-VI- :

(D-VI-5)

wherein each of R’ 1 and R 1 is, independently, benzyl or CH2CH(CH3)2; or a pharmaceutically acceptable salt thereof.

I n some embodiments, the conjugate is described by formula (D-VI-6):

(D-VI-6)

wherein each of R’ 1 and R 1 is, independently, benzyl or CH2CH(CH3)2; each of R’ 16 and R 16 is, independently, a lipophilic moiety, optionally substituted C1-C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1-C20 heteroalkyl, optionally substituted C3- C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl, or a pharmaceutically acceptable salt thereof.

In some embodiments, the conjugate is described by formula (D-VI-6a):

(D-VI-6a)

wherein each of R’ 1 and R 1 is, independently, benzyl or CH2CH(CH3)2; or a pharmaceutically acceptable salt thereof.

In some embodiments, the conjugate is described by formula (D-VI-6b):

(D-VI-6b) wherein each of R’ 1 and R 1 is, independently, benzyl or CH 2 CH(CH 3 ) 2 ; or a

salt thereof.

In some embodiments, the conjugate is described by formula (D-VI-6c):

( )

wherein each of R’ 1 and R 1 is, independently, benzyl or CH2CH(CH3)2; or a pharmaceutically acceptable salt thereof.

In some embodiments, the conjugate is described by formula (D-VI-6d):

wherein each of R’ 1 and R 1 is, independently, benzyl or CH2CH(CH3)2; or a pharmaceutically acceptable salt thereof. In some embodiments the con u ate is described b formula D-VI

(D-VI-7)

wherein each of R’ 1 and R 1 is, independently, benzyl or CH2CH(CH3)2; each of R’ 16 and R 16 is, independently, a lipophilic moiety, optionally substituted C1-C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1-C20 heteroalkyl, optionally substituted C3- C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl, or a pharmaceutically acceptable salt thereof.

In m m im n h n i ri f rm l D-VI- :

(D-VI-8)

wherein each of R’ 1 and R 1 is, independently, benzyl or CH2CH(CH3)2; each of R’ 16 and R 16 is, independently, a lipophilic moiety, optionally substituted C1-C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1-C20 heteroalkyl, optionally substituted C3- C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl, or a pharmaceutically acceptable salt thereof. In some embodiments the con u ate is described b formula D-VI

(D-VI-9)

wherein each of R’ 1 and R 1 is, independently, benzyl or CH2CH(CH3)2; each of R’ 16 and R 16 is, independently, a lipophilic moiety, optionally substituted C1-C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1-C20 heteroalkyl, optionally substituted C3- C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl, or a pharmaceutically acceptable salt thereof.

In m m im n h n i ri f rm l D-VI-1 :

(D-VI-10)

wherein each of R’ 1 and R 1 is, independently, benzyl or CH2CH(CH3)2; each of R’ 16 and R 16 is, independently, a lipophilic moiety, optionally substituted C1-C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1-C20 heteroalkyl, optionally substituted C3- C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl, or a pharmaceutically acceptable salt thereof. In some embodiments, each of R’ 16 and R 16 is, independently, C1-C

In some embodiments of this aspect, the conjugate is described by formula (D-VII):

(D-VII)

wherein each of R’ 1 and R 1 is, independently, benzyl or CH2CH(CH3)2; each of R 2 , R 6 , R 8 , R’ 2 , and R’ 6 is, independently, a positively charged moiety, a polar moiety, optionally substituted C1-C5 alkamino, optionally substituted C1-C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4- C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1-C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl, or a pharmaceutically acceptable salt thereof.

In some embodiments of this as ect the con u ate is described b formula D-VIII :

(D-VIII)

wherein each of R’ 1 and R 1 is, independently, benzyl or CH2CH(CH3)2; each of R 2 , R 6 , R 8 , and R’ 2 is, independently, a positively charged moiety, a polar moiety, optionally substituted C1-C5 alkamino, optionally substituted C1-C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4- C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1 C20 heteroalkyl optionally substituted C3 C20 heterocycloalkyl optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heter

substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl, or a pharmaceutically acceptable salt thereof.

In some embodiments of this aspect, the conjugate is described by formula (D-IX):

(D-IX)

wherein each of R’ 1 and R 1 is, independently, benzyl or CH2CH(CH3)2; each of R 2 , R 6 , and R’ 2 is, independently, a positively charged moiety, a polar moiety, optionally substituted C1-C5 alkamino, optionally substituted C1-C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4- C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1-C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl, or a pharmaceutically acceptable salt thereof.

In some embodiments the conu ate is described b formula D-IX-1 :

(D-IX-1)

wherein each of R’ 1 and R 1 is, independently, benzyl or CH2CH(CH3)2; or a pharmaceutically acceptable salt thereof. In some embodiments, the conjugate is described by formula (D-IX

(D-IX-2)

wherein each of R’ 1 and R 1 is, independently, benzyl or CH2CH(CH3)2; or a pharmaceutically acceptable salt thereof.

In some embodiments, the conjugate is described by formula (D-IX-3):

(D-IX-3)

wherein each of R’ 1 and R 1 is, independently, benzyl or CH2CH(CH3)2; each of R’ 16 and R 16 is, independently, a lipophilic moiety, optionally substituted C1-C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1-C20 heteroalkyl, optionally substituted C3- C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl, or a pharmaceutically acceptable salt thereof. In some embodiments, the conjugate is described by formula (D-IX

(D-IX-4)

wherein each of R’ 1 and R 1 is, independently, benzyl or CH2CH(CH3)2; each of R’ 16 and R 16 is, independently, a lipophilic moiety, optionally substituted C1-C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1-C20 heteroalkyl, optionally substituted C3- C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl, or a pharmaceutically acceptable salt thereof.

In some embodiments, each of R’ 16 and R 16 is, independently, C1-C6 alkyl or benzyl.

In some embodiments of this aspect, the conjugate is described by formula (D-X):

(D-X)

w herein each of R’1 and R1 is, independently, benzyl or CH2CH(CH3)2; each of R2 and R’2 is, independently, a positively charged moiety, a polar moiety, optionally substituted C1-C5 alkamino, optionally substituted C1-C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4- C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1-C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or opt

heteroalkaryl, or a pharmaceutically acceptable salt thereof.

In some embodiments of this aspect, the conjugate is described by formula (D-XI):

(D-XI)

wherein each of R’ 1 and R 1 is, independently, benzyl or CH2CH(CH3)2; each of R 2 and R 6 is, independently, a positively charged moiety, a polar moiety, optionally substituted C1-C5 alkamino, optionally substituted C1-C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4- C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1-C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl, or a pharmaceutically acceptable salt thereof.

In some embodiments the con u ate is described b formula D-XI-1 :

(D-XI-1)

wherein each of R’ 1 and R 1 is, independently, benzyl or CH2CH(CH3)2; or a pharmaceutically acceptable salt thereof. In some embodiments, the conjugate is described by formula (D-XI

(D-XI-2)

wherein each of R’ 1 and R 1 is, independently, benzyl or CH2CH(CH3)2; or a pharmaceutically acceptable salt thereof.

In some embodiments, the conjugate is described by formula (D-XI-3):

(D-XI-3)

wherein each of R’ 1 and R 1 is, independently, benzyl or CH2CH(CH3)2; R 16 is a lipophilic moiety, optionally substituted C1-C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1-C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4- C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl, or a pharmaceutically acceptable salt thereof. In some embodiments, the conjugate is described by formula (D-XI

(D-XI-4)

wherein each of R’ 1 and R 1 is, independently, benzyl or CH2CH(CH3)2; R 16 is a lipophilic moiety, optionally substituted C1-C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1-C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4- C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl, or a pharmaceutically acceptable salt thereof.

In some embodiments, R 16 is C1-C6 alkyl or benzyl.

In m m im n f hi h n i ri f rm l D-XII :

( D-XII)

wherein each of R’ 1 and R 1 is, independently, benzyl or CH2CH(CH3)2; each of R 2 and R’ 2 is,

independently, a positively charged moiety, a polar moiety, optionally substituted C1-C5 alkamino, optionally substituted C1-C20 alkyl, optionally substituted C3-C20 cycloalky

C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1-C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl; R 6 , R 7 , N 2 , and C 2 together form an optionally substituted 5-8 membered ring including optionally substituted C3-C7 heterocycloalkyl including an N heteroatom and additional 0-2 heteroatoms independently selected from N, O, and S, or optionally substituted C2-C7 heteroaryl including an N heteroatom and additional 0-2 heteroatoms independently selected from N, O, and S; R’ 6 , R’ 7 , N’ 2 , and C’ 2 together form an optionally substituted 5-8 membered ring including optionally substituted C3-C7 heterocycloalkyl including an N heteroatom and additional 0-2 heteroatoms independently selected from N, O, and S, or optionally substituted C2-C7 heteroaryl including an N heteroatom and additional 0-2 heteroatoms independently selected from N, O, and S; or a pharmaceutically acceptable salt thereof.

In some embodiments, the conjugate is described by formula (D-XII-1):

(D-XII-1)

wherein each of R’ 1 and R 1 is, independently, benzyl or CH2CH(CH3)2; or a pharmaceutically acceptable salt thereof.

In some embodiments of this aspect, the conjugate is described by

(D-XIII)

wherein each of R’ 1 and R 1 is, independently, benzyl or CH2CH(CH3)2, or a pharmaceutically acceptable salt thereof.

In some embodiments of this aspect, the conjugate is described by formula (D-XIV):

(D-XIV)

wherein each A1 is a 1-5 amino acid peptide covalently attached to the linking nitrogen in each M1; each A2 is a 1-5 amino acid peptide covalently attached to the linking nitrogen in each M2, or is absent; each of L and L 1 is a remainder of L’; or a pharmaceutically acceptable salt thereof.

In some embodiments, R 2 is optionally substituted C1-C5 alkamino. In some embodiments, R’ 2 is optionally substituted C1-C5 alkamino (e.g., CH2NH2 or CH2CH2NH2).

In some embodiments, R 2 is a polar moiety. In some embodiments, R’ 2 is a polar moiety. In some embodiments, R 6 is a polar moiety. In some embodiments, R’ 6 is a polar moiety. In some embodiments, the polar moiety includes a hydroxyl group, a carboxylic acid group, an ester group, or an amide group. In some embodiments, the polar moiety is hydroxyl substituted C1-C4 alkyl. In some embodiments, the polar moiety is CHCH3OH or CH2OH.

In some embodiments, R 8 is optionally substituted C1-C5 alkamino. In some embodiments, R’ 8 is optionally substituted C1-C5 alkamino (e.g., CH2NH2 or CH2CH2NH2). In some embodiments, R8 is optionally substituted C5-C15 aryl. In some embodiments, R’ 8 is optionally substituted C5-C15 aryl. In some embodiments, the optionally substituted C5-C15 aryl is naphthyl.

In some embodiments, R 6 , R 7 , N 2 , and C 2 together form a 5- or 6-membered ring including C4-C5 heterocycloalkyl including an N heteroatom and additional 0 or 1 heteroatom independently selected from N, O, and S; and wherein R’ 6 , R’ 7 , N’ 2 , and C’ 2 together form a 5- or 6-mem

heterocycloalkyl including an N heteroatom and additional 0 or 1 heteroatom independently selected from N, O, and S.

In some embodiments, L’, L, or L 1 includes one or more optionally substituted C1-C20 alkylene, optionally substituted C1-C20 heteroalkylene, optionally substituted C2-C20 alkenylene, optionally substituted C2-C20 heteroalkenylene, optionally substituted C2-C20 alkynylene, optionally substituted C2-C20 heteroalkynylene, optionally substituted C3-C20 cycloalkylene, optionally substituted C3-C20 heterocycloalkylene, optionally substituted C4-C20 cycloalkenylene, optionally substituted C4-C20 heterocycloalkenylene, optionally substituted C8-C20 cycloalkynylene, optionally substituted C8-C20 heterocycloalkynylene, optionally substituted C5-C15 arylene, optionally substituted C2-C15

heteroarylene, O, S, NR i , P, carbonyl, thiocarbonyl, sulfonyl, phosphate, phosphoryl, or imino, wherein R i is H, optionally substituted C1-C20 alkyl, optionally substituted C1-C20 heteroalkyl, optionally substituted C2-C20 alkenyl, optionally substituted C2-C20 heteroalkenyl, optionally substituted C2-C20 alkynyl, optionally substituted C2-C20 heteroalkynyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C5-C15 aryl, or optionally substituted C2-C15 heteroaryl.

In some embodiments, the backbone of L’, L, or L 1 consists of one or more optionally substituted C1-C20 alkylene, optionally substituted C1-C20 heteroalkylene, optionally substituted C2-C20 alkenylene, optionally substituted C2-C20 heteroalkenylene, optionally substituted C2-C20 alkynylene, optionally substituted C2-C20 heteroalkynylene, optionally substituted C3-C20 cycloalkylene, optionally substituted C3-C20 heterocycloalkylene, optionally substituted C4-C20 cycloalkenylene, optionally substituted C4- C20 heterocycloalkenylene, optionally substituted C8-C20 cycloalkynylene, optionally substituted C8-C20 heterocycloalkynylene, optionally substituted C5-C15 arylene, optionally substituted C2-C15

h eteroarylene, O, S, NRi, P, carbonyl, thiocarbonyl, sulfonyl, phosphate, phosphoryl, or imino, wherein Ri is H, optionally substituted C1-C20 alkyl, optionally substituted C1-C20 heteroalkyl, optionally substituted C2-C20 alkenyl, optionally substituted C2-C20 heteroalkenyl, optionally substituted C2-C20 alkynyl, optionally substituted C2-C20 heteroalkynyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C5-C15 aryl, or optionally substituted C2-C15 heteroaryl.

In some embodiments, L’, L, or L 1 is oxo substituted. In some embodiments, the backbone of L’, L, or L 1 includes no more than 250 atoms. In some embodiments, L’, L, or L 1 is capable of forming an amide, a carbamate, a sulfonyl, or a urea linkage. In some embodiments, L or L 1 is a bond.

In some embodiments, each L is described by formula (D-L-I):

(D-L-I) wherein L A is described by formula

G A2 ; L B is described by formula

L C is described by

is a bond attached to Q in formula (L-I); G A2 is a bond attached to A1 or M1 if A1 is absent; G B1 is a bond attached to Q in formula (L-I); GB2 is a bond attached to A2 or M2 if A2 is absent; GC1 is a bond attached to Q in formula (L-I); G C2 is a bond attached to E; each of Z A1 , Z A2 , Z A3 , Z A4 , Z A5 , Z B1 , Z B2 , Z B3 , Z B4 , Z B5 , Z C1 , Z C2 , Z C3 , Z C4 , and Z C5 is, independently, optionally substituted C1-C20 alkylene, optionally substituted C1- C20 heteroalkylene, optionally substituted C2-C20 alkenylene, optionally substituted C2-C20

heteroalkenylene, optionally substituted C2-C20 alkynylene, optionally substituted C2-C20

heteroalkynylene, optionally substituted C3-C20 cycloalkylene, optionally substituted C3-C20

heterocycloalkylene, optionally substituted C4-C20 cycloalkenylene, optionally substituted C4-C20 heterocycloalkenylene, optionally substituted C8-C20 cycloalkynylene, optionally substituted C8-C20 heterocycloalkynylene, optionally substituted C5-C15 arylene, or optionally substituted C2-C15 heteroarylene; each of Y A1 , Y A2 , Y A3 , Y A4 , Y B1 , Y B2 , Y B3 , Y B4 , Y C1 , Y C2 , Y C3 , and Y C4 is, independently, O, S, NR i , P, carbonyl, thiocarbonyl, sulfonyl, phosphate, phosphoryl, or imino; R i is H, optionally substituted C1-C20 alkyl, optionally substituted C1-C20 heteroalkyl, optionally substituted C2-C20 alkenyl, optionally substituted C2-C20 heteroalkenyl, optionally substituted C2-C20 alkynyl, optionally substituted C2-C20 heteroalkynyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C5-C15 aryl, or optionally substituted C2-C15 heteroaryl; each of g1, h1, i1, j1, k1, l1, m1, n1, o1, g2, h2, i2, j2, k2, l2, m2, n2, o2, g3, h3, i3, j3, k3, l3, m3, n3, and o3 is, independently, 0 or 1; Q is a nitrogen atom, optionally substituted C1-C20 alkylene, optionally substituted C1-C20 heteroalkylene, optionally substituted C2-C20 alkenylene, optionally substituted C2-C20 heteroalkenylene, optionally substituted C2-C20 alkynylene, optionally substituted C2-C20 heteroalkynylene, optionally substituted C3-C20 cycloalkylene, optionally substituted C3-C20 heterocycloalkylene, optionally substituted C4-C20 cycloalkenylene, optionally substituted C4-C20 heterocycloalkenylene, optionally substituted C8-C20 cycloalkynylene, optionally substituted C8-C20 heterocycloalkynylene, optionally substituted C5-C15 arylene, or optionally substituted C2-C15 heteroarylene.

In some embodiments, depending on the structure and chemical formula of L C , L C may have two points of attachment to the Fc domain (e.g., two G C2 ).

,

C20 heteroalkylene, optionally substituted C2-C20 alkenylene, optionally su

heteroalkenylene, optionally substituted C2-C20 alkynylene, optionally substituted C2-C20

heteroalkynylene, optionally substituted C3-C20 cycloalkylene, optionally substituted C3-C20

heterocycloalkylene, optionally substituted C4-C20 cycloalkenylene, optionally substituted C4-C20 heterocycloalkenylene, optionally substituted C8-C20 cycloalkynylene, optionally substituted C8-C20 heterocycloalkynylene, optionally substituted C5-C15 arylene, optionally substituted C2-C15

heteroarylene, O, S, NR i , P, carbonyl, thiocarbonyl, sulfonyl, phosphate, and imino, and wherein R i is H, optionally substituted C1-C20 alkyl, optionally substituted C1-C20 heteroalkyl, optionally substituted C2- C20 alkenyl, optionally substituted C2-C20 heteroalkenyl, optionally substituted C2-C20 alkynyl, optionally substituted C2-C20 heteroalkynyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C5-C15 aryl, or optionally substituted C2-C15 heteroaryl.

In some embodiments of this aspect, E has the sequence of any one of SEQ ID NOs: 1-14, 1-29, or 1-31. In some embodiments of this aspect, when n is 2, E dimerizes to form an Fc domain.

In another as ect the invention features a con u ate of formula D-XV :

(D-XV)

wherein: each A is an independently selected amino acid; each E is an Fc domain monomer; each L is a linker that, when each m is 2, 3, 4, or 5, is bound to any of A and covalently attached to E; each m is 0, 1, 2, 3, 4, or 5; n is 1 or 2; T is an integer from 1 to 20; and Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 are each

independently selected from the side chain of an amino acid; or a pharmaceutically acceptable salt thereof. In another as ect the invention features a con u ate of formula D-

,

(D-XVI)

wherein: each A 1 and A 2 is an independently selected amino acid; each L is a linker that, when each m is 1, 2, 3, 4, or 5, is bound to a nitrogen atom in any A 1 and a nitrogen atom in any A 2 and covalently linked to E; each E is an Fc domain monomer; each m is independently 0, 1, 2, 3, 4, or 5; n is 1 or 2; T is an integer from 1 to 20; and Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 are each independently selected from the side chain of an amino acid; or a pharmaceutically acceptable salt thereof.

In some embodiments of this aspect, each A 1 and A 2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H-tetrazol-5-yl)alanine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4-diaminobutyric acid, 3-hydroxyproline, threonine, 2-amino-4-phenylbutyric acid, 3-(2-naphthyl)alanine, 2-piperazinecarboxylic acid, 2-aminooctanoic acid, serine, 2-aminohexanoic acid, 4-amino-4-piperidinyl carboxylic acid, methionine, methionine sulfoxide, methionine sulfone, S-methylcysteine, S-ethylcysteine, S-propylhomocysteine, cyclopropylalanine, 3-fluoroalanine, 2-amino-5-methylhexanoic acid, 2-amino-5- methylhex-4-enoic acid, alpha-t-butylglycine, and alpha-neopentylglycine; and each m is independently selected from 1, 2, 3, 4, or 5; or a pharmaceutically acceptable salt thereof.

In some embodiments of this aspect, each m is independently 2, 3, or 4, or a pharmaceutically acceptable salt thereof.

In some embodiments of this aspect, each A 1 and A 2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H-tetrazol-5-yl)alanine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4-diaminobutyric acid, 3-hydroxyproline, threonine, 2-amino-4-phenylbutyric acid, 3-(2-naphthyl)alanine, 2-piperazinecarboxylic acid, 2-aminooctanoic acid, serine, 2-aminohexanoic acid, 4-amino-4-piperidinyl carboxylic acid, methionine, methionine sulfoxide, methionine sulfone, S-methylcysteine, S-ethylcysteine, S-propylhomocysteine, cyclopropylalanine, 3-fluoroalanine, 2-amino-5-methylhexanoic acid, 2-amino-5- methylhex-4-enoic acid, alpha-t-butylglycine, and alpha-neopentylglycine; each m is independently 1, 2, 3 or 4; the combination of Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 is selected from one of or a pharmaceutically acceptable salt thereof.

In another aspect, the invention features a conjugate of formula (D-XVII):

,

(D-XVII)

wherein: each A 1 and A 2 is an independently selected amino acid; each X is absent or

is -CH2CH2C(O)NH-; each E is an Fc domain monomer; each m is independently 0, 1, 2, 3, 4, or 5; n is 1 or 2; Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 are each independently selected from the side chain of an amino acid; each d is an integer from 0 to 10; each e is an integer from 0 to 10; and T is an integer from 1 to 20, or a p harmaceutically acceptable salt thereof.

In some embodiments of this aspect, each A 1 and A 2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H-tetrazol-5-yl)alanine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4-diaminobutyric acid, 3-hydroxyproline, threonine, 2-amino-4-phenylbutyric acid, 3-(2-naphthyl)alanine, 2-piperazinecarboxylic acid, 2-aminooctanoic acid, serine, 2-aminohexanoic acid, 4-amino-4-piperidinyl carboxylic acid, methionine, methionine sulfoxide, methionine sulfone, S-methylcysteine, S-ethylcysteine, S-propylhomocysteine, cyclopropylalanine, 3-fluoroalanine, 2-amino-5-methylhexanoic acid, 2-amino-5- methylhex-4-enoic acid, alpha-t-butylglycine, and alpha-neopentylglycine; each m is 2 or 3; each d is an integer from 1 to 10; each e is an integer from 1 to 10; or a pharmaceutically acceptable salt thereof.

In some embodiments of this aspect, each A 1 and A 2 is independently selected from 2,4- diaminobutyric acid and threonine; each m is 2 or 3; each d is an integer from 1 to 10; and each e is an integer from 1 to 10; or a pharmaceutically acceptable salt thereof.

In some embodiments of this aspect, each m is 2, or a pharmaceutically acceptable salt thereof. In some embodiments, each m is 3, or a pharmaceutically acceptable salt thereof.

In some embodiments of this aspect, each d is 10; each e is 10; and each X is absent; or a pharmaceutically acceptable salt thereof.

In some embodiments of this aspect, each m is 2; each d is 1; each e is 1; and each X is - CH2CH2C(O)NH-; or a pharmaceutically acceptable salt thereof. In another as ect the invention features a con u ate of formula D-

,

(D-XVIII)

wherein: each A 1 and A 2 is an independently selected amino acid; each E is an Fc domain monomer; each m is independently 0, 1, 2, 3, 4, or 5; n is 1 or 2; Q1, Q2, Q3, Q4, Q5 and Q6 are each independently selected from the side chain of an amino acid; each d is an integer from 0 to 10; each e is an integer from 0 to 10; each f is an integer from 0 to 10; each g is an integer from 0 to 10; and T is an integer from 1 to 20, or a pharmaceutically acceptable salt thereof.

In some embodiments of this aspect, each A 1 and A 2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H-tetrazol-5-yl)alanine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4-diaminobutyric acid, 3-hydroxyproline, threonine, 2-amino-4-phenylbutyric acid, 3-(2-naphthyl)alanine, 2-piperazinecarboxylic acid, 2-aminooctanoic acid, serine, 2-aminohexanoic acid, 4-amino-4-piperidinyl carboxylic acid, methionine, methionine sulfoxide, methionine sulfone, S-methylcysteine, S-ethylcysteine, S-propylhomocysteine, cyclopropylalanine, 3-fluoroalanine, 2-amino-5-methylhexanoic acid, 2-amino-5- methylhex-4-enoic acid, alpha-t-butylglycine, and alpha-neopentylglycine; each m is 2; each d is 1; each e is 1; each f is 3; and each g is 1; or a pharmaceutically acceptable salt thereof.

In some embodiments of this aspect, each A 1 and A 2 is independently selected from

2,4-diaminobutyric acid and threonine; each m is 2; each d is 1; each e is 1; each f is 3; and each g is 1; or a pharmaceutically acceptable salt thereof.

In another as ect the invention features a conu ate of formula D-XIX :

,

(D XIX) wherein: each A 1 and A 2 is an independently selected amino acid; each E is

each m is independently 0, 1, 2, 3, 4, or 5; n is 1 or 2; Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 are each independently selected from the side chain of an amino acid; each d is an integer from 0 to 10; each e is an integer from 0 to 10; each f is an integer from 0 to 10; each g is an integer from 0 to 25; and T is an integer from 1 to 20, or a pharmaceutically acceptable salt thereof.

In some embodiments, each A 1 and A 2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H-tetrazol-5-yl)alanine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4- diaminobutyric acid, 3-hydroxyproline, threonine, 2-amino-4-phenylbutyric acid, 3-(2-naphthyl)alanine, 2- piperazinecarboxylic acid, 2-aminooctanoic acid, serine, 2-aminohexanoic acid, 4-amino-4-piperidinyl carboxylic acid, methionine, methionine sulfoxide, methionine sulfone, S-methylcysteine, S-ethylcysteine, S-propylhomocysteine, cyclopropylalanine, 3-fluoroalanine, 2-amino-5-methylhexanoic acid, 2-amino-5- methylhex-4-enoic acid, alpha-t-butylglycine, and alpha-neopentylglycine; each m is 2; each d is 1; each e is 1; each f is 1; and each g is 8 to 25; or a pharmaceutically acceptable salt thereof.

In some embodiments of this aspect, each A 1 and A 2 is independently selected from 2,4- diaminobutyric acid and threonine; each m is 2; each d is 1; each e is 1; each f is 1; and each g is 8 to 25; or a pharmaceutically acceptable salt thereof.

In another aspect, the invention features a conjugate of formula (D-XX):

,

(D-XX)

wherein: each A 1 and A 2 is an independently selected amino acid; each E is an Fc domain monomer; each m is independently 0, 1, 2, 3, 4, or 5; n is independently 1 or 2; Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 are each independently selected from the side chain of an amino acid; each d is an integer from 0 to 15; and T is an integer from 1 to 20, or a pharmaceutically acceptable salt thereof.

In some embodiments of this aspect, each A 1 and A 2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H-tetrazol-5-yl)alanine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4-diaminobutyric acid, 3-hydroxyproline, threonine, 2-amino-4-phenylbutyric acid, 3-(2-naphthyl)alanine, 2-piperazinecarboxylic acid, 2-aminooctanoic acid, serine, 2-aminohexanoic acid, 4-amino-4-piperidinyl carboxylic acid methionine methionine sulfoxide methionine sulfone S-methylcysteine S-ethylcysteine S-propylhomocysteine, cyclopropylalanine, 3-fluoroalanine, 2-amino-5-meth

methylhex-4-enoic acid, alpha-t-butylglycine, and alpha-neopentylglycine; each m is 2; and each d is 10; or a pharmaceutically acceptable salt thereof.

In some embodiments of this aspect, each A 1 and A 2 is independently selected from 2,4- diaminobutyric acid and piperazine-2-carboxylic acid; each m is 2; and each d is 10; or a

pharmaceutically acceptable salt thereof.

In another aspect, the invention features a conjugate of formula (D-XXI):

(D-XXI)

wherein: each A 1 and A 2 is an independently selected amino acid; each E is an Fc domain monomer ;

each Y is , -C(O)CH2CH2-, -CH2-, or is absent; each X is -C(O)CH2CH2- or is absent; each m is independently 0, 1, 2, 3, 4, or 5; n is 1 or 2; Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 are each independently selected from the side chain of an amino acid; each d is an integer from 0 to 15; each e is an integer from 1 to 10; each f is an integer from 1 to 5; each g is an integer from 1 to 5; and T is an integer from 1 to 20, or a pharmaceutically acceptable salt thereof.

In some embodiments of this aspect, each A 1 and A 2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H-tetrazol-5-yl)alanine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4-diaminobutyric acid, 3-hydroxyproline, threonine, 2-amino-4-phenylbutyric acid, 3-(2-naphthyl)alanine, 2-piperazinecarboxylic acid, 2-aminooctanoic acid, serine, 2-aminohexanoic acid, 4-amino-4-piperidinyl carboxylic acid, methionine, methionine sulfoxide, methionine sulfone, S-methylcysteine, S-ethylcysteine, S-propylhomocysteine, cyclopropylalanine, 3-fluoroalanine, 2-amino-5-methylhexanoic acid, 2-amino-5- methylhex-4-enoic acid, alpha-t-butylglycine, and alpha-neopentylglycine; each m is 2, 3 or 4; each d is 1; when each Y is , each e is 4; each f is 1 or 2; and each g is 1 or 2; or a pharmaceutically acceptable salt thereof.

In some embodiments of this aspect, each A 1 and A 2 is independently selected from 2,4- diaminobutyric acid, 3-aminoalanine, 2-piperazinecarboxylic acid, 2-aminohexanoic acid, 2-aminooctanoic

acid, methionine, and threonine; each m is 2, 3 or 4; when each Y is , each e is 4; each d is 1; each f is 1 or 2; each g is 1 or 2; or a pharmaceutically acceptable salt thereof.

In some embodiments of this aspect, each A 1 and A 2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H-tetrazol-5-yl)alanine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4-diaminobutyric acid, 3-hydroxyproline, threonine, 2-amino-4-phenylbutyric acid, 3-(2-naphthyl)alanine, 2-piperazinecarboxylic acid, 2-aminooctanoic acid, serine, 2-aminohexanoic acid, 4-amino-4-piperidinyl carboxylic acid, methionine, methionine sulfoxide, methionine sulfone, S-methylcysteine, S-ethylcysteine, S-propylhomocysteine, cyclopropylalanine, 3-fluoroalanine, 2-amino-5-methylhexanoic acid, 2-amino-5- methylhex-4-enoic acid, alpha-t-butylglycine, and alpha-neopentylglycine; each m is 3 or 4; each d is 1; each Y is absent; each f is 1; and each g is 1; or a pharmaceutically acceptable salt thereof.

In some embodiments of this aspect, each A 1 and A 2 is independently selected from 2,4- diaminobutyric acid and threonine; and each m is 3; or a pharmaceutically acceptable salt thereof.

In some embodiments of this aspect, each A 1 and A 2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H-tetrazol-5-yl)alanine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4-diaminobutyric acid, 3-hydroxyproline, threonine, 2-amino-4-phenylbutyric acid, 3-(2-naphthyl)alanine, 2-piperazinecarboxylic acid, 2-aminooctanoic acid, serine, 2-aminohexanoic acid, 4-amino-4-piperidinyl carboxylic acid, methionine, methionine sulfoxide, methionine sulfone, S-methylcysteine, S-ethylcysteine, S-propylhomocysteine, cyclopropylalanine, 3-fluoroalanine, 2-amino-5-methylhexanoic acid, 2-amino-5- methylhex-4-enoic acid, alpha-t-butylglycine, and alpha-neopentylglycine; each m is 2, 3 or 4; each d is 1; each Y is -C(O)CH2CH2-; each f is 1 or 2; and each g is 1 or 2; or a pharmaceutically acceptable salt thereof.

In some embodiments of this aspect, each A 1 and A 2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H-tetrazol-5-yl)alanine, 2,4-diaminobutyric acid, 3-aminoalanine, 2- aminohexanoic acid, 2-piperazinecarboxylic acid, 2-aminooctanoic acid, methionine, methionine sulfoxide, methionine sulfone, S-methylcysteine, S-ethylcysteine, S-propylhomocysteine,

c yclopropylalanine, 3-fluoroalanine, 2-amino-5-methylhexanoic acid, 2-amino-5-methylhex-4-enoic acid, alpha-t-butylglycine, and alpha-neopentylglycine, or a pharmaceutically acceptable salt thereof.

In some embodiments, each m is 4, each d is 1, each f is 1, and each g is 1, or a

pharmaceutically acceptable salt thereof. In some embodiments, each m is 3, each f is 2, and each g is 1, or a pharmaceutically acceptable salt thereof. In some embodiments, each A 1 and A 2 is independently selected fr

asparagine, glutamine, 3-(2H-tetrazol-5-yl)alanine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4- diaminobutyric acid, 3-hydroxyproline, threonine, 2-amino-4-phenylbutyric acid, 3-(2-naphthyl)alanine, 2- piperazinecarboxylic acid, 2-aminooctanoic acid, serine, 2-aminohexanoic acid, 4-amino-4-piperidinyl carboxylic acid, methionine, methionine sulfoxide, methionine sulfone, S-methylcysteine, S-ethylcysteine, S-propylhomocysteine, cyclopropylalanine, 3-fluoroalanine, 2-amino-5-methylhexanoic acid, 2-amino-5- methylhex-4-enoic acid, alpha-t-butylglycine, and alpha-neopentylglycine; each m is 2, 3 or 4; each d is 1; each Y is -CH2-; each f is 1 or 2; and each g is 1 or 2; or a pharmaceutically acceptable salt thereof.

In some embodiments of this aspect, each A 1 and A 2 is independently selected from 2,4- diaminobutyric acid, 2-aminooctanoic acid and threonine; each f is 1; and each g is 1; or a

pharmaceutically acceptable salt thereof.

In some embodiments, each A 1 and A 2 is independently selected from 2,4-diaminobutyric acid, 2- aminohexanoic acid, 2-aminooctanoic acid and threonine, or a pharmaceutically acceptable salt thereof; each m is 4; each f is 1; and each g is 1; or a pharmaceutically acceptable salt thereof.

In some embodiments of this aspect, each A 1 and A 2 is independently selected from 2,4- diaminobutyric acid, 2-aminohexanoic acid, 2-aminooctanoic acid and threonine, or a pharmaceutically acceptable salt thereof; each m is 3; each f is 1; and each g is 1; or a pharmaceutically acceptable salt thereof.

In another aspect, the invention features a conjugate of formula (D-XXII):

,

(D-XXII)

wherein: each A 1 and A 2 is an independently selected amino acid; each X is -C(O)CH2CH2CH2-Y- or - C(O)CH2CH2C(O)NH-; each Y is heteroaryl; each E is an Fc domain monomer; each m is independently 0, 1, 2, 3, 4, or 5; n is 1 or 2; T is an integer from 1 to 20; Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 are each independently selected from the side chain of an amino acid; and each d is

pharmaceutically acceptable salt thereof.

In some embodiments of this aspect, each A 1 and A 2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H-tetrazol-5-yl)alanine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4-diaminobutyric acid, 3-hydroxyproline, threonine, 2-amino-4-phenylbutyric acid, 3-(2-naphthyl)alanine, 2-piperazinecarboxylic acid, 2-aminooctanoic acid, serine, 2-aminohexanoic acid, 4-amino-4-piperidinyl carboxylic acid, methionine, methionine sulfoxide, methionine sulfone, S-methylcysteine, S-ethylcysteine, S-propylhomocysteine, cyclopropylalanine, 3-fluoroalanine, 2-amino-5-methylhexanoic acid, 2-amino-5- methylhex-4-enoic acid, alpha-t-butylglycine, and alpha-neopentylglycine; each m is 2 or 3; each d is an integer from 1 to 3; or a pharmaceutically acceptable salt thereof.

In some embodiments, each A 1 and A 2 is independently selected from 2,4-diaminobutyric acid, 3- (2-naphthyl)alanine, and threonine, or a pharmaceutically acceptable salt thereof.

In some embodiments, each X is -C(O)CH 2 CH 2 CH 2- Y-; each m is 3; each d is 3; and each Y is 1,4-triazololyl; or a pharmaceutically acceptable salt thereof.

In some embodiments, each X is -C(O)CH2CH2C(O)-; each m is 2; and each d is 1; or a pharmaceutically acceptable salt thereof.

In another as ect the invention features a conu ate of formula D-XXIII :

,

(D-XXIII)

wherein: each A 1 and A 2 is an independently selected amino acid; each E is an Fc domain monomer; each R is C1-C20 alkyl; each m is independently 0, 1, 2, 3, 4, or 5; n is 1 or 2; Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 are each independently selected from the side chain of an amino acid; each d is an integer from 0 to 20; each e is an integer from 0 to 20; each f is an integer from 0 to 20; and T is an integer from 1 to 20, or a pharmaceutically acceptable salt thereof.

In some embodiments of this aspect, each A 1 and A 2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H-tetrazol-5-yl)alanine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4-diaminobutyric acid, 3-hydroxyproline, threonine, 2-amino-4-phenylbutyric acid, 3-(2-naphthyl)alanine, 2-piperazinecarboxylic acid, 2-aminooctanoic acid, serine, 2-aminohexanoic acid, 4-amino-4-piperidinyl carboxylic acid, methionine, methionine sulfoxide, methionine sulfone, S-methylcysteine, S-ethylcysteine, methylhex-4-enoic acid, alpha-t-butylglycine, and alpha-neopentylglycine; e

integer from 1 to 3; each e is an integer from 1 to 3; each f is an integer from 1 to 3; and each R is C1- C10 alkyl; or a pharmaceutically acceptable salt thereof.

In some embodiments, each A 1 and A 2 is independently selected from 2,4-diaminobutyric acid and threonine; each m is 2; each d is 1; each e is 1; each f is 1; each R is C1-C10 alkyl; and or a pharmaceutically acceptable salt thereof.

In anoth r n f f rm l D-XXIV :

,

(D-XXIV)

wherein: each A 1 and A 2 is an independently selected amino acid; each E is an Fc domain monomer; each R is C1-C20 alkyl; each m is independently 0, 1, 2, 3, 4, or 5; n is 1 or 2; Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 are each independently selected from the side chain of an amino acid; each d is an integer from 1 to 20; each e is an integer from 1 to 20; each f is an integer from 1 to 20; and T is an integer from 1 to 20, or a pharmaceutically acceptable salt thereof.

I n some embodiments of this aspect, each A1 and A2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H-tetrazol-5-yl)alanine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4-diaminobutyric acid, 3-hydroxyproline, threonine, 2-amino-4-phenylbutyric acid, 3-(2-naphthyl)alanine, 2-piperazinecarboxylic acid, 2-aminooctanoic acid, serine, 2-aminohexanoic acid, 4-amino-4-piperidinyl carboxylic acid, methionine, methionine sulfoxide, methionine sulfone, S-methylcysteine, S-ethylcysteine, S-propylhomocysteine, cyclopropylalanine, 3-fluoroalanine, 2-amino-5-methylhexanoic acid, 2-amino-5- methylhex-4-enoic acid, alpha-t-butylglycine, and alpha-neopentylglycine; each m is 2, 3, or 4; each d is an integer from 1 to 3; each e is an integer from 1 to 3; each f is an integer from 1 to 3; each R is C1-C10 alkyl; and or a pharmaceutically acceptable salt thereof.

In some embodiments, each A 1 and A 2 is independently selected from 2,4-diaminobutyric acid, 2- aminooctanoic acid, and threonine; each m is 2, 3, or 4; each d is 1; each e is 1; each f is 1; and each R is C1-C10 alkyl; or a pharmaceutically acceptable salt thereof. In some embodiments, each m is 2, or a pharmaceutically acceptab

embodiments, each m is 3, or a pharmaceutically acceptable salt thereof. In some embodiments, each m is 4, or a pharmaceutically acceptable salt thereof.

In another h inv n i n f r n f f rm l D-XXV :

,

(D-XXV)

wherein: each A 1 and A 2 is an independently selected amino acid; each E is an Fc domain monomer; , -C(O)CH2CH2-, -CH2-, or is absent; each e is an integer from 1 to 10; each m is independently 0, 1, 2, 3, 4, or 5; n is 1 or 2; Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 are each independently selected from the side chain of an amino acid; each d is an integer from 0 to 15; and T is an integer from 1 to 20, or a pharmaceutically acceptable salt thereof.

In some embodiments of this aspect, each Y is–C(O)CH2CH2-, or a pharmaceutically acceptable salt thereof.

In some embodiments of this aspect, each A 1 and A 2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H-tetrazol-5-yl)alanine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4-diaminobutyric acid, 3-hydroxyproline, threonine, 2-amino-4-phenylbutyric acid, 3-(2-naphthyl)alanine, 2-piperazinecarboxylic acid, 2-aminooctanoic acid, serine, 2-aminohexanoic acid, 4-amino-4-piperidinyl carboxylic acid, methionine, methionine sulfoxide, methionine sulfone, S-methylcysteine, S-ethylcysteine, S-propylhomocysteine, cyclopropylalanine, 3-fluoroalanine, 2-amino-5-methylhexanoic acid, 2-amino-5- methylhex-4-enoic acid, alpha-t-butylglycine, and alpha-neopentylglycine; each m is 2, 3, or 4; and each d is an integer from 1 to 3; or a pharmaceutically acceptable salt thereof.

In some embodiments, each A 1 and A 2 is independently selected from 2,4-diaminobutyric acid, 2- aminooctanoic acid, and threonine; and each d is 1; or a pharmaceutically acceptable salt thereof.

In some embodiments, each m is 2, or a pharmaceutically acceptable salt thereof. In some embodiments, each m is 3, or a pharmaceutically acceptable salt thereof. In some embodiments, each m is 4, or a pharmaceutically acceptable salt thereof.

In another aspect, the invention features a conjugate of formula (D-XXVI):

,

(D-XXVI)

wherein: each A 1 and A 2 is an independently selected amino acid; each E is an Fc domain monomer; each X is -CH2- or -C(O)-; each m is independently 0, 1, 2, 3, 4, or 5; n is 1 or 2; Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 are each independently selected from the side chain of an amino acid; each d is an integer from 0 to 15; and T is an integer from 1 to 20, or a pharmaceutically acceptable salt thereof.

In some embodiments of this aspect, each A 1 and A 2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H-tetrazol-5-yl)alanine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4-diaminobutyric acid, 3-hydroxyproline, threonine, 2-amino-4-phenylbutyric acid, 3-(2-naphthyl)alanine, 2-piperazinecarboxylic acid, 2-aminooctanoic acid, serine, 2-aminohexanoic acid, 4-amino-4-piperidinyl carboxylic acid, methionine, methionine sulfoxide, methionine sulfone, S-methylcysteine, S-ethylcysteine, S-propylhomocysteine, cyclopropylalanine, 3-fluoroalanine, 2-amino-5-methylhexanoic acid, 2-amino-5- methylhex-4-enoic acid, alpha-t-butylglycine, and alpha-neopentylglycine; each m is 2, 3, or 4; and each d is an integer from 1 to 3; or a pharmaceutically acceptable salt thereof.

In some embodiments, each A 1 and A 2 is independently selected from 2,4-diaminobutyric acid, 2- aminooctanoic acid, and threonine; and each d is 1; or a pharmaceutically acceptable salt thereof.

In some embodiments, each m is 2, or a pharmaceutically acceptable salt thereof. In some embodiments, each m is 3, or a pharmaceutically acceptable salt thereof. In some embodiments, each m is 4, or a pharmaceutically acceptable salt thereof.

In another aspect, the invention features a conjugate of formula (D-

,

(D-XXVII)

wherein: each A 1 and A 2 is an independently selected amino acid; each E is an Fc domain monomer;

each Y is , -C(O)CH2CH2-, -CH2-, or is absent; each m is independently 0, 1, 2, 3, 4, or 5; n is 1 or 2; Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 are each independently selected from the side chain of an amino acid; each d is an integer from 0 to 15; each e is an integer from 1 to 3; and T is an integer from 1 to 20, or a pharmaceutically acceptable salt thereof.

In some embodiments of this aspect, each Y is -C(O)CH2CH2-, or a pharmaceutically acceptable salt thereof.

In some embodiments of this aspect, each A 1 and A 2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H-tetrazol-5-yl)alanine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4-diaminobutyric acid, 3-hydroxyproline, threonine, 2-amino-4-phenylbutyric acid, 3-(2-naphthyl)alanine, 2-piperazinecarboxylic acid, 2-aminooctanoic acid, serine, 2-aminohexanoic acid, 4-amino-4-piperidinyl carboxylic acid, methionine, methionine sulfoxide, methionine sulfone, S-methylcysteine, S-ethylcysteine, S-propylhomocysteine, cyclopropylalanine, 3-fluoroalanine, 2-amino-5-methylhexanoic acid, 2-amino-5- methylhex-4-enoic acid, alpha-t-butylglycine, and alpha-neopentylglycine; each m is 2, 3, or 4; each d is an integer from 1 to 3; or a pharmaceutically acceptable salt thereof.

In some embodiments, each A 1 and A 2 is independently selected from 2,4-diaminobutyric acid, 2- aminooctanoic acid, and threonine; and each d is 1; and each e is 1 or 2; or a pharmaceutically acceptable salt thereof.

In some embodiments, each m is 2, or a pharmaceutically acceptable salt thereof. In some embodiments, each m is 3, or a pharmaceutically acceptable salt thereof. In some embodiments, each m is 4, or a pharmaceutically acceptable salt thereof. In another aspect, the invention features a conjugate of formula (D-

,

(D-XXVIII)

wherein: each A 1 and A 2 is an independently selected amino acid; each E is an Fc domain monomer;

each X is independently selected from , -C(O)CH 2 CH 2 C(O)-, -CH 2 CH 2 NHC(O)CH 2 CH 2 -, -C(O)-, and -CH2-, or is absent; each m is independently 0, 1, 2, 3, 4, or 5; n is 1 or 2; Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 are each independently selected from the side chain of an amino acid; each d is an integer from 1 to 15; each g is an integer from 1 to 15; each e is independently from 1 to 15; and T is an integer from 1 to 20, or a pharmaceutically acceptable salt thereof.

In some embodiments of this aspect, the conjugate is of the formula (D-XXVIII-1):

(D-XXVIII-1)

or a pharmaceutically acceptable salt thereof. In some embodiments the con u ate is of the formula D-XXVIII-2

,

(D-XXVIII-2)

or a pharmaceutically acceptable salt thereof.

In each A 1 and A 2 is independently selected from glycine, arginine, asparagine, glutamine, 3-(2H- tetrazol-5-yl)alanine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4-diaminobutyric acid, 3- hydroxyproline, threonine, 2-amino-4-phenylbutyric acid, 3-(2-naphthyl)alanine, 2-piperazinecarboxylic acid, 2-aminooctanoic acid, serine, 2-aminohexanoic acid, 4-amino-4-piperidinyl carboxylic acid, methionine, methionine sulfoxide, methionine sulfone, S-methylcysteine, S-ethylcysteine, S- propylhomocysteine, cyclopropylalanine, 3-fluoroalanine, 2-amino-5-methylhexanoic acid, 2-amino-5- methylhex-4-enoic acid, alpha-t-butylglycine, and alpha-neopentylglycine; each m is 2, 3, or 4; each d is an integer from 1 to 5; each e is 1; each X is -C(O)CH2CH2C(O)-; or a pharmaceutically acceptable salt thereof.

In some embodiments of this aspect, each A 1 and A 2 is independently selected from 2,4- diaminobutyric acid, 2-aminohexanoic acid, and threonine; and each d is 1; or a pharmaceutically acceptable salt thereof.

In some embodiments, each m is 2, or a pharmaceutically acceptable salt thereof. In some embodiments, each m is 3, or a pharmaceutically acceptable salt thereof. In some embodiments, each m is 4, or a pharmaceutically acceptable salt thereof.

In some embodiments of the conjugates described herein, Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 are each independently selected from the side chain of a natural amino acid, or a pharmaceutically acceptable salt thereof. In some embodiments, at least one of Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 is selected from the side chain of a non-natural amino acid, or a pharmaceutically acceptable salt thereof. In some embodiments, at least two of Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 are independently selected from the side chain of a non-natural amino acid, or a pharmaceutically acceptable salt thereof. In some embodiments, at least three of Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 are independently selected from the side chain of a non-natural amino acid, or a pharmaceutically acceptable salt thereof. In some embodiments, at least four of Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 are independently selected from the side chain of a non-natural amino acid, or a pharmaceutically acceptable salt thereof. In some embodiments, at least five of Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 are

i ndependently selected from the side chain of a non-natural amino acid, or a pharmaceutically acceptable salt thereof. In some embodiments, each of Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 is in

side chain of a non-natural amino acid, or a pharmaceutically acceptable salt thereof.

In some embodiments, each Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 is independently selected from the side chain of serine, threonine, cysteine, glycine, proline, alanine, valine, isoleucine, leucine, methionine, phenylalanine, tyrosine, and tryptophan, or a pharmaceutically acceptable salt thereof.

In some embodiments, each of Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 is independently selected from C1-C4 alkyl, C1-C2 hydroxyalkyl, C1-C5 alkamino, and C6-C35 alkaryl, or a pharmaceutically acceptable salt thereof.

In some embodiments, each of Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 is independently selected from 2- methyl-1-propyl, 2-propyl, 1-hydroxyethyl, butyl, benzyl, hydroxymethyl, propyl, 2-butyl, methyl, and 2- aminoethyl, or a pharmaceutically acceptable salt thereof.

In some embodiments, the combination of Q 1 , Q 2 , Q 3 , Q 4 , Q 5 and Q 6 is selected from one of

or a pharmaceutically acceptable salt thereof.

In another aspect, the invention features a conjugate described by formula (M-Ia):

wherein each M includes a cyclic heptapeptide including a linking nitrogen atom; each E is an Fc domain monomer; L’ in each L’-M is a linker, wherein L’ is covalently attached to the linking nitrogen atom in M and to E; n is 1 or 2; and T is an integer from1 to 20; or a pharmaceutically acceptable salt thereof.

In some embodiments, when n is 2, E dimerizes to form an Fc domain, and the conjugate is d escribed by formula (M-Ib):

wherein J is an Fc domain; and T is an integer from 2 to 20, or a pharmaceutically acceptable salt thereof.

In some embodiments, L’ in each L’-M is described by formula (M-L’):

wherein L is a remainder of L’; and A is 1 to 5 amino acids, wherein at least one A is covalently attached to the linking nitrogen atom in M, or a pharmaceutically acceptable salt thereof.

In some embodiments of this aspect, the conjugate is described by

(M-II)

wherein each L is a remainder of L’ and is covalently attached to or may take the place of R 2 , R 3 , R 4 , R 5 , R6, R7, R8, R9, R10, or R16; each of R1 and R12 is, independently, a lipophilic moiety, a polar moiety, or H; each of R 11 , R 13 , and R 14 is, independently, optionally substituted C1-C5 alkamino, a polar moiety, a positively charged moiety, or H; each R 15 is a lipophilic moiety or a polar moiety; each of R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , R 8 , R 9 , and R 10 is, independently, a lipophilic moiety, a positively charged moiety, a polar moiety, H, optionally substituted C1-C5 alkamino, optionally substituted C1-C20 alkyl, optionally substituted C3- C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1-C20 heteroalkyl, optionally substituted C3- C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl; or two R groups on the same or adjacent atoms join to form an optionally substituted 5-8 membered ring; each R 16 is H, optionally substituted C1-C5 alkamino, optionally substituted C1-C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4- C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1-C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl, or is absent; and each of a, b, and c is, independently, 0 or 1; or a pharmaceutically acceptable salt thereof.

In some embodiments of this aspect, the conjugate includes at least one optionally substituted 5-8 membered ring formed by joining (i) R 2 , R 3 , and C 1 ; (ii) R 3 , R 4 , N 1 , and C 1 ; (iii) R 5 , R 6 , and C 2 ; (iv) R 6 , R 7 , N2, and C2; (v) R8, R9, and C3; or (vi) R9, R10, N3, and C3, or a pharmaceutically acceptable salt thereof. In some embodiments the con u ate is described b formula M-III

( )

or a pharmaceutically acceptable salt thereof.

In some embodiments, each of R 1 and R 12 is a lipophilic moiety; each of R 11 , R 13 , and R 14 is, independently, optionally substituted C1-C5 alkamino, a polar moiety, or a positively charged moiety; and each R 15 is a polar moiety; or a pharmaceutically acceptable salt thereof.

In some embodiments, each lipophilic moiety is independently selected from optionally substituted C1-C20 alkyl, optionally substituted C5-C15 aryl, optionally substituted C6-C35 alkaryl, or optionally C5-C10 substituted heteroaryl, or a pharmaceutically acceptable salt thereof.

In some embodiments, each lipophilic moiety is, independently, C1-C8 alkyl, methyl substituted C2-C4 alkyl, (C1-C10)alkylene(C6)aryl, phenyl substituted (C1-C10)alkylene(C6)aryl, or alkyl substituted C4-C9 heteroaryl, or a pharmaceutically acceptable salt thereof.

In some embodiments, each lipophilic moiety is, independently, benzyl, isobutyl, sec-butyl, isopropyl, n-propyl, methyl, biphenylmethyl, n-octyl, or methyl substituted indolyl, or a pharmaceutically acceptable salt thereof.

In some embodiments, each of R 11 , R 13 , and R 14 is independently optionally substituted C1-C5 alkamino, or a pharmaceutically acceptable salt thereof.

In some embodiments, each of R 11 , R 13 , and R 14 is CH2CH2NH2, or a pharmaceutically acceptable salt thereof.

In some embodiments, each R 15 is a polar moiety, or a pharmaceutically acceptable salt thereof. In some embodiments, the polar moiety comprises C1-C4 hydroxyalkyl, a carboxylic acid group, an ester group, or an amide group, or a pharmaceutically acceptable salt thereof. In some embodiments, the polar moiety is C1-C4 hydroxyalkyl, or a pharmaceutically acceptable salt thereof. In some embodiments, the polar moiety is CHCH3OH, or a pharmaceutically acceptable salt thereof. In some embodiments of this aspect, the conjugate is described by ( )

wherein each R 1 is benzyl or CH2CH(CH3)2, or a pharmaceutically acceptable salt thereof. 5 In some embodiments, the conjugate is described by formula (M-IV-1):

or a pharmaceutically acceptable salt thereof. In some embodiments the con u ate is described b formula M-IV

,

(M-V)

wherein each R 1 is benzyl or CH 2 CH(CH 3 ) 2 ; and each R 2 is a positively charged moiety, a polar moiety, optionally substituted C1-C5 alkamino, optionally substituted C1-C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1-C20 heteroalkyl, optionally substituted C3- C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl; or a pharmaceutically acceptable salt thereof. In some embodiments the con u ate is described b formula M-V-

(M-VI)

wherein each R 1 is benzyl or CH2CH(CH3)2; each R 2 is optionally substituted C1-C20 alkylene, optionally substituted C1-C20 heteroalkylene, optionally substituted C2-C20 alkenylene, optionally substituted C2- C20 heteroalkenylene, optionally substituted C2-C20 alkynylene, optionally substituted C2-C20 heteroalkynylene, optionally substituted C3-C20 cycloalkylene, optionally substituted C3-C20 heterocycloalkylene, optionally substituted C4-C20 cycloalkenylene, optionally substituted C4-C20 heterocycloalkenylene, optionally substituted C8-C20 cycloalkynylene, optionally substituted C8-C20 heterocycloalkynylene, optionally substituted C5-C15 arylene, or optionally substituted C2-C15 h eteroarylene; and each R16 is H, optionally substituted C1-C5 alkamino, optionally substituted C1-C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1-C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20

heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C

pharmaceutically acceptable salt thereof.

In some embodiments, the con u ate is described b formula M-VI-1):

,

(M-VI-1)

wherein each R 16 is H, optionally substituted C1-C5 alkamino, optionally substituted C1-C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1-C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20

heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl; or a pharmaceutically acceptable salt thereof.

In some embodiment f hi n i ri f rmula (M-VII):

(M-VII)

wherein each R 1 is benzyl or CH 2 CH(CH 3 ) 2 ; each R 2 is optionally substituted C1-C20 alkylene, optionally substituted C1-C20 heteroalkylene, optionally substituted C2-C20 alkenylene, optionally substituted C2- C20 heteroalkenylene, optionally substituted C2-C20 alkynylene, optionally substituted C2-C20 heteroalkynylene, optionally substituted C3-C20 cycloalkylene, optionally substituted C3-C20 heterocycloalkylene optionally substituted C4-C20 cycloalkenylene optionally substituted C4-C20 heterocycloalkenylene, optionally substituted C8-C20 cycloalkynylene, optio

heterocycloalkynylene, optionally substituted C5-C15 arylene, or optionally substituted C2-C15 heteroarylene; each R 6 is a positively charged moiety, a polar moiety, optionally substituted C1-C5 alkamino, optionally substituted C1-C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5- C15 aryl, optionally substituted C1-C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl; and each R 16 is H, optionally substituted C1-C5 alkamino, optionally substituted C1-C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1- C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl; or a pharmaceutically acceptable salt thereof.

In some embodiments the con u ate is described b formula M-VII-1):

(M-VII-1)

or a pharmaceutically acceptable salt thereof.

In some embodiments the conu ate is described b formula M-VI

,

(M-VII-2)

wherein each R 16 is H, optionally substituted C1-C20 alkyl, or optionally substituted C1-C20 heteroalkyl, or a pharmaceutically acceptable salt thereof.

In some embodimen h n i ri f rm l M-VII- ):

,

(M-VII-3)

or a pharmaceutically acceptable salt thereof.

In some embodiments of this aspect, the conjugate is described by

,

(M-VIII)

wherein each R 1 is benzyl or CH2CH(CH3)2; and each R 2 is optionally substituted C1-C20 alkylene, optionally substituted C1-C20 heteroalkylene, optionally substituted C2-C20 alkenylene, optionally substituted C2-C20 heteroalkenylene, optionally substituted C2-C20 alkynylene, optionally substituted C2-C20 heteroalkynylene, optionally substituted C3-C20 cycloalkylene, optionally substituted C3-C20 heterocycloalkylene, optionally substituted C4-C20 cycloalkenylene, optionally substituted C4-C20 heterocycloalkenylene, optionally substituted C8-C20 cycloalkynylene, optionally substituted C8-C20 heterocycloalkynylene, optionally substituted C5-C15 arylene, or optionally substituted C2-C15 heteroarylene; each R 6 , R 7 , N 2 , and C 2 together form an optionally substituted 5-8 membered ring comprising optionally substituted C3-C7 heterocycloalkyl comprising a nitrogen atom and additional 0-2 heteroatoms independently selected from N, O, and S, or optionally substituted C2-C7 heteroaryl comprising a nitrogen atom and additional 0-2 heteroatoms independently selected from N, O, and S; or a pharmaceutically acceptable salt thereof.

In some embodiment h n i ri f rm l M-VIII-1):

,

(M-VIII-1)

or a pharmaceutically acceptable salt thereof. In some embodiments, the conjugate is described by formula (M-VI

(M-VIII-2)

or a pharmaceutically acceptable salt thereof.

In some embodiments, the conjugate is described by formula (M-VIII-3):

,

(M-VIII-3)

or a pharmaceutically acceptable salt thereof.

In some embodiments, the conjugate is described by formula (M-VIII-4):

(M-VIII-4)

or a pharmaceutically acceptable salt thereof. In some embodiments the con u ate is described b formula M-VI

,

(M-VIII-5)

or a pharmaceutically acceptable salt thereof.

I n some embodiments, the conjugate is described by formula (M-IX):

,

(M-IX)

wherein each R 1 is benzyl or CH2CH(CH3)2; each R 2 is optionally substituted C1-C20 alkylene, optionally substituted C1-C20 heteroalkylene, optionally substituted C2-C20 alkenylene, optionally substituted C2- C20 heteroalkenylene, optionally substituted C2-C20 alkynylene, optionally substituted C2-C20 heteroalkynylene, optionally substituted C3-C20 cycloalkylene, optionally substituted C3-C20

heterocycloalkylene, optionally substituted C4-C20 cycloalkenylene, optionally substituted C4-C20 heterocycloalkenylene, optionally substituted C8-C20 cycloalkynylene, optionally substituted C8-C20 heterocycloalkynylene, optionally substituted C5-C15 arylene, or optionally substituted C2-C15 heteroarylene; each of R 6 and R 8 is, independently, a positively charged moiety, a polar moiety, optionally substituted C1-C5 alkamino, optionally substituted C1-C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1-C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl; and each R 16 is H, optionally substituted C1-C5 alkamino, ti ll b tit t d C1 C20 lk l ti ll b tit t d C3 C20 l lk l ti ll b tit t d C4 C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally sub

optionally substituted C1-C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl; or a pharmaceutically acceptable salt thereof.

In some embo im n h n i ri f rm l M-IX-1 :

,

(M-IX-1)

or a pharmaceutically acceptable salt thereof.

I n some embodim - -

(M-IX-2)

wherein each R 16 is H, optionally substituted C1-C20 alkyl, or optionally substituted C1-C20 heteroalkyl; or a pharmaceutically acceptable salt thereof.

I n some embodiments, the conjugate is described by formula (M-IX-3):

(M-IX-3)

wherein each R 16 is H, optionally substituted C1-C20 alkyl, or optionally substituted C1-C20 heteroalkyl; or a pharmaceutically acceptable salt thereof.

In some embodiments of this as ect the con u ate is described b formula (M-X):

(M-X)

wherein each R 1 is benzyl or CH2CH(CH3)2; each R 2 is optionally substituted C1-C20 alkylene, optionally substituted C1-C20 heteroalkylene, optionally substituted C2-C20 alkenylene, optionally substituted C2- C20 heteroalkenylene, optionally substituted C2-C20 alkynylene, optionally substituted C2-C20 heteroalkynylene, optionally substituted C3-C20 cycloalkylene, optionally substituted C3-C20 heterocycloalkylene, optionally substituted C4-C20 cycloalkenylene, optionally substituted C4-C20 heterocycloalkenylene, optionally substituted C8-C20 cycloalkynylene, optionally substituted C8-C20 heterocycloalkynylene, optionally substituted C5-C15 arylene, or optionally substituted C2-C15 heteroarylene; each R 6 , R 7 , N 2 , and C 2 together form an optionally substituted 5-8 membered ring comprising optionally substituted C3-C7 heterocycloalkyl comprising a nitrogen atom and additional 0-2 heteroatoms independently selected from N, O, and S, or optionally substituted C2-C7 heteroaryl comprising a nitrogen atom and additional 0-2 heteroatoms independently selected from N, O, and S; each R 8 is a positively charged moiety, a polar moiety, optionally substituted C1-C5 alkamino, optionally substituted C1-C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1-C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4- C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C

is H, optionally substituted C1-C5 alkamino, optionally substituted C1-C20 alkyl, optionally substituted C3- C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1-C20 heteroalkyl, optionally substituted C3- C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl, or a pharmaceutically acceptable salt thereof.

In some embodiments the con u ate is described b formula M-X-1 :

( M-X-1)

or a pharmaceutically acceptable salt thereof.

In some embodiments the con u ate is described b formula M-XI):

,

(M-XI)

wherein each R 1 is benzyl or CH2CH(CH3)2; and each of R 2 and R 6 is, independently, a positively charged moiety, a polar moiety, optionally substituted C1-C5 alkamino, optionally substituted C1-C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1-C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20

heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl, or a h ti ll t bl lt th f In some embodiments the conu ate is described b formula M-XI

,

(M-XI-1)

or a pharmaceutically acceptable salt thereof.

In some embodim n h n i ri f rm l M-XI-2:

,

(M-XI-2)

or a pharmaceutically acceptable salt thereof.

In some embodim n h n i ri f rm l M-XII:

,

(M-XII) wherein each R 1 is benzyl or CH 2 CH(CH 3 ) 2 ; each R 2 is a positively charged

optionally substituted C1-C5 alkamino, optionally substituted C1-C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1-C20 heteroalkyl, optionally substituted C3- C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl; and each R 6 , R 7 , N 2 , and C 2 together form an optionally substituted 5-8 membered ring comprising optionally substituted C3-C7 heterocycloalkyl comprising a nitrogen atom and additional 0-2 heteroatoms independently selected from N, O, and S, or optionally substituted C2-C7 heteroaryl comprising a nitrogen atom and additional 0-2 heteroatoms independently selected from N, O, and S; or a pharmaceutically acceptable salt thereof.

In some embodim n f hi h n i ri f rmula (M-XIII):

(M-XIII)

w herein each R1 is benzyl or CH2CH(CH3)2; each R2 is a positively charged moiety, a polar moiety, optionally substituted C1-C5 alkamino, optionally substituted C1-C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1-C20 heteroalkyl, optionally substituted C3- C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl; each R 6 is optionally substituted C1-C20 alkylene, optionally substituted C1-C20 heteroalkylene, optionally substituted C2-C20 alkenylene, optionally substituted C2- C20 heteroalkenylene, optionally substituted C2-C20 alkynylene, optionally substituted C2-C20 heteroalkynylene, optionally substituted C3-C20 cycloalkylene, optionally substituted C3-C20 heterocycloalkylene, optionally substituted C4-C20 cycloalkenylene, optionally substituted C4-C20 heterocycloalkenylene, optionally substituted C8-C20 cycloalkynylene, optionally substituted C8-C20 heterocycloalkynylene, optionally substituted C5-C15 arylene, or optionally substituted C2-C15 heteroarylene; and each R 16 is H, optionally substituted C1-C5 alkamino, optionally substituted C1-C20 alkyl optionally substituted C3 C20 cycloalkyl optionally substituted C4 C20 cycloalkenyl optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optiona

heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20

heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl; or a pharmaceutically acceptable salt thereof.

In some embodiments, the conjugate is described by formula (M-XIII-1):

,

(M-XIII-1)

wherein each R 16 is H or optionally substituted C6-C35 heteroalkaryl; or a pharmaceutically acceptable salt thereof.

In some embodiments of this aspect, the conjugate is described by formula (M-XIV):

,

(M-XIV)

wherein each R 1 is benzyl or CH2CH(CH3)2; each R 2 is a positively charged moiety, a polar moiety, optionally substituted C1-C5 alkamino, optionally substituted C1-C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1-C20 heteroalkyl, optionally substituted C3- C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl optionally substituted C2 C15 heteroaryl optionally substituted C6 C35 alkaryl or optionally substituted C6-C35 heteroalkaryl; each R 6 , R 7 , N 2 , and C 2 togeth

substituted 5-8 membered ring comprising optionally substituted C3-C7 heterocycloalkyl comprising a nitrogen atom and additional 0-2 heteroatoms independently selected from N, O, and S, or optionally substituted C2-C7 heteroaryl comprising a nitrogen atom and additional 0-2 heteroatoms independently selected from N, O, and S; and each R16 is H, optionally substituted C1-C5 alkamino, optionally substituted C1-C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1-C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4- C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl; or a pharmaceutically acceptable salt thereof.

In some embodimen h n i ri f rm l M-XIV-1):

,

(M-XIV-1)

w herein each R16 is H or optionally substituted C6-C35 heteroalkaryl; or a pharmaceutically acceptable salt thereof.

In some embodiments of this aspect, the conjugate is described by formula (M-XV):

,

(M-XV)

wherein each R 1 is benzyl or CH2CH(CH3)2; and each of R 2 , R 6 , and R 8 is, independently, a positively charged moiety, a polar moiety, optionally substituted C1-C5 alkamino, optionally substituted C1-C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C2

substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1-C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl; or a pharmaceutically acceptable salt thereof.

In some embodimen h n i ri f rm l M-XV-1):

(M-XV-1)

or a pharmaceutically acceptable salt thereof.

In some embodiments the con u ate is described b formula M-XV-2 :

(M-XV-2)

or a pharmaceutically acceptable salt thereof.

In some embodiments of this as ect the con u ate is described b

(M-XVI)

wherein each R 1 is benzyl or CH2CH(CH3)2; each of R 2 and R 8 is, independently, a positively charged moiety, a polar moiety, optionally substituted C1-C5 alkamino, optionally substituted C1-C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1-C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20

heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl; and each R 6 , R 7 , N 2 , and C 2 together form an optionally substituted 5-8 membered ring comprising optionally substituted C3-C7 heterocycloalkyl comprising a nitrogen atom and additional 0-2 heteroatoms independently selected from N, O, and S, or optionally substituted C2-C7 heteroaryl comprising a nitrogen atom and additional 0-2 heteroatoms independently selected from N, O, and S; or a

p harmaceutically acceptable salt thereof.

In some embodiments of this as ect the con u ate is described b formula (M-XVII):

(M-XVII)

wherein each R 1 is benzyl or CH2CH(CH3)2; each of R 2 and R 6 is, independently, a positively charged moiety, a polar moiety, optionally substituted C1-C5 alkamino, optionally substituted C1-C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1-C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, option

heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl; each R 8 is optionally substituted C1-C20 alkylene, optionally substituted C1-C20 heteroalkylene, optionally substituted C3-C20 cycloalkylene, optionally substituted C3-C20 heterocycloalkylene, optionally substituted C4-C20 heterocycloalkenylene, optionally substituted C8-C20 heterocycloalkynylene, optionally substituted C5-C15 arylene, or optionally substituted C2-C15 heteroarylene; and each R 16 is H, optionally substituted C1-C5 alkamino, optionally substituted C1-C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1-C20 heteroalkyl, optionally substituted C3- C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl, or a pharmaceutically acceptable salt thereof.

In some embodiments, h n i ri f rm l M-XVII-1):

,

( M-XVII-1)

or a pharmaceutically acceptable salt thereof.

In some embodiments the con u ate is described b formula M-XVII-2):

(M-XVII-2)

or a pharmaceutically acceptable salt thereof. In some embodiments the con u ate is described b formula M-XV

(M-XVII-3)

or a pharmaceutically acceptable salt thereof.

In some embodim n h n i ri f rm l M-XVIII :

,

(M-XVIII)

wherein each R 1 is benzyl or CH2CH(CH3)2; each R 2 is a positively charged moiety, a polar moiety, optionally substituted C1-C5 alkamino, optionally substituted C1-C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1-C20 heteroalkyl, optionally substituted C3- C20 heterocycloalkyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl; each R 6 , R 7 , N 2 , and C 2 together form an optionally substituted 5-8 membered ring comprising optionally substituted C3-C7 heterocycloalkyl comprising a nitrogen atom and additional 0-2 heteroatoms independently selected from N, O, and S, or optionally substituted C2-C7 heteroaryl comprising a nitrogen atom and additional 0-2 heteroatoms independently selected from N, O, and S; each R 8 is optionally substituted C1-C20 alkylene, optionally substituted C1- C20 heteroalkylene, optionally substituted C2-C20 alkenylene, optionally substituted C2-C20 heteroalkenylene, optionally substituted C2-C20 alkynylene, optionally substituted C2-C20

heteroalkynylene, optionally substituted C3-C20 cycloalkylene, optionally substituted C3-C20 h t l lk l ti ll b tit t d C4 C20 l lk l ti ll b tit t d C4 C20 heterocycloalkenylene, optionally substituted C8-C20 cycloalkynylene, optio

heterocycloalkynylene, optionally substituted C5-C15 arylene, or optionally substituted C2-C15 heteroarylene; and each R 16 is H, optionally substituted C1-C5 alkamino, optionally substituted C1-C20 alkyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C5-C15 aryl, optionally substituted C1-C20 heteroalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20

heterocycloalkenyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C2-C15 heteroaryl, optionally substituted C6-C35 alkaryl, or optionally substituted C6-C35 heteroalkaryl, or a pharmaceutically acceptable salt thereof.

In some embodiments, each R 2 is optionally substituted C1-C5 alkamino (e.g., CH2NH2 or CH2CH2NH2) or a polar moiety, or a pharmaceutically acceptable salt thereof. In some embodiments, each R 6 is a polar moiety, or a pharmaceutically acceptable salt thereof. In some embodiments, the polar moiety comprises C1-C4 hydroxylalkyl group, a carboxylic acid group, an ester group, or an amide group, or a pharmaceutically acceptable salt thereof. In some embodiments, the polar moiety is C1-C4 hydroxyalkyl, or a pharmaceutically acceptable salt thereof. In some embodiments, the polar moiety is CHCH3OH or CH2OH, or a pharmaceutically acceptable salt thereof.

In some embodiments, each R 8 is optionally substituted C1-C5 alkamino or optionally substituted C5-C15 aryl, or a pharmaceutically acceptable salt thereof. In some embodiments, each R 8 is CH2NH2 or CH2CH2NH2, or a pharmaceutically acceptable salt thereof. In some embodiments, each R 8 is naphthyl, or a pharmaceutically acceptable salt thereof.

In some embodiments, each R 6 , R 7 , N 2 , and C 2 together form a 5- or 6-membered ring comprising C4-C5 heterocycloalkyl comprising a nitrogen atom and additional 0 or 1 heteroatom independently selected from N, O, and S, or a pharmaceutically acceptable salt thereof.

In some embodiments of this aspect, when n is 2, E dimerizes to form an Fc domain.

I n some embodiments, each L’ or L comprises one or more optionally substituted C1-C20 alkylene, optionally substituted C1-C20 heteroalkylene, optionally substituted C2-C20 alkenylene, optionally substituted C2-C20 heteroalkenylene, optionally substituted C2-C20 alkynylene, optionally substituted C2- C20 heteroalkynylene, optionally substituted C3-C20 cycloalkylene, optionally substituted C3-C20 heterocycloalkylene, optionally substituted C4-C20 cycloalkenylene, optionally substituted C4-C20 heterocycloalkenylene, optionally substituted C8-C20 cycloalkynylene, optionally substituted C8-C20 heterocycloalkynylene, optionally substituted C5-C15 arylene, optionally substituted C2-C15 heteroarylene, O, S, NR i , P, carbonyl, thiocarbonyl, sulfonyl, phosphate, phosphoryl, or imino; and R i is H, optionally substituted C1-C20 alkyl, optionally substituted C1-C20 heteroalkyl, optionally substituted C2-C20 alkenyl, optionally substituted C2-C20 heteroalkenyl, optionally substituted C2-C20 alkynyl, optionally substituted C2-C20 heteroalkynyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C5-C15 aryl, or optionally substituted C2-C15 heteroaryl; or a f In some embodiments, the backbone of each L’ or L consists of one

C1-C20 alkylene, optionally substituted C1-C20 heteroalkylene, optionally substituted C2-C20 alkenylene, optionally substituted C2-C20 heteroalkenylene, optionally substituted C2-C20 alkynylene, optionally substituted C2-C20 heteroalkynylene, optionally substituted C3-C20 cycloalkylene, optionally substituted C3-C20 heterocycloalkylene, optionally substituted C4-C20 cycloalkenylene, optionally substituted C4-C20 heterocycloalkenylene, optionally substituted C8-C20 cycloalkynylene, optionally substituted C8-C20 heterocycloalkynylene, optionally substituted C5-C15 arylene, optionally substituted C2-C15 heteroarylene, O, S, NR i , P, carbonyl, thiocarbonyl, sulfonyl, phosphate, phosphoryl, or imino; and R i is H, optionally substituted C1-C20 alkyl, optionally substituted C1-C20 heteroalkyl, optionally substituted C2-C20 alkenyl, optionally substituted C2-C20 heteroalkenyl, optionally substituted C2-C20 alkynyl, optionally substituted C2-C20 heteroalkynyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C5-C15 aryl, or optionally substituted C2-C15 heteroaryl; or a pharmaceutically acceptable salt thereof.

