US20150087810A1 - Matrix Metalloproteinase Substrates And Other Cleavable Moieties And Methods Of Use Thereof - Google Patents

Matrix Metalloproteinase Substrates And Other Cleavable Moieties And Methods Of Use Thereof Download PDF

Info

Publication number
US20150087810A1
US20150087810A1 US14/497,089 US201414497089A US2015087810A1 US 20150087810 A1 US20150087810 A1 US 20150087810A1 US 201414497089 A US201414497089 A US 201414497089A US 2015087810 A1 US2015087810 A1 US 2015087810A1
Authority
US
United States
Prior art keywords
seq
amino acid
acid sequence
isolated polypeptide
sequence
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US14/497,089
Other languages
English (en)
Inventor
Stephen James Moore
Margaret Thy Luu Nguyen
Daniel R. Hostetter
Olga Vasiljeva
Jeanne Grace Flandez
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Cytomx Therapeutics Inc
Original Assignee
Cytomx Therapeutics Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Cytomx Therapeutics Inc filed Critical Cytomx Therapeutics Inc
Priority to US14/497,089 priority Critical patent/US20150087810A1/en
Assigned to CYTOMX THERAPEUTICS, INC. reassignment CYTOMX THERAPEUTICS, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: HOSTETTER, DANIEL R., Vasiljeva, Olga, FLANDEZ, JEANNE GRACE, MOORE, STEPHEN JAMES, NGUYEN, MARGARET THY LUU
Publication of US20150087810A1 publication Critical patent/US20150087810A1/en
Priority to US16/179,670 priority patent/US11814410B2/en
Priority to US18/474,103 priority patent/US20240150478A1/en
Priority to US18/474,134 priority patent/US20240084022A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2866Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against receptors for cytokines, lymphokines, interferons
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2863Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against receptors for growth factors, growth regulators
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/44Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material not provided for elsewhere, e.g. haptens, metals, DNA, RNA, amino acids
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K7/00Peptides having 5 to 20 amino acids in a fully defined sequence; Derivatives thereof
    • C07K7/04Linear peptides containing only normal peptide links
    • C07K7/06Linear peptides containing only normal peptide links having 5 to 11 amino acids
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/50Fusion polypeptide containing protease site
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/95Fusion polypeptide containing a motif/fusion for degradation (ubiquitin fusions, PEST sequence)

Definitions

  • the invention relates generally to polypeptides that include a cleavable moiety that is a substrate for at least one matrix metalloprotease (MMP), to activatable antibodies and other larger molecules that include the cleavable moiety that is a substrate for at least one MMP protease, and to methods of making and using these polypeptides that include a cleavable moiety that is a substrate for at least one MMP protease in a variety of therapeutic, diagnostic and prophylactic indications.
  • MMP matrix metalloprotease
  • Proteases are enzymes that degrade proteins by cleaving the peptide bonds between amino acid residues. Proteases occur naturally in all organisms and are involved in a variety of physiological reactions from simple degradation to highly regulated pathways. Some proteases are known to break specific peptide bonds based on the presence of a particular amino acid sequence within a protein.
  • CM cleavable moiety
  • MMP matrix metalloprotease
  • the CM is a substrate for at least one matrix metalloprotease (MMP).
  • MMPs include MMP1; MMP2; MMP3; MMP7; MMP8; MMP9; MMP10; MMP11; MMP12; MMP13; MMP14; MMP15; MMP16; MMP17; MMP19; MMP20; MMP23; MMP24; MMP26; and MMP27.
  • the CM is a substrate for MMP9, MMP14, MMP1, MMP3, MMP13, MMP17, MMP11, and MMP19.
  • the CM is a substrate for MMP9.
  • the CM is a substrate for MMP14.
  • the CM is a substrate for two or more MMPs. In some embodiments, the CM is a substrate for at least MMP9 and MMP14. In some embodiments, the CM comprises two or more substrates for the same MMP. In some embodiments, the CM comprises at least two or more MMP9 substrates. In some embodiments, the CM comprises at least two or more MMP14 substrates.
  • the CM is a substrate for an MMP and includes the sequence ISSGLLSS (SEQ ID NO: 14); QNQALRMA (SEQ ID NO: 15); AQNLLGMV (SEQ ID NO: 16); STFPFGMF (SEQ ID NO: 17); PVGYTSSL (SEQ ID NO: 18); DWLYWPGI (SEQ ID NO: 19); MIAPVAYR (SEQ ID NO: 20); RPSPMWAY (SEQ ID NO: 21); WATPRPMR (SEQ ID NO: 22); FRLLDWQW (SEQ ID NO: 23); LKAAPRWA (SEQ ID NO: 24); GPSHLVLT (SEQ ID NO: 25); LPGGLSPW (SEQ ID NO: 26); MGLFSEAG (SEQ ID NO: 27); SPLPLRVP (SEQ ID NO: 28); RMHLRSLG (SEQ ID NO: 29); LAAPLGLL (SEQ ID NO: 30); AVGLLAPP (SEQ ID NO: 31); LLAPSHRA (SEQ ID NO
  • the CM comprises the amino acid sequence ISSGLLSS (SEQ ID NO: 14). In some embodiments, the CM comprises the amino acid sequence QNQALRMA (SEQ ID NO: 15). In some embodiments, the CM comprises the amino acid sequence AQNLLGMV (SEQ ID NO: 16). In some embodiments, the CM comprises the amino acid sequence STFPFGMF (SEQ ID NO: 17). In some embodiments, the CM comprises the amino acid sequence PVGYTSSL (SEQ ID NO: 18). In some embodiments, the CM comprises the amino acid sequence DWLYWPGI (SEQ ID NO: 19). In some embodiments, the CM comprises the amino acid sequence MIAPVAYR (SEQ ID NO: 20).
  • the CM comprises the amino acid sequence RPSPMWAY (SEQ ID NO: 21). In some embodiments, the CM comprises the amino acid sequence WATPRPMR (SEQ ID NO: 22). In some embodiments, the CM comprises the amino acid sequence FRLLDWQW (SEQ ID NO: 23). In some embodiments, the CM comprises the amino acid sequence LKAAPRWA (SEQ ID NO: 24). In some embodiments, the CM comprises the amino acid sequence GPSHLVLT (SEQ ID NO: 25). In some embodiments, the CM comprises the amino acid sequence LPGGLSPW (SEQ ID NO: 26). In some embodiments, the CM comprises the amino acid sequence MGLFSEAG (SEQ ID NO: 27).
  • the CM comprises the amino acid sequence SPLPLRVP (SEQ ID NO: 28). In some embodiments, the CM comprises the amino acid sequence RMHLRSLG (SEQ ID NO: 29). In some embodiments, the CM comprises the amino acid sequence LAAPLGLL (SEQ ID NO: 30). In some embodiments, the CM comprises the amino acid sequence AVGLLAPP (SEQ ID NO: 31). In some embodiments, the CM comprises the amino acid sequence LLAPSHRA (SEQ ID NO: 32). In some embodiments, the CM comprises the amino acid sequence PAGLWLDP (SEQ ID NO: 33). In some embodiments, the CM comprises the amino acid sequence ISSGLSS (SEQ ID NO: 159).
  • the CM is linked or otherwise attached to an antibody.
  • the CM is used to link one or more agents to the antibody or antigen binding fragment thereof (AB) that binds a given target, such that the CM is cleaved when exposed to the MMP and the agent is released from the AB.
  • AB antigen binding fragment thereof
  • Exemplary targets include, but are not limited to the targets shown in Table 1.
  • Exemplary ABs include, but are not limited to, the targets shown in Table 2.
  • the antibody in the uncleaved state has the structural arrangement from N-terminus to C-terminus as follows: Agent-CM-AB or AB-CM-Agent.
  • the antibody comprises a linking peptide between the AB and the CM.
  • the antibody comprises a linking peptide between the CM and the conjugated agent.
  • the antibody comprises a first linking peptide (LP1) and a second linking peptide (LP2), and the antibody in the uncleaved state has the structural arrangement from N-terminus to C-terminus as follows: Agent-LP1-CM-LP2-AB or AB-LP2-CM-LP1-Agent.
  • each of LP1 and LP2 is a peptide of about 1 to 20 amino acids in length.
  • the two linking peptides need not be identical to each other.
  • At least one of LP1 or LP2 comprises an amino acid sequence selected from the group consisting of (GS) n , (GGS) n , (GSGGS) n (SEQ ID NO: 1) and (GGGS) n (SEQ ID NO: 2), where n is an integer of at least one.
  • At least one of LP1 or LP2 comprises an amino acid sequence selected from the group consisting of GGSG (SEQ ID NO: 3), GGSGG (SEQ ID NO: 4), GSGSG (SEQ ID NO: 5), GSGGG (SEQ ID NO: 6), GGGSG (SEQ ID NO: 7), and GSSSG (SEQ ID NO: 8).
  • LP1 comprises the amino acid sequence GSSGGSGGSGGSG (SEQ ID NO: 9), GSSGGSGGSGG (SEQ ID NO: 10), GSSGGSGGSGGS (SEQ ID NO: 11), GSSGGSGGSGGSGGGS (SEQ ID NO: 155), GSSGGSGGSG (SEQ ID NO: 156), or GSSGGSGGSGS (SEQ ID NO: 157).
  • LP2 comprises the amino acid sequence GSS, GGS, GGGS (SEQ ID NO: 158), GSSGT (SEQ ID NO: 12) or GSSG (SEQ ID NO: 13).
  • the AB has an equilibrium dissociation constant of about 100 nM or less for binding to the target.
  • the antibody includes an antibody or antigen-binding fragment thereof that specifically binds a target.
  • the antibody or immunologically active fragment thereof that binds the target is a monoclonal antibody, domain antibody, single chain, Fab fragment, a F(ab′) 2 fragment, a scFv, a scab, a dAb, a single domain heavy chain antibody, or a single domain light chain antibody.
  • such an antibody or immunologically active fragment thereof that binds the target is a mouse, other rodent, chimeric, humanized or fully human monoclonal antibody.
  • the MMP protease is co-localized with the target in a tissue, and the MMP protease cleaves the CM in the antibody when the antibody is exposed to the protease.
  • the CM is a polypeptide of up to 15 amino acids in length.
  • the CM is a substrate for at least one matrix metalloprotease (MMP).
  • MMPs include MMP1; MMP2; MMP3; MMP7; MMP8; MMP9; MMP10; MMP11; MMP12; MMP13; MMP14; MMP15; MMP16; MMP17; MMP19; MMP20; MMP23; MMP24; MMP26; and MMP27.
  • the CM is a substrate for MMP9, MMP14, MMP1, MMP3, MMP13, MMP17, MMP11, and MMP19.
  • the CM is a substrate for MMP9.
  • the CM is a substrate for MMP14.
  • the CM is a substrate for two or more MMPs. In some embodiments, the CM is a substrate for at least MMP9 and MMP14. In some embodiments, the CM comprises two or more substrates for the same MMP. In some embodiments, the CM comprises at least two or more MMP9 substrates. In some embodiments, the CM comprises at least two or more MMP14 substrates.
  • the CM is a substrate for an MMP and includes the sequence ISSGLLSS (SEQ ID NO: 14); QNQALRMA (SEQ ID NO: 15); AQNLLGMV (SEQ ID NO: 16); STFPFGMF (SEQ ID NO: 17); PVGYTSSL (SEQ ID NO: 18); DWLYWPGI (SEQ ID NO: 19); MIAPVAYR (SEQ ID NO: 20); RPSPMWAY (SEQ ID NO: 21); WATPRPMR (SEQ ID NO: 22); FRLLDWQW (SEQ ID NO: 23); LKAAPRWA (SEQ ID NO: 24); GPSHLVLT (SEQ ID NO: 25); LPGGLSPW (SEQ ID NO: 26); MGLFSEAG (SEQ ID NO: 27); SPLPLRVP (SEQ ID NO: 28); RMHLRSLG (SEQ ID NO: 29); LAAPLGLL (SEQ ID NO: 30); AVGLLAPP (SEQ ID NO: 31); LLAPSHRA (SEQ ID NO
  • the CM comprises the amino acid sequence ISSGLLSS (SEQ ID NO: 14). In some embodiments, the CM comprises the amino acid sequence QNQALRMA (SEQ ID NO: 15). In some embodiments, the CM comprises the amino acid sequence AQNLLGMV (SEQ ID NO: 16). In some embodiments, the CM comprises the amino acid sequence STFPFGMF (SEQ ID NO: 17). In some embodiments, the CM comprises the amino acid sequence PVGYTSSL (SEQ ID NO: 18). In some embodiments, the CM comprises the amino acid sequence DWLYWPGI (SEQ ID NO: 19). In some embodiments, the CM comprises the amino acid sequence MIAPVAYR (SEQ ID NO: 20).
  • the CM comprises the amino acid sequence RPSPMWAY (SEQ ID NO: 21). In some embodiments, the CM comprises the amino acid sequence WATPRPMR (SEQ ID NO: 22). In some embodiments, the CM comprises the amino acid sequence FRLLDWQW (SEQ ID NO: 23). In some embodiments, the CM comprises the amino acid sequence LKAAPRWA (SEQ ID NO: 24). In some embodiments, the CM comprises the amino acid sequence GPSHLVLT (SEQ ID NO: 25). In some embodiments, the CM comprises the amino acid sequence LPGGLSPW (SEQ ID NO: 26). In some embodiments, the CM comprises the amino acid sequence MGLFSEAG (SEQ ID NO: 27).
  • the CM comprises the amino acid sequence SPLPLRVP (SEQ ID NO: 28). In some embodiments, the CM comprises the amino acid sequence RMHLRSLG (SEQ ID NO: 29). In some embodiments, the CM comprises the amino acid sequence LAAPLGLL (SEQ ID NO: 30). In some embodiments, the CM comprises the amino acid sequence AVGLLAPP (SEQ ID NO: 31). In some embodiments, the CM comprises the amino acid sequence LLAPSHRA (SEQ ID NO: 32). In some embodiments, the CM comprises the amino acid sequence PAGLWLDP (SEQ ID NO: 33). In some embodiments, the CM comprises the amino acid sequence ISSGLSS (SEQ ID NO: 159).
  • the CM is a substrate for at least one matrix metalloprotease (MMP) and includes a motif sequence that is recognized by MMP9. In some embodiments, the CM is a substrate for at least one MMP and includes a motif sequence that is recognized by MMP14.
  • MMP matrix metalloprotease
  • the CM is a substrate for at least one MMP
  • the CM polypeptide and/or the CM portion of any polypeptide that comprises the CM comprises a polypeptide having a length less than 50 amino acids, less than 40 amino acids, less than 30 amino acids, less than 25 amino acids, less than 20 amino acids, less than 19 amino acids, less than 18 amino acids, less than 17 amino acids, less than 16 amino acids, less than 15 amino acids long, less than 14 amino acids, less than 13 amino acids, less than 12 amino acids, less than 11 amino acids, or less than 10 amino acids long.
  • the CM is a substrate for at least one MMP and comprises a polypeptide sequence that is not substantially identical to any human polypeptide sequence that is naturally cleaved by the same MMP protease. In some embodiments, the CM is a substrate for at least one MMP and comprises a polypeptide sequence that is no more than 90% or more identical to any human polypeptide sequence that is naturally cleaved by the same MMP protease.
  • the motif sequence is a substrate for at least MMP and includes a core CM consensus sequence shown in Tables 8A-8M below. In some embodiments, the motif sequence includes a subgenus, i.e., a subset, of the core CM consensus sequence shown in Tables 8A-8M below.
  • the motif sequence is a substrate for at least MMP9 and includes a core CM consensus sequence shown in Tables 8A-8D. In some embodiments, the motif sequence is a substrate for at least MMP9 and includes a subgenus, i.e., a subset, of the core CM consensus sequence shown in Tables 8A-8D below.
  • the motif sequence is a substrate for at least MMP14 and includes a core CM consensus sequence shown in Tables 8E-8M. In some embodiments, the motif sequence is a substrate for at least MMP14 and includes a subgenus, i.e., a subset, of the core CM consensus sequence shown in Tables 8E-8M below.
  • X 23 is L, M, P, S, or T; Subgenus 1.2: X 22 X 23 X 24 X 25 X 26 X 27 X 28 X 29 (SEQ ID NO: 319), X 24 is A, D, F, G, L, M, N, wherein X 22 is G, P or R; X 23 is P; X 24 is L, M, or S; X 25 is G, P, R, S, T, or V; P, or S; X 26 is L, M, or R; X 27 is W; X 28 is A, G, or S; and X 29 X 25 is A, D, E, G, H, I, M, is R, V, or Y.
  • X 22 is P or R; X 23 is P; X 24 is M or S; X 25 is G or P; X 26 R, V, W, or Y; is L, M, or R; X 27 is W; X 28 is A, G, or S; and X 29 is R, V, or Y.
  • X 27 is C, F, G, H, P, Q, Subgenus 1.4: X 22 X 23 X 24 X 25 X 26 X 27 X 28 X 29 (SEQ ID NO: 321), R, T, V, or W; wherein X 22 is P or R; X 23 is P; X 24 is S; X 25 is G or P; X 26 is X 28 is A, D, G, L, M, S, T, M, or R; X 27 is W; X 28 is A or S; and X 29 is Y.
  • X 22 X 23 X 24 X 25 X 26 X 27 X 28 X 29 (SEQ ID NO: 322), X 29 is C, H, L, R, S, V, W, wherein X 22 is P or R; X 23 is P; X 24 is S; X 25 is G or P; X 26 is or Y. M, or R; X 27 is W; X 28 is A or S; and X 29 is Y.
  • Subgenus 1.6 X 22 X 23 X 24 X 25 X 26 X 27 X 28 X 29 (SEQ ID NO: 323), wherein X 22 is C, G, H, L, or R; X 23 is P, S or T; X 24 is N, R, S or T; X 25 is P or S; X 26 is C, M, R, V, or W; X 27 is C, P, R, or W; X 28 is A, D, or G; and X 29 is C or Y.
  • Subgenus 2.2 X 32 X 33 X 34 X 35 X 36 X 37 X 38 X 39 (SEQ ID X 34 is C, G, H, L, Q, S, T, W, NO: 326), wherein X 32 is W; X 33 is D; X 34 is H, Q, or W; X 35 X 35 is D, G, L, P; is D or P; X 36 is I or R; X 37 is G, S, or V; X 38 is L, M, X 36 is E, G, I, L, N, P, R, or V; or V; and X 39 is G, L, or S.
  • X 37 is G, L, P, R, S, or V; Subgenus 2.3: X 32 X 33 X 34 X 35 X 36 X 37 X 38 X 39 (SEQ ID X 38 is A, I, L, M, T, or V; and NO: 327), wherein X 32 is W; X 33 is D; X 34 is H, Q, or W; X 35 X 39 is A, G, L, P, Q, R, S, or V. is P; X 36 is I or R; X 37 is S; X 38 is L, M, or V; and X 39 is L.
  • X 46 is A, F, R, S, or T
  • X 47 is G, H, T or Y
  • X 42 is G, I, L, M, P, R, S, X 48 is G, I, M, V, or W
  • X 49 is F, L, or S.
  • Subgenus 3.2 X 42 X 43 X 44 X 45 X 46 X 47 X 48 X 49 (SEQ ID NO: 331), X 43 is A, D, H, I, L, P, S, wherein X 42 is L, M, or S; X 43 is S or T; X 44 is F or L; X 45 is P; X 46 or T; is A, F, or T; X 47 is G, H, T or Y; X 48 is I, M, or W; and X 49 is F.
  • X 44 is F, L, S, or V; Subgenus 3.3: X 42 X 43 X 44 X 45 X 46 X 47 X 48 X 49 (SEQ ID NO: 332), X 45 is H, L, M, P, Q, R, wherein X 42 is L, M, or S; X 43 is S or T; X 44 is F; X 45 is P; X 46 is S, or T; A, F, or T; X 47 is G, H, or Y; X 48 is I, M, or W; and X 49 is F.
  • X 46 is A, D, F, G, L, M, Subgenus 3.4: X 42 X 43 X 44 X 45 X 46 X 47 X 48 X 49 (SEQ ID NO: 333), R, S, T, or V; wherein X 42 is L or M; X 43 is S or T; X 44 is F; X 45 is P; X 46 is A or T; X 47 is A, C, G, H, Q, T or X 47 is H or Y; X 48 is I or W; and X 49 is F.
  • X 42 X 43 X 44 X 45 X 46 X 47 X 48 X 49 (SEQ ID NO: 334), X 48 is C, G, I, M, R, S, T, wherein X 42 is G, I, R, Or S; X 43 is H Or T; X 44 is F, L, S, or V; X 45 V, or W; and is L, P, or R; X 46 is F, L, or S; X 47 is A, C, or G; X 48 is I, M, or V; and X 49 is F, L, S, or Y. X 49 is F or L.
  • Subgenus 3.6 X 42 X 43 X 44 X 45 X 46 X 47 X 48 X 49 (SEQ ID NO: 335), wherein X 42 is S; X 43 is T; X 44 is F or V; X 45 is L or P; X 46 is F or L; X 47 is G; X 48 is I or M; and X 49 is F.
  • X 53 is A, C, D, G, L, R, Subgenus 4.2: X 52 X 53 X 54 X 55 X 56 X 57 X 58 X 59 (SEQ ID NO: 342), V, W, or Y; wherein X 52 is D or H; X 53 is W or Y; X 54 is H or L; X 55 is H, L, X 54 is D, H, L, P, Q, R, S, or Y; X 56 IS G or W; X 57 is P or R; X 58 is G, L, or P; and X 59 is G, or Y; I, S, or T.
  • X 55 is D, F, H, I, L, M, P, Subgenus 4.3: X 52 X 53 X 54 X 55 X 56 X 57 X 58 X 59 (SEQ ID NO: 343), S, or Y; wherein X 52 is H; X 53 is W; X 54 is H or L; X 55 is H, L, or Y; X 56 Is X 56 is A, C, E, F, G, K, G or W; X 57 is P; X 58 is L or P; and X 59 is G, I, S or T.
  • X 52 X 53 X 54 X 55 X 56 X 57 X 58 X 59 (SEQ ID NO: 344), X 57 is A, G, K L, M, N, wherein X 52 is H; X 53 is W; X 54 is H or L; X 55 is L or Y; X 56 Is G; P, R, S, or T; X 57 is P; X 58 is L or P; and X 59 is G, I, S, or T.
  • X 58 is A, F, G, H, L, P, R, Subgenus 4.5: X 52 X 53 X 54 X 55 X 56 X 57 X 58 X 59 (SEQ ID NO: 345), S, or T; and wherein X 52 is H; X 53 is W; X 54 is H or L X 55 is L or Y X 56 Is G; X 59 is A, G, H, I, N, P, S, X 57 is P; X 58 is P; and X 59 is T. T, or Y.
  • Subgenus 4.6 X 52 X 53 X 54 X 55 X 56 X 57 X 58 X 59 (SEQ ID NO: 346), wherein X 52 is D, G, S, or Y; X 53 is W; X 54 is L or P; X 55 is D or Y; X 56 is C, E, G, or W; X 57 is M or P; X 58 is G, R, or S; and X 59 is H, I, or Y.
  • Subgenus 4.7 X 52 X 53 X 54 X 55 X 56 X 57 X 58 X 59 (SEQ ID NO: 347), wherein X 52 is D, G, or S; X 53 is W; X 54 is L; X 55 is Y; X 56 is E or W; X 57 is M or P; X 58 is G or S; and X 59 is I or Y.
  • X 62 X 63 X 64 X 65 X 66 X 67 X 68 X 69 (SEQ ID NO: 354)
  • X 64 is A, C, E, F, G, H, K, wherein X 62 is A, I, S or T; X 63 is L, Q, S, or V; X 64 is A, L, R, L, P, Q, R, S, or V; or S; X 65 is G; X 66 is I or L; X 67 is E, L, R, or Y; X 68 is F, H, L, R, or X 65 is D, E, G, S, or V; S; and X 69 is H, L, P, or S.
  • X 66 is A, I, L, M, or V; Subgenus 5.3: X 62 X 63 X 64 X 65 X 66 X 67 X 68 X 69 (SEQ ID NO: 355), X 67 is C, E, G, I, K, L, M, wherein X 62 is A, I, S or T; X 63 is L, S, or V; X 64 is A, R or S; N, Q, R, or Y; X 65 is G; X 66 is L; X 67 is E, L or R; X 68 is F, H, or S; and X 69 is X 68 is A, F, H, I, L, M, N, L, P, or S.
  • X 62 X 63 X 64 X 65 X 66 X 67 X 68 X 69 (SEQ ID NO: 356), X 69 is A, C, G, H, I, L, N, wherein X 62 is A, I, S or T; X 63 is L, S, or V; X 64 is R or S; X 65 is P, Q, R, S, T, V, or W. G; X 66 is L; X 67 is L or R; X 68 is F, H, or S; and X 69 is P or S.
  • Subgenus 5.5 X 62 X 63 X 64 X 65 X 66 X 67 X 68 X 69 (SEQ ID NO: 357), wherein X 62 is A, I, S or T; X 63 is L, S, or V; X 64 is R or S; X 65 is G; X 66 is L; X 67 is L or R; X 68 is S; and X 69 is P or S.
  • Subgenus 5.6 X 62 X 63 X 64 X 65 X 66 X 67 X 68 X 69 (SEQ ID NO: 358), wherein X 62 is T; X 63 is L, S, or V; X 64 is S; X 65 is G; X 66 is L; X 67 is R; X 68 is S; and X 69 is P.
  • X 62 is A, G, I, M, P, S, T, or V;
  • X 63 is L, Q, S, or V;
  • X 64 is A, C, F, K, L, Q, R or S;
  • X 65 is D, G, S, or V;
  • X 66 is L or M;
  • X 67 is G, I, L, M, N, Q, or R;
  • X 68 is I, N, P, or S; and
  • X 69 is A, H, I, N, Q, or S.
  • Subgenus 5.8 X 62 X 63 X 64 X 65 X 66 X 67 X 68 X 69 (SEQ ID NO: 360), wherein X 62 is A, I, or S; X 63 is L, Q, S, or V; X 64 is L, R or S; X 65 is G; X 66 is L; X 67 is L, M, or R; X 68 is S; and X 69 is A, H, N, Q, or S.
  • Subgenus 5.11 X 62 X 63 X 64 X 65 X 66 X 67 X 68 X 69 (SEQ ID NO: 363), wherein X 62 is A or S; X 63 is L or V; X 64 is S; X 65 is G; X 66 is L; X 67 is L or R; X 68 is S; and X 69 is H, or S.
  • X 73 is A, C, E, F, H, L, N, Subgenus 6.2: X 72 X 73 X 74 X 75 X 76 X 77 X 78 X 79 (SEQ ID NO: 373), R, S, or V; wherein X 72 is G, L or R, or S; X 73 is A or L; X 74 is A, E, N, Q, X 74 is A, D, E, K, N, P Q, or S; X 75 is A, E, N, S, or T; X 76 is L or M; X 77 is L or R; X 78 is S, T, or Y; A, L, or T; and X 79 is F, G, L, R, or S.
  • X 75 is A, E, G, H, K, L, N, Subgenus 6.3: X 72 X 73 X 74 X 75 X 76 X 77 X 78 X 79 (SEQ ID NO: 374), P, R, S, or T; wherein X 72 is L; X 73 is A or L; X 74 is E, N, Q, or S; X 75 is A or X 76 is I, K, L, M, N, R, T, S; X 76 is L or M; X 77 is R; X 78 is A or T; and X 79 is F, L, or R.
  • X 72 X 73 X 74 X 75 X 76 X 77 X 78 X 79 (SEQ ID NO: 375),
  • X 77 is A, D, E, I, K, L, P, wherein X 72 is L; X 73 is A or L; X 74 is E, N, Q, or S; X 75 is A or Q, R, S, T, V, or Y; S; X 76 is L or M; X 77 is R; X 78 is A; and X 79 is L or R.
  • X 78 is A, C, D, E, G, I, L, M, Q, R, S, T, or V; and X 79 is A, F, G, H, I, L, P, Q, R, S, T, or Y.
  • X 74 is A, D, E, K, N, P Q, S, T, or Y; Subgenus 6A.2: X 72 X 73 X 74 X 75 X 76 X 77 X 78 (SEQ ID X 75 is A, E, G, H, K, L, N, P, R, S, or NO: 377), wherein X 72 is A, E, L, or Q; X 73 is F, H, T; or N; X 74 is Q; X 75 is A; X 76 is L or T; X 77 is Q or R; X 76 is I, K, L, M, N, R, T, V or Y; and X 78 is I or M.
  • X 77 is A, D, E, I, K, L, P, Q, R, S, T, Subgenus 6A.3: X 72 X 73 X 74 X 75 X 76 X 77 X 78 (SEQ ID V, or Y; and NO: 378), wherein X 72 is A; X 73 is F, H, or N; X 74 is X 78 is A, C, D, E, G, I, L, M, Q, R, S, Q; X 75 is A; X 76 is L; X 77 is R; and X 78 is M. T, or V.
  • X 82 is L; X 83 is H, K, R, or Y; X 84 is A, L, or V; X 85 is T, or V; A, I, or L; X 86 is P; X 87 is G, R, or V; X 88 is T or W; and X 89 is X 85 is A, D, F, G, I, L, N, P, A, F, L, or S.
  • X 82 X 83 X 84 X 85 X 86 X 87 X 88 X 89 (SEQ ID NO: 398),
  • X 88 is A, C, E, F, H, I, L, N, wherein X 82 is A, F, L, Q, or S;
  • X 83 is A, E, G, H, K, Q, or Y; R, S, T, W, or Y; and
  • X 84 is A, G, K, S, or V;
  • X 85 is A, I, L, P, or T;
  • X 86 is A, P, or X 89 is A, F, G, I, L, M, R, S, T, or R;
  • X 87 is A, L, M, R, V, or Y;
  • X 88 is C, H, R, T, or W; and X 89 V.
  • X 82 is F or L;
  • X 83 is G, K, Q, or Y;
  • X 84 is A, G, S, or V;
  • X 85 is A, I, or L;
  • X 86 is P;
  • X 87 is A, R, or V;
  • X 88 is R or W; and
  • X 89 is A, F, L, or R.
  • Subgenus 7.6 X 82 X 83 X 84 X 85 X 86 X 87 X 88 X 89 (SEQ ID NO: 400), wherein X 82 is L; X 83 is K or Y; X 84 is A or S; X 85 is A, I, or L; X 86 is P; X 87 is A, R, or V; X 88 is W; and X 89 is A or F.
  • Subgenus 7.7 X 82 X 83 X 84 X 85 X 86 X 87 X 88 X 89 (SEQ ID NO: 401), wherein X 82 is L; X 83 is K or Y; X 84 is A; X 85 is A or I; X 86 is P; X 87 is R or V; X 88 is W; and X 89 is A or F.
  • X 92 X 93 X 94 X 95 X 96 X 97 X 98 (SEQ ID NO: 412), wherein X 93 is A, P, R, or T; X 92 is F, G, L, S, T, or V; X 93 is P; X 94 is A, E, H, K, N, Q, R, S, T, X 94 is A, E, F, G, H, I, or V; X 95 is A, G, H, N, P, or S; X 96 is I, L, M, or V; X 97 is F, I, L, K, L, N, P, Q, R, S, T, R, S, T, V, or Y; and X 98 is A, F, L, R, T, V, or Y.
  • X 92 X 93 X 94 X 95 X 96 X 97 X 98 (SEQ ID NO: 415), wherein X 98 is A, D, E, F, G, H, X 92 is F, L, or S; X 93 is P; X 94 is A, Q or S; X 95 is G; X 96 is I, L, or I, K, L, M, N, P, Q, R, S, T, M; X 97 is L or V; and X 98 is L. V, or Y.
  • Subgenus 8.6 X 92 X 93 X 94 X 95 X 96 X 97 X 98 (SEQ ID NO: 416), wherein X 92 is F, L, or S; X 93 is P; X 94 is A or S; X 95 is G; X 96 is I, L, or M; X 97 is L or V; and X 98 is L.
  • Subgenus 8.8 X 92 X 93 X 94 X 95 X 96 X 97 X 98 (SEQ ID NO: 418), wherein X 92 is L, S, or V; X 93 is P; X 94 is A, N, Q, or S; X 95 is H, N, P, or S; X 96 is F, I, L, or M; X 97 is I, L, S, or V; and X 98 is A, L, or Q.
  • Subgenus 8.9 X 92 X 93 X 94 X 95 X 96 X 97 X 98 (SEQ ID NO: 419), wherein X 92 is L; X 93 is P; X 94 is A, N, Q, or S; X 95 is H; X 96 is I or L; X 97 is V; and X 98 is L.
  • X 93 is A, P, R, or T; Subgenus 8A.2: X 92 X 93 X 94 X 95 X 96 X 97 X 98 X 99 (SEQ ID NO: 488), X 94 is A, E, F, G, H, I, K, wherein X 92 is F, G, L, S, T, or V; X 93 is P; X 94 is A, E, H, K, N, L, N, P, Q, R, S, T, or Q, R, S, T, or V; X 95 is A, G, H, N, P, or S; X 96 is I, L, M, or V; V; X 97 is F, I, L, R, S, T, V, or Y; X 98 is A, F, L, R, T, V, or Y; and X 95 is A, D, E, G, H, K, X 99 is A, D, G, L,
  • X 92 is F, L, or S
  • X 93 is P
  • X 94 is A, K, Q, R, or S
  • X 95 is R, S, V, W, or Y
  • X 96 is I, L, M, or V
  • X 97 is F, L, R, S, T, V, or Y
  • X 97 is A, C, F, G, H, I, K, X 98 is F, L, T, or V
  • X 99 is A, D, G, L, R, T, or V.
  • Subgenus 8A.6 X 92 X 93 X 94 X 95 X 96 X 97 X 98 X 99 (SEQ ID NO: 492), wherein X 92 is F, L, or S; X 93 is P; X 94 is A or S; X 95 is G; X 96 is I, L, or M; X 97 is L or V; X 98 is L; and X 99 is R.
  • Subgenus 8A.7 X 92 X 93 X 94 X 95 X 96 X 97 X 98 X 99 (SEQ ID NO: 493), wherein X 92 is F, G, L, M, P, S, V, or W; X 93 is P; X 94 is A, N, Q, or S; X 95 is A, D, G, H, M, N, P, or S; X 96 is F, I, L, M, or V; X 97 is A, I, L, M, S, or V; X 98 is A, G, I, L, M, N, P, Q, R, S, T, or Y; and X 99 is A, F, H, I, L, Q, R, T, V, W, or Y.
  • Subgenus 8A.8 X 92 X 93 X 94 X 95 X 96 X 97 X 98 X 99 (SEQ ID NO: 494), wherein X 92 is L, S, or V; X 93 is P; X 94 is A, N, Q, or S; X 95 is H, N, P, or S; X 96 is F, I, L, or M; X 97 is I, L, S, or V; X 98 is A, L, or Q; and X 99 is L, T, V, or Y.
  • Subgenus 8A.9 X 92 X 93 X 94 X 95 X 96 X 97 X 98 X 99 (SEQ ID NO: 495), wherein X 92 is L; X 93 is P; X 94 is A, N, Q, or S; X 95 is H; X 96 is I or L; X 97 is V; X 98 is L; and X 99 is L or V.
  • X 104 is A, G, H, L, N, P, R, S, T, Subgenus 9.2: X 102 X 103 X 104 X 105 X 106 X 107 X 108 X 109 (SEQ ID or V; NO: 427), wherein X 102 is F, G, I, R, or S; X 103 is L, P, R, X 105 is A, D, E, H, L, M, N, P, Q, or V; X 104 is A or H; X 105 is A, D, or R; X 106 is A or G; R, S, T, or V; X 107 is L or V; X 108 is H, L, M, R, S, or V; and X 109 is A, X 106 is A, G, R, S, or T; L, S, or V.
  • X 107 is C, F, L, M, S, V, W, or Y; Subgenus 9.3: X 102 X 103 X 104 X 105 X 106 X 107 X 108 X 109 (SEQ ID X 108 is A, E, F, G, H, I, L, M, N, NO: 428), wherein X 102 is G, R or S; X 103 is R or V; X 104 is Q, R, S, V, W, or Y; and A or H; X 105 is A, D, or R; X 106 is A or G; X 107 is L or V; X 109 is A, E, G, L, P, R, S, or V.
  • X 108 is H or R; and X 109 is A, L, S, or V.
  • Subgenus 9.4 X 102 X 103 X 104 X 105 X 106 X 107 X 108 X 109 (SEQ ID NO: 429), wherein X 102 is R; X 103 is R; X 104 is A or H; X 105 is A or D; X 106 is G; X 107 is L or V; X 108 is R; and X 109 is A, S, or V.
  • Subgenus 9.5 X 102 X 103 X 104 X 105 X 106 X 107 X 108 X 109 (SEQ ID NO: 430), wherein X 102 is D, F, G, I, L, R, S, or T; X 103 is E, L, M, R, S, T, or V; X 104 is H or N; X 105 is A, D, L, M, R, or T; X 106 is A, G, R, or T; X 107 is C, L, M, S, V, or W; X 108 is A, E, F, G, L, R, S, or W; and X 109 is A, G, L, P, R, S, or V.
  • Subgenus 9.6 X 102 X 103 X 104 X 105 X 106 X 107 X 108 X 109 (SEQ ID NO: 431), wherein X 102 is F, I, R, or S; X 103 is E, L, R, or V; X 104 is H; X 105 is D, M, R, or T; X 106 is A or G; X 107 is L, M, S, or V; X 108 is E, R, or S; and X 109 is A, P, S, or V.
  • Subgenus 9.7 X 102 X 103 X 104 X 105 X 106 X 107 X 108 X 109 (SEQ ID NO: 432), wherein X 102 is I or R; X 103 is E, R, or V; X 104 is H; X 105 is D, M, R, or T; X 106 is A or G; X 107 is L or V; X 108 is R or S; and X 109 is A, P, S, or V.
  • Subgenus 9.8 X 102 X 103 X 104 X 105 X 106 X 107 X 108 X 109 (SEQ ID NO: 433), wherein X 102 is I or R; X 103 is R; X 104 is H; X 105 is D; X 106 is A or G; X 107 is L or V; X 108 is R or S; and X 109 is A or S.
  • X 114 is A, H, K, L, N, P, Q, R, S, Subgenus 10.2: X 112 X 113 X 114 X 115 X 116 X 117 X 118 X 119 (SEQ T, or V; ID NO: 438), wherein X 112 is A, I, T, or V; X 113 is A, L, M, X 115 is A, D, F, G, H, I, L, P, R, Q, R, V, or Y; X 114 is A, N, S, or T; X 115 is G, L, S, or V; S, V, or Y; X 116 is L or V; X 117 is A, F, G, K, or S; X 118 is M, N, Q, R, X 116 is C, F, I, L, P, V, or Y; or V; and X 119 is I, L, or V.
  • X 117 is A, D, E, F, G, I, K, M, N, Subgenus 10.3: X 112 X 113 X 114 X 115 X 116 X 117 X 118 X 119 (SEQ R, S, T, V, or W; ID NO: 439), wherein X 112 is A, I, T, or V; X 113 is M, Q, or X 118 is A, D, E, F, H, K, L, M, N, Y; X 114 is A, N, or S; X 115 is G, L, S, or V; X 116 is L or V; Q, R, V, or Y; and X 117 is A, F, G, or S; X 118 is M, N, Q, or R; and X 119 is I, L, X 119 is A, F, I, L, M, or V.
  • Subgenus 10.4 X 112 X 113 X 114 X 115 X 116 X 117 X 118 X 119 (SEQ ID NO: 440), wherein X 112 is A, I, or V; X 113 is Y; X 114 is N or S; X 115 is G, L, or V; X 116 is L; X 117 is A, G, or S; X 118 is M, Q, or R; and X 119 is L or V.
  • Subgenus 10.5 X 112 X 113 X 114 X 115 X 116 X 117 X 118 X 119 (SEQ ID NO: 441), wherein X 112 is A, I, or V; X 113 is Y; X 114 is N or S; X 115 is G, L, or V; X 116 is L; X 117 is G or S; X 118 is M or R; and X 119 is L or V.
  • Subgenus 10.6 X 112 X 113 X 114 X 115 X 116 X 117 X 118 X 119 (SEQ ID NO: 442), wherein X 112 is A, I, or V; X 113 is Y; X 114 is N or S; X 115 is G, L, or V; X 116 is L; X 117 is S, X 118 is M or R; and X 119 is L or V.
  • Subgenus 10.7 X 112 X 113 X 114 X 115 X 116 X 117 X 118 X 119 (SEQ ID NO: 443), wherein X 112 is A; X 113 is Y; X 114 is N or S; X 115 is G or L; X 116 is L; X 117 is S; X 118 is R; and X 119 is L or V.
  • Subgenus 10.8 X 112 X 113 X 114 X 115 X 116 X 117 X 118 X 119 (SEQ ID NO: 444), wherein X 112 is A, D, G, I, L, N, P, S, T, V, W, or Y; X 113 is A, D, G, L, M, Q, S, or V; X 114 is H, K, N, P, Q, R, S, or T; X 115 is H, I, L, R, or V; X 116 is I, L, P, or V; X 117 is A, D, E, G, I, K, M, N, S, or T; X 118 is D, F, L, M, Q, R, or V; and X 119 is A, F, I, L, or V.
  • Subgenus 10.9 X 112 X 113 X 114 X 115 X 116 X 117 X 118 X 119 (SEQ ID NO: 445), wherein X 112 is A, I, T, or V; X 113 is A, D, G, L, M, Q, S, or V; X 114 is H, K, N, S, or T; X 115 is H, I, L, or V; X 116 is L; X 117 is A, G, K, or S; X 118 is L, M, Q, R, or V; and X 119 is A, L, or V.
  • Subgenus 10.11 X 112 X 113 X 114 X 115 X 116 X 117 X 118 X 119 (SEQ ID NO: 447), wherein X 112 is A or I; X 113 is A, L, or Q; X 114 is N or S; X 115 is L or V; X 116 is L; X 117 is A or S; X 118 is M or R; and X 119 is L or V.
  • Subgenus 10.12 X 112 X 113 X 114 X 115 X 116 X 117 X 118 X 119 (SEQ ID NO: 448), wherein X 112 is I; X 113 is A, L, or Q; X 114 is N; X 115 is L or V; X 116 is L; X 117 is A or S; X 118 is M or R; and X 119 is L or V.
  • Subgenus 10.13 X 112 X 113 X 114 X 115 X 116 X 117 X 118 X 119 (SEQ ID NO: 449), wherein X 112 is I; X 113 is A, L, or Q; X 114 is N; X 115 is L or V; X 116 is L; X 117 is S, X 118 is M; and X 119 is L or V.
  • X 124 is A, G, P, R, S, or T; Subgenus 11.2: X 122 X 123 X 124 X 125 X 126 X 127 X 128 X 129 (SEQ ID X 125 is H, I, L, P, R, or V; NO: 455), wherein X 122 is G, P, R, or S; X 123 is A or R; X 124 X 126 is L or W; is G, P, or S; X 125 is L or V; X 126 is W; X 127 is L or Y; X 128 X 127 is F, H, L, M, Q, S, V, or Y; is E or T; and X 129 is Q.
  • X 128 is A, D, E, I, K, P, R, S, T, or Subgenus 11.3: X 122 X 123 X 124 X 125 X 126 X 127 X 128 X 129 (SEQ ID V; and NO: 456), wherein X 122 is P; X 123 is A; X 124 is P or S; X 125 X 129 is A, E, F, G, H, I, L, N, P, Q, is L or V; X 126 is W; X 127 is Y; X 128 is T; and X 129 is Q. R, or V.
  • X 2 X 3 X 4 X 5 X 6 X 7 X 8 X 9 (SEQ ID NO: 460), wherein T, V, or Y; X 2 is A, P, or S; X 3 is L, S or V; X 4 is G, N, R, or S; X 5 is L, P, or S; X 3 is A, E, F, G, H, I, X 6 is L; X 7 is A, G, R, or S; X 8 is L, P, or V; and X 9 is F, L, P, or S.
  • X 2 X 3 X 4 X 5 X 6 X 7 X 8 X 9 (SEQ ID NO: 461), wherein T, V, W, or Y; X 2 is A, P, or S; X 3 is L, S, or V; X 4 is G, N, R, or S; X 5 is L, P, or X 4 is A, E, G, H, K, N, S; X 6 is L; X 7 is A, G, R, or S; X 8 is L or P; and X 9 is F, P, or S.
  • X 6 is I, L, M, Q, T, V, W, or Subgenus 12.5: X 2 X 3 X 4 X 5 X 6 X 7 X 8 X 9 (SEQ ID NO: 463), wherein Y; X 2 is A or S; X 3 is L; X 4 is G, N, or S; X 5 is L or S; X 6 is L; X 7 is R X 7 is A, D, G, H, K, L, or S; X 8 is L; and X 9 is P.
  • X 2 X 3 X 4 X 5 X 6 X 7 X 8 X 9 (SEQ ID NO: 464), wherein X 8 is A, D, E, F, G, I, X 2 is A, E, G, H, I, L, M, P, or S; X 3 is A, E, G, H, I, K, L, P, Q, R, K, L, M, P, Q, R, S, S, T, V, W, or Y; X 4 is A, G, N, R, S, T, or V; X 5 is A, G, H, L, N, V, W, or Y; and P, R, S, T, or V; X 6 is I, L, M, or Q; X 7 is A, D, G, K, L, N, Q, R, S, X 9 is A, F, G, I, L, M, or V; X 8
  • Subgenus 12.8 X 2 X 3 X 4 X 5 X 6 X 7 X 8 X 9 (SEQ ID NO: 466), wherein X 2 is A, P, or S; X 3 is H, S, or V; X 4 is G, N, or S; X 5 is L, P, or S; X 6 is L; X 7 is A, G, R, or S; X 8 is F, I, M, or P; and X 9 is P or R.
  • Subgenus 12.9 X 2 X 3 X 4 X 5 X 6 X 7 X 8 X 9 (SEQ ID NO: 467), wherein X 2 is A, P, or S; X 3 is S or V; X 4 is G, N, or S; X 5 is L; X 6 is L; X 7 is A, G or R; X 8 is F, I, or P; and X 9 is P.
  • Subgenus 12.10 X 2 X 3 X 4 X 5 X 6 X 7 X 8 X 9 (SEQ ID NO: 468), wherein X 2 is A, P, or S; X 3 is S or V; X 4 is G, N, or S; X 5 is L; X 6 is L; X 7 is A or R; X 8 is F or P; and X 9 is P.
  • Subgenus 12.11 X 2 X 3 X 4 X 5 X 6 X 7 X 8 X 9 (SEQ ID NO: 469), wherein X 2 is A or P; X 3 is S; X 4 is G or N; X 5 is L; X 6 is L; X 7 is R; X 8 is F; and X 9 is P.
  • X 14 is A, D, E, F, G, H, I, K, Subgenus 13.2: X 12 X 13 X 14 X 15 X 16 X 17 X 18 X 19 (SEQ ID NO: 476), L, M, N, P, Q, R, S, T, V, wherein X 12 is L, M, or V; X 13 is A, H, L, N, Q, S, or V; X 14 is or Y; A, L, M, Q, S, T, or V; X 15 is P; X 16 is A, F, G, I, L, R, S, V, or X 15 is A, E, G, N, P, Q, S, T, Y; X 17 is H, L, M, Q, or S; X 18 is A, D, G, H, R, or S; and X 19 is V, or W; A, F, G, L, R, or S.
  • X 16 is A, F, G, H, I, K, L, Subgenus 13.3: X 12 X 13 X 14 X 15 X 16 X 17 X 18 X 19 (SEQ ID NO: 477), M, N, P, Q, R, S, T, V, or wherein X 12 is L, M, or V; X 13 is A or L; X 14 is A, L, or S; X 15 Y; is P; X 16 is L or V; X 17 is H, L, or Q; X 18 is G or S; and X 19 is X 17 is A, D, E, F, G, H, I, L, G, R, or S.
  • X 12 X 13 X 14 X 15 X 16 X 17 X 18 X 19 (SEQ ID NO: 479), X 19 is A, D, F, G, H, I, L, wherein X 12 is L or V; X 13 is A or L; X 14 is L or S; X 15 is P; X 16 M, N, P, Q, R, S, T, V, W, or Y. is L; X 17 is H or L; X 18 is G; and X 19 is S.
  • Subgenus 13.6 X 12 X 13 X 14 X 15 X 16 X 17 X 18 X 19 (SEQ ID NO: 480), wherein X 12 is F, I, L, M, S, T, or V; X 13 is A, E, G, H, L, M, S, V, W, or Y; X 14 is A, D, E, G, K, L, M, N, Q, R, S, T, or V; X 15 is E, G, N, P, S, T, or V; X 16 is A, F, G, L, N, P, Q, R, S, V, or Y; X 17 is A, E, H, P, Q, or R; X 18 is D, E, G, N, R, S, or T; and X 19 is A, D, G, Q, S, T, or V.
  • Subgenus 13 X 12 X 13 X 14 X 15 X 16 X 17 X 18 X 19 (SEQ ID NO: 481), wherein X 12 is L, M, or V; X 13 is A or L; X 14 is A, L, Q, or S; X 15 is G, P, or T; X 16 is A, S, or Y; X 17 is H or P; X 18 is D or G; and X 19 is A, G or S.
  • Subgenus 13.7 X 12 X 13 X 14 X 15 X 16 X 17 X 18 X 19 (SEQ ID NO: 482), wherein X 12 is L or M; X 13 is A or L; X 14 is L; X 15 is G or P; X 16 is A or S; X 17 is H; X 18 is G; and X 19 is A or G.
  • the CM comprises an amino acid sequence selected from the group consisting of SEQ ID NOs: 352, 371, 394, 410, 425, 436, 453, 458, 473, 485, and 486. In some embodiments, the CM comprises an amino acid sequence selected from the group consisting of SEQ ID NO: 353-363, 372-375, 376-378, 395-401, 411-419, 426-433, 437-449, 454-456, 459-469, 475-482, and 487-495. In some embodiments, the CM comprises an amino acid sequence selected from the group consisting of SEQ ID NO: 353-363. In some embodiments, the CM comprises an amino acid sequence selected from the group consisting of 372-375.
  • the CM comprises an amino acid sequence selected from the group consisting of 376-378. In some embodiments, the CM comprises an amino acid sequence selected from the group consisting of 395-401. In some embodiments, the CM comprises an amino acid sequence selected from the group consisting of 411-419. In some embodiments, the CM comprises an amino acid sequence selected from the group consisting of 426-433. In some embodiments, the CM comprises an amino acid sequence selected from the group consisting of 437-449. In some embodiments, the CM comprises an amino acid sequence selected from the group consisting of 454-456. In some embodiments, the CM comprises an amino acid sequence selected from the group consisting of 459-469. In some embodiments, the CM comprises an amino acid sequence selected from the group consisting of 475-482. In some embodiments, the CM comprises an amino acid sequence selected from the group consisting of 487-495.
  • the CM comprises an amino sequence selected from the group consisting of SEQ ID NOs: 317, 324, 329 and 340. In some embodiments, the CM comprises an amino acid sequence selected from the group consisting of SEQ ID NOs: 318-323, 325-327, 330-335, and 341-347. In some embodiments, the CM comprises an amino acid sequence selected from the group consisting of SEQ ID NOs: 318-323. In some embodiments, the CM comprises an amino acid sequence selected from the group consisting of SEQ ID NOs: 325-327. In some embodiments, the CM comprises an amino acid sequence selected from the group consisting of SEQ ID NOs: 330-335. In some embodiments, the CM comprises an amino acid sequence selected from the group consisting of SEQ ID NOs: 341-347.
  • the CM comprises a core CM consensus 1 sequence comprising the amino acid sequence RPSPMWAY (SEQ ID NO: 21).
  • the CM comprises a core CM consensus 2 sequence comprising the amino acid sequence WDHPISLL (SEQ ID NO: 328). In some embodiments, the CM comprises a core CM consensus 2 sequence comprising the amino acid sequence WATPRPMR (SEQ ID NO: 22).
  • the CM comprises a core CM consensus 3 sequence comprising the amino acid sequence LTFPTYIF (SEQ ID NO: 336). In some embodiments, the CM comprises a core CM consensus 3 sequence comprising the amino acid sequence MTFPTYIF (SEQ ID NO: 337). In some embodiments, the CM comprises a core CM consensus 3 sequence comprising the amino acid sequence LTFPTYWF (SEQ ID NO: 338). In some embodiments, the CM comprises a core CM consensus 3 sequence comprising the amino acid sequence MTFPTYWF (SEQ ID NO: 339). In some embodiments, the CM comprises a core CM consensus 3 sequence comprising the amino acid sequence STFPFGMF (SEQ ID NO: 17).
  • the CM comprises a core CM consensus 4 sequence comprising the amino acid sequence DWLYWMGI (SEQ ID NO: 348). In some embodiments, the CM comprises a core CM consensus 4 sequence comprising the amino acid sequence DWLYWPGI (SEQ ID NO: 19). In some embodiments, the CM comprises a core CM consensus 4 sequence comprising the amino acid sequence DWLYWMSI (SEQ ID NO: 349). In some embodiments, the CM comprises a core CM consensus 4 sequence comprising the amino acid sequence DWLYWPSI (SEQ ID NO: 350). In some embodiments, the CM comprises a core CM consensus 4 sequence comprising the amino acid sequence HWHLGPPT (SEQ ID NO: 351).
  • the CM comprises a core CM consensus 5 sequence comprising the amino acid sequence ISSGLLSS (SEQ ID NO: 14). In some embodiments, the CM comprises a core CM consensus 5 sequence comprising the amino acid sequence SVSGLLSH (SEQ ID NO: 364). In some embodiments, the CM comprises a core CM consensus 5 sequence comprising the amino acid sequence SVSGLLSS (SEQ ID NO: 365). In some embodiments, the CM comprises a core CM consensus 5 sequence comprising the amino acid sequence SVSGLRSH (SEQ ID NO: 366). In some embodiments, the CM comprises a core CM consensus 5 sequence comprising the amino acid sequence SVSGLRSS (SEQ ID NO: 367).
  • the CM comprises a core CM consensus 5 sequence comprising the amino acid sequence TLSGLRSP (SEQ ID NO: 368). In some embodiments, the CM comprises a core CM consensus 5 sequence comprising the amino acid sequence TSSGLRSP (SEQ ID NO: 369). In some embodiments, the CM comprises a core CM consensus 5 sequence comprising the amino acid sequence TVSGLRSP (SEQ ID NO: 370).
  • the CM comprises a core CM consensus 6 sequence comprising the amino acid sequence AFQALRM (SEQ ID NO: 379). In some embodiments, the CM comprises a core CM consensus 6 sequence comprising the amino acid sequence AHQALRM (SEQ ID NO: 380). In some embodiments, the CM comprises a core CM consensus 6 sequence comprising the amino acid sequence ANQALRM (SEQ ID NO: 381). In some embodiments, the CM comprises a core CM consensus 6 sequence comprising the amino acid sequence ANQALRMA (SEQ ID NO: 382). In some embodiments, the CM comprises a core CM consensus 6 sequence comprising the amino acid sequence LLEALRAL (SEQ ID NO: 383).
  • the CM comprises a core CM consensus 6 sequence comprising the amino acid sequence LLNALRAL (SEQ ID NO: 384). In some embodiments, the CM comprises a core CM consensus 6 sequence comprising the amino acid sequence LLQALRAL (SEQ ID NO: 385). In some embodiments, the CM comprises a core CM consensus 6 sequence comprising the amino acid sequence LLSALRAL (SEQ ID NO: 386). In some embodiments, the CM comprises a core CM consensus 6 sequence comprising the amino acid sequence LLESLRAL (SEQ ID NO: 387). In some embodiments, the CM comprises a core CM consensus 6 sequence comprising the amino acid sequence LLNSLRAL (SEQ ID NO: 388).
  • the CM comprises a core CM consensus 6 sequence comprising the amino acid sequence LLQSLRAL (SEQ ID NO: 389). In some embodiments, the CM comprises a core CM consensus 6 sequence comprising the amino acid sequence LLSSLRAL (SEQ ID NO: 390). In some embodiments, the CM comprises a core CM consensus 6 sequence comprising the amino acid sequence QFQALRM (SEQ ID NO: 391). In some embodiments, the CM comprises a core CM consensus 6 sequence comprising the amino acid sequence QHQALRM (SEQ ID NO: 392). In some embodiments, the CM comprises a core CM consensus 6 sequence comprising the amino acid sequence QNQALRM (SEQ ID NO: 393). In some embodiments, the CM comprises a core CM consensus 6 sequence comprising the amino acid sequence QNQALRMA (SEQ ID NO: 15).
  • the CM comprises a core CM consensus 7 sequence comprising the amino acid sequence LKAAPRWA (SEQ ID NO: 24). In some embodiments, the CM comprises a core CM consensus 7 sequence comprising the amino acid sequence LKAAPVWA (SEQ ID NO: 403). In some embodiments, the CM comprises a core CM consensus 7 sequence comprising the amino acid sequence LKAAPRWF (SEQ ID NO: 404). In some embodiments, the CM comprises a core CM consensus 7 sequence comprising the amino acid sequence LKAAPVWF (SEQ ID NO: 405). In some embodiments, the CM comprises a core CM consensus 7 sequence comprising the amino acid sequence LYAAPRWA (SEQ ID NO: 406).
  • the CM comprises a core CM consensus 7 sequence comprising the amino acid sequence LYAAPVWA (SEQ ID NO: 407). In some embodiments, the CM comprises a core CM consensus 7 sequence comprising the amino acid sequence LYAAPRWF (SEQ ID NO: 408). In some embodiments, the CM comprises a core CM consensus 7 sequence comprising the amino acid sequence LYAAPVWF (SEQ ID NO: 409).
  • the CM comprises a core CM consensus 8 sequence comprising the amino acid sequence GPSHLVLT (SEQ ID NO: 25). In some embodiments, the CM comprises a core CM consensus 8 sequence comprising the amino acid sequence LPAGLLL (SEQ ID NO: 402). In some embodiments, the CM comprises a core CM consensus 8 sequence comprising the amino acid sequence LPAGLLLR (SEQ ID NO: 420). In some embodiments, the CM comprises a core CM consensus 8 sequence comprising the amino acid sequence LPAHLVLL (SEQ ID NO: 421). In some embodiments, the CM comprises a core CM consensus 8 sequence comprising the amino acid sequence LPSHLVLL (SEQ ID NO: 422).
  • the CM comprises a core CM consensus 8 sequence comprising the amino acid sequence LPAHLVLV (SEQ ID NO: 423). In some embodiments, the CM comprises a core CM consensus 8 sequence comprising the amino acid sequence LPSHLVLV (SEQ ID NO: 424).
  • the CM comprises a core CM consensus 9 sequence comprising the amino acid sequence RMHLRSLG (SEQ ID NO: 29). In some embodiments, the CM comprises a core CM consensus 9 sequence comprising the amino acid sequence RRHDGLRA (SEQ ID NO: 434). In some embodiments, the CM comprises a core CM consensus 9 sequence comprising the amino acid sequence RRHDGLRS (SEQ ID NO: 435).
  • the CM comprises a core CM consensus 10 sequence comprising the amino acid sequence AQNLLGMV (SEQ ID NO: 16). In some embodiments, the CM comprises a core CM consensus 10 sequence comprising the amino acid sequence IANLLSMV (SEQ ID NO: 450). In some embodiments, the CM comprises a core CM consensus 10 sequence comprising the amino acid sequence ILNLLSMV (SEQ ID NO: 451). In some embodiments, the CM comprises a core CM consensus 10 sequence comprising the amino acid sequence IQNLLSMV (SEQ ID NO: 452).
  • the CM comprises a core CM consensus 11 sequence comprising the amino acid sequence PAGLWLDP (SEQ ID NO: 33). In some embodiments, the CM comprises a core CM consensus 11 sequence comprising the amino acid sequence PASLWYTQ (SEQ ID NO: 457).
  • the CM comprises a core CM consensus 12 sequence comprising the amino acid sequence ALGLLRLP (SEQ ID NO: 470). In some embodiments, the CM comprises a core CM consensus 12 sequence comprising the amino acid sequence ALGLLSLP (SEQ ID NO: 471). In some embodiments, the CM comprises a core CM consensus 12 sequence comprising the amino acid sequence ASGLLRFP (SEQ ID NO: 472). In some embodiments, the CM comprises a core CM consensus 12 sequence comprising the amino acid sequence AVGLLAPP (SEQ ID NO: 31).
  • the CM comprises a core CM consensus 13 sequence comprising the amino acid sequence LAAPLGLL (SEQ ID NO: 30). In some embodiments, the CM comprises a core CM consensus 13 sequence comprising the amino acid sequence LLAPSHRA (SEQ ID NO: 32).
  • the CM comprises a core CM consensus 13 sequence comprising the amino acid sequence LLLPAHGG (SEQ ID NO: 474). In some embodiments, the CM comprises a core CM consensus 13 sequence comprising the amino acid sequence LLLPLLGS (SEQ ID NO: 483).
  • the CM is a substrate for at least two proteases.
  • at least one protease is an MMP and at least one protease is selected from the group consisting of those shown in Table 7.
  • ADAMS, ADAMTS e.g. ADAM8 ADAM9 ADAM10 ADAM12 ADAM15 ADAM17/TACE ADAMDEC1 ADAMTS1 ADAMTS4 ADAMTS5 Aspartate proteases, e.g., BACE Renin Aspartic cathepsins, e.g., Cathepsin D Cathepsin E Caspases, e.g., Caspase 1 Caspase 2 Caspase 3 Caspase 4 Caspase 5 Caspase 6 Caspase 7 Caspase 8 Caspase 9 Caspase 10 Caspase 14 Cysteine cathepsins, e.g., Cathepsin B Cathepsin C Cathepsin K Cathepsin L Cathepsin S Cathepsin V/L2 Cathepsin X/Z/P Cysteine proteinases, e.
  • Cysteine cathepsins e.g
  • the antibody includes at least a first CM and a second CM.
  • the first CM and the second CM are each polypeptides of no more than 15 amino acids long.
  • the first CM and the second CM in the antibody in the uncleaved state have the structural arrangement from N-terminus to C-terminus as follows: Agent-CM1-CM2-AB, AB-CM2-CM1-Agent, Agent-CM2-CM1-AB, or AB-CM1-CM2-Agent.
  • the activatable antibody includes a linking peptide between the agent and CM1.
  • the activatable antibody includes a linking peptide between CM1 and CM2.
  • the activatable antibody includes a linking peptide between CM2 and AB. In some embodiments, the activatable antibody includes a linking peptide between the agent and CM1 and a linking peptide between CM2 and AB. In some embodiments, the activatable antibody includes a linking peptide between agent and CM1 and a linking peptide between CM1 and CM2. In some embodiments, the activatable antibody includes a linking peptide between CM1 and CM2 and a linking peptide between CM2 and AB. In some embodiments, the activatable antibody includes a linking peptide between agent and CM1, a linking peptide between CM1 and CM2, and a linking peptide between CM2 and AB.
  • the activatable antibody includes at least a first CM that includes a substrate for at least one matrix metalloprotease (MMP) and a second CM that includes a substrate sequence.
  • CM2 matrix metalloprotease
  • Exemplary substrates for the second CM (CM2) include but are not limited to substrates cleavable by one or more of the following enzymes or proteases listed in Table 7.
  • the CM2 is selected for use with a specific protease.
  • the CM2 is a substrate for at least one protease selected from the group consisting of a matrix metalloprotease (MMP), a neutrophil elastase, u-type plasminogen activator (uPA, also referred to as urokinase), legumain, matriptase (also referred to herein as MT-SP1 or MTSP1), thrombin, a cysteine protease such as a cathepsin, ADAM17, BMP-1, HtrA1, and a TMPRSS such as TMPRSS3 or TMPRSS4.
  • MMP matrix metalloprotease
  • uPA u-type plasminogen activator
  • legumain legumain
  • matriptase also referred to herein as MT-SP1 or MTSP1
  • thrombin a cysteine protease
  • the CM2 is a substrate for a neutrophil elastase. In some embodiments, the CM2 is a substrate for uPA. In some embodiments, the CM2 is a substrate for legumain. In some embodiments, the CM2 is a substrate for matriptase. In some embodiments, the CM2 is a substrate for thrombin. In some embodiments, the CM2 is a substrate for a cysteine protease. In some embodiments, the CM2 is a substrate for a cathepsin. In some embodiments, the CM2 is a substrate for ADAM17. In some embodiments, the CM2 is a substrate for BMP-1.
  • the CM2 is a substrate for HtrA1. In some embodiments, the CM2 is a substrate for a TMPRSS. In some embodiments, the CM2 is a substrate for TMPRSS3. In some embodiments, the CM2 is a substrate for TMPRSS4.
  • suitable CM2 are cleaved by at least one protease and include the sequence TGRGPSWV (SEQ ID NO: 34); SARGPSRW (SEQ ID NO: 35); TARGPSFK (SEQ ID NO: 36); LSGRSDNH (SEQ ID NO: 37); GGWHTGRN (SEQ ID NO: 38); HTGRSGAL (SEQ ID NO: 39); PLTGRSGG (SEQ ID NO: 40); AARGPAIH (SEQ ID NO: 41); RGPAFNPM (SEQ ID NO: 42); SSRGPAYL (SEQ ID NO: 43); RGPATPIM (SEQ ID NO: 44); RGPA (SEQ ID NO: 45); GGQPSGMWGW (SEQ ID NO: 46); FPRPLGITGL (SEQ ID NO: 47); VHMPLGFLGP (SEQ ID NO: 48); SPLTGRSG (SEQ ID NO: 49); SAGFSLPA (SEQ ID NO: 126); LAPLGLQRR (SEQ ID NO:
  • the CM2 comprises the amino acid sequence TGRGPSWV (SEQ ID NO: 34). In some embodiments, the CM2 comprises the amino acid sequence SARGPSRW (SEQ ID NO: 35). In some embodiments, the CM2 comprises the amino acid sequence TARGPSFK (SEQ ID NO: 36). In some embodiments, the CM2 comprises the amino acid sequence LSGRSDNH (SEQ ID NO: 37). In some embodiments, the CM2 comprises the amino acid sequence GGWHTGRN (SEQ ID NO: 38). In some embodiments, the CM2 comprises the amino acid sequence HTGRSGAL (SEQ ID NO: 39). In some embodiments, the CM2 comprises the amino acid sequence PLTGRSGG (SEQ ID NO: 40).
  • the CM2 comprises the amino acid sequence AARGPAIH (SEQ ID NO: 41). In some embodiments, the CM2 comprises the amino acid sequence RGPAFNPM (SEQ ID NO: 42). In some embodiments, the CM2 comprises the amino acid sequence SSRGPAYL (SEQ ID NO: 43). In some embodiments, the CM2 comprises the amino acid sequence RGPATPIM (SEQ ID NO: 44). In some embodiments, the CM2 comprises the amino acid sequence RGPA (SEQ ID NO: 45). In some embodiments, the CM2 comprises the amino acid sequence GGQPSGMWGW (SEQ ID NO: 46). In some embodiments, the CM2 comprises the amino acid sequence FPRPLGITGL (SEQ ID NO: 47).
  • the CM2 comprises the amino acid sequence VHMPLGFLGP (SEQ ID NO: 48). In some embodiments, the CM2 comprises the amino acid sequence SPLTGRSG (SEQ ID NO: 49). In some embodiments, the CM2 comprises the amino acid sequence LAPLGLQRR (SEQ ID NO: 50). In some embodiments, the CM2 comprises the amino acid sequence SGGPLGVR (SEQ ID NO: 51). In some embodiments, the CM2 comprises the amino acid sequence PLGL (SEQ ID NO: 52). In some embodiments, the CM2 comprises the amino acid sequence GPRSFGL (SEQ ID NO: 315). In some embodiments, the CM2 comprises the amino acid sequence GPRSFG (SEQ ID NO: 316).
  • the CM2 is a substrate for at least one MMP. In some embodiments, the CM2 is a substrate for at least one MMP listed in the Table 7. In some embodiments, the CM2 is a substrate for MMP9. In some embodiments, the CM2 is a substrate for MMP14. In some embodiments, CM1 is substrate for a first MMP, and CM2 is a substrate for a second MMP, where the first MMP and the second MMP are different MMPs. In some embodiments, CM1 is a first substrate sequence for a MMP, and CM2 is a second substrate for the same MMP, where the CM1 and CM2 have different substrate sequences.
  • the CM2 is a substrate for two or more MMPs. In some embodiments, the CM2 is a substrate for at least MMP9 or MMP14. In some embodiments, the CM2 is a substrate for two or more MMPs. In some embodiments, the CM2 is a substrate for at least MMP9 and MMP14. In some embodiments, CM1 and CM2 are both substrates for MMP9. In some embodiments, CM1 and CM2 are both substrates for MMP14. In some embodiments, CM1 is a substrate for MMP9 and CM2 is a substrate for MMP14. In some embodiments, CM1 is a substrate for MMP14 and CM2 is a substrate for MMP9.
  • CM1 and/or CM2 is a substrate for an MMP and includes the sequence ISSGLLSS (SEQ ID NO: 14); QNQALRMA (SEQ ID NO: 15); AQNLLGMV (SEQ ID NO: 16); STFPFGMF (SEQ ID NO: 17); PVGYTSSL (SEQ ID NO: 18); DWLYWPGI (SEQ ID NO: 19); MIAPVAYR (SEQ ID NO: 20); RPSPMWAY (SEQ ID NO: 21); WATPRPMR (SEQ ID NO: 22); FRLLDWQW (SEQ ID NO: 23); LKAAPRWA (SEQ ID NO: 24); GPSHLVLT (SEQ ID NO: 25); LPGGLSPW (SEQ ID NO: 26); MGLFSEAG (SEQ ID NO: 27); SPLPLRVP (SEQ ID NO: 28); RMHLRSLG (SEQ ID NO: 29); LAAPLGLL (SEQ ID NO: 30); AVGLLAPP (SEQ ID NO: 14); QNQALR
  • the first cleaving agent and the second cleaving agent are the same protease, and the first CM and the second CM are different substrates for the enzyme. In some embodiments, the first cleaving agent and the second cleaving agent are different proteases. In some embodiments, the first cleaving agent and the second cleaving agent are co-localized in the target tissue. In some embodiments, the first CM and the second CM are cleaved by at least one cleaving agent in the target tissue.
  • the agent conjugated to the AB is a therapeutic agent.
  • the agent is an antineoplastic agent.
  • the agent is a toxin or fragment thereof.
  • a fragment of a toxin is a fragment that retains toxic activity.
  • the agent is conjugated to the AB via a cleavable linker.
  • the agent is conjugated to the AB via a linker that includes at least one MMP-cleavable substrate sequence.
  • the agent is conjugated to the AB via a noncleavable linker.
  • the agent is a microtubule inhibitor.
  • the agent is a nucleic acid damaging agent, such as a DNA alkylator or DNA intercalator, or other DNA damaging agent.
  • the agent is an agent selected from the group listed in Table 3.
  • the agent is a dolastatin.
  • the agent is an auristatin or derivative thereof.
  • the agent is auristatin E or a derivative thereof.
  • the agent is monomethyl auristatin E (MMAE).
  • the agent is monomethyl auristatin D (MMAD).
  • the agent is a maytansinoid or maytansinoid derivative.
  • the agent is DM1 or DM4.
  • the agent is a duocarmycin or derivative thereof.
  • the agent is a calicheamicin or derivative thereof.
  • the agent is a pyrrolobenzodiazepine.
  • the agent is an anti-inflammatory agent.
  • the antibody also includes a detectable moiety.
  • the detectable moiety is a diagnostic agent.
  • the conjugated antibody and/or conjugated activatable antibody includes a detectable label.
  • the detectable label includes an imaging agent, a contrasting agent, an enzyme, a fluorescent label, a chromophore, a dye, one or more metal ions, or a ligand-based label.
  • the imaging agent comprises a radioisotope.
  • the radioisotope is indium or technetium.
  • the contrasting agent comprises iodine, gadolinium or iron oxide.
  • the enzyme comprises horseradish peroxidase, alkaline phosphatase, or ⁇ -galactosidase.
  • the fluorescent label comprises yellow fluorescent protein (YFP), cyan fluorescent protein (CFP), green fluorescent protein (GFP), modified red fluorescent protein (mRFP), red fluorescent protein tdimer2 (RFP tdimer2), HCRED, or a europium derivative.
  • the luminescent label comprises an N-methylacrydium derivative.
  • the label comprises an Alexa Fluor® label, such as Alex Fluor® 680 or Alexa Fluor® 750.
  • the ligand-based label comprises biotin, avidin, streptavidin or one or more haptens.
  • the AB of the antibody naturally contains one or more disulfide bonds. In some embodiments, the AB can be engineered to include one or more disulfide bonds.
  • the antibody and/or conjugated antibody is monospecific. In some embodiments, the antibody and/or conjugated antibody is multispecific, referred to herein as multispecific antibodies and/or conjugated multispecific antibodies. In some embodiments, the multispecific antibody and/or conjugated multispecific antibody is bispecific or trifunctional. In some embodiments, the antibody and/or conjugated antibody is formulated as part of a pro-Bispecific T Cell Engager (pro-BITE) molecule. In some embodiments, the antibody and/or conjugated antibody is formulated as part of a pro-Chimeric Antigen Receptor (pro-CAR) modified T cell or other engineered receptor.
  • pro-BITE pro-Bispecific T Cell Engager
  • pro-CAR pro-Chimeric Antigen Receptor
  • the activatable antibody and/or conjugated activatable antibody is monospecific. In some embodiments, the activatable antibody and/or conjugated activatable antibody is multispecific, referred to herein as multispecific activatable antibodies and/or conjugated multispecific activatable antibodies. As used herein, terms such as “activatable antibody” and all grammatical variations thereof, unless otherwise noted, are intended to encompass, but are not limited to embodiments where the activatable antibody is a multispecific activatable antibody of the disclosure.
  • conjugated activatable antibody and all grammatical variations thereof, unless otherwise noted, are intended to encompass, but are not limited to embodiments where the conjugated activatable antibody is a conjugated multispecific activatable antibody of the disclosure.
  • the multispecific activatable antibody and/or conjugated multispecific activatable antibody is bispecific or trifunctional.
  • the activatable antibody and/or conjugated activatable antibody is formulated as part of a pro-Bispecific T Cell Engager (pro-BITE) molecule.
  • pro-CAR pro-Chimeric Antigen Receptor
  • the activatable antibodies, conjugated activatable antibodies, multispecific activatable antibodies, and/or conjugated multispecific activatable antibodies described herein are used in conjunction with one or more additional agents or a combination of additional agents.
  • additional agents include current pharmaceutical and/or surgical therapies for an intended application, such as, for example, cancer.
  • the activatable antibodies, conjugated activatable antibodies, multispecific activatable antibodies, and/or conjugated multispecific activatable antibodies can be used in conjunction with an additional chemotherapeutic or anti-neoplastic agent.
  • the activatable antibodies described herein in an activated state bind a given target and include (i) an antibody or an antigen binding fragment thereof (AB) that specifically binds to the target; (ii) a masking moiety (MM) that inhibits the binding of the AB to the target in an uncleaved state; and (c) a cleavable moiety (CM) coupled to the AB, wherein the CM is a polypeptide that functions as a substrate for a matrix metalloprotease.
  • AB antibody or an antigen binding fragment thereof
  • MM masking moiety
  • CM cleavable moiety
  • the activatable antibody in the uncleaved state has the structural arrangement from N-terminus to C-terminus as follows: MM-CM-AB or AB-CM-MM.
  • the activatable antibody comprises a linking peptide between the MM and the CM.
  • the activatable antibody comprises a linking peptide between the CM and the AB.
  • the activatable antibody comprises a first linking peptide (LP1) and a second linking peptide (LP2), and the activatable antibody in the uncleaved state has the structural arrangement from N-terminus to C-terminus as follows: MM-LP1-CM-LP2-AB or AB-LP2-CM-LP 1-MM.
  • each of LP1 and LP2 is a peptide of about 1 to 20 amino acids in length.
  • the two linking peptides need not be identical to each other.
  • At least one of LP1 or LP2 comprises an amino acid sequence selected from the group consisting of (GS) n , (GGS) n , (GSGGS) n (SEQ ID NO: 1) and (GGGS) n (SEQ ID NO: 2), where n is an integer of at least one.
  • At least one of LP1 or LP2 comprises an amino acid sequence selected from the group consisting of GGSG (SEQ ID NO: 3), GGSGG (SEQ ID NO: 4), GSGSG (SEQ ID NO: 5), GSGGG (SEQ ID NO: 6), GGGSG (SEQ ID NO: 7), and GSSSG (SEQ ID NO: 8).
  • LP1 comprises the amino acid sequence GSSGGSGGSGGSG (SEQ ID NO: 9), GSSGGSGGSGG (SEQ ID NO: 10), GSSGGSGGSGGS (SEQ ID NO: 11), GSSGGSGGSGGSGGGS (SEQ ID NO: 155), GSSGGSGGSG (SEQ ID NO: 156), or GSSGGSGGSGS (SEQ ID NO: 157).
  • LP2 comprises the amino acid sequence GSS, GGS, GGGS (SEQ ID NO: 158), GSSGT (SEQ ID NO: 12) or GSSG (SEQ ID NO: 13).
  • the AB has an equilibrium dissociation constant of about 100 nM or less for binding to the target.
  • the activatable antibody includes an antibody or antigen-binding fragment thereof that specifically binds a target.
  • the antibody or immunologically active fragment thereof that binds the target is a monoclonal antibody, domain antibody, single chain, Fab fragment, a F(ab) 2 fragment, a scFv, a scab, a dAb, a single domain heavy chain antibody, or a single domain light chain antibody.
  • such an antibody or immunologically active fragment thereof that binds the target is a mouse, other rodent, chimeric, humanized or fully human monoclonal antibody.
  • the activatable antibody is a multispecific activatable antibody.
  • the multispecific activatable antibodies provided herein are multispecific antibodies that recognize two or more different antigens or epitopes and that include at least one masking moiety (MM) linked to at least one antigen- or epitope-binding domain of the multispecific antibody such that coupling of the MM reduces the ability of the antigen- or epitope-binding domain to bind its target.
  • the MM is coupled to the antigen- or epitope-binding domain of the multispecific antibody via a cleavable moiety (CM) that functions as a substrate for at least one MMP protease.
  • CM cleavable moiety
  • the activatable multispecific antibodies provided herein are stable in circulation, activated at intended sites of therapy and/or diagnosis but not in normal, i.e., healthy tissue, and, when activated, exhibit binding to a target that is at least comparable to the corresponding, unmodified multispecific antibody.
  • the activatable antibody and/or conjugated activatable antibody provided herein includes at least a first antibody or antigen binding fragment thereof (AB1) that specifically binds a Jagged target, e.g., Jagged 1 and/or Jagged 2, and that contains a combination of a VH CDR1 sequence, a VH CDR2 sequence, and a VH CDR3 sequence, wherein at least one of the VH CDR1 sequence, the VH CDR2 sequence, and the VH CDR3 sequence is selected from a VH CDR1 that sequence includes at least the amino acid sequence SYAMS (SEQ ID NO: 498); a VH CD2 sequence that includes at least the amino acid sequence SIDPEGRQTYYADSVKG (SEQ ID NO: 499); a VH CDR3 sequence that includes at least the amino acid sequence DIGGRSAFDY (SEQ ID NO: 500), and combinations thereof
  • AB1 antibody or antigen binding fragment thereof
  • the activatable antibody and/or conjugated activatable antibody provided herein includes at least a first antibody or antigen binding fragment thereof (AB1) that specifically binds a Jagged target, e.g., Jagged 1 and/or Jagged 2, and that contains a combination of a VL CDR1 sequence, a VL CDR2 sequence, and a VL CDR3 sequence, wherein at least one of the VL CDR1 sequence, the VL CDR2 sequence, and the VL CDR3 sequence is selected from a VL CDR1 sequence that includes at least the amino acid sequence RASQSISSY (SEQ ID NO: 501); a VL CDR2 sequence that includes at least the amino acid sequence AASSLQS (SEQ ID NO: 502); a VL CDR3 sequence that includes at least the amino acid sequence QQTVVAPPL (SEQ ID NO: 503), and combinations
  • the activatable antibody and/or conjugated activatable antibody provided herein includes at least a first antibody or antigen binding fragment thereof (AB1) that specifically binds a Jagged target, e.g., Jagged 1 and/or Jagged 2, and that contains a combination of a VH CDR1 sequence, a VH CDR2 sequence, and a VH CDR3 sequence, wherein at least one of the VH CDR1 sequence, the VH CDR2 sequence, and the VH CDR3 sequence is selected from a VH CDR1 sequence that includes a sequence that is at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more identical to the amino acid sequence SYAMS (SEQ ID NO: 498); a VH CD2 sequence that includes a sequence that is at least 90%, 91%, 92%, 93%, 94%
  • the activatable antibody and/or conjugated activatable antibody provided herein includes at least a first antibody or antigen binding fragment thereof (AB1) that specifically binds a Jagged target, e.g., Jagged 1 and/or Jagged 2, and that contains a combination of a VL CDR1 sequence, a VL CDR2 sequence, and a VL CDR3 sequence, wherein at least one of the VL CDR1 sequence, the VL CDR2 sequence, and the VL CDR3 sequence is selected from a VL CDR1 sequence that includes a sequence that is at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more identical to the amino acid sequence RASQSISSY (SEQ ID NO: 501); a VL CDR2 sequence that includes a sequence that is at least 90%, 91%, 92%, 9
  • the activatable antibody and/or conjugated activatable antibody provided herein includes at least a first antibody or antigen binding fragment thereof (AB1) that specifically binds a Jagged target, e.g., Jagged 1 and/or Jagged 2, and that contains a combination of a VH CDR1 sequence, a VH CDR2 sequence, a VH CDR3 sequence, a VL CDR1 sequence, a VL CDR2 sequence, and a VL CDR3 sequence, wherein the VH CDR1 sequence includes at least the amino acid sequence SYAMS (SEQ ID NO: 498); the VH CD2 sequence includes at least the amino acid sequence SIDPEGRQTYYADSVKG (SEQ ID NO: 499); the VH CDR3 sequence includes at least the amino acid sequence DIGGRSAFDY (SEQ ID NO: 500); the VL CDR1 sequence includes at least the amino acid
  • the activatable antibody and/or conjugated activatable antibody provided herein includes at least a first antibody or antigen binding fragment thereof (AB1) that specifically binds a Jagged target, e.g., Jagged 1 and/or Jagged 2, and that contains a combination of a VH CDR1 sequence, a VH CDR2 sequence, a VH CDR3 sequence, a VL CDR1 sequence, a VL CDR2 sequence, and a VL CDR3 sequence, wherein the VH CDR1 sequence includes a sequence that is at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more identical to the amino acid sequence SYAMS (SEQ ID NO: 498); the VH CD2 sequence includes a sequence that is at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 9
  • the activatable antibody and/or conjugated activatable antibody provided herein includes at least a first antibody or antigen binding fragment thereof (AB1) that specifically binds Epidermal Growth Factor Receptor (EGFR) and that contains a combination of a VH CDR1 sequence, a VH CDR2 sequence, and a VH CDR3 sequence, wherein at least one of the VH CDR1 sequence, the VH CDR2 sequence, and the VH CDR3 sequence is selected from a VH CDR1 sequence that includes at least the amino acid sequence NYGVH (SEQ ID NO: 504); a VH CD2 sequence that includes at least the amino acid sequence VIWSGGNTDYNTPFTS (SEQ ID NO: 505); a VH CDR3 sequence that includes at least the amino acid sequence ALTYYDYEFAY (SEQ ID NO: 506); and combinations thereof.
  • AB1 that specifically binds Epidermal Growth Factor Receptor
  • the activatable antibody and/or conjugated activatable antibody provided herein includes at least a first antibody or antigen binding fragment thereof (AB1) that specifically binds EGFR and that contains a combination of a VL CDR1 sequence, a VL CDR2 sequence, and a VL CDR3 sequence, wherein at least one of the VL CDR1 sequence, the VL CDR2 sequence, and the VL CDR3 sequence is selected from a VL CDR1 sequence that includes at least the amino acid sequence RASQSIGTNIH (SEQ ID NO: 507); a VL CDR2 sequence that includes at least the amino acid sequence KYASESIS (SEQ ID NO: 508); and a VL CDR3 sequence that includes at least the amino acid sequence QQNNNWPTT (SEQ ID NO: 509), and combinations thereof.
  • AB1 antibody or antigen binding fragment thereof
  • the activatable antibody and/or conjugated activatable antibody provided herein includes at least a first antibody or antigen binding fragment thereof (AB1) that specifically binds EGFR and that contains a combination of a VH CDR1 sequence, a VH CDR2 sequence, and a VH CDR3 sequence, wherein at least one of the VH CDR1 sequence, the VH CDR2 sequence, and the VH CDR3 sequence is selected from a VH CDR1 sequence that includes a sequence that is at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more identical to the amino acid sequence NYGVH (SEQ ID NO: 504); a VH CD2 sequence that includes a sequence that is at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more identical
  • the activatable antibody and/or conjugated activatable antibody provided herein includes at least a first antibody or antigen binding fragment thereof (AB1) that specifically binds EGFR and that contains a combination of a VL CDR1 sequence, a VL CDR2 sequence, and a VL CDR3 sequence, wherein at least one of the VL CDR1 sequence, the VL CDR2 sequence, and the VL CDR3 sequence is selected from a VL CDR1 sequence that includes a sequence that is at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more identical to the amino acid sequence RASQSIGTNIH (SEQ ID NO: 507); a VL CDR2 sequence that includes a sequence that is at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%
  • the activatable antibody and/or conjugated activatable antibody provided herein includes at least a first antibody or antigen binding fragment thereof (AB1) that specifically binds EGFR and that contains a combination of a VH CDR1 sequence, a VH CDR2 sequence, a VH CDR3 sequence, a VL CDR1 sequence, a VL CDR2 sequence, and a VL CDR3 sequence, wherein the VH CDR1 sequence includes at least the amino acid sequence NYGVH (SEQ ID NO: 504); the VH CD2 sequence includes at least the amino acid sequence VIWSGGNTDYNTPFTS (SEQ ID NO: 505); the VH CDR3 sequence includes at least the amino acid sequence ALTYYDYEFAY (SEQ ID NO: 506); the VL CDR1 sequence includes at least the amino acid sequence RASQSIGTNIH (SEQ ID NO: 507);
  • the activatable antibody and/or conjugated activatable antibody provided herein includes at least a first antibody or antigen binding fragment thereof (AB1) that specifically binds EGFR and that contains a combination of a VH CDR1 sequence, a VH CDR2 sequence, a VH CDR3 sequence, a VL CDR1 sequence, a VL CDR2 sequence, and a VL CDR3 sequence, wherein the VH CDR1 sequence includes a sequence that is at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more identical to the amino acid sequence NYGVH (SEQ ID NO: 504); the VH CD2 sequence includes a sequence that is at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more identical to the amino acid sequence VIWSGG
  • the activatable antibody and/or conjugated activatable antibody provided herein includes at least a heavy chain amino acid sequence selected from the group consisting of SEQ ID NOs: 54, 56, 57, 58, 61, 63, 65, 68, 70, 72, 76, 78, 80, 82, 84, 86, 88, 90, 92, 94, 96, 98, 100, 102, 104, 106, 108, 110, 112, and 114.
  • the activatable antibody and/or conjugated activatable antibody provided herein includes at least a light chain amino acid sequence selected from the group consisting of SEQ ID NOs: 55, 59, 60, 62, 64, 67, 69, 71, 73, 75, 77, 79, 81, 83, 85, 87, 89, 91, 93, 95, 97, 99, 101, 103, 105, 107, 109, 111, and 113.
  • the activatable antibody and/or conjugated activatable antibody provided herein includes at least a heavy chain amino acid sequence selected from the group consisting of SEQ ID NOs: 54, 56, 57, 58, 61, 63, 65, 68, 70, 72, 76, 78, 80, 82, 84, 86, 88, 90, 92, 94, 96, 98, 100, 102, 104, 106, 108, 110, 112, and 114 and a light chain amino acid sequence selected from the group consisting of SEQ ID NOs: 55, 59, 60, 62, 64, 67, 69, 71, 73, 75, 77, 79, 81, 83, 85, 87, 89, 91, 93, 95, 97, 99, 101, 103, 105, 107, 109, 111, and 113.
  • the activatable antibody and/or conjugated activatable antibody provided herein includes at least a heavy chain amino acid sequence that is at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more identical to an amino acid sequence selected from the group consisting of SEQ ID NOs: 54, 56, 57, 58, 61, 63, 65, 68, 70, 72, 76, 78, 80, 82, 84, 86, 88, 90, 92, 94, 96, 98, 100, 102, 104, 106, 108, 110, 112, and 114.
  • the activatable antibody and/or conjugated activatable antibody provided herein includes at least a light chain amino acid sequence that is at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more identical to an amino acid sequence selected from the group consisting of SEQ ID NOs: 55, 59, 60, 62, 64, 67, 69, 71, 73, 75, 77, 79, 81, 83, 85, 87, 89, 91, 93, 95, 97, 99, 101, 103, 105, 107, 109, 111, and 113.
  • the activatable antibody and/or conjugated activatable antibody provided herein includes at least a heavy chain amino acid sequence that is at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more identical to an amino acid sequence selected from the group consisting of SEQ ID NOs: 54, 56, 57, 58, 61, 63, 65, 68, 70, 72, 76, 78, 80, 82, 84, 86, 88, 90, 92, 94, 96, 98, 100, 102, 104, 106, 108, 110, 112, and 114 and a light chain amino acid sequence that is at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more identical to an amino acid sequence selected from the group consisting of SEQ ID NOs:
  • the MM has an equilibrium dissociation constant for binding to the AB that is greater than the equilibrium dissociation constant of the AB to the target.
  • the MM has an equilibrium dissociation constant for binding to the AB that is no more than the equilibrium dissociation constant of the AB to the target.
  • the MM does not interfere or compete with the AB for binding to the target in a cleaved state.
  • the MM is a polypeptide of about 2 to 40 amino acids in length.
  • the MM is a polypeptide of up to about 40 amino acids in length.
  • the MM polypeptide sequence is different from that of any natural binding partner of the AB. In some embodiments, the MM polypeptide sequence is no more than 50% identical to any natural binding partner of the AB. In some embodiments, the MM polypeptide sequence is no more than 40%, 30%, 25%, 20%, 15%, or 10% identical to any natural binding partner of the AB.
  • the coupling of the MM to the AB reduces the ability of the AB to bind its target such that the dissociation constant (K d ) of the AB when coupled to the MM towards the target is at least two times greater than the K d of the AB when not coupled to the MM towards the target.
  • the coupling of the MM to the AB reduces the ability of the AB to bind its target such that the dissociation constant (K d ) of the AB when coupled to the MM towards the target is at least three times greater than the K d of the AB when not coupled to the MM towards the target.
  • the coupling of the MM to the AB reduces the ability of the AB to bind its target such that the dissociation constant (K d ) of the AB when coupled to the MM towards the target is at least five times greater than the K d of the AB when not coupled to the MM towards the target.
  • the coupling of the MM to the AB reduces the ability of the AB to bind its target such that the dissociation constant (K d ) of the AB when coupled to the MM towards the target is at least 10 times greater than the K d of the AB when not coupled to the MM towards the target.
  • the coupling of the MM to the AB reduces the ability of the AB to bind its target such that the dissociation constant (K d ) of the AB when coupled to the MM towards the target is at least 20 times greater than the K d of the AB when not coupled to the MM towards the target.
  • the coupling of the MM to the AB reduces the ability of the AB to bind the target such that the dissociation constant (K d ) of the AB when coupled to the MM towards the target is at least 40 times greater than the K d of the AB when not coupled to the MM towards the target.
  • the coupling of the MM to the AB reduces the ability of the AB to bind the target such that the dissociation constant (K d ) of the AB when coupled to the MM towards the target is at least 100 times greater than the K d of the AB when not coupled to the MM towards the target.
  • the coupling of the MM to the AB reduces the ability of the AB to bind the target such that the dissociation constant (K d ) of the AB when coupled to the MM towards the target is at least 1000 times greater than the K d of the AB when not coupled to the MM towards the target.
  • the coupling of the MM to the AB reduces the ability of the AB to bind the target such that the dissociation constant (K d ) of the AB when coupled to the MM towards the target is at least 10,000 times greater than the K d of the AB when not coupled to the MM towards the target.
  • the MMP protease is co-localized with the target in a tissue, and the MMP cleaves the CM in the activatable antibody when the activatable antibody is exposed to the MMP.
  • the MM in the presence of the target, reduces the ability of the AB to bind the target by at least 90% when the CM is uncleaved, as compared to when the CM is cleaved when assayed in vitro using a target displacement assay such as, for example, the assay described in PCT Publication Nos. WO 2009/025846 and WO 2010/081173.
  • a target displacement assay such as, for example, the assay described in PCT Publication Nos. WO 2009/025846 and WO 2010/081173.
  • the CM is positioned in the activatable antibody such that in the uncleaved state, binding of the activatable antibody to the target is reduced to occur with an equilibrium dissociation constant that is at least twofold greater than the equilibrium dissociation constant of an unmodified AB binding to the target, whereas in the cleaved state (i.e., when the activatable antibody is in the cleaved state), the AB binds the target.
  • the CM is positioned in the activatable antibody such that in the uncleaved state, binding of the activatable antibody to the target is reduced to occur with an equilibrium dissociation constant that is at least five-fold greater than the equilibrium dissociation constant of an unmodified AB binding to the target, whereas in the cleaved state (i.e., when the activatable antibody is in the cleaved state), the AB binds the target.
  • the CM is positioned in the activatable antibody such that in the uncleaved state, binding of the activatable antibody to the target is reduced to occur with an equilibrium dissociation constant that is at least ten-fold greater than the equilibrium dissociation constant of an unmodified AB binding to the target, whereas in the cleaved state (i.e., when the activatable antibody is in the cleaved state), the AB binds the target.
  • the CM is positioned in the activatable antibody such that in the uncleaved state, binding of the activatable antibody to the target is reduced to occur with an equilibrium dissociation constant that is at least 20-fold greater than the equilibrium dissociation constant of an unmodified AB binding to the target, whereas in the cleaved state (i.e., when the activatable antibody is in the cleaved state), the AB binds the target.
  • the CM is positioned in the activatable antibody such that in the uncleaved state, binding of the activatable antibody to the target is reduced to occur with an equilibrium dissociation constant that is at least 40-fold greater than the equilibrium dissociation constant of an unmodified AB binding to the target, whereas in the cleaved state, the AB binds the target.
  • the CM is positioned in the activatable antibody such that in the uncleaved state, binding of the activatable antibody to the target is reduced to occur with an equilibrium dissociation constant that is at least 50-fold greater than the equilibrium dissociation constant of an unmodified AB binding to the target, whereas in the cleaved state, the AB binds the target.
  • the CM is positioned in the activatable antibody such that in the uncleaved state, binding of the activatable antibody to the target is reduced to occur with an equilibrium dissociation constant that is at least 100-fold greater than the equilibrium dissociation constant of an unmodified AB binding to the target, whereas in the cleaved state, the AB binds the target.
  • the CM is positioned in the activatable antibody such that in the uncleaved state, binding of the activatable antibody to the target is reduced to occur with an equilibrium dissociation constant that is at least 200-fold greater than the equilibrium dissociation constant of an unmodified AB binding to the target, whereas in the cleaved state, the AB binds the target.
  • the CM is a polypeptide of up to 15 amino acids in length.
  • the CM is a substrate for at least one matrix metalloprotease (MMP).
  • MMPs include MMP1; MMP2; MMP3; MMP7; MMP8; MMP9; MMP10; MMP11; MMP12; MMP13; MMP14; MMP15; MMP16; MMP17; MMP19; MMP20; MMP23; MMP24; MMP26; and MMP27.
  • the CM is a substrate for MMP9, MMP14, MMP1, MMP3, MMP13, MMP17, MMP11, and MMP19.
  • the CM is a substrate for MMP9.
  • the CM is a substrate for MMP14.
  • the CM is a substrate for two or more MMPs. In some embodiments, the CM is a substrate for at least MMP9 and MMP14. In some embodiments, the CM comprises two or more substrates for the same MMP. In some embodiments, the CM comprises at least two or more MMP9 substrates. In some embodiments, the CM comprises at least two or more MMP14 substrates.
  • the CM is a substrate for an MMP and includes the sequence ISSGLLSS (SEQ ID NO: 14); QNQALRMA (SEQ ID NO: 15); AQNLLGMV (SEQ ID NO: 16); STFPFGMF (SEQ ID NO: 17); PVGYTSSL (SEQ ID NO: 18); DWLYWPGI (SEQ ID NO: 19); MIAPVAYR (SEQ ID NO: 20); RPSPMWAY (SEQ ID NO: 21); WATPRPMR (SEQ ID NO: 22); FRLLDWQW (SEQ ID NO: 23); LKAAPRWA (SEQ ID NO: 24); GPSHLVLT (SEQ ID NO: 25); LPGGLSPW (SEQ ID NO: 26); MGLFSEAG (SEQ ID NO: 27); SPLPLRVP (SEQ ID NO: 28); RMHLRSLG (SEQ ID NO: 29); LAAPLGLL (SEQ ID NO: 30); AVGLLAPP (SEQ ID NO: 31); LLAPSHRA (SEQ ID NO
  • the CM comprises the amino acid sequence ISSGLLSS (SEQ ID NO: 14). In some embodiments, the CM comprises the amino acid sequence QNQALRMA (SEQ ID NO: 15). In some embodiments, the CM comprises the amino acid sequence AQNLLGMV (SEQ ID NO: 16). In some embodiments, the CM comprises the amino acid sequence STFPFGMF (SEQ ID NO: 17). In some embodiments, the CM comprises the amino acid sequence PVGYTSSL (SEQ ID NO: 18). In some embodiments, the CM comprises the amino acid sequence DWLYWPGI (SEQ ID NO: 19). In some embodiments, the CM comprises the amino acid sequence MIAPVAYR (SEQ ID NO: 20).
  • the CM comprises the amino acid sequence RPSPMWAY (SEQ ID NO: 21). In some embodiments, the CM comprises the amino acid sequence WATPRPMR (SEQ ID NO: 22). In some embodiments, the CM comprises the amino acid sequence FRLLDWQW (SEQ ID NO: 23). In some embodiments, the CM comprises the amino acid sequence LKAAPRWA (SEQ ID NO: 24). In some embodiments, the CM comprises the amino acid sequence GPSHLVLT (SEQ ID NO: 25). In some embodiments, the CM comprises the amino acid sequence LPGGLSPW (SEQ ID NO: 26). In some embodiments, the CM comprises the amino acid sequence MGLFSEAG (SEQ ID NO: 27).
  • the CM comprises the amino acid sequence SPLPLRVP (SEQ ID NO: 28). In some embodiments, the CM comprises the amino acid sequence RMHLRSLG (SEQ ID NO: 29). In some embodiments, the CM comprises the amino acid sequence LAAPLGLL (SEQ ID NO: 30). In some embodiments, the CM comprises the amino acid sequence AVGLLAPP (SEQ ID NO: 31). In some embodiments, the CM comprises the amino acid sequence LLAPSHRA (SEQ ID NO: 32). In some embodiments, the CM comprises the amino acid sequence PAGLWLDP (SEQ ID NO: 33). In some embodiments, the CM comprises the amino acid sequence ISSGLSS (SEQ ID NO: 159).
  • the CM is a substrate for at least two proteases.
  • at least one protease is an MMP and at least one protease is selected from the group consisting of those shown in Table 7.
  • the activatable antibody includes at least a first CM and a second CM.
  • the first CM and the second CM are each polypeptides of no more than 15 amino acids long.
  • the first CM and the second CM in the activatable antibody have the structural arrangement from N-terminus to C-terminus as follows in the uncleaved state: MM-CM 1-CM2-AB, AB-CM2-CM 1-MM, MM-CM2-CM1-AB, or AB-CM1-CM2-MM.
  • the activatable antibody includes a linking peptide between MM and CM1.
  • the activatable antibody includes a linking peptide between CM1 and CM2.
  • the activatable antibody includes a linking peptide between CM2 and AB. In some embodiments, the activatable antibody includes a linking peptide between MM and CM1 and a linking peptide between CM2 and AB. In some embodiments, the activatable antibody includes a linking peptide between MM and CM1 and a linking peptide between CM1 and CM2. In some embodiments, the activatable antibody includes a linking peptide between CM1 and CM2 and a linking peptide between CM2 and AB. In some embodiments, the activatable antibody includes a linking peptide between MM and CM1, a linking peptide between CM1 and CM2, and a linking peptide between CM2 and AB.
  • the activatable antibody includes at least a first CM that includes a substrate for at least one matrix metalloprotease (MMP) and a second CM that includes a substrate sequence.
  • CM2 matrix metalloprotease
  • Exemplary substrates for the second CM (CM2) include but are not limited to substrates cleavable by one or more of the following enzymes or proteases listed in Table 7.
  • the CM2 is selected for use with a specific protease.
  • the CM2 is a substrate for at least one protease selected from the group consisting of a matrix metalloprotease (MMP), a neutrophil elastase, u-type plasminogen activator (uPA, also referred to as urokinase), legumain, matriptase (MT-SP1), thrombin, a cysteine protease such as a cathepsin, ADAM17, BMP-1, HtrA1, and a TMPRSS such as TMPRSS3 or TMPRSS4.
  • MMP matrix metalloprotease
  • uPA u-type plasminogen activator
  • MT-SP1 matriptase
  • thrombin a cysteine protease
  • TMPRSS such as TMPRSS3 or TMPRSS4.
  • the CM2 is a substrate for a neutrophil elastase. In some embodiments, the CM2 is a substrate for uPA. In some embodiments, the CM2 is a substrate for legumain. In some embodiments, the CM2 is a substrate for matriptase. In some embodiments, the CM2 is a substrate for thrombin. In some embodiments, the CM2 is a substrate for a cysteine protease. In some embodiments, the CM2 is a substrate for a cathepsin. In some embodiments, the CM2 is a substrate for ADAM17. In some embodiments, the CM2 is a substrate for BMP-1.
  • the CM2 is a substrate for HtrA1. In some embodiments, the CM2 is a substrate for a TMPRSS. In some embodiments, the CM2 is a substrate for TMPRSS3. In some embodiments, the CM2 is a substrate for TMPRSS4.
  • suitable CM2 are cleaved by at least one protease and include the sequence TGRGPSWV (SEQ ID NO: 34); SARGPSRW (SEQ ID NO: 35); TARGPSFK (SEQ ID NO: 36); LSGRSDNH (SEQ ID NO: 37); GGWHTGRN (SEQ ID NO: 38); HTGRSGAL (SEQ ID NO: 39); PLTGRSGG (SEQ ID NO: 40); AARGPAIH (SEQ ID NO: 41); RGPAFNPM (SEQ ID NO: 42); SSRGPAYL (SEQ ID NO: 43); RGPATPIM (SEQ ID NO: 44); RGPA (SEQ ID NO: 45); GGQPSGMWGW (SEQ ID NO: 46); FPRPLGITGL (SEQ ID NO: 47); VHMPLGFLGP (SEQ ID NO: 48); SPLTGRSG (SEQ ID NO: 49); SAGFSLPA (SEQ ID NO: 126); LAPLGLQRR (SEQ ID NO:
  • the CM2 comprises the amino acid sequence TGRGPSWV (SEQ ID NO: 34). In some embodiments, the CM2 comprises the amino acid sequence SARGPSRW (SEQ ID NO: 35). In some embodiments, the CM2 comprises the amino acid sequence TARGPSFK (SEQ ID NO: 36). In some embodiments, the CM2 comprises the amino acid sequence LSGRSDNH (SEQ ID NO: 37). In some embodiments, the CM2 comprises the amino acid sequence GGWHTGRN (SEQ ID NO: 38). In some embodiments, the CM2 comprises the amino acid sequence HTGRSGAL (SEQ ID NO: 39). In some embodiments, the CM2 comprises the amino acid sequence PLTGRSGG (SEQ ID NO: 40).
  • the CM2 comprises the amino acid sequence AARGPAIH (SEQ ID NO: 41). In some embodiments, the CM2 comprises the amino acid sequence RGPAFNPM (SEQ ID NO: 42). In some embodiments, the CM2 comprises the amino acid sequence SSRGPAYL (SEQ ID NO: 43). In some embodiments, the CM2 comprises the amino acid sequence RGPATPIM (SEQ ID NO: 44). In some embodiments, the CM2 comprises the amino acid sequence RGPA (SEQ ID NO: 45). In some embodiments, the CM2 comprises the amino acid sequence GGQPSGMWGW (SEQ ID NO: 46). In some embodiments, the CM2 comprises the amino acid sequence FPRPLGITGL (SEQ ID NO: 47).
  • the CM2 comprises the amino acid sequence VHMPLGFLGP (SEQ ID NO: 48). In some embodiments, the CM2 comprises the amino acid sequence SPLTGRSG (SEQ ID NO: 49). In some embodiments, the CM2 comprises the amino acid sequence LAPLGLQRR (SEQ ID NO: 50). In some embodiments, the CM2 comprises the amino acid sequence SGGPLGVR (SEQ ID NO: 51). In some embodiments, the CM2 comprises the amino acid sequence PLGL (SEQ ID NO: 52). In some embodiments, the CM2 comprises the amino acid sequence GPRSFGL (SEQ ID NO: 315). In some embodiments, the CM2 comprises the amino acid sequence GPRSFG (SEQ ID NO: 316)
  • the CM2 is a substrate for at least one MMP. In some embodiments, the CM2 is a substrate for at least one MMP listed in the Table 7. In some embodiments, the CM2 is a substrate for MMP9. In some embodiments, the CM2 is a substrate for MMP14. In some embodiments, CM1 is substrate for a first MMP, and CM2 is a substrate for a second MMP, where the first MMP and the second MMP are different MMPs. In some embodiments, CM1 is a first substrate sequence for a MMP, and CM2 is a second substrate for the same MMP, where the CM1 and CM2 have different substrate sequences.
  • the CM2 is a substrate for two or more MMPs. In some embodiments, the CM2 is a substrate for at least MMP9 or MMP14. In some embodiments, the CM2 is a substrate for two or more MMPs. In some embodiments, the CM2 is a substrate for at least MMP9 and MMP14. In some embodiments, CM1 and CM2 are both substrates for MMP9. In some embodiments, CM1 and CM2 are both substrates for MMP14. In some embodiments, CM1 is a substrate for MMP9 and CM2 is a substrate for MMP14. In some embodiments, CM1 is a substrate for MMP14 and CM2 is a substrate for MMP9.
  • CM1 and/or CM2 is a substrate for an MMP and includes the sequence ISSGLLSS (SEQ ID NO: 14); QNQALRMA (SEQ ID NO: 15); AQNLLGMV (SEQ ID NO: 16); STFPFGMF (SEQ ID NO: 17); PVGYTSSL (SEQ ID NO: 18); DWLYWPGI (SEQ ID NO: 19); MIAPVAYR (SEQ ID NO: 20); RPSPMWAY (SEQ ID NO: 21); WATPRPMR (SEQ ID NO: 22); FRLLDWQW (SEQ ID NO: 23); LKAAPRWA (SEQ ID NO: 24); GPSHLVLT (SEQ ID NO: 25); LPGGLSPW (SEQ ID NO: 26); MGLFSEAG (SEQ ID NO: 27); SPLPLRVP (SEQ ID NO: 28); RMHLRSLG (SEQ ID NO: 29); LAAPLGLL (SEQ ID NO: 30); AVGLLAPP (SEQ ID NO: 14); QNQALR
  • the first cleaving agent and the second cleaving agent are the same matrix metalloprotease, and the first CM and the second CM are different substrates for the enzyme.
  • the first cleaving agent and the second cleaving agent are different proteases, where at least one protease is an MMP.
  • the first cleaving agent and the second cleaving agent are co-localized in the target tissue.
  • the first CM and the second CM are cleaved by at least one cleaving agent in the target tissue.
  • the activatable antibody is exposed to and cleaved by a MMP such that, in the activated or cleaved state, the activated antibody includes a light chain amino acid sequence that includes at least a portion of LP2 and/or CM sequence after the MMP has cleaved the CM.
  • the CM comprises the non-prime side of the protease cleavage site; that is, the CM comprises at least the P1 and P2 amino acids, and in some embodiments, comprises the P1, P2 and P3 amino acids and in some embodiments, comprises the P1, P2, P3, and P4 amino acids.
  • the CM comprises the non-prime side and the prime side of the protease cleavage site.
  • the CM comprises the non-prime side but lacks at least part of the prime side of the protease cleavage site.
  • the CM comprises the non-prime side but lacks the prime side of the protease cleavage site.
  • Such a CM can be linked directly or through a linker to an antibody or other molecule as disclosed herein, such as, but not limited to, a detection moiety.
  • the activatable antibody is an anti-EGFR activatable antibody that includes at least an AB that is or is derived from cetuximab or panitumumab; a MM comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 160, 167-200, and 497; and a CM comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 14-33, and 159.
  • the activatable antibody is an anti-EGFR activatable antibody that includes at least an AB that is or is derived from cetuximab or panitumumab; a MM comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 160, 167-200, and 497; and a CM comprising an amino acid sequence selected from the group consisting of the sequences presented in Tables 8A-8M.
  • the anti-EGFR activatable antibody also includes a first linking peptide (LP1) and a second linking peptide (LP2), and the activatable antibody in the uncleaved state has the structural arrangement from N-terminus to C-terminus as follows: MM-LP1-CM-LP2-AB or AB-LP2-CM-LP1-MM.
  • each of LP1 and LP2 is a peptide of about 1 to 20 amino acids in length.
  • the two linking peptides need not be identical to each other.
  • At least one of LP1 or LP2 comprises an amino acid sequence selected from the group consisting of (GS) n , (GGS) n , (GSGGS) n (SEQ ID NO: 1) and (GGGS) n (SEQ ID NO: 2), where n is an integer of at least one.
  • at least one of LP1 or LP2 comprises an amino acid sequence selected from the group consisting of GGSG (SEQ ID NO: 3), GGSGG (SEQ ID NO: 4), GSGSG (SEQ ID NO: 5), GSGGG (SEQ ID NO: 6), GGGSG (SEQ ID NO: 7), and GSSSG (SEQ ID NO: 8).
  • LP1 comprises the amino acid sequence GSSGGSGGSGGSG (SEQ ID NO: 9), GSSGGSGGSGG (SEQ ID NO: 10), GSSGGSGGSGGS (SEQ ID NO: 11), GSSGGSGGSGGSGGGS (SEQ ID NO: 155), GSSGGSGGSG (SEQ ID NO: 156), or GSSGGSGGSGS (SEQ ID NO: 157).
  • LP2 comprises the amino acid sequence GSS, GGS, GGGS (SEQ ID NO: 158), GSSGT (SEQ ID NO: 12) or GSSG (SEQ ID NO: 13).
  • the activatable antibody is an anti-EGFR activatable antibody that includes at least an AB comprising a heavy chain amino acid sequence comprising the VH CDR1 sequence of SEQ ID NO: 504, the VH CDR2 sequence of SEQ ID NO: 505, the VH CDR3 sequence of SEQ ID NO: 506, the VL CDR1 sequence of SEQ ID NO: 507, the VL CDR2 sequence of SEQ ID NO: 508, and the VL CDR2 sequence of SEQ ID NO: 509; a MM comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 160, 167-200, and 497; and a CM comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 14-33, and 159.
  • the activatable antibody is an anti-EGFR activatable antibody that includes at least an AB comprising a heavy chain amino acid sequence comprising the VH CDR1 sequence of SEQ ID NO: 504, the VH CDR2 sequence of SEQ ID NO: 505, the VH CDR3 sequence of SEQ ID NO: 506, the VL CDR1 sequence of SEQ ID NO: 507, the VL CDR2 sequence of SEQ ID NO: 508, and the VL CDR2 sequence of SEQ ID NO: 509; a MM comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 160, 167-200, and 497; and a CM comprising an amino acid sequence selected from the group consisting of the sequences presented in Tables 8A-8M.
  • the anti-EGFR activatable antibody also includes a first linking peptide (LP1) and a second linking peptide (LP2), and the activatable antibody in the uncleaved state has the structural arrangement from N-terminus to C-terminus as follows: MM-LP1-CM-LP2-AB or AB-LP2-CM-LP1-MM.
  • each of LP1 and LP2 is a peptide of about 1 to 20 amino acids in length.
  • the two linking peptides need not be identical to each other.
  • At least one of LP1 or LP2 comprises an amino acid sequence selected from the group consisting of (GS) n , (GGS) n , (GSGGS) n (SEQ ID NO: 1) and (GGGS) n (SEQ ID NO: 2), where n is an integer of at least one.
  • at least one of LP1 or LP2 comprises an amino acid sequence selected from the group consisting of GGSG (SEQ ID NO: 3), GGSGG (SEQ ID NO: 4), GSGSG (SEQ ID NO: 5), GSGGG (SEQ ID NO: 6), GGGSG (SEQ ID NO: 7), and GSSSG (SEQ ID NO: 8).
  • LP1 comprises the amino acid sequence GSSGGSGGSGGSG (SEQ ID NO: 9), GSSGGSGGSGG (SEQ ID NO: 10), GSSGGSGGSGGS (SEQ ID NO: 11), GSSGGSGGSGGSGGGS (SEQ ID NO: 155), GSSGGSGGSG (SEQ ID NO: 156), or GSSGGSGGSGS (SEQ ID NO: 157).
  • LP2 comprises the amino acid sequence GSS, GGS, GGGS (SEQ ID NO: 158), GSSGT (SEQ ID NO: 12) or GSSG (SEQ ID NO: 13).
  • the activatable antibody is an anti-EGFR activatable antibody that includes at least an AB comprising the heavy chain amino acid sequence of SEQ ID NO: 56, 57 or 58 and the light chain amino acid sequence of SEQ ID NO: 59; a MM comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 160, 167-200, and 497; and a CM comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 14-33, and 159.
  • the activatable antibody is an anti-EGFR activatable antibody that includes at least an AB comprising the heavy chain amino acid sequence of SEQ ID NO: 56, 57 or 58 and the light chain amino acid sequence of SEQ ID NO: 59; a MM comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 160, 167-200, and 497; and a CM comprising an amino acid sequence selected from the group consisting of the sequences presented in Tables 8A-8M.
  • the anti-EGFR activatable antibody also includes a first linking peptide (LP1) and a second linking peptide (LP2), and the activatable antibody in the uncleaved state has the structural arrangement from N-terminus to C-terminus as follows: MM-LP1-CM-LP2-AB or AB-LP2-CM-LP 1-MM.
  • each of LP1 and LP2 is a peptide of about 1 to 20 amino acids in length.
  • the two linking peptides need not be identical to each other.
  • At least one of LP1 or LP2 comprises an amino acid sequence selected from the group consisting of (GS) n , (GGS) n , (GSGGS) n (SEQ ID NO: 1) and (GGGS) n (SEQ ID NO: 2), where n is an integer of at least one.
  • at least one of LP1 or LP2 comprises an amino acid sequence selected from the group consisting of GGSG (SEQ ID NO: 3), GGSGG (SEQ ID NO: 4), GSGSG (SEQ ID NO: 5), GSGGG (SEQ ID NO: 6), GGGSG (SEQ ID NO: 7), and GSSSG (SEQ ID NO: 8).
  • LP1 comprises the amino acid sequence GSSGGSGGSGGSG (SEQ ID NO: 9), GSSGGSGGSGG (SEQ ID NO: 10), GSSGGSGGSGGS (SEQ ID NO: 11), GSSGGSGGSGGSGGGS (SEQ ID NO: 155), GSSGGSGGSG (SEQ ID NO: 156), or GSSGGSGGSGS (SEQ ID NO: 157).
  • LP2 comprises the amino acid sequence GSS, GGS, GGGS (SEQ ID NO: 158), GSSGT (SEQ ID NO: 12) or GSSG (SEQ ID NO: 13).
  • the activatable antibody is an anti-EGFR activatable antibody that includes at least an AB comprising the heavy chain amino acid sequence of SEQ ID NO: 56 and the light chain amino acid sequence of SEQ ID NO: 59; a MM comprising the amino acid sequence of SEQ ID NO: 160; and a CM comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 14-33, and 159.
  • the activatable antibody is an anti-EGFR activatable antibody that includes at least an AB comprising the heavy chain amino acid sequence of SEQ ID NO: 56 and the light chain amino acid sequence of SEQ ID NO: 59; a MM comprising the amino acid sequence of SEQ ID NO: 160; and a CM comprising an amino acid sequence selected from the group consisting of the sequences presented in Tables 8A-8M.
  • the anti-EGFR activatable antibody also includes a first linking peptide (LP1) and a second linking peptide (LP2), and the activatable antibody in the uncleaved state has the structural arrangement from N-terminus to C-terminus as follows: MM-LP1-CM-LP2-AB or AB-LP2-CM-LP1-MM.
  • each of LP1 and LP2 is a peptide of about 1 to 20 amino acids in length.
  • the two linking peptides need not be identical to each other.
  • At least one of LP1 or LP2 comprises an amino acid sequence selected from the group consisting of (GS) n , (GGS) n , (GSGGS) n (SEQ ID NO: 1) and (GGGS) n (SEQ ID NO: 2), where n is an integer of at least one.
  • at least one of LP1 or LP2 comprises an amino acid sequence selected from the group consisting of GGSG (SEQ ID NO: 3), GGSGG (SEQ ID NO: 4), GSGSG (SEQ ID NO: 5), GSGGG (SEQ ID NO: 6), GGGSG (SEQ ID NO: 7), and GSSSG (SEQ ID NO: 8).
  • LP1 comprises the amino acid sequence GSSGGSGGSGGSG (SEQ ID NO: 9), GSSGGSGGSGG (SEQ ID NO: 10), GSSGGSGGSGGS (SEQ ID NO: 11), GSSGGSGGSGGSGGGS (SEQ ID NO: 155), GSSGGSGGSG (SEQ ID NO: 156), or GSSGGSGGSGS (SEQ ID NO: 157).
  • LP2 comprises the amino acid sequence GSS, GGS, GGGS (SEQ ID NO: 158), GSSGT (SEQ ID NO: 12) or GSSG (SEQ ID NO: 13).
  • the activatable antibody is an anti-EGFR activatable antibody that includes at least an AB comprising a heavy chain amino acid sequence comprising the VH CDR1 sequence of SEQ ID NO: 504, the VH CDR2 sequence of SEQ ID NO: 505, the VH CDR3 sequence of SEQ ID NO: 506, the VL CDR1 sequence of SEQ ID NO: 507, the VL CDR2 sequence of SEQ ID NO: 508, and the VL CDR2 sequence of SEQ ID NO: 509; a MM comprising the amino acid sequence of SEQ ID NO: 160; and a CM comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 14-33, and 159.
  • the activatable antibody is an anti-EGFR activatable antibody that includes at least an AB comprising a heavy chain amino acid sequence comprising the VH CDR1 sequence of SEQ ID NO: 504, the VH CDR2 sequence of SEQ ID NO: 505, the VH CDR3 sequence of SEQ ID NO: 506, the VL CDR1 sequence of SEQ ID NO: 507, the VL CDR2 sequence of SEQ ID NO: 508, and the VL CDR2 sequence of SEQ ID NO: 509; a MM comprising the amino acid sequence of SEQ ID NO: 160; and a CM comprising an amino acid sequence selected from the group consisting of the sequences presented in Tables 8A-8M.
  • the anti-EGFR activatable antibody also includes a first linking peptide (LP1) and a second linking peptide (LP2), and the activatable antibody in the uncleaved state has the structural arrangement from N-terminus to C-terminus as follows: MM-LP1-CM-LP2-AB or AB-LP2-CM-LP 1-MM.
  • each of LP1 and LP2 is a peptide of about 1 to 20 amino acids in length.
  • the two linking peptides need not be identical to each other.
  • At least one of LP1 or LP2 comprises an amino acid sequence selected from the group consisting of (GS) n , (GGS) n , (GSGGS) n (SEQ ID NO: 1) and (GGGS) n (SEQ ID NO: 2), where n is an integer of at least one.
  • at least one of LP1 or LP2 comprises an amino acid sequence selected from the group consisting of GGSG (SEQ ID NO: 3), GGSGG (SEQ ID NO: 4), GSGSG (SEQ ID NO: 5), GSGGG (SEQ ID NO: 6), GGGSG (SEQ ID NO: 7), and GSSSG (SEQ ID NO: 8).
  • LP1 comprises the amino acid sequence GSSGGSGGSGGSG (SEQ ID NO: 9), GSSGGSGGSGG (SEQ ID NO: 10), GSSGGSGGSGGS (SEQ ID NO: 11), GSSGGSGGSGGSGGGS (SEQ ID NO: 155), GSSGGSGGSG (SEQ ID NO: 156), or GSSGGSGGSGS (SEQ ID NO: 157).
  • LP2 comprises the amino acid sequence GSS, GGS, GGGS (SEQ ID NO: 158), GSSGT (SEQ ID NO: 12) or GSSG (SEQ ID NO: 13).
  • the activatable antibody is an anti-Jagged activatable antibody that includes at least an AB comprising the heavy chain amino acid sequence of SEQ ID NO: 61, 63, 65, 68, 70, 72, 76, 78, 80, 82, 84, 86, 88, 90, 92, 94, 96, 98, 100, 102, 104, 106, 108, 110, 112, or 114 and the light chain amino acid sequence of SEQ ID NO: 60, 62, 64, 67, 69, 71, 73, 75, 77, 79, 81, 83, 85, 87, 89, 91, 93, 95, 97, 99, 101, 103, 105, 107, 109, 111, or 113; a MM comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 201-263, and 496; and a CM comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 14
  • the activatable antibody is an anti-Jagged activatable antibody that includes at least an AB comprising the heavy chain amino acid sequence of SEQ ID NO: 61, 63, 65, 68, 70, 72, 76, 78, 80, 82, 84, 86, 88, 90, 92, 94, 96, 98, 100, 102, 104, 106, 108, 110, 112, or 114 and the light chain amino acid sequence of SEQ ID NO: 60, 62, 64, 67, 69, 71, 73, 75, 77, 79, 81, 83, 85, 87, 89, 91, 93, 95, 97, 99, 101, 103, 105, 107, 109, 111, or 113; a MM comprising an amino acid sequence selected from the group consisting of SEQ ID NOs: 201-263, and 496; and a CM comprising an amino acid sequence selected from the group consisting of the sequences presented in Table
  • the anti-Jagged activatable antibody also includes a first linking peptide (LP1) and a second linking peptide (LP2), and the activatable antibody in the uncleaved state has the structural arrangement from N-terminus to C-terminus as follows: MM-LP1-CM-LP2-AB or AB-LP2-CM-LP1-MM.
  • each of LP1 and LP2 is a peptide of about 1 to 20 amino acids in length.
  • the two linking peptides need not be identical to each other.
  • At least one of LP1 or LP2 comprises an amino acid sequence selected from the group consisting of (GS) n , (GGS) n , (GSGGS) n (SEQ ID NO: 1) and (GGGS) n (SEQ ID NO: 2), where n is an integer of at least one.
  • at least one of LP1 or LP2 comprises an amino acid sequence selected from the group consisting of GGSG (SEQ ID NO: 3), GGSGG (SEQ ID NO: 4), GSGSG (SEQ ID NO: 5), GSGGG (SEQ ID NO: 6), GGGSG (SEQ ID NO: 7), and GSSSG (SEQ ID NO: 8).
  • LP1 comprises the amino acid sequence GSSGGSGGSGGSG (SEQ ID NO: 9), GSSGGSGGSGG (SEQ ID NO: 10), GSSGGSGGSGGS (SEQ ID NO: 11), GSSGGSGGSGGSGGGS (SEQ ID NO: 155), GSSGGSGGSG (SEQ ID NO: 156), or GSSGGSGGSGS (SEQ ID NO: 157).
  • LP2 comprises the amino acid sequence GSS, GGS, GGGS (SEQ ID NO: 158), GSSGT (SEQ ID NO: 12) or GSSG (SEQ ID NO: 13).
  • the activatable antibody is an anti-Jagged activatable antibody that includes at least an AB comprising the heavy chain amino acid sequence of SEQ ID NO: 112 and the light chain amino acid sequence of SEQ ID NO: 111; a MM comprising the amino acid sequence selected of SEQ ID NO: 217; and a CM comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 14-33, and 159.
  • the activatable antibody is an anti-Jagged activatable antibody that includes at least an AB comprising the heavy chain amino acid sequence of SEQ ID NO: 112 and the light chain amino acid sequence of SEQ ID NO: 111; a MM comprising the amino acid sequence selected of SEQ ID NO: 217; and a CM comprising an amino acid sequence selected from the group consisting of the sequences presented in Tables 8A-8M.
  • the anti-Jagged activatable antibody also includes a first linking peptide (LP1) and a second linking peptide (LP2), and the activatable antibody in the uncleaved state has the structural arrangement from N-terminus to C-terminus as follows: MM-LP1-CM-LP2-AB or AB-LP2-CM-LP 1-MM.
  • each of LP1 and LP2 is a peptide of about 1 to 20 amino acids in length.
  • the two linking peptides need not be identical to each other.
  • At least one of LP1 or LP2 comprises an amino acid sequence selected from the group consisting of (GS) n , (GGS) n , (GSGGS) n (SEQ ID NO: 1) and (GGGS) n (SEQ ID NO: 2), where n is an integer of at least one.
  • at least one of LP1 or LP2 comprises an amino acid sequence selected from the group consisting of GGSG (SEQ ID NO: 3), GGSGG (SEQ ID NO: 4), GSGSG (SEQ ID NO: 5), GSGGG (SEQ ID NO: 6), GGGSG (SEQ ID NO: 7), and GSSSG (SEQ ID NO: 8).
  • LP1 comprises the amino acid sequence GSSGGSGGSGGSG (SEQ ID NO: 9), GSSGGSGGSGG (SEQ ID NO: 10), GSSGGSGGSGGS (SEQ ID NO: 11), GSSGGSGGSGGSGGGS (SEQ ID NO: 155), GSSGGSGGSG (SEQ ID NO: 156), or GSSGGSGGSGS (SEQ ID NO: 157).
  • LP2 comprises the amino acid sequence GSS, GGS, GGGS (SEQ ID NO: 158), GSSGT (SEQ ID NO: 12) or GSSG (SEQ ID NO: 13).
  • the activatable antibody is an anti-Jagged activatable antibody that includes at least an AB comprising a heavy chain amino acid sequence comprising the VH CDR1 sequence of SEQ ID NO: 498, the VH CDR2 sequence of SEQ ID NO: 499, the VH CDR3 sequence of SEQ ID NO: 500, the VL CDR1 sequence of SEQ ID NO: 501, the VL CDR2 sequence of SEQ ID NO: 502, and the VL CDR2 sequence of SEQ ID NO: 503; a MM comprising the amino acid sequence selected from the group consisting of SEQ ID NO: 217; and a CM comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 14-33, and 159.
  • the activatable antibody is an anti-Jagged activatable antibody that includes at least an AB comprising a heavy chain amino acid sequence comprising the VH CDR1 sequence of SEQ ID NO: 498, the VH CDR2 sequence of SEQ ID NO: 499, the VH CDR3 sequence of SEQ ID NO: 500, the VL CDR1 sequence of SEQ ID NO: 501, the VL CDR2 sequence of SEQ ID NO: 502, and the VL CDR2 sequence of SEQ ID NO: 503; a MM comprising the amino acid sequence selected from the group consisting of SEQ ID NO: 217; and a CM comprising an amino acid sequence selected from the group consisting of the sequences presented in Tables 8A-8M.
  • the anti-Jagged activatable antibody also includes a first linking peptide (LP1) and a second linking peptide (LP2), and the activatable antibody in the uncleaved state has the structural arrangement from N-terminus to C-terminus as follows: MM-LP1-CM-LP2-AB or AB-LP2-CM-LP1-MM.
  • each of LP1 and LP2 is a peptide of about 1 to 20 amino acids in length.
  • the two linking peptides need not be identical to each other.
  • At least one of LP1 or LP2 comprises an amino acid sequence selected from the group consisting of (GS) n , (GGS) n , (GSGGS) n (SEQ ID NO: 1) and (GGGS) n (SEQ ID NO: 2), where n is an integer of at least one.
  • at least one of LP1 or LP2 comprises an amino acid sequence selected from the group consisting of GGSG (SEQ ID NO: 3), GGSGG (SEQ ID NO: 4), GSGSG (SEQ ID NO: 5), GSGGG (SEQ ID NO: 6), GGGSG (SEQ ID NO: 7), and GSSSG (SEQ ID NO: 8).
  • LP1 comprises the amino acid sequence GSSGGSGGSGGSG (SEQ ID NO: 9), GSSGGSGGSGG (SEQ ID NO: 10), GSSGGSGGSGGS (SEQ ID NO: 11), GSSGGSGGSGGSGGGS (SEQ ID NO: 155), GSSGGSGGSG (SEQ ID NO: 156), or GSSGGSGGSGS (SEQ ID NO: 157).
  • LP2 comprises the amino acid sequence GSS, GGS, GGGS (SEQ ID NO: 158), GSSGT (SEQ ID NO: 12) or GSSG (SEQ ID NO: 13).
  • the activatable antibody also includes an agent conjugated to the AB.
  • the agent is a therapeutic agent.
  • the agent is an antineoplastic agent.
  • the agent is a toxin or a fragment thereof.
  • the agent is conjugated to the AB via a linker.
  • the linker is a cleavable linker.
  • the agent is a microtubule inhibitor.
  • the agent is a nucleic acid damaging agent, such as a DNA alkylator or DNA intercalator, or other DNA damaging agent.
  • the linker is a cleavable linker.
  • the agent is conjugated to the AB via a linker that includes at least one MMP-cleavable substrate sequence.
  • the agent is an agent selected from the group listed in Table 3.
  • the agent is a dolastatin.
  • the agent is an auristatin or derivative thereof.
  • the agent is auristatin E or a derivative thereof.
  • the agent is monomethyl auristatin E (MMAE).
  • the agent is monomethyl auristatin D (MMAD).
  • the agent is a maytansinoid or maytansinoid derivative.
  • the agent is DM1 or DM4.
  • the agent is a duocarmycin or derivative thereof.
  • the agent is a calicheamicin or derivative thereof.
  • the agent is a pyrrolobenzodiazepine.
  • the agent is an anti-inflammatory agent.
  • the activatable antibody also includes a detectable moiety.
  • the detectable moiety is a diagnostic agent.
  • the conjugated antibody includes a detectable label.
  • the detectable label includes an imaging agent, a contrasting agent, an enzyme, a fluorescent label, a chromophore, a dye, one or more metal ions, or a ligand-based label.
  • the imaging agent comprises a radioisotope.
  • the radioisotope is indium or technetium.
  • the contrasting agent comprises iodine, gadolinium or iron oxide.
  • the enzyme comprises horseradish peroxidase, alkaline phosphatase, or ⁇ -galactosidase.
  • the fluorescent label comprises yellow fluorescent protein (YFP), cyan fluorescent protein (CFP), green fluorescent protein (GFP), modified red fluorescent protein (mRFP), red fluorescent protein tdimer2 (RFP tdimer2), HCRED, or a europium derivative.
  • the luminescent label comprises an N-methylacrydium derivative.
  • the label comprises an Alexa Fluor® label, such as Alex Fluor® 680 or Alexa Fluor® 750.
  • the ligand-based label comprises biotin, avidin, streptavidin or one or more haptens.
  • the activatable antibody also includes a signal peptide.
  • the signal peptide is conjugated to the activatable antibody via a spacer.
  • the spacer is conjugated to the activatable antibody in the absence of a signal peptide.
  • the spacer is joined directly to the MM of the activatable antibody.
  • the spacer is joined directly to the MM of the activatable antibody in the structural arrangement from N-terminus to C-terminus of spacer-MM-CM-AB.
  • An example of a spacer joined directly to the N-terminus of MM of the activatable antibody is QGQSGQ (SEQ ID NO: 53).
  • the spacer includes at least the amino acid sequence QGQSGQ (SEQ ID NO: 53).
  • the AB of the activatable antibody naturally contains one or more disulfide bonds.
  • the AB can be engineered to include one or more disulfide bonds.
  • the serum half-life of the activatable antibody is longer than that of the corresponding antibody; e.g., the pK of the activatable antibody is longer than that of the corresponding antibody. In some embodiments, the serum half-life of the activatable antibody is similar to that of the corresponding antibody. In some embodiments, the serum half-life of the activatable antibody is at least 15 days when administered to an organism. In some embodiments, the serum half-life of the activatable antibody is at least 12 days when administered to an organism. In some embodiments, the serum half-life of the activatable antibody is at least 11 days when administered to an organism. In some embodiments, the serum half-life of the activatable antibody is at least 10 days when administered to an organism.
  • the serum half-life of the activatable antibody is at least 9 days when administered to an organism. In some embodiments, the serum half-life of the activatable antibody is at least 8 days when administered to an organism. In some embodiments, the serum half-life of the activatable antibody is at least 7 days when administered to an organism. In some embodiments, the serum half-life of the activatable antibody is at least 6 days when administered to an organism. In some embodiments, the serum half-life of the activatable antibody is at least 5 days when administered to an organism. In some embodiments, the serum half-life of the activatable antibody is at least 4 days when administered to an organism. In some embodiments, the serum half-life of the activatable antibody is at least 3 days when administered to an organism.
  • the serum half-life of the activatable antibody is at least 2 days when administered to an organism. In some embodiments, the serum half-life of the activatable antibody is at least 24 hours when administered to an organism. In some embodiments, the serum half-life of the activatable antibody is at least 20 hours when administered to an organism. In some embodiments, the serum half-life of the activatable antibody is at least 18 hours when administered to an organism. In some embodiments, the serum half-life of the activatable antibody is at least 16 hours when administered to an organism. In some embodiments, the serum half-life of the activatable antibody is at least 14 hours when administered to an organism. In some embodiments, the serum half-life of the activatable antibody is at least 12 hours when administered to an organism.
  • the serum half-life of the activatable antibody is at least 10 hours when administered to an organism. In some embodiments, the serum half-life of the activatable antibody is at least 8 hours when administered to an organism. In some embodiments, the serum half-life of the activatable antibody is at least 6 hours when administered to an organism. In some embodiments, the serum half-life of the activatable antibody is at least 4 hours when administered to an organism. In some embodiments, the serum half-life of the activatable antibody is at least 3 hours when administered to an organism.
  • the activatable antibody and/or conjugated activatable antibody is monospecific. In some embodiments, the activatable antibody and/or conjugated activatable antibody is multispecific, e.g., by way of non-limiting example, bispecific or trifunctional. In some embodiments, the activatable antibody and/or conjugated activatable antibody is formulated as part of a pro-Bispecific T Cell Engager (pro-BITE) molecule. In some embodiments, the activatable antibody and/or conjugated activatable antibody is formulated as part of a pro-Chimeric Antigen Receptor (pro-CAR) modified T cell or other engineered receptor.
  • pro-BITE pro-Bispecific T Cell Engager
  • pro-CAR pro-Chimeric Antigen Receptor
  • compositions and methods that include an activatable antibody that includes an antibody or antibody fragment (AB) that specifically binds a given target, where the AB is coupled to a masking moiety (MM) that decreases the ability of the AB to bind its target.
  • the activatable antibody further includes a cleavable moiety (CM) that is a substrate for at least one MMP.
  • CM cleavable moiety
  • compositions and methods provided herein enable the attachment of one or more agents to one or more cysteine residues in the AB without reducing or otherwise disturbing one or more disulfide bonds within the MM.
  • the compositions and methods provided herein produce an activatable antibody that is conjugated to one or more agents, e.g., any of a variety of therapeutic, diagnostic and/or prophylactic agents, for example, in some embodiments, without any of the agent(s) being conjugated to the MM of the activatable antibody.
  • the compositions and methods provided herein produce conjugated activatable antibodies in which the MM retains the ability to effectively and efficiently mask the AB of the activatable antibody in an uncleaved state.
  • the compositions and methods provided herein produce conjugated activatable antibodies in which the activatable antibody is still activated, i.e., cleaved, in the presence of a MMP that can cleave the CM.
  • the activatable antibodies have at least one point of conjugation for an agent, but in the methods and compositions provided herein less than all possible points of conjugation are available for conjugation to an agent.
  • the one or more points of conjugation are sulfur atoms involved in disulfide bonds.
  • the one or more points of conjugation are sulfur atoms involved in interchain disulfide bonds.
  • the one or more points of conjugation are sulfur atoms involved in interchain sulfide bonds, but not sulfur atoms involved in intrachain disulfide bonds.
  • the one or more points of conjugation are sulfur atoms of cysteine or other amino acid residues containing a sulfur atom. Such residues may occur naturally in the antibody structure or may be incorporated into the antibody by site-directed mutagenesis, chemical conversion, or mis-incorporation of non-natural amino acids.
  • the method generally includes partially reducing interchain disulfide bonds in the activatable antibody with a reducing agent, such as, for example, TCEP; and conjugating the drug reactive with free thiols to the partially reduced activatable antibody.
  • a reducing agent such as, for example, TCEP
  • conjugating the drug reactive with free thiols to the partially reduced activatable antibody As used herein, the term partial reduction refers to situations where an activatable antibody is contacted with a reducing agent and less than all disulfide bonds, e.g., less than all possible sites of conjugation are reduced.
  • a method of reducing and conjugating an agent, e.g., a drug, to an activatable antibody resulting in selectivity in the placement of the agent is provided.
  • the method generally includes partially reducing the activatable antibody with a reducing agent such that any conjugation sites in the masking moiety or other non-AB portion of the activatable antibody are not reduced, and conjugating the agent to interchain thiols in the AB.
  • the conjugation site(s) are selected so as to allow desired placement of an agent to allow conjugation to occur at a desired site.
  • the reducing agent is, for example, TCEP.
  • the reduction reaction conditions such as, for example, the ratio of reducing agent to activatable antibody, the length of incubation, the temperature during the incubation, the pH of the reducing reaction solution, etc., are determined by identifying the conditions that produce a conjugated activatable antibody in which the MM retains the ability to effectively and efficiently mask the AB of the activatable antibody in an uncleaved state.
  • the ratio of reduction agent to activatable antibody will vary depending on the activatable antibody.
  • the ratio of reducing agent to activatable antibody will be in a range from about 20:1 to 1:1, from about 10:1 to 1:1, from about 9:1 to 1:1, from about 8:1 to 1:1, from about 7:1 to 1:1, from about 6:1 to 1:1, from about 5:1 to 1:1, from about 4:1 to 1:1, from about 3:1 to 1:1, from about 2:1 to 1:1, from about 20:1 to 1:1.5, from about 10:1 to 1:1.5, from about 9:1 to 1:1.5, from about 8:1 to 1:1.5, from about 7:1 to 1:1.5, from about 6:1 to 1:1.5, from about 5:1 to 1:1.5, from about 4:1 to 1:1.5, from about 3:1 to 1:1.5, from about 2:1 to 1:1.5, from about 1.5:1 to 1:1.5, or from about 1:1 to 1:1.5.
  • the ratio is in a range of from about 5:1 to 1:1. In some embodiments, the ratio is in a range of from about 5:1 to 1.5:1. In some embodiments, the ratio is in a range of from about 4:1 to 1:1. In some embodiments, the ratio is in a range from about 4:1 to 1.5:1. In some embodiments, the ratio is in a range from about 8:1 to about 1:1. In some embodiments, the ratio is in a range of from about 2.5:1 to 1:1.
  • a method of reducing interchain disulfide bonds in the AB of an activatable antibody and conjugating an agent, e.g., a thiol-containing agent such as a drug, to the resulting interchain thiols to selectively locate agent(s) on the AB is provided.
  • the method generally includes partially reducing the AB with a reducing agent to form at least two interchain thiols without forming all possible interchain thiols in the activatable antibody; and conjugating the agent to the interchain thiols of the partially reduced AB.
  • the AB of the activatable antibody is partially reduced for about 1 hour at about 37° C. at a desired ratio of reducing agent:activatable antibody.
  • the ratio of reducing agent to activatable antibody will be in a range from about 20:1 to 1:1, from about 10:1 to 1:1, from about 9:1 to 1:1, from about 8:1 to 1:1, from about 7:1 to 1:1, from about 6:1 to 1:1, from about 5:1 to 1:1, from about 4:1 to 1:1, from about 3:1 to 1:1, from about 2:1 to 1:1, from about 20:1 to 1:1.5, from about 10:1 to 1:1.5, from about 9:1 to 1:1.5, from about 8:1 to 1:1.5, from about 7:1 to 1:1.5, from about 6:1 to 1:1.5, from about 5:1 to 1:1.5, from about 4:1 to 1:1.5, from about 3:1 to 1:1.5, from about 2:1 to 1:1.5, from about 1.5:1 to 1:1.5, or from about 1:1 to 1:1.5.
  • the ratio is in a range of from about 5:1 to 1:1. In some embodiments, the ratio is in a range of from about 5:1 to 1.5:1. In some embodiments, the ratio is in a range of from about 4:1 to 1:1. In some embodiments, the ratio is in a range from about 4:1 to 1.5:1. In some embodiments, the ratio is in a range from about 8:1 to about 1:1. In some embodiments, the ratio is in a range of from about 2.5:1 to 1:1.
  • the thiol-containing reagent can be, for example, cysteine or N-acetyl cysteine.
  • the reducing agent can be, for example, TCEP.
  • the reduced activatable antibody can be purified prior to conjugation, using for example, column chromatography, dialysis, or diafiltration. In some embodiments, the reduced antibody is not purified after partial reduction and prior to conjugation.
  • the disclosure also provides partially reduced activatable antibodies in which at least one interchain disulfide bond in the activatable antibody has been reduced with a reducing agent without disturbing any intrachain disulfide bonds in the activatable antibody, wherein the activatable antibody includes an antibody or an antigen binding fragment thereof (AB) that specifically binds to the target, a masking moiety (MM) that inhibits the binding of the AB of the activatable antibody in an uncleaved state to the target, and a cleavable moiety (CM) coupled to the AB, wherein the CM is a polypeptide that functions as a substrate for at least one MMP.
  • the MM is coupled to the AB via the CM.
  • one or more intrachain disulfide bond(s) of the activatable antibody is not disturbed by the reducing agent. In some embodiments, one or more intrachain disulfide bond(s) of the MM within the activatable antibody is not disturbed by the reducing agent. In some embodiments, the activatable antibody in the uncleaved state has the structural arrangement from N-terminus to C-terminus as follows: MM-CM-AB or AB-CM-MM. In some embodiments, reducing agent is TCEP.
  • the disclosure also provides partially reduced activatable antibodies, including but not limited to multispecific activatable antibodies of the disclosure, in which at least one interchain disulfide bond in the activatable antibody has been reduced with a reducing agent without disturbing or otherwise compromising the activity and/or efficacy of the activatable antibody, wherein the activatable antibody includes an antibody or an antigen binding fragment thereof (AB) that specifically binds to a target, a masking moiety (MM) that inhibits the binding of the AB of the activatable antibody in an uncleaved state to the target, and a cleavable moiety (CM) coupled to the AB, and the CM is a polypeptide that functions as a substrate for a protease.
  • AB antigen binding fragment thereof
  • MM masking moiety
  • CM cleavable moiety
  • the activity and/or efficacy of the activatable antibody is, by way of nonlimiting example, masking activity, activation of the activatable antibody, and/or binding activity of the activated activatable antibody.
  • one or more intrachain disulfide bond(s) of the activatable antibody is not disturbed by the reducing agent.
  • one or more intrachain disulfide bond(s) of the MM within the activatable antibody is not disturbed by the reducing agent.
  • the activatable antibody in the uncleaved state has the structural arrangement from N-terminus to C-terminus as follows: MM-CM-AB or AB-CM-MM.
  • reducing agent is TCEP.
  • the disclosure also provides conjugated activatable antibodies that include an activatable antibody linked to monomethyl auristatin D (MMAD) payload, wherein the activatable antibody includes an antibody or an antigen binding fragment thereof (AB) that specifically binds to a target, a masking moiety (MM) that inhibits the binding of the AB of the activatable antibody in an uncleaved state to the target, and cleavable moiety (CM) coupled to the AB, and the CM is a polypeptide that functions as a substrate for at least one MMP protease.
  • MMAD monomethyl auristatin D
  • the MMAD-conjugated activatable antibody can be conjugated using any of several methods for attaching agents to ABs: (a) attachment to the carbohydrate moieties of the AB, or (b) attachment to sulfhydryl groups of the AB, or (c) attachment to amino groups of the AB, or (d) attachment to carboxylate groups of the AB.
  • the MMAD payload is conjugated to the AB via a linker. In some embodiments, the MMAD payload is conjugated to a cysteine in the AB via a linker. In some embodiments, the MMAD payload is conjugated to a lysine in the AB via a linker. In some embodiments, the MMAD payload is conjugated to another residue of the AB via a linker, such as those residues disclosed herein. In some embodiments, the linker is a thiol-containing linker. In some embodiments, the linker is a cleavable linker. In some embodiments, the linker is a non-cleavable linker.
  • the linker is selected from the group consisting of the linkers shown in Tables 5 and 6.
  • the activatable antibody and the MMAD payload are linked via a maleimide caproyl-valine-citrulline linker.
  • the activatable antibody and the MMAD payload are linked via a maleimide PEG-valine-citrulline linker.
  • the activatable antibody and the MMAD payload are linked via a maleimide caproyl-valine-citrulline-para-aminobenzyloxycarbonyl linker.
  • the activatable antibody and the MMAD payload are linked via a maleimide PEG-valine-citrulline-para-aminobenzyloxycarbonyl linker.
  • the MMAD payload is conjugated to the AB using the partial reduction and conjugation technology disclosed herein.
  • the target is selected from the group of targets listed in Table 1.
  • the target is EGFR.
  • the target is a Jagged protein, e.g., Jagged 1 and/or Jagged 2.
  • the target is interleukin 6 receptor (IL-6R).
  • the AB is or is derived from an antibody selected from the group of antibodies listed in Table 2.
  • the antigen binding fragment thereof is selected from the group consisting of a Fab fragment, a F(ab′) 2 fragment, a scFv, a scAb, a dAb, a single domain heavy chain antibody, and a single domain light chain antibody.
  • the AB has an equilibrium dissociation constant of about 100 nM or less for binding to the target. In some embodiments, the MM has an equilibrium dissociation constant for binding to the AB that is greater than the equilibrium dissociation constant of the AB to the target. In some embodiments, the MM does not interfere or compete with the AB of the activatable antibody in a cleaved state for binding to the target. In some embodiments, the MM is a polypeptide of no more than 40 amino acids in length. In some embodiments, the MM polypeptide sequence is different from that of the target, and the MM polypeptide sequence is no more than 50% identical to any natural binding partner of the AB.
  • the MM does not include more than 25% amino acid sequence identity to the target. In some embodiments, the MM does not include more than 10% amino acid sequence identity to the target. In some embodiments, the CM is a polypeptide of up to 15 amino acids in length. In some embodiments, the MMP protease is co-localized with the target in a tissue, and the MMP protease cleaves the CM in the activatable antibody when the activatable antibody is exposed to the MMP protease. In some embodiments, the MMP protease is a MMP9 protease. In some embodiments, the MMP protease is a MMP14 protease.
  • the activatable antibody includes a linking peptide between the MM and the CM. In some embodiments, the activatable antibody includes a linking peptide between the CM and the AB. In some embodiments, the activatable antibody includes a first linking peptide (LP1) and a second linking peptide (LP2), and the activatable antibody in an uncleaved state has the structural arrangement from N-terminus to C-terminus as follows: MM-LP1-CM-LP2-AB or AB-LP2-CM-LP 1-MM. In some embodiments, the two linking peptides need not be identical to each other. In some embodiments, each of LP1 and LP2 is a peptide of about 1 to 20 amino acids in length.
  • At least one of LP1 or LP2 includes an amino acid sequence selected from the group consisting of (GS) n , (GGS) n , (GSGGS) n (SEQ ID NO: 1) and (GGGS) n (SEQ ID NO: 2), where n is an integer of at least one.
  • at least one of LP1 or LP2 includes an amino acid sequence selected from the group consisting of GGSG (SEQ ID NO: 3), GGSGG (SEQ ID NO: 4), GSGSG (SEQ ID NO: 5), GSGGG (SEQ ID NO: 6), GGGSG (SEQ ID NO: 7), and GSSSG (SEQ ID NO: 8).
  • the activatable antibody includes a second CM; in some embodiments, the second CM is a substrate for an enzyme selected from the group consisting of those shown in Table 7.
  • the disclosure also provides polypeptides and other larger molecules that include one or more of the MMP-cleavable substrate sequences presented herein.
  • the MMP-cleavable substrate sequences presented herein are useful in prodrug compositions and methods of use thereof.
  • These MMP-cleavable substrate sequences presented herein are also useful in probes and other detection agents and methods of use thereof.
  • the MMP-cleavable substrate sequences presented herein can be used in conjunction with fluors and other quenchers to produce detection agents, such as imaging agents and/or other diagnostic agents.
  • MMP-cleavable substrate sequences presented herein are useful in any composition and/or method in the art that would use a substrate that is cleavable by one or more MMPs, such as MMP9 and/or MMP14.
  • the disclosure also provides an isolated nucleic acid molecule encoding an antibody and/or an activatable antibody described herein, as well as vectors that include these isolated nucleic acid sequences.
  • the disclosure provides methods of producing an antibody and/or activatable antibody by culturing a cell under conditions that lead to expression of the antibody and/or activatable antibody, wherein the cell comprises such a vector.
  • the disclosure provides a method of manufacturing a conjugated antibody of the disclosure that bind a given target by (a) culturing a cell comprising a nucleic acid construct that encodes the antibody under conditions that lead to expression of the antibody, (i) wherein the antibody includes a cleavable moiety (CM), and (ii) wherein the CM is a polypeptide that functions as a substrate for a matrix metalloprotease; (b) recovering the antibody; and (c) conjugating the recovered antibody to one or more additional agents.
  • CM cleavable moiety
  • the disclosure also provides a method of manufacturing the activatable antibodies of the disclosure that bind in an activated state a given target by (a) culturing a cell comprising a nucleic acid construct that encodes the activatable antibody under conditions that lead to expression of the activatable antibody, wherein the activatable antibody comprises a masking moiety (MM), a cleavable moiety (CM), and an antibody or an antigen binding fragment thereof (AB) that specifically binds the target, (i) wherein the CM is a polypeptide that functions as a substrate for a MMP; and (ii) wherein the CM is positioned in the activatable antibody such that, in an uncleaved state, the MM interferes with specific binding of the AB to the target and in a cleaved state the MM does not interfere or compete with specific binding of the AB to the target; and (b) recovering the activatable antibody.
  • MM masking moiety
  • CM cleavable moiety
  • AB
  • the disclosure provides methods of preventing, delaying the progression of, treating, alleviating a symptom of, or otherwise ameliorating a target-related disease in a subject by administering a therapeutically effective amount of a conjugated antibody, an activatable antibody and/or a conjugated activatable antibody described herein to a subject in need thereof.
  • the disclosure provides methods of preventing, delaying the progression of, treating, alleviating a symptom of, or otherwise ameliorating inflammation and/or an inflammatory disorder in a subject by administering a therapeutically effective amount of a conjugated antibody, an activatable antibody and/or a conjugated activatable antibody described herein to a subject in need thereof.
  • the disclosure also provides methods of preventing, delaying the progression of, treating, alleviating a symptom of, or otherwise ameliorating cancer in a subject by administering a therapeutically effective amount of a conjugated antibody, an activatable antibody and/or a conjugated activatable antibody described herein to a subject in need thereof.
  • the disclosure also provides methods of preventing, delaying the progression of, treating, alleviating a symptom of, or otherwise ameliorating an autoimmune disease in a subject by administering a therapeutically effective amount a conjugated antibody, an activatable antibody and/or a conjugated activatable antibody described herein to a subject in need thereof.
  • a conjugated antibody, an activatable antibody and/or a conjugated activatable antibody used in any of the embodiments of these methods and uses can be administered at any stage of the disease.
  • a conjugated antibody, activatable antibody and/or conjugated activatable antibody can be administered to a patient suffering cancer of any stage, from early to metastatic.
  • the terms subject and patient are used interchangeably herein.
  • the subject is a mammal, such as a human, non-human primate, companion animal (e.g., cat, dog, horse), farm animal, work animal, or zoo animal.
  • the subject is a rodent.
  • the subject is a human.
  • the subject is a companion animal.
  • the subject is an animal in the care of a veterinarian.
  • the conjugated antibody, activatable antibody and/or conjugated activatable antibody and therapeutic formulations thereof are administered to a subject suffering from or susceptible to a disease or disorder associated with aberrant target expression and/or activity.
  • a subject suffering from or susceptible to a disease or disorder associated with aberrant target expression and/or activity is identified using any of a variety of methods known in the art.
  • subjects suffering from cancer or other neoplastic condition are identified using any of a variety of clinical and/or laboratory tests such as, physical examination and blood, urine and/or stool analysis to evaluate health status.
  • subjects suffering from inflammation and/or an inflammatory disorder are identified using any of a variety of clinical and/or laboratory tests such as physical examination and/or bodily fluid analysis, e.g., blood, urine and/or stool analysis, to evaluate health status.
  • Administration of a conjugated antibody, an activatable antibody and/or a conjugated activatable antibody to a patient suffering from a disease or disorder associated with aberrant target expression and/or activity is considered successful if any of a variety of laboratory or clinical objectives is achieved.
  • administration of a conjugated antibody, an activatable antibody and/or a conjugated activatable antibody to a patient suffering from a disease or disorder associated with aberrant target expression and/or activity is considered successful if one or more of the symptoms associated with the disease or disorder is alleviated, reduced, inhibited or does not progress to a further, i.e., worse, state.
  • a conjugated antibody, an activatable antibody and/or a conjugated activatable antibody to a patient suffering from a disease or disorder associated with aberrant target expression and/or activity is considered successful if the disease or disorder enters remission or does not progress to a further, i.e., worse, state.
  • the conjugated antibody, activatable antibody and/or conjugated activatable antibody is administered during and/or after treatment in combination with one or more additional agents such as, by way of non-limiting example, an anti-inflammatory agent, an immunosuppressive agent, a chemotherapeutic agent, such as an alkylating agent, an anti-metabolite, an anti-microtubule agent, a topoisomerase inhibitor, a cytotoxic antibiotic, and/or any other nucleic acid damaging agent.
  • the additional agent is a taxane, such as paclitaxel (e.g., Abraxane®).
  • the additional agent is an anti-metabolite, such as gemcitabine.
  • the additional agent is an alkylating agent, such as platinum-based chemotherapy, such as carboplatin or cisplatin.
  • the additional agent is a targeted agent, such as a kinase inhibitor, e.g., sorafenib or erlotinib.
  • the additional agent is a targeted agent, such as another antibody, e.g., a monoclonal antibody (e.g., bevacizumab), a bispecific antibody, or a multispecific antibody.
  • the additional agent is a proteosome inhibitor, such as bortezomib or carfilzomib.
  • the additional agent is an immune modulating agent, such as lenolidominde or IL-2. In some embodiments, the additional agent is radiation. In some embodiments, the additional agent is an agent considered standard of care by those skilled in the art. In some embodiments, the additional agent is a chemotherapeutic agent well known to those skilled in the art.
  • the additional agent is an antibody, another conjugated antibody, another activatable antibody and/or another conjugated activatable antibody.
  • the conjugated antibody, activatable antibody and/or conjugated activatable antibody and the additional agent(s) are administered simultaneously.
  • the conjugated antibody, activatable antibody and/or conjugated activatable antibody and the additional agent(s) can be formulated in a single composition or administered as two or more separate compositions.
  • the conjugated antibody, activatable antibody and/or conjugated activatable antibody and the additional agent(s) are administered sequentially, or the antibody and/or conjugated antibodies and the additional agent are administered at different times during a treatment regimen.
  • the antibody and/or conjugated antibodies is administered prior to the administration of the additional agent, the antibody and/or conjugated antibodies is administered subsequent to the administration of the additional agent, or the antibody and/or conjugated antibodies and the additional agent are administered in an alternating fashion.
  • the antibody and/or conjugated antibodies and additional agent are administered in single doses or in multiple doses.
  • the CM is linked or otherwise attached to an activatable antibody that includes an antibody or antigen-binding fragment thereof that specifically binds a given target coupled to a masking moiety (MM), such that coupling of the MM to the AB reduces the ability of the antibody or antigen-binding fragment thereof to bind the target.
  • the MM is coupled via the CM.
  • Exemplary targets include, but are not limited to the targets shown in Table 1.
  • Exemplary ABs include, but are not limited to, the targets shown in Table 2.
  • the activatable antibodies provided herein are stable in circulation, activated at intended sites of therapy and/or diagnosis but not in normal, e.g., healthy tissue or other tissue not targeted for treatment and/or diagnosis, and, when activated, exhibit binding to the target that is at least comparable to the corresponding, unmodified antibody.
  • the disclosure also provides methods and kits for using the conjugated antibodies, activatable antibodies and/or conjugated activatable antibodies in a variety of diagnostic and/or prophylactic indications.
  • the disclosure provides methods and kits for detecting presence or absence of a cleaving agent and a target of interest in a subject or a sample by (i) contacting a subject or sample with an activatable antibody, wherein the activatable antibody comprises a masking moiety (MM), a cleavable moiety (CM) that is cleaved by the cleaving agent, and an antigen binding domain or fragment thereof (AB) that specifically binds the target of interest, wherein the activatable antibody in an uncleaved, non-activated state comprises a structural arrangement from N-terminus to C-terminus as follows: MM-CM-AB or AB-CM-MM; (a) wherein the MM is a peptide that inhibits binding of the AB to the target, and wherein the MM does not have an amino acid sequence of a naturally occurring binding partner of the AB and is not a modified form of a natural binding partner of the AB; and (b) wherein, in an uncleaved
  • the activatable antibody is an activatable antibody to which a therapeutic agent is conjugated. In some embodiments, the activatable antibody is not conjugated to an agent. In some embodiments, the activatable antibody comprises a detectable label. In some embodiments, the detectable label is positioned on the AB. In some embodiments, measuring the level of activatable antibody in the subject or sample is accomplished using a secondary reagent that specifically binds to the activated antibody, wherein the reagent comprises a detectable label. In some embodiments, the secondary reagent is an antibody comprising a detectable label.
  • the activatable antibody includes a detectable label.
  • the detectable label includes an imaging agent, a contrasting agent, an enzyme, a fluorescent label, a chromophore, a dye, one or more metal ions, or a ligand-based label.
  • the imaging agent comprises a radioisotope.
  • the radioisotope is indium or technetium.
  • the contrasting agent comprises iodine, gadolinium or iron oxide.
  • the enzyme comprises horseradish peroxidase, alkaline phosphatase, or ⁇ -galactosidase.
  • the fluorescent label comprises yellow fluorescent protein (YFP), cyan fluorescent protein (CFP), green fluorescent protein (GFP), modified red fluorescent protein (mRFP), red fluorescent protein tdimer2 (RFP tdimer2), HCRED, or a europium derivative.
  • the luminescent label comprises an N-methylacrydium derivative.
  • the label comprises an Alexa Fluor® label, such as Alex Fluor® 680 or Alexa Fluor® 750.
  • the ligand-based label comprises biotin, avidin, streptavidin or one or more haptens.
  • the subject is a mammal. In some embodiments of these methods, the subject is a human. In some embodiments, the subject is a non-human mammal, such as a non-human primate, companion animal (e.g., cat, dog, horse), farm animal, work animal, or zoo animal. In some embodiments, the subject is a rodent. In some embodiments, the subject is a human. In some embodiments, the subject is a companion animal. In some embodiments, the subject is an animal in the care of a veterinarian.
  • a non-human mammal such as a non-human primate, companion animal (e.g., cat, dog, horse), farm animal, work animal, or zoo animal.
  • the subject is a rodent.
  • the subject is a human.
  • the subject is a companion animal. In some embodiments, the subject is an animal in the care of a veterinarian.
  • the method is an in vivo method. In some embodiments of these methods, the method is an in situ method. In some embodiments of these methods, the method is an ex vivo method. In some embodiments of these methods, the method is an in vitro method.
  • the method is used to identify or otherwise refine a patient population suitable for treatment with an activatable antibody of the disclosure, followed by treatment by administering that activatable antibody and/or conjugated activatable antibody to a subject in need thereof.
  • patients that test positive for both the target and at least one MMP that cleaves the substrate in the cleavable moiety (CM) of the activatable antibody being tested in these methods are identified as suitable candidates for treatment with such an activatable antibody comprising such a CM, and the patient is then administered a therapeutically effective amount of the activatable antibody and/or conjugated activatable antibody that was tested.
  • CM cleavable moiety
  • patients that test negative for either or both of the target and the MMP that cleaves the substrate in the CM in the activatable antibody being tested using these methods might be identified as suitable candidates for another form of therapy.
  • such patients can be tested with other activatable antibodies until a suitable activatable antibody for treatment is identified (e.g., an activatable antibody comprising a CM that is cleaved by the patient at the site of disease).
  • the patient is then administered a therapeutically effective amount of the activatable antibody and/or conjugated for which the patient tested positive.
  • compositions according to the disclosure can include an antibody of the disclosure and a carrier. These pharmaceutical compositions can be included in kits, such as, for example, diagnostic kits.
  • FIGS. 1A and 1B are a series of graphs depicting the ability of the activatable anti-EGFR antibody containing a masking moiety comprising amino acid sequence CISPRGCPDGPYVMY (SEQ ID NO: 160), a cleavage moiety comprising the MMP14 substrate 520 (also referred to herein as MN520) ISSGLLSS (SEQ ID NO: 14), and the heavy and light chains of the anti-EGFR antibody C225v5, where the entire activatable antibody construct is referred to herein as Pb-MN520, to inhibit tumor growth in the H292 xenograft lung cancer model.
  • CISPRGCPDGPYVMY SEQ ID NO: 160
  • MN520 cleavage moiety comprising the MMP14 substrate 520
  • ISSGLLSS SEQ ID NO: 14
  • Pb-MN520 the entire activatable antibody construct
  • FIGS. 2A and 2B are a series of graphs depicting cleavage of the substrate pool referred to herein as SMP87 by 5 nM MMP9.
  • FIGS. 3A and 3B are a series of graphs depicting cleavage of substrate sequence VAGRSMRP (SEQ ID NO: 484) by 5 nM MMP9.
  • FIG. 4 is a graph depicting correlation of substrate sequence frequency and function.
  • FIGS. 5A and 5B are a series of graphs depicting cleavage of the substrate pool SMP39 by 60 nM MMP14.
  • FIGS. 6A and 6B are a series of graphs depicting cleavage of the substrate sequence QNQALRMA (SEQ ID NO: 15) by 30 nM MMP14.
  • FIGS. 7A and 7B are a series of schematic representations of the peptide display platforms used in the working examples provided herein.
  • FIG. 7A is a schematic representation of the sequence of the display platform referred to herein as “Display Platform CYTX-DP-XXXXXXX” or “CYTX-DP-XXXXXXX” (SEQ ID NO: 512).
  • FIG. 7A is a schematic representation of the sequence of the display platform referred to herein as “Display Platform CYTX-DP-XXXXXXXXX” or “CYTX-DP-XXXXXXXXX” (SEQ ID NO: 512).
  • FIG. 7B is a schematic representation of the sequence of the display platform referred to herein as “Display Platform SP-CYTX-DP-XXXXXXXX” or “SP-CYTX-DP-XXXXXXXX” (SEQ ID NO: 513), where SP-CYTX-DP-XXXXXXXX is the CYTX-DP-XXXXXXXX platform with a signal peptide.
  • CM cleavable moiety
  • MMP matrix metalloprotease
  • CM when displayed in a peptide display platform, exhibit a number of desirable cleavage characteristics when exposed to an MMP protease under specified conditions.
  • Table 9 depicts (a) the percentage of MMP9-selected substrates tested in the CYTX-DP display platform that exhibited at least 20% cleavage when incubated with 50 nM human MMP9 for 1 hour at 37° C.
  • a MMP9 substrate when displayed in the CYTX-DP platform exhibits at least 20% cleavage when incubated with 50 nM human MMP9 for 1 hour at 37° C. in 50 mM Tris-HCl, pH 7.4, supplemented with 150 mM NaCl, 10 mM CaCl 2 , and 0.05% (w/v) Brij-35. In some embodiments, a MMP9 substrate when displayed in the CYTX-DP platform exhibits less than 20% cleavage when incubated with 500 pM human plasmin for 1 hour at 37° C.
  • a MMP9 substrate when displayed in the CYTX-DP platform exhibits at least 20% cleavage when incubated with 50 nM human MMP9 for 1 hour at 37° C. in 50 mM Tris-HCl, pH 7.4, supplemented with 150 mM NaCl, 10 mM CaCl 2 , and 0.05% (w/v) Brij-35 and exhibits less than 20% cleavage when incubated with 500 pM human plasmin for 1 hour at 37° C. in 50 mM Tris-HCl, pH 7.4, supplemented with 100 mM NaCl, 0.01% Tween20 and 1 mM EDTA.
  • a MMP14 substrate exhibits at least 20% cleavage when incubated with 50 nM human MMP14 for 1 hour at 37° C. in 50 mM HEPES, pH 6.8, supplemented with 10 mM CaCl 2 , and 0.5 mM MgCl 2 .
  • a MMP14 substrate when displayed in the CYTX-DP platform exhibits less than 20% cleavage when incubated with 500 pM human plasmin for 1 hour at 37° C. in 50 mM Tris-HCl, pH 7.4, supplemented with 100 mM NaCl, 0.01% Tween20 and 1 mM EDTA.
  • a MMP14 substrate exhibits at least 20% cleavage when incubated with 50 nM human MMP14 for 1 hour at 37° C. in 50 mM HEPES, pH 6.8, supplemented with 10 mM CaCl 2 , and 0.5 mM MgCl 2 and exhibits less than 20% cleavage when incubated with 500 pM human plasmin for 1 hour at 37° C. in 50 mM Tris-HCl, pH 7.4, supplemented with 100 mM NaCl, 0.01% Tween20 and 1 mM EDTA.
  • the observed k cat /K M value of a substrate in an activatable antibody for MMP9 is greater than 100 M ⁇ 1 s ⁇ 1 . In some embodiments, the observed k cat /K M value of a substrate in an activatable antibody for MMP9 is greater than 1,000 M ⁇ 1 s ⁇ 1 . In some embodiments, the observed k cat /K M value of a substrate in an activatable antibody for MMP9 is greater than 10,000 M ⁇ 1 s ⁇ 1 .
  • the observed k cat /K M value of a substrate in an activatable antibody for MMP14 is greater than 100 M ⁇ 1 s ⁇ 1 . In some embodiments, the observed k cat /K M value of a substrate in an activatable antibody for MMP14 is greater than 1,000 M ⁇ 1 s ⁇ 1 . In some embodiments, the observed k cat /K M value of a substrate in an activatable antibody for MMP14 is greater than 10,000 M ⁇ 1 s ⁇ 1 .
  • the disclosure also provides antibodies that include one or more of these MMP-cleavable substrates.
  • these MMP-cleavable substrates are useful when conjugating antibodies to one or more additional agents to produce conjugated antibodies.
  • These MMP-cleavable are useful in activatable antibody constructs.
  • the conjugated antibodies and/or activatable antibodies include an antibody or antigen-binding fragment thereof (AB) that specifically binds a target.
  • AB antibody or antigen-binding fragment thereof
  • targets of an AB include, but are not necessarily limited to, cell surface receptors and secreted binding proteins (e.g., growth factors), soluble enzymes, structural proteins (e.g. collagen, fibronectin) and the like.
  • conjugated antibodies and/or activatable antibodies have an AB that binds an extracellular target, usually an extracellular protein target.
  • conjugated antibodies and/or activatable antibodies are designed for cellular uptake and are switchable inside a cell.
  • the AB is a binding partner for any target listed in Table 1.
  • the AB is or is derived from an antibody listed in Table 2.
  • Target Avastin TM (bevacizumab) VEGF Lucentis TM (ranibizumab) VEGF Erbitux TM (cetuximab) EGFR Vectibix TM (panitumumab) EGFR Remicade TM (infliximab) TNF ⁇ Humira TM (adalimumab) TNF ⁇ Tysabri TM (natalizumab) Integrin ⁇ 4 Simulect TM (basiliximab) IL2R Soliris TM (eculizumab) Complement C5 Raptiva TM (efalizumab) CD11a Bexxar TM (tositumomab) CD20 Zevalin TM (ibritumomab tiuxetan) CD20 Rituxan TM (rituximab) CD20 Ocrelizumab CD20 Arzerra TM (ofatumumumumum
  • Exemplary conjugated antibodies and/or activatable antibodies of the disclosure include, for example, antibodies that bind interleukin 6 receptor (IL-6R) and that include a heavy chain and a light chain that are, or are derived from, the antibody referred to herein as the“Av1” antibody, which binds interleukin-6 receptor (IL-6R).
  • IL-6R interleukin 6 receptor
  • the amino acid sequences for the Av1 heavy chain and the Av1 light chain are shown below in SEQ ID NO: 54 and SEQ ID NO: 55, respectively.
  • Exemplary conjugated antibodies and/or activatable antibodies of the disclosure include, for example, antibodies that bind interleukin 6 receptor (IL-6R) and that include a heavy chain and a light chain that are, or are derived from, the Av1 antibody and a masking moiety.
  • exemplary conjugated antibodies and/or activatable antibodies of the disclosure include an amino acid sequence attached to the N-terminus of the AV1 light chain.
  • N-terminal amino acid sequences include, for example, YGSCSWNYVHIFMDC (SEQ ID NO: 161); QGDFDIPFPAHWVPIT (SEQ ID NO: 162); MGVPAGCVWNYAHIFMDC (SEQ ID NO: 163); QGQSGQYGSCSWNYVHIFMDC (SEQ ID NO: 164); QGQSGQGDFDIPFPAHWVPIT (SEQ ID NO: 165); or QGQSGQMGVPAGCVWNYAHIFMDC (SEQ ID NO: 166). It is also to be appreciated that such amino acid sequences can be attached to the N-terminus of the AV1 heavy chain or to the C-terminus of the AV1 heavy or light chain.
  • Exemplary activatable antibodies of the disclosure include, for example, antibodies that bind Epidermal Growth Factor Receptor (EGFR) and that include a heavy chain and a light chain that are, or are derived from, an antibody selected from the group consisting of the antibody referred to herein as the“c225v5” antibody, the antibody referred to herein as the“c225v4” antibody, and the antibody referred to herein as the“c225v6” antibody, each of which binds EGFR.
  • EGFR Epidermal Growth Factor Receptor
  • the c225v5 antibody, the c225v4 antibody, and the c225v6 antibody share the same light chain sequence, referred to herein as “c225 light chain.”
  • the amino acid sequences for the c225v5 heavy chain, the c225v4 antibody, the c225v6 antibody, and the c225 light chain are shown below.
  • Exemplary conjugated antibodies and/or activatable antibodies of the disclosure include, for example, antibodies that bind a Jagged target, e.g., Jagged-1, Jagged-2 and/or both Jagged-1 and Jagged-2, and that include a combination of a variable heavy chain region and a variable light chain region that are, or are derived from, the variable heavy chain and variable light chain sequences shown below.
  • a Jagged target e.g., Jagged-1, Jagged-2 and/or both Jagged-1 and Jagged-2
  • Exemplary conjugated antibodies and/or activatable antibodies of the disclosure include, for example, antibodies that bind a Jagged target, e.g., Jagged-1, Jagged-2 and/or both Jagged-1 and Jagged-2, and that include a combination of a heavy chain region and a light chain region that are, or are derived from, the heavy chain and light chain sequences shown below.
  • a Jagged target e.g., Jagged-1, Jagged-2 and/or both Jagged-1 and Jagged-2
  • 4D11 Light Chain sequence (SEQ ID NO: 111) DIQMTQSPSSLSASVGDRVTITCRASQSISSYLNWYQQKPGKAPKLLIYA ASSLQSGVPSRFSGSGSGTDFTLTISSLQPEDFATYYCQQTVVAPPLFGQ GTKVEIKRTVAAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREAKVQWKV DNALQSGNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQG LSSPVTKSFNRGEC 4D11 Heavy Chain sequence: (SEQ ID NO: 112) EVQLLESGGGLVQPGGSLRLSCAASGFTFSSYAMSWVRQAPGKGLEWVSS IDPEGRQTYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCAKDI GGRSAFDYWGQGTLVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKD YFPEPVTVSWNSGALTSGV
  • the activatable antibodies and activatable antibody compositions provided herein contain at least an antibody or antibody fragment thereof (collectively referred to as AB throughout the disclosure) that specifically binds a target, e.g., a human target, wherein the AB is modified by a masking moiety (MM).
  • AB an antibody or antibody fragment thereof
  • MM masking moiety
  • the masking moiety is selected for use with a specific antibody or antibody fragment.
  • suitable masking moieties for use with antibodies that bind EGFR include MMs that include the sequence CISPRG (SEQ ID NO: 167).
  • the MM can include a sequence such as CISPRGC (SEQ ID NO: 497); CISPRGCG (SEQ ID NO: 168); CISPRGCPDGPYVMY (SEQ ID NO: 160); CISPRGCPDGPYVM (SEQ ID NO: 169), CISPRGCEPGTYVPT (SEQ ID NO: 170) and CISPRGCPGQIWHPP (SEQ ID NO: 171).
  • Suitable masking moieties include any of the EGFR-specific masks disclosed in PCT Publication No. WO 2010/081173, such as, by way of non-limiting example, GSHCLIPINMGAPSC (SEQ ID NO: 172); CISPRGCGGSSASQSGQGSHCLIPINMGAPSC (SEQ ID NO: 173); CNHHYFYTCGCISPRGCPG (SEQ ID NO: 174); ADHVFWGSYGCISPRGCPG (SEQ ID NO: 175); CHHVYWGHCGCISPRGCPG (SEQ ID NO: 176); CPHFTTTSCGCISPRGCPG (SEQ ID NO: 177); CNHHYHYYCGCISPRGCPG (SEQ ID NO: 178); CPHVSFGSCGCISPRGCPG (SEQ ID NO: 179); CPYYTLSYCGCISPRGCPG (SEQ ID NO: 180); CNHVYFGTCGCISPRGCPG (SEQ ID NO: 181); CNHFTLTTCGCISPRGC
  • Suitable masking moieties for use with antibodies that bind a Jagged target include, by way of non-limiting example, masking moieties that include a sequence such as QGQSGQCNIWLVGGDCRGWQG (SEQ ID NO: 496); QGQSGQGQQWCNIWINGGDCRGWNG (SEQ ID NO: 201); PWCMQRQDFLRCPQP (SEQ ID NO: 202); QLGLPAYMCTFECLR (SEQ ID NO: 203); CNLWVSGGDCGGLQG (SEQ ID NO: 204); SCSLWTSGSCLPHSP (SEQ ID NO: 205); YCLQLPHYMQAMCGR (SEQ ID NO: 206); CFLYSCTDVSYWNNT (SEQ ID NO: 207); PWCMQRQDYLRCPQP (SEQ ID NO: 208); CNLWISGGDCRGLAG (SEQ ID NO: 209); CNLW
  • Suitable masking moieties for use with antibodies that bind an interleukin 6 target include, by way of non-limiting example, masking moieties that include a sequence such as QGQSGQYGSCSWNYVHIFMDC (SEQ ID NO: 264); QGQSGQGDFDIPFPAHWVPIT (SEQ ID NO: 265); QGQSGQMGVPAGCVWNYAHIFMDC (SEQ ID NO: 266); YRSCNWNYVSIFLDC (SEQ ID NO: 267); PGAFDIPFPAHWVPNT (SEQ ID NO: 268); ESSCVWNYVHIYMDC (SEQ ID NO: 269); YPGCKWNYDRIFLDC (SEQ ID NO: 270); YRTCSWNYVGIFLDC (SEQ ID NO: 271); YGSCSWNYVHIFMDC (SEQ ID NO: 161); YGSCSWNYVHIFLDC (SEQ ID NO: 264).
  • the K d of the AB modified with a MM towards the target is at least 5, 10, 25, 50, 100, 250, 500, 1,000, 2,500, 5,000, 10,000, 50,000, 100,000, 500,000, 1,000,000, 5,000,000, 10,000,000, 50,000,000 or greater, or between 5-10, 10-100, 10-1,000, 10-10,000, 10-100,000, 10-1,000,000, 10-10,000,000, 100-1,000, 100-10,000, 100-100,000, 100-1,000,000, 100-10,000,000, 1,000-10,000, 1,000-100,000, 1,000-1,000,000, 1000-10,000,000, 10,000-100,000, 10,000-1,000,000, 10,000-10,000,000, 100,000-1,000,000, or 100,000-10,000,000 times greater than the K d of the AB not modified with an MM or of the parental AB towards the target.
  • the binding affinity of the AB modified with a MM towards the target is at least 2, 3, 4, 5, 10, 20, 25, 40, 50, 100, 250, 500, 1,000, 2,500, 5,000, 10,000, 50,000, 100,000, 500,000, 1,000,000, 5,000,000, 10,000,000, 50,000,000 or greater, or between 5-10, 10-100, 10-1,000, 10-10,000, 10-100,000, 10-1,000,000, 10-10,000,000, 100-1,000, 100-10,000, 100-100,000, 100-1,000,000, 100-10,000,000, 1,000-10,000, 1,000-100,000, 1,000-1,000,000, 1000-10,000,000, 10,000-100,000, 10,000-1,000,000, 10,000-10,000,000, 100,000-1,000,000, or 100,000-10,000,000 times lower than the binding affinity of the AB not modified with an MM or of the parental AB towards the target.
  • the dissociation constant (K d ) of the MM towards the AB is generally greater than the K d of the AB towards the target.
  • the K d of the MM towards the AB can be at least 5, 10, 25, 50, 100, 250, 500, 1,000, 2,500, 5,000, 10,000, 100,000, 1,000,000 or even 10,000,000 times greater than the K d of the AB towards the target.
  • the binding affinity of the MM towards the AB is generally lower than the binding affinity of the AB towards the target.
  • the binding affinity of MM towards the AB can be at least 5, 10, 25, 50, 100, 250, 500, 1,000, 2,500, 5,000, 10,000, 100,000, 1,000,000 or even 10,000,000 times lower than the binding affinity of the AB towards the target.
  • the AB When the AB is modified with a MM and is in the presence of the target specific binding of the AB to its target is reduced or inhibited, as compared to the specific binding of the AB not modified with an MM or the specific binding of the parental AB to the target.
  • the AB's ability to bind the target when modified with an MM can be reduced by at least 50%, 60%, 70%, 80%, 90%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% and even 100% for at least 2, 4, 6, 8, 12, 28, 24, 30, 36, 48, 60, 72, 84, or 96 hours, or 5, 10, 15, 30, 45, 60, 90, 120, 150, or 180 days, or 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, or 12 months or more when measured in vivo or in an in vitro assay.
  • the MM inhibits the binding of the AB to the target.
  • the MM binds the antigen binding domain of the AB and inhibits binding of the AB to the target.
  • the MM can sterically inhibit the binding of the AB to the target.
  • the MM can allosterically inhibit the binding of the AB to its target.
  • the AB when the AB is modified or coupled to a MM and in the presence of target there is no binding or substantially no binding of the AB to the target, or no more than 0.001%, 0.01%, 0.1%, 1%, 2%, 3%, 4%, 5%, 6%, 7%, 8%, 9%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, or 50% binding of the AB to the target, as compared to the binding of the AB not modified with an MM, the parental AB, or the AB not coupled to an MM to the target, for at least 2, 4, 6, 8, 12, 28, 24, 30, 36, 48, 60, 72, 84, or 96 hours, or 5, 10, 15, 30, 45, 60, 90, 120, 150, or 180 days, or 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, or 12 months or longer when measured in vivo or in an in vitro assay.
  • the MM When an AB is coupled to or modified by a MM, the MM ‘masks’ or reduces or otherwise inhibits the specific binding of the AB to the target. When an AB is coupled to or modified by a MM, such coupling or modification can effect a structural change that reduces or inhibits the ability of the AB to specifically bind its target.
  • An AB coupled to or modified with an MM can be represented by the following formulae (in order from an amino (N) terminal region to carboxyl (C) terminal region:
  • MM is a masking moiety
  • the AB is an antibody or antibody fragment thereof
  • the L is a linker.
  • linkers e.g., flexible linkers
  • the MM is not a natural binding partner of the AB. In some embodiments, the MM contains no or substantially no homology to any natural binding partner of the AB. In some embodiments, the MM is no more than 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, or 80% similar to any natural binding partner of the AB. In some embodiments, the MM is no more than 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, or 80% identical to any natural binding partner of the AB.
  • the MM is no more than 25% identical to any natural binding partner of the AB. In some embodiments, the MM is no more than 50% identical to any natural binding partner of the AB. In some embodiments, the MM is no more than 20% identical to any natural binding partner of the AB. In some embodiments, the MM is no more than 10% identical to any natural binding partner of the AB.
  • the activatable antibodies include an AB that is modified by an MM and also includes one or more cleavable moieties (CM). Such activatable antibodies exhibit activatable/switchable binding, to the AB's target.
  • Activatable antibodies generally include an antibody or antibody fragment (AB), modified by or coupled to a masking moiety (MM) and a modifiable or cleavable moiety (CM).
  • CM contains an amino acid sequence that serves as a substrate for at least one matrix metalloprotease of interest.
  • the elements of the activatable antibodies are arranged so that the MM and CM are positioned such that in a cleaved (or relatively active) state and in the presence of a target, the AB binds a target while in an uncleaved (or relatively inactive) state in the presence of the target, specific binding of the AB to its target is reduced or inhibited.
  • the specific binding of the AB to its target can be reduced due to the inhibition or masking of the AB's ability to specifically bind its target by the MM.
  • the K d of the AB modified with a MM and a CM towards the target is at least 5, 10, 20, 25, 40, 50, 100, 250, 500, 1,000, 2,500, 5,000, 10,000, 50,000, 100,000, 500,000, 1,000,000, 5,000,000, 10,000,000, 50,000,000 or greater, or between 5-10, 10-100, 10-1,000, 10-10,000, 10-100,000, 10-1,000,000, 10-10,000,000, 100-1,000, 100-10,000, 100-100,000, 100-1,000,000, 100-10,000,000, 1,000-10,000, 1,000-100,000, 1,000-1,000,000, 1000-10,000,000, 10,000-100,000, 10,000-1,000,000, 10,000-10,000,000, 100,000-1,000,000, or 100,000-10,000,000 times greater than the K d of the AB not modified with an MM and a CM or of the parental AB towards the target.
  • the binding affinity of the AB modified with a MM and a CM towards the target is at least 2, 3, 4, 5, 10, 20, 25, 40, 50, 100, 250, 500, 1,000, 2,500, 5,000, 10,000, 50,000, 100,000, 500,000, 1,000,000, 5,000,000, 10,000,000, 50,000,000 or greater, or between 5-10, 10-100, 10-1,000, 10-10,000, 10-100,000, 10-1,000,000, 10-10,000,000, 100-1,000, 100-10,000, 100-100,000, 100-1,000,000, 100-10,000,000, 1,000-10,000, 1,000-100,000, 1,000-1,000,000, 1000-10,000,000, 10,000-100,000, 10,000-1,000,000, 10,000-10,000,000, 100,000-1,000,000, or 100,000-10,000,000 times lower than the binding affinity of the AB not modified with an MM and a CM or of the parental AB towards the target.
  • the AB When the AB is modified with a MM and a CM and is in the presence of the target but not in the presence of a modifying agent (for example a MMP), specific binding of the AB to its target is reduced or inhibited, as compared to the specific binding of the AB not modified with an MM and a CM or of the parental AB to the target.
  • a modifying agent for example a MMP
  • the AB's ability to bind the target when modified with an MM and a CM can be reduced by at least 50%, 60%, 70%, 80%, 90%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% and even 100% for at least 2, 4, 6, 8, 12, 28, 24, 30, 36, 48, 60, 72, 84, or 96 hours or 5, 10, 15, 30, 45, 60, 90, 120, 150, or 180 days, or 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, or 12 months or longer when measured in vivo or in an in vitro assay.
  • cleaved state refers to the condition of the activatable antibodies following modification of the CM by at least one matrix metalloprotease.
  • uncleaved state refers to the condition of the activatable antibodies in the absence of cleavage of the CM by a MMP.
  • activatable antibodies is used herein to refer to an activatable antibody in both its uncleaved (native) state, as well as in its cleaved state.
  • a cleaved activatable antibody may lack an MM due to cleavage of the CM by protease, resulting in release of at least the MM (e.g., where the MM is not joined to the activatable antibodies by a covalent bond (e.g., a disulfide bond between cysteine residues).
  • activatable or switchable By activatable or switchable is meant that the activatable antibody exhibits a first level of binding to a target when in a inhibited, masked or uncleaved state (i.e., a first conformation), and a second level of binding to the target in the uninhibited, unmasked and/or cleaved state (i.e., a second conformation), where the second level of target binding is greater than the first level of binding.
  • the access of target to the AB of the activatable antibody is greater in the presence of a cleaving agent capable of cleaving the CM than in the absence of such a cleaving agent.
  • the AB when the activatable antibody is in the uncleaved state, the AB is inhibited from target binding and can be masked from target binding (i.e., the first conformation is such the AB cannot bind the target), and in the cleaved state the AB is not inhibited or is unmasked to target binding.
  • the CM and AB of the activatable antibodies are selected so that the AB represents a binding moiety for a given target, and the CM represents a substrate for a MMP that is co-localized with the target at a treatment site or diagnostic site in a subject.
  • the activatable antibodies disclosed herein find particular use where, for example, a MMP capable of cleaving a site in the CM is present at relatively higher levels in target-containing tissue of a treatment site or diagnostic site than in tissue of non-treatment sites (for example in healthy tissue).
  • activatable antibodies provide for reduced toxicity and/or adverse side effects that could otherwise result from binding of the AB at non-treatment sites if the AB were not masked or otherwise inhibited from binding to the target.
  • an activatable antibody can be designed by selecting an AB of interest and constructing the remainder of the activatable antibody so that, when conformationally constrained, the MM provides for masking of the AB or reduction of binding of the AB to its target. Structural design criteria can be to be taken into account to provide for this functional feature.
  • Dynamic range generally refers to a ratio of (a) a maximum detected level of a parameter under a first set of conditions to (b) a minimum detected value of that parameter under a second set of conditions.
  • the dynamic range refers to the ratio of (a) a maximum detected level of target protein binding to an activatable antibody in the presence of a MMP capable of cleaving the CM of the activatable antibodies to (b) a minimum detected level of target protein binding to an activatable antibody in the absence of the protease.
  • the dynamic range of an activatable antibody can be calculated as the ratio of the equilibrium dissociation constant of an activatable antibody cleaving agent (e.g., enzyme) treatment to the equilibrium dissociation constant of the activatable antibodies cleaving agent treatment.
  • the greater the dynamic range of an activatable antibody the better the switchable phenotype of the activatable antibody.
  • Activatable antibodies having relatively higher dynamic range values exhibit more desirable switching phenotypes such that target protein binding by the activatable antibodies occurs to a greater extent (e.g., predominantly occurs) in the presence of a cleaving agent (e.g., enzyme) capable of cleaving the CM of the activatable antibodies than in the absence of a cleaving agent.
  • Activatable antibodies can be provided in a variety of structural configurations. Exemplary formulae for activatable antibodies are provided below. It is specifically contemplated that the N- to C-terminal order of the AB, MM and CM may be reversed within an activatable antibody. It is also specifically contemplated that the CM and MM may overlap in amino acid sequence, e.g., such that the CM is contained within the MM.
  • activatable antibodies can be represented by the following formula (in order from an amino (N) terminal region to carboxyl (C) terminal region:
  • MM is a masking moiety
  • CM is a cleavable moiety
  • AB is an antibody or fragment thereof.
  • MM and CM are indicated as distinct components in the formulae above, in all exemplary embodiments (including formulae) disclosed herein it is contemplated that the amino acid sequences of the MM and the CM could overlap, e.g., such that the CM is completely or partially contained within the MM.
  • the formulae above provide for additional amino acid sequences that may be positioned N-terminal or C-terminal to the activatable antibodies elements.
  • the MM is not a natural binding partner of the AB. In some embodiments, the MM contains no or substantially no homology to any natural binding partner of the AB. In some embodiments, the MM is no more than 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, or 80% similar to any natural binding partner of the AB. In some embodiments, the MM is no more than 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, or 80% identical to any natural binding partner of the AB.
  • the MM is no more than 50% identical to any natural binding partner of the AB. In some embodiments, the MM is no more than 25% identical to any natural binding partner of the AB. In some embodiments, the MM is no more than 20% identical to any natural binding partner of the AB. In some embodiments, the MM is no more than 10% identical to any natural binding partner of the AB.
  • the activatable antibody construct may be desirable to insert one or more linkers, e.g., flexible linkers, into the activatable antibody construct so as to provide for flexibility at one or more of the MM-CM junction, the CM-AB junction, or both.
  • the AB, MM, and/or CM may not contain a sufficient number of residues (e.g., Gly, Ser, Asp, Asn, especially Gly and Ser, particularly Gly) to provide the desired flexibility.
  • the switchable phenotype of such activatable antibody constructs may benefit from introduction of one or more amino acids to provide for a flexible linker.
  • a flexible linker can be operably inserted to facilitate formation and maintenance of a cyclic structure in the uncleaved activatable antibody.
  • an activatable antibody comprises one of the following formulae (where the formula below represent an amino acid sequence in either N- to C-terminal direction or C- to N-terminal direction):
  • MM, CM, and AB are as defined above; wherein L1 and L2 are each independently and optionally present or absent, are the same or different flexible linkers that include at least 1 flexible amino acid (e.g., Gly).
  • the formulae above provide for additional amino acid sequences that may be positioned N-terminal or C-terminal to the activatable antibodies elements.
  • targeting moieties e.g., a ligand for a receptor of a cell present in a target tissue
  • serum half-life extending moieties e.g., polypeptides that bind serum proteins, such as immunoglobulin (e.g., IgG) or serum albumin (e.g., human serum albumin (HAS)).
  • immunoglobulin e.g., IgG
  • serum albumin e.g., human serum albumin (HAS)
  • the CM is specifically cleaved by at least one MMP at a rate of about 0.001-1500 ⁇ 10 4 M ⁇ 1 S ⁇ 1 or at least 0.001, 0.005, 0.01, 0.05, 0.1, 0.5, 1, 2.5, 5, 7.5, 10, 15, 20, 25, 50, 75, 100, 125, 150, 200, 250, 500, 750, 1000, 1250, or 1500 ⁇ 10 4 M ⁇ 1 S ⁇ 1 .
  • CM For specific cleavage by an enzyme, contact between the enzyme and CM is made.
  • the activatable antibody comprising an AB coupled to a MM and a CM
  • the CM can be cleaved.
  • Sufficient enzyme activity can refer to the ability of the enzyme to make contact with the CM and effect cleavage. It can readily be envisioned that an enzyme may be in the vicinity of the CM but unable to cleave because of other cellular factors or protein modification of the enzyme.
  • Linkers suitable for use in compositions described herein are generally ones that provide flexibility of the modified AB or the activatable antibodies to facilitate the inhibition of the binding of the AB to the target. Such linkers are generally referred to as flexible linkers.
  • Suitable linkers can be readily selected and can be of any of a suitable of different lengths, such as from 1 amino acid (e.g., Gly) to 20 amino acids, from 2 amino acids to 15 amino acids, from 3 amino acids to 12 amino acids, including 4 amino acids to 10 amino acids, 5 amino acids to 9 amino acids, 6 amino acids to 8 amino acids, or 7 amino acids to 8 amino acids, and may be 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 amino acids in length.
  • Exemplary flexible linkers include glycine polymers (G)n, glycine-serine polymers (including, for example, (GS)n, (GSGGS)n (SEQ ID NO: 1) and (GGGS)n (SEQ ID NO: 2), where n is an integer of at least one), glycine-alanine polymers, alanine-serine polymers, and other flexible linkers known in the art.
  • Glycine and glycine-serine polymers are relatively unstructured, and therefore may be able to serve as a neutral tether between components. Glycine accesses significantly more phi-psi space than even alanine, and is much less restricted than residues with longer side chains (see Scheraga, Rev.
  • Exemplary flexible linkers include, but are not limited to Gly-Gly-Ser-Gly (SEQ ID NO: 3), Gly-Gly-Ser-Gly-Gly (SEQ ID NO: 4), Gly-Ser-Gly-Ser-Gly (SEQ ID NO: 5), Gly-Ser-Gly-Gly-Gly (SEQ ID NO: 6), Gly-Gly-Gly-Ser-Gly (SEQ ID NO: 7), Gly-Ser-Ser-Ser-Gly (SEQ ID NO: 8), and the like.
  • an activatable antibodies can include linkers that are all or partially flexible, such that the linker can include a flexible linker as well as one or more portions that confer less flexible structure to provide for a desired activatable antibodies structure.
  • the activatable antibodies described herein also include an agent conjugated to the activatable antibody.
  • the conjugated agent is a therapeutic agent, such as an anti-inflammatory and/or an antineoplastic agent.
  • the agent is conjugated to a carbohydrate moiety of the activatable antibody, for example, in some embodiments, where the carbohydrate moiety is located outside the antigen-binding region of the antibody or antigen-binding fragment in the activatable antibody.
  • the agent is conjugated to a sulfhydryl group of the antibody or antigen-binding fragment in the activatable antibody.
  • the agent is a cytotoxic agent such as a toxin (e.g., an enzymatically active toxin of bacterial, fungal, plant, or animal origin, or fragments thereof), or a radioactive isotope (i.e., a radioconjugate).
  • a cytotoxic agent such as a toxin (e.g., an enzymatically active toxin of bacterial, fungal, plant, or animal origin, or fragments thereof), or a radioactive isotope (i.e., a radioconjugate).
  • the agent is a detectable moiety such as, for example, a label or other marker.
  • the agent is or includes a radiolabeled amino acid, one or more biotinyl moieties that can be detected by marked avidin (e.g., streptavidin containing a fluorescent marker or enzymatic activity that can be detected by optical or calorimetric methods), one or more radioisotopes or radionuclides, one or more fluorescent labels, one or more enzymatic labels, and/or one or more chemiluminescent agents.
  • detectable moieties are attached by spacer molecules.
  • the disclosure also pertains to immunoconjugates comprising an antibody conjugated to a cytotoxic agent such as a toxin (e.g., an enzymatically active toxin of bacterial, fungal, plant, or animal origin, or fragments thereof), or a radioactive isotope (i.e., a radioconjugate).
  • a cytotoxic agent such as a toxin (e.g., an enzymatically active toxin of bacterial, fungal, plant, or animal origin, or fragments thereof), or a radioactive isotope (i.e., a radioconjugate).
  • Suitable cytotoxic agents include, for example, dolastatins and derivatives thereof (e.g. auristatin E, AFP, MMAF, MMAE, MMAD, DMAF, DMAE).
  • the agent is monomethyl auristatin E (MMAE) or monomethyl auristatin D (MMAD).
  • the agent is an agent selected from
  • the agent is a dolastatin. In some embodiments, the agent is an auristatin or derivative thereof. In some embodiments, the agent is auristatin E or a derivative thereof. In some embodiments, the agent is monomethyl auristatin E (MMAE). In some embodiments, the agent is monomethyl auristatin D (MMAD). In some embodiments, the agent is a maytansinoid or maytansinoid derivative. In some embodiments, the agent is DM1 or DM4. In some embodiments, the agent is a duocarmycin or derivative thereof. In some embodiments, the agent is a calicheamicin or derivative thereof. In some embodiments, the agent is a pyrrolobenzodiazepine.
  • the agent is linked to the AB using a maleimide caproyl-valine-citrulline linker or a maleimide PEG-valine-citrulline linker. In some embodiments, the agent is linked to the AB using a maleimide caproyl-valine-citrulline linker.
  • the agent is linked to the AB using a maleimide PEG-valine-citrulline linker
  • the agent is monomethyl auristatin D (MMAD) linked to the AB using a maleimide PEG-valine-citrulline-para-aminobenzyloxycarbonyl linker, and this linker payload construct is referred to herein as “vc-MMAD.”
  • the agent is monomethyl auristatin E (MMAE) linked to the AB using a maleimide PEG-valine-citrulline-para-aminobenzyloxycarbonyl linker, and this linker payload construct is referred to herein as “vc-MMAE.”
  • vc-MMAD monomethyl auristatin E
  • Enzymatically active toxins and fragments thereof that can be used include diphtheria A chain, nonbinding active fragments of diphtheria toxin, exotoxin A chain (from Pseudomonas aeruginosa ), ricin A chain, abrin A chain, modeccin A chain, alpha-sarcin, Aleurites fordii proteins, dianthin proteins, Phytolaca americana proteins (PAPI, PAPII, and PAP-S), momordica charantia inhibitor, curcin, crotin, sapaonaria officinalis inhibitor, gelonin, mitogellin, restrictocin, phenomycin, enomycin, and the tricothecenes.
  • a variety of radionuclides are available for the production of radioconjugated antibodies. Examples include 212 Bi, 131 I, 131 In, 90 Y, and 186 Re.
  • Conjugates of the antibody and cytotoxic agent are made using a variety of bifunctional protein-coupling agents such as N-succinimidyl-3-(2-pyridyldithiol) propionate (SPDP), iminothiolane (IT), bifunctional derivatives of imidoesters (such as dimethyl adipimidate HCL), active esters (such as disuccinimidyl suberate), aldehydes (such as glutareldehyde), bis-azido compounds (such as bis(p-azidobenzoyl) hexanediamine), bis-diazonium derivatives (such as bis-(p-diazoniumbenzoyl)-ethylenediamine), diisocyanates (such as tolyene 2,6-diisocyanate), and bis-active fluorine compounds (such as 1,5-difluoro-2,4-dinitrobenzene).
  • SPDP N-succinimidyl-3-(
  • a ricin immunotoxin can be prepared as described in Vitetta et al., Science 238: 1098 (1987).
  • Carbon-14-labeled 1-isothiocyanatobenzyl-3-methyldiethylene triaminepentaacetic acid (MX-DTPA) is an exemplary chelating agent for conjugation of radionucleotide to the antibody. (See WO94/11026).
  • Table 3 lists some of the exemplary pharmaceutical agents that may be employed in the herein described disclosure but in no way is meant to be an exhaustive list.
  • Auristatin E Monomethyl auristatin D
  • MMAE Monomethyl auristatin E
  • DMAE Desmethyl auristatin E
  • MMAF Desmethyl auristatin F
  • DMAF Auristatin derivatives, e.g., amides thereof
  • Auristatin tyramine Auristatin quinoline
  • Dolastatins Dolastatin derivatives
  • Dolastatin 16 Dpv Maytansinoids e.g.
  • DM-1; DM-4 Maytansinoid derivatives Duocarmycin Duocarmycin derivatives Alpha-amanitin Anthracyclines Doxorubicin Daunorubicin Bryostatins Camptothecin Camptothecin derivatives 7-substituted Camptothecin 10,11- Difluoromethylenedioxycamptothecin Combretastatins Debromoaplysiatoxin Kahalalide-F Discodermolide Ecteinascidins ANTIVIRALS Acyclovir Vira A Symmetrel ANTIFUNGALS Nystatin ADDITIONAL ANTI-NEOPLASTICS Adriamycin Cerubidine Bleomycin Alkeran Velban Oncovin Fluorouracil Methotrexate Thiotepa Bisantrene Novantrone Thioguanine Procarabizine Cytarabine ANTI-BACTERIALS Aminoglycosides Streptomycin Neomycin Kanamycin Amikacin
  • Coupling may be accomplished by any chemical reaction that will bind the two molecules so long as the antibody and the other moiety retain their respective activities.
  • This linkage can include many chemical mechanisms, for instance covalent binding, affinity binding, intercalation, coordinate binding and complexation.
  • the binding is, however, covalent binding.
  • Covalent binding can be achieved either by direct condensation of existing side chains or by the incorporation of external bridging molecules.
  • Many bivalent or polyvalent linking agents are useful in coupling protein molecules, such as the antibodies of the present disclosure, to other molecules.
  • representative coupling agents can include organic compounds such as thioesters, carbodiimides, succinimide esters, diisocyanates, glutaraldehyde, diazobenzenes and hexamethylene diamines.
  • the conjugated activatable antibody can also be modified for site-specific conjugation through modified amino acid sequences inserted or otherwise included in the activatable antibody sequence. These modified amino acid sequences are designed to allow for controlled placement and/or dosage of the conjugated agent within a conjugated activatable antibody.
  • the activatable antibody can be engineered to include cysteine substitutions at positions on light and heavy chains that provide reactive thiol groups and do not negatively impact protein folding and assembly, nor alter antigen binding.
  • the activatable antibody can be engineered to include or otherwise introduce one or more non-natural amino acid residues within the activatable antibody to provide suitable sites for conjugation.
  • the activatable antibody can be engineered to include or otherwise introduce enzymatically activatable peptide sequences within the activatable antibody sequence.
  • Suitable linkers are described in the literature. (See, for example, Ramakrishnan, S. et al., Cancer Res. 44:201-208 (1984) describing use of MBS (M-maleimidobenzoyl-N-hydroxysuccinimide ester). See also, U.S. Pat. No. 5,030,719, describing use of halogenated acetyl hydrazide derivative coupled to an antibody by way of an oligopeptide linker.
  • MBS M-maleimidobenzoyl-N-hydroxysuccinimide ester
  • suitable linkers include: (i) EDC (1-ethyl-3-(3-dimethylamino-propyl) carbodiimide hydrochloride; (ii) SMPT (4-succinimidyloxycarbonyl-alpha-methyl-alpha-(2-pridyl-dithio)-toluene (Pierce Chem. Co., Cat. (21558G); (iii) SPDP (succinimidyl-6 [3-(2-pyridyldithio) propionamido]hexanoate (Pierce Chem.
  • Sulfo-LC-SPDP sulfosuccinimidyl 6 [3-(2-pyridyldithio)-propianamide]hexanoate
  • sulfo-NHS N-hydroxysulfo-succinimide: Pierce Chem. Co., Cat. #24510 conjugated to EDC.
  • Additional linkers include, but are not limited to, SMCC, sulfo-SMCC, SPDB, or sulfo-SPDB.
  • linkers described above contain components that have different attributes, thus leading to conjugates with differing physio-chemical properties.
  • sulfo-NHS esters of alkyl carboxylates are more stable than sulfo-NHS esters of aromatic carboxylates.
  • NHS-ester containing linkers are less soluble than sulfo-NHS esters.
  • the linker SMPT contains a sterically hindered disulfide bond, and can form conjugates with increased stability.
  • Disulfide linkages are in general, less stable than other linkages because the disulfide linkage is cleaved in vitro, resulting in less conjugate available.
  • Sulfo-NHS in particular, can enhance the stability of carbodimide couplings.
  • Carbodimide couplings (such as EDC) when used in conjunction with sulfo-NHS, forms esters that are more resistant to hydrolysis than the carbodimide coupling reaction alone.
  • the linkers are cleavable. In some embodiments, the linkers are non-cleavable. In some embodiments, two or more linkers are present. The two or more linkers are all the same, i.e., cleavable or non-cleavable, or the two or more linkers are different, i.e., at least one cleavable and at least one non-cleavable.
  • ABs may be covalently attached to an agent through an intermediate linker having at least two reactive groups, one to react with AB and one to react with the agent.
  • the linker which may include any compatible organic compound, can be chosen such that the reaction with AB (or agent) does not adversely affect AB reactivity and selectivity. Furthermore, the attachment of linker to agent might not destroy the activity of the agent.
  • Suitable linkers for reaction with oxidized antibodies or oxidized antibody fragments include those containing an amine selected from the group consisting of primary amine, secondary amine, hydrazine, hydrazide, hydroxylamine, phenylhydrazine, semicarbazide and thiosemicarbazide groups.
  • Such reactive functional groups may exist as part of the structure of the linker, or may be introduced by suitable chemical modification of linkers not containing such groups.
  • suitable linkers for attachment to reduced ABs include those having certain reactive groups capable of reaction with a sulfhydryl group of a reduced antibody or fragment.
  • reactive groups include, but are not limited to: reactive haloalkyl groups (including, for example, haloacetyl groups), p-mercuribenzoate groups and groups capable of Michael-type addition reactions (including, for example, maleimides and groups of the type described by Mitra and Lawton, 1979, J. Amer. Chem. Soc. 101: 3097-3110).
  • suitable linkers for attachment to neither oxidized nor reduced Abs include those having certain functional groups capable of reaction with the primary amino groups present in unmodified lysine residues in the Ab.
  • Such reactive groups include, but are not limited to, NHS carboxylic or carbonic esters, sulfo-NHS carboxylic or carbonic esters, 4-nitrophenyl carboxylic or carbonic esters, pentafluorophenyl carboxylic or carbonic esters, acyl imidazoles, isocyanates, and isothiocyanates.
  • suitable linkers for attachment to neither oxidized nor reduced Abs include those having certain functional groups capable of reaction with the carboxylic acid groups present in aspartate or glutamate residues in the Ab, which have been activated with suitable reagents.
  • suitable activating reagents include EDC, with or without added NHS or sulfo-NHS, and other dehydrating agents utilized for carboxamide formation.
  • the functional groups present in the suitable linkers would include primary and secondary amines, hydrazines, hydroxylamines, and hydrazides.
  • the agent may be attached to the linker before or after the linker is attached to the AB. In certain applications it may be desirable to first produce an AB-linker intermediate in which the linker is free of an associated agent. Depending upon the particular application, a specific agent may then be covalently attached to the linker. In some embodiments, the AB is first attached to the MM, CM and associated linkers and then attached to the linker for conjugation purposes.
  • branched linkers that have multiple sites for attachment of agents are utilized.
  • a single covalent attachment to an AB would result in an AB-linker intermediate capable of binding an agent at a number of sites.
  • the sites may be aldehyde or sulfhydryl groups or any chemical site to which agents can be attached.
  • higher specific activity can be achieved by attachment of a single site linker at a plurality of sites on the AB.
  • This plurality of sites may be introduced into the AB by either of two methods. First, one may generate multiple aldehyde groups and/or sulfhydryl groups in the same AB. Second, one may attach to an aldehyde or sulfhydryl of the AB a “branched linker” having multiple functional sites for subsequent attachment to linkers.
  • the functional sites of the branched linker or multiple site linker may be aldehyde or sulfhydryl groups, or may be any chemical site to which linkers may be attached. Still higher specific activities may be obtained by combining these two approaches, that is, attaching multiple site linkers at several sites on the AB.
  • Peptide linkers that are susceptible to cleavage by enzymes of the complement system, such as but not limited to urokinase, tissue plasminogen activator, trypsin, plasmin, or another enzyme having proteolytic activity may be used in one embodiment of the present disclosure.
  • an agent is attached via a linker susceptible to cleavage by complement.
  • the antibody is selected from a class that can activate complement. The antibody-agent conjugate, thus, activates the complement cascade and releases the agent at the target site.
  • an agent is attached via a linker susceptible to cleavage by enzymes having a proteolytic activity such as a urokinase, a tissue plasminogen activator, plasmin, or trypsin.
  • cleavable linkers are useful in conjugated activatable antibodies that include an extracellular toxin, e.g., by way of non-limiting example, any of the extracellular toxins shown in Table 3.
  • Non-limiting examples of cleavable linker sequences are provided in Table 4.
  • agents may be attached via disulfide bonds (for example, the disulfide bonds on a cysteine molecule) to the AB. Since many tumors naturally release high levels of glutathione (a reducing agent) this can reduce the disulfide bonds with subsequent release of the agent at the site of delivery.
  • glutathione a reducing agent
  • the reducing agent that would modify a CM would also modify the linker of the conjugated activatable antibody.
  • linker in such a way as to optimize the spacing between the agent and the AB of the activatable antibody. This may be accomplished by use of a linker of the general structure:
  • W is either —NH—CH 2 — or —CH 2 —;
  • Q is an amino acid, peptide; and
  • n is an integer from 0 to 20.
  • the linker may comprise a spacer element and a cleavable element.
  • the spacer element serves to position the cleavable element away from the core of the AB such that the cleavable element is more accessible to the enzyme responsible for cleavage.
  • Certain of the branched linkers described above may serve as spacer elements.
  • linker to agent or of spacer element to cleavable element, or cleavable element to agent
  • attachment of linker to agent need not be particular mode of attachment or reaction. Any reaction providing a product of suitable stability and biological compatibility is acceptable.
  • an AB that is an antibody of a class that can activate complement is used.
  • the resulting conjugate retains both the ability to bind antigen and activate the complement cascade.
  • an agent is joined to one end of the cleavable linker or cleavable element and the other end of the linker group is attached to a specific site on the AB.
  • the agent has an hydroxy group or an amino group, it may be attached to the carboxy terminus of a peptide, amino acid or other suitably chosen linker via an ester or amide bond, respectively.
  • such agents may be attached to the linker peptide via a carbodimide reaction.
  • the agent contains functional groups that would interfere with attachment to the linker, these interfering functional groups can be blocked before attachment and deblocked once the product conjugate or intermediate is made.
  • the opposite or amino terminus of the linker is then used either directly or after further modification for binding to an AB that is capable of activating complement.
  • Linkers may be of any desired length, one end of which can be covalently attached to specific sites on the AB of the activatable antibody.
  • the other end of the linker or spacer element may be attached to an amino acid or peptide linker.
  • conjugates when administered to a subject, will accomplish delivery and release of the agent at the target site, and are particularly effective for the in vivo delivery of pharmaceutical agents, antibiotics, antimetabolites, antiproliferative agents and the like as presented in but not limited to those in Table 3.
  • the activatable antibody may be conjugated to one or more therapeutic agents using certain biochemical cross-linkers.
  • Cross-linking reagents form molecular bridges that tie together functional groups of two different molecules.
  • hetero-bifunctional cross-linkers can be used that eliminate unwanted homopolymer formation.
  • Peptidyl linkers cleavable by lysosomal proteases are also useful, for example, Val-Cit, Val-Ala or other dipeptides.
  • acid-labile linkers cleavable in the low-pH environment of the lysosome may be used, for example: bis-sialyl ether.
  • Other suitable linkers include cathepsin-labile substrates, particularly those that show optimal function at an acidic pH.
  • hetero-bifunctional cross-linkers are referenced in Table 5.
  • the conjugate may be designed so that the agent is delivered to the target but not released. This may be accomplished by attaching an agent to an AB either directly or via a non-cleavable linker.
  • non-cleavable linkers may include amino acids, peptides, D-amino acids or other organic compounds that may be modified to include functional groups that can subsequently be utilized in attachment to ABs by the methods described herein.
  • A-general formula for such an organic linker could be
  • W is either —NH—CH 2 — or —CH 2 —;
  • Q is an amino acid, peptide; and
  • n is an integer from 0 to 20.
  • a compound may be attached to ABs that do not activate complement.
  • this attachment may be accomplished using linkers that are susceptible to cleavage by activated complement or using linkers that are not susceptible to cleavage by activated complement.
  • the antibodies disclosed herein can also be formulated as immunoliposomes.
  • Liposomes containing the antibody are prepared by methods known in the art, such as described in Epstein et al., Proc. Natl. Acad. Sci. USA, 82: 3688 (1985); Hwang et al., Proc. Natl Acad. Sci. USA, 77: 4030 (1980); and U.S. Pat. Nos. 4,485,045 and 4,544,545. Liposomes with enhanced circulation time are disclosed in U.S. Pat. No. 5,013,556.
  • Particularly useful liposomes can be generated by the reverse-phase evaporation method with a lipid composition comprising phosphatidylcholine, cholesterol, and PEG-derivatized phosphatidylethanolamine (PEG-PE). Liposomes are extruded through filters of defined pore size to yield liposomes with the desired diameter.
  • Fab′ fragments of the antibody of the present disclosure can be conjugated to the liposomes as described in Martin et al., J. Biol. Chem., 257: 286-288 (1982) via a disulfide-interchange reaction.
  • Standard techniques are used for recombinant DNA, oligonucleotide synthesis, and tissue culture and transformation (e.g., electroporation, lipofection). Enzymatic reactions and purification techniques are performed according to manufacturer's specifications or as commonly accomplished in the art or as described herein. The foregoing techniques and procedures are generally performed according to conventional methods well known in the art and as described in various general and more specific references that are cited and discussed throughout the present specification. See e.g., Sambrook et al. Molecular Cloning: A Laboratory Manual (2d ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y. (1989)).
  • antibody refers to immunoglobulin molecules and immunologically active portions of immunoglobulin (Ig) molecules, i.e., molecules that contain an antigen binding site that specifically binds (immunoreacts with) an antigen.
  • immunoglobulin immunoglobulin
  • immunoglobulin immunoglobulin molecules
  • immunologically active portions of immunoglobulin (Ig) molecules i.e., molecules that contain an antigen binding site that specifically binds (immunoreacts with) an antigen.
  • Ig immunoglobulin
  • immunoglobulin (Ig) molecules i.e., molecules that contain an antigen binding site that specifically binds (immunoreacts with) an antigen.
  • specifically bind or “immunoreacts with” or “immunospecifically bind” is meant that the antibody reacts with one or more antigenic determinants of the desired antigen and does not react with other polypeptides or binds at much lower affinity (K d >10
  • the basic antibody structural unit is known to comprise a tetramer.
  • Each tetramer is composed of two identical pairs of polypeptide chains, each pair having one “light” (about 25 kDa) and one “heavy” chain (about 50-70 kDa).
  • the amino-terminal portion of each chain includes a variable region of about 100 to 110 or more amino acids primarily responsible for antigen recognition.
  • the carboxy-terminal portion of each chain defines a constant region primarily responsible for effector function.
  • antibody molecules obtained from humans relate to any of the classes IgG, IgM, IgA, IgE and IgD, which differ from one another by the nature of the heavy chain present in the molecule. Certain classes have subclasses as well, such as IgG 1 , IgG 2 , and others.
  • the light chain may be a kappa chain or a lambda chain.
  • mAb monoclonal antibody
  • CDRs complementarity determining regions
  • antigen binding site refers to the part of the immunoglobulin molecule that participates in antigen binding.
  • the antigen binding site is formed by amino acid residues of the N-terminal variable (“V”) regions of the heavy (“H”) and light (“L”) chains.
  • V N-terminal variable
  • L light
  • hypervariable regions Three highly divergent stretches within the V regions of the heavy and light chains, referred to as “hypervariable regions,” are interposed between more conserved flanking stretches known as “framework regions,” or “FRs”.
  • FR refers to amino acid sequences that are naturally found between, and adjacent to, hypervariable regions in immunoglobulins.
  • the three hypervariable regions of a light chain and the three hypervariable regions of a heavy chain are disposed relative to each other in three dimensional space to form an antigen-binding surface.
  • the antigen-binding surface is complementary to the three-dimensional surface of a bound antigen, and the three hypervariable regions of each of the heavy and light chains are referred to as “complementarity-determining regions,” or “CDRs.”
  • CDRs complementarity-determining regions
  • epitopic determinants include any protein determinant capable of specific binding to an immunoglobulin, an scFv, or a T-cell receptor.
  • epitopic determinants usually consist of chemically active surface groupings of molecules such as amino acids or sugar side chains and usually have specific three dimensional structural characteristics, as well as specific charge characteristics.
  • antibodies may be raised against N-terminal or C-terminal peptides of a polypeptide.
  • An antibody is said to specifically bind an antigen when the dissociation constant is ⁇ 1 ⁇ M; in some embodiments, ⁇ 100 nM and in some embodiments, ⁇ 10 nM.
  • the terms “specific binding,” “immunological binding,” and “immunological binding properties” refer to the non-covalent interactions of the type which occur between an immunoglobulin molecule and an antigen for which the immunoglobulin is specific.
  • the strength, or affinity of immunological binding interactions can be expressed in terms of the dissociation constant (K d ) of the interaction, wherein a smaller K d represents a greater affinity.
  • Immunological binding properties of selected polypeptides can be quantified using methods well known in the art. One such method entails measuring the rates of antigen-binding site/antigen complex formation and dissociation, wherein those rates depend on the concentrations of the complex partners, the affinity of the interaction, and geometric parameters that equally influence the rate in both directions.
  • both the “on rate constant” (K on ) and the “off rate constant” (K off ) can be determined by calculation of the concentrations and the actual rates of association and dissociation. (See Nature 361:186-87 (1993)).
  • the ratio of K off /K on enables the cancellation of all parameters not related to affinity, and is equal to the dissociation constant K d . (See, generally, Davies et al. (1990) Annual Rev Biochem 59:439-473).
  • An antibody of the present disclosure is said to specifically bind to the target, when the equilibrium binding constant (K d ) is ⁇ 1 ⁇ M, in some embodiments 100 nM, in some embodiments ⁇ 10 nM, and in some embodiments ⁇ 100 pM to about 1 pM, as measured by assays such as radioligand binding assays or similar assays known to those skilled in the art.
  • K d equilibrium binding constant
  • isolated polynucleotide shall mean a polynucleotide of genomic, cDNA, or synthetic origin or some combination thereof, which by virtue of its origin the “isolated polynucleotide” (1) is not associated with all or a portion of a polynucleotide in which the “isolated polynucleotide” is found in nature, (2) is operably linked to a polynucleotide which it is not linked to in nature, or (3) does not occur in nature as part of a larger sequence.
  • Polynucleotides in accordance with the disclosure include the nucleic acid molecules encoding the heavy chain immunoglobulin molecules shown herein, and nucleic acid molecules encoding the light chain immunoglobulin molecules shown herein.
  • isolated protein means a protein of cDNA, recombinant RNA, or synthetic origin or some combination thereof, which by virtue of its origin, or source of derivation, the “isolated protein” (1) is not associated with proteins found in nature, (2) is free of other proteins from the same source, e.g., free of murine proteins, (3) is expressed by a cell from a different species, or (4) does not occur in nature.
  • polypeptide is used herein as a generic term to refer to native protein, fragments, or analogs of a polypeptide sequence. Hence, native protein fragments, and analogs are species of the polypeptide genus.
  • Polypeptides in accordance with the disclosure comprise the heavy chain immunoglobulin molecules shown herein, and the light chain immunoglobulin molecules shown herein, as well as antibody molecules formed by combinations comprising the heavy chain immunoglobulin molecules with light chain immunoglobulin molecules, such as kappa light chain immunoglobulin molecules, and vice versa, as well as fragments and analogs thereof.
  • naturally-occurring refers to the fact that an object can be found in nature.
  • a polypeptide or polynucleotide sequence that is present in an organism (including viruses) that can be isolated from a source in nature and that has not been intentionally modified by man in the laboratory or otherwise is naturally-occurring.
  • operably linked refers to positions of components so described are in a relationship permitting them to function in their intended manner.
  • a control sequence “operably linked” to a coding sequence is ligated in such a way that expression of the coding sequence is achieved under conditions compatible with the control sequences.
  • control sequence refers to polynucleotide sequences that are necessary to effect the expression and processing of coding sequences to which they are ligated. The nature of such control sequences differs depending upon the host organism in prokaryotes, such control sequences generally include promoter, ribosomal binding site, and transcription termination sequence in eukaryotes, generally, such control sequences include promoters and transcription termination sequence.
  • control sequences is intended to include, at a minimum, all components whose presence is essential for expression and processing, and can also include additional components whose presence is advantageous, for example, leader sequences and fusion partner sequences.
  • polynucleotide as referred to herein means nucleotides of at least 10 bases in length, either ribonucleotides or deoxynucleotides or a modified form of either type of nucleotide.
  • the term includes single and double stranded forms of DNA.
  • oligonucleotide referred to herein includes naturally occurring, and modified nucleotides linked together by naturally occurring, and non-naturally occurring oligonucleotide linkages.
  • Oligonucleotides are a polynucleotide subset generally comprising a length of 200 bases or fewer. In some embodiments, oligonucleotides are 10 to 60 bases in length and in some embodiments, 12, 13, 14, 15, 16, 17, 18, 19, or 20 to 40 bases in length. Oligonucleotides are usually single stranded, e.g., for probes, although oligonucleotides may be double stranded, e.g., for use in the construction of a gene mutant. Oligonucleotides of the disclosure are either sense or antisense oligonucleotides.
  • nucleotides includes deoxyribonucleotides and ribonucleotides.
  • modified nucleotides referred to herein includes nucleotides with modified or substituted sugar groups and the like.
  • oligonucleotide linkages includes oligonucleotide linkages such as phosphorothioate, phosphorodithioate, phosphoroselerloate, phosphorodiselenoate, phosphoroanilothioate, phoshoraniladate, phosphoronmidate, and the like. See e.g., LaPlanche et al. Nucl. Acids Res.
  • oligonucleotide can include a label for detection, if desired.
  • Examples of unconventional amino acids include: 4 hydroxyproline, ⁇ -carboxyglutamate, ⁇ -N,N,N-trimethyllysine, ⁇ -N-acetyllysine, O-phosphoserine, N-acetylserine, N-formylmethionine, 3-methylhistidine, 5-hydroxylysine, ⁇ -N-methylarginine, and other similar amino acids and imino acids (e.g., 4-hydroxyproline).
  • the left-hand direction is the amino terminal direction and the right-hand direction is the carboxy-terminal direction, in accordance with standard usage and convention.
  • the left-hand end of single-stranded polynucleotide sequences is the 5′ end the left-hand direction of double-stranded polynucleotide sequences is referred to as the 5′ direction.
  • the direction of 5′ to 3′ addition of nascent RNA transcripts is referred to as the transcription direction sequence regions on the DNA strand having the same sequence as the RNA and that are 5′ to the 5′ end of the RNA transcript are referred to as “upstream sequences”, sequence regions on the DNA strand having the same sequence as the RNA and that are 3′ to the 3′ end of the RNA transcript are referred to as “downstream sequences”.
  • the term “substantial identity” means that two peptide sequences, when optimally aligned, such as by the programs GAP or BESTFIT using default gap weights, share at least 80 percent sequence identity, in some embodiments, at least 90 percent sequence identity, in some embodiments, at least 95 percent sequence identity, and in some embodiments, at least 99 percent sequence identity.
  • residue positions that are not identical differ by conservative amino acid substitutions.
  • amino acid sequences of antibodies or immunoglobulin molecules are contemplated as being encompassed by the present disclosure, providing that the variations in the amino acid sequence maintain at least 75%, in some embodiments, at least 80%, 90%, 95%, and in some embodiments, 99%.
  • conservative amino acid replacements are contemplated. Conservative replacements are those that take place within a family of amino acids that are related in their side chains.
  • amino acids are generally divided into families: (1) acidic amino acids are aspartate, glutamate; (2) basic amino acids are lysine, arginine, histidine; (3) non-polar amino acids are alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, tryptophan, and (4) uncharged polar amino acids are glycine, asparagine, glutamine, cysteine, serine, threonine, tyrosine.
  • the hydrophilic amino acids include arginine, asparagine, aspartate, glutamine, glutamate, histidine, lysine, serine, and threonine.
  • the hydrophobic amino acids include alanine, cysteine, isoleucine, leucine, methionine, phenylalanine, proline, tryptophan, tyrosine and valine.
  • Other families of amino acids include (i) serine and threonine, which are the aliphatic-hydroxy family; (ii) asparagine and glutamine, which are the amide containing family; (iii) alanine, valine, leucine and isoleucine, which are the aliphatic family; and (iv) phenylalanine, tryptophan, and tyrosine, which are the aromatic family.
  • Suitable amino- and carboxy-termini of fragments or analogs occur near boundaries of functional domains.
  • Structural and functional domains can be identified by comparison of the nucleotide and/or amino acid sequence data to public or proprietary sequence databases.
  • computerized comparison methods are used to identify sequence motifs or predicted protein conformation domains that occur in other proteins of known structure and/or function. Methods to identify protein sequences that fold into a known three-dimensional structure are known. Bowie et al. Science 253:164 (1991).
  • Suitable amino acid substitutions are those which: (1) reduce susceptibility to proteolysis, (2) reduce susceptibility to oxidation, (3) alter binding affinity for forming protein complexes, (4) alter binding affinities, and (5) confer or modify other physicochemical or functional properties of such analogs.
  • Analogs can include various muteins of a sequence other than the naturally-occurring peptide sequence. For example, single or multiple amino acid substitutions (for example, conservative amino acid substitutions) may be made in the naturally-occurring sequence (for example, in the portion of the polypeptide outside the domain(s) forming intermolecular contacts.
  • a conservative amino acid substitution should not substantially change the structural characteristics of the parent sequence (e.g., a replacement amino acid should not tend to break a helix that occurs in the parent sequence, or disrupt other types of secondary structure that characterizes the parent sequence).
  • Examples of art-recognized polypeptide secondary and tertiary structures are described in Proteins, Structures and Molecular Principles (Creighton, Ed., W. H. Freeman and Company, New York (1984)); Introduction to Protein Structure (C. Branden and J. Tooze, eds., Garland Publishing, New York, N.Y. (1991)); and Thornton et at. Nature 354:105 (1991).
  • polypeptide fragment refers to a polypeptide that has an amino terminal and/or carboxy-terminal deletion and/or one or more internal deletion(s), but where the remaining amino acid sequence is identical to the corresponding positions in the naturally-occurring sequence deduced, for example, from a full length cDNA sequence. Fragments typically are at least 5, 6, 8 or 10 amino acids long, in some embodiments, at least 14 amino acids long, in some embodiments, at least 20 amino acids long, usually at least 50 amino acids long, and in some embodiments, at least 70 amino acids long.
  • analog refers to polypeptides that are comprised of a segment of at least 25 amino acids that has substantial identity to a portion of a deduced amino acid sequence and that has specific binding to the target, under suitable binding conditions.
  • polypeptide analogs comprise a conservative amino acid substitution (or addition or deletion) with respect to the naturally-occurring sequence.
  • Analogs typically are at least 20 amino acids long, in some embodiments, at least 50 amino acids long or longer, and can often be as long as a full-length naturally-occurring polypeptide.
  • agent is used herein to denote a chemical compound, a mixture of chemical compounds, a biological macromolecule, or an extract made from biological materials.
  • label refers to incorporation of a detectable marker, e.g., by incorporation of a radiolabeled amino acid or attachment to a polypeptide of biotinyl moieties that can be detected by marked avidin (e.g., streptavidin containing a fluorescent marker or enzymatic activity that can be detected by optical or calorimetric methods). In certain situations, the label or marker can also be therapeutic. Various methods of labeling polypeptides and glycoproteins are known in the art and may be used.
  • labels for polypeptides include, but are not limited to, the following: radioisotopes or radionuclides (e.g., 3 H, 14 C, 15 N, 35 S, 90 Y, 99 Tc, 111 In, 125 I, 131 I), fluorescent labels (e.g., FITC, rhodamine, lanthanide phosphors), enzymatic labels (e.g., horseradish peroxidase, p-galactosidase, luciferase, alkaline phosphatase), chemiluminescent, biotinyl groups, predetermined polypeptide epitopes recognized by a secondary reporter (e.g., leucine zipper pair sequences, binding sites for secondary antibodies, metal binding domains, epitope tags).
  • radioisotopes or radionuclides e.g., 3 H, 14 C, 15 N, 35 S, 90 Y, 99 Tc, 111 In, 125 I, 131 I
  • labels are attached by spacer arms of various lengths to reduce potential steric hindrance.
  • pharmaceutical agent or drug refers to a chemical compound or composition capable of inducing a desired therapeutic effect when properly administered to a patient.
  • substantially pure means an object species is the predominant species present (i.e., on a molar basis it is more abundant than any other individual species in the composition), and in some embodiments, a substantially purified fraction is a composition wherein the object species comprises at least about 50 percent (on a molar basis) of all macromolecular species present.
  • a substantially pure composition will comprise more than about 80 percent of all macromolecular species present in the composition, in some embodiments, more than about 85%, 90%, 95%, and 99%.
  • the object species is purified to essential homogeneity (contaminant species cannot be detected in the composition by conventional detection methods) wherein the composition consists essentially of a single macromolecular species.
  • patient includes human and veterinary subjects.
  • Activatable antibodies of the disclosure specifically bind a given target, e.g., a human target protein. Also included in the disclosure are activatable antibodies that bind to the same epitope as the activatable antibodies described herein.
  • a monoclonal antibody e.g., a murine monoclonal or humanized antibody
  • a monoclonal antibody has the same specificity as a monoclonal antibody used in the methods described herein by ascertaining whether the former prevents the latter from binding to the target. If the monoclonal antibody being tested competes with the monoclonal antibody of the disclosure, as shown by a decrease in binding by the monoclonal antibody of the disclosure, then the two monoclonal antibodies bind to the same, or a closely related, epitope.
  • a method for determining whether a monoclonal antibody has the specificity of a monoclonal antibody of the disclosure is to pre-incubate the monoclonal antibody of the disclosure with the target and then add the monoclonal antibody being tested to determine if the monoclonal antibody being tested is inhibited in its ability to bind the target. If the monoclonal antibody being tested is inhibited then, in all likelihood, it has the same, or functionally equivalent, epitopic specificity as the monoclonal antibody of the disclosure.
  • the disclosure also provides multispecific activatable antibodies.
  • the multispecific activatable antibodies provided herein are multispecific antibodies that recognize two or more different antigens or epitopes and that include at least one masking moiety (MM) linked to at least one antigen- or epitope-binding domain of the multispecific antibody such that coupling of the MM reduces the ability of the antigen- or epitope-binding domain to bind its target.
  • the MM is coupled to the antigen- or epitope-binding domain of the multispecific antibody via a cleavable moiety (CM) that functions as a substrate for at least one MMP protease.
  • CM cleavable moiety
  • the activatable multispecific antibodies provided herein are stable in circulation, activated at intended sites of therapy and/or diagnosis but not in normal, i.e., healthy tissue, and, when activated, exhibit binding to a target that is at least comparable to the corresponding, unmodified multispecific antibody.
  • the multispecific activatable antibodies are designed to engage immune effector cells, also referred to herein as immune-effector cell engaging multispecific activatable antibodies.
  • the multispecific activatable antibodies are designed to engage leukocytes, also referred to herein as leukocyte engaging multispecific activatable antibodies.
  • the multispecific activatable antibodies are designed to engage T cells, also referred to herein as T-cell engaging multispecific activatable antibodies.
  • the multispecific activatable antibodies engage a surface antigen on a leukocyte, such as on a T cell, on a natural killer (NK) cell, on a myeloid mononuclear cell, on a macrophage, and/or on another immune effector cell.
  • NK natural killer
  • the immune effector cell is a leukocyte. In some embodiments, the immune effector cell is a T cell. In some embodiments, the immune effector cell is a NK cell. In some embodiments, the immune effector cell is a mononuclear cell, such as a myeloid mononuclear cell.
  • the multispecific activatable antibodies are designed to bind or otherwise interact with more than one target and/or more than one epitope, also referred to herein as multi-antigen targeting activatable antibodies. As used herein, the terms “target” and “antigen” are used interchangeably.
  • immune effector cell engaging multispecific activatable antibodies of the disclosure include a targeting antibody or antigen-binding fragment thereof and an immune effector cell engaging antibody or antigen-binding portion thereof, where at least one of the targeting antibody or antigen-binding fragment thereof and/or the immune effector cell engaging antibody or antigen-binding portion thereof is masked.
  • the immune effector cell engaging antibody or antigen binding fragment thereof includes a first antibody or antigen-binding fragment thereof (AB1) that binds a first, immune effector cell engaging target, where the AB1 is attached to a masking moiety (MM1) such that coupling of the MM1 reduces the ability of the AB1 to bind the first target.
  • AB1 first antibody or antigen-binding fragment thereof
  • MM1 masking moiety
  • the targeting antibody or antigen-binding fragment thereof includes a second antibody or fragment thereof that includes a second antibody or antigen-binding fragment thereof (AB2) that binds a second target, where the AB2 is attached to a masking moiety (MM2) such that coupling of the MM2 reduces the ability of the AB2 to bind the second target.
  • AB2 second antibody or antigen-binding fragment thereof
  • MM2 masking moiety
  • the immune effector cell engaging antibody or antigen binding fragment thereof includes a first antibody or antigen-binding fragment thereof (AB1) that binds a first, immune effector cell engaging target, where the AB1 is attached to a masking moiety (MM1) such that coupling of the MM1 reduces the ability of the AB1 to bind the first target, and the targeting antibody or antigen-binding fragment thereof includes a second antibody or fragment thereof that includes a second antibody or antigen-binding fragment thereof (AB2) that binds a second target, where the AB2 is attached to a masking moiety (MM2) such that coupling of the MM2 reduces the ability of the AB2 to bind the second target.
  • AB1 antibody or antigen-binding fragment thereof
  • MM1 masking moiety
  • the non-immune effector cell engaging antibody is a cancer targeting antibody. In some embodiments the non-immune cell effector antibody is an IgG. In some embodiments the immune effector cell engaging antibody is a scFv. In some embodiments the targeting antibody (e.g., non-immune cell effector antibody) is an IgG and the immune effector cell engaging antibody is a scFv. In some embodiments, the immune effector cell is a leukocyte. In some embodiments, the immune effector cell is a T cell. In some embodiments, the immune effector cell is a NK cell. In some embodiments, the immune effector cell is a myeloid mononuclear cell.
  • T-cell engaging multispecific activatable antibodies of the disclosure include a targeting antibody or antigen-binding fragment thereof and a T-cell engaging antibody or antigen-binding portion thereof, where at least one of the targeting antibody or antigen-binding fragment thereof and/or the T-cell engaging antibody or antigen-binding portion thereof is masked.
  • the T-cell engaging antibody or antigen binding fragment thereof includes a first antibody or antigen-binding fragment thereof (AB1) that binds a first, T-cell engaging target, where the AB1 is attached to a masking moiety (MM1) such that coupling of the MM1 reduces the ability of the AB1 to bind the first target.
  • AB1 first antibody or antigen-binding fragment thereof
  • MM1 masking moiety
  • the targeting antibody or antigen-binding fragment thereof includes a second antibody or fragment thereof that includes a second antibody or antigen-binding fragment thereof (AB2) that binds a second target, where the AB2 is attached to a masking moiety (MM2) such that coupling of the MM2 reduces the ability of the AB2 to bind the second target.
  • AB2 second antibody or antigen-binding fragment thereof
  • MM2 masking moiety
  • the T-cell engaging antibody or antigen binding fragment thereof includes a first antibody or antigen-binding fragment thereof (AB1) that binds a first, T-cell engaging target, where the AB1 is attached to a masking moiety (MM1) such that coupling of the MM1 reduces the ability of the AB1 to bind the first target, and the targeting antibody or antigen-binding fragment thereof includes a second antibody or fragment thereof that includes a second antibody or antigen-binding fragment thereof (AB2) that binds a second target, where the AB2 is attached to a masking moiety (MM2) such that coupling of the MM2 reduces the ability of the AB2 to bind the second target.
  • AB1 antibody or antigen-binding fragment thereof
  • MM1 masking moiety
  • the T-cell engaging multispecific activatable antibodies include a cancer targeting antibody or antigen-binding fragment thereof and a T-cell engaging antibody or antigen-binding portion thereof, where at least one of the cancer targeting antibody or antigen-binding fragment thereof and/or the T-cell engaging antibody or antigen-binding portion thereof is masked.
  • the T-cell engaging antibody or antigen binding fragment thereof includes a first antibody or antigen-binding fragment thereof (AB1) that binds a first, T-cell engaging target, where the AB1 is attached to a masking moiety (MM1) such that coupling of the MM1 reduces the ability of the AB1 to bind the first target.
  • AB1 first antibody or antigen-binding fragment thereof
  • MM1 masking moiety
  • the cancer targeting antibody or antigen-binding fragment thereof includes a second antibody or fragment thereof that includes a second antibody or antigen-binding fragment thereof (AB2) that binds a second, cancer-related target, where the AB2 is attached to a masking moiety (MM2) such that coupling of the MM2 reduces the ability of the AB2 to bind the second, cancer-related target.
  • AB2 second antibody or antigen-binding fragment thereof
  • MM2 masking moiety
  • the T-cell engaging antibody or antigen binding fragment thereof includes a first antibody or antigen-binding fragment thereof (AB1) that binds a first, T-cell engaging target, where the AB1 is attached to a masking moiety (MM1) such that coupling of the MM1 reduces the ability of the AB1 to bind the first target, and the cancer targeting antibody or antigen-binding fragment thereof includes a second antibody or fragment thereof that includes a second antibody or antigen-binding fragment thereof (AB2) that binds a second, cancer-related target, where the AB2 is attached to a masking moiety (MM2) such that coupling of the MM2 reduces the ability of the AB2 to bind the second, cancer-related target.
  • AB1 antibody or antigen-binding fragment thereof
  • MM1 masking moiety
  • the T-cell engaging multispecific activatable antibodies include a cancer targeting IgG antibody or antigen-binding fragment thereof and a T-cell engaging scFv, where at least one of the cancer targeting IgG antibody or antigen-binding fragment thereof and/or the T-cell engaging antibody or antigen-binding portion thereof is masked.
  • the T-cell engaging antibody or antigen binding fragment thereof includes a first antibody or antigen-binding fragment thereof (AB1) that binds a first, T-cell engaging target, where the AB1 is attached to a masking moiety (MM1) such that coupling of the MM1 reduces the ability of the AB1 to bind the first target.
  • the cancer targeting IgG antibody or antigen-binding fragment thereof includes a second antibody or fragment thereof that includes a second antibody or antigen-binding fragment thereof (AB2) that binds a second, cancer-related target, where the AB2 is attached to a masking moiety (MM2) such that coupling of the MM2 reduces the ability of the AB2 to bind the second, cancer-related target.
  • AB2 second antibody or antigen-binding fragment thereof
  • MM2 masking moiety
  • the T-cell engaging antibody or antigen binding fragment thereof includes a first antibody or antigen-binding fragment thereof (AB1) that binds a first, T-cell engaging target, where the AB1 is attached to a masking moiety (MM1) such that coupling of the MM1 reduces the ability of the AB1 to bind the first target, and the cancer targeting IgG antibody or antigen-binding fragment thereof includes a second antibody or fragment thereof that includes a second antibody or antigen-binding fragment thereof (AB2) that binds a second, cancer-related target, where the AB2 is attached to a masking moiety (MM2) such that coupling of the MM2 reduces the ability of the AB2 to bind the second, cancer-related target.
  • AB1 first antibody or antigen-binding fragment thereof
  • MM1 masking moiety
  • one antigen is typically an antigen present on the surface of a tumor cell or other cell type associated with disease, such as, but not limited to, any target listed in Table 1, such as, but not limited to, EGFR, erbB2, EpCAM, Jagged, PD-L1, B7H3, or CD71 (transferrin receptor), and another antigen is typically a stimulatory or inhibitory receptor present on the surface of a T-cell, natural killer (NK) cell, myeloid mononuclear cell, macrophage, and/or other immune effector cell, such as, but not limited to, B7-H4, BTLA, CD3, CD4, CD8, CD16a, CD25, CD27, CD28, CD32, CD56, CD137, CTLA-4, GITR, HVEM, ICOS, LAG3, NKG2D, OX40, PD-1, TIGIT, TIM3, or VISTA.
  • a tumor cell or other cell type associated with disease such as, but not limited to, any target listed in Table 1, such as
  • the antigen is a stimulatory receptor present on the surface of a T cell or NK cell; examples of such stimulatory receptors include, but are not limited to, CD3, CD27, CD28, CD137 (also referred to as 4-1BB), GITR, HVEM, ICOS, NKG2D, and OX40.
  • the antigen is an inhibitory receptor present on the surface of a T-cell; examples of such inhibitory receptors include, but are not limited to, BTLA, CTLA-4, LAG3, PD-1, TIGIT, TIM3, and NK-expressed KIRs.
  • the antibody domain conferring specificity to the T-cell surface antigen may also be substituted by a ligand or ligand domain that binds to a T-cell receptor, a NK-cell receptor, a macrophage receptor, and/or other immune effector cell receptor, such as, but not limited to, B7-1, B7-2, B7H3, PD-L1, PD-L2, or TNFSF9.
  • a ligand or ligand domain that binds to a T-cell receptor, a NK-cell receptor, a macrophage receptor, and/or other immune effector cell receptor, such as, but not limited to, B7-1, B7-2, B7H3, PD-L1, PD-L2, or TNFSF9.
  • One embodiment of the disclosure is a multispecific activatable antibody that is activatable in a cancer microenvironment and that includes an antibody, for example a IgG or scFv, directed to a tumor target and an agonist antibody, for example an IgG or scFv, directed to a co-stimulatory receptor expressed on the surface of an activated T cell or NK cell, wherein at least one of the cancer target antibody and/or agonist antibody is masked.
  • co-stimulatory receptors include, but are not limited to, CD27, CD137, GITR, HVEM, NKG2D, and OX40.
  • the multispecific activatable antibody once activated by tumor-associated proteases, would effectively crosslink and activate the T cell or NK cell expressed co-stimulatory receptors in a tumor-dependent manner to enhance the activity of T cells that are responding to any tumor antigen via their endogenous T cell antigen or NK-activating receptors.
  • the activation-dependent nature of these T cell or NK cell costimulatory receptors would focus the activity of the activated multispecific activatable antibody to tumor-specific T cells, without activating all T cells independent of their antigen specificity.
  • At least the co-stimulatory receptor antibody of the multispecific activatable antibody is masked to prevent activation of auto-reactive T cells that may be present in tissues that also express the antigen recognized by the tumor target-directed antibody in the multispecific activatable antibody, but whose activity is restricted by lack of co-receptor engagement.
  • One embodiment of the disclosure is a multispecific activatable antibody that is activatable in a disease characterized by T cell overstimulation, such as, but not limited to, an autoimmune disease or inflammatory disease microenvironment.
  • a multispecific activatable antibody includes an antibody, for example a IgG or scFv, directed to a target comprising a surface antigen expressed in a tissue targeted by a T cell in autoimmune or inflammatory disease and an antibody, for example a IgG or scFv, directed to an inhibitory receptor expressed on the surface of a T cell or NK cell, wherein at least one of the disease tissue target antibody and/or T cell inhibitory receptor antibody is masked.
  • inhibitory receptors include, but are not limited to, BTLA, CTLA-4, LAG3, PD-1, TIGIT, TIM3, and NK-expressed KIRs.
  • tissue antigen targeted by T cells in autoimmune disease include, but are not limited to, a surface antigen expressed on myelin or nerve cells in multiple sclerosis or a surface antigen expressed on pancreatic islet cells in Type 1 diabetes.
  • the multispecific activatable antibody when localized in the tissue under autoimmune attack or inflammation is activated and co-engages the T cell or NK cell inhibitory receptor to suppress the activity of autoreactive T cells responding to any disease tissue-targeted antigens via their endogenous TCR or activating receptors.
  • at least one or multiple antibodies are masked to prevent suppression of T cell responses in non-disease tissues where the target antigen may also be expressed.
  • the T-cell engaging multispecific activatable antibody includes an anti-CD3 epsilon (CD3 ⁇ , also referred to herein as CD3e and CD3) scFv and a targeting antibody or antigen-binding fragment thereof, where at least one of the anti-CD3 ⁇ scFv and/or the targeting antibody or antigen-binding portion thereof is masked.
  • the CD3 ⁇ scFv includes a first antibody or antigen-binding fragment thereof (AB1) that binds CD3 ⁇ , where the AB1 is attached to a masking moiety (MM1) such that coupling of the MM1 reduces the ability of the AB1 to bind CD3 ⁇ .
  • the targeting antibody or antigen-binding fragment thereof includes a second antibody or fragment thereof that includes a second antibody or antigen-binding fragment thereof (AB2) that binds a second target, where the AB2 is attached to a masking moiety (MM2) such that coupling of the MM2 reduces the ability of the AB2 to bind the second target.
  • AB2 second antibody or antigen-binding fragment thereof
  • MM2 masking moiety
  • the CD3 ⁇ scFv includes a first antibody or antigen-binding fragment thereof (AB1) that binds CD3 ⁇ , where the AB1 is attached to a masking moiety (MM1) such that coupling of the MM1 reduces the ability of the AB1 to bind CD3 ⁇ , and the targeting antibody or antigen-binding fragment thereof includes a second antibody or fragment thereof that includes a second antibody or antigen-binding fragment thereof (AB2) that binds a second target, where the AB2 is attached to a masking moiety (MM2) such that coupling of the MM2 reduces the ability of the AB2 to bind the second target.
  • AB1 antibody or antigen-binding fragment thereof
  • MM1 masking moiety
  • the T-cell engaging multispecific activatable antibody includes an anti-CD3 ⁇ scFv and a cancer targeting antibody or antigen-binding fragment thereof, where at least one of the anti-CD3 ⁇ scFv and/or the cancer targeting antibody or antigen-binding portion thereof is masked.
  • the CD3 ⁇ scFv includes a first antibody or antigen-binding fragment thereof (AB1) that binds CD3 ⁇ , where the AB1 is attached to a masking moiety (MM1) such that coupling of the MM1 reduces the ability of the AB1 to bind CD3 ⁇ .
  • the cancer targeting antibody or antigen-binding fragment thereof includes a second antibody or fragment thereof that includes a second antibody or antigen-binding fragment thereof (AB2) that binds a second, cancer-related target, where the AB2 is attached to a masking moiety (MM2) such that coupling of the MM2 reduces the ability of the AB2 to bind the second, cancer-related target.
  • AB2 second antibody or antigen-binding fragment thereof
  • MM2 masking moiety
  • the CD3 ⁇ scFv includes a first antibody or antigen-binding fragment thereof (AB1) that binds CD3 ⁇ , where the AB1 is attached to a masking moiety (MM1) such that coupling of the MM1 reduces the ability of the AB1 to bind CD3 ⁇ , and the cancer targeting antibody or antigen-binding fragment thereof includes a second antibody or fragment thereof that includes a second antibody or antigen-binding fragment thereof (AB2) that binds a second, cancer-related target, where the AB2 is attached to a masking moiety (MM2) such that coupling of the MM2 reduces the ability of the AB2 to bind the second, cancer-related target.
  • AB1 antibody or antigen-binding fragment thereof
  • MM1 masking moiety
  • the T-cell engaging multispecific activatable antibody includes an anti-CD3 ⁇ scFv and a cancer targeting IgG antibody or antigen-binding fragment thereof, where at least one of the anti-CD3 ⁇ scFv and/or the cancer targeting IgG antibody or antigen-binding portion thereof is masked.
  • the CD3 ⁇ scFv includes a first antibody or antigen-binding fragment thereof (AB1) that binds CD3 ⁇ , where the AB1 is attached to a masking moiety (MM1) such that coupling of the MM1 reduces the ability of the AB1 to bind CD3 ⁇ .
  • the cancer targeting IgG antibody or antigen-binding fragment thereof includes a second antibody or fragment thereof that includes a second antibody or antigen-binding fragment thereof (AB2) that binds a second, cancer-related target, where the AB2 is attached to a masking moiety (MM2) such that coupling of the MM2 reduces the ability of the AB2 to bind the second, cancer-related target.
  • AB2 second antibody or antigen-binding fragment thereof
  • MM2 masking moiety
  • the CD3 ⁇ scFv includes a first antibody or antigen-binding fragment thereof (AB1) that binds CD3 ⁇ , where the AB1 is attached to a masking moiety (MM1) such that coupling of the MM1 reduces the ability of the AB1 to bind CD3 ⁇ , and the cancer targeting antibody IgG or antigen-binding fragment thereof includes a second antibody or fragment thereof that includes a second antibody or antigen-binding fragment thereof (AB2) that binds a second, cancer-related target, where the AB2 is attached to a masking moiety (MM2) such that coupling of the MM2 reduces the ability of the AB2 to bind the second, cancer-related target.
  • AB1 first antibody or antigen-binding fragment thereof
  • MM1 masking moiety
  • the T-cell engaging multispecific activatable antibody includes an anti-CD3 epsilon (CD3 ⁇ ) scFv that is derived from OKT3, where at least one of the targeting antibody or antigen-binding fragment thereof and/or the OKT3 scFv or OKT3-derived scFv is masked.
  • the OKT3 scFv or OKT3-derived scFv includes a first antibody or antigen-binding fragment thereof (AB1) that binds CD3 ⁇ , where the AB1 is attached to a masking moiety (MM1) such that coupling of the MM1 reduces the ability of the AB1 to bind CD3 ⁇ .
  • the targeting antibody or antigen-binding fragment thereof includes a second antibody or fragment thereof that includes a second antibody or antigen-binding fragment thereof (AB2) that binds a second target, where the AB2 is attached to a masking moiety (MM2) such that coupling of the MM2 reduces the ability of the AB2 to bind the second target.
  • AB2 second antibody or antigen-binding fragment thereof
  • MM2 masking moiety
  • the OKT3 scFv or OKT3-derived scFv includes a first antibody or antigen-binding fragment thereof (AB1) that binds CD3 ⁇ , where the AB1 is attached to a masking moiety (MM1) such that coupling of the MM1 reduces the ability of the AB1 to bind CD3 ⁇ , and the targeting antibody or antigen-binding fragment thereof includes a second antibody or fragment thereof that includes a second antibody or antigen-binding fragment thereof (AB2) that binds a second target, where the AB2 is attached to a masking moiety (MM2) such that coupling of the MM2 reduces the ability of the AB2 to bind the second target.
  • AB1 antibody or antigen-binding fragment thereof
  • MM1 masking moiety
  • the T-cell engaging multispecific activatable antibody includes an OKT3 scFv or OKT3-derived scFv and a cancer targeting antibody or antigen-binding fragment thereof, where at least one of the OKT3 scFv or OKT3-derived scFv and/or the cancer targeting antibody or antigen-binding portion thereof is masked.
  • the OKT3 scFv or OKT3-derived scFv includes a first antibody or antigen-binding fragment thereof (AB1) that binds CD3 ⁇ , where the AB1 is attached to a masking moiety (MM1) such that coupling of the MM1 reduces the ability of the AB1 to bind CD3 ⁇ .
  • the cancer targeting antibody or antigen-binding fragment thereof includes a second antibody or fragment thereof that includes a second antibody or antigen-binding fragment thereof (AB2) that binds a second, cancer-related target, where the AB2 is attached to a masking moiety (MM2) such that coupling of the MM2 reduces the ability of the AB2 to bind the second, cancer-related target.
  • AB2 second antibody or antigen-binding fragment thereof
  • MM2 masking moiety
  • the OKT3 scFv or OKT3-derived scFv includes a first antibody or antigen-binding fragment thereof (AB1) that binds CD3 ⁇ , where the AB1 is attached to a masking moiety (MM1) such that coupling of the MM1 reduces the ability of the AB1 to bind CD3 ⁇ , and the cancer targeting antibody or antigen-binding fragment thereof includes a second antibody or fragment thereof that includes a second antibody or antigen-binding fragment thereof (AB2) that binds a second, cancer-related target, where the AB2 is attached to a masking moiety (MM2) such that coupling of the MM2 reduces the ability of the AB2 to bind the second, cancer-related target.
  • AB1 antibody or antigen-binding fragment thereof
  • MM1 masking moiety
  • the T-cell engaging multispecific activatable antibody includes an OKT3 scFv or OKT3-derived scFv and a cancer targeting IgG antibody or antigen-binding fragment thereof, where at least one of the OKT3 scFv or OKT3-derived scFv and/or the cancer targeting IgG antibody or antigen-binding portion thereof is masked.
  • the OKT3 scFv or OKT3-derived scFv includes a first antibody or antigen-binding fragment thereof (AB1) that binds CD3 ⁇ , where the AB1 is attached to a masking moiety (MM1) such that coupling of the MM1 reduces the ability of the AB1 to bind CD3 ⁇ .
  • AB1 first antibody or antigen-binding fragment thereof
  • MM1 masking moiety
  • the cancer targeting IgG antibody or antigen-binding fragment thereof includes a second antibody or fragment thereof that includes a second antibody or antigen-binding fragment thereof (AB2) that binds a second, cancer-related target, where the AB2 is attached to a masking moiety (MM2) such that coupling of the MM2 reduces the ability of the AB2 to bind the second, cancer-related target.
  • AB2 second antibody or antigen-binding fragment thereof
  • MM2 masking moiety
  • the OKT3 scFv or OKT3-derived scFv includes a first antibody or antigen-binding fragment thereof (AB1) that binds CD3 ⁇ , where the AB1 is attached to a masking moiety (MM1) such that coupling of the MM1 reduces the ability of the AB1 to bind CD3 ⁇ , and the cancer targeting antibody IgG or antigen-binding fragment thereof includes a second antibody or fragment thereof that includes a second antibody or antigen-binding fragment thereof (AB2) that binds a second, cancer-related target, where the AB2 is attached to a masking moiety (MM2) such that coupling of the MM2 reduces the ability of the AB2 to bind the second, cancer-related target.
  • AB1 antibody or antigen-binding fragment thereof
  • MM1 masking moiety
  • the T-cell engaging multispecific activatable antibody includes an anti-CTLA-4 scFv, where at least one of the targeting antibody or antigen-binding fragment thereof and/or the anti-CTLA-4 scFv is masked.
  • the anti-CTLA-4 scFv includes a first antibody or antigen-binding fragment thereof (AB1) that binds CTLA-4, where the AB1 is attached to a masking moiety (MM1) such that coupling of the MM1 reduces the ability of the AB1 to bind CTLA-4.
  • the targeting antibody or antigen-binding fragment thereof includes a second antibody or fragment thereof that includes a second antibody or antigen-binding fragment thereof (AB2) that binds a second target, where the AB2 is attached to a masking moiety (MM2) such that coupling of the MM2 reduces the ability of the AB2 to bind the second target.
  • AB2 second antibody or antigen-binding fragment thereof
  • MM2 masking moiety
  • the anti-CTLA-4 scFv includes a first antibody or antigen-binding fragment thereof (AB1) that binds CTLA-4, where the AB1 is attached to a masking moiety (MM1) such that coupling of the MM1 reduces the ability of the AB1 to bind CTLA-4, and the targeting antibody or antigen-binding fragment thereof includes a second antibody or fragment thereof that includes a second antibody or antigen-binding fragment thereof (AB2) that binds a second target, where the AB2 is attached to a masking moiety (MM2) such that coupling of the MM2 reduces the ability of the AB2 to bind the second target.
  • AB1 antibody or antigen-binding fragment thereof
  • MM1 masking moiety
  • the T-cell engaging multispecific activatable antibody includes an anti-CTLA-4 scFv and a targeting IgG antibody or antigen-binding fragment thereof, where at least one of the anti-CTLA-4 scFv and/or the targeting IgG antibody or antigen-binding portion thereof is masked.
  • the anti-CTLA-4 scFv includes a first antibody or antigen-binding fragment thereof (AB1) that binds CTLA-4, where the AB1 is attached to a masking moiety (MM1) such that coupling of the MM1 reduces the ability of the AB1 to bind CTLA-4.
  • the targeting IgG antibody or antigen-binding fragment thereof includes a second antibody or fragment thereof that includes a second antibody or antigen-binding fragment thereof (AB2) that binds a second target, where the AB2 is attached to a masking moiety (MM2) such that coupling of the MM2 reduces the ability of the AB2 to bind the second target.
  • AB2 second antibody or antigen-binding fragment thereof
  • MM2 masking moiety
  • the anti-CTLA-4 scFv includes a first antibody or antigen-binding fragment thereof (AB1) that binds CTLA-4, where the AB1 is attached to a masking moiety (MM1) such that coupling of the MM1 reduces the ability of the AB1 to bind CTLA-4, and the targeting antibody IgG or antigen-binding fragment thereof includes a second antibody or fragment thereof that includes a second antibody or antigen-binding fragment thereof (AB2) that binds a second target, where the AB2 is attached to a masking moiety (MM2) such that coupling of the MM2 reduces the ability of the AB2 to bind the second target.
  • AB1 antibody or antigen-binding fragment thereof
  • MM1 masking moiety
  • the multi-antigen targeting antibodies and/or multi-antigen targeting activatable antibodies include at least a first antibody or antigen-binding fragment thereof that binds a first target and/or first epitope and a second antibody or antigen-binding fragment thereof that binds a second target and/or a second epitope. In some embodiments, the multi-antigen targeting antibodies and/or multi-antigen targeting activatable antibodies bind two or more different targets. In some embodiments, the multi-antigen targeting antibodies and/or multi-antigen targeting activatable antibodies bind two or more different epitopes on the same target. In some embodiments, the multi-antigen targeting antibodies and/or multi-antigen targeting activatable antibodies bind a combination of two or more different targets and two or more different epitopes on the same target.
  • a multispecific activatable antibody comprising an IgG has the IgG variable domains masked. In some embodiments, a multispecific activatable antibody comprising a scFv has the scFv domains masked. In some embodiments, a multispecific activatable antibody has both IgG variable domains and scFv domains, where at least one of the IgG variable domains is coupled to a masking moiety. In some embodiments, a multispecific activatable antibody has both IgG variable domains and scFv domains, where at least one of the scFv domains is coupled to a masking moiety.
  • a multispecific activatable antibody has both IgG variable domains and scFv domains, where at least one of the IgG variable domains is coupled to a masking moiety and at least one of the scFv domains is coupled to a masking moiety. In some embodiments, a multispecific activatable antibody has both IgG variable domains and scFv domains, where each of the IgG variable domains and the scFv domains is coupled to its own masking moiety. In some embodiments, one antibody domain of a multispecific activatable antibody has specificity for a target antigen and another antibody domain has specificity for a T-cell surface antigen.
  • one antibody domain of a multispecific activatable antibody has specificity for a target antigen and another antibody domain has specificity for another target antigen. In some embodiments, one antibody domain of a multispecific activatable antibody has specificity for an epitope of a target antigen and another antibody domain has specificity for another epitope of the target antigen.
  • a scFv in a multispecific activatable antibody, can be fused to the carboxyl terminus of the heavy chain of an IgG activatable antibody, to the carboxyl terminus of the light chain of an IgG activatable antibody, or to the carboxyl termini of both the heavy and light chains of an IgG activatable antibody.
  • a scFv in a multispecific activatable antibody, can be fused to the amino terminus of the heavy chain of an IgG activatable antibody, to the amino terminus of the light chain of an IgG activatable antibody, or to the amino termini of both the heavy and light chains of an IgG activatable antibody.
  • a scFv can be fused to any combination of one or more carboxyl termini and one or more amino termini of an IgG activatable antibody.
  • a masking moiety (MM) linked to a cleavable moiety (CM) is attached to and masks an antigen binding domain of the IgG.
  • a masking moiety (MM) linked to a cleavable moiety (CM) is attached to and masks an antigen binding domain of at least one scFv.
  • a masking moiety (MM) linked to a cleavable moiety (CM) is attached to and masks an antigen binding domain of an IgG and a masking moiety (MM) linked to a cleavable moiety (CM) is attached to and masks an antigen binding domain of at least one scFv.
  • the disclosure provides examples of multispecific activatable antibody structures which include, but are not limited to, the following: (VL-CL) 2 :(VH-CH1-CH2-CH3-L4-VH*-L3-VL*-L2-CM-L1-MM) 2 ; (VL-CL) 2 :(VH-CH1-CH2-CH3-L4-VL*-L3-VH*-L2-CM-L1-MM) 2 ; (MM-L1-CM-L2-VL-CL) 2 : (VH-CH1-CH2-CH3-L4-VH*-L3-VL*) 2 ; (MM-L1-CM-L2-VL-CL) 2 :(VH-CH1-CH2-CH3-L4-VL*-L3-VH*) 2 ; (VL-CL) 2 :(MM-L1-CM-L2-VL*-L3-VH*) 2 ; (VL-CL) 2 :
  • one antigen is typically an antigen present on the surface of a tumor cell or other cell type associated with disease, such as, but not limited to, any target listed in Table 1, such as, but not limited to, EGFR, erbB2, EpCAM, Jagged, PD-L1, B7H3, or CD71 (transferrin receptor), and another antigen is typically a stimulatory (also referred to herein as activating) or inhibitory receptor present on the surface of a T-cell, natural killer (NK) cell, myeloid mononuclear cell, macrophage, and/or other immune effector cell, such as, but not limited to, B7-H4, BTLA, CD3, CD4, CD8, CD16a, CD25, CD27, CD28, CD32, CD56, CD137 (also referred to as TNFRSF9), CTLA-4, GITR, HVEM, ICOS, LAG3, NKG2D, OX40, PD-1, TIG
  • the antibody domain conferring specificity to the T-cell surface antigen may also be substituted by a ligand or ligand domain that binds to a T-cell receptor, a NK-cell receptor, a macrophage receptor, and/or other immune effector cell receptor, such as, but not limited to, B7-1, B7-2, B7H3, PD-L1, PD-L2, or TNFSF9.
  • a ligand or ligand domain that binds to a T-cell receptor, a NK-cell receptor, a macrophage receptor, and/or other immune effector cell receptor, such as, but not limited to, B7-1, B7-2, B7H3, PD-L1, PD-L2, or TNFSF9.
  • one antigen is selected from the group of targets listed in Table 1
  • another antigen is selected from the group of targets listed in Table 1.
  • the targeting antibody is an anti-EGFR antibody. In some embodiments, the targeting antibody is C225v5, which is specific for binding to EGFR. In some embodiments, the targeting antibody is C225, which is specific for binding to EGFR. In some embodiments, the targeting antibody is C225v4, which is specific for binding to EGFR. In some embodiments, the targeting antibody is C225v6, which is specific for binding to EGFR. In some embodiments, the targeting antibody is an anti-Jagged antibody. In some embodiments, the targeting antibody is 4D11, which is specific for binding to human and mouse Jagged 1 and Jagged 2. In some embodiments, the targeting antibody is 4D11v2, which is specific for binding to human and mouse Jagged 1 and Jagged 2.
  • the targeting antibody can be in the form an activatable antibody.
  • the scFv(s) can be in the form of a Pro-scFv (see, e.g., WO 2009/025846, WO 2010/081173).
  • the scFv is specific for binding CD3 ⁇ , and is or is derived from an antibody or fragment thereof that binds CD3 ⁇ , e.g., CH2527, FN18, H2C, OKT3, 2C11, UCHT1, or V9.
  • the scFv is specific for binding CTLA-4 (also referred to herein as CTLA and CTLA4).
  • the anti-CTLA-4 scFv includes the amino acid sequence:
  • the anti-CTLA-4 scFv includes the amino acid sequence that is at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more identical to the amino acid sequence of SEQ ID NO: 510.
  • the anti-CD3 ⁇ scFv includes the amino acid sequence:
  • the anti-CD3 ⁇ scFv includes the amino acid sequence that is at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more identical to the amino acid sequence of SEQ ID NO: 511.
  • the scFv is specific for binding one or more T-cells, one or more NK-cells and/or one or more macrophages. In some embodiments, the scFv is specific for binding a target selected from the group consisting of B7-H4, BTLA, CD3, CD4, CD8, CD16a, CD25, CD27, CD28, CD32, CD56, CD137, CTLA-4, GITR, HVEM, ICOS, LAG3, NKG2D, OX40, PD-1, TIGIT, TIM3, or VISTA.
  • the multispecific activatable antibody also includes an agent conjugated to the AB.
  • the agent is a therapeutic agent.
  • the agent is an antineoplastic agent.
  • the agent is a toxin or fragment thereof.
  • the agent is conjugated to the multispecific activatable antibody via a linker.
  • the agent is conjugated to the AB via a cleavable linker.
  • the agent is conjugated to the AB via a linker that includes at least one MMP-cleavable substrate sequence.
  • the linker is a non-cleavable linker.
  • the agent is a microtubule inhibitor.
  • the agent is a nucleic acid damaging agent, such as a DNA alkylator or DNA intercalator, or other DNA damaging agent.
  • the linker is a cleavable linker.
  • the agent is an agent selected from the group listed in Table 4.
  • the agent is a dolastatin.
  • the agent is an auristatin or derivative thereof.
  • the agent is auristatin E or a derivative thereof.
  • the agent is monomethyl auristatin E (MMAE).
  • the agent is monomethyl auristatin D (MMAD).
  • the agent is a maytansinoid or maytansinoid derivative. In some embodiments, the agent is DM1 or DM4. In some embodiments, the agent is a duocarmycin or derivative thereof. In some embodiments, the agent is a calicheamicin or derivative thereof. In some embodiments, the agent is a pyrrolobenzodiazepine.
  • the multispecific activatable antibody also includes a detectable moiety.
  • the detectable moiety is a diagnostic agent.
  • the multispecific activatable antibody naturally contains one or more disulfide bonds. In some embodiments, the multispecific activatable antibody can be engineered to include one or more disulfide bonds.
  • the disclosure also provides an isolated nucleic acid molecule encoding a multispecific activatable antibody described herein, as well as vectors that include these isolated nucleic acid sequences.
  • the disclosure provides methods of producing a multispecific activatable antibody by culturing a cell under conditions that lead to expression of the activatable antibody, wherein the cell comprises such a nucleic acid molecule.
  • the cell comprises such a vector.
  • the disclosure also provides a method of manufacturing multispecific activatable antibodies of the disclosure by (a) culturing a cell comprising a nucleic acid construct that encodes the multispecific activatable antibody under conditions that lead to expression of the multispecific activatable, and (b) recovering the multispecific activatable antibody.
  • the disclosure also provides multispecific activatable antibodies and/or multispecific activatable antibody compositions that include at least a first antibody or antigen-binding fragment thereof (AB1) that specifically binds a first target or first epitope and a second antibody or antigen-biding fragment thereof (AB2) that binds a second target or a second epitope, where at least AB1 is coupled or otherwise attached to a masking moiety (MM1), such that coupling of the MM1 reduces the ability of AB1 to bind its target.
  • AB1 antibody or antigen-binding fragment thereof
  • AB2 second antibody or antigen-biding fragment thereof
  • the MM1 is coupled to AB1 via a first cleavable moiety (CM1) sequence that includes a substrate for a protease, for example, a protease that is co-localized with the target of AB1 at a treatment site or a diagnostic site in a subject.
  • CM1 first cleavable moiety
  • the multispecific activatable antibodies provided herein are stable in circulation, activated at intended sites of therapy and/or diagnosis but not in normal, i.e., healthy tissue, and, when activated, exhibit binding to the target of AB1 that is at least comparable to the corresponding, unmodified multispecific antibody.
  • the multispecific activatable antibody comprises a linking peptide between the MM1 and the CM1.
  • the multispecific activatable antibody comprises a linking peptide between the CM1 and the AB1.
  • the activatable antibody comprises a first linking peptide (LP1) and a second linking peptide (LP2), and at least a portion of the multispecific activatable antibody has the structural arrangement from N-terminus to C-terminus as follows in the uncleaved state: MM1-LP1-CM1-LP2-AB1 or AB1-LP2-CM1-LP1-MM1.
  • the two linking peptides need not be identical to each other.
  • At least one of LP1 or LP2 includes an amino acid sequence selected from the group consisting of (GS) n , (GGS) n , (GSGGS) n (SEQ ID NO: 1) and (GGGS) n (SEQ ID NO: 2), where n is an integer of at least one.
  • at least one of LP1 or LP2 includes an amino acid sequence selected from the group consisting of GGSG (SEQ ID NO: 3), GGSGG (SEQ ID NO: 4), GSGSG (SEQ ID NO: 5), GSGGG (SEQ ID NO: 6), GGGSG (SEQ ID NO: 7), and GSSSG (SEQ ID NO: 8).
  • the multispecific activatable antibody includes at least a first antibody or antigen-binding fragment thereof (AB1) that specifically binds a first target or first epitope and a second antibody or antigen-binding fragment thereof (AB2) that specifically binds a second target or second epitope.
  • each of the AB in the multispecific activatable antibody is independently selected from the group consisting of a monoclonal antibody, domain antibody, single chain, Fab fragment, a F(ab′) 2 fragment, a scFv, a scAb, a dAb, a single domain heavy chain antibody, and a single domain light chain antibody.
  • each of the AB in the multispecific activatable antibody is a rodent (e.g., mouse or rat), chimeric, humanized or fully human monoclonal antibody.
  • each of the AB in the multispecific activatable antibody has an equilibrium dissociation constant of about 100 nM or less for binding to its corresponding target or epitope.
  • MM1 has an equilibrium dissociation constant for binding to its corresponding AB that is greater than the equilibrium dissociation constant of the AB to its corresponding target or epitope.
  • MM1 has an equilibrium dissociation constant for binding to its corresponding AB that is no more than the equilibrium dissociation constant of the AB to its corresponding target or epitope.
  • MM1 does not interfere or compete with its corresponding AB for binding to the corresponding target or epitope when the multispecific activatable antibody is in a cleaved state.
  • MM1 is a polypeptide of about 2 to 40 amino acids in length. In some embodiments, each of the MM in the multispecific activatable antibody is a polypeptide of no more than 40 amino acids in length.
  • MM1 has a polypeptide sequence that is different from that of target of the corresponding AB.
  • MM1 has a polypeptide sequence that is no more than 50% identical to any natural binding partner of the corresponding AB. In some embodiments, MM1 has a polypeptide sequence that is no more than 25% identical to any natural binding partner of the corresponding AB. In some embodiments, MM1 has a polypeptide sequence that is no more than 10% identical to any natural binding partner of the corresponding AB.
  • the coupling of MM1 reduces the ability of the corresponding AB to bind its target or epitope such that the dissociation constant (K d ) of the AB when coupled to the MM1 towards its corresponding target or epitope is at least 20 times greater than the K d of the AB when not coupled to the MM1 towards its corresponding target or epitope.
  • K d dissociation constant
  • the coupling of MM1 reduces the ability of the corresponding AB to bind its target or epitope such that the dissociation constant (K d ) of the AB when coupled to the MM1 towards its corresponding target or epitope is at least 40 times greater than the K d of the AB when not coupled to the MM1 towards its corresponding target or epitope.
  • K d dissociation constant
  • the coupling of MM1 reduces the ability of the corresponding AB to bind its target or epitope such that the dissociation constant (K d ) of the AB when coupled to the MM1 towards its corresponding target or epitope is at least 100 times greater than the K d of the AB when not coupled to the MM1 towards its corresponding target or epitope.
  • K d dissociation constant
  • the coupling of MM1 reduces the ability of the corresponding AB to bind its target or epitope such that the dissociation constant (K d ) of the AB when coupled to the MM1 towards its corresponding target or epitope is at least 1000 times greater than the K d of the AB when not coupled to the MM1 towards its corresponding target or epitope.
  • the coupling of MM1 reduces the ability of the corresponding AB to bind its target or epitope such that the dissociation constant (K d ) of the AB when coupled to the MM1 towards its corresponding target or epitope is at least 10,000 times greater than the K d of the AB when not coupled to the MM1 towards its corresponding target or epitope.
  • K d dissociation constant
  • MM1 is an amino acid sequence selected from a MM disclosed herein.
  • the multispecific activatable antibody includes at least a second masking moiety (MM2) that inhibits the binding of the AB2 to its target when the multispecific activatable antibody is in an uncleaved state, and a second cleavable moiety (CM2) coupled to the AB2, wherein the CM2 is a polypeptide that functions as a substrate for a second protease.
  • CM2 is a polypeptide of no more than 15 amino acids long.
  • the second protease is co-localized with the second target or epitope in a tissue, and wherein the second protease cleaves the CM2 in the multispecific activatable antibody when the multispecific activatable antibody is exposed to the second protease.
  • the first protease and the second protease are co-localized with the first target or epitope and the second target or epitope in a tissue.
  • the first protease and the second protease are the same protease.
  • CM1 and CM2 are different substrates for the same protease.
  • the protease is selected from the group consisting of those shown in Table 7.
  • the first protease and the second protease are different proteases.
  • the first protease and the second protease are different proteases selected from the group consisting of those shown in Table 7.
  • each of the MM in the multispecific activatable antibody e.g., MM1 and at least MM2
  • each of the MM in the multispecific activatable antibody has an equilibrium dissociation constant for binding to its corresponding AB that is no more than the equilibrium dissociation constant of the AB to its corresponding target or epitope.
  • each of the MM in the multispecific activatable antibody does not interfere or compete with its corresponding AB for binding to the corresponding target or epitope when the multispecific activatable antibody is in a cleaved state.
  • each of the MM in the multispecific activatable antibody is a polypeptide of about 2 to 40 amino acids in length. In some embodiments, each of the MM in the multispecific activatable antibody is a polypeptide of no more than 40 amino acids in length.
  • each of the MM in the multispecific activatable antibody has a polypeptide sequence that is different from that of target of the corresponding AB.
  • each of the MM in the multispecific activatable antibody has a polypeptide sequence that is no more than 50% identical to any natural binding partner of the corresponding AB. In some embodiments, each of the MM in the multispecific activatable antibody has a polypeptide sequence that is no more than 25% identical to any natural binding partner of the corresponding AB. In some embodiments, each of the MM in the multispecific activatable antibody has a polypeptide sequence that is no more than 10% identical to any natural binding partner of the corresponding AB.
  • the coupling of each of the MM reduces the ability of the corresponding AB to bind its target or epitope such that the dissociation constant (K d ) of the AB when coupled to the MM towards its corresponding target or epitope is at least 20 times greater than the K d of the AB when not coupled to the MM towards its corresponding target or epitope.
  • K d dissociation constant
  • the coupling of each of the MM reduces the ability of the corresponding AB to bind its target or epitope such that the dissociation constant (K d ) of the AB when coupled to the MM towards its corresponding target or epitope is at least 40 times greater than the K d of the AB when not coupled to the MM towards its corresponding target or epitope.
  • K d dissociation constant
  • the coupling of each of the MM reduces the ability of the corresponding AB to bind its target or epitope such that the dissociation constant (K d ) of the AB when coupled to the MM towards its corresponding target or epitope is at least 100 times greater than the K d of the AB when not coupled to the MM towards its corresponding target or epitope.
  • K d dissociation constant
  • the coupling of each of the MM reduces the ability of the corresponding AB to bind its target or epitope such that the dissociation constant (K d ) of the AB when coupled to the MM towards its corresponding target or epitope is at least 1000 times greater than the K d of the AB when not coupled to the MM towards its corresponding target or epitope.
  • the coupling of each of the MM reduces the ability of the corresponding AB to bind its target or epitope such that the dissociation constant (K d ) of the AB when coupled to the MM towards its corresponding target or epitope is at least 10,000 times greater than the K d of the AB when not coupled to the MM towards its corresponding target or epitope.
  • K d dissociation constant
  • each of the MM is an amino acid sequence selected from a MM disclosed herein.
  • CM1 and/or CM2 is cleaved by at least one MMP protease.
  • at least one of CM1 and/or CM2 includes an amino acid sequence selected from the group consisting of ISSGLLSS (SEQ ID NO: 14); QNQALRMA (SEQ ID NO: 15); AQNLLGMV (SEQ ID NO: 16); STFPFGMF (SEQ ID NO: 17); PVGYTSSL (SEQ ID NO: 18); DWLYWPGI (SEQ ID NO: 19); MIAPVAYR (SEQ ID NO: 20); RPSPMWAY (SEQ ID NO: 21); WATPRPMR (SEQ ID NO: 22); FRLLDWQW (SEQ ID NO: 23); LKAAPRWA (SEQ ID NO: 24); GPSHLVLT (SEQ ID NO: 25); LPGGLSPW (SEQ ID NO: 26); MGLFSEAG (SEQ ID NO: 27); SPLPLRVP (SEQ ID NO:
  • CM1 and/or CM2 includes an amino acid sequence selected from the group consisting of SEQ ID NOs: 364-370, 379-393, 402-409, 420-424, 434, 435, 450-452, 457, 470-472, 474, and 483.
  • CM1 and/or CM2 includes an amino acid sequence selected from the group consisting of SEQ ID NOs: 328, 336-339, and 348-351.
  • the protease that cleaves the first cleavable moiety (CM1) sequence is co-localized with the target of the AB1 in the multispecific activatable antibody in a tissue, and the protease cleaves the CM1 in the multispecific activatable antibody when the multispecific activatable antibody is exposed to the protease.
  • the multispecific activatable antibody includes more than one cleavable moiety sequence, and the protease that cleaves at least one cleavable moiety sequence is co-localized with the target of at least one of the AB regions in the multispecific activatable antibody in a tissue, and the protease cleaves the CM in the multispecific activatable antibody when the multispecific activatable antibody is exposed to the protease.
  • each CM e.g., CM1 and at least CM2 is positioned in the multispecific activatable antibody such that in the uncleaved state, binding of the multispecific activatable antibody to a target of one of the AB regions is reduced to occur with an equilibrium dissociation constant that is at least twofold greater than the equilibrium dissociation constant of an unmodified AB binding to its target, and whereas in the cleaved state, the AB binds its target.
  • each CM e.g., CM1 and at least CM2 is positioned in the multispecific activatable antibody such that in the uncleaved state, binding of the multispecific activatable antibody to a target of one of the AB regions is reduced to occur with an equilibrium dissociation constant that is at least threefold greater than the equilibrium dissociation constant of an unmodified AB binding to its target, and whereas in the cleaved state, the AB binds its target.
  • each CM e.g., CM1 and at least CM2 is positioned in the multispecific activatable antibody such that in the uncleaved state, binding of the multispecific activatable antibody to a target of one of the AB regions is reduced to occur with an equilibrium dissociation constant that is at least fourfold greater than the equilibrium dissociation constant of an unmodified AB binding to its target, and whereas in the cleaved state, the AB binds its target.
  • each CM e.g., CM1 and at least CM2 is positioned in the multispecific activatable antibody such that in the uncleaved state, binding of the multispecific activatable antibody to a target of one of the AB regions is reduced to occur with an equilibrium dissociation constant that is at least fivefold greater than the equilibrium dissociation constant of an unmodified AB binding to its target, and whereas in the cleaved state, the AB binds its target.
  • each CM e.g., CM1 and at least CM2 is positioned in the multispecific activatable antibody such that in the uncleaved state, binding of the multispecific activatable antibody to a target of one of the AB regions is reduced to occur with an equilibrium dissociation constant that is at least tenfold greater than the equilibrium dissociation constant of an unmodified AB binding to its target, and whereas in the cleaved state, the AB binds its target.
  • each CM e.g., CM1 and at least CM2 is positioned in the multispecific activatable antibody such that in the uncleaved state, binding of the multispecific activatable antibody to a target of one of the AB regions is reduced to occur with an equilibrium dissociation constant that is at least 20-fold greater than the equilibrium dissociation constant of an unmodified AB binding to its target, and whereas in the cleaved state, the AB binds its target.
  • each CM is positioned in the multispecific activatable antibody such that in the uncleaved state, binding of the multispecific activatable antibody to a target of one of the AB regions is reduced to occur with an equilibrium dissociation constant that is at least 40-fold greater than the equilibrium dissociation constant of an unmodified AB binding to its target, and whereas in the cleaved state, the AB binds its target.
  • each CM is positioned in the multispecific activatable antibody such that in the uncleaved state, binding of the multispecific activatable antibody to a target of one of the AB regions is reduced to occur with an equilibrium dissociation constant that is at least 50-fold greater than the equilibrium dissociation constant of an unmodified AB binding to its target, and whereas in the cleaved state, the AB binds its target.
  • each CM is positioned in the multispecific activatable antibody such that in the uncleaved state, binding of the multispecific activatable antibody to a target of one of the AB regions is reduced to occur with an equilibrium dissociation constant that is at least 100-fold greater than the equilibrium dissociation constant of an unmodified AB binding to its target, and whereas in the cleaved state, the AB binds its target.
  • each CM is positioned in the multispecific activatable antibody such that in the uncleaved state, binding of the multispecific activatable antibody to a target of one of the AB regions is reduced to occur with an equilibrium dissociation constant that is at least 200-fold greater than the equilibrium dissociation constant of an unmodified AB binding to its target, and whereas in the cleaved state, the AB binds its target.
  • each CM in the multispecific activatable antibody is a polypeptide of up to 15 amino acids in length.
  • At least one CM in the multispecific activatable antibody includes an amino acid sequence selected from the group consisting of SEQ ID NOs: 14-33 and 159 and the other CM includes the amino acid sequence LSGRSDNH (SEQ ID NO: 26). In some embodiments, at least one CM includes the amino acid sequence LSGRSDNH (SEQ ID NO: 26). In some embodiments, at least one cleavable moiety is selected for use with a specific protease, for example a protease that is known to be co-localized with at least one target of the multispecific activatable antibody.
  • cleavable moieties for use in the multispecific activatable antibodies of the disclosure are cleaved by at least a protease such as urokinase, legumain, and/or matriptase (also referred to herein as MT-SP1 or MTSP1).
  • a protease such as urokinase, legumain, and/or matriptase (also referred to herein as MT-SP1 or MTSP1).
  • a suitable cleavable moiety includes at least one of the following sequences: TGRGPSWV (SEQ ID NO: 27); SARGPSRW (SEQ ID NO: 28); TARGPSFK (SEQ ID NO: 29); LSGRSDNH (SEQ ID NO: 26); GGWHTGRN (SEQ ID NO: 30); HTGRSGAL (SEQ ID NO: 31); PLTGRSGG (SEQ ID NO: 32); AARGPAIH (SEQ ID NO: 33); RGPAFNPM (SEQ ID NO: 34); SSRGPAYL (SEQ ID NO: 35); RGPATPIM (SEQ ID NO: 36); RGPA (SEQ ID NO: 37); GGQPSGMWGW (SEQ ID NO: 38); FPRPLGITGL (SEQ ID NO: 39); VHMPLGFLGP (SEQ ID NO: 40); SPLTGRSG (SEQ ID NO: 41); SAGFSLPA (SEQ ID NO: 42); LAPLGLQRR (SEQ ID NO:
  • one CM is a substrate for at least one MMP protease and the other CM in the multispecific activatable antibody is a substrate for a protease selected from the group consisting of those shown in Table 7.
  • the protease is selected from the group consisting of uPA, legumain, matriptase, ADAM17, BMP-1, TMPRSS3, TMPRSS4, neutrophil elastase, MMP-7, MMP-9, MMP-12, MMP-13, and MMP-14.
  • the protease is a cathepsin, such as, but not limited to, cathepsin S.
  • each CM in the multispecific activatable antibody is a substrate for a protease selected from the group consisting of uPA (urokinase plasminogen activator), legumain and matriptase.
  • the protease comprises uPA.
  • the protease comprises legumain.
  • the protease comprises matriptase.
  • the protease comprises a matrix metalloproteinase (MMP).
  • MMP matrix metalloproteinase
  • At least one CM in the multispecific activatable antibody is a substrate for at least two proteases.
  • each protease is selected from the group consisting of those shown in Table 7.
  • at least one CM in the multispecific activatable antibody is a substrate for at least two proteases, wherein one of the proteases is selected from the group consisting of uPA, legumain and matriptase and the other protease is selected from the group consisting of those shown in Table 7.
  • at least one CM in the multispecific activatable antibody is a substrate for at least two proteases selected from the group consisting of uPA, legumain and matriptase.
  • the multispecific activatable antibody includes at least a first CM (CM1) and a second CM (CM2).
  • CM1 and CM2 are part of a single cleavable linker that joins an MM to an AB.
  • CM1 is part of a cleavable linker that joins MM1 to AB1
  • CM2 is part of a separate cleavable linker that joins an MM2 to AB2.
  • a multispecific activatable antibody comprises more than two CMs. In some embodiments, such a multispecific activatable antibody comprises more than two CMs and more than two MMs.
  • CM1 and CM2 are each polypeptides of no more than 15 amino acids long.
  • at least one of the first CM and the second CM is a polypeptide that functions as a substrate for a protease selected from the group consisting of those listed in Table 7.
  • at least one of the first CM and the second CM is a polypeptide that functions as a substrate for a protease selected from the group consisting of uPA, legumain, and matriptase.
  • the first CM is cleaved by a first cleaving agent selected from the group consisting of uPA, legumain, and matriptase in a target tissue and the second CM is cleaved by a second cleaving agent in a target tissue.
  • the other protease is selected from the group consisting of those shown in Table 7.
  • the first cleaving agent and the second cleaving agent are the same protease selected from the group consisting of those listed in Table 7, and the first CM and the second CM are different substrates for the enzyme.
  • the first cleaving agent and the second cleaving agent are the same protease selected from the group consisting of uPA, legumain, and matriptase, and the first CM and the second CM are different substrates for the enzyme.
  • the first cleaving agent and the second cleaving agent are the same protease selected from the group listed in Table 7, and the first CM and the second CM are the same substrate.
  • the first cleaving agent and the second cleaving agent are different proteases.
  • the first cleaving agent and the second cleaving agent are different proteases selected from the group consisting of those shown in Table 7.
  • the first cleaving agent and the second cleaving agent are co-localized in the target tissue. In some embodiments, the first CM and the second CM are cleaved by at least one cleaving agent in the target tissue.
  • the multispecific activatable antibody is exposed to and cleaved by a protease such that, in the activated or cleaved state, the activated multispecific activatable antibody includes a light chain amino acid sequence that includes at least a portion of LP2 and/or CM sequence after the protease has cleaved the CM.
  • compositions and methods that include a multispecific activatable antibody that includes at least a first antibody or antibody fragment (AB1) that specifically binds a target and a second antibody or antibody fragment (AB2), where at least the first AB in the multispecific activatable antibody is coupled to a masking moiety (MM1) that decreases the ability of AB1 to bind its target.
  • a multispecific activatable antibody that includes at least a first antibody or antibody fragment (AB1) that specifically binds a target and a second antibody or antibody fragment (AB2), where at least the first AB in the multispecific activatable antibody is coupled to a masking moiety (MM1) that decreases the ability of AB1 to bind its target.
  • MM1 masking moiety
  • each AB is coupled to a MM that decreases the ability of its corresponding AB to each target.
  • AB1 is coupled to a first masking moiety (MM1) that decreases the ability of AB1 to bind its target
  • AB2 is coupled to a second masking moiety (MM2) that decreases the ability of AB2 to bind its target.
  • the multispecific activatable antibody comprises more than two AB regions; in such embodiments, AB1 is coupled to a first masking moiety (MM1) that decreases the ability of AB1 to bind its target, AB2 is coupled to a second masking moiety (MM2) that decreases the ability of AB2 to bind its target, AB3 is coupled to a third masking moiety (MM3) that decreases the ability of AB3 to bind its target, and so on for each AB in the multispecific activatable antibody.
  • MM1 first masking moiety
  • MM2 is coupled to a second masking moiety
  • MM3 is coupled to a third masking moiety (MM3) that decreases the ability of AB3 to bind its target
  • the multispecific activatable antibody further includes at least one cleavable moiety (CM) that is a substrate for a protease, where the CM links a MM to an AB.
  • the multispecific activatable antibody includes at least a first antibody or antibody fragment (AB1) that specifically binds a target and a second antibody or antibody fragment (AB2), where at least the first AB in the multispecific activatable antibody is coupled via a first cleavable moiety (CM1) to a masking moiety (MM1) that decreases the ability of AB1 to bind its target.
  • AB1 is coupled via CM1 to MM1, and AB2 is coupled via a second cleavable moiety (CM2) to a second masking moiety (MM2) that decreases the ability of AB2 to bind its target.
  • the multispecific activatable antibody comprises more than two AB regions; in some of these embodiments, AB1 is coupled via CM1 to MM1, AB2 is coupled via CM2 to MM2, and AB3 is coupled via a third cleavable moiety (CM3) to a third masking moiety (MM3) that decreases the ability of AB3 to bind its target, and so on for each AB in the multispecific activatable antibody.
  • the disclosure also provides activatable antibodies that include non-binding steric moieties (NB) or binding partners (BP) for non-binding steric moieties, where the BP recruits or otherwise attracts the NB to the activatable antibody.
  • the activatable antibodies provided herein include, for example, an activatable antibody that includes a non-binding steric moiety (NB), a cleavable linker (CL) and antibody or antibody fragment (AB) that binds a target; an activatable antibody that includes a binding partner for a non-binding steric moiety (BP), a CL and an AB; and an activatable antibody that includes a BP to which an NB has been recruited, a CL and an AB that binds the target.
  • NB non-binding steric moiety
  • CL cleavable linker
  • AB antibody or antibody fragment
  • NB-containing activatable antibodies Activatable antibodies in which the NB is covalently linked to the CL and AB of the activatable antibody or is associated by interaction with a BP that is covalently linked to the CL and AB of the activatable antibody are referred to herein as “NB-containing activatable antibodies.”
  • activatable or switchable is meant that the activatable antibody exhibits a first level of binding to a target when the activatable antibody is in an inhibited, masked or uncleaved state (i.e., a first conformation), and a second level of binding to the target when the activatable antibody is in an uninhibited, unmasked and/or cleaved state (i.e., a second conformation, i.e., activated antibody), where the second level of target binding is greater than the first level of target binding.
  • the activatable antibody compositions can exhibit increased bioavailability and more favorable biodistribution compared to conventional antibody therapeutics.
  • activatable antibodies provide for reduced toxicity and/or adverse side effects that could otherwise result from binding of the at non-treatment sites and/or non-diagnostic sites if the AB were not masked or otherwise inhibited from binding to such a site.
  • the activatable antibody includes a non-binding steric moiety (NB); a cleavable linker (CL); and an antibody or antibody fragment (AB) that binds specifically to the target, wherein the NB is a polypeptide that does not bind specifically to the AB; the CL is a polypeptide that includes a substrate (S) for an enzyme; the CL is positioned such that in an uncleaved state, the NB interferes with binding of the AB to the target and in a cleaved state, the NB does not interfere with binding of the AB to the target; and the NB does not inhibit cleavage of the CL by the enzyme.
  • NB non-binding steric moiety
  • CL cleavable linker
  • AB antibody or antibody fragment
  • polypeptide refers to any polypeptide that includes at least two amino acid residues, including larger polypeptides, full-length proteins and fragments thereof, and the term polypeptide is not limited to single-chain polypeptides and can include multi-unit, e.g., multi-chain, polypeptides.
  • polypeptide and polypeptide are used interchangeably herein, and in cases where the polypeptide is of a longer length, e.g., 50 amino acids or greater, the terms polypeptide and protein are used interchangeably herein.
  • the activatable antibody includes a non-binding steric moiety (NB); a cleavable linker (CL); and an antibody or antibody fragment (AB) that binds specifically to the target, wherein (i) the NB includes a polypeptide that does not bind specifically to the AB; (ii) CL is a polypeptide of up to 50 amino acids in length that includes a substrate (S) for an enzyme; (iii) the CL is positioned such that in an uncleaved state, the NB interferes with binding of the AB to the target and in a cleaved state, the NB does not interfere with binding of the AB to the target; and (iv) the NB does not inhibit cleavage of the CL by the enzyme.
  • NB non-binding steric moiety
  • CL cleavable linker
  • AB antibody or antibody fragment
  • the CL has a length of up to 15 amino acids, a length of up to 20 amino acids, a length of up to 25 amino acids, a length of up to 30 amino acids, a length of up to 35 amino acids, a length of up to 40 amino acids, a length of up to 45 amino acids, a length of up to 50 amino acids, a length in the range of 10-50 amino acids, a length in the range of 15-50 amino acids, a length in the range of 20-50 amino acids, a length in the range of 25-50 amino acids, a length in the range of 30-50 amino acids, a length in the range of 35-50 amino acids, a length in the range of 40-50 amino acids, a length in the range of 45-50 amino acids, a length in the range of 10-40 amino acids, a length in the range of 15-40 amino acids, a length in the range of 20-40 amino acids, a length in the range of 25-40 amino acids, a length in the range of 30-40 amino acids, a length in the range of 35-40 amino acids, a length
  • the activatable antibody includes a non-binding steric moiety (NB); a cleavable linker (CL); and an antibody or antibody fragment (AB) that binds specifically to the target, wherein (i) the NB includes a polypeptide that does not bind specifically to the AB; (ii) the CL is a polypeptide that includes a substrate (S) for an enzyme; (iii) the CL is positioned such that in an uncleaved state, the NB interferes with binding of the AB to the target and in a cleaved state, the NB does not interfere with binding of the AB to the target; (iv) the NB does not inhibit cleavage of the CL by the enzyme; and (v) the activatable antibody has the structural arrangement from N-terminus to C-terminus as follows in the uncleaved state: NB-CL-AB or AB-CL-NB.
  • the activatable antibody includes a non-binding steric moiety (NB); a cleavable linker (CL); and an antibody or antibody fragment (AB) that binds specifically to the target, wherein (i) the NB includes a polypeptide that does not bind specifically to the AB; (ii) the CL is a polypeptide that includes a substrate (S) for an enzyme; (iii) the CL is positioned such that in an uncleaved state, the NB interferes with binding of the AB to the target and in a cleaved state, the NB does not interfere with binding of the AB to the target, and wherein the NB in the uncleaved activatable antibody reduces the ability of the AB to bind the target by at least 50%, for example, by at least 60%, by at least 70%, by at least 75%, by at least 80%, by at least 85%, by at least 90%, by at least 95%, by at least 96%, by at least 97%, by at least
  • the reduction in the ability of the AB to bind the target is determined, e.g., using an assay as described herein or an in vitro target displacement assay such as, for example, the assay described in PCT Publication Nos. WO 2009/025846 and WO 2010/081173.
  • the activatable antibody includes a binding partner (BP) for a non-binding steric moiety (NB); a cleavable linker (CL); and an antibody or antibody fragment (AB) that binds specifically to the target, wherein the BP is a polypeptide that binds to the NB when exposed thereto; the NB does not bind specifically to the AB; the CL is a polypeptide that includes a substrate (S) for an enzyme; the CL is positioned such that in an uncleaved state in the presence of the NB, the NB interferes with binding of the AB to the target and in a cleaved state, the NB does not interfere with binding of the AB to the target and the BP does not interfere with binding of the AB to the target; and the NB and the BP do not inhibit cleavage of the CL by the enzyme.
  • the BP of the activatable antibody is optionally bound to the NB.
  • the NB is recruited by
  • the activatable antibody is formulated as a composition.
  • the composition also includes the NB, where the NB is co-formulated with the activatable antibody that includes the BP, the CL, and the AB.
  • the BP is selected from the group consisting of an albumin binding peptide, a fibrinogen binding peptide, a fibronectin binding peptide, a hemoglobin binding peptide, a transferrin binding peptide, an immunoglobulin domain binding peptide, and other serum protein binding peptides.
  • the NB is a soluble, globular protein. In some examples of any of these activatable antibody embodiments, the NB is a protein that circulates in the bloodstream. In some examples of any of these activatable antibody embodiments, the NB is selected from the group consisting of albumin, fibrinogen, fibronectin, hemoglobin, transferrin, an immunoglobulin domain, and other serum proteins.
  • the CL is a polypeptide that includes a substrate (S) for a protease.
  • the protease is co-localized with the in a tissue, and the protease cleaves the CL in the activatable antibody when the activatable antibody is exposed to the protease.
  • the CL is a polypeptide of up to 50 amino acids in length.
  • the CL is a polypeptide that includes a substrate (S) having a length of up to 15 amino acids, e.g., 3 amino acids long, 4 amino acids long, 5 amino acids long, 6 amino acids long, 7 amino acids long, 8 amino acids long, 9 amino acids long, 10 amino acids long, 11 amino acids long, 12 amino acids long, 13 amino acids long, 14 amino acids long, or 15 amino acids long.
  • S substrate
  • the activatable antibody has the structural arrangement from N-terminus to C-terminus as follows in the uncleaved state: NB-CL-AB, AB-CL-NB, BP-CL-AB or AB-CL-BP.
  • the activatable antibody includes a BP and the activatable antibody is in the presence of the corresponding NB
  • the activatable antibody has a structural arrangement from N-terminus to C-terminus as follows in the uncleaved state: NB:BP-CM-AB or AB-CM-BP:NB, where “:” represents an interaction, e.g., binding, between the NB and BP.
  • the activatable antibody includes an antibody or antigen-binding fragment thereof that specifically binds a given target and is a monoclonal antibody, domain antibody, single chain, Fab fragment, a F(ab′) 2 fragment, a scFv, a scab, a dAb, a single domain heavy chain antibody, or a single domain light chain antibody.
  • an antibody or immunologically active fragment thereof that binds the target a mouse, other rodent, chimeric, humanized or fully human monoclonal antibody.
  • the activatable antibody includes a combination of a variable heavy chain region comprising an amino acid sequence presented herein and a variable light chain region comprising an amino acid sequence presented herein.
  • the activatable antibody includes a combination of a variable heavy chain region comprising an amino acid sequence that is at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more identical to an amino acid sequence presented herein, and a variable light chain region comprising an amino acid sequence that is at least 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or more identical to an amino acid sequence presented herein.
  • the activatable antibody also includes an agent conjugated to the AB.
  • the agent is a therapeutic agent.
  • the agent is an antineoplastic agent.
  • the agent is a toxin or fragment thereof.
  • the agent is conjugated to the AB via a linker.
  • the linker is a cleavable linker.
  • the agent is conjugated to the AB via a noncleavable linker.
  • the agent is an agent selected from the group listed in Table 3.
  • the agent is a microtubule inhibitor.
  • the agent is a nucleic acid damaging agent, such as a DNA alkylator or DNA intercalator, or other DNA damaging agent.
  • the agent is a dolastatin.
  • the agent is an auristatin or derivative thereof.
  • the agent is auristatin E or a derivative thereof.
  • the agent is monomethyl auristatin E (MMAE).
  • the agent is monomethyl auristatin D (MMAD).
  • the agent is a maytansinoid or maytansinoid derivative.
  • the agent is DM1 or DM4.
  • the agent is a duocarmycin or derivative thereof.
  • the agent is a calicheamicin or derivative thereof.
  • the agent is a pyrrolobenzodiazepine.
  • the activatable antibody also includes a detectable moiety.
  • the detectable moiety is a diagnostic agent.
  • the activatable antibody also includes a spacer. In some examples of any of these activatable antibody embodiments, the activatable antibody also includes a signal peptide. In some embodiments, the signal peptide is conjugated to the activatable antibody via a spacer. In some examples of any of these activatable antibody embodiments, the spacer is joined directly to the MM of the activatable antibody.
  • the serum half-life of the activatable antibody is longer than that of the corresponding antibody; e.g., the pK of the activatable antibody is longer than that of the corresponding antibody. In some embodiments, the serum half-life of the activatable antibody is similar to that of the corresponding antibody. In some embodiments, the serum half-life of the activatable antibody is at least 15 days when administered to an organism. In some embodiments, the serum half-life of the activatable antibody is at least 12 days when administered to an organism. In some embodiments, the serum half-life of the activatable antibody is at least 11 days when administered to an organism. In some embodiments, the serum half-life of the activatable antibody is at least 10 days when administered to an organism.
  • the serum half-life of the activatable antibody is at least 9 days when administered to an organism. In some embodiments, the serum half-life of the activatable antibody is at least 8 days when administered to an organism. In some embodiments, the serum half-life of the activatable antibody is at least 7 days when administered to an organism. In some embodiments, the serum half-life of the activatable antibody is at least 6 days when administered to an organism. In some examples of any of these activatable antibody embodiments, the serum half-life of the activatable antibody is at least 5 days when administered to an organism. In some embodiments, the serum half-life of the activatable antibody is at least 4 days when administered to an organism.
  • the serum half-life of the activatable antibody is at least 3 days when administered to an organism. In some embodiments, the serum half-life of the activatable antibody is at least 2 days when administered to an organism. In some embodiments, the serum half-life of the activatable antibody is at least 24 hours when administered to an organism. In some embodiments, the serum half-life of the activatable antibody is at least 20 hours when administered to an organism. In some embodiments, the serum half-life of the activatable antibody is at least 18 hours when administered to an organism. In some embodiments, the serum half-life of the activatable antibody is at least 16 hours when administered to an organism. In some embodiments, the serum half-life of the activatable antibody is at least 14 hours when administered to an organism.
  • the serum half-life of the activatable antibody is at least 12 hours when administered to an organism. In some embodiments, the serum half-life of the activatable antibody is at least 10 hours when administered to an organism. In some embodiments, the serum half-life of the activatable antibody is at least 8 hours when administered to an organism. In some embodiments, the serum half-life of the activatable antibody is at least 6 hours when administered to an organism. In some embodiments, the serum half-life of the activatable antibody is at least 4 hours when administered to an organism. In some embodiments, the serum half-life of the activatable antibody is at least 3 hours when administered to an organism.
  • the disclosure also provides an isolated nucleic acid molecule encoding any of these activatable antibodies, as well as vectors that include these isolated nucleic acid sequences.
  • the disclosure provides methods of producing an activatable antibody by culturing a cell under conditions that lead to expression of the activatable antibody, wherein the cell comprises such a nucleic acid sequence. In some embodiments, the cell comprises such a vector.
  • the dissociation constant (K d ) of the NB-containing activatable antibody toward the target is greater than the K d of the AB towards the target when it is not associated with the NB or NB:BP.
  • the dissociation constant (K d ) of the NB-containing activatable antibody toward the target is greater than the K d of the parental AB towards the target.
  • the K d of the NB-containing activatable antibody toward the target is at least 5, 10, 25, 50, 100, 250, 500, 1,000, 2,500, 5,000, 10,000, 50,000, 100,000, 500,000, 1,000,000, 5,000,000, 10,000,000, 50,000,000 or greater, or between 5-10, 10-100, 10-1,000, 10-10,000, 10-100,000, 10-1,000,000, 10-10,000,000, 100-1,000, 100-10,000, 100-100,000, 100-1,000,000, 100-10,000,000, 1,000-10,000, 1,000-100,000, 1,000-1,000,000, 1000-10,000,000, 10,000-100,000, 10,000-1,000,000, 10,000-10,000,000, 100,000-1,000,000, or 100,000-10,000,000 times greater than the K d of the AB when it is not associated with the NB or NB:BP or the K d of the parental AB towards the target.
  • the binding affinity of the NB-containing activatable antibody towards the target is lower than the binding affinity of the AB when it is not associated with the NB or NB:BP or lower than the binding affinity of the parental AB towards the target.
  • the binding affinity of the NB-containing activatable antibody toward the target is at least 5, 10, 25, 50, 100, 250, 500, 1,000, 2,500, 5,000, 10,000, 50,000, 100,000, 500,000, 1,000,000, 5,000,000, 10,000,000, 50,000,000 or greater, or between 5-10, 10-100, 10-1,000, 10-10,000, 10-100,000, 10-1,000,000, 10-10,000,000, 100-1,000, 100-10,000, 100-100,000, 100-1,000,000, 100-10,000,000, 1,000-10,000, 1,000-100,000, 1,000-1,000,000, 1000-10,000,000, 10,000-100,000, 10,000-1,000,000, 10,000-10,000,000, 100,000-1,000,000, or 100,000-10,000,000 times lower than the binding affinity of the AB when it is not associated with the NB or NB:BP or lower than the binding affinity of the parental AB towards the target.
  • the NB-containing activatable antibody When the NB-containing activatable antibody is in the presence of the target, specific binding of the AB to the target is reduced or inhibited, as compared to the specific binding of the AB when it is not associated with the NB or NB:BP. When the NB-containing activatable antibody is in the presence of the target, specific binding of the AB to the target is reduced or inhibited, as compared to the specific binding of the parental AB to the target.
  • the ability of the NB-containing activatable antibody to bind the target is reduced, for example, by at least 50%, 60%, 70%, 80%, 90%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or even 100% for at least 2, 4, 6, 8, 12, 28, 24, 30, 36, 48, 60, 72, 84, or 96 hours, or 5, 10, 15, 30, 45, 60, 90, 120, 150, or 180 days, or 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, or 12 months or longer when measured in vitro and/or in vivo.
  • a modifying agent for example a protease or other enzyme
  • specific binding of the AB to the target is reduced or inhibited, as compared to the specific binding of the AB when it is not associated with the NB or NB:BP.
  • a modifying agent for example a protease, other enzyme, reduction agent, or light
  • specific binding of the AB to the target is reduced or inhibited, as compared to the specific binding of the parental AB to the target.
  • the ability of the NB-containing activatable antibody to bind the target is reduced, for example, by at least 50%, 60%, 70%, 80%, 90%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or even 100% for at least 2, 4, 6, 8, 12, 28, 24, 30, 36, 48, 60, 72, 84, or 96 hours, or 5, 10, 15, 30, 45, 60, 90, 120, 150, or 180 days, or 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, or 12 months or longer when measured in vitro and/or in vivo.
  • the activatable antibody includes an agent conjugated to the AB to produce an activatable antibody conjugate.
  • the agent is a therapeutic agent.
  • the agent is a diagnostic agent.
  • the agent is a detectable marker.
  • the agent is an antineoplastic agent.
  • the agent is a toxin or fragment thereof.
  • the agent is conjugated to the AB via a linker.
  • the linker is a cleavable linker.
  • the agent is conjugated to the AB via a noncleavable linker.
  • the agent is a microtubule inhibitor.
  • the agent is a nucleic acid damaging agent, such as a DNA alkylator or DNA intercalator, or other DNA damaging agent.
  • the agent is an agent selected from the group listed in Table 3.
  • the agent is a dolastatin.
  • the agent is an auristatin or derivative thereof.
  • the agent is auristatin E or a derivative thereof.
  • the agent is monomethyl auristatin E (MMAE).
  • the agent is monomethyl auristatin D (MMAD).
  • the agent is a maytansinoid or maytansinoid derivative. In some embodiments, the agent is DM1 or DM4. In some embodiments, the agent is a duocarmycin or derivative thereof. In some embodiments, the agent is a calicheamicin or derivative thereof. In some embodiments, the agent is a pyrrolobenzodiazepine.
  • the activatable antibodies are dual-target binding activatable antibodies.
  • Such dual target binding activatable antibodies contain two Abs that may bind the same or different targets.
  • dual-targeting activatable antibodies contain bispecific antibodies or antibody fragments.
  • Dual target binding activatable antibodies are designed so as to have a CL cleavable by a cleaving agent that is co-localized in a target tissue with one or both of the targets capable of binding to the ABs of the activatable antibodies.
  • Dual target binding activatable antibodies with more than one AB to the same or different targets can be designed so as to have more than one CL, wherein the first CL is cleavable by a cleaving agent in a first target tissue and wherein the second CL is cleavable by a cleaving agent in a second target tissue, with one or more of the targets binding to the ABs of the activatable antibodies.
  • the first and second target tissues are spatially separated, for example, at different sites in the organism.
  • the first and second target tissues are the same tissue temporally separated, for example the same tissue at two different points in time, for example the first time point is when the tissue is an early stage tumor, and the second time point is when the tissue is a late stage tumor.
  • the disclosure also provides nucleic acid molecules encoding the activatable antibodies described herein.
  • the disclosure also provides vectors that include these nucleic acids.
  • the activatable antibodies described herein are produced by culturing a cell under conditions that lead to expression of the activatable antibody, wherein the cell includes these nucleic acid molecules or vectors.
  • the disclosure also provides methods of manufacturing activatable antibodies.
  • the method includes the steps of (a) culturing a cell that includes a nucleic acid construct that encodes the activatable antibody under conditions that lead to expression of the activatable antibody, wherein the activatable antibody includes (i) a non-binding steric moiety (NB); (ii) a cleavable linker (CL); and (iii) an antibody or an antigen binding fragment thereof (AB) that specifically binds a target, wherein (1) the NB does not bind specifically to the AB; (2) the CL is a polypeptide that includes a substrate (S) for an enzyme; (3) the CL is positioned such that in an uncleaved state, the NB interferes with binding of the AB to the target and in a cleaved state, the NB does not interfere with binding of the AB to the target; and (4) the NB does not inhibit cleavage of the CL by the enzyme; and (b) recovering the activatable antibody.
  • the method includes the steps of (a) culturing a cell that includes a nucleic acid construct that encodes the activatable antibody under conditions that lead to expression of the activatable antibody, wherein the activatable antibody includes (i) a binding partner (BP) for a non-binding steric moiety (NB); (ii) a cleavable linker (CL); and (iii) an antibody or an antigen binding fragment thereof (AB) that specifically binds a target, wherein (1) the NB does not bind specifically to the AB; (2) the CL is a polypeptide that includes a substrate (S) for an enzyme; (3) the CL is positioned such that in an uncleaved state in the presence of the NB, the NB interferes with binding of the AB to the target and in a cleaved state, the NB does not interfere with binding of the AB to the target and the BP does not interfere with binding of the AB to the target; and (4) the NB and the BP do
  • formulations include, for example, powders, pastes, ointments, jellies, waxes, oils, lipids, lipid (cationic or anionic) containing vesicles (such as LipofectinTM), DNA conjugates, anhydrous absorption pastes, oil-in-water and water-in-oil emulsions, emulsions carbowax (polyethylene glycols of various molecular weights), semi-solid gels, and semi-solid mixtures containing carbowax. Any of the foregoing mixtures may be appropriate in treatments and therapies in accordance with the present disclosure, provided that the active ingredient in the formulation is not inactivated by the formulation and the formulation is physiologically compatible and tolerable with the route of administration.
  • Therapeutic formulations of the disclosure which include a conjugated antibody, an activatable antibody and/or a conjugated activatable antibody, are used to prevent, treat or otherwise ameliorate a disease or disorder associated with aberrant target expression and/or activity.
  • therapeutic formulations of the disclosure which include a conjugated antibody, an activatable antibody and/or a conjugated activatable antibody, are used to treat or otherwise ameliorate inflammation, an inflammatory disorder, an autoimmune disease and/or a cancer or other neoplastic condition.
  • the cancer is a solid tumor or a hematologic malignancy where the target is expressed. In some embodiments, the cancer is a solid tumor where the target is expressed.
  • the cancer is a hematologic malignancy where the target is expressed.
  • the target is expressed on parenchyma (e.g., in cancer, the portion of an organ or tissue that often carries out function(s) of the organ or tissue).
  • the target is expressed on a cell, tissue, or organ.
  • the target is expressed on stroma (i.e., the connective supportive framework of a cell, tissue, or organ).
  • the target is expressed on an osteoblast.
  • the target is expressed on the endothelium (vasculature).
  • the target is expressed on a cancer stem cell.
  • the agent to which the activatable antibody is conjugated is a microtubule inhibitor.
  • the agent to which the activatable antibody is conjugated is a nucleic acid damaging agent.
  • Efficaciousness of prevention, amelioration or treatment is determined in association with any known method for diagnosing or treating the disease or disorder associated with target expression and/or activity, such as, for example, aberrant target expression and/or activity.
  • Prolonging the survival of a subject or otherwise delaying the progression of the disease or disorder associated with target expression and/or activity, e.g., aberrant target expression and/or activity, in a subject indicates that the conjugated antibody, activatable antibody and/or conjugated activatable antibody confers a clinical benefit.
  • a conjugated antibody, an activatable antibody and/or a conjugated activatable antibody can be administered in the form of pharmaceutical compositions.
  • Principles and considerations involved in preparing such compositions, as well as guidance in the choice of components are provided, for example, in Remington: The Science And Practice Of Pharmacy 19th ed. (Alfonso R. Gennaro, et al., editors) Mack Pub. Co., Easton, Pa.: 1995; Drug Absorption Enhancement: Concepts, Possibilities, Limitations, And Trends, Harwood Academic Publishers, Langhorne, Pa., 1994; and Peptide And Protein Drug Delivery (Advances In Parenteral Sciences, Vol. 4), 1991, M. Dekker, New York.
  • the smallest fragment that specifically binds to the binding domain of the target protein is selected.
  • peptide molecules can be designed that retain the ability to bind the target protein sequence.
  • Such peptides can be synthesized chemically and/or produced by recombinant DNA technology. (See, e.g., Marasco et al., Proc. Natl. Acad. Sci. USA, 90: 7889-7893 (1993)).
  • the formulation can also contain more than one active compounds as necessary for the particular indication being treated, for example, in some embodiments, those with complementary activities that do not adversely affect each other.
  • the composition can comprise an agent that enhances its function, such as, for example, a cytotoxic agent, cytokine, chemotherapeutic agent, or growth-inhibitory agent.
  • an agent that enhances its function such as, for example, a cytotoxic agent, cytokine, chemotherapeutic agent, or growth-inhibitory agent.
  • Such molecules are suitably present in combination in amounts that are effective for the purpose intended.
  • the active ingredients can also be entrapped in microcapsules prepared, for example, by coacervation techniques or by interfacial polymerization, for example, hydroxymethylcellulose or gelatin-microcapsules and poly-(methylmethacrylate) microcapsules, respectively, in colloidal drug delivery systems (for example, liposomes, albumin microspheres, microemulsions, nano-particles, and nanocapsules) or in macroemulsions.
  • colloidal drug delivery systems for example, liposomes, albumin microspheres, microemulsions, nano-particles, and nanocapsules
  • the formulations to be used for in vivo administration must be sterile. This is readily accomplished by filtration through sterile filtration membranes.
  • sustained-release preparations can be prepared. Suitable examples of sustained-release preparations include semipermeable matrices of solid hydrophobic polymers containing the antibody, which matrices are in the form of shaped articles, e.g., films, or microcapsules. Examples of sustained-release matrices include polyesters, hydrogels (for example, poly(2-hydroxyethyl-methacrylate), or poly(vinylalcohol)), polylactides (U.S. Pat. No.
  • copolymers of L-glutamic acid and ⁇ ethyl-L-glutamate copolymers of L-glutamic acid and ⁇ ethyl-L-glutamate, non-degradable ethylene-vinyl acetate, degradable lactic acid-glycolic acid copolymers such as the LUPRON DEPOTTM (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.
  • the conjugated antibody, activatable antibody and/or conjugated activatable antibody contains a detectable label.
  • An intact antibody, or a fragment thereof e.g., Fab, scFv, or F(ab) 2 ) is used.
  • the term “labeled”, with regard to the probe or antibody is intended to encompass direct labeling of the probe or antibody by coupling (i.e., physically linking) a detectable substance to the probe or antibody, as well as indirect labeling of the probe or antibody by reactivity with another reagent that is directly labeled.
  • Examples of indirect labeling include detection of a primary antibody using a fluorescently-labeled secondary antibody and end-labeling of a DNA probe with biotin such that it can be detected with fluorescently-labeled streptavidin.
  • bio sample is intended to include tissues, cells and biological fluids isolated from a subject, as well as tissues, cells and fluids present within a subject. Included within the usage of the term “biological sample”, therefore, is blood and a fraction or component of blood including blood serum, blood plasma, or lymph. That is, the detection method of the disclosure can be used to detect an analyte mRNA, protein, or genomic DNA in a biological sample in vitro as well as in vivo.
  • in vitro techniques for detection of an analyte mRNA include Northern hybridizations and in situ hybridizations.
  • In vitro techniques for detection of an analyte protein include enzyme linked immunosorbent assays (ELISAs), Western blots, immunoprecipitations, immunochemical staining, and immunofluorescence.
  • In vitro techniques for detection of an analyte genomic DNA include Southern hybridizations. Procedures for conducting immunoassays are described, for example in “ELISA: Theory and Practice: Methods in Molecular Biology”, Vol. 42, J. R. Crowther (Ed.) Human Press, Totowa, N.J., 1995; “Immunoassay”, E. Diamandis and T.
  • in vivo techniques for detection of an analyte protein include introducing into a subject a labeled anti-analyte protein antibody.
  • the antibody can be labeled with a radioactive marker whose presence and location in a subject can be detected by standard imaging techniques.
  • conjugated antibodies, activatable antibodies and/or conjugated activatable antibodies of the disclosure are also useful in a variety of diagnostic and prophylactic formulations.
  • a conjugated antibody, an activatable antibody and/or a conjugated activatable antibody is administered to patients that are at risk of developing one or more of the aforementioned disorders.
  • a patient's or organ's predisposition to one or more of the aforementioned disorders can be determined using genotypic, serological or biochemical markers.
  • a conjugated antibody, an activatable antibody and/or a conjugated activatable antibody is administered to human individuals diagnosed with a clinical indication associated with one or more of the aforementioned disorders. Upon diagnosis, a conjugated antibody, an activatable antibody and/or a conjugated activatable antibody is administered to mitigate or reverse the effects of the clinical indication.
  • a conjugated antibody, an activatable antibody and/or a conjugated activatable antibody of the disclosure is also useful in the detection of a target in patient samples and accordingly are useful as diagnostics.
  • the antibodies and/or activatable antibodies, and conjugated versions thereof, of the disclosure are used in in vitro assays, e.g., ELISA, to detect target levels in a patient sample.
  • a conjugated antibody, an activatable antibody and/or a conjugated activatable antibody of the disclosure is immobilized on a solid support (e.g., the well(s) of a microtiter plate).
  • the immobilized conjugated antibody, activatable antibody and/or conjugated activatable antibody serves as a capture antibody for any target that may be present in a test sample.
  • the solid support Prior to contacting the immobilized antibody with a patient sample, the solid support is rinsed and treated with a blocking agent such as milk protein or albumin to prevent nonspecific adsorption of the analyte.
  • the wells are treated with a test sample suspected of containing the antigen, or with a solution containing a standard amount of the antigen.
  • a sample is, e.g., a serum sample from a subject suspected of having levels of circulating antigen considered to be diagnostic of a pathology.
  • the solid support is treated with a second antibody that is detectably labeled.
  • the labeled second antibody serves as a detecting antibody. The level of detectable label is measured, and the concentration of target antigen in the test sample is determined by comparison with a standard curve developed from the standard samples.
  • a conjugated antibody, an activatable antibody and/or a conjugated activatable antibody can also be used in diagnostic and/or imaging methods.
  • such methods are in vitro methods.
  • such methods are in vivo methods.
  • such methods are in situ methods.
  • such methods are ex vivo methods.
  • activatable antibodies having an enzymatically cleavable CM can be used to detect the presence or absence of an enzyme that is capable of cleaving the CM.
  • Such activatable antibodies can be used in diagnostics, which can include in vivo detection (e.g., qualitative or quantitative) of enzyme activity (or, in some embodiments, an environment of increased reduction potential such as that which can provide for reduction of a disulfide bond) through measured accumulation of activated antibodies (i.e., antibodies resulting from cleavage of an activatable antibody) in a given cell or tissue of a given host organism.
  • activated antibodies i.e., antibodies resulting from cleavage of an activatable antibody
  • Such accumulation of activated antibodies indicates not only that the tissue expresses enzymatic activity (or an increased reduction potential depending on the nature of the CM) but also that the tissue expresses target to which the activated antibody binds.
  • the CM can be selected to be substrate for a matrix metalloprotease (MMP) found at the site of a tumor, at the site of a viral or bacterial infection at a biologically confined site (e.g., such as in an abscess, in an organ, and the like), and the like.
  • MMP matrix metalloprotease
  • the AB can be one that binds a target antigen.
  • a detectable label e.g., a fluorescent label or radioactive label or radiotracer
  • Suitable detectable labels are discussed in the context of the above screening methods and additional specific examples are provided below.
  • activatable antibodies will exhibit an increased rate of binding to disease tissue relative to tissues where the CM specific enzyme is not present at a detectable level or is present at a lower level than in disease tissue or is inactive (e.g., in zymogen form or in complex with an inhibitor). Since small proteins and peptides are rapidly cleared from the blood by the renal filtration system, and because the enzyme specific for the CM is not present at a detectable level (or is present at lower levels in non-disease tissues or is present in inactive conformation), accumulation of activated antibodies in the disease tissue is enhanced relative to non-disease tissues.
  • activatable antibodies can be used to detect the presence or absence of a cleaving agent in a sample.
  • the activatable antibodies can be used to detect (either qualitatively or quantitatively) the presence of an enzyme in the sample.
  • the activatable antibodies can be used to detect (either qualitatively or quantitatively) the presence of reducing conditions in a sample.
  • the activatable antibodies can be detectably labeled, and can be bound to a support (e.g., a solid support, such as a slide or bead).
  • the detectable label can be positioned on a portion of the activatable antibody that is not released following cleavage, for example, the detectable label can be a quenched fluorescent label or other label that is not detectable until cleavage has occurred.
  • the assay can be conducted by, for example, contacting the immobilized, detectably labeled activatable antibodies with a sample suspected of containing an enzyme and/or reducing agent for a time sufficient for cleavage to occur, then washing to remove excess sample and contaminants.
  • the presence or absence of the cleaving agent (e.g., enzyme or reducing agent) in the sample is then assessed by a change in detectable signal of the activatable antibodies prior to contacting with the sample e.g., the presence of and/or an increase in detectable signal due to cleavage of the activatable antibody by the cleaving agent in the sample.
  • the cleaving agent e.g., enzyme or reducing agent
  • Such detection methods can be adapted to also provide for detection of the presence or absence of a target that is capable of binding the AB of the activatable antibodies when cleaved.
  • the assays can be adapted to assess the presence or absence of a cleaving agent and the presence or absence of a target of interest.
  • the presence or absence of the cleaving agent can be detected by the presence of and/or an increase in detectable label of the activatable antibodies as described above, and the presence or absence of the target can be detected by detection of a target-AB complex e.g., by use of a detectably labeled anti-target antibody.
  • Activatable antibodies are also useful in in situ imaging for the validation of activatable antibody activation, e.g., by protease cleavage, and binding to a particular target.
  • In situ imaging is a technique that enables localization of proteolytic activity and target in biological samples such as cell cultures or tissue sections. Using this technique, it is possible to confirm both binding to a given target and proteolytic activity based on the presence of a detectable label (e.g., a fluorescent label).
  • a detectable label e.g., a fluorescent label
  • an activatable antibody is labeled with a detectable label.
  • the detectable label may be a fluorescent dye, (e.g. a fluorophore, Fluorescein Isothiocyanate (FITC), Rhodamine Isothiocyanate (TRITC), an Alexa Fluor® label), a near infrared (NIR) dye (e.g., Qdot® nanocrystals), a colloidal metal, a hapten, a radioactive marker, biotin and an amplification reagent such as streptavidin, or an enzyme (e.g. horseradish peroxidase or alkaline phosphatase).
  • FITC Fluorescein Isothiocyanate
  • TRITC Rhodamine Isothiocyanate
  • Alexa Fluor® label Alexa Fluor® label
  • NIR near infrared
  • colloidal metal e.g., a hapten, a radioactive marker, biotin and an
  • Detection of the label in a sample that has been incubated with the labeled, activatable antibody indicates that the sample contains the target and contains a matrix metalloprotease (MMP) that is specific for the CM of the activatable antibody.
  • MMP matrix metalloprotease
  • the presence of the MMP can be confirmed using broad spectrum protease inhibitors such as those described herein, and/or by using an agent that is specific for the protease, for example, an antibody such as A11, which is specific for the protease matriptase (MT-SP1) and inhibits the proteolytic activity of matriptase; see e.g., International Publication Number WO 2010/129609, published 11 Nov. 2010.
  • the same approach of using broad spectrum protease inhibitors such as those described herein, and/or by using a more selective inhibitory agent can be used to identify a MMP specific for the CM of the activatable antibody.
  • the presence of the target can be confirmed using an agent that is specific for the target, e.g., another antibody, or the detectable label can be competed with unlabeled target.
  • unlabeled activatable antibody could be used, with detection by a labeled secondary antibody or more complex detection system.
  • Similar techniques are also useful for in vivo imaging where detection of the fluorescent signal in a subject, e.g., a mammal, including a human, indicates that the disease site contains the target and contains a MMP that is specific for the CM of the activatable antibody.
  • the disclosure provides methods of using the antibodies and/or activatable antibodies in a variety of diagnostic and/or prophylactic indications.
  • the disclosure provides methods of detecting presence or absence of a cleaving agent and a target of interest in a subject or a sample by (i) contacting a subject or sample with an activatable antibody, wherein the activatable antibody comprises a masking moiety (MM), a cleavable moiety (CM) that is cleaved by the cleaving agent, and an antigen binding domain or fragment thereof (AB) that specifically binds the target of interest, wherein the activatable antibody in an uncleaved, non-activated state comprises a structural arrangement from N-terminus to C-terminus as follows: MM-CM-AB or AB-CM-MM; (a) wherein the MM is a peptide that inhibits binding of the AB to the target, and wherein the MM does not have an amino acid sequence of a naturally occurring binding partner of the AB and is not a
  • the activatable antibody is an activatable antibody to which a therapeutic agent is conjugated. In some embodiments, the activatable antibody is not conjugated to an agent. In some embodiments, the activatable antibody comprises a detectable label. In some embodiments, the detectable label is positioned on the AB. In some embodiments, measuring the level of activatable antibody in the subject or sample is accomplished using a secondary reagent that specifically binds to the activated antibody, wherein the reagent comprises a detectable label. In some embodiments, the secondary reagent is an antibody comprising a detectable label.
  • the disclosure also provides methods of detecting presence or absence of a cleaving agent in a subject or a sample by (i) contacting a subject or sample with an activatable antibody in the presence of a target of interest, e.g., the target, wherein the activatable antibody comprises a masking moiety (MM), a cleavable moiety (CM) that is cleaved by the cleaving agent, and an antigen binding domain or fragment thereof (AB) that specifically binds the target of interest, wherein the activatable antibody in an uncleaved, non-activated state comprises a structural arrangement from N-terminus to C-terminus as follows: MM-CM-AB or AB-CM-MM; (a) wherein the MM is a peptide that inhibits binding of the AB to the target, and wherein the MM does not have an amino acid sequence of a naturally occurring binding partner of the AB and is not a modified form of a natural binding partner of the AB; and (b) wherein
  • the activatable antibody is an activatable antibody to which a therapeutic agent is conjugated. In some embodiments, the activatable antibody is not conjugated to an agent. In some embodiments, the activatable antibody comprises a detectable label. In some embodiments, the detectable label is positioned on the AB. In some embodiments, measuring the level of activatable antibody in the subject or sample is accomplished using a secondary reagent that specifically binds to the activated antibody, wherein the reagent comprises a detectable label. In some embodiments, the secondary reagent is an antibody comprising a detectable label.
  • kits for use in methods of detecting presence or absence of a cleaving agent and the target in a subject or a sample
  • the kits include at least an activatable antibody comprises a masking moiety (MM), a cleavable moiety (CM) that is cleaved by the cleaving agent, and an antigen binding domain or fragment thereof (AB) that specifically binds the target of interest
  • the activatable antibody in an uncleaved, non-activated state comprises a structural arrangement from N-terminus to C-terminus as follows: MM-CM-AB or AB-CM-MM; (a) wherein the MM is a peptide that inhibits binding of the AB to the target, and wherein the MM does not have an amino acid sequence of a naturally occurring binding partner of the AB and is not a modified form of a natural binding partner of the AB; and (b) wherein, in an uncleaved, non-activated state, the MM interferes with specific binding of the MM
  • the activatable antibody is an activatable antibody to which a therapeutic agent is conjugated. In some embodiments, the activatable antibody is not conjugated to an agent. In some embodiments, the activatable antibody comprises a detectable label. In some embodiments, the detectable label is positioned on the AB. In some embodiments, measuring the level of activatable antibody in the subject or sample is accomplished using a secondary reagent that specifically binds to the activated antibody, wherein the reagent comprises a detectable label. In some embodiments, the secondary reagent is an antibody comprising a detectable label.
  • the disclosure also provides methods of detecting presence or absence of a cleaving agent in a subject or a sample by (i) contacting a subject or sample with an activatable antibody, wherein the activatable antibody comprises a masking moiety (MM), a cleavable moiety (CM) that is cleaved by the cleaving agent, an antigen binding domain (AB) that specifically binds the target, and a detectable label, wherein the activatable antibody in an uncleaved, non-activated state comprises a structural arrangement from N-terminus to C-terminus as follows: MM-CM-AB or AB-CM-MM; wherein the MM is a peptide that inhibits binding of the AB to the target, and wherein the MM does not have an amino acid sequence of a naturally occurring binding partner of the AB and is not a modified form of a natural binding partner of the AB; wherein, in an uncleaved, non-activated state, the MM interferes with specific binding of the
  • the activatable antibody is an activatable antibody to which a therapeutic agent is conjugated. In some embodiments, the activatable antibody is not conjugated to an agent. In some embodiments, the activatable antibody comprises a detectable label. In some embodiments, the detectable label is positioned on the AB. In some embodiments, measuring the level of activatable antibody in the subject or sample is accomplished using a secondary reagent that specifically binds to the activated antibody, wherein the reagent comprises a detectable label. In some embodiments, the secondary reagent is an antibody comprising a detectable label.
  • kits for use in methods of detecting presence or absence of a cleaving agent and the target in a subject or a sample where the kits include at least an activatable antibody and/or conjugated activatable antibody (e.g., an activatable antibody to which a therapeutic agent is conjugated) described herein for use in contacting a subject or biological sample and means for detecting the level of activated activatable antibody and/or conjugated activatable antibody in the subject or biological sample, wherein a detectable level of activated activatable antibody in the subject or biological sample indicates that the cleaving agent and the target are present in the subject or biological sample and wherein no detectable level of activated activatable antibody in the subject or biological sample indicates that the cleaving agent, the target or both the cleaving agent and the target are absent and/or not sufficiently present in the subject or biological sample, such that the target binding and/or protease cleavage of the activatable antibody cannot be detected in the subject or biological sample.
  • the disclosure also provides methods of detecting presence or absence of a cleaving agent in a subject or a sample by (i) contacting a subject or biological sample with an activatable antibody in the presence of the target, and (ii) measuring a level of activated activatable antibody in the subject or biological sample, wherein a detectable level of activated activatable antibody in the subject or biological sample indicates that the cleaving agent is present in the subject or biological sample and wherein no detectable level of activated activatable antibody in the subject or biological sample indicates that the cleaving agent is absent and/or not sufficiently present in the subject or biological sample at a detectable level, such that protease cleavage of the activatable antibody cannot be detected in the subject or biological sample.
  • Such an activatable antibody includes a masking moiety (MM), a cleavable moiety (CM) that is cleaved by the cleaving agent, and an antigen binding domain or fragment thereof (AB) that specifically binds the target, wherein the activatable antibody in an uncleaved (i.e., non-activated) state comprises a structural arrangement from N-terminus to C-terminus as follows: MM-CM-AB or AB-CM-MM; (a) wherein the MM is a peptide that inhibits binding of the AB to the target, and wherein the MM does not have an amino acid sequence of a naturally occurring binding partner of the AB; and (b) wherein the MM of the activatable antibody in an uncleaved state interferes with specific binding of the AB to the target, and wherein the MM of an activatable antibody in a cleaved (i.e., activated) state does not interfere or compete with specific binding of the AB to the target.
  • the activatable antibody is an activatable antibody to which a therapeutic agent is conjugated. In some embodiments, the activatable antibody is not conjugated to an agent.
  • the detectable label is attached to the masking moiety. In some embodiments, the detectable label is attached to the cleavable moiety N-terminal to the protease cleavage site. In some embodiments, a single antigen binding site of the AB is masked. In some embodiments wherein an antibody of the disclosure has at least two antigen binding sites, at least one antigen binding site is masked and at least one antigen binding site is not masked. In some embodiments, all antigen binding sites are masked. In some embodiments, the measuring step includes use of a secondary reagent comprising a detectable label.
  • kits for use in methods of detecting presence or absence of a cleaving agent and the target in a subject or a sample where the kits include at least an activatable antibody and/or conjugated activatable antibody described herein for use in contacting a subject or biological sample with an activatable antibody in the presence of the target, and measuring a level of activated activatable antibody in the subject or biological sample, wherein a detectable level of activated activatable antibody in the subject or biological sample indicates that the cleaving agent is present in the subject or biological sample and wherein no detectable level of activated activatable antibody in the subject or biological sample indicates that the cleaving agent is absent and/or not sufficiently present in the subject or biological sample at a detectable level, such that protease cleavage of the activatable antibody cannot be detected in the subject or biological sample.
  • Such an activatable antibody includes a masking moiety (MM), a cleavable moiety (CM) that is cleaved by the cleaving agent, and an antigen binding domain or fragment thereof (AB) that specifically binds the target, wherein the activatable antibody in an uncleaved (i.e., non-activated) state comprises a structural arrangement from N-terminus to C-terminus as follows: MM-CM-AB or AB-CM-MM; (a) wherein the MM is a peptide that inhibits binding of the AB to the target, and wherein the MM does not have an amino acid sequence of a naturally occurring binding partner of the AB; and (b) wherein the MM of the activatable antibody in an uncleaved state interferes with specific binding of the AB to the target, and wherein the MM of an activatable antibody in a cleaved (i.e., activated) state does not interfere or compete with specific binding of the AB to the target.
  • the activatable antibody is an activatable antibody to which a therapeutic agent is conjugated. In some embodiments, the activatable antibody is not conjugated to an agent.
  • the detectable label is attached to the masking moiety. In some embodiments, the detectable label is attached to the cleavable moiety N-terminal to the protease cleavage site. In some embodiments, a single antigen binding site of the AB is masked. In some embodiments wherein an antibody of the disclosure has at least two antigen binding sites, at least one antigen binding site is masked and at least one antigen binding site is not masked. In some embodiments, all antigen binding sites are masked. In some embodiments, the measuring step includes use of a secondary reagent comprising a detectable label.
  • kits for use in methods of detecting presence or absence of a cleaving agent in a subject or a sample where the kits include at least an activatable antibody and/or conjugated activatable antibody described herein for use in contacting a subject or biological sample and means for detecting the level of activated activatable antibody and/or conjugated activatable antibody in the subject or biological sample, wherein the activatable antibody includes a detectable label that is positioned on a portion of the activatable antibody that is released following cleavage of the CM, wherein a detectable level of activated activatable antibody in the subject or biological sample indicates that the cleaving agent is absent and/or not sufficiently present in the subject or biological sample such that the target binding and/or protease cleavage of the activatable antibody cannot be detected in the subject or biological sample, and wherein no detectable level of activated activatable antibody in the subject or biological sample indicates that the cleaving agent is present in the subject or biological sample at a detectable level.
  • the disclosure provides methods of detecting presence or absence of a cleaving agent and the target in a subject or a sample by (i) contacting a subject or biological sample with an activatable antibody, wherein the activatable antibody includes a detectable label that is positioned on a portion of the activatable antibody that is released following cleavage of the CM and (ii) measuring a level of activated activatable antibody in the subject or biological sample, wherein a detectable level of activated activatable antibody in the subject or biological sample indicates that the cleaving agent, the target or both the cleaving agent and the target are absent and/or not sufficiently present in the subject or biological sample, such that the target binding and/or protease cleavage of the activatable antibody cannot be detected in the subject or biological sample, and wherein a reduced detectable level of activated activatable antibody in the subject or biological sample indicates that the cleaving agent and the target are present in the subject or biological sample.
  • a reduced level of detectable label is, for example, a reduction of about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95% and/or about 100%.
  • Such an activatable antibody includes a masking moiety (MM), a cleavable moiety (CM) that is cleaved by the cleaving agent, and an antigen binding domain or fragment thereof (AB) that specifically binds the target, wherein the activatable antibody in an uncleaved (i.e., non-activated) state comprises a structural arrangement from N-terminus to C-terminus as follows: MM-CM-AB or AB-CM-MM; (a) wherein the MM is a peptide that inhibits binding of the AB to the target, and wherein the MM does not have an amino acid sequence of a naturally occurring binding partner of the AB; and (b) wherein the MM of the activatable antibody in an uncleaved state interferes with specific binding of the AB to the target, and wherein the MM of an activatable antibody in a cleaved (i.e., activated) state does not interfere or compete with specific binding of the AB to the target.
  • the activatable antibody is an activatable antibody to which a therapeutic agent is conjugated. In some embodiments, the activatable antibody is not conjugated to an agent. In some embodiments, the activatable antibody comprises a detectable label. In some embodiments, the detectable label is positioned on the AB. In some embodiments, measuring the level of activatable antibody in the subject or sample is accomplished using a secondary reagent that specifically binds to the activated antibody, wherein the reagent comprises a detectable label. In some embodiments, the secondary reagent is an antibody comprising a detectable label.
  • kits for use in methods of detecting presence or absence of a cleaving agent and the target in a subject or a sample where the kits include at least an activatable antibody and/or conjugated activatable antibody described herein for use in contacting a subject or biological sample and means for detecting the level of activated activatable antibody and/or conjugated activatable antibody in the subject or biological sample, wherein a detectable level of activated activatable antibody in the subject or biological sample indicates that the cleaving agent, the target or both the cleaving agent and the target are absent and/or not sufficiently present in the subject or biological sample, such that the target binding and/or protease cleavage of the activatable antibody cannot be detected in the subject or biological sample, and wherein a reduced detectable level of activated activatable antibody in the subject or biological sample indicates that the cleaving agent and the target are present in the subject or biological sample.
  • a reduced level of detectable label is, for example, a reduction of about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95% and/or about 100%.
  • the disclosure also provides methods of detecting presence or absence of a cleaving agent in a subject or a sample by (i) contacting a subject or biological sample with an activatable antibody, wherein the activatable antibody includes a detectable label that is positioned on a portion of the activatable antibody that is released following cleavage of the CM; and (ii) measuring a level of detectable label in the subject or biological sample, wherein a detectable level of the detectable label in the subject or biological sample indicates that the cleaving agent is absent and/or not sufficiently present in the subject or biological sample at a detectable level, such that protease cleavage of the activatable antibody cannot be detected in the subject or biological sample, and wherein a reduced detectable level of the detectable label in the subject or biological sample indicates that the cleaving agent is present in the subject or biological sample.
  • a reduced level of detectable label is, for example, a reduction of about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95% and/or about 100%.
  • Such an activatable antibody includes a masking moiety (MM), a cleavable moiety (CM) that is cleaved by the cleaving agent, and an antigen binding domain or fragment thereof (AB) that specifically binds the target, wherein the activatable antibody in an uncleaved (i.e., non-activated) state comprises a structural arrangement from N-terminus to C-terminus as follows: MM-CM-AB or AB-CM-MM; (a) wherein the MM is a peptide that inhibits binding of the AB to the target, and wherein the MM does not have an amino acid sequence of a naturally occurring binding partner of the AB; and (b) wherein the MM of the activatable antibody in an uncleaved state interferes with specific binding of the AB to the target, and wherein the MM of an activatable antibody in a cleaved (i.e., activated) state does not interfere or compete with specific binding of the AB to the target.
  • the activatable antibody is an activatable antibody to which a therapeutic agent is conjugated. In some embodiments, the activatable antibody is not conjugated to an agent. In some embodiments, the activatable antibody comprises a detectable label. In some embodiments, the detectable label is positioned on the AB. In some embodiments, measuring the level of activatable antibody in the subject or sample is accomplished using a secondary reagent that specifically binds to the activated antibody, wherein the reagent comprises a detectable label. In some embodiments, the secondary reagent is an antibody comprising a detectable label.
  • kits for use in methods of detecting presence or absence of a cleaving agent of interest in a subject or a sample where the kits include at least an activatable antibody and/or conjugated activatable antibody described herein for use in contacting a subject or biological sample and means for detecting the level of activated activatable antibody and/or conjugated activatable antibody in the subject or biological sample, wherein the activatable antibody includes a detectable label that is positioned on a portion of the activatable antibody that is released following cleavage of the CM, wherein a detectable level of the detectable label in the subject or biological sample indicates that the cleaving agent, the target, or both the cleaving agent and the target are absent and/or not sufficiently present in the subject or biological sample, such that the target binding and/or protease cleavage of the activatable antibody cannot be detected in the subject or biological sample, and wherein a reduced detectable level of the detectable label in the subject or biological sample indicates that the cleaving agent and
  • a reduced level of detectable label is, for example, a reduction of about 5%, about 10%, about 15%, about 20%, about 25%, about 30%, about 35%, about 40%, about 45%, about 50%, about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95% and/or about 100%.
  • the activatable antibody includes a detectable label.
  • the detectable label includes an imaging agent, a contrasting agent, an enzyme, a fluorescent label, a chromophore, a dye, one or more metal ions, or a ligand-based label.
  • the imaging agent comprises a radioisotope.
  • the radioisotope is indium or technetium.
  • the contrasting agent comprises iodine, gadolinium or iron oxide.
  • the enzyme comprises horseradish peroxidase, alkaline phosphatase, or ⁇ -galactosidase.
  • the fluorescent label comprises yellow fluorescent protein (YFP), cyan fluorescent protein (CFP), green fluorescent protein (GFP), modified red fluorescent protein (mRFP), red fluorescent protein tdimer2 (RFP tdimer2), HCRED, or a europium derivative.
  • the luminescent label comprises an N-methylacrydium derivative.
  • the label comprises an Alexa Fluor® label, such as Alex Fluor® 680 or Alexa Fluor® 750.
  • the ligand-based label comprises biotin, avidin, streptavidin or one or more haptens.
  • the subject is a mammal. In some embodiments of these methods and kits, the subject is a human. In some embodiments, the subject is a non-human mammal, such as a non-human primate, companion animal (e.g., cat, dog, horse), farm animal, work animal, or zoo animal. In some embodiments, the subject is a rodent.
  • a non-human mammal such as a non-human primate, companion animal (e.g., cat, dog, horse), farm animal, work animal, or zoo animal. In some embodiments, the subject is a rodent.
  • the method is an in vivo method. In some embodiments of these methods, the method is an in situ method. In some embodiments of these methods, the method is an ex vivo method. In some embodiments of these methods, the method is an in vitro method.
  • in situ imaging and/or in vivo imaging are useful in methods to identify which patients to treat.
  • the activatable antibodies are used to screen patient samples to identify those patients having the appropriate protease(s) and target(s) at the appropriate location, e.g., at a tumor site.
  • in situ imaging is used to identify or otherwise refine a patient population suitable for treatment with an activatable antibody of the disclosure.
  • patients that test positive for both the target (e.g., the target) and a protease that cleaves the substrate in the cleavable moiety (CM) of the activatable antibody being tested are identified as suitable candidates for treatment with such an activatable antibody comprising such a CM.
  • patients that test negative for either or both of the target (e.g., the target) and the protease that cleaves the substrate in the CM in the activatable antibody being tested using these methods might be identified as suitable candidates for another form of therapy.
  • such patients that test negative with respect to a first activatable antibody can be tested with other activatable antibodies comprising different CMs until a suitable activatable antibody for treatment is identified (e.g., an activatable antibody comprising a CM that is cleaved by the patient at the site of disease).
  • a suitable activatable antibody for treatment e.g., an activatable antibody comprising a CM that is cleaved by the patient at the site of disease.
  • the patient is then administered a therapeutically effective amount of the conjugated activatable antibody for which the patient tested positive.
  • in vivo imaging is used to identify or otherwise refine a patient population suitable for treatment with an activatable antibody of the disclosure.
  • patients that test positive for both the target e.g., the target
  • a protease that cleaves the substrate in the cleavable moiety (CM) of the activatable antibody being tested e.g., accumulate activated antibodies at the disease site
  • CM cleavable moiety
  • patients that test negative might be identified as suitable candidates for another form of therapy.
  • such patients that test negative with respect to a first activatable antibody can be tested with other activatable antibodies comprising different CMs until a suitable activatable antibody for treatment is identified (e.g., an activatable antibody comprising a CM that is cleaved by the patient at the site of disease).
  • a suitable activatable antibody for treatment e.g., an activatable antibody comprising a CM that is cleaved by the patient at the site of disease.
  • the patient is then administered a therapeutically effective amount of the conjugated activatable antibody for which the patient tested positive.
  • the method or kit is used to identify or otherwise refine a patient population suitable for treatment with an activatable antibody of the disclosure. For example, patients that test positive for both the target (e.g., the target) and a protease that cleaves the substrate in the cleavable moiety (CM) of the activatable antibody being tested in these methods are identified as suitable candidates for treatment with such an activatable antibody comprising such a CM. Likewise, patients that test negative for both of the targets (e.g., the target) and the protease that cleaves the substrate in the CM in the activatable antibody being tested using these methods might be identified as suitable candidates for another form of therapy.
  • the target e.g., the target
  • CM cleavable moiety
  • such patients can be tested with other activatable antibodies until a suitable activatable antibody for treatment is identified (e.g., an activatable antibody comprising a CM that is cleaved by the patient at the site of disease).
  • a suitable activatable antibody for treatment e.g., an activatable antibody comprising a CM that is cleaved by the patient at the site of disease.
  • patients that test negative for either of the target are identified as suitable candidates for treatment with such an activatable antibody comprising such a CM.
  • patients that test negative for either of the target are identified as not being suitable candidates for treatment with such an activatable antibody comprising such a CM.
  • such patients can be tested with other activatable antibodies until a suitable activatable antibody for treatment is identified (e.g., an activatable antibody comprising a CM that is cleaved by the patient at the site of disease).
  • the activatable antibody is an activatable antibody to which a therapeutic agent is conjugated.
  • the activatable antibody is not conjugated to an agent.
  • the activatable antibody comprises a detectable label.
  • the detectable label is positioned on the AB.
  • measuring the level of activatable antibody in the subject or sample is accomplished using a secondary reagent that specifically binds to the activated antibody, wherein the reagent comprises a detectable label.
  • the secondary reagent is an antibody comprising a detectable label.
  • a method or kit is used to identify or otherwise refine a patient population suitable for treatment with an anti-the target activatable antibody and/or conjugated activatable antibody (e.g., activatable antibody to which a therapeutic agent is conjugated) of the disclosure, followed by treatment by administering that activatable antibody and/or conjugated activatable antibody to a subject in need thereof.
  • an anti-the target activatable antibody and/or conjugated activatable antibody e.g., activatable antibody to which a therapeutic agent is conjugated
  • patients that test positive for both the targets (e.g., the target) and a protease that cleaves the substrate in the cleavable moiety (CM) of the activatable antibody and/or conjugated activatable antibody being tested in these methods are identified as suitable candidates for treatment with such antibody and/or such a conjugated activatable antibody comprising such a CM, and the patient is then administered a therapeutically effective amount of the activatable antibody and/or conjugated activatable antibody that was tested.
  • patients that test negative for either or both of the target (e.g., the target) and the protease that cleaves the substrate in the CM in the activatable antibody being tested using these methods might be identified as suitable candidates for another form of therapy.
  • such patients can be tested with other antibody and/or conjugated activatable antibody until a suitable antibody and/or conjugated activatable antibody for treatment is identified (e.g., an activatable antibody and/or conjugated activatable antibody comprising a CM that is cleaved by the patient at the site of disease).
  • a suitable antibody and/or conjugated activatable antibody for treatment e.g., an activatable antibody and/or conjugated activatable antibody comprising a CM that is cleaved by the patient at the site of disease.
  • the patient is then administered a therapeutically effective amount of the activatable antibody and/or conjugated for which the patient tested positive.
  • the MM is a peptide having a length from about 4 to 40 amino acids.
  • the activatable antibody comprises a linker peptide, wherein the linker peptide is positioned between the MM and the CM. In some embodiments of these methods and kits, the activatable antibody comprises a linker peptide, where the linker peptide is positioned between the AB and the CM.
  • the activatable antibody comprises a first linker peptide (L1) and a second linker peptide (L2), wherein the first linker peptide is positioned between the MM and the CM and the second linker peptide is positioned between the AB and the CM.
  • each of L1 and L2 is a peptide of about 1 to 20 amino acids in length, and wherein each of L1 and L2 need not be the same linker.
  • one or both of L1 and L2 comprises a glycine-serine polymer.
  • At least one of L1 and L2 comprises an amino acid sequence selected from the group consisting of (GS)n, (GSGGS)n (SEQ ID NO: 1) and (GGGS)n (SEQ ID NO: 2), where n is an integer of at least one.
  • at least one of L1 and L2 comprises an amino acid sequence having the formula (GGS)n, where n is an integer of at least one.
  • At least one of L1 and L2 comprises an amino acid sequence selected from the group consisting of Gly-Gly-Ser-Gly (SEQ ID NO: 3), Gly-Gly-Ser-Gly-Gly (SEQ ID NO: 4), Gly-Ser-Gly-Ser-Gly (SEQ ID NO: 5), Gly-Ser-Gly-Gly-Gly (SEQ ID NO: 6), Gly-Gly-Gly-Ser-Gly (SEQ ID NO: 7), and Gly-Ser-Ser-Ser-Gly (SEQ ID NO: 8).
  • the AB comprises an antibody or antibody fragment sequence selected from the cross-reactive antibody sequences presented herein. In some embodiments of these methods and kits, the AB comprises a Fab fragment, a scFv or a single chain antibody (scAb).
  • the cleaving agent is a protease that is co-localized in the subject or sample with the target and the CM is a polypeptide that functions as a substrate for the protease, wherein the protease cleaves the CM in the activatable antibody when the activatable antibody is exposed to the protease.
  • the CM is a polypeptide of up to 15 amino acids in length.
  • the CM is coupled to the N-terminus of the AB.
  • the CM is coupled to the C-terminus of the AB.
  • the CM is coupled to the N-terminus of a VL chain of the AB.
  • activatable antibodies and/or conjugated activatable antibodies of the disclosure are used in diagnostic and prophylactic formulations.
  • an activatable antibody is administered to patients that are at risk of developing one or more of the aforementioned inflammation, inflammatory disorders, cancer or other disorders.
  • a patient's or organ's predisposition to one or more of the aforementioned disorders can be determined using genotypic, serological or biochemical markers.
  • an activatable antibody and/or conjugated activatable antibodies is administered to human individuals diagnosed with a clinical indication associated with one or more of the aforementioned disorders. Upon diagnosis, an activatable antibody and/or conjugated activatable antibodies is administered to mitigate or reverse the effects of the clinical indication.
  • Activatable antibodies and/or conjugated activatable antibodies of the disclosure are also useful in the detection of the target in patient samples and accordingly are useful as diagnostics.
  • the activatable antibodies and/or conjugated activatable antibodies of the disclosure are used in in vitro assays, e.g., ELISA, to detect target levels in a patient sample.
  • an activatable antibody of the disclosure is immobilized on a solid support (e.g., the well(s) of a microtiter plate).
  • the immobilized activatable antibody serves as a capture antibody for any target that may be present in a test sample.
  • the solid support Prior to contacting the immobilized antibody with a patient sample, the solid support is rinsed and treated with a blocking agent such as milk protein or albumin to prevent nonspecific adsorption of the analyte.
  • the wells are treated with a test sample suspected of containing the antigen, or with a solution containing a standard amount of the antigen.
  • a sample is, e.g., a serum sample from a subject suspected of having levels of circulating antigen considered to be diagnostic of a pathology.
  • the solid support is treated with a second antibody that is detectably labeled.
  • the labeled second antibody serves as a detecting antibody. The level of detectable label is measured, and the concentration of target antigen in the test sample is determined by comparison with a standard curve developed from the standard samples.
  • Activatable antibodies and/or conjugated activatable antibodies can also be used in diagnostic and/or imaging methods.
  • such methods are in vitro methods.
  • such methods are in vivo methods.
  • such methods are in situ methods.
  • such methods are ex vivo methods.
  • activatable antibodies having an enzymatically cleavable CM can be used to detect the presence or absence of an enzyme that is capable of cleaving the CM.
  • Such activatable antibodies can be used in diagnostics, which can include in vivo detection (e.g., qualitative or quantitative) of enzyme activity (or, in some embodiments, an environment of increased reduction potential such as that which can provide for reduction of a disulfide bond) through measured accumulation of activated antibodies (i.e., antibodies resulting from cleavage of an activatable antibody) in a given cell or tissue of a given host organism.
  • activated antibodies i.e., antibodies resulting from cleavage of an activatable antibody
  • Such accumulation of activated antibodies indicates not only that the tissue expresses enzymatic activity (or an increased reduction potential depending on the nature of the CM) but also that the tissue expresses target to which the activated antibody binds.
  • the CM can be selected to be a protease substrate for a protease found at the site of a tumor, at the site of a viral or bacterial infection at a biologically confined site (e.g., such as in an abscess, in an organ, and the like), and the like.
  • the AB can be one that binds a target antigen.
  • a detectable label e.g., a fluorescent label or radioactive label or radiotracer
  • Suitable detectable labels are discussed in the context of the above screening methods and additional specific examples are provided below.
  • activatable antibodies will exhibit an increased rate of binding to disease tissue relative to tissues where the CM specific enzyme is not present at a detectable level or is present at a lower level than in disease tissue or is inactive (e.g., in zymogen form or in complex with an inhibitor). Since small proteins and peptides are rapidly cleared from the blood by the renal filtration system, and because the enzyme specific for the CM is not present at a detectable level (or is present at lower levels in non-disease tissues or is present in inactive conformation), accumulation of activated antibodies in the disease tissue is enhanced relative to non-disease tissues.
  • activatable antibodies can be used to detect the presence or absence of a cleaving agent in a sample.
  • the activatable antibodies can be used to detect (either qualitatively or quantitatively) the presence of an enzyme in the sample.
  • the activatable antibodies can be used to detect (either qualitatively or quantitatively) the presence of reducing conditions in a sample.
  • the activatable antibodies can be detectably labeled, and can be bound to a support (e.g., a solid support, such as a slide or bead).
  • the detectable label can be positioned on a portion of the activatable antibody that is not released following cleavage, for example, the detectable label can be a quenched fluorescent label or other label that is not detectable until cleavage has occurred.
  • the assay can be conducted by, for example, contacting the immobilized, detectably labeled activatable antibodies with a sample suspected of containing an enzyme and/or reducing agent for a time sufficient for cleavage to occur, then washing to remove excess sample and contaminants.
  • the presence or absence of the cleaving agent (e.g., enzyme or reducing agent) in the sample is then assessed by a change in detectable signal of the activatable antibodies prior to contacting with the sample e.g., the presence of and/or an increase in detectable signal due to cleavage of the activatable antibody by the cleaving agent in the sample.
  • the cleaving agent e.g., enzyme or reducing agent
  • Such detection methods can be adapted to also provide for detection of the presence or absence of a target that is capable of binding the AB of the activatable antibodies when cleaved.
  • the assays can be adapted to assess the presence or absence of a cleaving agent and the presence or absence of a target of interest.
  • the presence or absence of the cleaving agent can be detected by the presence of and/or an increase in detectable label of the activatable antibodies as described above, and the presence or absence of the target can be detected by detection of a target-AB complex e.g., by use of a detectably labeled anti-target antibody.
  • Activatable antibodies are also useful in in situ imaging for the validation of activatable antibody activation, e.g., by protease cleavage, and binding to a particular target.
  • In situ imaging is a technique that enables localization of proteolytic activity and target in biological samples such as cell cultures or tissue sections. Using this technique, it is possible to confirm both binding to a given target and proteolytic activity based on the presence of a detectable label (e.g., a fluorescent label).
  • a detectable label e.g., a fluorescent label
  • an activatable antibody is labeled with a detectable label.
  • the detectable label may be a fluorescent dye, (e.g. Fluorescein Isothiocyanate (FITC), Rhodamine Isothiocyanate (TRITC), a near infrared (NIR) dye (e.g., Qdot® nanocrystals), a colloidal metal, a hapten, a radioactive marker, biotin and an amplification reagent such as streptavidin, or an enzyme (e.g. horseradish peroxidase or alkaline phosphatase).
  • FITC Fluorescein Isothiocyanate
  • TRITC Rhodamine Isothiocyanate
  • NIR near infrared
  • colloidal metal e.g., a colloidal metal, a hapten, a radioactive marker, biotin and an amplification reagent such as streptavidin, or an enzyme (e.g. horseradish
  • Detection of the label in a sample that has been incubated with the labeled, activatable antibody indicates that the sample contains the target and contains a protease that is specific for the CM of the activatable antibody.
  • the presence of the protease can be confirmed using broad spectrum protease inhibitors such as those described herein, and/or by using an agent that is specific for the protease, for example, an antibody such as A11, which is specific for the protease matriptase (MT-SP1) and inhibits the proteolytic activity of matriptase; see e.g., International Publication Number WO 2010/129609, published 11 Nov. 2010.
  • the same approach of using broad spectrum protease inhibitors such as those described herein, and/or by using a more selective inhibitory agent can be used to identify a protease or class of proteases specific for the CM of the activatable antibody.
  • the presence of the target can be confirmed using an agent that is specific for the target, e.g., another antibody, or the detectable label can be competed with unlabeled target.
  • unlabeled activatable antibody could be used, with detection by a labeled secondary antibody or more complex detection system.
  • Similar techniques are also useful for in vivo imaging where detection of the fluorescent signal in a subject, e.g., a mammal, including a human, indicates that the disease site contains the target and contains a protease that is specific for the CM of the activatable antibody.
  • in situ imaging and/or in vivo imaging are useful in methods to identify which patients to treat.
  • the activatable antibodies are used to screen patient samples to identify those patients having the appropriate protease(s) and target(s) at the appropriate location, e.g., at a tumor site.
  • in situ imaging is used to identify or otherwise refine a patient population suitable for treatment with an activatable antibody of the disclosure.
  • patients that test positive for both the target and a protease that cleaves the substrate in the cleavable moiety (CM) of the activatable antibody being tested e.g., accumulate activated antibodies at the disease site
  • CM cleavable moiety
  • patients that test negative for either or both of the target and the protease that cleaves the substrate in the CM in the activatable antibody being tested using these methods are identified as suitable candidates for another form of therapy (i.e., not suitable for treatment with the activatable antibody being tested).
  • such patients that test negative with respect to a first activatable antibody can be tested with other activatable antibodies comprising different CMs until a suitable activatable antibody for treatment is identified (e.g., an activatable antibody comprising a CM that is cleaved by the patient at the site of disease).
  • a suitable activatable antibody for treatment e.g., an activatable antibody comprising a CM that is cleaved by the patient at the site of disease.
  • in vivo imaging is used to identify or otherwise refine a patient population suitable for treatment with an activatable antibody of the disclosure.
  • patients that test positive for both the target and a protease that cleaves the substrate in the cleavable moiety (CM) of the activatable antibody being tested e.g., accumulate activated antibodies at the disease site
  • CM cleavable moiety
  • patients that test negative are identified as suitable candidates for another form of therapy (i.e., not suitable for treatment with the activatable antibody being tested).
  • such patients that test negative with respect to a first activatable antibody can be tested with other activatable antibodies comprising different CMs until a suitable activatable antibody for treatment is identified (e.g., an activatable antibody comprising a CM that is cleaved by the patient at the site of disease).
  • a suitable activatable antibody for treatment e.g., an activatable antibody comprising a CM that is cleaved by the patient at the site of disease.
  • compositions suitable for administration can be incorporated into pharmaceutical compositions suitable for administration.
  • Such compositions typically comprise the conjugated antibody, activatable antibody and/or conjugated activatable antibody and a pharmaceutically acceptable carrier.
  • pharmaceutically acceptable carrier is intended to include any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like, compatible with pharmaceutical administration. Suitable carriers are described in the most recent edition of Remington's Pharmaceutical Sciences, a standard reference text in the field, which is incorporated herein by reference.
  • Suitable examples of such carriers or diluents include, but are not limited to, water, saline, ringer's solutions, dextrose solution, and 5% human serum albumin. Liposomes and non-aqueous vehicles such as fixed oils may also be used.
  • the use of such media and agents for pharmaceutically active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the active compound, use thereof in the compositions is contemplated. Supplementary active compounds can also be incorporated into the compositions.
  • a pharmaceutical composition of the disclosure is formulated to be compatible with its intended route of administration.
  • routes of administration include parenteral, e.g., intravenous, intradermal, subcutaneous, oral (e.g., inhalation), transdermal (i.e., topical), transmucosal, and rectal administration.
  • Solutions or suspensions used for parenteral, intradermal, or subcutaneous application can include the following components: a sterile diluent such as water for injection, saline solution, fixed oils, polyethylene glycols, glycerine, propylene glycol or other synthetic solvents; antibacterial agents such as benzyl alcohol or methyl parabens; antioxidants such as ascorbic acid or sodium bisulfite; chelating agents such as ethylenediaminetetraacetic acid (EDTA); buffers such as acetates, citrates or phosphates, and agents for the adjustment of tonicity such as sodium chloride or dextrose.
  • the pH can be adjusted with acids or bases, such as hydrochloric acid or sodium hydroxide.
  • the parenteral preparation can be enclosed in ampoules, disposable syringes or multiple dose vials made of glass or plastic.
  • compositions suitable for injectable use include sterile aqueous solutions (where water soluble) or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersion.
  • suitable carriers include physiological saline, bacteriostatic water, Cremophor ELTM (BASF, Parsippany, N.J.) or phosphate buffered saline (PBS).
  • the composition must be sterile and should be fluid to the extent that easy syringeability exists. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms such as bacteria and fungi.
  • the carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), and suitable mixtures thereof.
  • the proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants.
  • Prevention of the action of microorganisms can be achieved by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, ascorbic acid, thimerosal, and the like.
  • isotonic agents for example, sugars, polyalcohols such as manitol, sorbitol, sodium chloride in the composition.
  • Prolonged absorption of the injectable compositions can be brought about by including in the composition an agent that delays absorption, for example, aluminum monostearate and gelatin.
  • Sterile injectable solutions can be prepared by incorporating the active compound in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, followed by filtered sterilization.
  • dispersions are prepared by incorporating the active compound into a sterile vehicle that contains a basic dispersion medium and the required other ingredients from those enumerated above.
  • methods of preparation are vacuum drying and freeze-drying that yields a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.
  • Oral compositions generally include an inert diluent or an edible carrier. They can be enclosed in gelatin capsules or compressed into tablets. For the purpose of oral therapeutic administration, the active compound can be incorporated with excipients and used in the form of tablets, troches, or capsules. Oral compositions can also be prepared using a fluid carrier for use as a mouthwash, wherein the compound in the fluid carrier is applied orally and swished and expectorated or swallowed. Pharmaceutically compatible binding agents, and/or adjuvant materials can be included as part of the composition.
  • the tablets, pills, capsules, troches and the like can contain any of the following ingredients, or compounds of a similar nature: a binder such as microcrystalline cellulose, gum tragacanth or gelatin; an excipient such as starch or lactose, a disintegrating agent such as alginic acid, Primogel, or corn starch; a lubricant such as magnesium stearate or Sterotes; a glidant such as colloidal silicon dioxide; a sweetening agent such as sucrose or saccharin; or a flavoring agent such as peppermint, methyl salicylate, or orange flavoring.
  • a binder such as microcrystalline cellulose, gum tragacanth or gelatin
  • an excipient such as starch or lactose, a disintegrating agent such as alginic acid, Primogel, or corn starch
  • a lubricant such as magnesium stearate or Sterotes
  • a glidant such as colloidal silicon dioxide
  • the compounds are delivered in the form of an aerosol spray from pressured container or dispenser that contains a suitable propellant, e.g., a gas such as carbon dioxide, or a nebulizer.
  • a suitable propellant e.g., a gas such as carbon dioxide, or a nebulizer.
  • Systemic administration can also be by transmucosal or transdermal means.
  • penetrants appropriate to the barrier to be permeated are used in the formulation.
  • penetrants are generally known in the art, and include, for example, for transmucosal administration, detergents, bile salts, and fusidic acid derivatives.
  • Transmucosal administration can be accomplished through the use of nasal sprays or suppositories.
  • the active compounds are formulated into ointments, salves, gels, or creams as generally known in the art.
  • the compounds can also be prepared in the form of suppositories (e.g., with conventional suppository bases such as cocoa butter and other glycerides) or retention enemas for rectal delivery.
  • suppositories e.g., with conventional suppository bases such as cocoa butter and other glycerides
  • retention enemas for rectal delivery.
  • the active compounds are prepared with carriers that will protect the compound against rapid elimination from the body, such as a controlled release formulation, including implants and microencapsulated delivery systems.
  • a controlled release formulation including implants and microencapsulated delivery systems.
  • Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid. Methods for preparation of such formulations will be apparent to those skilled in the art.
  • the materials can also be obtained commercially from Alza Corporation and Nova Pharmaceuticals, Inc.
  • Liposomal suspensions (including liposomes targeted to infected cells with monoclonal antibodies to viral antigens) can also be used as pharmaceutically acceptable carriers. These can be prepared according to methods known to those skilled in the art, for example, as described in U.S. Pat. No. 4,522,811.
  • Dosage unit form refers to physically discrete units suited as unitary dosages for the subject to be treated; each unit containing a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier.
  • the specification for the dosage unit forms of the disclosure are dictated by and directly dependent on the unique characteristics of the active compound and the particular therapeutic effect to be achieved, and the limitations inherent in the art of compounding such an active compound for the treatment of individuals.
  • compositions can be included in a container, pack, or dispenser together with instructions for administration.
  • This Example demonstrates the ability of synovial fluid samples to cleave MMP substrate sequences of the disclosure.
  • the MMP cleavable sequences were tested in the context of an activatable antibody construct comprising a masking moiety linked to an anti-IL-6R antibody sequence via a linker region that includes the MMP cleavable sequence being evaluated.
  • MMP-cleavable activatable antibodies were incubated with synovial fluid:
  • the extent of activatable antibody activation was determined by an ELISA format that measured the ability of the activatable antibody, following incubation in synovial fluid, to bind to human IL6R as compared to the binding of anti-IL6R parental antibody to IL6R. Briefly, Nunc Maxisorp plates were coated overnight at 4° C. with 100 ⁇ l/well (microliters/well) of a 500-ng/mL solution of human IL6R (R and D Systems, Cat No. 227-SR/CF) in PBS, pH 7.4. Plates were washed 3 times with PBST (PBS, pH 7.4, 0.05% Tween-20).
  • PBST PBS, pH 7.4, 0.05% Tween-20
  • IL6R-coated plates were washed 3 times with PBST (PBS, pH 7.4, 0.05% Tween-20).
  • PBST PBS, pH 7.4, 0.05% Tween-20
  • the plates were incubated 1 hour at room temperature, and then washed 3 times with PBST (PBS, pH 7.4, 0.05% Tween-20).
  • Table 6 provides the results of this experiment.
  • the data indicate that anti-IL6R activatable antibodies comprising the substrates in Table 6 are cleaved by at least some synovial fluid samples (SyF) obtained from RA patients.
  • This Example demonstrates the ability of an activatable anti-EGFR antibody that contains a masking moiety comprising the amino acid sequence CISPRGCPDGPYVMY (SEQ ID NO: 160), a cleavage moiety comprising the MMP14 substrate 520 (also referred to herein as MN520) ISSGLLSS (SEQ ID NO: 14), and the heavy chain (SEQ ID NO: 56) and light chain (SEQ ID NO: 59) of the anti-EGFR antibody C225v5, where the entire activatable antibody construct is referred to herein as Pb-MN520, to inhibit tumor growth in the H292 xenograft lung cancer model.
  • the configuration of the light chain of the activatable antibody was masking moiety—MMP substrate—light chain of C225v5.
  • FIG. 1A is a graph depicting the effects seen in H292 xenograft tumor-bearing mice that were treated using Pb-520 (12.5 mg/kg, solid blue line) and IVIG (12.5 mg/kg, green dashed line) dosed at different times. Data are presented as mean tumor volume ⁇ SEM.
  • FIG. 1B is a graph depicting systemic stability of the Pb-520 activatable antibody in H292 tumor bearing mice. Blood samples were taken through retro-orbital bleeds at Day 7 and the circulating stability of substrate 520 was determined by analysis of IgG pull-downs with capillary electrophoresis (GXII; Caliper LifeSciences). Concentrations of cleaved and uncleaved light chain were determined using LabChip GX software (Caliper LifeSciences).
  • SA-PE Streptavidin-conjugated phycoerythrin
  • Human MMP9 Research & Diagnostics Systems, Inc.
  • Human MMP14 Research & Diagnostics Systems, Inc.
  • Human Plasmin Human Plasmin (Haematologic Technologies Inc.) was used without modifications.
  • Human tPA Human tPA (Molecular Innovations) was used without modifications.
  • YPet fused to the SH3 domain of Mona was produced at CytomX Therapeutics and used without modifications.
  • MMP14 Buffer HCM 50 mM HEPES (pH 6.8), 10 mM CaCl 2 , 0.5 mM MgCl 2
  • MMP9 Buffer TCNB 50 mM Tris-HCl, 10 mM CaCl 2 , 150 mM NaCl, 0.05% (w/v) Brij-35, pH 7.5
  • Plasmin Buffer 50 mM Tris-Cl pH 7.5, 100 mM NaCl, 0.01% Tween20 and 1 mM EDTA was used.
  • TBST 50 mM Tris-HCl, 150 mM NaCl, 0.05% Tween20, pH 7.4 was used.
  • coli MC1061 (Casadaban et al., JMB 138(2):179-207 (1980) was used. All bacterial growth was performed at 37° C. with vigorous shaking in Luria-Bertani broth (LB) supplemented with 34 ⁇ g/mL chloramphenicol, unless another antibiotic is specified.
  • LB Luria-Bertani broth
  • MMP9 protease cleavage assays cultures were induced for 45 minutes to 1 hour.
  • the reaction buffer for MMP9 was TCNB.
  • Assays for MMP9 hydrolysis were performed after fresh cells were incubated with 5 nM-25 nM MMP9 for 1 hr. Background hydrolysis of the regions flanking the substrate site (using platform eCLiPS3.0-NSUB_SP described in PCT patent application PCT/US13/54378, filed Aug. 9, 2013, which was published as International Publication No. WO 2014/026136 on 13 Feb. 2014, the contents of which are hereby incorporated by reference in their entirety) was measured under each reaction condition to ensure that hydrolysis occurred in the designated substrate region.
  • MMP14 protease cleavage assays cultures were induced for 45 minutes to 1 hour.
  • the reaction buffer for MMP14 was HCM.
  • Assays for MMP14 hydrolysis were performed after reactions with 3 nM-250 nM MMP14 or 1 hr. Background hydrolysis of the regions flanking the substrate site (using platform eCLiPS3.0-NSUB_SP described herein) was measured under each reaction condition to ensure that hydrolysis occurred in the designated substrate region.
  • reaction buffer for plasmin is 50 mM Tris-HCl pH 7.5 supplemented with 100 mM NaCl, 0.01% Tween20 and 1 mM EDTA.
  • Assays for plasmin hydrolysis are performed after reactions with plasmin for 1 hr.
  • platform eCLiPS3.0-NSUB_SP is used; cultures are induced for 45 minutes to 1 hr.
  • the reaction buffer for tPA is TBST.
  • Assays for tPA hydrolysis are performed after reactions with tPA for 1 hr.
  • Streptavidin conjugated phycoerythrin was used for labeling streptavidin binding affinity ligand on the N-termini of CPX.
  • Fluorescent protein YPet fused to the SH3 domain of Mona was used for labeling the MONA binding affinity ligand on the C-termini of CPX.
  • the cells were incubated for 30 min at 4° C. with SAPE (20 ⁇ g/mL) or YPet-MONA (50 nM). For the described example below 30 min incubation was used.
  • C product conversion
  • t time
  • p protease concentration
  • Substrates were submitted to Ion TorrentTM sequencing (see, e.g., Rothenberg, J M, Nature 475, 348-352).
  • Raw Ion Torrent reads were cropped by invariant vector sequences to obtain just the variable peptide insert. Insert sequences were translated, and sequences with stop codons were excluded from further analysis. The frequency of each sequence was obtained by number of times observed out of all viable peptide reads observed. Enrichment of sequences was obtained by comparison of observed frequency of each sequence post selection to the frequency of each sequence pre-selection. Individual sequences were identified and isolated from these data, and sequences were aligned in CLC main lab (CLC Main Workbench 6.6.2, available online).
  • the alignment file was imported to Jalview (see, e.g., Waterhouse, A. M., et al., 2009, Bioinformatics 9, 1189-1191), and an average distance tree was assembled using the BLOSUM62 algorithm (S Henikoff S et al., 1992, Proc Natl Acad Sci USA. 89, 10915-10919).
  • the restricted group of sequences includes members of the cluster closest to the sequence of interest.
  • the extended group of sequences includes the restricted group of sequences plus members of the branch that shares the closest common ancestor (where applicable).
  • FIGS. 2A and 2B show cleavage of pool SMP87 by MMP9 at 5 nM in TCNB buffer.
  • FIGS. 3A and 3B show cleavage of a substrate comprising amino acid sequence VAGRSMRP (SEQ ID NO: 484) by 5 nM MMP9 in TBST.
  • FIG. 4 shows the correlation between frequency of particular cleavage moieties (Copy Number) and their abilities to be cleaved by MMP9 (MMP9 k cat /K M M ⁇ 1 s ⁇ 1 ).
  • FIGS. 5A and 5B show cleavage of pool SMP39 by MMP14 at 60 nM in HCM buffer.
  • FIGS. 6A and 6B show cleavage of a substrate comprising amino acid sequence QNQALRMA (SEQ ID NO: 15) by 30 nM MMP14 in HCM buffer.
  • This Example demonstrates the in vitro activity of substrates of the disclosure when they are incorporated into activatable antibodies.
  • MMP9 protease digests were performed in TCNB, 50 mM Tris-HCl, 10 mM CaCl 2 , 150 mM NaCl, 0.05% (w/v) Brij-35, pH 7.5.
  • MMP14 digests were performed in 50 mM HEPES (pH 6.8), 10 mM CaCl 2 , 0.5 mM MgCl 2 . Varying concentrations of active site titrated MMP9 or MMP14 were combined with a fixed activatable antibody concentration to maintain a substrate to protease ratio of at least 50.
  • C product conversion
  • t time (s)
  • p protease concentration (M)
  • Resultant activatable antibodies comprising substrates selected for cleavage by MMP14 tested for cleavage by MMP14 had k cat /K M values ranging from about 400 to 60,000 M ⁇ 1 s ⁇ 1 for MMP14.
  • This Example demonstrates the in vivo stability of substrates of the disclosure when they are incorporated into activatable antibodies and injected into mice.
  • mice received a single IP dose of each activatable antibody at 12.5 mg/kg on Day 0. Mice were euthanized on day 4 ( ⁇ 96 h post-dose) by CO 2 asphyxiation, and blood was collected immediately as plasma-EDTA and stored at ⁇ 80° C.
  • Activatable antibodies were purified from plasma by anti-human IgG immunoprecipitation using magnetic beads. Eluted activatable antibodies were prepared for analysis by capillary electrophoresis as described in the k cat /K M section. Briefly, 5 ⁇ l of eluted IgG was added to 7 ⁇ l Protein Express Sample Buffer with 2-mercaptoethanol. Quantification of circulating stability was identical to quantification of product conversion.
  • activatable antibodies comprising substrates of the disclosure selected for cleavage by MMP14, seven demonstrated less than 20% cleavage in the collected plasma samples. Of seven activatable antibodies comprising substrates of the disclosure selected for cleavage by MMP9, four demonstrated no more than 20% cleavage in the collected plasma samples.
  • Human MMP9 (catalog no. 911-MP, Research & Diagnostics Systems, Inc.) was activated following the supplied protocol and used without modifications.
  • Human MMP14 (catalog no. 918-M), Research & Diagnostics Systems, Inc.) was activated following the supplied protocol and used without modifications.
  • Human Plasmin (catalog no. HCPM-0140, Haematologic Technologies Inc.) was used without modifications.
  • Anti-EE monoclonal antibody (Covance, Princeton, N.J.) was labeled with Alexa 647 (Life Sciences) and used with no other modifications (named EE647).
  • coli MC1061 or MC1061 derived strains were used for all experiments (Casadaban et al., JMB 138(2):179-207 (1980)). All bacterial growth was performed at 37° C. with vigorous shaking in Luria-Bertani broth (LB) supplemented with 34 ⁇ g/mL chloramphenicol (cm), unless another antibiotic is specified.
  • LB Luria-Bertani broth
  • chloramphenicol cm
  • the amino acid sequence of CYTX-DP-XXXXXXXXXXX display platform also including its signal peptide, i.e., SP-CYTX-DP-XXXXXXXX display platform (SEQ ID NO: 513) is shown in FIG. 7B .
  • MMP9 protease cleavage assays cultures were induced for 45 minutes.
  • the reaction buffer for MMP9 was 50 mM Tris-HCl, pH 7.4, supplemented with 150 mM NaCl, 10 mM CaCl 2 and 0.05% (w/v) Brij-35.
  • Assays for MMP9 hydrolysis were performed after cleavage with 5 nM-150 nM MMP9 for 1 hour. Background hydrolysis of the regions flanking the substrate site (using, e.g., CYTX-DP-NSUB, a display platform in which the “Substrate” is non-cleavable linker GGGSGGGS) was measured under each reaction condition to ensure that hydrolysis occurred in the designated substrate region.
  • MMP14 protease cleavage assays cultures were induced for 45 minutes.
  • the reaction buffer for MMP14 was 50 mM HEPES, pH 6.8, supplemented with 10 mM CaCl 2 and 0.5 mM MgCl 2 .
  • Assays for MMP14 hydrolysis were performed after cleavage with 5 nM-150 nM MMP14 for 1 hr. Background hydrolysis of the regions flanking the substrate site (using, e.g., CYTX-DP-NSUB) was measured under each reaction condition to ensure that hydrolysis occurred in the designated substrate region.
  • plasmin stability assays cultures were induced for 45 minutes.
  • the reaction buffer for plasmin was 50 mM Tris-HCl pH 7.4, supplemented with 100 mM NaCl, 0.01% Tween20 and 1 mM EDTA.
  • Assays for plasmin hydrolysis were performed after cleavage with 500 pM plasmin for 1 hr. Background hydrolysis of the regions flanking the substrate site (using, e.g., CYTX-DP-NSUB) was measured under each reaction condition to ensure that hydrolysis occurred in the designated substrate region.
  • Alexa-647 conjugated anti-EE antibody (EE647) was used for labeling EE binding affinity ligand on the N-termini of the CYTX-DP display platform.
  • Alexa-647 conjugated anti-His antibody (His647) was used for labeling the 8His binding affinity ligand on the C-termini of the CYTX-DP display platform.
  • the cells were incubated for 1 hour at 4° C. with EE647 (20 ⁇ g/mL) or His647 (2 ⁇ g/mL). For the example described below, a 1-hour incubation was used.
  • This Example demonstrates the ability of substrates of the embodiments to be cleaved by MMP but not by plasmin.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Immunology (AREA)
  • Medicinal Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Biochemistry (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biophysics (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Microbiology (AREA)
  • Mycology (AREA)
  • Epidemiology (AREA)
  • Peptides Or Proteins (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
US14/497,089 2013-09-25 2014-09-25 Matrix Metalloproteinase Substrates And Other Cleavable Moieties And Methods Of Use Thereof Abandoned US20150087810A1 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
US14/497,089 US20150087810A1 (en) 2013-09-25 2014-09-25 Matrix Metalloproteinase Substrates And Other Cleavable Moieties And Methods Of Use Thereof
US16/179,670 US11814410B2 (en) 2013-09-25 2018-11-02 Matrix metalloproteinase substrates and other cleavable moieties and methods of use thereof
US18/474,103 US20240150478A1 (en) 2013-09-25 2023-09-25 Matrix metalloproteinase substrates and other cleavable moieties and methods of use thereof
US18/474,134 US20240084022A1 (en) 2013-09-25 2023-09-25 Matrix metalloproteinase substrates and other cleavable moieties and methods of use thereof

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201361882377P 2013-09-25 2013-09-25
US201461971332P 2014-03-27 2014-03-27
US14/497,089 US20150087810A1 (en) 2013-09-25 2014-09-25 Matrix Metalloproteinase Substrates And Other Cleavable Moieties And Methods Of Use Thereof

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US16/179,670 Division US11814410B2 (en) 2013-09-25 2018-11-02 Matrix metalloproteinase substrates and other cleavable moieties and methods of use thereof

Publications (1)

Publication Number Publication Date
US20150087810A1 true US20150087810A1 (en) 2015-03-26

Family

ID=51691175

Family Applications (4)

Application Number Title Priority Date Filing Date
US14/497,089 Abandoned US20150087810A1 (en) 2013-09-25 2014-09-25 Matrix Metalloproteinase Substrates And Other Cleavable Moieties And Methods Of Use Thereof
US16/179,670 Active US11814410B2 (en) 2013-09-25 2018-11-02 Matrix metalloproteinase substrates and other cleavable moieties and methods of use thereof
US18/474,103 Pending US20240150478A1 (en) 2013-09-25 2023-09-25 Matrix metalloproteinase substrates and other cleavable moieties and methods of use thereof
US18/474,134 Pending US20240084022A1 (en) 2013-09-25 2023-09-25 Matrix metalloproteinase substrates and other cleavable moieties and methods of use thereof

Family Applications After (3)

Application Number Title Priority Date Filing Date
US16/179,670 Active US11814410B2 (en) 2013-09-25 2018-11-02 Matrix metalloproteinase substrates and other cleavable moieties and methods of use thereof
US18/474,103 Pending US20240150478A1 (en) 2013-09-25 2023-09-25 Matrix metalloproteinase substrates and other cleavable moieties and methods of use thereof
US18/474,134 Pending US20240084022A1 (en) 2013-09-25 2023-09-25 Matrix metalloproteinase substrates and other cleavable moieties and methods of use thereof

Country Status (12)

Country Link
US (4) US20150087810A1 (ru)
EP (1) EP3049111A2 (ru)
JP (4) JP6915987B2 (ru)
KR (2) KR102488220B1 (ru)
CN (1) CN106163556B (ru)
AU (3) AU2014324884B2 (ru)
CA (2) CA2925106C (ru)
IL (2) IL244744B (ru)
MX (2) MX2016003957A (ru)
NZ (2) NZ718283A (ru)
RU (2) RU2715232C2 (ru)
WO (1) WO2015048329A2 (ru)

Cited By (27)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9650445B2 (en) 2012-02-28 2017-05-16 The University Of Birmingham Immunotherapeutic molecules and uses
WO2017106630A1 (en) 2015-12-18 2017-06-22 The General Hospital Corporation Polyacetal polymers, conjugates, particles and uses thereof
WO2017218533A1 (en) 2016-06-13 2017-12-21 Torque Therapeutics, Inc. Methods and compositions for promoting immune cell function
WO2018106738A1 (en) 2016-12-05 2018-06-14 Massachusetts Institute Of Technology Brush-arm star polymers, conjugates and particles, and uses thereof
US10035856B2 (en) 2015-11-19 2018-07-31 Revitope Limited Functional antibody fragment complementation for a two-components system for redirected killing of unwanted cells
WO2018191438A1 (en) 2017-04-11 2018-10-18 Inhibrx, Inc. Multispecific polypeptide constructs having constrained cd3 binding and methods of using the same
WO2018213335A1 (en) * 2017-05-16 2018-11-22 Scalmibio, Inc. Activatable antibodies and methods of use thereof
US10138272B2 (en) 2014-01-31 2018-11-27 Cytomx Therapeutics, Inc. Matriptase and u-plasminogen activator substrates and other cleavable moieties and methods of use thereof
WO2019036433A2 (en) 2017-08-16 2019-02-21 Bristol-Myers Squibb Company ANTIBODIES IN THE FORM OF PRODRUGS, PRODRUGS BASED THEREON, THEIR METHODS OF USE AND PREPARATION
WO2020023553A1 (en) 2018-07-24 2020-01-30 Inhibrx, Inc. Multispecific polypeptide constructs containing a constrained cd3 binding domain and a receptor binding region and methods of using the same
WO2020037236A1 (en) 2018-08-17 2020-02-20 Massachusetts Institute Of Technology Degradable polymers of a cyclic silyl ether and uses thereof
WO2020076970A1 (en) 2018-10-11 2020-04-16 Inhibrx, Inc. B7h3 single domain antibodies and therapeutic compositions thereof
WO2020076977A2 (en) 2018-10-11 2020-04-16 Inhibrx, Inc. Dll3 single domain antibodies and therapeutic compositions thereof
WO2020077257A1 (en) 2018-10-11 2020-04-16 Inhibrx, Inc. Pd-1 single domain antibodies and therapeutic compositions thereof
WO2020076992A1 (en) 2018-10-11 2020-04-16 Inhibrx, Inc. 5t4 single domain antibodies and therapeutic compositions thereof
US10696724B2 (en) 2018-05-14 2020-06-30 Werewolf Therapeutics, Inc. Activatable interleukin-2 polypeptides
US10696723B2 (en) 2018-05-14 2020-06-30 Werewolf Therapeutics, Inc. Activatable interleukin 12 polypeptides
WO2019222275A3 (en) * 2018-05-14 2020-08-20 TCR2 Therapeutics Inc. Compositions and methods for tcr reprogramming using inducible fusion proteins
WO2020236253A1 (en) 2019-05-20 2020-11-26 Massachusetts Institute Of Technology Boronic ester prodrugs and uses thereof
WO2020251878A1 (en) 2019-06-11 2020-12-17 Bristol-Myers Squibb Company Anti-ctla4 antibody prodruggable (probody) at a cdr position
US11028142B2 (en) 2015-05-18 2021-06-08 TCR2 Therapeutics Inc. Compositions and methods for TCR reprogramming using fusion proteins
US11046759B2 (en) 2015-01-20 2021-06-29 Cytomx Therapeutics, Inc. Matrix metalloprotease-cleavable and serine protease-cleavable substrates and methods of use thereof
WO2021141662A1 (en) 2020-01-10 2021-07-15 Massachusetts Institute Of Technology Proteolysis targeting chimeric molecules (protacs) with functional handles and uses thereof
WO2021155071A1 (en) 2020-01-29 2021-08-05 Inhibrx, Inc. Cd28 single domain antibodies and multivalent and multispecific constructs thereof
WO2022029420A1 (en) 2020-08-03 2022-02-10 Bicycletx Limited Peptide-based linkers
US11739132B2 (en) 2019-05-14 2023-08-29 Werewolf Therapeutics, Inc. Separation moieties and methods of use thereof
US11814410B2 (en) 2013-09-25 2023-11-14 Cytomx Therapeutics, Inc. Matrix metalloproteinase substrates and other cleavable moieties and methods of use thereof

Families Citing this family (41)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
SG11201509595PA (en) 2013-05-28 2015-12-30 Dcb Usa Llc Antibody locker for the inactivation of protein drug
MY185014A (en) 2015-08-07 2021-04-30 Alx Oncology Inc Contructs having a sirp-alpha domain or variant thereof
CN106519037B (zh) * 2015-09-11 2019-07-23 科济生物医药(上海)有限公司 可活化的嵌合受体
TW202323287A (zh) 2016-11-28 2023-06-16 日商中外製藥股份有限公司 包含抗原結合域與運送部分的多胜肽
WO2018097308A1 (ja) 2016-11-28 2018-05-31 中外製薬株式会社 リガンド結合活性が調整可能なリガンド結合分子
CA3042276A1 (en) 2016-12-09 2018-06-14 Seattle Genetics, Inc. Bivalent antibodies masked by coiled coils
CN107501394B (zh) * 2017-06-19 2021-02-26 天津医科大学 高特异性mmp-14底物肽及其制备方法和应用
CN111511762A (zh) 2017-08-21 2020-08-07 天演药业公司 抗cd137分子及其用途
US20210206845A1 (en) 2017-11-28 2021-07-08 Chugai Seiyaku Kabushiki Kaisha Ligand-binding molecule having adjustable ligand-binding activity
TW202348632A (zh) 2017-11-28 2023-12-16 日商中外製藥股份有限公司 含有抗原結合域及運輸部分之多胜肽
SG11202004864TA (en) 2017-12-01 2020-06-29 Seattle Genetics Inc Cd47 antibodies and uses thereof for treating cancer
WO2019148444A1 (en) 2018-02-02 2019-08-08 Adagene Inc. Anti-ctla4 antibodies and methods of making and using the same
WO2019148445A1 (en) * 2018-02-02 2019-08-08 Adagene Inc. Precision/context-dependent activatable antibodies, and methods of making and using the same
EP3769309A1 (en) 2018-03-20 2021-01-27 Cytomx Therapeutics Inc. Systems and methods for quantitative pharmacological modeling of activatable antibody species in mammalian subjects
US20210253672A1 (en) 2018-05-30 2021-08-19 Chugai Seiyaku Kabushiki Kaisha Ligand-binding molecule containing single domain antibody
WO2020118109A2 (en) * 2018-12-06 2020-06-11 Cytomx Therapeutics, Inc. Matrix metalloprotease-cleavable and serine or cysteine protease-cleavable substrates and methods of use thereof
CA3133802A1 (en) 2019-03-21 2020-09-24 Immunogen, Inc. Methods of preparing cell-binding agent-drug conjugates
SG11202110922QA (en) 2019-04-26 2021-10-28 Immunogen Inc Camptothecin derivatives
CA3141130A1 (en) 2019-05-31 2020-12-03 ALX Oncology Inc. Methods of treating cancer with sirp alpha fc fusion in combination with an immune checkpoint inhibitor
WO2020247572A1 (en) 2019-06-05 2020-12-10 Seattle Genetics, Inc. Masked antibody formulations
CN113905757A (zh) 2019-06-05 2022-01-07 中外制药株式会社 抗体切割位点结合分子
AR119097A1 (es) 2019-06-05 2021-11-24 Seattle Genetics Inc Métodos de purificación de anticuerpos enmascarados
CN114945597A (zh) * 2020-01-17 2022-08-26 艾提欧生物疗法有限公司 降低脱靶毒性的前抗体
EP4132656A1 (en) 2020-04-09 2023-02-15 CytomX Therapeutics, Inc. Compositions containing activatable antibodies
CN115667523A (zh) 2020-04-10 2023-01-31 西托姆克斯治疗公司 可活化细胞因子构建体和相关组合物以及方法
JP2024511387A (ja) 2021-03-16 2024-03-13 シートムエックス セラピューティクス,インコーポレイテッド マスクした活性化可能サイトカイン構成体ならびに関連する組成物及び方法
EP4370211A1 (en) 2021-07-14 2024-05-22 Seagen Inc. Antibody masking domains
WO2023034825A1 (en) 2021-08-30 2023-03-09 Cytomx Therapeutics, Inc. Method for determining protease activity in a biological sample
WO2023049825A1 (en) 2021-09-24 2023-03-30 Seagen Inc. Improved antibody masking domains
CA3233707A1 (en) 2021-10-08 2023-04-13 Alexey Yevgenyevich Berezhnoy Activatable cytokine constructs and combination methods
WO2023060156A2 (en) 2021-10-08 2023-04-13 Cytomx Therapeutics, Inc. Activatable cytokine constructs and related compositions and methods
US20230174995A1 (en) 2021-10-15 2023-06-08 Cytomx Therapeutics, Inc. Activatable polypeptide complex
CA3234609A1 (en) 2021-10-15 2023-04-20 Leila M. BOUSTANY Activatable polypeptide complex
WO2023064955A1 (en) 2021-10-15 2023-04-20 Cytomx Therapeutics, Inc. Activatable anti-cd3, anti-egfr, heteromultimeric bispecific polypeptide complex
WO2023183888A1 (en) 2022-03-23 2023-09-28 Cytomx Therapeutics, Inc. Activatable antigen-binding protein constructs and uses of the same
WO2023183923A1 (en) 2022-03-25 2023-09-28 Cytomx Therapeutics, Inc. Activatable dual-anchored masked molecules and methods of use thereof
WO2024030847A1 (en) 2022-08-01 2024-02-08 Cytomx Therapeutics, Inc. Protease-cleavable moieties and methods of use thereof
WO2024030858A1 (en) 2022-08-01 2024-02-08 Cytomx Therapeutics, Inc. Protease-cleavable substrates and methods of use thereof
WO2024030845A1 (en) 2022-08-01 2024-02-08 Cytomx Therapeutics, Inc. Protease-cleavable moieties and methods of use thereof
WO2024030850A1 (en) 2022-08-01 2024-02-08 Cytomx Therapeutics, Inc. Protease-cleavable substrates and methods of use thereof
WO2024030843A1 (en) 2022-08-01 2024-02-08 Cytomx Therapeutics, Inc. Protease-cleavable moieties and methods of use thereof

Citations (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6558728B1 (en) * 1996-05-10 2003-05-06 Danisco A/S α-glucuronidases of aspergillus, production thereof and their uses
US20040123343A1 (en) * 2000-04-19 2004-06-24 La Rosa Thomas J. Rice nucleic acid molecules and other molecules associated with plants and uses thereof for plant improvement
US20100018965A1 (en) * 2008-07-25 2010-01-28 Schmidt-Seeger Gmbh Round container for germinating and drying malt
US20100041588A1 (en) * 2003-07-01 2010-02-18 Keay Susan K Derivatives of apf and methods of use
US20100189651A1 (en) * 2009-01-12 2010-07-29 Cytomx Therapeutics, Llc Modified antibody compositions, methods of making and using thereof
WO2010088691A2 (en) * 2009-02-02 2010-08-05 Washington State University Compositions and methods for treating or preventing conditions and diseases associated with mannheimia haemolytica
WO2011028698A2 (en) * 2009-09-02 2011-03-10 Kansas State University Research Foundation Mri and optical assays for proteases
US20110214205A1 (en) * 2010-02-26 2011-09-01 Monsanto Technology Llc. Isolated Novel Nucleic Acid and Protein Molecules from Foxtail Millet and Methods of Using Those Molecules to Generate Transgenic Plants with Enhanced Agronomic Traits
US20110287517A1 (en) * 2010-05-20 2011-11-24 Allergan, Inc. Degradable Clostridial Toxins
US20140363430A1 (en) * 2012-09-25 2014-12-11 Cytomx Therapeutics, Inc. Activatable Antibodies that Bind Interleukin-6 Receptor and Methods of Use Thereof

Family Cites Families (59)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3773919A (en) 1969-10-23 1973-11-20 Du Pont Polylactide-drug mixtures
US4485045A (en) 1981-07-06 1984-11-27 Research Corporation Synthetic phosphatidyl cholines useful in forming liposomes
US4522811A (en) 1982-07-08 1985-06-11 Syntex (U.S.A.) Inc. Serial injection of muramyldipeptides and liposomes enhances the anti-infective activity of muramyldipeptides
US4544545A (en) 1983-06-20 1985-10-01 Trustees University Of Massachusetts Liposomes containing modified cholesterol for organ targeting
EP0279862B1 (en) 1986-08-28 1993-11-03 Teijin Limited Cytocidal antibody complex and process for its preparation
US5013556A (en) 1989-10-20 1991-05-07 Liposome Technology, Inc. Liposomes with enhanced circulation time
US5151510A (en) 1990-04-20 1992-09-29 Applied Biosystems, Inc. Method of synethesizing sulfurized oligonucleotide analogs
JP3095175B2 (ja) 1992-11-13 2000-10-03 アイデック ファーマシューティカルズ コーポレイション B細胞リンパ腫の治療のためのヒトbリンパ球限定分化抗原に対するキメラ抗体と放射能標識抗体の療法利用
AUPO930697A0 (en) * 1997-09-19 1997-10-09 Walter And Eliza Hall Institute Of Medical Research, The Catalytic antibodies and a method of producing same
US6551795B1 (en) 1998-02-18 2003-04-22 Genome Therapeutics Corporation Nucleic acid and amino acid sequences relating to pseudomonas aeruginosa for diagnostics and therapeutics
JP4493846B2 (ja) * 1998-05-13 2010-06-30 ドマンティス リミテッド 選択系
WO2001055309A2 (en) 2000-01-31 2001-08-02 Human Genome Sciences, Inc. Nucleic acids, proteins, and antibodies
WO2001091798A2 (en) 2000-06-01 2001-12-06 Universite Catholique De Louvain Tumor activated prodrug compounds
AU2001280464A1 (en) 2000-08-04 2002-02-18 Eli Lilly And Company C1q-related factor, homologous polypeptides and therapeutic uses thereof
JP4334866B2 (ja) 2000-10-09 2009-09-30 アイシス イノヴェイション リミテッド 治療用抗体
US7465790B2 (en) 2000-10-09 2008-12-16 Isis Innovation, Inc. Therapeutic antibodies
WO2002038796A2 (en) 2000-11-08 2002-05-16 Beth Israel Deaconess Medical Center, Inc. Methods for determining protease cleavage site motifs
US20050208602A1 (en) * 2001-08-10 2005-09-22 Rosen Craig A 89 human secreted proteins
US7439319B2 (en) 2001-09-14 2008-10-21 Burnham Institute For Medical Research Selective substrates for matrix metalloproteinases
WO2003038083A1 (en) 2001-10-29 2003-05-08 Bayer Healthcare Ag Regulation of human type i adenylate cyclase
CN101044154A (zh) 2002-02-14 2007-09-26 威廉·J·鲁特 治疗宿主中切割的嵌合分子
US20040109855A1 (en) 2002-07-23 2004-06-10 Herman Waldmann Therapeutic antibodies with reduced side effect
US8809504B2 (en) 2002-09-03 2014-08-19 Vit Lauermann Inhibitor which is deactivatable by a reagent produced by a target cell
US7276497B2 (en) 2003-05-20 2007-10-02 Immunogen Inc. Cytotoxic agents comprising new maytansinoids
GB0417430D0 (en) 2004-08-05 2004-09-08 Uc3 A novel HPV vaccine comprising peptides from host cell proteins
JP2008535857A (ja) 2005-04-07 2008-09-04 ノバルティス ヴァクシンズ アンド ダイアグノスティクス インコーポレイテッド 癌の診断、検出および処置におけるcacna1e
CA2620886C (en) 2005-08-31 2017-03-14 The Regents Of The University Of California Cellular libraries of peptide sequences (clips) and methods of using the same
JP2009529522A (ja) 2006-03-10 2009-08-20 ディアト 酵素で切断可能なリンカーを介して抗体に複合された抗癌剤
SI2502938T1 (sl) * 2006-10-27 2015-05-29 Genentech, Inc. Protitelesa in imunokonjugati in njihove uporabe
CA2697032C (en) * 2007-08-22 2021-09-14 The Regents Of The University Of California Activatable binding polypeptides and methods of identification and use thereof
GB0819287D0 (en) 2008-10-22 2008-11-26 Univ Bradford Compounds
US8895702B2 (en) * 2008-12-08 2014-11-25 City Of Hope Development of masked therapeutic antibodies to limit off-target effects; application to anti-EGFR antibodies
EP2356131A4 (en) * 2008-12-08 2012-09-12 Tegopharm Corp MASKING LIGANDS FOR REVERSIBLE INHIBITION OF VERSATILE COMPOUNDS
CN102481341B (zh) 2009-02-23 2017-05-17 希托马克斯医疗有限公司 蛋白原及其使用方法
CA2761310C (en) 2009-05-07 2017-02-28 Charles S. Craik Antibodies and methods of use thereof
US8524220B1 (en) 2010-02-09 2013-09-03 David Gordon Bermudes Protease inhibitor: protease sensitivity expression system composition and methods improving the therapeutic activity and specificity of proteins delivered by bacteria
ES2920373T3 (es) 2011-05-16 2022-08-03 Tagworks Pharmaceuticals B V Activación de fármaco bioortogonal
CN103842383B (zh) 2011-05-16 2017-11-03 健能隆医药技术(上海)有限公司 多特异性fab融合蛋白及其使用方法
CA2871458C (en) 2012-04-27 2020-06-23 Cytomx Therapeutics, Inc. Activatable antibodies that bind epidermal growth factor receptor and methods of use thereof
WO2013192546A1 (en) 2012-06-22 2013-12-27 Cytomx Therapeutics, Inc. Activatable antibodies having non-binding steric moieties and mehtods of using the same
SG11201408554QA (en) 2012-06-22 2015-02-27 Cytomx Therapeutics Inc Anti-jagged 1/jagged 2 cross-reactive antibodies, activatable anti-jagged antibodies and methods of use thereof
US9309510B2 (en) 2012-08-10 2016-04-12 Cytomx Therapeutics, Inc. Protease-resistant systems for polypeptide display and methods of making and using thereof
WO2014107559A1 (en) 2013-01-03 2014-07-10 Branders.Com, Inc. Methods and apparatus for determining a score value for criteria associated with a gift
WO2014107599A2 (en) * 2013-01-04 2014-07-10 Cytomx Therapeutics, Inc. Compositions and methods for detecting protease activity in biological systems
EP2988786A4 (en) 2013-04-22 2016-12-21 Avelas Biosciences Inc COMPOSITIONS AND METHODS OF USE FOR SELECTIVE DRUG DELIVERY
SG11201509595PA (en) 2013-05-28 2015-12-30 Dcb Usa Llc Antibody locker for the inactivation of protein drug
CA2925106C (en) 2013-09-25 2023-11-14 Cytomx Therapeutics, Inc. Matrix metalloproteinase substrates and other cleavable moieties and methods of use thereof
WO2015116933A2 (en) 2014-01-31 2015-08-06 Cytomx Therapeutics, Inc. Matriptase and u-plasminogen activator substrates and other cleavable moieties and methods of use thereof
CA2955947A1 (en) 2014-07-25 2016-01-28 Cytomx Therapeutics, Inc. Anti-cd3 antibodies, activatable anti-cd3 antibodies, multispecific anti-cd3 antibodies, multispecific activatable anti-cd3 antibodies, and methods of using the same
MA41374A (fr) 2015-01-20 2017-11-28 Cytomx Therapeutics Inc Substrats clivables par métalloprotéase matricielle et clivables par sérine protéase et procédés d'utilisation de ceux-ci
MA42971A (fr) 2015-03-13 2018-08-15 Cytomx Therapeutics Inc Anticorps anti-pdl1, anticorps anti-pld1 activables, et leurs procédés d'utilisation
DK3292149T3 (da) 2015-05-04 2022-02-28 Cytomx Therapeutics Inc Aktiverbare anti-cd71-antistoffer og fremgangsmåder til anvendelse deraf
AU2016258988A1 (en) 2015-05-04 2017-12-07 Cytomx Therapeutics, Inc Anti-ITGa3 antibodies, activatable anti-ITGa3 antibodies, and methods of use thereof
MX2017014136A (es) 2015-05-04 2018-07-06 Cytomx Therapeutics Inc Anticuerpos anti-cd166, anticuerpos anti-cd166 activables, y metodos de uso de los mismos.
BR112018000768A2 (pt) 2015-07-13 2018-09-25 Cytomx Therapeutics Inc anticorpos anti-pd-1, anticorpos anti-pd-1 ativáveis e métodos de uso dos mesmos
BR112019008223A2 (pt) 2016-11-03 2019-07-16 Bristol-Myers Squibb Company anticorpos anti-ctla-4 ativáveis e usos dos mesmos
US20180303952A1 (en) 2017-03-09 2018-10-25 Cytomx Therapeutics, Inc. Cd147 antibodies, activatable cd147 antibodies, and methods of making and use thereof
BR112019025188A2 (pt) 2017-06-01 2020-06-23 Cytomx Therapeutics, Inc. Anticorpos anti-pdl1 ativáveis e métodos de uso dos mesmos
WO2020118109A2 (en) 2018-12-06 2020-06-11 Cytomx Therapeutics, Inc. Matrix metalloprotease-cleavable and serine or cysteine protease-cleavable substrates and methods of use thereof

Patent Citations (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6558728B1 (en) * 1996-05-10 2003-05-06 Danisco A/S α-glucuronidases of aspergillus, production thereof and their uses
US20040123343A1 (en) * 2000-04-19 2004-06-24 La Rosa Thomas J. Rice nucleic acid molecules and other molecules associated with plants and uses thereof for plant improvement
US20100041588A1 (en) * 2003-07-01 2010-02-18 Keay Susan K Derivatives of apf and methods of use
US20100018965A1 (en) * 2008-07-25 2010-01-28 Schmidt-Seeger Gmbh Round container for germinating and drying malt
US20100189651A1 (en) * 2009-01-12 2010-07-29 Cytomx Therapeutics, Llc Modified antibody compositions, methods of making and using thereof
WO2010088691A2 (en) * 2009-02-02 2010-08-05 Washington State University Compositions and methods for treating or preventing conditions and diseases associated with mannheimia haemolytica
WO2011028698A2 (en) * 2009-09-02 2011-03-10 Kansas State University Research Foundation Mri and optical assays for proteases
US20110214205A1 (en) * 2010-02-26 2011-09-01 Monsanto Technology Llc. Isolated Novel Nucleic Acid and Protein Molecules from Foxtail Millet and Methods of Using Those Molecules to Generate Transgenic Plants with Enhanced Agronomic Traits
US20110287517A1 (en) * 2010-05-20 2011-11-24 Allergan, Inc. Degradable Clostridial Toxins
US20140363430A1 (en) * 2012-09-25 2014-12-11 Cytomx Therapeutics, Inc. Activatable Antibodies that Bind Interleukin-6 Receptor and Methods of Use Thereof

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
retrieved on 7/25/18 *
UniProtKB UniProtKB B1FZS3 (B1FZS3_9Burk)[online]. [retrieved on 7/25/18]. Retrieved from internet < https //www.uniprot.org/uniprot/B1FZS3 > *

Cited By (44)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9650445B2 (en) 2012-02-28 2017-05-16 The University Of Birmingham Immunotherapeutic molecules and uses
US9822180B2 (en) 2012-02-28 2017-11-21 The University Of Birmingham Immunotherapeutic molecules and uses
US10106621B2 (en) 2012-02-28 2018-10-23 The University Of Birmingham Immunotherapeutic molecules and uses
US11814410B2 (en) 2013-09-25 2023-11-14 Cytomx Therapeutics, Inc. Matrix metalloproteinase substrates and other cleavable moieties and methods of use thereof
US11884746B2 (en) 2014-01-31 2024-01-30 Cytomx Therapeutics, Inc. Matriptase and u-plasminogen activator substrates and other cleavable moieties and methods of use thereof
US10138272B2 (en) 2014-01-31 2018-11-27 Cytomx Therapeutics, Inc. Matriptase and u-plasminogen activator substrates and other cleavable moieties and methods of use thereof
US11028126B2 (en) 2014-01-31 2021-06-08 Cytomx Therapeutics, Inc. Matriptase and u-plasminogen activator substrates and other cleavable moieties and methods of use thereof
US11548944B2 (en) 2015-01-20 2023-01-10 Cytomx Therapeutics, Inc. Matrix metalloprotease-cleavable and serine protease-cleavable substrates and methods of use thereof
US11548943B2 (en) 2015-01-20 2023-01-10 Cytomx Therapeutics, Inc. Matrix metalloprotease-cleavable and serine protease-cleavable substrates and methods of use thereof
US11472875B1 (en) 2015-01-20 2022-10-18 Cytomx Therapeutics, Inc. Matrix metalloprotease-cleavable and serine protease-cleavable substrates and methods of use thereof
US11046759B2 (en) 2015-01-20 2021-06-29 Cytomx Therapeutics, Inc. Matrix metalloprotease-cleavable and serine protease-cleavable substrates and methods of use thereof
US11028142B2 (en) 2015-05-18 2021-06-08 TCR2 Therapeutics Inc. Compositions and methods for TCR reprogramming using fusion proteins
US11965012B2 (en) 2015-05-18 2024-04-23 TCR2 Therapeutics Inc. Compositions and methods for TCR reprogramming using fusion proteins
US10035856B2 (en) 2015-11-19 2018-07-31 Revitope Limited Functional antibody fragment complementation for a two-components system for redirected killing of unwanted cells
WO2017106630A1 (en) 2015-12-18 2017-06-22 The General Hospital Corporation Polyacetal polymers, conjugates, particles and uses thereof
WO2017218533A1 (en) 2016-06-13 2017-12-21 Torque Therapeutics, Inc. Methods and compositions for promoting immune cell function
WO2018106738A1 (en) 2016-12-05 2018-06-14 Massachusetts Institute Of Technology Brush-arm star polymers, conjugates and particles, and uses thereof
US11866507B2 (en) 2017-04-11 2024-01-09 Inhibrx, Inc. Multispecific polypeptide constructs having constrained CD3 binding and methods of using the same
WO2018191438A1 (en) 2017-04-11 2018-10-18 Inhibrx, Inc. Multispecific polypeptide constructs having constrained cd3 binding and methods of using the same
US11939385B2 (en) 2017-05-16 2024-03-26 ALX Oncology Inc. Activatable antibodies and methods of use thereof
WO2018213335A1 (en) * 2017-05-16 2018-11-22 Scalmibio, Inc. Activatable antibodies and methods of use thereof
WO2019036433A2 (en) 2017-08-16 2019-02-21 Bristol-Myers Squibb Company ANTIBODIES IN THE FORM OF PRODRUGS, PRODRUGS BASED THEREON, THEIR METHODS OF USE AND PREPARATION
US11623965B2 (en) 2017-08-16 2023-04-11 Bristol-Myers Squibb Company Prodruggable antibodies, prodrugs thereof, and methods of use and making
US10696723B2 (en) 2018-05-14 2020-06-30 Werewolf Therapeutics, Inc. Activatable interleukin 12 polypeptides
US10696724B2 (en) 2018-05-14 2020-06-30 Werewolf Therapeutics, Inc. Activatable interleukin-2 polypeptides
US11981716B2 (en) 2018-05-14 2024-05-14 Werewolf Therapeutics, Inc. Activatable interleukin-2 polypeptides and methods of use thereof
US11535658B2 (en) 2018-05-14 2022-12-27 Werewolf Therapeutics, Inc. Activatable interleukin-2 polypeptides and methods of use thereof
WO2019222275A3 (en) * 2018-05-14 2020-08-20 TCR2 Therapeutics Inc. Compositions and methods for tcr reprogramming using inducible fusion proteins
US11453710B2 (en) 2018-05-14 2022-09-27 Werewolf Therapeutics, Inc. Activatable interleukin 12 polypeptides and methods of use thereof
US11352403B2 (en) 2018-05-14 2022-06-07 Werewolf Therapeutics, Inc. Activatable interleukin-2 polypeptides and methods of use thereof
WO2020023553A1 (en) 2018-07-24 2020-01-30 Inhibrx, Inc. Multispecific polypeptide constructs containing a constrained cd3 binding domain and a receptor binding region and methods of using the same
WO2020037236A1 (en) 2018-08-17 2020-02-20 Massachusetts Institute Of Technology Degradable polymers of a cyclic silyl ether and uses thereof
US11208485B2 (en) 2018-10-11 2021-12-28 Inhibrx, Inc. PD-1 single domain antibodies and therapeutic compositions thereof
US11945869B2 (en) 2018-10-11 2024-04-02 Inhibrx, Inc. PD-1 single domain antibodies and therapeutic compositions thereof
WO2020076992A1 (en) 2018-10-11 2020-04-16 Inhibrx, Inc. 5t4 single domain antibodies and therapeutic compositions thereof
WO2020077257A1 (en) 2018-10-11 2020-04-16 Inhibrx, Inc. Pd-1 single domain antibodies and therapeutic compositions thereof
WO2020076977A2 (en) 2018-10-11 2020-04-16 Inhibrx, Inc. Dll3 single domain antibodies and therapeutic compositions thereof
WO2020076970A1 (en) 2018-10-11 2020-04-16 Inhibrx, Inc. B7h3 single domain antibodies and therapeutic compositions thereof
US11739132B2 (en) 2019-05-14 2023-08-29 Werewolf Therapeutics, Inc. Separation moieties and methods of use thereof
WO2020236253A1 (en) 2019-05-20 2020-11-26 Massachusetts Institute Of Technology Boronic ester prodrugs and uses thereof
WO2020251878A1 (en) 2019-06-11 2020-12-17 Bristol-Myers Squibb Company Anti-ctla4 antibody prodruggable (probody) at a cdr position
WO2021141662A1 (en) 2020-01-10 2021-07-15 Massachusetts Institute Of Technology Proteolysis targeting chimeric molecules (protacs) with functional handles and uses thereof
WO2021155071A1 (en) 2020-01-29 2021-08-05 Inhibrx, Inc. Cd28 single domain antibodies and multivalent and multispecific constructs thereof
WO2022029420A1 (en) 2020-08-03 2022-02-10 Bicycletx Limited Peptide-based linkers

Also Published As

Publication number Publication date
EP3049111A2 (en) 2016-08-03
WO2015048329A2 (en) 2015-04-02
JP2023159099A (ja) 2023-10-31
AU2014324884A1 (en) 2016-04-21
JP2016538240A (ja) 2016-12-08
IL244744B (en) 2022-04-01
JP2020015749A (ja) 2020-01-30
AU2023254931A1 (en) 2023-11-16
MX2016003957A (es) 2017-02-02
CN106163556B (zh) 2024-04-09
CN106163556A (zh) 2016-11-23
IL291329A (en) 2022-05-01
AU2014324884B2 (en) 2020-03-26
AU2020203910B2 (en) 2023-10-05
BR112016006665A8 (pt) 2018-01-30
KR102488220B1 (ko) 2023-01-12
RU2715232C2 (ru) 2020-02-26
IL244744A0 (en) 2016-04-21
CA2925106C (en) 2023-11-14
KR20230011497A (ko) 2023-01-20
WO2015048329A3 (en) 2015-06-04
NZ756892A (en) 2022-05-27
JP2021138707A (ja) 2021-09-16
US20240084022A1 (en) 2024-03-14
BR112016006665A2 (pt) 2017-09-12
JP6915987B2 (ja) 2021-08-11
RU2020106752A (ru) 2020-03-11
NZ718283A (en) 2022-05-27
AU2020203910A1 (en) 2020-07-02
CA2925106A1 (en) 2015-04-02
US20190284283A1 (en) 2019-09-19
US20240150478A1 (en) 2024-05-09
CA3214529A1 (en) 2015-04-02
RU2016115542A3 (ru) 2018-11-01
MX2021000416A (es) 2021-03-25
US11814410B2 (en) 2023-11-14
RU2016115542A (ru) 2017-10-30
KR20160074510A (ko) 2016-06-28

Similar Documents

Publication Publication Date Title
US20240084022A1 (en) Matrix metalloproteinase substrates and other cleavable moieties and methods of use thereof
US11884746B2 (en) Matriptase and u-plasminogen activator substrates and other cleavable moieties and methods of use thereof
US11548943B2 (en) Matrix metalloprotease-cleavable and serine protease-cleavable substrates and methods of use thereof
US20200377602A1 (en) Matrix metalloprotease-cleavable and serine or cysteine protease-cleavable substrates and methods of use thereof
US10745481B2 (en) Anti-CD166 antibodies, activatable anti-CD166 antibodies, and methods of use thereof
US20210025877A1 (en) Methods of qualitatively and/or quantitatively analyzing properties of activatable antibodies and uses thereof

Legal Events

Date Code Title Description
AS Assignment

Owner name: CYTOMX THERAPEUTICS, INC., CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:MOORE, STEPHEN JAMES;NGUYEN, MARGARET THY LUU;HOSTETTER, DANIEL R.;AND OTHERS;SIGNING DATES FROM 20141218 TO 20150107;REEL/FRAME:034802/0940

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION