Heteromultimeric proteins and methods of use thereof

文档序号:395295 发布日期:2021-12-14 浏览:20次 中文

阅读说明:本技术 异多聚体蛋白质及其使用方法 (Heteromultimeric proteins and methods of use thereof ) 是由 Y.刘 J.董 B.王 于 2020-03-27 设计创作,主要内容包括:本申请提供了异多聚体蛋白质,诸如双特异性抗体,其包含相对于野生型CH3结构域在氨基酸位置354处具有利用大疏水氨基酸进行的取代的含有第一抗体重链恒定结构域3(CH3)的多肽,和/或包含相对于野生型CH3结构域在氨基酸位置347处利用带负电荷的氨基酸残基进行的取代的含有第二CH3的多肽。还提供了多肽、编码此类多肽的核酸和载体、药物组合物、制备方法和使用异多聚体蛋白质的治疗方法。(The present application provides heteromultimeric proteins, such as bispecific antibodies, comprising a first antibody heavy chain constant domain 3(CH3) -containing polypeptide having a substitution with a large hydrophobic amino acid at amino acid position 354 relative to the wild-type CH3 domain, and/or a second CH 3-containing polypeptide comprising a substitution with a negatively charged amino acid residue at amino acid position 347 relative to the wild-type CH3 domain. Also provided are polypeptides, nucleic acids and vectors encoding such polypeptides, pharmaceutical compositions, methods of preparation, and methods of treatment using heteromultimeric proteins.)

1. A heteromultimeric protein comprising a first polypeptide comprising a first heavy chain constant domain 3(CH3) domain and a second polypeptide comprising a second CH3 domain, wherein the first CH3 domain comprises a substitution with a large hydrophobic amino acid at amino acid position 354 relative to the wild-type CH3 domain and/or the second CH3 domain comprises a substitution with a negatively charged amino acid at amino acid position 347 relative to the wild-type CH3 domain, and wherein the amino acid residue numbering is based on EU numbering.

2. The heteromultimeric protein of claim 1, wherein the large hydrophobic amino acid at amino acid position 354 forms a hydrophobic interaction with an amino acid residue in the second CH3 domain.

3. The heteromultimeric protein of claim 1 or 2, wherein the second CH3 domain comprises a tyrosine (Y) at amino acid position 349.

4. The heteromultimeric protein of any one of claims 1-3, wherein the negatively charged amino acid at amino acid position 347 forms an ionic bond with an amino acid residue in the first CH3 domain.

5. The heteromultimeric protein of any one of claims 1-4, wherein the first CH3 domain comprises a lysine (K) at amino acid position 360.

6. The heteromultimeric protein of any one of claims 1-5, wherein the first CH3 domain and the second CH3 domain are human CH3 domains.

7. The heteromultimeric protein of any one of claims 1-6, wherein the first CH3 domain comprises a substitution selected from the group consisting of: S354Y, S354F and S354W.

8. The heteromultimeric protein of claim 7, wherein the first CH3 domain comprises S354Y.

9. The heteromultimeric protein of any one of claims 1-8, wherein the second CH3 domain comprises a substitution selected from the group consisting of: Q347E and Q347D.

10. The heteromultimeric protein of claim 9, wherein the second CH3 domain comprises Q347E.

11. The heteromultimeric protein of any one of claims 1-10, wherein the first CH3 domain and the second CH3 domain further comprise knob entry hole residues.

12. The heteromultimeric protein of claim 11, wherein the knob entry well residues are T366Y and Y407T.

13. The heteromultimeric protein of claim 12, wherein the first CH3 domain comprises T366Y and S354Y, and the second CH3 domain comprises Y407T and Q347E.

14. The heteromultimeric protein of any one of claims 1-13, wherein the first polypeptide and/or the second polypeptide comprises heavy chain constant domain 2(CH 2).

15. The heteromultimeric protein of claim 14, wherein said heteromultimeric protein comprises an IgG Fc region.

16. The heteromultimeric protein of claim 15, wherein the IgG Fc region is an IgG1, IgG2, IgG3, or IgG4 Fc region.

17. The heteromultimeric protein of any one of claims 1-16, wherein the first polypeptide is an antibody heavy chain and/or the second polypeptide is an antibody heavy chain.

18. The heteromultimeric protein of claim 17, wherein said heteromultimeric protein comprises one or more antibody light chains.

19. The heteromultimeric protein of claim 18, wherein said heteromultimeric protein is a multispecific antibody.

20. The heteromultimeric protein of claim 18 or 19, comprising:

(a) a first heavy chain comprising, from N-terminus to C-terminus: a first heavy chain variable domain (VH1), a first heavy chain constant domain 1(CH1), a first heavy chain constant domain 2(CH2) and the first CH3 domain;

(b) a first light chain comprising, from N-terminus to C-terminus: a first light chain variable domain (VL1) and a first light chain constant domain (CL);

(c) a second heavy chain comprising from N-terminus to C-terminus: a second heavy chain variable domain (VH2), a second CH1, a second CH2 and the second CH3 domain; and

(d) a second light chain comprising, from N-terminus to C-terminus: a second light chain variable domain (VL2) and a second CL;

wherein VH1 associates with VL1 to form a first antigen binding site that specifically binds to a first target, and VH2 associates with VL2 to form a second antigen binding site that specifically binds to a second target.

21. The heteromultimeric protein of claim 20, wherein VL1 and VL2 have the same amino acid sequence.

22. The heteromultimeric protein of claim 20, wherein VL1 and VL2 have different amino acid sequences.

23. The heteromultimeric protein of any one of claims 20-22, wherein the first target and the second target are different epitopes of the same antigen.

24. The heteromultimeric protein of any one of claims 20-22, wherein the first target and the second target are different antigens.

25. The heteromultimeric protein of claim 24, wherein the first antigen-binding site specifically binds a tumor antigen and the second antigen-binding site specifically binds CD 3.

26. The heteromultimeric protein of claim 25, wherein the first antigen binding site specifically binds CD 20.

27. The heteromultimeric protein of claim 25, wherein the first antigen binding site specifically binds HER 2.

28. The heteromultimeric protein of any one of claims 20-27, comprising:

(a) a first heavy chain comprising, from N-terminus to C-terminus: a third triple chain variable domain (VH3), a third CH1, VH1, a first CH1, said first CH2 and said first CH3 domain;

(b) a first light chain comprising, from N-terminus to C-terminus: a third light chain variable domain (VL2), a third CL, the VL1, and the first CL;

wherein VH3 associates with VL3 to form a third antigen binding site that specifically binds to a third target.

29. The heteromultimeric protein of claim 28, wherein the first antigen binding site and the third antigen binding site specifically bind to the same antigen.

30. The heteromultimeric protein of claim 29, wherein the first and third antigen binding sites specifically bind to HER2, and the second antigen binding site specifically binds to CD 3.

31. The heteromultimeric protein of claim 18 or 19, comprising:

(a) a first heavy chain comprising, from N-terminus to C-terminus: a first VHH, a first heavy chain constant domain 2(CH2) and a first CH3 domain;

(b) a second heavy chain comprising from N-terminus to C-terminus: a first heavy chain variable domain (VH1), a second CH1, a second CH2 and the second CH3 domain; and

(d) a light chain comprising, from N-terminus to C-terminus: a first light chain variable domain (VL1) and a first CL;

wherein VH1 associates with VL1 to form a first antigen binding site that specifically binds to a first target, and the first VHH specifically binds to a second target.

32. The heteromultimeric protein of claim 31, wherein the first antigen binding site specifically binds to CD3, and the first VHH specifically binds to a tumor antigen.

33. The heteromultimeric protein of claim 32, wherein the first VHH protein specifically binds to BCMA.

34. The heteromultimeric protein of any one of claims 31-33, wherein the first heavy chain comprises, from N-terminus to C-terminus: a second VHH, the first CH2 and the first CH3 domain, wherein the second VHH domain specifically binds to a third target.

35. The heteromultimeric protein of claim 34, wherein the first VHH and the second VHH specifically bind to the same antigen.

36. The heteromultimeric protein of claim 35, wherein the first VHH and the second VHH specifically bind to BCMA.

37. The heteromultimeric protein of any one of claims 1-16, wherein said heteromultimeric protein is an immunoadhesin or an antibody-immunoadhesin chimera.

38. One or more nucleic acids encoding the heteromultimeric protein of any one of claims 1-37.

39. A vector comprising one or more nucleic acids of claim 38.

40. A host cell comprising one or more nucleic acids of claim 38 or the vector of claim 39.

41. A method for making a multispecific antibody or heteromultimeric protein, the method comprising:

(a) culturing the host cell of claim 40 under conditions that allow expression of one or more nucleic acids or vectors; and

(b) recovering the multispecific antibody or the heteromultimeric protein from the host cell culture.

42. A pharmaceutical composition comprising the heteromultimeric protein of any one of claims 1-37 and a pharmaceutically acceptable excipient.

43. A method for treating a subject in need thereof, the method comprising administering to the subject an effective amount of the pharmaceutical composition of claim 42.

44. A method of generating a heteromultimeric protein that specifically binds to a first target and a second target, the method comprising:

(a) providing a first polypeptide comprising a first binding domain that specifically binds to the first target and a first CH3 domain; and

(b) providing a second polypeptide comprising a second binding domain that specifically binds to the second target and a second CH3 domain;

wherein:

(i) the first CH3 domain comprises a substitution with a large hydrophobic amino acid at amino acid position 354 relative to the wild-type CH3 domain and/or the second CH3 domain comprises a substitution with a negatively charged amino acid at amino acid position 347 relative to the wild-type CH3 domain; or

(ii) The first CH3 domain comprises a substitution with a negatively charged amino acid at amino acid position 347 relative to the wild-type CH3 domain and/or the second CH3 domain comprises a substitution with a large hydrophobic amino acid at amino acid position 354 relative to the wild-type CH3 domain;

and wherein amino acid residue numbering is based on EU numbering.

Technical Field

The present application relates to heteromultimeric proteins, such as bispecific antibodies, compositions, methods of making, and methods of use.

Background

The classical approach to generating bispecific antibodies by co-expressing two different IgG molecules in hybridomas results in up to 10 possible heavy and light chain combinations. This reduces the yield and presents purification challenges. To overcome these challenges, a variety of bispecific antibody formats have been developed that promote heterodimer formation. Many known formats employ single chain variable (scFv) modules or similar structures that rely on engineered linkers to force the antigen-binding components into the desired configuration. However, many of these bispecific antibody formats suffer from unfavorable properties compared to the native antibody, including ease of aggregation, difficulty of production, short serum half-life, and potential immunogenicity.

Bispecific antibody designs have been developed in several native antibody formats (i.e., antibodies consisting of two light chains and two heavy chains). For example, the heavy chain Fc-Fc interface can be engineered with interacting amino acid pairs, such as knob-into-holes (KIH) residues, disulfide-forming cysteines, or residues with opposite electrostatic charges, to actively promote heterodimer formation from different heavy chains when they are co-expressed. However, the classical KIH strategy still results in significant homodimer formation and low yields of bispecific antibodies.

The disclosures of all publications, patents, patent applications, and published patent applications mentioned herein are hereby incorporated by reference in their entirety.

Disclosure of Invention

Heteromultimeric proteins, such as Fc-containing heterodimeric proteins, multispecific antibodies, and multispecific immunoadhesins, methods of making and methods of using the same are provided.

One aspect of the present application provides a heteromultimeric (e.g., heterodimeric) protein comprising a first polypeptide comprising a first heavy chain constant domain 3(CH3) domain and a second polypeptide comprising a second CH3 domain, wherein the first CH3 domain comprises a substitution with a large hydrophobic amino acid at amino acid position 354 relative to the wild-type CH3 domain, and/or the second CH3 domain comprises a substitution with a negatively charged amino acid at amino acid position 347 relative to the wild-type CH3 domain, and wherein the amino acid residue numbering is based on EU numbering. In some embodiments, the large hydrophobic amino acid at amino acid position 354 forms a hydrophobic interaction with an amino acid residue in the second CH3 domain. In some embodiments, the second CH3 domain comprises a large hydrophobic residue at amino acid position 349 (e.g., Y349). In some embodiments, the negatively charged amino acid at amino acid position 347 forms an ionic bond with an amino acid residue in the first CH3 domain. In some embodiments, the first CH3 domain comprises a positively charged residue at amino acid position 360 (e.g., K360). In some embodiments, the first CH3 domain and the second CH3 domain are human CH3 domains. In some embodiments, the first CH3 domain comprises a substitution selected from the group consisting of: S354Y, S354F and S354W. In some embodiments, the first CH3 domain comprises S354Y. In some embodiments, the second CH3 domain does not comprise a compensatory substitution (e.g., substitution at Y349) for the substitution of S354 in the first CH3 domain. In some embodiments, the second CH3 domain comprises a substitution selected from the group consisting of: Q347E and Q347D. In some embodiments, the second CH3 domain comprises Q347E.

In some embodiments of any of the heteromultimeric proteins described above, the first CH3 domain and the second CH3 domain further comprise a knob-hole (KIH) residue. In some embodiments, the buttonhole residues are T366Y and Y407T. In some embodiments, the first CH3 domain comprises T366Y and S354Y, and the second CH3 domain comprises Y407T and Q347E. In some embodiments, the first CH3 domain comprises Y407T and S354Y, and the second CH3 domain comprises T366Y and Q347E. And is

In some embodiments of any of the heteromultimeric proteins described above, the first polypeptide and/or the second polypeptide comprises heavy chain constant domain 2(CH 2). In some embodiments, the heteromultimeric protein comprises an IgG Fc region. In some embodiments, the IgG Fc region is an IgG1, IgG2, IgG3, or IgG4 Fc region. In some embodiments, the first polypeptide is an antibody heavy chain and/or the second polypeptide is an antibody heavy chain. In some embodiments, the heteromultimeric protein comprises one or more antibody light chains.

In some embodiments according to any of the above heteromultimeric proteins, the heteromultimeric protein is a multispecific (e.g., bispecific) antibody.

In some embodiments of any of the heteromultimeric proteins described above, the heteromultimeric protein comprises: (a) a first heavy chain comprising, from N-terminus to C-terminus: a first heavy chain variable domain (VH1), a first heavy chain constant domain 1(CH1), a first heavy chain constant domain 2(CH2) and a first CH3 domain; (b) a first light chain comprising, from N-terminus to C-terminus: a first light chain variable domain (VL1) and a first light chain constant domain (CL); (c) a second heavy chain comprising from N-terminus to C-terminus: a second heavy chain variable domain (VH2), a second CH1, a second CH2 and a second CH3 domain; and (d) a second light chain comprising from N-terminus to C-terminus: a second light chain variable domain (VL2) and a second CL; wherein VH1 associates with VL1 to form a first antigen binding site that specifically binds to a first target and VH2 binds with VL2 to form a second antigen binding site that specifically binds to a second target. In some embodiments, VL1 and VL2 have the same amino acid sequence. In some embodiments, VL1 and VL2 have different amino acid sequences. In some embodiments, the first target and the second target are the same epitope. In some embodiments, the first target and the second target are different epitopes of the same antigen. In some embodiments, the first target and the second target are different antigens. In some embodiments, the first antigen-binding site specifically binds to a tumor antigen and the second antigen-binding site specifically binds to CD3, or the first antigen-binding site specifically binds to CD3 and the second antigen-binding site specifically binds to a tumor antigen. In some embodiments, the first antigen-binding site specifically binds CD20 and the second antigen-binding site specifically binds CD3, or the first antigen-binding site specifically binds CD3 and the second antigen-binding site specifically binds CD 20. In some embodiments, the first antigen binding site specifically binds to HER2 and the second antigen binding site specifically binds to CD3, or the first antigen binding site specifically binds to CD3 and the second antigen binding site specifically binds to HER 2. In some embodiments, the heteromultimeric protein comprises: (a) a first heavy chain comprising, from N-terminus to C-terminus: a third triple chain variable domain (VH3), a third CH1, VH1, a first CH1, a first CH2 and a first CH3 domain; (b) a first light chain comprising, from N-terminus to C-terminus: a third light chain variable domain (VL2), a third CL, VL1, and a first CL; wherein VH3 associates with VL3 to form a third antigen binding site that specifically binds a third target. In some embodiments, the first antigen binding site and the third antigen binding site specifically bind to the same antigen. In some embodiments, the first antigen binding site and the third antigen binding site specifically bind HER2, and the second antigen binding site specifically binds CD 3.

In some embodiments of any of the heteromultimeric proteins described above, the heteromultimeric protein comprises: (a) a first heavy chain comprising, from N-terminus to C-terminus: a first VHH, a first heavy chain constant domain 2(CH2) and a first CH3 region; (b) a second heavy chain comprising from N-terminus to C-terminus: a first heavy chain variable domain (VH1), a second CH1, a second CH2 and a second CH3 domain; and (d) a light chain comprising from N-terminus to C-terminus: a first light chain variable domain (VL1) and a first CL; wherein VH1 associates with VL1 to form a first antigen binding site that specifically binds to a first target, and a first VHH that specifically binds to a second target. In some embodiments, the first antigen binding site specifically binds CD3 and the first VHH specifically binds a tumor antigen, or the first antigen binding site specifically binds a tumor antigen and the first VHH specifically binds CD 3. In some embodiments, the first VHH specifically binds BCMA. In some embodiments, the first heavy chain comprises, from N-terminus to C-terminus: a second VHH, a first CH2, and a first CH3 domain, wherein the second VHH specifically binds a third target. In some embodiments, the first VHH and the second VHH specifically bind to the same antigen. In some embodiments, the first VHH and the second VHH specifically bind BCMA.

In some embodiments according to any of the above heteromultimeric proteins, the heteromultimeric protein is an immunoadhesin or an antibody-immunoadhesin chimera.

Another aspect of the application provides a polypeptide comprising an antibody CH3 domain, wherein the CH3 domain comprises a substitution with a large hydrophobic amino acid at amino acid position 354 relative to the wild-type CH3 domain and/or a substitution with a negatively charged amino acid at amino acid position 347 relative to the wild-type CH3 domain, and wherein the polypeptide has a reduced ability to form a homodimer as compared to a polypeptide comprising the wild-type CH3 domain. In some embodiments, the CH3 domain is a human CH3 domain. In some embodiments, the CH3 domain comprises a substitution selected from the group consisting of: S354Y, S354F and S354W. In some embodiments, the CH3 domain comprises S354Y. In some embodiments, the CH3 domain comprises a substitution selected from the group consisting of: Q347E and Q347D. In some embodiments, the CH3 domain comprises Q347E. In some embodiments, the CH3 domain further comprises knob entry residues, such as T366Y or S407T. In some embodiments, the polypeptide further comprises heavy chain constant domain 2(CH 2). In some embodiments, the polypeptide comprises an antibody heavy chain.

In some embodiments, polypeptides comprising the CH3 domain of any one of SEQ ID NOs 1-4 are provided. In some embodiments, polypeptides comprising the amino acid sequence of any one of SEQ ID NOs 1-4 are provided.

In some embodiments, an antibody (e.g., a bispecific antibody) comprising a polypeptide according to any of the above is provided.

Another aspect of the present application provides a method of producing a heteromultimeric protein that specifically binds a first target and a second target, the method comprising: (a) providing a first polypeptide comprising a first binding domain that specifically binds to a first target and a first CH3 domain; and (b) providing a second polypeptide comprising a second binding domain that specifically binds to a second target and a second CH3 domain; wherein: (i) the first CH3 domain comprises a substitution with a large hydrophobic amino acid at amino acid position 354 relative to the wild-type CH3 domain and/or the second CH3 domain comprises a substitution with a negatively charged amino acid at amino acid position 347 relative to the wild-type CH3 domain; or (ii) the first CH3 domain comprises a substitution with a negatively charged amino acid at amino acid position 347 relative to the wild-type CH3 domain and/or the second CH3 domain comprises a substitution with a large hydrophobic amino acid at amino acid position 354 relative to the wild-type CH3 domain; and wherein amino acid residue numbering is based on EU numbering. In some embodiments, the large hydrophobic amino acid at amino acid position 354 in one CH3 domain forms a hydrophobic interaction with an amino acid residue in another CH3 domain. In some embodiments, another CH3 domain comprises a large hydrophobic residue at amino acid position 349 (e.g., Y349). In some embodiments, the negatively charged amino acid at amino acid position 347 in one CH3 domain forms an ionic bond with an amino acid residue in another CH3 domain. In some embodiments, another CH3 domain comprises a positively charged residue at amino acid position 360 (e.g., K360). In some embodiments, the first CH3 domain and the second CH3 domain are human CH3 domains. In some embodiments, the first or second CH3 domain comprises a substitution selected from the group consisting of: S354Y, S354F and S354W. In some embodiments, the first or second CH3 domain comprises S354Y. In some embodiments, the first or second CH3 domain comprises a substitution selected from the group consisting of: Q347E and Q347D. In some embodiments, the first or second CH3 domain comprises Q347E. In some embodiments, the first CH3 domain and the second CH3 domain further comprise KIH residues, such as T366Y and Y407T. In some embodiments, the first CH3 domain comprises T366Y and S354Y, and the second CH3 domain comprises Y407T and Q347E. In some embodiments, the first CH3 domain comprises Y407T and S354Y, and the second CH3 domain comprises T366Y and Q347E. And is

In some embodiments, a heteromultimeric protein prepared using any one of the methods described above is provided.

One aspect of the present application provides one or more nucleic acids encoding a heteromultimeric protein according to any one of the heteromultimeric proteins described above or a polypeptide according to any one of the polypeptides described above. In some embodiments, there is provided a vector comprising one or more nucleic acids according to any one of the above. In some embodiments, there is provided a host cell comprising one or more nucleic acids according to any one of the above nucleic acids or a vector according to any one of the above vectors.

One aspect of the present application provides a method for making a multispecific (e.g., bispecific) antibody or heteromultimeric (e.g., heterodimeric) protein, the method comprising: (a) culturing a host cell according to any one of the above host cells under conditions that allow expression of one or more nucleic acids or vectors; and (b) recovering the multispecific antibody or heteromultimeric protein from the host cell culture.

Another aspect of the application provides a pharmaceutical composition comprising a heteromultimeric protein according to any of the heteromultimeric proteins described above or an antibody according to any of the antibodies described above, and a pharmaceutically acceptable excipient. In some embodiments, there is provided a method for treating a subject in need thereof, the method comprising administering to the subject an effective amount of a pharmaceutical composition according to any one of the pharmaceutical compositions described above.

Also provided are kits and articles of manufacture comprising any of the heteromultimeric proteins described above (e.g., bispecific antibodies) or for use in any of the methods described above.

Brief Description of Drawings

Figure 1 shows the schematic structure of two bispecific antibody constructs. In some embodiments, the first heavy chain of the bispecific antibody construct has an engineered residue, such as Q347E, that forms an ionic bond with a native residue in the second heavy chain (e.g., K360); and the second heavy chain has an engineered residue, such as S354Y, that forms a hydrophobic interaction with a native residue in the first heavy chain (e.g., Y349). The constructs on the left (S1) have a common light chain and the construct on the right (S2) has two different light chains. The engineered ionic bonds and hydrophobic interactions described herein can be combined with conventional knob entry hole (KIH) mutations to promote heterodimer formation.

Figure 2A shows a partial view of the crystal structure of the CH3-CH3 interface of two heavy chains in an antibody. S354 in the first heavy chain (i.e., S375 in the figure, numbered according to Kabat) does not form any contact with a residue in the second heavy chain.

Figure 2B shows a partial view of the simulated crystal structure of the CH3-CH3 interface of a heterodimeric Fc with the S354Y mutation in the first heavy chain (i.e., Y375 according to Kabat numbering in the figure). S354Y forms a hydrophobic interaction with Y349 in the second heavy chain (i.e. Y370 in the figure according to Kabat numbering).

Figure 2C shows a partial view of the simulated crystal structure of the CH3-CH3 interface of a heterodimeric Fc with a Q347E mutation in the first heavy chain (i.e., E368 according to Kabat numbering in the figure). Q347E forms an ionic bond with K360 in the second heavy chain (i.e. K383 in the figure according to Kabat numbering). Q347 may or may not form very weak hydrogen bonds with K360.

Figure 3 shows a schematic workflow for the production and quality control of CD20/CD3 bispecific antibody.

FIG. 4 shows the chromatogram of the CD20/CD3 antibody on a MonoQ column after MonoQ purification. V2 is a common light chain bispecific antibody with KIH (i.e., T366Y and Y407T) residues. V4b is a common light chain bispecific antibody with KIH residues identical to V2, Q347E and S354Y mutations in both heavy chains, respectively.

FIG. 5A shows the chromatogram of the CD20/CD 3V 4b bispecific antibody on a MonoQ column after protein A purification. FIG. 5B shows SDS gels and protein concentrations of MonoQ purified fractions. Fig. 5C shows the results of T cell activation assay of the MonoQ purified fraction. FIG. 5D shows the results of capillary electrophoresis of sodium dodecyl sulfate (CE-SDS) under reducing conditions for purified CD20/CD 3V 4b bispecific antibody. FIG. 5E shows the results of CE-SDS under non-reducing conditions for the purified CD20/CD 3V 4b bispecific antibody. FIG. 5F shows the results of Differential Scanning Fluorometry (DSF) and Static Light Scattering (SLS) of purified CD20/CD 3V 4b bispecific antibody.

FIG. 6 shows chromatograms of four batches of CD20/CD 3V 4b bispecific antibody on a MonoQ column after protein A purification.

FIG. 7 shows sequence alignments of IgG constant region sequences from different species, human IgG1(SEQ ID NO:5), human IgG2(SEQ ID NO:6), human IgG3(SEQ ID NO:7), human IgG4(SEQ ID NO:8), murine IgG1(SEQ ID NO:9), murine IgG2a (SEQ ID NO:10), murine IgG3(SEQ ID NO:11), rat IgG1(SEQ ID NO:12), rat IgG2a (SEQ ID NO:13), rat IgG (SEQ ID NO:14), bovine IgG1(SEQ ID NO:15), bovine IgG2(SEQ ID NO:16), feline IgG (SEQ ID NO:17), and canine IgG (SEQ ID NO: 18). Amino acid residues 347, 349, 354, and 360 are highly conserved among IgG molecules of various species.

Figure 8 shows a schematic of a Her2-B3/CD3 bispecific antibody comprising one anti-Her 2Fab, one anti-CD 3 Fab and one Fc domain.

FIG. 9 shows the purification of Her2-B3/CD3 bispecific antibody by cation exchange Chromatography (CEX). Retention time is shown in minutes on the x-axis and relative protein abundance is shown in milliabsorbance units (mAU) on the y-axis. Individual peak fractions (i.e., A1-A15 and B15-B6) were labeled.

FIG. 10 shows sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) of fractions of Her2-B3/CD3 bispecific antibody purified by CEX, as shown in FIG. 9. Lanes 1-8 show the fraction of the main peak labeled A1-A8, lanes 9 and 10 show the 2 nd and 3 rd fractions of the small peaks labeled A14 and B7, and lane 11 shows the Her2-B3/CD3 bispecific antibody after protein A purification. Lane M shows the protein marker ladder, with the Kilodalton (KD) mass of the protein marker shown on the right.