In some embodiments, each L’ or L is oxo substituted, or a pharmaceutically acceptable salt thereof. In some embodiments, the backbone of each L’ or L comprises no more than 120 atoms, or a pharmaceutically acceptable salt thereof. In some embodiments, each L’ or L is capable of forming an amide, a carbamate, a sulfonyl, or a urea linkage, or a pharmaceutically acceptable salt thereof. In some embodiments, each L is a bond, or a pharmaceutically acceptable salt thereof.

In some embodiments, each L is described by formula (M-L-1):

wherein: J 1 is a bond attached to A or M if A is absent; J 2 is a bond attached to E; each of Q 1 , Q 2 , Q 3 , Q 4 , and Q 5 is, independently, optionally substituted C1-C20 alkylene, optionally substituted C1-C20 heteroalkylene, optionally substituted C2-C20 alkenylene, optionally substituted C2-C20

heteroalkenylene, optionally substituted C2-C20 alkynylene, optionally substituted C2-C20

heteroalkynylene, optionally substituted C3-C20 cycloalkylene, optionally substituted C3-C20

heterocycloalkylene, optionally substituted C4-C20 cycloalkenylene, optionally substituted C4-C20 heterocycloalkenylene, optionally substituted C8-C20 cycloalkynylene, optionally substituted C8-C20 heterocycloalkynylene, optionally substituted C5-C15 arylene, or optionally substituted C2-C15 heteroarylene; each of T 1 , T 2 , T 3 , T 4 is, independently, O, S, NR i , P, carbonyl, thiocarbonyl, sulfonyl, phosphate, phosphoryl, or imino; R i is H, optionally substituted C1-C20 alkyl, optionally substituted C1- C20 heteroalkyl, optionally substituted C2-C20 alkenyl, optionally substituted C2-C20 heteroalkenyl, optionally substituted C2-C20 alkynyl, optionally substituted C2-C20 heteroalkynyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20

cycloalkynyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C5-C15 aryl, or optionally substituted C2-C15 heteroaryl; and each of g, h, i, j, k, l, m, n, and o is, independently, 0 or 1; or a pharmaceutically acceptable sal tthere of. In some embodiments, depending on the structure and chemical fo points of attachment to the Fc domain (e.g., two J 2 ).

In some embodiments, L is

wherein each of d and e is, independently, an integer from 1 to 26; or a ph

thereof. In another aspect, the invention features a conjugate of formula (M-XIX):

,

(M-XIX)

wherein:each A is an independently selected amino acid; each L is a linker that, when each m is 2, 3, 4, or 5, is bound to any of A; each E is an Fc domain monomer; each m is 0, 1, 2, 3, 4, or 5; n is 1 or 2; Tis an integer from 1 to 20; and Q 1 , Q 2 , and Q 3 are each independently selected from the side chain of an amino acid; or a pharmaceutically acceptable salt thereof.

In some embodiments of this aspect, Q 1 , Q 2 , and Q 3 are each independently selected from the side chain of a natural amino acid, or a pharmaceutically acceptable salt thereof.

In some embodiments, at least one of Q 1 , Q 2 , and Q 3 is selected from the side chain of a non- natural amino acid, or a pharmaceutically acceptable salt thereof. In some embodiments, at least two of Q 1 , Q 2 , and Q 3 are independently selected from the side chain of a non-natural amino acid, or a pharmaceutically acceptable salt thereof. In some embodiments, each of Q 1 , Q 2 , and Q 3 is independently selected from the side chain of a non-natural amino acid, or a pharmaceutically acceptable salt thereof.

In some embodiments, each of Q 1 , Q 2 , and Q 3 is independently selected from the side chain of serine, threonine, cysteine, glycine, proline, alanine, valine, isoleucine, leucine, methionine,

p henylalanine, tyrosine, and tryptophan, or a pharmaceutically acceptable salt thereof.

In some embodiments, each of Q 1 , Q 2 , and Q 3 is independently selected from C1-C4 alkyl, C1-C2 hydroxyalkyl, C1-C5 alkamino, and C6-C35 alkaryl, or a pharmaceutically acceptable salt thereof.

In some embodiments, each of Q 1 , Q 2 , and Q 3 is independently selected from 2-methyl-1-propyl, 2-propyl, 1-hydroxyethyl, butyl, benzyl, hydroxymethyl, propyl, 2-butyl, methyl, and 2-aminoethyl, or a pharmaceutically acceptable salt thereof. In some embodiments, the combination of Q 1 , Q 2 , and Q 3 is selecte

In some embodiments, each m is 1, 2 or 3, or a pharmaceutically acceptable salt thereof. In some embodiments, each m is 1, 2, 3, or 4; or a pharmaceutically acceptable salt thereof.

In some embodiments, each A is independently selected from glycine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4-diaminobutyric acid, 3-hydroxyproline, threonine and 2-amino-4- phenylbutyric acid; and each m is 1, 2, 3, 4, or 5; or a pharmaceutically acceptable salt thereof.

In some embodiments, each m is 1, 2, or 3, or a pharmaceutically acceptable salt thereof. In some embodiments, each A is independently selected from glycine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4-diaminobutyric acid, 3-hydroxyproline, threonine and 2-amino-4- phenylbutyric acid; and each m is 1, 2, 3, 4, or 5; or a pharmaceutically acceptable salt thereof.

In some embodiments, each m is 1, 2, or 3; or a pharmaceutically acceptable salt thereof. In some embodiments of this aspect, when n is 2, E dimerizes to form an Fc domain, or a pharmaceutically acceptable salt thereof.

I n another aspect, the invention features a conjugate of formula (M-XX):

,

(M-XX)

wherein: each A is an independently selected amino acid; each E is an Fc domain monomer; each m is 0, 1, 2, 3, 4, or 5; n is 1 or 2; Q 1 , Q 2 , and Q 3 are each independently selected from the side chain of an amino acid; each d is 0 to 20; and T is an integer from 1 to 20; or a pharmaceutically acceptable salt thereof.

In some embodiments of this aspect, each m is 1, 2 or 3, or a pharmaceutically acceptable salt thereof. In some embodiments, each m is 1, 2, 3, or 4; or a pharmaceutically acceptable salt thereof.

In some embodiments, each A is independently selected from glycine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4-diaminobutyric acid, 3-hydroxyproline, threonine, 3-(2-naphthyl)alanine, and 2-amino-4-phenylbutyric acid; and each m is 1, 2, 3, 4, or 5; or a pharmaceutically acceptable salt thereof.

In some embodiments, each m is 1, 2, or 3, or a pharmaceutically acceptable salt thereof.

In some embodiments, each A is independently selected from glycine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4-diaminobutyric acid, 3-hydroxyproline, threonine, 3-(2-naphthyl)alanine, and 2-amino-4-phenylbutyric acid; each m is 1, 2, 3, 4, or 5; or a pharmaceutically acceptable salt thereof.

In some embodiments, each m is 1, 2, or 3; or a pharmaceutically acceptable salt thereof.

In some embodiments, each d is 0 to 10, or a pharmaceutically acceptable salt thereof. In some embodiments, each d is 0 to 5, or a pharmaceutically acceptable salt thereof. In some embodiments, each d is 0 or 1, or a pharmaceutically acceptable salt thereof.

In some embodiments, each A is independently selected from glycine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4-diaminobutyric acid, 3-hydroxyproline, threonine, 3-(2-naphthyl)alanine, and 2-amino-4-phenylbutyric acid, and 2-aminopentanoic acid; each m is 1, 2, 3, or 4; Q 1 , Q 2 , and Q 3 are each independently selected from 2-methyl-1-propyl, 2-propyl, 1-hydroxyethyl, butyl, benzyl,

hydroxymethyl, propyl, 2-butyl, methyl, and 2,4-diaminobutyric acid; and each d is 0 to 10; or a pharmaceutically acceptable salt thereof.

In some embodiments, Q 1 , Q 2 , and Q 3 are each independently selected from 2-methyl-1-propyl, benzyl, and 1-hydroxyethyl, or a pharmaceutically acceptable salt thereof. In some embodiments, each A is independently selected from glyci

hydroxyproline, 3-(2-naphthyl)alanine, and threonine, and 2-aminopentanoic acid; each m is 2 to 4; each d is 0 or 1; or a pharmaceutically acceptable salt thereof.

In another aspect, the invention features a conjugate of formula (M-XXI):

,

(M-XXI)

wherein: each A is an independently selected amino acid; each E is an Fc domain monomer; each m is 0, 1, 2, 3, 4, or 5; n is 1 or 2; Q1, Q2, and Q3 are each independently selected from the side chain of an amino acid; each d is an integer from 1 to 26; each e is an integer from 1 to 26; and T is an integer from 1 to 20; or a pharmaceutically acceptable salt thereof.

In some embodiments of this aspect, each m is 1, 2 or 3, or a pharmaceutically acceptable salt thereof. In some embodiments, each m is 1, 2, 3, or 4; or a pharmaceutically acceptable salt thereof.

In some embodiments, each A is independently selected from glycine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4-diaminobutyric acid, 3-hydroxyproline, threonine and 2-amino-4- phenylbutyric acid; and each m is 1, 2, 3, 4, or 5; or a pharmaceutically acceptable salt thereof.

In some embodiments, each m is 1, 2, or 3, or a pharmaceutically acceptable salt thereof.

In some embodiments, each A is independently selected from glycine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4-diaminobutyric acid, 3-hydroxyproline, threonine and 2-amino-4- phenylbutyric acid; and each m is 1, 2, 3, 4, or 5; or a pharmaceutically acceptable salt thereof.

In some embodiments, each m is 1, 2, or 3; or a pharmaceutically acceptable salt thereof.

In some embodiments, each d is an integer from 1 to 10, and each e is an integer from 1 to 10, or a pharmaceutically acceptable salt thereof.

In some embodiments, each d is an integer from 1 to 5, and each e is an integer from 1 to 6, or a pharmaceutically acceptable salt thereof. In some embodiments, each d is 1, and each e is an integer from 1 to 6, or a pharmaceutically acceptable salt thereof. In some embodiments, each d is 1, and each e is 6, or a pharmaceutically acceptable salt thereof.

In some embodiments of this aspect, each A is independently selected from glycine, 3- aminoalanine, piperazine-2-carboxylic acid, 2,4-diaminobutyric acid, 3-hydroxyproline, threonine and 2- amino-4-phenylbutyric acid; each m is 1, 2 or 3; Q 1 , Q 2 , and Q 3 are each independently selected from 2- diaminobutyric acid; each d is an integer from 1 to 10; and each e is an inte

pharmaceutically acceptable salt thereof.

In some embodiments, Q 1 , Q 2 , and Q 3 are each independently selected from 2-methyl-1-propyl and 1-hydroxyethyl, or a pharmaceutically acceptable salt thereof.

I n some embodiments, each A is independently selected from 2,4-diaminobutyric acid and threonine; each m is 2; each d is 1; and each e is 6; or a pharmaceutically acceptable salt thereof.

In n h r h inv n i n f r n f f rm l M-XXII :

,

(M-XXII)

wherein: each A is an independently selected amino acid; each X is -CH2- or -C(O)-; each Z is -C(O)NH- , -NHC(O)-,-CH2NH-, or O; each E is an Fc domain monomer; each m is 0, 1, 2, 3, 4, or 5; n is 1 or 2; Q1, Q2, and Q3 are each independently selected from the side chain of an amino acid; each d is an integer from 1 to 26; each e is an integer from 1 to 26; and T is an integer from 1 to 20; or a pharmaceutically acceptable salt thereof.

I n some embodiments, each m is 1, 2 or 3, or a pharmaceutically acceptable salt thereof. In some embodiments, each m is 1, 2, 3, or 4; or a pharmaceutically acceptable salt thereof.

In some embodiments, each A is independently selected from glycine, 2-aminoalanine, 3- aminoalanine, piperazine-2-carboxylic acid, 2,4-diaminobutyric acid, 3-hydroxyproline, threonine and 2- amino-4-phenylbutyric acid; and each m is 1, 2, 3, 4, or 5; or a pharmaceutically acceptable salt thereof.

In some embodiments, each m is 1, 2, or 3, or a pharmaceutically acceptable salt thereof.

In some embodiments, each A is independently selected from glycine, 2-aminoalanine, 3- aminoalanine, piperazine-2-carboxylic acid, 2,4-diaminobutyric acid, 3-hydroxyproline, threonine and 2- amino-4-phenylbutyric acid; each m is 1, 2, 3, 4, or 5; or a pharmaceutically acceptable salt thereof.

In some embodiments, each m is 1, 2, or 3; or a pharmaceutically acceptable salt thereof.

In some embodiments, each d is 1, and each e is 6 to 24, or a pharmaceutically acceptable salt thereof.

In some embodiments, each A is independently selected from glycine, 2-aminoalanine, 3- aminoalanine, piperazine-2-carboxylic acid, 2,4-diaminobutyric acid, 3-hydroxyproline, threonine and 2- amino-4-phenylbutyric acid; each m is 1, 2 or 3; Q 1 , Q 2 , and Q 3 are each independently selected from 2- diaminobutyric acid; each d is an integer from 1 to 10; and each e is an inte

pharmaceutically acceptable salt thereof.

In some embodiments, Q 1 , Q 2 , and Q 3 are each independently selected from benzyl, 2-methyl-1- propyl and 1-hydroxyethyl, or a pharmaceutically acceptable salt thereof.

I n some embodiments, each A is independently selected from 2-aminoalanine, 2,4-diaminobutyric acid, 3-hydroxyproline, piperazine-2-carboxylic acid, and threonine; each Z is, -CH2NH-, -C(O)NH- or - NHC(O)-; each m is 2 or 3; each d is 1; and each e is 4 to 26; or a pharmaceutically acceptable salt thereof.

In some embodiments, each A is independently selected from glycine, 2-aminoalanine, 2,4- diaminobutyric acid, 3-hydroxyproline, and threonine; each Z is -CH2NH-, or O; each m is 2; each d is 1; and each e is 4 to 26; or a pharmaceutically acceptable salt thereof.

In n h r n f f rm l M-XXIV :

,

(M-XXIV)

w herein: each A is an independently selected amino acid; each E is an Fc domain monomer; each m is 0, 1, 2, 3, 4, or 5; n is 1 or 2; Q1, Q2, and Q3 are each independently selected from the side chain of an amino acid; and each d is an integer from 1 to 26; each e is an integer from 1 to 26; and T is an integer from 1 to 20; or a pharmaceutically acceptable salt thereof.

In some embodiments, each m is 1, 2 or 3, or a pharmaceutically acceptable salt thereof. In some embodiments, each m is 1, 2, 3, or 4; or a pharmaceutically acceptable salt thereof.

In some embodiments, each A is independently selected from glycine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4-diaminobutyric acid, 3-hydroxyproline, threonine and 2-amino-4- phenylbutyric acid; and each m is 1, 2, 3, 4, or 5; or a pharmaceutically acceptable salt thereof.

In some embodiments, each m is 1, 2, or 3, or a pharmaceutically acceptable salt thereof.

In some embodiments, each A is independently selected from glycine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4-diaminobutyric acid, 3-hydroxyproline, threonine and 2-amino-4- phenylbutyric acid; each m is 1, 2, or 3; or a pharmaceutically acceptable salt thereof.

In some embodiments, each d is an integer from 1 to 10, and each e is an integer from 1 to 10, or a pharmaceutically acceptable salt thereof. In some embodiments, each d is an integer from 1 to 5, and each e is an integer from 1 to 5, or a pharmaceutically acceptable salt there

each d is 1, and each e is 4, or a pharmaceutically acceptable salt thereof.

In some embodiments, each A is independently selected from glycine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4-diaminobutyric acid, 3-hydroxyproline, threonine and 2-amino-4- phenylbutyric acid; each m is 1, 2 or 3; Q1, Q2, and Q3 are each independently selected from 2-methyl-1- propyl, 2-propyl, 1-hydroxyethyl, butyl, benzyl, hydroxymethyl, propyl, 2-butyl, methyl, and 2,4- diaminobutyric acid; each d is an integer from 1 to 10; and each e is an integer from 1 to 10; or a pharmaceutically acceptable salt thereof.

In some embodiments, Q 1 , Q 2 , and Q 3 are each independently selected from benzyl, 2-methyl-1- propyl and 1-hydroxyethyl, or a pharmaceutically acceptable salt thereof.

In some embodiments, each A is independently selected from 2,4-diaminobutyric acid and threonine; each m is 2; each d is 1; and each e is 4; or a pharmaceutically acceptable salt thereof.

In n h r h inv n i n f r n f f rm l M-XXV :

,

(M-XXV)

wherein: each A is an independently selected amino acid; each E is an Fc domain monomer; each m is 0, 1, 2, 3, 4, or 5; n is 1 or 2; Q1, Q2, and Q3 are each independently selected from the side chain of an amino acid; each d is an integer from 1 to 26; each e is an integer from 1 to 26; T is an integer from 1 to 20; or a pharmaceutically acceptable salt thereof.

In some embodiments, each m is 1, 2 or 3, or a pharmaceutically acceptable salt thereof. In some embodiments, each m is 1, 2, 3, or 4; or a pharmaceutically acceptable salt thereof.

In some embodiments, each A is independently selected from glycine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4-diaminobutyric acid, 3-hydroxyproline, threonine, 2-aminooctanoic acid, and 2-amino-4-phenylbutyric acid; and each m is 1, 2, 3, 4, or 5; or a pharmaceutically acceptable salt thereof.

In some embodiments, each m is 1, 2, or 3, or a pharmaceutically acceptable salt thereof.

In some embodiments, each A is independently selected from glycine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4-diaminobutyric acid, 3-hydroxyproline, threonine, 2-aminooctanoic acid, d 2 i 4 h lb t i id h i 1 2 3 4 h ti ll t bl lt th f In some embodiments, each d is an integer from 1 to 10, and each

a pharmaceutically acceptable salt thereof. In some embodiments, each d is an integer from 1 to 5, and each e is an integer from 1 to 5, or a pharmaceutically acceptable salt thereof.

In some embodiments, each m is 4, each d is 1, and each e is 4, or a pharmaceutically acceptable salt thereof.

In some embodiments, each A is independently selected from glycine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4-diaminobutyric acid, 3-hydroxyproline, threonine, 2-aminooctanoic acid, and 2-amino-4-phenylbutyric acid; each m is 1, 2, 3, or 4; Q 1 , Q 2 , and Q 3 are each independently selected from 2-methyl-1-propyl, 2-propyl, 1-hydroxyethyl, butyl, benzyl, hydroxymethyl, propyl, 2-butyl, methyl, and 2,4-diaminobutyric acid; each d is 1; and each e is an integer from 1 to 5; or a pharmaceutically acceptable salt thereof.

In some embodiments, Q 1 , Q 2 , and Q 3 are each independently selected from benzyl, 2-methyl-1- propyl and 1-hydroxyethyl, or a pharmaceutically acceptable salt thereof.

In some embodiments, each A is independently selected from 2,4-diaminobutyric acid, 2- aminooctanoic acid, and threonine; each m is 4; each d is 1; and each e is 4; or a pharmaceutically acceptable salt thereof.

In another as ect the invention features a con u ate of formula M-XXVI :

,

(M-XXVI)

wherein: each A is an independently selected amino acid; each X is heteroaryl or heterocyclyl; each E is an Fc domain monomer; each m is 0, 1, 2, 3, 4, or 5; n is 1 or 2; Q1, Q2, and Q3 are each independently selected from the side chain of an amino acid; each d is an integer from 1 to 26; each e is an integer from 1 to 26; and T is an integer from 1 to 20; or a pharmaceutically acceptable salt thereof.

In some embodiments, each m is 1, 2 or 3, or a pharmaceutically acceptable salt thereof. In some embodiments, each m is 1, 2, 3, or 4; or a pharmaceutically acceptable salt thereof.

In some embodiments, each A is independently selected from glycine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4-diaminobutyric acid, 3-hydroxyproline, threonine, 2-aminooctanoic acid, 3-(2-naphthyl)alanine, and 2-amino-4-phenylbutyric acid; and each m is 1, 2, 3, 4, or 5; or a

pharmaceutically acceptable salt thereof. In some embodiments, each A is independently selected from glyci

piperazine-2-carboxylic acid, 2,4-diaminobutyric acid, 3-hydroxyproline, threonine, 2-aminooctanoic acid, 3-(2-naphthyl)alanine, and 2-amino-4-phenylbutyric acid; each m is 1, 2, 3, or 4; or a pharmaceutically acceptable salt thereof.

I n some embodiments, each d is an integer from 1 to 10, and each e is an integer from 1 to 10, or a pharmaceutically acceptable salt thereof. In some embodiments, each d is an integer from 1 to 5, and each e is an integer from 1 to 5, or a pharmaceutically acceptable salt thereof. In some embodiments, each m is 4, each d is 1, and each e is 4, or a pharmaceutically acceptable salt thereof.

In some embodiments, each A is independently selected from glycine, 3-aminoalanine, piperazine-2-carboxylic acid, 2,4-diaminobutyric acid, 3-hydroxyproline, threonine, 2-aminooctanoic acid, 3-(2-naphthyl)alanine, and 2-amino-4-phenylbutyric acid; each X is heteroaryl; each m is 1, 2, 3, or 4; Q 1 , Q 2 , and Q 3 are each independently selected from 2-methyl-1-propyl, 2-propyl, 1-hydroxyethyl, butyl, benzyl, hydroxymethyl, propyl, 2-butyl, methyl, and 2,4-diaminobutyric acid; each d is an integer from 1 to 5; and each e is an integer from 1 to 5; or a pharmaceutically acceptable salt thereof.

In some embodiments, each A is independently selected from 2,4-diaminobutyric acid, threonine, 2-aminooctanoic acid, and 2-amino-4-phenylbutyric acid; each m is 2, 3, or 4; and each e is an integer from 1 to 3; or a pharmaceutically acceptable salt thereof.

In some embodiments, each X is triazolyl, or a pharmaceutically acceptable salt thereof. In some embodiments, each X is 1,4-triazolyl, or a pharmaceutically acceptable salt thereof. In some

embodiments, Q 1 , Q 2 , and Q 3 are each independently selected from benzyl, 2-methyl-1-propyl and 1- hydroxyethyl, or a pharmaceutically acceptable salt thereof.

In some embodiments, each A is independently selected from 2,4-diaminobutyric acid, 2- aminooctanoic acid, and threonine; each m is 4; each d is 1; and each e is 3; or a pharmaceutically acceptable salt thereof.

I n some embodiments of the conjugates described herein, Q1, Q2, and Q3 are each independently selected from the side chain of a natural amino acid, or a pharmaceutically acceptable salt thereof. In some embodiments, at least one of Q 1 , Q 2 , and Q 3 is selected from the side chain of a non-natural amino acid, or a pharmaceutically acceptable salt thereof. In some embodiments, at least two of Q 1 , Q 2 , and Q 3 are independently selected from the side chain of a non-natural amino acid, or a pharmaceutically acceptable salt thereof. In some embodiments, each of Q 1 , Q 2 , and Q 3 is independently selected from the side chain of a non-natural amino acid, or a pharmaceutically acceptable salt thereof. In some embodiments, each of Q 1 , Q 2 , and Q 3 is independently selected from the side chain of serine, threonine, cysteine, glycine, proline, alanine, valine, isoleucine, leucine, methionine, phenylalanine, tyrosine, and tryptophan, or a pharmaceutically acceptable salt thereof. In some embodiments, each of Q 1 , Q 2 , and Q 3 is independently selected from C1-C4 alkyl, C1-C2 hydroxyalkyl, C1-C5 alkamino, and C6-C35 alkaryl, or a pharmaceutically acceptable salt thereof.

In some embodiments, each of Q 1 , Q 2 , and Q 3 is independently selected from 2-methyl-1-propyl, 2-propyl, 1-hydroxyethyl, butyl, benzyl, hydroxymethyl, propyl, 2-butyl, methyl, and 2-aminoethyl, or a f In some embodiments, the combination of Q 1 , Q 2 , and Q 3 is selecte

In some embodiments of the conjugates described herein, when n is 2, E dimerizes to form an Fc domain, or a pharmaceutically acceptable salt thereof.

In some embodiments of any of the aspects of a conjugate described herein, a concentration of the conjugate, or a pharmaceutically acceptable salt thereof, that activates an immune cell is less than or equal to 10,000 nM. In some embodiments, a concentration of the conjugate, or a pharmaceutically acceptable salt thereof, that activates an immune cell is less than or equal to equal to 1,000 nM. In some embodiments, a concentration of the conjugate, or a pharmaceutically acceptable salt thereof, that activates an immune cell is less than or equal to equal to 100 nM.

In another aspect, the invention features a pharmaceutical composition including a conjugate described herein, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient. In some embodiments, the pharmaceutical composition further includes an antibacterial agent.

In some embodiments, the antibacterial agent is selected from the group consisting of linezolid, tedizolid, posizolid, radezolid, retapamulin, valnemulin, tiamulin, azamulin, lefamulin, plazomicin, amikacin, gentamicin, gamithromycin, kanamycin, neomycin, netilmicin, tobramycin, paromomycin, streptomycin spectinomycin geldanamycin herbimycin rifaximin loracarbef ertapenem doripenem imipenem/cilastatin, meropenem, cefadroxil, cefazolin, cefalotin, cefalexin,

cefoxitin, cefprozil, cefuroxime, cefixime, cefdinir, cefditoren, cefoperazone, cefotaxime, cefpodoxime, ceftazidime, ceftibuten, ceftizoxime, ceftriaxone, cefepime, ceftaroline fosamil, ceftobiprole, teicoplanin, vancomycin, telavancin, dalbavancin, oritavancin, clindamycin, lincomycin, daptomycin, solithromycin, azithromycin, clarithromycin, dirithromycin, erythromycin, roxithromycin, troleandomycin, telithromycin, spiramycin, aztreonam, furazolidone, nitrofurantoin, amoxicillin, ampicillin, azlocillin, carbenicillin, cloxacillin, dicloxacillin, flucloxacillin, mezlocillin, methicillin, nafcillin, oxacillin, penicillin g, penicillin v, piperacillin, penicillin g, temocillin, ticarcillin, amoxicillin clavulanate, ampicillin/sulbactam,

piperacillin/tazobactam, ticarcillin/clavulanate, bacitracin, ciprofloxacin, enoxacin, gatifloxacin, gemifloxacin, levofloxacin, lomefloxacin, moxifloxacin, nalidixic acid, norfloxacin, ofloxacin, trovafloxacin, grepafloxacin, sparfloxacin, temafloxacin, mafenide, sulfacetamide, sulfadiazine, silver sulfadiazine, sulfadimethoxine, sulfamethizole, sulfamethoxazole, sulfanilimide, sulfasalazine, sulfisoxazole, trimethoprim-sulfamethoxazole (tmp-smx), sulfonamidochrysoidine, eravacycline, demeclocycline, doxycycline, minocycline, oxytetracycline, tetracycline, clofazimine, dapsone, capreomycin, cycloserine, ethambutol(bs), ethionamide, isoniazid, pyrazinamide, rifampicin, rifabutin, rifapentine, streptomycin, arsphenamine, chloramphenicol, fosfomycin, fusidic acid, metronidazole, mupirocin, platensimycin, quinupristin/dalfopristin, thiamphenicol, tigecycline, tinidazole, and trimethoprim, prodrugs thereof, and pharmaceutically acceptable salts thereof. In some embodiments, a prodrug of tedizolid is tedizolid phosphate.

In some embodiments, the antibacterial agent is tedizolid, azithromycin, meropenem, amikacin, levofloxacin, rifampicin, linezolid, erythromycin, or solithromycin. In some embodiments, the antibacterial agent is tedizolid, azithromycin, meropenem, amikacin, or levofloxacin.

In another aspect, the invention features a method of protecting against or treating a bacterial infection in a subject, the method including administering to the subject a conjugate described herein. In some embodiments, the method further includes administering to the subject an antibacterial agent.

In another aspect, the invention features a method of protecting against or treating a bacterial infection in a subject, the method including administering to the subject (1) a conjugate described herein and (2) an antibacterial agent.

In another aspect, the invention features a method of inducing immune cell activation of the immune response in a subject having a bacterial infection, the method including administering to the subject a conjugate described herein. In some embodiments, the method further includes administering to the subject an antibacterial agent.

In another aspect, the invention features a method of inducing immune cell activation of the immune response in a subject having a bacterial infection, the method including administering to the subject (1) a conjugate described herein and (2) an antibacterial agent.

In some embodiments of the methods, the conjugate and the antibacterial agent are administered substantially simultaneously. In some embodiments, the conjugate and the antibacterial agent are administered separately. In some embodiments, the conjugate is administered first, followed by

f administered first, followed by administering of the conjugate alone. In som

and the antibacterial agent are administered substantially simultaneously, followed by administering of the conjugate or the antibacterial agent alone. In some embodiments, the conjugate or the antibacterial agent is administered first, followed by administering of the conjugate and the antibacterial agent substantially simultaneously.

In some embodiments of the methods, administering the conjugate and the antibacterial agent together lowers the MIC of each of the conjugate and the antibacterial agent relative to the MIC of each of the conjugate and the antibacterial agent when each is used alone.

In some embodiments of the methods, the conjugate and/or the antibacterial agent is

administered intramuscularly, intravenously, intradermally, intraarterially, intraperitoneally, intralesionally, intracranially, intraarticularly, intraprostatically, intrapleurally, intratracheally, intranasally, intravitreally, intravaginally, intrarectally, topically, intratumorally, peritoneally, subcutaneously, subconjunctival, intravesicularlly, mucosally, intrapericardially, intraumbilically, intraocularally, orally, locally, by inhalation, by injection, or by infusion.

In another aspect, the invention features a method of preventing, stabilizing, or inhibiting the growth of bacteria, or killing bacteria, including contacting the bacteria or a site susceptible to bacterial growth with a conjugate described herein.

In some embodiments, the method further includes contacting the bacteria or the site susceptible to bacterial growth with an antibacterial agent.

In another aspect, the invention features a method of preventing, stabilizing, or inhibiting the growth of bacteria, or killing bacteria, including contacting the bacteria or a site susceptible to bacterial growth with (1) a conjugate described herein and (2) an antibacterial agent.

In some embodiments of the methods described herein, the antibacterial agent is selected from the group consisting of linezolid, tedizolid, posizolid, radezolid, retapamulin, valnemulin, tiamulin, azamulin, lefamulin, plazomicin, amikacin, gentamicin, gamithromycin, kanamycin, neomycin, netilmicin, tobramycin, paromomycin, streptomycin, spectinomycin, geldanamycin, herbimycin, rifaximin, loracarbef, ertapenem, doripenem, imipenem/cilastatin, meropenem, cefadroxil, cefazolin, cefalotin, cefalexin, cefaclor, cefamandole, cefoxitin, cefprozil, cefuroxime, cefixime, cefdinir, cefditoren, cefoperazone, cefotaxime, cefpodoxime, ceftazidime, ceftibuten, ceftizoxime, ceftriaxone, cefepime, ceftaroline fosamil, ceftobiprole, teicoplanin, vancomycin, telavancin, dalbavancin, oritavancin, clindamycin, lincomycin, daptomycin, solithromycin, azithromycin, clarithromycin, dirithromycin, erythromycin, roxithromycin, troleandomycin, telithromycin, spiramycin, aztreonam, furazolidone, nitrofurantoin, amoxicillin, ampicillin, azlocillin, carbenicillin, cloxacillin, dicloxacillin, flucloxacillin, mezlocillin, methicillin, nafcillin, oxacillin, penicillin g, penicillin v, piperacillin, penicillin g, temocillin, ticarcillin, amoxicillin clavulanate,

ampicillin/sulbactam, piperacillin/tazobactam, ticarcillin/clavulanate, bacitracin, ciprofloxacin, enoxacin, gatifloxacin, gemifloxacin, levofloxacin, lomefloxacin, moxifloxacin, nalidixic acid, norfloxacin, ofloxacin, trovafloxacin, grepafloxacin, sparfloxacin, temafloxacin, mafenide, sulfacetamide, sulfadiazine, silver sulfadiazine, sulfadimethoxine, sulfamethizole, sulfamethoxazole, sulfanilimide, sulfasalazine,

f f ( ) f demeclocycline, doxycycline, minocycline, oxytetracycline, tetracycline, clof

capreomycin, cycloserine, ethambutol(bs), ethionamide, isoniazid, pyrazinamide, rifampicin, rifabutin, rifapentine, streptomycin, arsphenamine, chloramphenicol, fosfomycin, fusidic acid, metronidazole, mupirocin, platensimycin, quinupristin/dalfopristin, thiamphenicol, tigecycline, tinidazole, and

t rimethoprim, prodrugs thereof, and pharmaceutically acceptable salts thereof. In some embodiments, a prodrug of tedizolid is tedizolid phosphate.

In some embodiments of the methods, the antibacterial agent is tedizolid, azithromycin, meropenem, amikacin, levofloxacin, rifampicin, linezolid, erythromycin, or solithromycin. In some embodiments of the methods, the antibacterial agent is tedizolid, azithromycin, meropenem, amikacin, or levofloxacin.

In some embodiments of the methods, the bacterial infection is caused by Gram-negative bacteria. In some embodiments, the bacterial infection is caused by a resistant strain of bacteria. In some embodiments, the resistant strain of bacteria possesses the mcr-1 gene, the mcr-2 gene, and/or a chromosomal mutation conferring polymyxin resistance. In some embodiments, the resistant strain of bacteria possesses the mcr-1 gene. In some embodiments, the resistant strain of bacteria possesses the mcr-2 gene. In some embodiments, the resistant strain of bacteria possesses a chromosomal mutation conferring polymyxin resistance. In some embodiments, the resistant strain of bacteria is a resistant strain of E. coli.

In another aspect, the invention features a method of preventing lipopolysaccharides (LPS) in Gram-negative bacteria from activating an immune system in a subject, including administering to the subject a conjugate described herein. In some embodiments, the method prevents LPS from activating a macrophage. In some embodiments, the method prevents LPS-induced nitric oxide (NO) production from a macrophage. Definitions

The term“cyclic heptapeptide” or“cycloheptapeptide,” as used herein, refers to certain compounds that bind to lipopolysaccharides (LPS) in the cell membrane of Gram-negative bacteria to disrupt and permeabilize the cell membrane, leading to cell death and/or sensitization to other antibiotics. Cyclic heptapeptides or cycloheptapeptides comprise seven natural or non-natural α-amino acid residues, such as D- or L-amino acid residues, in a closed ring. Generally, cyclic heptapeptides are formed by linking the α-carboxyl group of one amino acid to the α-amino group or the γ-amino group of another amino acid and cyclizing. The cyclic heptapeptide comprises a heterocycle comprising carbon and nitrogen ring members, which may be substituted, for example, with amino acid side chains. One nitrogen from an α-amino group in the cyclic heptapeptide, however, is not a ring member and is branched from a ring member of the heterocycle. Thus, this nitrogen is directly attached to a ring member, such as a carbon atom (e.g., an α-carbon atom). This nitrogen atom serves as an attachment point for the cyclic heptapeptide to a linker and/or to a peptide (e.g., a peptide including 1-5 amino acid residue(s)), and thus is referred to herein as a“linking nitrogen.” The linking nitrogen is directly attached to the ring of the cyclic heptapeptide and is not derived from a side chain, s

chain. The linking nitrogens in a conjugate of, e.g., formula (II) or (III), are N 4 and N’ 4 .

In some embodiments, a peptide including one or more (e.g., 1-5; 1, 2, 3, 4, or 5) amino acid residues (e.g., natural and/or non-natural amino acid residues) may be covalently attached to a linking nitrogen (e.g., N4 and/or N’4, the nitrogen from an α-amino group) in the cyclic heptapeptide ring. Cyclic heptapeptides may be derived from polymyxins (e.g., naturally existing polymyxins and non-natural polymyxins) and/or octapeptins (e.g., naturally existing octapeptins and non-natural octapeptins).

Examples of naturally existing polymyxins include, but are not limited to, polymyxin B1, polymyxin B2, polymyxin B 3 , polymyxin B 4 , polymyxin B 5 , polymyxin B 6 , polymyxin B 1 -Ile, polymyxin B 2 -Ile, polymyxin C 1 , polymyxin C2, polymyxin S1, polymyxin T1, polymyxin T2, polymyxin A1, polymyxin A2, polymyxin D1, polymyxin D2, polymyxin E1 (colistin A), polymyxin E2 (colistin B), polymyxin E3, polymyxin E4, polymyxin E7, polymyxin E1-Ile, polymyxin E1-Val, polymyxin E1-Nva, polymyxin E2-Ile, polymyxin E2-Val, polymyxin E 2 -Nva, polymyxin E 8 -Ile, polymyxin M 1 , and polymyxin M 2 . In other embodiments, a cyclic heptapeptide may be entirely synthetic and prepared by standard peptide methodology as known in the art.

As used herein, the term“Fc domain monomer” refers to a polypeptide chain that includes at least a hinge domain and second and third antibody constant domains (CH2 and CH3) or functional fragments thereof (e.g., fragments that that capable of (i) dimerizing with another Fc domain monomer to form an Fc domain, and (ii) binding to an Fc receptor. The Fc domain monomer can be any

immunoglobulin antibody isotype, including IgG, IgE, IgM, IgA, or IgD (e.g., IgG). Additionally, the Fc domain monomer can be an IgG subtype (e.g., IgG1, IgG2a, IgG2b, IgG3, or IgG4) (e.g., IgG1). An Fc domain monomer does not include any portion of an immunoglobulin that is capable of acting as an antigen-recognition region, e.g., a variable domain or a complementarity determining region (CDR). Fc domain monomers in the conjugates as described herein can contain one or more changes from a wild- type Fc domain monomer sequence (e.g., 1-10, 1-8, 1-6, 1-4 amino acid substitutions, additions, or deletions) that alter the interaction between an Fc domain and an Fc receptor. Examples of suitable changes are known in the art.

As used herein, the term“Fc domain” refers to a dimer of two Fc domain monomers that is capable of binding an Fc receptor. In the wild-type Fc domain, the two Fc domain monomers dimerize by the interaction between the two C H 3 antibody constant domains, in some embodiments, one or more disulfide bonds form between the hinge domains of the two dimerizing Fc domain monomers.

The term“covalently attached” refers to two parts of a conjugate that are linked to each other by a covalent bond formed between two atoms in the two parts of the conjugate. For example, in the conjugate described herein (e.g., a conjugate of any one of formulas (I)-(XXXIII)), when a’ is 0, L is covalently attached to N’ 4 , which means that when a’ is 0, an atom in L forms a covalent bond with N’ 4 in the conjugate. Similarly, when a’ is 1 and b’ is 0, L is covalently attached to N’ 1 ; when b’ is 1, and c’ is 0, L is covalently attached to N’ 2 ; when c’ is 1, L is covalently attached to N’ 3 ; when a is 0, L is covalently attached to N 4 ; when a is 1 and b is 0, L is covalently attached to N 1 ; when b is 1, and c is 0, L is covalently attached to N 2 ; and when c is 1, L is covalently attached to N 3 . The terms“linker,”“L’,”“L,” and“L 1 ,” as used herein, refer to a cova

between two or more components in a conjugate (e.g., between two cyclic heptapeptides in a conjugate described herein, between a cyclic heptapeptide and an Fc domain in a conjugate described herein, and betwee a dimer of two cyclic peptapeptide and an Fc domain in a conjugate described herein). In some embodiments, a conjugate described herein may contain a linker that has a trivalent structure (e.g., a trivalent linker). A trivalent linker has three arms, in which each arm is covalently linked to a component of the conjugate (e.g., a first arm conjugated to a first cyclic heptapeptide, a second arm conjugated to a second cyclic heptapeptide, and a third arm conjugated to an Fc domain).

Molecules that may be used as linkers include at least two functional groups, which may be the same or different, e.g., two carboxylic acid groups, two amine groups, two sulfonic acid groups, a carboxylic acid group and a maleimide group, a carboxylic acid group and an alkyne group, a carboxylic acid group and an amine group, a carboxylic acid group and a sulfonic acid group, an amine group and a maleimide group, an amine group and an alkyne group, or an amine group and a sulfonic acid group. The first functional group may form a covalent linkage with a first component in the conjugate and the second functional group may form a covalent linkage with the second component in the conjugate. In some embodiments of a trivalent linker, two arms of a linker may contain two dicarboxylic acids, in which the first carboxylic acid may form a covalent linkage with the first cyclic heptapeptide in the conjugate and the second carboxylic acid may form a covalent linkage with the second cyclic heptapeptide in the conjugate, and the third arm of the linker may for a covalent linkage with an Fc domain in the conjugate. Examples of dicarboxylic acids are described further herein. In some embodiments, a molecule containing one or more maleimide groups may be used as a linker, in which the maleimide group may form a carbon-sulfur linkage with a cysteine in a component (e.g., an Fc domain) in the conjugate. In some embodiments, a molecule containing one or more alkyne groups may be used as a linker, in which the alkyne group may form a 1,2,3-triazole linkage with an azide in a component (e.g., an Fc domain) in the conjugate. In some embodiments, a molecule containing one or more azide groups may be used as a linker, in which the azide group may form a 1,2,3-triazole linkage with an alkyne in a component (e.g., an Fc domain) in the conjugate. In some embodiments, a molecule containing one or more bis-sulfone groups may be used as a linker, in which the bis-sulfone group may form a linkage with an amine group a component (e.g., an Fc domain) in the conjugate. In some embodiments, a molecule containing one or more sulfonic acid groups may be used as a linker, in which the sulfonic acid group may form a sulfonamide linkage with a component in the conjugate. In some embodiments, a molecule containing one or more isocyanate groups may be used as a linker, in which the isocyanate group may form a urea linkage with a component in the conjugate. In some embodiments, a molecule containing one or more haloalkyl groups may be used as a linker, in which the haloalkyl group may form a covalent linkage, e.g., C-N and C-O linkages, with a component in the conjugate.

In some embodiments, a linker provides space, rigidity, and/or flexibility between the two or more components. In some embodiments, a linker may be a bond, e.g., a covalent bond. The term“bond” refers to a chemical bond, e.g., an amide bond, a disulfide bond, a C-O bond, a C-N bond, a N-N bond, a C S f f embodiments, a linker includes no more than 250 atoms. In some embodim

more than 250 non-hydrogen atoms. In some embodiments, the backbone of a linker includes no more than 250 atoms. The“backbone” of a linker refers to the atoms in the linker that together form the shortest path from one part of a conjugate to another part of the conjugate (e.g., the shortest path linking a first cyclic heptapeptide and a second cyclic heptapeptide). The atoms in the backbone of the linker are directly involved in linking one part of a conjugate to another part of the conjugate (e.g., linking a first cyclic heptapeptide and a second cyclic heptapeptide). For examples, hydrogen atoms attached to carbons in the backbone of the linker are not considered as directly involved in linking one part of the conjugate to another part of the conjugate.

In some embodiments, a linker may comprise a synthetic group derived from, e.g., a synthetic polymer (e.g., a polyethylene glycol (PEG) polymer). In some embodiments, a linker may comprise one or more amino acid residues, such as D- or L-amino acid residues. In some embodiments, a linker may be a residue of an amino acid sequence (e.g., a 1-25 amino acid, 1-10 amino acid, 1-9 amino acid, 1-8 amino acid, 1-7 amino acid, 1-6 amino acid, 1-5 amino acid, 1-4 amino acid, 1-3 amino acid, 1-2 amino acid, or 1 amino acid sequence). In some embodiments, a linker may comprise one or more, e.g., 1-100, 1-50, 1-25, 1-10, 1-5, or 1-3, optionally substituted alkylene, optionally substituted heteroalkylene (e.g., a PEG unit), optionally substituted alkenylene, optionally substituted heteroalkenylene, optionally substituted alkynylene, optionally substituted heteroalkynylene, optionally substituted cycloalkylene, optionally substituted heterocycloalkylene, optionally substituted cycloalkenylene, optionally substituted heterocycloalkenylene, optionally substituted cycloalkynylene, optionally substituted

heterocycloalkynylene, optionally substituted arylene, optionally substituted heteroarylene (e.g., pyridine), O, S, NR i (R i is H, optionally substituted alkyl, optionally substituted heteroalkyl, optionally substituted alkenyl, optionally substituted heteroalkenyl, optionally substituted alkynyl, optionally substituted heteroalkynyl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl, optionally substituted cycloalkenyl, optionally substituted heterocycloalkenyl, optionally substituted cycloalkynyl, optionally substituted heterocycloalkynyl, optionally substituted aryl, or optionally substituted heteroaryl), P, carbonyl, thiocarbonyl, sulfonyl, phosphate, phosphoryl, or imino. For example, a linker may comprise one or more optionally substituted C1-C20 alkylene, optionally substituted C1-C20 heteroalkylene (e.g., a PEG unit), optionally substituted C2-C20 alkenylene (e.g., C2 alkenylene), optionally substituted C2-C20 heteroalkenylene, optionally substituted C2-C20 alkynylene, optionally substituted C2-C20

heteroalkynylene, optionally substituted C3-C20 cycloalkylene (e.g., cyclopropylene, cyclobutylene), optionally substituted C3-C20 heterocycloalkylene, optionally substituted C4-C20 cycloalkenylene, optionally substituted C4-C20 heterocycloalkenylene, optionally substituted C8-C20 cycloalkynylene, optionally substituted C8-C20 heterocycloalkynylene, optionally substituted C5-C15 arylene (e.g., C6 arylene), optionally substituted C2-C15 heteroarylene (e.g., imidazole, pyridine), O, S, NR i (R i is H, optionally substituted C1-C20 alkyl, optionally substituted C1-C20 heteroalkyl, optionally substituted C2- C20 alkenyl, optionally substituted C2-C20 heteroalkenyl, optionally substituted C2-C20 alkynyl, optionally substituted C2-C20 heteroalkynyl, optionally substituted C3-C20 cycloalkyl, optionally

C C C C C4-C20 heterocycloalkenyl, optionally substituted C8-C20 cycloalkynyl, opt

heterocycloalkynyl, optionally substituted C5-C15 aryl, or optionally substituted C2-C15 heteroaryl), P, carbonyl, thiocarbonyl, sulfonyl, phosphate, phosphoryl, or imino.

The term“lipophilic moiety,” as used herein, refers to a portion, substituent, or functional group of a compound that is, in general, hydrophobic and non-polar. A moiety is lipophilic if it has a hydrophobicity determined using a cLogP value of greater than 0, such as about 0.25 or greater, about 0.5 or greater, about 1 or greater, about 2 or greater, 0.25-5, 0.5-4 or 2-3. As used herein, the term“cLogP” refers to the calculated partition coefficient of a molecule or portion of a molecule. The partition coefficient is the ratio of concentrations of a compound in a mixture of two immiscible phases at equilibrium (e.g., octanol and water) and measures the hydrophobicity or hydrophilicity of a compound. A variety of methods are available in the art for determining cLogP. For example, in some embodiments, cLogP can be determined using quantitative structure-property relationship algorithims known in the art (e.g., using fragment based prediction methods that predict the logP of a compound by determining the sum of its non-overlapping molecular fragments). Several algorithims for calculating cLogP are known in the art including those used by molecular editing software such as CHEMDRAW® Pro, Version 12.0.2.1092 (Camrbridgesoft, Cambridge, MA) and MARVINSKETCH® (ChemAxon, Budapest, Hungary). A moiety is considered lipophilic if it has a cLogP value described above in at least one of the above methods. A lipophilic moiety having the stated cLogP value will be considered lipophilic, even though it may have a positive charge or a polar substituent.

In some embodiments, a lipophilic moiety contains entirely hydrocarbons. In some embodiments, a lipophilic moiety may contain one or more, e.g., 1-4, 1-3, 1, 2, 3, or 4, heteroatoms independently selected from N, O, and S (e.g., an indolyl), or one or more, e.g., 1-4, 1-3, 1, 2, 3, or 4, halo groups, which, due to the structure of the moiety and/or small differences in electronegativity between the heteroatoms or halo groups and the hydrocarbons, do not induce significant chemical polarity into the lipophilic moiety. Thus, in some embodiments, a lipophilic moiety having, e.g., 1-4, 1-3, 1, 2, 3, or 4, heteroatoms and/or, e.g., 1-4, 1-3, 1, 2, 3, or 4, halo atoms may still be considered non-polar. In some embodiments, a lipophilic moiety may be optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, optionally substituted heteroalkyl, optionally substituted heteroalkenyl, optionally substituted heteroalkynyl, or optionally substituted heteroaryl, or halo forms thereof, wherein the optional substituents are also lipophilic (such as alkyl, alkenyl, alkynyl, heteroalkyl, heteroalkenyl, heteroalkynyl, aryl, or heteroaryl) or are not lipophilic but do not change the overall lipophilic character of the moiety, i.e., the moiety has a cLogP value of greater than 0. For example, octanol contains a polar group, OH, but is still a lipophilic moiety. In some embodiments, a lipophilic moiety may be benzyl, isobutyl, sec-butyl, isopropyl, n-propyl, methyl, biphenylmethyl, n-octyl, or substituted indolyl (e.g., alkyl substituted indolyl). In some embodiments, a lipophilic moiety may be the side chain of a hydrophobic amino acid residue, e.g., leucine, isoleucine, alanine, phenylalanine, valine, and proline, or groups such as methyl, ethyl, propyl, isopropyl, butyl, isobutyl, and pyrrolidinyl. In some embodiments, lipophilic moieties of the conjugates described herein may interact with the hydrophobic f ( f f ) f bacterial cells (e.g., Gram-negative bacterial cells). Due to its position on the cyclic heptapeptide, one or more of R 1 , R 12 , R 15 , R’ 1 , R’ 12 , and R’ 15 may be a lipophilic moiety.

The term“positively charged moiety,” as used herein, refers to a portion, substituent, or functional group of a compound that contains at least one positive charge. In some embodiments, a positively charged moiety contains one or more (e.g., 1-4, 1-3, 1, 2, 3, or 4) heteroatoms independently selected from N, O, and S, for example. In some embodiments, a positively charged moiety may possess a pH- dependent positive charge, e.g., the moiety becomes a positively charged moiety at physiological pH (e.g., pH 7), such as -NH3 + , -(CH2)4NH2, -(CH2)3NH2, -(CH2)2NH2, -CH2NH2, -(CH2)4N(CH3)2,

-(CH 2 ) 3 N(CH 3 ) 2 , -(CH 2 ) 2 N(CH 3 ) 2 , -CH 2 N(CH 3 ) 2, -(CH 2 ) 4 NH(CH 3 ), -(CH 2 ) 3 NH(CH 3 ),-(CH 2 ) 2 NH(CH 3 ) , and -CH2NH(CH3). In some embodiments, a positively charged moiety may be optionally substituted alkamino, optionally substituted heteroalkyl (e.g., optionally substituted heteroalkyl containing 1-3 nitrogens; -(CH2)4-guanidinium, -(CH2)3-guanidinium, -(CH2)2-guanidinium, -CH2-guanidinium), optionally substituted heterocycloalkyl (e.g., optionally substituted heterocycloalkyl containing 1-3 nitrogens), or optionally substituted heteroaryl (e.g., optionally substituted heteroaryl containing 1-3

nitrogens; -(CH2)4-imidazole, -(CH2)3-imidazole, -(CH2)2-imidazole, -CH2-imidazole). In some

embodiments, a positively charged moiety may be pH independent such

as -CH 2 N(CH 3 ) 3 + , -(CH 2 ) 2 N(CH 3 ) 3 + , -(CH 2 ) 3 N(CH 3 ) 3 + , or -(CH 2 ) 4 N(CH 3 ) 3 + . Thus, substituents may transform an otherwise lipophilic moiety such as optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, optionally substituted heteroalkyl, optionally substituted heteroalkenyl, optionally substituted heteroalkynyl, or optionally substituted heteroaryl, or halo forms thereof, to a positively charged moiety with the addition of a substituent that imparts a positive charge or a pH dependent positive charge, such as guanidinyl, -NH3 + , -NH2, -NH(CH3), -N(CH3)2, and/or -N(CH3)3 + . In some embodiments, a positively charged moiety may be the side chain of an amino acid residue (e.g., a natural or non-natural amino acid residue, such as a D- or L-amino acid residue, that is positively charged at physiological pH (e.g., pH 7), such as the side chain of a basic amino acid residue (e.g., arginine, lysine, histidine, ornithine, diaminobuteric acid, or diaminopropionic acid). In some embodiments, positively charged moieties of the conjugates described herein interact with the negatively charged portions of lipid A (e.g., phosphates of lipid A) when the conjugates bind to the membrane of bacterial cells (e.g., Gram-negative bacterial cells). Due to its position on the cyclic heptapeptide, one or more of R 11 , R 13 , R 14 , R’ 11 , R’ 13 , and R’ 14 may be a positively charged moiety.

The term“polar moiety,” as used herein, refers to a portion, substituent, or functional group of a compound that has a chemical polarity induced by atoms with different electronegativity. The polarity of a polar moiety is dependent on the electronegativity between atoms within the moiety and the asymmetry of the structure of the moiety. In some embodiments, a polar moiety contains one or more (e.g., 1-4, 1-3, 1, 2, 3, or 4) heteroatoms independently selected from N, O, and S, which may induce chemical polarity in the moiety by having different electronegativity from carbon and hydrogen. In general, a polar moiety interacts with other polar or charged molecules. In some embodiments, a polar moiety may be optionally substituted alkamino, optionally substituted heteroalkyl (e.g., N- and/or O-containing

heteroalkyl; -(CH2)4-carboxylic acid, -(CH2)3-carboxylic acid, -(CH2)2-carboxylic acid, -CH2-carboxylic acid), optionally substituted heterocycloalkyl (e.g., N- and/or O-containing h

substituted heteroaryl (e.g., N- and/or O-containing heteroaryl). In some embodiments, a polar moiety may–CH(CH3)OH, -CH2OH, -(CH2)2CONH2, -CH2CONH2, -CH2COOH, or -(CH2)2COOH. Thus, substituents may transform an otherwise lipophilic moiety such optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, optionally substituted heteroalkyl, optionally substituted heteroalkenyl, optionally substituted heteroalkynyl, or optionally substituted heteroaryl, or halo forms thereof, to a polar moiety with the addition of a substituent that imparts polarity, such as -OH, -COOH, -COOR, or -CONR2, in which R is H or C1-C4 alkyl. In some embodiments, a polar moiety may be the side chain or a polar or charged amino acid residue (e.g., threonine, serine, glutamine, asparagine, arginine, lysine histidine, aspartic acid, and glutamic acid). In some embodiments, a polar moiety is the side chain of threonine. In some embodiments, polar moieties of the conjugates described herein interact with the negatively charged portions of lipid A (e.g., phosphates of lipid A) when the conjugates bind to the membrane of bacterial cells (e.g., Gram-negative bacterial cells). Due to its position on the cyclic heptapeptide, one or more of R 1 , R 12 , R 15 , R’ 1 , R’ 12 , and R’ 15 may be a polar moiety.

The term“polymyxin core,” as used herein means a cyclic heptapeptide having the structure:

,

wherein Q 1 , Q 2 and Q 3 are as follows:

attachment of the polymyxin core to the remainder of the conjugates disclosed herein, including the second polymyxin core (in conjugates that containg an Fc domain covalently linked to one or more dimers of polymyxin cores), the linker, and the Fc domain of the conjugates disclosed herein.

The terms“alkyl,”“alkenyl,” and“alkynyl,” as used herein, include straight-chain and branched- chain monovalent substituents, as well as combinations of these, containing only C and H when unsubstituted. When the alkyl group includes at least one carbon-carbon double bond or carbon-carbon triple bond, the alkyl group can be referred to as an“alkenyl” or“alkynyl” group respectively. The monovalency of an alkyl, alkenyl, or alkynyl group does not include the optional substituents on the alkyl, alkenyl, or alkynyl group. For example, if an alkyl, alkenyl, or alkynyl group is attached to a compound, monovalency of the alkyl, alkenyl, or alkynyl group refers to its attachment to the compound and does not include any additional substituents that may be present on the alkyl, alkenyl, or alkynyl group. In some embodiments, the alkyl or heteroalkyl group may contain, e.g., 1-20.1-18, 1-16, 1-14, 1-12, 1-10, 1-8, 1- 6, 1-4, or 1-2 carbon atoms (e.g., C1-C20, C1-C18, C1-C16, C1-C14, C1-C12, C1-C10, C1-C8, C1-C6, C1-C4, or C1-C2). In some embodiments, the alkenyl, heteroalkenyl, alkynyl, or heteroalkynyl group may contain, e.g., 2-20, 2-18, 2-16, 2-14, 2-12, 2-10, 2-8, 2-6, or 2-4 carbon atoms (e.g., C2-C20, C2-C18, C2-C16, C2-C14, C2-C12, C2-C10, C2-C8, C2-C6, or C2-C4). Examples include, but are not limited to, methyl, ethyl, isobutyl, sec-butyl, tert-butyl, 2-propenyl, and 3-butynyl.

The term“cycloalkyl,” as used herein, represents a monovalent saturated or unsaturated non- aromatic cyclic alkyl group. A cycloalkyl may have, e.g., three to twenty carbons (e.g., a C3-C7, C3-C8, C3-C9, C3-C10, C3-C11, C3-C12, C3-C14, C3-C16, C3-C18, or C3-C20 cycloalkyl). Examples of cycloalkyls include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, and cycloheptyl. When the cycloalkyl group includes at least one carbon-carbon double bond, the cycloalkyl group can be referred to as a“cycloalkenyl” group. A cycloalkenyl may have, e.g., four to twenty carbons (e.g., a C4- C7, C4-C8, C4-C9, C4-C10, C4-C11, C4-C12, C4-C14, C4-C16, C4-C18, or C4-C20 cycloalkenyl).

Exemplary cycloalkenyl groups include, but are not limited to, cyclopentenyl, cyclohexenyl, and cycloheptenyl. When the cycloalkyl group includes at least one carbon-carbon triple bond, the cycloalkyl group can be referred to as a“cycloalkynyl” group. A cycloalkynyl may have, e.g., eight to twenty carbons (e.g., a C8-C9, C8-C10, C8-C11, C8-C12, C8-C14, C8-C16, C8-C18, or C8-C20 cycloalkynyl). The term“cycloalkyl” also includes a cyclic compound having a bridged multicyclic structure in which one or more carbons bridges two non-adjacent members of a monocyclic ring, e.g., bicyclo[2.2.1.]heptyl and adamantane. The term“cycloalkyl” also includes bicyclic, tricyclic, and tetracyclic fused ring structures, e.g., decalin and spiro cyclic compounds.

The term“aryl,” as used herein, refers to any monocyclic or fused ring bicyclic or tricyclic system which has the characteristics of aromaticity in terms of electron distribution throughout the ring system, e g phenyl naphthyl or phenanthrene In some embodiments a ring system contains 5-15 ring member atoms or 5-10 ring member atoms. An aryl group may have, e.g., five to fift

C5-C7, C5-C8, C5-C9, C5-C10, C5-C11, C5-C12, C5-C13, C5-C14, or C5-C15 aryl). The term “heteroaryl” also refers to such monocyclic or fused bicyclic ring systems containing one or more, e.g., 1- 4, 1-3, 1, 2, 3, or 4, heteroatoms selected from O, S and N. A heteroaryl group may have, e.g., two to fifteen carbons (e.g., a C2-C3, C2-C4, C2-C5, C2-C6, C2-C7, C2-C8, C2-C9. C2-C10, C2-C11, C2-C12, C2-C13, C2-C14, or C2-C15 heteroaryl). The inclusion of a heteroatom permits inclusion of 5-membered rings to be considered aromatic as well as 6-membered rings. Thus, typical heteroaryl systems include, e.g., pyridyl, pyrimidyl, indolyl, benzimidazolyl, benzotriazolyl, isoquinolyl, quinolyl, benzothiazolyl, benzofuranyl, thienyl, furyl, pyrrolyl, thiazolyl, oxazolyl, isoxazolyl, benzoxazolyl, benzoisoxazolyl, and imidazolyl. Because tautomers are possible, a group such as phthalimido is also considered heteroaryl. In some embodiments, the aryl or heteroaryl group is a 5- or 6-membered aromatic rings system optionally containing 1-2 nitrogen atoms. In some embodiments, the aryl or heteroaryl group is an optionally substituted phenyl, pyridyl, indolyl, pyrimidyl, pyridazinyl, benzothiazolyl, benzimidazolyl, pyrazolyl, imidazolyl, isoxazolyl, thiazolyl, or imidazopyridinyl. In some embodiments, the aryl group is phenyl. In some embodiments, an aryl group may be optionally substituted with a substituent such an aryl substituent, e.g., biphenyl.

The term“alkaryl,” refers to an aryl group that is connected to an alkylene, alkenylene, or alkynylene group. In general, if a compound is attached to an alkaryl group, the alkylene, alkenylene, or alkynylene portion of the alkaryl is attached to the compound. In some embodiments, an alkaryl is C6- C35 alkaryl (e.g., C6-C16, C6-C14, C6-C12, C6-C10, C6-C9, C6-C8, C7, or C6 alkaryl), in which the number of carbons indicates the total number of carbons in both the aryl portion and the alkylene, alkenylene, or alkynylene portion of the alkaryl. Examples of alkaryls include, but are not limited to, (C1- C8)alkylene(C6-C12)aryl, (C2-C8)alkenylene(C6-C12)aryl, or (C2-C8)alkynylene(C6-C12)aryl. In some embodiments, an alkaryl is benzyl or phenethyl. In a heteroalkaryl, one or more heteroatoms selected from N, O, and S may be present in the alkylene, alkenylene, or alkynylene portion of the alkaryl group and/or may be present in the aryl portion of the alkaryl group. In an optionally substituted alkaryl, the substituent may be present on the alkylene, alkenylene, or alkynylene portion of the alkaryl group and/or may be present on the aryl portion of the alkaryl group.

The term“amino,” as used herein, represents–N(R x ) 2 or–N + (R x ) 3 , where each R x is, independently, H, alkyl, alkenyl, alkynyl, aryl, alkaryl, cycloalkyl, or two R x combine to form a

heterocycloalkyl. In some embodiment, the amino group is -NH2.

The term“alkamino,” as used herein, refers to an amino group, described herein, that is attached to an alkylene (e.g., C1-C5 alkylene), alkenylene (e.g., C2-C5 alkenylene), or alkynylene group (e.g., C2- C5 alkenylene). In general, if a compound is attached to an alkamino group, the alkylene, alkenylene, or alkynylene portion of the alkamino is attached to the compound. The amino portion of an alkamino refers to–N(R x )2 or–N + (R x )3, where each R x is, independently, H, alkyl, alkenyl, alkynyl, aryl, alkaryl, cycloalkyl, or two R x combine to form a heterocycloalkyl. In some embodiment, the amino portion of an alkamino is -NH2. An example of an alkamino group is C1-C5 alkamino, e.g., C2 alkamino (e.g., CH2CH2NH2 or C C (C ) ) selected from N, O, and S may be present in the alkylene, alkenylene, or al

heteroalkamino group. In some embodiments, an alkamino group may be optionally substituted. In a substituted alkamino group, the substituent may be present on the alkylene, alkenylene, or alkynylene portion of the alkamino group and/or may be present on the amino portion of the alkamino group.

T he term“alkamide,” as used herein, refers to an amide group that is attached to an alkylene (e.g., C1-C5 alkylene), alkenylene (e.g., C2-C5 alkenylene), or alkynylene (e.g., C2-C5 alkenylene) group. In general, if a compound is attached to an alkamide group, the alkylene, alkenylene, or alkynylene portion of the alkamide is attached to the compound. The amide portion of an alkamide refers to–C(O)-N(R x ) 2 , where each R x is, independently, H, alkyl, alkenyl, alkynyl, aryl, alkaryl, cycloalkyl, or two R x combine to form a heterocycloalkyl. In some embodiment, the amide portion of an alkamide is -C(O)NH2. An alkamide group may be -(CH2)2-C(O)NH2 or -CH2-C(O)NH2. In a heteroalkamide group, one or more, e.g., 1-4, 1-3, 1, 2, 3, or 4, heteroatoms selected from N, O, and S may be present in the alkylene, alkenylene, or alkynylene portion of the heteroalkamide group. In some embodiments, an alkamide group may be optionally substituted. In a substituted alkamide group, the substituent may be present on the alkylene, alkenylene, or alkynylene portion of the alkamide group and/or may be present on the amide portion of the alkamide group.

The terms“alkylene,”“alkenylene,” and“alkynylene,” as used herein, refer to divalent groups having a specified size. In some embodiments, an alkylene may contain, e.g., 1-20, 1-18, 1-16, 1-14, 1- 12, 1-10, 1-8, 1-6, 1-4, or 1-2 carbon atoms (e.g., C1-C20, C1-C18, C1-C16, C1-C14, C1-C12, C1-C10, C1-C8, C1-C6, C1-C4, or C1-C2). In some embodiments, an alkenylene or alkynylene may contain, e.g., 2-20, 2-18, 2-16, 2-14, 2-12, 2-10, 2-8, 2-6, or 2-4 carbon atoms (e.g., C2-C20, C2-C18, C2-C16, C2- C14, C2-C12, C2-C10, C2-C8, C2-C6, or C2-C4). Alkylene, alkenylene, and/or alkynylene includes straight-chain and branched-chain forms, as well as combinations of these. The divalency of an alkylene, alkenylene, or alkynylene group does not include the optional substituents on the alkylene, alkenylene, or alkynylene group. For example, two cyclic heptapeptides may be attached to each other by way of a linker that includes alkylene, alkenylene, and/or alkynylene, or combinations thereof. Each of the alkylene, alkenylene, and/or alkynylene groups in the linker is considered divalent with respect to the two attachments on either end of alkylene, alkenylene, and/or alkynylene group. For example, if a linker includes -(optionally substituted alkylene)-(optionally substituted alkenylene)-(optionally substituted alkylene)-, the alkenylene is considered divalent with respect to its attachments to the two alkylenes at the ends of the linker. The optional substituents on the alkenylene are not included in the divalency of the alkenylene. The divalent nature of an alkylene, alkenylene, or alkynylene group (e.g., an alkylene, alkenylene, or alkynylene group in a linker) refers to both of the ends of the group and does not include optional substituents that may be present in an alkylene, alkenylene, or alkynylene group. Because they are divalent, they can link together multiple (e.g., two) parts of a conjugate, e.g., a first cyclic heptapeptide and a second cyclic heptapeptide. Alkylene, alkenylene, and/or alkynylene groups can be substituted by the groups typically suitable as substituents for alkyl, alkenyl and alkynyl groups as set forth herein. For example, C=O is a C1 alkylene that is substituted by an oxo (=O). For example, -HCR-C≡C- may be an optional substituent, R. Heteroalkylene, heteroalkenylene, and/or hetero

alkylene, alkenylene, and/or alkynylene groups including one or more, e.g., 1-4, 1-3, 1, 2, 3, or 4, heteroatoms, e.g., N, O, and S. For example, a polyethylene glycol (PEG) polymer or a PEG unit -(CH2)2- O- in a PEG polymer is considered a heteroalkylene containing one or more oxygen atoms.

T he term“cycloalkylene,” as used herein, refers to a divalent cyclic group linking together two parts of a compound. For example, one carbon within the cycloalkylene group may be linked to one part of the compound, while another carbon within the cycloalkylene group may be linked to another part of the compound. A cycloalkylene group may include saturated or unsaturated non-aromatic cyclic groups. A cycloalkylene may have, e.g., three to twenty carbons in the cyclic portion of the cycloalkylene (e.g., a C3-C7, C3-C8, C3-C9, C3-C10, C3-C11, C3-C12, C3-C14, C3-C16, C3-C18, or C3-C20 cycloalkylene). When the cycloalkylene group includes at least one carbon-carbon double bond, the cycloalkylene group can be referred to as a“cycloalkenylene” group. A cycloalkenylene may have, e.g., four to twenty carbons in the cyclic portion of the cycloalkenylene (e.g., a C4-C7, C4-C8, C4-C9. C4-C10, C4-C11, C4- C12, C4-C14, C4-C16, C4-C18, or C4-C20 cycloalkenylene). When the cycloalkylene group includes at least one carbon-carbon triple bond, the cycloalkylene group can be referred to as a“cycloalkynylene” group. A cycloalkynylene may have, e.g., four to twenty carbons in the cyclic portion of the

cycloalkynylene (e.g., a C4-C7, C4-C8, C4-C9. C4-C10, C4-C11, C4-C12, C4-C14, C4-C16, C4-C18, or C8-C20 cycloalkynylene). A cycloalkylene group can be substituted by the groups typically suitable as substituents for alkyl, alkenyl and alkynyl groups as set forth herein. Heterocycloalkylene refers to a cycloalkylene group including one or more, e.g., 1-4, 1-3, 1, 2, 3, or 4, heteroatoms, e.g., N, O, and S. Examples of cycloalkylenes include, but are not limited to, cyclopropylene and cyclobutylene. A tetrahydrofuran may be considered as a heterocycloalkylene.

The term“arylene,” as used herein, refers to a multivalent (e.g., divalent or trivalent) aryl group linking together multiple (e.g., two or three) parts of a compound. For example, one carbon within the arylene group may be linked to one part of the compound, while another carbon within the arylene group may be linked to another part of the compound. An arylene may have, e.g., five to fifteen carbons in the aryl portion of the arylene (e.g., a C5-C6, C5-C7, C5-C8, C5-C9. C5-C10, C5-C11, C5-C12, C5-C13, C5- C14, or C5-C15 arylene). An arylene group can be substituted by the groups typically suitable as substituents for alkyl, alkenyl and alkynyl groups as set forth herein. Heteroarylene refers to an aromatic group including one or more, e.g., 1-4, 1-3, 1, 2, 3, or 4, heteroatoms, e.g., N, O, and S. A heteroarylene group may have, e.g., two to fifteen carbons (e.g., a C2-C3, C2-C4, C2-C5, C2-C6, C2-C7, C2-C8, C2- C9. C2-C10, C2-C11, C2-C12, C2-C13, C2-C14, or C2-C15 heteroarylene).

The term“optionally substituted,” as used herein, refers to having 0, 1, or more substituents, such as 0-25, 0-20, 0-10 or 0-5 substituents. Substituents include, but are not limited to, alkyl, alkenyl, alkynyl, aryl, alkaryl, acyl, heteroaryl, heteroalkyl, heteroalkenyl, heteroalkynyl, heteroalkaryl, halogen, oxo, cyano, nitro, amino, alkamino, hydroxy, alkoxy, alkanoyl, carbonyl, carbamoyl, guanidinyl, ureido, amidinyl, any of the groups or moieties described above, and hetero versions of any of the groups or moieties described above. Substituents include, but are not limited to, F, Cl, methyl, phenyl, benzyl, OR, S SO SO OCO CO CO COO OCO CO COO OOC SO 3 R, CONR 2 , SO 2 NR 2 , NRSO 2 NR 2 , CN, CF 3 , OCF 3 , SiR 3 , and NO 2 , wher

H, alkyl, alkenyl, aryl, heteroalkyl, heteroalkenyl, or heteroaryl, and wherein two of the optional substituents on the same or adjacent atoms can be joined to form a fused, optionally substituted aromatic or nonaromatic, saturated or unsaturated ring which contains 3–8 members, or two of the optional substituents on the same atom can be joined to form an optionally substituted aromatic or nonaromatic, saturated or unsaturated ring which contains 3–8 members.

An optionally substituted group or moiety refers to a group or moiety (e.g., any one of the groups or moieties described above) in which one of the atoms (e.g., a hydrogen atom) is optionally replaced with another substituent. For example, an optionally substituted alkyl may be an optionally substituted methyl, in which a hydrogen atom of the methyl group is replaced by, e.g., OH. As another example, a substituent on a heteroalkyl or its divalent counterpart, heteroalkylene, may replace a hydrogen on a carbon or a hydrogen on a heteroatom such as N. For example, the hydrogen atom in the

group -R-NH-R- may be substituted with an alkamide substituent, e.g., -R-N[(CH 2 C(O)N(CH 3 ) 2 ]-R.

Generally, an optional substituent is a noninterfering substituent. A“noninterfering substituent” refers to a substituent that leaves the ability of the conjugates described herein (e.g., conjugates of any one of formulas (I)-(XXXIII)) to either bind to lipopolysaccharides (LPS) or to kill or inhibit the growth of Gram- negative bacteria qualitatively intact. Thus, in some embodiments, the substituent may alter the degree of such activity. However, as long as the conjugate retains the ability to bind to LPS and/or to kill or inhibit the growth of Gram-negative bacteria, the substituent will be classified as“noninterfering.” In some aspects, a noninterfering substituent leaves the ability of a conjugate described herein (e.g., a conjugate of any one of formulas (I)-(XXXIII)) to kill or inhibit the growth of Gram-negative bacteria qualitatively intact as determined by measuring the minimum inhibitory concentration (MIC) against at least one Gram- negative bacteria as known in the art, wherein the MIC is 128 µg/mL or less. In some aspects, a noninterfering substituent leaves the ability of a conjugate described herein (e.g., a conjugate of any one of formulas (I)-(XXXIII)) to bind to lipopolysaccharides (LPS) from the cell membrane of Gram-negative bacteria qualitatively intact, as determined by an LPS binding assay, wherein the conjugate shows a value of about 10% or greater displacement of a fluorogenic substrate at 250 µM of the conjugate.

The term“hetero,” when used to describe a chemical group or moiety, refers to having at least one heteroatom that is not a carbon or a hydrogen, e.g., N, O, and S. Any one of the groups or moieties described above may be referred to as hetero if it contains at least one heteroatom. For example, a heterocycloalkyl, heterocycloalkenyl, or heterocycloalkynyl group refers to a cycloalkyl, cycloalkenyl, or cycloalkynyl group that has one or more heteroatoms independently selected from, e.g., N, O, and S. An example of a heterocycloalkenyl group is a maleimido. For example, a heteroaryl group refers to an aromatic group that has one or more heteroatoms independently selected from, e.g., N, O, and S. One or more heteroatoms may also be included in a substituent that replaced a hydrogen atom in a group or moiety as described herein. For example, in an optionally substituted heteroaryl group, if one of the hydrogen atoms in the heteroaryl group is replaced with a substituent (e.g., methyl), the substituent may also contain one or more heteroatoms (e.g., methanol). The term“acyl,” as used herein, refers to a group having the structure: , wherein R z is an optionally substituted alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, alkaryl, alkamino, heteroalkyl, heteroalkenyl, heteroalkynyl, heterocycloalkyl, heterocycloalkenyl,

heterocycloalkynyl, heteroaryl, heteroalkaryl, or heteroalkamino.

The term“halo” or“halogen,” as used herein, refers to any halogen atom, e.g., F, Cl, Br, or I. Any one of the groups or moieties described herein may be referred to as a“halo moiety” if it contains at least one halogen atom, such as haloalkyl.

The term“hydroxyl,” as used herein, represents an -OH group.

The term“oxo,” as used herein, refers to a substituent having the structure =O, where there is a d ouble bond between an atom and an oxygen atom. The term“carbonyl,” as used herein, refers to a group having the structure: The term“thiocarbonyl,” as used herein, refers to a group having the structure:

The term“phosphate,” as used herein, represents the group having the structure: .

The term“phosphoryl,” as used herein, represents the group having the structure: . The term“sulfonyl,” as used herein, represents the group having the structure: . The term“imino,” as used herein, represents the group having the structure: wherein

R is an optional substituent.

