Multispecific antibodies targeting HIV GP120 and CD3

文档序号:1471632 发布日期:2020-02-21 浏览:13次 中文

阅读说明:本技术 靶向hiv gp120和cd3的多特异性抗体 (Multispecific antibodies targeting HIV GP120 and CD3 ) 是由 T·西拉 J·戈德史密斯 C·S·佩斯 D·斯隆 H·斯蒂芬森 N·D·汤姆森 G·库克 于 2018-06-21 设计创作,主要内容包括:公开了结合HIV gp120和CD3的多特异性抗体(例如,双特异性抗体)。还公开了使用这些抗体治疗或预防HIV感染的方法。(Multispecific antibodies (e.g., bispecific antibodies) that bind HIV gp120 and CD3 are disclosed. Methods of using these antibodies to treat or prevent HIV infection are also disclosed.)

1. A multispecific antibody that binds human immunodeficiency virus-1 (HIV-1) envelope (Env) glycoprotein gp120(gp120) and human CD3, wherein the antibody comprises:

(a) a first antigen-binding domain comprising a first heavy chain variable domain (VH) and a first light chain variable domain (VL), wherein the first antigen-binding domain binds gp120 and comprises:

(i) a first VH-Complementarity Determining Region (CDR)1 comprising the amino acid sequence of SEQ ID NO 1;

(ii) a first VH-CDR2 comprising the amino acid sequence of SEQ ID NO 2;

(iii) a first VH-CDR3 comprising the amino acid sequence of SEQ ID NO. 3;

(iv) a first VL-CDR1 comprising the amino acid sequence of SEQ ID NO. 4;

(v) a first VL-CDR2 comprising the amino acid sequence of SEQ ID NO. 5; and

(vi) a first VL-CDR3 comprising the amino acid sequence of SEQ ID NO 6; and

(b) a second antigen binding domain that binds human CD 3.

2. The antibody of claim 1, wherein the first VH comprises an amino acid sequence at least 70% identical to SEQ ID No. 7.

3. The antibody of claim 1 or 2, wherein the first VL comprises an amino acid sequence at least 70% identical to SEQ ID No. 8.

4. The antibody of claim 3, wherein the amino acids at positions 66, 67A and 67C (Kabat numbering) of SEQ ID NO. 8 are unchanged.

5. The antibody of claim 1 or 2, wherein the first VL comprises an amino acid sequence at least 70% identical to SEQ ID No. 81.

6. The antibody of claim 1 or 2, wherein the first VL comprises an amino acid sequence at least 70% identical to SEQ ID No. 82.

7. The antibody of claim 1 or 2, wherein the first VL comprises an amino acid sequence at least 70% identical to SEQ ID NO 83.

8. The antibody of claim 1 or 2, wherein the first VL comprises an amino acid sequence at least 70% identical to SEQ ID NO: 84.

9. The antibody of any one of claims 1-8, wherein the first antigen binding domain comprises a heavy chain having an amino acid sequence at least 70% identical to SEQ id No. 9.

10. The antibody of any one of claims 1-9, wherein the first antigen binding domain comprises a light chain having an amino acid sequence at least 70% identical to SEQ id No. 10.

11. The antibody of claim 1, wherein the first VH comprises the amino acid sequence of SEQ ID No. 7 and the first VL comprises the amino acid sequence of SEQ ID No. 8.

12. The antibody of claim 1, wherein the first VH comprises the amino acid sequence of SEQ ID No. 7 and the first VL comprises the amino acid sequence of SEQ ID No. 81.

13. The antibody of claim 1, wherein the first VH comprises the amino acid sequence of SEQ ID No. 7 and the first VL comprises the amino acid sequence of SEQ ID No. 82.

14. The antibody of claim 1, wherein the first VH comprises the amino acid sequence of SEQ ID No. 7 and the first VL comprises the amino acid sequence of SEQ ID No. 83.

15. The antibody of claim 1, wherein the first VH comprises the amino acid sequence of SEQ ID No. 7 and the first VL comprises the amino acid sequence of SEQ ID No. 84.

16. The antibody of any one of claims 1-15, wherein the multispecific antibody is a bispecific antibody.

17. The antibody of any one of claims 1-16, wherein the second antigen-binding domain comprises a second VH and a second VL, wherein the second VH comprises:

(i) a second VH-CDR1 comprising the amino acid sequence of SEQ ID NO. 11;

(ii) a second VH-CDR2 comprising the amino acid sequence of SEQ ID NO 12; and

(iii) a second VH-CDR3 comprising the amino acid sequence of SEQ ID NO 13.

18. The antibody of any one of claims 1-16, wherein the second antigen-binding domain comprises a second VH and a second VL, wherein the second VL comprises:

(i) a second VL-CDR1 comprising the amino acid sequence of SEQ ID NO. 14;

(ii) a second VL-CDR2 comprising the amino acid sequence of SEQ ID NO. 15; and

(iii) a second VL-CDR3 comprising the amino acid sequence of SEQ ID NO 16.

19. The antibody of any one of claims 1-16, wherein the second antigen-binding domain comprises a second VH and a second VL, wherein

The second VH comprises:

(i) a second VH-CDR1 comprising the amino acid sequence of SEQ ID NO. 11;

(ii) a second VH-CDR2 comprising the amino acid sequence of SEQ ID NO 12; and

(iii) a second VH-CDR3 comprising the amino acid sequence of SEQ ID NO 13; and is

The second VL comprises:

(i) a second VL-CDR1 comprising the amino acid sequence of SEQ ID NO. 14;

(ii) a second VL-CDR2 comprising the amino acid sequence of SEQ ID NO. 15; and

(iii) a second VL-CDR3 comprising the amino acid sequence of SEQ ID NO 16.

20. The antibody of any one of claims 17-19, wherein the second VH comprises an amino acid sequence at least 70% identical to SEQ ID NO 17.

21. The antibody of any one of claims 17-19, wherein the second VL comprises an amino acid sequence that is at least 70% identical to SEQ ID No. 18.

22. The antibody of any one of claims 17-19, wherein the second VH comprises the amino acid sequence of SEQ ID No. 17 and the second VL comprises the amino acid sequence of SEQ ID No. 18.

23. The antibody of any one of claims 17-19, wherein the second antigen-binding domain comprises a heavy chain having an amino acid sequence at least 70% identical to SEQ id No. 19.

24. The antibody of any one of claims 17-19, wherein the second antigen-binding domain comprises a light chain having an amino acid sequence at least 70% identical to SEQ id No. 20.

25. The antibody of any one of claims 1-24, wherein the antibody is a kappa-lambda entity, an amphiphilic and re-targeting molecule (DART), a "knob" structure antibody, a chain exchange engineered domain antibody (SEEDbody), a bispecific T cell engager (BiTe), a CrossMab, an Fcab, a diabody, a tandem diabody (TandAb), or a DuoBody.

26. The antibody of any one of claims 1-24, wherein the first antigen binding domain is linked to a sequence selected from human IgG, either directly or by insertion of an amino acid sequence1Human IgG2Human IgG3Human IgG4Human IgA1And human IgA2And wherein the second antigen-binding domain is fused, directly or by insertion of an amino acid sequence, to a first heavy chain constant region selected from human IgG1Human IgG2Human IgG3Human IgG4Human IgA1And human IgA2Is fused to the second heavy chain constant region.

27. The antibody of claim 26, wherein the first heavy chain constant region is a human IgG1And wherein the second heavy chain constant region is human IgG1

28. The antibody of claim 26 or 27, wherein the first antigen-binding domain is fused, directly or by way of an intervening amino acid sequence, to a first light chain constant region that is a human λ constant region, and wherein the second antigen-binding domain is fused, directly or by way of an intervening amino acid sequence, to a second light chain constant region that is a human λ constant region.

29. The antibody of any one of claims 26-28, wherein

(a) The first heavy chain constant region comprises an F405L amino acid mutation; and

(b) the second heavy chain constant region comprises a K409R amino acid mutation.

30. The antibody of any one of claims 26-28, wherein

(a) The first heavy chain constant region comprises a K409R amino acid mutation; and

(b) the second heavy chain constant region comprises an F405L amino acid mutation.

31. The antibody of any one of claims 26-30, wherein effector functions of the first heavy chain constant region and the second heavy chain constant region are reduced or eliminated.

32. The antibody of any one of claims 26-30, wherein the first heavy chain constant region comprises a human IgG comprising the N297A mutation or the N297Q mutation1Heavy chainThe constant region and/or the second heavy chain constant region comprises a human IgG comprising the N297A mutation or the N297Q mutation1A heavy chain constant region.

33. A bispecific antibody that binds gp120 and human CD3, wherein the antibody comprises:

(A) a first arm that binds gp120, comprising:

(i) a first heavy chain comprising:

(a) a first heavy chain constant region comprising a first mutation that is an F405L or K409R mutation; and

(b) a first VH comprising a VH-CDR1 comprising the amino acid sequence of SEQ ID NO. 1, a VH-CDR2 comprising the amino acid sequence of SEQ ID NO.2, and a VH-CDR3 comprising the amino acid sequence of SEQ ID NO. 3; and

(ii) a first light chain comprising:

(a) a first light chain constant region; and

(b) a first VL comprising VL-CDR1 comprising the amino acid sequence of SEQ ID NO.4, VL-CDR2 comprising the amino acid sequence of SEQ ID NO.5 and VL-CDR3 comprising the amino acid sequence of SEQ ID NO. 6; and

(B) a second arm that binds human CD3, comprising:

(i) a second heavy chain comprising:

(a) a second heavy chain constant region comprising a second mutation that is an F405L or K409R mutation; and

(b) a second VH comprising a VH-CDR1 comprising the amino acid sequence of SEQ ID NO. 11, a VH-CDR2 comprising the amino acid sequence of SEQ ID NO. 12, and a VH-CDR3 comprising the amino acid sequence of SEQ ID NO. 13; and

(ii) a second light chain comprising:

(a) a second light chain constant region; and

(b) a second VL comprising VL-CDR1 comprising the amino acid sequence of SEQ ID NO. 14, VL-CDR2 comprising the amino acid sequence of SEQ ID NO. 15 and VL-CDR3 comprising the amino acid sequence of SEQ ID NO. 16,

wherein the first mutation and the second mutation are different mutations.

34. The antibody of claim 33, wherein the first VH comprises an amino acid sequence at least 70% identical to SEQ ID No. 7.

35. The antibody of claim 33 or 34, wherein the first VL comprises an amino acid sequence that is at least 70% identical to SEQ ID No. 8.

36. The antibody of claim 35, wherein the amino acids at positions 66, 67A and 67C (Kabat numbering) of SEQ ID NO. 8 are unchanged.

37. The antibody of any one of claims 33-36, wherein the first heavy chain comprises an amino acid sequence at least 70% identical to SEQ ID No. 9.

38. The antibody of any one of claims 33-37, wherein the first light chain comprises an amino acid sequence at least 70% identical to SEQ ID No. 10.

39. The antibody of any one of claims 33-38, wherein the second VH comprises an amino acid sequence at least 70% identical to SEQ ID NO 17.

40. The antibody of any one of claims 33-39, wherein the second VL comprises an amino acid sequence at least 70% identical to SEQ ID NO 18.

41. The antibody of any one of claims 33-40, wherein the second heavy chain comprises an amino acid sequence at least 70% identical to SEQ ID NO 19.

42. The antibody of any one of claims 33-41, wherein the second light chain comprises an amino acid sequence at least 70% identical to SEQ ID NO 20.

43. The antibody of claim 33, wherein the first VH comprises the amino acid sequence of SEQ ID No. 7 and the first VL comprises the amino acid sequence of SEQ ID No. 8, and/or the second VH comprises the amino acid sequence of SEQ ID No. 17 and the second VL comprises the amino acid sequence of SEQ ID No. 18.

44. The antibody of claim 33, wherein the first VH comprises the amino acid sequence of SEQ ID No. 7 and the first VL comprises the amino acid sequence of SEQ ID No. 81, and/or the second VH comprises the amino acid sequence of SEQ ID No. 17 and the second VL comprises the amino acid sequence of SEQ ID No. 18.

45. The antibody of claim 33, wherein the first VH comprises the amino acid sequence of SEQ ID No. 7 and the first VL comprises the amino acid sequence of SEQ ID No. 82, and/or the second VH comprises the amino acid sequence of SEQ ID No. 17 and the second VL comprises the amino acid sequence of SEQ ID No. 18.

46. The antibody of claim 33, wherein the first VH comprises the amino acid sequence of SEQ ID No. 7 and the first VL comprises the amino acid sequence of SEQ ID No. 83, and/or the second VH comprises the amino acid sequence of SEQ ID No. 17 and the second VL comprises the amino acid sequence of SEQ ID No. 18.

47. The antibody of claim 33, wherein the first VH comprises the amino acid sequence of SEQ ID No. 7 and the first VL comprises the amino acid sequence of SEQ ID No. 84, and/or the second VH comprises the amino acid sequence of SEQ ID No. 17 and the second VL comprises the amino acid sequence of SEQ ID No. 18.

48. The antibody of claim 33, wherein the first heavy chain comprises the amino acid sequence of SEQ ID NO 9 and the first light chain comprises the amino acid sequence of SEQ ID NO 10, and/or the second heavy chain comprises the amino acid sequence of SEQ ID NO 19 and the second light chain comprises the amino acid sequence of SEQ ID NO 20.

49. The antibody of claim 33, wherein the first heavy chain comprises the amino acid sequence of SEQ ID NO 9 and the first light chain comprises the amino acid sequence of SEQ ID NO 40, and/or the second heavy chain comprises the amino acid sequence of SEQ ID NO 19 and the second light chain comprises the amino acid sequence of SEQ ID NO 20.

50. The antibody of claim 33, wherein the first heavy chain comprises the amino acid sequence of SEQ ID No. 9 and the first light chain comprises the amino acid sequence of SEQ ID No. 78, and/or the second heavy chain comprises the amino acid sequence of SEQ ID No. 19 and the second light chain comprises the amino acid sequence of SEQ ID No. 20.

51. The antibody of claim 33, wherein the first heavy chain comprises the amino acid sequence of SEQ ID NO 9 and the first light chain comprises the amino acid sequence of SEQ ID NO 79, and/or the second heavy chain comprises the amino acid sequence of SEQ ID NO 19 and the second light chain comprises the amino acid sequence of SEQ ID NO 20.

52. The antibody of claim 33, wherein the first heavy chain comprises the amino acid sequence of SEQ ID NO 9 and the first light chain comprises the amino acid sequence of SEQ ID NO 80, and/or the second heavy chain comprises the amino acid sequence of SEQ ID NO 19 and the second light chain comprises the amino acid sequence of SEQ ID NO 20.

53. The antibody of any one of claims 1-52, further comprising a cytotoxic agent, a radioisotope, a therapeutic agent, an antiviral agent, or a detectable label.

54. A composition comprising (i) a nucleic acid molecule encoding a first light chain variable region or a first light chain of a first antigen-binding fragment of the antibody of any one of claims 1-52, (ii) a nucleic acid molecule encoding a first heavy chain variable region or a first heavy chain of a first antigen-binding fragment of the antibody of any one of claims 1-52, (iii) a nucleic acid molecule encoding a first light chain variable region or a first light chain of a second antigen-binding fragment of the antibody of any one of claims 1-52, and (iv) a nucleic acid molecule encoding a second heavy chain variable region or a second heavy chain of a second antigen-binding fragment of the antibody of any one of claims 1-52.

55. A host cell comprising (i) a nucleic acid molecule encoding a first light chain variable region or a first light chain of a first antigen-binding fragment of the antibody of any one of claims 1-52, (ii) a nucleic acid molecule encoding a first heavy chain variable region or a first heavy chain of a first antigen-binding fragment of the antibody of any one of claims 1-52, and/or (iii) a nucleic acid molecule encoding a first light chain variable region or a first light chain of a second antigen-binding fragment of the antibody of any one of claims 1-52, and (iv) a nucleic acid molecule encoding a second heavy chain variable region or a second heavy chain of a second antigen-binding fragment of the antibody of any one of claims 1-52.

56. The host cell of claim 55 selected from the group consisting of E.coli, Pseudomonas, Bacillus, Streptomyces, yeast, CHO, YB/20, NS0, PER-C6, HEK-293T, NIH-3T3, HeLa, BHK, Hep G2, SP2/0, R1.1, B-W, L-M, COS1, COS 7, BSC1, BSC40, BMT10 cells, plant cells, insect cells, and human cells in tissue culture.

57. A method of producing an antibody that binds gp120 and human CD3, the method comprising culturing the host cell of claim 55 or 56 under conditions such that the nucleic acid molecule is expressed and the antibody is produced.

58. A method for detecting gp120 and CD 3-expressing cells in a sample, the method comprising contacting the sample with the antibody of any one of claims 1-53.

59. A pharmaceutical composition comprising the antibody of any one of claims 1-53 and a pharmaceutically acceptable excipient.

60. A kit comprising an antibody of any one of claims 1-53 or a pharmaceutical composition of claim 59, and a) a detection reagent, b) a gp120 and/or CD3 antigen, c) a notice reflecting approval for use or sale for human administration, or d) a combination thereof.

61. A method of treating or preventing a human immunodeficiency virus infection in a human subject in need thereof, the method comprising administering to the human subject a therapeutically effective amount of the antibody of any one of claims 1-53 or the pharmaceutical composition of claim 59.

62. An antibody that binds gp120, wherein the antibody comprises a heavy chain variable domain (VH) and a light chain variable domain (VL), wherein the VH comprises a VH-CDR1 comprising the amino acid sequence of SEQ ID No. 1, a VH-CDR2 comprising the amino acid sequence of SEQ ID No.2, a VH-CDR3 comprising the amino acid sequence of SEQ ID No. 3, wherein the VL comprises a VL-CDR1 comprising the amino acid sequence of SEQ ID No.4, a VL-CDR2 comprising the amino acid sequence of SEQ ID No.5, and a VL-CDR3 comprising the amino acid sequence of SEQ ID No. 6, and wherein the VL comprises a tyrosine at position 67A (Kabat numbering), a phenylalanine at position 67A (Kabat numbering), a threonine at position 67A (Kabat numbering), or a glycine at position 67 (Kabat numbering).

63. The antibody of claim 62, wherein the VH comprises the amino acid sequence of SEQ ID NO 7.

64. The antibody of claim 62, wherein the VL comprises the amino acid sequence of SEQ ID NO 81, SEQ ID NO 82, SEQ ID NO 83 or SEQ ID NO 84.

65. The antibody of claim 62, wherein the VH comprises the amino acid sequence of SEQ ID NO 7 and the VL comprises the amino acid sequence of SEQ ID NO 81, SEQ ID NO 82, SEQ ID NO 83 or SEQ ID NO 84.

66. The antibody of claim 62, wherein the antibody comprises a heavy chain comprising the amino acid sequence set forth in SEQ ID NO. 9.

67. The antibody of claim 62, wherein the antibody comprises a light chain comprising an amino acid sequence set forth in any one of SEQ ID NOs 40, 78, 79, or 80.

68. The antibody of claim 62, wherein the antibody comprises a heavy chain comprising the amino acid sequence set forth in SEQ ID NO. 9 and a light chain comprising the amino acid sequence set forth in any one of SEQ ID NO. 40, 78, 79, or 80.

69. The antibody of any one of claims 62-68, further comprising a cytotoxic agent, a radioisotope, a therapeutic agent, an antiviral agent, or a detectable label.

70. A pharmaceutical composition comprising the antibody of any one of claims 62-69 and a pharmaceutically acceptable carrier.

71. One or more nucleic acids encoding the antibody of any one of claims 62-68.

72. One or more vectors comprising one or more nucleic acids of claim 71.

73. A host cell comprising one or more vectors of claim 72.

74. A method of producing an anti-gp 120 antibody, the method comprising culturing the host cell of claim 73 under conditions such that the one or more nucleic acids are expressed and the antibody is produced.

75. A method of treating or preventing a human immunodeficiency virus infection in a human subject in need thereof, the method comprising administering to the human subject a therapeutically effective amount of the antibody of any one of claims 62-69 or the pharmaceutical composition of claim 70.

76. An antibody fragment that binds gp120, wherein the antibody fragment comprises a heavy chain variable domain (VH) and a light chain variable domain (VL), wherein the VH comprises a VH-CDR1 comprising the amino acid sequence of SEQ ID NO:1, a VH-CDR2 comprising the amino acid sequence of SEQ ID NO:2, a VH-CDR3 comprising the amino acid sequence of SEQ ID NO:3, wherein the VL comprises a VL-CDR1 comprising the amino acid sequence of SEQ ID NO:4, a VL-CDR2 comprising the amino acid sequence of SEQ ID NO:5, and a VL-CDR3 comprising the amino acid sequence of SEQ ID NO:6, and wherein the VL comprises a tyrosine at position 67A (Kabat numbering), a phenylalanine at position 67A (Kabat numbering), a threonine at position 67A (Kabat numbering), or a glycine at position 67 (Kabat numbering).

77. The antibody fragment of claim 76, wherein said VH comprises the amino acid sequence of SEQ ID NO 7.

78. The antibody fragment of claim 76, wherein said VL comprises the amino acid sequence of SEQ ID NO 81, SEQ ID NO 82, SEQ ID NO 83 or SEQ ID NO 84.

79. The antibody fragment of claim 76, wherein said VH comprises the amino acid sequence of SEQ ID NO 7 and said VL comprises the amino acid sequence of SEQ ID NO 81, SEQ ID NO 82, SEQ ID NO 83 or SEQ ID NO 84.

80. The antibody fragment of any one of claims 76 to 79, which is a Fab, F (ab)2, Fv, scFv, sc (Fv)2, or diabody.

81. The antibody fragment of any one of claims 76-80, further comprising a cytotoxic agent, a radioisotope, a therapeutic agent, an antiviral agent, or a detectable label.

82. A pharmaceutical composition comprising the antibody fragment of any one of claims 76-81 and a pharmaceutically acceptable carrier.

83. One or more nucleic acids encoding the antibody fragment of any one of claims 76-80.

84. One or more vectors comprising one or more nucleic acids of claim 83.

85. A host cell comprising one or more vectors of claim 84.

86. A method of producing an anti-gp 120 antibody fragment, the method comprising culturing the host cell of claim 85 under conditions such that the one or more nucleic acids are expressed and the antibody fragment is produced.

Technical Field

The present disclosure relates to antibodies for the treatment and prevention of Human Immunodeficiency Virus (HIV) infection. In particular, provided herein are multispecific antibodies including broadly neutralizing anti-HIV antibodies, and methods of using these antibodies to reduce HIV replication and to treat and prevent HIV infection.

Background

Human Immunodeficiency Virus (HIV) infection and related diseases are a major public health problem worldwide. Most currently approved therapies for HIV infection target viral reverse transcriptase, protease, and integrase, but resistance of HIV to these existing drugs, long-term toxicity, and inadequate patient compliance with daily dosing regimens have been problems associated with these therapies. Therefore, it is important to discover and develop new HIV drugs.

WO2012/030904 describes human anti-HIV antibodies derived from memory B cells of HIV-infected donors, which are capable of inhibiting infection of HIV-1 species from multiple clades. However, the therapeutic use of these antibodies is limited due to problems with immunogenicity, pharmacokinetics, antigen specificity, effector function, and manufacture. Thus, there is a need in the art for novel anti-HIV antibodies with advantageous properties for therapeutic use.

Disclosure of Invention

The present disclosure provides, inter alia, compositions and methods for treating or preventing HIV. More specifically, provided herein are multispecific antibodies targeting Human Immunodeficiency Virus (HIV) envelope (Env) glycoprotein GP120(GP120) and a second antigen (e.g., cluster of differentiation 3(CD 3); anti-IgA receptor (CD89)), and uses thereof.

In one aspect, the disclosure provides multispecific antibodies that bind human immunodeficiency virus-1 (HIV-1) envelope (Env) glycoprotein gp120(gp120) and human CD3 (e.g., human CD3 epsilon). The antibody comprises a first antigen binding domain comprising a first heavy chain variable domain (VH) and a first light chain variable domain (VL). The first antigen binding domain binds gp120 and comprises a first VH-Complementarity Determining Region (CDR)1 comprising an amino acid sequence of SEQ ID NO 1; a first VH-CDR2 comprising the amino acid sequence of SEQ ID NO. 2; a first VH-CDR3 comprising the amino acid sequence of SEQ ID NO. 3; a first VL-CDR1 comprising the amino acid sequence of SEQ ID NO. 4; a first VL-CDR2 comprising the amino acid sequence of SEQ ID NO. 5; and a first VL-CDR3 comprising the amino acid sequence of SEQ ID NO. 6. The antibody further comprises a second antigen-binding domain that binds human CD3 (e.g., human CD3 epsilon). In certain embodiments, the anti-gp 120 antibody binds to a polypeptide comprising SEQ ID NO: 21 or a protein consisting thereof. In some cases, the anti-gp 120 antibody binds to a polypeptide comprising SEQ ID NO: 38 or a protein consisting thereof. In some cases, the anti-gp 120 antibody binds to free HIV-1 virus. In some cases, anti-gp 120 antibodies bind to HIV-1 infected cells. In some cases, the anti-gp 120 antibody binds to both free HIV-1 virus and HIV-1 infected cells. In certain instances, the anti-gp 120 antibody binds to at least two different HIV-1 strains (e.g., group M, group N, group O, or group P). In one embodiment, the anti-gp 120 antibody binds pwito.c/2474 (accession No. JN944948 and NIH AIDS Reagent Program catalog No. 11739). In another embodiment, the anti-gp 120 antibody binds to pCH058.c/2960 (accession No. JN944940 and NIH AIDS Reagent Program catalog No. 700010058).

In another aspect, the disclosure provides multispecific antibodies that bind human immunodeficiency virus-1 (HIV-1) envelope (Env) glycoprotein gp120(gp120) and IgA receptor CD 89. The antibody comprises a first antigen binding domain comprising a first heavy chain variable domain (VH) and a first light chain variable domain (VL). The first antigen binding domain binds gp120 and comprises a heavy chain variable region comprising seq id NO:1, a first VH-Complementarity Determining Region (CDR)1 comprising the amino acid sequence of SEQ ID NO: 2, a first VH-CDR2 comprising the amino acid sequence of SEQ ID NO:3, a first VH-CDR3 comprising the amino acid sequence of SEQ ID NO:4, a first VL-CDR1 comprising the amino acid sequence of SEQ ID NO:5 and a first VL-CDR2 comprising the amino acid sequence of SEQ ID NO:6, or a first VL-CDR3 of the amino acid sequence of seq id No. 6. The antibody also comprises a second antigen-binding domain that binds CD89 (e.g., human CD 89/FCAR; UniProtKB-P24071). In certain embodiments, the anti-gp 120 antibody binds to a polypeptide comprising SEQ ID NO: 21 or a protein consisting thereof. In some cases, the anti-gp 120 antibody binds to a polypeptide comprising SEQ ID NO: 38 or a protein consisting thereof. In some cases, the anti-gp 120 antibody binds to free HIV-1 virus. In some cases, anti-gp 120 antibodies bind to HIV-1 infected cells. In some cases, anti-gp 120 antibodies bind to free HIV-1 virus and HIV-1 infected cells. In certain instances, the anti-gp 120 antibody binds to at least two different HIV-1 strains (e.g., group M, group N, group O, or group P). In one embodiment, the anti-gp 120 antibody binds pwito.c/2474 (accession No. JN944948 and NIH AIDS Reagent Program catalog No. 11739). In another embodiment, the anti-gp 120 antibody binds to pCH058.c/2960 (accession No. JN944940 and NIH AIDS Reagent Program catalog No. 700010058).

In some embodiments of the above two aspects, the first VH comprises a VH sequence identical to SEQ ID NO:7 at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identical. In some embodiments, the first VL comprises a sequence identical to SEQ ID NO: 8 at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identical. In certain instances of these embodiments, SEQ ID NO: 8 at one or more of positions 66, 67A and 67C (Kabat numbering). In certain embodiments, the first VL comprises an amino acid sequence identical to SEQ ID NO: 81 at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identical. In certain embodiments, the first VL comprises an amino acid sequence identical to SEQ ID NO: 82 at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identical. In certain embodiments, the first VL comprises an amino acid sequence identical to SEQ ID NO: 83 at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identical. In certain embodiments, the first VL comprises an amino acid sequence identical to SEQ ID NO: 84 are at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identical. In some cases, the VH described above is identical to SEQ ID NO: 56-65. In other cases, the VH described above is identical to SEQ ID NO: 66-75, to each other. In some cases, the VH described above is directly linked to a VH having the amino acid sequence of SEQ id no:77 (e.g., substitutions that increase half-life and/or decrease effector function). In certain embodiments, the aforementioned VH is linked, directly or by intervening amino acids (e.g., a G-S linker sequence), to an amino acid sequence comprising a CH1 domain, a CH2 domain, and a CH3 domain from IgG1 (e.g., human IgG1, e.g., IgG1m3 allotype), and an IgG3 hinge region (e.g., the "open" IgG3 hinge variant "IgG 3C-" described in WO 2017/096221).

In some embodiments of the above two aspects, the first antigen binding domain comprises a polypeptide having an amino acid sequence identical to SEQ ID NO: 9, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identical. In some embodiments, the first antigen binding domain comprises a polypeptide having an amino acid sequence identical to SEQ ID NO: 10 at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identical. In some embodiments, the first antigen binding domain comprises a polypeptide having an amino acid sequence identical to SEQ ID NO: 40, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identical. In some embodiments, the first antigen binding domain comprises a polypeptide having an amino acid sequence identical to SEQ ID NO: 78 at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identical. In some embodiments, the first antigen binding domain comprises a polypeptide having an amino acid sequence identical to SEQ ID NO: 79, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%. In some embodiments, the first antigen binding domain comprises a polypeptide having an amino acid sequence identical to SEQ ID NO: 80, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identical.

In certain embodiments of the above two aspects, the first VH comprises SEQ ID NO:7 and the first VL comprises the amino acid sequence of SEQ ID NO: 8. In other embodiments, the first VH comprises SEQ ID NO:7 and the first VL comprises the amino acid sequence of SEQ ID NO: 81, or a pharmaceutically acceptable salt thereof. In other embodiments, the first VH comprises seq id NO:7 and the first VL comprises the amino acid sequence of SEQ ID NO: 82. In other embodiments, the first VH comprises SEQ ID NO:7 and the first VL comprises the amino acid sequence of SEQ ID NO: 83 of the sequence listing. In other embodiments, the first VH comprises SEQ ID NO:7 and the first VL comprises the amino acid sequence of SEQ ID NO: 84. In some cases, the VH described above is identical to SEQ ID NO: 56-65. In other cases, the VH described above is identical to SEQ ID NO: 66-75, to each other. In some cases, the VH described above may be directly linked to a VH having the amino acid sequence of SEQ ID NO:77 (e.g., substitutions that increase half-life and/or decrease effector function). In certain instances, the aforementioned VH is linked, directly or through one or more intermediate amino acids (e.g., a G-S linker sequence), to an amino acid sequence comprising the CH1, CH2 and CH3 domains of IgG1 (e.g., human IgG1, e.g., IgG1m3 allotype), and an IgG3 hinge region (e.g., the "open" IgG3 hinge variant "IgG 3C-" described in WO 2017/096221).

In some embodiments of the above two aspects, the first antigen binding domain comprises a polypeptide having the amino acid sequence of SEQ ID NO: 9 and comprises a light chain having the amino acid sequence set forth in SEQ ID NO: 10, or a light chain of the amino acid sequence set forth in seq id No. 10. In some embodiments, the first antigen binding domain comprises a polypeptide having the amino acid sequence of SEQ ID NO: 9 and comprises a light chain having the amino acid sequence set forth in SEQ ID NO: 40, or a light chain of the amino acid sequence set forth in seq id no. In some embodiments, the first antigen binding domain comprises a polypeptide having the amino acid sequence of SEQ ID NO: 9 and comprises a light chain having the amino acid sequence set forth in SEQ ID NO: 78, or a light chain of the amino acid sequence set forth in seq id no. In some embodiments, the first antigen binding domain comprises a polypeptide having the amino acid sequence of SEQ ID NO: 9 and comprises a light chain having the amino acid sequence set forth in SEQ ID NO: 79, or a pharmaceutically acceptable salt thereof. In some embodiments, the first antigen binding domain comprises a polypeptide having the amino acid sequence of SEQ ID NO: 9 and comprises a heavy chain having the amino acid sequence set forth in seq id NO: 80, or a light chain of the amino acid sequence set forth in seq id no.

In certain embodiments, the multispecific antibody is a bispecific antibody.