Figure 11 shows a schematic of a Her2-B3-V3/CD3 bispecific antibody comprising two anti-Her 2fabs, one anti-CD 3 Fab and one Fc domain, as described above, with Her2-B3-V3/CD3 bispecific antibody.

FIG. 12 shows CEX purification of Her2-B3-V3/CD3 bispecific antibody. The sample was previously purified on a protein a column. Retention time is shown in minutes on the x-axis and relative protein abundance is shown in milliabsorbance units (mAU) on the y-axis. Individual peak fractions were labeled.

FIG. 13 shows SDS-PAGE of the Her2-B3-V3/CD3 bispecific antibody fraction after CEX purification. Lane 1 shows Her2-B3-V3/CD3 expressed in the supernatant. Lanes 2-6 show purified fractions A3-A7 (non-reduced) (see FIG. 12). Lanes 7-11 show purified fractions A3-A7 (reduced) (see FIG. 12). Lane M shows the protein marker ladder and the Kilodalton (KD) mass of the protein marker is shown on the left. The position of the assembled Her2/CD3 bispecific antibody ("Her 2/CD3 BsAb"), as well as the individual components (i.e., anti-Her 2 heavy chain with two Fab "Her 2(2Fab) HCs", anti-CD 3 heavy chain "CD 3 HC" and light chain "common LC") are shown on the right.

FIG. 14 shows a schematic representation of a BCMA-Fc/CD3 bispecific antibody. On the left is BCMA-3E5/CD3, which consists of a BCMA-3E5 VHH domain, a CD3 Fab and an Fc domain. The right one is BCMA-3E1B2/CD3, which comprises two BCMA VHH domains (3E1 and 3B2), one CD3 Fab and one Fc domain.

FIG. 15 shows SDS-PAGE of protein A purified BCMA-3E1B2/CD3 and BCMA-3E5/CD 3. Lane 1 shows non-reduced BCMA-3E1B2/CD3, lane 2 shows non-reduced BCMA-3E5/CD3, lane 3 shows reduced BCMA-3E1B2/CD3, and lane 4 shows reduced BCMA-3E5/CD 3. Lane M shows the protein marker ladder and the Kilodalton (KD) mass of the protein marker is shown on the left. The position of the BCMA-Fc/CD3 bispecific antibody component is shown on the right.

Figure 16 shows a schematic structure of a multispecific antibody construct with engineered ionic bonds and hydrophobic interactions in the Fc region. The bispecific antibody formats are shown in the top row, and the trispecific and tetraspecific antibody formats are shown in the bottom row. As described above, the oval structures were obtained from llama VHH libraries.

Detailed Description

The present application provides heteromultimeric (e.g., heterodimeric) proteins comprising an antibody heavy chain constant domain 3(CH3) domain with novel engineered ionic bonds and/or hydrophobic interactions, which antibody heavy chain constant domain 3 can be combined with knob-entry hole (KIH) residues, facilitating the formation of heterodimers. Methods of making and using the heteromultimeric proteins are also provided. The preparation methods described herein can be used to enhance heterodimer formation and hinder homodimer formation when two different CH 3-containing polypeptides are co-expressed. The CH 3-based heteromultimeric protein strategies described herein are applicable to all Fc-containing heteromultimeric proteins, such as bispecific antibodies and bispecific immunoadhesins.

Accordingly, one aspect of the present application provides a heteromultimeric protein comprising a first polypeptide comprising a first CH3 domain and a second polypeptide comprising a second CH3 domain, wherein the first CH3 domain comprises a substitution at amino acid position 354 with a large hydrophobic amino acid (e.g., S354Y) relative to the wild-type CH3 domain, and/or the second CH3 domain comprises a substitution at amino acid position 347 with a negatively charged amino acid (e.g., Q347E) relative to the wild-type CH3 domain, and wherein the amino acid residue numbering is based on EU numbering.

Compositions (such as pharmaceutical compositions), methods of preparation, methods of treatment, kits and articles of manufacture are also provided.

I. Definition of

As used herein, a "heteromultimer" or "heteromultimeric protein" is a molecule comprising at least a first polypeptide and a second polypeptide, wherein the second polypeptide differs in amino acid sequence from the first polypeptide by at least one amino acid residue. In some embodiments, the heteromultimer has binding specificity for at least two different ligands or binding sites. The heteromultimer may comprise a "heterodimer" formed by the first polypeptide and the second polypeptide, or may form a higher order tertiary structure in which polypeptides other than the first polypeptide and the second polypeptide are present.

As used herein, "binding domain" includes any region of a polypeptide that is responsible for specifically binding a target molecule (e.g., an antigen, ligand, receptor, substrate, or inhibitor). Exemplary binding domains include antibody variable domains, receptor binding domains, ligand binding domains, and enzymatic domains.

As used herein, the terms "specific binding," "specific recognition," or "specific for" refer to a measurable and reproducible interaction, such as binding between a target and an antibody, which determines the presence of the target in the presence of a heterogeneous population of molecules, including biomolecules. For example, an antibody that specifically recognizes a target (which may be an epitope) is an antibody that binds the target with greater affinity, avidity, more readily, and/or for a longer duration than it binds to other targets. In some embodiments, an antibody that specifically recognizes an antigen reacts with one or more antigenic determinants of the antigen with a binding affinity that is at least about 10-fold greater than its binding affinity to other targets.

The term "antibody" is used herein in the broadest sense and includes full-length antibodies and antigen-binding fragments thereof. The term "antibody" includes monoclonal antibodies (including full-length 4 chain antibodies or full-length heavy chain-only antibodies with immunoglobulin Fc regions), antibody compositions with polyepitopic specificity, multispecific antibodies (e.g., bispecific antibodies, diabodies, and single chain molecules), and antibody fragments (e.g., Fab, F (ab')2And Fv). Antibodies contemplated herein include single domain antibodies, such as heavy chain-only antibodies.

Full-length four-chain antibodies comprise two heavy chains and two light chains. The variable regions of the light and heavy chains are responsible for antigen binding. The variable regions in both chains typically comprise three highly variable loops called Complementarity Determining Regions (CDRs) (light chain (LC) CDRs, including LC-CDR1, LC-CDR2, and LC-CDR3, and Heavy Chain (HC) CDRs, including HC-CDR1, HC-CDR2, and HC-CDR 3). CDR boundaries of the antibodies and antigen-binding fragments disclosed herein can be defined or identified by the Kabat, Chothia, or Al-Lazikani convention (Al-Lazikani 1997; Chothia 1985; Chothia 1987; Chothia 1989; Kabat 1987; Kabat 1991). The three CDRs of the heavy or light chain are interleaved between flanking segments called Framework Regions (FRs) that are more highly conserved than the CDRs and form a scaffold that supports hypervariable loops. The constant regions of the heavy and light chains are not involved in antigen binding, but exhibit various effector functions. Antibodies are classified according to the amino acid sequence of their heavy chain constant region. The five major classes or isotypes of antibodies are IgA, IgD, IgE, IgG and IgM, which are characterized by the presence of alpha, delta, epsilon, gamma and mu heavy chains, respectively. Several major antibody classes are divided into subclasses, such as IgG1(γ 1 heavy chain), IgG2(γ 2 heavy chain), IgG3(γ 3 heavy chain), IgG4(γ 4 heavy chain), IgA1(α 1 heavy chain), or IgA2(α 2 heavy chain).

The "variable region" or "variable domain" of an antibody refers to the amino-terminal domain of a heavy or light chain of the antibody. The variable domains of the heavy and light chains may be referred to as "V" respectivelyH"and" VL". These domains are usually the most variable part of an antibody (relative to other antibodies of the same class) and contain an antigen binding site. Heavy chain-only antibodies from camelid species have a single heavy chain variable region, which is referred to as a "VHH". Thus, VHH is a special type of VH

The term "heavy chain-only antibody" or "HCAb" refers to a functional antibody that comprises a heavy chain but lacks the light chain typically found in a 4-chain antibody. Camelids (such as camels, llamas or alpacas) are known to produce hcabs.

The term "single domain antibody" or "sdAb" refers to a single antigen-binding polypeptide having three Complementarity Determining Regions (CDRs). The sdAb alone is capable of binding to an antigen without pairing with a corresponding CDR-containing polypeptide. In some cases, single domain antibodies are engineered from camelid hcabs, and their heavy chain variable domains are referred to herein as "VHHs" (variable domains of the heavy chain of heavy chain antibodies). Camel sdabs are one of the smallest known antigen-binding antibody fragments (see, e.g., Hamers-Casterman et al, Nature 363:446-8 (1993); Greenberg et al, Nature 374:168-73 (1995); Hassanzadeh-Ghassabeh et al, nanomedicine (Lond),8:1013-26 (2013)). Basic VHH has the following structure from N-terminus to C-terminus: FR1-CDR1-FR2-CDR2-FR3-CDR3-FR4, wherein FR1 to FR4 refer to framework regions 1 to 4, respectively, and wherein CDR1 to CDR3 refer to complementarity determining regions 1 to 3.

As used herein, the term "antigen-binding fragment" refers to antibody fragments including, for example, diabodies, fabs, Fab ', F (ab ') 2, Fv fragments, disulfide-stabilized Fv fragments (dsFv), (dsFv)2, bispecific dsFv (dsFv-dsFv '), disulfide-stabilized diabodies (ds diabodies), single-chain Fv (scFv), scFv dimers (bivalent diabodies); single domain antibodies (such as V)HH) And multispecific antibodies formed from antibody portions comprising one or more CDRs, or any other antibody fragment that binds an antigen but does not comprise an intact antibody structure. The antigen-binding fragment is capable of binding to the same antigen to which the parent antibody or parent antibody fragment (e.g., parent scFv) binds. In some embodiments, an antigen-binding fragment can comprise one or more CDRs from a particular human antibody grafted to a framework region from one or more different human antibodies. Papain digestion of antibodies produces two identical antigen-binding fragments (called "Fab" fragments) and a residual "Fc" fragment (the name reflects the ability to crystallize readily). Fab fragments consist of the entire variable domains of the L and H chains (V)H) And the first constant domain of a heavy chain (C)H1) And (4) forming. For antigen binding, each Fab fragment is monovalent, i.e., it has only a single antigen binding site. Pepsin treatment of antibodies produced a single large F (ab')2Fragments which correspond approximately to two disulfide-linked Fab fragments with different antigen binding activity and which are still capable of cross-linking the antigen. Fab' fragments differ from Fab fragments in that they are at CHThe carboxy terminus of domain 1 has some additional residues, including one or more cysteines from the antibody hinge region. Fab '-SH is herein the name for Fab',wherein one or more cysteine residues of the constant domain have a free thiol group. F (ab')2Antibody fragments were originally produced as pairs of Fab' fragments with hinge cysteines between them. Other chemical couplings of antibody fragments are also known.

The term "constant domain" refers to a portion of an immunoglobulin molecule that has a more conserved amino acid sequence relative to another portion of the immunoglobulin (the variable domain) that comprises the antigen binding site. Constant Domain comprising heavy chain CH1、CH2 and CH3 Domain (collectively referred to as C)H) And CHL (or C) of light chainL) A domain.

The "light chains" of antibodies (immunoglobulins) from any mammalian species can be assigned to one of two distinctly different classes, termed kappa ("κ") and lambda ("λ"), based on the amino acid sequence of their constant domains.

"Fv" is the smallest antibody fragment that contains the entire antigen recognition and binding site. The fragment consists of a dimer of one heavy chain variable region domain and one light chain variable region domain in close, non-covalent association. From the folding of these two domains, six hypervariable loops (3 loops for each of the H and L chains) are generated, which provide amino acid residues for antigen binding and confer antigen-binding specificity to the antibody. However, even a single variable domain (or half of an Fv comprising only three CDRs specific for an antigen) has the ability to recognize and bind antigen, although with less avidity than the entire binding site.

"Single-chain Fv", also abbreviated as "sFv" or "scFv", is a polypeptide comprising V joined into a single polypeptide chainHAnd VLAntibody fragments of antibody domains. Preferably, the scFv polypeptide is further comprised at VHDomains with VLA polypeptide linker between the domains, which enables the scFv to form the structure required for antigen binding. For a review of scFv, see Pluckthun in The Pharmacology of Monoclonal Antibodies, Vol.113, edited by Rosenburg and Moore, Springer-Verlag, New York, pp.269-315 (1994).

The term "diabodies" refers to diabodies that are constructed at VHDomains with VLSmall antibody fragments made from sFv fragments (see preceding paragraph) with short linkers (about 5-10 residues) between the domains that allow inter-chain pairing rather than intra-chain pairing of the V domains, resulting in a bivalent fragment, i.e., a fragment with two antigen binding sites. Bispecific diabodies are heterodimers of two "cross" sFv fragments, where the V of both antibodiesHDomains and VLThe domains are present on different polypeptide chains. Diabodies are described in more detail, for example, in EP 404,097, WO 93/11161, Hollinger et al, Proc. Natl. Acad. Sci. USA90: 6444-.

The term "specificity" refers to the selective recognition of a particular epitope of an antigen by an antigen binding protein. For example, natural antibodies are monospecific. As used herein, the term "multispecific" means that an antigen binding protein has polyepitopic specificity (i.e., has two, three, or more antigen binding sites capable of specifically binding to two, three, or more different epitopes on one biomolecule, or capable of specifically binding to epitopes on two, three, or more different biomolecules). As used herein, "bispecific" means that the antigen binding protein has two different antigen binding specificities. Unless otherwise indicated, the order of antigens bound by the listed bispecific antibodies is arbitrary.

The term "chimeric antibody" refers to antibodies in which a portion of the heavy and/or light chain is identical or homologous to corresponding sequences in antibodies derived from a particular species or belonging to a particular antibody class or subclass, while the remainder of one or more chains are identical or homologous to corresponding sequences in antibodies derived from another species or belonging to another antibody class or subclass, so long as they exhibit the biological activity of the present invention (see U.S. Pat. No. 4,816,567; and Morrison et al, Proc. Natl. Acad. Sci. USA,81:6851-6855(1984)), as well as fragments of such antibodies.

A "humanized" form of a non-human (e.g., rodent) antibody is a chimeric antibody containing minimal sequences derived from a non-human antibody. For the most part, humanized antibodies are human immunoglobulins (recipient antibody) in which residues from a hypervariable region (HVR) of the recipient are replaced by residues from a hypervariable region of a non-human species (donor antibody), such as mouse, rat, rabbit or nonhuman primate having the desired antibody specificity, affinity, and capacity. In some cases, Framework Region (FR) residues of the human immunoglobulin are replaced with corresponding non-human residues. In addition, humanized antibodies may comprise residues that are not present in the recipient antibody or in the donor antibody. These modifications were made to further improve antibody performance. In general, a humanized antibody will comprise substantially all of at least one and typically two variable domains, in which all or substantially all of the hypervariable loops correspond to those of a non-human immunoglobulin and all or substantially all of the FRs are those of a human immunoglobulin sequence. The humanized antibody will also optionally comprise at least a portion of an immunoglobulin constant region (Fc), typically that of a human immunoglobulin. For further details, see Jones et al, Nature 321:522-525 (1986); riechmann et al, Nature 332: 323-E329 (1988); and Presta, curr, Op, Structure, biol.2:593-596 (1992).

A "multispecific antibody" is a molecule having binding specificity for at least two different antigens or epitopes. "multispecific antibodies" encompass bispecific antibodies ("BsAbs") that bind two different antigens or epitopes, as well as antibodies with more than two specificities, such as trispecific antibodies.

As used herein, the term "immunoadhesin" refers to antibody-like molecules that combine the binding domain of a heterologous protein (an "adhesion", e.g., a receptor, ligand or enzyme) with the effector function of an immunoglobulin constant domain. Structurally, immunoadhesins comprise fusions of the amino acid sequence of an adhesin with the desired binding specificity, which is different from the antigen-binding site of an antibody, and the sequence of an immunoglobulin constant domain. The immunoglobulin constant domain sequence in the immunoadhesin may be obtained from any immunoglobulin, such as IgG1, IgG2, IgG3 or IgG4 subtype, IgA, IgE, IgD or IgM.

As used herein, the phrase "multispecific immunoadhesin" refers to an immunoadhesin having at least two binding specificities (i.e., combining two or more adhesin binding domains). Multispecific immunoadhesins can assemble into heterodimers, heterotrimers, or heterotetramers, for example, as disclosed in WO 89/02922, EP314,317, and US5,116,964. In some embodiments, the multispecific immunoadhesin is bispecific.

An "antibody-immunoadhesin chimera" comprises a molecule that combines at least one binding domain of an antibody with at least one immunoadhesin.

The "CH 1 domain" of the human IgG Fc region (also referred to as "C1" of the "H1" domain) typically extends from about amino acid 118 to about amino acid 215(EU numbering system).

A "hinge region" is generally defined as extending from Glu216 to Pro230 of human IgG1 (Burton, molecular. Immunol.22:161-206 (1985)). The hinge region of other IgG isotypes can be aligned to the IgG1 sequence by placing the first and last cysteine residues that form the S-S bond between heavy chains at the same position.

The "CH 2 domain" of the human IgG Fc region (also referred to as "C2" of the "H2" domain) typically extends from about amino acid 231 to about amino acid 340. The CH2 domain is unique in that it is not closely paired with another domain. In contrast, two N-linked branched carbohydrate chains are interposed between the two CH2 domains of an intact native IgG molecule. It is speculated that carbohydrates may provide an alternative to domain-domain pairings and help stabilize the CH2 domain. Burton, Molec Immunol.22:161-206 (1985).

The "CH 3 domain" (also referred to as the "C2" or "H3" domain) comprises an extension of residues C-terminal to the CH2 domain in the Fc region (i.e., from about amino acid residue 341 to C-terminal of the antibody sequence (typically at amino acid residue 446 or 447 of an IgG)).

The term "knob-entry hole" or "KIH" refers to a pair of engineered amino acid residues in the CH3 domain that result in a spatial modification of the contact surface of the first CH3 domain, which first CH3 domain is preferentially attached to the corresponding contact surface of the second CH3 domain by a complementary spatial modification. Such steric modifications are derived primarily from different amino acid residues and side chains, e.g., to create "knob" or "pore" structures that complement each other to form a "knob-entry" dimer. See, for example, Ridgway, J.B., L.G.Presta et al (1996) "Knobs-int-holes' engineering of antibodies CH3 domains for latent channel synthesis" "Protein Eng 9(7): 617-21; atwell, S., J.B.Ridgway et al (1997), "Stable peripherals from the domain interface of a host using a phase display library," J Mol Biol 270(1): 26-35; merchant, A.M., Z.Zhu et al (1998). "An effective route to human biospecific IgG." Nat Biotechnol 16(7): 677-81; carter, P. (2001). "Bispecific human IgG by design." J immunological Methods 248(1-2): 7-15; and U.S. Pat. nos. 5,731,168 and 7,183,076, which are incorporated herein by reference.

An "isolated" heteromultimer refers to a heteromultimer that has been identified and isolated and/or recovered from a component of its native cell culture environment. Contaminant components of their natural environment are substances that would interfere with diagnostic or therapeutic uses of the heteromultimer and may include enzymes, hormones, and other proteinaceous or nonproteinaceous solutes. The heteromultimer of the invention is typically purified to substantial homogeneity. The phrases "substantially homogeneous," "substantially homogeneous form," and "substantially homogeneous" are used to indicate that the product is substantially free of by-products derived from undesired combinations of polypeptides (e.g., homomultimers). By substantially homogeneous, in terms of purity, it is meant that the amount of by-products does not exceed about 10%, 5%, 1%, 0.5% or less, where the percentages are by weight.

As used herein, the term "isolated nucleic acid" is intended to refer to a nucleic acid of genomic, cDNA, or synthetic origin, or some combination thereof, from which the "isolated nucleic acid" (1) does not bind to all or a portion of the polynucleotide to which it is naturally occurring, (2) is operably linked to a polynucleotide to which it is not naturally linked, or (3) does not naturally occur as part of a larger sequence.

Unless otherwise indicated, "nucleotide sequences encoding amino acid sequences" includes all nucleotide sequences that are degenerate versions of each other and that encode the same amino acid sequence. The phrase nucleotide sequence encoding a protein or RNA may also include introns to the extent that the nucleotide sequence encoding a protein may contain one or more introns in some forms.

The term "operably linked" refers to a functional linkage between a regulatory sequence and a heterologous nucleic acid sequence that results in the expression of the latter. For example, a first nucleic acid sequence is operably linked to a second nucleic acid sequence when the first nucleic acid sequence is in a functional relationship with the second nucleic acid sequence. For example, a promoter is operably linked to a coding sequence if it affects the transcription or expression of the coding sequence. Generally, operably linked DNA sequences are contiguous and, where necessary, join two protein coding regions in the same reading frame.

As used herein, "treatment" or "treating" is a method of obtaining beneficial or desired results, including clinical results. For purposes of the present invention, beneficial or desired clinical results include, but are not limited to, one or more of the following: alleviating one or more symptoms caused by the disease, reducing the extent of the disease, stabilizing the disease (e.g., preventing or delaying the worsening of the disease), preventing or delaying the spread of the disease, preventing or delaying the recurrence of the disease, delaying or slowing the progression of the disease, ameliorating the disease state, providing remission (partial or complete) of the disease, reducing the dose of one or more other drugs required to treat the disease, delaying the progression of the disease, increasing or improving the quality of life, weight gain, and/or prolonging survival. The methods of the invention contemplate any one or more of these aspects of treatment.

An "effective amount" of an antibody or composition disclosed herein is an amount sufficient to achieve the stated purpose. An "effective amount" can be determined empirically and by known methods relevant to the stated purpose.

As used herein, "pharmaceutically acceptable" or "pharmacologically compatible" refers to materials that are not biologically or otherwise undesirable, e.g., the materials may be incorporated into a pharmaceutical composition for administration to a patient without causing any significant undesirable biological effects or interacting in a deleterious manner with any of the additional components of the composition in which they are contained. The pharmaceutically acceptable carrier or excipient preferably meets the required standards for toxicological and manufacturing testing and/or is included in the inactive ingredient guidelines as set out by the U.S. food and drug administration.

It is to be understood that the embodiments of the invention described herein include "consisting of" and/or "consisting essentially of an embodiment.

Reference herein to "about" a value or parameter includes (and describes) variations that are directed to that value or parameter itself. For example, a description referring to "about X" includes a description of "X".

As used herein, reference to "not" a value or parameter generally means and describes "in addition to a value or parameter.

As used herein and in the appended claims, the singular forms "a", "an" and "the" include plural referents unless the context clearly dictates otherwise.

Heteromultimeric proteins

The present application provides heteromultimeric proteins, such as heterodimeric proteins, comprising a first polypeptide comprising a first antibody heavy chain constant domain 3(CH3) domain and a second polypeptide comprising a second CH3 domain, wherein the first CH3 domain and the second CH3 domain comprise engineered hydrophobic interactions and electrostatic interactions. In some embodiments, the first CH3 domain and the second CH3 domain further comprise knob entry hole ("KIH") residues. Exemplary structures of heteromultimeric proteins include heterodimers (e.g., bispecific immunoadhesins), heterotrimers (e.g., antibody-immunoadhesin chimeras), heterotetramers (e.g., bispecific antibodies), and other oligomeric structures. In some embodiments, the heteromultimeric protein is a bispecific antibody, such as a common light chain antibody or an antibody having two different light chains. See, for example, fig. 1).

In some embodiments, a heteromultimeric protein (e.g., a heterodimeric protein) is provided comprising a first polypeptide comprising a first CH3 domain and a second polypeptide comprising a second CH3 domain, wherein the first CH3 domain comprises engineered amino acid residues that form hydrophobic interactions with native amino acid residues of the second CH3 domain, and the second CH3 domain comprises engineered amino acid residues that form ionic bonds with native amino acid residues in the first CH3 domain. In some embodiments, a heteromultimeric protein (e.g., a heterodimeric protein) is provided comprising a first polypeptide comprising a first CH3 domain and a second polypeptide comprising a second CH3 domain, wherein the first CH3 domain comprises a first engineered amino acid residue that forms a hydrophobic interaction with a first native amino acid residue of the second CH3 domain and a second engineered amino acid residue that forms an ionic bond with a second native amino acid residue in the second CH3 domain. In some embodiments, the first CH3 domain and the second CH3 domain further comprise a KIH residue.

In some embodiments, a heteromultimeric protein (e.g., a heterodimeric protein) is provided comprising a first polypeptide comprising a first CH3 domain and a second polypeptide comprising a second CH3 domain, wherein the first CH3 domain comprises a substitution with a large hydrophobic amino acid at amino acid position 354 relative to the wild-type CH3 domain, and/or the second CH3 domain comprises a substitution with a negatively charged amino acid at amino acid position 347 relative to the wild-type CH3 domain, and wherein the amino acid residue numbering is based on EU numbering. In some embodiments, a heteromultimeric protein (e.g., a heterodimeric protein) is provided comprising a first polypeptide comprising a first CH3 domain and a second polypeptide comprising a second CH3 domain, wherein the first CH3 domain comprises a substitution with a large hydrophobic amino acid at amino acid position 354 relative to the wild-type CH3 domain, and/or a substitution with a negatively charged amino acid at amino acid position 347 relative to the wild-type CH3 domain, and wherein the amino acid residue numbering is based on EU numbering. In some embodiments, the large hydrophobic amino acid is tyrosine (Y), phenylalanine (F), or tryptophan (W). In some embodiments, the negatively charged amino acid is aspartic acid (D) or glutamic acid (E). In some embodiments, the large hydrophobic amino acid at amino acid position 354 in one CH3 domain forms a hydrophobic interaction with an amino acid residue in another CH3 domain. In some embodiments, another CH3 domain comprises a large hydrophobic residue at amino acid position 349 (e.g., Y349). In some embodiments, the negatively charged amino acid at amino acid position 347 in one CH3 domain forms an ionic bond with an amino acid residue in another CH3 domain. In some embodiments, another CH3 domain comprises a positively charged residue at amino acid position 360 (e.g., K360). In some embodiments, the first CH3 domain and the second CH3 domain further comprise KIH residues, such as T366Y and Y407T. In some embodiments, the first and second CH3 domains are a human CH3 domain, a murine CH3 domain, a rat CH3 domain, a camelid CH3 domain, or a rabbit CH3 domain. Figure 7 shows a sequence alignment of the Fc region sequences of IgG molecules from different species.

In some embodiments, a heteromultimeric protein (e.g., a heterodimeric protein) is provided comprising a first polypeptide comprising a first CH3 domain and a second polypeptide comprising a second CH3 domain, wherein the first CH3 domain comprises a substitution of a large hydrophobic amino acid for S354 and/or the second CH3 domain comprises a substitution of a negatively charged amino acid for Q347, and wherein the amino acid residue numbering is based on EU numbering. In some embodiments, a heteromultimeric protein (e.g., a heterodimeric protein) is provided comprising a first polypeptide comprising a first CH3 domain and a second polypeptide comprising a second CH3 domain, wherein the first CH3 domain comprises a substitution of a large hydrophobic amino acid for S354, and/or a substitution of a negatively charged amino acid for Q347, and wherein the amino acid residue numbering is based on EU numbering. In some embodiments, the large hydrophobic amino acid is Y, F or W. In some embodiments, the negatively charged amino acid is D or E. In some embodiments, the large hydrophobic amino acid at amino acid position 354 in one CH3 domain forms a hydrophobic interaction with an amino acid residue in another CH3 domain. In some embodiments, another CH3 domain comprises a large hydrophobic residue at amino acid position 349 (e.g., Y349). In some embodiments, the negatively charged amino acid at amino acid position 347 in one CH3 domain forms an ionic bond with an amino acid residue in another CH3 domain. In some embodiments, another CH3 domain comprises a positively charged residue at amino acid position 360 (e.g., K360). In some embodiments, the CH3 domain is a human CH3 domain. In some embodiments, the first CH3 domain and the second CH3 domain further comprise KIH residues, such as T366Y and Y407T.

In some embodiments, a heteromultimeric protein (e.g., a heterodimeric protein) is provided comprising a first polypeptide comprising a first human CH3 domain and a second polypeptide comprising a second human CH3 domain, wherein the first CH3 domain comprises S354Y and the second CH3 domain comprises Q347E, and wherein the amino acid residue numbering is based on EU numbering. In some embodiments, S354Y in the first CH3 domain forms a hydrophobic interaction with an amino acid residue in the second CH3 domain. In some embodiments, the second CH3 domain comprises a large hydrophobic residue at amino acid position 349 (e.g., Y349). In some embodiments, Q347E in the second CH3 domain forms an ionic bond with an amino acid residue in the first CH3 domain. In some embodiments, the first CH3 domain comprises a positively charged residue at amino acid position 360 (e.g., K360). In some embodiments, the first CH3 domain and the second CH3 domain further comprise a KIH residue. In some embodiments, the second CH3 domain does not comprise a substitution of Y349, for example Y349S.

The substitutions in the CH3 domain at amino acid positions 354 and 347 relative to the wild-type CH3 domain described herein can be combined with any known mutation and/or engineered residue in the CH3 domain, including knob-entry-hole ("KIH") mutations, which can facilitate heterodimer formation. Any KIH residue compatible with substitution at amino acid positions 354 and 347 can be used, including for example T366Y and Y407T. In some embodiments, the heteromultimeric protein does not comprise a KIH residue in the CH3 domain. In some embodiments, the heteromultimeric proteins comprise additional non-KIH mutations that promote heterodimer formation, such as cysteine residues that form disulfide bonds, and/or engineered residues that form electrostatic interactions. See, for example, U.S. patent No. 8,592,562.

In some embodiments, a heteromultimeric protein (e.g., a heterodimeric protein) is provided comprising a first polypeptide comprising a first human CH3 domain and a second polypeptide comprising a second human CH3 domain, wherein the first CH3 domain comprises S354Y and T366Y and the second CH3 domain comprises Q347E and Y407T, and wherein the amino acid residue numbering is based on EU numbering. In some embodiments, S354Y in the first CH3 domain forms a hydrophobic interaction with an amino acid residue in the second CH3 domain. In some embodiments, the second CH3 domain comprises a large hydrophobic residue at amino acid position 349 (e.g., Y349). In some embodiments, Q347E in the second CH3 domain forms an ionic bond with an amino acid residue in the first CH3 domain. In some embodiments, the first CH3 domain comprises a positively charged residue at amino acid position 360 (e.g., K360). In some embodiments, the second CH3 domain does not comprise a substitution of Y349, for example Y349S. In some embodiments, the first CH3 and the second CH3 do not comprise further mutations compared to the wild-type human CH3 domain.

In some embodiments, a heteromultimeric protein (e.g., a heterodimeric protein) is provided comprising a first polypeptide comprising a first human CH3 domain and a second polypeptide comprising a second human CH3 domain, wherein the first CH3 domain comprises S354Y and Y407T and the second CH3 domain comprises Q347E and T366Y, and wherein the amino acid residue numbering is based on EU numbering. In some embodiments, S354Y in the first CH3 domain forms a hydrophobic interaction with an amino acid residue in the second CH3 domain. In some embodiments, the second CH3 domain comprises a large hydrophobic residue at amino acid position 349 (e.g., Y349). In some embodiments, Q347E in the second CH3 domain forms an ionic bond with an amino acid residue in the first CH3 domain. In some embodiments, the first CH3 domain comprises a positively charged residue at amino acid position 360 (e.g., K360). In some embodiments, the second CH3 domain does not comprise a substitution of Y349, for example Y349S. In some embodiments, the first CH3 and the second CH3 do not comprise further mutations compared to the wild-type human CH3 domain.

The first polypeptide is any polypeptide associated with the second polypeptide. The first polypeptide and the second polypeptide interact at an interface that includes the CH3-CH3 interface. In some embodiments, each of the first and second polypeptides may comprise one or more additional domains, such as binding domains (e.g., antibody variable domains, receptor binding domains, ligand binding domains, or enzymatic domains) and antibody constant domains (or portions thereof), including CH2, CH1, hinges, and CL domains. Exemplary first and second polypeptides include antibody heavy chain polypeptides, chimeras combining antibody constant domains with binding domains of heterologous polypeptides (i.e., immunoadhesins, such as receptor-Fc fusion polypeptides, ligand-Fc fusion polypeptides), and antibody variable domain polypeptides (e.g., diabodies, bispecific bulky (maxibody) or bispecific peptibodies).

In some embodiments, the first polypeptide and/or the second polypeptide comprises a CH2 domain. In some embodiments, the first polypeptide and/or the second polypeptide comprises a CH1 domain and a CH2 domain. In some embodiments, the first polypeptide and the second polypeptide each comprise an Fc region of an IgG, such as a human IgG. In some embodiments, the first polypeptide and the second polypeptide each comprise an Fc region of IgG1, IgG2, IgG3, or IgG 4. Exemplary sequences of the wild-type Fc region of IgG molecules are shown in figure 7. Exemplary mutant Fc sequences are shown in table 1. The exact amino acids corresponding to the various domains of an IgG molecule may be understood differently by those skilled in the art. Thus, the N-terminus or C-terminus of a domain as outlined herein may be extended or shortened by 1, 2, 3, 4, 5, 6, 7, 8, 9 or even 10 amino acids. Furthermore, it is noted that there are a number of known numbering schemes for specifying amino acid positions in IgG, which may differ from the EU numbering scheme used in the present patent application. In some embodiments, the Fc region is from the constant region of an IgA, IgD, IgE, or IgM heavy chain.

In some embodiments, heteromultimeric proteins (e.g., heterodimeric proteins) are provided comprising a first polypeptide comprising the CH3 domain of SEQ ID NO:1 (e.g., amino acids 116-217) and a second polypeptide comprising the CH3 domain of SEQ ID NO:2 (e.g., amino acids 116-217). In some embodiments, heteromultimeric proteins (e.g., heterodimeric proteins) are provided comprising a first polypeptide comprising the CH3 domain of SEQ ID NO:3 (e.g., amino acids 116-217) and a second polypeptide comprising the CH3 domain of SEQ ID NO:4 (e.g., amino acids 116-217). In some embodiments, a heteromultimeric protein (e.g., a heterodimeric protein) is provided comprising a first polypeptide comprising the amino acid sequence of SEQ ID NO:1 and a second polypeptide comprising the amino acid sequence of SEQ ID NO: 2. In some embodiments, a heteromultimeric protein (e.g., a heterodimeric protein) is provided comprising a first polypeptide comprising the amino acid sequence of SEQ ID NO. 3 and a second polypeptide comprising the amino acid sequence of SEQ ID NO. 4.

Table 1 exemplary IgG Fc sequences.

Exemplary embodiments of the invention include, but are not limited to, antibodies, multispecific antibodies (e.g., bispecific antibodies), monospecific multivalent antibodies, bispecific bulky (i.e., scFv-Fc fusion proteins), monomeric (i.e., Fab-Fc), peptibodies (i.e., one peptide fused to one arm of a heterodimeric Fc molecule, including monovalent, bivalent, monospecific, and bispecific peptibodies), immunoadhesins, antibody-immunoadhesin chimeras, receptor-Fc fusion proteins, and ligand-Fc fusion proteins.

In some embodiments, the heteromultimeric protein is an antibody, such as a multispecific (e.g., bispecific) antibody. In some embodiments, the heteromultimeric protein comprises one or more antibody light chains. In some embodiments, the heteromultimeric protein is a common light chain antibody, i.e., a bispecific antibody comprising two identical light chains. In some embodiments, the heteromultimeric protein is a common variable light chain antibody, i.e., a bispecific antibody comprising two light chains having the same light chain variable region. In some embodiments, the heteromultimeric protein comprises light chains having the same light chain variable region. In some embodiments, the heteromultimeric protein comprises a light chain comprising a light chain variable region derived from the same parent antibody. In some embodiments, the heteromultimeric protein comprises two different light chains. In some embodiments, the heteromultimeric protein comprises light chains derived from two different antibodies. In some embodiments, the heteromultimeric protein comprises a lambda light chain. In some embodiments, the heteromultimeric protein comprises a kappa light chain. In some embodiments, the heteromultimeric protein comprises a lambda light chain and a kappa light chain. Common light chain antibodies, including common variable light chain antibodies, are known in the art. See, for example, U.S. patent No. 8642745B2 and U.S. patent application publication No. 2016/0319036a1, which are incorporated herein by reference. Any known common variable light chain strategy can be used in combination with the CH 3-based heterodimerization strategy described herein to provide multispecific antibodies.

In some embodiments, the heteromultimeric protein is an antibody, such as a multispecific (e.g., bispecific) antibody, comprising a first antigen-binding fragment that binds a first antigen and a second antigen-binding fragment that binds a second antigen. In some embodiments, the heteromultimeric protein comprises a first antigen-binding fragment that binds a first antigen and a second antigen-binding fragment, and a third antigen-binding fragment that binds a second antigen. In some embodiments, the heteromultimeric protein comprises a first antigen-binding fragment that binds a tumor antigen and a second antigen-binding fragment, and a third antigen-binding fragment that binds a second antigen (such as CD 3). In some embodiments, the heteromultimeric protein comprises a tumor antigen binding region having a relatively low affinity for a tumor antigen, and thus binds much less strongly to healthy cells having a lower density of tumor antigens. In some embodiments, the first, second, and/or third antigen-binding fragment is a Fab. In some embodiments, the first, second and/or third antigen-binding fragment is a VHH. In some embodiments, the first, second, and/or third antigen-binding fragment is an scFv. In some embodiments, the multispecific antibody comprises two Fab domains. In some embodiments, the multispecific antibody comprises three Fab domains. In some embodiments, the multispecific antibody comprises two VHH domains. In some embodiments, the multispecific antibody comprises three VHH domains. In some embodiments, the multispecific antibody comprises a Fab domain and a VHH domain. In some embodiments, the multispecific antibody comprises two Fab domains and a VHH domain. In some embodiments, the multispecific antibody comprises a Fab domain and two VHH domains. Exemplary structures of multispecific antibodies described herein are shown in figure 16.

In some embodiments, a multispecific (e.g., bispecific) antibody is provided comprising: (a) a first heavy chain comprising, from N-terminus to C-terminus: a first heavy chain variable domain (VH1), a first heavy chain constant domain 1(CH1), a first heavy chain constant domain 2(CH2) and a first CH3 domain; (b) a first light chain comprising, from N-terminus to C-terminus: a first light chain variable domain (VL1) and a first light chain constant domain (CL); (c) a second heavy chain comprising from N-terminus to C-terminus: a second heavy chain variable domain (VH2), a second CH1, a second CH2 and a second CH3 domain; and (d) a second light chain comprising from the N-terminus to the C-carbon terminus: a second light chain variable domain (VL2) and a second CL; wherein VH1 associates with VL1 to form a first antigen binding site that specifically binds to a first target, and VH2 associates with VL2 to form a second antigen binding site that specifically binds to a second target; wherein the first CH3 domain comprises a substitution with a large hydrophobic amino acid at amino acid position 354 relative to the wild-type CH3 domain and/or the second CH3 domain comprises a substitution with a negatively charged amino acid at amino acid position 347 relative to the wild-type CH3 domain, and wherein the amino acid residue numbering is based on EU numbering. In some embodiments, the large hydrophobic amino acid is Y, F or W. In some embodiments, the negatively charged amino acid is D or E. In some embodiments, the large hydrophobic amino acid at amino acid position 354 forms a hydrophobic interaction with an amino acid residue in the second CH3 domain. In some embodiments, the second CH3 domain comprises a large hydrophobic residue at amino acid position 349 (e.g., Y349). In some embodiments, the negatively charged amino acid at amino acid position 347 forms an ionic bond with an amino acid residue in the first CH3 domain. In some embodiments, the first CH3 domain comprises a positively charged residue at amino acid position 360 (e.g., K360). In some embodiments, the first and second CH3 domains are a human CH3 domain, a murine CH3 domain, a rat CH3 domain, a camelid CH3 domain, or a rabbit CH3 domain. In some embodiments, the first CH3 domain and the second CH3 domain further comprise KIH residues, such as T366Y and Y407T. In some embodiments, the multispecific antibody is chimeric, human, or humanized. In some embodiments, VL1 and VL2 have the same amino acid sequence. In some embodiments, VL1 and VL2 have different amino acid sequences.

In some embodiments, a multispecific (e.g., bispecific) antibody is provided comprising: (a) a first heavy chain comprising, from N-terminus to C-carbon terminus: a VH1 domain, a first CH1 domain, a first CH2 domain, and a first CH3 domain; (b) a first light chain comprising, from N-terminus to C-terminus: a VL1 domain and a first CL domain; (c) a second heavy chain comprising from N-terminus to C-terminus: a VH2 domain, a second CH1 domain, a second CH2 domain, and a second CH3 domain; and (d) a second light chain comprising from N-terminus to C-terminus: a VL2 domain and a second CL domain; wherein VH1 associates with VL1 to form a first antigen binding site that specifically binds to a first target, and VH2 associates with VL2 to form a second antigen binding site that specifically binds to a second target; wherein the first CH3 domain comprises a substitution of a large hydrophobic amino acid for S354 and/or the second CH3 domain comprises a substitution of a negatively charged amino acid for Q347, and wherein the amino acid residue numbering is based on EU numbering. In some embodiments, the large hydrophobic amino acid is Y, F or W. In some embodiments, the negatively charged amino acid is D or E. In some embodiments, the large hydrophobic amino acid at amino acid position 354 forms a hydrophobic interaction with an amino acid residue in the second CH3 domain. In some embodiments, the second CH3 domain comprises a large hydrophobic residue at amino acid position 349 (e.g., Y349). In some embodiments, the negatively charged amino acid at amino acid position 347 forms an ionic bond with an amino acid residue in the first CH3 domain. In some embodiments, the first CH3 domain comprises a positively charged residue at amino acid position 360 (e.g., K360). In some embodiments, the first CH3 domain and the second CH3 domain further comprise KIH residues, such as T366Y and Y407T. In some embodiments, the multispecific antibody is chimeric, human, or humanized. In some embodiments, VL1 and VL2 have the same amino acid sequence. In some embodiments, VL1 and VL2 have different amino acid sequences.

In some embodiments, a multispecific (e.g., bispecific) antibody is provided comprising: (a) a first heavy chain comprising, from N-terminus to C-carbon terminus: a VH1 domain, a first CH1 domain, a first CH2 domain, and a first CH3 domain; (b) a first light chain comprising, from N-terminus to C-terminus: a VL1 domain and a first CL domain; (c) a second heavy chain comprising from N-terminus to C-terminus: a VH2 domain, a second CH1 domain, a second CH2 domain, and a second CH3 domain; and (d) a second light chain comprising from N-terminus to C-terminus: a VL2 domain and a second CL domain; wherein VH1 associates with VL1 to form a first antigen binding site that specifically binds to a first target, and VH2 associates with VL2 to form a second antigen binding site that specifically binds to a second target; wherein the first CH3 domain comprises S354Y and T366Y and the second CH3 domain comprises Q347E and Y407T, wherein the numbering of the amino acid residues is based on EU numbering. In some embodiments, S354Y of the first CH3 domain forms a hydrophobic interaction with an amino acid residue in the second CH3 domain. In some embodiments, the second CH3 domain comprises a large hydrophobic residue at amino acid position 349 (e.g., Y349). In some embodiments, Q347E of the second CH3 domain forms an ionic bond with an amino acid residue in the first CH3 domain. In some embodiments, the first CH3 domain comprises a positively charged residue at amino acid position 360 (e.g., K360). In some embodiments, the second CH3 domain does not comprise a substitution of Y349, for example Y349S. In some embodiments, the first CH3 and the second CH3 do not comprise further mutations compared to the wild-type human CH3 domain. In some embodiments, the multispecific antibody is chimeric, human, or humanized. In some embodiments, VL1 and VL2 have the same amino acid sequence. In some embodiments, VL1 and VL2 have different amino acid sequences. In some embodiments, the first antigen-binding site specifically binds CD3 and the second antigen-binding site specifically binds to a tumor antigen, or the first antigen-binding site specifically binds CD3 and the second antigen-binding site specifically binds to a tumor antigen. In some embodiments, the first antigen-binding site specifically binds CD20 and the second antigen-binding site specifically binds CD3, or the first antigen-binding site specifically binds CD3 and the second antigen-binding site specifically binds CD 20. In some embodiments, the first antigen binding site specifically binds to HER2 and the second antigen binding site specifically binds to CD3, or the first antigen binding site specifically binds to CD3 and the second antigen binding site specifically binds to HER 2.

In some embodiments, a multispecific (e.g., bispecific) antibody is provided comprising: (a) a first heavy chain comprising, from N-terminus to C-carbon terminus: a VH1 domain, a first CH1 domain, a first CH2 domain, and a first CH3 domain; (b) a first light chain comprising, from N-terminus to C-terminus: a VL1 domain and a first CL domain; (c) a second heavy chain comprising from N-terminus to C-terminus: a VH2 domain, a second CH1 domain, a second CH2 domain, and a second CH3 domain; and (d) a second light chain comprising from N-terminus to C-terminus: a VL2 domain and a second CL domain; wherein VH1 associates with VL1 to form a first antigen binding site that specifically binds to a first target, and VH2 associates with VL2 to form a second antigen binding site that specifically binds to a second target; wherein the first CH3 domain comprises S354Y and Y407T and the second CH3 domain comprises Q347E and T366Y, wherein the numbering of the amino acid residues is based on EU numbering. In some embodiments, S354Y of the first CH3 domain forms a hydrophobic interaction with an amino acid residue in the second CH3 domain. In some embodiments, the second CH3 domain comprises a large hydrophobic residue at amino acid position 349 (e.g., Y349). In some embodiments, Q347E of the second CH3 domain forms an ionic bond with an amino acid residue in the first CH3 domain. In some embodiments, the first CH3 domain comprises a positively charged residue at amino acid position 360 (e.g., K360). In some embodiments, the second CH3 domain does not comprise a substitution of Y349, for example Y349S. In some embodiments, the first CH3 and the second CH3 do not comprise further mutations compared to the wild-type human CH3 domain. In some embodiments, the multispecific antibody is chimeric, human, or humanized. In some embodiments, VL1 and VL2 have the same amino acid sequence. In some embodiments, VL1 and VL2 have different amino acid sequences. In some embodiments, the first antigen-binding site specifically binds CD3 and the second antigen-binding site specifically binds to a tumor antigen, or the first antigen-binding site specifically binds CD3 and the second antigen-binding site specifically binds to a tumor antigen. In some embodiments, the first antigen-binding site specifically binds CD20 and the second antigen-binding site specifically binds CD3, or the first antigen-binding site specifically binds CD3 and the second antigen-binding site specifically binds CD 20. In some embodiments, the first antigen binding site specifically binds to HER2 and the second antigen binding site specifically binds to CD3, or the first antigen binding site specifically binds to CD3 and the second antigen binding site specifically binds to HER 2.

In some embodiments, a multispecific (e.g., bispecific) antibody is provided comprising: (a) a first heavy chain comprising, from N-terminus to C-terminus: a first VHH domain (VHH1), a first heavy chain constant domain 2(CH2) and a first CH3 domain; and (b) a second heavy chain comprising from N-terminus to C-terminus: a second VHH domain (VHH2), a second CH2 and a second CH3 domain; wherein VHH1 specifically binds to a first target and VHH2 specifically binds to a second target; wherein the first CH3 domain comprises a substitution with a large hydrophobic amino acid at amino acid position 354 relative to the wild-type CH3 domain and/or the second CH3 domain comprises a substitution with a negatively charged amino acid at amino acid position 347 relative to the wild-type CH3 domain, and wherein amino acid residue numbering is based on EU numbering. In some embodiments, the large hydrophobic amino acid is Y, F or W. In some embodiments, the negatively charged amino acid is D or E. In some embodiments, the large hydrophobic amino acid at amino acid position 354 forms a hydrophobic interaction with an amino acid residue in the second CH3 domain. In some embodiments, the second CH3 domain comprises a large hydrophobic residue at amino acid position 349 (e.g., Y349). In some embodiments, the negatively charged amino acid at amino acid position 347 forms an ionic bond with an amino acid residue in the first CH3 domain. In some embodiments, the first CH3 domain comprises a positively charged residue at amino acid position 360 (e.g., K360). In some embodiments, the first and second CH3 domains are a human CH3 domain, a murine CH3 domain, a rat CH3 domain, a camelid CH3 domain, or a rabbit CH3 domain. In some embodiments, the first CH3 domain and the second CH3 domain further comprise KIH residues, such as T366Y and Y407T. In some embodiments, the multispecific antibody is chimeric, human, or humanized.

In some embodiments, a multispecific (e.g., bispecific) antibody is provided comprising: (a) a first heavy chain comprising, from N-terminus to C-terminus: a first VHH domain (VHH1), a first heavy chain constant domain 2(CH2) and a first CH3 domain; and (b) a second heavy chain comprising from N-terminus to C-terminus: a second VHH domain (VHH2), a second CH2 and a second CH3 domain; wherein VHH1 specifically binds to a first target and VHH2 specifically binds to a second target; wherein the first CH3 domain comprises a substitution of a large hydrophobic amino acid for S354 and/or the second CH3 domain comprises a substitution of a negatively charged amino acid for Q347, and wherein the amino acid residue numbering is based on EU numbering. In some embodiments, the large hydrophobic amino acid is Y, F or W. In some embodiments, the negatively charged amino acid is D or E. In some embodiments, the large hydrophobic amino acid at amino acid position 354 forms a hydrophobic interaction with an amino acid residue in the second CH3 domain. In some embodiments, the second CH3 domain comprises a large hydrophobic residue at amino acid position 349 (e.g., Y349). In some embodiments, the negatively charged amino acid at amino acid position 347 forms an ionic bond with an amino acid residue in the first CH3 domain. In some embodiments, the first CH3 domain comprises a positively charged residue at amino acid position 360 (e.g., K360). In some embodiments, the first CH3 domain and the second CH3 domain further comprise KIH residues, such as T366Y and Y407T. In some embodiments, the multispecific antibody is chimeric, human, or humanized.

In some embodiments, a multispecific (e.g., bispecific) antibody is provided comprising: (a) a first heavy chain comprising, from N-terminus to C-terminus: a first VHH domain (VHH1), a first heavy chain constant domain 2(CH2) and a first CH3 domain; and (b) a second heavy chain comprising from N-terminus to C-terminus: a second VHH domain (VHH2), a second CH2 and a second CH3 domain; wherein VHH1 specifically binds to a first target and VHH2 specifically binds to a second target; wherein the first CH3 domain comprises S354Y and T366Y, the second CH3 domain comprises Q347E and Y407T, and wherein amino acid residue numbering is based on EU numbering. In some embodiments, S354Y of the first CH3 domain forms a hydrophobic interaction with an amino acid residue in the second CH3 domain. In some embodiments, the second CH3 domain comprises a large hydrophobic residue at amino acid position 349 (e.g., Y349). In some embodiments, Q347E of the second CH3 domain forms an ionic bond with an amino acid residue in the first CH3 domain. In some embodiments, the first CH3 domain comprises a positively charged residue at amino acid position 360 (e.g., K360). In some embodiments, the second CH3 domain does not comprise a substitution of Y349, for example Y349S. In some embodiments, the first CH3 and the second CH3 do not comprise further mutations compared to the wild-type human CH3 domain. In some embodiments, the multispecific antibody is chimeric, human, or humanized.

In some embodiments, a multispecific (e.g., bispecific) antibody is provided comprising: (a) a first heavy chain comprising, from N-terminus to C-terminus: a first VHH domain (VHH1), a first heavy chain constant domain 2(CH2) and a first CH3 domain; and (b) a second heavy chain comprising from N-terminus to C-terminus: a second VHH domain (VHH2), a second CH2 and a second CH3 domain; wherein VHH1 specifically binds to a first target and VHH2 specifically binds to a second target; wherein the first CH3 domain comprises S354Y and Y407T, the second CH3 domain comprises Q347E and T366Y, and wherein amino acid residue numbering is based on EU numbering. In some embodiments, S354Y of the first CH3 domain forms a hydrophobic interaction with an amino acid residue in the second CH3 domain. In some embodiments, the second CH3 domain comprises a large hydrophobic residue at amino acid position 349 (e.g., Y349). In some embodiments, Q347E of the second CH3 domain forms an ionic bond with an amino acid residue in the first CH3 domain. In some embodiments, the first CH3 domain comprises a positively charged residue at amino acid position 360 (e.g., K360). In some embodiments, the second CH3 domain does not comprise a substitution of Y349, for example Y349S. In some embodiments, the first CH3 and the second CH3 do not comprise further mutations compared to the wild-type human CH3 domain. In some embodiments, the multispecific antibody is chimeric, human, or humanized.

In some embodiments, a multispecific (e.g., bispecific) antibody is provided comprising: (a) a first heavy chain comprising, from N-terminus to C-terminus: a VHH domain, a first heavy chain constant domain 2(CH2) and a first CH3 domain; (b) a second heavy chain comprising, from N-terminus to C-carbon terminus: a first heavy chain variable domain (VH1), a first CH1, a second CH2 and a second CH3 domain; and (d) a light chain comprising from N-terminus to C-terminus: a first light chain variable domain (VL1) and a first CL; wherein VH1 associates with VL1 to form a first antigen binding site that specifically binds to a first target, and the VHH domain specifically binds to a second target; wherein the first CH3 domain comprises a substitution with a large hydrophobic amino acid at amino acid position 354 relative to the wild-type CH3 domain and/or the second CH3 domain comprises a substitution with a negatively charged amino acid at amino acid position 347 relative to the wild-type CH3 domain or wherein the second CH3 domain comprises a substitution with a large hydrophobic amino acid at amino acid position 354 relative to the wild-type CH3 domain and/or the first CH3 domain comprises a substitution with a negatively charged amino acid at amino acid position 347 relative to the wild-type CH3 domain; and wherein amino acid residue numbering is based on EU numbering. In some embodiments, the large hydrophobic amino acid is Y, F or W. In some embodiments, the negatively charged amino acid is D or E. In some embodiments, the large hydrophobic amino acid at amino acid position 354 forms a hydrophobic interaction with an amino acid residue in the second CH3 domain. In some embodiments, the second CH3 domain comprises a large hydrophobic residue at amino acid position 349 (e.g., Y349). In some embodiments, the negatively charged amino acid at amino acid position 347 forms an ionic bond with an amino acid residue in the first CH3 domain. In some embodiments, the first CH3 domain comprises a positively charged residue at amino acid position 360 (e.g., K360). In some embodiments, the first and second CH3 domains are a human CH3 domain, a murine CH3 domain, a rat CH3 domain, a camelid CH3 domain, or a rabbit CH3 domain. In some embodiments, the first CH3 domain and the second CH3 domain further comprise KIH residues, such as T366Y and Y407T. In some embodiments, the multispecific antibody is chimeric, human, or humanized.

In some embodiments, a multispecific (e.g., bispecific) antibody is provided comprising: (a) a first heavy chain comprising, from N-terminus to C-terminus: a VHH domain, a first heavy chain constant domain 2(CH2) and a first CH3 domain; (b) a second heavy chain comprising, from N-terminus to C-carbon terminus: a first heavy chain variable domain (VH1), a first CH1, a second CH2 and a second CH3 domain; and (d) a light chain comprising from N-terminus to C-terminus: a first light chain variable domain (VL1) and a first CL; wherein VH1 associates with VL1 to form a first antigen binding site that specifically binds to a first target, and the VHH domain specifically binds to a second target; wherein the first CH3 domain comprises a substitution of the large hydrophobic amino acid for S354 and/or the second CH3 domain comprises a substitution of the negatively charged amino acid for Q347, or wherein the second CH3 domain comprises a substitution of the large hydrophobic amino acid for S354 and/or the first CH3 domain comprises a substitution of the negatively charged amino acid for Q347; and wherein amino acid residue numbering is based on EU numbering. In some embodiments, the large hydrophobic amino acid is Y, F or W. In some embodiments, the negatively charged amino acid is D or E. In some embodiments, the large hydrophobic amino acid at amino acid position 354 forms a hydrophobic interaction with an amino acid residue in the second CH3 domain. In some embodiments, the second CH3 domain comprises a large hydrophobic residue at amino acid position 349 (e.g., Y349). In some embodiments, the negatively charged amino acid at amino acid position 347 forms an ionic bond with an amino acid residue in the first CH3 domain. In some embodiments, the first CH3 domain comprises a positively charged residue at amino acid position 360 (e.g., K360). In some embodiments, the first CH3 domain and the second CH3 domain further comprise KIH residues, such as T366Y and Y407T.

In some embodiments, a multispecific (e.g., bispecific) antibody is provided comprising: (a) a first heavy chain comprising, from N-terminus to C-terminus: a VHH domain, a first heavy chain constant domain 2(CH2) and a first CH3 domain; (b) a second heavy chain comprising, from N-terminus to C-carbon terminus: a first heavy chain variable domain (VH1), a first CH1, a second CH2 and a second CH3 domain; and (d) a light chain comprising from N-terminus to C-terminus: a first light chain variable domain (VL1) and a first CL; wherein VH1 associates with VL1 to form a first antigen binding site that specifically binds to a first target, and the VHH domain specifically binds to a second target; wherein the first CH3 domain comprises S354Y and T366Y and the second CH3 domain comprises Q347E and Y407T, or wherein the second CH3 domain comprises S354Y and T366Y and the first CH3 domain comprises Q347E and Y407T; and wherein amino acid residue numbering is based on EU numbering. In some embodiments, S354Y of the first CH3 domain forms a hydrophobic interaction with an amino acid residue in the second CH3 domain. In some embodiments, the second CH3 domain comprises a large hydrophobic residue at amino acid position 349 (e.g., Y349). In some embodiments, Q347E of the second CH3 domain forms an ionic bond with an amino acid residue in the first CH3 domain. In some embodiments, the first CH3 domain comprises a positively charged residue at amino acid position 360 (e.g., K360). In some embodiments, the second CH3 domain does not comprise a substitution of Y349, for example Y349S. In some embodiments, the first CH3 and the second CH3 do not comprise further mutations compared to the wild-type human CH3 domain. In some embodiments, the multispecific antibody is chimeric, human, or humanized. In some embodiments, the VHH specifically binds a tumor antigen (e.g., BCMA) and the first antigen binding site specifically binds CD 3.

In some embodiments, a multispecific (e.g., bispecific) antibody is provided comprising: (a) a first heavy chain comprising, from N-terminus to C-terminus: a VHH domain, a first heavy chain constant domain 2(CH2) and a first CH3 domain; (b) a second heavy chain comprising, from N-terminus to C-carbon terminus: a first heavy chain variable domain (VH1), a first CH1, a second CH2 and a second CH3 domain; and (d) a light chain comprising from N-terminus to C-terminus: a first light chain variable domain (VL1) and a first CL; wherein VH1 associates with VL1 to form a first antigen binding site that specifically binds to a first target, and the VHH domain specifically binds to a second target; wherein the first CH3 domain comprises S354Y and Y407T and the second CH3 domain comprises Q347E and T366Y, or wherein the second CH3 domain comprises S354Y and Y407T and the first CH3 domain comprises Q347E and T366Y; and wherein amino acid residue numbering is based on EU numbering. In some embodiments, S354Y of the first CH3 domain forms a hydrophobic interaction with an amino acid residue in the second CH3 domain. In some embodiments, the second CH3 domain comprises a large hydrophobic residue at amino acid position 349 (e.g., Y349). In some embodiments, Q347E of the second CH3 domain forms an ionic bond with an amino acid residue in the first CH3 domain. In some embodiments, the first CH3 domain comprises a positively charged residue at amino acid position 360 (e.g., K360). In some embodiments, the second CH3 domain does not comprise a substitution of Y349, for example Y349S. In some embodiments, the first CH3 and the second CH3 do not comprise further mutations compared to the wild-type human CH3 domain. In some embodiments, the multispecific antibody is chimeric, human, or humanized. In some embodiments, the VHH specifically binds a tumor antigen (e.g., BCMA) and the first antigen binding site specifically binds CD 3.

In some embodiments, multispecific (e.g., bispecific or trispecific) antibodies are provided comprising: (a) a first heavy chain comprising, from N-terminus to C-terminus: a first heavy chain variable domain (VH1), a first CH1, a second heavy chain variable domain (VH2), a second CH1, a first CH2 and a first CH3 domain; (b) a first light chain comprising, from N-terminus to C-terminus: a first light chain variable domain (VL1), a first CL, a second light chain variable domain (VL2), and a second CL; (c) a second heavy chain comprising from N-terminus to C-terminus: a third triple chain variable domain (VH3), a third CH1, a second CH2 and a second CH3 domain; and (d) a second light chain comprising from N-terminus to C-terminus: a third light chain variable domain (VL3) and a third CL; wherein VH1 associates with VL1 to form a first antigen binding site that specifically binds to a first target, and VH2 associates with VL2 to form a second antigen binding site that specifically binds to a second target, and VH3 associates with VL3 to form a third antigen binding site that specifically binds to a third target; wherein the first CH3 domain comprises a substitution with a large hydrophobic amino acid at amino acid position 354 relative to the wild-type CH3 domain and/or the second CH3 domain comprises a substitution with a negatively charged amino acid at amino acid position 347 relative to the wild-type CH3 domain or wherein the second CH3 domain comprises a substitution with a large hydrophobic amino acid at amino acid position 354 relative to the wild-type CH3 domain and/or the first CH3 domain comprises a substitution with a negatively charged amino acid at amino acid position 347 relative to the wild-type CH3 domain; and wherein amino acid residue numbering is based on EU numbering. In some embodiments, the large hydrophobic amino acid is Y, F or W. In some embodiments, the negatively charged amino acid is D or E. In some embodiments, the large hydrophobic amino acid at amino acid position 354 forms a hydrophobic interaction with an amino acid residue in the second CH3 domain. In some embodiments, the second CH3 domain comprises a large hydrophobic residue at amino acid position 349 (e.g., Y349). In some embodiments, the negatively charged amino acid at amino acid position 347 forms an ionic bond with an amino acid residue in the first CH3 domain. In some embodiments, the first CH3 domain comprises a positively charged residue at amino acid position 360 (e.g., K360). In some embodiments, the first and second CH3 domains are a human CH3 domain, a murine CH3 domain, a rat CH3 domain, a camelid CH3 domain, or a rabbit CH3 domain. In some embodiments, the first CH3 domain and the second CH3 domain further comprise KIH residues, such as T366Y and Y407T. In some embodiments, the multispecific antibody is chimeric, human, or humanized. In some embodiments, VL1, VL2, and/or VL3 have the same amino acid sequence. In some embodiments, VL1, VL2, and VL3 have different amino acid sequences.

In some embodiments, multispecific (e.g., bispecific or trispecific) antibodies are provided comprising: (a) a first heavy chain comprising, from N-terminus to C-terminus: a first heavy chain variable domain (VH1), a first CH1, a second heavy chain variable domain (VH2), a second CH1, a first CH2 and a first CH3 domain; (b) a first light chain comprising, from N-terminus to C-terminus: a first light chain variable domain (VL1), a first CL, a second light chain variable domain (VL2), and a second CL; (c) a second heavy chain comprising from N-terminus to C-terminus: a third triple chain variable domain (VH3), a third CH1, a second CH2 and a second CH3 domain; and (d) a second light chain comprising from N-terminus to C-terminus: a third light chain variable domain (VL3) and a third CL; wherein VH1 associates with VL1 to form a first antigen binding site that specifically binds to a first target, VH2 associates with VL2 to form a second antigen binding site that specifically binds to a second target, and VH3 associates with VL3 to form a third antigen binding site that specifically binds to a third target; wherein the first CH3 domain comprises a substitution of the large hydrophobic amino acid for S354 and/or the second CH3 domain comprises a substitution of the negatively charged amino acid for Q347, or wherein the second CH3 domain comprises a substitution of the large hydrophobic amino acid for S354 and/or the first CH3 domain comprises a substitution of the negatively charged amino acid for Q347; and wherein amino acid residue numbering is based on EU numbering. In some embodiments, the large hydrophobic amino acid is Y, F or W. In some embodiments, the negatively charged amino acid is D or E. In some embodiments, the large hydrophobic amino acid at amino acid position 354 forms a hydrophobic interaction with an amino acid residue in the second CH3 domain. In some embodiments, the second CH3 domain comprises a large hydrophobic residue at amino acid position 349 (e.g., Y349). In some embodiments, the negatively charged amino acid at amino acid position 347 forms an ionic bond with an amino acid residue in the first CH3 domain. In some embodiments, the first CH3 domain comprises a positively charged residue at amino acid position 360 (e.g., K360). In some embodiments, the first CH3 domain and the second CH3 domain further comprise KIH residues, such as T366Y and Y407T. In some embodiments, the multispecific antibody is chimeric, human, or humanized. In some embodiments, VL1, VL2, and/or VL3 have the same amino acid sequence. In some embodiments, VL1, VL2, and VL3 have different amino acid sequences.

In some embodiments, multispecific (e.g., bispecific or trispecific) antibodies are provided comprising: (a) a first heavy chain comprising, from N-terminus to C-terminus: a first heavy chain variable domain (VH1), a first CH1, a second heavy chain variable domain (VH2), a second CH1, a first CH2 and a first CH3 domain; (b) a first light chain comprising, from N-terminus to C-terminus: a first light chain variable domain (VL1), a first CL, a second light chain variable domain (VL2), and a second CL; (c) a second heavy chain comprising from N-terminus to C-terminus: a third triple chain variable domain (VH3), a third CH1, a second CH2 and a second CH3 domain; and (d) a second light chain comprising from N-terminus to C-terminus: a third light chain variable domain (VL3) and a third CL; wherein VH1 associates with VL1 to form a first antigen binding site that specifically binds to a first target, VH2 associates with VL2 to form a second antigen binding site that specifically binds to a second target, and VH3 associates with VL3 to form a third antigen binding site that specifically binds to a third target; wherein the first CH3 domain comprises S354Y and T366Y and the second CH3 domain comprises Q347E and Y407T, or wherein the second CH3 domain comprises S354Y and T366Y and the first CH3 domain comprises Q347E and Y407T; and wherein amino acid residue numbering is based on EU numbering. In some embodiments, S354Y of the first CH3 domain forms a hydrophobic interaction with an amino acid residue in the second CH3 domain. In some embodiments, the second CH3 domain comprises a large hydrophobic residue at amino acid position 349 (e.g., Y349). In some embodiments, Q347E of the second CH3 domain forms an ionic bond with an amino acid residue in the first CH3 domain. In some embodiments, the first CH3 domain comprises a positively charged residue at amino acid position 360 (e.g., K360). In some embodiments, the second CH3 domain does not comprise a substitution of Y349, for example Y349S. In some embodiments, the first CH3 and the second CH3 do not comprise further mutations compared to the wild-type human CH3 domain. In some embodiments, the multispecific antibody is chimeric, human, or humanized. In some embodiments, VL1, VL2, and/or VL3 have the same amino acid sequence. In some embodiments, VL1, VL2, and VL3 have different amino acid sequences. In some embodiments, the first and second antigen-binding sites specifically bind to a tumor antigen (e.g., HER2), and the third antigen-binding site specifically binds to CD 3.

In some embodiments, multispecific (e.g., bispecific or trispecific) antibodies are provided comprising: (a) a first heavy chain comprising, from N-terminus to C-terminus: a first heavy chain variable domain (VH1), a first CH1, a second heavy chain variable domain (VH2), a second CH1, a first CH2 and a first CH3 domain; (b) a first light chain comprising, from N-terminus to C-terminus: a first light chain variable domain (VL1), a first CL, a second light chain variable domain (VL2), and a second CL; (c) a second heavy chain comprising from N-terminus to C-terminus: a third triple chain variable domain (VH3), a third CH1, a second CH2 and a second CH3 domain; and (d) a second light chain comprising from N-terminus to C-terminus: a third light chain variable domain (VL3) and a third CL; wherein VH1 associates with VL1 to form a first antigen binding site that specifically binds to a first target, VH2 associates with VL2 to form a second antigen binding site that specifically binds to a second target, and VH3 associates with VL3 to form a third antigen binding site that specifically binds to a third target; wherein the first CH3 domain comprises S354Y and Y407T and the second CH3 domain comprises Q347E and T366Y, or wherein the second CH3 domain comprises S354Y and Y407T and the first CH3 domain comprises Q347E and T366Y; and wherein amino acid residue numbering is based on EU numbering. In some embodiments, S354Y of the first CH3 domain forms a hydrophobic interaction with an amino acid residue in the second CH3 domain. In some embodiments, the second CH3 domain comprises a large hydrophobic residue at amino acid position 349 (e.g., Y349). In some embodiments, Q347E of the second CH3 domain forms an ionic bond with an amino acid residue in the first CH3 domain. In some embodiments, the first CH3 domain comprises a positively charged residue at amino acid position 360 (e.g., K360). In some embodiments, the second CH3 domain does not comprise a substitution of Y349, for example Y349S. In some embodiments, the first CH3 and the second CH3 do not comprise further mutations compared to the wild-type human CH3 domain. In some embodiments, the multispecific antibody is chimeric, human, or humanized. In some embodiments, VL1, VL2, and/or VL3 have the same amino acid sequence. In some embodiments, VL1, VL2, and VL3 have different amino acid sequences. In some embodiments, the first and second antigen-binding sites specifically bind to a tumor antigen (e.g., HER2), and the third antigen-binding site specifically binds to CD 3.

In some embodiments, multispecific (e.g., bispecific or trispecific) antibodies are provided comprising: (a) a first heavy chain comprising, from N-nitrogen terminus to C-terminus: a first VHH domain (VHH1), a second VHH domain (VHH2), a first CH2 and a first CH3 domain; (b) a second heavy chain comprising from N-terminus to C-terminus: a third VHH domain (VHH3), a second CH2 and a second CH3 domain; wherein VHH1 specifically binds to a first target, VHH2 specifically binds to a second target, and VHH3 specifically binds to a third target; wherein the first CH3 domain comprises a substitution with a large hydrophobic amino acid at amino acid position 354 relative to the wild-type CH3 domain and/or the second CH3 domain comprises a substitution with a negatively charged amino acid at amino acid position 347 relative to the wild-type CH3 domain or wherein the second CH3 domain comprises a substitution with a large hydrophobic amino acid at amino acid position 354 relative to the wild-type CH3 domain and/or the first CH3 domain comprises a substitution with a negatively charged amino acid at amino acid position 347 relative to the wild-type CH3 domain; and wherein amino acid residue numbering is based on EU numbering. In some embodiments, the large hydrophobic amino acid is Y, F or W. In some embodiments, the negatively charged amino acid is D or E. In some embodiments, the large hydrophobic amino acid at amino acid position 354 forms a hydrophobic interaction with an amino acid residue in the second CH3 domain. In some embodiments, the second CH3 domain comprises a large hydrophobic residue at amino acid position 349 (e.g., Y349). In some embodiments, the negatively charged amino acid at amino acid position 347 forms an ionic bond with an amino acid residue in the first CH3 domain. In some embodiments, the first CH3 domain comprises a positively charged residue at amino acid position 360 (e.g., K360). In some embodiments, the first and second CH3 domains are a human CH3 domain, a murine CH3 domain, a rat CH3 domain, a camelid CH3 domain, or a rabbit CH3 domain. In some embodiments, the first CH3 domain and the second CH3 domain further comprise KIH residues, such as T366Y and Y407T. In some embodiments, the multispecific antibody is chimeric, human, or humanized.

In some embodiments, multispecific (e.g., bispecific or trispecific) antibodies are provided comprising: (a) a first heavy chain comprising, from N-nitrogen terminus to C-terminus: a first VHH domain (VHH1), a second VHH domain (VHH2), a first CH2 and a first CH3 domain; (b) a second heavy chain comprising from N-terminus to C-terminus: a third VHH domain (VHH3), a second CH2 and a second CH3 domain; wherein VHH1 specifically binds to a first target, VHH2 specifically binds to a second target, and VHH3 specifically binds to a third target; wherein the first CH3 domain comprises a substitution of the large hydrophobic amino acid for S354 and/or the second CH3 domain comprises a substitution of the negatively charged amino acid for Q347, or wherein the second CH3 domain comprises a substitution of the large hydrophobic amino acid for S354 and/or the first CH3 domain comprises a substitution of the negatively charged amino acid for Q347; and wherein amino acid residue numbering is based on EU numbering. In some embodiments, the large hydrophobic amino acid is Y, F or W. In some embodiments, the negatively charged amino acid is D or E. In some embodiments, the large hydrophobic amino acid at amino acid position 354 forms a hydrophobic interaction with an amino acid residue in the second CH3 domain. In some embodiments, the second CH3 domain comprises a large hydrophobic residue at amino acid position 349 (e.g., Y349). In some embodiments, the negatively charged amino acid at amino acid position 347 forms an ionic bond with an amino acid residue in the first CH3 domain. In some embodiments, the first CH3 domain comprises a positively charged residue at amino acid position 360 (e.g., K360). In some embodiments, the first CH3 domain and the second CH3 domain further comprise KIH residues, such as T366Y and Y407T. In some embodiments, the multispecific antibody is chimeric, human, or humanized.

In some embodiments, multispecific (e.g., bispecific or trispecific) antibodies are provided comprising: (a) a first heavy chain comprising, from N-nitrogen terminus to C-terminus: a first VHH domain (VHH1), a second VHH domain (VHH2), a first CH2 and a first CH3 domain; (b) a second heavy chain comprising from N-terminus to C-terminus: a third VHH domain (VHH3), a second CH2 and a second CH3 domain; wherein VHH1 specifically binds to a first target, VHH2 specifically binds to a second target, and VHH3 specifically binds to a third target; wherein the first CH3 domain comprises S354Y and T366Y and the second CH3 domain comprises Q347E and Y407T, or wherein the second CH3 domain comprises S354Y and T366Y and the first CH3 domain comprises Q347E and Y407T; and wherein amino acid residue numbering is based on EU numbering. In some embodiments, S354Y of the first CH3 domain forms a hydrophobic interaction with an amino acid residue in the second CH3 domain. In some embodiments, the second CH3 domain comprises a large hydrophobic residue at amino acid position 349 (e.g., Y349). In some embodiments, Q347E of the second CH3 domain forms an ionic bond with an amino acid residue in the first CH3 domain. In some embodiments, the first CH3 domain comprises a positively charged residue at amino acid position 360 (e.g., K360). In some embodiments, the second CH3 domain does not comprise a substitution of Y349, for example Y349S. In some embodiments, the first CH3 and the second CH3 do not comprise further mutations compared to the wild-type human CH3 domain. In some embodiments, the multispecific antibody is chimeric, human, or humanized.

In some embodiments, multispecific (e.g., bispecific or trispecific) antibodies are provided comprising: (a) a first heavy chain comprising, from N-nitrogen terminus to C-terminus: a first VHH domain (VHH1), a second VHH domain (VHH2), a first CH2 and a first CH3 domain; (b) a second heavy chain comprising from N-terminus to C-terminus: a third VHH domain (VHH3), a second CH2 and a second CH3 domain; wherein VHH1 specifically binds to a first target, VHH2 specifically binds to a second target, and VHH3 specifically binds to a third target; wherein the first CH3 domain comprises S354Y and Y407T and the second CH3 domain comprises Q347E and T366Y, or wherein the second CH3 domain comprises S354Y and Y407T and the first CH3 domain comprises Q347E and T366Y; and wherein amino acid residue numbering is based on EU numbering. In some embodiments, S354Y of the first CH3 domain forms a hydrophobic interaction with an amino acid residue in the second CH3 domain. In some embodiments, the second CH3 domain comprises a large hydrophobic residue at amino acid position 349 (e.g., Y349). In some embodiments, Q347E of the second CH3 domain forms an ionic bond with an amino acid residue in the first CH3 domain. In some embodiments, the first CH3 domain comprises a positively charged residue at amino acid position 360 (e.g., K360). In some embodiments, the second CH3 domain does not comprise a substitution of Y349, for example Y349S. In some embodiments, the first CH3 and the second CH3 do not comprise further mutations compared to the wild-type human CH3 domain. In some embodiments, the multispecific antibody is chimeric, human, or humanized.

In some embodiments, multispecific (e.g., bispecific or trispecific) antibodies are provided comprising: (a) a first heavy chain comprising, from N-terminus to C-terminus: a first VHH domain (VHH1), a second VHH domain (VHH2), a first CH2 and a first CH3 domain; (b) a second heavy chain comprising from N-terminus to C-terminus: a first heavy chain variable domain (VH1), a first CH1, a second CH2 and a second CH3 domain; and (C) a light chain comprising from N-terminus to C-terminus: a first light chain variable domain (VL1) and a first CL; wherein VH1 associates with VL1 to form a first antigen binding site that specifically binds to a first target, VHH1 specifically binds to a second target, and VHH2 specifically binds to a third target; wherein the first CH3 domain comprises a substitution with a large hydrophobic amino acid at amino acid position 354 relative to the wild-type CH3 domain and/or the second CH3 domain comprises a substitution with a negatively charged amino acid at amino acid position 347 relative to the wild-type CH3 domain or wherein the second CH3 domain comprises a substitution with a large hydrophobic amino acid at amino acid position 354 relative to the wild-type CH3 domain and/or the first CH3 domain comprises a substitution with a negatively charged amino acid at amino acid position 347 relative to the wild-type CH3 domain; and wherein amino acid residue numbering is based on EU numbering. In some embodiments, the large hydrophobic amino acid is Y, F or W. In some embodiments, the negatively charged amino acid is D or E. In some embodiments, the large hydrophobic amino acid at amino acid position 354 forms a hydrophobic interaction with an amino acid residue in the second CH3 domain. In some embodiments, the second CH3 domain comprises a large hydrophobic residue at amino acid position 349 (e.g., Y349). In some embodiments, the negatively charged amino acid at amino acid position 347 forms an ionic bond with an amino acid residue in the first CH3 domain. In some embodiments, the first CH3 domain comprises a positively charged residue at amino acid position 360 (e.g., K360). In some embodiments, the first and second CH3 domains are a human CH3 domain, a murine CH3 domain, a rat CH3 domain, a camelid CH3 domain, or a rabbit CH3 domain. In some embodiments, the first CH3 domain and the second CH3 domain further comprise KIH residues, such as T366Y and Y407T. In some embodiments, the multispecific antibody is chimeric, human, or humanized.

In some embodiments, multispecific (e.g., bispecific or trispecific) antibodies are provided comprising: (a) a first heavy chain comprising, from N-terminus to C-terminus: a first VHH domain (VHH1), a second VHH domain (VHH2), a first CH2 and a first CH3 domain; (b) a second heavy chain comprising from N-terminus to C-terminus: a first heavy chain variable domain (VH1), a first CH1, a second CH2 and a second CH3 domain; and (C) a light chain comprising from N-terminus to C-terminus: a first light chain variable domain (VL1) and a first CL; wherein VH1 associates with VL1 to form a first antigen binding site that specifically binds to a first target, VHH1 specifically binds to a second target, and VHH2 specifically binds to a third target; wherein the first CH3 domain comprises a substitution of the large hydrophobic amino acid for S354 and/or the second CH3 domain comprises a substitution of the negatively charged amino acid for Q347, or wherein the second CH3 domain comprises a substitution of the large hydrophobic amino acid for S354 and/or the first CH3 domain comprises a substitution of the negatively charged amino acid for Q347; and wherein amino acid residue numbering is based on EU numbering. In some embodiments, the large hydrophobic amino acid is Y, F or W. In some embodiments, the negatively charged amino acid is D or E. In some embodiments, the large hydrophobic amino acid at amino acid position 354 forms a hydrophobic interaction with an amino acid residue in the second CH3 domain. In some embodiments, the second CH3 domain comprises a large hydrophobic residue at amino acid position 349 (e.g., Y349). In some embodiments, the negatively charged amino acid at amino acid position 347 forms an ionic bond with an amino acid residue in the first CH3 domain. In some embodiments, the first CH3 domain comprises a positively charged residue at amino acid position 360 (e.g., K360). In some embodiments, the first CH3 domain and the second CH3 domain further comprise KIH residues, such as T366Y and Y407T. In some embodiments, the multispecific antibody is chimeric, human, or humanized.

In some embodiments, multispecific (e.g., bispecific or trispecific) antibodies are provided comprising: (a) a first heavy chain comprising, from N-terminus to C-terminus: a first VHH domain (VHH1), a second VHH domain (VHH2), a first CH2 and a first CH3 domain; (b) a second heavy chain comprising from N-terminus to C-terminus: a first heavy chain variable domain (VH1), a first CH1, a second CH2 and a second CH3 domain; and (C) a light chain comprising from N-terminus to C-terminus: a first light chain variable domain (VL1) and a first CL; wherein VH1 associates with VL1 to form a first antigen binding site that specifically binds to a first target, VHH1 specifically binds to a second target, and VHH2 specifically binds to a third target; wherein the first CH3 domain comprises S354Y and T366Y and the second CH3 domain comprises Q347E and Y407T, or wherein the second CH3 domain comprises S354Y and T366Y and the first CH3 domain comprises Q347E and Y407T; and wherein amino acid residue numbering is based on EU numbering. In some embodiments, S354Y of the first CH3 domain forms a hydrophobic interaction with an amino acid residue in the second CH3 domain. In some embodiments, the second CH3 domain comprises a large hydrophobic residue at amino acid position 349 (e.g., Y349). In some embodiments, Q347E of the second CH3 domain forms an ionic bond with an amino acid residue in the first CH3 domain. In some embodiments, the first CH3 domain comprises a positively charged residue at amino acid position 360 (e.g., K360). In some embodiments, the second CH3 domain does not comprise a substitution of Y349, for example Y349S. In some embodiments, the first CH3 and the second CH3 do not comprise further mutations compared to the wild-type human CH3 domain. In some embodiments, the multispecific antibody is chimeric, human, or humanized. In some embodiments, VHH1 and VHH2 specifically bind to a tumor antigen (e.g., BCMA) and the first antigen binding site specifically binds CD 3.

In some embodiments, multispecific (e.g., bispecific or trispecific) antibodies are provided comprising: (a) a first heavy chain comprising, from N-terminus to C-terminus: a first VHH domain (VHH1), a second VHH domain (VHH2), a first CH2 and a first CH3 domain; (b) a second heavy chain comprising from N-terminus to C-terminus: a first heavy chain variable domain (VH1), a first CH1, a second CH2 and a second CH3 domain; and (C) a light chain comprising from N-terminus to C-terminus: a first light chain variable domain (VL1) and a first CL; wherein VH1 associates with VL1 to form a first antigen binding site that specifically binds to a first target, VHH1 specifically binds to a second target, and VHH2 specifically binds to a third target; wherein the first CH3 domain comprises S354Y and Y407T and the second CH3 domain comprises Q347E and T366Y, or wherein the second CH3 domain comprises S354Y and Y407T and the first CH3 domain comprises Q347E and T366Y; and wherein amino acid residue numbering is based on EU numbering. In some embodiments, S354Y of the first CH3 domain forms a hydrophobic interaction with an amino acid residue in the second CH3 domain. In some embodiments, the second CH3 domain comprises a large hydrophobic residue at amino acid position 349 (e.g., Y349). In some embodiments, Q347E of the second CH3 domain forms an ionic bond with an amino acid residue in the first CH3 domain. In some embodiments, the first CH3 domain comprises a positively charged residue at amino acid position 360 (e.g., K360). In some embodiments, the second CH3 domain does not comprise a substitution of Y349, for example Y349S. In some embodiments, the first CH3 and the second CH3 do not comprise further mutations compared to the wild-type human CH3 domain. In some embodiments, the multispecific antibody is chimeric, human, or humanized. In some embodiments, VHH1 and VHH2 specifically bind to a tumor antigen (e.g., BCMA) and the first antigen binding site specifically binds CD 3.

In some embodiments, multispecific (e.g., bispecific, trispecific, or tetraspecific) antibodies are provided comprising: (a) a first heavy chain comprising, from N-terminus to C-terminus: a first VHH domain (VHH1), a second VHH domain (VHH2), a first CH2 and a first CH3 domain; (b) a second heavy chain comprising from N-terminus to C-terminus: a third VHH domain (VHH3), a fourth VHH domain (VHH4), a second CH2 and a second CH3 domain; wherein VHH1 specifically binds to a first target, VHH2 specifically binds to a second target, VHH3 specifically binds to a third target, and VHH4 specifically binds to a fourth target; wherein the first CH3 domain comprises a substitution with a large hydrophobic amino acid at amino acid position 354 relative to the wild-type CH3 domain and/or the second CH3 domain comprises a substitution with a negatively charged amino acid at amino acid position 347 relative to the wild-type CH3 domain, and wherein amino acid residue numbering is based on EU numbering. In some embodiments, the large hydrophobic amino acid is Y, F or W. In some embodiments, the negatively charged amino acid is D or E. In some embodiments, the large hydrophobic amino acid at amino acid position 354 forms a hydrophobic interaction with an amino acid residue in the second CH3 domain. In some embodiments, the second CH3 domain comprises a large hydrophobic residue at amino acid position 349 (e.g., Y349). In some embodiments, the negatively charged amino acid at amino acid position 347 forms an ionic bond with an amino acid residue in the first CH3 domain. In some embodiments, the first CH3 domain comprises a positively charged residue at amino acid position 360 (e.g., K360). In some embodiments, the first and second CH3 domains are a human CH3 domain, a murine CH3 domain, a rat CH3 domain, a camelid CH3 domain, or a rabbit CH3 domain. In some embodiments, the first CH3 domain and the second CH3 domain further comprise KIH residues, such as T366Y and Y407T. In some embodiments, the multispecific antibody is chimeric, human, or humanized.

In some embodiments, multispecific (e.g., bispecific, trispecific, or tetraspecific) antibodies are provided comprising: (a) a first heavy chain comprising, from N-terminus to C-terminus: a first VHH domain (VHH1), a second VHH domain (VHH2), a first CH2 and a first CH3 domain; (b) a second heavy chain comprising from N-terminus to C-terminus: a third VHH domain (VHH3), a fourth VHH domain (VHH4), a second CH2 and a second CH3 domain; wherein VHH1 specifically binds to a first target, VHH2 specifically binds to a second target, VHH3 specifically binds to a third target, and VHH4 specifically binds to a fourth target; wherein the first CH3 domain comprises a substitution of a large hydrophobic amino acid for S354 and/or the second CH3 domain comprises a substitution of a negatively charged amino acid for Q347, and wherein the amino acid residue numbering is based on EU numbering. In some embodiments, the large hydrophobic amino acid is Y, F or W. In some embodiments, the negatively charged amino acid is D or E. In some embodiments, the large hydrophobic amino acid at amino acid position 354 forms a hydrophobic interaction with an amino acid residue in the second CH3 domain. In some embodiments, the second CH3 domain comprises a large hydrophobic residue at amino acid position 349 (e.g., Y349). In some embodiments, the negatively charged amino acid at amino acid position 347 forms an ionic bond with an amino acid residue in the first CH3 domain. In some embodiments, the first CH3 domain comprises a positively charged residue at amino acid position 360 (e.g., K360). In some embodiments, the first CH3 domain and the second CH3 domain further comprise KIH residues, such as T366Y and Y407T. In some embodiments, the multispecific antibody is chimeric, human, or humanized.

In some embodiments, multispecific (e.g., bispecific, trispecific, or tetraspecific) antibodies are provided comprising: (a) a first heavy chain comprising, from N-terminus to C-terminus: a first VHH domain (VHH1), a second VHH domain (VHH2), a first CH2 and a first CH3 domain; (b) a second heavy chain comprising from N-terminus to C-terminus: a third VHH domain (VHH3), a fourth VHH domain (VHH4), a second CH2 and a second CH3 domain; wherein VHH1 specifically binds to a first target, VHH2 specifically binds to a second target, VHH3 specifically binds to a third target, and VHH4 specifically binds to a fourth target; wherein the first CH3 domain comprises S354Y and T366Y, the second CH3 domain comprises Q347E and Y407T, and wherein amino acid residue numbering is based on EU numbering. In some embodiments, S354Y of the first CH3 domain forms a hydrophobic interaction with an amino acid residue in the second CH3 domain. In some embodiments, the second CH3 domain comprises a large hydrophobic residue at amino acid position 349 (e.g., Y349). In some embodiments, Q347E of the second CH3 domain forms an ionic bond with an amino acid residue in the first CH3 domain. In some embodiments, the first CH3 domain comprises a positively charged residue at amino acid position 360 (e.g., K360). In some embodiments, the second CH3 domain does not comprise a substitution of Y349, for example Y349S. In some embodiments, the first CH3 and the second CH3 do not comprise further mutations compared to the wild-type human CH3 domain. In some embodiments, the multispecific antibody is chimeric, human, or humanized.

In some embodiments, multispecific (e.g., bispecific, trispecific, or tetraspecific) antibodies are provided comprising: (a) a first heavy chain comprising, from N-terminus to C-terminus: a first VHH domain (VHH1), a second VHH domain (VHH2), a first CH2 and a first CH3 domain; (b) a second heavy chain comprising from N-terminus to C-terminus: a third VHH domain (VHH3), a fourth VHH domain (VHH4), a second CH2 and a second CH3 domain; wherein VHH1 specifically binds to a first target, VHH2 specifically binds to a second target, VHH3 specifically binds to a third target, and VHH4 specifically binds to a fourth target; wherein the first CH3 domain comprises S354Y and Y407T, the second CH3 domain comprises Q347E and T366Y, and wherein amino acid residue numbering is based on EU numbering. In some embodiments, S354Y of the first CH3 domain forms a hydrophobic interaction with an amino acid residue in the second CH3 domain. In some embodiments, the second CH3 domain comprises a large hydrophobic residue at amino acid position 349 (e.g., Y349). In some embodiments, Q347E of the second CH3 domain forms an ionic bond with an amino acid residue in the first CH3 domain. In some embodiments, the first CH3 domain comprises a positively charged residue at amino acid position 360 (e.g., K360). In some embodiments, the second CH3 domain does not comprise a substitution of Y349, for example Y349S. In some embodiments, the first CH3 and the second CH3 do not comprise further mutations compared to the wild-type human CH3 domain. In some embodiments, the multispecific antibody is chimeric, human, or humanized.

Also provided are multispecific antibodies comprising any one of the first CH3 domain and the second CH3 domain described herein. In some embodiments, the present application provides a bispecific T cell adaptor (BiTE) molecule comprising a first antigen-binding fragment that specifically binds a tumor antigen, a second antigen-binding fragment that specifically binds CD3, and any one of the mutant Fc regions described herein. In some embodiments, the present application provides a bispecific T cell adaptor (BiTE) molecule comprising first and second antigen-binding fragments that specifically bind to a tumor antigen, a third antigen-binding fragment that specifically binds to CD3, and any one of the mutant Fc regions described herein. In some embodiments, the present application provides multispecific (e.g., bispecific or trispecific) antibodies that specifically bind CD20 and CD 3. In some embodiments, the present application provides bispecific antibodies that specifically bind HER2 and CD 3. In some embodiments, the present application provides multispecific (e.g., bispecific or trispecific) antibodies that specifically bind CD3 and BCMA. Any suitable antigen-binding fragment may be used for the multispecific antibodies described herein, including, for example, anti-CD 20 and anti-CD 3 antigen-binding fragments described in international application No. PCT/US2018/044778 or international application No. PCT/US 2020/015311.

In some embodiments, the heteromultimeric protein is a heteromultimeric immunoadhesin. Immunoadhesins are antibody-like molecules that combine the binding domain or ligand of a protein (such as a cell surface receptor) ("adhesin") with the effector functions of immunoglobulin constant domains. Immunoadhesins can possess many of the valuable chemical and biological properties of human antibodies. Since immunoadhesins can be constructed from human protein sequences of desired specificity linked to appropriate human immunoglobulin hinge and constant domain (Fc) sequences, the human component can be used entirely to achieve the target binding specificity. In some embodiments, the heteromultimeric protein is a multispecific immunoadhesin, e.g., a bispecific immunoadhesin, i.e., the two arms of the immunoadhesin have different specificities.

In some embodiments, the heteromultimeric protein is an antibody-immunoadhesin chimera. These molecules combine the binding region of the immunoadhesin with the binding domain of an antibody. Exemplary antibody-immunoadhesin chimeras have been described, for example, in Berg et al, PNAS (USA)88: 4723-.

In some embodiments, the heteromultimeric protein is an immunoadhesin or antibody-immunoadhesin chimera comprising one or more binding domains of a non-antibody fragment, such as a ligand binding domain, a receptor binding domain, or an enzyme domain. As used herein, the term "ligand binding domain" refers to any native cell surface receptor or any region or derivative thereof (which at least retains qualitative ligand binding capability, preferably the biological activity of the corresponding native receptor). In some embodiments, the receptor is from a cell surface polypeptide having an extracellular domain homologous to a member of the immunoglobulin superfamily. Other typical receptors are not members of the immunoglobulin superfamily, but are still specifically covered by this definition, being cytokine receptors, receptors with tyrosine kinase activity (receptor tyrosine kinases), members of the hematopoeitin and nerve growth factor receptor superfamily and cell adhesion molecules, e.g., (E-, L-and P-) selectins.

The term "receptor binding domain" refers to any natural ligand of a receptor, including a cell adhesion molecule, or any region or derivative of such a natural ligand (which at least retains qualitative receptor binding capacity, preferably the biological activity of the corresponding natural ligand). This definition specifically includes, among other things, binding sequences for ligands from the above-mentioned receptors.

In some embodiments, a multispecific (e.g., bispecific) immunoadhesin is provided comprising: (a) a first polypeptide comprising, from N-terminus to C-terminus: a first binding domain that specifically binds to a first target, a first CH2 domain, and a first CH3 domain; (b) a second polypeptide comprising from N-terminus to C-terminus: a second binding domain that specifically binds a second target, a second CH2 domain, and a second CH3 domain; wherein the first CH3 domain comprises a substitution with a large hydrophobic amino acid at amino acid position 354 relative to the wild-type CH3 domain and/or the second CH3 domain comprises a substitution with a negatively charged amino acid at amino acid position 347 relative to the wild-type CH3 domain, and wherein amino acid residue numbering is based on EU numbering. In some embodiments, the large hydrophobic amino acid is Y, F or W. In some embodiments, the negatively charged amino acid is D or E. In some embodiments, the large hydrophobic amino acid at amino acid position 354 forms a hydrophobic interaction with an amino acid residue in the second CH3 domain. In some embodiments, the second CH3 domain comprises a large hydrophobic residue at amino acid position 349 (e.g., Y349). In some embodiments, the negatively charged amino acid at amino acid position 347 forms an ionic bond with an amino acid residue in the first CH3 domain. In some embodiments, the first CH3 domain comprises a positively charged residue at amino acid position 360 (e.g., K360). In some embodiments, the first and second CH3 domains are a human CH3 domain, a murine CH3 domain, a rat CH3 domain, a camelid CH3 domain, or a rabbit CH3 domain. In some embodiments, the first CH3 domain and the second CH3 domain further comprise KIH residues, such as T366Y and Y407T. In some embodiments, the first binding domain and the second binding domain are receptor binding domains. In some embodiments, the first binding domain and the second binding domain are ligand binding domains. In some embodiments, the first binding domain is a receptor binding domain and the second binding domain is a ligand binding domain, or the first binding domain is a ligand binding domain and the second binding domain is a receptor binding domain.

In some embodiments, a multispecific (e.g., bispecific) immunoadhesin is provided comprising: (a) a first polypeptide comprising, from N-terminus to C-terminus: a first binding domain that specifically binds to a first target, a first CH2 domain, and a first CH3 domain; (b) a second polypeptide comprising from N-terminus to C-terminus: a second binding domain that specifically binds a second target, a second CH2 domain, and a second CH3 domain; wherein the first CH3 domain comprises a substitution of a large hydrophobic amino acid for S354 and/or the second CH3 domain comprises a substitution of a negatively charged amino acid for Q347, and wherein the amino acid residue numbering is based on EU numbering. In some embodiments, the large hydrophobic amino acid is Y, F or W. In some embodiments, the negatively charged amino acid is D or E. In some embodiments, the large hydrophobic amino acid at amino acid position 354 forms a hydrophobic interaction with an amino acid residue in the second CH3 domain. In some embodiments, the second CH3 domain comprises a large hydrophobic residue at amino acid position 349 (e.g., Y349). In some embodiments, the negatively charged amino acid at amino acid position 347 forms an ionic bond with an amino acid residue in the first CH3 domain. In some embodiments, the first CH3 domain comprises a positively charged residue at amino acid position 360 (e.g., K360). In some embodiments, the first CH3 domain and the second CH3 domain further comprise KIH residues, such as T366Y and Y407T. In some embodiments, the first binding domain and the second binding domain are receptor binding domains. In some embodiments, the first binding domain and the second binding domain are ligand binding domains. In some embodiments, the first binding domain is a receptor binding domain and the second binding domain is a ligand binding domain, or the first binding domain is a ligand binding domain and the second binding domain is a receptor binding domain.

In some embodiments, a multispecific (e.g., bispecific) immunoadhesin is provided comprising: (a) a first polypeptide comprising, from N-terminus to C-terminus: a first binding domain that specifically binds to a first target, a first CH2 domain, and a first CH3 domain; (b) a second polypeptide comprising from N-terminus to C-terminus: a second binding domain that specifically binds a second target, a second CH2 domain, and a second CH3 domain; wherein: (i) the first CH3 domain comprises S354Y and T366Y, the second CH3 domain comprises Q347E and Y407T, or (ii) the first CH3 domain comprises S354Y and Y407T, the second CH3 domain comprises Q347E and T366Y, and wherein the amino acid residue numbering is based on EU numbering. In some embodiments, S354Y of the first CH3 domain forms a hydrophobic interaction with an amino acid residue in the second CH3 domain. In some embodiments, the second CH3 domain comprises a large hydrophobic residue at amino acid position 349 (e.g., Y349). In some embodiments, Q347E of the second CH3 domain forms an ionic bond with an amino acid residue in the first CH3 domain. In some embodiments, the first CH3 domain comprises a positively charged residue at amino acid position 360 (e.g., K360). In some embodiments, the second CH3 domain does not comprise a substitution of Y349, for example Y349S. In some embodiments, the first CH3 and the second CH3 do not comprise further mutations compared to the wild-type human CH3 domain. In some embodiments, the first binding domain and the second binding domain are receptor binding domains. In some embodiments, the first binding domain and the second binding domain are ligand binding domains. In some embodiments, the first binding domain is a receptor binding domain and the second binding domain is a ligand binding domain, or the first binding domain is a ligand binding domain and the second binding domain is a receptor binding domain.

The multispecific antibodies and multispecific immunoadhesins described herein can specifically bind to any suitable combination of epitopes, antigens, or target molecules. In some embodiments, the first target, the second target, the third target, and the fourth target are the same epitope. In some embodiments, the first target, the second target, the third target, and/or the fourth target are different epitopes of the same antigen. In some embodiments, the first target, the second target, the third target, and the fourth target are different antigens. In some embodiments, the first target, the second target, the third target, and the fourth target are different target molecules.

In some embodiments, the first target, the second target, the third target, and/or the fourth target are cell surface molecules. In some embodiments, the first target, the second target, the third target, and/or the fourth target are tumor antigens. Tumor antigens are proteins produced by tumor cells that can elicit an immune response, particularly a T cell-mediated immune response. The choice of the targeted antigen of the invention will depend on the particular type of cancer to be treated. Exemplary tumor antigens include, for example, glioma-associated antigen, carcinoembryonic antigen (CEA), β -human chorionic gonadotropin, alpha-fetoprotein (AFP), lectin-reactive AFP, thyroglobulin, RAGE-1, MN-CAIX, human telomerase reverse transcriptase, RU1, RU2(AS), intestinal carboxyesterase, mut hsp70-2, M-CSF, prostatase, Prostate Specific Antigen (PSA), PAP, NY-ESO-1, LAGE-la, p53, prostaglandins, PSMA, HER2/neu, survivin and telomerase, prostate cancer tumor antigen-1 (PCTA-1), MAGE, ELF2M, neutrophil elastase, ephrinB2, CD22, insulin growth factor (IGF-I), IGF-II, IGF-I receptor, and mesothelin.

In some embodiments, the tumor antigen comprises one or more antigenic cancer epitopes associated with a malignancy. Malignant tumors express a number of proteins that can serve as target antigens for immune attack. These molecules include, but are not limited to, tissue-specific antigens such as MART-1, tyrosinase and gp100 in melanoma and Prostatic Acid Phosphatase (PAP) and prostate-specific antigen (PSA) in prostate cancer. Other target molecules belong to the group of transformation-related molecules such as the oncogene HER 2/Neu/ErbB-2. Another group of target antigens are the onco-fetal antigens (onco-total antigen), such as carcinoembryonic antigen (CEA). In B-cell lymphomas, tumor-specific idiotypic immunoglobulins constitute a true tumor-specific immunoglobulin antigen that is unique to an individual's tumor. B cell differentiation antigens such as CD 19, CD20, and CD37 are other candidates as target antigens in B cell lymphomas.

In some embodiments, the tumor antigen is a Tumor Specific Antigen (TSA) or a Tumor Associated Antigen (TAA). TSA is unique to tumor cells and does not occur on other cells of the body. TAA is not unique to tumor cells, but instead it is also expressed on normal cells under conditions that do not induce an immune-tolerant state to the antigen. Expression of the antigen on the tumor may occur under conditions that enable the immune system to react to the antigen. TAAs may be antigens expressed on normal cells during fetal development when the immune system is immature and unable to respond, or they may be antigens that are normally present at very low levels on normal cells but are expressed at much higher levels on tumor cells.

Non-limiting examples of TSA or TAA antigens include the following: differentiation antigens such as MART-1/Melana (MART-I), gp100 (Pmel 17), tyrosinase, TRP-1, TRP-2, and tumor-specific multispectral antigens such as MAGE-1, MAGE-3, BAGE, GAGE-1, GAGE-2, pl 5; overexpressed embryonic antigens such as CEA; overexpressed oncogenes such as p53, Ras, HER2/neu, and mutated tumor suppressor genes; unique tumor antigens resulting from chromosomal translocations such as BCR-ABL, E2A-PRL, H4-RET, IGH-IGK, MYL-RAR; and viral antigens such as the epstein-barr virus antigen EBVA and the Human Papilloma Virus (HPV) antigens E6 and E7. Other protein-based macroantigens include TSP-180, MAGE-4, MAGE-5, MAGE-6, RAGE, NY-ESO, pl85erbB2, pl80erbB-3, C-met, nm-23HI, PSA, TAG-72, CA 19-9, CA 72-4, CAM 17.1, NuMa, K-ras, β -catenin, CDK4, Mum-1, P15, P16, 43-9F, 5T4, 791T 72, alpha fetoprotein, β -HCG, BCA225, BTA, CA 125, CA 15-3\ CA 27.29\ BCAA, CA 195, CA 242, CA-50, CAM43, CD68\ P1, CO-029, Ga-5, G250, Ga733 CAM, gp CAM 175-MG, MG 175-MG 50, EpMA-50, MOV 28-50, SDC-70, MOV-23, MCAS-C-1, CG493-35, CGTCK-4, CGTCK-13, CGTCK-9, CGTCK-6, CGTCK-3, CGTCK, and TCK-9, and TCK-3, and TCK 23, and TCK-7, TAAL6, TAG72, TLP and TPS.

In some embodiments, the first target, the second target, the third target, and/or the fourth target are immune checkpoint molecules, such as stimulatory immune checkpoint molecules or inhibitory immune checkpoint molecules. Exemplary stimulatory immune checkpoint molecules include, but are not limited to, CD28, OX40, ICOS, GITR, 4-1BB, CD27, CD40, CD3, HVEM, and TCR (e.g., MHC class I or class II molecules). Exemplary inhibitory immune checkpoint molecules include, but are not limited to, CTLA-4, TIM-3, A2a receptor, LAG-3, BTLA, KIR, PD-1, IDO, CD47 and ligands thereof, such as B7.1, B7.2, PD-L1, PD-L2, HVEM, B7-H4, NKTR-218 and SIRP-alpha receptor.

In some embodiments, the first target, the second target, the third target, and/or the fourth target are antigens on immune effector cells (such as T cells, B cells, macrophages, or natural killer cells). In some embodiments, the first, second, third, or fourth target is CD 3. In some embodiments, the first target is CD3 and the second target is a tumor antigen, or the first target is a tumor antigen and the second target is CD 3. In some embodiments, the first target is CD3, and the second and third targets are tumor antigens.

Also provided is a single polypeptide of any of the heteromultimeric proteins described herein.

In some embodiments, a polypeptide comprising an antibody CH3 domain is provided, wherein the CH3 domain comprises a substitution with a large hydrophobic amino acid at amino acid position 354 relative to the wild-type CH3 domain and/or a substitution with a negatively charged amino acid at amino acid position 347 relative to the wild-type CH3 domain, and wherein the polypeptide has a reduced ability to form a homodimer as compared to a polypeptide comprising the wild-type CH3 domain. In some embodiments, the large hydrophobic amino acid is Y, F or W. In some embodiments, the negatively charged amino acid is D or E. In some embodiments, the CH3 domain is a human CH3 domain, a murine CH3 domain, a rat CH3 domain, a camelid CH3 domain, or a rabbit CH3 domain. In some embodiments, the CH3 domain further comprises a KIH residue, such as T366Y or Y407T. In some embodiments, the polypeptide comprises a CH2 domain. In some embodiments, the polypeptide comprises an antibody heavy chain.

In some embodiments, polypeptides are provided comprising a human antibody CH3 domain, wherein the CH3 domain comprises a substitution of a large hydrophobic amino acid for S354 and/or a substitution of a negatively charged amino acid for Q347, and wherein the polypeptide has a reduced ability to form a homodimer as compared to a polypeptide comprising a wild-type CH3 domain. In some embodiments, the CH3 domain comprises a substitution selected from the group consisting of: S354Y, S354F and S354W. In some embodiments, the CH3 domain comprises a substitution selected from the group consisting of: Q347E and Q347D. In some embodiments, the CH3 domain further comprises a KIH residue, such as T366Y or Y407T. In some embodiments, the polypeptide comprises a CH2 domain. In some embodiments, the polypeptide comprises an antibody heavy chain.

Also provided herein are variants and derivatives of any of the aforementioned heteromultimeric proteins or polypeptides.

In some embodiments, amino acid sequence variants of the heteromultimeric proteins (e.g., multispecific antibodies) provided herein are contemplated. For example, it may be desirable to increase the binding affinity and/or other biological properties of the heteromultimeric protein. Amino acid sequence variants of heteromultimeric proteins can be prepared by introducing appropriate modifications into the nucleotide sequence encoding the heteromultimeric protein or by peptide synthesis. Such modifications include, for example, deletions from and/or insertions into and/or substitutions of residues within the amino acid sequence of the heteromultimeric protein. Any combination of deletions, insertions, and substitutions can be made to arrive at the final construct, provided that the final construct possesses the desired properties, e.g., antigen binding.

In some embodiments, heteromultimeric protein variants having one or more amino acid substitutions are provided. The target sites for substitution mutagenesis include the CDRs and FRs of multispecific antibodies. Amino acid substitutions may be introduced into the multispecific antibody of interest and the product screened for a desired activity, e.g., maintaining/increasing antigen binding and lysis, reducing immunogenicity, or improving ADCC or CDC.

Conservative substitutions are shown in table 2 below.

Table 2: conservative substitutions

Amino acids can be classified into different classes according to common side chain properties:

a. and (3) hydrophobic: norleucine, Met, Ala, Val, Leu, Ile

b. Neutral hydrophilicity: cys, Ser, Thr, Asn, Gln

c. Acidity: asp and Glu

d. Alkalinity: his, Lys, Arg

e. Residues that influence chain orientation: gly, Pro

f. Aromatic: trp, Tyr, Phe.

Non-conservative substitutions will entail replacing one of these classes with a member of the other class.

In some embodiments, substitutions, insertions, or deletions may occur within one or more CDRs so long as such alterations do not substantially reduce the ability of the multispecific antibody to bind antigen. For example, conservative changes (e.g., conservative substitutions as provided herein) may be made to the CDRs that do not substantially reduce binding affinity. Such changes may be outside of HVR "hotspots" or SDRs.

One method that can be used to identify residues or regions of an antibody that can be targeted for mutagenesis is referred to as "alanine scanning mutagenesis" as described by Cunningham and Wells (1989) Science,244: 1081-1085. In this method, a residue or group of target residues (e.g., charged residues such as arg, asp, his, lys, and glu) are identified and replaced with a neutral or negatively charged amino acid (e.g., alanine or polyalanine) to determine whether the interaction of the antibody with the antigen is affected. Further substitutions may be introduced at amino acid positions that show functional sensitivity to the initial substitution. Alternatively or additionally, the crystal structure of the antigen-antibody complex may be determined to identify the contact points between the antibody and the antigen. Such contact residues and adjacent residues may be targeted or eliminated as candidates for substitution. Variants can be screened to determine if they contain the desired property.

Amino acid sequence insertions include amino-and/or carboxy-terminal fusions ranging in length from one residue to polypeptides containing one hundred or more residues, as well as intrasequence insertions of single or multiple amino acid residues. Examples of terminal insertions include heteromultimeric proteins with an N-terminal methionyl residue. Other insertional variants of the heteromultimeric protein include fusion of the N-or C-terminus of the heteromultimeric protein with an enzyme (e.g., for ADEPT) or a polypeptide that increases the serum half-life of the heteromultimeric protein.

Heteromultimeric protein variants are also provided with amino-terminal leader extensions. For example, one or more amino acid residues of the amino-terminal leader sequence are present on the amino terminus of any one or more of the heavy or light chains of the antibody.

Covalent modification of heteromultimeric proteins is also included within the scope of the invention. Covalent modifications of heteromultimeric proteins can be introduced into molecules by reacting targeted amino acid residues of the heteromultimeric proteins or fragments thereof with organic derivatizing agents capable of reacting with selected side chains or N-or C-terminal residues. Another type of covalent modification of heteromultimeric proteins involves altering the native glycosylation pattern of the polypeptide. For example, one or more carbohydrate moieties found in the original heteromultimeric protein can be deleted and/or one or more glycosylation sites not present in the original heteromultimeric protein can be added. The addition of glycosylation sites to heteromultimeric proteins can be conveniently accomplished by altering the amino acid sequence to include one or more N-linked glycosylation sites. The alteration may also be made by adding or substituting one or more serine or threonine residues to the original heteromultimeric protein sequence (for O-linked glycosylation sites). For example, the amino acid sequence of a heteromultimeric protein can be altered by changes at the DNA level, e.g., by mutating the DNA encoding the heteromultimeric protein at preselected bases such that codons are generated that will translate into the desired amino acids. Another method of increasing the number of carbohydrate moieties on a heteromultimeric protein is by chemical or enzymatic coupling of a glycoside to a polypeptide. These methods are described in WO 87/05330 and Aplin and Wriston, CRC Crit. Rev. biochem, pp 259-306 (1981). Removal of the carbohydrate moieties present on the heteromultimeric protein can be accomplished chemically or enzymatically.

Another type of covalent modification of heteromultimeric proteins involves linking the heteromultimeric proteins to a variety of non-protein properties

One of the sections. Moieties suitable for heteromultimeric protein derivatization include, but are not limited to, water soluble polymers. Non-limiting examples of water-soluble polymers include, but are not limited to, polyethylene glycol (PEG), copolymers of ethylene glycol/propylene glycol, carboxymethylcellulose, dextran, polyvinyl alcohol, polyvinylpyrrolidone, poly-1, 3-dioxolane, poly-1, 3, 6-trioxane, ethylene/maleic anhydride copolymers, polyaminoacids (homopolymers or random copolymers) and dextran or poly (n-vinyl pyrrolidone) polyethylene glycol, propylene glycol homopolymers, propylene oxide/ethylene oxide copolymers, polyoxyethylene polyols (e.g., glycerol), polyvinyl alcohol, and mixtures thereof. Polyethylene glycol propionaldehyde may have advantages in production due to its stability in water. The polymer may have any molecular weight and may be branched or unbranched. The number of polymers attached to the heteromultimeric protein can vary, and if more than one polymer is attached, they can be the same or different molecules. In general, the number and/or type of polymers used for derivatization can be determined based on considerations including, but not limited to, the particular properties or functions of the heteromultimeric protein to be improved, whether the heteromultimeric protein derivative will be used in determining therapy under conditions, and the like.

Preparation method

The present application also provides methods of making heteromultimeric (e.g., heterodimeric) proteins, such as multispecific antibodies or immunoadhesins. Nucleic acids, vectors, and host cells for making heteromultimeric proteins or polypeptides thereof are also provided.

The heteromultimeric proteins described herein can be prepared using any method known in the art, including the methods described below and in the examples. Such methods may comprise culturing a host cell comprising a nucleic acid encoding a first and second CH 3-containing polypeptide such that the polypeptides are co-expressed by the cell. In certain embodiments, the nucleic acids encoding the first and second CH 3-containing polypeptides are provided to the host cell in any ratio of, for example, about 1:1, 1:2, 2:1, 1:3, 3:1, 1:4, 4:1, 1:5, 5:1, 1:6, 6:1, 1:7, 7:1, 1:8, 8:1, 1:9, 9:1, 1:10, or 10:1 (mole: mole). In some embodiments, the heteromultimeric protein comprises one or more antibody light chains. In some embodiments, the heteromultimeric protein comprises a first heavy chain, a second heavy chain, and a common light chain co-expressed by a cell. In some embodiments, the nucleic acid encoding the common light chain and the nucleic acid encoding the first or second heavy chain are provided to the host cell in a ratio of at least any one of about 1:1, 2:1, 3:1, 4:1, 5:1, 6:1, 7:1, 8:1, 9:1, or 10: 1. In some embodiments, the nucleic acid encoding the first heavy chain, the nucleic acid encoding the second heavy chain, and the nucleic acid encoding the common light chain are provided to the host cell in a ratio of about 1: 5. It is contemplated that varying the ratio of nucleic acids may increase the yield of heterodimeric molecules relative to homodimeric molecules.

In some embodiments, there is provided a method of producing a heteromultimeric protein that specifically binds a first target and a second target, the method comprising: (a) providing a first polypeptide comprising a first binding domain that specifically binds to a first target and a first CH3 domain; and (b) providing a second polypeptide comprising a second binding domain that specifically binds to a second target and a second CH3 domain; wherein: (i) the first CH3 domain comprises a substitution with a large hydrophobic amino acid at amino acid position 354 relative to the wild-type CH3 domain and/or the second CH3 domain comprises a substitution with a negatively charged amino acid at amino acid position 347 relative to the wild-type CH3 domain; or (ii) the first CH3 domain comprises a substitution with a negatively charged amino acid at amino acid position 347 relative to the wild-type CH3 domain and/or the second CH3 domain comprises a substitution with a large hydrophobic amino acid at amino acid position 354 relative to the wild-type CH3 domain; and wherein amino acid residue numbering is based on EU numbering. In some embodiments, the large hydrophobic amino acid is Y, F or W. In some embodiments, the negatively charged amino acid is D or E. In some embodiments, the large hydrophobic amino acid at amino acid position 354 forms a hydrophobic interaction with an amino acid residue in the second CH3 domain. In some embodiments, the second CH3 domain comprises a large hydrophobic residue at amino acid position 349 (e.g., Y349). In some embodiments, the negatively charged amino acid at amino acid position 347 forms an ionic bond with an amino acid residue in the first CH3 domain. In some embodiments, the first CH3 domain comprises a positively charged residue at amino acid position 360 (e.g., K360). In some embodiments, the first and second CH3 domains are human CH3 domains. In some embodiments, the substitution at amino acid position 354 is S354Y. In some embodiments, the substitution at amino acid position 347 is Q347E. In some embodiments, the first CH3 domain and the second CH3 domain further comprise a KIH residue, such as T366Y or Y407T.

In some embodiments, there is provided a method of generating a multispecific antibody that specifically binds a first target and a second target, the method comprising: (a) providing a first heavy chain comprising, from N-terminus to C-terminus: a VH1, a first CH1, a first CH2, and a first CH3 domain; (b) providing a first light chain comprising, from N-terminus to C-terminus: VL1 and CL; (c) a second heavy chain comprising from N-terminus to C-terminus: a VH2, a second CH1, a second CH2, and a second CH3 domain; and (d) a second light chain comprising from N-terminus to C-terminus: VL2 and a second CL; wherein VH1 associates with VL1 to form a first antigen binding site that specifically binds to a first target, and VH2 associates with VL2 to form a second antigen binding site that specifically binds to a second target; wherein: (i) the first CH3 domain comprises a substitution with a large hydrophobic amino acid at amino acid position 354 relative to the wild-type CH3 domain and/or the second CH3 domain comprises a substitution with a negatively charged amino acid at amino acid position 347 relative to the wild-type CH3 domain; or (ii) the first CH3 domain comprises a substitution with a negatively charged amino acid at amino acid position 347 relative to the wild-type CH3 domain and/or the second CH3 domain comprises a substitution with a large hydrophobic amino acid at amino acid position 354 relative to the wild-type CH3 domain; and wherein amino acid residue numbering is based on EU numbering. In some embodiments, the large hydrophobic amino acid is Y, F or W. In some embodiments, the negatively charged amino acid is D or E. In some embodiments, the large hydrophobic amino acid at amino acid position 354 forms a hydrophobic interaction with an amino acid residue in the second CH3 domain. In some embodiments, the second CH3 domain comprises a large hydrophobic residue at amino acid position 349 (e.g., Y349). In some embodiments, the negatively charged amino acid at amino acid position 347 forms an ionic bond with an amino acid residue in the first CH3 domain. In some embodiments, the first CH3 domain comprises a positively charged residue at amino acid position 360 (e.g., K360). In some embodiments, the first and second CH3 domains are human CH3 domains. In some embodiments, the substitution at amino acid position 354 is S354Y. In some embodiments, the substitution at amino acid position 347 is Q347E. In some embodiments, the first CH3 domain and the second CH3 domain further comprise a KIH residue, such as T366Y or Y407T. In some embodiments, VL1 and VL2 have the same amino acid sequence. In some embodiments, VL1 and VL2 have different amino acid sequences.

Nucleic acids

The present application also provides isolated nucleic acid molecules comprising polynucleotides encoding one or more polypeptide chains of a heteromultimeric protein described herein, such as a multispecific antibody.

Oligonucleotide-mediated mutagenesis can be used to prepare substituted variants of the DNA encoding the first or second polypeptide. This technique is well known in the art, as described in Adelman et al, DNA,2:183 (1983). Briefly, the first or second polypeptide DNA is altered by hybridizing an oligonucleotide encoding the desired mutation to a DNA template, wherein the template is a single stranded form of a plasmid or phage containing the unaltered or native heteromultimeric DNA sequence. Following hybridization, the entire second complementary strand of the template is synthesized using a DNA polymerase, thereby incorporating the oligonucleotide primer and encoding the selected alteration in the heteromultimeric DNA. Cassette mutagenesis can be performed by replacing the region of the target DNA with a synthetic mutant fragment generated by annealing a complementary oligonucleotide, as described by Wells et al, Gene34:315 (1985). PCR mutagenesis is also suitable for making variants of the first or second polypeptide DNA.

In some embodiments, the nucleic acid molecule comprises a polynucleotide encoding a first polypeptide or a second polypeptide of a heteromultimeric protein. In some embodiments, the nucleic acid molecule comprises a polynucleotide encoding a first polypeptide of a heteromultimeric protein and a polynucleotide encoding a second polypeptide of a heteromultimeric protein. In some embodiments, the nucleic acid molecule comprises a polynucleotide encoding a heavy chain or a light chain of a multispecific antibody. In some embodiments, the nucleic acid molecule comprises a polynucleotide encoding a heavy chain and one or more light chains of a multispecific antibody. In some embodiments, the first nucleic acid molecule comprises a first polynucleotide encoding a first heavy chain, the second nucleic acid molecule comprises a second polynucleotide encoding a second heavy chain, and the third nucleic acid molecule comprises a third polynucleotide encoding a common light chain. In some embodiments, one or more nucleic acid molecules are operably linked to a promoter. In some embodiments, different nucleic acid molecules are operably linked to different promoters.

Additional promoter elements, such as enhancers, regulate the frequency of transcription initiation. Typically, these promoters are located in the region 30-110bp upstream of the start site, although many promoters have recently been shown to also contain functional elements downstream of the start site. The spacing between promoter elements is generally flexible, so that promoter function is retained when the elements are inverted or moved relative to one another. In the thymidine kinase (tk) promoter, the spacing between promoter elements can be increased to 50bp apart before activity begins to decline.

An example of a suitable promoter is the immediate early Cytomegalovirus (CMV) promoter sequence. The promoter sequence is a strong constitutive promoter sequence capable of driving high levels of expression of any polynucleotide sequence to which it is operably linked. Another example of a suitable promoter is elongation growth factor-1 α (EF-1 α). However, other constitutive promoter sequences may also be used, including, but not limited to, the simian virus 40(SV40) early promoter, the Mouse Mammary Tumor Virus (MMTV), the Human Immunodeficiency Virus (HIV) Long Terminal Repeat (LTR) promoter, the MoMuLV promoter, the avian leukemia virus promoter, the EB virus immediate early promoter, the rous sarcoma virus promoter, and human gene promoters such as, but not limited to, the actin promoter, the myosin promoter, the hemoglobin promoter, and the creatine kinase promoter. In addition, the present invention should not be limited to the use of constitutive promoters. Inducible promoters are also envisaged as part of the invention. The use of an inducible promoter provides a molecular switch that can turn on expression of the polynucleotide sequence, which is operably linked when such expression is desired, or turn off expression when expression is not desired. Examples of inducible promoters include, but are not limited to, metallothionein promoters, glucocorticoid promoters, progesterone promoters, and tetracycline promoters.

In some embodiments, the polynucleotide encoding the first polypeptide and/or the second polypeptide and/or the one or more light chains of the heteromultimeric protein comprises a nucleotide sequence encoding a leader sequence that, when translated, is N-terminal to the first polypeptide and/or the second polypeptide and/or the one or more light chains. The leader sequence may be that of the native heavy or light chain, or may be another heterologous leader sequence. In some embodiments, the nucleic acid (or set of nucleic acids) encoding the heteromultimeric protein may further comprise a nucleic acid sequence encoding a peptide tag (such as a protein purification tag, e.g., a His tag, an HA tag).

The present application also includes variants of these nucleic acid sequences. For example, variants include nucleotide sequences that hybridize under at least moderately stringent hybridization conditions to a nucleic acid sequence encoding any of the heteromultimeric proteins described herein.

Nucleic acid molecules can be constructed using recombinant DNA techniques conventional in the art. In some embodiments, the nucleic acid molecule is an expression vector suitable for expression in a selected host cell.

Carrier

Vectors are provided comprising polynucleotides encoding a first polypeptide, a second polypeptide, and/or one or more light chains of any of the heteromultimeric proteins described herein, such as multispecific antibodies. Such vectors include, but are not limited to, DNA vectors, phage vectors, viral vectors, retroviral vectors, and the like. '

In some embodiments, the vector comprises a first polynucleotide sequence encoding a first polypeptide and a second polynucleotide sequence encoding a second polypeptide. In some embodiments, the first polypeptide and the second polypeptide are expressed from the vector as two separate polypeptides. In some embodiments, the first polypeptide and the second polypeptide are expressed as part of a single polypeptide.

In some embodiments, the vector comprises a first polynucleotide sequence encoding a first heavy chain, a second polynucleotide sequence encoding a second heavy chain, and a third polynucleotide sequence encoding a common light chain. In some embodiments, the heavy chain and one or more light chains are expressed from the vector as separate polypeptides. In some embodiments, the heavy chain and one or more light chains are expressed as part of a single polypeptide.

In some embodiments, the first vector comprises a polynucleotide encoding a first polypeptide and the second vector comprises a polynucleotide encoding a second polypeptide. In some embodiments, the first vector and the second vector are transfected into the host cell in similar amounts (such as similar molar amounts or similar mass amounts). In some embodiments, the first vector and the second vector are transfected into a host cell at a molar or mass ratio of 5:1 to 1: 5.

Nucleic acid clones can be cloned into many types of vectors. For example, the nucleic acid can be cloned into a vector (including but not limited to plasmids, phagemids, phage derivatives, animal viruses, and cosmids). Vectors of particular interest include expression vectors, replication vectors, probe generation vectors, and sequencing vectors.

In addition, the expression vector may be provided to the cell in the form of a viral vector. Viral vector technology is well known in the art and is described, for example, in Green and Sambrook (2013, Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory, New York), as well as other virology and Molecular biology manuals. Viruses that can be used as vectors include, but are not limited to, retroviruses, adenoviruses, adeno-associated viruses, herpes viruses, and lentiviruses. Generally, suitable vectors comprise an origin of replication, a promoter sequence, a convenient restriction endonuclease site, and one or more selectable markers that function in at least one organism (see, e.g., WO 01/96584; WO 01/29058; and U.S. Pat. No. 6,326,193).

Expression of heteromultimeric proteins by one or more nucleic acids encoding a polypeptide can be accomplished by inserting the nucleic acid into a suitable expression vector such that the nucleic acid is operably linked to 5 ' and 3 ' regulatory elements, including, for example, a promoter and a3 ' untranslated region (UTR). The vector may be adapted for replication and integration in a eukaryotic host cell. Typical cloning and expression vectors contain transcription and translation terminators, initiation sequences, and promoters for regulating the expression of the desired nucleic acid sequences.

In some embodiments, a vector optimized for expression of a polypeptide in CHO cells or CHO-derived cells or NSO cells is selected. Exemplary such methods are described, for example, in Running der et al, biotechnol. prog.20:880-889 (2004).

To assess the expression of the polypeptide or portion thereof, the expression vector to be introduced into the cells may also comprise a selectable marker gene or a reporter gene or both to facilitate identification and selection of expressing cells from a population of cells sought to be transfected or infected by the viral vector. In other aspects, the selectable marker may be carried on a separate piece of DNA and used in a co-transfection procedure. Both the selectable marker and the reporter gene may be flanked by appropriate regulatory sequences to enable expression in a host cell. Useful selectable markers include, for example, antibiotic resistance genes such as neo and the like.

Reporter genes are used to identify potentially transfected cells and to evaluate the functionality of regulatory sequences. In general, a reporter gene is a gene that is absent or not expressed in a recipient organism or tissue and that encodes a polypeptide whose expression exhibits some easily detectable property (e.g., enzymatic activity). Expression of the reporter gene is detected at a suitable time after introduction of the DNA into the recipient cells. Suitable reporter genes may include genes encoding luciferase, beta-galactosidase, chloramphenicol acetyltransferase, secreted alkaline phosphatase, or green fluorescent protein (e.g., Ui-Tel et al, 2000FEBS Letters 479: 79-82). Suitable expression systems are well known and can be prepared using known techniques or obtained commercially. Generally, the construct with the smallest 5' flanking region that showed the highest reporter expression level was identified as the promoter. Such promoter regions may be linked to a reporter gene and used to assess the ability of an agent to modulate promoter-driven transcription.

Host cell

The present application provides an isolated host cell comprising any of the heteromultimeric proteins (such as multispecific antibodies), nucleic acid molecules, or vectors described herein.

The heteromultimeric proteins described herein, such as multispecific antibodies, can be expressed in prokaryotic cells, such as bacterial cells, or in eukaryotic cells, such as fungal cells, such as yeast, plant cells, insect cells, and mammalian cells. Such expression can be performed, for example, according to procedures known in the art. Exemplary eukaryotic cells that can be used to express the polypeptide include, but are not limited to, COS cells, including COS 7 cells; 293 cells, including 293-6E cells; CHO cells, including CHO-S, DG 44. Lec13 CHO cells and FUT8 CHO cells; PER.Cells (Crucell); and NSO cells. Suitable non-mammalian host cells include prokaryotes such as E.coli (E.coli) or Bacillus subtilis (B.subtilis) and yeasts such as Saccharomyces cerevisiae (S.cerevisae), Schizosaccharomyces pombe (S.pombe); or Kluyveromyces lactis (K.lactis). In some embodiments, a particular eukaryotic host cell is selected based on its ability to perform a desired post-translational modification of the heavy and/or light chain of an antibody. For example, in some embodiments, a polypeptide produced by a CHO cell has a higher level of sialylation compared to the same polypeptide produced in a 293 cell.

One or more nucleic acids can be introduced into a desired host cell by any method, including but not limited to calcium phosphate transfection, DEAE-dextran mediated transfection, cationic lipid mediated transfection, electroporation, transduction, infection, and the like. Non-limiting exemplary methods are described, for example, in Sambrook et al, Molecular Cloning, A Laboratory Manual, 3 rd edition Cold Spring Harbor Laboratory Press (2001). The nucleic acid may be transiently or stably transfected into the desired host cell according to any suitable method.

In some embodiments, the heteromultimeric protein is produced in a cell-free system. For example, in Sitaraman et al, Methods mol. biol.498:229-44 (2009); spirin, Trends Biotechnol.22:538-45 (2004); a non-limiting exemplary cell-free system is described in Endo et al, Biotechnol. adv.21: 695-one 713 (2003).

Purification of

The heteromultimeric proteins can be purified from the host cell culture using standard techniques. The heteromultimeric protein can be recovered from the culture medium as a secreted protein, although it can also be recovered from host cell lysates when produced directly in the absence of a secretion signal. If the heteromultimeric protein is membrane-bound, it can be released from the membrane using a suitable detergent solution.

When the heteromultimeric protein is produced in recombinant cells other than those of human origin, it is completely free of human-derived proteins or polypeptides. However, it is necessary to purify the heteromultimeric protein from the recombinant cellular protein or polypeptide to obtain a substantially homogeneous preparation with respect to the heteromultimeric protein. As a first step, the culture medium or lysate is typically centrifuged to remove particulate cell debris.

Heteromultimeric proteins with antibody constant domains can be conveniently purified by hydroxyapatite chromatography, gel electrophoresis, dialysis or affinity chromatography. Other purification techniques include, but are not limited to, chromatographic methods such as size exclusion, ion exchange (e.g., MonoQ), hydrophobic interaction chromatography, mixed mode chromatography (e.g., reverse phase/anion exchange, reverse phase/cation exchange, hydrophilic interaction/anion exchange, hydrophilic interaction/cation exchange, etc.), silica gel chromatography, heparin sepharose chromatography, anion or cation exchange resin chromatography (such as polyaspartic acid columns), reverse phase HPLC, ultracentrifugation, ethanol precipitation, chromatofocusing, SDS-PAGE, and ammonium sulfate precipitation. Suitable affinity ligands include ligands that bind to the constant region of an antibody. For example, protein a, protein G, protein a/G, or an antibody affinity column can be used to bind the constant region and purify antibodies comprising an Fc fragment. In some embodiments, the heteromultimeric protein is purified using protein a beads, followed by purification with a MonoQ column.

Method of use

The heteromultimeric proteins described herein (e.g., multispecific antibodies) are useful in therapy and diagnosis.

Also provided herein are compositions (such as pharmaceutical compositions) comprising any of the heteromultimeric proteins, nucleic acids, vectors, or host cells described herein. In some embodiments, the heteromultimeric protein can be formulated in a pharmaceutical composition comprising one or more pharmaceutically acceptable buffers or excipients. Such pharmaceutical compositions can be administered to an individual in need thereof to treat a disease or condition, prevent a disease or condition, or arrest the development of symptoms of a disease or condition.

Pharmaceutical compositions of the heteromultimeric proteins described herein can be obtained by mixing the heteromultimeric protein of the desired purity with optional pharmaceutically acceptable carriers, excipients, or stabilizers in the form of lyophilized formulations or aqueous solutions (Remington's Pharmaceutical Sciences 16 th edition, Osol, a. editor (1980)). Acceptable carriers, excipients, or stabilizers are nontoxic to recipients at the dosages and concentrations employed, and include buffers such as phosphate, citrate, and other organic acids; antioxidants (including ascorbic acid and methionine); preservatives (such as octadecyl dimethyl benzyl ammonium chloride; hexa-hydrocarbyl quaternary ammonium chloride; benzalkonium chloride, benzethonium chloride; phenol, butanol or benzyl alcohol; alkyl parabens such as methyl or propyl paraben; catechol; resorcinol; cyclohexanol; 3-pentanol; and m-cresol); low molecular weight (less than about 10 residues) polypeptides; proteins such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, histidine, arginine or lysine; monosaccharides, disaccharides, and other carbohydrates including glucose, mannose, or dextrins; chelating agents such as EDTA; sugars such as sucrose, mannitol, trehalose, or sorbitol; salt-forming counterions such as sodium; metal complexes (such as Zn-protein complexes); and/or non-ionic surfactants such as TWEENTM、PLURONICSTMOr polyethylene glycol (PEG). Adaptation to subcutaneous tissue is described in WO97/04801Lyophilized formulations for administration. Such lyophilized formulations can be reconstituted with a suitable diluent to a high protein concentration, and the reconstituted formulations can be administered to an individual to be imaged, diagnosed, or treated herein. Pharmaceutical compositions for in vivo administration must be sterile. This is readily accomplished by filtration, for example, through sterile filtration membranes.

In some embodiments, there is provided a method of treating a disease in an individual in need thereof, the method comprising administering to the individual an effective amount of a heteromultimeric protein comprising: (a) a first polypeptide comprising a first binding domain that specifically binds to a first target and a first CH3 domain; and (b) a second polypeptide comprising a second binding domain that specifically binds a second target and a second CH3 domain; wherein the first CH3 domain comprises a substitution with a large hydrophobic amino acid at amino acid position 354 relative to the wild-type CH3 domain and/or the second CH3 domain comprises a substitution with a negatively charged amino acid at amino acid position 347 relative to the wild-type CH3 domain, and wherein amino acid residue numbering is based on EU numbering. In some embodiments, the large hydrophobic amino acid is Y, F or W. In some embodiments, the negatively charged amino acid is D or E. In some embodiments, the large hydrophobic amino acid at amino acid position 354 forms a hydrophobic interaction with an amino acid residue in the second CH3 domain. In some embodiments, the second CH3 domain comprises a large hydrophobic residue at amino acid position 349 (e.g., Y349). In some embodiments, the negatively charged amino acid at amino acid position 347 forms an ionic bond with an amino acid residue in the first CH3 domain. In some embodiments, the first CH3 domain comprises a positively charged residue at amino acid position 360 (e.g., K360). In some embodiments, the first and second CH3 domains are human CH3 domains. In some embodiments, the substitution at amino acid position 354 is S354Y. In some embodiments, the substitution at amino acid position 347 is Q347E. In some embodiments, the first CH3 domain and the second CH3 domain further comprise a KIH residue, such as T366Y or Y407T. In some embodiments, the first target is a tumor antigen (e.g., CD20, HER2, or BCMA) and the second target is CD3, or the first target is CD3 and the second target is a tumor antigen (e.g., CD20, HER2, or BCMA). In some embodiments, the disease is cancer.

