Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
FUSION PROTEINS FOR HIV THERAPY
Document Type and Number:
WIPO Patent Application WO/2012/065055
Kind Code:
A2
Abstract:
Disclosed herein are fusion antibodies created to provide both an antigen-binding site that targets the CD4 receptor and an antigen-binding site that targets the HIV envelope. The fusion antibodies disclosed herein provide improved potency and breadth against HIV as compared to monospecific antibodies, and additionally provide high barrier against viral resistance. Also disclosed are pharmaceutical formulations and therapeutic methods utilizing such fusion proteins.

Inventors:
HO DAVID (US)
HUANG YAOXING (US)
PACE CRAIG (US)
SONG RUIJIANG (US)
FANG QING (US)
Application Number:
PCT/US2011/060357
Publication Date:
May 18, 2012
Filing Date:
November 11, 2011
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
UNIV ROCKEFELLER (US)
HO DAVID (US)
HUANG YAOXING (US)
PACE CRAIG (US)
SONG RUIJIANG (US)
FANG QING (US)
International Classes:
C07K16/46; A61K39/42; A61P31/18
Foreign References:
JPH0768276B21995-07-26
JP2004518624A2004-06-24
Other References:
See references of EP 2638072A4
Attorney, Agent or Firm:
GROLZ, Edward W. (Scott Murphy & Presser,400 Garden City Plaza,Suite 30, Garden City New York, US)
Download PDF:
Claims:
WHAT IS CLAIMED IS:

1. A fusion antibody, comprising a first antigen-binding site which binds to the CD4 receptor, conjugated to a second antigen-binding site which binds to the HIV envelope.

2. The fusion antibody of claim 1, comprising an intact anti-CD4 antibody which provides said first antigen-binding site, conjugated to an antigen-binding fragment of an anti-HIV antibody which provides said second antigen-binding site.

3. The fusion antibody of claim 2, wherein said first antigen-binding site binds to the D2 domain of the CD4 receptor.

4. The fusion antibody of claim 3, wherein said anti-CD4 antibody is humanized or monkeynized.

5. The fusion antibody of claim 4, wherein said anti-CD4 antibody is ibalizumab or a monkeynized derivative thereof.

6. The fusion antibody of claim 2 wherein said anti-CD4 antibody is an affinity matured antibody that has a greater binding affinity to CD4 than ibalizumab.

7. The fusion antibody of claim 2, wherein said anti-HIV antibody is a broad and potent neutralizing antibody.

8. The fusion antibody of claim 7, wherein said anti-HIV antibody binds to an HIV envelope protein.

9. The fusion antibody of claim 8, wherein said anti-HIV antibody binds to a quaternary epitope on the HIV envelope trimer.

10. The fusion antibody of claim 9, wherein said quaternary epitope comprises the V1/V2 and V3 regions of the HIV envelope trimer.

11. The fusion antibody of claim 10, wherein said anti-HIV antibody is PG9.

12. The fusion antibody of claim 8, wherein said anti-HIV antibody binds to a CD4-induced epitope in the HIV gpl20 protein.

13. The fusion antibody of claim 12, wherein said anti-HIV antibody is m36.

14. The fusion antibody of claim 8, wherein said anti-HIV antibody binds to an epitope in the HIV envelope involved in binding to CD4.

15. The fusion antibody of claim 14, wherein anti-HIV antibody is VRC01.

16. The fusion antibody of claim 1, wherein the Fc region is the Fc fragment of human IgGl or IgG4 containing the LALA mutation.

17. The fusion antibody of claim 16, wherein the Fc region is additionally modified to include one or more mutations that improve recycling of the antibody.

18. A method of treating a subject infected with HIV comprising administering to the subject a therapeutically effective amount of the fusion protein according to any one of claims 1-17.

19. A method of preventing HIV infection in a subject in need thereof comprising administering to the subject a therapeutically effective amount of the fusion protein according to any one of claims 1-17.

20. A method of preventing a HIV-positive pregnant subject from transmitting the HIV virus to the child, comprising administering to the subject a therapeutically effective amount of the fusion protein according to any one of claims 1-17.

Description:
Fusion Proteins for HIV Therapy

CROSS REFERENCE TO RELATED APPLICATION

[0001] This application claims the benefit of priority from U.S. Provisional Application No. 61/413, 178, filed November 12, 2010, the entire content of which is incorporated by reference.

FIELD OF THE DISCLOSURE

[0002] This disclosure generally relates to fusion proteins comprised of an antigen-binding site that binds to an epitope of the CD4 receptor and another antigen-binding site that binds an epitope of HIV. The fusion proteins are useful for treating HIV infection in a subject, decreasing the viral load of HIV in a patient, and/or preventing the transmission of HIV to subjects.

BACKGROUND ART

[0003] HrV-1 entry is triggered by interaction of the viral envelope (Env) glycoprotein gpl20 with domain 1 (D l) of the T-cell receptor CD4. Binding of CD4 by gpl 20 induces extensive conformational changes in gpl20 leading to formation and exposure of a structure called the co-receptor (coR) binding site, also known as the CD4-induced (CD4i) epitope, in the gpl20 protein (Moore et al., Proc Natl Acad Sci USA 100(19): 10598-602, 2003). The bridging sheet of gpl20 is a critical component of the coR binding site that is highly conserved across genetically diverse HIV- 1 isolates from different clades (Huang et al., Proc Natl Acad Sci USA 101 (9):2706-1 1 , 2004). The coR binding site is typically unformed on free (non-CD4-bound) gpl20. It forms after attachment of viruses to target cells through CD4 binding. The bridging sheet is highly immunogenic and elicits a class of antibodies known as CD4-induced (CD4i) antibodies in vivo. However, access of full-size Abs to the CD4i epitope (bridging sheet) is sterically restricted during viral entry into cells, most likely because the large size of an Ab cannot access the tight crypt within gpl20 where the bridging sheet resides. Thus, most known full-size CD4i Abs do not have potent antiviral activity. Fragments of CD4i Abs that are smaller in size could potentially gain access to the CD4i epitope during viral entry. For example two Ab fragments, known as Fab and scFv, of known CD4i Abs have been shown to significantly inhibit HIV entry more potently than full-size Abs (Labrijn et al., J. Virol. 77: 10557-10565, 2003).

[0004] Ibalizumab (iMab) is a potent and broad HIV-1 neutralizing Ab (Jacobson et al., Antimicrob. Agents Chemother. 53:450-457, 2009; Kuritzkes et al., J. Infect. Dis. 189:286-291 , 2004). iMab neutralizes HIV by binding mainly to domain 2 (D2) of the CD4 receptor on host T-cells, thus blocking the ability of HIV to use these CD4 receptors to gain entry into T-cells and produce infection (Burkly et al., J. Immunol. 149: 1779- 178, 1992). In a large panel of primary isolates (more than 100 viruses) tested recently, iMab neutralized 92% of all viruses as defined by 50% inhibition of infection, and 47.4% of viruses as defined by 90% inhibition of infection (Figure 1). These data, while promising, indicate that there is still a need to further improve the potency and breadth of iMab.

[0005] Several antibodies have been reported that target epitopes on HIV Env. m36 polypeptide is a human heavy chain domain Ab fragment that targets the highly conserved, but sterically restricted, CD4i epitope on HIV Env. m36 is a potent and broad cross-reactive HIV- 1 inhibitor with CD4i Ab activity in vitro, with a mean IC50 in the hundred nanomolar range (Chen et al., Proc Natl Acad Sci USA 105(44): 17121-6, 2008). However, similar to other small-size antibody fragments, the m36 polypeptide is predicted to have a short half life in circulation. A long half life is an important indicator for long-lasting antiviral activity. PG9 is another potent and broad anti-HIV-1 antibody, targeting a quaternary epitope of the HIV envelope trimer (Walker et al., Science 326, 285-289 (2009)). VRCOl is also a potent and broad anti-HIV-1 antibody, targeting a CD4 binding site epitope on the HIV envelope.

SUMMARY OF THE DISCLOSURE

[0006] Disclosed herein are fusion antibodies created to provide both an antigen-binding site that targets the CD4 receptor and an antigen-binding site that targets the HIV envelope. The fusion antibodies disclosed herein provide improved potency and breadth against HIV as compared to monospecific antibodies, and additionally provide high barrier against viral resistance. [0007] In some embodiments, the fusion proteins assume the configuration of an intact IgG molecule directed to a first antigen (which can be any isotype, i.e., IgGi, IgG 2 , IgG 3 , and IgG 4 ), connected via a linker at either the C or N terminus of its heavy or light chain to an antigen- binding domain of a second antibody directed to a second antigen.

[0008] In specific embodiments, the fusion protein is composed of an anti-CD4 antibody conjugated to an antigen-binding domain of an anti-HIV antibody. Examples of such fusion antibodies include Ibalizumab conjugated to m36, PG9scFv, or VRCOlscFv.

[0009] In additional embodiments, the anti-CD4 antibody portion of a bispecific fusion protein has been modified and fusion variants with improved affinity for CD4 have been selected by affinity maturation.

