Multispecific binding proteins with mutant Fab domains

文档序号:395287 发布日期:2021-12-14 浏览:36次 中文

阅读说明:本技术 具有突变型Fab结构域的多特异性结合蛋白 (Multispecific binding proteins with mutant Fab domains ) 是由 M·阿马拉尔 C·贝尔 I·福肯 G·赫斯勒 S·霍尔珀 J·荣格 C·兰格 W-D· 于 2019-12-23 设计创作,主要内容包括:提供结合蛋白,其包含与VH区配对的VL区和与CL区配对的CH1区,其中所述VL区和所述VH区包含带相反电荷的突变以促进配对,并且其中所述CH1区和所述CL区包含突变以促进配对。还提供结合蛋白,其包含一个或多个半胱氨酸残基,所述半胱氨酸残基被工程化到VH/VL对中以形成一个或多个二硫键。还提供多特异性结合蛋白、编码结合蛋白和多特异性结合蛋白的核酸、表达载体、宿主细胞、药物组合物以及施用本文所述的结合蛋白或多特异性结合蛋白的治疗方法。(Binding proteins are provided comprising a VL region paired with a VH region and a CH1 region paired with a CL region, wherein the VL region and the VH region comprise oppositely charged mutations to facilitate pairing, and wherein the CH1 region and the CL region comprise mutations to facilitate pairing. Also provided are binding proteins comprising one or more cysteine residues engineered into a VH/VL pair to form one or more disulfide bonds. Also provided are multispecific binding proteins, nucleic acids encoding binding proteins and multispecific binding proteins, expression vectors, host cells, pharmaceutical compositions, and therapeutic methods of administering a binding protein or multispecific binding protein described herein.)

1. A multispecific antigen-binding protein comprising at least two VL regions and at least two CH1 regions, the at least two VL regions being paired separately with at least two VH regions to form at least two antigen-binding sites, the at least two CH1 regions being paired separately with two CL regions,

wherein at least one CH1/CL pair comprises a CH1/CL mutation selected from one or more of the following to facilitate pairing:

(1) the T192E (CH1) mutation and the N137K and S114A (CL) mutations, and

(2) the L143Q and S188V (CH1) mutations and the V133T and S176V (CL) mutations, and

(3) the T192E, L143Q and S188V (CH1) mutations and the N137K, S114A, V133T and S176V (CL) mutations,

(4) the K221E (CH1) mutation and the E123K (CL) mutation,

(5) T192E and K221E (CH1) mutations and N137K, S114A and E123K (CL) mutations, (6) L143E, L143D, L143K, L143R or L143H (CH1) mutations and S176E, S176D, S176K, S176R or S176H (CL) mutations,

(7) L124E, L124D, L124K, L124R or L124H (CH1) mutations and V133E, V133D, V133K, V133R or V133H (CL) mutations,

(8) the K228D (CH1) mutation and the D122K (CL) mutation, and

(9) the K221E and K228D (CH1) mutations and the D122K and E123K (CL) mutations,

wherein when two CH1/CL pairs contain a mutation to facilitate pairing of two different VH/VL pairs, the two CH1/CL pairs do not contain the same mutation, and

Wherein at least one VH/VL pair comprises an oppositely charged mutation to facilitate pairing, the oppositely charged mutation comprising (1) a mutant residue at Kabat position 39 in the VH region selected from E, D, K, R or H, and (2) a mutant residue at Kabat position 38 in the VL region selected from E, D, K, R or H, and

wherein a mutated residue in the VH domain has an opposite charge to a mutated residue in the VL domain.

2. A multispecific antigen-binding protein comprising:

a) a first light chain (LC 1)/heavy chain (HC1) pair, the first light chain (LC 1)/heavy chain (HC1) pair comprising:

(1) a first VL region (VL1) that pairs with a first VH region (VH1) to form a first antigen binding site (VL 1);

(2) a first constant heavy chain region 1(CH1-1) operably linked to VH1 and a first constant light chain region (CL1) operably linked to VL 1; and

(3) a first heterodimerization domain (HD 1); and

b) a second light chain (LC 2)/heavy chain (HC2) pair, the second light chain (LC 2)/heavy chain (HC2) pair comprising:

(4) a second VL region (VL2) that pairs with a second VH region (VH2) to form a second antigen binding site;

(5) a second constant heavy chain region 1(CH1-2) operably linked to VH2 and a second constant light chain region (CL2) operably linked to VL 2; and

(6) A second heterodimerization domain (HD 2);

wherein HD1 and HD2 heterodimerize,

wherein at least one or both of the pair VL1 and VH1 and at least one or both of the pair VL2 and VH2 comprise oppositely charged mutations to facilitate pairing, the oppositely charged mutations comprising (1) a mutant residue at Kabat position 39 in the VH region selected from E, D, K, R or H, and (2) a mutant residue at Kabat position 38 in the VL region selected from E, D, K, R or H, and wherein the mutant residue in the VH region has an opposite charge to the mutant residue in the VL region,

wherein at least one or both of the pair CL1 and CH1-1 and at least one or both of the pair CL2 and CH1-2 comprise mutations to facilitate pairing, and

wherein when both of the pair CL1 and CH1-1 and both of the pair CL2 and CH1-2 comprise mutations to facilitate pairing, the mutations that facilitate pairing in CH1-1 and CL1 are different from the mutations that facilitate pairing in CH1-2 and CL 2.

3. The multispecific antigen-binding protein of claim 1, wherein at least one CH1 region is operably linked to a heterodimerization domain.

4. A multispecific antigen-binding protein comprising at least two polypeptide chains and forming at least two antigen-binding sites, wherein one polypeptide chain comprises a structure represented by the formula:

VL1-L1-VL2-L2-CL [I]

And one polypeptide chain comprises a structure represented by the formula:

VH2-L3-VH1-L4-CH1 [II]

wherein:

VL1 is a first immunoglobulin light chain variable domain;

VL2 is a second immunoglobulin light chain variable domain;

VH1 is a first immunoglobulin heavy chain variable domain;

VH2 is a second immunoglobulin heavy chain variable domain;

CL is an immunoglobulin light chain constant domain;

CH1 is an immunoglobulin CH1 heavy chain constant domain; and is

L1, L2, L3 and L4 are amino acid linkers,

wherein VH1 is paired with VL1, VH2 is paired with VL2, and CH1 is paired with CL,

wherein said polypeptide of formula I and said polypeptide of formula II form a cross light-heavy chain pair,

wherein one or more cysteine residues are engineered into one or both of the VH1/VL1 pair and the VH2/VL2 pair to form one or more disulfide bonds,

wherein at least one or both of the pair VL1 and VH1 and at least one or both of the pair VL2 and VH2 comprise oppositely charged mutations that facilitate pairing comprising (1) a mutant residue at Kabat position 39 in the VH region selected from E, D, K, R or H, and (2) a mutant residue at Kabat position 38 in the VL region selected from E, D, K, R or H,

Wherein a mutated residue in the VH region has an opposite charge to a mutated residue in the VL region, and

wherein the CH1 and CL domain pair comprises a mutation that facilitates pairing.

5. A multispecific antigen-binding protein comprising four polypeptide chains forming four antigen-binding sites, wherein each of the two polypeptide chains comprises a structure represented by the formula:

VL1-L1-VL2-L2-CL [I]

and each of the two polypeptide chains comprises a structure represented by the formula:

VH2-L3-VH1-L4-CH1-Fc [II]

wherein:

VL1 is a first immunoglobulin light chain variable domain;

VL2 is a second immunoglobulin light chain variable domain;

VH1 is a first immunoglobulin heavy chain variable domain;

VH2 is a second immunoglobulin heavy chain variable domain;

CL is an immunoglobulin light chain constant domain;

CH1 is an immunoglobulin CH1 heavy chain constant domain;

fc comprises an immunoglobulin hinge region and CH2 and CH3 immunoglobulin heavy chain constant domains; and is

L1, L2, L3 and L4 are amino acid linkers,

wherein VH1 pairs with VL1 to form a first antigen binding site, VH2 pairs with VL2 to form a second antigen binding site, and CH1 pairs with CL,

wherein said polypeptide of formula I and said polypeptide of formula II form a cross light-heavy chain pair,

Wherein one or more cysteine residues are engineered into one or both of the VH1/VL1 pair and the VH2/VL2 pair to form one or more disulfide bonds,

wherein one or both of the pair VL1 and VH1 and one or both of the pair VL2 and VH2 comprise oppositely charged mutations that facilitate pairing, the oppositely charged mutations comprising (1) a mutated residue at Kabat position 39 in the VH region selected from E, D, K, R or H, and (2) a mutated residue at Kabat position 38 in the VL region selected from E, D, K, R or H, and wherein the mutated residue in the VH region has an opposite charge to the mutated residue in the VL region, and

wherein one or both of the CH1 and CL domain pairs comprise a mutation that facilitates pairing, and

wherein when at least two CH1/CL pairs contain mutations to promote pairing, the set of mutations in one CH1/CL pair is different from the set of mutations in the other CH1/CL pair.

6. A multispecific antigen-binding protein comprising four polypeptide chains forming three antigen-binding sites, wherein

The first polypeptide chain comprises a structure represented by the formula:

VL2-L1-VL1-L2-CL1 [I],

the second polypeptide chain comprises a structure represented by the formula:

VH1-L3-VH2-L4-CH 1-1-hinge-CH 2-CH3 [ II ],

The third polypeptide chain comprises a structure represented by the formula:

VH3-CH 1-2-hinge-CH 2-CH3 [ III ], and

the fourth polypeptide chain comprises a structure represented by the formula:

VL3-CL2 [IV],

wherein:

VL1 is a first immunoglobulin light chain variable domain;

VL2 is a second immunoglobulin light chain variable domain;

VL3 is a third immunoglobulin light chain variable domain;

VH1 is a first immunoglobulin heavy chain variable domain;

VH2 is a second immunoglobulin heavy chain variable domain;

VH3 is a third immunoglobulin heavy chain variable domain;

CL1 is a first immunoglobulin light chain constant domain;

CL2 is a second immunoglobulin light chain constant domain;

CH1-1 is a first immunoglobulin CH1 heavy chain constant domain;

CH1-2 is a second immunoglobulin CH1 heavy chain constant domain;

CH2 is an immunoglobulin CH2 heavy chain constant domain;

CH3 is an immunoglobulin CH3 heavy chain constant domain;

a hinge is an immunoglobulin hinge region connecting the CH1 domain and the CH2 domain; and is

L1, L2, L3 and L4 are amino acid linkers,

wherein said polypeptide of formula I and said polypeptide of formula II form a cross light-heavy chain pair, and

wherein one or more cysteine residues are engineered into one or more of the VH1/VL1 pair, the VH2/VL2 pair and the VH3/VL3 pair to form one or more disulfide bonds,

Wherein one or both of the pair VL1 and VH1 and one or both of the pair VL2 and VH2 comprise oppositely charged mutations that facilitate pairing, the oppositely charged mutations comprising (1) a mutated residue at Kabat position 39 in the VH region selected from E, D, K, R or H, and (2) a mutated residue at Kabat position 38 in the VL region selected from E, D, K, R or H, and wherein the mutated residue in the VH region has an opposite charge to the mutated residue in the VL region,

wherein one or both of the pair CL1 and CH1-1 and one or both of the pair CL2 and CH1-2 comprise mutations that promote pairing, and

wherein the mutations in CH1-1 and CL1 are different from the mutations in CH1-2 and CL2 when both of the CL1 and CH1-1 pairs and both of the CL2 and CH1-2 pairs contain mutations to facilitate pairing.

7. A multispecific antigen-binding protein comprising

a) A first light chain (LC 1)/heavy chain (HC1) pair, the first light chain (LC 1)/heavy chain (HC1) pair comprising:

(1) a first VL region (VL1) that pairs with a first VH region (VH1) to form a first antigen binding site (VL 1);

(2) a first constant heavy chain region 1(CH1-1) operably linked to VH1 and a first constant light chain region (CL1) operably linked to VL1, and

(b) A second light chain (LC 2)/heavy chain (HC2) pair, the second light chain (LC 2)/heavy chain (HC2) pair comprising:

(3) a second VL region (VL2) that pairs with a second VH region (VH2) to form a second antigen binding site; and

(4) a second constant heavy chain region 1(CH1-2) operably linked to VH2 and a second constant light chain region (CL2) operably linked to VL2,

wherein the C-terminus of CH1-1 is operably linked to the N-terminus of VH2,

wherein one or more cysteine residues are engineered into one or both of the VH1/VL1 pair and the VH2/VL2 pair to form one or more disulfide bonds,

wherein one or both of the pair VL1 and VH1 and one or both of the pair VL2 and VH2 comprise oppositely charged mutations that facilitate pairing, the oppositely charged mutations comprising (1) a mutated residue at Kabat position 39 in the VH region selected from E, D, K, R or H, and (2) a mutated residue at Kabat position 38 in the VL region selected from E, D, K, R or H, and wherein the mutated residue in the VH region has an opposite charge to the mutated residue in the VL region,

wherein one or both of the pair CL1 and CH1-1 and one or both of the pair CL2 and CH1-2 comprise mutations that promote pairing, and

Wherein the mutations in CH1-1 and CL1 are different from the mutations in CH1-2 and CL2 when both of the CL1 and CH1-1 pairs and both of the CL2 and CH1-2 pairs contain mutations to facilitate pairing.

8. The multispecific antigen-binding protein of any one of claims 2-7, wherein CH1 comprises a T192E mutation, and CL comprises N137K and S114A mutations.

9. The multispecific antigen-binding protein of any one of claims 2-8, wherein CH1 comprises L143Q and S188V mutations, and CL comprises V133T and S176V mutations.

10. The multispecific antigen-binding protein of any one of claims 2 to 9, wherein CH1 comprises T192E, L143Q, and S188V mutations, and CL comprises N137K, S114A, V133T, and S176V mutations.

11. The multispecific antigen-binding protein of any one of claims 2-10, wherein CH1 comprises a K221E mutation, and CL comprises an E123K mutation.

12. The multispecific antigen-binding protein of any one of claims 2-11, wherein CH1 comprises the K228D mutation and CL comprises the D122K mutation.

13. The multispecific antigen-binding protein of any one of claims 2 to 12, wherein CH1 comprises K221E and K228D mutations, and CL comprises D122K and E123K mutations.

14. The multispecific antigen-binding protein of any one of claims 2 to 13, wherein CH1 comprises an L143E, L143D, L143K, L143R, or L143H mutation, and CL comprises an S176E, S176D, S176K, S176R, or S176H mutation.

15. The multispecific antigen-binding protein of any one of claims 2-14, wherein CH1 comprises an L124E, L124D, L124K, L124R, or L124H mutation, and CL comprises a V133E, V133D, V133K, V133R, or V133H mutation.

16. The multispecific antigen-binding protein of any one of claims 2 to 15, wherein VH comprises a Q39E, Q39D, Q39K, Q39R, or Q39H mutation, and VL comprises a Q38E, Q38D, Q38K, Q38R, or Q38H mutation.

17. The multispecific antigen-binding protein of any one of claims 2 to 16, wherein at least one CH1/CL pair comprises a CH1/CL mutation selected from one or more of:

(1) the T192E (CH1) mutation and the N137K and S114A (CL) mutations,

(2) the L143Q and S188V (CH1) mutations and the V133T and S176V (CL) mutations,

(3) the T192E, L143Q and S188V (CH1) mutations and the N137K, S114A, V133T and S176V (CL) mutations,

(4) the K221E (CH1) mutation and the E123K (CL) mutation,

(5) the T192E and K221E (CH1) mutations and the N137K, S114A and E123K (CL) mutations,

(6) L143E, L143D, L143K, L143R or L143H (CH1) mutations and S176E, S176D, S176K, S176R or S176H (CL) mutations,

(7) L124E, L124D, L124K, L124R or L124H (CH1) mutations and V133E, V133D, V133K, V133R or V133H (CL) mutations,

(8) the K228D (CH1) mutation and the D122K (CL) mutation, and

(9) the K221E and K228D (CH1) mutations and the D122K and E123K (CL) mutations,

wherein when at least two CH1/CL pairs comprise a set of mutations to facilitate pairing of two different VH/VL pairs, then the at least two CH1/CL pairs do not comprise the same mutation, and

wherein at least one VH/VL pair comprises an oppositely charged mutation to facilitate pairing, the oppositely charged mutation comprising (1) a mutated residue at Kabat position 39 in the VH region selected from E, D, K, R or H, and (2) a mutated residue at Kabat position 38 in the VL region selected from E, D, K, R or H, and wherein the mutated residue in the VH region has an opposite charge to the mutated residue in the VL region.

18. An antigen binding protein comprising:

an antigen binding domain; and

a constant heavy chain CH1 region paired with a constant light chain CL region,

wherein the antigen binding domain selectively binds to a target antigen, and wherein the CH1 region and the CL region comprise one or both of:

a) L143E, L143D, L143K, L143R or L143H mutations in the CH1 region and S176E, S176D, S176K, S176R or S176H mutations in the CL region; and

b) the L124E, L124D, L124K, L124R or L124H mutations in the CH1 region and the V133E, V133D, V133K, V133R or V133H mutations in the CL region,

wherein the mutant residue in the CH1 region has an opposite charge to the mutant residue in the CL region.

19. The antigen binding protein of claim 18, further comprising a CH1/CL mutation selected from one or more of the following to facilitate pairing:

(1) the T192E (CH1) mutation and the N137K and S114A (CL) mutations,

(2) the L143Q and S188V (CH1) mutations and the V133T and S176V (CL) mutations,

(3) the T192E, L143Q and S188V (CH1) mutations and the N137K, S114A, V133T and S176V (CL) mutations,

(4) the K221E (CH1) mutation and the E123K (CL) mutation,

(5) the K228D (CH1) mutation and the D122K (CL) mutation, and

(6) the K221E and K228D (CH1) mutations and the D122K and E123K (CL) mutations,

wherein when two CH1/CL pairs comprise a mutation to facilitate pairing of two different VH/VL pairs, the two CH1/CL pairs do not comprise the same mutation.

20. The antigen binding protein of claim 18 or 19, further comprising at least one VH/VL pair comprising an oppositely charged mutation to facilitate pairing, the oppositely charged mutation comprising (1) a mutated residue at Kabat position 39 in the VH region selected from E, D, K, R or H, and (2) a mutated residue at Kabat position 38 in the VL region selected from E, D, K, R or H, and wherein the mutated residue in the VH region has an opposite charge to the mutated residue in the VL region.

21. An antigen binding protein comprising:

an antigen binding domain; and

a constant heavy chain CH1 region paired with a constant light chain CL region,

wherein the antigen binding domain selectively binds to a target antigen, and wherein the CH1 region and the CL region comprise one or both of:

a) L143E, L143D, L143K, L143R or L143H mutations in the CH1 region and S176E, S176D, S176K, S176R or S176H mutations in the CL region; and

b) the K221E and K228D mutations in the CH1 region and the D122K and E123K mutations in the CL region,

wherein the mutant residue in the CH1 region has an opposite charge to the mutant residue in the CL region.

22. The antigen binding protein of claim 21, further comprising at least one VH/VL pair comprising oppositely charged mutations to facilitate pairing, the oppositely charged mutations comprising (1) a mutant residue at Kabat position 39 in the VH region selected from E, D, K, R or H, and (2) a mutant residue at Kabat position 38 in the VL region selected from E, D, K, R or H, and wherein the mutant residue in the VH region has an opposite charge to the mutant residue in the VL region.

23. The multispecific antigen-binding protein of any one of claims 1 to 22, further comprising one or more cysteine residues engineered into one or several VH/VL pairs to form one or more disulfide bonds.

24. The multispecific antigen-binding protein of claim 23, wherein one or both VH regions comprise one or both of the 44C and 105C mutations and one or both VL regions comprise one or both of the 100C and 43C mutations.

25. A kit comprising one or more isolated nucleic acid molecules comprising one or more nucleotide sequences encoding the multispecific antigen-binding protein of any one of claims 1-24.

26. A kit comprising one or more expression vectors comprising one or more nucleic acid molecules according to claim 25.

27. An isolated host cell comprising one or more nucleic acid molecules according to claim 25 or one or more expression vectors according to claim 26.

28. A pharmaceutical composition comprising a pharmaceutically acceptable carrier and the multispecific antigen-binding protein of any one of claims 1-24.

29. A method of treating a disorder in which antigenic activity is detrimental, comprising administering to a subject in need thereof an effective amount of a multispecific antigen-binding protein of any one of claims 1-24.

30. The multispecific antigen-binding protein or antigen-binding protein of any one of claims 1 to 24, comprising three HCDRs per VH region and three LCDRs per VL region, and further comprising binding specificity for one or more target antigens or one or more target epitopes.

Background

Classical antibodies are Y-proteins or immunoglobulins, which are heterotetramers formed from two heterodimers consisting of a light chain portion and a heavy chain portion. The "arm" of an antibody contains the antigen binding site and is referred to as the Fab region. Classical antibodies (e.g., those produced by the host immune system) have two identical fabs and can recognize and bind to a particular antigen. The generation of asymmetry in the native antibody structure is a prerequisite for the production of multispecific binding proteins with two (e.g., bispecific antibodies) or more binding specificities. For example, bispecific antibodies can be made flexible to bind two different antigens simultaneously by separating one or more fvs on different asymmetric binding arms or fabs. However, despite these advantages, many multispecific antibody technologies have process and manufacturing problems due to the fact that each is not paired incorrectly with a weighed chain and a light chain. For example, many of these techniques have a so-called "light chain problem," in which random pairing of two different light chains with a heavy chain results in various chain pairing combinations in addition to the desired combination. In some cases, light chain problems can be avoided by using a common light chain that enables binding to both antigens. However, this may not be feasible for many antibodies, as this format requires de novo antibody production in transgenic mice or by display technology. Furthermore, rare antibodies such as broadly neutralizing anti-HIV antibodies derived from human patients cannot adapt to such formats. Thus, there remains a need for an alternative and inventive solution to the problem of wrong pairing. A series of mutations in the dimer interface have been carefully designed to allow these antibodies to heterodimerize.

Disclosure of Invention

The present disclosure provides antigen binding proteins, which may optionally comprise a plurality of mutations along the dimer interface that allow the antigen binding protein to effectively heterodimerize.

In one aspect, the present disclosure provides an antigen binding protein comprising

A VL region paired with a VH region to form an antigen binding site; and

a CH1 region, the CH1 region being paired with a CL region,

wherein the VL region and the VH region comprise oppositely charged mutations to facilitate pairing, and wherein the CH1 region and the CL region comprise mutations to facilitate pairing.

In some embodiments, the antigen binding protein further comprises a second VL region paired with a second VH region to form a second antigen binding site; and a second CH1 region, the second CH1 region paired with a second CL region.

In some embodiments, one or both of the first VH and VL pair and the second VH and VL pair comprise oppositely charged mutations to facilitate pairing, and one or both of the first CH1 and CL pair and the second CH1 and CL pair comprise mutations to facilitate pairing.

In some embodiments, one or both CH1 regions comprise the T192E mutation and one or both CL regions comprise the N137K and S114A mutations.

In some embodiments, one or both CH1 regions comprise the L143Q and S188V mutations, and one or both CL regions comprise the V133T and S176V mutations.

In some embodiments, one or both CH1 regions comprise T192E, L143Q, and S188V mutations, and one or both CL regions comprise N137K, S114A, V133T, and S176V mutations.

In some embodiments, one or both CH1 regions comprise an L143E, L143D, L143K, L143R, or L143H mutation, and one or both CL regions comprise an S176E, S176D, S176K, S176R, or S176H mutation, wherein the mutation in CH1 is oppositely charged to the mutation in CL.

In some embodiments, one or both CH1 regions comprise an L124E, L124D, L124K, L124R, or L124H mutation, and one or both CL regions comprise a V133E, V133D, V133K, V133R, or V133H mutation, wherein the mutation in CH1 is oppositely charged to the mutation in CL.

In some embodiments, one or both VH regions comprise a 39E, 39D, 39K, 39R, or 39H mutation, and one or both VL regions comprise a 38E, 38D, 38K, 38R, or Q38H mutation, wherein the mutations in VH are oppositely charged to the mutations in VL.

In some embodiments, one or both VH regions comprise a Q39E, Q39D, Q39K, Q39R, or Q39H mutation, and one or both VL regions comprise a Q38E, Q38D, Q38K, Q38R, or Q38H mutation, wherein the mutation in VH is oppositely charged to the mutation in VL.

In some embodiments, the antigen binding protein further comprises one or more cysteine residues engineered into one or both of the VH/VL pairs to form one or more disulfide bonds.

In some embodiments, one or both VH regions comprise one or both of the 44C and 105C mutations, and one or both VL regions comprise one or both of the 100C and 43C mutations.

In some embodiments, one or both VH regions comprise the 44C mutation, and one or both VL regions comprise the 100C mutation.

In some embodiments, one or both VH regions comprise the 105C mutation, and one or both VL regions comprise the 43C mutation.

In some embodiments, the antigen binding protein further comprises oppositely charged mutations in one or both CH1/CL pairs.

In some embodiments, the oppositely charged mutations in one or both of the CH1/CL pairs are selected from: K221E in the CH1 region and E123K in the CL region; K228D in the CH1 region and D122K in the CL region; L145E in the CH1 region and S176K in the CL region; and L128E in the CH1 region and V133K in the CL region.

In another aspect, the present disclosure provides a multispecific antigen-binding protein comprising at least two VL regions that are paired separately with at least two VH regions to form at least two antigen-binding sites, and at least two CH1 regions that are paired separately with two CL regions, wherein at least one CH1/CL pair comprises a CH1/CL mutation selected from:

(1) The T192E (CH1) mutation and the N137K and S114A (CL) mutations, and/or

(2) The L143Q and S188V (CH1) mutations and the V133T and S176V (CL) mutations, and/or

(3) T192E, L143Q and S188V (CH1) mutations and N137K, S114A, V133T and S176V (CL) mutations, and/or

(4) The K221E (CH1) mutation and the E123K (CL) mutation, and/or

(5) The T192E and K221E (CH1) mutations and the N137K, S114A and E123K (CL) mutations, and/or

(6) L143E, L143D, L143K, L143R or L143H (CH1) mutations and S176E, S176D, S176K, S176R or S176H (CL) mutations, and/or

(7) L124E, L124D, L124K, L124R or L124H (CH1) mutations and V133E, V133D, V133K, V133R or V133H (CL) mutations, and

(8) wherein when two CH1/CL pairs comprise a mutation to facilitate pairing of two different VH/VL pairs, the two CH1/CL pairs do not comprise the same mutation,

and is

Wherein at least one VH/VL pair comprises an oppositely charged mutation to facilitate pairing, the oppositely charged mutation comprising (1) a mutated residue at Kabat position 39 in the VH region selected from E, D, K, R or H, and (2) a mutated residue at Kabat position 38 in the VL region selected from E, D, K, R or H, and wherein the mutated residue in the VH region has an opposite charge to the mutated residue in the VL region.

In another aspect, the present disclosure provides a multispecific antigen-binding protein comprising at least two VL regions that are paired separately with at least two VH regions to form at least two antigen-binding sites, and at least two CH1 regions that are paired separately with two CL regions,

wherein at least one CH1/CL pair comprises a CH1/CL mutation selected from one or more of the following to facilitate pairing:

(1) the T192E (CH1) mutation and the N137K and S114A (CL) mutations, and

(2) the L143Q and S188V (CH1) mutations and the V133T and S176V (CL) mutations, and

(3) the T192E, L143Q and S188V (CH1) mutations and the N137K, S114A, V133T and S176V (CL) mutations,

(4) the K221E (CH1) mutation and the E123K (CL) mutation,

(5) the T192E and K221E (CH1) mutations and the N137K, S114A and E123K (CL) mutations,

(6) L143E, L143D, L143K, L143R or L143H (CH1) mutations and S176E, S176D, S176K, S176R or S176H (CL) mutations,

(7) L124E, L124D, L124K, L124R or L124H (CH1) mutations and V133E, V133D, V133K, V133R or V133H (CL) mutations,

(8) the K228D (CH1) mutation and the D122K (CL) mutation, and

(9) the K221E and K228D (CH1) mutations and the D122K and E123K (CL) mutations,

wherein when two CH1/CL pairs contain a mutation to facilitate pairing of two different VH/VL pairs, the two CH1/CL pairs do not contain the same mutation, and

Wherein at least one VH/VL pair comprises an oppositely charged mutation to facilitate pairing, the oppositely charged mutation comprising (1) a mutant residue at Kabat position 39 in the VH region selected from E, D, K, R or H, and (2) a mutant residue at Kabat position 38 in the VL region selected from E, D, K, R or H, and

wherein a mutated residue in the VH domain has an opposite charge to a mutated residue in the VL domain.

In some embodiments, in the multispecific antigen-binding protein, when at least two different VH/VL pairs comprise a mutant set to facilitate pairing, then the at least two different VH/VL pairs do not comprise the same mutant set.

In some embodiments, the multispecific antigen-binding protein comprises at least two VL regions that are paired separately with at least two VH regions to form at least two antigen-binding sites, and at least two CH1 regions that are paired separately with two CL regions,

wherein at least one CH1/CL pair comprises a CH1/CL mutation selected from one or more of the following to facilitate pairing:

(1) the T192E (CH1) mutation and the N137K and S114A (CL) mutations, and/or

(2) The L143Q and S188V (CH1) mutations and the V133T and S176V (CL) mutations, and/or

(3) T192E, L143Q and S188V (CH1) mutations and N137K, S114A, V133T and S176V (CL) mutations, and/or

(4) The K221E (CH1) mutation and the E123K (CL) mutation, and/or

(5) L143E, L143D, L143K, L143R or L143H (CH1) mutations and S176E, S176D, S176K, S176R or S176H (CL) mutations, and/or

(6) L124E, L124D, L124K, L124R or L124H (CH1) mutations and V133E, V133D, V133K, V133R or V133H (CL) mutations, and

(7) when two CH1/CL pairs contain mutations to facilitate the pairing of two different VH/VL pairs, the two CH1/CL pairs do not contain the same mutations,

and is

Wherein at least one VH/VL pair comprises oppositely charged mutations selected from: 39E, Q39D, Q39K, Q39R or Q39H mutations and Q38E, Q38D, Q38K, Q38R or Q38H mutations, and wherein the mutations in VH are oppositely charged to the mutations in VL; and when several VH/VL pairs (where VL or VH are not in the same polypeptide chain) contain mutations to facilitate pairing of different VH/VL pairs, then each VH/VL pair does not contain the same oppositely charged mutation.

In some embodiments, one or both CH1 regions are operably linked to a heterodimerization domain.

In some embodiments, the heterodimerization domain comprises a first Fc domain.

In some embodiments, the first Fc domain is heterodimerized with a second Fc domain, and wherein the first Fc domain comprises a first CH3 region and the second Fc domain comprises a second CH3 region.

In some embodiments, the first CH3 region comprises one or both of the S354C and T366W mutations and the second CH3 region comprises one or more of the Y349C, T366S, L368A, and Y407V mutations, wherein the mutations promote Fc domain heterodimerization.

