Polypeptides targeting HIV fusion

Information

  • Patent Grant
  • 11155602
  • Patent Number
    11,155,602
  • Date Filed
    Wednesday, August 7, 2019
    4 years ago
  • Date Issued
    Tuesday, October 26, 2021
    2 years ago
Abstract
The invention is directed to polypeptides comprising a CD4 binding moiety, a gp41 binding moiety, a HIV fusion peptide inhibitor moiety and combinations thereof. More specifically, the present invention relates to polypeptides comprising a fibronectin-based scaffold domain protein that binds CD4, a fibronectin-based scaffold domain protein that binds the N17 domain of gp41, and a HIV fusion peptide inhibitor or combinations thereof. The invention also relates to the use of the innovative proteins in therapeutic applications to treat HIV.
Description
FIELD OF THE INVENTION

The invention is directed to polypeptides comprising a CD4 binding moiety, a gp41 binding moiety, a HIV fusion peptide inhibitor moiety and combinations thereof. More specifically, the present invention relates to polypeptides comprising a fibronectin-based scaffold domain protein that binds CD4, a fibronectin-based scaffold domain protein that binds the N17 domain of gp41, and a HIV fusion peptide inhibitor or combinations thereof. The invention also relates to the use of the innovative proteins in therapeutic applications to treat HIV.


BACKGROUND OF THE INVENTION

Acquired immunodeficiency syndrome (AIDS) is the result of infection by the retrovirus known as human immunodeficiency virus (HIV). It remains a major medical problem, with an estimated 35 million people infected worldwide at the end of 2013. During that year, there were 2.1 million new infections, with 1.5 million people dying from complications due to AIDS.


Current therapy for HIV-infected individuals consists of a combination of approved anti-retroviral agents. Over two dozen drugs are currently approved for HIV infection, either as single agents, fixed dose combinations or single tablet regimens, the latter two containing 2-4 approved agents. These agents belong to a number of different classes, targeting either a viral enzyme or the function of a viral protein during the virus life cycle. Thus, agents are classified as either nucleotide reverse transcriptase inhibitors (NRTIs), non-nucleotide reverse transcriptase inhibitors (NNRTIs), protease inhibitors (PIs), integrase inhibitors (INIs), or entry inhibitors (one entry inhibitor, maraviroc, targets the host CCR5 protein, while the other, enfuvirtide, is a peptide that targets the gp41 region of the viral gp160 protein). In addition, a pharmacokinetic enhancer with no antiviral activity (cobicistat) has been approved for use in combinations with antiretroviral agents (ARVs) that require boosting.


Despite the armamentarium of agents and drug combinations, there remains a medical need for new anti-retroviral agents, due in part to the need for chronic dosing to combat infection. Significant problems related to long-term toxicities are documented, creating a need to address and prevent these co-morbidities (e.g., CNS, CV/metabolic, renal disease). Also, increasing failure rates on current therapies continue to be a problem, due either to the presence or emergence of resistant strains or to non-compliance attributed to drug holidays or adverse side effects. For example, despite therapy, it has been estimated that 63% of subjects receiving combination therapy remained viremic, as they had viral loads>500 copies/ml (Oette, M. et al., “Primary HIV Drug Resistance and Efficacy of First-Line Antiretroviral Therapy Guided by Resistance Testing”, J. Acq. Imm. Def. Synd., 41(5):573-581 (2006)). Among these patients, 76% had viruses that were resistant to one or more classes of antiretroviral agents. As a result, new drugs are needed that are more convenient, have high genetic barriers to the development of resistance and have improved safety over current agents.


It is now well known that cells can be infected by HIV through a process by which fusion occurs between the cellular membrane and the viral membrane. The generally accepted model of this process is that the viral envelope glycoprotein complex (gp120/gp41) interacts with cell surface receptors on the membranes of the target cells. Following binding of gp120 to cellular receptors (e.g., CD4 in combination with a chemokine co-receptor such as CCR5 or CXCR4), a conformational change is induced in the gp120/gp41 complex that allows gp41 to insert into the membrane of the target cell and mediate membrane fusion. As these entry processes occur on the cell membrane, they are amenable for inhibition by macrocmolecules, which include biologic peptides (Haqqani et al., Antiviral Res., 98:158 (2013)). For instance, the approved antiviral peptide enfuvirtide (FUZEON®) targets a region of gp41 involved in membrane fusion. Larger polypeptides such as monoclonal antibodies can also inhibit different aspects of virus entry. A monoclonal antibody targeting the first step of virus entry, interaction with the cellular receptor CD4 (ibalizumab; Bruno et al., J. Antimicrob. Chemother., 65:1839 (2010)), as well as a monoclonal antibody targeting the co-receptor CCR5 (PRO-140; Tenorio, Curr. HIV/AIDS Rep., 8:1 (2011)) have both shown positive results in Phase 2a trials. These antibodies also have the property of being long acting antiretrovirals, with potential dosing regimens of weekly to monthly (Jacobson et al., J. Infect. Dis., 201:1481 (2010); Jacobson et al., Antimicrob. Agents Chemother., 53:450 (2009)).


Another property of peptide entry inhibitors is that enhanced or synergistic potency can be obtained if two peptide inhibitors are attached to each other, or if a single inhibitor is localized near the site of action through binding to membrane biomolecules. Thus, attaching a fusion peptide inhibitor to a monoclonal antibody targeting CCR5 (Kopetzki et al., Virol. J., 5:56 (2008)) or attaching a cholesterol moiety to the C-terminus of a peptide fusion inhibitor to place it at the surface of the target cell membrane (Ingallinela et al., Proc. Natl. Acad. Sci. USA, 106:5801 (2009); Augusto et al., J. Antimicrob. Chemother., 69:1286 (2014)) drastically increases the potency of the combined molecule compared to the separate molecules. Similarly, bispecific antibodies consisting of anti-HIV-1 neutralizing antibody fragments targeting gp120 fused to ibalizumab showed synergistic increases in potency compared to the individual inhibitors (Sun et al., J. Acquir. Immune Defic. Syndr., 66:473 (2014)). The Combinectin molecules of the invention makes use of these various properties.


SUMMARY OF THE INVENTION

The invention is directed to polypeptides comprising a CD4 binding moiety, a gp41 binding moiety, a HIV fusion peptide inhibitor moiety and combinations thereof.


One embodiment of the invention is directed to polypeptides comprising a fibronectin-based scaffold domain protein that binds CD4, a fibronectin-based scaffold domain protein that binds the N17 domain of gp41, and a HIV fusion peptide inhibitor.


In one embodiment of the invention, the three domains are connected to each other by linkers. In another embodiment of the invention, the three domains may be connected to each other in any order. In another embodiment of the invention, the polypeptide comprises an amino acid sequence at least 80%, 85%, 90%, 95%, 98%, 99% or 100% identical to the non-linker regions of SEQ ID NO: 3, 5, 7 or 9.


The invention is also directed to polypeptides comprising a fibronectin-based scaffold domain protein that binds CD4 and a fibronectin-based scaffold domain protein that binds the N17 domain of gp41. In one embodiment of the invention, the two domains are connected to each other by linkers. In another embodiment of the invention, the two domains may be connected to each other in any order.


The invention is also directed to polypeptides comprising a fibronectin-based scaffold domain protein that binds CD4 and a HIV fusion peptide inhibitor. In one embodiment of the invention, the two domains are connected to each other by linkers. In another embodiment of the invention, the two domains may be connected to each other in any order.


The invention is also directed to polypeptides comprising a fibronectin-based scaffold domain protein that binds the N17 domain of gp41, and a HIV fusion peptide inhibitor. In one embodiment of the invention, the two domains are connected to each other by linkers. In another embodiment of the invention, the two domains may be connected to each other in any order. In another embodiment of the invention, the polypeptide comprises an amino acid sequence at least 80%, 85%, 90%, 95%, 98%, 99% or 100% identical to the non-linker regions of SEQ ID NO: 410-428.


Another embodiment of the invention is also directed to polypeptides comprising three active domains, a fibronectin-based scaffold domain protein that binds CD4, a gp41 binding moiety and a HIV fusion peptide inhibitor moiety. The invention is also directed to polypeptides comprising a fibronectin-based scaffold domain protein that binds gp41, a CD4 binding moiety and a HIV fusion peptide inhibitor moiety. The invention is also directed to polypeptides comprising a CD4 binding moiety, a gp41 binding moiety and a HIV fusion peptide inhibitor. In one embodiment of the invention, the two domains are connected to each other by linkers. In another embodiment of the invention, the two domains may be connected to each other in any order.


The invention is also directed to polypeptides comprising two active domains, a fibronectin-based scaffold domain protein that binds CD4 and a gp41 binding moiety. The invention is also directed to polypeptides comprising a fibronectin-based scaffold domain protein that binds gp41 and a CD4 binding moiety. The invention is also directed to polypeptides comprising a CD4 binding moiety and a HIV fusion peptide inhibitor. The invention is also directed to polypeptides comprising a gp41 binding moiety and a HIV fusion peptide inhibitor. In one embodiment of the invention, the two domains are connected to each other by linkers. In another embodiment of the invention, the two domains may be connected to each other in any order.


Another embodiment of the invention is also directed to anti-CD4 Adnectin™ anti-N17 Adnectin™, or HIV fusion peptide inhibitors. In another embodiment of the invention, the polypeptide comprises an amino acid sequence at least 80%, 85%, 90%, 95%, 98%, 99% or 100% identical to the non-linker regions of SEQ ID NO: 95-114 or SEQ ID NO: 115-371 or SEQ ID NO: 372-392, respectively.


In another embodiment of the invention, a pharmacokinetic (PK) moiety is attached to the polypeptides of the invention. Examples of a PK moiety include but are not limited to polyethylene glycol, sialic acid, Fc, Fc fragment, transferrin, serum albumin (HSA), a serum albumin binding protein, and a serum immunoglobulin binding protein. In one embodiment of the invention, the PK moiety may be attached to the linker regions, or the N- or C-terminus of the polypeptide of the invention. In another embodiment of the invention, the polypeptide comprises an amino acid sequence at least 80%, 85%, 90%, 95%, 98%, 99% or 100% identical to the non-linker regions of SEQ ID NO: 4, 6, 8 or 10.


The invention is also directed to a polypeptide comprising the amino acid sequence of any one of the sequences shown in SEQ ID NO: 3-10.


The invention is also directed to a pharmaceutical composition comprising one or more of the polypeptides of the invention and a carrier.


The invention also provides a method of treating HIV in a subject comprising administering an effective amount of the polypeptides of the invention.





BRIEF DESCRIPTION OF THE FIGURES


FIG. 1 is a diagram of alternative Combinectins. One of the Adnectin™ binds CD4, a second Adnectin™ binds the HR1 region of gp41. The peptide also binds in the HR1 region of gp41. The different components of the Combinectin are connected to each other in any order by linkers. Any of the Combinectins may have a PK moiety attached, such as HSA or Fc.



FIG. 2 shows the amino acid sequences of Fc fusion-Combinectin 3137_(SEQ ID NO:4), 3151_(SEQ ID NO:6) and human serum albumin (HSA) fusion-Combinectin 3191_(SEQ ID NO:8), and 3202_(SEQ ID NO:10). The Fc and HSA sequences are in bold. The anti-CD4 Adnectin sequences are underlined. The anti-N17 Adnectin sequences are double underlined. The HIV inhibitor peptide sequences are underlined in bold. The linker sequences are italicized.



FIG. 3 shows the potency (EC50 and EC90) of Combinectin 3137, 3151, 3191 and 3202, as described in Example 2.



FIG. 4 shows the PK properties of Combinectin 3137, 3151, 3191 and 3202 as described in Example 3.



FIG. 5 shows the amino acid sequences of anti-N17 Adnectin™ in combination with an HIV fusion peptide inhibitor, which correspond to the sequences described in Table 4 minus the N-terminus and C-terminus extended regions. The anti-N17 Adnectin™ sequences are double underlined. The HIV fusion peptide inhibitor sequences are underlined in bold. The linker sequences are italicized.



FIG. 6 shows the WebLogo for the CD loop of anti-CD4 Adnectin™. WebLogo generates sequence logos, graphical representations of the patterns within a multiple sequence alignment. Each logo consists of stacks of letters, one stack for each position in the sequence. The overall height of each stack indicates the sequence conservation at that position (measured in bits), whereas the height of symbols within the stack reflects the relative frequency of the corresponding amino or nucleic acid at that position (Crooks, G. E. et al., “WebLogo: A sequence logo generator”, Genome Research, 14:1188-1190 (2004)).



FIG. 7 shows the WebLogo for the FG loop of anti-CD4 Adnectin™ (Schneider, T. D. et al., “Sequence Logos: A New Way to Display Consensus Sequences”, Nucleic Acids Res., 18:6097-6100 (1990)).



FIG. 8 shows point mutation data for the HIV fusion peptide inhibitor. The aspartic acid (D) near the C-terminus of the HIV fusion peptide inhibitor was replaced with the amino acids listed along the x axis.





DETAILED DESCRIPTION OF THE INVENTION
Definitions

Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by the skilled artisan. Although any methods and compositions similar or equivalent to those described herein can be used in practice or testing of the present invention, the preferred methods and compositions are described herein.


“Polypeptide” as used herein refers to any sequence of two or more amino acids, regardless of length, post-translation modification, or function. “Polypeptide”, “peptide”, and “protein” are used interchangeably herein. Polypeptides can be modified in any of a variety of standard chemical ways (e.g., an amino acid can be modified with a protecting group; the carboxy-terminal amino acid can be made into a terminal amide group; the amino-terminal residue can be modified with groups to, e.g., enhance lipophilicity; or the polypeptide can be chemically glycosylated or otherwise modified to increase stability or in vivo half-life). Polypeptide modifications can include the attachment of another structure such as a cyclic compound or other molecule to the polypeptide and can also include polypeptides that contain one or more amino acids in an altered configuration (i.e., R or S; or, L or D).


The peptides of the invention may include, for example, proteins derived from the tenth type III domain of fibronectin that have been modified to bind to CD4 or N17 domain of gp41 and are referred to herein as, “anti-CD4 Adnectin™”, “anti-N17 Adnectin™”, “CD4 Adnectin™” or “gp41 Adnectin™”. The polypeptides of the invention may also include peptides modeled after the heptad repeat 2 (HR2) region of HIV envelope glycoprotein gp41, which inhibit fusion by binding the heptad repeat 1 (HR1) region of gp41 and are referred to herein as “HIV fusion peptide inhibitor” or “fusion peptide inhibitor”. The polypeptides of the invention also include “Combinectins” comprising an anti-CD4 Adnectin™ linked to an anti-N17 Adnectin™ linked to a HIV fusion peptide inhibitor (FIG. 1). Alternatively, the Combinectin comprises an anti-CD4 Adnectin™ linked to an anti-N17 Adnectin™ or an anti-N17 Adnectin™ linked to a HIV fusion peptide inhibitor or an anti-CD4 Adnectin™ linked to a HIV fusion peptide inhibitor.


A “polypeptide chain”, as used herein, refers to a polypeptide wherein each of the domains thereof is joined to other domain(s) by peptide bond(s), as opposed to non-covalent interactions or disulfide bonds.


An “isolated” polypeptide is one that has been identified and separated and/or recovered from a component of its natural environment. Contaminant components of its natural environment are materials that would interfere with diagnostic or therapeutic uses for the polypeptide, and may include recombinant host cell proteins and other proteinaceous or nonproteinaceous solutes. In one embodiment, the polypeptides will be purified (1) to greater than 95% by weight of polypeptide as determined by the Lowry method, and most preferably more than 99% by weight, or (2) to homogeneity by SDS-PAGE under reducing or nonreducing condition using Coomassie blue or, silver stain. Ordinarily, isolated polypeptide will be prepared by at least one purification step.


“Percent (%) amino acid sequence identity” herein is defined as the percentage of amino acid residues in a candidate sequence that are identical with the amino acid residues in a selected sequence, after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent sequence identity, and not considering any conservative substitutions as part of the sequence identity. Alignment for purposes of determining percent amino acid sequence identity can be achieved in various ways that are within the skill in the art, for instance, using publicly available computer software such as BLAST, BLAST-2, ALIGN, ALIGN-2 or Megalign (DNASTAR®) software. Those skilled in the art can readily determine appropriate parameters for measuring alignment, including any algorithms needed to achieve maximal alignment over the full-length of the sequences being compared. For example, the % amino acid sequence identity of a given amino acid sequence A to, with, or against a given amino acid sequence B (which can alternatively be phrased as a given amino acid sequence A that has or comprises a certain % amino acid sequence identity to, with, or against a given amino acid sequence B) is calculated as follows: 100 times the fraction X/Y where X is the number of amino acid residues scored as identical matches by the sequence alignment program ALIGN-2 in that program's alignment of A and B, and where Y is the total number of amino acid residues in B. It will be appreciated that where the length of amino acid sequence A is not equal to the length of amino acid sequence B, the % amino acid sequence identity of A to B will not equal the % amino acid sequence identity of B to A.


As used herein, “conservative substitution” denotes the replacement of an amino acid residue by another, without altering the overall conformation and function of the peptide, including, but not limited to, replacement of an amino acid with one having similar properties (such as, for example, polarity, hydrogen bonding potential, acidic, basic, shape, hydrophobic, aromatic, and the like). Amino acids with similar properties are well known in the art. For example, arginine, histidine and lysine are hydrophilic-basic amino acids and may be interchangeable. Similarly, isoleucine, a hydrophobic amino acid, may be replaced with leucine, methionine or valine. Neutral hydrophilic amino acids, which can be substituted for one another, include asparagine, glutamine, serine and threonine. By “substituted” or “modified” the present invention includes those amino acids that have been altered or modified from naturally occurring amino acids. As such it should be understood that in the context of the present invention a conservative substitution is recognized in the art as a substitution of one amino acid for another amino acid that has similar properties.


As used herein, the term “binding site” refers to the site or portion of a protein (e.g., CD4, gp41) that interacts or binds to a particular protein of the invention (e.g., as an epitope is recognized by an antibody). Binding sites can be formed from contiguous amino acids or noncontiguous amino acids juxtaposed by tertiary folding of a protein. Binding sites formed by contiguous amino acids are typically retained on exposure to denaturing solvents, whereas binding sites formed by tertiary folding are typically lost on treatment of denaturing solvents.


The binding site for an anti-CD4 moiety or anti-N17moiety of the invention may be determined by application of standard techniques typically used for epitope mapping of antibodies including, but not limited to protease mapping and mutational analysis. Alternatively, a binding site can be determined by competition assay using a reference protein (e.g., another Adnectin™ or antibody) which binds to the same polypeptide, e.g., CD4 or gp41. If the test protein and reference molecule (e.g., another Adnectin™ or antibody) compete, then they bind to the same binding site or to binding sites sufficiently proximal such that binding of one molecule interferes with the other.


The terms “specifically binds”, “specific binding”, “selective binding”, and “selectively binds”, as used interchangeably herein refers to a protein that exhibits affinity for a CD4 or gp41, but does not significantly bind (e.g., less than about 10% binding) to a different polypeptide as measured by a technique available in the art such as, but not limited to, Scatchard analysis and/or competitive binding assays (e.g., competition ELISA, BIACORE® SPR assay). The term is also applicable where e.g., a binding domain of a protein of the invention is specific for CD4 or gp41.


The term “preferentially binds” as used herein refers to the situation in which the peptides of the invention bind CD4 or gp41 at least about 20% greater than it binds a different polypeptide as measured by a technique available in the art such as, but not limited to, Scatchard analysis and/or competitive binding assays (e.g., competition ELISA, BIACORE® SPR assay).


As used herein, the term “cross-reactivity” refers to a protein which binds to more than one distinct protein having identical or very similar binding sites.


The term “Kd”, as used herein, is intended to refer to the dissociation equilibrium constant of a particular Adnectin™-protein, fusion peptide inhibitor-protein or Combinectin-protein (e.g., CD4 and/or gp41) interaction or the affinity of an Adnectin™ fusion peptide inhibitor or Combinectin for a protein (e.g., CD4 and/or gp41), as measured using a surface plasmon resonance assay or a cell binding assay. A “desired Kd”, as used herein, refers to a Kd of a protein of the invention that is sufficient for the purposes contemplated. For example, a desired Kd may refer to the Kd of a Combinectin required to elicit a functional effect in an in vitro assay, e.g., a cell-based luciferase assay.


The term “kon”, as used herein, is intended to refer to the association rate constant for the association of, for example, a Combinectin into the Combinectin/protein complex.


The term “koff”, as used herein, is intended to refer to the dissociation rate constant for the dissociation of, for example, a Combinectin from the Combinectin/protein complex.


The term “IC50”, as used herein, refers to the concentration of, for example, a Combinectin that inhibits a response, either in an in vitro or an in vivo assay, to a level that is 50% of the maximal inhibitory response, i.e., halfway between the maximal inhibitory response and the untreated response.


The terms “inhibit” or “neutralize” as used herein with respect to an activity of a protein of the invention means the ability to substantially antagonize, prohibit, prevent, restrain, slow, disrupt, eliminate, stop, reduce or reverse e.g., progression or severity of that which is being inhibited including, but not limited to, a biological activity or property, a disease or a condition. The inhibition or neutralization is preferably at least about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95% or higher.


The term “PK” is an acronym for “pharmacokinetic” and encompasses properties of a compound including, by way of example, absorption, distribution, metabolism, and elimination by a subject. A “PK modulation protein” or “PK moiety” as used herein refers to any protein, peptide, or moiety that affects the pharmacokinetic properties of a biologically active molecule when fused to or administered together with the biologically active molecule. Examples of a PK modulation protein or PK moiety include PEG, human serum albumin (HSA) binders (as disclosed in U.S. Publication No. 2005/0287153, U.S. Pat. No. 7,696,320, PCT Publication Nos. WO 2009/083804 and WO 2009/133208), human serum albumin, Fc or Fc fragments and variants thereof, and sugars (e.g., sialic acid).


The term “CD4 binding moiety” refers to any moiety that blocks HIV surface protein gp120 binding to the CD4 receptor on CD4+ T cells. The CD4 binding moiety may be anti-CD4-Adnectin™, -antibody (such as ibalizumab), -domain antibody (dAb), -antibody fragments (Fab), -bispecific antibody and fusion protein thereof.


The term “gp41 binding moiety” refers to any moiety that interferes with the interaction of the viral envelope glycoprotein complex (gp120/gp41) with T cells. The gp41 binding moiety may be anti-gp41-Adnectin™, -antibody (Ab), -domain antibody (dAb), -antibody fragments (Fab), -bispecific antibody and fusion protein thereof.


A “HIV fusion peptide inhibitor moiety” refers to any moiety that inhibits fusion by binding the heptad repeat 1 (HR1) region of gp41. Examples of fusion peptide inhibitor moiety include peptides derived from the NHR and CHR regions of gp41, designated NHR and CHR peptides, respectively. Enfuvirtide is an example of a CHR peptide.


The peptides of the invention may include, for example, the CD4 monoclonal antibody ibalizumab, an anti-N17 Adnectin™ and a HIV fusion peptide inhibitor. Alternatively, the peptides of the invention may include an anti-CD4 Adnectin™, an anti-N17 Adnectin™ and the HIV fusion peptide inhibitor enfuvirtide.


The “half-life” of an amino acid sequence or compound can generally be defined as the time taken for the serum concentration of the polypeptide to be reduced by 50%, in vivo, for example, due to degradation of the sequence or compound and/or clearance or sequestration of the sequence or compound by natural mechanisms. The half-life can be determined in any manner known per se, such as by pharmacokinetic analysis. Suitable techniques will be clear to the person skilled in the art, and may, for example, generally involve the steps of suitably administering to a subject a suitable dose of the amino acid sequence or compound of the invention; collecting blood samples or other samples from the subject at regular intervals; determining the level or concentration of the amino acid sequence or compound of the invention in said blood sample; and calculating, from (a plot of) the data thus obtained, the time until the level or concentration of the amino acid sequence or compound of the invention has been reduced by 50% compared to the initial level upon dosing. Reference is, for example, made to the standard handbooks, such as Kenneth, A. et al., Chemical Stability of Pharmaceuticals: A Handbook for Pharmacists and in Peters et al., Pharmacokinetic Analysis: A Practical Approach (1996). Reference is also made to Gibaldi, M. et al., Pharmacokinetics, Second Rev. Edition, Marcel Dekker (1982).


Half-life can be expressed using parameters such as the t1/2-alpha, t1/2-beta, HL_Lambda_z, and the area under the curve (AUC). In the present specification, an “increase in half-life” refers to an increase in any one of these parameters, any two of these parameters, any three of these parameters or all four of these parameters.


The notations “mpk”, “mg/kg”, or “mg per kg” refer to milligrams per kilogram. All notations are used interchangeably throughout the present disclosure.


The terms “individual”, “subject”, and “patient”, used interchangeably herein, refer to an animal, preferably a mammalian (including a nonprimate and a primate), including, but not limited to, murines, simians, humans, mammalian farm animals (e.g., bovine, porcine, ovine), mammalian sport animals (e.g., equine), and mammalian pets (e.g., canine and feline); preferably the term refers to humans. In a certain embodiment, the subject, is a mammal, is preferably a human and is infected with HIV.


The term “therapeutically effective amount” refers to at least the minimal dose, but less than a toxic dose, of an agent which is necessary to impart a therapeutic benefit to a subject. For example, a therapeutically effective amount of a Combinectin of the invention is an amount which in mammals, preferably humans, results in a significant decline in circulating HIV within the infected individual.


Overview


The present invention provides novel polypeptides that bind to CD4 and/or gp41. The polypeptides comprising a CD4 binding moiety, a gp41 binding moiety, a HIV fusion peptide inhibitor moiety and combinations thereof. More specifically, the present invention relates to polypeptides comprising a fibronectin-based scaffold domain protein that binds CD4, a fibronectin-based scaffold domain protein that binds the N17 domain of gp41, and a HIV fusion peptide inhibitor or combinations thereof (herein referred to as “Combinectins”).


In order to identify CD4 and gp41 Adnectins™, soluble CD4 (extracellular domain) and gp41 (various artificial constructs designed to display a triple-helical segment mimicking a portion of gp41) were presented to large synthetic libraries of Adnectins™. Adnectins™ that survived several rounds of selection were screened for CD4 or gp41 binding, for biophysical properties, and for HIV-1 inhibitory activity. The best anti-CD4 and anti-N17 Adnectin™ sequences that emerged from the screenings were mutated and subjected to further rounds of selection with increased selective pressure, achieved by lowering the target concentration and/or selecting for anti-CD4 or gp41 Adnectins™ with fast on-rates and/or slow off-rates. From this optimization process, multiple families of Adnectins™, some that targeted CD4, and others targeting gp41, were identified as HIV-1 specific inhibitors with favorable biochemical and biophysical activity.


An optimized gp41-targeting helical peptide was developed starting with sequences related to gp41 HR2 and containing changes for improving potency and breadth across HIV strains as well as increasing the resistance barrier. Peptides were produced at times synthetically and at other times as genetic fusions to inert or active Adnectins™. Optimal N- and C-terminal positions were determined in fusions to a member of the gp41 Adnectin™ family. To identify mutations that would further increase potency, an N-terminally trimmed peptide with modest potency was used, so that improvements would be more easily detected. Small libraries of the inert Adnectin™-peptide fusion were made comprising single and multiple point mutations, then the proteins were expressed and screened for biophysical properties and HIV-1 inhibitory activity. The final family of peptides consisted of optimal length peptides with various combinations of the sequences that had the most favorable profiles.


I. Fibronectin Based Scaffolds—Adnectins™


One aspect of the application provides anti-CD4 and anti-N17 Adnectins™comprising a fibronectin type III (Fn3) domain in which part or all of one or more of the solvent accessible loops has been randomized or mutated. In some embodiments, one or more residues in one or more of the non-loop beta strands has also been randomized or mutated. In some embodiments, the Fn3 domain is an Fn3 domain derived from the tenth type 3 module of human fibronectin (10Fn3):


VSDVPRDLEVVAATPTSLLISWDAPAVTVRYYRITYGETGGNSPVQEFTVPGS KSTATISGLKPGVDYTITVYAVTGRGDSPASSKPISINYRT (SEQ ID NO:1) (BC, CD, DE, and FG loops are underlined)









(BC, CD, DE, and FG loops are underlined)


(SEQ ID NO: 1)


VSDVPRDLEVVAATPTSLLISWDAPAVTVRYYRITYGETGGNSPVQE





FTVPGSKSTATISGLKPGVDYTITVYAVTGRGDSPASSKPISINYRT






In other embodiments, the non-ligand binding sequences of 10Fn3, i.e., the “10Fn3 scaffold”, may be altered, provided that the 10Fn3 retains ligand binding function and/or structural stability. A variety of mutant 10Fn3 scaffolds have been reported. In one aspect, one or more of Asp 7, Glu 9, and Asp 23 is replaced by another amino acid, such as, for example, a non-negatively charged amino acid residue (e.g., Asn, Lys, etc.). These mutations have been reported to have the effect of promoting greater stability of the mutant 10Fn3 at neutral pH as compared to the wild-type form (see, e.g., PCT Publication No. WO 02/04523). A variety of additional alterations in the 10Fn3 scaffold that are either beneficial or neutral have been disclosed. See, for example, Baton et al., Protein Eng., 15(12):1015-1020 (December 2002); Koide et al., Biochemistry, 40(34):10326-10333 (Aug. 28, 2001).


Both variant and wild-type 10Fn3 proteins are characterized by the same structure, namely seven beta-strand domain sequences designated A through G and six loop regions (AB loop, BC loop, CD loop, DE loop, EF loop, and FG loop) which connect the seven beta-strand domain sequences. The beta strands positioned closest to the N- and C-termini may adopt a beta-like conformation in solution. In SEQ ID NO:1, the AB loop corresponds to residues 14-17, the BC loop corresponds to residues 23-31, the CD loop corresponds to residues 37-47, the DE loop corresponds to residues 51-56, the EF loop corresponds to residues 63-67, and the FG loop corresponds to residues 75-87.


Accordingly, in some embodiments, the anti-CD4 or anti-N17 Adnectin™ of the invention is a 10Fn3 polypeptide that is at least 40%, 50%, 60%, 65%, 70%, 75%, 80%, 85%, or 90% identical to the human 10Fn3 domain, shown in SEQ ID NO:1. Much of the variability will generally occur in one or more of the loops. Each of the beta or beta-like strands of a 10Fn3 polypeptide may consist essentially of an amino acid sequence that is at least 40%, 50%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95% or 100% identical to the sequence of a corresponding beta or beta-like strand of SEQ ID NO:1, provided that such variation does not disrupt the stability of the polypeptide in physiological conditions.


In some embodiments, the invention provides one or more Adnectins™ comprising a tenth fibronectin type III (10Fn3) domain, wherein the 10Fn3 domain comprises a loop, AB; a loop, BC; a loop, CD; a loop, DE; a loop EF; and a loop FG; and has at least one loop selected from loop BC, CD, DE, and FG with an altered amino acid sequence relative to the sequence of the corresponding loop of the human 10Fn3 domain. In some embodiments, the Adnectins™ of the present invention comprise an 10Fn3 domain comprising an amino acid sequence at least 40%, 50%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99% or 100% identical to the non-loop regions of SEQ ID NO:1, wherein at least one loop selected from BC, CD, DE, and FG is altered. In some embodiments, the BC and FG loops are altered, and in some embodiments, the BC, DE, and FG loops are altered, i.e., the 10Fn3 domains comprise non-naturally occurring loops. In some embodiments, the AB, CD and/or the EF loops are altered. In some embodiments the CD and FG loops are altered. In some embodiments the solvent-accessible amino acids in the strands between loops are altered, with or without alteration of the adjoining loops. By “altered” is meant one or more amino acid sequence alterations relative to a template sequence (corresponding human fibronectin domain) and includes amino acid additions, deletions, substitutions or a combination thereof. Altering an amino acid sequence may be accomplished through intentional, blind, or spontaneous sequence variation, generally of a nucleic acid coding sequence, and may occur by any technique, for example, PCR, error-prone PCR, or chemical DNA synthesis.


In some embodiments, one or more loops selected from BC, CD, DE, and FG may be extended or shortened in length relative to the corresponding human fibronectin loop. In some embodiments, the length of the loop may be extended by 1-25 amino acids. In some embodiments, the length of the loop may be decreased by 1-11 amino acids. To optimize antigen binding, therefore, a loop of 10Fn3 may be altered in length as well as in sequence to obtain the greatest possible flexibility and affinity in antigen binding.


In some embodiments, the Adnectins™ comprise a Fn3 domain that comprises an amino acid sequence at least 40%, 50%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99%, or 100% identical to the non-loop regions of SEQ ID NO:1, wherein at least one loop selected from BC, CD, DE, and FG is altered. In some embodiments, the altered BC loop has up to 1, 2, 3, 4, 5, 6, 7, 8 or 9 amino acid substitutions, up to 1, 2, 3, or 4 amino acid deletions, up to 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 amino acid insertions, or a combination thereof. In some embodiments, the altered CD loop has up to 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or 11 amino acid substitutions, up to 1, 2, 3, 4, 5, or 6 amino acid deletions, up to 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 amino acid insertions, or a combination thereof. In some embodiments, the altered DE loop has up to 1, 2, 3, 4, 5, or 6 amino acid substitutions, up to 1, 2, 3, or 4 amino acid deletions, up to 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, or 13 amino acid insertions, or a combination thereof. In some embodiments, the FG loop has up to 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12 or 13 amino acid substitutions, up to 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or 11 amino acid deletions, up to 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, or 25 amino acid insertions, or a combination thereof.


Extension Sequences


In certain embodiments, the Adnectin™ molecules of the present invention may be modified to comprise an N-terminal extension sequence and/or a C-terminal extension. For example, an MG sequence may be placed at the N-terminus of the 10Fn3 defined by SEQ ID NO: 1. The M will usually be cleaved off, leaving a G at the N-terminus. The Adnectins™ described herein may also comprise alternative C-terminal tail sequences, referred to herein as truncated C-terminal or C-terminal extension sequences. Further, truncated version may be used as therapeutic molecules in the truncated form, or alternative C-terminal extensions, such as His6 tag, may be added to the truncated version. In certain embodiments, the C-terminal extension sequences (also called “tails”), comprise E and D residues, and may be between 8 and 50, 10 and 30, 10 and 20, 5 and 10, and 2 and 4 amino acids in length. In certain embodiments, the first residue of a C-terminal extension is a proline. In certain other embodiments, the first residue of a C-terminal extension is a glutamic acid.


In some embodiments, the N-terminus may be extended by up to 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or more amino acids, which may be altered in any way, before or after rounds of selection, in order to improve target binding, stability, or both. In other embodiments, the C-terminus may be extended by up to 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or more amino acids, which may be altered in any way, before or after rounds of selection, in order to improve target binding, stability, or both. In still other embodiments, both the N- and C-termini may be extended in this manner.


Anti-CD4 Adnectin™


The amino acid sequence of anti-CD4 Adnectin™ loop region CD and FG of the invention include but are not limited to those listed in Table 1 below. The CD loops described in Table 1 replace R30 through T49 of 10Fn3 defined by SEQ ID NO:1. The FG loops described in Table 1 replace D67 through N91 of 10Fn3 defined by SEQ ID NO:1.


Table 1 also lists the IC50 values for each anti-CD4 Adnectin™ comprising the listed CD/FG loop combinations.