In some embodiments, there is provided a method of treating a disease in an individual in need thereof, the method comprising administering to the individual an effective amount of a multispecific antibody comprising: (a) a first heavy chain comprising, from N-terminus to C-terminus: a VH1, a first CH1, a first CH2, and a first CH3 domain; (b) a first light chain comprising, from N-terminus to C-terminus: VL1 and CL; (c) a second heavy chain comprising from N-terminus to C-terminus: a VH2, a second CH1, a second CH2, and a second CH3 domain; and (d) a second light chain comprising from N-terminus to C-terminus: VL2 and a second CL; wherein VH1 associates with VL1 to form a first antigen binding site that specifically binds to a first target, and VH2 associates with VL2 to form a second antigen binding site that specifically binds to a second target; wherein the first CH3 domain comprises a substitution with a large hydrophobic amino acid at amino acid position 354 relative to the wild-type CH3 domain and/or the second CH3 domain comprises a substitution with a negatively charged amino acid at amino acid position 347 relative to the wild-type CH3 domain; and wherein amino acid residue numbering is based on EU numbering. In some embodiments, the large hydrophobic amino acid is Y, F or W. In some embodiments, the negatively charged amino acid is D or E. In some embodiments, the large hydrophobic amino acid at amino acid position 354 forms a hydrophobic interaction with an amino acid residue in the second CH3 domain. In some embodiments, the second CH3 domain comprises a large hydrophobic residue at amino acid position 349 (e.g., Y349). In some embodiments, the negatively charged amino acid at amino acid position 347 forms an ionic bond with an amino acid residue in the first CH3 domain. In some embodiments, the first CH3 domain comprises a positively charged residue at amino acid position 360 (e.g., K360). In some embodiments, the first and second CH3 domains are human CH3 domains. In some embodiments, the substitution at amino acid position 354 is S354Y. In some embodiments, the substitution at amino acid position 347 is Q347E. In some embodiments, the first CH3 domain and the second CH3 domain further comprise a KIH residue, such as T366Y or Y407T. In some embodiments, VL1 and VL2 have the same amino acid sequence. In some embodiments, VL1 and VL2 have different amino acid sequences. In some embodiments, the first target is a tumor antigen (e.g., CD20, HER2, or BCMA) and the second target is CD3, or the first target is CD3 and the second target is a tumor antigen (e.g., CD20, HER2, or BCMA). In some embodiments, the disease is cancer.

In some embodiments, the heteromultimeric proteins are used in diagnostic assays. For example, heteromultimeric proteins can be used in sandwich assays that involve the use of two molecules, each molecule capable of binding a different immunogenic portion or epitope of the sample to be detected. In a sandwich assay, a test sample analyte is bound by a first arm of a heteromultimeric protein immobilized on a solid support, after which a second arm of the heteromultimeric protein binds to the analyte, thereby forming an insoluble three-part complex. The second arm of the heteromultimer can itself be labeled with a detectable moiety (direct sandwich assay) or can be measured using an anti-immunoglobulin antibody labeled with a detectable moiety (indirect sandwich assay). For example, one type of sandwich assay is an ELISA assay, in which case the detectable moiety is an enzyme.

V. kits and articles of manufacture

Also provided are kits that can be used in any of the preparation, diagnostic, and therapeutic methods described herein, including kits comprising any of the heteromultimeric proteins (e.g., multispecific antibodies) described herein.

The kits of the present application are in suitable packaging. Suitable packaging includes, but is not limited to, vials, bottles, jars, flexible packaging (e.g., sealed Mylar or plastic bags), and the like. The kit may optionally provide additional components such as reagents, buffers, and explanatory information.

Accordingly, the present application also provides articles. The article may comprise a container and a label or package inserted on or accompanying the container. Suitable containers include vials (such as sealed vials), bottles, cans, flexible packages, and the like. In some embodiments, the container contains the pharmaceutical composition and may have a sterile access port (e.g., the container may be an intravenous solution bag or vial having a stopper pierceable by a hypodermic injection needle). The label or package insert indicates that the composition is for use in diagnosing (including determining risk), treating or preventing a disease or condition in an individual. The label may indicate the reconstitution and/or direction of use of the various components. The container holding the pharmaceutical composition may be a multi-purpose vial that allows for repeated administration (e.g., 2 to 6 administrations) of the reconstituted formulation. Package insert refers to an insert, typically contained in commercial packages of diagnostic and/or therapeutic products, containing information about indications, usage, dosage, administration, contraindications and/or warnings of use of such products. In addition, the article of manufacture may also include a second container comprising a pharmaceutically acceptable buffer, such as bacteriostatic water for injection (BWFI), phosphate buffered saline, Ringer's solution, and dextrose solution. It may also include other materials that appear desirable to the commercial and user, including other buffers, diluents, filters, needles, and syringes.

The kit or article of manufacture may include a plurality of unit doses of the pharmaceutical composition and instructions for use packaged in amounts sufficient for storage and use in pharmacies, such as hospital pharmacies and compounding pharmacies.

Examples

The following examples are merely illustrative of the present invention and therefore should not be considered as limiting the invention in any way. The following examples and detailed description are provided by way of illustration and not limitation.

Example 1 preparation, purification and characterization of a CD20/CD3 bispecific antibody

Bispecific CD20/CD3 antibodies were prepared and characterized according to the scheme of figure 3.

In the first format (S1 of fig. 1), two common light chain CD20/CD3 antibodies were prepared. The V2 CD20/CD3 antibody has KIH residues T366Y in the first heavy chain and Y407T residues in the second heavy chain. The V4b CD20/CD3 antibody has T336Y and S354Y in the first heavy chain and Y407T and Q347E in the second heavy chain.

Vectors encoding the heavy and light chains were transiently transfected into expi293 or CHO cells at a 1:1:2 or 1:1:5 first heavy chain (H1) to second heavy chain (H2) to common light chain ratio. The bispecific antibody is cultured and induced to secrete from the host cell. The supernatant of the cell culture containing the bispecific antibody was centrifuged at 3000rpm for 10 minutes, and then the supernatant sample was filtered with a 0.45 μm membrane.

The bispecific antibody was then purified from the supernatant sample using a two-step chromatography protocol comprising a first protein a purification step and a MonoQ step. In the protein a purification step, the AKTA purification system was first equilibrated with 10 Column Volumes (CV) of buffer a (PBS pH 7.4). The supernatant was loaded onto a protein a purification column and the column was washed with 10CV of buffer a. Bispecific antibody was eluted with buffer B (0.1M glycine, pH 2.5) and peak fractions were collected and pooled. Pooled antibody samples were dialyzed twice against PBS.

In the MonoQ step, antibody samples were first exchanged into buffer a' (20mM Tris-Cl, pH 9). The AKTA purification system was then equilibrated with buffer a'. The antibody sample was loaded onto a MonoQ column, which was subsequently washed with 5CV of buffer A'. The bispecific antibody was eluted from the MonoQ column with 0-25% buffer B ' over 50 minutes at a flow rate of 0.4ml/min using a salt gradient established from buffer a ' and buffer B ' ((20mM Tris-Cl,1M NaCl, pH 9).

The purified bispecific antibody fraction from the MonoQ column was confirmed using a T cell activation assay. Briefly, Raji (human B lymphocytes) and Jurkat (human T lymphocytes) cells were plated at 1X10, respectively5Individual cells/well were seeded into 96-well plates. Diluted bispecific antibody samples were added to the plates and incubated at 37 ℃ with 5% CO2Incubate in incubator for 17 hours. The plate was washed once with PBS containing 0.1% BSA at 1200rpm for 5 minutes. CD69-PE was then added to the plates and incubated at room temperature for 30 minutes. The plate was then washed once with PBS containing 0.1% BSA at 1200rpm for 5 minutes. The CD69+ signal was read from the plate in the FL2 channel on a BD Calibur plate reader.

The expression levels of the three batches of the CD20/CD3 bispecific antibody and the yield of each purification step are shown in Table 3 below. For reference, rituximab is expressed at about 130mg/L from the same expression system. FIG. 4 shows chromatograms from MonoQ purification steps for three CD20/CD3 bispecific antibodies. The introduction of the S354Y and Q349E mutations into the Fc increased the overall yield of bispecific antibody from 67.8% (KIH mutation only) to 86.8% (KIH + S354Y and Q349E). As shown in figure 6, the high yield and increased heterodimer formation rate of the V4b construct was reproducible in different batches of bispecific antibody preparations.

Table 3 expression levels and purification yields of CD20/CD3 bispecific antibodies.

Characterization of the purified CD20/CD 3V 4B bispecific antibody showed high purity (fig. 5B, fig. 5D, fig. 5E), T cell activation activity (fig. 5C), thermostability, and anti-aggregation properties (fig. 5F).

Example 2 preparation, purification and characterization of Her2/CD3 bispecific antibody

Her2/CD3 bispecific antibody comprising one anti-Her 2Fab

A Her2/CD3 bispecific antibody comprising an anti-Her 2Fab, an anti-CD 3 Fab and an Fc domain ("Her 2-B3/CD3 BsAb", FIG. 8) was prepared by co-transfecting expi293 cells with plasmids expressing Her2-B3 HC-v 4B-knob, CD3-HC-v 4B-well and common light chain. Expression levels were determined to be 152ug/ml 96 hours after transfection. Culture supernatants were collected and IgG purified on a protein a column. The purified sample was then loaded onto a cation exchange chromatography Column (CEX). After gradient elution, bispecific antibody appeared as the main peak detected by CEX (fig. 9). The purified fractions were further analyzed by sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE). The SDS-PAGE results showed relatively high fractional purity of the main CEX peak (FIG. 10).

Her2/CD3 bispecific antibody comprising two anti-Her 2Fab

A Her2/CD3 bispecific antibody comprising two anti-Her 2fabs ("Her 2-B3-V3/CD 3") was prepared (fig. 11).

The anti-tumor antigen arm of bispecific antibodies with high affinity for tumor cells can generally kill healthy non-tumor cells expressing normal levels of antigen. To eliminate this type of off-tumor effect, a new form of bispecific antibody was developed, in which the tumor targeting arm has two copies of the Fab domain and thus becomes bivalent to the tumor antigen (fig. 11). In this three-domain bispecific (TDB) format (2:1), the affinity of the tumor-binding region for the tumor antigen is relatively low and therefore its binding to healthy cells with a lower density of tumor antigens is weaker.

Expi293 cells were co-transfected with plasmids expressing Her2(2Fabs) -CH-v4 b-knob, CD3-CH-v4 b-well and common light chain DNA. The medium was collected and the expression level was determined to be 158ug/ml 96 hours after transfection using ProbeLife. The bispecific antibody Her2-B3-V3/CD3 was purified using a protein A column. The purified sample was then loaded onto a cation exchange Column (CEX) for a second purification. After gradient elution, bispecific antibody appeared as a major peak (fig. 12). The purified fractions were further analyzed by SDS-PAGE (FIG. 13). Non-reducing SDS-PAGE results showed only one band of about 200kD in size after CEX purification, which is consistent with the size of Her2-B3-V3/CD3 (with two Her2 Fab). The reducing SDS-PAGE results clearly show the three chains of the bispecific antibody, which is also consistent with their corresponding size.

Example 3 preparation, purification and characterization of a BCMA/CD3 bispecific antibody

A BCMA/CD3 bispecific antibody was prepared. anti-BCMA VHH panned from the llama phage library was linked to human Fc-v4 b-knob to form BCMA-Fc chains. The BCMA-Fc chain together with CD3-HC-v4 b-well and its light chain form a novel IgG-like bispecific antibody. Schematic representations of two BCMA/CD3 IgG-like bispecific antibodies are shown in figure 14. On the left side of FIG. 14 is BCMA-3E5/CD3, containing only one BCMA-VHH (3E 5). On the right of FIG. 14 is BCMA-3E1B2/CD3, containing two BCMA-VHHs (3E1 and 3B 2).

To prepare the BCMA/CD3 bispecific antibody, expi293 cells were co-transfected with plasmids expressing BCMA (3E5) -Fc-v 4B-knob (or BCMA (3E1B2) -Fc-v 4B-knob), CD3-CH-v 4B-well, and CD3 LC. The medium was collected and the expression level of BCMA-3E5/CD3 was determined to be 62ug/ml and the expression level of BCMA-3E1B2/CD3 was determined to be 54ug/ml using ProbeLife.

Culture supernatants were collected and both bispecific antibodies were purified on a protein a column. SDS-PAGE was performed on two BCMA-Fc/CD3 bispecific antibodies (FIG. 15). The results indicated that both BCMA/CD3 bispecific antibodies were successfully expressed and that some CD3 homodimers were formed (size approximately 150 Kd; see FIG. 15). In the reducing SDS-PAGE results, CD3-VH-v4 b-well, BCMA-Fc and common light chain were detected based on their molecular weights (FIG. 15).

Further experiments were performed to purify and characterize the BCMA/CD3 bispecific antibody. First, the ratio of three plasmids expressing three strands in transfection was optimized. Optimization of the three plasmid ratios increased the percentage of bispecific antibody heterodimers. BCMA/CD3 bispecific antibody was expressed using optimized plasmid ratios and purified on a protein a column and by CEX. BCMA/CD3 bispecific antibody purity was assessed by SDS-PAGE.

Example 4 preparation, purification and characterization of CD20/CD3 bispecific antibody with Fc mutation

A CD20/CD3 bispecific antibody having a mutation in the Fc region was generated. Mutations to the anti-CD 20 heavy chain are shown in table 4. Mutations directed against the heavy chain of CD3 are shown in table 5.

TABLE 4 anti-CD 20 heavy chain Fc mutation.

TABLE 5 anti-CD 3 heavy chain Fc mutation.

All 13 combinations of anti-CD 20 heavy chain and anti-CD 3 heavy chain with Fc mutations were recombinantly expressed with their common light chain. The concentration of the antibody in the supernatant was measured, and the antibody concentration was shown in table 6.

TABLE 6 IgG supernatant concentrations of mutant combinations.

In further experiments, CD20/CD3 bispecific antibodies with the above-described combinations of Fc mutations were characterized. The activity of the CD20/CD3 bispecific antibody on T cell activation was evaluated. Combinations of Fc mutants with T cell activation activity similar to or higher than CD20/CD3 wild type (A1B1) were identified and selected.

Selected combinations of CD20/CD3 mutants were recombinantly expressed, purified, and analyzed by SDS-PAGE. Fc mutant combinations with similar or higher purity (i.e., percentage heterodimers) than A1B1 were identified and selected.

The selected CD20/CD3 mutant combinations were evaluated for thermostability and aggregation potential using the methods described in example 5 below. Thermal stability was assessed by differential scanning fluorimetry and static light scattering. Aggregation potential was assessed by Dynamic Light Scattering (DLS). Mutant combinations with thermostability similar to or better than A1B1 and aggregation potential similar to or lower than A1B1 were identified and selected.

Example 5 materials and methods

The following examples demonstrate materials and methods for producing and characterizing bispecific antibodies.

By transfection with EXPI293FTMCell recombinant expression of bispecific antibodies

As described below, GIBCO was usedTMEXPIFECTAMINETM293 turnThe staining kit (cat # a14524) expresses bispecific antibody.

For each 30-mL transfection, at 25.5mL EXPI293TMThe expression medium used was 7.5X 107And (4) cells. To transfect the cells the following day, the cells were plated at 2.0X 106Viable cells/mL, and 8% CO in air at 37 ℃2Was incubated with rotation on an orbital shaker at 125rpm in a humid atmosphere. On the day of transfection, the number and viability of cells were determined using an automated cell counter or trypan blue dye exclusion. For transfection, the viability of the cells must be greater than 95%. The number of cells required to contain one transfection was calculated (7.5X 10 per 30-mL transfection)7Individual cells) of the cell suspension. To each sterile disposable 125-mL Erlenmeyer flask was added an appropriate volume of cell suspension and fresh, pre-warmed EXPI293 was added for each 30-mL transfectionTMThe medium was expressed to a volume of 25.5 mL. The cells were returned to the incubator.

For each 30-mL transfection, lipid-DNA complexes were prepared as follows: 30. mu.g of OPTI-MEMTMI reduction of plasmid DNA in serum medium (Cat. No. 31985-. Mixing 80 μ L EXPIFECTAMINETM293 reagent in OPTI-MEMTMI medium was diluted to a total volume of 1.5mL, gently mixed and incubated at room temperature for 5 minutes. After 5 minutes of incubation, the diluted DNA was added to the diluted EXPIFECTAMINETM293, to obtain a total volume of 3mL, and mix gently. Mixing DNA-EXPIFECTAMINETM293 reagent mixture incubated at room temperature for 20-30 min to allow DNA-EXPIFECTAMINETM293 reagent complex formation.

After completion of DNA-EXPIFECTAMINETM293 reagent Complex incubation, 3mL of DNA-EXPIFECTAMINE was added to each flask from the lipid-DNA complexes prepared aboveTM293 reagent complex. To a negative control flask was added 3mL of OPTI-MEMTMI Medium, not DNA-EXPIFECTAMINETM293 reagent complex. Each flask contained a total volume of 28.5 mL. Cells were incubated at 37 ℃ in an incubator with 8% CO in air2Of moist airIncubate at 125rpm on an orbital shaker in an atmosphere.

About 16-18 hours after transfection, 150. mu.L of EXPIFECTAMINE was added to each flaskTM293 transfection enhancer 1 and 1.5mL EXPIFECTAMINETM293 transfection enhancer 2. The final volume in each 125-mL flask was approximately 30 mL. The culture medium was harvested approximately 72-96 hours after transfection and assayed for recombinant protein expression.

Purification of bispecific antibodies with protein A column

To purify bispecific antibodies on a protein a column, the expression medium was centrifuged at 3000rpm for 10 minutes 72-96 hours after transfection, and the supernatant was then filtered through a 0.45 μm membrane.

Use as followsA pure protein purification system. BalancingPurification system, pump B was filled with buffer B (0.1M glycine, pH 2.5), and pump a and sample pump were filled with buffer a (PBS, pH 7.4). A protein A purification column (HiTrap protein A HP column from GE, Cat: 17040201 or 17040301) was then set up and equilibrated with 10 Column Volumes (CV) of buffer A. The supernatant was loaded into the column by a sample pump at a flow rate of 1 ml/min for a 1ml column and 3 ml/min for a 5ml column. After sample loading, the column was washed with 10CV of buffer A. The antibody was eluted with 100% buffer B and the peak fractions were collected with 1/10 volumes of 1M Tris pH 8. The column was washed with 5CV of buffer B and then 10CV of buffer A. The column was packed with 20% ethanol at 4 ℃ and stored, andthe system was filled with 20% ethanol for storage.

Selected peak fractions of eluted Ab were pooled and dialyzed twice in PBS. The purified antibody is then ready for a second purification step.

Second purification of bispecific antibody by cation exchange Chromatography (CEX)

The antibody was diluted 1:10 (in PBS) to buffer 1(20mM NaxH (3-x) PO4, pH 7.4). BalancingPurification system, pump B was filled with buffer 2(20mM NaxH (3-x) PO4,1M NaCl, pH 7.4), and pump a and sample pump were filled with buffer 1. Setting POROSTMGOPURETMHS pre-filled column (Thermofoisher, Cat. No: A36637) and buffer 1 was equilibrated.

The diluted antibody sample was loaded to the column by a sample pump at a flow rate of 1.6 ml/min and washed with 5CV of buffer 1 after sample loading. For salt gradient elution, 0-20% buffer 2 was used at a flow rate of 1.6 ml/min over 40 minutes. The peak fraction (typically 1 ml/vial) was collected. The column was washed with 5CV of buffer 2 and then 10CV of buffer 1. The column was packed with 20% ethanol at 4 ℃ for storage and equilibrated in 20% ethanolProvided is a system. Peak fractions were pooled and dialyzed against PBS.

SDS-PAGE assay

For non-reducing SDS-PAGE, 4 fold loading buffer was added to the samples to obtain 1 fold sample ready for loading. For reducing SDS-PAGE, 8% by volume of beta-mercaptoethanol was added to the sample, mixed well, heated at 95 ℃ for 5 minutes, and the sample was then ready for loading. The sample was loaded into the gel well, next to which was a Protein Marker (Protein Marker). The sample was run at 200V for 50 minutes. INSTANTBBLUE for gelTMThe protein staining solution was stained for 1 hour and then washed once with water.

Thermostability (DSF/SLS) and aggregation potential (DLS) assays

Purified bispecific antibody samples were submitted to the UNcle system (Uncariamed Labs) for analysis. Dynamic Light Scattering (DLS) was measured at 25 ℃ and data was calculated and analyzed using UNcle analysis software. For the differential scanning fluorimetry/static light scattering (DSF/SLS) assay,a temperature ramp of 1 deg.C/min was carried out with monitoring from 25 deg.C to 95 deg.C. UNcle measured SLS at 266nm and 473 nm. T ismAnd TaggCalculations and analyses were also performed using UNcle analysis software.

Sequence listing

<110> AB Studio Co., Ltd (AB Studio Inc.)

<120> heteromultimeric proteins and methods of use thereof

<130> 792702000240

<140> not yet allocated

<141> simultaneous accompanying submission

<150> US 62/825,726

<151> 2019-03-28

<160> 18

<170> FastSEQ for Windows version 4.0

<210> 1

<211> 221

<212> PRT

<213> Artificial Sequence (Artificial Sequence)

<220>

<223> synthetic construct

<400> 1

Pro Pro Cys Pro Ala Pro Glu Leu Leu Gly Gly Pro Ser Val Phe Leu

1 5 10 15

Phe Pro Pro Lys Pro Lys Asp Thr Leu Met Ile Ser Arg Thr Pro Glu

20 25 30

Val Thr Cys Val Val Val Asp Val Ser His Glu Asp Pro Glu Val Lys

35 40 45

Phe Asn Trp Tyr Val Asp Gly Val Glu Val His Asn Ala Lys Thr Lys

50 55 60

Pro Arg Glu Glu Gln Tyr Asn Ser Thr Tyr Arg Val Val Ser Val Leu

65 70 75 80

Thr Val Leu His Gln Asp Trp Leu Asn Gly Lys Glu Tyr Lys Cys Lys

85 90 95

Val Ser Asn Lys Ala Leu Pro Ala Pro Ile Glu Lys Thr Ile Ser Lys

100 105 110

Ala Lys Gly Gln Pro Arg Glu Pro Gln Val Tyr Thr Leu Pro Pro Tyr

115 120 125

Arg Glu Glu Met Thr Lys Asn Gln Val Ser Leu Tyr Cys Leu Val Lys

130 135 140

Gly Phe Tyr Pro Ser Asp Ile Ala Val Glu Trp Glu Ser Asn Gly Gln

145 150 155 160

Pro Glu Asn Asn Tyr Lys Thr Thr Pro Pro Val Leu Asp Ser Asp Gly

165 170 175

Ser Phe Phe Leu Tyr Ser Lys Leu Thr Val Asp Lys Ser Arg Trp Gln

180 185 190

Gln Gly Asn Val Phe Ser Cys Ser Val Met His Glu Ala Leu His Asn

195 200 205

His Tyr Thr Gln Lys Ser Leu Ser Leu Ser Pro Gly Lys

210 215 220

<210> 2

<211> 221

<212> PRT

<213> Artificial Sequence (Artificial Sequence)

<220>

<223> synthetic construct

<400> 2

Pro Pro Cys Pro Ala Pro Glu Leu Leu Gly Gly Pro Ser Val Phe Leu

1 5 10 15

Phe Pro Pro Lys Pro Lys Asp Thr Leu Met Ile Ser Arg Thr Pro Glu

20 25 30

Val Thr Cys Val Val Val Asp Val Ser His Glu Asp Pro Glu Val Lys

35 40 45

Phe Asn Trp Tyr Val Asp Gly Val Glu Val His Asn Ala Lys Thr Lys

50 55 60

Pro Arg Glu Glu Gln Tyr Asn Ser Thr Tyr Arg Val Val Ser Val Leu

65 70 75 80

Thr Val Leu His Gln Asp Trp Leu Asn Gly Lys Glu Tyr Lys Cys Lys

85 90 95

Val Ser Asn Lys Ala Leu Pro Ala Pro Ile Glu Lys Thr Ile Ser Lys

100 105 110

Ala Lys Gly Gln Pro Arg Glu Pro Glu Val Tyr Thr Leu Pro Pro Ser

115 120 125

Arg Glu Glu Met Thr Lys Asn Gln Val Ser Leu Thr Cys Leu Val Lys

130 135 140

Gly Phe Tyr Pro Ser Asp Ile Ala Val Glu Trp Glu Ser Asn Gly Gln

145 150 155 160

Pro Glu Asn Asn Tyr Lys Thr Thr Pro Pro Val Leu Asp Ser Asp Gly

165 170 175

Ser Phe Phe Leu Thr Ser Lys Leu Thr Val Asp Lys Ser Arg Trp Gln

180 185 190

Gln Gly Asn Val Phe Ser Cys Ser Val Met His Glu Ala Leu His Asn

195 200 205

His Tyr Thr Gln Lys Ser Leu Ser Leu Ser Pro Gly Lys

210 215 220

<210> 3

<211> 221

<212> PRT

<213> Artificial Sequence (Artificial Sequence)

<220>

<223> synthetic construct

<400> 3

Pro Pro Cys Pro Ala Pro Glu Leu Leu Gly Gly Pro Ser Val Phe Leu

1 5 10 15

Phe Pro Pro Lys Pro Lys Asp Thr Leu Met Ile Ser Arg Thr Pro Glu

20 25 30

Val Thr Cys Val Val Val Asp Val Ser His Glu Asp Pro Glu Val Lys

35 40 45

Phe Asn Trp Tyr Val Asp Gly Val Glu Val His Asn Ala Lys Thr Lys

50 55 60

Pro Arg Glu Glu Gln Tyr Asn Ser Thr Tyr Arg Val Val Ser Val Leu

65 70 75 80

Thr Val Leu His Gln Asp Trp Leu Asn Gly Lys Glu Tyr Lys Cys Lys

85 90 95

Val Ser Asn Lys Ala Leu Pro Ala Pro Ile Glu Lys Thr Ile Ser Lys

100 105 110

Ala Lys Gly Gln Pro Arg Glu Pro Gln Val Tyr Thr Leu Pro Pro Tyr

115 120 125

Arg Glu Glu Met Thr Lys Asn Gln Val Ser Leu Thr Cys Leu Val Lys

130 135 140

Gly Phe Tyr Pro Ser Asp Ile Ala Val Glu Trp Glu Ser Asn Gly Gln

145 150 155 160

Pro Glu Asn Asn Tyr Lys Thr Thr Pro Pro Val Leu Asp Ser Asp Gly

165 170 175

Ser Phe Phe Leu Thr Ser Lys Leu Thr Val Asp Lys Ser Arg Trp Gln

180 185 190

Gln Gly Asn Val Phe Ser Cys Ser Val Met His Glu Ala Leu His Asn

195 200 205

His Tyr Thr Gln Lys Ser Leu Ser Leu Ser Pro Gly Lys

210 215 220

<210> 4

<211> 221

<212> PRT

<213> Artificial Sequence (Artificial Sequence)

<220>

<223> synthetic construct

<400> 4

Pro Pro Cys Pro Ala Pro Glu Leu Leu Gly Gly Pro Ser Val Phe Leu

1 5 10 15

Phe Pro Pro Lys Pro Lys Asp Thr Leu Met Ile Ser Arg Thr Pro Glu

20 25 30

Val Thr Cys Val Val Val Asp Val Ser His Glu Asp Pro Glu Val Lys

35 40 45

Phe Asn Trp Tyr Val Asp Gly Val Glu Val His Asn Ala Lys Thr Lys

50 55 60

Pro Arg Glu Glu Gln Tyr Asn Ser Thr Tyr Arg Val Val Ser Val Leu

65 70 75 80

Thr Val Leu His Gln Asp Trp Leu Asn Gly Lys Glu Tyr Lys Cys Lys

85 90 95

Val Ser Asn Lys Ala Leu Pro Ala Pro Ile Glu Lys Thr Ile Ser Lys

100 105 110

Ala Lys Gly Gln Pro Arg Glu Pro Glu Val Tyr Thr Leu Pro Pro Ser

115 120 125

Arg Glu Glu Met Thr Lys Asn Gln Val Ser Leu Tyr Cys Leu Val Lys

130 135 140

Gly Phe Tyr Pro Ser Asp Ile Ala Val Glu Trp Glu Ser Asn Gly Gln

145 150 155 160

Pro Glu Asn Asn Tyr Lys Thr Thr Pro Pro Val Leu Asp Ser Asp Gly

165 170 175

Ser Phe Phe Leu Tyr Ser Lys Leu Thr Val Asp Lys Ser Arg Trp Gln

180 185 190

Gln Gly Asn Val Phe Ser Cys Ser Val Met His Glu Ala Leu His Asn

195 200 205

His Tyr Thr Gln Lys Ser Leu Ser Leu Ser Pro Gly Lys

210 215 220

<210> 5

<211> 206

<212> PRT

<213> Intelligent (Homo sapiens)

<400> 5

Pro Ser Val Phe Leu Phe Pro Pro Lys Pro Lys Asp Thr Leu Met Ile

1 5 10 15

Ser Arg Thr Pro Glu Val Thr Cys Val Val Val Asp Val Ser His Glu

20 25 30

Asp Pro Glu Val Lys Phe Asn Trp Tyr Val Asp Gly Val Glu Val His

35 40 45

Asn Ala Lys Thr Lys Pro Arg Glu Glu Gln Tyr Asn Ser Thr Tyr Arg

50 55 60

Val Val Ser Val Leu Thr Val Leu His Gln Asp Trp Leu Asn Gly Lys

65 70 75 80

Glu Tyr Lys Cys Lys Val Ser Asn Lys Ala Leu Pro Ala Pro Ile Glu

85 90 95

Lys Thr Ile Ser Lys Ala Lys Gly Gln Pro Arg Glu Pro Gln Val Tyr

100 105 110

Thr Leu Pro Pro Ser Arg Asp Glu Leu Thr Lys Asn Gln Val Ser Leu

115 120 125

Thr Cys Leu Val Lys Gly Phe Tyr Pro Ser Asp Ile Ala Val Glu Trp

130 135 140

Glu Ser Asn Gly Gln Pro Glu Asn Asn Tyr Lys Thr Thr Pro Pro Val

145 150 155 160

Leu Asp Ser Asp Gly Ser Phe Phe Leu Tyr Ser Lys Leu Thr Val Asp

165 170 175

Lys Ser Arg Trp Gln Gln Gly Asn Val Phe Ser Cys Ser Val Met His

180 185 190

Glu Ala Leu His Asn His Tyr Thr Gln Lys Ser Leu Ser Leu

195 200 205

<210> 6

<211> 206

<212> PRT

<213> Intelligent (Homo sapiens)

<400> 6

Pro Ser Val Phe Leu Phe Pro Pro Lys Pro Lys Asp Thr Leu Met Ile

1 5 10 15

Ser Arg Thr Pro Glu Val Thr Cys Val Val Val Asp Val Ser His Glu

20 25 30

Asp Pro Glu Val Gln Phe Asn Trp Tyr Val Asp Gly Val Glu Val His

35 40 45

Asn Ala Lys Thr Lys Pro Arg Glu Glu Gln Phe Asn Ser Thr Phe Arg

50 55 60

Val Val Ser Val Leu Thr Val Val His Gln Asp Trp Leu Asn Gly Lys

65 70 75 80

Glu Tyr Lys Cys Lys Val Ser Asn Lys Gly Leu Pro Ala Pro Ile Glu

85 90 95

Lys Thr Ile Ser Lys Thr Lys Gly Gln Pro Arg Glu Pro Gln Val Tyr

100 105 110

Thr Leu Pro Pro Ser Arg Glu Glu Met Thr Lys Asn Gln Val Ser Leu

115 120 125

Thr Cys Leu Val Lys Gly Phe Tyr Pro Ser Asp Ile Ala Val Glu Trp

130 135 140

Glu Ser Asn Gly Gln Pro Glu Asn Asn Tyr Lys Thr Thr Pro Pro Met

145 150 155 160

Leu Asp Ser Asp Gly Ser Phe Phe Leu Tyr Ser Lys Leu Thr Val Asp

165 170 175

Lys Ser Arg Trp Gln Gln Gly Asn Val Phe Ser Cys Ser Val Met His

180 185 190

Glu Ala Leu His Asn His Tyr Thr Gln Lys Ser Leu Ser Leu

195 200 205

<210> 7

<211> 206

<212> PRT

<213> Intelligent (Homo sapiens)

<400> 7

Pro Ser Val Phe Leu Phe Pro Pro Lys Pro Lys Asp Thr Leu Met Ile

1 5 10 15

Ser Arg Thr Pro Glu Val Thr Cys Val Val Val Asp Val Ser His Glu

20 25 30

Asp Pro Glu Val Gln Phe Lys Trp Tyr Val Asp Gly Val Glu Val His

35 40 45

Asn Ala Lys Thr Lys Pro Arg Glu Glu Gln Phe Asn Ser Thr Phe Arg

50 55 60

Val Val Ser Val Leu Thr Val Leu His Gln Asp Trp Leu Asn Gly Lys

65 70 75 80

Glu Tyr Lys Cys Lys Val Ser Asn Lys Ala Leu Pro Ala Pro Ile Glu

85 90 95

Lys Thr Ile Ser Lys Thr Lys Gly Gln Pro Arg Glu Pro Gln Val Tyr

100 105 110

Thr Leu Pro Pro Ser Arg Glu Glu Met Thr Lys Asn Gln Val Ser Leu

115 120 125

Thr Cys Leu Val Lys Gly Phe Tyr Pro Ser Asp Ile Ala Val Glu Trp

130 135 140

Glu Ser Ser Gly Gln Pro Glu Asn Asn Tyr Asn Thr Thr Pro Pro Met

145 150 155 160

Leu Asp Ser Asp Gly Ser Phe Phe Leu Tyr Ser Lys Leu Thr Val Asp

165 170 175

Lys Ser Arg Trp Gln Gln Gly Asn Ile Phe Ser Cys Ser Val Met His

180 185 190

Glu Ala Leu His Asn Arg Phe Thr Gln Lys Ser Leu Ser Leu

195 200 205

<210> 8

<211> 206

<212> PRT

<213> Intelligent (Homo sapiens)

<400> 8

Pro Ser Val Phe Leu Phe Pro Pro Lys Pro Lys Asp Thr Leu Met Ile

1 5 10 15

Ser Arg Thr Pro Glu Val Thr Cys Val Val Val Asp Val Ser Gln Glu

20 25 30

Asp Pro Glu Val Gln Phe Asn Trp Tyr Val Asp Gly Val Glu Val His

35 40 45

Asn Ala Lys Thr Lys Pro Arg Glu Glu Gln Phe Asn Ser Thr Tyr Arg

50 55 60

Val Val Ser Val Leu Thr Val Leu His Gln Asp Trp Leu Asn Gly Lys

65 70 75 80

Glu Tyr Lys Cys Lys Val Ser Asn Lys Gly Leu Pro Ser Ser Ile Glu

85 90 95

Lys Thr Ile Ser Lys Ala Lys Gly Gln Pro Arg Glu Pro Gln Val Tyr

100 105 110

Thr Leu Pro Pro Ser Gln Glu Glu Met Thr Lys Asn Gln Val Ser Leu

115 120 125

Thr Cys Leu Val Lys Gly Phe Tyr Pro Ser Asp Ile Ala Val Glu Trp

130 135 140

Glu Ser Asn Gly Gln Pro Glu Asn Asn Tyr Lys Thr Thr Pro Pro Val

145 150 155 160

Leu Asp Ser Asp Gly Ser Phe Phe Leu Tyr Ser Arg Leu Thr Val Asp

165 170 175

Lys Ser Arg Trp Gln Glu Gly Asn Val Phe Ser Cys Ser Val Met His

180 185 190

Glu Ala Leu His Asn His Tyr Thr Gln Lys Ser Leu Ser Leu

195 200 205

<210> 9

<211> 206

<212> PRT

<213> little mouse (Mus musculus)

<400> 9

Ser Ser Val Phe Ile Phe Pro Pro Lys Pro Lys Asp Val Leu Thr Ile

1 5 10 15

Thr Leu Thr Pro Lys Val Thr Cys Val Val Val Asp Ile Ser Lys Asp

20 25 30

Asp Pro Glu Val Gln Phe Ser Trp Phe Val Asp Asp Val Glu Val His

35 40 45

Thr Ala Gln Thr Gln Pro Arg Glu Glu Gln Phe Asn Ser Thr Phe Arg

50 55 60

Ser Val Ser Glu Leu Pro Ile Met His Gln Asp Trp Leu Asn Gly Lys

65 70 75 80

Glu Phe Lys Cys Arg Val Asn Ser Ala Ala Phe Pro Ala Pro Ile Glu

85 90 95

Lys Thr Ile Ser Lys Thr Lys Gly Arg Pro Lys Ala Pro Gln Val Tyr

100 105 110

Thr Ile Pro Pro Pro Lys Glu Gln Met Ala Lys Asp Lys Val Ser Leu

115 120 125

Thr Cys Met Ile Thr Asp Phe Phe Pro Glu Asp Ile Thr Val Glu Trp

130 135 140

Gln Trp Asn Gly Gln Pro Ala Glu Asn Tyr Lys Asn Thr Gln Pro Ile

145 150 155 160

Met Asp Thr Asp Gly Ser Tyr Phe Val Tyr Ser Lys Leu Asn Val Gln

165 170 175

Lys Ser Asn Trp Glu Ala Gly Asn Thr Phe Thr Cys Ser Val Leu His

180 185 190

Glu Gly Leu His Asn His His Thr Glu Lys Ser Leu Ser His

195 200 205

<210> 10

<211> 206

<212> PRT

<213> little mouse (Mus musculus)

<400> 10

Pro Ser Val Phe Ile Phe Pro Pro Lys Ile Lys Asp Val Leu Met Ile

1 5 10 15

Ser Leu Ser Pro Ile Val Thr Cys Val Val Val Asp Val Ser Glu Asp

20 25 30

Asp Pro Asp Val Gln Ile Ser Trp Phe Val Asn Asn Val Glu Val His

35 40 45

Thr Ala Gln Thr Gln Thr His Arg Glu Asp Tyr Asn Ser Thr Leu Arg

50 55 60

Val Val Ser Ala Leu Pro Ile Gln His Gln Asp Trp Met Ser Gly Lys

65 70 75 80

Glu Phe Lys Cys Lys Val Asn Asn Lys Asp Leu Pro Ala Pro Ile Glu

85 90 95

Arg Thr Ile Ser Lys Pro Lys Gly Ser Val Arg Ala Pro Gln Val Tyr

100 105 110

Val Leu Pro Pro Pro Glu Glu Glu Met Thr Lys Lys Gln Val Thr Leu

115 120 125

Thr Cys Met Val Thr Asp Phe Met Pro Glu Asp Ile Tyr Val Glu Trp

130 135 140

Thr Asn Asn Gly Lys Thr Glu Leu Asn Tyr Lys Asn Thr Glu Pro Val

145 150 155 160

Leu Asp Ser Asp Gly Ser Tyr Phe Met Tyr Ser Lys Leu Arg Val Glu

165 170 175

Lys Lys Asn Trp Val Glu Arg Asn Ser Tyr Ser Cys Ser Val Val His

180 185 190

Glu Gly Leu His Asn His His Thr Thr Lys Ser Phe Ser Arg

195 200 205

<210> 11

<211> 206

<212> PRT

<213> little mouse (Mus musculus)

<400> 11

Pro Ser Val Phe Ile Phe Pro Pro Lys Pro Lys Asp Ala Leu Met Ile

1 5 10 15

Ser Leu Thr Pro Lys Val Thr Cys Val Val Val Asp Val Ser Glu Asp

20 25 30

Asp Pro Asp Val His Val Ser Trp Phe Val Asp Asn Lys Glu Val His

35 40 45

Thr Ala Trp Thr Gln Pro Arg Glu Ala Gln Tyr Asn Ser Thr Phe Arg

50 55 60

Val Val Ser Ala Leu Pro Ile Gln His Gln Asp Trp Met Arg Gly Lys

65 70 75 80

Glu Phe Lys Cys Lys Val Asn Asn Lys Ala Leu Pro Ala Pro Ile Glu

85 90 95

Arg Thr Ile Ser Lys Pro Lys Gly Arg Ala Gln Thr Pro Gln Val Tyr

100 105 110

Thr Ile Pro Pro Pro Arg Glu Gln Met Ser Lys Lys Lys Val Ser Leu

115 120 125

Thr Cys Leu Val Thr Asn Phe Phe Ser Glu Ala Ile Ser Val Glu Trp

130 135 140

Glu Arg Asn Gly Glu Leu Glu Gln Asp Tyr Lys Asn Thr Pro Pro Ile

145 150 155 160

Leu Asp Ser Asp Gly Thr Tyr Phe Leu Tyr Ser Lys Leu Thr Val Asp

165 170 175

Thr Asp Ser Trp Leu Gln Gly Glu Ile Phe Thr Cys Ser Val Val His

180 185 190

Glu Ala Leu His Asn His His Thr Gln Lys Asn Leu Ser Arg

195 200 205

<210> 12

<211> 206

<212> PRT

<213> Brown rat (Rattus norvegicus)

<400> 12

Pro Ser Val Phe Ile Phe Pro Pro Lys Pro Lys Asp Ile Leu Leu Ile

1 5 10 15

Ser Gln Asn Ala Lys Val Thr Cys Val Val Val Asp Val Ser Glu Glu

20 25 30

Glu Pro Asp Val Gln Phe Ser Trp Phe Val Asn Asn Val Glu Val His

35 40 45

Thr Ala Gln Thr Gln Pro Arg Glu Glu Gln Tyr Asn Ser Thr Phe Arg

50 55 60

Val Val Ser Ala Leu Pro Ile Gln His Gln Asp Trp Met Ser Gly Lys

65 70 75 80

Glu Phe Lys Cys Lys Val Asn Asn Lys Ala Leu Pro Ser Pro Ile Glu

85 90 95

Lys Thr Ile Ser Lys Pro Lys Gly Leu Val Arg Lys Pro Gln Val Tyr

100 105 110

Val Met Gly Pro Pro Thr Glu Gln Leu Thr Glu Gln Thr Val Ser Leu

115 120 125

Thr Cys Leu Thr Ser Gly Phe Leu Pro Asn Asp Ile Gly Val Glu Trp

130 135 140

Thr Ser Asn Gly His Ile Glu Lys Asn Tyr Lys Asn Thr Glu Pro Val

145 150 155 160

Met Asp Ser Asp Gly Ser Phe Phe Met Tyr Ser Lys Leu Asn Val Glu

165 170 175

Arg Ser Arg Trp Asp Ser Arg Ala Pro Phe Val Cys Ser Val Val His

180 185 190

Glu Gly Leu His Asn His His Val Glu Lys Ser Ile Ser Arg

195 200 205

<210> 13

<211> 206

<212> PRT

<213> Brown rat (Rattus norvegicus)

<400> 13

Ser Ser Val Phe Ile Phe Pro Pro Lys Thr Lys Asp Val Leu Thr Ile

1 5 10 15

Thr Leu Thr Pro Lys Val Thr Cys Val Val Val Asp Ile Ser Gln Asn

20 25 30

Asp Pro Glu Val Arg Phe Ser Trp Phe Ile Asp Asp Val Glu Val His

35 40 45

Thr Ala Gln Thr His Ala Pro Glu Lys Gln Ser Asn Ser Thr Leu Arg

50 55 60

Ser Val Ser Glu Leu Pro Ile Val His Arg Asp Trp Leu Asn Gly Lys

65 70 75 80

Thr Phe Lys Cys Lys Val Asn Ser Gly Ala Phe Pro Ala Pro Ile Glu

85 90 95

Lys Ser Ile Ser Lys Pro Glu Gly Thr Pro Arg Gly Pro Gln Val Tyr

100 105 110

Thr Met Ala Pro Pro Lys Glu Glu Met Thr Gln Ser Gln Val Ser Ile

115 120 125

Thr Cys Met Val Lys Gly Phe Tyr Pro Pro Asp Ile Tyr Thr Glu Trp

130 135 140

Lys Met Asn Gly Gln Pro Gln Glu Asn Tyr Lys Asn Thr Pro Pro Thr

145 150 155 160

Met Asp Thr Asp Gly Ser Tyr Phe Leu Tyr Ser Lys Leu Asn Val Lys

165 170 175

Lys Glu Thr Trp Gln Gln Gly Asn Thr Phe Thr Cys Ser Val Leu His

180 185 190

Glu Gly Leu His Asn His His Thr Glu Lys Ser Leu Ser His

195 200 205

<210> 14

<211> 206

<212> PRT

<213> Rabbit (Oryctolagus cuniculus)

<400> 14

Pro Ser Val Phe Ile Phe Pro Pro Lys Pro Lys Asp Thr Leu Met Ile

1 5 10 15

Ser Arg Thr Pro Glu Val Thr Cys Val Val Val Asp Val Ser Glu Asp

20 25 30

Asp Pro Glu Val Gln Phe Thr Trp Tyr Ile Asn Asn Glu Gln Val Arg

35 40 45

Thr Ala Arg Pro Pro Leu Arg Glu Gln Gln Phe Asn Ser Thr Ile Arg

50 55 60

Val Val Ser Thr Leu Pro Ile Ala His Glu Asp Trp Leu Arg Gly Lys

65 70 75 80

Glu Phe Lys Cys Lys Val His Asn Lys Ala Leu Pro Ala Pro Ile Glu

85 90 95

Lys Thr Ile Ser Lys Ala Arg Gly Gln Pro Leu Glu Pro Lys Val Tyr

100 105 110

Thr Met Gly Pro Pro Arg Glu Glu Leu Ser Ser Arg Ser Val Ser Leu

115 120 125

Thr Cys Met Ile Asn Gly Phe Tyr Pro Ser Asp Ile Ser Val Glu Trp

130 135 140

Glu Lys Asn Gly Lys Ala Glu Asp Asn Tyr Lys Thr Thr Pro Ala Val

145 150 155 160

Leu Asp Ser Asp Gly Ser Tyr Phe Leu Tyr Ser Lys Leu Ser Val Pro

165 170 175

Thr Ser Glu Trp Gln Arg Gly Asp Val Phe Thr Cys Ser Val Met His

180 185 190

Glu Ala Leu His Asn His Tyr Thr Gln Lys Ser Ile Ser Arg

195 200 205

<210> 15

<211> 208

<212> PRT

<213> general cow (Bos taurus)

<400> 15

Pro Ser Val Phe Ile Phe Pro Pro Lys Pro Lys Asp Thr Leu Thr Ile

1 5 10 15

Ser Gly Thr Pro Glu Val Thr Cys Val Val Val Asp Val Gly His Asp

20 25 30

Asp Pro Glu Val Lys Phe Ser Trp Phe Val Asp Asp Val Glu Val Asn

35 40 45

Thr Ala Thr Thr Lys Pro Arg Glu Glu Gln Phe Asn Ser Thr Tyr Arg

50 55 60

Val Val Ser Ala Leu Arg Ile Gln His Gln Asp Trp Thr Gly Gly Lys

65 70 75 80

Glu Phe Lys Cys Lys Val His Asn Glu Gly Leu Pro Ala Pro Ile Val

85 90 95

Arg Thr Ile Ser Arg Thr Lys Gly Pro Ala Arg Glu Pro Gln Val Tyr

100 105 110

Val Leu Ala Pro Pro Gln Glu Glu Leu Ser Lys Ser Thr Val Ser Leu

115 120 125

Thr Cys Met Val Thr Ser Phe Tyr Pro Asp Tyr Ile Ala Val Glu Trp

130 135 140

Gln Arg Asn Gly Gln Pro Glu Ser Glu Asp Lys Tyr Gly Thr Thr Pro

145 150 155 160

Pro Gln Leu Asp Ala Asp Gly Ser Tyr Phe Leu Tyr Ser Arg Leu Arg

165 170 175

Val Asp Arg Asn Ser Trp Gln Glu Gly Asp Thr Tyr Thr Cys Val Val

180 185 190

Met His Glu Ala Leu His Asn His Tyr Thr Gln Lys Ser Thr Ser Lys

195 200 205

<210> 16

<211> 208

<212> PRT

<213> general cow (Bos taurus)

<400> 16

Pro Ser Val Phe Ile Phe Pro Pro Lys Pro Lys Asp Thr Leu Met Ile

1 5 10 15

Thr Gly Thr Pro Glu Val Thr Cys Val Val Val Asn Val Gly His Asp

20 25 30

Asn Pro Glu Val Gln Phe Ser Trp Phe Val Asp Asp Val Glu Val His

35 40 45

Thr Ala Arg Ser Lys Pro Arg Glu Glu Gln Phe Asn Ser Thr Tyr Arg

50 55 60

Val Val Ser Ala Leu Pro Ile Gln His Gln Asp Trp Thr Gly Gly Lys

65 70 75 80

Glu Phe Lys Cys Lys Val Asn Asn Lys Gly Leu Ser Ala Pro Ile Val

85 90 95

Arg Ile Ile Ser Arg Ser Lys Gly Pro Ala Arg Glu Pro Gln Val Tyr

100 105 110

Val Leu Asp Pro Pro Lys Glu Glu Leu Ser Lys Ser Thr Leu Ser Val

115 120 125

Thr Cys Met Val Thr Gly Phe Tyr Pro Glu Asp Val Ala Val Glu Trp

130 135 140

Gln Arg Asn Arg Gln Thr Glu Ser Glu Asp Lys Tyr Arg Thr Thr Pro

145 150 155 160

Pro Gln Leu Asp Thr Asp Arg Ser Tyr Phe Leu Tyr Ser Lys Leu Arg

165 170 175

Val Asp Arg Asn Ser Trp Gln Glu Gly Asp Ala Tyr Thr Cys Val Val

180 185 190

Met His Glu Ala Leu His Asn His Tyr Met Gln Lys Ser Thr Ser Lys

195 200 205

<210> 17

<211> 208

<212> PRT

<213> domestic Cat (Felis cat)

<400> 17

Pro Ser Ile Phe Ile Phe Pro Pro Lys Pro Lys Asp Thr Leu Ser Ile

1 5 10 15

Ser Arg Thr Pro Glu Val Thr Cys Leu Val Val Asp Leu Gly Pro Asp

20 25 30

Asp Ser Asp Val Gln Ile Thr Trp Phe Val Asp Asn Thr Gln Val Tyr

35 40 45

Thr Ala Lys Thr Ser Pro Arg Glu Glu Gln Phe Asn Ser Thr Tyr Arg

50 55 60

Val Val Ser Val Leu Pro Ile Leu His Gln Asp Trp Leu Lys Gly Lys

65 70 75 80

Glu Phe Lys Cys Lys Val Asn Ser Lys Ser Leu Pro Ser Pro Ile Glu

85 90 95

Arg Thr Ile Ser Lys Asp Lys Gly Gln Pro His Glu Pro Gln Val Tyr

100 105 110

Val Leu Pro Pro Ala Gln Glu Glu Leu Ser Arg Asn Lys Val Ser Val

115 120 125

Thr Cys Leu Ile Glu Gly Phe Tyr Pro Pro Asp Ile Ala Val Glu Trp

130 135 140

Glu Ile Thr Gly Gln Pro Glu Pro Glu Asn Asn Tyr Arg Thr Thr Pro

145 150 155 160

Pro Gln Leu Asp Ser Asp Gly Thr Tyr Phe Leu Tyr Ser Arg Leu Ser

165 170 175

Val Asp Arg Ser Arg Trp Gln Arg Gly Asn Thr Tyr Thr Cys Ser Val

180 185 190

Ser His Glu Ala Leu His Ser His His Thr Gln Lys Ser Leu Thr Gln

195 200 205

<210> 18

<211> 209

<212> PRT

<213> dog family (Canis lupus family)

<400> 18

Pro Ser Val Leu Ile Phe Pro Pro Lys Pro Lys Asp Ile Leu Arg Ile

1 5 10 15

Thr Arg Thr Pro Glu Val Thr Cys Val Val Leu Asp Leu Gly Arg Glu

20 25 30

Asp Pro Glu Val Gln Ile Ser Trp Phe Val Asp Gly Lys Glu Val His

35 40 45

Thr Ala Lys Thr Gln Ser Arg Glu Gln Gln Phe Asn Gly Thr Tyr Arg

50 55 60

Val Val Ser Val Leu Pro Ile Glu His Gln Asp Trp Leu Thr Gly Lys

65 70 75 80

Glu Phe Lys Cys Arg Val Asn His Ile Asp Leu Pro Ser Pro Ile Glu

85 90 95

Arg Thr Ile Ser Lys Ala Arg Gly Arg Ala His Lys Pro Ser Val Tyr

100 105 110

Val Leu Pro Pro Ser Pro Lys Glu Leu Ser Ser Ser Asp Thr Val Ser

115 120 125

Ile Thr Cys Leu Ile Lys Asp Phe Tyr Pro Pro Asp Ile Asp Val Glu

130 135 140

Trp Gln Ser Asn Gly Gln Gln Glu Pro Glu Arg Lys His Arg Met Thr

145 150 155 160

Pro Pro Gln Leu Asp Glu Asp Gly Ser Tyr Phe Leu Tyr Ser Lys Leu

165 170 175

Ser Val Asp Lys Ser Arg Trp Gln Gln Gly Asp Pro Phe Thr Cys Ala

180 185 190

Val Met His Glu Thr Leu Gln Asn His Tyr Thr Asp Leu Ser Leu Ser

195 200 205

His

90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:制备热改性淀粉的方法

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!