In another aspect, the disclosure provides a multispecific antibody comprising

a) A first light chain (LC 1)/heavy chain (HC1) pair, said first light chain (LC 1)/heavy chain (HC1) pair comprising

(1) A first VL region (VL1) that pairs with a first VH region (VH1) to form a first antigen binding site (VL 1);

(2) a first constant heavy chain region 1(CH1-1) and a first constant light chain region (CL 1); and

(3) a first heterodimerization domain (HD 1); and

b) a second light chain (LC 2)/heavy chain (HC2) pair comprising

(4) A second VL region (VL2) that pairs with a second VH region (VH2) to form a second antigen binding site;

(5) a second constant heavy chain region 1(CH1-2) and a second constant light chain region (CL 2); and

(6) A second heterodimerization domain (HD 2);

wherein HD1 and HD2 heterodimerize,

wherein at least one or both of the VL1 and VH1 pairs and at least one or both of the VL2 and VH2 pairs comprise oppositely charged mutations to facilitate pairing,

wherein at least one or both of the pair CL1 and CH1-1 and at least one or both of the pair CL2 and CH1-2 comprise mutations to facilitate pairing, and

wherein when both of the pair CL1 and CH1-1 and both of the pair CL2 and CH1-2 comprise mutations to facilitate pairing, the mutations that facilitate pairing in CH1-1 and CL1 are different from the mutations that facilitate pairing in CH1-2 and CL 2.

In another aspect, the present disclosure provides a multispecific antibody comprising:

a) a first light chain (LC 1)/heavy chain (HC1) pair, said first light chain (LC 1)/heavy chain (HC1) pair comprising

(1) A first VL region (VL1) that pairs with a first VH region (VH1) to form a first antigen binding site (VL 1);

(2) a first constant heavy chain region 1(CH1-1) operably linked to VH1 and a first constant light chain region (CL1) operably linked to VL 1; and

(3) a first heterodimerization domain (HD 1); and

b) a second light chain (LC 2)/heavy chain (HC2) pair comprising

(4) A second VL region (VL2) that pairs with a second VH region (VH2) to form a second antigen binding site;

(5) a second constant heavy chain region 1(CH1-2) operably linked to VH2 and a second constant light chain region (CL2) operably linked to VL 2; and

(6) a second heterodimerization domain (HD 2);

wherein HD1 and HD2 heterodimerize,

wherein at least one or both of the VL1 and VH1 pairs and at least one or both of the VL2 and VH2 pairs comprise oppositely charged mutations to facilitate pairing,

wherein at least one or both of the pair CL1 and CH1-1 and at least one or both of the pair CL2 and CH1-2 comprise mutations to facilitate pairing, and

wherein when both of the pair CL1 and CH1-1 and both of the pair CL2 and CH1-2 comprise mutations to facilitate pairing, the mutations that facilitate pairing in CH1-1 and CL1 are different from the mutations that facilitate pairing in CH1-2 and CL 2.

In another aspect, the present disclosure provides a multispecific antigen-binding protein comprising:

a) a first light chain (LC 1)/heavy chain (HC1) pair, said first light chain (LC 1)/heavy chain (HC1) pair comprising

(1) A first VL region (VL1) that pairs with a first VH region (VH1) to form a first antigen binding site (VL 1);

(2) A first constant heavy chain region 1(CH1-1) operably linked to VH1 and a first constant light chain region (CL1) operably linked to VL 1; and

(3) a first heterodimerization domain (HD 1); and

b) a second light chain (LC 2)/heavy chain (HC2) pair comprising

(4) A second VL region (VL2) that pairs with a second VH region (VH2) to form a second antigen binding site;

(5) a second constant heavy chain region 1(CH1-2) operably linked to VH2 and a second constant light chain region (CL2) operably linked to VL 2; and

(6) a second heterodimerization domain (HD 2);

wherein HD1 and HD2 heterodimerize,

wherein at least one or both of the pair VL1 and VH1 and at least one or both of the pair VL2 and VH2 comprise oppositely charged mutations to facilitate pairing, the oppositely charged mutations comprising (1) a mutant residue at Kabat position 39 in the VH region selected from E, D, K, R or H, and (2) a mutant residue at Kabat position 38 in the VL region selected from E, D, K, R or H, and wherein the mutant residue in the VH region has an opposite charge to the mutant residue in the VL region,

wherein at least one or both of the pair CL1 and CH1-1 and at least one or both of the pair CL2 and CH1-2 comprise mutations to facilitate pairing, and

Wherein when both of the pair CL1 and CH1-1 and both of the pair CL2 and CH1-2 comprise mutations to facilitate pairing, the mutations that facilitate pairing in CH1-1 and CL1 are different from the mutations that facilitate pairing in CH1-2 and CL 2.

In some embodiments, CH1-1 comprises the T192E mutation, and CL1 comprises the N137K and S114A mutations.

In some embodiments, CH1-1 comprises L143Q and S188V mutations, and CL1 comprises V133T and S176V mutations.

In some embodiments, CH1-1 comprises T192E, L143Q, and S188V mutations, and CL1 comprises N137K, S114A, V133T, and S176V mutations.

In some embodiments, CH1-2 comprises the T192E mutation, and CL2 comprises the N137K and S114A mutations.

In some embodiments, CH1-2 comprises L143Q and S188V mutations, and CL2 comprises V133T and S176V mutations.

In some embodiments, CH1-2 comprises T192E, L143Q, and S188V mutations, and CL2 comprises N137K, S114A, V133T, and S176V mutations.

In some embodiments, one or both of CH1-1 and CH1-2 comprises an L143E, L143D, L143K, L143R, or L143H mutation, and one or both of CL1 and CL2 comprises an S176E, S176D, S176K, S176R, or S176H mutation, wherein the mutation in one or both of CH1-1 or CH1-2 is oppositely charged than the mutation in one or both of CL1 and CL 2.

In some embodiments, one or both of CH1-1 and CH1-2 comprises an L124E, L124D, L124K, L124R, or L124H mutation, and one or both of CL1 and CL2 comprises a V133E, V133D, V133K, V133R, or V133H mutation, wherein the mutation in one or both of CH1-1 or CH1-2 is oppositely charged than the mutation in one or both of CL1 and CL 2.

In some embodiments, one or both of VH1 and VH2 comprises a 39E, 39D, 39K, 39R, or 39H mutation, and one or both of VL1 and VL2 comprises a 38E, 38D, 38K, 38R, or 38H mutation, wherein the mutation in one or both of VH1 and VH2 is oppositely charged to the mutation in one or both of VL1 and VL 2.

In some embodiments, one or both of VH1 and VH2 comprises a Q39E, Q39D, Q39K, Q39R, or Q39H mutation, and one or both of VL1 and VL2 comprises a Q38E, Q38D, Q38K, Q38R, or Q38H mutation, wherein the mutation in one or both of VH1 and VH2 is oppositely charged to the mutation in one or both of VL1 and VL 2.

In some embodiments, the multispecific antibody further comprises one or more cysteine residues engineered into one or both of the VH1/VL1 and VH2/VL2 pairs to form one or more disulfide bonds.

In some embodiments, one or both of VH1 and VH2 comprises 44C and 105C mutations, and one or both of VL1 and VL2 comprises 100C and 43C mutations.

In some embodiments, VH1 comprises the 44C mutation, and VL1 comprises the 100C mutation.

In some embodiments, VH1 comprises the 105C mutation and VL1 comprises the 43C mutation.

In some embodiments, VH2 comprises the 44C mutation, and VL2 comprises 100C.

In some embodiments, VH2 comprises the 105C mutation and VL2 comprises the 43C mutation.

In some embodiments, VH1 comprises 39E and 44C mutations, and VL1 comprises 38K and 100C mutations.

In some embodiments, VH1 comprises the 39E and 105C mutations and VL1 comprises the 38K and 43C mutations.

In some embodiments, the multispecific antibody further comprises an oppositely charged mutation in the CH1-1/CL1 pair.

In some embodiments, the oppositely charged mutations in the CH1-1/CL1 pair are selected from: K221E in the CH1-1 region and E123K in the CL1 region; K228D in the CH1-1 region and D122K in the CL1 region; L145E in the CH1-1 region and S176K in the CL1 region; and L128E in the CH1-1 region and V133K in the CL1 region.

In some embodiments, the multispecific antibody further comprises an oppositely charged mutation in the CH1-2/CL2 pair.

In some embodiments, the oppositely charged mutations in the CH1-2/CL2 pair are selected from: K221E in the CH1-2 region and E123K in the CL2 region; K228D in the CH1-2 region and D122K in the CL2 region; L145E in the CH1-2 region and S176K in the CL2 region; and L128E in the CH1-2 region and V133K in the CL2 region.

In some embodiments, the first heterodimerization domain and the second heterodimerization domain comprise Fc domains.

In some embodiments, the first heterodimerization domain comprises a first CH3 domain, the first CH3 domain comprises one or both of the S354C and T366W mutations, and the second heterodimerization domain comprises a second CH3 domain, the second CH3 domain comprises one or both of the Y349C, T366S, L368A, and Y407V mutations, wherein the mutations promote Fc domain heterodimerization.

In some embodiments, the CH1-1 domain is linked to a first CH2 domain and a first CH3 domain, the CH1-2 domain is linked to a second CH2 domain and a second CH3 domain, and wherein the first CH2 and CH3 domains and the second CH2 and CH3 domains dimerize to form an Fc domain.

In some embodiments, the first CH3 domain comprises one or both of the S354C and T366W mutations, the second CH3 domain comprises one or more of the Y349C, T366S, L368A, and Y407V mutations, and wherein the mutations promote heterodimerization of Fc domains.

In another aspect, the present disclosure provides an antigen binding protein or multispecific antigen binding protein comprising at least two polypeptide chains and forming at least two antigen binding sites, wherein one polypeptide chain comprises a structure represented by the formula:

VL1-L1-VL2-L2-CL[I]

and one polypeptide chain comprises a structure represented by the formula:

VH2-L3-VH1-L4-CH1[II]

wherein:

VL1 is a first immunoglobulin light chain variable domain;

VL2 is a second immunoglobulin light chain variable domain;

VH1 is a first immunoglobulin heavy chain variable domain;

VH2 is a second immunoglobulin heavy chain variable domain;

CL is an immunoglobulin light chain constant domain;

CH1 is an immunoglobulin CH1 heavy chain constant domain; and is

L1, L2, L3 and L4 are amino acid linkers,

wherein said polypeptide of formula I and said polypeptide of formula II form a cross light-heavy chain pair,

wherein one or more cysteine residues are engineered into one or both of the VH1/VL1 pair and the VH2/VL2 pair to form one or more disulfide bonds,

Wherein at least one or both of the pair VL1 and VH1 and at least one or both of the pair VL2 and VH2 comprise oppositely charged mutations that promote pairing, and

wherein the CH1 and CL domain pair comprises a mutation that facilitates pairing.

In some embodiments, VH1 is paired with VL1, VH2 is paired with VL2, and CH1 is paired with CL.

In some embodiments, the present disclosure provides a multispecific antigen-binding protein comprising at least two polypeptide chains and forming at least two antigen-binding sites, wherein one polypeptide chain comprises a structure represented by the formula:

VL1-L1-VL2-L2-CL[I]

and one polypeptide chain comprises a structure represented by the formula:

VH2-L3-VH1-L4-CH1[II]

wherein:

VL1 is a first immunoglobulin light chain variable domain;

VL2 is a second immunoglobulin light chain variable domain;

VH1 is a first immunoglobulin heavy chain variable domain;

VH2 is a second immunoglobulin heavy chain variable domain;

CL is an immunoglobulin light chain constant domain;

CH1 is an immunoglobulin CH1 heavy chain constant domain; and is

L1, L2, L3 and L4 are amino acid linkers,

wherein VH1 is paired with VL1, VH2 is paired with VL2, and CH1 is paired with CL,

wherein said polypeptide of formula I and said polypeptide of formula II form a cross light-heavy chain pair,

Wherein one or more cysteine residues are engineered into one or both of the VH1/VL1 pair and the VH2/VL2 pair to form one or more disulfide bonds,

wherein at least one or both of the pair VL1 and VH1 and at least one or both of the pair VL2 and VH2 comprise oppositely charged mutations that facilitate pairing, the oppositely charged mutations comprising (1) a mutant residue at Kabat position 39 in the VH region selected from E, D, K, R or H, and (2) a mutant residue at Kabat position 38 in the VL region selected from E, D, K, R or H, and wherein the mutant residue in the VH region has an opposite charge to the mutant residue in the VL region, and

wherein the CH1 and CL domain pair comprises a mutation that facilitates pairing.

In some embodiments, one or both of VH1 and VH2 comprises VH44C and VH105C mutations.

In some embodiments, one or both of VL1 and VL2 comprises VL43C and VL100C mutations.

In some embodiments, one or both of VH1 and VH2 comprises a VH44C mutation, and one or both of VL1 and VL2 comprises a VL100C mutation.

In some embodiments, one or both of VH1 and VH2 comprises a VH105C mutation, and one or both of VL1 and VL2 comprises a VL43C mutation.

In some embodiments, CH1 comprises the T192E mutation and CL comprises the N137K and S114A mutations.

In some embodiments, CH1 comprises L143Q and S188V mutations, and CL comprises V133T and S176V mutations.

In some embodiments, CH1 comprises T192E, L143Q, and S188V mutations, and CL comprises N137K, S114A, V133T, and S176V mutations.

In some embodiments, CH1 comprises an L143E, L143D, L143K, L143R, or L143H mutation, CL comprises an S176E, S176D, S176K, S176R, or S176H mutation, and wherein the mutation in CH1 is oppositely charged to the mutation in CL.

In some embodiments, CH1 comprises an L124E, L124D, L124K, L124R, or L124H mutation, CL comprises a V133E, V133D, V133K, V133R, or V133H mutation, and wherein the mutation in CH1 is oppositely charged to the mutation in CL.

In some embodiments, one or both of VH1 and VH2 comprises a 39E, 39D, 39K, 39R, or 39H mutation, one or both of VL1 and VL2 comprises a 38E, 38D, 38K, 38R, or 38H mutation, and wherein the mutation in one or both of VH1 and VH2 is oppositely charged to the mutation in one or both of VL1 and VL 2.

In some embodiments, one or both of VH1 and VH2 comprises a Q39E, Q39D, Q39K, Q39R, or Q39H mutation, one or both of VL1 and VL2 comprises a Q38E, Q38D, Q38K, Q38R, or Q38H mutation, and wherein the mutation in one or both of VH1 and VH2 is oppositely charged to the mutation in one or both of VL1 and VL 2.

In some embodiments, CH1 is operably linked to a dimerization domain.

In some embodiments, the dimerization domain is an Fc domain comprising a CH2 domain and a CH3 domain.

In another aspect, the present disclosure provides a multispecific antigen-binding protein comprising four polypeptide chains forming four antigen-binding sites, wherein each of the two polypeptide chains comprises a structure represented by the formula:

VL1-L1-VL2-L2-CL[I]

and each of the two polypeptide chains comprises a structure represented by the formula:

VH2-L3-VH1-L4-CH1-Fc[II]

wherein:

VL1 is a first immunoglobulin light chain variable domain;

VL2 is a second immunoglobulin light chain variable domain;

VH1 is a first immunoglobulin heavy chain variable domain;

VH2 is a second immunoglobulin heavy chain variable domain;

CL is an immunoglobulin light chain constant domain;

CH1 is an immunoglobulin CH1 heavy chain constant domain;

Fc comprises an immunoglobulin hinge region and CH2 and CH3 immunoglobulin heavy chain constant domains; and is

L1, L2, L3 and L4 are amino acid linkers,

wherein VH1 pairs with VL1 to form a first antigen binding site, VH2 pairs with VL2 to form a second antigen binding site, and CH1 pairs with CL,

wherein said polypeptide of formula I and said polypeptide of formula II form a cross light-heavy chain pair,

wherein one or more cysteine residues are engineered into one or both of the VH1/VL1 pair and the VH2/VL2 pair to form one or more disulfide bonds,

wherein one or both of the pair VL1 and VH1 and one or both of the pair VL2 and VH2 comprise oppositely charged mutations that facilitate pairing, the oppositely charged mutations comprising (1) a mutated residue at Kabat position 39 in the VH region selected from E, D, K, R or H, and (2) a mutated residue at Kabat position 38 in the VL region selected from E, D, K, R or H, and wherein the mutated residue in the VH region has an opposite charge to the mutated residue in the VL region, and

wherein one or both of the CH1 and CL domain pairs comprise a mutation that facilitates pairing, and

wherein when at least two CH1/CL pairs contain mutations to promote pairing, the set of mutations in one CH1/CL pair is different from the set of mutations in the other CH1/CL pair.

In another aspect, the present disclosure provides an antigen binding protein comprising four polypeptide chains forming four antigen binding sites, wherein each of the two polypeptide chains comprises a structure represented by the formula:

VL1-L1-VL2-L2-CL[I]

and each of the two polypeptide chains comprises a structure represented by the formula:

VH2-L3-VH1-L4-CH1-Fc[II]

wherein:

VL1 is a first immunoglobulin light chain variable domain;

VL2 is a second immunoglobulin light chain variable domain;

VH1 is a first immunoglobulin heavy chain variable domain;

VH2 is a second immunoglobulin heavy chain variable domain;

CL is an immunoglobulin light chain constant domain;

CH1 is an immunoglobulin CH1 heavy chain constant domain;

fc comprises an immunoglobulin hinge region and CH2 and CH3 immunoglobulin heavy chain constant domains; and is

L1, L2, L3 and L4 are amino acid linkers,

wherein said polypeptide of formula I and said polypeptide of formula II form a cross light-heavy chain pair,

wherein VH1 pairs with VL1 to form a first antigen-binding site, VH2 pairs with VL2 to form a second antigen-binding site, and CH1 pairs with CL, and more particularly, the VH1/VL1 pair comprises a first antigen-binding specificity and the VH2/VL2 pair comprises a second antigen-binding specificity,

wherein one or more cysteine residues are engineered into one or both of the VH1/VL1 pair and the VH2/VL2 pair to form one or more disulfide bonds,

Wherein one or both of the pair VL1 and VH1 and one or both of the pair VL2 and VH2 comprise oppositely charged mutations that promote pairing, and

wherein one or both of the CH1 and CL domain pairs comprise a mutation that facilitates pairing, and

wherein when both CH1 and CL pairs contain mutations to promote pairing, the mutations in one CH1 and CL pair are different from the mutations in the other CH1 and CL pair to promote pairing.

In some embodiments, one or both of VH1 and VH2 comprises VH44C and VH105C mutations.

In some embodiments, one or both of VL1 and VL2 comprises VL43C and VL100C mutations.

In some embodiments, one or both of VH1 and VH2 comprises a VH44C mutation, and one or both of VL1 and VL2 comprises a VL100C mutation.

In some embodiments, one or both of VH1 and VH2 comprises a VH105C mutation, and one or both of VL1 and VL2 comprises a VL43C mutation.

In some embodiments, CH1 comprises the T192E mutation and CL comprises the N137K and S114A mutations.

In some embodiments, CH1 comprises L143Q and S188V mutations, and CL comprises V133T and S176V mutations.

In some embodiments, CH1 comprises T192E, L143Q, and S188V mutations, and CL comprises N137K, S114A, V133T, and S176V mutations.

In some embodiments, CH1 comprises an L143E, L143D, L143K, L143R, or L143H mutation, CL comprises an S176E, S176D, S176K, S176R, or S176H mutation, and wherein the mutation in CH1 is oppositely charged to the mutation in CL.

In some embodiments, CH1 comprises an L124E, L124D, L124K, L124R, or L124H mutation, CL comprises a V133E, V133D, V133K, V133R, or V133H mutation, and wherein the mutation in CH1 is oppositely charged to the mutation in CL.

In some embodiments, one or both of VH1 and VH2 comprises a 39E, 39D, 39K, 39R, or 39H mutation, one or both of VL1 and VL2 comprises a 38E, 38D, 38K, 38R, or 38H mutation, and wherein the mutation in one or both of VH1 and VH2 is oppositely charged to the mutation in one or both of VL1 and VL 2.

In some embodiments, one or both of VH1 and VH2 comprises a Q39E, Q39D, Q39K, Q39R, or Q39H mutation, one or both of VL1 and VL2 comprises a Q38E, Q38D, Q38K, Q38R, or Q38H mutation, and wherein the mutation in one or both of VH1 and VH2 is oppositely charged to the mutation in one or both of VL1 and VL 2.

In another aspect, the present disclosure provides a multispecific antigen-binding protein comprising four polypeptide chains forming three antigen-binding sites, wherein

The first polypeptide chain comprises a structure represented by the formula:

VL2-L1-VL1-L2-CL1[I],

the second polypeptide chain comprises a structure represented by the formula:

VH1-L3-VH2-L4-CH 1-1-hinge-CH 2-CH3[ II ],

the third polypeptide chain comprises a structure represented by the formula:

VH3-CH 1-2-hinge-CH 2-CH3[ III ], and

the fourth polypeptide chain comprises a structure represented by the formula:

VL3-CL2[IV],

wherein:

VL1 is a first immunoglobulin light chain variable domain;

VL2 is a second immunoglobulin light chain variable domain;

VL3 is a third immunoglobulin light chain variable domain;

VH1 is a first immunoglobulin heavy chain variable domain;

VH2 is a second immunoglobulin heavy chain variable domain;

VH3 is a third immunoglobulin heavy chain variable domain;

CL1 is a first immunoglobulin light chain constant domain;

CL2 is a second immunoglobulin light chain constant domain;

CH1-1 is a first immunoglobulin CH1 heavy chain constant domain;

CH1-2 is a second immunoglobulin CH1 heavy chain constant domain;

CH2 is an immunoglobulin CH2 heavy chain constant domain;

CH3 is an immunoglobulin CH3 heavy chain constant domain;

A hinge is an immunoglobulin hinge region connecting the CH1 domain and the CH2 domain; and L1, L2, L3 and L4 are amino acid linkers,

wherein said polypeptide of formula I and said polypeptide of formula II form a cross light-heavy chain pair, and

wherein one or more cysteine residues are engineered into one or more of the VH1/VL1 pair, the VH2/VL2 pair and the VH3/VL3 pair to form one or more disulfide bonds,

wherein one or both of the pair VL1 and VH1 and one or both of the pair VL2 and VH2 comprise oppositely charged mutations that facilitate pairing, the oppositely charged mutations comprising (1) a mutated residue at Kabat position 39 in the VH region selected from E, D, K, R or H, and (2) a mutated residue at Kabat position 38 in the VL region selected from E, D, K, R or H, and wherein the mutated residue in the VH region has an opposite charge to the mutated residue in the VL region,

wherein one or both of the pair CL1 and CH1-1 and one or both of the pair CL2 and CH1-2 comprise mutations that promote pairing, and

wherein the mutations in CH1-1 and CL1 are different from the mutations in CH1-2 and CL2 when both of the CL1 and CH1-1 pairs and both of the CL2 and CH1-2 pairs contain mutations to facilitate pairing.

In some embodiments, one or both of VH1 and VH2 comprises VH44C and VH105C mutations.

In some embodiments, one or both of VL1 and VL2 comprises VL43C and VH100C mutations.

In some embodiments, one or both of VH1 and VH2 comprises a VH44C mutation, and one or both of VL1 and VL2 comprises a VL100C mutation.

In some embodiments, one or both of VH1 and VH2 comprises a VH105C mutation, and one or both of VL1 and VL2 comprises a VL43C mutation.

In some embodiments, one or both of CH1-1 and CL1 comprise a mutation that facilitates pairing and one or both of CH1-2 and CL2 comprise a mutation that facilitates pairing.

In some embodiments, CH1-1 comprises the T192E mutation, and CL1 comprises the N137K and S114A mutations.

In some embodiments, CH1-1 comprises L143Q and S188V mutations, and CL1 comprises V133T and S176V mutations.

In some embodiments, CH1-1 comprises T192E, L143Q, and S188V mutations, and CL1 comprises N137K, S114A, V133T, and S176V mutations.

In some embodiments, CH1-2 comprises the T192E mutation, and CL2 comprises the N137K and S114A mutations.

In some embodiments, CH1-2 comprises L143Q and S188V mutations, and CL2 comprises V133T and S176V mutations.

In some embodiments, CH1-2 comprises T192E, L143Q, and S188V mutations, and CL2 comprises N137K, S114A, V133T, and S176V mutations.

In some embodiments, one or both of CH1-1 and CH1-2 comprises an L143E, L143D, L143K, L143R, or L143H mutation, one or both of CL1 and CL2 comprises an S176E, S176D, S176K, S176R, or S176H mutation, and wherein the mutation in one or both of CH1-1 or CH1-2 is oppositely charged to the mutation in one or both of CL1 and CL 2.

In some embodiments, one or both of CH1-1 and CH1-2 comprises an L124E, L124D, L124K, L124R, or L124H mutation, one or both of CL1 and CL2 comprises a V133E, V133D, V133K, V133R, or V133H mutation, and wherein the mutation in one or both of CH1-1 or CH1-2 is oppositely charged than the mutation in one or both of CL1 and CL 2.

In some embodiments, one or more of VH1, VH2, and VH3 comprises a 39E, 39D, 39K, 39R, or 39H mutation, one or more of VL1, VL2, and VL3 comprises a 38E, 38D, 38K, 38R, or 38H mutation, and wherein the mutation in one or more of VH1, VH2, and VH3 is oppositely charged to the mutation in one or more of VL1, VL2, and VL 3.

In some embodiments, one or more of VH1, VH2, and VH3 comprises a Q39E, Q39D, Q39K, Q39R, or Q39H mutation, one or more of VL1, VL2, and VL3 comprises a Q38E, Q38D, Q38K, Q38R, or Q38H mutation, and wherein the mutation in one or more of VH1, VH2, and VH3 is oppositely charged to the mutation in one or more of VL1, VL2, and VL 3.

In another aspect, the present disclosure provides a multispecific antigen-binding protein or antibody comprising:

a) a first light chain (LC 1)/heavy chain (HC1) pair, the first light chain (LC 1)/heavy chain (HC1) pair comprising:

(1) a first VL region (VL1) that pairs with a first VH region (VH1) to form a first antigen binding site (VL 1);

(2) a first constant heavy chain region 1(CH1-1) operably linked to VH1 and a first constant light chain region (CL1) operably linked to VL1, and

b) a second light chain (LC 2)/heavy chain (HC2) pair, the second light chain (LC 2)/heavy chain (HC2) pair comprising:

(3) a second VL region (VL2) that pairs with a second VH region (VH2) to form a second antigen binding site; and

(4) a second constant heavy chain region 1(CH1-2) operably linked to VH2 and a second constant light chain region (CL2) operably linked to VL2,

Wherein the C-terminus of CH1-1 is operably linked to the N-terminus of VH2, and

wherein one or more cysteine residues are engineered into one or both of the VH1/VL1 pair and the VH2/VL2 pair to form one or more disulfide bonds,

wherein one or both of the VL1 and VH1 pairs and one or both of the VL2 and VH2 pairs comprise oppositely charged mutations that promote pairing,

wherein one or both of the pair CL1 and CH1-1 and one or both of the pair CL2 and CH1-2 comprise mutations that promote pairing, and

wherein the mutations in CH1-1 and CL1 are different from the mutations in CH1-2 and CL2 when both of the CL1 and CH1-1 pairs and both of the CL2 and CH1-2 pairs contain mutations to facilitate pairing.

In some embodiments, CH1-1 comprises the T192E mutation, and CL1 comprises the N137K and S114A mutations.

In some embodiments, CH1-1 comprises L143Q and S188V mutations, and CL1 comprises V133T and S176V mutations.

In some embodiments, CH1-1 comprises T192E, L143Q, and S188V mutations, and CL1 comprises N137K, S114A, V133T, and S176V mutations.

In some embodiments, CH1-2 comprises the T192E mutation, and CL2 comprises the N137K and S114A mutations.

In some embodiments, CH1-2 comprises L143Q and S188V mutations, and CL2 comprises V133T and S176V mutations.

In some embodiments, CH1-2 comprises T192E, L143Q, and S188V mutations, and CL2 comprises N137K, S114A, V133T, and S176V mutations.

In some embodiments, one or both of CH1-1 and CH1-2 comprises an L143E, L143D, L143K, L143R, or L143H mutation, one or both of CL1 and CL2 comprises an S176E, S176D, S176K, S176R, or S176H mutation, and wherein the mutation in one or both of CH1-1 or CH1-2 is oppositely charged to the mutation in one or both of CL1 and CL 2.

In some embodiments, one or both of CH1-1 and CH1-2 comprises an L124E, L124D, L124K, L124R, or L124H mutation, one or both of CL1 and CL2 comprises a V133E, V133D, V133K, V133R, or V133H mutation, and wherein the mutation in one or both of CH1-1 or CH1-2 is oppositely charged than the mutation in one or both of CL1 and CL 2.

In some embodiments, one or both of VH1 and VH2 comprises a 39E, 39D, 39K, 39R, or 39H mutation, one or both of VL1 and VL2 comprises a 38E, 38D, 38K, 38R, or 38H mutation, and wherein the mutation in one or both of VH1 and VH2 is oppositely charged to the mutation in one or both of VL1 and VL 2.

In some embodiments, one or both of VH1 and VH2 comprises a Q39E, Q39D, Q39K, Q39R, or Q39H mutation, one or both of VL1 and VL2 comprises a Q38E, Q38D, Q38K, Q38R, or Q38H mutation, and wherein the mutation in one or both of VH1 and VH2 is oppositely charged to the mutation in one or both of VL1 and VL 2.

In some embodiments, the present disclosure provides a multispecific antigen-binding protein comprising:

a) a first light chain (LC 1)/heavy chain (HC1) pair, the first light chain (LC 1)/heavy chain (HC1) pair comprising:

(1) a first VL region (VL1) that pairs with a first VH region (VH1) to form a first antigen binding site (VL 1);

(2) a first constant heavy chain region 1(CH1-1) operably linked to VH1 and a first constant light chain region (CL1) operably linked to VL1, and

b) a second light chain (LC 2)/heavy chain (HC2) pair, the second light chain (LC 2)/heavy chain (HC2) pair comprising:

(3) a second VL region (VL2) that pairs with a second VH region (VH2) to form a second antigen binding site; and

(4) a second constant heavy chain region 1(CH1-2) operably linked to VH2 and a second constant light chain region (CL2) operably linked to VL2,

Wherein the C-terminus of CH1-1 is operably linked to the N-terminus of VH2, and

wherein one or more cysteine residues are engineered into one or both of the VH1/VL1 pair and the VH2/VL2 pair to form one or more disulfide bonds,

wherein one or both of the pair VL1 and VH1 and one or both of the pair VL2 and VH2 comprise oppositely charged mutations that facilitate pairing, the oppositely charged mutations comprising (1) a mutated residue at Kabat position 39 in the VH region selected from E, D, K, R or H, and (2) a mutated residue at Kabat position 38 in the VL region selected from E, D, K, R or H, and wherein the mutated residue in the VH region has an opposite charge to the mutated residue in the VL region,

wherein one or both of the pair CL1 and CH1-1 and one or both of the pair CL2 and CH1-2 comprise mutations that promote pairing, and

wherein the mutations in CH1-1 and CL1 are different from the mutations in CH1-2 and CL2 when both of the CL1 and CH1-1 pairs and both of the CL2 and CH1-2 pairs contain mutations to facilitate pairing.

In some embodiments, the multispecific antibody further comprises one or more cysteine residues engineered into one or both of the VH1/VL1 and VH2/VL2 pairs to form one or more disulfide bonds.

In some embodiments, one or both of VH1 and VH2 comprises 44C and 105C mutations, and one or both of VL1 and VL2 comprises 100C and 43C mutations.

In some embodiments, VH1 comprises the 44C mutation, and VL1 comprises the 100C mutation.

In some embodiments, VH1 comprises the 105C mutation and VL1 comprises the 43C mutation.

In some embodiments, VH2 comprises the 44C mutation, and VL2 comprises the 100C mutation.

In some embodiments, VH2 comprises the 105C mutation and VL2 comprises the 43C mutation.

In some embodiments, VH1 comprises 39E and 44C mutations, and VL1 comprises 38K and 100C mutations.

In some embodiments, VH1 comprises the 39E and 105C mutations and VL1 comprises the 38K and 43C mutations.

In some embodiments, the multispecific antibody further comprises an oppositely charged mutation in the CH1-1/CL1 pair.

In some embodiments, the oppositely charged mutations in the CH1-1/CL1 pair are selected from: K221E in the CH1-1 region and E123K in the CL1 region; K228D in the CH1-1 region and D122K in the CL1 region; L145E in the CH1-1 region and S176K in the CL1 region; and L128E in the CH1-1 region and V133K in the CL1 region.

In some embodiments, the multispecific antibody further comprises an oppositely charged mutation in the CH1-2/CL2 pair.

In some embodiments, the oppositely charged mutations in the CH1-2/CL2 pair are selected from: K221E in the CH1-2 region and E123K in the CL2 region; K228D in the CH1-2 region and D122K in the CL2 region; L145E in the CH1-2 region and S176K in the CL2 region; and L128E in the CH1-2 region and V133K in the CL2 region.

In some embodiments, the CH1-1 domain is linked to a first CH2 domain and a first CH3 domain, the CH1-2 domain is linked to a second CH2 domain and a second CH3 domain, and wherein the first CH2 and CH3 domains and the second CH2 and CH3 domains dimerize to form an Fc domain.

In some embodiments, the first CH3 domain comprises one or both of the S354C and T366W mutations, and the second CH3 domain comprises one or both of the Y349C, T366S, L368A, and Y407V mutations to promote Fc domain heterodimerization.

In some embodiments, the C-terminus of CH1-1 is operably linked to the N-terminus of VH2 via a peptide linker.

In some embodiments, the peptide linker comprises (GGGGS)n(SEQ ID NO: X) linker, wherein n is any integer from 1 to 5.

In some embodiments, the peptide linker comprises all or part of the sequence of the hinge region of one or more immunoglobulins selected from IgA, IgG, and IgD.

In some embodiments, the peptide linker comprises the following sequence:

EPKSCDKTHTSPPSPAPELLGGPSTPPTPSPSGG(SEQ ID NO:X)。

in another aspect, the present disclosure provides a multispecific antigen-binding protein or antibody comprising:

a) a first light chain (LC 1)/heavy chain (HC1) pair, said first light chain (LC 1)/heavy chain (HC1) pair comprising

(1) A first VL region (VL1) that pairs with a first VH region (VH1) to form a first antigen binding site (VL 1);

(2) a first constant heavy chain region 1(CH1-1) operably linked to VH1 and a first constant light chain region (CL1) operably linked to VL 1; and

(3) a first heterodimerization domain (HD 1); and

b) a second light chain (LC 2)/heavy chain (HC2) pair comprising

(4) A second VL region (VL2) that pairs with a second VH region (VH2) to form a second antigen binding site;

(5) a second constant heavy chain region 1(CH1-2) operably linked to VH2 and a second constant light chain region (CL2) operably linked to VL 2; and

(6) a second heterodimerization domain (HD 2);

wherein HD1 and HD2 heterodimerize,

wherein at least one or both of VH1 and VH2 comprises a Q39E, Q39D, Q39K, Q39R, or Q39H mutation, and one or both of VL1 and VL2 comprises a Q38E, Q38D, Q38K, Q38R, or Q38H mutation, wherein the mutation in one or both of VH1 and VH2 is oppositely charged to the mutation in one or both of VL1 and VL2 to facilitate pairing, and

Wherein at least one or both of CH1-1 and CH1-2 comprises one or more of the T192E, L143Q, and S188V mutations, and at least one or both of CL1 and CL2 comprises one or more of the N137K, S114A, V133T, and S176V mutations.

In another aspect, the present disclosure provides a multispecific antigen-binding protein or antibody comprising:

a) a first light chain (LC 1)/heavy chain (HC1) pair, said first light chain (LC 1)/heavy chain (HC1) pair comprising

(1) A first VL region (VL1) that pairs with a first VH region (VH1) to form a first antigen binding site (VL 1);

(2) a first constant heavy chain region 1(CH1-1) and a first constant light chain region (CL 1); and

b) a second light chain (LC 2)/heavy chain (HC2) pair comprising

(3) A second VL region (VL2) that pairs with a second VH region (VH2) to form a second antigen binding site; and

(4) a second constant heavy chain region 1(CH1-2) and a second constant light chain region (CL 2);

wherein at least one or both of VH1 and VH2 comprises a Q39E, Q39D, Q39K, Q39R, or Q39H mutation, and one or both of VL1 and VL2 comprises a Q38E, Q38D, Q38K, Q38R, or Q38H mutation, wherein the mutation in one or both of VH1 and VH2 is oppositely charged to the mutation in one or both of VL1 and VL2 to facilitate pairing, and

Wherein at least one or both of CH1-1 and CH1-2 comprises one or more of the T192E, L143Q, and S188V mutations, and at least one or both of CL1 and CL2 comprises one or more of the N137K, S114A, V133T, and S176V mutations.

In another aspect, the present disclosure provides a multispecific antigen-binding protein or antibody comprising:

a) a first light chain (LC 1)/heavy chain (HC1) pair, said first light chain (LC 1)/heavy chain (HC1) pair comprising

(1) A first VL region (VL1) that pairs with a first VH region (VH1) to form a first antigen binding site (VL 1);

(2) a first constant heavy chain region 1(CH1-1) operably linked to VH1 and a first constant light chain region (CL1) operably linked to VL 1; and

(3) a first heterodimerization domain (HD 1); and

b) a second light chain (LC 2)/heavy chain (HC2) pair comprising

(4) A second VL region (VL2) that pairs with a second VH region (VH2) to form a second antigen binding site;

(5) a second constant heavy chain region 1(CH1-2) operably linked to VH2 and a second constant light chain region (CL2) operably linked to VL 2; and

(6) a second heterodimerization domain (HD 2);

wherein HD1 and HD2 heterodimerize,

Wherein at least one or both of CH1-1 and CH1-2 comprises a K221E, K221D, K221K, K221R, or K221H mutation, and one or both of CL1 and CL2 comprises an E123E, E123D, E123K, E123R, or E123H mutation, wherein the mutation in one or both of CH1-1 and CH1-2 is oppositely charged to the mutation in one or both of CL1 and CL2 to facilitate pairing, and

wherein at least one or both of CH1-1 and CH1-2 comprises one or more of the T192E, L143Q, and S188V mutations, and at least one or both of CL1 and CL2 comprises one or more of the N137K, S114A, V133T, and S176V mutations.

In another aspect, the present disclosure provides a multispecific antigen-binding protein or antibody comprising:

a) a first light chain (LC 1)/heavy chain (HC1) pair, said first light chain (LC 1)/heavy chain (HC1) pair comprising

(1) A first VL region (VL1) that pairs with a first VH region (VH1) to form a first antigen binding site (VL 1);

(2) a first constant heavy chain region 1(CH1-1) and a first constant light chain region (CL 1); and

b) a second light chain (LC 2)/heavy chain (HC2) pair comprising

(3) A second VL region (VL2) that pairs with a second VH region (VH2) to form a second antigen binding site; and

(4) A second constant heavy chain region 1(CH1-2) and a second constant light chain region (CL 2);

wherein at least one or both of CH1-1 and CH1-2 comprises a K221E, K221D, K221K, K221R, or K221H mutation, and one or both of CL1 and CL2 comprises an E123E, E123D, E123K, E123R, or E123H mutation, wherein the mutation in one or both of CH1-1 and CH1-2 is oppositely charged to the mutation in one or both of CL1 and CL2 to facilitate pairing, and

wherein at least one or both of CH1-1 and CH1-2 comprises one or more of the T192E, L143Q, and S188V mutations, and at least one or both of CL1 and CL2 comprises one or more of the N137K, S114A, V133T, and S176V mutations.

In another aspect, the present disclosure provides a multispecific antigen-binding protein or antibody comprising:

a) a first light chain (LC 1)/heavy chain (HC1) pair, said first light chain (LC 1)/heavy chain (HC1) pair comprising

(1) A first VL region (VL1) that pairs with a first VH region (VH1) to form a first antigen binding site (VL 1);

(2) a first constant heavy chain region 1(CH1-1) operably linked to VH1 and a first constant light chain region (CL1) operably linked to VL 1; and

(3) A first heterodimerization domain (HD 1); and

b) a second light chain (LC 2)/heavy chain (HC2) pair comprising

(4) A second VL region (VL2) that pairs with a second VH region (VH2) to form a second antigen binding site;

(5) a second constant heavy chain region 1(CH1-2) operably linked to VH2 and a second constant light chain region (CL2) operably linked to VL 2; and

(6) a second heterodimerization domain (HD 2);

wherein HD1 and HD2 heterodimerize,

wherein at least one or both of VH1 and VH2 comprises a Q39E, Q39D, Q39K, Q39R, or Q39H mutation, and one or both of VL1 and VL2 comprises a Q38E, Q38D, Q38K, Q38R, or Q38H mutation, wherein the mutation in one or both of VH1 and VH2 is oppositely charged to the mutation in one or both of VL1 and VL2 to facilitate pairing,

wherein at least one or both of CH1-1 and CH1-2 comprises a K221E, K221D, K221K, K221R, or K221H mutation, and one or both of CL1 and CL2 comprises an E123E, E123D, E123K, E123R, or E123H mutation, wherein the mutation in one or both of CH1-1 and CH1-2 is oppositely charged to the mutation in one or both of CL1 and CL2 to facilitate pairing, and wherein the mutations in one or both of CH1-1 and CH1-2 are oppositely charged to facilitate pairing, and wherein the mutations in one or both of CH1 and CL2 are oppositely charged to facilitate pairing

Wherein at least one or both of CH1-1 and CH1-2 comprises one or more of the T192E, L143Q, and S188V mutations, and at least one or both of CL1 and CL2 comprises one or more of the N137K, S114A, V133T, and S176V mutations.

In another aspect, the present disclosure provides a multispecific antigen-binding protein or antibody comprising:

a) a first light chain (LC 1)/heavy chain (HC1) pair, said first light chain (LC 1)/heavy chain (HC1) pair comprising

(1) A first VL region (VL1) that pairs with a first VH region (VH1) to form a first antigen binding site (VL 1);

(2) a first constant heavy chain region 1(CH1-1) and a first constant light chain region (CL 1); and

b) a second light chain (LC 2)/heavy chain (HC2) pair comprising

(3) A second VL region (VL2) that pairs with a second VH region (VH2) to form a second antigen binding site; and

(4) a second constant heavy chain region 1(CH1-2) and a second constant light chain region (CL 2);

wherein at least one or both of VH1 and VH2 comprises a Q39E, Q39D, Q39K, Q39R, or Q39H mutation, and one or both of VL1 and VL2 comprises a Q38E, Q38D, Q38K, Q38R, or Q38H mutation, wherein the mutation in one or both of VH1 and VH2 is oppositely charged to the mutation in one or both of VL1 and VL2 to facilitate pairing,

Wherein at least one or both of CH1-1 and CH1-2 comprises a K221E, K221D, K221K, K221R, or K221H mutation, and one or both of CL1 and CL2 comprises an E123E, E123D, E123K, E123R, or E123H mutation, wherein the mutation in one or both of CH1-1 and CH1-2 is oppositely charged to the mutation in one or both of CL1 and CL2 to facilitate pairing, and

wherein at least one or both of CH1-1 and CH1-2 comprises one or more of the T192E, L143Q, and S188V mutations, and at least one or both of CL1 and CL2 comprises one or more of the N137K, S114A, V133T, and S176V mutations.

In another aspect, the present disclosure provides a multispecific antigen-binding protein or antibody comprising:

a) a first light chain (LC 1)/heavy chain (HC1) pair, said first light chain (LC 1)/heavy chain (HC1) pair comprising

(1) A first VL region (VL1) that pairs with a first VH region (VH1) to form a first antigen binding site (VL 1);

(2) a first constant heavy chain region 1(CH1-1) operably linked to VH1 and a first constant light chain region (CL1) operably linked to VL 1; and

(3) a first heterodimerization domain (HD 1); and

b) a second light chain (LC 2)/heavy chain (HC2) pair comprising

(4) A second VL region (VL2) that pairs with a second VH region (VH2) to form a second antigen binding site;

(5) a second constant heavy chain region 1(CH1-2) operably linked to VH2 and a second constant light chain region (CL2) operably linked to VL 2; and

(6) a second heterodimerization domain (HD 2);

wherein HD1 and HD2 heterodimerize,

wherein at least one or both of VH1 and VH2 comprises a Q39E, Q39D, Q39K, Q39R, or Q39H mutation, and one or both of VL1 and VL2 comprises a Q38E, Q38D, Q38K, Q38R, or Q38H mutation, wherein the mutation in one or both of VH1 and VH2 is oppositely charged to the mutation in one or both of VL1 and VL2 to facilitate pairing, and

wherein at least one or two of CH1-1 and CH1-2 comprises one or more of the T192E, L143Q, and S188V mutations, and at least one or two of CL1 and CL2 comprises one or more of the N137K, S114A, V133T, and S176V mutations, and

wherein at least one or both of VH1 and VH2 comprises a 44C mutation and one or both of VL1 and VL2 comprises a 100C mutation.

In another aspect, the present disclosure provides a multispecific antigen-binding protein or antibody comprising:

a) A first light chain (LC 1)/heavy chain (HC1) pair, said first light chain (LC 1)/heavy chain (HC1) pair comprising

(1) A first VL region (VL1) that pairs with a first VH region (VH1) to form a first antigen binding site (VL 1);

(2) a first constant heavy chain region 1(CH1-1) and a first constant light chain region (CL 1); and

b) a second light chain (LC 2)/heavy chain (HC2) pair comprising

(3) A second VL region (VL2) that pairs with a second VH region (VH2) to form a second antigen binding site; and

(4) a second constant heavy chain region 1(CH1-2) and a second constant light chain region (CL 2);

wherein at least one or both of VH1 and VH2 comprises a Q39E, Q39D, Q39K, Q39R, or Q39H mutation, and one or both of VL1 and VL2 comprises a Q38E, Q38D, Q38K, Q38R, or Q38H mutation, wherein the mutation in one or both of VH1 and VH2 is oppositely charged to the mutation in one or both of VL1 and VL2 to facilitate pairing, and

wherein at least one or two of CH1-1 and CH1-2 comprises one or more of the T192E, L143Q, and S188V mutations, and at least one or two of CL1 and CL2 comprises one or more of the N137K, S114A, V133T, and S176V mutations, and

Wherein at least one or both of VH1 and VH2 comprises a 44C mutation and one or both of VL1 and VL2 comprises a 100C mutation.

In another aspect, the present disclosure provides a multispecific antigen-binding protein or antibody comprising:

a) a first light chain (LC 1)/heavy chain (HC1) pair, said first light chain (LC 1)/heavy chain (HC1) pair comprising

(1) A first VL region (VL1) that pairs with a first VH region (VH1) to form a first antigen binding site (VL 1);

(2) a first constant heavy chain region 1(CH1-1) and a first constant light chain region (CL 1); and

b) a second light chain (LC 2)/heavy chain (HC2) pair comprising

(3) A second VL region (VL2) that pairs with a second VH region (VH2) to form a second antigen binding site; and

(4) a second constant heavy chain region 1(CH1-2) and a second constant light chain region (CL 2);

wherein at least one or both of CH1-1 and CH1-2 comprises an L143E, L143D, L143K, L143R, or L143H mutation, and at least one or both of CL1 and CL2 comprises an S176E, S176D, S176K, S176R, or S176H mutation, and wherein the mutation in one or both of CH1-1 or CH1-2 is oppositely charged to the mutation in one or both of CL1 and CL 2.

In another aspect, the present disclosure provides a multispecific antigen-binding protein or antibody comprising:

a) a first light chain (LC 1)/heavy chain (HC1) pair, said first light chain (LC 1)/heavy chain (HC1) pair comprising

(1) A first VL region (VL1) that pairs with a first VH region (VH1) to form a first antigen binding site (VL 1);

(2) a first constant heavy chain region 1(CH1-1) and a first constant light chain region (CL 1); and

b) a second light chain (LC 2)/heavy chain (HC2) pair comprising

(3) A second VL region (VL2) that pairs with a second VH region (VH2) to form a second antigen binding site; and

(4) a second constant heavy chain region 1(CH1-2) and a second constant light chain region (CL 2);

wherein at least one or both of CH1-1 and CH1-2 comprises a L124E, L124D, L124K, L124R, or L124H mutation, and at least one or both of CL1 and CL2 comprises a V133E, V133D, V133K, V133R, or V133H mutation, and wherein the mutation in one or both of CH1-1 or CH1-2 is oppositely charged to the mutation in one or both of CL1 and CL 2.

In some embodiments, CH1-1 further comprises a T192E mutation, and CL1 comprises N137K and S114A mutations.

In some embodiments, CH1-2 comprises the T192E mutation, and CL2 comprises the N137K and S114A mutations.

In some embodiments, one or both of VH1 and VH2 comprises a Q39E, Q39D, Q39K, Q39R, or Q39H mutation, and one or both of VL1 and VL2 comprises a Q38E, Q38D, Q38K, Q38R, or Q38H mutation, wherein the mutation in one or both of VH1 and VH2 is oppositely charged to the mutation in one or both of VL1 and VL 2.

In some embodiments, the multispecific antibody further comprises one or more cysteine residues engineered into one or both of the VH1/VL1 and VH2/VL2 pairs to form one or more disulfide bonds.

In some embodiments, one or both of VH1 and VH2 comprises 44C and 105C mutations, and one or both of VL1 and VL2 comprises 100C and 43C mutations.

In some embodiments, VH1 comprises the 44C mutation, and VL1 comprises the 100C mutation.

In some embodiments, VH1 comprises the 105C mutation and VL1 comprises the 43C mutation.

In some embodiments, VH2 comprises the 44C mutation, and VL2 comprises 100C.

In some embodiments, VH2 comprises the 105C mutation and VL2 comprises the 43C mutation.

In some embodiments, VH1 comprises 39E and 44C mutations, and VL1 comprises 38K and 100C mutations.

In some embodiments, VH1 comprises the 39E and 105C mutations and VL1 comprises the 38K and 43C mutations.

In some embodiments, the multispecific antibody further comprises oppositely charged mutations in one or both of the CH1-1/CL1 pair and one or both of the CH1-2/CL2 pair.

In some embodiments, the oppositely charged mutations in one or both of the pair CH1-1/CL1 and one or both of the pair CH1-2/CL2 comprise K221E in one or both of the CH1-1 and CH2-2 regions and E123K in one or both of the CL1 and CL2 regions.

In some embodiments, the CH1-1 domain is operably linked to a first Fc domain comprising a first CH2 domain and a first CH3 domain, and the CH1-2 domain is operably linked to a second Fc domain comprising a second CH2 domain and a second CH3 domain, and wherein the first Fc domain and the second Fc domain dimerize.

In some embodiments, the first CH3 domain comprises one or both of the S354C and T366W mutations, the second CH3 domain comprises one or more of the Y349C, T366S, L368A, and Y407V mutations, and wherein the mutations promote heterodimerization of Fc domains.

In another aspect, the present disclosure provides an antigen binding protein comprising:

an antigen binding domain and a constant heavy chain CH1 region paired with a constant light chain CL region,

wherein the antigen binding domain selectively binds to a target antigen, and wherein the CH1 region and the CL region comprise one or both of:

a) L143E, L143D, L143K, L143R or L143H mutations in the CH1 region and S176E, S176D, S176K, S176R or S176H mutations in the CL region; and

b) the L124E, L124D, L124K, L124R or L124H mutations in the CH1 region and the V133E, V133D, V133K, V133R or V133H mutations in the CL region,

wherein the mutant residue in the CH1 region has an opposite charge to the mutant residue in the CL region.

In another aspect, the disclosure provides a binding protein comprising

A protein binding domain; and

a CH1 region, the CH1 region being paired with a CL region,

wherein the protein binding domain selectively binds to a target antigen, and wherein the CH1 region and the CL region comprise one or both of:

a) L143E, L143D, L143K, L143R or L143H mutations in the CH1 region and S176E, S176D, S176K, S176R or S176H mutations in the CL region; and

b) The L124E, L124D, L124K, L124R or L124H mutations in the CH1 region and the V133E, V133D, V133K, V133R or V133H mutations in the CL region,

wherein the mutation in the CH1 region is oppositely charged to the mutation in the CL region.

In another aspect, the disclosure provides an antigen binding protein comprising a constant heavy chain CH1 region paired with a constant light chain CL region,

wherein the antigen binding domain selectively binds to a target antigen, and wherein the CH1 region and the CL region comprise one or both of:

a) L143E, L143D, L143K, L143R or L143H mutations in the CH1 region and S176E, S176D, S176K, S176R or S176H mutations in the CL region; and

b) the K221E and K228D mutations in the CH1 region and the D122K and E123K mutations in the CL region,

wherein the mutant residue in the CH1 region has an opposite charge to the mutant residue in the CL region.

In some embodiments, the binding protein further comprises a K221E mutation in the CH1 region and an E123K mutation in the CL region.

In some embodiments, the present disclosure provides a multispecific antigen-binding protein comprising a first Fab comprising CH1-1, VH1, CL1, and VL1 domains; and a second Fab comprising CH1-2, VH2, CL2 and VL2 domains, wherein the first and second Fab are selected from one of the following alternatives:

i. The first Fab comprises a 143R mutation in CH1-1, a 39K mutation in VH1, a 176E mutation in CL1, a 38E mutation in VL1, and the second Fab comprises a 143E mutation in CH1-2, a 39E mutation in VH2, a 176R mutation in CL2, and a 38K mutation in VL 2;

the first Fab comprises a 143K mutation in CH1-1, a 39K mutation in VH1, a 176E mutation in CL1, a 38E mutation in VL1, and the second Fab comprises a 143E mutation in CH1-2, a 39E mutation in VH2, a 176K mutation in CL2, and a 38K mutation in VL 2;

the first Fab comprises a 143H mutation in CH1-1, a 39K mutation in VH1, a 176E mutation in CL1, a 38E mutation in VL1, and the second Fab comprises a 143E mutation in CH1-2, a 39E mutation in VH2, a 176H mutation in CL2, and a 38K mutation in VL 2;

the first Fab comprises a 143R mutation in CH1-1, a 39K mutation in VH1, a 176D mutation in CL1, a 38E mutation in VL1, and the second Fab comprises a 143D mutation in CH1-2, a 39E mutation in VH2, a 176R mutation in CL2, and a 38K mutation in VL 2;

v. the first Fab comprises a 143K mutation in CH1-1, a 39K mutation in VH1, a 176D mutation in CL1, a 38E mutation in VL1, and the second Fab comprises a 143D mutation in CH1-2, a 39E mutation in VH2, a 176K mutation in CL2, and a 38K mutation in VL 2;

The first Fab comprises a 143H mutation in CH1-1, a 39K mutation in VH1, a 176D mutation in CL1, a 38E mutation in VL1, and the second Fab comprises a 143D mutation in CH1-2, a 39E mutation in VH2, a 176H mutation in CL2, and a 38K mutation in VL 2.

In some embodiments, the CH1 region is operably linked to a heterodimerization domain.

In some embodiments, the heterodimerization domain comprises a first Fc domain.

In some embodiments, the first Fc domain is heterodimerized with a second Fc domain, and wherein the first Fc domain comprises a first CH3 region and the second Fc domain comprises a second CH3 region.

In some embodiments, the first CH3 region comprises one or both of the S354C and T366W mutations and the second CH3 region comprises one or more of the Y349C, T366S, L368A, and Y407V mutations, wherein the mutations promote Fc domain heterodimerization.

In some embodiments, the antigen binding protein further comprises at least one VH/VL pair comprising oppositely charged mutations that facilitate pairing, the oppositely charged mutations comprising (1) a mutant residue at Kabat position 39 in the VH region selected from E, D, K, R or H, and (2) a mutant residue at Kabat position 38 in the VL region selected from E, D, K, R or H, and wherein the mutant residue in the VH region has an opposite charge to the mutant residue in the VL region.

In some embodiments, the multispecific antigen-binding protein or antigen-binding protein comprises three HCDRs per VH region and three LCDRs per VL region, and further comprises binding specificity for one or more target antigens or one or more target epitopes. In some embodiments, the HCDR, the LCDR and/or the antigen are disclosed herein. In some embodiments, the HCDR, the LCDR and/or the antigen are known in the art. In some embodiments, the HCDR, the LCDR and/or the antigen are newly identified or newly discovered.

In some embodiments, the present disclosure provides an antigen binding protein comprising:

an antigen binding domain; and

a constant heavy chain CH1 region paired with a constant light chain CL region,

wherein the antigen binding domain selectively binds to a target antigen, and wherein the CH1 region and the CL region comprise one or both of:

a) L143E, L143D, L143K, L143R or L143H mutations in the CH1 region and S176E, S176D, S176K, S176R or S176H mutations in the CL region; and

b) the L124E, L124D, L124K, L124R or L124H mutations in the CH1 region and the V133E, V133D, V133K, V133R or V133H mutations in the CL region,

Wherein the mutant residue in the CH1 region has an opposite charge to the mutant residue in the CL region.

In some embodiments, the antigen binding protein further comprises a CH1/CL mutation selected from one or more of the following to facilitate pairing:

(1) the T192E (CH1) mutation and the N137K and S114A (CL) mutations,

(2) the L143Q and S188V (CH1) mutations and the V133T and S176V (CL) mutations,

(3) the T192E, L143Q and S188V (CH1) mutations and the N137K, S114A, V133T and S176V (CL) mutations,

(4) the K221E (CH1) mutation and the E123K (CL) mutation,

(5) the K228D (CH1) mutation and the D122K (CL) mutation, and

(6) the K221E and K228D (CH1) mutations and the D122K and E123K (CL) mutations,

wherein when two CH1/CL pairs comprise a mutation to facilitate pairing of two different VH/VL pairs, the two CH1/CL pairs do not comprise the same mutation.

In some embodiments, the CH1 region is operably linked to an Fc domain.

In some embodiments, an isolated nucleic acid molecule is provided that comprises a nucleotide sequence encoding the multispecific antibody or the antigen-binding protein. In some embodiments, a kit is provided comprising one or more isolated nucleic acid molecules comprising one or more nucleotide sequences encoding the multispecific antigen-binding protein or the antigen-binding protein.

In some embodiments, an expression vector is provided comprising the nucleic acid molecule. In some embodiments, a kit is provided comprising one or more expression vectors comprising one or more of the nucleic acid molecules.

In some embodiments, an isolated host cell is provided comprising the one or more nucleic acid molecules or the one or more expression vectors. In some embodiments, an isolated host cell is provided comprising the kit of nucleic acid molecules or the kit of expression vectors.

In some embodiments, the host cell is a mammalian cell or an insect cell.

In some embodiments, a pharmaceutical composition is provided comprising a pharmaceutically acceptable carrier and a therapeutically effective amount of the multispecific antibody or the antigen-binding protein.

In some embodiments, there is provided a method of treating a disorder in which antigen activity is detrimental, the method comprising administering to a subject in need thereof an effective amount of a multispecific antibody or antigen-binding protein.

In some embodiments, a polynucleotide is provided that encodes a multispecific antibody or antigen-binding protein.

In some embodiments, a host cell is provided that expresses a multispecific antibody or antigen-binding protein.

In some embodiments, a method of producing a multispecific antibody or antigen-binding protein is provided, the method comprising culturing a host cell under conditions such that the multispecific antibody or antigen-binding protein is expressed. In some embodiments, a multispecific antibody or antigen-binding protein is provided for use as a medicament.

The above summary is not limiting, and other features and advantages of the disclosed antigen binding proteins and methods will be apparent from the following brief description of the drawings, detailed description, and claims.

Drawings

FIGS. 1A-1C schematically depict a form of cross-duplex variable (CODV) antigen binding protein with several different mutations to enhance heterodimerization. FIG. 1A depicts the CH 1/kappa mutation: the "O" on the CODV-Fab (CH 1: L143Q, S188V; Ck: V133T, S176V) and the "Δ" on the Fab2 (CH 1: T192E; Ck: N137K, S114A). FIG. 1B depicts mutations on the CODV-Fab (VH39E/VL38K + O) and on the Fab2 (VH39K/VL38E + Δ). FIG. 1C depicts mutations in the CODV-Fab ([ VH39E/VL38K + O ] + [ VH44Cys/VL100Cys ] or [ VH105Cys/VL43Cys ]) and in the Fab2 ([ VH39K/VL38E + Δ ] + [ VH44Cys/VL100Cys ] or [ VH105Cys/VL43Cys ]).

Fig. 2A-2G graphically depict the results of table 5. FIG. 2A depicts the results of WT CODV antibodies. FIG. 2B depicts the results for a CODV antibody with only the CH1/Ck mutation. FIG. 2C depicts the results of CH1/Ck and Charge Mutation (CM) combinations. Figure 2D depicts the results for disulfide-only stabilized (ds) CODV antibodies. FIG. 2E depicts the results of the combination of CH1/Ck and ds mutations. Figure 2F depicts the results of CM and ds mutation combinations. FIG. 2G depicts the results of the combination of all three mutation groups (i.e., CH1/Ck, CM and ds-stabilized) in the CODV antibody.

Fig. 3A-3D schematically depict tandem Fab antibody formats in an open configuration (fig. 3A, 3B) and a closed configuration (fig. 3C, 3D). FIG. 3A shows an open configuration with the CH 1/kappa mutation ("Δ" on Fab1 (CH 1: T192E; Ck: N137K, S114A) and "O" on Fab2 (CH 1: L143Q, S188V; Ck: V133T, S176V)). FIG. 3B shows an open configuration with a mutation on Fab1 (VH39K/VL38E + Δ) and a mutation on Fab2 (VH39E/VL38K + O). FIG. 3C shows the closed configuration with the CH 1/kappa mutation (Δ on Fab1 (CH 1: T192E; Ck: N137K, S114A) and O on Fab2 (CH 1: L143Q, S188V; Ck: V133T, S176V)). FIG. 3D shows the closed configuration with the mutations at Fab1 (VH39K/VL38E + Δ) and Fab2 (VH39E/VL38K + O).

FIG. 4 depicts the results of SEC and HIC analysis of tandem Fab in open configuration with WT, CH1/Ck and CH1/Ck + CM forms.

Fig. 5A-5D depict results of HIC analysis, yield and binding affinity of anti-CD 40 x anti-PD-L1 tandem Fab antibodies in open and closed configurations. FIG. 5A shows the closed configuration with only the CH1/Ck mutation. Figure 5B shows the closed configuration with Fab domain exchange with only CH1/Ck mutation. FIG. 5C shows the closed configuration with the CH1/Ck and CM mutations. FIG. 5D shows the open configuration with CH1/Ck and CM mutations.

Fig. 6A-6C depict results of HIC analysis, yield, and binding affinity of anti-PD-1 x anti-OX 40 tandem Fab antibodies in open and closed configurations. FIG. 6A shows the closed configuration with only the CH1/Ck mutation. FIG. 6B shows an open configuration with only the CH1/Ck mutation. FIG. 6C shows the open configuration with CH1/Ck and CM mutations.

Fig. 7A-7B depict purity results for anti-4-1 BB x anti-PD-L1 tandem Fab antibody (fig. 7A) and anti-4-1 BB x anti-PD-1 tandem Fab antibody (fig. 7B). Both antibodies were in an open configuration with CH1/Ck and CM mutations.