[0010] In other embodiments, the Fc region of a fusion protein has been engineered to provide a better PK profile, including improved stability (e.g., via introduced LALA mutations) and improved recycling capability.

[0011] Pharmaceutical formulations and therapeutic methods utilizing the bispecific fusion proteins disclosed herein are also provided.

BRIEF DESCRIPTION OF THE DRAWINGS

[0012] This patent or application file contains drawings executed in color. Copies of this patent or patent application publication with color drawings will be provided by the Office upon request and payment of the necessary fee.

[0013] Figure 1. Ibalizumab neutralization profile on a diverse panel of Env-pseudotyped viruses.

[0014] Figure 2. Schematic representation of the iMab-m36 fusion construct. Orange, Yellow, and Blue represent the Variable Heavy, Variable Light, and Constant chains of ibalizumab, respectively. Red represents the m36 polypeptide. [0015] Figure 3. SDS-PAGE showing expression of purified m36, iMab36 fusion, and iMab. Left-most lane contains a size marker.

[0016] Figure 4. iMabm36 binds sCD4. X-axis indicates increasing levels of the unlabeled constructs for iMab36 (pink), iMab (black), or m36 (green). Y-axis indicates the competitive level of binding of HRP-labeled ibalizumab to sCD4.

[0017] Figure 5. iMab36 was shown to be active against ibalizumab-sensitive and -resistant viruses. iMab-sensitive (left column) or iMab-resistant (right two columns) viruses were tested for their viral activity in the presence of the neutralizing constructs iMabm36 (pink), iMab (black), or m36 (green).

[0018] Figure 6. iMabm36 was shown to have greater potency against iMab-resistant viruses as compared to iMab alone.

[0019] Figure 7. Cell surface anchoring of m36 by fusing it to iMab was shown to improve its antiviral potency.

[0020] Figure 8. Sensitivity of iMabm36 was determined by the iMab-resistant virus sensitivity to m36.

[0021] Figures 9. Anti-ΗΓΥ activity of iMabm36 was shown to be dependent on its CD4 binding ability.

[0022] Figure 10. iMabm36 fusion was shown to be stable in vitro.

[0023] Figure 1 1. Amino acid sequence of an iMabm36 fusion peptide. (A) SEQ ED NO: 1 : composed of 1 -473 = MV1 heavy chain (SEQ ID NO: 2, the first 19 amino acid residues constituting a leader sequence), 474-489 = linker (SEQ ED NO: 3), and 490-606 = m36 variable heavy chain (SEQ ID NO: 4). (B) SEQ ED NO: 5: composed of residues 1 -19 = leader sequence (SEQ ED NO: 6), residues 20-131 = MV 1 variable light chain (SEQ ED NO: 7), and residues 132-238 = MV 1 constant light chain (SEQ ED NO: 8). [0024] Figure 12. Schematic representation of C-terminal and N-terminal ibalizumab-PG9 bispecific antibodies.

[0025] Figure 13. Ibalizumab-PG9 fusion antibody was shown to bind to human CD4 and gpl20 by ELISA.

[0026] Figure 14. Ibalizumab-PG9 fusion antibody was found to be active against ibalizumab- sensitive and -resistant viruses. Neutralization curves of ibalizumab (left column) and ibalizumab-PG9 (right column) against ibalizumab-resistant (top) and ibalizumab-sensitive (bottom) pseudoviruses. Each colored line represents the neutralization profile of an individual virus. Dashed black line indicates 50% inhibition of infection. Red line indicates the lowest IC50 observed with ibalizumab to highlight the potency of the ibalizumab-PG9 fusion antibody.

[0027] Figure 15. Amino acid sequence of a PG9-ibalizumab fusion peptide. (A) SEQ ID NO: 9: composed residues 1-19 (Purple) = Signal Peptide Sequence (SEQ ID NO: 10), residues 20- 155 (Black) = PG9 variable heavy chain sequence (SEQ ID NO: 1 1 ), residues 156-175 (Green) = Linker (SEQ ID NO: 12), residues 176-285 (Red) = PG9 variable light chain sequence (SEQ ID NO: 13), residues 286-301 (Blue) = Linker (SEQ ID NO: 14), residues 302-423 (Underlined Black = Ibalizumab variable heavy chain sequence (SEQ ID NO: 15), and residues 424-756 (Orange) = Fc sequence (SEQ ID NO: 16). (B) SEQ ID NO: 17: composed of residues 1 - 20 = signal peptide sequence (SEQ ID NO: 18), residues 21 - 133 = ibalizumab variable light chain (SEQ ID NO: 19), and residues 134 - 236 = ibalizumab constant light chain (SEQ ID NO: 20).

[0028] Figure 16. Schematic representation of C-terminal and N-terminal ibalizumab- VRCOl bispecific antibodies.

[0029] Figure 17. Ibalizumab-VRCOl fusion antibody was shown to bind to human CD4 and gpl 20 by ELISA.

[0030] Figure 18. Ibalizumab-VRCOl fusion antibody was found to be active against ibalizumab-resistant viruses. Neutralization curves of ibalizumab (left column) and ibalizumab- VRCOl (right column) against ibalizumab-resistant pseudoviruses. Each colored line represents the neutralization profile of an individual virus. Dashed black line indicates 100% inhibition of infection.

[0031] Figure 19. Amino acid sequence of a VRCOl -ibalizumab fusion peptide. (A) SEQ ED NO: 21 : composed of residues 1 -121 (Black) = VRCOl variable heavy chain sequence (SEQ ED NO: 22), residues 122-141 (Green) = Linker (SEQ ID NO: 23), residues 142-245 (Red) = VRCO l variable light chain sequence (SEQ ED NO: 24), residues 246-255 (Blue) = Linker (SEQ ID NO: 23), residues 256-387 (Underlined Black) = Ibalizumab variable heavy chain sequence (SEQ ED NO: 15), and residues 388-720 (Orange) = Fc sequence (SEQ ED NO: 16). (B) SEQ ED NO: 17: composed of residues 1 - 20 = signal peptide sequence (SEQ ED NO: 18), residues 21 - 133 = ibalizumab variable light chain (SEQ ED NO: 19), and residues 134 - 236 (ibalizumab constant light chain) (SEQ ED NO: 20).

[0032] Figure 20. HIV-neutralizing properties of ibalizumab and three bispecific fusion antibodies against a panel of 1 18 HEV isolates, as reflected by maximum percent inhibition (MPI in grey) and IC50 (in red).

[0033] Figure 21. Comparison of antiviral breadth and potency of select bispecific constructs versus their parental mAbs.

[0034] Figure 22. Enhanced potency of the PG9-iMab fusion construct in the right-most column. Red bars indicate median + interquartile IC50 range of all viruses tested.

[0035] Figure 23. Stability of ibalizumab-GlLALA in monkeys was shown to be superior to the original ibalizumab.

[0036] Figure 24. Improved PK profile for ibalizumab 428/434 variant in mice transgenic for human FcRn. Ab refers to IgGl -LALA version of ibalizumab; Tg signifies mice transgenic for human FcRn, whereas Wt signifies wild-type mice.

[0037] Figure 25. (A) 5A8 (yellow), the murine progenitor of ibalizumab, was humanized with human-specific sequences (green) to create ibalizumab (iMab), which was then "monkeynized" with rhesus-specific sequences (blue) in two steps to yield RhiMab. (B) Using RhiMab, rhesus bispecific antibodies were constructed using (B) the scFv fusion approach. Note that the human residues in the additional antigen-binding Fv will also be replaced by their rhesus counterparts.

[0038] Figures 26. (A) The amino acid sequences of the light chain (SEQ ID NO: 25) and heavy chain (SEQ ID NO: 26) of Rhesus Ibalizumab (RhiMab). (B) The amino acid sequence of Rhesus PG9 scFv (SEQ ID NO: 27): residues 1-137 = heavy chain (SEQ ID NO: 28), residues 138-157 = linker (SEQ ID NO: 12), and residues 158-267 = light chain (SEQ ED NO: 29).

[0039] Figure 27. Schematic summary of the method used to conduct "in vitro affinity maturation." Example shown here is for mutations directed to CDR-H1 , but a similar approach was taken for other CDRs.

DETAILED DESCRIPTION

[0040] It has been identified in accordance with the present invention that fusion antibodies created to include both an antigen-binding site that targets the CD4 receptor (such as domain 2 of the CD4 receptor) and an antigen-binding site that targets the HIV envelope (such as glycoprotein gpl20 or gp41) provide improved potency and breadth against HIV as compared to monospecific antibodies. Such fusion antibodies also provide high barrier against viral resistance. The basic structure of the fusion proteins disclosed herein is typically a whole IgG molecule directed to a first antigen, connected via a linker at either the C or N terminus of the heavy or light chain to an antigen-binding domain or a single-chain antibody (scFv) directed to a second antigen. The features and applications of these bispecific fusion antibodies are described in details below.

[0041] Definitions

[0042] An "antigen" refers to a molecule which contains one or more epitopes and which is capable of eliciting an immunological response. "Antigenic molecules" are also used in a general sense to refer to molecules that are binding targets of the fusion proteins disclosed herein.

[0043] An "epitope", also known as antigenic determinant, is the portion of an antigenic molecule or molecules that is recognized by the immune system, i.e., B cells, T cells or antibodies. An epitope can be a conformational epitope or a linear epitope. A conformational epitope is formed by discontinuous sections of an antigenic molecule, or formed by multiple molecules. In the case where the antigen is a protein, a conformational epitope can be formed by discontinuous amino acid residues of the same protein molecule, or by amino acid residues on different molecules of the protein (e.g., a quaternary epitope formed by a multimer of the protein). A linear epitope is formed by continuous sections of an antigen, e.g., a continuous sequence of. amino acids of a protein antigen.

[0044] The term "antibody" is used herein broadly and encompasses intact antibody molecules, which include intact polyclonal, monoclonal, monospecific, polyspecific, chimeric, humanized, human, primatized, single-chain, single-domain, synthetic and recombinant antibodies, and antibody fragments that have a desired activity or function.

[0045] The term "chimeric antibody" refers to antibodies containing polypeptides from different sources, e.g., different species or different antibody class or subclass. Examples of chimeric antibodies include an antigen-binding portion of a murine monoclonal antibody fused an Fc fragment of a human immunoglobulin. Methods for making chimeric antibodies are known in the art; for example, methods described in patents by U.S. Patent 4,816,397 to Boss et al. and U.S. Patent 4,816,567 to Cabilly et al.

[0046] The term "humanized antibody" refers to antibodies that contain non-human sequence elements in a human immunoglobulin backbone or framework. Generally, humanized antibodies are human immunoglobulins (recipient antibody) in which residues from a hypervariable region (CDRs) of the recipient are replaced by residues from a hypervariable region of a non-human species (donor antibody) such as mouse, rat, rabbit or nonhuman primate having a desired specificity, affinity and capacity. In some instances, framework region (FR) residues of the human immunoglobulin are also replaced by non-human residues. Humanized antibodies may also, in some instances, contain residues that are not found in either the recipient antibody or the donor antibody and introduced to further refine antibody performance. In general, a humanized antibody contains substantially all of at least one, and typically two, variable domains, in which all or substantially all of the hypervariable regions correspond to those of a non-human immunoglobulin and all or substantially all of the framework regions are those of a human immunoglobulin sequence. A humanized antibody optionally also contains at least a portion of an immunoglobulin constant region (Fc), typically that of a human immunoglobulin. Methods for making humanized antibodies are documented in the art; see, for example, by U.S. Patent 5,225,539 to Winter and U.S. Patent 4,816,397 to Boss et al.

[0047] The term "primatized antibody" refers to antibodies that contain non-primate sequence elements in a primate immunoglobulin backbone or framework. For example, primatized antibodies can be made from a primate immunoglobulin (recipient antibody) by replacing residues in a hypervariable region (CDRs) of the recipient antibody with residues from a hypervariable region of a donor antibody from a non-primate species such as mouse, rat, rabbit or nonhuman primate having a desired specificity, affinity and capacity. Alternatively, primatized antibodies can be made suitable for administration to a desirable primate species by using a recipient immunoglobulin having non-primate sequences or sequences from a different primate species and introducing the Fc fragment, and/or residues, including particularly framework region residues, from the desirable primate, into the recipient immunoglobulin. ^ Examples of primatized antibodies include "monkeynized" antibodies disclosed herein in the Examples section.

[0048] The term "monospecific antibody" refers to antibodies that recognize and bind to one epitope.

[0049] The term "polyspecific antibody" refers to antibodies formed from at least two separate antibodies and binding to multiple (i.e., two or more) separate epitopes.

[0050] As described above, the term "antibody" also includes fragments of an intact antibody, or "antibody fragments", including particularly antigen-binding fragments of an intact antibody. Examples of antigen-binding fragments include, but are not limited to, Fab fragments

(consisting of the VL, VH, CL and CH I domains), Fab' fragments (which differs from Fab fragments by having an additional few residues at the C-terminus of the CHI domain including one or more cysteines from the antibody hinge region), (Fab') 2 fragments (formed by two Fab' fragments linked by a disulphide bridge at the hinge region), Fd fragments (consisting of the VH and CHI domains), Fv fragments (referring to a dimer of one heavy and one light chain variable domain in tight, non-covalent association which contains a complete antigen recognition and binding site), dAb fragments (consisting of a VH domain), single domain fragments (VH domain, VL domain, VHH domain, or VNAR domain), isolated CDR regions, scFv (or "single chain Fv", referring to a fusion of the VL and VH domains, linked together via a linker), and other antibody fragments that retain antigen-binding function.

[0051] The term "CDR" or "complementarity determining region" refers to the hypervariable regions within the variable domain of an antibody. There are 3 CDRs in each of the heavy chain and light chain variable domains, and are composed of amino acid residues responsible for antigen-binding. The term "framework region" or "FR" refers to the more conserved portions of the variable domains and is composed of residues other than the hypervariable region residues.

[0052] The term "antigen-binding site" of an antibody means a conformation and/or configuration formed by amino acids of the antibody to which an antigen binds. For example, the three CDRs of each of the VH and VL domains interact to define an antigen-binding site on the surface of the VH-VL dimer. Together, the six CDRs confer antigen-binding specificity to the antibody. It should be noted, however, a single variable domain (i.e., VH or VL) can also recognize and bind antigen, albeit often less effectively than the whole binding site with all six CDRs.

[0053] The term "neutralizing antibody" refers to an antibody that inhibits, reduces or completely prevents ΗΓΥ-1 infection. Whether an antibody is a neutralizing antibody can be determined by in vitro assays described in the Examples section hereinbelow. [0054] The term "potent neutralizing antibody" refers to an antibody which, when used at a low concentration, reduces HIV-1 infection by at least 50%, 60%, 70%, 80%, 90%, 95%, 99% or greater. Concentrations below 50μg/ml, between 1 and 50μg/ml, or even below 1 μg/ml, are considered "low concentrations". In some embodiments, low concentrations are concentrations in the picomolar range, such as 10-900 ng/ml, and include any concentration in that range, such as 800, 700, 600, 500, 400, 300, 200, 100, 75, 50, 25, 10 ng/ml, or even less than 10 ng/ml.

[0055] The term "broad neutralizing antibody" refers to an antibody which inhibits HIV-1 infection, as defined by a 50% inhibition of infection in vitro, in more than 50%, 60%, 70%, 80%, 90%, 95%, 99% or greater, of a large panel of (greater than 100) HIV-1 envelope pseudotyped viruses and viral isolates.

[0056] The term "fragment" as used herein refers to a physically contiguous portion of the primary structure of a biomolecule. In the case of proteins, a fragment may be defined by a contiguous portion of the amino acid sequence of a protein and may be at least 3-5 amino acids, at least 6-10 amino acids, at least 1 1 -15 amino acids, at least 16-24 amino acids, at least 25-30 amino acids, at least 30-45 amino acids and up to the full length of the protein minus a few amino acids. For example, the m36 fragment is 1 17 amino acids long. In the case of polynucleotides, a fragment is defined by a contiguous portion of the nucleic acid sequence of a polynucleotide and may be at least 9- 15 nucleotides, at least 15-30 nucleotides, at least 31-45 nucleotides, at least 46-74 nucleotides, at least 75-90 nucleotides, and at least 90-130 nucleotides. In some embodiments, fragments of biomolecules are immunogenic fragments.

[0057] A "fusion protein" refers to two or more peptides of different origins connected to each other via a linker or linkers. For example, a fusion protein can include a protein conjugated to an antibody. Other examples include, an antibody conjugated to a different antibody or an antibody conjugated to a Fab fragment. The Fab fragment can be conjugated to the N terminus or C terminus of the heavy or light chain of the antibody, or other regions within the antibody. The term fusion protein and fusion construct are used herein interchangeably.

[0058] A "peptide" is any compound formed by the linkage of two or more amino acids by amide (peptide) bonds, usually a polymer of alpha-amino acids in which the alpha-amino group of each amino acid residue (except the NH2 terminus) is linked to the alpha-carboxyl group of the next residue in a linear chain. The terms peptide, polypeptide and poly(amino acid) are used synonymously herein to refer to this class of compounds without restriction as to size, unless indicated to the contrary. Members of this class having a large size are also referred to as proteins and include antibodies.

[0059] The term "linker" refers to a chemical moiety that connects one peptide to another, e.g., one antibody to another. Linkers can also be used to attach antibodies to labels or solid substrates. A linker can include amino acids. Linkers can be straight or branched, saturated or unsaturated carbon chains. They can also include one or more heteroatoms within the chain or at the termini of the chains. By "heteroatoms" is meant atoms other than carbon which are chosen from the group comprising of oxygen, nitrogen, sulfur, phosphorus, boron and halogen. In specific embodiments, linkers are peptides. The use of a linker may or may not be advantageous or needed, depending on the specific antibody pairs. Methods and techniques for the attachment of a linker to an antibody are known in the art. For a treatise on this subject, the reader is referred to Bioconjugate Techniques, G. Hermanson, Academic Press, 1996.

[0060] Fusion Antibody Targeting Both CD4 and HIV

[0061] This disclosure is directed to fusion antibodies that contain an antigen-binding site that targets the CD4 receptor and another antigen-binding site that targets the HIV envelope.

Fusion antibodies having specificities towards simply two different antigens are also referred to herein as bispecific antibodies. Bispecific fusion antibodies can be modified to include additional antigen binding sites to provide other polyspecific antibodies.

[0062] Generally speaking, the structure of the fusion proteins disclosed herein is composed of an intact IgG molecule directed to a first antigen, connected via a linker at either the C or N terminus of its heavy chain or light chain to an antigen-binding fragment of a second antibody directed to a second antigen. For example, a fusion antibody having dual specificities towards the CD4 receptor and HIV can be formed by conjugating an intact anti-CD4 antibody with an antigen-binding fragment of anti-HIV antibody. Conversely, a fusion antibody having dual specificities towards the CD4 receptor and HIV can be formed by conjugating an intact anti- HIV antibody with an antigen-binding fragment of anti-CD4 antibody. In either scenario, a bispecific fusion antibody contains an antigen-binding site that binds to an epitope on the CD4 receptor and another antigen-binding site that binds to an epitope on the HIV envelope.

[0063] Anti-CD4 antibodies have been described in the art and can also be readily generated as the protein sequence of the CD4 receptor is available to those skilled in the art. As a member of the immunoglobulin superfamily, CD4 has four immunoglobulin domains (Dl to D4) that are located on the extracellular surface of the cell. CD4 uses its D l domain to interact with the p 2 -domain of MHC class Π molecules.

[0064] In some embodiments, antibodies directed principally to the second immunoglobulin- like domain (D2) (amino acid positions 98-180) of the CD4 receptor are used in forming the bispecific fusion antibodies herein. Antibodies directed to the D2 domain of CD4 have the desirable property of blocking HIV infection without interfering with immune functions mediated by interaction of CD4 with the major histocompatibility complex (MHC) class II molecules. In specific embodiments, the anti-CD4 antibody used in forming a bispecific fusion binds to an epitope located in the BC-loop of D2 near the D1 -D2 junction of the CD4 receptor (amino acids 121-1.27). In other embodiments, the anti-CD4 antibody binds to the FG-loop of D2 (amino acids 163-165) and part of Dl (amino acids 77-96). The anti-CD4 antibody used in forming a fusion protein can bind to one or more or all of these regions, i.e., Dl , D1 -D2 junction, D2, the BC or FG loop of D2, or any combination thereof. The above amino acid numbering corresponds to positions of the mature form of the receptor, not including the signal peptide. The amino acid sequence of the human CD4 receptor is available in GenBank under Accession No. AAA 16069.1 and_is also set forth in SEQ ED NO: 34, in which amino acids 1- 25 represent a signal peptide, amino acids 26-122 constitute Dl , and amino acids 123-205 constitute D2.

[0065] A specific example of an anti-CD4 antibody suitable for use in forming a bispecific fusion antibody is Ibalizumab (previously known as TNX-355, or hu5A8), which is a humanized, anti-CD4 monoclonal antibody. Ibalizumab potently blocks infection by a broad spectrum of HIV- 1 isolates and targets an epitope located in the BC-loop of D2 near the D1 -D2 junction of the CD4 receptor, without interfering with immune functions mediated by interaction of CD4 with the major histocompatibility complex (MHC) class II molecules.

[0066] Anti-HIV Env antibodies have also been described in the art and can also be readily generated by those skilled in the art. The env gene encodes a precursor protein, gpl60. During HIV reproduction, the endogenous enzymes of the host cell cleave gpl60 into gpl20 and gp41. Three gpl20 molecules form a trimer which is anchored to the viral membrane, or envelope, via non-covalent interactions with the transmembrane protein, gp41. Binding of CD4 by gpl20 induces extensive conformational changes in gpl20 leading to formation and exposure of a structure called the co-receptor (coR) binding site, also known as the CD4-induced (CD4i) epitope, in the gpl20 protein (Moore et al., Proc Natl Acad Sci USA 100(19): 10598-602, 2003). The bridging sheet of gp 120 is a critical component of the coR binding site that is highly conserved across genetically diverse HIV-1 isolates from different clades (Huang et al., Proc Natl Acad Sci USA 101 (9):2706- 1 1 , 2004). The coR binding site is typically unformed on free (non-CD4-bound) gpl20. It forms after attachment of viruses to target cells through CD4 binding. The bridging sheet is highly immunogenic and elicits a class of antibodies known as CD4-induced (CD4i) antibodies in vivo. However, access of full-size Abs to the CD4i epitope (bridging sheet) is sterically restricted during viral entry into cells, most likely because the large size of an Ab often cannot access the tight crypt within gpl20 where the bridging sheet resides. Fragments of CD4i Abs that are smaller in size could potentially gain access to the CD4i epitope during viral entry. For example, two Ab fragments, known as Fab and scFv, of known CD4i Abs have been shown to significantly inhibit HIV entry more potently than full- size Abs (Labrijn et al., J. Virol. 77: 10557- 10565, 2003), and such antibody fragments are suitable for use in forming a bispecific fusion antibody in accordance with this disclosure.

[0067] In specific embodiments, anti-HIV antibodies used herein in forming a bispecific fusion antibody are directed to an epitope on an envelope protein of HIV, e.g., gpl20 monomer or trimer, or gp41. In more specific embodiments, the anti-HIV antibodies used in forming a bispecific fusion may bind to the CD4 binding site on the HIV envelope at amino acids 279 - 460, the V I V2 and V3 regions of the HIV envelope trimer at amino acids 125-196 and 296- 331 , and/or the CD4 bridging sheet site on the HIV envelope at amino acids 332 - 444. Examples of anti-HIY antibodies suitable for use herein include, but are not limited to, m36, PG9, and VRC01. The amino acid sequence of HIV gpl60 is available in GenBank under Accession No. AAB60578. L and is also set forth in SEQ ID NO: 35, in which amino acids 1- 30 represent a signal peptide, and amino acids 31 -509 represent the mature gpl20.

[0068] In certain embodiments, a fusion antibody having dual specificities towards the CD4 receptor and HIV is composed of an intact anti-CD4 antibody, conjugated with (i.e., covalently linked to) an antigen-binding fragment of anti-HIV antibody. As defined above, an antigen- binding fragment of anti-HIV antibody including Fab fragments, Fab' fragments, (Fab') 2 fragments, Fd fragments, Fv fragments, dAb fragments, single domain fragments, isolated CDR regions, scFvs, and other antibody fragments that retain HIV -binding function of an anti-HIV antibody.

[0069] In a specific embodiment, a single domain antibody fragment of an anti-HIV antibody, e.g., m36, is fused to an intact anti-CD4 antibody. In some embodiments, the single domain antibody fragment is fused to the C-terminus of the heavy chain or light chain of the anti-CD4 antibody. In other embodiments, the single domain antibody fragment is fused to the N- terminus of the heavy or light chain of the anti-CD4 antibody.

[0070] In another specific embodiment, a single chain Fv fragment (i.e., scFv) of an anti-HIV antibody is fused to an intact anti-CD4 antibody. Similarly, the scFv fragment can be fused to the C-terminus or N-terminus of either the heavy chain or light chain of the anti-CD4 antibody. In some embodiments, the scFv fragment is a single chain of the VH domain linked to the N- terminus of the VL domain of a relevant anti-HIV antibody; while in other embodiments, the scFv fragment is a single chain of the VH domain linked to the C-terminus of the VL domain of a relevant anti-HIV antibody.

[0071] The linkage between the two antibody components, i.e., between the anti-CD4 antibody portion and the anti-HIV antibody portion, and also between the VH and VL domains of an scFv fragment, is achieved by a peptide linker. The length of a linker is generally in the range of 5 to 50 amino acids, and in specific embodiments, in the range of 9-25 amino acids, such as 9, 12, 16, 20 or 24 amino acids. The linkers can be synthetic or native human antibody-derived sequences, or a combination of both. Generally speaking, the linkers are principally composed of relatively small, neutral amino acids, such as Glycine, Serine, Alanine, and can include multiple copies of a sequence enriched in Glycine and Serine, such as multiple copies of GGGGS (SEQ ID NO: 33). Examples of linkers suitable for use herein include those set forth in SEQ ID NO: 3, 12, 14, 23 and 32.

[0072] The bispecific fusion antibodies disclosed herein can be produced by utilizing techniques available to those skilled in the art. For example, DNA molecules encoding a desirable bispecific fusion antibody can be constructed based on the coding sequence of the two antibody components of the fusion using molecular cloning techniques. The resulting DNAs can be placed into expression vectors which are then transfected into host cells such as E. coli cells, simian COS cells, Chinese Hamster Ovary (CHO) cells, human embryonic kidney 293 cells, or myeloma cells including murine myeloma cells that do not otherwise produce immunoglobulin protein, to obtain the synthesis of monoclonal antibodies in the recombinant host cells. For example, an expression vector encoding a heavy chain of an anti-CD4 antibody fused to an anti-HIV antibody fragment, and an expression vector encoding the light chain of the anti-CD4 antibody, can be co-transfected into a host cell for expression. Antibody- containing culture supernatants can be collected for purification of antibodies.