In certain embodiments, the second antigen-binding domain binds human CD3 and comprises a second VH and a second VL. The second VH comprises a VH comprising SEQ ID NO: 11, a second VH-CDR1 comprising the amino acid sequence of SEQ ID NO:12 and a second VH-CDR2 comprising the amino acid sequence of SEQ ID NO: 13, and a second VH-CDR3 of the amino acid sequence of seq id No. 13. In some embodiments, the second VL comprises a vh comprising SEQ ID NO: 14, a second VL-CDR1 comprising the amino acid sequence of SEQ ID NO: 15 and a second VL-CDR2 comprising the amino acid sequence of SEQ ID NO: 16, or a second VL-CDR3 of the amino acid sequence of seq id No. 16.

In certain embodiments, the second antigen-binding domain binds human CD3 and comprises a second VH and a second VL, wherein the second VH comprises a VH comprising SEQ ID NO: 11, a second VH-CDR1 comprising the amino acid sequence of SEQ ID NO:12 and a second VH-CDR2 comprising the amino acid sequence of SEQ ID NO: 13, a second VH-CDR3 of the amino acid sequence of seq id no; and wherein the second VL comprises a vh comprising SEQ ID NO: 14, a second VL comprising the amino acid sequence of SEQ ID NO: 15 and a second VL-CDR2 comprising the amino acid sequence of SEQ ID NO: 16, or a second VL-CDR3 of the amino acid sequence of seq id No. 16.

In certain embodiments, the second antigen-binding domain binds human CD89 and comprises a second VH and a second VL. The second VH comprises a VH comprising SEQ ID NO: 98, a second VH-CDR1 comprising the amino acid sequence of SEQ ID NO: 99 and a second VH-CDR2 comprising the amino acid sequence of SEQ ID NO: 100, and a second VH-CDR3 of the amino acid sequence of seq id no. In some embodiments, the second VL comprises a vh comprising SEQ ID NO: 103, a second VL-CDR1 comprising the amino acid sequence of SEQ ID NO: 104 and a second VL-CDR2 comprising the amino acid sequence of SEQ ID NO: 105, or a second VL-CDR3 of the amino acid sequence of seq id no.

In certain embodiments, the second antigen-binding domain binds human CD89 and comprises a second VH and a second VL, wherein the second VH comprises a VH comprising SEQ ID NO: 98, a second VH-CDR1 comprising the amino acid sequence of SEQ ID NO: 99 and a second VH-CDR2 comprising the amino acid sequence of SEQ ID NO: 100, a second VH-CDR3 of the amino acid sequence; and wherein the second VL comprises a vh comprising SEQ ID NO: 103, a second VL-CDR1 comprising the amino acid sequence of SEQ ID NO: 104 and a second VL-CDR2 comprising the amino acid sequence of SEQ ID NO: 105, or a second VL-CDR3 of the amino acid sequence of seq id no.

In some embodiments, the second VH comprises a VH sequence identical to SEQ ID NO: 17, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identical. In some embodiments, the second VL comprises a sequence identical to SEQ ID NO: 18, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identical.

In one embodiment, the second VH comprises SEQ ID NO: 17, and the second VL comprises the amino acid sequence set forth in SEQ ID NO: 18, or a pharmaceutically acceptable salt thereof.

In some embodiments, the second VH comprises a VH sequence identical to SEQ ID NO: 96 at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identical. In some embodiments, the second VL comprises a sequence identical to SEQ ID NO: 101 at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identical.

In one embodiment, the second VH comprises SEQ ID NO: 96 and the second VL comprises the amino acid sequence set forth in SEQ ID NO: 101, or a pharmaceutically acceptable salt thereof.

In certain embodiments, the second antigen-binding domain comprises a polypeptide having an amino acid sequence that is identical to SEQ ID NO: 19 at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identical. In some embodiments, the second antigen-binding domain comprises a polypeptide having an amino acid sequence that is identical to SEQ ID NO: 20, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identical.

In certain embodiments, the second antigen-binding domain comprises a polypeptide having an amino acid sequence that is identical to SEQ ID NO: 97 at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identical. In some embodiments, the second antigen-binding domain comprises a polypeptide having an amino acid sequence that is identical to SEQ ID NO: 102, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identical.

In some embodiments, the antibody is a kappa-lambda entity, an amphipathic and retargeting molecule (DART), "knob-in-hole" structural antibody (knob-in-hole), a chain exchange engineered domain antibody (SEEDbody), a bispecific T cell engager (BiTe), a CrossMab, an Fcab, a diabody, a tandem diabody (TandAb), or

Figure BDA0002328692870000091

In certain embodiments, the first antigen binding domain is fused, directly or by insertion of an amino acid sequence, to a first heavy chain constant region selected from the group consisting of human IgG1, human IgG2, human IgG3, human IgG4, human IgA1, and human IgA 2. In some cases, the first antigen-binding domain is fused, directly or by insertion of an amino acid sequence, to a first heavy chain constant region, wherein the constant region is from human IgG1 (e.g., an IgG1m3 allotype), except that the IgG1 hinge region is replaced with an IgG3 hinge region (e.g., the "open" IgG3 hinge variant "IgG 3C-" described in WO 2017/096221). In some embodiments, the second antigen-binding domain is fused, directly or by insertion of an amino acid sequence, to a second heavy chain constant region selected from the group consisting of human IgG1, human IgG2, human IgG3, human IgG4, human IgA1, and human IgA 2. In some cases, the second antigen-binding domain is fused, directly or by insertion of an amino acid sequence, to a second heavy chain constant region, wherein the constant region is from human IgG1 (e.g., an IgG1m3 allotype), except that the IgG1 hinge region is replaced by an IgG3 hinge region (e.g., the "open" IgG3 hinge variant "IgG 3C-" described in WO 2017/096221).

In a specific embodiment, the first heavy chain constant region is human IgG1 and the second heavy chain constant region is human IgG 1.

In some embodiments, the first antigen binding domain is fused, directly or by insertion of an amino acid sequence, to a first light chain constant region that is a human λ constant region. In other embodiments, the second antigen-binding domain is fused, directly or by insertion of an amino acid sequence, to a second light chain constant region that is a human lambda constant region.

In some embodiments, the first heavy chain constant region comprises one of: F405L, F405A, F405D, F405E, F405H, F405I, F405K, F405M, F405N, F405Q, F405S, F405T, F405V, F405W, or F405Y amino acid mutation and the second heavy chain constant region comprises a K409R amino acid mutation. In one instance, the first heavy chain constant region comprises the F405L amino acid mutation and the second heavy chain constant region comprises the K409R amino acid mutation. In another instance, the first heavy chain constant region comprises the F405A amino acid mutation and the second heavy chain constant region comprises the K409R amino acid mutation. In another instance, the first heavy chain constant region comprises the F405D amino acid mutation and the second heavy chain constant region comprises the K409R amino acid mutation. In another instance, the first heavy chain constant region comprises the F405E amino acid mutation and the second heavy chain constant region comprises the K409R amino acid mutation. In another instance, the first heavy chain constant region comprises the F405H amino acid mutation and the second heavy chain constant region comprises the K409R amino acid mutation. In another instance, the first heavy chain constant region comprises the F405I amino acid mutation and the second heavy chain constant region comprises the K409R amino acid mutation. In another instance, the first heavy chain constant region comprises the F405K amino acid mutation and the second heavy chain constant region comprises the K409R amino acid mutation. In another instance, the first heavy chain constant region comprises the F405M amino acid mutation and the second heavy chain constant region comprises the K409R amino acid mutation. In another instance, the first heavy chain constant region comprises the F405N amino acid mutation and the second heavy chain constant region comprises the K409R amino acid mutation. In another instance, the first heavy chain constant region comprises the F405Q amino acid mutation and the second heavy chain constant region comprises the K409R amino acid mutation. In another instance, the first heavy chain constant region comprises the F405S amino acid mutation and the second heavy chain constant region comprises the K409R amino acid mutation. In another instance, the first heavy chain constant region comprises the F405T amino acid mutation and the second heavy chain constant region comprises the K409R amino acid mutation. In another instance, the first heavy chain constant region comprises the F405V amino acid mutation and the second heavy chain constant region comprises the K409R amino acid mutation. In another instance, the first heavy chain constant region comprises the F405W amino acid mutation and the second heavy chain constant region comprises the K409R amino acid mutation. In another instance, the first heavy chain constant region comprises the F405Y amino acid mutation and the second heavy chain constant region comprises the K409R amino acid mutation.

In another embodiment, the first heavy chain constant region comprises the K409R amino acid mutation and the second heavy chain constant region comprises one of: F405L, F405A, F405D, F405E, F405H, F405I, F405K, F405M, F405N, F405Q, F405S, F405T, F405V, F405W, or F405Y amino acid mutation. In one instance, the first heavy chain constant region comprises the K409R amino acid mutation and the second heavy chain constant region comprises the F405L amino acid mutation. In another instance, the first heavy chain constant region comprises the K409R amino acid mutation and the second heavy chain constant region comprises the F405A amino acid mutation. In one instance, the first heavy chain constant region comprises the K409R amino acid mutation and the second heavy chain constant region comprises the F405D amino acid mutation. In one instance, the first heavy chain constant region comprises the K409R amino acid mutation and the second heavy chain constant region comprises the F405E amino acid mutation. In one instance, the first heavy chain constant region comprises the K409R amino acid mutation and the second heavy chain constant region comprises the F405H amino acid mutation. In one instance, the first heavy chain constant region comprises the K409R amino acid mutation and the second heavy chain constant region comprises the F405I amino acid mutation. In one instance, the first heavy chain constant region comprises the K409R amino acid mutation and the second heavy chain constant region comprises the F405K amino acid mutation. In one instance, the first heavy chain constant region comprises the K409R amino acid mutation and the second heavy chain constant region comprises the F405M amino acid mutation. In one instance, the first heavy chain constant region comprises the K409R amino acid mutation and the second heavy chain constant region comprises the F405N amino acid mutation. In one instance, the first heavy chain constant region comprises the K409R amino acid mutation and the second heavy chain constant region comprises the F405Q amino acid mutation. In one instance, the first heavy chain constant region comprises the K409R amino acid mutation and the second heavy chain constant region comprises the F405S amino acid mutation. In one instance, the first heavy chain constant region comprises the K409R amino acid mutation and the second heavy chain constant region comprises the F405T amino acid mutation. In one instance, the first heavy chain constant region comprises the K409R amino acid mutation and the second heavy chain constant region comprises the F405V amino acid mutation. In one instance, the first heavy chain constant region comprises the K409R amino acid mutation and the second heavy chain constant region comprises the F405W amino acid mutation. In one instance, the first heavy chain constant region comprises the K409R amino acid mutation and the second heavy chain constant region comprises the F405Y amino acid mutation.

In certain embodiments, the effector function of the first heavy chain constant region and the second heavy chain constant region is reduced or eliminated (e.g., relative to the effector function of an antibody having wild-type IgG1 Fc).

In some embodiments, the first heavy chain constant region comprises a human IgG1 heavy chain constant region comprising an N297A mutation or an N297Q mutation, and/or the second heavy chain constant region comprises a human IgG1 heavy chain constant region comprising an N297A mutation or an N297Q mutation.

In another aspect, the disclosure provides bispecific antibodies that bind gp120 and human CD 3. The bispecific antibody comprises a first arm that binds gp 120. The first arm comprises a first heavy chain comprising a first heavy chain constant region comprising a first mutation that is any one of an F405L, F405A, F405D, F405E, F405H, F405I, F405K, F405M, F405N, F405Q, one of F405T, F405V, F405W, or F405Y amino acid mutations in F405S, or a K409R mutation; and a first VH comprising a VH comprising SEQ ID NO:1, VH-CDR1 comprising the amino acid sequence of SEQ ID NO: 2 comprises and comprises the amino acid sequence of SEQ ID NO:3, VH-CDR3 of the amino acid sequence of seq id no. In one instance, the first arm comprises a first heavy chain comprising a first heavy chain constant region comprising a first mutation that is one of the F405L or K409R mutations. The first arm further comprises a first light chain comprising a first light chain constant region; and a first VL comprising a vh comprising SEQ ID NO:4, VL-CDR1 comprising the amino acid sequence of SEQ ID NO:5 and a VL-CDR2 comprising the amino acid sequence of SEQ ID NO:6, VL-CDR3 of the amino acid sequence of seq id no. The bispecific antibody comprises a second arm that binds CD3 (e.g., human CD3 (e.g., human CD3 epsilon)). The second arm comprises a second heavy chain comprising a second heavy chain constant region comprising a second mutation that is any one of an amino acid mutation of F405L, F405A, F405D, F405E, F405H, F405I, F405K, F405M, F405N, F405Q, F405S, F405T, F405V, F405W, or F405Y, or a K409R mutation; and a second VH comprising a VH comprising SEQ ID NO: 11, VH-CDR1 comprising the amino acid sequence of SEQ ID NO:12 and a VH-CDR2 comprising the amino acid sequence of SEQ ID NO: 13, VH-CDR3 of the amino acid sequence of seq id No. 13. In one instance, the second arm comprises a second heavy chain comprising a second mutated second heavy chain constant region comprising one of the F405L or K409R mutations. The second arm comprises a second light chain comprising a second light chain constant region; and a second VL comprising a vh comprising SEQ ID NO: 14, VL-CDR1 comprising the amino acid sequence of SEQ ID NO: 15 and VL-CDR2 comprising the amino acid sequence of SEQ ID NO: 16, VL-CDR3 of the amino acid sequence of seq id No. 16. The first mutation and the second mutation are different mutations.

In another aspect, the disclosure provides bispecific antibodies that bind gp120 and human CD 89. The bispecific antibody comprises a first arm that binds gp 120. The first arm comprises a first heavy chain comprising a first heavy chain constant region comprising a first mutation that is any one of an amino acid mutation of F405L, F405A, F405D, F405E, F405H, F405I, F405K, F405M, F405N, F405Q, F405S, F405T, F405V, F405W, or F405Y, or a K409R mutation; and a first VH comprising a VH comprising SEQ ID NO:1, VH-CDR1 comprising the amino acid sequence of SEQ ID NO: 2 and VH-CDR2 comprising the amino acid sequence of SEQ ID NO:3, VH-CDR3 of the amino acid sequence of seq id no. In one instance, the first arm comprises a first heavy chain comprising a first heavy chain constant region comprising a first mutation that is either a F405L or a K409R mutation. The first arm further comprises a first light chain comprising a first light chain constant region; and a first VL comprising a vh comprising SEQ ID NO:4, VL-CDR1 comprising the amino acid sequence of SEQ ID NO:5 and a VL-CDR2 comprising the amino acid sequence of SEQ ID NO:6, VL-CDR3 of the amino acid sequence of seq id no. The bispecific antibody comprises a second arm that binds CD89 (e.g., human CD 89). The second arm comprises a second heavy chain comprising a second mutant second heavy chain constant region comprising a second mutation that is any one of an F405L, F405A, F405D, F405E, F405H, F405I, F405K, F405M, F405N, F405Q, F405S, F405T, F405V, F405W, or F405Y amino acid mutation, or a K409R mutation; a second VH comprising a VH comprising SEQ ID NO: 98, VH-CDR1 comprising the amino acid sequence of SEQ ID NO: 99 and a VH-CDR2 comprising the amino acid sequence of SEQ ID NO: 100, or a VH-CDR3 of the amino acid sequence of seq id no. In one instance, the second arm comprises a second heavy chain comprising a second heavy chain constant region comprising a second mutation that is either a F405L or a K409R mutation. The second arm comprises a second light chain comprising a second light chain constant region; and a second VL comprising a vh comprising SEQ ID NO: 103, VL-CDR1 comprising the amino acid sequence of SEQ ID NO: 104 and VL-CDR2 comprising the amino acid sequence of SEQ ID NO: 105, VL-CDR3 of the amino acid sequence. The first mutation and the second mutation are different mutations.

In certain embodiments of the two aspects above, the anti-gp 120 antibody arm binds to a polypeptide comprising or consisting of SEQ ID NO: 21, and a protein consisting of the amino acid sequence shown in the figure 21. In some cases, the anti-gp 120 antibody arm binds to a polypeptide comprising or consisting of SEQ ID NO: 38, or a pharmaceutically acceptable salt thereof. In some cases, the anti-gp 120 antibody arm binds to free HIV-1 virus. In some cases, anti-gp 120 antibodies bind to HIV-1 infected cells. In some cases, the anti-gp 120 antibody arm binds free HIV-1 virus and HIV-1 infected cells. In certain instances, the anti-gp 120 antibody binds to at least two different HIV-1 strains (e.g., group M, group N, group O, or group P). In one embodiment, the anti-gp 120 antibody arm binds pwito.c/2474 (accession No. JN944948 and NIH AIDS Reagent Program catalog No. 11739). In another embodiment, the anti-gp 120 antibody arm binds to pCH058.c/2960 (accession No. JN944940 and NIH AIDS Reagent Program catalog No. 700010058).

In some embodiments, the first VH comprises a VH sequence identical to SEQ ID NO:7 at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identical. In some embodiments, the first VL comprises a sequence identical to SEQ ID NO: 8 at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identical. In certain instances of these embodiments, SEQ ID NO: 8 (Kabat numbering) is unchanged at one or more of positions 66, 67A and 67C (Kabat numbering). In certain embodiments, the first VL comprises an amino acid sequence identical to SEQ ID NO: 81 at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identical. In certain embodiments, the first VL comprises an amino acid sequence identical to SEQ ID NO: 82 at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identical. In certain embodiments, the first VL comprises an amino acid sequence identical to SEQ ID NO: 83 at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identical. In certain embodiments, the first VL comprises an amino acid sequence identical to SEQ ID NO: 84, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identical. In some cases, the VH described above is directly or through insertion of an amino acid (e.g., a G-S linker sequence) to a VH having the sequence of SEQ ID NO:77 (e.g., substitutions that increase half-life and/or decrease effector function).

In some embodiments, the first heavy chain comprises a heavy chain variable region identical to SEQ ID NO: 9, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identical. In some embodiments, the first light chain comprises a heavy chain variable region identical to SEQ ID NO: 10 at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identical. In some embodiments, the first light chain comprises a heavy chain variable region identical to SEQ ID NO: 40, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identical. In some embodiments, the first light chain comprises a heavy chain variable region identical to SEQ ID NO: 78 at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identical. In some embodiments, the first light chain comprises a heavy chain variable region identical to SEQ ID NO: 79, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100%. In some embodiments, the first light chain comprises a heavy chain variable region identical to SEQ ID NO: 80 at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identical.

In certain embodiments, the second VH comprises a VH sequence identical to SEQ ID NO: 17, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identical. In some embodiments, the second VL comprises a sequence identical to SEQ ID NO: 18, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identical.

In certain embodiments, the second VH comprises a VH sequence identical to SEQ ID NO: 96 at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identical. In some embodiments, the second VL comprises a sequence identical to SEQ ID NO: 101 at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identical.

In some embodiments, the second heavy chain comprises a heavy chain variable region identical to SEQ ID NO: 19 at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identical. In certain embodiments, the second light chain comprises a sequence identical to SEQ ID NO: 20 at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identical.

In some embodiments, the second heavy chain comprises a heavy chain variable region identical to SEQ ID NO: 97 at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identical. In certain embodiments, the second light chain comprises a sequence identical to SEQ ID NO: 102 at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identical.

In one embodiment, the first VH comprises SEQ ID NO:7 and the first VL comprises the amino acid sequence of SEQ id no: 8, and/or the second VH comprises the amino acid sequence of SEQ ID NO: 17 and the second VL comprises the amino acid sequence of SEQ id no: 18.

In one embodiment, the first VH comprises SEQ ID NO:7 and the first VL comprises the amino acid sequence of SEQ id no: 8, and/or the second VH comprises the amino acid sequence of SEQ ID NO: 96 and the second VL comprises the amino acid sequence of SEQ id no: 101, or a pharmaceutically acceptable salt thereof.

In another embodiment, the first VH comprises SEQ ID NO:7 and the first VL comprises seq id NO: 81, and/or the second VH comprises the amino acid sequence of SEQ ID NO: 17 and the second VL comprises the amino acid sequence of seq id NO: 18.

In another embodiment, the first VH comprises SEQ ID NO:7 and the first VL comprises seq id NO: 81, and/or the second VH comprises the amino acid sequence of SEQ ID NO: 96 and the second VL comprises seq id NO: 101, or a pharmaceutically acceptable salt thereof.

In further embodiments, the first VH comprises SEQ ID NO:7 and the first VL comprises seq id NO: 82, and/or the second VH comprises the amino acid sequence of SEQ ID NO: 17 and the second VL comprises the amino acid sequence of seq id NO: 18.

In further embodiments, the first VH comprises SEQ ID NO:7 and the first VL comprises seq id NO: 82, and/or the second VH comprises the amino acid sequence of SEQ ID NO: 96 and the second VL comprises seq id NO: 101, or a pharmaceutically acceptable salt thereof.

In another embodiment, the first VH comprises SEQ ID NO:7 and the first VL comprises seq id NO: 83, and/or the second VH comprises the amino acid sequence of SEQ ID NO: 17 and the second VL comprises the amino acid sequence of seq id NO: 18.

In another embodiment, the first VH comprises SEQ ID NO:7 and the first VL comprises seq id NO: 83, and/or the second VH comprises the amino acid sequence of SEQ ID NO: 96 and the second VL comprises seq id NO: 101, or a pharmaceutically acceptable salt thereof.

In yet another embodiment, the first VH comprises SEQ ID NO:7 and the first VL comprises seq id NO: 84, and/or the second VH comprises the amino acid sequence of SEQ ID NO: 17 and the second VL comprises the amino acid sequence of seq id NO: 18.

In yet another embodiment, the first VH comprises SEQ ID NO:7 and the first VL comprises seq id NO: 84, and/or the second VH comprises the amino acid sequence of SEQ ID NO: 96 and the second VL comprises seq id NO: 101, or a pharmaceutically acceptable salt thereof.

In another embodiment, the first heavy chain comprises SEQ ID NO: 9 and the first light chain comprises the amino acid sequence of SEQ ID NO: 10, and/or the second heavy chain comprises the amino acid sequence of SEQ ID NO: 19 and the second light chain comprises SEQ ID NO: 20.

In yet another embodiment, the first VH comprises SEQ ID NO:7 and the first VL comprises seq id NO: 84, and/or the second VH comprises the amino acid sequence of SEQ ID NO: 96 and the second VL comprises seq id NO: 101, or a pharmaceutically acceptable salt thereof.

In another embodiment, the first heavy chain comprises SEQ ID NO: 9 and the first light chain comprises the amino acid sequence of SEQ ID NO: 40, and/or the second heavy chain comprises the amino acid sequence of SEQ ID NO: 19 and the second light chain comprises SEQ ID NO: 20.

In another embodiment, the first heavy chain comprises SEQ ID NO: 9 and the first light chain comprises the amino acid sequence of SEQ ID NO: 40, and/or the second heavy chain comprises the amino acid sequence of SEQ ID NO: 97 and the second light chain comprises SEQ ID NO: 102, or a pharmaceutically acceptable salt thereof.

In another embodiment, the first heavy chain comprises SEQ ID NO: 9 and the first light chain comprises seq id NO: 78, and/or the second heavy chain comprises the amino acid sequence of SEQ ID NO: 19 and the second light chain comprises SEQ ID NO: 20.

In another embodiment, the first heavy chain comprises SEQ ID NO: 9 and the first light chain comprises the amino acid sequence of SEQ ID NO: 78, and/or the second heavy chain comprises the amino acid sequence of SEQ ID NO: 97 and the second light chain comprises SEQ ID NO: 102, or a pharmaceutically acceptable salt thereof.

In yet another embodiment, the first heavy chain comprises SEQ ID NO: 9 and the first light chain comprises the amino acid sequence of SEQ ID NO: 79, and/or the second heavy chain comprises the amino acid sequence of SEQ ID NO: 19 and the second light chain comprises SEQ ID NO: 20.

In yet another embodiment, the first heavy chain comprises SEQ ID NO: 9 and the first light chain comprises the amino acid sequence of SEQ ID NO: 79, and/or the second heavy chain comprises the amino acid sequence of SEQ ID NO: 97 and the second light chain comprises SEQ ID NO: 102, or a pharmaceutically acceptable salt thereof.

In further embodiments, the first heavy chain comprises SEQ ID NO: 9 and the first light chain comprises the amino acid sequence of SEQ ID NO: 80, and/or the second heavy chain comprises the amino acid sequence of SEQ ID NO: 19 and the second light chain comprises SEQ ID NO: 20.

In further embodiments, the first heavy chain comprises SEQ ID NO: 9 and the first light chain comprises the amino acid sequence of SEQ ID NO: 80, and/or the second heavy chain comprises the amino acid sequence of SEQ ID NO: 97 and the second light chain comprises SEQ ID NO: 102, or a pharmaceutically acceptable salt thereof.

In certain embodiments of all of the above aspects and embodiments, the antibody further comprises a cytotoxic agent, a radioisotope, a therapeutic agent, an antiviral agent, or a detectable label.

In a further aspect, the present disclosure provides a composition of nucleic acid molecules comprising a first light chain variable region or a first light chain encoding a first antigen binding domain of an antibody disclosed above. In another aspect, the present disclosure provides compositions comprising a nucleic acid molecule encoding a first heavy chain variable region or a first heavy chain of a first antigen binding domain of an antibody disclosed above. In yet another aspect, the present disclosure provides compositions comprising a nucleic acid molecule encoding the first light chain variable region or the first light chain of the second antigen-binding domain of the antibody disclosed above. In yet another aspect, the present disclosure provides compositions comprising a nucleic acid molecule encoding a second heavy chain variable region or a second heavy chain of a second antigen-binding domain of an antibody disclosed above.

In another aspect, the disclosure features a host cell comprising one or more of the foregoing nucleic acids. In some cases, the host cell comprises all four chains of the bispecific antibody. In other cases, the host cell comprises a nucleic acid encoding the gp 120-binding arm of the bispecific antibody. In other cases, the host cell comprises a nucleic acid encoding the CD3 binding arm of the bispecific antibody. In still other cases, the host cell comprises a nucleic acid encoding the CD89 binding arm of the bispecific antibody. In some embodiments, the host cell is selected from the group consisting of E.coli, Pseudomonas, Bacillus, Streptomyces, yeast (e.g., Pichia, Saccharomyces), CHO, YB/20, NS0, PER-C6, HEK-293T, NIH-3T3, HeLa, BHK, Hep G2, SP2/0, R1.1, B-W, L-M, COS 1, COS 7, BSC1, BSC40, BMT10 cells, plant cells, insect cells, and human cells in tissue culture.

In yet another aspect, features are methods of generating antibodies that bind gp120 and human CD3 (or antibodies that bind gp120 and human CD 89). The method comprises culturing the above-described host cell under conditions such that the nucleic acid molecule is expressed and the antibody is produced.

In another aspect, a method of detecting cells expressing gp120 and CD3 (or CD89) in a sample is disclosed. The method comprises contacting the sample with an antibody described herein.

In yet another aspect, the present disclosure provides a pharmaceutical composition comprising an antibody described herein and a pharmaceutically acceptable excipient.

In further embodiments, the disclosure features kits comprising an antibody described herein and a) a detection reagent, b) a gp120 and/or CD3 and/or CD89 antigen, c) a notice reflective of approved use or sale for human administration, or d) a combination thereof.

Also provided are methods of treating or preventing human immunodeficiency virus infection in a human subject in need thereof. The method comprises administering to a human subject a therapeutically effective amount of an antibody or pharmaceutical composition disclosed herein. In some embodiments, the human immunodeficiency virus infection is an HIV-1 infection. In some embodiments, the virus in the patient has Env positive for N332 PNG. In certain embodiments, HIV is of clade B, G, A, AC or AE.

In another aspect, the disclosure features an antibody that binds gp 120. The antibody comprises a VH and a VL. The VH comprises a VH comprising SEQ ID NO:1, VH-CDR1 comprising the amino acid sequence of SEQ ID NO: 2, VH-CDR2 comprising the amino acid sequence of SEQ ID NO:3, VH-CDR3 of the amino acid sequence of seq id no. VL comprises a vh comprising SEQ ID NO:4, VL-CDR1 comprising the amino acid sequence of SEQ ID NO:5 and a VL-CDR2 comprising the amino acid sequence of SEQ ID NO:6, VL-CDR3 of the amino acid sequence of seq id no. In addition, the VL comprises a tyrosine, phenylalanine or threonine at position 67A (Kabat numbering), or a glycine at position 67 (Kabat numbering).

In certain embodiments, the anti-gp 120 antibody binds to a polypeptide comprising or consisting of SEQ ID NO: 21, and a protein consisting of the amino acid sequence shown in the figure. In some cases, the anti-gp 120 antibody binds to a polypeptide comprising or consisting of SEQ ID NO: 38. In some cases, the anti-gp 120 antibody binds to free HIV-1 virus. In some cases, anti-gp 120 antibodies bind to HIV-1 infected cells. In some cases, anti-gp 120 antibodies bind to free HIV-1 virus and HIV-1 infected cells. In certain instances, the anti-gp 120 antibody binds to at least two different HIV-1 strains (e.g., group M, group N, group O, or group P). In one embodiment, the anti-gp 120 antibody binds pwito.c/2474 (accession No. JN944948 and NIH aid sreagent Program catalog No. 11739). In another embodiment, the anti-gp 120 antibody binds to pCH058.c/2960 (accession No. JN944940 and NIH AIDS Reagent Program catalog No. 700010058).

In some embodiments, the VH comprises a sequence identical to SEQ ID NO:7 at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identical. In some embodiments, the VH is linked, directly or by insertion of an amino acid sequence (e.g., a G-S linker), to a human IgG1 constant region (e.g., an IgG1m3 allotype) comprising 0-10 (e.g., 1,2, 3, 4, 5, 6,7, 8,9, or 10) amino acid substitutions that reduce effector function and/or increase the pharmacokinetic half-life of the antibody. In some cases, the antibody has a hinge region from an IgG3 antibody (e.g., the "open" IgG 3C-hinge variant disclosed in WO 2017/096221) and CH1, CH2, and CH3 regions from a human IgG1 antibody (e.g., an IgG1m3 allotype).

In certain embodiments, the VL comprises a sequence identical to SEQ ID NO: 81. 82, 83 or 84, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99% or 100%.

In some embodiments, the heavy chain comprises a heavy chain sequence identical to SEQ ID NO: 9, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identical.

In certain embodiments, the light chain comprises a heavy chain variable region identical to SEQ ID NO: 40. 78, 79 or 80, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99% or 100% identical.

In one embodiment, the antibody comprises a heavy chain comprising SEQ ID NO: 9, and a light chain comprising the amino acid sequence set forth in SEQ ID NO: 40 of any one of the amino acid sequences set forth in seq id no.

In another embodiment, the antibody comprises a heavy chain variable region comprising SEQ ID NO: 9, and a light chain comprising the amino acid sequence set forth in SEQ ID NO: 78 to any one of the amino acid sequences set forth herein.

In yet another embodiment, the antibody comprises a heavy chain comprising SEQ ID NO: 9, and a light chain comprising the amino acid sequence set forth in SEQ ID NO: 79 of any one of the amino acid sequences set forth herein.

In another embodiment, the antibody comprises a heavy chain variable region comprising SEQ ID NO: 9, and a light chain comprising the amino acid sequence set forth in SEQ ID NO: 80, or a light chain of an amino acid sequence as set forth in any one of seq id nos.

In some embodiments, the above-disclosed antibodies further comprise a cytotoxic agent, a radioisotope, a therapeutic agent, an antiviral agent, or a detectable label.

In another aspect, the disclosure features a pharmaceutical composition comprising the antibody of this aspect and a pharmaceutically acceptable carrier.

In another aspect, the disclosure relates to one or more nucleic acids encoding an antibody of this aspect.

In another aspect, the present disclosure provides one or more vectors comprising one or more of the nucleic acids described above.

In yet another aspect, the disclosure features a host cell comprising one or more of the vectors described above.

In a further aspect, the disclosure provides methods for producing an anti-gp 120 antibody. The method comprises culturing the above-described host cell under conditions in which one or more nucleic acids are expressed and antibodies are produced.

Also featured are methods of treating or preventing human immunodeficiency virus infection in a human subject in need thereof. The method comprises administering to a human subject a therapeutically effective amount of an antibody or pharmaceutical composition of this aspect. In some embodiments, the human immunodeficiency virus infection is an HIV-1 infection. In some embodiments, HIV in the patient has Env positive for N332 PNG. In certain embodiments, HIV is of clade B, G, A, AC or AE.

In another aspect, the disclosure features an antibody fragment that binds gp 120. The antibody fragment comprises VH and VL. The VH comprises a VH comprising SEQ ID NO:1, VH-CDR1 comprising the amino acid sequence of SEQ ID NO: 2, VH-CDR2 comprising the amino acid sequence of SEQ ID NO:3, VH-CDR3 of the amino acid sequence of seq id no. VL comprises a vh comprising SEQ ID NO:4, VL-CDR1 comprising the amino acid sequence of SEQ ID NO:5 and a VL-CDR2 comprising the amino acid sequence of SEQ ID NO:6, VL-CDR3 of the amino acid sequence of seq id no. In addition, the VL comprises a tyrosine, phenylalanine or threonine at position 67A (Kabat numbering), or a glycine at position 67 (Kabat numbering).

