Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
BISPECIFIC MOLECULES COMPRISING AN HIV-1 ENVELOPE TARGETING ARM
Document Type and Number:
WIPO Patent Application WO/2018/053328
Kind Code:
A1
Abstract:
The invention is directed to multispecific molecules comprising an HIV-1 envelope targeting arm and an arm targeting an effector cell, compositions comprising these molecules and methods of use.

Inventors:
HAYNES BARTON F (US)
NORDSTROM JEFFREY LEE (US)
LAM CHIA-YING KAO (US)
MOODY M ANTHONY (US)
WILLIAMS LATONYA (US)
Application Number:
PCT/US2017/051877
Publication Date:
March 22, 2018
Filing Date:
September 15, 2017
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
UNIV DUKE (US)
MACROGENICS INC (US)
International Classes:
A61K38/00; A61K39/395; A61K39/42; C07K16/10; C07K16/46; C07K19/00
Domestic Patent References:
WO2016054101A12016-04-07
WO2015184207A12015-12-03
WO2013070776A12013-05-16
Foreign References:
US20150152167A12015-06-04
US20120269821A12012-10-25
US20160222105A12016-08-04
Attorney, Agent or Firm:
KIM, William W. et al. (US)
Download PDF:
Claims:
CLAIMS

1. A bispecific molecule comprising a first polypeptide chain and a second polypeptide chain, covalently bonded to one another, wherein:

(I) the first polypeptide chain comprises in the N- to C-terminal direction:

(i) a domain (A) comprising a binding region of the light chain variable domain of a first immunoglobulin (VLl) having the binding specificity of a gp41 MPER binding HIV- 1 antibody (1);

(ii) a domain (B) comprising a binding region of a heavy chain variable domain of a second immunoglobulin (VH2) specific for an epitope (2); and

(iii) a domain (C) comprising a heterodimer promoting domain;

(II) the second polypeptide chain comprises in the N- to C-terminal direction:

(i) a domain (D) comprising a binding region of a light chain variable domain of the second immunoglobulin (VL2) specific for the epitope (2);

(ii) a domain (E) comprising a binding region of a heavy chain variable domain of the first immunoglobulin (VHl) having the binding specificity of a gp41 MPER binding HIV- 1 antibody (1); and

(iii) a domain (F) comprising a heterodimer promoting domain; and wherein:

the domains (A) and (B) are linked so that they do not associate with one another to form an epitope binding site;

the domains (D) and (E) are linked so that they do not associate with one another to form an epitope binding site; and

the domains (A) and (E) associate to form a binding site that binds the HIV-1 envelope like a gp41 MPER binding HIV-1 antibody (1); and the domains (B) and (D) associate to form a binding site that binds the epitope (2).

2. The bispecific molecule of claim 1, wherein:

(i) domains (A) and (B) are separated by a peptide linker 1 ;

(ii) domains (C) and (B) are separated by a peptide linker 2;

(iii) domains (D) and (E) are separated by a peptide linker 1 ; and

(iv) domains (F) and (E) are separated by a peptide linker 2.

3. A bispecific molecule comprising a first polypeptide chain, a second polypeptide chain, and a third polypeptide chain, wherein the first and second polypeptide chains are covalently bonded and the second and third polypeptide chains are covalently bonded, and wherein:

(I) the first polypeptide chain comprises in the N- to C-terminal direction:

(i) a domain (A) comprising a binding region of the light chain variable domain of a first immunoglobulin (VLl) having the binding specificity of a gp41 MPER binding HIV- 1 antibody (1);

(ii) a domain (B) comprising a binding region of a heavy chain variable domain of a second immunoglobulin (VH2) specific for an epitope (2);

(iii) a domain (C) comprising a heterodimer promoting domain; and

(iv) a CH2-CH3 domain;

(II) the second polypeptide chain comprises in the N- to C-terminal direction:

(i) a domain (D) comprising a binding region of a light chain variable domain of the second immunoglobulin (VL2) specific for the epitope (2);

(ii) a domain (E) comprising a binding region of a heavy chain variable domain of the first immunoglobulin (VHl) having the binding specificity of a gp41 MPER binding HIV- 1 antibody (1); and

(iii) a domain (F) comprising a heterodimer promoting domain;

(III) the third polypeptide chain comprises in the N- to C-terminal direction:

(i) a CH2-CH3 domain, and wherein:

the domains (A) and (B) are linked so that they do not associate with one another to form an epitope binding site;

the domains (D) and (E) are linked so that they do not associate with one another to form an epitope binding site;

the domains (A) and (E) associate to form a binding site that binds the HIV-1 envelope like a gp41 MPER binding HIV-1 antibody (1);

the domains (B) and (D) associate to form a binding site that binds the epitope (2); and the CH2-CH3 domains of the first and third polypeptide form an Fc Domain.

4. The bispecific molecule of claim 3, wherein: (i) the third polypeptide chain further comprises a peptide linker 3 N-terminal to the CH2-CH3 domain;

(ii) domains (A) and (B) are separated by a peptide linker 1;

(iii) domains (C) and (B) are separated by a peptide linker 2;

(iv) the CH2-CH3 domain and domain (C) are separated by a peptide linker 3 or a spacer linker 3;

(v) domains (D) and (E) are separated by a peptide linker 1 ; and

(vi) domains (F) and (E) are separated by a peptide linker 2.

5. A bispecific molecule comprising a first polypeptide chain, a second polypeptide chain and a third polypeptide chain, wherein the first and second polypeptide chains are covalently bonded and the second and third polypeptide chains are covalently bonded, and wherein:

(I) the first polypeptide chain comprises in the N- to C-terminal direction:

(i) a CH2-CH3 domain;

(ii) a domain (A) comprising a binding region of the light chain variable domain of a first immunoglobulin (VLl) having the binding specificity of a gp41 MPER binding HIV- 1 antibody (1);

(iii) a domain (B) comprising a binding region of a heavy chain variable domain of a second immunoglobulin (VH2) specific for an epitope (2); and

(iv) a domain (C) comprising a heterodimer promoting domain;

(II) the second polypeptide chain comprises in the N- to C-terminal direction:

(i) a domain (D) comprising a binding region of a light chain variable domain of the second immunoglobulin (VL2) specific for the epitope (2);

(ii) a domain (E) comprising a binding region of a heavy chain variable domain of the first immunoglobulin (VHl) having the binding specificity of a gp41 MPER binding HIV- 1 antibody (1); and

(iii) a domain (F) comprising a heterodimer promoting domain;

(III) the third polypeptide chain comprises in the N- to C-terminal direction:

(i) a CH2-CH3 domain, and wherein:

the domains (A) and (B) are linked so that they do not associate with one another to form an epitope binding site; the domains (D) and (E) are linked so that they do not associate with one another to form an epitope binding site;

the domains (A) and (E) associate to form a binding site that binds the HIV-1 envelope like a gp41 MPER bindingHIV-1 antibody (1);

the domains (B) and (D) associate to form a binding site that binds the epitope (2); and the CH2-CH3 domains of the first and third polypeptide form an Fc Domain.

6. The bispecific molecule of claim 5, wherein:

(i) the CH2-CH3 domain and domain (A) are separated by a peptide linker 4;

(ii) domains (A) and (B) are separated by a peptide linker 1 ;

(iii) domains (C) and (B) are separated by a peptide linker 2;

(iv) domains (D) and (E) are separated by a peptide linker 1 ;

(v) domains (F) and (E) are separated by a peptide linker 2;

(vi) the first polypeptide chain further comprises a peptide linker 3 N-terminal to the CH2-CH3 domain; and

(vii) the third polypeptide chain further comprises a peptide linker 3 N-terminal to the CH2-CH3 domain.

7. A bispecific molecule comprising a first polypeptide chain, a second polypeptide chain, a third polypeptide chain, and a fourth polypeptide chain, wherein the first and second polypeptide chains are covalently bonded and the first and third polypeptide chains are covalently bonded, and wherein:

(I) the first and the third polypeptide chains each comprise in the N- to C-terminal direction:

(i) a domain (A) comprising a binding region of the light chain variable domain of a first immunoglobulin (VLl) having the binding specificity of a gp41 MPER binding HIV- 1 antibody (1);

(ii) a domain (B) comprising a binding region of a heavy chain variable domain of a second immunoglobulin (VH2) specific for an epitope (2);

(iii) a domain (C) comprising a heterodimer promoting domain; and

(iv) a CH2-CH3 domain;

(II) the second and fourth polypeptide chains each comprise in the N- to C-terminal direction: (i) a domain (D) comprising a binding region of a light chain variable domain of the second immunoglobulin (VL2) specific for the epitope (2);

(ii) a domain (E) comprising a binding region of a heavy chain variable domain of the first immunoglobulin (VH1) having the binding specificity of a gp41 MPER bindingHIV- 1 antibody (1);

(iii) a domain (F) comprising a heterodimer promoting domain; and wherein the domains (A) and (B) are linked so that they do not associate with one another to form an epitope binding site;

the domains (D) and (E) are linked so that they do not associate with one another to form an epitope binding site;

the domains (A) and (E) associate to form a binding site that binds the HIV-1 envelope like a gp41 MPER binding HIV-1 antibody (1);

the domains (B) and (D) associate to form a binding site that binds the epitope (2); and the CH2-CH3 domains of the first and third polypeptide form an Fc Domain.

8. The bispecific molecule of claim 7, wherein:

(i) domains (A) and (B) are separated by a peptide linker 1 ;

(ii) domains (C) and (B) are separated by a peptide linker 2;

(iii) the CH2-CH3 domain and domain (C) are separated by a peptide linker 3 or a spacer linker 3;

(iv) domains (D) and (E) are separated by a peptide linker 1 ; and

(v) domains (F) and (E) are separated by a peptide linker 2.

9. A trivalent binding molecule comprising a first, second, third and fourth polypeptide chain wherein:

(I) the first polypeptide chain comprises in the N-terminus to C-terminus direction:

(i) a domain (A) comprising a binding region of a light chain variable domain of a first immunoglobulin (VL1) specific for an epitope (1);

(ii) a domain (B) comprising a binding region of a heavy chain variable domain of a second immunoglobulin (VH2) specific for an epitope (2);

(iii) a domain (C) comprising a heterodimer promoting domain; and

(iv) a CH2-CH3 Domain;

(II) the second polypeptide chain comprises, in the N-terminus to C-terminus direction: (i) a domain (D) comprising a binding region of a light chain variable domain of the first immunoglobulin (VL1) specific for the epitope (2);

(ii) a domain (E) comprising a binding region of a heavy chain variable domain of the second immunoglobulin (VH1) specific for the epitope (1);

(iii) a domain (F) comprising a heterodimer promoting domain; and

(III) the third polypeptide chain that comprises, in the N-terminus to C-terminus direction:

(i) a domain (G) comprising a binding region of a heavy chain variable domain of a third immunoglobulin (VH3) specific for an epitope (3); and

(ii) a CHI -Hinge Domain, and a CH2-CH3 Domain; and

(IV) the fourth polypeptide chain that comprises, in the N-terminus to C-terminus direction:

(i) a domain (H) comprising a binding region of a light chain variable domain of the third immunoglobulin (VL3) specific for the epitope (3); and

(ii) CL Kappa Domain or a CL Lambda Domain; and wherein

the domains (A) and (B) are linked so that they do not associate with one another to form an epitope binding site;

the domains (D) and (E) are linked so that they do not associate with one another to form an epitope binding site;

the domains (A) and (E) associate to form a binding site that binds the epitope (1);

the domains (B) and (D) associate to form a binding site that binds the epitope (2);

the domains (G) and (H) associate to form a binding site that bind the epitope (3);

at least one of epitope (1), epitope (2), and epitope (3) is an epitope bound by an gp41 MPER binding HIV- 1 antibody, and at least one of epitope (1), epitope (2), and epitope (3) is an epitope of CD3, CD8, or CD 16;

the CH2-CH3 domains of the first and third polypeptide form an Fc Domain;

said first and second polypeptide chains are covalently bonded to one another;

said first and third polypeptide chains are covalently bonded to one another; and

said third and fourth polypeptide chains are covalently bonded to one another.

10. The trivalent binding molecule of claim 9, wherein:

(i) domains (A) and (B) are separated by a peptide linker 1 ;

(ii) domains (C) and (B) are separated by a peptide linker 2 or a peptide linker 2-C;

(iii) the CH2-CH3 domain and domain (C) are separated by a peptide linker 3 or a spacer linker 3; (iv) domains (D) and (E) are separated by a peptide linker 1 ; and

(v) domains (F) and (E) are separated by a peptide linker 2 or a peptide linker 2-C.

11. A trivalent binding molecule comprising a first, second, and third polypeptide chain wherein:

(I) the first polypeptide chain comprises in the N-terminus to C-terminus direction:

(i) a domain (A) comprising a binding region of a light chain variable domain of a first immunoglobulin (VL1) specific for an epitope (1);

(ii) a domain (B) comprising a binding region of a heavy chain variable domain of a second immunoglobulin (VH2) specific for an epitope (2);

(iii) a domain (C) comprising a heterodimer promoting domain; and

(iv) a CH2-CH3 Domain;

(II) the second polypeptide chain comprises, in the N-terminus to C-terminus direction:

(i) a domain (D) comprising a binding region of a light chain variable domain of the first immunoglobulin (VL1) specific for the epitope (2);

(ii) a domain (E) comprising a binding region of a heavy chain variable domain of the second immunoglobulin (VH1) specific for the epitope (1);

(iii) a domain (F) comprising a heterodimer promoting domain; and

(III) the third polypeptide chain that comprises, in the N-terminus to C-terminus direction:

(i) a domain (H) comprising a binding region of a light chain variable domain of a third immunoglobulin (VL3) specific for an epitope (3)

(ii) a domain (G) comprising a binding region of a heavy chain variable domain of the third immunoglobulin (VH3) specific for the epitope (3);

(iii) a CH2-CH3 Domain; and wherein

the domains (A) and (B) are linked so that they do not associate with one another to form an epitope binding site;

the domains (D) and (E) are linked so that they do not associate with one another to form an epitope binding site;

the domains (A) and (E) associate to form a binding site that binds the epitope (1);

the domains (B) and (D) associate to form a binding site that binds the epitope (2);

the domains (G) and (H) associate to form a binding site that bind the epitope (3); at least one of epitope (1), epitope (2), and epitope (3) is an epitope bound by a gp41 MPER binding HIV-1 antibody, and at least one of epitope (1), epitope (2), and epitope (3) is and epitope of CD3, CD8, or CD 16;

the CH2-CH3 domains of the first and third polypeptide form an Fc Domain;

said first and second polypeptide chains are covalently bonded to one another;

said first and third polypeptide chains are covalently bonded to one another; and

said third and fourth polypeptide chains are covalently bonded to one another.

12. The trivalent binding molecule of claim 1 1, wherein:

(i) domains (A) and (B) are separated by a peptide linker 1 ;

(ii) domains (C) and (B) are separated by a peptide linker 2 or a peptide linker 2-C;

(iii) the CH2-CH3 domain and domain (C) are separated by a peptide linker 3 or a spacer linker 3;

(iv) domains (D) and (E) are separated by a peptide linker 1 ;

(v) domains (F) and (E) are separated by a peptide linker 2 or a peptide linker 2-C;

(vi) domains (H) and (G) are separated by a peptide linker 5; and

(vii) the CH2-CH3 domain and domain (G) are separated by a peptide linker 3.

13. The trivalent binding molecule of claims 9-12, wherein one of epitope ( 1), epitope (2), or epitope (3) is an epitope of CD8.

14. The molecules of claims 3-8 and 9-12, wherein the CH2-CH3 domain of the first

polypeptide chain is the of the "knob" design (SEQ ID NOs: 531 or 532) and the CH2- CH3 domain of the third polypeptide chain is of the "hole" design (SEQ ID NOs: 533 or 534).

15. The molecules of claim 14, wherein the CH2-CH3 domain of the first polypeptide

comprises SEQ ID NO: 531 and the CH2-CH3 domain of the third polypeptide chain comprises SEQ ID NO: 533.

16. The molecules of claims 3-8 and 9-12, wherein the CH2-CH3 domain of the third

polypeptide chain is the of the "knob" design (SEQ ID NOs: 531 or 532) and the CH2- CH3 domain of the first polypeptide chain is of the "hole" design (SEQ ID NOs: 533 or 534).

17. The molecules of claim 16, wherein the CH2-CH3 domain of the third polypeptide comprises SEQ ID NO: 531 and the CH2-CH3 domain of the first polypeptide chain comprises SEQ ID NO: 533.

18. The molecules of claims 1-12 wherein the epitope (2) is a CD3 epitope, CD8 epitope, or a CD 16 epitope.

19. The molecules of claims 1-12, wherein the gp41 MPER binding HIV-1 antibody is

DH51 1, DH51 1 2 K3, DH512, DH513, DH514, DH515, DH516, DH517, DH518, DH536, or DH537or any of the antibodies of the DH511 lineage.

20. The molecules of claims 1-12, wherein the molecule binds HIV-1 envelope with the specificity of the gp 41 MPER binding antibody DH512, DH511 2 K3 or any antibody of the DH51 1 lineage and binds CD3, CD8, or CD 16.

21. The molecules of claims 1-12, wherein domain (A) comprises the CDR1, CDR2, and CDR3 of the light chain variable domain of immunoglobulin DH512, DH51 1 2 K3 or any immunoglobulin of the DH51 1 lineage.

22. The molecules of claims 1-12, wherein domain (E) comprises the CDR1, CDR2, and CDR3 of the heavy chain variable domain of immunoglobulin DH512, or any immunoglobulin of the DH51 1 lineage.

23. The molecules of claims 1-12, wherein domain (A) comprises the light chain variable domain of immunoglobulin DH512, DH51 1 2 K3 or any immunoglobulin of the DH511 lineage.

24. The molecules of claims 1-12, wherein domain (E) comprises the heavy chain variable domain of immunoglobulin DH512, or any immunoglobulin of the DH51 1 lineage.

25. The molecules of claims 1-12, wherein domain (B) comprises the heavy chain variable domain of an anti-CD3 antibody.

26. The molecules of claims 1-12, wherein the domain (D) comprises the light chain variable domain of an anti-CD3 antibody.

27. The trivalent binding molecule of claims 9-12, wherein domain (G) comprises heavy chain variable domain of an anti-CD8 antibody.

28. The trivalent binding molecule of claims 9-12, wherein domain (H) comprises light chain variable domain of an anti-CD8 antibody.

29. The bispecific molecule of claim 3, wherein the first polypeptide comprises SEQ ID NO:

555, the second polypeptide comprises SEQ ID NO: 557, and the third polypeptide comprises SEQ ID NO: 559.

30. The trivalent binding molecule of claim 9, wherein a. the first polypeptide comprises SEQ ID NO: 555, the second polypeptide comprises SEQ ID NO: 557, the third polypeptide comprises SEQ ID NO: 561, and the fourth polypeptide comprises SEQ ID NO: 562; or

b. the first polypeptide comprises SEQ ID NO: 555, the second polypeptide comprises SEQ ID NO: 557, the third polypeptide comprises SEQ ID NO: 627, and the fourth polypeptide comprises SEQ ID NO: 628; or

c. comprises SEQ ID NO: 629, the second polypeptide comprises SEQ ID NO: 630, the third polypeptide comprises SEQ ID NO: 631, and the fourth polypeptide comprises SEQ ID NO: 632.

31. The trivalent binding molecule of claim 1 1, wherein the first polypeptide comprises SEQ ID NO: 555, the second polypeptide comprises SEQ ID NO: 557, and the third polypeptide comprises SEQ ID NO: 563.

32. A composition comprising any one of the molecules of claims 1-3 lor any combination thereof and a carrier.

33. A composition comprising a bispecific molecule or trivalent molecule which comprises at least one arm with the binding specificity of HIV- 1 antibody DH512, DH51 1 2 K3 or any antibody of the DH511 lineage, and a second arm targeting CD3, CD8 or CD 16.

34. The composition of claim 33, further comprising a second bispecific molecule or trivalent molecule comprising a first arm with the binding specificity of an HIV- 1 antibody different from the binding specificity of the first bispecific molecule or trivalent molecule and a second arm targeting CD3, CD8 or CD 16, wherein the first and second molecules are different.

35. A method to treat or prevent HIV- 1 infection in a subject in need thereof comprising

administering to the subject a composition comprising any one of the molecules of claim 1-31 or a combination of any one of these molecules in a therapeutically effective amount.

36. The method of claim 35, further comprising administering a latency activating agent.

37. The method of claim 36, wherein the latency activating agent is vorinostat, romidepsin, panobinostat, disulfiram, JQ 1, bryostatin, PMA, inonomycin, or any combination thereof.

38. A vector comprising nucleic acids comprising nucleotides encoding the molecules of any one of claim 1-31.

39. A composition comprising a vector comprising a nucleic acid encoding the molecules of any one of claim 1-31.

Description:
Bispecific Molecules Comprising an HIV-1 Envelope Targeting Arm

[0001] This invention claims the benefit of and priority to U.S. Serial No. 62/395,788, filed September 16, 2016, the contents of which is hereby incorporated by reference in its entirety.

[0002] This patent disclosure contains material that is subject to copyright protection. The copyright owner has no objection to the facsimile reproduction by anyone of the patent document or the patent disclosure as it appears in the U.S. Patent and Trademark Office patent file or records, but otherwise reserves any and all copyright rights.

[0003] All patents, patent applications and publications cited herein are hereby incorporated by reference in their entirety. The disclosure of these publications in their entireties are hereby incorporated by reference into this application in order to more fully describe the state of the art as known to those skilled therein as of the date of the invention described herein.

FIELD OF THE INVENTION

[0004] The invention is directed to multispecific molecules, such as but not limited to bispecific and trispecific molecules (e.g. bispecific antibodies, bispecific diabodies, and trivalent binding molecules) comprising an HIV-1 binding domain and an effector cell binding domain, and their uses.

BACKGROUND

[0005] Highly Active Antiretroviral Therapy (HAART) has been effective in reducing the viral burden and ameliorating the effects of HIV-1 infection in infected individuals. However, despite this therapy the virus persists in the individual due to latent reservoir of HIV-1 infected cells which evade this treatment. Thus, there is a need for therapeutic agents for treatment of HIV-1 infected individuals, as well as agents that target virus infected cells and have the potential to reduce the latent reservoir of HIV-1 infected cells.

GOVERNMENT SUPPORT

[0006] This invention was made with government support under Grant No. UM1AI126619 awarded by National Institute of Allergy and Infectious Diseases. The Government has certain rights to the invention.

SUMMARY OF THE INVENTION

[0007] In some aspects the invention is directed to multispecific molecules, such as but not limited to bispecific and trispecific molecules (e.g. , bispecific antibodies, bispecific diabodies, trivalent binding molecules, etc.) which comprise epitope-binding fragments of antibodies (e.g. , VL and VH Domains) that enable them to coordinately bind immunospecifically to at least one target on HIV-1 envelope (e.g. but not limited to a membrane-proximal extracellar region of HIV-1 gp41 epitope) and at least one epitope of a second molecule that is not HIV-1 Env, for example but not limited to an effector cell which expresses CD2, CD3, CD8, CD 16, T-Cell Receptor (TCR), NKG2D, etc. epitope. In some embodiments, the MPER binding specificity of the molecules of the invention is different from the specificity of the MPER antibody 10E8.

[0008] Selection of the VL and VH Domains of the polypeptide domains of the multispecific molecules of the invention is coordinated so that the polypeptides chains that make up such multispecific molecules assemble to form at least one functional epitope-binding site that is specific for at least one epitope of HIV-1 Env and at least one functional epitope-binding site that is specific for at least one epitope of a molecule that is not HIV-1 Env. In some embodiments, the multispecific molecules of the invention comprise an Fc Domain (Fc bearing multispecific molecules of the invention).

[0009] In certain aspects, the invention provides multispecific molecules comprise an epitope- binding site that specifically binds the membrane-proximal extracellular region (MPER) of HIV-1 gp41. In non-limiting embodiments the multispecific molecules comprise the MPER binding portions from an antibody from Figure 13, Figure 55, Figure 56, Figure 57 or Figures 30-33 (antibodies with mutations in the DH512 or DH51 1 VH chain). In non-limiting embodiments, combination mutations in the DH512 or DH51 1 VHCDR3 could include VH LlOOdF together with TlOOaW Figures 31 and 32); VH LlOOdW together with TlOOaW (Figures 31 and 32). In some embodiments, the MPER binding portions of the multispecific molecule do not comprise portions for the antibody 10E8.

[0010] Non-limiting examples include multispecific molecules comprising VH or VL chains from DH511, DH512, DH512 K3, DH512-L100dF, DH513, DH514, DH515, DH516, DH517, DH518, DH536, DH537, lineage members, chimeric and mutant antibodies with non- limiting embodiments described in Examples 10, 1 1 or 12.

[0011] In non-limiting embodiments, the multispecific molecules comprise 1, 2 or all 3 of the CDRHS of a VH Domain with the specificity of the gp41 MPER binding antibody DH51 1 , DH512, DH512 K3, DH512-L100dF, DH513, DH514, DH515, DH516, DH517, DH518, DH 536, DH537, lineage members, chimeric and mutant antibodies with non-limiting

embodiments described in Example 1 1, and/or 1, 2 or all 3 of the CDRLS of a VL Domain of the gp41 MPER binding antibody DH511, DH512, DH512 K3, DH512-L100dF, DH513, DH514, DH515, DH516, DH517, DH518, DH536, DH537, lineage members, chimeric and mutant antibodies with non-limiting embodiments described in Examples 10, 1 1 or 12.

[0012] In non-limiting embodiments, the multispecific molecules comprise the VH Domain with the specificity of the gp41 MPER binding antibody DH51 1, DH512, DH512 K3, DH512-L100dF, DH513, DH514, DH515, DH516, DH517, DH518, DH536, DH537, lineage members, chimeric and mutant antibodies with non-limiting embodiments described in Example 1 1, and/or the VL Domain, of the gp41 MPER binding antibody DH51 1, DH512, DH512 K3, DH512-L100dF, DH513, DH514, DH515, DH516, DH517, DH518, DH536, DH537, lineage members, chimeric and mutant antibodies with non-limiting embodiments described in Examples 10, 1 1 or 12.

[0013] In certain embodiments an antibody (or a molecule comprising the CDRs, or the variable domains of such antibody) binds specifically to a particular target, peptide, or polysaccharide (such as an antigen present on the surface of a pathogen, for example gpl20, gp41, or CD3), even where the specific epitope may not be known, and do not bind in a significant amount to other proteins or polysaccharides present in the sample or subject. Specific binding can be determined by methods known in the art. Various competitive binding assays are known in the art. With reference to an antibody antigen complex, in certain embodiments specific binding of the antigen and antibody has a KD of less than about 10 6 Molar, such as less than about 10 6 Molar, 10 7 Molar, 10 8 Molar, 10 9 , or even less than about 10 10 Molar.

[0014] In some aspects, the present invention is directed to bispecific molecules, e.g.

covalently linked polypeptide chains to form bispecific antibodies, covalently linked diabodies and/or trivalent binding molecules and their use in the treatment of HIV- 1. In certain aspects, the bispecific molecules of the present invention can bind to two different targets or epitopes on two different cells wherein the first epitope is expressed on a different cell type than the second epitope, such that the bispecific molecules can bring the two cells together. In certain aspects, the bispecific molecules of the present invention can bind to two different cells, wherein the bispecific molecules comprises an arm with the binding specificity for an HIV-1 envelope, for example as provided by the binding specificity of the gp41 MPER binding antibody DH511.2, a variant of DH51 1.2 called DH511 2 K3 and/or other antibodies from the DH51 1 lineage, and/or provided by any of the antibodies DH51 1, DH513, DH514, DH515, DH516, DH517, DH518, DH536, or DH537, which arm binds to the HIV-1 envelope expressed on a first cell, e.g. HIV-1 infected cell, and a second arm with the binding specificity for an epitope expressed on a different cell type than the first cell, such that the bispecific molecules can bring the two cells together. In certain embodiment, the second cell is in effector cell which expresses CD2, CD3, CD8, CD 16, T-Cell Receptor (TCR), NKG2D, etc. epitope.

[0015] In certain aspects, the invention provides a bispecific molecule comprising a first polypeptide chain and a second polypeptide chain, covalently bonded to one another, wherein:

(I) the first polypeptide chain comprises in the N- to C-terminal direction:

(i) a domain (A) comprising a binding region of the light chain variable domain of a first immunoglobulin (VL1) having the binding specificity of a gp41 MPER binding HIV-1 antibody (1);

(ii) a domain (B) comprising a binding region of a heavy chain variable domain of a second immunoglobulin (VH2) specific for an epitope (2); and

(iii) a domain (C) comprising a heterodimer promoting domain;

(II) the second polypeptide chain comprises in the N- to C-terminal direction:

(i) a domain (D) comprising a binding region of a light chain variable domain of the second immunoglobulin (VL2) specific for the epitope (2);

(ii) a domain (E) comprising a binding region of a heavy chain variable domain of the first immunoglobulin (VH1) having the binding specificity of a gp41 MPER binding HIV- 1 antibody (1); and

(iii) a domain (F) comprising a heterodimer promoting domain; and wherein:

the domains (A) and (B) are linked so that they do not associate with one another to form an epitope binding site;

the domains (D) and (E) are linked so that they do not associate with one another to form an epitope binding site; and

the domains (A) and (E) associate to form a binding site that binds the HIV-1 envelope like a gp41 MPER binding HIV-1 antibody (1); and

the domains (B) and (D) associate to form a binding site that binds the epitope (2).

[0016] In certain aspects the invention provides such bispecific molecules wherein:

(i) domains (A) and (B) are separated by a peptide linker 1 ;

(ii) domains (C) and (B) are separated by a peptide linker 2; (iii) domains (D) and (E) are separated by a peptide linker 1 ; and

(iv) domains (F) and (E) are separated by a peptide linker 2.

[0017] In certain aspects the invention provides bispecific molecules comprising a first polypeptide chain and a second polypeptide chain, covalently bonded to one another, wherein:

(I) the first polypeptide chain comprises in the N- to C-terminal direction:

(i) a domain (A) comprising a binding region of the light chain variable domain of a first immunoglobulin (VL1) having the binding specificity of the gp41 MPER binding antibody DH511.2, a variant of DH511.2 called DH511 2 K3, or any antibody of the DH511 lineage (1);

(ii) a domain (B) comprising a binding region of a heavy chain variable domain of a second immunoglobulin (VH2) specific for an epitope (2), wherein domains (A) and (B) are separated from one another by a peptide linker 1 ; and

(iii) a domain (C) comprising a heterodimer promoting domain, including but not limited to a K coil or E coil; wherein domain (C) and domain (B) are separated by a peptide linker

2;

(II) the second polypeptide chain comprises in the N- to C-terminal direction:

(i) a domain (D) comprising a binding region of a light chain variable domain of the second immunoglobulin (VL2) specific for the epitope (2);

(ii) a domain (E) comprising a binding region of a heavy chain variable domain of the first immunoglobulin (VH1) having the binding specificity of the gp41 MPER binding antibody DH511.2, a variant of DH511.2 called DH511 2 K3, or any antibody of the DH511 lineage (1), wherein domains (D) and (E) are separated from one another by a peptide linker 1 ; and

(iii) a domain (F) comprising a heterodimer promoting domain, including but not limited to a K coil or E coil; wherein domain (F) and domain (E) are separated by a peptide linker 2; and wherein:

the domains (A) and (B) do not associate with one another to form an epitope binding site; the domains (D) and (E) do not associate with one another to form an epitope binding site; and

the domains (A) and (E) associate to form a binding site that binds the HIV-1 envelope like the gp41 MPER binding antibody DH511.2, a variant of DH511.2 called DH511 2 K3, or any antibody of the DH51 1 lineage (1); and the domains (B) and (D) associate to form a binding site that binds the epitope (2).

[0018] In certain aspects the invention provides such bispecific molecules, wherein the first or second polypeptide chain further comprises an Fc Domain. The invention also provides such bispecific molecules wherein the first or second polypeptide chain further comprises an Fc Domain and the bispecific molecule further comprises a third polypeptide chain.

[0019] In certain aspects, the invention provides bispecific molecules comprising a first polypeptide chain and a second polypeptide chain, covalently bonded to one another, wherein:

(I) the first polypeptide chain comprises in the N- to C-terminal direction:

(i) a domain (A) comprising SEQ ID NO: 553, 570, 574, 578, 582, 586, 590, 594, 598, 602, 606, or 625;

(ii) a domain (B) comprising SEQ ID NO: 500 or 504; and

(iii) a domain (C) comprising SEQ ID NO: 520, 521, 518, or 519;

(II) the second polypeptide chain comprises in the N- to C-terminal direction:

(i) a domain (D) comprising SEQ ID NO: 502, or 506;

(ii) a domain (E) comprising SEQ ID NO: 551, 568, 572, 576, 580, 584, 588, 592, 596, 600, 604, 608-624, or 626 (Table 4); and

(iii) a domain (F) comprising SEQ ID NO: 518, 519, 520, or 521 ; and wherein:

the domains (A) and (B) are linked so that they do not associate with one another to form an epitope binding site;

the domains (D) and (E) are linked so that they do not associate with one another to form an epitope binding site; and

the domains (A) and (E) associate to form a binding site that binds the HIV-1 envelope like a gp41 MPER binding antibody ( 1); and

the domains (B) and (D) associate to form a binding site that binds an epitope (2).

[0020] In certain aspects the invention provides such bispecific molecules wherein:

(i) domains (A) and (B) are separated by SEQ ID NO: 508;

(ii) domains (C) and (B) are separated by SEQ ID NO: 509 or 510;

(iii) domains (D) and (E) are separated by SEQ ID NO: 508; and

(iv) domains (F) and (E) are separated by SEQ ID NO: 509 or 510. [0021] In certain aspects the invention provides bispecific molecules comprising a first polypeptide chain and a second polypeptide chain, covalently bonded to one another, wherein:

(I) the first polypeptide chain comprises in the N- to C-terminal direction:

(i) a domain (A) comprising SEQ ID NO: 553, 570, 574, 578, 582, 586, 590, 594, 598, 602, 606, or 625;

(ii) a domain (B) comprising SEQ ID NO:500 or 504, wherein domains (A) and (B) are separated from one another by SEQ ID NO: 508; and

(iii) a domain (C) comprising SEQ ID NO: 520, 521, 518, or 519; wherein domain (C) and domain (B) are separated by SEQ ID NO: 509 or 510;

(II) the second polypeptide chain comprises in the N- to C-terminal direction:

(i) a domain (D) comprising SEQ ID NO: 502, or 506;

(ii) a domain (E) comprising SEQ ID NO: 551, 568, 572, 576, 580, 584, 588, 592, 596, 600, 604 608-624, or 626 (Table 4), wherein domains (D) and (E) are separated from one another by SEQ ID NO: 508; and

(iii) a domain (F) comprising SEQ ID NO: 518, 519, 520, or 521; wherein domain (F) and domain (E) are separated by SEQ ID NO: 509 or 510; and wherein:

the domains (A) and (B) do not associate with one another to form an epitope binding site; the domains (D) and (E) do not associate with one another to form an epitope binding site; and

wherein if the domain (A) comprises SEQ ID NO: 553 and domain (E) comprises SEQ ID NO: 551 they associate to form a binding site that binds the HIV-1 envelope like antibody DH511.2 K3;

wherein if the domain (A) comprises SEQ ID NO: 570 and domain (E) comprises SEQ ID NO: 568 they associate to form a binding site that binds the HIV-1 envelope like antibody DH511;

wherein if the domain (A) comprises SEQ ID NO: 574 and domain (E) comprises SEQ ID NO: 572 they associate to form a binding site that binds the HIV-1 envelope like antibody DH512 (also referred to as DH511.2);

wherein if the domain (A) comprises SEQ ID NO: 578 and domain (E) comprises SEQ ID NO: 576 they associate to form a binding site that binds the HIV-1 envelope like antibody DH513; wherein if the domain (A) comprises SEQ ID NO: 582 and domain (E) comprises SEQ ID NO: 580 they associate to form a binding site that binds the HIV-1 envelope like antibody DH514;

wherein if the domain (A) comprises SEQ ID NO: 586 and domain (E) comprises SEQ ID NO: 584 they associate to form a binding site that binds the HIV-1 envelope like antibody DH515;

wherein if the domain (A) comprises SEQ ID NO: 590 and domain (E) comprises SEQ ID NO: 588 they associate to form a binding site that binds the HIV-1 envelope like antibody DH516;

wherein if the domain (A) comprises SEQ ID NO: 594 and domain (E) comprises SEQ ID NO: 592 they associate to form a binding site that binds the HIV-1 envelope like antibody DH517;

wherein if the domain (A) comprises SEQ ID NO: 598 and domain (E) comprises SEQ ID NO: 596 they associate to form a binding site that binds the HIV-1 envelope like antibody DH518;

wherein if the domain (A) comprises SEQ ID NO: 602 and domain (E) comprises SEQ ID NO: 600 they associate to form a binding site that binds the HIV-1 envelope like antibody DH536; or

wherein if the domain (A) comprises SEQ ID NO: 606 and domain (E) comprises SEQ ID NO: 604 they associate to form a binding site that binds the HIV-1 envelope like antibody DH537; or

wherein if the domain (A) comprises SEQ ID NO: 625 and domain (E) comprises SEQ ID NO: 624 or 626 they associate to form a binding site that binds the HIV-1 envelope like antibody DH51 l_5a or DH51 l_5b; and

wherein if the domain (B) comprises SEQ ID NO: 500 and domain (D) comprises SEQ ID NO: 502 they associate to form a binding site that binds CD3; or

wherein if the domain (B) comprises SEQ ID NO: 504 and domain (D) comprises SEQ ID NO: 506 they associate to form a binding site that binds CD 16.

[0022] In certain aspects the invention provides a bispecific molecule comprising a first polypeptide chain, a second polypeptide chain, and a third polypeptide chain, wherein some of the polypeptides are covalently bonded (See Figure 103), and wherein:

(I) the first polypeptide chain comprises in the N- to C-terminal direction: (i) a domain (A) comprising a binding region of the light chain variable domain of a first immunoglobulin (VL1) having the binding specificity of a gp41 MPER binding antibody

(i) ;

(ii) a domain (B) comprising a binding region of a heavy chain variable domain of a second immunoglobulin (VH2) specific for an epitope (2);

(iii) a domain (C) comprising a heterodimer promoting domain; and

(iv) a CH2-CH3 domain;

(II) the second polypeptide chain comprises in the N- to C-terminal direction:

(i) a domain (D) comprising a binding region of a light chain variable domain of the second immunoglobulin (VL2) specific for the epitope (2);

(ii) a domain (E) comprising a binding region of a heavy chain variable domain of the first immunoglobulin (VH1) having the binding specificity of a gp41 MPER binding antibody (l); and

(iii) a domain (F) comprising a heterodimer promoting domain;

(III) the third polypeptide chain comprises in the N- to C-terminal direction:

(i) a CH2-CH3 domain, and wherein:

the domains (A) and (B) are linked so that they do not associate with one another to form an epitope binding site;

the domains (D) and (E) are linked so that they do not associate with one another to form an epitope binding site;

the domains (A) and (E) associate to form a binding site that binds the HIV-1 envelope like a gp41 MPER binding antibody (1);

the domains (B) and (D) associate to form a binding site that binds the epitope (2); and the CH2-CH3 domains of the first and third polypeptide form an Fc Domain.

[0023] In certain aspects the invention provides such bispecific molecules wherein:

(i) the third polypeptide chain further comprises a peptide linker 3 N-terminal to the CH2-CH3 domain;

(ii) domains (A) and (B) are separated by a peptide linker 1;

(iii) domains (C) and (B) are separated by a peptide linker 2;

(iv) the CH2-CH3 domain and domain (C) are separated by a peptide linker 3 or a spacer linker 3;

(v) domains (D) and (E) are separated by a peptide linker 1 ; and (vi) domains (F) and (E) are separated by a peptide linker 2.

[0024] In certain aspects the invention provides bispecific molecules comprising a first polypeptide chain, a second polypeptide chain, and a third polypeptide chain, wherein some of the polypeptides are covalently bonded (See Figure 103), and wherein:

(I) the first polypeptide chain comprises in the N- to C-terminal direction:

(i) a domain (A) comprising a binding region of the light chain variable domain of a first immunoglobulin (VL1) having the binding specificity of the gp41 MPER binding antibody DH511.2, a variant of DH511.2 called DH511 2 K3, or any antibody of the DH511 lineage (1);

(ii) a domain (B) comprising a binding region of a heavy chain variable domain of a second immunoglobulin (VH2) specific for an epitope (2), wherein domains (A) and (B) are separated from one another by a peptide linker 1 ;

(iii) a domain (C) comprising a heterodimer promoting domain, including but not limited to a K coil or E coil; wherein domain (C) and domain (B) are separated by a peptide linker

2;

(iv) a CH2-CH3 domain, wherein the CH2-CH3 domain and domain (C) are separated by a peptide linker 3 or a spacer-linker 3;

(II) the second polypeptide chain comprises in the N- to C-terminal direction:

(i) a domain (D) comprising a binding region of a light chain variable domain of the second immunoglobulin (VL2) specific for the epitope (2);

(ii) a domain (E) comprising a binding region of a heavy chain variable domain of the first immunoglobulin (VH1) having the binding specificity of the gp41 MPER binding antibody DH511.2, a variant of DH511.2 called DH511 2 K3, or any antibody of the DH511 lineage (1), wherein domains (D) and (E) are separated from one another by a peptide linker 1 ; and

(iii) a domain (F) comprising a heterodimer promoting domain, including but not limited to a K coil or E coil; wherein domain (F) and domain (E) are separated by a peptide linker

2;

(III) the third polypeptide chain comprises in the N- to C-terminal direction:

(i) a peptide linker 3,

(ii) a CH2-CH3 domain, and wherein: the domains (A) and (B) do not associate with one another to form an epitope binding site; the domains (D) and (E) do not associate with one another to form an epitope binding site; the domains (A) and (E) associate to form a binding site that binds the HIV-1 envelope like the gp41 MPER binding antibody DH51 1.2, a variant of DH51 1.2 called DH51 1 2 K3, or any antibody of the DH51 1 lineage ( 1);

the domains (B) and (D) associate to form a binding site that binds the epitope (2); and the CH2-CH3 domains of the first and third polypeptide form an Fc Domain.

[0025] In certain aspects said first and second polypeptide chains are covalently bonded to one another; and said first and third polypeptide chains are covalently bonded to one another.

[0026] In certain aspects the invention provides a bispecific molecule comprising a first polypeptide chain, a second polypeptide chain, and a third polypeptide chain, wherein the first and second polypeptide chains are covalently bonded and the first and third polypeptide chains are covalently bonded, and wherein:

(I) the first polypeptide chain comprises in the N- to C-terminal direction:

(i) a domain (A) comprising SEQ ID NO: 553, 570, 574, 578, 582, 586, 590, 594, 598, 602, 606, or 625;

(ii) a domain (B) comprising SEQ ID NO:500 or 504;

(iii) a domain (C) comprising SEQ ID NO: 520, 521, 518, or 519; and

(iv) a CH2-CH3 domain comprising SEQ ID NO: 531, 532, 533, or 534;

(II) the second polypeptide chain comprises in the N- to C-terminal direction:

(i) a domain (D) comprising SEQ ID NO: 502, or 506;

(ii) a domain (E) comprising SEQ ID NO: 551, 568, 572, 576, 580, 584, 588, 592, 596, 600, 604, 608-624, or 626 (Table 4); and

(iii) a domain (F) comprising SEQ ID NO: 518, 519, 520, or 521;

(III) the third polypeptide chain comprises in the N- to C-terminal direction:

(i) a CH2-CH3 domain comprising SEQ ID NO: 531, 532, 533, or 534; and wherein: the domains (A) and (B) are linked so that they do not associate with one another to form an epitope binding site;

the domains (D) and (E) are linked so that they do not associate with one another to form an epitope binding site; the domains (A) and (E) associate to form a binding site that binds the HIV-1 envelope like a gp41 MPER binding antibody (1);

the domains (B) and (D) associate to form a binding site that binds an epitope (2); and the CH2-CH3 domains of the first and third polypeptide form an Fc Domain.

[0027] In certain aspects the invention provides such bispecific molecules wherein:

(i) the third polypeptide chain further comprises SEQ ID NO: 523 N-terminal to the CH2-CH3 domain;

(ii) domains (A) and (B) are separated by SEQ ID NO: 508;

(iii) domains (C) and (B) are separated by SEQ ID NO: 509 or 510;

(iv) the CH2-CH3 domain and domain (C) are separated by SEQ ID NO: 523 or 522;

(v) domains (D) and (E) are separated by SEQ ID NO: 508; and

(vi) domains (F) and (E) are separated by SEQ ID NO: 509 or 510.

[0028] In certain aspects the invention provides bispecific molecules comprising a first polypeptide chain, a second polypeptide chain, and a third polypeptide chain, wherein the first and second polypeptide chains are covalently bonded and the first and third polypeptide chains are covalently bonded, and wherein:

(I) the first polypeptide chain comprises in the N- to C-terminal direction:

(i) a domain (A) comprising SEQ ID NO: 553, 570, 574, 578, 582, 586, 590, 594, 598, 602, 606, or 625;

(ii) a domain (B) comprising SEQ ID NO:500 or 504, wherein domains (A) and (B) are separated SEQ ID NO: 508;

(iii) a domain (C) comprising SEQ ID NO: 513, 520, 51 1, or 518; wherein domain (C) and domain (B) are separated by SEQ ID NO: 509 or 510;

(iv) a CH2-CH3 domain comprising SEQ ID NO: 531, 532, 533, or 534, wherein the CH2-CH3 domain and domain (C) are separated by SEQ ID NO: 523 or 522;

(II) the second polypeptide chain comprises in the N- to C-terminal direction:

(i) a domain (D) comprising SEQ ID NO: 502, or 506;

(ii) a domain (E) comprising SEQ ID NO: 551, 568, 572, 576, 580, 584, 588, 592, 596, 600, 604, 608-624, or 626 (Table 4), wherein domains (D) and (E) are separated from one another by SEQ ID NO: 508; and (iii) a domain (F) comprising SEQ ID NO: 513, 520, 511, or 518; wherein domain (F) and domain (E) are separated by SEQ ID NO: 509 or 510;

(III) the third polypeptide chain comprises in the N- to C-terminal direction:

(i) SEQ ID NO: 523; and

(ii) a CH2-CH3 domain comprising SEQ ID NO: 531, 532, 533, or 534; and wherein: the domains (A) and (B) do not associate with one another to form an epitope binding site; the domains (D) and (E) do not associate with one another to form an epitope binding site; and

wherein if the domain (A) comprises SEQ ID NO: 553 and domain (E) comprises SEQ ID NO: 551 they associate to form a binding site that binds the HIV-1 envelope like antibody DH511.2 K3;

wherein if the domain (A) comprises SEQ ID NO: 570 and domain (E) comprises SEQ ID NO: 568 they associate to form a binding site that binds the HIV-1 envelope like antibody DH511;

wherein if the domain (A) comprises SEQ ID NO: 574 and domain (E) comprises SEQ ID NO: 572 they associate to form a binding site that binds the HIV-1 envelope like antibody DH512 (also referred to as DH511.2);

wherein if the domain (A) comprises SEQ ID NO: 578 and domain (E) comprises SEQ ID NO: 576 they associate to form a binding site that binds the HIV-1 envelope like antibody DH513;

wherein if the domain (A) comprises SEQ ID NO: 582 and domain (E) comprises SEQ ID NO: 580 they associate to form a binding site that binds the HIV-1 envelope like antibody DH514;

wherein if the domain (A) comprises SEQ ID NO: 586 and domain (E) comprises SEQ ID NO: 584 they associate to form a binding site that binds the HIV-1 envelope like antibody DH515;

wherein if the domain (A) comprises SEQ ID NO: 590 and domain (E) comprises SEQ ID NO: 588 they associate to form a binding site that binds the HIV-1 envelope like antibody DH516;

wherein if the domain (A) comprises SEQ ID NO: 594 and domain (E) comprises SEQ ID NO: 592 they associate to form a binding site that binds the HIV-1 envelope like antibody DH517; wherein if the domain (A) comprises SEQ ID NO: 598 and domain (E) comprises SEQ ID NO: 596 they associate to form a binding site that binds the HIV-1 envelope like antibody DH518;

wherein if the domain (A) comprises SEQ ID NO: 602 and domain (E) comprises SEQ ID NO: 600 they associate to form a binding site that binds the HIV-1 envelope like antibody DH536;

wherein if the domain (A) comprises SEQ ID NO: 606 and domain (E) comprises SEQ ID NO: 604 they associate to form a binding site that binds the HIV-1 envelope like antibody DH537; or

wherein if the domain (A) comprises SEQ ID NO: 625 and domain (E) comprises SEQ ID NO: 624 or 626 they associate to form a binding site that binds the HIV-1 envelope like antibody DH51 l_5a or DH51 l_5b; and

wherein if the domain (B) comprises SEQ ID NO: 500 and domain (D) comprises SEQ ID NO: 502 they associate to form a binding site that binds CD3; or

wherein if the domain (B) comprises SEQ ID NO: 504 and domain (D) comprises SEQ ID NO: 506 they associate to form a binding site that binds CD16; and

the CH2-CH3 domains of the first and third polypeptide form an Fc Domain.

[0029] In certain aspects the invention provides bispecific molecules comprising a first polypeptide chain, a second polypeptide chain, and a third polypeptide chain, wherein some of the polypeptides are covalently bonded (See Figure 103), and wherein:

(I) the first polypeptide chain comprises in the N- to C-terminal direction:

(i) a CH2-CH3 domain;

(ii) a domain (A) comprising a binding region of the light chain variable domain of a first immunoglobulin (VL1) having the binding specificity of a gp41 MPERbinding antibody (1);

(iii) a domain (B) comprising a binding region of a heavy chain variable domain of a second immunoglobulin (VH2) specific for an epitope (2); and

(iv) a domain (C) comprising a heterodimer promoting domain;

(II) the second polypeptide chain comprises in the N- to C-terminal direction:

(i) a domain (D) comprising a binding region of a light chain variable domain of the second immunoglobulin (VL2) specific for the epitope (2); (ii) a domain (E) comprising a binding region of a heavy chain variable domain of the first immunoglobulin (VH1) having the binding specificity of a gp41 MPERbinding antibody (1); and

(iii) a domain (F) comprising a heterodimer promoting domain;

(III) the third polypeptide chain comprises in the N- to C-terminal direction:

(i) a CH2-CH3 domain, and wherein:

the domains (A) and (B) are linked so that they do not associate with one another to form an epitope binding site;

the domains (D) and (E) are linked so that they do not associate with one another to form an epitope binding site;

the domains (A) and (E) associate to form a binding site that binds the HIV-1 envelope like a gp41 MPER binding antibody (1); the domains (B) and (D) associate to form a binding site that binds the epitope (2); and

the CH2-CH3 domains of the first and third polypeptide form an Fc Domain.