[0073] The produced fusion antibodies can be evaluated in in vitro assays to assess their functionality, e.g., binding to the CD4 receptor, binding to the HIV envelope, anti-HIV potency and breadth. In specific embodiments, this disclosure provides potent and broad bispecific fusion antibodies, with potency and breadth being defined as hereinabove. These antibodies are described in more details in the Examples section.

[0074] Modifications To Antibodies

[0075] Humanization and Primatization

[0076] In cases where the fusion antibody or the two antibodies forming the bispecific fusion antibody are non-human antibodies, the antibody can be "humanized" to reduce

immunogenicity to a human recipient. Methods for humanizing non-human antibodies have been described in the art. See, e.g., Jones et al., Nature 321 :522-525 (1986); Riechmann et al, Nature 332:323-327 (1988); Verhoeyen et al., Science 239: 1534-1536 (1988), and U.S. Patent 4,816,567. Generally, residues from the variable domain of a non-human antibody are "imported" into a human immunoglobulin molecule, resulting in antibodies in which some hypervariable region residues and possibly some FR residues of a human antibody are substituted by residues from analogous sites of non-human antibodies. It is important to humanize a non-human antibody while retaining high affinity for the antigen. To this end, three dimensional immunoglobulin models are commonly available and suitable for use in analyzing proposed humanized sequences in comparison to the parental non-human antibodies. Such analysis permits identification of residues likely involved in recognition and binding of the antigen, and therefore rational design of humanized sequences that retain the specificity and affinity for the antigen.

[0077] In specific embodiments, bispecific fusion antibodies are formed from an anti-CD4 antibody which has been humanized and an anti-HIV human or humanized antibody. For example, ibalizumab is an example of a humanized anti-CD4 antibody, and m36 is an example of the variable domain of a human anti-HIV antibody.

[0078] Similarly, a fusion antibody or the two antibodies forming the fusion can be

"primatized" to reduce immunogenicity to another primate, non-human recipient, e.g., a rhesus recipient. Residues from the variable domain of a donor antibody (such as a non-primate antibody or an antibody of a primate species different from the recipient primate) are

"imported" into a nonhuman primate recipient immunoglobulin molecule, resulting in antibodies in which some hypervariable region residues and possibly some FR residues of a nonhuman primate antibody are substituted by residues from analogous sites of donor antibodies. Alternatively, primatized antibodies can be made for use in a desirable primate species by using a recipient immunoglobulin having non-primate sequences or sequences from a different primate species by introducing the Fc fragment, and/or residues, including particularly framework region residues, from the desirable primate, into the recipient immunoglobulin. A "monkeynized" version of Ibalizumab, and a monkeynized version of anti- HIV antibody PG9 scFv, are disclosed in the Examples hereinbelow. [0079] Affinity Maturation

[0080] One or more hypervariable region residues of an antibody can be substituted to select for variants that have improved biological properties relative to the parent antibody by employing, e.g., affinity maturation using phage or yeast display. For example, the Fab region of an anti-CD4 antibody or an anti-HIV antibody can be mutated at several sites selected based on available structural information to generate all possible amino substitutions at each site. The antibody variants thus generated are displayed in a monovalent fashion from phage particles or on the surface of yeast cells. The displayed variants are then screened for their biological activity (e.g. binding affinity). Examples of ibalizumab variants having high affinities are described herein below. These ibalizumab variants can be used in forming bispecific fusion antibodies.

[0081] Modifications to the Fc Region

[0082] The fusion antibody or the intact antibody used in forming the fusion, which can be of any IgG isotype from any primate species including human, can be modified to improve certain biological properties.of the antibody, e.g., to improve stability, to enhance or reduce effector functions such as antigen-dependent cell-mediated cyotoxicity (ADCC) and/or complement dependent cytotoxicity (CDC) of the antibody, improved or decreased internalization and/or recycling, among others.

[0083] For example, the Fc fragment of ibalizumab (derived from human Ig4) can be replaced with human IgG l carrying the so-called LALA mutations that reduce effector function mediated through FcRs (except FcRn). Such modification has been shown herein to improve the stability of the resulting antibody by about 5 fold. The LALA mutation can also be introduced into an IgG4 background. In another example, the IgG l Fc fragment can be modified with the pair of mutations, M428L/N434S, to improve the recycling of the antibody via the antibody salvage pathway. This pair of mutations, introduced in the IgGl -LALA version of ibalizumab, has led to an additional about 3 fold improvement in PK of the resulting antibody. [0084] Still another type of modification involves alteration of the glycosylation pattern of a parent antibody, including deletions of one or more carbohydrate moieties found in the parent antibody, or addition of one or more carbohydrates (via addition of one or more glycosylation sites) that are not present in the parent antibody

[0085] Pharmaceutical Formulations

[0086] Pharmaceutical formulations of the fusion antibody proteins disclosed can be prepared by mixing a fusion protein with optional pharmaceutically acceptable carriers.

Pharmaceutically acceptable carriers include solvents, dispersion media, isotonic agents and the like. The carrier can be liquid, semi-solid, e.g. pastes, or solid carriers. Examples of carriers include water, saline solutions or other buffers (such as phosphate, citrate buffers), oil, alcohol, proteins (such as serum albumin, gelatin), carbohydrates (such as monosaccharides, disaccharides, and other carbohydrates including glucose, sucrose, trehalose, mannose, mannitol, sorbitol or dextrins), gel, lipids, liposomes, resins, porous matrices, binders, fillers, coatings, stabilizers, preservatives, antioxidants including ascorbic acid and methionine, chelating agents such as EDTA; salt forming counter-ions such as sodium; non-ionic surfactants such as TWEEN™' PLURONICS™ or polyethylene glycol (PEG), or combinations thereof.

[0087] The formulation can contain more than one active compound, e.g., one or more fusion antibodies, in combination with one or more additional beneficial compound for preventing and treating HIV infections.

[0088] The active ingredients can be combined with the carrier in any convenient and practical manner, e.g., by admixture, solution, suspension, emulsification, encapsulation, absorption and the like, and can be made in formulations such as tablets, capsules, powder (including lyophilized powder), syrup, suspensions that are suitable for injections, ingestions, infusion, or the like. Sustained-release preparations can also be prepared. [0089] Methods of Treatment and Prevention

[0090] In a further aspect, the bispecific fusion antibodies disclosed herein, optionally provided in pharmaceutically acceptable carrier, are employed for the treatment and prevention of HIV infection in a subject, as well as prevention of HIV transmission.

[0091] The term "treatment" of HIV infection refers to effective inhibition of the HIV infection so as to delay the onset, slow down the progression, reduce viral load, and/or ameliorate the symptoms caused by HIV infection.

[0092] The term "prevention" of HIV infection means the onset of HIV infection is delayed, and/or the incidence or likelihood of HIV infection is reduced or eliminated.

[0093] The term "prevention" of HIV transmission means the incidence or likelihood of HIV being transmitted from one individual to another (e.g., from an HIV-positive woman to the child during pregnancy, labor or delivery, or breastfeeding) is reduced or eliminated.

[0094] The term "subject" refers to any primate subject, including human and rhesus subjects.

[0095] To treat and/or prevent HIV infection, a therapeutic amount of a fusion antibody disclosed herein is administered to a subject in need.

[0096] The term "therapeutically effective amount" means the dose required to effect an inhibition of HIV infection so as to treat and/or prevent HIV infection. The dosage of a fusion antibody depends oii the disease state and other clinical factors, such as weight and condition of the subject, the subject's response to the therapy, the type of formulations and the route of administration. The precise dosage to be therapeutically effective and non-detrimental can be determined by those skilled in the art. As a general rule, a suitable dose of a fusion antibody for the administration to adult humans parenterally is in the range of about 0.1 to 20 mg/kg of patient body weight per day, once a week, or even once a month, with the typical initial range used being in the range of about 2 to 10 mg/kg. Since the antibodies will eventually be cleared from the bloodstream, re-administration may be required. Alternatively, implantation or injection of the fusion antibodies provided in a controlled release matrix can be employed. [0097] The fusion antibodies can be administered to the subject by standard routes, including the oral, transdermal, parenteral (e.g., intravenous, intraperitoneal, intradermal, subcutaneous or intramuscular). In addition, the antibodies can be introduced into the body, by injection or by surgical implantation or attachment such that a significant amount of a desirable antibody is able to enter blood stream in a controlled release fashion.

[0098] The description of some specific embodiments provides sufficient information that others can, by applying current knowledge, readily modify or adapt for various applications such specific embodiments without departing from the generic concept, and, therefore, such adaptations and modifications should and are intended to be comprehended within the meaning and range of equivalents of the disclosed embodiments. It is to be understood that the phraseology or terminology employed herein is for the purpose of description and not of limitation. In the drawings and the description, there have been disclosed exemplary embodiments and, although specific terms may have been employed, they are unless otherwise stated used in a generic and descriptive sense only and not for purposes of limitation, the scope of the claims therefore not being so limited. Moreover, one skilled in the art will appreciate that certain steps of the methods discussed herein may be sequenced in alternative order or steps may be combined. Therefore, it is intended that the appended claims not be limited to the particular embodiment disclosed herein.