Fig. 8A-8B schematically depict a Y-shaped bispecific antibody format with several different mutations to enhance heterodimerization. FIG. 8A shows the CH 1/kappa mutation: o "on Fab1 (CH 1: L143Q, S188V; Ck: V133T, S176V) and Δ on Fab2 (CH 1: T192E; Ck: N137K, S114A). FIG. 8B shows the mutations at Fab1 (VH39E/VL38K + O) and at Fab2 (VH39K/VL38E + Δ).

Fig. 9A-9C depict purity results for anti-PD-1 x anti-OX 40 antibodies. The thermal stability of the antibody at the onset of T was measured using NanoDSF (differential scanning fluorimetry). Purity was measured using HIC. The wild-type antibody is shown in fig. 9A. The CH1/Ck mutations (Fab1 ═ CH 1: L143Q, S188V; Ck: V133T, S176V) and (Fab2 ═ CH 1: T192E; Ck: N137K, S114A) are shown in fig. 9B. CH1/Ck and CM mutations (Fab1 ═ CH 1: L143Q, S188V; Ck: V133T, S176V; VH39E/VL38K) and (Fab2 ═ CH 1: T192E; Ck: N137K, S114A; VH39K/VL38E) are shown in fig. 9C.

FIGS. 10A-10L depict SEC and HIC profiles of various anti-PD-1 x anti-OX 40 antibodies with several different combinations of mutations. Specific mutations are listed in table 9 below.

Fig. 11A-11E depict chain mismatch data for various Y-shaped antibodies. FIG. 11A depicts mispaired data for anti-PD-1 x anti-GITR antibody. Figure 11B depicts mismatching data for anti-TNF x anti-GITR antibodies. FIG. 11C depicts mismatching data for anti-TNF x anti-OX 40 antibodies. Figure 11D depicts mismatching data for anti-CD 40 x anti-PD-L1 antibody. Figure 11E depicts the mismatching data for anti-CD 3 x anti-CD 123 antibodies. Specific mutations and biophysical characterization data are listed in table 11 below.

FIG. 12 depicts the cytotoxicity assay of human panT cells against THP-1 target cells co-incubated with bispecific CD3 x CD123-hIgG1-LALA molecules. T effector cells and CFSE-labeled THP-1 target cells were seeded at an effector to target ratio of 10:1 and incubated with serial dilutions (10nM to 0nM) of the corresponding bispecific molecule for 20h at 37 ℃. Dead cells were stained with 7-AAD and measured by flow cytometry. Cytotoxic activity was calculated based on the percentage of dead THP-1 target cells (7-AAD/CFSE double positive). The data show the relationship of dead target cells [% ] to bispecific molecule concentration [ pM ] as the average of two representative healthy donors.

Detailed Description

I.Definition of

In order that the disclosure may be more readily understood, selected terms are defined below.

As used herein, sequence position numbering refers to Kabat numbering (Kabat, E.A. et al, Sequences of proteins of immunological interest. 5 th edition-US Department of Health and Human Services, NIH publication No. 91-3242, pages 662, 680, 689, 1991).

As used herein, the twenty conventional amino acids and their abbreviations follow conventional usage. Stereoisomers of twenty conventional amino acids (e.g., D-amino acids); unnatural amino acids (e.g., a-disubstituted amino acids, N-alkyl amino acids, lactic acid, and other non-conventional amino acids) can also be suitable components of the polypeptide chains of the binding proteins described herein. Examples of unconventional amino acids include: 4-hydroxyproline, y-carboxyglutamic acid, c-N, N, N-trimethyllysine, c-N-acetyl lysine, O-phosphoserine, N-acetyl serine, N-formyl methionine, 3-methylhistidine, 5-hydroxylysine, u-N-methyl arginine, and other similar amino and imino acids (e.g., 4-hydroxyproline). In the polypeptide notation used herein, the left-hand direction is the amino-terminal direction and the right-hand direction is the carboxy-terminal direction, according to standard usage and convention. Naturally occurring residues can be classified into a number of classes based on common side chain properties (see table 1).

Table 1-amino acid residues provided by category.

Conservative amino acid substitutions may involve the exchange of a member of one of these classes for another member of the same class. Conservative amino acid substitutions may encompass non-naturally occurring amino acid residues that are typically incorporated by chemical peptide synthesis rather than by biological system synthesis. These include peptidomimetics and other inverted (reversed) or inverted (inverted) forms of amino acid residues. Non-conservative substitutions may involve the exchange of a member of one of these classes for a member from another class.

As used herein, the term "mutation" or "mutated" refers to a change in the amino acid sequence by deletion, insertion and/or substitution of one or more amino acids. In particular, the term refers to substitution. Mutations are introduced with respect to a given sequence (e.g., the amino acid sequence of a VL1 and/or VH1 pair that specifically recognizes a first antigen).

As used herein, a "T192E (CH1) mutation" refers to a substitution of a threonine (T) residue to a glutamic acid (E) residue at Kabat position 192 in the immunoglobulin CH1 heavy chain constant domain of an antigen binding protein.

As used herein, "set of mutations" refers to a set of different mutations present in a sequence.

The term "variant" as used herein refers to an amino acid sequence that is at least 80%, 85%, 90%, 95%, 96%, 97%, 98% or 99% identical to the amino acid sequence from which it is derived. The determination of the percent identity between two sequences is accomplished using the mathematical algorithm of Karlin and Altschul, Proc.Natl.Acad.Sci.USA 90,5873-5877, 1993. Such algorithms are incorporated into the BLASTN and BLASTP programs of Altschul et al (1990) J.mol.biol.215, 403-410. To obtain a gapped alignment for comparison purposes, gapped BLAST was used, as described in Altschul et al (1997) Nucleic Acids Res.25, 3389-3402. When utilizing BLAST and gapped BLAST programs, the default parameters of the respective programs are used. Alternatively, variants may also be defined as having up to 20, 15, 10, 5,4, 3, 2 or 1 amino acid substitutions, in particular conservative amino acid substitutions. Conservative substitutions are well known in the art (see, e.g., Creighton (1984) proteins. W.H.Freeman and Company). A summary of the physical and chemical properties of the amino acids is given in table 1 above. In particular embodiments, a conservative substitution is a substitution with amino acids that share at least one property according to table 1 (i.e., column 1 and/or column 2). The term "variant" also includes fragments. Fragments have N-terminal and/or C-terminal deletions amounting to up to 20, 15, 10, 5,4, 3, 2 or 1 amino acids. Additionally or alternatively, the variant may be modified, for example by N-terminal and/or C-terminal amino acid additions of up to a total of 50, 40, 30, 20, 10, 5,4, 3, 2 or 1 amino acids.

As used herein, the term "antigen" or "target antigen" or "antigen target" refers to a molecule or a portion of a molecule (e.g., an epitope) that is capable of being specifically bound by a binding protein described herein and, in addition, is capable of being used in an animal to produce an antibody that is capable of specifically binding to an epitope of the antigen. The target antigen may have one or more epitopes. For each target antigen recognized by a binding protein, the binding protein is capable of competing with an intact antibody that recognizes the target antigen.

The term "epitope" as used herein means capable of interacting withAny determinant, such as a polypeptide determinant, to which an immunoglobulin or T cell receptor specifically binds. In certain embodiments, epitope determinants include chemically active surface groups (groupings) of molecules, such as amino acids, sugar side chains, phosphoryl groups, or sulfonyl groups, and in certain embodiments, may have specific three-dimensional structural characteristics and/or specific charge characteristics. An epitope is a region of an antigen bound by an antibody or by an antigen-binding fragment of an antibody or by a binding protein. In certain embodiments, a binding protein is said to specifically bind an antigen when it preferentially recognizes its target antigen in a complex mixture of proteins and/or macromolecules. In some embodiments, when the equilibrium dissociation constant is <10-8When M is equal to<1-9M, or when the dissociation constant is<10-10M, the binding protein is said to specifically bind to the antigen.

As used herein, the term "antigen binding protein" or "binding polypeptide" refers to a polypeptide (e.g., an antibody or fragment thereof) that contains at least one binding site responsible for selectively binding to a target antigen of interest (e.g., a human antigen). Exemplary binding sites include, but are not limited to, antibody variable domains, ligand binding sites of receptors, or receptor binding sites of ligands. In certain aspects, a binding polypeptide comprises a plurality (e.g., two, three, four, or more) binding sites. In certain aspects, the binding protein is not a therapeutic enzyme.

As used herein, "heterodimerization domain" or "HD" refers to a subunit of a bispecific, trispecific, or multispecific binding protein that facilitates, directs, or forces the correct assembly of a light chain and its cognate heavy chain to produce a desired protein while preventing mispairing of the corresponding light or heavy chains.

As used herein, a "multispecific" binding protein is a binding protein that binds two or more antigens, and/or two or more different epitopes. Multispecific binding proteins that bind two antigens and/or two different epitopes are also referred to herein as "bispecific" binding proteins. Multispecific binding proteins that bind three antigens and/or three different epitopes are also referred to herein as "trispecific" binding proteins.

As used herein, the term "heterodimeric Fc" or "functional fragment of a heterodimeric Fc" refers to a mutant form of a constant domain (e.g., CH2-CH3 or CH2-CH3-CH4) that is mutated with respect to a naturally occurring Fc portion, wherein it no longer forms a homodimer, but rather forms a heterodimer with a corresponding mutated Fc portion. Thus, the term refers to a portion of the two chains that form the heterodimer. Several such pairs are known in the art and include, for example, knob-in-hole (KIH) variants or EV-RWT variants.

Ridgeway and coworkers generated the CH3 interface that favors heterodimer assembly by: the small side chain on one CH3 interface was replaced with the larger side chain to create a pestle, and the large side chain on the other CH3 domain was replaced with the smaller side chain to create a hole. Testing such variants showed preferential heterodimerization. The original knob structure mutations were further expanded by phage display to identify additional suitable combinations for generating bispecific IgG antibodies to test for additional substitutions that allow disulfide bond formation. Variations of the knob and hole structure are further described in U.S. patent No. 5,732,168 and U.S. patent No. 8,216,805, which are incorporated herein by reference. Thus, in one embodiment, the CH3 domain of one Fc domain or heterodimerization domain contains the mutations Y349C, T366S, L368A and Y407V, and the CH3 domain of the other Fc domain or heterodimerization domain contains the mutations S354C and T366W (amino acid positions are indicated by reference to the IgG1 sequence).

As used herein, the term "homodimerization domain" refers to a domain that mediates homodimerization of two similar domains (e.g., two heavy chains). Heavy chain pairing is mediated by the last domain of the constant region (i.e., CH3 in IgG molecules) that forms a high affinity homodimeric complex (K)DAbout 10 pM). Additional interactions are present in the hinge region responsible for covalent linkage of the two heavy chains formed upon heavy chain assembly. The interaction in the CH3 homodimer involves approximately 16 residues at the CH3-CH3 interfaceGroup, as shown for human γ 1CH3, where a patch (patch) formed by 6 residues (T366, L368, F405, Y407 and K409) at the center of the interface strongly promotes stability. Homodimerization domains include, but are not limited to, an Fc region and effector modified variants thereof, as well as fragments of any of the Fc region and effector modified variants thereof, a CH2 domain or fragment thereof, a CH3 domain or fragment thereof, a CH4 domain or fragment thereof, and the like.

Naturally occurring antibodies typically comprise tetramers. Each such tetramer is typically composed of two identical pairs of polypeptide chains, each pair having one full length "light" chain (typically having a molecular weight of about 25 kDa) and one full length "heavy" chain (typically having a molecular weight of about 50-70 kDa). As used herein, the terms "heavy chain" and "light chain" refer to any immunoglobulin polypeptide having a variable domain sequence sufficient to confer specificity for a target antigen. The amino-terminal portion of each light and heavy chain typically includes a variable domain of about 100 to 110 or more amino acids, which is typically responsible for antigen recognition. The carboxy-terminal portion of each chain typically defines a constant domain responsible for effector function. Thus, in naturally occurring antibodies, a full-length heavy chain IgG immunoglobulin polypeptide comprises one variable domain (VH) and three constant domains (CH1, CH2, and CH3), wherein the VH domain is located amino-terminal to the polypeptide and the CH3 domain is located carboxy-terminal, and a full-length light chain immunoglobulin polypeptide comprises one variable domain (VL) and one constant domain (CL), wherein the VL domain is located amino-terminal to the polypeptide and the CL domain is located carboxy-terminal.

In some embodiments, the multispecific antigen-binding proteins of the present disclosure comprise one or more VH domains from any one of the VH domain sequences listed in tables 2, 3, and 4. In some embodiments, the multispecific antigen-binding proteins of the present disclosure comprise one or more VL domains from any one of the VL domain sequences listed in tables 2, 3, and 4. In some embodiments, the multispecific antigen-binding proteins of the present disclosure comprise one or more VH domains from any one of the VH domain sequences listed in tables 2, 3, and 4 paired with one or more VL domains from any one of the VL domain sequences listed in tables 2, 3, and 4.

Human light chains are generally classified as kappa and lambda light chains, and human heavy chains are generally classified as mu, delta, gamma, alpha or epsilon, and the isotypes of antibodies are defined as IgM, IgD, IgG, IgA, and IgE, respectively. IgG has several subclasses, including but not limited to IgG1, IgG2, IgG3, and IgG 4. IgM has multiple subclasses, including but not limited to IgM1 and IgM 2. IgA is similarly subdivided into multiple subclasses, including but not limited to IgA1 and IgA 2. Within full-length light and heavy chains, the variable and constant domains are typically joined by a "J" region of about 12 or more amino acids, and the heavy chain also includes a "D" region of about 10 or more amino acids. See, e.g., Fundamental Immunology (Paul, w. editor, Raven Press, 2 nd edition, 1989), which is incorporated by reference in its entirety for all purposes. The variable region of each light/heavy chain pair typically forms an antigen binding site. The variable domains of naturally occurring antibodies typically exhibit the same overall structure of relatively conserved Framework Regions (FRs) joined by three hypervariable regions (also known as complementarity determining regions or CDRs). The CDRs from both chains of each pair are typically aligned by the framework regions, which may enable binding to a specific epitope. From amino-terminus to carboxy-terminus, both light and heavy chain variable domains typically comprise the domains FR1, CDR1, FR2, CDR2, FR3, CDR3 and FR 4.

As used herein, the term "set of CDRs" refers to a set of three CDRs present in a single variable region capable of binding antigen. The exact boundaries of these CDRs have been defined in different ways depending on the different systems. It has been found by the system described by Kabat (Kabat et al, SEQUENCES OF PROTEINS OF IMMUNOLOGICAL INTEREST (National Institutes OF Health, Bethesda, MD (1987) and (1991)) not only provides a clear residue numbering system for any variable region OF an antibody, but also provides the exact residue boundaries defining the three CDRs which can be referred to as Kabat CDRs, Chothia and coworkers (Chothia and Lesk,1987, J.Affol.biol.196: 901-17; Chothia et al, 1989, Nature 342:877-83), despite significant diversity at the amino acid sequence level, certain sub-portions within the Kabat CDRs adopt almost identical peptide backbone conformations which are designated L1, L2 and L3 or H1, H2 and H3, where "L" and "H" designate light chain regions respectively which can be referred to light chain regions which overlap with other CDRs in the Kabat CDRs which are described by Kabat et al Kabat CDRs, 1995, FASEB J.9: 133-39; MacCallum,1996, J.mol.biol.262(5): 732-45; and Lefranc,2003, Dev.Comp.Immunol.27: 55-77. Still other CDR boundary definitions may not strictly follow the system described herein, but still overlap with the Kabat CDRs, although they may be shortened or lengthened given that a particular residue or group of residues, or even the entire CDR, does not significantly affect prediction or experimental findings of antigen binding. The methods used herein may utilize CDRs defined according to any of these systems, but certain embodiments use Kabat or Chothia defined CDRs. The use of amino acid sequences to identify predicted CDRs is well known in the art, as in the following references: martin, a.c. "Protein sequence and structure analysis of Antibody variable domains," Antibody Engineering, vol.2. Kontermann r., Dikel s. editor Springer-Verlag, Berlin, p.33-51 (2010). The amino acid sequences of the heavy and/or light chain variable domains can also be examined to identify the sequence of the CDRs by other conventional methods (e.g., by comparison to known amino acid sequences of other heavy and light chain variable regions to determine regions with sequence hyperdegeneration). The numbered sequences can be aligned by visual inspection, or by using an alignment program (such as one of the CLUSTAL program suite), as described in: thompson,1994, Nucleic Acids Res.22: 4673-80. Molecular models are routinely used to correctly delineate the framework and CDR regions and thereby correct sequence-based alignments.

In some embodiments, the CDR/FR of an immunoglobulin light or heavy chain is determined based on the IMGT definition (Lefranc et al Dev. Comp. Immunol.,2003,27(1): 55-77; website: IMGT. org).

In some embodiments, the multispecific antigen-binding protein of the present disclosure comprises 3 variable heavy chain cdrs (hcdrs) from any one of the variable heavy chain sequences listed in tables 2, 3, and 4. In some embodiments, the multispecific antigen-binding protein of the present disclosure comprises 3 variable light chain cdrs (lcdrs) from any one of the variable light chain sequences listed in tables 2, 3, and 4. In some embodiments, the multispecific antigen-binding protein of the present disclosure comprises 3 HCDRs from any one of the variable heavy chain sequences listed in tables 2, 3, and 4 and 3 LCDRs from any one of the variable light chain sequences listed in tables 2, 3, and 4. The CDR sequences of the heavy and light chain variable sequences from tables 2, 3 and 4 can be readily determined by those skilled in the art using art-recognized methods for identifying CDR sequences.

As used herein, the term "Fc" refers to a molecule (in monomeric or multimeric form) that comprises a sequence of a non-antigen-binding fragment produced by antibody digestion or by other means, and may contain a hinge region. While IgG1 and IgG2 are used in the exemplary embodiment, the original immunoglobulin source of the native Fc is generally of human origin and can be any immunoglobulin. Fc molecules consist of monomeric polypeptides that can be joined into dimeric or multimeric forms by covalent (i.e., disulfide) and non-covalent bonding. The number of intermolecular disulfide bonds between the monomeric subunits of a native Fc molecule ranges from 1 to 4, depending on the class (e.g., IgG, IgA, and IgE) or subclass (e.g., IgG1, IgG2, IgG3, IgA1, and IgGA 2). An example of an Fc is a disulfide-bonded dimer resulting from papain digestion of IgG. As used herein, the term "native Fc" is generic to monomeric, dimeric and multimeric forms.

Fab fragments typically comprise one light chain and one heavy chain of the VH and CH1 domains, where the VH-CH1 heavy chain portion of the F (ab) fragment is incapable of forming a disulfide bond with another heavy chain polypeptide. As used herein, a Fab fragment may also include a light chain comprising two variable domains separated by an amino acid linker and a CL domain, and a heavy chain comprising two variable domains separated by an amino acid linker and a CH1 domain.

F (ab ') fragments typically comprise a light chain and a portion of a heavy chain containing more constant regions (between the CH1 and CH2 domains) such that interchain disulfide bonds can be formed between the two heavy chains to form a F (ab') 2 molecule.

As used herein, the term "binding protein" refers to a non-naturally occurring (or recombinant, engineered, or substituted) molecule that specifically binds to at least one target antigen.

As used herein, the term "Tm" refers to the melting temperature of a binding protein, antigen binding protein, antibody, and is a key parameter for the thermal stability of an antigen binding protein. Tm is generally related to the thermal stability of the Fv fragment, i.e., the variable region heavy and light chains (VH/VL). Tm can be measured by Differential Scanning Calorimetry (DSC) or Differential Scanning Fluorimetry (DSF).

One embodiment of the present disclosure provides binding proteins having biological and immunological specificity for between one and four target antigens and/or specificity for between one and four target epitopes. Another embodiment of the disclosure provides a nucleic acid molecule comprising a nucleotide sequence encoding a polypeptide chain forming such a binding protein. Another embodiment of the present disclosure provides an expression vector comprising a nucleic acid molecule comprising a nucleotide sequence encoding a polypeptide chain forming such a binding protein. Yet another embodiment of the present disclosure provides a host cell that expresses such a binding protein (i.e., a nucleic acid molecule or vector comprising a polypeptide chain encoding such a binding protein).

The term "linker" as used herein refers to 0-100 consecutive amino acid residues. Linkers are present or absent and are the same or different. The linkers included in the protein or polypeptide may all have the same amino acid sequence or may have different amino acid sequences.

In some embodiments, the peptide linker comprises the following sequence: EPKSCDKTHTSPPSPAPELLGGPSTPPTPSPSGG (SEQ ID NO: X).

In some embodiments, the term "linker" refers to 1-15 contiguous amino acid residues. Typically, the linker provides flexibility and spatial separation between two amino acids or between two polypeptide domains. Linkers may be inserted between the VH, VL, CH and/or CL domains depending on the form of the molecule to provide sufficient flexibility and mobility for the domains of the light and heavy chains, e.g., to fold into a crossed double variable region immunoglobulin. At the amino sequence level, linkers are typically inserted between the variable domains, between the variable domain and the knockout domain, or at the transition between the variable domain and the constant domain, respectively. Since the approximate size of immunoglobulin domains is well understood, transitions between domains can be identified. The exact location of the domain transition can be determined by locating a peptide stretch that does not form a secondary structural element (such as a beta sheet or alpha helix) as evidenced by experimental data or as can be determined by techniques of modeling or secondary structure prediction. In certain exemplary embodiments, a linker may be inserted between the Fab domains to generate a tandem Fab antibody. In particular embodiments, a linker may be inserted between the N-terminus of the VH domain of the first Fab and the C-terminus of the CH1 domain of the second Fab.

The identity and sequence of the amino acid residues in the linker may vary depending on the type of secondary structural element or elements that are desired to be achieved in the linker. For example, glycine, serine and alanine are suitable for the linker with the greatest flexibility. Certain combinations of glycine, proline, threonine, and serine are useful if a more rigid and extended linker is desired. Depending on the desired properties, any amino acid residue in combination with other amino acid residues may be considered a linker as necessary to construct a larger peptide linker.

In some embodiments, the linker comprises: a single glycine (Gly) residue; diglycine peptide (Gly-Gly); tripeptide (Gly-Gly); a peptide having four glycine residues (Gly-Gly-Gly-Gly; SEQ ID NO: x); a peptide having five glycine residues (Gly-Gly-Gly-Gly-Gly; SEQ ID NO: x); a peptide having six glycine residues (Gly-Gly-Gly-Gly-Gly-Gly; SEQ ID NO: x); a peptide having seven glycine residues (Gly-Gly-Gly-Gly-Gly-Gly-Gly-Gly; SEQ ID NO: x); and a peptide having eight glycine residues (Gly-Gly-Gly-Gly-Gly; SEQ ID NO: x).

In some embodiments, the linker comprises a small amino acid, such as Gly, Ala, or Ser.

In some embodiments, the linker comprises gly (g) and ser(s), or GS, GGS, GGGS, or GGGGS. In some embodiments, the linker comprises (Gly-Gly-Gly-Gly-Ser)2(i.e., (GGGGS)2). In some embodiments, the linker comprises (Gly-Gly-Gly-Gly-Ser)3(i.e., (GGGGS)3)。

In some embodiments, the linker comprises a Gly-Gly-Gly-Gly-Ser (SEQ ID NO: x), a peptide Gly-Gly-Gly-Gly-Ser (SEQ ID NO: x), and a peptide Gly-Gly-Ser-Gly-Ser-Ser-Ser-Gly-Ser-Gly-Ser-Gly-Gly-Gly (SEQ ID NO: x).

In some embodiments, the linker comprises a single Ser residue; a single Val residue; a dipeptide selected from: Arg-Thr, Gln-Pro, Ser-Ser, Thr-Lys, and Ser-Leu; or a polypeptide selected from: Thr-Lys-Gly-Pro-Ser (SEQ ID NO: x), Thr-Val-Ala-Ala-Pro (SEQ ID NO: x), Gln-Pro-Lys-Ala-Ala (SEQ ID NO: x), Gln-Arg-Ile-Glu-Gly (SEQ ID NO: x), Ala-Ser-Thr-Lys-Gly-Pro-Ser (SEQ ID NO: x), Arg-Thr-Val-Ala-Ala-Pro-Ser (SEQ ID NO: x), Gly-Gln-Pro-Lys-Ala-Ala-Pro (SEQ ID NO: x), His-Ile-Asp-Ser-Pro-Asn-Lys (SEQ ID NO: x), and Asp-Lys-Thr-His-Thr (SEQ ID NO: x).

In some embodiments, two tandem Fab passes through (Gly-Gly-Gly-Gly-Ser)2And (4) connecting by using a joint.

In some embodiments with a CODV-Fab portion, wherein L1 and L2 are located on the light chain and L3 and L4 are located on the heavy chain, L1 is 3 to 12 amino acid residues in length, L2 is 3 to 14 amino acid residues in length, L3 is 1 to 8 amino acid residues in length, and L4 is 1 to 3 amino acid residues in length. In some embodiments, L1 is 5 to 10 amino acid residues in length, L2 is 5 to 8 amino acid residues in length, L3 is 1 to 5 amino acid residues in length, and L4 is 1 to 2 amino acid residues in length. In some embodiments, L1 is 7 amino acid residues in length, L2 is 5 amino acid residues in length, L3 is 1 amino acid residue in length, and L4 is 2 amino acid residues in length. In some embodiments, L1 is 10 amino acid residues in length, L2 is 10 amino acid residues in length, L3 is 0 amino acid residues in length, and L4 is 0 amino acid residues in length. In some embodiments, L1, L2, L3, and L4 each have an independently selected length of 0 to 15 amino acids (e.g., 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15 amino acids), wherein at least two linkers have a length of 1 to 15 amino acids (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15 amino acids). In some embodiments, L1, L2, L3, and L4 are Asp-Lys-Thr-His-Thr (SEQ ID NO: x). In some embodiments, one or more linkers comprise the sequence Gly-Gln-Pro-Lys-Ala-Ala-Pro (SEQ ID NO: x). In some embodiments, L1 comprises the sequence Gly-Gln-Pro-Lys-Ala-Ala-Pro (SEQ ID NO: x). In some embodiments, L1 comprises the sequence Gly-Gln-Pro-Lys-Ala-Ala-Pro (SEQ ID NO: x), L2 comprises the sequence Thr-Lys-Gly-Pro-Ser-Arg (SEQ ID NO: x), L3 comprises the sequence Ser, and L4 comprises the sequence Arg-Thr. In some embodiments, L3 comprises the sequence Gly-Gln-Pro-Lys-Ala-Ala-Pro (SEQ ID NO: x). In some embodiments, L1 comprises the sequence Ser, L2 comprises the sequence Arg-Thr, L3 comprises the sequence Gly-Gln-Pro-Lys-Ala-Ala-Pro (SEQ ID NO: x), and L4 comprises the sequence Thr-Lys-Gly-Pro-Ser-Arg (SEQ ID NO: x).

In some embodiments, L1, L2, L3, and L4 each independently comprise a sequence selected from: (Gly-Gly-Gly-Gly-Ser)n(wherein n is an integer between 0 and 5; SEQ ID NO: x), Gly-Gly-Gly-Gly-Ser-Gly-Gly-Gly-Gly-Gly-Ser (SEQ ID NO: x), Gly-Gly-Gly-Gly-Ser-Gly-Gly-Gly-Gly-Gly-Ser (SEQ ID NO: x), Ser, Arg-Thr, Thr-Lys-Gly-Pro-Ser (SEQ ID NO: x), Gly-Gln-Pro-Lys-Ala-Pro (SEQ ID NO: x), and Gly-Gly-Ser-Gly-Ser-Ser-Gly-Ser-Gly-Ser-Gly-Gly (SEQ ID NO: x). In some embodiments, L1 comprises the sequence Gly-Gln-Pro-Lys-Ala-Ala-Pro (SEQ ID NO: x), L2 comprises the sequence Thr-Lys-Gly-Pro-Ser (SEQ ID NO: x), L3 comprises the sequence Ser, and L4 comprises the sequence Arg-Thr. In some embodiments, L1 comprises the sequence Gly-Gly-Gly-Gly-Gly-Ser-Gly-Gly-Gly-Gly-Gly-Ser (SEQ ID)X), L2 comprises the sequence Gly-Gly-Gly-Gly-Gly-Ser (SEQ ID NO: x), L3 is 0 amino acids in length, and L4 is 0 amino acids in length. In some embodiments, L1 comprises the sequence Gly-Ser-Gly (SEQ ID NO: x), L2 comprises the sequence Gly-Ser-Gly (SEQ ID NO: x), L3 is 0 amino acids in length, and L4 is 0 amino acids in length. In some embodiments, L1 comprises the sequence Gly-Ser-Gly-Ser (SEQ ID NO: x), L2 is 0 amino acids in length, L3 comprises the sequence Gly-Ser-Gly-Ser (SEQ ID NO: x), and L4 is 0 amino acids in length. In some embodiments, L1 and L2 are zero amino acids in length, and L3 and L4 each comprise a sequence independently selected from: (Gly-Gly-Gly-Gly-Ser) n(SEQ ID NO: x) (wherein n is an integer between 0 and 5; SEQ ID NO: x), Gly-Gly-Gly-Gly-Gly-Ser-Gly-Gly-Gly-Gly-Ser (SEQ ID NO: x), Gly-Gly-Gly-Gly-Ser-Gly-Gly-Gly-Gly-Ser (SEQ ID NO: x), Ser, Arg-Thr, Thr-Lys-Gly-Pro-Ser (SEQ ID NO: x), Gly-Gln-Pro-Lys-Ala-Ala-Pro (SEQ ID NO: x), and Gly-Gly-Ser-Gly-Ser-Ser-Gly-Ser-Gly-Gly (SEQ ID NO: x). In some embodiments, L3 and L4 are zero amino acids in length, and L1 and L2 each comprise a sequence independently selected from: (Gly-Gly-Gly-Gly-Ser)n(wherein n is an integer between 0 and 5; SEQ ID NO: x), Gly-Gly-Gly-Gly-Ser-Gly-Gly-Gly-Gly-Gly-Ser (SEQ ID NO: x), Gly-Gly-Gly-Gly-Ser-Gly-Gly-Gly-Gly-Gly-Ser (SEQ ID NO: x), Ser, Arg-Thr, Thr-Lys-Gly-Pro-Ser (SEQ ID NO: x), Gly-Gln-Pro-Lys-Ala-Pro (SEQ ID NO: x), and Gly-Gly-Ser-Gly-Ser-Ser-Gly-Ser-Gly-Ser-Gly-Gly (SEQ ID NO: x).

In some embodiments, one or more linkers comprise a sequence derived from a naturally occurring sequence located at the junction between an antibody variable domain and an antibody constant domain (e.g., as described in WO 2012/135345, which is incorporated by reference). For example, in some embodiments, the linker comprises a sequence found at the transition between an endogenous VH domain and a CH1 domain or between an endogenous VL domain and a CL domain (e.g., κ or λ). In some embodiments, the linker comprises a sequence found at the transition between an endogenous human VH domain and a CH1 domain or between an endogenous human VL domain and a CL domain (e.g., human κ or λ).

The examples listed above are not intended to limit the scope of the present disclosure in any way, and linkers comprising randomly selected amino acids selected from valine, leucine, isoleucine, serine, threonine, lysine, arginine, histidine, aspartic acid, glutamic acid, asparagine, glutamine, glycine, and proline are suitable for use in the binding proteins described herein. For additional description of linker sequences see, e.g., WO 2012/135345, WO 2017/180913, which are incorporated by reference.

As used herein, the term "valency" refers to the number of binding sites of a binding protein, epitope, antigen binding protein, or antibody. For example, the term "monovalent binding protein" refers to a binding protein having one antigen binding site. The term "bivalent binding protein" refers to a binding protein having two binding sites. The term "trivalent binding protein" refers to a binding protein having three binding sites. The term "tetravalent binding protein" refers to a binding protein having four binding sites. In particular embodiments, the bivalent binding protein may bind to one antigen target. In other embodiments, a bivalent binding protein may bind two different antigen targets. In particular embodiments, the trivalent binding protein may bind to one antigen target, i.e., is monospecific. In other embodiments, a trivalent binding protein may bind two different antigen targets, i.e., be bispecific. In other embodiments, a trivalent binding protein may bind three different antigen targets, i.e., is trispecific. In particular embodiments, the tetravalent binding protein may bind to one antigen target, i.e., be monospecific. In other embodiments, the tetravalent binding protein may bind to two different antigen targets, i.e. be bispecific. In other embodiments, the tetravalent binding protein may bind to three different antigen targets, i.e., is trispecific. In other embodiments, the tetravalent binding protein may bind to four different antigen targets, i.e., is tetraspecific.

As used herein, the term "specificity" refers to the amount of binding specificity of a binding protein, epitope, antigen binding protein, or antibody. For example, the term "monospecific binding protein" refers to a binding protein that specifically binds to one antigen target. The term "bispecific binding protein" refers to a binding protein that specifically binds to two different antigen targets. The term "trispecific binding protein" refers to a binding protein that specifically binds to three different antigen targets. The term "tetraspecific binding protein" refers to a binding protein that specifically binds to four different antigen targets, and so forth.

As used herein, the term "selective recognition site" refers to a modification in a binding protein that allows for selective recognition by an affinity reagent that binds to the selective recognition site. Examples of selective recognition sites include binding sites for protein a in the Fc portion of an immunoglobulin.

As used herein, the term "affinity reagent" refers to a reagent containing a ligand that is immobilized on a matrix and specifically binds to a surface group of a molecule, such as an amino acid or sugar side chain, and generally has a specific three-dimensional structural characteristic as well as a specific charge characteristic. Affinity reagents are tools in affinity chromatography where purification is enabled by specific interactions between ligands and products. "protein L" as an example of an affinity reagent refers to a recombinant protein L which is immobilized on a matrix to form a ligand having affinity for a subset of the variable domains of immunoglobulin kappa light chains. Such a matrix may be a resin. Another example of an affinity reagent is "kappa-select", which refers to a recombinant camelid derived 13kDa single chain antibody immobilized on a matrix to form a ligand with affinity for the constant domain of the kappa light chain of a human immunoglobulin. Another example of an affinity reagent is protein A. Protein A is a 42 kDa surface protein originally found on the cell wall of the bacterium Staphylococcus aureus (Staphylococcus aureus). It has been shown through crystallographic refinement that the major binding site for protein a is located on the Fc region, between the CH2 and CH3 domains. In addition, protein a has been shown to bind to human IgG molecules containing IgG F (ab') 2 fragments from the human VH3 gene family. Protein a binds with strong affinity to the Fc portion of immunoglobulins of certain species.

For example, the dissociation constant (KD) of a binding protein can be determined by surface plasmon resonance. In general, surface plasmon resonance analysis measures real-time binding interactions between a ligand (target antigen on a Biosensor matrix) and an analyte (binding protein in solution) by Surface Plasmon Resonance (SPR) using a BIAcore system (Pharmacia Biosensor; Piscataway, N.J.). Surface plasmon analysis can also be performed by immobilizing the analyte (binding protein on the biosensor matrix) and presenting the ligand (target antigen). As used herein, the term "KD" refers to the dissociation constant of the interaction between a particular binding protein and a target antigen.

The term "specifically binds" as used herein refers to a binding protein or antigen-binding fragment thereof that binds at least about 1x10-6M、1x10-7M、1x10-8M、1x10-9M、1x10-10M、1x10-11M、1x10-12M or greater Kd, and/or the ability to bind an epitope with an affinity that is at least two times greater than the affinity of the binding protein or antigen binding fragment thereof for a non-specific antigen. The binding affinity of an antigen to a binding protein or antibody can be performed by Surface Plasmon Resonance (SPR) using a BIAcore instrument.

The term "nucleic acid" as used herein refers to a polymeric or oligomeric macromolecule or a biological macromolecule that is essential for all known life forms. Nucleic acids, including DNA (deoxyribonucleic acid) and RNA (ribonucleic acid), are made from monomers called nucleotides. Most naturally occurring DNA molecules consist of two complementary biopolymer strands that are coiled around each other to form a double helix. A DNA strand is also referred to as a polynucleotide consisting of nucleotides. Each nucleotide is composed of a nitrogen-containing nucleobase, as well as monosaccharide and phosphate groups called deoxyribose or ribose. Naturally occurring nucleobases comprise guanine (G), adenine (A), thymine (T), uracil (U) or cytosine (C). Nucleotides are joined to each other in the chain by covalent bonds between the sugar of one nucleotide and the phosphate of the next, resulting in an alternating sugar-phosphate backbone. If fructose is deoxyribose, the polymer is DNA. If fructose is ribose, the polymer is RNA. Typically, polynucleotides are formed by phosphodiester bonds between individual nucleotide monomers.

The term "polynucleotide" as used herein refers to a single-or double-stranded nucleic acid polymer of at least 10 nucleotides in length. It is understood that a nucleotide comprising a polynucleotide may be a ribonucleotide or a deoxyribonucleotide or a modified form of either type of nucleotide. Such modifications include base modifications (e.g., bromouridine), ribose modifications (e.g., cytarabine and 2 ', 3' -dideoxyribose), and internucleotide linkage modifications (e.g., phosphorothioate, phosphorodithioate, phosphoroselenoate, phosphorodiselenoate, phosphoroanilothioate, phosphoroanillylate, and phosphoroamidate). The term "polynucleotide" specifically includes single-stranded and double-stranded forms of DNA.

An "isolated polynucleotide" is a polynucleotide of genomic, cDNA, or synthetic origin, or some combination thereof, which isolated polynucleotide: (1) not associated with all or a portion of the polynucleotide in which it is found in nature, (2) linked to a polynucleotide that it is not linked to in nature, or (3) not present as part of a larger sequence in nature.

An "isolated polypeptide" is a polypeptide which: (1) free of at least some other polypeptides with which it is typically found, (2) substantially free of other polypeptides from the same source (e.g., from the same species), (3) expressed by cells from a different species, (4) has been isolated from at least about 50% of the polynucleotides, lipids, carbohydrates or other materials with which the isolated polypeptide binds in nature, (5) does not bind (via covalent or non-covalent interactions) to the portion of the polypeptide with which the "isolated polypeptide" binds in nature, (6) is operably bound (via covalent or non-covalent interactions) to a polypeptide with which the isolated polypeptide does not bind in nature, or (7) does not occur in nature. Such isolated polypeptides may be encoded by genomic DNA, cDNA, mRNA, or other RNA of synthetic origin, or any combination thereof. In exemplary embodiments, the isolated polypeptide is substantially free of polypeptides or other contaminants found in its natural environment that would interfere with its use (therapeutic, diagnostic, prophylactic, research or other use).

As used herein, the term "expression vector" also referred to as an expression construct, generally refers to a plasmid or virus designed for protein expression in a cell. The term "vector" refers to a protein or polynucleotide or a mixture thereof that is capable of being introduced into a cell or of introducing the proteins and/or nucleic acids contained therein into a cell. Examples of vectors include, but are not limited to, plasmids, cosmids, phages, viruses or artificial chromosomes. In particular, a vector is used to transport a gene product of interest (such as, for example, foreign or heterologous DNA) into a suitable host cell. The vector may contain a "replicon" polynucleotide sequence which facilitates autonomous replication of the vector in a host cell. Foreign DNA is defined as heterologous DNA that is DNA not naturally found in the host cell, e.g., a replicating vector molecule, encoding a selectable or screenable marker, or encoding a transgene. Once in the host cell, the vector may replicate independently of the host chromosomal DNA or simultaneously with the host chromosomal DNA, and several copies of the vector and its inserted DNA may be produced. In addition, the vector may also contain the necessary elements to permit transcription of the inserted DNA into an mRNA molecule or otherwise cause replication of the inserted DNA into multiple copies of RNA. The vector may further encompass "expression control sequences" that regulate the expression of the gene of interest. Typically, the expression control sequence is a polypeptide or polynucleotide, such as, but not limited to, a promoter, enhancer, silencer, insulator, or repressor. In a vector comprising more than one polynucleotide encoding one or more gene products of interest, expression can be controlled together or separately by one or more expression control sequences. More specifically, each polynucleotide contained on the vector may be controlled by a separate expression control sequence, or all polynucleotides contained on the vector may be controlled by a single expression control sequence. An open reading frame may be formed by a polynucleotide contained on a single vector controlled by a single expression control sequence. Some expression vectors additionally contain sequence elements adjacent to the inserted DNA that increase the half-life of the expressed mRNA and/or allow translation of the mRNA into a protein molecule. Many mRNA and polypeptide molecules encoded by the inserted DNA can thus be synthesized rapidly.

The term "host cell" as used herein refers to a cell into which a recombinant expression vector has been introduced. Recombinant host cells or host cells are intended to refer not only to the particular subject cell, but also to the progeny of such a cell. Because certain modifications may occur in succeeding generations due to either mutation or environmental influences, such progeny may not, in fact, be identical to the parent cell, but such cells are still included within the scope of the term "host cell" as used herein. A wide variety of host cell expression systems can be used to express the binding protein, including bacterial, yeast, baculovirus and mammalian expression systems (as well as phage display expression systems). An example of a suitable bacterial expression vector is pUC 19. For recombinant expression of a binding protein, a host cell is transformed or transfected with one or more recombinant expression vectors carrying DNA segments encoding polypeptide chains of the binding protein such that the polypeptide chains are expressed in the host cell and, in exemplary embodiments, secreted into a culture medium in which the host cell is cultured, whereby the binding protein can be recovered from the culture medium.

As used herein, the term "pharmaceutical composition" refers to a compound or composition that is capable of inducing a desired therapeutic effect when properly administered to a subject (e.g., a human subject).

As used herein, the term "pharmaceutically acceptable carrier" or "physiologically acceptable carrier" refers to one or more formulation materials suitable for achieving or enhancing delivery of a binding protein.

The terms "effective amount" and "therapeutically effective amount" when used in reference to a pharmaceutical composition comprising one or more binding proteins (e.g., antibodies or antigen-binding fragments thereof) refer to an amount or dose sufficient to produce a desired therapeutic result. More specifically, a therapeutically effective amount is an amount of a binding protein (e.g., an antibody or antigen-binding fragment thereof) sufficient to inhibit one or more clinically-defined pathological processes associated with the condition being treated for a period of time. The effective amount may vary depending on the particular antibody-like binding protein used and also depends on a variety of factors and conditions related to the patient being treated and the severity of the disorder. For example, if the binding protein or multispecific binding protein is to be administered in vivo, factors such as the age, weight, and health of the patient, as well as dose response curves and toxicity data obtained in preclinical animal work, will be among those considered. Determination of an effective or therapeutically effective amount of a given pharmaceutical composition is well within the ability of those skilled in the art.

As used herein, the term "method of producing a binding protein" refers to recombinant methods of protein expression using techniques well known in the art.

II.Pair-promoting CH1/CL mutations

In certain embodiments, mutations can be made in the CH1 and CL interface to promote specific pairing and prevent CH1/CL mis-pairing. Such mutations are described below, and are further described in detail in: WO 2013/005194 and Golay et al (2016) J.Immunol. 196. 3199-3211, each of which is incorporated herein by reference.

A first set of mutations can be made to the interacting polar interface amino acid pairs in the CH1 and CL domains. The polar amino acids can be exchanged for neutral and salt-forming amino acid pairs. In a certain embodiment, the first set of mutations may comprise the T192E CH1 mutation and the N137K, S114A clk mutations. The T192E CH1 mutation and the N137K clk mutation form salt bridges that can potentiate association specificity, however, undesirable pairing should be avoided by the lack of space and charge complementarity between the wild type and variant CH1 and clk domains. In addition, the S114A mutation on the clk domain was made to avoid steric clashes with the larger lysine side chain. Alternatively, the CH 1T 192E and CL N137K/S114A mutant groups may be referred to as "CR 3" mutant groups.

A second set of mutations can be made to the interacting hydrophobic and polar interfacial amino acid residue pairs in the CH1 and clk domains. One mutation may constitute a switch from hydrophobic interaction to polar interaction. In a certain embodiment, the second set of mutations may comprise L143Q, S188V CH1 mutations, and V133T, S176V clk mutations. The L134Q CH1 mutation and the V133T clk mutation constitute switches from hydrophobic to polar interactions. The simultaneous presence of the S188V CH1 mutation and the S176V clk mutation constitutes a switch from polar interaction to hydrophobic interaction. The exchange of the polar/hydrophobic character of the interfacial interactions is expected to keep the affinity between the mutated CH1 and clk domains unchanged while reducing their respective affinities for the other wild-type counterparts CH1 and clk domains, thereby preventing mis-pairing by virtue of the unfavorable interactions that occur on the mismatched (variant/wild-type) domains. Alternatively, the CH 1L 143Q/S188V and CL V133T/S176V mutant groups may be referred to as "MUT 4" mutant groups.

The third and fourth sets of mutations were "knob and hole" mutations. More specifically, in the third group of mutations (KH1), L124A, L143E CH1 mutations were performed and V133W clk mutations were performed. In the fourth group of mutations (KH2), the V190A CH1 mutation was performed and the L135W, N137A clk mutations were performed. The first, second, third and fourth sets of mutations are described in further detail in WO 2013/005194 a 1.

The fifth and sixth sets of mutations can be made to exchange electrostatic charges in the CH1 and clk domains. In a certain embodiment, the fifth set of mutations may comprise a K221E CH1 mutation and an E123K clk mutation. Alternatively, the fifth set of mutations may be referred to as the "K221E/E123K opposite charge" mutation set or the "NN 1" mutation set. In a certain embodiment, the sixth set of mutations may comprise the K228D CH1 mutation and the D122K clk mutation. Alternatively, the sixth set of mutations may be referred to as the "K228D/D122K opposite charge" mutation set or the "NN 2" mutation set. Group five and sixth mutations are described in further detail in WO 2007/147901 a 1. In certain embodiments, the "K221E/E123K oppositely charged" mutation group and the "K228D/D122K oppositely charged" mutation group may be combined. Thus, the combination may comprise the K221E and K228D CH1 mutant pair and the E123K and D122K clk mutant pair. Alternatively, the combined mutant group may be referred to as the "K221E: K228D/E123K: D122K oppositely charged" mutant group or "NN 3" mutant group.

The seventh and eighth sets of mutations can be made to exchange electrostatic charges in the CH1 domain and the clk domain. In a certain embodiment, the seventh set of mutations may comprise L143E, L143D, L143R, L143K, or L143H CH1 mutations and S176E, S176D, S176R, S176K, or S176H clk mutations, provided that the CH1 mutation is oppositely charged to the clk mutation. Alternatively, the seventh set of mutations may be referred to as the "L143/S176 oppositely charged" set of mutations. In certain embodiments, the CH 1L 143E or L143D mutations may be paired with CL S176R or S176K mutations and may be referred to as a "CM 3" mutation group. In certain embodiments, the CH 1L 143R or L143K mutations may be paired with CL S176E or S176D mutations and may be referred to as a "CM 4" mutation group. In a certain embodiment, the eighth set of mutations can comprise L124E, L124D, L124R, L124K, or L124H CH1 mutations and V133E, V133D, V133R, V133K, or V133H clk mutations, provided that the CH1 mutations are oppositely charged to the clk mutations. Alternatively, the eighth set of mutations can be referred to as the "L124/V133 oppositely charged" set of mutations. In certain embodiments, the CH 1L 124E or L124D mutations may be paired with CL V133R or V133K mutations and may be referred to as the "CM 5" mutation group. In certain embodiments, the CH 1L 124R or L124K mutations may be paired with CL V133E or V133D mutations and may be referred to as the "CM 6" mutation group.

In further embodiments, any one or more of the above-mentioned mutations may be combined with each other and/or with the mutations described below. As an example, the CH1 domain of the first Fab contains the T192E, K221E mutations and the clk domain of the first Fab contains the E123K, N137K, S114A mutations. The CH1 domain of the second Fab may further comprise L143Q, S188V mutations, and the clk domain of the second Fab may comprise V133T, S176V mutations. Alternatively, the second Fab may be wild-type.

Sequence position numbering as used herein with respect to the CH1 domain and the CL κ domain refers to Kabat numbering (Kabat, E.A. et al, Sequences of proteins of immunological interest. 5 th edition-US Department of Health and Human Services, NIH publication No. 91-3242, pages 662, 680, 689, 1991).

III.VH/VL opposite charge mutations that promote pairing

In certain embodiments, mutations may be made in the VH and VL interfaces to promote specific pairing and prevent VH/VL mis-pairing. In particular embodiments, mutations made to the VH and VL domains introduce oppositely charged amino acid residues to promote heterodimerization through electrostatic interactions.

In a certain embodiment, the set of opposite charge mutations may comprise a mutation at Kabat position 39 in the VH domain and a mutation at Kabat position 38 in the VL domain. Any known positively or negatively charged residue may be introduced at VH Kabat position 39, provided that the mutation introduced at VL Kabat position 38 is oppositely charged. For example, one possible mutation pair comprises a VH K39 mutant residue (introducing a positive charge) and a VL E38 mutant residue (introducing a negative charge). Alternatively, a set of back mutations may be performed in which a VH E39 mutant residue (introducing a negative charge) and a VL K38 mutant residue (introducing a positive charge) are introduced. In certain embodiments, the VH domain may comprise a mutation at Kabat position 39 to a residue of E, D, K, R or H, and the VL domain may comprise a mutation at Kabat position 38 to a residue of E, D, K, R or H, provided that the mutated residue in the VH domain is oppositely charged to the mutated residue in the VL domain. For example, when the VH domain comprises a mutated residue E or D at Kabat position 39, then the VL domain comprises a mutated residue K, R or H at Kabat position 38. Alternatively, when the VH domain comprises a mutated residue K, R or H at Kabat position 39, then the VL domain comprises a mutated residue E or D at Kabat position 38. In a certain embodiment wherein two VH/VL pairs are not in the same polypeptide chain, if one VH/VL pair comprises a positive charge (e.g., E or D) in VH39 and a negative charge (e.g., K, R or H) in VL38, the other VH/VL pair can comprise a negative charge (e.g., K, R or H) in VH39 and a positive charge (e.g., E or D) in VL 38. For example, if a first VH/VL pair comprises VH39K and VL38E mutations, a second VH/VL pair may comprise VH39E and VL38K mutations. Alternatively, this set of opposite charge mutations may be referred to as the "VH 39/VL38 opposite charge" mutation set. This group of oppositely charged mutations is further described in detail in the following documents: tan et al, Biophysic journal, Vol.75, p.1473-1482, 1998.

In a certain embodiment, the set of opposite charge mutations may comprise a Q39 mutation in the VH domain and a Q38 mutation in the VL domain. Any known positively or negatively charged residue may be introduced at the VH Q39 position, provided that the mutation introduced at the VL Q38 position is oppositely charged. For example, one possible mutation pair comprises a VH Q39K mutation (introducing a positive charge) and a VL Q38E mutation (introducing a negative charge). Alternatively, a set of back mutations may be performed in which the VH Q39E mutation (negative charge introduced) and the VL Q38K mutation (positive charge introduced) are introduced. In certain embodiments, the VH domain may comprise a Q39E, Q39D, Q39K, Q39R, or Q39H mutation, and the VL domain may comprise a Q38E, Q38D, Q38K, Q38R, or Q38H mutation, provided that the mutation in the VH domain is oppositely charged to the mutation in the VL domain.

In further embodiments, any one or more of the above-mentioned mutations may be combined with each other and/or with the mutations described below.

IV.VH/VL disulfide stabilization mutations

In certain embodiments, mutations may be made in the VH and VL interfaces to improve stability between the VH/VL interfaces. Specific sets of amino acid mutations in the VH and VL domains can improve stability by introducing non-native cysteine residues that form disulfide bridges.

A first set of disulfide stabilizing mutations may be made to amino acid residues in the VH and VL domains. In a certain embodiment, the first set of disulfide stabilizing mutations may comprise a 44C mutation in the VH domain and a 100C mutation in the VL domain. Alternatively, the first set of disulfide stabilizing mutations may be referred to as the "VH 44C/VL 100C" mutation set. The first group of disulfide stabilizing mutations is further described in detail in: reiter et al Nature Biotechnology, Vol.14, pp.1239-1245, 1996, which are incorporated herein by reference for all purposes.

A second set of disulfide stabilizing mutations may be made to amino acid residues in the VH and VL domains. In a certain embodiment, the second set of disulfide stabilizing mutations may comprise a 105C mutation in the VH domain and a 43C mutation in the VL domain. Alternatively, the second set of disulfide stabilizing mutations may be referred to as the "VH 105C/VL 43C" mutation set. A second set of disulfide stabilizing mutations is further described in detail in U.S. patent No. 9,527,927, which is incorporated herein by reference for all purposes.

V.Antibody mutation combinations

In certain embodiments, combinations of sets of mutations can be located in the CH1 and clk interface and/or in the VH and VL interface to further facilitate pairing and improve stability.

In particular embodiments, the first Fab domain in the antibody may comprise a combination of one or both of the CR3 and MUT4 mutation sets with opposite charge mutation sets to the VH/VL. In further embodiments, the second Fab domain in the antibody may comprise a combination of one or both of the CR3 and MUT4 mutation sets with opposite charge to the VH/VL mutation set.

In particular embodiments, the first Fab domain may comprise a combination of one or both of the CR3 and MUT4 mutation sets with opposite charge to the VH/VL mutation sets and disulfide bond stabilizing mutation sets. In further embodiments, the second Fab domain in the antibody may comprise a combination of one or both of the CR3 and MUT4 mutant sets of opposite charge to VH/VL and disulfide bond stabilizing mutant sets.

Any of the above embodiments may further comprise sets of opposite charge mutations in the CH1/CL interface. For example, the first Fab may comprise the K221E CH1 mutation and the E123K clk mutation. The second Fab may comprise the K221E CH1 mutation and the E123K clk mutation.

VI.Antibody formats and sets of mutations therein

Any of the above-listed sets of mutations and combinations thereof may be applied to the multispecific antigen-binding proteins described herein.

Cross double variable

In particular embodiments, "cross-double variable" or "CODV" refers to an antigen binding domain that specifically binds to at least one target antigen or at least one target epitope, and comprises at least two polypeptide chains forming at least two antigen binding sites, wherein at least one polypeptide chain comprises a structure represented by the formula:

VL1-L1-VL2-L2-CL[I]

and at least one polypeptide chain comprises a structure represented by the formula:

VH2-L3-VH1-L4-CH1[II]

wherein:

VL1 is a first immunoglobulin light chain variable domain;

VL2 is a second immunoglobulin light chain variable domain;

VH1 is a first immunoglobulin heavy chain variable domain;

VH2 is a second immunoglobulin heavy chain variable domain;

CL is an immunoglobulin light chain constant domain;

CH1 is an immunoglobulin CH1 heavy chain constant domain; and is

L1, L2, L3 and L4 are amino acid linkers, and

wherein the polypeptide of formula I and the polypeptide of formula II form a cross light-heavy chain pair.

In particular embodiments, the CODV antigen binding domain specifically binds to at least one target antigen or at least one target epitope and comprises four polypeptide chains forming four antigen binding sites, wherein each of the two polypeptide chains comprises a structure represented by the formula:

VL1-L1-VL2-L2-CL[I]

And each of the two polypeptide chains comprises a structure represented by the formula:

VH2-L3-VH1-L4-CH1-Fc[II]

wherein:

VL1 is a first immunoglobulin light chain variable domain;

VL2 is a second immunoglobulin light chain variable domain;

VH1 is a first immunoglobulin heavy chain variable domain;

VH2 is a second immunoglobulin heavy chain variable domain;

CL is an immunoglobulin light chain constant domain;

CH1 is an immunoglobulin CH1 heavy chain constant domain;

fc is an immunoglobulin hinge region and CH2 and CH3 immunoglobulin heavy chain constant domains; and is

L1, L2, L3 and L4 are amino acid linkers, and

wherein said polypeptide of formula I and said polypeptide of formula II form a cross light-heavy chain pair,

wherein the VH1/VL1 pair comprises a first antigen binding specificity and the VH2/VL2 pair comprises a second antigen binding specificity.

In certain embodiments, the antigen binding proteins described herein are trispecific and/or trivalent antigen binding proteins comprising four polypeptide chains forming three antigen binding sites that specifically bind to one or more different antigen targets, wherein a first polypeptide chain comprises a structure represented by the formula:

VL2-L1-VL1-L2-CL[I]

the second polypeptide chain comprises a structure represented by the formula:

VH1-L3-VH2-L4-CH 1-hinge-CH 2-CH3[ II ]

The third polypeptide chain comprises a structure represented by the formula:

VH3-CH 1-hinge-CH 2-CH3[ III ]

And the fourth polypeptide chain comprises a structure represented by the formula:

VL3-CL[IV],

wherein:

VL1 is a first immunoglobulin light chain variable domain;

VL2 is a second immunoglobulin light chain variable domain;

VL3 is a third immunoglobulin light chain variable domain;

VH1 is a first immunoglobulin heavy chain variable domain;

VH2 is a second immunoglobulin heavy chain variable domain;

VH3 is a third immunoglobulin heavy chain variable domain;

CL is an immunoglobulin light chain constant domain;

CH1 is an immunoglobulin CH1 heavy chain constant domain;

CH2 is an immunoglobulin CH2 heavy chain constant domain;

CH3 is an immunoglobulin CH3 heavy chain constant domain;

a hinge is an immunoglobulin hinge region connecting the CH1 domain and the CH2 domain; and is

L1, L2, L3 and L4 are amino acid linkers, and

wherein the polypeptide of formula I and the polypeptide of formula II form a cross light-heavy chain pair.

In certain embodiments, the first polypeptide chain and the second polypeptide chain have a cross-orientation that forms two different antigen binding sites. In some embodiments, VH1 and VL1 form a binding pair and form a first antigen binding site. In some embodiments, VH2 and VL2 form a binding pair and form a second antigen binding site. In some embodiments, the third polypeptide and the fourth polypeptide form a third antigen binding site. In some embodiments, VH3 and VL3 form a binding pair and form a third antigen binding site.

Such antigen binding proteins comprise at least three antigen binding sites. The antigen binding protein is at least a trivalent antigen binding molecule. In particular embodiments, the antigen binding protein specifically binds to one antigen target, i.e., the antigen binding protein is a monospecific antigen binding molecule. In another embodiment, the antigen binding protein specifically binds to two different antigen targets, i.e., the antigen binding protein is a bispecific antigen binding molecule. In another embodiment, the antigen binding protein specifically binds to three different antigen targets, i.e., the antigen binding protein is a trispecific antigen binding molecule.

The examples listed above are not intended to limit the scope of the invention in any way, and it has been shown that linkers comprising randomly selected amino acids selected from the group consisting of: valine, leucine, isoleucine, serine, threonine, lysine, arginine, histidine, aspartic acid, glutamic acid, asparagine, glutamine, glycine and proline.

CODV antibody formats, various permutations of CODV antibody formats, and other details regarding linkers are further described in WO 2012/135345a1 and WO 2017/180913a2, which are incorporated herein by reference in their entirety.

Tandem Fab

In certain embodiments, a "tandem Fab" refers to an antigen binding protein in which the C-terminus of one CH1 region of a first Fab domain is operably linked to the N-terminus of a VH region of a second Fab domain. In certain embodiments, the tandem Fab antibodies may be tetravalent and monospecific (each of the four fabs binds the same antigen). In certain embodiments, the tandem Fab antibodies may be tetravalent and bispecific (two of the four fabs bind a first antigen or epitope and the other two fabs bind a second antigen or epitope).

The tandem fab may be operably linked using any peptide linker known in the art for linking two or more antigen binding domains. In a particular embodiment, the peptide linker is a Gly-to-Ser linker, i.e. a linker comprising only one or more glycine amino acids and one or more serine amino acids. In a particular embodiment, the peptide linker is (Gly-Gly-Gly-Gly-Ser)n(SEQ ID NO: x) linker, wherein n is any integer from 1 to 5. In a particular embodiment, the peptide linker is (Gly-Gly-Gly-Gly-Ser)3(SEQ ID NO: x) linker. In a particular embodiment, the peptide linker is (Gly-Gly-Gly-Gly-Ser)2(SEQ ID NO: x) linker.

Alternatively, or in combination with the Gly-to-Ser linkers listed above, the peptide linker may comprise all or part of the sequence of the hinge region of one or more immunoglobulins selected from IgA, IgG and IgD. The sequences of the hinge regions of human IgG, IgA and IgD are indicated below:

IgA1(SEQ ID NO:X):

Val-Pro-Ser-Thr-Pro-Pro-Thr-Pro-Ser-Pro-Ser-Thr-Pro-Pro-Thr-Pro-Ser-Pro-Ser。

IgA2(SEQ ID NO:X):

Val-Pro-Pro-Pro-Pro-Pro。

IgD(SEQ ID NO:X):Glu-Ser-Pro-Lys-Ala-Gln-Ala-Ser-Ser-Val-Pro-Thr-Ala-Gln-Pro-Gln-Ala-Glu-Gly-Ser-Leu-Ala-Lys-Ala-Thr-Thr-Ala-Pro-Ala-Thr-Thr-Arg-Asn-Thr-Gly-Arg-Gly-Gly-Glu-Glu-Lys-Lys-Lys-Glu-Lys-Glu-Lys-Glu-Glu-Gln-Glu-Glu-Arg-Glu-Thr-Lys-Thr-Pro。

IgG1(SEQ ID NO:X):

Glu-Pro-Lys-Ser-Cys-Asp-Lys-Thr-His-Thr-Cys-Pro-Pro-Cys-Pro。

IgG2(SEQ ID NO:X):

Glu-Arg-Lys-Cys-Cys-Val-Glu-Cys-Pro-Pro-Cys-Pro。

IgG3:

Glu-Leu-Lys-Thr-Pro-Leu-Gly-Asp-Thr-Thr-His-Thr-Cys-Pro-Arg-Cys-Pro (SEQ ID NO: X), followed by 0 or 1 to 4 repeats of Glu-Pro-Lys-Ser-Cys-Asp-Thr-Pro-Pro-Cys-Pro-Arg-Cys-Pro (SEQ ID NO: X).

IgG4:

Glu-Ser-Lys-Tyr-Gly-Pro-Pro-Cys-Pro-Ser-Cys-Pro(SEQ ID NO:X)。

The peptide linker may comprise all or part of the sequence of the hinge region of only one immunoglobulin. In this case, the immunoglobulin may be of the same isotype and subclass as the immunoglobulin from which the adjacent CH1 domain is derived, or of a different isotype or subclass.