[0030] In certain aspects the invention provides such bispecific molecules wherein:

(i) the CH2-CH3 domain and domain (A) are separated by a peptide linker 4;

(ii) domains (A) and (B) are separated by a peptide linker 1;

(iii) domains (C) and (B) are separated by a peptide linker 2;

(iv) domains (D) and (E) are separated by a peptide linker 1 ;

(v) domains (F) and (E) are separated by a peptide linker 2;

(vi) the first polypeptide chain further comprises a peptide linker 3 N-terminal to the CH2- CH3 domain; and

(vii) the third polypeptide chain further comprises a peptide linker 3 N-terminal to the CH2-CH3 domain.

[0031] In other aspects the invention provides bispecific molecules comprising a first polypeptide chain, a second polypeptide chain, and a third polypeptide chain, wherein some of the polypeptides are covalently bonded (See Figure 103), and wherein:

(I) the first polypeptide chain comprises in the N- to C-terminal direction:

(i) a peptide linker 3 followed by a CH2-CH3 domain;

(ii) a domain (A) comprising a binding region of the light chain variable domain of a first immunoglobulin (VL1) having the binding specificity of the gp41 MPER binding antibody DH511.2, a variant of DH511.2 called DH511 2 K3, or any antibody of the DH511 lineage (1), wherein the CH2-CH3 domain and domain (A) are separated by a peptide linker 4;

(iii) a domain (B) comprising a binding region of a heavy chain variable domain of a second immunoglobulin (VH2) specific for an epitope (2), wherein domains (A) and (B) are separated by a peptide linker 1 ;

(iv) a domain (C) comprising a heterodimer promoting domain, including but not limited to a K coil or E coil; wherein domain (C) and domain (B) are separated by a peptide linker 2;

(II) the second polypeptide chain comprises in the N- to C-terminal direction:

(i) a domain (D) comprising a binding region of a light chain variable domain of the second immunoglobulin (VL2) specific for the epitope (2);

(ii) a domain (E) comprising a binding region of a heavy chain variable domain of the first immunoglobulin (VH1) having the binding specificity of the gp41 MPER binding antibody DH511.2, a variant of DH511.2 called DH511 2 K3, or any antibody of the DH511 lineage (1), wherein domains (D) and (E) are separated by a peptide linker 1; and

(iii) a domain (F) comprising a heterodimer promoting domain, including but not limited to a K coil or E coil; wherein domain (F) and domain (E) are separated by a peptide linker 2;

(III) the third polypeptide chain comprises in the N- to C-terminal direction:

(i) a peptide linker 3,

(ii) a CH2-CH3 domain, and wherein:

the domains (A) and (B) do not associate with one another to form an epitope binding site; the domains (D) and (E) do not associate with one another to form an epitope binding site; the domains (A) and (E) associate to form a binding site that binds the HIV-1 envelope like the gp41 MPER binding antibody DH511.2, a variant of DH511.2 called DH511 2 K3, or any antibody of the DH511 lineage (1);

the domains (B) and (D) associate to form a binding site that binds the epitope (2); and the CH2-CH3 domains of the first and third polypeptide form an Fc Domain. [0032] In certain aspects said first and second polypeptide chains are covalently bonded to one another; and said first and third polypeptide chains are covalently bonded to one another.

[0033] In certain aspects the invention provides bispecific molecules comprising a first polypeptide chain, a second polypeptide chain, and a third polypeptide chain, wherein the first and second polypeptide chains are covalently bonded and the first and third polypeptide chains are covalently bonded (See Figure 103), and wherein:

(I) the first polypeptide chain comprises in the N- to C-terminal direction:

(i) a CH2-CH3 domain comprising SEQ ID NO: 531, 532, 533, or 534;

(ii) a domain (A) comprising SEQ ID NO: 553, 570, 574, 578, 582, 586, 590, 594, 598, 602, 606, or 625;

(iii) a domain (B) comprising SEQ ID NO:500 or 504; and

(iv) a domain (C) comprising SEQ ID NO: 520, 521, 518, or 519;

(II) the second polypeptide chain comprises in the N- to C-terminal direction:

(i) a domain (D) comprising SEQ ID NO: 502, or 506;

(ii) a domain (E) comprising SEQ ID NO: 551, 568, 572, 576, 580, 584, 588, 592, 596, 600, 604, 608-624, or 626 (Table 4); and

(iii) a domain (F) comprising SEQ ID NO: 518, 519, 520, or 521 ;

(III) the third polypeptide chain comprises in the N- to C-terminal direction:

(i) a CH2-CH3 domain comprising SEQ ID NO: 531, 532, 533, or 534, and wherein: the domains (A) and (B) are linked so that they do not associate with one another to form an epitope binding site;

the domains (D) and (E) are linked so that they do not associate with one another to form an epitope binding site;

the domains (A) and (E) associate to form a binding site that binds the HIV-1 envelope like a gp41 MPER binding antibody (1); the domains (B) and (D) associate to form a binding site that binds an epitope (2); and

the CH2-CH3 domains of the first and third polypeptide form an Fc Domain.

[0034] In certain aspects the invention provides such bispecific molecules wherein:

(i) the CH2-CH3 domain and domain (A) are separated by SEQ ID NO: 524 or 525;

(ii) domains (A) and (B) are separated by SEQ ID NO: 508;

(iii) domains (C) and (B) are separated by SEQ ID NO: 509 or 510; (iv) domains (D) and (E) are separated by SEQ ID NO: 508;

(v) domains (F) and (E) are separated by SEQ ID NO: 509 or 510;

(vi) the first polypeptide chain further comprises SEQ ID NO: 523 N-terminal to the CH2- CH3 domain; and

(vii) the third polypeptide chain further comprises SEQ ID NO: 523 N-terminal to the CH2-CH3 domain.

[0035] In other aspects the invention provides bispecific molecules comprising a first polypeptide chain, a second polypeptide chain, and a third polypeptide chain, wherein the first and second polypeptide chains are covalently bonded and the first and third polypeptide chains are covalently bonded (See Figure 103), and wherein:

(I) the first polypeptide chain comprises in the N- to C-terminal direction:

(i) SEQ ID NO: 523 followed by a CH2-CH3 domain comprising SEQ ID NO: 531, 532, 533, or 534;

(ii) a domain (A) comprising SEQ ID NO: 553, 570, 574, 578, 582, 586, 590, 594, 598, 602, 606, or 625, wherein the CH2-CH3 domain and domain (A) are separated SEQ ID NO: 524 or 525;

(iii) a domain (B) comprising SEQ ID NO: 500 or 504, wherein domains (A) and (B) are separated by SEQ ID NO: 508;

(iv) a domain (C) comprising SEQ ID NO: 520, 521, 518, or 519; wherein domain (C) and domain (B) are separated by SEQ ID NO: 509 or 510;

(II) the second polypeptide chain comprises in the N- to C-terminal direction:

(i) a domain (D) comprising SEQ ID NO: 502, or 506;

(ii) a domain (E) comprising SEQ ID NO: 551, 568, 572, 576, 580, 584, 588, 592, 596, 600, 604, 608-624, or 626 (Table 4), wherein domains (D) and (E) are separated by SEQ ID NO: 508; and

(iii) a domain (F) comprising SEQ ID NO: 518, 519, 520, or 521; wherein domain (F) and domain (E) are separated by SEQ ID NO: 509 or 510;

(III) the third polypeptide chain comprises in the N- to C-terminal direction:

(i) SEQ ID NO: 523,

(ii) a CH2-CH3 domain comprising SEQ ID NO: 531, 532, 533, or 534, and wherein: the domains (A) and (B) do not associate with one another to form an epitope binding site; the domains (D) and (E) do not associate with one another to form an epitope binding site; wherein if the domain (A) comprises SEQ ID NO: 553 and domain (E) comprises SEQ ID NO: 551 they associate to form a binding site that binds the HIV-1 envelope like antibody DH511.2 K3;

wherein if the domain (A) comprises SEQ ID NO: 570 and domain (E) comprises SEQ ID NO: 568 they associate to form a binding site that binds the HIV-1 envelope like antibody DH511;

wherein if the domain (A) comprises SEQ ID NO: 574 and domain (E) comprises SEQ ID NO: 572 they associate to form a binding site that binds the HIV-1 envelope like antibody DH512 (also referred to as DH511.2);

wherein if the domain (A) comprises SEQ ID NO: 578 and domain (E) comprises SEQ ID NO: 576 they associate to form a binding site that binds the HIV-1 envelope like antibody DH513;

wherein if the domain (A) comprises SEQ ID NO: 582 and domain (E) comprises SEQ ID NO: 580 they associate to form a binding site that binds the HIV-1 envelope like antibody DH514;

wherein if the domain (A) comprises SEQ ID NO: 586 and domain (E) comprises SEQ ID NO: 584 they associate to form a binding site that binds the HIV-1 envelope like antibody DH515;

wherein if the domain (A) comprises SEQ ID NO: 590 and domain (E) comprises SEQ ID NO: 588 they associate to form a binding site that binds the HIV-1 envelope like antibody DH516;

wherein if the domain (A) comprises SEQ ID NO: 594 and domain (E) comprises SEQ ID NO: 592 they associate to form a binding site that binds the HIV-1 envelope like antibody DH517;

wherein if the domain (A) comprises SEQ ID NO: 598 and domain (E) comprises SEQ ID NO: 596 they associate to form a binding site that binds the HIV-1 envelope like antibody DH518;

wherein if the domain (A) comprises SEQ ID NO: 602 and domain (E) comprises SEQ ID NO: 600 they associate to form a binding site that binds the HIV-1 envelope like antibody DH536; or wherein if the domain (A) comprises SEQ ID NO: 606 and domain (E) comprises SEQ ID NO: 604 they associate to form a binding site that binds the HIV-1 envelope like antibody DH537; or

wherein if the domain (A) comprises SEQ ID NO: 625 and domain (E) comprises SEQ ID NO: 624 or 626 they associate to form a binding site that binds the HIV-1 envelope like antibody DH51 l_5a or DH51 l_5b; and

wherein if the domain (B) comprises SEQ ID NO: 500 and domain (D) comprises SEQ ID NO: 502 they associate to form a binding site that binds CD3; or

wherein if the domain (B) comprises SEQ ID NO: 504 and domain (D) comprises SEQ ID NO: 506 they associate to form a binding site that binds CD16; and

the CH2-CH3 domains of the first and third polypeptide form an Fc Domain.

[0036] In certain aspects the invention provides a bispecific molecule comprising a first polypeptide chain, a second polypeptide chain, a third polypeptide chain, and a fourth polypeptide chain, wherein some of the polypeptides are covalently bonded, and wherein:

(I) the first and the third polypeptide chains each comprise in the N- to C-terminal direction:

(i) a domain (A) comprising a binding region of the light chain variable domain of a first immunoglobulin (VL1) having the binding specificity of a gp41 MPER binding antibody

(i) ;

(ii) a domain (B) comprising a binding region of a heavy chain variable domain of a second immunoglobulin (VH2) specific for an epitope (2);

(iii) a domain (C) comprising a heterodimer promoting domain; and

(iv) a CH2-CH3 domain;

(II) the second and fourth polypeptide chains each comprise in the N- to C-terminal direction:

(i) a domain (D) comprising a binding region of a light chain variable domain of the second immunoglobulin (VL2) specific for the epitope (2);

(ii) a domain (E) comprising a binding region of a heavy chain variable domain of the first immunoglobulin (VH1) having the binding specificity of a gp41 MPER binding antibody

(i);

(iii) a domain (F) comprising a heterodimer promoting domain; and wherein

the domains (A) and (B) are linked so that they do not associate with one another to form an epitope binding site; the domains (D) and (E) are linked so that they do not associate with one another to form an epitope binding site;

the domains (A) and (E) associate to form a binding site that binds the HIV-1 envelope like a gp41 MPER binding antibody (1);

the domains (B) and (D) associate to form a binding site that binds the epitope (2); and the CH2-CH3 domains of the first and third polypeptide form an Fc Domain.

[0037] In certain aspects the invention provides such bispecific molecules wherein:

(i) domains (A) and (B) are separated by a peptide linker 1 ;

(ii) domains (C) and (B) are separated by a peptide linker 2;

(iii) the CH2-CH3 domain and domain (C) are separated by a peptide linker 3 or a spacer linker 3;

(iv) domains (D) and (E) are separated by a peptide linker 1 ; and

(v) domains (F) and (E) are separated by a peptide linker 2.

[0038] In certain aspects the invention provides a bispecific molecule comprising a first polypeptide chain, a second polypeptide chain, a third polypeptide chain, and a fourth polypeptide chain, wherein some of the polypeptides are covalently bonded, and wherein:

(I) the first and the third polypeptide chains each comprise in the N- to C-terminal direction:

(i) a domain (A) comprising a binding region of the light chain variable domain of a first immunoglobulin (VL1) having the binding specificity of the gp41 MPER binding antibody DH511.2, a variant of DH51 1.2 called DH51 1 2 K3, or any antibody of the DH51 1 lineage (1);

(ii) a domain (B) comprising a binding region of a heavy chain variable domain of a second immunoglobulin (VH2) specific for an epitope (2), wherein domains (A) and (B) are separated by a peptide linker 1;

(iii) a domain (C) comprising a heterodimer promoting domain, wherein domains (C) and (B) are separated by a peptide linker 2; and

(iv) a CH2-CH3 domain, wherein the CH2-CH3 domain and domain (C) are separated by a peptide linker 3 or a spacer linker 3;

(II) the second and fourth polypeptide chains each comprise in the N- to C-terminal direction:

(i) a domain (D) comprising a binding region of a light chain variable domain of the second immunoglobulin (VL2) specific for the epitope (2); (ii) a domain (E) comprising a binding region of a heavy chain variable domain of the first immunoglobulin (VH1) having the binding specificity of the gp41 MPER binding antibody DH51 1.2, a variant of DH51 1.2 called DH51 1 2 K3, or any antibody of the DH51 1 lineage (1), wherein domains (D) and (E) are separated by a peptide linker 1 ;

(iii) a domain (F) comprising a heterodimer promoting domain, wherein domains (F) and (E) are separated by a peptide linker 2; and wherein

the domains (A) and (B) do not associate with one another to form an epitope binding site; the domains (D) and (E) do not associate with one another to form an epitope binding site; the domains (A) and (E) associate to form a binding site that binds the HIV-1 envelope like the gp41 MPER binding antibody DH51 1.2, a variant of DH51 1.2 called DH51 1 2 K3, or any antibody of the DH51 1 lineage ( 1);

the domains (B) and (D) associate to form a binding site that binds the epitope (2); and the CH2-CH3 domains of the first and third polypeptide form an Fc Domain.

[0039] In certain aspects said first and second polypeptide chains are covalently bonded to one another; said third and fourth polypeptide chains are covalently bonded to one another; and said first and third chains are covalently bonded to one another.

[0040] In certain aspects the invention provides a bispecific molecule comprising a first polypeptide chain, a second polypeptide chain, a third polypeptide chain, and a fourth polypeptide chain, wherein the first and second polypeptide chains are covalently bonded, the third and fourth polypeptide chains are covalently bonded, and the first and third chains are covalently bonded, and wherein:

(I) the first and the third polypeptide chains each comprise in the N- to C-terminal direction:

(i) a domain (A) comprising SEQ ID NO: 553, 570, 574, 578, 582, 586, 590, 594, 598, 602, 606, or 625;

(ii) a domain (B) comprising SEQ ID NO: 500 or 504;

(iii) a domain (C) comprising SEQ ID NO: 520, 521, 518, or 519; and

(iv) a CH2-CH3 domain comprising SEQ ID NO: 527, 528, or 529;

(II) the second and fourth polypeptide chains each comprise in the N- to C-terminal direction:

(i) a domain (D) comprising SEQ ID NO: 502, or 506;

(ii) a domain (E) comprising SEQ ID NO: 551, 568, 572, 576, 580, 584, 588, 592, 596, 600, 604, 608-624, or 626 (Table 4); (iii) a domain (F) comprising SEQ ID NO: 518, 519, 520, or 521 ; and wherein the domains (A) and (B) are linked so that they do not associate with one another to form an epitope binding site;

the domains (D) and (E) are linked so that they do not associate with one another to form an epitope binding site;

the domains (A) and (E) associate to form a binding site that binds the HIV-1 envelope like a gp41 MPER binding antibody (1);

the domains (B) and (D) associate to form a binding site that binds an epitope (2); and the CH2-CH3 domains of the first and third polypeptide form an Fc Domain.

[0041] In certain aspects the invention provides such bispecific molecules wherein:

(i) domains (A) and (B) are separated by SEQ ID NO: 508;

(ii) domains (C) and (B) are separated by SEQ ID NO: 509 or 510;

(iii) the CH2-CH3 domain and domain (C) are separated by SEQ ID NO: 523 or 522;

(iv) domains (D) and (E) are separated by SEQ ID NO: 508; and

(v) domains (F) and (E) are separated by SEQ ID NO: 509 or 510.

[0042] In certain aspects the invention provides a bispecific molecule comprising a first polypeptide chain, a second polypeptide chain, a third polypeptide chain, and a fourth polypeptide chain, wherein the first and second polypeptide chains are covalently bonded, the third and fourth polypeptide chains are covalently bonded, and the first and third chains are covalently bonded, and wherein:

(I) the first and the third polypeptide chains each comprise in the N- to C-terminal direction:

(i) a domain (A) comprising SEQ ID NO: 553, 570, 574, 578, 582, 586, 590, 594, 598, 602, 606, or 625;

(ii) a domain (B) comprising SEQ ID NO: 500 or 504, wherein domains (A) and (B) are separated by SEQ ID NO: 508;

(iii) a domain (C) comprising SEQ ID NO: 520, 521, 518, or 519, wherein domains (C) and (B) are separated by SEQ ID NO: 509 or 510; and

(iv) a CH2-CH3 domain comprising SEQ ID NO: 527, 528, or 529, wherein the CH2- CH3 domain and domain (C) are separated by SEQ ID NO: 523 or 522;

(II) the second and fourth polypeptide chains each comprise in the N- to C-terminal direction: (i) a domain (D) comprising SEQ ID NO: 502, or 506;

(ii) a domain (E) comprising SEQ ID NO: 551, 568, 572, 576, 580, 584, 588, 592, 596, 600, 604, 608-624, or 626 (Table 4), wherein domains (D) and (E) are separated by SEQ ID NO: 508;

(iii) a domain (F) comprising SEQ ID NO: 518, 519, 520, or 521, wherein domains (F) and (E) are separated by SEQ ID NO: 509 or 510; and wherein

the domains (A) and (B) do not associate with one another to form an epitope binding site; the domains (D) and (E) do not associate with one another to form an epitope binding site; wherein if the domain (A) comprises SEQ ID NO: 553 and domain (E) comprises SEQ ID NO: 551 they associate to form a binding site that binds the HIV-1 envelope like antibody DH511.2 K3;

wherein if the domain (A) comprises SEQ ID NO: 570 and domain (E) comprises SEQ ID NO: 568 they associate to form a binding site that binds the HIV-1 envelope like antibody DH511;

wherein if the domain (A) comprises SEQ ID NO: 574 and domain (E) comprises SEQ ID NO: 572 they associate to form a binding site that binds the HIV-1 envelope like antibody DH512 (also referred as DH511.2);

wherein if the domain (A) comprises SEQ ID NO: 578 and domain (E) comprises SEQ ID NO: 576 they associate to form a binding site that binds the HIV-1 envelope like antibody DH513;

wherein if the domain (A) comprises SEQ ID NO: 582 and domain (E) comprises SEQ ID NO: 580 they associate to form a binding site that binds the HIV-1 envelope like antibody DH514;

wherein if the domain (A) comprises SEQ ID NO: 586 and domain (E) comprises SEQ ID NO: 584 they associate to form a binding site that binds the HIV-1 envelope like antibody DH515;

wherein if the domain (A) comprises SEQ ID NO: 590 and domain (E) comprises SEQ ID NO: 588 they associate to form a binding site that binds the HIV-1 envelope like antibody DH516;

wherein if the domain (A) comprises SEQ ID NO: 594 and domain (E) comprises SEQ ID NO: 592 they associate to form a binding site that binds the HIV-1 envelope like antibody DH517; wherein if the domain (A) comprises SEQ ID NO: 598 and domain (E) comprises SEQ ID NO: 596 they associate to form a binding site that binds the HIV-1 envelope like antibody DH518;

wherein if the domain (A) comprises SEQ ID NO: 602 and domain (E) comprises SEQ ID NO: 600 they associate to form a binding site that binds the HIV-1 envelope like antibody DH536; or

wherein if the domain (A) comprises SEQ ID NO: 606 and domain (E) comprises SEQ ID NO: 604 they associate to form a binding site that binds the HIV-1 envelope like antibody DH537; or

wherein if the domain (A) comprises SEQ ID NO: 625 and domain (E) comprises SEQ ID NO: 624 or 626 they associate to form a binding site that binds the HIV-1 envelope like antibody DH51 l_5a or DH51 l_5b; and

wherein if the domain (B) comprises SEQ ID NO: 500 and domain (D) comprises SEQ ID NO: 502 they associate to form a binding site that binds CD3; or

wherein if the domain (B) comprises SEQ ID NO: 504 and domain (D) comprises SEQ ID NO: 506 they associate to form a binding site that binds CD16; and

the CH2-CH3 domains of the first and third polypeptide form an Fc Domain.

[0043] In certain aspects the invention provides trivalent binding molecules comprising a first, second, third and fourth polypeptide chain, wherein some of the polypeptides are covalently bonded and wherein:

(I) the first polypeptide chain comprises in the N-terminus to C-terminus direction:

(i) a domain (A) comprising a binding region of a light chain variable domain of a first immunoglobulin (VL1) specific for an epitope (1);

(ii) a domain (B) comprising a binding region of a heavy chain variable domain of a second immunoglobulin (VH2) specific for an epitope (2);

(iii) a domain (C) comprising a heterodimer promoting domain; and

(iv) a CH2-CH3 Domain;

(II) the second polypeptide chain comprises, in the N-terminus to C-terminus direction:

(i) a domain (D) comprising a binding region of a light chain variable domain of the first immunoglobulin (VL1) specific for the epitope (2); (ii) a domain (E) comprising a binding region of a heavy chain variable domain of the second immunoglobulin (VH1) specific for the epitope (1); and

(iii) a domain (F) comprising a heterodimer promoting domain;

(III) the third polypeptide chain that comprises, in the N-terminus to C-terminus direction:

(i) a domain (G) comprising a binding region of a heavy chain variable domain of a third immunoglobulin (VH3) specific for an epitope (3); and

(ii) a CHI -Hinge Domain, and a CH2-CH3 Domain; and

(IV) the fourth polypeptide chain that comprises, in the N-terminus to C-terminus direction:

(i) a domain (H) comprising a binding region of a light chain variable domain of the third immunoglobulin (VL3) specific for the epitope (3); and

(ii) CL Kappa Domain or a CL Lambda Domain; and wherein

the domains (A) and (B) are linked so that they do not associate with one another to form an epitope binding site;

the domains (D) and (E) are linked so that they do not associate with one another to form an epitope binding site;

the domains (A) and (E) associate to form a binding site that binds the epitope (1);

the domains (B) and (D) associate to form a binding site that binds the epitope (2);

the domains (G) and (H) associate to form a binding site that bind the epitope (3);

at least one of epitope (1), epitope (2), and epitope (3) is an epitope bound by a gp41 MPER binding antibody, and at least one of epitope (1), epitope (2), and epitope (3) is an epitope of, for example, but not limited to, CD3, CD8, or CD16, or an epitope on any suitable effector cell;

the CH2-CH3 domains of the first and third polypeptide form an Fc Domain;

said first and second polypeptide chains are covalently bonded to one another;

said first and third polypeptide chains are covalently bonded to one another; and

said third and fourth polypeptide chains are covalently bonded to one another.

[0044] In certain aspects the invention provides such trivalent molecules wherein:

(i) domains (A) and (B) are separated by a peptide linker 1 ;

(ii) domains (C) and (B) are separated by a peptide linker 2 or a peptide linker 2-C;

(iii) the CH2-CH3 domain and domain (C) are separated by a peptide linker 3 or a spacer linker 3; (iv) domains (D) and (E) are separated by a peptide linker 1 ; and

(v) domains (F) and (E) are separated by a peptide linker 2 or a peptide linker 2-C.

[0045] In certain aspects the invention provides trivalent binding molecules comprising a first, second, third and fourth polypeptide chain, wherein some of the polypeptides are covalently bonded and wherein:

(I) the first polypeptide chain comprises in the N-terminus to C-terminus direction:

(i) a domain (A) comprising a binding region of a light chain variable domain of a first immunoglobulin (VL1) specific for an epitope (1);

(ii) a domain (B) comprising a binding region of a heavy chain variable domain of a second immunoglobulin (VH2) specific for an epitope (2), wherein domains (A) and (B) are separated by a Peptide Linker 1;

(iii) a domain (C) comprising:

(a) a heterodimer promoting domain; wherein domain (C) and domain (B) are separated by a Peptide Linker 2-C; or

(b) a heterodimer promoting domain; wherein domain (C) and domain (B) are separated by a Peptide Linker 2; and

(iv) a CH2-CH3 Domain, wherein the CH2-CH3 domain and domain (C) are separated by a Peptide Linker 3 or a Spacer-Linker 3;

(II) the second polypeptide chain comprises, in the N-terminus to C-terminus direction:

(i) a domain (D) comprising a binding region of a light chain variable domain of the first immunoglobulin (VL1) specific for the epitope (2);

(ii) a domain (E) comprising a binding region of a heavy chain variable domain of the second immunoglobulin (VH1) specific for the epitope (1), wherein domains (D) and (E) are separated by a Peptide Linker 1 ;

(iii) a domain (F) comprising

(a) a heterodimer promoting domain; wherein domain (F) and domain (E) are separated by a Peptide Linker 2-C; or

(b) a heterodimer promoting domain; wherein domain (F) and domain (E) are separated by a Peptide Linker 2;

(III) the third polypeptide chain that comprises, in the N-terminus to C-terminus direction: (i) a domain (G) comprising a binding region of a heavy chain variable domain of a third immunoglobulin (VH3) specific for an epitope (3); and

(ii) a CHI -Hinge Domain, and a CH2-CH3 Domain; and

(IV) the fourth polypeptide chain that comprises, in the N-terminus to C-terminus direction:

(i) a domain (H) comprising a binding region of a light chain variable domain of the third immunoglobulin (VL3) specific for the epitope (3); and

(ii) CL Kappa Domain or a CL Lambda Domain; and wherein

the domains (A) and (B) do not associate with one another to form an epitope binding site; the domains (D) and (E) do not associate with one another to form an epitope binding site; the domains (A) and (E) associate to form a binding site that binds the epitope (1);

the domains (B) and (D) associate to form a binding site that binds the epitope (2);

the domains (G) and (H) associate to form a binding site that bind the epitope (3);

at least one of epitope (1), epitope (2), and epitope (3) is an epitope bound by the gp41 MPER binding antibody DH511.2, a variant of DH511.2 called DH511 2 K3, or any antibody of the

DH511 lineage, and at least one of epitope (1), epitope (2), and epitope (3) is an epitope of for example, but not limited to, CD3, CD8, or CD16, or an epitope on any suitable effector cell; the CH2-CH3 domains of the first and third polypeptide form an Fc Domain;

said first and second polypeptide chains are covalently bonded to one another;

said first and third polypeptide chains are covalently bonded to one another; and

said third and fourth polypeptide chains are covalently bonded to one another.

[0046] In certain aspects the invention provides trivalent binding molecules comprising a first, second, third and fourth polypeptide chain, wherein some of the polypeptides are covalently bonded and wherein:

(I) the first polypeptide chain comprises in the N-terminus to C-terminus direction:

(i) a domain (A) comprising SEQ ID NO: 553, 570, 574, 578, 582, 586, 590, 594, 598, 602, 606, or 625;

(ii) a domain (B) comprising SEQ ID NO:500;

(iii) a domain (C) comprising SEQ ID NO: 520, 521, 518, or 519; and

(iv) a CH2-CH3 Domain comprising SEQ ID NO: 531, 532, 533, or 534;

(II) the second polypeptide chain comprises, in the N-terminus to C-terminus direction:

(i) a domain (D) comprising SEQ ID NO: 502; (ii) a domain (E) comprising SEQ ID NO: 551, 568, 572, 576, 580, 584, 588, 592, 596, 600, 604, 608-624, or 626 (Table 4); and

(iii) a domain (F) comprising SEQ ID NO: 518, 519, 520, or 521;

(III) the third polypeptide chain that comprises, in the N-terminus to C-terminus direction:

(i) a domain (G) comprising SEQ ID NO: 551, 568, 572, 576, 580, 584, 588, 592, 596, 600, 604, 608-624, or 626 (Table 4), 543, or 547; and

(ii) a CHI -Hinge Domain comprising SEQ ID NO: 515, and a CH2-CH3 Domain comprising SEQ ID NO: 531, 532, 533, or 534; and

(IV) the fourth polypeptide chain that comprises, in the N-terminus to C-terminus direction:

(i) a domain (H) comprising SEQ ID NO: 553, 570, 574, 578, 582, 586, 590, 594, 598, 602, 606, 545, or 549; and

(ii) CL Kappa Domain comprising SEQ ID NO: 516 or a CL Lambda Domain comprising SEQ ID NO: 517; and wherein

the domains (A) and (B) are linked so that they do not associate with one another to form an epitope binding site;

the domains (D) and (E) are linked so that they do not associate with one another to form an epitope binding site;

the domains (A) and (E) associate to form a binding site that binds an epitope (1);

the domains (B) and (D) associate to form a binding site that binds an epitope (2);

the domains (G) and (H) associate to form a binding site that bind an epitope (3);

wherein epitope (1) is an epitope bound by a gp41 MPER binding antibody, epitope (2) is an epitope of CD3, and epitope (3) is an epitope bound by a gp41 MPER binding antibody or is an epitope of CD 8;

the CH2-CH3 domains of the first and third polypeptide form an Fc Domain;

said first and second polypeptide chains are covalently bonded to one another;

said first and third polypeptide chains are covalently bonded to one another; and

said third and fourth polypeptide chains are covalently bonded to one another.

[0047] In certain aspects the invention provides such trivalent molecules wherein:

(i) domains (A) and (B) are separated by SEQ ID NO: 508;

(ii) domains (C) and (B) are separated by SEQ ID NO: 509 or 510;

(iii) the CH2-CH3 domain and domain (C) are separated by SEQ ID NO: 522, or 523; (iv) domains (D) and (E) are separated by SEQ ID NO: 508; and

(v) domains (F) and (E) are separated by SEQ ID NO: 509 or 510.

[0048] In certain aspects the invention provides trivalent binding molecules comprising a first, second, third and fourth polypeptide chain, wherein some of the polypeptides are covalently bonded and wherein:

(I) the first polypeptide chain comprises in the N-terminus to C-terminus direction:

(i) a domain (A) comprising SEQ ID NO: 553, 570, 574, 578, 582, 586, 590, 594, 598, 602, 606, or 625;

(ii) a domain (B) comprising SEQ ID NO: 500, wherein domains (A) and (B) are separated by SEQ ID NO: 508;

(iii) a domain (C) comprising SEQ ID NO: 520, 521, 518, or 519, wherein domains (C) and (B) are separated by SEQ ID NO: 509 or 510;

(iv) a CH2-CH3 Domain comprising SEQ ID NO: 531, 532, 533, or 534, wherein the CH2-CH3 domain and domain (C) are separated by SEQ ID NO: 522, or 523;

(II) the second polypeptide chain comprises, in the N-terminus to C-terminus direction:

(i) a domain (D) SEQ ID NO: 502;

(ii) a domain (E) comprising SEQ ID NO: 551, 568, 572, 576, 580, 584, 588, 592, 596, 600, 604, 608-624, or 626 (Table 4), wherein domains (D) and (E) are separated from one another by SEQ ID NO: 508;

(iii) a domain (F) comprising SEQ ID NO: 518, 519, 520, or 521, wherein domain (F) and domain (E) are separated by SEQ ID NO: 509 or 510;

(III) the third polypeptide chain that comprises, in the N-terminus to C-terminus direction:

(i) a domain (G) comprising SEQ ID NO: 551, 568, 572, 576, 580, 584, 588, 592, 596, 600, 604, 608-624, or 626 (Table 4), 543, or 547; and

(ii) a CHI -Hinge Domain SEQ ID NO: 515, and a CH2-CH3 Domain comprising SEQ ID NO: 531, 532, 533, or 534; and

(IV) the fourth polypeptide chain that comprises, in the N-terminus to C-terminus direction:

(i) a domain (H) comprising SEQ ID NO: 553, 570, 574, 578, 582, 586, 590, 594, 598, 602, 606, 625, 545, or 549; and

(ii) CL Kappa Domain comprising SEQ ID NO: 516 or a CL Lambda Domain comprising SEQ ID NO: 517; and wherein the domains (A) and (B) do not associate with one another to form an epitope binding site; the domains (D) and (E) do not associate with one another to form an epitope binding site; the domains (A) and (E) associate to form a binding site that binds an epitope (1);

the domains (B) and (D) associate to form a binding site that binds an epitope (2);

the domains (G) and (H) associate to form a binding site that bind an epitope (3);

wherein if the domain (A) comprises SEQ ID NO: 553 and domain (E) comprises SEQ ID NO: 551 they associate to form a binding site that binds the HIV-1 envelope like antibody DH511.2 K3;

wherein if the domain (A) comprises SEQ ID NO: 570 and domain (E) comprises SEQ ID NO: 568 they associate to form a binding site that binds the HIV-1 envelope like antibody DH511;

wherein if the domain (A) comprises SEQ ID NO: 574 and domain (E) comprises SEQ ID NO: 572 they associate to form a binding site that binds the HIV-1 envelope like antibody DH512 (also referred as DH511.2);

wherein if the domain (A) comprises SEQ ID NO: 578 and domain (E) comprises SEQ ID NO: 576 they associate to form a binding site that binds the HIV-1 envelope like antibody DH513;

wherein if the domain (A) comprises SEQ ID NO: 582 and domain (E) comprises SEQ ID NO: 580 they associate to form a binding site that binds the HIV-1 envelope like antibody DH514;

wherein if the domain (A) comprises SEQ ID NO: 586 and domain (E) comprises SEQ ID NO: 584 they associate to form a binding site that binds the HIV-1 envelope like antibody DH515;

wherein if the domain (A) comprises SEQ ID NO: 590 and domain (E) comprises SEQ ID NO: 588 they associate to form a binding site that binds the HIV-1 envelope like antibody DH516;

wherein if the domain (A) comprises SEQ ID NO: 594 and domain (E) comprises SEQ ID NO: 592 they associate to form a binding site that binds the HIV-1 envelope like antibody DH517;

wherein if the domain (A) comprises SEQ ID NO: 598 and domain (E) comprises SEQ ID NO: 596 they associate to form a binding site that binds the HIV-1 envelope like antibody DH518; wherein if the domain (A) comprises SEQ ID NO: 602 and domain (E) comprises SEQ ID NO: 600 they associate to form a binding site that binds the HIV-1 envelope like antibody DH536; or

wherein if the domain (A) comprises SEQ ID NO: 606 and domain (E) comprises SEQ ID NO: 604 they associate to form a binding site that binds the HIV-1 envelope like antibody DH537; or

wherein if the domain (A) comprises SEQ ID NO: 625 and domain (E) comprises SEQ ID NO: 624 or 626 they associate to form a binding site that binds the HIV-1 envelope like antibody DH51 l_5a or DH51 l_5b; and

wherein if the domain (B) comprises SEQ ID NO: 500 and domain (D) comprises SEQ ID NO: 502 they associate to form a binding site that binds CD3; and

wherein if the domain (H) comprises SEQ ID NO: 553 and domain (G) comprises SEQ ID NO: 551 they associate to form a binding site that binds the HIV-1 envelope like antibody DH51 1.2 K3;

wherein if the domain (H) comprises SEQ ID NO: 570 and domain (G) comprises SEQ ID NO: 568 they associate to form a binding site that binds the HIV- 1 envelope like antibody DH51 1 ;

wherein if the domain (H) comprises SEQ ID NO: 574 and domain (G) comprises SEQ ID NO: 572 they associate to form a binding site that binds the HIV-1 envelope like antibody DH512 (also referred as DH51 1.2);

wherein if the domain (H) comprises SEQ ID NO: 578 and domain (G) comprises SEQ ID NO: 576 they associate to form a binding site that binds the HIV-1 envelope like antibody DH513;

wherein if the domain (H) comprises SEQ ID NO: 582 and domain (G) comprises SEQ ID NO: 580 they associate to form a binding site that binds the HIV-1 envelope like antibody DH514;

wherein if the domain (H) comprises SEQ ID NO: 586 and domain (G) comprises SEQ ID NO: 584 they associate to form a binding site that binds the HIV-1 envelope like antibody DH515;

wherein if the domain (H) comprises SEQ ID NO: 590 and domain (G) comprises SEQ ID NO: 588 they associate to form a binding site that binds the HIV-1 envelope like antibody DH516; wherein if the domain (H) comprises SEQ ID NO: 594 and domain (G) comprises SEQ ID NO: 592 they associate to form a binding site that binds the HIV-1 envelope like antibody DH517;

wherein if the domain (H) comprises SEQ ID NO: 598 and domain (G) comprises SEQ ID NO: 596 they associate to form a binding site that binds the HIV-1 envelope like antibody DH518;

wherein if the domain (H) comprises SEQ ID NO: 602 and domain (G) comprises SEQ ID NO: 600 they associate to form a binding site that binds the HIV-1 envelope like antibody DH536; or

wherein if the domain (H) comprises SEQ ID NO: 606 and domain (G) comprises SEQ ID NO: 604 they associate to form a binding site that binds the HIV-1 envelope like antibody DH537; or

wherein if the domain (A) comprises SEQ ID NO: 625 and domain (E) comprises SEQ ID NO: 624 or 626 they associate to form a binding site that binds the HIV-1 envelope like antibody DH51 l_5a or DH51 l_5b; or

wherein if the domain (H) comprises SEQ ID NO: 545 and domain (G) comprises SEQ ID NO: 543 they associate to form a binding site that binds CD8; or

wherein if the domain (H) comprises SEQ ID NO: 549 and domain (G) comprises SEQ ID NO: 547 they associate to form a binding site that binds CD8; and wherein

epitope (1) is an epitope bound by the antibody DH511, DH511 2 K3, DH512 (also referred to as DH511.2), DH513, DH514, DH515, DH516, DH517, DH518, DH536, or DH537, epitope (2) is an epitope of CD3, and epitope (3) is an epitope bound by the antibody DH511, DH511 2 K3, DH512 (also referred to as DH511.2), DH513, DH514, DH515, DH516, DH517, DH518, DH536, or DH537 or is an epitope of CD8; and wherein the CH2-CH3 domains of the first and third polypeptide form an Fc Domain;

said first and second polypeptide chains are covalently bonded to one another;

said first and third polypeptide chains are covalently bonded to one another; and

said third and fourth polypeptide chains are covalently bonded to one another.

[0049] In certain aspects the invention also provides trivalent binding molecules comprising a first, second, and third polypeptide chain, wherein some of the polypeptides are covalently bonded and wherein:

(I) the first polypeptide chain comprises in the N-terminus to C-terminus direction: (i) a domain (A) comprising a binding region of a light chain variable domain of a first immunoglobulin (VL1) specific for an epitope (1);

(ii) a domain (B) comprising a binding region of a heavy chain variable domain of a second immunoglobulin (VH2) specific for an epitope (2);

(iii) a domain (C) comprising a heterodimer promoting domain; and

(iv) a CH2-CH3 Domain;

(II) the second polypeptide chain comprises, in the N-terminus to C-terminus direction:

(i) a domain (D) comprising a binding region of a light chain variable domain of the first immunoglobulin (VL1) specific for the epitope (2);

(ii) a domain (E) comprising a binding region of a heavy chain variable domain of the second immunoglobulin (VH1) specific for the epitope (1);

(iii) a domain (F) comprising a heterodimer promoting domain; and

(III) the third polypeptide chain that comprises, in the N-terminus to C-terminus direction:

(i) a domain (H) comprising a binding region of a light chain variable domain of a third immunoglobulin (VL3) specific for an epitope (3)

(ii) a domain (G) comprising a binding region of a heavy chain variable domain of the third immunoglobulin (VH3) specific for the epitope (3);

(iii) a CH2-CH3 Domain; and wherein

the domains (A) and (B) are linked so that they do not associate with one another to form an epitope binding site;

the domains (D) and (E) are linked so that they do not associate with one another to form an epitope binding site;

the domains (A) and (E) associate to form a binding site that binds the epitope (1);

the domains (B) and (D) associate to form a binding site that binds the epitope (2);

the domains (G) and (H) associate to form a binding site that bind the epitope (3);

at least one of epitope (1), epitope (2), and epitope (3) is an epitope bound by a gp41 MPER antibody, and at least one of epitope (1), epitope (2), and epitope (3) is and epitope of for example, but not limited to, CD3, CD8, or CD16, or any other suitable epitope on an effector cell;

the CH2-CH3 domains of the first and third polypeptide form an Fc Domain;

said first and second polypeptide chains are covalently bonded to one another;

said first and third polypeptide chains are covalently bonded to one another; and said third and fourth polypeptide chains are covalently bonded to one another.

[0050] In certain aspects the invention provides such trivalent molecules wherein:

(i) domains (A) and (B) are separated by a peptide linker 1 ;

(ii) domains (C) and (B) are separated by a peptide linker 2 or a peptide linker 2-C;

(iii) the CH2-CH3 domain and domain (C) are separated by a peptide linker 3 or a spacer linker 3;

(iv) domains (D) and (E) are separated by a peptide linker 1 ;

(v) domains (F) and (E) are separated by a peptide linker 2 or a peptide linker 2-C;

(vi) domains (H) and (G) are separated by a peptide linker 5; and

(vii) the CH2-CH3 domain and domain (G) are separated by a peptide linker 3.

[0051] In certain aspects the invention also provides trivalent binding molecules comprising a first, second, and third polypeptide chain, wherein some of the polypeptides are covalently bonded and wherein:

(I) the first polypeptide chain comprises in the N-terminus to C-terminus direction:

(i) a domain (A) comprising a binding region of a light chain variable domain of a first immunoglobulin (VL1) specific for an epitope (1);

(ii) a domain (B) comprising a binding region of a heavy chain variable domain of a second immunoglobulin (VH2) specific for an epitope (2), wherein domains (A) and (B) are separated by a Peptide Linker 1;

(iii) a domain (C) comprising:

(a) a heterodimer promoting domain; wherein domain (C) and domain (B) are separated by a Peptide Linker 2-C; or

(b) a heterodimer promoting domain; wherein domain (C) and domain (B) are separated by a Peptide Linker 2; and

(iv) a CH2-CH3 Domain, wherein the CH2-CH3 domain and domain (C) are separated by a Peptide Linker 3 or a Spacer-Linker 3;

(II) the second polypeptide chain comprises, in the N-terminus to C-terminus direction:

(i) a domain (D) comprising a binding region of a light chain variable domain of the first immunoglobulin (VL1) specific for the epitope (2); (ii) a domain (E) comprising a binding region of a heavy chain variable domain of the second immunoglobulin (VH1) specific for the epitope (1), wherein domains (D) and (E) are separated by a Peptide Linker 1 ;

(iii) a domain (F) comprising:

(a) a heterodimer promoting domain; wherein domain (F) and domain (E) are separated by a Peptide Linker 2-C; or

(b) a heterodimer promoting domain; wherein domain (F) and domain (E) are separated by a Peptide Linker 2;

(III) the third polypeptide chain that comprises, in the N-terminus to C-terminus direction:

(i) a domain (H) comprising a binding region of a light chain variable domain of a third immunoglobulin (VL3) specific for an epitope (3)

(ii) a domain (G) comprising a binding region of a heavy chain variable domain of the third immunoglobulin (VH3) specific for the epitope (3), wherein domains (H) and (G) are separated by a Peptide Linker 5;

(iii) a Peptide Linker 3; and

(iv) a CH2-CH3 Domain; and wherein

the domains (A) and (B) do not associate with one another to form an epitope binding site; the domains (D) and (E) do not associate with one another to form an epitope binding site; the domains (A) and (E) associate to form a binding site that binds the epitope (1);

the domains (B) and (D) associate to form a binding site that binds the epitope (2);

the domains (G) and (H) associate to form a binding site that bind the epitope (3);

at least one of epitope (1), epitope (2), and epitope (3) is an epitope bound by the gp41 MPER binding antibody DH511.2, a variant of DH511.2 called DH511 2 K3, or any antibody of the

DH511 lineage, and at least one of epitope (1), epitope (2), and epitope (3) is and epitope of for example, but not limited to, CD3, CD8 or CD16, or an epitope on any suitable effector cell;

the CH2-CH3 domains of the first and third polypeptide form an Fc Domain;

said first and second polypeptide chains are covalently bonded to one another; and said first and third polypeptide chains are covalently bonded to one another.

[0052] In certain aspects the invention also provides trivalent binding molecules comprising a first, second, and third polypeptide chain, wherein some of the polypeptides are covalently bonded and wherein: (I) the first polypeptide chain comprises in the N-terminus to C-terminus direction:

(i) a domain (A) comprising SEQ ID NO: 553, 570, 574, 578, 582, 586, 590,594, 598, 602, 606, or 625;

(ii) a domain (B) comprising SEQ ID NO: 500;

(iii) a domain (C) comprising SEQ ID NO: 520, 521, 518, or 519; and

(iv) a CH2-CH3 Domain comprising SEQ ID NO: 531, 532, 533, or 534;

(II) the second polypeptide chain comprises, in the N-terminus to C-terminus direction:

(i) a domain (D) comprising SEQ ID NO: 502;

(ii) a domain (E) comprising SEQ ID NO: 551, 568, 572, 576, 580, 584, 588, 592, 596, 600, 604, 608-624, or 626 (Table 4);

(iii) a domain (F) comprising SEQ ID NO: 518, 519, 520, or 521; and

(III) the third polypeptide chain that comprises, in the N-terminus to C-terminus direction:

(i) a domain (H) comprising SEQ ID NO: 545 or 549;

(ii) a domain (G) comprising SEQ ID NO: 543 or 547;

(iii) a CH2-CH3 Domain comprising SEQ ID NO: 531, 532, 533, or 534; and wherein

the domains (A) and (B) are linked so that they do not associate with one another to form an epitope binding site;

the domains (D) and (E) are linked so that they do not associate with one another to form an epitope binding site;

the domains (A) and (E) associate to form a binding site that binds an epitope bound by a gp41 MPER antibody;

the domains (B) and (D) associate to form a binding site that binds CD3;

the domains (G) and (H) associate to form a binding site that binds CD8; wherein the CH2-CH3 domains of the first and third polypeptide form an Fc Domain;

said first and second polypeptide chains are covalently bonded to one another;

said first and third polypeptide chains are covalently bonded to one another; and

said third and fourth polypeptide chains are covalently bonded to one another.

[0053] In certain aspects the invention provides such trivalent molecules wherein:

(i) domains (A) and (B) are separated by SEQ ID NO: 508;

(ii) domains (C) and (B) are separated by SEQ ID NO: 509 or 510; (iii) the CH2-CH3 domain and domain (C) are separated by SEQ ID NO: 522, or 523;

(iv) domains (D) and (E) are separated by SEQ ID NO: 508;

(v) domains (F) and (E) are separated by SEQ ID NO: 509 or 510;

(vi) domains (H) and (G) are separated by SEQ ID NO: 526; and

(vii) the CH2-CH3 domain and domain (G) are separated by SEQ ID NO: 523 or a CH1- Hinge Domain comprising SEQ ID NO: 515.

[0054] In certain aspects the invention also provides trivalent binding molecules comprising a first, second, and third polypeptide chain, wherein some of the polypeptides are covalently bonded and wherein:

(I) the first polypeptide chain comprises in the N-terminus to C-terminus direction:

(i) a domain (A) comprising SEQ ID NO: 553, 570, 574, 578, 582, 586, 590, 594, 598, 602, 606, or 625;

(ii) a domain (B) comprising SEQ ID NO: 500, wherein domains (A) and (B) are separated by SEQ ID NO: 508;

(iii) a domain (C) comprising SEQ ID NO: 520, 521, 518, and 519, wherein domains (C) and (B) are separated by SEQ ID NO: 509 or 510; and

(iv) a CH2-CH3 Domain comprising SEQ ID NO: 531, 532, 533, or 534, wherein the CH2-CH3 domain and domain (C) are separated by SEQ ID NO: 522, or 523;

(II) the second polypeptide chain comprises, in the N-terminus to C-terminus direction:

(i) a domain (D) comprising SEQ ID NO: 502;

(ii) a domain (E) comprising SEQ ID NO: 551, 568, 572, 576, 580, 584, 588, 592, 596, 600, 604, 608-624, or 626 (Table 4), wherein domains (D) and (E) are separated by SEQ ID NO: 508;

(iii) a domain (F) comprising SEQ ID NO: 518, 519, 520, or 521, wherein domains

(F) and (E) are separated by SEQ ID NO: 509 or 510;

(III) the third polypeptide chain that comprises, in the N-terminus to C-terminus direction:

(i) a domain (H) comprising SEQ ID NO: 545 or 549;

(ii) a domain (G) comprising SEQ ID NO: 543 or 547, wherein domains (H) and

(G) are separated by SEQ ID NO: 526;

(iii) SEQ ID NO: 523, or a CHI-Hinge Domain comprising SEQ ID NO: 515; and

(iv) a CH2-CH3 Domain comprising SEQ ID NO: 531, 532, 533, or 534; and wherein the domains (A) and (B) do not associate with one another to form an epitope binding site; the domains (D) and (E) do not associate with one another to form an epitope binding site; the domains (A) and (E) associate to form a binding site that binds the epitope (1);

the domains (B) and (D) associate to form a binding site that binds CD3;

the domains (G) and (H) associate to form a binding site that binds CD8; and

wherein if the domain (A) comprises SEQ ID NO: 553 and domain (E) comprises SEQ ID NO: 551 they associate to form a binding site that binds the HIV-1 envelope like antibody DH51 1.2 K3;

wherein if the domain (A) comprises SEQ ID NO: 570 and domain (E) comprises SEQ ID NO: 568 they associate to form a binding site that binds the HIV- 1 envelope like antibody DH51 1 ;

wherein if the domain (A) comprises SEQ ID NO: 574 and domain (E) comprises SEQ ID NO: 572 they associate to form a binding site that binds the HIV-1 envelope like antibody DH512 (also referred as DH51 1.2);

wherein if the domain (A) comprises SEQ ID NO: 578 and domain (E) comprises SEQ ID NO: 576 they associate to form a binding site that binds the HIV-1 envelope like antibody DH513;

wherein if the domain (A) comprises SEQ ID NO: 582 and domain (E) comprises SEQ ID NO: 580 they associate to form a binding site that binds the HIV-1 envelope like antibody DH514;

wherein if the domain (A) comprises SEQ ID NO: 586 and domain (E) comprises SEQ ID NO: 584 they associate to form a binding site that binds the HIV-1 envelope like antibody DH515;

wherein if the domain (A) comprises SEQ ID NO: 590 and domain (E) comprises SEQ ID NO: 588 they associate to form a binding site that binds the HIV-1 envelope like antibody DH516;

wherein if the domain (A) comprises SEQ ID NO: 594 and domain (E) comprises SEQ ID NO: 592 they associate to form a binding site that binds the HIV-1 envelope like antibody DH517;

wherein if the domain (A) comprises SEQ ID NO: 598 and domain (E) comprises SEQ ID NO: 596 they associate to form a binding site that binds the HIV-1 envelope like antibody DH518; wherein if the domain (A) comprises SEQ ID NO: 602 and domain (E) comprises SEQ ID NO: 600 they associate to form a binding site that binds the HIV-1 envelope like antibody DH536; or

wherein if the domain (A) comprises SEQ ID NO: 606 and domain (E) comprises SEQ ID NO: 604 they associate to form a binding site that binds the HIV-1 envelope like antibody DH537; or

wherein if the domain (A) comprises SEQ ID NO: 625 and domain (E) comprises SEQ ID NO: 624 or 626 they associate to form a binding site that binds the HIV-1 envelope like antibody DH51 l_5a or DH51 l_5b; and

wherein if the domain (H) comprises SEQ ID NO: 545 and domain (G) comprises SEQ ID NO: 543 they associate to form a binding site that binds CD8; or

wherein if the domain (H) comprises SEQ ID NO: 549 and domain (G) comprises SEQ ID NO: 547 they associate to form a binding site that binds CDS;

wherein the CH2-CH3 domains of the first and third polypeptide form an Fc Domain;

said first and second polypeptide chains are covalently bonded to one another; and said first and third polypeptide chains are covalently bonded to one another.