The term“N-protecting group,” as used herein, represents those groups intended to protect an a mino group against undesirable reactions during synthetic procedures. Commonly used N-protecting groups are disclosed in Greene,“Protective Groups in Organic Synthesis,” 5th Edition (John Wiley & Sons, New York, 2014), which is incorporated herein by reference. N-protecting groups include, e.g., acyl, aryloyl, and carbamyl groups such as formyl, acetyl, propionyl, pivaloyl, t-butylacetyl, 2-chloroacetyl, 2-bromoacetyl, trifluoroacetyl, trichloroacetyl, phthalyl, o-nitrophenoxyacetyl, α-chlorobutyryl, benzoyl, carboxybenzyl (CBz), 4-chlorobenzoyl, 4-bromobenzoyl, 4-nitrobenzoyl, and chiral auxiliaries such as protected or unprotected D, L or D, L-amino acid residues such as alanine,

sulfonyl-containing groups such as benzenesulfonyl and p-toluenesulfonyl; carbamate forming groups such as benzyloxycarbonyl, p-chlorobenzyloxycarbonyl, p-methoxybenzyloxycarbonyl, p- nitrobenzyloxycarbonyl, 2-nitrobenzyloxycarbonyl, p-bromobenzyloxycarbonyl, 3,4- dimethoxybenzyloxycarbonyl, 3,5-dimethoxybenzyl oxycarbonyl, 2,4-dimethoxybenzyloxycarbonyl, 4-methoxybenzyloxycarbonyl, 2-nitro-4,5-dimethoxybenzyloxycarbonyl,

3,4,5-trimethoxybenzyloxycarbonyl, 1-(p-biphenylyl)-1-methylethoxycarbonyl, α,α-dimethyl- 3,5-dimethoxybenzyloxycarbonyl, benzhydryloxy carbonyl, t-butyloxycarbonyl (BOC),

diisopropylmethoxycarbonyl, isopropyloxycarbonyl, ethoxycarbonyl, methoxycarbonyl, allyloxycarbonyl, 2,2,2,-trichloroethoxycarbonyl, phenoxycarbonyl, 4-nitrophenoxy carbonyl, fluorenyl-9-methoxycarbonyl (Fmoc), cyclopentyloxycarbonyl, adamantyloxycarbonyl, cyclohexyloxycarbonyl, and phenylthiocarbonyl; alkaryl groups such as benzyl, triphenylmethyl, and benzyloxymethyl; and silyl groups such as trimethylsilyl.

The term“amino acid,” as used herein, means naturally occurring amino acids and non-naturally occurring amino acids.

The term“naturally occurring amino acids,” as used herein, means amino acids including Ala, Arg, Asn, Asp, Cys, Gln, Glu, Gly, His, Ile, Leu, Lys, Met, Phe, Pro, Ser, Thr, Trp, Tyr, and Val.

The term“non-naturally occurring amino acid,” as used herein, means an alpha amino acid that is not naturally produced or found in a mammal. Examples of non-naturally occurring amino acids include D-amino acids; an amino acid having an acetylaminomethyl group attached to a sulfur atom of a cysteine; a pegylated amino acid; the omega amino acids of the formula NH 2 (CH 2 ) n COOH where n is 2-6, neutral nonpolar amino acids, such as sarcosine, t-butyl alanine, t-butyl glycine, N-methyl isoleucine, and norleucine; oxymethionine; phenylglycine; citrulline; methionine sulfoxide; cysteic acid; ornithine;

diaminobutyric acid; 3-aminoalanine; 3-hydroxy-D-proline; 2,4-diaminobutyric acid; 2-aminopentanoic acid; 2-aminooctanoic acid, 2-carboxy piperazine; piperazine-2-carboxylic acid, 2-amino-4-phenylbutanoic acid; 3-(2-naphthyl)alanine, and hydroxyproline. Other amino acids are α-aminobutyric acid, α-amino-α- methylbutyrate, aminocyclopropane-carboxylate, aminoisobutyric acid, aminonorbornyl-carboxylate, L- cyclohexylalanine, cyclopentylalanine, L-N-methylleucine, L-N-methylmethionine, L-N-methylnorvaline, L- N-methylphenylalanine, L-N-methylproline, L-N-methylserine, L-N-methyltryptophan, D-ornithine, L-N- methylethylglycine, L-norleucine, α-methyl-aminoisobutyrate, α-methylcyclohexylalanine, D-α- methylalanine, D-α-methylarginine, D-α-methylasparagine, D-α-methylaspartate, D-α-methylcysteine, D- α-methylglutamine, D-α-methylhistidine, D-α-methylisoleucine, D-α-methylleucine, D-α-methyllysine, D-α- methylmethionine, D-α-methylornithine, D-α-methylphenylalanine, D-α-methylproline, D-α-methylserine, D-N-methylserine, D-α-methylthreonine, D-α-methyltryptophan, D-α-methyltyrosine, D-α-methylvaline, D- N-methylalanine, D-N-methylarginine, D-N-methylasparagine, D-N-methylaspartate, D-N-methylcysteine, D-N-methylglutamine, D-N-methylglutamate, D-N-methylhistidine, D-N-methylisoleucine, D-N- methylleucine, D-N-methyllysine, N-methylcyclohexylalanine, D-N-methylornithine, N-methylglycine, N- methylaminoisobutyrate, N-(1-methylpropyl)glycine, N-(2-methylpropyl)glycine, D-N-methyltryptophan, D- homophenylalanine, L-α-methylarginine, L-α-methylaspartate, L-α-methylcy

L-α-methylhistidine, L-α-methylisoleucine, L-α-methylleucine, L-α-methylmethionine, L-α-methylnorvaline, L-α-methylphenylalanine, L-α-methylserine, L-α-methyltryptophan, L-α-methylvaline, N-(N-(2,2- diphenylethyl) carbamylmethylglycine, 1-carboxy-1-(2,2-diphenyl-ethylamino) cyclopropane, 4- hydroxyproline, ornithine, 2-aminobenzoyl (anthraniloyl), D-cyclohexylalanine, 4-phenyl-phenylalanine, L- citrulline, α-cyclohexylglycine, L-1,2,3,4-tetrahydroisoquinoline-3-carboxylic acid, L-thiazolidine-4- carboxylic acid, L-homotyrosine, L-2-furylalanine, L-histidine (3-methyl), N-(3-guanidinopropyl)glycine, O- methyl-L-tyrosine, O-glycan-serine, meta-tyrosine, nor-tyrosine, L-N,N′,N″-trimethyllysine, homolysine, norlysine, N-glycan asparagine, 7-hydroxy-1,2,3,4-tetrahydro-4-fluorophenylalanine, 4- methylphenylalanine, bis-(2-picolyl)amine, pentafluorophenylalanine, indoline-2-carboxylic acid, 2- aminobenzoic acid, 3-amino-2-naphthoic acid, asymmetric dimethylarginine, L-tetrahydroisoquinoline-1- carboxylic acid, D-tetrahydroisoquinoline-1-carboxylic acid, 1-amino-cyclohexane acetic acid, D/L- allylglycine, 4-aminobenzoic acid, 1-amino-cyclobutane carboxylic acid, 2 or 3 or 4-aminocyclohexane carboxylic acid, 1-amino-1-cyclopentane carboxylic acid, 1-aminoindane-1-carboxylic acid, 4-amino- pyrrolidine-2-carboxylic acid, 2-aminotetraline-2-carboxylic acid, azetidine-3-carboxylic acid, 4-benzyl- pyrolidine-2-carboxylic acid, tert-butylglycine, b-(benzothiazolyl-2-yl)-alanine, b-cyclopropyl alanine, 5,5- dimethyl-1,3-thiazolidine-4-carboxylic acid, (2R,4S)4-hydroxypiperidine-2-carboxylic acid, (2S,4S) and (2S,4R)-4-(2-naphthylmethoxy)-pyrolidine-2-carboxylic acid, (2S,4S) and (2S,4R)4-phenoxy-pyrrolidine-2- carboxylic acid, (2R,5S)and(2S,5R)-5-phenyl-pyrrolidine-2-carboxylic acid, (2S,4S)-4-amino-1-benzoyl- pyrrolidine-2-carboxylic acid, t-butylalanine, (2S,5R)-5-phenyl-pyrrolidine-2-carboxylic acid, 1- aminomethyl-cyclohexane-acetic acid, 3,5-bis-(2-amino)ethoxy-benzoic acid, 3,5-diamino-benzoic acid, 2- methylamino-benzoic acid, N-methylanthranylic acid, L-N-methylalanine, L-N-methylarginine, L-N- methylasparagine, L-N-methylaspartic acid, L-N-methylcysteine, L-N-methylglutamine, L-N- methylglutamic acid, L-N-methylhistidine, L-N-methylisoleucine, L-N-methyllysine, L-N-methylnorleucine, L-N-methylornithine, L-N-methylthreonine, L-N-methyltyrosine, L-N-methylvaline, L-N-methyl-t- butylglycine, L-norvaline, α-methyl-γ-aminobutyrate, 4,4′-biphenylalanine, α-methylcylcopentylalanine, α- methyl-α-napthylalanine, α-methylpenicillamine, N-(4-aminobutyl)glycine, N-(2-aminoethyl)glycine, N-(3- aminopropyl)glycine, N-amino-α-methylbutyrate, α-napthylalanine, N-benzylglycine, N-(2- carbamylethyl)glycine, N-(carbamylmethyl)glycine, N-(2-carboxyethyl)glycine, N-(carboxymethyl)glycine, N-cyclobutylglycine, N-cyclodecylglycine, N-cycloheptylglycine, N-cyclohexylglycine, N-cyclodecylglycine, N-cylcododecylglycine, N-cyclooctylglycine, N-cyclopropylglycine, N-cycloundecylglycine, N-(2,2- diphenylethyl)glycine, N-(3,3-diphenylpropyl)glycine, N-(3-guanidinopropyl)glycine, N-(1- hydroxyethyl)glycine, N-(hydroxyethyl))glycine, N-(imidazolylethyl))glycine, N-(3-indolylyethyl)glycine, N- methyl-γ-aminobutyrate, D-N-methylmethionine, N-methylcyclopentylalanine, D-N-methylphenylalanine, D-N-methylproline, D-N-methylthreonine, N-(1-methylethyl)glycine, N-methyl-napthylalanine, N- methylpenicillamine, N-(p-hydroxyphenyl)glycine, N-(thiomethyl)glycine, penicillamine, L-α-methylalanine, L-α-methylasparagine, L-α-methyl-t-butylglycine, L-methylethylglycine, L-α-methylglutamate, L-α- methylhomophenylalanine, N-(2-methylthioethyl)glycine, L-α-methyllysine, L-α-methylnorleucine, L-α- homophenylalanine, N-(N-(3,3-diphenylpropyl) carbamylmethylglycine, L-py

pyroglutamic acid, O-methyl-L-serine, O-methyl-L-homoserine, 5-hydroxylysine, α-carboxyglutamate, phenylglycine, L-pipecolic acid (homoproline), L-homoleucine, L-lysine (dimethyl), L-2-naphthylalanine, L- dimethyldopa or L-dimethoxy-phenylalanine, L-3-pyridylalanine, L-histidine (benzoyloxymethyl), N- cycloheptylglycine, L-diphenylalanine, O-methyl-L-homotyrosine, L-β-homolysine, O-glycan-threoine, Ortho-tyrosine, L-N,N′-dimethyllysine, L-homoarginine, neotryptophan, 3-benzothienylalanine, isoquinoline-3-carboxylic acid, diaminopropionic acid, homocysteine, 3,4-dimethoxyphenylalanine, 4- chlorophenylalanine, L-1,2,3,4-tetrahydronorharman-3-carboxylic acid, adamantylalanine, symmetrical dimethylarginine, 3-carboxythiomorpholine, D-1,2,3,4-tetrahydronorharman-3-carboxylic acid, 3- aminobenzoic acid, 3-amino-1-carboxymethyl-pyridin-2-one, 1-amino-1-cyclohexane carboxylic acid, 2- aminocyclopentane carboxylic acid, 1-amino-1-cyclopropane carboxylic acid, 2-aminoindane-2-carboxylic acid, 4-amino-tetrahydrothiopyran-4-carboxylic acid, azetidine-2-carboxylic acid, b-(benzothiazol-2-yl)- alanine, neopentylglycine, 2-carboxymethyl piperidine, b-cyclobutyl alanine, allylglycine, diaminopropionic acid, homo-cyclohexyl alanine, (2S,4R)- 4-hydroxypiperidine-2-carboxylic acid, octahydroindole-2- carboxylic acid, (2S,4R) and (2S,4R)-4-(2-naphthyl), pyrrolidine-2-carboxylic acid, nipecotic acid, (2S,4R)and (2S,4S)-4-(4-phenylbenzyl) pyrrolidine-2-carboxylic acid, (3S)-1-pyrrolidine-3-carboxylic acid, (2S,4S)-4-tritylmercapto-pyrrolidine-2-carboxylic acid, (2S,4S)-4-mercaptoproline, t-butylglycine, N,N- bis(3-aminopropyl)glycine, 1-amino-cyclohexane-1-carboxylic acid, N-mercaptoethylglycine, and selenocysteine. In some embodiments, amino acid residues may be charged or polar. Charged amino acids include alanine, lysine, aspartic acid, or glutamic acid, or non-naturally occurring analogs thereof. Polar amino acids include glutamine, asparagine, histidine, serine, threonine, tyrosine, methionine, or tryptophan, or non-naturally occurring analogs thereof.

It is specifically contemplated that in some embodiments, a terminal amino group in the amino acid may be an amido group or a carbamate group.

T he term“antibacterial agent,” as used herein, refers to an agent that is used in addition to one or more of the conjugates described herein (e.g., conjugates of any one of formulas (I)-(XXXIII)) in methods of treating a bacterial infection (e.g., Gram-negative bacterial infection) and/or preventing, stabilizing, or inhibiting the growth of bacteria, or killing bacteria. An antibacterial agent may be an agent that prevents the entrance of a bacteria (e.g., a Gram-negative bacteria) into a subject’s cells, tissues, or organs, inhibits the growth of a bacteria (e.g., a Gram-negative bacteria) in a subject’s cells, tissues, or organs, and/or kills a bacteria (e.g., a Gram-negative bacteria) that is inside a subject’s cells, tissues, or organs. Examples of antibacterial agents are described in detail further herein. In some embodiments, an antibacterial agent used in addition to a conjugate described herein is linezolid or tedizolid (e.g., tedizolid phosphate).

The term“bacterial infection,” as used herein, refers to the invasion of a subject’s cells, tissues, and/or organs by bacteria (e.g., Gram-negative bacteria), thus, causing an infection. In some

embodiments, the bacteria may grow, multiply, and/or produce toxins in the subject’s cells, tissues, and/or organs. In some embodiments, a bacterial infection can be any situation in which the presence of a

( ) “ ff ” f bacterial infection when a latent bacterial population is detectable in or on t

excessive amount of a bacterial population is present in or on the subject’s body, or when the presence of a bacterial population(s) is damaging the cells, tissues, and/or organs of the subject.

The term“protecting against,” as used herein, refers to preventing a subject from developing a bacterial infection (e.g., a Gram-negative bacterial infection) or decreasing the risk that a subject may develop a bacterial infection (e.g., a Gram-negative bacterial infection). Prophylactic drugs used in methods of protecting against a bacterial infection in a subject are often administered to the subject prior to any detection of the bacterial infection. In some embodiments of methods of protecting against a bacterial infection, a subject (e.g., a subject at risk of developing a bacterial infection) may be

administered a conjugate described herein (e.g., a conjugate having any one of formulas (I)-(XXXIII)) to prevent the bacterial infection development or decrease the risk of the bacterial infection development.

The term“treating” or“to treat,” as used herein, refers to a therapeutic treatment of a bacterial infection (e.g., a Gram-negative bacterial infection) in a subject. In some embodiments, a therapeutic treatment may slow the progression of the bacterial infection, improve the subject’s outcome, and/or eliminate the infection. In some embodiments, a therapeutic treatment of a bacterial infection (e.g., a Gram-negative bacterial infection) in a subject may alleviate or ameliorate of one or more symptoms or conditions associated with the bacterial infection, diminish the extent of the bacterial infection, stabilize (i.e., not worsening) the state of the bacterial infection, prevent the spread of the bacterial infection, and/or delay or slow the progress of the bacterial infection, as compare the state and/or the condition of the bacterial infection in the absence of the therapeutic treatment.

The phrase“LPS-induced nitric oxide (NO) production from a macrophage,” as used herein, refers to the ability of the lipopolysaccharides (LPS) in Gram-negative bacteria to activate a macrophage and induce NO production from the macrophage. NO production from a macrophage in response to LPS is a signal of macrophage activation, which may lead to sepsis in a subject, e.g., a Gram-negative bacteria infected subject. The disclosure features conjugates (e.g., conjugates of any one of formulas (I)- (XXXIII)) that are able to bind to LPS in the cell membrane of Gram-negative bacteria to disrupt and permeabilize the cell membrane, thus neutralizing an immune response to LPS. NO production from a macrophage may be measured using available techniques in the art, e.g., a Griess assay.

The term“resistant strain of bacteria,” as used herein, refers to a strain of bacteria (e.g., Gram- negative or Gram-positive bacteria) that is refractory to treatment with an antibiotic, such as an antibiotic described in the Detailed Description. Antibiotics that a strain of bacteria is resistant to do not include the conjugates described herein (e.g., conjugates of any one of formulas (I)-(XXXIII)). Resistance may arise through natural resistance in certain types of bacteria, spontaneous random genetic mutations, and/or by inter- or intra-species horizontal transfer of resistance genes. In some embodiments, a resistant strain of bacteria contains a mcr-1 gene and/or a mcr-2 gene. In some embodiments, a resistant strain of bacteria contains a chromosomal mutation conferring polymyxin resistance. In some embodiments, a resistant strain of bacteria contains a mcr-1 gene and/or a mcr-2 gene in combination with other antibiotic resistance genes. In some embodiments, a resistant strain of bacteria is a resistant strain of E. coli (e.g.,

) The term "activating an immune cell," as used herein, refers to the ability of a conjugate to bind to an immune cell to produce an effective immune response. The ability of a conjugate to directly or indirectly bind to an immune cell to produce an effective immune response may be quantified by measuring the concentration of the conjugate at which such immune response is produced. In some embodiments, the concentration of a conjugate that binds to an FcyR receptor on an immune cell to trigger an effective immune response may be less than or equal to 1 0,000 nM as measured in accordance with, e.g., an enzyme-linked immunosorbent assay (ELISA). In some embodiments, the concentration of a conjugate that binds to an immune cell receptor to trigger an effective immune response may be less than or equal to 1 000 nM or less than or equal to 100 nM as measured in accordance with an ELISA. In an ELISA, an FcyR may be immobilized on a support or surface using conventional techniques in the art. After the FcyR is immobilized to the surface, a conjugate described herein may be applied over the surface so it is captured by the FcyR through binding of the Fc domain in the conjugate to the FcyR. In some embodiments, the conjugate may be detected using a secondary antibody, which is linked to an enzyme (e.g., horseradish peroxidase) for subsequent signal amplification. During signal amplification, the enzyme's substrate (e.g., 3,3'-diaminobenzidine) may be added to produce a measurable signal (e.g., color change).

The term "average value of T," as used herein, refers to the mean number of monomers of cyclic heptapeptides or dimers of cyclic heptapeptides conjugated to an Fc domain monomer within a population of conjugates. In some embodiments, within a population of conjugates, the average number of monomers of cyclic heptapeptides or dimers of cyclic heptapeptides conjugated to an Fc domain monomer may be from 1 to 5 (e.g., 1 to 2).

The term "subject," as used herein, can be a human, non-human primate, or other mammal, such as but not limited to dog, cat, horse, cow, pig, turkey, goat, fish, monkey, chicken, rat, mouse, and sheep.

The term "substantially simultaneously," as used herein, refers to two or more events that occur at the same time or within a narrow time frame of each other. As disclosed herein, a conjugate described herein (e.g., a conjugate of any one of formulas (l)-(XXXIII)) and an antibacterial agent (e.g., linezolid or tedizolid) may be administered substantially simultaneously, which means that the conjugate and the antibacterial agent are administered together (e.g., in one pharmaceutical composition) or separately but within a narrow time frame of each other, e.g., within 10 minutes, e.g., 1 0, 9, 8, 7, 6, 5, 4, 3, 2, or 1 minute, or 45, 30, 15, or 1 0 seconds of each other.

The term "therapeutically effective amount," as used herein, refers to an amount, e.g., pharmaceutical dose, effective in inducing a desired effect in a subject or in treating a subject having a condition or disorder described herein (e.g., a bacterial infection (e.g., a Gram-negative bacterial infection)). It is also to be understood herein that a "therapeutically effective amount" may be interpreted as an amount giving a desired therapeutic and/or preventative effect, taken in one or more doses or in any dosage or route, and/or taken alone or in combination with other therapeutic agents (e.g., an antibacterial agent described herein). For example, in the context of administering a conjugate described herein (e.g., a conjugate of any one of formulas (l)-(XXXIII)) that is used for the treatment of a bacterial infection, an effective amount of a conjugate is, for example, an amount sufficient to prevent, slow down, or reverse the progression of the bacterial infection as compared to the res

administration of the conjugate.

As used herein, the term“pharmaceutical composition” refers to a medicinal or pharmaceutical formulation that contains at least one active ingredient (e.g., a conjugate of any one of formulas (I)- (XXXIII)) as well as one or more excipients and diluents to enable the active ingredient suitable for the method of administration. The pharmaceutical composition of the present disclosure includes pharmaceutically acceptable components that are compatible with a conjugate described herein (e.g., a conjugate of any one of formulas (I)-(XXXIII)).

As used herein, the term“pharmaceutically acceptable carrier” refers to an excipient or diluent in a pharmaceutical composition. For example, a pharmaceutically acceptable carrier may be a vehicle capable of suspending or dissolving the active conjugate (e.g., a conjugate of any one of formulas (I)- (XXXIII)). The pharmaceutically acceptable carrier must be compatible with the other ingredients of the formulation and not deleterious to the recipient. In the present disclosure, the pharmaceutically acceptable carrier must provide adequate pharmaceutical stability to a conjugate described herein. The nature of the carrier differs with the mode of administration. For example, for oral administration, a solid carrier is preferred; for intravenous administration, an aqueous solution carrier (e.g., WFI, and/or a buffered solution) is generally used.

The term“pharmaceutically acceptable salt,” as used herein, represents salts of the conjugates described herein (e.g., conjugates of any one of formulas (I)-(XXXIII)) that are, within the scope of sound medical judgment, suitable for use in methods described herein without undue toxicity, irritation, and/or allergic response. Pharmaceutically acceptable salts are well known in the art. For example, pharmaceutically acceptable salts are described in: Pharmaceutical Salts: Properties, Selection, and Use (Eds. P.H. Stahl and C.G. Wermuth), Wiley-VCH, 2008. The salts can be prepared in situ during the final isolation and purification of the conjugates described herein or separately by reacting the free base group with a suitable organic acid.

The term“about,” as used herein, indicates a deviation of ±5%. For example, about 10% refers to from 9.5% to 10.5%.

Definitions of abbreviations used in the disclosure are provided in T

Table A

Other features and advantages of the conjugates described herein will be apparent from the following Detailed Description and the claims. Description of the Drawings

FIG.1 shows non-reducing and reducing SDS-PAGE and a schematic illustration of an Fc domain formed from Fc domain monomers having the sequene of SEQ ID NO: 1.

FIG.2 shows non-reducing and reducing SDS-PAGE and a schematic illustration of an Fc domain formed from Fc domain monomers having the sequene of SEQ ID NO: 3.

FIG.3 shows non-reducing and reducing SDS-PAGE and a schematic illustration of an Fc domain formed from Fc domain monomers having the sequene of SEQ ID NO: 5.

FIG.4 shows non-reducing and reducing SDS-PAGE and a schematic illustration of an Fc domain formed from Fc domain monomers having the sequene of SEQ ID NO: 7.

FIG.5 shows non-reducing and reducing SDS-PAGE and a schematic illustration of an Fc domain formed from Fc domain monomers having the sequene of SEQ ID NO: 9.

FIG.6 shows non-reducing and reducing SDS-PAGE and a schematic illustration of an Fc domain formed from Fc domain monomers having the sequene of SEQ ID NO: 11.

FIG.7 shows non-reducing and reducing SDS-PAGE and a schematic illustration of an Fc domain formed from Fc domain monomers having the sequene of SEQ ID NO: 13 FIG.8 shows a non-reducing SDS-PAGE of Conjugate 3.

FIG.9 shows a non-reducing SDS-PAGE of Conjugate 4.

FIG.10 shows a non-reducing SDS-PAGE of Conjugate 5.

FIG.11 shows a non-reducing SDS-PAGE of Conjugate 6.

F IG. 12 shows a non-reducing SDS-PAGE of Conjugate 7.

FIG.13 shows a non-reducing SDS-PAGE of Conjugate 8.

FIG.14 shows a non-reducing SDS-PAGE of Conjugate 9.

FIG.15 shows a non-reducing SDS-PAGE of Conjugate 10.

FIG.16 shows a non-reducing SDS-PAGE of Conjugate 11.

FIG.17 shows a non-reducing SDS-PAGE of Conjugate 12.

FIG.18 shows a non-reducing SDS-PAGE of Conjugate 13.

FIG.19 shows a non-reducing SDS-PAGE of Conjugate 14.

FIG.20 shows a non-reducing SDS-PAGE of Conjugate 15.

FIG.21 shows a non-reducing SDS-PAGE of Conjugate 16.

FIG.22 shows a non-reducing SDS-PAGE of Conjugate 17.

FIG.23 shows a non-reducing SDS-PAGE of Conjugate 18.

FIG.24 shows a non-reducing SDS-PAGE of Conjugate 19.

FIG.25 shows a non-reducing SDS-PAGE of Conjugate 20.

FIG.26 shows a non-reducing SDS-PAGE of Conjugate 21.

FIG.27 shows a non-reducing SDS-PAGE of Conjugate 22.

FIG.28 shows a non-reducing SDS-PAGE of Conjugate 23.

FIG.29 shows a non-reducing SDS-PAGE of Conjugate 24.

FIG.30 shows a non-reducing SDS-PAGE of Conjugate 25.

FIG.31 shows a non-reducing SDS-PAGE of Conjugate 26.

F IGS. 32A-32C are plots showing the binding of conjugate 3 (FIG. 32A), conjugate 5 (FIG. 32B), and conjugate 7 (FIG.32C) to bacteria as measured by flow cytometry.

FIGS.32D-32F are images showing the binding of Conjugate 13 (FIG.32D), Conjugate 7 (FIG. 32E), and a negative control (FIG.32F) to E. coli, as determined by antibody binding.

FIGS.33A-33F are graphs showing that drug conjugation to an Fc domain does not interfere with FcRn interaction. Binding to mouse FcRn is shown for conjugate 15 (FIG.33A), conjugate 4 (FIG.33B), conjugate 5 (FIG.33C), and conjugate 7 (FIG.33D). Binding to human FcRn is shown for conjugate 5 (FIG.33E) and conjugate 7 (FIG.33F) .

FIG.34A is a graph depicting the mean plasma concentration-time profile following administration of 10 mg/kg of conjugate 23 by intravenous (IV) or intraperitoneal (IP) administration in mice.

FIG.34B is a table which corresponds to the graph of FIG.34A, and which shows the individual and mean plasma concentrations and pharmacokinetics following administration of 10 mg/kg of conjugate 23 by intravenous (IV) or intraperitoneal (IP) administration in mice. FIG.35 is a graph that shows conjugate-mediated bacterial comple

(CDC).

FIG.36 is a survival curve that shows survival data for treated and untreated mice in a lethal model of septicemia.

F IG. 37 is a survival curve that shows expanded dose ranging study in the E. coli

peritonitis/septicemia model of infection.

FIGS.38A-38C are a series of graphs showing that ADC conjugates reduce CFU burden in pneumonia model against A. baumannii AB5075. Efficacy of ADC conjugates was determined upon intratracheal infection of A. baumannii AB5075 (3 x 10 7 CFU) at 24 h time point. CFU burden was reduced by conjugate 6 (FIG.38A), conjugate 20 (FIG.38B) and conjugate 21 (FIG.38C). Combination treatment with azithromycin (AZM) further reduced CFU burden beyond activity of AZM or conjugates alone. Conjugates (50 mpk) were administered intraperitoneally at t=-12 h and t=+1 h. AZM (25 mpk) was administered subcutaneously once at t=+1 h.

FIGS.39A and 39B are a set of survival curves showing that ADC conjugates protect against lethal challenge in pneumonia model against A. baumannii AB5075. Efficacy of ADC conjugates was determined upon intratracheal infection with A. baumannii AB5075 (5 x 10 7 CFU) over 5 days. FIG.39A shows that conjugate 22 and colistin showed 100% protection, whereas mice treated with control IgG1 resulted in 20% survival. FIG 39B shows that conjugate 23 resulted in 100% protection and IgG1 control was 0% survival. Colistin resulted in 20% protection at day 5. Conjugates or IgG1 (50 mpk) were administered intraperitoneally at (A) t=-12 h, t=+1 h, t=+24 h and t=+48 h or (B) t=-12 h and t=+1h. Colistin (10 mpk) and PBS were administered intraperitoneally at (A) t=+1 h, t=+7 h, t=+24 h and t=+48 h or (B) t=+1 h, t=+5 h, t=+24 h, t=+36 h, t=+48 h and t=+60 h. Survival was monitored twice daily for 5 days.

FIGS.40A-40C are a series of graphs showing that ADC conjugates bind purified LPS.

Conjugate 23 binds to purified LPS from E. coli O111:B4 in dose-dependency from 0.05– 5 µM (FIG. 40A). Correlation of DAR-dependent LPS displacement ranging from DAR 1.5 to 4.5 at 1 µM and 5 µM (FIG.40B and 40C, repsectively). The percent LPS displacement was normalized to vehicle control.

FIGS.41A-41C are a series of graphs showing that ADC conjugates inhibit LPS-induced nitrite oxide production by RAW macrophages. Conjugate 23 inhibit NO production by RAW macrophages in response to LPS (10 µg/mL) in dose-dependency from 0.05– 5 µM at 24 h time point (FIG.41A).

Correlation of DAR-dependent inhibition ranging from DAR 1.5 to 4.5 at 1 µM and 5 µM (FIGS.41B and 41C, respectively). The percent inhibition was normalized to vehicle control.

FIG.42 shows a non-reducing SDS-PAGE of Conjugate 27.

FIG.43 shows a non-reducing SDS-PAGE of Conjugate 28.

FIG.44 shows a non-reducing SDS-PAGE of Conjugate 29.

FIG.45 shows a non-reducing SDS-PAGE of Conjugate 30.

FIG.46 shows a non-reducing SDS-PAGE of Conjugate 31.

FIG 47 h d i SDS PAGE f C j t 32 FIG.48 shows a non-reducing SDS-PAGE of Conjugate 33. FIG.49 shows a non-reducing SDS-PAGE of Conjugate 34. FIG.50 shows a non-reducing SDS-PAGE of Conjugate 35. FIG.51 shows a non-reducing SDS-PAGE of Conjugate 36. FIG. 52 shows a non-reducing SDS-PAGE of Conjugate 37. FIG.53 shows a non-reducing SDS-PAGE of Conjugate 38. FIG.54 shows a non-reducing SDS-PAGE of Conjugate 39. FIG.55 shows a non-reducing SDS-PAGE of Conjugate 40. FIG.56 shows a non-reducing SDS-PAGE of Conjugate 41. FIG.57 shows a non-reducing SDS-PAGE of Conjugate 42. FIG.58 shows a non-reducing SDS-PAGE of Conjugate 43. FIG.59 shows a non-reducing SDS-PAGE of Conjugate 44. FIG.60 shows a non-reducing SDS-PAGE of Conjugate 45. FIG.61 shows a non-reducing SDS-PAGE of Conjugate 46. FIG.62 shows a non-reducing SDS-PAGE of Conjugate 47. FIG.63 shows a non-reducing SDS-PAGE of Conjugate 48. FIG.64 shows a non-reducing SDS-PAGE of Conjugate 49. FIG.65 shows a non-reducing SDS-PAGE of Conjugate 50. FIG.66 shows a non-reducing SDS-PAGE of Conjugate 51. FIG.67 shows a non-reducing SDS-PAGE of Conjugate 52. FIG.68 shows a non-reducing SDS-PAGE of Conjugate 53. FIG.69 shows a non-reducing SDS-PAGE of Conjugate 54. FIG.70 shows a non-reducing SDS-PAGE of Conjugate 55. FIG.71 shows a non-reducing SDS-PAGE of Conjugate 56. FIG. 72 shows a non-reducing SDS-PAGE of Conjugate 57. FIG.73 shows a non-reducing SDS-PAGE of Conjugate 58. FIG.74 shows a non-reducing SDS-PAGE of Conjugate 59. FIG.75 shows a non-reducing SDS-PAGE of Conjugate 60. FIG.76 shows a non-reducing SDS-PAGE of Conjugate 61. FIG.77 shows a non-reducing SDS-PAGE of Conjugate 62. FIG.78 shows a non-reducing SDS-PAGE of Conjugate 63. FIG.79 shows a non-reducing SDS-PAGE of Conjugate 64. FIG.80 shows a non-reducing SDS-PAGE of Conjugate 65. FIG.81 shows a non-reducing SDS-PAGE of Conjugate 66. FIG.82 shows a non-reducing SDS-PAGE of Conjugate 67. FIG.83 shows a non-reducing SDS-PAGE of Conjugate 68. FIG.84 shows a non-reducing SDS-PAGE of Conjugate 69. FIG.85 shows a non-reducing SDS-PAGE of Conjugate 70. G S S G f C FIG.87 shows a non-reducing SDS-PAGE of Conjugate 72.

FIG.88 shows a non-reducing SDS-PAGE of Conjugate 73.

FIG.89 shows a non-reducing SDS-PAGE of Conjugate 74.

FIG.90 shows a non-reducing SDS-PAGE of Conjugate 75.

F IG. 91 shows a non-reducing SDS-PAGE of Conjugate 76.

FIG.92 shows a non-reducing SDS-PAGE of Conjugate 77.

FIG.93 shows a non-reducing SDS-PAGE of Conjugate 78.

FIG.94 shows a non-reducing SDS-PAGE of Conjugate 79.

FIG.95 is a survival curve showing the efficacy of ADC conjugate 23 in mouse pneumonia model vs. AB5075. Survival of mice (n=5 per group) was monitored for 5 days. ADC conjugate 23 showed a dose-dependent protection: 0% protection at 1 mpk, 20% protection at 10 mpk and 80% protection at 25 and 50 mpk. Colistin used as positive control resulting in 80% protection. In comparison, control IgG1 and PBS were not protective and mice succumbed to infection between day 1.5– 2.

FIG.96 is a survival curve showing the efficacy of ADC conjugate 34 in mouse pneumonia model vs. PAO1. Survival of mice (n=5 per group) was monitored for 5 days. A single dose of ADC conjugate 34 was protective (80% survival) against lethal challenge with P. aeruginosa PAO1. Colistin used as positive control resulting in complete protection. In comparison, control IgG1 and PBS were not protective and mice succumbed to infection between day 2– 2.5.

FIG.97 is a survival curve showing the efficacy of ADC conjugate 34 in mouse pneumonia model vs. AB5075. Survival of mice (n=4 per group) was monitored for 5 days. A single dose of ADC conjugate 34 was protective (75% survival) against lethal challenge with A. baumannii AB5075. Control IgG1 was not protective and mice succumbed to infection on day 2.

FIG.98 is a survival curve showing the efficacy of ADC conjugate 23 in mouse pneumonia model vs. PAO1. Survival of mice (n=5 per group) was monitored for 5 days. A single dose of ADC conjugate 23 was completely protective against challenge with P. aeruginosa PAO1. Positive control, colistin, was was also protective (100%). Controls IgG1 (60% survival) and PBS (20% survival) were only partially protective.

FIG.99 is a survival curve showing the efficacy of ADC conjugate 23 in mouse pneumonia model vs. PAO1. Survival of mice (n=5 per group) was monitored for 5 days. A single dose of ADC conjugates 23, 34, 35, 38 or 44 was completely protective against challenge with P. aeruginosa PAO1. Positive control, colistin, was was also protective (100%). Controls IgG1 (60% survival) was only partially protective and mice in PBS group succumbed to infection on day 2.

FIG.100 is a survival curve showing the protective effect of Conjugate 23 against endotoxemia. Mice dosed with the vehicle or hIgG1 reached 100% mortality within 48 hours of LPS challenge. Mice receiving multiple doses of colistin were partially protected with ~30% of animals reaching death after 5 days. In contrast, all mice treated with conjugate 23 were rescued with 0% mortality.

FIG.101 is a graph showing the in vitro killing of E. coli ATCC 25922 following treatment with either conjugate 23 alone, or conjugate 23 and purified human neutrophils. FIG.102 is a FACS plot showing the neutrophil phagocytosis of FIT

BioParticles opsonized with conjugate 23, conjugate 41, or media alone.

FIG.103 shows a graph of the DAR values of the azido-hIgG1(Myc)Fc that were measured by MALDI-TOF at varying equivalents of azido-PEG4-NHS ester. Detailed Description

The disclosure features conjugates, compositions, and methods for the treatment of bacterial infections (e.g., Gram-negative bacterial infections). The conjugates disclosed herein include monomers or dimers of cyclic heptapeptides (e.g., two polymyxin cores) conjugated to Fc domains. The dimers of cyclic heptapeptides are linked to each other through a linker and/or one or two peptides (e.g., a peptide including a 1-5 amino acid residue(s)). The monomers or dimers of cyclic heptapeptides in the conjugates described herein bind to lipopolysaccharides (LPS) in the cell membrane of Gram-negative bacteria to disrupt and permeabilize the cell membrane, leading to cell death and/or sensitization of the Gram-negtaive bacteria to other antibiotics. The Fc domains in the conjugates bind to FcγRs (e.g., FcRn, FcγRI, FcγRIIa, FcγRIIc, FcγRIIIa, and FcγRIIIb) on immune cells, e.g., neutrophils, to activate phagocytosis and effector functions, such as antibody-dependent cell-mediated cytotoxicity (ADCC), thus leading to the engulfment and destruction of bacterial cells by immune cells and further enhancing the antibacterial activity of the conjugates. I. Bacterial Infections

Pathogenic bacteria cause bacterial infections and diseases such as tuberculosis, pneumonia, and foodborne illnesses. Bacteria may be categorized into two major types: Gram-positive bacteria and Gram-negative bacteria. Gram-positive bacteria possess a thick cell wall containing multiple layers of peptidoglycan and teichoic acids, while Gram-negative bacteria have a relatively thin cell wall containing fewer layers of peptidoglycan that are surrounded by a second lipid membrane containing

lipopolysaccharides (LPS) and lipoproteins. LPS, also called endotoxins, are composed of

polysaccharides and lipid A. These differences in bacterial cell wall structure can produce differences in antibiotic susceptibility. Examples of Gram-positive bacteria include, but are not limited to, bacteria in the genus Streptococcus (e.g., Streptococcus pyogenes), bacteria in the genus Staphylococcus (e.g., Staphylococcus cohnii), bacteria in the genus Corynebacterium (e.g., Corynebacterium auris), bacteria in the genus Listeria (e.g., Listeria grayi), bacteria in the genus Bacillus (e.g., Bacillus aerius), and bacteria in the genus Clostridium (e.g., Clostridium acetium). Examples of Gram-negative bacteria include, but are not limited to, bacteria in the genus Escherichia (e.g., Escherichia coli), bacteria in the genus Klebsiella (e.g., Klebsiella granulomatis, Klebsiella oxytoca, Klebsiella pneumoniae, Klebsiella terrigena, and Klebsiella variicola), bacteria in the genus Acinetobacter (e.g., Acinetobacter baumannii,

Acinetobacter calcoaceticus, Acinetobacter kookii, and Acinetobacter junii), bacteria in the genus Pseudomonas (e.g., Pseudomonas aeruginosa), bacteria in the genus Neisseria (e.g., Neisseria gonorrhoeae), bacteria in the genus Yersinia (e.g., Yersinia pestis), bacteria in the genus Vibrio (e.g., Vibrio cholerae), bacteria in the genus Campylobacter (e.g., Campylobacte

genus Salmonella (e.g., Salmonella enterica).

Bacteria may evolve to become more or fully resistant to antibiotics. Resistance may arise through natural resistance in certain types of bacteria, spontaneous random genetic mutations, and/or by inter- or intra-species horizontal transfer of resistance genes. Resistant bacteria are increasingly difficult to treat, requiring alternative medications or higher doses, which may be more costly or more toxic.

Bacteria resistant to multiple antibiotics are referred to as multidrug resistant (MDR) bacteria. For example, the mcr-1 gene encodes a phosphoethanolamine transferase (MCR-1) which confers resistance to colistin, a natural polymyxin, through modification of LPS. This is the first known horizontally- transferable resistance determinant for the polymyxin class of antibiotics. The mcr-1 gene has also been found in bacterial strains which already possess resistance to other classes of antibiotics, such as in carbapenem-resistant Enterobacteriaceae (CRE). An mcr-1 resistance plasmid refers to a bacterial plasmid that carries mcr-1 alone or in combination with other antibiotic resistance genes. A mcr-1 resistance plasmid refers to a bacterial plasmid that carries one or more antibiotic resistance genes. Examples of mcr-1 resistance plasmids include, but are not limited to, pHNSHP45, pMR0516mcr, pESTMCR, pAF48, pAF23, pmcr1-IncX4, pmcr1-IncI2, pA31-12, pVT553, pICBEC72Hmcr, pE15004, pE15015, and pE15017.

In another example, the mcr-2 gene also confers resistance to colistin. The mcr-2 gene was identified in porcine and bovine colistin-resistance E.coli that did not contain mcr-1 (Xavier et al., Euro Surveill 21(27), 2016). The mcr-2 gene is a 1,617 bp phspoethanolamine transferase harbored on an IncX4 plasmid. The mcr-2 gene has 76.7% nucleotide identity to mcr-1. Analysis of mcr-2 harboring plasmids from E.coli isolates shows that the mobile element harboring mcr-2 is an IS element of the IS1595 superfamily, which are distinguished by the presence of an ISXO2-like transposase domain (Xavier et al., supra). The MCR-2 protein was predicted to have two domains, with domain 1 (1-229 residues) as a transporter and domain 2 (230-538 residues) as a transferase domain. An mcr-2 resistance plasmid refers to a bacterial plasmid that carries mcr-2 alone or in combination with other antibiotic resistance genes. A mcr-2 resistance plasmid refers to a bacterial plasmid that carries one or more antibiotic resistance genes. Mcr-2 resistance plasmids include, but are not limited to, pKP37-BE and pmcr2-IncX4.

Furthermore, resistant strain E. coli BAA-2469 possesses the New Delhi metallo-β-lactamase (NDM-1) enzyme, which makes bacteria resistant to a broad range of β-lactam antibiotics. Additionally, E. coli BAA-2469 is also known to be resistatnt to penicillins (e.g., ticarcillin, ticarcillin/clavulanic acid, piperacillin, ampicillin, and ampicillin/sulbactam), cephalosporins (e.g., cefalotin, cefuroxime, cefuroxime, cefotetan, cefpodoxime, cefotaxime, ceftizoxime, cefazolin, cefoxitin, ceftazidime, ceftriaxone, and cefepime), carbapenems (e.g., doripenem, meropenem, ertapenem, imipenem), quinolones (e.g., nalidixic acid, moxifloxacin, norfloxacin, ciprofloxacin, and levofloxacin), aminoglycosides (e.g., amikacin, gentamicin, and tobramycin), and other antibiotics (e.g., tetracycline, tigecycline, nitrofurantoin, aztreonam, trimethoprim/sulfamethoxazole). In some embodiments, a resistant strain of bacteria possesses the

and/or a chromosomal mutation conferring polymyxin resistance. In some embodiments, a resistant strain of bacteria is a resistant strain of E. coli (e.g., E. coli BAA-2469). II. Conjugates of the Disclosure

Provided herein are synthetic conjugates useful in the treatment of bacterial infections (e.g., Gram-negative bacterial infections). The conjugates disclosed herein include an Fc domain conjugated to one or more monomers of cyclic heptapeptides or one or more dimers of two cyclic heptapeptides. The dimers of two cyclic heptapeptides include a first cyclic heptapeptide (e.g., a first polymyxin core) and a second cyclic heptapeptide (e.g., a second polymyxin core). The first and second cyclic heptapeptides are linked to each other by way of a linker and/or one or two peptides (e.g., each peptide including a 1-5 amino acid residue(s)).

Without being bound by theory, in some aspects, conjugates described herein bind to the cell membrane of Gram-negative bacteria (e.g., bind to LPS in the cell membrane of Gram-negative bacteria) through the interactions between the cyclic heptapeptides in the conjugates and the cell membrane of Gram-negative bacteria. The binding of the conjugates to the cell membrane of Gram-negative bacteria disrupt and permeabilize the cell membrane, leading to cell death and/or sensitization of the Gram- negative bacteria to other antibiotics. In some embodiments, the initial association of the conjugates with the bacterial cell membrane occurs through electrostatic interactions between the cyclic heptapepdies in the conjugates and the anionic LPS in the outer membrane of Gram-negative bacteria, disrupting the arrangement of the cell membrane. Specifically, conjugates described herein may bind to lipid A in the LPS. More specifically, the cyclic heptapeptides in the conjugates described herein may bind to one or both phosphate groups in lipid A. In some embodiments, antibiotic-resistant bacteria (e.g., antibiotic- resistant, Gram-negative bacteria) has one phosphate group in lipid A. In some embodiments, conjugates described herein may bind to multiple Gram-negative bacterial cells at the same time. The binding of the conjugates described herein to the LPS may also displace Mg 2+ and Ca 2+ cations that bridge adjacent LPS molecules, causing, e.g., membrane permeabilization, leakage of cellular molecules, inhibition of cellular respiration, and/or cell death. Furthermore, in addition to disrupting the cell membrane of Gram-negative bacteria, the Fc domain in the conjugates described herein binds to the FcγRs (e.g., FcRn, FcγRI, FcγRIIa, FcγRIIc, FcγRIIIa, and FcγRIIIb) on immune cells and serves as a gradient against which immune cells chemotax to the site of bacterial infection and/or growth. The binding of the Fc domain in the conjugates described herein to the FcγRs on immune cells activates phagocytosis and effector functions, such as antibody-dependent cell-mediated cytotoxicity (ADCC), thus leading to the engulfment and destruction of Gram-negative bacterial cells by immune cells and further enhancing the antibacterial activity of the conjugates.

Conjugates provided herein are described by any one of formulas (I)-(XXXIII). In some embodiments, the conjugates described herein include one or more monomers of cyclic heptapeptides conjugated to an Fc domain. In some embodiments, the conjugates described herein include one or f (an Fc domain monomer) dimerizes to form an Fc domain. Conjugates des

synthesized using available chemical synthesis techniques in the art. In some embodiments, available functional groups in the cyclic heptapeptides and the linker, e.g., amines, carboxylic acids, maleimides, bis-sulfones, azides, alkynes, and/or hydroxyl groups, may be used in making the conjugates described herein. For example, the linking nitrogen (described further herein) in a cyclic heptapeptide may form an amide bond with the carbon in a carboxylic acid group in the linker. A peptide including one or more (e.g., 1-3; 1, 2, or 3) amino acid residues (e.g., natural and/or non-natural amino acid residues) may be also be covalently attached to the linking nitrogen of the cyclic heptapeptide through forming an amide bond between the carbon in a carboxylic acid group in the peptide and the linking nitrogen. In cases where a functional group is not available for conjugation, a molecule may be derivatized using conventional chemical synthesis techniques that are well known in the art. In some embodiments, the conjugates described herein contain one or more chiral centers. The conjugates include each of the isolated stereoisomeric forms as well as mixtures of stereoisomers in varying degrees of chiral purity, including racemic mixtures. It also encompasses the various diastereomers, enantiomers, and tautomers that can be formed. Cyclic heptapeptide or polymyxin core

In some aspects, a cyclic heptapeptide or polymyxin core, as used herein, refers to certain compounds that bind to lipopolysaccharides (LPS) in the cell membrane of Gram-negative bacteria to disrupt and permeabilize the cell membrane, leading to cell death and/or sensitization to other antibiotics. In some aspects, cyclic heptapeptide, as used herein, refers to certain compounds that kill or inhibit the growth of Gram-negative bacteria as determined by measuring the minimum inhibitory concentration (MIC) against at least one Gram-negative bacteria as known in the art, wherein the MIC is 128 µg/mL or less.

C yclic heptapeptides are composed of, at least, amino acid residues, each of which may, independently, may have a D- or L- configuration, assembled as a cyclic heptapeptide ring. A cyclic heptapeptide includes seven natural or non-natural amino acid residues attached to each other in a closed ring. The ring contains six bonds formed by linking the carbon in the α-carboxyl group of one amino acid residue to the nitrogen in the α-amino group of the adjacent amino acid residue and one bond formed by linking the carbon in the α-carboxyl group of one amino acid residue to the nitrogen in the γ- amino group in the side chain of the adjacent amino acid residue. For the amino acid residue whose nitrogen in the γ-amino group in the side chain participates in forming the ring, the nitrogen in the α-amino group of this amino acid residue does not participate directly in forming the ring and serves as the linking nitrogen (thus, referred to as the“linking nitrogen” herein) that links one cyclic heptapeptide or polymyxin core to an Fc domain by way of a linker and/or one or two peptides (e.g., a peptide including a 1-5 amino acid residue(s)), or in the case of a conjugate including an Fc domain covalently linked to one or more dimers of cyclic heptapeptides or polymyxin cores, the linking nitrogen serves to link one cyclic heptapeptide or polymyxin core to another cyclic heptapeptide or polymyxin core. Conjugates described

( ) f domain and (2) one or more monomers of cyclic heptapeptides conjugated

of cyclic heptapeptides (e.g., two polymyxin cores) are linked to each other at their linking nitrogens through a linker and/or one or two peptides (e.g., a peptide including a 1-5 amino acid residue(s)).

In some embodiments, a peptide including one or more (e.g., 1-5; 1, 2, 3, 4, or 5) amino acid residues (e.g., natural and/or non-natural amino acid residues) may be covalently attached to the linking nitrogen of the cyclic heptapeptide or the polymyxin core.

Cyclic heptapeptides or polymyxin cores may be derived from polymyxins (e.g., naturally existing polymyxins and non-natural polymyxins) and/or octapeptins (e.g., naturally existing octapeptins and non- natural octapeptins). In some embodiments, cyclic heptapeptides may be compounds described in Gallardo-Godoy et al., J. Med. Chem.59:1068, 2016 (e.g., compounds 11-41 in Table 1 of Gallardo- Godoy et al.), which is incorporated herein by reference in its entirety. Examples of naturally existing polymyxins and their structures are shown in Table 1A. Examples of non-natural polymyxins and their structures are shown in Table 1B. Table 1A. Natural polymyxins and their structures

Table 1B. Non-natural polymyxins and their structures (Dab: diaminobutyric acid; Dap: diaminopropionic acid; Orn: ornithine;

Abu: 2-aminobutyric acid; Nle: norleucine)

Conjguates of dimers of cyclic heptapeptides linked to an Fc domain

The conjugates described herein an Fc domain covalently linked to one or more dimers of cyclic heptapeptides. The dimers of two cyclic heptapeptides include a first cyclic heptapeptide (e.g., a first polymyxin core) and a second cyclic heptapeptide (e.g., a second polymyxin core). The first and second cyclic heptapeptides are linked to each other by way of a linker and/or one or two peptides (e.g., each peptide including a 1-5 amino acid residue(s)). In some embodiments of the dimers of cyclic

heptapeptides, the first and second cyclic heptapeptides are the same. In some embodiments, the first and second cyclic heptapeptides are different. The disclosure provides a conjugate, or a

pharmaceutically acceptable salt thereof, described by formula (I):

In some embodiments, the disclosure provides a conjugate, or a pharmaceutically acceptable salt thereof, described by the formulae below:

;

ĨDV6)

(D-VI-7)

(D-VI-10)

 

;

(D-XVII)

ĨDXX)

;

;

(D-XXVI)

.

(D-XXVIII-2)

In some embodiments, when a is 1 in the conjugate of formula (II), (i) each of R 2 , R 3 , and R 4 is, independently, a lipophilic moiety, a positively charged moiety, a polar moiety, H, optionally substituted alkamino, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted

cycloalkenylene, optionally substituted cycloalkynylene, optionally substituted aryl, optionally substituted heteroalkyl, optionally substituted heterocycloalkyl, optionally substituted heterocycloalkenylene, optionally substituted heterocycloalkynylene, optionally substituted heteroaryl, optionally substituted alkaryl, or optionally substituted heteroalkaryl; or (ii) R 2 , R 3 , and C 1 together form a ring (e.g., an optionally substituted 5-8 membered ring) comprising optionally substituted cycloalkyl, optionally substituted heterocycloalkyl comprising 1 or 2 heteroatoms independently selected from N, O, and S, and R 4 is a lipophilic moiety, a positively charged moiety, a polar moiety, H, optionally substituted alkamino, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted cycloalkenylene, optionally substituted cycloalkynylene, optionally substituted aryl, optionally substituted heteroalkyl, optionally substituted heterocycloalkyl, optionally substituted heterocycloalkenylene, optionally substituted heterocycloalkynylene, optionally substituted heteroaryl, optionally substituted alkaryl, or optionally substituted heteroalkaryl; or (iii) R 3 , R 4 , N 1 , and C 1 together form a ring (e.g., an optionally substituted 5-8 membered ring) comprising optionally substituted heterocycloalkyl comprising an N heteroatom and additional 0-2 heteroatoms independently selected from N, O, and S, or optionally substituted heteroaryl comprising an N heteroatom and additional 0-2 heteroatoms independently selected from N, O, and S, and R 2 is a lipophilic moiety, a positively charged moiety, a polar moiety, H, optionally substituted alkamino, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted

cycloalkenylene, optionally substituted cycloalkynylene, optionally substituted aryl, optionally substituted heteroalkyl, optionally substituted heterocycloalkyl, optionally substituted heterocycloalkenylene, optionally substituted heterocycloalkynylene, optionally substituted heteroaryl, optionally substituted alkaryl, or optionally substituted heteroalkaryl.

In some embodiments, when b is 1 in the conjugate of formula (II), (i) each of R 5 , R 6 , and R 7 is, independently, a lipophilic moiety, a positively charged moiety, a polar moiety, H, optionally substituted alkamino, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted

c ycloalkenylene, optionally substituted cycloalkynylene, optionally substituted aryl, optionally substituted heteroalkyl optionally substituted heterocycloalkyl optionally substituted heterocycloalkenylene optionally substituted heterocycloalkynylene, optionally substituted heteroar

alkaryl, or optionally substituted heteroalkaryl; or (ii) R 5 , R 6 , and C 2 together form a ring (e.g., an optionally substituted 5-8 membered ring) comprising optionally substituted cycloalkyl, optionally substituted heterocycloalkyl comprising 1 or 2 heteroatoms independently selected from N, O, and S, and R7 is a lipophilic moiety, a positively charged moiety, a polar moiety, H, optionally substituted alkamino, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted cycloalkenylene, optionally substituted cycloalkynylene, optionally substituted aryl, optionally substituted heteroalkyl, optionally substituted heterocycloalkyl, optionally substituted heterocycloalkenylene, optionally substituted heterocycloalkynylene, optionally substituted heteroaryl, optionally substituted alkaryl, or optionally substituted heteroalkaryl; or (iii) R 6 , R 7 , N 2 , and C 2 together form a ring (e.g., an optionally substituted 5-8 membered ring) comprising optionally substituted heterocycloalkyl comprising an N heteroatom and additional 0-2 heteroatoms independently selected from N, O, and S, or optionally substituted heteroaryl comprising an N heteroatom and additional 0-2 heteroatoms independently selected from N, O, and S, and R 5 is a lipophilic moiety, a positively charged moiety, a polar moiety, H, optionally substituted alkamino, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted

cycloalkenylene, optionally substituted cycloalkynylene, optionally substituted aryl, optionally substituted heteroalkyl, optionally substituted heterocycloalkyl, optionally substituted heterocycloalkenylene, optionally substituted heterocycloalkynylene, optionally substituted heteroaryl, optionally substituted alkaryl, or optionally substituted heteroalkaryl.

In some embodiments, when c is 1 in the conjugate of formula (II), (i) each of R 8 , R 9 , and R 10 is, independently, a lipophilic moiety, a positively charged moiety, a polar moiety, H, optionally substituted alkamino, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted

cycloalkenylene, optionally substituted cycloalkynylene, optionally substituted aryl, optionally substituted heteroalkyl, optionally substituted heterocycloalkyl, optionally substituted heterocycloalkenylene, optionally substituted heterocycloalkynylene, optionally substituted heteroaryl, optionally substituted alkaryl, or optionally substituted heteroalkaryl; or (ii) R 8 , R 9 , and C 3 together form a ring (e.g., an optionally substituted 5-8 membered ring) comprising optionally substituted cycloalkyl, optionally substituted heterocycloalkyl comprising 1 or 2 heteroatoms independently selected from N, O, and S, and R 10 is a lipophilic moiety, a positively charged moiety, a polar moiety, H, optionally substituted alkamino, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted cycloalkenylene, optionally substituted cycloalkynylene, optionally substituted aryl, optionally substituted heteroalkyl, optionally substituted heterocycloalkyl, optionally substituted heterocycloalkenylene, optionally substituted heterocycloalkynylene, optionally substituted heteroaryl, optionally substituted alkaryl, or optionally substituted heteroalkaryl; or (iii) R 9 , R 10 , N 3 , and C 3 together form a ring (e.g., an optionally substituted 5-8 membered ring) comprising optionally substituted heterocycloalkyl comprising an N heteroatom and additional 0-2 heteroatoms independently selected from N, O, and S, or optionally substituted heteroaryl comprising an N heteroatom and additional 0-2 heteroatoms independently selected from N, O, and S, and R 8 is a lipophilic moiety, a positively charged moiety, a polar moiety, H, optionally substituted cycloalkenylene, optionally substituted cycloalkynylene, optionally substitute

heteroalkyl, optionally substituted heterocycloalkyl, optionally substituted heterocycloalkenylene, optionally substituted heterocycloalkynylene, optionally substituted heteroaryl, optionally substituted alkaryl, or optionally substituted heteroalkaryl.

I n some embodiments, when a’ is 1 in the conjugate of formula (II), (i) each of R’2, R’3, and R’4 is, independently, a lipophilic moiety, a positively charged moiety, a polar moiety, H, optionally substituted alkamino, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted

cycloalkenylene, optionally substituted cycloalkynylene, optionally substituted aryl, optionally substituted heteroalkyl, optionally substituted heterocycloalkyl, optionally substituted heterocycloalkenylene, optionally substituted heterocycloalkynylene, optionally substituted heteroaryl, optionally substituted alkaryl, or optionally substituted heteroalkaryl; or (ii) R’ 2 , R’ 3 , and C’ 1 together form a ring (e.g., an optionally substituted 5-8 membered ring) comprising optionally substituted cycloalkyl, optionally substituted heterocycloalkyl comprising 1 or 2 heteroatoms independently selected from N, O, and S, and R’ 4 is a lipophilic moiety, a positively charged moiety, a polar moiety, H, optionally substituted alkamino, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted cycloalkenylene, optionally substituted cycloalkynylene, optionally substituted aryl, optionally substituted heteroalkyl, optionally substituted heterocycloalkyl, optionally substituted heterocycloalkenylene, optionally substituted heterocycloalkynylene, optionally substituted heteroaryl, optionally substituted alkaryl, or optionally substituted heteroalkaryl; or (iii) R’ 3 , R’ 4 , N’ 1 , and C’ 1 together form a ring (e.g., an optionally substituted 5- 8 membered ring) comprising optionally substituted heterocycloalkyl comprising an N heteroatom and additional 0-2 heteroatoms independently selected from N, O, and S, or optionally substituted heteroaryl comprising an N heteroatom and additional 0-2 heteroatoms independently selected from N, O, and S, and R’ 2 is a lipophilic moiety, a positively charged moiety, a polar moiety, H, optionally substituted alkamino, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted

c ycloalkenylene, optionally substituted cycloalkynylene, optionally substituted aryl, optionally substituted heteroalkyl, optionally substituted heterocycloalkyl, optionally substituted heterocycloalkenylene, optionally substituted heterocycloalkynylene, optionally substituted heteroaryl, optionally substituted alkaryl, or optionally substituted heteroalkaryl.

In some embodiments, when b’ is 1 in the conjugate of formula (II), (i) each of R’ 5 , R’ 6 , and R’ 7 is, independently, a lipophilic moiety, a positively charged moiety, a polar moiety, H, optionally substituted alkamino, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted

cycloalkenylene, optionally substituted cycloalkynylene, optionally substituted aryl, optionally substituted heteroalkyl, optionally substituted heterocycloalkyl, optionally substituted heterocycloalkenylene, optionally substituted heterocycloalkynylene, optionally substituted heteroaryl, optionally substituted alkaryl, or optionally substituted heteroalkaryl; or (ii) R’ 5 , R’ 6 , and C’ 2 together form a ring (e.g., an optionally substituted 5-8 membered ring) comprising optionally substituted cycloalkyl, optionally substituted heterocycloalkyl comprising 1 or 2 heteroatoms independently selected from N, O, and S, and R’ 7 is a lipophilic moiety, a positively charged moiety, a polar moiety, H, optionally substituted alkamino, optionally substituted cycloalkynylene, optionally substituted aryl, optionally

optionally substituted heterocycloalkyl, optionally substituted heterocycloalkenylene, optionally substituted heterocycloalkynylene, optionally substituted heteroaryl, optionally substituted alkaryl, or optionally substituted heteroalkaryl; or (iii) R’ 6 , R’ 7 , N’ 2 , and C’ 2 together form a ring (e.g., an optionally substituted 5- 8 membered ring) comprising optionally substituted heterocycloalkyl comprising an N heteroatom and additional 0-2 heteroatoms independently selected from N, O, and S, or optionally substituted heteroaryl comprising an N heteroatom and additional 0-2 heteroatoms independently selected from N, O, and S, and R’ 5 is a lipophilic moiety, a positively charged moiety, a polar moiety, H, optionally substituted alkamino, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted

cycloalkenylene, optionally substituted cycloalkynylene, optionally substituted aryl, optionally substituted heteroalkyl, optionally substituted heterocycloalkyl, optionally substituted heterocycloalkenylene, optionally substituted heterocycloalkynylene, optionally substituted heteroaryl, optionally substituted alkaryl, or optionally substituted heteroalkaryl.

In some embodiments, when c’ is 1 in the conjugate of formula (II), (i) each of R’ 8 , R’ 9 , and R’ 10 is, independently, a lipophilic moiety, a positively charged moiety, a polar moiety, H, optionally substituted alkamino, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted

cycloalkenylene, optionally substituted cycloalkynylene, optionally substituted aryl, optionally substituted heteroalkyl, optionally substituted heterocycloalkyl, optionally substituted heterocycloalkenylene, optionally substituted heterocycloalkynylene, optionally substituted heteroaryl, optionally substituted alkaryl, or optionally substituted heteroalkaryl; or (ii) R’ 8 , R’ 9 , and C’ 3 together form a ring (e.g., an optionally substituted 5-8 membered ring) comprising optionally substituted cycloalkyl, optionally substituted heterocycloalkyl comprising 1 or 2 heteroatoms independently selected from N, O, and S, and R’ 10 is a lipophilic moiety, a positively charged moiety, a polar moiety, H, optionally substituted alkamino, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted cycloalkenylene, optionally substituted cycloalkynylene, optionally substituted aryl, optionally substituted heteroalkyl, optionally substituted heterocycloalkyl, optionally substituted heterocycloalkenylene, optionally substituted heterocycloalkynylene, optionally substituted heteroaryl, optionally substituted alkaryl, or optionally substituted heteroalkaryl; or (iii) R’ 9 , R’ 10 , N’ 3 , and C’ 3 together form a ring (e.g., an optionally substituted 5-8 membered ring) comprising optionally substituted heterocycloalkyl comprising an N heteroatom and additional 0-2 heteroatoms independently selected from N, O, and S, or optionally substituted heteroaryl comprising an N heteroatom and additional 0-2 heteroatoms independently selected from N, O, and S, and R’ 8 is a lipophilic moiety, a positively charged moiety, a polar moiety, H, optionally substituted alkamino, optionally substituted alkyl, optionally substituted cycloalkyl, optionally substituted

cycloalkenylene, optionally substituted cycloalkynylene, optionally substituted aryl, optionally substituted heteroalkyl, optionally substituted heterocycloalkyl, optionally substituted heterocycloalkenylene, optionally substituted heterocycloalkynylene, optionally substituted heteroaryl, optionally substituted alkaryl, or optionally substituted heteroalkaryl.

In the conjugates described herein, the squiggly line connected to E indicates that one or more ( ) f may be attached to an Fc domain monomer or an Fc domain. In some emb

more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) dimers of cyclic heptapeptides may be attached to an Fc domain monomer. In some embodiments, when n is 2, one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20) dimers of cyclic heptapeptides may be attached to an Fc domain. The squiggly line in the conjugates described herein is not to be construed as a single bond between one or more dimers of cyclic heptapeptides and an atom in the Fc domain monomer. In some embodiments, when T is 1, one dimer of cyclic heptapeptides may be attached to an atom in the Fc domain monomer or Fc domain. In some embodiments, when T is 2, two dimers of cyclic heptapeptides may be attached to an atom in the Fc domain monomer or Fc domain.

As described further herein, a linker in a conjugate described herein (e.g., L’, L, or L 1 ) may be a branched structure. As described further herein, a linker in a conjugate described herein (e.g., L’, L, or L 1 ) may be a multivalent structure, e.g., a divalent or trivalent structure having two or thee arms, respectively. In some embodiments when the linker has three arms, two of the arms may be attached to the first and second cyclic heptapeptides and the third arm may be attached to the Fc domain monomer or Fc domain.

In conjugates having an Fc domain covalently linked to one or more dimers of cyclic

heptapeptides, as represented by the formulae above, when n is 2, two Fc domain monomers (each Fc domain monomer is represented by E) dimerize to form an Fc domain. Conjguates of monomers of cyclic heptapeptides linked to an Fc domain

In some embodiments, the conjugats described herein include an Fc domain covalentnly linked to one or more monomers of cyclic heptapeptides. Conjugates of an Fc domain and one or more monomers of cyclic heptapeptides may be formed by linking the Fc domain to each of the monomers of cyclic heptapeptides through a linker and/or one or more amino acids (e.g., a peptide including a 1-5 amino acid residue(s)).

In the conjugates having an Fc domain covalently linked to one or more monomers of cyclic heptapeptides described herein, the squiggly line connected to E indicates that one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20) monomers of cyclic heptapeptides may be attached to an Fc domain monomer or an Fc domain. In some embodiments, when n is 1, one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10) monomers of cyclic heptapeptides may be attached to an Fc domain monomer. In some embodiments, when n is 2, one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20) monomers of cyclic heptapeptides may be attached to an Fc domain. The squiggly line in the conjugates described herein is not to be construed as a single bond between one or more monomers of cyclic heptapeptides and an atom in the Fc domain monomer or Fc domain. In some embodiments, when T is 1, one monomer of cyclic heptapeptide may be attached to an atom in the Fc domain monomer or Fc domain. In some embodiments, when T is 2, two monomers of cyclic heptapeptides may be attached to an atom in the Fc domain monomer or Fc domain.

As described further herein, a linker in a conjugate having an Fc domain covalently linked to one f ( ’ ) having two arms. One arm in a divalent linker may be attached to the mono

and the other arm may be attached to the Fc domain monomer or Fc domain.

In some embodiments, a conjugate containing an Fc domain covalently linked to one or more monomers of cyclic heptapeptides provided herein is described by any one of formulae below:

(M-III)

(M-IV-2)

;

(M-VIII-3)

(M-XI)

 

;

;

;

( )

In conjugates having an Fc domain covalently linked to one or more monomers of cyclic heptapeptides, as represented by the formulae above, when n is 2, two Fc domain monomers (each Fc domain monomer is represented by E) dimerize to form an Fc domain. III. Fc domain monomers and Fc domains

An Fc domain monomer includes a hinge domain, a CH2 antibody constant domain, and a CH3 antibody constant domain. The Fc domain monomer can be of immunoglobulin antibody isotype IgG, IgE, IgM, IgA, or IgD. The Fc domain monomer can also be of any immunoglobulin antibody isotype (e.g., IgG1, IgG2a, IgG2b, IgG3, or IgG4). The Fc domain monomer can also be of any species, e.g., human, murine, or mouse. A dimer of Fc domain monomers is an Fc domain that can bind to an Fc receptor, which is a receptor located on the surface of leukocytes.

In some embodiments, an Fc domain monomer in the conjugates described herein may contain one or more amino acid substitutions, additions, and/or deletion relative to an Fc domain monomer having a sequence of any one of SEQ ID NOs: 1-14, 1-29, or 1-31. In some embodiments, an Asn in an Fc domain monomer in the conjugates as described herein may be replaced by Ala in order to prevent N- linked glycosylation (see, e.g., SEQ ID NOs: 11-14, where Asn to Ala substitutuion is labled with *). In some embodiments, an Fc domain monomer in the conjugates described h

additional Cys additions (see, e.g., SEQ ID NOs: 9 and 10, where Cys additions are labled with *).

In some embodiments, an Fc domain monomer in the conjugates as described herein includes an additional moiety, e.g., an albumin-binding peptide, a purification peptide (e.g., a hexa-histidine peptide (HHHHHH (SEQ ID NO: 32)), or a signal sequence (e.g., IL2 signal sequence

MYRMQLLSCIALSLALVTNS (SEQ ID NO: 33)) attached to the N- or C-terminus of the Fc domain monomer. In some embodiments, an Fc domain monomer in the conjugate does not contain any type of antibody variable region, e.g., VH, VL, a complementarity determining region (CDR), or a hypervariable region (HVR).