Alternatively, the peptide linker may comprise all or part of the sequences of the hinge regions of at least two immunoglobulins of different isotypes or subclasses. In this case, the N-terminal part of the peptide linker directly following the CH1 domain may consist of all or part of the hinge region of an immunoglobulin of the same isotype and subclass as the immunoglobulin from which the CH1 domain is derived. Optionally, the peptide linker may further comprise a sequence of 2 to 15 or 5 to 10N-terminal amino acids of the CH2 domain of an immunoglobulin.

In certain embodiments, sequences from a native hinge region may be used. In other embodiments, point mutations may be introduced into these sequences, in particular alanine or serine in place of one or more cysteine residues in the native IgG1, IgG2, or IgG3 hinge sequences, in order to avoid undesirable intra-or interchain disulfide bonds.

Non-limiting examples of peptide linkers that can be used in the antigen binding proteins of the present disclosure are peptide linkers having the following sequences: Glu-Pro-Lys-Ser-Cys-Asp-Lys-Thr-His-Thr-Cys-Pro-Ala-Pro-Glu-Leu-Leu-Gly-Gly-Pro-Ser-Thr-Pro-Pro-Thr-Pro-Ser-Pro-Ser-Gly-Gly-Gly (SEQ ID NO: x). The peptide linker consists of: the full length sequence of the human IgG1 hinge, followed by the 9N-terminal amino acids of human IgG1 CH2 (Ala-Pro-Glu-Leu-Leu-Gly-Gly-Pro-Ser, SEQ ID NO: X), followed by a portion of the sequence of the human IgA1 hinge (Thr-Pro-Pro-Thr-Pro-Ser-Pro-Ser, SEQ ID NO: X), and followed by dipeptide GG added to provide supplemental flexibility to the linker. In certain embodiments, the peptide linker Glu-Pro-Lys-Ser-Cys-Asp-Lys-Thr-His-Thr-Cys-Pro-Ala-Pro-Glu-Leu-Leu-Gly-Gly-Pro-Ser-Thr-Pro-Pro-Thr-Pro-Ser-Pro-Ser-Gly-Gly (SEQ ID NO: x) may have one or more cysteine residues substituted to eliminate disulfide bond formation. In particular embodiments, the peptide linker comprises the following sequence: Glu-Pro-Lys-Ser-Cys-Asp-Lys-Thr-His-Thr-Ser-Pro-Pro-Ser-Pro-Ala-Pro-Glu-Leu-Leu-Gly-Gly-Pro-Ser-Thr-Pro-Pro-Thr-Pro-Ser-Pro-Ser-Gly-Gly (SEQ ID NO: X). Hinge-derived peptide linkers are further described in WO 2013/005194 a2, which is incorporated by reference herein in its entirety.

VII.Exemplary antibody mutation and antibody Format combinations

Any of the antibody mutations listed above may be combined with any of the antibody formats listed above.

In particular embodiments, the antigen binding proteins of the present disclosure may comprise CODV antibody forms having one or more of a CR3 mutant panel, a MUT4 mutant panel, an L143/S176 oppositely charged mutant panel, and an L124/V133 oppositely charged mutant panel. The antigen binding protein may further comprise one or more VH/VL oppositely charged mutant groups. The one or more VH/VL opposing charge mutation sets include, but are not limited to, the VH39/VL38 opposing charge mutation set. The antigen binding protein may further comprise one or more VH/VL disulfide stabilization mutants. The one or more VH/VL disulfide bond mutation sets include, but are not limited to, the VH44C/VL100C mutation set and the VH105C/VL43C mutation set. The antigen binding protein may further comprise one or more CH1/CL oppositely charged mutant groups. The one or more CH1/CL oppositely charged mutant groups may include, but are not limited to, the K221E/E123K oppositely charged mutant group.

In certain embodiments, the antigen binding proteins of the present disclosure comprise two polypeptide chains and form two antigen binding sites, wherein one polypeptide chain has a structure represented by the formula:

VL1-L1-VL2-L2-CL[I]

And one polypeptide chain has a structure represented by the formula:

VH2-L3-VH1-L4-CH1[II]

wherein:

VL1 is a first immunoglobulin light chain variable domain;

VL2 is a second immunoglobulin light chain variable domain;

VH1 is a first immunoglobulin heavy chain variable domain;

VH2 is a second immunoglobulin heavy chain variable domain;

CL is an immunoglobulin light chain constant domain;

CH1 is an immunoglobulin CH1 heavy chain constant domain; and is

L1, L2, L3 and L4 are amino acid linkers,

wherein said polypeptide of formula I and said polypeptide of formula II form a cross light-heavy chain pair,

wherein one or both of VH1 and VH2 comprises a VH44C mutation and one or both of VL1 and VL2 comprises a VL100C mutation to form a disulfide bond,

wherein one or both of VH1 and VH2 comprises a Q39E, Q39D, Q39K, Q39R, or Q39H mutation, and one or both of VL1 and VL2 comprises a Q38E, Q38D, Q38K, Q38R, or Q38H mutation, wherein the mutation in one or both of VH1 and VH2 is oppositely charged to the mutation in one or both of VL1 and VL2, and

wherein the CH1 domain comprises one or more of the T192E, L143Q, and S188V mutations, and the CL domain comprises one or more of the N137K, S114A, V133T, and S176V mutations.

In particular embodiments, the antigen binding proteins of the present disclosure may comprise a tandem Fab antibody format having one or more of a CR3 mutant panel, a MUT4 mutant panel, an L143/S176 oppositely charged mutant panel, and an L124/V133 oppositely charged mutant panel. The antigen binding protein may further comprise one or more VH/VL oppositely charged mutant groups. The one or more VH/VL opposing charge mutation sets include, but are not limited to, the VH39/VL38 opposing charge mutation set. The antigen binding protein may further comprise one or more VH/VL disulfide stabilization mutants. The one or more VH/VL disulfide bond mutation sets include, but are not limited to, the VH44C/VL100C mutation set and the VH105C/VL43C mutation set. The antigen binding protein may further comprise one or more CH1/CL oppositely charged mutant groups. The one or more CH1/CL oppositely charged mutant groups may include, but are not limited to, the K221E/E123K oppositely charged mutant group.

In particular embodiments, the multispecific antibodies of the present disclosure may comprise:

a) a first light chain (LC 1)/heavy chain (HC1) pair, the first light chain (LC 1)/heavy chain (HC1) pair comprising:

(1) a first VL region (VL1) that pairs with a first VH region (VH1) to form a first antigen binding site (VL 1);

(2) A first constant heavy chain region 1(CH1-1) operably linked to VH1 and a first constant light chain region (CL1) operably linked to VL 1; and

b) a second light chain (LC 2)/heavy chain (HC2) pair, the second light chain (LC 2)/heavy chain (HC2) pair comprising:

(3) a second VL region (VL2) that pairs with a second VH region (VH2) to form a second antigen binding site; and

(4) a second constant heavy chain region 1(CH1-2) operably linked to VH2 and a second constant light chain region (CL2) operably linked to VL2,

wherein the C-terminus of CH1-1 is operably linked to the N-terminus of VH2, and

wherein one or both of VH1 and VH2 comprises a Q39E, Q39D, Q39K, Q39R, or Q39H mutation, and one or both of VL1 and VL2 comprises a Q38E, Q38D, Q38K, Q38R, or Q38H mutation, wherein the mutation in one or both of VH1 and VH2 is oppositely charged to the mutation in one or both of VL1 and VL2, and

wherein one or both of the CH1-1 domain and the CH1-2 domain comprise one or more of the T192E, L143Q, and S188V mutations, and one or both of the CL1 domain and the CL2 domain comprise one or more of the N137K, S114A, V133T, and S176V mutations,

Wherein the mutations that promote pairing in CH1-1 and CL1 are different from the mutations that promote pairing in CH1-2 and CL 2.

In particular embodiments, the antigen binding proteins of the present disclosure may comprise a traditional Y-shaped antibody format having one or more of a CR3 mutant panel, a MUT4 mutant panel, a L143/S176 oppositely charged mutant panel, and a L124/V133 oppositely charged mutant panel. The antigen binding protein may further comprise one or more VH/VL oppositely charged mutant groups. The one or more VH/VL opposing charge mutation sets include, but are not limited to, the VH39/VL38 opposing charge mutation set. The antigen binding protein may further comprise one or more VH/VL disulfide stabilization mutants. The one or more VH/VL disulfide stabilization mutant groups include, but are not limited to, the VH44C/VL100C mutant group and the VH105C/VL43C mutant group. The antigen binding protein may further comprise one or more CH1/CL oppositely charged mutant groups. The one or more CH1/CL oppositely charged mutant groups may include, but are not limited to, the K221E/E123K oppositely charged mutant group.

In particular embodiments, the multispecific antibodies of the present disclosure may comprise:

a) a first light chain (LC 1)/heavy chain (HC1) pair, said first light chain (LC 1)/heavy chain (HC1) pair comprising

(1) A first VL region (VL1) that pairs with a first VH region (VH1) to form a first antigen binding site (VL 1);

(2) a first constant heavy chain region 1(CH1-1) operably linked to VH1 and a first constant light chain region (CL1) operably linked to VL 1; and

(3) a first heterodimerization domain (HD 1); and

b) a second light chain (LC 2)/heavy chain (HC2) pair comprising

(4) A second VL region (VL2) that pairs with a second VH region (VH2) to form a second antigen binding site;

(5) a second constant heavy chain region 1(CH1-2) operably linked to VH2 and a second constant light chain region (CL2) operably linked to VL 2; and

(6) a second heterodimerization domain (HD 2);

wherein HD1 and HD2 heterodimerize,

wherein one or both of VH1 and VH2 comprises a Q39E, Q39D, Q39K, Q39R, or Q39H mutation, and one or both of VL1 and VL2 comprises a Q38E, Q38D, Q38K, Q38R, or Q38H mutation, wherein the mutation in one or both of VH1 and VH2 is oppositely charged to the mutation in one or both of VL1 and VL2,

wherein one or both of the CH1-1 domain and the CH1-2 domain comprise one or more of the T192E, L143Q, and S188V mutations, and one or both of the CL1 domain and the CL2 domain comprise one or more of the N137K, S114A, V133T, and S176V mutations, and

Wherein the mutations that promote pairing in CH1-1 and CL1 are different from the mutations that promote pairing in CH1-2 and CL 2.

In particular embodiments, the multispecific antibodies of the present disclosure may comprise:

a) a first light chain (LC 1)/heavy chain (HC1) pair, said first light chain (LC 1)/heavy chain (HC1) pair comprising

(1) A first VL region (VL1) that pairs with a first VH region (VH1) to form a first antigen binding site (VL 1);

(2) a first constant heavy chain region 1(CH1-1) operably linked to VH1 and a first constant light chain region (CL1) operably linked to VL 1; and

(3) a first heterodimerization domain (HD 1); and

b) a second light chain (LC 2)/heavy chain (HC2) pair comprising

(4) A second VL region (VL2) that pairs with a second VH region (VH2) to form a second antigen binding site;

(5) a second constant heavy chain region 1(CH1-2) operably linked to VH2 and a second constant light chain region (CL2) operably linked to VL 2; and

(6) a second heterodimerization domain (HD 2);

wherein HD1 and HD2 heterodimerize,

wherein one or both of VH1 and VH2 comprises a Q39E, Q39D, Q39K, Q39R, or Q39H mutation, and one or both of VL1 and VL2 comprises a Q38E, Q38D, Q38K, Q38R, or Q38H mutation, wherein the mutation in one or both of VH1 and VH2 is oppositely charged to the mutation in one or both of VL1 and VL2,

Wherein one or both of the CH1-1 domain and the CH1-2 domain comprise one or more of the T192E, L143Q, and S188V mutations, and one or both of the CL1 domain and the CL2 domain comprise one or more of the N137K, S114A, V133T, and S176V mutations, and

wherein the mutations that promote pairing in CH1-1 and CL1 are different from the mutations that promote pairing in CH1-2 and CL2,

wherein one or both of the CH1-1 domain and the CH1-2 domain further comprises a K221E mutation, and one or both of the CL1 domain and the CL2 domain further comprises an E123K mutation.

In particular embodiments, the multispecific antibodies of the present disclosure may comprise:

a) a first light chain (LC 1)/heavy chain (HC1) pair, said first light chain (LC 1)/heavy chain (HC1) pair comprising

(1) A first VL region (VL1) that pairs with a first VH region (VH1) to form a first antigen binding site (VL 1);

(2) a first constant heavy chain region 1(CH1-1) operably linked to VH1 and a first constant light chain region (CL1) operably linked to VL 1; and

(3) a first heterodimerization domain (HD 1); and

b) a second light chain (LC 2)/heavy chain (HC2) pair comprising

(4) A second VL region (VL2) that pairs with a second VH region (VH2) to form a second antigen binding site;

(5) a second constant heavy chain region 1(CH1-2) operably linked to VH2 and a second constant light chain region (CL2) operably linked to VL 2; and

(6) a second heterodimerization domain (HD 2);

wherein HD1 and HD2 heterodimerize,

wherein one or both of VH1 and VH2 comprises a Q39E, Q39D, Q39K, Q39R, or Q39H mutation, and one or both of VL1 and VL2 comprises a Q38E, Q38D, Q38K, Q38R, or Q38H mutation, wherein the mutation in one or both of VH1 and VH2 is oppositely charged to the mutation in one or both of VL1 and VL2,

wherein one or both of the CH1-1 domain and CH1-2 domain comprises an L143E, L143D, L143K, L143R, or L143H mutation, and one or both of the CL1 domain and CL2 domain comprises an S176E, S176D, S176K, S176R, or S176H mutation, and wherein the mutation in one or both of CH1-1 or CH1-2 is oppositely charged to the mutation in one or both of CL1 and CL 2.

In particular embodiments, the multispecific antibodies of the present disclosure may comprise:

a) A first light chain (LC 1)/heavy chain (HC1) pair, said first light chain (LC 1)/heavy chain (HC1) pair comprising

(1) A first VL region (VL1) that pairs with a first VH region (VH1) to form a first antigen binding site (VL 1);

(2) a first constant heavy chain region 1(CH1-1) operably linked to VH1 and a first constant light chain region (CL1) operably linked to VL 1; and

(3) a first heterodimerization domain (HD 1); and

b) a second light chain (LC 2)/heavy chain (HC2) pair comprising

(4) A second VL region (VL2) that pairs with a second VH region (VH2) to form a second antigen binding site;

(5) a second constant heavy chain region 1(CH1-2) operably linked to VH2 and a second constant light chain region (CL2) operably linked to VL 2; and

(6) a second heterodimerization domain (HD 2);

wherein HD1 and HD2 heterodimerize,

wherein one or both of VH1 and VH2 comprises a Q39E, Q39D, Q39K, Q39R, or Q39H mutation, and one or both of VL1 and VL2 comprises a Q38E, Q38D, Q38K, Q38R, or Q38H mutation, wherein the mutation in one or both of VH1 and VH2 is oppositely charged to the mutation in one or both of VL1 and VL2,

Wherein one or both of the CH1-1 domain and CH1-2 domain comprises an L124E, L124D, L124K, L124R, or L124H mutation, and one or both of the CL1 domain and CL2 domain comprises a V133E, V133D, V133K, V133R, or V133H mutation, and wherein the mutation in one or both of CH1-1 or CH1-2 is oppositely charged to the mutation in one or both of CL1 and CL 2.

In particular embodiments, the multispecific antibodies of the present disclosure may comprise:

a) a first light chain (LC 1)/heavy chain (HC1) pair, said first light chain (LC 1)/heavy chain (HC1) pair comprising

(1) A first VL region (VL1) that pairs with a first VH region (VH1) to form a first antigen binding site (VL 1);

(2) a first constant heavy chain region 1(CH1-1) operably linked to VH1 and a first constant light chain region (CL1) operably linked to VL 1; and

(3) a first heterodimerization domain (HD 1); and

b) a second light chain (LC 2)/heavy chain (HC2) pair comprising

(4) A second VL region (VL2) that pairs with a second VH region (VH2) to form a second antigen binding site;

(5) a second constant heavy chain region 1(CH1-2) operably linked to VH2 and a second constant light chain region (CL2) operably linked to VL 2; and

(6) A second heterodimerization domain (HD 2);

wherein HD1 and HD2 heterodimerize,

wherein the CH1-1 domain comprises an L143E or L143D mutation and the CL1 domain comprises an S176R or S176K mutation.

In particular embodiments, the multispecific antibodies of the present disclosure may comprise:

a) a first light chain (LC 1)/heavy chain (HC1) pair, said first light chain (LC 1)/heavy chain (HC1) pair comprising

(1) A first VL region (VL1) that pairs with a first VH region (VH1) to form a first antigen binding site (VL 1);

(2) a first constant heavy chain region 1(CH1-1) operably linked to VH1 and a first constant light chain region (CL1) operably linked to VL 1; and

(3) a first heterodimerization domain (HD 1); and

b) a second light chain (LC 2)/heavy chain (HC2) pair comprising

(4) A second VL region (VL2) that pairs with a second VH region (VH2) to form a second antigen binding site;

(5) a second constant heavy chain region 1(CH1-2) operably linked to VH2 and a second constant light chain region (CL2) operably linked to VL 2; and

(6) a second heterodimerization domain (HD 2);

wherein HD1 and HD2 heterodimerize,

wherein the CH1-1 domain comprises an L143R or L143K mutation and the CL1 domain comprises an S176E or S176D mutation.

In particular embodiments, the multispecific antibodies of the present disclosure may comprise:

a) a first light chain (LC 1)/heavy chain (HC1) pair, said first light chain (LC 1)/heavy chain (HC1) pair comprising

(1) A first VL region (VL1) that pairs with a first VH region (VH1) to form a first antigen binding site (VL 1);

(2) a first constant heavy chain region 1(CH1-1) operably linked to VH1 and a first constant light chain region (CL1) operably linked to VL 1; and

(3) a first heterodimerization domain (HD 1); and

b) a second light chain (LC 2)/heavy chain (HC2) pair comprising

(4) A second VL region (VL2) that pairs with a second VH region (VH2) to form a second antigen binding site;

(5) a second constant heavy chain region 1(CH1-2) operably linked to VH2 and a second constant light chain region (CL2) operably linked to VL 2; and

(6) a second heterodimerization domain (HD 2);

wherein HD1 and HD2 heterodimerize,

wherein the CH1-1 domain comprises an L143E or L143D mutation, and the CH1-2 domain comprises an L143R or L143K mutation, and the CL1 domain comprises an S176R or S176K mutation, and the CL2 domain comprises an S176E or S176D mutation.

In particular embodiments, the multispecific antibodies of the present disclosure may comprise:

a) A first light chain (LC 1)/heavy chain (HC1) pair, said first light chain (LC 1)/heavy chain (HC1) pair comprising

(1) A first VL region (VL1) that pairs with a first VH region (VH1) to form a first antigen binding site (VL 1);

(2) a first constant heavy chain region 1(CH1-1) operably linked to VH1 and a first constant light chain region (CL1) operably linked to VL 1; and

(3) a first heterodimerization domain (HD 1); and

b) a second light chain (LC 2)/heavy chain (HC2) pair comprising

(4) A second VL region (VL2) that pairs with a second VH region (VH2) to form a second antigen binding site;

(5) a second constant heavy chain region 1(CH1-2) operably linked to VH2 and a second constant light chain region (CL2) operably linked to VL 2; and

(6) a second heterodimerization domain (HD 2);

wherein HD1 and HD2 heterodimerize,

wherein the CH1-1 domain comprises an L143E or L143D mutation and the CL1 domain comprises an S176R or S176K mutation,

wherein one or both of the CH1-1 domain and the CH1-2 domain comprises a K221E mutation and one or both of the CL1 domain and the CL2 domain comprises an E123K mutation.

In particular embodiments, the multispecific antibodies of the present disclosure may comprise:

a) A first light chain (LC 1)/heavy chain (HC1) pair, said first light chain (LC 1)/heavy chain (HC1) pair comprising

(1) A first VL region (VL1) that pairs with a first VH region (VH1) to form a first antigen binding site (VL 1);

(2) a first constant heavy chain region 1(CH1-1) operably linked to VH1 and a first constant light chain region (CL1) operably linked to VL 1; and

(3) a first heterodimerization domain (HD 1); and

b) a second light chain (LC 2)/heavy chain (HC2) pair comprising

(4) A second VL region (VL2) that pairs with a second VH region (VH2) to form a second antigen binding site;

(5) a second constant heavy chain region 1(CH1-2) operably linked to VH2 and a second constant light chain region (CL2) operably linked to VL 2; and

(6) a second heterodimerization domain (HD 2);

wherein HD1 and HD2 heterodimerize,

wherein the CH1-1 domain comprises an L143R or L143K mutation and the CL1 domain comprises an S176E or S176D mutation,

wherein one or both of the CH1-1 domain and the CH1-2 domain comprises a K221E mutation and one or both of the CL1 domain and the CL2 domain comprises an E123K mutation.

In particular embodiments, the multispecific antibodies of the present disclosure may comprise:

a) A first light chain (LC 1)/heavy chain (HC1) pair, said first light chain (LC 1)/heavy chain (HC1) pair comprising

(1) A first VL region (VL1) that pairs with a first VH region (VH1) to form a first antigen binding site (VL 1);

(2) a first constant heavy chain region 1(CH1-1) operably linked to VH1 and a first constant light chain region (CL1) operably linked to VL 1; and

(3) a first heterodimerization domain (HD 1); and

b) a second light chain (LC 2)/heavy chain (HC2) pair comprising

(4) A second VL region (VL2) that pairs with a second VH region (VH2) to form a second antigen binding site;

(5) a second constant heavy chain region 1(CH1-2) operably linked to VH2 and a second constant light chain region (CL2) operably linked to VL 2; and

(6) a second heterodimerization domain (HD 2);

wherein HD1 and HD2 heterodimerize,

wherein the CH1-1 domain comprises an L143E or L143D mutation, and the CH1-2 domain comprises an L143R or L143K mutation, and the CL1 domain comprises an S176R or S176K mutation, and the CL2 domain comprises an S176E or S176D mutation,

wherein one or both of the CH1-1 domain and the CH1-2 domain comprises a K221E mutation and one or both of the CL1 domain and the CL2 domain comprises an E123K mutation.

In particular embodiments, the multispecific antibodies of the present disclosure may comprise:

a) a first light chain (LC 1)/heavy chain (HC1) pair, said first light chain (LC 1)/heavy chain (HC1) pair comprising

(1) A first VL region (VL1) that pairs with a first VH region (VH1) to form a first antigen binding site (VL 1);

(2) a first constant heavy chain region 1(CH1-1) operably linked to VH1 and a first constant light chain region (CL1) operably linked to VL 1; and

(3) a first heterodimerization domain (HD 1); and

b) a second light chain (LC 2)/heavy chain (HC2) pair comprising

(4) A second VL region (VL2) that pairs with a second VH region (VH2) to form a second antigen binding site;

(5) a second constant heavy chain region 1(CH1-2) operably linked to VH2 and a second constant light chain region (CL2) operably linked to VL 2; and

(6) a second heterodimerization domain (HD 2);

wherein HD1 and HD2 heterodimerize,

wherein the CH1-1 domain comprises an L143E or L143D mutation and the CL1 domain comprises an S176R or S176K mutation,

wherein one or both of the CH1-1 domain and the CH1-2 domain comprises a K228D mutation and one or both of the CL1 domain and the CL2 domain comprises a D122K mutation.

In particular embodiments, the multispecific antibodies of the present disclosure may comprise:

a) a first light chain (LC 1)/heavy chain (HC1) pair, said first light chain (LC 1)/heavy chain (HC1) pair comprising

(1) A first VL region (VL1) that pairs with a first VH region (VH1) to form a first antigen binding site (VL 1);

(2) a first constant heavy chain region 1(CH1-1) operably linked to VH1 and a first constant light chain region (CL1) operably linked to VL 1; and

(3) a first heterodimerization domain (HD 1); and

b) a second light chain (LC 2)/heavy chain (HC2) pair comprising

(4) A second VL region (VL2) that pairs with a second VH region (VH2) to form a second antigen binding site;

(5) a second constant heavy chain region 1(CH1-2) operably linked to VH2 and a second constant light chain region (CL2) operably linked to VL 2; and

(6) a second heterodimerization domain (HD 2);

wherein HD1 and HD2 heterodimerize,

wherein the CH1-1 domain comprises an L143R or L143K mutation and the CL1 domain comprises an S176E or S176D mutation,

wherein one or both of the CH1-1 domain and the CH1-2 domain comprises a K228D mutation and one or both of the CL1 domain and the CL2 domain comprises a D122K mutation.

In particular embodiments, the multispecific antibodies of the present disclosure may comprise:

a) a first light chain (LC 1)/heavy chain (HC1) pair, said first light chain (LC 1)/heavy chain (HC1) pair comprising

(1) A first VL region (VL1) that pairs with a first VH region (VH1) to form a first antigen binding site (VL 1);

(2) a first constant heavy chain region 1(CH1-1) operably linked to VH1 and a first constant light chain region (CL1) operably linked to VL 1; and

(3) a first heterodimerization domain (HD 1); and

b) a second light chain (LC 2)/heavy chain (HC2) pair comprising

(4) A second VL region (VL2) that pairs with a second VH region (VH2) to form a second antigen binding site;

(5) a second constant heavy chain region 1(CH1-2) operably linked to VH2 and a second constant light chain region (CL2) operably linked to VL 2; and

(6) a second heterodimerization domain (HD 2);

wherein HD1 and HD2 heterodimerize,

wherein the CH1-1 domain comprises an L143E or L143D mutation, and the CH1-2 domain comprises an L143R or L143K mutation, and the CL1 domain comprises an S176R or S176K mutation, and the CL2 domain comprises an S176E or S176D mutation,

wherein one or both of the CH1-1 domain and the CH1-2 domain comprises a K228D mutation and one or both of the CL1 domain and the CL2 domain comprises a D122K mutation.

In particular embodiments, the multispecific antibodies of the present disclosure may comprise:

a) a first light chain (LC 1)/heavy chain (HC1) pair, said first light chain (LC 1)/heavy chain (HC1) pair comprising

(1) A first VL region (VL1) that pairs with a first VH region (VH1) to form a first antigen binding site (VL 1);

(2) a first constant heavy chain region 1(CH1-1) operably linked to VH1 and a first constant light chain region (CL1) operably linked to VL 1; and

(3) a first heterodimerization domain (HD 1); and

b) a second light chain (LC 2)/heavy chain (HC2) pair comprising

(4) A second VL region (VL2) that pairs with a second VH region (VH2) to form a second antigen binding site;

(5) a second constant heavy chain region 1(CH1-2) operably linked to VH2 and a second constant light chain region (CL2) operably linked to VL 2; and

(6) a second heterodimerization domain (HD 2);

wherein HD1 and HD2 heterodimerize,

wherein the CH1-1 domain comprises an L143E or L143D mutation and the CL1 domain comprises an S176R or S176K mutation,

wherein one or both of the CH1-1 domain and the CH1-2 domain comprises K221E and K228D mutations, and one or both of the CL1 domain and the CL2 domain comprises D122K and E123K mutations.

In particular embodiments, the multispecific antibodies of the present disclosure may comprise:

a) a first light chain (LC 1)/heavy chain (HC1) pair, said first light chain (LC 1)/heavy chain (HC1) pair comprising

(1) A first VL region (VL1) that pairs with a first VH region (VH1) to form a first antigen binding site (VL 1);

(2) a first constant heavy chain region 1(CH1-1) operably linked to VH1 and a first constant light chain region (CL1) operably linked to VL 1; and

(3) a first heterodimerization domain (HD 1); and

b) a second light chain (LC 2)/heavy chain (HC2) pair comprising

(4) A second VL region (VL2) that pairs with a second VH region (VH2) to form a second antigen binding site;

(5) a second constant heavy chain region 1(CH1-2) operably linked to VH2 and a second constant light chain region (CL2) operably linked to VL 2; and

(6) a second heterodimerization domain (HD 2);

wherein HD1 and HD2 heterodimerize,

wherein the CH1-1 domain comprises an L143R or L143K mutation and the CL1 domain comprises an S176E or S176D mutation,

wherein one or both of the CH1-1 domain and the CH1-2 domain comprises K221E and K228D mutations, and one or both of the CL1 domain and the CL2 domain comprises D122K and E123K mutations.

In particular embodiments, the multispecific antibodies of the present disclosure may comprise:

a) a first light chain (LC 1)/heavy chain (HC1) pair, said first light chain (LC 1)/heavy chain (HC1) pair comprising

(1) A first VL region (VL1) that pairs with a first VH region (VH1) to form a first antigen binding site (VL 1);

(2) a first constant heavy chain region 1(CH1-1) operably linked to VH1 and a first constant light chain region (CL1) operably linked to VL 1; and

(3) a first heterodimerization domain (HD 1); and

b) a second light chain (LC 2)/heavy chain (HC2) pair comprising

(4) A second VL region (VL2) that pairs with a second VH region (VH2) to form a second antigen binding site;

(5) a second constant heavy chain region 1(CH1-2) operably linked to VH2 and a second constant light chain region (CL2) operably linked to VL 2; and

(6) a second heterodimerization domain (HD 2);

wherein HD1 and HD2 heterodimerize,

wherein the CH1-1 domain comprises an L143E or L143D mutation, and the CH1-2 domain comprises an L143R or L143K mutation, and the CL1 domain comprises an S176R or S176K mutation, and the CL2 domain comprises an S176E or S176D mutation,

wherein one or both of the CH1-1 domain and the CH1-2 domain comprises K221E and K228D mutations, and one or both of the CL1 domain and the CL2 domain comprises D122K and E123K mutations.

In particular embodiments, the multispecific antibodies of the present disclosure may comprise:

a) a first light chain (LC 1)/heavy chain (HC1) pair, said first light chain (LC 1)/heavy chain (HC1) pair comprising

(1) A first VL region (VL1) that pairs with a first VH region (VH1) to form a first antigen binding site (VL 1);

(2) a first constant heavy chain region 1(CH1-1) operably linked to VH1 and a first constant light chain region (CL1) operably linked to VL 1; and

(3) a first heterodimerization domain (HD 1); and

b) a second light chain (LC 2)/heavy chain (HC2) pair comprising

(4) A second VL region (VL2) that pairs with a second VH region (VH2) to form a second antigen binding site;

(5) a second constant heavy chain region 1(CH1-2) operably linked to VH2 and a second constant light chain region (CL2) operably linked to VL 2; and

(6) a second heterodimerization domain (HD 2);

wherein HD1 and HD2 heterodimerize,

wherein the CH1-1 domain comprises an L143E or L143D mutation and the CL1 domain comprises an S176R or S176K mutation,

wherein the VH1 domain comprises a VH39K mutation and the VL1 domain comprises a VL38E mutation.

In particular embodiments, the multispecific antibodies of the present disclosure may comprise:

a) A first light chain (LC 1)/heavy chain (HC1) pair, said first light chain (LC 1)/heavy chain (HC1) pair comprising

(1) A first VL region (VL1) that pairs with a first VH region (VH1) to form a first antigen binding site (VL 1);

(2) a first constant heavy chain region 1(CH1-1) operably linked to VH1 and a first constant light chain region (CL1) operably linked to VL 1; and

(3) a first heterodimerization domain (HD 1); and

b) a second light chain (LC 2)/heavy chain (HC2) pair comprising

(4) A second VL region (VL2) that pairs with a second VH region (VH2) to form a second antigen binding site;

(5) a second constant heavy chain region 1(CH1-2) operably linked to VH2 and a second constant light chain region (CL2) operably linked to VL 2; and

(6) a second heterodimerization domain (HD 2);

wherein HD1 and HD2 heterodimerize,

wherein the CH1-1 domain comprises an L143R or L143K mutation and the CL1 domain comprises an S176E or S176D mutation,

wherein the VH1 domain comprises a VH39E mutation and the VL1 domain comprises a VL38K mutation.