[0055] In certain aspects the invention provides such trivalent binding molecules wherein one of epitope (1), epitope (2), and epitope (3) is an epitope of HIV-1 Envelope, one of epitope (1), epitope (2), and epitope (3) is an epitope of CD3, and one of epitope (1), epitope (2), and epitope (3) is an epitope of CD8. In particular aspects of such trivalent binding molecules:

(a) epitope (1) is an epitope of HIV-1 Envelope gp41 MPER, epitope (2) is an epitope of CD3, and epitope (3) is an epitope of CD8;

(b) epitope (1) is an epitope of HIV-1 Envelope gp41 MPER, epitope (2) is an epitope of CD8, and epitope (3) is an epitope of CD3;

(c) epitope (1) is an epitope of CD3, epitope (2) is an epitope of HIV-1 envelope gp41 MPER, and epitope (3) is an epitope of CD8;

(d) epitope (1) is an epitope of CD3, epitope (2) is an epitope of CD8, and epitope (3) is an epitope of HIV- 1 Envelope gp41 MPER;

(e) epitope (1) is an epitope of CD8, epitope (2) is an epitope of CD3, and epitope (3) is an epitope of HIV- 1 Envelope gp41 MPER; or

(f) epitope (1) is an epitope of CD8, epitope (2) is an epitope of HIV-1 Envelope gp41 MPER, and epitope (3) is an epitope of CD3. [0056] In certain aspects the invention provides such trivalent binding molecules wherein two of epitope (1), epitope (2), and epitope (3) are epitopes of HIV-1 Envelope, and one of epitope (1), epitope (2), and epitope (3) is an epitope of CD3, where said epitopes of HIV-1 Envelope may be the same epitope or different epitopes. In particular aspects of such trivalent binding molecules:

(a) epitope (1) is an epitope of HIV-1 Envelope gp41 MPER, epitope (2) is an epitope of HIV-1 Envelope gp41 MPER, and epitope (3) is an epitope of CD3;

(b) epitope (1) is an epitope of HIV-1 Envelope gp41 MPER, epitope (2) is an epitope of CD3, and epitope (3) is an epitope of HIV-1 Envelope gp41 MPER; or

(c) epitope (1) is an epitope of CD3, epitope (2) is an epitope of HIV-1 Envelope gp41 MPER, and epitope (3) is an epitope of HIV-1 Envelope gp41 MPER.

[0057] In certain aspects, domain (H) comprises a binding region of a light chain variable domain of an anti-CD 8 antibody, an anti-CD3 antibody, an anti-CD 16 antibody, an HIV-1 envelope gp41 MPER antibody. In certain aspects, domain (G) comprises a binding region of a heavy chain variable domain of an anti-CB8 antibody, an anti-CD3 antibody, an anti-CD 16 antibody, an HIV-1 envelope gp41 MPER antibody.

[0058] In certain aspects, domain (B) comprises the heavy chain variable domain of an anti- CD3 antibody, an anti-CD8 antibody, or an anti-CD 16 antibody. In certain embodiment, domain (D) comprises the light chain variable domain of an anti-CD3 antibody, an anti-CD8 antibody, or an anti-CD 16 antibody.

[0059] In certain aspects, the CH2-CH3 domain of the first polypeptide chain of any of the multispecific molecules of the invention is the of the "knob" design and the CH2-CH3 domain of the third polypeptide chain of any of the multivalent molecules of the invention is of the "hole" design.

[0060] In certain aspects, the CH2-CH3 domain of the third polypeptide chain of any of the multispecific molecules of the invention is the of the "knob" design and the CH2-CH3 domain of the first polypeptide chain of any of the multivalent molecules of the invention is of the "hole" design.

[0061] In certain aspects, the CH2-CH3 domain of the first polypeptide chain is the of the "knob" design (SEQ ID NOs: 531 or 532) and the CH2-CH3 domain of the third polypeptide chain is of the "hole" design (SEQ ID NOs: 533 or 534). In certain aspects, the CH2-CH3 domain of the first polypeptide comprises SEQ ID NO: 531 and the CH2-CH3 domain of the third polypeptide chain comprises SEQ ID NO: 533. In certain aspects, the CH2-CH3 domain of the third polypeptide chain is the of the "knob" design (SEQ ID NOs: 531 or 532) and the CH2-CH3 domain of the first polypeptide chain is of the "hole" design (SEQ ID NOs: 533 or 534). In certain aspects, the CH2-CH3 domain of the third polypeptide comprises SEQ ID NO: 531 and the CH2-CH3 domain of the first polypeptide chain comprises SEQ ID NO: 533.

[0062] In certain aspects, the epitope (2) is a CD3 epitope, CD 8 epitope or a CD 16 epitope. In certain embodiments, the bispecific or trivalent molecule binds HIV-1 envelope with the specificity of the gp41 MPER binding antibody DH51 1.2, a variant of DH51 1.2 called DH51 1.2 K3, or any antibody of the DH51 1 lineage and also binds CD3. In certain embodiments, the bispecific or trivalent molecule binds HIV-1 envelope with the specificity of the gp41 MPER binding antibody DH51 1.2, a variant of DH51 1.2 called DH51 1 2 K3, or any antibody of the DH51 1 lineage and also binds CD3, CD8, or CD 16.

[0063] In certain aspects, domain (A) comprises the CDR1, CDR2, and CDR3 of the light chain variable domain of any one of the immunoglobulin DH51 1, DH512, DH51 1 2 K3, DH513, DH514, DH515, DH516, DH517, DH518, DH536, or DH537, or any of the other antibodies described herein. In certain aspects, the domain (E) comprises the CDR1, CDR2, and CDR3 of the heavy chain variable domain of any one of the immunoglobulin DH51 1, DH512, DH51 1.2 K3, DH513, DH514, DH515, DH516, DH517, DH518, DH536, or DH537, or any of the other antibodies described herein. In certain aspects, domain (A) comprises the light chain variable domain of any one of the immunoglobulin DH511, DH512, DH511 2 K3, DH513, DH514, DH515, DH516, DH517, DH518, DH536, or DH537. In certain aspects, domain (E) comprises the heavy chain variable domain of any one of the immunoglobulin DH51 1, DH512, DH51 1.2 K3, DH513, DH514, DH515, DH516, DH517, DH518, DH536, or DH537.

[0064] In certain aspects, domain (A) comprises the CDR1, CDR2, and CDR3 of the light chain variable domain of immunoglobulin DH51 1.2 or DH51 1 2 K3. In certain aspects, the domain (E) comprises the CDR1, CDR2, and CDR3 of the heavy chain variable domain of immunoglobulin DH51 1.2 or DH51 1.2 K3. In certain aspects, domain (A) comprises the light chain variable domain of immunoglobulin DH51 1.2 or DH51 1 2 K3. In certain aspects, domain (E) comprises the heavy chain variable domain of immunoglobulin DH51 1.2 or DH51 1.2 K3. [0065] In certain aspects, the first polypeptide comprises SEQ ID NO: 555. In certain aspects, the second polypeptide comprises SEQ ID NO: 557. In certain aspects, the third polypeptide comprises SEQ ID NO: 559.

[0066] In certain aspects, the bispecific molecule comprises the first polypeptide of SEQ ID NO: 555, the second polypeptide of SEQ ID NO: 557, and the third polypeptide of SEQ ID NO: 559.

[0067] In certain aspects, the bispecific molecule consists essentially of the first polypeptide of SEQ ID NO: 555, the second polypeptide of SEQ ID NO: 557, and the third polypeptide of SEQ ID NO: 559. In certain aspects, the bispecific molecule consists of the first polypeptide of SEQ ID NO: 555, the second polypeptide of SEQ ID NO: 557, and the third polypeptide of SEQ ID NO: 559.

[0068] In certain aspects, a four chain trivalent binding molecule is a trispecific molecule and comprises the first polypeptide of SEQ ID NO: 555, the second polypeptide of SEQ ID NO: 557, the third polypeptide of SEQ ID NO: 561, the fourth polypeptide of SEQ ID NO: 562 (See Figure 104A).

[0069] In certain aspects, a three chain trivalent binding molecule is a trispecific molecule and comprises the first polypeptide of SEQ ID NO: 555, the second polypeptide of SEQ ID NO: 557, the third polypeptide of SEQ ID NO: 563 (See Figure 104D).

[0070] In certain aspects, the invention provides a composition comprising any one of the multispecific molecules or any combination thereof. In certain aspects, the composition comprises a composition comprising a bispecific molecule comprising a first arm with the binding specificity of a HIV- 1 envelope gp41 MPER binding site antibody and a second arm targeting CD3, CD8, or CD 16. In certain aspects, the bispecific molecule comprises an Fc portion or any other modification which extends its serum half-life. In certain aspects, the composition further comprises a second bispecific molecule or trivalent binding molecule comprising a first arm with an HIV-1 envelope binding specificity different from the HIV-1 binding specificity of the first multispecific molecule, and a second arm targeting CD3, CD8, or CD 16, wherein the first and second multispecific molecules are different in either the HIV-

1 binding specificity and/or the specificity of the second arm. [0071] In certain aspects, the invention provides a method to treat or prevent HIV-1 infection in a subject in need thereof comprising administering to the subject a composition comprising any one of the multispecific molecules of the invention or a combination of any one of the multispecific molecules in a therapeutically effective amount. In certain embodiments, the methods further comprise administering a latency activating agent. In some embodiments, the latency activating agent is vorinostat, romidepsin, panobinostat, disulfiram, JQ 1, bryostatin, PMA, inonomycin, or any combination thereof.

[0072] In certain aspects, the invention provides nucleic acids comprising nucleotides encoding the multispecific molecules of the invention. In certain aspects, the invention provides a vector comprising nucleic acids comprising nucleotides encoding the multispecific molecules of the invention. Provided are also compositions comprising a vector comprising a nucleic acid encoding the multispecific molecules. In certain aspects the invention provides a cell line comprising vectors or nucleic acids encoding the multispecific molecules of the invention, wherein the vectors encode polypeptide chains for expression of the multispecific molecules of the invention, e.g. but not limited to, polypeptide chain 1 and polypeptide chain 2, or polypeptide chain 1, polypeptide chain 2 and polypeptide chain 3. In certain embodiments, the vector is suitable for gene delivery and expression. In certain embodiment, the vector is an adenoviral vector, an adeno associated virus based vector, or a combination thereof.

[0073] In certain embodiments, the multispecific molecule binds to the HIV-1 envelope like the HIV- 1 antibody from which it is derived. In certain embodiments, the multispecific molecule binds to the DH51 1 -HIV-1 envelope epitope, i.e. the multispecific molecule binds to the HIV-1 envelope like the DH511 antibody, and also binds CD3, CD8, or CD 16.

[0074] In certain embodiments a multispecific molecule of the invention comprises, consists essentially of or consists of sequences as described herein, (e.g. , Table 4).

[0075] In certain aspects the invention provides compositions comprising any of the multispecific molecules described herein, or a combination thereof. In certain embodiments, these compositions are formulated as pharmaceutical composition for therapeutic use. [0076] In certain aspects the invention is directed to nucleic acids which encode the multispecific molecule of the invention. In certain embodiments, these nucleic acids are comprised in a vector, and are operably linked to a promoter. In certain aspects the invention provides cell lines, or isolated cells, which comprise nucleic acids for the expression of the multispecific molecule of the invention.

[0077] In certain aspects, the invention provides compositions comprising the multispecific molecule of the invention or nucleic acids encoding the same for use in methods of treating or preventing HIV-1 infection. In some embodiments, these methods further comprise administering a Latency Activating Reagent. Non-limiting examples of these include HDAC inhibitors, e.g., vorinostat, romidepsin, panobinostat, disulfiram, JQ1, bryostatin, PMA, monomycin, or any combination thereof. In some embodiments, this combination therapy targets the pool of latently infected HIV-1 cells.

[0078] In certain aspects, the invention provides methods of treating or preventing an HIV-1 infection in a subject, the method comprising administering to the subject a composition comprising any one of the multispecific molecules the invention, or a combination thereof in a therapeutically sufficient amount. In certain embodiments, the methods further comprise administering a latency activating agent.

BRIEF DESCRIPTION OF THE DRAWINGS

[0079] To conform to the requirements for PCT patent applications, many of the figures presented herein are black and white representations of images originally created in color.

[0080] Figure 1 shows Neutralization-based Epitope Prediction (NEP) Analysis.

Neutralization-based epitope prediction analysis. The predicted relevant prevalence of antibody clusters [(10 epitopes targeting sites of vulnerability (CD4 binding site, V1/V2, MPER, glycan V3)] is shown as a heat map, with dark color intensity (higher fractional number) corresponding to a stronger neutralization signal. Plasma neutralization breadth is shown, and numbers in each row add up to 1.00. NEP algorithm reference: [Georgiev IS et al Science 340: 751-756].

[0081] Figure 2 shows MPR.03 Hook sequence (SEQ ID NOs: 1-2). MPR.03 is a biotinylated peptide containing lysines at both ends for solubility

(KKKNEQELLELDKWASLWT^FDITOWLWYIRKKK-biotin) (SEQ ID NO: 463) used to pull out gp41 antibodies from blood memory B cell sorts. See Morris L. et al. (2011) PLoS ONE 6(9): e23532.

[0082] Figure 3 shows a representative CH0210 mper03 sort (sort #1).

[0083] Figure 4 shows V(D)J Rearrangement of MPER Antibodies Isolated from Four HIV-1 Infected Individuals. * indicates that these mAbs neutralized the tier 1 isolate MN in TZM-bl cells. Mutation refers to VH nucleotide sequence somatic mutation percentages in the variable heavy (VH) immunoglobulin (Ig) genes.

[0084] Figure 5 shows Neutralization Titers of MPER Antibodies Isolated from Four HIV- 1 Infected Individuals using a small panel of HIV- 1 isolates in the TZMbl pseudovirus inhibition assay.

[0085] Figure 6 shows the MPER BnAb DH511 VH Phylogram of the B Cell Clonal Lineage Derived from Subject 0210. Antibodies in clone DH511 include the following: DH511, DH512, DH513, DH514, DH515, DH516 and DH520.

[0086] Figure 7 shows summary results of neutralization of gp41 antibodies against a panel of

30 HIV-1 tier 2 isolates in the TZMbl pseudovirus neutralization assay. Data show that antibodies in the DH511 B cell clonal lineage (DH511-DH516) all neutralize 100% of 30

HIV-1 isolates tested in the TZMbl Env pseudovirus neutralization assay.

[0087] Figure 8 shows Neutralizing Breadth and Potency of DH512, DH517 and DH518

HIV- 1 BnAbs compared to 10E8, VRC01 and a mixture of CH01 and CH31 bnAbs . DH512 neutralizes 100% of HIV strains and is as at least as potent as 10E8.

[0088] Figure 9 shows Neutralizing Breadth and Potency of various HIV-1 BnAbs that are candidates for being combined with DH512 or other antibodies in Figure 4 for a potent mixture of bnAbs. DH270IA1 is II in the DH270 lineage (See Figure 26, and US Ser. No.

62/056,568 filed September 28, 214)

[0089] Figure 10 shows Neutralizing Breadth and Potency of some candidate bnAbs for single or combination use.

[0090] Figure 11 shows summary of Clone DH511 binding to the indicated peptides (SEQ ID NOs: 3-14) in ELISA. Clone DH511 antibodies bind at the C-terminus of the MPER. "+" indicates that antibodies in the Clone DH511 bind to the peptide. The summary shows that DH511 clone antibodies do not bind the peptides when D674 is mutated to S674. The twelve sequences of the peptides (without the three lysines at the N- and C- end) are shown in SEQ ID NOs: 703 to 714. The twelve sequences of the peptides (with the three lysines at the N- and C- end) are shown in SEQ ID NOs: 3 to 14. Thus, antibody DH51 1 requires an aspartic acid at amino acid position 674 for binding.

[0091] Figure 12 shows nucleic acid sequences of antibodies DH511-5 18, DH536 and 537 (SEQ ID Nos: 15 to 34).

[0092] Figure 13 shows amino acid sequences of antibodies DH51 1-518, DH536 and 537. (SEQ ID Nos: 35 to 55)

[0093] Figures 14A-B show Alignment of VH (Fig. 14A; (SEQ ID Nos: 56-61)) and VL (Fig. 14B (SEQ ID Nos: 62-67)) Sequences of BnAb DH51 1 Clonal Lineage. Bolded is the sequence of CDR1, underlined is the sequence of CDR2 and italicized is the sequence of CDR3 of the DH51 1 VH chain and DH51 1 VL chain. The CDRs of the VH and VL sequences of the other antibodies DH512, DH513, DH514, DH515, and DH516 can be readily determined based on the sequence alignment.

[0094] Figures 15A-B show Alignment of VH (Fig. 15A (SEQ ID Nos: 68-76)) and VL (Fig. 15B (SEQ ID Nos: 77-85)) sequences of MPER BnAbs. Bolded is the sequence of CDR1, italicized is the sequence of CDR2 and underlined is the sequence of CDR3 of VH or VL of the listed MPER antibodies.

[0095] Figure 16 shows sequences of MPER alanine mutants (SEQ ID NOs: 86-1 12) screened in ELISA. All antibodies in the DH51 clone showed weak binding to this peptide set. DH517 (Ab510053) strongly bound to MPER656 peptide and showed decreased binding to several residues (A4, A6-A13, A16-A 18, A20, A23, A24, A26) using the ala substituted peptides in table.

[0096] Figure 17 shows Binding of DH517 (Ab510053) to alanine substituted MPER-26 peptides. The binding studies do not conclusively map the DH517epitope.

[0097] Figure 18 shows MPER656 variants (SEQ ID NOs: 1 13-124) screened in ELISA.

Residues shown in light blue (underlined) indicate positions that differ from MPER656- biotin.

[0098] Figure 19 shows Binding of DH51 1 (Ab510056) to MPER656 variants

[0099] Figure 20 shows Binding of DH512 (Ab510049) to MPER656 variants

[0100] Figure 21 shows Binding of DH513 (Ab570022) to MPER656 variants

[0101] Figure 22 shows Binding of DH514 (Ab570029) to MPER656 variants

[0102] Figure 23 shows Binding of DH515 (Ab510052) to MPER656 variants

[0103] Figure 24 shows Binding of DH516 (Ab510048) to MPER656 variants [0104] Figure 25 shows Binding of DH518 (Ab570010) to MPER656 variants.

[0105] Figure 26 shows the amino acids sequences of VH (SEQ ID NOs: 137-148) and VL (SEQ ID NOs: 161-172) chains of antibodies of the DH270 lineage, and nucleic acid sequences (SEQ ID NOs: 125-136 (VH); SEQ ID NOs: 149-160 (VL)) encoding these amino acids. CDRs are highlighted and underlined in the UCA.

[0106] Figure 27A shows amino acid (SEQ ID Nos: 173 and 174) and nucleic acid sequences (SEQ ID Nos: 175 and 176) of CD4bs antibody CH557. Figure 27B shows amino acid sequences of VH chains of antibodies from CH235 lineage (SEQ ID NOs: 177-188). Figure 27C shows amino acid sequences of VL chains of antibodies from CH235 lineage (SEQ ID NOs: 189-198).

[0107] Figure 28A shows neutralization Breadth and Potency of Plasma and Memory B cell (MB C) -derived MPER bnAbs. Figure 28B shows neutralization Breadth and Potency of chimeric MPER bnAbs (n=30 cross-clade HIV-1 isolates)

[0108] Figure 29A and B show neutralization data from TZM-bl assay (Titer in TZM.bl cells (μg/ml) for DH512 K3 and other chimeric antibodies compared to DH512 and 10E8. The data in the first column is historic data when DH512 was run in this panel previously. DH512 was run at the same time as DH512_K3 but is listed as Ab510049 in this assay; therefore, data from columns DH512 K3 and AA&AB DH512/Ab510049 should be compared.

[0109] Figure 30 shows positions in the VHCDR3 chain of DH511 (SEQ ID NO.: 471) which could be mutated. Amino acid positions refer to Kabat numbering. Most mutations are to changes to W, but F, L or possibly other substitutions can be tried.

[0110] Figure 31 shows positions in the VHCDR3 chain of DH512 (SEQ ID NO.: 472) which could be mutated. Amino acid positions refer to Kabat numbering for the DH512VH chain: QVQLVQSGGGLVKPGGSLTLSCSASGFFFDNSWMGWVRQAPGKGLEWVGRIRRLK DGATGEYGAAVKDRFTISRDDSRNMLYLHMRTLKTEDSGTYYCTMDEGTPVTRFLE WGYFYYYMAVWGRGTTVIVSS (SEQ ID NO.: 469). Most mutations are to changes to W, but F, L or possibly other substitutions can also be tried. Position VI 00 can be changed to I. Position LlOOd can be changed to F.

[0111] Figure 32 shows positions outside of VHCDR3 which could be mutated (SEQ ID NOS: 473-478, resepectively, in order of appearance). Most mutations are to changes to W, but F, L or possibly other substitutions can also be tried. [0112] Figure 33 shows amino acid sequences (SEQ ID NOs: 199-216) of some of the DH512 mutants from Figure 31.

[0113] Figure 34 shows neutralization data for a set of 16 mutations from Figure 31. In this figure DH512 is referred to as DH512 (Ab510049_4A): its heavy chain is H510049 4 and its light chain is K510032

[0114] Figure 35 shows summary of anti-cardiolipin activity of various antibodies as measured by QUANTA Lite® ACA IgG III kit. Data plotted are representative of 2 independent experiments. mAb were run in duplicate in the second assay. Mean error and standard deviation are shown. Data were consistent between assays. Dotted line indicates positivity cut-off of 0.18. mAbs with OD values above 0.18 are bolded in the figure legend (DH514, DH518-315 HC, DH51 1-I6-4a through DH511 I1 4A; 4E10).

[0115] Figure 36 shows a summary of self-reactivity data of MPER antibodies.

[0116] Figure 37 shows summary results of neutralization data of DH512 and 10E8 against a panel of HIV-1 isolates in the TZMbl pseudovirus neutralization assay. Values represent IC50 in μg/ml. Figure 37 also shows the mean IC50 and percent of isolates neutralized at different IC50 values.

[0117] Figure 38 shows summary results of neutralization data of DH512 and 10E8 against a panel of HIV-1 isolates in the TZMbl pseudovirus neutralization assay. Values represent IC80 in μg/ml. Figure 38 also shows the mean IC80 and percent of isolates neutralized at different IC80 values.

[0118] Figure 39 shows Experimental Overview of Paired VH-VL Sequencing and antibody identification (Example 10). V gene repertoire sequencing. Identification of individual monoclonal antibodies requires the generation of a sample-specific database of IgG VH sequences constructed by next-generation sequencing of mature B cells isolated from the PBMCs of the donor. Reads are processed bioinformatically to obtain a database of unique VH sequences, which then are clustered into clonotypes according to their CDR3 sequences. The obtained database is used to interpret the MS spectra. F(ab)2 purification and proteomic analysis. F(ab)2 fragments are prepared from total serum IgG and subjected to antigen-affinity chromatography (monomelic gp l20). Proteins in the elution and flow-through are denatured and reduced, alkylated, trypsin-digested and analyzed by high resolution LC-MS/MS. Spectra are interpreted with the sample-specific VH database and peptides uniquely associated with a single CDR3 are used to identify full-length VH sequences. [0119] Figure 40 shows MPER BnAb DH51 1 Clonal Lineage Derived from African

Individual CH0210 (the heavy chain for DH51 1 1A is not included).

[0120] Figure 41 shows Neutralization Activity (IC50) of MPER Antibodies Identified by

Paired VH:VL Sequencing Technology (Example 10). Summary data of two independent assays.

[0121] Figure 42 shows Neutralization Activity (IC80) of MPER Antibodies Identified by Paired VH:VL Sequencing Technology (Example 10). Summary data of two independent assays.

[0122] Figure 43 shows Nucleotide Alignment of MPER Antibody Heavy Chain Sequences (SEQ ID NOs: 217-229).

[0123] Figure 44 shows Amino Acid Alignment of MPER Antibody Heavy Chain Sequences (SEQ ID NOs: 230-242).

[0124] Figure 45 shows Nucleotide Alignment of MPER Antibody Light Chain Sequences (SEQ ID NOs: 243-252).

[0125] Figure 46 shows Amino Acid Alignment of MPER Antibody Light Chain Sequences (SEQ ID NOs: 253-262).

[0126] Figure 47 shows Immunogenetic Characteristics of MPER Antibodies - Original Pairings.

[0127] Figures 48 shows epitope mapping of antibodies of Example 10. Binding to various MPER peptides in an ELISA assay was used to map the epitopes of these MPER antibodies.

[0128] Figures 49 show epitope mapping of antibodies of Example 10. Binding to various MPER peptides in an ELISA assay was used to map the epitopes of these MPER antibodies.

[0129] Figures 50 show epitope mapping of antibodies of Example 10. Binding to various MPER peptides in an ELISA assay was used to map the epitopes of these MPER antibodies.

[0130] Figures 51 show epitope mapping of antibodies of Example 10. Binding to various MPER peptides in an ELISA assay was used to map the epitopes of these MPER antibodies.

[0131] Figures 52 show epitope mapping of antibodies of Example 10. Binding to various MPER peptides in an ELISA assay was used to map the epitopes of these MPER antibodies.

[0132] Figure 53 shows Poly/Autoreactivity analysis of DH51 1_5a. Antibody DH51 1_5a appears to be autoreactive with one protein (NUDC).

[0133] Figure 54 shows Poly/Autoreactivity analysis of DH51 l_5b. Antibody DH51 l_5b appears to be polyreactive. [0134] Figure 55 shows Antibody Pairings - Heavy and Light Chain Chimeric Antibodies from Example 11.

[0135] Figure 56A shows neutralization activity of Heavy and Light Chain Chimeric Antibodies chimeric pairings 1-32 (from Figure 55). Figure 56B shows Neutralization Activity on New Pairings in rows 33-67 (from Figure 55). Figure 56C shows Neutralization Activity on New Pairings in rows 68-91 (from Figure 55). Figure 56D shows that 8 chimeric antibodies were selected for large scale expression and neutralization activity analysis.

[0136] Figure 57 shows nucleic acid and amino acid sequences of VH and VL sequences of antibodies from Example 10 (SEQ ID NOs: 263-300).

[0137] Figure 58 shows sequences of DH51 l_5a and 5b as Fabs (SEQ ID NOs: 301-304).

[0138] Figures 59A-F show isolation of MPER-directed broadly neutralizing antibodies, (a) Fluorescently-labeled MPR.03 peptide tetramers were used to stain peripheral blood mononuclear cells from donor CH0210. A representative flow cytometric plot is shown. Square represents frequency of MPR.03 double positive memory B cells that were single-cell sorted for Ig gene amplification and expression. Colored dots within the square show individual cells that yielded MPER-specific monoclonal antibodies DH511.1-DH511.6 as revealed by index sorting. Memory B cells were gated as live CD16-CD14-CD3-CD235- CD19+IgD-CD38hi. (b) Phylogenetic tree of VHDHJH sequences of the DH511 clonal lineage. Ancestral reconstruction of the evolutionary pathway from the inferred unmutated common ancestor (UCA) to the mature mAbs including 6 maturational intermediates (circles, 11-16) is indicated, (c) Neutralization activity of probe -identified MPER antibodies against a panel of 199 cross-clade HIV-1 isolates. Median and geometric mean neutralization potency against viruses neutralized with a median IC50/IC80<50 μg/ml is indicated. Percentage of 199 viruses neutralized by mAbs DH511.1-DH511.6, 10E8, and VRCOl at IC50<50 μg/ml, IC50<1 μg/ml, and IC50O.1 μg/ml. (d) Neutralization potency and breadth of DH511.2 compared to 10E8 and VRCOl against a 199 HIV-1 Env pseudovirus panel displayed as potency-breadth curves. Percentage of isolates neutralized at IC50 (top panel) and IC80 (bottom panel) values is plotted against mAb concentration, (e) Percent maximum neutralization of each isolate by DH511.2 is shown, (f) Identification of MPER-directed broadly neutralizing plasma antibodies by proteomics. Phylogenetic tree of heavy chain sequences identified in the plasma (black) and in the memory B cell compartment (grey, see Figure 59b). The bar on the right shows the relative abundance of the three identified clonotypes in serum (IV: 95%, II: 4%, III: 1%).

[0139] Figures 60A-E shows structural analysis of the DH51 1 lineage, (a) Ribbon model of crystal structures of DH51 1.1 and DH51 1.2 Fabs in complex with gp41 MPER peptides 656- 683 and 662-683, respectively, oriented based on Ca-atom superposition of distal MPER residues 671-683. (b) Close-up view of antibody-peptide contacts. gp41 residues that interact with antibody VH3-15 region residues, HCDR3 residues, or both, are shown in cyan, red, and brown, respectively, (c) Ribbon model of crystal structures of Fabs of plasma-derived variants DH51 1.1 IP and DH51 1.12P are shown in complex with gp41 MPER peptide 662-683

[51 1.1 IP is placeholder here] . Residues shown in surface representation differ in sequence from DH51 1.1 or DH51 1.2. Of the residues that are unique to DH51 1.1 IP and DH511.12P, those at the interface with gp41 are colored red and are predominantly located within their HCDR3 loops, (d) Close-up view of DH51 1.1 IP and DH51 1.12P antibody-peptide contacts, with gp41 contacting residues colored as in b. (e) Sequence alignment of DH51 1 lineage antibodies (SEQ ID NOs: 305-310), antibody 10E8, and their shared VH3-15 germ line gene precursor. Residues that contact gp41 are labeled with closed circles, and somatically -mutated residues shaded red, orange blue, and green, for 10E8, DH51 1.1, DH51 1.2, and DH51 1.1 IP and DH51 1.12P, respectively.

[0140] Figures 61A-E shows comparison with other MPER-specific antibodies, (a) Crystal structures of DH511.1 and DH51 1.2 Fab in complex with gp41 MPER peptides 656-683 and 662-683, respectively, oriented based on Ca-atom superposition of distal MPER residues 671- 683. (b) Crystal structures of antibodies 10E8 and 4E10 in complex with MPER peptide epitopes, oriented as in (a), (c) Surface representations of antibodies DH51 1.1, DH511.2, and 10E8, colored as in (a) and (b) and rotated by 60°. gp41 contact footprints within the HCDR3 loops are colored red and those within the variable heavy chain VH3- 15 regions are colored green. VH3-15 contacting residues positions that are shared by antibodies DH51 1.1 and DH51 1.2 and antibody 10E8 are colored cyan, (d) Angles of approach to distal gp41 MPER by antibodies DH51 1.1, DH51 1.2, 10E8, and 4E10. Shown is a superposition of the structures of antibody -bound gp41 MPER, with lines representing the longitudinal and latitudinal axes of antibody variable regions colored as in (a) and (b). The longitudinal axis is drawn to the Ca atom of gp41 residue 672 from the center of the latitudinal axis, defined as the point midway between heavy and light chain intra-chain disulfide bonds (spheres). [0141] Figures 62A-C show standard experimental mapping and neutralization-based epitope prediction analysis to delineate the specificities that mediate plasma neutralization breadth, (a) Plasma from donor CH0210 showed potent MPER-directed neutralizing activity against the HIV-2/HIV-1 MPER chimeric pseudovirus C 1C. Neutralization titer is reported as median inhibitory dilution (ID50). (b) Neutralization activity adsorbed with MPER peptide. Anti-MPER antibodies were depleted from plasma using MPER peptide-coated magnetic beads. The depleted fraction was tested for neutralization activity against the indicated heterologous viruses. Neutralization was considerably diminished by removal of anti-MPER from both plasmas, indicating that MPER antibodies were largely responsible for neutralization breadth. ND, not determined, (c) Neutralization-based epitope prediction (NEP) analysis. The predicted relative prevalence of antibody clusters [(10 epitopes targeting sites of vulnerability (CD4 binding site, V1 V2, MPER, glycan V3)] is shown as a heat map, with dark color intensity (higher fractional number) corresponding to a stronger neutralization signal. Plasma neutralization breadth is shown, and numbers in each row add up to 1.00. Shown below are the locations on the Env trimer of the epitopes identified by NEP for this donor and confirmed to be targeted by standard experimental mapping methods.

[0142] Figures 63A-B show frequency and identity of CDR3 peptides from MPER affinity chromatography, (a) Representative histogram of antibody clonotype frequencies identified proteomically in the F(ab)'2 elution and flow through fractions following MPER affinity purification. Clonotypes were defined as genes with the same V- and J- gene usage and >85% sequence identity in the HCDR3. Frequencies of the identified clonotypes were based on the average peak areas of the detected CDR peptides, (b) Identified clonotypes and gene usage (SEQ ID NOs: 31 1-320).

[0143] Figure 64 shows Phylogenetic tree of VHDHJH sequences of memory B cell and plasma-derived DH51 1 clonal lineage members.

[0144] Figure 65A and 65B show Epitope mapping by alanine scanning mutagenesis of C- terminal MPER residues. Values listed are mean measurements from two independent experiments. Epitope residues were defined as residues where log AUC relative to wild-type (WT) for alanine mutations was reduced by 50%.

[0145] Figure 66A-C show Surface-plasmon resonance analysis of binding of the DH511 clonal lineage to MPR.03 peptide. Figure 66C shows Association (ka) and dissociation (kd) rate constants and binding affinities (Kd) for each Fab. [0146] Figure 67A-C show Surface-plasmon resonance analysis of binding of the DH511 clonal lineage to MPER liposomes (SEQ ID NOs: 321-325).

[0147] Figure 68A-C show poly/autoreactivity analysis of MPER bNAbs. Reactivity of DH511 clonal lineage members with self-antigens as measured by indirect

immunofluorescence Hep-2 cell staining (b) and a multiplex bead array anti-nuclear antibody (ANA) assay (a) panel consisting of several autoantigens: SSA, SSB, Smith antigen (Sm), ribonucleoprotein (RNP), Scl-70, Jo-1, double -stranded DNA (dsDNA), Cent B, Histone, and anti-cardiolipin. None of the antibodies were identified as reactive with Hep-2 cells.

DH511.1 UCA reacted with ribonucleoprotein, and DH511 16 reacted with dsDNA. (c) Protein microarrays were used to assess binding to >9400 human proteins. Autoantigens identified: PPP1R1C (protein phosphatase 1, regulatory (inhibitor) subunit 1C) [DH511.1]; FYN (FYN oncogene related to SRC, FGR, YES, transcription variant 1 [DH511.1, DH511.3, DH511.6, DH511 I3, DH511 I4]; NECAP endocytosis associated 1 (NECAP1) [DH511.1, DH11.6]; SIAHBP1 (fuse-binding protein-interacting repressor, transcription variant 1, mRNA) [DH511.1]; STUB 1 (STIP1 homology and U-box containing protein 1) [DH511.2, DH511.6] STIP1 (stress-induced phosphoprotein 1) [DH511 I1, DH511 I2]; OR1F1 (olfactory receptor, family 1, subfamily F, member 1) [DH511]; C6orfl45 (Px-domain containing protein) [DH511.1]; FLJ36032 [DH511 UCA]; TTCl(tetratricopeptide repeat domain 1) [DH511 11], nuclear distribution gene C homolog (A. nidulans) (NUDC)

[DH511.1 IP, DH511.12P], Scm-like with four MBT domains protein 1 [DH511.12P] .

[0148] Figure 69 shows ELISA binding of DH511 lineage members to Ul snRNP components. The DH51 l UCA bound specifically to Ul-snRNPA while no binding was observed to the other components. Results shown represent one experiment.

[0149] Figure 70 shows potential mechanistic differences in binding of 4E10 versus

DH511.2/10E8 to MPER liposomes. 4E10 bound to MPER656.1 in a biphasic

association/dissociation mode and the binding could be fit to a 2-step conformational change model. DH512 appears to have a different mechanistic mode and its binding could be fit to a 1: 1 Langmuir model.

[0150] Figure 71A-C show DH511.2 recognizes a transiently exposed intermediate state of gp41, and the lifetime of DH511.2 epitope exposure is the same as that of 10E8 and 4E10. Time course of neutralization of tier 2 HIV-1 isolate B.BGl 168 was measured by addition of mAbs to TZM-bl cells pre-incubated with virus. Half-life values were similar among the three antibodies.

[0151] Figure 72 shows Sequence Comparison of DH511, DH512, and 10E8 HCDR3 Loops (SEQ ID NOs: 326-328). The figure shows that while HCDR3 loops of DH51 1 and 10E8 lineages are both encoded by D3-3 precursor, substantial differences are observed in their final matured lengths and sequences. One conserved sequence motif between DH51 1/DH512 and 10E8 HCDR3s appears to be a hydrophobic residue doublet at the center of the loop (boxed).

[0152] Figure 73A-D shows Structural Comparison of DH51 1 (A), DH512 (B), and 10E8 (QHCDR3 Loops. Conserved DH51 1/DH512 and 10E8 hydrophobic residue doublets at apex of HCDR3 loops are spatially co-localized (D), relative to MPER. Comparison is based on Ca superposition of MPER residues 671-683.

[0153] Figures 74A-B shows Comparison of DH51 1, DH512, and 10E8 HCDR3 Loops, (a) Sequence alignment of HCDR3 loops of DH51 1, DH512, and 10E8 (SEQ ID NOs: 329-331). (b) Structural comparison of HCDR3 loops based on alignment of distal MPER gp41 residues (that CDRH3 orientation differs from Figure 73). The HCDR3 loops of bNabs that target the gp41 MPER have been shown to be critical for their capacity to neutralize the HIV-1 virus, largely through interactions with the viral membrane. Mutations that reduce hydrophobicity of the HCDR3 loops ablate virus neutralization, while mutations that augment hydrophobicity in turn augment neutralization potency. Given that the DH51 1 lineage shares a common D3-3 gene with 10E8, we sought to compare the sequences and structures of their respective HCDR3 loops to assess whether common characteristics could be discerned. While sequence alignment of their matured amino acid sequences were quite different, as were their lengths, a conserved hydrophobic residue doublet at the centers of both loops was observed. These two residues have previously been shown to be critical for 10E8 epitope binding and

neutralization. Remarkably, despite the overall differences in sequence and length of the DH51 1/12 and 10E8 HCDR3 loops, when they were compared structurally based on an alignment of MPER distal residues, the conserved hydrophobic residue doublets at their tips ended up spatially co-localized relative to MPER. Studies are underway to assess the importance of these two residues in the DH511 context, and the structures are being utilized to introduce additional mutations that are aimed at improving the neutralization potency of DH51 1-lineage antibodies as immunotherapeutics. lHuang, J. et al. Broad and potent neutralization of HIV- 1 by a gp41 -specific human antibody. Nature 491, 406-412, doi: 10.1038/nature l l544 (2012).

[0154] Figure 75 shows sequence characteristics of MPER antibodies isolated from memory B cells (SEQ ID NOs: 332-359). Figure 75 corresponds to Supplementary Table 1 as referenced in Example 12.

[0155] Figure 76 shows neutralization activity of MPER mAbs against a cross-clade 30 isolate HIV-1 Env-pseudovirus panel (IC50 values). Figure 76 corresponds to Supplementary Table 2a as referenced in Example 12.

[0156] Figure 77 shows neutralization activity of MPER mAbs against a cross-clade 30 isolate HIV-1 Env-pseudovirus panel (IC80 values). Figure 77 corresponds to Supplementary Table 2b as referenced in Example 12.

[0157] Figure 78 shows neutralization activity of DH511.2 against a cross-clade 199 isolate HIV-1 Env-pseudovirus panel. Figure 78 corresponds to Supplementary Table 3 as referenced in Example 12.

[0158] Figure 79 shows neutralization activity of DH511.2 against a panel of 200 clade C HIV-1 primary isolates. Figure 79 corresponds to Supplementary Table 4 as referenced in Example 12.

[0159] Figure 80 shows neutralization activity of 16 DH51 1.2 heavy chain mutant antibodies. Figure 80 corresponds to Supplementary Table 27 as referenced in Example 12.

[0160] Figure 81 shows sequence characteristics and pairing of plasma-derived heavy and light chains identified by mass spectrometry and paired VH-VL next-generation sequencing (SEQ ID NOs: 360-367 and 479-489, respectively, in order of appearance). Figure 81 corresponds to Supplementary Table 6 as referenced in Example 12.

[0161] Figure 82 shows neutralization activity of 16 plasma mAbs against a 4 indicator HIV- 1 Env pseudovirus panel. Figure 82 corresponds to Supplementary Table 7 as referenced in Example 12.

[0162] Figure 83 shows neutralization activity of plasma mAbs DH51 1.1 IP and DH51 1.12P against a cross-clade 203 isolate HIV-1 Env-pseudovirus panel. Figure 83 corresponds to Supplementary Table 8 as referenced in Example 12.

[0163] Figure 84 shows sequences of alanine substituted MPR.03 peptides (SEQ ID NOs: 368-381). Figure 84 corresponds to Supplementary Table 9 as referenced in Example 12. [0164] Figure 85 shows sequences of COT6.15 MPER mutant viruses (SEQ ID NOs: 382- 403). Figure 85 corresponds to Supplementary Table 10 as referenced in Example 12.

[0165] Figure 86 shows neutralization Activity Against a series of MPER alanine mutant pseudoviruses in the COT6.15 Env background. Figure 86 corresponds to Supplementary Table 11 as referenced in Example 12.

[0166] Figure 87 shows crystallization peptides (SEQ ID NOs: 404-406). Figure 87 corresponds to Supplementary Table 12 as referenced in Example 12.

[0167] Figure 88 shows crystallographic data collection and refinement statistics. Figure 88 corresponds to Supplementary Table 13 as referenced in Example 12.

[0168] Figure 89 shows antibody contact interfaces by CDR loop. Figure 89 corresponds to

Supplementary Table 14 as referenced in Example 12.

[0169] Figure 90 shows bonded and non-bonded contacts DH511.1-MPER. (Non-Kabat numbering). Figure 90 corresponds to Supplementary Table 15 as referenced in Example 12.

[0170] Figure 91 shows bonded and non-bonded contacts DH511.2-MPER. (Non-Kabat numbering). Figure 91 corresponds to Supplementary Table 16 as referenced in Example 12.

[0171] Figure 92 shows bonded and non-bonded contacts DH511.11P-MPER. Figure 92 corresponds to Supplementary Table 17 as referenced in Example 12.

[0172] Figure 93 shows bonded and non-bonded contacts DH511.12P-MPER. (Non-Kabat numbering). Figure 93 corresponds to Supplementary Table 18 as referenced in Example 12.

[0173] Figure 94 shows neutralization of the DH511 clonal lineage against a panel of 12 global HIV-1 reference strains. Figure 94 corresponds to Supplementary Table 19 as referenced in Example 12.

[0174] Figures 95A-C show primers and PCR conditions for paired VH:VL NGS. Figure 95 A shows overlap extension oligonucleotides for framework region 1 (5' - 3') (SEQ ID NOs: 407-427). Figure 95B shows overlap extension oligonucleotides for leader peptide (5' - 3') (SEQ ID NOs: 428-441). Figure 95C shows nested constant region oligonucleotides (5' - 3') (SEQ ID NOs: 442-446). Figure 95 A corresponds to Supplementary Table 28 as referenced in Example 12. Figure 95B corresponds to Supplementary Table 29 as referenced in Example 12. Figure 95C corresponds to Supplementary Table 30 as referenced in Example 12.

[0175] Figure 96 shows DH511 clonal lineage membrane insertion scores and HCDR3 analysis (SEQ ID NOs: 447-455). The membrane insertion scores can be recalculated to exclude the C in the CDR3. HCDR3s score for the .P antibodies will be calculated. Figure 96 corresponds to Supplementary Table 21 as referenced in Example 12.

[0176] Figure 97 shows cardiolipin reactivity of the DH511 clonal lineage. Figure 97 corresponds to Supplementary Table 22 as referenced in Example 12.

[0177] Figure 98 shows neutralization activity of 91 chimeric MPER mAbs against the tier 2 HIV-1 isolate B.BG1168. Figure 98 corresponds to Supplementary Table 23 as referenced in Example 12.

[0178] Figure 99 shows neutralization activity of chimeric mAb DH511.2 K3 against a cross- clade 30 isolate Env-pseudovirus panel. Figure 99 corresponds to Supplementary Table 24 as referenced in Example 12.

[0179] Figures lOOA-C show primers and PCR conditions for paired VH:VL NGS. Figure 100A shows PCR conditions for isotype specific amplification. Figure 100B shows oligonucleotides for isotype specific amplification (5' - 3') (SEQ ID NOs: 456-462). Figure lOOC shows PCR conditions for MiSeq Barcoding. Figure 100A corresponds to

Supplementary Table 30 as referenced in Example 12. Figure 100B corresponds to

Supplementary Table 31 as referenced in Example 12. Figure lOOC corresponds to

Supplementary Table 32 as referenced in Example 12.

[0180] Figure 101 provides a schematic of a representative covalently bonded diabody having two epitope-binding sites composed of two polypeptide chains, each having an E-coil or K-coil Heterodimer-Promoting Domain (alternative Heterodimer-Promoting Domains are provided below). A cysteine residue may be present in a linker and/or in the Heterodimer- Promoting Domain as shown in Figure 102B. VL and VH Domains that recognize the same epitope are shown using the same shading or fill pattern.

[0181] Figures 102A-102C provide schematics showing representative covalently bonded tetravalent diabodies having four epitope-binding sites composed of two heterodimer pairs of polypeptide chains (i.e., four polypeptide chains in all). One polypeptide of each heterodimer pair possesses a CH2 and CH3 Domain, such that the associated chains form all or part of an Fc Domain. VL and VH Domains that recognize the same epitope are shown using the same shading or fill pattern. The two pairs of polypeptide chains may be the same. In such embodiments wherein the two pairs of polypeptide chains are the same and the VL and VH Domains recognize different epitopes (as shown in Figures 102A-102B), the resulting molecule possesses four epitope-binding sites and is bispecific and bivalent with respect to each bound epitope. In such embodiments wherein the VL and VH Domains recognize the same epitope (e.g., the same VL Domain CDRs and the same VH Domain CDRs are used on all chains) the resulting molecule possesses four epitope-binding sites and is monospecific and tetravalent with respect to a single epitope. Alternatively, the two pairs of polypeptides may be different. In such embodiments wherein the two pairs of polypeptide chains are different and the VL and VH Domains of each pair of polypeptides recognize different epitopes (as shown by the different shading and patterns in Figure 102C), the resulting molecule possesses four epitope-binding sites and is tetraspecific and monovalent with respect to each bound epitope. Figure 102A shows an Fc Domain-containing diabody which contains a peptide Heterodimer-Promoting Domain comprising a cysteine residue. Figure 102B shows an Fc Domain-containing diabody, which contains E-coil and K-coil

Heterodimer-Promoting Domains comprising a cysteine residue and a linker (with an optional cysteine residue). Figure 102C, shows an Fc-Region-Containing diabody, which contains antibody CHI and CL domains which could serve as Heterodimer Promoting Domains.

[0182] Figures 103A and 103B provide schematics of a representative covalently bonded Fc bearing diabody molecule having two epitope-binding sites composed of three polypeptide chains. Two of the polypeptide chains comprise a CH2 and CH3 Domain, such that the associated chains form all or part of an Fc Domain. The polypeptide chains comprising the VL and VH Domain further comprise a Heterodimer-Promoting Domain. VL and VH Domains that recognize the same epitope are shown using the same shading or fill pattern.

[0183] Figures 104A-104F provide schematics of representative Fc Domain-containing trivalent binding molecules having three epitope-binding sites. Figures 104A and 104B, respectively, illustrate schematically the domains of trivalent binding molecules comprising two diabody-type binding domains and a Fab-type binding domain having different domain orientations in which the diabody-type binding domains are N-terminal or C-terminal to an Fc Domain. The molecules in Figures 104A and 104B comprise four chains. Figures 104C and 104D, respectively, illustrate schematically the domains of trivalent binding molecules comprising two diabody-type binding domains N-terminal to an Fc Domain, and a Fab-type binding domain in which the light chain and heavy chain are linked via a polypeptide spacer, or an scFv-type binding domain. The trivalent binding molecules in Figures 104E and 104F, respectively, illustrate schematically the domains of trivalent binding molecules comprising two diabody-type binding domains C-terminal to an Fc Domain, and a Fab-type binding domain in which the light chain and heavy chain are linked via a polypeptide spacer, or an scFv-type binding domain. The trivalent binding molecules in Figures 104C-104F comprise three chains. VL and VH Domains that recognize the same epitope are shown using the same shading or fill pattern.

[0184] Figures 105A-105B provide schematics of a representative HIVxCD3 bispecific monovalent diabody comprising three polypeptide chains. Figure 105A shows the domains of each of the three polypeptide chains, dashed lines represent disulfide bonds which form between the chains, and the arrows indicate the interactions of the Variable Domains. Figure 105B provides a schematic of the assembled chains. Such diabodies contain an anti-HIV-1 binding arm (e.g. , DH511, DH51 1.2 K3, DH512, DH513, DH514, DH515, DH516, DH517, DH518, DH536, or DH537) combined with an anti-CD3 binding arm (e.g., hXR32). They are composed of two polypeptide chains: one with the VL of an anti-CD3 antibody linked to the VH of an anti-HIV- 1 antibody; the second with the VL of an anti-HIV-1 antibody linked to the VH of an anti-CD3 antibody. The first and the second polypeptide chains are linked by interchain disulfide bond and paired via oppositely charged E-coil/K-coil Heterodimer- Promoting Domains. The amino acid and nucleotide sequences of Chain 1, 2 and 3 which form the bispecific monovalent diabody designated DH511 2_K3xCD3 Fc are provided in Table 4 (SEQ ID NOs: 555-560). Control molecules have one of the arms replaced by a non- HIV-1 envelope binding arm derived, for example, from an anti-FITC antibody (4420) or from an anti-RSV antibody (palivizumab).