In some embodiments, an Fc domain monomer in the conjugates as described herein may have a sequence that is at least 95% identical (e.g., 97%, 99%, or 99.5% identical) to the sequence of any one of SEQ ID NOs: 1-14, 1-29, or 1-31 shown below. In some embodiments, an Fc domain monomer in the conjugates as described herein may have a sequence of any one of SEQ ID NOs: 1-14, 1-29, or 1-31 shown below. SEQ ID NO: 1: murine Fc-IgG2a with IL2 signal sequence at the N-terminus (bold)

MYRMQLLSCIALSLALVTNSPRGPTIKPCPPCKCPAPNLLGGPSVFIFPPKIKDVLM ISLSPIVTCVVVDVS EDDPDVQISWFVNNVEVHTAQTQTHREDYNSTLRVVSALPIQHQDWMSGKEFKCKVNNKD LPAPIERTIS KPKGSVRAPQVYVLPPPEEEMTKKQVTLTCMVTDFMPEDIYVEWTNNGKTELNYKNTEPV LDSDGSYFM YSKLRVEKKNWVERNSYSCSVVHEGLHNHHTTKSFSRTPGK SEQ ID NO: 2: mature murine Fc-IgG2a

PRGPTIKPCPPCKCPAPNLLGGPSVFIFPPKIKDVLMISLSPIVTCVVVDVSEDDPD VQISWFVNNVEVHTA QTQTHREDYNSTLRVVSALPIQHQDWMSGKEFKCKVNNKDLPAPIERTISKPKGSVRAPQ VYVLPPPEEE MTKKQVTLTCMVTDFMPEDIYVEWTNNGKTELNYKNTEPVLDSDGSYFMYSKLRV EKKNWVERNSYSC SVVHEGLHNHHTTKSFSRTPGK SEQ ID NO: 3: human Fc-IgG1 with IL2 signal sequence at the N-terminus (bold)

MYRMQLLSCIALSLALVTNSMVRSDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMI SRTPEVTCVVVDV SHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNK ALPAPIEKT ISKAKGQPREPQVYTLPPSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTP PVLDSDGSF FLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK

(SEQ ID NO: 3) SEQ ID NO: 4: mature human Fc-IgG1

MVRSDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEV KFNWYVDGVEVH NAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPRE PQVYTLPPSR EEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKS RWQQGNVFS CS Q S S S G SEQ ID NO: 5: murine Fc-IgG2a with IL2 signal sequence (bold) at the N-terminus and hexa- histidine peptide (italicized) at the C-terminus

MYRMQLLSCIALSLALVTNSPRGPTIKPCPPCKCPAPNLLGGPSVFIFPPKIKDVLM ISLSPIVTCVVVDVS EDDPDVQISWFVNNVEVHTAQTQTHREDYNSTLRVVSALPIQHQDWMSGKEFKCK VNNKDLPAPIERTIS KPKGSVRAPQVYVLPPPEEEMTKKQVTLTCMVTDFMPEDIYVEWTNNGKTELNYKNTEPV LDSDGSYFM YSKLRVEKKNWVERNSYSCSVVHEGLHNHHTTKSFSRTPGKHHHHHH SEQ ID NO: 6: mature murine Fc-IgG2a with hexa-histidine peptide (italicized) at the C-terminus PRGPTIKPCPPCKCPAPNLLGGPSVFIFPPKIKDVLMISLSPIVTCVVVDVSEDDPDVQI SWFVNNVEVHTA QTQTHREDYNSTLRVVSALPIQHQDWMSGKEFKCKVNNKDLPAPIERTISKPKGSVRAPQ VYVLPPPEEE MTKKQVTLTCMVTDFMPEDIYVEWTNNGKTELNYKNTEPVLDSDGSYFMYSKLRVEKKNW VERNSYSC SVVHEGLHNHHTTKSFSRTPGKHHHHHH SEQ ID NO: 7: human Fc-IgG1 with IL2 signal sequence (bold) at the N-terminus and hexa- histidine peptide (italicized) at the C-terminus

MYRMQLLSCIALSLALVTNSMVRSDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMI SRTPEVTCVVVDV SHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNK ALPAPIEKT ISKAKGQPREPQVYTLPPSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTP PVLDSDGSF FLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGKHHHHHH SEQ ID NO: 8: mature human Fc-IgG1 with hexa-histidine peptide (italicized) at the C-terminus MVRSDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFN WYVDGVEVH NAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPRE PQVYTLPPSR EEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKL TVDKSRWQQGNVFS CSVMHEALHNHYTQKSLSLSPGKHHHHHH SEQ ID NO: 9: human Fc-IgG1 with IL2 signal sequence (bold) at the N-terminus, two additional cysteines in the hinge region (*), and hexa-histidine peptide (italicized) at the C-terminus

MYRMQLLSCIALSLALVTNSMVRSDKTHTCPPCPPC*KC*PAPELLGGPSVFLFPPK PKDTLMISRTPEVT CVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYK CKVSNKAL PAPIEKTISKAKGQPREPQVYTLPPSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPE NNYKTTPPVL DSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGKHHHHHH SEQ ID NO: 10: mature human Fc-IgG1 with two additional cysteines in the hinge region (*) and hexa-histidine peptide (italicized) at the C-terminus

MVRSDKTHTCPPCPPC*KC*PAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDV SHEDPEVKFNWYV DGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKA KGQPREPQV YTLPPSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVL

QQGNVFSCSVMHEALHNHYTQKSLSLSPGKHHHHHH SEQ ID NO: 11: murine Fc-IgG2a with IL2 signal sequence (bold) at the N-terminus, Asn to Ala substitution (*), and hexa-histidine peptide (italicized) at the C-terminus

MYRMQLLSCIALSLALVTNSPRGPTIKPCPPCKCPAPNLLGGPSVFIFPPKIKDVLM ISLSPIVTCVVVDVS EDDPDVQISWFVNNVEVHTAQTQTHREDYA*STLRVVSALPIQHQDWMSGKEFKCKVNNK DLPAPIERTI SKPKGSVRAPQVYVLPPPEEEMTKKQVTLTCMVTDFMPEDIYVEWTNNGKTELNYKNTEP VLDSDGSYF MYSKLRVEKKNWVERNSYSCSVVHEGLHNHHTTKSFSRTPGKHHHHHH SEQ ID NO: 12: mature murine Fc-IgG2a with Asn to Ala substitution (*) and hexa-histidine peptide (italicized) at the C-terminus

PRGPTIKPCPPCKCPAPNLLGGPSVFIFPPKIKDVLMISLSPIVTCVVVDVSEDDPD VQISWFVNNVEVHTA QTQTHREDYA*STLRVVSALPIQHQDWMSGKEFKCKVNNKDLPAPIERTISKPKGSVRAP QVYVLPPPEE EMTKKQVTLTCMVTDFMPEDIYVEWTNNGKTELNYKNTEPVLDSDGSYFMYSKLRVEKKN WVERNSYS CSVVHEGLHNHHTTKSFSRTPGKHHHHHH SEQ ID NO: 13: human Fc-IgG1 with IL2 signal sequence (bold) at the N-terminus, Asn to Ala substitution (*), and hexa-histidine peptide (italicized) at the C-terminus

MYRMQLLSCIALSLALVTNSMVRSDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMI SRTPEVTCVVVDV SHEDPEVKFNWYVDGVEVHNAKTKPREEQYA*STYRVVSVLTVLHQDWLNGKEYKCKVSN KALPAPIEK TISKAKGQPREPQVYTLPPSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTT PPVLDSDGS FFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGKHHHHHH S EQ ID NO: 14: mature human Fc-IgG1 with Asn to Ala substitution (*) and hexa-histidine peptide (italicized) at the C-terminus

MVRSDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEV KFNWYVDGVEVH NAKTKPREEQYA*STYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPR EPQVYTLPPS REEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDK SRWQQGNV FSCSVMHEALHNHYTQKSLSLSPGKHHHHHH SEQ ID NO: 15: human IgG1 Fc with Human Serum Albumin Signal Sequence (bold) at the N- terminus

MKWVTFISLLFLFSSAYSISAMVRSDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLM ISR TPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLN GKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSREEMTKNQVSLTCLVKGFYPS DIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNH YTQKSLSLSPGK SEQ ID NO: 16: human IgG1 Fc with Human Serum Albumin Signa

terminus then terminal K removed, G4S linker (italicized) and then c-Myc tag

MKWVTFISLLFLFSSAYSISAMVRSDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLM ISR TPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLN GKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSREEMTKNQVSLTCLVK GFYPS DIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNH YTQKSLSLSPGGGGGSEQKLISEEDL SEQ ID NO: 17: mature human IgG1 Fc with terminal K removed, G4S linker (italicized) and then c-Myc tag

ISAMVRSDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHED PEVKFNWYVDGVE VHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQP REPQVYTLPP SREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVD KSRWQQGN VFSCSVMHEALHNH YTQKSLSLSPGGGGGSEQKLISEEDL SEQ ID NO: 18: human IgG1 Fc with Human Serum Albumin Signal Sequence (bold), with lysine to serine modification (*) to prevent lysine conjugation at this site

MKWVTFISLLFLFSSAYSISAMVRSDKTHTCPPCPAPELLGGPSVFLFPPKPS*DTL MISR

TPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQD WLN GKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSREEMTKNQVSLTCLVKGFYPS DIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNH YTQKSLSLSPGK S EQ ID NO: 19: mature human IgG1 Fc with lysine to serine modification (*) to prevent lysine conjugation at this site

ISAMVRSDKTHTCPPCPAPELLGGPSVFLFPPKPS*DTLMISRTPEVTCVVVDVSHE DPEVKFNWYVDGV EVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQ PREPQVYTLP PSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTV DKSRWQQG NVFSCSVMHEALHNHYTQKSLSLSPGK SEQ ID NO: 20: human IgG1 Fc with Human Serum Albumin Signal Sequence (bold) at the N- terminus

with lysine to serine modification (*) to prevent lysine conjugation at this site, G4S linker (italicized) and C-Myc tag

MKWVTFISLLFLFSSAYSISAMVRSDKTHTCPPCPAPELLGGPSVFLFPPKPS(*)D TLMISR

TPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQD WLN GKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSREEMTKNQVSLTCLVKGFYPS S GQ S GS S S QQG SCS YTQKSLSLSPGGGGGSEQKLISEEDL SEQ ID NO: 21: mature human IgG1 Fc with lysine to serine modification (*) to prevent lysine conjugation at this site, G4S linker (italicized) and C-Myc tag

I SAMVRSDKTHTCPPCPAPELLGGPSVFLFPPKPS(*)DTLMISRTPEVTCVVVDV SHEDPEVKFNWYVDG VEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKG QPREPQVYTL PPSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLT VDKSRWQQ GNVFSCSVMHEALHNHYTQKSLSLSPGGGGGSEQKLISEEDL SEQ ID NO: 22: human IgG1 Fc with Human Serum Albumin Signal Sequence (bold) at the N- terminus, Asn to Ala substitution (*), G4S linker (italicized) and C-myc tag

MKWVTFISLLFLFSSAYSISAMVRSDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLM ISR TPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYA(*)STYRVVSVLTVLHQD WLN

GKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSREEMTKNQVSLTCLVKGF YPS DIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNH YTQKSLSLSPGGGGGSEQKLISEEDL SEQ ID NO: 23: mature human IgG1 Fc with Asn to Ala substitution (*), G4S linker (italicized) and C-myc tag

ISAMVRSDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHED PEVKFNWYVDGVE VHNAKTKPREEQYA(*)STYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAK GQPREPQVYTL PPSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLT VDKSRWQQ GNVFSCSVMHEALHNHYTQKSLSLSPGGGGGSEQKLISEEDL S EQ ID NO: 24: human IgG1 Fc with Human Serum Albumin Signal Sequence (bold) at the N- terminus, with H310A (*) and H435A (*) mutations to impede FcRn binding G4S (italicized) and C-myc tag MKWVTFISLLFLFSSAYSISAMVRSDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISR TPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLA(*)QD WLN

GKEYKCKVSNKALPAPIEKTISKA(*)KGQPREPQVYTLPPSREEMTKNQVSLTCLV KGFYPS

DIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEAL HNA YTQKSLSLSPGGGGGSEQKLISEEDL SEQ ID NO: 25: mature human IgG1 Fc with Human Serum Album

the N-terminus, with H310A (*) and H435A (*) mutations to impede FcRn binding G4S (italicized) and C- myc tag

ISAMVRSDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHED PEVKFNWYVDGVE VHNAKTKPREEQYNSTYRVVSVLTVLA(*)QDWLNGKEYKCKVSNKALPAPIEKT ISKA(*)KGQPREPQVYT LPPSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKL TVDKSRWQQ GNVFSCSVMHEALHNAYTQKSLSLSPGGGGGSEQKLISEEDL SEQ ID NO: 26: human IgG1 Fc with Human Serum Albumin Signal Sequence (bold) at the N- terminus, G4S linker (italicized) and mutated (lysine to phenylalanine, bold) C-myc tag

MKWVTFISLLFLFSSAYSISAMVRSDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLM ISR TPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLN GKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSREEMTKNQVSLTCLVKGFYPS DIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNH YTQKSLSLSPGGGGGSEQFLISEEDL SEQ ID NO: 27: mature human IgG1 Fc with G4S linker (italicized) and mutated (lysine to phenylalanine, bold) C-myc tag

ISAMVRSDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHED PEVKFNWYVDGVE VHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQP REPQVYTLPP SREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVD KSRWQQGN VFSCSVMHEALHNHYTQKSLSLSPGGGGGSEQFLISEEDL SEQ ID NO: 28: human IgG1 Fc with Human Serum Albumin Signal Sequence (bold) at the N- terminus, Asn to Ala substitution (*), G4S linker (italicized) and mutated (lysine to phenylalanine, bold) C- myc tag

MKWVTFISLLFLFSSAYSISAMVRSDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLM ISR TPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYA(*)STYRVVSVLTVLHQD WLN

GKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSREEMTKNQVSLTCLVKGF YPS DIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNH YTQKSLSLSPGGGGGSEQFLISEEDL SEQ ID NO: 29: mature human IgG1 Fc with Asn to Ala substitution

mutated (lysine to phenylalanine, bold) C-myc tag

ISAMVRSDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHED PEVKFNWYVDGVE VHNAKTKPREEQYA(*)STYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAK GQPREPQVYTL PPSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFL YSKLTVDKSRWQQ GNVFSCSVMHEALHNHYTQKSLSLSPGGGGGSEQFLISEEDL SEQ ID NO: 30: human IgG1 Fc with Human Serum Albumin Signal Sequence (bold) at the N- terminus, removal of the residues ISAMVRS from the N-terminus of the mature human IgG1 Fc, Glu to Asp substitution (*), Met to Leu substitution (**), G4S linker (italicized), and mutated (lysine to phenylalanine, bold) C-myc tag

MKWVTFISLLFLFSSAYSDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEV TCVVVDVSHEDPE VKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPI EKTISKAKG QPREPQVYTLPPSRD(*)EL(**)TKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTT PPVLDSDGSFFLY SKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGGGGGSEQFLISEEDL SEQ ID NO: 31: human IgG1 Fc with Human Serum Albumin Signal Sequence (bold) at the N- terminus, removal of the residues ISAMVRS from the N-terminus of the mature human IgG1 Fc, Glu to Asp substitution (*), and Met to Leu substitution (**)

MKWVTFISLLFLFSSAYSDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEV TCVVVDVSHEDPE VKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPI EKTISKAKG QPREPQVYTLPPSRD(*)EL(**)TKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTT PPVLDSDGSFFLY SKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK A s defined herein, an Fc domain includes two Fc domain monomers that are dimerized by the interaction between the CH3 antibody constant domains, as well as one or more disulfide bonds that form between the hinge domains of the two dimerizing Fc domain monomers. An Fc domain forms the minimum structure that binds to an Fc receptor, e.g., Fc-gamma receptors (i.e., Fcγ receptors (FcγR)), Fc-alpha receptors (i.e., Fcα receptors (FcαR)), Fc-epsilon receptors (i.e., Fcε receptors (FcεR)), and/or the neonatal Fc receptor (FcRn). In some embodiments, an Fc domain of the present invention binds to an Fcγ receptor (e.g., FcRn, FcγRI (CD64), FcγRIIa (CD32), FcγRIIb (CD32), FcγRIIIa (CD16a), FcγRIIIb (CD16b)), and/or FcγRIV and/or the neonatal Fc receptor (FcRn).

As used herein, a sulfur atom“corresponding to” a particular cysteine residue of a particular SEQ ID NO. should be understood to include the sulfur atom of any cysteine residue that one of skill in the art would understand to align to the particular cysteine of the particular sequence. The protein sequence alignment of human IgG1 (UniProtKB: P01857), human IgG2 (UniProtKB: P01859), human IgG3 (UniProtKB: P01860), and human IgG4 (UniProtKB: P01861) is provided below (aligned with Clustal Omega Multiple Pairwise Alignment). The alignment indicates cysteine residues (e.g., sulfur atoms of

) “ ” ( ) O f in the art would readily be able to perform such an alignment with any IgG v

determine the sulfur atom of a cysteine that corresponds to any sulfur atom of a particular cysteine of a particular SEQ ID NO. described herein. For example, one of skill in the art would readily be able to determine that Cys10 of SEQ ID NO: 10 (the first cysteine of the conserved CPPC motif of the hinge region of the Fc domain) corresponds to, for example, Cys109 of IgG1, Cys106 of IgG2, Cys156 of IgG3, Cys29 of SEQ ID NO: 1, Cys9 of SEQ ID NO: 2, Cys30 of SEQ ID NO: 3, or Cys10 of SEQ ID NO: 10.

As used herein, a nitrogen atom“corresponding to” a particular lysine residue of a particular SEQ ID NO. should be understood to include the nitrogen atom of any lysine residue that one of skill in the art would understand to align to the particular lysine of the particular sequence. The protein sequence alignment of human IgG1 (UniProtKB: P01857), human IgG2 (UniProtKB: P01859), human IgG3 (UniProtKB: P01860), and human IgG4 (UniProtKB: P01861) is provided below (aligned with Clustal Omega Multiple Pairwise Alignment). The alignment indicates lysine residues (e.g., nitrogen atoms of lysine residues) that“correspond to” one another (in boxes and indicated by the * symbol). One of skill in the art would readily be able to perform such an alignment with any IgG variant of the invention to determine the nitrogen atom of a lysine that corresponds to any nitrogen atom of a particular lysine of a particular SEQ ID NO. described herein. For example, one of skill in the art would readily be able to determine that Lys35 of SEQ ID NO: 10 corresponds to, for example, Lys129 of IgG1, Lys126 of IgG2, Lys176 of IgG3, Lys51 of SEQ ID NO: 1, Lys31 of SEQ ID NO: 2, Lys50 of SEQ ID NO: 3, or Lys30 of SEQ ID NO: 10.

Activation of Immune Cells

Fc-gamma receptors (FcγRs) bind the Fc portion of immunoglobulin G (IgG) and play important roles in immune activation and regulation. For example, the IgG Fc domains in immune complexes (ICs) engage FcγRs with high avidity, thus triggering signaling cascades that regulate immune cell activation. The human FcγR family contains several activating receptors (FcγRI, FcγRIIa, FcγRIIc, FcγRIIIa, and FcγRIIIb) and one inhibitory receptor (FcγRIIb). FcγR signaling is mediated

contain immune tyrosine activating motifs (ITAMs) for activating FcγRs and immune tyrosine inhibitory motifs (ITIM) for inhibitory receptor FcγRIIb. In some embodiments, FcγR binding by Fc domains results in ITAM phosphorylation by Src family kinases; this activates Syk family kinases and induces downstream signaling networks, which include PI3K and Ras pathways.

In the conjugates described herein, the portion of the conjugates including monomers or dimers of cyclic heptapeptides bind to lipopolysaccharides (LPS) in the cell membrane of Gram-negative bacteria to disrupt and permeabilize the cell membrane, leading to cell death and/or sensitization to other antibiotics, while the Fc domain portion of the conjugates bind to FcγRs (e.g., FcRn, FcγRI, FcγRIIa, FcγRIIc, FcγRIIIa, and FcγRIIIb) on immune cells and activate phagocytosis and effector functions, such as antibody-dependent cell-mediated cytotoxicity (ADCC), thus leading to the engulfment and destruction of bacterial cells by immune cells and further enhancing the antibacterial activity of the conjugates.

Examples of immune cells that may be activated by the conjugates described herein include, but are not limited to, macrophages, neutrophils, eosinophils, basophils, lymphocytes, follicular dendritic cells, natural killer cells, and mast cells. IV. Linkers

A linker refers to a linkage or connection between two or more components in a conjugate described herein (e.g., between two cyclic heptapeptides in a conjugate described herein, between a cyclic heptapeptide and an Fc domain in a conjugate described herein, and betwee a dimer of two cyclic peptapeptide and an Fc domain in a conjugate described herein). Linkers in conjguates having an Fc domain covalently linked to dimers of cyclic heptapeptides

I n a conjugate containing an Fc domain covalently linked to one or more dimers of cyclic heptapeptides as described herein, a linker in the conjugate (e.g., L’, L, or L 1 ) may be a branched structure. As described further herein, a linker in a conjugate described herein (e.g., L’, L, or L 1 ) may be a multivalent structure, e.g., a divalent or trivalent structure having two or thee arms, respectively. In some embodiments when the linker has three arms, two of the arms may be attached to the first and second cyclic heptapeptides and the third arm may be attached to the Fc domain monomer or Fc domain. In some embodiments when the linker has two arms, one arm may be attached to an Fc domain and the other arm may be attached to one of the two cyclic heptapeptides. In other embodiments, a linker with two arms may be used to attach the two cyclic heptapeptides on a conjugate containing an Fc domain covalently linked to one or more dimers of cyclic heptapeptides.

In some embodiments, the linker L’ in the conjugate described by formula (I) is described by formula (L):

in which L is a remainder of L’; A1 is a 1-5 amino acid peptide (e.g., a 1-4, 1-3, or 1-2 amino acid peptide) covalently attached to the linking nitrogen in each M1 or is absent; and A2 is a 1-5 amino acid peptide (e.g., a 1-4, 1-3, or 1-2 amino acid peptide) covalently attached to the linking nitrogen in each M2 or is absent. In some embodiments, a conjugate described herein may contain a linker that has a trivalent structure (e.g., a trivalent linker; a linker of formula (L-I)). A trivalent linker has three arms, in which each arm is conjugated to a component of the conjugate (e.g., a first arm conjugated to a first cyclic heptapeptide, a second arm conjugated to a second heptapeptide, and a third arm conjugated to an Fc domain in the conjugate described herein). In some embodiments, the one or more dimers of cyclic heptapeptides in the conjugates described herein may each be, independently, connected to an atom in the Fc domain.

In some embodiments, a linker in a conjugate having an Fc domain covalently linked to one or more dimers of cyclic heptapeptides is described by formula (L-I):

wherein L A is described by formula G A1 -(Z A1 )g1-(Y A1 )h1-(Z A2 )i1-(Y A2 )j1-(Z A3 )k1-(Y A3 )l1-(Z A4 )m1-(Y A4 )n1-(Z A5 )o1- G A2 ; L B is described by formula G B1 -(Z B1 )g2-(Y B1 )h2-(Z B2 )i2-(Y B2 )j2-(Z B3 )k2-(Y B3 )l2-(Z B4 )m2-(Y B4 )n2-(Z B5 )o2-G B2 ; L C is described by formula G C1 -(Z C1 )g3-(Y C1 )h3-(Z C2 )i3-(Y C2 )j3-(Z C3 )k3-(Y C3 )l3-(Z C4 )m3-(Y C4 )n3-(Z C5 )o3-G C2 ; G A1 is a bond attached to Q in formula (L-I); G A2 is a bond attached to the first cyclic heptapeptide or a peptide (e.g., a peptide including 1-5 amino acid residue(s)) attached to the first cyclic heptapeptide, if the peptide is present; G B1 is a bond attached to Q in formula (L-I); G B2 is a bond attached to the second cyclic heptapeptide or a peptide (e.g., a peptide including 1-5 amino acid residue(s)) attached to the second cyclic heptapeptide, if the peptide is present; G C1 is a bond attached to Q in formula (L-I); G C2 is a bond attached to an Fc domain; each of Z A1 , Z A2 , Z A3 , Z A4 , Z A5 , Z B1 , Z B2 , Z B3 , Z B4 , Z B5 , Z C1 , Z C2 , Z C3 , Z C4 , and Z C5 is, independently, optionally substituted C1-C20 alkylene, optionally substituted C1-C20 heteroalkylene, optionally substituted C2-C20 alkenylene, optionally substituted C2-C20 heteroalkenylene, optionally substituted C2-C20 alkynylene, optionally substituted C2-C20 heteroalkynylene, optionally substituted C3-C20 cycloalkylene, optionally substituted C3-C20 heterocycloalkylene, optionally substituted C4-C20 cycloalkenylene, optionally substituted C4-C20 heterocycloalkenylene, optionally substituted C8-C20 cycloalkynylene, optionally substituted C8-C20 heterocycloalkynylene, optionally substituted C5-C15 arylene, or optionally substituted C2-C15 heteroarylene; each of Y A1 , Y A2 , Y A3 , Y A4 , Y B1 , Y B2 , Y B3 , Y B4 , Y C1 , Y C2 , Y C3 , and Y C4 is, independently, O, S, NR i , P, carbonyl, thiocarbonyl, sulfonyl, phosphate, phosphoryl, or imino; R i is H, optionally substituted C1-C20 alkyl, optionally substituted C1-C20 heteroalkyl, optionally substituted C2-C20 alkenyl, optionally substituted C2-C20 heteroalkenyl, optionally substituted C2-C20 alkynyl, optionally substituted C2-C20 heteroalkynyl, optionally substituted C

substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C5-C15 aryl, or optionally substituted C2-C15 heteroaryl; each of g1, h1, i1, j1, k1, l1, m1, n1, o1, g2, h2, i2, j2, k2, l2, m2, n2, o2, g3, h3, i3, j3, k3, l3, m3, n3, and o3 is, independently, 0 or 1; Q is a nitrogen atom, optionally substituted C1-C20 alkylene, optionally substituted C1-C20 heteroalkylene, optionally substituted C2-C20 alkenylene, optionally substituted C2-C20 heteroalkenylene, optionally substituted C2-C20 alkynylene, optionally substituted C2-C20

heteroalkynylene, optionally substituted C3-C20 cycloalkylene, optionally substituted C3-C20 heterocycloalkylene, optionally substituted C4-C20 cycloalkenylene, optionally substituted C4-C20 heterocycloalkenylene, optionally substituted C8-C20 cycloalkynylene, optionally substituted C8-C20 heterocycloalkynylene, optionally substituted C5-C15 arylene, or optionally substituted C2-C15 heteroarylene.

In some embodiments, depending on the structure and chemical formula of L C , L C may have two points of attachment to the Fc domain (e.g., two G C2 ).

Linkers of formula (L-I) that may be used in conjugates described herein include, but are not limi

,

Linkers in conjugates having an Fc domain covalently linked to monomers of

cyclic heptapeptides

In a conjugate containing an Fc domain covalently linked to one or more monomers of cyclic heptapeptides as described herein, a linker in the conjugate (e.g., L’, or L) may be a divalent structure having two arms. One arm in a divalent linker may be attached to the monomer of cyclic heptapeptide and the other arm may be attached to the Fc domain. In some embodiments, the one or more monomers of cyclic heptapeptides in the conjugates described herein may each be, independently, connected to an atom in the Fc domain. In some embodiments, a linker is described by formula (L-I):

wherein J 1 is a bond attached to a cyclic heptapeptide or a peptide (e.g., a peptide including 1-5 amino acid residue(s)) attached to the cyclic heptapeptide, if the peptide is present; J 2 is a bond attached to an Fc domain; each of Q1, Q2, Q3, Q4, and Q5 is, independently, optionally substituted C1-C20 alkylene, optionally substituted C1-C20 heteroalkylene, optionally substituted C2-C20 alkenylene, optionally substituted C2-C20 heteroalkenylene, optionally substituted C2-C20 alkynylene, optionally substituted C2-C20 heteroalkynylene, optionally substituted C3-C20 cycloalkylene, optionally substituted C3-C20 heterocycloalkylene, optionally substituted C4-C20 cycloalkenylene, optionally substituted C4-C20 heterocycloalkenylene, optionally substituted C8-C20 cycloalkynylene, optionally substituted C8-C20 heterocycloalkynylene, optionally substituted C5-C15 arylene, or optionally substituted C2-C15 heteroarylene; each of T 1 , T 2 , T 3 , T 4 is, independently, O, S, NR i , P, carbonyl, thiocarbonyl, sulfonyl, phosphate, phosphoryl, or imino; R i is H, optionally substituted C1-C20 alkyl, optionally substituted C1- C20 heteroalkyl, optionally substituted C2-C20 alkenyl, optionally substituted C2-C20 heteroalkenyl, optionally substituted C2-C20 alkynyl, optionally substituted C2-C20 heteroalkynyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C5-C15 aryl, or optionally substituted C2-C15 heteroaryl; and each of g, h, i, j, k, l, m, n, and o is, independently, 0 or 1.

In some embodiments, depending on the structure and chemical formula of L-I, J 2 may have two points of attachment to the Fc domain (e.g., two J 2 ).

Linkers of formula (L-I) that may be used in conjugates described herein include, but are not

limited to, ,

,

to 26. In some embodiments, a linker provides space, rigidity, and/or flexibility between the cyclic heptapeptides and the Fc domain in the conjguates described here or between two cyclic heptapeptides in the conjugates described herein. In some embodiments, a linker may be a bond, e.g., a covalent bond, e.g., an amide bond, a disulfide bond, a C-O bond, a C-N bond, a N-N bond, a C-S bond, or any kind of bond created from a chemical reaction, e.g., chemical conjugation. In some embodiments, a linker (L’, L, or L 1 as shown in any one of formulas (I)-(XVII)) includes no more than 250 atoms (e.g., 1-2, 1-4, 1-6, 1-8, 1-10, 1-12, 1-14, 1-16, 1-18, 1-20, 1-25, 1-30, 1-35, 1-40, 1-45, 1-50, 1-55, 1-60, 1-65, 1-70, 1-75, 1-80, 1-85, 1-90, 1-95, 1-100, 1-110, 1-120, 1-130, 1-140, 1-150, 1-160, 1-170, 1-180, 1-190, 1-200, 1-210, 1- 220, 1-230, 1-240, or 1-250 atom(s); 250, 240, 230, 220, 210, 200, 190, 180, 170, 160, 150, 140, 130, 120, 110, 100, 95, 90, 85, 80, 75, 70, 65, 60, 55, 50, 45, 40, 35, 30, 28, 26, 24, 22, 20, 18, 16, 14, 12, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 atom(s)). In some embodiments, a linker (L’, L, or L 1 ) includes no more than 250 non-hydrogen atoms (e.g., 1-2, 1-4, 1-6, 1-8, 1-10, 1-12, 1-14, 1-16, 1-18, 1-20, 1-25, 1-30, 1-35, 1-40, 1- 45, 1-50, 1-55, 1-60, 1-65, 1-70, 1-75, 1-80, 1-85, 1-90, 1-95, 1-100, 1-110, 1-120, 1-130, 1-140, 1-150, 1-160 1-170 1-180 1-190 1-200 1-210 1-220 1-230 1-240 or 1-250 non-hydrogen atom(s); 250 240 230, 220, 210, 200, 190, 180, 170, 160, 150, 140, 130, 120, 110, 100, 95, 9

50, 45, 40, 35, 30, 28, 26, 24, 22, 20, 18, 16, 14, 12, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 non-hydrogen atom(s)). In some embodiments, the backbone of a linker (L’, L, or L 1 ) includes no more than 250 atoms (e.g., 1-2, 1-4, 1-6, 1-8, 1-10, 1-12, 1-14, 1-16, 1-18, 1-20, 1-25, 1-30, 1-35, 1-40, 1-45, 1-50, 1-55, 1-60, 1-65, 1-70, 1-75, 1-80, 1-85, 1-90, 1-95, 1-100, 1-110, 1-120, 1-130, 1-140, 1-150, 1-160, 1-170, 1-180, 1-190, 1-200, 1-210, 1-220, 1-230, 1-240, or 1-250 atom(s); 250, 240, 230, 220, 210, 200, 190, 180, 170, 160, 150, 140, 130, 120, 110, 100, 95, 90, 85, 80, 75, 70, 65, 60, 55, 50, 45, 40, 35, 30, 28, 26, 24, 22, 20, 18, 16, 14, 12, 10, 9, 8, 7, 6, 5, 4, 3, 2, or 1 atom(s)). The“backbone” of a linker refers to the atoms in the linker that together form the shortest path from one part of the conjugate to another part of the conjugate. The atoms in the backbone of the linker are directly involved in linking one part of the conjugate to another part of the conjugate. For examples, hydrogen atoms attached to carbons in the backbone of the linker are not considered as directly involved in linking one part of the conjugate to another part of the conjugate.

Molecules that may be used to make linkers (L’, L, or L 1 ) include at least two functional groups, e.g., two carboxylic acid groups. In some embodiments of a trivalent linker, two arms of a linker may contain two dicarboxylic acids, in which the first carboxylic acid may form a covalent linkage with the first cyclic heptapeptide in the conjugate and the second carboxylic acid may form a covalent linkage with the second cyclic heptapeptide in the conjugate, and the third arm of the linker may for a covalent linkage (e.g., a C-O bond) with an Fc domain in the conjugate. In some embodiments of a divalent linker, the divalent linker may contain two carboxylic acids, in which the first carboxylic acid may form a covalent linkage with one component (e.g., a cyclic heptapeptide) in the conjugate and the second carboxylic acid may form a covalent linkage (e.g., a C-S bond or a C-N bond) with another component (e.g., an Fc domain) in the conjugate. In some embodiments, when the cyclic heptapeptide is attached to a peptide (e.g., a peptide including a 1-5 amino acid residue(s)) at the linking nitrogen, the linker may form a covalent linkage with the peptide.

In some embodiments, dicarboxylic acid molecules may be used as linkers (e.g., a dicarboxylic acid linker). For example, in a conjugate containing an Fc domain covalently linked to one or more dimers of cyclic heptapeptides, the first carboxylic acid in a dicarboxylic acid molecule may form a covalent linkage with the linking nitrogen of the first cyclic heptapeptide and the second carboxylic acid may form a covalent linkage with the linking nitrogen of the second cyclic heptapeptide.

In some embodiments, when the first and/or second cyclic heptapeptide is attached to a peptide (e.g., a peptide including a 1-5 amino acid residue(s)) at the linking nitrogen, the first carboxylic acid in a dicarboxylic acid linker may form a covalent linkage with the terminal amine group at the end of the peptide that is attached to the first cyclic heptapeptide and the second carboxylic acid in the dicarboxylic acid linker may form a covalent linkage with the terminal amine group at the end of the peptide that is attached to the second cyclic heptapeptide. Examples of dicarboxylic acids molecules that may be used to form

limited to,

,

,

Other examples of dicarboxylic acids molecules that may be used to form linkers include, but are not limited to,

,

In some embodiments, dicarboxylic acid molecules, such as the ones described herein, may be further functionalized to contain one or more additional functional groups.

In some embodiments, when the cyclic heptapeptide is attached to a peptide (e.g., a peptide including a 1-5 amino acid residue(s)) at the linking nitrogen, the linking group may comprise a moiety comprising a carboxylic acid moiety and an amino moiety that are spaced by from 1 to 25 atoms. Exam les of such linkin rou s include but are not limited to

In some embodiments, a molecule containing an azide group may be used to form a linker, in which the azide group may undergo cycloaddition with an alkyne to form a 1,2,3-triazole linkage. In some embodiments, a molecule containing an alkyne group may be used to form a linker, in which the alkyne group may undergo cycloaddition with an azide to form a 1,2,3-triazole linkage. In some embodiments, a molecule containing a maleimide group may be used to form a linker, in which the maleimide group may react with a cysteine to form a C-S linkage. In some embodiments, a molecule containing one or more sulfonic acid groups may be used to form a linker, in which the sulfonic acid group may form a sulfonamide linkage with the linking nitrogen in a cyclic heptapeptide. In some embodiments, a molecule containing one or more isocyanate groups may be used to form a linker, in which the isocyanate group may form a urea linkage with the linking nitrogen in a cyclic heptapeptide. In some embodiments, a molecule containing one or more haloalkyl groups may be used to form a linker, in which the haloalkyl group may form a covalent linkage, e.g., C-N and C-O linkages, with a cyclic heptapeptide.

In some embodiments, a linker (L’, L, or L 1 ) may comprise a synthetic group derived from, e.g., a synthetic polymer (e.g., a polyethylene glycol (PEG) polymer). In some embodiments, a linker may comprise one or more amino acid residues. In some embodiments, a linker may be an amino acid sequence (e.g., a 1-25 amino acid, 1-10 amino acid, 1-9 amino acid, 1-8 amino acid, 1-7 amino acid, 1-6 amino acid, 1-5 amino acid, 1-4 amino acid, 1-3 amino acid, 1-2 amino acid, or 1 amino acid sequence). In some embodiments, a linker (L’, L, or L 1 ) may include one or more optionally substituted C1-C20 alkylene, optionally substituted C1-C20 heteroalkylene (e.g., a PEG unit), optionally substituted C2-C20 alkenylene (e.g., C2 alkenylene), optionally substituted C2-C20 heteroalkenylene, optionally substituted C2-C20 alkynylene, optionally substituted C2-C20 heteroalkynylene, option

cycloalkylene (e.g., cyclopropylene, cyclobutylene), optionally substituted C3-C20 heterocycloalkylene, optionally substituted C4-C20 cycloalkenylene, optionally substituted C4-C20 heterocycloalkenylene, optionally substituted C8-C20 cycloalkynylene, optionally substituted C8-C20 heterocycloalkynylene, optionally substituted C5-C15 arylene (e.g., C6 arylene), optionally substituted C2-C15 heteroarylene (e.g., imidazole, pyridine), O, S, NR i (R i is H, optionally substituted C1-C20 alkyl, optionally substituted C1-C20 heteroalkyl, optionally substituted C2-C20 alkenyl, optionally substituted C2-C20 heteroalkenyl, optionally substituted C2-C20 alkynyl, optionally substituted C2-C20 heteroalkynyl, optionally substituted C3-C20 cycloalkyl, optionally substituted C3-C20 heterocycloalkyl, optionally substituted C4-C20 cycloalkenyl, optionally substituted C4-C20 heterocycloalkenyl, optionally substituted C8-C20 cycloalkynyl, optionally substituted C8-C20 heterocycloalkynyl, optionally substituted C5-C15 aryl, or optionally substituted C2-C15 heteroaryl), P, carbonyl, thiocarbonyl, sulfonyl, phosphate, phosphoryl, or imino.

Covalent conjugation of two or more components in a conjugate using a linker may be accomplished using well-known organic chemical synthesis techniques and methods. Complementary functional groups on two components may react with each other to form a covalent bond. Examples of complementary reactive functional groups include, but are not limited to, e.g., maleimide and cysteine, amine and activated carboxylic acid, thiol and maleimide, activated sulfonic acid and amine, isocyanate and amine, azide and alkyne, and alkene and tetrazine. Site-specific conjugation to a polypeptide (e.g., an Fc domain monomer or an Fc domain) may accomplished using techniques known in the art.

Exemplary techniques for site-specific conjugation of a small molecule to an Fc domain are provided in Agarwall. P., et al. Bioconjugate Chem.26:176-192 (2015).

Other examples of functional groups capable of reacting with amino groups include, e.g., alkylating and acylating agents. Representative alkylating agents include: (i) an α-haloacetyl group, e.g., XCH 2 CO- (where X=Br, Cl, or I); (ii) a N-maleimide group, which may react with amino groups either through a Michael type reaction or through acylation by addition to the ring carbonyl group; (iii) an aryl halide, e.g., a nitrohaloaromatic group; (iv) an alkyl halide; (v) an aldehyde or ketone capable of Schiff’s base formation with amino groups; (vi) an epoxide, e.g., an epichlorohydrin and a bisoxirane, which may react with amino, sulfhydryl, or phenolic hydroxyl groups; (vii) a chlorine-containing of s-triazine, which is reactive towards nucleophiles such as amino, sufhydryl, and hydroxyl groups; (viii) an aziridine, which is reactive towards nucleophiles such as amino groups by ring opening; (ix) a squaric acid diethyl ester; and (x) an α-haloalkyl ether.

Examples of amino-reactive acylating groups include, e.g., (i) an isocyanate and an

isothiocyanate; (ii) a sulfonyl chloride; (iii) an acid halide; (iv) an active ester, e.g., a nitrophenylester or N- hydroxysuccinimidyl ester; (v) an acid anhydride, e.g., a mixed, symmetrical, or N-carboxyanhydride; (vi) an acylazide; and (vii) an imidoester. Aldehydes and ketones may be reacted with amines to form Schiff’s bases, which may be stabilized through reductive amination.

It will be appreciated that certain functional groups may be converted to other functional groups f f f f f this purpose include conversion of amines to carboxyls using reagents such

conversion of amines to thiols using reagents such as N-acetylhomocysteine thiolactone, S- acetylmercaptosuccinic anhydride, 2-iminothiolane, or thiol-containing succinimidyl derivatives;

conversion of thiols to carboxyls using reagents such as α -haloacetates; conversion of thiols to amines using reagents such as ethylenimine or 2-bromoethylamine; conversion of carboxyls to amines using reagents such as carbodiimides followed by diamines; and conversion of alcohols to thiols using reagents such as tosyl chloride followed by transesterification with thioacetate and hydrolysis to the thiol with sodium acetate. V. Antibacterial Agents

In some embodiments, one or more antibacterial agents may be administered in combination (e.g., administered substantially simultaneously (e.g., in the same pharmaceutical composition or in separate pharmaceutical compositions) or administered separately at different times) with a conjugate described herein (e.g., a conjugate of any one of formulas (I)-(XXXIII)).

Antibacterial agents may be grouped into several classes, e.g., quinolones, carbapenems, macrolides, DHFR inhibitors, aminoglycosides, ansamycins (e.g., geldanamycin, herimycin, and rifaximin), carbacephem (e.g., loracarbef), cephalosporins (e.g., cefadroxil, cefaolin, cefalotin, cefalothin, cephalexin, e.g., cefaclor, cefamandole, cefoxitin, cefprozil, cefuroxime, cefdinir, cefditoren,

cefoperazone, cefotaxime, cefpodoxime, ceftazidime, and ceftobiprole), glycopeptides (e.g., teicoplanin, vancomycin, telavancin, dalbavancin, and oritavancin), lincosamides (e.g., clindamycin and lincomycin), lipopeptides (e.g., daptomycin), monobactams (e.g., aztreonam), nitrofurans (e.g., furazolidone and nitrofurantoin), oxazolidinones, pleuromutilins, penicillins , sulfonamides, and tetracyclines (e.g., eravacycline, demeclocycline, doxycycline, minocycline, oxytetracycline, and tetracycline). Quinolones include, but are not limited to, ciprofloxacin, enoxacin, gatifloxacin, gemifloxacin, levofloxacin, lomefloxacin, moxifloxacin, nalidixic acid, norfloxacin, ofloxacin, trovafloxacin, grepafloxacin, sparfloxacin, and temafloxacin. Carbapenems include, but are not limited to, ertapenem, doripenem,

imipenem/cilastatin, and meropenem. Macrolides include, but are not limited to, solithromycin, azithromycin, clarithromycin, dirithromycin, erythromycin, roxithromycin, troleandomycin, telithromycin, and spiramycin. In some embodiments, a macrolide is solithromycin. DHFR inhibitors include, but are not limited to, diaminoquinazoline, diaminopyrroloquinazoline, diaminopyrimidine, diaminopteridine, and diaminotriazines. Aminoglycosides include, but are not limited to, plazomicin, amikacin, gentamicin, gamithromycin, kanamycin, neomycin, netilmicin, tobramycin, paromomycin, streptomycin, and spectinomycin. Oxazolidinones include, but are not limited to, linezolid, tedizolid, posizolid, radezolid, and furazolidone. Pleuromutilins include, but are not limited to, retapamulin, valnemulin, tiamulin, azamulin, and lefamulin. Penicillins include, but are not limited to, amoxicillin, ampicillin, azlocillin, carbenicillin, cloxacillin, dicloxacillin, flucloxacillin, mezlocillin, methicillin, nafcillin, oxacillin, penicillin G, penicillin V, piperacillin, penicillin G, temocillin, and ticarcillin. Sulfonamides include, but are not limited to, mafenide, sulfacetamide, sulfadiazine, silver sulfadiazine, sulfadimethoxine, sulfamethizole, sulfamethoxazole, sulfanilimide, sulfasalazine, sulfisoxazole, trimethoprim-sulfamethoxazole (C

and sulfonamidochrysoidine.

In some embodiments, the antibacterial agent used in combination with a conjugate described herein (e.g., a conjugate of any one of formulas (I)-(XXXIII)) is selected from the group consisting of linezolid, tedizolid, posizolid, radezolid, retapamulin, valnemulin, tiamulin, azamulin, lefamulin, plazomicin, amikacin, gentamicin, gamithromycin, kanamycin, neomycin, netilmicin, tobramycin, paromomycin, streptomycin, spectinomycin, geldanamycin, herbimycin, rifaximin, loracarbef, ertapenem, doripenem, imipenem/cilastatin, meropenem, cefadroxil, cefazolin, cefalotin, cefalexin, cefaclor, cefamandole, cefoxitin, cefprozil, cefuroxime, cefixime, cefdinir, cefditoren, cefoperazone, cefotaxime, cefpodoxime, ceftazidime, ceftibuten, ceftizoxime, ceftriaxone, cefepime, ceftaroline fosamil, ceftobiprole, teicoplanin, vancomycin, telavancin, dalbavancin, oritavancin, clindamycin, lincomycin, daptomycin, solithromycin, azithromycin, clarithromycin, dirithromycin, erythromycin, roxithromycin, troleandomycin, telithromycin, spiramycin, aztreonam, furazolidone, nitrofurantoin, amoxicillin, ampicillin, azlocillin, carbenicillin, cloxacillin, dicloxacillin, flucloxacillin, mezlocillin, methicillin, nafcillin, oxacillin, penicillin g, penicillin v, piperacillin, penicillin g, temocillin, ticarcillin, amoxicillin clavulanate, ampicillin/sulbactam,

piperacillin/tazobactam, ticarcillin/clavulanate, bacitracin, ciprofloxacin, enoxacin, gatifloxacin, gemifloxacin, levofloxacin, lomefloxacin, moxifloxacin, nalidixic acid, norfloxacin, ofloxacin, trovafloxacin, grepafloxacin, sparfloxacin, temafloxacin, mafenide, sulfacetamide, sulfadiazine, silver sulfadiazine, sulfadimethoxine, sulfamethizole, sulfamethoxazole, sulfanilimide, sulfasalazine, sulfisoxazole, trimethoprim-sulfamethoxazole (tmp-smx), sulfonamidochrysoidine, eravacycline, demeclocycline, doxycycline, minocycline, oxytetracycline, tetracycline, clofazimine, dapsone, capreomycin, cycloserine, ethambutol(bs), ethionamide, isoniazid, pyrazinamide, rifampicin, rifabutin, rifapentine, streptomycin, arsphenamine, chloramphenicol, fosfomycin, fusidic acid, metronidazole, mupirocin, platensimycin, quinupristin/dalfopristin, thiamphenicol, tigecycline, tinidazole, and trimethoprim, prodrugs thereof, and pharmaceutically acceptable salts thereof.

In some embodiments, the antibacterial agent used in combination with a conjugate described herein (e.g., a conjugate of any one of formulas (I)-(XXXIII)) is tedizolid, azithromycin, meropenem, amikacin, levofloxacin, rifampicin, linezolid, erythromycin, or solithromycin. In some embodiments, the antibacterial agent used in combination with a conjugate described herein (e.g., a conjugate of any one of formulas (I)-(XXXIII)) is tedizolid, azithromycin, meropenem, amikacin, or levofloxacin. In some embodiments, the antibacterial agent used in combination with a conjugate described herein (e.g., a conjugate of any one of formulas (I)-(XXXIII)) is tiamulin. In some embodiments, the antibacterial agent used in combination with a conjugate described herein (e.g., a conjugate of any one of formulas (I)- (XXXIII)) is solithromycin. VI. Methods

Methods described herein include, e.g., methods of protecting against or treating a bacterial infection (e.g., a Gram-negative bacterial infection) in a subject and methods of preventing, stabilizing, or f ( G ) f bacterial infection (e.g., a Gram-negative bacterial infection) in a subject inc

subject a conjugate described herein (e.g., a conjugate of any one of formulas (I)-(XXXIII)) or a pharmaceutical composition thereof. In some embodiments, the bacterial infection is caused by Gram- negative bacteria. In some embodiments, the bacterial infection is caused by a resistant strain of bacteria. In some embodiments, the resistant strain of bacteria is a resistant strain of E. coli. A method of preventing, stabilizing, or inhibiting the growth of bacteria, or killing bacteria (e.g., Gram-negative bacteria) includes contacting the bacteria (e.g., Gram-negative bacteria) or a site susceptible to bacterial growth (e.g., Gram-negative bacterial growth) with a conjugate described herein (e.g., a conjugate of any one of formulas (I)-(XXXIII)) or a pharmaceutical composition thereof. In some embodiments, the bacterial infection is caused by Gram-negative bacteria. In some embodiments, the bacterial infection is caused by a resistant strain of bacteria. In some embodiments, the resistant strain of bacteria is a resistant strain of E. coli. In some embodiments, a conjugate used in any methods described herein (e.g., a conjugate of any one of formulas (I)-(XXXIII)) may bind to LPS in the cell membrane of Gram-negative bacteria to disrupt and permeabilize the cell membrane, leading to cell death and/or sensitization to other antibiotics. Furthermore, in addition to disrupting the cell membrane of Gram-negative bacteria, the Fc domain in the conjugates described herein binds to the FcγRs (e.g., FcRn, FcγRI, FcγRIIa, FcγRIIc, FcγRIIIa, and FcγRIIIb) on immune cells and serves as a gradient against which immune cells chemotax to the site of bacterial infection and/or growth. The binding of the Fc domain in the conjugates described herein to the FcγRs on immune cells activates phagocytosis and effector functions, such as antibody- dependent cell-mediated cytotoxicity (ADCC), thus leading to the engulfment and destruction of Gram- negative bacterial cells by immune cells and further enhancing the antibacterial activity of the conjugates.

Moreover, methods described herein also include methods of protecting against or treating sepsis in a subject by administering to the subject a conjugate described herein (e.g., a conjugate of any one of formulas (I)-(XXXIII)). In some embodiments, the method further includes administering to the subject an antibacterial agent. Methods described herein also include methods of preventing lipopolysaccharides (LPS) in Gram-negative bacteria (e.g., a resistant strain of Gram-negative bacteria or a resistant strain of E. coli (e.g., E. coli BAA-2469)) from activating a immune system in a subject by administering to the subject a conjugate described herein (e.g., a conjugate of any one of formulas (I)-(XXXIII)). In some embodiments of the method, the method prevents LPS from activating a macrophage. In some embodiments, the method further includes administering to the subject an antibacterial agent. In some embodiments, a conjugate used in any methods described herein (e.g., a conjugate of any one of formulas (I)-(XXXIII)) may bind to LPS in the cell membrane of Gram-negative bacteria to disrupt and permeabilize the cell membrane, leading to cell death and/or sensitization to other antibiotics.

In some embodiments, the methods described herein may further include administering to the subject an antibacterial agent in addition to a conjugate described herein (e.g., a conjugate of any one of formulas (I)-(XXXIII)). Methods described herein also include methods of protecting against or treating a bacterial infection in a subject by administering to said subject (1) a conjugate described herein (e.g., a conjugate of any one of formulas (I)-(XXXIII)) and (2) an antibacterial agent. Methods described herein f f contacting the bacteria or a site susceptible to bacterial growth with (1) a co

(e.g., a conjugate of any one of formulas (I)-(XXXIII)) and (2) an antibacterial agent.

In some embodiments, the conjugate described herein (e.g., a conjugate of any one of formulas (I)-(XXXIII)) is administered first, followed by administering of the antibacterial agent alone. In some embodiments, the antibacterial agent is administered first, followed by administering of the conjugate described herein alone. In some embodiments, the conjugate described herein and the antibacterial agent are administered substantially simultaneously (e.g., in the same pharmaceutical composition or in separate pharmaceutical compositions). In some embodiments, the conjugate described herein or the antibacterial agent is administered first, followed by administering of the conjugate described herein and the antibacterial agent substantially simultaneously (e.g., in the same pharmaceutical composition or in separate pharmaceutical compositions). In some embodiments, the conjugate described herein and the antibacterial agent are administered first substantially simultaneously (e.g., in the same pharmaceutical composition or in separate pharmaceutical compositions), followed by administering of the conjugate described herein or the antibacterial agent alone. In some embodiments, when a conjugate described herein (e.g., a conjugate of any one of formulas (I)-(XXXIII)) and an antibacterial agent are administered together (e.g., substantially simultaneously in the same or separate pharmaceutical compositions, or separately in the same treatment regimen), the MIC of each of the conjugate and the antibacterial agent may be lower than the MIC of each of the conjugate and the antibacterial agent when each is used alone in a treatment regimen. VII. Pharmaceutical Compositions and Preparations

A conjugate described herein may be formulated in a pharmaceutical composition for use in the methods described herein. In some embodiments, a conjugate described herein may be formulated in a pharmaceutical composition alone. In some embodiments, a conjugate described herein may be formulated in combination with an antibacterial agent in a pharmaceutical composition. In some embodiments, the pharmaceutical composition includes a conjugate described herein (e.g., a conjugate described by any one of formulas (I)-(XXXIII)) and pharmaceutically acceptable carriers and excipients. Acceptable carriers and excipients in the pharmaceutical compositions are nontoxic to recipients at the dosages and concentrations employed. Acceptable carriers and excipients may include buffers such as phosphate, citrate, HEPES, and TAE, antioxidants such as ascorbic acid and methionine, preservatives such as hexamethonium chloride, octadecyldimethylbenzyl ammonium chloride, resorcinol, and benzalkonium chloride, proteins such as human serum albumin, gelatin, dextran, and immunoglobulins, hydrophilic polymers such as polyvinylpyrrolidone, amino acid residues such as glycine, glutamine, histidine, and lysine, and carbohydrates such as glucose, mannose, sucrose, and sorbitol.

Examples of other excipients include, but are not limited to, antiadherents, binders, coatings, compression aids, disintegrants, dyes, emollients, emulsifiers, fillers (diluents), film formers or coatings, flavors, fragrances, glidants (flow enhancers), lubricants, sorbents, suspensing or dispersing agents, or sweeteners. Exemplary excipients include, but are not limited to: butylated hydroxytoluene (BHT),

( ) pyrrolidone, citric acid, crospovidone, cysteine, ethylcellulose, gelatin, hydro

hydroxypropyl methylcellulose, lactose, magnesium stearate, maltitol, mannitol, methionine,

methylcellulose, methyl paraben, microcrystalline cellulose, polyethylene glycol, povidone, pregelatinized starch, propyl paraben, retinyl palmitate, shellac, silicon dioxide, sodium carboxymethyl cellulose, sodium citrate, sodium starch glycolate, sorbitol, starch (corn), stearic acid, stearic acid, sucrose, talc, titanium dioxide, vitamin A, vitamin E, vitamin C, and xylitol.

The conjugates herein may have ionizable groups so as to be capable of preparation as pharmaceutically acceptable salts. These salts may be acid addition salts involving inorganic or organic acids or the salts may, in the case of acidic forms of the conjugates herein be prepared from inorganic or organic bases. Frequently, the conjugates are prepared or used as pharmaceutically acceptable salts prepared as addition products of pharmaceutically acceptable acids or bases. Suitable pharmaceutically acceptable acids and bases are well-known in the art, such as hydrochloric, sulphuric, hydrobromic, acetic, lactic, citric, or tartaric acids for forming acid addition salts, and potassium hydroxide, sodium hydroxide, ammonium hydroxide, caffeine, various amines, and the like for forming basic salts. Methods for preparation of the appropriate salts are well-established in the art.

Representative acid addition salts include, but are not limited to, acetate, adipate, alginate, ascorbate, aspartate, benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphorate,

camphorsulfonate, citrate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, fumarate, glucoheptonate, glycerophosphate, hemisulfate, heptonate, hexanoate, hydrobromide, hydrochloride, hydroiodide, 2-hydroxy-ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, malate, maleate, malonate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oleate, oxalate, palmitate, pamoate, pectinate, persulfate, 3-phenylpropionate, phosphate, picrate, pivalate, propionate, stearate, succinate, sulfate, tartrate, thiocyanate, toluenesulfonate, undecanoate, and valerate salts. Representative alkali or alkaline earth metal salts include, but are not limited to, sodium, lithium, potassium, calcium, and magnesium, as well as nontoxic ammonium, quaternary ammonium, and amine cations, including, but not limited to ammonium, tetramethylammonium, tetraethylammonium, methylamine, dimethylamine, trimethylamine, triethylamine, and ethylamine.

Depending on the route of administration and the dosage, a conjugate herein or a pharmaceutical composition thereof used in the methods described herein will be formulated into suitable pharmaceutical compositions to permit facile delivery. A conjugate (e.g., a conjugate of any one of formulas (I)-(XXXIII)) or a pharmaceutical composition thereof may be formulated to be administered intramuscularly, intravenously (e.g., as a sterile solution and in a solvent system suitable for intravenous use), intradermally, intraarterially, intraperitoneally, intralesionally, intracranially, intraarticularly,

intraprostatically, intrapleurally, intratracheally, intranasally, intravitreally, intravaginally, intrarectally, topically, intratumorally, peritoneally, subcutaneously, subconjunctival, intravesicularlly, mucosally, intrapericardially, intraumbilically, intraocularally, orally (e.g., a tablet, capsule, caplet, gelcap, or syrup), topically (e.g., as a cream, gel, lotion, or ointment), locally, by inhalation, by injection, or by infusion (e.g., continuous infusion, localized perfusion bathing target cells directly, catheter, lavage, in cremes, or lipid

) f composition thereof may be in the form of, e.g., tablets, capsules, pills, pow

suspensions, emulsions, solutions, gels including hydrogels, pastes, ointments, creams, plasters, drenches, osmotic delivery devices, suppositories, enemas, injectables, implants, sprays, preparations suitable for iontophoretic delivery, or aerosols. The compositions may be formulated according to conventional pharmaceutical practice.

A conjugate described herein may be formulated in a variety of ways that are known in the art. For use as treatment of human and animal subjects, a conjugate described herein can be formulated as pharmaceutical or veterinary compositions. Depending on the subject (e.g., a human) to be treated, the mode of administration, and the type of treatment desired, e.g., prophylaxis or therapy, a conjugate described herein is formulated in ways consonant with these parameters. A summary of such techniques is found in Remington: The Science and Practice of Pharmacy, 22nd Edition, Lippincott Williams & Wilkins (2012); and Encyclopedia of Pharmaceutical Technology, 4th Edition, J. Swarbrick and J. C. Boylan, Marcel Dekker, New York (2013), each of which is incorporated herein by reference.

Formulations may be prepared in a manner suitable for systemic administration or topical or local administration. Systemic formulations include those designed for injection (e.g., intramuscular, intravenous or subcutaneous injection) or may be prepared for transdermal, transmucosal, or oral administration. The formulation will generally include a diluent as well as, in some cases, adjuvants, buffers, and preservatives. The conjugates can be administered also in liposomal compositions or as microemulsions. Systemic administration may also include relatively noninvasive methods such as the use of suppositories, transdermal patches, transmucosal delivery and intranasal administration. Oral administration is also suitable for conjugates herein. Suitable forms include syrups, capsules, and tablets, as is understood in the art.

The pharmaceutical compositions can be administered parenterally in the form of an injectable formulation. Pharmaceutical compositions for injection can be formulated using a sterile solution or any pharmaceutically acceptable liquid as a vehicle. Formulations may be prepared as solid forms suitable for solution or suspension in liquid prior to injection or as emulsions. Pharmaceutically acceptable vehicles include, but are not limited to, sterile water, physiological saline, and cell culture media (e.g., Dulbecco’s Modified Eagle Medium (DMEM), α-Modified Eagles Medium (α-MEM), F-12 medium). Such injectable compositions may also contain amounts of nontoxic auxiliary substances such as wetting or emulsifying agents, pH buffering agents, such as sodium acetate and sorbitan monolaurate. Formulation methods are known in the art, see e.g., Pharmaceutical Preformulation and Formulation, 2nd Edition, M. Gibson, Taylor & Francis Group, CRC Press (2009).

The pharmaceutical compositions can be prepared in the form of an oral formulation.

Formulations for oral use include tablets containing the active ingredient(s) in a mixture with non-toxic pharmaceutically acceptable excipients. These excipients may be, for example, inert diluents or fillers (e.g., sucrose, sorbitol, sugar, mannitol, microcrystalline cellulose, starches including potato starch, calcium carbonate, sodium chloride, lactose, calcium phosphate, calcium sulfate, or sodium phosphate); granulating and disintegrating agents (e.g., cellulose derivatives including microcrystalline cellulose,

) ( sucrose, glucose, sorbitol, acacia, alginic acid, sodium alginate, gelatin, sta

microcrystalline cellulose, magnesium aluminum silicate, carboxymethylcellulose sodium, methylcellulose, hydroxypropyl methylcellulose, ethylcellulose, polyvinylpyrrolidone, or polyethylene glycol); and lubricating agents, glidants, and antiadhesives (e.g., magnesium stearate, zinc stearate, stearic acid, silicas, hydrogenated vegetable oils, or talc). Formulations for oral use may also be provided as chewable tablets, or as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent (e.g., potato starch, lactose, microcrystalline cellulose, calcium carbonate, calcium phosphate or kaolin), or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium, for example, peanut oil, liquid paraffin, or olive oil. Powders, granulates, and pellets may be prepared using the ingredients mentioned above under tablets and capsules in a conventional manner using, e.g., a mixer, a fluid bed apparatus or a spray drying equipment.

Other pharmaceutically acceptable excipients for oral formulations include, but are not limited to, colorants, flavoring agents, plasticizers, humectants, and buffering agents. Formulations for oral use may also be provided as chewable tablets, or as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent (e.g., potato starch, lactose, microcrystalline cellulose, calcium carbonate, calcium phosphate or kaolin), or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium, for example, peanut oil, liquid paraffin, or olive oil. Powders, granulates, and pellets may be prepared using the ingredients mentioned above under tablets and capsules in a conventional manner using, e.g., a mixer, a fluid bed apparatus or a spray drying equipment.

Dissolution or diffusion controlled release of a conjugate described herein (e.g., a conjugate of any one of formulas (I)-(XXXIII)) or a pharmaceutical composition thereof can be achieved by appropriate coating of a tablet, capsule, pellet, or granulate formulation of the conjugate, or by incorporating the conjugate into an appropriate matrix. A controlled release coating may include one or more of the coating substances mentioned above and/or, e.g., shellac, beeswax, glycowax, castor wax, carnauba wax, stearyl alcohol, glyceryl monostearate, glyceryl distearate, glycerol palmitostearate, ethylcellulose, acrylic resins, dl-polylactic acid, cellulose acetate butyrate, polyvinyl chloride, polyvinyl acetate, vinyl pyrrolidone, polyethylene, polymethacrylate, methylmethacrylate, 2-hydroxymethacrylate, methacrylate hydrogels, 1,3 butylene glycol, ethylene glycol methacrylate, and/or polyethylene glycols. In a controlled release matrix formulation, the matrix material may also include, e.g., hydrated methylcellulose, carnauba wax and stearyl alcohol, carbopol 934, silicone, glyceryl tristearate, methyl acrylate-methyl methacrylate, polyvinyl chloride, polyethylene, and/or halogenated fluorocarbon.

The pharmaceutical composition may be formed in a unit dose form as needed. The amount of active component, e.g., a conjugate described herein (e.g., a conjugate of any one of formulas (I)- (XXXIII)), included in the pharmaceutical compositions are such that a suitable dose within the designated range is provided (e.g., a dose within the range of 0.01-100 mg/kg of body weight). VIII. Routes of Administration and Dosages

In any of the methods described herein, conjugates herein may be administered by any appropriate route for treating or protecting against a bacterial infection (e.g., a Gram-negative bacterial infection), or for preventing, stabilizing, or inhibiting the growth of bacteria, or killing bacteria (e.g., Gram- negative bacteria). Conjugates described herein may be administered to humans, domestic pets, livestock, or other animals with a pharmaceutically acceptable diluent, carrier, or excipient. In some embodiments, administering comprises administration of any of the conjugates described herein (e.g., conjugates of any one of formulas (I)-(XXXIII)) or compositions intramuscularly, intravenously (e.g., as a sterile solution and in a solvent system suitable for intravenous use), intradermally, intraarterially, intraperitoneally, intralesionally, intracranially, intraarticularly, intraprostatically, intrapleurally, intratracheally, intranasally, intravitreally, intravaginally, intrarectally, topically, intratumorally, peritoneally, subcutaneously, subconjunctival, intravesicularlly, mucosally, intrapericardially, intraumbilically, intraocularally, orally (e.g., a tablet, capsule, caplet, gelcap, or syrup), topically (e.g., as a cream, gel, lotion, or ointment), locally, by inhalation, by injection, or by infusion (e.g., continuous infusion, localized perfusion bathing target cells directly, catheter, lavage, in cremes, or lipid compositions). In some embodiments, if an antibacterial agent is also administered in addition to a conjugate described herein, the antibacterial agent or a pharmaceutical composition thereof may also be administered in any of the routes of administration described herein.

The dosage of a conjugate described herein (e.g., a conjugate of any one of formulas (I)-(XXXIII)) or pharmaceutical compositions thereof depends on factors including the route of administration, the disease to be treated (e.g., the extent and/or condition of the bacterial infection), and physical characteristics, e.g., age, weight, general health, of the subject. Typically, the amount of the conjugate or the pharmaceutical composition thereof contained within a single dose may be an amount that effectively prevents, delays, or treats the bacterial infection without inducing significant toxicity. A pharmaceutical composition may include a dosage of a conjugate described herein ranging from 0.01 to 500 mg/kg (e.g., 0.01, 0.1, 0.2, 0.3, 0.4, 0.5, 1, 2, 3, 4, 5, 10, 15, 20, 25, 30, 35, 40, 45, 50, 100, 150, 200, 250, 300, 350, 400, 450, or 500 mg/kg) and, in a more specific embodiment, about 0.1 to about 30 mg/kg and, in a more specific embodiment, about 1 to about 30 mg/kg. In some embodiments, when a conjugate described herein (e.g., a conjugate of any one of formulas (I)-(XXXIII)) and an antibacterial agent are administered together (e.g., substantially simultaneously in the same or separate pharmaceutical compositions, or separately in the same treatment regimen), the dosage needed of the conjugate described herein may be lower than the dosage needed of the conjugate if the conjugate was used alone in a treatment regimen.

A conjugate described herein (e.g., a conjugate of any one of formulas (I)-(XXXIII)) or a pharmaceutical composition thereof may be administered to a subject in need thereof, for example, one or more times (e.g., 1-10 times or more; 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 times) daily, weekly, monthly, biannually, annually, or as medically necessary. Dosages may be provided in either a single or multiple dosage regimens. The timing between administrations may decrease as the medical condition improves or increase as the health of the patient declines. The dosage and frequency of administration may be adapted by the physician in accordance with conventional factors such as t

different parameters of the subject. EXAMPLES

E xample 1– Preparation of Fc Constructs

Reverse translations of the amino acids comprising the protein constructs (SEQ ID NOs: 1, 3, 5, 7, 9, 11, and 13) were synthesized by solid-phase synthesis. The oligonucleotide templates were cloned into pcDNA3.1 (Life Technologies, Carlsbad, CA, USA) at the cloning sites BamHI and XhoI (New England Biolabs, Ipswich, MA, USA) and included signal sequences derived from the human Interleukin-2 or human albumin. The pcDNA3.1 plasmids were transformed into Top10 E. coli cells (LifeTech). DNA was amplified, extracted, and purified using the PureLink® HiPure Plasmid Filter Maxiprep Kit (LifeTech). The plasmid DNA is delivered, using the ExpiFectamine™ 293 Transfection Kit (LifeTech), into HEK-293 cells per the manufacturer’s protocol. Cells were centrifuged, filtered, and the supernatants were purified using MabSelect Sure Resin (GE Healthcare, Chicago, IL, USA). Purified molecules were analyzed using 4-12% Bis Tris SDS PAGE gels by loading 1-2 µg of each molecule into the gel, and staining using instant Blue staining. Each gel included a molecular weight ladder with the indicated molecular weight standards (FIGS.1-7). Reduced and non-reduced lanes are denoted by“R” and“NR”. Example 2– Synthesis of Intermediate 1 (Int-1)

De radation of ol m xin B to tri-Boc ol m xin B c clohe ta e tide

Polymyxin B (100 g, 72.2 mmol) was dissolved in acetonitrile (1000 mL) and water (500 mL) and stirred at room temperature for 10 mins. TEA (58.5 g, 8.0 eq) was added and the mixture stirred for a further 10 mins. Boc2O (94.6 g, 6.0 eq) was subsequently added in one portion and the mixture stirred for 6 hrs at 20 o C. Savinase ® (300 mL) was then added. The pH of the resulting mixture was adjusted to 9.0 with aq 4M sodium hydroxide solution (10 mL) and the reaction mixture stirred at 25 o C. Additional Savinase ® (100 mL) was added after 17 hrs, and another quantity of Savinase ® (100 mL x2) was added after 26 hrs. After an overall reaction time of 80 hrs, the mixture was diluted with ethyl acetate (2000 mL). After separation of the layers, the organic phase was washed with 0.1 M NaOH solution (1000 mL x2, 10V x2), then water (1000 mL, 10V). The organic layer was dried over anhy

solvent evaporated at reduced pressure. The residue was purified by silica gel chromatography eluting with 80% (EtOAc : MeOH : H2O·NH3 = 40:10:1) in ethyl acetate to give the title compound (49.8 g, 65.0%). LCMS: m/z (M + H) + calcd for C50H83N11O14:1061.61; found:1062.5 Example 3– Synthesis of Int-2 (Tripeptide (Dab-Thr-Dab))

Step a.

NH2-Dab(Boc)-OMe (HCl salt) (5.000 g, 1eq.), Z-NH-Thr-OH (5.049 g, 1.05eq.), EDCI (5.350 g, 1.5eq.), HOBt (3.733 g, 1.5eq.) and NaHCO3 (3.095 g, 2eq.) were weighed into a 100- mL round bottom flask.24 mL of DCM/DMF (4:1) was added into the flask. The mixture was stirred at room temperature for less than 3 hrs. (TLC or LC/MS monitoring). After completion, EtOAc (200 mL) was added to dilute the reaction mixture and washed with 1N aq. HCl, saturated NaHCO3 and brine. Dried with Na2SO4 and condensed. The residue was purified with normal phase silica (2~7% MeOH/DCM) to give 8.24 g pure desired product (>95%). Step b.

Starting material (SM) (8.24 g) was dissolved in 100 mL of MeOH/EtOAc. And 5%Pd/C (3.75 g, 0.1eq.) was added. Under H2 balloon, the reaction mixture was stirred for 2h. Checked LC/MS. Celite filtration and MeOH wash. Dried to give 5.87 g of free amine (>99%). The material was used in the next step without purification. Step c.

NH2-Thr-Dab(Boc)-OMe (1.46 g, 1eq.), Z-NH-Dab(Boc)-OH(DCHA salt) (2.463, 1.05eq.), EDCI (1.260 g, 1.5eq.), HOBt (0.888 g, 1.5eq.) and NaHCO3 (0.738, 2 eq.) were weighed into a 100- mL round bottom flask.20 mL of DCM/DMF (4:1) was added into the flask. The mixture was stirred at room added to dilute the reaction mixture and washed with 1N aq. HCl, saturated

with Na2SO4 and condensed. The residue was purified with normal phase silica (2~7% MeOH/DCM) to give 2.78 g pure desired product (>95% isolated yield). (The reaction was repeated at 4.4 g scale and gave the similar result.) Step d.

A solution of the Cbz-tripeptide (1.55 g, 2.32 mmol) in methanol (10 mL) was charged with 5% Pd/C (0.300 g) and flushed with hydrogen from a balloon. After stirring overnight under hydrogen atmosphere, LCMS showed complete conversion. Pd/C was removed by filtration through Celite. The filtrate was concentrated and used in the next Step without further purification. Example 4–

A solution of the Cbz-tripeptide-OMe (7.85 g, 11.76 mmol, described in Step 3 of Int-2 synthesis), dissolved in THF (30 mL), and water (30 mL), was treated with powdered LiOH (0.338 g, 14.1 mmol) and stirred at room temperature. After 15 minutes LCMS showed consumption of starting material. The reaction was made slightly acidic by adding concentrated HCl (aq), then extracted into ethyl acetate, dried over sodium sulfate, concentrated, and purified by reversed phase liquid chromatography (RPLC) using an Isco CombiFlash liquid chromatograph eluted with 5% to 100% methanol and water, using no modifier. Yield 7.2 g, 94%. Ion(s) found by LCMS: (M+H)+ = 654.2. Step a.

A stirring solution of methyl (2S)-2-[(N-{(2S)-2-amino-4-[(tert-butoxycarbonyl)amino]butan oyl}-L- threonyl)amino]-4-[(tert-butoxycarbonyl)amino]butanoate (0.399 g, 0.748 mmol), 2,2'- {[(benzyloxy)carbonyl]azanediyl}diacetic acid (0.100, 0.374 mmol), and DIEA (0.261 mL, 1.50 mmol), in DMF (1 mL), were treated with a solution of HATU (0.285 g, 0.748 mmol), dropwise over 30 minutes, at room temperature. The desired product was isolated by RPLC using an Isco CombiFlash liquid chromatograph eluted with 10% to 100% methanol and water, using no modifier. Yield 0.200 g, 41%. Ion(s) found by LCMS: (M+H)+ = 1298.7. Step b.

A solution of dimethyl (2S,5S,8S,16S,19S,22S)-12-[(benzyloxy)carbonyl]-2,8,16,22-te trakis{2- [(tert-butoxycarbonyl)amino]ethyl}-5,19-bis[(1R)-1-hydroxyet hyl]-4,7,10,14,17,20-hexaoxo- 3,6,9,12,15,18,21-heptaazatricosane-1,23-dioate (0.380 g, 0.242 mmol), in methanol(1 mL) was treated with a solution of lithium hydroxide(0.028 g, 1.171 mmol), in water(1 mL), then stirred at room

temperature for 30 minutes. The reaction was made slightly acidic(pH=5) with concentrated HCl (several drops). The desired product was isolated directly by RPLC using an Isco CombiFlash liquid

chromatograph eluted with 10% to 100% methanol and water, using no modifier. Yield 0.164 g, 44%. Ion(s) found by LCMS: [(M-2Boc)/2]+H+ = 535.8, [(M-3Boc)/2]+H+ =485.8 .

A solution of (2S,5S,8S,16S,19S,22S)-12-[(benzyloxy)carbonyl]-2,8,16,22-te trakis{2-[(tert- butoxycarbonyl)amino]ethyl}-5,19-bis[(1R)-1-hydroxyethyl]-4, 7,10,14,17,20-hexaoxo-3,6,9,12,15,18,21- heptaazatricosane-1,23-dioic acid (0.164 g, 0.129 mmol), tri-Boc polymyxin heptapeptide (0.329 g, 0.310 mmol, Int-1), DIEA (0.146 mL, 0.839 mmol), and DMF (1 mL), was treated with a solution of HATU (0.175 g, 0.460 mmol) in DMF (1 mL), dropwise over 30 minutes. The crude reaction mixture was taken on to the next Step without purification. Ion(s) found by LCMS: [(M-2Boc)/3]+H+ = 1053.3, [(M-3Boc)/3]+H+ = 1019.9, [(M-4Boc)/3]+H+ = 986.6. .

Crude Cbz deca Boc intermediate (DMF solution) was diluted with methanol (10 mL), charged with 5%Pd/C (0.150 g), and hydrogen from a balloon. The reaction was monitored by LCMS. After 2hr the mixture was filtered through celite, concentrated and purified by RPLC using an Isco CombiFlash liquid chromatograph eluted with 10% to 100% methanol and water, using no modifier. Yield 0.252 g, 61% (two steps). Ion(s) found by LCMS: [M/3]+H+ = 1075.3.

A solution of Step d product (0.135 g, 0.042 mmol), 3-(2-{2-[2-(2,5-dioxo-2,5-dihydro-1H-pyrrol-1- yl)ethoxy]ethoxy}ethoxy)propanoic acid (0.015 g, 0.050 mmol), and DIEA (0.026 mL, 0.150 mmol), in DMF (1 mL), were treated with HATU (0.019 g, 0.050 mmol), while stirring at room temperature. After 30 minutes, the product was purified by RPLC using an Isco CombiFlash liquid chromatograph eluted with 10% to 100% methanol and water, using no modifier. Yield 0.112 g, 76% yield. Ion(s) found by LCMS: [(M-3Boc)/3]+H+ = 1069.6, [(M-4Boc)/3]+H+ = 1036.3, [(M-5Boc)/3]+H+ = 1003.0. Ste f.

A solution of Deca Boc Int-4 (0.112 g, 0.032 mmol), dissolved in DC

TFA (1 mL), while stirring at room temperature. After 5 minutes, the product was concentrated and purified by RPLC using an Isco CombiFlash liquid chromatograph eluted with 0% to 100% acetonitrile and water, using trifluoroacetic acid as the modifier. Yield of Int-4 was 0.071 g, 88% yield. Ion(s) found by LCMS: [M/3]+H+ = 836.1, [M/4]+H+ =627.3, [M/5]+H+ = 502.1.

Step a. Synthesis of Int-5a

HATU (1.56 g, 4.11 mmol) in DMF (1.5 mL) was added, dropwise, to a solution of 2,2'- {[(benzyloxy)carbonyl]azanediyl}diacetic acid (0.5 g, 1.87 mmol), norleu-OMe hydrochloride salt (0.71 g, 3.93 mmol), and triethylamine (1.13 g, 11.23 mmol) in DMF (5 mL) over a period of 30 minutes. The mixture was stirred for an additional 20 minutes then applied directly to RPLC using an Isco Combiflash liquid chromatograph eluted with 10% to 95% acetonitrile and water using 0.1 % TFA modifier. The pure fractions were pooled and lyophilized to afford the CBZ-protected di-ester as a clear oil, ion found by LC/MS [M+H]+ = 522.6. The di-ester intermediate was stirred in a 1/1/2 mixture (10 mL) of

THF/methanol/water containing LiOH (0.18 g, 7.48 mmol) for 20 minutes. The mixture was applied directly to RPLC using an Isco Combiflash liquid chromatograph eluted with 10% to 95% acetonitrile and water using 0.1 % TFA modifier. The pure fractions were pooled and lyophilized to afford (2S,2'S)-2,2'- ({[(benzyloxy)carbonyl]azanediyl}bis[(1-oxoethane-2,1-diyl)a zanediyl])dihexanoic acid as a white solid. Yield: 43%, 2 Steps. Ion found by LC/MS [m-H]- = 492.3. Step b. Synthesis of Int-5b.

EDC (0.38 g, 2.0 mmol) was added to a solution of Int-1 (2.0 g, 1.88 mmol), Cbz-D-Ser-OH (0.48 g, 2.0 mmol), and HOBt (0.31 g, 2.0 mmol) in a 5/1 mixture of DCM/DMF (15 mL). The mixture was stirred for an additional 2 hours and applied directly to RPLC using an Isco Combiflash liquid chromatograph eluted with 10% to 95% acetonitrile and water using 0.1 % TFA modifier. The pure fractions were pooled and lyophilized to afford the CBZ-protected intermediate a white solid. Ion found by LC/MS [m/2+H]+ = 542.4 (loss of 1 Boc group). The CBZ-protected intermediate was stirred in methanol (50 mL) containing 1 g of 5% Pd/C under 1 atmosphere of hydrogen for 6 hours. The mixture was filtered and concentrated to afford the Int-5b (free amine) as a white solid. Yield: 73%, 2 Steps. Ion fo

525.6 (loss of 1 Boc group). Step c. Synthesis of Int-5c.

HATU (370 mg, 0.97 mmol) in DMF (2 mL) was added, dropwise over 30 minutes, to a stirring solution of Int-5b (1.10 g, 0.95 mmol), CBZ-Thr-OH (253 mg, 1.0 mmol), and triethylamine (300 mg, 2.97 mmol) in DMF (6 mL). The reaction was stirred for 30 minutes and then applied directly to RPLC using an Isco Combiflash liquid chromatograph eluted with 10% to 95% acetonitrile and water using 0.1 % TFA modifier. The pure fractions were pooled and lyophilized to afford 1.1 g of the CBZ-protected intermediate a white solid. LC/MS [m/2+H] + = 593.0 (loss of 2 Boc groups). The CBZ-protected intermediate was stirred in methanol (50 mL) containing 0.30 g of 5% Pd/C under 1 atmosphere of hydrogen for 6 hours. The mixture was filtered and concentrated to afford the Int-5c (free amine) as a white solid. Yield: 66%, 2 Steps. Ion found by LC/MS [m/2+H] + = 525.8 (loss of 2 Boc groups). Step d. Synthesis of Int-5d.

The title compound was prepared from Int-5c and Z-(γBoc)-Dab-OH in an analogous manner as described in Step c. Yield: 78%. Ion found by LC/MS [M/2+H] + 676.2 (loss of 1 Boc group). Step e. Synthesis of Int-5e.