In particular embodiments, the multispecific antibodies of the present disclosure may comprise:

a) a first light chain (LC 1)/heavy chain (HC1) pair, said first light chain (LC 1)/heavy chain (HC1) pair comprising

(1) A first VL region (VL1) that pairs with a first VH region (VH1) to form a first antigen binding site (VL 1);

(2) a first constant heavy chain region 1(CH1-1) operably linked to VH1 and a first constant light chain region (CL1) operably linked to VL 1; and

(3) a first heterodimerization domain (HD 1); and

b) a second light chain (LC 2)/heavy chain (HC2) pair comprising

(4) A second VL region (VL2) that pairs with a second VH region (VH2) to form a second antigen binding site;

(5) a second constant heavy chain region 1(CH1-2) operably linked to VH2 and a second constant light chain region (CL2) operably linked to VL 2; and

(6) a second heterodimerization domain (HD 2);

wherein HD1 and HD2 heterodimerize,

wherein the CH1-1 domain comprises an L143E or L143D mutation, and the CH1-2 domain comprises an L143R or L143K mutation, and the CL1 domain comprises an S176R or S176K mutation, and the CL2 domain comprises an S176E or S176D mutation,

wherein the VH1 domain comprises a VH39K mutation and the VL1 domain comprises a VL38E mutation.

In particular embodiments, the multispecific antibodies of the present disclosure may comprise:

a) a first light chain (LC 1)/heavy chain (HC1) pair, said first light chain (LC 1)/heavy chain (HC1) pair comprising

(1) A first VL region (VL1) that pairs with a first VH region (VH1) to form a first antigen binding site (VL 1);

(2) a first constant heavy chain region 1(CH1-1) operably linked to VH1 and a first constant light chain region (CL1) operably linked to VL 1; and

(3) a first heterodimerization domain (HD 1); and

b) a second light chain (LC 2)/heavy chain (HC2) pair comprising

(4) A second VL region (VL2) that pairs with a second VH region (VH2) to form a second antigen binding site;

(5) a second constant heavy chain region 1(CH1-2) operably linked to VH2 and a second constant light chain region (CL2) operably linked to VL 2; and

(6) a second heterodimerization domain (HD 2);

wherein HD1 and HD2 heterodimerize,

wherein the CH1-1 domain comprises an L143E or L143D mutation, and the CH1-2 domain comprises an L143R or L143K mutation, and the CL1 domain comprises an S176R or S176K mutation, and the CL2 domain comprises an S176E or S176D mutation,

wherein the VH1 domain comprises a VH39K mutation and the VL1 domain comprises a VL38E mutation,

wherein the VH2 domain comprises a VH39E mutation and the VL2 domain comprises a VL38K mutation.

In particular embodiments, the multispecific antibodies of the present disclosure may comprise:

a) A first light chain (LC 1)/heavy chain (HC1) pair, said first light chain (LC 1)/heavy chain (HC1) pair comprising

(1) A first VL region (VL1) that pairs with a first VH region (VH1) to form a first antigen binding site (VL 1);

(2) a first constant heavy chain region 1(CH1-1) operably linked to VH1 and a first constant light chain region (CL1) operably linked to VL 1; and

(3) a first heterodimerization domain (HD 1); and

b) a second light chain (LC 2)/heavy chain (HC2) pair comprising

(4) A second VL region (VL2) that pairs with a second VH region (VH2) to form a second antigen binding site;

(5) a second constant heavy chain region 1(CH1-2) operably linked to VH2 and a second constant light chain region (CL2) operably linked to VL 2; and

(6) a second heterodimerization domain (HD 2);

wherein HD1 and HD2 heterodimerize,

wherein the CH1-1 domain comprises an L143E or L143D mutation and the CL1 domain comprises an S176R or S176K mutation,

wherein one or both of the CH1-1 domain and the CH1-2 domain comprises a K221E mutation and one or both of the CL1 domain and the CL2 domain comprises an E123K mutation,

wherein the VH1 domain comprises a VH39K mutation and the VL1 domain comprises a VL38E mutation.

In particular embodiments, the multispecific antibodies of the present disclosure may comprise:

a) a first light chain (LC 1)/heavy chain (HC1) pair, said first light chain (LC 1)/heavy chain (HC1) pair comprising

(1) A first VL region (VL1) that pairs with a first VH region (VH1) to form a first antigen binding site (VL 1);

(2) a first constant heavy chain region 1(CH1-1) operably linked to VH1 and a first constant light chain region (CL1) operably linked to VL 1; and

(3) a first heterodimerization domain (HD 1); and

b) a second light chain (LC 2)/heavy chain (HC2) pair comprising

(4) A second VL region (VL2) that pairs with a second VH region (VH2) to form a second antigen binding site;

(5) a second constant heavy chain region 1(CH1-2) operably linked to VH2 and a second constant light chain region (CL2) operably linked to VL 2; and

(6) a second heterodimerization domain (HD 2);

wherein HD1 and HD2 heterodimerize,

wherein the CH1-1 domain comprises an L143R or L143K mutation and the CL1 domain comprises an S176E or S176D mutation,

wherein one or both of the CH1-1 domain and the CH1-2 domain comprises a K221E mutation and one or both of the CL1 domain and the CL2 domain comprises an E123K mutation,

Wherein the VH1 domain comprises a VH39E mutation and the VL1 domain comprises a VL38K mutation.

In particular embodiments, the multispecific antibodies of the present disclosure may comprise:

a) a first light chain (LC 1)/heavy chain (HC1) pair, said first light chain (LC 1)/heavy chain (HC1) pair comprising

(1) A first VL region (VL1) that pairs with a first VH region (VH1) to form a first antigen binding site (VL 1);

(2) a first constant heavy chain region 1(CH1-1) operably linked to VH1 and a first constant light chain region (CL1) operably linked to VL 1; and

(3) a first heterodimerization domain (HD 1); and

b) a second light chain (LC 2)/heavy chain (HC2) pair comprising

(4) A second VL region (VL2) that pairs with a second VH region (VH2) to form a second antigen binding site;

(5) a second constant heavy chain region 1(CH1-2) operably linked to VH2 and a second constant light chain region (CL2) operably linked to VL 2; and

(6) a second heterodimerization domain (HD 2);

wherein HD1 and HD2 heterodimerize,

wherein the CH1-1 domain comprises an L143E or L143D mutation, and the CH1-2 domain comprises an L143R or L143K mutation, and the CL1 domain comprises an S176R or S176K mutation, and the CL2 domain comprises an S176E or S176D mutation,

Wherein one or both of the CH1-1 domain and the CH1-2 domain comprises a K221E mutation and one or both of the CL1 domain and the CL2 domain comprises an E123K mutation,

wherein the VH2 domain comprises a VH39E mutation and the VL2 domain comprises a VL38K mutation.

In particular embodiments, the multispecific antibodies of the present disclosure may comprise:

a) a first light chain (LC 1)/heavy chain (HC1) pair, said first light chain (LC 1)/heavy chain (HC1) pair comprising

(1) A first VL region (VL1) that pairs with a first VH region (VH1) to form a first antigen binding site (VL 1);

(2) a first constant heavy chain region 1(CH1-1) operably linked to VH1 and a first constant light chain region (CL1) operably linked to VL 1; and

(3) a first heterodimerization domain (HD 1); and

b) a second light chain (LC 2)/heavy chain (HC2) pair comprising

(4) A second VL region (VL2) that pairs with a second VH region (VH2) to form a second antigen binding site;

(5) a second constant heavy chain region 1(CH1-2) operably linked to VH2 and a second constant light chain region (CL2) operably linked to VL 2; and

(6) a second heterodimerization domain (HD 2);

wherein HD1 and HD2 heterodimerize,

Wherein the CH1-1 domain comprises an L143E or L143D mutation, and the CH1-2 domain comprises an L143R or L143K mutation, and the CL1 domain comprises an S176R or S176K mutation, and the CL2 domain comprises an S176E or S176D mutation,

wherein one or both of the CH1-1 domain and the CH1-2 domain comprises a K221E mutation and one or both of the CL1 domain and the CL2 domain comprises an E123K mutation,

wherein the VH1 domain comprises a VH39K mutation and the VL1 domain comprises a VL38E mutation,

wherein the VH2 domain comprises a VH39E mutation and the VL2 domain comprises a VL38K mutation.

In particular embodiments, the multispecific antibodies of the present disclosure may comprise:

a) a first light chain (LC 1)/heavy chain (HC1) pair, said first light chain (LC 1)/heavy chain (HC1) pair comprising

(1) A first VL region (VL1) that pairs with a first VH region (VH1) to form a first antigen binding site (VL 1);

(2) a first constant heavy chain region 1(CH1-1) operably linked to VH1 and a first constant light chain region (CL1) operably linked to VL 1; and

(3) a first heterodimerization domain (HD 1); and

b) a second light chain (LC 2)/heavy chain (HC2) pair comprising

(4) A second VL region (VL2) that pairs with a second VH region (VH2) to form a second antigen binding site;

(5) A second constant heavy chain region 1(CH1-2) operably linked to VH2 and a second constant light chain region (CL2) operably linked to VL 2; and

(6) a second heterodimerization domain (HD 2);

wherein HD1 and HD2 heterodimerize,

wherein the CH1-1 domain comprises an L143E or L143D mutation and the CL1 domain comprises an S176R or S176K mutation,

wherein one or both of the CH1-1 domain and the CH1-2 domain comprises a K228D mutation and one or both of the CL1 domain and the CL2 domain comprises a D122K mutation,

wherein the VH1 domain comprises a VH39K mutation and the VL1 domain comprises a VL38E mutation.

In particular embodiments, the multispecific antibodies of the present disclosure may comprise:

a) a first light chain (LC 1)/heavy chain (HC1) pair, said first light chain (LC 1)/heavy chain (HC1) pair comprising

(1) A first VL region (VL1) that pairs with a first VH region (VH1) to form a first antigen binding site (VL 1);

(2) a first constant heavy chain region 1(CH1-1) operably linked to VH1 and a first constant light chain region (CL1) operably linked to VL 1; and

(3) a first heterodimerization domain (HD 1); and

b) a second light chain (LC 2)/heavy chain (HC2) pair comprising

(4) A second VL region (VL2) that pairs with a second VH region (VH2) to form a second antigen binding site;

(5) a second constant heavy chain region 1(CH1-2) operably linked to VH2 and a second constant light chain region (CL2) operably linked to VL 2; and

(6) a second heterodimerization domain (HD 2);

wherein HD1 and HD2 heterodimerize,

wherein the CH1-1 domain comprises an L143R or L143K mutation and the CL1 domain comprises an S176E or S176D mutation,

wherein one or both of the CH1-1 domain and the CH1-2 domain comprises a K228D mutation and one or both of the CL1 domain and the CL2 domain comprises a D122K mutation,

wherein the VH1 domain comprises a VH39E mutation and the VL1 domain comprises a VL38K mutation.

In particular embodiments, the multispecific antibodies of the present disclosure may comprise:

a) a first light chain (LC 1)/heavy chain (HC1) pair, said first light chain (LC 1)/heavy chain (HC1) pair comprising

(1) A first VL region (VL1) that pairs with a first VH region (VH1) to form a first antigen binding site (VL 1);

(2) a first constant heavy chain region 1(CH1-1) operably linked to VH1 and a first constant light chain region (CL1) operably linked to VL 1; and

(3) a first heterodimerization domain (HD 1); and

b) A second light chain (LC 2)/heavy chain (HC2) pair comprising

(4) A second VL region (VL2) that pairs with a second VH region (VH2) to form a second antigen binding site;

(5) a second constant heavy chain region 1(CH1-2) operably linked to VH2 and a second constant light chain region (CL2) operably linked to VL 2; and

(6) a second heterodimerization domain (HD 2);

wherein HD1 and HD2 heterodimerize,

wherein the CH1-1 domain comprises an L143E or L143D mutation, and the CH1-2 domain comprises an L143R or L143K mutation, and the CL1 domain comprises an S176R or S176K mutation, and the CL2 domain comprises an S176E or S176D mutation,

wherein one or both of the CH1-1 domain and the CH1-2 domain comprises a K228D mutation and one or both of the CL1 domain and the CL2 domain comprises a D122K mutation,

wherein the VH2 domain comprises a VH39E mutation and the VL2 domain comprises a VL38K mutation.

In particular embodiments, the multispecific antibodies of the present disclosure may comprise:

a) a first light chain (LC 1)/heavy chain (HC1) pair, said first light chain (LC 1)/heavy chain (HC1) pair comprising

(1) A first VL region (VL1) that pairs with a first VH region (VH1) to form a first antigen binding site (VL 1);

(2) A first constant heavy chain region 1(CH1-1) operably linked to VH1 and a first constant light chain region (CL1) operably linked to VL 1; and

(3) a first heterodimerization domain (HD 1); and

b) a second light chain (LC 2)/heavy chain (HC2) pair comprising

(4) A second VL region (VL2) that pairs with a second VH region (VH2) to form a second antigen binding site;

(5) a second constant heavy chain region 1(CH1-2) operably linked to VH2 and a second constant light chain region (CL2) operably linked to VL 2; and

(6) a second heterodimerization domain (HD 2);

wherein HD1 and HD2 heterodimerize,

wherein the CH1-1 domain comprises an L143E or L143D mutation, and the CH1-2 domain comprises an L143R or L143K mutation, and the CL1 domain comprises an S176R or S176K mutation, and the CL2 domain comprises an S176E or S176D mutation,

wherein one or both of the CH1-1 domain and the CH1-2 domain comprises a K228D mutation and one or both of the CL1 domain and the CL2 domain comprises a D122K mutation,

wherein the VH1 domain comprises a VH39K mutation and the VL1 domain comprises a VL38E mutation,

wherein the VH2 domain comprises a VH39E mutation and the VL2 domain comprises a VL38K mutation.

In particular embodiments, the multispecific antibodies of the present disclosure may comprise:

a) a first light chain (LC 1)/heavy chain (HC1) pair, said first light chain (LC 1)/heavy chain (HC1) pair comprising

(1) A first VL region (VL1) that pairs with a first VH region (VH1) to form a first antigen binding site (VL 1);

(2) a first constant heavy chain region 1(CH1-1) operably linked to VH1 and a first constant light chain region (CL1) operably linked to VL 1; and

(3) a first heterodimerization domain (HD 1); and

b) a second light chain (LC 2)/heavy chain (HC2) pair comprising

(4) A second VL region (VL2) that pairs with a second VH region (VH2) to form a second antigen binding site;

(5) a second constant heavy chain region 1(CH1-2) operably linked to VH2 and a second constant light chain region (CL2) operably linked to VL 2; and

(6) a second heterodimerization domain (HD 2);

wherein HD1 and HD2 heterodimerize,

wherein the CH1-1 domain comprises an L143E or L143D mutation and the CL1 domain comprises an S176R or S176K mutation,

wherein one or both of the CH1-1 domain and the CH1-2 domain comprises K221E and K228D mutations, and one or both of the CL1 domain and the CL2 domain comprises D122K and E123K mutations,

Wherein the VH1 domain comprises a VH39K mutation and the VL1 domain comprises a VL38E mutation.

In particular embodiments, the multispecific antibodies of the present disclosure may comprise:

a) a first light chain (LC 1)/heavy chain (HC1) pair, said first light chain (LC 1)/heavy chain (HC1) pair comprising

(1) A first VL region (VL1) that pairs with a first VH region (VH1) to form a first antigen binding site (VL 1);

(2) a first constant heavy chain region 1(CH1-1) operably linked to VH1 and a first constant light chain region (CL1) operably linked to VL 1; and

(3) a first heterodimerization domain (HD 1); and

b) a second light chain (LC 2)/heavy chain (HC2) pair comprising

(4) A second VL region (VL2) that pairs with a second VH region (VH2) to form a second antigen binding site;

(5) a second constant heavy chain region 1(CH1-2) operably linked to VH2 and a second constant light chain region (CL2) operably linked to VL 2; and

(6) a second heterodimerization domain (HD 2);

wherein HD1 and HD2 heterodimerize,

wherein the CH1-1 domain comprises an L143R or L143K mutation and the CL1 domain comprises an S176E or S176D mutation,

wherein the VH1 domain comprises a VH39E mutation and the VL1 domain comprises a VL38K mutation,

Wherein one or both of the CH1-1 domain and the CH1-2 domain comprises K221E and K228D mutations, and one or both of the CL1 domain and the CL2 domain comprises D122K and E123K mutations.

In particular embodiments, the multispecific antibodies of the present disclosure may comprise:

a) a first light chain (LC 1)/heavy chain (HC1) pair, said first light chain (LC 1)/heavy chain (HC1) pair comprising

(1) A first VL region (VL1) that pairs with a first VH region (VH1) to form a first antigen binding site (VL 1);

(2) a first constant heavy chain region 1(CH1-1) operably linked to VH1 and a first constant light chain region (CL1) operably linked to VL 1; and

(3) a first heterodimerization domain (HD 1); and

b) a second light chain (LC 2)/heavy chain (HC2) pair comprising

(4) A second VL region (VL2) that pairs with a second VH region (VH2) to form a second antigen binding site;

(5) a second constant heavy chain region 1(CH1-2) operably linked to VH2 and a second constant light chain region (CL2) operably linked to VL 2; and

(6) a second heterodimerization domain (HD 2);

wherein HD1 and HD2 heterodimerize,

wherein the CH1-1 domain comprises an L143E or L143D mutation, and the CH1-2 domain comprises an L143R or L143K mutation, and the CL1 domain comprises an S176R or S176K mutation, and the CL2 domain comprises an S176E or S176D mutation,

Wherein one or both of the CH1-1 domain and the CH1-2 domain comprises K221E and K228D mutations, and one or both of the CL1 domain and the CL2 domain comprises D122K and E123K mutations,

wherein the VH2 domain comprises a VH39E mutation and the VL2 domain comprises a VL38K mutation.

In particular embodiments, the multispecific antibodies of the present disclosure may comprise:

a) a first light chain (LC 1)/heavy chain (HC1) pair, said first light chain (LC 1)/heavy chain (HC1) pair comprising

(1) A first VL region (VL1) that pairs with a first VH region (VH1) to form a first antigen binding site (VL 1);

(2) a first constant heavy chain region 1(CH1-1) operably linked to VH1 and a first constant light chain region (CL1) operably linked to VL 1; and

(3) a first heterodimerization domain (HD 1); and

b) a second light chain (LC 2)/heavy chain (HC2) pair comprising

(4) A second VL region (VL2) that pairs with a second VH region (VH2) to form a second antigen binding site;

(5) a second constant heavy chain region 1(CH1-2) operably linked to VH2 and a second constant light chain region (CL2) operably linked to VL 2; and

(6) a second heterodimerization domain (HD 2);

Wherein HD1 and HD2 heterodimerize,

wherein the CH1-1 domain comprises an L143E or L143D mutation, and the CH1-2 domain comprises an L143R or L143K mutation, and the CL1 domain comprises an S176R or S176K mutation, and the CL2 domain comprises an S176E or S176D mutation,

wherein one or both of the CH1-1 domain and the CH1-2 domain comprises K221E and K228D mutations, and one or both of the CL1 domain and the CL2 domain comprises D122K and E123K mutations,

wherein the VH1 domain comprises a VH39K mutation and the VL1 domain comprises a VL38E mutation,

wherein the VH2 domain comprises a VH39E mutation and the VL2 domain comprises a VL38K mutation.

In particular embodiments, the multispecific antibodies of the present disclosure may comprise:

a) a first light chain (LC 1)/heavy chain (HC1) pair, said first light chain (LC 1)/heavy chain (HC1) pair comprising

(1) A first VL region (VL1) that pairs with a first VH region (VH1) to form a first antigen binding site (VL 1);

(2) a first constant heavy chain region 1(CH1-1) operably linked to VH1 and a first constant light chain region (CL1) operably linked to VL 1; and

(3) a first heterodimerization domain (HD 1); and

b) a second light chain (LC 2)/heavy chain (HC2) pair comprising

(4) A second VL region (VL2) that pairs with a second VH region (VH2) to form a second antigen binding site;

(5) a second constant heavy chain region 1(CH1-2) operably linked to VH2 and a second constant light chain region (CL2) operably linked to VL 2; and

(6) a second heterodimerization domain (HD 2);

wherein HD1 and HD2 heterodimerize,

wherein one or both of the CH1-1 domain and the CH1-2 domain comprises K221E and K228D mutations, and one or both of the CL1 domain and the CL2 domain comprises D122K and E123K mutations,

wherein the VH1 domain comprises a VH39K mutation and the VL1 domain comprises a VL38E mutation.

In particular embodiments, the multispecific antibodies of the present disclosure may comprise:

a) a first light chain (LC 1)/heavy chain (HC1) pair, said first light chain (LC 1)/heavy chain (HC1) pair comprising

(1) A first VL region (VL1) that pairs with a first VH region (VH1) to form a first antigen binding site (VL 1);

(2) a first constant heavy chain region 1(CH1-1) operably linked to VH1 and a first constant light chain region (CL1) operably linked to VL 1; and

(3) a first heterodimerization domain (HD 1); and

b) a second light chain (LC 2)/heavy chain (HC2) pair comprising

(4) A second VL region (VL2) that pairs with a second VH region (VH2) to form a second antigen binding site;

(5) a second constant heavy chain region 1(CH1-2) operably linked to VH2 and a second constant light chain region (CL2) operably linked to VL 2; and

(6) a second heterodimerization domain (HD 2);

wherein HD1 and HD2 heterodimerize,

wherein one or both of the CH1-1 domain and the CH1-2 domain comprises K221E and K228D mutations, and one or both of the CL1 domain and the CL2 domain comprises D122K and E123K mutations,

wherein the VH1 domain comprises a VH39E mutation and the VL1 domain comprises a VL38K mutation,

in particular embodiments, the multispecific antibodies of the present disclosure may comprise:

a) a first light chain (LC 1)/heavy chain (HC1) pair, said first light chain (LC 1)/heavy chain (HC1) pair comprising

(1) A first VL region (VL1) that pairs with a first VH region (VH1) to form a first antigen binding site (VL 1);

(2) a first constant heavy chain region 1(CH1-1) operably linked to VH1 and a first constant light chain region (CL1) operably linked to VL 1; and

(3) a first heterodimerization domain (HD 1); and

b) a second light chain (LC 2)/heavy chain (HC2) pair comprising

(4) A second VL region (VL2) that pairs with a second VH region (VH2) to form a second antigen binding site;

(5) a second constant heavy chain region 1(CH1-2) operably linked to VH2 and a second constant light chain region (CL2) operably linked to VL 2; and

(6) a second heterodimerization domain (HD 2);

wherein HD1 and HD2 heterodimerize,

wherein one or both of the CH1-1 domain and the CH1-2 domain comprises K221E and K228D mutations, and one or both of the CL1 domain and the CL2 domain comprises D122K and E123K mutations,

wherein the VH1 domain comprises a VH39K mutation and the VL1 domain comprises a VL38E mutation,

wherein the VH2 domain comprises a VH39E mutation and the VL2 domain comprises a VL38K mutation.

VIII.Formulation/pharmaceutical composition

In certain embodiments, there is provided a pharmaceutical composition comprising a pharmaceutically acceptable carrier and a therapeutically effective amount of an antigen binding protein described herein. Some embodiments include pharmaceutical compositions comprising a therapeutically effective amount of any one of the binding proteins or binding protein-drug conjugates as described herein, mixed with a pharmaceutically or physiologically acceptable formulation selected for suitability with the mode of administration.

Acceptable formulation materials are generally non-toxic to recipients at the dosages and concentrations employed.

In some embodiments, the pharmaceutical composition may contain formulation materials for modifying, maintaining or maintaining, for example, the pH, osmolarity, viscosity, clarity, color, isotonicity, odor, sterility, stability, dissolution or release rate, adsorption or permeation of the composition. Suitable formulation materials include, but are not limited to, amino acids (e.g., glycine, glutamine, asparagine, arginine, or lysine), antimicrobials, antioxidants (e.g., ascorbic acid, sodium sulfite, or sodium bisulfite), buffers (e.g., borates, bicarbonates, Tris-HCl, citrates, phosphates, or other organic acids), bulking agents (e.g., mannitol or glycine), chelating agents (e.g., ethylenediaminetetraacetic acid (EDTA)), complexing agents (e.g., caffeine, polyvinylpyrrolidone, beta-cyclodextrin, or hydroxypropyl-beta-cyclodextrin), bulking agents, monosaccharides, disaccharides, and other carbohydrates (e.g., glucose, mannose, or dextrin), proteins (e.g., serum albumin, gelatin, or immunoglobulins), colorants, flavors and diluents, emulsifiers, hydrophilic polymers (e.g., polyvinylpyrrolidone), Low molecular weight polypeptides, salt-forming counterions (such as sodium), preservatives (such as benzalkonium chloride, benzoic acid, salicylic acid, thimerosal, phenylethyl alcohol, methyl paraben, propyl paraben, chlorhexidine, sorbic acid or hydrogen peroxide), solvents (such as glycerol, propylene glycol or polyethylene glycol), sugar alcohols (such as mannitol or sorbitol), suspending agents, surfactants or wetting agents (such as pluronics), PEG, sorbitan esters, polysorbates (such as polysorbate 20 or polysorbate 80), triton (triton), tromethamine, lecithin, cholesterol or tyloxapol (tyloxapal)), stability enhancers (such as sucrose or sorbitol), tonicity enhancers (such as alkali metal halides (e.g., sodium or potassium chloride) or mannitol sorbitol), delivery vehicles, diluents, excipients and/or pharmaceutical adjuvants (see, for example, REMINGTON' S PHARMACEUTICAL SCIENCES (18 th edition, edited by a.r. gennaro, Mack Publishing Company 1990) and subsequent versions thereof, which are incorporated herein by reference for any purpose).

In some embodiments, the optimal pharmaceutical composition will be determined by the skilled artisan based on, for example, the intended route of administration, the form of delivery, and the desired dosage. Such compositions may affect the physical state, stability, rate of in vivo release, and rate of in vivo clearance of the binding protein.

In some embodiments, the primary vehicle or carrier in the pharmaceutical composition may be aqueous or non-aqueous in nature. For example, a vehicle or carrier suitable for injection may be water, physiological saline solution, or artificial cerebrospinal fluid, possibly supplemented with other materials commonly used in compositions for parenteral administration. Neutral buffered saline or saline mixed with serum albumin are additional exemplary vehicles. Other exemplary pharmaceutical compositions comprise Tris buffer at about pH 7.0-8.5 or acetate buffer at about pH 4.0-5.5, which may further include sorbitol or a suitable substituent. In one embodiment of the present disclosure, the binding protein composition may be prepared for storage by mixing the composition of selected purity with an optional formulation in the form of a lyophilized cake or an aqueous solution. In addition, the binding proteins can be formulated as a lyophilizate using suitable excipients such as sucrose.

In some embodiments, the pharmaceutical compositions of the present disclosure may be selected for parenteral or subcutaneous delivery. Alternatively, the composition may be selected for inhalation or for delivery via the digestive tract, such as oral administration. The preparation of such pharmaceutically acceptable compositions is within the skill of the art.

In some embodiments, the formulation components are present at a concentration acceptable to the site of administration. For example, buffers are used to maintain the composition at physiological pH or slightly lower pH, typically in the pH range of about 5 to about 8.

Where parenteral administration is contemplated, the therapeutic composition employed may be in the form of a pyrogen-free parenterally acceptable aqueous solution comprising the desired binding protein in a pharmaceutically acceptable vehicle. A particularly suitable vehicle for parenteral injection is sterile distilled water in which the binding protein is formulated as a sterile isotonic solution for proper storage. Yet another preparation may involve formulating the desired molecule with an agent that provides controlled or sustained release of the product, such as injectable microspheres, bioerodible particles, polymeric compounds (such as polylactic or polyglycolic acid), beads, or liposomes, which can then be delivered via depot injection. Hyaluronic acid may also be used, and this may have the effect of promoting a sustained duration in circulation. Other suitable means for introducing the desired molecule include implantable drug delivery devices.

In one embodiment, the pharmaceutical composition may be formulated for inhalation. For example, the binding protein may be formulated as a dry powder for inhalation. The binding protein inhalation solution may also be formulated with a propellant for aerosol delivery. In yet another embodiment, the solution may be atomized.

It is also contemplated that certain formulations may be administered orally. In one embodiment of the present disclosure, the multispecific binding proteins administered in this manner may be formulated with or without those carriers typically used in the compounding of solid dosage forms (e.g., tablets and capsules). For example, the capsule may be designed to release the active portion of the formulation while maximizing bioavailability and minimizing pro-systemic degradation in the gastrointestinal tract. Other agents may be included to facilitate absorption of the binding protein. Diluents, flavoring agents, low melting waxes, vegetable oils, lubricants, suspending agents, tablet disintegrating agents, and binding agents may also be employed.

Another pharmaceutical composition may involve an effective amount of a multispecific binding protein in a mixture with non-toxic excipients suitable for the manufacture of tablets. Solutions can be prepared in unit dosage form by dissolving the tablets in sterile water or another suitable vehicle. Suitable excipients include, but are not limited to, inert diluents such as calcium carbonate, sodium carbonate or bicarbonate, lactose or calcium phosphate; or binding agents, such as starch, gelatin or acacia; or a lubricant such as magnesium stearate, stearic acid or talc.

Additional pharmaceutical compositions of the present disclosure will be apparent to those skilled in the art, including formulations comprising binding proteins in sustained or controlled delivery formulations. Techniques for formulating a variety of other sustained or controlled delivery means (e.g., liposome carriers, bioerodible microparticles or porous beads, and depot injections) are also known to those skilled in the art. Further examples of sustained-release formulations include semipermeable polymer matrices in the form of shaped articles (e.g., films) or microcapsules. The sustained release matrix may comprise a polyester, a hydrogel, a polylactide, a copolymer of L-glutamic acid and gamma ethyl-L-glutamate, poly (2-hydroxyethyl-methacrylate), ethylene vinyl acetate, or poly D (-) -3-hydroxybutyric acid. Sustained release compositions may also include liposomes, which may be prepared by any of several methods known in the art.

In some embodiments, the pharmaceutical composition to be used for in vivo administration must generally be sterile. This can be achieved by filtration through sterile filtration membranes. In the case of lyophilization of the composition, sterilization can be performed using this method either before or after lyophilization and reconstitution. Compositions for parenteral administration may be stored in lyophilized form or in solution form. In addition, parenteral compositions are typically placed into a container having a sterile access port, such as an intravenous bag or vial having a stopper pierceable by a hypodermic injection needle.

Once the pharmaceutical composition has been formulated, it can be stored in sterile vials as a solution, suspension, gel, emulsion, solid, or as a dehydrated or lyophilized powder. Such formulations can be stored in a ready-to-use form or in a form that requires reconstitution prior to administration (e.g., lyophilization).

The present disclosure also encompasses kits for producing single dose administration units. The kits may each contain both a first container having a dried multispecific binding protein and a second container having an aqueous formulation. Also included within the scope of the present disclosure are kits containing single and multi-chamber pre-filled syringes (e.g., liquid syringes and lyophilizate syringes).

The effective amount of the binding protein pharmaceutical composition to be employed therapeutically will depend, for example, on the context of the treatment and the target. Those skilled in the art will recognize that appropriate dosage levels for treatment will thus vary, in part, according to the following: the molecule delivered, the indication for which the binding protein is used, the route of administration, and the size (body weight, body surface or organ size) and condition (age and general health) of the patient. Thus, the clinician can gradually adjust the dosage and modify the route of administration to obtain the best therapeutic effect.