TABLE 1







CD4 Adnectin ™-CD and FG loop combinations










CD loop
FG loop
IC50, μM
SEQ ID NO





HSYHIQYWPLGSYQRY
EYQIRVYAETGGADSDQSM
0.0025
13, 14


QVFS
GWIQIG







LSYHIQYWPLGLYQAY
EYQIRVYAETGRGESPASFG
0.0060
15, 16


QVFS
WIQIG







HAYHIQYWPLGFYQGY
EYQIRVYAETGLGDAHQSLG
0.0072
17, 18


QVFS
WIQIG







LAYHIQYWPLGWYQR
EYQIRVYAETGRGESPASFG
0.0075
19, 20


YQIFS
WIQIG







LAYHIQYWPLGWYQR
EYQIRVYAETGRGESPASFG
0.0082
21, 22


YQVFS
WIQIG







HFYHIQYWPLGLYHLY
EYQIRVYAETGRGESPASFG
0.0087
23, 24


QVFS
WIQIG







YSYHIQYWPLGWYHR
EYQIRVYAETGADDPVQAL
0.0099
25, 26


YQVFS
GWIQIG







RCYHIQYWPLGLYPLY
EYQIRVYAETGDESSVQPFG
0.0115
27, 28


QVFS
WIQIG







YSYHIQYWPLGWYQR
EYQIRVYAETDGGRSQQSFG
0.0118
29, 30


YQVFS
WIQIG







SSYHIQYWPLGAYQRY
EYQIRVYAETGRGESPASFG
0.0158
31, 32


QVFS
WIQIG







HAYHIQYWPLGLYQRY
EYQIRVYAETGRGESPASFG
0.0165
33, 34


QVFS
WIQIG







HAYYIQYWPLGSYQFY
EYQIRVYAETGRGESPASFG
0.0213
35, 36


QVFA
WIQIG







HSYHIQYWPLGSYLRY
EYQIRVYAETGRGESPASFG
0.0214
37, 38


QVFS
WIQIG







LSYHIQYWPLGFYQRY
EYQIRVYAETGRGESPASFG
0.0230
39, 40


QVFS
WIQIG







SAYHIQYWPLGWYHR
EYQIRVYAETGRGESPASFG
0.0239
41, 42


YQIFS
WIQIG







YSYHIQYWPLGAYSRH
EYQIRVYAETGGDGSEMYF
0.0260
43, 44


QLFS
GWIQIG







LAYHIQYWPLGWYHL
EYQIRVYAETGRGESPASFG
0.0272
45, 46


YQVFS
WIQIG







LAYHIQYWPLGWYQL
DYQIRVYAETSGESSEQYLG
0.0287
47, 48


YKVFS
WIQIG







HSYHIQYWPLGWYQL
EYQIRVYAETEVDSGQHSFG
0.0290
49, 50


YQVFS
WIQIG







LAYHIQYWPLGWYQR
EYQIRVYAETGESGAQQSFG
0.0297
51, 52


YQIFS
WIQIG







QSYHIQYWPLGAYQLY
EYQIRVYAETGRGESPASFG
0.0323
53, 54


QLFS
WIQIG







HAYHIQYWPLGFYQGY
EYQIRVYADTGRGYQLSYS
0.0353
55, 56


QVFS
WIQIGY







FRYHIQYWPLGGYERY
EYQIRVYAETGRGESPASFG
0.0410
57, 58


QVFT
WIQIG







HSYHIQYWPLGSYHLY
EYQIRVYAETGRGESPASFG
0.0411
59, 60


QLFS
WIQIG







HSYHIQYWPLGWYQL
EYQIRVYAETGGFGSPPNFG
0.0436
61, 62


YQVFT
WIQIG







QFYHIQYWPLGSYQRY
EYQIRVYAETGRGESPASFG
0.0509
63, 64


QVFS
WIQIG







NSYHIQYWPLGWYHR
EYQIRVYAETGRGESPASFG
0.0517
65, 66


YQVFS
WIQIG







HSYHIQYWPLGRYQLY
EYQIRVYAETGRGESPASFG
0.0562
67, 68


QVFS
WIQIG







LAYHIQYWPLGWYHL
EYQIRVYAETGGVGWHHSF
0.0587
69, 70


YQIFS
GWIQIG







HVYHIQYWPLGWYPR
EYQIRVYAETGRGESPASFG
0.0604
71, 72


YQVFS
WIQIG







HSYHIPYWELAWYQRY
EYQIRVYAETGRGESPASFG
0.0637
73, 74


QVFS
WIQIG







ESYHIQYWPLGLYHRY
EYQIRVYAETGRGESPASFG
0.0688
75, 76


QVFS
WIQIG







LAYHIQYWPLGWYQA
EYQIRVYAETGRGESPASFG
0.0703
77, 78


YQVFS
WIQIG







YLYHIQYWPLGWYHR
EYQIRVYAETGRGESPASFG
0.0715
79, 80


YQVFT
WIQIG







RFYHIQYWPLGWYHC
EYQIRVYAQTGDGSSQEYFG
0.0839
81, 82


YQVFV
WIQIG







HSYHIQYWPLGWYYR
EYQIRVYAETGGSGSQQYVV
0.0869
83, 84


YQVFS
GWIQIG







HAYHIQYWPLGFYQGY
EYQIRVYAETGRGESPASFG
0.0875
85, 86


QVFS
WIQIG







HSYHIQYWPLGLYVLY
EYQIRVYAETGAGGSEHSFG
0.1059
87, 88


QVFS
WIQIG







LSYHIQYWPLGRYERY
EYQIRVYAETVGGESLDSFS
0.1277
89, 90


QVFS
WIQIG







LSYHIQYWPLGWYQLY
EYQIRVYAETRVGGSVASFG
0.1600
91, 92


QVFY
WIQIG







LAYHIQYWPLGRYQLY
EYQIRVYAETGRGESPASFG
0.4281
93, 94


QVFS
WIQIG









In some embodiments, anti-CD4 Adnectin™ of the invention comprises an amino acid sequence at least 80%, 85%, 90%, 95%, 98%, 99% or 100% identical to the CD/FG loop region combinations of SEQ ID NOs: 13-94.


In some embodiments, anti-CD4 Adnectin™ of the invention comprises an amino acid sequence at least 80%, 85%, 90%, 95%, 98%, 99% or 100% identical to any one of the CD loop regions of SEQ ID NOs: 13, 15, 17, 19, 21, 23, 25, 27, 29, 31, 33, 35, 37, 39, 41, 43, 45, 47, 49, 51, 53, 55, 57, 59, 61, 63, 65, 67, 69, 71, 73, 75, 77, 79, 81, 83, 85, 87, 89, 91 and 93.


In some embodiments, anti-CD4 Adnectin™ of the invention comprises an amino acid sequence at least 80%, 85%, 90%, 95%, 98%, 99% or 100% identical to any one of the FG loop regions of SEQ ID NOs: 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 34, 36, 38, 40, 42, 44, 46, 48, 50, 52, 54, 56, 58, 60, 62, 64, 66, 68, 70, 72, 74, 76, 78, 80, 82, 84, 86, 88, 90, 92 and 94.


WebLogo (≤weblogo.berkeley.edu≥) was used to identify consensus sequences for anti-CD4 Adnectin™. Y32, I34, Y36, Q46 and F48 of the anti-CD4 Adnectin™ CD loop are conserved amino acids (see FIG. 6). In some embodiments, the anti-CD4 Adnectin™ comprises one or more of the conserved amino acids Y32, I34, Y36, Q46 and F48.


WebLogo identified Y68, I70, V72, A74, T76, I88 and I90 of the anti-CD4 Adnectin™ FG loop as conserved amino acids (see FIG. 7). In some embodiments, the anti-CD4 Adnectin™ comprises one or more of the conserved amino acids Y68, I70, V72, A74, T76, I88 and 190.


In some embodiments, the anti-CD4 Adnectin™ comprises one or more of the conserved amino acids Y32, I34, Y36, Q46, F48, Y68, I70, V72, A74, T76, I88 and 190.


The full length amino acid sequence of anti-CD4 Adnectin™ of the invention include but are not limited to those listed in Table 2 below. Table 2 also lists the antiviral EC50 values for each anti-CD4 Adnectin™









TABLE 2







Anti-CD4 Adnectin ™









SEQ ID
Antiviral



NO
EC50 (nM)
Anti-CD4 Adnectin ™ Protein Sequence





 95
   48
MASTSGVSDVPRDLEVVAATPTSLLISWDAPAVTVHS




YHIQYWPLGWYQRYQVFSVPGSKSTATISGLKPEVEY




QIRVYAETGGGGSQQSFGWIQIGYRTEGSGSHHHHHH





 96
    7.8
MGVSDVPRDLEVVAATPTSLLISWDAPAVTVQSYHIQ




YWPLGSYQRYQVFSVPGSKSTATISGLKPGVEYQIRV




YAETGGADSDQSMGWIQIGYRTEGDKPSQHHHHHH





 97
   11
MGVSDVPRDLEVVAATPTSLLISWDAPAVTVHSYHIQ




YWPLGWYQRYQVFSVPGSKSTATISGLKPGVEYQIRV




YAETRSGLADESFGWIQIGYRTEGDKPSQHHHHHH





 98
    4.9
MGVSDVPRDLEVVAATPTSLLISWDAPAVTVHSYHIQ




YWPLGSYQRYQVFSVPGSKSTATISGLKPGVEYQIRV




YAETGGADSDQSMGWIQIGYRTEGDKPSQHHHHHH





 99
    8.5
MGHHHHHHGGVSDVPRDLEVVAATPTSLLISWDAPA




VTVHSYHIQYWPLGSYQRYQVFSVPGSKSTATISGLK




PGVEYQIRVYAETGGADSDQSFGWIQIGYRTPES





100
    6
MASTSGSSSYLMPSDLEVVAATPTSLYIHWYPIASTIIN




FRITYGETGGNSPVQEFTVPGSQVHATISGLKPGVDYT




ITVYAVHYEHKYSELWMGHPISINYRTEGSGSHHHHH




H





101
 >400
MASTSGSASYLIPSDLEVVAATPTSLSIYWYPVASTIIN




FRITYVETGGNSPVQEFTVPGSKSTATISGLKPGVDYTI




TVYAVHYEQKYSEYWIGHPISINYRTEGSGSHHHHHH





102
>5500
MASTSGSSPYLMPYDLEVVAATPTSLFIRWYGSASSIV




KFRITYGETGGNSPVQEFTVGGTQLHATISGLKPGVD




YTITVYAVHFEHKYSELWIGHPISINYRTEGSGSHHHH




RH





103
  255
MASTSGYTSYPIPYDLEVVAATPTSLYIHWYWIAATII




SFRITYGETGGNSPVQEFTVPAGQDHATISGLKPGVDY




TITVYAVHYEEEYSEFWTGHPISINYRTEGSGSHHHHH




H





104
  500
MASTSGTHWFYSIPHDLEVVAATPTSLTIAWEPPHHT




AMGYRITYGETGGNSPVQEFTVPGGYTTAYISGLKPG




VDYTITVYAAYYEREYSEHWISHPISINYRTEGSGSHH




HHHH





105
  300
MASTSGEFYHTKYPYDLEVVAATPTSLEISWRSPTRD




WQWFRITYGETGGNSPVQEFTVAGPYRNAIISGLKPG




VDYTITVYADVYMPSEGGLVVDTYHPISINYRTEGSG




SHHHHHH





106
 2400
MASTSGQAYPEYYFVDLEVVAATPTSLLISWSKPYYN




AYSYRITYGETGGNSPVQEFTVLGHDTRAVISGLKPG




VDYTITVYAMFIEYIDQEIWHAHPISINYRTEGSGSHH




HHHH





107
  500
MGVSDVPRDLEVVAATPTSLLISWDEHTDIYRYYRIT




YGETGGNSPVQEFTVPAMEHTATISGLKPGVDYTITV




YAVTHVYPIMIHQYPISINYRTEIDKPSQHHHHHH





108
  170
MASTSGVSDVPRDLEVVAATPTSLLISWDAPAVTVLE




YQIDYHPAAVWHALQRFTVPGSKSTATISGLKPGVHY




KISVTATTHADNESIMWHPISIYYRTEGSGSHHHHHH





109
  320
MGVSDVPRDLEVVAATPTSLLISWDYPTVTPRYYRIT




YGETGGNSPVQEFTVPEYIGTATISGLKPGVDYTITVY




AVTNDTTIYSISRPISINYRTEIDKPSQHHHHHH





110
   28.6
MGHHHHHHGGVSDVPRDLEVVAATPTSLLISWDAPA




VTVHSYHIQYWPLGSYQRYQVFSVPGSKSTATISGLK




PGVEYQIRVYAETGRGESDQSLGWIQIGYRTEES





111
   20.0
MGHHHHHHGGVSDVPRDLEVVAATPTSLLISWDAPA




VTVHSYHIQYWPLGSYQRYQVFSVPGSKSTATISGLK




PGVEYQIRVYAETGRGESDQSFGWIQIGYRTEES





112
   18.4
MGHHHHHHGGVSDVPRDLEVVAATPTSLLISWDAPA




VTVHSYHIQYWPLGSYQRYQVFSVPGSKSTATISGLK




PGVEYQIRVYAETGRGESDQSLGWIQIGYRTPES





113
   15.4
MGHHHHHHGGVSDVPRDLEVVAATPTSLLISWDAPA




VTVHSYHIQYWPLGSYQRYQVFSVPGSKSTATISGLK




PGVEYQIRVYAETGRGESDQSFGWIQIGYRTPES





114
   12.6
MGHHHHHHGGVSDVPRDLEVVAATPTSLLISWDAPA




VTVHAYHIQYWPLGFYQGYQVFSVPGSKSTATISGLK




PGVEYQIRVYAETGLGDAHQSLGWIQIGYRTPES









In some embodiments, the anti-CD4 Adnectin™ of the invention comprises an amino acid sequence at least 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99% or 100% identical to any one of SEQ ID NOs: 95-114.


In some embodiments, the anti-CD4 Adnectin™ of the invention comprises an amino acid sequence at least 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99% or 100% identical to any one of SEQ ID NOs: 95-114, excluding any N-terminus extended region.


In some embodiments, the anti-CD4 Adnectin™ of the invention comprises an amino acid sequence at least 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99% or 100% identical to any one of SEQ ID NOs: 95-114, excluding any C-terminus extended region.


In some embodiments, the anti-CD4 Adnectin™ of the invention comprises an amino acid sequence at least 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99% or 100% identical to any one of SEQ ID NOs: 95-114, excluding both the N-terminus and C-terminus extended regions.


In other embodiments, anti-CD4 Adnectin™ comprises an amino acid sequence at least 80%, 85%, 90%, 95%, 98%, 99% or 100% identical to the CD loop and FG loop regions of SEQ ID NOs: 95-114.


Anti-N17 Adnectin™


The full length amino acid sequence of anti-N17 Adnectin™ protein of the invention include but are not limited to those listed in Table 3 below. Table 3 also lists the antiviral EC50 values for each anti-N17 Adnectin™.









TABLE 3







Anti-N17 Adnectin ™









SEQ ID
Antiviral



NO
EC50 (nM)
Anti-N17 Adnectin ™ Protein Sequence












115
130
MGVSDVPRDLEVVAATPTSLLISWEYKVNNYRYYRITY




GETGGNSPVQEFTVPSVLSTATISGLKPGVDYTITVYAV




TYGVHSSPISINYRTEIDKPSQHHHHHH





116
50
MASTSGVSDVPRDLEVVAATPTSLLISWDAPAVTVEQY




YIAYYVEGEPSSYQYFRVPGSKSTATISGLKPGVLYHIY




VNAVTGSGLRPEFSLPIRIKYRTEGSGSHHHHHH





117
42
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDDSAPISINYRTEID





118
28
MGVSDVPRDLEVVAATPTSLLISWKYKVHPYRYYRITY




GETGGNSPVQEFTVPSVLSTATISGLKPGVDYTITVYAV




TYGVNSLPISINYRTEIDKPSQHHHHHH





119
>1,000
MGVSDVPRDLEVVAATPTSLLISWDAPAVTVGWYHIGY




NVEGEPASYQYFRVPGSKSTATISGLKPGVEYMIFVNAV




TGSGAREEFSLPISINYRTEGSGSHHHHHH





120
39
MGHHHHHHGGVSDVPRDLEVVAATPTSLLISWEYNVN




PYRYYRITYGETGGNSPVQEFTVPSVLSSAQISGLKPGV




DYTITVYAVTRGVDSAPISINYRTPGG





121
57
MGHHHHHHGGVSDVPRDLEVVAATPTSLLISWEYNVN




PYRYYRITYGETGGNSPVQEFTVPSVLSTAEISGLKPGV




DYTITVYAVTYGVDSDPISINYRTPGG





122
6
MGHHHHHHGGVSDVPRDLEVVAATPTSLLISWQYKVH




PYRYYRITYGETGGNSPVQEFTVPSVLSTAEISGLKPGV




DYTITVYAVTRGVDSAPISINYRTPGG





123
28
MGVSDVPRDLEVVAATPTSLLISWEYKVHPYRYYRITY




GETGGNSPVQEFTVPSVLSTATISGLKPGVDYTITVYAV




TRGVDSAPISINYRTEIDKPSQHHHHHH





124
6
MGVSDVPRDLEVVAATPTSLLISWEYKVHPYRYYRITY




GETGGNSPVQEFTVPSVLSTAEISGLKPGVDYTITVYAV




TRGVDSAPISINYRTPIDKPSQHHHHHH





125
4342
MGHHHHHHGGVSDVPRDLEVVAATPTSLLISWEYKVH




PYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPGV




DYTITVYAVTYGVQSDPISINYRTPGG





126
8
MGVSDVPRDLEVVAATPTSLLISWEYKVHPYRYYRITY




GETGGNSPVQEFTVPSVLSTATISGLKPGVDYTITVYAV




TYGIQSPPISINYRTEIDKPSQHHHHHH





127
9
MGHHHHHHGGVSDVPRDLEVVAATPTSLLISWQYKVH




PYRYYRITYGETGGNSPVQEFTVPSVLSSAQISGLKPGV




DYTITVYAVTYGIESSPISINYRTPGG





128
18
MGHHHHHHGGVSDVPRDLEVVAATPTSLLISWEYKVH




PYRYYRITYGETGGNSPVQEFTVPSVLSSAEISGLKPGVD




YTITVYAVTYGIDSSPISINYRTPGG





129
11
MGVSDVPRDLEVVAATPTSLLISWEYKVHPYRYYRITY




GETGGNSPVQEFTVPSVLSSAEISGLKPGVDYTITVYAV




TYGIDSPPISINYRTEIDKPSQHHHHHH





130
42
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYNV




HYDRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTDGVHSSPISINYRTEID





131
5
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDLSAPISINYRTEID





132
5
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWDAPA




VTVEEYYIGYYVEFEPSSYQWFTVPGSKSTATISGLKPG




VEYSIYVNAVTGMGMQPEMSLPISINYRTEGS





133
42
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HYDRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGVDSDPISINYRTEID





134
5
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWDAPA




VTVEQYYIAYYDEKEPSSYQYFRVPGSKSTATISGLKPG




VEYAIFVNAVTRSGVLPEFSLPISINYRTEGS





135
>1,000
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWKYNV




NAYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGVHSSPISINYRTEID





136
5
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWDAPA




VVTVEQYYIGYYVEAEPSSYQYFFVPGSKSTATISGLKPG




VDYAIFVNAVTASGRGPEYSLPISINYRTEGS





137
80
MGVSDVPRDLEVVAATPTSLLISWHDGIGEERYYRITYG




ETGGNSPVQEFTVPMDDITATISGLKPGVDYTITVYAVT




VGDVISVLHEPISINYRTEIDKPSQHHHHHH





138
3300
MGVSDVPRDLEVVAATPTSLLISWHYPFEGYVTYYRIT




YGETGGNSHVQEFTVPVGYTTATISGLKPGVDYTITVYA




VTSSKGYVYFPISINYRTEIDKPSQHHHHHH





139
130
MGVSDVPRDLEVVAATPTSLLISWEDPEAAVRYYRITY




GETGGNSPVQEFTVPINDLHSYLSGLKPGVDYTITVYAV




TEATVMYVLDEPISINYRTEIDKPSQHHHHHH





140
950
MGVSDVPRDLEVVAATPTSLLISWDLLEDMSRYYRITY




GETGGNSPVQEFTVPTDAYTATISGLKPGVDYTITVYAV




TQDSHVIELSYPISINYRTEIDKPSQHHHHHH





141
3
MGVSDVPRDLEVVAATPTSLLISWQYKVHPYRYYRITY




GETGGNSPVQEFTVPSVLSTATISGLKPGVDYTITVYAV




TYGIQSPPISINYRTEIDKPSQHHHHHH





142
55
MGVSDVPRDLEVVAATPTSLLISWRYRVHPYRYYRITY




GETGGNSPVQEFTVPSVLSTATISGLKPGVDYTITVYAV




TDGVQSSPISINYRTEIDKPSQHHHHHH





143
20
MGVSDVPRDLEVVAATPTSLLISWEYNVNPYRYYRITY




GETGGNSPVQEFTVPSVLSTATISGLKPGVDYTITVYAV




TYGIESSPISINYRTEIDKPSQHHHHHH





144
20
MGVSDVPRDLEVVAATPTSLLISWQYKVHPYRYYRITY




GETGGNSPVQEFTVPSVLSTATISGLKPGVDYTITVYAV




TYGVESSPISINYRTEIDKPSQHHHHHH





145
11
MGVSDVPRDLEVVAATPTSLLISWEYKVHPYRYYRITY




GETGGNSPVQEFTVPSVLSTATISGLKPGVDYTITVYAV




TYGVNSLPISINYRTEIDKPSQHHHHHH





146
11
MGVSDVPRDLEVVAATPTSLLISWEYKVHPYRYYRITY




GETGGNSPVQEFTVPSVLSTATISGLKPGVDYTITVYAV




TYGIDSPPISINYRTEIDKPSQHHHHHH





147
28
MGVSDVPRDLEVVAATPTSLLISWEYKVHPYRYYRITY




GETGGNSPVQEFTVPSVLSTATISGLKPGVDYTITVYAV




TYGVDSDPISINYRTEIDKPSQHHHHHH





148
14
MGVSDVPRDLEVVAATPTSLLISWEYNVNPYRYYRITY




GETGGNSPVQEFTVPSVLSTATISGLKPGVDYTITVYAV




TYGIDSPPISINYRTEIDKPSQHHHHHH





149
3
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGKDSAPISINYRTEID





150
18
MGVSDVPRDLEVVAATPTSLLISWEYNVNPYRYYRITY




GETGGNSPVQEFTVPSVLSTATISGLKPGVDYTITVYAV




TYGVESSPISINYRTEIDKPSQHHHHHH





151
11
MGVSDVPRDLEVVAATPTSLLISWEYKVHPYRYYRITY




GETGGNSPVQEFTVPSVLSTATISGLKPGVDYTITVYAV




TYGIDSSPISINYRTEIDKPSQHHHHHH





152
20
MGVSDVPRDLEVVAATPTSLLISWEYNVNPYRYYRITY




GETGGNSPVQEFTVPSVLSTATISGLKPGVDYTITVYAV




TYGVQSDPISINYRTEIDKPSQHHHHHH





153
8
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTAQISGLKPG




VDYTITVYAVTYGDDSAPISINYRTEID





154
8
MGVSDVPRDLEVVAATPTSLLISWEYNVNPYRYYRITY




GETGGNSPVQEFTVPSVLSTATISGLKPGVDYTITVYAV




TYGVNSLPISINYRTEIDKPSQHHHHHH





155
9
MGVSDVPRDLEVVAATPTSLLISWQYKVHPYRYYRITY




GETGGNSPVQEFTVPSVLSTATISGLKPGVDYTITVYAV




THGVHSAPISINYRTEIDKPSQHHHHHH





156
43
MGVSDVPRDLEVVAATPTSLLISWEYKVNPWRYYRITY




GETGGNSPVQEFTVPSVLSTATISGLKPGVDYTITVYAV




TYGIESSPISINYRTEIDKPSQHHHHHH





157
43
MGVSDVPRDLEVVAATPTSLLISWQYKVHPYRYYRITY




GETGGNSPVQEFTVPSVLSTATISGLKPGVDYTITVYAV




TYGVNSIPISINYRTEIDKPSQHHHHHH





158
85
MGVSDVPRDLEVVAATPTSLLISWEYKVHYDRYYRITY




GETGGNSPVQEFTVPSVLSTATISGLKPGVDYTITVYAV




TYGIQSPPISINYRTEIDKPSQHHHHHH





159
23
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDDSAPISIPYRTPGG





160
58
MGVSDVPRDLEVVAATPTSLLISWEYKVDPYRYYRITY




GETGGNSPVQEFTVPSVLSTATISGLKPGVDYTITVYAV




TYGVESSPISINYRTEIDKPSQHHHHHH





161
13
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSSAEISGLKPGV




DYTITVYAVTYGDDSAPISINYRTEID





162
35
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSSAQISGLKPG




VDYTITVYAVTYGDDSAPISINYRTEID





163
35
MGHHHHHHGGSVSDVPRYLEVVAATPTSLLISWQYKV




HPYRWYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGVNSIPISINYRTEID





164
35
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWKYQV




HAYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGVYSAPISINYRTEID





165
42
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




NPYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGVHSSPISINYRTEID





166
50
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWKYNL




HPYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGIISEPISINYRTEID





167
42
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYRV




NPYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGVQSPPISINYRTEID





168
42
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




NAYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGVLSPPISINYRTEID





169
5
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYNV




NPYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTRGVDSAPISINYRTEID





170
5
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKVS




PYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPGV




DYTITVYAVTFGIRSSPISINYRTEID





171
35
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWKYQV




HAYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGIISEPISINYRTEID





172
5
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




DPYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGIDSSPISINYRTEID





173
5
MGHHHHHHGGAVSDVPRDLEVVAATPTSLLISWEYKV




NPWRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGVHSSPISINYRTEID





174
5
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYNV




NAYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGIISEPISINYRTEID





175
5
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HYDRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGVQSTPISINYRTEID





176
>1,000
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HHDRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGIESSPISINYRTEID





177
252
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDDSAPISIPYRTPGG





178
631
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGXDSXPISINYRTEID





179
>1,000
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDDSRPISINYRTEID





180
>1,000
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGKDSAPISINYRTEID





181
631
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDHSAPISINYRTEID





182
>1,000
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDQSAPISINYRTEID





183
20
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGIDSAPISINYRTEID





184
20
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDVSAPISINYRTEID





185
252
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGXDSAPISINYRTEID





186
2830
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGEDSAPISINYRTEID





187
86
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFKVPSVLSTATISGLKPG




VDYTITVYAVTYGDDSAPISINYRTEID





188
86
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDLSAPISINYRTEID





189
>1,000
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGKDSAPISINYRTEID





190
>1,000
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGADSAPISINYRTEID





191
>1,000
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGTDSAPISINYRTEID





192
2830
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGTDSAPISINYRTEID





193
252
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGKDSAPISINYRTEID





194
>1,000
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDPSAPISINYRTEID





195
>1,000
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDDSQPISINYRTEID





196
86
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDVSAPISINYRTEID





197
81
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDDSGPISINYRTEID





198
2830
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDDSAPISIQYRTEID





199
>1,000
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDDSQPISINYRTEID





200
>1,000
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYTETGGNSPVQEFTVPSVLSTATISGLKPGV




DYTITVYAVTYGDDSAPISINYRTEID





201
20
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDQSAPISINYRTEID





202
>1,000
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDDSKPISINYRTEID





203
86
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDDSGPISINYRTEID





204
0
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYRETGGNSPVQEFTVPSVLSTATISGLKPGV




DYTITVYAVTYGDDSAPISINYRTEID





205
86
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYDV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDDSAPISINYRTEID





206
237
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDDSKPISINYRTEID





207
>1,000
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYWETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDDSAPISINYRTEID





208
>1,000
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDDSTPISINYRTEID





209
20
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDDSTPISINYRTEID





210
143
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDDSSPISINYRTEID





211
631
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTRGDDSAPISINYRTEID





212
86
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTHGDDSAPISINYRTEID





213
>1,000
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDDSWPISINYRTEID





214
>1,000
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTHGDDSAPISINYRTEID





215
>1,000
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWKYDV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDDSAPISINYRTEID





216
>1,000
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTATISDLKPG




VDYTITVYAVTYGDDSHPISINYRTEID





217
>1,000
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTRGDDSAPISINYRTEID





218
62
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTATIRGLKPG




VDYTITVYAVTYGDDSAPISINYRTEID





219
>1,000
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWKYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDDSAPISINYRTEID





220
86
MGHHHHHHCGSVSDVPRDLEVVAATPTSLLISWEYKV




AHNRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDDSAPISINYRTEID





221
>1,000
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




YGYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDDSAPISINYRTEID





222
>1,000
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




DHQRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDDSAPISINYRTEID





223
>1,000
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




DYRRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDDSAPISINYRTEID





224
>1,000
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




AYDRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDDSAPISINYRTEID





225
>1,000
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




TSYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPGV




DYTITVYAVTYGDDSAPISINYRTEID





226
631
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRIIHETGGNSPVQEFTVPSVLSTATISGLKPGVD




YTITVYAVTYGDDSAPISINYRTEID





227
>1,000
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDDSAPISINYRKEAEL





228
>1,000
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




NHQRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDDSAPISINYRTEID





229
>1,000
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




DHRRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDDSAPISINYRTEID





230
17
MGVSDVPRDLEVVAATPTSLLISWEYKVHPYRYYRITY




GETGGNSPVQEFTVPSVLSTATISGLKPGVDYTITVYAV




TYGVDSDPISINYRTDDKPSQHHHHHH





231
10
MGVSDVPRDLEVVAATPTSLLISWEYKVHPYRYYRITY




GETGGNSPVQEFTVPSVLSTATISGLKPGVDYTITVYAV




TYGVNSIPISINYRTEIDKPSQHHHHHH





232
47
MGVSDVPRDLEVVAATPTSLLISWEYKVNAYRYYRITY




GETGGNSPVQEFTVPSVLSTATISGLKPGVDYTITVYAV




TYGIESSPISINYRTEIDKPSQHHHHHH





233
55
MGVSDVPRDLEVVAATPTSLLISWEYKVHPYRYYRITY




GETGGNSPVQEFTVPSVLSTATISGLKPGVDYTITVYAV




TDGVQSSPISINYRTEIDKPSQHHHHHH





234
20
MGVSDVPRDLEVVAATPTSLLISWEYKVNAYRYYRITY




GETGGNSPVQEFTVPSVLSTATISGLKPGVDYTITVYAV




TYGIDSSPISINYRTEIDKPSQHHHHHH





235
29
MGVSDVPRDLEVVAATPTSLLISWEYKVHYDRYYRITY




GETGGNSPVQEFTVPSVLSTATISGLKPGVDYTITVYAV




TNGVLSSPISINYRTEIDKPSQHHHHHH





236
85
MGVSDVPRDLEVVAATPTSLLISWEYKVNPWRYYRITY




GETGGNSPVQEFTVPSVLSTATISGLKPGVDYTITVYAV




TRGVDSAPISINYRTEIDKPSQHHHHHH





237
31
MGVSDVPRDLEVVAATPTSLLISWEYKVHPXRYYRITY




GETGGNSPVQEFTVPSVLSTATISGLKPGVDYTITVYAV




TYGVBSXPISINYRTEIDKPSQHHHHHH





238
11
MGVSDVPRDLEVVAATPTSLLISWEYKVNPWRYYRITY




GETGGNSPVQEFTVPSVLSTATISGLKPGVDYTITVYAV




TYGIQSPPISINYRTEIDKPSQHHHHHH





239
4
MGVSDVPRDLEVVAATPTSLLISWQYKVHPYRWYRITY




GETGGNSPVQEFTVPSVLSTATISGLKPGVDYTITVYAV




TYGVNSIPISINYRTEIDKPSQHHHHHH





240
5
MGVSDVPRDLEVVAATPTSLLISWEYKVSPYRYYRITY




GETGGNSPVQEFTVPSVLSTATISGLKPGVDYTITVYAV




TYGVNSIPISINYRTEIDKPSQHHHHHH





241
8
MGVSDVPRDLEVVAATPTSLLISWEYKVNPWRYYRITY




GETGGNSPVQEFTVPSVLSTATISGLKPGVDYTITVYAV




TYGVNSLPISINYRTEIDKPSQHHHHHH





242
0
MGVSDVPRDLEVVAATPTSLLISWEYNVHYDRYYRITY




GETGGNSPVQEFTVPSVLSTATISGLKPGVDYTITVYAV




TYGVDSDPISINYRTEIDKPSQHHHHHH





243
48
MGVSDVPRDLEVVAATPTSLLISWEYKVNPWRYYRITY




GETGGNSPVQEFTVPSVLSTATISGLKPGVDYTITVYAV




TYGVDSDPISINYRTEIDKPSQHHHHHH





244
58
MGVSDVPRDLEVVAATPTSLLISWEYKVDPYRYYRITY




GETGGNSPVQEFTVPSVLSTATISGLKPGVDYTITVYAV




TYGVDSDPISINYRTEIDKPSQHHHHHH





245
50
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWKYQV




HAYRYYRITYGETGGNSPVQEFTVPSVLSTATINGLKPG




VDYTITVYAVTYGIISEPISINYRTEID





246
>1,000
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




NYNRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTEGVQSAPISINYRTEID





247
2
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDDSAPISINYRTPGG





248
>1,000
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNTPVQEFRVPSVLSTATISGLKPG




VDYTITVYAVTYADASAPISINYRTEID





249
83
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSSATLNGLKPG




VDYTITVYAVTYGDDSAPISINYRTEID





250
218
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSSAKISGLKPG




VDYTITVYAVTYGDDSAPISINYRTEID





251
186
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFNVPSVLSTATISGLKPG




VDYTITVYAVTYGDDSAPISINYRTEID





252
>1,000
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




AHRRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDDSAPISINYRTEID





253
>1,000
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




DAYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDDSAPISINYRTEID





254
>1,000
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




DSYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDDSAPISINYRTEID





255
136
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDDSAPISINYRTEID





256
170
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDASAPISINYRTEID





257
>1,000
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTNGDDSAPISINYRTEID





258
195
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSSATIRGLKPG




VDYTITVYAVTYGDDSAPISINYRTEID





259
135
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTAKISGLKPG




VDYTITVYAVTYGDDSAPISINYRTEID





260
188
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFRVPSVLSTATISGLKPG




VDYTITVYAVTYGDDSAPISINYRTEID





261
>1,000
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




APWRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDDSAPISINYRTEID





262
>1,000
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




DGWRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDDSAPISINYRTEID





263
>1,000
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




DTWRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDDSAPISINYRTEID





264
209
MGHHHHHHGGSVSDVPRDLEVVAATPTXLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDDSAPISINYRTEID





265
63
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGADSAPISINYRTEID





266
>1,000
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTRGDDSAPISINYRTEID





267
>1,000
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQDFTVPSVLSTATIRGLKPG




VDYTITVYAVTYGDDSAPISINYRTEID





268
2
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTANISGLKPG




VDYTITVYAVTYGDDSAPISINYRTEID





269
184
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQAFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDDSAPISINYRTEID





270
>1,000
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




APYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDDSAPISINYRTEID





271
>1,000
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




DGYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDDSAPISINYRTEID





272
>1,000
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




DTYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDDSAPISINYRTEID





273
>1,000
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYEV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDDSAPISINYRTEID





274
115
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYRV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDDSAPISINYRTEID





275
180
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQSFTVPSVLSTATISGLKPGV




DYTITVYAVTYGADSAPISINYRTEID





276
16
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTATIRGLKPG




VDYTITVYAVTYGDDSAPISINYRTEID





277
>1,000
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQDFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDDSAPISINYRTEID





278
>1,000
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




ARWRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDDSAPISINYRTEID





279
>1,000
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




DHQRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDDSAPISINYRTEID





280
>1,000
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




DYGRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDDSAPISINYRTEID





281
>1,000
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWKYGV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDDSAPISINYRTEID





282
56
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYSVH




PYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPGV




DYTITVYAVTYGDDSAPISINYRTEID





283
>1,000
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTDGDDSAPISINYRTEID





284
>1,000
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTSGDDSAPISINYRTEID





285
397
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSSATLRGLKPG




VDYTITVYAVTYGDDSAPISINYRTEID





286
>1,000
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




AAWRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDDSAPISINYRTEID





287
>1,000
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




ATWRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDDSAPISINYRTEID





288
>1,000
MGHHHHHHGGSVSDVPRDLEVVAATTTSLLISWEYKV




DYHRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDDSAPISINYRTEID





289
>1,000
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYGV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDDSAPISINYRTEID





290
138
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWKYXV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDDSAPISINYRTEID





291
>1,000
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTGGDDSAPISINYRTEID





292
52
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTATIHGLKPG




VDYTITVYAVTYGDDSAPISINYRTEID





293
>1,000
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTATLRGLKPG




VDYTITVYAVTYGDDSAPISINYRTEID





294
101
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSSATISGLKPGV




DYTITVYAVTYGDDSAPISINYRTEID





295
>1,000
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




AAYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDDSAPISINYRTEID





296
>1,000
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




ATYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDDSAPISINYRTEID





297
30
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




DPWRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDDSAPISINYRTEID





298
>1,000
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




DYQRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDDSAPISINYRTEID





299
>1,000
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWKYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDDSAPISINYRTEID





300
132
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYXV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDDSAPISINYRTEID





301
>1,000
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTHGDDFAPISINYRTEMLI





302
4
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSSATINGLKPG




VDYTITVYAVTYGDDSAPISINYRTEID





303
109
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFKVPSVLSTATISGLKPG




VDYTITVYAVTYGDDSAPISINYRTEID





304
>1,000
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




AHDRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDDSAPISINYRTEID





305
83
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




DPYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDDSAPISINYRTEID





306
>1,000
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




DYRRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDDSAPISINYRTEID





307
205
MGHHHHHHGGSVSDVPRDLEVVAATPTGLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDDSAPISINYRTEID





308
>1,000
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




NAWRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDDSAPISINYRTEID





309
>1,000
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTHGDDSAPISINYRTEID





310
62
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTATINGLKPG




VDYTITVYAVTYGDDSAPISINYRTEID





311
42
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTAEISGLKPG




VDYTITVYAVTYGDDSAPISINYRTEID





312
37
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFLVPSVLSTATISGLKPG




VDYTITVYAVTYGDDSAPISINYRTEID





313
>1,000
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




AHGRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDDSAPISINYRTEID





314
>1,000
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




AYKRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDDSAPISINYRTEID





315
>1,000
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




DRWRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDDSAPISINYRTEID





316
331
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYAV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDDSAPISINYRTEID





317
>1,000
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




NAYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDDSAPISINYRTEID





318
>1,000
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




NGYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDDSAPISINYRTEID





319
>1,000
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKVS




HGRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPGV




DYTITVYAVTYGDDSAPISINYRTEID





320
>1,000
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




TTWRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDDSAPISINYRTEID





321
55
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSSAZLSGLKPG




VDYTITVYAVTYGDDSAPISINYRTEID





322
96
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGTDSAPISINYRTEID





323
126
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDQSAPISINYRTEID





324
487
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDDSNPISINYRTEID





325
>1,000
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




NHDRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDDSAPISINYRTEID





326
>1,000
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKVS




HRRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPGV




DYTITVYAVTYGDDSAPISINYRTEID





327
>1,000
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




TYERYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPGV




DYTITVYAVTYGDDSAPISINYRTEID





328
63
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSSATXXGLKPG




VDYTITVYAVTYGDDSAPISINYRTEID





329
102
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDVSAPISINYRTEID





330
99
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDDSPPISINYRTEID





331
100
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDDSXPISINYRTEID





332
>1,000
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




NRWRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDDSAPISINYRTEID





333
>1,000
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKVS




PWRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPGV




DYTITVYAVTYGDDSAPISINYRTEID





334
>1,000
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




TYRRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDDSAPISINYRTEID





335
112
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDESAPISINYRTEID





336
171
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDDSDPISINYRTEID





337
>1,000
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDDSYPISINYRTEID





338
>1,000
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




NRYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDDSAPISINYRTEID





339
>1,000
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKVS




RWRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPGV




DYTITVYAVTYGDDSAPISINYRTEID





340
>1,000
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




XXYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDDSAPISINYRTEID





341
>1,000
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQSFTVPSVLSTATISGLKPGV




DYTITVYAVTYGDDSAPISINYRTEID





342
>1,000
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGIDSAPISINYRTEID





343
122
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDHSAPISINYRTEID





344
>1,000
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDDSEPISINYRTEID





345
>1,000
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDDSRPISINYRTEID





346
83
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDDSAPISIQYRTEID





347
>1,000
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




NYRRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDDSAPISINYRTEID





348
>1,000
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKVS




TWRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPGV




DYTITVYAVTYGDDSAPISINYRTEID





349
>1,000
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




YAYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDDSAPISINYRTEID





350
191
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQTFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDDSAPISINYRTEID





351
215
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQAFTVPSVLSTATISGLKPG




VDYTITVYAVTYGKDSAPISINYRTEID





352
77
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDLSAPISINYRTEID





353
19
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDDSGPISINYRTEID





354
>1,000
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKVS




AWRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPGV




DYTITVYAVTYGDDSAPISINYRTEID





355
>1,000
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKVS




TYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPGV




DYTITVYAVTYGDDSAPISINYRTEID





356
31
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTAELSGLKPG




VDYTITVYAVTYGDDSAPISINYRTEID





357
178
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDLSAPISINYRTEID





358
>1,000
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDDSTPISINYRTEID





359
>1,000
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKVS




AYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPGV




DYTITVYAVTYGDDSAPISINYRTEID





360
>1,000
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




TGYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDDSAPISINYRTEID





361
>1,000
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




YHGRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDDSAPISINYRTEID





362
121
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTAKLSGLKPG




VDYTITVYAVTYGDDSAPISINYRTEID





363
130
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGNDSAPISINYRTEID





364
89
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDPSAPISINYRTEID





365
>1,000
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKVS




GYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPGV




DYTITVYAVTYGDDSAPISINYRTEID





366
>1,000
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




YHNRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDDSAPISINYRTEID





367
300
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSSAQLSGLKPG




VDYTITVYAVTYGDDSAPISINYRTEID





368
62
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGTDSAPISINYRTEID





369
155
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDPSAPISINYRTEID





370
335
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGDDSKPISINYRTEID





371
114
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKV




HPYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPG




VDYTITVYAVTYGIDSTPISINYRTEID









In some embodiments, the anti-N17 Adnectin™ of the invention comprises an amino acid sequence at least 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99% or 100% identical to any one of SEQ ID NOs: 115-371.


In some embodiments, the anti-N17 Adnectin™ of the invention comprises an amino acid sequence at least 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99% or 100% identical to any one of SEQ ID NOs: 115-371, excluding any N-terminus extended region.


In some embodiments, the anti-N17 Adnectin™ of the invention comprises an amino acid sequence at least 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99% or 100% identical to any one of SEQ ID NOs: 115-371, excluding any C-terminus extended region.


In some embodiments, the anti-N17 Adnectin™ of the invention comprises an amino acid sequence at least 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99% or 100% identical to any one of SEQ ID NOs: 115-371, excluding both the N-terminus and C-terminus extended regions.


In other embodiments, anti-N17 Adnectin™ comprises an amino acid sequence at least 80%, 85%, 90%, 95%, 98%, 99% or 100% identical to the BC loop, DE loop and FG loop regions of SEQ ID NOs: 115-371.


Analysis of the sequences above indicate that S52, V53, L54 and S55 of the anti-N17 Adnectin™ DE loop are conserved amino acids. In some embodiments, the anti-N17 Adnectin™ comprises one or more of the conserved amino acids S52, V53, L54 and S55.


Additionally, analysis of the sequences above indicate that Y24 of the anti N17 Adnectin™ BC loop is a conserved amino acid. In some embodiments, position 26 of the anti-N17 Adnectin™ BC loop is valine or leucine.


Analysis of the sequences above indicate that G78 and S85 of the anti-N17 Adnectin™ FG loop are conserved amino acids. In some embodiments, the anti-N17 Adnectin™ comprises one or more of the conserved amino acids G78 and S85. In some embodiments, position 79 of the anti-N17 Adnectin™ FG loop is valine or isoleucine.


In some embodiments, the anti-N17 Adnectin™ comprises one or more of the conserved amino acids Y24, V26, L26, S52, V53, L54, S55 G78, V79, 179 and S85.


Point mutation analysis of the anti-N17 Adnectin™ showed advantages of mutating several 10Fn3 non-loop scaffold positions. Specifically, mutating positions T56 and T58 boosted potency. In some embodiments, the anti-N17 Adnectin™ comprises mutating T58 to Asn, Glu, or Gln.


II. HIV Fusion Peptide Inhibitors


The amino acid sequence of gp41, and its variation among different strains of HIV-1, is well known. The fusogenic domain (often called the fusion peptide, or FP) is believed to be involved in insertion into and disruption of the target cell membrane. The transmembrane domain, containing the transmembrane anchor sequence, is located toward the C-terminal end of the protein. Between the fusogenic domain and transmembrane anchor are two distinct regions, known as heptad repeat (HR) regions, each region having a plurality of heptads. The amino acid sequence comprising the HR1 region and the amino acid sequence comprising the HR2 region are each relatively conserved regions in the HIV-1 envelope protein. A representative sequence of the external domain of gp41 (clade B consensus) is as follows:










(SEQ ID NO: 2)



512 AVGIGAMFL GFLGAAGSTM GAASVTLTVQ ARQLLSGIVQ QQNNLLRAIE






561 AQQHLLQLTV WGIKQLQARV LAVERYLKDQ QLLGIWGCSG KLICTTAVPW





611 NASWSNKSLD EIWNNMTWME WEREIDNYTG LIYTLIEESQ NQQEKNEQEL





661 LELDKWASLW NWFDITNWLW YIK






The fusion peptide consists of approximately the first 23 amino acids, Ala512-Ser534. The HR1 region has a plurality of contiguous 7 amino acid residue stretches or “heptads” (the 7 amino acids in each heptad designated “a” through “g”), with a predominance of hydrophobic residues at the first (“a”) and fourth (“d”) positions which interact homotypically to form the core of the 3-helix bundle structure. Neutral polar amino acids such as Glutamine may also occupy these positions. One representative heptad begins with Leu545. A highly conserved portion of HR1 consists of the 17 residues from Leu565 to Leu581, and is called “N17”.


The C-terminal portion of gp41 comprises the HR2 region, which is believed to form an alpha helical structure during the fusion process, and bind into the grooves of the HR1 triple helical structure. HR2 also comprises heptads, though they do not interact homotypically but rather interact with amino acids from HR1. One representative heptad begins at Trp628.


HIV Fusion Peptide Inhibitor


The HIV fusion peptide inhibitor of the invention include but are not limited to the following sequences:














SEQ



ID


HIV Fusion Peptide Inhibitor Sequence
NO







SEYEARIEALIRAAQEQQEKNEAALRELYKWAL
372





SEYEARIEALIRAAQEQQEKNEAALRELWKWAS
373





TIAEYAARIEALIRAAQEQQEKNEAALRELYKWAS
374





ARIEEYAARIEALIRAAQEQQEKNEAALRELYKWAS
375





SEYEARIEALIRAAQEQQEKNEAALRELYKWAS
376





TEYEARIEALIRAAQEQQEKNEAALRELKEWASIWN
377





SEYEARIEALIRAAQEQQEKNEAALRELDKWTGVWGNYEKV
378





SRIEALIRAAQEQQEKNEAALRELFKWAS
379





SRIEALIRAAQEQQQKNEAALRELDKWAS
380





SRIEALIRAAQEQQEKNEAALRELYKWAS
381





SRIEALIRAAQEQQEKNEAALRELLKWAS
382





SRIEALIRAAQEQQEKNEAALRELQKWAS
383





SRIEALIRAAQEQQEKNEAALRELDKWAS
384





AIAEYAARIEALIRAAQEQQEKNEAALRELDKWAS
385





TEYEARIEALIRAAQEQQEKNEAALRELDK
386





SRIEALIRAAQEQQEKNEAALRELYKWTS
387





SRIEALIRAAQEQQEKNEAALRELYKWASLWI
388





SRIEALIRAAQEQQEKNEAALRELYKWASRWN
389





SRIEALIRAAQEQQEKNEAALRELYKWASSWN
390





SRIEALIRAAQEQQEKNEAALRELYKWGS
391





SRIEALIRAAQEQQEKNEAALRELDK
392









In some embodiments, the HIV fusion peptide inhibitor comprises an amino acid sequence at least 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99% or 100% identical to any one of SEQ ID NOs: 372-392.


Point mutations in the C-terminal region of the HIV fusion peptide inhibitor were seen to boost potency. In some embodiments, a hydrophobic replacement of the aspartic acid (D) near the C-terminus of the HIV fusion peptide inhibitor provided at least a 10× increase in potency (FIG. 8). In some embodiments, the HIV fusion peptide inhibitor comprises replacing “DK” with “YK”, “LK”, “FK” or “WK”.


Other point mutation studies in the C-terminal region showed how the C-terminal amino acids “WAS” can be mutated with good effects on potency. In some embodiments, the HIV fusion peptide inhibitor comprises replacing C-terminal amino acids “WAS” to “WFS” or “WAL”.


III. Linkers


The various components of the Combinectin of the invention may be covalently or non-covalently linked. In some embodiments, the PK moiety may be directly or indirectly linked to a Combinectin via a polypeptide linker.


Suitable linkers are those which allow the separate domains to fold independently of each other forming a three dimensional structure that permits high affinity binding to a target molecule.


The disclosure provides a number of suitable linkers that meet these requirements, including glycine-serine based linkers, glycine-proline based linkers. The Examples described herein demonstrate that the Combinectin domains joined via polypeptide linkers retain their target binding function. In some embodiments, the linker is a glycine-serine based linker. These linkers comprise glycine and serine residues and may be between 8 and 50, 10 and 30, and 10 and 20 amino acids in length. Examples include linkers having an amino acid sequence (GS)7 (SEQ ID NO: 393), G(GS)6 (SEQ ID NO: 394), and G(GS)7G (SEQ ID NO: 395). Other linkers contain glutamic acid, and include, for example, (GSE)5 (SEQ ID NO: 396) and GGSEGGSE (SEQ ID NO: 397). Other exemplary glycine-serine linkers include (GS)4 (SEQ ID NO: 398), (GGGGS)7 (SEQ ID NO: 399), (GGGGS)5 (SEQ ID NO: 400), (GGGGS)4 (SEQ ID NO: 401 (GGGGS)3G (SEQ ID NO: 402). In some embodiments, the linker is a glycine-proline based linker. These linkers comprise glycine and proline residues and may be between 3 and 30, 10 and 30, and 3 and 20 amino acids in length. Examples include linkers having an amino acid sequence (GP)3G (SEQ ID NO: 403) and (GP)5G (SEQ ID NO: 404). In other embodiments, the linker may be a proline-alanine based linker having between 3 and 30, 10 and 30, and 3 and 20 amino acids in length. Examples of proline alanine based linkers include, for example, (PA)3 (SEQ ID NO: 405), (PA)6 (SEQ ID NO: 406) and (PA)9 (SEQ ID NO: 407). In some embodiments, the linker is a glutamic acid-proline based linker. These linkers comprise glutamic acid and proline residues and may be between 3 and 30, 10 and 30, and 3 and 20 amino acids in length. Examples include linkers having an amino acid sequence ESPEPETPEDE (SEQ ID NO: 408) and (ESPEPETPED)2E (SEQ ID NO: 409). It is contemplated, that the optimal linker length and amino acid composition may be determined by routine experimentation by methods well known in the art.


Linkers or spacers, may be introduced at the N-terminus of the anti-CD4 moiety, the C-terminus of the anti-CD4 moiety, the N-terminus of the anti-gp41 moiety, the C-terminus of the anti-gp41 moiety, the N-terminus of the HIV fusion peptide inhibitor, or combinations thereof. In some embodiments the preferred linkers between the anti-CD4 Adnectin™ and anti-N17 Adnectin™ are short glutamine-proline rich linkers. In some embodiments the preferred linker between the anti-N17 Adnectin™ and HIV fusion peptide inhibitor are flexible glycine-serine rich linkers.


IV. Anti-N17 Adnectin™ Linked to a HIV Fusion Peptide Inhibitor Combinectin


The amino acid sequences of anti-N17 Adnectin™-HIV fusion peptide inhibitor Combinectin of the invention include but are not limited to those listed in Table 4 below. Table 4 also lists the antiviral EC50 values for each anti-N17 Adnectin™-HIV fusion peptide inhibitor Combinectin.









TABLE 4







Anti-N17 Adnectin TM - HIV Fusion


Peptide Inhibitor Combinectin









SEQ
Anti-



ID
viral
Anti-N17 Adnectin ™ - HIV Fusion


NO
EC50 (nM)
Peptide Inhibitor Protein Sequence












410
2
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKVN




AYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPGVD




YTITVYAVTYGVDSDPISINYRTEIDGGGGSGGGGSGGG




GSGGGGARIEEYAARIEALIRAAQEQQEKNEAALRELYK




WAS





411
4342
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKVN




AYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPGVD




YTITVYAVTYGVDSDPISINYRTEIDGGGGSGGGGSGGG




GSGGGGTIAEYAARIEALIRAAQEQQEKNEAALRELYK




WAS





412
2980
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKVN




AYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPGVD




YTITVYAVTYGVDSDPISINYRTEIDGGGGSGGGGSGGG




GSGGGGSEYEARIEALIRAAQEQQEKNEAALRELYKWAS





413
6
MGHHHHHHGVSDVPRDLEVVAATPTSLLISWKYKVHP




YRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPGVDY




TITVYAVTYGVNSLPISINYRTEIDGGGGSGGGGSGGGGS




GGGGTEYEARIEALIRAAQEQQEKNEAALRELKEWASI




WN





414
4
MGHHHHHHTSGVSDVPRDLEVVAATPTSLLISWDAPAV




TVGWYHIGYNVEGEPASYQYFRVPGSKSTATISGLKPGV




EYMIFVNAVTGSGAREEFSLPISINYRTEIDGGGGSGGGG




SGGGGSGGGGSEYEARIEALIRAAQEQQEKNEAALRELD




KWTGVWGNYEKV





415
>1,000
MGHHHHHHGVSDVPRDLEVVAATPTSLLISWDAPAVTV




RYYRITYGETGGNSPVQEFTVPGSKSTATISGLKPGVDYT




ITVYAVTGRGESPASSKPISINYRTEIDGGGGSGGGGSGG




GGSGGGGSRIEALIRAAQEQQEKNEAALRELFKWAS





416
>1,000
MGHHHHHHGVSDVPRDLEVVAATPTSLLISWDAPAVTV




RYYRITYGETGGNSPVQEFTVPGSKSTATISGLKPGVDYT




ITVYAVTGRGESPASSKPISINYRTEIDGGGGSGGGGSGG




GGSGGGGSRIEALIRAAQEQQQKNEAALRELDKWAS





417
428
MGHHHHHHGVSDVPRDLEVVAATPTSLLISWDAPAVTV




RYYRITYGETGGNSPVQEFTVPGSKSTATISGLKPGVDYT




ITVYAVTGRGESPASSKPISINYRTEIDGGGGSGGGGSGG




GGSGGGGSRIEALIRAAQEQQEKNEAALRELYKWAS





418
218
MGHHHHHHGVSDVPRDLEVVAATPTSLLISWDAPAVTV




RYYRITYGETGGNSPVQEFTVPGSKSTATISGLKPGVDYT




ITVYAVTGRGESPASSKPISINYRTEIDGGGGSGGGGSGG




GGSGGGGSRIEALIRAAQEQQEKNEAALRELLKWAS





419
>1,000
MGHHHHHHGVSDVPRDLEVVAATPTSLLISWDAPAVTV




RYYRITYGETGGNSPVQEFTVPGSKSTATISGLKPGVDYT




ITVYAVTGRGESPASSKPISINYRTEIDGGGGSGGGGSGG




GGSGGGGSRIEALIRAAQEQQEKNEAALRELQKWAS





420
8
MGHHHHHHGGSVSDVPRDLEVVAATPTSLLISWEYKVH




PYRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPGVD




YTITVYAVTYGDDSAPISINYRTEIDGGGGSGGGGSGGG




GSGGGGSRIEALIRAAQEQQEKNEAALRELDKWAS





421
50
MGHHHHHHGVSDVPRDLEVVAATPTSLLISWVAGAED




YQYYRITYGETGGNSPVQEFTVPHDLVTATISGLKPGVD




YTITVYAVTDMMHVEYTEHPISINYRTEIDGGGGSGGGG




SGGGGSGGAIAEYAARIEALIRAAQEQQEKNEAALRELD




KWAS





422
3
MGHHHHHHGVSDVPRDLEVVAATPTSLLISWKYKVHP




YRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPGVDY




TITVYAVTYGVNSLPISINYRTEIDGGGGSGGGGSGGGGS




GGGGTEYEARIEALIRAAQEQQEKNEAALRELDK





423
122
MGHHHHHHGVSDVPRDLEVVAATPTSLLISWDAPAVTV




RYYRITYGETGGNSPVQEFTVPGSKSTATISGLKPGVDYT




ITVYAVTGRGESPASSKPISINYRTEIDGGGGSGGGGSGG




GGSGGGGSRIEALIRAAQEQQEKNEAALRELYKWTS





424
39
MGHHHHHHGVSDVPRDLEVVAATPTSLLISWDAPAVTV




RYYRITYGETGGNSPVQEFTVPGSKSTATISGLKPGVDYT




ITVYAVTGRGESPASSKPISINYRTEIDGGGGSGGGGSGG




GGSGGGGSRIEALIRAAQEQQEKNEAALRELYKWASLWI





425
523
MGHHHHHHGVSDVPRDLEVVAATPTSLLISWDAPAVTV




RYYRITYGETGGNSPVQEFTVPGSKSTATISGLKPGVDYT




ITVYAVTGRGESPASSKPISINYRTEIDGGGGSGGGGSGG




GGSGGGGSRIEALIRAAQEQQEKNEAALRELYKWASRWN





426
139
MGHHHHHHGVSDVPRDLEVVAATPTSLLISWDAPAVTV




RYYRITYGETGGNSPVQEFTVPGSKSTATISGLKPGVDYT




ITVYAVTGRGESPASSKPISINYRTEIDGGGGSGGGGSGG




GGSGGGGSRIEALIRAAQEQQEKNEAALRELYKWASSWN





427
143
MGHHHHHHGVSDVPRDLEVVAATPTSLLISWDAPAVTV




RYYRITYGETGGNSPVQEFTVPGSKSTATISGLKPGVDYT




ITVYAVTGRGESPASSKPISINYRTEIDGGGGSGGGGSGG




GGSGGGGSRIEALIRAAQEQQEKNEAALRELYKWGS





428
50
MGHHHHHHGVSDVPRDLEVVAATPTSLLISWEYKVNN




YRYYRITYGETGGNSPVQEFTVPSVLSTATISGLKPGVDY




TITVYAVTVGVHSSPISINYRTEIDGGGGSGGGGSGGGGS




GGGGSRIEALIRAAQEQQEKNEAALRELDK









In some embodiments, the anti-N17 Adnectin™-HIV fusion peptide inhibitor Combinectin or fusion proteins thereof comprise an amino acid sequence at least 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99% or 100% identical to the non-linker regions of any one of SEQ ID NOs: 410-428.


In some embodiments, the anti-N17 Adnectin™-HIV fusion peptide inhibitor Combinectin or fusion proteins thereof comprise an amino acid sequence at least 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99% or 100% identical to the non-linker regions of any one of SEQ ID NOs: 410-428, excluding any N-terminus extended region.


In other embodiments, anti-N17 Adnectin™-HIV fusion peptide inhibitor Combinectin comprises an anti-N17 Adnectin™ comprising an amino acid sequence at least 80%, 85%, 90%, 95%, 98%, 99% or 100% identical to the BC loop, DE loop and FG loop regions of SEQ ID NOs: 410-428.


In other embodiments, the anti-N17 Adnectin™-HIV fusion peptide inhibitor Combinectin comprises a HIV fusion peptide inhibitor comprising an amino acid sequence at least 80%, 85%, 90%, 95%, 98%, 99% or 100% identical to the HIV fusion peptide inhibitor of SEQ ID NOs: 410-428.


V. Anti-CD4 Adnectin™ Linked to an Anti-N17 Adnectin™ Linked to a HIV Fusion Peptide Inhibitor Combinectin


The anti-CD4 Adnectin™ linked to an anti-N17 Adnectin™ linked to a HIV fusion peptide inhibitor Combinectin of the invention include but are not limited to the following sequences:









Combinectin 3137


(SEQ ID NO: 3)



GVSDVPRDLEVVAATPTSLLISWDAPAVTVHSYHIQYWPLGSYQRYQ







VFSVPGSKSTATISGLKPGVEYQIRVYAETGRGESDQSLGWIQIGYR







TE
ESPEPETPEDE
GVSDVPRDLEVVAATPTSLLISWQYKVHPYRYYR







ITYGETGGNSPVQEFTVPSVLSTAEISGLKPGVDYTITVYAVTRGVD







SAPISINYRTP
GGGGSGGGGSGGGGSGGGG
SEYEARIEALIRAAQEQ







QEKNEAALRELYKWAL






Combinectin 3151


(SEQ ID NO: 5)



GVSDVPRDLEVVAATPTSLLISWDAPAVTVHSYHIQYWPLGSYQRYQ







VFSVPGSKSTATISGLKPGVEYQIRVYAETGRGESDQSLGWIQIGYR







TE
ESPEPETPEDE
GVSDVPRDLEVVAATPTSLLISWEYNVNPYRYYR






ITYGETGGNSPVQEFTVPSVLSSAQISGLKPGVDYTITVYAVTRGVD






SAPISINYRTP
GGGGSGGGGSGGGGSGGGG
SEYEARIEALIRAAQEQ







QEKNEAALRELWKWAS






Combinectin 3191


(SEQ ID NO: 7)



GVSDVPRDLEVVAATPTSLLISWDAPAVTVHSYHIQYWPLGSYQRYQ







VFSVPGSKSTATISGLKPGVEYQIRVYAETGRGESDQSLGWIQIGYR







TP
ESPEPETPEDE
GVSDVPRDLEVVAATPTSLLISWEYKVHPYRYYR







ITYGETEGGNSPVQEFTVPSVLSSAEISGLKPGVDYTITVYAVTYGI







DSPPISINYRTE
GGGGSGGGGSGGGGSGGGG
SEYEARIEALIRAAQE







QQEKNEAALRELYKWAL






Combinectin 3202


(SEQ ID NO: 9)



GVSDVPRDLEVVAATPTSLLISWDAPAVTVHSYHIQYWPLGSYQRYQ







VFSVPGSKSTATISGLKPGVEYQIRVYAETGGADSDQSFGWIQIGYR







TP
ESPEPETPEDE
GVSDVPRDLEVVAATPTSLLISWEYKVHPYRYYR







ITYGETGGNSPVQEFTVPSVLSTAEISGLKPGVDYTITVYAVTRGVD







SAPISINYRTP
GGGGSGGGGSGGGGSGGGG
TIAEYAARIEALIRAAQ







EQQEKNEAALRELYKWAS







In the sequences above, the anti-CD4 Adnectin™ sequences are underlined. The anti-N17 Adnectin™ sequences are double underlined. The HIV fusion peptide inhibitor sequences are underlined in bold. The linker sequences are italicized.


In some embodiments, the Combinectin or fusion proteins thereof comprises an amino acid sequence at least 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99% or 100% identical to any one of SEQ ID NOs: 3, 5, 7 and 9.


In some embodiments, the Combinectin or fusion proteins thereof comprises an amino acid sequence at least 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99% or 100% identical to the non-linker regions of any one of SEQ ID NOs: 3, 5, 7 and 9.


In other embodiments, the Combinectin comprises an anti-CD4 Adnectin™ comprising an amino acid sequence at least 80%, 85%, 90%, 95%, 98%, 99% or 100% identical to the CD loop and FG loop regions of SEQ ID NOs: 3, 5, 7 and 9.


In other embodiments, the Combinectin comprises an anti-N17 Adnectin™ comprising an amino acid sequence at least 80%, 85%, 90%, 95%, 98%, 99% or 100% identical to the BC loop, DE loop and FG loop regions of SEQ ID NOs: 3, 5, 7 and 9.


In other embodiments, the Combinectin comprises a HIV fusion peptide inhibitor comprising an amino acid sequence at least 80%, 85%, 90%, 95%, 98%, 99% or 100% identical to the HIV fusion peptide inhibitor of SEQ ID NOs: 3, 5, 7 and 9.


VI. Pharmacokinetic Moieties


In one aspect, the application provides for an anti-CD4 moiety, an anti-gp41 moiety, a HIV fusion peptide inhibitor and combinations thereof further comprising a pharmacokinetic (PK) moiety. Improved pharmacokinetics may be assessed according to the perceived therapeutic need. Often it is desirable to increase bioavailability and/or increase the time between doses, possibly by increasing the time that a protein remains available in the serum after dosing. In some instances, it is desirable to improve the continuity of the serum concentration of the protein over time (e.g., decrease the difference in serum concentration of the protein shortly after administration and shortly before the next administration). The Combinectin of the invention may be attached to a moiety that reduces the clearance rate of the polypeptide in a mammal (e.g., mouse, rat, or human) by at least two-fold, at least 10-fold, at least 20-fold, at least 30-fold, at least 40-fold, at least 50-fold, at least 60-fold, at least 70-fold, at least 80-fold, at least 90-fold, at least 100-fold relative to the unmodified Combinectin. Other measures of improved pharmacokinetics may include serum half-life, which is often divided into an alpha phase and a beta phase. Either or both phases may be improved significantly by addition of an appropriate moiety. For example, the PK moiety may increase the serum half-life of the polypeptide by more than 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, 120%, 150%, 200%, 400%, 600%, 800%, 1000% or more relative to the Combinectin alone.


Moieties that slow clearance of a protein from the blood, herein referred to as “PK moieties”, include polyoxyalkylene moieties (e.g., polyethylene glycol), sugars (e.g., sialic acid), and well-tolerated protein moieties (e.g., Fc and fragments and variants thereof, transferrin, or serum albumin). The Combinectin of the invention may also be fused to albumin or a fragment (portion) or variant of albumin as described in U.S. Publication No. 2007/0048282, or may be fused to one or more Adnectins™ or other moieties that bind serum albumin, Fc, or transferrin.


Other PK moieties that can be used in the invention include those described in Kontermann et al., (Current Opinion in Biotechnology, 22:868-876 (2011)), herein incorporated by reference. Such PK moieties include, but are not limited to, human serum albumin fusions, human serum albumin conjugates, human serum albumin binders (e.g., Adnectin™ PKE, AlbudAb, ABD), XTEN fusions, PAS fusions (i.e., recombinant PEG mimetics based on the three amino acids proline, alanine, and serine), carbohydrate conjugates (e.g., hydroxyethyl starch (HES)), glycosylation, polysialic acid conjugates, and fatty acid conjugates.


Accordingly, in some embodiments the invention provides a Combinectin fused to a PK moiety that is a polymeric sugar. In some embodiments, the PK moiety is a polyethylene glycol moiety or an Fc region. In some embodiments the PK moiety is a serum albumin binding protein such as those described in U.S. Publication Nos. 2007/0178082 and 2007/0269422. In some embodiments the PK moiety is human serum albumin. In some embodiments, the PK moiety is transferrin.


The Fc fusion-Combinectin of the invention include the sequences shown in 3137 (SEQ ID NO: 4) and 3151 (SEQ ID NO: 6) of FIG. 2.


In some embodiments, the Fc fusion-Combinectin or fusion proteins thereof comprises an amino acid sequence at least 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99% or 100% identical to any one of SEQ ID NOs: 4 and 6.


In some embodiments, the Fc fusion-Combinectin or fusion proteins thereof comprises an amino acid sequence at least 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99% or 100% identical to the non-linker regions of any one of SEQ ID NOs: 4 and 6.


In other embodiments, the Fc fusion-Combinectin comprises an anti-CD4 Adnectin™ comprising an amino acid sequence at least 80%, 85%, 90%, 95%, 98%, 99% or 100% identical to the CD loop and FG loop regions of SEQ ID NOs: 4 and 6.


In other embodiments, the Fc fusion-Combinectin comprises an anti-N17 Adnectin™ comprising an amino acid sequence at least 80%, 85%, 90%, 95%, 98%, 99% or 100% identical to the BC loop, DE loop and FG loop regions of SEQ ID NOs: 4 and 6.


In other embodiments, the Fc fusion-Combinectin comprises a fusion peptide inhibitor comprising an amino acid sequence at least 80%, 85%, 90%, 95%, 98%, 99% or 100% identical to the HIV fusion peptide inhibitor of SEQ ID NOs: 4 and 6.


The HSA fusion-Combinectin of the invention include the sequences shown in 3191 (SEQ ID NO: 8) and 3202 (SEQ ID NO: 10) of FIG. 2.


In some embodiments, the HSA fusion—Combinectin or fusion proteins thereof comprises an amino acid sequence at least 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99% or 100% identical to any one of SEQ ID NOs: 8 and 10.


In some embodiments, the HSA fusion-Combinectin or fusion proteins thereof comprises an amino acid sequence at least 70%, 75%, 80%, 85%, 90%, 95%, 98%, 99% or 100% identical to the non-linker regions of any one of SEQ ID NOs: 8 and 10.


In other embodiments, the HSA fusion-Combinectin comprises an anti-CD4 Adnectin™ comprising an amino acid sequence at least 80%, 85%, 90%, 95%, 98%, 99% or 100% identical to the CD loop and FG loop regions of SEQ ID NOs: 8 and 10.


In other embodiments, the HSA fusion-Combinectin comprises an anti-N17 Adnectin™ comprising an amino acid sequence at least 80%, 85%, 90%, 95%, 98%, 99% or 100% identical to the BC loop, DE loop and FG loop regions of SEQ ID NOs: 8 and 10.


In other embodiments, the HSA fusion-Combinectin comprises a fusion peptide inhibitor comprising an amino acid sequence at least 80%, 85%, 90%, 95%, 98%, 99% or 100% identical to the HIV fusion peptide inhibitor of SEQ ID NOs: 8 and 10.


Polyethylene Glycol


In some embodiments, the anti-CD4 moiety, anti-gp41 moiety, HIV fusion peptide inhibitor and combinations thereof comprises polyethylene glycol (PEG). PEG is a well-known, water soluble polymer that is commercially available or can be prepared by ring-opening polymerization of ethylene glycol according to methods well known in the art (Sandler et al., Polymer Synthesis, Vol. 3, pp. 138-161, Academic Press, New York). The term “PEG” is used broadly to encompass any polyethylene glycol molecule, without regard to size or to modification at an end of the PEG, and can be represented by the formula: X—O(CH2CH2O)n-1CH2CH2OH, where n is 20 to 2300 and X is H or a terminal modification, e.g., a C1-4 alkyl. PEG can contain further chemical groups which are necessary for binding reactions, which result from the chemical synthesis of the molecule; or which act as a spacer for optimal distance of parts of the molecule. In addition, such a PEG can consist of one or more PEG side-chains which are linked together. PEGs with more than one PEG chain are called multiarmed or branched PEGs. Branched PEGs are described in, for example, European Published Application No. 473084A and U.S. Pat. No. 5,932,462.


One or more PEG molecules may be attached at different positions on the protein, and such attachment may be achieved by reaction with amines, thiols or other suitable reactive groups. The amine moiety may be, for example, a primary amine found at the N-terminus of a polypeptide or an amine group present in an amino acid, such as lysine or arginine. In some embodiments, the PEG moiety is attached at a position on the polypeptide selected from the group consisting of: a) the N-terminus; b) between the N-terminus and the most N-terminal beta strand or beta-like strand; c) a loop or strand residue positioned on a face of the polypeptide opposite the target-binding site; d) between the C-terminus and the most C-terminal beta strand or beta-like strand; e) within a linker sequence connecting two binding domains, and f) at the C-terminus.


PEGylation may be achieved by site-directed PEGylation, wherein a suitable reactive group is introduced into the protein to create a site where PEGylation preferentially occurs. In some embodiments, the protein is modified to introduce a cysteine residue at a desired position, permitting site-directed PEGylation on the cysteine. Mutations may be introduced into a protein coding sequence to generate cysteine residues. This might be achieved, for example, by mutating one or more amino acid residues to cysteine. Preferred amino acids for mutating to a cysteine residue include serine, threonine, alanine and other hydrophilic residues. Preferably, the residue to be mutated to cysteine is a surface-exposed residue. Algorithms are well-known in the art for predicting surface accessibility of residues based on primary sequence or a protein. Alternatively, surface residues may be predicted by comparing the amino acid sequences of binding polypeptides, given that the crystal structure of the framework, based on which binding polypeptides are designed and evolved, has been solved (see Himanen et al., Nature, 414:933-938 (2001)) and thus the surface-exposed residues identified. PEGylation of cysteine residues may be carried out using, for example, PEG-maleimide, PEG-vinylsulfone, PEG-iodoacetamide, or PEG-orthopyridyl disulfide.


The PEG is typically activated with a suitable activating group appropriate for coupling to a desired site on the polypeptide. PEGylation methods are well-known in the art and further described in Zalipsky, S. et al., “Use of Functionalized Poly(Ethylene Glycols) for Modification of Polypeptides” in Harris, J. M., Polyethylene Glycol Chemistry: Biotechnical and Biomedical Applications, Plenus Press, New York (1992), and in Zalipsky, Advanced Drug Reviews, 16:157-182 (1995).


PEG may vary widely in molecular weight and may be branched or linear. Typically, the weight-average molecular weight of PEG is from about 100 Daltons to about 150,000 Daltons. Exemplary weight-average molecular weights for PEG include about 20,000 Daltons, about 40,000 Daltons, about 60,000 Daltons and about 80,000 Daltons. In certain embodiments, the molecular weight of PEG is 40,000 Daltons. Branched versions of PEG having a total molecular weight of any of the foregoing can also be used. In some embodiments, the PEG has two branches. In other embodiments, the PEG has four branches. In another embodiment, the PEG is a bis-PEG (NOF Corporation, DE-200MA), to which two Adnectins™ are conjugated.


Conventional separation and purification techniques known in the art can be used to purify PEGylated Adnectin™, such as size exclusion (e.g., gel filtration) and ion exchange chromatography. Products may also be separated using SDS-PAGE. Products that may be separated include mono-, di-, tri-, poly- and un-PEGylated Adnectins™, as well as free PEG. The percentage of mono-PEG conjugates can be controlled by pooling broader fractions around the elution peak to increase the percentage of mono-PEG in the composition. About 90% mono-PEG conjugates represent a good balance of yield and activity.


In some embodiments, the PEGylated anti-CD4 moiety, anti-gp41 moiety, HIV fusion peptide inhibitor and combinations thereof will preferably retain at least about 25%, 50%, 60%, 70%, 80%, 85%, 90%, 95% or 100% of the biological activity associated with the unmodified anti-CD4 moiety, anti-gp41 moiety, Adnectin™ or Combinectin. In some embodiments, biological activity refers to its ability to bind to CD4 or gp41, as assessed by Kd, kon, or koff. In some embodiments, the PEGylated anti-CD4 moiety, anti-gp41 moiety, Adnectin™ or Combinectin thereof shows an increase in binding to CD4 or gp41 relative to unPEGylated anti-CD4 moiety, anti-gp41 moiety, Adnectin™ or Combinectin.


Immunoglobulin Fc Domain (and Fragments)


In some embodiments, the peptide of the invention is fused to an immunoglobulin Fc domain, or a fragment or variant thereof. As used herein, a “functional Fc region” is an Fc domain or fragment thereof which retains the ability to bind FcRn. In some embodiments, a functional Fc region binds to FcRn, but does not possess effector functions. The ability of the Fc region or fragment thereof to bind to FcRn can be determined by standard binding assays known in the art. In other embodiments, the Fc region or fragment thereof binds to FcRn and possesses at least one “effector function” of a native Fc region. Exemplary “effector functions” include C1q binding; complement dependent cytotoxicity (CDC); Fc receptor binding; antibody-dependent cell-mediated cytotoxicity (ADCC); phagocytosis; down regulation of cell surface receptors (e.g., B cell receptor; BCR), etc. Such effector functions generally require the Fc region to be combined with a binding domain (e.g., an anti-CD4 or ant-N-17 Adnectin™) and can be assessed using various assays known in the art for evaluating such antibody effector functions.


A “native sequence Fc region” comprises an amino acid sequence identical to the amino acid sequence of an Fc region found in nature. A “variant Fc region” comprises an amino acid sequence which differs from that of a native sequence Fc region by virtue of at least one amino acid modification. Preferably, the variant Fc region has at least one amino acid substitution compared to a native sequence Fc region or to the Fc region of a parent polypeptide, e.g., from about one to about ten amino acid substitutions, and preferably from about one to about five amino acid substitutions in a native sequence Fc region or in the Fc region of the parent polypeptide. The variant Fc region herein will preferably possess at least about 80% sequence identity with a native sequence Fc region and/or with an Fc region of a parent polypeptide, and most preferably at least about 90% sequence identity therewith, more preferably at least about 95% sequence identity therewith.


In an exemplary embodiment, the Fc domain is derived from an IgG1 subclass, however, other subclasses (e.g., IgG2, IgG3, and IgG4) may also be used. Shown below is the sequence of a human IgG1 immunoglobulin Fc domain:









(SEQ ID NO: 11)



DKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVS






HEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWL





NGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSREEMTKN





QVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYS





KLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK






The core hinge sequence is underlined, and the CH2 and CH3 regions are in regular text. It should be understood that the C-terminal glycine and lysine are optional in the Combinectin of the invention.


The fusion may be formed by attaching an Adnectin™ of the invention to either end of the Fc molecule, i.e., Fc-Adnectin™ or Adnectin™-Fc arrangements. In certain embodiments, the Fc and Adnectin™ are fused via a linker.


In some embodiments, the Fc region used in the Adnectin™ fusion comprises the hinge region of an Fc molecule. As used herein, the “hinge” region comprises the core hinge residues spanning positions 1-16 of SEQ ID NO: 11 (DKTHTCPPCPAPELLG; SEQ ID NO: 12) of the IgG1 Fc region. In certain embodiments, the Adnectin™-Fc fusion adopts a multimeric structure (e.g., dimer) owing, in part, to the cysteine residues at positions 6 and 9 of SEQ ID NO: 11 within the hinge region.


In certain embodiments, an Adnectin-Fc fusion may have the following configurations: 1) Adnectin™-hinge-Fc or 2) hinge-Fc-Adnectin™. Therefore, any Adnectin™ of the present invention can be fused to an Fc region comprising a hinge sequence according to these configurations. In some embodiments, a linker may be used to join the Adnectin™ to the hinge-Fc moiety, for example, an exemplary fusion protein may have the configuration Adnectin™-linker-hinge-Fc or hinge-Fc-linker-Adnectin™ Additionally, depending on the system in which the fusion polypeptide is produced, a leader sequence may be placed at the N-terminus of the fusion polypeptide. For example, if the fusion is produced in a mammalian system, a leader sequence may be added to the N-terminus of the fusion molecule. If the fusion is produced in E. coli, the fusion sequence will be preceded by a methionine.


VII. Nucleic Acid-Protein Fusion Technology


In one aspect, the invention provides an Adnectin™ comprising fibronectin type III domains that binds CD4 or the n17 domain of gp41. One way to rapidly make and test Fn3 domains with specific binding properties is the nucleic acid-protein fusion technology. This disclosure utilizes the in vitro expression and tagging technology, termed ‘PROfusion’ which exploits nucleic acid-protein fusions (RNA- and DNA-protein fusions) to identify novel polypeptides and amino acid motifs that are important for binding to proteins. Nucleic acid-protein fusion technology is a technology that covalently couples a protein to its encoding genetic information. For a detailed description of the RNA-protein fusion technology and fibronectin-based scaffold protein library screening methods see Szostak et al., U.S. Pat. Nos. 6,258,558, 6,261,804, 6,214,553, 6,281,344, 6,207,446, 6,518,018 and 6,818,418; Roberts et al., Proc. Natl. Acad. Sci., 94:12297-12302 (1997); and Kurz et al., Molecules, 5:1259-1264 (2000), all of which are herein incorporated by reference.


VIII. Vectors and Polynucleotides


Nucleic acids encoding any of the various proteins or polypeptides disclosed herein may be synthesized chemically. Codon usage may be selected so as to improve expression in a cell. Such codon usage will depend on the cell type selected. Specialized codon usage patterns have been developed for E. coli and other bacteria, as well as mammalian cells, plant cells, yeast cells and insect cells. See, for example, Mayfield et al., Proc. Natl. Acad. Sci. USA, 100(2):438-442 (Jan. 21, 2003); Sinclair et al., Protein Expr. Purif., 26(I):96-105 (October 2002); Connell, N. D., Curr. Opin. Biotechnol., 12(5):446-449 (October 2001); Makrides et al., Microbiol. Rev., 60(3):512-538 (September 1996); and Sharp et al., Yeast, 7(7):657-678 (October 1991).


General techniques for nucleic acid manipulation are described in, for example, Sambrook et al., Molecular Cloning: A Laboratory Manual, Second Edition, Vols. 1-3, Cold Spring Harbor Laboratory Press (1989), or Ausubel, F. et al., Current Protocols in Molecular Biology, Green Publishing and Wiley-Interscience, New York (1987) and periodic updates, herein incorporated by reference. Generally, the DNA encoding the polypeptide is operably linked to suitable transcriptional or translational regulatory elements derived from mammalian, viral, or insect genes. Such regulatory elements include a transcriptional promoter, an optional operator sequence to control transcription, a sequence encoding suitable mRNA ribosomal binding site, and sequences that control the termination of transcription and translation. The ability to replicate in a host, usually conferred by an origin of replication, and a selection gene to facilitate recognition of transformants is additionally incorporated.


The proteins described herein may be produced recombinantly not only directly, but also as a fusion polypeptide with a heterologous polypeptide, which is preferably a signal sequence or other polypeptide having a specific cleavage site at the N-terminus of the mature protein or polypeptide. The heterologous signal sequence selected preferably is one that is recognized and processed (i.e., cleaved by a signal peptidase) by the host cell.


For prokaryotic host cells that do not recognize and process a native signal sequence, the signal sequence is substituted by a prokaryotic signal sequence selected, for example, from the group of the alkaline phosphatase, penicillinase, 1 pp, or heat-stable enterotoxin II leaders.


For yeast secretion the native signal sequence may be substituted by, e.g., a yeast invertase leader, a factor leader (including Saccharomyces and Kluyveromyces alpha-factor leaders), or acid phosphatase leader, the C. albicans glucoamylase leader, or the signal sequence described in U.S. Pat. No. 5,631,144. In mammalian cell expression, mammalian signal sequences as well as viral secretory leaders, for example, the herpes simplex gD signal, are available. The DNA for such precursor regions may be ligated in reading frame to DNA encoding the protein.


Both expression and cloning vectors contain a nucleic acid sequence that enables the vector to replicate in one or more selected host cells. Generally, in cloning vectors this sequence is one that enables the vector to replicate independently of the host chromosomal DNA, and includes origins of replication or autonomously replicating sequences. Such sequences are well known for a variety of bacteria, yeast, and viruses. The origin of replication from the plasmid pBR322 is suitable for most Gram-negative bacteria, the 2 micron plasmid origin is suitable for yeast, and various viral origins (SV40, polyoma, adenovirus, VSV or BPV) are useful for cloning vectors in mammalian cells. Generally, the origin of replication component is not needed for mammalian expression vectors (the SV40 origin may typically be used only because it contains the early promoter).


Expression and cloning vectors may contain a selection gene, also termed a selectable marker. Typical selection genes encode proteins that (a) confer resistance to antibiotics or other toxins, e.g., ampicillin, neomycin, methotrexate, or tracycline, (b) complement auxotrophic deficiencies, or (c) supply critical nutrients not available from complex media, e.g., the gene encoding D-alanine racemase for Bacilli.


Expression and cloning vectors usually contain a promoter that is recognized by the host organism and is operably linked to the nucleic acid encoding the protein of the invention, e.g., a fibronectin-based scaffold protein. Promoters suitable for use with prokaryotic hosts include the phoA promoter, beta-lactamase and lactose promoter systems, alkaline phosphatase, a tryptophan (trp) promoter system, and hybrid promoters such as the tan promoter. However, other known bacterial promoters are suitable. Promoters for use in bacterial systems also will contain a Shine-Dalgarno (S.D.) sequence operably linked to the DNA encoding the protein of the invention. Promoter sequences are known for eukaryotes. Virtually all eukaryotic genes have an AT-rich region located approximately 25 to 30 bases upstream from the site where transcription is initiated. Another sequence found 70 to 80 bases upstream from the start of transcription of many genes is a CNCAAT region where N may be any nucleotide. At the 3′ end of most eukaryotic genes is an AATAAA sequence that may be the signal for addition of the poly A tail to the 3′ end of the coding sequence. All of these sequences are suitably inserted into eukaryotic expression vectors.


Examples of suitable promoting sequences for use with yeast hosts include the promoters for 3-phosphoglycerate kinase or other glycolytic enzymes, such as enolase, glyceraldehyde-3-phosphate dehydrogenase, hexokinase, pyruvate decarboxylase, phosphofructokinase, glucose-6-phosphate isomerase, 3-phosphoglycerate mutase, pyruvate kinase, triosephosphate isomerase, phosphoglucose isomerase, and glucokinase.


Transcription from vectors in mammalian host cells can be controlled, for example, by promoters obtained from the genomes of viruses such as polyoma virus, fowlpox virus, adenovirus (such as Adenovirus 2), bovine papilloma virus, avian sarcoma virus, cytomegalovirus, a retrovirus, hepatitis-B virus and most preferably Simian Virus 40 (SV40), from heterologous mammalian promoters, e.g., the actin promoter or an immunoglobulin promoter, from heat-shock promoters, provided such promoters are compatible with the host cell systems.


Transcription of a DNA encoding protein of the invention by higher eukaryotes is often increased by inserting an enhancer sequence into the vector. Many enhancer sequences are now known from mammalian genes (globin, elastase, albumin, α-fetoprotein, and insulin). Typically, however, one will use an enhancer from a eukaryotic cell virus. Examples include the SV40 enhancer on the late side of the replication origin (bp 100-270), the cytomegalovirus early promoter enhancer, the polyoma enhancer on the late side of the replication origin, and adenovirus enhancers. See also Yaniv, Nature, 297:17-18 (1982) on enhancing elements for activation of eukaryotic promoters. The enhancer may be spliced into the vector at a position 5′ or 3′ to the peptide-encoding sequence, but is preferably located at a site 5′ from the promoter.


Expression vectors used in eukaryotic host cells (e.g., yeast, fungi, insect, plant, animal, human, or nucleated cells from other multicellular organisms) will also contain sequences necessary for the termination of transcription and for stabilizing the mRNA. Such sequences are commonly available from the 5′ and, occasionally 3′, untranslated regions of eukaryotic or viral DNAs or cDNAs. These regions contain nucleotide segments transcribed as polyadenylated fragments in the untranslated portion of mRNA encoding the protein of the invention. One useful transcription termination component is the bovine growth hormone polyadenylation region. See WO 94/11026 and the expression vector disclosed therein.


The recombinant DNA can also include any type of protein tag sequence that may be useful for purifying the protein. Examples of protein tags include, but are not limited to, a histidine tag, a FLAG tag, a myc tag, an HA tag, or a GST tag. Appropriate cloning and expression vectors for use with bacterial, fungal, yeast, and mammalian cellular hosts can be found in Cloning Vectors: A Laboratory Manual, Elsevier, New York (1985), the relevant disclosure of which is hereby incorporated by reference.


The expression construct is introduced into the host cell using a method appropriate to the host cell, as will be apparent to one of skill in the art. A variety of methods for introducing nucleic acids into host cells are known in the art, including, but not limited to, electroporation; transfection employing calcium chloride, rubidium chloride, calcium phosphate, DEAE-dextran, or other substances; microprojectile bombardment; lipofection; and infection (where the vector is an infectious agent).


Suitable host cells include prokaryotes, yeast, mammalian cells, or bacterial cells. Suitable bacteria include gram negative or gram positive organisms, for example, E. coli or Bacillus spp. Yeast, preferably from the Saccharomyces species, such as S. cerevisiae, may also be used for production of polypeptides. Various mammalian or insect cell culture systems can also be employed to express recombinant proteins. Baculovirus systems for production of heterologous proteins in insect cells are reviewed by Luckow et al. (Bio/Technology, 6:47 (1988)). Examples of suitable mammalian host cell lines include endothelial cells, COS-7 monkey kidney cells, CV-1, L cells, C127, 3T3, Chinese hamster ovary (CHO), human embryonic kidney cells, HeLa, 293, 293T, and BHK cell lines. Purified polypeptides are prepared by culturing suitable host/vector systems to express the recombinant proteins. For many applications, the small size of many of the polypeptides disclosed herein would make expression in E. coli as the preferred method for expression. The protein is then purified from culture media or cell extracts.


IX. Protein Production


The present invention is also directed to cell lines that express a Combinectin or fusion protein thereof. Creation and isolation of cell lines producing a Combinectin can be accomplished using standard techniques known in the art, such as those described herein.


Host cells are transformed with the herein-described expression or cloning vectors for protein production and cultured in conventional nutrient media modified as appropriate for inducing promoters, selecting transformants, or amplifying the genes encoding the desired sequences. In the examples shown here, the host cells used for high-throughput protein production (HTPP) and mid-scale production were those from the HMS174-bacterial strain.


The host cells used to produce the proteins of this invention may be cultured in a variety of media. Commercially available media such as Ham's F10 (Sigma), Minimal Essential Medium ((MEM), (Sigma), RPMI-1640 (Sigma), and Dulbecco's Modified Eagle's Medium ((DMEM), Sigma)) are suitable for culturing the host cells. In addition, many of the media described in Ham et al., Meth. Enzymol., 58:44 (1979), Barites et al., Anal. Biochem., 102:255 (1980), U.S. Pat. Nos. 4,767,704, 4,657,866, 4,927,762, 4,560,655, 5,122,469, 6,048,728, 5,672,502, or U.S. Patent No. RE 30,985 may be used as culture media for the host cells. Any of these media may be supplemented as necessary with hormones and/or other growth factors (such as insulin, transferrin, or epidermal growth factor), salts (such as sodium chloride, calcium, magnesium, and phosphate), buffers (such as HEPES), nucleotides (such as adenosine and thymidine), antibiotics (such as Gentamycin drug), trace elements (defined as inorganic compounds usually present at final concentrations in the micromolar range), and glucose or an equivalent energy source. Any other necessary supplements may also be included at appropriate concentrations that would be known to those skilled in the art. The culture conditions, such as temperature, pH, and the like, are those previously used with the host cell selected for expression, and will be apparent to the ordinarily skilled artisan.


Proteins disclosed herein can also be produced using cell-free translation systems. For such purposes the nucleic acids encoding the polypeptide must be modified to allow in vitro transcription to produce mRNA and to allow cell-free translation of the mRNA in the particular cell-free system being utilized (eukaryotic such as a mammalian or yeast cell-free translation system or prokaryotic such as a bacterial cell-free translation system.


Proteins of the invention can also be produced by chemical synthesis (e.g., by the methods described in Solid Phase Peptide Synthesis, Second Edition, The Pierce Chemical Co., Rockford, Ill. (1984)). Modifications to the protein can also be produced by chemical synthesis.


The proteins of the present invention can be purified by isolation/purification methods for proteins generally known in the field of protein chemistry. Non-limiting examples include extraction, recrystallization, salting out (e.g., with ammonium sulfate or sodium sulfate), centrifugation, dialysis, ultrafiltration, adsorption chromatography, ion exchange chromatography, hydrophobic chromatography, normal phase chromatography, reversed-phase chromatography, get filtration, gel permeation chromatography, affinity chromatography, electrophoresis, countercurrent distribution or any combinations of these. After purification, polypeptides may be exchanged into different buffers and/or concentrated by any of a variety of methods known to the art, including, but not limited to, filtration and dialysis.


The purified polypeptide is preferably at least 85% pure, or preferably at least 95% pure, and most preferably at least 98% pure. Regardless of the exact numerical value of the purity, the polypeptide is sufficiently pure for use as a pharmaceutical product.


X. Biophysical and Biochemical Characterization


Binding of the protein of the invention to a target molecule (e.g., CD4 or gp41) may be assessed in terms of equilibrium constants (e.g., dissociation, Kd) and in terms of kinetic constants (e.g., on-rate constant, kon and off-rate constant, koff). The protein of the invention will generally bind to a target molecule with a Kd of less than 500 nM, 100 nM, 10 nM, 1 nM, 500 pM, 200 pM, or 100 pM, although higher Kd values may be tolerated where the koff is sufficiently low or the kon, is sufficiently high.


In Vitro Assays for Binding Affinity


Proteins that bind CD4 or gp41 can be identified using various in vitro assays. Preferably, the assays are high-throughput assays that allow for screening multiple candidates simultaneously.


In some embodiments, biomolecular interactions can be monitored in real time with the BIACORE® system, which uses SPR to detect changes in the resonance angle of light at the surface of a thin gold film on a glass support due to changes in the refractive index of the surface up to 300 nm away. BIACORE® analysis generates association rate constants, dissociation rate constants, equilibrium dissociation constants, and affinity constants. Binding affinity is obtained by assessing the association and dissociation rate constants using a BIACORE® surface plasmon resonance system (Biacore, Inc.). A biosensor chip is activated for covalent coupling of the target. The target is then diluted and injected over the chip to obtain a signal in response units of immobilized material. Since the signal in resonance units (RU) is proportional to the mass of immobilized material, this represents a range of immobilized target densities on the matrix. Association and dissociation data are fit simultaneously in a global analysis to solve the net rate expression for a 1:1 bimolecular interaction, yielding best fit values for kon, koff and Rmax (maximal response at saturation). Equilibrium dissociation constants for binding, Kd's are calculated from SPR measurements as koff/kon.


In some embodiments, the Combinectin of the invention exhibit a Kd of 100 nM or less. Preferably, the Kd is 10 nM or less. More preferably, the Kd is 1 nM or less.


In some embodiments, the Combinectin of the invention exhibit an IC50 of 5 nM or less, 4 nM or less, 3 nM or less, 2.5 nM or less, 2 nM or less, 1.5 nM or less, 1 nM or less, 0.5 nM or less, 0.2 nM or less, or 0.1 nM or less. Preferably, the IC50 is 1.5 nM or less. More preferably, the IC50 is 0.5 nM or less.


It should be understood that the assays described herein above are exemplary, and that any method known in the art for determining the binding affinity between proteins (e.g., fluorescence based-transfer (FRET), enzyme-linked immunosorbent assay, and competitive binding assays (e.g., radioimmunoassays)) can be used to assess the binding affinities of the Combinectin of the invention.


In the present invention, ELISA assays were utilized for identifying Adnectins™ that bind to CD4 or gp41, with affinities determined by BIACORE® SPR. FACS assays were also used to determine the EC50 of binding of the CD4 Adnectin™ (alone and as part of the full Combinectin) to CD4 as presented naturally on T-cell surfaces. Peptide affinities were measured by BIACORE® SPR.


As described in Table 5 below, the range of binding affinities (by SPR) for CD4 Adnectins™ to CD4 was 0.3 nM to 140 nM; the range for N17 Adnectin™ binding to artificial gp41-based targets was 0.5 nM to 40 nM; the range for peptide binding was 0.2 nM to 70 nM. SPR-based affinities for the Combinectin of the invention and for the individual components thereof are shown in Table 5 below.









TABLE 5







Binding Affinities for Combinectin and Individual Components















Kon
Koff
Kd


Protein
ID
Target
(1/Ms)
(1/s)
(nM)















CD4
ADX_
CD4
1.9E+05
7.6E−04
4


Adnectin ™
6940_B01






N17
ADX_
gp41
7.0E+06
3.3E−03
0.5


Adnectin ™
6200_A08
(IZN24)





peptide
203613-24
gp41
2.3E+06
2.5E−04
0.1




(PRD-828)





HSA-
BMT-
CD4
7.6E+03
8.3E−04
109


Combinectin
180280






HSA-
BMT-
gp41
8.1E+05
1.7E−03
2


Combinectin
180280
(IZN24)





HSA-
BMT-
gp41
9.6E+05
1.8E−04
0.2


Combinectin
180280
(PRD-828)













In Vitro Assays for Inhibition Activity


Various art-recognized in vitro systems exist that allow for examination of the potency of the Combinectin (or of individual inhibitors or combinations thereof) against HIV-1 infection. These include systems that allow for complete replication of laboratory-derived virus or clinical isolates of various strains in cultured cells or peripheral blood monocyte cultures. In addition, systems that recapitulate the early cell entry stages of infection, without using viable virus, could be used to analyze the effectiveness of the Combinectin, individual inhibitors or combinations thereof. These include, but are not limited to, “pseudotyped” viruses that contain deletions that make them unable to produce infectious virions or cells that express only the HIV gp160 gene that can be used to monitor the HIV-1specific fusion reaction to target cells.


In Vivo Models


One skilled in the art would know of various art-recognized animal models that allow for replication and in some cases recapitulate the symptoms of HIV infection. These models can be used to test the efficacy of the Combinectin, individual inhibitors or combinations thereof of the invention.


XI. Therapeutic Applications


In one aspect, the present invention provides Combinectins useful for the treatment of HIV. Accordingly, in certain embodiments the invention provides methods for attenuating or inhibiting HIV fusion in a subject comprising administering an effective amount of the Combinectin of the invention to a subject. In some embodiments, the subject is a human. In some embodiments, the Combinectin of the invention is pharmaceutically acceptable to a mammal, in particular a human. A “pharmaceutically acceptable” polypeptide refers to a polypeptide that is administered to an animal without significant adverse medical consequences.


In some embodiments, the Combinectin of the present invention will be administered to a subject in combination (concurrently or separately) with an agent known in the art to be useful for the particular disorder or disease being treated.


In some embodiments, the target patient population for Combinectin therapy is one that is not amenable to standard therapy for the disease being treated due to, e.g., age, pre-existing conditions, genetic makeup, and/or co-morbidities. The Combinectin of the invention can serve as an alternative to existing therapies that are associated with substantial side effects or safety concerns.


In some embodiments, the target patient population for Combinectin therapy is comprised of uninfected individuals at high risk of infection, due to lifestyle or other aggravating factors. The Combinectin is used to protect these individuals from getting infected by HIV (pre-exposure prophylaxis).


XII. Pharmaceutical Compositions


The present invention further provides pharmaceutical compositions comprising a Combinectin or fusion proteins thereof described herein, wherein the composition is essentially endotoxin free, or at least contain no more than acceptable levels of endotoxins as determined by the appropriate regulatory agency (e.g., FDA).


Compositions of the present invention can be in the form of a pill, tablet, capsule, liquid, or sustained release tablet for oral administration; a liquid for intravenous, subcutaneous or parenteral administration; or a gel, lotion, ointment, cream, or a polymer or other sustained release vehicle for local administration, or an atomizable suspension suitable for inhaled or intranasal administration.


Methods well known in the art for making compositions are found, for example, in Gennaro, A. R., ed., Remington: The Science and Practice of Pharmacy, 20th Edition, Lippincott Williams & Wilkins, Philadelphia, Pa. (2000). Compositions for parenteral administration may, for example, contain excipients, sterile water, saline, polyalkylene glycols such as polyethylene glycol, oils of vegetable origin, or hydrogenated naphthalenes. Biocompatible, biodegradable lactide polymer, lactide/glycolide copolymer, or polyoxyethylene-polyoxypropylene copolymers may be used to control the release of the compounds. Nanoparticulate compositions (e.g., biodegradable nanoparticles, solid lipid nanoparticles, liposomes) may be used to control the biodistribution of the compounds. Other potentially useful parenteral delivery systems include ethylene-vinyl acetate copolymer particles, osmotic pumps, implantable infusion systems, and liposomes. The concentration of the compound in the composition varies depending upon a number of factors, including the dosage of the drug to be administered, and the route of administration.


Acceptable carriers, excipients, or stabilizers are nontoxic to recipients at the dosages and concentrations employed, and include buffers such as phosphate, citrate, and other organic acids; antioxidants including ascorbic acid and methionine; preservatives (such as octadecyldimethylbenzyl ammonium chloride; hexamethonium chloride; benzalkonium chloride, benzethonium chloride; phenol, butyl or benzyl alcohol; alkyl parabens such as methyl or propyl paraben; catechol; resorcinol; cyclohexanol; 3-pentanol; and m-cresol); low molecular weight (less than about 10 residues) polypeptides; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, histidine, arginine, or lysine; monosaccharides, disaccharides, and other carbohydrates including glucose, mannose, or dextrans; chelating agents such as EDTA; sugars such as sucrose, mannitol, trehalose or sorbitol; salt-forming counter-ions such as sodium; metal complexes (e.g., Zn-protein complexes); and/or non-ionic surfactants such as Tween, PLURONIC® or polyethylene glycol (PEG).


The active ingredients may also be entrapped in a microcapsule prepared, for example, by coacervation techniques or by interfacial polymerization, for example, hydroxymethylcellulose or gelatin-microcapsule and poly-(methylmethacylate) microcapsule, respectively, in colloidal drug delivery systems (for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules) or in macroemulsions. Such techniques are disclosed in Osol, A., ed., Remington's Pharmaceutical Sciences, 16th Edition (1980).


Sustained-release preparations may be prepared. Suitable examples of sustained-release preparations include semipermeable matrices of solid hydrophobic polymers containing the proteins of the invention, which matrices are in the form of shaped articles, e.g., films, or microcapsule. Examples of sustained-release matrices include polyesters, hydrogels (for example, poly(2-hydroxyethyl-methacrylate), or poly(vinylalcohol)), polylactides (U.S. Pat. No. 3,773,919), copolymers of L-glutamic acid and y ethyl-L-glutamate, non-degradable ethylene-vinyl acetate, degradable lactic acid-glycolic acid copolymers such as the LUPRON DEPOT® (injectable microspheres composed of lactic acid-glycolic acid copolymer and leuprolide acetate), and poly-D-(−)-3-hydroxybutyric acid. While polymers such as ethylene-vinyl acetate and lactic acid-glycolic acid enable release of molecules for over 100 days, certain hydrogels release proteins for shorter time periods. When encapsulated proteins of the invention may remain in the body for a long time, they may denature or aggregate as a result of exposure to moisture at 37° C., resulting in a loss of biological activity and possible changes in immunogenicity. Rational strategies can be devised for stabilization depending on the mechanism involved. For example, if the aggregation mechanism is discovered to be intermolecular S—S bond formation through thio-disulfide interchange, stabilization may be achieved by modifying sulfhydryl residues, lyophilizing from acidic solutions, controlling moisture content, using appropriate additives, and developing specific polymer matrix compositions.


Compositions of the present invention for oral use include tablets containing the active ingredient(s) in a mixture with non-toxic pharmaceutically acceptable excipients. These excipients may be, for example, inert diluents or fillers (e.g., sucrose and sorbitol), lubricating agents, glidants, and anti-adhesives (e.g., magnesium stearate, zinc stearate, stearic acid, silicas, hydrogenated vegetable oils, or talc). Compositions for oral use may also be provided as chewable tablets, or as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent, or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium.


The pharmaceutical composition to be used for in vivo administration typically must be sterile. This may be accomplished by filtration through sterile filtration membranes. Where the composition is lyophilized, sterilization using this method may be conducted either prior to or following lyophilization and reconstitution. The composition for parenteral administration may be stored in lyophilized form or in solution. In addition, parenteral compositions generally are placed into a container having a sterile access port, for example, an intravenous solution bag or vial having a stopper pierceable by a hypodermic injection needle.


Once the pharmaceutical composition has been formulated, it may be stored in sterile vials as a solution, suspension, gel, emulsion, solid, or a dehydrated or lyophilized powder. Such formulations may be stored either in a ready-to-use form or in a form (e.g., lyophilized) requiring reconstitution prior to administration.


The compositions herein may also contain more than one active compound as necessary for the particular indication being treated, preferably those with complementary activities that do not adversely affect each other. Such molecules are suitably present in combination in amounts that are effective for the purpose intended.


XIII. Administration


A pharmaceutical composition comprising a Combinectin or fusion protein thereof of the present invention can be administered to a subject with HIV using standard administration techniques including oral, parenteral, pulmonary, transdermal, intramuscular, intranasal, buccal, sublingual, or suppository administration. Preferably, administration of the Combinectins of the invention is parenteral. The term parenteral as used herein includes intravenous, intramuscular, subcutaneous, rectal, vaginal, or intraperitoneal administration. Peripheral systemic delivery by intravenous or intraperitoneal or subcutaneous injection is preferred.


A therapeutically effective dose refers to a dose that produces the therapeutic effects for which it is administered. An effective amount of a pharmaceutical composition to be employed therapeutically will depend, for example, upon the therapeutic context and objectives. One skilled in the art will appreciate that the appropriate dosage levels for treatment will thus vary depending, in part, upon the molecule delivered, the indication for which the binding agent molecule is being used, the route of administration, and the size (body weight, body surface or organ size) and condition (the age and general health) of the patient.


For example, the therapeutically effective dose can be estimated initially either in cell culture assays or in animal models such as mice, rats, rabbits, dogs, pigs, or monkeys. An animal model may also be used to determine the appropriate concentration range and route of administration. Such information can then be used to determine useful doses and routes for administration in humans.


The exact dosage will be determined in light of factors related to the subject requiring treatment, and may be ascertained using standard techniques. Dosage and administration are adjusted to provide sufficient levels of the active compound or to maintain the desired effect. Factors that may be taken into account include the severity of the disease state, the general health of the subject, the age, weight, and gender of the subject, time and frequency of administration, drug combination(s), reaction sensitivities, and response to therapy. In general, the Combinectin of the present invention are administered at about 0.01 mg/kg to about 50 mg/kg per day, preferably about 0.01 mg/kg to about 30 mg/kg per day, most preferably about 0.01 mg/kg to about 20 mg/kg per day. In some embodiments, the Combinectin of the present invention are administered at weekly dosages of about 0.01 mg/kg to about 10 mg/kg, more preferably about 0.01 to about 5 mg/kg, most preferably about 0.01 to about 1 mg/kg. Alternatively, the Combinectin of the invention are administered at about 15 to about 100 mg/week, from about 20 to about 80 mg/week, from about 20 to about 60 mg/week, or about 20 to about 25 mg/week.


The frequency of dosing will depend upon the pharmacokinetic parameters of the binding agent molecule in the formulation used. Typically, a composition is administered until a dosage is reached that achieves the desired effect. The composition may therefore be administered as a single dose or as multiple doses (at the same or different concentrations/dosages) over time, or as a continuous infusion. Further refinement of the appropriate dosage is routinely made. Appropriate dosages may be ascertained through use of appropriate dose-response data. For example, the Combinectin may be given daily (e.g., once, twice, three times, or four times daily) or less frequently (e.g., once every other day, once or twice weekly, or monthly). In addition, as is known in the art, adjustments for age as well as the body weight, general health, sex, diet, time of administration, drug interaction, and the severity of the disease may be necessary, and will be ascertainable with routine experimentation by those skilled in the art. The Combinectin is suitably administered to the patient at one time or over a series of treatments.


Administration of a Combinectin or a fusion thereof, and one or more additional therapeutic agents, whether co-administered or administered sequentially, may occur as described above for therapeutic applications. Suitable pharmaceutically acceptable carriers, diluents, and excipients for co-administration will be understood by the skilled artisan to depend on the identity of the particular therapeutic agent being administered.


XIV. Kits and Articles of Manufacture


The Combinectin of the invention can be provided in a kit, a packaged combination of reagents in predetermined amounts with instructions for use in the therapeutic or diagnostic methods of the invention.


For example, in one embodiment of the invention, an article of manufacture containing materials useful for the treatment or prevention of the disorders or conditions described above is provided. The article of manufacture comprises a container and a label. Suitable containers include, for example, bottles, vials, syringes, and test tubes. The containers may be formed from a variety of materials such as glass or plastic. The container holds a composition of the invention which is effective for treating HIV and may have a sterile access port (for example, the container may be an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle). The active agent in the composition is a Combinectin of the invention. The label on, or associated with, the container indicates that the composition is used for treating HIV. The article of manufacture may further comprise a second container comprising a pharmaceutically-acceptable buffer, such as phosphate-buffered saline, Ringer's solution and dextrose solution. It may further include other materials desirable from a commercial and user standpoint, including other buffers, diluents, filters, needles, syringes, and package inserts with instructions for use.


INCORPORATION BY REFERENCE

All documents and references, including patent documents and websites, described herein are individually incorporated by reference to into this document to the same extent as if there were written in this document in full or in part.


The invention is now described by reference to the following examples, which are illustrative only, and are not intended to limit the present invention. While the invention has been described in detail and with reference to specific embodiments thereof, it will be apparent to one of skill in the art that various changes and modifications can be made thereto without departing from the spirit and scope thereof.


EXAMPLES
Example 1—Combinectin Production/Purification

HIV Combinectin Tandem—Bacterial


Transformed DNA into BL21(DE3) bacterial cells.


Cells grown up in bacterial culture at ˜37° C. to a target OD600


Culture temperature dropped to ˜30° C., and culture is induced with IPTG and harvested after several hours.


Harvest is performed using a centrifuge.


Recovery of protein is performed using a chemical lysis and a MICROFLUIDIZER®, followed by clarification by centrifugation or tangential flow filtration. Lysate is processed immediately or frozen for later use.


Purification by Hydrophobic Interaction Chromatography followed by hydroxyapatite chromatography and/or ion exchange chromatography. Formulated and concentrated using tangential flow filtration.


HIV Combinectin-Fc Construct; Mammalian Cell Culture


DNA is transfected into appropriate mammalian cells.


Cells grown in cell culture.


Harvested by centrifugation and/or filtration.


Purification using affinity chromatography and ion exchange chromatography.


Formulated and concentrated using tangential flow filtration.


HIV Combinectin-HuSA construct; Mammalian Cell Culture


DNA is transfected into appropriate mammalian cells.


Cells grown in cell culture.


Harvested by centrifugation and/or filtration.


Purification by Hydrophobic Interaction Chromatography followed by hydroxyapatite chromatography and/or ion exchange chromatography.


Formulated and concentrated using tangential flow filtration.


Example 2—Combinectin Potency Assay

MT-2 cells, HEK 293T cells and the proviral DNA clone of NU-3 were obtained from the NIH AIDS Research and Reference Reagent Program. MT-2 cells were propagated in RPMI 1640 medium supplemented with 10% heat-inactivated fetal bovine serum (FBS), 10 mM HEPES buffer pH 7.55, and 2 mM L-glutamine. HEK 293T cells were propagated in DMEM media supplemented with 10% heat-inactivated FBS, 10 mM HEPES buffer pH 7.55 and 2 mM L-glutamine. Recombinant NL-Rluc virus, in which a section of the nef gene from the proviral clone of NL4-3 was replaced with the Renilla luciferase gene, was constructed at Bristol-Myers Squibb. The replication-competent virus was harvested 3 days after transfection of HEK 293T cells with the modified pNL-Rluc proviral clone. Transfections were performed using Lipofectamine Plus (Invitrogen, Carlsbad, Calif.), according to manufacturer's instruction. Virus was titered in MT-2 cells using luciferase enzyme activity as a biomarker. The NL-Rluc virus used to infect MT-2 cells a multiplicity of 0.01 for 1 hour before adding to the peptides in 96-well plates. Peptides were serially diluted three-fold and 11 concentrations were plated in triplicate. After 4-5 days of incubation, cells were processed and quantitated for virus growth by the amount of expressed luciferase. Luciferase was quantitated using the Dual Luciferase kit from Promega (Madison, Wis.), with modifications to the manufacturer's protocol. The diluted Passive Lysis solution was pre-mixed with the re-suspended Luciferase Assay Reagent and then re-suspended in STOP & GLO® Substrate (2:1:1 ratio). A total of 50 pt of the mixture was added to each aspirated well on assay plates and luciferase activity was measured immediately on a Wallac TriLux (Perkin-Elmer, Waltham, Mass.). The 50% effective concentration (EC50) was calculated by comparing the amount of luciferase produced in the presence of inhibitory peptide compared to wells where no peptide was added.


Example 3—Evaluation of Combinectin Pharmacokinetics

Human CD4 Transgenic Mouse Model


Male and female heterozygous human CD4 mice were obtained from Jackson Laboratories, Bar Harbor, Me.


WT Mouse PK Studies


8-21 day single IV bolus dose studies were run in female C57Bl/6 WT mice to assess the PK properties of the various Combinectins. Fc-Combinectin fusions were dosed at 10 mg/kg and HSA-Combinectin fusions were dosed at 8.8 mg/kg. Plasma samples were collected in CPD and stored at −80° C. until analysis.


hCD4 Mouse PK Studies


7-10 day single IV bolus dose studies were run in heterozygous hCD4 mice to assess the PK properties of various Combinectins in the presence of target. Combinectins doses and sample collection methods were the same as described above for the WT mice.


Cynomolgus Monkey Studies


A 1-week single dose study was conducted in female cynos to determine the PK of Combinectins. Following a 1 mg/kg dose, serum samples were collected at the indicated times, aliquoted and quick frozen for MSD or LC/MS analysis.


Pharmacokinetic Measurements


Drug levels were measured in mouse or cyno plasma using the Mesoscale technology platform or colorimetric ELISA formats. Fc-Combinectin fusions were captured via the protein PRD828 (BMS) that specifically binds the peptide component of Combinectins and detected using a Goat anti-Human IgG Fc-HRP conjugated pAb (Pierce #31413). HSA-Combinectin fusions were captured via PRD828 and detected using a goat pAb against HSA (Bethyl, TX #A80-229A) that was ruthenium labeled. Sample concentrations were calculated from a standard curve using a 5-parameter logarithmic fit. Non-compartmental analyses were performed using Phoenix WINNONLIN® 6.3 (Pharsight Corporation, Mountain View, Calif.) using a plasma model and linear up/log down calculation method.

Claims
  • 1. A polypeptide comprising two active domains, wherein one domain is a fibronectin-based scaffold domain protein that binds the N17 domain of glycoprotein 41 (gp41) and comprises the amino acid sequence that is at least 90% identical to the amino acid sequence of any one of SEQ ID NOs: 115-371; and the other domain is a cluster of differentiation 4 (CD4) binding moiety or a human immunodeficiency virus (HIV) fusion peptide inhibitor, wherein the CD4 binding moiety is selected from the group consisting of an anti-CD4 antibody or fragment thereof, and a fibronectin-based scaffold domain protein that binds CD4 and comprises the amino acid sequence that is at least 90% identical to the amino acid sequence of any one of SEQ ID NOs: 95-114.
  • 2. The polypeptide of claim 1, wherein the two domains are connected to each other in any order by linkers.
  • 3. The polypeptide of claim 1, further comprising one or more pharmacokinetic (PK) moieties selected from the group consisting of polyethylene glycol, sialic acid, Fc, Fc fragment, transferrin, serum albumin, a serum albumin binding protein, and a serum immunoglobulin binding protein.
  • 4. The polypeptide of claim 3, wherein the PK moiety is Fc.
  • 5. The polypeptide of claim 4, wherein the Fc is attached to the N-terminus of the polypeptide.
  • 6. The polypeptide of claim 3, wherein the PK moiety is human serum albumin.
  • 7. The polypeptide of claim 6, wherein the human serum albumin is attached to the N-terminus of the polypeptide.
CROSS-REFERENCE TO RELATED APPLICATIONS

This application is a divisional of U.S. application Ser. No. 15/568,830, filed on 24 Oct. 2017, now U.S. Pat. No. 10,407,490, which is a 371 of lntemational PCT/US2016/027424 filed on 14 Apr. 2016, which claims priority to U.S. Provisional Application Ser. Nos. 62/152,271, filed Apr. 24, 2015, and 62/257,474, filed Nov. 19, 2015; the entire contents of which are incorporated herein by reference.

US Referenced Citations (2)
Number Name Date Kind
20070048282 Rosen et al. Mar 2007 A1
20090022720 Fischer et al. Jan 2009 A1
Foreign Referenced Citations (4)
Number Date Country
WO 2011077093 Jun 2011 WO
WO 2011130354 Oct 2011 WO
WO-2014165093 Oct 2014 WO
WO 2014206336 Dec 2014 WO
Non-Patent Literature Citations (5)
Entry
Eckert, D.M. and Kim, P.S., “Design of P potent inhibitors of HIV-1 entry from the gp41-N-peptide region”, Proc. Natl. Acad. Sci. USA, 98(20):11187-92 (2001).
Greenberg, et al, Resistance to enfuvirtide, the first HIV fusion inhibitor, Journal of Antimicrobial Chemotherapy, 54:333-340 (2004).
Haqqani, et al., “Entry inhibitors and their use in the treatment of HIV-1 infection” Antiviral Research; 98(2):158-170 (2013).
Jain, et al, “Mutual prodrugs containing bio-cleavable and drug releasable disulfide linkers”, Bioorganic Chemistry, 49:40-48 (2013).
Krystal, M., et al. HIV-1 Combinectin BMS-986197: A Long-Acting Inhibitor With Multiple Modes of Action. Conference on Retroviruses and Opportunistic Infections (CROI): Conference Abstract, Feb. 22, 2016.
Related Publications (1)
Number Date Country
20190359688 A1 Nov 2019 US
Provisional Applications (2)
Number Date Country
62257474 Nov 2015 US
62152271 Apr 2015 US
Divisions (1)
Number Date Country
Parent 15568830 US
Child 16533941 US