DETAILED DESCRIPTION

[0185] Highly active anti-retroviral therapy (HAART) alone or in combination with latency reversing agents fails to reduce the pool of latently infected cells. This is due to limited ability of the CD8+ T cells to eliminate HIV-1 latently infected cells. Dual Affinity Re-Targeting proteins (DARTs) are multispecific, antibody-based diabody molecules that can bind at least two distinct antigens simultaneously. HIV-1 diabodies contain an HIV- 1 binding arm combined with an effector cell binding arm (e.g., but not limited to CD3 effector cells) are designed to redirect effector cells (e.g. but not limited to cytotoxic CD3+ T cells) to engage and kill HIV-1 -infected cells. Additionally, multispecific molecules such as trivalent binding molecules containing one or more HIV-1 binding arms combined with one or more effector cell binding arms, may also be designed to redirected effector cells to engage and kill HIV-1- infected cells.

[0186] The provision of multispecific/non-mono-specific molecules provides a significant advantage over typical mono-specific antibodies: the capacity to co-ligate and co-localize cells that express different epitopes. Bivalent diabodies have wide-ranging applications including therapy and immunodiagnosis. Bi-valency allows for great flexibility in the design and engineering of the diabody in various applications, providing enhanced avidity to multimeric antigens, the cross-linking of differing antigens, and directed targeting to specific cell types relying on the presence of both target antigens. Due to their increased valency, low dissociation rates and rapid clearance from the circulation (for diabodies of small size, at or below -50 kDa), diabody molecules known in the art have also shown particular use in the field of tumor imaging (Fitzgerald et al. (1997) "Improved Tumour Targeting By Disulphide Stabilized Diabodies Expressed In Pichia pastoris, " Protein Eng. 10: 1221). Of particular importance is the co-ligating of differing cells, for example, the cross-linking of cytotoxic T cells to tumor cells (Staerz et al. (1985) "Hybrid Antibodies Can Target Sites For Attack By T Cells, " Nature 314:628-631, and Holliger et al. ( 1996) "Specific Killing Of Lymphoma Cells By Cytotoxic T-Ce lis Mediated By A Bispecific Diabody, " Protein Eng. 9:299-305).

[0187] Diabody epitope binding domains may also be directed to a surface determinant of a B cell, such as CD 19, CD20, CD22, CD30, CD37, CD40, and CD74 (Moore, P.A. et al. (201 1) 'Application Of Dual Affinity Retargeting Molecules To Achieve Optimal Redirected T-Cell Killing Of ' B-Cell Lymphoma " Blood 1 17( 17):4542-4551; Cheson, B.D. et al. (2008) "Monoclonal Antibody Therapy For B-Cell Non-Hodgkin 's Lymphoma " N. Engl. J. Med. 359(6):613-626; Castillo, J. et al. (2008) "Newer monoclonal antibodies for hematological malignancies," Exp. Hematol. 36(7):755-768. In many studies, diabody binding to effector cell determinants, e.g. , Fey receptors (FcyR), was also found to activate the effector cell (Holliger et al. ( 1996) "Specific Killing Of Lymphoma Cells By Cytotoxic T -Cells Mediated By A Bispecific Diabody, " Protein Eng. 9:299-305; Holliger et al. (1999) "Carcinoembryonic Antigen (CE A) -Specific T -Cell Activation In Colon Carcinoma Induced By Anti-CD 3 x Anti- CEA Bispecific Diabodies And B7 x Anti-CEA Bispecific Fusion Proteins, " Cancer Res. 59:2909-2916; WO 2006/1 13665; WO 2008/157379; WO 2010/080538; WO 2012/018687; WO 2012/162068). Normally, effector cell activation is triggered by the binding of an antigen bound antibody to an effector cell via Fc-FcyR interaction; thus, in this regard, diabody molecules may exhibit Ig-like functionality independent of whether they comprise an Fc Domain (e.g., as assayed in any effector function assay known in the art or exemplified herein (e.g. , ADCC assay)). By cross-linking tumor and effector cells, the diabody not only brings the effector cell within the proximity of the tumor cells but leads to effective tumor killing (see e.g. , Cao et al. (2003) "Bispecific Antibody Conjugates In Therapeutics, " Adv. Drug. Deliv. Rev. 55 : 171-197).

[0188] The formation of such non-mono-specific diabodies requires the successful assembly of two or more distinct and different polypeptides (i.e. , such formation requires that the diabodies be formed through the heterodimerization of different polypeptide chain species). This fact is in contrast to mono-specific diabodies, which are formed through the homodimerization of identical polypeptide chains. Because at least two dissimilar polypeptides (/ ' . e. , two polypeptide species) must be provided in order to form a non-mono- specific diabody, and because homodimerization of such polypeptides leads to inactive molecules (Takemura, S. et al. (2000) "Construction Of A Diabody (Small Recombinant Bispecific Antibody) Using A Refolding System, " Protein Eng. 13(8):583-588), the production of such polypeptides must be accomplished in such a way as to prevent covalent bonding between polypeptides of the same species (i. e. , so as to prevent homodimerization)

(Takemura, S. et al. (2000) "Construction Of A Diabody (Small Recombinant Bispecific Antibody) Using A Refolding System, " Protein Eng. 13(8):583-588). The art has therefore taught the non-covalent association of such polypeptides (see, e.g. , Olafsen et al. (2004) "Covalent Disulfide-Linked Anti-CEA Diabody Allows Site-Specific Conjugation And Radiolabeling For Tumor Targeting Applications, " Prot. Engr. Des. Sel. 17:21-27; Asano et al. (2004) "A Diabody For Cancer Immunotherapy And Its Functional Enhancement By Fusion Of Human Fc Domain," Abstract 3P-683, J. Biochem. 76(8):992; Takemura, S. et al. (2000) "Construction Of A Diabody (Small Recombinant Bispecific Antibody) Using A Refolding System, " Protein Eng. 13(8):583-588; Lu, D. et al. (2005) "A Fully Human Recombinant IgG-Like Bispecific Antibody To Both The Epidermal Growth Factor Receptor And The Insulin-Like Growth Factor Receptor For Enhanced Antitumor Activity," J. Biol. Chem. 280(20): 19665-19672).

[0189] The art has recognized that bispecific diabodies composed of non-covalently associated polypeptides are unstable and readily dissociate into non-functional monomers (see, e.g. , Lu, D. et al. (2005) "A Fully Human Recombinant IgG-Like Bispecific Antibody To Both The Epidermal Growth Factor Receptor And The Insulin-Like Growth Factor Receptor For Enhanced Antitumor Activity " i. Biol. Chem. 280(20): 19665-19672).

[0190] In the face of this challenge, the invention provides stable, covalently bonded heterodimeric multispecific diabodies, termed DARTs™ (see, e.g. , United States Patent Publications No. 2014-0099318; 2013-0295121 ; 2010-0174053 and 2009-0060910; European Patent Publication No. EP 2714079; EP 2601216; EP 2376109; EP 2158221 and PCT Publications No. WO 2015/026894; WO2015/026892; WO 2015/021089; WO 2014/159940; WO 2012/162068; WO 2012/018687; WO 2010/080538; Moore, P.A. et al. (2011)

'Application Of Dual Affinity Retargeting Molecules To Achieve Optimal Redirected T-Cell Killing Of B-Cell Lymphoma;' Blood 1 17( 17):4542-4551 ; Veri, M.C. et al. (2010)

"Therapeutic Control OfB Cell Activation Via Recruitment OfFcgamma Receptor lib (CD32B) Inhibitory Function With A Novel Bispecific Antibody Scaffold " Arthritis Rheum. 62(7): 1933-1943; Johnson, S. et al. (2010) "Effector Cell Recruitment With Novel Fv-Based Dual-Affinity Re-Targeting Protein Leads To Potent Tumor Cytolysis And in vivo B-Cell Depletion " J. Mol. Biol. 399(3):436-449), the contents of which publications are herein incorporated by reference in their entirety). Such multispecific molecules comprise two or more covalently complexed polypeptides and involve engineering one or more cysteine residues into each of the employed polypeptide species. For example, the addition of a cysteine residue to the C-terminus of such constructs has been shown to allow disulfide bonding between the polypeptide chains, stabilizing the resulting heterodimer without interfering with the binding characteristics of the bivalent molecule.

[0191] The invention provides multispecific, antibody-based molecules that can bind at least two distinct antigens simultaneously, wherein at least one of the antigens is comprised in an HIV-1 envelope. In certain aspects, the present invention is directed to HIV- 1 multispecific molecules that are capable of simultaneous binding to an epitope of HIV- 1 envelope and an epitope of an antigen on a number of effector cells, e.g. but not limited to an effector cell which expresses CD2, CD3, CD8, CD 16, T-Cell Receptor (TCR), NKG2D, etc. epitope) and to the uses of such molecules in the treatment of HIV-1 infection.

[0192] In certain embodiments the invention provides molecules with dual targeting specificity (including but not limited to bispecific antibodies, bispecific diabodies, and trivalent binding molecules). In certain aspects the invention provides bispecific molecules that are capable of localizing an immune effector cell to an HIV-1 envelope expressing cell, so as facilitate the killing of the HIV-1 envelope expressing cell. In this regard, bispecific molecules bind with one "arm" to an epitope of a surface antigen on target cells, and with the second "arm" to an epitope of an activating, invariant component of the T cell receptor (TCR) complex. The simultaneous binding of such a bispecific molecule to both of its targets will force a temporary interaction between target cell and T cell, causing activation of any cytotoxic T cell and subsequent lysis of the target cell. Hence, the immune response is redirected to the target cells and is independent of peptide antigen presentation by the target cell or the specificity of the T cell as would be relevant for normal MHC-restricted activation of CTLs. In this context it is crucial that CTLs are only activated when a target cell is presenting the bispecific molecule to them, i.e. the immunological synapse is mimicked. Particularly desirable are bispecific molecules that do not require lymphocyte preconditioning or co- stimulation in order to elicit efficient lysis of target cells. In certain embodiments, such molecule may further comprise a third binding "arm" and be trivalent. In some embodiments, the third arm binds to an epitope of a surface antigen on target cells, which may be the same epitope or a different epitope as bound by the first arm. In some embodiments, the third arm binds to an epitope of an activating, invariant component of the TCR complex, which may be the same epitope or a different epitope as bound by the second arm. In alternative

embodiments, the third arm binds to a different epitope to that bound by the first arm or second arm, such as an epitope of a surface antigen on target cells or an epitope expressed on the surface of an effector cell (e.g. , but not limited to an epitope of CD2, CD3, CD8, CD 16, T-Cell Receptor (TCR), NKG2D, etc.).

[0193] In certain embodiments, such bispecific molecules comprise one portion which targets HIV-1 envelope and a second portion which binds a second target. In certain embodiments, the first portion comprises VH and VL sequences, or CDRs from the antibodies described herein. In certain embodiments, the second target could be, for example but not limited to an effector cell. In certain embodiments the second portion is a T-cell engager. In certain embodiments, the second portion comprises a sequence/paratope which targets CD3. In certain embodiments, the second portion is an antigen-binding region derived from a CD3 antibody, optionally a known CD3 antibody. In certain embodiments, the anti-CD3 antibody induces T cell-mediated killing. In certain embodiments, the bispecific molecules comprise whole antibodies. In other embodiments, the dual targeting bispecific molecules consist essentially of Fab fragments. In other embodiments, the dual targeting bispecific molecules comprise a heavy chain constant region (CHI). In certain embodiments, the bispecific molecule does not comprise Fc Domain. In certain embodiments, the bispecific molecules have improved effector function. In certain embodiments, the bispecific molecules have improved cell killing activity. Various methods and platforms for design of bispecific molecules (including but not limited to bispecific antibodies, bispecific diabodies, etc. ) are known in the art. See for example US Pub. 20140206846, US Pub. 20140170149,

20100174053, US Pub. 20090060910, US Pub 20130295121, US Pub. 20140099318, US Pub. 20140088295 which contents are herein incorporated by reference in their entirety.

[0194] A bispecific or Afunctional molecule is an artificial hybrid antibody that can comprise two different heavy /light chain pairs and two different binding sites (see, e.g. , Romain Rouet & Daniel Christ "Bispecific antibodies with native chain structure" Nature Biotechnology 32, 136-137 (2014); Byrne et al. "A tale of two specificities: bispecific antibodies for therapeutic and diagnostic applications" Trends in Biotechnology, Volume 31, Issue 1 1, November 2013, Pages 621-632 Songsivilai and Lachmann, Clin. Exp. Immunol., 79:315-321 (1990);

Kostelny et al , J. Immunol. 148: 1547-53 (1992) (and references therein)). In certain embodiments the bispecific molecule is a whole antibody of any isotype. In other embodiments the bispecific molecule is a bispecific fragment, for example but not limited to F(ab)2 fragment. In some embodiments, the bispecific molecules do not include Fc portion, which makes these bispecific molecules relatively small in size and easy to penetrate tissues.

[0195] In some embodiments, the invention encompasses polypeptide chains, each of which polypeptide chains comprise a VH and VL domain, comprising CDRs as described herein. In certain embodiments, the VL and VH domains comprising each polypeptide chain have the same specificity, and the multimer molecule is bivalent and monospecific. In other embodiments, the VL and VH domains comprising each polypeptide chain have differing specificity and the multimer is bivalent and bispecific. In still other embodiments, the VH and VL domains of each polypeptide chain have differing specificity and the multimer is trivalent and bispecific or trivalent and trispecific.

[0196] The multispecific molecules of the invention can simultaneously bind two separate and distinct epitopes. In certain embodiments the epitopes are from the same antigen. In other embodiments, the epitopes are from different antigens. In non-limiting embodiments at least one epitope binding site is specific for a determinant expressed on an immune effector cell (e.g. CD3, CD 16, CD32, CD64, etc. ) which are expressed on T lymphocytes, natural killer (NK) cells or other mononuclear cells. In one embodiment, the multispecific molecule binds to the effector cell determinant and also activates the effector cell. In this regard, multispecific molecules of the invention (e.g. , bispecific antibodies, bispecific diabodies, trivalent binding molecules, etc.) may exhibit Ig-like functionality independent of whether they further comprise an Fc domain (e.g. , as assayed in any effector function assay known in the art or exemplified herein).

[0197] In certain embodiments, the multispecific molecule comprises an HIV- 1 envelope binding fragment, for example but not limited to an HIV-1 envelope binding fragment from any of the antibodies described herein. In other embodiments, the multispecific molecule further comprises a second antigen-interaction-site/fragment. In other embodiments, the multispecific molecule further comprises at least one effector cell targeting arm.

[0198] In certain embodiments the multispecific molecules engage cells for Antibody- Dependent Cell-mediated Cytotoxicity (ADCC). In certain embodiments the multispecific molecules engage natural killer cells, neutrophil polymorphonuclear leukocytes, monocytes and macrophages. In certain embodiments the multispecific molecules are T-cell engagers. In certain embodiments, the bispecific molecule comprises an HIV-1 envelope binding fragment and CD3 binding fragment. Various CD3 antibodies are provided herein (see, e.g. , Table 4) and others are known in the art. See for example US Patent 8,784,821, and United States Patent Publications No. 2014-0099318 providing various disclosure on various CD3 antibodies, which disclosure is incorporated by reference in its entirety. In certain embodiments, the bispecific molecule comprises an HIV-1 envelope binding fragment and CD 16 binding fragment. Various CD 16 antibodies are provided herein (see e.g. , Table 4) and others are known in the art. See for example WO 03/101485, which disclosure is

incorporated by reference in its entirety.

[0199] In certain embodiments, the invention provides molecules or fragments comprising a CDR(s) of the VH and/or VL chains, or VH and/or VL chains of any suitable HIV-1 antibody, as the HIV-1 binding arm(s) of multispecific molecules, e.g. , but not limited to bispecific antibodies, bispecific diabodies, trivalent binding molecules, etc. , or toxin labeled HIV-1 binding molecules. Exemplary HIV-1 antibodies are provided in Table 4.

[0200] The invention also includes variants of the antibodies (and fragments) disclosed herein, including variants that retain the ability to bind to recombinant Env protein, the ability to bind to the surface of virus-infected cells and/or ADCC-mediating properties of the antibodies specifically disclosed, and methods of using same to, for example, reduce HIV- 1 infection risk. Combinations of the antibodies, or fragments thereof, disclosed herein can also be used in the generation of the multispecific molecules of the invention.

[0201] Homologs and variants of a VL or a VH of an antibody that specifically binds a polypeptide are typically characterized by possession of at least about 75%, for example at least about 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity counted over the full length alignment with the amino acid sequence of interest. Proteins with even greater similarity to the reference sequences will show increasing percentage identities when assessed by this method, such as at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, or at least 99% sequence identity. When less than the entire sequence is being compared for sequence identity, homologs and variants will typically possess at least 80% sequence identity over short windows of 10-20 amino acids, and may possess sequence identities of at least 85% or at least 90% or 95% depending on their similarity to the reference sequence. Methods for determining sequence identity over such short windows are available at the NCBI website on the internet. One of skill in the art will appreciate that these sequence identity ranges are provided for guidance only; it is entirely possible that strongly significant homologs could be obtained that fall outside of the ranges provided.

[0202] In certain embodiments, the invention provides multispecific molecules comprising the VL and VH domains of antibodies which are 99%, 98%, 97%, 96%, 95%, 94%, 93%, 92%, 91%, 90%, 89%, 88%, 87%, 86%, 85%, 84%, 83%, 82%, 81%, 80% identical to the VH and VL amino acid sequences of the antibodies described herein and still maintain their epitope binding breadth and/or potency. In certain embodiments, the invention provides multispecific molecules comprising the CDR 1, 2, and/or 3 of the VH and CDR1, 2, and/or 3 of the VL which are 99%, 98%, 97%, 96%, 95%, 94%, 93%, 92%, 91%, 90%, 89%, 88%, 87%, 86%, 85%, 84%, 83%, 82%, 81%, 80% identical to the CDR1, 2, and/or 3 of VH and CDR1, 2, and/or 3 VL amino acid sequences of the antibodies described herein and still maintain their epitope binding breadth and/or potency.

[0203] In certain embodiments, the invention provides multispecific molecules comprising polypeptide chains which are 99%, 98%, 97%, 96%, 95%, 94%, 93%, 92%, 91%, 90%, 89%, 88%, 87%, 86%, 85%, 84%, 83%, 82%, 81%, 80% identical to SEQ ID NOs:551. [0204] In certain embodiments, the invention provides multispecific molecules comprising polypeptide chains which are 99%, 98%, 97%, 96%, 95%, 94%, 93%, 92%, 91%, 90%, 89%, 88%, 87%, 86%, 85%, 84%, 83%, 82%, 81%, 80% identical to SEQ ID NOs:553.

[0205] In some aspects the invention provides recombinant, multispecific molecules, polyclonal or monoclonal antibodies, variants, fusion proteins comprising an antibody portion with an antigen recognition site of the required specificity, humanized antibodies, and chimeric antibodies, and any other modified configuration of the immunoglobulin molecule that comprises an antigen recognition site of the required specificity. Throughout this application, the numbering of amino acid residues of the light and heavy chains of antibodies is according to the EU index as in Kabat et al. (1992) SEQUENCES OF PROTEINS OF

IMMUNOLOGICAL INTEREST, National Institutes of Health Publication No. 91-3242. Amino acids from the Variable Domains of the mature heavy and light chains of immunoglobulins are designated by the position of an amino acid in the chain. Kabat described numerous amino acid sequences for antibodies, identified an amino acid consensus sequence for each subgroup, and assigned a residue number to each amino acid. The Kabat numbering scheme is extendible to antibodies not included in his compendium by aligning the antibody in question with one of the consensus sequences in Kabat by reference to conserved amino acids. This method for assigning residue numbers has become standard in the field and readily identifies amino acids at equivalent positions in different antibodies, including chimeric or humanized variants.

[0206] In some embodiments, antigen-binding fragment of an antibody is a portion of an antibody that possesses an at least one antigen recognition site. Fragments include for example but not limited to, Fab, Fab', F(ab')2 Fv), and single chain (scFv).

[0207] In certain embodiments the invention provides recombinant molecules. In certain embodiments, recombinant molecules encompasses not only intact monoclonal antibodies and full-length monoclonal antibodies, but also fragments thereof (such as Fab, Fab', F(ab')2 Fv), single chain (scFv), mutants thereof, fusion proteins and multispecific molecules comprising an antibody portion, humanized monoclonal antibodies, chimeric monoclonal antibodies, and any other modified configuration of the immunoglobulin molecule that comprises an antigen recognition site of the required specificity and the ability to bind to an antigen. Recombinant molecules are not limited as regards to the source of the molecule or the manner in which it is made (e.g., by hybridoma, phage selection, recombinant expression, transgenic animals, etc.). [0208] Methods of making recombinant molecules are known in the art. In certain embodiments, the molecules are produced recombinantly by any means known in the art. In one embodiment, the polynucleotide sequence encoding such a recombinant molecule is cloned into a vector for expression or propagation. The sequence encoding an antibody of interest may be maintained in a vector in a host cell and the host cell can then be expanded and frozen for future use. The polynucleotide sequence of such antibodies may be used for genetic manipulation to generate the multispecific molecules of the invention (e.g. , bispecific antibodies, bispecific diabodies, trivalent binding molecules, etc.) as well as a chimeric antibody, a humanized antibody, or a caninized antibody, to improve the affinity, or other characteristics of the antibody. The general principle in humanizing an antibody involves retaining the basic sequence of the antigen-binding portion of the antibody, while swapping the non-human remainder of the antibody with human antibody sequences. There are four general steps to humanize a monoclonal antibody. These are: (1) determining the nucleotide and predicted amino acid sequence of the starting antibody light and heavy variable Domains (2) designing the humanized antibody or caninized antibody, i.e., deciding which antibody framework region to use during the humanizing or canonizing process (3) the actual humanizing or caninizing methodologies/techniques and (4) the transfection and expression of the humanized antibody. See, for example, U.S. Patents Nos. 4,816,567; 5,807,715;

5,866,692; and 6,331,415.

[0209] The antibodies described herein, or fragments thereof, or molecules comprising such fragments, may be recombinantly produced in prokaryotic or eukaryotic expression systems. These systems are well described in the art. In general, protein therapeutics are produced from mammalian cells. The most widely used host mammalian cells are Chinese hamster ovary (CHO) cells and mouse myeloma cells, including NS0 and Sp2/0 cells. Two derivatives of the CHO cell line, CHO-K1 and CHO pro-3, gave rise to the two most commonly used cell lines in large scale production, DUKX-X11 and DG44. (See, e.g., Kim, J., et al, "CHO cells in biotechnology for production of recombinant proteins: current state and further potential," Appl. Microbiol. Biotechnol, 2012, 93:917-30, which is hereby incorporated-by-reference.) Other mammalian cell lines for recombinant protein expression include, but are not limited to, COS, HeLa, HEK293T, U20S, A549, HT1080, CAD, P19, NIH 3T3, L929, N2a, HEK 293, MCF-7, Y79, SO-Rb50, HepG2, J558L, and BHK. If the aim is large-scale production, the most currently used cells for this application are CHO cells. Guidelines to cell engineering for mAbs production were also reported. (Costa et al. , "Guidelines to cell engineering for monoclonal antibody production," Eur J Pharm

Biopharm, 2010, 74: 127-38, which is hereby incorporated-by-reference.) Using heterologous promoters, enhancers and amplifiable genetic markers, the yields of antibody and antibody fragments can be increased. Similar methods are utilized for the expression of the multispecific molecules of the invention. Thus, in certain embodiments, the invention provides an antibody, or antibody fragment, or molecule comprising such fragment, that is recombinantly produced from a mammalian cell-line, including a CHO cell-line. In certain embodiments, the invention provides a composition comprising an antibody, or antibody fragment, or molecule comprising such fragment, wherein the antibody, or antibody fragment, or molecule comprising such fragment was recombinantly produced in a mammalian cell-line, and wherein the antibody, or antibody fragment, or molecule comprising such fragment is present in the composition at a concentration of at least 1, 10, 100, 1000 micrograms/mL, or at a concentration of at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 20, 50, or 100 milligrams/mL.

[0210] Furthermore, large-scale production of therapeutic-grade molecules are much different than those for laboratory scale. There are extreme purity requirements for therapeutic- grade. Large-scale production of therapeutic-grade molecules requires multiples steps, including product recovery for cell-culture harvest (removal of cells and cell debris), one or more chromatography steps for antibody purification, and formulation (often by tangential filtration). Because mammalian cell culture and purification steps can introduce variants that are unique to the recombinant production process (i.e., protein aggregates, N- and C- terminal variants, acidic variants, basic variants, different glycosylation profiles), there are recognized approaches in the art for analyzing and controlling these variants. (See, Fahrner, et al. , Industrial purification of pharmaceutical antibodies: Development, operation, and validation of chromatography processes, Biotech. Gen. Eng. Rev., 2001, 18:301-327, which is hereby incorporated-by-reference.) In certain embodiments of the invention, the composition comprises less than 1, 2, 3, 4, 5, 10, 15, 20, 25, 30, 35, 50, or 100 nanograms of host cell protein (i.e., proteins from the cell-line used to recombinantly produce the molecule). In other embodiments, the composition comprises less than 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, or 25 ng of protein A per milligram of molecule (i.e., protein A is a standard approach for purifying antibodies and other Fc bearing molecules from recombinant cell culture, but steps should be done to limit the amount of protein A in the composition, as it may be immunogenic). (See, e.g. , U.S. Patent No. 7,458,704, Reduced protein A leaching during protein A affinity chromatography; which is hereby incorporated-by-reference.)

[0211] Multispecific molecules

[0212] Various non-limiting multispecific molecule designs are provides in Figures 101-105.

[0213] As provided herein, the invention contemplates designs of multispecific molecules, which include, but are not limited to bispecific antibodies, bispecific diabodies, Fc Domain bearing diabodies, trivalent binding molecules, Fc Domain bearing trivalent bind molecules etc. The multispecific molecules provided herein comprise various domains, including, but not limited to peptide linkers, Heterodimer Promoting Domains, VL and VH domains, and Fc Domains. Specific non-limiting embodiments of exemplary multispecific molecules are provided herein. Alternative combinations of the various domains described herein can be employed in the multispecific molecules of the invention.

[0214] As provided herein, the invention contemplates designs of multispecific molecules with various peptide linkers (also referred to herein as "intervening peptide linkers") separating the domains comprised in the polypeptide chains. Any of a variety of peptide linkers can be used to separate the domains in the polypeptide chains of the multispecific molecules of the invention. Typically, such peptide linkers will comprise 1-20, 1-19, 1-18, 1- 17, 1-16, 1-15, 1- 14, 1- 13, 1-12, 1-1 1, 1- 10, 1-9, 1-8, 1-7, 1-6, 1-5, 1-4, 1-3, 1-2, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1, 12, 13, 14, 15, 16, 17, 18, 19, or 20 amino acid residues. Such polypeptide linkers can include a series of glycine residues (Gly) and/or Serine (Ser) residues and may optionally comprise cysteine residue(s). Specific non-limiting embodiments of exemplary polypeptide linkers (e.g. , Peptide Linker 1, Peptide Linker 2, Spacer Linker 3, etc. ) are provided herein. Alternative peptide linkers are well-known in the art and can be employed in the multispecific molecules of the invention. Other linkers can be readily determined. Some additional examples of linkers are disclosed in US Pub 20100174053, incorporated by reference in its entirety.

[0215] Typically, the VH and VL domains of a polypeptide chain of the multispecific molecules are linked so that they do not associate with each other. In certain embodiments the length of the peptide linker, which separates such VL and VH domains of a polypeptide chain is selected to substantially or completely prevent such VL and VH domains from binding to one another. Thus the VL and VH domains of a polypeptide chain are substantially or completely incapable of binding to one another. In certain embodiments this is due to the peptide linker which separates the VH and VL domains. As provided herein, the invention also contemplates designs of multispecific molecules wherein the domains comprising the polypeptide chains (e.g., Heterodimer Promoting Domains, VL and VH domains, and Fc Domains etc.) are directly linked (i. e. no peptide linker is used between the domain). In such multispecific molecules the domains can be linked by a peptide bond.

[0216] One embodiment of the present invention relates to multispecific molecules, which are bispecific, that are capable of binding to a "first epitope" and a "second epitope," such epitopes not being identical to one another. Such bispecific molecules comprise "VLl" / "VHl" domains that are capable of binding to the first epitope, and "VL2" / "VH2" domains that are capable of binding to the second epitope. The notation "VLl" and "VHl" denote respectively, the Variable Light Chain Domain and Variable Heavy Chain Domain of such bispecific molecules that bind the "first" epitope. Similarly, the notation "VL2" and "VH2" denote respectively, the Light Chain Variable Domain and Heavy Chain Variable Domain of such bispecific molecules that bind the "second" epitope. It is irrelevant whether a particular epitope is designated as the first vs. the second epitope; such notation having relevance only with respect to the presence and orientation of domains of the polypeptide chains of such multispecific molecules of the present invention. In one embodiment, one of such epitopes is an epitope of HIV- 1 Env (for example but not limited to a gp41 MPER epitope), and the other is an epitope of a molecule that is not HIV-1 Env. In particular embodiments, one of such epitopes is an epitope of HIV-1 Env and the other is an epitope of a molecule (e.g. , CD2, CD3, CD8, CD 16, T-Cell Receptor (TCR), NKG2D, etc. ) present on the surface of an effector cell, for example but not limited to a T lymphocyte, a natural killer (NK) cell or other mononuclear cell (see, e.g. , Figures 101 and 103A-103B). In certain embodiments, a bispecific molecule comprises more than two epitope-binding sites (see, e.g. , Figures 102A- 102C). Such bispecific molecules will bind at least one epitope of HIV-1 Env and at least one epitope of a molecule that is not HIV-1 Env.

[0217] One embodiment of the present invention also relates to trivalent binding molecules that are capable of binding to a "first epitope," a "second epitope," and a "third epitope," wherein at least one of such epitopes is not identical to another. Such trivalent binding molecules comprise VLl / VHl domains that are capable of binding to the first epitope, VL2 / VH2 domains that are capable of binding to the second epitope, and further comprise "VL3" / "VH3" domains that are capable of binding to the third epitope, wherein the notation "VL3" and "VH3" denote respectively, the Variable Light Chain Domain and Variable Heavy Chain Domain of such trivalent binding molecules that bind the "third" epitope. The capacity to bind a third epitope provides additional and/or enhanced functionality. In one embodiment, one (or two) of such epitopes is an epitope of HIV- 1 Env (particularly a gp41 MPER epitope), and two (or one) of such epitopes is an epitope of a molecule that is not HIV- 1 Env. In particular embodiments, one (or two) of such epitopes is an epitope of HIV- 1 Env and two (or one) of such epitopes is an epitope of a molecule (e.g. , CD2, CD3, CD8, CD 16, T-Cell Receptor (TCR), NKG2D, etc.) present on the surface of an effector cell, such as a T lymphocyte, a natural killer (NK) cell or other mononuclear cell (see, e.g. , Figures 104A- 104F). Such trivalent binding molecules will bind at least one epitope of HIV- 1 Env and at least one epitope of a molecule that is not HIV- 1 Env, and may bind two epitopes of HIV- 1 Env and one epitope of a molecule that is not HIV-1 Env or may bind one epitope of HIV-1 Env and two epitopes that are not epitopes of HIV-1 Env.

[0218] In some embodiments, such molecules comprise two polypeptide chains, wherein each of the two polypeptide chains comprises three Domains (Figure 101). The first polypeptide chain comprises: (i) a Domain that comprises a binding region of a light chain variable Domain of a first immunoglobulin (VL1), (ii) a second Domain that comprises a binding region of a heavy chain variable Domain of a second immunoglobulin (VH2), and (iii) a third Domain that serves to promote heterodimerization with the second polypeptide chain and to covalently bond the first polypeptide to the second polypeptide chain of the molecule. The second polypeptide chain contains a complementary first Domain (a VL2 Domain), a complementary second Domain (a VH1 Domain) and a third Domain that complexes with the third Domain of the first polypeptide chain in order to promote heterodimerization and covalent bonding with the first polypeptide chain. Such molecules are stable, potent and have the ability to simultaneously bind two or more antigens. They are able to promote redirected T cell (CD3) or NK (CD 16) cell mediated killing of cells expressing target antigens.

[0219] In certain embodiments, the HIV-1 multispecific molecules of the present invention are composed of two polypeptide chains which associate with one another to form one binding site specific for an epitope of HIV-1, and one binding site specific for an epitope of CD2, CD3, CD8, CD 16, T-Cell Receptor (TCR), NKG2D, etc. , so as to be capable of simultaneously binding to HIV- 1 and, for example, to CD3. Thus, such diabodies bind to a "first epitope," which may be either an epitope of CD3 or HIV-1, and a "second epitope," which is an epitope of HIV- 1 when the first epitope is an epitope of CD3, and is an epitope of CD3 when the first epitope is from HIV-1. Alternatively, such diabodies bind to a "first epitope," which may be either an epitope of CD 16 or HIV-1, and a "second epitope," which is an epitope of HIV-1 when the first epitope is of CD 16, and is an epitope of CD 16 when the first epitope is an epitope of HIV-1.

[0220] In certain embodiments, the first of such two polypeptide chains will contain, in the N- terminal to C-terminal direction, an N-terminus, the Antigen-Binding Domain of a Light Chain Variable Domain (VL) of an antibody that binds to a "first" epitope of a "first" antigen (e.g. , either CD3 or HIV-1 envelope), the Antigen-Binding Domain of a Heavy Chain Variable Domain (VH) of an antibody that binds to a "second" epitope of a "second" antigen (HIV-1, if the first antigen was CD3; CD3, if the first antigen was HIV-1), a

Heterodimerization-Promoting Domain, and a C-terminus. An intervening peptide linker (Peptide Linker 1) separates the Antigen-Binding Domain of the Light Chain Variable Domain from the Antigen-Binding Domain of the Heavy Chain Variable Domain. In certain embodiments the Antigen-Binding Domain of the Heavy Chain Variable Domain is linked to the Heterodimerization-Promoting Domain by an intervening peptide linker (Peptide Linker 2). In certain embodiments the first of the two polypeptide chains will thus contain, in the N- terminal to C-terminal direction: VLFirst Antigen - Peptide Linker 1 - VHsecond Antigen - Peptide Linker 2 - Heterodimerization-Promoting Domain.

[0221] The terms VL/VH for first and second antig ens, VLfirst antigen VHfirst antigen VLsecond antigen/VHsecond antigen, and VL1/VH1 and VL2/VH2 are used interchangeably throughout the application.

[0222] In certain embodiments, the second of such two polypeptide chains will contain, in the N-terminal to C-terminal direction, an N-terminus, the Antigen-Binding Domain of a Light Chain Variable Domain (VL) of an antibody that binds to the second epitope of the second antigen, the Antigen-Binding Domain of a Heavy Chain Variable Domain (VH) of an antibody that binds to the first epitope of the first antigen, a Heterodimerization-Promoting Domain and a C-terminus. An intervening peptide linker (Peptide Linker 1) separates the Antigen-Binding Domain of the Light Chain Variable Domain from the Antigen-Binding Domain of the Heavy Chain Variable Domain. In certain embodiments, the Antigen-Binding Domain of the Heavy Chain Variable Domain is linked to the Heterodimerization-Promoting Domain by an intervening peptide linker (Peptide Linker 2). In certain embodiments the second of the two polypeptide chains will thus contain, in the N-terminal to C-terminal direction: VLsecond Antigen Peptide Linker 1 - VHFirst Antigen Peptide Linker 2 - Heterodimerization-Promoting Domain.

[0223] The Antigen-Binding Domain of the Light Chain Variable Domain of the first polypeptide chain interacts with the Antigen-Binding Domain of the Heavy Chain Variable Domain of the second polypeptide chain in order to form a functional antigen-binding site that is specific for the first antigen (e.g., either HIV-1 envelope or CD3). Likewise, the Antigen- Binding Domain of the Light Chain Variable Domain of the second polypeptide chain interacts with the Antigen-Binding Domain of the Heavy Chain Variable Domain of the first polypeptide chain in order to form a second functional antigen-binding site that is specific for the second antigen (e.g., either CD3 or HIV- 1 envelope, depending upon the identity of the first antigen). Thus, the selection of the Antigen-Binding Domain of the Light Chain Variable Domain and the Antigen-Binding Domain of the Heavy Chain Variable Domain of the first and second polypeptide chains are coordinated, such that the two polypeptide chains collectively comprise Antigen-Binding Domains of light and Heavy Chain Variable Domains capable of binding to the intended targets, in certain embodiments e.g. HIV-1 envelope and CD3, or CD 16.

[0224] In certain embodiments the length of Peptide Linker 1, which separates such VL and VH domains of a polypeptide chain is selected to substantially or completely prevent such VL and VH domains from binding to one another. Thus the VL and VH domains of the first polypeptide chain are substantially or completely incapable of binding to one another.

Likewise, the VL and VH domains of the second polypeptide chain are substantially or completely incapable of binding to one another. In certain embodiments this is due to the peptide linker which separates the VH and VL domains. In certain embodiments, the peptide linker is 5, 6, 7, 8, 9, 10, 1 1, 12, 13, 14, 15, 16, 17, 18, 19, 20, but no more than 20 amino acids. In some embodiments, the peptide linker is less than 12 amino acids in length. In certain embodiments an intervening spacer peptide (Peptide Linker 1) has the sequence (SEQ ID NO:508): GGGSGGGG.

[0225] Peptide Linker 2 separates the VH Domain of a polypeptide chain from the

Heterodimer-Promoting Domain of that polypeptide chain. Any of a variety of linkers can be used for the purpose of Peptide Linker 2. The length and composition of Peptide Linker 2 may be selected based on the choice of heterodimer-promoting domains. Typically, the second intervening peptide linker (Peptide Linker 2) will comprise 1-20 amino acid residues. In certain embodiments, where the heterodimer-promoting domains do not comprise a cysteine residue a cysteine-containing second intervening peptide linker (Peptide Linker 2) is utilized. Optionally, both a cysteine-containing Peptide Linker 2 (Peptide Linker 2-C) and a cysteine-containing Heterodimer-Promoting Domain are used. In certain embodiments a sequence for such Peptide Linker 2 has the amino acid sequence: GGCGGG (SEQ ID NO:509), which has a cysteine residue that may be used to covalently bond the first and second polypeptide chains to one another via a disulfide bond. In certain embodiments, a sequence for Peptide Linker 2-C has the amino acid sequence: ASTKG (SEQ ID NO: 510). Peptide Linker 2 and Peptide Linker 2-C could be used interchangeably.

[0226] The formation of heterodimers of the first and second polypeptide chains can be driven by the inclusion of specific sequences referred to as Heterodimer-Promoting Domains (HPDs). Such domains include without limitation GVEPKSC (SEQ ID NO:51 1) or VEPKSC (SEQ ID NO:512) on one polypeptide chain and GFNRGEC (SEQ ID NO:513) or FNRGEC (SEQ ID NO:514) on the other polypeptide chain (See US2007/0004909 herein incorporated by reference in its entirety). Various HPD sequences are contemplated by the invention and disclosed in the specification. Table 4 discloses non-limiting embodiments of various HPDs. In some embodiments the HPD include E/K-coils (SEQ ID NOs: 518, 520) or cysteine engineered E/K-coils (SEQ ID NOs: 519, 521). In some embodiments HPD includes combinations of SEQ ID NOs: 511, 512, 513, and 514 (e.g., SEQ ID NOs: 511 and 513; SEQ ID NOs: 512 and 513; SEQ ID NOs: 51 1 and 514; SEQ ID NOs: 512 and 514). In some embodiments HPDs include any suitable sequences with a Cysteine residue to permit disulfide bond. In some embodiments HPDs includes suitable CHI and CL domains (See for example CH domain (SEQ ID NO: 515) and kappa and lambda light chain constant domains (SEQ ID NOs: 516 and 517).

[0227] In certain embodiments, the Heterodimer-Promoting Domains of the present invention are formed from one, two, three or four tandemly repeated coil domains of opposing charge that comprise a sequence of at least six, at least seven or at least eight charged amino acid residues (Apostolovic, B. et al. (2008) "pH-Sensitivity of the E3/K3 Heterodimeric Coiled Coil " Biomacromolecules 9:3173-3180; Arndt, K M. et al. (2001) "Helix-stabilized Fv (hsFv) Antibody Fragments: Substituting the Constant Domains of a Fab Fragment for a Heterodimeric Coiled-coi I Domain " J. Molec. Biol. 312:221-228; Arndt, K.M. et al. (2002) "Comparison of In Vivo Selection and Rational Design ofHeterodimeric Coiled Coils " Structure 10: 1235-1248; Boucher, C. et al. (2010) "Protein Detection By Western Blot Via Coiled-Coil Interactions " Analytical Biochemistry 399: 138-140; Cachia, P.J. et al. (2004) "Synthetic Peptide Vaccine Development: Measurement Of Polyclonal Antibody Affinity And Cross-Reactivity Using A New Peptide Capture And Release System For Surface Plasmon Resonance Spectroscopy " J. Mol. Recognit. 17:540-557; De Crescenzo, G.D. et al. (2003) "Real-Time Monitoring of the Interactions of Two-Stranded de novo Designed Coiled-Coils: Effect of Chain Length on the Kinetic and Thermodynamic Constants of Binding "

Biochemistry 42: 1754-1763; Fernandez-Rodriquez, J. et al. (2012) "Induced

Heterodimerization And Purification Of Two Target Proteins By A Synthetic Coiled-Coil Tag " Protein Science 21 :511-519; Ghosh, T.S. et al. (2009) "End-To-End And End-To- Middle Interhelical Interactions: New Classes Of Interacting Helix Pairs In Protein

Structures " Acta Crystallographica D65: 1032-1041; Grigoryan, G. et al. (2008) "Structural Specificity In Coiled-Coil Interactions " Curr. Opin. Struc. Biol. 18:477-483; Litowski, J.R. et al. (2002) "Designing Heterodimeric Two-Stranded a-Helical Coiled-Coils: The Effects Of Hydrophobi city And a-Helical Propensity On Protein Folding, Stability, And Specificity " J. Biol. Chem. 277:37272-37279; Steinkruger, J.D. et al. (2012) "The d'-d-d' Vertical Triad is Less Discriminating Than the a'— a— a' Vertical Triad in the Antiparallel Coiled-coil Dimer Motif" J. Amer. Chem. Soc. 134(5):2626-2633; Straussman, R. et al. (2007) "Kinking the Coiled Coil - Negatively Charged Residues at the Coiled-coil Interface " J. Molec. Biol. 366: 1232-1242; Tripet, B. et al. (2002) "Kinetic Analysis of the Interactions between Troponin C and the C-terminal Troponin I Regulatory Region and Validation of a New Peptide Delivery/Capture System used for Surface Plasmon Resonance " J. Molec. Biol. 323:345-362; Woolfson, D.N. (2005) "The Design Of Coiled-Coil Structures And

Assemblies " Adv. Prot. Chem. 70:79-112; Zeng, Y. et al. (2008) "A Ligand-Pseudoreceptor System Based On de novo Designed Peptides For The Generation Of Adenoviral Vectors With Altered Tropism " J. Gene Med. 10:355-367).

[0228] Such repeated coil domains may be exact repeats or may have substitutions. For example, the Heterodimer-Promoting Domain of the first polypeptide chain may comprise a sequence of eight negatively charged amino acid residues and the Heterodimerization- Promoting Domain of the second polypeptide chain may comprise a sequence of eight negatively charged amino acid residues. It is immaterial which coil is provided to the first or second polypeptide chains, provided that a coil of opposite charge is used for the other polypeptide chain.

[0229] In certain embodiments a multispecific molecule of the present invention has a first polypeptide chain having a negatively charged coil. The positively charged amino acid may be lysine, arginine, histidine, etc. and/or the negatively charged amino acid may be glutamic acid, aspartic acid, etc. In certain embodiments the positively charged amino acid is lysine and/or the negatively charged amino acid is glutamic acid. It is possible for only a single Heterodimer-Promoting Domain to be employed (since such domain will inhibit

homodimerization and thereby promote heterodimerization). In certain embodiments both the first and second polypeptide chains of the multispecific molecules of the present invention contain Heterodimer-Promoting Domains.

[0230] In certain embodiments, one of the Heterodimer-Promoting Domains will comprise four tandem "E-coil" helical domains (SEQ ID NO:518 (EVAALEK-EVAALEK- EVAALEK-EVAALEK)), whose glutamate residues will form a negative charge at pH 7, while the other of the Heterodimer-Promoting Domains will comprise four tandem "K-coil" domains (SEQ ID NO:520 (KVAALKE-KVAALKE-KVAALKE-KVAALKE)), whose lysine residues will form a positive charge at pH 7. The presence of such charged domains promotes association between the first and second polypeptides, and thus fosters

heterodimerization. In other embodiments, one of the four tandem "E-coil" helical domains of SEQ ID NO: 518 has been modified to contain a cysteine residue: EVAA£EK- EVAALEK-EVAALEK-EVAALEK (SEQ ID NO: 519). In some embodiments, a Heterodimer-Promoting Domain in which one of the four tandem "K-coil" helical domains of SEQ ID NO: 520 has been modified to contain a cysteine residue: KVAACKE-KVAALKE- KVAALKE-KVAALKE (SEQ ID NO: 521). Such cysteine modified Heterodimer- Promoting Domains may be used to covalently bond the first and second polypeptide chains to one another via a disulfide bond.

[0231] In some embodiments, the number of K coil and E coil domains can vary and a skilled artisan can readily determine whether a different number of K-coil or E-coil domain lead to heterodimerization .

[0232] In certain embodiments, the multispecific molecules of the invention, for example but not limited to bispecific monovalent diabodies and trivalent binding molecules, are engineered so that their first and second polypeptide chains covalently bond to one another via one or more cysteine residues positioned along their length. Such cysteine residues may be introduced into the intervening peptide linker that separates the VL and VH domains of the polypeptides. Alternatively, as provided above Peptide Linker 2 and/or the HPDs may contain such cysteine residues.

[0233] The invention also includes variants of the multispecific molecules, or fragments thereof disclosed herein, including variants that retain the ability to bind to recombinant Env protein, the ability to bind to the surface of virus-infected cells and/or ADCC-mediating properties of the antibodies specifically disclosed, and methods of using same to, for example, reduce HIV- 1 infection risk. Combinations of the multispecific molecules, antibodies, or fragments thereof, disclosed herein can also be used in the methods of the invention.

[0234] Formation of multispecific molecule as described herein requires the interaction of differing polypeptide chains. Such interactions are difficult to achieve with efficiency within a single cell recombinant production system, due to the many variants of potential chain mispairings. One solution to decrease the probability of mispairings, is to engineer "knobs- into-holes" type mutations into the desired polypeptide chain pairs. Such mutations favor heterodimerization over homodimerization. For example, with respect to Fc-Fc-interactions, an amino acid substitution (preferably a substitution with an amino acid comprising a bulky side group forming a knob\ e.g. , tryptophan) can be introduced into the CH2 or CH3 domain such that steric interference will prevent interaction with a similarly mutated domain and will obligate the mutated domain to pair with a domain into which a complementary, or accommodating mutation has been engineered, i. e. , ' the hole ' (e.g. , a substitution with glycine). Such sets of mutations can be engineered into any pair of polypeptides comprising the multispecific molecule, and further, engineered into any portion of the polypeptides chains that comprise a multispecific molecule. Methods of protein engineering to favor

heterodimerization over homodimerization are well known in the art, in particular with respect to the engineering of immunoglobulin-like molecules, and are encompassed herein (see e.g. , Ridgway et al. (1996) " ' Knobs-Into-Holes ' Engineering Of Antibody CH3 Domains For Heavy Chain Heterodimerization," Protein Engr. 9:617-621, Atwell et al. (1997) "Stable Heterodimers From Remodeling The Domain Interface Of A Homodimer Using A Phage Display Library," J. Mol. Biol. 270: 26-35, and Xie et al. (2005) "A New Format Of

Bispecific Antibody: Highly Efficient Heterodimerization, Expression And Tumor Cell Lysis," J. Immunol. Methods 296:95-101 ; each of which is hereby incorporated herein by reference in its entirety).

[0235] In some embodiments the invention provides multispecific molecules comprising variant Fc domain (or portions thereof), which variant Fc domain comprises at least one amino acid modification (e.g. substitution, insertion deletion) relative to a comparable wild- type Fc domain or hinge-Fc domain (or portion thereof). Molecules comprising variant Fc domains or hinge-Fc domains (or portion thereof) (e.g. , antibodies) normally have altered phenotypes relative to molecules comprising wild-type Fc domains or hinge-Fc domains or portions thereof. The variant phenotype may be expressed as altered serum half-life, altered stability, altered susceptibility to cellular enzymes or altered effector function as assayed in an NK dependent or macrophage dependent assay. Fc domain variants identified as altering effector function are known in the art. For example International Application WO04/063351, U.S. Patent Application Publications 2005/0037000 and 2005/0064514.

[0236] In some embodiments the invention provides multispecific molecules comprising a hinge domain. The hinge domain be derived from any immunoglobulin isotype or allotype including IgA, IgD, IgG, IgE and IgM. In preferred embodiments, the hinge domain is derived from IgG, wherein the IgG isotype is IgGl, IgG2, IgG3 or IgG4, or an allotype thereof. The hinge domain may be engineered into a polypeptide chain comprising the multispecific molecule together with an Fc domain such that the multispecific molecule comprises a hinge-Fc domain. In certain embodiments, the hinge and Fc domain are independently selected from any immunoglobulin isotype known in the art or exemplified herein. In other embodiments the hinge and Fc domain are separated by at least one other domain of the polypeptide chain, e.g., the VL domain.

[0237] In another aspect, the invention provides multispecific molecules which include Fc domain(s)-- Fc bearing multispecific molecules. While some of the disclosure regarding Fc domain(s) refers to specific designs, a skilled artisan appreciates that the Fc disclosure is pertinent to any Fc bearing design of multispecific molecules, including but not limited to the designs described in Figures 101-105.

[0238] Fc bearing multispecific molecules, for example but not limited to Fc bearing diabodies are heavier, and could bind neonatal Fc receptor, increasing their circulating half- life. See Garber "Bispecific antibodies rise again" Nature Reviews Drug Discovery 13, 799- 801 (2014), Figure la; See US Pub 20130295121, US Pub 20140099318 incorporated by reference in their entirety. In certain embodiments, the invention encompasses multispecific molecules comprising an Fc domain or portion thereof (e.g. a CH2 domain, or CH3 domain). The Fc domain or portion thereof may be derived from any immunoglobulin isotype or allotype including, but not limited to, IgA, IgD, IgG, IgE and IgM. In some embodiments, the Fc domain (or portion thereof) is derived from IgG. In some embodiments, the IgG isotype is IgGl, IgG2, IgG3 or IgG4 or an allotype thereof. In some embodiments, the multispecific molecule comprises an Fc domain, which Fc domain comprises a CH2 domain and CH3 domain independently selected from any immunoglobulin isotype (i.e. an Fc domain comprising the CH2 domain derived from IgG and the CH3 domain derived from IgE, or the CH2 domain derived from IgGl and the CH3 domain derived from IgG2, etc.). In some embodiments, the Fc domain may be engineered into a polypeptide chain comprising the multispecific molecule of the invention in any position relative to other domains or portions of the polypeptide chain (e.g. , the Fc domain, or portion thereof, may be C-terminal to both the VL and VH domains of the polypeptide of the chain; may be N-terminal to both the VL and VH domains; or may be N-terminal to one domain and C-terminal to another (i.e. , between two domains of the polypeptide chain)).