[0099] The present description is further illustrated by the following examples, which should not be construed as limiting in any way. The contents of all cited references (including literature references, issued patents, and published patent applications as cited throughout this application) are hereby expressly incorporated by reference.

Example-1. Construction and Testing of iMabm36

[00100] This Example describes the construction and testing of a novel bispecific HIV-1 neutralizing antibody (Ab) for the prevention and treatment of HIV/AIDS which was named iMabm36. iMabm36 is composed of the anti-CD4 Ab ibalizumab (iMab) linked to two copies of the anti-CD4-induced Ab m36. iMabm36 is shown herein to have significantly increased antiviral potency and breadth over iMab and m36 alone. In particular, it can potently inhibit viral entry of all iMab-resistant viruses tested. Mechanistically, it is shown therein that iMabm36 activity requires CD4 binding and is m36 sensitivity dependent. The inter- dependency of this dual mechanism of action enables the high potency and breadth of iMabm36. As almost all HIV-l isolates use CD4 as a primary entry receptor and the m36 targeting site is highly conserved across all HIV-l isolates, targeting these two sites is believed to provide a high barrier against viral resistance. iMabm36 has been shown to be stable in vitro, and is believed to have improved pharmacokinetics as compared to m36 alone and is unlikely to be immunogenic.

[00101] Construction and expression of iMabm36 fusion Ab. A bi-specific fusion Ab was constructed based on a derivative IgG l version of iMab called MV 1 and m36. As shown in the schematic in Figure 2 and the sequence in Figure 1 1 , m36 was linked to the C-terminus of the heavy chain of MV1 via a flexible (G4S)x3 linker peptide (GGGGSGGGGSGGGGSG, SEQ ID NO: 3). In brief, cDNA sequence of the fusion construct was generated by overlap PCR and subsequently cloned into the pVAX expression plasmid through unique restriction sites.

[00102] The iMabm36 fusion Ab was expressed by transient co-transfection of pVAX vectors expressing 1 ) heavy chain iMabm36 fusion and 2) light chain ibalizumab into human embryonic kidney 293 cells. Ab-containing culture supematants were filtered and purified by affinity chromatography using a Protein A Sepharose column (Figure 3). The average yield was of culture medium.

[00103] iMabm36 retains CD4 binding activity in vitro. Purified iMabm36 was assessed for soluble CD4 (sCD4) binding in a competition ELISA assay. In brief, a 96-well plate was coated with sCD4. A fixed concentration of HRP labeled iMab was then mixed with increasing concentrations of iMabm36 or unlabeled iMab and measured for sCD4 binding competition. As shown in Figure 4, iMabm36 competed with iMab-HRP labeled Ab for CD4 binding with equal potency as compared to the parental unlabeled iMab, indicating the functional CD4 binding properties of the iMabm36 fusion protein. As expected, m36 alone did not bind to sCD4. [00104] iMabm36 fusion Ab improves antiviral potency and breadth as compared to iMab or m36 alone. To test whether the fusion of m36 to the C-terminal of iMab could result in more potent antiviral activity, iMabm36, iMab and m36 were each examined in a TZ -bl based HIV- 1 Env pseudotyped neutralization assay against a panel of 6 viruses that included iMab sensitive and resistant viruses. As shown in Figure 5, iMabm36 demonstrated a similar potency against iMab sensitive viruses (G 19 and G23) as compared to iMab alone. While iMab can only achieve a maximal percent of inhibition (MPI) of 25% to 75% for iMab resistant viruses (G02, G09, G20, and B09), the iMabm36 fusion could neutralize all four of these viruses and achieved 100% MPI at low nanomolar concentrations. Consistent with previous publication, the 50% inhibitory activity of m36 is within the range of hundreds of nanomolar concentrations.

[00105] In a separate experiment, iMabm36 activity was further examined against all iMab-resistant viruses in the inventors' collection (Figure 6). Among 13 iMab-resistant viruses tested (defined as a MPI of less than 100%), all could be neutralized by iMabm36 with 100% MPI, except for the virus G12 which was neutralized by up to 90% MPI. The mean concentration required to achieve 100% viral neutralization by iMabm36 is around 0.5pg/ml (2.8 nM). Also, iMabm36 was active in a PMBC based neutralization assay (data not shown).

[00106] Increased antiviral potency of iMabm36 compared to iMab and m36 mixture. The results of Figure 5 indicated that iMabm36 was more active than its individual components iMab and m36. To investigate if the high antiviral activity of iMabm36 could also be achieved by simply mixing iMab and m36, a neutralizing assay was performed using iMab and m36 at a molar ratio of 1 :2 because one iMab carries two m36 peptides in the iMabm36 fusion. As shown in Figure 7a, mixing iMab and m36 improved not only the antiviral activity of the respective components, but also achieved 100% neutralization of the iMab-resistant viruses tested. These results indicate that iMab and m36 contribute to the antiviral activity in an additive or synergistic manner. In comparison with the mixing of iMab and m36 components, the iMabm36 fusion Ab demonstrated even greater antiviral activity with at least 10-fold enhancement at the same molar concentrations tested. [00107] To test the hypothesis that iMabm36 acts through increasing m36 local concentration, iMab was mixed with an excess of m36 at a molar ratio of 1 : 10. As shown in Figure 7b, mixing iMab and m36 at a 1 : 10 ratio was more potent than that of the 1 :2 ratio in Figure 7a. Notably, even with a five times excess of m36, the mixture using individual components was still less potent than m36 fused to iMab.

[00108] Improved potency of the iMabm36 fusion against iMab-resistant viruses is conferred by viral sensitivity to m36. To better understand the contribution of m36 in the context of the iMab36 fusion, the inventors analyzed the correlation of ibalizumab-resistant virus sensitivity to m36 versus iMabm36. As shown in Figure 8, a strong correlation was observed between the IC50 of m36 and the IC50 of iMabm36. This result supports the notion that, in the context of iMab-resistant viruses, the sensitivity of iMabm36 is determined by the virus sensitivity to m36.

[00109] Antiviral potency of iMabm36 is dependent on its CD4 binding. To understand the mechanism of antiviral activity of the iMab36 fusion, two contact amino acid residues in the iMab heavy chain variable CDR3 region were substituted with alanine residues. The CDR3 regions of the wt and mutant heavy chains of iMab are set forth in Fig. 9a and SEQ ID NOS: 35 and 36 respectively. The mutant fusion Ab (iMabAm36) was purified and tested for its CD4 binding ability in vitro. As shown in Figure 9a, iMabAm36 could no longer compete with iMab-HRP for sCD4 binding, suggesting that these two mutations abolished the CD4 binding activity of iMabm36. In a virus neutralization assay, iMabAm36 lost its virus neutralization activity (Figure 9b). This result suggests that the antiviral activity of iMabm36 is dependent on its CD4 binding. In other words, anchoring of m36 via iMab binding to CD4 is critical for iMabm36 antiviral activity.

[00110] In vitro stability of iMabm36. To assess the stability of iMabm36, the inventors first generated high titers of rabbit anti-m36 immune serum. To do so, rabbits were immunized with purified m36 protein (300μg/dose) in CFA at week 0 and subsequently boosted in IFA twice at weeks 4 and 8. Anti-m36 Ab titers were determined in the serum sample collected 4 weeks post last boost immunization. The in vitro stability of the iMabm36 fusion Ab was determined by incubation of the fusion Ab in 20% serum in PBS at 37°C for up to 7 days. Aliquots of the untreated (day 0) and treated fusion Ab were taken at the indicated time points and stored at -20°C. The presence of intact iMabm36 was examined by the functional binding activity of iMabm36 to sCD4 and determined by anti-iMab Fc direct ELISA and anti-m36 sandwich ELISA, respectively.

[00111] Data from Figure 10 demonstrates that, over time, there was no loss of CD4 binding as detected by a secondary Ab against iMab Fc, which indicates that the iMab portion of this fusion was still present. The m36 portion of this fusion was also still present as detected by an anti-m36 rabbit immune serum and a secondary Ab against rabbit serum Ig. These results suggest that iMabm36 was stable for up to at least 7 days in the condition tested.

Example-2. Construction and Testing of Ibalizumab-PG9

[00112] This Example describes the construction and testing of Ibalizumab-PG9 (or also referred to as "PG9-Ibalizumab"), a novel bispecific HIV-1 neutralizing antibody (NAb) for the prevention and treatment of HIV/AIDS. Ibalizumab-PG9 was created as a fusion of the anti- CD4 Ab ibalizumab fused to a single-chain Fv (scFv) version of the anti-HIV envelope NAb, PG9. Ibalizumab-PG9 showed significantly increased antiviral potency and breadth over ibalizumab or PG9 alone. In particular, it potently inhibited viral entry of all ibalizumab- resistant viruses tested. Since ibalizumab and PG9 inhibit HIV entry using two distinct mechanisms, the chance of emerging HIV resistance to the ibalizumab-PG9 fusion is diminished in comparison to either ibalizumab or PG9 alone.