In certain embodiments, the anti-gp 120 antibody fragment binds to a polypeptide comprising or consisting of SEQ ID NO: 21, and a protein consisting of the amino acid sequence shown in the figure 21. In some cases, the anti-gp 120 antibody fragment binds to a polypeptide comprising or consisting of SEQ ID NO: 38. In some cases, the anti-gp 120 antibody fragment binds to free HIV-1 virus. In some cases, anti-gp 120 antibodies bind to HIV-1 infected cells. In some cases, the anti-gp 120 antibody fragment binds to free HIV-1 virus and HIV-1 infected cells. In some cases, the anti-gp 120 antibody will bind to at least two different HIV-1 strains (e.g., group M, group N, group O, or group P). In one embodiment, the anti-gp 120 antibody fragment binds pwito.c/2474 (accession No. JN944948 and NIH AIDS Reagent Program catalog No. 11739). In another embodiment, the anti-gp 120 antibody fragment binds to pCH058.c/2960 (accession No. JN944940 and NIH AIDS Reagent Program catalog No. 700010058).

In some embodiments, the VH comprises a sequence identical to SEQ ID NO:7 at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identical.

In certain embodiments, the VL comprises a sequence identical to SEQ ID NO: 81. 82, 83 or 84, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99% or 100%.

In some embodiments, the antibody fragment is a Fab, F (ab)2, Fv, scFv, sc (Fv)2, or diabody.

In some embodiments, the above-described antibody fragment further comprises a cytotoxic agent, a radioisotope, a therapeutic agent, an antiviral agent, or a detectable label.

In another aspect, the disclosure features a pharmaceutical composition comprising the antibody fragment of this aspect and a pharmaceutically acceptable carrier.

In another aspect, the disclosure relates to one or more nucleic acids encoding the antibody fragments of this aspect.

In another aspect, the present disclosure provides one or more vectors comprising one or more of the nucleic acids described above.

In yet another aspect, the disclosure features a host cell comprising one or more of the vectors described above.

In a further aspect, the disclosure provides methods of producing anti-gp 120 antibody fragments. The method comprises culturing the above-described host cell under conditions in which one or more nucleic acids are expressed and antibody fragments are produced.

In another aspect, the disclosure features a method of treating or preventing HIV in a human subject in need thereof. The method comprises administering to a human subject a therapeutically effective amount of the antibody fragment or pharmaceutical composition of this aspect. In some embodiments, the human immunodeficiency virus infection is an HIV-1 infection. In some embodiments, HIV in the patient has Env positive for N332 PNG. In certain embodiments, HIV is of clade B, G, A, AC or AE.

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

Other features and advantages of the invention will be apparent from the following detailed description, and from the claims.

Detailed Description

The multispecific antibodies described herein bind to the Human Immunodeficiency Virus (HIV) envelope (Env) protein gp120(gp120) and cluster of differentiation 3(CD3) (e.g., CD3 epsilon) and effectively kill HIV-infected cells. For example, a multispecific antibody is a bispecific antibody having two antigen-binding arms, wherein the bispecific antibody binds an antigen on an HIV-infected cell (e.g., gp120) with one arm and an antigen on a T cell (e.g., CD3) with the other arm, a T cell (e.g., CD 4) can be conjugated to a bispecific antibody+T cells and/or CD8+T cells) are targeted to HIV-infected cells, resulting in killing of HIV-infected cells. Bispecific antibodies that bind CD3 and gp120 can redirect CD8+And CD4+T cells to kill gp120 expressing cells (e.g., HIV infected cells). The T cells kill HIV infected cells regardless of their T cell receptor specificity. In one embodiment, the bispecific antibody is anti-gp 120X anti-CD 3

Figure BDA0002328692870000251

The duobodies of the present disclosure have significant advantages over other bispecific platforms known in the artAnd (4) the advantages are achieved.

Figure BDA0002328692870000252

A significant advantage of the platform over, for example, DART platforms is that the duobodies disclosed herein can recruit CD4+T cells to kill target cells (e.g., HIV-infected cells). No CD4 observed with DART+T cell mediated killing (see Sloan et al, PLOS Patholoens, 11 (11): e1005233. doi: 10.1371/journal. ppat.1005233 (2015)). This is a significant advantage, since the target in HIV therapy is also CD4+T cells. One problem with using antibodies that require innate effector cells for activity in HIV therapy is that the effector cells may not be present in latently infected CD4+Tissue in which T cells reside, and if CD4+T cells themselves may be effector cells, which is not a problem.

HIV-1 is the major family of HIV and accounts for 95% of all infections worldwide. HIV-2 is found primarily in some Western Africa countries.

The HIV virus is divided into specific groups, M, N, O and P, where M is the "main" group and contributes to the majority of HIV/AIDS worldwide. The M groups are further subdivided into subtypes (also called clades) that are prevalent in different geographical locations, according to their genetic sequence.

The M group "subtype" or "clade" is the HIV-1 group M subtype defined by the genetic sequence data. Examples of group M subtypes include subtypes A-K. Certain subtypes are known to be more virulent or resistant to different drug treatments. There are also "circulating recombinant forms" or CRFs derived from recombination between different subtypes of viruses, each assigned a number. For example, CRF12_ BF is a recombination between subtypes B and F. Subtype a is not very common in the west. Subtype B is the predominant form in Europe, America, Japan, Thailand and Australia. Subtype C is the predominant form in southern Africa, eastern Africa, India, Nepal and parts of China. Subtype D is usually only seen in eastern and central africa. Subtype E was never identified as non-recombinant, it only recombined with subtype a as CRF01_ AE. Subtype F is found in the middle of Africa, south America and eastern Europe. Subtype G (and CRF02_ AG) is found in Africa and Central Europe. Subtype H is restricted to central Africa. Subtype I was originally used to describe the strain now designated CRF04_ cpx, of which cpx was used for "recombination" of several subtypes. Subtype J is mainly seen in north africa, central africa and west africa, and the caribbean subtype K is limited to the democratic republic of congo and karilong. These subtypes are sometimes further divided into sub-subtypes, such as a1 and a2 or F1 and F2. In 2015, CRF19 strain, a recombinant of subtype a, subtype D and subtype G and subtype D proteases, was found in cuba, closely associated with the rapid development of aids.

The present disclosure specifically provides neutralizing antibodies (e.g., broadly neutralizing abs) that target gp120 polypeptides on the surface of HIV-infected cells. Neutralizing antibodies against viral envelope proteins can provide an adaptive immune defense against HIV-1 exposure by blocking infection of susceptible cells. Extensive neutralization indicated that the antibody could neutralize HIV-1 isolates from different clades. Thus, antibodies encompassed by the present disclosure have cross-clade binding activity.

gp120

The envelope glycoprotein gp120 (or gp120) is a 120kDa glycoprotein that is part of the outer layer of HIV. It exists as a viral membrane spike (viral membrane spike) itself, consisting of three gp120 molecules linked together and anchored to the membrane by the gp41 protein. gp120 is important for viral infection because it facilitates entry of HIV into host cells through interaction with cell surface receptors. These receptors include the DC-SIGN, heparan sulfate proteoglycans and CD4 receptors. Binding to CD4 on helper T cells induces the initiation of a cascade of conformational changes in gp120 and gp41, which results in fusion of the virus to the host cell membrane.

gp120 is encoded by the HIV env gene. The env gene codes for a gene product of about 850 amino acids. The major env product is the protein gp160, which is cleaved in the endoplasmic reticulum by the cellular proteases furin to gp120 (about 480 amino acids) and gp41 (about 345 amino acids).

The amino acid sequence of an exemplary gp160 polypeptide of HIV clone WITO is provided below (V3 hypervariable loop bold, and N332 potential N-linked glycosylation sites bold and underlined):

the amino acid sequence of an exemplary gp120 polypeptide is provided below:

Figure BDA0002328692870000272

the amino acid sequence of another exemplary gp120 polypeptide is provided below (see www.bioafrica.net/proteomics/ENV-gp120prot. html):

Figure BDA0002328692870000282

genomic diversity between independent human immunodeficiency virus type 1(HIV-1) isolates, in successive isolates from the same patient to a lesser extent, and even within single patient isolates, is a well-known feature of HIV-1. Although this sequence heterogeneity is distributed throughout the genome, most of the heterogeneity is located in the env gene. Comparison of the predicted amino acid sequences from several different isolates revealed that sequence heterogeneity was clustered in five variable regions of the surface glycoprotein gp120 (designated V1 to V5). The V3 region, although only 35 amino acids long, exhibits considerable sequence variability. Interestingly, despite this variability, the V3 region includes mediation and CD4+Determinants of cellular interactions. The increase in gp120 variability results in higher levels of viral replication, indicating increased viral fitness in individuals infected with diverse HIV-1 variants. Variability in potential N-linked glycosylation sites (PNGS) also leads to increased viral adaptability. PNGS allows long-chain carbohydrates to bind to the hypervariable region of gp 120. Thus, the number of PNGS in env may influence the virus' adaptability by providing more or less sensitivity to neutralizing antibodies.

The consensus sequence for the gp 120V 3 region is provided below (Milich et al, J Virol.,67(9): 5623-5631 (1993)):

CTRPNNNTRKSIHIGPGRAFYTTGEIIGDIRQAHC(SEQ ID NO:22)。

anti-gp 120 antibodies

The disclosure features anti-gp 120 antibodies. In certain embodiments, these antibodies bind to HIV-1 antigen expressed on the cell surface and eliminate or kill infected cells.

In certain embodiments, these antibodies are neutralizing antibodies (e.g., monoclonal) targeting HIV-1. A "neutralizing antibody" is an antibody that can neutralize the ability of HIV to initiate and/or perpetuate an infection in a host and/or in a target cell in vitro. The present disclosure provides neutralizing monoclonal human antibodies, wherein the antibodies recognize an antigen from HIV, such as a gp120 polypeptide. In certain embodiments, a "neutralizing antibody" can inhibit entry of an HIV-1 virus (e.g., SF162 and/or JR-CSF) at a neutralization index of >1.5 or >2.0 (Kostrikikis LG et al J.Virol.,70(1): 445-458 (1996)).

In some embodiments, these antibodies are broadly neutralizing antibodies (e.g., monoclonal) targeting HIV-1. "broadly neutralizing antibody" refers to an antibody that neutralizes more than one HIV-1 virus species (from a diverse clade and different strains within a clade) in a neutralization assay. Broadly neutralizing antibodies can neutralize at least 2, 3, 4, 5, 6,7, 8,9 or more different HIV-1 strains belonging to the same or different clades. In particular embodiments, the broadly neutralizing antibody can neutralize multiple HIV-1 species belonging to at least 2, 3, 4, 5, or 6 different clades. In certain embodiments, the inhibitory concentration of the antibody can be less than about 0.0001. mu.g/ml, less than about 0.001. mu.g/ml, less than about 0.01. mu.g/ml, less than about 0.1. mu.g/ml, less than about 0.5. mu.g/ml, less than about 1.0. mu.g/ml, less than about 5. mu.g/ml, less than about 10. mu.g/ml, less than about 25. mu.g/ml, less than about 50. mu.g/ml or less than about 100. mu.g/ml to neutralize about 50% of the input virus in a neutralization assay.

In one embodiment, the anti-gp 120 antibodies of the present disclosure are related to the antibody described as PGT-121LO6 in PCT application publication No. WO 2012/030904. Table 1 below provides the relevant sequence information for the PGT-121LO6 antibody.

TABLE 1

Figure BDA0002328692870000301

Figure BDA0002328692870000311

Crystal structure and experimental analysis of an antibody highly related to the PGT-121LO6 antibody (i.e., PGT-122) showed that PGT122 also utilized amino acid residues other than the CDRs to bind antigen (along with the CDRs). For example, the antibody appears to have additional regions in the framework regions that contact the antigen (see, e.g., PLOS Pathogens,9, e1003754(2013), such as Experimental differentiation for PGT121 and primed antibodies: Sok). The high resolution structure of PGT122 binding to Env viral antigens has been determined (see, e.g., Julien JP et al, Science, 342, 14777-. The structure of PGT121 is described in Julie JP et al, PLOS Pathologens 9, E1003342(2013) and Mouquet H et al, PNAS, 109, E3268-E3277 (2012). The structure of the PGT122 is described in Julie JP et al, PLOS Pathologens 9, e1003342(2013), PDB ID 4JY 5; and the structure of PGT123 is described in Julie JP et al, PLOS Pathologens 9, e1003342 (2013). PGT122 and PGT123 antibodies are closely related to PGT121 antibodies, so PGT122/Env structure and knowledge of PGT121, PGT122 and PGT123 structure can be used to model the structure of PGT121 bound to Env very accurately and predict with high confidence the residues of PGT121 involved in binding to Env. Residues of the PGT121 LO6 antibody contacting gp120 antigen predicted based on similarity to PGT122 and PGT122/Env structures are provided below, with framework residues shown in bold (Kabat numbering):

VH (Kabat numbering):

33. 56, 58, 99, 100A, 100B, 100C, 100D, 100E, 100G, 100I, 100J, 100K, 100L; and

VL (Kabat numbering):

28、29、30、50、51、52、66、67、67A、67C、91、92、93、 94、95、95A、95B。

the PGT-121LO6 antibody has been shown to bind to a variety of different antigen variants (e.g., different virus strains) that can contact the antibody at unknown amino acid positions other than those listed above. Different virus strains have different Env (i.e. antigen) sequences and different glycosylation patterns, and even a single Env sequence may have heterogeneous glycosylation patterns, requiring extensive binding or neutralizing antibodies to recognize different Env proteins of different HIV-1 variants or even different glycosylation patterns on the same Env protein. For example, the epitope for PGT121 consists of the Env V3 loop, in particular the N-linked glycan at position N332. The V3 loop is the primary determinant for cell tropism and virus clade. Among the 117 CCR5 tropic viruses of various clades, the presence of a potential N-linked glycosylation (PNG) motif in the viral DNA sequence encoding N332 glycan was significantly correlated with susceptibility to PGT121 neutralization in viruses of clade B, G, A, AC and AE. Of the 50 clade B Env sequences isolated from patients enrolled in a girlidard (Gilead) -sponsored clinical trial, 94% of the CCR5 tropism Env with the N332 PNG motif were readily neutralized by PGT121, compared to only 26% of the non-CCR 5 tropism, N332 PNG positive virus predispositions (P < 0.0001). Thus, genetic determination of Env clade, tropism and presence of the N332 PNG motif are highly predictive of susceptibility to neutralization by PGT121 and can be used as markers to predict viral susceptibility to neutralization by PGT121 and its derivatives.

The present disclosure provides variants of the PGT-121LO6 antibody. In certain embodiments, these variants have substantially the same or increased binding affinity for gp120 as compared to the PGT-121LO6 antibody. Binding affinity can be determined using any assay known in the art, including ELISA, SPR, BLI, or flow cytometry. In certain embodiments, these variants have increased binding affinity for FcRn at pH 6.0 as compared to the PGT-121LO6 antibody. In some embodiments, these variants have increased HIV-1 neutralization relative to the PGT-121LO6 antibody. In certain embodiments, the variant has reduced immunogenicity as compared to the PGT-121LO6 antibody. In certain embodiments, the Env region in or around the following residues (HIV Env HXB2 numbering) is implicated in predicting binding of a variant of the disclosure to an Env protein: the V3 loop (324-328, 330) and the related N332 glycan as well as a portion of the V1-loop (135-137) and the related N137 glycan, residue 415-417. Paratope prediction for Env binding involves residues in direct contact with antigen in the following regions (Kabat numbering): CDRH1(33), CDRH2(50, 56, 58), CDRH3(99, 100A, 100B, 100C, 100D, 100E, 100G, 100I, 100L), CDRL1(28, 29, 30), CDRL2(50, 51, 52), LFR3(66, 67A, 67B, and 67C), and CDRL3(93, 94, 95A, 95B).

PGT-121LO6 heavy chain variable domain sequence (with Kabat numbering) (SEQ ID NO: 126)

Figure BDA0002328692870000331

PGT-121LO6 light chain variable domain sequence (with Kabat numbering; note VL terminates at position 107 (V); i.e., G108 is not part of VL) (SEQ ID NO:127)

Figure BDA0002328692870000341

An exemplary variant of the PGT-121LO6 antibody is the PGT-121.60 antibody, the relevant sequence information of which is provided in table 2 below.

TABLE 2

Figure BDA0002328692870000351

Exemplary anti-gp 120 antibody 1

Exemplary anti-gp 120 antibody 1 is related to the PGT-121.60 antibody. Table 3 provides the relevant amino acid sequences of an exemplary anti-gp 120 antibody 1(PGT121.60 human IgG1 FEARLS/human λ).

TABLE 3

Figure BDA0002328692870000352

Figure BDA0002328692870000361

Figure BDA0002328692870000371

The anti-gp 120 antibody can comprise the heavy chain CDR1, CDR2, and CDR3 and the light chain CDR1, CDR2, and CDR3 of exemplary anti-gp 120 antibody 1. In one embodiment, the CDRs are defined based on the Kabat definition. In another embodiment, the CDRs are defined based on the Chothia definition. In particular embodiments, Chothia is defined from Discovery Studio using Chothia and Lesk, J Mol biol.196 (4): 901-17(1987) and Morea et al, Methods, 20: 267-279 (2000). In another specific embodiment, the Chothia definition is based on Chothia from the Abysis definition. In another embodiment, the CDRs are defined based on IMGT definitions. In another embodiment, the CDRs are defined based on the honeygger definition. In another embodiment, the CDRs are defined based on contact definitions. In some cases, an anti-gp 120 antibody of the present disclosure binds to a polypeptide comprising or consisting of SEQ ID NO: 21, and a protein consisting of the amino acid sequence shown in the figure 21. In some cases, an anti-gp 120 antibody of the present disclosure binds to a polypeptide comprising or consisting of SEQ ID NO: 38. In some cases, the anti-gp 120 antibodies of the present disclosure bind to free HIV-1 virus. In some cases, an anti-gp 120 antibody of the disclosure binds to an HIV-1 infected cell. In some cases, the anti-gp 120 antibodies of the present disclosure bind to free HIV-1 virus and HIV-1 infected cells. In some cases, an anti-gp 120 antibody of the present disclosure binds to at least two different HIV-1 strains (e.g., group M, group N, group O, or group P). In one embodiment, an anti-gp 120 antibody of the disclosure binds pwito.c/2474 (accession No. JN944948 and NIH AIDS Reagent Program catalog No. 11739). In another embodiment, an anti-gp 120 antibody of the disclosure binds to pCH058.c/2960 (accession No. JN944940 and NIH AIDS Reagent Program catalog No. 700010058).

In certain instances, the anti-gp 120 antibody comprises an amino acid sequence that is at least 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the variable heavy chain of exemplary anti-gp 120 antibody 1. In some embodiments, the anti-gp 120 antibody comprises an amino acid sequence that is at least 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the heavy chain of exemplary anti-gp 120 antibody 1. In certain instances, the anti-gp 120 antibody comprises an amino acid sequence that is at least 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the variable light chain of exemplary anti-gp 120 antibody 1. In certain instances, the anti-gp 120 antibody comprises an amino acid sequence that is at least 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the light chain of exemplary anti-gp 120 antibody 1. In certain embodiments, the anti-gp 120 antibody comprises an amino acid sequence that is at least 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the variable heavy and variable light chains of exemplary anti-gp 120 antibody 1. In some embodiments, the anti-gp 120 antibody comprises an amino acid sequence at least 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the heavy chain of exemplary anti-gp 120 antibody 1 and comprises an amino acid sequence at least 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to light chain 1 of the exemplary anti-gp 120 antibody. In some cases, an anti-gp 120 antibody of the present disclosure binds to a polypeptide comprising or consisting of SEQ ID NO: 21, and a protein consisting of the amino acid sequence shown in the figure. In some cases, an anti-gp 120 antibody of the disclosure binds to a polypeptide comprising SEQ ID NO: 38 or a protein consisting thereof. In some cases, the anti-gp 120 antibodies of the present disclosure bind to free HIV-1 virus. In some cases, an anti-gp 120 antibody of the disclosure binds to an HIV-1 infected cell. In some cases, the anti-gp 120 antibodies of the present disclosure bind to free HIV-1 virus and HIV-1 infected cells. In some cases, an anti-gp 120 antibody of the present disclosure binds to at least two different HIV-1 strains (e.g., group M, group N, group O, or group P). In one embodiment, an anti-gp 120 antibody of the disclosure binds pwito.c/2474 (accession No. JN944948 and NIH AIDS Reagent Program catalog No. 11739). In another embodiment, an anti-gp 120 antibody of the disclosure binds to pch058.c/2960 (accession No. JN944940 and NIH aid diagnostic Program catalog No. 700010058).

In some embodiments, the variable heavy chain of exemplary anti-gp 120 antibody 1 is linked to a heavy chain constant region comprising a CH1 domain and a hinge region. In some embodiments, the variable heavy chain of exemplary anti-gp 120 antibody 1 is linked to a heavy chain constant region comprising a CH3 domain. In certain embodiments, the variable heavy chain of exemplary anti-gp 120 antibody 1 is linked to a heavy chain constant region comprising a CH1 domain, a hinge region, and a CH2 domain from IgG4 and a CH3 domain (e.g., IgG1, for example). In certain embodiments, the variable heavy chain of exemplary anti-gp 120 antibody 1 is linked to a heavy chain constant region comprising a CH1 domain, a CH2 domain, and a CH3 domain from IgG1 (e.g., human IgG1, e.g., IgG1m3 allotype), and an IgG3 hinge region (e.g., the "open" IgG3 hinge variant named "IgG 3C" in WO2017/096221 (see, e.g., fig. 2A of the PCT publication)). This IgG3 hinge variant is expected to exhibit improved Fab arm flexibility and cross-200A0Distance (sufficient for trimeric internal interaction) capability. In certain embodiments, such chimeric antibodies comprise one or more additional mutations in the heavy chain constant region that increase the stability of the chimeric antibody. In certain embodiments, the heavy chain constant region comprises a substitution that alters a property of the antibody (e.g., reduces Fc receptor binding, increases or decreases antibody glycosylation, decreases binding to C1q, increases half-life, decreases effector function).

In certain embodiments, the anti-gp 120 antibody is an IgG antibody. In one embodiment, the antibody is IgG 1. In another embodiment, the antibody is IgG 2. In some embodiments, the antibody has a chimeric heavy chain constant region (e.g., having the CH1, hinge, and CH2 regions of IgG4 and the CH3 region of IgG 1).

IgG antibodies exist in various allotypes and allotypes (isoallotypes). In particular embodiments, the antibodies of the present disclosure comprise an IgG1 heavy chain having an allotype G1m1, nG1m2, G1m3, G1m17,1, G1m17,1,2, G1m3,1, or G1m 17. Each of these allotypes or heteroallotypes is characterized by the following amino acid residues at designated positions (EU numbering) in the heavy chain constant region (Fc) of IgG 1:

G1m1:D356,L358;

nG1m1:E356,M358;

G1m3:R214,E356,M358,A431;

G1m17,1:K214,D356,L358,A431;

G1m17,1,2:K214,D356,L358,G431;

g1m3,1: R214, D356, L358, A431; and

G1m17:K214,E356,M358,A431。

in particular embodiments, the VH of exemplary anti-gp 120 antibody 1 is linked, either directly or by insertion of an amino acid sequence (e.g., G-S linker), to the wild-type IgG1m3 sequence provided below (representative allotype-determining residues are shown in bold type)

Figure BDA0002328692870000401

In certain embodiments, the VH of exemplary anti-gp 120 antibody 1 (e.g., an amino acid sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO:7 or having 0 to 5 (i.e., 1,2, 3, 4, or 5) amino acid substitutions (e.g., conservative substitutions in SEQ ID NO: 7)) is identical to a VH having SEQ ID NO: the mutated IgG1m3 sequences of 1 to 10 (e.g., 1,2, 3, 4, 5, 6,7, 8,9, 10) amino acid substitutions in 77 (e.g., substitutions that reduce effector function and/or increase half-life) are linked directly or by intervening amino acid sequences (e.g., G-S linkers). Exemplary amino acid substitutions are described later in this disclosure.

In certain embodiments, the anti-gp 120 antibody is a human IgG 1/human kappa antibody. In some embodiments, an antibody of the present disclosure comprises a kappa light chain having an allotype selected from Km1, Km1,2, or Km 3. Each of these allotypes was characterized by the following amino acid residues at the designated positions (EU numbering) within the IgG1 light chain:

Km1:V153,L191;

km1,2: A153, L191; and

Km3:A153,V191。

in certain embodiments, the antibodies of the present disclosure comprise an IgG1 kappa light chain comprising one of the following amino acid sequences, wherein representative allotype determining residues are indicated in bold:

Km1:

Figure BDA0002328692870000411

Km1,2:

Figure BDA0002328692870000412

Figure BDA0002328692870000413

or

Km3:

Figure BDA0002328692870000414

In certain embodiments, the anti-gp 120 antibody is a human IgG 1/human λ antibody. Each human individual contains 7 to 11 different λ light chain genes encoding light chains selected from λ 1, λ 2, λ 3, λ 4, λ 5, λ 6 and λ 7. In particular embodiments, the antibodies of the present disclosure comprise a λ light chain selected from λ 1, λ 2, λ 3, λ 4, λ 5, λ 6, and λ 7. In particular embodiments, the antibodies described herein comprise a lambda light chain comprising one of the following amino acid sequences, wherein representative lambda determining residues are represented in bold:

λ1:

Figure BDA0002328692870000415

λ2:

Figure BDA0002328692870000416

λ3:

Figure BDA0002328692870000423

or

λ7:

Figure BDA0002328692870000424

In particular embodiments, the VL of exemplary anti-gp 120 antibody 1 (e.g., an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% identity to SEQ ID NO: 8, or having from 0 to 5 (i.e., 1,2, 3, 4, or 5) amino acid substitutions (e.g., conservative substitutions in SEQ ID NO: 8)) is linked directly to a wild-type human λ 2 sequence (SEQ ID NO: 89) or via an intervening amino acid sequence (e.g., a G-S linker). In certain embodiments, the VL of exemplary anti-gp 120 antibody 1 is identical to a VL having SEQ ID NO: 89 (i.e., 1,2, 3, 4, 5) substituted mutant human λ 2 sequences are linked directly or via an intervening amino acid sequence (e.g., a G-S linker).

In a particular embodiment, the anti-gp 120 antibody is a human IgG1m 3/human λ 2 antibody.

For example, an antibody such as exemplary anti-gp 120 antibody 1 can be prepared by preparing and expressing a nucleic acid encoding an antibody amino acid sequence.

Exemplary anti-gp 120 antibody 2

Another exemplary anti-gp 120 antibody, exemplary anti-gp 120 antibody 2, has the same six CDRs as exemplary anti-gp 120 antibody 1. The antibody comprises a heavy chain variable region comprising SEQ ID NO:7 or a VH sequence consisting of it, and a VL sequence comprising or consisting of the amino acid sequence set forth in seq id no:

Figure BDA0002328692870000425

Figure BDA0002328692870000431

in some cases, the VL is linked to the human λ constant region directly or through an intervening amino acid sequence (e.g., a G-S linker).

An exemplary anti-gp 120 antibody 2 comprises a heavy chain comprising SEQ ID NO: 9 and a light chain comprising or consisting of the amino acid sequence set forth in seq id no:

Figure BDA0002328692870000432

in some cases, an anti-gp 120 antibody of the present disclosure binds to a polypeptide comprising or consisting of SEQ ID NO: 21, and a protein consisting of the amino acid sequence shown in the figure 21. In some cases, an anti-gp 120 antibody of the present disclosure binds to a polypeptide comprising or consisting of SEQ ID NO: 38. In some cases, the anti-gp 120 antibodies of the present disclosure bind to free HIV-1 virus. In some cases, an anti-gp 120 antibody of the disclosure binds to an HIV-1 infected cell. In some cases, the anti-gp 120 antibodies of the present disclosure bind to free HIV-1 virus and HIV-1 infected cells. In some cases, an anti-gp 120 antibody of the present disclosure binds to at least two different HIV-1 strains (e.g., group M, group N, group O, or group P). In one embodiment, an anti-gp 120 antibody of the disclosure binds pwito.c/2474 (accession No. JN944948 and NIH AIDS Reagent Program catalog No. 11739). In another embodiment, an anti-gp 120 antibody of the disclosure binds to pCH058.c/2960 (accession No. JN944940 and NIH AIDS Reagent Program catalog No. 700010058).

In some embodiments, the variable heavy chain of exemplary anti-gp 120 antibody 2 (e.g., an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% identity to SEQ ID NO:7 or having 0 to 5 (i.e., 1,2, 3, 4, or 5) amino acid substitutions (e.g., conservative substitutions in SEQ ID NO: 7)) is linked to a heavy chain constant region comprising a CH1 domain and a hinge region. In some embodiments, the variable heavy chain of exemplary anti-gp 120 antibody 2 (e.g., an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% identity to SEQ ID NO:7 or having 0 to 5 (i.e., 1,2, 3, 4, or 5) amino acid substitutions (e.g., conservative substitutions in SEQ ID NO: 7)) is linked to a heavy chain constant region comprising a CH3 domain. In certain embodiments, the variable heavy chain of exemplary anti-gp 120 antibody 2 (e.g., an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% identity to SEQ ID NO:7 or having from 0 to 5 (i.e., 1,2, 3, 4, or 5) amino acid substitutions (e.g., a conservative substitution in SEQ ID NO: 7)) is linked to a heavy chain constant region comprising a CH1 domain, a hinge region, and a CH2 domain from IgG4 and a CH3 domain (e.g., from IgG 1). In certain embodiments, the variable heavy chain of exemplary anti-gp 120 antibody 1 is linked to a heavy chain constant region comprising a CH1 domain, a CH2 domain, and a CH3 domain from IgG1 (e.g., human IgG1, e.g., IgG1m3 allotype), and an IgG3 hinge region (e.g., "open" IgG3 hinge variant "IgG 3C-" described in WO 2017/096221). In certain embodiments, such chimeric antibodies comprise one or more additional mutations in the heavy chain constant region that increase the stability of the chimeric antibody. In certain embodiments, the heavy chain constant region comprises a substitution that alters a property of the antibody (e.g., reduces Fc receptor binding, increases or decreases antibody glycosylation, decreases binding to C1q, increases half-life, decreases effector function).

In certain embodiments, the anti-gp 120 antibody is an IgG antibody. In one embodiment, the antibody is IgG 1. In another embodiment, the antibody is IgG 2. In some embodiments, the antibody has a chimeric heavy chain constant region (e.g., having the CH1, hinge, and CH2 regions of IgG4 and the CH3 region of IgG 1).

In particular embodiments, the antibodies of the present disclosure comprise an IgG1 heavy chain having an allotype G1m1, nG1m2, G1m3, G1m17,1, G1m17,1,2, G1m3,1, or G1m 17.

In certain embodiments, the anti-gp 120 antibody is a human IgG 1/human kappa antibody. In some embodiments, an antibody of the present disclosure comprises a kappa light chain having an allotype selected from Km1, Km1,2, or Km 3. In certain embodiments, the anti-gp 120 antibody is a human IgG 1/human λ antibody. In some embodiments, an antibody of the present disclosure comprises a light chain selected from λ 1, λ 2, λ 3, λ 4, λ 5, λ 6, and λ 7.

In a particular embodiment, the anti-gp 120 antibody is a human IgG1m 3/human λ 2 antibody.

In some embodiments, the VH of exemplary anti-gp 120 antibody 2 (e.g., an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% identity to SEQ ID NO:7, or having from 0 to 5 (i.e., 1,2, 3, 4, or 5) amino acid substitutions (e.g., a conservative substitution within SEQ ID NO: 7)) is linked to a wild-type IgG1m3 sequence (SEQ ID NO: 77) either directly or through an intervening amino acid sequence (e.g., a G-S linker). In certain embodiments, the VH of exemplary anti-gp 120 antibody 2 (e.g., an amino acid sequence at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% identical to SEQ ID NO:7, or having from 0 to 5 (i.e., 1,2, 3, 4, or 5) amino acid substitutions (e.g., a conservative substitution in SEQ ID NO: 7)) is identical to a VH having SEQ ID NO: the mutated IgG1m3 sequences of 1 to 10 (e.g., 1,2, 3, 4, 5, 6,7, 8,9, 10) amino acid substitutions in 77 (e.g., substitutions that reduce effector function and/or increase half-life) are linked directly or by intervening amino acid sequences (e.g., G-S linkers). Exemplary amino acid substitutions are described later in this disclosure.