[0239] Other modifications of the multispecific molecules are contemplated to increase the half-life of the molecules. In some embodiments, these modifications include addition of a polypeptide portion of a serum binding protein. See US20100174053 Al, incorporated by reference.

[0240] In some embodiments, the Fc variants of the multispecific molecules of the invention are expected to have increased serum half-life compared to the non-Fc variants. Skilled artisan can readily carry out various assays, including pharmacokinetic studies, to determine the half-life of these molecules.

[0241] In some embodiments, the polypeptide chains in multispecific molecules further comprise an Fc domain. Dimerization of the Fc domains leads to formation of a multispecific molecule that exhibits immunoglobulin-like functionality, i. e. , Fc mediated function (e.g. , Fc- Fc.gamma.R interaction, complement binding, etc.).

[0242] As provided in Figures 102-103, one or both of the polypeptide chains of bispecific diabodies may additionally comprise the sequence of a CH2-CH3 Domain, such that complexing between the two diabody polypeptides forms an Fc Domain that may be capable of binding to the Fc receptor of cells (such as B lymphocytes, dendritic cells, natural killer cells, macrophages, neutrophils, eosinophils, basophils and mast cells). Similarly, the first and third polypeptide chains of trivalent binding molecules can comprise the sequence of a CH2- CH3 Domain, such that complexing between these two polypeptide chains forms an Fc Domain. As provided in more detail below, the CH2 and/or CH3 Domains of such polypeptide chains need not be identical in sequence, and advantageously are modified to foster complexing between the two polypeptide chains. Many variations of such molecules have been described (see, e.g., United States Patent Publications No. 2014-0099318; 2013- 0295121 ; 2010-0174053 and 2009-0060910; European Patent Publication No. EP 2714079; EP 2601216; EP 2376109; EP 2158221 and PCT Publications No. WO 2015/026894; WO 2015/026892; WO 2015/021089; WO 2014/159940; WO 2012/162068; WO 2012/018687; WO 2010/080538, the content of each of these publications in herein incorporated by reference in its entirety).

[0243] In some embodiments, Fc -bearing bispecific diabodies may comprise two pairs of polypeptide chains (or four different chains, as provided below). The first and third polypeptide chains of such a bispecific molecule (e.g., diabodies) contain three domains: (i) a VL1 -containing Domain, (ii) a VH2-containing Domain, (iii) Heterodimer-Promoting Domain and (iv) a Domain containing a CH2-CH3 sequence. The second and fourth polypeptide chains contain: (i) a VL2 -containing Domain, (ii) a VHl -containing Domain and (iii) a Heterodimer-Promoting Domain, where the Heterodimer-Promoting Domains promote the dimerization of the first/third chains with the second/fourth chains. The VL and/or VH Domains of the third and fourth polypeptide chains, and VL and/or VH Domains of the first and second polypeptide chains may be the same or different so as to permit tetravalent binding that is either monospecific, bispecific or tetraspecific. Such molecules are tetravalent and have enhanced potency. The general structure of the polypeptide chains of a

representative four-chain Fc Domain-containing multispecific molecules of invention is provided in Table 1 :

[0244] HIV- 1 bispecific bivalent Fc bearing diabodies can be composed of two pairs of polypeptide chains (i.e., two first polypeptide chain and two second polypeptide chains) which associate with one another to form two binding sites specific for an epitope of HIV-1 and two binding sites specific for an epitope, for example but not limited to CD3 (see, Figures 102A-102C), so as to be capable of simultaneously binding to HIV-1 and to CD3. Thus such molecules binding to a "first" epitope, on a "first" antigen which may be either CD3 or HIV-1, and a "second" epitope on a "second" antigen, which is HIV-1 when the first epitope is CD3, and is CD3 when the first epitope is HIV-1.

[0245] As shown in Figures 102A-102C, the first polypeptide chain comprises (in the N- terminal to C-terminal direction): an N-terminus, the Antigen-Binding Domain of a Light Chain Variable Domain (VL1) of an antibody that binds to a "first" epitope of a "first" antigen (either an effector cell epitope such as but not limited to CD3, or HIV-1), the Antigen- Binding Domain of a Heavy Chain Variable Domain (VH2) of an antibody that binds to a "second" epitope of a "second" antigen (HIV-1, if the first antigen as CD3; CD3, if the first antigen was HIV-1), a Heterodimer-Promoting Domain which may comprise a cysteine residue, the CH2-CH3 domains of an Fc Domain ("Fc Domain") and a C-terminus. The second polypeptide contains (in the N-terminal to C-terminal direction): an N-terminus, the Antigen-Binding Domain the Light Chain Variable Domain (VL2) of an antibody that binds to the second epitope of the second antigen (VL2), the Antigen-Binding Domain of the Heavy Chain Variable Domain (VH1) of an antibody that binds to the first epitope of the first antigen, a Heterodimer-Promoting Domain that promotes heterodimerization with the first polypeptide chain, and a C-terminus. Here, two first polypeptides complex with each other to form an Fc Domain. An intervening peptide linker (Peptide Linker 1) separates the Antigen- Binding Domain of the Light Chain Variable Domain from the Antigen-Binding Domain of the Heavy Chain Variable Domain. In non-limiting embodiments the Antigen-Binding Domain of the Heavy Chain Variable Domain is linked to the Heterodimer-Promoting Domain by an intervening peptide linker (Peptide Linker 2). In other non-limiting embodiments, the Heterodimer-Promoting Domain is linked to the Fc Domain by an intervening peptide linker (Peptide Linker 3) or by an intervening spacer-linker peptide (Spacer-Linker 3). In certain embodiments, the first and second polypeptide chains form a disulfide bond between cysteine residues, which may be present in Peptide Linker 2 (e.g., but not limited to Peptide Linker 2-C) and/or in the Heterodimer-Promoting Domains (e.g. but not limited to E-coil-C/K-coil-C). Figures 102A-102C provide schematics of three variations of such diabodies utilizing different Heterodimer-Promoting Domains. In non- limiting embodiments, the first polypeptide chains will contain, in the N-terminal to C- terminal direction: VL1 - Peptide Linker 1 - VH2 - Peptide Linker 2 - Heterodimer- Promoting Domain - Spacer-Linker 3 - Fc Domain, and the second polypeptide chains will contain, in the N-terminal to C-terminal direction: VL2 - Peptide Linker 1 - VH1 - Peptide Linker 2 - Heterodimer-Promoting Domain.

[0246] In some embodiments, Fc bearing diabodies may comprise three polypeptide chains. The first polypeptide of such a molecule contains three Domains: (i) a VL1 -containing Domain, (ii) a VH2 -containing Domain, (iii) a Domain that promotes heterodimerization and covalent bonding with the diabody 's first polypeptide chain and (iv) a Domain containing a CH2-CH3 sequence. The second polypeptide of such diabodies contains: (i) a VL2- containing Domain, (ii) a VH1 -containing Domain and (iii) a Domain that promotes heterodimerization and covalent bonding with the diabody's first polypeptide chain. The third polypeptide of such diabodies comprises a CH2-CH3 sequence. Thus, the first and second polypeptide chains of such diabodies associate together to form a VL1/VH1 binding site that is capable of binding to the epitope, as well as a VL2/VH2 binding site that is capable of binding to the second epitope. The first and second polypeptides are bonded to one another through a disulfide bond involving cysteine residues in their respective third Domains.

Notably, the first and third polypeptide chains complex with one another to form an Fc Domain that is stabilized via a disulfide bond. Such diabodies have enhanced potency. Such Fc bearing diabodies may have either of two orientations (Table 2):

[0247] HIV- 1 bispecific monovalent Fc bearing diabodies can be composed of three polypeptide chains which associate with one another to form one binding site specific for an epitope of HIV-1 and one binding site specific for another epitope, for example but not limited to an epitope of CD3 (see, Figure 103A-103B), so as to be capable of simultaneously binding to HIV-1 and to CD3. Thus, such molecules bind to a "first" antigen, which may be either CD3 or HIV-1, and a "second" antigen, which is HIV-1 when the first epitope is CD3, and is CD3 when the first epitope is HIV-1.

[0248] As shown in Figure 103A, the first of such three polypeptide chains will contain, in the N-terminal to C-terminal direction, an N-terminus, the Antigen-Binding Domain of a Light Chain Variable Domain (VL) of an antibody that binds to a "first" epitope of a "first" antigen (for example but not limited to either CD3 or HIV-1), the Antigen -Binding Domain of a Heavy Chain Variable Domain (VH) of an antibody that binds to a "second" epitope of a "second" antigen (for example but not limited to HIV-1, if the first antigen was CD3; CD3, if the first antigen was HIV-1), a Heterodimerization-Promoting Domain, and a C-terminus. An intervening peptide linker (Peptide Linker 1) separates the Antigen-Binding Domain of the Light Chain Variable Domain from the Antigen-Binding Domain of the Heavy Chain Variable Domain. In non-limiting embodiments, the Antigen-Binding Domain of the Heavy Chain Variable Domain is linked to the Heterodimerization-Promoting Domain by an intervening peptide linker (Peptide Linker 2). In a non-limiting example of an HIV- 1 bispecific monovalent Fc bearing diabody, the C-terminus of the Heterodimerization- Promoting Domain is linked to the CH2-CH3 domains of an Fc Domain ("Fc Domain") by an intervening peptide linker (Peptide Linker 3) or by an intervening spacer-linker peptide (Spacer-Linker 3). In non-limiting embodiments, the first of the three polypeptide chains will thus contain, in the N-terminal to C-terminal direction: VLFirst Antigen - Peptide Linker 1 - VHsecond Antigen - Peptide Linker 2 - Heterodimerization-Promoting Domain - Spacer-Linker 3 - Fc Domain.

[0249] Alternatively, as shown in Figure 103B, the first of such three polypeptide chains will contain, in the N-terminal to C-terminal direction, an N-terminus, Peptide Linker 3, the CH2-CH3 domains of an Fc Domain ("Fc Domain"), an intervening spacer peptide (Peptide Linker 4), having, for example the amino acid sequence: APSSS (SEQ ID NO:524) or the amino acid sequence APSSSPME (SEQ ID NO:525), the Antigen-Binding Domain of a Light Chain Variable Domain (VL) of an antibody that binds to the first epitope of the first antigen (for example but not limited to CD3 or HIV- 1), the Antigen-Binding Domain of a Heavy Chain Variable Domain (VH) of an antibody that binds to the second epitope of the second antigen (for example but not limited to HIV- 1, if the first antigen was CD3; CD3, if the first antigen was HIV-1), a Heterodimerization-Promoting Domain, and a C-terminus. An intervening peptide linker (Peptide Linker 1) separates the Antigen-Binding Domain of the Light Chain Variable Domain from the Antigen-Binding Domain of the Heavy Chain Variable Domain. In non-limiting embodiments, the Antigen-Binding Domain of the Heavy Chain Variable Domain is linked to the Heterodimerization-Promoting Domain by an intervening peptide linker peptide (Peptide Linker 2). In non-limiting embodiments, the first of the three polypeptide chains will thus contain, in the N-terminal to C-terminal direction: Peptide Linker 3 - Fc Domain - Peptide Linker 4 - VLFirst Antigen - Peptide Linker 1 - VHsecond Antigen - Peptide Linker 2 - Heterodimerization-Promoting Domain.

[0250] In non-limiting embodiments, the second of such three polypeptide chains will contain, in the N-terminal to C-terminal direction, an N-terminus, the Antigen-Binding Domain of a Light Chain Variable Domain (VL) of an antibody that binds to the second epitope of the second antigen, the Antigen-Binding Domain of a Heavy Chain Variable Domain (VH) of an antibody that binds to the first epitope of the first antigen, a

Heterodimerization-Promoting Domain and a C-terminus. An intervening peptide linker (Peptide Linker 1) separates the Antigen-Binding Domain of the Light Chain Variable Domain from the Antigen-Binding Domain of the Heavy Chain Variable Domain. In non- limiting embodiments, the Antigen-Binding Domain of the Heavy Chain Variable Domain is linked to the Heterodimerization-Promoting Domain by an intervening peptide linker (Peptide Linker 2). In non-limiting embodiments, the second of the three polypeptide chains will thus contain, in the N-terminal to C-terminal direction: VLsecond Antigen - Peptide Linker 1 - VHFirst Antigen - Peptide Linker 2 - Heterodimerization-Promoting Domain.

[0251] In non-limiting embodiments, the third of such three polypeptide chains will contain a peptide linker (Peptide Linker 3) and the CH2-CH3 domains of an Fc Domain ("Fc

Domain").

[0252] For the various Fc bearing multispecific molecules described herein, the Antigen- Binding Domain of the Light Chain Variable Domain of the first polypeptide chain interacts with the Antigen-Binding Domain of the Heavy Chain Variable Domain of the second polypeptide chain in order to form a functional antigen-binding site that is specific for the first antigen (e.g., either HIV- 1 or CD3). Likewise, the Antigen-Binding Domain of the Light Chain Variable Domain of the second polypeptide chain interacts with the Antigen-Binding Domain of the Heavy Chain Variable Domain of the first polypeptide chain in order to form a second functional antigen-binding site that is specific for the second antigen (e.g., either CD3 or HIV-1, depending upon the identity of the first antigen). Thus, the selection of the Antigen-Binding Domain of the Light Chain Variable Domain and the Antigen-Binding Domain of the Heavy Chain Variable Domain of the first and second polypeptide chains are coordinated, such that the two polypeptide chains collectively comprise Antigen-Binding Domains of light and Heavy Chain Variable Domains capable of binding to the first and second antigens (e.g. , HIV-1 and CD3).

[0253] The Fc Domain of the Fc bearing multispecific molecules, including but not limited to bispecific and trispecific molecules (e.g. bispecific antibodies, bispecific diabodies, and trivalent binding molecules), of the present invention may be either a complete Fc Domain (e.g. , a complete IgG Fc Domain) or only a fragment of a complete Fc Domain. In some embodiments the Fc Domain of the molecules of the present invention may possess the ability to bind to one or more Fc receptors (e.g. , FcyR(s)). In non-limiting embodiments the Fc Domain will cause reduced binding to FcyRIA (CD64), FcyRIIA (CD32A), FcyRIIB

(CD32B), FcyRIIIA (CD 16a) or FcyRIIIB (CD 16b) (relative to the binding exhibited by a wild-type Fc Domain) or will substantially eliminate the ability of such Fc Domain to bind to such receptor(s). The Fc bearing multispecific molecules of the present invention may include some or all of the CH2 Domain and/or some or all of the CH3 Domain of a complete Fc Domain, or may comprise a variant CH2 and/or a variant CH3 sequence (that may include, for example, one or more insertions and/or one or more deletions with respect to the CH2 or CH3 domains of a complete Fc Domain). The Fc Domain of the Fc bearing multispecific molecules of the present invention may comprise non-Fc polypeptide portions, or may comprise portions of non-naturally complete Fc Domains, or may comprise non-naturally occurring orientations of CH2 and/or CH3 domains (such as, for example, two CH2 domains or two CH3 domains, or in the N-terminal to C-terminal direction, a CH3 Domain linked to a CH2 Domain, etc.).

[0254] Polymorphisms have been observed at a number of different positions within antibody constant regions (e.g. , Fc positions, including but not limited to positions 270, 272, 312, 315, 356, and 358 as numbered by the EU index as set forth in Kabat), and thus slight differences between the presented sequence and sequences in the prior art can exist. Polymorphic forms of human immunoglobulins have been well-characterized. At present, 18 Gm allotypes are known: Glm (1, 2, 3, 17) or Glm (a, x, f, z), G2m (23) or G2m (n), G3m (5, 6, 10, 1 1, 13, 14, 15, 16, 21, 24, 26, 27, 28) or G3m (b l, c3, b3, bO, b3, b4, s, t, gl, c5, u, v, g5) (Lefranc, et. al. , "The Human IgG Subclasses: Molecular Analysis Of Structure, Function And

Regulation " Pergamon, Oxford, pp. 43-78 (1990); Lefranc, G. et. al. , 1979, Hum. Genet. : 50, 199-21 1). It is specifically contemplated that the molecules of the present invention may be incorporate any allotype, isoallotype, or haplotype of any immunoglobulin gene, and are not limited to the allotype, isoallotype or haplotype of the sequences provided herein.

Furthermore, in some expression systems the C-terminal amino acid residue (bolded above) of the CH3 Domain may be post-translationally removed. Accordingly, the C-terminal residue of the CH3 Domain is an optional amino acid residue. Specifically encompassed by the instant invention are molecules lacking the C-terminal residue of the CH3 Domain. Also specifically encompassed by the instant invention are such constructs comprising the C- terminal lysine residue of the CH3 Domain. [0255] In non-limiting embodiments the first and third polypeptide chains of the Fc bearing multispecific molecules of the present invention each comprise CH2-CH3 domains that complex together to form an immunoglobulin (IgG) Fc Domain. The amino acid sequence of the CH2-CH3 domain of human IgGl is (SEQ ID NO: 527):

APELLGGPSV FLFPPKPKDT LMISRTPEVT CVVVDVSHED PEVKFNWYVD GVEVHNAKTK PREEQYNSTY RVVSVLTVLH QDWLNGKEYK CKVSNKALPA PIEKTISKAK GQPREPQVYT LPPSREEMTK NQVSLTCLVK GFYPSDIAVE WESNGQPENN YKTTPPVLDS DGSFFLYSKL TVDKSRWQQG NVFSCSVMHE ALHNHYTQKS LSLSPGK

[0256] Thus the CH2 and/or CH3 Domains of the first and third polypeptide chains may both be composed of SEQ ID NO: 527, or a variant thereof (e.g., SEQ ID NO: 528, 529, 530).

[0257] In non-limiting embodiments the CH2-CH3 domains of the first and third polypeptide chains of the Fc bearing multispecific molecules of the present invention exhibit decreased (or substantially no) binding to FcyRIA (CD64), FcyRIIA (CD32A), FcyRIIB (CD32B), FcyRIIIA (CD 16a) or FcyRIIIB (CD 16b) (relative to the binding exhibited by the wild-type Fc Domain). Fc variants and mutant forms capable of mediating such altered binding are well known in the art and include amino acid substitutions at positions 234 and 235, a substitution at position 265 or a substitution at position 297 (see, for example, US Patent No. 5,624,821, herein incorporated by reference). In non-limiting embodiments the CH2-CH3 Domain of the first and/or third polypeptide chains of the Fc bearing multispecific molecules of the present invention include a substitution at position 234 with alanine and 235 with alanine.

[0258] The CH2 and/or CH3 Domains of the first and third polypeptide chains need not be identical in sequence, and in some embodiment are modified to foster complexing between the two polypeptide chains. For example, an amino acid substitution (for example a substitution with an amino acid comprising a bulky side group forming a 'knob', e.g., tryptophan) can be introduced into the CH2 or CH3 Domain such that steric interference will prevent interaction with a similarly mutated domain and will obligate the mutated domain to pair with a domain into which a complementary, or accommodating mutation has been engineered, i.e. , 'the hole' (e.g. , a substitution with glycine). Such sets of mutations can be engineered into any pair of polypeptides comprising Fc bearing multispecific molecules of the invention, and further, engineered into any portion of the polypeptides chains of the pair. Methods of protein engineering to favor heterodimerization over homodimerization are well known in the art, in particular with respect to the engineering of immunoglobulin-like molecules, and are encompassed herein (see e.g., Ridgway et al. (1996) " 'Knobs-Into-Holes ' Engineering Of Antibody CH3 Domains For Heavy Chain Heterodimerization, " Protein Engr. 9:617-621, Atwell et al. (1997) "Stable Heterodimers From Remodeling The Domain Interface Of A Homodimer Using A Phage Display Library, " J. Mol. Biol. 270: 26-35, and Xie et al. (2005) Ά New Format Of Bispecific Antibody: Highly Efficient

Heterodimerization, Expression And Tumor Cell Lysis, " J. Immunol. Methods 296:95-101; each of which is hereby incorporated herein by reference in its entirety). In non-limiting embodiments the 'knob' is engineered into the CH2-CH3 Domains of the first polypeptide chain and the 'hole' is engineered into the CH2-CH3 Domains of the third polypeptide chain. Thus, the 'knob' will help in preventing the first polypeptide chain from homodimerizing via its CH2 and/or CH3 Domains. In non-limiting embodiments, as the third polypeptide chain contains the 'hole' substitution it will heterodimerize with the first polypeptide chain as well as homodimerize with itself. In non-limiting embodiments a knob is created by modifying a native IgG Fc Domain to contain the modification T366W. In non-limiting embodiments a hole is created by modifying a native IgG Fc Domain to contain the modification T366S, L368A and Y407V. To aid in purifying the third polypeptide chain homodimer from the final bispecific monovalent Fc bearing diabody comprising the first, second and third polypeptide chains, the protein A binding site of the CH2 and CH3 Domains of the third polypeptide chain is mutated by amino acid substitution at position 435 (H435R). Thus, the third polypeptide chain homodimer will not bind to protein A, whereas the bispecific monovalent Fc bearing diabody will retain its ability to bind protein A via the protein A binding site on the first polypeptide chain.

[0259] In non-limiting embodiments a sequence for the CH2 and CH3 Domains of the first polypeptide chain of the Fc bearing multispecific molecules of the present invention will have the "knob-bearin " se uence (SEQ ID NO: 531):

[0260] In non-limiting embodiments a sequence for the CH2 and CH3 Domains of the third polypeptide chain of the Fc bearing multispecific molecules of the present invention will have the "hole-bearin " se uence SE ID NO: 533 :

[0261] As will be noted, the CH2-CH3 Domains of SEQ ID NO: 531 and SEQ ID NO: 533 include a substitution at position 234 with alanine and 235 with alanine, and thus form an Fc Domain exhibit decreased (or substantially no) binding to FcyRIA (CD64), FcyRIIA

(CD32A), FcyRIIB (CD32B), FcyRIIIA (CD 16a) or FcyRIIIB (CD 16b) (relative to the binding exhibited by the wild-type Fc Domain (SEQ ID NO: 527).

[0262] In non-limiting embodiments, the first polypeptide chain will have a "knob-bearing" CH2-CH3 sequence, such as that of SEQ ID NO: 531 or 532. However, as will be recognized, a "hole-bearing" CH2-CH3 Domain (e.g. , SEQ ID NO: 533 or 534) could be employed in the first polypeptide chain, in which case, a "knob-bearing" CH2-CH3 Domain (e.g. , SEQ ID NO: 531 or 532) would be employed in the third polypeptide chain.

[0263] In non-limiting embodiments, the Fc domain can be modified by amino acid substitution to increase binding to the neonatal Fc receptor and therefore the half-life of the antibody when administered to a subject. The Fc domain can be an IgA, IgM, IgD, IgE or IgG Fc domain. The Fc domain can be an optimized Fc domain, as described in U.S.

Published Patent Application No. 20100093979, incorporated herein by reference. In certain embodiments the Fc bearing multispecific molecules comprise amino acid alterations, or combinations thereof, for example in the Fc domain(s) outside of epitope binding, which alterations can improve their properties. Various Fc modifications are known in the art. Amino acid numbering is according to the EU Index in Kabat. In some embodiments, the invention contemplates antibodies comprising mutations that affect neonatal Fc receptor (FcRn) binding, antibody half-life, and localization and persistence of antibodies at mucosal sites. See e.g. Ko SY et al. , Nature 514: 642-45, 2014, at Figure la and citations therein; Kuo, T. and Averson, V., mAbs 3(5): 422-430, 201 1, at Table 1, US Pub 20110081347 (an aspartic acid at Kabat residue 288 and/or a lysine at Kabat residue 435), US Pub

20150152183 for various Fc Domain mutation, incorporated by reference in their entirety. [0264] In certain embodiments, the Fc bearing multispecific molecules comprise AAAA substitution in and around the Fc Domain of the Fc bearing multispecific molecule that has been reported to enhance ADCC via NK cells (AAA mutations) containing the Fc Domain aa of S298A as well as E333A and K334A (Shields RI et al. JBC, 276: 6591-6604, 2001) and the 4 th A (N434A) is to enhance FcR neonatal mediated transport of the IgG to mucosal sites (Shields RI et al. ibid).

[0265] Other mutations have been reported to improve antibody half-life or function or both and can be incorporated into the Fc Domain of the Fc bearing multispecific molecules. These include the DLE set of mutations (Romain G, et al. Blood 124: 3241, 2014), the LS mutations M428L/N434S, alone or in a combination with other Fc Domain mutations, (Ko SY et al. Nature 514: 642-45, 2014, at Figure la and citations therein; Zlevsky et al, Nature

Biotechnology, 28(2): 157-159, 2010; US Pub 20150152183); the YTE Fc mutations (Robbie G et al, Antimicrobial Agents and Chemotherapy 12: 6147-53, 2013) as well as other engineered mutations to the Fc Domain such as QL mutations, IHH mutations (Ko SY et al. Nature 514: 642-45, 2014, at Figure la and relevant citations; See also Rudicell R et al, J. Virol 88: 12669-82, 201). In some embodiments, modifications, such as but not limited to fucosylation, which may affect interaction with Fc receptors (See e.g. Moldt, et al. JVI 86(11): 66189-6196, 2012). In some embodiments, the Fc bearing multispecific molecules can comprise modifications, for example but not limited to glycosylation, which reduce or eliminate polyreactivity of such a molecule. See e.g. Chuang, et al. Protein Science 24: 1019- 1030, 2015. In some embodiments the Fc bearing multispecific molecules can comprise modifications in the Fc domain such that the Fc domain exhibits, as compared to an unmodified Fc domain enhanced antibody dependent cell mediated cytotoxicity (ADCC); increased binding to FcyRIIA or to FcyRIIIA; decreased binding to FcyRIIB; or increased binding to FcyRIIB. See e.g. US Pub 20140328836.

[0266] In another aspect, the invention provides trivalent structures incorporating two diabody-type binding domains and one non-diabody-type domain and an Fc Domain (see, e.g., Figures 104A-104F and PCT Publication Nos. WO 2015/184207 and WO

2015/184203). Such trivalent binding molecules may be utilized to generate monospecific, bispecific or trispecific molecules. The ability to bind three different epitopes provides enhanced capabilities. [0267] A further embodiment of the present invention relates to trivalent binding molecules comprising an Fc Domain. The Fc Domain bearing trivalent binding molecules can simultaneously bind a first epitope, a second epitope, and a third epitope, wherein at least one of such epitopes is not identical to another. Such trivalent binding molecules comprise three epitope-binding sites, two of which are Diabody-Type Binding Domains, which provide binding Site A and binding Site B, and one of which is a Fab-Type Binding Domain, or an scFv-Type Binding Domain, which provides binding Site C (see, e.g. , Figures 104A-104F, and PCT Publication Nos. WO 2015/184207 and WO 2015/184203). Such trivalent binding molecules thus comprise "VL1" / "VH1" domains that are capable of binding to the first epitope and "VL2" / "VH2" domains that are capable of binding to the second epitope and "VL3" and "VH3" domains that are capable of binding to the "third" epitope of such trivalent binding molecule. A "Diabody-Type Binding Domain" is the type of epitope-binding site present in a diabody, and especially, a DART® diabody, as described above. Each of a "Fab- Type Binding Domain" and an "scFv-Type Binding Domain" are epitope-binding sites that are formed by the interaction of the VL Domain of an immunoglobulin light chain and a complementing VH Domain of an immunoglobulin heavy chain. Fab-Type Binding Domains differ from Diabody-Type Binding Domains in that the two polypeptide chains that form a Fab-Type Binding Domain comprise only a single epitope-binding site, whereas the two polypeptide chains that form a Diabody-Type Binding Domain comprise at least two epitope- binding sites. Similarly, scFv-Type Binding Domains also differ from Diabody-Type Binding Domains in that they comprise only a single epitope-binding site. Thus, as used herein Fab- Type, and scFv-Type Binding Domains are distinct from Diabody-Type Binding Domains.

[0268] Typically, the trivalent binding molecules of the present invention will comprise four different polypeptide chains (see Figures 104A-104B), however, the molecules may comprise fewer or greater numbers of polypeptide chains, for example by fusing such polypeptide chains to one another (e.g., via a peptide bond) or by dividing such polypeptide chains to form additional polypeptide chains, or by associating fewer or additional polypeptide chains via disulfide bonds. Figures 104C-104F illustrate this aspect of the present invention by schematically depicting such molecules having three polypeptide chains. As provided in Figures 104A-104F, the trivalent binding molecules of the present invention may have alternative orientations in which the Diabody-Type Binding Domains are N-terminal (Figures 104A, 104C and 104D) or C-terminal (Figures 104B, 104E and 104F) to an Fc Domain. [0269] In certain embodiments, the first polypeptide chain of such trivalent binding molecules of the present invention contains: (i) a VL1 -containing Domain, (ii) a VH2 -containing Domain, (iii) a Heterodimer-Promoting Domain, and (iv) a Domain containing a CH2-CH3 sequence. The VL1 and VL2 Domains are located N-terminal or C-terminal to the CH2-CH3- containing domain as presented in Table 3 (also see, Figures 104A and 104B). The second polypeptide chain of such embodiments contains: (i) a VL2-containing Domain, (ii) a VH1- containing Domain, and (iii) a Heterodimer-Promoting Domain. The third polypeptide chain of such embodiments contains: (i) a VH3 -containing Domain, (ii) a CHI -containing Domain and (iii) a Domain containing a CH2-CH3 sequence. The third polypeptide chain may be the heavy chain of an antibody that contains a VH3 and a heavy chain constant region, or a polypeptide that contains such domains. The fourth polypeptide of such embodiments contains: (i) a VL3 -containing Domain and (ii) a CL-containing Domain. The fourth polypeptide chains may be a light chain of an antibody that contains a VL3 complementary to the VH3 of the third polypeptide chain, or a polypeptide that contains such domains. The third or fourth polypeptide chains may be isolated from naturally occurring antibodies.

Alternatively, they may be constructed recombinantly, synthetically or by other means.

[0270] The Light Chain Variable Domain of the first and second polypeptide chains are separated from the Heavy Chain Variable Domains of such polypeptide chains by an intervening spacer peptide having a length that is too short to permit their VL1/VH2 (or their VL2/VH1) domains to associate together to form epitope-binding site capable of binding to either the first or second epitope. A preferred intervening peptide linker (Peptide Linker 1) for this purpose has the sequence (SEQ ID NO:508): GGGSGGGG. Other Domains of the trivalent binding molecules may be separated by one or more intervening peptide linkers (Peptide Linkers), optionally comprising a cysteine residue. In particular, as provided above, such Peptide Linkers will typically be incorporated between Variable Domains (i.e. , VH or VL) and peptide Heterodimer-Promoting Domains (e.g. , an E-coil or K-coil) and between such peptide Heterodimer-Promoting Domains (e.g. , an E-coil or K-coil) and CH2-CH3 Domains. Exemplary peptide linkers (e.g. , Peptide Linker 2, Peptide Linker 2-C, Peptide Linker 3, Spacer Linker 3, etc. ) useful for the generation of trivalent binding molecules are provided above. Such linkers are also provided in PCT Publication Nos. WO 2015/184207 and WO 2015/184203. Thus, the first and second polypeptide chains of such trivalent binding molecules associate together to form a VL1/VH1 binding site capable of binding a first epitope, as well as a VL2/VH2 binding site that is capable of binding to a second epitope. The third and fourth polypeptide chains of such trivalent binding molecules associate together to form a VL3/VH3 binding site that is capable of binding to a third epitope.

[0271] As described above, the trivalent binding molecules of the present invention may comprise three polypeptides. Trivalent binding molecules comprising three polypeptide chains may be obtained by linking the domains of the fourth polypeptide N-terminal to the VH3 -containing Domain of the third polypeptide (e.g., using an intervening spacer peptide (Peptide Linker 5)). Alternatively, a third polypeptide chain of a trivalent binding molecule of the invention containing the following domains is utilized: (i) a VL3 -containing Domain, (ii) a VH3 -containing Domain, and (iii) a Domain containing a CH2-CH3 sequence, wherein the VL3 and VH3 are spaced apart from one another by an intervening spacer peptide that is sufficiently long (at least 9 or more amino acid residues) so as to allow the association of these domains to form an epitope-binding site. One preferred intervening spacer peptide for this purpose has the sequence: GGGGSGGGGSGGGGS (SEQ ID NO: 526).

[0272] It will be understood that the VLl/VHl, VL2/VH2, and VL3/VH3 Domains of such trivalent binding molecules may be different so as to permit binding that is monospecific, bispecific, or trispecific. In particular, the VL and VH Domains may be selected such that a trivalent binding molecule comprises two binding sites for a first epitope and one binding sites for a second epitope, or one binding site for a first epitope and two binding sites for a second epitope, or one binding site for a first epitope, one binding site for a second epitope and one binding site for a third epitope.

[0273] In one embodiment, these domains are selected so as to bind an epitope of HIV- 1 Env, an epitope of second molecule, and an epitope of a third molecule, wherein the second molecule and the third molecule (e.g. , CD2, CD3, CD 8, CD16, T-Cell Receptor (TCR), NKG2D, etc.) are present on the surface of an effector cell, such as a T lymphocyte, a natural killer (NK) cell or other mononuclear cell.

[0274] The general structure of the polypeptide chains of representative trivalent binding molecules of invention is provided in Figures 104A-104F and in Table 3:

[0275] One embodiment of the present invention relates to trivalent binding molecules that comprise two epitope-binding sites for HIV-1 Env and one epitope-binding site for a second molecule. The two epitope-binding sites for HIV-1 Env may bind the same epitope or different epitopes. Another embodiment of the present invention relates to trivalent binding molecules that comprise, one epitope-binding site for HIV- 1 Env and two epitope-binding sites for a second molecule. The two epitope-binding sites for the second molecule may bind the same epitope or different epitopes of the second molecule. A further embodiment of the present invention relates to trispecific trivalent binding molecules that comprise, one epitope- binding site for HIV-1 Env, one epitope-binding site for a second molecule, and one epitope- binding site for a third molecule. In certain embodiments, the second molecule is a molecule (e.g. , CD!, CD3, CD8, CD 16, T-Cell Receptor (TCR), NKG2D, etc.) present on the surface of an effector cell, such as a T lymphocyte, a natural killer (NK) cell or other mononuclear cell. In certain embodiments, the second molecule is CD3 and the third molecule is CD8. As provided above, such trivalent binding molecules may comprise three, four, five, or more polypeptide chains. [0276] In one embodiment, these domains are selected so as to bind two epitopes of HIV- 1 Env, which may be the same epitopes or different epitopes, and an epitope of second molecule, wherein the second molecule is a molecule (e.g., CD2, CD3, CD8, CD 16, T-Cell Receptor (TCR), NKG2D, etc.) present on the surface of an effector cell, such as a T lymphocyte, a natural killer (NK) cell or other mononuclear cell. In a specific embodiment, the two epitopes of HIV- 1 Env are the same, and the second molecule is CD3. In an alternative embodiment, the two epitopes of HIV- 1 Env are different, and the second molecule is CD3.

[0277] The molecules, and fragments thereof, described above can be formulated as a composition (e.g., a pharmaceutical composition). Suitable compositions can comprise the molecules (or fragments thereof) in a pharmaceutically acceptable carrier e.g., dissolved or dispersed in an aqueous medium, or lyophilized. The compositions can be sterile and can be in an injectable form (e.g. but not limited to a form suitable for intravenous injection, or intramuscular injection). The molecules (and fragments thereof) can also be formulated as a composition appropriate for topical administration to the skin or mucosa. Such compositions can take the form of liquids, ointments, creams, gels and pastes. The molecules (and fragments thereof) can also be formulated as a composition appropriate for intranasal administration. The molecules (and fragments thereof) can be formulated so as to be administered as a post-coital douche or with a condom. Standard formulation techniques can be used in preparing suitable compositions.

[0278] In certain embodiments the invention provides multispecific molecules such as but not limited to bispecific and trispecific molecules (e.g. , bispecific antibodies, bispecific diabodies, trivalent binding molecules, etc.) comprising the binding domains from human, humanized and/or chimeric antibodies. Methods to construct such antibodies are well known in the art.

[0279] In certain aspects the invention provides use of the multispecific molecules of the invention such as but not limited to bispecific and trispecific molecules (e.g., bispecific antibodies, bispecific diabodies, trivalent binding molecules, etc.), in methods of treating and preventing HIV-1 infection in an individual, comprising administering to the individual a therapeutically effective amount of a composition comprising a multispecific molecule of the invention in a pharmaceutically acceptable form. In certain embodiment, the methods include a composition which includes more than one HIV-1 targeting multispecific molecule. In certain embodiments, the HIV- 1 targeting multispecific molecule in such combination bind different epitopes on the HIV-1 envelope. In certain embodiments, such combinations of multispecific molecule targeting more than one HIV- 1 epitope provide increased killing of HIV-1 infected cells. In other embodiments, such combinations of multispecific molecule targeting more than one HIV-1 epitope provide increased breadth in recognition of different HIV-1 subtypes.

[0280] The various multispecific molecule described herein have utility, for example, in settings including but not limited to the following:

i) in the setting of anticipated known exposure to HIV-1 infection, the multispecific molecule described herein can be administered prophylactically (e.g. , IV, topically or intranasally) as a microbiocide,

ii) in the setting of known or suspected exposure, such as occurs in the setting of rape victims, or commercial sex workers, or in any homosexual or heterosexual transmission without condom protection, the multispecific molecule described herein can be administered as post-exposure prophylaxis, e.g. , IV or topically, and

iii) in the setting of Acute HIV-1 infection (AHI), the multispecific molecule described herein can be administered as a treatment for AHI to control the initial viral load, or for the elimination of virus-infected CD4 T cells.

[0281] In accordance with the invention, the multispecific molecules described herein can be administered prior to contact of the subject or the subject's immune system/cells with HIV-1 or within about 48 hours of such contact. Administration within this time frame can maximize inhibition of infection of vulnerable cells of the subject with HIV-1.

[0282] In addition, various forms of the multispecific molecules described herein can be administered to chronically or acutely infected HIV-1 patients and used to kill remaining virus infected cells by virtue of these multispecific molecule binding to the surface of virus infected cells and being able to mediate redirected cell killing of such infected cells.

[0283] In certain embodiments, the multispecific molecules of the invention can be administered in combination with latency activating agents, so as to activate latent reservoir of HIV-1 -infected cells. The expectation is that by activating latent proviral HIV-1 DNA in resting cells, once inactive cells will start producing new virus and they will be recognized and eliminated by the immune system. Non-limiting examples of latency activating agents are HDAC inhibitors, e.g., vorinostat, romidepsin, panobinostat, disulfiram, JQ 1, bryostatin, PMA, inonomycin, or any combination thereof. See Bullen et al. Nature Medicine 20, 425- 429 (2014).

[0284] In certain embodiments the multispecific molecules of the invention can be administered in combination with anti -retroviral agents.

[0285] Suitable dose ranges can depend on the multispecific molecule and on the nature of the formulation and route of administration. Optimum doses can be determined by one skilled in the art without undue experimentation. For example, doses of antibodies in the range of 1-50 mg/kg of unlabeled or labeled antibody (with toxins or radioactive moieties) can be used. If antibody fragments, with or without toxins are used or antibodies are used that can be targeted to specific CD4 infected T cells, then less antibody can be used (e.g. , from 5 mg/kg to 0.01 mg/kg). If multispecific molecules are used, doses in the range of 0.01 μg/kg to about 30 mg/kg or more of the subject's body weight can be used. Suitable dose ranges can depend on the antibody (or fragment, or multispecific molecule) and on the nature of the formulation and route of administration. Optimum doses can be determined by one skilled in the art without undue experimentation. For example but not limited, doses of multispecific molecules in the range of O.01-100 μg/kg, 0.1-50 mg/kg, 1-50 mg/kg, 1- lO mg/kg, 1, 1.25, 1.5, 2, 2.5, 3, 3.5, 4, 4.5, 5, 5.5, 6, 6.5, 7, 7.5, 8, 8.5, 9, 9.5, 10, 1 1, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 mg/kg of unlabeled or labeled multispecific molecule (with toxins or radioactive moieties) can be used. If antibody fragments, with or without toxins are used or antibodies are used that can be targeted to specific CD4 infected T cells, then less antibody can be used (e.g. , from 5 mg/kg to 0.01 mg/kg). In other embodiments, the molecules of the invention can be administered at a suitable fixed dose, regardless of body size or weight. See Bai et al. Clinical Pharmacokinetics February 2012, Volume 51, Issue 2, pp 1 19-135.

[0286] Multispecific molecules of the invention can be produced recombinantly using nucleic acids comprising nucleotide sequences encoding VH and VL sequences selected from those shown in the figures and examples, or those known in the art.

[0287] In certain embodiments the invention provides multispecific binding molecules comprising antigen binding fragments. Typically, multispecific binding molecules compete with the intact antibody from which they were derived for specific binding to the target including separate heavy chains, light chains Fab, Fab', F(ab').sub.2, F(ab)c, diabodies, Dabs, nanobodies, and Fv. Fragments that can be produced by recombinant DNA techniques, or by enzymatic or chemical separation of intact immunoglobulins.

[0288] Nucleic acid sequences encoding polypeptides for the production of multispecific molecules with specificities as described herein can be used to produce plasmids for stable expression of such multispecific molecules. Methods for recombinant expression and purification are known in the art. In certain embodiments of Fc bearing multispecific molecules, the plasmids also comprise any of the changes to the Fc portion described herein.

[0289] In certain embodiments, the nucleic acids are optimized for recombinant expression in a suitable host cell. In certain embodiments, the vector is suitable for gene delivery and expression. There are numerous expression systems available for expression of proteins including E. coli, other bacterial hosts, yeast, and various higher eukaryotic cells such as the COS, CHO, HeLa and myeloma cell lines.

[0290] Any suitable cell line can be used for expression of the polypeptides of the invention, including but not limited to CHO cells, 293T cells. In some aspects, the invention provides nucleic acids encoding these antibodies, expression cassettes and vectors including these nucleic acids, and isolated cells that express the nucleic acids which encode the multispecific molecules of the invention are also provided. The polypeptides of the invention can be purified by any suitable method for purification of polypeptides and/or antibodies.

[0291] Table 4. Summary listing of various sequences listed throughout the specification; starting at SEQ ID NO: 500

[0292] Various antigen binding domains capable of binding to an epitope of HIV- 1, CD3, CD 16 and CD8 are contemplated by the invention and are disclosed in the specification. Table 4 discloses non-limiting embodiments of such antigen binding domains which may be incorporated into the multispecific molecules of the invention. Additional, alternative antigen binding domains from other antibodies having specificity for the desired antigens may be utilized. Many such antibodies are known in the art, for example additional anti-CD3 antibodies are described in WO2012/162067 and WO 2014/110601 the contents of each of which are hereby incorporated by reference; additional anti-CD 16 antibodies are described in WO 03/101485 the contents of which is hereby incorporated by reference; and additional anti- CD8 antibodies are described in WO 2014/164553 the contents of which is hereby incorporated by reference.

[0293] Various Heterodimer-Promoting Domain (HPD) sequences are contemplated by the invention and disclosed in the specification. Table 4 discloses non-limiting embodiments of various HPDs. In some embodiments the HPD include E/K-coils (SEQ ID NOs: 518, 510) or cysteine engineered E/K-coils (SEQ ID NOs: 519, 521). In some embodiments HPD includes combinations of SEQ ID NOs: 51 1, 512, 513, and 514 sequences (e.g., SEQ ID NOs: 511 and 513; SEQ ID NOs: 512 and 513; SEQ ID NOs: 51 1 and 514; SEQ ID NOs: 512 and 514); kappa and lambda light chain constant domains (SEQ ID NOs: 516 and 517). In some embodiments HPD include any suitable sequences with a Cysteine residue to permit disulfide bond. In some embodiments HPD includes suitable CHI and CL domains.

[0294] A skilled artisan readily appreciates that the disclosure throughout the application of various design elements such as but not limited to HPDs and sequences, linkers, CH2-CH3 domain and Fc domain and their variants is applicable and could be used in any of the designs of the multispecific molecules of the invention that comprise these design elements.

[0295] HIV-1 Antibodies

[0296] Broadly neutralizing and potent HIV-1 envelope glycoprotein (Env) antibodies are now being developed for both prevention of HIV-1 (Rudicell RS et al. J. Virol 88: 12669-82, 2014) and for treatment of HIV-1 infected individuals (Barouch DH, et al. Nature 503 : 224-8, 2013; Shingai M et al. Nature 503 : 277-80, 2013). Thus, human recombinant antibodies either alone or in combinations have great prophylactic and therapeutic potential for the prevention and treatment of HIV-1 infection. Moreover, antibodies that bind with high affinity to Env may be useful in eliminating the latent pool of HIV- 1 -infected CD4 T cells and curing HIV-1 infection, when either used to sensitize HIV-1 expressing target cells with bispecific bnAbs for NK or CD8 T cell killing or when bnAbs are conjugated with toxins or radionucleotides.

[0297] In certain aspects the invention provides fully human antibodies and fragments that specifically bind to and potently neutralize various isolates of HIV-1. In some embodiments, the antibodies bind to HIV-1 env gp41 membrane-proximal extracellular region (MPER). In some embodiments, the antibodies of the invention are combined in compositions with antibodies to other HIV-1 env binding sites. [0298] In certain aspects the invention provides pharmaceutical compositions including these human antibodies and a pharmaceutically acceptable carrier. In certain aspects the invention provides antibodies for passive immunization against HIV/AIDS. Nucleic acids encoding these antibodies, expression cassettes and vectors including these nucleic acids, and isolated cells that express the nucleic acids which encode the antibodies of the invention are also provided.

[0299] In some embodiments, the invention provides antibodies which are clonal variants. In some embodiments, clonal variants are sequences that differ by one or more nucleotides or amino acids, and have a V region with shared mutations compared to the germline, identical VHDJH or VJH gene usage, identical or similar HCDR3 length, and the same VL and JL usage. The germline sequence (unmutated common ancestor "UCA") is intended to be the sequence coding for the antibody/immunoglobulin (or of any fragment thereof) deprived of mutations, for example somatic mutations. Antibodies in a clone that are designated as UCA and/or I (for "Intermediate") are typically not identified from a biological sample, but are derived computationally based on VH and/or VL sequences isolated from subjects infected with HIV- 1.

[0300] Compositions including the human antibodies of the invention, including gp41 MPER binding antibodies, can be used for any purpose including but not limited to research, diagnostic and therapeutic purposes.

[0301] The neutralization breadth of the inventive antibodies is demonstrated by the diversity of viruses which are neutralized in the TZMbl Env pseudovirus inhibition assay. In certain embodiments, the neutralization breadth and/or binding of the antibodies of the invention can be maintained in the presence of tolerate changes to the epitope. Comparing the sequences of the neutralized viruses, versus viruses that are not neutralized, a skilled artisan can readily determine the % virus changes, including changes in the epitope, which can be tolerated while neutralization and/or binding is maintained.

[0302] Comparing the sequences of the antibodies and their neutralization properties, a skilled artisan can readily determine sequence identity, compare sequence length and determine the % sequence identity and/or changes, including % sequence identity and/or changes in the VH and VL sequences, including % sequence identity and/or changes in the CDRs, as well as the specific positions and types of substitutions which can be tolerated while neutralization potency and breadth is maintained. [0303] Various algorithms for sequence alignment are known in the art. The similarity between amino acid sequences is expressed in terms of the similarity between the sequences, otherwise referred to as sequence identity. Sequence identity is frequently measured in terms of percentage identity (or similarity or homology); the higher the percentage, the more similar the two sequences are. Homologs or variants of a polypeptide will possess a relatively high degree of sequence identity when aligned using standard methods.

[0304] Methods of alignment of sequences for comparison are well known in the art. Various programs and alignment algorithms are described in: Smith and Waterman, Adv. Appl. Math. 2:482, 1981 ; Needleman and Wunsch, J. Mol. Biol. 48:443, 1970; Pearson and Lipman, Proc. Natl. Acad. Sci. U.S.A. 85 :2444, 1988; Higgins and Sharp, Gene 73 :237, 1988; Higgins and Sharp, CABIOS 5 : 151, 1989; Corpet et al. , Nucleic Acids Research 16: 10881, 1988; and Pearson and Lipman, Proc. Natl. Acad. Sci. U.S.A. 85:2444, 1988. Altschul et al , Nature Genet. 6: 1 19, 1994, presents a detailed consideration of sequence alignment methods and homology calculations.

[0305] The NCBI Basic Local Alignment Search Tool (BLAST) (Altschul et al , J. Mol. Biol. 215 :403, 1990) is available from several sources, including the National Center for

Biotechnology Information (NCBI, Bethesda, Md.) and on the internet, for use in connection with the sequence analysis programs blastp, blastn, blastx, tblastn and tblastx. A description of how to determine sequence identity using this program is available on the NCBI website on the internet.

[0306] Homologs and variants of a VL or a VH of an antibody that specifically binds a polypeptide are typically characterized by possession of at least about 75%, for example at least about 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% sequence identity counted over the full length alignment with the amino acid sequence of interest. Proteins with even greater similarity to the reference sequences will show increasing percentage identities when assessed by this method, such as at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, or at least 99% sequence identity. When less than the entire sequence is being compared for sequence identity, homologs and variants will typically possess at least 80% sequence identity over short windows of 10-20 amino acids, and may possess sequence identities of at least 85% or at least 90% or 95% depending on their similarity to the reference sequence. Methods for determining sequence identity over such short windows are available at the NCBI website on the internet. One of skill in the art will appreciate that these sequence identity ranges are provided for guidance only; it is entirely possible that strongly significant homologs could be obtained that fall outside of the ranges provided.

[0307] As used herein, reference to antibodies, without explicit mention of antibody fragments and antibody-fragment comprising molecules, may encompass antibody fragments and antibody-fragment comprising molecules.

[0308] In certain embodiments, the invention provides antibodies and antibody-fragment comprising molecules, including multispecific molecules, such as, but not limited to bispecific and trispecific molecules (e.g., bispecific antibodies, bispecific diabodies, trivalent binding molecules, etc.) which are 99%, 98%, 97%, 96%, 95%, 94%, 93%, 92%, 91%, 90%, 89%, 88%, 87%, 86%, 85%, 84%, 83%, 82%, 81%, 80% identical to the VH and VL amino acid sequences of the antibodies described herein and still maintain the neutralization breadth, biding and/or potency. In certain embodiments, the invention provides antibodies and antibody-fragment comprising molecules, including multispecific molecules, such as, but not limited to bispecific and trispecific molecules (e.g., bispecific antibodies, bispecific diabodies, trivalent binding molecules, etc.) which are 99%, 98%, 97%, 96%, 95%, 94%, 93%, 92%, 91%, 90%, 89%, 88%, 87%, 86%, 85%, 84%, 83%, 82%, 81%, 80% identical to the CDRl, 2, and/or 3 of VH and CDRl, 2, and/or 3 VL amino acid sequences of the antibodies described herein and still maintain the neutralization breadth, biding and/or potency.