The frequency of administration will depend on the pharmacokinetic parameters of the binding protein in the formulation used. Typically, the clinician will administer the composition until a dosage is reached that achieves the desired effect. Thus, the compositions may be administered as a single dose, as two or more doses over time (which may or may not contain equal amounts of the desired molecule), or as a continuous infusion via an implanted device or catheter. Further modification of the appropriate dosage is routinely performed by those of ordinary skill in the art and is within the scope of tasks routinely performed by those of ordinary skill in the art. Appropriate dosages may be determined by using appropriate dose-response data.

The route of administration of the pharmaceutical composition is consistent with known methods, e.g., oral; by injection via intravenous, intraperitoneal, intracerebral (intraparenchymal), intracerebroventricular, intramuscular, intraocular, intraarterial, intraportal, or intralesional routes; by a sustained release system; or by an implant device. Where desired, the composition may be administered by bolus injection, or continuously by infusion, or by an implanted device.

In some embodiments, the composition may also be administered topically via implantation of a membrane, sponge, or other suitable material onto which the desired molecule has been adsorbed or encapsulated. Where an implanted device is used, the device may be implanted into any suitable tissue or organ, and delivery of the desired molecule may be via diffusion, timed release bolus injection, or continuous administration.

VIII.Methods of treatment/use

Another aspect of the present disclosure is a multispecific antibody and/or antigen-binding protein for use as a medicament as described herein.

In certain embodiments, there is provided a method of treating a disorder in which antigen activity is detrimental, the method comprising administering to a subject in need thereof an effective amount of an antigen binding protein described herein.

The binding proteins can be used in any known assay method, such as competitive binding assays, direct and indirect sandwich assays, and immunoprecipitation assays, for the detection and quantification of one or more target antigens. The binding protein will bind the one or more target antigens with an affinity suitable for the assay method used.

For diagnostic applications, in some embodiments, the binding protein may be labeled with a detectable moiety. The detectable moiety may be capable of producing a detectable signal, either directly or indirectlyAny portion of the signal is measured. For example, the detectable moiety may be a radioisotope, such as3H、14C、32P、35S、125I、99Tc、111In or67Ga; fluorescent or chemiluminescent compounds, such as fluorescein isothiocyanate, rhodamine or luciferin; or an enzyme such as alkaline phosphatase, beta-galactosidase or horseradish peroxidase.

Binding proteins may also be used for in vivo imaging. Binding proteins labeled with a detectable moiety can be administered to an animal, for example, into the bloodstream, and the presence and location of the labeled antibody in the host determined. The binding protein may be labeled with any moiety detectable in animals by nuclear magnetic resonance, radiology, or other detection means known in the art.

For clinical or research applications, in some embodiments, the binding protein may be conjugated to a cytotoxic agent. A variety of antibodies (i.e., antibody-drug conjugates) coupled to cytotoxic agents have been used to target cytotoxic payloads to specific tumor cells. Cytotoxic agents and linkers that conjugate the agents to antibodies are known in the art; see, e.g., Parslow, A.C. et al (2016) Biomedicines 4:14 and Kalim, M.et al (2017) Drug Des.Devel.Ther.11: 2265-2276.

The present disclosure also relates to kits comprising binding proteins and other reagents useful for detecting the level of a target antigen in a biological sample. Such reagents may include detectable labels, blocking sera, positive and negative control samples, and detection reagents. In some embodiments, the kit comprises a composition comprising any of the binding proteins, polynucleotides, vectors, vector systems, and/or host cells described herein. In some embodiments, a kit comprises a container and a label or package insert on or associated with the container. Suitable containers include, for example, bottles, vials, syringes, IV bags, and the like. The container may be made of a variety of materials such as glass or plastic. The container contains a composition that is effective, by itself or in combination with another composition, in the treatment, prevention and/or diagnosis of a condition, and the container may have a sterile access port (e.g., the container may be an intravenous bag or a vial having a stopper pierceable by a hypodermic injection needle). In some embodiments, the label or package insert indicates that the composition is for use in the prevention, diagnosis, and/or treatment of a selected condition. Alternatively or additionally, the article of manufacture or kit may further comprise a second (or third) container comprising a pharmaceutically acceptable buffer, such as bacteriostatic water for injection (BWFI), phosphate buffered saline, Ringer's solution, and dextrose solution. The article or kit may further include other materials as desired from a commercial and user perspective, including other buffers, diluents, filters, needles and syringes.

In some embodiments, the binding proteins of the disclosure are administered to a patient in need thereof for the treatment or prevention of cancer. In some embodiments, the disclosure relates to methods of preventing and/or treating a proliferative disease or disorder (e.g., cancer). In some embodiments, the method comprises administering to the patient a therapeutically effective amount of at least one binding protein described herein or a pharmaceutical composition related thereto. In some embodiments, the patient is a human.

In some embodiments, the at least one binding protein is administered in combination with one or more anti-cancer therapies (e.g., any anti-cancer therapy known in the art, such as a chemotherapeutic agent or therapy). In some embodiments, the at least one binding protein is administered prior to the one or more anti-cancer therapies. In some embodiments, the at least one binding protein is administered concurrently with the one or more anti-cancer therapies. In some embodiments, the at least one binding protein is administered after one or more anti-cancer therapies.

Examples

Before the present invention is described in detail below, it is to be understood that this invention is not limited to the particular methodology, protocols, and reagents described herein as these may vary. It is also to be understood that the terminology used herein is for the purpose of describing particular embodiments only, and is not intended to limit the scope of the present invention, which will be limited only by the appended claims. Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art. In certain embodiments, the terms used herein are defined as described in the following documents: "A multilingual gloss of biotechnology terms (IUPAC Recommendations)," Leuenberger, H.G.W, Nagel, B. and Kolb, eds. H.1995, Helvetica Chimica Acta, CH-4010Basel, Switzerland. Unless defined otherwise herein, scientific and technical terms used herein have the same meaning as commonly understood by one of ordinary skill in the art. In the case of any potential ambiguity, the definitions provided herein take precedence over any dictionary or extrinsic definitions. Unless the context requires otherwise, singular terms shall include the plural and plural terms shall include the singular. The use of "or" means "and/or" unless stated otherwise. The use of the term "including" as well as other forms such as "includes" and "included" is not limiting. As used herein, the singular forms "a", "an" and "the" include plural referents unless otherwise specified. Thus, for example, reference to "a protein" includes a plurality of protein molecules.

Furthermore, unless otherwise indicated, the experiments described herein employ conventional molecular and cellular biological and immunological techniques within the skill of the art. Such techniques are well known to the skilled worker and are explained fully in the literature. See, for example, Ausubel et al, Current Protocols in Molecular Biology, John Wiley & Sons, Inc., NY, N.Y. (1987. Act. 2008), including all supplements, Molecular Cloning: A Laboratory Manual (fourth edition) MR Green and J.Sambrook, and Harlow et al, Antibodies: A Laboratory Manual, Chapter 14, Cold Spring Harbor Laboratory, Cold Spring Harbor (2013, 2 nd edition).

Several documents are cited throughout the text of this specification. Each document (including all patents, patent applications, scientific publications, manufacturer specifications, instructions, etc.) cited herein, supra or infra, is hereby incorporated by reference in its entirety. Nothing herein is to be construed as an admission that the invention is not entitled to antedate such disclosure by virtue of prior invention.

Hereinafter, elements of the present invention will be described. These elements are listed with particular embodiments, however, it is understood that these particular embodiments can be combined in any manner and in any number to produce additional embodiments. The examples and specific embodiments described differently should not be construed as limiting the invention to only the embodiments explicitly described. This description should be understood to support and encompass embodiments combining the explicitly described embodiments with any number of the disclosed elements. Moreover, any permutation and combination of all the elements described in this application should be considered to be disclosed in the specification of this application unless the context indicates otherwise.

Generally, the terms used in connection with cell and tissue culture, molecular biology, immunology, microbiology, genetics and protein and nucleic acid chemistry and hybridization described herein are terms well known and commonly used in the art. Unless otherwise indicated, the methods and techniques provided herein are generally performed according to conventional methods well known in the art and as described in various general and more specific references that are cited and discussed throughout the present specification. The enzymatic reactions and purification techniques are performed according to the manufacturer's instructions, as is commonly done in the art or as described herein. The terminology used in connection with, and the laboratory procedures and techniques of, analytical chemistry, synthetic organic chemistry, and medical and pharmaceutical chemistry described herein are those well known and commonly used in the art. Conventional techniques are used for chemical synthesis, chemical analysis, pharmaceutical preparation, formulation and delivery, and treatment of patients.

Fab assembly is driven by VH/VL and CH1/CL domain interactions. Multiple multispecific constructs were generated with different structures containing different Fab interface mutations. The ability of the combined mutant pairs within the CH1/CL and VH/VL interfaces to direct correct pairing was investigated and described in the examples below.

Example 1: general expression and purification scheme

Expression of bispecific and trispecific molecules

HEK 293-FS cells grown in F17 serum free suspension medium (Invitrogen) were transfected with the indicated light chain encoding plasmid and heavy chain encoding plasmid using polyethyleneimine transfection reagent. After 7 days of incubation at 37 ℃, cells were removed by centrifugation and the supernatant was passed through a 0.22 μm filter to remove particles.

For purification, the antibody was captured on a MabSelect Sure column (Cat: 11-0034-93, GE Healthcare) and eluted with 0.1M citrate buffer (pH 3.0) and directly desalted using a HiPrep 26/10 desalting column (Cat: 17-05087-02, GE Healthcare). After fine purification (polising) of the protein by Size Exclusion Chromatography (SEC) using Superdex 20026/60 (GE) and a final ultrafiltration concentration step, the protein was used for further characterization.

Analytical Size Exclusion Chromatography (SEC)

Analytical SEC was performed using a BioSECcurity instrument (PSS Polymer) with a TSKgel SuperSW3000 column (4,6 mm. times.300 mm) and a TSKgel SuperSW HPLC protective column (Tosoh Bioscience) at 25 ℃. The assay was run at a flow rate of 0.25ml/min using 250mM NaCl, 100mM sodium phosphate (pH 6.7), with detection at 280nm and 260 nm. Static light scattering was detected at 436 nm. Mu.l of protein sample (1mg/ml) was applied to the column. Data evaluation was performed using WinGPC software version 8.1 (PSS Polymer). For molecular weight estimation, the SEC column was calibrated with protein standards ranging in molecular weight from 6.5 to 670 kDa.

Analytical Hydrophobic Interaction Chromatography (HIC)

Analytical HIC was performed using an LC10 HPLC instrument (Shimadzu) with TSKgel Ether-5PW (10 μm, 2X75mm) (Tosoh Bioscience) at 25 ℃. The assay was run at a flow rate of 0.1ml/min, with detection at 280 nm. Mu.g of undiluted protein sample was applied to the column. Gradient elution was from 0 to 30min (0% to 100% B), followed by re-equilibration for 10min at 100% B and 15 min. Buffer A consisted of 1.5M ammonium sulfate, 25mM sodium phosphate (pH 7.0). Buffer B consisted of 25mM sodium phosphate (pH 7.0). Data evaluation was performed using Labsolutions software version 5.85 (Shimadzu).

Mass Spectrum (MS)

Protein integrity was analyzed by LC-MS. Protein samples were deglycosylated with 12.5 μ g protein (diluted to 0.5mg/ml in D-PBS buffer treated with 0.5 μ l PNGaseF (glycerol free, New England Biolabs)) at 37 ℃ for 15 hours. LC-MS analysis was performed using a 6540UHD Accurate-Mass Q-TOF LC/MS instrument (Agilent). Reverse Phase (RP) chromatography was performed using Poroshell 300SB-C8(5 μm, 75x0.5mm) (Agilent) and a guard column Poroshell 300SB-C8(5 μm, 2.1x12.5mm) (Agilent) at 180 μ L/min. Eluents were LC water, 0.1% formic acid (a), and 90% acetonitrile, 10% LC water, 0.1% formic acid (B). Mu.g of protein was injected onto the column and eluted using a linear gradient from 0% to 100% B over 13 min. Data analysis was performed using MassHunter software b.06 (Agilent). Molecular masses were calculated based on the amino acid sequence of the protein using the GPMAW software version 9.13a2 (Lighthouse Data).

Surface Plasmon Resonance (SPR)

Binding of antigen to the antibody construct was measured using Surface Plasmon Resonance (SPR) with BIAcore 3000 instrument (GE Healthcare) and HBS-EP buffer (GE Healthcare). Anti-human Fc capture antibodies (human antibody capture kit, GE Life Sciences) were immobilized via primary amine groups (11000RU) on a research grade CM5 chip (GE Life Sciences) using standard procedures. The ligand was captured at a flow rate of 10 μ l/min at an adjusted RU value resulting in a maximum analyte binding of 30 RU. The tested antibody constructs were used as analytes and injected at 100nM concentration for 240 seconds at 300 seconds dissociation time and 30 μ L/min flow rate. Binding kinetics measurements were performed with the captured antibody by injecting two-fold serial dilutions of 3nM to 100nM of the analyte. The chip surface was regenerated by 2min injection with the regeneration buffer provided by the capture kit. The sensorgrams were double referenced with a blank chip surface and a HBS-EP buffer blank. Data analysis was performed using BIAevaluation software version 4.1.

Differential Scanning Fluorometry (DSF)

Melting point Tm data was determined using Differential Scanning Fluorimetry (DSF). Samples were diluted in white 96-well half-skirt (BIORAD) in D-PBS buffer (Invitrogen) to a final concentration of 0.2 μ g/μ l, including a 4x concentrated solution of SYPRO-Orange dye (Invitrogen, 5000x stock in DMSO) in D-PBS. All measurements were performed in duplicate using a myq 2 real-time PCR instrument (BIORAD). The negative first derivative curve (-d (RFU)/dT) of the melting curve was generated in iQ5 software version 2.1 (BIORAD). The data were then exported into Excel for Tm determination and graphical display of the data.

The sequences of selected tandem Fab, trispecific CODV and bispecific Y-shaped antibodies are listed in tables 2, 3 and 4 below.

Table 2-amino acid sequences of selected tandem Fab antibodies.

Table 3-amino acid sequence of trispecific CODV antibodies.

Table 4-amino acid sequence of bispecific Y-antibodies.

Example 2: mutations in CODV forms that enable heterodimerization

Three different mutants of the CODV antibody format were explored for expression, yield and homogeneity. The first form comprises a CODV antibody with a knob structure mutation in the Fc domain and a CH1/κ MUT4 mutation in CODV-Fab (CH 1: L143Q, S188V; Ck: V133T, S176V) and a CH1/κ CR3 mutation in Fab2 (CH 1: T192E; Ck: N137K, S114A) (fig. 1A). The second form comprises a CODV antibody with a knob-and-hole structural mutation in the Fc domain and a MUT4/CR3 mutation in the first form, in combination with electrostatic mutations VH39E/VL38K in both CODV-Fab and Fab2 (fig. 1B). The third form comprises a CODV antibody with a knob-and-hole structural mutation in the Fc domain and a second form of MUT4/CR3 mutation and electrostatic mutation in combination with the disulfide stabilizing mutation VH44C/VL100C on the CODV-Fab (fig. 1C).

A summary of the electrostatic mutations and disulfide bond formation mutations in the variable regions is shown in table 4. A summary of the CH1/CL κ mutations is shown in Table 5. The details of the expression and purification scheme are described in example 1. Fig. 2A-2G graphically depict data from various CODV antibody formats tested.

Table 5 below summarizes the results of the characterization of various CODV antibody formats tested on ([ OX40 x PD1] + GITR) trispecific CODV antibodies. CH1/Ck mutation: CODV-Fab (CH 1: L143Q, S188V; Ck: V133T, S176V) and Fab (CH 1: T192E; Ck: N137K, S114A). CM (charged mutation): CODV-Fab (VH39E/VL38K) and Fab2(VH39K/VL 38E). ds (disulfide stabilization mutation): VH44Cys/VL100 Cys. As shown below in table 5, introducing a mutation within the CH1/CL interface increased the onset melting point (Tm onset), indicating an increase in thermal stability. Further incorporation of VH/VL modifications significantly increased correct pairing as shown by HIC and MS (see fig. 2A-2G). The combination of the CH 1/clk mutant group (CR3 and MUT4) with disulfide stabilizing mutations showed promising results, exhibiting high HIC monomer content and increased thermal stability. The combination of the CH 1/clk mutant group (CR3 and MUT4), disulfide-stabilized mutations and opposite charge mutations also showed promising results.

Table 5-summary of expression and biophysical characterization of trispecific CODV antibodies containing different Fab interface mutations ([ OX40 x PD1] + GITR).

Example 3: mutations in tandem Fab formats enabling heterodimerization

Two conformations of tandem Fab antibodies were explored. Each conformation was further tested for efficient expression, purification and homogeneity. The first format consists of an open conformation of tandem fabs, where the Y-shaped antibody is attached to another Fab fragment through a flexible linker on the VH domain (see fig. 3A and 3B). The second tandem Fab consists of a closed conformation of tandem Fab in which the linkers in the VH domains described above can form disulfide bonds (see fig. 3C and 3D).

Two mutants, one each in the open-tandem Fab and closed-tandem Fab conformations, were explored for efficient expression, purification, and homogeneity. The first mutant consisted of an open fab with only MUT4 and CR3 mutations (fig. 3A). The second mutant consisted of an open fab with a combination of MUT4 and CR3 mutations and oppositely charged mutations (fig. 3B).

Two mutants, each with a closed-tandem Fab conformation, were explored for efficient expression, purification and homogeneity. The first mutant consisted of a closed fab with only MUT4 and CR3 mutations (fig. 3C). The second mutant consisted of a closed fab with a combination of MUT4 and CR3 mutations and oppositely charged mutations (fig. 3D).

As shown in table 6 below, the combination of the CH1/CL mutation with the VH/VL interface mutation increased the target HIC profile compared to the examples where only the CH1/CL modification was introduced. Thus, combining mutations reduces the amount of mis-paired species and increases the amount of correctly paired molecules (target HIC peaks). The strength of the pairing result depends on the antibody sequence and domain alignment.

Table 6-yield, SEC, HIC and binding affinity data for tandem Fab antibodies.

Only open configuration tandem fabs with MUT4/CR3 mutations yielded approximately 68% correct pairings. However, it was observed that the introduction of oppositely charged mutations increased the yield and homogeneity of the correct polypeptide from 68% to 96% (fig. 4).

The open and closed configurations were compared side-by-side with anti-CD 40 x anti-PD-L1 antibody. As can be seen in fig. 5A-5D, the closed configuration results in lower yield and purity compared to the open configuration as measured by HIC. The addition of oppositely charged mutations in combination with the CH1/CL κ mutation (CR3/MUT4) also improved purity. anti-PD-1 x anti-OX 40 antibodies supported these effects, as shown in fig. 6A-6C. The open configuration results in higher yield and purity. The inclusion of the opposite charge mutations further improves yield and purity.

Example 4: y-shaped antibody mutation

Orthogonal Fab design in bispecific antibodies with Y-shaped structures was achieved by introducing knob-and-hole structure mutations (knobs: S354C, T366W; knobs: Y349C, T366S, L368A, Y407V) into the CH3 domain of the corresponding heavy chain. Two different Fab arms contained mutations as described in fig. 8A and fig. 8B, as well as the mutations shown in tables 7-9 below. The mutations in FIG. 8A are the CH 1/kappa MUT4 mutations in Fab1 (CH 1: L143Q, S188V; Ck: V133T, S176V) and the CH 1/kappa CR3 mutations in Fab2 (CH 1: T192E; Ck: N137K, S114A). The mutations in figure 8B are the MUT4/CR3 mutations of figure 8A, in combination with the electrostatic mutations VH39E/VL38K in both Fab1 and Fab 2.

anti-PD-1 x anti-OX 40 antibodies with various mutations were tested. Table 7 below provides a summary of the expression and biophysical characterization of variants of PD-1x OX40 bispecific antibodies whose Fab interface mutations were varied. Modification of only CH1/CL or modification of only VH/VL is ineffective to prevent mis-pairing. The proposed combination of CH1/CL mutations with VH39/VL38 interfacial mutations significantly affected guided pairing as determined by HIC and MS (see fig. 9A-9C).

Table 7-expression and biophysical characterization of variants of anti-PD-1 x anti-OX 40Y-shaped antibodies.

anti-CD 40 x anti-PD-L1 antibodies with various mutations were tested. Table 8 below provides a summary of the expression and biophysical characterization of variants of the CD40 x PD-L1 bispecific antibody whose Fab interface mutations were varied. Modification of only CH1/CL or modification of only VH/VL is ineffective to prevent mis-pairing. The combination of the CH1/CL mutation with the VH39/VL38 interfacial mutation significantly affected guided pairing as determined by HIC and MS.

Table 8-expression and biophysical characterization of variants of anti-CD 40 x anti-PD-L1Y-shaped antibody.

anti-PD-1 x anti-OX 40 antibodies were tested with various mutations that differed from previous anti-PD-1 x anti-OX 40 antibodies. Table 9 below provides a summary of the expression and biophysical characterization of variants of PD 1x OX40 bispecific antibodies with altered Fab interface mutations. As shown in table 9, the paired CH1/CL mutations L143/S176 or L124/V133 combined with the VH/VL interface mutation at position VH38/VL39 significantly increased the amount of correctly paired species as observed by HIC profiling and verified by MS analysis (fig. 9A-9C and fig. 10A-10L).

Table 9-expression and biophysical characterization of variants of anti-PD-1 x anti-OX 40Y-shaped antibodies.

For better comparison between constructs, the antigen binding levels in the SPR data in tables 7-9 are reported relative to the captured antibody levels (RU antigen/RU Fc capture). SPR data in tables 7-9 show that all antibody constructs tested bind to their respective antigens at comparable levels of binding.

Example 5: general expression and purification scheme for examples 6 and 7

Analytical Size Exclusion Chromatography (SEC)

Analytical SEC was performed using a BioSECcurity instrument (PSS Polymer) with an AdvanceBio 300 column (4.6 mm. times.300 mm) and an AdvanceBio 300 guard column (Agilent Technologies) at 25 ℃. The assay was run at a flow rate of 0.5ml/min using 2 Xconcentrated D-PBS buffer (Thermo Fisher Scientific) and detection was at 280 nm. A10. mu.l protein sample (1mg/ml) was applied to the column. Data evaluation was performed using WinGPC software version 8.1 (PSS Polymer). For molecular weight estimation, the SEC columns were calibrated with a protein calibration standard mixture (Agilent Technologies).

Analytical Hydrophobic Interaction Chromatography (HIC)

Analytical HIC was performed using an LC10 HPLC instrument (Shimadzu) or a Vanquish HPLC instrument (Thermo Fisher Scientific) equipped with a TSKgel Butyl-NPR column (2.5 μm, 4.6 × 35mm) (Tosoh Bioscience) at 25 ℃. The assay was run at a flow rate of 1ml/min, with detection at 280 nm. Mu.g of undiluted protein sample was applied to the column. Gradient elution was from 15% B to 85% B, followed by 1min to 100% B, then 1min to 15% B, then 3 min at 15% B equilibration over 7 min. Buffer A consisted of 1.5M ammonium sulfate, 25mM sodium phosphate (pH 7.0). Buffer B consisted of 25mM sodium phosphate (pH 7.0). Data evaluation was performed using Labsolutions software version 5.85 (Shimadzu) or Chromeleon 7 software (Thermo Fisher Scientific).

Mass Spectrum (MS)

Protein integrity and potential mis-pairing of heterodimeric constructs were analyzed by LC-mass spectrometry (LC-MS). Protein samples were deglycosylated with 12.5 μ g protein (diluted to 0.17mg/ml in LC-MS grade water (Thermo Scientific) treated with 0.5 μ l PNGaseF (glycerol free, New England Biolabs)) at 37 ℃ for 16 h. LC-MS analysis was performed using a Thermo Fisher Orbitrap Lumos LC/MS instrument. Reversed Phase (RP) chromatography was performed using a MabPac RP HPLC column (analytical 4 μm particle size, 2.1X100mm) (Thermo Scientific) at 300 μ L/min. Eluents were LC water, 0.1% formic acid (a), and 90% acetonitrile, 10% LC water, 0.1% formic acid (B). Mu.g of protein was injected onto the column and eluted using a linear gradient from 0% to 95% B over 12 min. Data analysis was performed using the expression software 13.0.3 (Genedata). Molecular masses were calculated based on the amino acid sequence of the protein using the GPMAW software version 10.32b1 (Lighthouse data).

Surface Plasmon Resonance (SPR)

Binding of antigen to the antibody construct was measured using Surface Plasmon Resonance (SPR) with BIAcore T200 instrument (GE Healthcare) and HBS-EP + buffer (GE Healthcare). For the assessment of the relative binding level (% Rmax) of the antibodies to their respective antigens, the antibodies were captured to the sensor chip by anti-Fc affinity capture. In this assay, recombinant human antigens (PD-L1-His (9049-B7-100, R & D Systems), CD40-His (10774-H08H, Sino Biological), TNF α (130-094-022, Miltenyi), GITR-His (internal production), PD1-His (8986-PD-100, R & D Systems), CD3 ε -FLAG-His (# CT038-H2508H, Sino Biological) and human CD123(#301-R3/CF, R & D Systems)) were used. Anti-human Fc capture antibodies (human antibody capture kit, GE Life Sciences) were immobilized via primary amine groups (11000RU) on a research grade CM5 chip (GE Life Sciences) using standard procedures. The antibody was captured at a flow rate of 10 μ Ι/min at an adjusted RU value resulting in maximum analyte binding of 10 to 30 RU. Antigens were used as analytes and injected at 400nM and 100nM concentration (for CD3 ∈ δ -FLAG-His) or at 100nM concentration (for all other antigens used). The antigen was injected for 240 seconds at a flow rate of 30. mu.L/min with a dissociation time of 300 seconds. The chip surface was regenerated by 2min injection with the regeneration buffer provided by the capture kit. The sensorgrams were double referenced with a blank chip surface and a HBS-EP buffer blank. Data analysis and binding level determination were performed using Biacore 8K evaluation software version 1.11.7442 (GE Healthcare). The% Rmax value was calculated using the maximum binding level divided by the theoretical Rmax value. From the capture level rcrap, binding stoichiometry N, and antibody molecular weight mw (ab) and antigen molecular weight mw (ag), Rmax values were calculated from Rmax (rcrap) N (mw (ag)/mw (ab).

Nanometer differential scanning fluorimetry (NanoDSF)

The onset temperature (tstart) and melting point (Tm) of protein denaturation was determined using nano differential scanning fluorimetry (nanoDSF). Samples were diluted in formulation buffer to a final concentration of 0.5 μ g/μ l and loaded in duplicate into nanoDSF capillaries (Nanotemper Technologies). All measurements were performed using a Prometheus nt. From 20 ℃ to 95 ℃ at a heating rate of 1 ℃/min. Data were recorded using pr. thermcontrol software version 2.3.1 (nanotepmper Technologies) and analyzed using PR. stability analysis software version 1.0.3 (nanotepmper Technologies).

Example 6: additional Y-shaped antibody mutations

Additional bispecific antibodies with Y-shaped structures were generated and tested with different sets of mutations. Specifically, anti-PD-1 x anti-OX 40 antibodies having the mutations listed in table 10 below were employed. The data below shows that all Y-antibodies with Fab mutations have reduced mis-pairing compared to the wild type control. Antibodies (protein ID Y61) with the VH39/VL38 oppositely charged mutant group, the L143/S176 oppositely charged mutant group and the combined K221E: K228D/E123K: D122K oppositely charged mutant group showed excellent results, no mis-pairings were detected.

Table 10-expression and biophysical characterization of variants of anti-PD-1 x anti-OX 40Y-shaped antibodies.

Example 7: additional mutations in the Y-shaped antibody format that enable heterodimerization

Additional Y-shaped antibodies with different heterodimerization mutation sets were tested. Table 11 below lists the biophysical characterization data for each Y-shaped antibody tested. As enumerated in table 11 and fig. 11A-11E, "CM 1" refers to the VH39K/VL38E mutant pair, "CM 2" refers to the VH39E/VL38K mutant pair, "CM 3" refers to the CH 1L 143E or L143D/CL S176R or S176K mutant pair, "CM 4" refers to the CH 1L 143R or L143K/CL S176E or S176D mutant pair, and "NN 3" refers to the CH 1K 221E and K228D/CL D122K and E123K mutant pairs.

Table 11-heterodimerization mutations of tandem Fab antibodies.

As shown in table 11 and fig. 11A-11E, the various heterodimerization mutations used each reduced mis-pairing compared to the wild-type antibody. In the case of an anti-TNF x anti-OX 40 antibody (Y49) that is known to have no mispairings in its wild-type configuration, inclusion of the heterodimerization mutation had no negative effect on the mispairings (fig. 11C).

In addition to the biophysical characterization above, the T cell engager antibody CD3 x CD123 was in a cell-based cytotoxicity assay. Bispecific antibody molecules were analyzed in cytotoxicity assays using primary human T cells. Human peripheral monocytes from healthy donor blood were isolated in Leucosep tubes (Greiner Bio-One, #227290) using 15ml Histopaque (Sigma-Aldrich, #10771) and centrifugation at 1000Xg for 10 min. The isolated PBMCs were washed twice in autoMACS wash buffer (Miltenyi Biotec, # 130-. Primary human T cells were isolated from human PBMC using the manufacturer's protocol using a MACSpro separator (Miltenyi Biotec) and a Pan T cell isolation kit (Miltenyi Biotec, # 130-. Isolating human T cells at 5x10 6The cells/mL were resuspended in RPMI GlutaMAX I medium (Gibco, #72400) supplemented with 10% FCS HI (Gibco, # 10082-147). THP-1 target cells (ADCC TIB-202) were stained with 1. mu.M CFSE (Invitrogen, # C1157) for 15min at 37 ℃ prior to cytotoxicity assays. Cells were washed twice in RPMI + GlutaMAX I medium and centrifuged at 400x g for 5 min. THP-1 target cells were treated at 5X105Individual cells/mL were resuspended in RPMI medium supplemented with 10% FCS HI. Combining CFSE-labeled THP-1 cells with humansThe pan T cells were mixed at an effector to target ratio of 10:1 and seeded in 96-well assay plates (Greiner BioOne, #650185) in a total volume of 100. mu.l/well. Bispecific antibody molecules were added to cells in a volume of 5 μ L/well in 11 dilution series starting from 10nM to 0nM (1:6 dilution), and at 37 ℃ and 5% CO2The mixture was incubated for 20 hours. After incubation, cells were stained with 5. mu.g/ml 7-AAD (Invitrogen, # A1310) for 30min at 4 ℃. To determine cytotoxicity, dead target cells were measured by gating CFSE/7-AAD double positive THP-1 cells on a LSRII flow cytometer (BD) and EC50 values were determined with Xlfit software.

As shown in figure 12, all three CD3 x CD123 bispecific antibodies displayed comparable and robust activity. The presence of heterodimerization mutations in Y56 and Y57 did not negatively affect activity, however, chain mispairing was reduced.

196页详细技术资料下载
上一篇:一种医用注射器针头装配设备
下一篇:抗-SEMA3A抗体及其用于治疗眼或眼部疾病的用途

网友询问留言

已有0条留言

还没有人留言评论。精彩留言会获得点赞!

精彩留言,会给你点赞!