In some embodiments, the VL of exemplary anti-gp 120 antibody 2 (e.g., an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% identity to SEQ ID NO: 81, or having from 0 to 5 (i.e., 1,2, 3, 4, or 5) amino acid substitutions (e.g., a conservative substitution in SEQ ID NO: 81)) is linked directly to a wild-type human λ 2 sequence (SEQ ID NO: 89) or via an intervening amino acid sequence (e.g., a G-S linker). In certain embodiments, the VL of exemplary anti-gp 120 antibody 2 (e.g., an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% identity to SEQ ID NO: 81, or having from 0 to 5 (i.e., 1,2, 3, 4, or 5) amino acid substitutions (e.g., a conservative substitution in SEQ ID NO: 81)) is identical to a VL having SEQ ID NO: 89 (i.e., 1,2, 3, 4, 5) substituted mutant human λ 2 sequences are linked directly or via an intervening amino acid sequence (e.g., a G-S linker).

Exemplary anti-gp 120 antibody 2 can be used as a monospecific antibody or a multispecific antibody (e.g., bispecific antibody). The present disclosure encompasses whole antibodies or antigen-binding fragments (e.g., Fab, F (ab)2, Fv, scFv, sc (Fv)2, diabodies).

Antibodies such as exemplary anti-gp 120 antibody 2 can be prepared, for example, by making and expressing nucleic acids encoding the amino acid sequences of the antibodies.

Exemplary anti-gp 120 antibody 3

Another exemplary anti-gp 120 antibody, exemplary anti-gp 120 antibody 3, has the same six CDRs as exemplary anti-gp 120 antibody 1. The antibody comprises a heavy chain variable region comprising SEQ ID NO:7 or a VH sequence consisting of it, and a VL sequence comprising or consisting of the amino acid sequence set forth in seq id no:

Figure BDA0002328692870000461

in some cases, the VL is linked to the human λ constant region directly or through an intervening amino acid sequence (e.g., a G-S linker).

Exemplary anti-gp 120 antibody 3 comprises a heavy chain comprising SEQ ID NO: 9 and a light chain comprising or consisting of the amino acid sequence set forth in seq id no:

Figure BDA0002328692870000462

in some cases, an anti-gp 120 antibody of the present disclosure binds to a polypeptide comprising or consisting of SEQ ID NO: 21, and a protein consisting of the amino acid sequence shown in the figure 21. In some cases, an anti-gp 120 antibody of the present disclosure binds to a polypeptide comprising or consisting of SEQ ID NO: 38. In some cases, the anti-gp 120 antibodies of the present disclosure bind to free HIV-1 virus. In some cases, an anti-gp 120 antibody of the disclosure binds to an HIV-1 infected cell. In some cases, the anti-gp 120 antibodies of the present disclosure bind to free HIV-1 virus and HIV-1 infected cells. In some cases, an anti-gp 120 antibody of the present disclosure binds to at least two different HIV-1 strains (e.g., group M, group N, group O, or group P). In one embodiment, an anti-gp 120 antibody of the disclosure binds pwito.c/2474 (accession No. JN944948 and NIH AIDS Reagent Program catalog No. 11739). In another embodiment, an anti-gp 120 antibody of the disclosure binds to pCH058.c/2960 (accession No. JN944940 and NIH AIDS Reagent Program catalog No. 700010058).

In some embodiments, the variable heavy chain of exemplary anti-gp 120 antibody 3 is linked to a heavy chain constant region comprising a CH1 domain and a hinge region. In some embodiments, the variable heavy chain of exemplary anti-gp 120 antibody 3 is linked to a heavy chain constant region comprising a CH3 domain. In certain embodiments, the variable heavy chain of exemplary anti-gp 120 antibody 3 is linked to a heavy chain constant region comprising a CH1 domain from IgG4, a hinge region, and a CH2 domain and a CH3 domain (e.g., from IgG 1). In certain embodiments, the variable heavy chain of exemplary anti-gp 120 antibody 1 is linked to a heavy chain constant region comprising a CH1 domain, a CH2 domain, and a CH3 domain from IgG1 (e.g., human IgG1, e.g., IgG1m3 allotype), and an IgG3 hinge region (e.g., "open" IgG3 hinge variant "IgG 3C-" described in WO 2017/096221). In certain embodiments, such chimeric antibodies comprise one or more additional mutations in the heavy chain constant region that increase the stability of the chimeric antibody. In certain embodiments, the heavy chain constant region comprises a substitution that alters a property of the antibody (e.g., reduces Fc receptor binding, increases or decreases antibody glycosylation, decreases binding to C1q, increases half-life, decreases effector function).

In certain embodiments, the anti-gp 120 antibody is an IgG antibody. In one embodiment, the antibody is IgG 1. In another embodiment, the antibody is IgG 2. In some embodiments, the antibody has a chimeric heavy chain constant region (e.g., having the CH1, hinge, and CH2 regions of IgG4 and the CH3 region of IgG 1).

In particular embodiments, the antibodies of the present disclosure comprise an IgG1 heavy chain having an allotype G1m1, nG1m2, G1m3, G1m17,1, G1m17,1,2, G1m3,1, or G1m 17.

In certain embodiments, the anti-gp 120 antibody is a human IgG 1/human kappa antibody. In some embodiments, an antibody of the present disclosure comprises a kappa light chain having an allotype selected from Km1, Km1,2, or Km 3. In certain embodiments, the anti-gp 120 antibody is a human IgG 1/human λ antibody. In some embodiments, an antibody of the present disclosure comprises a light chain selected from λ 1, λ 2, λ 3, λ 4, λ 5, λ 6, and λ 7.

In a particular embodiment, the anti-gp 120 antibody is a human IgG1m 3/human λ 2 antibody.

In some embodiments, the VH of exemplary anti-gp 120 antibody 3 (e.g., an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% identity to SEQ ID NO:7, or having from 0 to 5 (i.e., 1,2, 3, 4, or 5) amino acid substitutions (e.g., a conservative substitution within SEQ ID NO: 7)) is linked directly to a wild-type IgG1m3 sequence (SEQ ID NO: 77) or via an intervening amino acid sequence (e.g., a G-S linker). In certain embodiments, the VH (e.g., at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% identity to SEQ ID NO:7, or having 0 to 5 (i.e., 1,2, 3, 4, or 5) amino acid substitutions (e.g., conservative substitutions in SEQ ID NO: 7) amino acid sequence) of exemplary anti-gp 120 antibody 3 is identical to a VH having SEQ ID NO: the mutated IgG1m3 sequences of 1 to 10 (e.g., 1,2, 3, 4, 5, 6,7, 8,9, 10) amino acid substitutions in 77 (e.g., to reduce effector function and/or extend half-life) are linked directly or by intervening amino acid sequences (e.g., G-S linkers). Exemplary amino acid substitutions are described later in this disclosure.

In some embodiments, the VL of exemplary anti-gp 120 antibody 3 (e.g., an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% identity to SEQ ID NO: 82, or having from 0 to 5 (i.e., 1,2, 3, 4, or 5) amino acid substitutions (e.g., a conservative substitution in SEQ ID NO: 82)) is linked directly to a wild-type human λ 2 sequence (SEQ ID NO: 89) or via an intervening amino acid sequence (e.g., a G-S linker). In certain embodiments, the VL of exemplary anti-gp 120 antibody 3 (e.g., an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% identity to SEQ ID NO: 82, or having from 0 to 5 (i.e., 1,2, 3, 4, or 5) amino acid substitutions (e.g., a conservative substitution in SEQ ID NO: 82)) is identical to a VL having SEQ ID NO: 89 (i.e., 1,2, 3, 4, 5) substituted mutant human λ 2 sequences are linked directly or via an intervening amino acid sequence (e.g., a G-S linker).

Exemplary anti-gp 120 antibody 3 can be used as a monospecific antibody or a multispecific antibody (e.g., bispecific antibody). The present disclosure encompasses whole antibodies or antigen-binding fragments (e.g., Fab, F (ab)2, Fv, scFv, sc (Fv)2, diabodies).

For example, antibodies such as exemplary anti-gp 120 antibody 3 can be made by making and expressing nucleic acids encoding antibody amino acid sequences.

Exemplary anti-gp 120 antibody 4

Another exemplary anti-gp 120 antibody, exemplary anti-gp 120 antibody 4, has the same six CDRs as exemplary anti-gp 120 antibody 1. The antibody comprises a heavy chain variable region comprising SEQ ID NO:7 or a VH sequence consisting of it, and a VL sequence comprising or consisting of the amino acid sequence set forth in seq id no:

Figure BDA0002328692870000491

in some cases, the VL is linked to the human λ constant region directly or through an intervening amino acid sequence (e.g., a G-S linker).

An exemplary anti-gp 120 antibody 4 comprises a heavy chain comprising SEQ ID NO: 9 and a light chain comprising or consisting of the amino acid sequence set forth in seq id no:

in some cases, an anti-gp 120 antibody of the present disclosure binds to a polypeptide comprising or consisting of SEQ ID NO: 21, and a protein consisting of the amino acid sequence shown in the figure 21. In some cases, an anti-gp 120 antibody of the present disclosure binds to a polypeptide comprising or consisting of SEQ ID NO: 38. In some cases, the anti-gp 120 antibodies of the present disclosure bind to free HIV-1 virus. In some cases, an anti-gp 120 antibody of the disclosure binds to an HIV-1 infected cell. In some cases, the anti-gp 120 antibodies of the present disclosure bind to free HIV-1 virus and HIV-1 infected cells. In some cases, an anti-gp 120 antibody of the present disclosure binds to at least two different HIV-1 strains (e.g., group M, group N, group O, or group P). In one embodiment, an anti-gp 120 antibody of the disclosure binds pwito.c/2474 (accession No. JN944948 and NIH AIDS Reagent Program catalog No. 11739). In another embodiment, an anti-gp 120 antibody of the disclosure binds to pCH058.c/2960 (accession No. JN944940 and NIH AIDS Reagent Program catalog No. 700010058).

In some embodiments, the variable heavy chain of exemplary anti-gp 120 antibody 4 is linked to a heavy chain constant region comprising a CH1 domain and a hinge region. In some embodiments, the variable heavy chain of exemplary anti-gp 120 antibody 4 is linked to a heavy chain constant region comprising a CH3 domain. In certain embodiments, the variable heavy chain of exemplary anti-gp 120 antibody 4 is linked to a heavy chain constant region comprising a CH1 domain from IgG4, a hinge region, and a CH2 domain and a CH3 domain (e.g., from IgG 1). In certain embodiments, the variable heavy chain of exemplary anti-gp 120 antibody 1 is linked to a heavy chain constant region comprising a CH1 domain, a CH2 domain, and a CH3 domain from IgG1 (e.g., human IgG1, e.g., IgG1m3 allotype), and an IgG3 hinge region (e.g., "open" IgG3 hinge variant "IgG 3C-" described in WO 2017/096221). In certain embodiments, such chimeric antibodies comprise one or more additional mutations in the heavy chain constant region that increase the stability of the chimeric antibody. In certain embodiments, the heavy chain constant region comprises a substitution that alters a property of the antibody (e.g., reduces Fc receptor binding, increases or decreases antibody glycosylation, decreases binding to C1q, increases half-life, decreases effector function).

In certain embodiments, the anti-gp 120 antibody is an IgG antibody. In one embodiment, the antibody is IgG 1. In another embodiment, the antibody is IgG 2. In some embodiments, the antibody has a chimeric heavy chain constant region (e.g., having the CH1, hinge, and CH2 regions of IgG4 and the CH3 region of IgG 1).

In particular embodiments, the antibodies of the present disclosure comprise an IgG1 heavy chain having an allotype G1m1, nG1m2, G1m3, G1m17,1, G1m17,1,2, G1m3,1, or G1m 17.

In certain embodiments, the anti-gp 120 antibody is a human IgG 1/human kappa antibody. In some embodiments, an antibody of the present disclosure comprises a kappa light chain having an allotype selected from Km1, Km1,2, or Km 3. In certain embodiments, the anti-gp 120 antibody is a human IgG 1/human λ antibody. In some embodiments, an antibody of the present disclosure comprises a λ light chain selected from λ 1, λ 2, λ 3, λ 4, λ 5, λ 6, and λ 7.

In some embodiments, the VH of exemplary anti-gp 120 antibody 4 (e.g., an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% identity to SEQ ID NO:7, or having from 0 to 5 (i.e., 1,2, 3, 4, or 5) amino acid substitutions (e.g., a conservative substitution within SEQ ID NO: 7)) is linked directly to or through an intervening amino acid sequence (e.g., a G-S linker) with the wild-type IgG1m3 sequence (SEQ ID NO: 77). In certain embodiments, the VH of exemplary anti-gp 120 antibody 4 (e.g., an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% identity to SEQ ID NO:7, or having from 0 to 5 (i.e., 1,2, 3, 4, or 5) amino acid substitutions (e.g., a conservative substitution in SEQ ID NO: 7)) is identical to a VH having SEQ ID NO: the mutated IgG1m3 sequences with 1 to 10 (e.g., 1,2, 3, 4, 5, 6,7, 8,9, 10) amino acid substitutions in 77 (e.g., to reduce effector function and/or increase half-life) are linked directly or by insertion of an amino acid sequence (e.g., a G-S linker). Exemplary amino acid substitutions are described later in this disclosure.

In some embodiments, the VL of exemplary anti-gp 120 antibody 4 (e.g., an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% identity to SEQ ID NO: 83, or having from 0 to 5 (i.e., 1,2, 3, 4, or 5) amino acid substitutions (e.g., a conservative substitution in SEQ ID NO: 83)) is linked directly to a wild-type human λ 2 sequence (SEQ ID NO: 89) or via an intervening amino acid sequence (e.g., a G-S linker). In certain embodiments, the VL of exemplary anti-gp 120 antibody 4 (e.g., an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% identity to SEQ ID NO: 83, or having from 0 to 5 (i.e., 1,2, 3, 4, or 5) amino acid substitutions (e.g., a conservative substitution in SEQ ID NO: 83)) is identical to the VL of SEQ ID NO: 89 with 1 to 5 (i.e., 1,2, 3, 4, 5) substitutions are linked directly or via an intervening amino acid sequence (e.g., a G-S linker).

In a particular embodiment, the anti-gp 120 antibody is a human IgG1m 3/human λ 2 antibody.

Exemplary anti-gp 120 antibody 4 can be used as a monospecific antibody or a multispecific antibody (e.g., bispecific antibody). Intact antibodies or antigen-binding fragments (e.g., Fab, F (ab)2, Fv, scFv, sc (Fv)2, diabodies) can be used.

For example, an antibody such as exemplary anti-gp 120 antibody 4 can be made by making and expressing a nucleic acid encoding the amino acid sequence of the antibody.

Exemplary anti-gp 120 antibody 5

Another exemplary anti-gp 120 antibody, exemplary anti-gp 120 antibody 5, has the same six CDRs as exemplary anti-gp 120 antibody 1. The antibody comprises a heavy chain variable region comprising SEQ ID NO:7 or a VH sequence consisting of it, and a VL sequence comprising or consisting of the amino acid sequence set forth in seq id no:

Figure BDA0002328692870000521

in some cases, the VL is linked to the human λ constant region directly or through an intervening amino acid sequence (e.g., a G-S linker).

An exemplary anti-gp 120 antibody 5 comprises a heavy chain comprising SEQ ID NO: 9 or a heavy chain consisting of the amino acid sequence shown below and a light chain comprising or consisting of the amino acid sequence shown below

Figure BDA0002328692870000522

In some cases, an anti-gp 120 antibody of the present disclosure binds to a polypeptide comprising or consisting of SEQ ID NO: 21, and a protein consisting of the amino acid sequence shown in the figure 21. In some cases, an anti-gp 120 antibody of the present disclosure binds to a polypeptide comprising or consisting of SEQ ID NO: 38. In some cases, the anti-gp 120 antibodies of the present disclosure bind to free HIV-1 virus. In some cases, an anti-gp 120 antibody of the disclosure binds to an HIV-1 infected cell. In some cases, the anti-gp 120 antibodies of the present disclosure bind to free HIV-1 virus and HIV-1 infected cells. In some cases, an anti-gp 120 antibody of the present disclosure binds to at least two different HIV-1 strains (e.g., group M, group N, group O, or group P). In one embodiment, an anti-gp 120 antibody of the disclosure binds pwito.c/2474 (accession No. JN944948 and NIH AIDS Reagent Program catalog No. 11739). In another embodiment, an anti-gp 120 antibody of the disclosure binds to pCH058.c/2960 (accession No. JN944940 and NIH AIDS Reagent Program catalog No. 700010058).

In some embodiments, the variable heavy chain of exemplary anti-gp 120 antibody 5 is linked to a heavy chain constant region comprising a CH1 domain and a hinge region. In some embodiments, the variable heavy chain of exemplary anti-gp 120 antibody 5 is linked to a heavy chain constant region comprising a CH3 domain. In certain embodiments, the variable heavy chain of exemplary anti-gp 120 antibody 5 is linked to a heavy chain constant region comprising a CH1 domain from IgG4, a hinge region, and a CH2 domain and a CH3 domain (e.g., from IgG 1). In certain embodiments, the variable heavy chain of exemplary anti-gp 120 antibody 1 is linked to a heavy chain constant region comprising a CH1 domain, a CH2 domain, and a CH3 domain from IgG1 (e.g., a human IgG1, e.g., an IgG1m3 allotype), and an IgG3 hinge region (e.g., an "open" IgG3 hinge variant "IgG 3C-" described in WO 2017/096221). In certain embodiments, such chimeric antibodies comprise one or more additional mutations in the heavy chain constant region that increase the stability of the chimeric antibody. In certain embodiments, the heavy chain constant region comprises a substitution that alters a property of the antibody (e.g., reduces Fc receptor binding, increases or decreases antibody glycosylation, decreases binding to C1q, increases half-life, decreases effector function).

In certain embodiments, the anti-gp 120 antibody is an IgG antibody. In one embodiment, the antibody is IgG 1. In another embodiment, the antibody is IgG 2. In some embodiments, the antibody has a chimeric heavy chain constant region (e.g., having the CH1, hinge, and CH2 regions of IgG4 and the CH3 region of IgG 1).

In particular embodiments, the antibodies of the present disclosure comprise an IgG1 heavy chain having an allotype G1m1, nG1m2, G1m3, G1m17,1, G1m17,1,2, G1m3,1, or G1m 17.

In certain embodiments, the anti-gp 120 antibody is a human IgG 1/human kappa antibody. In some embodiments, an antibody of the present disclosure comprises a kappa light chain having an allotype selected from Km1, Km1,2, or Km 3. In certain embodiments, the anti-gp 120 antibody is a human IgG 1/human λ antibody. In some embodiments, an antibody of the present disclosure comprises a λ light chain selected from λ 1, λ 2, λ 3, λ 4, λ 5, λ 6, and λ 7.

In some embodiments, the VH of exemplary anti-gp 120 antibody 5 (e.g., an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% identity to SEQ ID NO:7, or having from 0 to 5 (i.e., 1,2, 3, 4, or 5) amino acid substitutions (e.g., a conservative substitution within SEQ ID NO: 7)) is linked directly to a wild-type IgG1m3 sequence (SEQ ID NO: 77) or via an intervening amino acid sequence (e.g., a G-S linker). In certain embodiments, the VH (e.g., an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% identity to SEQ ID No. 7, or having from 0 to 5 (i.e., 1,2, 3, 4, or 5) amino acid substitutions (e.g., a conservative substitution in SEQ ID No. 7)) of exemplary anti-gp 120 antibody 5 is identical to a VH having SEQ ID NO:1 to 10 (e.g., 1,2, 3, 4, 5, 6,7, 8,9, 10) amino acid substitutions in 77 (e.g., to reduce effector function and/or extend half-life) are linked directly or by intervening amino acid sequences (e.g., G-S linkers). Exemplary amino acid substitutions are described later in this disclosure.

In some embodiments, the VL of exemplary anti-gp 120 antibody 5 (e.g., an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% identity to SEQ ID NO: 84, or having from 0 to 5 (i.e., 1,2, 3, 4, or 5) amino acid substitutions (e.g., a conservative substitution in SEQ ID NO: 84)) is linked directly to a wild-type human λ 2 sequence (SEQ ID NO: 89) or via an intervening amino acid sequence (e.g., a G-S linker). In certain embodiments, the VL of exemplary anti-gp 120 antibody 5 (e.g., an amino acid sequence having at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, or 100% identity to SEQ ID NO: 84, or having from 0 to 5 (i.e., 1,2, 3, 4, or 5) amino acid substitutions (e.g., a conservative substitution in SEQ ID NO: 84)) is identical to the VL of SEQ ID NO: 89 with 1 to 5 (i.e., 1,2, 3, 4, 5) substitutions are linked directly or via an intervening amino acid sequence (e.g., a G-S linker).

In a particular embodiment, the anti-gp 120 antibody is a human IgG1m 3/human λ 2 antibody.

Exemplary anti-gp 120 antibody 5 can be used as a monospecific antibody or a multispecific antibody (e.g., bispecific antibody). The present disclosure encompasses whole antibodies or antigen-binding fragments (e.g., Fab, F (ab)2, Fv, scFv, sc (Fv)2, diabodies).

Antibodies such as exemplary anti-gp 120 antibody 5 can be prepared, for example, by preparing and expressing nucleic acids encoding the amino acid sequences of the antibodies.

CD3

DifferentiationThe cluster (CD3) is a multimeric protein complex, which consists of four different polypeptide chains: epsilon (epsilon), gamma (gamma), delta (delta), and zeta (zeta) which assemble and function as three pairs of dimers (epsilon gamma, epsilon delta, zeta). The CD3 protein has an N-terminal extracellular region, a transmembrane domain, and a cytoplasmic tail in which an Immunoreceptor Tyrosine Activation Motif (ITAM) is located. The extracellular domains of CD3 epsilon, gamma, and delta comprise immunoglobulin-like domains and are therefore considered to be part of the immunoglobulin superfamily. CD3/T cell co-receptor contributes to activation of CD8+T cells and CD4+T cells.

The amino acid sequence of human CD3 epsilon can be found in unaprotkb-P07766 and is provided below (signal sequence underlined):

Figure BDA0002328692870000551

the amino acid sequence of human CD3 δ can be found in unaprotkb-P04234 and is provided below (signal sequence underlined):

Figure BDA0002328692870000552

Figure BDA0002328692870000562

antibodies that bind human CD3 are well known in the art (see, e.g., Kuhn & Weiner, Immunotherapy, 8 (8): 889-906 (2016); WO 2015/104346). OKT3(Muromab), an anti-CD 3 antibody against CD3 epsilon, has been approved clinically for use in humans to induce immunosuppression in solid organ transplants for the prevention and treatment of rejection (Norman, Therapeutic Drug Monitoring, 17, 615-620 (1995)). Teplizumab, also known as hOKT3 γ 1(Ala-Ala) and MGA031, is a humanized IgG1 antibody developed by grafting the complementarity determining regions of OKT3 into the human IgG1 framework. Introduction of two point mutations into the Fc portion thereof reduces FcR binding. Otelixizumab (chaclycd 3, TRX4, GSK2136525) was derived from the rat antibody YTH 12.5. The humanized IgG1 carries a single mutation in the γ 1Fc portion to avoid glycosylation and thus inhibit FcR binding. Visilizumab (Nuvion, HuM291) is a humanized IgG2 antibody that is made non-mitogenic by two point mutations in its Fc region. Foralumab (28F 11-AE; NI-0401) is a fully human anti-CD 3 mAb; the Fc portion of this human IgG1 was mutated so that the mAb was non-FcR binding in vitro and showed only a small amount of cytokine release in vivo while maintaining CD3/TCR modulation and T cell depletion.

Non-limiting examples of anti-CD 3 antibodies are also disclosed in US 2016/0333095a 1.

In certain embodiments, the anti-CD 3 antibodies of the present disclosure bind to human CD 3. In some cases, an anti-CD 3 antibody of the present disclosure binds to human CD3 epsilon. In other embodiments, the anti-CD 3 antibodies of the present disclosure bind to human CD3 δ.

Exemplary anti-CD 3 antibody 1

Relevant sequence information for exemplary anti-CD 3 antibody 1 is provided in table 4.

TABLE 4

Figure BDA0002328692870000571

Figure BDA0002328692870000581

anti-CD 3 antibodies may include the heavy chain CDR1, CDR2, and CDR3 and light chain CDR1, CDR2, and CDR3 of exemplary anti-CD 3 antibody 1. In one embodiment, the CDRs are defined based on the Kabat definition. In another embodiment, the CDRs are defined based on the Chothia definition. In another embodiment, the CDRs are defined based on IMGT definitions. In another embodiment, the CDRs are defined based on the honeygger definition. In another embodiment, the CDRs are based on Chothia definitions from the Abysis definition. In another embodiment, the CDR based on Chothia/AbM CDR definition. In another embodiment, the CDRs are defined based on contact definitions. For example, these CDRs can be determined using the abbis database (www.bioinf.org.uk/analysis/sequence _ input/key _ annotation.

In certain instances, an anti-CD 3 antibody comprises an amino acid sequence that is at least 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the variable heavy chain of exemplary anti-CD 3 antibody 1. In some embodiments, the anti-CD 3 antibody comprises an amino acid sequence that is at least 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the heavy chain of exemplary anti-CD 3 antibody 1. In some cases, an anti-CD 3 antibody comprises an amino acid sequence that is at least 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the variable light chain of exemplary anti-CD 3 antibody 1. In some cases, an anti-CD 3 antibody comprises an amino acid sequence that is at least 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the light chain of exemplary anti-CD 3 antibody 1. In certain embodiments, the anti-CD 3 antibody comprises an amino acid sequence that is at least 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the variable heavy and variable light chains of exemplary anti-CD 3 antibody 1. In some embodiments, the anti-CD 3 antibody comprises an amino acid sequence at least 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the heavy chain of exemplary anti-CD 3 antibody 1 and comprises an amino acid sequence at least 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the light chain of exemplary anti-CD 3 antibody 1.

In some embodiments, the variable heavy chain of exemplary anti-CD 3 antibody 1 is linked to a heavy chain constant region comprising a CH1 domain and a hinge region. In some embodiments, the variable heavy chain of exemplary anti-CD 3 antibody 1 is linked to a heavy chain constant region comprising a CH3 domain. In certain embodiments, the variable heavy chain of exemplary anti-CD 3 antibody 1 is linked to a heavy chain constant region comprising a CH1 domain from IgG4, a hinge region, and a CH2 domain and a CH3 domain (e.g., from IgG 1). In certain embodiments, the variable heavy chain of exemplary anti-gp 120 antibody 1 is linked to a heavy chain constant region comprising a CH1 domain, a CH2 domain, and a CH3 domain from IgG1 (e.g., human IgG1, e.g., IgG1m3 allotype), and an IgG3 hinge region (e.g., "open" IgG3 hinge variant "IgG 3C" described in WO 2017/096221). In certain embodiments, such chimeric antibodies comprise one or more additional mutations in the heavy chain constant region that increase the stability of the chimeric antibody. In certain embodiments, the heavy chain constant region comprises a substitution that alters a property of the antibody (e.g., reduces Fc receptor binding, increases or decreases antibody glycosylation, decreases binding to C1q, increases half-life, decreases effector function).

In certain embodiments, the anti-CD 3 antibody is an IgG antibody. In one embodiment, the antibody is IgG 1. In another embodiment, the antibody is IgG 2. In some embodiments, the antibody has a chimeric heavy chain constant region (e.g., having the CH1, hinge, and CH2 regions of IgG4 and the CH3 region of IgG 1).

In particular embodiments, the antibodies of the present disclosure comprise an IgG1 heavy chain having an allotype G1m1, nG1m2, G1m3, G1m17,1, G1m17,1,2, G1m3,1, or G1m 17.

In particular embodiments, the VH of exemplary anti-CD 3 antibody 1 is linked to wild-type IgG1m3 Fc (SEQ ID NO: 77). In certain instances, the VH of exemplary anti-CD 3 antibody 1 is linked to a mutant IgG1m3 sequence having 1 to 10 (e.g., 1,2, 3, 4, 5, 6,7, 8,9, 10) amino acid substitutions (e.g., decreasing effector function and/or increasing half-life) in SEQ ID NO: 77. Exemplary amino acid substitutions are described below.

In a specific embodiment, the VL of exemplary anti-CD 3 antibody 1 is linked to a human λ 2 sequence (SEQ ID NO: 89). In some cases, the VL of anti-CD 3 antibody 1 is identical to a VL having SEQ ID NO: 89 (e.g., 1,2, 3, 4, 5, 6,7, 8,9, 10) amino acid substitutions in a human λ 2 sequence. Such amino acid substitutions are described below.

In certain embodiments, the anti-CD 3 antibody is a human IgG 1/human λ antibody.

For example, an antibody such as exemplary anti-CD 3 antibody 1 can be prepared by preparing and expressing a nucleic acid encoding the amino acid sequence of the antibody.

CD89

CD89 (cluster of differentiation 89), also known as the Fc fragment of IgA receptor (FCAR), is the transmembrane receptor Fc α RI. Fc α RI binds the heavy chain constant region of immunoglobulin a (IgA) antibodies Fc α RI is expressed on the cell surface of myeloid lineage cells, including neutrophils, monocytes, macrophages and eosinophils.

The amino acid sequence of human CD89 from UniProtKB-P24071 is provided below:

Figure BDA0002328692870000601

antibodies that bind human CD89 are well known in the art (see, e.g., Fishwild et al, Nature Biotechnol., 14 (7): 845-. In some embodiments, the anti-CD 89 antibody is one of 14.1, 7.4, or 8.2 (also referred to as 14a8, 7F12, and 8D2, respectively). Any of these antibodies or variants thereof may be used in the multispecific antibodies disclosed herein.

In certain embodiments, an anti-CD 89 antibody of the present disclosure or a multispecific antibody disclosed herein binds to a polypeptide comprising seq id NO: 95 or a polypeptide consisting thereof.

Exemplary anti-CD 89 antibody 1

The following table provides relevant sequence information for exemplary anti-CD 89 antibody 1.

Figure BDA0002328692870000611

anti-CD 89 antibodies may include the heavy chain CDR1, CDR2, and CDR3 and light chain CDR1, CDR2, and CDR3 of exemplary anti-CD 89 antibody 1. In one embodiment, the CDRs are defined based on the Kabat definition. In another embodiment, the CDRs are defined based on the Chothia definition. In another embodiment, the CDRs are defined based on IMGT definitions. In another embodiment, the CDRs are defined based on the honeygger definition. In another embodiment, the CDRs are defined based on Chothia from the Abysis definition. In another embodiment, the CDRs are defined based on Chothia/AbM CDRs. In another embodiment, the CDRs are defined based on contact definitions. For example, these CDRs can be determined using the AbYsis database (www.bioinf.org.uk/analysis/sequence _ input/key _ annotation n.

In certain instances, an anti-CD 89 antibody comprises an amino acid sequence that is at least 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the variable heavy chain of exemplary anti-CD 89 antibody 1. In some embodiments, the anti-CD 89 antibody comprises an amino acid sequence that is at least 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the heavy chain of exemplary anti-CD 89 antibody 1. In some cases, an anti-CD 89 antibody comprises an amino acid sequence that is at least 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the light chain of exemplary anti-CD 89 antibody 1. In certain instances, an anti-CD 89 antibody comprises an amino acid sequence that is at least 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the light chain of exemplary anti-CD 89 antibody 1. In certain embodiments, the anti-CD 89 antibody comprises an amino acid sequence that is at least 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the variable heavy and variable light chains of exemplary anti-CD 89 antibody 1. In some embodiments, the anti-CD 89 antibody comprises an amino acid sequence at least 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the heavy chain of exemplary anti-CD 89 antibody 1 and comprises an amino acid sequence at least 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the light chain of exemplary anti-CD 89 antibody 1.

In some embodiments, the variable heavy chain of exemplary anti-CD 89 antibody 1 is linked to a heavy chain constant region comprising a CH1 domain and a hinge region. In some embodiments, the variable heavy chain of exemplary anti-CD 89 antibody 1 is linked to a heavy chain constant region comprising a CH3 domain. In certain embodiments, the variable heavy chain of exemplary anti-CD 89 antibody 1 is linked to a heavy chain constant region comprising a CH1 domain from IgG4, a hinge region, and a CH2 domain and a CH3 domain (e.g., from IgG 1). In certain embodiments, the variable heavy chain of exemplary anti-CD 89 antibody 1 is linked to a heavy chain constant region comprising a CH1 domain, a CH2 domain, and a CH3 domain from IgG1 (e.g., human IgG1, e.g., IgG1m3 allotype), and an IgG3 hinge region (e.g., "open" IgG3 hinge variant "IgG 3C-" described in WO 2017/096221). In certain embodiments, such chimeric antibodies comprise one or more additional mutations in the heavy chain constant region that increase the stability of the chimeric antibody. In certain embodiments, the heavy chain constant region comprises a substitution that alters a property of the antibody (e.g., reduces Fc receptor binding, increases or decreases antibody glycosylation, decreases binding to C1q, increases half-life, decreases effector function).

In certain embodiments, the anti-CD 89 antibody is an IgG antibody. In one embodiment, the antibody is IgG 1. In another embodiment, the antibody is IgG 2. In some embodiments, the antibody has a chimeric heavy chain constant region (e.g., having the CH1, hinge, and CH2 regions of IgG4 and the CH3 region of IgG 1).

In particular embodiments, the antibodies of the present disclosure comprise an IgG1 heavy chain having an allotype G1m1, nG1m2, G1m3, G1m17,1, G1m17,1,2, G1m3,1, or G1m 17.

In particular embodiments, the VH of exemplary anti-CD 89 antibody 1 is linked to wild-type IgG1m3 Fc (SEQ ID NO: 77). In certain instances, the VH of exemplary anti-CD 89 antibody 1 is identical to the VH having SEQ ID NO:1 to 10 (e.g., 1,2, 3, 4, 5, 6,7, 8,9, 10) amino acid substitutions in 77 (e.g., to reduce effector function and/or increase half-life) are linked to the mutant IgG1m3 sequence. Exemplary amino acid substitutions are described below.

In a specific embodiment, the VL of exemplary anti-CD 89 antibody 1 is linked to a human λ 2 sequence (SEQ ID NO: 89). In certain instances, the VL of exemplary anti-CD 89 antibody 1 is identical to the VL having SEQ ID NO: 89 (e.g., 1,2, 3, 4, 5, 6,7, 8,9, 10) amino acid substitutions in a human λ 2 sequence. Such amino acid substitutions are described below.

In certain embodiments, the anti-CD 89 antibody is a human IgG 1/human λ antibody.

For example, an antibody such as exemplary anti-CD 89 antibody 1 can be prepared by preparing and expressing a nucleic acid encoding the amino acid sequence of the antibody.

Multispecific antibodies

In another aspect, the disclosure features a multispecific antibody. Multispecific antibodies are antibodies that bind two or more different epitopes (e.g., bispecific antibodies, trivalent antibodies, tetravalent antibodies). The anti-gp 120 and anti-CD 3 antibodies described above or anti-gp 120 and anti-CD 89 may be included as part of a multispecific antibody. The multispecific antibody may have a binding site for at least one other antigen or one other epitope that is not bound by the anti-gp 120 or anti-CD 3 (or anti-CD 89) antibody binding site of the multispecific antibody. The anti-gp 120/anti-CD 3 multispecific antibody or anti-gp 120/anti-CD 89 multispecific antibody may comprise a dimerization domain and three or more (e.g., three, four, five, six) antigen binding sites. Exemplary dimerizationThe domain comprises (or consists of) an Fc region. The anti-gp 120/anti-CD 3 multispecific antibody or anti-gp 120/anti-CD 89 multispecific antibody may comprise (or consist of) three to about eight (i.e., three, four, five, six, seven, eight) antigen binding sites. The multispecific antibody optionally comprises at least one polypeptide chain (e.g., two polypeptide chains, three polypeptide chains), wherein the polypeptide chain comprises three or more variable domains. For example, a polypeptide chain can comprise, for example, VD1- (X1)n-VD2-(X2)nFc or VD1- (X1)n-VD2-(X2)n-VD3-(X3)n-Fc, wherein VD1 is a first variable domain, VD2 is a second variable domain, VD3 is a third variable domain, Fc is a polypeptide chain of an Fc region, X1, X2 and X3 represent amino acid or peptide spacers, and n is 0 or 1. In certain instances, the variable domains may each be an scFv. Multispecific antibodies can be readily produced by recombinant expression of nucleic acids encoding the polypeptide chains of the antibody.

Bispecific antibodies

In one aspect, the multispecific antibody is a bispecific antibody. Bispecific antibodies are antibodies that have binding specificities for two different epitopes. Bispecific antibodies have two "arms". One arm of the bispecific antibody binds to one epitope and the other arm binds to the other epitope. In one embodiment, one arm of the bispecific antibody binds a first antigen and the other arm of the bispecific antibody binds a second antigen. In another embodiment, the two arms of the bispecific antibody bind two different epitopes of the same antigen.

In one aspect, the disclosure features bispecific antibodies that specifically bind gp120 and specifically bind a second antigen (e.g., a trigger molecule on a leukocyte, such as a T cell receptor molecule (e.g., CD3), or an Fc receptor of IgG (fcyr), such as fcyri (CD64), fcyrii (CD32), fcyriii (CD16), or CD89), thereby concentrating and localizing cellular defense mechanisms to infected cells.

In particular embodiments, one arm of the bispecific antibody specifically binds gp120, while the other arm specifically binds CD3 (e.g., human CD3 (e.g., human CD3 epsilon, human CD3 delta)). In another specific embodiment, one arm of the bispecific antibody specifically binds gp120 and the other arm specifically binds CD89 (e.g., human CD 89). In certain embodiments, the arms of the bispecific antibody that binds gp120 comprise the six CDRs of the exemplary anti-gp 120 antibody 1. In some cases, the CDRs are defined according to Kabat. In other embodiments, the CDRs are defined according to Chothia. In still other embodiments, the CDRs are defined according to the IMGT definition. In still other embodiments, the CDRs are defined according to the honeyger definition. In certain embodiments, the arm of a bispecific antibody that binds gp120 comprises an amino acid sequence that is at least 75%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the VH (SEQ ID NO: 7) of exemplary anti-gp 120 antibody 1. In certain embodiments, the arm of a bispecific antibody that binds gp120 comprises an amino acid sequence that is at least 75%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the VL (SEQ ID NO: 8) of exemplary anti-gp 120 antibody 1. In certain embodiments, the arm of a bispecific antibody that binds gp120 comprises an amino acid sequence that is at least 75%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the VH (SEQ ID NO: 7) and VL (SEQ ID NO: 8), respectively, of exemplary anti-gp 120 antibody 1. In certain embodiments, the arm of a bispecific antibody that binds gp120 comprises an amino acid sequence that is at least 75%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the VH (SEQ ID NO: 7) and VL (SEQ ID NO: 81), respectively, of exemplary anti-gp 120 antibody 2. In certain embodiments, the arm of a bispecific antibody that binds gp120 comprises an amino acid sequence that is at least 75%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the VH (SEQ ID NO: 7) and VL (SEQ ID NO: 82), respectively, of exemplary anti-gp 120 antibody 3. In certain embodiments, the arm of a bispecific antibody that binds gp120 comprises an amino acid sequence that is at least 75%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the VH (SEQ ID NO: 7) and VL (SEQ ID NO: 83), respectively, of exemplary anti-gp 120 antibody 4. In certain embodiments, the arm of a bispecific antibody that binds gp120 comprises an amino acid sequence that is at least 75%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identical to the VH (SEQ ID NO: 7) and VL (SEQ ID NO: 84), respectively, of exemplary anti-gp 120 antibody 5. In certain embodiments, the arm of a bispecific antibody that binds gp120 comprises an amino acid sequence that is at least 75%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the heavy chain of exemplary anti-gp 120 antibody 1 (SEQ ID NO: 9). In certain embodiments, the arm of a bispecific antibody that binds gp120 comprises an amino acid sequence that is at least 75%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the light chain of an exemplary anti-gp 120 antibody (SEQ ID NO: 10). In certain embodiments, the arm of a bispecific antibody that binds gp120 comprises an amino acid sequence that is at least 75%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% identical to the heavy chain (SEQ ID NO: 9) and light chain (SEQ ID NO: 10), respectively, of exemplary anti-gp 120 antibody 1. In certain instances, the arm of a bispecific antibody that binds gp120 comprises an amino acid sequence that is at least 75%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the heavy chain (SEQ ID NO: 9) and light chain (SEQ ID NO: 40), respectively, of exemplary anti-gp 120 antibody 2. In certain instances, the arm of a bispecific antibody that binds gp120 comprises an amino acid sequence that is at least 75%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the heavy chain (SEQ ID NO: 9) and light chain (SEQ ID NO: 78), respectively, of exemplary anti-gp 120 antibody 3. In certain instances, the arm of a bispecific antibody that binds gp120 comprises an amino acid sequence that is at least 75%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the heavy chain (SEQ ID NO: 9) and light chain (SEQ ID NO: 79), respectively, of exemplary anti-gp 120 antibody 4. In certain instances, the arm of a bispecific antibody that binds gp120 comprises an amino acid sequence that is at least 75%, 80%, identical to 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the heavy chain (SEQ ID NO: 9) and light chain (SEQ ID NO: 80), respectively, of exemplary anti-gp 120 antibody 5. In one embodiment, the arm of the bispecific antibody that binds gp120 comprises SEQ ID NO:7 and SEQ ID NO: 8. In another embodiment, the arm of the bispecific antibody that binds gp120 comprises SEQ ID NO:7 and SEQ ID NO: 81, or a pharmaceutically acceptable salt thereof. In another embodiment, the arm of the bispecific antibody that binds gp120 comprises SEQ ID NO:7 and SEQ ID NO: 82. In yet another embodiment, the arm of the bispecific antibody that binds gp120 comprises SEQ ID NO:7 and SEQ ID NO: 83 of the sequence listing. In a further embodiment, the arm of the bispecific antibody that binds gp120 comprises SEQ ID NO:7 and SEQ ID NO: 84. In a particular embodiment, the arm of the bispecific antibody that binds gp120 comprises SEQ ID NO: 9 and SEQ ID NO: 10. In another specific embodiment, the arm of the bispecific antibody that binds gp120 comprises SEQ ID NO: 9 and SEQ ID NO: 40. In yet another specific embodiment, the arm of the bispecific antibody that binds gp120 comprises SEQ ID NO: 9 and SEQ ID NO: 78, or a pharmaceutically acceptable salt thereof. In a further embodiment, the arm of the bispecific antibody that binds gp120 comprises SEQ ID NO: 9 and SEQ ID NO: 79. In another embodiment, the arm of the bispecific antibody that binds gp120 comprises SEQ ID NO: 9 and SEQ ID NO: 80. In some cases, the arm of a bispecific antibody that binds gp120 binds to a polypeptide comprising or consisting of SEQ ID NO: 21, and a protein consisting of the amino acid sequence shown in the figure 21. In some cases, the arm of a bispecific antibody that binds gp120 binds to a polypeptide comprising or consisting of SEQ ID NO: 38, or a pharmaceutically acceptable salt thereof. In some cases, the arms of the bispecific antibody that bind gp120 bind free HIV-1 virus. In some cases, arms of bispecific antibodies that bind gp120 bind to HIV-1 infected cells. In some cases, arms of bispecific antibodies that bind gp120 bind to free HIV-1 virus and HIV-1 infected cells. In some cases, the arms of a bispecific antibody that binds gp120 bind to at least two HIV-1 strains (e.g., group M, group N, group O, or group P). In one embodiment, the arms of the bispecific antibody that binds gp120 bind pwito.c/2474 (accession No. JN944948 and NIH AIDS Reagent Program catalog No. 11739). In another embodiment, the arms of the bispecific antibody that binds gp120 bind pCH058.c/2960 (accession No. JN944940 and NIH AIDS Reagent Program catalog No. 700010058).

In certain embodiments, the arms of a bispecific antibody that binds human CD3 comprise the six CDRs of exemplary anti-human CD3 antibody 1. In some cases, the CDRs are according to the Kabat definition. In other embodiments, the CDRs are defined according to Chothia. In still other embodiments, the CDRs are defined according to the IMGT definition. In still other embodiments, the CDRs are defined according to the honeyger definition. In certain embodiments, the arm of a bispecific antibody that binds human CD3 comprises an amino acid sequence that is at least 75%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the VH (SEQ ID NO: 17) of exemplary anti-CD 3 antibody 1. In certain embodiments, the arm of a bispecific antibody that binds human CD3 comprises an amino acid sequence that is at least 75%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the VL of exemplary anti-CD 3 antibody 1 (SEQ ID NO: 18). In certain embodiments, the arm of a bispecific antibody that binds human CD3 comprises an amino acid sequence that is at least 75%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the VH (SEQ ID NO: 17) and VL (SEQ ID NO: 18), respectively, of exemplary anti-human CD3 antibody 1. In certain embodiments, the arm of a bispecific antibody that binds human CD3 comprises an amino acid sequence that is at least 75%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the heavy chain of exemplary anti-human CD3 antibody 1 (SEQ ID NO: 19). In certain embodiments, the arm of a bispecific antibody that binds CD3 comprises an amino acid sequence that is at least 75%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the light chain of exemplary anti-human CD3 antibody 1 (SEQ id no: 20). In certain embodiments, the arm of a bispecific antibody that binds human CD3 comprises an amino acid sequence that is at least 75%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the heavy chain (SEQ ID NO: 19) and light chain (SEQ ID NO: 20), respectively, of exemplary anti-human CD3 antibody 1. In a particular embodiment, the arm of the bispecific antibody that binds human CD3 comprises SEQ ID NO: 17 and SEQ ID NO: 18. In another specific embodiment, the arm of the bispecific antibody that binds human CD3 comprises SEQ ID NO: 19 and SEQ ID NO: 20. In certain embodiments, the arms of a bispecific antibody that binds human CD3 bind human CD3 epsilon.

In certain embodiments, the arms of a bispecific antibody that binds human CD89 comprise the six CDRs of exemplary anti-human CD89 antibody 1. In some cases, the CD is according to Kabat. In other embodiments, the CDRs are defined according to Chothia. In still other embodiments, the CDRs are defined according to the IMGT definition. In still other embodiments, the CDRs are defined according to the honeyger definition. In certain embodiments, the arm of a bispecific antibody that binds human CD89 comprises an amino acid sequence that is at least 75%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the VH (SEQ ID NO: 96) of exemplary anti-CD 89 antibody 1. In certain embodiments, the arm of a bispecific antibody that binds human CD89 comprises an amino acid sequence that is at least 75%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the VL of exemplary anti-CD 89 antibody 1 (SEQ ID NO: 101). In certain embodiments, the arm of a bispecific antibody that binds human CD89 comprises an amino acid sequence that is at least 75%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the VH (SEQ ID NO: 96) and VL (SEQ ID NO: 101), respectively, of exemplary anti-human CD89 antibody 1. In certain embodiments, the arm of a bispecific antibody that binds human CD89 comprises an amino acid sequence that is at least 75%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the heavy chain of exemplary anti-human CD89 antibody 1 (SEQ ID NO: 97). In certain embodiments, the arm of a bispecific antibody that binds CD89 comprises an amino acid sequence that is at least 75%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the light chain of exemplary anti-human CD89 antibody 1 (SEQ ID NO: 102). In certain embodiments, the arm of a bispecific antibody that binds human CD89 comprises an amino acid sequence that is at least 75%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% identical to the heavy chain (SEQ ID NO: 97) and light chain (SEQ ID NO: 102), respectively, of exemplary anti-human CD89 antibody 1. In a particular embodiment, the arm of the bispecific antibody that binds human CD89 comprises SEQ ID NO: 96 and SEQ ID NO: 101, or a pharmaceutically acceptable salt thereof. In another specific embodiment, the arm of the bispecific antibody that binds human CD89 comprises SEQ ID NO: 97 and SEQ ID NO: 102. In certain embodiments, the arm of a bispecific antibody that binds human CD89 binds human CD 89.

In certain embodiments, one arm of the bispecific antibody comprises an scFv that binds gp 120. In certain embodiments, one arm of the bispecific antibody comprises an scFv that binds human CD 3. In certain embodiments, one arm of the bispecific antibody comprises an scFv that binds human CD 89. In certain embodiments, a bispecific antibody may comprise a chimeric antibody or a humanized antibody. In certain embodiments, a bispecific antibody can comprise a F (ab') 2 fragment.

In one aspect, bispecific antibodies of the disclosure bind gp120 and human CD3 (e.g., CD3 epsilon, CD3 delta) and can achieve killing of HIV-1 infected cells. In one instance, such bispecific antibody that binds gp120 comprises SEQ id no:1, VH-CDR1, SEQ ID NO: 2, VH-CDR2 of SEQ ID NO:3, VH-CDR3 of SEQ ID NO:4, VL-CDR1 of SEQ ID NO:5 and the VL-CDR2 of SEQ ID NO:6 VL-CDR 3. In one instance, such a bispecific antibody that binds human CD3 comprises SEQ ID NO: 11, VH-CDR1 of SEQ ID NO:12, VH-CDR2 of SEQ ID NO: 13, VH-CDR3 of SEQ ID NO: 14, VL-CDR1 of SEQ ID NO: 15 and VL-CDR2 of SEQ ID NO: 16 VL-CDR 3. In another instance, a bispecific antibody that binds gp120 and human CD3 comprises on its gp 120-binding arm: SEQ ID NO:1, VH-CDR1, SEQ ID NO: 2, VH-CDR2 of SEQ ID NO:3, VH-CDR3 of SEQ ID NO:4, seq id NO:5 and the VL-CDR2 of SEQ ID NO:6 VL-CDR 3; and comprises on its human CD3 binding arm: SEQ ID NO: 11, VH-CDR1 of SEQ ID NO:12, VH-CDR2 of SEQ ID NO: 13, VH-CDR3 of SEQ ID NO: 14, VL-CDR1 of SEQ ID NO: 15 and VL-CDR2 of SEQ ID NO: 16 VL-CDR 3.

In another instance, such a bispecific antibody comprises a heavy chain variable region comprising a heavy chain variable region having a sequence identical to SEQ ID NO:7 VH having an amino acid sequence at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97% or at least 98% identical. In another instance, such a bispecific antibody comprises a substitution, insertion, and/or deletion in its gp 120-binding arm that is identical to SEQ ID NO:7 VH of the same amino acid sequence. In another instance, such a bispecific antibody comprises a substitution in its gp 120-binding arm that is identical to SEQ ID NO:7 VH of the same amino acid sequence. In another instance, such a bispecific antibody comprises a peptide having a sequence identical to SEQ ID NO: 8. 81, 82, 83 or 84, at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97% or at least 98% identical. In another instance, such a bispecific antibody comprises a heavy chain variable region having substitutions, insertions, and/or deletions in its gp 120-binding arm other than 1 to 10 (i.e., 1,2, 3, 4, 5, 6,7, 8,9, 10) with SEQ ID NO: 8. 81, 82, 83 or 84, or a pharmaceutically acceptable salt thereof. In another instance, such a bispecific antibody comprises a heavy chain variable region having a sequence identical to seq id NO: 8. 81, 82, 83 or 84, or a pharmaceutically acceptable salt thereof. In another instance, such a bispecific antibody comprises a peptide having a sequence identical to SEQ ID NO: 9, a heavy chain of an amino acid sequence that is at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97% or at least 98% identical. In another instance, such a bispecific antibody comprises, in its gp120 binding arm, a substitution, insertion and/or deletion with SEQ id no: 9 the same amino acid sequence. In another instance, such a bispecific antibody comprises a substitution in its gp 120-binding arm that is identical to SEQ ID NO: 9 the same amino acid sequence. In another instance, such a bispecific antibody comprises a peptide having a sequence identical to SEQ ID NO: 10. 40, 78, 79 or 80, at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97% or at least 98% identical. In another instance, such a bispecific antibody comprises a heavy chain variable region having from 1 to 10 (i.e., 1,2, 3, 4, 5, 6,7, 8,9, 10) substitutions, insertions, and/or deletions in its gp120 binding arm that is complementary to SEQ ID NO: 10. 40, 78, 79 or 80, or a pharmaceutically acceptable salt thereof. In another instance, such a bispecific antibody comprises a substitution in its gp 120-binding arm that is identical to SEQ ID NO: 10. 40, 78, 79 or 80, or a pharmaceutically acceptable salt thereof.

In another instance, such a bispecific antibody comprises on its human CD3 binding arm a polypeptide having an amino acid sequence identical to SEQ ID NO: 17 VH of an amino acid sequence that is at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97% or at least 98% identical. In another instance, such a bispecific antibody comprises on its human CD3 binding arm an amino acid sequence that differs from SEQ ID NO: 17 VH of the same amino acid sequence. In another instance, such a bispecific antibody comprises on its human CD3 binding arm a sequence having an amino acid sequence identical to SEQ ID NO: 17 VH of the same amino acid sequence. In another instance, such a bispecific antibody comprises on its human CD3 binding arm a polypeptide having an amino acid sequence identical to SEQ ID NO: 18, VL of an amino acid sequence that is at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, or at least 98% identical. In another instance, such a bispecific antibody comprises on its human CD3 binding arm a sequence having an amino acid sequence identical to SEQ ID NO: 18, VL of the same amino acid sequence. In another instance, such a bispecific antibody comprises on its human CD3 binding arm a sequence having an amino acid sequence identical to SEQ ID NO: 18, VL of the same amino acid sequence. In another instance, such a bispecific antibody comprises a heavy chain variable region having a sequence identical to SEQ id no: 19, at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97% or at least 98% identical to the amino acid sequence of the light chain. In another instance, such a bispecific antibody comprises on its human CD3 binding arm a sequence having an amino acid sequence identical to SEQ ID NO: 19 the same amino acid sequence. In another instance, such a bispecific antibody comprises on its human CD3 binding arm a sequence having an amino acid sequence identical to SEQ ID NO: 19 the same amino acid sequence. In another instance, such a bispecific antibody comprises on its human CD3 binding arm a polypeptide having an amino acid sequence identical to SEQ ID NO: 20, a light chain of an amino acid sequence that is at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97% or at least 98% identical. In another instance, such a bispecific antibody comprises on its human CD3 binding arm a sequence having an amino acid sequence identical to SEQ ID NO: 20, and a light chain of the same amino acid sequence. In another instance, such a bispecific antibody comprises a heavy chain variable region having a sequence identical to SEQ id no: 20, and a light chain of the same amino acid sequence.

In one aspect, the bispecific antibodies of the present disclosure bind gp120 and human CD89 and can effect killing of HIV-1 infected cells. CD89 is an IgA receptor expressed primarily on neutrophils, and thus this bispecific antibody enhances recruitment of neutrophils to kill HIV-infected cells. In one instance, such bispecific antibodies that bind gp120 comprise SEQ ID NO:1, VH-CDR1, SEQ ID NO: 2, VH-CDR2 of SEQ ID NO:3, VH-CDR3 of SEQ ID NO:4, VL-CDR1 of SEQ ID NO:5 and the VL-CDR2 of SEQ ID NO:6 VL-CDR 3. In one instance, such a bispecific antibody that binds human CD89 comprises SEQ ID NO: 98, VH-CDR1 of SEQ ID NO: 99, VH-CDR2, SEQ id no: 100, VH-CDR3 of SEQ ID NO: 103, VL-CDR1, SEQ ID NO: 104 and VL-CDR2 of SEQ ID NO: 105, VL-CDR 3. In another instance, a bispecific antibody that binds gp120 and human CD89 comprises on its gp 120-binding arm the amino acid sequence of SEQ ID NO:1, VH-CDR1, SEQ ID NO: 2, VH-CDR2 of SEQ ID NO:3, VH-CDR3 of SEQ ID NO:4, VL-CDR1 of SEQ ID NO:5 and the VL-CDR2 of SEQ ID NO:6 VL-CDR 3; and comprises on its human CD89 binding arm: SEQ ID NO: 98, VH-CDR1 of SEQ ID NO: 99, VH-CDR2 of SEQ ID NO: 100, VH-CDR3 of SEQ ID NO: 103, VL-CDR1, SEQ ID NO: 104 and VL-CDR2 of SEQ ID NO: 105, VL-CDR 3.

In another instance, such a bispecific antibody comprises a peptide having a sequence identical to SEQ ID NO:7 VH of an amino acid sequence which is at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97% or at least 98% identical. In another instance, such a bispecific antibody comprises, in its gp120 binding arm, a sequence having an amino acid sequence identical to SEQ ID NO:7 VH of the same amino acid sequence. In another instance, such a bispecific antibody comprises a heavy chain variable region having a sequence identical to SEQ ID NO:7 VH of the same amino acid sequence. In another instance, such a bispecific antibody comprises a peptide having a sequence identical to SEQ ID NO: 8. 81, 82, 83 or 84 has an amino acid sequence that is at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97% or at least 98% identical to the VL. In another instance, such a bispecific antibody comprises in its gp120 binding arm an amino acid sequence having an amino acid sequence identical to SEQ ID NO: 8. 81, 82, 83 or 84, or a pharmaceutically acceptable salt thereof. In another instance, such a bispecific antibody comprises a heavy chain variable region having a sequence identical to SEQ ID NO: 8. 81, 82, 83 or 84, or a pharmaceutically acceptable salt thereof. In another instance, such a bispecific antibody comprises a peptide having a sequence identical to SEQ ID NO: 9, a heavy chain of an amino acid sequence that is at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97% or at least 98% identical. In another instance, such a bispecific antibody comprises, in its gp120 binding arm, a sequence having an amino acid sequence identical to SEQ ID NO: 9 the same amino acid sequence. In another instance, such a bispecific antibody comprises a heavy chain variable region having a sequence identical to SEQ ID NO: 9 the same amino acid sequence. In another instance, such a bispecific antibody comprises a heavy chain variable region having a sequence identical to seq id NO: 10. 40, 78, 79 or 80, at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97% or at least 98% identical. In another instance, such a bispecific antibody comprises in its gp120 binding arm an amino acid sequence having a sequence identical to SEQ ID NO: 10. 40, 78, 79 or 80, or a pharmaceutically acceptable salt thereof. In another instance, such a bispecific antibody comprises a heavy chain variable region having a sequence identical to SEQ ID NO: 10. 40, 78, 79 or 80, or a pharmaceutically acceptable salt thereof.

In another instance, such a bispecific antibody comprises on its human CD89 binding arm a polypeptide having an amino acid sequence identical to SEQ ID NO: 96 VH having an amino acid sequence at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97% or at least 98% identical. In another instance, such a bispecific antibody comprises on its human CD89 binding arm a sequence having an amino acid sequence identical to SEQ ID NO: 96 VH of the same amino acid sequence. In another instance, such a bispecific antibody comprises on its human CD89 binding arm a sequence having an amino acid sequence identical to SEQ ID NO: 96 VH of the same amino acid sequence. In another instance, such a bispecific antibody comprises on its human CD89 binding arm a polypeptide having an amino acid sequence identical to SEQ ID NO: 101 has an amino acid sequence that is at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, or at least 98% identical. In another instance, such a bispecific antibody comprises on its human CD89 binding arm a sequence having an amino acid sequence identical to SEQ ID NO: 101, VL of the same amino acid sequence. In another instance, such a bispecific antibody comprises a heavy chain variable region having a sequence identical to SEQ id no: 101, VL of the same amino acid sequence. In another instance, such a bispecific antibody comprises on its human CD89 binding arm a polypeptide having an amino acid sequence identical to SEQ ID NO: 97, at least 90%, 91%, 92%, 93%, 94%, 95% 96%, 97% or at least 98% identical to the amino acid sequence of the heavy chain. In another example, such a bispecific antibody comprises on its human CD89 binding arm a sequence having an amino acid sequence identical to SEQ ID NO: 97 of the same amino acid sequence. In another instance, such a bispecific antibody comprises on its human CD89 binding arm a sequence having an amino acid sequence identical to SEQ ID NO: 97 of the same amino acid sequence. In another instance, such a bispecific antibody comprises on its human CD89 binding arm a polypeptide having an amino acid sequence identical to SEQ ID NO: 102 have an amino acid sequence that is at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, or at least 98% identical. In another instance, such a bispecific antibody comprises on its human CD89 binding arm a sequence having an amino acid sequence identical to SEQ ID NO: 102, and a light chain of the same amino acid sequence. In another instance, such a bispecific antibody comprises on its human CD89 binding arm a sequence having an amino acid sequence identical to SEQ ID NO: 102, and a light chain of the same amino acid sequence.

In certain embodiments, the bispecific antibody has an Fc domain from a human IgG1 antibody with 0-10 amino acid substitutions therein. The Fc domain comprises one "branch" (hinge-CH 2-CH3) from one component of the bispecific antibody (e.g., gp 120-binding moiety) and another "branch" from a second component of the bispecific antibody (e.g., CD 3-or CD 89-binding moiety). The permutation may be in one or two "branches". In certain embodiments, the Fc domain has one or more (1, 2, 3, 4, or 5) of the following mutations (EU numbering) in one or both "branches": N297A or N297Q, L234F, L235E, D265A or P331S.

Bispecific antibodies that bind gp120 and CD3 or bind gp120 and CD89 as disclosed herein can be prepared by chemically linking two different monoclonal antibodies or by fusing two hybridoma cell lines to produce a hybrid hybridoma. Other bispecific antibody technology platforms that can be used include, for exampleKlamda antibodies, SMIP, DNL, Covx bodies, peptide antibodies, chain exchange engineered domain antibodies (SEEDbodes), dAbs, diabodies, Affibodies, Fynomes, KunitzDomains, TandAbs, nanobodies, Albu-dabs, DARTs, DVD-IG, scFv-Igs, SVD-Igs, dAb-Igs, Knobs-in-Holes, BiTe platforms,

Figure BDA0002328692870000781

A platform,

Figure BDA0002328692870000782

Andnon-limiting examples of bispecific formats that can be used to prepare the bispecific Antibodies disclosed herein are described in Del Bano et al, Antibodies, 5: 1 (2016); garber et al, Nature Reviews Drug Discovery, 13: 799 and 801 (2014).

In one embodiment, a bispecific antibody molecule of the present disclosure comprises a single antibody having two arms comprising different antigen-binding regions, one arm specific for a first antigen, such as gp120, and a second arm specific for a second antigen, such as human CD3 or human CD 89. In another embodiment, the bispecific antibody molecules of the present disclosure comprise a single antibody having one antigen-binding region or arm with specificity for a first antigen, e.g., gp120, and a second antigen-binding region or arm with specificity for a second antigen, e.g., human CD3 or human CD 89. In yet another embodiment, a bispecific antibody molecule of the present disclosure comprises a single chain antibody having a first specificity for a first antigen, such as gp120, and a second specificity for a second antigen, such as human CD3 or human CD89, e.g., via two scfvs linked in series by an additional peptide linker. In a further embodiment, bispecific antibody molecules of the present disclosure include double Variable Domain antibodies (DVD-Ig), in which each light and heavy chain comprises two Variable domains in tandem connected by a short peptide (Wu et al, Generation and Characterization of Dual Variable Domain Immunoglobulin (DVD-Ig)TM) Molecule, Antibody Engineering, Springer Berlin Heidelberg (2010)). At one endIn some embodiments, the bispecific antibody is a chemically linked bispecific (Fab')2 fragment. In other embodiments, the bispecific antibody comprises Tandab (i.e., a fusion of two single chain diabodies, thereby producing a tetravalent bispecific antibody with two binding sites for each target antigen). In certain embodiments, the bispecific antibody is a flexible body (flexobody), which is a combination of a scFv and a diabody, resulting in a multivalent molecule. In yet another embodiment, the bispecific antibody comprises a "docking and locking" molecule based on a "dimerization and docking domain" in Protein Kinase (Protein Kinase) a, which when applied to Fab, can produce a trivalent bispecific binding Protein consisting of two identical Fab fragments linked to different Fab fragments. In another instance, the bispecific antibody of the present disclosure comprises a "Scorpion molecule" (which comprises, e.g., two scfvs fused to the two ends of a human Fab arm. In yet another embodiment, a bispecific antibody of the present disclosure comprises a diabody.

Exemplary classes of bispecific antibodies include, but are not limited to, IgG-like molecules with complementary CH3 domains to promote heterodimerization; an IgG fusion molecule in which a full-length IgG antibody is fused to an additional Fab fragment or portion of a Fab fragment; an Fc fusion molecule in which a single chain Fv molecule or a stable diabody is fused to a heavy chain constant domain, Fc region, or portion thereof; a Fab fusion molecule in which different Fab fragments are fused together; a recombinant IgG-like dual targeting molecule, wherein the molecule is flanked by at least two Fab fragments or portions of Fab fragments of different antibodies; scFv-based and diabody-based and heavy chain antibodies (e.g., domain antibodies, nanobodies), wherein different single chain Fv molecules or different diabodies or different heavy chain antibodies (e.g., domain antibodies, nanobodies) are fused to each other or to another protein or carrier molecule.

Examples of Fab fusion bispecific antibodies include, but are not limited to, F (ab)2 (Metarex/AMGEN), Dual-Action or Bis-Fab (genentech), Dock-and-Lock (DNL) (ImmunoMedics), Bivalent bispecific (Biotecnol) and Fab-Fv (UCB-Celltech).

Examples of scFv-based, diabody-based, and domain antibodies include, but are not limited to, bispecific T cell adaptors (BITE) (Micromet), tandem diabodies (Tandab) (affected), parental and retargeting techniques (DART) (macrogenetics), single chain diabodies (Academic), TCR-like antibodies (AIT, ReceptorLogics), human serum albumin scFv fusions (Merrimack), and combody (epigen biotech), double targeting nanobodies (Ablynx), and double targeting heavy chain-only domain antibodies.

Duobodies

Bispecific antibodies of the disclosure can be those that bind gp120 and a second antigen (e.g., human CD3, e.g., human CD3 epsilon or human CD3 delta; human CD89)

Figure BDA0002328692870000791

Is a bispecific IgG1 antibody comprising a K409R mutation in the CH3 region of the constant region of one heavy chain and a mutation in the CH3 region of the constant region of the other heavy chain selected from the group consisting of F405L, F405A, F405D, F405E, F405H, F405I, F405K, F405M, F405N, F405Q, F405S, F405T, F405V, F405W and F405Y of the bispecific antibody. In a particular embodiment of the process of the present invention,

Figure BDA0002328692870000803

is a bispecific IgG1 antibody comprising a K409R mutation in the CH3 region of the constant region of one heavy chain and a F405L mutation in the CH3 region of the constant region of the other heavy chain of the bispecific antibody.

In certain embodiments, the first antigen binding domain that binds gp120 as described above comprises a human IgG1 heavy chain constant region comprising a mutation selected from the group consisting of F405L, F405A, F405D, F405E, F405H, F405I, F405K, F405M, F405N, F405Q, F405S, F405T, F405V, F405W, and F405Y, and the second antigen binding domain that binds human CD3 (or human CD89) as described above comprises a human IgG1 heavy chain constant region comprising a K409R mutation.

In one embodiment, the first antigen-binding domain that binds gp120 as described above comprises a human IgG1 heavy chain constant region comprising the F405L mutation, and the second antigen-binding domain that binds human CD3 as described above comprises a human IgG1 heavy chain constant region comprising the K409R mutation.

In other embodiments, the first antigen-binding domain that binds gp120 as described above comprises a human IgG1 heavy chain constant region comprising a K409R mutation, and the second antigen-binding domain that binds human CD3 (or human CD89) as described above comprises a human IgG1 heavy chain constant region comprising a mutation selected from the group consisting of F405L, F405A, F405D, F405E, F405H, F405I, F405K, F405M, F405N, F405Q, F405S, F405T, F405V, F405W, and F405Y.

In one embodiment, the first antigen-binding domain that binds gp120 as described above comprises a human IgG1 heavy chain constant region comprising a K409R mutation, and the second antigen-binding domain that binds human CD3 (or human CD89) as described above comprises a human IgG1 heavy chain constant region comprising an F405L mutation.

Figure BDA0002328692870000801

The CDRs of (a) and the VH and VL may be any of the anti-gp 120, anti-CD 3 or anti-CD 89 amino acid sequences described in detail above.

In one embodiment of the process of the present invention,

Figure BDA0002328692870000802

the gp 120-binding arm of (a) comprises SEQ ID NO:1, VH-CDR1, SEQ ID NO: 2, VH-CDR2 of SEQ ID NO:3, VH-CDR3 of SEQ ID NO:4, VL-CDR1 of SEQ ID NO:5 and the VL-CDR2 of SEQ ID NO:6 VL-CDR 3. In one embodiment of the process of the present invention,

Figure BDA0002328692870000811

the CD 3-binding arm of (a) comprises SEQ ID NO: 11, VH-CDR1 of SEQ ID NO:12, VH-CDR2 of SEQ ID NO: 13, VH-CDR3 of SEQ ID NO: 14, VL-CDR1 of SEQ ID NO: 15 and VL-CDR2 of SEQ ID NO: 16 VL-CDR 3. In another instance, binding gp120 and human CD3

Figure BDA0002328692870000812

On its gp120 binding armComprises the following steps: SEQ ID NO:1, VH-CDR1, SEQ id no: 2, VH-CDR2 of SEQ ID NO:3, VH-CDR3 of SEQ ID NO:4, VL-CDR1 of SEQ ID NO:5 and the VL-CDR2 of SEQ ID NO:6 VL-CDR 3; and comprises on its human CD3 binding arm: SEQ ID NO: 11, VH-CDR1 of SEQ id no:12, VH-CDR2 of SEQ ID NO: 13, VH-CDR3 of SEQ ID NO: 14, VL-CDR1 of SEQ ID NO: 15 and VL-CDR2 of SEQ ID NO: 16 VL-CDR 3.

In one embodiment of the process of the present invention,

Figure BDA0002328692870000813

the gp120 binding arm of (a) comprises SEQ ID NO:7 and SEQ ID NO: 8 VL. In one embodiment of the process of the present invention,

Figure BDA0002328692870000814

the gp120 binding arm of (a) comprises SEQ ID NO:7 and the VH of SEQ ID NO: VL of 81. In one embodiment of the process of the present invention,

Figure BDA0002328692870000815

the gp120 binding arm of (a) comprises SEQ ID NO:7 and the VH of SEQ ID NO: 82 VL. In one embodiment of the process of the present invention,

Figure BDA0002328692870000816

the gp120 binding arm of (a) comprises SEQ id no:7 and the VH of SEQ ID NO: 83 VL of seq id no. In one embodiment of the process of the present invention,

Figure BDA0002328692870000817

comprises seq id NO:7 and the VH of SEQ ID NO: 84 VL of the series. In one embodiment of the process of the present invention,

Figure BDA0002328692870000818

the CD 3-binding arm of (a) comprises SEQ ID NO: 17 and SEQ ID NO: 18 VL of. In one embodiment of the process of the present invention,

Figure BDA0002328692870000819

the CD89 binding arm of (a) comprises SEQ ID NO: VH of 96 and SEQ ID NO: 101 VL. In another instance, binding gp120 and human CD3

Figure BDA00023286928700008110

Comprising in its gp120 binding arm: SEQ ID NO:7 and the VH of SEQ ID NO: 8, and comprises in its human CD3 binding arm: SEQ ID NO: 17 and SEQ ID NO: 18 VL of. In another instance, binding gp120 and human CD89

Figure BDA00023286928700008111

Comprising in its gp120 binding arm: SEQ ID NO:7 and the VH of SEQ ID NO: 8, and comprises in its human CD89 binding arm: SEQ ID NO: VH of 96 and SEQ ID NO: 101 VL. In one instance, binding gp120 and human CD3

Figure BDA00023286928700008112

Comprising in its gp120 binding arm: SEQ ID NO:7 and the VH of SEQ ID NO: 81, and comprises in its human CD3 binding arm: SEQ ID NO: 17 and SEQ ID NO: 18 VL of. In one instance, binding gp120 and human CD89

Figure BDA0002328692870000821

Comprising in its gp120 binding arm: SEQ ID NO:7 and the VH of SEQ ID NO: 81, and comprises in its human CD3 binding arm: SEQ ID NO: VH of 96 and SEQ ID NO: 101 VL. In one instance, binding gp120 and human CD3

Figure BDA0002328692870000822

Comprising in its gp120 binding arm: SEQ ID NO:7 and the VH of SEQ ID NO: 82, and comprises in its human CD3 binding arm: SEQ ID NO: 17 and SEQ id no: 18 VL of. In one instance, binding gp120 and human CD89

Figure BDA0002328692870000823

Comprising in its gp120 binding arm: SEQ ID NO:7 and the VH of SEQ ID NO: 82, and comprises in its human CD3 binding arm: SEQ ID NO: VH of 96 and SEQ ID NO: 101 VL. In one instance, binding gp120 and human CD3

Figure BDA0002328692870000824

Comprising in its gp120 binding arm: SEQ ID NO:7 and the VH of SEQ ID NO: 83, and comprises in its human CD3 binding arm: SEQ ID NO: 17 and SEQ ID NO: 18 VL of. In one instance, binding gp120 and human CD89

Figure BDA0002328692870000825

Comprising in its gp120 binding arm: SEQ ID NO:7 and the VH of SEQ ID NO: 83, and comprises in its human CD89 binding arm: SEQ ID NO: VH of 96 and SEQ ID NO: 101 VL. In one instance, binding gp120 and human CD3

Figure BDA0002328692870000826

Comprising in its gp120 binding arm: SEQ ID NO:7 and the VH of SEQ ID NO: 84, and comprises in its human CD3 binding arm: SEQ ID NO: 17 and SEQ ID NO: 18 VL of. In one instance, binding gp120 and human CD89

Figure BDA0002328692870000827

Comprising in its gp120 binding arm: SEQ ID NO:7 and the VH of SEQ ID NO: 84, and comprises in its human CD89 binding arm: SEQ ID NO: VH of 96 and SEQ ID NO: 101 VL. In another embodiment, binding gp120 and human CD3Comprises the amino acid sequence of SEQ ID NO: 9 and the first heavy chain of SEQ ID NO: 10, a first light chain; and SEQ ID NO: 19 and the second heavy chain of SEQ ID NO: 20, a second light chain. In another embodiment, binding gp120 and human CD89

Figure BDA0002328692870000829

Comprises the amino acid sequence of SEQ ID NO: 9 and the first heavy chain of SEQ ID NO: 10, a first light chain; and SEQ ID NO: 97 and the second heavy chain of SEQ ID NO: 102, a second light chain. In another embodiment, binding gp120 and human CD3

Figure BDA00023286928700008210

ComprisesSEQ ID NO: 9 and the first heavy chain of SEQ ID NO: 40; and SEQ ID NO: 19 and the second heavy chain of SEQ ID NO: 20, a second light chain. In another embodiment, binding gp120 and human CD89

Figure BDA0002328692870000831

Comprises the amino acid sequence of SEQ ID NO: 9 and the first heavy chain of SEQ ID NO: 40; and SEQ ID NO: 97 and the second heavy chain of SEQ ID NO: 102, a second light chain. In another embodiment, binding gp120 and human CD3

Figure BDA0002328692870000832

Comprises the amino acid sequence of SEQ ID NO: 9 and the first heavy chain of SEQ ID NO: 78 with a first light chain; and SEQ ID NO: 19 and the second heavy chain of SEQ ID NO: 20, a second light chain. In another embodiment, binding gp120 and human CD89Comprises the amino acid sequence of SEQ ID NO: 9 and the first heavy chain of SEQ ID NO: 78 with a first light chain; and SEQ ID NO: 97 and the second heavy chain of SEQ ID NO: 102, a second light chain. In another embodiment, binding gp120 and human CD3

Figure BDA0002328692870000834

Comprises the amino acid sequence of SEQ ID NO: 9 and the first heavy chain of SEQ ID NO: 79 of a first light chain; and SEQ ID NO: 19 and the second heavy chain of SEQ ID NO: 20, a second light chain. In another embodiment, binding gp120 and human CD89

Figure BDA0002328692870000835

Comprises the amino acid sequence of SEQ ID NO: 9 and the first heavy chain of SEQ ID NO: 79 of a first light chain; and SEQ ID NO: 97 and the second heavy chain of SEQ ID NO: 102, a second light chain. In another embodiment, binding gp120 and human CD3

Figure BDA0002328692870000836

Comprises the amino acid sequence of SEQ ID NO: 9 and the first heavy chain of SEQ ID NO: 80; and SEQ ID NO: 19 and the second heavy chain of SEQ ID NO: 20 second ofThe light chain. In another embodiment, binding gp120 and human CD89

Figure BDA0002328692870000837

Comprises the amino acid sequence of SEQ ID NO: 9 and the first heavy chain of SEQ ID NO: 80; and SEQ ID NO: 97 and the second heavy chain of SEQ ID NO: 102, a second light chain. In one embodiment, anti-gp 120 x CD3

Figure BDA0002328692870000838

Or anti-gp 120 x CD89

Figure BDA0002328692870000839

Comprises an amino acid sequence that is at least 80%, 85%, 90%, 91%, 92%, 93%, 94% or 95%, 96%, 97%, 98%, 99% or 100% identical to one of the amino acid sequences set forth below, or identical to one of the amino acid sequences provided below except for 1 to 10 (i.e., 1,2, 3, 4, 5, 6,7, 8,9 or 10) amino acid substitutions:

Figure BDA00023286928700008310

Figure BDA0002328692870000841

Figure BDA0002328692870000851

Figure BDA0002328692870000863

in one embodiment, anti-gp 120 x CD3

Figure BDA0002328692870000861

Or anti-gp 120 x CD89

Figure BDA0002328692870000862

One of the heavy chains of (1) comprisesAn amino acid sequence that is at least 80%, 85%, 90%, 91%, 92%, 93%, 94%, or 95%, 96%, 97%, 98%, 99%, or 100% identical, or is identical (except for 1 to 10 (i.e., 1,2, 3, 4, 5, 6,7, 8,9, or 10) amino acid substitutions) to an amino acid sequence provided below:

Figure BDA0002328692870000871

Figure BDA0002328692870000881

Figure BDA00023286928700008915

in one embodiment of the process of the present invention,

Figure BDA0002328692870000891

comprises the amino acid sequence of SEQ ID NO: 64, and

Figure BDA0002328692870000892

comprises the amino acid sequence of SEQ ID NO: 74. In a further embodiment of the process of the present invention,comprises the amino acid sequence of SEQ ID NO: 65, and

Figure BDA0002328692870000894

comprises the amino acid sequence of SEQ ID NO: 75. In a further embodiment of the process of the present invention,

Figure BDA0002328692870000895

one ofThe heavy chain constant region comprises SEQ ID NO: 62, and

Figure BDA0002328692870000896

comprises the amino acid sequence of SEQ ID NO: 72, or a pharmaceutically acceptable salt thereof. In yet another embodiment of the present invention,

Figure BDA0002328692870000897

comprises the amino acid sequence of SEQ ID NO: 63, and

Figure BDA0002328692870000898

comprises the amino acid sequence of SEQ ID NO: 73, or a pharmaceutically acceptable salt thereof.

In one embodiment of the process of the present invention,

Figure BDA0002328692870000899

the heavy chain of the gp 120-binding arm of (a) has the amino acid sequence of SEQ ID NO: 9, andthe light chain of the gp120 binding arm of (a) has the amino acid sequence of SEQ ID NO: 10, or a pharmaceutically acceptable salt thereof. In a further embodiment of the process of the present invention,

Figure BDA00023286928700008911

the heavy chain of the gp 120-binding arm of (a) has the amino acid sequence of SEQ ID NO: 9, and

Figure BDA00023286928700008912

the light chain of the gp120 binding arm of (a) has the amino acid sequence of SEQ ID NO: 40, or a pharmaceutically acceptable salt thereof. In a further embodiment of the process of the present invention,

Figure BDA00023286928700008913

the heavy chain of the gp 120-binding arm of (a) has the amino acid sequence of SEQ ID NO: 9, and

Figure BDA00023286928700008914

the light chain of the gp120 binding arm of (a) has the amino acid sequence of SEQ ID NO: 78 toThe amino acid sequence of (a). In a further embodiment of the process of the present invention,the heavy chain of the gp 120-binding arm of (a) has the amino acid sequence of SEQ ID NO: 9, andthe light chain of the gp120 binding arm of (a) has the amino acid sequence of SEQ ID NO: 79, or a pharmaceutically acceptable salt thereof. In yet another embodiment of the present invention,the heavy chain of the gp 120-binding arm of (a) has the amino acid sequence of SEQ ID NO: 9, and

Figure BDA0002328692870000904

the light chain of the gp120 binding arm of (a) has the amino acid sequence of SEQ ID NO: 80, or a pharmaceutically acceptable salt thereof.

In one embodiment, gp 120X CD3

Figure BDA0002328692870000905

The heavy chain of the CD3 binding arm of (a) comprises SEQ id no: 19 or consists thereof, andthe light chain of the CD3 binding arm of (a) comprises SEQ ID NO: 20 or consists thereof.

In one embodiment, gp 120X CD89

Figure BDA0002328692870000907

The heavy chain of the CD89 binding arm of (a) comprises SEQ id no: 97 or consists thereof, and

Figure BDA0002328692870000908

the light chain of the CD3 binding arm of (a) comprises SEQ ID NO: 102 or consists thereof.

In one embodiment, gp 120X CD3

Figure BDA0002328692870000909

Comprising two arms, the first arm comprising a heavy chain and a light chain that bind gp120, wherein

Figure BDA00023286928700009010

The heavy chain of the gp 120-binding arm of (a) comprises a heavy chain identical to SEQ ID NO: 9, and the light chain of the gp120 binding arm comprises an amino acid sequence at least 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, or 98% identical to the amino acid sequence set forth in SEQ ID NO: 10. 40, 78, 79 or 80, is at least 80%, 81%, 82%, an amino acid sequence that is at least 80%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97% or 98% identical, and a second arm comprising heavy and light chains that bind CD3, wherein the second arm comprises a heavy chain and a light chain that bind CD3

Figure BDA00023286928700009011

The heavy chain of the CD3 binding arm of (a) comprises a heavy chain identical to SEQ ID NO: 19, and the light chain of the CD3 binding arm comprises an amino acid sequence at least 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, or 98% identical to the amino acid sequence set forth in SEQ ID NO: 20, or an amino acid sequence at least 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, or 98% identical to the amino acid sequence set forth in seq id no.

In one embodiment, gp 120X CD89

Figure BDA0002328692870000915

Comprising two arms, the first arm comprising a heavy chain and a light chain that bind gp120, wherein

Figure BDA0002328692870000916

The heavy chain of the gp 120-binding arm of (a) comprises a heavy chain identical to SEQ ID NO: 9 is at least 80%, 81%, 82%, 83%, 84%, 85%,86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97% or 98% identical to the amino acid sequence of SEQ ID NO: 10. 40, 78, 79 or 80, is at least 80%, 81%, 82%, an amino acid sequence that is at least 80%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97% or 98% identical, and a second arm comprising heavy and light chains that bind CD89, wherein the second arm comprises a heavy chain and a light chain that bind CD89The heavy chain of the CD89 binding arm of (a) comprises a heavy chain identical to SEQ ID NO: 97, and the light chain of a CD89 binding arm comprises an amino acid sequence at least 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, or 98% identical to the amino acid sequence set forth in SEQ ID NO: 102, or an amino acid sequence at least 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, or 98% identical thereto.

In a specific embodiment, gp 120X CD3

Figure BDA0002328692870000911

Comprising two arms, the first arm comprising a heavy chain and a light chain that bind gp120, wherein

Figure BDA0002328692870000912

The heavy chain of the gp 120-binding arm of (a) comprises SEQ ID NO: 9 and the light chain of the gp120 binding arm comprises the amino acid sequence set forth in SEQ ID NO: 10 and the second arm comprises a heavy chain and a light chain that bind CD3, wherein

Figure BDA0002328692870000913

The heavy chain of the CD3 binding arm of (a) comprises SEQ ID NO: 19, and the light chain of the CD3 binding arm comprises the amino acid sequence set forth in SEQ ID NO: 20, or a pharmaceutically acceptable salt thereof. In certain embodiments, can be

Figure BDA0002328692870000914

From 1 to 10 (i.e., 1,2, 3, 4, 5, 6,7, 8,9, or 10) amino acid substitutions are made in any component of one or both heavy chain constant regions of (e.g., CH1, hinge, CH2, CH 3). In certain instances, amino acid substitutions alter (e.g., decrease) effector function and/or increase half-life relative to an unaltered polypeptide. In certain embodiments, these substitutions may be conservative amino acid substitutions.

In a specific embodiment, gp 120X CD89

Figure BDA0002328692870000921

Comprising two arms, the first arm comprising a heavy chain and a light chain that bind gp120, wherein

Figure BDA0002328692870000922

The heavy chain of the gp 120-binding arm of (a) comprises SEQ ID NO: 9, and the light chain of the gp120 binding arm comprises the amino acid sequence set forth in SEQ ID NO: 10 and the second arm comprises a heavy chain and a light chain that bind CD89, wherein

Figure BDA0002328692870000923

The heavy chain of the CD89 binding arm of (a) comprises SEQ ID NO: 97, and the light chain of the CD89 binding arm comprises the amino acid sequence set forth in SEQ ID NO: 102, or a pharmaceutically acceptable salt thereof. In certain embodiments, can be

Figure BDA0002328692870000924

From 1 to 10 (i.e., 1,2, 3, 4, 5, 6,7, 8,9, or 10) amino acid substitutions are made in any component of one or both heavy chain constant regions of (e.g., CH1, hinge, CH2, CH 3). In certain instances, amino acid substitutions alter (e.g., decrease) effector function and/or increase half-life relative to an unaltered polypeptide. In certain embodiments, these substitutions may be conservative amino acid substitutions.

In another specific embodiment, gp 120X CD3

Figure BDA0002328692870000925

Comprising two arms, the first arm comprising a heavy chain and a light chain that bind gp120, wherein

Figure BDA0002328692870000926

The heavy chain of the gp 120-binding arm of (a) comprises SEQ ID NO: 9, and the light chain of the gp120 binding arm comprises the amino acid sequence set forth in SEQ ID NO: 40 and the second arm comprises a heavy chain and a light chain that bind CD3, wherein

Figure BDA0002328692870000927

The heavy chain of the CD3 binding arm of (a) comprises SEQ ID NO: 19, and the light chain of the CD3 binding arm comprises the amino acid sequence set forth in SEQ ID NO: 20, or a pharmaceutically acceptable salt thereof. In certain embodiments, can be

Figure BDA0002328692870000928

From 1 to 10 (i.e., 1,2, 3, 4, 5, 6,7, 8,9, or 10) amino acid substitutions are made in any component of one or both heavy chain constant regions of (e.g., CH1, hinge, CH2, CH 3). In certain instances, amino acid substitutions alter (e.g., decrease) effector function and/or increase half-life relative to an unaltered polypeptide. In certain embodiments, these substitutions may be conservative amino acid substitutions.

In another specific embodiment, gp 120X CD89

Figure BDA0002328692870000929

Comprising two arms, the first arm comprising a heavy chain and a light chain that bind gp120, whereinThe heavy chain of the gp 120-binding arm of (a) comprises SEQ ID NO: 9, and the light chain of the gp120 binding arm comprises the amino acid sequence set forth in SEQ ID NO: 40 and the second arm comprises a heavy chain and a light chain that bind CD89, whereinThe heavy chain of the CD89 binding arm of (a) comprises SEQ ID NO: 97, and the light chain of the CD89 binding arm comprises the amino acid sequence set forth in SEQ ID NO: 102, or a pharmaceutically acceptable salt thereof. In certain embodiments, can be

Figure BDA0002328692870000939

From 1 to 10 (i.e., 1,2, 3, 4, 5, 6,7, 8,9, or 10) amino acid substitutions are made in any component of one or both heavy chain constant regions of (e.g., CH1, hinge, CH2, CH 3). In certain instances, amino acid substitutions alter (e.g., decrease) effector function and/or increase half-life relative to an unaltered polypeptide. In certain embodiments, these substitutions may be conservative amino acid substitutions.

In another specific embodiment, gp 120X CD3

Figure BDA0002328692870000931

Comprising two arms, the first arm comprising a heavy chain and a light chain that bind gp120, wherein

Figure BDA0002328692870000932

The heavy chain of the gp 120-binding arm of (a) comprises SEQ ID NO: 9, and the light chain of the gp120 binding arm comprises the amino acid sequence set forth in SEQ ID NO: 78 and the second arm comprises a heavy chain and a light chain that bind CD3, wherein

Figure BDA0002328692870000933

The CD3 binding arm of (a) comprises SEQ ID NO: 19, and the light chain of the CD3 binding arm comprises the amino acid sequence set forth in SEQ ID NO: 20, or a pharmaceutically acceptable salt thereof. In certain embodiments, can beFrom 1 to 10 (i.e., 1,2, 3, 4, 5, 6,7, 8,9, or 10) amino acid substitutions are made in any component of one or both heavy chain constant regions of (e.g., CH1, hinge, CH2, CH 3). In certain instances, amino acid substitutions alter (e.g., decrease) effector function and/or increase half-life relative to an unaltered polypeptide. In certain embodimentsThese substitutions may be conservative amino acid substitutions.

In another specific embodiment, gp 120X CD89

Figure BDA0002328692870000935

Comprising two arms, the first arm comprising a heavy chain and a light chain that bind gp120, wherein

Figure BDA0002328692870000936

The heavy chain of the gp 120-binding arm of (a) comprises SEQ ID NO: 9, and the light chain of the gp120 binding arm comprises the amino acid sequence set forth in SEQ ID NO: 78 and the second arm comprises a heavy chain and a light chain that bind CD89, whereinThe heavy chain of the CD89 binding arm of (a) comprises SEQ ID NO: 97, and the light chain of the CD89 binding arm comprises the amino acid sequence set forth in SEQ ID NO: 102, or a pharmaceutically acceptable salt thereof. In certain embodiments, can be

Figure BDA0002328692870000938

From 1 to 10 (i.e., 1,2, 3, 4, 5, 6,7, 8,9, or 10) amino acid substitutions are made in any component of one or both heavy chain constant regions of (e.g., CH1, hinge, CH2, CH 3). In certain instances, amino acid substitutions alter (e.g., decrease) effector function and/or increase half-life relative to an unaltered polypeptide. In certain embodiments, these substitutions may be conservative amino acid substitutions.

In another specific embodiment, gp 120X CD3

Figure BDA0002328692870000941

Comprising two arms, the first arm comprising a heavy chain and a light chain that bind gp120, wherein

Figure BDA0002328692870000942

The heavy chain of the gp 120-binding arm of (a) comprises SEQ ID NO: 9, and the light chain of the gp120 binding arm comprises the amino acid sequence set forth in SEQ ID NO: 79, andthe second arm comprises a heavy chain and a light chain which bind CD3, wherein

Figure BDA0002328692870000943

The heavy chain of the CD3 binding arm of (a) comprises SEQ ID NO: 19, and the light chain of the CD3 binding arm comprises the amino acid sequence set forth in SEQ ID NO: 20, or a pharmaceutically acceptable salt thereof. In certain embodiments, can beFrom 1 to 10 (i.e., 1,2, 3, 4, 5, 6,7, 8,9, or 10) amino acid substitutions are made in any component of one or both heavy chain constant regions of (e.g., CH1, hinge, CH2, CH 3). In certain instances, amino acid substitutions alter (e.g., decrease) effector function and/or increase half-life relative to an unaltered polypeptide. In certain embodiments, these substitutions may be conservative amino acid substitutions.

In another specific embodiment, gp 120X CD89

Figure BDA0002328692870000945

Comprising two arms, the first arm comprising a heavy chain and a light chain that bind gp120, wherein

Figure BDA0002328692870000946

The heavy chain of the gp 120-binding arm of (a) comprises SEQ ID NO: 9, and the light chain of the gp120 binding arm comprises the amino acid sequence set forth in SEQ ID NO: 79 and the second arm comprises a heavy chain and a light chain that bind CD89, wherein

Figure BDA0002328692870000947

The heavy chain of the CD89 binding arm of (a) comprises SEQ ID NO: 97, and the light chain of the CD89 binding arm comprises the amino acid sequence set forth in SEQ ID NO: 102, or a pharmaceutically acceptable salt thereof. In certain embodiments, can be

Figure BDA0002328692870000948

From 1 to 10 (i.e., 1,2, 3, 4, 5) of any component of one or both heavy chain constant regions (e.g., CH1, hinge, CH2, CH3)6,7, 8,9, or 10) amino acid substitutions. In some cases, the amino acid substitution alters (e.g., decreases) effector function and/or increases half-life relative to an unaltered polypeptide. In certain embodiments, these substitutions may be conservative amino acid substitutions.

In another specific embodiment, gp 120X CD3

Figure BDA0002328692870000949

Comprising two arms, the first arm comprising a heavy chain and a light chain that bind gp120, wherein

Figure BDA00023286928700009410

The heavy chain of the gp 120-binding arm of (a) comprises SEQ ID NO: 9, and the light chain of the gp120 binding arm comprises the amino acid sequence set forth in SEQ ID NO: 80 and the second arm comprises a heavy chain and a light chain that bind CD3, whereinThe heavy chain of the CD3 binding arm of (a) comprises SEQ ID NO: 19, and the light chain of the CD3 binding arm comprises the amino acid sequence set forth in SEQ ID NO: 20, or a pharmaceutically acceptable salt thereof. In certain embodiments, can be

Figure BDA0002328692870000952

From 1 to 10 (i.e., 1,2, 3, 4, 5, 6,7, 8,9, or 10) amino acid substitutions are made in any component of one or both heavy chain constant regions of (e.g., CH1, hinge, CH2, CH 3). In certain instances, amino acid substitutions alter (e.g., decrease) effector function and/or increase half-life relative to an unaltered polypeptide. In certain embodiments, these substitutions may be conservative amino acid substitutions.

In another specific embodiment, gp 120X CD89

Figure BDA0002328692870000953

Comprising two arms, the first arm comprising a heavy chain and a light chain that bind gp120, wherein

Figure BDA0002328692870000954

The heavy chain of the gp 120-binding arm of (a) comprises SEQ ID NO: 9, and the light chain of the gp120 binding arm comprises the amino acid sequence set forth in SEQ ID NO: 80 and the second arm comprises a heavy chain and a light chain that bind CD89, wherein

Figure BDA0002328692870000955

The CD89 binding arm of (a) comprises SEQ ID NO: 97, and the light chain of the CD89 binding arm comprises the amino acid sequence set forth in SEQ ID NO: 102, or a pharmaceutically acceptable salt thereof. In certain embodiments, can be

Figure BDA0002328692870000956

From 1 to 10 (i.e., 1,2, 3, 4, 5, 6,7, 8,9, or 10) amino acid substitutions are made in any component of one or both heavy chain constant regions of (e.g., CH1, hinge, CH2, CH 3). In certain instances, amino acid substitutions alter (e.g., decrease) effector function and/or increase half-life relative to an unaltered polypeptide. In certain embodiments, these substitutions may be conservative amino acid substitutions.

In particular embodiments, the antibody is defucosylated. In some embodiments, the antibody comprises one or more tags. In certain embodiments, the one or more labels comprise an avidin label.

Fc modification

In certain embodiments, an antibody of the present disclosure (e.g.,

Figure BDA0002328692870000957

) One or more amino acid sequence modifications are included in the heavy chain constant region (Fc). In certain embodiments, an antibody of the present disclosure (e.g.,

Figure BDA0002328692870000961

) One or more amino acid sequence modifications are included in the heavy chain constant region (Fc). In certain embodiments, these modifications are increased as compared to the PGT-121LO6 antibodyThe stability of the modified antibody or the binding affinity is improved. In particular embodiments, these modifications increase the stability or improve binding affinity of the modified antibody compared to the PGT-121.60 antibody. In particular embodiments, certain of these modifications increase the half-life of the antibody. In certain embodiments, certain of these modifications reduce antibody effector function. In other embodiments, certain of these modifications reduce antibody effector function and increase the half-life of the antibody.

In certain embodiments, the one or more modifications are selected from the following Fc amino acid substitutions (EU numbering) or combinations thereof: L234F; L235E; G236A; S239D; F243L; D265E; D265A; S267E; H268F; R292P; N297Q; N297A; S298A; S324T; I332E; S239D; A330L; L234F; L235E; P331S; F243L; Y300L; V305I; P396L; S298A; E333A; K334A; E345R; L235V; F243L; R292P; Y300L; P396L; M428L; E430G; N434S; G236A, S267E, H268F, S324T and I332E; G236A, S239D, and I332E; S239D, a330L, I332E; L234F, L235E and P331S; F243L, R292P, Y300L, V305I and P396L; G236A, H268F, S324T and I332E; S239D, H268F, S324T and I332E; S298A, E333A and K334A; L235V, F243L, R292P, Y300L and P396L; S239D, I332E; S239D, S298A, and I332E; G236A, S239D, I332E, M428L and N434S; G236A, S239D, a330L, I332E, M428L and N434S; S239D, I332E, G236A and a 330L; M428L and N4343S; M428L, N434S; G236A, S239D, a330L and I332E; and G236A and I332E. In certain embodiments, the one or more modifications are selected from: N297A, D265A, L234F, L235E, N297Q and P331S. In certain embodiments, the one or more modifications are N297A or D265A. In certain embodiments, the one or more modifications are L234F and L235E. In certain embodiments, the one or more modifications are L234F, L234E, and D265A. In certain embodiments, the one or more modifications are L234F, L234E, and N297Q. In certain embodiments, the one or more modifications are L234F, L235E, and P331S. In certain embodiments, the one or more modifications are D265A and N297Q. In certain embodiments, the one or more modifications are L234F, L235E, D265A, N297Q and P331S.

Mutations that reduce Fc receptor binding include, for example, N297A; N297Q; D265A; L234F/L235E; L234F/L235E/N297Q; L234F/L235E/P331S; D265A/N297Q; and L234F/L235E/D265A/N297Q/P331S (all EU numbering). In certain embodiments, an antibody disclosed herein (e.g., Duobodies) comprises L234F and L235E mutations. In certain embodiments, an antibody disclosed herein (e.g., Duobodies) comprises L234F, L235E, and D265A mutations. In certain embodiments, an antibody disclosed herein (e.g., Duobodies) comprises L234F, L235E, and D265A mutations. In certain embodiments, an antibody disclosed herein (e.g., Duobodies) comprises a N297A or N297Q mutation. In certain embodiments, an antibody disclosed herein (e.g., Duobodies) comprises an N297A or N297Q mutation, as well as L234F, L235E, and D265A mutations. In certain embodiments, one, two, three, four, or more amino acid substitutions are introduced into the Fc region to alter the effector function of the antibody. For example, these substitutions are at a position selected from the group consisting of amino acid residues 234, 235, 236, 237, 265, 297, 318, 320 and 322 (according to EU numbering). These positions may be substituted with different amino acid residues to give the antibody an altered (e.g., reduced) affinity for an effector ligand (e.g., an Fc receptor or the C1 component of complement), but retain the antigen binding ability of the parent antibody. In certain embodiments, an antibody disclosed herein (e.g., Duobodies) comprises the E233P, L234V, L235A, and G236A mutations (EU numbering). In some embodiments, the antibody comprises a327G, a330S, and P331S mutations (EU numbering). In some embodiments, the antibody comprises the K322A mutation (EU numbering). In some embodiments, the antibody comprises E318A, K320A, and K322A (EU numbering) mutations. In certain embodiments, the antibody comprises an L235E (EU numbering) mutation.

Mutations that increase the half-life of an antibody are known in the art. In one embodiment, an antibody described herein (e.g.,

Figure BDA0002328692870000971

) The constant region of (A) comprises a methionine to tyrosine substitution at position 252 (EU numbering), a serine to threonine substitution at position 254 (EU numbering)And a threonine to glutamic acid substitution at position 256 (EU numbering). See, for example, U.S. patent No. 7,658,921. This type of mutant, referred to as the "YTE mutant", exhibits a four-fold increase in half-life relative to the wild-type form of the same antibody (Dall' Acqua et al, J Biol Chem, 281: 23514-24 (2006); Robbie et al, anti Agents Chemotherap, 57 (12): 6147-. In certain embodiments, the antibody comprises an IgG constant domain comprising amino acid substitutions of one, two, three, or more amino acid residues at positions 251, 257, 285, 290, 308, 314, 385, 389, and 428 (EU numbering). In other embodiments, the antibodies described herein (e.g.,

Figure BDA0002328692870000981

) Comprising substitutions M428L and N4343S (EU numbering). In other embodiments, the antibodies described herein (e.g.,

Figure BDA0002328692870000982

) Contains the T250Q and M428L (EU numbering) mutations. In other embodiments, the antibodies described herein (e.g.,) Comprising H433K and N434F (EU numbering) mutations.

In particular embodiments, an antibody (e.g.,

Figure BDA0002328692870000984

) Comprising two or more, three or more, four or more, five or more, six or less, five or less, four or less, three or less, two or less, or one modified Fc amino acid residue. In certain embodiments, the antibody comprises the L234F, L235E, D264A mutations collectively referred to as "FEA". In certain embodiments, the antibody comprises the L234F, L235E, D264A, and F405L mutations collectively referred to as "FEAL". In certain embodiments, the antibody comprises L234F, L235E, D264A, and is selected from F405L, F405A, F405D, F405E, F405H, F405I, F405K, F405M, F405N, F405Q, F405S, F405T,mutations of F405V, F405W and F405Y. In certain embodiments, the antibody comprises the L234F, L235E, D264A, and K409R mutations collectively referred to as "FEAR. In certain embodiments, the FEAL and FEAR comprise a bispecific antibody described herein (e.g.,) In (1). In certain embodiments, the antibody comprises M428L and N434S mutations, collectively referred to as LS. In certain embodiments, the antibody comprises L234F, L235E, D264A, F405L, M428L, and N434S mutations, collectively referred to as "FEALLS". In certain embodiments, the antibody comprises L234F, L235E, D264A, M428L, and N434S mutations and one additional mutation selected from F405L, F405A, F405D, F405E, F405H, F405I, F405K, F405M, F405N, F405Q, F405S, F405T, F405V, F405W, and F405Y. In certain embodiments, the antibody comprises L234F, L235E, D264A, K409R, M428L, and N434S mutations collectively referred to as "FEARLS". In certain embodiments, FEALLS and FEARLS comprise a bispecific antibody described herein (e.g.,) In (1). In certain embodiments, the antibody comprises S239D, I332E, G236A, a330L ("DEAL"). In certain embodiments, the antibody comprises the S239D, I332E, G236A, a330L, M428L, and N434S mutations ("DEALLS"). FEA mutations reduce or eliminate effector function, while DEAL mutations increase or enhance effector function by enhancing Fc binding to activated Fc γ R. The LS mutation increases the pharmacokinetic half-life of the antibody.

By reducing or eliminating effector function, the CD 3X gp120 multispecific/bispecific antibody (i) ensures that T cells bound by the bispecific molecule, including T cells not infected with HIV, are not killed by innate effector cells (e.g., NK cells, macrophages); and (ii) also ensures that T cells are not activated in the absence of target cells by not binding or reducing binding to Fc γ Rs on innate effector cells. Activation of T cells in the absence of target cells leads to a cytokine response and is not tolerated. Binding of bispecific molecules to Fc γ Rs on innate effector cells results in clustering of CD3 molecules on T cells, leading to antigen-independent T cell activation.

Conjugated antibodies

Any of the antibodies disclosed herein can be conjugated antibodies that bind to a variety of molecules, including macromolecular species such as polymers (e.g., polyethylene glycol (PEG), polyethyleneimine modified with PEG (PEI-PEG), polyglutamic acid (PGA) (N- (2-hydroxypropyl) methacrylamide (HPMA) copolymer), hyaluronic acid, radioactive species (e.g., a polymer such as polyethylene glycol (PEG), polyethylene imine modified with PEG (PEI-PEG), and poly (N- (2-hydroxypropyl) methacrylamide (HPMA))90Y、131I、125I、35S、3H、121In、99Tc), fluorescent substances (e.g., fluorescein and rhodamine), luminescent substances (e.g., luminol), haptens, enzymes (e.g., glucose oxidase), metal chelates, biotin, avidin, and drugs.

Such conjugated antibodies can be prepared by chemical modification of an antibody or lower molecular weight version thereof as described herein. Methods of modifying antibodies are well known in the art (e.g., US 5,057,313 and US 5,156,840).

Nucleic acids

The disclosure also features polynucleotides comprising nucleotide sequences encoding polynucleotide antibodies described herein that bind to gp120 and human CD3 antigens or bind to gp120 and CD89 antigens, vectors comprising such polynucleotides, and host cells (e.g., mammalian cells, yeast, e. Provided herein are polynucleotides comprising nucleotide sequences encoding any of the antibodies provided herein, as well as vectors comprising such polynucleotide sequences, e.g., expression vectors for efficient expression in a host cell, e.g., a mammalian cell.

In one aspect, the disclosure provides a polynucleotide comprising a nucleotide sequence encoding the VH, VL, or VH and VL of an antibody that binds gp120 (e.g., exemplary anti-gp 120 antibody 2; exemplary anti-gp 120 antibody 3; exemplary anti-gp 120 antibody 4; exemplary anti-gp 120 antibody 5).

In another aspect, the disclosure provides a polynucleotide comprising a nucleotide sequence encoding an antibody that binds gp120 and human CD3 polypeptides and comprises an amino acid sequence described herein.

In another aspect, the disclosure provides a polynucleotide comprising a nucleotide sequence encoding an antibody that binds to gp120 and a human CD89 polypeptide and comprises an amino acid sequence described herein.

In another aspect, the disclosure provides a polynucleotide or nucleic acid molecule encoding an antibody or antigen-binding fragment thereof according to the invention. In some embodiments, the nucleic acid molecule encodes an antibody light chain (or fragment thereof) or both of the present application. In other embodiments, the nucleic acid is DNA, cDNA, or mRNA. In some other embodiments, the nucleic acid molecule is codon optimized to enhance expression in the host cell.

In another aspect, provided herein is a polynucleotide comprising a nucleotide sequence encoding a CDR, light chain or heavy chain of an antibody described herein. The polynucleotide may comprise a nucleotide sequence encoding a light chain or light chain variable domain comprising the VL CDRs of the antibodies described herein (see, e.g., the tables above). The polynucleotide may comprise a nucleotide sequence encoding a heavy chain or a heavy chain variable domain comprising the VH CDRs of the antibodies described herein (see, e.g., the tables above). In one embodiment, the polynucleotides described herein encode a polypeptide having a sequence comprising SEQ ID NOs: 4. 5 and 6 or SEQ ID NO: 14. 15 and 16, or a variable light chain of the VL-CDR. In another embodiment, the polynucleotides described herein encode a polypeptide having a sequence comprising SEQ ID NOs: 1. 2 and 3 or SEQ ID NOs: 11. 12 and 13, or a variable heavy chain or heavy chain of a VH-CDR of the amino acid sequences set forth in seq id no. In one embodiment, the polynucleotide described herein encodes a polypeptide comprising SEQ ID NO: 8. 18 or 101, or a VL domain of an amino acid sequence set forth in seq id No. 18 or 101. In another embodiment, the polynucleotide described herein encodes a polypeptide comprising SEQ ID NO: 7. 17 or 96. In yet another embodiment, the polynucleotide described herein encodes a polypeptide comprising SEQ ID NO: 10. 20 or 102, or a light chain of an amino acid sequence as set forth in seq id no. In another embodiment, the polynucleotide described herein encodes a polypeptide comprising SEQ ID NO: 9. 19 or 97, or a pharmaceutically acceptable salt thereof.

The disclosure also includes polynucleotides encoding anti-gp 120 and anti-CD 3 (or anti-CD 89) antibodies optimized, for example, by codon optimization, substitution of heterologous signal sequences, and elimination of mRNA instability elements. Methods of producing optimized nucleic acids can be achieved by employing, for example, U.S. patent No.5,965,726; 6,174,666, respectively; 6,291,664, respectively; 6,414,132, respectively; and 6,794,498, respectively.

Vectors and host cells

The disclosure also includes vectors comprising the nucleic acids disclosed herein. The vector may be of any type, e.g., a recombinant vector, such as an expression vector. Vectors include, but are not limited to, plasmids, cosmids, Bacterial Artificial Chromosomes (BACs) and Yeast Artificial Chromosomes (YACs), as well as vectors derived from bacteriophages or plant or animal (including human) viruses. The vector may contain an origin of replication recognized by the proposed host cell, and in the case of an expression vector, a promoter and other regulatory regions that can be recognized by the host cell. In particular embodiments, the vector comprises a polynucleotide encoding an antibody of the present disclosure operably linked to a promoter and optionally additional regulatory elements. Certain vectors are capable of autonomous replication in a host into which they are introduced (e.g., vectors having a bacterial origin of replication may replicate in bacteria). Other vectors may be integrated into the genome of a host upon introduction into the host, and thereby replicated together with the host genome. Vectors include, but are not limited to, those suitable for recombinant production of the antibodies disclosed herein.

The choice of vector depends on the recombinant process followed and the host used.vectors can be introduced into host cells, inter alia, by calcium phosphate transfection, viral infection, DEAE-dextran mediated transfection, lipofectamine transfection or electroporation.

In a particular embodiment, the vector used is pcDNATM3.1+ (ThermoFisher,MA)。

The present disclosure also provides host cells comprising a nucleic acid or vector described herein. Any of a variety of host cells may be used. In one embodiment, the host cell is a prokaryotic cell, such as E.coli. In another embodiment, the host cell is a eukaryotic cell, e.g., a mammalian cell, such as a Chinese Hamster Ovary (CHO) cell, a COS cell, a BHK cell, an NSO cell, or a bowden melanoma cell. Examples of human host cells are inter alia HeLa, 911, AT1080, a549, 293 and HEK293T cells.

The term "nucleic acid molecule" refers to a polymeric form of nucleotides, and includes both sense and antisense strands of RNA, cDNA, genomic DNA, as well as synthetic forms and mixed polymers of the foregoing.

The term "operably linked" refers to two or more nucleic acid sequence elements that are typically physically linked and in a functional relationship with each other. For example, a promoter is operably linked to a coding sequence if it is capable of promoting or regulating the transcription or expression of the coding sequence, in which case the coding sequence is understood to be "under the control" of the promoter.

As used herein, "substitution" means the replacement of one or more amino acids or nucleotides with a different amino acid or nucleotide, respectively.

An "isolated" nucleic acid refers to a nucleic acid molecule that has been separated from components of its natural environment. An isolated nucleic acid includes a nucleic acid molecule contained in a cell that normally contains the nucleic acid molecule, but which is present extrachromosomally or at a chromosomal location different from its natural chromosomal location. "isolated nucleic acid encoding an antibody or fragment thereof" refers to one or more nucleic acid molecules encoding the heavy and light chains of an antibody (or fragment thereof), including such nucleic acid molecules in a single vector or separate vectors, as well as such nucleic acid molecules present at one or more locations in a host cell.

As used herein, the term "vector" refers to a nucleic acid molecule capable of propagating another nucleic acid to which it is linked. The term includes vectors which are self-replicating nucleic acid structures, as well as vectors which are incorporated into the genome of a host cell into which they are introduced. Some vectors are suitable for delivery of the nucleic acid molecules or polynucleotides of the present application. Certain vectors are capable of directing the expression of a nucleic acid to which they are operably linked. Such vectors are referred to herein as expression vectors.

The terms "host cell," "host cell line," and "host cell culture" are used interchangeably and refer to a cell into which an exogenous nucleic acid has been introduced, including the progeny of such a cell. Host cells include "transformants" and "transformed cells," which include the primary transformed cell and progeny derived therefrom (regardless of the number of passages). Progeny may not be identical in nucleic acid content to the parent cell, but may contain mutations. Progeny of mutants having the same function or biological activity as screened or selected in the originally transformed cell are included herein.

The term "variant" of a polynucleotide as used herein is a polynucleotide that differs from the polynucleotide specifically disclosed herein, typically in one or more substitutions, deletions, additions and/or insertions. Such variants may be naturally occurring, or may be synthetically produced, for example, by modifying one or more polynucleotide sequences of the invention and assessing one or more biological activities of the encoded polypeptide as described herein and/or using any of a variety of techniques well known in the art.

The term "variant" of a polypeptide as used herein is a polypeptide that differs from the polypeptide specifically disclosed herein, typically in one or more substitutions, deletions, additions and/or insertions. Such variants may be naturally occurring, or may be synthetically produced, for example, by modifying one or more of the above-described polypeptide sequences of the invention and assessing one or more biological activities of the polypeptide as described herein and/or using any of a variety of techniques well known in the art. In one embodiment, the antibody or antigen binding fragment thereof comprises variant 1, variant 2, variant 3, and/or variant 4. In some embodiments, the antibody or antigen-binding fragment thereof comprises variant 1. In some embodiments, the antibody or antigen-binding fragment thereof comprises variant 2. In some embodiments, the antibody or antigen-binding fragment thereof comprises variant 3. In some embodiments, the antibody or antigen-binding fragment thereof comprises variant 4.

The term "variant" may also refer to any naturally occurring or engineered molecule comprising one or more nucleotide or amino acid mutations. In one embodiment, the molecule is an antibody. For example, a somatic variant may include all relevant naturally occurring antibodies that are part of or derived from the same B cell lineage. Engineered variants may include all single mutations or combined mutations made to an antibody.

Method for producing antibody

Monospecific antibodies that bind gp120 and bispecific antibodies that bind gp120 and human CD3 (e.g., human CD3 epsilon or human CD3 delta) can be produced by any method known in the art for antibody synthesis, e.g., by chemical synthesis or by recombinant expression techniques.

Methods for making monospecific antibodies are well known in the art. Methods for making bispecific antibodies are described, for example, in U.S. Pat. nos. 5,731,168; 5,807,706, respectively; 5,821,333, respectively; and U.S. application publication nos. 2003/020734 and 2002/0155537. Bispecific tetravalent antibodies and methods for making them are described, for example, in WO 02/096948 and WO 00/44788, the disclosures of both of which are incorporated herein by reference in their entirety. In addition, other publications relevant to the preparation of bispecific antibodies include WO 91/00360, WO 92/08802, WO92/05793 and WO 93/17715; tutt et al, j.immunol.147: 60-69 (1991); U.S. patent nos. 4,474,893; 4,714,681, respectively; 4,925,648; 5,573,920, respectively; 5,601,819 and 9,212,230; and Kostelny et al, j.immunol.148: 1547-1553(1992).

In one embodiment, the bispecific antibody of the present disclosure isDuobodies can be identified by, for example, those described in International publications Nos. WO 2008/119353, WO 2011/131746, WO 2011/147986 and WO 2013/060867, Labrijn AF et al PNAS,110(13):5145-

Figure BDA0002328692870001052

Preparation of technical platform (Genmab A/S). This technique can be used to bind half of a first monospecific antibody comprising two heavy chains and two light chains to half of a second monospecific antibody comprising two heavy chains and two light chains. The resulting heterodimer comprises one heavy chain and one light chain from the first antibody paired with one heavy chain and one light chain from the second antibody. When two monospecific antibodies recognize different epitopes on different antigens, the resulting heterodimer is a bispecific antibody.

For thePlatform, each monospecific antibody includes a heavy chain constant region with a single point mutation in the CH3 domain. These point mutations allow for stronger interactions between the CH3 domains in the resulting bispecific antibody than between the CH3 domains in any monospecific antibody without the mutation. A single point mutation in each monospecific antibody may be at residue 366, 368, 370, 399, 405, 407 or 409(EU numbering) in the CH3 domain of the heavy chain constant region (see WO 2011/131746). Furthermore, a single point mutation is located at a different residue in one monospecific antibody relative to another monospecific antibody. For example, one monospecific antibody may comprise the mutation F405L (EU numbering; phenylalanine to leucine mutation at residue 405), or one of the F405A, F405D, F405E, F405H, F405I, F405K, F405M, F405N, F405Q, F405S, F405T, F405V, F405W, and F405Y mutations, while another monospecific antibody may comprise the mutation K409R (EU numbering; lysine to arginine mutation at residue 409). The heavy chain constant region of a monospecific antibody may be of the IgG1, IgG2, IgG3 or IgG4 isotype (e.g., human IgG1 isotype) and is encoded by

Figure BDA0002328692870001061

Bispecific antibodies produced by the technology can be modified to alter (e.g., reduce) Fc-mediated effector function and/or improve half-life. Preparation method

Figure BDA0002328692870001062

The method comprises the following steps: (i) separately expressing two parent IgG1 comprising a single matched point mutation in the CH3 domain (i.e., K409R and F405L (or F405A, F405D, F405E, F405H, F405I, F405K, F405M, F405N, F405Q, F405S, F405T, F405V, F405W, and F405Y mutations) (EU numbering)); (ii) in vitro blending of parental IgG1 under permissive redox conditions to allow half-molecule recombination; (iii) removing the reducing agent to reoxidize the interchain disulfide bonds; and (iv) analysis of exchange efficiency and final product using chromatography-based or Mass Spectrometry (MS) -based methods (see Labrijn et al, Nature Protocols,9(10): 2450-.

Another exemplary method for making bispecific antibodies is by the knob-and-hole technology (Ridgway et al, Protein Eng., 9: 617-621 (1996); WO 2006/028936). The major drawback in making bispecific antibodies is the reduction of the mismatch problem of Ig heavy chains by mutating selected amino acids that form the interface of the CH3 domain in IgG. At the position within the CH3 domain where the two heavy chains interact directly, an amino acid with a small side chain (hole) is introduced into the sequence of one heavy chain, while an amino acid with a large side chain (knob) is introduced into the corresponding interaction residue position of the other heavy chain. In some cases, the antibodies of the present disclosure have immunoglobulin chains in which the CH3 domain is modified by mutation of selected amino acids that interact at the interface between the two polypeptides, thereby preferentially forming bispecific antibodies. Bispecific antibodies can be composed of immunoglobulin chains of the same subclass or of different subclasses. In one instance, bispecific antibodies that bind gp120 and CD3 contain a T366W (EU numbering) mutation in the "knob chain" and a T366S, L368A, Y407V (EU numbering) mutation in the "hole chain". In certain embodiments, additional interchain disulfide bridges are introduced between the CH3 domains, for example, by introducing the Y349C mutation into the "knob chain" and the E356C mutation or S354C mutation into the "hole chain". In certain embodiments, the R409D, K370E mutations are introduced in the "pestle chain" and the D399K, E357K mutations are introduced in the "mortar chain". In other embodiments, the Y349C, T366W mutations were introduced into one strand, and the E356C, T366S, L368A, Y407V mutations were introduced into the corresponding strand. In some embodiments, the Y349C, T366W mutations are introduced into one chain, and the S354C, T366S, L368A, Y407V mutations are introduced into the corresponding chain. In some embodiments, the Y349C, T366W mutations are introduced into one chain, while the S354C, T366S, L368A, Y407V mutations are introduced into the corresponding chain. In yet other embodiments, the Y349C, T366W mutations are introduced in one chain and the S354C, T366S, L368A, Y407V mutations are introduced in the corresponding chain (all EU numbering).

Another exemplary method of making bispecific antibodies is by using bispecific T cell adaptors

Figure BDA0002328692870001071

A platform. BiTE is made by genetically fusing a first scFv (e.g., an scFv that binds gp120) to a second scFv (e.g., an scFv that binds human CD3) via a flexible peptide linker (e.g., GGGGS (SEQ ID NO: 76)). See, e.g., Staerz et al, Nature, 314: 628-631 (1985); mack et al, PNAS, 92: 7021-7025 (1995); huehls et al, immunol. cell biol., 93: 290-296(2015).

Another exemplary method of making bispecific antibodies is by using a Dual Affinity Retargeting (DART) platform. This technique is based on a diabody version of Holliger et al (PNAS,90: 6444-.

Yet another exemplary method for making bispecific antibodies is through the use of a trifunctional hybrid antibody platform

Figure BDA0002328692870001072

The platform employs a chimeric structure consisting of half of full-length antibodies of two different isotypes (mouse IgG2a and rat IgG2 b). This technique relies on species-preferred heavy/light chain pairing. See Lindhofer et al, jimmunol, 155: 219-225 (1995).

Preparation of bispecificA further exemplary method of antibody is through the use of

Figure BDA0002328692870001073

A platform. This technology is based on a diabody concept, but is designed as a single polypeptide chain comprising a short linker, VH1-VL2-VH2-VL1, to prevent intra-chain pairing. The head-to-tail dimerization of this single chain results in the formation of a tetravalent homodimer (Kipriyanov et al, J mol. biol., 293: 41-56 (1999)).

Yet another method for making bispecific antibodies is CrossMab technology. CrossMab is a chimeric antibody composed of half of two full-length antibodies. For proper strand pairing, it combines two techniques: (i) a knob and mortar structure which facilitates correct pairing between the two heavy chains; and (ii) exchange between the heavy and light chains of one of the two fabs to introduce an asymmetry that avoids light chain mispairing. See, Ridgway et al, Protein eng, 9: 617 + 621 (1996); schaefer et al, PNAS, 108: 11187-11192(2011). CrossMab can combine two or more antigen binding domains to target two or more targets or to introduce a bivalent to one target, e.g., 2:1 format.

Multispecific antibodies of the present disclosure may be produced in bacteria or eukaryotic cells. Antibodies can also be produced in eukaryotic cells, such as transformed cell lines (e.g., CHO, 293E, 293T, COS, NIH3T 3). In addition, antibodies (e.g., scFv) can be expressed in yeast cells such as Pichia pastoris (see, e.g., Powers et al, J Immunol methods.251: 123-35(2001)), Hansenula, or Saccharomyces. In one embodiment, the bispecific antibody described herein is produced in a CHO cell line. To produce the antibody of interest, a polynucleotide encoding the antibody is constructed, introduced into an expression vector, and then expressed in a suitable host cell. Standard molecular biology techniques are used to prepare recombinant expression vectors, transfect host cells, select transformants, culture the host cells and recover the antibodies.

In addition, when Escherichia coli such as JM109, DH5 α, HB101 or XL1-Blue is used as a host, the vector must have a promoter which can be efficiently expressed in Escherichia coli, such as lacZ promoter (Ward et al, 341: 544-546 (1989)), araB promoter (Better et al, Science, 240: 1041-1043(1988)), or T7 promoter.examples of such vectors include, for example, M13-series vectors, pUC-series vectors, pBR322, pBluescript, pCR-Script, pGEX-5X-1 (Pharmax), "Aexpress system" (QIAGAGEN), pEGFP and pET (when using the expression vector, the host is preferably BL21 expressing T7 RNA polymerase). the expression vector may contain a signal sequence for antibody secretion in order to generate a signal sequence for the expression of Escherichia coli, the bacterial cell expression vector may be used as a cell-producing signal sequence (Leu) for the expression of Bacillus coli strain 4379, and the expression vector may be used as a cell-secreting signal sequence for the E.

If the antibody is expressed in animal cells such as CHO, COS and NIH3T3 cells, the expression vector includes a promoter required for expression in these cells, such as the SV40 promoter (Mullingan et al, Nature, 277: 108(1979)), MMLV-LTR promoter, EF1 α promoter (Mizushima et al, Nucleic Acids Res., 18: 5322(1990)) or CMV promoter in addition to the Nucleic acid sequence encoding the immunoglobulin or a domain thereof, the recombinant expression vector may carry additional sequences such as a sequence regulating replication of the vector in the host cell (e.g., origin of replication) and a selection marker gene which facilitates selection of the host cell into which the vector has been introduced (see, for example, U.S. Pat. Nos. 4,399,216, 4,634,665 and 5,179,017.) examples of vectors having selection markers generally include pBAM, CMV-2, CMV-pBAM, PCR-2, and OPAM-pBAM 13.

In one embodiment, the antibody is produced in a mammalian cell. Exemplary mammalian host cells for expression of antibodies include Chinese Hamster Ovary (CHO) cells (including DHFR-CHO cells, as described in Urlaub and Chasin (1980) Proc. Natl. Acad. Sci. USA 77: 4216-. For example, the cell is a mammary epithelial cell.

In an exemplary system for antibody expression, recombinant expression vectors encoding the antibody heavy and light chains of bispecific antibodies of the disclosure are introduced into dhfr by calcium phosphate-mediated transfection-In CHO cells. In a specific embodiment, the dhfr-CHO cell is a cell of the DG44 cell line, such as DG44i (see, e.g., Derouaz et al, Biochem BiophyS Res Commun, 340 (4): 1069-77 (2006)). In recombinant expression vectors, the antibody heavy and light chain genes are each operably linked to enhancer/promoter regulatory elements (e.g., derived from SV40, CMV, adenovirus, etc., such as CMV enhancer/AdMLP promoter regulatory element or SV40 enhancer/AdMLP promoter regulatory element) to drive high levels of transcription of the genes. The recombinant expression vector also carries the DHFR gene, which allows the use of methotrexate selection/amplification for selection of CHO cells transfected with the vector. The selected transformant host cells are cultured to allow expression of the heavy and light chains of the antibody, and the antibody is recovered from the culture medium.

Multispecific antibodies can also be produced by transgenic animals. For example, U.S. patent No.5,849,992 describes a method of expressing an antibody in the mammary gland of a transgenic mammal. A transgene comprising a milk-specific promoter and nucleic acid encoding the antibody of interest and a signal sequence for secretion is constructed. The milk produced by the female of such a transgenic mammal includes the antibody of interest secreted therein. The antibody may be purified from milk or, for some applications, used directly. Also provided are animals comprising one or more of the nucleic acids described herein.

Multispecific antibodies of the present disclosure can be isolated from the interior or exterior (e.g., culture medium) of a host cell and purified as substantially pure and homogeneous antibodies. The method of separation and purification generally used for antibody purification may be used for separation and purification of antibodies, and is not limited to any particular method. The antibody can be isolated and purified by appropriately selecting and combining, for example, column chromatography, filtration, ultrafiltration, salting out, solvent precipitation, solvent extraction, distillation, immunoprecipitation, SDS-polyacrylamide gel electrophoresis, isoelectric focusing, dialysis, and recrystallization. Chromatography includes, for example, affinity chromatography, ion exchange chromatography, hydrophobic chromatography, gel filtration, reverse phase chromatography and adsorption chromatography (stratgies for Protein Purification and chromatography: analytical Course Manual. Ed Daniel R. Marshak et al, Cold Spring Harbor laboratory Press, 1996). The chromatographic analysis can be performed using liquid chromatography such as HPLC and FPLC. Columns for affinity chromatography include protein a columns and protein G columns. Examples of chromatography columns using protein A columns include Hyper D, POROS and Sepharose FF (GEHealthcare Biosciences). The present disclosure also includes antibodies that are highly purified using these purification methods.

Pharmaceutical composition

The disclosure also includes pharmaceutical compositions comprising an antibody described herein or a polynucleotide encoding an antibody described herein and a pharmaceutically acceptable diluent, carrier, or excipient. In certain embodiments, the pharmaceutical composition comprises a therapeutically effective amount of an antibody or polynucleotide.

In light of this disclosure, those skilled in the art are aware of various pharmaceutically acceptable diluents, carriers, and excipients, as well as techniques for preparing and using pharmaceutical compositions. Illustrative pharmaceutical compositions and pharmaceutically acceptable diluents, carriers and excipients are also described in Remington: The Science and Practice of Pharmacy 20th Ed. (Lippincott, Williams & Wilkins 2003). In particular embodiments, each carrier, diluent or excipient is "acceptable" in the sense of being compatible with the other ingredients of the pharmaceutical composition and not injurious to the subject. Typically, the pharmaceutically acceptable carrier is a pH buffered aqueous solution. Some examples of materials that may be used as pharmaceutically acceptable carriers, diluents or excipients include: sterile water; buffers, for example, phosphate buffered saline; sugars such as lactose, glucose and sucrose; starches, such as corn starch and potato starch; cellulose and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; radix astragali powder; malt; gelatin; talc; excipients, such as cocoa butter and suppository waxes; oils such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; glycols, such as propylene glycol; polyols such as glycerol, sorbitol, mannitol and polyethylene glycol; esters such as ethyl oleate and ethyl laurate; agar; buffering agents such as magnesium hydroxide and aluminum hydroxide; alginic acid; pyrogen-free water; isotonic saline; ringer's solution; ethanol; a phosphate buffer solution; and other non-toxic compatible substances used in pharmaceutical formulations. Wetting, emulsifying and lubricating agents, such as sodium lauryl sulfate and magnesium stearate, as well as coloring, mold release, coating, sweetening, flavoring and perfuming agents, preservatives and antioxidants can also be present in the composition.

The methods of formulation and delivery of the pharmaceutical compositions are generally tailored to the site and disease being treated. Exemplary formulations include, but are not limited to, those suitable for parenteral administration, such as intravenous, intraarterial, intramuscular, or subcutaneous administration, including formulations encapsulated in micelles, liposomes, or drug release capsules (with the active agent incorporated into a biocompatible coating designed for sustained release); an ingestible formulation; formulations for topical use, such as creams, ointments and gels; and other formulations such as inhalants, aerosols and sprays.

Method of treatment

The present disclosure provides methods for treating or preventing HIV infection or a related disease or disorder in a subject in need thereof (e.g., a human subject), comprising providing to a subject in need thereof an effective amount of an antibody or a polynucleotide encoding an antibody described herein. As used herein, the term "effective amount" in the context of administering a therapy to a subject refers to the amount of the therapy that achieves the desired prophylactic or therapeutic effect. The polynucleotide may be present in a vector, such as a viral vector. In a particular embodiment, the associated disease or condition is caused by HIV infection. In a particular embodiment, it is acquired immunodeficiency syndrome (AIDS). In particular embodiments, the subject is virally inhibited HIV infectionA dyed mammal, while in other embodiments, the subject is an untreated HIV-infected mammal. In certain embodiments, the viral load of the untreated subject is at 103To 105Between copies/ml, and in certain embodiments, the virologically suppressed subject has a viral load < 50 copies/ml. In particular embodiments, the subject is a mammal, e.g., a human. In certain embodiments, the subject is diagnosed with, or is considered at risk for developing, an HIV (such as HIV-1 or HIV-2) infection or associated disease or condition (such as AIDS). Subjects at risk for an HIV-associated disease or disorder include patients who have been in contact with or otherwise exposed to HIV from an infected person. Administration of the prophylactic agent can be carried out prior to the onset of symptoms characteristic of the HIV-associated disease or disorder, such that the disease or disorder is prevented or its development is optionally delayed.

Also provided are methods for preventing or inhibiting HIV viral titer, viral replication, viral propagation, or an increase in the amount of HIV viral DNA, HIV proviral DNA, or HIV viral protein in a subject (e.g., a human subject). In one embodiment, the method comprises providing to a subject in need thereof an antibody described herein or a polynucleotide encoding the antibody effective to prevent an increase in HIV titer, viral replication, or HIV protein amount of one or more HIV strains or isolates in the subject. In certain embodiments, the method further comprises measuring the amount of HIV viral or proviral DNA or protein at one or more time points before and after the subject is provided with the antibodies of the present disclosure. Methods and biomarkers for determining the amount of HIV viral or proviral DNA or protein in a subject are known in the art and are available and are described, for example, in silicano, j.d. et al, Curr Opin HIV AIDS,5(6):491-7(2010) and Rouzioux, c. et al, Curr Opin HIV AIDS,8(3):170-5 (2013).

As described herein

Figure BDA0002328692870001121

CD4 isolatable from patients with combined antiretroviral therapy (cART) inhibition+T is thinThe cells reactivate latent HIV ex vivo and thus can actually increase the titer of the HIV virus. This is that

Figure BDA0002328692870001122

Because it can activate latently infected cells to express gp120 and then potentially be targeted for clearance. Binding to CD3 induces a partial T cell activation phenotype, whereas latently infected CD4+Activation of T cells then leads to viral expression. Thus, also featured are methods of reversing HIV latency in a subject in need thereof. The method comprises administering gp120X CD3 as described herein to a human subject in need thereof

Figure BDA0002328692870001131

In certain embodiments, the method further comprises, e.g., in providing the subject with a composition of the present disclosure

Figure BDA0002328692870001132

The amount of HIV RNA, viral or proviral DNA or protein is measured at one or more time points before and after.

In certain aspects, the antibodies of the present disclosure may be used, for example, in methods of inhibiting certain viruses, such as HIV isolates described herein, prophylactically inhibiting or preventing infection of certain viruses, such as HIV isolates described herein, detecting certain viruses in a sample, such as HIV isolates described herein, inhibiting certain viruses, such as HIV isolates described herein, diagnosing certain viruses, such as HIV isolates described herein.

For in vivo treatment of a mammalian subject, e.g., a human, the subject may be administered or provided with a pharmaceutical composition comprising a multispecific antibody described herein. When used for in vivo therapy, the antibodies described herein are typically administered or provided to a patient in a therapeutically effective amount (i.e., to eliminate or reduce the viral load and/or viral pool of the patient). The antibody is administered or provided to a mammalian subject, e.g., a human, according to known methods, such as, but not limited to, intravenous administration (e.g., bolus injection or by continuous infusion over a period of time), by intramuscular, intraperitoneal, intracerobrospinal, subcutaneous, intraarticular, intrasynovial, intrathecal, oral, topical, or inhalation routes. The antibody may be administered parenterally or intravenously at the site of the target cell where possible. In one embodiment, the antibody is administered to the subject by an intravenous route. In another embodiment, the antibody is administered to the subject by a subcutaneous route. In particular embodiments, the pharmaceutical compositions of the present disclosure are administered to a subject systemically, parenterally, or topically.

In certain embodiments, the present disclosure provides a method for treating HIV infection comprising administering to a patient in need thereof a therapeutically effective amount of an antibody disclosed herein.

Combination therapy

In certain embodiments, a method is provided for treating or preventing HIV infection in a human having or at risk of having the infection, the method comprising administering to the human a therapeutically effective amount of an antibody disclosed herein, or a pharmaceutical composition thereof, in combination with a therapeutically effective amount of one or more (e.g., one, two, three, one or two, or one to three) other therapeutic agents. In one embodiment, a method is provided for treating an HIV infection in a human having or at risk of having the infection, comprising administering to the human a therapeutically effective amount of an antibody disclosed herein, or a pharmaceutically acceptable salt thereof, in combination with an effective amount of one or more (e.g., one, two, three, one or two, or one to three) other therapeutic agents.

In one embodiment, a pharmaceutical composition is provided comprising an antibody disclosed herein, or a pharmaceutical composition thereof, in combination with one or more (e.g., one, two, three, one or two, or one to three) additional therapeutic agents, and a pharmaceutically acceptable carrier, diluent, or excipient.

In certain embodiments, the methods provided by the present disclosure for treating HIV infection comprise administering to a patient in need thereof a therapeutically effective amount of an antibody disclosed herein, or a pharmaceutical composition thereof, in combination with a therapeutically effective amount of one or more other therapeutic agents useful for treating HIV infection.

In certain embodiments, the antibodies disclosed herein or pharmaceutical compositions thereof are combined with one, two, three, four, or more additional therapeutic agents. In certain embodiments, the antibodies disclosed herein or pharmaceutical compositions thereof are combined with two additional therapeutic agents. In other embodiments, the antibodies disclosed herein or pharmaceutical compositions thereof are combined with three additional therapeutic agents. In further embodiments, the antibodies disclosed herein or pharmaceutical compositions thereof are combined with four additional therapeutic agents. The one, two, three, four or more additional therapeutic agents may be different therapeutic agents selected from the same therapeutic agent class, and/or they may be selected from different therapeutic agent classes.

In certain embodiments, the antibodies disclosed herein are administered with one or more additional therapeutic agents. Co-administration of an antibody disclosed herein with one or more additional therapeutic agents generally refers to the simultaneous or sequential administration of a compound disclosed herein and one or more additional therapeutic agents such that a therapeutically effective amount of both the antibody disclosed herein and the one or more additional therapeutic agents are present in the patient. When administered sequentially, the combination may be administered two or more times.

Co-administration includes administering a unit dose of an antibody disclosed herein before or after administration of a unit dose of one or more other therapeutic agents. For example, the antibodies disclosed herein can be administered within seconds, minutes, or hours of administration of the one or more additional therapeutic agents. In some embodiments, a unit dose of an antibody disclosed herein is administered first, followed by a unit dose of one or more additional therapeutic agents within seconds or minutes. Alternatively, a unit dose of one or more additional therapeutic agents is administered first, followed by administration of a unit dose of an antibody disclosed herein within seconds or minutes. In other embodiments, a unit dose of an antibody disclosed herein is administered first, followed by administration of a unit dose of one or more additional therapeutic agents several hours (e.g., 1-12 hours) later. In still other embodiments, a unit dose of one or more additional therapeutic agents is administered first, followed by administration of a unit dose of an antibody disclosed herein after a few hours (e.g., 1-12 hours).

In certain embodiments, the antibodies disclosed herein are combined with one or more additional therapeutic agents in a single dosage form for simultaneous administration to a patient, e.g., as a solid dosage form for oral administration.

In certain embodiments, the antibodies of the present disclosure are formulated as liquids, which may optionally comprise additional therapeutic agents for the treatment of HIV. In certain embodiments, the liquid may comprise another active ingredient for the treatment of HIV, such as HIV protease inhibitors, HIV non-nucleoside or non-nucleotide reverse transcriptase inhibitors, HIV nucleoside or nucleotide reverse transcriptase inhibitors, HIV integrase inhibitors, HIV non-catalytic site (or allosteric) integrase inhibitors, pharmacokinetic enhancers, and combinations thereof.

In certain embodiments, such formulations are suitable for once-a-day administration. In the above embodiments, the additional therapeutic agent may be an anti-HIV agent. In some cases, the additional therapeutic agent may be an HIV protease inhibitor, an HIV non-nucleoside or non-nucleotide reverse transcriptase inhibitor, an HIV nucleoside or nucleotide reverse transcriptase inhibitor, an HIV integrase inhibitor, an HIV non-catalytic site (or allosteric) integrase inhibitor, an HIV entry inhibitor, an HIV maturation inhibitor, a latency reversal agent, a compound targeting the HIV capsid, an immune-based therapy, a phosphatidylinositol 3-kinase (PI3K) inhibitor, an HIV antibody, a bispecific antibody and "antibody-like" therapeutic protein, an HIV p17 matrix protein inhibitor, an IL-13 antagonist, a peptidyl-prolyl cis-trans isomerase a modulator, a protein disulfide isomerase inhibitor, a complement C5a receptor antagonist, a DNA methyltransferase inhibitor, a HIVvif gene modulator, a Vif dimerization antagonist, an HIV-1 viral infection factor inhibitor, TAT protein inhibitors, HIV-1Nef modulators, Hck tyrosine kinase modulators, mixed lineage kinase 3(MLK-3) inhibitors, HIV-1 splice inhibitors, Rev protein inhibitors, integrin antagonists, nucleoprotein inhibitors, splice factor modulators, COMM domain containing protein 1 modulators, HIV ribonuclease H inhibitors, reverse cyclin (retrocyclin) modulators, CDK-9 inhibitors, dendritic ICAM-3 trap non-integrin 1 inhibitors, HIV GAG protein inhibitors, HIV POL protein inhibitors, complement factor H modulators, ubiquitin ligase inhibitors, deoxycytidine kinase inhibitors, cyclin dependent kinase inhibitors, proprotein convertase PC9 stimulators, ATP dependent RNA helicase DDX3X inhibitors, reverse transcriptase priming complex inhibitors, G6PD and NADH-oxidase inhibitors, Pharmacokinetic enhancers, HIV gene therapy, HIV vaccines, and combinations thereof.

In some embodiments, the additional therapeutic agent is selected from the group consisting of combination drugs for HIV, other drugs for HIV, HIV protease inhibitors, HIV reverse transcriptase inhibitors, HIV integrase inhibitors, HIV non-catalytic site (or allosteric) integrase inhibitors, HIV entry (fusion) inhibitors, HIV maturation inhibitors, latency reversal agents, capsid inhibitors, immune-based therapies, PI3K inhibitors, HIV antibodies and bispecific antibodies, and "antibody-like" therapeutic proteins, and combinations thereof.

Examples of combination drugs that can be used with the antibodies of the present disclosure include

Figure BDA0002328692870001161

(efavirenz, tenofovir disoproxil fumarate and emtricitabine);

Figure BDA0002328692870001162

(

Figure BDA0002328692870001163

rilpivirine, tenofovir disoproxil fumarate, and emtricitabine);

Figure BDA0002328692870001164

(

Figure BDA0002328692870001165

rilpivirine, tenofovir disoproxil fumarate, and emtricitabine);(elvitegravir, coltstat, tenofovir disoproxil fumarate, and emtricitabine);(tenofovir disoproxil fumarate and emtricitabine; TDF + FTC);

Figure BDA0002328692870001168

(tenofovir alafenamide and emtricitabine);

Figure BDA0002328692870001169

(tenofovir alafenamide, emtricitabine, and rilpivirine);

Figure BDA00023286928700011610

(tenofovir alafenamide, emtricitabine, cosotal and elvitegravir); darunavir, tenofovir alafenamide hemifumarate, emtricitabine, and costatate; efavirenz, lamivudine and tenofovir disoproxil fumarate; lamivudine and tenofovir disoproxil fumarate; tenofovir and lamivudine; tenofovir alafenamide and emtricitabine; tenofovir alafenamide hemifumarate and emtricitabine; tenofovir alafenamide hemifumarate, emtricitabine and rilpivirine; tenofovir alafenamide hemifumarate, emtricitabine, cosotal and elvitegravir;(zidovudine and lamivudine; AZT +3 TC);(

Figure BDA00023286928700011613

abacavir sulfate and lamivudine; ABC +3 TC);

Figure BDA00023286928700011614

(

Figure BDA00023286928700011615

lopinavir and ritonavir);

Figure BDA00023286928700011616

(dolutegravir, abacavir and lamivudine);

Figure BDA00023286928700011617

(abacavir sulfate, zidovudine and lamivudine; ABC + AZT +3 TC); atazanavir and comparastat; atazanavir sulfate and comparastat; atazanavir sulfate and ritonavir; darunavir and costal; dolutegravir and rilpivirine; dolutegravir and rilpivirine hydrochloride; dolutegravir, abacavir sulfate and lamivudine; lamivudine, nevirapine, and zidovudine; rosiglivir and lamivudine; dolavine, lamivudine and tenofovir disoproxil fumarate; dolavine, lamivudine and tenofovir disoproxil; dolutegravir + lamivudine, lamivudine + abacavir + zidovudine, lamivudine + abacavir, lamivudine + tenofovir fumarate, lamivudine + zidovudine + nevirapine, lopinavir + ritonavir + abacavir + lamivudine, lopinavir + ritonavir + zidovudine + lamivudine, tenofovir + lamivudine, and tenofovir fumarate + ritonavir + rillitabine + rilpivirine hydrochloride, lopinavir, ritonavir, zidovudine, and lamivudine; vacc-4x and romidepsin; and APH-0812.

Examples of other drugs that may be used in combination with the antibodies of the present disclosure for the treatment of HIV include: acymphopyran, alisporivir, BanLec, deferiprone, Gamimone, maytansinoid, naltrexone, prolactin, REP 9, RPI-MN, VSSP, H1viral, SB-728-T, 1, 5-dicaffeoylquinic acid, rHIV7-shl-TAR-CCR5RZ, AAV-eCD4-Ig gene therapy, MazF gene therapy, BlockAide, ABX-464, AG-1105, APH-0812, BIT-225, CYT-107, HGTV-43, HPH-116, HS-10234, IMO-635, IND-02, MK-1376, MK-8507, MK-8591, NOV-205, PA-1050040 (PA-PGV), N-007, SCY-3100, SB-9200, SCB-90452, TETR-TEV-90110, 719-TEV-90112, MK-111, 111-111V-111, RN-18, Immuglo and VIR-576.

Examples of HIV protease inhibitors that may be combined with the antibodies of the present disclosure include amprenavir, atazanavir, brecanavir, daranavir, fosamprenavir calcium, indinavir sulfate, lopinavir, nelfinavir mesylate, ritonavir, saquinavir mesylate, tipranavir, DG-17, TMB-657(PPL-100), T-169, BL-008, and TMC-310911.

Examples of HIV nucleoside or non-nucleotide reverse transcriptase inhibitors that may be combined with the antibodies of the present disclosure include dapivirine, delavirdine mesylate, dolavine, efavirenz, etravirine, lentinan, nevirapine, rilpivirine, ACC-007, AIC-292, KM-023, and VM-1500.

Examples of HIV nucleoside or nucleotide reverse transcriptase inhibitors that may be combined with an antibody of the present disclosure include adefovir, adefovir dipivoxil, alfvudine, emtricitabine, tenofovir alafenamide fumarate, tenofovir alafenamide hemifumarate, tenofovir disoproxil fumarate, tenofovir disoproxil hemifumarate,

Figure BDA0002328692870001181

and

Figure BDA0002328692870001182

(didanosine, ddl), abacavir sulfate, alovudine, aplidine, censvudine, didanosine, elvitein, filtinivir, fosalvudine tidoxil, CMX-57, dapivirine, dolavine, etravirine, OCR-5753, tenofovir disoproxil orotate, fosalvudine tidoxil, lamivudine, phosphazene, stavudine, zalcitabine, zidovudine, GS-9131, GS-9148, and KP-1461.

Examples of HIV integrase inhibitors that may be combined with the antibodies of the present disclosure include elvitegravir, curcumin derivatives, sebacic acid derivatives, 3, 5-dicaffeoylquinic acid, aurintricarboxylic acid, derivatives of aurintricarboxylic acid, caffeic acid phenethyl ester, derivatives of caffeic acid phenethyl ester, tyrphostin (tyrphostin), tyrphostin derivatives, quercetin derivatives, raltegravir, dolutegravir, JTK-351, bictegravir, AVX-15567, caboteravivir (depot injection), diketimine 4-1 derivatives, integrase LEDGF inhibitors, ledgins, M-522, M-532, NSC-310217, NSC-371056, NSC-48240, NSC-642710, NSC-699171, NSC-699172, NSC-699173, NSC-699174, stilbene disulfonic acid, T-169, and cabotegravir.

Examples of HIV non-catalytic sites or allosteric integrase inhibitors (NCINIs) that may be combined with the antibodies of the present disclosure include CX-05045, CX-05168, and CX-14442.

Examples of HIV entry (fusion) inhibitors that may be combined with the antibodies of the present disclosure include cericiviroc, CCR5 inhibitors, gp41 inhibitors, CD4 attachment inhibitors, gp120 inhibitors, and CXCR4 inhibitors.

Examples of CCR5 inhibitors that may be combined with the antibodies of the present disclosure include apvirovir, vicriviroc, maravir oc, cenicriviroc, PRO-140, adaptavir (RAP-101), nifeviroc (TD-0232), anti-GP 120/CD4 or CCR5 bispecific antibodies, B-07, MB-66, the polypeptides C25P, TD-0680, and vMIP (Haimivu).

Examples of gp41 inhibitors that can bind to the antibodies of the present disclosure include abacavir peptide, enfuvirtide, BMS-986197, enfuvirtide biologics, enfuvirtide biosimiders, HIV-1 fusion inhibitors (P26-Bapc), ITV-1, ITV-2, ITV-3, ITV-4, PIE-12 trimer, and Cifuvirtide.

Examples of CD4 attachment inhibitors that can be combined with the antibodies of the present disclosure include ibalizumab and CADA analogs.

Examples of gp120 inhibitors that can bind to the antibodies of the present disclosure include Radha-108 (receptor) 3B3-PE38, BanLec, bentonite-based nanomedicines, fostemavir tromethamine, IQP-0831, and BMS-663068

Examples of CXCR4 inhibitors that can be combined with the antibodies of the present disclosure include plexafort, ALT-1188, N15 peptide, and vmip (haiipipu).

Examples of HIV maturation inhibitors that can be combined with the antibodies of the present disclosure include BMS-955176 and GSK-2838232.

Examples of latency reversing agents that can bind to the antibodies of the present disclosure include Histone Deacetylase (HDAC) inhibitors, proteasome inhibitors such as velcade, protein kinase c (pkc) activators, Smyd2 inhibitors, BET-bromodomain 4(BRD4) inhibitors, ionomycin, PMA, SAHA (mercaptohydroxamic acid or sulfinyl, aniline and hydroxamic acid), NIZ-985, IL-15, JQ1, disulfiram, amphotericin B, and ubiquitin inhibitors (e.g., lagranol analogs and GSK-343). Examples of HDAC inhibitors include romidepsin, vorinostat and panobinostat. Examples of PKC activators include indomethacin, prostratin, eugenol B, and DAG lactone.

Examples of capsid inhibitors that may be combined with the antibodies of the present disclosure include capsid polymerization inhibitors or capsid disrupting compounds, HIV nucleocapsid p7(NCp7) inhibitors such as azodicarbonamide, HIV p24 capsid protein inhibitors, AVI-621, AVI-101, AVI-201, AVI-301, and AVI-CAN1-15 series.

Examples of immune-based therapies that may be combined with the antibodies of the present disclosure include toll-like receptor modulators such as TLR1, TLR2, TLR3, TLR4, TLR5, TLR6, TLR7, TLR8, TLR9, TLR10, TLR11, TLR12, and TLR13, programmed cell death protein 1(PD-1) modulators, programmed death ligand 1(PD-L1) modulators, IL-15 agonists, DermaVir, interleukin 7, quinil (hydroxychloroquine), proleukin (aldesleukin, IL-2), interferon α, interferon α -2b, interferon α -n3, pegylated interferon α, interferon gamma, hydroxyurea, mycophenolic acid (MPA) and its ester derivatives Mycophenolate Mofetilate (MMF), ribavirin, ritolimod, Polyethyleneimine (PEI), gelonin, pegylated interferon 12, interferon-grafted interferon gamma, MPF-962, MPF, MPE-7, MPS-15, MPS-7, MPS-2, MPS-7, MPS-2, and MPS-.

Examples of PI3K inhibitors that may be combined with the antibodies of the present disclosure include idarasib (idelalisib), Abelisib (aleglisib), Buparlisib (buparlisib), CAI orotic acid, copanlisib, duvelisib, gedatolisib, neratinib, panculisib, periplosine, pictilisib, pilalaisib, puquitinib mesylate, rigosertib sodium, sonolisib, taselisib, AMG-319, AZD-8186, BAY-1082439, CLR-1401, CLR-457, CUDC-907, DS-7423, SAR-3342, GSK-2126458, GSK-2269577, GSK-2636771, GSC-040093, CLR-3023414, MLN-309, MLR-1726, SAR-1726, RV-17230, TGSR-559, ZSR-559, TGSF-1126, TGSF-XL-1126, TGASR-19, SAR-RG-4480, SAR-4417, SAR-RG-102, SAR-LSR-4417, SAR-102, SAR-LSR-102, SAR-RG-102, and ZSR-LSR-4417.

Examples of integrin α -4/β -7 antagonists that can be combined with the antibodies of the present disclosure include PTG-100, TRK-170, Abbruzumab (abrilumab), Epoluzumab (etrolizumab), Caroglustrat methyl (carotegrast methyl), and Vidolizumab (vedolizumab).

Examples of HIV antibodies, bispecific antibodies, and "antibody-like" therapeutic proteins that can be combined with the antibodies of the present disclosure include

Figure BDA0002328692870001201

Figure BDA0002328692870001202

Fab derivatives, bNAbs (broadly neutralizing HIV-1 antibody), BMS-936559, TMB-360 and antibodies targeting HIV GP120 or GP41, HIV-targeting antibody recruiting molecules, anti-CD 63 monoclonal antibodies, anti-GB virus C antibodies, anti-GP 120/CD4, CCR5 bispecific antibodies, anti-nef single domain antibodies, anti-Rev antibodies, camel-derived anti-CD 18 antibodies, camel-derived anti-ICAM-1 antibodies, DCVax-001, GP140 targeting antibodies, GP 41-based HIV therapeutic antibodies, human recombinant mAb (PGT-121), ebazumab, Immuglo, MB-66. Examples of those that target HIV in this manner include bavimab, UB-421, C2F5, C2G12, C4E10, C2F5+ C2G12+ C4E10, 3BNC-117, PGT145, PGT121, PGDM1400, MDX010 (ipilimumab), VRC01, A32, 7B2, 10E8, VRC-07-523, VRC-HIVMAB080-00-AB, MGD-014, and VRC 07. Other examples of antibodies targeting HIV include PGT122, PGT123, PGT124, 10-1074, PGT133, PGT134, PG16, PG9, PGT151, and the like.

Examples of pharmacokinetic enhancers that may be combined with the antibodies of the present disclosure include cobicistat and ritonavir.

Examples of other therapeutic agents that may be combined with the antibodies of the present disclosure include compounds disclosed in WO 2004/096286(GileadSciences), WO 2006/015261(Gilead Sciences), WO 2006/110157(Gilead Sciences), WO2012/003497(Gilead Sciences), WO 2012/003498(Gilead Sciences), WO 2012/145728(Gilead Sciences), WO 2013/006738(Gilead Sciences), WO 2013/159064(GileadSciences), WO 2014/100323(Gilead Sciences), US 2013/0165489(University of pennsylania), US 2014/0221378 (Japan tobacademy), US 2014/0221380(Japan tobaco), WO 2009/062285 (Boehringer Ingelheim), WO 2010/130034(Boehringer Ingelheim), WO2013/006792(Pharma resco), US 20140221356 (gileahrengenhence 20100143301), WO 596 (giliewic), and sci 3 (giliewic) are disclosed.

Examples of HIV vaccines that can be combined with the antibodies of the present disclosure include peptide vaccines, recombinant subunit protein vaccines, live vector vaccines, DNA vaccines, CD 4-derived peptide vaccines, vaccine combinations, rgp120(AIDSVAX), ALVAC HIV (vCP1521)/AIDSVAX B/E (GP120) (RV144), monomeric GP120 HIV-1 subtype C vaccines, Remune, ITV-1, Contre Vir, Ad5-ENVA-48, DCVax-001(CDX-2401), Vacc-4x, Vacc-C5, VAC-3S, multiclade DNA recombinant adenovirus 5(rAd5), Pennvax-G, Pennvax-GP, HIV-Trimix-mRNA vaccines, HIV-LAMP-vax, Ad35, Ad35-GRIN, NAcGM3/VSSP ISA-51, poly-ICLC adjuvants, Tatmultine, GTU-06, HIV (FIT-06), VV [ 1 ] GP 2.140 + VV 59, rVSVIN HIV-1 Gag vaccine, SeV-Gag vaccine, AT-20, DNK-4, Ad35-Grin/ENV, TBC-M4, HIVAX-2, NYVAC-HIV-PT1, NYVAC-HIV-PT4, DNA-HIV-PT123, rAAV1-PG9DP, GOVX-B11, GOVX-B21, TVI-HIV-1, Ad-4(Ad4-ENV clade C + Ad4-mGag), EN41-UGR7C, EN41-FPA2, PreVaxtat, GOAE-H, MYM-V101, CombiHIac, ADVAX, MYM-V201, MVA-CMDR, DNA-5 Gag/Mod/nef/nev (HVD 505), MVGTG-1, MVGTV-01, HIV-Gag vaccine, HIV-26-AVS-26, AVS-64201, AVS-AGX-26, AVS-26 SAV-001, ThV-01, TL-01, TUTI-16, VGX-3300, IHV-001, and virus-like particle vaccines such as pseudovirion vaccines, CombiVICHvac, LFn-p 24B/C fusion vaccines, GTU-based DNA vaccines, HIV gag/pol/nef/Env DNA vaccines, anti-TAT HIV vaccines, binding polypeptide vaccines, dendritic cell vaccines, gag-based DNA vaccines, GI-2010, GP41 HIV-1 vaccines, HIV vaccines (PIKA adjuvant), Ii-key/MHC class II epitope hybrid peptide vaccines, ITV-2, ITV-3, ITV-4, LIPO-5, multi-clade Env vaccines, MVA vaccines, Pennvax-GP, pp71 deficient MV HCV vector HIV gag vaccines, recombinant peptide vaccines (HIV infection), NCI, rgp HIV, RNActive HIV 160, SCB-703 HIV-703 vaccine, and HIV-like particle vaccines, Tat Oyi vaccine, TBC-M4, therapeutic HIV vaccine, UBI HIVgp120, Vacc-4x + romidepsin, variant gp120 polypeptide vaccine, rAd5 gag-pol env A/B/C vaccine.

Therapeutic agents for birth control (contraceptives) that can be combined with the antibodies of the present disclosure include cyproterone acetate, desogestrel, dienogest, drospirenone, estradiol valerate, ethinyl estradiol, norethindrone, etonogestrel, levofolinic acid, levonorgestrel, ethinyl estrenol, medroxyprogesterone acetate, mestranol, mifepristone, misoprostol acetate, nomegestrol acetate, norelgestromin, norethindrone, norgestimate, oxymetafene, segastersone acetate, ulipristal acetate, and any combination thereof.

In certain embodiments, the antibodies disclosed herein, or pharmaceutically acceptable salts thereof, are conjugated with one, two, three, four, or more members selected from the group consisting of

Figure BDA0002328692870001221

(efavirenz, tenofovir fumarate and emtricitabine);

Figure BDA0002328692870001222

(

Figure BDA0002328692870001223

rilpivirine, tenofovir disoproxil fumarate, and emtricitabine);

Figure BDA0002328692870001224

(elvitegravir, coltstat, tenofovir disoproxil fumarate, and emtricitabine);

Figure BDA0002328692870001231

(tenol fumarate)Fuvirate, emtricitabine; TDF + FTC);

Figure BDA0002328692870001232

(tenofovir alafenamide and emtricitabine);(tenofovir alafenamide, emtricitabine, and rilpivirine);

Figure BDA0002328692870001234

(tenofovir alafenamide, emtricitabine, cosotal and elvitegravir); adefovir dipivoxil; adefovir dipivoxil; can be compared with the sitagliptin; emtricitabine; tenofovir disoproxil fumarate; tenofovir disoproxil fumarate; tenofovir disoproxil fumarate; tenofovir alafenamide; tenofovir alafenamide hemifumarate;

Figure BDA0002328692870001235

(dolutegravir, abacavir and lamivudine); dolutegravir, abacavir sulfate and lamivudine; leegievir; rosiglivir and lamivudine; maravir oc; enfuvirtide;

Figure BDA0002328692870001236

(

Figure BDA0002328692870001237

lopinavir and ritonavir);

Figure BDA0002328692870001238

(zidovudine and lamivudine; AZT +3 TC);

Figure BDA0002328692870001239

(

Figure BDA00023286928700012310

abacavir sulfate and lamivudine; ABC +3 TC);(Abacavir sulfate, zidovudine andlamivudine; ABC + AZT +3 TC); rilpivirine; rilpivirine hydrochloride; atazanavir sulfate and comparastat; atazanavir and comparastat; darunavir and costal; atazanavir; atazanavir sulfate; dolutegravir; elvitegravir; ritonavir; atazanavir sulfate and ritonavir; darunavir; lamivudine; prolactin; fosamprenavir; fosamprenavir calcium efavirenz; etravirine; nelfinavir; nelfinavir mesylate; an interferon; didanosine; stavudine; indinavir; indinavir sulfate; tenofovir and lamivudine; zidovudine; nevirapine; saquinavir; saquinavir mesylate; aldesleukin; zalcitabine; tipranavir; apravir; delavirdine; delavirdine mesylate; radha-108 (receptor); lamivudine and tenofovir disoproxil fumarate; efavirenz, lamivudine and tenofovir disoproxil fumarate; phosphazenes; lamivudine, nevirapine, and zidovudine; abacavir; and abacavir sulfate.

In a specific embodiment, the antibodies disclosed herein or pharmaceutical compositions thereof are combined with an HIV nucleoside or nucleotide inhibitor of reverse transcriptase and an HIV non-nucleoside inhibitor of reverse transcriptase. In another specific embodiment, the antibodies disclosed herein or pharmaceutical compositions thereof are combined with an HIV nucleoside or nucleotide inhibitor of reverse transcriptase and an HIV protease inhibiting compound. In additional embodiments, the antibodies disclosed herein or pharmaceutical compositions thereof are combined with an HIV nucleoside or nucleotide inhibitor of reverse transcriptase, an HIV non-nucleoside inhibitor of reverse transcriptase, and a pharmacokinetic enhancer. In certain embodiments, the antibodies disclosed herein or pharmaceutical compositions thereof are combined with at least one HIV nucleoside inhibitor of reverse transcriptase, an integrase inhibitor, and a pharmacokinetic enhancer. In another embodiment, the antibodies disclosed herein or pharmaceutical compositions thereof are combined with HIV nucleoside or nucleotide inhibitors of two reverse transcriptases.

In particular embodiments, the antibodies disclosed herein or pharmaceutical compositions thereof are combined with abacavir sulfate, tenofovir disoproxil, tenofovir fumarate, tenofovir alafenamide hemifumarate, tenofovir alafenamide, or tenofovir alafenamide hemifumarate.

In particular embodiments, the antibodies disclosed herein or pharmaceutical compositions thereof are combined with tenofovir, tenofovir disoproxil fumarate, tenofovir alafenamide, or tenofovir alafenamide hemifumarate.

In a particular embodiment, the antibody or pharmaceutical composition thereof disclosed herein is combined with a first additional therapeutic agent selected from abacavir sulfate, tenofovir disoproxil fumarate, tenofovir alafenamide, and tenofovir alafenamide hemifumarate, and a second additional therapeutic agent selected from emtricitabine and lamivudine.

In a particular embodiment, the antibody disclosed herein or a pharmaceutical composition thereof is combined with a first additional therapeutic agent selected from the group consisting of tenofovir, tenofovir disoproxil, tenofovir alafenamide, and tenofovir alafenamide hemifumarate, and a second additional therapeutic agent, wherein the second additional therapeutic agent is emtricitabine.

In another specific embodiment, the antibody disclosed herein or a pharmaceutical composition thereof is combined with a first additional therapeutic agent (contraceptive) of cyproterone acetate, desogestrel, dienogest, drospirenone, estradiol valerate, ethinyl estradiol, norethindrone, etonogestrel, levofolinic acid, levonorgestrel, ethinyl estrenol acetate, medroxyprogesterone acetate, mestranol, mifepristone, misoprostol acetate, nomegestrol, norelgestromin, norethindrone, norgestimate, oxymetafil, segetssone acetate, ulipristal acetate, and any combination thereof.

Reagent kit

The present disclosure also encompasses kits comprising one or more of the antibodies described herein or conjugates thereof. In one instance, provided herein is a pharmaceutical package or kit comprising one or more containers filled with one or more ingredients of a pharmaceutical composition described herein, e.g., one or more antibodies provided herein. In some cases, a kit comprises a pharmaceutical composition described herein. In one embodiment, a kit is provided comprising an antibody disclosed herein or a pharmaceutical composition thereof in combination with one or more (e.g., one, two, three, one or two, or one to three) additional therapeutic agents (e.g., those described above). Optionally, associated with such containers may be a notice in a form prescribed by an institution for regulating the production, use, or sale of a pharmaceutical or biological product, the notice reflecting approval by the institution for manufacture, use, or sale for human administration.

250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:纯化抗体的方法

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!