[0309] In certain embodiments, the invention provides antibodies and antibody-fragment comprising molecules, including multispecific molecules, such as, but not limited to bispecific and trispecific molecules (e.g., bispecific antibodies, bispecific diabodies, trivalent binding molecules, etc.) which can tolerate a larger percent variation in the sequences outside of the VH and/VL sequences of the antibodies. In certain embodiments, the invention provides antibodies which are 99%, 98%, 97%, 96%, 95%, 94%, 93%, 92%, 91%, 90%, 89%, 88%, 87%, 86%, 85%, 84%, 83%, 82%, 81%, 80%, 79%, 78%, 77%, 76%, 75%, 74%, 73%, 72%, 71%, 70%, 69%, 68%, 67%, 66%, 65% identical, wherein the identity is outside of the VH or VL regions, or the CDRs of the VH or VL chains of the antibodies described herein.

[0310] Antibodies of the invention are expected to have the same binding specificity, for example as intact immunoglobulins and antigen binding variants or fragments e.g. as a number of well characterized fragments produced by digestion with various peptidases. For instance, and without limitation, Fabs, Fvs, scFvs are fragments which are expected to have the same binding specificities as intact antibodies. Binding specificity can be determined by any suitable assay in the art, for example but not limited competition binding assays, epitope mapping, etc. Assays to determine glycan dependence and glycan specificity binding are also known in the art. A scFv protein is a fusion protein in which a light chain variable region of an immunoglobulin and a heavy chain variable region of an immunoglobulin are bound by a linker, while in dsFvs, the chains have been mutated to introduce a disulfide bond to stabilize the association of the chains. Provided are also genetically engineered forms such as chimeric antibodies and heteroconjugate antibodies such as bispecific antibodies. See also, Pierce Catalog and Handbook, 1994-1995 (Pierce Chemical Co., Rockford, III); Kuby,

Immunology, 3.sup.rd Ed., W.H. Freeman & Co., New York, 1997.

[0311] In certain embodiments the invention provides antibody fragments and molecules comprising antibody fragments, which have the binding specificity and/or properties of the inventive antibodies. Non-limiting examples include: (1) Fab, the fragment which contains a monovalent antigen-binding fragment of an antibody molecule produced by digestion of whole antibody with the enzyme papain to yield an intact light chain and a portion of one heavy chain; (2) Fab', the fragment of an antibody molecule obtained by treating whole antibody with pepsin, followed by reduction, to yield an intact light chain and a portion of the heavy chain; two Fab' fragments are obtained per antibody molecule; (3) (Fab').sub.2, the fragment of the antibody obtained by treating whole antibody with the enzyme pepsin without subsequent reduction; (4) F(ab').sub.2, a dimer of two Fab' fragments held together by two disulfide bonds; (5) Fv, a genetically engineered fragment containing the variable region of the light chain and the variable region of the heavy chain expressed as two chains; and (6) single chain antibody ("SCA"), a genetically engineered molecule containing the variable region of the light chain, the variable region of the heavy chain, linked by a suitable polypeptide linker as a genetically fused single chain molecule. In certain embodiments, the antibody fragments can be produces recombinantly.

[0312] In certain embodiments, VH refers to the variable region of an immunoglobulin heavy chain, including but not limited to that of an antibody fragment, such as Fv, scFv, dsFv or Fab. In certain embodiments, VL refers to the variable region of an immunoglobulin light chain, including but not limited to that of an Fv, scFv, dsFv or Fab.

[0313] Any of the nucleic acids encoding any of the antibodies, or fragment thereof can be expressed in a recombinantly engineered cell such as bacteria, plant, yeast, insect and mammalian cells. The nucleic acid sequences include any sequence necessary for expression, including but not limited to a promoter, a leader sequence. These antibodies can be expressed as individual VH and/or VL chain, or can be expressed as a fusion protein. In certain embodiments, the antibodies can be expressed by viral vector mediated delivery of genes encoding the antibodies of the invention (See e.g. Yang et al. Viruses 2014, 6, 428-447).

[0314] The present invention also encompasses molecules comprising a hinge domain. The hinge domain be derived from any immunoglobulin isotype or allotype including IgA, IgD, IgG, IgE and IgM. In preferred embodiments, the hinge domain is derived from IgG, wherein the IgG isotype is IgGl, IgG2, IgG3 or IgG4, or an allotype thereof. The hinge domain may be engineered into a polypeptide chain comprising the diabody molecule together with an Fc domain such that the diabody molecule comprises a hinge-Fc domain. In certain

embodiments, the hinge and Fc domain are independently selected from any immunoglobulin isotype known in the art or exemplified herein. In other embodiments the hinge and Fc domain are separated by at least one other domain of the polypeptide chain, e.g. , the VL domain.

[0315] Various antibodies names are used throughout the application. Antibodies names correlation is as follows: Memory B cell antibodies: DH511=DH51 1.1 ; DH512=DH51 1.2; DH513=DH51 1.3; DH514=DH51 1.4; DH515=DH51 1.5; DH516=DH51 1.6;

[0316] Plasma antibodies: DH51 l_la=DH51 1 7P; DH51 l_2a=DH51 1.8P;

DH51 l_3a=DH51 1 9P; DH51 l_4a=DH51 1.10P; DH51 l_5a=DH51 1. I IP;

DH51 1_5b=DH51 1.12P.

[0317] Chimeric antibodies which combine a heavy and light chain from different antibodies are typically indicated by the designation of the heavy and light chain of each parent antibody.

[0318] Mutations in the VH chain are referenced with respect to Kabat numbering of the indicated VH chain.

[0319] It is readily understood that the nucleic acid sequences disclosed in the application are non-limiting embodiments of representative nucleotide sequences encoding the respective amino acid sequences.

[0320] The contents of the various publications cited throughout the specification are incorporated by reference in their entirety.

[0321] The following examples are provided to illustrate particular features of certain embodiments, but the scope of the claims should not be limited to those features exemplified. EXAMPLES

[0322] The following examples are provided to illustrate particular features of certain embodiments, but the scope of the claims should not be limited to those features exemplified.

Example 1: MPER antibodies

[0323] Figure 1 shows the three HIV infected individual plasma that was evaluated for HIV neutralizing activity and the specificities profiled by the Georgiev algorithm (Georgiev IS et al Science 340: 751-6, 2013). From this analysis we found three subjects (CH0210, CH0536, CH1244) with gp41 bnAb activity (Figure 1).

[0324] Methods to identify and isolate MPER reactive antibodies were carried out as described in Liao HX et al. J. Virol. Methods 158: 171-9, 2009. MPER specific hooks were designed to identify to antibodies which bind to HIV-1 gp41 MPER region. Using one such hook, the MPR.03-biotin hook tetramerized (Figure 2), with fluorophor labeled streptavidin in two colors (Figure 3), we sorted by flow cytometry into single wells, the diagonally (that reacted with both colors hooks) reactive memory B cells (Figure 3). B cells from 10 million PBMC were sorted and PCR was carried out according to the protocol in Liao HX et al. J. Virol. Methods 158: 171-9, 2009. PCR amplifications were carried out to amplify rearranged VH and VL fragment pairs from the diagonally sorted memory B cells (Liao et al JVM). Overlapping PCR was used to construct full length Ig heavy and Ig light linear genes comprising the rearranged VH and VL fragment pairs. RT-PCR and PCR reactions was carried out essentially as described in Liao HX et al. J. Virol. Methods 158: 171-9, 2009, see for example Figure 1, Section 3.3. Sequence analysis of the VH and VL genes was carried out to determine the VH and VL gene usage, CDR lengths, the % mutation of HCDR3 and LCDR3. Based on this sequence analysis, one to two pairs of linear VH and VL genes were selected and made in linear cassettes (essentially as described in Liao HX et al. J. Virol. Methods 158: 171-9, 2009, see for example Figure 1, Section 3.3) to produce recombinant monoclonal antibodies by transient transfection in 293T cells.

[0325] Pairs of VH and VL genes as selected above can also be used to produce plasmids for stable expression of recombinant antibodies.

[0326] In certain embodiments, the plasmids or linear constructs for recombinant antibody expression also comprise AAAA substitution in and around the Fc region of the antibody that has been reported to enhance ADCC via NK cells (AAA mutations) containing the Fc region aa of S298A as well as E333A and K334A (Shields RI et al JBC, 276: 6591-6604, 2001) and the 4 th A (N434A) is to enhance FcR neonatal mediated transport of the IgG to mucosal sites (Shields RI et al. ibid).

[0327] The antibodies of the invention were selected based on a combination of criteria including sequence analyses, and functional analyses including but not limited as neutralization breadth, and potency.

[0328] In certain embodiments, the antibodies of the invention comprise naturally rearranged VH and VL fragment pairs, wherein the rest of the Ig gene is not naturally occurring with the isolated rearranged VH and VL fragments. In certain embodiments, the antibodies of the invention are recombinantly produced by synth

[0329] Figure 4 and Example 12 shows a summary of some of the characteristics of the recombinant MPER antibodies of the invention. DH51 1-DH517 are antibodies with VH and VL chains from individual CH0210. DH518 is an antibody with VH and VL chains from individual CH0536. DH536 is an antibody with VH and VL chains from individual CH1244. CH537 is an antibody with VH and VL chains from individual CH0585. DH 51 1-DH516 antibodies are all members of the same B cell clonal lineage (Figure 6). Figure 5 shows the neutralizing capacity of these antibodies with all but DH536 and DH537 able to neutralize difficult to neutralized (tier 2) HIV strains B.BG1 168, C.CH505, and C.DU172). Figure 6 shows the phylogram of the DH51 1 clonal lineage.

Example 2: TZMbl neutralization assay

[0330] TZMbl neutralization assay is a standard way to evaluate antibody breadth and potency. See Montefiori, D. Methods Mol Biol. 2009;485 :395-405; HIV-1 Env- pseudoviruses infection of TZM-bl cells. Exemplary pseudovirus neutralization assays and panels of HIV-1 pseudovirus are described for example, in Li et al., J Virol 79, 10108- 10125, 2005, Seaman et al, J. Virol., 84: 1439-1452, 2010; Sarzotti-Kelsoe et al, J. Immunol.

Methods, 409: 131-46, 2014; and WO201 1/038290, each of which is incorporated by reference herein. Various HIV-1 isolates, both Tier 1 and Tier 2 viruses can be included in this assay.

[0331] The TZMbl assay was conducted to determine neutralization potency and breadth of the various antibodies of the invention on different HIV-1 pseudoviruses. [0332] Figure 7 shows the results of neutralization of 8 of the gp41 antibodies against a panel of 30 HIV tier 2 isolates in the TZMbl pseudovirus neutralization assay. The DH511 clonal lineage members all neutralized 100% (30/30) isolates while DH517 neutralized 50% and DH518 neutralized 83%. This in contrast to 10E8 gp41 antibody that only neutralized 29/30 isolates. Figure 8 shows the mean IC50, IC80 and percent of isolates neutralized at an IC50 <50ug/ml and at an IC80 of <5ug/ml (confirm). Thus, mAb DH512 is equally as potent and slightly more broad in neutralization breadth than the mAb 10E8. Figure 9 shows other mAbs and their breadth and potency. Various figures, including without limitation, Figures 37, 38, 28, 56 and 34, and Figures from Example 12 show neutralization data of various antibodies against various panels of pseudoviruses.

Example 3: Epitope mapping of MPER antibodies

[0333] Binding of antibodies to various MPER peptides in an ELISA assay was used to map the epitopes of the MPER antibodies.

[0334] Figure 11 shows that Antibody epitopes maps to the C-terminus of gp41 to a similar region where 10E8 binds (Huang J et al. Nature 491 406, 2012; See US Pub 20140348785). Figures 1 1, 15-25 show binding of antibodies to MPER peptide variants. These mapping studies show that the antibodies of the invention are 10E8 like Abs. In non-limiting embodiments, DH512 shows the broadest and most potent neutralization among the antibodies tested.

[0335] Figure 12 shows an alanine substituted gp41 peptide set used to map DH517 mab and Figure 13 shows a summary of ala mutants to which the antibody is sensitive for binding to gp41. Figures 14 and 15 show the VH and VL sequences of the DH51 1-DH516 antibodies.

[0336] Figures 12-13 show the nucleotide and amino acid sequences of all the certain antibodies of the invention.

[0337] Figures 16-25 show that DH517 displayed a unique mapping pattern in that it depends on DKW at the N terminus and several residues at the C terminus important for 10E8 binding and neutralization. Clone DH51 1 mAbs bound strongly to the majority of the MPER656 variants, showing decreased binding to MPER656.2 and MPER656.2dYIK683R-biotin. These data indicate that the asparagine at position 674 is critical for binding, thus providing evidence that these mAbs bind at the C-terminus. [0338] All the antibodies used in the above Examples had the AAAA substitution in and around the Fc region of the antibody that has been reported to enhance ADCC via NK cells (AAA mutations) containing the Fc region aa of S298A as well as E333A and K334A (Shields RI et al JBC, 276: 6591-6604, 2001) and the 4 th A (N434A) is to enhance FcR neonatal mediated transport of the IgG to mucosal sites (Shields RI et al. ibid).

[0339] Epitope mapping studies are also described in Example 12.

Example 4: Binding assays and Kd determination

[0340] Kd measurements of antibody binding to HIV-1 envelope, e.g. gp41 or any other suitable peptide for the MPER antibodies, will be determined by Surface Plasmon Resonance measurements, for example using Biacore, or any other suitable technology which permits detection of interaction between two molecules in a quantitative way.

Example 5: Various assays

[0341] Various assays for self-reactivity of human antibodies are known in the art. AtheNA Multi-Lyte ANA Plus Test System is one such assay. ELISA cardiolipin assay is another assay to measure autoreactivity.

[0342] The stability and properties of the antibodies, for example as formulated in a composition for treatment will be tested.

[0343] Animal studies (PK and PD studies) could be conducted to determine the distribution and half-life of the antibodies.

[0344] Various assays and experiments can be designed to analyze prevention, treatment and/or cure.

Example 6: Antibodies from CH235 lineage

[0345] CH557 is one example of a CD4bs broad neutralizing HIV- 1 antibody, from a series of clonal antibodies (Figure 28) which can be used in combination with the antibodies of the invention.

Example 7: V3 glycan Antibodies from DH270 lineage

[0346] Antibodies from DH270 lineage are shown in Figure 26. II (DH270IA1), 12, 14, 13 and UCA in Figure 26 are not isolated from human subjects but are derived computationally based on VH and VL sequences of other observed antibodies from the clone: DH471, DH429,

DH473, DH391 and DH270. The VH and VL sequences of DH471, DH429, DH473, DH391 and DH270 are derived from a human subject infected with HIV-1.

[0347] The VH and VL sequences of DH471, DH429, DH473, DH391 and DH270 are derived essentially as described in Example 1, except that cell were sorted with a different hook.

[0348] Neutralization data for antibodies II (DH270IA1) and DH429 is summarized in Figure 9, and Figure 10.

[0349] DH542, DH542-QSA, DH542 K3 are non-limiting examples of V3 antibodies, which can be used in combination with the antibodies of the invention. The nucleotide and amino acid sequences of the VH and VL of DH542 QSA are shown below. DH542 QSA antibody has the VH of DH542 and the VL called DH542-QSA

[0350] >DH542_HC_nt (SEQ ID NO: 465)

CAGGTGCAGCTGGTGCAGTCTGGGGCTCAAATGAAGAACCCTGGGGCCTCAGTGAAGGTC TC CTGCGCGCCTTCTGGATATACCTTCACCGACTTTTACATACATTGGTTGCGCCAGGCCCC TG GCCAGGGGCTTCAGTGGATGGGATGGATGAACCCTCAGACTGGTCGCACAAACACTGCAC GA AACTTTCAGGGGAGGGTCACCATGACCAGGGACACGTCCATCGGCACAGCCTACATGGAG TT GAGAAGCCTGACATCTGACGACACGGCCATATATTACTGTACGACAGGGGGATGGATCAG TC TTTACTATGATAGTAGTTATTACCCCAACTTTGACCACTGGGGTCAGGGAACCCTGCTCA CC GTCTCCTCAG

[0351] >DH542_HC_aa (SEQ ID NO: 466)

QVQLVQSGAQMKNPGASVKVSCAPSGYTFTDFYIHWLRQAPGQGLQWMGWMNPQTGRTNT AR NFQGRVTMTRDTSIGTAYMELRSLTSDDTAIYYCTTGGWISLYYDSSYYPNFDHWGQGTL LT VSS

[0352] >DH542_LC_nt_corrected (DH542 QSA) (SEQ ID NO: 467)

CAGTCTGCCCTGACTCAGCCTGCCTCCGTGTCTGGGTCTCCTGGACAGTCGATCACCATC TC CTGCACTGGAACCAAGTATGATGTTGGGAGTCATGACCTTGTCTCCTGGTACCAACAGTA CC CAGGCAAAGTCCCCAAATACATGATTTATGAAGTCAATAAACGGCCCTCAGGAGTTTCTA AT CGCTTCTCTGGCTCCAAATCTGGCAACACGGCCTCCCTGACAATCTCTGGGCTCCGGGCT GA GGACGAGGCTGACTATTATTGCTGTTCATTTGGAGGGAGTGCCACCGTGGTCTGCGGCGG CG GGACCAAGGTGACCGTCCTAg

[0353] >DH542_LC_aa_corrected (DH542 QSA) (SEQ ID NO: 468) QSALTQPASVSGSPGQSI ISCTGTKYDVGSHDLVSWYQQYPGKVPKYMIYEVNKRPSGVSN

RFSGSKSGNTASLTISGLRAEDEADYYCCSFGGSATVVCGGGTKVTVL

[0354] DH542-L4 is an antibody that has a VH of DH542 and VL of DH429 (Figure 26)

Example 8: DH540 antibody is described elsewhere.

[0355] DH540 antibody is described in detail in US Ser. No. 62/170,558, filed June 3, 2015. Example 9: TZMbl neutralization assay

[0356] TZMbl neutralization assay was conducted to determine neutralization potency and breadth of different HIV-1 viral species by DH512 and mAb 10E8. Figures 37 and 38 show the results of neutralization against a panel of HIV isolates in the TZMbl pseudovirus neutralization assay. Figures 37 and 38 also show the mean IC50, IC80 and percent of isolates neutralized at different IC50 or IC80 values.

Example 10: Isolation of additional antibodies from the DH511 lineage

[0357] High throughput native VH:VL sequencing from single B cells

[0358] Additional antibodies were isolated from the individual CH0210 by high-throughput sequencing of the paired human immunoglobulin heavy and light chain repertoire. See Figure 39. For detailed methods, see DeKosky et al. Nature Biotechnology, 31, 166-169 (2013), and DeKosky et al. Nature Medicine, 21, 86-91 (2015). Briefly B cells were isolated from PBMCs via negative depletion. The heavy and light chain transcripts were co-localized on RNA binding beads, and then physically tied together using overlap extension RT-PCR. The paired VH:VL amplicons were then used to generate 3 libraries for sequencing: a heavy chain database, a light chain database, and a paired database. The necessity for three databases stems from the fact that MiSeq currently limits the forward and reverse reads to -300 bp each (approximate read lengths are shown below as arrows). As the heavy and light chains are both longer than 300 nucleotides, the full length heavy and full length light chains were sequenced separately and the paired database was used as a key to stitch the heavy and light chains together by matching unique CDR3 sequences.

[0359] F(ab)2 fragments were prepared from total serum IgG and subjected to antigen-affinity chromatography using the MPER peptide. Proteins in the elution and flow-through were denatured and reduced, alkylated, trypsin-digested and analyzed by high resolution LC- MS/MS. Spectra were interpreted with the heavy chain database obtained from next- generation sequencing, and peptides uniquely associated with a single CDR ("informative peptides") were used to identify full-length VH sequences. Clonotypes are defined as VH sequences having the same germline V and J and at least 85% aa identity in the CDRH3. To identify the MPER-binding antibodies, the focus was on the clonotypes that contain the identified CDR3 peptides and were highly enriched in the elution. This identified three clonotypes: 137, 335 and 195. All three clonotypes use the same VDJ combination (VH3-15, DH3-3, and JH6), which was also utilized by the DH511 series MPER lineages.

[0360] Based on VH sequences it was apparent that the antibodies pulled out by the paired VH:VL sequencing technology were members of the DH511 clonal lineage. Therefore, all of the antibodies are named starting with DH511. The numbers after the underscore correspond to the cluster names that were designated by the VH:VL sequencing. Antibodies were clustered by 96% nucleotide identity in the CDR3.

[0361] The above analysis identified additional MPER antibodies listed below:

[0362] VH and VL genes were selected and made in linear cassettes (essentially as described in Liao HX et al. J. Virol. Methods 158: 171-9, 2009, see for example Figure 1, Section 3.3) to produce recombinant monoclonal antibodies by transient transfection in 293T cells. See also Example 1 for variations in the backbone.

Example 11: Heavy and Light Chain Chimeric Antibodies; Antibodies with changes in the amino acids of the VH chain

[0363] This example describes chimeric antibodies comprising non-natural VH and VL chain pairs. Naturally occurring VH or VL chain are combined in non-natural pairs as described in Figure 55, chimeras 1-91.

[0364] Chimeras 1-91 were recombinantly expressed and their neutralization profile was determined in the TZMB1 assay (Figure 56). Based on neutralization data for chimeras 68-91 as shown in Figure 56, three antibodies DH512 K2 4A (VH: H510049 4A (DH512) and VL: DH511 1AVK), DH512 K3 4A (VH: H510049 4A (DH512) and VL: DH511 2AVK) and DH512 K4 4A (VH: H510049 4A (DH512) and VL: DH511 5AVK) antibodies were produced large scale and will be tested for neutralization against a larger panel of viruses (see panels for DU512). DH512 K3 4A is also referred to as DH512 K3 and DH511 2 K3.

[0365] The invention contemplates antibodies which comprise amino acid changes, or combination of such changes, in the VH chains of antibodies form the DH511 lineage. Non- limiting examples of antibodies with mutations are provided in Figures 30-33, or any combination thereof. Most mutations are to changes to W, but can also try F, L or possibly other substitutions, e.g. without limitation I, V, A. Additional mutations include without limitation the following: TlOOaF; TlOOaL; TlOOal; TlOOaV; TlOOaA; LlOOdW, or any combination thereof.

[0366] In some embodiment, such double mutants: TlOOaW-LlOOdF; TlOOaW-LlOOdW; TlOOaF-LlOOdF; TlOOaL-LlOOdF; TlOOaL-LlOOdW.

[0367] Neutralization data for a subset of these antibodies is provided in Figure 34. The data show that some of the mutations abrogate neutralization while others enhance potency. One candidate, DH512_L100dF_4A, is more potent than 10E8 and has similar potency to DH512 K3.

[0368] In some embodiments, LlOOd could be changed to Trp.

[0369] Data in Figure 34 and 80 show that single mutant LlOOdF, and single mutant TlOOaW have improved neutralization. These single mutants will be tested against a panel of additional viruses (see panel for DH512, DH512 K3).

[0370] Contemplated are also combination mutations, for example but not limited combination TlOOaW with LlOOdF, combination LlOOdW with TlOOaW.

[0371] Mutated VH chain as contemplated above could be combined with VH chain from DH512, or with VL chain from DH512 K3 (DH511 2AVK).

Example 12: Shared Memory and Plasma Repertoires of HIV- 1 Neutralizing Antibodies

[0372] Shared Memory and Plasma Repertoires of HIV-1 Neutralizing Antibodies

[0373] Understanding the relationship of the memory B cell and plasma immunoglobulin repertoires of HIV-1 -infected individuals who develop broadly neutralizing antibodies (bnAbs) is important, since plasma antibody responses are required to achieve maximum protection from infectious agents. Using HIV-1 envelope gp41 membrane-proximal external region (MPER)-specific memory B cell sorting and next-generation sequencing, coupled with mass spectrometry analysis of plasma antibodies, we probed the memory B cell and plasma antibody repertoires of an HIV-1 -infected donor with a plasma bnAb signature that mapped to Env gp41 distal MPER. We found potent IgG bnAbs from the same B cell clonal lineage in memory B cells and plasma that neutralized 99% of HIV-1 isolates. Structural analysis demonstrated clonal lineage antibodies from memory B cells and plasma both recognized the envelope gp41 epitope identically in an alpha helical conformation. Thus, a major source of potentially protective plasma HIV-1 bnAbs is the memory B cell pool. [0374] Introduction

[0375] Inducing broadly reactive neutralizing antibodies (bnAbs) is critical for developing a protective HIV-1 vaccine. Some of the broadest bnAbs isolated are to the envelope gp41 membrane proximal external region (MPER), with two of these, 10E8 and 4E10, the most broad (1, 2). Monoclonal antibody (mAb) 4E10, while extremely broad in neutralization breadth, is not potent, and is highly polyreactive with many non-HIV-1 proteins and autoreactive with the human protein splicing factor 3b subunit 3 (SF3B3) (3) as well as with lipids (4). In contrast, mAb 10E8 is not as polyreactive as 4E10, and is both more broad and potent (1), although it does have a degree of lipid reactivity (5) and is autoreactive with the host protein family of sequence similarity 84 member A (FAM84A) (6).

[0376] To date, all HIV-1 broadly neutralizing antibodies have been isolated from memory B cells, either with clonal memory B cell cultures or using fluorophore-labeled Env and flow cytometry cell sorting. However, most correlates of protection for infectious agents with successful vaccines are the levels of plasma neutralizing antibodies. Moreover, the correlate of decreased transmission risk in the only HIV-1 vaccine trial to demonstrate a degree of efficacy was plasma antibodies to the second variable loop (V2) region (7).

[0377] In HIV-1 infection, 60% of HIV-1 -specific antibodies derive from abnormal B cell subsets, that are either activated or exhausted and express Fc receptor-like-4 (FcRL4) (8, 9). However, many of the antibodies reflected in HIV-1 memory B cells are not expressed in plasma (8). Similarly, many of the memory B cell specificities of antibodies in other settings are also not represented in plasma (10-12). Thus, it is not known if envelope-reactive memory B cells with bnAb B cell receptors are a major source of plasma broad neutralizing activity.

[0378] Here we have isolated memory B cell and plasma broad and potent envelope gp41 bnAbs from an African donor and demonstrated broad and potent plasma gp41 bnAbs to be in the same B cell clonal lineage as those isolated from memory B cells. Chimeric antibodies consisting of memory bnAb VH and plasma bnAb VL as well as engineering memory bnAb heavy chain complementary determining regions yield antibodies with greater potency than naturally paired antibodies. Thus, the class-switched memory B cell pool contributes to plasma bnAbs.

[0379] Results

[0380] Isolation of Memory B Cell gp41 Neutralizing Antibodies [0381] Neutralization-based epitope prediction analysis revealed that plasma from HIV-1 clade C-infected individual CH0210 contained C-terminal MPER bnAb activity (13) (Figure 62). Six clonally-related mAbs, designated DH511.1-DH511.6, were isolated by antigen- specific single memory B cell sorting using MPER peptide fluorophore-labeled probes (14) (Fig. 59a, 59b, and Supplementary Table 1). The DH511 B cell clonal lineage was distinguished by HCDR3 loops of 24 amino acids in DH511.1, DH511.3, and DH511.6, while DH511.2, DH511.4, and DH511.5 antibodies had a one amino acid deletion in the HCDR3, resulting in a length of 23 amino acids (Supplementary Table 1). VH and VL somatic mutation rates were 15-22% and 14-18%, respectively. The DH511 clonal lineage was derived from the same heavy-chain germ line gene as previously isolated gp41 neutralizing antibody 10E8 (VH3-15), but utilized a different VL germ line gene (DH511 : VK1-39, 10E8: VL3-19) (1) (Supplementary Table 1). Antibody DH517, derived from a second clonal lineage arising from the same donor, was similarly isolated. DH517 utilized VH 4-34 and VL3-19 germ line genes, was 22.8% and 14.3% mutated, respectively, and had a long HCDR3 comprised of 24 amino acids.

[0382] DH511.1-DH511.6 and DH517 mAbs were assessed for neutralization breadth and potency against a panel of 30 cross-clade HIV-1 isolates. All six DH511 clonal members neutralized 30 of 30 isolates tested with median 50% inhibitory concentrations (IC50) ranging from 0.7 to 4.2 μg/ml (Supplementary Table 2a). DH517 had less breadth than DH511 clone antibodies, neutralizing 15 of 30 isolates with a median IC50 of 5.7 μg/ml

(Supplementary Table 2a). The most potent DH511 clone bnAb (DH511.2) in a large cross- clade panel of 199 geographically and genetically diverse HIV-1 Env pseudoviruses, neutralized 197/199 (99%) viruses but was less potent than 10E8 (195/200, 98%) (median IC50, DH511.2 = 1.1 μg/ml and 10E8 = 0.4 μg/ml) (Fig. 59c, 59d, and Supplementary Table 3). Neutralization curves revealed that DH511.2 achieved >99% maximal neutralization for 93% of the isolates (Fig. 59e), and showed similar potency and breadth of neutralization against a second panel of 200 clade C primary HIV-1 isolates (Supplementary Table 4).

[0383] Isolation of Plasma gp41 Neutralizing Antibodies

[0384] We next analyzed the MPER-specific plasma antibody repertoire from donor CH0210 using an independent proteomics-based approach for the identification and semi-quantitative determination of antigen-specific antibodies in human serum (15, 16). MPER-specific antibodies were isolated from a 2 ml plasma sample by affinity chromatography, processed for proteomics (10) and subjected to liquid chromatography high-resolution tandem mass spectrometry (LC-MS/MS) analysis. For peptide identification, a donor-specific VH database comprising 98,413 unique high quality sequences was derived from a natively paired VH:VL repertoire from 845,000 peripheral single B cells from total PBMCs (isolated using MACS negative selection: CD2-CD14-CD16-CD43-CD235a-) (17-19). These VH sequences were then clustered into 4,428 clonotypes, using a cut-off of≥85% amino acid identity in the HCDR3 region.

[0385] Using stringent data filtering protocols (10), high confidence peptide-spectrum matches (PSMs) from HCDR3 peptides were identified and their respective LC peak intensities were used for relative quantification. As we have shown previously, an estimated >80% of all HCDR3 peptides within a sample are typically identified in this manner

(detection limit approximately 0.4 ng/ml), and peak intensities correlate well with absolute peptide concentrations (10, 15). Plasma Ig clonotypes were defined as VH sequences having the same germline V and J and 85% aa identity in the HCDR3.

[0386] We found that the MPER-specific plasma antibody repertoire consisted of 10 clonotypes, three of which used the same VDJ combination (VH3-15, DH3-3, JH6) as the DH511 clonal lineage (Figure 63). Clonotype IV comprised 95% of the total intensity of HCDR3 peptides detected in the MPER-specific antibody repertoire (i.e. in antibodies eluted following affinity chromatography with immobilized MPR.03 peptide); we noted that detection of HCDR1 and HCDR2 peptides unique to Clonotype IV provided further unambiguous support for the prevalence of these antibodies in the CH0210 plasma (Fig. 59f). Clonotype II, which included antibodies DH511.2, DH511.4 and DH511.5 isolated by single- cell sorting, and Clonotype III were detected at 4% and 1% relative abundancy, respectively (Fig. 59f). All three HCDR3 clonotypes utilized the same VDJ genes (VH3-15, DH 3-3 and JH6), displayed similar HCDR3 lengths of 23-24 amino acids and VHgene mutation rates of 15-20% (Supplementary Table 6). Whereas 11 VH DH511 clonal lineage members were found by mass spectrometry (Supplementary Table 6, Figure 64), the phylogram was collapsed to represent the most prevalent members (Fig. 59f). It is noteworthy that Clonotype I (Fig. 59g), that includes DH511.1, DH511.3 and DH511.6, was isolated by memory B cell sorting but was not detected in the plasma; we validated that recombinant DH511.1, DH511.3 and DH511.6 antibodies were readily detectable by mass spectrometry, indicating that their absence from the CH0210 plasma was not a technical artifact. [0387] Using the proteomically identified HCDR3 sequences, we searched the native VH:VL sequence database comprising -200,000 heavy-light chain pairs from single B cells to determine the respective full-length light-chain sequence belonging to each clonotype (Supplementary Table 6). For clonotypes in which multiple VH:VL somatic variants were detected, only the two most frequent variants, as quantified by the number of sequencing reads, were selected for expression and characterization (Supplementary Table 6). The light-chains belonging to these three clonotypes all shared the same V- and J- gene identity (IGKV1-39, IGKJ2) as the light-chains of the DH511 clonal lineage isolated by memory B cell single-cell sorting. Six plasma mAbs belonging to the DH511 clonal lineage (designated DH511 7P-DH511.12P), showed potent tier 2 neutralizing activity against a panel of four HIV-1 isolates (Supplementary Table 7), with mAbs DH511.1 IP and DH511.12P demonstrating the most potent neutralizing activity. DH511.1 IP and DH511.12P were selected for further characterization of their neutralization breadth and potency against a panel of 203 cross-clade isolates and had slightly more breadth (99.5% of isolates tested) and greater potency than memory B cell-derived DH511.2 but were less potent than 10E8 (median IC 50 : 0.7 μg/ml for DH511.1 IP and DH511.12P versus 0.4 μg ml for 10E8) (Supplementary Table 8).

[0388] Structural Analysis of DH511 Lineage Antibodies

[0389] We used a panel of alanine substituted MPER peptides that span gp41 residues 671- 683 (Supplementary Table 9) to define the epitopes of DH511.1-DH511.12P by enzyme linked immunosorbent assay (ELISA). Similar to the epitopes of 4E10 and 10E8 (1), DH511.1-DH511.12P binding was sensitive to alanine mutations at Asn671 gp41 and

Trp672 gp41 , but unlike 4E10 and 10E8, was also sensitive to Asp674Ala gp41 , and to a lesser extent Leu679Ala gp41 mutations (Figure 63). Assessment of the neutralization activity of DH511.1 -DH511.12P (not DH511.7-DH511.10) mAbs against clade C COT6.15 Env pseudoviruses bearing alanine substitutions across the MPER (20, 21) (Supplementary Table 10) demonstrated sensitivity of neutralization to Env mutations of Phe673Ala gp41 , Asp674Ala gp41 , and Asp674Ser gp41 , with the most prominent resistance observed against the Trp672Ala gp41 mutant virus (Supplementary Table 11). These data demonstrated that the epitope recognized by DH511 lineage antibodies was similar to but distinct with those of gp41 bnAbs 4E10 and 10E8, requiring the aspartic acid at position 674 for binding and neutralization. [0390] Crystal structures of the antigen-binding fragments (Fab) of the DH511.1 antibody in complex with a peptide spanning the full gp41 MPER (residues 656-683) and of the DH511.2 antibody in complex with gp41 peptides spanning residues 662-683 and 670-683 were determined to 2.7 A, 2.6 A and 2.2 A resolution, respectively (Fig. 60, Figure 62 and Supplementary Tables 12 and 13). Both DH511.1 and DH511.2 recognized an alpha- helical conformation of the distal portion of the gp41 MPER (residues 671-683) (Fig. 60a), similar to the conformation recognized by neutralizing antibodies 10E8 and 4E10 (Fig. 61b). RMSDs for this region of gp41 across all four antibody-bound structures did not exceed 0.46 A. Ordered electron density for the bound peptides was also observed upstream of the distal gp41 MPER helix. In the case of DH511.1 , an additional□ -helix was present between residues 656-661, followed by an extended conformation between residues 662-670 (Fig. 60a). DH511.2-bound MPER also adopted an extended conformation between residues 662- 670, upstream of the distal helix, with the highest degree of overall structural homology to DH511.1 -bound MPER occurring between gp41 residues 668-683 (CD RMSD = 0.39 A) (Fig. 60a). Interactions between DH511.1 and DH511.2 and gp41 MPER were mediated exclusively by their heavy chains, with VH3-15-encoded regions accounting for 45-50% of the antibody contact interface with gp41, and HCDR3 loops accounting for 50% or more of the remaining interface (Fig. 60b and 61c, Supplementary Table 14). A total of 751.1 and 681.4 A 2 interactive surface area was buried on DH511.1 and DH511.2, respectively, and 797.2 and 780.1 A 2 on gp41 MPER in the two respective structures (Supplementary Table 14). The larger interface observed for the DH511.1 complex was due to the longer gp41 MPER peptide of that complex and the additional interface observed between its N-terminus and the antibody. It is likely that this additional interface is due to crystal lattice constraints, since alanine scan mutagenesis of N-terminal gp41 MPER residues did not result in reduction of antibody binding (Figure 65) . Contacts between DH511.1 and DH511.2 and gp41 MPER were highly conserved in both structures (Fig. 60c and Supplementary Tables 15-16). VH3- 15-encoded residues of both DH511.1 and DH511.2 mediated interactions with gp41 residues L669, W670, N671, W672, F673, and D674, while their HCDR3 loop residues contacted gp41 residues W672, T676, L678, W679 and R683, as well as 1675 in the case of DH511.2 (Fig. 60c and Supplementary Tables 15-16). The interactions observed in the structures were consistent with alanine scan analyses that revealed reduced antibody binding upon mutation of gp41 residues 671-674 and 679 (Figure 65). Interactions between DH511.1 and DH51 1.2 and main-chain atoms of gp41, which would be difficult to detect in alanine scan analyses, were also observed, including between antibody residue N3 1 and the carbonyl oxygen of gp41 W670 (Fig. 60c and Supplementary Tables 15-16).

[0391] To compare atomic-level recognition of gp41 MPER by plasma-derived versus memory B-cell-derived antibodies, structural studies of the plasma-derived DH511 -lineage antibodies DH51 1.1 IP and DH51 1.12P were undertaken in complex with gp41 MPER peptides. Crystal structures of DH511.1 IP and DH51 1.12P Fabs were determined in complex with a peptide spanning gp41 MPER residues 662-683, to 2.47 and 1.88 A, respectively (Fig. 60c and Supplementary Tables 12 and 13). The structures revealed that both plasma derived variants recognized a conformation of the MPER similar to that recognized by DH51 1.1 and DH51 1.2, adopting an□ -helix between residues 671-683 and an extended conformation upstream, between residues 662-670. The highest degree of structural homology occurred between residues 668-683. As in the case of DH51 1.1 and DH51 1.2, interactions between DH51 1.1 IP and DH511.12P and gp41 were mediated exclusively by their heavy chains (Fig. 60d and Supplementary Table 14). The plasma-derived variants recognized the very same gp41 residues as those recognized in common by DH51 1.1 and DH51 1.2, although the respective antibody residues that mediated these contacts with gp41 differed in some cases (Fig. 60b, 60d, 60e and Supplementary Tables 15-18). While contacts between HCDR1 loop residues of the DH51 1.1 IP and DH51 1.12P and gp41 were largely conserved relative to those of DH51 1.1 and DH51 1.2, gp41 contacts mediated by their HCDR2 loops diverged relative to those of DH51 1.1 and DH51 1.2 (Fig. 60). The substitution of DH51 1.1 and DH511.2 HCDR2 residue K52c with a glycine in DH51 1.1 IP and

DH51 1.12P, led to the loss of a salt bridge mediated by K52c and gp41 residue D674 - one that was replaced by an additional salt bridge mediated by conserved residue R52a (Figures 60b, 60d, 60e and Supplementary Tables 15-18). Examination of additional gp41- contacting residues that were unique to the plasma-derived variants revealed that unique residues of their HCDR3 loops, which differed from the DH51 1.1 and DH51 1.2 HCDR3 loops at ~7 residue positions, mediated many of these contacts (Fig. 60c and Supplementary Tables 17-18). Despite their overall sequence divergence from DH511.1 and DH51 1.2, -26- 28% in heavy chain variable regions, the structures of the DH511.1 IP and DH51 1.12P were highly homologous to those of DH51 1.1 and DH51 1.2. In sum, the plasma-derived variants examined here recognized a similar conformation of the gp41 MPER as that recognized by memory B-cell derived variants, contacted a similar set of gp41 residues, and did so through modified antibody contacts that did not significantly alter the backbone conformations of their paratopes or common epitope.

[0392] We next compared the structures of DH51 1 lineage antibodies to those of other antibodies that target the distal gp41 MPER (Fig. 61a and 61b). Since the DH51 1 lineage shares a common VH3-15 heavy chain precursor as the 10E8 lineage, we were especially interested in determining if a structural basis for usage of this precursor to target the MPER could be discerned. As a first step, we compared the directions of approach of DH511 lineage antibodies to the distal MPER helix, relative to those of 10E8 and 4E10. All four antibodies were oriented by superimposing residues 671-683 of their respective epitopes, and their directions of approach were defined by a line drawn from the Ca atom of epitope residue 672 to a point midway between the variable region intra-chain heavy and light chain disulfide bonds, which represented the longitudinal axis of the antibody variable regions. Pairwise comparison of the directions of approach of DH511.1 versus those of DH51 1.2, 10E8 and 4E10 yielded differences of 4.7°, 13.4° and 25.2°, respectively, suggesting the DH51 1 lineage most closely resembled 10E8 in its approach to the epitope (Fig. 61d). While the longitudinal axes of the DH511.1 and DH511.2 variable regions and that of 10E8 were highly similar, the orientations of their heavy and light chains relative to this longitudinal axis differed more substantially - by -54° (Fig. 61d). This difference resulted in a rotational shift of the gp41 footprint on 10E8 relative to the footprint on DH51 1 lineage antibodies (Fig. 61c). Thus, while DH511 lineage antibodies share an identical heavy chain VH3- 15 precursor as antibody 10E8, and approached gp41 MPER from similar angles, the orientations of their heavy and light chains relative to the epitope differed more substantially.

[0393] To determine if a common structural basis for VH3-15 precursor usage could nonetheless be discerned between the two lineages, we compared VH3-15-encoded gp41- contacting residues in DH51 1.1, DH51 1.2 and 10E8. Of the total number of residue interactions that exist between the VH3-15 regions of three respective antibodies and gp41 (8 for DH51 1.1, 10 for DH51 1.2, and 10 for 10E8), five common residue positions were involved interactions with gp41 in all three antibodies: 28, 31, and 33 within the HCDR1 and 52c and 53 within the HCDR2 (Fig. 61c and 60e). Heavy chain residues 31 and 33 are asparagine and tryptophan in all three antibodies and are un-mutated from the germ-line precursor. Residue 53 is aspartate in DH5 11.1 and DH51 1.2, as it is in the germ-line precursor, and a chemically similar glutamate in 10E8. Residue positions 28 and 52c are somatically mutated from germ-line in all three antibodies, to disparate amino acids (Fig. 61e). While all five residues maintain contact with gp41 in both the DH511.1 and 10E8 lineages, the rotational shift in the orientations of the heavy and light chains between the two lineages results in distinct modes of gp41 recognition (Fig. 60b and 61e). Yet, the five common VH3-15 encoded gp41 -contacting residues in both lineages end up interacting with many of the same gp41 MPER residues, including L669, W670, N671, W672, and F673 (Fig. 60b, 60e, and 61e). VH3-15 germ line encoded residue W33, shown in previous studies to be required for 10E8 recognition of gp41 (1), interacts with gp41 residues W672 and F673 in both the DH511.1 and 10E8 lineages, although from a distinct spatial position in each case (Fig. 60b and 61e). Thus, despite a relative shift in heavy and light chain orientations, a common subset of DH511.1 and 10E8 lineage VH3-15 residues interact with the same subset of distal MPER residues. It remains to be determined if the observed differences in the heavy and light chain orientations of two lineages, relative to gp41 MPER, were determined at inception of naive antibody recognition or if they were added during antibody development and maturation.

[0394] Origin and Development of the DH511 Clonal Lineage

[0395] A maximum likelihood phylogenetic tree was constructed from the VDJ sequences recovered from memory B cell sorting and was used to infer the unmutated common ancestor (UCA) of clone DH511 and six maturational intermediate antibodies (Fig. 59b). A global panel of 12 HIV-1 isolates was used to assess the development of neutralization breadth in the DH511 clonal lineage. None of the isolates were neutralized by the UCA or intermediate (I) 6 antibody that was most closely related to the DH511 UCA. Antibody 12 and later members of the lineage acquired the ability to neutralize 12/12 isolates (Supplementary Table 19). DH511 clone acquisition of breadth was associated with the accumulation of somatic mutations, but neutralization potency did not directly correlate with percent VH mutation frequency. Analysis of a panel of MPER peptides and MPER peptide liposomes did not reveal constructs that bound to the UCA. Binding to the MPER peptides was acquired at the 15 stage of maturation (Figures 66 and 67).

[0396] Polyreactivity/autoreactivity of the DH511 Clonal Lineage

[0397] The DH511 inferred UCA and intermediates 11-13 and 16 reacted with several autoantigens as measured by ELISA (Figures 68-69) and were found to exhibit polyreactivity in a protein microarray against 9,400 human proteins (3) (Figure 68). The mature members of the lineage were not polyreactive by ELISA, although some members demonstrated polyreactivity by microarray analysis (DH51 1.1, DH51 1.5, DH51 1.6, and DH51 1.12P). All DH51 1 lineage members lacked reactivity by indirect immunofluorescence human epithelial (HEp-2) cell staining assay. Regarding higher affinity autoreactivity with single proteins, mature bnAb DH51 1.2 reacted with the E3 ubiquitin ligase STIP1 Homology and U-Box Containing Protein 1 (STUB 1) while both DH511.1 IP and DH51 1.12P reacted with nuclear distribution gene C homolog (A. nidulans) (NUDC); DH511.12 also reacted with Scm-like with four MBT domains protein 1 (SFMBT1) (Figure 68).

[0398] To characterize the lipid reactivity of the DH51 1 clonal lineage, we first determined propensity for lipid membrane binding/insertion of DH51 1.1 -DH5 11.6 based on HCDR3 hydrophobicity. Three or more Phe or Trp amino acid residues were contained within the HCDR3 sequences of each DH511 clonal lineage member, and several members were found to have at least one Pro, with the exception of DH51 1.3 and DH51 1.6. A membrane insertion score was calculated based on the Wimley-White hydrophobicity scale, which measures the propensity of amino acids to sit at the interface of the head and tail group in a lipid bilayer. Notably, membrane insertion scores were similar between the most potent neutralizer DH51 1.2 and 4E10/10E8 but differed from 2F5 (Supplementary Table 21).

[0399] To further delineate the interaction of DH51 1 clonal members with the lipid bilayer interface, we determined cardiolipin reactivity and kinetics of binding to MPER peptide versus MPER peptide-liposome conjugates. The UCA and members of the memory B cell clonal lineage did not bind cardiolipin in ELISA (Supplementary Table 22). The binding of gp41 bnAbs 2F5 and 4E10 to gp41 -lipid complex has been proposed as a sequential two-step process, in which encountering the lipid membrane takes place first, presumably to aid in docking of the antibody with the transiently exposed gp41 intermediate neutralizing epitope during the virion-host cell fusion process (4, 22, 23). Surface plasmon resonance (SPR) analysis of DH51 1 lineage fragments of antigen binding (Fabs) demonstrated that DH51 1.1- DH51 1.6 and intermediates 11-15 bound the MPER peptide

(NEQELLELDKWASLWNWFDITNWLWYIR (SEQ ID NO:2)) with nanomolar affinity (Kd range: 1 1.1-99.9 nm), while the inferred UCA and intermediate 6 (most closely related to the UCA) did not bind (Figure 66). Binding kinetics studies as show in Figure 71), support the hypothesis that like 2F5 and 4E10, DH511 lineage antibodies bind in a two-step conformational change model.

[0400] To determine the impact of timing of the gp41 intermediate epitope exposure on HIV- 1 neutralization (24), we compared the window of time in which bnAbs DH51 1.2, 10E8, and 4E10 could neutralize the tier 2 HIV-1 strain B.BG1 168 after virus addition to TZM-bl cells. The lifetime of neutralization for DH51 1.2 (t1/2: 26.8 ± 2.3 min) was the same as that for bnAbs 10E8 (ti/2: 25.6 ± 2.5 min) and 4E10 (t1/2: 28.2 ± 3.5 min), similar to the published half- life of fusion inhibition by the gp41 intermediate mimic T20 (20.2 ± 0.5 min) (24). These results suggest that DH51 1.2 recognizes a transiently exposed intermediate state of gp41 (25).

[0401] Engineering DH511 Clonal Lineage Members for Enhanced Potency

[0402] To identify more potent variants of the DH511 clonal lineage, we generated 91 chimeric mAbs by swapping the heavy and light-chains of DH51 1.2 with those of DH51 1 lineage members derived from the plasma. Of the 91 chimeric antibodies, one variant, DH51 1 2 K3 (comprised of the DH5 11.2 heavy-chain reconstituted with the plasma light- chain of DH51 1.8P), showed greater potency than 10E8 (Supplementary Table 24) .

DH51 1.2 K3 neutralization data are shown in Figure 28 and 58.

[0403] Sixteen HCDR3 mutations of DH51 1.2 were made (Figures 30-33) to determine effect on DH51 1.2 potency. Figure 34 shows neutralization data for sixteen of these antibodies. Additional mutations will be made, including combinations of mutations, from the mutations listed in Figures 30-31.

[0404] Discussion

[0405] We have used a combination of memory B cell sorting (26, 27) and plasma antigen- specific antibody characterization by HCDR3 mass spectrometry sequencing to

simultaneously characterize class-switched memory B cell antibodies and plasma antibodies (15, 28-30). The memory B cell repertoire contains multiple specificities of antibodies reflective of an individual's immune history (30) whereas primary contributors to plasma antibodies are both long lived plasma cells as well as shorter lived plasma cells derived from terminally differentiated memory B cells in response to current antigens ( 16). However, evidence exists that for non-HIV-1 antigens such as influenza ( 1 1) and West Nile virus (12), not all of the memory B cell repertoire is found in plasma. Here we demonstrate that class- switched memory B cells and plasma shared the same clonal lineage members of highly broad and potent HIV-1 gp41 neutralizing antibodies. [0406] In the case of HIV- 1 antibody responses, the relationship of the memory B cell and plasma antibody pools is complicated by the damage that HIV-1 inflicts on the B cell lineage with disruption of the germinal center in the earliest stages of infection (31), and the accumulation of FcRL4+ memory B cells in chronic infection (8). Interestingly, HIV-1- specific B cell responses are enriched in the FcRL4+ memory B cell compartment and exhibit many features of premature exhaustion (8). Regarding antibodies that target the Env bnAb epitope at the CD4 binding site, it has been shown that -60% of this response is contained within the exhausted FcRL4+ memory B cell compartment, thus preventing their progression to plasma cells and production of secreted antibody (8, 9). In contrast, Scheid and colleagues studied antigen-specific memory B cell repertoires in HIV-1 infected individuals and found broad diversity of neutralizing antibodies (32). Moreover, analysis has demonstrated bnAb activity in plasma can predict isolation of bnAb variable heavy (VH) and variable light (VL) from memory B cells from the same individual (13, 33-38). Moreover, only a limited number of bnAb specificities are generally present in HIV-1 -infected plasma (38, 39), and when bnAbs are isolated from memory B cells in clonal memory B cell cultures, the bnAbs are the minority of the Env specifities isolated (26, 37, 40). Thus, in spite of early damage to B cell follicles and accumulation of memory B cells with an exhaustion phenotype, HIV-1 infected individuals can make productive, albeit subdominant, bnAb responses that progress to plasma cell differentiation and secretion into blood plasma.