[00113] Construction and expression of the ibalizumab-PG9 fusion antibodies. To improve ibalizumab's breadth and potency, bispecific ibalizumab-PG9 was created to simultaneously target both the CD4 receptor on host T-cells and the HIV envelope protein gpl20. PG9 is currently one of the most potent and broadest anti-HIV- 1 antibodies, targeting a quaternary epitope of the HIV envelope trimer (Walker et al., Science 326, 285-289 (2009)). To create the fusion antibody, the inventors fused the heavy chain variable domain of PG9 to the light chain variable domain of PG9 via a flexible linker (SEQ ID NO: 12) to create a PG9 single chain variable fragment (scFv). This PG9 scFv was then fused via a flexible linker (SEQ ED NO: 14) to the N-terminal and/or C-terminal of the ibalizumab heavy chain to create bispecific ibalizumab-PG9 (Figure 12). The bispecific fusion antibody with PG9 scFv fused to the N-terminus of the ibalizumab heavy chain (Figure 15) was used in the following experiments. The Fc region of the ibalizumab of this fusion construct also contained a LALA mutation. The LALA mutation is further described hereinbelow.

[00114] Ibalizumab-PG9 fusion antibody retains its binding functions. The ability of the fusion antibody to bind its cognate antigens was assessed using ELISA. As seen in Figure 13 (left side), the fusion antibody was able to bind human CD4 comparably to ibalizumab. Biacore studies indicate that the addition of the PG9 scFv had no effect on the affinity at which the antibody binds CD4 (data not shown). To determine if the ibalizumab-PG9 fusion retained the inherent binding activity of PG9, the inventors demonstrated that the fusion antibody was able to bind gpl20 comparably to the anti-gpl20 antibody F105 (Figure 13, right side). As expected, ibalizumab was unable to bind gpl20, even when tested at concentrations 10-fold higher than that of the fusion antibody.

[00115] Ibalizumab-PG9 fusion antibody retains and improves virus neutralization activity as compared to ibalizumab alone. To assess the antiviral efficacy of the ibalizumab- PG9 fusion antibody, the inventors determined its HIV- l neutralization capacity using the standardized single cycle TZM-bl reporter assay. The ibalizumab-PG9 fusion antibody was able to neutralize 9 out of 9 ibalizumab-resistant viruses to greater than 95% inhibition (Figure 14, top right). In addition to improving the breadth of ibalizumab, the fusion antibody exhibited significantly enhanced potency (p=0.002), up to 150-fold compared to ibalizumab.

Example-3. Construction and Testing of Ibalizumab-VRCOl

[00116] This Example describes the generation and testing of Ibalizumab-VRCOl (also referred to as "VRCOl -Ibalizumab"), a novel bispecific HIV- l neutralizing antibody (NAb) for the prevention and treatment of HIV/AIDS. Ibalizumab-VRCOl was created as a fusion of a single-chain Fv (scFv) version of the anti-HIV envelope NAb, VRCOl , fused to the anti-CD4 Ab ibalizumab. Ibalizumab-VRCOl demonstrated significantly increased antiviral breadth over ibalizumab or VRCOl alone. In particular, it potently inhibited viral entry of all ibalizumab- resistant viruses tested. In addition, complementary resistance to ibalizumab and VRCOl occur via a mutually exclusive genotype. The dominant pathway to ibalizumab resistance involves abrogation of potential N-linked glycosylation (PNG) of the V5 N-terminus. It has been previously demonstrated from a large panel of viruses and from site-directed mutants that the presence of a V5 N-terminal PNG site is sufficient to confer sensitivity to ibalizumab. In contrast, the presence of a V5 N-terminal PNG site substantially reduces (>20-fold) the sensitivity to VRCOl . Therefore, combination therapy with ibalizumab and VRCOl, either as a fusion antibody or co-administered, is believed to provide a substantially higher genetic barrier to resistance compared to such combinations of any other broadly NAbs against HIV described to date.

[00117] Construction and expression of the ibalizumab- VRCOl fusion antibodies.

To improve the breadth of ibalizumab and VRCOl , the inventors created bispecific ibalizumab- VRC01 to simultaneously target both the CD4 receptor on host T-cells and the HIV envelope protein gpl20. VRCOl is currently one of the most potent and broadest anti-HIV-1 antibodies, targeting the CD4 binding site of the HIV envelope trimer. To create the fusion antibody, the inventors fused the heavy chain variable domain of VRCOl to the light chain variable domain of VRCOl via a flexible (GGGGS) 4 (SEQ Π) NO: 22) linker to create a VRCOl single chain variable fragment (scFv). This VRCOl scFv was then fused via the flexible (GGGGS) 4 (SEQ ID NO: 22) linker to the N-terminal and/or C-terminal of the ibalizumab heavy chain to create bispecific ibalizumab-VRCOl (Figure 16). The bispecific fusion antibody with VRCOl scFv fused to the N-terminus of the ibalizumab heavy chain (Figure 19) was used in the following experiments. The Fc region of the ibalizumab of this fusion construct also contained a LALA mutation. The LALA mutation is further described hereinbelow.

[00118] Ibalizumab-VRCOl fusion antibody retains its binding functions. The ability of the fusion antibody to bind its cognate antigens was assessed using ELISA. As seen in Figure 17 (left side), the fusion antibody was able to bind human CD4 comparably to ibalizumab. To determine if the ibalizumab-VRCOl fusion retained the inherent binding activity of VRCOl , the inventors demonstrated that the fusion antibody was able to bind gpl20 (Figure 17, right side). As expected, ibalizumab alone was unable to bind gpl20. [00119] Ibaiizumab- VRCOl fusion antibody retains and improves virus

neutralization activity as compared to ibaiizumab alone. To assess the antiviral efficacy of the ibaiizumab- VRCOl fusion antibody, the inventors determined its HIV- 1 neutralization capacity using the standardized single cycle TZM-bl reporter assay. The ibaiizumab- VRCOl fusion antibody was able to neutralize 10 out of 10 ibalizumab-resistant viruses to 100% inhibition (Figure 18, right).

ExampIe-4. Improvements of Bispecific Fusion Antibodies Over Ibaiizumab In Breadth and Potency.

[00120] The improvement of the fusion antibodies constructed herein as compared to ibaiizumab is illustrated as follows.

[00121] The top-left panel of Fig. 20 recapitulates the in vitro neutralizing activity of unmodified ibaiizumab (iMab) against a panel of 1 18 HIV isolates, illustrating the "hole" in its repertoire. An IC50 of >10 ug/mL was noted for a small fraction of the viruses, but high-level neutralization (MPI >80%) was not achieved for many more viruses. iMab-m36 (Example 1) demonstrated a modest increase in breadth (top-right, Fig. 20). VRCOl -iMab (Example 3) showed even greater breadth and potency with IC50s consistently between 0.01 and 0.1 μg/mL (bottom-left). Most notable were the neutralizing activities exhibited by PG9-iMab (Example 2) (bottom-right), where breadth of 100% (as measured by >80% neutralization) was observed, as was a significant increase (> 200-fold by IC90) in potency (median IC50, IC90 and IC99 of 0.005 Hg/mL, 0.028 μg/mL, and 0.080 μg/mL, respectively).

[00122] The remarkable gain in anti-HIV activity of the fusion antibodies is even more evident when shown as percentage coverage of viruses neutralized over a range of

concentrations (Fig. 21). In particular, 99% of the viruses were neutralized by PG9-iMab at concentrations <0.1 μg/mL.

[00123] The antiviral potency of the bispecific antibodies was also displayed by showing their IC50s on a scattered plot in comparison with results for ibaiizumab (Fig. 22). The improvement is readily evident. Example-5. PK Improvements on the Ibalizumab Backbone

[00124] Ibalizumab has a relatively short half-life in vivo (3.5 days for IV administration and -10 days for SC administration). mAbs humanized with IgG4 (such as ibalizumab) are known to have a strong tendency to switch their inter-chain disulfide bonds to intra-chain disulfide bonds, resulting in the dissociation of the antibody into monomers. In order to improve stability, the Fc region of the ibalizumab was modified to introduce the so-called "LALA" (Leu to Ala and Leu to Ala) mutations. The Fc regions of PG9-ibalizumab (Figure 15) and VRCOl -ibalizumab (Figure 19) each contained a set of LALA mutations ((Leu to Ala and Leu to Ala). In an additional construct, the ibalizumab backbone was modified by replacing IgG4 with human IgGl carrying the LALA mutations (L234A/L235A) (Hessell et al., Nature 449(7158): 101 -4, 2007) that reduced effector function mediated through FcRs (with exception of FcRn, see below). The particular "LALA" modifications made in this additional construct are L239F, L240E (the numbering system does not account for the 19 amino acid leader sequence) and correspond to position 258 and 259 of SEQ ID NO: 30. When infused into rhesus monkeys in short-term experiments, this new variant was more stable by about 5- fold relative to ibalizumab (Fig. 23).

[00125] In addition, the inventors also modified the backbone of ibalizumab in order to increase the recycling of ibalizumab via the antibody salvage pathway. FcRn, the so-called neonatal FcR, has been shown to be involved in the transcytosis of IgG across epithelium, allowing transfer of maternal antibodies to the fetus (Ghetie et al., Immunol. Res. 25(2):97- 1 13, 2002). Considerable evidence exists to support the additional role of FcRn in regulating levels of circulating IgG (Roopenian et al., Nat. Rev. Immunol. 7(9):715-25, 2007). Serum IgG is taken up by endothelial cells into their endosomes via fluid-phase pinocytosis. Typically, the Fc region of IgG binds FcRn only at pH 6.0-6.5 (endosomes), while dissociation of the IgG- FcRn complex is facilitated at pH 7.0-7.4 (cell surface). Therefore, upon acidification within endosomes, IgG binds to FcRn, which is then recycled to the cell surface and released from FcRn. IgG molecules that are not bound to FcRn within the endosomes are degraded. FcRn thus functions as a salvage receptor, rescuing IgG from the lysosomal degradation pathway. Therefore, manipulations to enhance the IgG-FcRn interaction could improve the PK of a mAb in vivo. Indeed, it has been reported that M428L N434S mutations have beneficial effects on PK (Zalevsky et al., Nat. Biotechnol. 28(2): 157-9, 2010). With this combination of mutations, affinity for FcRn at pH 6.0 increased by 1 1 -fold, largely due to a slower off rate. When placed into the context of bevacizumab (Avastin, IgG l , anti-VEGF, approved for treatment of colon, lung, and breast cancer) or cetuximab (Erbitux, IgGl , anti-EGFR, approved for treatment of colon and head and neck cancers), the PK profile of both antibodies improved by 5 -fold in human FcRn transgenic mice and by 3-fold in cynomolgus monkeys. The inventors made this set of mutations (M433L, N439S) in the IgGl -LALA version of ibalizumab (the numbering system does not account for the 19 amino acid leader sequence), which correspond to positions 452 and 458, respectively of SEQ ID NO: 30. This modification led to a ~3-fold improvement in PK in human-FcRn-transgenic mice versus wild-type mice (Fig. 24).

[00126] SEQ ED NO: 30 sets forth the amino acid sequence of iMabm36 heavy chain with the modifications discussed above. Residues 1 -19 of SEQ ED NO: 30 constitute a leader sequence (SEQ ED NO: 10); residues 20-471 represent the iMab heavy chain with IgG l-LALA and FcRn mutations (SEQ ED NO: 31); residues 472-489 represent a linker (SEQ ED NO: 32), and residues 490-606 represent m36 variable heavy chain (SEQ ID NO: 4).

Example-6. PK Evaluation, Immunogenicity Assessment, and SHIV Prevention Studies In Monkeys.

[00127] Bispecific fusion antibody candidates with the desired antiviral and PK properties as determined by in vitro assessment of the binding affinity for both CD4 and gpl20 are further assessed in vivo in rhesus macaques. However, to avoid the immunogenicity of the humanized antibody molecules in monkeys, rhesus equivalents of the bispecific humanized antibodies were generated. For example, for ibalizumab containing the PK enhancement mutations (Gl -LALA and M428IJN434S), the CHI, CH2, and CH3 domains were first replaced with their rhesus counterparts, as depicted in Fig. 25A. Such a chimeric construct could still have a high probability of being immunogenic in monkeys because of the "human" framework residues in the Fv region. Thus, these framework residues were systematically replaced with rhesus amino acids, thereby creating a full "monkeynized" version of the antibody, which was called Rhi ab (depicted in Fig. 25A). During this process, care was taken to avoid any loss in the antigen-binding property of the antibody. More specifically, based on the sequence alignment of iMab with GenBank available IgG of rhesus macaque sequences, a total of 8 amino acid residues (3 from heavy chain and 5 from light variable regions) of iMab were identified. All eight amino acid residues and combinations thereof were substituted with amino acids that were conserved in rhesus IgG. The amino acid sequences of the heavy and light chains of one of the resulting antibodies (RhiMab) are shown in Fig. 26 A. RhiMab was functionally characterized by a) sCD4 competition binding ELISA, 2) BiaCore binding, and 3) and a viral neutralization activity assay.

[00128] The PK profile of RhiMab in monkeys is determined as follows. A range of doses (from 1 mg/kg to 10 mg kg) of this antibody is administered SC to macaques. Blood is drawn serially in the ensuing month to measure the serum level of the antibody over time. PBMCs are isolated to assess the percentage of CD4 receptors on T-cells bound by the antibody, and the resultant receptor occupancy values are correlated with serum antibody concentrations. Rectal and cervico-vaginal (if available) biopsies are obtained one week after RhiMab administration to quantify the extent of antibody coating of mucosal CD4 T-cells. It is crucially important to have adequate antibody penetration into these mucosal sites, through which HIV enters a new host. Three additional SC doses of RhiMab are given to each monkey at 1 , 2, and 3 months following the initial injection. Again serial blood samples are drawn to determine whether there is accumulation of antibody over repeated dosing as well as to evaluate changes in the PK profile or receptor occupancy values during chronic administration. During this latter period, mucosal biopsies are only performed sparingly, as dictated by the PK findings.

[00129] An unusually low PK profile could hint at the development of an immunogenic response. An assay is currently under development to detect anti-RhiMab antibodies in macaque serum over time. A negative result would confirm that the RhiMab scaffold on which to build the bispecific constructs was indeed non-immunogenic. If immunogenicity is a frequent occurrence, attempts will be made to map the site(s) on RhiMab that is (are) responsible for inducing an antibody response. Once defined, substitutions at the site(s) on RhiMab would be made with the hope of eliminating the immunogenic potential. [00130] PG9 scFv was selected for conversion into the rhesus equivalents. Specifically, amino acids in human PG9 scFv were identified based on the conserved amino acid usage in a panel of rhesus macaque antibody sequences found from GenBank. A total of 19 amino acid residues ( 14 from heavy chain and 5 from light chain) were identified and changed to generate rhesus PG9 scFv. The sequences of the heavy and light chains of the rhesus PG9 scFv are shown in Fig. 26B.

[00131] A bispecific fusion was then created with rhesus PG9 scFv and RhiMab, as shown in Fig. 25B. The bispecific antibody is tested in an initial PK study in monkeys similar to those already described. A dose is then chosen based on the maintenance for 2 months of serum levels above the IC90 and/or high receptor occupancy. At week 0, the antibody is administered SC to six rhesus macaques, while six additional animals serve as untreated controls. Each antibody is given again at the same dose SC at week 8 and week 16. Beginning on day 4, all monkeys are challenged intrarectally (IR) with low-dose SHIV. The IR inoculations are repeated weekly for 1 1 more times. All animals are monitored frequently over 6 months for seroconversion, plasma viremia, CD4 cell counts, and clinical manifestations of immunodeficiency. Protection conferred by the bispecific antibodies are scored by three different measures. First is complete protection against SHIV infection; second is the number of extra viral inoculations needed to achieve infection; and the third is an attenuation of infection as reflected by low viral loads, high CD4 cell counts, and lack of disease progression.

Example-7. Affinity Maturation to Alter the Fab Binding Recognition Region of

Ibalizumab.

[00132] Random mutations were generated in CDR residues chosen on the basis of the structure information from the ibalizumab-CD4 complex. A standard yeast display screening system was utilized to identify variants with higher or lower affinities. Guided by structural information, random mutations were introduced at specified positions within the CDRs to generate a single-chain Fv (scFv) library. Variants in the library were then transformed into yeast and subsequently expressed on the cell surface. Human soluble CD4 (sCD4) was used to bind cells expressing scFv in competition with unmodified scFv. CD4 binding was detected by staining with an anti-CD4 antibody followed by secondary detection reagents. The binding signal was measured by flow cytometry and the higher affinity CD4 binding clones were isolated by fluorescent-labeled cell sorting. The N-terminal c-myc epitope tag on scFv was also simultaneously stained by a second color with an anti-c-myc antibody in order to ensure that the full-length scFv expressed clones were properly separated by cell sorting. Selected clones were grown again and assessed for higher affinity CD4 binding in multiple rounds of selection to ensure enrichment of the highest affinity forms. After the final round of selection, the best yeast clones were isolated and their respective scFvs were subjected to DNA sequencing in order to identify the mutations present. . ·

[00133] Figure 27 is a schematic summary of the method used to conduct "in vitro affinity maturation". Example shown therein is for mutations directed to CDR-H1 , but a similar approach was taken for other CDRs.

[00134] Higher affinity forms were generated, including one CDR-H1 variant of ibalizumab that has a 12.5-fold improvement in affinity (KD of 10 pM) (Table 1). iMab mutants tested by yeast platform demonstrated increased or decreased affinity to CD4 (Table 2). Variants with the highest and lowest affinity were confirmed by Biacore (Table 3).

Affinity improvement after 2 rounds of screening

- tested by yeast display platform

Table 2: Afflnity tested by yeast platform for increased or decreased iMab mutants