HATU (0.11 g, 0.30 mmol) in DMF (0.5 mL) was added, dropwise, to a stirring mixture of Int-5d (0.44 g, 0.30 mmol), and INT-A1 (0.70 g, 0.14 mmol) in DMF (1.5 mL) over a period of 20 minutes. The mixture was applied directly to RPLC using an Isco Combiflash liquid chromatograph eluted with 30% to 95% methanol and water using 0.1 % TFA modifier. The pure fractions were pooled and lyophilized to afford the CBZ-protected intermediate, LC/MS [m/3+H] + 1020.0 (loss of 3 Boc groups). The CBZ- intermediate was stirred in methanol (10 mL) in the presence of 5% Pd/C (100 mg) under 1 atmosphere of hydrogen for 2 hours. The mixture was filtered and concentrated then purified by RPLC using an Isco Combiflash liquid chromatograph eluted with 30% to 95% acetonitrile and water using no modifier. The pure fractions were pooled and lyophilized to afford the title compound as a white solid. Yield: 51%, 2 Steps. Ion found by LC/MS [m/2+H] + = 1562.6 (loss of 2 Boc groups). f. n h i f In - .

HATU (57 mg, 0.15 mmol) in DMF (1 mL) was added, dropwise, to a stirring mixture of propargyl- peg-4-carboxylic acid (42 mg 016 mmol) and Int-5e (345 mg 011 mmol) in DMF (15 mL) over a period of 20 minutes. The mixture was applied directly to RPLC using an Isco Com

eluted with 30% to 95% methanol and water using 0.1 % TFA modifier. The pure fractions were pooled and lyophilized to afford the octa Boc-protected intermediate. The octa Boc-protected intermediate was stirred in a 1/1 mixture of TFA/DCM (5 mL) containing thioanisole (106 mg, 0.85 mmol) for 20 minutes at ambient temperature. The mixture was concentrated then purified by RPLC using an Isco Combiflash liquid chromatograph eluted with 0% to 70% acetonitrile and water using no modifier. The pure fractions were pooled and lyophilized to afford the title compound Int-5 as a white solid. Yield: 36%, 2 Steps.

LC/MS [m/4+H] + = 667.2. Example 7– Synthesis of Int-6 (azido-peg4-hexyl pyrolidine-3,4-dicarboxamide)

Step a. Synthesis of Int-6a (more polar diastereomer)

HATU (3.1 g, 8.1 mmol) in DMF (5 mL) was added, dropwise, to a solution of racemic-trans1- (tert-butoxycarbonyl)pyrrolidine-3,4-dicarboxylic acid (1 g, 3.9 mmol), H-Norleu-OMe-hydrochloride (1.5 g, 8.1 mmol), and triethylamine (4.0 g, 39.5 mmol) in DMF (10 mL) over a period of 20 minutes. The mixture was stirred for an additional 20 minutes then applied directly to RPLC using an Isco Combiflash liquid chromatograph eluted with 10% to 95% acetonitrile and water using 0.1 % TFA modifier. The two diasteomers were separated, pooled and lyophilized separately into the more polar diasteomer (a) and the less polar diastereomer (b):Ion found by LC/MS [M+H]+ = 414.2 (loss of 1 Boc group) for both Boc- protected intermediates. Each diastereomer was stirred in a 1/1 mixture of DCM/TFA (8 mL) for 20 minutes then concentrated and purified by RPLC using an Isco Combiflash liquid chromatograph eluted with 0% to 75% acetonitrile and water using no modifier. Yield: 88%, 2 Steps. Ion found by LC/MS

[M+H]+ = 414.2.

(The stereochemistry of the polar diastereomer was assigned using single crystal x-ray crystallography) S

Azido-PEG4-NHS ester (254 mg, 0.65 mmol) in DMF (1 mL) was added, dropwise to a mixture of Int-6a (more polar diastereomer from Step A), TFA salt (265 mg, 0.50 mmol) and triethylamine (66 mg, 0.65 mmol) in DMF (1 mL) and the mixture was stirred at ambient temperature for 2 hours. The mixture was applied directly to RPLC using an Isco Combiflash liquid chromatograph eluted with 10% to 95% acetonitrile and water using 0.1 % TFA modifier.The pure fractions were pooled and lyophilized to afford the di-ester intermediate. LC/MS [M+H] + = 687.4. The di-ester intermediate was stirred in a 1/1/2 mixture of MeOH/THF/DI water containing LiOH (36 mg, 1.5 mmol) at ambient temperature for 5 minutes. The mixture was acidified with glcial acetic acid, concentrated and applied directly to RPLC using an Isco Combiflash liquid chromatograph eluted with 10% to 95% acetonitrile and water using 0.1 % TFA modifier.The pure fractions were pooled and lyophilized to afford the title compound Int-6 as a clear oil. Yield: 69%, 2 Steps. Ion found by LC/MS [M+H] + = 659.4.

Example 8– Synthesis of Int-7

Step a. penta-Boc-PMB-decapeptide (Int-7a)

Boc polymyxin B heptapeptide (0.464 g, 0.437 mmol, Int-1), and tripeptide acid (0.316 g, 0.398 mmol, Int-3), were dissolved in DMF (1 mL), DIEA (0.229 mL, 1.31 mmol), then treated with HATU (0.166 g, 0.437 mmol). After stirring for 30 min LCMS showed complete conversion. The crude Cbz-product was diluted with methanol (4 mL), and charged with 5%Pd/C(0.250 g) and placed under a hydrogen atmosphere. After 2hr the mixture was filtered through Celite, concentrated, and purified by RPLC using an Isco CombiFlash liquid chromatograph eluted with 10% to 100% methanol and water, using no modifier. Yield 0.640 g, 94% (two steps). Ion(s) found by LCMS: [(M-1Boc)/2]+H+ = 1054.6, [(M- 2Boc)/2]+H+ = 1004.6. Step b. Coupling of Int-6 and Int-7a to give Int-7

HATU (150 mg, 0.40 mmol) in DMF (1 mL) was added, dropwise, to a stirring mixture of Int-6 (120 mg 030 mmol) and penta-Boc-PMB-decapeptide (626 mg 040 mmol Int-7a) in DMF (2 mL) over a period of 20 minutes. The mixture was applied directly to RPLC using an

chromatograph eluted with 30% to 95% methanol and water using 0.1 % TFA modifier. The pure fractions were pooled and lyophilized to afford the Boc-protected intermediate, ion found by LC/MS [m/3+H] + 1150.4 (loss of 3 Boc groups). The Boc-protected intermediate was stirred in a 1/1 mixture of TFA/DCM (6 mL) containing thioanisole (225 mg, 1.81 mmol) for 20 minutes at ambient temperature. The mixture was concentrated then purified by RPLC using an Isco Combiflash liquid chromatograph eluted with 0% to 70% acetonitrile and water using no modifier. The pure fractions were pooled and lyophilized to afford the title compound (Int-7) as a white solid. Yield: 68%, 2 Steps. Ion found by LC/MS [m/3+H] + = 917.2.

Step a. Synthesis of benzyloxycarbonyl -3,4-bis{[(2S)-1-methoxy-1-oxooctan-2- yl]carbamoyl}pyrrolidine

A solution of racemic 1-[(benzyloxy)carbonyl]pyrrolidine-3,4-dicarboxylic acid (3.69 g, 12.6 mmol), methyl (2S)-2-aminooctanoate (4.58 g, 26.4 mmol), and DIEA (13.8 mL, 79.3 mmol), in DMF (20 mL) was treated with HATU (10.0 g, 26.4 mmol) at room temperature for 30 minutes. The crude product was purified by RPLC using an Isco CombiFlash liquid chromatograph eluted with 5% to 100% methanol and water, using no modifier. Two diastereomers where observed and were separable. The desired more polar isomer (Int-8a) had the shorter retention time and was taken on to Step b. Yield 1.44 g, 19%. Step b. Synthesis of Int-8

A solution of benzyloxycarbonyl -3,4-bis{[(2S)-1-methoxy-1-oxooctan-2-yl]carbamoyl}pyrrolidi ne (1.44 g, 2.38 mmol, more polar isomer from Step A, Int-8a) in THF (10 mL) and water (10 mL) was treated with powdered lithium hydroxide (0.143 g, 5.96 mmol) at room temperature

showed complete conversion. The reaction was made slightly acidic by adding concentrated HCl, and concentrated to an oil. The oil was purified by RPLC using an Isco CombiFlash liquid chromatograph eluted with 5% to 100% acetonitrile and water, using TFA as the modifier. Yield 1.14 g, 83% yield. Example 10– S nthesis of aminooctano l enta Boc ol m xin B deca e tide Int-18

Step a. Synthesis of penta Boc polymyxin B decapeptide (Int-7a)

Tri-Boc polymyxin B heptapeptide (0.464, 0.437 mmol, Int-1), and tripeptide acid (0.316, 0.398 mmol, Int-3), were dissolved in DMF (1 mL), DIEA (0.229 mL, 1.31 mmol), then treated with HATU (0.166 g, 0.437 mmol). After stirring for 30 min LCMS showed complete conversion. The crude Cbz-product was diluted with methanol (4 mL), and charged with 5%Pd/C (0.250 g) and placed under a hydrogen atmosphere. After 2 hr the mixture was filtered through Celite, concentrated, and purified by RPLC using an Isco CombiFlash liquid chromatograph eluted with 10% to 100% methanol and water, using no modifier. Yield of Int-7a was 0.640 g, 94% (two steps). Ion(s) found by LCMS: [(M-1Boc)/2]+H+ = 1054.6, [(M-2Boc)/2]+H+ = 1004.6. Step b. Synthesis of aminooctanoyl penta Boc polymyxin B decapeptide (Int-18)

A solution of Int-7a (3.00 g, 1.918 mmol), Cbz-aminooctanoic acid (0.591 g, 2.01 mmol), and DIEA (1.05 mL, 6.04 mmol), in DMF (10 mL), was treated with a solution of HATU (0.766 g, 2.01 mmol) dissolved in DMF (3 mL), dropwise over 60 minutes. After 2h, LCMS showed complete consumption of starting material. The crude mixture was treated with 5% Pd/C (1.7 g), vacuum flushed with hydrogen, and stirred under a hydrogen atmosphere for 2hr or until complete by LCMS. The crude reaction was filtered through Celite, concentrated, and purified by RPLC using an Isco CombiFlash liquid

chromatograph eluted with 20% to 100% methanol and water, using no modifier. Yield of Int-18 was 2.59 g, 79% (two steps). Ion(s) found by LCMS: [M/2]+H+ = 853.4. Example 11– Synthesis of Int-9

A solution of Int-8 (0.401 g, 0.70 mmol), Int-7a (2.18 g, 1.39 mmol), and DIEA (0.76 mL, 4.39 mmol) in DMF, was treated with a solution of HATU (0.557 g, 1.46 mmol) in DMF (5 mL), dropwise over 1h. After stirring for an additional 30 minutes the reaction was charged with 5% Pd/C (1.5 g), vacuum flushed with hydrogen, and stirred under a hydrogen atmosphere for 2h. The resulting mixture was filtered through celite, concentrated, and purified by RPLC using an Isco CombiFlash liquid

chromatograph eluted with 30% to 100% methanol and water, using no modifier. Yield 1.39 g, 56% yield. .

A solution of product from Step a (0.300 g, 0.085 mmol), Cbz-PEG8-acid (0.049 g, 0.085 mmol), and DIEA (0.049 mL, 0.280 mmol), in DMF (2 mL), was treated with a solution of HATU (0.036 g, 0.0934 mmol) in DMF (1 mL), dropwise over 1h. After stirring an additional 30 minutes the reaction was charged with 5% Pd/C(0.35 g), vacuum flushed with hydrogen, and stirred under a hydrogen atmosphere for 2h. The resulting mixture was filtered through celite, concentrated and purified by RPLC using an Isco CombiFlash liquid chromatograph eluted with 30% to 100% methanol and water, using no modifier. Yield 0.315 g, 94% yield. Ion(s) found by LCMS: [M-2(Boc)/3]+H+ = 1252.4. .

A solution of product from Step b (0.315 g, 0.080 mmol), bis-sulfone acid (0.044 g, 0.088 mmol), and N-methylmorpholine (0.020 mL, 0.175 mmol), in DMF (3 mL), was treated with a solution of HATU (0.033 g, 0.088 mmol), in DMF (1 mL), dropwise over 1h. After stirring for an additional 30 minutes, the reaction was concentrated and purified by RPLC using an Isco CombiFlash liquid chromatograph eluted with 30% to 100% methanol and water, using no modifier. Yield 0.243 g, 6

LCMS: did not ionize. Ste d.

A solution of product from Step c(0.216 g, 0.049 mmol), suspended in DCM (2 mL) was treated with TFA (2 mL) at room temperature. After stirring for 5 minutes, the reaction was concentrated and purified by RPLC using an Isco CombiFlash liquid chromatograph eluted with 5% to 100% acetonitrile and water, using TFA as the modifier. Yield of Int-9 was 0.226 g, quantitative yield. Ion(s) found by LCMS: [M/3]+H+ = 1146.3, [M/4]+H+ = 860.0, [M/5]+H+ = 688.2.

Step a. Synthesis of amino Peg 8 penta Boc polymyxin B undecapeptide

A solution of Int-18 (0.600 g, 0.352 mmol), Cbz-amino Peg 8 acid (0.223 g, 0.387 mmol), and DIEA (0.202 mL, 1.161 mmol), in DMF (3 mL) was treated with a solution of HATU (0.147 g, 0.387 mmol), in DMF (1 mL), dropwise over 1hr. After stirring for an additional 30 minutes, the reaction was charged with 5% Pd/C (0.400 g), vacuum flushed with hydrogen, and stirred under a hydrogen atmosphere until complete by LCMS (2h). The resulting mixture was filtered through celite, concentrated, and purified by RPLC using an Isco CombiFlash liquid chromatograph eluted with 30% to 100% methanol and water, using no modifier. Yield 0.450 g, 64%. Ion(s) found by LCMS: [M/2]+H+ = 1064.6, [M-1(Boc)/2]+H+ = 1014.6. Step b. Synthesis of deca Boc PEG8 polymyxin B dimer

A solution of amino PEG8 penta Boc polymyxin B undecapeptide (0.746 g, 0.350 mmol), Cbz iminodiacetic acid(0.046 g, 0.171 mmol), and DIEA(0.197 mL, 1.13 mmol), in DMF (5 mL), was treated with a solution of HATU (0.133 g, 0.350 mmol) in DMF (2 mL), dropwise over 1h. After stirring an additional 30 minutes, the reaction was charged with 5% Pd/C (0.350 g), vacuum flushed with hydrogen, and stirred under a hydrogen atmosphere until complete by LCMS. The crude reaction was purified by RPLC using an Isco CombiFlash liquid chromatograph eluted with 30% to 100% methanol and water, using no modifier. Yield 0.465 g, 62% (two steps). Ion(s) found by LCMS: [M-3(Boc)/4]+H+ = 897.3, [M- 4(Boc)/4]+H+ = 872.3. Step c. Synthesis of deca Boc PEG 8 polymyxin B dimer bis su

A solution of deca Boc PEG 8 polymyxin B dimer(0.465 g, 0.107 mmol), bis sulfone acid(0.059 g, 0.117 mmol), and N-methylmorpholine (0.026 mL, 0.235 mmol), in DMF (5 mL) was treated with a solution of HATU (0.045 g, 0.117 mmol) and DMF (1 mL), dropwise over 1h. The resulting solution was stirred for an additional 30 minutes, then concentrated to a viscous oil. The crude oil was used in the next Step without further purification. Yield 0.465 g, 62% (two steps). Ion(s) found by LCMS: did not ionize. Step d. Synthesis of Int-10

A solution of crude deca Boc PEG 8 polymyxin B dimer bis sulfone, suspended in DCM (4 mL), was treated with TFA (4 mL), and stirred at room temperature for 5 min. The resulting solution was concentrated, and purified by RPLC using an Isco CombiFlash liquid chromatograph eluted with 5% to 100% methanol and water, using TFA as the modifier. Int-10 was obtained in a yield of 0.099 g, 27% (three steps). Ion(s) found by LCMS: [M/2]+H+ = 1917.5, [M/3]+H+ = 1278.7. E

A solution of Int-18 (0.500 g, 0.291 mmol), Cbz-amino PEG 8 diacid (0.089 g, 0.138 mmol), and DIEA (0.167 mL, 0.960 mmol), in DMF (4 mL), was treated with a solution of HATU (0.122 g, 0.320 mmol), in DMF (1 mL), dropwise over 1h. After stirring an additional 30 minutes, the reaction was charged with 5% Pd/C (0.5 g), vacuum flushed with hydrogen, and stirred under a hydrogen atmosphere until complete by LCMS. The crude mixture was purified by RPLC using an Isco CombiFlash liquid chromatograph eluted with 30% to 100% methanol and water, using no mo

steps). Ion(s) found by LCMS: [M-3(Boc)/4]+H+ = 897.3, [M-4(Boc)/4]+H+ = 872.3. Step b. Synthesis of bis sulfone deca Boc amino PEG 8 dimer

A solution of deca Boc amino PEG 8 dimer (0.212 g, 0.0545 mmol), bis-sulfone acid(0.033 g, 0.0654 mmol), and N-methylmorpholine(0.022 mL, 0.196 mmol), in DMF (3 mL), were treated with a solution of HATU (0.025 g, 0.065 mmol), in DMF (1.0 mL), dropwise over 1h. The resulting crude reaction mixture was concentrated to an oil and used in the next Step without purification. The product did not ionize by LCMS.

The crude oil was dissolved in DCM (5 mL), and treated with TFA (5 mL), for 5 minutes at room temperature. The resulting solution was concentrated and purified by RPLC using an Isco CombiFlash liquid chromatograph eluted with 10% to 100% methanol and water, using 0.1% TFA as the modifier. Yield 0.099 g, 40% (two steps). Ion(s) found by LCMS: [M/3]+H+ = 1123.3, [M/4]+H+ = 842.7. Example 14– Conjugate 1 (Conjugate of h-IgG1 Fc-2(Int-4))

A solution of h-IgG1 Fc (SEQ ID NO: 4) (4.66 mg in 175 µL of pH 7.4 PBS, MW=54,731 Da), Tris pH=8.0 (155 µL), and TCEP (68 µL of 10 mM stock in pH 8.0 Tris) were mixed at room temperature. LCMS after 1hr showed complete reduction. Int-4 was added as solution (153 µL of 8.04 mg dissolved in 250 µL of pH 8.0 Tris, 8 eq/disulfide). LCMS after 12h showed that all the reduced Fc was consumed, and a product peak with mass = 32,380 Da (DAR=2). The solution was dilu

and concentrated using a centrifugal concentrator (10,000 MWCO) to a volume of 1 mL, two times. The crude mixture was diluted 1:10 in PBS pH 7.4, and purified using MabSelect Sure Resin (GE Healthcare, Chicago, IL, USA), followed by size exclusion chromatography. Purified material was quantified using a Nanodrop™ UV visible spectrophotomer (using a calculated extinction coefficient based on the amino acid sequence of h-IgG1), and concentrated to approximately 10 mg/mL using a centrifugal concentrator (10,000 MWCO). Yield 0.001 g, 38% after purification. Product peak found by LCMS 32,380 Da

(DAR=2/Fc monomer, 4/Fc dimer). Final product: Conjugate 1. Example 15– Conjugate 2 (Conjugate of m-IgG2a Fc-3(Int-4))

A solution of m-IgG2a Fc (SEQ ID NO: 2) (4.5 mg in 315 µL pH 7.4 PBS, MW~54,000 Da), and TCEP (66 µL of 10mM stock in pH 8.0 Tris) were mixed at room temperature. LCMS after 1hr showed complete reduction. Int-4 was added as powder (7.9 mg, 8 eq/hinge disulfide). LCMS after 12h showed that all the reduced Fc was consumed, and a product peak with mass = 36,512 Da (DAR=3). The solution was diluted with 15 mL pH 7.4 PBS and ultrafiltered (10,000 MWCO) to a volume of 1 mL, two times. The crude mixture was diluted 1:10 in PBS pH 7.4, and purified using MabSelect Sure Resin (GE Healthcare, Chicago, IL, USA), followed by size exclusion chromatography. Purified material was quantified using a Nanodrop™ UV visible spectrophotomer (using a calculated extinction coefficient based on the amino acid sequence of m-IgG2a), and concentrated to approximately 10 mg/mL using a centrifugal concentrator (10,000 MWCO). Yield 0.004 g, 69% after purification. Product peak found by LCMS 36,512 Da (DAR=3/Fc monomer, 6/Fc dimer). Final product: Conjugate 2. Example 16– Conjugate 3 (Conjugate of m-IgG2a Fc-8(Int-5))

Peg4-azidoNHS ester (2.2 mg in 2.2 µL of pH 7.4 PBS 1x buffer solution) was added to a solution of m-IgG2a Fc (SEQ ID NO: 2) (28 mg in 300 µL pH 7.4 PBS, MW~54,000 Da) and the mixture was shaken gently for 12 hours at ambient temperature.

The crude mixture was diluted 1:10 in PBS pH 7.4, and purified using MabSelect Sure Resin (GE Healthcare, Chicago, IL, USA). Purified material was quantified using a Nanodrop™ UV visible spectrophotomer (using a calculated extinction coefficient based on the amino acid sequence of m- IgG2a), and concentrated to approximately 10 mg/mL using a centrifugal concentrator (10,000 MWCO). Int-5 (28 mg, 7.8 µmol) in a solution of 10 nM sodium ascorbate, 10 mM Copper (II) sulfate and 10 mM tris(3-hydroxylpropyltriazolylmethyl)amine in pH 7.4 PBS 1x buffer (3 mL) was added to the purified azido- m-IgG2a conjugate in pH 7.4 PBS 1x buffer and the mixture was shaken gently at ambient temperature for 24 hours. The crude mixture was diluted 1:10 in PBS pH 7.4, and purified using MabSelect Sure Resin (GE Healthcare, Chicago, IL, USA), followed by size exclusion chromatography. Purified material was quantified using a Nanodrop™ UV visible spectrophotomer (using a calculated extinction coefficient based on the amino acid sequence of m-IgG2a), and concentrated to approximately 10 mg/mL using a centrifugal concentrator (10,000 MWCO). Purified molecules were analyzed using 4-12% Bis Tris SDS G f gel included a molecular weight ladder with the indicated molecular weight

inducates non-reduced SDS-PAGE gel condition. Yield 0.016 g, 38% after purification. MALDI MS analysis showed a broad range peak (75,683– 87079) with an average of mass of 81,375 (DAR=4/Fc monomer, 8/Fc dimer). Final product: Conjugate 3. Example 17– Conjugate 4 (Conjugate of m-IgG2a(His)6 Fc-3(Int-9))

A solution of m-IgG2a(His)6 Fc (SEQ ID NO: 6) (6.15 mg in 0.5 mL pH 7.4 PBS, MW~59,880 Da), and TCEP (1.85 µL of 500mM stock in pH 7.4 PBS, 3eq/disulfide) were mixed at room temperature. LCMS after 1hr showed complete reduction. The solution was diluted with 15 mL pH 7.8 PBS with 2mM EDTA and ultrafiltered (10,000 MWCO) to a volume of 1 mL, two times. Int-9 was added as powder (2.11 mg, 1.5 eq/hinge disulfide). LCMS after 1h at room temperature showed all of the reduced Fc was consumed, and a broad peak with masses corresponding to DAR=2, 3, and 4. The solution was treated with DTT (0.142 mg, 9eq) and stored at 4C for 48h. LCMS showed DAR 3 prior to purification. The crude mixture was diluted 1:10 in PBS pH 7.4, and purified using MabSelect Sure Resin (GE Healthcare, Chicago, IL, USA), followed by size exclusion chromatography. Purified material was quantified using a Nanodrop™ UV visible spectrophotomer (using a calculated extinction coefficient based on the amino acid sequence of m-IgG2a(His) 6 ), and concentrated to approximately 10 mg/mL using a centrifugal concentrator (10,000 MWCO). Purified molecules were analyzed using 4-12% Bis Tris SDS PAGE gels by loading 1-2 µg of each molecule into the gel, and staining using instant Blue staining. Each gel included a molecular weight ladder with the indicated molecular weight standards (FIG.9).“NR” inducates non-reduced SDS-PAGE gel condition. Yield 0.006 g, 83% after purification. Product peak found by LCMS 69,262 Da (DAR=3/Fc dimer). Final product: Conjugate 4. Example 18– Conjugate 5 (Conjugate of h-IgG1 Fc-2(Int-9))

A solution of h-IgG1 Fc (SEQ ID NO: 2) (10.5 mg in 0.6 mL pH 7.4 PBS, MW~54,731 Da), and TCEP (4.6 µL of 500mM stock in pH 7.4 PBS, 6eq/disulfide) were mixed at room temperature. LCMS after 1hr showed complete reduction. The solution was diluted with 15 mL pH 7.8 PBS with 2mM EDTA and ultrafiltered (10,000 MWCO) to a volume of 1 mL, two times. Int-9 was added as powder (5.2 mg, 3.0 eq/hinge disulfide). LCMS after 1h at room temperature showed all of the reduced Fc was consumed, and a broad product peak with a mass = 61,306 Da(DAR=2). The crude mixture was diluted 1:10 in PBS pH 7.4, and purified using MabSelect Sure Resin (GE Healthcare, Chicago, IL, USA), followed by size exclusion chromatography. Purified material was quantified using a Nanodrop™ UV visible

spectrophotomer (using a calculated extinction coefficient based on the amino acid sequence of h-IgG1), and concentrated to approximately 10 mg/mL using a centrifugal concentrator (10,000 MWCO). Purified molecules were analyzed using 4-12% Bis Tris SDS PAGE gels by loading 1-2 µg of each molecule into the gel, and staining using instant Blue staining. Each gel included a molecular weight ladder with the indicated molecular weight standards (FIG.10).“NR” inducates non-reduced SDS-PAGE gel condition. Yield 0.010 g, 86% after purification. Product peak found by LCMS 61,306 Da (DAR=2/Fc dimer). Final C . Example 19– Conjugate 6 (Conjugate of m-IgG2a(His)6 Fc-2(Int-5))

Peg4-azido-NHS ester (4.4 mg in 2.2 mL of pH 7.4 PBS 1x buffer solution) was added to a solution of m-IgG2a(His) 6 Fc (SEQ ID NO: 6) (30 mg in 2.4 mL pH 7.4 PBS, MW~55,600 Da) and the mixture was shaken gently for 12 hours at ambient temperature. The crude mixture was diluted 1:10 in PBS pH 7.4, and purified using MabSelect Sure Resin (GE Healthcare, Chicago, IL, USA). Purified material was quantified using a Nanodrop™ UV visible spectrophotomer (using a calculated extinction coefficient based on the amino acid sequence of m-IgG2a(His) 6 ), and concentrated to approximately 10 mg/mL using a centrifugal concentrator (10,000 MWCO). Int-5 (28 mg, 7.8 µmol) in a solution of 10 nM sodium ascorbate, 10 mM Copper (II) sulfate and 10 mM tris(3-hydroxylpropyltriazolylmethyl)amine in pH 7.4 PBS 1x buffer (3 mL) was added to the purified azido-m-IgG2a conjugate in pH 7.4 PBS 1x buffer and the mixture was shaken gently at ambient temperature for 24 hours. The crude mixture was diluted 1:10 in PBS pH 7.4, and purified using MabSelect Sure Resin (GE Healthcare, Chicago, IL, USA), followed by size exclusion chromatography. Purified material was quantified using a Nanodrop™ UV visible spectrophotomer (using a calculated extinction coefficient based on the amino acid sequence of m-IgG2a(His) 6 ), and concentrated to approximately 10 mg/mL using a centrifugal concentrator (10,000 MWCO). Purified molecules were analyzed using 4-12% Bis Tris SDS PAGE gels by loading 1-2 µg of each molecule into the gel, and staining using instant Blue staining. Each gel included a molecular weight ladder with the indicated molecular weight standards (FIG.11).“NR” inducates non-reduced SDS-PAGE gel condition. Yield 0.018 g, 37% after purification. MALDI MS analysis showed a broad range peak (86310– 98106) with an average of mass of 89,434 (DAR=6/Fc monomer, 12/Fc dimer). Final product: Conjugate 6. Example 20– Conjugate 7 (Conjugate of h-IgG1(His) 6 Fc-2(Int-10))

A solution of h-IgG1(His)6 Fc (SEQ ID NO: 8) (10.0 mg in 1.0 mL pH 7.4 PBS, MW~56,633 Da), and TCEP (6.3 µL of 500mM stock in pH 7.4 PBS, 9eq/disulfide) were mixed at room temperature. LCMS after 1hr showed complete reduction. The solution was diluted with 15 mL pH 7.8 PBS with 2mM EDTA and ultrafiltered (10,000 MWCO) to a volume of 1 mL, two times. Int-10 was added as powder (5.3 mg, 3.0 eq/hinge disulfide). LCMS after 2h at room temperature showed all of the reduced Fc was consumed, and a broad product peak, mass = 63,682 Da(DAR=2). The reaction was treated with DTT(0.272 mg, 10eq) and stored at 4C. The crude mixture was diluted 1:10 in PBS pH 7.4, and purified using MabSelect Sure Resin (GE Healthcare, Chicago, IL, USA), followed by size exclusion chromatography. Purified material was quantified using a Nanodrop™ UV visible spectrophotomer (using a calculated extinction coefficient based on the amino acid sequence of h-IgG1(His)6), and concentrated to approximately 10 mg/mL using a centrifugal concentrator (10,000 MWCO). Purified molecules were analyzed using 4-12% Bis Tris SDS PAGE gels by loading 1-2 µg of each molecule into the gel, and staining using instant Blue staining. Each gel included a molecular weight ladder with the indicated molecular weight standards (FIG. 12).“NR” inducates non-reduced SDS-PAGE gel condition. Yield 0.003 g,

Product peak found by LCMS 63,682 Da (DAR=2/Fc dimer). Final product: Conjugate 7. Example 21– Conjugate 8 (Conjugate of h-IgG1(His) 6 Fc-2(Int-7))

D BCO-peg4-NHS ester (14.4 mg in 2.5 mL of pH 7.4 PBS 1x buffer solution) was added to a solution of h-IgG1(His)6 Fc (SEQ ID NO: 8) (30 mg in 3 mL pH 7.4 PBS, MW~56,633 Da) and the mixture was shaken gently for 12 hours at ambient temperature. Some precipitate was observed during at the end of this time period. The crude mixture was diluted 1:10 in PBS pH 7.4, and purified using MabSelect Sure Resin (GE Healthcare, Chicago, IL, USA). Purified material was quantified using a Nanodrop™ UV visible spectrophotomer (using a calculated extinction coefficient based on the amino acid sequence of h- IgG1(His)6), and concentrated to approximately 10 mg/mL using a centrifugal concentrator (10,000 MWCO). Int-7 (28 mg, 7.1 µmol) in a solution of in pH 7.4 PBS 1x buffer (2 mL) was added to the purified azido- h-IgG1 conjugate (4.97 mg) in pH 7.4 PBS 1x buffer (3 mL). Acetonitrile (2 mL) was added when solution began to turn cloudy. The mixture was shaken gently at ambient temperature for 24 hours. The crude mixture was diluted 1:10 in PBS pH 7.4, and purified using MabSelect Sure Resin (GE Healthcare, Chicago, IL, USA), followed by size exclusion chromatography. Purified material was quantified using a Nanodrop™ UV visible spectrophotomer (using a calculated extinction coefficient based on the amino acid sequence of h-IgG1(His)6), and concentrated to approximately 10 mg/mL using a centrifugal concentrator (10,000 MWCO). Purified molecules were analyzed using 4-12% Bis Tris SDS PAGE gels by loading 1-2 µg of each molecule into the gel, and staining using instant Blue staining. Each gel included a molecular weight ladder with the indicated molecular weight standards (FIG.13).“NR” inducates non-reduced SDS-PAGE gel condition. Yield 0.005 g, 14% after purification. MALDI MS analysis showed a broad range peak (60893– 79801) with an average of mass of 68,815 (DAR=2/Fc monomer, 4/Fc dimer). Final product: Conjugate 8. Example 22– Conjugate 9 (Conjugate of h-IgG1(His)6 Fc-4(Int-10))

A solution of engineered h-IgG1(His)6 Fc (SEQ ID NO: 10) (10.0 mg in 0.84 mL pH 7.4 PBS, MW~57,676 Da), and TCEP (12.5 µL of 500mM stock in pH 7.4 PBS, 9eq/disulfide) were mixed at room temperature. LCMS after 1hr showed complete reduction. The solution was diluted with 15 mL pH 7.8 PBS with 2mM EDTA and ultrafiltered (10,000 MWCO) to a volume of 1 mL, two times. Int-10 was added as powder (6.87 mg, 2.0 eq/hinge disulfide). LCMS after 24h at room temperature showed all the reduced Fc was consumed, and a broad product peak with masses = 64,726 Da(DAR=2), 71,915 Da(DAR=4), and 78,818 Da (DAR=6). The crude mixture was diluted 1:10 in PBS pH 7.4, and purified using MabSelect Sure Resin (GE Healthcare, Chicago, IL, USA), followed by size exclusion

chromatography. Purified material was quantified using a Nanodrop™ UV visible spectrophotomer (using a calculated extinction coefficient based on the amino acid sequence of h-IgG1(His)6), and concentrated to approximately 10 mg/mL using a centrifugal concentrator (10,000 MWCO). Purified molecules were analyzed using 4-12% Bis Tris SDS PAGE gels by loading 1-2 µg of each molecule into the gel, and molecular weight standards (FIG.14).“NR” inducates non-reduced SDS-P

0.003 g, 24% after purification. Product peak (major) found by LCMS 71915 Da (DAR=4/engineered Fc dimer). Final product: Conjugate 9. Example 23– Conjugate 10 (Conjugate of h-IgG1(His) 6 Fc-4(Int-9))

A solution of h-IgG1(His)6 Fc (SEQ ID NO: 10) (10.0 mg in 0.84 mL pH 7.4 PBS, MW~57,676 Da), and TCEP (12.5 µL of 500mM stock in pH 7.4 PBS, 9eq/disulfide) were mixed at room temperature. LCMS after 1hr showed complete reduction. The solution was diluted with 15 mL pH 7.8 PBS with 2mM EDTA and ultrafiltered (10,000 MWCO) to a volume of 1 mL, two times. Int-9 was added as powder (6.87 mg, 2.0 eq/hinge disulfide). LCMS after 1h at room temperature showed all of the reduced Fc was consumed, and a broad product peak with a mass = 70,223 Da(DAR=4). The crude mixture was diluted 1:10 in PBS pH 7.4, and purified using MabSelect Sure Resin (GE Healthcare, Chicago, IL, USA), followed by size exclusion chromatography. Purified material was quantified using a Nanodrop™ UV visible spectrophotomer (using a calculated extinction coefficient based on the amino acid sequence of h- IgG1(His)6), and concentrated to approximately 10 mg/mL using a centrifugal concentrator (10,000 MWCO). Purified molecules were analyzed using 4-12% Bis Tris SDS PAGE gels by loading 1-2 µg of each molecule into the gel, and staining using instant Blue staining. Each gel included a molecular weight ladder with the indicated molecular weight standards (FIG.15).“NR” inducates non-reduced SDS-PAGE gel condition. Yield 0.010 g, 82% after purification. Product peak (major) found by LCMS 70,223 Da (DAR=4/engineered Fc dimer). Final product: Conjugate 10. Example 24– Conjugate 11 (Conjugate of h-IgG1(His)6 Fc-4(Int-11))

A solution of h-IgG1(His)6 Fc (SEQ ID NO: 10) (10.0 mg in 0.84 mL pH 7.4 PBS, MW~57,676 Da), and TCEP (12.5 µL of 500mM stock in pH 7.4 PBS, 9eq/disulfide) were mixed at room temperature. LCMS after 1hr showed complete reduction. The solution was diluted with 15 mL pH 7.8 PBS with 2mM EDTA and ultrafiltered (10,000 MWCO) to a volume of 1 mL, two times. Int-11 was added as powder (6.31 mg, 2.0 eq/hinge disulfide). LCMS after 1h at room temperature showed all of the reduced Fc was consumed, and a broad product peak with a mass = 69,913 Da(DAR=4). The crude mixture was diluted 1:10 in PBS pH 7.4, and purified using MabSelect Sure Resin (GE Healthcare, Chicago, IL, USA), followed by size exclusion chromatography. Purified material was quantified using a Nanodrop™ UV visible spectrophotomer (using a calculated extinction coefficient based on the amino acid sequence of h- IgG1(His)6), and concentrated to approximately 10 mg/mL using a centrifugal concentrator (10,000 MWCO). Purified molecules were analyzed using 4-12% Bis Tris SDS PAGE gels by loading 1-2 µg of each molecule into the gel, and staining using instant Blue staining. Each gel included a molecular weight ladder with the indicated molecular weight standards (FIG.16).“NR” inducates non-reduced SDS-PAGE gel condition. Yield 0.010 g, 82% after purification. Product peak (major) found by LCMS 69,913 Da (DAR=4/engineered Fc dimer). Final product: Conjugate 11. Example 25– Synthesis of Int-12 (maleimide-PEG 8-amino-polymyxin

Step a. Synthesis of penta Boc polymyxin B decapeptide (Int-7a)

Tri-Boc polymyxin B heptapeptide (0.464 g, 0.437 mmol, Int-1), and tripeptide acid (0.316 g, 0.398 mmol, Int-3), were dissolved in DMF (1 mL), DIEA (0.229 mL, 1.31 mmol), then treated with HATU (0.166 g, 0.437 mmol). After stirring for 30 min LCMS showed complete conversion. The crude Cbz- product was diluted with methanol (4 mL), and charged with 5%Pd/C (0.250 g) and placed under a hydrogen atmosphere. After 2 hr the mixture was filtered through Celite, concentrated, and purified by RPLC using an Isco CombiFlash liquid chromatograph eluted with 10% to 100% methanol and water, using no modifier. Yield of Int-7a was 0.640 g, 94% (two steps). Ion(s) found by LCMS: [(M-1Boc)/2]+H+ = 1054.6, [(M-2Boc)/2]+H+ = 1004.6.

Step b. Synthesis of aminooctanoyl penta Boc polymyxin B de

A solution of Int-7a (3.00 g, 1.918 mmol), Cbz-aminooctanoic acid (0.591 g, 2.01 mmol), and DIEA (1.05 mL, 6.04 mmol), in DMF (10 mL), was treated with a solution of HATU (0.766 g, 2.01 mmol) dissolved in DMF (3 mL), dropwise over 60 minutes. After 2h, LCMS showed complete consumption of starting material. The crude mixture was treated with 5% Pd/C (1.7 g), vacuum flushed with hydrogen, and stirred under a hydrogen atmosphere for 2hr or until complete by LCMS. The crude reaction was filtered through Celite, concentrated, and purified by RPLC using an Isco CombiFlash liquid

chromatograph eluted with 20% to 100% methanol and water, using no modifier. Yield of Int-18 was 2.59 g, 79% (two steps). Ion(s) found by LCMS: [M/2]+H+ = 853.4. Step c. Synthesis of maleimide penta Boc polymyxin B undecapeptide

A solution of aminooctanoyl penta Boc polymyxin B undecapeptide (0.5 g, 0.293 mmol), maleimide-acid (0.168 g, 0.323 mmol), DIEA (0.153 mL, 0.880 mmol), in DMF (3 mL), was treated with a solution of HATU (0.123 mg, 0.323 mmol) in DMF (1 mL) dropwise over 60 minutes. The resulting mixture was purified by RPLC using an Isco CombiFlash liquid chromatograph eluted with 20% to 100% methanol and water, using no modifier. Yield 0.595 g, 92%. Ion(s) found by LCMS: [M-1(Boc)/2]+H+ = 1054.6, [M-2(Boc)/2]+H+ =1004.6

A solution of maleimide penta Boc polymyxin B undecapeptide (0.595 g, 0.269 mmol), dissolved in DCM (2 mL), was treated with TFA (2 mL) while stirring for 5 minutes at room temperature. The resulting solution was concentrated and purified by RPLC using an Isco CombiFlash liquid

chromatograph eluted with 5% to 100% methanol and water, using TFA (0.1%) as the modifier. Yield 0.560 g, 73%. Ion(s) found by LCMS: [M/3]+H+ = 570.0, [M/4]+H+ = 427.7. Exa

A solution of aminooctanoyl penta Boc polymyxin B decapeptide (0.600 g, 0.352 mmol, Int-18), Cbz-amino Peg 8 acid (0.223 g, 0.387 mmol), and DIEA (0.202 mL, 1.161 mmol), in DMF (3 mL) was treated with a solution of HATU (0.147 g, 0.387 mmol), in DMF (1 mL), dropwise over 1hr. After stirring for an additional 30 minutes, the reaction was charged with 5% Pd/C (0.400 g), vacuum flushed with hydrogen, and stirred under a hydrogen atmosphere until complete by LCMS (2h). The resulting mixture was filtered through celite, concentrated, and purified by RPLC using an Isco CombiFlash liquid chromatograph eluted with 30% to 100% methanol and water, using no modifier. Yield 0.450 g, 64%. Ion(s) found by LCMS: [M/2]+H+ = 1064.6, [M-1(Boc)/2]+H+ = 1014.6. Step b. Synthesis of PEG 8 penta Boc polymyxin B undecapeptide

A solution of amino Peg 8 penta Boc polymyxin B undecapeptide (0.450 g, 0.211 mmol), bissulfone acid (0.116 g, 0.233 mmol), and N-methylmorpholine(0.051 mL, 0.465 mmol) in DMF (4 mL), was treated with a solution of HATU (0.0884 g, 0.232 mmol), in DMF (1.0 mL), dropwise over 1hr. After stirring for an additional 30 minutes the reaction was purified by RPLC using an Isco CombiFlash liquid chromatograph eluted with 30% to 100% methanol and water, using no mo

Ion(s) found by LCMS: No ions observed.

A solution of Peg 8 penta Boc polymyxin B undecapeptide(0.475 mg, 0.182 mmol), suspended in DCM (3 mL) was treated with TFA (3 mL) for 5 minutes at room temperature, then concentrated to an oil. The crude oil was purified by RPLC using an Isco CombiFlash liquid chromatograph eluted with 5% to 100% methanol and water, using TFA (0.1%) as the modifier. Yield 0.475 g, 97%. Ion(s) found by LCMS: [M/3]+H+ = 704.0, [M/4]+H+ = 528.3.

The title compound was prepared analogously to Int-13, where Cbz-amino PEG 4 acid was used in place of Cbz-amino Peg 8 acid in the first Step of that sequence. Ion(s) found by LCMS: [M/2]+H+ = 967.5, [M/3]+H+ = 645.3. Example 28– Conjugate 12 (Conjugate of m-IgG2a Fc-3(Int-12))

A solution of m-IgG2a Fc (SEQ ID NO: 2) (10 mg in 169 µL pH 7.4 PBS, MW~54,000 Da), and TCEP (2.8 µL of 50mM stock in pH 8.0 Tris) were mixed at room temperature. LCMS after 1hr showed complete reduction. Int-12 was added as powder (19.0 mg, 6 eq/hinge disulfide). LCMS after 48h at 4C showed that all the reduced Fc was consumed. The solution was diluted with 15 mL pH 7.4 PBS and ultrafiltered (10,000 MWCO) to a volume of 1 mL, two times. The crude mixture was diluted 1:10 in PBS pH 7.4, and purified using MabSelect Sure Resin (GE Healthcare, Chicago, IL, USA), followed by size exclusion chromatography. Purified material was quantified using a Nanodrop™ UV visible

spectrophotomer (using a calculated extinction coefficient based on the amino acid sequence of m- IgG2a), and concentrated to approximately 10 mg/mL using a centrifugal concentrator (10,000 MWCO). Purified molecules were analyzed using 4-12% Bis Tris SDS PAGE gels by loading 1-2 µg of each molecule into the gel, and staining using instant Blue staining. Each gel included a molecular weight ladder with the indicated molecular weight standards (FIG.17).“R” inducates reduced SDS-PAGE gel condition. Yield 0.008 g, 70% after purification. Product peak found by LCMS 31,272 Da (DAR=3/Fc monomer, 6/Fc dimer). Final product: Conjugate 12. Example 29– Conjugate 13 (Conjugate of h-IgG1 Fc-2(Int-13))

A solution of h-IgG1 Fc (SEQ ID NO: 4) (25 mg in 1 mL pH 7.4 PBS, MW=54,731 Da), and TCEP (9.1 µL of 50mM stock in pH 7.4 PBS, 5eq/disulfide) were mixed at room temperature. LCMS after 1hr showed complete reduction. The solution was diluted with 15 mL pH 7.8 PBS w/ 2mM EDTA and ultrafiltered (10,000 MWCO) to a volume of 1 mL, two times. Int-13 was added as powder (14.7 mg, 6 eq/hinge disulfide). LCMS after 12h at room temperature showed all the reduced Fc was consumed, and a product peak with mass = 58,329 Da(DAR=2). The solution was diluted with 15 mL pH 7.4 PBS and ultrafiltered (10,000 MWCO) to a volume of 1 mL, two times. The crude mixture was diluted 1:10 in PBS pH 7.4, and purified using MabSelect Sure Resin (GE Healthcare, Chicago, IL, USA), followed by size exclusion chromatography. Purified material was quantified using a Nanodrop™ UV visible

spectrophotomer (using a calculated extinction coefficient based on the amino acid sequence of h-IgG1), and concentrated to approximately 10 mg/mL using a centrifugal concentrator (10,000 MWCO). Purified molecules were analyzed using 4-12% Bis Tris SDS PAGE gels by loading 1-2 µg of each molecule into the gel, and staining using instant Blue staining. Each gel included a molecular weight ladder with the indicated molecular weight standards (FIG.18). Yield 0.022 g, 83% after purification. Product peak found by LCMS 58,329 Da (DAR=2/Fc dimer). Final product: Conjugate 13. Example 30– Conjugate 14 (Conjugate of h-IgG1 Fc-2(Int-14))

A solution of h-IgG1 Fc (SEQ ID NO: 4) (25 mg in 1 mL pH 7.4 PBS, MW=54,731 Da), and TCEP (3.6 µL of 500mM stock in pH 7.4 PBS, 2eq/disulfide) were mixed at room temperature. LCMS after 1hr showed complete reduction. The solution was diluted with 15 mL pH 7.8 PBS w/ 2mM EDTA and ultrafiltered (10,000 MWCO) to a volume of 1 mL, two times. Int-14 was added as powder (13.7 mg, 6 / f ) C S f a broad product peak with mass = 57,977 Da(DAR=2). The solution was di

and ultrafiltered (10,000 MWCO) to a volume of 1 mL, two times. The crude mixture was diluted 1:10 in PBS pH 7.4, and purified using MabSelect Sure Resin (GE Healthcare, Chicago, IL, USA), followed by size exclusion chromatography. Purified material was quantified using a Nanodrop™ UV visible spectrophotomer (using a calculated extinction coefficient based on the amino acid sequence of h-IgG1), and concentrated to approximately 10 mg/mL using a centrifugal concentrator (10,000 MWCO). Purified molecules were analyzed using 4-12% Bis Tris SDS PAGE gels by loading 1-2 µg of each molecule into the gel, and staining using instant Blue staining. Each gel included a molecular weight ladder with the indicated molecular weight standards (FIG.19). Yield 0.021 g, 78% after purification. Product peak found by LCMS 57,977 Da (DAR=2/Fc dimer). Final product: Conjugate 14. Example 31– Conjugate 15 (Conjugate of m-IgG2a(His) 6 Fc-3(Int-13))

A solution of m-IgG2a(His) 6 Fc (SEQ ID NO: 6) (12.8 mg in 1 mL pH 7.4 PBS, MW~59,880 Da), and TCEP (3.8 µL of 500mM stock in pH 7.4 PBS, 3eq/disulfide) were mixed at room temperature. LCMS after 1hr showed complete reduction. The solution was diluted with 15 mL pH 7.8 PBS w/ 2mM EDTA and ultrafiltered (10,000 MWCO) to a volume of 1 mL, two times. Int-13 was added as powder (3.4 mg, 2 eq/hinge disulfide). LCMS after 1h at room temperature showed all of the reduced Fc was consumed, and a product peak with mass = 65,438 Da(DAR=3). The solution was diluted with 15 mL pH 7.4 PBS and ultrafiltered (10,000 MWCO) to a volume of 1 mL, two times. The crude mixture was diluted 1:10 in PBS pH 7.4, and purified using MabSelect Sure Resin (GE Healthcare, Chicago, IL, USA), followed by size exclusion chromatography. Purified material was quantified using a Nanodrop™ UV visible spectrophotomer (using a calculated extinction coefficient based on the amino acid sequence of m- IgG2a(His)6), and concentrated to approximately 10 mg/mL using a centrifugal concentrator (10,000 MWCO). Purified molecules were analyzed using 4-12% Bis Tris SDS PAGE gels by loading 1-2 µg of each molecule into the gel, and staining using instant Blue staining. Each gel included a molecular weight ladder with the indicated molecular weight standards (FIG.20). Yield 0.013 g, 94% after purification. Product peak found by LCMS 65,438 Da (DAR=3/Fc dimer). Final product: Conjugate 15. Example 32– Conjugate 16 (Conjugate of h-IgG1(His) 6 Fc-2(Int-5))

Peg4-azido-NHS ester (38.4 mg in 150 uL of DMF) was added to a solution of h-IgG1(His)6 Fc (SEQ ID NO: 8 (140 mg in 15 mL pH 7.4 PBS 1x buffer, MW~55,111 Da) and the mixture was shaken gently for 12 hours at ambient temperature.

The crude mixture was diluted 1:10 in PBS pH 7.4, and purified using MabSelect Sure Resin (GE Healthcare, Chicago, IL, USA). Purified material was quantified using a Nanodrop™ UV visible spectrophotomer (using a calculated extinction coefficient based on the amino acid sequence of h- IgG1(His)6), and concentrated to approximately 10 mg/mL using a centrifugal concentrator (10,000 MWCO). Int-5 (150 mg, 41.9 µmol) was added to the purified azido-h-IgG1 conjugate (56.9 mg in 7 mL of pH 7.4 PBS 1x buffer, MW~58703 Da by MALDI) followed by 2 mL of a solution of 10 mM sodium ascorbate, 10 mM Copper (II) sulfate and 10 mM tris(3-hydroxylpropyltriazo

1x buffer and the mixture was shaken gently at ambient temperature for 4 hours at which point an additional 0.6 mL of the solution of 10 mM sodium ascorbate, 10 mM Copper (II) sulfate and 10 mM tris(3- hydroxylpropyltriazolylmethyl)amine in pH 7.4 PBS 1x buffer was added and the mixture was shaken gently for 12 hours. The crude mixture was diluted 1:10 in PBS pH 7.4, and purified using MabSelect Sure Resin (GE Healthcare, Chicago, IL, USA), followed by size exclusion chromatography. Purified material was quantified using a Nanodrop™ UV visible spectrophotomer (using a calculated extinction coefficient based on the amino acid sequence of h-IgG1(His)6), and concentrated to approximately 10 mg/mL using a centrifugal concentrator (10,000 MWCO). Purified molecules were analyzed using 4-12% Bis Tris SDS PAGE gels by loading 1-2 µg of each molecule into the gel, and staining using instant Blue staining. Each gel included a molecular weight ladder with the indicated molecular weight standards (FIG. 21).“NR” inducates non-reduced SDS-PAGE gel condition. Yield 0.057 g, 78% after purification. MALDI MS analysis showed a broad range peak (60893– 79801) with an average of mass of 80443 (DAR=4/Fc monomer, 8/Fc dimer). Final product: Conjugate 16. Example 33– Conjugate 17 (Conjugate of aglycosylated h-IgG1(His) 6 Fc-2(Int-5))

Peg4-azido-NHS ester (20.4 mg in 82 uL of DMF) was added to a solution of aglycosylated h- IgG1(His)6 Fc (SEQ ID NO: 14) (74 mg in 8 mL pH 7.4 PBS 1x buffer, MW~53,637 Da) and the mixture was shaken gently for 12 hours at ambient temperature. The crude mixture was diluted 1:10 in PBS pH 7.4, and purified using MabSelect Sure Resin (GE Healthcare, Chicago, IL, USA). Purified material was quantified using a Nanodrop™ UV visible spectrophotomer (using a calculated extinction coefficient based on the amino acid sequence of h-IgG1(His)6), and concentrated to approximately 10 mg/mL using a centrifugal concentrator (10,000 MWCO). Int-5 (150 mg, 41.9 µmol) was added to the purified azido-h- IgG2a conjugate(azido-CTP-125) (56.9 mg in 7.5 mL of pH 7.4 PBS 1x buffer, MW~57,118 Da by MALDI) followed by 2 mL of a solution of 10 mM sodium ascorbate, 10 mM Copper (II) sulfate and 10 mM tris(3-hydroxylpropyltriazolylmethyl)amine in pH 7.4 PBS 1x buffer and the mixture was shaken gently at ambient temperature for 4 hours at which point an additional 0.6 mL of the solution of 10 mM sodium ascorbate, 10 mM Copper (II) sulfate and 10 mM tris(3-hydroxylpropyltriazolylmethyl)amine in pH 7.4 PBS 1x buffer was added and the mixture was shaken gently for an additional 12 hours.The crude mixture was diluted 1:10 in PBS pH 7.4, and purified using MabSelect Sure Resin (GE Healthcare, Chicago, IL, USA), followed by size exclusion chromatography. Purified material was quantified using a Nanodrop™ UV visible spectrophotomer (using a calculated extinction coefficient based on the amino acid sequence of h-IgG1(His) 6 ), and concentrated to approximately 10 mg/mL using a centrifugal concentrator (10,000 MWCO). Purified molecules were analyzed using 4-12% Bis Tris SDS PAGE gels by loading 1-2 µg of each molecule into the gel, and staining using instant Blue staining. Each gel included a molecular weight ladder with the indicated molecular weight standards (FIG.22).“NR” inducates non-reduced SDS-PAGE gel condition. Yield 0.055 g, 50% after purification. MALDI MS analysis showed a broad range peak (60893– 79801) with an average of mass of 80899 (DAR=4.5/Fc monomer, 9/Fc dimer). Final product: C Step a.

A solution of Cbz-iminodiacetic acid (0.149 g, 0.557 mmol), Int-18 (2.00 g, 1.173 mmol, described in Example 10 Step b.), and DIEA (0.644 mL, 3.69 mmol) in DMF, was treated with a solution of HATU (0.468 g, 1.231 mmol) in DMF (5 mL), dropwise over 1h. After stirring for an additional 30 minutes the reaction was charged with 5% Pd/C (1.0 g), vacuum flushed with hydrogen, and stirred under a hydrogen atmosphere for 2h. The resulting mixture was filtered through celite, concentrated, and purified by RPLC using an Isco CombiFlash liquid chromatograph eluted with 30% to 100% methanol and water, using no modifier. Yield 1.0 g, 48% yield (two steps). Step b.

Product from Step a (1.03 g, 0.294 mmol), Cbz-N-amido-PEG4-acid (0.117 g, 0.294 mmol) and DIEA (0.110 mL, 0.617 mmol) in DMF (15 mL), was treated HATU (0.117 g, 0.308 mmol). After stirring for an additional 30 minutes the reaction was charged with 5% Pd/C (1.0 g), vacuum flushed with hydrogen, and stirred under a hydrogen atmosphere for 2 h. The resulting mixture was filtered through celite, concentrated, and purified by RPLC using an Isco CombiFlash liquid chromatograph eluted with 30% to 100% methanol and water, using no modifier. Yield 0.450 g, 41% yield (two steps). Step c.

Product from Step b (0.450 g, 0.114 mmol), 4-(3-tosyl-2-(tosylmethyl)propanoyl)benzoic acid (0.023 g, 0.229 mmol) in DMF (10 mL), was treated HATU (0.043 g, 0.114 mmol). After stirring for 30 minutes the reaction was purified by RPLC using an Isco CombiFlash liquid chromatograph eluted with 30% to 100% methanol and water, using no modifier. Yield 0.158 g, 31% yield. Step d.

P roduct from Step c (0.158 g, 0.037 mmol) was treated with TFA (5 mL) for 5 minutes then concentrated to an oil. The oil was purified by RPLC using an Isco CombiFlash liquid chromatograph eluted with 10% to 100% methanol and water, using 0.1% TFA as the modifier. Yield 0.47 g, 27% yield. Example 35– Conjugate 18 (Conjugate of h-IgG1(His) 6 Fc-2(Int-15))

A solution of h-IgG1(His)6 Fc (SEQ ID NO: 8) (857 µL, 10.20 mg, 0.1757 µmol, 11.90 mg/mL, MW 58046 D ) i H 74 PBS t t d ith t l ti f TCEP (126 L 6324 l 500 M) for 1 h, when Q-TOF analysis returned MW=28,773 Da. Excess TCEP was

with a 10,000 MWCO centrifugal filter (2 x 15 mL pH 7.4 PBS). The resulting solution was treated with Int-15 (6.15 mg, 1.405 µmol, 8 eq), then agitated gently for 1.5 h, when Q-TOF analysis showed full disappearance of the starting protein. Freshly prepared DTT water solution (54 µL, 3.513 µmol, 1 mg/100 µL) was added. The crude mixture was diluted 1:10 in PBS pH 7.4, and purified using MabSelect Sure Resin (GE Healthcare, Chicago, IL, USA), followed by size exclusion chromatography. Purified material was quantified using a Nanodrop™ UV visible spectrophotomer (using a calculated extinction coefficient based on the amino acid sequence of h-IgG1), and concentrated to approximately 10 mg/mL using a centrifugal concentrator (10,000 MWCO). Purified molecules were analyzed using 4-12% Bis Tris SDS PAGE gels by loading 1-2 µg of each molecule into the gel, and staining using instant Blue staining. Each gel included a molecular weight ladder with the indicated molecular weight standards (FIG.23).“NR” inducates non-reduced SDS-PAGE gel condition. Q-TOF analysis of the purified final product gave an average mass of 34,720 Da (DAR = 4). Yield = 9.38 mg, 77% yield. Example 36– Conjugate 19 (Conjugate of m-IgG2a(His)6 Fc-3(Int-15))

A solution of mIgG2a(His)6 Fc (SEQ ID NO: 6) (900 µL, 8.69 mg, 0.1408 µmol, 9.658 mg/mL, MW=61,739 Da) in pH 7.4 PBS was treated with a water solution of TCEP (7.6 µL, 3.802 µmol, 500 mM) for 1 h, when Q-TOF analysis returned MW=29,998 Da. Excess of TCEP was removed by buffer exchange with a 10,000 MWCO centrifugal filter (2 x 15 mL pH 7.4 PBS). The resulting solution was treated with Int-15 (3.70 mg, 0.8447 µmol, 6 eq), then agitated gently for 1.5 h, when Q-TOF analysis showed full disappearance of the starting protein. Freshly prepared DTT water solution (43 µL, 2.816 µmol, 1 mg/100 µL) was added. The crude mixture was diluted 1:10 in PBS pH 7.4, and purified using MabSelect Sure Resin (GE Healthcare, Chicago, IL, USA), followed by size exclusion chromatography. Purified material was quantified using a Nanodrop™ UV visible spectrophotomer (using a calculated extinction coefficient based on the amino acid sequence of m-IgG2a), and concentrated to approximately 10 mg/mL using a centrifugal concentrator (10,000 MWCO). Purified molecules were analyzed using 4- 12% Bis Tris SDS PAGE gels by loading 1-2 µg of each molecule into the gel, and staining using instant Blue staining. Each gel included a molecular weight ladder with the indicated molecular weight standards (FIG.24). Q-TOF analysis of the purified final product gave an average mass of 36,013 Da (DAR = 4). Yield = 8.69 mg, 86% yield. Example 37– Synthesis of Int-16

Ste a.

A solution of Cbz-iminodiacetic acid (0.149 g, 0.557 mmol), Int-18 (2.00 g, 1.173 mmol, described in Example 10 Step b.), and DIEA (0.644 mL, 3.69 mmol) in DMF, was treated with a solution of HATU (0.468 g, 1.231 mmol) in DMF (5 mL), dropwise over 1h. After stirring for a

reaction was charged with 5% Pd/C (1.0 g), vacuum flushed with hydrogen, and stirred under a hydrogen atmosphere for 2h. The resulting mixture was filtered through celite, concentrated, and purified by RPLC using an Isco CombiFlash liquid chromatograph eluted with 30% to 100% methanol and water, using no modifier. Yield 1.0 g, 48% yield (two steps). Ste b.

Product from Step a (0.800 g, 0.228 mmol), alkyne-peg4-acid (0.0833 g, 0.274 mmol) and DIEA (0.131 mL, 0.753 mmol) in DMF (5 mL), was treated HATU (0.0954 g, 0.251 mmol). After stirring for 30 minutes the reaction was purified by RPLC using an Isco CombiFlash liquid chromatograph eluted with 30% to 100% methanol and water, using no modifier. Yield 0.859 g, 99% yield. Ste c.

Product from Step b (0.859 g, 0.229 mmol) was treated with TFA (5 mL) for 5 minutes then concentrated to an oil. The oil was purified by RPLC using an Isco CombiFlash liquid chromatograph eluted with 10% to 100% methanol and water, using 0.1% TFA as the modifier. Yield 0.863 g, 97% yield. Example 38– Synthesis of Int-17

A solution of Int-18 (0.800 g, 0.469 mmol, described in Example 10 Step b.), alkyne-peg4 acid(0.134 g, 0.516 mmol), and DIEA (0.294 mL, 1.69 mmol) in DMF (3 mL), was treated with HATU (0.214 g, 0.563 mmol). After stirring for 30 minutes the reaction was purified by RPLC using an Isco C ombiFlash liquid chromatograph eluted with 30% to 100% methanol and water, using no modifier. Yield 0.83 g, 91% yield.

A solution of penta Boc product from the previous Step (0.830 g, 0.426 mmol), was treated with TFA (5 mL). After stirring for 5 minutes the reaction was concentrated and purified by RPLC using an Isco CombiFlash liquid chromatograph eluted with 10% to 100% methanol and water, using TFA as the modifier. Yield 0.689 g, 80% yield. Example 39– Conjugate 20 (Conjugate of m-IgG2a(His) 6 Fc-3(Int-16))

A solution of mIgG2a(His)6 Fc (SEQ ID NO: 6) (42.6 mg, 0.0707 µmol, 10.65 mg/mL,

MW=60,252da) in pH7.4 PBS was treated with azido-peg4-NHS ester (32.9 mg, 84.8 µmol, 120 eq), then agitated gently overnight at RT. Unreacted azido-peg4-NHS ester was removed by buffer exchange with a 10,000 MWCO centrifugal filter (6x15 mL pH 7.4 PBS). The final volume of the azide functionalized Fc was 3.0 mL. Maldi TOF analysis gave a molecular weight of 64,396 (azide DAR = 15.1). To this was added freshly prepared solutions of CuSO4 (300 µL of 20 mM), THPTA (300 µL of 50 mM, Tris(3- hydroxypropyltriazolylmethyl)amine), ascorbic acid (300 µL of 100 mM), and alkyne derivatized small molecule (109.9 mg, 28.3 µmol, Int-16)). The crude mixture was diluted 1:10 in PBS pH 7.4, and purified using MabSelect Sure Resin (GE Healthcare, Chicago, IL, USA), followed by size exclusion

chromatography. Purified material was quantified using a Nanodrop™ UV visible spectrophotomer (using a calculated extinction coefficient based on the amino acid sequence of m-IgG2a), and concentrated to approximately 10 mg/mL using a centrifugal concentrator (10,000 MWCO). Purified molecules were analyzed using 4-12% Bis Tris SDS PAGE gels by loading 1-2 µg of each molecule into the gel, and staining using instant Blue staining. Each gel included a molecular weight ladder with the indicated molecular weight standards (FIG.25). Maldi TOF analysis of the purified final product gave an average mass of 86,577 Da (DAR = 10). Yield = 72 mg, 70% yield. Example 40– Conjugate 21 (Conjugate of h-IgG1(His)6 Fc-2(Int-17))

A solution of hIgG1(His)6 Fc (SEQ ID NO: 8) (67.2 mg, 1.19 µmol, 16.8 mg/mL, MW=56,633 Da) in pH7.4 PBS was treated with azido-peg4-NHS ester (55.0 mg, 142.5 µmol, 120 eq), then agitated gently overnight at RT. Unreacted azido-peg4-NHS ester was removed by buffer exchange with a 10,000 MWCO centrifugal filter (6x15 mL pH 7.4 PBS). The final volume of the azide functionalized Fc was 5.0 mL. To this was added freshly prepared solutions of CuSO 4 (500 µL of 20m

Tris(3-hydroxypropyltriazolylmethyl)amine), ascorbic acid (500 µL of 100 mM), and alkyne derivatized small molecule (95.7 mg, 47.5 µmol, Int-17). The resulting solution was agitated gently overnight. The crude mixture was diluted 1:10 in PBS pH 7.4, and purified using MabSelect Sure Resin (GE Healthcare, Chicago, IL, USA), followed by size exclusion chromatography. Purified material was quantified using a Nanodrop™ UV visible spectrophotomer (using a calculated extinction coefficient based on the amino acid sequence of h-IgG1), and concentrated to approximately 10 mg/mL using a centrifugal concentrator (10,000 MWCO). Purified molecules were analyzed using 4-12% Bis Tris SDS PAGE gels by loading 1-2 µg of each molecule into the gel, and staining using instant Blue staining. Each gel included a molecular weight ladder with the indicated molecular weight standards (FIG.26).“NR” inducates non-reduced SDS- PAGE gel condition. Maldi TOF analysis of the purified final product gave a mass of an average mass of 86,892 Da (DAR = 15). Yield 67 mg, 77% yield.

Ex m l 41– nh i f In-1 In-2 In-21 n In-22

Synthesis of Int-19 and Int-20

HATU (198 mg, 0.52 mmol, in 1 mL of DMF) was added to a stirring mixture of propargyl-PEG4- acid (148 mg, 0.57 mmol), Int-6a, TFA salt (250 mg, 0.47 mmol) and triethylamine (158 mg, 1.57 mmol) in DMF (2 mL) and the mixture was stirred at ambient temperature for 30 minutes. The mixture was applied directly to RPLC using an Isco Combiflash liquid chromatograph eluted with 10% to 95% acetonitrile and water using 0.1 % TFA modifier. The pure fractions were pooled and lyophilized to afford the di-ester intermediate. LC/MS [M+H]+= 656.4. The di-ester intermediate was stirred in a 1/1/2 mixture of

MeOH/THF/DI water containing LiOH (36 mg, 1.5 mmol) at ambient temperature for 5 minutes. The mixture was acidified with glacial acetic acid, concentrated and applied directly to RPLC using an Isco Combiflash liquid chromatograph eluted with 10% to 95% acetonitrile and water using 0.1 % TFA modifier. The pure fractions were pooled and lyophilized to afford the title compound Int-19 as a clear oil. Yield: 74%, 2 Steps. Ion found by LC/MS [M-H]- = 626.2.

Int-20 was synthesized in a similar way as described for Int-19. Isolated yield was 73%. Positive mass ions were found by LC/MS at tr = 4.10 with 8-minute 5~95% CAN/water method: (M + 2H + )/2 = 402.5. Synthesis of Int-21

A solid mixture of PMBD (example 10 Step b., 7.444 g, 4.760 mmol), Int-191.494 g, 2.380 mmol), EDCI (1.141 g, 5.950 mmol) and HOAt (0.8099 g, 5.950 mmol) were dissolved in 20 mL of dry DMF immediately followed by adding triethylamine (1.66 mL, 9.52 mmol). The mixture was stirred for overnight (~15 hours) at room temperature. HPLC analysis showed the completion of the reaction. Most of the solvent (DMF) of the reaction mixture was removed by reduced pressure rotovap to give a viscous oily residue. To this residue methanol was added with gentle stirring to triturate the reaction product. After some crystalline solid started to precipitate the flask was put standstill for more than 3 hours. Simple filtration collected the crystalline white solid with methanol wash (3 x 5 mL). The solid was dried to give 6.500 grams of pure product (73% isolated yield). The solid was dissolved with 100% TFA and stirred for 30 minutes. TFA was removed by rotovap. The residue was dissolved in minimum amount of water and purified by RPLC using an Isco Combiflash liquid chromatograph eluted with 5 % to 35% acetonitrile and water using 0.1 % TFA modifier. The pure fractions were lyophilized to afford the title product Int-21 as a white powder (TFA salt, 6.08 g, 90% isolated yield, Positive mass ions were found (M + 6H + )/6 = 453.8 and (M + 5H + )/5 = 544.2. Synthesis of Int-22

A solid mixture of PMBD (example 10 Step b, 2.505 g, 1.602 mmol), Int-20 (0.644 g, 0.801 mmol), EDCI (0.3839 g, 2.003 mmol) and HOAt (0.0.2726 g, 2.003 mmol) were dissolved in 20 mL of dry DMF immediately followed by adding triethylamine (0.56 mL, 3.2 mmol). The mixture was stirred for overnight (~15 hours) at room temperature. HPLC analysis showed the completion of the reaction. Most of the solvent (DMF) of the reaction mixture was removed by reduced pressure rotovap to give a viscous f some crystalline solid started to precipitate the flask was put standstill for more than 3 hours. Simple filtration collected the crystalline white solid with methanol wash (3 x 5 mL). The solid was dried to give 2.183 grams of pure product (70% isolated yield). The solid (2.020 g) was dissolved with 100% TFA and stirred for 30 minutes. TFA was removed by rotovap. The residue was dissolved in minimum amount of water and purified by RPLC using an Isco Combiflash liquid chromatograph eluted with 5 % to 35% acetonitrile and water using 0.1 % TFA modifier. The pure fractions were lyophilized to afford the product lnt-22 as a white powder (TFA salt, 2.297 g, 90% isolated yield,

Positive mass ions were found by LC/MS at tr = 1 .87 with 8-minute 5-95% CAN/water method:

Example 42 - Conjugate 22 (Conjugate of h-lgG1 (His) 6 Fc-2(lnt-21))

A solution of hlgG1 (His)eFc (SEQ ID NO: 8) (1 5.0 mL, 2.54 μιτιοΙ, 9.33 mg/mL, MW = 551 1 1 Da) in pH 7.4 PBSxl was treated with azido-peg4-NHS ester (0.0384 g in 150 μL of DMF, 96.9 μιτιοΙ, 38 eq), then agitated gently overnight at RT. Unreacted azido-peg4-NHS ester was removed by buffer exchange with a 10,000 MWCO centrifugal filter (6 x 15 mL pH 7.4 PBSxl ). The final volume of the azide functionalized Fc was 9.00 mL with concentration at 5.16 mg/mL, MW = 60302. To this Fc-azido PBSxl buffer solution (0.0781 g, 1 .50 μιηοΙ, 15.15 mL) were added alkyne derivatized small molecule (TFA salt, 209.7 mg, 54.4 μιτιοΙ, lnt-21 ) and freshly prepared pH 7.4 PBSxl solutions of CuSC (4.49 mL of 10.0 mM, 30 eq), tris(3-hydroxypropyltriazolylmethyl)-amine (THPTA, 4.49 mL of 10.0 mM, 30 eq), and ascorbic acid (4.49 mL of 10.0 mM, 30eq). The resulting solution was agitated gently for 4 hours at room temperature. Another portion of freshly prepared pH 7.4 PBSxl solutions of CuSC (2.25 mL, of 10.0 mM, 15 eq) was added and the reaction mixture was gently agitated for overnight (~ 15 hours) at room temperature, then the final portion of freshly prepared pH 7.4 PBSxl solutions of CuSC (2.25 mL, of 10.0 mM, 15 eq) was added and the reaction mixture was gently agitated for 2 hours. The crude mixture was diluted 1 :10 in PBS pH 7.4, and purified using MabSelect Sure Resin (GE Healthcare, Chicago, IL, USA), followed by size exclusion chromatography. Purified material was quantified using a Nanodrop™ UV visible spectrophotomer (using a calculated extinction coefficient based on the amino acid sequence of h- lgG1 ), and concentrated to approximately 10 mg/mL using a centrifugal concentrator (10,000 MWCO). Purified molecules were analyzed using 4-12% Bis Tris SDS PAGE gels by loading 1 -2 μg of each molecule into the gel, and staining using instant Blue staining. Each gel included a molecular weight ladder with the indicated molecular weight standards (FIG. 27). "NR" inducates non-reduced SDS-PAGE gel condition. MALDI TOF analysis of the purified final product gave a mass of an average mass of 82535 Da (DAR = 8). 89% yield. Example 43 - Conjugate 23 (Conjugate of h-lgGI (c-myc) Fc-2(lnt-21))

A solution of hlgG1 (c-myc) Fc (SEQ ID NO: 17) (9.50 mL, 1 .68 μιτιοΙ, 10.3 mg/mL, MW = 58,065

Da) in pH 7.4 PBSxl was treated with azido-peg4-NHS ester (98%, 6.66 mg, 16.8 μιτιοΙ, 10 eq), then agitated gently overnight at RT. Unreacted azido-peg4-NHS ester was removed by buffer exchange with a 10,000 MWCO centrifugal filter (6 x 15 mL pH 7.4 PBSxl ). The final volume of the azide functionalized Fc was 9.00 mL with concentration at 8.91 mg/mL, MW = 59006. To this F

were added freshly prepared pH 7.4 PBSx1 solutions of CuSO4 (2.04 mL of 10.0 mM, 15 eq), Tris(3- hydroxypropyltriazolylmethyl)-amine (THPTA, 2.04 mL of 10.0 mM, 15 eq), ascorbic acid(2.04 mL of 10.0 mM, 15eq), and alkyne derivatized small molecule (TFA salt, 209.7 mg, 54.4 µmol, Int-21). The resulting solution was agitated gently for 4 hours at room temperature. Another portion of freshly prepared pH 7.4 PBSx1 solutions of CuSO4 (1.02 mL, of 10.0 mM, 7.5 eq) was added and the reaction mixture was gently agitated for overnight (~ 15 hours) at room temperature, then the final portion of freshly prepared pH 7.4 PBSx1 solutions of CuSO4 (1.02 mL, of 10.0 mM, 7.5 eq) was added and the reaction mixture was gently agitated for 2 hours. The crude mixture was diluted 1:10 in PBS pH 7.4, and purified using MabSelect Sure Resin (GE Healthcare, Chicago, IL, USA), followed by size exclusion chromatography. Purified material was quantified using a Nanodrop™ UV visible spectrophotomer (using a calculated extinction coefficient based on the amino acid sequence of h-IgG1), and concentrated to approximately 10 mg/mL using a centrifugal concentrator (10,000 MWCO). Purified molecules were analyzed using 4-12% Bis Tris SDS PAGE gels by loading 1-2 µg of each molecule into the gel, and staining using instant Blue staining. Each gel included a molecular weight ladder with the indicated molecular weight standards (FIG.28). “NR” inducates non-reduced SDS-PAGE gel condition. MALDI TOF analysis of the purified final product gave a mass of an average mass of 69010 Da (DAR = 3.7). 75% yield.

Example 44– Synthesis of Int-23

Step a. Synthesis of Int-1-Gly Conjugate

To a mixture of Int-1 (4.24 g, 4 mmol) and Z-Gly-OH (1 g, 4.8 mmol) in anhydrous DMF (8 mL) was added HATU (1.87 g, 4.9 mmol) in portions over 20 minutes, followed by DIPEA (936 mg, 7.2 mmol). After the reaction mixture was stirred for 15 minutes, it was poured into water (100 mL). The white solid product was collected by filtration and washed with water. The material was re-dissolved in MeOH (50 mL) and treated with Pd/C (1 g). The mixture was stirred under hydrogen overnight. Pd/C was then filtered off, and the filtrate was concentrated and purified through RPLC (150 g, 15 to 75 % MeOH and water). Yield 4.02 g, 89.8 %. Ion found by LCMS: [(M– Boc + 2H)/2] + = 510.4, [(M– 3Boc + 2H)/2] + = 410.2. Step b.

To a mixture of the step-a product (4.02 g, 3.592 mmol) and Z-Thr-OH (980.3 mg, 3.87 mmol) in anhydrous DMF (5 mL) was added HATU (1.47 g, 3.87 mmol) in portions over 10 minutes, followed by DIPEA (755 mg, 5.8 mmol). After the addition, the reaction was stirred for 20 minutes and then poured into water (100 mL). The white solid product was collected by filtration and washed with water. The material was re-dissolved in MeOH (50 mL) and added with Pd/C (1 g). The mixture was stirred under hydrogen overnight. Pd/C was then filtered, and the filtrate was concentrated and purified through RPLC (150 g, 15 to 80 % MeOH and water). Yield 3.68 g, 83.9 %. Ions found by LCMS: [(M - 2Boc + 2H)/2] + = 511. Step c.

A mixture of the step-b product (1.2 g, 0.984 mmol) and Z-Dab(Boc)-OH.DCHA (605 mg, 1.13 mmol) was dissolved in anhydrous NMP (3 mL). It was added with HATU (430 mg, 1.13 mmol) in portions over 5 minutes, followed by DIPEA (150 mg, 1.13 mmol). The reaction was stirred for 30 minutes and then directly purified through RPLC (100 g, 40 to 100 % MeOH and water). The collected fractions were concentrated by rotary evaporation to a white solid (Ion found by LCMS: [M - 2Boc + 2H)/2] + = 678). The material was re-dissolved in MeOH (30 mL) and treated with Pd/C. The mixture was stirred under hydrogen overnight. Pd/C was filtered, and the filtrate was concentrated by rotary evaporation and further dried under high vacuum. Yield 1.07 g, 76.6%. Ion found by LCMS: [(M - 2Boc + 2H)/2] + = 611.

A mixture of the step-c product (313.6 mg, 0.2207 mmol) and INT-8 alkyne-PEG4- Pyrrolidine- (NLeu-CO2H)2 (65.9 mg, 0.105 mmol) was dissolved in anhydrous DMF (1 mL). It was added with DIPEA (65 mg, 0.5 mmol) followed by HATU (83.9 mg, 0.2207 mmol) in portions over 5 minutes. The reaction was stirred for 30 minutes and then directly purified through RPLC (50 g, 40 to 100 % MeOH and water). Yield 165 mg, 49.6 %. Ions found by LCMS: [M - 3Boc + 3H)/3] + = 1044.6, [

[M - 6Boc + 3H)/3] + = 944.4.

The step-d product (165 mg, 0.0521 mmol) was dissolved in TFA/DCM (1:1, 1 mL), and the solution was stirred for 30 minutes. It was directly purified by RPLC (50 g, 5 to 30 % acetonitrile and water, using 0.1 % TFA as modifier). Yield 160 mg, 86.6 %. Ions found by LCMS: [M + 4H)/4] + = 658.4, [M + 5H)/5] + = 527.1, [M + 6H)/6] + = 439.4, [M + 7H)/7] + = 376.8. Example 45– Conjugate 24 (Conjugate of h-IgG1(c-myc) Fc-2(Int-23))

Int-21 (60 mg, 0.0169 mmol) was added to h-IgG1(c-myc) Fc (SEQ ID NO: 17) -(azido)5 conjugate (0.0002079 M, 2 mL) solution, followed by 0.832 mL of 0.01 M CuSO4 solution, 0.832 mL of 0.01 M sodium ascorbate solution, and 0.832 mL of 0.01 M THPTA solution. The reaction was gently shaken at room temperature of 4 hours, then added with an additional amount of freshly prepared CuSO4 solution (0.01M, 0.416 mL). The reaction was shaken overnight. Additional CuSO4 solution (0.01 M, 0.416 mL, freshly prepared) was added, and the reaction mixture was shaken for another 2 hours. The crude mixture was diluted 1:10 in PBS pH 7.4, and purified using MabSelect Sure Resin (GE Healthcare, Chicago, IL, USA), followed by size exclusion chromatography. Purified material was quantified using a Nanodrop™ UV visible spectrophotomer (using a calculated extinction coefficient based on the amino acid sequence of h-IgG1), and concentrated to approximately 10 mg/mL using a centrifugal concentrator (10,000 MWCO). Purified molecules were analyzed using 4-12% Bis Tris SDS PAGE gels by loading 1-2 µg of each molecule into the gel, and staining using instant Blue staining. Each gel included a molecular weight ladder with the indicated molecular weight standards (FIG.29).“NR” inducates non-reduced SDS- PAGE gel condition. Maldi TOF analysis of the purified final product gave a

(DAR = 4.2). Yield 24.5 mg, 99 % yield. Example 46– Conjugate 25 (Conjugate of h-IgG1(c-myc) Fc-2(Int-22))

A solution of hIgG1(His) 6 Fc (SEQ ID NO: 17) (20.0 mL, 3.62 µmol, 10.5 mg/mL, MW = 57810Da) in pH 7.4 PBSx1 was treated with azido-peg4-NHS ester (98%, 6.66 mg, 16.8 µmol, 10 eq), then agitated gently overnight at RT. Unreacted azido-peg4-NHS ester was removed by buffer exchange with a 10,000 MWCO centrifugal filter (6 x 15 mL pH 7.4 PBSx1). The final volume of the azide functionalized Fc was 9.00 mL with concentration at 12.4 mg/mL, MW = 59641. To this Fc-azido PBSx1 buffer solution (0.0248 g, 0.416 µmol, 2.00 mL were added alkyne derivatized small molecule (TFA salt, 67.1 mg, 16.6 µmol, Int- 22) and freshly prepared pH 7.4 PBSx1 solutions of CuSO4 (0.832 mL of 10.0 mM, 20 eq), tris(3- hydroxypropyltriazolylmethyl)-amine (THPTA, 0.832 mL of 10.0 mM, 20 eq), ascorbic acid (0.832 mL of 10.0 mM, 20eq). The resulting solution was agitated gently for 4 hours at room temperature. Another portion of freshly prepared pH 7.4 PBSx1 solutions of CuSO4 (0.420 mL, of 10.0 mM, 10 eq) was added and the reaction mixture was gently agitated for overnight (~ 15 hours) at room temperature, then the final portion of freshly prepared pH 7.4 PBSx1 solutions of CuSO4 (0.420 mL, of 10.0 mM, 20 eq) was added and the reaction mixture was gently agitated for 2 hours. The crude mixture was diluted 1:10 in PBS pH 7.4, and purified using MabSelect Sure Resin (GE Healthcare, Chicago, IL, USA), followed by size exclusion chromatography. Purified material was quantified using a Nanodrop™ UV visible

spectrophotomer (using a calculated extinction coefficient based on the amino acid sequence of h-IgG1), and concentrated to approximately 10 mg/mL using a centrifugal concentrator (10,000 MWCO). Purified molecules were analyzed using 4-12% Bis Tris SDS PAGE gels by loading 1-2 µg of each molecule into the gel, and staining using instant Blue staining. Each gel included a molecular weight ladder with the indicated molecular weight standards (FIG.30).“NR” inducates non-reduced SDS-PAGE gel condition. MALDI TOF analysis of the purified final product gave a mass of an average mass of 71842 Da (DAR = 4.2). Yield 23 mg, 77% yield. Example 47– Conjugate 26 (Conjugate of m-IgG2a(His) 6 Fc-3(Int-13))

A solution of mIgG2a(His) 6 Fc (SEQ ID NO: 6) (3.1 mg in 0.6 mL pH 7.4 PBS, MW~53,782 Da), and TCEP (2.1 µL of 500 mM stock in pH 7.4 PBS, 9eq/disulfide) were mixed at room temperature.

LCMS after 1hr showed complete reduction. The solution was diluted with 15 mL pH 7.8 PBS w/ 2mM EDTA and ultrafiltered (10,000 MWCO) to a volume of 1 mL, two times. Int-10 (described in Example 12) was added as powder (1.16 mg, 2 eq/hinge disulfide). LCMS after 1h at room temperature showed all of the reduced Fc was consumed. The crude mixture was diluted 1:10 in PBS pH 7.4, and purified using MabSelect Sure Resin (GE Healthcare, Chicago, IL, USA), followed by size exclusion chromatography. Purified material was quantified using a Nanodrop™ UV visible spectrophotomer (using a calculated extinction coefficient based on the amino acid sequence of m-IgG2a(His) 6 ), and concentrated to approximately 10 mg/mL using a centrifugal concentrator (10,000 MWCO). Purified molecules were

% S S G f staining using instant Blue staining. Each gel included a molecular weight la

molecular weight standards (FIG.31). Yield 2.9 mg, 82% after purification. Product peak found by LCMS 60,764 Da (DAR=2/Fc dimer). Example 48– Minimum Inhibitory Concentration (MIC) assays

Generation of E. coli BW25113 + pUC18-mcr-1.

An mcr-1 expression plasmid was constructed (GenScript; Piscataway, NJ) using the pUC18 high copy number plasmid (Yanisch-Perron, et al., 1985). A 1649-bp fragment was synthesized containing, in order from 5’ to 3’: EcoR1 restriction site (5’-GAATTC-3’), a ribosomal binding site (5’-AGGAGG-3’), a 5- bp native start codon upstream sequence (5’-TTCTC-3’) from the mcr-1-bearing plasmid pHNSHP45 (Liu, YY, et al., 2015; GenBank Accession # KP347127), the 1626-bp mcr-1 open reading frame with stop codon (“orf00073” of GenBank Accession # KP347127), and finally an Xba1 restriction site sequence (5’- TCTAGA-3’). Restriction digestion and subsequent ligation into the multiple cloning site of the pUC18 backbone resulted in directional integration of the mcr-1 gene. The pUC18-mcr-1 plasmid was transformed into chemically-competent E. coli BW25113 (Coli Genetic Stock Center #7636) as described previously (Chung, et al., PNAS.1989), recovered for 1 h shaking at 37°C in Super Optimal broth with Catabolite repression (SOC) media and then aliquots were plated on Luria-Bertani (LB) media containing 100 µg/mL of carbenicillin. Putative transformant colonies were then verified in MIC assays. Strains and culture conditions.

Bacterial strains were stored as glycerol stocks at -80°C prior to culturing on cation-adjusted Mueller-Hinton agar (MHA; BD cat. no.211438) at 37°C. Antibacterial activity was assessed against a panel consisting of Escherichia coli, Acinetobacter baumannii, Pseudomonas aeruginosa, and Klebsiella pneumoniae strains (see Table 2).

Table 2. Strains used in MIC assays

Abbreviations: CGSC - Coli Genetic Stock Center; FBS - fetal bovine serum; COL-R - colistin-resistant; ATCC - American Type Culture Collection; MDR - multidrug-resistant; CDC - Centers for Disease Control and Prevention; BEI - Biodefense and Emerging Infections Research Resou

Belgian Co-Ordinated Collections of Micro-Organisms Minimum Inhibitory Concentration (MIC) assays.

M IC assays were performed according to CLSI broth microdilution guidelines (M07-A9, M100- S23) with the exception of using a 100 µL assay volume, using RPMI media instead of Mueller-Hinton media and preparing stock compounds at 50X final concentration. Briefly, stock solutions of all antibacterial agents were prepared fresh in appropriate solvents (i.e. PBS, pH 7.4, DMSO, DI, etc.). Stock concentrations were made at 50X the highest final assay concentration and serially diluted 2- fold, 8 or 12 times in a 96-well PCR plate (VWR cat. no.83007-374). Bacterial cell suspensions generated from MHA plate cultures were prepared in 0.85% saline and adjusted to ~0.1 OD600 nm. Next, cell suspensions were diluted 1:200 in RPMI containing 5% Luria Broth (LB) to improve bacterial growth (BD Difco, cat. no.244620) to a concentration of ~5 x 10 5 CFU (colony-forming units)/mL. Due to poor solubility in Mueller-Hinton broth media, conjugates 1-15, 18-19 and 26 were evaluated initially in RPMI 1640 medium (+) L-glutamine, (-) sodium bicarbonate (“RPMI A”; MP Biomedicals, cat. no.1060124), adjusted to pH 7.0 with NaOH and buffered with MOPS (Sigma, cat. no. M3183-500G) as is specified per CLSI guidelines for antifungal susceptibility testing (M27-A3, M27-S4). Later, conjugates 16 and 17, and 20-25 were evaluated in RPMI 1640 medium (+) L-glutamine, (+) sodium bicarbonate, pH 7.4 (“RPMI B”; Gibco, cat. no.11875-085) as has been described as a more physiologically-relevant media for susceptibility testing of antibacterial compounds. Dorschner RA et al., The mammalian ionic environment dictates microbial susceptibility to antimicrobial defense peptides. FASEB J.20(1):35-42 (2017). Ninety- eight microliters of each cell suspension in MHB were added to test wells in a 96-well assay plate (Costar cat. no.3370). A Beckman Multimek 96 liquid handling robot was used to dispense 2 µL of each 50X stock compound into the plate containing 98 µL of each strain in MHB (2% final solvent concentration). E. coli strains BW25113 and ATCC 25922 were run in the presence and absence of 50% heat-inactivated fetal bovine serum (FBS; Sigma, cat. no. F4135-100 mL). Plates were mixed by shaking then incubated at 37°C overnight (16-20 h). MIC values were read visually at 100% growth inhibition for all conjugates and control drugs (Tables 3A and 3B).

Table 3C. MIC values for conjugates

Table 3D. MIC values for conjugates

Table 3E. MIC values for conjugates

Table 3F. MIC values for conjugates

Table 3G. MIC values for conjugates

Example 49– Antibacterial Activity of Conjugates in Human Blood

Overnight stationary-phase cultures were centrifuged at 3,500 x g for 10 min at room temperature and resuspended in 1X PBS to an optical density at 600 nm (OD600) of 0.4 (1 x 10 8 colony forming units [CFU]/ mL) in RPMI with L-glutamine and phenol red (Life Technologies). Bacteria were diluted 1:25 in RPMI (4 x 106 CFU/ mL) and 10 µl added to wells of a non-binding 96-well plate (Corning 3641) containing 80 ul of 50% hirudin anti-coagulated blood (complement active) or 50% heparin anti- coagulated blood (complement inactive) in RPMI. After adding 5 ul of 20X solutions of each conjugate diluted in RPMI (128– 2 ug/ mL final concentration), and 5 ul RPMI for a final blood concentration 40%, the plate was incubated under static conditions at 37°C for 90 min. Minimum bactericidal concentration (MBC) was determined by 10-fold serial dilution in 1X PBS, then plating 4 ul spots onto rectangular LB agar plates and overnight incubation at 37°C. MBC was defined as the minimum conjugate concentration required for complete bacterial clearance. An untreated growth control (40% blood, 0 µg/ mL conjugate) was included in all assays. Conjugate 23 MBCs are lower in complement active hirudin-treated blood compared to complement inactive heparin-treated blood, indicating that conjugate 23-mediated killing is complement dependent (Table 4). Table 4. Conjugate MBCs (µg/mL) in complement active hirudin-treated human blood vs.

(complement inactive heparin-treated human blood).

Abbreviations: KP, Klebsiella pneumoniae; AB, Acinetobacter baumannii; PA, Pseudomonas aeruginosa; EC, Escherichia coli; COL, colistin; R, resistant; S, sensitive. Example 50– Binding of Conjugates to Bacteria Measured by Flow Cytometry

Binding of conjugates to gram negative bacteria was measured via flow cytometry. Gram negative bacteria (E. coli ATCC 25922) were grown overnight in Luria-Bertani (LB) (BD Difco™ Cat. 244620) at 37°C while shaking. The following day, bacteria were grown to log-phase in LB until reaching an OD600 of 0.4 (~10 8 CFU/ mL). 1 mL of bacteria per condition were washed in DPBS pH 7.4 (Gibco 10010-023) and treated with 5 µM of Conjugate 3 (FIG.32A), Conjugate 5 (FIG.32B), or Conjugate 7 (FIG.32C) (shown as open, black histograms in FIGS 32A, 32B, and 32C) or control molecule without mIg2a-Fc (FIG.32A) and control molecule without hIgG1-Fc (FIGS.32B nand 32C) (shown as filled, gray histograms in FIGS.32A-32C) for 15 minutes. Bacteria were washed and molecules bound to the surface of the bacteria were detected via flow cytometry using the appropriate fluorescently-conjugated secondary antibodies. Specifically, bacteria were washed 1X in DPBS and 1X in staining buffer (20mM HEPES (Gibco 15630-080) + 1% Bovine Serum Albumin (Sigma A5611) in

blocked in staining buffer for 20 minutes at room temperature, then appropriate fluorescently conjugated secondary antibody was added (Alexa Fluor 647 conjugated, at 1:400-1:1000). Bacteria were incubated for 30 minutes in dark, washed 1X in DPBS and fixed in a 2% solution of paraformaldehyde/DPBS. 50,000 events/ sample were acquired on a BD FACS Calibur and analysis was performed using FlowJo software. Viable bacteria were gated on based on FSC VS SSC properties and conjugate binding was detected as fluorescence in the PE channel. The data shown in FIGS.32A-32C demonstrate that Conjugate 3, Conjugate 5, and Conjugate 7 are able to bind to lipopolysaccharides on the surface of E. coli ATCC 25922.

Furthermore, FIGS.32D-32F are fluorescent images showing the binding of Conjugate 13 (FIG. 32D), Conjugate 7 (FIG.32E), and a negative control (FIG.32F) to E. coli, as determined by antibody binding. Example 51– Binding of Conjugates to FcRn

Binding of Conjugates Conjugate 15 or Conjugate 4 compared to unconjugated control Fc molecule (m-IgG2a type; SEQ ID NO: 2), to neonatal Fc receptor (FcRn) was analyzed by ELISA to determine the impact of drug conjugation on interaction with FcRn.

96-well ELISA plates (Fisher, Cat. No.12-565-136) were coated with 0.25 µg/well of recombinant mouse FcRn (R&D, Cat. No.6775-FC-050) or recombinant human FcRn (R&D, Cat. No.8639-FC-050) in carbonate-bicarbonate buffer (Sigma, Cat. No. C34041) at pH 6.25 overnight at 37°C. A pH of 6.25 was maintained during the complete experiment. Blocking was carried out with 5% skim milk in PBS with 0.05% Tween 20. Purified conjugates were added at 20 µg/ mL starting concentration and serially 2-fold diluted in blocking buffer. For detection, HRP-labeled secondary anti-mouse IgG-HRP (GE Healthcare, Cat. No. NA931V) or anti-human IgG-HRP (GE Healthcare, Cat. No. NA933V) was used to detect the bound conjugates. Absorbance was measured at 450 nm with a 96-well microplate reader and data were analyzed using GraphPad Prism software, FIGS.33A-33F. The results demonstrate that drug conjugation in the hinge region of an Fc domain does not interfere with the conjugate’s interaction with and binding to FcRn. Furthermore, hIgG1-Fc conjugates, Conjugate 5 and Conjugate 7 bind to both mouse and human FcRn. Example 52– Pharmacokinetics of Conjugate 23

The pharmacokinetics of conjugate 23 was evaluated in male CD-1 mice (N = 3 animals/group) after 10 mg/kg intravenous (IV) or intraperitoneal (IP) administration. Whole blood samples (by tail snip) were collected and the plasma harvested at 0.0833, 0.25, 0.5, 1, 2, 4, 8, 24, 48 and 72 hours post injection. Plasma concentrations of conjugate 23 were measured by sandwich ELISA using an Anti-Myc tag capture antibody (Abcam ab9132) at and an HRP-conjugated anti-hIgG-Fcg F(ab’)2 (Jackson ImmunoResearch 709-036-098) for detection. The amount of conjugate 23 in plasma samples was quantified using a conjugate 23 standard curve. Data were analyzed using GraphPad Prism 6 and non- linear regression, Sigmoidal 4PL. Plasma pharmacokinetic parameters wer

compartmental analysis using Phoenix WinNonlin software (Figure 34). Example 53– Complement-Dependent Cytotoxicity (CDC)

T o demonstrate that the conjugates can facilitate immune-mediated killing of compound bound bacteria CDC assays were performed. Gram-negative bacteria were cultured overnight in Luria-Bertani (LB) (BD Difco™ Cat.244620) at 37 °C under constant agitation. The next day, bacteria were grown to log phase until the OD600 reached 0.4 (~10 8 CFU/ mL). Bacteria were washed in DPBS pH 7.4 (Gibco 10010-023) resuspended in RPMI 1640 (Gibco 11-835-055) at 10 6 CFU/ mL and 100 mL/well was added to the wells of a 96-well, TC-treated, polystyrene plate (Fisher 08-772-2C). Then 10% normal human serum ((NHS) Innovative research, IPLA-CSER), 10% heat-inactivated human serum (HIS), which is complement inactive (Innovative research, IPLA-CSER, 56C for 30 minutes) or RPMI was added to wells. Finally, Conjugate 7 (cyclic heptapeptide hIgG1-Fc) or control (cyclic heptapeptide negative hIgG1-Fc aglycosylated hIgG1-Fc negative control molecule) were diluted to 0.2 uM final in RPMI and added to wells.

The plates were incubated at 37 °C for 90 minutes. At the end of the assay, wells were serially diluted in sterile water and remaining bacteria were cultured overnight at 37 °C on agar plates. Colonies were counted the next day and CFU/mL and % survival normalized to RPMI control was determined using the following equations: CFU/mL = (colonies*dilution factor)/volume and % inhibition normalized to RPMI= ([CFU/mL of test condition] / [CFU/ mL of molecule in RPMI])*100.

FIG.35 shows potent antibacterial activity of Conjugate 7 in the presence of 10% complement active NHS while no antibacterial activity is seen with the control molecule. Approximately 50% of this activity is lost in the presence of 10% complement inactive HIS demonstrating the contribution of complement-dependent cytotoxicity (CDC) to the antibacterial effect. These results demonstrate Fc- mediated antibacterial activity of Conjugate 7 against E. coli ATCC 25922. Example 54– In Vivo Efficacy Testing of Conjugates in a Mouse Model of Septicemia

In order to better determine the potency of the conjugates, several were tested in a mouse model of peritonitis/septicemia. These studies were conducted using immunocompetent C57BL/6 mice (an inbred strain). Table 5 outlines the experimental design for a study with Conjugate 12. In this experiment the challenge pathogen was a mouse virulent isolate of E. coli (ATCC 25922) which was administered IP with 5% mucin to inhibit local macrophages (0.5 mL total volume with 0.9% saline as the vehicle; 1.4E3 CFUs of bacteria delivered). Test articles were dissolved in 0.9% saline which also served as the vehicle only control and dosed IP once as indicated in the table. The end point for the study was survival over 4- days. Table 5. In Vivo Efficacy Testing of Conjugate 12 in a Mouse Model of

The study results were plotted as a Kaplan-Meier graph (FIG.36). In this study the death phase occurred over the first 48 hours post-bacterial challenge. All tested concentrations of Conjugate 12 (20, 10, & 5 mg/kg) showed increased survival over the vehicle only control group; as did colistin at 1 mg/kg, the positive control in the study. The difference in survival between vehicle only and the group receiving 5 mg/kg of Conjugate 12 was significantly different (Mantel-Cox test). Expanded dose titration of in vivo conjugate activity in a mouse model of septicemia.

In a follow up to the first efficacy study a dose titration study was run using Conjugate 3. This study was essentially performed as the previous study (Table 4) except the final challenge inoculum was 3.4E3 and all test articles/controls were dosed IV, 1 hour prior to bacterial challenge. Study results are illustrated in FIG.37 and show substantial differences between the vehicle only control group and all concentrations of Conjugate 3 ranging from 1 to 20 mg/kg (all groups, including the colistin control are statistically different relative to the vehicle control (Mantel-Cox test). Importantly, significant efficacy was even achieved at the lowest concentration of test article run (1 mg/kg) indicating the ED50 is below this level. Screening and dose titration of conjugate 23 in a mouse model of septicemia.

The activity of conjugate 23 was evaluated in vivo using an established mouse septicemia model. In this model CD-1 mice (Charles River Labs; females, 20 g) were challenged intraperitoneally (0.2 mL) with approximately 1x10 7 CFU of E. coli 25922 grown in LB media. Prior to challenge bacteria were diluted (1:1) in fresh LB + PBS with 5% gastric hog mucin to locally inhibit macrophages.

In the first study (Table 6, study 1) conjugate 23 was administered as a single dose therapeutically 1 hr. following bacterial challenge. All animals treated with the ADC survived over the course of the study (5 days) while mice treated with vehicle only (1x PBS) reached 100%

mortality. Based on the strength of these results conjugate 23 was further evaluated in this model (Table 6, study 2). In this shorter experiment an IgG1 Fc only negative control was included to exclude the possibility that the efficacy achieved in study 1 was the result of non-specific priming of the immune system by the Fc component of the ADC. As expected 4 of 5 mice treated with the negative control to 50 mg/kg demonstrated a Stepwise (dose dependent) increase in surviva

concentration.

Collectively, Conjugates 3, 12 and 23 showed significant efficacy in lethal models of E. coli sepsis. Table 6. Screening of Conjugate 23 in a lethal mouse peritonitis/septicemia model

Example 55– Synergy of ADC conjugates with Azithromycin (AZM) in an A. baumannii lung infection model

The murine pneumonia model was adapted from Lin et al., 2015 (PMID: 26288841). A. baumannii AB5075 was grown to log-phase in LB at 37 °C, 200 rpm shaking. Bacteria were washed once with PBS via centrifugation at 4,000 rpm. Bacteria were adjusted to desired concentration in PBS.

Prior to infection, 8-week-old female C57Bl/6J mice (Jackson Labs) were anesthetized with 100 mg/kg ketamine and 10 mg/kg xylazine. Using an otoscope, mice were infected directly into the trachea. For CFU burden studies mice were infected with 2 × 10 7 CFU in 30 µL PBS. At 24 h post-infection mice were sacrificed by CO2 and both lung lobes were harvested. Lungs were homogenized with 1 mm silica beads in 1 mL PBS using a MagNA Lyser (Roche). Homogenization was carried out at 6,000 rpm for 60 s and chilled on ice for 60 s in-between runs. Lung homogenate was diluted in PBS ranging from 10 -1 to 10- 8, plated on LB plates and incubated at 37 °C overnight. The next day, CFU were enumerated and surviving CFU calculated relative to weight of the lung. Mice were treated intraperitoneally with colistin (10 mpk) or PBS at t=+1 h and 5 h post-infection. Conjugates or hIgG1 (50 mpk) were administered intraperitoneally at t=-12 h pre-infection and t=+1 h post-infection. Azithromycin (Fresenius-Kabi) was administered subcutaneously once at t=+1 h post-infection. Efficacy of ADC conjugates (50 mpk) was determined upon intratracheal infection of A. baumannii AB5075. For CFU burden studies, mice (n=5 per group) were infected with 3 x 107 bacteria, sacrificed and lung CFU burden determined at 24 h time point. All ADC conjugates tested reduced CFU burden of A. baumannii AB5075– conjugate 6 by 1.12 logs (FIG. 38A), conjugate 20 by 0.91 logs (FIG. 38B) and conjugate 21 by 0.78 logs (FIG. 38C). Treatment with azithromycin (AZM) reduced CFU burden by 0.76 logs. Combination treatment of conjugates + AZM showed additive efficacy and reduced CFU burden for conjugate 6 + A

conjugate 20 + AZM by 1.81 logs (FIG.38B) and conjugate 21 + AZM by 1.50 logs (FIG.38C). Example 56– Efficacy of ADC conjugates in a lethal A. baumannii lung infection model

T he murine pneumonia model was adapted from Lin et al., 2015 (PMID: 26288841). A. baumannii AB5075 was grown to log-phase in LB at 37 °C, 200 rpm shaking. Bacteria were washed once with PBS via centrifugation at 4,000 rpm. Bacteria were adjusted to desired concentration in PBS.

Prior to infection, 8-week-old female C57Bl/6J mice (Jackson Labs) were anesthetized with 100 mg/kg ketamine and 10 mg/kg xylazine. Using an otoscope, mice were infected directly into the trachea.

Mice were treated intraperitoneally with colistin (10 mpk) or PBS at t=+1 h and 5 h post-infection. Conjugates or hIgG1 (50 mpk) were administered intraperitoneally at t=-12 h pre-infection and t=+1 h post- infection. For survival studies mice were infected with 5 × 10 7 CFU in 30 µL of PBS. Mice were treated intraperitoneally with colistin (10 mpk) or PBS at t=+1 h, 5 h, 24 h, 36 h, 48 h and 60 h post-infection. Conjugates or hIgG1 (50 mpk) were administered intraperitoneally at t=-12 h pre-infection and t=+1 h post- infection. For the survival model, survival of mice (n=5 per group) was monitored for 5 days. ADC conjugate 22 showed 100% protection which is en par with colistin as positive control that was dosed at t=+1 h, t=+7 h, t=+24 h and t=+48 h. In comparison, control IgG1 resulted in only 20% protection (FIG.39A). We found that conjugate 23 showed 100% protection and control IgG1 was not protective in this model (0%). Colistin was dosed at t=+1 h, t=+5 h, t=+24 h, t=+36 h, t=+48 h and t=+60 h and resulted in 20% survival at day 5 with mice starting to die at day 2 (FIG.39B). Example 57– Measurement of binding of ADC conjugates to LPS using an LPS displacement assay To assess the ability of ADC conjugates to bind to LPS, we incubated purified LPS with dansyl- PMB, which becomes fluorescent upon binding to LPS. Next, we measured the change in fluorescence intensity upon addition of ADC conjugates. A decrease in fluorescence intensity by ADC conjugates reflects displaced dansyl-PMB from LPS and correlates with binding of ADC conjugates to LPS.

LPS displacement was determined with fluorescent dansyl-polymyxin B (PMB). For that, 2 µL purified LPS from E. coli O111:B4 (Sigma) at 1 mg/mL was added to 45 µL of PBS and incubated with 3 µL of 200 µM dansyl-PMB (in-house) in a black 96 well plate (Costar) for 5 min at room temperature (RT) in the dark. Test articles were added to corresponding wells in 25 µL at concentrations ranging from 20 µM to 0.2 µM. After 5 min incubation at RT in the dark, the relative fluorescence unit (RFU) is determined at excitation at 340 nm and emission at 485 nm on EnSpire plate reader (Perkin Elmer).

The % LPS displacement is calculated relative to vehicle control. % LPS displacement = (1-(RFU test article– dansyl-PMB in PBS) / (RFU no test article– dansyl-PMB in PBS)) x 100.

ADC conjugate 23 binds to purified LPS from E. coli O111:B4 in dose-dependency tested from 0.05– 5 µM (FIG.40A). A dependence on drug-to-antibody-ratio (DAR) on displacement was noted. Displacement was determined from conjugate 23 variants with DARs ranging from 1.5 to 4.5 (FIG. 40B) at 1 µM and (FIG. 40C) 5 µM with displaced LPS at DAR 4.5 of over 90%. Example 58– Measurement of binding of ADC conjugates to LPS us

assay

To address the question if binding to LPS by ADC as described in Figs. 40A-C is sufficient to prevent down-stream LPS-induced activation of immune cells, we used a functional macrophage activation assay in which RAW macrophages produce nitrite oxide (NO) in response to LPS exposure. RAW 264.7 (ATCC TIB-71) murine macrophage cells were seeded at 2 x 10 5 cells in DMEM + 10% FBS per well in a 96-well plate one day prior to experiment and incubated at at 37 °C / 5% CO2. The next day, the media was aspirate and cells were washed once with PBS. Test articles were prepared in RPMI-1640 without phenol- red + 5% FBS and added in 100 µL to cells. After 15 min, 100 µL of 10 µg/ mL LPS from E. coli O111:B4 was added to each well and incubated for 24 h at 37 °C / 5% CO2. 24 h post-treatment, 150 µL of supernatant was transferred to a new 96-well plate and nitrite oxide processed according to manufacture’s instructions of the Griess reagent system kit (Promega).

The % inhibition is calculated relative to PBS as vehicle control. % Inhibition = (1- (NO produced in µM test article) / (NO produced in µM vehicle control)) x 100. We tested the impact of ADC conjugate 23 to macrophages that were stimulated with 10 µg/ mL LPS from E. coli O111:B4 to maximally induce NO production. We found that ADC conjugate 23 prevented LPS-induced macrophage activation in dose- dependency from 0.05– 5 µM (FIG.41A). Furthermore, we observed a DAR-dependent reduction in NO production from DAR 1.5 to 4.5 (FIG.41B) at 1 µM and (FIG.41C) 5 µM.

Step a. A solution of Boc-piperazine (0.316 g, 1.70 mmol), propargyl-PEG-4-bromide (0.500 g, 1.70 mmol), and potassium carbonate (0.352 g, 2.55 mmol) dissolved in acetonitrile (8 mL) was heated in a 65 °C oil bath for 15h. The reaction was concentrated and purified by RPLC using an Isco CombiFlash liquid chromatograph eluted with 10% to 100% acetonitrile and water, using 0.1% TFA as the modifier. Yield of mono-TFA salt 0.837 g, 96% yield. Ion(s) found by LCMS: (M+H) + = 401.4 Step b. A solution of Boc-piperazine-PEG-4-alkyne (0.837 g, 1.63 mmol), and TFA (5 mL) was stirred for 5 minutes, concentrated under vacuum, and purified by RPLC using an Isco CombiFlash liquid chromatograph eluted with 10% to 100% acetonitrile and water, using 0.1% TFA as the modifier. Yield of bis-TFA salt 0.783 g, 91% yield. Ion(s) found by LCMS: (M+H) + = 301.4 Step c.

A solution of racemic trans-cyclopentanone-3, 4-dicarboxylic acid (0.500 g, 2.90 mmol), L- norleucine methylester HCl (1.16 g, 6.39 mmol), DIEA (3.03 mL, 17.4 mmol), and HATU (2.43 g, 6.39 mmol) in DMF (5 mL) was stirred at room temperature for 1hr. The diastereomeric products were separated by RPLC using an Isco CombiFlash liquid chromatograph eluted with 10% to 100% acetonitrile and water, using 0.1% TFA as the modifier. The diastereomer with the shorter retention time was collected and taken on in the sequence. Yield of desired diastereomer 0.419 g, 34% yield. Ion(s) found by LCMS: did not ionize. Step d.

A solution of product from the previous step (0.280 g, 0.657 mmol), piperazine-PEG4-alkyne (0.416 g, 0.788 mmol), acetic acid (75 µL, 1.313 mmol), and dichloromethane (2 mL) was stirred for 30 min at room temperature, then treated with sodium triacetoxyborohydride (0.278 g, 1.31 mmol) in two equal portions 60 minutes apart. The reaction was stirred overnight, then purified by RPLC using an Isco CombiFlash liquid chromatograph eluted with 10% to 100% acetonitrile and water, using 0.1% TFA as the modifier. Yield of product was 0.275 g, 45% yield. Ion(s) found by LCMS: (M+H) + =711.4 Step e.

A solution of product from the previous step (0.275 g, 0.293 mmol) dissolved in THF (4 mL), was treated with a solution of lithium hydroxide (0.042 g, 1.76 mmol) dissolved in water (2 mL). LCMS after 30 min showed complete hydrolysis. The reaction was made slightly acidic with concentrated HCl, then purified by RPLC using an Isco CombiFlash liquid chromatograph eluted with 10% to 100% acetonitrile and water, using no modifier. Yield of product was 0.170 g, 85% yield. Ion(s) found by LCMS: (M+H) + = 683.0 f.

A solution of the di-acid product from the previous step (0.073 g, 0.107 mmol), INT-7a (0.351 g, 0.225 mmol, Example 8, Step a), and DIEA (0.123 mL, 0.708 mmol) dissolved in DMF (1 mL) was treated with a solution of HATU (0.090 g, 0.236 mmol), in DMF (1 mL), dropwise over 60 minutes. Product was isolated by RPLC using an Isco CombiFlash liquid chromatograph eluted with 30% to 100% methanol and water, using 0.1% TFA as the modifier. Yield of 0.350 g, 82% yield. Ion(s) found by LCMS: [M- 2(Boc)/3]+H+ = 1191.7, [M-3(Boc)/3]+H+ = 1158.4, [M-3(Boc)/4]+H+ = 869.0, [M-4(Boc)/4]+H+ = 843.7. .

A solution of the deca-boc-protected product from the previous step (0.350 g, 0.093 mmol), was stirred in TFA (3 mL) for five minutes, and then stripped of TFA using a rotary evaporator. Product was isolated by RPLC using an Isco CombiFlash liquid chromatograph eluted with 5% to 100% methanol and water, using 0.1% TFA as the modifier. Yield of the product (Int-24) as a TFA salt was 0.340 g, 88% yield. Ion(s) found by LCMS: [M/4]+H+ = 924.6, [M/5]+H+ = 694.1, [M/6]+H+ = 555.3, [M/7]+H+ = 463.0. Example 60– Synthesis of Int-25

Step a.

A flask charged with L-dimethyltartrate (1.25 g, 7.02 mmol), 5-pentynal (1.23 mL, 10.5 mmol), p- toluenesulfonic acid (0.120 g, 0.702 mmol), and toluene (35 mL), was equipped with a Dean-Stark trap and heated in a 155 °C oil bath. After 30 minutes, approximately 0.4 mL of water was collected. The reaction was cooled down, then stripped of toluene and purified by RPLC using an Isco CombiFlash liquid chromatograph eluted with 10% to 100% methanol and water, using no modifier. Yield of product was 0.854 g, 47% yield. Ion(s) found by LCMS: (M+H) + = 257.2

A solution of the bis-ester from the previous step (0.850 g, 3.32 mmol), dissolved in THF (6 mL), was treated with a solution of lithium hydroxide (0.238 g, 9.95 mmol) dissolved in water (3 mL). LCMS after 30 minutes indicated complete reaction. The reaction was made slightly acid with conc. HCl, stripped of THF, and purified by RPLC using an Isco CombiFlash liquid chromatograph eluted with 5% to 100% acetonitrile and water, using no modifier. Yield of product was 0.817 g, 108% yield. The product was contaminated with salt, and was used in the next step without further purification. Ion(s) found by LCMS: (M+H) + = 229.2

.

A solution of di-acid product from the previous step (0.035 g, 0.153 mmol), Int-7a (0.505 g, 0.323 mmol, Example 8, Step b), and DIEA (0.177 mL, 1.02 mmol) dissolved in DMF (2 mL) was treated with a solution of HATU (0.129 g, 0.339 mmol), in DMF (1 mL), dropwise over 60 minutes. The product was isolated by RPLC using an Isco CombiFlash liquid chromatograph eluted with 30% to 100% methanol and water, using no modifier. Yield of product was 0.361 g, 67% yield. Ion(s) found by LCMS: [M- 3(Boc)/3]+H+ = 1007.4, [M-4(Boc)/3]+H+ = 973.7, [M-5(Boc)/3]+H+ = 940.3. Ste d.

A solution of the deca-boc-protected product from the previous step (0.361 g, 0.109 mmol), was stirred in TFA (3 mL) for five minutes and then stripped of TFA using a rotary evaporator. Product was isolated by RPLC using an Isco CombiFlash liquid chromatograph eluted with 5% to 100% methanol and water, using 0.1% TFA as the modifier. Yield of Int-25 as a TFA salt was 0.350 g, 93% yield. Ion(s) found by LCMS: [M/3]+H+ = 773.3, [M/4]+H+ = 580.4, [M/5]+H+ = 464.7. Example 61- Synthesis of Int-26

Step a.

A solution of diacid (0.200 g, 0.876 mmol, Example 60, step b), L-norleucine methyl ester hydrochloride (0.255 g, 1.753 mmol), and DIEA (0.962 mL, 5.52 mmol), in DMF (4 mL) was treated with HATU (0.700 g, 1.841 mmol) in one portion, at room temperature. After stirring for 30 minutes, the reaction was purified by RPLC using an Isco CombiFlash liquid chromatograph eluted with 5% to 100% methanol and water, using no modifier. The product containing fractions were combined and concentrated to yield 0.210 g, 50%. Ion(s) found by LCMS: (M+H) + = 483.3 S tep b.

A solution of the diester from Step a (0.210 g, 0.435 mmol), in THF (2 mL) was treated with a solution of lithium hydroxide (0.026 g, 1.088 mmol) dissolved in water (1 mL). LCMS after 30 minutes showed complete hydrolysis. Product was purified by RPLC using an Isco CombiFlash liquid chromatograph eluted with 5% to 100% methanol and water, using no modifier. Yield of the desired diacid was 0.195 g, 98%. Ion(s) found by LCMS: (M+H) + = 455.2

.

A solution of diacid product from the previous step (0.050 g, 0.110 mmol), Int-7a (0.326 g, 0.232 mmol, Example 8, Step b), and DIEA (0.121 mL, 0.695 mmol) dissolved in DMF (2 mL) was treated with a solution of HATU (0.093 g, 0.243 mmol), in DMF (1 mL), dropwise over 60 minutes. Product was isolated by RPLC using an Isco CombiFlash liquid chromatograph eluted with 30% to 100% methanol and water, using no modifier. Yield of the desired product was 0.303 g, 74% yield. Ion(s) found by LCMS: [M- 2(Boc)/3]+H+ = 1115.5, [M-3(Boc)/3]+H+ = 1082.7, [M-4(Boc)/3]+H+ = 1049.2. Ste d.

A solution of the deca-boc-protected product from the previous step (0.303 g, 0.085 mmol), was stirred in TFA (2 mL) for five minutes, and then stripped of TFA using a rotary evaporator. The product was isolated by RPLC using an Isco CombiFlash liquid chromatograph eluted with 5% to 100% methanol and water, using 0.1% TFA as the modifier. Yield of Int-26 as a TFA salt was 0.274 g, 87% yield. Ion(s) found by LCMS: [M/3]+H+ = 849.1, [M/4]+H+ = 637.2, [M/5]+H+ = 510.0. Step a.

Neat 4-chloro butanal dimethyl acetal (1.85 g, 12.1 mmol), L-dibenzyl tartrate (1.0 g, 3.03 mmol), and methanesulfonic acid (0.029 mL, 0.454 mmol) were heated in an 80 °C oil bath for 1 hr. The crude reaction was purified directly by RPLC using an Isco CombiFlash liquid chromatograph eluted with 30% to 100% acetonitrile and water, using no modifier. Yield 0.122 g, 9.6% yield. Ion(s) found by LCMS: (M+H) + =419.2 Step b.

A solution of the diester from Step a (0.244 g, 0.583 mmol), Boc-piperazine (0.434 g, 2.33 mmol), potassium iodide (0.10 g, 0.583 mmol), and sodium bicarbonate (0.098 g, 1.17 mmol) dissolved in DMF (0.5 mL) was heated in an 80 °C oil bath for 3 hours. The crude reaction was purified directly by RPLC using an Isco CombiFlash liquid chromatograph eluted with 10% to 100% acetonitrile and water, using no modifier. Yield 0.134 g, 40% yield. Ion(s) found by LCMS: (M+H) + = 569.4 Step c.

Boc protected diester from Step b (0.215 g, 0.378 mmol) was treate

minutes, then stripped of TFA under vacuum, and purified by directly by RPLC using an Isco CombiFlash liquid chromatograph eluted with 10% to 100% acetonitrile and water, using no modifier. Yield 0.116 g, 65% yield. Ion(s) found by LCMS: (M+H) + = 469.2. Step d.

A solution of product from the previous step (0.116 g, 0.248 mmol), propargyl-PEG4-bromide (0.110 g, 0.371 mmol), and sodium bicarbonate (0.062 g, 0.743 mmol) were dissolved in DMF (2 mL) and heated in an 80 °C oil bath for 4 hr. Product was isolated by RPLC using an Isco CombiFlash liquid chromatograph eluted with 10% to 100% acetonitrile and water, using no modifier. Yield 0.072 g, 43% yield. Ion(s) found by LCMS: (M+H) + = 683.3 Step e.

A solution of the dibenzyl ester from Step d (0.072 g, 0.105 mmol), in THF (1 mL) was treated with a solution of lithium hydroxide (0.0076 g, 0.316 mmol) dissolved in water (0.5 mL). LCMS after 30 minutes showed complete hydrolysis. The reaction was made slightly acidic with conc. HCl then purified by RPLC using an Isco CombiFlash liquid chromatograph eluted with 5% to 100% methanol and water, using no modifier. Yield 0.052 g, 98%. Ion(s) found by LCMS: (M+H)+ = 503.5. f.

A solution of di-acid product from the previous step (0.065 g, 0.089 mmol), Int-75 (0.326 g, 0.232 mmol, Example 104, step e), and DIEA (0.155 mL, 0.890 mmol) dissolved in DMF (2 mL) was treated with a solution of HATU (0.117 g, 0.306 mmol), in DMF (1 mL), dropwise over 60minutes. Product was isolated by RPLC using an Isco CombiFlash liquid chromatograph eluted with 30% to 100% methanol and water, using no modifier. Yield of 0.228 g, 57% yield. Ion(s) found by LCMS: [M-3(Boc)/4]+H+ = 880.6, [M-4(Boc)/4]+H+ = 855.5, [M-5(Boc)/4]+H+ = 830.5. .

A solution of deca-boc-protected product from the previous step (0.228 g, 0.060 mmol), was stirred in TFA (2 mL) for five minutes, and then stripped of TFA using a rotary evaporator. Product was isolated by RPLC using an Isco CombiFlash liquid chromatograph eluted with 5% to 100% methanol and water, using 0.1% TFA as the modifier. The yield of Int-27 as a TFA salt was 0.157 g, 66% yield. Ion(s) found by LCMS: [M/3]+H+ = 940.4, [M/4]+H+ = 705.5, [M/5]+H+ = 564.8.

To a 60 °C stirring solution of L-dimethyltartrate (1.64 g, 9.22 mmol), 4-ethynylbenzaldehyde (600 mg, 4.61 mmol) and trimethylorthoformate (1.01 mL, 9.22 mmol), was added methane sulfonic acid (0.021 mL, 0.323 mmol) and the temperature was raised to 100 °C. After 2 h, the reaction was cooled to room temperature and diluted with 20 mL of ethyl acetate. The organic layer was carefully added to a f ( ) ( monitoring) the organic layer was separated and washed with water (50 mL

mL) and solid magnesium sulfate. The resulting mixture was filtered through celite, concentrated, and purified by normal phase liquid chromatography (silica gel) using an Isco CombiFlash liquid

chromatograph eluted with 1% to 25% hexanes and ethyl acetate. Yield 0.416 g of (4R,5R)-dimethyl 2- (4-ethynylphenyl)-1,3-dioxolane-4,5-dicarboxylate, 31% yield.1H NMR (Methanol-d4) δ: 7.57 (d, J = 8.3 Hz, 2H), 7.50 (d, J = 8.3 Hz, 2H), 6.08 (s, 1H), 5.05 (d, J = 3.8 Hz, 1H), 4.91 (d, J = 3.8 Hz, 1H), 3.85 (s, 3H), 3.81 (s, 3H), 3.56 (s, 1H). Step b.

To a stirring solution of (4R,5R)-dimethyl 2-(4-ethynylphenyl)-1,3-dioxolane-4,5-dicarboxylate (0.416 g, 1.433 mmol) in tetrahydrofuran (5.0 mL) and water (5.0 mL) it was added lithium hydroxide (0.103 g, 4.299 mmol) and stirring was continued overnight. The reaction was concentrated to about half of its volume per rotatory evaporation. Ethyl acetate (15 mL) was added and subsequently a 1.0 M solution of sulfuric acid (5.0 mL), while the mixture was stirred vigorously for 10 minutes. The organic phase was separated and then washed with water (10 mL) and brine (10 mL). The organic layer was then dried with sodium sulfate, filtered, and concentrated to dryness per vacuum techniques. HPLC analysis for the residues revealed one component in high purity, so it was used in the next step without further purification. Yield 0.306 g of (4R,5R)-2-(4-ethynylphenyl)-1,3-dioxolane-4,5-dicarboxylic acid, 82% yield. Ste c.

To a 0 °C stirring solution of (4R,5R)-2-(4-ethynylphenyl)-1,3-dioxo

(0.045 g, 0.172 mmol), Int-7a (0.537 g, 0.343 mmol, described in Example 8 step a), and DIEA (0.126 mL, 0.721 mmol) in DMF (5.0 mL), was treated with a solution of HATU (0.134 g, 0.352 mmol) in DMF (3.0 mL), dropwise. Temperature was slowly allowed to reach ambient temperature after 15 min. Upon completion, all the volatiles were rotary evaporated. The residue was purified by RPLC using an Isco CombiFlash liquid chromatograph eluted with 50% to 100% water and methanol, using no modifier. Yield 0.400 g, 69% yield. 1 H NMR diagnostic peaks (Methanol-d4) δ: 7.61 (d, J = 8.3 Hz, 2H), 7.51 (d, J = 8.3 Hz, 2H), 7.27 (m, 10H), 6.14 (s, 1H), 1.44 (br s, 90H), 1.21 (t, J = 5.1 Hz, 12H), 0.69 (d, J = 18.8 Hz, 12H). .

Product from step c (0.400 g, 0.119 mmol) was treated with dichloromethane (4.0 mL), 2-methyl- 2-butene (0.25 mL), and TFA (2.0 mL) until gas evolution ceased (bubbler monitoring), then concentrated to an oil. The oil was purified by RPLC using an Isco CombiFlash liquid chromatograph eluted with 10% to 100% water and methanol, using 0.1% TFA as the modifier. Yield 0.085 g, 20% yield desired product Int-28. Ion(s) found by LCMS: (M+3H)/3 = 784.5, (M+4H)/4 = 588.8, (M+5H)/5 = 471.2.

To a 0 °C stirring solution of (4R,5R)-2-(4-ethynylphenyl)-1,3-dioxolane-4,5-dicarboxylic acid (0.254 g, 0.969 mmol), L-norleucine methyl ester hydrochloride (0.370 g, 2.034 mmol), and DIEA (1.05 mL, 6.006 mmol) in DMF (5.0 mL), was treated with a solution of HATU (0.134 g, 0.352 mmol) in DMF (5.0 mL), dropwise. Temperature was slowly allowed to reach ambient after 15 min. Upon completion, all the volatiles were evaporated. The residue was purified by RPLC using an Isco CombiFlash liquid chromatograph eluted with 10% to 100% water and methanol using no modifier Yield 0217 g 43% yield of bis-(dimethyl ester). 1 H NMR (Methanol-d4) δ: 7.62 (d, J = 8.3 Hz, 2

6.13 (s, 1H), 4.89 (d, J = 4.4 Hz, 1H), 4.84 (d, J = 4.4 Hz, 1H), 4.49 (td, J = 9.1, 8.5, 5.0 Hz, 2H), 3.75 (s, 1H), 3.74 (s, 3H), 3.58 (s, 1H), 1.79 (m, 4H), 1.34 (m, 8H), 0.91 (m, 6H). S tep b.

To a stirring solution of bis-(dimethyl ester) product from step a (0.217 g, 0.420 mmol) in tetrahydrofuran (4.0 mL) and water (1.0 mL) it was added lithium hydroxide (0.030 g, 1.260 mmol) and stirring was continued overnight. The reaction was concentrated to about 1 mL per rotatory evaporation. Dichloromethane (15 mL) was added and subsequently a 1.0 M solution of sulfuric acid (5.0 mL), while the mixture was stirred vigorously for 10 minutes. The organic phase was separated and then washed with water (10 mL) and brine (10 mL). The organic layer was then dried with sodium sulfate, filtered, and concentrated to dryness per vacuum techniques. HPLC analysis for the residues revealed one component in high purity that was used in the next step. Yield 0.207 g of dicarboxylic acid, quantitative yield. Ste c.

To a 0 °C stirring solution of dicarboxylic acid product from step b (0.047 g, 0.096 mmol), Int-7a (0.300 g, 0.192 mmol, described in Example 8 step a), and DIEA (0.104 mL, 0.595 mmol) in DMF (5.0 mL), was treated with a solution of HATU (0.076 g, 0.201 mmol) in DMF (3.0 mL), dropwise. Temperature was slowly allowed to reach ambient temperature after 15 min. Upon completion, all the volatiles were evaporated. The residue was purified by RPLC using an Isco CombiFlash

with 50% to 100% water and methanol, using no modifier. Yield 0.287 g, 84% yield. 1 H NMR for the diagnostic peaks (Methanol-d4) δ: 7.63 (d, J = 8.2 Hz, 2H), 7.53 (d, J = 8.2 Hz, 2H), 7.35– 7.15 (m, 10H), 6.15 (s, 1H), 4.85 (br s, 2H), 1.44 (s, 90H), 1.20 (br d, J = 6.0 Hz, 12H), 0.99– 0.83 (m, 6H), 0.70 (br d, J = 17.6 Hz, 12H). Ste d.

Product from step c (0.287 g, 0.080 mmol) was treated with dichloromethane (4.0 mL), 2-methyl- 2-butene (0.25 mL), and TFA (2.0 mL) until gas evolution ceased (bubbler monitoring), then concentrated to an oil. The oil was purified by RPLC using an Isco CombiFlash liquid chromatograph eluted with 10% to 100% water and methanol, using 0.1% TFA as the modifier. Yield 0.185 g, 62% yield of desired product Int-29. Ion(s) found by LCMS: (M+4H)/4 = 645.4, (M+5H)/5 = 516.4.

Step a.

To a 0 °C stirring solution of 4-formylbenzoic acid (0.260 g, 1.729 mmol), propargyl-PEG4-amine (0.400 g, 0.729 mmol), and DIEA (0.632 mL, 3.632 mmol) in DMF (2.0 mL), was treated with HATU (0.671 g, 0.1.764 mmol). Temperature was slowly allowed to reach ambient after 15 min. Upon completion, all the volatiles were evaporated. The residue was purified by RPLC using an Isco

CombiFlash liquid chromatograph eluted with 10% to 100% water and methanol, using no modifier. Yield 0.570 g of N-(2-(2-(2-(2-(prop-2-ynyloxy)ethoxy)ethoxy)ethoxy)ethyl)-4- formylbenzamide, 91% yield. 1 H NMR (Methanol-d4) δ: 10.07 (s, 1H), 8.01 (s, 4H), 4.16 (d, J = 2.4 Hz, 2H), 3.76– 3.51 (m, 16H), 2.84 (t, J = 2.4 Hz, 1H). Step b.

To a 60 °C stirring solution of L-dimethyltartrate (0.838 g, 4.706 mmol), N-(2-(2-(2-(2-(prop-2- ynyloxy)ethoxy)ethoxy)ethoxy)ethyl)-4-formylbenzamide (0.570 mg, 1.568 mmol) and trimethyl orthoformate (1.03 mL, 9.412 mmol), it was added methane sulfonic acid (0.005 mL, 0.078 mmol) and the temperature was raised to 100 °C. After 18 h, the reaction was cooled to room temperature and diluted with 20 mL of ethyl acetate. The organic layer was carefully added to a saturated aqueous solution of sodium bicarbonate (50 mL). Upon gas evolution ceased (bubble monitoring) the organic layer was separated and washed with water (50 mL), then dried with brine (50 mL) and solid magnesium sulfate. The resulting mixture was filtered through celite, concentrated, and purified by RPLC using an Isco CombiFlash liquid chromatograph eluted with 25% to 100% water and methanol, using no modifier. Yield 0.360 g of (4R,5R)-dimethyl 2-(4-(2-(2-(2-(2-(prop-2- ynyloxy)ethoxy)ethoxy)ethoxy)ethylcarbamoyl)phenyl)-1,3-diox olane-4,5-dicarboxylate, 44% yield. 1 H NMR (Methanol-d4) δ: 8.56 (br s, 1H), 7.87 (d, J = 8.4 Hz, 2H), 7.69 (d, J = 8.4 Hz, 2H), 6.14 (s, 1H), 5.07 (d, J = 3.7 Hz, 1H), 4.94 (d, J = 3.7 Hz, 1H), 4.15 (d, J = 2.4 Hz, 2H), 3.86 (s, 3H), 3.81 (s, 3H), 3.72– 3.50 (m, 16H), 2.84 (t, J = 2.4 Hz, 1H). Step c.

To a stirring solution of (4R,5R)-dimethyl 2-(4-(2-(2-(2-(2-(prop-2- ynyloxy)ethoxy)ethoxy)ethoxy)ethylcarbamoyl)phenyl)-1,3-diox olane-4,5-dicarboxylate (0.360 g, 0.688 mmol) in tetrahydrofuran (5.0 mL) and water (2.5 mL) it was added lithium hydroxide (0.066 g, 2.751 mmol) and stirring was continued overnight. The reaction was concentrated to about 2.5 mL per rotatory evaporation. Ethyl acetate (15 mL) was added and subsequently a 1.0 M solution of sulfuric acid (4.0 mL), while the mixture was stirred vigorously for 10 minutes. The organic phase was separated and then washed with water (10 mL) and brine (10 mL). The organic layer was then dried with sodium sulfate, filtered, and concentrated to dryness. HPLC analysis revealed one component in high purity, that was used in next step without further purification. Yield 0.342 g of (4R,5R)-2-(4-(2-(2-(2-(2-(prop-2- ynyloxy)ethoxy)ethoxy)ethoxy)ethylcarbamoyl)phenyl)-1,3-diox olane-4,5-dicarboxylic acid, quantitative yield. 1 H NMR (Methanol-d4) δ: 7.87 (d, J = 8.2 Hz, 2H), 7.71 (d, J = 8.2 Hz, 2H), 6.14 (s, 1H), 4.98 (d, J = 3.9 Hz, 1H), 4.87 (d, J = 3.9 Hz, 1H), 4.16 (d, J = 2.4 Hz, 2H), 3.73– 3.52 (m, 16H), 2.83 (t, J = 2.4 Hz, 1H). Step d.

To a 0 °C stirring solution of (4R,5R)-2-(4-(2-(2-(2-(2-(prop-2- ynyloxy)ethoxy)ethoxy)ethoxy)ethylcarbamoyl)phenyl)-1,3-diox olane-4,5-dicarboxylic acid (0.400 g, 0.807 mmol), L-norleucine methyl ester hydrochloride (0.246 g, 1.695 mmol), and DIEA (0.703 mL, 4.036 mmol) in DMF (5.0 mL), was treated with a solution of HATU (0.645 g, 1.695 mmol) in DMF (5.0 mL), dropwise. Temperature was slowly allowed to reach ambient temperature after 15 min. Upon completion, all the volatiles were evaporated. The residue was purified by RPLC using an Isco CombiFlash liquid chromatograph eluted with 10% to 100% water and methanol, using no modifier. Yield 0.328 g, 54% yield of bis-(dimethyl ester) 1 H NMR (Methanol-d4) δ: 791 (d J = 83 Hz 2H) 775 (d J = 83 Hz 2H) 6.18 (s, 1H), 4.93 (d, J = 4.5 Hz, 1H), 4.86 (d, J = 4.5 Hz, 1H), 4.50 (td, J =

J = 2.4 Hz, 2H), 3.76 (s, 3H), 3.74 (s, 3H), 3.70– 3.51 (m, 16H), 2.84 (t, J = 2.4 Hz, 1H), 1.97– 1.63 (m, 4H), 1.46– 1.22 (m, 8H), 0.91 (dt, J = 9.5, 7.1 Hz, 6H) S tep e.

To a stirring solution of bis-(dimethyl ester) product from step d (0.328 g, 0.437 mmol) in tetrahydrofuran (5.0 mL) and water (2.5 mL) it was added lithium hydroxide (0.031 g, 1.312 mmol) and stirring was continued overnight. The reaction was concentrated to about 2.5 mL per rotatory evaporation. Ethyl acetate (15 mL) was added and subsequently a 1.0 M solution of sulfuric acid (4.0 mL), while the mixture was stirred vigorously for 10 minutes. The organic phase was separated and then washed with water (10 mL) and brine (10 mL). The organic layer was then dried with sodium sulfate, filtered, and concentrated to dryness. HPLC analysis revealed one component in high purity that was used in the next step without further purification. Yield 0.325 g of dicarboxylic acid, quantitative yield. f.

To a 0 °C stirring solution of dicarboxylic acid product from step e (0.058 g, 0.080 mmol), Int-7a (0250 0160 l d ib d i E l 8 t ) d DIEA (0070 L 0400 l) i DMF (30 mL), was treated with a solution of HATU (0.063 g, 0.168 mmol) in DMF (1.

was allowed to reach ambient temperature over a 15 min period. Upon completion, all the volatiles were evaporated. The residue was purified by RPLC using an Isco CombiFlash liquid chromatograph eluted with 50% to 100% water and methanol, using no modifier. Yield 0.232 g, 76% yield. 1 H NMR (Methanol- d 4) δ: 7.90 (d, J = 7.9 Hz, 2H), 7.73 (d, J = 7.9 Hz, 2H), 7.25 (m, 10H), 6.19 (s, 1H), 3.64 (br d, J = 7.8 Hz, 16H), 2.74 (br s, 1H), 1.43 (br d, J = 2.7 Hz, 90H), 1.19 (br d, J = 6.2 Hz, 12H), 0.89 (br d, J = 7.9 Hz, 6H), 0.70 (br d, J = 18.6 Hz, 12H). Step g. Deprotection to give Int-30

Product from the previous step (0.232 g, 0.061 mmol) was treated with dichloromethane (4.0 mL), 2-methyl-2-butene (0.25 mL), and TFA (2.0 mL) until gas evolution ceased (bubbler monitoring), then concentrated to an oil. The oil was purified by RPLC using an Isco CombiFlash liquid chromatograph eluted with 10% to 100% water and methanol, using 0.1% TFA as the modifier. Yield of Int-30 was 0.208 g, 86%. Ion(s) found by LCMS: (M+4H)/4 = 703.5, (M+5H)/5 = 563.2.

Step a.

In a sealed tube, a stirring mixture of methyl 3-bromo-5-(bromomethyl)benzoate (3.08 g, 10.00 mmol) and sodium azide (0.715 g, 11.00 mmol) in DMF (10 mL) was heated at 80 °C for 1 h, when HPLC analysis showed full conversion of the bromide. All the volatiles were evaporated. The residue was taken up in DCM (40 mL) and water (40 mL). The water layer was washed with additional DCM (2 x 30 mL). The combined organic layers were dried with brine and solid sodium sulfate. Upon filtration, all the volatiles were evaporated, affording 2.73 g of the crude azide derivative in high purity, as confirmed per 1H-NMR and LC-MS analysis. 1H NMR (DMSO-d6) δ: 8.03 (t, J = 1.6 Hz, 1H), 7.98– 7.93 (m, 1H), 7.90 (t, J = 1.5 Hz, 1H), 4.60 (s, 2H), 3.88 (s, 3H). This azide (2.73 g) was dissolved under stirring in THF (30 mL) and water (2 mL), and triphenylphosphine (3.148 g, 12.00 mmol) was added. The reaction was stirred at room temperature overnight, allowing gas evolution through a bubbler. All the volatiles were evaporated per vacuum techniques. The desired product was isolated by normal phase liquid chromatography using an Isco CombiFlash liquid chromatograph eluted with 1% to 20% hexanes and ethylacetate. From the desired fractions, the desired compound was obtained still contaminated with triphenylphenylphosphine oxide. This material was therefore dissolved in DCM (50 mL) and extracted an acidic solution (12 mL of 1.0 M H2SO4 and 30 mL of water). The water layer was added dropwise to a vigorously stirring solution of saturated sodium bicarbonate (60 mL) and stirring was continued overnight. The obtained mixture was extracted with DCM (3 x 30 mL). The combined organic layers were dried with brine and solid sodium sulfate. Upon filtration, all the volatiles were evaporated per vacuum techniques, affording 1.31 g, 54% of 3-(aminomethyl)-5-bromobenzoate. Ions found by LCMS: (M+H)+ = 244.0; 246.0. Step b.

A solution of methyl 3-(aminomethyl)-5-bromobenzoate (1.306 g, 5.350 mmol) and methyl 3- bromo-5-formylbenzoate (1.238 g, 5.096 mmol) in DCM (10 mL) was evaporated and left over high vacuum overnight. The residue was taken up in THF (30 mL) and treated under vigorous stirring with sodium triacetoxyborohydride (3.240 g, 15.29 mmol). This mixture was quenched after 18 h by the addition of a saturated aqueous solution of ammonium chloride (50 mL). The obtained mixture was extracted with DCM (3 x 30 mL). The combined organic layers were dried with brine and solid sodium sulfate. Upon filtration, all the volatiles were evaporated. The desired product was isolated by normal phase liquid chromatography using an Isco CombiFlash liquid chromatograph eluted with 1% to 50% hexanes and ethylacetate. Yield 1.611 g, 67% of 3-((N-(1'-(3'-carboxymethyl-5- bromo)phenylene)methylene)methyl)-5-bromobenzoate. Ions found by LCM

474.0. Step c

To a 0˚C stirring solution of methyl 3-((N-(1'-(3'-carboxymethyl-5- bromo)phenylene)methylene)methyl)-5-bromobenzoate (1.611 g, 3.419 mmol) and DIPEA (0.953 mL, 5.471 mmol) in THF (20 mL) it was added Cbz-Cl (0.732 mL, 5.129 mmol) dropwise. The temperature was raised to ambient temperature after 10 minutes and stirring was continued until complete

disappearance of the starting amine by LC-MS analysis. All the volatiles were removed by rotatory evaporation. The desired product was isolated by normal phase liquid chromatography using an Isco CombiFlash liquid chromatograph eluted with 1% to 20% hexanes and ethylacetate. Yield 2.070 g, quantitative yield of N-Cbz protected methyl 3-((N-(1'-(3'-carboxymethyl-5- bromo)phenylene)methylene)methyl)-5-bromobenzoate. Ions found by LCMS: (M+H)+ = 603.0, 605.0, 607.0. Step d

Under nitrogen, in a capped microwave reaction vessel stirring mixture of N-Cbz protected methyl 3-((N-(1'-(3'-carboxymethyl-5-bromo)phenylene)methylene)meth yl)-5-bromobenzoate (0.299 g, 0.494 mmol), potassium phenyltrifluoroborate (0.200 g, 1.087 mmol), sodium carbonate (0.209 g, 1.976 mmol) and bis(triphenylphosphine)palladium (II) dichloride (0.035 g, 0.049 mmol) in MeCN (4 mL) and water (4 mL) were irradiated with microwaves for 10 mim at 130 °C. Upon cooling, the reaction was treated under stirring with SiliaMetS® (0.200 g, 0.240 mmol) for 1 h, and TFA was added (0.380 mL, 5.0 mmol). Upon filtration, all the volatiles were evaporated per vacuum techniques. The desired product was isolated as a dodeca-TFA salt by RPLC using an Isco CombiFlash liquid chromatograph eluted with 0% to 100% methanol and water, using TFA as the modifier. Yield 0.072 g, 25% yield of N-Cbz protected 3-((N-(1'-(3'- carboxy-5-phenyl)phenylene)methylene)methyl)-5-phenylbenzoic acid. 1 H NMR (0.500 mL Chloroform-d + 0.050 mL TFA) δ: 8.18 (d, J = 5.5 Hz, 2H), 7.82 (d, J = 16.6 Hz, 2H), 7.60

7.27 (m, 15H), 5.32 (s, 2H), 4.70 (d, J = 19.9 Hz, 4H).

To a 0 °C stirring solution of N-Cbz protected 3-((N-(1'-(3'-carboxy-5- phenyl)phenylene)methylene)methyl)-5-phenylbenzoic acid (0.075 g, 0.131 mmol), Int-7a (0.466 g, 0.262 mmol, described in Example 8 step a), and DIEA (0.137 mL, 0.787 mmol) in DMF (5.0 mL), was treated with a solution of HATU (0.102 g, 0.269 mmol) in DMF (1.5 mL), dropwise. Temperature was slowly allowed to reach ambient temperature after 15 min. Upon completion, all the volatiles were evaporated. The residue was purified by RPLC using an Isco CombiFlash liquid chromatograph eluted with 50% to 100% water and methanol, using no modifier. Yield 0.451 g, 94% yield. 1 H NMR for the diagnostic peaks 1 H NMR (Methanol-d4) δ: 8.12– 7.09 (m, 31H), 5.25 (s, 2H), 1.41 (br d, J = 10.2 Hz, 90H), 1.24– 1.04 (m, 12H), 0.69 (br d, J = 18.7 Hz, 12H).

Product from step e was dissolved in methanol (25 mL) and suspended with SiliaCatPd(0)® (0.123 g, 0.025 mmol) under hydrogen atmosphere (~1 atm). When all the starting material was consumed by HPLC analysis, the mixture was filtered over a short Celite pad, and all the volatiles were evaporated per vacuum techniques. HPLC analysis for the residues revealed one component in high purity, so it was used in the next step without further purification. Yield 0.43

the diagnostic peaks 1 H NMR (Methanol-d4) δ: 8.07 (d, J = 9.0 Hz, 2H), 7.87 (d, J = 8.4 Hz, 4H), 7.74– 7.70 (m, 4H), 7.55– 7.11 (m, 18H), 1.42 (d, J = 11.1 Hz, 90H), 1.20 (br t, J = 6.7 Hz, 12H), 0.69 (br d, J = 17.9 Hz, 12H) Ste

To a 0 °C stirring solution of propargyl-PEG4-acid (0.032 g, 0.122 mmol), product from step f (0.431 g, 0.122 mmol), and DIEA (0.064 mL, 0.366 mmol) in DMF (5.0 mL), was treated with a solution of HATU (0.049 g, 0.128 mmol) in DMF (1.5 mL), dropwise. Temperature was slowly allowed to reach ambient after 15 min. Upon completion, all the volatiles were evaporated per vacuum techniques. The residue was purified by RPLC using an Isco CombiFlash liquid chromatograph eluted with 50% to 100% water and methanol, using no modifier. Yield 0.326 g, 71% yield. h

Product from step g (0.341 g, 0.086 mmol) was treated with dichloromethane (4.0 mL), 2-methyl- 2-butene (0.25 mL), and TFA (2.0 mL) until gas evolution ceased (bubbler monitoring), then concentrated t o an oil. The oil was purified by RPLC using an Isco CombiFlash liquid chromatograph eluted with 10% to 100% water and methanol, using 0.1% TFA as the modifier. Yield of In

Ion(s) found by LCMS: (M+4H)/4 = 693.3, (M+5H)/5 = 554.8. Example 67– Synthesis of Int-32

T he general method for the preparation of the extended Threonines is described in the following reference: David Aiker, Giles Hamblett, Laurence M. Harwood, Sarah M. Robertson, David J. Watkin and Eleri Williams Tetrahedron 54 1998 6089-6098.

Step a.

Propionaldehyde (7.4 g, 127 mmol, 3 eq.) was added to a solution of the 5-phenyl-morpholin-2- one (7.5 g, 42 mmol, 1 eq.) in 100 mL toluene along with 15 grams of activated 4Å sieves and a condenser. The reaction mixture was heated to reflux for 24 hours under nitrogen. The reaction was filtered and concentrated to give the crude product as an oil, which was used in the next step without any purification. Ion(s) found by LCMS: (M+H) + = 276.2. Step b.

To the crude material from step a in methanol (50 mL) was added 2 M HCI (10 mL). The reaction mixture was heated to reflux under nitrogen until LCMS indicated the absence of starting material and the presence of a new compound (1-2h). The solvent was removed in vacuo to yield a yellow oil, which was th ifi d b RPLC i I C biFl h li id h t h l t d ith 10% t 100% acetonitrile and water, using 0.1% TFA as the modifier. Yield of the produc

yield in two steps. Ion(s) found by LCMS: (M+H)+ = 267.2. Step c.

The compound from step b (1.3 g, 5 mmol) was dissolved into 30 mL methanol, Pearlman's catalyst (1.3 g) and ammonium formate (3.1 g, 10 eq.) were added to the reaction vessel and the solution degassed and heated to reflux for 4 hours. The solution was cooled and filtered to remove the catalyst. Solvent was removed in vacuo and the crude mixture was purified by RPLC using an Isco CombiFlash liquid chromatograph eluted with 10% to 100% acetonitrile and water, using 0.1% TFA as the modifier. Yield of product as yellow oil as TFA salt, 1.30 g, 100% yield. Ion(s) found by LCMS: (M+H) + = 147.1. Step d.

L ithium hydroxide (120 mg, 5 mmol) in 2 mL water was added into a solution of the amino acid ester from the previous step (0.3 g, 2 mmol) in 2 mL water, 2 mL methanol and 2 mL THF. The reaction was monitored by LCMS. After completion, the solution was treated with Amberlite® IRN-77, ion exchange resin to adjust to pH 1, then filtered, concentrated, and used in next step without further purification. Step e.

The amino acid from the previous step (0.3 g, 2 mmol) was dissolved in 5 mL methanol and 5 mL Sat. NaHCO3 solution, followed added CbzCl (510 mg, 3 mmol, 1.5 eq.), the solution was stirred for 1hour then neutralized by 1N HCl. The solution was extracted with ethyl acetate and dried over Na2SO4. The solution was concentrated and purified by RPLC using an Isco CombiFlash liquid chromatograph eluted with 10% to 100% acetonitrile and water, using 0.1% TFA as the modifier. Yield of the product as a yellow oil, 450 mg, 92% yield. Ion(s) found by LCMS: (M+H)+ = 268.1.

To a solution of Int-1 (2.1 g, 2 mmol, Example 2) and (2S)-2-{[(benzyloxy) carbonyl]-amino}-4- [(tert-butoxycarbonyl)amino]butanoic acid (740 mg, 2 mmol) in DMF (30 mL) was added EDC (0.6 g, 3 mmol), HOBT (0.45 g, 3 mmol), and DIEA (0.7 mL, 5 mmol) at room temperature. The solution was stirred overnight. The resulting solution was concentrated and re-dissolved into 5 mL methanol, then added drop-wise to 200 mL vigorously stirred water, this heterogeneous solution was stirred for 1 hour, then filtered with Buchner funnel with Filter paper. The collected crude product was re-dissolved into 20 mL methanol, then 1 g of 5% Pd/C was added to the above solution, and the mixture was stirred at room temperature under a hydrogen atmosphere overnight. Palladium was removed by filtration after the reaction was complete as determined by LCMS. The filtrate was concentrated and used next step without further purification. Ion(s) found by LCMS: [M/2]+H+= 631.9, [M/3]+H+ = 421.6. Step g.

To a solution of product from the previous step f and (3R)-N-[(benzyloxy)carbonyl]-3-hydroxy-L- norvaline (540 mg, 2 mmol) in DMF (20 mL) was added EDC (0.6 g, 3 mmol), HOBT (0.45 g, 3 mmol), DIEA (0.56 mL, 4 mmol) at room temperature. The solution was stirred overnight. The resulting solution was concentrated and re-dissolved into 5 mL methanol and added dropwise into 100 mL vigorously stirred water, this heterogeneous solution was stirred for 1 hour, then filtered with Buchner funnel with filter paper. The collected crude product was re-dissolved in 20 mL methanol, then treated with 1 g of 5% Pd/C. The mixture was stirred at room temperature under a hydrogen atmosphere overnight. The palladium charcoal was removed by filtration. The filtrate was concentrated and used next step without further purification. Ion(s) found by LCMS: [M/2]+H+= 689.4, [M/3]+H+ = 459.9. Step h.