[0407] A critical question is whether memory B cells in HIV-1 infected individuals are differentiating into the long-lived plasma cell pool that resides in bone marrow and is responsible for long-lived plasma antibody responses (41). We have previously studied the effect of anti-retroviral treatment in HIV-1 infection on the half-lives of Env gpl20 and gp41 as well as Gag antibody responses, and demonstrated whereas Env antibody half-life was short for gp 120 (81 weeks) and gp41 (33 weeks), antibody half-life was longer for Gag (648 weeks). In contrast, in the same individuals, the half-life of influenza antibodies did not decay over the time studied (42). These data demonstrate that in chronic HIV-1 -infection, the cells making plasma gp41 antibodies are not long-lived plasma cells.

[0408] Thus, by directly measuring the gp41 broad neutralizing repertoire in memory B cells and plasma, we have directly demonstrated the survival from immune damage of memory B cells to produce plasma broadly neutralizing antibodies. Finally, we show that blood plasma is a rich source for isolation of potent bnAbs for recombinant antibody production and for constructing chimeric memory B cell/plasma antibodies for enhancing antibody potency and breadth.

[0409] References

[0410] 1. Huang J, Ofek G, Laub L, Louder MK, Doria-Rose NA, Longo NS, Imamichi H, Bailer RT, Chakrabarti B, Sharma SK, Alam SM, Wang T, Yang Y, Zhang B, Migueles SA, Wyatt R, Haynes BF, Kwong PD, Mascola JR, Connors M. 2012. Broad and potent neutralization of HIV-1 by a gp41-specific human antibody. Nature 491 :406- 412.

[0411] 2. Zwick MB, Labrijn AF, Wang M, Spenlehauer C, Saphire EO, Binley JM, Moore JP, Stiegler G, Katinger H, Burton DR, Parren PW. 2001. Broadly neutralizing antibodies targeted to the membrane -proximal external region of human

immunodeficiency virus type 1 glycoprotein gp41. Journal of virology 75: 10892-10905.

[0412] 3. Yang G, Holl TM, Liu Y, Li Y, Lu X, Nicely NI, Kepler TB, Alam SM, Liao HX, Cain DW, Spicer L, VandeBerg JL, Haynes BF, Kelsoe G. 2013. Identification of autoantigens recognized by the 2F5 and 4E10 broadly neutralizing HIV-1 antibodies. The Journal of experimental medicine 210:241-256.

[0413] 4. Alam SM, McAdams M, Boren D, Rak M, Scearce RM, Gao F, Camacho ZT, Gewirth D, Kelsoe G, Chen P, Haynes BF. 2007. The role of antibody polyspecificity and lipid reactivity in binding of broadly neutralizing anti-HIV-1 envelope human monoclonal antibodies 2F5 and 4E10 to glycoprotein 41 membrane proximal envelope epitopes. Journal of immunology 178:4424-4435.

[0414] 5. Chen J, Frey G, Peng H, Rits-Volloch S, Garrity J, Seaman MS, Chen B. 2014.

Mechanism of HIV-1 neutralization by antibodies targeting a membrane-proximal region of gp41. Journal of virology 88: 1249-1258.

[0415] 6. Liu M, Yang G, Wiehe K, Nicely NI, Vandergrift NA, Rountree W,

Bonsignori M, Alam SM, Gao J, Haynes BF, Kelsoe G. 2015. Polyreactivity and autoreactivity among HIV-1 antibodies. Journal of virology 89:784-798.

[0416] 7. Haynes BF, Gilbert PB, McElrath MJ, Zolla-Pazner S, Tomaras GD, Alam SM, Evans DT, Montefiori DC, Karnasuta C, Sutthent R, Liao HX, DeVico AL, Lewis GK, Williams C, Pinter A, Fong Y, Janes H, DeCamp A, Huang Y, Rao M, Billings E, Karasavvas N, Robb ML, Ngauy V, de Souza MS, Paris R, Ferrari G, Bailer RT, Soderberg KA, Andrews C, Berman PW, Frahm N, De Rosa SC, Alpert MD, Yates NL, Shen X, Koup RA, Pitisuttithum P, Kaewkungwal J, Nitayaphan S, Rerks-Ngarm S, Michael NL, Kim JH. 2012. Immune-correlates analysis of an HIV-1 vaccine efficacy trial. The New England journal of medicine 366: 1275-1286.

[0417] 8. Moir S, Ho J, Malaspina A, Wang W, DiPoto AC, O'Shea MA, Roby G, Kottilil S, Arthos J, Proschan MA, Chun TW, Fauci AS. 2008. Evidence for HIV- associated B cell exhaustion in a dysfunctional memory B cell compartment in HIV- infected viremic individuals. The Journal of experimental medicine 205: 1797-1805.

[0418] 9. Kardava L, Moir S, Shah N, Wang W, Wilson R, Buckner CM, Santich BH, Kim LJ, Spurlin EE, Nelson AK, Wheatley AK, Harvey CJ, McDermott AB,

Wucherpfennig KW, Chun TW, Tsang JS, Li Y, Fauci AS. 2014. Abnormal B cell memory subsets dominate HIV-specific responses in infected individuals. The Journal of clinical investigation 124:3252-3262.

[0419] 10. Boutz DR, Horton AP, Wine Y, Lavinder JJ, Georgiou G, Marcotte EM. 2014.

Proteomic identification of monoclonal antibodies from serum. Analytical chemistry 86:4758-4766.

[0420] 11. Wrammert J, Koutsonanos D, Li GM, Edupuganti S, Sui J, Morrissey M, McCausland M, Skountzou I, Hornig M, Lipkin WI, Mehta A, Razavi B, Del Rio C, Zheng NY, Lee JH, Huang M, Ali Z, Kaur K, Andrews S, Amara RR, Wang Y, Das SR, O'Donnell CD, Yewdell JW, Subbarao K, Marasco WA, Mulligan MJ, Compans R, Ahmed R, Wilson PC. 2011. Broadly cross-reactive antibodies dominate the human B cell response against 2009 pandemic H1N1 influenza virus infection. The Journal of experimental medicine 208: 181-193.

[0421] 12. Purtha WE, Tedder TF, Johnson S, Bhattacharya D, Diamond MS. 2011.

Memory B cells, but not long-lived plasma cells, possess antigen specificities for viral escape mutants. The Journal of experimental medicine 208:2599-2606.

[0422] 13. Georgiev IS, Doria-Rose NA, Zhou T, Kwon YD, Staupe RP, Moquin S, Chuang GY, Louder MK, Schmidt SD, Altae-Tran HR, Bailer RT, McKee K, Nason M, O'Dell S, Ofek G, Pancera M, Srivatsan S, Shapiro L, Connors M, Migueles SA, Morris L, Nishimura Y, Martin MA, Mascola JR, Kwong PD. 2013. Delineating antibody recognition in polyclonal sera from patterns of HIV-1 isolate neutralization. Science 340:751-756. [0423] 14. Morris L, Chen X, Alam M, Tomaras G, Zhang R, Marshall DJ, Chen B, Parks R, Foulger A, Jaeger F, Donathan M, Bilska M, Gray ES, Abdool Karim SS, Kepler TB, Whitesides J, Montefiori D, Moody MA, Liao HX, Haynes BF. 2011. Isolation of a human anti-HIV gp41 membrane proximal region neutralizing antibody by antigen- specific single B cell sorting. PloS one 6:e23532.

[0424] 15. Lavinder JJ, Wine Y, Giesecke C, Ippolito GC, Horton AP, Lungu OI, Hoi KH, DeKosky BJ, Murrin EM, Wirth MM, Ellington AD, Dorner T, Marcotte EM, Boutz DR, Georgiou G. 2014. Identification and characterization of the constituent human serum antibodies elicited by vaccination. Proceedings of the National Academy of Sciences of the United States of America 111 :2259-2264.

[0425] 16. Wine Y, Horton AP, Ippolito GC, Georgiou G. 2015. Serology in the 21st century: the molecular-level analysis of the serum antibody repertoire. Current opinion in immunology 35:89-97.

[0426] 17. McDaniel JR, DeKosky BJ, Tanno H, Ellington AD, Georgiou G. 2016. Ultra- high-throughput sequencing of the immune receptor repertoire from millions of lymphocytes. Nature protocols 11 :429-442.

[0427] 18. DeKosky B J, Ippolito GC, Deschner RP, Lavinder JJ, Wine Y, Rawlings BM, Varadarajan N, Giesecke C, Dorner T, Andrews SF, Wilson PC, Hunicke-Smith SP, Willson CG, Ellington AD, Georgiou G. 2013. High-throughput sequencing of the paired human immunoglobulin heavy and light chain repertoire. Nature biotechnology 31 : 166- 169.

[0428] 19. DeKosky BJ, Kojima T, Rodin A, Charab W, Ippolito GC, Ellington AD, Georgiou G. 2015. In-depth determination and analysis of the human paired heavy- and light-chain antibody repertoire. Nature medicine 21 :86-91.

[0429] 20. Gray ES, Madiga MC, Moore PL, Mlisana K, Abdool Karim SS, Binley JM, Shaw GM, Mascola JR, Morris L. 2009. Broad neutralization of human

immunodeficiency virus type 1 mediated by plasma antibodies against the gp41 membrane proximal external region. Journal of virology 83: 11265-11274.

[0430] 21. Gray ES, Meyers T, Gray G, Montefiori DC, Morris L. 2006. Insensitivity of paediatric HIV-1 subtype C viruses to broadly neutralising monoclonal antibodies raised against subtype B. PLoS medicine 3:e255. [0431] 22. Alam SM, Morelli M, Dennison SM, Liao HX, Zhang R, Xia SM, Rits- Volloch S, Sun L, Harrison SC, Haynes BF, Chen B. 2009. Role of HIV membrane in neutralization by two broadly neutralizing antibodies. Proceedings of the National Academy of Sciences of the United States of America 106:20234-20239.

[0432] 23. Alam SM, Liao HX, Dennison SM, Jaeger F, Parks R, Anasti K, Foulger A, Donathan M, Lucas J, Verkoczy L, Nicely N, Tomaras GD, Kelsoe G, Chen B, Kepler TB, Haynes BF. 2011. Differential reactivity of germ line allelic variants of a broadly neutralizing HIV-1 antibody to a gp41 fusion intermediate conformation. Journal of virology 85: 11725-11731.

[0433] 24. Shen X, Dennison SM, Liu P, Gao F, Jaeger F, Montefiori DC, Verkoczy L, Haynes BF, Alam SM, Tomaras GD. 2010. Prolonged exposure of the HIV-1 gp41 membrane proximal region with L669S substitution. Proceedings of the National Academy of Sciences of the United States of America 107:5972-5977.

[0434] 25. Frey G, Chen J, Rits-Volloch S, Freeman MM, Zolla-Pazner S, Chen B. 2010.

Distinct conformational states of HIV-1 gp41 are recognized by neutralizing and non- neutralizing antibodies. Nature structural & molecular biology 17: 1486-1491.

[0435] 26. Liao HX, Lynch R, Zhou T, Gao F, Alam SM, Boyd SD, Fire AZ, Roskin KM, Schramm CA, Zhang Z, Zhu J, Shapiro L, Program NCS, Mullikin JC, Gnanakaran S, Hraber P, Wiehe K, Kelsoe G, Yang G, Xia SM, Montefiori DC, Parks R, Lloyd KE, Scearce RM, Soderberg KA, Cohen M, Kamanga G, Louder MK, Tran LM, Chen Y, Cai

F, Chen S, Moquin S, Du X, Joyce MG, Srivatsan S, Zhang B, Zheng A, Shaw GM, Hahn BH, Kepler TB, Korber BT, Kwong PD, Mascola JR, Haynes BF. 2013. Co-evolution of a broadly neutralizing HIV-1 antibody and founder virus. Nature 496:469-476.

[0436] 27. Moody MA, Yates NL, Amos JD, Drinker MS, Eudailey JA, Gurley TC, Marshall DJ, Whitesides JF, Chen X, Foulger A, Yu JS, Zhang R, Meyerhoff RR, Parks R, Scull JC, Wang L, Vandergrift NA, Pickeral J, Pollara J, Kelsoe G, Alam SM, Ferrari

G, Montefiori DC, Voss G, Liao HX, Tomaras GD, Haynes BF. 2012. HIV-1 gpl20 vaccine induces affinity maturation in both new and persistent antibody clonal lineages. Journal of virology 86:7496-7507.

[0437] 28. Cheung WC, Beausoleil SA, Zhang X, Sato S, Schieferl SM, Wieler JS,

Beaudet JG, Ramenani RK, Popova L, Comb MJ, Rush J, Polakiewicz RD. 2012. A proteomics approach for the identification and cloning of monoclonal antibodies from serum. Nature biotechnology 30:447-452.

[0438] 29. Wine Y, Boutz DR, Lavinder JJ, Miklos AE, Hughes RA, Hoi KH, Jung ST, Horton AP, Murrin EM, Ellington AD, Marcotte EM, Georgiou G. 2013. Molecular deconvolution of the monoclonal antibodies that comprise the polyclonal serum response. Proceedings of the National Academy of Sciences of the United States of America 110:2993-2998.

[0439] 30. Lavinder JJ, Horton AP, Georgiou G, Ippolito GC. 2015. Next-generation sequencing and protein mass spectrometry for the comprehensive analysis of human cellular and serum antibody repertoires. Current opinion in chemical biology 24: 112-120.

[0440] 31. Levesque MC, Moody MA, Hwang KK, Marshall DJ, Whitesides JF, Amos JD, Gurley TC, Allgood S, Haynes BB, Vandergrift NA, Plonk S, Parker DC, Cohen MS, Tomaras GD, Goepfert PA, Shaw GM, Schmitz JE, Eron JJ, Shaheen NJ, Hicks CB, Liao HX, Markowitz M, Kelsoe G, Margolis DM, Haynes BF. 2009. Polyclonal B cell differentiation and loss of gastrointestinal tract germinal centers in the earliest stages of HIV-1 infection. PLoS medicine 6:el000107.

[0441] 32. Scheid JF, Mouquet H, Feldhahn N, Seaman MS, Velinzon K, Pietzsch J, Ott RG, Anthony RM, Zebroski H, Hurley A, Phogat A, Chakrabarti B, Li Y, Connors M, Pereyra F, Walker BD, Wardemann H, Ho D, Wyatt RT, Mascola JR, Ravetch JV, Nussenzweig MC. 2009. Broad diversity of neutralizing antibodies isolated from memory B cells in HIV-infected individuals. Nature 458:636-640.

[0442] 33. Tomaras GD, Binley JM, Gray ES, Crooks ET, Osawa K, Moore PL, Tumba N, Tong T, Shen X, Yates NL, Decker J, Wibmer CK, Gao F, Alam SM, Easterbrook P, Abdool Karim S, Kamanga G, Crump JA, Cohen M, Shaw GM, Mascola JR, Haynes BF, Montefiori DC, Morris L. 2011. Polyclonal B cell responses to conserved neutralization epitopes in a subset of HIV-1 -infected individuals. Journal of virology 85: 11502-11519.

[0443] 34. Simek MD, Rida W, Priddy FH, Pung P, Carrow E, Laufer DS, Lehrman JK, Boaz M, Tarragona-Fiol T, Miiro G, Birungi J, Pozniak A, McPhee DA, Manigart O, Karita E, Inwoley A, Jaoko W, Dehovitz J, Bekker LG, Pitisuttithum P, Paris R, Walker LM, Poignard P, Wrin T, Fast PE, Burton DR, Koff WC. 2009. Human immunodeficiency virus type 1 elite neutralizers: individuals with broad and potent neutralizing activity identified by using a high-throughput neutralization assay together with an analytical selection algorithm. Journal of virology 83:7337-7348.

[0444] 35. Walker LM, Phogat SK, Chan-Hui PY, Wagner D, Phung P, Goss JL, Wrin T, Simek MD, Fling S, Mitcham JL, Lehrman JK, Priddy FH, Olsen OA, Frey SM,

Hammond PW, Protocol GPI, Kaminsky S, Zamb T, Moyle M, Koff WC, Poignard P, Burton DR. 2009. Broad and potent neutralizing antibodies from an African donor reveal a new HIV-1 vaccine target. Science 326:285-289.

[0445] 36. Walker LM, Huber M, Doores KJ, Falkowska E, Pejchal R, Julien JP, Wang SK, Ramos A, Chan-Hui PY, Moyle M, Mitcham JL, Hammond PW, Olsen OA, Phung P, Fling S, Wong CH, Phogat S, Wrin T, Simek MD, Protocol GPI, Koff WC, Wilson IA, Burton DR, Poignard P. 2011. Broad neutralization coverage of HIV by multiple highly potent antibodies. Nature 477:466-470.

[0446] 37. Bonsignori M, Hwang KK, Chen X, Tsao CY, Morris L, Gray E, Marshall DJ, Crump JA, Kapiga SH, Sam NE, Sinangil F, Pancera M, Yongping Y, Zhang B, Zhu J, Kwong PD, O'Dell S, Mascola JR, Wu L, Nabel GJ, Phogat S, Seaman MS, Whitesides JF, Moody MA, Kelsoe G, Yang X, Sodroski J, Shaw GM, Montefiori DC, Kepler TB, Tomaras GD, Alam SM, Liao HX, Haynes BF. 2011. Analysis of a clonal lineage of HIV- 1 envelope V2/V3 conformational epitope-specific broadly neutralizing antibodies and their inferred unmutated common ancestors. Journal of virology 85:9998-10009.

[0447] 38. Bonsignori M, Montefiori DC, Wu X, Chen X, Hwang KK, Tsao CY, Kozink DM, Parks RJ, Tomaras GD, Crump JA, Kapiga SH, Sam NE, Kwong PD, Kepler TB, Liao HX, Mascola JR, Haynes BF. 2012. Two distinct broadly neutralizing antibody specificities of different clonal lineages in a single HIV- 1 -infected donor: implications for vaccine design. Journal of virology 86:4688-4692.

[0448] 39. Walker LM, Simek MD, Priddy F, Gach JS, Wagner D, Zwick MB, Phogat SK, Poignard P, Burton DR. 2010. A limited number of antibody specificities mediate broad and potent serum neutralization in selected HIV-1 infected individuals. PLoS pathogens 6:el001028.

[0449] 40. Gao F, Bonsignori M, Liao HX, Kumar A, Xia SM, Lu X, Cai F, Hwang KK, Song H, Zhou T, Lynch RM, Alam SM, Moody MA, Ferrari G, Berrong M, Kelsoe G, Shaw GM, Hahn BH, Montefiori DC, Kamanga G, Cohen MS, Hraber P, Kwong PD, Korber BT, Mascola JR, Kepler TB, Haynes BF. 2014. Cooperation of B cell lineages in induction of HIV- 1 -broadly neutralizing antibodies. Cell 158:481-491.

[0450] 41. Amanna I J, Carlson NE, Slifka MK. 2007. Duration of humoral immunity to common viral and vaccine antigens. The New England journal of medicine 357: 1903- 1915.

[0451] 42. Bonsignori M, Moody MA, Parks RJ, Holl TM, Kelsoe G, Hicks CB,

Vandergrift N, Tomaras GD, Haynes BF. 2009. HIV-1 envelope induces memory B cell responses that correlate with plasma antibody levels after envelope gpl20 protein vaccination or HIV-1 infection. Journal of immunology 183:2708-2717.

[0452] Supplementary Materials and Methods

[0453] Donor Information

[0454] Plasma and peripheral blood mononuclear cells were collected from South African donor CH0210, chronically infected with a clade C virus for an unknown period at the time of enrollment in the Center for HIV/AIDS Vaccine Immunology (CHAVI) 001 chronic HIV-1 infection cohort (previously described in (33). Informed consent was obtained under clinical protocols approved by the Institutional Review Board of the Duke University Health System and clinical site in South Africa. The DH511 bnAb lineage was isolated from PBMC and plasma collected at 8 weeks post-study enrollment, where the viral load was 5, 180 copies/ml and CD4 T cell count was unknown, at which time donor CH0210 had not initiated anti- retro viral therapy (ART).

[0455] Epitope Mapping and Neutralization-based Epitope Prediction Analysis

[0456] Donor CH0210 plasma was screened for neutralization breadth utilizing standard experimental mapping and computational methods for epitope prediction (13, 43). Anti- MPER bnAb activity was detected using two different assays: plasma neutralization of the HIV-2/HIV-1 MPER chimeric pseudovirus C1C and plasma adsorption with MPER peptide coated magnetic beads, followed by testing of adsorbed plasmas for reduction of neutralization activity as described previously (44). An algorithm for Neutralization-based Epitope Prediction (NEP) (13, 43) was used to delineate the specificities mediating breadth against a panel of 21 diverse HIV-1 strains. The resulting linear coefficients on a scale of (0 to 1) from the computational procedure was used to predict the relative prevalence of each of the reference antibody specificities in donor CH0210 plasma. [0457] Antigen-specific Single Memory B Cell Sorting and Antibody Expression

[0458] As previously described (14), fluorescently-labeled MPER peptide tetramer probes were generated using biotinylated MPR.03 peptide

(KKKNEQELLELDKWASLWNWFDITNWLWYIRKKK-biotin SEQ ID NO: 463) (CPC Scientific Inc., San Jose, CA) conjugated to fluorophore-labeled streptavidins, yielding a tetramer with four MPER epitopes for surface Ig cross-linking. Eleven and a half million PBMC from donor CH0210 were stained with MPR.03 -Alexa647 and MPR.03-Brilliant Violet 421 peptide tetramers and a cocktail of antibodies to identify MPER-specific memory B cells: surface IgM (FITC), surface IgD (phycoerythrin [PE]), CD3 (PE-Cy5), CD 16 (Brilliant Violet 570), CD235a (PE-Cy5), and CD 19 (allophycocyanin [APC]-Cy7) (BD Biosciences, San Jose, CA); CD 14 (Brilliant Violet 605) (Invitrogen, Carlsbad, CA); CD27 (PE-Cy7), CD38 (APC-Alexa Fluor 700) (Beckman Coulter, Brea, CA), and CD 10 (ECD) (Beckman Coulter, Brea, CA). Aqua blue vital dye (Invitrogen, Carlsbad, CA) was used to stain dead cells. Using a four laser FACS Aria cell sorter and FACSDiva software (BD Biosciences, San Jose, CA), MPR.03 double positive CD 16-CD 14-CD3-CD235-CD 19+IgD- CD38hi memory B cells were single cell sorted into individual wells of a 96-well plate containing reverse transcription (RT) reaction buffer (5 of 5' first-strand cDNA buffer, 0.5 of RNaseOUT [Invitrogen, Carlsbad, CA], 1.25 μL of dithiothreitol, 0.0625 μL Igepal CA-630 [Sigma, St. Louis, MO], 13.25 μL of distilled H20 [dH20; Invitrogen, Carlsbad, CA]). Data were further analyzed using FlowJo software (TreeStar, Ashland, OR). Plates were stored at -80°C until PCR could be performed.

[0459] PCR Amplification and Expression of Ig Genes

[0460] Immunoglobulin genes were amplified from RNA of isolated cells by reverse transcription- polymerase chain reaction (RT-PCR). For RT, 10 mM dNTPs (New England Biolabs, Ipswich, MA), 3 μL random hexamers at 150 ng/ml (Gene Link, Hawthorne, NY), and 1 μl Superscript ® III (Invitrogen, Carlsbad, CA) were added to each well and subjected to thermocy cling under the following conditions: 42°C for 10 minutes, 25 °C for 10 minutes, 50°C for 60 minutes and 94°C for 5 minutes. IgH, g g variable region genes were separately amplified from the cDNA by nested PCR, using AmpliTaq Gold ® 360 Mastermix (Invitrogen, Carlsbad, CA), heavy-chain (45) and light-chain gene-specific primers as previously described (46). PCR amplicons were purified and sequenced, and VHDJH and VLJL genes, mutation frequencies, and CDR3 lengths were determined using the Clonanalyst software (47). Clonal relatedness and inference of the unmutated common ancestor (UCA) and intermediate antibodies were determined by computational methods as described in (26, 40, 48). Maximum likelihood phylogenetic trees were constructed from V(D)J sequences using the Phylogeny Inference Package (PHYLIP) (version 3.69; (49). Transient small-scale expression of antibodies was achieved by overlapping PCR assembly of variable heavy and light-chain gene pairs into IgH, linear expression cassettes for production of full length IgGl mAbs by transfection into 293T cells as described previously (46). Supernatants were screened for HIV- 1 Env binding by ELISA and neutralization activity in TZM-bl cells. For large scale antibody production, antibody variable heavy-chain and light-chain genes were de novo synthesized (GenScript, Township, NJ), cloned into pcDNA3.1 expression vectors containing the constant regions of IgGl (46), and co-transfected at equal ratios in Expi 293i cells using ExpiFectamine 293 transfection reagents (Thermo Fischer Scientific, Waltham, MA) according to the manufacturer's instructions. Culture supernatants were harvested and concentrated after 4-5 days incubation at 37°C and 8% CO2, followed by affinity purification by protein A column (Pierce, Thermo Fisher Scientific, Waltham, MA). Antibody purity was evaluated by SDS-Page and Coomassie Blue staining for heavy and light-chains of the appropriate size.

[0461] ELISA Assays

[0462] Binding of transiently transfected supernatants and mAbs to HIV-1 Env proteins and peptides was detected by enzyme-linked immunosorbent assay (ELISA). High-binding 384- well plates (Corning, Oneonta, NY) were coated overnight at 4°C or for 2 hours at room temperature with 2 μg/ml HIV-1 protein or streptavidin (for detection of binding to biotinylated peptides) in 0.1 M sodium bicarbonate (Sigma Aldrich, St. Louis, MO). Plates were blocked for 1 hour at room temperature with assay diluent comprised of phosphate buffered saline (PBS), 4% (weight/volume) whey protein (BiPro USA, Prarie, MN), 15% normal goat serum (Invitrogen, Carlsbad, CA), 0.5% Tween 20, and 0.05% sodium azide (Sigma Aldrich, St. Louis, MO), followed by a 1 hour incubation with antibody at a starting concentration of 100 μg/ml, serially diluted 3-fold. Horseradish peroxidase-conjugated goat anti-human IgG Fc antibody (Jackson ImmunoRe search Laboratories, West Grove, PA) was added to each well and incubated for 1 hour, after which plates were washed with PBS/0.1% Tween 20 and developed with SureBlue Reserve TMB One Component Microwell Peroxidase Substrate for 15 minutes (KPL, Gaithersburg, MD). Development was stopped with 0.1 M HC1, and plates were read at 450 nm. Experiments were performed in duplicate, and results were reported as logarithm area under the curve (Log AUC). For epitope mapping, purified mAbs were screened as listed above against a panel of MPR.03 alanine scanned peptides. Epitope positions were defined by MPR.03 alanine scan mutations that reduced the Log AUC by >50% compared to the wild-type peptide.

[0463] Neutralization Assays

[0464] Neutralization assays were performed using HIV-1 Env pseudoviruses to infect TZM- bl cells as previously described (50, 51). A five-parameter hill slope equation was used to fit neutralization curves by non-linear regression and for determination of maximum percent inhibition (MPI) values. Titers were calculated as 50% or 80% inhibitory concentrations (IC50 and IC80) and reported as the concentration of antibody causing a 50% or 80% reduction in relative luminescence units compared to virus control wells. Mapping of the MPER residues critical for neutralization was performed using a panel of alanine scanned COT6.15 Env pseudoviruses as described previously (20, 21).

[0465] Poly/autoreactivity Analysis

[0466] Antibody binding to a panel of nine autoantigens, including Sjogren's syndrome antigen (SSA), SSB, Smith antigen (Sm), ribonucleoprotein (RNP), scleroderma 70 (Scl-70), Jo-1, double-stranded DNA (dsDNA), centromere B (Cent B), and histone, was quantified by ELISA. Anti-cardiolipin reactivity was measured using the QUANTA Lite ACA IgG III ELISA kit (Nova Diagnostics, San Diego, CA) per the manufacturer's instructions as previously described (52). Antibodies were assayed for reactivity to the human epithelial cell line (HEp-2) by indirect immunofluorescence staining using the IFA ANA/Hep-2 Test System (Zeus Scientific, Somerville, NJ) per the manufacturer's protocol. Antibodies were diluted to 50 μg/ml and 25 μg/ml and scored negative or positive (1+ to 4+) at each dilution. Antibodies were also screened for binding to a panel of >9,400 human proteins using a Protoarray microarray (Invitrogen, Carlsbad, CA) according to the manufacturer's instructions and as described in (6). Briefly, the array was blocked and incubated on ice with 2 μg/ml HIV-1 antibody or the isotype control antibody, human myeloma protein, 15 IK (Southern Biotech, Birmingham, AL) for 90 minutes. Antibody binding was detected with 1 μg/ml anti- human IgG-Alexa-647 secondary antibody (Invitrogen). Arrays were scanned using a GenePix 4000B scanner (Molecular Devices, Sunnyvale, CA) at a wavelength of 635 nm, 10 μπι resolution, using 100% power and 650 gain. The fluorescence intensity of antibody binding was measured with the GenePix Pro 5.0 program (Molecular Devices, Sunnyvale, CA).

[0467] Surface Plasmon Resonance Affinity and Kinetics Measurements

[0468] Surface plasmon resonance analysis was performed on a Biacore 3000 instrument (GE Healthcare, Little Chalfont, UK) at 25°C and data analyzed using the BIAevaluation 4.1 software (BIAcore) as described previously (Alam et al. JI 2007). To determine the affinity, association and dissociation rate constants of the DH511 clonal lineage to MPER, biotinylated MPR.03 peptide was coated on streptavidin sensors at a density of 58 response units (RUs). DH511 lineage Fabs were injected over flow cells at increasing concentrations at a flow and minute dissociation steps. Curves were blank surface and CH58 Fab analyte subtracted. Peptide-liposome conjugates were generated with MPER656.1-GTH1 peptides using an extrusion method (4) and analyzed for binding in a two-step encounter docking model as described previously (4).

[0469] Time Course of DH511.2 Neutralization

[0470] The time course of DH511.2 neutralization was determined using a post-attachment HIV-1 pseudotyped virus neutralization assay described previously (53). Inhibitory concentrations of DH511.2, 10E8, and 4E10 mAb were added to TZM-bl cells incubated with B.BG1168 virus at different time intervals after infection. Infectivity was measured in relative light units (RLUs).

[0471] High-throughput Paired VH:VL Sequencing of Immunoglobulin Transcripts [0472] Material & reagents. Protein G Plus agarose, NeutrAvidin agarose, immobilized pepsin resin and Hypersep SpinTip C18 columns (C18-SpinTips) were acquired from Pierce (Thermo Fisher Scientific, Rockford, IL). TRIS hydrocholoride (Tris-HCl), ammonium bicarbonate (NH4HC03), 2,2,2-trifluoroethanol (TFE), dithiothrietol (DTT), and iodoacetamide (IAM) were obtained from Sigma-Aldrich (St. Louis, MO). LC-MS grade water, acetonitrile (ACN), and formic acid were purchased from EMD (Billerica, MA).

[0473] Isolation of memory B cells. Frozen PBMCs (10 million cells in 1 mL) were thawed at 37°C, resuspended in 50 mL of RPMI 1640 (Lonza) supplemented with 10% Fetal Bovine Serum, lx non-essential amino acids, lx sodium pyruvate, lx glutamine, lx penicillin/streptomycin, and 20 U/mL DNAse I, and recovered via centrifugation (300 g for 10 min at 20°C). The cells were then resuspended in 4 mL of RPMI and allowed to recover at 37°C for 30 min. The cells were diluted with 10 mL of cold MACS buffer (PBS supplemented with 0.5% BSA and 2 mM EDTA), collected by centrifugation (300 g for 10 min at 4°C), and depleted of non-B cells using the Human Memory B Cell Isolation Kit with an LD column (Miltenyi Biotec) as per the manufacturer's instructions. This yielded 400,000 - 500,000 B cells per vial.

[0474] Amplification of the paired VH:VL repertoire. The paired VH and VL sequences were then determined using a custom designed axisymmetric flow focusing device (19) that is comprised of three concentric tubes. Total B cells were suspended in 6 mL of cold PBS and passed through the innermost tube at a rate of 0.5 mL/min. Oligo d(T)25 magnetic beads (1 μιη diameter at a concentration of 45 beads/mL solution; NEB) were washed, subjected to focused ultrasonication (Covaris) to dissociate any aggregates, resuspended in 6 mL of lysis buffer ( 100 mM Tris-HCl pH 7.5, 500 mM LiCl, 10 mM EDTA, 1% Lithium dodecyl sulfate (LiDS), 5 mM DTT), and passed through the middle tube at a rate of 0.5 mL/min. The outer tubing contained an oil phase (mineral oil containing 4.5% Span-80, 0.4% Tween-80, and 0.05% Triton X-100; Sigma-Aldrich) flowing at 3 mL/min. The cells, beads, and lysis buffer were emulsified as they passed through a custom designed 120 μιη diameter orifice, and were subsequently collected in 2 mL microcentrifuge tubes. Each tube was inverted several times, incubated at 20°C for 3 minutes, and then placed on ice. Following the collection phase, emulsions were pooled into 50 mL conicals, and centrifuged (4,000 g for 5 min at 4°C). The mineral oil (upper phase) was decanted, and the emulsions (bottom phase) were broken with water-saturated cold diethyl ether (Fischer). Magnetic beads were recovered following a second centrifugation step (4,000 g for 5 min at 4°C) and resuspended in 1 mL of cold Buffer 1 (100 mM Tris pH 7.5, 500 mM LiCl, 10 mM EDTA, 1% LiDS, 5 mM DTT). The beads were then serially pelleted using a magnetic rack, and washed with the following buffers: 1 mL lysis buffer, 1 mL Buffer 1, and 0.5 mL Buffer 2 (20 mM Tris pH 7.5, 50 mM KC1, 3 mM MgCl). The beads were split into two aliquots, and each was then pelleted one final time and resuspended in an RT-PCR mixture (19) containing VH and VL Framework Region 1 (FR1) linkage primers or VH and VL leader peptide (LP) linkage primers (Supplementary Tables 28 and 29). The RT-PCR mixtures were then added dropwise to 9 mL of chilled oil phase in an IKA dispersing tube (DT-20, VWR) and emulsified using an emulsion dispersing apparatus (Ultra-Turrax® Tube Drive; IKA) for 5 min. The emulsions were aliquoted into 96- well PCR plates ( 100 uL/well), and subjected to RT-PCR under the following conditions: 30 min at 55°C followed by 2 min at 94°C; 4 cycles of 94°C for 30 s, 50°C for 30 s, 72°C for 2 min; 4 cycles of 94°C for 30 s, 55°C for 30 s, 72°C for 2 min; 32 cycles of 94°C for 30 s, 60°C for 30 s, 72°C for 2 min; 72°C for 7 min; held at 4°C.

[0475] Following RT-PCR, the emulsions were collected in 2 mL microcentrifuge tubes and centrifuged (16000 g for 10 min at 20°C). The mineral oil (upper phase) was decanted, and water-saturated ether was used to break the emulsions. The aqueous phase (containing the DNA) was extracted three times by sequentially adding ether, centrifuging the samples (16000 g for 30 s at 20°C), and removing the upper ether phase. Trace amounts of ether were removed using a SpeedVac for 30 min at 20°C. The DNA amplicons were purified using a silica spin column (Zymo-Spin™ I, Zymo Research) according to the manufacturer's instructions, and eluted in 40 H2O. The two samples were then amplified through a nested PCR (see Supplementary Table 30 for primers) using Platinum Taq (Life Technologies) under the following conditions: (FRl primer derived sample) 2 min at 94°C, 32 cycles of 94°C for 30 s, 62°C for 30 s, 72°C for 20 s; 72°C for 7 min; held at 4°C; (LP primer derived sample) 2 min at 94°C, 27 cycles of 94°C for 30 s, 62°C for 30 s, 72°C for 20 s; 72°C for 7 min; held at 4°C. The amplicons, approximately 850 bp in length, were gel purified from 1% agarose using a gel extraction kit (Zymo Research) according to the manufacturer's instructions, and eluted in 20 H 2 O.

[0476] To determine the full length VH and VL reads for antibody expression studies, the paired amplicon was subjected to an additional PCR using NEBNext high fidelity polymerase (NEB) to specifically amplify the full VH chain and the full VL chain separately in addition to the paired chains (Note: the paired reads sequence the entire J- and D-regions, and the fragment of the V regions spanning FR2 to CDR3). Each sample was split into 5 reactions and subjected to the following PCR conditions: 30 s at 98°C, X cycles of 98°C for 10 s, 62°C for 30 s, 72°C for Y s; 72°C for 7 min; held at 4°C (See Supplementary Table 31 for the PCR conditions and Supplementary Table 32 for the primer sequences). Finally, these sequences were amplified one final time with TSBC compatible barcoding primers following the protocol shown in Supplementary Table 33, gel purified from 1% agarose using a gel purification kit according to manufacturer's instructions, and submitted for paired-end Illumina NGS.

[0477] Bioinformatic analysis of NGS data. Raw 2x300 MiSeq reads were quality filtered (minimum Phred score of 20 over half of the nucleotide sequence) and submitted to MiXCR (54) for CDR3 identification and gene annotation. Productive VH and VL reads were paired by Illumina MiSeq ID using a custom python script. Full length VH and VL reads were stitched together using FLAsH (55) and then quality filtered. Full length VH and VL constructs were designed by matching the paired CDRH3:CDRL3 nucleotide sequences to the respective CDR3 in the full length VH and VL libraries.

[0478] Sample preparation & LC-MS/MS analysis. Serum IgG from donor 0210 was purified by Protein G Plus agarose affinity chromatography, and F(ab')2 fragments were generated by digestion with immobilized pepsin. Antigen-specific F(ab')2 was isolated by affinity chromatography with the biotinylated MPER peptide coupled to NeutrAvidin agarose and eluted in 100 mM glycine pH 2.7. The collected fractions were neutralized and the protein containing fractions were pooled and prepared for LC-MS/MS as described previously (10). Briefly, protein samples were concentrated and resuspended in 50% (v/v) TFE, 50 mM NH4HCO3 and 2.5 mM DTT and incubated at 55°C for 45 min. The reduced samples were then alkylated with IAM in the dark, at room temperature for 30 min. The reaction was quenched by addition of DTT and the samples were diluted to 5% TFE and digested with trypsin (trypsin/protein ration of 1 :75 at 37°C for 5 h). The digestion was stopped by addition of formic acid to 1% (v/v). The samples were then concentrated by SpeedVac, resuspended in 5% ACN, 0.1% formic acid and the peptides were washed on C18-SpinTips according to the manufacturer's protocol. Subsequently, the peptides were separated by reverse phase chromatography (Dionex UltiMate 3000 RSLCnano system with Dionex Acclaim PepMapRSLC C18 column, Thermo Scientific) and analyzed on-line by nano-ESI tandem MS on an Orbitrap Velos Pro (Thermo Scientific). MSI scans were collected in the orbitrap at 60,000 resolution and ions with >+l charge were fragmented by CID with up to 20 MS2 spectra collected per MS 1.

[0479] Computational interpretation of peptide mass spectra. Full length VH and VL sequencing data (see above) was submitted to the IMGT/HighV-Quest Tool (56) for annotation and unique full length VH sequences were clustered into clonotypes according to their CDRH3 sequences with a cut-off of 85% identity as described previously (29). The sample-specific target protein sequence database was constructed from the full-length VH and VL sequences mentioned above (>2 reads), Ensembl human protein-coding sequences and common contaminants (maxquant.org). The spectra were then searched against this database using the SEQUEST (Proteome Discoverer 1.4, Thermo Scientific) with previously described settings (15). The resulting PSMs were filtered with Percolator (Proteome Discoverer 1.4) to control false discovery rates (FDR) to <1% and the average mass deviation (AMD) was calculated for all high-confidence PSMs and peptides with an AMD of <1.5 ppm were kept for the final dataset. Informative peptides, as defined previously (15), were grouped by their CDRHl, 2 or 3 association and for each group the abundances of the corresponding clonotypes were determined by the sum of the extracted-ion chromatograms of the respective precursor ions.

[0480] Crystallization, Structure Determination, and Structural Analysis.

[0481] Purified DH511.1 and DH511.2 fragments of antigen binding (Fabs) were set up in crystallization trials in complex with a panel of gp41 MPER peptides. For each complex, 576 initial conditions from commercially available screens (Hampton Research, Rigaku) were set up as vapor diffusion sitting drops robotically (TTP Labtech). Crystals of DH511 Fab in complex with gp41 MPER peptide 656-683 were obtained in a condition composed of 30% PEG 1500, while those of DH511.2 Fab in complex with peptides MPR.03.DN4 and MPR.03.DN14, were obtained in 30% PEG 1500, 10% Isopropanol, 0.1 M CaCl2, 0.1 M Imidazole pH 6.5 and in 20% PEG 8000, 10% PEG 400, 0.5 M NaCl, 0.1 M C2H3NaO2 pH 5.5, respectively. Crystal hits were hand optimized and X-ray diffraction data extended to 2.8, 2.65, and 2.2 A, respectively. Data was processed with HKL-2000 (57) and structures were solved by molecular replacement using the DH514 Fab unliganded structure as a search model in Phaser (58). The structures were refined to Rcrystal/Rfree of 21.28/25.57, 25.61/28.99, and 19.03/22.63%, respectively, using Phenix (59) combined with iterative model building in Coot (60). Interactive surfaces were determined using Pisa (61) and structural alignments using LSQKAB (62). All graphical images were prepared with Pymol (PyMOL Molecular Graphics System). X-ray diffraction data was collected at SER CAT ID-22 or BM-22 beamlines of the Advanced Photon Source (Argonne, IL), under General User Proposal 44127 (G O ).

[0482] Supplemental References

[0483] 1. Huang J, Ofek G, Laub L, Louder MK, Doria-Rose NA, Longo NS, Imamichi H, Bailer RT, Chakrabarti B, Sharma SK, Alam SM, Wang T, Yang Y, Zhang B, Migueles SA, Wyatt R, Haynes BF, Kwong PD, Mascola JR, Connors M. 2012. Broad and potent neutralization of HIV-1 by a gp41-specific human antibody. Nature 491 :406- 412. [0484] 2. Zwick MB, Labrijn AF, Wang M, Spenlehauer C, Saphire EO, Binley JM, Moore JP, Stiegler G, Katinger H, Burton DR, Parren PW. 2001. Broadly neutralizing antibodies targeted to the membrane -proximal external region of human

immunodeficiency virus type 1 glycoprotein gp41. Journal of virology 75: 10892-10905.

[0485] 3. Yang G, Holl TM, Liu Y, Li Y, Lu X, Nicely NI, Kepler TB, Alam SM, Liao HX, Cain DW, Spicer L, VandeBerg JL, Haynes BF, Kelsoe G. 2013. Identification of autoantigens recognized by the 2F5 and 4E10 broadly neutralizing HIV-1 antibodies. The Journal of experimental medicine 210:241-256.

[0486] 4. Alam SM, McAdams M, Boren D, Rak M, Scearce RM, Gao F, Camacho ZT, Gewirth D, Kelsoe G, Chen P, Haynes BF. 2007. The role of antibody polyspecificity and lipid reactivity in binding of broadly neutralizing anti-HIV-1 envelope human monoclonal antibodies 2F5 and 4E10 to glycoprotein 41 membrane proximal envelope epitopes. Journal of immunology 178:4424-4435.

[0487] 5. Chen J, Frey G, Peng H, Rits-Volloch S, Garrity J, Seaman MS, Chen B. 2014.

Mechanism of HIV-1 neutralization by antibodies targeting a membrane-proximal region of gp41. Journal of virology 88: 1249-1258.

[0488] 6. Liu M, Yang G, Wiehe K, Nicely NI, Vandergrift NA, Rountree W,

Bonsignori M, Alam SM, Gao J, Haynes BF, Kelsoe G. 2015. Polyreactivity and autoreactivity among HIV-1 antibodies. Journal of virology 89:784-798.

[0489] 7. Haynes BF, Gilbert PB, McElrath MJ, Zolla-Pazner S, Tomaras GD, Alam SM, Evans DT, Montefiori DC, Karnasuta C, Sutthent R, Liao HX, DeVico AL, Lewis GK, Williams C, Pinter A, Fong Y, Janes H, DeCamp A, Huang Y, Rao M, Billings E, Karasavvas N, Robb ML, Ngauy V, de Souza MS, Paris R, Ferrari G, Bailer RT, Soderberg KA, Andrews C, Berman PW, Frahm N, De Rosa SC, Alpert MD, Yates NL, Shen X, Koup RA, Pitisuttithum P, Kaewkungwal J, Nitayaphan S, Rerks-Ngarm S, Michael NL, Kim JH. 2012. Immune-correlates analysis of an HIV-1 vaccine efficacy trial. The New England journal of medicine 366: 1275-1286.

[0490] 8. Moir S, Ho J, Malaspina A, Wang W, DiPoto AC, O'Shea MA, Roby G,

Kottilil S, Arthos J, Proschan MA, Chun TW, Fauci AS. 2008. Evidence for HIV- associated B cell exhaustion in a dysfunctional memory B cell compartment in HIV- infected viremic individuals. The Journal of experimental medicine 205: 1797-1805. [0491] 9. Kardava L, Moir S, Shah N, Wang W, Wilson R, Buckner CM, Santich BH, Kim LJ, Spurlin EE, Nelson AK, Wheatley AK, Harvey CJ, McDermott AB,

Wucherpfennig KW, Chun TW, Tsang JS, Li Y, Fauci AS. 2014. Abnormal B cell memory subsets dominate HIV-specific responses in infected individuals. The Journal of clinical investigation 124:3252-3262.

[0492] 10. Boutz DR, Horton AP, Wine Y, Lavinder JJ, Georgiou G, Marcotte EM. 2014.

Proteomic identification of monoclonal antibodies from serum. Analytical chemistry 86:4758-4766.

[0493] 11. Wrammert J, Koutsonanos D, Li GM, Edupuganti S, Sui J, Morrissey M, McCausland M, Skountzou I, Hornig M, Lipkin WI, Mehta A, Razavi B, Del Rio C, Zheng NY, Lee JH, Huang M, Ali Z, Kaur K, Andrews S, Amara RR, Wang Y, Das SR, O'Donnell CD, Yewdell JW, Subbarao K, Marasco WA, Mulligan MJ, Compans R, Ahmed R, Wilson PC. 2011. Broadly cross-reactive antibodies dominate the human B cell response against 2009 pandemic H1N1 influenza virus infection. The Journal of experimental medicine 208: 181-193.

[0494] 12. Purtha WE, Tedder TF, Johnson S, Bhattacharya D, Diamond MS. 2011.

Memory B cells, but not long-lived plasma cells, possess antigen specificities for viral escape mutants. The Journal of experimental medicine 208:2599-2606.

[0495] 13. Georgiev IS, Doria-Rose NA, Zhou T, Kwon YD, Staupe RP, Moquin S, Chuang GY, Louder MK, Schmidt SD, Altae-Tran HR, Bailer RT, McKee K, Nason M, O'Dell S, Ofek G, Pancera M, Srivatsan S, Shapiro L, Connors M, Migueles SA, Morris L, Nishimura Y, Martin MA, Mascola JR, Kwong PD. 2013. Delineating antibody recognition in polyclonal sera from patterns of HIV- 1 isolate neutralization. Science 340:751-756.

[0496] 14. Morris L, Chen X, Alam M, Tomaras G, Zhang R, Marshall DJ, Chen B, Parks R, Foulger A, Jaeger F, Donathan M, Bilska M, Gray ES, Abdool Karim SS, Kepler TB, Whitesides J, Montefiori D, Moody MA, Liao HX, Haynes BF. 2011. Isolation of a human anti-HIV gp41 membrane proximal region neutralizing antibody by antigen- specific single B cell sorting. PloS one 6:e23532.

[0497] 15. Lavinder JJ, Wine Y, Giesecke C, Ippolito GC, Horton AP, Lungu OI, Hoi KH, DeKosky BJ, Murrin EM, Wirth MM, Ellington AD, Dorner T, Marcotte EM, Boutz DR, Georgiou G. 2014. Identification and characterization of the constituent human serum antibodies elicited by vaccination. Proceedings of the National Academy of Sciences of the United States of America 11 1 :2259-2264.

[0498] 16. Wine Y, Horton AP, Ippolito GC, Georgiou G. 2015. Serology in the 21st century: the molecular-level analysis of the serum antibody repertoire. Current opinion in immunology 35 : 89-97.

[0499] 17. McDaniel JR, DeKosky BJ, Tanno H, Ellington AD, Georgiou G. 2016. Ultra- high-throughput sequencing of the immune receptor repertoire from millions of lymphocytes. Nature protocols 1 1 :429-442.

[0500] 18. DeKosky B J, Ippolito GC, Deschner RP, Lavinder JJ, Wine Y, Rawlings BM, Varadarajan N, Giesecke C, Dorner T, Andrews SF, Wilson PC, Hunicke-Smith SP, Willson CG, Ellington AD, Georgiou G. 2013. High-throughput sequencing of the paired human immunoglobulin heavy and light chain repertoire. Nature biotechnology 31 : 166- 169.

[0501] 19. DeKosky BJ, Kojima T, Rodin A, Charab W, Ippolito GC, Ellington AD, Georgiou G. 2015. In-depth determination and analysis of the human paired heavy- and light-chain antibody repertoire. Nature medicine 21 :86-91.

[0502] 20. Gray ES, Madiga MC, Moore PL, Mlisana K, Abdool Karim SS, Binley JM, Shaw GM, Mascola JR, Morris L. 2009. Broad neutralization of human

immunodeficiency virus type 1 mediated by plasma antibodies against the gp41 membrane proximal external region. Journal of virology 83 : 1 1265- 1 1274.

[0503] 21. Gray ES, Meyers T, Gray G, Montefiori DC, Morris L. 2006. Insensitivity of paediatric HIV- 1 subtype C viruses to broadly neutralising monoclonal antibodies raised against subtype B. PLoS medicine 3 :e255.

[0504] 22. Alam SM, Morelli M, Dennison SM, Liao HX, Zhang R, Xia SM, Rits- Volloch S, Sun L, Harrison SC, Haynes BF, Chen B. 2009. Role of HIV membrane in neutralization by two broadly neutralizing antibodies. Proceedings of the National Academy of Sciences of the United States of America 106:20234-20239.

[0505] 23. Alam SM, Liao HX, Dennison SM, Jaeger F, Parks R, Anasti K, Foulger A, Donathan M, Lucas J, Verkoczy L, Nicely N, Tomaras GD, Kelsoe G, Chen B, Kepler TB, Haynes BF. 2011. Differential reactivity of germ line allelic variants of a broadly neutralizing HIV-1 antibody to a gp41 fusion intermediate conformation. Journal of virology 85 : 1 1725- 11731. [0506] 24. Shen X, Dennison SM, Liu P, Gao F, Jaeger F, Montefiori DC, Verkoczy L, Haynes BF, Alam SM, Tomaras GD. 2010. Prolonged exposure of the HIV-1 gp41 membrane proximal region with L669S substitution. Proceedings of the National Academy of Sciences of the United States of America 107:5972-5977.

[0507] 25. Frey G, Chen J, Rits-Volloch S, Freeman MM, Zolla-Pazner S, Chen B. 2010.

Distinct conformational states of HIV-1 gp41 are recognized by neutralizing and non- neutralizing antibodies. Nature structural & molecular biology 17: 1486-1491.

[0508] 26. Liao HX, Lynch R, Zhou T, Gao F, Alam SM, Boyd SD, Fire AZ, Roskin KM, Schramm CA, Zhang Z, Zhu J, Shapiro L, Program NCS, Mullikin JC, Gnanakaran S, Hraber P, Wiehe K, Kelsoe G, Yang G, Xia SM, Montefiori DC, Parks R, Lloyd KE, Scearce RM, Soderberg KA, Cohen M, Kamanga G, Louder MK, Tran LM, Chen Y, Cai

F, Chen S, Moquin S, Du X, Joyce MG, Srivatsan S, Zhang B, Zheng A, Shaw GM, Hahn BH, Kepler TB, Korber BT, Kwong PD, Mascola JR, Haynes BF. 2013. Co-evolution of a broadly neutralizing HIV-1 antibody and founder virus. Nature 496:469-476.

[0509] 27. Moody MA, Yates NL, Amos JD, Drinker MS, Eudailey JA, Gurley TC, Marshall DJ, Whitesides JF, Chen X, Foulger A, Yu JS, Zhang R, Meyerhoff RR, Parks R, Scull JC, Wang L, Vandergrift NA, Pickeral J, Pollara J, Kelsoe G, Alam SM, Ferrari

G, Montefiori DC, Voss G, Liao HX, Tomaras GD, Haynes BF. 2012. HIV-1 gpl20 vaccine induces affinity maturation in both new and persistent antibody clonal lineages. Journal of virology 86:7496-7507.

[0510] 28. Cheung WC, Beausoleil SA, Zhang X, Sato S, Schieferl SM, Wieler JS,

Beaudet JG, Ramenani RK, Popova L, Comb MJ, Rush J, Polakiewicz RD. 2012. A proteomics approach for the identification and cloning of monoclonal antibodies from serum. Nature biotechnology 30:447-452.

[0511] 29. Wine Y, Boutz DR, Lavinder JJ, Miklos AE, Hughes RA, Hoi KH, Jung ST, Horton AP, Murrin EM, Ellington AD, Marcotte EM, Georgiou G. 2013. Molecular deconvolution of the monoclonal antibodies that comprise the polyclonal serum response. Proceedings of the National Academy of Sciences of the United States of America 110:2993-2998.

[0512] 30. Lavinder JJ, Horton AP, Georgiou G, Ippolito GC. 2015. Next-generation sequencing and protein mass spectrometry for the comprehensive analysis of human cellular and serum antibody repertoires. Current opinion in chemical biology 24: 112-120. [0513] 31. Levesque MC, Moody MA, Hwang KK, Marshall DJ, Whitesides JF, Amos JD, Gurley TC, Allgood S, Haynes BB, Vandergrift NA, Plonk S, Parker DC, Cohen MS, Tomaras GD, Goepfert PA, Shaw GM, Schmitz JE, Eron JJ, Shaheen NJ, Hicks CB, Liao HX, Markowitz M, Kelsoe G, Margolis DM, Haynes BF. 2009. Polyclonal B cell differentiation and loss of gastrointestinal tract germinal centers in the earliest stages of HIV-1 infection. PLoS medicine 6:el000107.

[0514] 32. Scheid JF, Mouquet H, Feldhahn N, Seaman MS, Velinzon K, Pietzsch J, Ott RG, Anthony RM, Zebroski H, Hurley A, Phogat A, Chakrabarti B, Li Y, Connors M, Pereyra F, Walker BD, Wardemann H, Ho D, Wyatt RT, Mascola JR, Ravetch JV, Nussenzweig MC. 2009. Broad diversity of neutralizing antibodies isolated from memory B cells in HIV-infected individuals. Nature 458:636-640.

[0515] 33. Tomaras GD, Binley JM, Gray ES, Crooks ET, Osawa K, Moore PL, Tumba N, Tong T, Shen X, Yates NL, Decker J, Wibmer CK, Gao F, Alam SM, Easterbrook P, Abdool Karim S, Kamanga G, Crump JA, Cohen M, Shaw GM, Mascola JR, Haynes BF, Montefiori DC, Morris L. 2011. Polyclonal B cell responses to conserved neutralization epitopes in a subset of HIV-1 -infected individuals. Journal of virology 85: 11502-11519.

[0516] 34. Simek MD, Rida W, Priddy FH, Pung P, Carrow E, Laufer DS, Lehrman JK, Boaz M, Tarragona-Fiol T, Miiro G, Birungi J, Pozniak A, McPhee DA, Manigart O, Karita E, Inwoley A, Jaoko W, Dehovitz J, Bekker LG, Pitisuttithum P, Paris R, Walker LM, Poignard P, Wrin T, Fast PE, Burton DR, Koff WC. 2009. Human immunodeficiency virus type 1 elite neutralizers: individuals with broad and potent neutralizing activity identified by using a high-throughput neutralization assay together with an analytical selection algorithm. Journal of virology 83:7337-7348.

[0517] 35. Walker LM, Phogat SK, Chan-Hui PY, Wagner D, Phung P, Goss JL, Wrin T, Simek MD, Fling S, Mitcham JL, Lehrman JK, Priddy FH, Olsen OA, Frey SM,

Hammond PW, Protocol GPI, Kaminsky S, Zamb T, Moyle M, Koff WC, Poignard P, Burton DR. 2009. Broad and potent neutralizing antibodies from an African donor reveal a new HIV-1 vaccine target. Science 326:285-289.

[0518] 36. Walker LM, Huber M, Doores KJ, Falkowska E, Pejchal R, Julien JP, Wang SK, Ramos A, Chan-Hui PY, Moyle M, Mitcham JL, Hammond PW, Olsen OA, Phung P, Fling S, Wong CH, Phogat S, Wrin T, Simek MD, Protocol GPI, Koff WC, Wilson IA, Burton DR, Poignard P. 2011. Broad neutralization coverage of HIV by multiple highly potent antibodies. Nature 477:466-470.

[0519] 37. Bonsignori M, Hwang KK, Chen X, Tsao CY, Morris L, Gray E, Marshall DJ, Crump JA, Kapiga SH, Sam NE, Sinangil F, Pancera M, Yongping Y, Zhang B, Zhu J, Kwong PD, O'Dell S, Mascola JR, Wu L, Nabel GJ, Phogat S, Seaman MS, Whitesides JF, Moody MA, Kelsoe G, Yang X, Sodroski J, Shaw GM, Montefiori DC, Kepler TB, Tomaras GD, Alam SM, Liao HX, Haynes BF. 2011. Analysis of a clonal lineage of HIV- 1 envelope V2/V3 conformational epitope-specific broadly neutralizing antibodies and their inferred unmutated common ancestors. Journal of virology 85:9998-10009.

[0520] 38. Bonsignori M, Montefiori DC, Wu X, Chen X, Hwang KK, Tsao CY, Kozink DM, Parks RJ, Tomaras GD, Crump JA, Kapiga SH, Sam NE, Kwong PD, Kepler TB, Liao HX, Mascola JR, Haynes BF. 2012. Two distinct broadly neutralizing antibody specificities of different clonal lineages in a single HIV-1 -infected donor: implications for vaccine design. Journal of virology 86:4688-4692.

[0521] 39. Walker LM, Simek MD, Priddy F, Gach JS, Wagner D, Zwick MB, Phogat SK, Poignard P, Burton DR. 2010. A limited number of antibody specificities mediate broad and potent serum neutralization in selected HIV-1 infected individuals. PLoS pathogens 6:el001028.

[0522] 40. Gao F, Bonsignori M, Liao HX, Kumar A, Xia SM, Lu X, Cai F, Hwang KK, Song H, Zhou T, Lynch RM, Alam SM, Moody MA, Ferrari G, Berrong M, Kelsoe G, Shaw GM, Hahn BH, Montefiori DC, Kamanga G, Cohen MS, Hraber P, Kwong PD, Korber BT, Mascola JR, Kepler TB, Haynes BF. 2014. Cooperation of B cell lineages in induction of HIV-1 -broadly neutralizing antibodies. Cell 158:481-491.

[0523] 41. Amanna I J, Carlson NE, Slifka MK. 2007. Duration of humoral immunity to common viral and vaccine antigens. The New England journal of medicine 357: 1903- 1915.

[0524] 42. Bonsignori M, Moody MA, Parks RJ, Holl TM, Kelsoe G, Hicks CB,

Vandergrift N, Tomaras GD, Haynes BF. 2009. HIV-1 envelope induces memory B cell responses that correlate with plasma antibody levels after envelope gpl20 protein vaccination or HIV-1 infection. Journal of immunology 183:2708-2717.

[0525] 43. Chuang GY, Acharya P, Schmidt SD, Yang Y, Louder MK, Zhou T, Kwon YD, Pancera M, Bailer RT, Doria-Rose NA, Nussenzweig MC, Mascola JR, Kwong PD, Georgiev IS. 2013. Residue-level prediction of HIV- 1 antibody epitopes based on neutralization of diverse viral strains. Journal of virology 87: 10047-10058.

[0526] 44. Gray ES, Taylor N, Wycuff D, Moore PL, Tomaras GD, Wibmer CK, Puren A, DeCamp A, Gilbert PB, Wood B, Montefiori DC, Binley JM, Shaw GM, Haynes BF, Mascola JR, Morris L. 2009. Antibody specificities associated with neutralization breadth in plasma from human immunodeficiency virus type 1 subtype C-infected blood donors. Journal of virology 83:8925-8937.

[0527] 45. Tiller T, Meffire E, Yurasov S, Tsuiji M, Nussenzweig MC, Wardemann H.

2008. Efficient generation of monoclonal antibodies from single human B cells by single cell RT-PCR and expression vector cloning. Journal of immunological methods 329: 112- 124.

[0528] 46. Liao HX, Levesque MC, Nagel A, Dixon A, Zhang R, Walter E, Parks R,

Whitesides J, Marshall DJ, Hwang KK, Yang Y, Chen X, Gao F, Munshaw S, Kepler TB,

Denny T, Moody MA, Haynes BF. 2009. High-throughput isolation of immunoglobulin genes from single human B cells and expression as monoclonal antibodies. Journal of virological methods 158: 171-179.

[0529] 47. Kepler TB. 2013. Reconstructing a B-cell clonal lineage. I. Statistical inference of unobserved ancestors. FlOOOResearch 2: 103.

[0530] 48. Kepler TB, Munshaw S, Wiehe K, Zhang R, Yu JS, Woods CW, Denny TN,

Tomaras GD, Alam SM, Moody MA, Kelsoe G, Liao HX, Haynes BF. 2014.

Reconstructing a B-Cell Clonal Lineage. II. Mutation, Selection, and Affinity Maturation.

Frontiers in immunology 5: 170.

[0531] 49. Felstein J. 2009. PHYLIP (Phylogeny Inference Package) version 3.69.

Distributed by the author. Department of Genome Sciences, University of Washington,

Seattle.

[0532] 50. Montefiori DC. 2005. Evaluating neutralizing antibodies against HIV, SIV, and SHIV in luciferase reporter gene assays. Current protocols in immunology / edited by John E. Coligan ... [et al.] Chapter 12:Unit 12 11.

[0533] 51. Seaman MS, Janes H, Hawkins N, Grandpre LE, Devoy C, Giri A, Coffey RT, Harris L, Wood B, Daniels MG, Bhattacharya T, Lapedes A, Polonis VR, McCutchan FE, Gilbert PB, Self SG, Korber BT, Montefiori DC, Mascola JR. 2010. Tiered categorization of a diverse panel of HIV- 1 Env pseudoviruses for assessment of neutralizing antibodies.

Journal of virology 84: 1439-1452.

[0534] 52. Haynes BF, Fleming J, St Clair EW, Katinger H, Stiegler G, Kunert R,

Robinson J, Scearce RM, Plonk K, Staats HF, Ortel TL, Liao HX, Alam SM. 2005.

Cardiolipin poly specific autoreactivity in two broadly neutralizing HIV-1 antibodies.

Science 308: 1906-1908.

[0535] 53. Sun ZY, Oh KJ, Kim M, Yu J, Brusic V, Song L, Qiao Z, Wang JH, Wagner

G, Reinherz EL. 2008. HIV-1 broadly neutralizing antibody extracts its epitope from a kinked gp41 ectodomain region on the viral membrane. Immunity 28:52-63.

[0536] 54. Bolotin DA, Poslavsky S, Mitrophanov I, Shugay M, Mamedov IZ, Putintseva

EV, Chudakov DM. 2015. MiXCR: software for comprehensive adaptive immunity profiling. Nature methods 12:380-381.

[0537] 55. Magoc T, Salzberg SL. 2011. FLASH: fast length adjustment of short reads to improve genome assemblies. Bioinformatics 27:2957-2963.

[0538] 56. Alamyar E, Duroux P, Lefranc MP, Giudicelli V. 2012. IMGT((R)) tools for the nucleotide analysis of immunoglobulin (IG) and T cell receptor (TR) V-(D)-J repertoires, polymorphisms, and IG mutations: IMGT/V-QUEST and IMGT/HighV-

QUEST for NGS. Methods in molecular biology 882:569-604.

[0539] 57. Otwinowski Z, Minor W. 1997. Processing of X-ray diffraction data collected in oscillation mode. Methods Enzymol. 276:307-326.

[0540] 58. Adams PD, Afonine PV, Bunkoczi G, Chen VB, Davis IW, Echols N, Headd

JJ, Hung LW, Kapral GJ, Grosse-Kunstleve RW, McCoy AJ, Moriarty NW, Oeffher R,

Read RJ, Richardson DC, Richardson JS, Terwilliger TC, Zwart PH. 2010. PHENIX: a comprehensive Python-based system for macromolecular structure solution. Acta crystallographica. Section D, Biological crystallography 66:213-221.

[0541] 59. Adams PD, Grosse-Kunstleve RW, Hung LW, Ioerger TR, McCoy AJ,

Moriarty NW, Read RJ, Sacchettini JC, Sauter NK, Terwilliger TC. 2002. PHENIX: building new software for automated crystallographic structure determination. Acta

Crystallogr. Sect. D-Biol. Crystallogr. 58: 1948-1954.

[0542] 60. Emsley P, Cowtan K. 2004. Coot: model-building tools for molecular graphics.

Acta Crystallogr. Sect. D-Biol. Crystallogr. 60:2126-2132. [0543] 61. Krissinel E, Henrick K. 2007. Inference of macromolecular assemblies from crystalline state. Journal of molecular biology 372:774-797.

[0544] 62. Winn MD, Ballard CC, Cowtan KD, Dodson EJ, Emsley P, Evans PR, Keegan RM, Krissinel EB, Leslie AG, McCoy A, McNicholas SJ, Murshudov GN, Pannu NS, Potterton EA, Powell HR, Read RJ, Vagin A, Wilson KS. 201 1. Overview of the CCP4 suite and current developments. Acta crystallographica. Section D, Biological

crystallography 67:235-242.

Example 13: Exemplary HIVxCD3 Bispecific Diabodies with Fc Domains

[0545] Provided herein are stable, structurally compact, bispecific diabodies that have interchain disulfide bonds that may be engineered with an Fc Domain to extend serum half-life. The antigen binding arms of such bispecific diabodies are advantageously selected to co- engage immune effector cells (e.g. , T cells, NK cells, etc. ) with antigen-expressing target cells (e.g. , HIV-1 infected cells) and activate and redirect the cytolytic activity of immune effector cell against the antigen expressing target cells. Provided herein are multispecific molecules which comprise fully human antigen binding portions of HIV-1 antibodies, and variants thereof, that specifically bind to and potently neutralize various isolates of HIV- 1. The HIV- 1 antibodies provided herein (e.g. , DH51 1, DH512, DH51 1.2 K3, DH513, DH514, DH515, DH516, DH517, DH518, DH536, or DH537) specifically bind the membrane-proximal extracelluar region (MPER) of gp41. Such antibodies are utilized in the generation of HIVxCD3 bispecific diabodies (and/or trivalent binding molecule as provided herein) having binding specificity for the MPER of gp41. Exemplary, HIVxCD3 bispecific diabodies, having three polypeptide chains, an HIV-1 binding arm derived from an anti-gp41 MPER antibody described herein, a CD3 effector cell binding arm derived from a humanized anti- CD3e mAb (hXRCD3), and CH2-CH3 IgGl Fc Domains may have the general structure shown in Figures 105A-105B.

[0546] Three exemplary HIVxCD3 bispecific diabodies with an Fc Domain designated "DH511.2 K3xCD3 Fc,' "DH511.11PxCD3 Fc,' and "DH511.12P Fc' were designed and were expressed and tested. These molecules comprise three polypeptide chains and include an HIV-1 binding arm derived from the DH511 2 K3, DH511.1 IP, or DH511.12P antibodies described herein, a CD3 effector cell binding arm derived from a humanized anti-CD3e mAb (hXRCD3), and CH2-CH3 IgGl Fc Domains. The amino acid sequences and a representative polynucleotide encoding each polypeptide chain are presented in Table 4. Corresponding negative control bispecific diabodies with an irrelevant binding arm [e.g. , aRSV derived from palivizumab, or afluorescein derived from 4-4-20] instead of the DH511.2 K3 HIV-1 arm, or having an HIV-1 arm [e.g., aHIV derived from DH511.2 K3] with an irrelevant binding arm [aRSV or afluorescein] instead of the CD3 arm may also be designed. In addition, a comparator bispecific diabody having an HIV-1 binding arm derived from an alternative anti- HIV-1 env antibody [e.g., the A32 antibody (Protein Data Bank (PBD) ID Code 3TNM)] instead of DH511.2 K3 may be designed. The HIVxCD3 Fc bispecific diabodies are capable of simultaneously binding to HIV-1 and CD3. The control bispecific diabodiesare capable of simultaneously binding to RSV (or any other non-HIV antigen) and CD3, or of simultaneously binding to HIV-1 and an irrelevant antigen. Each of the exemplary bispecific diabodies is a heterotrimer of polypeptide Chains 1, 2 and 3 have the general structure provided in Figure 103A (also see, e.g. , Figures 105A-105B). Chains 1 and 2 comprise the VH and VL Domains of DH511.2 K3, XRCD3, 4-4-20, palivizumab, or an alternative anti- HIV env antibody, as detailed above, while Chain 3 is common to other diabody molecules with this configuration.

[0547] Methods for forming bispecific diabodies, including such diabodies comprising an Fc Domain, are provided in WO 2006/113665, WO 2008/157379, WO 2010/080538, WO 2012/018687, WO 2012/162068, WO 2012/162067, WO 2014/159940, WO 2015/021089, WO 2015/026892 and WO 2015/026894.

[0548] Table 5 provides the SEQ ID NOs for each of the polypeptide chains present in DH511.2_K3xCD3 Fc molecule. Table 4 provides SEQ ID NOs for the VL and VH Domains of DH511.2 K3 (as provided herein), A32 (additional HIV-1 Env antibodies are known in the art, see, e.g. , WO 2016/054101), hXRCD3 (additional anti-CD3 antibodies are known in the art, see, e.g. , WO 2012/162067), 4-4-20, and palivizumab (additional anti-RSV antibodies are known in the art, see, e.g., US 6,818,216), the SEQ ID NOs for the VL and VH Domains of the exemplary anti-CD16 mAb h3G8, anti-CD8 mAb OKT8 and anti-CD8 mAb TRX2 are also provided. In addition, the SEQ ID NOs for representative nucleic acid encoding sequences are provided.

[0549] Chain 1 of the DH511.2_K3xCD3 Fc exemplary molecule comprises: an N- terminus, the VL Domain of DH511.2 K3 (SEQ ID NO: 553), a Peptide Linker 1 (SEQ ID NO: 508), the VH Domain of hXR32 (SEQ ID NO: 500), a Peptide Linker 2 (SEQ ID NO: 510), a cysteine engineered E-coil, a Spacer Linker 3 (SEQ ID NO: 522), a knob bearing CH2-CH3 (SEQ ID NO: 531), and a C-terminus. Chain 1 of DH511.11PxCD3 Fc is the same as for DH511.12PxCD3 Fc and differs from that of DH511.2_K3xCD3 Fc only in it comprises VL Domain DH511 5AVK (SEQ ID NO: 625). [0550] Chain 2 of the DH511.2_K3xCD3 Fc exemplary molecule comprises: an N- terminus, the VL Domain of hXR32 (SEQ ID NO: 503), a Peptide Linker 1 (SEQ ID NO: 508), the VH Domain of DH511.2 K3 (SEQ ID NO: 551), a Peptide Linker 2 (SEQ ID NO: 510), a cysteine engineered K-coil, a Spacer Linker 3 (SEQ ID NO: 521), and a C-terminus. Chain 2 of DH511.11PxCD3 Fc differs from that of DH511.2_K3xCD3 Fc only in that it comprises VH Domain DH511 5AVH-4A (SEQ ID NO: 624). Chain 2 of DH511.12PxCD3 Fc differs from that of DH511.2_K3xCD3 Fc only in that it comprises VH Domain DH511 5BVH-4A (SEQ ID NO: 626).

[0551] Chain 3 of the DH511.2_K3xCD3 Fc exemplary molecule comprises: an N- terminus, a Peptide Linker 3 (SEQ ID NO: 523), a hole bearing CH2-CH3 (SEQ ID NO: 533), and a C-terminus. Such bispecific diabodies, may be generated using the methods as provided above. This Chain 3 is common to each of DH511.2_K3xCD3 Fc, DH511.11PxCD3 Fc, and DH511.12P Fc

[0552] For additional sequences related to this example see Table 4.

[0553] The exemplary bispecific molecules were expressed and tested in an MPER peptide binding ELISA essentially as described above to confirm HIV antigen binding activity. The exemplary bispecific binding molecules were also tested in sCD3 binding ELISA to confirm CD3 binding activity. Briefly, soluble CD3 (1 μg/mL) was coated onto a solid support, and then incubated with the exemplary bispecific molecules (at a starting concentration of 2 nM, followed by 1 :4 serial dilutions). At the conclusion of the assay, binding was blocked with phosphate buffered saline (PBS) containing bovine serum albumin (BSA). Binding was detected by ELISA using anti-human IgG-that had been conjugated to horseradish peroxidase (HRP). All the molecules were found to bound to both the MPER peptide and sCD3.

[0554] DH511.11PxCD3 Fc and DH511.2_K3xCD3 Fc were also tested in a cytotoxic T lymphocyte (CTL) assay. Briefly, Jurkat 522F/Y GF HIV-1 Env-expressing target cells were treated with serial dilutions of the exemplary bispecific molecules, together with Pan T cells (isolated from healthy human PBMCs) at an E/T ratio of 10: 1, and incubated at 37°C for 48 hours. Cytotoxicity was measured by luminescence. Specific lysis is calculated from luminiscence counts (RLU): cytotoxicity (%) = 100 x (1-[RLU of Sample/RLU of Control]), where control equals the average RLU of target cells incubated with effector cells in the absence of a test article. Minimal or no activity was observed for these bispecific molecules in these assays under these conditions using Pan T cells from two different donors.

[0555] The cytotoxic activity of these and other molecules of the invention will be tested in a killing assay using HIV-1 infected cells expressing HIV- 1 envelope on their surface.

[0556] Without being bound by theory, other bispecific formats, for example but not limited to trivalent molecules having two HIV- 1 binding arms, are expected to have improved properties, including but not limited to improved activity in the CTL assay. Non-limiting embodiments of such trivalent molecules are described in Example 14B.

Example 14A: Additional multispecific molecules

[0557] Sequences from any one of the MPER antibodies contemplated by this disclosure, including but not limited to DH51 1, DH512, DH512 K3, DH513, DH514, DH515, DH516, DH517, DH518, DH536, DH537, lineage members, chimeric and mutant antibodies with non- limiting embodiments shown in Example 1 1 could be used to construct any one of the multispecific molecule formats described herein. The effector arm could target be any one of the non-limiting examples of CD2, CD3, CD8, CD 16, T-Cell Receptor (TCR), NKG2D, etc. epitopes. Non-limiting examples are provided in the Table below. The information in the specification can be readily used for alternative designs of the listed molecules, and for design of other bispecific molecules, for example molecules based on DH51 1, DH512, DH512 K3, DH513, DH514, DH515, DH516, DH517, DH518, DH536, DH537, lineage members chimeric and mutant antibodies with non-limiting embodiments shown in Example 1 1 lineage members using CDRs, or VH and VL chains from these HIV-1 antibodies.

[0558] Table 6 shows a summary of some additional non-limiting embodiments of bispecific diabody (with and without Fc Domains) that may be generated comprising alternative HIV-1 and/or alternative effector cell binding specificities. The information in specification can be readily used for alternative design of the listed bispecific diabodies, and for design of other bispecific diabodies, for example bispecific diabodies comprising a CD 16 binding specificity instead of CD3 and/or incorporating the HIV-1 binding specificity of alternative DH51 1 lineage members, DH512, DH512 K3, DH513, DH514, DH515, DH516, DH517, DH518, DH536, DH537 using CDRs, or VH and VL chains from these antibodies as provided herein.

[0559] Wherein SEQ ID NOs: 570, 574, 578, 582, 586, 590, 594, 598, 602, and 606 correspond to the VL chain of antibody DH511, DH512, DH513, DH514, DH515, DH516, DH517, DH518, DH536, and DH537, respectively and as described in Table 4. Wherein SEQ ID NOs: 553, and 625 correspond to the VL chain of antibody DH511 2 K3, and DH51 l_5a/5b, respectively and as described in Table 4.

[0560] Wherein SEQ ID NOs: 568, 572, 576, 580, 584, 588, 592, 596, 600, 604, 608-623 correspond to the VH chain of antibody DH511, DH512, DH513, DH514, DH515, DH516, DH517, DH518, DH536, DH537, chimeras and mutants as described in Example 11, respectively and as described in Table 4. Wherein SEQ ID NOs: 551, 624, and 626 correspond to the VH chain of antibody DH511.2 K3, DH51 l_5a, and DH51 l_5b, respectively and as described in Table 4.

Example 14B: Exemplary Trivalent and Control Molecules with Fc Domains

[0561] As provided herein trivalent binding molecules having three Antigen-Binding Domains provide additional functionality as they can co-engage multiple epitopes present on the surface of an effector cell, such as a T lymphocyte (e.g. , CD3 and CD8), or they can bind multiple epitopes of HIV-1 (e.g., epitopes of different HIV-1 isolates or different epitopes of HIV-1 Env, or the same epitope on multiple HIV-1 proteins). The antigen binding arms of such trivalent binding molecules are advantageously selected to co-engage immune effector cells (e.g. , T cells, NK cells, etc.) with antigen-expressing target cells (e.g., HIV-1 infected cells) and activate and redirect the cytolytic activity of immune effector cell against the antigen expressing target cells. [0562] Table 7 shows a summary of some non-limiting embodiments of trivalent binding molecules (having 3 or 4 polypeptide chain) that may be generated. The information in specification can be readily used for alternative design of the listed trivalent binding molecules, and for design of other trivalent binding molecules, for example trivalent binding molecules comprising alternative CD 8 binding specificities and/or incorporating the HIV-1 binding specificity of additional and/or alternative DH511 lineage members, DH512, DH512 K3, DH513, DH514, DH515, DH516, DH517, DH518, DH536, or DH537 using CDRs, or VH and VL chains from these antibodies.

[0563] Wherein SEQ ID NOs:570, 574, 578, 582, 586, 590, 594, 598, 602, and 606 correspond to the VL chain of antibody DH511, DH512, DH513, DH514, DH515, DH516, DH517, DH518, DH536, and DH537, respectively and as described in Table 4. Wherein SEQ ID NOs: 553, and 625 correspond to the VL chain of antibody DH511 2 K3, and DH51 l_5a/5b, respectively and as described in Table 4.

[0564] Wherein SEQ ID NOs:568, 572, 576, 580, 584, 588, 592, 596, 600, 604, 608-623 correspond to the VH chain of antibody DH511, DH512, DH513, DH514, DH515, DH516, DH517, DH518, DH536, DH537, chimeric and mutant variants as described in Example 11, respectively and as described in Table 4. Wherein SEQ ID NOs: 551, 624, and 626 correspond to the VH chain of antibody DH511.2 K3, DH51 l_5a, and DH51 l_5b, respectively and as described in Table 4.

[0565] Table 4 provides the amino acid sequences of one such exemplary trivalent binding molecule comprising antigen binding sites for HIV-1, CD3 and CD8 (HIVxCD3xCD8), and having four polypeptide chains which may be generated are provided in SEQ ID NOs: 555, 557, 561, and 562. Chain 1 of this exemplary molecule comprises: an N-terminus, the VL Domain of DH511.2_K3 (SEQ ID NO: 553), a Peptide Linker 1 (SEQ ID NO: 508), the VH Domain of hXR32 (SEQ ID NO: 500), a Peptide Linker 2 (SEQ ID NO: 510), a cysteine engineered E-coil, a Spacer Linker 3 (SEQ ID NO: 522), a knob bearing CH2-CH3 (SEQ ID NO: 531), and a C-terminus. Chain 2 of this exemplary molecule comprises: an N-terminus, the VL Domain of hXR32 (SEQ ID NO: 503), a Peptide Linker 1 (SEQ ID NO: 508), the VH Domain of DH511.2_K3 (SEQ ID NO: 551), a Peptide Linker 2 (SEQ ID NO: 510), a cysteine engineered K-coil, a Spacer Linker 3 (SEQ ID NO: 521), and a C-terminus. Chain 3 of this exemplary molecule comprises: an N-terminus, the VH Domain of OKT8 (SEQ ID NO: 543), a CHI-Hinge Domain (SEQ ID NO: 515), a hole bearing CH2-CH3 (SEQ ID NO: 533), and a C-terminus. Chain 4 of this exemplary molecule comprises: an N-terminus, the VL Domain of OKT8 (SEQ ID NO: 545), a CL Kappa Domain (SEQ ID NO: 516), and a C- terminus. It will be noted that polypeptide chains 1 and 2 of such an exemplary trivalent binding molecule are the same as those present in the bispecific Fc bearing diabody

DH511.2_K3xCD3 Fc described above.

[0566] Table 4 provides the amino acid sequences of another such exemplary trivalent binding molecule comprising antigen binding sites for HIV-1, CD3 and CD8

(HIVxCD3xCD8), and having three polypeptide chain which may be generated are provided in SEQ ID NOs: 555, 557, and 563. Chain 1 of this exemplary molecule comprises: an N- terminus, the VL Domain of DH511 2 K3 (SEQ ID NO: 553), a Peptide Linker 1 (SEQ ID NO: 508), the VH Domain of hXR32 (SEQ ID NO: 500), a Peptide Linker 2 (SEQ ID NO: 510), a cysteine engineered E-coil, a Spacer Linker 3 (SEQ ID NO: 522), a knob bearing CH2-CH3 (SEQ ID NO: 531), and a C-terminus. Chain 2 of this exemplary molecule comprises: an N-terminus, the VL Domain of hXR32 (SEQ ID NO: 503), a Peptide Linker 1 (SEQ ID NO: 508), the VH Domain of DH511 2 K3 (SEQ ID NO: 551), a Peptide Linker 2 (SEQ ID NO: 510), a cysteine engineered K-coil, a Spacer Linker 3 (SEQ ID NO: 521), and a C-terminus. Chain 3 of this exemplary molecule comprises: an N-terminus, the VL Domain of OKT8 (SEQ ID NO: 545), a Peptide Linker 5 (SEQ ID NO: 526), the VH Domain of OKT8 (SEQ ID NO: 543), a CHI-Hinge Domain (SEQ ID NO: 515), a hole bearing CH2- CH3 (SEQ ID NO: 533), and a C-terminus. It will be noted that polypeptide chains 1 and 2 of such an exemplary trivalent binding molecule are the same as those present in the bispecific Fc bearing diabody DH511 2_K3xCD3 Fc described above.

[0567] Exemplary trivalent binding molecules having three or four polypeptide chains, comprising two HIV-1 Env binding sites and one CD3 binding site are readily generated, for example by replace the VL and VH Domains of the anti-CD antibody in the above described molecules with the VL and VH Domains of an anti-HIV-1 Env antibody provided herein (e.g., DH511.2 K3) or known in the art. As provided above the HIV-1 Env binding arms may comprise the VL and VH Domains of the same or different anti-HIV-1 Env antibodies.

[0568] Table 4 provides the amino acid sequences of one exemplary trivalent binding molecules comprising two HIV-1 Env binding arms having the same VH and VL Domains, and one CD3 binding arm (HIVxCD3xHIV), and having four polypeptide chains which may be generated are provided in SEQ ID NOs: 555, 557, 627, and 628. Chain 1 of this exemplary trispecific molecule comprises: an N-terminus, the VL Domain of DH511 2 K3 (SEQ ID NO: 553), a Peptide Linker 1 (SEQ ID NO: 508), the VH Domain of hXR32 (SEQ ID NO: 500), a Peptide Linker 2 (SEQ ID NO: 510), a cysteine engineered E-coil, a Spacer Linker 3 (SEQ ID NO: 522), a knob bearing CH2-CH3 (SEQ ID NO: 531), and a C-terminus. Chain 2 of this exemplary molecule comprises: an N-terminus, the VL Domain of hXR32 (SEQ ID NO: 503), a Peptide Linker 1 (SEQ ID NO: 508), the VH Domain of DH511.2 K3 (SEQ ID NO: 551), a Peptide Linker 2 (SEQ ID NO: 510), a cysteine engineered K-coil, a Spacer Linker 3 (SEQ ID NO: 521), and a C-terminus. Chain 3 of this exemplary molecule comprises: an N- terminus, the VH Domain of DH511.2 K3 (SEQ ID NO: 551), a CHI-Hinge Domain (SEQ ID NO: 515), a hole bearing CH2-CH3 (SEQ ID NO: 533), and a C-terminus. Chain 4 of this exemplary molecule comprises: an N-terminus, the VL Domain of DH511 2 K3 (SEQ ID NO: 553), a CL Kappa Domain (SEQ ID NO: 516), and a C-terminus. It will be noted that polypeptide chains 1 and 2 of such an exemplary trivalent binding molecule are the same as those present in the bispecific Fc bearing diabody DH511.2_K3xCD3 Fc described above.

[0569] Table 4 provides the amino acid sequences of another exemplary trivalent binding molecules comprising two HIV-1 Env binding arms having the same VH and VL Domains, and one CD3 binding arm (HIVxHIVxCD3), and having four polypeptide chains which may be generated are provided in SEQ ID NOs: 629, 630, 631 and 632. Chain 1 of this exemplary molecule comprises: an N-terminus, the VL Domain of DH511 2 K3 (SEQ ID NO: 553), a

Peptide Linker 1 (SEQ ID NO: 508), the VH Domain of DH511.2 K3 (SEQ ID NO: 551), a

Peptide Linker 2 (SEQ ID NO: 510), a cysteine engineered E-coil, a Spacer Linker 3 (SEQ ID

NO: 522), a knob bearing CH2-CH3 (SEQ ID NO: 531), and a C-terminus. Chain 2 of this exemplary molecule comprises: an N-terminus, the VL Domain of DH511 2 K3 (SEQ ID

NO: 553), a Peptide Linker 1 (SEQ ID NO: 508), the VH Domain of DH511 2 K3 (SEQ ID

NO: 551), a Peptide Linker 2 (SEQ ID NO: 510), a cysteine engineered K-coil, a Spacer

Linker 3 (SEQ ID NO: 521), and a C-terminus. Chain 3 of this exemplary molecule comprises: an N-terminus, the VH Domain of hXR32 (SEQ ID NO: 500), a CHI-Hinge

Domain (SEQ ID NO: 515), a hole bearing CH2-CH3 (SEQ ID NO: 533), and a C-terminus.

Chain 4 of this exemplary molecule comprises: an N-terminus, the VL Domain of hXR32

(SEQ ID NO: 503), a CL Kappa Domain (SEQ ID NO: 516), and a C-terminus. [0570] As described above, alternative trivalent binding molecules may be generated by replacing the VL and VH Domains of DH511 2 K3 with the VL and VH Domains of any of the anti-HIV-1 antibodies provided herein. In particular, the VL Domain of DH511 2 K3 (SEQ ID NO: 553) may be replaced with the VL Domain DH511 5AVK (SEQ ID NO: 625), and the VH Domain of DH511 2 K3 (SEQ ID NO: 551) may be replaced with the VH Domain DH511 5AVH-4A (SEQ ID NO: 624) or the VH Domain DH511 5BVH-4A (SEQ ID NO: 626).

[0571] The ability of such exemplary trivalent binding molecules to bind to HIV-1 Env, CD3, and/or CD8 may be evaluated using the methods describe above, or well known in the art (see, e.g. , WO2016/054101). The activity of such trivalent binding molecules to mediate redirected cell killing of target cells expressing HIV-1 Env may be examined using the cytotoxic T lymphocyte (CTL) assay described above, or similar assays known in the art (see, e.g., Sung, JAM et al. Dual -Affinity Re-Targeting proteins direct T cell-mediated cytolysis of latently HIV-1 -infected cells. J Clin Invest. 2015; 125(11):4077-4090;

and WO2016/054101).

Example 15: Combinations of multispecific antibodies

[0572] Multispecific antibodies with different HIV-1 specificity, e.g. but not limited toCD4 binding site and V3 glycan binding, could also be tested in combination.

[0573] Various combinations of HIV-1 multispecific antibodies to mediate redirected cell killing of target cells expressing HIV-1 Env may be examined using the cytotoxic T lymphocyte (CTL) assay described above, or similar assays known in the art (see, e.g. , Sung, JAM et al. Dual -Affinity Re-Targeting proteins direct T cell-mediated cytolysis of latently HIV-1 -infected cells. J Clin Invest. 2015; 125(l l):4077-4090; and WO2016/054101).

Combinations of multispecific molecules, either with different HIV-1 specificity and/or different effector cell specificity, will be tested whether they provide enhanced benefits.

Example 16: Prophetic examples [0574] All multispecific molecules described herein could be tested in any other suitable assay. For non-limiting examples of further studies and characterization See Sung et al. J Clin Invest. 2015;125(ll):4077-4090; Sloan DD, Lam C-YK, Irrinki A, Liu L, Tsai A, Pace CS, et al. (2015) Targeting HIV Reservoir in Infected CD4 T Cells by Dual-Affinity Re- Targeting Molecules (DARTs) that Bind HIV Envelope and Recruit Cytotoxic T Cells. PLoS Pathog 11(11): el005233. doi: 10.1371/journal.ppat.1005233

[0575] Non-limiting examples of these assays include: determination of binding properties of the multispecific molecules to HIV-1 envelope expressing cells and/or HIV-1 infected cells; determining whether the multispecific molecules of the invention induce redirected T-cell killing of cell lines expressing various Envelopes and concomitant T-cell activation;

determining whether multispecific molecules bind to the surface of HIV-1- infected CD4 + T cells and redirect CD8 + T-cells to kill HIV-1 infected CD4 + cells using lymphocytes from HIV-1 seronegative donors; determining whether multispecific molecules redirect CD8 + T- cells to clear HIV-1 (e.g. JR-CSF, or any other suitable HIV-1 type)-superinfected CD4 + cells using lymphocytes from patients on suppressive ART; determining whether multispecific molecules redirect T cells from HIV-1 -infected individuals on suppressive ART to clear virus from resting CD4 + T cells following induction of latent virus expression.

[0576] Methods and Reagents

[0577] Generation of Infectious Molecular Clones (IMCs). HIV-1 IMCs for any subtype B BaL, subtype AE CM235 and subtype C 1086. C were generated with the backbone derived from NHL4-3 isolate as previously described See Edmonds TG et al. Replication competent molecular clones of HIV-1 expressing Renilla luciferase facilitate the analysis of antibody inhibition in PBMC. Virology. 2010;408(1): 1-13; Adachi A et al. Production of acquired immunodeficiency syndrome-associated retrovirus in human and nonhuman cells transfected with an infectious molecular clone. J Virol. 1986;59(2):284-291 . All IMCs expressed the Renilla luciferase reporter gene and preserved all nine viral open reading frames. The Renilla luciferase reporter gene was expressed under the control of the HIV-1 Tat gene. Upon HIV-1 infection of CD4+ T cells, expression of Tat during HIV-1 replication will induce luciferase expression, which allows quantitation of infected cells by measuring relative luminescence units (RLU).

[0578] Cell Lines. Jurkat-522 F/Y GF cells, which constitutively express a fusion protein of Copepod Green Fluorescent Protein (copGFP) and Firefly Luciferase (System Biosciences), were generated at Macrogenics from Jurkat-522 F/Y cells by transduction and clone selection. HEK293-D371 cells, which have doxycycline-inducible expression of HIV- 1 CM244 (subtype AE) gp l40 or CH505 T/F envelope, were obtained from Dr. John Kappes

(University of Alabama at Birmingham).

[0579] Flow Cytometric Analysis of diabody or mAb Binding to Cells. Diabodies at a desired concentration, e.g. 4 μg/mL are incubated with 10 5 cells in 200 FACS buffer containing 10% human AB serum for 30 minutes at room temperature. After washing, cells are resuspended in 100 of 1 μg/mL biotin-conjugated mouse anti-EK antibody (recognizes the E/K heterodimerization region of diabody proteins and/or trivalent binding molecules), are mixed with 1 :500 diluted streptavidin-PE and incubated in the dark for 45 minutes at 2-8°C. Cells are washed, resuspended with FACS buffer, and analyzed with a BD Calibur flow cytometer and Flow Jo software (Tree Star, Ashland OR). Binding to IMC-infected CD4 + T cells from normal human donors is conducted as previously described (54) for the A32 and 7B2 mAbs, and with biotin-conjugated mouse anti-EK antibody and 1 :500 diluted streptavidin-PE for the HIVx4420 control diabodies.

[0580] The ability of DH51 1 2_K3xCD3 Fc (or any of the bispecific diabodies and/or trivalent binding molecules) to mediate redirected cell killing of HIV-1 IMC-infected CD4 + Cells may be examined using assays well known in the art (see, e.g. , Sung, JAM et al. Dual- Affinity Re-Targeting proteins direct T cell-mediated cytolysis of latently HIV-infected cells. J Clin Invest. 2015; 125( 1 1):4077-4090; and WO2016/054101). Briefly, resting PBMC from normal healthy HIV-1 seronegative donors are activated (e.g. , with anti-human CD3 and anti-human CD28 antibodies). A CD4+ enriched cell population is obtained by depletion of CD8+ T cells (e.g. , using commercially available magnetic beads) and is spinoculated in the presence of the luciferase-expressing IMC -representing an HIV-1 subtype (e.g. , AE (CM235); B (BaL); or C (1086. C)) and cultured for -72 hours. The CD4+-infected target cells are then incubated with resting CD8+ effector cells (which may be isolated by negative selection from autologous PBMC, using a commercial CD8+ T cell isolation kit) at different E/T ratios (e.g. , 33 : 1, 1 1 : 1, 3 : 1, and 0: 1) in the absence or presence of bispecific diabodies for 6-48 hours at concentrations ranging from 0.0001-1,000 ng/ml. Uninfected and infected target cells alone are included as additional controls. Each condition is tested in duplicate. After incubation, the percentage of specific lysis (%SL) of target cells is determined, for example by adding ViviRen Live Cell Substrate (Promega) and measuring the RLU on a luminometer. The %SL of target cells may be determined as described previously (Pollara J et al. HIV-1 Vaccine- Induced CI and V2 Env-Specific Antibodies Synergize for Increased Antiviral Activities. J Virol. 2014;88(14):7715-7726).

[0581] Redirected T-Cell Cytotoxicity Assay Against HIV-1 Env-expressing Cell Lines and Assessment of T-Cell Activation. Pan T cells are isolated from healthy human PBMCs with the Dynabeads® Untouched™ Human T Cells Kit (Invitrogen). HIV-1 Env expressing cell lines (1-4 xlO 5 cells/mL) are treated with serial dilutions of diabodies (or trivalent binding molecules), together with human T cells at an effectortarget (E:T) ratio = 10: 1, or otherwise at varying E:T ratios as indicated, and incubated at 37°C, 5% CO2 overnight. Cytotoxicity is measured by lactate dehydrogenase (LDH) release (CytoTox 96® Non-Radioactive

Cytotoxicity Assay, Promega) as described previously (32). With the Jurkat-522 F/Y GF cell line, cytotoxicity is also measured by luminescence using Luciferase-Glo substrate

(Promega). Specific lysis is calculated from luminiscence counts (RLU): cytotoxicity (%) = 100 x (1-(RLU of Sample ÷ RLU of Control)), where Control = average RLU of target cells incubated with effector cells in the absence of DART. Data are fit to a sigmoidal dose- response function to obtain 50% effective concentration (EC50) and percent maximum specific lysis values. T-cell activation is measured by FACS analysis after cells in the assay plate are labeled with CD8-FITC, CD4-APC, and CD25-PE antibodies (BD Biosciences), followed by cell collection by FACS Calibur flow cytometer equipped with acquisition software CellQuest Pro Version 5.2.1 (BD Biosciences). Data analysis is performed using Flow Jo software (Tree star, Inc).

[0582] Redirected T-Cell Cytotoxicity Assay Against HIV-1 IMC-Infected CD4 + Cells.

Cryopreserved resting PBMC from normal healthy HIV-1 seronegative donors are activated for 72 hours with anti -human CD3 (clone OKT3; eBioscience) and anti-human CD28 (clone

CD28.2; BD Pharmingen). Subsequently, a CD4 + enriched cell population (purity >92.3%; average±standard deviation 95.73±2.6%) is obtained by depletion of CD8 + T cells using magnetic beads (Miltenyi Biosciences), spinoculated in presence of the luciferase-expressing

IMC representing HIV-1 subtype AE (CM235), B (BaL) or C (1086.C) and cultured for 72 hours. CD4 + infected target cells are incubated with resting CD8 + effector cells (isolated by negative selection from autologous PBMC, CD8 + T cell Isolation Kit, Miltenyi Biosciences) at 33: 1, 11 : 1, 3: 1, and 0: 1 E:T ratios in the absence or presence of diabodies (and/or trivalent binding molecules) for 6-48 hours at concentration ranging from 1,000 to 0.0001 ng/mL. Uninfected and infected target cells alone are included as additional controls. Each condition is tested in duplicate. After incubation, ViviRen™ Live Cell Substrate (Promega) is added and RLU measured on a luminometer; percentage specific lysis (%SL) of target cells is determined as described previously. See Pollara J et al. HIV-1 Vaccine-Induced CI and V2 Env-Specific Antibodies Synergize for Increased Antiviral Activities. J Virol.

2014;88(14):7715-7726.

[0583] T-Cell Degranulation (CD 107) Assay. As described for the cytotoxicity assay with HIV-1 IMC-infected cells as targets, activated CD4 + cells infected with HIV-1 BaL IMC are plated with resting CD8 + effector cells at a 33: 1 E:T ratio in the absence or presence of Ing/mL diabodies and incubated for 6 hours. For the CD4 T cell degranulation, activated CD4 + T cells are either infected with JR-CSF and labeled with the viability (NFLl) and target specific (TFL4) markers utilized in an ADCC assay or added to targets as effectors at a 10: 1 ratio prior to addition of diabodies. Each condition is tested in duplicate. CD107 PE-Cy5 (clone H4A3; eBioscience) is titered and added during the last six hours of the incubation along with Monensin solution (BD GolgiStop). A panel of antibodies consisting of

LIVE/DEAD Aqua stain, anti-CD3 APC-H7 (clone SK7; BD Pharmingen), anti-CD4 BV605 (clone OKT4; Biolegend), anti-CD8 BV650 (clone RPA-T8; Biolegend) are used to detect CD107 + CD8 + T cells. After washing and fixation, samples are acquired on a custom made LSRII (BD Bioscience, San Jose, CA) within the next 24 hours. A minimum of 300,000 total viable events is acquired for each test. The analysis of the data is performed using the Flow-Jo software (Treestar, Ashland, OR).

[0584] T-Cell Viability and Activation Assays. CD8 + T cells and CD 8 depleted PBMCs obtained from HIV infected ART suppressed patients are plated at 5xl0 4 cells per well in 96 well plates with lOOng/mL of the indicated DART. Cells are cultured in 0.2mL of cIMDM media supplemented with 10% FBS, 1% Penicillin/Streptomycin and 5U/mL IL-2 for 7 days, and then are stained with the following antibodies: HLA-DR-PerCP (clone L243), CD25-PE (clone M-A251), CD8-FITC (clone HIT8a), CD8-PE (clone HIT8a), CD4-FITC (clone RPA- T4), and Annexin V-PE and 7-AAD (all BD biosciences, San Jose, CA).

[0585] Redirected T-Cell Viral Clearance Assay. CD8 + T-cells are isolated from PBMCs by positive selection (EasySep human CD8 + Selection Kit, Stem Cell). CD8-depleted PBMCs are first activated with 2μg/mL of PHA (Remel, Lenexa, KS) and 60U/mL of IL-2, and then infected by spinoculation at 1200xg for 90 minutes with either JR-CSF or autologous reservoir virus (AR) at an MOI of 0.01 AR virus is obtained from pooled supernatants of replicate wells from outgrowth assays of resting CD4+ T-cells for each patient performed. Fifty-thousand (5xl0 4 ) targets/well are co-cultured with CD8 + T cells in triplicate at the indicated E:T ratio in the absence or presence of 100 ng/mL of diabody in 0.2m of cIMDM media supplemented with 10% FBS, 1% Penicillin/Streptomycin and 5 U/mL IL-2. For experiments performed in the presence of antiretrovirals (ARVs), 24 hours after spinoculation cells are washed and ΙμΜ of raltegravir and 4μΜ of abacavir are added, and then diabodies and CD8 + T-cells are added to cultures. Supernatant is assayed on day 7 by p24 ELISA (ABL, Rockville, MD). Results are calculated as the log (p24 of infected target cells only control divided by p24 of the test condition).

[0586] Latency Clearance Assay (LCA). The reduction of virus recovery from CD4 + infected cells is assessed by a standard quantitative viral outgrowth assay using the resting CD4 + T cells of aviremic, ART-treated patients, following the addition of antiviral effector cells and/or molecules, as previously described. See Sung JA et al. Expanded Cytotoxic T-Cell Lymphocytes Target the Latent Hiv Reservoir. J Infect Dis . 2015;: 1-15. . In this case the LCA is used to model the ability of diabodies to clear virus emerging from the latent reservoir under clinically and pharmacologically relevant conditions. Resting CD4 + T-cells are isolated from a leukapheresis product as previously described (72) and exposed to PHA ^g/mL) and IL-2 (60U/mL) for 24 hours or vorinostat (VOR) (335nM, 6 hours) (Merck Research Laboratories), and plated at 0.5 to 1 x 10 6 cells/well in 12 to 36 replicate wells depending on the size of the reservoir. The VOR is then washed off and CD8s added at an E:T of 1 : 10 as well as 100 ng/mL of the indicated DART. Cells are co-cultured for 24 hours (unless specified otherwise) following which the diabody (and/or trivalent binding molecule) proteins are washed off and allogeneic CD 8 -depleted PBMCs from an HIV negative donor are added to amplify residual virus. Supernatant is assayed for the presence of p24 antigen on day 15 for each well. Results are calculated as % viral recovery [(# of positive wells/total number plated)xl00], normalized to a control in which no CD8 + T cells are added.

[0587] Additional animal studies will be conducted to evaluate toxicity, safety, PK and PD profiles, and efficacy in preventing or treating HIV infection of the multispecific molecules of the invention.

[0588] All documents and other information sources cited herein are hereby incorporated in their entirety by reference.