To a solution of PMB nonapeptide-NH2 from the previous step g and (2S)-2-{[(benzyloxy) carbonyl]-amino}-4-[(tert-butoxycarbonyl)amino]butanoic acid (720 mg, 2 mmol) in DMF (20 mL) was added EDC (0.6 g, 3 mmol), HOBT (0.45 g, 3 mmol), DIEA (0.56 mL, 4 mmol) at room temperature. The solution was stirred overnight. The resulting solution was concentrated and re-dissolved into 5 mL methanol, then added drop-wise into 100 mL vigorously stirred water. This heterogeneous solution was stirred for 1hour, then filtered with Buchner funnel with filter paper. The collected crude product was dissolved in 20 mL methanol, then treated with1 g of 5% Pd/C. The mixture

temperature under a hydrogen atmosphere overnight. The palladium charcoal was removed by filtration. The filtrate was concentrated and purified by RPLC using an Isco CombiFlash liquid chromatograph eluted with 10% to 100% acetonitrile and water, using 0.1% TFA as the modifier. Yield of product as a TFA salt: 2.3 g, 73% yield. Ion(s) found by LCMS: [M/2]+H+= 789.5, [M/3]+H+ = 526,7, [M/4]+H+ = 395.3.

Step i.

To a solution of cbz-diacetic acid (2 g, 7 mmol), and methyl L-norleucine HCl salt (2.1 g, 14.2 mmol) in DMF (40 mL) was added EDC (4.0 g, 20 mmol), HOBT (3.1 g, 20 mmol), and DIEA (4.2 mL, 30 mmol) at room temperature. The solution was stirred overnight. The resulting solution was concentrated and purified by RPLC using an Isco CombiFlash liquid chromatograph eluted with 5% to 100% acetonitrile and water, using 0.1% TFA as the modifier. Yield of the product as an oil 2.7 g, 72% yield. Ion(s) found by LCMS: (M+H)+ = 536.3. Step j.

The compound from step i (536 mg, 1 mmol) was dissolved in 10 mL methanol, then 200 mg of 5% Pd/C was added to the solution. The mixture was stirred at room temperature under a hydrogen atmosphere overnight. The palladium on charcoal was filtered, concentrated, and used in next step without further purification. Step k.

A solution of the compound from step j (450 mg, 1 mmol), propargyl-PEG4-NH2 (230 mg, 1 mmol) in DMF (5 mL) was treated with EDC (300 mg, 1.5 mmol), HOBT (230 mg, 1.5 mmol), and DIEA (0.3 mL, 1.5 mmol) at room temperature. The solution was stirred overnight. The res

concentrated and purified by RPLC using an Isco CombiFlash liquid chromatograph eluted with 5% to 100% acetonitrile and water, using 0.1% TFA as the modifier. Yield of the product as an oil, 520 mg, 78% yield. Ion(s) found by LCMS: (M+H)+ = 659.4. Step l.

Lithium hydroxide (120 mg, 5 mmol) dissolved in 2 mL water was added to a solution of the amino acid ester from step k (0.65 g, 1 mmol) in 2 mL methanol and 2 mL THF. The progress of the reaction was monitored by LCMS. After completion, the solution was treated with Amberlite® IRN-77 ion exchange resin to adjust the pH to 1. The resulting solution was filtered and concentrated then used in next step without further purification. Ion(s) found by LCMS: (M+H)+ = 631.4. Step m.

The compound from the previous step l (56 mg, 0.089 mmol), and penta-Boc-polymyxin B decapeptide (295 mg, 0.18 mmol, Example 8, step a) in DMF (5 mL) were added EDC (60 mg, 0.3 mmol), HOBT (45 mg, 0.3 mmol), and DIEA (0.14 mL, 1.0 mmol) at room temperature. The solution was stirred overnight. The resulting solution was concentrated and re-dissolved into 2 mL methanol, The resulting solution was added dropwise into 50 mL vigorously stirred water, this heterogeneous solution was stirred for 1 hour, then filtered with Buchner funnel with filter paper. The crude product was used in next step without further purification.

Product from step m was treated in 2 mL DCM and 2 mL TFA at room temperature. The solution was stirred 10 min, then concentrated and purified by RPLC using an Isco CombiFlash liquid chromatograph eluted with 0% to 100% acetonitrile and water, using formic acid as the modifier. Yield of Int-32 was 0.120 g, 67% yield. Ion(s) found by LCMS: [M/4]+H+ = 691.4, [M/5]+H+ = 553.3, [M/6]+H+ =461.3, [M/7]+H+=395.5.

The title compound Int-33 was prepared analogously to Example 67, where propionaldehyde was substituted with butyraldehyde in step a of the sequence. Ion(s) found by LCMS: [M/4]+H+ = 698.4, [M/5]+H+ = 558.9, [M/6]+H+ =465.9, [M/7]+H+=399.5.

The title compound Int-34 was prepared analogously to Example 67, where propionaldehyde was substituted with heptanal in step a of the sequence. Ion(s) found by LCMS: [M/4]+H+ = 719.4.4, [M/5]+H+ = 575.7, [M/6]+H+ =479.9, [M/7]+H+=411.5. Example 70– Synthesis of Int-35

The title compound Int-35 was prepared analogously to Example 67, where propionaldehyde was substituted with 3-phenylpropionaldehyde in step a of the sequence. Ion(s) found by LCMS: [M/4]+H+ = 729.4, [M/5]+H+ = 582.7, [M/6]+H+ =486.6, [M/7]+H+=417.2.

The title compound Int-36 was prepared analogously to Example 68, where the di-acid intermediate was substituted with the di-acid described in Example 41, step m. Ion(s) found by LCMS: [M/4]+H+ = 693.2, [M/5]+H+ = 554.5, [M/6]+H+ =463.1, [M/7]+H+=397.1.

The title compound Int-37 was prepared analogously to Example 69, where the di-acid intermediate was substituted with di-acid described in the example 41, step m. Ion(s) found by LCMS: [M/4]+H+ = 714.2, [M/5]+H+ = 572.3, [M/6]+H+ =477.1, [M/7]+H+=409.1.

The title compound Int-38 was prepared analogously to Example 67, where penta-Boc-polymyxin B decapeptide containing an extended threonine was substituted with the Int-7a (described in Example 8). Ion(s) found by LCMS: [M/4]+H+ = 684.4, [M/5]+H+ = 547.7, [M/6]+H+ =456.6.

The title compound Int-39 was prepared analogously to Example 69, where the di-acid intermediate was replaced with the di-acid intermediate described in step a of Example 90. Ion(s) found by LCMS: [M/4]+H+ = 662.9, [M/5]+H+ = 530.5, [M/6]+H+ =442.3, [M/7]+H+=379.2.

The title compound Int-40 was prepared analogously to Example 68, where the di-acid intermediate was replaced with the di-acid intermediate described in step a of Example 90. Ion(s) found by LCMS: [M/4]+H+ = 641.9, [M/5]+H+ = 513.7, [M/6]+H+ =428.3, [M/7]+H+=367.2.

Step a.

Methyl N-(oxomethylidene)-L-norleucinate (510 mg, 3 mmol) was added into a solution of benzyl tert-butyl 2,2'-azanediyldiacetate (830 mg, 3 mmol) and triethyl amine (0.52 mL, 3 mmol) in 5 mL DMF at room temperature, then stirred overnight. The reaction was concentrated then purified by RPLC using an Isco CombiFlash liquid chromatograph eluted with 5% to 100% acetonitrile and water, using 0.1% TFA as the modifier. Yield of the products as an oil, 920 mg, 69% yield. Ion(s) found by LCMS: M+H = 452.3. Step b.

Product from the previous step (450 mg, 1 mmol) was dissolved into 10 mL methanol containing 5% water, then treated with 200 mg of 5% Pd/C and stirred at room temperature under a hydrogen atmosphere for 2 hours. The palladium charcoal was removed by filtration, and the filtrate was concentrated and used in next step without further purification. Step c.

The compound from step b (450 mg, 1 mmol), methyl L-norleucinate (230 mg, 1 mmol) in DMF (5 mL) was added EDC (300 mg, 1.5 mmol), HOBT (230 mg, 1.5 mmol), DIEA (0.3 mL, 1.5 mmol) at room temperature. The solution was stirred overnight. The resulting solution was concentrated and purified by RPLC using an Isco CombiFlash liquid chromatograph eluted with 5% to 100% acetonitrile and water, using 0.1% TFA as the modifier. Yield of the product as an oil, 420 mg, 86% yield. Ion(s) found by LCMS: (M+H)+ 4883 Step d.

The compound drom step c (500 mg,1 mmol) was treated with 5 mL TFA at room temperature, after the completion the reaction, the solution was concentrated and used in next step without further purification. Step e.

The compound from step d (430 mg, 1 mmol), propargyl-PEG-4-NH2 (230 mg, 1 mmol) in DMF (5 mL) was added EDC (300 mg, 1.5 mmol), HOBT (230 mg, 1.5 mmol), DIEA (0.3 mL, 1.5 mmol) at room temperature. The solution was stirred overnight. The resulting solution was concentrated and purified by RPLC using an Isco CombiFlash liquid chromatograph eluted with 5% to 100% acetonitrile and water, using 0.1% TFA as the modifier. Yield of the product as an oil, 470 mg, 73% yield. Ion(s) found by LCMS: (M+H)+ = 645.4. Step f.

Lithium hydroxide (24 mg, 1 mmol) in 1 mL water was added into a solution of the compound from step e (70 mg, 1 mmol) in 1 mL methanol and 1 mL THF. The reaction was monitored by LCMS. After completion, the solution was adjusted to pH 1 with Amberlite® IRN-77, then the resulting solution was filtered and concentrated and used in next step without further purification. Ion(s) found by LCMS: (M+H)+ = 616.4. .

The compound from step f (62 mg, 0.1 mmol), and penta-Boc-polymyxin B decapeptide (320 mg, 0.2 mmol, 2 eq) in DMF (5 mL) were added EDC (60 mg, 0.3 mmol), HOBT (45 mg, 0.3 mmol), DIEA (0.14 mL, 1.0 mmol) at room temperature. The solution was stirred overnight. The resulting solution was concentrated and re-dissolved into 2 mL methanol, the resulting solution was added drop wise into 50 mL vigorously stirred water, this heterogeneous solution was stirred for 1 hour, then filtered with Buchner funnel with Whatman filter paper. The crude product was used for next step without purification. h.

The compound from step g was treated in 2 mL DCM and 2 mL TFA at room temperature, the solution was stirred 10 min, then concentrated and purified by RPLC using an Isco CombiFlash liquid chromatograph eluted with 0% to 100% acetonitrile and water, using formic acid as the modifier. Yield of Int-41 was 0.070 g, 26% yield. Ion(s) found by LCMS: [M/4]+H+ = 694.9, [M/5]+H+ = 555.1, [M/6]+H+ =463.6, [M/7]+H+=397.5. E

Step a.

t-butyl Bromo-acetate (1.2 mg, 6 mmol) was added into a solution of propargyl-PEG-4-NH2 (460 mg, 2 mmol) and DIPEA (1.4 mL, 10 mmol) in 20 mL DMF, The resulting solution was stirred at room temperature for overnight. The reaction solution was concentrated and purified by flash chromatography to provide products. Yield of product AS AN OIL, 700 mg, 76%. Ion(s) found by LC/MS [M+H]+ =460.3. Step b.

t ert-butyl 3-(2-tert-butoxy-2-oxoethyl)-6,9,12,15-tetraoxa-3-azaoctadec -17-yn-1-oate (230 mg, 0.5 mmol) was treated with 5 mL TFA at room temperature, after the completion the reaction, the solution was concentrated and used in next step without further purification. Step c.

The compound from step b (700 mg, 2 mmol), methyl L-norleucinate (630 mg, 4.2 mmol) in DMF (40 mL) was added EDC (1.0 g, 5 mmol), HOBT (0.9 g, 6 mmol), DIEA (1.0 mL, 7 mmol) at room temperature. The solution was stirred overnight. The resulting solution was concentrated and purified by RPLC using an Isco CombiFlash liquid chromatograph eluted with 5% to 100% acetonitrile and water, using 0.1% TFA as the modifier. Yield of product as an oil, 910 mg, 75%. Ion(s) found by LCMS: (M+H)+ = 602.4. Step d.

Lithium hydroxide 24 mg, 1 mmol) in 2 mL water was added into a solution of the amino acid ester from step c (0.06 g, 0.1 mmol) in 2 mL methanol and 2 mL THF. The reaction was monitored by LCMS. After completion, the solution was adjusted to pH 1 with Amberlite® IRN-77, then the resulting solution was filtered and concentrated and used in next step without further purification. Ion(s) found by LCMS: (M+H)+ = 574.4.

The compound from step d (57 mg, 0.1 mmol), and penta-Boc-polymyxin B decapeptide (350 mg, 0.22 mmol, 2.1 eq) in DMF (5 mL) were added EDC (60 mg, 0.3 mmol), HOBT (45 mg, 0.3 mmol), DIEA (0.14 mL, 1.0 mmol) at room temperature. The solution was stirred overnight. The resulting solution was concentrated and re-dissolved into 2 mL methanol, the resulting solution was added drop wise into 50 mL vigorously stirred water, this heterogeneous solution was stirred for 1 hour, then filtered with Buchner funnel with Whatman filter paper. The crude product was used for next step without purification. f.

Product from step e was treated in 2 mL DCM and 2 mL TFA at room temperature, the solution was stirred 10 min, then concentrated and purified by RPLC using an Isco CombiFlash liquid chromatograph eluted with 0% to 100% acetonitrile and water, using formic acid as the modifier. Yield of Int-42 was 0.120 g, 67% yield. Ion(s) found by LCMS: [M/4]+H+ = 666.6, [M/5]+H+ = 533.5, [M/6]+H+ =444.7, [M/7]+H+=381.4.

Exa TFA

LiOH

O O O O N O O N NH 2 O

O EDC/HOBt H

N N

O

O NH O

O

O

Step a.

Propargyl-PEG4-acid (850 mg, 3.2 mmol), tert-butyl [2-(piperazin-1-yl)ethyl]carbamate (750 mg, 3.2 mmol) in DMF (20 mL) was added EDC (1.0 g, 5 mmol), HOBT (0.9 g, 6 mmol), DIEA (1.0 mL, 7 mmol) at room temperature. The solution was stirred overnight. The resulting solution was concentrated and purified by RPLC using an Isco CombiFlash liquid chromatograph eluted with 5% to 100% acetonitrile and water, using 0.1% TFA as the modifier. Yield of product as an oil, 850 mg, 56%. Ion(s) found by LCMS: (M+H) + = 472.3. Step b.

Intermediate from the previous step (470 mg, 1 mmol) was treated with 5 mL TFA at room temperature. After completion of the reaction, the solution was concentrated and used in next step without further purification. Step c.

Benzyl Bromo-acetate (0.8 mg, 3.5 mmol) was added into a solution of compounds from step b (470 mg, 1 mmol, TFA salt) and DIPEA (1.0 mL, 7 mmol) in 10 mL DMF, The resulting solution was stirred at room temperature for overnight. The reaction solution was concentrated and purified by flash chromatography to provide products. Yield of product as an oil, 620 mg, 92%. Ion(s) found by LC/MS [M+H] + =668.3. Step d.

Lithium hydroxide (120 mg, 5 mmol) in 2 mL water was added into a solution of the amino acid ester from step d (0.500 g, 0.74 mmol) in 2 mL methanol and 2 mL THF. The reaction was monitored by LCMS. After completion, the solution was adjusted to pH 1 with Amberlite® IRN-77, then the resulting solution was filtered and concentrated and used in next step without further purification. Yield of product as an oil 480 mg, 100% yield. Ion(s) found by LCMS: (M+H) + = 488.3. Step e.

To a solution of intermediate from the previous step (360 mg, 0.73 mmol) in DMF (20 mL) was added EDC (300 mg, 1.5 mmol), HOBT (230 mg, 1.5 mmol), DIEA (0.28 mL, 2 mmol) at room temperature. The solution was stirred overnight. The resulting solution was concentrated and purified by RPLC using an Isco CombiFlash liquid chromatograph eluted with 5% to 100% acetonitrile and water, using 0.1% TFA as the modifier. Yield of product as an oil, 420 mg, 77%. Ion(s) found by LCMS: (M+H) + = 742.5. Step f.

Lithium hydroxide (24 mg, 1 mmol) in 2 mL water was added into a solution of the amino acid f ( ) LCMS. After completion, the solution was adjusted to pH 1 with Amberlite®

solution was filtered and concentrated and used in next step without further purification. Yield of product as an oil, 140 mg, 100% yield. Ion(s) found by LCMS: (M+H) + = 714.4.

The compound from step f (50 mg, 0.067 mmol), and penta-Boc-polymyxin B decapeptide (240 mg, 0.15 mmol, 2.1 eq) in DMF (5 mL) were added EDC (60 mg, 0.3 mmol), HOBT (45 mg, 0.3 mmol), DIEA (0.14 mL, 1.0 mmol) at room temperature. The solution was stirred overnight then concentrated and re-dissolved into 2 mL methanol. This solution was added dropwise to 50 mL vigorously stirred water. This heterogeneous solution was stirred for 1 hour, then filtered with a Buchner funnel with Whatman filter paper. The crude product was used for next step without purification. Ste h.

Intermediate from step g was treated with 2 mL DCM and 2 mL TFA at room temperature. The solution was stirred 10 min, then concentrated and purified by RPLC using an Isco CombiFlash liquid chromatograph eluted with 0% to 100% acetonitrile and water, using formic

Int-43 was 0.080 g as TFA salt, 67% yield. Ion(s) found by LCMS: [M/4]+H+ = 715.7, [M/5]+H+ = 572.7, [M/6]+H+ =477.5, [M/7]+H+=409.4.

Step a.

To a solution propargyl-PEG-4-acid (0.52 g 2 mmol), and methyl L-norleucinate (0.35 g, 2 mmol) in DMF (20 mL) was added EDC (0.60 g, 3 mmol), HOBT (0.45 g, 3 mmol), DIEA (0.56 mL, 4 mmol) at room temperature. The solution was stirred overnight. The resulting solution was concentrated and purified by RPLC using an Isco CombiFlash liquid chromatograph eluted with 5% to 100% acetonitrile and water, using 0.1% TFA as the modifier. Ion(s) found by LCMS: (M+H)+ =416.2. Step b.

Lithium hydroxide (60 mg, 2.5 mmol) in 2 mL water was added into a solution of the amino acid ester from the previous step (0.4 g, 1 mmol) in 2 mL methanol and 2 mL THF. The reaction was monitored by LCMS. After completion, the solution was adjusted to pH 1 with Amberlite® IRN-77, then the resulting solution was filtered and concentrated and used in next step without further purification. Ion(s) found by LCMS: (M+H)+ = 402.2. Step c.

The compound from step d (40 mg, 0.1 mmol), and penta-Boc-poly

0.1 mmol, 1 eq) in DMF (5 mL) were added EDC (40 mg, 0.2 mmol), HOBT (30 mg, 0.2 mmol), DIEA (0.14 mL, 1.0 mmol) at room temperature. The solution was stirred overnight. The resulting solution was concentrated and re-dissolved into 2 mL methanol. The resulting solution was added dropwise into 50 mL vigorously stirred water, this heterogeneous solution was stirred for 1 hour, then filtered with Buchner funnel with Filter paper. The crude product was used in next step without further purification. .

Intermediate form the previous step was treated with 2 mL DCM and 2 mL TFA at room temperature, stirred for 10 min, then concentrated and purified by RPLC using an Isco CombiFlash liquid chromatograph eluted with 0% to 100% acetonitrile and water, using formic acid as the modifier. Yield of Int-44 was 0.060 g, 42% yield. Ion(s) found by LCMS: [M/2]+H+ = 737.9, [M/3]+H+ = 492.3, [M/4]+H+ =369.5. Exam

The title compound Int-45 was prepared analogously to Example 79, where the where penta-Boc polymyxin B decapeptide was substituted with decapeptide described in Example 69, step c of the sequence. Ion(s) found by LCMS: [M/2]+H+ = 758.9, [M/3]+H+ = 506.3, [M/4]+H+ =380.0. Example 81– Synthesis of Int-46

The title compound Int-46was prepared analogously to Examples 80, where the acid intermediate was substituted by propargyl-PEG-4-acid in step c of the sequence. Ion(s) found by LCMS: [M/2]+H+ = 681.4, [M/3]+H+ = 454.6. Example 82– Synthesis of Int-47

Step a.

A flask was charged with and (S)-Methyl 2-amino-5-methylhexanoate (0.010 g, 0.627 mmol), diacid (0.115 g, 0.285 mmol, described in Example 90, step a), DIEA (0.40 mL, 2.28 mmol) in anhydrous DMF (1 mL). To this reaction mixture was added a solution of HATU (0.238 g, 0.627 mmol) in DMF (1 mL) at a rate of 2.5 mL/h. After the addition, the reaction mixture was stirred for an addition hour then was purified by RPLC using an Isco CombiFlash liquid chromatograph eluted with 5% to 95% acetonitrile and water, using 0.1% TFA as modifier. Yield of 0.126 g, 64%. Ion(s) found by LCMS: (M+H)+ = 687.4 Step b.

A solution of bis-ester product from the previous step (0.126 g, 0.183 mmol), dissolved in MeOH:THF:H2O (1:1:2, 8 mL), was treated with lithium hydroxide solid (0.026 g, 1.10 mmol). The reaction mixture was continued stirring overnight. After the reaction was completed by monitoring with LCMS, the reaction was quenched with glacial acetic acid to pH 4, stripped of methanol and tetrahydrofuran then purified by RPLC using an Isco Combiflash liquid chromatograph eluted with 5% to 55% acetonitrile and water, using 0.1% TFA as modifier. Yield of 0.088 g, 72%. Ion(s) found by LCMS: (M+H) + = 659.3

Step c.

A flask was charged with di-acid product from the previous step (0.048 g, 0.073 mmol), HOAt (0.030 g, 0.220 mmol), EDC (0.042 g, 0.220 mmol) and Int-7a (0.252 g, 0.161 mmol, Example 25, step a) in DMF (2 mL). The reaction mixture was stirred at room temperature for 10 minutes followed by addition of DIEA (0.08 mL, 0.439 mmol) then continued to stir overnight. The deca-Boc-protected intermediate was purified by RPLC using an Isco Combiflash liquid chromatograph eluted with 5% to 25% then 100% methanol and water, using 0.1% TFA as modifier. Pure fractions were combined and concentrated to yield deca-boc-protected intermediate.

A solution of deca-Boc-protected intermediate from the previous step was stirred in TFA/CH2Cl2 (1:1, 6 mL) at ambient temperature for 1 hour then stripped of TFA and CH2Cl2 using the rotary evaporator. The product was purified by RPLC using an Isco Combiflash liquid chromatograph eluted with 5% to 30% acetonitrile and water, using 0.1% TFA as modifier. Yield of 0.087 g as TFA salt, 31%, 2 steps. Ion(s) found by LCMS: [(M+5H)/5]+ = 551.20, [(M+6H)/6]+ = 458.9

The title compound Int-48 was prepared from the di-acid product from the Example 82, step a (0.04 g, 0.061 mmol) and the extended threonine decapeptide (0.213 g, 0.134 mmol, Example 67, step h) as described in Example 67, step m. Yield 0.0767 g as TFA salt, 32%, 2 steps. Ion(s) found by LCMS: [(M+5H)/5]+ = 561.8, [(M+6H)/6]+ = 468.6. Step a.

A flask was charged with L-valine methyl ester HCl (0.970 g, 5.786 mmol), racemic-trans-(tert- butoxycarbonyl)pyrrolidine-3,4-dicarboxylic acid (0.500 g, 1.929 mmol), DIEA (2.0 mL, 11.57 mmol) in anhydrous DMF (8 mL). To this reaction mixture was added a solution of HATU (2.2 g, 5.86 mmol) in DMF (8 mL) at a rate of 4 mL/h. After stirring overnight solvent was stripped to half of the volume then purified by RPLC using an Isco Combiflash liquid chromatograph eluted with 5% to 45% acetonitrile and water, using 0.1% TFA as modifier. Yield of 0.0845 g polar diastereomer 9% (used in the next step). Yield of less polar isomer 0.18 g, 14% yield (discarded). Ion(s) found by LCMS: (M-Boc)+ = 386.2 Step b.

A solution of the Boc-Pyrrolidine product from the previous step (0.085 g, 0.174 mmol) in dioxane (3 mL) was charged with HCl (4M in dioxane, 2 mL). The reaction mixture was stirred at ambient temperature for 1 hour then concentrated under reduced pressure. The residue was rinsed with ethyl acetate (20 mL, 2x) then concentrated and dried under reduced pressure. Yield 0.160 g as HCl salt, 189%. Ion(s) found by LCMS: [M+H]+ = 386.0.

Step c.

A flask was charged with propargyl-PEG4-acid (0.054 g, 0.19 mmol), the product from the previous step (0.073 g, 0.173 mmol) and DIEA (0.09 mL, 0.519 mmol) was stirred at ambient temperature in DMF (1 mL). To this solution was added a solution of HATU (0.072 g, 0.19 mmol) in DMF (0.6 mL) via a syringe pump at a rate of 2.5 mL/hr. After stirring for 1 hr product was purified by RPLC using an Isco Combiflash liquid chromatograph eluted with 5% to 50% acetonitrile and water, using 0.1% TFA as modifier. Yield 0.114 g, 105%. Ion(s) found by LCMS: (M+H)+ = 628.4 Step d.

A solution of bis-ester from the previous step (0.10 g, 0.159 mmol), dissolved in MeOH:THF:H2O (1:1:2, 8 mL), was treated with lithium hydroxide solid (0.011 g, 0.478 mmol). The reaction mixture was continued stirring overnight. After the reaction was completed by monitoring with LCMS, the reaction was quenched with glacial acetic acid to pH 4, stripped of methanol and tetrahydrofuran then purified by RPLC using an Isco Combiflash liquid chromatograph eluted with 5% to 45% acetonitrile and water, using 0.1% TFA as modifier. Yield of 0.073 g, 77%. Ion(s) found by LCMS: (M+H)+ = 598.2

.

A 4 mL scintillation vial was charged with di-acid product from the previous step (0.073 g, 0.122 mmol), HOAt (0.051 g, 0.367 mmol), EDC (0.070 g, 0.367 mmol) and PMB-decapeptide-D-Ser (0.390 g, 0.269 mmol, Example 6, Int-5d) in DMF (2 mL). The reaction mixture was stirred at room temperature for 10 minutes followed by addition of DIEA (0.13 mL, 0.733 mmol), then continued to stir overnight. Crude product was purified by RPLC using an Isco Combiflash liquid chromatograph eluted with 5% to 25% then 100% methanol and water, using 0.1% TFA as modifier. Pure fractions were combined and concentrated to yield deca-boc-protected intermediate.

A solution of deca-Boc-protected intermediate from the previous step was stirred in TFA/CH2Cl2 (1:1, 6 mL) at ambient temperature for 1 hour then stripped of TFA and CH2Cl2 using a rotary evaporator. The product was purified by RPLC using an Isco Combiflash liquid chromatograph eluted with 5% to 30% acetonitrile and water, using 0.1% TFA as modifier. Yield of Int-49 was 0.131 g as a TFA salt, 30%, 2 steps. Ion(s) found by LCMS: [(M+4)/4]+ = 667.0, [(M+5H)/5]+ = 534.0, [(M+6H)/6]+ = 445.0

Example 85– Synthesis of Int-50

Step a.

A flask was charged with L-Valine methyl ester HCl (0.776 g, 4.63 mmol), racemic-trans1-(tert- butoxycarbonyl)pyrrolidine-3,4-dicarboxylic acid (0.40 g, 1.543 mmol), HOAt (0.861 g, 6.326 mmol), and EDC (1.21 g, 6.326 mmol) in anhydrous DMF (6 mL). The reaction mixture was stirred at ambient temperature for 10 minutes following by addition of DIEA (2.42 mL, 13.89 mmol) then continued to stir overnight. The reaction mixture was reduced by half using a rotatory evaporator, then purified by RPLC using an Isco CombiFlash liquid chromatograph eluted with 5% to 50% acetonitrile and water, using 0.1% TFA as modifier. Yield 0.144 g, 19% of the polar diastereomer (less polar isomer discarded) as a clear oil. Ion(s) found by LCMS: (M-Boc)+ = 386.2 and (M-H)+ = 484.2. Steps b, c & d.

The diacid was prepared as described in Example 84, steps b, c, d. Yield 0.17 g, 135% yield; ion(s) found by LCMS: [M+H]+ = 385.2 (Step b). Yield 0.201 g, 79% yield; ion(s) found by LCMS: [M+H]+ = 628.4 (Step c). Yield 0.138 g, 72% yield; ion(s) found by LCMS: [M+H]+ = 601.3 (Step d). Ste e.

The title compound Int-50 was prepared from the di-acid product of step d (0.069 g, 0.115 mmol) and Int-7a (0.395 g, 0.253 mmol, Example 10, step a) following the procedure described in Example 84, step e. Yield 0.042 g as TFA salt, 10%, 2 steps. Ion(s) found by LCMS: [(M+5H)/5]+ = 539.6, [(M+6H)/6]+ = 449.4. Example 86– Synthesis of Int-51

Step a.

A flask was charged with tri-Boc-PMBH-(NH2) (2.00 g, 1.883 mmol, Example 2, Int-1), 1-Cbz- aminocyclopropane carboxylic acid (0.542 g, 2.259 mmol) and DIEA (0.98 mL, 5.65 mmol) in DMF (19 mL). To this solution was added a solution of HATU (0.877 g, 2.259 mmol) in DMF (3.5 mL) via a syringe pump at rate of 2.5 mL/hr. The reaction was stirred overnight at ambient temperature. The reaction mixture concentrated to half of its volume using the rotary evaporator then purified by RPLC using an Isco CombiFlash eluting with 5% to 25% to 90% methanol and water, using 0.1% TFA as modifier. Yield 2.159 g, 90%. Ion(s) found by LCMS [(M-2Boc+2H)/2]+ = 540.5. Step b.

A solution of the product from the previous step (2.08 g, 1.62 mmol) in methanol (15 mL) was charged with Pd/SiO2 (0.405 g, 0.081 mmol) and hydrogen gas balloon. The reaction mixture was stirred at ambient temperature overnight. It was filtered through a pad of Celite® and washed with methanol, then concentrated to the title compound as white solid. Yield 1.94 g, 104%. Ion(s) was found by LCMS: : [(M-2Boc+2H)/2]+ = 473.4, [(M-3Boc+2H)/2]+ = 423.3 Step c.

A flask was charged with product from the previous step (0.979 g, 0.855 mmol), N-Cbz-L- threonine (0.227 g, 0.898 mmol), HOAt (0.175 g, 1.282 mmol) and EDC (0.246 g, 1.28 mmol) in DMF (2.6 mL) then stirred for 10 minutes following by addition of DIEA (0.45 mL, 2.56 mmol). The reaction mixture was stirred at ambient temperature overnight. It was purified by RPLC using an Isco CombiFlash eluting with 5% to 25% then 100% acetonitrile and water, using 0.1%TFA as modifier. Yield 0.84 g, 71%. Ion(s) found by LCMS: [(M-2Boc+2H)/2]+ = 590.8 Step d.

The title compound was prepared from the product of the previous step using the procedure described in Step b of this example. Yield 0.725 g, 96%. Ion(s) found by LCMS: [M-2Boc+2H)/2]+ = 523.6 Step e.

The title compound was prepared from the product of the previous (0.725 g, 0.581 mmol) and Z- dab( ^Boc)-OH DCHA salt (0.341 g, 0.639 mmol) as described in procedure from Step c. Yield 0.626 g, 68%. Ion(s) found by LCMS: [(M-2Boc+2H)/2]+ = 690.4 Step f.

The title compound was prepared from the product of the previous step (0.626 g, 0.396 mmol) and Pd/SiO2 (0.099 g, 0.02 mmol) as described in Step b of this example. Yield 0.575 g. 100%. Ion(s) found by LCMS: (M+Na)+ = 646.0 .

The title compound Int-51 was prepared from intermediate from Example 84, step d (0.069 g, 0.115 mmol) and intermediate from the previous step (0.366 g, 0.253 mmol), following the procedure from Example 84, step e. Yield 0.0478 g as TFA salt, 12%, 2 steps. Ion(s) found LCMS: [(M+4H)/4]+ = 664.8, [(M+5H)/5]+ = 532.0, [(M+6H)/6]+ = 443.6.

Step a.

The diester was prepared from methyl (2S)-2-amino-5-methylhexanoate HCl salt (0.557 g, 2.85 mmol) and racemic-trans-1-(tert-butoxycarbonyl)pyrrolidine-3,4-dicarbo xylic acid (0.352 g, 1.36 mmol) as described in Example 84, step a. Yield 0.303 g of the polar diastereomer 41%,. Ion(s) found by LCMS: [M-Boc +H]+ = 442.2. Step b.

The title compound was prepared from the product of the previous step (0.303 g, 0.56 mmol) and HCl (4N in dioxane, 6.40 mL, 46.0 mmol) as described in Example 84, step b. Yield 0.29 g, 108%. Ion(s) found by LCMS: [M+H]+ = 442.4 Step c.

The title compound was prepared from the product of the previous (0.29 g, 0.657 mmol) and propargyl-PEG4-acid (0.188 g, 0.722 mmol) as described in Example 84, step c. Yield 0.133 g, 30%. Ion(s) found by LCMS: (pos) (M+H)+ = 684.4, and (neg) (M-H)- = 682.3. Step d.

The title compound was prepared from the bis-methyl esters product of the previous step (0.133 g, 0.194 mmol) and lithium hydroxide (0.014 g, 0.582 mmol) as described by the Example 84, step d. Yield 0.12 g, 94%. Ion(s) found by LCMS: (pos) (M+H)+ = 656.3 and (neg) (M-H)+ = 655.3 .

The title compound Int-52 was prepared from the di-acid product of the previous step (0.06 g, 0.092 mmol), and the product described in Example 6, Int-5d (0.292 g, 0.201 mmol) using the procedure described in Example 84, step e. Yield 0.135 g as TFA salt, 41%, 2 steps. Ion(s) found by LCMS: [(M+5H)/5]+ = 544.9, [(M+6H)/6]+ = 454.3 Example 88– Synthesis of Int-53

The title compound Int-53 was prepared from the di-acid product described in Example 87, step d (0.06 g, 0.092 mmol) and Int-7a (0.315 g, 0.212 mmol, Example 8, step a) using the procedure described in Example 84, step e. Yield 0.116 g as TFA salt, 35%, 2 steps. Ion(s) found by LCMS: [(M+4H)/4]+ = 687.2, [(M+5H)/5]+ = 550.1, [(M+6H)/6]+ = 458.6. Example 89– Synthesis of Int-54

Step a.

The title compound was prepared from methyl (2S,3R)-2-amino-3-methylpentanoate (0.242 g, 1.66 mmol) and racemic-trans1-(tert-butoxycarbonyl)pyrrolidine-3,4-dicarbox ylic acid (0.210 g, 0.811 mmol) prepared analogously to Example 84, step a. Yield 0.121 g of polar isomer, 29%. Ion(s) found by LCMS: (M+H)+ = 514.2. Note: Only the polar isomer was taken forward to the next steps. Step b.

The title compound was prepared from the product of the previous step (0.121 g, 0.235 mmol) and HCl (4M in Dioxane, 0.59 mL, 2.35 mmol) as described in the procedure for Example 84, step b. Yield 0.122 g, 116%. Ion(s) found by LCMS: (M+H)+ = 414.2 Step c.

The title compound was prepared from the product of the previous step (0.118 g, 0.261 mmol) and propargyl-PEG4-acid (0.188 g, 0.287 mmol) as described in Example 84, step c. Yield 0.075 g, 44%. Ion(s) found by LCMS: (pos) (M+H)+ = 656.4, (M+Na)+ = 679.3 and (neg) (M-H)+ = 654.2 Step d.

The title compound was prepared from bis-methyl ester product in t

0.114 mmol) and lithium hydroxide (0.008 g, 0.34 mmol) as described by a procedure of Example 84 Step d. Yield 0.056 g, 78%. Ion(s) found by LCMS: (M+H)+ = 628.3

The title compound Int-54 was prepared from the di-acid product of the previous step (0.056 g, 0.088 mmol) and Int-7a (0.305 g, 0.195 mmol, Example 8, step a) using the procedure from Example 84, step e. Yield of 0.0326 g as TFA salt, 10%. Ion(s) found LCMS: [(M+5H)/5]+ = 544.7, [(M+6H)/6]+ = 454.3

EDC (0.77 g, 4.0 mmol) was added to a stirring mixture of diester (1.0 g, 4.0 mmol), HOBt (0.62 g, 4.0 mmol), and propargyl-peg4-amine (0.94 g, 4.0 mmol) in DMF (8 mL) and stirred for 2 hours. The mixture was reduced by half on a rotary evaporator and then purified by RPLC using an Isco Combiflash liquid chromatograph eluted with 5% to 95% acetonitrile and water using 0.1 % TFA modifier. The pure fractions were pooled and lyophilized to afford the di-ester as a clear oil. Positive ions found: by LCMS (M+H + ) = 461.2.

The di-ester was stirred in a 1:1:2 mixture (5 mL) of THF/MeOH/DI water containing LiOH (0.39 g, 16.2 mmol) for 1 hour. The mixture was acidified with glacial acetic acid (~0.7 mL) and concentrated to half the volume on the rotary evaporator. The di-acid was purified RPLC using an Isco Combiflash liquid chromatograph eluted with 10% to 95% acetonitrile and water using 0.1 % TFA modifier. The pure fractions were pooled and lyophilized to afford the di-acid as a viscous clear oil. Yield 953 mg, 59.8 %, 2 t P iti i f d b LCMS (M+H + ) 4052 Step b.

EDC (0.12 g, 0.65 mmol) was added to a stirring mixture of Isoleucine-methyl ester hydrochloride salt (0.17 g, 0.93 mmol), HOBt (0.099 g, 0.65 mmol), intermediate from the previous step (0.13 g, 0.31 mmol), and triethylamine (0.097 g, 0.96 mmol) in DMF (1.5 mL) and the reaction was stirred for 12 hours. The mixture was applied directly to RPLC using an Isco Combiflash liquid chromatograph eluted with 20% to 95% acetonitrile and water using 0.1 % TFA modifier. The pure fractions were pooled and lyophilized to afford the 200 mg of the di-ester as a white solid. Positive ions found: by LCMS (M+H + ) = 659.4. The di- acid was hydrolyzed by stirring in a 1:1:2 mixture THF/MeOH/DI water (8 mL) containing LiOH (0.029 g, 1.2 mmol) for 30 minutes at ambient temperature. Glacial acetic acid (~1 mL) was added and the volume was reduced by 70% on the rotary evaporator. The residue was purified by RPLC using an Isco

Combiflash liquid chromatograph eluted with 5% to 85% acetonitrile and water using 0.1 % TFA modifier. The pure fractions were pooled and lyophilized to afford di-acid as a clear oil. Yield 73%, 2 steps. Positive ions found: by LCMS (M+H + ) = 631.4.

.

EDC (89 mg, 0.47 mol) was added to a stirring mixture of intermediate from previous step (140 mg, 0.22 mmol), Int-7a (760 mg, 0.49 mmol), and HOBt (71 mg, 0.47 mmol) in 1.5 mL of DMF. The reaction was stirred for 12 hours at ambient temperature. The mixture was then applied directly to RPLC using an Isco Combiflash liquid chromatograph eluted with 30% to 100% methanol and water using 0.1 % TFA modifier. The fractions containing the positive ion mass observed by LCMS [(M-(3Boc)+3H + )/3] = 1141.2 were pooled and concentrated. The Boc-protected dimer was stirred in a 1/1 mixture of TFA/DCM (10 mL) at ambient temperature for 30 minutes. The solvent was removed by rotary evaporator and dried under high vacuum. The crude deprotected dimer was purified by RPLC using an Isco Combiflash liquid chromatograph eluted with 0% to 90% acetonitrile and water using 0.1 % TFA modifier. The pure fractions were pooled and lyophilized to afford desired product as a white solid deca-TFA salt. Yield of Int- 55 was 405 mg, 67%, 2 steps. Positive mass ions were found by LCMS (M+4H + )/4 = 680.8, LCMS (M+5H + )/5 = 544.6, and LCMS (M+6H + )/6 = 454.2.

E

The title compound Int-56 was prepared analogously to Example 90, where L-leucine was substituted for L-isoleucine in step A of that example. Yield 430 mg, 66%, 2 steps. Positive ions found by LCMS (M+4H + )/4 = 680.8, (M+5H + )/5 = 544.6, and (M+6H + )/6 = 454.2. E

The title compound Int-57 was prepared analogously to Example 90 where Int-7a was substituted with Int-7a described in Example 67, step h and the di-acid intermediate described in step b of Example 90. Yield of 210 mg, 69%, 2 steps. Ions found by LCMS (M+3H + )/3 = 926.

and LCMS (M+5H + )/5 = 556.4.

EDC (3.2 g, 17 mmol) was added to a stirring mixture of racemic-trans1-(tert- butoxycarbonyl)pyrrolidine-3,4-dicarboxylic acid (2 g, 7.7 mmol), Isoluecine methyl ester HCl salt (3.5 g, 19.3 mmol), HOBt (2.6 g, 17 mmol), and triethylamine (2 g, 20 mmol) in 15 mL of DMF. The reaction was stirred for 12 hours, diluted with aqueous 1N HCl (100 mL), extracted into ethyl acetate, dried over sodium sulfate and concentrated. The diastereomers were separated by RPLC using an Isco Combiflash liquid chromatograph eluted with 15% to 95% acetonitrile and water using 0.1 % TFA modifier. The polar i somer was pooled and lyophilized to afford 1.2 g of the boc-protected ester. Yield, combined 59%.LCMS [M (1boc)+H + ) = 4142 The polar isomer boc protected ester (12 g 23 mmol)) was stirred in 30 mL of 4M HCl (g) in dioxane for 30 minutes. The solvent was removed by rotary e

high vacuum to afford the di-ester-Ile-pyrolidine HCl salt as a white solid. Yield of 0.60 g, 75%. LCMS [M+H + ) = 414.2. S tep b.

HATU (389 mg, 0.87 mmol) was added to a stirring mixture of the trans- di-methyl ester-Ile- pyrolidine HCl salt (325 mg, 0.72 mmol), propargyl-peg4 acid (226 mg, 0.86 mmol), and triethylamine (291 mg.2.9 mmol) and stirred at ambient temperature for 30 minutes. The mixture was applied directly to RPLC using an Isco Combiflash liquid chromatograph eluted with 20% to 95% acetonitrile and water using 0.1 % TFA modifier. Fractions showing desired ions by LCMS (M+H + ) = 656.4 were pooled and concentrated. The residue was stirred in a 1:1:2 mixture of methanol/THF/DI water containing LiOH (52 mg, 2.2 mmol) at ambient temperature for 30 minutes. The reaction mixture was acidified with a few drops of acetic acid and the volume was reduced by half on the rotary evaporator. The mixture was purified by RPLC using an Isco Combiflash liquid chromatograph eluted with 10% to 95% acetonitrile and water using 0.1 % TFA modifier. The pure fractions were pooled and lyophilized to afford the propargyl peg4 di- acid intermediate as a clear oil. Yield of 375 mg, 83%, 2 steps. Positive ions were found by LCMS [M+H + ) = 626.2.

Step c.

The title compound Int-58 was prepared analogously to Example 90 where Int-7a was substituted with tetra Boc PMBH-Gly-Thr-Dab (described in Example 100, step c), and the intermediate described in step b of this example. Yield; 54%, 2 steps. Positive mass ions were found by LCMS (M+3H + )/3 = 877.8, LCMS (M+4H + )/4 = 658.8, LCMS (M+5H + )/5 = 527.4, and LCMS (M+6H + )/6 = 439.8.

Example 94– Synthesis of Int-59

The title compound Int-59 was prepared analogously to Example 90 step c from the intermediate from Example 93, step b and Int-5d Example 6. Yield; 44%, 2 steps. Positive mass ions were found by LCMS (M+3H + )/3 = 898.0, LCMS (M+4H + )/4 = 673.8, LCMS (M+5H + )/5 = 539.4, and LCMS (M+6H + )/6 = 449.8.

The title compound Int-60 was prepared analogously to Example 90, step c from Int-7a (Example2, step a) and the intermediate from Example 93, step b. Yield of 195 mg, 49%, 2 steps.

Positive mass ions were found by LCMS (M+3H + )/3 = 906.4, LCMS (M+4H + )/4 = 680.3, LCMS (M+5H + )/5 = 544.5.

The dimethyl ester was prepared analogously to intermediate Example 93, step a substituting L- leucine for L-isoleucine. Yield of 0.85 g, 51%, 2 steps. LCMS (M+H)+ = 414.2.

Step b.

The diacid was prepared analogously to Example 93, step b. Yield of 0.82 g, 77%, 2 steps. Negative Ion found by LCMS [M-H]- = 626.2.

The title compound Int-61 was prepared analogously to Example 90, step c from the previous intermediate and Int-7a. Yield of 179 mg, 39%, 2 steps. Positive mass ions were found by LCMS (M+3H + )/3 = 906.6, LCMS (M+4H + )/4 = 680.3, LCMS (M+5H + )/5 = 544.5. The title compound Int-62 was prepared analogously to Example 90, step c from the intermediates described in Example 96 step b, and Example 6, Int-5d. Yield of 225 mg 36%, 2 steps. Positive mass ions were found by LCMS (M+3H + )/3 = 897.8, LCMS (M+4H + )/4 = 674.0, LCMS (M+5H + )/5 = 539.4.

EDC (175 mg, 0.92 mmol) was added to a stirring mixture of di acid (synthesis described in Example 60, step b ,100 mg, 0.43 mmol), isoluecine methyl ester hydrochloride (239 mg, 1.3 mmol), HOBt (141 mg, 0.92 mmol), and triethylamine (132 mg, 1.3 mmol) in 1.5 mL of DMF. The reaction was stirred at ambient temperature for 12 hours then purified by RPLC using an Isco Combiflash liquid chromatograph eluted with 10% to 95% acetonitrile and water using 0.1 % TFA modifier. Pure fractions were pooled and concentrated to afford an oil; ion(s) found by LCMS (M+H + ) = 483.3. The oil was taken up in a 1:1:2 mixture of methanol/THF/DI water (5 mL) containing LiOH (31 mg, 1.3 mmol) and stirred at ambient temperature for 30 minutes. The mixture was acidified (pH~5) with a few drops of glacial acetic acid and concentrated to half volume. The mixture was purified by RPLC using an Isco Combiflash liquid chromatograph eluted with 5% to 80% acetonitrile and water using 01 % TFA modifier Pure fractions were pooled and lyophilized to afford the di-acid intermediate as a clear oil.

steps. Positive Ion found by LCMS (M+H + ) = 455.3. Step b.

The title compound Int-63 was prepared analogously from Int-7a and the previous intermediate from this example. Yield, 260 mg, 77%, 2 steps. Positive mass ions were found by LCMS (M+3H + )/3 = 848.8, LCMS (M+4H + )/4 = 637.0, LCMS (M+5H + )/5 = 509.9.

Example 99: Synthesis of Int-64

EDC (2.04 g, 10.7 mmol) was added to a stirring mixture of racemic-4-oxocyclopentane-1,2- dicarboxylic acid (0.8 g, 4.6 mmol), HOBt (1.6 g, 10.7 mmol), and triethylamine (1.5 g, 14.4 mmol) in DMF (12 mL) and the reaction was stirred for 12 hours. The mixture was diluted with ethyl acetate (75 mL) washed with 1N HCl (100 mL). The aqueous phase was back extracted with ethyl acetate (2x, 50 mL). The combined organics were washed with brine (100 mL), dried over sodium sulfate and concentrated. The residue was purified by ISCO Combi Flash reversed phase HPLC (acetonitrile in DI water with 0.1% TFA as modifier, 10-95% acetonitrile, 40 minute gradient. The 2 diastereomers were separated and the more polar diastereomer (by HPLC) was taken forward in subsequent reactions. Yield of 1.7 g, 85%. LCMS (M+H + ) = 427.8. The shown stereochemistry was assigned by X-ray crystallography. Step b.

Tert-butyl [ethoxy(ethyl)phosphoryl]acetate (0.43 g, 1.7 mmol) was dissolved in dry THF (15 mL) and cooled to 0°C under an atmosphere of nitrogen. NaHMDS (1.7 mL, 1.7 mmol) was added and the mixture was stirred for 30 minutes. The intermediate from step a of this example (0.36 g, 0.85 mmol, in 10 mL of THF ) was added dropwise and the reaction was stirred for 30 minutes. The reaction was quenched with 1N aqueous HCL (100 mL) and extracted into ethyl acetate (3x, 50 mL). The combined organics were dried over sodium sulfate and concentrated. The residue was purified by by ISCO Combi Flash reversed phase HPLC (25-95% acetonitrile in DI water with 0.1% TFA as modifier, 40 minute gradient. The pure fractions were pooled and lyophilized to afford 425 mg of di-ester cyclopentyl acetic acid as a clear oil. Positive ions found by LCMS [M+H]+ = 525.4. The alkene was reduced by stirring in methanol (25 mL) in the presence of 5% Pd/C (150 mg) under 1 atmosphere of hydrogen for 4 hours. The mixture was filtered through celite and concentrated. The residue was stirred in TFA (15 mL) at ambient temperature for 12 hours. The TFA was removed by rotary evaporator and the residue was purified by ISCO Combi Flash reversed phase HPLC (5-95% acetonitrile in DI water with 0.1% TFA as modifier, 40 minute gradient. The pure fractions were pooled and lyophilized to afford di methyl ester cyclopentyl acid as a clear oil. Yield of 334 mg, 83%, 3 steps. Positive ions found byLCMS [M+H]+ = 471.2 Step c.

A flask was charged with the intermediate from the previous step (0.303 g, 0.644 mmol), propagyl-PEG4-amine (0.179 g, 0.773 mmol) and DIEA (0.34 mL, 1.93 mmol) in DMF (2 mL) at room temperature. To this solution was added a prepared solution of HATU (0.294 g, 0.773 mmol) in DMF (1.22 mL) via a syringe pump at a rate of 2.5 mL/hr. The reaction mixture was continued stirring overnight then purified by RPLC without aqueous work up using Isco CombiFlash reversed phase HPLC (5-60% acetonitrile in DI water with 0.1% TFA as modifier). The pure fractions were combined and lyophilized to give propargyl-peg4-di-ester intermediate (0.41 g, 93%). Positive ions found by LCMS: [M+H]+ = 684.4 To a solution of dimethyl ester (0.41 g, 0.60 mmol) from a previous step in

m ethanol/tetrahydrofuran/DI water (1:1:2, 15 mL) was added lithium hydroxide (0.043 g, 1.80 mmol). The reaction mixture was stirred at ambient temperature for 1 hour then acidified

pH4. Methanol and tetrahydrofuran were stripped under reduced pressure. The mixture was purified by RPLC using Isco CombiFlash reversed phase HPLC (5-40% acetonitrile and DI water, using 0.1% TFA as modifier) to yield the title di-acid compound after lyophilizing, 0.411 g, 94%. Positive ions found by LCMS: [M+H]+ = 656.4. Step d.

The title compound Int-64 was prepared from the previous di-acid intermediate and Int-7a with Thr2 extended (described in Example 67, step h), analogously to Example 90 step C. Yield 125 mg, 49%, 2 steps. Positive ions found by LCMS (M+3H + )/3 = 934.6, LCMS (M+4H + )/4 = 701.3, LCMS (M+5H + )/5 = 561.5.

Example 100– Synthesis of Int-65

Step a.

To a mixture of PMB heptapeptide (4.24 g, 4 mmol, Example 2) and Z-Gly-OH (1 g, 4.8 mmol) in anhydrous DMF (8 mL) was added HATU (1.87 g, 4.9 mmol) in portions over 20 minutes, followed by DIPEA (936 mg, 7.2 mmol). After the reaction mixture was stirred for 15 minutes, it was poured into water (100 mL). The white solid product was collected by filtration and washed with water. The material was re- dissolved in MeOH (50 mL) and treated with Pd/C (5%) (1 g), then stirred under hydrogen overnight. Pd/C (5%) was then filtered, concentrated and purified by RPLC (150 g, 15 to 75 % MeOH and water). Yield 4.02 g, 90 %. Ions found by LCMS: [(M– Boc + 2H)/2] + = 510.4, [(M– 3Boc + 2H)/2] + = 410.2 Step b.

To a mixture of the step-a product (4.02 g, 3.592 mmol) and Z-Thr-OH (980.3 mg, 3.87 mmol) in anhydrous DMF (5 mL) was added HATU (1.47 g, 3.87 mmol) in portions over 10 minutes, followed by DIPEA (755 mg, 5.8 mmol). After the addition, the reaction was stirred for 20 minutes and then poured into water (100 mL). The white solid product was collected by filtration and washed with water. The material was re-dissolved in MeOH (50 mL) and treated with Pd/C (5%) (1 g), and stirred under hydrogen overnight. Pd/C was then filtered, and the filtrate was concentrated and purified by RPLC (150 g column, 15 to 80 % MeOH and water). Yield 3.68 g, 84%. Ion found by LCMS: [(M - 2Boc + 2H)/2] + = 511.0 Step c.

A mixture of the step-b product (1.2 g, 0.984 mmol) and Z-Dab(Boc)-OH DCHA salt (605 mg, 1.13 mmol) was dissolved in anhydrous NMP (3 mL). It was treated with HATU (430 mg, 1.13 mmol) in portions over 5 minutes, followed by DIPEA (150 mg, 1.13 mmol). The reaction was stirred for 30 minutes and then directly purified by RPLC (100 g column, 40 to 100 % MeOH and water). The collected fractions were concentrated by rotary evaporation to a white solid (Ion found by LCMS: [M - 2Boc + 2H)/2] + = 678). The material was re-dissolved in MeOH (30 mL) and treated with Pd/C (5%), then stirred under hydrogen overnight. Pd/C was filtered, and the filtrate was concentrated by rotary evaporation and further dried under high vacuum. Yield 1.07 g, 76.6 %. Ion found by LCMS: [(M - 2Boc + 2H)/2] + = 611. Ste d.

A mixture of the step-c product (313.6 mg, 0.2207 mmol) and diacid central linker (65.9 mg, 0.105 mmol, Example 41, INT-19) was dissolved in anhydrous DMF (1 mL). DIPEA (65 mg, 0.5 mmol) and HATU (83.9 mg, 0.2207 mmol) were added in portions over 5 minutes. The reaction was stirred for 30 minutes and then directly purified by RPLC (50 g, 40 to 100 % MeOH and water). Yield 165 mg, 50%. Ions found by LCMS: [M - 3Boc + 3H)/3] + = 1044.6, [M - 4Boc + 3H)/3] + = 1011.3, [M - 6Boc + 3H)/3] + = 944.4. .

The step-d product (165 mg, 0.0521 mmol) was dissolved in TFA/DCM (1:1, 1 mL), and the solution was stirred for 30 minutes. It was concentrated and directly purified by RPLC (50 g column, 5 to 30 % acetonitrile and water, using 0.1 % TFA as modifier). Yield of Int-65 was 160 mg, 87%. Ions found by LCMS: [M + 4H)/4] + = 658.4, [M + 5H)/5] + = 527.1, [M + 6H)/6] + = 439.4, [M + 7H)/7] + = 376.8.

E

To a solution of intermediate from Example 7, INT-6a (228.1 mg, 0.507 mmol) in anhydrous ethanol (2 mL) was added DIPEA (198 mg, 1.52 mmol) and propargyl-PEG4-bromide (180 mg, 0.608 mmol). After the mixture was heated at 80 0 C for 30 hours, it was cooled to room temperature and then placed in an ice-water bath. THF (1 mL) and a solution of LiOH (105 mg, 2.5 mmol) in water (2 mL) were added. The reaction was stirred for 3 hours. It was acidified by 4N HCl solu

directly purified by RPLC (50 g, 5 to 70 % acetonitrile and water). Yield 92.9 mg, 30.6 %. Ion found by LCMS: [M + H] + = 600.

To a solution of the step-a product (32 mg, 0.05336 mmol) and Int-7a (Example 8, step a) (183.6 mg, 0.1174 mmol) in anhydrous DMF (1 mL), was added DIPEA (42 mg, 0.4 mmol) and HATU (40.6 mg, 0.107 mmol). The resulting solution was stirred for 30 minutes and was purified by RPLC (50 g column, 20 to 100 % MeOH and water). Yield 189 mg, 96%. Ion found by LCMS: [(M– 2Boc + 3H)/3] + = 1164.4, [(M– 3Boc + 3H)/3] + = 1131, [(M– 4Boc + 3H)/3] + = 1097. .

The step-b product (189 mg, 0.0512 mmol) was dissolved in TFA (0.5 mL) and stirred for 15 minutes. It was directly purified by RPLC (50 g column, 0 to 50 % acetonitrile and water, using 0.1 % TFA as modifier). Yield of Int-66 was 103 mg, 51 %. Ions found by LCMS: [(M + 5H)/5] + = 538.6, [(M + 6H)/6] + = 449.2, [(M + 7H)/7] + = 385.0, [(M + 8H)/8] + = 337.0.

To a solution of dimethyl itaconate (1.58 g, 10 mmol) in MeOH (8 mL) was treated with t-butyl piperazine-1-carboxylate (1.86 g, 10 mmol). The mixture was heated at 60 o C for 1 day. It was then purified by HPLC (0 to 20 % acetonitrile and water, using 0.1 % TFA as modifier). Yield 1.175 g, 34.1 %. Ion found by LCMS: [M + H] + = 345.0. Step b.

The step-b product (1.175 g, 3.41 mmol) was dissolved in MeOH (5 mL) and cooled in an ice- water bath. It was treated with a solution of LiOH monohydrate (378 mg, 9 mmol) in water (9 mL). After the reaction was stirred at 0 o C to room temperature overnight, it was re-cooled in an ice-water bath and acidified by 4N HCl solution in dioxane (2 mL). The mixture was partially concentrated by rotary evaporation at room temperature. The residue was purified by RPLC (150 g column, 0 to 80 % MeOH and water). Yield 336.5 mg, 31.2 %. Ion found by LCMS: [M + H] + = 317.2. Step c.

A mixture of the step b product (336.5 mg, 1.064 mmol) and H-Nle-OMe HCl (464 mg, 2.552 mmol) was dissolved in anhydrous NMP (3 mL) and DIPEA (520 mg, 4 mmol). To this was added HATU (810 mg, 2.13 mmol) in portions over 10 minutes. The reaction was stirred for another 30 minutes, then purified by RPLC (100 g column, 10 to 75 % acetonitrile and water, using 0.1 % TFA as modifier). Yield 261.7 mg, 43.2 %. Ion found by LCMS: [M + H] + = 571.4. Step d.

The step c product (261.7 g, 0.459 mmol) was dissolved in THF (5 mL), and 4N HCl in dioxane (2.5 mL). The mixture was stirred at room temperature for 4 hours and then directly purified by RPLC (50 g, 0 to 80 % acetonitrile and water). Yield 144.3 mg, 66.9 %. Ion found by LCMS: [M + H] + = 471.4. Step e.

A mixture of the step d product (144.3 mg, 0.307 mmol) and propargyl-PEG4-acid (104 mg, 0.4 mmol) was dissolved in anhydrous DMF (1 mL) and DIPEA (130 mg, 1 mmol). HATU (152 mg, 0.4 mmol) was added, and the reaction was stirred for 30 minutes. It was then purified by RPLC (100 g, 5 to 100 % acetonitrile and water). Yield 165 mg, 75.6 %. Ion found by LCMS: [M + H] + = 713.4. Step f.

The step e product (165 mg, 0.232 mmol) was dissolved in MeOH (2 mL) and cooled in an ice- water bath. It was treated with a solution of LiOH monohydrate (21 mg, 0.5 mmol) in water (1 mL). After the reaction was stirred for 4 hours, it was acidified by 4N HCl solution in dioxane (0.2 mL) and directly purified by HPLC (5 to 35 % acetonitrile and water). Yield 46.4 mg, 29.2 %. Ion found by LCMS: [M + H] + = 685. Ste .

The title compound was prepared analogously to Example 101, ste

and Int-7a (Example 8, step a). Yield 246 mg, 96.1 %. Ions found by LCMS: [(M– 2Boc + 3H)/3] + = 1192.4, [(M– 3Boc + 3H)/3] + = 1159.1, [(M– 4Boc + 3H)/3] + = 1125.8. S tep h.

The title compound Int-67 was prepared analogously to Example 101, step-c, using the product from step-g of this example. Yield 115 mg, 43.6 %. Ions found by LCMS: [(M + 4H)/4] + = 694.4, [(M + 4H)/5] + = 555.8, [(M + 6H)/6] + = 463.2, [(M + 7H)/7] + = 397.2.

Example 103– Synthesis of Int-68

Step a.

A mixture of 3-(carboxymethyl)pentanedioic acid (190.2 mg, 1.0 mmol) and H-Nle-OMe HCl (400 mg, 2.2 mmol) was dissolved in anhydrous NMP (2 mL) and DIPEA (546 mg, 4.2 mmol). HATU (760.4 mg, 2.0 mmol) was added in portions over 10 minutes, and the reaction was stirred for 1 hour. It was then purified by RPLC (100 g column, 5 to 80 % acetonitrile and water). Yield 162.5 mg, 36.6 %. Ion found by LCMS: [M + H] + = 445.2. Step b.

A mixture of the step a product (162.5 mg, 0.366 mmol) and propargyl-PEG4-amine (101.8 mg, 0.392 mmol) was dissolved in anhydrous DMF (1 mL) and DIPEA (78 mg, 0.6 mmol). HATU (140 mg, 0.366 mmol) was added, and the reaction was stirred for 30 minutes. It was then purified by RPLC (50 g column, 10 to 80 % acetonitrile and water). Yield 159.1 mg, 66.1 %. Ion found by LCMS: [M + H] + = 658.4. Step c.

The step-b product (159.1 mg, 0.242 mmol) was dissolved in MeOH

water bath. It was treated with a solution of LiOH monohydrate (25.4 mg, 0.6 mmol) in water (1 mL). After the reaction stirred for 4 hours, it was acidified with 4N HCl solution in dioxane (0.3 mL) and directly purified by RPLC (50 g, 5 to 60 % acetonitrile and water). Yield 67.1 mg, 44 %. Ion found by LCMS: [M + H]+ = 630.0. Ste d.

The title compound was prepared analogously to Example 101, step-b, using the diacid from step c and Int-7a (described in Example 8, step a). Yield 247.4 mg, 62.4 %. Ions found by LCMS: [(M - 2Boc + 3H)/3] + = 1174, [(M - 3Boc + 3H)/3] + = 1140.7, [(M - 4Boc + 3H)/3] + = 1107.4, [(M - 5Boc + 3H)/3] + = 1074, [(M - 6Boc + 3H)/3] + = 1040.7. Ste e.

The title compound Int-68 was prepared analogously to Example 101, step-c, using the step-d product from this example. Yield 197.5 mg, 76.9 %. Ions found by LCMS: [(M + 4H)/4] + = 680.6, [(M + 4H)/5] + = 544.6, [(M + 6H)/6] + = 454.2, [(M + 7H)/7] + = 389.4, [(M + 8H)/8] + = 340.8. Step a.

To a suspension of (4S)-Boc-amino-L-Proline methyl ester HCl salt (1.222 g, 5 mmol) in DCM (6 mL) and DMF (2 mL) was added benzyl 2-bromoacetate (1.375 g, 6 mmol) and DIPEA (975 mg, 7.5 mmol). The mixture was stirred at room temperature for 3 days. It was then concentrated and purified by RPLC (100 g column, 15 to 75 % acetonitrile and water, using 0.1 % TFA as the modifier). Yield 1.71 g, 87.2 %. Ion found by LCMS: [M + H] + = 393.2. Step b.

The step a product was dissolved in DCM (5 mL) and treated with 4N HCl solution in dioxane (5 mL). The mixture was heated at ~ 40 o C for 30 minutes. It was then concentrated and re-dissolved in acetonitrile and water for lyophilization. Yield 1.6 g as di-HCl salt. Ion found by LCMS: [M + H] + = 293.2. Step c.

To a solution of propargyl-PEG4 acid (520.6 mg, 2 mmol) in anhydrous DMF (3 mL) was added HATU (760.4 mg, 2 mmol). After the solid dissolved, the step b product (804 mg, 2.2 mmol) was added, followed by DIPEA (910 mg, 7 mmol). The resulting mixture was stirred for 1 hour and then directly purified by RPLC (100 g, 15 to 67 % acetonitrile and water). Yield 629 mg, 58.9 %. Ion found by LCMS: [M + H] + = 535.2. Step d.

The step c product (629.1 mg, 1.178 mmol) was dissolved in MeOH (3 mL). This solution was cooled in an ice-water bath then treated with a solution of LiOH monohydrate (126 mg, 3 mmol) in water (3 mL). After the mixture was stirred for 3 hours, it was then acidified by 4N HCl solution in dioxane (1 mL). The organic solvents were removed by rotary evaporation, and the res

(100 g column, 0 to 50 % acetonitrile and water). Yield 478 mg, 94.3 %. Ion found by LCMS: [M + H] + = 431.2 S tep e. Preparation of Int-75

To a mixture of Int-7a (2 g, 1.28 mmol, Example 8, step a) and Z-L-norleucine (407.1 mg, 1.535 mmol) in anhydrous DMF (3 mL) was added HATU (583.6 g, 1.535 mmol) in portions over 10 minutes, followed by DIPEA (390 mg, 3 mmol). After the reaction mixture was stirred for 1 hour, it was poured into water (60 mL). The white solid product was collected by filtration and washed with water. The material was re-dissolved in MeOH (30 mL) and treated with Pd/C (5%) (1 g). The mixture was stirred under hydrogen overnight. Pd/C was then filtered, and the filtrate was concentrated and purified by RPLC (150 g, 40 to 95 % MeOH and water). Yield of Int-75 was 2.02 g, 94.3 %. Ion found by LCMS: [(M– Boc + 2H)/2]+ = 789.2. Step f.

The title compound was prepared analogously to Example 101, step-b, using the step-e and step- d intermediates described in this example. Yield 244.4 mg, 65.3 %. Ion found by LCMS: [(M– 2Boc + 3H)/3]+ = 1183.0, [(M– 3Boc + 3H)/3]+ = 1149.7, [(M– 4Boc + 3H)/3]+ = 1116.4, [(M– 5Boc + 3H)/3]+ = 1083.3, [(M– 6Boc + 3H)/3] + = 1049.2, [(M– 7Boc + 3H)/3] + = 1016.2. .

The title compound Int-69 was prepared analogously to Example 101, step-c, using the step-f product from this example. Yield 177.8 mg, 67.9 %. Ions found by LCMS: [(M + 4H)/4] + = 687.5, [(M + 5H)/5] + = 550.1, [(M + 6H)/6] + = 458.6, [(M + 7H)/7] + = 393.2. Example 105– Synthesis of Int-70

Step a.

To a solution of Z-L-norleucine (1.062 g, 4 mmol) in anhydrous NMP (5 mL) was added (S)- methyl 2-amino-4-((tert-butoxycarbonyl)amino)butanoate HCl salt (1.182 g, 4.4 mmol) and DIPEA (1.14 g, 8.8 mmol). The resulting mixture was treated with HATU (760.4 mg, 2.0 mmol) in portions over 10 minutes, and then stirred for 1 hour. It was then purified by RPLC (100 g column, 20 to 75 % acetonitrile and water). Yield 1.9 g, 99.4 %. Ion found by LCMS: [M - Boc + H] + = 380.2. Step b.

The step-a product (1.9 g, 3.975 mmol) was dissolved in MeOH (5 mL) and THF (10 mL). After the solution was cooled in an ice-water bath, it was treated with a solution of LiOH monohydrate (250 mg, 6 mmol) in water (10 mL). After the reaction was stirred for 30 minutes, DCM (100 mL) was added, and the reaction was continued stirring for 30 minutes. It was then acidified with 4N HCl solution in dioxane (1.5 mL), and diluted with water (30 mL). Two layers were separated, and the aqueous layer was back- extracted with EtOAc (80 mL x 2). The combined organic layers were dried over Na2SO4, concentrated by rotary evaporation, and further dried under high vacuum. Yield 1.72 g, 93 %. Ion found by LCMS: [M - Boc + H] + = 366. Step c.

To a solution of the step-b product (535.9 mg, 1.2 mmol) and PMB heptapeptide (1.06 g, 1 mmol, Example 2) in anhydrous DMF (2 mL) was added HATU (456.2 mg, 1.2 mmol) in portions over 10 minutes, followed by DIPEA (312 mg, 2.4 mmol). After the reaction was stir

by RPLC (100 g, 40 to 95 % MeOH and water). The collected fractions were concentrated by rotary evaporation to a white solid (Ion found by LCMS: [(M -2Boc + 2H)/2] + = 655.6). The material was re- dissolved in MeOH (30 mL) and treated with Pd/C (5%). The mixture was stirred at room temperature overnight. Pd/C was filtered, and the filtrate was concentrated by rotary evaporation to dryness. Yield 1.177 g, 85 % over two steps. Ion found by LCMS: [(M - Boc + 2H)/2] + = 638.4. Step d.

To a solution of the step-b product (475.1 mg, 1.02 mmol) and the step-c product (1.177 g, 0.851 mmol) in anhydrous DMF (2 mL) was added HATU (388 mg, 1.02 mmol) in portions over 10 minutes, followed by DIPEA (260 mg, 2 mmol), After the reaction was stirred for 1 hour, it was purified by RPLC (100 g, 40 to 95 % MeOH and water). The collected fractions were concentrated by rotary evaporation to a white solid (Ion found by LCMS: [(M - 2Boc + 2H)/2] + = 821.1). The material was re-dissolved in MeOH (30 mL) and treated with Pd/C (5%). The mixture was stirred at room temperature overnight. Pd/C was filtered, and the filtrate was concentrated by rotary evaporation to dryness. Yield 1.196 g, 83.3 % over two steps. Ion found by LCMS: [(M - Boc + 2H)/2] + = 795. Ste e.

To a mixture of diacid (40.4 mg, 0.1 mmol, described in Example 67, step i) and the step-d product from this example (371.6 mg, 0.22 mmol) in anhydrous DMF (2 mL) was added HATU (76 mg, 0.2 mmol) and DIPEA (52 mg, 0.4 mmol). The resulting mixture was stirred for 30 minutes and was purified by RPLC (50 g, column 40 to 95 % MeOH and water, using 0.1 % T

fractions were concentrated to a white solid (Ions found by LCMS: [(M– 2Boc + 3H)/3] + = 1182.0, [(M– 4Boc + 3H)/3] + = 1116.0, [(M– 5Boc + 3H)/3] + = 1083. The material was re-dissolved in TFA (0.5 mL) and DCM (~ 1 mL). The solution was stirred for 30 minutes and directly purified through RPLC (50 g column, 3 t o 50% acetonitrile and water, using 0.1% TFA as modifier). Yield of Int-70 was 102.4 mg, 25.6 % over two steps. Ions found by LCMS: [(M + 5H)/5] + = 549.6, [(M + 6H)/6] + = 458.2, [(M + 7H)/7] + = 393.0. Example 106– Synthesis of Int-71

.

The compound was prepared analogously to Example 101, step-b, using intermediates from Example 105, step-d, and Example 104, step-d. Yield 171 mg, 45.3%. Ions found by LCMS: [(M– 2Boc + 3H)/3] + = 1191.0, [(M– 3Boc + 3H)/3] + = 1157.6, [(M– 5Boc + 3H)/3] + = 1091.2. .

The title compound Int-71 was prepared analogously to Example 101, step-d, using the step-a intermediate described in this example. Yield 147 mg, 81.6 %. Ions found by LCMS: [(M + 5H)/5] + = 554.9, [(M + 6H)/6] + = 462.6, [(M + 7H)/7] + = 396

A solid mixture of norleucine methyl ester HCI salt (2.1 10 g, 1 1 .62 mmol), racemic trans-4- oxocyclopentane-1 ,2-dicarboxylic acid (1 .000 g, 5.81 mmol), EDCI (2.783 g, 14.52 mmol), HOAt (1 .976 g, 14.52 mmol) and NaHCC (1 .952 g, 23.23 mmol) were mixed in 20 mL of DCM/DMF (5:1 ). The mixture was stirred for overnight (-15 hours) at room temperature. LC/MS analysis showed the completion of the reaction. Two diastereomers were separated by reversed phase HPLC. The polar diastereomer and less polar diastereomer have the retention time at 4.83 min. and 5.04 min. respectively with 8-minute 5-95% ACN/water method: (M + H) + = 427.0 and M - H + = 425.0. The reaction mixture was diluted with EtOAc (200 mL) and washed with 1 N aq. HCI, saturated NaHCCb and brine respectively. The solution was dried with anhy. Sodium sulfate and the solvents were removed. The residue was dissolved in minimum amount of NMP and purified by RPLC using an Isco CombiFlash liquid chromatograph eluted with 30 - 60% acetonitrile and water. The pure polar diastereomer dimethyl ester, 1 .071 g, was obtained in 46% yield (50% in theory). The absolute stereochemistry of the polar and less po

by X-ray. The polar isomer was used in the next step. Step b.

The polar dimethyl ester (1.071 g, 2.511 mmol) was treated with LiOH (0.1302 g, 5.273 mmol) in 20 mL of THF/Water (1:1). Stirred for less than 1 hrs.1N HCl to acidify. Removed THF and extracted with EtOAc to give diacid (1.000 g, 100%). Positive mass ions was found as (M + H + ): 399.0, tr = 1.3 minutes with 5-minute 40~95% ACN/water method.

A solid mixture of diacid (1.000 g, 2.510 mmol), Dab( ^-Boc) methyl ester HCl salt (1.349 g, 5.020 mmol), EDCI (1.203 g, 6.274 mmol), HOAt (0.8540 g, 6.274 mmol) and NaHCO3 (0.8424 g, 10.04 mmol) were mixed in 20 mL of DCM/DMF (5:1). The mixture was stirred for overnight (~15 hours) at room temperature. LC/MS analysis showed the completion of the reaction. The reaction mixture was diluted with EtOAc (200 mL) and washed with 1 N aq. HCl, saturated NaHCO3 and brine respectively. The solution was dried with anhy. Sodium sulfate and the solvents were removed. The residue was dissolved in minimum amount of NMP and purified by RPLC using an Isco CombiFlash liquid chromatograph eluted with 30 - 60% acetonitrile and water. The cyclopentanonyl central linker, 1.760 g, was obtained in 85% yield. Ions found by LCMS: (M– 2Boc + 2H + )/2 = 314.0. Step c.

A mixture of cyclopentanonyl central linker (0.8360 g, 1.011 mmol), aminooxy-PEG4-propargyl (0.500 g, 2.022 mmol), and potassium acetate (0.2977 g, 3.033 mmol) were dissolved in 20 mL of methanol/water (5:1). The mixture was stirred for overnight (~15 hours) at 6

the completion of the reaction. Most of the solvents of the reaction mixture was removed by reduced pressure rotovap to give a viscous oily residue. The residue was dissolved in minimum amount of NMP and purified by RPLC using an Isco CombiFlash liquid chromatograph eluted with 30 - 60% acetonitrile and water. The pure PEG4-oxime central linker dimethyl ester, 1.0457 g, was obtained in 98% yield. The dimethyl ester has the retention time at 5.47 min. with 8-minute 5~95% ACN/water method: (M– 2Boc + 2H + )/2 = 428.6.

The dimethyl ester (1.0457 g, 0.9900 mmol) was treated with LiOH (0.0.0513 g, 2.079 mmol) in 10 mL of THF/Water (1:1). Stirred for less than 1 hrs.1N HCl to acidify. Removed THF and extracted with EtOAc to give PEG4-oxime central linker (1.011 g, 99%). Positive mass ions was found as (M -2Boc + 2H + )/2 = 414.6, tr = 4.50 minutes with 8-minute 5~95% ACN/water method. .

A solid mixture of Int-7a (0.6631 g, 0.4863 mmol, Example 8, step a), PEG4-oxime central linker (0.250 g, 0.2432 mmol), EDCI (0.1165 g, 0.6079 mmol) and HOAt (0.0827 g, 0.608 mmol) were dissolved in 5 mL of dry DMF immediately followed by adding triethylamine (0.17 mL, 0.97 mmol). The mixture was stirred for overnight (~15 hours) at room temperature. HPLC analysis showed the completion of the reaction. Most of the solvent (DMF) of the reaction mixture was removed by reduced pressure rotovap to give a viscous oily residue. The residue was triturated with methanol. After some crystalline solid started to precipitate the flask was put standstill for more than 3 hours. Simple filtration collected the crystalline white solid with methanol wash (3 x 5 mL). The solid was dried to give 0.675 grams of pure product, 75% isolated yield. The solid (0.440 g) was dissolved with 100% TFA and stirred for 30 minutes. TFA was removed by rotovap. The residue was dissolved in minimum amount of water and purified by RPLC using an Isco CombiFlash liquid chromatograph eluted with 5 % to 35% acetonitrile and water using 0.1 % TFA modifier. The pure fractions were lyophilized to afford the product Int-72 as a white powder (TFA salt, 0.360 g, 65% isolated yield, Positive mass ions were found by LC/MS: (M + 6H + )/6 = 454.0, (M + 5H + )/5 = ( )/ E

Step a.

A mixture of the polar dimethyl ester (from Synthesis of , Step 1, 0.9959 g, 2.335 mmol), aminooxy-PEG4-propargyl (0.6745 g, 2.916 mmol) and acetic acid (0.26 mL, 4.5 mmol) were dissolved in 20 mL of DCM. The mixture was stirred for 1 hour at room temperature followed by addition of sodium triacetatoxyborohydride (0.9509 g, 4.486 mmol). The reaction mixture was stirred for overnight (~15 hours) at room temperature. HPLC analysis showed the completion of the reaction. Positive mass ion was found by LC/MS: (M + H) + = 642.4. Most of the solvents of the reaction mixture was removed by reduced pressure rotovap to give a viscous oily residue. The residue was dissolved in 20 mL of THF/10%N HCO (11) d t t d ith B h d id (071 L 31 l) d dii l th l i (0.54 mL, 3.1 mmol). The mixture was stirred for overnight (~15 hours) at ro

was removed and the residue was extracted with EtOAc. After removal of EtOAc, the residue was dissolved in minimum amount of NMP and purified by RPLC using an Isco CombiFlash liquid

chromatograph eluted with 30 - 60% acetonitrile and water. The pure PEG4-amino cyclopentanyl dimethyl ester, 0.7405 g, was obtained in 65% yield. The dimethyl ester has the retention time at 2.88 min. with 5- minute 40~95% ACN/water method: (M + H) + = 742.4 and M– Boc + H + = 643.0.

The dimethyl ester (0.7405 g, 0.9980 mmol) was treated with LiOH (0.0.0616 g, 2.495 mmol) in 10 mL of THF/Water (1:1). Stirred for less than 1 hrs then acidified with 1N HCl. The solution was concentrated and extracted with EtOAc to give Boc-PEG4-central linker (0.705 g, 99%). Positive mass ions was found as (M + H) + = 714.4, tr = 3.30 minutes with 5-minute 5~95% ACN/water method. Step b.

A solid mixture of Int-5d (0.6544 g, 0.4511 mmol), Boc-PEG4 central linker (0.1610 g, 0.2255 mmol), EDCI (0.1081 g, 0.5636 mmol) and HOAt (0.07675 g, 0.5638 mmol) were dissolved in 5 mL of dry DMF immediately followed by adding triethylamine (0.16 mL, 0.90 mmol). The mixture was stirred for overnight (~15 hours) at room temperature. HPLC analysis showed the completion of the reaction. Most of the solvent (DMF) of the reaction mixture was removed by reduced pressure rotovap to give a viscous oily residue. To this residue methanol was added with gentle stirring to triturate the reaction product. After some crystalline solid started to precipitate the flask was put standstill for more than 3 hours. Simple filtration collected the crystalline white solid with methanol wash (3 x 5 mL). The solid was dried to give 0.607 grams of pure product, 75% isolated yield. Positive ions found: (M– 3Boc +3 H + )/3 = 1093.8. The solid (0.440 g) was dissolved with 100% TFA and stirred for 30 minutes. TFA was removed by rotovap. The residue was dissolved in minimum amount of water and purified by RPLC using an Isco CombiFlash liquid chromatograph eluted with 5 % to 35% acetonitrile and water using 0.1 % TFA modifier. The pure fractions were lyophilized to afford the desired product Int-73 as a white powder (TFA salt, 0.670 g, 87% isolated yield). Positive ions found by LC/MS: (M + 7H + )/7 = 383.4, (M + 6H

536.2, (M + 4H + )/4 = 670.1. Example 109– Synthesis of conjugate 27

A solution of azido-hIgG1(Myc)Fc (50 mg, 0.85 µmol, 15.9 mg/mL, MW=58,743 Da, DAR = 3.3) in pH 7.4 PBS was treated with freshly prepared PBS 7.4 solutions of alkyne derivatized small molecule (37.0 mg, 25.5 µmol, Example 38), THPTA(5.5 mg, 12.8 µmol, tris(3-hydroxypropyltriazolylmethyl)amine), sodium ascorbate(5.0 mg, 25.5 µmol), and CuSO4 (2.0 mg, 12.8 µmol). The resulting solution was agitated on a rocker table overnight at room temperature, then purified by affinity chromatography over a protein-A column (See Example 166). Maldi TOF analysis of the purified final product gave an average mass of 63,234 Da (DAR = 3.1). Yield 33 mg, 61% yield. Example 110– Synthesis of conjugate 38

A solution of azido-hIgG1(Myc)Fc (30 mg, 0.50 µmol, 15.9 mg/mL, MW=58,743 Da, DAR = 4.0) in pH 7.4 PBS was treated with freshly prepared PBS 7.4 solutions of alkyne derivatized small molecule (52.0 mg, 12.6 µmol, See Example 59, Synthesis of int-24), THPTA(3.3 mg, 7.5 µmol, tris(3- hydroxypropyltriazolylmethyl)amine), sodium ascorbate(3.0 mg, 15.1 µmol), and CuSO 4 (1.2 mg, 7.5 µmol). The resulting solution was agitated on a rocker table overnight at room temperature, then purified by affinity chromatography over a protein-A column (See Example 166). Maldi TOF analysis of the purified final product gave an average mass of 67,320 Da (DAR = 3.8 calculated from the tallest peak). Yield 17.8 mg, 59% yield. Example 111– Synthesis of conjugate 39

A solution of azido-hIgG1(Myc)Fc (30 mg, 0.50 µmol, 15.9 mg/mL, MW=59,641 Da, DAR = 4.0) in pH 7.4 PBS was treated with freshly prepared PBS 7.4 solutions of alkyne derivatized small molecule (43.4 mg, 12.6 µmol, See Example 60), THPTA(3.3 mg, 7.5 µmol, tris(3- hydroxypropyltriazolylmethyl)amine), sodium ascorbate(3.0 mg, 15.1 µmol), and CuSO4(1.2 mg, 7.5 µmol). The resulting solution was agitated on a rocker table overnight at room temperature, then purified by protein-A affinity chromatography, followed by size exclusion chromatography (See Example 166 for purification procedure). Maldi TOF analysis of the purified final product gave an average mass of 68,535 Da (DAR = 3.8 calculated from a weighted average of the Maldi peaks). Yield 16.7 mg, 48% yield. Example 112– Synthesis of conjugate 40

A solution of azido-hIgG1(Myc)Fc (30 mg, 0.50 µmol, 15.9 mg/mL, MW=59,641 Da, DAR = 4.0) in pH 7.4 PBS was treated with freshly prepared PBS 7.4 solutions of alkyne derivatized small molecule (46.3 mg, 12.6 µmol, See Example 61), THPTA(3.3 mg, 7.5 µmol, tris(3-hydroxypropyltriazolylmethyl)amine), sodium ascorbate(3.0 mg, 15.1 µmol), and CuSO 4 (1.2 mg, 7.5 µmol). CuSO 4 was added last and caused formation of a cloudy white mixture. The resulting solution was agitated on a rocker table overnight at room f ff f chromatography (See Example 166 for purification procedure). Maldi TO

product gave an average mass of 69,074 Da (DAR = 3.7 calculated from a weighted average of the Maldi peaks). Yield 12.3 mg, 41% yield. Example 113– Synthesis of conjugate 74

A solution of hIgG1(Myc)Fc (30 mg, 0.51 µmol, 15.9 mg/mL, MW=58,270 Da) in pH 7.4 PBS was treated with TCEP (18.5 µL, 0.5 M, 9.3 µmol) at room temperature. LCMS after 1h showed complete reduction of the disulfides. This solution was buffer exchanged with PBS, pH = 7.8, EDTA 2 mM (3x15 mL) using a 10,000 mw cutoff filter (final volume 4 mL). To this was added a freshly prepared solution of azido-PEG-4-bissulfone linker (1.44 mg, 20.6 µmol, in 0.2 mL DMF) resulting in a cloudy solution. Azido-PEG-4-bissulfone linker:

Addition of 1 mL methanol removed the cloudiness. The resulting reaction was agitated overnight on a rocker table at room temperature. The azido Fc was purified by protein-A affinity chromatography (See Example 166). LCMS showed clean incorporation of two bissulfone linker units (mass = 59,050, azide DAR = 2.0).

A solution of cysteine-azido-hIgG1(Myc)Fc (28.6 mg, 0.48 µmol, 11.7 mg/mL, DAR = 2.0) in pH 7.4 PBS was treated with freshly prepared PBS 7.4 solutions of alkyne derivatized small molecule (22.4 mg, 5.8 µmol, See Example 41, INT-21), THPTA(2.1 mg, 4.8 µmol, tris(3- hydroxypropyltriazolylmethyl)amine), sodium ascorbate (2.9 mg, 14.5 µmol), and CuSO4 (0.77 mg, 4.8 µmol). CuSO4 was added last and caused formation of a slightly cloudy solution. The resulting solution was agitated on a rocker table overnight at room temperature, then purified by protein-A affinity chromatography , followed by size exclusion chromatography (See Example 166 for purification procedure). Maldi TOF analysis of the purified final product gave an average mass of 64,485 Da (DAR = 3.7 calculated from a weighted average of the Maldi peaks). Yield 16 mg, 52% yield. Example 114– Synthesis of conjugate 75

A solution of azido-hIgG1(Myc)Fc (30 mg, 0.50 µmol, 15.9 mg/mL, MW=59,641 Da, DAR = 2.8) in pH 7.4 PBS was treated with freshly prepared PBS 7.4 solutions of alkyne derivatized small molecule (15.8 mg, 5.6 µmol, See Example 62), THPTA (3.3 mg, 7.5 µmol, tris(3- hydroxypropyltriazolylmethyl)amine), sodium ascorbate (3.0 mg, 15.1 µmol), and CuSO4 (1.2 mg, 7.5 µmol). CuSO4 was added last and caused formation of a cloudy white mixture. The resulting solution f ff chromatography, followed by size exclusion chromatography (See Example 166 for purification procedure). Maldi TOF analysis of the purified final product gave an average mass of 69,074 Da DAR = 3.7 calculated from a weighted average of the Maldi peaks). Yield 12.3 mg, 41 % yield. Example 115 - Synthesis of conjugate 28

To a room temperature solution of alkyne functionalized small molecule (Example 64, 0.050 mg,

13.5 μιτιοΙ) in pH 7.4 PBS (500 μΙ_), was added in order: a 10 mM solution of THPTA in pH 7.4 PBS (700 μΙ_, 7.0 μιηοΙ), a 20 mM solution of sodium ascorbate in pH 7.4 PBS (700 μΙ_, 14.0 μιηοΙ), and last a 10 mM solution of copper sulfate in water (700 μΙ_, 7.0 μιηοΙ). Upon mixing, a solution of azido- hlgG1 (Myc)Fc (29.4 mg, 0.500 μιτιοΙ, 10 mg/mL, MW = 58,837.00 Da, DAR = 2.7) in pH 7.4 PBS was gently dispersed. The resulting solution was agitated on a rocker table overnight at room temperature, then purified by affinity chromatography over a protein-A column (See Example 166). Maldi TOF analysis of the purified final product gave an average mass of 63,812 Da (DAR = 2.4). Yield 10.35 mg, 52% yield. Example 116 - Synthesis of conjugate 29

To a room temperature solution of material described in Example 63 (0.047 mg, 13.5 μιηοΙ) in pH 7.4 PBS (500 μΙ_), was added in order: a 10 mM solution of THPTA in pH 7.4 PBS (700 μΙ_, 7.0 μιτιοΙ), a 20 mM solution of sodium ascorbate in pH 7.4 PBS (700 μΙ_, 14.0 μmol), and last a 10 mM solution of copper sulfate in water (700 μΙ_, 7.0 μιηοΙ). Upon mixing, a solution of azido-hlgG1 (Myc)Fc (29.4 mg, 0.500 μιτιοΙ, 10 mg/mL, MW = 58,837.00 Da, DAR = 2.7) in pH 7.4 PBS was gently dispersed. The resulting solution was agitated on a rocker table overnight at room temperature, then purified by affinity chromatography over a protein-A column (See Example 166). Maldi TOF analysis of the purified final product gave an average mass of 60,951 Da (DAR = 1 .6). Yield 1 5.75 mg, 78% yield. Example 117 - Synthesis of conjugate 55

To a room temperature solution of alkyne functionalized small molecule (Example 65, 0.054 mg,

13.6 μιτιοΙ) in pH 7.4 PBS (500 μΙ_), was added in order: a 20 mM solution of THPTA in pH 7.4 PBS (340 μΙ_, 6.8 μιηοΙ), a 20 mM solution of sodium ascorbate in pH 7.4 PBS (2,720 μΙ_, 54.4 μιτιοΙ), and last a 20 mM solution of copper sulfate in water (340 μL, 6.8 μιηοΙ). Upon mixing, the solution of azido- hlgG1 (Myc)Fc (29.4 mg, 0.340 μιτιοΙ, 10 mg/mL, MW = 58,821 .00 Da, DAR = 2) in pH 7.4 PBS was gently dispersed. The resulting solution was agitated on a rocker table overnight at room temperature, then purified by affinity chromatography over a protein-A column (See Example 166). Maldi TOF analysis of the purified final product gave an average mass of 61 ,939Da (DAR = 1 .1 ). Yield 5.92 mg, 30% yield. Example 118 - Synthesis of conjugate 76

To a room temperature solution of alkyne functionalized small molecule (Example 66, 0.056 mg, 10.2 μιτιοΙ) in pH 7.4 PBS (500 μΐ), it was added in the order: a 20 mM solution of THPTA in pH 7.4 PBS (340 μL, 6.8 μmol), a 20 mM solution of sodium ascorbate in pH 7.4 PBS (680 μΐ, 13.6 μιτιοΙ), and last a 20 mM solution of copper sulfate in water (340 μί, 6.8 μιηοΙ). Upon mixing, a solution of azido- hIgG1(Myc)Fc (0.020 mg, 0.340 µmol, 10 mg/mL, MW = 58,833.00 Da, DA

gently dispensed. The resulting solution was agitated on a rocker table overnight at room temperature, then purified by affinity chromatography over a protein-A column (See Example 166). Maldi TOF analysis of the purified final product gave an average mass of 63,911 Da (DAR = 1.7). Yield 10.81 mg, 54% yield. Example 119– Synthesis of conjugate 69

A solution of azido-hIgG1(Myc)Fc in PBS buffer (20 mg, 0.34 µmol, 1.40 mL, MW=58,721 Da, DAR=2.7) was treated with alkyne derivatized small molecule (TFA salt, 36 mg, 9.2 µmol, Example 67) and freshly prepared pH 7.4 PBS solutions of CuSO 4 (0.7 mL of 10.0 mM, 20 eq), tris(3- hydroxypropyltriazolylmethyl)-amine (THPTA, 0.7 mL of 10.0 mM, 20 eq), and sodium ascorbate (1.02 mL of 10.0 mM, 30eq). The resulting solution was agitated gently for 12 hours at room temperature. The reaction mixture was purified by affinity chromatography over a protein A column, followed size exclusion chromatography (See Example 166). MALDI TOF analysis of the purified final product gave an average mass of 64,845 Da (DAR = 2.3). Yield 9.36 mg, 67% yield. Example 120– Synthesis of conjugate 34

A solution of azido-hIgG1(Myc)Fc in PBS buffer (30 mg, 0.50 µmol, 2.0 mL, MW=58,548 Da, DAR=2.7) was treated with alkyne derivatized small molecule (TFA salt, 54 mg, 13.5 µmol, Example 68) and freshly prepared pH 7.4 PBS solutions of CuSO4 (1.0 mL of 10.0 mM, 20 eq), tris(3- hydroxypropyltriazolylmethyl)-amine (THPTA, 1.0 mL of 10.0 mM, 20 eq), and sodium ascorbate (1.4 mL of 10.0 mM, 30 eq). The resulting solution was agitated gently for 12 hours at room temperature. The reaction mixture was purified by affinity chromatography over a protein A column, followed size exclusion chromatography (See Example 166). MALDI TOF analysis of the purified final product gave an average mass of 64,717 Da (DAR = 2.26). Yield 12.7 mg, 42% yield. Example 121– Synthesis of conjugate 43

A solution of azido-hIgG1(Myc)Fc in PBS buffer (30 mg, 0.50 umol, 2.0 mL, MW=58,935 Da, DAR=5) was treated with alkyne derivatized small molecule (TFA salt, 60 mg, 15 µmol, Example 69) and freshly prepared pH 7.4 PBS solutions of CuSO 4 (1.0 mL of 10.0 mM, 20 eq), tris(3- hydroxypropyltriazolylmethyl)-amine (THPTA, 1.0 mL of 10.0 mM, 20 eq), and sodium ascorbate (1.5 mL of 10.0 mM, 30eq). The resulting solution was agitated gently for 12 hours at room temperature. The reaction mixture was purified by affinity chromatography over a protein A column, followed size exclusion chromatography (See Example 166). MALDI TOF analysis of the purified final product gave an average mass of 70,947 Da (DAR = 4.4). Yield 27 mg, 90% yield Example 122– Synthesis of conjugate 78

A solution of azido-hIgG1(Myc)Fc in PBS buffer (20 mg, 0.34 umol, 1.78 mL, MW=58,833 Da, DAR=3) was treated with alkyne derivatized small molecule (TFA salt, 41 mg, 15 µmol, Example 70) and f S f C SO ( f ) ( hydroxypropyltriazolylmethyl)-amine (THPTA, 0.68 mL of 10.0 mM, 20 eq),

of 10.0 mM, 30eq). The resulting solution was agitated gently for 12 hours at room temperature. The reaction mixture was purified by affinity chromatography over a protein A column, followed size exclusion chromatography (See Example 166). MALDI TOF analysis of the purified final product gave an average mass of 69,330 Da (DAR = 3.3). Yield 10 mg, 50% yield. Example 123– Synthesis of conjugate 31

A solution of azido-hIgG1(Myc)Fc in PBS buffer (30 mg, 0.50 µmol, 2.0 mL, MW=58,743 Da, DAR=3.3) was treated with alkyne derivatized small molecule (TFA salt, 54 mg, 13.5 µmol, Example 71) and freshly prepared pH 7.4 PBS solutions of CuSO4 (1.0 mL of 10.0 mM, 20 eq), tris(3- hydroxypropyltriazolylmethyl)-amine (THPTA, 1.0 mL of 10.0 mM, 20 eq), and sodium ascorbate (1.4 mL of 10.0 mM, 30eq). The resulting solution was agitated gently for 12 hours at room temperature. The reaction mixture was purified by affinity chromatography over a protein A column, followed size exclusion chromatography (See Example 166). MALDI TOF analysis of the purified final product gave an average mass of 64462 Da (DAR = 2.17). Yield 26.1 mg, 65% yield. Example 124– Synthesis of conjugate 33

A solution of azido-hIgG1(Myc)Fc in PBS buffer (30 mg, 0.50 µmol, 2.0 mL, MW=58,548 Da, DAR=2.7) was treated with alkyne derivatized small molecule (TFA salt, 54 mg, 13.5 µmol, Example 72) and freshly prepared pH 7.4 PBS solutions of CuSO4 (1.0 mL of 10.0 mM, 20 eq), tris(3- hydroxypropyltriazolylmethyl)-amine (THPTA, 1.0 mL of 10.0 mM, 20 eq), and sodium ascorbate (1.4 mL of 10.0 mM, 30eq). The resulting solution was agitated gently for 12 hours at room temperature. The reaction mixture was purified by affinity chromatography over a protein A column, followed size exclusion chromatography (See Example 166). MALDI TOF analysis of the purified final product gave an average mass of 64,223 Da (DAR = 2.05). Yield 14.5 mg, 48% yield. Example 125– Synthesis of conjugate 44

A solution of azido-hIgG1(Myc)Fc in PBS buffer (30 mg, 0.50 µmol, 2.0 mL, MW=58,935 Da, DAR=3.88) was treated with alkyne derivatized small molecule (TFA salt, 58 mg, 15 µmol, Example 73) and freshly prepared pH 7.4 PBS solutions of CuSO4 (1.0 mL of 10.0 mM, 20 eq), tris(3- hydroxypropyltriazolylmethyl)-amine (THPTA, 1.0 mL of 10.0 mM, 20 eq), and sodium ascorbate (1.5 mL of 10.0 mM, 30eq). The resulting solution was agitated gently for 12 hours at room temperature. The reaction mixture was purified by affinity chromatography over a protein A column, followed size exclusion chromatography (See Example 166). MALDI TOF analysis of the purified final product gave an average mass of 69,725 Da (DAR = 4). Yield 27 mg, 91% yield. Example 126– Synthesis of conjugate 66

A solution of azido-hIgG1(Myc)Fc in PBS buffer (20 mg, 0.34 µmol, 2.0 mL, MW=58,721 Da, ) ( ) and freshly prepared pH 7.4 PBS solutions of CuSO 4 (0.7 mL of 10.0 mM, 2

hydroxypropyltriazolylmethyl)-amine (THPTA, 0.7 mL of 10.0 mM, 20 eq), and sodium ascorbate (1.0 mL of 10.0 mM, 30eq). The resulting solution was agitated gently for 12 hours at room temperature. The reaction mixture was purified by affinity chromatography over a protein A column, followed size exclusion chromatography (See Example 166). MALDI TOF analysis of the purified final product gave an average mass of 64,614 Da (DAR = 2.3). Yield 15.3 mg, 77% yield. Example 127– Synthesis of conjugate 67

A solution of azido-hIgG1(Myc)Fc in PBS buffer (20 mg, 0.34 µmol, 2.0 mL, MW=58,721 Da, DAR=2.75) was treated with alkyne derivatized small molecule (TFA salt, 34 mg, 9.2 µmol, Example 75) and freshly prepared pH 7.4 PBS solutions of CuSO4 (0.7 mL of 10.0 mM, 20 eq), tris(3- hydroxypropyltriazolylmethyl)-amine (THPTA, 0.7 mL of 10.0 mM, 20 eq), and sodium ascorbate (1.0 mL of 10.0 mM, 30eq). The resulting solution was agitated gently for 12 hours at room temperature. The reaction mixture was purified by affinity chromatography over a protein A column, followed size exclusion chromatography (See Example 166). MALDI TOF analysis of the purified final product gave an average mass of 64,792 Da (DAR = 2.4). Yield 14.4 mg, 72% yield. Example 128– Synthesis of conjugate 68

A solution of azido-hIgG1(Myc)Fc in PBS buffer (20 mg, 0.34 µmol, 1.40 mL, MW=58,721 Da, DAR=2.7) was treated with alkyne derivatized small molecule (TFA salt, 36 mg, 9.2 µmol, Example 76) and freshly prepared pH 7.4 PBS solutions of CuSO 4 (0.7 mL of 10.0 mM, 20 eq), tris(3- hydroxypropyltriazolylmethyl)-amine (THPTA, 0.7 mL of 10.0 mM, 20 eq), and sodium ascorbate (1.02 mL of 10.0 mM, 30eq). The resulting solution was agitated gently for 12 hours at room temperature. The reaction mixture was purified by affinity chromatography over a protein A column, followed size exclusion chromatography (See Example 166). MALDI TOF analysis of the purified final product gave an average mass of 65,317 Da (DAR = 2.4). Yield 13.4 mg, 67% yield. Example 129– Synthesis of conjugate 56

A solution of azido-hIgG1(Myc)Fc in PBS buffer (20 mg, 0.34 µmol, 1.40 mL, MW=58526 Da, DAR=2.59) was treated with alkyne derivatized small molecule (TFA salt, 36 mg, 9.2 µmol, Example 77) and freshly prepared pH 7.4 PBS solutions of CuSO4 (0.7 mL of 10.0 mM, 20 eq), tris(3- hydroxypropyltriazolylmethyl)-amine (THPTA, 0.7 mL of 10.0 mM, 20 eq), and sodium ascorbate (1.02 mL of 10.0 mM, 30eq). The resulting solution was agitated gently for 12 hours at room temperature. The reaction mixture was purified by affinity chromatography over a protein A column, followed size exclusion chromatography (See Example 166). MALDI TOF analysis of the purified final product gave an average mass of 63,622 Da (DAR = 2.21). Yield 19.2 mg, 96% yield. Example 130 - Synthesis of conjugate 79

A solution of azido-hlgG1 (Myc)Fc in PBS buffer (20 mg, 0.34 μιτιοΙ, 1 .40 mL, MW=58833 Da, DAR=3.1 6) was treated with alkyne derivatized small molecule (TFA salt, 43 mg, 10.2 μιτιοΙ, Example 78) and freshly prepared pH 7.4 PBS solutions of CuSCU (0.7 mL of 10.0 mM, 20 eq), tris(3- hydroxypropyltriazolylmethyl)-amine (THPTA, 0.7 mL of 10.0 mM, 20 eq), and sodium ascorbate (1 .02 mL of 10.0 mM, 30eq). The resulting solution was agitated gently for 12 hours at room temperature. The reaction mixture was purified by affinity chromatography over a protein A column, followed size exclusion chromatography (See Example 166). MALDI TOF analysis of the purified final product gave an average mass of 65,326 Da (DAR = 2.1 ). Yieldl 1 .8 mg, 59% yield.

Example 131 - Synthesis of conjugate 32

A solution of azido-hlgG1 (Myc)Fc in PBS buffer (40 mg, 0.67 μιτιοΙ, 2.50 mL, MW=58743 Da, DAR=3.3) was treated with alkyne derivatized small molecule (TFA salt, 46 mg, 9.2 μιτιοΙ, Example 79) and freshly prepared pH 7.4 PBS solutions of CuSCU (1 .4 mL of 10.0 mM, 20 eq), tris(3- hydroxypropyltriazolylmethyl)-amine (THPTA, 1 .4 mL of 10.0 mM, 20 eq), and sodium ascorbate (1 .4 mL of 10.0 mM, 30eq). The resulting solution was agitated gently for 12 hours at room temperature. The reaction mixture was purified by affinity chromatography over a protein A column, followed size exclusion chromatography (See Example 166). MALDI TOF analysis of the purified final product gave an average mass of 62,465 Da (DAR = 2.65). Yield 13.8 mg, 35% yield.

Example 132 - Synthesis of conjugate 45

A solution of azido-hlgG1 (Myc)Fc in PBS buffer (22.5 mg, 0.38 μιτιοΙ, 1 .50 mL, MW=58,835 Da, DAR=5) was treated with alkyne derivatized small molecule (TFA salt, 23 mg, 1 1 μιτιοΙ, Example 80) and freshly prepared pH 7.4 PBS solutions of CuSCU (0.75 mL of 10.0 mM, 20 eq), tris(3- hydroxypropyltriazolylmethyl)-amine (THPTA, 0.75 mL of 10.0 mM, 20 eq), and sodium ascorbate (1 .13 mL of 10.0 mM, 30eq). The resulting solution was agitated gently for 12 hours at room temperature. The reaction mixture was purified by affinity chromatography over a protein A column, followed size exclusion chromatography (See Example 166). MALDI TOF analysis of the purified final product gave an average mass of 65,241 Da (DAR = 4.18). Yield 15 mg, 75% yield.

Example 133 - Synthesis of conjugate 46

A solution of azido-hlgG1 (Myc)Fc in PBS buffer (20 mg, 0.32 μιτιοΙ, 2.50 mL, MW=59641 Da,

DAR=3.9) was treated with alkyne derivatized small molecule (TFA salt, 19 mg, 1 0 μιηοΙ, Example 81 ) and freshly prepared pH 7.4 PBS solutions of CuS0 4 (0.65 mL of 10.0 mM, 20 eq), tris(3- hydroxypropyltriazolylmethyl)-amine (THPTA, 0.65 mL of 10.0 mM, 20 eq), and sodium ascorbate (1 .0 mL of 10.0 mM, 30eq). The resulting solution was agitated gently for 12 hours at room temperature. The reaction mixture was purified by affinity chromatography over a protein A column, followed size exclusion chromatography (See Example 166). MALDI TOF analysis of the purified final product gave an average mass of 64,789 Da (DAR = 4.26). Yield 17.88 mg, 89% yield. Example 134– Synthesis of conjugate 42

A solution was freshly prepared with CuSO4 (10 mM, 16.07 mg), sodium ascorbate (20 mM, 39.7 mg) and tris(3-hydroxypropyltriazolylmethyl)amine) (10 mM, 43.86 mg) in PBS 7.4 buffer solution of (10 mL).

A solution of azido-hIgG1(Myc)Fc (40 mg, 0.679 µmol, 15.0 mg/mL, MW=58,935 Da, DAR = 5.0) in pH 7.4 PBS was treated with freshly prepared PBS 7.4 solutions (1.68 mL) of alkyne derivatized small molecule TFA salt (97.0 mg, 27.1 µmol Example 84). To this solution was added a freshly prepared mixed solution of CuSO 4 [(10 mM), sodium ascorbate (20 mM) and tris(3-hydroxypropyltriazolylmethyl)amine) (10 mM), 1.4 mL]. The resulting solution was gently agitated on a rocker table at room temperature for 3 hours then another 0.5 mL of the mixed solution [CuSO4 (10 mM), sodium ascorbate (20 mM) and tris(3- hydroxypropyltriazolylmethyl)amine) (10 mM)]. The resulting solution was continued to agitate overnight then purified by affinity chromatography over a protein-A column. See Example 166 for purification procedure. Maldi TOF analysis of the purified final product gave an average mass of 69,638 Da (DAR = 4.0). Yield 37.53 mg, 79% yield. Example 135– Synthesis of conjugate 53

A solution was freshly prepared with CuSO4 (10 mM, 15.96 mg), sodium ascorbate (20 mM, 39.6 mg) and tris(3-hydroxypropyltriazolylmethyl)amine) (10 mM, 43.86 mg) in PBS 7.4 buffer solution of (10 mL).

A solution of azido-hIgG1(Myc)Fc (20 mg, 1.43 mL, 0.342 µmol, 14.0 mg/mL, MW=58,526 Da, DAR = 2.6) in pH 7.4 PBS was diluted with PBS 7.4 solutions (0.57 mL) to make a 10.0 mg/mL solution of (2 mL).

A solution of alkyne derivatized small molecule TFA salt (27.5 mg, 7.08 µmol, Example 88) in PBS 7.4 solutions (1.5 mL) was prepared in a 15 mL centrifuge tube. To this solution was added A solution of (0.68 mL, 10 mM of CuSO4, 20 mM of sodium ascorbate and 10 mM tris(3- hydroxypropyltriazolylmethyl)amine)) followed by addition of a solution of azido-hIgG1(Myc)Fc (20 mg, 2.0 mL, 0.342 µmol, 10.0 mg/mL, MW=58,526 Da, DAR = 2.59) in pH 7.4 PBS. The resulting solution was gently agitated on a rocker table at room temperature overnight then purified by affinity chromatography over a protein-A column. See Example 166 for purification procedure. Maldi TOF analysis of the purified final product gave an average mass of 65,320 Da (DAR = 2.5). Yield 19.15 mg, 87% yield. Example 136– Synthesis of conjugate 54

A solution was freshly prepared with CuSO4 (10 mM, 15.96 mg), sodium ascorbate (20 mM, 39.6 mg) and tris(3-hydroxypropyltriazolylmethyl)amine) (10 mM, 43.86 mg) in PBS 7.4 buffer solution of (10 mL).

A solution of azido-hIgG1(Myc)Fc (20 mg, 1.43 mL, 0.342 µmol, 14.0 mg/mL, MW=58,526 Da, DAR = 2.59) in pH 7.4 PBS was diluted with PBS 7.4 solutions (0.57 mL) to make a 10.0 mg/mL solution of (2mL) A solution of alkyne derivatized small molecule TFA salt (25.7 mg,

PBS 7.4 solutions (1.5 mL) was prepared in a 15 mL centrifuge tube. To this solution was added A solution of (0.68 mL, 10 mM of CuSO4, 20 mM of sodium ascorbate and 10 mM tris(3- hydroxypropyltriazolylmethyl)amine)) followed by addition of a solution of azido-hIgG1(Myc)Fc (20 mg, 2.0 mL, 0.342 µmol, 10.0 mg/mL, MW=58,526 Da, DAR = 2.59) in pH 7.4 PBS. The resulting solution was gently agitated on a rocker table at room temperature overnight then purified by affinity chromatography over a protein-A column. See Example 166 for purification procedure. Maldi TOF analysis of the purified final product gave an average mass of 64,708 Da (DAR = 2.3). Yield 19.32 mg, 88% yield. Example 137– Synthesis of conjugate 62

A solution was freshly prepared with CuSO4 (10 mM, 15.96 mg), sodium ascorbate (20 mM, 39.6 mg) and tris(3-hydroxypropyltriazolylmethyl)amine) (10 mM, 43.86 mg) in PBS 7.4 buffer solution of (10 mL).

A solution of azido-hIgG1(Myc)Fc (20 mg, 1.43 mL, 0.342 µmol, 14.0 mg/mL, MW=58,721 Da, DAR = 2.75) in pH 7.4 PBS was diluted with PBS 7.4 solutions (0.57 mL) to make a 10.0 mg/mL solution of (2 mL).

A solution of alkyne derivatized small molecule TFA salt (36.1 mg, 9.36 µmol, Example 89) in PBS 7.4 solutions (1.5 mL) was prepared in a 15 mL centrifuge tube. To this solution was added A solution of (0.68 mL, 10 mM of CuSO4, 20 mM of sodium ascorbate and 10 mM tris(3- hydroxypropyltriazolylmethyl)amine)) followed by addition of a solution of azido-hIgG1(Myc)Fc (20 mg, 2.0 mL, 0.342 µmol, 10.0 mg/mL, MW=58,721 Da, DAR = 2.75) in pH 7.4 PBS. The resulting solution was gently agitated on a rocker table at room temperature overnight then purified by affinity chromatography over a Superdex200 column. See Example 166 for purification procedure. Maldi TOF analysis of the purified final product gave an average mass of 66,835 Da (DAR = 3.0). Yield 9.86 mg, 41% yield. Example 138– Synthesis of conjugate 63

A solution was freshly prepared with CuSO4 (10 mM, 15.96 mg), sodium ascorbate (20 mM, 39.6 mg) and tris(3-hydroxypropyltriazolylmethyl)amine) (10 mM, 43.86 mg) in PBS 7.4 buffer solution of (10 mL).

A solution of azido-hIgG1(Myc)Fc (20 mg, 1.43 mL, 0.342 µmol, 14.0 mg/mL, MW=58,721 Da, DAR = 2.75) in pH 7.4 PBS was diluted with PBS 7.4 solutions (0.57 mL) to make a 10.0 mg/mL solution of (2 mL).

A solution of alkyne derivatized small molecule TFA salt (32.8 mg, 9.2 µmol, Example 86) in PBS 7.4 solutions (1.5 mL) was prepared in a 15 mL centrifuge tube. To this solution was added A solution of (0.68 mL, 10 mM of CuSO4, 20 mM of sodium ascorbate and 10 mM tris(3- hydroxypropyltriazolylmethyl)amine)) followed by addition of a solution of azido-hIgG1(Myc)Fc (20 mg, 2.0 mL, 0.342 µmol, 10.0 mg/mL, MW=58,721 Da, DAR = 2.75) in pH 7.4 PBS. The resulting solution was gently agitated on a rocker table at room temperature overnight then purified by protein-A affinity chromatography. See Example 166 for purification procedure. Maldi TOF a

product gave an average mass of 66,609 Da (DAR = 2.9). Yield 9.55 mg, 41% yield. Example 139– Synthesis of conjugate 64

A solution was freshly prepared with CuSO 4 (10 mM, 15.96 mg), sodium ascorbate (20 mM, 39.6 mg) and tris(3-hydroxypropyltriazolylmethyl)amine) (10 mM, 43.86 mg) in PBS 7.4 buffer solution of (10 mL).

A solution of azido-hIgG1(Myc)Fc (20 mg, 1.43 mL, 0.342 µmol, 14.0 mg/mL, MW=58,721 Da, DAR = 2.75) in pH 7.4 PBS was diluted with PBS 7.4 solutions (0.57 mL) to make a 10.0 mg/mL solution of (2 mL).

A solution of alkyne derivatized small molecule TFA salt (35.9 mg, 9.37 µmol, Example 85) in PBS 7.4 solutions (1.5 mL) was prepared in a 15 mL centrifuge tube. To this solution was added A solution of (0.68 mL, 10 mM of CuSO 4 , 20 mM of sodium ascorbate and 10 mM tris(3- hydroxypropyltriazolylmethyl)amine)) followed by addition of a solution of azido-hIgG1(Myc)Fc (20 mg, 2.0 mL, 0.342 µmol, 10.0 mg/mL, MW=58,721 Da, DAR = 2.75) in pH 7.4 PBS. The resulting solution was gently agitated on a rocker table at room temperature overnight then purified by affinity chromatography over a Superdex200 column. See Example 166 for purification procedure. Maldi TOF analysis of the purified final product gave an average mass of 66,863 Da (DAR = 3.0). Yield 8.62 mg, 36% yield. Example 140– Synthesis of conjugate 65

A solution was freshly prepared with CuSO 4 (10 mM, 15.96 mg), sodium ascorbate (20 mM, 39.6 mg) and tris(3-hydroxypropyltriazolylmethyl)amine) (10 mM, 43.86 mg) in PBS 7.4 buffer solution of (10 mL).

A solution of azido-hIgG1(Myc)Fc (20 mg, 1.43 mL, 0.342 µmol, 14.0 mg/mL, MW=58,721 Da, DAR = 2.75) in pH 7.4 PBS was diluted with PBS 7.4 solutions (0.57 mL) to make a 10.0 mg/mL solution of (2 mL).

A solution of alkyne derivatized small molecule TFA salt (36.4 mg, 9.37 µmol, Example 82) in PBS 7.4 solutions (1.5 mL) was prepared in a 15 mL centrifuge tube. To this solution was added A solution of (0.68 mL, 10 mM of CuSO 4 , 20 mM of sodium ascorbate and 10 mM tris(3- hydroxypropyltriazolylmethyl)amine)) followed by addition of a solution of azido-hIgG1(Myc)Fc (20 mg, 2.0 mL, 0.342 µmol, 10.0 mg/mL, MW=58,721 Da, DAR = 2.75) in pH 7.4 PBS. The resulting solution was gently agitated on a rocker table at room temperature overnight then purified by protein-A affinity chromatography. See Example 166 for purification procedure. Maldi TOF analysis of the purified final product gave an average mass of 65,998 Da (DAR = 2.7). Yield 10.48 mg, 46% yield. Example 141– Synthesis of conjugate 73

A solution was freshly prepared with CuSO 4 (10 mM, 8.2 mg), sodium ascorbate (20 mM, 22.27 mg) and tris(3-hydroxypropyltriazolylmethyl)amine) (10 mM, 22.0 mg) in PBS 7.4 buffer solution of (5 mL). A solution of azido-hIgG1(Myc)Fc (20 mg, 1.43 mL, 0.342 µmol, 14

DAR = 2.75) in pH 7.4 PBS was diluted with PBS 7.4 solutions (0.57 mL) to make a 10.0 mg/mL solution of (2 mL).

A solution of alkyne derivatized small molecule TFA salt (36.96 mg, 9.37 µmol, Example 83) in PBS 7.4 solutions (1.5 mL) was prepared in a 15 mL centrifuge tube. To this solution was added A solution of (0.68 mL, 10 mM of CuSO4, 20 mM of sodium ascorbate and 10 mM tris(3- hydroxypropyltriazolylmethyl)amine)) followed by addition of a solution of azido-hIgG1(Myc)Fc (20 mg, 2.0 mL, 0.342 µmol, 10.0 mg/mL, MW=58,721 Da, DAR = 2.75) in pH 7.4 PBS. The resulting solution was gently agitated on a rocker table at room temperature overnight then purified by affinity chromatography over a protein-A column. See Example 166 for purification procedure. Maldi TOF analysis of the purified final product gave an average mass of 68,785 Da (DAR = 3.5). Yield 16.64 mg, 58% yield. Example 142– Synthesis of conjugate 35

Alkyne derivatized small molecule TFA salt (53 mg, 0.13 mmol, See Example 95) was dissolved in 1 mL of pH 7.4 PBS buffer and added to azido-hIgG1(Myc)Fc (30 mg, 51 µmol, MW=58548, DAR 2.7) in 4 mL of PBS 7.4 buffer followed by (1.02 mL) of a premixed solution which was 20 mM sodium ascorbate (0.02 mmol), 10 mM Copper (II) sulfate (0.01 mmol) and 10 mM tris(3- hydroxypropyltriazolylmethyl)amine (0.01 mmol) and the reaction was shaken gently at ambient temperature for 16 hours. See Example 166 for purification procedure. Yield of 19.9 mg, 60%. MALDI MS analysis showed a range of masses (58264-72608) with an average of mass of 64013. Average DAR=2.4. Example 143– Synthesis of conjugate 36

Alkyne derivatized small molecule TFA salt (49 mg, 0.13 mmol, See Example 93) was dissolved in 1 mL of pH 7.4 PBS buffer and added to azido-hIgG1(Myc)Fc (30 mg, 51 µmol, MW=58548, DAR 2.7) in 4 mL of PBS 7.4 buffer followed by (1.02 mL) of a premixed solution which was 20 mM sodium ascorbate (0.02 mmol), 10 mM Copper (II) sulfate (0.01 mmol) and 10 mM tris(3- hydroxypropyltriazolylmethyl)amine (0.01 mmol) and the reaction was shaken gently at ambient temperature for 16 hours. See Example 166 for purification procedure. Yield of 18.9 mg, 61%. MALDI MS analysis showed a range of masses (58270-72456) with an average of mass of 63816. Average DAR=2.3. Example 144– Synthesis of conjugate 37

Alkyne derivatized small molecule TFA salt (49 mg, 0.13 mmol, See Example 94) was dissolved in 1 mL of pH 7.4 PBS buffer and added to azido-hIgG1(Myc)Fc (30 mg, 51 µmol, MW=58548, DAR 2.7) in 4 mL PBS 7.4 buffer followed by (1.02 mL) of a premixed solution which was 20 mM sodium ascorbate (0.02 mmol), 10 mM Copper (II) sulfate (0.01 mmol) and 10 mM tris(3-hydroxypropyltriazolylmethyl)amine (0.01 mmol) and the reaction was shaken gently at ambient temperature for 16 hours. See Example 166 for purification procedure. Yield of 23.4 mg, 71%. MALDI MS analysis show

(58273-72500) with an average of mass of 63921. Average DAR=2.1. Example 145– Synthesis of conjugate 49

A lkyne derivatized small molecule TFA salt (53 mg, 0.0085 mmol, Example 90) was dissolved in 1 mL of pH 7.4 PBS buffer and added to azido-hIgG1(Myc)Fc (20 mg, 34 µmol, MW=58526, DAR 2.6) in 3 mL PBS 7.4 buffer followed by (0.68 mL) of a premixed solution which was 20 mM sodium ascorbate (0.014 mmol), 10 mM Copper (II) sulfate (0.0068 mmol) and 10 mM tris(3- hydroxypropyltriazolylmethyl)amine (0.0068 mmol)and the reaction was shaken gently at ambient temperature for 16 hours. See Example 166 for purification procedure. Yield of 17.9 mg, 81%. MALDI MS analysis showed a range of masses (58344-72906) with an average of mass of 64071. Average DAR=2.4. Example 146– Synthesis of conjugate 50

Alkyne derivatized small molecule TFA salt (31 mg, 0.0085 mmol, See Example 97) was dissolved in 1 mL of pH 7.4 PBS buffer and added to azido-hIgG1(Myc)Fc (20 mg, 34 µmol, MW=58526, DAR 2.6) in 3 mL PBS 7.4 buffer followed by (0.68 mL) of a premixed solution which was 20 mM sodium ascorbate (0.014 mmol), 10 mM Copper (II) sulfate (0.0068 mmol) and 10 mM tris(3- hydroxypropyltriazolylmethyl)amine (0.0068 mmol)and the reaction was shaken gently at ambient temperature for 16 hours. See Example 166 for purification procedure. Yield of 20.6 mg, 94%. MALDI MS analysis showed a range of masses (58346-72992) with an average of mass of 64001. Average DAR=2.5. Example 147– Synthesis of conjugate 51

A lkyne derivatized small molecule TFA salt (33 mg, 0.0085 mmol, See Example 96) was dissolved in 1 mL of pH 7.4 PBS buffer and added to azido-hIgG1(Myc)Fc (20 mg, 34 µmol, MW=58526, DAR 2.6) in 3 mL PBS 7.4 buffer followed by (0.68 mL) of a premixed solution which was 20 mM sodium ascorbate (0.014 mmol), 10 mM Copper (II) sulfate (0.0068 mmol) and 10 mM tris(3- hydroxypropyltriazolylmethyl)amine (0.0068 mmol)and the reaction was shaken gently at ambient temperature for 16 hours. See Example 166 for purification procedure. Yield of 19.3 mg, 88%. MALDI MS analysis showed a range of masses (58289-72667) with an average of mass of 64060. Average DAR=2.6. Example 148– Synthesis of conjugate 57

Alkyne derivatized small molecule TFA salt (33 mg, 0.0085 mmol, See Example 91) was dissolved in 1 mL of pH 7.4 PBS buffer and added to azido-hIgG1(Myc)Fc (20 mg, 34 µmol, MW=58526, DAR 2.6) in 3 mL PBS 7.4 buffer followed by (0.68 mL) of a premixed solution which was 20 mM sodium ascorbate (0.014 mmol), 10 mM Copper (II) sulfate (0.0068 mmol) and 10 mM tris(3- ) ( ) temperature for 16 hours. See Example 166 for purification procedure. Yie

MS analysis showed a range of masses (58980-76291) with an average of mass of 64526. Average DAR=3.0. Example 149– Synthesis of conjugate 61

Alkyne derivatized small molecule TFA salt (32 mg, 0.0092 mmol, See Example 98) was dissolved in 1 mL of pH 7.4 PBS buffer and added to azido-hIgG1(Myc)Fc (20 mg, 34 µmol, MW=58721, DAR 2.8) in 3 mL PBS 7.4 buffer followed by (0.68 mL) of a premixed solution which was 20 mM sodium ascorbate (0.014 mmol), 10 mM Copper (II) sulfate (0.0068 mmol) and 10 mM tris(3- hydroxypropyltriazolylmethyl)amine (0.0068 mmol)and the reaction was shaken gently at ambient temperature for 16 hours. See Example 166 for purification procedure. Yield of 9.86 mg, 42%. MALDI MS analysis showed a range of masses (58258-75079) with an average of mass of 66516. Average DAR=2.9. Example 150– Synthesis of conjugate 72

Alkyne derivatized small molecule TFA salt (34 mg, 0.0092 mmol, See Example 99) was dissolved in 1 mL of pH 7.4 PBS buffer and added to azido-hIgG1(Myc)Fc (20 mg, 34 µmol, MW=58721, DAR 2.8) in 3 mL PBS 7.4 buffer followed by (0.68 mL) of a premixed solution which was 20 mM sodium ascorbate (0.014 mmol), 10 mM Copper (II) sulfate (0.0068 mmol) and 10 mM tris(3- hydroxypropyltriazolylmethyl)amine (0.0068 mmol)and the reaction was shaken gently at ambient temperature for 16 hours. See Example 166 for purification procedure. Yield of 14.7 mg mg, 63%. MALDI MS analysis showed a range of masses (58687-76383) with an average of mass of 67633. Average DAR=3.4. Example 151– Synthesis of conjugate 77

Alkyne derivatized small molecule TFA salt (34 mg, 0.0092 mmol, See Example 92) was dissolved in 1 mL of pH 7.4 PBS buffer and added to azido-hIgG1(Myc)Fc (20 mg, 34 µmol, MW=58721, DAR 2.8) in 3 mL PBS 7.4 buffer followed by (0.68 mL) of a premixed solution which was 20 mM sodium ascorbate (0.015 mmol), 10 mM Copper (II) sulfate (0.0076 mmol) and 10 mM tris(3- hydroxypropyltriazolylmethyl)amine (0.076 mmol) and the reaction was shaken gently at ambient temperature for 16 hours. See Example 166 for purification procedure. Yield of 16.42 mg, 63%. MALDI MS analysis showed a range of masses (58521-76178) with an average of mass of 68384. Average DAR = 3.1. Example 152– Synthesis of conjugate 24

Note: all the solutions were prepared in PBS 7.4 buffer solution. Product described in Example 100 (60 mg, 0.0169 mmol) was added to azido-hIgG1(Myc)Fc (25 mg, 0.42 µmol, 12.4 mg/mL, MW=59,641 Da, DAR = 5) solution, followed by 0.832 mL of 0.01 M CuSO4 solution, 0.832 mL of 0.01 M f room temperature for 4 hours, then treated with an additional amount of fre

(0.01 M, 0.416 mL). The reaction was shaken overnight. Additional CuSO4 solution (0.01 M, 0.416 mL, freshly prepared) was added, and the reaction mixture was shaken for another 2 hours. The resulting solution was purified by affinity chromatography over a protein A column, followed size exclusion chromatography (See Example 166). Maldi TOF analysis of the purified final product gave an average mass of 71,189. Da (DAR = 4.2). Yield 24.5 mg, 99 % yield. Example 153– Synthesis of conjugate 48

Note: All the solutions were prepared in PBS 7.4 buffer solution. The material described in Example 101(16 mg, 0.004043 mmol) was added to azido-hIgG1(Myc)Fc (30 mg, 0.51 µmol, 13.6 mg/mL, MW = 59367 Da, DAR = 4), followed by 1.263 mL of 0.01 M CuSO4, 2.021 mL of 0.01 M sodium ascorbate, and 1.263 mL of 0.01 M THPTA solutions. The reaction was gently shaken at room

temperature overnight. It was then treated with an additional amount of alkyne (48 mg, 0.01213 mmol), followed by 1.263 mL of 0.01 M CuSO4, 2.021 mL of 0.01 M sodium ascorbate, and 1.263 mL of 0.01 M THPTA solutions. The reaction was shaken for 1 more day. The resulting solution was purified by affinity chromatography over a protein A column, followed size exclusion chromatography (See Example 166). Maldi TOF analysis of the purified final product gave an average mass of 68611. Da (DAR = 3.6). Yield 28.56 mg, 95 % yield. Example 154– Synthesis of conjugate 58

Note: All the solutions were prepared in PBS 7.4 buffer solution. To a solution of azido- hIgG1(Myc)Fc (26 mg, 0.44 µmol, 14 mg/mL, MW = 58526 Da, DAR = 2.59) was added product from Example 102 (37.1 mg, 0.009205 mmol, 0.5 mL). After the resulting mixture was gently mixed, it was treated with 1.1106 mL of 0.01 M CuSO4, 1.777 mL of 0.01 M sodium ascorbate, and 1.1106 mL of 0.01 M THPTA solutions. The reaction was gently shaken at room temperature overnight. It was then purified by affinity chromatography over a protein A column, followed size exclusion chromatography (See Example 166). Maldi TOF analysis of the purified final product gave an average mass of 66062. Da (DAR = 2.7). Yield 23.46 mg, 90 % yield Example 155– Synthesis of conjugate 59

The title compound was prepared analogously to Example 155 substituting product from Example 103. Maldi TOF analysis of the purified final product gave an average mass of 66111. Da (DAR = 2.7). Yield 22.72 mg, 84 % yield. Example 156– Synthesis of conjugate 60

The title compound was prepared analogously to Example 155 using product from Example 104. Maldi TOF analysis of the purified final product gave an average mass of 65,962. Da (DAR = 2.6). Yield 16.37 mg, 82 % yield Example 157– Synthesis of conjugate 70

Note: all the solutions were prepared in PBS 7.4 buffer solution. To a solution of product from Example 105(30 mg, 0.007518 mmol, 0.5 mL) was added azido-hIgG1(Myc)Fc (20 mg, 0.34 µmol, 14.3 mg/mL, MW = 58721 Da, DAR = 2.75). The resulting mixture was gently mixed and then treated with 0.3 mL of 0.05 M CuSO 4 and 0.6 mL of 0.05 M sodium ascorbate solutions. After gently mixed, the reaction mixture was treated with 0.3 mL of 0.05 M THPTA solution and gently shaken at room temperature for 4 hours. It was then treated with an additional amount CuSO4 (2 mg, 0.0125 mmol), and the reaction was continued shaken overnight. The resulting solution was purified by affinity chromatography over a protein A column, followed size exclusion chromatography (See Example 166). Maldi TOF analysis of the purified final product gave an average mass of 65391. Da (DAR = 2.5). Yield 5.27 mg, 39 % yield. Example 158– Synthesis of conjugate 71

The title compound was prepared analogously to Example 158 using the product from Example 106. Maldi TOF analysis of the purified final product gave an average mass of 64,733. Da (DAR = 2.2). Yield 4.07 mg, 42 % yield. Example 159– Synthesis of azido-hIgG1(Myc)Fc with specific DAR values

Preparation of solutions of azido-PEG4 NHS ester in DMF/PBS solution:

100.0 mg of Azido-PEG4 NHS ester was dissolved in 1.00 mL of DMF and diluted with 2.57 mL of PBS buffer solution in ice bath to give 0.1 M azido-PEG4 NHS ester solution.1.00 mL of 0.1 M azido-PEG4 NHS ester was diluted to 0.050M PBS buffer solution. A solution of hIgG1(Myc)Fc (described in Example 1) in pH 7.4 PBS solution (2.31 mL, 5.17 µmol, 13.0 mg/mL, MW = 58009 Da) was treated with azido- PEG4-NHS ester as shown in the table below, then agitated gently overnight at RT to give azido- hlgG1(Myc)Fc with specific DAR values. Unreacted azido-PEG4-NHS ester was removed by buffer exchange with a 10,000 MWCO centrifugal filter (6 x 15 mL pH 7.4 PBS). The final average molecular weight of the azido functionalized Fc was measured by MALDI-TOF (Table 7 and FIG.103). The DAR values were calculated with the following formula: DAR (MW of azido-Fc– MW of Fc) ÷ (MW azido PEG4

fragment = 273.3). Using 0 to 20 equivalents of azido PEG4 NHS ester, giv

DAR values.

Table 7.

Example 160– Synthesis of conjugate 30

A solution of hIgG1(Myc)Fc (22.00 mL, 4.68 µmol, 12.3 mg/mL, MW = 57812 Da) in pH 7.4 PBS was treated with azido-PEG4-NHS ester (0.0107 g, 562 µL of 0.050 M solution in DMF/PBS (1:3), 281 ^µmol, 6 eq), then agitated gently overnight at RT. Unreacted azido-PEG4-NHS ester was removed by buffer exchange with a 10,000 MWCO centrifugal filter (6 x 15 mL pH 7.4 PBS). The final volume of the azide functionalized Fc (143-4A-azido-Fc) was 19 mL with concentration at 14.7 mg/mL, MW (MALDI) = 58548, DAR (MALDI) = 2.7. To this azido-Fc PBS buffer solution (0.0294 g, 0.502 µmol, 2.00 mL) were added alkyne derivatized small molecule (TFA salt, 52.3 mg, 13.56 ^µmol, Example 107) and freshly prepared pH 7.4 PBS solutions of CuSO 4 (1.004 mL of 10.0 mM, 20 eq), tris(3- hydroxypropyltriazolylmethyl)-amine (1.004 mL of 10.0 mM, 20 eq), and sodium ascorbate (1.004 mL of 10.0 mM, 20 eq). The resulting solution was agitated gently for 4 hours at room temperature. Another portion of freshly prepared pH 7.4 PBS solutions of CuSO4 (0.500 mL, of 10.0 mM, 10 eq) was added and the reaction mixture was gently agitated for overnight (~ 15 hours) at room temperature, then the final portion of freshly prepared pH 7.4 PBS solutions of CuSO4 (0.500 mL, of 10.0 mM, 10 eq) was added and the reaction mixture was gently agitated for 2 hours. The reaction mixture was purified by affinity chromatography over a protein A column, followed size exclusion chromatography (See Example 166). MALDI TOF analysis of the purified final product gave an average mass of 63146 Da (DAR = 2.0). Yield 25.0 mg, 83% yield. Example 161– Synthesis of conjugate 41

A solution of aglycosylated hIgG1(Myc)Fc (CTP-146-2A), 3.80 mL, 0.603 µmol, 8.8 mg/mL, MW = 54868 Da) in pH 7.4 PBS was treated with azido-PEG4-NHS ester (0.0019 g, 512 µL of 0.010 M solution in DMF/PBS (1:9), 5.12 ^µmol, 8.5 eq), then agitated gently overnight at RT. Unreacted azido-PEG4- NHS ester was removed by buffer exchange with a 10,000 MWCO centrifugal filter (6 x 15 mL pH 7.4 PBS). The final volume of the azide functionalized Fc was 7.5 mL with con

average MW (MALDI) = 55634, DAR (MALDI) = 2.8. To this Fc-azido PBS buffer solution (0.0275 g, 0.495 µmol, 6.40 mL) were added alkyne derivatized small molecule (TFA salt, 53.4 mg, 13.9 ^µmol, Example 41, INT-21) and freshly prepared pH 7.4 PBS solutions of CuSO4 (1.484 mL of 10.0 mM, 30 eq), tris(3-hydroxypropyltriazolylmethyl)-amine (THPTA, 1.484 mL of 10.0 mM, 30 eq), sodium ascorbate (2.226 mL of 10.0 mM, 45 eq). The resulting solution was agitated gently for ~15 hours at room temperature. The reaction mixture was purified by affinity chromatography over a protein A column, followed size exclusion chromatography (See Example 166 for purification procedure). MALDI TOF analysis of the purified final product gave an average mass of 61,747 Da (DAR = 2.3). Yield 17.8 mg, 65% yield. Example 162– Synthesis of conjugate 47

A solution of aglycosylated hIgG1(Myc)Fc (CTP-143-5A), 17.8 mL, 5.20 µmol, 16.9 mg/mL, MW = 57,874 Da) in pH 7.4 PBS was treated with azido-PEG4-NHS ester (0.0019 g, 512 µL of 0.010 M solution in DMF/PBS (1:9), 5.12 ^µmol, 8.5 eq), then agitated gently overnight at RT. Unreacted azido-PEG4- NHS ester was removed by buffer exchange with a 10,000 MWCO centrifugal filter (6 x 15 mL pH 7.4 PBS). The final volume of the azide functionalized Fc was 20 mL with concentration at 14.9 mg/mL, MW (MALDI) = 59303, DAR (MALDI) = 5.2. To this Fc-azido PBS buffer solution (0.0300 g, 0.503 µmol, 2.00 mL) were added alkyne derivatized small molecule (TFA salt, 55.9 mg, 15.1 ^µmol, Example 108) and freshly prepared pH 7.4 PBS solutions of CuSO4 (2.012 mL of 10.0 mM, 40 eq), tris(3- hydroxypropyltriazolylmethyl)-amine (THPTA, 2.012 mL of 10.0 mM, 40 eq), sodium ascorbate (3.018 mL of 10.0 mM, 60 eq). The resulting solution was agitated gently for ~15 hours at room temperature. The reaction mixture was purified by affinity chromatography over a protein A column, followed size exclusion chromatography (See Example 166). MALDI TOF analysis of the purified final product gave an average mass of 69833 Da (DAR = 4.1). Yield 26.5 mg, 88% yield. Example 163– Synthesis of conjugate 52

A solution of aglycosylated BE-0096hIgG1(Myc)Fc (BE-0096, 8.50 mL, 0.603 µmol, 9.73 mg/mL, MW = 54420 Da) in pH 7.4 PBS was treated with azido-PEG4-NHS ester (0.0412 g, 1056 µL of 0.010 M solution in DMF/PBS (1:9), 10.6 ^µmol, 7 eq), then agitated gently overnight at RT. Unreacted azido- PEG4-NHS ester was removed by buffer exchange with a 10,000 MWCO centrifugal filter (4 x 15 mL pH 7.4 PBS). The final volume of the azide functionalized Fc was 5.5 mL with concentration at 15.2 mg/mL, MW (MALDI) = 55409, DAR (MALDI) = 3.6. To this Fc-azido PBS buffer solution (0.0200 g, 0.261 µmol, 1.315 mL) were added alkyne derivatized small molecule (TFA salt, 40.4 mg, 10.5 ^µmol, (C-801, WJ- 052-048)) and freshly prepared pH 7.4 PBS solutions of CuSO4 (0.721 mL of 10.0 mM, 20 eq), tris(3- hydroxypropyltriazolylmethyl)-amine (THPTA, 0.721 mL of 10.0 mM, 20 eq), sodium ascorbate (1.082 mL of 10.0 mM, 30 eq). The resulting solution was agitated gently for ~15 hours at room temperature. The reaction mixture was purified by affinity chromatography over a protein A column, followed size exclusion chromatography (See Example 166). MALDI TOF analysis of the purified fi

mass of 63851 Da (DAR = 3.1). Yield 18.8 mg, 94% yield. Example 164– ADC purification protocols

T he crude mixtures were diluted 1:10 in PBS pH 7.4, and purified using MabSelect Sure Resin (GE Healthcare, Chicago, IL, USA), followed by size exclusion chromatography. Purified material was quantified using a Nanodrop™ UV visible spectrophotomer (using a calculated extinction coefficient based on the amino acid sequence of h-IgG1), and concentrated to approximately 10 mg/mL using a centrifugal concentrator (10,000 MWCO). Purified molecules were analyzed using 4-12% Bis Tris SDS PAGE gels by loading 1-2 µg of each molecule into the gel, and staining using instant Blue staining. Each gel included a molecular weight ladder with the indicated molecular weight standards. Example 165– Efficacy of ADC conjugate 23 in mouse pneumonia model vs. AB5075

The murine pneumonia model was adapted from Lin et al., 2015 (PMID: 26288841). A. baumannii AB5075 was grown to log-phase in LB at 37 °C, 200 rpm shaking. Bacteria were washed once with PBS via centrifugation at 4,000 rpm. Bacteria were adjusted to the desired concentration in PBS.

Prior to infection, 8-week-old female C57Bl/6J mice (Jackson Labs) were anesthetized with 100 mg/kg ketamine and 10 mg/kg xylazine. Using an otoscope, mice were infected non-invasively directly into the trachea.

Mice were infected with 5 × 10 7 CFU in 30 µL of PBS. Mice were treated intraperitoneally with different doses of conjugate 23 or hIgG1 (50 mpk) at t=-12 h pre-infection and t=+1 h post-infection or with colistin (10 mpk) or PBS at t=+1 h, 5 h, 24 h, 36 h, 48 h and 60 h post-infection.

Survival of mice (n=5 per group) was monitored for 5 days. ADC conjugate 23 showed a dose- dependent protection: 0% protection at 1 mpk, 20% protection at 10 mpk and 80% protection at 25 and 50 mpk. Colistin was used as positive control resulting in 80% protection. In comparison, control IgG1 and PBS were not protective and mice succumbed to infection between day 1.5 - 2 (FIG.95). Example 166– Efficacy of ADC conjugate 34 in mouse pneumonia model vs. PAO1

The murine pneumonia model was adapted from Lin et al., 2015 (PMID: 26288841). P.

aeruginosa PAO1 was grown to log-phase in LB at 37 °C, 200 rpm shaking. Bacteria were washed once with PBS via centrifugation at 4,000 rpm. Bacteria were adjusted to the desired concentration in PBS. Prior to infection, 8-week-old female C57Bl/6J mice (Jackson Labs) were anesthetized with 100 mg/kg ketamine and 10 mg/kg xylazine. Using an otoscope, mice were infected non-invasively directly into the trachea.

Mice were infected with 5 × 10 6 CFU in 30 µL of PBS. Mice were treated intraperitoneally with conjugate 34 (25 mpk) or hIgG1 (50 mpk) at t=-12 h prior to infection or with colistin (10 mpk) or PBS at t=+1 h, 5 h, 24 h, 36 h, 48 h and 60 h post-infection.

Survival of mice (n=5 per group) was monitored for 5 days. A single dose of ADC conjugate 34 ( % ) O C positive control resulting in complete protection. In comparison, control IgG

and mice succumbed to infection between day 2– 2.5 (FIG.96). Example 167– Efficacy of ADC conjugate 34 in mouse pneumonia model vs. AB5075

T he murine pneumonia model was adapted from Lin et al., 2015 (PMID: 26288841). A. baumannii AB5075 was grown to log-phase in LB at 37 °C, 200 rpm shaking. Bacteria were washed once with PBS via centrifugation at 4,000 rpm. Bacteria were adjusted to the desired concentration in PBS.

Prior to infection, 8-week-old female C57Bl/6J mice (Jackson Labs) were anesthetized with 100 mg/kg ketamine and 10 mg/kg xylazine. Using an otoscope, mice were infected non-invasively directly into the trachea.

Mice were infected with 5 × 10 7 CFU in 30 µL of PBS. Mice were treated intraperitoneally with conjugate 34 or hIgG1 (50 mpk) at t=-12 h pre-infection.

Survival of mice (n=4 per group) was monitored for 5 days. A single dose of ADC conjugate 34 was protective (75% survival) against a lethal challenge with A. baumannii AB5075. Control IgG1 was not protective and mice succumbed to infection on day 2 (FIG.97). Example 168– Efficacy of ADC conjugate 23 in mouse pneumonia model vs. PAO1

The murine pneumonia model was adapted from Lin et al., 2015 (PMID: 26288841). P.

aeruginosa PAO1 was grown to log-phase in LB at 37 °C, 200 rpm shaking. Bacteria were washed once with PBS via centrifugation at 4,000 rpm. Bacteria were adjusted to desired concentration in PBS.

Prior to infection, 8-week-old female C57Bl/6J mice (Jackson Labs) were anesthetized with 100 mg/kg ketamine and 10 mg/kg xylazine. Using an otoscope, mice were infected non-invasively directly into the trachea.

Mice were infected with 5 × 10 6 CFU in 30 µL of PBS. Mice were treated intraperitoneally with conjugate 23 or hIgG1 (50 mpk) at t=-12 h prior to infection or with colistin (10 mpk) or PBS at t=+1 h and 5 h post-infection.

Survival of mice (n=5 per group) was monitored for 5 days. A single dose of ADC conjugate 23 was completely protective against challenge with P. aeruginosa PAO1. Positive control, colistin, was was also protective (100%). Controls IgG1 (60% survival) and PBS (20% survival) were only partially protective (FIG.98). Example 169– Efficacy of ADC conjugate 23 in mouse pneumonia model vs. PAO1

The murine pneumonia model was adapted from Lin et al., 2015 (PMID: 26288841). P.

aeruginosa PAO1 was grown to log-phase in LB at 37 °C, 200 rpm shaking. Bacteria were washed once with PBS via centrifugation at 4,000 rpm. Bacteria were adjusted to the desired concentration in PBS. Prior to infection, 8-week-old female C57Bl/6J mice (Jackson Labs) were anesthetized with 100 mg/kg ketamine and 10 mg/kg xylazine. Using an otoscope, mice were infected non-invasively directly into the trachea. Mice were infected with 5 × 10 6 CFU in 30 µL of PBS. Mice were tre

conjugate 23, 34, 35, 38, 44 or hIgG1 (25 mpk) at t=-12 h prior to infection or with colistin (10 mpk) or PBS at t=+1 h and 5 h post-infection.

Survival of mice (n=5 per group) was monitored for 5 days. Single doses of ADC conjugates 23, 34, 35, 38 or 44 were completely protective against challenge with P. aeruginosa PAO1. Positive control, colistin, was was also protective (100%). The IgG1 control (60% survival) was only partially protective and mice in the PBS group all succumbed to infection on day 2 (FIG.99). Example 170– Protective effect of Conjugate 23 against endotoxemia

Septic shock, resulting in systemic organ failure, is an important component of many bacterial infections. In infections caused by Gm-negative pathogens, or in some mixed infections, LPS (aka endotoxin) is the dominate mediator of this process which results from hyper-stimulation of the immune system. In a mouse model of endotoxemia we were able to demonstrate the protective effect of

Conjugate 23 to rescue mice given a lethal dose of LPS.

Mouse model of endotoxemia. These studies used female immune competent C57BL/6 mice between 8 and 10 weeks old. After acclimation for 7 days mice were challenged IP with 12.5 mg/kg of purified LPS (Escherichia coli 0111:B4) in PBS from Sigma (#L4130) in a volume of 0.2 ml. Previously this dose was shown to result in 100% mortality of challenged mice within 5 days. Study mice were housed and cared for following standard IACUC approved protocols, and clearly moribund mice were humanely euthanized and scored as a death.

Efficacy of Conjugate 23 in endotoxemia. C57BL/6 mice were challenged with a lethal dose of LPS as described above. Immediately following LPS challenge 2 groups of animals received vehicle (PBS) or colistin (10 mg/kg) as a separate IP injection. Due to the short half-life of colistin both groups were dosed twice daily (bid, separated by 4-6 hours) for 2 days. In contrast, conjugate 23 was administered as a single IP dose at 25 mg/kg 4 hours prior to LPS challenge. The F c -only control for conjugate 23 which lacks the LPS recognition moiety was dosed following the same procedure (hIgG1).

As illustrated in Figure 100, mice dosed with the vehicle or hIgG1 reached 100% mortality within 48 hours of LPS challenge. Mice receiving multiple doses of colistin were partially protected with ~30% of animals reaching death after 5 days. In contrast, all mice treated with conjugate 23 were rescued with 0% mortality. This study demonstrates the LPS neutralizing capabilities of this class of compounds and suggests an additional mechanism of action beyond direct killing of bacteria. Example 171– Conjugate activity in human blood

Overnight stationary-phase cultures were centrifuged at 3,500 x g for 10 min at room temperature and resuspended in 1X PBS to an optical density at 600 nm (OD600) of 0.4 (1 x 10 8 colony forming units [CFU]/ml) in RPMI with L-glutamine and phenol red (Life Technologies). Bacteria were diluted 1:25 in RPMI (4 x 10 6 CFU/ml) and 10 ul added to wells of a non-binding 96-well plate (Corning 3641) containing 80 ul of 50% hirudin anti-coagulated blood (complement active) in RPMI. After adding 5 ul of 20X

( / f ) f f concentration of 40% in 100 ul, the plate was incubated under static conditi

minimum bactericidal concentration (MBC) was determined by 10-fold serial dilution in 1X PBS, then plating 4 ul spots onto rectangular LB agar plates for overnight incubation at 37°C. MBC was defined as the minimum conjugate concentration required for bacterial clearance (Table 8A and Table 8B). An untreated growth control (40% blood, 0 ug/ml conjugate) was included in all assays as a growth comparator.

Table 8A. MBC values of conjugates in bloo

Table 8B. MBC values of conjugates in bloo

Example 172– Conjugate activity in human serum

Overnight stationary-phase cultures were centrifuged at 3,500 x g for 10 min at room temperature and resuspended in 1X PBS to an optical density at 600 nm (OD600) of 0.4 (1 x 108 colony forming units [CFU]/ml) in RPMI with L-glutamine and phenol red (Life Technologies). Bacteria were diluted 1:25 in RPMI (4 x 10 6 CFU/ml) and 10 uL added to wells of a non-binding 96-well plate (Corning 3641) containing 80 ul of 50% normal human serum (complement-active; Innovative Researc

50% heat-inactivated human serum (56°C for 30 min; complement inactive) in RPMI. After adding 5 ul of 20X conjugate diluted in RPMI (128– 2 ug/ml final concentration), and 5 ul RPMI for a final serum concentration of 40% in 100 ul, the plate was incubated under static conditions at 37°C for 90 min. The minimum bactericidal concentration (MBC) was determined by 10-fold serial dilution in 1X PBS, then plating 4 ul spots onto rectangular LB agar plates for overnight incubation at 37°C. MBC was defined as the minimum conjugate concentration required for bacterial clearance (Table 9). An untreated growth control (40% serum, 0 ug/ml conjugate) was included in all assays as a growth comparator. MBCs are typically lower in complement active normal human serum compared to complement inactive heat- inactivated human serum, indicating that conjugate-mediated killing is complement dependent (Table 9).

Table 9. MBC values of conjugates in serum

Example 173– In vitro neutrophil killing assay

5 x 10 4 log phase E. coli ATCC 25922 were added to 96-well, round-bottom, non-treated tissue culture plates in the presence or absence of 25 nM ADC. 5 x 10 4 polymorphprep (AXIS-SHIELD) purified human neutrophils were added to a portion of the wells and plates were incubated at 37°C shaking at 800 rpm for the indicated time points Aliquots were removed at each time point serially diluted in water to lyse neutrophils and incubated overnight on agar plates at 37°C. Surviving

manually counting colonies and data were graphed using the following formula: % of inoculum= ([CFU/mL of test condition] / [CFU/mL of inoculum])*100 (FIG.101). Example 174– Neutrophil phagocytosis of FITC-labeled E. coli (K-12) BioParticles

10 5 FITC-labeled E. coli BioParticles (Invitrogen) were opsonized with 250 nM ADC or media only by co-incubation for 25 min at 37°C shaking (650 rpm).2 x 10 5 polymorphprep purified human neutrophils were then added and plates were incubated at 37°C shaking at 650 rpm for 10 min. Neutrophils were placed on ice for 5 minutes to stop phagocytosis and then washed in cold PBS followed by fixation in 2% PFA. Neutrophils were incubated in 0.2% trypan blue for 2 minutes to quench extracellular fluorescence, washed once and analyzed on a FACSCalibur (BD Biosciences) in the presence of residual trypan blue. Fluorescence was measured in the Blu FL1 channel (FIG.102). Other Embodiments

W hile the disclosure has been described in connection with specific embodiments thereof, it will be understood that it is capable of further modifications and this application is intended to cover any variations, uses, or adaptations of the disclosure following, in general, the principles of the disclosure and including such departures from the present disclosure come within known or customary practice within the art to which the disclosure pertains and may be applied to the essential features hereinbefore set forth.

All publications, patents and patent applications are herein incorporated by reference in their entirety to the same extent as if each individual publication, patent or patent application was specifically and individually indicated to be incorporated by reference in its entirety.

What is claimed is: