WO2016073879A2 - Transforming growth factor-related antibodies and uses thereof - Google Patents

Transforming growth factor-related antibodies and uses thereof Download PDF

Info

Publication number
WO2016073879A2
WO2016073879A2 PCT/US2015/059515 US2015059515W WO2016073879A2 WO 2016073879 A2 WO2016073879 A2 WO 2016073879A2 US 2015059515 W US2015059515 W US 2015059515W WO 2016073879 A2 WO2016073879 A2 WO 2016073879A2
Authority
WO
WIPO (PCT)
Prior art keywords
antibody
amino acid
acid sequence
set forth
seq
Prior art date
Application number
PCT/US2015/059515
Other languages
French (fr)
Other versions
WO2016073879A3 (en
Inventor
Gregory J. Carven
Justin W. JACKSON
Michelle STRAUB
Dong Yun Lee
William K. MCCONAUGHY
Jared J. SHEEHAN
Katherine Jane Turner
Nagesh K. Mahanthappa
Original Assignee
Scholar Rock, 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 Scholar Rock, Inc. filed Critical Scholar Rock, Inc.
Publication of WO2016073879A2 publication Critical patent/WO2016073879A2/en
Publication of WO2016073879A3 publication Critical patent/WO2016073879A3/en

Links

Classifications

    • 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
    • C07K16/22Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against growth factors ; against growth regulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/33Crossreactivity, e.g. for species or epitope, or lack of said crossreactivity
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/60Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments
    • C07K2317/62Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments comprising only variable region components
    • C07K2317/622Single chain antibody (scFv)
    • 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
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value

Definitions

  • Embodiments of the present disclosure may include modulators of growth factor activity.
  • such modulators may include antibodies and may modulate TGF- ⁇ family member activity and/or biology.
  • Cell signaling molecules stimulate a variety of cellular activities. Such signaling is often tightly regulated, often through interactions with other biomolecules, the extracellular and/or cellular matrix or within a particular cell environment or niche. Such interactions may be direct or indirect.
  • Cell signaling cascades are involved in a number of diverse biological pathways including, but not limited to modulation of cell growth, modulation of tissue homeostasis, extracellular matrix (ECM) dynamics, modulation of cell migration, invasion and immune modulation/suppression.
  • ECM extracellular matrix
  • proteins involved in cell signaling are synthesized and/or are sequestered in latent form, requiring stimulus of some kind to participate in signaling events.
  • antibodies have selected binding profiles to TGFP family member proteins and/or forms thereof.
  • antibodies with selected binding profiles specifically bind to different forms of TGFP family member proteins.
  • antibodies provided herein specifically bind to one or more of a pro-form, a latent-form, a primed-form, and/or a mature- form of a TGFP family member protein, such as proMyostatin, latent Myostatin, primed Myostatin and/or mature Myostatin. In some embodiments, such antibodies inhibit
  • Myostatin signaling In some embodiments, inhibition of Myostatin signaling is useful for increasing muscle mass or preventing muscle atrophy.
  • antibodies provided herein bind to and prevent cleavage of Myostatin by a proprotein convertase and/or a tolloid protease. Preventing cleavage of proMyostatin or latent Myostatin, in some embodiments, prevents Myostatin activation. Further aspects of the disclosure relate to antibodies having selected binding profiles and an affinity to an antigen that is sensitive to pH. Furthermore, in some embodiments, antibodies provided herein are sweeping antibodies that can efficiently clear antigens (e.g., proMyostatin and/or latent Myostatin) from serum.
  • antigens e.g., proMyostatin and/or latent Myostatin
  • the present disclosure includes an antibody that specifically binds human proMyostatin, human latent Myostatin, human GDF-8 prodomain, human GDF- 11 ARM8 prodomain, and human proGDFl 1 ARM8, but does not specifically bind human mature GDF-8 having an amino acid sequence as set forth in SEQ ID NO: 42.
  • Another aspect of the invention includes an antibody that specifically binds human proMyostatin, murine proMyostatin, human latent Myostatin, human GDF-8 prodomain, human GDF-11 ARM8 prodomain, and human proGDFl 1 ARM8, but does not specifically bind human mature GDF-8 having an amino acid sequence as set forth in SEQ ID NO: 42.
  • human proMyostatin has an amino acid sequence as set forth in SEQ ID NO: 5.
  • murine proMyostatin has an amino acid sequence as set forth in SEQ ID NO: 88.
  • human latent Myostatin has an amino acid sequence as set forth in SEQ ID NO: 5.
  • human GDF-8 prodomain has an amino acid sequence as set forth in SEQ ID NO: 38.
  • human GDF- 11 ARM8 prodomain has an amino acid sequence as set forth in SEQ ID NO: 181.
  • human proGDFl 1 ARM8 has an amino acid sequence as set forth in SEQ ID NO: 179.
  • an antibody specifically binds human latent proGDFl 1 ARM8, but does not specifically bind human proGDFl 1 having an amino acid sequence as set forth in SEQ ID NO: 4.
  • an antibody specifically binds murine latent myostatin and human latent proGDFl 1 ARM8, but does not specifically bind human proGDFl 1 having an amino acid sequence as set forth in SEQ ID NO: 4.
  • a murine latent myostatin has an amino acid sequence as set forth in SEQ ID NO: 88.
  • a human latent proGDFl 1 ARM8 has an amino acid sequence as set forth in SEQ ID NO: 179.
  • an antibody comprises a CDR-H3 amino acid sequence set forth in SEQ ID NO: 752, 780, 815, 772, 784, 770, 774, 756, 796, 785, 761, 788, 767, 765, 799, 749, 802, 809, 750, 803, 800, 806, 759, 805, 797, 787, or 766, 782, 967, 776, 755, 876, 891, 963, 996, 993, 970, 968 or 1008.
  • an antibody comprises a CDR-L3 amino acid sequence set forth in any one of SEQ ID NO: 1367, 1394, 1660, 1387, 1397, 1385, 1388, 1371, 1409, 1398, 1376, 1401, 1382, 1380, 1656, 1364, 1413, 1418, 1365, 1658, 1657, 1415, 1374, 1659, 1410, 1400, 1381, 1395, 1582, 1390, 1370, 1670, 1485, 1578, 1611, 1608, 1585, 1583, or 1623.
  • an antibody comprises a CDR-H1, a CDR-H2, and/or a CDR-H3 from the variable heavy chain (VH) amino acid sequence of clone BP6a-l, BP6a-2, BP6a-3, BP6a-4, BP6a-5, BP6a-6, BP6a-7, BP6a-8, BP6a-9, BP6a-10, BP6a-l l, BP6a-12, BP6a-13, BP6a-14, BP6a-15, BP6a-16, BP6a-17, BP6a-18, BP6a-19, BP6a-20, BP6a-21, BP6a-22, BP6a-23, BP6a-24, BP6a-25, BP6a-26, BP6a-27, BP6b-l, BP6b-2, BP6b-3, BP6b- 4, BP6b-5, BP
  • an antibody comprises a variable heavy chain amino acid sequence of clone BP6a-l, BP6a-2, BP6a-3, BP6a-4, BP6a-5, BP6a-6, BP6a-7, BP6a-8, BP6a-9, BP6a-10, BP6a-l l, BP6a-12, BP6a-13, BP6a-14, BP6a-15, BP6a-16, BP6a-17, BP6a-18, BP6a-19, BP6a-20, BP6a-21, BP6a-22, BP6a-23, BP6a-24, BP6a-25, BP6a-26, BP6a-27, BP6b-l, BP6b-2, BP6b-3, BP6b-4, BP6b-5, BP6b-6, BP6b-7, BP6b-8, BP6b-9, BP6b-10,
  • an antibody comprises the variable light chain amino acid sequence of clone BP6a-l, BP6a-2, BP6a-3, BP6a-4, BP6a-5, BP6a-6, BP6a-7, BP6a-8, BP6a-9, BP6a-10, BP6a-l l, BP6a-12, BP6a-13, BP6a-14, BP6a-15, BP6a-16, BP6a-17, BP6a-18, BP6a-19, BP6a-20, BP6a-21, BP6a-22, BP6a-23, BP6a-24, BP6a-25, BP6a-26, BP6a-27, BP6b-l, BP6b-2, BP6b-3, BP6b-4, BP6b-5, BP6b-6, BP6b-7, BP6b-8, BP6b-9, BP6b-10, BP6b-
  • Further aspects of the present disclosure include an antibody that specifically binds human proMyostatin, and human proGDFl 1 ARM8 but does not specifically bind human latent Myostatin having an amino acid sequence as set forth in SEQ ID NO: 5, human GDF-8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 38, human GDF-11 ARM8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 181, or human mature GDF-8 having an amino acid sequence as set forth in SEQ ID NO: 42.
  • Another aspect of the present disclosure includes an antibody that specifically binds human proMyostatin, murine proMyostatin and human proGDFl 1 ARM8 but does not specifically bind human latent Myostatin having an amino acid sequence as set forth in SEQ ID NO: 5, human GDF-8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 38, human GDF-11 ARM8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 181, or human mature GDF-8 having an amino acid sequence as set forth in SEQ ID NO: 42.
  • the human proMyostatin has an amino acid sequence as set forth in SEQ ID NO: 5.
  • the murine proMyostatin has an amino acid sequence as set forth in SEQ ID NO: 88.
  • the human proGDFl 1 ARM8 has an amino acid sequence as set forth in SEQ ID NO: 179.
  • the antibody comprises a CDR-H3 amino acid sequence set forth in SEQ ID NO: 813. In other embodiments, the antibody comprises a CDR-L3 amino acid sequence set forth in any one of SEQ ID NO: 1422.
  • the antibody comprises a CDR-H1, a CDR-H2, and/or a CDR-H3 from the variable heavy chain (VH) amino acid sequence of clone BP2a-l as set forth in Table 14, and/or a CDR-L1, a CDR-L2, and/or a CDR-L3 from the variable light chain (VL) amino acid sequence of clone BP2a-l as set forth in Table 16.
  • the antibody comprises a variable heavy chain amino acid sequence of clone BP2a-l as set forth in Table 14.
  • the antibody comprises the variable light chain amino acid sequence of clone BP2a-l as set forth in Table 16.
  • the present disclosure includes an antibody that specifically binds human proMyostatin, and human latent Myostatin, but does not specifically bind human GDF-8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 38, human GDF- 11 ARM8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 181, human proGDFl 1 ARM8 having an amino acid sequence as set forth in SEQ ID NO: 179, or human mature GDF-8 having an amino acid sequence as set forth in SEQ ID NO: 42.
  • Another aspect of the disclosure includes an antibody that specifically binds human proMyostatin, murine proMyostatin, and human latent Myostatin, but does not specifically bind human GDF-8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 38, human GDF-11 ARM8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 181, human proGDFl 1 ARM8 having an amino acid sequence as set forth in SEQ ID NO: 179, or human mature GDF-8 having an amino acid sequence as set forth in SEQ ID NO: 42.
  • a human proMyostatin has an amino acid sequence as set forth in SEQ ID NO: 5.
  • a murine proMyostatin has an amino acid sequence as set forth in SEQ ID NO: 88.
  • a human latent Myostatin has an amino acid sequence as set forth in SEQ ID NO: 5.
  • a antibody comprises a CDR-H3 amino acid sequence set forth in SEQ ID NO: 794, 804, or 751.
  • the antibody comprises a CDR-L3 amino acid sequence set forth in any one of SEQ ID NO: 1407, 1414, or 1366.
  • the antibody comprises a CDR-H1, a CDR-H2, and/or a CDR-H3 from the variable heavy chain (VH) amino acid sequence of clone BP3a-l, BP3a-2 or BP3a-3 as set forth in Table 14, and/or a CDR-L1, a CDR-L2, and/or a CDR-L3 from the variable light chain (VL) amino acid sequence of clone BP3a-l, BP3a-2 or BP3a-3 as set forth in Table 16.
  • the antibody comprises a variable heavy chain amino acid sequence of clone BP3a-l, BP3a-2 or BP3a-3 as set forth in Table 14.
  • the antibody comprises the variable light chain amino acid sequence of clone BP3a-l, BP3a-2 or BP3a-3 as set forth in Table 16.
  • aspects of the present disclosure relate to an antibody that specifically binds human proMyostatin, human latent Myostatin and human GDF8 prodomain but does not specifically bind human GDF-11 ARM 8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 181, human proGDFl 1 ARM8 having an amino acid sequence as set forth in SEQ ID NO: 179 or human mature GDF-8 having an amino acid sequence as set forth in SEQ ID NO: 42.
  • Further aspects of the disclosure include an antibody that specifically binds human proMyostatin, murine proMyostatin, human latent Myostatin and human GDF8 prodomain but does not specifically bind human GDF- 11 ARM8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 181, human proGDFl 1 ARM8 having an amino acid sequence as set forth in SEQ ID NO: 179 or human mature GDF-8 having an amino acid sequence as set forth in SEQ ID NO: 42.
  • the human proMyo statin has an amino acid sequence as set forth in SEQ ID NO: 5.
  • the murine proMyo statin has an amino acid sequence as set forth in SEQ ID NO: 88.
  • the human latent Myostatin has an amino acid sequence as set forth in SEQ ID NO: 5.
  • the human GDF-8 prodomain has an amino acid sequence as set forth in SEQ ID NO: 38.
  • the antibody comprises a CDR-H3 amino acid sequence set forth in SEQ ID NO: 768.
  • the antibody comprises a CDR-L3 amino acid sequence set forth in any one of SEQ ID NO: 1383.
  • the antibody comprises a CDR-H1, a CDR-H2, and/or a CDR-H3 from the variable heavy chain (VH) amino acid sequence of clone BP4a-l as set forth in Table 14, and/or a CDR-L1, a CDR-L2, and/or a CDR-L3 from the variable light chain (VL) amino acid sequence of clone BP4a- 1 as set forth in Table 16.
  • the antibody comprises a variable heavy chain amino acid sequence of clone BP4a-l as set forth in Table 14.
  • the antibody comprises the variable light chain amino acid sequence of clone BP4a- 1 as set forth in Table 16.
  • aspects of the disclosure further relate to an antibody that specifically binds human proMyostatin, human latent Myostatin, and human proGDFl 1 ARM8, but does not specifically bind human GDF-8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 38, human GDF-11 ARM8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 181, or human mature GDF-8 having an amino acid sequence as set forth in SEQ ID NO: 42.
  • Additional aspects of the disclosure include an antibody that specifically binds human proMyostatin, murine proMyostatin, human latent Myostatin, and human proGDFl 1 ARM8, but does not specifically bind human GDF-8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 38, human GDF-11 ARM8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 181, or human mature GDF-8 having an amino acid sequence as set forth in SEQ ID NO: 42.
  • the human proMyostatin has an amino acid sequence as set forth in SEQ ID NO: 5.
  • the murine proMyostatin has an amino acid sequence as set forth in SEQ ID NO: 88.
  • the human latent Myostatin has an amino acid sequence as set forth in SEQ ID NO: 5.
  • the human proGDFl 1 ARM8 has an amino acid sequence as set forth in SEQ ID NO: 179.
  • the antibody specifically binds human latent proGDFl 1 ARM8, but does not specifically bind human proGDFl 1 having an amino acid sequence as set forth in SEQ ID NO: 4.
  • the antibody specifically binds murine latent myostatin and human latent proGDFl 1 ARM8, but does not specifically bind human proGDFl 1 having an amino acid sequence as set forth in SEQ ID NO: 4.
  • the murine latent myostatin has an amino acid sequence as set forth in SEQ ID NO: 88.
  • the human latent proGDFl 1 ARM8 has an amino acid sequence as set forth in SEQ ID NO: 179.
  • the antibody comprises a CDR-H3 amino acid sequence set forth in SEQ ID NO: 760, 798, 792, 789, 801, 790, 812, 758, 762, 775, 834, 974, 985, or 1002.
  • the antibody comprises a CDR-L3 amino acid sequence set forth in any one of SEQ ID NO: 1375, 1411, 1405, 1402, 1412, 1403, 1421, 1373, 1377, 1389, 1661, 1589, 1600, or 1617.
  • the antibody comprises a CDR-H1, a CDR-H2, and/or a CDR-H3 from the variable heavy chain (VH) amino acid sequence of clone BP5a-l, BP5a-2, BP5a-3, BP5a-4 or BP5a-5, BP5b-l, BP5b-2, BP5b-3, BP5b-4, BP5b-5, BP5b-6, BP5b-7, BP5b-8 or BP5b-9 as set forth in Table 14, and/or a CDR-L1, a CDR-L2, and/or a CDR-L3 from the variable light chain (VL) amino acid sequence of clone BP5a-l, BP5a-2, BP5a-3, BP5a-4 or BP5a-5, BP5b-l, BP5b-2, BP5b-3, BP5b-4, BP5b-5, BP5b-6,
  • the antibody comprises a variable heavy chain amino acid sequence of clone BP5a-l, BP5a-2, BP5a-3, BP5a-4 or BP5a-5, BP5b-l, BP5b-2, BP5b-3, BP5b-4, BP5b-5, BP5b-6, BP5b-7, BP5b-8 or BP5b-9 as set forth in Table 14.
  • the antibody comprises the variable light chain amino acid sequence of clone BP5a-l, BP5a-2, BP5a-3, BP5a-4 or BP5a-5, BP5b-l, BP5b-2, BP5b-3, BP5b-4, BP5b-5, BP5b-6, BP5b-7, BP5b-8 or BP5b-9 as set forth in Table 16.
  • aspects of the present disclosure include an antibody that specifically binds human proMyo statin but does not specifically bind human latent Myostatin having an amino acid sequence as set forth in SEQ ID NO: 5, human GDF-8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 38, human GDF-11 ARM8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 181, human proGDFl 1 ARM8 having an amino acid sequence as set forth in SEQ ID NO: 179, or human mature GDF-8 having an amino acid sequence as set forth in SEQ ID NO: 42.
  • Further aspects of the invention include an antibody that specifically binds human proMyostatin and murine proMyostatin but does not specifically bind human latent Myostatin having an amino acid sequence as set forth in SEQ ID NO: 5, human GDF-8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 38, human GDF-11 ARM8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 181, human proGDFl 1 ARM8 having an amino acid sequence as set forth in SEQ ID NO: 179, or human mature GDF-8 having an amino acid sequence as set forth in SEQ ID NO: 42.
  • the human proMyo statin has an amino acid sequence as set forth in SEQ ID NO: 5.
  • the murine proMyo statin has an amino acid sequence as set forth in SEQ ID NO: 88).
  • the antibody does not specifically bind human proGDFl 1 having an amino acid sequence as set forth in SEQ ID NO: 4, or human latent proGDFl 1 ARM8 having an amino acid sequence as set forth in SEQ ID NO: 179.
  • the antibody does not specifically bind human proGDFl 1 having an amino acid sequence as set forth in SEQ ID NO: 4, murine latent myostatin having an amino acid sequence as set forth in SEQ ID NO: 88 or human latent proGDFl 1 ARM8 having an amino acid sequence as set forth in SEQ ID NO: 179.
  • the antibody comprises a CDR-H3 amino acid sequence as set forth in SEQ ID NO: 769 or 827.
  • the antibody comprises a CDR-L3 amino acid sequence set forth in any one of SEQ ID NO: 1384 or 1435.
  • the antibody comprises a CDR-H1, a CDR-H2, and/or a CDR-H3 from the variable heavy chain (VH) amino acid sequence of clone BPla-1 or BPlb-1 as set forth in Table 14, and/or a CDR-L1, a CDR-L2, and/or a CDR-L3 from the variable light chain (VL) amino acid sequence of clone BPla-1 or BPlb-1 as set forth in Table 16.
  • the antibody comprises a variable heavy chain amino acid sequence of clone BPla-1 or BPlb-1 as set forth in Table 14.
  • the antibody comprises the variable light chain amino acid sequence of clone BPla-1 or BPlb-1 as set forth in Table 16.
  • FIG. 1 Further aspects of the invention relate to an antibody that specifically binds human latent Myostatin but does not specifically bind human proMyo statin having an amino acid sequence as set forth in SEQ ID NO: 5, human GDF-8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 38, human GDF-11 ARM8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 181, human proGDFl 1 ARM8 having an amino acid sequence as set forth in SEQ ID NO: 179, human mature GDF-8 having an amino acid sequence as set forth in SEQ ID NO: 42 or human proGDFl 1 having an amino acid sequence as set forth in SEQ ID NO: 4).
  • Additional aspects of the invention include an antibody that specifically binds human latent Myostatin but does not specifically bind murine proMyostatin having an amino acid sequence as set forth in SEQ ID NO: 88, human proMyostatin having an amino acid sequence as set forth in SEQ ID NO: 5, human GDF-8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 38, human GDF-11 ARM8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 181, human proGDFl 1 ARM8 having an amino acid sequence as set forth in SEQ ID NO: 179, human mature GDF-8 having an amino acid sequence as set forth in SEQ ID NO: 42 or human proGDFl 1 having an amino acid sequence as set forth in SEQ ID NO: 4).
  • the human latent Myostatin has an amino acid sequence as set forth in SEQ ID NO: 5.
  • the antibody specifically binds human latent proGDFl 1 ARM8.
  • the antibody specifically binds murine latent myostatin and human latent proGDFl 1 ARM8.
  • the murine latent myostatin has an amino acid sequence as set forth in SEQ ID NO: 88.
  • the human latent proGDFl 1 ARM8 has an amino acid sequence as set forth in SEQ ID NO: 179.
  • the antibody comprises a CDR-H3 amino acid sequence set forth in SEQ ID NO: 990.
  • the antibody comprises a CDR-L3 amino acid sequence set forth in any one of SEQ ID NO: 1605. In other embodiments, the antibody comprises a CDR-H1, a CDR-H2, and/or a CDR-H3 from the variable heavy chain (VH) amino acid sequence of clone BPlOb-1 as set forth in Table 14, and/or a CDR-L1, a CDR-L2, and/or a CDR-L3 from the variable light chain (VL) amino acid sequence of clone BPlOb-1 as set forth in Table 16. In further embodiments, the antibody comprises a variable heavy chain amino acid sequence of clone BPlOb-1 as set forth in Table 14. In other embodiments, the antibody comprises the variable light chain amino acid sequence of clone BPlOb-las set forth in Table 16.
  • Some aspects of the present disclosure include an antibody that specifically binds human proMyostatin, human latent Myostatin, human proGDFl 1 ARM8 and human mature GDF-8 but does not specifically bind human GDF-8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 38 or human GDF-11 ARM8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 181.
  • Another aspect of the present disclosure relates to an antibody that specifically binds human proMyostatin, murine proMyostatin, human latent Myostatin, human proGDFl 1 ARM8 and human mature GDF-8 but does not specifically bind human GDF-8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 38 or human GDF-11 ARM8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 181.
  • the human proMyostatin has an amino acid sequence as set forth in SEQ ID NO: 5.
  • the murine proMyostatin has an amino acid sequence as set forth in SEQ ID NO: 88.
  • the human latent Myostatin has an amino acid sequence as set forth in SEQ ID NO: 5.
  • the human proGDFl 1 ARM8 has an amino acid sequence as set forth in SEQ ID NO: 179.
  • the human mature GDF-8 has an amino acid sequence as set forth in SEQ ID NO: 42.
  • the antibody comprises a CDR-H3 amino acid sequence set forth in SEQ ID NO: 764.
  • the antibody comprises a CDR-L3 amino acid sequence set forth in any one of SEQ ID NO: 1379. In other embodiments, the antibody comprises a CDR-H1, a CDR-H2, and/or a CDR-H3 from the variable heavy chain (VH) amino acid sequence of clone BPl la-1 as set forth in Table 14, and/or a CDR-L1, a CDR-L2, and/or a CDR-L3 from the variable light chain (VL) amino acid sequence of clone BPl la-1 as set forth in Table 16. In another embodiment, the antibody comprises a variable heavy chain amino acid sequence of clone BPl la-1 as set forth in Table 14. In some embodiments, the antibody comprises the variable light chain amino acid sequence of clone BPl la-1 as set forth in Table 16.
  • Additional aspects of the present disclosure include an antibody that specifically binds human latent Myostatin and human mature GDF-8 but does not specifically bind human proMyostatin having an amino acid sequence as set forth in SEQ ID NO: 5, human GDF-8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 38, human GDF- 11 ARM8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 181 or human proGDFl 1 ARM8 having an amino acid sequence as set forth in SEQ ID NO: 179.
  • FIG. 1 Further aspects of the present disclosure relate to an antibody that specifically binds human latent Myostatin and human mature GDF-8 but does not specifically bind murine proMyostatin having an amino acid sequence as set forth in SEQ ID NO: 88, human proMyostatin having an amino acid sequence as set forth in SEQ ID NO: 5, human GDF-8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 38, human GDF-11 ARM8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 181 or human proGDFl 1 ARM8 having an amino acid sequence as set forth in SEQ ID NO: 179.
  • the human latent Myostatin has an amino acid sequence as set forth in SEQ ID NO: 5.
  • the human mature GDF-8 has an amino acid sequence as set forth in SEQ ID NO: 42.
  • the antibody does not specifically bind human proGDFl 1 having an amino acid sequence as set forth in SEQ ID NO: 4 or human latent proGDFl 1 ARM8 having an amino acid sequence as set forth in SEQ ID NO: 179.
  • the antibody specifically binds murine latent myostatin but does not specifically bind human proGDFl 1 having an amino acid sequence as set forth in SEQ ID NO: 4 or human latent proGDFl 1 ARM8 having an amino acid sequence as set forth in SEQ ID NO: 179.
  • the murine latent myostatin has an amino acid sequence as set forth in SEQ ID NO: 88.
  • the antibody comprises a CDR-H3 amino acid sequence set forth in SEQ ID NO: 989.
  • the antibody comprises a CDR-L3 amino acid sequence set forth in any one of SEQ ID NO: 1604. In other embodiments, the antibody comprises a CDR-H1, a CDR-H2, and/or a CDR-H3 from the variable heavy chain (VH) amino acid sequence of clone BP12b-l as set forth in Table 14, and/or a CDR-L1, a CDR-L2, and/or a CDR-L3 from the variable light chain (VL) amino acid sequence of clone BP 12b- las set forth in Table 16. In another embodiment, the antibody comprises a variable heavy chain amino acid sequence of clone BP12b-l as set forth in Table 14. In some embodiments, the antibody comprises the variable light chain amino acid sequence of clone BP12b-l as set forth in Table 16.
  • Further aspects of the present disclosure include an antibody that specifically binds human proMyo statin and human GDF-8 prodomain, but does not specifically bind human latent Myostatin having an amino acid sequence as set forth in SEQ ID NO: 5, human GDF-11 ARM8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 181, human proGDFl 1 ARM8 having an amino acid sequence as set forth in SEQ ID NO: 179 or human mature GDF-8 having an amino acid sequence as set forth in SEQ ID NO: 42.
  • Another aspect of the present disclosure relates to an antibody that specifically binds human proMyostatin and human GDF-8 prodomain, but does not specifically bind murine proMyostatin having an amino acid sequence as set forth in SEQ ID NO: 88, human latent Myostatin having an amino acid sequence as set forth in SEQ ID NO: 5, human GDF- 11 ARM8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 181, human proGDFl 1 ARM8 having an amino acid sequence as set forth in SEQ ID NO: 179 or human mature GDF-8 having an amino acid sequence as set forth in SEQ ID NO: 42.
  • the human proMyostatin has an amino acid sequence as set forth in SEQ ID NO: 5.
  • the human GDF-8 prodomain has an amino acid sequence as set forth in SEQ ID NO: 38.
  • the antibody comprises a CDR-H3 amino acid sequence set forth in SEQ ID NO: 786.
  • the antibody comprises a CDR-L3 amino acid sequence set forth in any one of SEQ ID NO: 1399.
  • the antibody comprises a CDR-H1, a CDR-H2, and/or a CDR-H3 from the variable heavy chain (VH) amino acid sequence of clone BP15a-l as set forth in Table 14, and/or a CDR-L1, a CDR-L2, and/or a CDR-L3 from the variable light chain (VL) amino acid sequence of clone BP15a-l as set forth in Table 16.
  • the antibody comprises a variable heavy chain amino acid sequence of clone BP15a-l as set forth in Table 14.
  • the antibody comprises the variable light chain amino acid sequence of clone BP15a-l as set forth in Table 16.
  • Another aspect of the present disclosure includes an antibody that specifically binds human proMyo statin and human latent Myostatin but does not specifically bind human GDF-8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 38, human GDF-11 ARM8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 181, human proGDFl 1 ARM8 having an amino acid sequence as set forth in SEQ ID NO: 179 or human mature GDF-8 having an amino acid sequence as set forth in SEQ ID NO: 42.
  • An additional aspect of the present disclosure relates to an antibody that specifically binds human proMyo statin and human latent Myostatin but does not specifically bind murine proMyostatin having an amino acid sequence as set forth in SEQ ID NO: 88, human GDF-8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 38, human GDF- 11 ARM8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 181, human proGDFl 1 ARM8 having an amino acid sequence as set forth in SEQ ID NO: 179 or human mature GDF-8 having an amino acid sequence as set forth in SEQ ID NO: 42.
  • the human proMyostatin has an amino acid sequence as set forth in SEQ ID NO: 5.
  • the human latent Myostatin has an amino acid sequence as set forth in SEQ ID NO: 5.
  • the antibody does not specifically bind human proGDFl 1 having an amino acid sequence as set forth in SEQ ID NO: 4 or human latent proGDFl 1 ARM8 having an amino acid sequence as set forth in SEQ ID NO: 179.
  • the antibody does not specifically bind human proGDFl 1 having an amino acid sequence as set forth in SEQ ID NO: 4, murine latent myostatin having an amino acid sequence as set forth in SEQ ID NO: 88 or human latent proGDFl 1 ARM8 having an amino acid sequence as set forth in SEQ ID NO: 179.
  • the antibody comprises a CDR-H3 amino acid sequence set forth in SEQ ID NO: 810, 855, 971, 983, or 986.
  • the antibody comprises a CDR-L3 amino acid sequence set forth in any one of SEQ ID NO: 1419, 1662, 1586, 1598, or 1601.
  • the antibody comprises a CDR-H1, a CDR-H2, and/or a CDR-H3 from the variable heavy chain (VH) amino acid sequence of clone BP16a-l, BP16b-l, BP16b-2, BP16b-3 or BP16b-4 as set forth in Table 14, and/or a CDR-Ll, a CDR-L2, and/or a CDR-L3 from the variable light chain (VL) amino acid sequence of clone BP16a-l, BP 16b- 1, BP16b-2, BP16b-3 or BP16b-4 as set forth in Table 16.
  • VH variable heavy chain
  • the antibody comprises a variable heavy chain amino acid sequence of clone BP16a-l, BP16b-l, BP16b-2, BP16b-3 or BP16b-4 as set forth in Table 14. In some embodiments, the antibody comprises the variable light chain amino acid sequence of clone BP16a-l, BP16b-l, BP 16b-
  • Additional aspects of the present disclosure include an antibody that specifically binds human proMyostatin and human latent Myostatin but does not specifically bind human GDF-8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 38, human GDF-11 ARM8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 181, human proGDFl 1 ARM8 having an amino acid sequence as set forth in SEQ ID NO: 179, human mature GDF-8 having an amino acid sequence as set forth in SEQ ID NO: 42 or human proGDFl 1 having an amino acid sequence as set forth in SEQ ID NO: 4.
  • the human proMyostatin has amino acid sequence as set forth in SEQ ID NO: 5. In other embodiments, the human latent Myostatin has an amino acid sequence as set forth in SEQ ID NO: 5. In another embodiment, the antibody specifically binds human latent proGDFl 1 ARM8. In other embodiments, the human latent proGDFl 1 ARM8 has an amino acid sequence as set forth in SEQ ID NO: 179. In some embodiments, the antibody does not specifically bind human latent proGDFl 1 ARM8 having an amino acid sequence as set forth in SEQ ID NO: 179. In another embodiment, the antibody comprises a CDR-H3 amino acid sequence set forth in SEQ ID NO: 982.
  • the antibody comprises a CDR-L3 amino acid sequence set forth in any one of SEQ ID NO: 1597.
  • the antibody comprises a CDR-H1, a CDR-H2, and/or a CDR-H3 from the variable heavy chain (VH) amino acid sequence of clone BP17b-l as set forth in Table 14, and/or a CDR-L1, a CDR-L2, and/or a CDR-L3 from the variable light chain (VL) amino acid sequence of clone BP17b-l as set forth in Table 16.
  • the antibody comprises a variable heavy chain amino acid sequence of clone BP17b-l as set forth in Table 14.
  • the antibody comprises the variable light chain amino acid sequence of clone BP17b-l as set forth in Table 16.
  • FIG. 1 Another aspect of the present disclosure relate to an antibody that specifically binds human proMyostatin, human latent Myostatin and human proGDFl 1 ARM8 but does not specifically bind human GDF-8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 38, human GDF-11 ARM8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 181 or human mature GDF-8 having an amino acid sequence as set forth in SEQ ID NO: 42.
  • Another aspect of the present disclosure includes an antibody that specifically binds human proMyostatin, human latent Myostatin and human proGDFl 1 ARM8 but does not specifically bind murine proMyostatin having an amino acid sequence as set forth in SEQ ID NO: 88, human GDF-8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 38, human GDF-11 ARM8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 181 or human mature GDF-8 having an amino acid sequence as set forth in SEQ ID NO: 42.
  • the human proMyostatin has an amino acid sequence as set forth in SEQ ID NO: 5.
  • the human latent Myostatin has an amino acid sequence as set forth in SEQ ID NO: 5.
  • the human proGDFl 1 ARM8 has an amino acid sequence as set forth in SEQ ID NO: 179.
  • the antibody specifically binds human latent proGDFl 1 ARM8 but does not specifically bind human proGDFl 1 having an amino acid sequence as set forth in SEQ ID NO: 4.
  • the antibody specifically binds human latent proGDFl 1 ARM8 but does not specifically bind human proGDFl 1 having an amino acid sequence as set forth in SEQ ID NO: 4 or murine latent myostatin having an amino acid sequence as set forth in SEQ ID NO: 88.
  • the human latent proGDFl 1 ARM 8 has an amino acid sequence as set forth in SEQ ID NO: 179.
  • the antibody comprises a CDR-H3 amino acid sequence set forth in SEQ ID NO: 763, 814, 753, 777, 795, 969, 972, 976, or 981.
  • the antibody comprises a CDR-L3 amino acid sequence set forth in any one of SEQ ID NO: 1378, 1423, 1368, 1391, 1408, 1584, 1587, 1591, or 1596.
  • the antibody comprises a CDR-H1, a CDR-H2, and/or a CDR-H3 from the variable heavy chain (VH) amino acid sequence of clone BP18a-l or BP18a-2, BP18b-l, BP18b-2, BP18b-3, BP18b-4, BP18b-5, BP18b-6 or BP18b-7 as set forth in Table 14, and/or a CDR-L1, a CDR-L2, and/or a CDR-L3 from the variable light chain (VL) amino acid sequence of clone BP18a-l or BP18a-2, BP18b-l, BP18b-2, BP18b-3, BP18b-4, BP18b-5, BP18b-6 or BP18b-7as set forth in Table 16.
  • VH variable heavy chain
  • the antibody comprises a variable heavy chain amino acid sequence of clone BP18a-l or BP 18a- 2, BP18b-l, BP18b-2, BP18b-3, BP18b-4, BP18b-5, BP18b-6 or BP18b-7 as set forth in Table 14.
  • the antibody comprises the variable light chain amino acid sequence of clone BP18a-l or BP18a-2, BP18b-l, BP18b-2, BP18b-3, BP18b-4, BP18b-5, BP18b-6 or BP18b-7 as set forth in Table 16.
  • Further aspects of the present disclosure include an antibody that specifically binds human proMyostatin, human latent Myostatin, human GDF-8 prodomain, human GDF-11 ARM8 prodomain and human proGDFl 1 ARM8 but does not specifically bind human mature GDF-8 having an amino acid sequence as set forth in SEQ ID NO: 42.
  • Additional aspects of the present disclosure relate to an antibody that specifically binds human proMyostatin, human latent Myostatin, human GDF-8 prodomain, human GDF- 11 ARM8 prodomain and human proGDFl 1 ARM8 but does not specifically bind murine proMyostatin having an amino acid sequence as set forth in SEQ ID NO: 88 or human mature GDF-8 having an amino acid sequence as set forth in SEQ ID NO: 42.
  • human proMyostatin having an amino acid sequence as set forth in SEQ ID NO: 88 or human mature GDF-8 having an amino acid sequence as set forth in SEQ ID NO: 42.
  • the human proMyostatin has an amino acid sequence as set forth in SEQ ID NO: 5.
  • the human latent Myostatin has an amino acid sequence as set forth in SEQ ID NO: 5.
  • the human GDF-8 prodomain has an amino acid sequence as set forth in SEQ ID NO: 38.
  • the human GDF-11 ARM8 prodomain has an amino acid sequence as set forth in SEQ ID NO: 181.
  • the human proGDFl 1 ARM8 has an amino acid sequence as set forth in SEQ ID NO: 179.
  • the antibody specifically binds human latent proGDFl 1 ARM8 but does not specifically bind human proGDFl 1 having an amino acid sequence as set forth in SEQ ID NO: 4. In another embodiment, the antibody specifically binds human latent proGDFl 1 ARM8 but does not specifically bind human proGDFl 1 having an amino acid sequence as set forth in SEQ ID NO: 4 or murine latent myostatin having an amino acid sequence as set forth in SEQ ID NO: 88. In some embodiments, the human latent proGDFl 1 ARM8 has an amino acid sequence as set forth in SEQ ID NO: 179.
  • the antibody comprises a CDR-H3 amino acid sequence set forth in SEQ ID NO: 773, 807, 811, 754, 771, or 978. In other embodiments, the antibody comprises a CDR-L3 amino acid sequence set forth in any one of SEQ ID NO: 1655, 1416, 1420, 1369, 1386, or 1593.
  • the antibody comprises a CDR-H1, a CDR-H2, and/or a CDR-H3 from the variable heavy chain (VH) amino acid sequence of clone BP19a-l, BP19a-2 or BP19a-3, BP19b-l, BP19b-2 or BP19b-3 as set forth in Table 14, and/or a CDR-L1, a CDR-L2, and/or a CDR-L3 from the variable light chain (VL) amino acid sequence of clone BP19a-l, BP 19a- 2 or BP19a-3, BP19b-l, BP19b-2 or BP19b-3 as set forth in Table 16.
  • VH variable heavy chain
  • the antibody comprises a variable heavy chain amino acid sequence of clone BP19a-l, BP19a-2 or BP19a-3, BP19b-l, BP19b-2 or BP19b-3 as set forth in Table 14. In other embodiments, the antibody comprises the variable light chain amino acid sequence of clone BP19a-l, BP19a-2 or BP19a-3, BP19b-l, BP19b-2 or BP19b-3 as set forth in Table 16.
  • FIG. 1 Another aspect of present disclosure relate to an antibody that specifically binds human proMyostatin, human latent Myostatin, human proGDFl 1 ARM8, human proGDFl 1 or human latent proGDFl 1 ARM8 but does not specifically bind human GDF-8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 38, human GDF-11 ARM8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 181 or human mature GDF-8 having an amino acid sequence as set forth in SEQ ID NO: 42.
  • Additional aspects of present disclosure include an antibody that specifically binds human proMyostatin, murine proMyostatin, human latent Myostatin, human proGDFl 1 ARM8, human proGDFl 1, murine latent myostatin or human latent proGDFl 1 ARM8 but does not specifically bind human GDF-8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 38, human GDF-11 ARM8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 181 or human mature GDF-8 having an amino acid sequence as set forth in SEQ ID NO: 42.
  • the human proMyostatin has an amino acid sequence as set forth in SEQ ID NO: 5.
  • the murine proMyostatin has an amino acid sequence as set forth in SEQ ID NO: 88.
  • the human latent Myostatin has an amino acid sequence as set forth in SEQ ID NO: 5.
  • the human proGDFl 1 ARM8 has an amino acid sequence as set forth in SEQ ID NO: 179.
  • the human proGDFl 1 has an amino acid sequence as set forth in SEQ ID NO: 4.
  • the murine latent myostatin has an amino acid sequence as set forth in SEQ ID NO: 88.
  • the human latent proGDFl 1 ARM8 has an amino acid sequence as set forth in SEQ ID NO: 179.
  • the antibody comprises a CDR-H3 amino acid sequence set forth in SEQ ID NO: 779. In other embodiments, the antibody comprises a CDR-L3 amino acid sequence set forth in any one of SEQ ID NO: 1393. In some embodiments, the antibody comprises a CDR-H1, a CDR-H2, and/or a CDR-H3 from the variable heavy chain (VH) amino acid sequence of clone BP21b-l as set forth in Table 14, and/or a CDR-L1, a CDR-L2, and/or a CDR-L3 from the variable light chain (VL) amino acid sequence of clone BP21b-l as set forth in Table 16.
  • VH variable heavy chain
  • VL variable light chain
  • the antibody comprises a variable heavy chain amino acid sequence of clone BP21b-l as set forth in Table 14. In other embodiments, the antibody comprises the variable light chain amino acid sequence of clone BP21b-l as set forth in Table 16.
  • Additional aspects of the present disclosure relate to an antibody that specifically binds human proMyostatin, human latent Myostatin, human proGDFl 1, and human latent proGDFl 1 ARM8 but does not specifically bind human GDF-8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 38, human GDF-11 ARM8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 181, human proGDFl 1 ARM8 having an amino acid sequence as set forth in SEQ ID NO: 179 or human mature GDF-8 having an amino acid sequence as set forth in SEQ ID NO: 42.
  • Further aspects of the present disclosure include an antibody that specifically binds human proMyostatin, murine proMyostatin, human latent Myostatin, human proGDFl 1, murine latent myostatin and human latent proGDFl 1 ARM8 but does not specifically bind human GDF-8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 38, human GDF-11 ARM8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 181, human proGDFl 1 ARM8 having an amino acid sequence as set forth in SEQ ID NO: 179 or human mature GDF-8 having an amino acid sequence as set forth in SEQ ID NO: 42.
  • the human proMyostatin has an amino acid sequence as set forth in SEQ ID NO: 5. In other embodiments, the murine proMyostatin has an amino acid sequence as set forth in SEQ ID NO: 88. In another embodiment, the human latent Myostatin has an amino acid sequence as set forth in SEQ ID NO: 5. In some embodiments, the human proGDFl 1 has an amino acid sequence as set forth in SEQ ID NO: 4. In another embodiment, the murine latent myostatin has an amino acid sequence as set forth in SEQ ID NO: 88. In some embodiments, the human latent proGDFl 1 ARM8 has an amino acid sequence as set forth in SEQ ID NO: 179.
  • the antibody comprises a CDR-H3 amino acid sequence set forth in SEQ ID NO: 1006. In some embodiments, the antibody comprises a CDR-L3 amino acid sequence set forth in any one of SEQ ID NO: 1621. In some embodiments, the antibody comprises a CDR-H1, a CDR-H2, and/or a CDR-H3 from the variable heavy chain (VH) amino acid sequence of clone BP22b-l as set forth in Table 14, and/or a CDR-L1, a CDR-L2, and/or a CDR-L3 from the variable light chain (VL) amino acid sequence of clone BP22b-las set forth in Table 16.
  • VH variable heavy chain
  • VL variable light chain
  • the antibody comprises a variable heavy chain amino acid sequence of clone BP22b-l as set forth in Table 14. In other embodiments, the antibody comprises the variable light chain amino acid sequence of clone BP22b-l as set forth in Table 16. [0054] Additional aspects of the present disclosure relate to an antibody that specifically binds proMyostatin and/or latent Myostatin, in which the antibody has an affinity to the proMyostatin and/or latent Myostatin that is pH sensitive.
  • Further aspects of the present disclosure include an antibody that specifically binds proMyostatin and/or latent Myostatin with a greater affinity at a pH in a range from 6.5 to 7.5 than it does at a pH in a range from 4.6 to 6.0.
  • the antibody has a Kd of binding to pro/latent-Myostatin in a range fromlO-3 M to 10-8 M under binding conditions having a pH in a range from 4.6 to 6.0.
  • the antibody has a Kd of binding to pro/latent-Myostatin in a range fromlO-6 M to 10-11 M, when the pH is in a range from 6.5 to 7.5.
  • the antibody comprises a CDR-H3 amino acid sequence set forth in SEQ ID NO: 813, 804, 790, 762, 784, 770, 764, 986, 777, or 1006.
  • the antibody comprises a CDR-L3 amino acid sequence set forth in any one of SEQ ID NO: 1422, 1414, 1403, 1377, 1397, 1385, 1379, 1601, 1391, or 1621.
  • the antibody comprises a CDR-H1, a CDR-H2, and/or a CDR-H3 from the variable heavy chain (VH) amino acid sequence of clone BP2a-l, BP3a-2, BP5b-l, BP5b- 4, BP6a-5, BP6a-6, BPl la-1, BP16b-4, BP18b-2 or BP22b-l as set forth in Table 14, and/or a CDR-L1, a CDR-L2, and/or a CDR-L3 from the variable light chain (VL) amino acid sequence of clone BP2a-l, BP3a-2, BP5b-l, BP5b-4, BP6a-5, BP6a-6, BPl la-1, BP16b-4, BP18b-2 or BP22b-l as set forth in Table 16.
  • VH variable heavy chain
  • the antibody comprises a variable heavy chain amino acid sequence of clone BP2a-l, BP3a-2, BP5b-l, BP5b-4, BP6a-5, BP6a-6, BPl la-1, BP16b-4, BP18b-2 or BP22b-las set forth in Table 14.
  • the antibody comprises the variable light chain amino acid sequence of clone BP2a-l, BP3a-2, BP5b-l, BP5b-4, BP6a-5, BP6a-6, BPl la-1, BP16b-4, BP18b-2 or BP22b- las set forth in Table 16.
  • aspects of the present disclosure include an isolated antibody or antigen binding fragment thereof having at least one variable domain with an amino acid sequence having at least 95% sequence identity to one or more of those listed in Table 14 or 16.
  • the at least one variable domain comprises a variable domain pair, the variable domain pair comprising a heavy chain variable domain (VH) and a light chain variable domain (VL), in which the variable domain pair comprises at least 95% sequence identity to a variable domain pair present in an scFv selected from any of those listed in Table 13.
  • Additional aspects of the present disclosure include an isolated antibody or antigen binding fragment thereof comprising at least one variable domain, the at least one variable domain comprising at least one complementarity determining region (CDR) sequence comprising at least 70% sequence identity to one or more of those listed in Table 15 or 17.
  • the at least one CDR sequence comprises at least 70% sequence identity to a heavy chain variable domain CDR (CDR-H) selected from any of those listed in Table 15.
  • the isolated antibody or antigen binding fragment thereof comprises at least one variable domain pair, the variable domain pair comprising a VH and a VL.
  • the VH comprises at least one CDR-H sequence with at least 70% sequence identity to a sequence selected from any of those listed in Table 15.
  • the VL comprises at least one CDR-L sequence with at least 70% sequence identity to a CDR-L sequence selected from any of those listed in Table 17.
  • Further aspects of the present disclosure include an isolated antibody or antigen binding fragment thereof that comprises: a VH sequence with at least 95% sequence identity to a sequence listed in Table 14, and a VL sequence with at least 95% sequence identity to a sequence listed in Table 16.
  • an isolated antibody or antigen binding fragment thereof that comprises: a CDR-H1 sequence with at least 70% sequence identity to a sequence listed in Table 15, a CDR-H2 sequence with at least 70% sequence identity to a sequence listed in Table 15, a CDR-H3 sequence with at least 70% sequence identity to a sequence listed in Table 15, a CDR-L1 sequence with at least 70% sequence identity to a sequence listed in Table 17, a CDR-L2 sequence with at least 70% sequence identity to a sequence listed in Table 17, and a CDR-L3 sequence with at least 70% sequence identity to a sequence listed in Table 17.
  • the isolated antibody or antigen binding fragment thereof comprising an antibody, in which the antibody is a monoclonal antibody.
  • the antibody comprises an IgGl isotype.
  • the antibody binds to one or more recombinant antigens listed in Table 24 and/or one or more polypeptides comprising an amino acid sequence with at least 95% sequence identity to any of those listed in Tables 1, 2, 4, 5, 9, 10, 19, 20 or 25.
  • the polypeptide comprises pro-growth differentiation factor (GDF)-8 (proGDF-8) (SEQ ID NO: 5).
  • GDF pro-growth differentiation factor
  • the antibody binds to a latent GDF-8 growth factor prodomain complex (GPC).
  • Further aspects of the present disclosure include a method of modulating growth factor activity in a biological system comprising contacting the biological system with the antibody or antigen binding fragment thereof.
  • the growth factor activity comprises GDF-8 activity.
  • the antibody is a stabilizing antibody and in which contacting the biological system with the stabilizing antibody results in inhibition of release of at least 5% of total GDF-8 mature growth factor in the biological system.
  • the stabilizing antibody comprises at least one variable domain comprising a VH and VL pair selected from the group consisting of the VH of SEQ ID NO: 462 and the VL of SEQ ID NO: 1037, the VH of SEQ ID NO: 472 and the VL of SEQ ID NO: 1047, the VH of SEQ ID NO: 448 and the VL of SEQ ID NO: 1023, the VH of SEQ ID NO: 502 and the VL of SEQ ID NO: 1077, the VH of SEQ ID NO: 581 and the VL of SEQ ID NO: 1153, the VH of SEQ ID NO: 442 and the VL of SEQ ID NO: 1017, the VH of SEQ ID NO: 464 and the VL of SEQ ID NO: 1039, the VH of SEQ ID NO: 593 and the VL of SEQ ID NO: 1166, the VH of SEQ ID NO: 598 and the VL of SEQ ID NO: 1171, the VH of SEQ ID NO: 462
  • TGF- ⁇ -related indication comprises a muscle disorder and/or injury.
  • the muscle disorder and/or injury comprises a muscle disorder selected from the group consisting of cachexia, inclusion body myositis (IBM) and sarcopenia.
  • the TGF-P-related indication comprises chronic obstructive pulmonary disease (COPD).
  • COPD chronic obstructive pulmonary disease
  • GDF growth differentiation factor
  • Fc-fusion protein comprises the sequence:
  • the recombinant antigen comprises a sequence selected from any of those presented in Table 12.
  • Additional aspects of the present disclosure include a recombinant antigen comprising a chimeric protein selected from any of those in Tables 20 and 21.
  • Yet another aspect of the present disclosure relates to an isolated antibody or antigen binding fragment thereof that binds to a recombinant antigen described above.
  • the recombinant antigen comprises latent GDF- 11 growth factor prodomain complex (GPC).
  • a further aspect of the present disclosure includes a method of modulating growth factor activity in a biological system comprising contacting the biological system with the antibody or antigen binding fragment thereof described above.
  • growth factor activity comprises GDF-11 activity.
  • an isolated antibody or antigen binding fragment thereof is a stabilizing antibody and in which contacting the biological system with the stabilizing antibody results in inhibition of release of at least 5% of total GDF-11 mature growth factor in the biological system.
  • an antibody is a releasing antibody and in which contacting the biological system with the releasing antibody results in release of at least 5% of total GDF-11 mature growth factor in the biological system.
  • a further aspect of the present disclosure includes a method of treating a TGF- ⁇ - related indication in a subject.
  • the methods involve administering to the subject any one or more of the antibodies, including antigen binding fragments, disclosed herein.
  • the TGF-P-related indication comprises a cardiovascular indication selected from the group consisting of cardiac hypertrophy, cardiac atrophy, atherosclerosis and restenosis.
  • the TGF-P-related indication comprises a GDF-11 -related indication.
  • the GDF-11 -related indication comprises anemia and/or ⁇ -thalassemia.
  • method of enhancing skeletal muscle rejuvenation, cerebral circulation, neurogenesis and/or erythropoiesis involve administering to a subject any one or more of the antibodies, including antigen binding fragments, disclosed herein.
  • the antibody is a monoclonal antibody.
  • the antibody comprises an IgG isotype.
  • the antibody is a bispecific antibody.
  • the isolated antibody or antigen binding fragment comprises at least one variable domain comprising a VH and VL pair selected from the group consisting of the VH of SEQ ID NO: 462 and the VL of SEQ ID NO: 1037, the VH of SEQ ID NO: 472 and the VL of SEQ ID NO: 1047, the VH of SEQ ID NO: 448 and the VL of SEQ ID NO: 1023, the VH of SEQ ID NO: 502 and the VL of SEQ ID NO: 1077, the VH of SEQ ID NO: 581 and the VL of SEQ ID NO: 1153, the VH of SEQ ID NO: 442 and the VL of SEQ ID NO: 1017, the VH of SEQ ID NO: 464 and the VL of SEQ ID NO: 1039, the VH of SEQ ID NO
  • compositions that comprise an antibody (or antigen binding fragment) and an excipient or carrier.
  • the composition is a pharmaceutical composition comprising (i) a therapeutically effective amount of an antibody and (ii) a pharmaceutically acceptable carrier.
  • the composition is for use in preventing muscle wasting or increasing muscle mass, and comprises a therapeutically effective amount of an antibody.
  • the carrier is a pharmaceutically acceptable carrier.
  • the antibody and carrier are in a lyophilized form.
  • the antibody and carrier are in solution.
  • the antibody and carrier are frozen. In some embodiments, the antibody and carrier are frozen at a temperature less than or equal to -65°C.
  • nucleic acids are provided that encode an antibody (or antigen binding fragment) or recombinant antigen.
  • Vectors and cells comprising such nucleic acids are also provided.
  • Kits comprising any of the compositions disclosed herein are also provided.
  • Methods of developing an antibody comprise: selecting a protein/antigen, subjecting the protein/antigen to structural analysis, identifying at least one epitope, and developing at least one antibody against the at least one epitope.
  • the protein is subjected to prodomain convertase cleavage prior to structural analysis.
  • the prodomain convertase cleaved protein is further subjected to cleavage with an enzyme selected from the group consisting of BMP- 1, mammalian tolloid protein (mTLD), mammalian tolloid-like 1 (mTLLl) and mammalian tolloid-like 2 (mTLL2).
  • the protein comprises proGDF-8 (SEQ ID NO: 5) or proGDF-11 (SEQ ID NO: 4).
  • an inhibitor of at least one BMP- 1/Tolloid- like proteinase (B/TP) is provided.
  • the inhibitor comprises a polypeptide.
  • the inhibitor comprises an inhibiting antibody, e.g., an anti-primed complex antibody.
  • the inhibitor prevents cleavage of a latent GPC.
  • the latent GPC is selected from the group consisting of latent GDF-8 and latent GDF-11.
  • the inhibitor prevents cleavage of a BMP/Tolloid cleavage site on the latent GPC.
  • the B/TP is selected from the group consisting of BMP- 1, mammalian tolloid protein (mTLD), mammalian tolloid-like 1 (mTLLl), and mammalian tolloid-like 2 (mTLL2).
  • antibodies are provided that bind at or within 10 amino acid residues of a proprotein convertase cleavage site of proMyostatin or latent Myostatin.
  • the proprotein convertase cleavage site comprises the amino acid sequence (SEQ ID NO: 1667 or 1668).
  • antibodies are provided that bind at or within 10 amino acid residues of a proprotein convertase docking site of proMyostatin or latent Myostatin.
  • the proprotein convertase docking site comprises the amino acid sequence (SEQ ID NO: 1667 or 1668).
  • antibodies are provided that bind at or within 10 amino acid residues of a tolloid protease cleavage site of proMyostatin or latent Myostatin.
  • the tolloid protease cleavage site comprises the amino acid sequence (SEQ ID NO: 1665 or 1666).
  • antibodies are provided that bind at or within 10 amino acid residues of a tolloid protease docking site of proMyostatin or latent Myostatin.
  • the tolloid protease docking site comprises the amino acid sequence (SEQ ID NO: 1665 or 1666).
  • binding of the antibody to the proMyostatin or the latent Myostatin inhibits proteolytic cleavage of the proMyostatin or the latent Myostatin by a proprotein convertase or a tolloid protease.
  • the disclosure relates to antibodies that compete for binding to an epitope with any one of the antibodies disclosed herein.
  • the competing antibodies bind to an epitope of human proMyostatin or an epitope of human latent Myostatin.
  • the competing antibodies specifically bind to an epitope of human proMyostatin or human latent Myostatin.
  • the competing antibodies bind to the epitope with an equilibrium dissociation constant (Kd) between the antibody and the epitope of less than 10 "6 M or in a range of 10 "11 M to 10 "6 M.
  • Kd equilibrium dissociation constant
  • the antibodies comprise an Fc portion.
  • the antibodies bind the neonatal Fc receptor FcRn. In some embodiments, the Fc portion binds the neonatal Fc receptor FcRn. In some embodiments, the antibodies bind FcRn at a pH greater than 6.0. In some embodiments, the antibodies bind FcRn at a pH in a range from 7.0 to 7.5. In some embodiments, the Kd of binding of the antibody to the FcRN is in a range from 10 " 3 J M to 10 8 M. In some embodiments, the Kd of binding of the antibody to the FcRN is in a range from HT -4 M to 10 -8 M.
  • the Kd of binding of the antibody to the FcRN is in a range from 10 -5 J M to 10-8 M. In some embodiments, the Kd of binding of the antibody to the FcRN is in a range from 10 "6 M to 10 "8 M.
  • the antibody comprises a CDR-H3 amino acid sequence set forth in SEQ ID NO: 813, 804, 790, 762, 784, 770, 764, 986, 777, or 1006. In some embodiments, the antibody comprises a CDR-L3 amino acid sequence set forth in any one of SEQ ID NO: 1422, 1414, 1403, 1377, 1397, 1385, 1379, 1601, 1391, or 1621.
  • the antibody comprises a CDR-H1, a CDR-H2, and/or a CDR-H3 from the variable heavy chain (VH) amino acid sequence of clone BP2a-l, BP3a-2, BP5b-l, BP5b-4, BP6a-5, BP6a-6, BPl la-1, BP16b-4, BP18b-2 or BP22b-l as set forth in Table 14, and/or a CDR-Ll, a CDR-L2, and/or a CDR-L3 from the variable light chain (VL) amino acid sequence of clone BP2a-l, BP3a-2, BP5b-l, BP5b-4, BP6a-5, BP6a-6, BPl la-1, BP16b-4, BP18b-2 or BP22b-l as set forth in Table 16.
  • VH variable heavy chain
  • the antibody comprises a variable heavy chain amino acid sequence of clone BP2a-l, BP3a-2, BP5b-l, BP5b-4, BP6a-5, BP6a-6, BPl la-1, BP16b-4, BP18b-2 or BP22b-las set forth in Table 14.
  • the antibody comprises a variable light chain amino acid sequence of clone BP2a-l, BP3a-2, BP5b-l, BP5b-4, BP6a-5, BP6a-6, BPl la-1, BP16b-4, BP18b-2 or BP22b-l as set forth in Table 16.
  • FIG. 1 is a diagram of the TGF-beta superfamily tree, where divergence is proportional to branch length.
  • FIG. 2 is a schematic of one embodiment of a linear representation of a translated growth factor monomer.
  • translated growth factors may comprise secretion signal peptides, prodomains and growth factor domains.
  • translated growth factors may also comprise a cleavage site between prodomain and growth factor regions.
  • FIG. 3 is a schematic of one embodiment of a GDF growth factor-prodomain complex (GPC) made up of proGDF monomers as well as an embodiment of a free growth factor dimer and free prodomains, separated after proteolytic cleavage of the GPC.
  • GPC GDF growth factor-prodomain complex
  • FIG. 4 is a schematic of one embodiment of a GDF GPC as well as an embodiment of a free growth factor dimer and free prodomains, separated after proteolytic cleavage.
  • FIG. 5 presents 3 tables showing the percent identity between amino acid sequences found in the TGF- ⁇ family.
  • FIG. 5A demonstrates percent identity among pro- proteins (prodomain and growth factor). Percent identity among growth factor domains is presented in FIG. 5B while percent identity among prodomains is presented in FIG. 5C.
  • FIG. 6 shows structure-based alignment between TGF- ⁇ family member proteins [adapted from Shi et al (Shi, M. et al., Latent TGF- ⁇ structure and activation. Nature. 2011 Jun 15; 474(7351):343-9, the contents of which are herein incorporated by reference in their entirety)] .
  • Cysteine residues required for interaction with LTBPs and/or GARPs are boxed.
  • Residues mutated in Camurati-Engelmann syndrome are indicated with a star.
  • Protease cleavage sites are indicated with an up arrow.
  • Protein modules and secondary structural elements are indicated with solid bars.
  • Residues underlined at the N-terminus of GDF-8 correspond to alternatively predicted signal peptide processing sites.
  • “Chimeric module breakpoints” indicate regions where structural features are conserved and provide modules for chimeric protein construction (swapping of modules between family members) in all family members. N-terminal regions are shown in (A), internal regions are shown in (B) and C-terminal regions are shown in (C).
  • FIG. 7 is a schematic of an embodiment of a recombinant GPC.
  • FIG. 8 is a schematic of embodiments of mutant recombinant proteins of the disclosure.
  • FIG. 9 depicts the formation of a latent complex after furin cleavage of a pro- protein dimer.
  • the latent complex retains the growth factor dimer until subjected to an activating cleavage [e.g. by members of the BMP- 1/Tolloid- like proteinase (B/TP) family.]
  • FIG. 10 presents an alignment conducted between GDF-8 (myostatin), GDF-11, Inhibin A and a GDF-8 dimer. Arrows indicate cleavage sites. Regions involved in internal interactions are boxed. Solid rectangles appear above residues predicted to be involved in steric clashes in chimeric constructs. Stars denote important break points in protein modules.
  • FIG. 11 depicts recombinant histidine-tagged proGDF-8 (referred to in the figure as proMyostatin), separated by SDS-PAGE under reducing and non-reducing conditions, as visualized by colloidal blue staining.
  • proMyostatin histidine-tagged proGDF-8
  • FIG. 12 is a graph showing CAGA promoter-dependent luciferase activity is the presence of GDF-11 or proGDF-11 after treatment with proprotein convertase, Tolloid proteinase or a combination of proprotein convertase and Tolloid proteinase.
  • FIG. 13 is a stained gel showing separation of proteinase treated proGDF-11 under reducing and non-reducing conditions.
  • FIG. 14 presents results of a luciferase-based growth factor activity assay.
  • FIG. 15 is a schematic showing formation of the "primed" complex.
  • the primed complex is an open, active conformation of GDF-8 GPCs or GDF-11 GPCs. Primed complexes are capable of receptor binding and inducing growth factor activity.
  • FIG. 16 is a graph showing that BP6a-5 dissociates rapidly at pH 5. The antibody blocks myostatin activation and promotes muscle growth in vivo.
  • FIG. 17 is a graph showing that BP6a-6 dissociates rapidly at pH 5.
  • the antibody does not block Myostatin activation or signaling and is in the same epitope bin (by ELISA binding profile) as BP6a-5.
  • FIG. 18 displays eight antibodies that dissociate more rapidly at pH5 that at pH 7.4.
  • the asterisk indicates some cross-reactivity with proGDFl 1 in ELISA and the bold indicates that the antibody inhibits GDF8 activation.
  • FIG. 19 shows CDR-H3 sequences of pH-sensitive antibodies. All listed antibodies have a histidine in the CDR-H3 sequence except for BP5b- 1. The sequences, from top to bottom, correspond to SEQ ID NOs: 762, 764, 770, 777, 784, 986, 1006, and 790. Note that BP18b-2 only includes one histidine in the entire variable region, while BP5b-l does not contain any histidines in the variable region. These two antibodies have the weakest pH sensitivity profiles.
  • FIG. 20 is a table of four antibodies for in vivo characterization of Myostatin sweeping. All antibodies will be produced both with and without the "sweeping" mutations in the constant region. Light grey indicates the antibody inhibits GDF8 activation.
  • FIG. 21 is a graph showing that BP1 la- 1 dissociates rapidly at pH5. The antibody does not block Myostatin activation or signaling, but does bind to proMyostatin, latent myostatin, and mature myostatin with equal affinity.
  • FIG. 22 is a graph showing that BP5b-4 dissociates rapidly at pH5.
  • the antibody does not block Myostatin activation or signaling; however, it is a different epitope group from BP1 la- 1 and BP6a-5.
  • FIG. 23 shows an example of an antibody (BP6a-3) with no pH-sensitive binding.
  • FIGs. 24A-24L show the different pH sensitivities of specific antibodies: BP6a-3 (FIG. 24A), BP5a-l (FIG. 24B), BP6a- l l (FIG. 24C), BP5b-4 (FIG. 24D), BPl la-1 (FIG. 24E), BP6a-6 (FIG. 24F), BP18b-2 (FIG. 24G), BP6a-2 (FIG. 24H), BP6a-5 (FIG. 241), BP14a-l (FIG. 24J), BP5b-2 (FIG. 24K), and BP18b-7 (FIG. 24L).
  • FIGs. 25A-25K show the different pH sensitivities of specific antibodies: BP16b-4 (FIG. 25 A), BP6b-9 (FIG. 25B), BP22b- l (FIG. 25C), BP3a-2 (FIG. 25D), BP5b- l (FIG. 25E), BP6a-4 (FIG. 25F), BP6a-7 (FIG. 25G), BPla-1 (FIG. 25H), BP2a- l (FIG. 251), BP5a- 2 (FIG. 25J), and BP6a- l (FIG. 25K).
  • antibodies are selected for use (e.g. , in a particular assay) based on the fact that they have a known and/or desired binding profile (a selected binding profile).
  • binding profile refers to a set of one or more parameters (e.g. , symbols, quantities, measurements, etc.) indicative of the extent to which an antibody specifically binds to one or more antigens.
  • a parameter indicative of the binding of an antibody for a target antigen is an IC50 or EC50 value.
  • a parameter indicative of the binding of an antibody for a target antigen is an equilibrium dissociation constant (Kd).
  • a parameter indicative of the binding is an equilibrium association constant (Ka).
  • Ka equilibrium association constant
  • the disclosure relates to antibodies having a selected myo statin-related binding profile, which comprises a set of one or more parameters indicative of the extent to which an antibody specifically binds to one or more antigens, at least one of which antigens is a myostatin-related protein (e.g., Myostatin, GDF11, etc.).
  • a binding profile comprises one or more parameters indicative of whether or not an antibody exhibits a threshold level of binding (e.g., specific binding) to one or more antigens.
  • a threshold level of binding is a level of binding that above (or below, depending on the parameter) a control or reference level of binding (e.g., background or non-specific binding). In some embodiments, a threshold level of binding is a level of binding that is at least 1, at least 2, at least 3, at least 4, at least 5, at least 6, at least 7 or more standard deviations greater than (or less than, depending on the assay and/or parameter) a control or reference level of binding (e.g., background or non-specific binding), as measured by an appropriate immunoassay.
  • a threshold level of binding is a level of binding that is in a range of 1 to 10 standard deviations, 2 to 10 standard deviations , or 4 to 6 standard deviations greater than (or less than, depending on the assay and/or parameter) a control or reference level of binding (e.g., background or non-specific binding), as measured by an appropriate immunoassay.
  • a threshold level of binding is determined through an appropriate immunoassay.
  • an appropriate immunoassay assesses the binding affinity of an antibody for a target antigen.
  • an appropriate immunoassay is an enzyme linked immune- sorbent assay.
  • an appropriate immunoassay is an assay that determines a kinetic measurement (e.g., on rate, off rate) indicative of binding between an antibody and antigen.
  • an appropriate immunoassay is an assay, such as an Octet assay, that determines one or more a kinetic parameters indicative of binding between an antibody and antigen.
  • an appropriate immunoassay is a cell-based assay that determines one or more a parameters indicative of specific binding between an antibody and antigen based on cellular activity, e.g., growth factor signaling (such as SMAD signaling), cell growth, cell survival, gene expression, reporter expression, protein production, protein secretion, etc.
  • an appropriate immunoassay is an in vivo assay that determines one or more a parameters indicative of specific binding between an antibody and antigen based on cellular, tissue or other physiological activity.
  • a binding profile comprises a set of one or more parameters indicative of the extent to which an antibody specifically binds to one or more one or more TGFP family member proteins or forms thereof, one or more portions or domains of TGFP family member proteins and/or one or more chimeras of TGFP family member proteins.
  • a binding profile comprises a set of one or more parameters indicative of the extent to which an antibody specifically binds or does not specifically bind to one or more different antigens.
  • a binding profile relates to the extent to which an antibody specifically binds or does not specifically bind to 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, or more different antigens. In some embodiments, a binding profile relates to the extent to which an antibody specifically binds or does not specifically bind to 5, 6, 7, 8, 9, 10 or 11 different antigens.
  • a particular binding profile comprises a set of one or more parameters indicative of the extent to which an antibody specifically binds to one or more TGFP family member proteins or forms thereof.
  • a TGFP family member proteins is selected from the group consisting of AMH, ARTN, BMP10, BMP15, BMP2, BMP3, BMP4, BMP5, BMP6, BMP7, BMP8A, BMP8B, GDF1, GDF10, GDF11, GDF15, GDF2, GDF3, GDF3A, GDF5, GDF6, GDF7, GDF8, GDF9, GDNF, INHA, INHBA, INHBB, INHBC, INHBE, LEFTY 1, LEFTY2, NODAL, NRTN, PSPN, TGFpi, TGFP2, and TGFP3 protein.
  • TGFP family member proteins or forms thereof are from a vertebrate organism.
  • TGFP family member proteins or forms thereof are from a vertebrate organism.
  • TGFP family member proteins or forms thereof
  • TGFP family member proteins or forms thereof are from a human or a mouse. In some embodiments, TGFP family member proteins or forms thereof are from a human. Examples of sequences of human and non-human TGFP family member proteins are shown in Tables 1, 4 and 5, provided herein. In some embodiments, TGFP family member proteins or forms thereof may include any naturally- occurring isoforms or variants of TGFP family member proteins. In some embodiments, TGFP family member proteins comprise an amino acid sequence that is at least 70%, 75%, 80%, 85%, 90%, 95%, 98%, or 99% identical to any of the amino acid sequences as set forth in any one of SEQ ID NOs: 1-37 and 70-115.
  • TGFP family member proteins comprise an amino acid sequence as set forth in any one of SEQ ID NOs: 1-37 and 70-115. In some embodiments, TGFP family member proteins consist of an amino acid sequence as set forth in any one of SEQ ID NOs: 1-37 and 70-115.
  • a particular binding profile may also comprise one or more parameters indicative of to the extent to which antibodies specifically bind to or do not specifically bind to a portion or domain of one or more TGFP family member proteins.
  • the portion or domain of a TGFP family member protein is a prodomain, a straight jacket region, a growth factor domain, a fastener region, a furin cleavage site region, a bmp/tolloid cleavage site, an arm region, a fingers region 1, a fingers region 2, a latency loop, an alpha 1 helical region, and/or a bowtie region.
  • Exemplary portions or domains of TGFP family member proteins are shown in Tables 2, 3, 9 and 11, provided herein.
  • portions or domains of TGFP family member proteins are portions or domains of GDF proteins. In some embodiments, portions or domains of TGFP family member proteins are portions or domains of GDF8 and/or GDF11. In some embodiments, portions or domains of TGFP family member proteins are portions or domains of Inhibin beta A. However, it should be appreciated that the portions or domains of TGFP family member proteins may be from any TGFP family member protein provided herein.
  • the portion or domain of a TGFP family member protein comprises an amino acid sequence that is at least 70%, 75%, 80%, 85%, 90%, 95%, 98%, or 99% identical to any of the amino acid sequences as set forth in any one of SEQ ID NOs: 38-65, 162-177 and 202-221.
  • portions or domains of TGFP family member proteins comprise an amino acid sequence as set forth in any one of SEQ ID NOs: 38-65, 162-177 and 202-221.
  • portions or domains of TGFP family member proteins consist of an amino acid sequence as set forth in any one of SEQ ID NOs: 38-65, 162- 177 and 202-221.
  • forms of TGFP family member proteins refer to complexes of TGFP family member proteins.
  • forms of TGFP family member proteins may include pro-forms, latent-forms, primed-forms, or mature forms of dimeric TGFP family member proteins, such as, for example, proMyostatin, proGDFl 1, latent Myostatin, latent GDF11, primed Myostatin, and primed GDF11.
  • TGFP family member proteins form dimeric complexes.
  • TGFP family member proteins form homodimeric complexes.
  • TGFP family member proteins form heterodimeric complexes.
  • Dimeric forms of TGFP family member proteins may include TGFP family member proteins that are full-length or TGFP family member proteins that have been cleaved (e.g., by a proprotein convertase and/or a tolloid protease).
  • forms of TGFP family member proteins are pro forms of TGFP family member proteins (e.g., proMyostatin or proGDFl 1).
  • forms of TGFP family member proteins include full-length TGFP family member proteins.
  • pro forms of TGFP family member proteins include, without limitation, proMyostatin and proGDFl 1 that have not been cleaved at a proprotein convertase cleavage site (e.g., by a proprotein convertase such as furin) or a tolloid protease cleavage site (e.g., by a tolloid protease such as BMP-1).
  • forms of TGFP family member proteins are latent forms of TGFP family member proteins (e.g., latent Myostatin or latent GDF11).
  • forms of TGFP family member proteins include TGFP family member proteins that have been cleaved (e.g., by a proprotein convertase).
  • latent forms of TGFP family member proteins include, without limitation, latent Myostatin and latent GDFl 1 that have been cleaved at a proprotein convertase cleavage site (e.g., by a proprotein convertase such as furin) but not at a tolloid protease cleavage site.
  • forms of TGFP family member proteins are primed forms of TGFP family member proteins (e.g., primed Myostatin or primed GDFl 1).
  • forms of TGFP family member proteins include TGFP family member proteins that have been cleaved (e.g., by a proprotein convertase and/or a tolloid protease).
  • primed forms of TGFP family member proteins include, without limitation, primed Myostatin and primed GDFl 1 that have been cleaved at a proprotein convertase cleavage site (e.g., by a proprotein convertase such as furin) and a tolloid protease cleavage site (e.g., by a tolloid protease such as BMP-1).
  • forms of TGFP family member proteins are mature forms of TGFP family member proteins (e.g., mature Myostatin or mature GDFl 1).
  • forms of TGFP family member proteins include TGFP family member proteins that have been cleaved (e.g., by a proprotein convertase and/or a tolloid protease) and are not in complex with one or more portions of a prodomain of a TGFP family member protein.
  • mature forms of TGFP family member proteins include, without limitation, mature Myostatin and mature GDFl 1 that have been cleaved at a proprotein convertase cleavage site (e.g., by a proprotein convertase such as furin), a tolloid protease cleavage site (e.g., by a tolloid protease such as BMP-1), and are not in complex with a prodomain of a TGFP family member protein.
  • a proprotein convertase cleavage site e.g., by a proprotein convertase such as furin
  • a tolloid protease cleavage site e.g., by a tolloid protease such as BMP-1
  • a particular binding profile comprises a set of one or more parameters indicative of the extent to which antibodies specifically bind to or do not specifically bind to chimeras of TGFP family member proteins.
  • chimeras of TGFP family member proteins can be used to provide information relating to particular epitopes to which any of the antibodies provided herein specifically bind or do not specifically bind.
  • chimeric TGFP family member proteins comprise at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14 or 15 portions or domains of different TGFp family member protein.
  • a chimeric TGFP family member protein may comprise a prodomain, a straight jacket region, a growth factor domain, a fastener region, a furin cleavage site region, a bmp/tolloid cleavage site, an arm region, a fingers region 1, a fingers region 2, a latency loop, an alpha 1 helical region, and/or a bowtie region of one TGFP family member protein and at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14 or 15 of a prodomain, a straight jacket region, a growth factor domain, a fastener region, a furin cleavage site region, a bmp/tolloid cleavage site, an arm region, a fingers region 1, a fingers region 2, a latency loop, an alpha 1 helical region, and/or a bowtie region from one or more different TGFP family member proteins.
  • chimeras of TGFP family member proteins are shown in Table 10, provided herein.
  • chimeras of TGFP family member proteins comprise an amino acid sequence that is at least 70%, 75%, 80%, 85%, 90%, 95%, 98%, or 99% identical to any of the amino acid sequences as set forth in any one of SEQ ID NOs: 178-201.
  • chimeras of TGFP family member proteins comprise an amino acid sequence as set forth in any one of SEQ ID NOs: 178-201.
  • chimeras of TGFP family member proteins consist of an amino acid sequence as set forth in any one of SEQ ID NOs: 178-201.
  • TGFP family member proteins include Fc fusion proteins to any of the TGFP family member proteins or forms or domains or portions thereof.
  • TGFP family member Fc fusion proteins are shown in Table 12, provided herein.
  • TGFP family member Fc fusion proteins comprise an amino acid sequence that is at least 70%, 75%, 80%, 85%, 90%, 95%, 98%, or 99% identical to any of the amino acid sequences as set forth in any one of SEQ ID NOs: 223-226.
  • TGFP family member Fc fusion proteins comprise an amino acid sequence as set forth in any one of SEQ ID NOs: 223-226.
  • TGFP family member Fc fusion proteins consist of an amino acid sequence as set forth in any one of SEQ ID NOs: 223-226.
  • a particular binding profile comprises a set of one or more parameters indicative of the extent to which antibodies specifically bind to or do not specifically bind to one or more of a human proMyo statin, a murine proMyostatin, a human latent Myostatin, a human GDF-8 prodomain, a human GDF-11 ARM8 prodomain, a human proGDFl 1 ARM8, a human mature GDF-8, a human proGDFl 1, a murine latent myostatin and/or a human latent proGDFl 1 ARM8.
  • parameters indicative of the extent of binding to one or more murine antigens can be removed from a binding profile.
  • a particular binding profile comprises a set of one or more parameters indicative of the extent to which antibodies specifically bind to or do not specifically bind to one or more of a human proMyostatin, a human latent
  • Myostatin a human GDF-8 prodomain, a human GDF-11 ARM8 prodomain, a human proGDFl 1 ARM8, a human mature GDF-8, a human proGDFl 1, and/or a human latent proGDFl 1 ARM8.
  • a particular binding profile relates to an extent to which antibodies specifically bind to or do not specifically bind to one or more of a protein that comprises an amino acid sequence that is at least 70%, 75%, 80%, 85%, 90%, 95%, 98%, or 99% identical to a human proMyostatin, a murine proMyostatin, a human latent Myostatin, a human GDF-8 prodomain, a human GDF- 11 ARM8 prodomain, a human proGDFl 1 ARM8, a human mature GDF-8, a human proGDFl 1, a murine latent myostatin and/or a human latent proGDFl 1 ARM8.
  • the human proMyostatin comprises an amino acid sequence as set forth in SEQ ID NO: 5.
  • the murine proMyostatin comprises an amino acid sequence as set forth in SEQ ID NO: 88.
  • the human latent Myostatin comprises an amino acid sequence as set forth in SEQ ID NO: 5.
  • the human GDF-8 prodomain comprises an amino acid sequence as set forth in SEQ ID NO: 38.
  • the human GDF- 11 ARM8 prodomain comprises an amino acid sequence as set forth in SEQ ID NO: 181.
  • the human proGDFl 1 ARM8 comprises an amino acid sequence as set forth in SEQ ID NO: 179.
  • the human mature GDF-8 comprises an amino acid sequence as set forth in SEQ ID NO: 42.
  • the human proGDFl 1 comprises an amino acid sequence as set forth in SEQ ID NO: 4.
  • the murine latent myostatin comprises an amino acid sequence as set forth in SEQ ID NO: 88.
  • the human latent proGDFl 1 ARM8 comprises an amino acid sequence as set forth in SEQ ID NO: 179.
  • antibodies provided herein have a binding profile as set forth in Table 33.
  • a binding profile may comprises one or more symbols (e.g., +, -, +/-) indicative of the extent to which an antibody binds to an antigen.
  • binding of an antibody to an antigen at a level detectable beyond a threshold level may be indicated by a "+”.
  • a "-" indicates that the antibody does not bind the antigen at level detectable beyond a threshold in a particular assay (e.g. , is less than 2, 3, 4 or 5 standard deviations beyond a reference level, e.g. , an assay background level).
  • a "+/-" indicates that an antibody is at or near a threshold of binding the antigen as determined by a particular assay (e.g. , within 2 to 5, 3 to 5, or 4 to 5 standard deviations of a reference level).
  • antibodies that "specifically binds" to a target antigen binds to the target antigen with greater affinity, avidity, more readily, and/or with greater duration than it binds to non- target antigens.
  • antibodies provided herein have particular binding profiles, e.g. , based on whether they specifically bind or do not specifically bind to one or more TGFP family member proteins or forms thereof, one or more portions or domains of TGFP family member proteins and/or one or more chimeras of TGFP family member proteins.
  • an antibody specifically binds an antigen if binding to that antigen is detected above a background level (e.g., of a control antigen) using an in vitro binding assay (e.g., an ELISA). In some embodiments, an antibody specifically binds an antigen if binding to that antigen is detected at least one, at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, or at least 10 standard deviations above a background level (e.g., of a control antigen) using an in vitro binding assay.
  • an antibody specifically binds an antigen if binding to that antigen is detected at least one, at least 5 standard deviations above a background level (e.g., of a control antigen) using an in vitro binding assay.
  • the in vitro binding assay is an enzyme linked immunosorbent assay (ELISA).
  • ELISA enzyme linked immunosorbent assay
  • the ELISA is performed as described in Example 29, provided herein. However, it should be appreciated that additional methods for determining the binding affinity of a protein to an antigen are also within the scope of this disclosure
  • an antibody specifically binds to an antigen (e.g., proMyo statin) if it binds that antigen with a higher affinity as compared to another antigen (e.g., latent Myostatin). In some embodiments, an antibody specifically binds to an antigen if it binds to that antigen by at least 2-fold, 5-fold, 10-fold, 50-fold, 100-fold, 200-fold, 500- fold, or 1,000-fold higher than another antigen.
  • an antigen e.g., proMyo statin
  • an antibody specifically binds to an antigen (e.g., proMyostatin) if it binds that antigen with a higher affinity as compared to another antigen (e.g., latent Myostatin). In some embodiments, an antibody specifically binds to an antigen if it binds to that antigen with a dissociation constant (Kd) that is less than 10 "3 M, 10 "4 M, 10 "5 M, 10 "6 M, 10 "7 M, 10 "8 M
  • Kd dissociation constant
  • antibodies bind an antigen but cannot effectively eliminate the antigen from the plasma.
  • the concentration of the antigen in the plasma may be increased by reducing the clearance of the antigen.
  • antibodies e.g., sweeping antibodies
  • antibodies have an affinity to an antigen that is sensitive to pH. Such pH sensitive antibodies may bind to the antigen in plasma at neutral pH and dissociate from the antigen in an acidic endosome, thus reducing antibody-mediated antigen accumulation and/or promoting antigen clearance from the plasma.
  • sweeping antibodies refer to antibodies having both pH-sensitive antigen binding and at least a threshold level of binding to cell surface neonatal Fc receptor (FcRn) at neutral or
  • sweeping antibodies bind to the neonatal Fc receptor FcRn at neutral pH.
  • sweeping antibodies may bind to the FcRn at a pH ranging from 7.0 to 7.6.
  • sweeping antibodies can bind to an antigen at an antigen binding site and bind to a cellular FcRn via an Fc portion of the antibody.
  • sweeping antibodies may then be internalized, releasing antigen in an acidic endosome, which may be degraded.
  • a sweeping antibody, no longer bound to the antigen may then be released (e.g., by exocytosis) by the cell back into the serum.
  • FcRn in the vascular endothelia extends the half-life of a sweeping antibody.
  • vascular endothelial cells internalize sweeping antibodies, which in some embodiments are bound to an antigen such as Myostatin (e.g., proMyostatin, latent Myostatin or primed Myostatin).
  • Myostatin e.g., proMyostatin, latent Myostatin or primed Myostatin.
  • a sweeping antibody is recycled back into the bloodstream.
  • a sweeping antibody has an increased half-life (e.g. , in the serum of a subject) as compared to its conventional counterpart.
  • a conventional counterpart of a sweeping antibody refers the antibody from which the sweeping antibody was derived (e.g., prior to engineering the Fc portion of the conventional antibody to bind FcRn with greater affinity at pH 7).
  • a sweeping antibody has a half- life in the serum of a subject that is at least 1%, 5%, 10%, 15%, 20%, 25%, 35%, 50%, 75%, 100%, 150%, 200% or 250% longer as compared to its conventional counterpart.
  • an Fc portion of a sweeping antibody binds FcRn. In some embodiments, the Fc portion of a sweeping antibody binds to FcRn at a pH of 7.4 with a Kd
  • a sweeping antibody binds to FcRn at a pH of 7.4 with a Kd ranging fromlO "3 M to 10 "7 M, from 10 "3 M to 10 "6 M, from 10 "3 M to 10 "5 M, from 10 "3 M to 10 "4 M, from 10 "4 M to 10 ⁇ 8 M, from 10 "4 M to 10 "7 M, from 10 "4 M to 10 "6 M, from 10 "4 M to 10 "5 M, from 10 "5 M to 10 ⁇ 8 M, from 10 "5 M to 10 "7 M, from 10 "5 M to 10 “6 M, from 10 “6 M to 10 “8 M, from 10 “6 M to 10 "7 M, or from 10 "7 M to 10 “8 M.
  • FcRn binds to the CH2-CH3 hinge region of a sweeping antibody.
  • FcRn binds to the same region as proteinA or protein G.
  • FcRn binds to a different binding site from FcyRs.
  • the amino acid residues AA of a sweeping antibody Fc region are required for binding to FcRn. In some embodiments, the amino acid residues AA of a sweeping antibody Fc region affect binding to FcRn.
  • any of the antibodies provided herein are engineered to bind FcRn with greater affinity. In some embodiments, any of the antibodies provided herein are engineered to bind FcRn with greater affinity at pH 7.4. In some embodiments, the affinity of sweeping antibodies to FcRn is increased to extend their pharmacokinetic (PK) properties as compared to their conventional counterparts. For example, in some embodiments, sweeping antibodies elicit less adverse reactions due to their efficacy at lower doses. In some embodiments, sweeping antibodies are administered less frequently. In some embodiments, transcytosis of sweeping antibodies to certain tissue types are increased. In some
  • sweeping antibodies enhance efficiency of trans-placental delivery. In some embodiments, sweeping antibodies are less costly to produce.
  • any of the antibodies provided herein are engineered to bind FcRn with lower affinity. In some embodiments, any of the antibodies provided herein are engineered to bind FcRn with lower affinity at pH 7.4. In some embodiments, the affinity of sweeping antibodies to FcRn is decreased to shorten their pharmacokinetic (PK) properties as compared to their conventional counterparts. For example, in some embodiments, sweeping antibodies are more rapidly cleared for imaging and/or radioimmunotherapy. In some embodiments, sweeping antibodies promote clearance of endogenous pathogenic antibodies as a treatment for autoimmune diseases. In some embodiments, sweeping antibodies reduce the risk of adverse pregnancy outcome, which may be caused by transplacental transport of material fetus- specific antibodies.
  • PK pharmacokinetic
  • sweeping antibodies have decreased affinity to an antigen at low pH as compared to a neutral or physiological pH (e.g., pH 7.4). In some embodiments, sweeping antibodies have a decreased affinity to an antigen at an acidic pH (e.g. a pH ranging from 5.5 to 6.5) as compared to a physiological pH (e.g., pH 7.4). It should be appreciated that any of the antibodies provided herein can be engineered to dissociate from the antigen depending on changes in pH (e.g., pH sensitive antibodies). In some embodiments, sweeping antibodies provided herein are engineered to bind antigen dependent on pH. In some embodiments, sweeping antibodies provided herein are engineered to bind FcRn dependent on pH.
  • sweeping antibodies provided herein are internalized by endocytosis. In some embodiments, sweeping antibodies provided here are internalized by FcRn binding. In some embodiments, endocytosed sweeping antibodies release antigen in an endosome. In some embodiments, sweeping antibodies are recycled back to the cell surface. In some embodiments, sweeping antibodies remain attached to cells. In some embodiments, endocytosed sweeping antibodies are recycled back to the plasma. It should be appreciated that the Fc portion of any of the antibodies provided herein may be engineered to have different FcRn binding activity. In some embodiments, FcRn binding activity affects the clearance time of an antigen by a sweeping antibody. In some embodiments, sweeping antibodies may be long-acting or rapid-acting sweeping antibodies.
  • converting a conventional therapeutic antibody into a sweeping antibody reduces the efficacious dose. In some embodiments, converting a conventional therapeutic antibody into a sweeping antibody reduces the efficacious dose by at least 1%, 2%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, or 99%. In some embodiments, converting a conventional therapeutic antibody into a sweeping antibody reduces the efficacious dose by at least 1.5 fold, 2 fold, 3 fold, 4 fold, 5 fold, 6 fold, 8 fold, 10 fold, 15 fold, 20 fold, 50 fold or 100 fold.
  • selecting an appropriate dose of a sweeping antibody for therapy may be performed empirically.
  • a high dose of a sweeping antibody may saturate FcRn, resulting in antibodies which stabilize antigen in serum without being internalized.
  • a low dose of a sweeping antibody may not be therapeutically effective.
  • sweeping antibodies are administered once a day, once a week, once every two weeks, once every three weeks, once every four weeks, once every 6 weeks, once every 8 weeks, once every 10 weeks, once every 12 weeks, once every 16 weeks, once every 20 weeks, or once every 24 weeks.
  • any of the antibodies provided herein may be modified or engineered to be sweeping antibodies.
  • any of the antibodies provided herein may be converted into a sweeping antibody using any suitable method.
  • suitable methods for making sweeping antibodies have been previously described in Igawa et ah, (2013) "Engineered Monoclonal Antibody with Novel Antigen-Sweeping Activity In Vivo," PLoS ONE 8(5): e63236; and Igawa et al., "pH-dependent antigen-binding antibodies as a novel
  • Aspects of the disclosure relate to antibodies that compete or cross-compete with any of the antibodies provided herein.
  • the term "compete”, as used herein with regard to an antibody means that a first antibody binds to an epitope of a protein (e.g., latentMyo statin) in a manner sufficiently similar to the binding of a second antibody, such that the result of binding of the first antibody with its epitope is detectably decreased in the presence of the second antibody compared to the binding of the first antibody in the absence of the second antibody.
  • a first antibody can inhibit the binding of a second antibody to its epitope without that second antibody inhibiting the binding of the first antibody to its respective epitope.
  • each antibody detectably inhibits the binding of the other antibody with its epitope or ligand, whether to the same, greater, or lesser extent, the antibodies are said to "cross-compete” with each other for binding of their respective epitope(s).
  • Both competing and cross-competing antibodies are within the scope of this disclosure. Regardless of the mechanism by which such competition or cross-competition occurs (e.g., steric hindrance, conformational change, or binding to a common epitope, or portion thereof), the skilled artisan would appreciate that such competing and/or cross-competing antibodies are encompassed and can be useful for the methods and/or compositions provided herein.
  • an antibody binds at or near the same epitope as any of the antibodies provided herein. In some embodiments, an antibody binds near an epitope if it binds within 15 or fewer amino acid residues of the epitope. In some embodiments, any of the antibodies provided herein bind within 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14 or 15 amino acid residues of an epitope that is bound by any of the antibodies provided herein.
  • any of the antibodies provided herein bind at or near a tolloid cleavage site or at or near a tolloid docking site of a TGFP family member protein (e.g., proMyo statin or latent Myostatin).
  • a TGFP family member protein e.g., proMyo statin or latent Myostatin.
  • an antibody binds near a tolloid cleavage site or near a tolloid docking site if it binds within 15 or fewer amino acid residues of the tolloid cleavage site or tolloid docking site.
  • any of the antibodies provided herein bind within 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14 or 15 amino acid residues of a tolloid cleavage site or tolloid docking site.
  • an antibody binds at or near a tolloid cleavage site of GDF11.
  • an antibody may bind an amino acid sequence as set forth in SEQ ID NO: 1665.
  • an antibody binds at or near a tolloid cleavage site of GDF8.
  • an antibody may bind an amino acid sequence as set forth in SEQ ID NO: 1666.
  • PKAPPLRELIDQYDVQRDDSSDGSLEDDDYHAT SEQ ID NO: 1666.
  • binding of an antibody at or near a tolloid cleavage site or at or near a tolloid docking site of a TGFP family member protein inhibits cleavage of the TGFP family member protein, for example, by a tolloid protease (e.g., BMP-1).
  • a tolloid protease e.g., BMP-1
  • any of the antibodies provided herein bind at or near a proprotein convertase cleavage site or at or near a proprotein convertase docking site of a TGFP family member protein (e.g., proMyo statin or latent Myostatin)
  • a TGFP family member protein e.g., proMyo statin or latent Myostatin
  • an antibody binds near a proprotein convertase cleavage site or near a proprotein convertase docking site if it binds within 15 or fewer amino acid residues of the proprotein convertase cleavage site or proprotein convertase docking site.
  • any of the antibodies provided herein bind within 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14 or 15 amino acid residues of a proprotein convertase cleavage site or proprotein convertase docking site.
  • an antibody binds at or near a proprotein convertase cleavage site of GDF11.
  • an antibody may bind an amino acid sequence as set forth in SEQ ID NO: 1667.
  • GLHPFMELRVLENTKRSRRNLGLDCDEHSSESRC SEQ ID NO: 1667.
  • an antibody binds at or near a proprotein convertase cleavage site of GDF8.
  • an antibody may bind an amino acid sequence as set forth in SEQ ID NO: 1668.
  • GLNPFLEVKVTDTPKRSRRDFGLDCDEHSTESRC SEQ ID NO: 1668.
  • binding of an antibody at or near a proprotein convertase cleavage site or at or near a proprotein convertase docking site of a TGFP family member protein inhibits cleavage of the TGFP family member protein, for example, by a proprotein convertase (e.g., furin).
  • a proprotein convertase e.g., furin
  • an antibody competes or cross-competes for binding to any of the antigens provided hererin (e.g., one or more TGFP family member proteins or forms thereof, one or more portions or domains of TGFP family member proteins and/or one or more chimeras of TGFP family member proteins) with an equilibrium dissociation constant, Kd, between the antibody and the protein of less than 10 ⁇ 6 M.
  • the antigens provided hererin e.g., one or more TGFP family member proteins or forms thereof, one or more portions or domains of TGFP family member proteins and/or one or more chimeras of TGFP family member proteins
  • an antibody competes or cross-competes for binding to any of the antigens provided herein with a Kd in a range from 10 "11 M to 10 "6 M.
  • Any of the antibodies provided herein can be characterized using any suitable methods. For example, one method is to identify the epitope to which the antigen binds, or "epitope mapping.” There are many suitable methods for mapping and characterizing the location of epitopes on proteins, including solving the crystal structure of an antibody- antigen complex, competition assays, gene fragment expression assays, and synthetic peptide-based assays, as described, for example, in Chapter 11 of Harlow and Lane, Using Antibodies, a Laboratory Manual, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., 1999.
  • epitope mapping can be used to determine the sequence to which an antibody binds.
  • the epitope can be a linear epitope, i.e., contained in a single stretch of amino acids, or a conformational epitope formed by a three-dimensional interaction of amino acids that may not necessarily be contained in a single stretch (primary structure linear sequence).
  • Peptides of varying lengths e.g., at least 4-6 amino acids long
  • the epitope to which the antibody binds can be determined in a systematic screen by using overlapping peptides derived from the target antigen sequence and determining binding by the antibody.
  • the open reading frame encoding the target antigen is fragmented either randomly or by specific genetic constructions and the reactivity of the expressed fragments of the antigen with the antibody to be tested is determined.
  • the gene fragments may, for example, be produced by PCR and then
  • telomere binding domains transcribed and translated into protein in vitro, in the presence of radioactive amino acids.
  • the binding of the antibody to the radioactively labeled antigen fragments is then determined by immunoprecipitation and gel electrophoresis.
  • Certain epitopes can also be identified by using large libraries of random peptide sequences displayed on the surface of phage particles (phage libraries). Alternatively, a defined library of overlapping peptide fragments can be tested for binding to the test antibody in binding assays.
  • mutagenesis of an antigen binding domain, domain swapping experiments and alanine scanning mutagenesis can be performed to identify residues required, sufficient, and/or necessary for epitope binding.
  • domain swapping experiments can be performed using a mutant of a target antigen in which various fragments of TGFP family member proteins have been replaced (swapped) with sequences from related, but antigenically distinct proteins, such as another member of a TGFP family member protein.
  • a binding profile of an antigen By assessing a binding profile of an antigen, the importance of the particular antigen fragment to antibody binding can be assessed.
  • competition assays can be performed using other antibodies known to bind to the same antigen to determine whether an antibody binds to the same epitope as the other antibodies. Such competition assays would be apparent to the skilled artisan.
  • any of the suitable methods can be applied to determine whether any of the antibodies provided hererin binds one or more of the specific residues/segments of one or more TGFP family member proteins as described herein. Further, the interaction of an antibody with one or more of those defined residues in TGFP family member proteins can be determined by routine technology. For example, a crystal structure can be determined, and the distances between the residues in TGFP family member proteins and one or more residues in an antibody can be determined accordingly. Based on such distance, whether a specific residue in a TGFP family member protein interacts with one or more residues in an antibody can be determined. Further, suitable methods, such as competition assays and target mutagenesis assays can be applied to determine the preferential binding of a candidate antibody to a TGFP family member protein as compared to another TGFP family member protein.
  • aspects of the disclosure relate to methods for assessing a biological sample using one or more antibodies having a selected binding profile (e.g., a binding profile as outlined in Table 33).
  • antibodies are selected for use (e.g., in an immunoassay) based on the fact that they have a known and/or desired binding profile (a selected binding profile).
  • antibodies may have a selected binding profile to one or more TGFP family member proteins or forms thereof, one or more portions or domains of TGFP family member proteins and/or one or more chimeras of TGFP family member proteins.
  • antibodies with selected binding profiles may be used as research and/or diagnostic tools.
  • antibodies with selected binding profiles may be used to assess a level of a protein, a localization pattern of a protein ⁇ e.g., at the tissue, cellular, and/or sub-cellular level) and/or a ratio of a protein with respect to another protein.
  • antibodies with selected binding profiles may be used to assess the level of proMyo statin and the level of latent Myostatin, which may be used to determine a ratio of proMyo statin to latent Myostatin.
  • methods include obtaining one or more antibodies that have a selected binding profile and subjecting a biological sample to an immunoassay using the one or more antibodies with selected binding profiles.
  • At least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25 or more antibodies having a selected binding profile are used in an assay to assess a biological sample. In some embodiments, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24 or 25 antibodies having a selected binding profile are used in an assay to assess a biological sample.
  • a biological sample is subjected to an immunoassay using two or more antibodies having different selected binding profiles.
  • a biological sample is subjected to an immunoassay using 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24 or 25 antibodies having different selected binding profiles.
  • antibodies have a selected binding profile to one or more of a human proMyostatin, a human latent Myostatin, a human GDF-8 prodomain, a human GDF-11 ARM8 prodomain, a human proGDFl 1 ARM8, a human mature GDF-8, a human proGDFl 1, and/or a human latent proGDFl 1 ARM8.
  • antibodies have a selected binding profile to one or more of a human proMyostatin, a murine proMyostatin, a human latent Myostatin, a human GDF-8 prodomain, a human GDF-11 ARM8 prodomain, a human proGDFl 1 ARM8, a human mature GDF-8, a human proGDFl 1, a murine latent Myostatin and/or a human latent proGDFl 1 ARM8.
  • the human proMyostatin comprises an amino acid sequence as set forth in SEQ ID NO: 5.
  • the murine proMyostatin comprises an amino acid sequence as set forth in SEQ ID NO: 88.
  • the human latent Myostatin comprises an amino acid sequence as set forth in SEQ ID NO: 5.
  • the human GDF-8 prodomain comprises an amino acid sequence as set forth in SEQ ID NO: 38.
  • the human GDF- 11 ARM8 prodomain comprises an amino acid sequence as set forth in SEQ ID NO: 181.
  • the human proGDFl 1 ARM8 comprises an amino acid sequence as set forth in SEQ ID NO: 179.
  • the human mature GDF-8 comprises an amino acid sequence as set forth in SEQ ID NO: 42.
  • the human proGDFl 1 comprises an amino acid sequence as set forth in SEQ ID NO: 4.
  • the murine latent Myostatin comprises an amino acid sequence as set forth in SEQ ID NO: 88.
  • the human latent proGDFl 1 ARM8 comprises an amino acid sequence as set forth in SEQ ID NO: 179.
  • a biological sample is subjected to an immunoassay using two or more antibodies having different selected binding profiles.
  • antibodies have selected binding profiles as shown in Table 33.
  • antibodies have selected binding profiles of any one of the antibodies Bla-1, BPlb-1, BP2a- 1, BP3a-l, BP3a-2, BP3a-3, BP4a-l, BP5a-l, BP5b-l, BP5a-2, BP5a-3, BP5a-4, BP5a-5, BP5b-2, BP5b-3, BP5b-4, BP5b-5, BP5b-6, BP5b-7, BP5b-8, BP5b-9, BP6a-l, BP6a-2, BP6a-3, BP6a-4, BP6a-5, BP6a-6, BP6b-l, BP6b-2, BP6a-7, BP6a-8,
  • a biological sample is subjected to one or more
  • immunoassays using a set of two or more (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10 or more) antibodies in which the set comprises two antibodies having binding profiles, as outlined in Table 33, selected from the following pairs: ⁇ BPla, BPlb ⁇ , ⁇ BPla, BP2a ⁇ , ⁇ BPla, BP3a ⁇ , ⁇ BPla, BP4a ⁇ , ⁇ BPla, BP5a ⁇ , ⁇ BPla, BP5b ⁇ , ⁇ BPla, BP6a ⁇ , ⁇ BPla, BP6b ⁇ , ⁇ BPla, BP7b ⁇ , ⁇ BPla, BP8b ⁇ , ⁇ BPla, BP9b ⁇ , ⁇ BPla, BPlOb ⁇ , ⁇ BPla, BPl la ⁇ , ⁇ BPla, BP12b ⁇ , ⁇ BPla, BP13a ⁇ , ⁇ BPla, BP14a ⁇ , ⁇ BPla, BP15a ⁇ , ⁇ BPla, BP16b ⁇
  • a biological sample is subjected to an immunoassay using a set of one or more antibodies comprising of any three, four , five, six, seven, eight, nine, ten or more of the antibodies of Table 33.
  • a biological sample is subjected to an immunoassay using three or more antibodies having different selected binding profiles.
  • antibodies have selected binding profiles as shown in Table 33.
  • antibodies have selected binding profiles of any one of the antibodies Bla-1, BPlb-1, BP2a- 1, BP3a-l, BP3a-2, BP3a-3, BP4a-l, BP5a-l, BP5b-l, BP5a-2, BP5a-3, BP5a-4, BP5a-5, BP5b-2, BP5b-3, BP5b-4, BP5b-5, BP5b-6, BP5b-7, BP5b-8, BP5b-9, BP6a-l, BP6a-2, BP6a-3, BP6a-4, BP6a-5, BP6a-6, BP6b-l, BP6b-2, BP6a-7, BP6a-8,
  • methods provided herein relate to methods for detecting one or more TGFP family member proteins or forms thereof, one or more portions or domains of TGFP family member proteins and/or one or more chimeras of TGFP family member proteins in a sample obtained from a subject.
  • a subject refers to an individual organism, for example, an individual mammal.
  • the subject is a human.
  • the subject is a non-human mammal.
  • the subject is a non-human primate.
  • the subject is a rodent (e.g. , a mouse or rat).
  • the subject is a sheep, a goat, a cattle, a cat, or a dog. In some embodiments, the subject is a vertebrate, an amphibian, a reptile, a fish, an insect, a fly, or a nematode. In some
  • the subject is a research animal.
  • the subject is genetically engineered, e.g. , a genetically engineered non-human subject.
  • the subject may be of either sex and at any stage of development.
  • the subject is a patient or a healthy volunteer.
  • a method for detecting one or more TGFP family member proteins or forms thereof, one or more portions or domains of TGFP family member proteins and/or one or more chimeras of TGFP family member proteins in a sample obtained from a subject involves (a) contacting the sample with an antibody having a selected binding profile under conditions suitable for binding of the antibody to the antigen, if the antigen is present in the sample, thereby forming binding complexes; and (b) determining the level of the antibody or antigen binding fragment bound to the antigen (e.g. , determining the level of the binding complexes).
  • a binding complex refers to a biomolecular complex of antibody bound to antigen (e.g. , one or more TGFP family member proteins or forms thereof, one or more portions or domains of TGFP family member proteins and/or one or more chimeras of TGFP family member proteins).
  • Binding complexes may comprise antibodies with a single specificity or two or more antibodies or antigen binding fragments with different specificities.
  • a binding complex comprises two or more antibodies recognizing different antigenic sites on the same antigen.
  • an antibody may be bound to an antigen, having bound to it other biomolecules such as RNA, DNA, polysaccharides or proteins.
  • a binding complex comprises two or more antibodies recognizing different antigens.
  • an antibody in a binding complex e.g. , an immobilized antibody bound to antigen
  • an antibody e.g. , a detectably labeled antibody.
  • binding complexes may, in some instances, comprise multiple antigens and multiple antibodies or antigen binding fragments.
  • Antigens present in binding complexes may or may not be in their native in situ conformation.
  • a binding complex is formed between an antibody and a purified protein antigen, or isolated proteins comprising antigen, in which the antigen is not in its native in situ conformation.
  • a binding complex is formed between an antibody and a purified protein antigen, in which the antigen is not in its native in situ conformation and is immobilized on solid support (e.g. , a PVDF membrane).
  • a binding complex is formed with an antibody and, for example, a cell surface protein that is present in situ in a native confirmation (e.g. , on the surface of a cell).
  • binding complexes may or may not be detectably labeled.
  • binding complexes comprise detectably labeled antibodies and non-labeled antibodies.
  • binding complexes comprise detectably labeled antigen.
  • antibodies, in binding complexes are immobilized to one or more solid supports.
  • antigens, in binding complexes are immobilized to one or more solid supports. Exemplary solid supports are disclosed herein and will be apparent to one of ordinary skill in the art. The foregoing examples of binding complexes are not intended to be limiting. Other examples of binding complexes will be apparent to one or ordinary skill in the art.
  • antibody or antigen may be conjugated to a solid support surface, either directly or indirectly.
  • Methods for conjugation to solid supports are standard and can be accomplished via covalent and non- covalent interactions.
  • Non-limiting examples of conjugation methods include: adsorption, cross-linking, protein A/G - antibody interactions, and streptavidin-biotin interactions. Other methods of conjugation will be readily apparent to one of ordinary skill in the art.
  • detection, diagnosis, and monitoring methods include comparing the level of the antibody (including antigen binding fragments) bound to the antigen (e.g. , one or more TGFP family member proteins or forms thereof, one or more portions or domains of TGFP family member proteins and/or one or more chimeras of TGFP family member proteins) to one or more reference standards.
  • the reference standard may be, for example, the level of a corresponding TGFP family member protein in a subject that does or does not have the TGFP family member protein.
  • the reference standard is the level of a TGFP family member protein detected in a sample that does not contain the TGFP family member protein (e.g. , a background level).
  • a background level can be determined from a sample that contains a particular pro/latent-Myostatin, by contacting the sample with non-specific antibodies (e.g. , antibodies obtained from non-immune serum).
  • the reference standard may be the level of a TGFP family member protein detected in a sample that does contain the TGFP family member protein (e.g. , a positive control).
  • the reference standard may be a series of levels associated with varying concentrations of a TGFP family member protein in a sample and useful for quantifying the concentration of the TGFP family member protein in the test sample.
  • the foregoing examples of reference standards are not limiting and other suitable reference standard will be readily apparent to one of ordinary skill in the art.
  • the level of one or more TGFP family member proteins or forms thereof, one or more portions or domains of TGFP family member proteins and/or one or more chimeras of TGFP family member proteins may be measured, as provided herein, from a biological sample using any of the antibodies having a selected binding profile.
  • a biological sample refers to any biological material which may be obtained from a subject or cell.
  • a biological sample may be whole blood, plasma, serum, saliva, cerebrospinal fluid, urine, cells (or cell lysate) or tissue (e.g. , normal tissue or diseased tissue).
  • a biological sample is a fluid sample.
  • a biological sample is a solid tissue sample.
  • a tissue sample may include, without limitation skeletal muscle, cardiac muscle, adipose tissue as well as tissue from other organs.
  • a biological sample is a biopsy sample.
  • a solid tissue sample may be made into a fluid sample using routine methods in the art.
  • a biological sample may also include one or more cells of a cell line.
  • a cell line includes human cells, primate cells (e.g. , vero cells), rat cells (e.g. , GH3 cells, OC23 cells) or mouse cells (e.g. , MC3T3 cells).
  • human cell lines including, without limitation, human embryonic kidney (HEK) cells, HeLa cells, cancer cells from the National Cancer Institute's 60 cancer cell lines (NCI60), DU145 (prostate cancer) cells, Lncap (prostate cancer) cells, MCF-7 (breast cancer) cells, MDA-MB-438 (breast cancer) cells, PC3 (prostate cancer) cells, T47D (breast cancer) cells, THP- 1 (acute myeloid leukemia) cells, U87 (glioblastoma) cells, SHSY5Y human neuroblastoma cells (cloned from a myeloma) and Saos-2 (bone cancer) cells.
  • HEK human embryonic kidney
  • HeLa cells cancer cells from the National Cancer Institute's 60 cancer cell lines (NCI60)
  • DU145 (prostate cancer) cells Lncap (prostate cancer) cells
  • MCF-7 breast cancer
  • MDA-MB-438 breast cancer
  • PC3 prostate cancer
  • a further embodiment relates to a method for monitoring a disease, a condition, or any treatment thereof (e.g., myopathy or myopathy treatment) in a subject having, or at risk of having, the disease or condition comprising: (a) obtaining a biological sample from the subject, (b) determining the level of one or more TGFP family member proteins or forms thereof, one or more portions or domains of TGFP family member proteins and/or one or more chimeras of TGFP family member proteins in the biological sample using an antibody having a selected binding profile and (c) repeating steps (a) and (b) on one or more occasions.
  • a disease, a condition, or any treatment thereof e.g., myopathy or myopathy treatment
  • Biomarkers such as Myostatin may be used as for assessing disease conditions, however, available commercial reagents (e.g., antibodies for ELISAs and Western Blots) are often either not specific for Myostatin, detect only mature Myostatin or do not detect
  • the level of proMyostatin or latent Myostatin may be measured in a subject, or biological sample therefrom, to detect or monitor the progression of a disease or condition.
  • the level of proMyostatin or latent Myostatin may be measured in a subject, or biological sample therefrom, to monitor the response to a treatment for a disease or condition. It should be appreciated that the level of proMyostatin or latent Myostatin may be monitored over any suitable period of time, which may differ depending on the disease or condition, the subject has or any treatment regimen that the subject may be subject to.
  • Another embodiment relates to a diagnostic composition
  • a diagnostic composition comprising any one of the above described antibodies, antigen binding fragments, polynucleotides, vectors or cells and optionally suitable means for detection.
  • the antibodies are, for example, suited for use in immunoassays in which they can be utilized in liquid phase or bound to a solid phase carrier.
  • immunoassays which can utilize the antibody are competitive and noncompetitive immunoassays in either a direct or indirect format. Examples of such
  • immunoassays are the Enzyme Linked Immunoassay (ELISA), radioimmunoassay (RIA), the sandwich (immunometric assay), flow cytometry, the western blot assay,
  • the antigens and antibodies can be bound to many different solid supports (e.g. , carriers, membrane, columns, proteomics array, etc.).
  • solid support materials include glass, polystyrene, polyvinyl chloride, polyvinylidene difluoride, polypropylene, polyethylene, polycarbonate, dextran, nylon, amyloses, natural and modified celluloses, such as nitrocellulose, polyacrylamides, agaroses, and magnetite.
  • the nature of the support can be either fixed or suspended in a solution (e.g. , beads).
  • antibodies may also be used in a method for evaluating expression of one or more TGFP family member proteins or forms thereof in a subject by obtaining a biological sample from the subject which may be a tissue sample, a blood sample or any other appropriate body fluid sample.
  • the procedure may comprise contacting the blood sample (whole blood, serum, plasma), a tissue sample, or protein sample isolated therefrom, with an antibody having a selected binding profile, under conditions enabling the formation of binding complexes between antibody and antigen.
  • the level of such binding complexes may then be determined by any suitable method.
  • the biological sample is contacted with the antibody under conditions suitable for binding of the antibody to one or more TGFP family member proteins or forms thereof, if the antigen is present in the sample, and formation of binding complexes consisting of antibody, bound to the antigen.
  • this contacting step is performed in a reaction chamber, such as a tube, plate well, membrane bath, cell culture dish, microscope slide, and the like.
  • an antibody having a selected binding profile is immobilized on a solid support.
  • the antigen is immobilized on a solid support.
  • the solid support is the surface of a the reaction chamber.
  • the solid support is of a polymeric membrane (e.g. , nitrocellulose strip, Polyvinylidene Difluoride (PVDF) membrane, etc.). Other appropriate solid supports may be used.
  • an antibody having a selected binding profile is
  • immobilized on the solid support prior to contacting with the antigen In other embodiments, immobilization of the antibody is performed after formation of binding complexes. In still other embodiments, antigen is immobilized on a solid support prior to formation of binding complexes.
  • a detection reagent is added to the reaction chamber to detect immobilized binding complexes. In some embodiments, the detection reagent comprises a detectably labeled secondary antibody directed against the antigen. In some embodiments, the primary antibody is itself detectable labeled, and is thereby the detection reagent.
  • detection methods comprise the steps of immobilizing antibodies to a solid support; applying a sample (e.g. , a biological sample or isolated protein sample) to the solid support under conditions that permit binding of antigen to the antibodies, if present in the sample; removing the excess sample from the solid support; applying detectably labeled antibodies under conditions that permit binding of the detectably labeled antibodies to the antigen -bound immobilized antibodies; washing the solid support and assaying for the presence of label on the solid support.
  • the antigen is immobilized on the solid support, such as a PVDF membrane, prior to contacting with the antibody in a reaction chamber (e.g. , a membrane bath).
  • a detection reagent is added to the reaction chamber to detect immobilized binding complexes.
  • the detection reagent comprises a detectably labeled secondary antibody directed against the antigen.
  • the detection reagent comprises a detectably labeled secondary antibody directed against the primary antibody.
  • the detectable label may be, for example, a radioisotope, a fluorophore, a luminescent molecule, an enzyme, a biotin-moiety, an epitope tag, or a dye molecule.
  • the primary antibody is itself detectable labeled, and is thereby the detection reagent. Suitable detectable labels are described herein, and will be readily apparent to one of ordinary skill in the art.
  • diagnostic kits suitable for home or clinical use (point of care service), comprise (a) detectably labeled and/or non-labeled antibodies having a selected binding profile, as antigen binding reagents (e.g. , reagents that bind one or more TGFP family member proteins or forms thereof); (b) a detection reagent; and, optionally, (c) complete instructions for using the reagents to detect antigens in a sample.
  • the diagnostic kit includes the antibody having a selected binding profile, and/or one or more TGFP family member proteins or forms thereof immobilized on a solid support. Any of the solid supports described herein are suitable for incorporation in the diagnostic kits.
  • the solid support is the surface of a reaction chamber of a plate well.
  • the plate well is in a multi-well plate having a number of wells selected from: 6, 12, 24, 96, 384, and 1536, but it is not so limited. In other words,
  • the diagnostic kits provide a detectably labeled antibody. Diagnostic kits are not limited to these embodiments and other variations in kit composition will be readily apparent to one of ordinary skill in the art.
  • Myostatin protein is present in muscle tissue. In some embodiments, Myostatin protein is present in circulation. In some embodiments, Myostatin protein is present in muscle tissue and in circulation. In some embodiments, Myostatin expression is altered in diseases or conditions involving muscle atrophy. Exemplary diseases or conditions involving muscle atrophy include, without limitation, muscular dystrophy, cachexia, sarcopenia, spinal chord injury and atrophy associated with muscle disuse. Thus, Myostatin in tissues or circulating Myostatin, can be a biomarker for muscular atrophic disease.
  • Myostatin as biomarker has been hampered by the paucity of available antibodies to Myostatin, which, because of the high homology between mature Myostatin growth factor and other members of the TGFP family, are not highly selective for Myostatin. Furthermore, many Myostatin antibodies cannot distinguish between the pro-, latent, and mature forms of Myostatin. Thus, detecting the location and expression levels of various Myostatin forms (e.g., proMyostatin, latent Myostatin, primed Myostatin, and mature Myostatin) may provide significant insight into Myostatin biology in normal and disease states.
  • various Myostatin forms e.g., proMyostatin, latent Myostatin, primed Myostatin, and mature Myostatin
  • antibodies to pro-, latent, and mature Myostatin may be useful in biomarker assays for muscle atrophy.
  • antibodies provided herein can be incorporated into assays for MSD, ELISA, RIA, quantitative western blotting (capillary western, Odyssey or similar technology), immunohistochemistry, and any other technology requiring specific binding of antibodies.
  • assays are be applied to analysis of Myostatin levels and isoform distribution in muscle or other tissue, blood and blood derivatives (e.g., serum, plasma, exosome-like vesicles (ELVs) from blood), and urine and derivatives (e.g., urinary ELVs).
  • blood and blood derivatives e.g., serum, plasma, exosome-like vesicles (ELVs) from blood
  • urine and derivatives e.g., urinary ELVs.
  • any of the antibodies provided herein can be used in an immunohistochemistry (IHC) assay.
  • an immunohistochemistry assay is performed on a tissue sample (e.g., a muscle biopsy) to identify a level or localization of one or more TGFP family member proteins or forms thereof (e.g., proMyostatin, latent Myostatin , primed Myostatin, and/or mature Myostatin).
  • a tissue sample e.g., a muscle biopsy
  • TGFP family member proteins or forms thereof e.g., proMyostatin, latent Myostatin , primed Myostatin, and/or mature Myostatin.
  • an IHC assay begins with antigen retrieval, which may vary in terms of reagents and methods.
  • the antigen retrieval process may involve pressure cooking, protease treatment, microwaving, or heating histologic sections in baths of appropriate buffers, with the standard goal of unmasking antigens hidden by formalin crosslinks or other fixation. See, e.g., Leong et al. Appl. Immnunohistochem. 4(3):201 (1996).
  • an IHC assay is a direct assay.
  • an IHC assay is an indirect assay.
  • binding of antibody to the target antigen can be determined directly.
  • a direct assay may use a labeled reagent, such as a fluorescent tag or an enzyme-labeled primary antibody, which can be visualized without further antibody interaction.
  • a labeled reagent such as a fluorescent tag or an enzyme-labeled primary antibody, which can be visualized without further antibody interaction.
  • unconjugated primary antibody binds to the antigen and then a labeled secondary antibody binds to the primary antibody. Where the secondary antibody is conjugated to an enzymatic label, a chromagenic or fluorogenic substrate is added to provide visualization of the antigen.
  • a primary and/or secondary antibody used for IHC is labeled with a detectable moiety.
  • labels include, without limitation, radioisotopes, colloidal gold particles, fluorescent or chemiluminescent labels.
  • a primary and/or secondary antibody used for IHC is labeled with a, radioisotope (e.g. , S, C, I or H) using the techniques described in Current Protocols in Immunology, Volumes 1 and 2, Coligen et al, Ed. Wiley- Interscience, New York, N.Y., Pubs.
  • radioactivity can be measured using scintillation counting.
  • Other radionuclides include, without limitation, 99Tc,90Y, mln, 32P, UC, 150, 13N, 18F, 51Cr, 57To, 225Ra, 60Co, 59Fe, 57Se, 152Eu, 67CU, 217Ci, 211 At, 212Pb, 47Sc, 109Pd, 234Th, and 40K, 157Gd, 55Mn, 52Tr, and 56Fe.
  • a primary and/or secondary antibody used for IHC is labeled with Colloidal gold particles.
  • a primary and/or secondary antibody used for IHC is labeled with fluorescent or chemiluminescent labels including, but not limited to, rare earth chelates (europium chelates), fluorescein and its derivatives, rhodamine and its derivatives, isothiocyanate, phycoerythrin, phycocyanin, allophycocyanin, o-phthaladehyde, fluorescamine, dansyl, umbelliferone, luciferin, luminal label, isoluminal label, an aromatic acridinium ester label, an imidazole label, an acridimium salt label, an oxalate ester label, an aequorin label, 2,3- dihydrophthalazinediones, Texas Red, dansyl, Lissamine, umbelliferone, phycocrytherin, phycocyanin, or commercially available fluorophores such SPECTRUM ORANGE® and SPECTRUM
  • a primary and/or secondary antibody used for IHC is labeled with an enzyme.
  • an enzyme catalyzes a chemical alteration of a chromogenic substrate that can be measured using various techniques. For example, an enzyme may catalyze a color change in a substrate, which can be measured
  • an enzyme may alter the fluorescence or
  • chemiluminescence of the substrate involves techniques for quantifying a change in fluorescence would be apparent to the skilled artisan.
  • a chemiluminescent substrate becomes electronically excited by a chemical reaction and may then emit light which can be measured (e.g., using a chemiluminometer) or donates energy to a fluorescent acceptor.
  • enzymatic labels include luciferases (e.g. firefly luciferase and bacterial luciferase; U.S. Pat. No. 4,737,456), luciferin, 2,3-dihydrophthalazinediones, malate dehydrogenase, urease, peroxidase such as horseradish peroxidase (HRPO), alkaline phosphatase, ⁇ -galactosidase, glucoamylase, lysozyme, saccharide oxidases (e.g., glucose oxidase, galactose oxidase, and glucose-6-phosphate dehydrogenase), heterocyclic oxidases (such as uricase and xanthine oxidase), lactoperoxidase, microperoxidase, and the like.
  • luciferases e.g. firefly luciferase and bacterial luciferase
  • a label is indirectly conjugated to any of the antibodies with a selected binding profile provided herein.
  • an antibody can be conjugated with biotin and any suitable label can be conjugated with avidin, or vice versa. Biotin binds selectively to avidin and thus, the label can be conjugated with the antibody in this indirect manner.
  • a tissue section e.g., an FFPE section or a frozen section
  • a tissue section is exposed to one or more of the antibodies provided herein as primary antibodies for a sufficient period of time and under suitable conditions to allow one or more primary antibody to bind to one or more TGFP family member proteins or forms thereof of the tissue section.
  • suitable conditions for achieving suitable binding can be determined by routine experimentation,
  • a slide containing a biological sample is subjected to a wash step to remove unbound and excess amounts of primary antibody.
  • primary antibody is linked to a detectable label, such as paramagnetic ions, radioactive isotopes, fluorochromes, and NM -detectable substances, and the slide is evaluated for staining of one or more TGFP family member proteins or forms thereof using an appropriate imaging apparatus.
  • a detectable label such as paramagnetic ions, radioactive isotopes, fluorochromes, and NM -detectable substances
  • tissue sample used to prepare stained tissue sections for evaluating expression of one or more TGFP family member proteins or forms thereof can be collected from a subject before and/or after exposure of the subject to one or more therapeutic agents, e.g., an anti-latent Myostatin antibody. Accordingly, tissue samples may be collected from a subject over a period of time.
  • a tissues sample e.g., a muscle tissue sample
  • a tissue sample is obtained by any suitable procedure, which includes, but not limited to, surgical excision, aspiration or biopsy.
  • a tissue sample is sectioned and examined for one or more TGFP family member proteins or forms thereof as a fresh specimen. In other embodiments, a tissue sample is frozen for further sectioning.
  • a tissue sample is preserved by fixing and embedding in paraffin (FFPE) or the like.
  • FFPE paraffin
  • a tissue sample may be fixed by conventional methodology, with the length of fixation depending on the size of the tissue sample and the fixative used.
  • exemplary fixatives include, but are not limited to neutral buffered formalin, glutaraldehyde, Bouin's or paraformaldehyde.
  • a tissue sample is fixed with formalin.
  • a fixed tissue sample is also embedded in paraffin to prepare a formalin-fixed and paraffin- embedded (FFPE) tissue sample. Examples of paraffin include, but are not limited to, Paraplast, Broloid and Tissuemay.
  • a tissue sample is fixed and dehydrated through an ascending series of alcohols, infiltrated and embedded with paraffin or other sectioning media so that the tissue sample may be sectioned.
  • a tissue sample is first sectioned and then the individual sections are fixed.
  • Growth factors are cell signaling molecules that stimulate a variety of cellular activities. Due to their broad-reaching influence within biological systems, growth factor signaling is tightly regulated, often through interactions with other biomolecules, the extracellular and/or cellular matrix or within a particular cell environment or niche. These interactions may be direct or indirect.
  • TGF- ⁇ transforming growth factor beta
  • Activated type I receptors may in turn phosphorylate receptor-associated SMADs (R-SMADs) promoting co-SMAD (e.g. SMAD4) dimer/trimer formation and nuclear translocation.
  • R-SMADs receptor-associated SMADs
  • co-SMAD e.g. SMAD4 dimer/trimer formation and nuclear translocation.
  • SMAD complexes collaborate with cofactors to modulate expression of TGF- ⁇ family member target genes.
  • TGF- ⁇ family member signaling cascades are involved in a number of diverse biological pathways including, but not limited to inhibition of cell growth, tissue
  • TGF- ⁇ signaling related to growth inhibition and tissue homeostasis may affect epithelial, endothelial, hematopoietic and immune cells through the activation of p21 and pi to mediate cell cycle arrest and repress myc.
  • ECM remodeling TGF- ⁇ signaling may increase fibroblast populations and ECM deposition (e.g. collagen).
  • TGF- ⁇ signaling related to cell migration and invasion may affect epithelial and/or endothelial cells, inducing stem cell-like phenotypes. This aspect of signaling may play a role in smooth muscle cell proliferation following vascular surgery and/or stenting.
  • TGF- ⁇ ligand is necessary for T regulatory cell function and maintenance of immune precursor cell growth and homeostasis.
  • Nearly all immune cells comprise receptors for TGF- ⁇ and TGF- ⁇ knockout mice die postnataly due in part to inflammatory pathologies.
  • TGF- ⁇ suppresses interferon gamma-induced activation of natural killer cells (Wi, J. et al., 2011. Hepatology. 53(4): 1342-51, the contents of which are herein incorporated by reference in their entirety).
  • phosphorylate type I receptors which in turn phosphorylate receptor-regulated Smads that translocate to and accumulate in the nucleus and regulate transcription.
  • BMP bone morphogenetic proteins
  • GDF growth and differentiation factor
  • myostatin nodal, anti-Mullerian hormone, and lefty proteins.
  • mature growth factors are synthesized along with their prodomains as single polypeptide chains (see FIG. 2).
  • polypeptide chains may comprise cleavage sites for separation of prodomains from mature growth factors.
  • cleavage sites are furin cleavage sites recognized and cleaved by proprotein convertases.
  • prodomain homology In general, homology among TGF- ⁇ family member growth factor domains is relatively high. Interestingly, prodomain homology is much lower. This lack of homology may be an important factor in altered growth factor regulation among family members. In some cases, prodomains may guide proper folding and/or dimerization of growth factor domains. Prodomains have very recently been recognized, in some cases, to have important functions in directing growth factors (after secretion) to specific locations in the extracellular matrix (ECM) and/or cellular matrix, until other signals are received that cause growth factor release from latency. Release from latency may occur in highly localized environments whereby growth factors may act over short distances (e.g.
  • growth factor-prodomain complexes are secreted as homodimers.
  • prodomain-growth factor complexes may be secreted as heterodimers.
  • TGF-P-related protein refers to a TGF- ⁇ isoform, a TGF- ⁇ family member or a TGF- ⁇ family member-related protein.
  • TGF- ⁇ family members may include, but are not limited to any of those shown in in FIG. 1 and/or listed in Table 1. These include, but are not limited to TGF- ⁇ proteins, BMPs, myostatin, GDFs and inhibins.
  • Aspects of the present disclosure provide tools and/or methods for characterizing and/or modulating cellular activities related to growth factor signaling.
  • tools of the present disclosure may comprise antigens comprising one or more components of one or more TGF ⁇ -related proteins.
  • tools of the present disclosure may comprise antibodies directed toward antigens of the present disclosure.
  • tools of the present disclosure may comprise assays for the detection and/or characterization of TGF ⁇ -related proteins, the detection and/or characterization of antibodies directed toward TGF ⁇ -related proteins and/or the detection and/or characterization of cellular activities and/or their cellular signaling related to TGF ⁇ -related proteins.
  • GDF growth differentiation factor
  • TGF- ⁇ family member proteins involved in a number of cellular and developmental activities.
  • GDF-modulatory antibodies are capable of distinguishing between various growth factor complexes allowing for growth factor activity modulation that occurs only at sites of specific complex formations.
  • TGF ⁇ -related proteins are involved in a number of cellular processes. In embryogenesis, the 33 members of the TGF- ⁇ family of proteins are involved in regulating major developmental processes and the details of the formation of many organs. Much of this regulation occurs before birth; however, the family continues to regulate many processes after birth, including, but not limited to immune responses, wound healing, bone growth, endocrine functions and muscle mass. TGF-P-related proteins are listed and described in International Patent Application No. WO2014074532, the contents of which are herein incorporated by reference in their entirety.
  • TGF- ⁇ family pro-proteins i.e. the protein after removal of the secretion signal sequence
  • the pro-protein contains, and is the precursor of, the prodomain and the growth factor. Shown in the Table are the names of the originating TGF- ⁇ family member and the pro-protein sequence. Also identified in "bold" and
  • proprotein convertase cleavage sites underlined are proprotein convertase cleavage sites. Upon cleavage, the resulting prodomain retains this site, whereas the mature growth factor begins following the cleavage site. It is noted that Leftyl and Lefty2 are not cleaved by proprotein convertases just prior to the start of the mature growth factor.
  • TGF- 2 SLSTCSTLDMDQFMRKRIEAIRGQILSKLKLTSPPEDYPEPEEV 2
  • GDF-8 NENSEQKENVEKEGLCNACTWRQNTKSSRIEAIKIQILSKLRL 5 (myostatin) ETAPNISKDVIRQLLPKAPPLRELIDQYDVQRDDSSDGSLEDD
  • GDF-7 RDGLEAAAVLRAAGAGPVRSPGGGGGGGGRTLAQAAGA 19
  • proprotein convertase refers to an enzyme that cleaves a prodomain from a translated protein to facilitate protein maturation.
  • Some proprotein convertases of the present disclosure include the subtilisin-like proprotein convertase (SPC) family member enzymes.
  • SPC family comprises calcium-dependent serine endopro teases that include, but are not limited to furin/PACE, PC 1/3, PC2, PC4, PC5/6, PACE4 and PC7 (Fuller et al, 2009. Invest Ophthalmol Vis Sci. 50(12):5759-68, the contents of which are herein incorporated by reference in their entirety).
  • GDF-11 may in some cases, be cleaved by PC5/6.
  • proprotein convertases may cleave proproteins at additional sites, other than those indicated in Table 1.
  • pro-proteins may be cleaved at a first cleavage site (the first site being the site closest to the N-terminus).
  • pro-proteins may be cleaved at a cleavage site other than a first cleavage site.
  • proprotein convertase cleavage may occur intracellularly.
  • proprotein convertase cleavage may occur extracellularly.
  • TGF- ⁇ family member proteins are synthesized in conjunction with prodomains. Some prodomains may remain associated with growth factors after cleavage. Such associations may form latent growth factor-prodomain complexes (GPCs) that modulate the availability of growth factors for cell signaling. Growth factors may be released from latency in GPCs through associations with one or more extracellular proteins. In some cases, growth factor release may rely on force applied to GPCs through extracellular protein interactions. Such forces may pull from C-terminal and/or N-terminal regions of GPCs resulting in the release of associated growth factors.
  • GPCs latent growth factor-prodomain complexes
  • the prodomain portion of the GPC is responsible for growth factor retention and blocking the interaction of retained growth factors with their receptors.
  • Such GPCs where the bound growth factor is unable to promote signaling activity, are also referred to herein as "latent complexes.”
  • Prodomain portions of GPCs that function to block growth factor signaling activity are referred to as latency associated peptides (LAPs).
  • LAPs latency associated peptides
  • TGF- ⁇ , 2 and 3 are know to comprise LAPs.
  • GDF prodomains also function to block growth factor activity.
  • Some prodomains may comprise LAP-like domains.
  • LAP-like domain refers to prodomain portions of GPCs and/or free prodomains that may be structurally similar or synthesized in a similar manner to LAPs, but that may not function to prevent growth factor/receptor interactions.
  • FIG. 3 is a schematic depicting an embodiment of a GPC before and after growth factor release.
  • growth factor dimers may associate with prodomain modules to form a GPC.
  • GPCs comprise protein modules necessary for different aspects of growth factor signaling, secretion, latency and/or release from latent GPCs.
  • protein module refers to any component, region and/or feature of a protein. Protein modules may vary in length, comprising one or more amino acids.
  • Protein modules may be from about 2 amino acid residues in length to about 50 amino acid residues in length, from about 5 amino acid residues in length to about 75 amino acid residues in length, from about 10 amino acid residues in length to about 100 amino acid residues in length, from about 25 amino acid residues in length to about 150 amino acid residues in length, from about 125 amino acid residues in length to about 250 amino acid residues in length, from about 175 amino acid residues in length to about 400 amino acid residues in length, from about 200 amino acid residues in length to about 500 amino acid residues in length and/or at least 500 amino acid residues in length.
  • protein modules comprise one or more regions with known functional features (e.g. protein binding domain, nucleic acid binding domain, hydrophobic pocket, etc). Protein modules may comprise functional protein domains necessary for different aspects of growth factor signaling, secretion, latency and/or release from latent conformations.
  • protein modules may be derived from TGF-P-related proteins.
  • Such protein modules may include, but are not limited to prodomains (e.g. latency- associated peptides (LAPs) or LAP-like domains), growth factor domains, fastener regions, proprotein convertase cleavage sites (e.g. furin cleavage sites), B/TP cleavage sites, arm regions, finger regions, residues (such as cysteine residues for example) for extracellular protein associations, latency loops (also referred to herein as latency lassos), alpha 1 helical regions, alpha 2 helical regions, RGD sequences and bowtie regions.
  • FIG. 4 is a schematic diagram showing some of the protein modules present in a GPC as well as in the prodomains and growth factor dimer after activating cleavage of the GPC.
  • prodomains may associate with growth factors in GPCs. Some prodomains may sterically prevent growth factor association with one or more cellular receptors. Prodomains may comprise arm regions and/or straight jacket regions. Some prodomains may comprise C-terminal regions referred to herein as "bowtie regions.” In some prodomain dimers, bowtie regions of each monomer may associate and/or interact. Such associations may comprise disulfide bond formation, as is found between monomers of TGF- ⁇ isoform LAPs.
  • arm regions may comprise trigger loop regions.
  • Trigger loops may comprise regions that associate with integrins. Such regions may comprise amino acid sequences comprising RGD (Arg-Gly-Asp). Regions comprising RGD sequences are referred to herein as RGD sequence regions.
  • prodomains comprise latency loops (also referred to herein as latency lassos). Some latency loops may maintain associations between prodomains and growth factors present within GPCs.
  • Prodomains may also comprise fastener regions. Such fastener regions may promote associations between prodomains and growth factors present within GPCs by maintaining prodomain
  • GPCs may require enzymatic cleavage to promote dissociation of bound growth factors and growth factor activity. Such enzymatic cleavage events are referred to herein as "activating cleavage" events. Activating cleavage of GPCs may be carried out in some instances by members of the BMP- 1/Tolloid- like proteinase (B/TP) family (Muir et al., 2011. J Biol Chem. 286(49):41905-11, the contents of which are herein incorporated by reference in their entirety).
  • B/TP BMP- 1/Tolloid- like proteinase
  • metaloproteinases may include, but are not limited to BMP- 1, mammalian tolloid protein (mTLD), mammalian tolloid-like 1 (mTLLl) and mammalian tolloid-like 2 (mTLL2).
  • Exemplary GPCs that may be subjected to activating cleavage by such metalloproteinases may include, but are not limited to GDF-8 and GDF-11.
  • GDF-8 may be cleaved by mTLL2.
  • activating cleavages may occur intracellularly. In some cases, activating cleavages may occur extracellularly.
  • Growth factor release from GPCs may require cleavage by a proprotein convertase enzyme followed by an activating cleavage [e.g. by one or more members of the BMP- 1/Tolloid-like proteinase (B/TP) family.]
  • GDF-8 and GDF-11 GPCs may be transformed by furin cleavage into a latent complex that further requires cleavage by
  • the present disclosure provides polypeptide inhibitors (e.g., antibodies) that inhibit one or more members of the B/TP family. Such inhibitors may block cleavage of BMP-l/Tolloid cleavage sites, including, but not limited to BMP/Tolloid cleavage sites on one or more latent complexes (e.g., GDF-8 latent complexes and/or GDF-11 latent complexes).
  • polypeptide inhibitors e.g., antibodies
  • Such inhibitors may block cleavage of BMP-l/Tolloid cleavage sites, including, but not limited to BMP/Tolloid cleavage sites on one or more latent complexes (e.g., GDF-8 latent complexes and/or GDF-11 latent complexes).
  • activating cleavage may not lead to dissociation of bound growth factor, but instead may promote an active conformation of the GPC.
  • active conformation refers to a GPC protein confirmation that allows the growth factor to engage in receptor interaction.
  • proBMP-7 and proBMP-9 See FIG. 1 .
  • Active conformations of GDF-8 GPCs or GDF- 11 GPCs are referred to herein as "primed" complexes, and can be produced by the sequential cleavage of GPCs at the furin cleavage site and the BMP/Tolloid cleavage site.
  • Primed complexes (either the entire complex or portions of the complex) may bind receptors resulting in receptor signaling.
  • prodomains may be dissociated from growth factors upon receptor binding and/or signaling activity.
  • prodomains may remain associated with growth factors upon receptor binding and/or signaling activity.
  • prodomains may become partially dissociated from growth factors during receptor binding and/or signaling activity.
  • primed complexes may bind preferentially to one or more receptors over one or more other receptors.
  • receptor activity resulting from primed complex interactions may be quenched or competed for by excess prodomain or fragments thereof.
  • the present disclosure provides polypeptide inhibitors (e.g., inhibiting antibodies) that block the formation of primed complexes from latent complexes.
  • such inhibitors bind BMP/Tolloid cleavage sites on latent GPCs (e.g., latent GDF-8 or latent GDF-11).
  • such inhibitors prevent cleavage of the BMP/Tolloid cleavage site.
  • Straight jacket regions may comprise alpha 1 helical regions.
  • alpha 1 helical regions may be positioned between growth factor monomers. Some alpha 1 helical regions comprise N-terminal regions of prodomains. Alpha 1 helical regions may also comprise N-terminal regions for extracellular associations. Such
  • extracellular associations may comprise extracellular matrix proteins and/or proteins associated with the extracellular matrix. Some extracellular associations may comprise associations with proteins that may include, but are not limited to LTBPs (e.g. LTBP1, LTBP2, LTBP3 and/or LTBP4), fibrillins (e.g. fibrillin- 1, fibrillin-2, fibrillin-3 and/or fibrillin-4), perlecan, decorin and/or GASPs.
  • LTBPs e.g. LTBP1, LTBP2, LTBP3 and/or LTBP4
  • fibrillins e.g. fibrillin- 1, fibrillin-2, fibrillin-3 and/or fibrillin-4
  • perlecan e.g., decorin and/or GASPs.
  • N-terminal extracellular associations may comprise disulfide bonds between cysteine residues.
  • extracellular matrix proteins and/or proteins associated with the extraceullar matrix may comprise bonds or interactions with one or more regions of prodomains other than N-termin
  • growth factor domains comprise one or more growth factor monomers. Some growth factor domains comprise growth factor dimers. Such growth factor domains may comprise growth factor homodimers or heterodimers (comprising growth factor monomers from different TGF-P-related proteins). Some growth factor domains may comprise fingers regions. Such fingers regions may comprise ⁇ -pleated sheets. Fingers regions may associate with prodomains. Some fingers regions may maintain association between growth factor domains and prodomains.
  • recombinant proteins of the present disclosure may comprise protein modules from growth differentiation factor (GDF) proteins.
  • GDF protein modules may comprise the protein modules and/or amino acid sequences listed in Table 2.
  • protein modules of the present disclosure may comprise amino acid sequences similar to those in Table 2, but comprise additional or fewer amino acids than those listed.
  • Some such amino acid sequences may comprise about 1 more or fewer amino acids, about 2 more or fewer amino acids, about 3 more or fewer amino acids, about 4 more or fewer amino acids, about 5 more or fewer amino acids, about 6 more or fewer amino acids, about 7 more or fewer amino acids, about 8 more or fewer amino acids, about 9 more or fewer amino acids, about 10 more or fewer amino acids or greater than 10 more or fewer amino acids on N-terminal and/or C-terminal ends.
  • NPRGSAGPCCTPTKMSPINMLYFNGKEQIIYGK IPAMVVDRCGCS GDF-11 growth factor NLGLDCDEHSSESRCCRYPLTVDFEAFGWDWI 43 domain IAPKRYKANYCSGQCEYMFMQKYPHTHLVQQ
  • GDF-8 fingers region 1 CRYPLTVDFEAFGWDWIIAPKRYKANYCS 47 and GDF- 11
  • GDF-15 is a TGF- ⁇ family protein that is highly expressed in liver. Expression of GDF-15 is dramatically upregulated following liver injury (Hsiao et al. 2000. Mol Cell Biol. 20(10):3742-51).
  • GDF-15 comprises less than 30% homology with other members, making it the most divergent member of the family (Tanno et al., 2010. Curr Opin Hematol. 17(3): 184-90, the contents of which are incorporated herein by reference in their entirety).
  • the mature form is soluble and can be found in the blood stream.
  • GDF-15 levels in circulation have been found to negatively correlate with hepcidin levels, suggesting a role for GDF-15 in iron load and/or metabolism (Finkenstedt et al., 2008. British Journal of Haematology. 144:789-93). Elevated GDF-15 in the blood is also associated with ineffective and/or apoptotic erythropoiesis, such as in subjects suffering from beta-thalassemia or dyserythropoietic anemias.
  • recombinant proteins of the present disclosure may comprise protein modules from activin subunits.
  • Such protein modules may comprise the protein modules and/or amino acid sequences of the activin subunit inhibin beta A, listed in Table 3.
  • protein modules of the present disclosure may comprise amino acid sequences similar to those in Table 3, but comprise additional or fewer amino acids than those listed.
  • Some such amino acid sequences may comprise about 1 more or fewer amino acids, about 2 more or fewer amino acids, about 3 more or fewer amino acids, about 4 more or fewer amino acids, about 5 more or fewer amino acids, about 6 more or fewer amino acids, about 7 more or fewer amino acids, about 8 more or fewer amino acids, about 9 more or fewer amino acids, about 10 more or fewer amino acids or greater than 10 more or fewer amino acids on N-terminal and/or C-terminal ends.
  • Prodomains may vary in length from about 50 to about 200, from about 100 to about 400 or from about 300 to about 500 amino acids residues. In some embodiments, prodomains range from about 169 to about 433 residues. Prodomains may be unrelated in sequence and/or low in homology. Some prodomains may have similar folds and/or three dimensional structures. Prodomains of TGF- ⁇ family members may comprise latency loops. Such loops may be proline-rich. Latency loop length may determine the ability of such loops to encircle growth factor finger regions.
  • protein modules from some TGF- ⁇ family members comprise low sequence identity with protein modules from other TGF- ⁇ family members. Such low sequence identity may indicate specialized roles for such family members with distinct protein modules.
  • GPCs Association of GPCs with extracellular proteins may strengthen prodomain-growth factor interactions.
  • extracellular proteins may include, but are not limited to LTBPs, fibrillins, GASPs, perlecan and/or decorin.
  • extracellular protein associations are required to keep growth factors latent in GPCs.
  • GARP expression has been shown to be required for surface expression of GPCs on the surface of cells of hematopoietic origin (Tran, D.Q. et al., GARP (LRRC32) is essential for the surface expression of latent TGF- ⁇ on platelets and activated FOXP3+ regulatory T cells. PNAS. 2009, Jun 2. 106(32): 13445-50). GARP may act as a tether to hold GPCs in place on the surface of these cells, including, but not limited to regulatory T-cells and/or platelets.
  • recombinant proteins of the present disclosure may comprise bone morphogenetic proteins (BMPs), a family of TGF-P-related proteins.
  • BMPs bone morphogenetic proteins
  • Protein modules comprising sequences from BMPs may comprise sequences from any of those BMP modules disclosed in FIG. 6. While related to other TGF- ⁇ family member proteins, BMPs generally signal through SMAD1, 5 and 8 proteins while TGF- ⁇ isoforms (e.g. TGF- ⁇ , TGF-P2 and TGF-P3) signal through SMAD2 and SMAD3.
  • Some BMP receptors and/or co-receptors are also distinct from other TGF- ⁇ family member proteins.
  • RGM repulsive guidance molecule
  • RGM proteins act as co-receptors for BMP signaling.
  • RGMA, RGMB and RGMC also known as hemojuvelin (Hjv)].
  • Recombinant proteins of the present disclosure comprising one or more BMP protein module may be useful for the development of antibodies and/or assays to study, enhance and/or perturb BMP interactions with RGM proteins.
  • CTCK domain-containing proteins Another family of GDF/BMP interacting proteins is C-terminal cysteine knot-like (CTCK) domain-containing proteins.
  • CTCK domain-containing proteins may act antagonistically with regard to GDF/BMP signal transduction.
  • CTCK domain-containing proteins include, but are not limited to Cerberus, Connective tissue growth factor (CTGF), DAN domain family member 5 (DAND5), Gremlin- 1 (GREM1), Gremlin-2 (GREM2), Mucin-19 (MUC19), Mucin-2 (MUC2), Mucin-5AC (MUC5AC), Mucin-5B (MUC5B), Mucin-6 (MUC6), Neuroblastoma suppressor of tumorigenicity 1 (NBL1), Norrin (NDP), Otogelin (OTOG), Otogelin-like protein (OTOGL), Protein CYR61 (CYR61), Protein NOV homolog (NOV), Sclerostin (SOST), Sclerostin domain-containing protein 1 (SOSTDC
  • the present disclosure provides recombinant proteins.
  • the term "recombinant protein” refers to a protein produced by an artificial gene and/or process (e.g. genetic engineering). Such recombinant proteins may comprise one or more protein modules from one or more TGF-P-related proteins. Some recombinant proteins disclosed herein may be useful as recombinant antigens.
  • the term "recombinant antigen” refers to a recombinant protein that may be used to immunize one or more hosts for the production of antibodies directed toward one or more epitopes present on such recombinant antigens. Some recombinant antigens may be cell-based antigens.
  • cell-based antigen refers to recombinant antigens that are expressed in cells for presentation of such antigens on the cell surface. Such cells may be used to immunize hosts for the production of antibodies directed to such cell-based antigens.
  • recombinant proteins disclosed herein may be used as therapeutics.
  • Recombinant proteins disclosed herein may modulate growth factor (e.g. growth factors comprising TGF-P-related proteins) levels and/or activity (e.g. signaling) upon administration and/or introduction to one or more subjects and/or niches.
  • growth factor e.g. growth factors comprising TGF-P-related proteins
  • activity e.g. signaling
  • recombinant proteins disclosed herein may be used to assay growth factor (e.g. growth factors comprising TGF-P-related proteins) levels and/or activity (e.g. signaling). Some recombinant proteins disclosed herein may be used in the isolation of antibodies directed to TGF-P-related proteins. Recombinant proteins of the present disclosure may also be used as recombinant antigens in the development of stabilizing [reducing or preventing dissociation between two agents, (e.g. growth-factor release from GPCs, GPC release from one or more protein interactions)] and/or releasing [enhancing the dissociation between two agents (e.g. growth-factor release from GPCs, GPC release from one or more protein interactions)] antibodies.
  • stabilizing reducing or preventing dissociation between two agents, (e.g. growth-factor release from GPCs, GPC release from one or more protein interactions)] and/or releasing [enhancing the dissociation between two agents (e.g. growth-factor release from GPCs, GPC release from
  • Recombinant proteins of the present disclosure may include TGF- ⁇ family member proteins as well as components and/or protein modules thereof. Some recombinant proteins of the present disclosure may comprise prodomains without associated growth factors, furin cleavage-deficient mutants, mutants deficient in extracellular protein associations and/or combinations thereof.
  • recombinant proteins may comprise detectable labels.
  • Detectable labels may be used to allow for detection and/or isolation of recombinant proteins.
  • Some detectable labels may comprise biotin labels, polyhistidine tags and/or flag tags. Such tags may be used to isolate tagged proteins.
  • Proteins produced may comprise additional amino acids encoding one or more 3C protease cleavage site. Such sites allow for cleavage at the 3C protease cleavage site upon treatment with 3C protease, including, but not limited to rhinovirus 3C protease. Such cleavage sites are introduced to allow for removal of detectable labels from recombinant proteins.
  • FIG. 7 is a schematic depicting an embodiment of a recombinant GPC.
  • Recombinant proteins according to FIG. 7 comprising TGF-P-family member proteins may comprise features including, but not limited to C-terminal regions of the mature growth factor, N-terminal regions of the prodomain and/or proprotein cleavage sites.
  • the proprotein cleavage site of recombinant GPCs may, for example, comprise the furin consensus sequences RXXR, or RXR wherein R is arginine and X indicates amino acid residues that may vary among TGF- ⁇ family members.
  • Furin cleavage site sequences (although not limited to cleavage by furin alone and may include cleavage by other proprotein convertase enzymes) for each TGF- ⁇ family member are indicated in Table 1.
  • prodomains of GPCs may comprise one or more cysteine residues within and/or near the N-terminal region of the prodomain.
  • cysteine residues may be from about 1 to about 10 amino acids, from about 4 to about 15 amino acids, from about 5 to about 20 amino acids and/or from about 7 to about 50 amino acids from the N-terminus of the prodomain.
  • recombinant GPCs may also be expressed with detectable labels. Such detectable labels may be useful for detection and/or isolation of recombinant GPCs. Detectable labels may comprise 2 or more histidine (His) residues. Such detectable labels may also be referred to herein as polyhistidine tags.
  • Polyhistidine tags may include hexa histidine tags or HIS-TAGTM (EMD Biosciences, Darmstadt, Germany) comprising a chain of six histidine residues. Some polyhistidine tags may be present at the N-terminus of recombinant proteins disclosed herein.
  • polyhistidine tags may be present at the C-terminus of recombinant proteins disclosed herein. Proteins produced may comprise additional amino acids encoding one or more 3C protease cleavage site. Such sites allow for cleavage at the 3C protease cleavage site upon treatment with 3C protease, including, but not limited to rhinovirus 3C protease. Some cleavage sites may be introduced to allow for removal of detectable labels from recombinant proteins.
  • recombinant GPCs may comprise mutations in one or more amino acids as compared to wild type sequences.
  • one or more regions of proteolytic processing may be mutated (see FIG. 8).
  • Such regions may comprise proprotein convertase cleavage sites.
  • Proprotein convertase e.g. furin
  • cleavage site mutations prevent enzymatic cleavage at that site and/or prevent enzymatic cleavage of growth factors from their prodomains.
  • Some proprotein convertase cleavage sites comprising RXXR sequences may be mutated to RXG (wherein X indicates a site where amino acid residues may be variable).
  • Such mutations are herein abbreviated as "D2G” mutations and may be resistant to enzymatic cleavage.
  • furin cleavage sites comprising RXXR sequences are mutated to AXXA.
  • AXXA sequences may also be resistant to enzymatic cleavage.
  • regions of proteolytic processing by tolloid and/or tolloid- like proteins may be mutated to prevent such proteolytic processing.
  • tolloid processing regions on GDF-8 and/or GDF-11 may be mutated.
  • mutation of aspartic acid residues to alanine residues within tolloid processing regions prevents tolloid processing. Mutation of aspartic acid residue 76 (D76) of the GDF-8 (myostatin) proprotein has been shown to prevent proteolytic activation of latent GDF-8 (Wolfman, N.M. et al.,PNAS. 2003, Oct 6. 100(26): 15842-6).
  • Asp 120 (D120, residue number counted from the translated protein, D98 from the proprotein of SEQ ID NO: 4) in GDF-11 may be mutated to prevent tolloid processing (Ge et al., 2005. Mol Cell Biol. 25(14):5846-58, the contents of which are herein incorporated by reference in their entirety).
  • one or more amino acids may be mutated in order to form recombinant GPCs with reduced latency. Such mutations are referred to herein as "activating mutations.” These mutations may introduce one or more regions of steric clash between complex prodomains and growth factor domains.
  • steric clash when referring to the interaction between two proteins or between two domains and/or epitopes within the same protein, refers to a repulsive interaction between such proteins, domains and/or epitopes due to overlapping position in three-dimensional space.
  • Steric clash within GPCs may reduce the affinity between prodomains and growth factor domains, resulting in elevated ratios of free growth factor to latent growth factor.
  • one or more amino acids may be mutated in order to form recombinant GPCs with increased latency. Such mutations are referred to herein as "stabilizing mutations.” These mutations may increase the affinity between prodomains and growth factor domains, resulting in decreased ratios of free growth factor to latent growth factor.
  • GDFs growth differentiation factors
  • GDFs Growth differentiation factors
  • activins and inhibins are TGF- ⁇ family member proteins involved in a number of cellular and/or developmental activities.
  • recombinant proteins may comprise one or more protein modules from one or more GDFs, activins and/or inhibins.
  • GDF protein modules may comprise GDF-8 and/or GDF-11 protein modules.
  • GDF-8 and GDF-11 which are secreted as latent complexes (Sengle et al., 2011. J Biol Chem. 286(7):5087-99; Ge et al., 2005. Mol Cel Biol. 25(14):5846-58), show
  • GDF-8 (also referred to herein as myostatin) is involved in regulating muscle mass, and its deficiency increases muscle mass in multiple species, including humans (Rodino-Klapac, L.R. et al., 2009. Muscle Nerve. 39(3):283-96). GDF-8 may be found in the circulation in latent form, but may also be stored in the extracellular matrix, bound to LTBP3 (Anderson et al., 2007. J Biol Chem. 283(11):7027-35) or perlecan (Sengle et al., 2011. J Biol Chem. 286(7):5087-99).
  • GDF-8 While complexed with its prodomain, GDF-8 is unable to participate in receptor binding with the type II receptor, ActRIIB (Sengle et al., 2008. J Mol Biol. 381(4): 1025-39). While GDF-8 is expressed primarily in muscle, GDF-11 expression is more systemic and its activity is thought to be involved in multiple processes (Lee et al., 2013. PNAS. 110(39):E3713-22). It is believed to be involved in development of multiple tissues, including, but not limited to the retina, kidney, pancreas and olfactory system. It is also believed to be a circulating factor in the blood. Recent studies indicate that GDF-11 may rejuvenate skeletal muscle, improve cerebral circulation and promote neurogenesis (Sinha, M. et al., 2014. Science.
  • antibodies of the disclosure may promote skeletal muscle rejuvenation, improve cerebral circulation and promote neurogenesis by promoting the release of GDF- 11 growth factor from latent complexes.
  • GDF-11 has also been identified as a regulator of erythropoiesis.
  • antibodies of the invation that inhibit GDF-11 may restore the ability of erythroid progenitors to differentiate and hence alleviate anemia (Paulson, RF. 2014. Nature Medicine. 20:334-5, the contents of which are herein incorporated by reference in their entirety).
  • GDF-8 and GDF-11 also share considerable homology. While the prodomains only share 48% homology, GDF-8 and GDF-11 growth factor domains share 90% homology (60% homology when prodomains and growth factors are taken together). Given the high degree of sequence similarity, it is not surprising that GDF- 11 and 8 growth factors bind and signal through the same receptors consisting of a Type I receptor (ALK4/5) in association with a type II receptor (ACTRIIA/B). The high degree of conservation in the mature growth factors has made it challenging to identify reagents and monoclonal antibodies that can differentiate between mature GDF-11 and 8. Consequently, there are no therapies in clinical trials today that are specific for GDF-11.
  • ALK4/5 Type I receptor
  • ACTRIIA/B type II receptor
  • proGDF proteins require furin cleavage in order to form latent complexes (see FIG. 9).
  • furin cleavage is carried out by cells in which proGDF proteins are expressed.
  • furin cleavage may be carried out through furin addition to cultures or solutions comprising proGDF proteins.
  • Furin cleavage of recombinant proteins of the disclosure may in some cases occur intracellularly. In some cases furin cleavage of recombinant proteins of the disclosure may occur extracellularly. Release of GDF-8 and GDF-11 from latent GPCs requires activating cleavage of latent complexes.
  • An activating cleavage refers to a proteolytic cleavage of a latent complex that leads to growth factor release and/or activation.
  • Activating cleavage may include prodomain cleavage at a BMP/tolloid/tolloid-like proteinase cleavage site.
  • sites may include the site located between Arg 75 and Asp 76 in GDF-8 and between Gly 97 and Asp 98 in GDF-11. This cleavage is between the 2 helix and the fastener.
  • recombinant proteins of the present disclosure comprising GDFs may comprise sequences listed in Table 4 or fragments thereof. In some cases, these sequences are expressed in association with N- and/or C-terminal secretion signal sequences [e.g. human Ig kappa chains with amino acid sequence MDMRVPAQLLGLLLLWFSGVLG (SEQ ID NO: 66)], flag tag sequences [e.g. DYKDDDDK (SEQ ID NO: 67)], one or more 3C protease cleavage site [e.g. LEVLFQGP (SEQ ID NO: 68)], one or more biotinylation site and/or His-tag sequences [e.g. HHHHHH (SEQ ID NO: 69)].
  • N- and/or C-terminal secretion signal sequences e.g. human Ig kappa chains with amino acid sequence MDMRVPAQLLGLLLLWFSGVLG (SEQ ID NO: 66)]
  • flag tag sequences e.
  • proGDF-8 AXXA NENSEQKENVEKEGLCNACTWRQNTKSSRIEAIKIQILSKLRLE 73 D76A TAPNISKDVIRQLLPKAPPLRELIDQYDVQRADSSDGSLEDDDY
  • proGDF-11 AEGPAAAAAAAAAAAAAGVGGERSSRPAPSVAPEPDGCPVC 4
  • proGDF-11 D98A AEGPAAAAAAAAAAAAAGVGGERSSRPAPSVAPEPDGCPVC 74
  • proGDF-11 AxxA AEGPAAAAAAAAAAAAAGVGGERSSRPAPSVAPEPDGCPVC 76
  • proGDF-11 AxxA AEGPAAAAAAAAAAAGVGGERSSRPAPSVAPEPDGCPVC 77 D98A VWRQHSRELRLESIKSQILSKLRLKEAPNISREVVKQLLPKAPP
  • Activins and inhibins are TGF- ⁇ family member proteins, the activity of each of which often results in opposing functions (Bilezikjian, L.M. et al., 2012. Mol Cell
  • Activins and inhibins are constructed in part from the same ⁇ - subunits, that may include inhibin-beta A, inhibin-beta B, inhibin-beta C and inhibin-beta E (referred to herein as ⁇ -subunit A, B, C and E, respectively).
  • ⁇ -subunit A, B, C and E The difference between activins and inhibins, structurally, is that activins are ⁇ -subunit dimers while inhibins are
  • Activins are named for their subunit pairs, such that activin A comprises a homodimer of two A subunits, activin AB comprises a dimer of A and B subunits, B comprises a dimer of B subunits, etc. (Muenster, U. et al., 2011. Vitam Horm. 85: 105-28). Activins are involved in a variety of functions that may include, but are not limited to cell growth, differentiation, programmed cell death, endocrine functions, cellular metabolism, bone growth, etc. They are especially recognized for their control of reproductive hormone cycles. Activin and inhibin signaling often functions antagonistically in this regard.
  • recombinant proteins of the present disclosure may comprise integrins.
  • Integrins are cell surface heterodimers formed by alpha and beta subunits, each of which has a transmembrane domain and in the N-terminal portion of the extracellular domain come together to form the ligand binding site.
  • Recombinant proteins of the present disclosure may comprise integrins and/or integrin subunits.
  • Such integrins and/or integrin subunits may comprise any of those disclosed in International Patent Application No.
  • Recombinant proteins of the disclosure may include intercellular adhesion molecule 1 (ICAM-1).
  • ICAM-1 proteins of the present disclosure may be used as control proteins during antibody development and/or antibody testing.
  • ICAM-1 may be used as a control during selection of binding molecules using phage display technologies.
  • ICAM-1 proteins of the disclosure comprise one or more detectable label. Detectable labels may include, for example, histidine tags.
  • recombinant GPCs of the present disclosure may comprise mutations in one or more N-terminal regions for extracellular associations.
  • N-terminal region for extracellular association refers to regions at or near protein N-termini that may be necessary for extracellular associations with one or more N-terminal regions. Such regions may comprise at least the first N-terminal residue, at least the first 5 N- terminal residues, at least the first 10 N-terminal residues, at least the first 20 amino acid residues and/or at least the first 50 amino acid residues.
  • Some mutations may comprise from about 1 amino acid residue to about 30 amino acid residues, from about 5 amino acid residues to about 40 amino acid residues and/or from about 10 amino acid residues to about 50 amino acid residues at or near protein N-termini.
  • Such regions may comprise residues for association with extracellular proteins (e.g. LTBPs, fibrillins, GASPs, perlecan and/or decorin).
  • LTBPs extracellular proteins
  • GASPs perlecan and/or decorin
  • cysteine residues present within and/or near N-terminal regions for extracellular associations may be necessary for such associations.
  • extracellular associations are present within about the first 5 N-terminal residues, about the first 10 N-terminal residues, about the first 20 N-terminal residues, about the first 50 N- terminal residues, about the first 60 N-terminal residues, about the first 70 N-terminal residues and/or at least the first 100 N-terminal residues.
  • Some mutations in one or more N- terminal regions for extracellular associations comprise substitution and/or deletion of such cysteine residues.
  • Such mutations may modulate the association of GPCs and/or prodomains with one or more extracellular proteins, including, but not limited to LTBPs, fibrillins, GASPs, perlecan and/or decorin. These mutations may also comprise substitution of one or more cysteine with another amino acid.
  • Cysteine residue substitutions are abbreviated herein as "C#X" wherein # represents the residue number [counting from the N-terminus of the pro- protein (without the signal peptide)] of the original cysteine residue and X represents the one letter amino acid code for the amino acid that is used for substitution. Any amino acid may be used for such substitutions.
  • serine (S) residues are used to substitute cysteine residues.
  • Nonlimiting examples of such mutations in GDF-8 may include C16S, C19S, C38S and/or C41S.
  • one or more cysteine in one or more other region of GPCs may be substituted or deleted.
  • such GPC modifications may promote the release of mature growth factor from prodomains.
  • cysteines or other residues may include those present in one or more of mature growth factors, alpha 2 helices, fasteners, latency lassos and/or bow-tie regions.
  • recombinant proteins of the present disclosure may comprise protein modules derived from one or more species, including mammals, including, but not limited to mice, rats, rabbits, pigs, monkeys and/or humans.
  • Recombinant proteins may comprise one or more amino acids from one or more amino acid sequences derived from one or more non-human protein sequences listed in Table 5.
  • recombinant proteins of the present disclosure may comprise such sequences with or without the native signal peptide. In some cases, these sequences are expressed in association with N- and/or C- terminal secretion signal sequences [e.g. human Ig kappa chains with amino acid sequence MDMRVPAQLLGLLLLWFSGVLG (SEQ ID NO: 66)], flag tag sequences [e.g.
  • DYKDDDDK (SEQ ID NO: 67)]
  • one or more 3C protease cleavage site e.g. LEVLFQGP (SEQ ID NO: 68)
  • one or more biotinylation site and/or His-tag sequences e.g. HHHHHH (SEQ ID NO: 69)].
  • proGDF-8 Mouse NEGSEREENVEKEGLCNACAWRQNTRYSRIEAIKIQILS 88
  • proGDF-8 Mouse NEGSEREENVEKEGLCNACAWRQNTRYSRIEAIKIQILS 89 AxxA KLRLETAPNISKDAIRQLLPRAPPLRELIDQYDVQRDDSS
  • proGDF-8 Mouse NEGSEREENVEKEGLCNACAWRQNTRYSRIEAIKIQILS 90 D76A KLRLETAPNISKDAIRQLLPRAPPLRELIDQYDVQRADSS
  • proGDF-8 Mouse NEGSEREENVEKEGLCNACAWRQNTRYSRIEAIKIQILS 91 AxxA D76A KLRLETAPNISKDAIRQLLPRAPPLRELIDQYDVQRADSS

Abstract

Provided herein are proteins, antibodies, assays, compositions and methods useful for modulating growth factor levels and/or activities. In some embodiments, such growth factors are members of the TGF-β superfamily of proteins.

Description

TRANSFORMING GROWTH FACTOR-RELATED ANTIBODIES AND USES
THEREOF
CROSS REFERENCE TO RELATED APPLICATIONS
[0001] This application claims priority under 35 U.S.C. § 119(e) of U.S. Provisional Patent Application No. 62/076,230, filed on November 6, 2014 and entitled
"TRANSFORMING GROWTH FACTOR-RELATED ANTIBODIES AND USES
THEREOF"; U.S. Provisional Patent Application No. 62/100,361, filed on January 6, 2015 and entitled "TRANSFORMING GROWTH FACTOR-RELATED ANTIBODIES AND USES THEREOF"; and U.S. Provisional Patent Application No. 62/187,348, filed on July 1, 2015 and entitled "TRANSFORMING GROWTH FACTOR-RELATED ANTIBODIES AND USES THEREOF". Each of these applications is incorporated herein by reference in its entirety for all purposes.
FIELD OF THE DISCLOSURE
[0002] Embodiments of the present disclosure may include modulators of growth factor activity. In some embodiments, such modulators may include antibodies and may modulate TGF-β family member activity and/or biology.
BACKGROUND OF THE DISCLOSURE
[0003] Cell signaling molecules stimulate a variety of cellular activities. Such signaling is often tightly regulated, often through interactions with other biomolecules, the extracellular and/or cellular matrix or within a particular cell environment or niche. Such interactions may be direct or indirect.
[0004] Cell signaling cascades are involved in a number of diverse biological pathways including, but not limited to modulation of cell growth, modulation of tissue homeostasis, extracellular matrix (ECM) dynamics, modulation of cell migration, invasion and immune modulation/suppression. In some cases, proteins involved in cell signaling are synthesized and/or are sequestered in latent form, requiring stimulus of some kind to participate in signaling events. There remains a need in the art for agents, tools and methods for modulating cell signaling and/or cellular activities. SUMMARY OF THE DISCLOSURE
[0005] Aspects of the disclosure relate to a recognition that in some embodiments antibodies have selected binding profiles to TGFP family member proteins and/or forms thereof. In some embodiments antibodies with selected binding profiles specifically bind to different forms of TGFP family member proteins. For example, antibodies provided herein specifically bind to one or more of a pro-form, a latent-form, a primed-form, and/or a mature- form of a TGFP family member protein, such as proMyostatin, latent Myostatin, primed Myostatin and/or mature Myostatin. In some embodiments, such antibodies inhibit
Myostatin signaling. In some embodiments, inhibition of Myostatin signaling is useful for increasing muscle mass or preventing muscle atrophy. In some embodiments, antibodies provided herein bind to and prevent cleavage of Myostatin by a proprotein convertase and/or a tolloid protease. Preventing cleavage of proMyostatin or latent Myostatin, in some embodiments, prevents Myostatin activation. Further aspects of the disclosure relate to antibodies having selected binding profiles and an affinity to an antigen that is sensitive to pH. Furthermore, in some embodiments, antibodies provided herein are sweeping antibodies that can efficiently clear antigens (e.g., proMyostatin and/or latent Myostatin) from serum.
[0006] The present disclosure, in some aspects, includes an antibody that specifically binds human proMyostatin, human latent Myostatin, human GDF-8 prodomain, human GDF- 11 ARM8 prodomain, and human proGDFl 1 ARM8, but does not specifically bind human mature GDF-8 having an amino acid sequence as set forth in SEQ ID NO: 42.
[0007] Another aspect of the invention includes an antibody that specifically binds human proMyostatin, murine proMyostatin, human latent Myostatin, human GDF-8 prodomain, human GDF-11 ARM8 prodomain, and human proGDFl 1 ARM8, but does not specifically bind human mature GDF-8 having an amino acid sequence as set forth in SEQ ID NO: 42.
[0008] In some embodiments, human proMyostatin has an amino acid sequence as set forth in SEQ ID NO: 5. In some embodiments, murine proMyostatin has an amino acid sequence as set forth in SEQ ID NO: 88. In some embodiments, human latent Myostatin has an amino acid sequence as set forth in SEQ ID NO: 5. In some embodiments, human GDF-8 prodomain has an amino acid sequence as set forth in SEQ ID NO: 38. In some
embodiments, human GDF- 11 ARM8 prodomain has an amino acid sequence as set forth in SEQ ID NO: 181. In some embodiments, human proGDFl 1 ARM8 has an amino acid sequence as set forth in SEQ ID NO: 179. In some embodiments, an antibody specifically binds human latent proGDFl 1 ARM8, but does not specifically bind human proGDFl 1 having an amino acid sequence as set forth in SEQ ID NO: 4. In other embodiments, an antibody specifically binds murine latent myostatin and human latent proGDFl 1 ARM8, but does not specifically bind human proGDFl 1 having an amino acid sequence as set forth in SEQ ID NO: 4. In some embodiments, a murine latent myostatin has an amino acid sequence as set forth in SEQ ID NO: 88. In some embodiments, a human latent proGDFl 1 ARM8 has an amino acid sequence as set forth in SEQ ID NO: 179.
[0009] In another embodiment, an antibody comprises a CDR-H3 amino acid sequence set forth in SEQ ID NO: 752, 780, 815, 772, 784, 770, 774, 756, 796, 785, 761, 788, 767, 765, 799, 749, 802, 809, 750, 803, 800, 806, 759, 805, 797, 787, or 766, 782, 967, 776, 755, 876, 891, 963, 996, 993, 970, 968 or 1008. In a further embodiment, an antibody comprises a CDR-L3 amino acid sequence set forth in any one of SEQ ID NO: 1367, 1394, 1660, 1387, 1397, 1385, 1388, 1371, 1409, 1398, 1376, 1401, 1382, 1380, 1656, 1364, 1413, 1418, 1365, 1658, 1657, 1415, 1374, 1659, 1410, 1400, 1381, 1395, 1582, 1390, 1370, 1670, 1485, 1578, 1611, 1608, 1585, 1583, or 1623.
[0010] In other embodiments, an antibody comprises a CDR-H1, a CDR-H2, and/or a CDR-H3 from the variable heavy chain (VH) amino acid sequence of clone BP6a-l, BP6a-2, BP6a-3, BP6a-4, BP6a-5, BP6a-6, BP6a-7, BP6a-8, BP6a-9, BP6a-10, BP6a-l l, BP6a-12, BP6a-13, BP6a-14, BP6a-15, BP6a-16, BP6a-17, BP6a-18, BP6a-19, BP6a-20, BP6a-21, BP6a-22, BP6a-23, BP6a-24, BP6a-25, BP6a-26, BP6a-27, BP6b-l, BP6b-2, BP6b-3, BP6b- 4, BP6b-5, BP6b-6, BP6b-7, BP6b-8, BP6b-9, BP6b-10, BP6b-l l or BP6b-12 as set forth in Table 14, and/or a CDR-L1, a CDR-L2, and/or a CDR-L3 from the variable light chain (VL) amino acid sequence of clone BP6a-l, BP6a-2, BP6a-3, BP6a-4, BP6a-5, BP6a-6, BP6a-7, BP6a-8, BP6a-9, BP6a-10, BP6a-l l, BP6a-12, BP6a-13, BP6a-14, BP6a-15, BP6a-16, BP6a- 17, BP6a-18, BP6a-19, BP6a-20, BP6a-21, BP6a-22, BP6a-23, BP6a-24, BP6a-25, BP6a-26, BP6a-27, BP6b-l, BP6b-2, BP6b-3, BP6b-4, BP6b-5, BP6b-6, BP6b-7, BP6b-8, BP6b-9, BP6b-10, BP6b-l l or BP6b-12 as set forth in Table 16.
[0011] In some embodiments, an antibody comprises a variable heavy chain amino acid sequence of clone BP6a-l, BP6a-2, BP6a-3, BP6a-4, BP6a-5, BP6a-6, BP6a-7, BP6a-8, BP6a-9, BP6a-10, BP6a-l l, BP6a-12, BP6a-13, BP6a-14, BP6a-15, BP6a-16, BP6a-17, BP6a-18, BP6a-19, BP6a-20, BP6a-21, BP6a-22, BP6a-23, BP6a-24, BP6a-25, BP6a-26, BP6a-27, BP6b-l, BP6b-2, BP6b-3, BP6b-4, BP6b-5, BP6b-6, BP6b-7, BP6b-8, BP6b-9, BP6b-10, BP6b-l 1 or BP6b-12 as set forth in Table 14. In other embodiments, an antibody comprises the variable light chain amino acid sequence of clone BP6a-l, BP6a-2, BP6a-3, BP6a-4, BP6a-5, BP6a-6, BP6a-7, BP6a-8, BP6a-9, BP6a-10, BP6a-l l, BP6a-12, BP6a-13, BP6a-14, BP6a-15, BP6a-16, BP6a-17, BP6a-18, BP6a-19, BP6a-20, BP6a-21, BP6a-22, BP6a-23, BP6a-24, BP6a-25, BP6a-26, BP6a-27, BP6b-l, BP6b-2, BP6b-3, BP6b-4, BP6b-5, BP6b-6, BP6b-7, BP6b-8, BP6b-9, BP6b-10, BP6b-l l or BP6b-12 as set forth in Table 16.
[0012] Further aspects of the present disclosure include an antibody that specifically binds human proMyostatin, and human proGDFl 1 ARM8 but does not specifically bind human latent Myostatin having an amino acid sequence as set forth in SEQ ID NO: 5, human GDF-8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 38, human GDF-11 ARM8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 181, or human mature GDF-8 having an amino acid sequence as set forth in SEQ ID NO: 42.
[0013] Another aspect of the present disclosure includes an antibody that specifically binds human proMyostatin, murine proMyostatin and human proGDFl 1 ARM8 but does not specifically bind human latent Myostatin having an amino acid sequence as set forth in SEQ ID NO: 5, human GDF-8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 38, human GDF-11 ARM8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 181, or human mature GDF-8 having an amino acid sequence as set forth in SEQ ID NO: 42.
[0014] In some embodiments, the human proMyostatin has an amino acid sequence as set forth in SEQ ID NO: 5. In other embodiments, the murine proMyostatin has an amino acid sequence as set forth in SEQ ID NO: 88. In another embodiment, the human proGDFl 1 ARM8 has an amino acid sequence as set forth in SEQ ID NO: 179. In some embodiments, the antibody comprises a CDR-H3 amino acid sequence set forth in SEQ ID NO: 813. In other embodiments, the antibody comprises a CDR-L3 amino acid sequence set forth in any one of SEQ ID NO: 1422. In another embodiment, the antibody comprises a CDR-H1, a CDR-H2, and/or a CDR-H3 from the variable heavy chain (VH) amino acid sequence of clone BP2a-l as set forth in Table 14, and/or a CDR-L1, a CDR-L2, and/or a CDR-L3 from the variable light chain (VL) amino acid sequence of clone BP2a-l as set forth in Table 16. In some embodiments, the antibody comprises a variable heavy chain amino acid sequence of clone BP2a-l as set forth in Table 14. In some embodiments, the antibody comprises the variable light chain amino acid sequence of clone BP2a-l as set forth in Table 16.
[0015] The present disclosure, in some aspects, includes an antibody that specifically binds human proMyostatin, and human latent Myostatin, but does not specifically bind human GDF-8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 38, human GDF- 11 ARM8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 181, human proGDFl 1 ARM8 having an amino acid sequence as set forth in SEQ ID NO: 179, or human mature GDF-8 having an amino acid sequence as set forth in SEQ ID NO: 42.
[0016] Another aspect of the disclosure includes an antibody that specifically binds human proMyostatin, murine proMyostatin, and human latent Myostatin, but does not specifically bind human GDF-8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 38, human GDF-11 ARM8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 181, human proGDFl 1 ARM8 having an amino acid sequence as set forth in SEQ ID NO: 179, or human mature GDF-8 having an amino acid sequence as set forth in SEQ ID NO: 42.
[0017] In some embodiments, a human proMyostatin has an amino acid sequence as set forth in SEQ ID NO: 5. In other embodiments, a murine proMyostatin has an amino acid sequence as set forth in SEQ ID NO: 88. In some embodiments, a human latent Myostatin has an amino acid sequence as set forth in SEQ ID NO: 5. In another embodiment, a antibody comprises a CDR-H3 amino acid sequence set forth in SEQ ID NO: 794, 804, or 751. In some embodiments, the antibody comprises a CDR-L3 amino acid sequence set forth in any one of SEQ ID NO: 1407, 1414, or 1366. In other embodiments, the antibody comprises a CDR-H1, a CDR-H2, and/or a CDR-H3 from the variable heavy chain (VH) amino acid sequence of clone BP3a-l, BP3a-2 or BP3a-3 as set forth in Table 14, and/or a CDR-L1, a CDR-L2, and/or a CDR-L3 from the variable light chain (VL) amino acid sequence of clone BP3a-l, BP3a-2 or BP3a-3 as set forth in Table 16. In some embodiments, the antibody comprises a variable heavy chain amino acid sequence of clone BP3a-l, BP3a-2 or BP3a-3 as set forth in Table 14. In some embodiments, the antibody comprises the variable light chain amino acid sequence of clone BP3a-l, BP3a-2 or BP3a-3 as set forth in Table 16.
[0018] Aspects of the present disclosure relate to an antibody that specifically binds human proMyostatin, human latent Myostatin and human GDF8 prodomain but does not specifically bind human GDF-11 ARM 8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 181, human proGDFl 1 ARM8 having an amino acid sequence as set forth in SEQ ID NO: 179 or human mature GDF-8 having an amino acid sequence as set forth in SEQ ID NO: 42.
[0019] Further aspects of the disclosure include an antibody that specifically binds human proMyostatin, murine proMyostatin, human latent Myostatin and human GDF8 prodomain but does not specifically bind human GDF- 11 ARM8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 181, human proGDFl 1 ARM8 having an amino acid sequence as set forth in SEQ ID NO: 179 or human mature GDF-8 having an amino acid sequence as set forth in SEQ ID NO: 42.
[0020] In some embodiments, the human proMyo statin has an amino acid sequence as set forth in SEQ ID NO: 5. In another embodiment, the murine proMyo statin has an amino acid sequence as set forth in SEQ ID NO: 88. In a further embodiment, the human latent Myostatin has an amino acid sequence as set forth in SEQ ID NO: 5. In some embodiments, the human GDF-8 prodomain has an amino acid sequence as set forth in SEQ ID NO: 38. In other embodiments, the antibody comprises a CDR-H3 amino acid sequence set forth in SEQ ID NO: 768. In further embodiments, the antibody comprises a CDR-L3 amino acid sequence set forth in any one of SEQ ID NO: 1383. In some embodiments, the antibody comprises a CDR-H1, a CDR-H2, and/or a CDR-H3 from the variable heavy chain (VH) amino acid sequence of clone BP4a-l as set forth in Table 14, and/or a CDR-L1, a CDR-L2, and/or a CDR-L3 from the variable light chain (VL) amino acid sequence of clone BP4a- 1 as set forth in Table 16. In other embodiments, the antibody comprises a variable heavy chain amino acid sequence of clone BP4a-l as set forth in Table 14. In another embodiment, the antibody comprises the variable light chain amino acid sequence of clone BP4a- 1 as set forth in Table 16.
[0021] Aspects of the disclosure further relate to an antibody that specifically binds human proMyostatin, human latent Myostatin, and human proGDFl 1 ARM8, but does not specifically bind human GDF-8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 38, human GDF-11 ARM8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 181, or human mature GDF-8 having an amino acid sequence as set forth in SEQ ID NO: 42.
[0022] Additional aspects of the disclosure include an antibody that specifically binds human proMyostatin, murine proMyostatin, human latent Myostatin, and human proGDFl 1 ARM8, but does not specifically bind human GDF-8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 38, human GDF-11 ARM8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 181, or human mature GDF-8 having an amino acid sequence as set forth in SEQ ID NO: 42.
[0023] In some embodiments, the human proMyostatin has an amino acid sequence as set forth in SEQ ID NO: 5. In other embodiments, the murine proMyostatin has an amino acid sequence as set forth in SEQ ID NO: 88. In a further embodiment, the human latent Myostatin has an amino acid sequence as set forth in SEQ ID NO: 5. In another embodiment, the human proGDFl 1 ARM8 has an amino acid sequence as set forth in SEQ ID NO: 179. In some embodiments, the antibody specifically binds human latent proGDFl 1 ARM8, but does not specifically bind human proGDFl 1 having an amino acid sequence as set forth in SEQ ID NO: 4. In some embodiments, the antibody specifically binds murine latent myostatin and human latent proGDFl 1 ARM8, but does not specifically bind human proGDFl 1 having an amino acid sequence as set forth in SEQ ID NO: 4. In other embodiments, the murine latent myostatin has an amino acid sequence as set forth in SEQ ID NO: 88. In another
embodiment, the human latent proGDFl 1 ARM8 has an amino acid sequence as set forth in SEQ ID NO: 179. In other embodiments, the antibody comprises a CDR-H3 amino acid sequence set forth in SEQ ID NO: 760, 798, 792, 789, 801, 790, 812, 758, 762, 775, 834, 974, 985, or 1002. In further embodiments, the antibody comprises a CDR-L3 amino acid sequence set forth in any one of SEQ ID NO: 1375, 1411, 1405, 1402, 1412, 1403, 1421, 1373, 1377, 1389, 1661, 1589, 1600, or 1617.
[0024] In another embodiment, the antibody comprises a CDR-H1, a CDR-H2, and/or a CDR-H3 from the variable heavy chain (VH) amino acid sequence of clone BP5a-l, BP5a-2, BP5a-3, BP5a-4 or BP5a-5, BP5b-l, BP5b-2, BP5b-3, BP5b-4, BP5b-5, BP5b-6, BP5b-7, BP5b-8 or BP5b-9 as set forth in Table 14, and/or a CDR-L1, a CDR-L2, and/or a CDR-L3 from the variable light chain (VL) amino acid sequence of clone BP5a-l, BP5a-2, BP5a-3, BP5a-4 or BP5a-5, BP5b-l, BP5b-2, BP5b-3, BP5b-4, BP5b-5, BP5b-6, BP5b-7, BP5b-8 or BP5b-9 as set forth in Table 16. In other embodiments, the antibody comprises a variable heavy chain amino acid sequence of clone BP5a-l, BP5a-2, BP5a-3, BP5a-4 or BP5a-5, BP5b-l, BP5b-2, BP5b-3, BP5b-4, BP5b-5, BP5b-6, BP5b-7, BP5b-8 or BP5b-9 as set forth in Table 14. In still other embodiments, the antibody comprises the variable light chain amino acid sequence of clone BP5a-l, BP5a-2, BP5a-3, BP5a-4 or BP5a-5, BP5b-l, BP5b-2, BP5b-3, BP5b-4, BP5b-5, BP5b-6, BP5b-7, BP5b-8 or BP5b-9 as set forth in Table 16.
[0025] Aspects of the present disclosure include an antibody that specifically binds human proMyo statin but does not specifically bind human latent Myostatin having an amino acid sequence as set forth in SEQ ID NO: 5, human GDF-8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 38, human GDF-11 ARM8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 181, human proGDFl 1 ARM8 having an amino acid sequence as set forth in SEQ ID NO: 179, or human mature GDF-8 having an amino acid sequence as set forth in SEQ ID NO: 42.
[0026] Further aspects of the invention include an antibody that specifically binds human proMyostatin and murine proMyostatin but does not specifically bind human latent Myostatin having an amino acid sequence as set forth in SEQ ID NO: 5, human GDF-8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 38, human GDF-11 ARM8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 181, human proGDFl 1 ARM8 having an amino acid sequence as set forth in SEQ ID NO: 179, or human mature GDF-8 having an amino acid sequence as set forth in SEQ ID NO: 42.
[0027] In some embodiments, the human proMyo statin has an amino acid sequence as set forth in SEQ ID NO: 5. In other embodiments, the murine proMyo statin has an amino acid sequence as set forth in SEQ ID NO: 88). In another embodiment, the antibody does not specifically bind human proGDFl 1 having an amino acid sequence as set forth in SEQ ID NO: 4, or human latent proGDFl 1 ARM8 having an amino acid sequence as set forth in SEQ ID NO: 179. In yet another embodiment, the antibody does not specifically bind human proGDFl 1 having an amino acid sequence as set forth in SEQ ID NO: 4, murine latent myostatin having an amino acid sequence as set forth in SEQ ID NO: 88 or human latent proGDFl 1 ARM8 having an amino acid sequence as set forth in SEQ ID NO: 179. In some embodiments, the antibody comprises a CDR-H3 amino acid sequence as set forth in SEQ ID NO: 769 or 827. In other embodiments, the antibody comprises a CDR-L3 amino acid sequence set forth in any one of SEQ ID NO: 1384 or 1435. In another embodiment, the antibody comprises a CDR-H1, a CDR-H2, and/or a CDR-H3 from the variable heavy chain (VH) amino acid sequence of clone BPla-1 or BPlb-1 as set forth in Table 14, and/or a CDR-L1, a CDR-L2, and/or a CDR-L3 from the variable light chain (VL) amino acid sequence of clone BPla-1 or BPlb-1 as set forth in Table 16. In a further embodiment, the antibody comprises a variable heavy chain amino acid sequence of clone BPla-1 or BPlb-1 as set forth in Table 14. In some embodiments, the antibody comprises the variable light chain amino acid sequence of clone BPla-1 or BPlb-1 as set forth in Table 16.
[0028] Further aspects of the invention relate to an antibody that specifically binds human latent Myostatin but does not specifically bind human proMyo statin having an amino acid sequence as set forth in SEQ ID NO: 5, human GDF-8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 38, human GDF-11 ARM8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 181, human proGDFl 1 ARM8 having an amino acid sequence as set forth in SEQ ID NO: 179, human mature GDF-8 having an amino acid sequence as set forth in SEQ ID NO: 42 or human proGDFl 1 having an amino acid sequence as set forth in SEQ ID NO: 4).
[0029] Additional aspects of the invention include an antibody that specifically binds human latent Myostatin but does not specifically bind murine proMyostatin having an amino acid sequence as set forth in SEQ ID NO: 88, human proMyostatin having an amino acid sequence as set forth in SEQ ID NO: 5, human GDF-8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 38, human GDF-11 ARM8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 181, human proGDFl 1 ARM8 having an amino acid sequence as set forth in SEQ ID NO: 179, human mature GDF-8 having an amino acid sequence as set forth in SEQ ID NO: 42 or human proGDFl 1 having an amino acid sequence as set forth in SEQ ID NO: 4).
[0030] In some embodiments, the human latent Myostatin has an amino acid sequence as set forth in SEQ ID NO: 5. In other embodiments, the antibody specifically binds human latent proGDFl 1 ARM8. In a further embodiment, the antibody specifically binds murine latent myostatin and human latent proGDFl 1 ARM8. In another embodiment, the murine latent myostatin has an amino acid sequence as set forth in SEQ ID NO: 88. In some embodiments, the human latent proGDFl 1 ARM8 has an amino acid sequence as set forth in SEQ ID NO: 179. In other embodiments, the antibody comprises a CDR-H3 amino acid sequence set forth in SEQ ID NO: 990. In some embodiments, the antibody comprises a CDR-L3 amino acid sequence set forth in any one of SEQ ID NO: 1605. In other embodiments, the antibody comprises a CDR-H1, a CDR-H2, and/or a CDR-H3 from the variable heavy chain (VH) amino acid sequence of clone BPlOb-1 as set forth in Table 14, and/or a CDR-L1, a CDR-L2, and/or a CDR-L3 from the variable light chain (VL) amino acid sequence of clone BPlOb-1 as set forth in Table 16. In further embodiments, the antibody comprises a variable heavy chain amino acid sequence of clone BPlOb-1 as set forth in Table 14. In other embodiments, the antibody comprises the variable light chain amino acid sequence of clone BPlOb-las set forth in Table 16.
[0031] Some aspects of the present disclosure include an antibody that specifically binds human proMyostatin, human latent Myostatin, human proGDFl 1 ARM8 and human mature GDF-8 but does not specifically bind human GDF-8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 38 or human GDF-11 ARM8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 181.
[0032] Another aspect of the present disclosure relates to an antibody that specifically binds human proMyostatin, murine proMyostatin, human latent Myostatin, human proGDFl 1 ARM8 and human mature GDF-8 but does not specifically bind human GDF-8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 38 or human GDF-11 ARM8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 181.
[0033] In some embodiments, the human proMyostatin has an amino acid sequence as set forth in SEQ ID NO: 5. In other embodiments, the murine proMyostatin has an amino acid sequence as set forth in SEQ ID NO: 88. In another embodiment, the human latent Myostatin has an amino acid sequence as set forth in SEQ ID NO: 5. In some embodiments, the human proGDFl 1 ARM8 has an amino acid sequence as set forth in SEQ ID NO: 179. In another embodiment, the human mature GDF-8 has an amino acid sequence as set forth in SEQ ID NO: 42. In other embodiments, the antibody comprises a CDR-H3 amino acid sequence set forth in SEQ ID NO: 764. In some embodiments, the antibody comprises a CDR-L3 amino acid sequence set forth in any one of SEQ ID NO: 1379. In other embodiments, the antibody comprises a CDR-H1, a CDR-H2, and/or a CDR-H3 from the variable heavy chain (VH) amino acid sequence of clone BPl la-1 as set forth in Table 14, and/or a CDR-L1, a CDR-L2, and/or a CDR-L3 from the variable light chain (VL) amino acid sequence of clone BPl la-1 as set forth in Table 16. In another embodiment, the antibody comprises a variable heavy chain amino acid sequence of clone BPl la-1 as set forth in Table 14. In some embodiments, the antibody comprises the variable light chain amino acid sequence of clone BPl la-1 as set forth in Table 16.
[0034] Additional aspects of the present disclosure include an antibody that specifically binds human latent Myostatin and human mature GDF-8 but does not specifically bind human proMyostatin having an amino acid sequence as set forth in SEQ ID NO: 5, human GDF-8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 38, human GDF- 11 ARM8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 181 or human proGDFl 1 ARM8 having an amino acid sequence as set forth in SEQ ID NO: 179.
[0035] Further aspects of the present disclosure relate to an antibody that specifically binds human latent Myostatin and human mature GDF-8 but does not specifically bind murine proMyostatin having an amino acid sequence as set forth in SEQ ID NO: 88, human proMyostatin having an amino acid sequence as set forth in SEQ ID NO: 5, human GDF-8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 38, human GDF-11 ARM8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 181 or human proGDFl 1 ARM8 having an amino acid sequence as set forth in SEQ ID NO: 179.
[0036] In some embodiments, the human latent Myostatin has an amino acid sequence as set forth in SEQ ID NO: 5. In other embodiments, the human mature GDF-8 has an amino acid sequence as set forth in SEQ ID NO: 42. In another embodiment, the antibody does not specifically bind human proGDFl 1 having an amino acid sequence as set forth in SEQ ID NO: 4 or human latent proGDFl 1 ARM8 having an amino acid sequence as set forth in SEQ ID NO: 179. In some embodiments, the antibody specifically binds murine latent myostatin but does not specifically bind human proGDFl 1 having an amino acid sequence as set forth in SEQ ID NO: 4 or human latent proGDFl 1 ARM8 having an amino acid sequence as set forth in SEQ ID NO: 179. In other embodiments, the murine latent myostatin has an amino acid sequence as set forth in SEQ ID NO: 88. In another embodiment, the antibody comprises a CDR-H3 amino acid sequence set forth in SEQ ID NO: 989. In some
embodiments, the antibody comprises a CDR-L3 amino acid sequence set forth in any one of SEQ ID NO: 1604. In other embodiments, the antibody comprises a CDR-H1, a CDR-H2, and/or a CDR-H3 from the variable heavy chain (VH) amino acid sequence of clone BP12b-l as set forth in Table 14, and/or a CDR-L1, a CDR-L2, and/or a CDR-L3 from the variable light chain (VL) amino acid sequence of clone BP 12b- las set forth in Table 16. In another embodiment, the antibody comprises a variable heavy chain amino acid sequence of clone BP12b-l as set forth in Table 14. In some embodiments, the antibody comprises the variable light chain amino acid sequence of clone BP12b-l as set forth in Table 16.
[0037] Further aspects of the present disclosure include an antibody that specifically binds human proMyo statin and human GDF-8 prodomain, but does not specifically bind human latent Myostatin having an amino acid sequence as set forth in SEQ ID NO: 5, human GDF-11 ARM8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 181, human proGDFl 1 ARM8 having an amino acid sequence as set forth in SEQ ID NO: 179 or human mature GDF-8 having an amino acid sequence as set forth in SEQ ID NO: 42.
[0038] Another aspect of the present disclosure relates to an antibody that specifically binds human proMyostatin and human GDF-8 prodomain, but does not specifically bind murine proMyostatin having an amino acid sequence as set forth in SEQ ID NO: 88, human latent Myostatin having an amino acid sequence as set forth in SEQ ID NO: 5, human GDF- 11 ARM8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 181, human proGDFl 1 ARM8 having an amino acid sequence as set forth in SEQ ID NO: 179 or human mature GDF-8 having an amino acid sequence as set forth in SEQ ID NO: 42.
[0039] In some embodiments, the human proMyostatin has an amino acid sequence as set forth in SEQ ID NO: 5. In other embodiments, the human GDF-8 prodomain has an amino acid sequence as set forth in SEQ ID NO: 38. In further embodiments, the antibody comprises a CDR-H3 amino acid sequence set forth in SEQ ID NO: 786. In another embodiment, the antibody comprises a CDR-L3 amino acid sequence set forth in any one of SEQ ID NO: 1399. In some embodiments, the antibody comprises a CDR-H1, a CDR-H2, and/or a CDR-H3 from the variable heavy chain (VH) amino acid sequence of clone BP15a-l as set forth in Table 14, and/or a CDR-L1, a CDR-L2, and/or a CDR-L3 from the variable light chain (VL) amino acid sequence of clone BP15a-l as set forth in Table 16. In another embodiment, the antibody comprises a variable heavy chain amino acid sequence of clone BP15a-l as set forth in Table 14. In other embodiments, the antibody comprises the variable light chain amino acid sequence of clone BP15a-l as set forth in Table 16.
[0040] Another aspect of the present disclosure includes an antibody that specifically binds human proMyo statin and human latent Myostatin but does not specifically bind human GDF-8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 38, human GDF-11 ARM8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 181, human proGDFl 1 ARM8 having an amino acid sequence as set forth in SEQ ID NO: 179 or human mature GDF-8 having an amino acid sequence as set forth in SEQ ID NO: 42.
[0041] An additional aspect of the present disclosure relates to an antibody that specifically binds human proMyo statin and human latent Myostatin but does not specifically bind murine proMyostatin having an amino acid sequence as set forth in SEQ ID NO: 88, human GDF-8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 38, human GDF- 11 ARM8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 181, human proGDFl 1 ARM8 having an amino acid sequence as set forth in SEQ ID NO: 179 or human mature GDF-8 having an amino acid sequence as set forth in SEQ ID NO: 42.
[0042] In some embodiments, the human proMyostatin has an amino acid sequence as set forth in SEQ ID NO: 5. In other embodiments, the human latent Myostatin has an amino acid sequence as set forth in SEQ ID NO: 5. In further embodiments, the antibody does not specifically bind human proGDFl 1 having an amino acid sequence as set forth in SEQ ID NO: 4 or human latent proGDFl 1 ARM8 having an amino acid sequence as set forth in SEQ ID NO: 179. In additional embodiments, the antibody does not specifically bind human proGDFl 1 having an amino acid sequence as set forth in SEQ ID NO: 4, murine latent myostatin having an amino acid sequence as set forth in SEQ ID NO: 88 or human latent proGDFl 1 ARM8 having an amino acid sequence as set forth in SEQ ID NO: 179. In some embodiments, the antibody comprises a CDR-H3 amino acid sequence set forth in SEQ ID NO: 810, 855, 971, 983, or 986. In other embodiments, the antibody comprises a CDR-L3 amino acid sequence set forth in any one of SEQ ID NO: 1419, 1662, 1586, 1598, or 1601. In further embodiments, the antibody comprises a CDR-H1, a CDR-H2, and/or a CDR-H3 from the variable heavy chain (VH) amino acid sequence of clone BP16a-l, BP16b-l, BP16b-2, BP16b-3 or BP16b-4 as set forth in Table 14, and/or a CDR-Ll, a CDR-L2, and/or a CDR-L3 from the variable light chain (VL) amino acid sequence of clone BP16a-l, BP 16b- 1, BP16b-2, BP16b-3 or BP16b-4 as set forth in Table 16. In another embodiment, the antibody comprises a variable heavy chain amino acid sequence of clone BP16a-l, BP16b-l, BP16b-2, BP16b-3 or BP16b-4 as set forth in Table 14. In some embodiments, the antibody comprises the variable light chain amino acid sequence of clone BP16a-l, BP16b-l, BP 16b-
2, BP16b-3 or BP16b-4 as set forth in Table 16.
[0043] Additional aspects of the present disclosure include an antibody that specifically binds human proMyostatin and human latent Myostatin but does not specifically bind human GDF-8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 38, human GDF-11 ARM8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 181, human proGDFl 1 ARM8 having an amino acid sequence as set forth in SEQ ID NO: 179, human mature GDF-8 having an amino acid sequence as set forth in SEQ ID NO: 42 or human proGDFl 1 having an amino acid sequence as set forth in SEQ ID NO: 4.
[0044] Further aspects of the present disclosure relate to an antibody that specifically binds human proMyostatin and human latent Myostatin but does not specifically bind murine proMyostatin having an amino acid sequence as set forth in SEQ ID NO: 88, human GDF-8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 38, human GDF-11 ARM8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 181, human proGDFl 1 ARM8 having an amino acid sequence as set forth in SEQ ID NO: 179, human mature GDF-8 having an amino acid sequence as set forth in SEQ ID NO: 42, human proGDFl 1 having an amino acid sequence as set forth in SEQ ID NO: 4 or murine latent myostatin having an amino acid sequence as set forth in SEQ ID NO: 88. In some embodiments, the human proMyostatin has amino acid sequence as set forth in SEQ ID NO: 5. In other embodiments, the human latent Myostatin has an amino acid sequence as set forth in SEQ ID NO: 5. In another embodiment, the antibody specifically binds human latent proGDFl 1 ARM8. In other embodiments, the human latent proGDFl 1 ARM8 has an amino acid sequence as set forth in SEQ ID NO: 179. In some embodiments, the antibody does not specifically bind human latent proGDFl 1 ARM8 having an amino acid sequence as set forth in SEQ ID NO: 179. In another embodiment, the antibody comprises a CDR-H3 amino acid sequence set forth in SEQ ID NO: 982. In further embodiments, the antibody comprises a CDR-L3 amino acid sequence set forth in any one of SEQ ID NO: 1597. In some embodiments, the antibody comprises a CDR-H1, a CDR-H2, and/or a CDR-H3 from the variable heavy chain (VH) amino acid sequence of clone BP17b-l as set forth in Table 14, and/or a CDR-L1, a CDR-L2, and/or a CDR-L3 from the variable light chain (VL) amino acid sequence of clone BP17b-l as set forth in Table 16. In some embodiments, the antibody comprises a variable heavy chain amino acid sequence of clone BP17b-l as set forth in Table 14. In other embodiments, the antibody comprises the variable light chain amino acid sequence of clone BP17b-l as set forth in Table 16.
[0045] Other aspects of the present disclosure relate to an antibody that specifically binds human proMyostatin, human latent Myostatin and human proGDFl 1 ARM8 but does not specifically bind human GDF-8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 38, human GDF-11 ARM8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 181 or human mature GDF-8 having an amino acid sequence as set forth in SEQ ID NO: 42.
[0046] Another aspect of the present disclosure includes an antibody that specifically binds human proMyostatin, human latent Myostatin and human proGDFl 1 ARM8 but does not specifically bind murine proMyostatin having an amino acid sequence as set forth in SEQ ID NO: 88, human GDF-8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 38, human GDF-11 ARM8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 181 or human mature GDF-8 having an amino acid sequence as set forth in SEQ ID NO: 42. In some embodiments, the human proMyostatin has an amino acid sequence as set forth in SEQ ID NO: 5. In other embodiments, the human latent Myostatin has an amino acid sequence as set forth in SEQ ID NO: 5. In other embodiments, the human proGDFl 1 ARM8 has an amino acid sequence as set forth in SEQ ID NO: 179. In another embodiment, the antibody specifically binds human latent proGDFl 1 ARM8 but does not specifically bind human proGDFl 1 having an amino acid sequence as set forth in SEQ ID NO: 4. In some embodiments, the antibody specifically binds human latent proGDFl 1 ARM8 but does not specifically bind human proGDFl 1 having an amino acid sequence as set forth in SEQ ID NO: 4 or murine latent myostatin having an amino acid sequence as set forth in SEQ ID NO: 88. In further embodiments, the human latent proGDFl 1 ARM 8 has an amino acid sequence as set forth in SEQ ID NO: 179. In another embodiment, the antibody comprises a CDR-H3 amino acid sequence set forth in SEQ ID NO: 763, 814, 753, 777, 795, 969, 972, 976, or 981. In other embodiments, the antibody comprises a CDR-L3 amino acid sequence set forth in any one of SEQ ID NO: 1378, 1423, 1368, 1391, 1408, 1584, 1587, 1591, or 1596.
[0047] In some embodiments, the antibody comprises a CDR-H1, a CDR-H2, and/or a CDR-H3 from the variable heavy chain (VH) amino acid sequence of clone BP18a-l or BP18a-2, BP18b-l, BP18b-2, BP18b-3, BP18b-4, BP18b-5, BP18b-6 or BP18b-7 as set forth in Table 14, and/or a CDR-L1, a CDR-L2, and/or a CDR-L3 from the variable light chain (VL) amino acid sequence of clone BP18a-l or BP18a-2, BP18b-l, BP18b-2, BP18b-3, BP18b-4, BP18b-5, BP18b-6 or BP18b-7as set forth in Table 16. In some embodiments, the antibody comprises a variable heavy chain amino acid sequence of clone BP18a-l or BP 18a- 2, BP18b-l, BP18b-2, BP18b-3, BP18b-4, BP18b-5, BP18b-6 or BP18b-7 as set forth in Table 14. In some embodiments, the antibody comprises the variable light chain amino acid sequence of clone BP18a-l or BP18a-2, BP18b-l, BP18b-2, BP18b-3, BP18b-4, BP18b-5, BP18b-6 or BP18b-7 as set forth in Table 16.
[0048] Further aspects of the present disclosure include an antibody that specifically binds human proMyostatin, human latent Myostatin, human GDF-8 prodomain, human GDF-11 ARM8 prodomain and human proGDFl 1 ARM8 but does not specifically bind human mature GDF-8 having an amino acid sequence as set forth in SEQ ID NO: 42.
[0049] Additional aspects of the present disclosure relate to an antibody that specifically binds human proMyostatin, human latent Myostatin, human GDF-8 prodomain, human GDF- 11 ARM8 prodomain and human proGDFl 1 ARM8 but does not specifically bind murine proMyostatin having an amino acid sequence as set forth in SEQ ID NO: 88 or human mature GDF-8 having an amino acid sequence as set forth in SEQ ID NO: 42. In some
embodiments, the human proMyostatin has an amino acid sequence as set forth in SEQ ID NO: 5. In other embodiments, the human latent Myostatin has an amino acid sequence as set forth in SEQ ID NO: 5. In further embodiments, the human GDF-8 prodomain has an amino acid sequence as set forth in SEQ ID NO: 38. In some embodiments, the human GDF-11 ARM8 prodomain has an amino acid sequence as set forth in SEQ ID NO: 181. In some embodiments, the human proGDFl 1 ARM8 has an amino acid sequence as set forth in SEQ ID NO: 179. In other embodiments, the antibody specifically binds human latent proGDFl 1 ARM8 but does not specifically bind human proGDFl 1 having an amino acid sequence as set forth in SEQ ID NO: 4. In another embodiment, the antibody specifically binds human latent proGDFl 1 ARM8 but does not specifically bind human proGDFl 1 having an amino acid sequence as set forth in SEQ ID NO: 4 or murine latent myostatin having an amino acid sequence as set forth in SEQ ID NO: 88. In some embodiments, the human latent proGDFl 1 ARM8 has an amino acid sequence as set forth in SEQ ID NO: 179. In another embodiment, the antibody comprises a CDR-H3 amino acid sequence set forth in SEQ ID NO: 773, 807, 811, 754, 771, or 978. In other embodiments, the antibody comprises a CDR-L3 amino acid sequence set forth in any one of SEQ ID NO: 1655, 1416, 1420, 1369, 1386, or 1593. In some embodiments, the antibody comprises a CDR-H1, a CDR-H2, and/or a CDR-H3 from the variable heavy chain (VH) amino acid sequence of clone BP19a-l, BP19a-2 or BP19a-3, BP19b-l, BP19b-2 or BP19b-3 as set forth in Table 14, and/or a CDR-L1, a CDR-L2, and/or a CDR-L3 from the variable light chain (VL) amino acid sequence of clone BP19a-l, BP 19a- 2 or BP19a-3, BP19b-l, BP19b-2 or BP19b-3 as set forth in Table 16. In some embodiments, the antibody comprises a variable heavy chain amino acid sequence of clone BP19a-l, BP19a-2 or BP19a-3, BP19b-l, BP19b-2 or BP19b-3 as set forth in Table 14. In other embodiments, the antibody comprises the variable light chain amino acid sequence of clone BP19a-l, BP19a-2 or BP19a-3, BP19b-l, BP19b-2 or BP19b-3 as set forth in Table 16.
[0050] Other aspects of present disclosure relate to an antibody that specifically binds human proMyostatin, human latent Myostatin, human proGDFl 1 ARM8, human proGDFl 1 or human latent proGDFl 1 ARM8 but does not specifically bind human GDF-8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 38, human GDF-11 ARM8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 181 or human mature GDF-8 having an amino acid sequence as set forth in SEQ ID NO: 42.
[0051] Additional aspects of present disclosure include an antibody that specifically binds human proMyostatin, murine proMyostatin, human latent Myostatin, human proGDFl 1 ARM8, human proGDFl 1, murine latent myostatin or human latent proGDFl 1 ARM8 but does not specifically bind human GDF-8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 38, human GDF-11 ARM8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 181 or human mature GDF-8 having an amino acid sequence as set forth in SEQ ID NO: 42. In some embodiments, the human proMyostatin has an amino acid sequence as set forth in SEQ ID NO: 5. In other embodiments, the murine proMyostatin has an amino acid sequence as set forth in SEQ ID NO: 88. In another embodiment, the human latent Myostatin has an amino acid sequence as set forth in SEQ ID NO: 5. In some embodiments, the human proGDFl 1 ARM8 has an amino acid sequence as set forth in SEQ ID NO: 179. In other embodiments, the human proGDFl 1 has an amino acid sequence as set forth in SEQ ID NO: 4. In other embodiments, the murine latent myostatin has an amino acid sequence as set forth in SEQ ID NO: 88. In some embodiments, the human latent proGDFl 1 ARM8 has an amino acid sequence as set forth in SEQ ID NO: 179. In another embodiment, the antibody comprises a CDR-H3 amino acid sequence set forth in SEQ ID NO: 779. In other embodiments, the antibody comprises a CDR-L3 amino acid sequence set forth in any one of SEQ ID NO: 1393. In some embodiments, the antibody comprises a CDR-H1, a CDR-H2, and/or a CDR-H3 from the variable heavy chain (VH) amino acid sequence of clone BP21b-l as set forth in Table 14, and/or a CDR-L1, a CDR-L2, and/or a CDR-L3 from the variable light chain (VL) amino acid sequence of clone BP21b-l as set forth in Table 16. In some embodiments, the antibody comprises a variable heavy chain amino acid sequence of clone BP21b-l as set forth in Table 14. In other embodiments, the antibody comprises the variable light chain amino acid sequence of clone BP21b-l as set forth in Table 16.
[0052] Additional aspects of the present disclosure relate to an antibody that specifically binds human proMyostatin, human latent Myostatin, human proGDFl 1, and human latent proGDFl 1 ARM8 but does not specifically bind human GDF-8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 38, human GDF-11 ARM8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 181, human proGDFl 1 ARM8 having an amino acid sequence as set forth in SEQ ID NO: 179 or human mature GDF-8 having an amino acid sequence as set forth in SEQ ID NO: 42.
[0053] Further aspects of the present disclosure include an antibody that specifically binds human proMyostatin, murine proMyostatin, human latent Myostatin, human proGDFl 1, murine latent myostatin and human latent proGDFl 1 ARM8 but does not specifically bind human GDF-8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 38, human GDF-11 ARM8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 181, human proGDFl 1 ARM8 having an amino acid sequence as set forth in SEQ ID NO: 179 or human mature GDF-8 having an amino acid sequence as set forth in SEQ ID NO: 42. In some embodiments, the human proMyostatin has an amino acid sequence as set forth in SEQ ID NO: 5. In other embodiments, the murine proMyostatin has an amino acid sequence as set forth in SEQ ID NO: 88. In another embodiment, the human latent Myostatin has an amino acid sequence as set forth in SEQ ID NO: 5. In some embodiments, the human proGDFl 1 has an amino acid sequence as set forth in SEQ ID NO: 4. In another embodiment, the murine latent myostatin has an amino acid sequence as set forth in SEQ ID NO: 88. In some embodiments, the human latent proGDFl 1 ARM8 has an amino acid sequence as set forth in SEQ ID NO: 179. In other embodiments, the antibody comprises a CDR-H3 amino acid sequence set forth in SEQ ID NO: 1006. In some embodiments, the antibody comprises a CDR-L3 amino acid sequence set forth in any one of SEQ ID NO: 1621. In some embodiments, the antibody comprises a CDR-H1, a CDR-H2, and/or a CDR-H3 from the variable heavy chain (VH) amino acid sequence of clone BP22b-l as set forth in Table 14, and/or a CDR-L1, a CDR-L2, and/or a CDR-L3 from the variable light chain (VL) amino acid sequence of clone BP22b-las set forth in Table 16. In some embodiments, the antibody comprises a variable heavy chain amino acid sequence of clone BP22b-l as set forth in Table 14. In other embodiments, the antibody comprises the variable light chain amino acid sequence of clone BP22b-l as set forth in Table 16. [0054] Additional aspects of the present disclosure relate to an antibody that specifically binds proMyostatin and/or latent Myostatin, in which the antibody has an affinity to the proMyostatin and/or latent Myostatin that is pH sensitive.
[0055] Further aspects of the present disclosure include an antibody that specifically binds proMyostatin and/or latent Myostatin with a greater affinity at a pH in a range from 6.5 to 7.5 than it does at a pH in a range from 4.6 to 6.0. In some embodiments, the antibody has a Kd of binding to pro/latent-Myostatin in a range fromlO-3 M to 10-8 M under binding conditions having a pH in a range from 4.6 to 6.0. In other embodiments, the antibody has a Kd of binding to pro/latent-Myostatin in a range fromlO-6 M to 10-11 M, when the pH is in a range from 6.5 to 7.5. In another embodiment, the antibody comprises a CDR-H3 amino acid sequence set forth in SEQ ID NO: 813, 804, 790, 762, 784, 770, 764, 986, 777, or 1006. In other embodiments, the antibody comprises a CDR-L3 amino acid sequence set forth in any one of SEQ ID NO: 1422, 1414, 1403, 1377, 1397, 1385, 1379, 1601, 1391, or 1621. In some embodiments, the antibody comprises a CDR-H1, a CDR-H2, and/or a CDR-H3 from the variable heavy chain (VH) amino acid sequence of clone BP2a-l, BP3a-2, BP5b-l, BP5b- 4, BP6a-5, BP6a-6, BPl la-1, BP16b-4, BP18b-2 or BP22b-l as set forth in Table 14, and/or a CDR-L1, a CDR-L2, and/or a CDR-L3 from the variable light chain (VL) amino acid sequence of clone BP2a-l, BP3a-2, BP5b-l, BP5b-4, BP6a-5, BP6a-6, BPl la-1, BP16b-4, BP18b-2 or BP22b-l as set forth in Table 16. In some embodiments, the antibody comprises a variable heavy chain amino acid sequence of clone BP2a-l, BP3a-2, BP5b-l, BP5b-4, BP6a-5, BP6a-6, BPl la-1, BP16b-4, BP18b-2 or BP22b-las set forth in Table 14. In other embodiments, the antibody comprises the variable light chain amino acid sequence of clone BP2a-l, BP3a-2, BP5b-l, BP5b-4, BP6a-5, BP6a-6, BPl la-1, BP16b-4, BP18b-2 or BP22b- las set forth in Table 16.
[0056] Aspects of the present disclosure include an isolated antibody or antigen binding fragment thereof having at least one variable domain with an amino acid sequence having at least 95% sequence identity to one or more of those listed in Table 14 or 16. In some embodiments, the at least one variable domain comprises a variable domain pair, the variable domain pair comprising a heavy chain variable domain (VH) and a light chain variable domain (VL), in which the variable domain pair comprises at least 95% sequence identity to a variable domain pair present in an scFv selected from any of those listed in Table 13.
[0057] Additional aspects of the present disclosure include an isolated antibody or antigen binding fragment thereof comprising at least one variable domain, the at least one variable domain comprising at least one complementarity determining region (CDR) sequence comprising at least 70% sequence identity to one or more of those listed in Table 15 or 17. In some embodiments, the at least one CDR sequence comprises at least 70% sequence identity to a heavy chain variable domain CDR (CDR-H) selected from any of those listed in Table 15. In some embodiments, the isolated antibody or antigen binding fragment thereof comprises at least one variable domain pair, the variable domain pair comprising a VH and a VL. In some embodiments, the VH comprises at least one CDR-H sequence with at least 70% sequence identity to a sequence selected from any of those listed in Table 15. In some embodiments, the VL comprises at least one CDR-L sequence with at least 70% sequence identity to a CDR-L sequence selected from any of those listed in Table 17.
[0058] Further aspects of the present disclosure include an isolated antibody or antigen binding fragment thereof that comprises: a VH sequence with at least 95% sequence identity to a sequence listed in Table 14, and a VL sequence with at least 95% sequence identity to a sequence listed in Table 16.
[0059] Further aspects of the present disclosure relate to an isolated antibody or antigen binding fragment thereof that comprises: a CDR-H1 sequence with at least 70% sequence identity to a sequence listed in Table 15, a CDR-H2 sequence with at least 70% sequence identity to a sequence listed in Table 15, a CDR-H3 sequence with at least 70% sequence identity to a sequence listed in Table 15, a CDR-L1 sequence with at least 70% sequence identity to a sequence listed in Table 17, a CDR-L2 sequence with at least 70% sequence identity to a sequence listed in Table 17, and a CDR-L3 sequence with at least 70% sequence identity to a sequence listed in Table 17. In some embodiments, the isolated antibody or antigen binding fragment thereof, comprising an antibody, in which the antibody is a monoclonal antibody. In some embodiments, the isolated antibody or antigen binding fragment thereof, comprising an antibody, in which the antibody comprises an IgG isotype. In other embodiments, the antibody comprises an IgGl isotype. In another embodiment, the antibody binds to one or more recombinant antigens listed in Table 24 and/or one or more polypeptides comprising an amino acid sequence with at least 95% sequence identity to any of those listed in Tables 1, 2, 4, 5, 9, 10, 19, 20 or 25. In a further embodiment, the polypeptide comprises pro-growth differentiation factor (GDF)-8 (proGDF-8) (SEQ ID NO: 5). In some embodiments, the antibody binds to a latent GDF-8 growth factor prodomain complex (GPC).
[0060] Further aspects of the present disclosure include a method of modulating growth factor activity in a biological system comprising contacting the biological system with the antibody or antigen binding fragment thereof. In some embodiments, the growth factor activity comprises GDF-8 activity. In other embodiments, the antibody is a stabilizing antibody and in which contacting the biological system with the stabilizing antibody results in inhibition of release of at least 5% of total GDF-8 mature growth factor in the biological system. In further embodiments, the stabilizing antibody comprises at least one variable domain comprising a VH and VL pair selected from the group consisting of the VH of SEQ ID NO: 462 and the VL of SEQ ID NO: 1037, the VH of SEQ ID NO: 472 and the VL of SEQ ID NO: 1047, the VH of SEQ ID NO: 448 and the VL of SEQ ID NO: 1023, the VH of SEQ ID NO: 502 and the VL of SEQ ID NO: 1077, the VH of SEQ ID NO: 581 and the VL of SEQ ID NO: 1153, the VH of SEQ ID NO: 442 and the VL of SEQ ID NO: 1017, the VH of SEQ ID NO: 464 and the VL of SEQ ID NO: 1039, the VH of SEQ ID NO: 593 and the VL of SEQ ID NO: 1166, the VH of SEQ ID NO: 598 and the VL of SEQ ID NO: 1171, the VH of SEQ ID NO: 626 and the VL of SEQ ID NO: 1199 and the VH of SEQ ID NO: 645 and the VL of SEQ ID NO: 1218. In some embodiments, the antibody is a releasing antibody and in which contacting the biological system with the releasing antibody results in release of at least 5% of total GDF-8 mature growth factor in the biological system.
[0061] Further aspects of the present disclosure include a method of treating a TGF-β- related indication in a subject comprising contacting the subject with the antibody or antigen binding fragment thereof of. In some embodiments, the TGF-P-related indication comprises a muscle disorder and/or injury. In other embodiments, the muscle disorder and/or injury comprises a muscle disorder selected from the group consisting of cachexia, inclusion body myositis (IBM) and sarcopenia. In another embodiment, the TGF-P-related indication comprises chronic obstructive pulmonary disease (COPD).
[0062] Further aspects of the present disclosure include a recombinant antigen comprising a growth differentiation factor (GDF) prodomain fused to an Fc-fusion protein. In some embodiments, the GDF is selected from the group consisting of GDF-8 and GDF-11. In other embodiments, the Fc-fusion protein comprises the sequence:
DKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWY VDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIE KTISKAKGQPREPQVYTLPPSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENN YKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG
K (SEQ ID NO: 222). In some embodiments, the recombinant antigen comprises a sequence selected from any of those presented in Table 12. [0063] Additional aspects of the present disclosure include a recombinant antigen comprising a chimeric protein selected from any of those in Tables 20 and 21.
[0064] Yet another aspect of the present disclosure relates to an isolated antibody or antigen binding fragment thereof that binds to a recombinant antigen described above. In some embodiments, the recombinant antigen comprises latent GDF- 11 growth factor prodomain complex (GPC).
[0065] A further aspect of the present disclosure includes a method of modulating growth factor activity in a biological system comprising contacting the biological system with the antibody or antigen binding fragment thereof described above. In some embodiments, growth factor activity comprises GDF-11 activity. In some embodiments, an isolated antibody or antigen binding fragment thereof is a stabilizing antibody and in which contacting the biological system with the stabilizing antibody results in inhibition of release of at least 5% of total GDF-11 mature growth factor in the biological system. In some embodiments, an antibody is a releasing antibody and in which contacting the biological system with the releasing antibody results in release of at least 5% of total GDF-11 mature growth factor in the biological system.
[0066] A further aspect of the present disclosure includes a method of treating a TGF-β- related indication in a subject. In some embodiments, the methods involve administering to the subject any one or more of the antibodies, including antigen binding fragments, disclosed herein. In some embodiments, the TGF-P-related indication comprises a cardiovascular indication selected from the group consisting of cardiac hypertrophy, cardiac atrophy, atherosclerosis and restenosis. In some embodiments, the TGF-P-related indication comprises a GDF-11 -related indication. In some embodiments, the GDF-11 -related indication comprises anemia and/or β-thalassemia.
[0067] In some embodiments, method of enhancing skeletal muscle rejuvenation, cerebral circulation, neurogenesis and/or erythropoiesis are provided. In some embodiments, the methods involve administering to a subject any one or more of the antibodies, including antigen binding fragments, disclosed herein. In some embodiments, the antibody is a monoclonal antibody. In some embodiments, the antibody comprises an IgG isotype. In some embodiments, the IgGl isotype. In some embodiments, the antibody is a bispecific antibody.
[0068] In some aspects, methods of increasing muscle mass in a subject are provided that comprise administering to the subject any one or more of the antibodies or antigen binding fragments disclosed herein. In some embodiments, the isolated antibody or antigen binding fragment comprises at least one variable domain comprising a VH and VL pair selected from the group consisting of the VH of SEQ ID NO: 462 and the VL of SEQ ID NO: 1037, the VH of SEQ ID NO: 472 and the VL of SEQ ID NO: 1047, the VH of SEQ ID NO: 448 and the VL of SEQ ID NO: 1023, the VH of SEQ ID NO: 502 and the VL of SEQ ID NO: 1077, the VH of SEQ ID NO: 581 and the VL of SEQ ID NO: 1153, the VH of SEQ ID NO: 442 and the VL of SEQ ID NO: 1017, the VH of SEQ ID NO: 464 and the VL of SEQ ID NO: 1039, the VH of SEQ ID NO: 593 and the VL of SEQ ID NO: 1166, the VH of SEQ ID NO: 598 and the VL of SEQ ID NO: 1171, the VH of SEQ ID NO: 626 and the VL of SEQ ID NO: 1199 and the VH of SEQ ID NO: 645 and the VL of SEQ ID NO: 1218.
[0069] Assays involving the use of one or more of the recombinant antigens and/or one or more of the antibodies or antigen binding fragments thereof disclosed herein are also provided.
[0070] In some aspects, compositions are provided that comprise an antibody (or antigen binding fragment) and an excipient or carrier. In some embodiments, the composition is a pharmaceutical composition comprising (i) a therapeutically effective amount of an antibody and (ii) a pharmaceutically acceptable carrier. In some embodiments, the composition is for use in preventing muscle wasting or increasing muscle mass, and comprises a therapeutically effective amount of an antibody. In some embodiments, the carrier is a pharmaceutically acceptable carrier. In some embodiments, the antibody and carrier are in a lyophilized form. In some embodiments, the antibody and carrier are in solution. In some embodiments, the antibody and carrier are frozen. In some embodiments, the antibody and carrier are frozen at a temperature less than or equal to -65°C.
[0071] In some embodiments, nucleic acids are provided that encode an antibody (or antigen binding fragment) or recombinant antigen. Vectors and cells comprising such nucleic acids are also provided. Kits comprising any of the compositions disclosed herein are also provided.
[0072] Methods of developing an antibody are also provided. In some embodiments, the methods comprise: selecting a protein/antigen, subjecting the protein/antigen to structural analysis, identifying at least one epitope, and developing at least one antibody against the at least one epitope. In some embodiments, the protein is subjected to prodomain convertase cleavage prior to structural analysis. In some embodiments, the prodomain convertase cleaved protein is further subjected to cleavage with an enzyme selected from the group consisting of BMP- 1, mammalian tolloid protein (mTLD), mammalian tolloid-like 1 (mTLLl) and mammalian tolloid-like 2 (mTLL2). In some embodiments, the protein comprises proGDF-8 (SEQ ID NO: 5) or proGDF-11 (SEQ ID NO: 4).
[0073] In some embodiments, an inhibitor of at least one BMP- 1/Tolloid- like proteinase (B/TP) is provided. In some embodiments, the inhibitor comprises a polypeptide. In some embodiments, the inhibitor comprises an inhibiting antibody, e.g., an anti-primed complex antibody. In some embodiments, the inhibitor prevents cleavage of a latent GPC. In some embodiments, the latent GPC is selected from the group consisting of latent GDF-8 and latent GDF-11. In some embodiments, the inhibitor prevents cleavage of a BMP/Tolloid cleavage site on the latent GPC. In some embodiments, the B/TP is selected from the group consisting of BMP- 1, mammalian tolloid protein (mTLD), mammalian tolloid-like 1 (mTLLl), and mammalian tolloid-like 2 (mTLL2).
[0074] In some aspects, antibodies are provided that bind at or within 10 amino acid residues of a proprotein convertase cleavage site of proMyostatin or latent Myostatin. In some embodiments, the proprotein convertase cleavage site comprises the amino acid sequence (SEQ ID NO: 1667 or 1668). In some aspects, antibodies are provided that bind at or within 10 amino acid residues of a proprotein convertase docking site of proMyostatin or latent Myostatin. In some embodiments, the proprotein convertase docking site comprises the amino acid sequence (SEQ ID NO: 1667 or 1668). In some aspects, antibodies are provided that bind at or within 10 amino acid residues of a tolloid protease cleavage site of proMyostatin or latent Myostatin. In some embodiments, the tolloid protease cleavage site comprises the amino acid sequence (SEQ ID NO: 1665 or 1666). In some aspects, antibodies are provided that bind at or within 10 amino acid residues of a tolloid protease docking site of proMyostatin or latent Myostatin. In some embodiments, the tolloid protease docking site comprises the amino acid sequence (SEQ ID NO: 1665 or 1666). In some embodiments, binding of the antibody to the proMyostatin or the latent Myostatin inhibits proteolytic cleavage of the proMyostatin or the latent Myostatin by a proprotein convertase or a tolloid protease.
[0075] In some embodiments, the disclosure relates to antibodies that compete for binding to an epitope with any one of the antibodies disclosed herein. In some embodiments, the competing antibodies bind to an epitope of human proMyostatin or an epitope of human latent Myostatin. In some embodiments, the competing antibodies specifically bind to an epitope of human proMyostatin or human latent Myostatin. In some embodiments, the competing antibodies bind to the epitope with an equilibrium dissociation constant (Kd) between the antibody and the epitope of less than 10"6 M or in a range of 10"11 M to 10"6 M. [0076] Aspects of the disclosure relate to sweeping antibodies or recycling antibodies. In some embodiments, the antibodies comprise an Fc portion. In some embodiments, the antibodies bind the neonatal Fc receptor FcRn. In some embodiments, the Fc portion binds the neonatal Fc receptor FcRn. In some embodiments, the antibodies bind FcRn at a pH greater than 6.0. In some embodiments, the antibodies bind FcRn at a pH in a range from 7.0 to 7.5. In some embodiments, the Kd of binding of the antibody to the FcRN is in a range from 10" 3J M to 10 8 M. In some embodiments, the Kd of binding of the antibody to the FcRN is in a range from HT -4 M to 10 -8 M. In some embodiments, the Kd of binding of the antibody to the FcRN is in a range from 10 -5 J M to 10-8 M. In some embodiments, the Kd of binding of the antibody to the FcRN is in a range from 10"6 M to 10"8 M. In some embodiments, the antibody comprises a CDR-H3 amino acid sequence set forth in SEQ ID NO: 813, 804, 790, 762, 784, 770, 764, 986, 777, or 1006. In some embodiments, the antibody comprises a CDR-L3 amino acid sequence set forth in any one of SEQ ID NO: 1422, 1414, 1403, 1377, 1397, 1385, 1379, 1601, 1391, or 1621. In some embodiments, the antibody comprises a CDR-H1, a CDR-H2, and/or a CDR-H3 from the variable heavy chain (VH) amino acid sequence of clone BP2a-l, BP3a-2, BP5b-l, BP5b-4, BP6a-5, BP6a-6, BPl la-1, BP16b-4, BP18b-2 or BP22b-l as set forth in Table 14, and/or a CDR-Ll, a CDR-L2, and/or a CDR-L3 from the variable light chain (VL) amino acid sequence of clone BP2a-l, BP3a-2, BP5b-l, BP5b-4, BP6a-5, BP6a-6, BPl la-1, BP16b-4, BP18b-2 or BP22b-l as set forth in Table 16. In some embodiments, the antibody comprises a variable heavy chain amino acid sequence of clone BP2a-l, BP3a-2, BP5b-l, BP5b-4, BP6a-5, BP6a-6, BPl la-1, BP16b-4, BP18b-2 or BP22b-las set forth in Table 14. In some embodiments, the antibody comprises a variable light chain amino acid sequence of clone BP2a-l, BP3a-2, BP5b-l, BP5b-4, BP6a-5, BP6a-6, BPl la-1, BP16b-4, BP18b-2 or BP22b-l as set forth in Table 16.
BRIEF DESCRIPTION OF THE FIGURES
[0077] The foregoing and other objects, features and advantages will be apparent from the following description of particular embodiments of the disclosure, as illustrated in the accompanying drawings. The drawings are not necessarily to scale, emphasis instead being placed upon illustrating the principles of various embodiments of the disclosure.
[0078] FIG. 1 is a diagram of the TGF-beta superfamily tree, where divergence is proportional to branch length. [0079] FIG. 2 is a schematic of one embodiment of a linear representation of a translated growth factor monomer. In such embodiments, translated growth factors may comprise secretion signal peptides, prodomains and growth factor domains. In embodiments according to embodiment depicted here, translated growth factors may also comprise a cleavage site between prodomain and growth factor regions.
[0080] FIG. 3 is a schematic of one embodiment of a GDF growth factor-prodomain complex (GPC) made up of proGDF monomers as well as an embodiment of a free growth factor dimer and free prodomains, separated after proteolytic cleavage of the GPC.
[0081] FIG. 4 is a schematic of one embodiment of a GDF GPC as well as an embodiment of a free growth factor dimer and free prodomains, separated after proteolytic cleavage.
Protein modules are indicated
[0082] FIG. 5 presents 3 tables showing the percent identity between amino acid sequences found in the TGF-β family. FIG. 5A demonstrates percent identity among pro- proteins (prodomain and growth factor). Percent identity among growth factor domains is presented in FIG. 5B while percent identity among prodomains is presented in FIG. 5C.
[0083] FIG. 6 shows structure-based alignment between TGF-β family member proteins [adapted from Shi et al (Shi, M. et al., Latent TGF-β structure and activation. Nature. 2011 Jun 15; 474(7351):343-9, the contents of which are herein incorporated by reference in their entirety)] . Cysteine residues required for interaction with LTBPs and/or GARPs are boxed. Residues mutated in Camurati-Engelmann syndrome are indicated with a star. Protease cleavage sites are indicated with an up arrow. Protein modules and secondary structural elements are indicated with solid bars. Residues underlined at the N-terminus of GDF-8 correspond to alternatively predicted signal peptide processing sites. "Chimeric module breakpoints" indicate regions where structural features are conserved and provide modules for chimeric protein construction (swapping of modules between family members) in all family members. N-terminal regions are shown in (A), internal regions are shown in (B) and C-terminal regions are shown in (C).
[0084] FIG. 7 is a schematic of an embodiment of a recombinant GPC.
[0085] FIG. 8 is a schematic of embodiments of mutant recombinant proteins of the disclosure.
[0086] FIG. 9 depicts the formation of a latent complex after furin cleavage of a pro- protein dimer. The latent complex retains the growth factor dimer until subjected to an activating cleavage [e.g. by members of the BMP- 1/Tolloid- like proteinase (B/TP) family.] [0087] FIG. 10 presents an alignment conducted between GDF-8 (myostatin), GDF-11, Inhibin A and a GDF-8 dimer. Arrows indicate cleavage sites. Regions involved in internal interactions are boxed. Solid rectangles appear above residues predicted to be involved in steric clashes in chimeric constructs. Stars denote important break points in protein modules.
[0088] FIG. 11 depicts recombinant histidine-tagged proGDF-8 (referred to in the figure as proMyostatin), separated by SDS-PAGE under reducing and non-reducing conditions, as visualized by colloidal blue staining.
[0089] FIG. 12 is a graph showing CAGA promoter-dependent luciferase activity is the presence of GDF-11 or proGDF-11 after treatment with proprotein convertase, Tolloid proteinase or a combination of proprotein convertase and Tolloid proteinase.
[0090] FIG. 13 is a stained gel showing separation of proteinase treated proGDF-11 under reducing and non-reducing conditions.
[0091] FIG. 14 presents results of a luciferase-based growth factor activity assay.
[0092] FIG. 15 is a schematic showing formation of the "primed" complex. The primed complex is an open, active conformation of GDF-8 GPCs or GDF-11 GPCs. Primed complexes are capable of receptor binding and inducing growth factor activity.
[0093] FIG. 16 is a graph showing that BP6a-5 dissociates rapidly at pH 5. The antibody blocks myostatin activation and promotes muscle growth in vivo.
[0094] FIG. 17 is a graph showing that BP6a-6 dissociates rapidly at pH 5. The antibody does not block Myostatin activation or signaling and is in the same epitope bin (by ELISA binding profile) as BP6a-5.
[0095] FIG. 18 displays eight antibodies that dissociate more rapidly at pH5 that at pH 7.4. The asterisk indicates some cross-reactivity with proGDFl 1 in ELISA and the bold indicates that the antibody inhibits GDF8 activation.
[0096] FIG. 19 shows CDR-H3 sequences of pH-sensitive antibodies. All listed antibodies have a histidine in the CDR-H3 sequence except for BP5b- 1. The sequences, from top to bottom, correspond to SEQ ID NOs: 762, 764, 770, 777, 784, 986, 1006, and 790. Note that BP18b-2 only includes one histidine in the entire variable region, while BP5b-l does not contain any histidines in the variable region. These two antibodies have the weakest pH sensitivity profiles.
[0097] FIG. 20 is a table of four antibodies for in vivo characterization of Myostatin sweeping. All antibodies will be produced both with and without the "sweeping" mutations in the constant region. Light grey indicates the antibody inhibits GDF8 activation. [0098] FIG. 21 is a graph showing that BP1 la- 1 dissociates rapidly at pH5. The antibody does not block Myostatin activation or signaling, but does bind to proMyostatin, latent myostatin, and mature myostatin with equal affinity.
[0099] FIG. 22 is a graph showing that BP5b-4 dissociates rapidly at pH5. The antibody does not block Myostatin activation or signaling; however, it is a different epitope group from BP1 la- 1 and BP6a-5.
[00100] FIG. 23 shows an example of an antibody (BP6a-3) with no pH-sensitive binding.
[00101] FIGs. 24A-24L show the different pH sensitivities of specific antibodies: BP6a-3 (FIG. 24A), BP5a-l (FIG. 24B), BP6a- l l (FIG. 24C), BP5b-4 (FIG. 24D), BPl la-1 (FIG. 24E), BP6a-6 (FIG. 24F), BP18b-2 (FIG. 24G), BP6a-2 (FIG. 24H), BP6a-5 (FIG. 241), BP14a-l (FIG. 24J), BP5b-2 (FIG. 24K), and BP18b-7 (FIG. 24L).
[00102] FIGs. 25A-25K show the different pH sensitivities of specific antibodies: BP16b-4 (FIG. 25 A), BP6b-9 (FIG. 25B), BP22b- l (FIG. 25C), BP3a-2 (FIG. 25D), BP5b- l (FIG. 25E), BP6a-4 (FIG. 25F), BP6a-7 (FIG. 25G), BPla-1 (FIG. 25H), BP2a- l (FIG. 251), BP5a- 2 (FIG. 25J), and BP6a- l (FIG. 25K).
DETAILED DESCRIPTION
[00103] Aspects of the disclosure relate to antibodies having particular binding profiles. In some embodiments, antibodies are selected for use (e.g. , in a particular assay) based on the fact that they have a known and/or desired binding profile (a selected binding profile). As used herein the term "binding profile" refers to a set of one or more parameters (e.g. , symbols, quantities, measurements, etc.) indicative of the extent to which an antibody specifically binds to one or more antigens. In some embodiments, a parameter indicative of the binding of an antibody for a target antigen is an IC50 or EC50 value. However, in some embodiments, a parameter indicative of the binding of an antibody for a target antigen is an equilibrium dissociation constant (Kd). In some embodiments, a parameter indicative of the binding is an equilibrium association constant (Ka). Other suitable parameters indicative of binding may be used in some embodiments. In some embodiments, the disclosure relates to antibodies having a selected myo statin-related binding profile, which comprises a set of one or more parameters indicative of the extent to which an antibody specifically binds to one or more antigens, at least one of which antigens is a myostatin-related protein (e.g., Myostatin, GDF11, etc.). [00104] In some embodiments, a binding profile comprises one or more parameters indicative of whether or not an antibody exhibits a threshold level of binding (e.g., specific binding) to one or more antigens. In some embodiments, a threshold level of binding is a level of binding that above (or below, depending on the parameter) a control or reference level of binding (e.g., background or non-specific binding). In some embodiments, a threshold level of binding is a level of binding that is at least 1, at least 2, at least 3, at least 4, at least 5, at least 6, at least 7 or more standard deviations greater than (or less than, depending on the assay and/or parameter) a control or reference level of binding (e.g., background or non-specific binding), as measured by an appropriate immunoassay. In some embodiments, a threshold level of binding is a level of binding that is in a range of 1 to 10 standard deviations, 2 to 10 standard deviations , or 4 to 6 standard deviations greater than (or less than, depending on the assay and/or parameter) a control or reference level of binding (e.g., background or non-specific binding), as measured by an appropriate immunoassay.
[00105] In some embodiments, a threshold level of binding is determined through an appropriate immunoassay. In some embodiments, an appropriate immunoassay assesses the binding affinity of an antibody for a target antigen. In some embodiments, an appropriate immunoassay is an enzyme linked immune- sorbent assay. In some embodiments, an appropriate immunoassay is an assay that determines a kinetic measurement (e.g., on rate, off rate) indicative of binding between an antibody and antigen. In some embodiments, an appropriate immunoassay is an assay, such as an Octet assay, that determines one or more a kinetic parameters indicative of binding between an antibody and antigen. In some embodiments, an appropriate immunoassay is a cell-based assay that determines one or more a parameters indicative of specific binding between an antibody and antigen based on cellular activity, e.g., growth factor signaling (such as SMAD signaling), cell growth, cell survival, gene expression, reporter expression, protein production, protein secretion, etc. In some embodiments, an appropriate immunoassay is an in vivo assay that determines one or more a parameters indicative of specific binding between an antibody and antigen based on cellular, tissue or other physiological activity.
[00106] In some embodiments, a binding profile comprises a set of one or more parameters indicative of the extent to which an antibody specifically binds to one or more one or more TGFP family member proteins or forms thereof, one or more portions or domains of TGFP family member proteins and/or one or more chimeras of TGFP family member proteins. In some embodiments, a binding profile comprises a set of one or more parameters indicative of the extent to which an antibody specifically binds or does not specifically bind to one or more different antigens. In some embodiments, a binding profile relates to the extent to which an antibody specifically binds or does not specifically bind to 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, or more different antigens. In some embodiments, a binding profile relates to the extent to which an antibody specifically binds or does not specifically bind to 5, 6, 7, 8, 9, 10 or 11 different antigens.
[00107] In some embodiments, a particular binding profile comprises a set of one or more parameters indicative of the extent to which an antibody specifically binds to one or more TGFP family member proteins or forms thereof. In some embodiments, a TGFP family member proteins is selected from the group consisting of AMH, ARTN, BMP10, BMP15, BMP2, BMP3, BMP4, BMP5, BMP6, BMP7, BMP8A, BMP8B, GDF1, GDF10, GDF11, GDF15, GDF2, GDF3, GDF3A, GDF5, GDF6, GDF7, GDF8, GDF9, GDNF, INHA, INHBA, INHBB, INHBC, INHBE, LEFTY 1, LEFTY2, NODAL, NRTN, PSPN, TGFpi, TGFP2, and TGFP3 protein. In some embodiments, TGFP family member proteins or forms thereof are from a vertebrate organism. In some embodiments, TGFP family member proteins or forms thereof are from a human, a monkey, a mouse or a rat. In some
embodiments, TGFP family member proteins or forms thereof are from a human or a mouse. In some embodiments, TGFP family member proteins or forms thereof are from a human. Examples of sequences of human and non-human TGFP family member proteins are shown in Tables 1, 4 and 5, provided herein. In some embodiments, TGFP family member proteins or forms thereof may include any naturally- occurring isoforms or variants of TGFP family member proteins. In some embodiments, TGFP family member proteins comprise an amino acid sequence that is at least 70%, 75%, 80%, 85%, 90%, 95%, 98%, or 99% identical to any of the amino acid sequences as set forth in any one of SEQ ID NOs: 1-37 and 70-115. In some embodiments, TGFP family member proteins comprise an amino acid sequence as set forth in any one of SEQ ID NOs: 1-37 and 70-115. In some embodiments, TGFP family member proteins consist of an amino acid sequence as set forth in any one of SEQ ID NOs: 1-37 and 70-115.
[00108] In some embodiments, a particular binding profile may also comprise one or more parameters indicative of to the extent to which antibodies specifically bind to or do not specifically bind to a portion or domain of one or more TGFP family member proteins. In some embodiments, the portion or domain of a TGFP family member protein is a prodomain, a straight jacket region, a growth factor domain, a fastener region, a furin cleavage site region, a bmp/tolloid cleavage site, an arm region, a fingers region 1, a fingers region 2, a latency loop, an alpha 1 helical region, and/or a bowtie region. Exemplary portions or domains of TGFP family member proteins are shown in Tables 2, 3, 9 and 11, provided herein. In some embodiments, portions or domains of TGFP family member proteins are portions or domains of GDF proteins. In some embodiments, portions or domains of TGFP family member proteins are portions or domains of GDF8 and/or GDF11. In some embodiments, portions or domains of TGFP family member proteins are portions or domains of Inhibin beta A. However, it should be appreciated that the portions or domains of TGFP family member proteins may be from any TGFP family member protein provided herein. In some embodiments, the portion or domain of a TGFP family member protein comprises an amino acid sequence that is at least 70%, 75%, 80%, 85%, 90%, 95%, 98%, or 99% identical to any of the amino acid sequences as set forth in any one of SEQ ID NOs: 38-65, 162-177 and 202-221. In some embodiments, portions or domains of TGFP family member proteins comprise an amino acid sequence as set forth in any one of SEQ ID NOs: 38-65, 162-177 and 202-221. In some embodiments, portions or domains of TGFP family member proteins consist of an amino acid sequence as set forth in any one of SEQ ID NOs: 38-65, 162- 177 and 202-221.
[00109] In some embodiments, forms of TGFP family member proteins refer to complexes of TGFP family member proteins. For example, forms of TGFP family member proteins may include pro-forms, latent-forms, primed-forms, or mature forms of dimeric TGFP family member proteins, such as, for example, proMyostatin, proGDFl 1, latent Myostatin, latent GDF11, primed Myostatin, and primed GDF11. In some embodiments, TGFP family member proteins form dimeric complexes. In some embodiments, TGFP family member proteins form homodimeric complexes. In some embodiments, TGFP family member proteins form heterodimeric complexes. Dimeric forms of TGFP family member proteins may include TGFP family member proteins that are full-length or TGFP family member proteins that have been cleaved (e.g., by a proprotein convertase and/or a tolloid protease). In some embodiments, forms of TGFP family member proteins are pro forms of TGFP family member proteins (e.g., proMyostatin or proGDFl 1). In some embodiments, forms of TGFP family member proteins include full-length TGFP family member proteins. For example, pro forms of TGFP family member proteins include, without limitation, proMyostatin and proGDFl 1 that have not been cleaved at a proprotein convertase cleavage site (e.g., by a proprotein convertase such as furin) or a tolloid protease cleavage site (e.g., by a tolloid protease such as BMP-1). In some embodiments, forms of TGFP family member proteins are latent forms of TGFP family member proteins (e.g., latent Myostatin or latent GDF11). In some embodiments, forms of TGFP family member proteins include TGFP family member proteins that have been cleaved (e.g., by a proprotein convertase). For example, latent forms of TGFP family member proteins include, without limitation, latent Myostatin and latent GDFl 1 that have been cleaved at a proprotein convertase cleavage site (e.g., by a proprotein convertase such as furin) but not at a tolloid protease cleavage site. In some embodiments, forms of TGFP family member proteins are primed forms of TGFP family member proteins (e.g., primed Myostatin or primed GDFl 1). In some embodiments, forms of TGFP family member proteins include TGFP family member proteins that have been cleaved (e.g., by a proprotein convertase and/or a tolloid protease). For example, primed forms of TGFP family member proteins include, without limitation, primed Myostatin and primed GDFl 1 that have been cleaved at a proprotein convertase cleavage site (e.g., by a proprotein convertase such as furin) and a tolloid protease cleavage site (e.g., by a tolloid protease such as BMP-1). In some embodiments, forms of TGFP family member proteins are mature forms of TGFP family member proteins (e.g., mature Myostatin or mature GDFl 1). In some embodiments, forms of TGFP family member proteins include TGFP family member proteins that have been cleaved (e.g., by a proprotein convertase and/or a tolloid protease) and are not in complex with one or more portions of a prodomain of a TGFP family member protein. For example, mature forms of TGFP family member proteins include, without limitation, mature Myostatin and mature GDFl 1 that have been cleaved at a proprotein convertase cleavage site (e.g., by a proprotein convertase such as furin), a tolloid protease cleavage site (e.g., by a tolloid protease such as BMP-1), and are not in complex with a prodomain of a TGFP family member protein.
[00110] In some embodiments, a particular binding profile comprises a set of one or more parameters indicative of the extent to which antibodies specifically bind to or do not specifically bind to chimeras of TGFP family member proteins. In some embodiments, chimeras of TGFP family member proteins can be used to provide information relating to particular epitopes to which any of the antibodies provided herein specifically bind or do not specifically bind. In some embodiments, chimeric TGFP family member proteins comprise at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14 or 15 portions or domains of different TGFp family member protein. For example, a chimeric TGFP family member protein may comprise a prodomain, a straight jacket region, a growth factor domain, a fastener region, a furin cleavage site region, a bmp/tolloid cleavage site, an arm region, a fingers region 1, a fingers region 2, a latency loop, an alpha 1 helical region, and/or a bowtie region of one TGFP family member protein and at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14 or 15 of a prodomain, a straight jacket region, a growth factor domain, a fastener region, a furin cleavage site region, a bmp/tolloid cleavage site, an arm region, a fingers region 1, a fingers region 2, a latency loop, an alpha 1 helical region, and/or a bowtie region from one or more different TGFP family member proteins. Exemplary chimeras of TGFP family member proteins are shown in Table 10, provided herein. In some embodiments, chimeras of TGFP family member proteins comprise an amino acid sequence that is at least 70%, 75%, 80%, 85%, 90%, 95%, 98%, or 99% identical to any of the amino acid sequences as set forth in any one of SEQ ID NOs: 178-201. In some embodiments, chimeras of TGFP family member proteins comprise an amino acid sequence as set forth in any one of SEQ ID NOs: 178-201. In some embodiments, chimeras of TGFP family member proteins consist of an amino acid sequence as set forth in any one of SEQ ID NOs: 178-201.
[00111] In some embodiments, TGFP family member proteins include Fc fusion proteins to any of the TGFP family member proteins or forms or domains or portions thereof.
Exemplary TGFP family member Fc fusion proteins are shown in Table 12, provided herein. In some embodiments, TGFP family member Fc fusion proteins comprise an amino acid sequence that is at least 70%, 75%, 80%, 85%, 90%, 95%, 98%, or 99% identical to any of the amino acid sequences as set forth in any one of SEQ ID NOs: 223-226. In some embodiments, TGFP family member Fc fusion proteins comprise an amino acid sequence as set forth in any one of SEQ ID NOs: 223-226. In some embodiments, TGFP family member Fc fusion proteins consist of an amino acid sequence as set forth in any one of SEQ ID NOs: 223-226.
[00112] In some embodiments, a particular binding profile comprises a set of one or more parameters indicative of the extent to which antibodies specifically bind to or do not specifically bind to one or more of a human proMyo statin, a murine proMyostatin, a human latent Myostatin, a human GDF-8 prodomain, a human GDF-11 ARM8 prodomain, a human proGDFl 1 ARM8, a human mature GDF-8, a human proGDFl 1, a murine latent myostatin and/or a human latent proGDFl 1 ARM8. However, in certain embodiments, parameters indicative of the extent of binding to one or more murine antigens can be removed from a binding profile. For example, in some embodiments, a particular binding profile comprises a set of one or more parameters indicative of the extent to which antibodies specifically bind to or do not specifically bind to one or more of a human proMyostatin, a human latent
Myostatin, a human GDF-8 prodomain, a human GDF-11 ARM8 prodomain, a human proGDFl 1 ARM8, a human mature GDF-8, a human proGDFl 1, and/or a human latent proGDFl 1 ARM8. In some embodiments, a particular binding profile relates to an extent to which antibodies specifically bind to or do not specifically bind to one or more of a protein that comprises an amino acid sequence that is at least 70%, 75%, 80%, 85%, 90%, 95%, 98%, or 99% identical to a human proMyostatin, a murine proMyostatin, a human latent Myostatin, a human GDF-8 prodomain, a human GDF- 11 ARM8 prodomain, a human proGDFl 1 ARM8, a human mature GDF-8, a human proGDFl 1, a murine latent myostatin and/or a human latent proGDFl 1 ARM8. In some embodiments, the human proMyostatin comprises an amino acid sequence as set forth in SEQ ID NO: 5. In some embodiments, the murine proMyostatin comprises an amino acid sequence as set forth in SEQ ID NO: 88. In some embodiments, the human latent Myostatin comprises an amino acid sequence as set forth in SEQ ID NO: 5. In some embodiments, the human GDF-8 prodomain comprises an amino acid sequence as set forth in SEQ ID NO: 38. In some embodiments, the human GDF- 11 ARM8 prodomain comprises an amino acid sequence as set forth in SEQ ID NO: 181. In some embodiments, the human proGDFl 1 ARM8 comprises an amino acid sequence as set forth in SEQ ID NO: 179. In some embodiments, the human mature GDF-8 comprises an amino acid sequence as set forth in SEQ ID NO: 42. In some embodiments, the human proGDFl 1 comprises an amino acid sequence as set forth in SEQ ID NO: 4. In some embodiments, the murine latent myostatin comprises an amino acid sequence as set forth in SEQ ID NO: 88. In some embodiments, the human latent proGDFl 1 ARM8 comprises an amino acid sequence as set forth in SEQ ID NO: 179. In some embodiments, antibodies provided herein have a binding profile as set forth in Table 33.
[00113] In some embodiments, a binding profile may comprises one or more symbols (e.g., +, -, +/-) indicative of the extent to which an antibody binds to an antigen. For example, in some embodiments, binding of an antibody to an antigen at a level detectable beyond a threshold level (e.g. , 5 standard deviations beyond a reference level) may be indicated by a "+". In some embodiments, a "-" indicates that the antibody does not bind the antigen at level detectable beyond a threshold in a particular assay (e.g. , is less than 2, 3, 4 or 5 standard deviations beyond a reference level, e.g. , an assay background level). In some embodiments, a "+/-" indicates that an antibody is at or near a threshold of binding the antigen as determined by a particular assay (e.g. , within 2 to 5, 3 to 5, or 4 to 5 standard deviations of a reference level).
[00114] An antibody that "specifically binds" to a target antigen, binds to the target antigen with greater affinity, avidity, more readily, and/or with greater duration than it binds to non- target antigens. In some embodiments, antibodies provided herein have particular binding profiles, e.g. , based on whether they specifically bind or do not specifically bind to one or more TGFP family member proteins or forms thereof, one or more portions or domains of TGFP family member proteins and/or one or more chimeras of TGFP family member proteins. In some embodiments an antibody specifically binds an antigen if binding to that antigen is detected above a background level (e.g., of a control antigen) using an in vitro binding assay (e.g., an ELISA). In some embodiments, an antibody specifically binds an antigen if binding to that antigen is detected at least one, at least 2, at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, or at least 10 standard deviations above a background level (e.g., of a control antigen) using an in vitro binding assay. In some embodiments an antibody specifically binds an antigen if binding to that antigen is detected at least one, at least 5 standard deviations above a background level (e.g., of a control antigen) using an in vitro binding assay. In some embodiments, the in vitro binding assay is an enzyme linked immunosorbent assay (ELISA). In some embodiments, the ELISA is performed as described in Example 29, provided herein. However, it should be appreciated that additional methods for determining the binding affinity of a protein to an antigen are also within the scope of this disclosure
[00115] In some embodiments, an antibody specifically binds to an antigen (e.g., proMyo statin) if it binds that antigen with a higher affinity as compared to another antigen (e.g., latent Myostatin). In some embodiments, an antibody specifically binds to an antigen if it binds to that antigen by at least 2-fold, 5-fold, 10-fold, 50-fold, 100-fold, 200-fold, 500- fold, or 1,000-fold higher than another antigen.
[00116] In some embodiments, an antibody specifically binds to an antigen (e.g., proMyostatin) if it binds that antigen with a higher affinity as compared to another antigen (e.g., latent Myostatin). In some embodiments, an antibody specifically binds to an antigen if it binds to that antigen with a dissociation constant (Kd) that is less than 10"3 M, 10"4 M, 10"5 M, 10"6M, 10"7 M, 10"8 M
Sweeping Antibodies
[00117] In some embodiments, antibodies bind an antigen but cannot effectively eliminate the antigen from the plasma. Thus, in some embodiments, the concentration of the antigen in the plasma may be increased by reducing the clearance of the antigen. However, in some embodiments, antibodies (e.g., sweeping antibodies) provided herein have an affinity to an antigen that is sensitive to pH. Such pH sensitive antibodies may bind to the antigen in plasma at neutral pH and dissociate from the antigen in an acidic endosome, thus reducing antibody-mediated antigen accumulation and/or promoting antigen clearance from the plasma. [00118] Aspects of the disclosure relate to sweeping antibodies. As used herein "sweeping antibodies" refer to antibodies having both pH-sensitive antigen binding and at least a threshold level of binding to cell surface neonatal Fc receptor (FcRn) at neutral or
physiological pH. In some embodiments, sweeping antibodies bind to the neonatal Fc receptor FcRn at neutral pH. For example sweeping antibodies may bind to the FcRn at a pH ranging from 7.0 to 7.6. In some embodiments, sweeping antibodies can bind to an antigen at an antigen binding site and bind to a cellular FcRn via an Fc portion of the antibody. In some embodiments, sweeping antibodies may then be internalized, releasing antigen in an acidic endosome, which may be degraded. In some embodiments, a sweeping antibody, no longer bound to the antigen, may then be released (e.g., by exocytosis) by the cell back into the serum.
[00119] In some embodiments, FcRn in the vascular endothelia (e.g., of a subject) extends the half-life of a sweeping antibody. In some embodiments, vascular endothelial cells internalize sweeping antibodies, which in some embodiments are bound to an antigen such as Myostatin (e.g., proMyostatin, latent Myostatin or primed Myostatin). In some
embodiments, a sweeping antibody is recycled back into the bloodstream. In some embodiments, a sweeping antibody has an increased half-life (e.g. , in the serum of a subject) as compared to its conventional counterpart. In some embodiments, a conventional counterpart of a sweeping antibody refers the antibody from which the sweeping antibody was derived (e.g., prior to engineering the Fc portion of the conventional antibody to bind FcRn with greater affinity at pH 7). In some embodiments, a sweeping antibody has a half- life in the serum of a subject that is at least 1%, 5%, 10%, 15%, 20%, 25%, 35%, 50%, 75%, 100%, 150%, 200% or 250% longer as compared to its conventional counterpart.
[00120] In some embodiments, an Fc portion of a sweeping antibody binds FcRn. In some embodiments, the Fc portion of a sweeping antibody binds to FcRn at a pH of 7.4 with a Kd
-3 -8
ranging from 10"J M to 10 ° M. In some embodiments, a sweeping antibody binds to FcRn at a pH of 7.4 with a Kd ranging fromlO"3 M to 10"7 M, from 10"3 M to 10"6 M, from 10"3 M to 10"5 M, from 10"3 M to 10"4 M, from 10"4 M to 10~8 M, from 10"4 M to 10"7 M, from 10"4 M to 10"6 M, from 10"4 M to 10"5 M, from 10"5 M to 10~8 M, from 10"5 M to 10"7 M, from 10"5 M to 10"6 M, from 10"6 M to 10"8 M, from 10"6 M to 10"7 M, or from 10"7 M to 10"8 M. In some embodiments, FcRn binds to the CH2-CH3 hinge region of a sweeping antibody. In some embodiments, FcRn binds to the same region as proteinA or protein G. In some
embodiments, FcRn binds to a different binding site from FcyRs. In some embodiments, the amino acid residues AA of a sweeping antibody Fc region are required for binding to FcRn. In some embodiments, the amino acid residues AA of a sweeping antibody Fc region affect binding to FcRn.
[00121] In some embodiments, any of the antibodies provided herein are engineered to bind FcRn with greater affinity. In some embodiments, any of the antibodies provided herein are engineered to bind FcRn with greater affinity at pH 7.4. In some embodiments, the affinity of sweeping antibodies to FcRn is increased to extend their pharmacokinetic (PK) properties as compared to their conventional counterparts. For example, in some embodiments, sweeping antibodies elicit less adverse reactions due to their efficacy at lower doses. In some embodiments, sweeping antibodies are administered less frequently. In some embodiments, transcytosis of sweeping antibodies to certain tissue types are increased. In some
embodiments, sweeping antibodies enhance efficiency of trans-placental delivery. In some embodiments, sweeping antibodies are less costly to produce.
[00122] In some embodiments, any of the antibodies provided herein are engineered to bind FcRn with lower affinity. In some embodiments, any of the antibodies provided herein are engineered to bind FcRn with lower affinity at pH 7.4. In some embodiments, the affinity of sweeping antibodies to FcRn is decreased to shorten their pharmacokinetic (PK) properties as compared to their conventional counterparts. For example, in some embodiments, sweeping antibodies are more rapidly cleared for imaging and/or radioimmunotherapy. In some embodiments, sweeping antibodies promote clearance of endogenous pathogenic antibodies as a treatment for autoimmune diseases. In some embodiments, sweeping antibodies reduce the risk of adverse pregnancy outcome, which may be caused by transplacental transport of material fetus- specific antibodies.
[00123] In some embodiments, sweeping antibodies have decreased affinity to an antigen at low pH as compared to a neutral or physiological pH (e.g., pH 7.4). In some embodiments, sweeping antibodies have a decreased affinity to an antigen at an acidic pH (e.g. a pH ranging from 5.5 to 6.5) as compared to a physiological pH (e.g., pH 7.4). It should be appreciated that any of the antibodies provided herein can be engineered to dissociate from the antigen depending on changes in pH (e.g., pH sensitive antibodies). In some embodiments, sweeping antibodies provided herein are engineered to bind antigen dependent on pH. In some embodiments,, sweeping antibodies provided herein are engineered to bind FcRn dependent on pH. In some embodiments, sweeping antibodies provided herein are internalized by endocytosis. In some embodiments, sweeping antibodies provided here are internalized by FcRn binding. In some embodiments, endocytosed sweeping antibodies release antigen in an endosome. In some embodiments, sweeping antibodies are recycled back to the cell surface. In some embodiments, sweeping antibodies remain attached to cells. In some embodiments, endocytosed sweeping antibodies are recycled back to the plasma. It should be appreciated that the Fc portion of any of the antibodies provided herein may be engineered to have different FcRn binding activity. In some embodiments, FcRn binding activity affects the clearance time of an antigen by a sweeping antibody. In some embodiments, sweeping antibodies may be long-acting or rapid-acting sweeping antibodies.
[00124] In some embodiments, converting a conventional therapeutic antibody into a sweeping antibody reduces the efficacious dose. In some embodiments, converting a conventional therapeutic antibody into a sweeping antibody reduces the efficacious dose by at least 1%, 2%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, or 99%. In some embodiments, converting a conventional therapeutic antibody into a sweeping antibody reduces the efficacious dose by at least 1.5 fold, 2 fold, 3 fold, 4 fold, 5 fold, 6 fold, 8 fold, 10 fold, 15 fold, 20 fold, 50 fold or 100 fold.
[00125] In some embodiments, selecting an appropriate dose of a sweeping antibody for therapy may be performed empirically. In some embodiments, a high dose of a sweeping antibody may saturate FcRn, resulting in antibodies which stabilize antigen in serum without being internalized. In some embodiments, a low dose of a sweeping antibody may not be therapeutically effective. In some embodiments, sweeping antibodies are administered once a day, once a week, once every two weeks, once every three weeks, once every four weeks, once every 6 weeks, once every 8 weeks, once every 10 weeks, once every 12 weeks, once every 16 weeks, once every 20 weeks, or once every 24 weeks.
[00126] In some embodiments, any of the antibodies provided herein may be modified or engineered to be sweeping antibodies. In some embodiments, any of the antibodies provided herein may be converted into a sweeping antibody using any suitable method. For example, suitable methods for making sweeping antibodies have been previously described in Igawa et ah, (2013) "Engineered Monoclonal Antibody with Novel Antigen-Sweeping Activity In Vivo," PLoS ONE 8(5): e63236; and Igawa et al., "pH-dependent antigen-binding antibodies as a novel
[00127] therapeutic modality," Biochimica et Biophysica Acta 1844 (2014) 1943-1950; the contents of each of which are hereby incorporated by reference. It should be appreciated, however, that the methods for making sweeping antibodies as provided herein are not meant to be limiting. Thus, additional methods for making sweeping antibodies are within the scope of this disclosure. Competing and Cross-Competing Antibodies
[00128] Aspects of the disclosure relate to antibodies that compete or cross-compete with any of the antibodies provided herein. The term "compete", as used herein with regard to an antibody, means that a first antibody binds to an epitope of a protein (e.g., latentMyo statin) in a manner sufficiently similar to the binding of a second antibody, such that the result of binding of the first antibody with its epitope is detectably decreased in the presence of the second antibody compared to the binding of the first antibody in the absence of the second antibody. The alternative, where the binding of the second antibody to its epitope is also detectably decreased in the presence of the first antibody, can, but need not be the case. That is, a first antibody can inhibit the binding of a second antibody to its epitope without that second antibody inhibiting the binding of the first antibody to its respective epitope.
However, where each antibody detectably inhibits the binding of the other antibody with its epitope or ligand, whether to the same, greater, or lesser extent, the antibodies are said to "cross-compete" with each other for binding of their respective epitope(s). Both competing and cross-competing antibodies are within the scope of this disclosure. Regardless of the mechanism by which such competition or cross-competition occurs (e.g., steric hindrance, conformational change, or binding to a common epitope, or portion thereof), the skilled artisan would appreciate that such competing and/or cross-competing antibodies are encompassed and can be useful for the methods and/or compositions provided herein.
[00129] Aspects of the disclosure relate to antibodies that compete or cross-compete with any of the antibodies provided herein. In some embodiments, an antibody binds at or near the same epitope as any of the antibodies provided herein. In some embodiments, an antibody binds near an epitope if it binds within 15 or fewer amino acid residues of the epitope. In some embodiments, any of the antibodies provided herein bind within 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14 or 15 amino acid residues of an epitope that is bound by any of the antibodies provided herein. For example, in some embodiments any of the antibodies provided herein bind at or near a tolloid cleavage site or at or near a tolloid docking site of a TGFP family member protein (e.g., proMyo statin or latent Myostatin). In some
embodiments, an antibody binds near a tolloid cleavage site or near a tolloid docking site if it binds within 15 or fewer amino acid residues of the tolloid cleavage site or tolloid docking site. In some embodiments, any of the antibodies provided herein bind within 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14 or 15 amino acid residues of a tolloid cleavage site or tolloid docking site. In some embodiments, an antibody binds at or near a tolloid cleavage site of GDF11. For example, an antibody may bind an amino acid sequence as set forth in SEQ ID NO: 1665. PKAPPLQQILDLHDFQGDALQPEDFLEEDEYHA (SEQ ID NO: 1665). In some embodiments, an antibody binds at or near a tolloid cleavage site of GDF8. For example, an antibody may bind an amino acid sequence as set forth in SEQ ID NO: 1666. PKAPPLRELIDQYDVQRDDSSDGSLEDDDYHAT (SEQ ID NO: 1666). In some embodiments, binding of an antibody at or near a tolloid cleavage site or at or near a tolloid docking site of a TGFP family member protein inhibits cleavage of the TGFP family member protein, for example, by a tolloid protease (e.g., BMP-1).
[00130] In other embodiments, any of the antibodies provided herein bind at or near a proprotein convertase cleavage site or at or near a proprotein convertase docking site of a TGFP family member protein (e.g., proMyo statin or latent Myostatin) In some embodiments, an antibody binds near a proprotein convertase cleavage site or near a proprotein convertase docking site if it binds within 15 or fewer amino acid residues of the proprotein convertase cleavage site or proprotein convertase docking site. In some embodiments, any of the antibodies provided herein bind within 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14 or 15 amino acid residues of a proprotein convertase cleavage site or proprotein convertase docking site. In some embodiments, an antibody binds at or near a proprotein convertase cleavage site of GDF11. For example, an antibody may bind an amino acid sequence as set forth in SEQ ID NO: 1667. GLHPFMELRVLENTKRSRRNLGLDCDEHSSESRC (SEQ ID NO: 1667). In some embodiments, an antibody binds at or near a proprotein convertase cleavage site of GDF8. For example, an antibody may bind an amino acid sequence as set forth in SEQ ID NO: 1668. GLNPFLEVKVTDTPKRSRRDFGLDCDEHSTESRC (SEQ ID NO: 1668). In some embodiments, binding of an antibody at or near a proprotein convertase cleavage site or at or near a proprotein convertase docking site of a TGFP family member protein inhibits cleavage of the TGFP family member protein, for example, by a proprotein convertase (e.g., furin).
[00131] In another embodiment, an antibody competes or cross-competes for binding to any of the antigens provided hererin (e.g., one or more TGFP family member proteins or forms thereof, one or more portions or domains of TGFP family member proteins and/or one or more chimeras of TGFP family member proteins) with an equilibrium dissociation constant, Kd, between the antibody and the protein of less than 10~6 M. In other
embodiments, an antibody competes or cross-competes for binding to any of the antigens provided herein with a Kd in a range from 10"11 M to 10"6 M. [00132] Any of the antibodies provided herein can be characterized using any suitable methods. For example, one method is to identify the epitope to which the antigen binds, or "epitope mapping." There are many suitable methods for mapping and characterizing the location of epitopes on proteins, including solving the crystal structure of an antibody- antigen complex, competition assays, gene fragment expression assays, and synthetic peptide-based assays, as described, for example, in Chapter 11 of Harlow and Lane, Using Antibodies, a Laboratory Manual, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., 1999. In an additional example, epitope mapping can be used to determine the sequence to which an antibody binds. The epitope can be a linear epitope, i.e., contained in a single stretch of amino acids, or a conformational epitope formed by a three-dimensional interaction of amino acids that may not necessarily be contained in a single stretch (primary structure linear sequence). Peptides of varying lengths (e.g., at least 4-6 amino acids long) can be isolated or synthesized (e.g., recombinantly) and used for binding assays with an antibody. In another example, the epitope to which the antibody binds can be determined in a systematic screen by using overlapping peptides derived from the target antigen sequence and determining binding by the antibody. According to the gene fragment expression assays, the open reading frame encoding the target antigen is fragmented either randomly or by specific genetic constructions and the reactivity of the expressed fragments of the antigen with the antibody to be tested is determined. The gene fragments may, for example, be produced by PCR and then
transcribed and translated into protein in vitro, in the presence of radioactive amino acids. The binding of the antibody to the radioactively labeled antigen fragments is then determined by immunoprecipitation and gel electrophoresis. Certain epitopes can also be identified by using large libraries of random peptide sequences displayed on the surface of phage particles (phage libraries). Alternatively, a defined library of overlapping peptide fragments can be tested for binding to the test antibody in binding assays. In additional examples, mutagenesis of an antigen binding domain, domain swapping experiments and alanine scanning mutagenesis can be performed to identify residues required, sufficient, and/or necessary for epitope binding. For example, domain swapping experiments can be performed using a mutant of a target antigen in which various fragments of TGFP family member proteins have been replaced (swapped) with sequences from related, but antigenically distinct proteins, such as another member of a TGFP family member protein. By assessing a binding profile of an antigen, the importance of the particular antigen fragment to antibody binding can be assessed. [00133] Alternatively, competition assays can be performed using other antibodies known to bind to the same antigen to determine whether an antibody binds to the same epitope as the other antibodies. Such competition assays would be apparent to the skilled artisan.
[00134] Any of the suitable methods, e.g., the epitope mapping methods as described herein, can be applied to determine whether any of the antibodies provided hererin binds one or more of the specific residues/segments of one or more TGFP family member proteins as described herein. Further, the interaction of an antibody with one or more of those defined residues in TGFP family member proteins can be determined by routine technology. For example, a crystal structure can be determined, and the distances between the residues in TGFP family member proteins and one or more residues in an antibody can be determined accordingly. Based on such distance, whether a specific residue in a TGFP family member protein interacts with one or more residues in an antibody can be determined. Further, suitable methods, such as competition assays and target mutagenesis assays can be applied to determine the preferential binding of a candidate antibody to a TGFP family member protein as compared to another TGFP family member protein.
Immunoassays and Related Binding Profiles
[00135] Aspects of the disclosure relate to methods for assessing a biological sample using one or more antibodies having a selected binding profile (e.g., a binding profile as outlined in Table 33). In some embodiments, antibodies are selected for use (e.g., in an immunoassay) based on the fact that they have a known and/or desired binding profile (a selected binding profile). For example, antibodies may have a selected binding profile to one or more TGFP family member proteins or forms thereof, one or more portions or domains of TGFP family member proteins and/or one or more chimeras of TGFP family member proteins. In some embodiments, antibodies with selected binding profiles may be used as research and/or diagnostic tools. In some embodiments, antibodies with selected binding profiles may be used to assess a level of a protein, a localization pattern of a protein {e.g., at the tissue, cellular, and/or sub-cellular level) and/or a ratio of a protein with respect to another protein. For example, antibodies with selected binding profiles may be used to assess the level of proMyo statin and the level of latent Myostatin, which may be used to determine a ratio of proMyo statin to latent Myostatin. In some embodiments, methods include obtaining one or more antibodies that have a selected binding profile and subjecting a biological sample to an immunoassay using the one or more antibodies with selected binding profiles. In some embodiments, at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25 or more antibodies having a selected binding profile are used in an assay to assess a biological sample. In some embodiments, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24 or 25 antibodies having a selected binding profile are used in an assay to assess a biological sample.
[00136] In some embodiments, a biological sample is subjected to an immunoassay using two or more antibodies having different selected binding profiles. In some embodiments, a biological sample is subjected to an immunoassay using 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24 or 25 antibodies having different selected binding profiles. In some embodiments, antibodies have a selected binding profile to one or more of a human proMyostatin, a human latent Myostatin, a human GDF-8 prodomain, a human GDF-11 ARM8 prodomain, a human proGDFl 1 ARM8, a human mature GDF-8, a human proGDFl 1, and/or a human latent proGDFl 1 ARM8. In some embodiments, antibodies have a selected binding profile to one or more of a human proMyostatin, a murine proMyostatin, a human latent Myostatin, a human GDF-8 prodomain, a human GDF-11 ARM8 prodomain, a human proGDFl 1 ARM8, a human mature GDF-8, a human proGDFl 1, a murine latent Myostatin and/or a human latent proGDFl 1 ARM8. In some embodiments, In some embodiments, the human proMyostatin comprises an amino acid sequence as set forth in SEQ ID NO: 5. In some embodiments, the murine proMyostatin comprises an amino acid sequence as set forth in SEQ ID NO: 88. In some embodiments, the human latent Myostatin comprises an amino acid sequence as set forth in SEQ ID NO: 5. In some embodiments, the human GDF-8 prodomain comprises an amino acid sequence as set forth in SEQ ID NO: 38. In some embodiments, the human GDF- 11 ARM8 prodomain comprises an amino acid sequence as set forth in SEQ ID NO: 181. In some embodiments, the human proGDFl 1 ARM8 comprises an amino acid sequence as set forth in SEQ ID NO: 179. In some embodiments, the human mature GDF-8 comprises an amino acid sequence as set forth in SEQ ID NO: 42. In some embodiments, the human proGDFl 1 comprises an amino acid sequence as set forth in SEQ ID NO: 4. In some embodiments, the murine latent Myostatin comprises an amino acid sequence as set forth in SEQ ID NO: 88. In some embodiments, the human latent proGDFl 1 ARM8 comprises an amino acid sequence as set forth in SEQ ID NO: 179.
[00137] In some embodiments, a biological sample is subjected to an immunoassay using two or more antibodies having different selected binding profiles. In some embodiments, antibodies have selected binding profiles as shown in Table 33. In some embodiments, antibodies have selected binding profiles of any one of the antibodies Bla-1, BPlb-1, BP2a- 1, BP3a-l, BP3a-2, BP3a-3, BP4a-l, BP5a-l, BP5b-l, BP5a-2, BP5a-3, BP5a-4, BP5a-5, BP5b-2, BP5b-3, BP5b-4, BP5b-5, BP5b-6, BP5b-7, BP5b-8, BP5b-9, BP6a-l, BP6a-2, BP6a-3, BP6a-4, BP6a-5, BP6a-6, BP6b-l, BP6b-2, BP6a-7, BP6a-8, BP6a-9, BP6a-10, BP6a-l l, BP6a-12, BP6a-13, BP6a-14, BP6a-15, BP6a-16, BP6a-17, BP6a-18, BP6a-19, BP6a-20, BP6a-21, BP6a-22, BP6a-23, BP6a-24, BP6a-25, BP6a-26, BP6a-27, BP6b-3, BP6b-4, BP6b-5, BP6b-6, BP6b-7, BP6b-8, BP6b-9, BP6b-10, BP6b-l l, BP6b-12, BP7b-l, BP8b-l, BP9b-l, BPlOb-1, BPl la-1, BP12b-l, BP13a-l, BP14a-l, BP15a-l, BP16b-l, BP16b-2, BP16b-3, BP16b-4, BP16a-l, BP17b-l, BP18b-l, BP18b-2, BP18b-3, BP18b-4, BP18b-5, BP18b-6, BP18b-7, BP18a-l, BP18a-2, BP19b-l, BP19b-2, BP19b-3, BP19a-l, BP19a-2, BP19a-3, BP20b-l, BP21b-l or BP22b-l.
[00138] In some embodiments, a biological sample is subjected to one or more
immunoassays using a set of two or more (e.g., 2, 3, 4, 5, 6, 7, 8, 9, 10 or more) antibodies, in which the set comprises two antibodies having binding profiles, as outlined in Table 33, selected from the following pairs: {BPla, BPlb}, {BPla, BP2a}, {BPla, BP3a}, {BPla, BP4a}, {BPla, BP5a}, {BPla, BP5b}, {BPla, BP6a}, {BPla, BP6b}, {BPla, BP7b}, {BPla, BP8b}, {BPla, BP9b}, {BPla, BPlOb}, {BPla, BPl la}, {BPla, BP12b}, {BPla, BP13a}, {BPla, BP14a}, {BPla, BP15a}, {BPla, BP16b}, {BPla, BP16a}, {BPla, BP17b}, {BPla, BP18b}, {BPla, BP18a}, {BPla, BP19b}, {BPla, BP19a}, {BPla, BP20b}, {BPla, BP21b}, {BPla, BP22b}, {BPlb, BP2a}, {BPlb, BP3a}, {BPlb, BP4a}, {BPlb, BP5a}, {BPlb, BP5b}, {BPlb, BP6a}, {BPlb, BP6b}, {BPlb, BP7b}, {BPlb, BP8b}, {BPlb, BP9b}, {BPlb, BPlOb}, {BPlb, BPl la}, {BPlb, BP12b}, {BPlb, BP13a}, {BPlb, BP14a}, {BPlb, BP15a}, {BPlb, BP16b}, {BPlb, BP16a}, {BPlb, BP17b}, {BPlb, BP18b}, {BPlb, BP18a}, {BPlb, BP19b}, {BPlb, BP19a}, {BPlb, BP20b}, {BPlb, BP21b}, {BPlb, BP22b}, {BP2a, BP3a}, {BP2a, BP4a}, {BP2a, BP5a}, {BP2a, BP5b}, {BP2a, BP6a}, {BP2a, BP6b}, {BP2a, BP7b}, {BP2a, BP8b}, {BP2a, BP9b}, {BP2a, BPlOb}, {BP2a, BPl la}, {BP2a, BP12b}, {BP2a, BP13a}, {BP2a, BP14a}, {BP2a, BP15a}, {BP2a, BP16b}, {BP2a, BP16a}, {BP2a, BP17b}, {BP2a, BP18b}, {BP2a, BP18a}, {BP2a, BP19b}, {BP2a, BP19a}, {BP2a, BP20b}, {BP2a, BP21b}, {BP2a, BP22b}, {BP3a, BP4a}, {BP3a, BP5a}, {BP3a, BP5b}, {BP3a, BP6a}, {BP3a, BP6b}, {BP3a, BP7b}, {BP3a, BP8b}, {BP3a, BP9b}, {BP3a, BPlOb}, {BP3a, BPl la}, {BP3a, BP12b}, {BP3a, BP13a}, {BP3a, BP14a}, {BP3a, BP15a}, {BP3a, BP16b}, {BP3a, BP16a}, {BP3a, BP17b}, {BP3a, BP18b}, {BP3a, BP18a}, {BP3a, BP19b}, {BP3a, BP19a}, {BP3a, BP20b}, {BP3a, BP21b}, {BP3a, BP22b}, {BP4a, BP5a}, {BP4a, BP5b}, {BP4a, BP6a}, {BP4a, BP6b}, {BP4a, BP7b}, {BP4a, BP8b}, {BP4a, BP9b}, {BP4a, BPlOb}, {BP4a, BPl la}, {BP4a, BP12b}, {BP4a, BP13a}, {BP4a, BP14a}, {BP4a, BP15a}, {BP4a, BP16b}, {BP4a, BP16a}, {BP4a, BP17b}, {BP4a, BP18b}, {BP4a, BP18a}, {BP4a, BP19b}, {BP4a, BP19a}, {BP4a, BP20b}, {BP4a, BP21b}, {BP4a, BP22b}, {BP5a, BP5b}, {BP5a, BP6a}, {BP5a, BP6b}, {BP5a, BP7b}, {BP5a, BP8b}, {BP5a, BP9b}, {BP5a, BPlOb}, {BP5a, BPl la}, {BP5a, BP12b}, {BP5a, BP13a}, {BP5a, BP14a}, {BP5a, BP15a}, {BP5a, BP16b}, {BP5a, BP16a}, {BP5a, BP17b}, {BP5a, BP18b}, {BP5a, BP18a}, {BP5a, BP19b}, {BP5a, BP19a}, {BP5a, BP20b}, {BP5a, BP21b}, {BP5a, BP22b}, {BP5b, BP6a}, {BP5b, BP6b}, {BP5b, BP7b}, {BP5b, BP8b}, {BP5b, BP9b}, {BP5b, BPlOb}, {BP5b, BPl la}, {BP5b, BP12b}, {BP5b, BP13a}, {BP5b, BP14a}, {BP5b, BP15a}, {BP5b, BP16b}, {BP5b, BP16a}, {BP5b, BP17b}, {BP5b, BP18b}, {BP5b, BP18a}, {BP5b, BP19b}, {BP5b, BP19a}, {BP5b, BP20b}, {BP5b, BP21b}, {BP5b, BP22b}, {BP6a, BP6b}, {BP6a, BP7b}, {BP6a, BP8b}, {BP6a, BP9b}, {BP6a, BPlOb}, {BP6a, BPl la}, {BP6a, BP12b}, {BP6a, BP13a}, {BP6a, BP14a}, {BP6a, BP15a}, {BP6a, BP16b}, {BP6a, BP16a}, {BP6a, BP17b}, {BP6a, BP18b}, {BP6a, BP18a}, {BP6a, BP19b}, {BP6a, BP19a}, {BP6a, BP20b}, {BP6a, BP21b}, {BP6a, BP22b}, {BP6b, BP7b}, {BP6b, BP8b}, {BP6b, BP9b}, {BP6b, BPlOb}, {BP6b, BPl la}, {BP6b, BP12b}, {BP6b, BP13a}, {BP6b, BP14a}, {BP6b, BP15a}, {BP6b, BP16b}, {BP6b, BP16a}, {BP6b, BP17b}, {BP6b, BP18b}, {BP6b, BP18a}, {BP6b, BP19b}, {BP6b, BP19a}, {BP6b, BP20b}, {BP6b, BP21b}, {BP6b, BP22b}, {BP7b, BP8b}, {BP7b, BP9b}, {BP7b, BPlOb}, {BP7b, BPl la}, {BP7b, BP12b}, {BP7b, BP13a}, {BP7b, BP14a}, {BP7b, BP15a}, {BP7b, BP16b}, {BP7b, BP16a}, {BP7b, BP17b}, {BP7b, BP18b}, {BP7b, BP18a}, {BP7b, BP19b}, {BP7b, BP19a}, {BP7b, BP20b}, {BP7b, BP21b}, {BP7b, BP22b}, {BP8b, BP9b}, {BP8b, BPlOb}, {BP8b, BPl la}, {BP8b, BP12b}, {BP8b, BP13a}, {BP8b, BP14a}, {BP8b, BP15a}, {BP8b, BP16b}, {BP8b, BP16a}, {BP8b, BP17b}, {BP8b, BP18b}, {BP8b, BP18a}, {BP8b, BP19b}, {BP8b, BP19a}, {BP8b, BP20b}, {BP8b, BP21b}, {BP8b, BP22b}, {BP9b, BPlOb}, {BP9b, BPl la}, {BP9b, BP12b}, {BP9b, BP13a}, {BP9b, BP14a}, {BP9b, BP15a}, {BP9b, BP16b}, {BP9b, BP16a}, {BP9b, BP17b}, {BP9b, BP18b}, {BP9b, BP18a}, {BP9b, BP19b}, {BP9b, BP19a}, {BP9b, BP20b}, {BP9b, BP21b}, {BP9b, BP22b}, {BPlOb, BPl la}, {BPlOb, BP12b}, {BPlOb, BP13a}, {BPlOb, BP14a}, {BPlOb, BP15a}, {BPlOb, BP16b}, {BPlOb, BP16a}, {BPlOb, BP17b}, {BPlOb, BP18b}, {BPlOb, BP18a}, {BPlOb, BP19b}, {BPlOb, BP19a}, {BPlOb, BP20b}, {BPlOb, BP21b}, {BPlOb, BP22b}, {BPl la, BP12b}, {BPl la, BP13a}, {BPl la, BP14a}, {BPl la, BP15a}, {BPl la, BP16b}, {BPl la, BP16a}, {BPl la, BP17b}, {BPl la, BP18b}, {BPl la, BP18a}, {BPl la, BP19b}, {BPl la, BP19a}, {BPl la, BP20b}, {BPl la, BP21b}, {BPl la, BP22b}, {BP12b, BP13a}, {BP12b, BP14a}, {BP12b, BP15a}, {BP12b, BP16b}, {BP12b, BP16a}, {BP12b, BP17b}, {BP12b, BP18b}, {BP12b, BP18a}, {BP12b, BP19b}, {BP12b, BP19a}, {BP12b, BP20b}, {BP12b, BP21b}, {BP12b, BP22b}, {BP13a, BP14a}, {BP13a, BP15a}, {BP13a, BP16b}, {BP13a, BP16a}, {BP13a, BP17b}, {BP13a, BP18b}, {BP13a, BP18a}, {BP13a, BP19b}, {BP13a, BP19a}, {BP13a, BP20b}, {BP13a, BP21b}, {BP13a, BP22b}, {BP14a, BP15a}, {BP14a, BP16b}, {BP14a, BP16a}, {BP14a, BP17b}, {BP14a, BP18b}, {BP14a, BP18a}, {BP14a, BP19b}, {BP14a, BP19a}, {BP14a, BP20b}, {BP14a, BP21b}, {BP14a, BP22b}, {BP15a, BP16b}, {BP15a, BP16a}, {BP15a, BP17b}, {BP15a, BP18b}, {BP15a, BP18a}, {BP15a, BP19b}, {BP15a, BP19a}, {BP15a, BP20b}, {BP15a, BP21b}, {BP15a, BP22b}, {BP16b, BP16a}, {BP16b, BP17b}, {BP16b, BP18b}, {BP16b, BP18a}, {BP16b, BP19b}, {BP16b, BP19a}, {BP16b, BP20b}, {BP16b, BP21b}, {BP16b, BP22b}, {BP16a, BP17b}, {BP16a, BP18b}, {BP16a, BP18a}, {BP16a, BP19b}, {BP16a, BP19a}, {BP16a, BP20b}, {BP16a, BP21b}, {BP16a, BP22b}, {BP17b, BP18b}, {BP17b, BP18a}, {BP17b, BP19b}, {BP17b, BP19a}, {BP17b, BP20b}, {BP17b, BP21b}, {BP17b, BP22b}, {BP18b, BP18a}, {BP18b, BP19b}, {BP18b, BP19a}, {BP18b, BP20b}, {BP18b, BP21b}, {BP18b, BP22b}, {BP18a, BP19b}, {BP18a, BP19a}, {BP18a, BP20b}, {BP18a, BP21b}, {BP18a, BP22b}, {BP19b, BP19a}, {BP19b, BP20b}, {BP19b, BP21b}, {BP19b, BP22b}, {BP19a, BP20b}, {BP 19a, BP21b}, {BP 19a, BP22b}, {BP20b, BP21b}, {BP20b, BP22b}, and {BP21b, BP22b}. In some embodiments, a biological sample is subjected to an
immunoassay using a set of one or more antibodies comprising any two of the antibodies of Table 33. In some embodiments, a biological sample is subjected to an immunoassay using a set of one or more antibodies comprising of any three, four , five, six, seven, eight, nine, ten or more of the antibodies of Table 33.
[00139] In some embodiments, a biological sample is subjected to an immunoassay using three or more antibodies having different selected binding profiles. In some embodiments, antibodies have selected binding profiles as shown in Table 33. In some embodiments, antibodies have selected binding profiles of any one of the antibodies Bla-1, BPlb-1, BP2a- 1, BP3a-l, BP3a-2, BP3a-3, BP4a-l, BP5a-l, BP5b-l, BP5a-2, BP5a-3, BP5a-4, BP5a-5, BP5b-2, BP5b-3, BP5b-4, BP5b-5, BP5b-6, BP5b-7, BP5b-8, BP5b-9, BP6a-l, BP6a-2, BP6a-3, BP6a-4, BP6a-5, BP6a-6, BP6b-l, BP6b-2, BP6a-7, BP6a-8, BP6a-9, BP6a-10, BP6a-l l, BP6a-12, BP6a-13, BP6a-14, BP6a-15, BP6a-16, BP6a-17, BP6a-18, BP6a-19, BP6a-20, BP6a-21, BP6a-22, BP6a-23, BP6a-24, BP6a-25, BP6a-26, BP6a-27, BP6b-3, BP6b-4, BP6b-5, BP6b-6, BP6b-7, BP6b-8, BP6b-9, BP6b-10, BP6b-l l, BP6b-12, BP7b-l, BP8b-l, BP9b-l, BPlOb-1, BPl la-1, BP12b-l, BP13a-l, BP14a-l, BP15a-l, BP16b-l, BP16b-2, BP16b-3, BP16b-4, BP16a-l, BP17b-l, BP18b-l, BP18b-2, BP18b-3, BP18b-4, BP18b-5, BP18b-6, BP18b-7, BP18a-l, BP18a-2, BP19b-l, BP19b-2, BP19b-3, BP19a-l, BP19a-2, BP19a-3, BP20b-l, BP21b-l or BP22b-l. In some embodiments, methods provided herein relate to methods for detecting one or more TGFP family member proteins or forms thereof, one or more portions or domains of TGFP family member proteins and/or one or more chimeras of TGFP family member proteins in a sample obtained from a subject. In some embodiments, a subject refers to an individual organism, for example, an individual mammal. In some embodiments, the subject is a human. In some embodiments, the subject is a non-human mammal. In some embodiments, the subject is a non-human primate. In some embodiments, the subject is a rodent (e.g. , a mouse or rat). In some embodiments, the subject is a sheep, a goat, a cattle, a cat, or a dog. In some embodiments, the subject is a vertebrate, an amphibian, a reptile, a fish, an insect, a fly, or a nematode. In some
embodiments, the subject is a research animal. In some embodiments, the subject is genetically engineered, e.g. , a genetically engineered non-human subject. The subject may be of either sex and at any stage of development. In some embodiments, the subject is a patient or a healthy volunteer.
[00140] In some embodiments, a method for detecting one or more TGFP family member proteins or forms thereof, one or more portions or domains of TGFP family member proteins and/or one or more chimeras of TGFP family member proteins in a sample obtained from a subject involves (a) contacting the sample with an antibody having a selected binding profile under conditions suitable for binding of the antibody to the antigen, if the antigen is present in the sample, thereby forming binding complexes; and (b) determining the level of the antibody or antigen binding fragment bound to the antigen (e.g. , determining the level of the binding complexes).
[00141] As used herein a binding complex refers to a biomolecular complex of antibody bound to antigen (e.g. , one or more TGFP family member proteins or forms thereof, one or more portions or domains of TGFP family member proteins and/or one or more chimeras of TGFP family member proteins). Binding complexes may comprise antibodies with a single specificity or two or more antibodies or antigen binding fragments with different specificities. In one embodiment, a binding complex comprises two or more antibodies recognizing different antigenic sites on the same antigen. In some instances, an antibody may be bound to an antigen, having bound to it other biomolecules such as RNA, DNA, polysaccharides or proteins. In one embodiment, a binding complex comprises two or more antibodies recognizing different antigens. In some embodiments, an antibody in a binding complex (e.g. , an immobilized antibody bound to antigen), may itself by bound, as an antigen, to an antibody (e.g. , a detectably labeled antibody). Thus, binding complexes may, in some instances, comprise multiple antigens and multiple antibodies or antigen binding fragments.
[00142] Antigens present in binding complexes may or may not be in their native in situ conformation. In some embodiments, a binding complex is formed between an antibody and a purified protein antigen, or isolated proteins comprising antigen, in which the antigen is not in its native in situ conformation. In some embodiments, a binding complex is formed between an antibody and a purified protein antigen, in which the antigen is not in its native in situ conformation and is immobilized on solid support (e.g. , a PVDF membrane). In some embodiments, a binding complex is formed with an antibody and, for example, a cell surface protein that is present in situ in a native confirmation (e.g. , on the surface of a cell).
[00143] Antibodies in binding complexes may or may not be detectably labeled. In some embodiments, binding complexes comprise detectably labeled antibodies and non-labeled antibodies. In some embodiments, binding complexes comprise detectably labeled antigen. In some embodiments, antibodies, in binding complexes, are immobilized to one or more solid supports. In some embodiments, antigens, in binding complexes, are immobilized to one or more solid supports. Exemplary solid supports are disclosed herein and will be apparent to one of ordinary skill in the art. The foregoing examples of binding complexes are not intended to be limiting. Other examples of binding complexes will be apparent to one or ordinary skill in the art.
[00144] In any of the detection, diagnostic, and monitoring methods, antibody or antigen may be conjugated to a solid support surface, either directly or indirectly. Methods for conjugation to solid supports are standard and can be accomplished via covalent and non- covalent interactions. Non-limiting examples of conjugation methods include: adsorption, cross-linking, protein A/G - antibody interactions, and streptavidin-biotin interactions. Other methods of conjugation will be readily apparent to one of ordinary skill in the art.
[00145] In some aspects, detection, diagnosis, and monitoring methods include comparing the level of the antibody (including antigen binding fragments) bound to the antigen (e.g. , one or more TGFP family member proteins or forms thereof, one or more portions or domains of TGFP family member proteins and/or one or more chimeras of TGFP family member proteins) to one or more reference standards. The reference standard may be, for example, the level of a corresponding TGFP family member protein in a subject that does or does not have the TGFP family member protein. In one embodiment, the reference standard is the level of a TGFP family member protein detected in a sample that does not contain the TGFP family member protein (e.g. , a background level). Alternatively, a background level can be determined from a sample that contains a particular pro/latent-Myostatin, by contacting the sample with non-specific antibodies (e.g. , antibodies obtained from non-immune serum). Then again, the reference standard may be the level of a TGFP family member protein detected in a sample that does contain the TGFP family member protein (e.g. , a positive control). In some cases, the reference standard may be a series of levels associated with varying concentrations of a TGFP family member protein in a sample and useful for quantifying the concentration of the TGFP family member protein in the test sample. The foregoing examples of reference standards are not limiting and other suitable reference standard will be readily apparent to one of ordinary skill in the art.
[00146] The level of one or more TGFP family member proteins or forms thereof, one or more portions or domains of TGFP family member proteins and/or one or more chimeras of TGFP family member proteins may be measured, as provided herein, from a biological sample using any of the antibodies having a selected binding profile. A biological sample refers to any biological material which may be obtained from a subject or cell. For example, a biological sample may be whole blood, plasma, serum, saliva, cerebrospinal fluid, urine, cells (or cell lysate) or tissue (e.g. , normal tissue or diseased tissue). In some embodiments, a biological sample is a fluid sample. In some embodiments, a biological sample is a solid tissue sample. For example, a tissue sample may include, without limitation skeletal muscle, cardiac muscle, adipose tissue as well as tissue from other organs. In some embodiments, a biological sample is a biopsy sample. In some embodiments, a solid tissue sample may be made into a fluid sample using routine methods in the art.
[00147] A biological sample may also include one or more cells of a cell line. In some embodiments, a cell line includes human cells, primate cells (e.g. , vero cells), rat cells (e.g. , GH3 cells, OC23 cells) or mouse cells (e.g. , MC3T3 cells). There are a variety of human cell lines, including, without limitation, human embryonic kidney (HEK) cells, HeLa cells, cancer cells from the National Cancer Institute's 60 cancer cell lines (NCI60), DU145 (prostate cancer) cells, Lncap (prostate cancer) cells, MCF-7 (breast cancer) cells, MDA-MB-438 (breast cancer) cells, PC3 (prostate cancer) cells, T47D (breast cancer) cells, THP- 1 (acute myeloid leukemia) cells, U87 (glioblastoma) cells, SHSY5Y human neuroblastoma cells (cloned from a myeloma) and Saos-2 (bone cancer) cells.
[00148] A further embodiment relates to a method for monitoring a disease, a condition, or any treatment thereof (e.g., myopathy or myopathy treatment) in a subject having, or at risk of having, the disease or condition comprising: (a) obtaining a biological sample from the subject, (b) determining the level of one or more TGFP family member proteins or forms thereof, one or more portions or domains of TGFP family member proteins and/or one or more chimeras of TGFP family member proteins in the biological sample using an antibody having a selected binding profile and (c) repeating steps (a) and (b) on one or more occasions.
[00149] Biomarkers such as Myostatin may be used as for assessing disease conditions, however, available commercial reagents (e.g., antibodies for ELISAs and Western Blots) are often either not specific for Myostatin, detect only mature Myostatin or do not detect
Myostatin at all. Thus, provided herein are methods and reagents (e.g., antibodies with selected binding profiles) for detecting one or more TGFP family member proteins or forms thereof, in the context of diseases and/or conditions (e.g., muscle atrophy) for diagnostic purposes. As one example, the level of proMyostatin or latent Myostatin may be measured in a subject, or biological sample therefrom, to detect or monitor the progression of a disease or condition. As another example, the level of proMyostatin or latent Myostatin may be measured in a subject, or biological sample therefrom, to monitor the response to a treatment for a disease or condition. It should be appreciated that the level of proMyostatin or latent Myostatin may be monitored over any suitable period of time, which may differ depending on the disease or condition, the subject has or any treatment regimen that the subject may be subject to.
[00150] Another embodiment relates to a diagnostic composition comprising any one of the above described antibodies, antigen binding fragments, polynucleotides, vectors or cells and optionally suitable means for detection. The antibodies are, for example, suited for use in immunoassays in which they can be utilized in liquid phase or bound to a solid phase carrier. Examples of immunoassays which can utilize the antibody are competitive and noncompetitive immunoassays in either a direct or indirect format. Examples of such
immunoassays are the Enzyme Linked Immunoassay (ELISA), radioimmunoassay (RIA), the sandwich (immunometric assay), flow cytometry, the western blot assay,
immunoprecipitation assays, immunohistochemistry, immuno-microscopy, lateral flow immuno-chromatographic assays, and proteomics arrays. The antigens and antibodies can be bound to many different solid supports (e.g. , carriers, membrane, columns, proteomics array, etc.). Examples of solid support materials include glass, polystyrene, polyvinyl chloride, polyvinylidene difluoride, polypropylene, polyethylene, polycarbonate, dextran, nylon, amyloses, natural and modified celluloses, such as nitrocellulose, polyacrylamides, agaroses, and magnetite. The nature of the support can be either fixed or suspended in a solution (e.g. , beads). [00151] By a further embodiment, antibodies (including antigen binding fragments) provided herein may also be used in a method for evaluating expression of one or more TGFP family member proteins or forms thereof in a subject by obtaining a biological sample from the subject which may be a tissue sample, a blood sample or any other appropriate body fluid sample. The procedure may comprise contacting the blood sample (whole blood, serum, plasma), a tissue sample, or protein sample isolated therefrom, with an antibody having a selected binding profile, under conditions enabling the formation of binding complexes between antibody and antigen. The level of such binding complexes may then be determined by any suitable method. In some embodiments, the biological sample is contacted with the antibody under conditions suitable for binding of the antibody to one or more TGFP family member proteins or forms thereof, if the antigen is present in the sample, and formation of binding complexes consisting of antibody, bound to the antigen. In some embodiments, this contacting step is performed in a reaction chamber, such as a tube, plate well, membrane bath, cell culture dish, microscope slide, and the like. In some embodiments, an antibody having a selected binding profile is immobilized on a solid support. In some embodiments, the antigen is immobilized on a solid support. In some embodiments, the solid support is the surface of a the reaction chamber. In some embodiments, the solid support is of a polymeric membrane (e.g. , nitrocellulose strip, Polyvinylidene Difluoride (PVDF) membrane, etc.). Other appropriate solid supports may be used.
[00152] In some embodiments, an antibody having a selected binding profile is
immobilized on the solid support prior to contacting with the antigen. In other embodiments, immobilization of the antibody is performed after formation of binding complexes. In still other embodiments, antigen is immobilized on a solid support prior to formation of binding complexes. A detection reagent is added to the reaction chamber to detect immobilized binding complexes. In some embodiments, the detection reagent comprises a detectably labeled secondary antibody directed against the antigen. In some embodiments, the primary antibody is itself detectable labeled, and is thereby the detection reagent.
[00153] In one aspect, detection methods comprise the steps of immobilizing antibodies to a solid support; applying a sample (e.g. , a biological sample or isolated protein sample) to the solid support under conditions that permit binding of antigen to the antibodies, if present in the sample; removing the excess sample from the solid support; applying detectably labeled antibodies under conditions that permit binding of the detectably labeled antibodies to the antigen -bound immobilized antibodies; washing the solid support and assaying for the presence of label on the solid support. [00154] In some embodiments, the antigen is immobilized on the solid support, such as a PVDF membrane, prior to contacting with the antibody in a reaction chamber (e.g. , a membrane bath). A detection reagent is added to the reaction chamber to detect immobilized binding complexes. In some embodiments, the detection reagent comprises a detectably labeled secondary antibody directed against the antigen. In some embodiments, the detection reagent comprises a detectably labeled secondary antibody directed against the primary antibody. As disclosed herein, the detectable label may be, for example, a radioisotope, a fluorophore, a luminescent molecule, an enzyme, a biotin-moiety, an epitope tag, or a dye molecule. In some embodiments, the primary antibody is itself detectable labeled, and is thereby the detection reagent. Suitable detectable labels are described herein, and will be readily apparent to one of ordinary skill in the art.
[00155] Accordingly, diagnostic kits, suitable for home or clinical use (point of care service), are provided that comprise (a) detectably labeled and/or non-labeled antibodies having a selected binding profile, as antigen binding reagents (e.g. , reagents that bind one or more TGFP family member proteins or forms thereof); (b) a detection reagent; and, optionally, (c) complete instructions for using the reagents to detect antigens in a sample. In some embodiments, the diagnostic kit includes the antibody having a selected binding profile, and/or one or more TGFP family member proteins or forms thereof immobilized on a solid support. Any of the solid supports described herein are suitable for incorporation in the diagnostic kits. In a preferred embodiment, the solid support is the surface of a reaction chamber of a plate well. Typically, the plate well is in a multi-well plate having a number of wells selected from: 6, 12, 24, 96, 384, and 1536, but it is not so limited. In other
embodiments, the diagnostic kits provide a detectably labeled antibody. Diagnostic kits are not limited to these embodiments and other variations in kit composition will be readily apparent to one of ordinary skill in the art.
[00156] In some embodiments, Myostatin protein is present in muscle tissue. In some embodiments, Myostatin protein is present in circulation. In some embodiments, Myostatin protein is present in muscle tissue and in circulation. In some embodiments, Myostatin expression is altered in diseases or conditions involving muscle atrophy. Exemplary diseases or conditions involving muscle atrophy include, without limitation, muscular dystrophy, cachexia, sarcopenia, spinal chord injury and atrophy associated with muscle disuse. Thus, Myostatin in tissues or circulating Myostatin, can be a biomarker for muscular atrophic disease. Without wishing to be bound by any particular theory, development of Myostatin as biomarker has been hampered by the paucity of available antibodies to Myostatin, which, because of the high homology between mature Myostatin growth factor and other members of the TGFP family, are not highly selective for Myostatin. Furthermore, many Myostatin antibodies cannot distinguish between the pro-, latent, and mature forms of Myostatin. Thus, detecting the location and expression levels of various Myostatin forms (e.g., proMyostatin, latent Myostatin, primed Myostatin, and mature Myostatin) may provide significant insight into Myostatin biology in normal and disease states. In addition to their therapeutic potential, antibodies to pro-, latent, and mature Myostatin may be useful in biomarker assays for muscle atrophy. Thus, in some embodiments, antibodies provided herein can be incorporated into assays for MSD, ELISA, RIA, quantitative western blotting (capillary western, Odyssey or similar technology), immunohistochemistry, and any other technology requiring specific binding of antibodies. In some embodiments, assays (e.g., any of the immunoassays provided herein) are be applied to analysis of Myostatin levels and isoform distribution in muscle or other tissue, blood and blood derivatives (e.g., serum, plasma, exosome-like vesicles (ELVs) from blood), and urine and derivatives (e.g., urinary ELVs).
[00157] In some embodiments, any of the antibodies provided herein can be used in an immunohistochemistry (IHC) assay. In some embodiments, an immunohistochemistry assay is performed on a tissue sample (e.g., a muscle biopsy) to identify a level or localization of one or more TGFP family member proteins or forms thereof (e.g., proMyostatin, latent Myostatin , primed Myostatin, and/or mature Myostatin). In some embodiments, an IHC assay begins with antigen retrieval, which may vary in terms of reagents and methods. The antigen retrieval process may involve pressure cooking, protease treatment, microwaving, or heating histologic sections in baths of appropriate buffers, with the standard goal of unmasking antigens hidden by formalin crosslinks or other fixation. See, e.g., Leong et al. Appl. Immnunohistochem. 4(3):201 (1996).
[00158] In some embodiments, an IHC assay is a direct assay. In some embodiments, an IHC assay is an indirect assay. In a direct IHC assay, binding of antibody to the target antigen can be determined directly. A direct assay may use a labeled reagent, such as a fluorescent tag or an enzyme-labeled primary antibody, which can be visualized without further antibody interaction. In a typical indirect assay, unconjugated primary antibody binds to the antigen and then a labeled secondary antibody binds to the primary antibody. Where the secondary antibody is conjugated to an enzymatic label, a chromagenic or fluorogenic substrate is added to provide visualization of the antigen. Signal amplification may occur because several secondary antibodies may react with different epitopes on the primary antibody. [00159] In some embodiments, a primary and/or secondary antibody used for IHC is labeled with a detectable moiety. Numerous labels are available and include, without limitation, radioisotopes, colloidal gold particles, fluorescent or chemiluminescent labels. In some embodiments, a primary and/or secondary antibody used for IHC is labeled with a, radioisotope (e.g. , S, C, I or H) using the techniques described in Current Protocols in Immunology, Volumes 1 and 2, Coligen et al, Ed. Wiley- Interscience, New York, N.Y., Pubs. (1991) for example and radioactivity can be measured using scintillation counting. Other radionuclides include, without limitation, 99Tc,90Y, mln, 32P, UC, 150, 13N, 18F, 51Cr, 57To, 225Ra, 60Co, 59Fe, 57Se, 152Eu, 67CU, 217Ci, 211 At, 212Pb, 47Sc, 109Pd, 234Th, and 40K, 157Gd, 55Mn, 52Tr, and 56Fe. In some embodiments, a primary and/or secondary antibody used for IHC is labeled with Colloidal gold particles. In some embodiments, a primary and/or secondary antibody used for IHC is labeled with fluorescent or chemiluminescent labels including, but not limited to, rare earth chelates (europium chelates), fluorescein and its derivatives, rhodamine and its derivatives, isothiocyanate, phycoerythrin, phycocyanin, allophycocyanin, o-phthaladehyde, fluorescamine, dansyl, umbelliferone, luciferin, luminal label, isoluminal label, an aromatic acridinium ester label, an imidazole label, an acridimium salt label, an oxalate ester label, an aequorin label, 2,3- dihydrophthalazinediones, Texas Red, dansyl, Lissamine, umbelliferone, phycocrytherin, phycocyanin, or commercially available fluorophores such SPECTRUM ORANGE® and SPECTRUM GREEN® and/or derivatives of any one or more of the above. In some embodiments, fluorescence can be quantified using a fluorimeter.
[00160] In some embodiments, a primary and/or secondary antibody used for IHC is labeled with an enzyme. In some embodiments, an enzyme catalyzes a chemical alteration of a chromogenic substrate that can be measured using various techniques. For example, an enzyme may catalyze a color change in a substrate, which can be measured
spectrophotometrically. Alternatively, an enzyme may alter the fluorescence or
chemiluminescence of the substrate. Techniques for quantifying a change in fluorescence would be apparent to the skilled artisan. In some embodiments, a chemiluminescent substrate becomes electronically excited by a chemical reaction and may then emit light which can be measured (e.g., using a chemiluminometer) or donates energy to a fluorescent acceptor.
[00161] In some embodiments, enzymatic labels include luciferases (e.g. firefly luciferase and bacterial luciferase; U.S. Pat. No. 4,737,456), luciferin, 2,3-dihydrophthalazinediones, malate dehydrogenase, urease, peroxidase such as horseradish peroxidase (HRPO), alkaline phosphatase, β-galactosidase, glucoamylase, lysozyme, saccharide oxidases (e.g., glucose oxidase, galactose oxidase, and glucose-6-phosphate dehydrogenase), heterocyclic oxidases (such as uricase and xanthine oxidase), lactoperoxidase, microperoxidase, and the like.
Techniques for conjugating enzymes to antibodies are described in O'Sullivan et al, Methods for the Preparation of Enzyme- Antibody Conjugates for use in Enzyme Immunoassay, in Methods in Enzym. (ed J. Langbne & H. Van Vunakis), Academic press, New York, 73: 147- 166 (1981).
[00162] In some embodiments, a label is indirectly conjugated to any of the antibodies with a selected binding profile provided herein. For example, in some embodiments, an antibody can be conjugated with biotin and any suitable label can be conjugated with avidin, or vice versa. Biotin binds selectively to avidin and thus, the label can be conjugated with the antibody in this indirect manner.
[00163] In some embodiments, after antigen retrieval and an optional blocking step, a tissue section (e.g., an FFPE section or a frozen section) is exposed to one or more of the antibodies provided herein as primary antibodies for a sufficient period of time and under suitable conditions to allow one or more primary antibody to bind to one or more TGFP family member proteins or forms thereof of the tissue section. In some embodiments, appropriate conditions for achieving suitable binding can be determined by routine experimentation, In some embodiments, a slide containing a biological sample is subjected to a wash step to remove unbound and excess amounts of primary antibody.
[00164] In some embodiments, primary antibody is linked to a detectable label, such as paramagnetic ions, radioactive isotopes, fluorochromes, and NM -detectable substances, and the slide is evaluated for staining of one or more TGFP family member proteins or forms thereof using an appropriate imaging apparatus.
[00165] In some embodiments, a tissue sample used to prepare stained tissue sections for evaluating expression of one or more TGFP family member proteins or forms thereof can be collected from a subject before and/or after exposure of the subject to one or more therapeutic agents, e.g., an anti-latent Myostatin antibody. Accordingly, tissue samples may be collected from a subject over a period of time. In some embodiments, a tissues sample (e.g., a muscle tissue sample) is obtained by any suitable procedure, which includes, but not limited to, surgical excision, aspiration or biopsy. In some embodiments, a tissue sample is sectioned and examined for one or more TGFP family member proteins or forms thereof as a fresh specimen. In other embodiments, a tissue sample is frozen for further sectioning. In other embodiments, a tissue sample is preserved by fixing and embedding in paraffin (FFPE) or the like. [00166] In some embodiments, a tissue sample may be fixed by conventional methodology, with the length of fixation depending on the size of the tissue sample and the fixative used. Exemplary fixatives include, but are not limited to neutral buffered formalin, glutaraldehyde, Bouin's or paraformaldehyde. In some embodiments, a tissue sample is fixed with formalin. In some embodiments, a fixed tissue sample is also embedded in paraffin to prepare a formalin-fixed and paraffin- embedded (FFPE) tissue sample. Examples of paraffin include, but are not limited to, Paraplast, Broloid and Tissuemay.
[00167] In some embodiments, a tissue sample is fixed and dehydrated through an ascending series of alcohols, infiltrated and embedded with paraffin or other sectioning media so that the tissue sample may be sectioned. Alternatively, a tissue sample is first sectioned and then the individual sections are fixed.
TGF-β family of growth factors
[00168] Growth factors are cell signaling molecules that stimulate a variety of cellular activities. Due to their broad-reaching influence within biological systems, growth factor signaling is tightly regulated, often through interactions with other biomolecules, the extracellular and/or cellular matrix or within a particular cell environment or niche. These interactions may be direct or indirect.
[00169] Growth factors of the transforming growth factor beta (TGF-β) family are involved in a variety of cellular processes. Growth factor binding to type II receptors leads to type I receptor phosphorylation and activation (Denicourt, C. et al., Another twist in the
transforming growth factor β-induced cell-cycle arrest chronicle. PNAS. 2003.
100(26): 15290-1). Activated type I receptors may in turn phosphorylate receptor-associated SMADs (R-SMADs) promoting co-SMAD (e.g. SMAD4) dimer/trimer formation and nuclear translocation. SMAD complexes collaborate with cofactors to modulate expression of TGF-β family member target genes.
[00170] TGF-β family member signaling cascades are involved in a number of diverse biological pathways including, but not limited to inhibition of cell growth, tissue
homeostasis, extracellular matrix (ECM) remodeling, endothelial to mesenchymal transition (EMT) in cell migration and invasion and immune modulation/suppression as well as in mesenchymal to epithelial transition. TGF-β signaling related to growth inhibition and tissue homeostasis may affect epithelial, endothelial, hematopoietic and immune cells through the activation of p21 and pi to mediate cell cycle arrest and repress myc. In relation to ECM remodeling, TGF-β signaling may increase fibroblast populations and ECM deposition (e.g. collagen). TGF-β signaling related to cell migration and invasion may affect epithelial and/or endothelial cells, inducing stem cell-like phenotypes. This aspect of signaling may play a role in smooth muscle cell proliferation following vascular surgery and/or stenting. In the immune system, TGF-β ligand is necessary for T regulatory cell function and maintenance of immune precursor cell growth and homeostasis. Nearly all immune cells comprise receptors for TGF- β and TGF-β knockout mice die postnataly due in part to inflammatory pathologies. Finally, TGF-β suppresses interferon gamma-induced activation of natural killer cells (Wi, J. et al., 2011. Hepatology. 53(4): 1342-51, the contents of which are herein incorporated by reference in their entirety).
[00171] The solution of the crystal structure of the latent form of TGF-beta is a first for the entire TGF-beta family and offers deep insights into these complexes (Shi, M. et al., Latent TGF-β structure and activation. Nature. 2011 Jun 15; 474(7351):343-9). Almost all signaling in the TGF-beta family goes through a common pathway whereby a dimeric ligand is recognized by a heterotetrameric receptor complex containing two type I and two type II receptors. Each receptor has a serine-threonine kinase domain. Type II receptors
phosphorylate type I receptors, which in turn phosphorylate receptor-regulated Smads that translocate to and accumulate in the nucleus and regulate transcription.
[00172] There are 33 different members of the TGF-beta family in humans (FIG. 1).
Members include the bone morphogenetic proteins (BMP), inhibin, activin, growth and differentiation factor (GDF), myostatin, nodal, anti-Mullerian hormone, and lefty proteins. A review of TGF-β family members, related signaling molecules as well as their relationships can be found in Massague., 2000. Nature Reviews Molecular Cell Biology. 1: 169-78, the contents of which are herein incorporated by reference in their entirety. In some
embodiments, mature growth factors are synthesized along with their prodomains as single polypeptide chains (see FIG. 2). In some embodiments, such polypeptide chains may comprise cleavage sites for separation of prodomains from mature growth factors. In some embodiments, such cleavage sites are furin cleavage sites recognized and cleaved by proprotein convertases.
[00173] In general, homology among TGF-β family member growth factor domains is relatively high. Interestingly, prodomain homology is much lower. This lack of homology may be an important factor in altered growth factor regulation among family members. In some cases, prodomains may guide proper folding and/or dimerization of growth factor domains. Prodomains have very recently been recognized, in some cases, to have important functions in directing growth factors (after secretion) to specific locations in the extracellular matrix (ECM) and/or cellular matrix, until other signals are received that cause growth factor release from latency. Release from latency may occur in highly localized environments whereby growth factors may act over short distances (e.g. from about 1 cell diameter to about a few cell diameters, from about 2 cell diameters to about 100 cell diameters and/or from about 10 cell diameters to about 10,000 cell diameters) and cleared once they reach the circulation. Some growth factor-prodomain complexes are secreted as homodimers. In some embodiments, prodomain-growth factor complexes may be secreted as heterodimers.
[00174] As used herein, the term "TGF-P-related protein" refers to a TGF-β isoform, a TGF-β family member or a TGF-β family member-related protein. TGF-β family members may include, but are not limited to any of those shown in in FIG. 1 and/or listed in Table 1. These include, but are not limited to TGF-β proteins, BMPs, myostatin, GDFs and inhibins. Aspects of the present disclosure provide tools and/or methods for characterizing and/or modulating cellular activities related to growth factor signaling. In other embodiments, tools of the present disclosure may comprise antigens comprising one or more components of one or more TGF^-related proteins. Some tools may comprise antibodies directed toward antigens of the present disclosure. In additional embodiments, tools of the present disclosure may comprise assays for the detection and/or characterization of TGF^-related proteins, the detection and/or characterization of antibodies directed toward TGF^-related proteins and/or the detection and/or characterization of cellular activities and/or their cellular signaling related to TGF^-related proteins.
[00175] Described herein are compounds for the modulation of growth factor activity and/or levels. Such growth factors include growth differentiation factor (GDF) proteins which are TGF-β family member proteins involved in a number of cellular and developmental activities. Part of the disclosure provides GDF-modulatory antibodies as well as methods for generating, optimizing and using such antibodies. Further antibodies include GDF- modulatory antibodies that are capable of distinguishing between various growth factor complexes allowing for growth factor activity modulation that occurs only at sites of specific complex formations.
Proteins of interest
[00176] TGF^-related proteins are involved in a number of cellular processes. In embryogenesis, the 33 members of the TGF-β family of proteins are involved in regulating major developmental processes and the details of the formation of many organs. Much of this regulation occurs before birth; however, the family continues to regulate many processes after birth, including, but not limited to immune responses, wound healing, bone growth, endocrine functions and muscle mass. TGF-P-related proteins are listed and described in International Patent Application No. WO2014074532, the contents of which are herein incorporated by reference in their entirety.
[00177] A list of exemplary TGF-β family pro-proteins, i.e. the protein after removal of the secretion signal sequence, is shown in Table 1. The pro-protein contains, and is the precursor of, the prodomain and the growth factor. Shown in the Table are the names of the originating TGF-β family member and the pro-protein sequence. Also identified in "bold" and
"underlined" are proprotein convertase cleavage sites. Upon cleavage, the resulting prodomain retains this site, whereas the mature growth factor begins following the cleavage site. It is noted that Leftyl and Lefty2 are not cleaved by proprotein convertases just prior to the start of the mature growth factor.
Table 1. Pro-proteins of the TGF-beta family
TGF Member Prodomain and growth factor Sequence SEQ
ID NO
TGF-βΙ LSTCKTIDMELVKRKRIEAIRGQILSKLRLASPPSQGEVPPGPL 1
PEAVLALYNSTRDRVAGESAEPEPEPEADYYAKEVTRVLMV
ETHNEIYDKFKQSTHSIYMFFNTSELREAVPEPVLLSRAELRL
LRLKLKVEQHVELYQKYSNNSWRYLSNRLLAPSDSPEWLSF
DVTGVVRQWLSRGGEIEGFRLSAHCSCDSRDNTLQVDINGFT
TGRRGDLATIHGMNRPFLLLMATPLERAOHLOSSRHRRALD
TNYCFSSTEKNCCVRQLYIDFRKDLGWKWIHEPKGYHANFC
LGPCPYIWSLDTQYSKVLALYNQHNPGASAAPCCVPQALEPL
PIVYYVGRKPKVEQLSNMIVRSCKCS
TGF- 2 SLSTCSTLDMDQFMRKRIEAIRGQILSKLKLTSPPEDYPEPEEV 2
PPEVISIYNSTRDLLQEKASRRAAACERERSDEEYYAKEVYKI
DMPPFFPSENAIPPTFYRPYFRIVRFDVSAMEKNASNLVKAEF
RVFRLQNPKARVPEQRIELYQILKSKDLTSPTQRYIDSKVVKT
RAEGEWLSFDVTDAVHEWLHHKDRNLGFKISLHCPCCTFVP
SNNYIIPNKSEELEARFAGIDGTSTYTSGDQKTIKSTRKKNSG
KTPHLLLMLLPSYRLESOOTNRRKKRALDAAYCFRNVODN
CCLRPLYIDFKRDLGWKWIHEPKGYNANFCAGACPYLWSSD
TQHSRVLSLYNTINPEASASPCCVSQDLEPLTILYYIGKTPKIE
QLSNMIVKSCKCS
TGF- 3 SLSLSTCTTLDFGHIKKKRVEAIRGQILSKLRLTSPPEPTVMTH 3
VPYQVLALYNSTRELLEEMHGEREEGCTQENTESEYYAKEIH
KFDMIQGLAEHNELAVCPKGITSKVFRFNVSSVEKNRTNLFR
AEFRVLRVPNPSSKRNEQRIELFQILRPDEHIAKQRYIGGKNL
PTRGTAEWLSFDVTDTVREWLLRRESNLGLEISIHCPCHTFQP
NGDILENIHEVMEIKFKGVDNEDDHGRGDLGRLKKQKDHHN
PHLILMMIPPHRLDNPGOGGORKKRALDTNYCFRNLEENCC
VRPLYIDFRQDLGWKWVHEPKGYYANFCSGPCPYLRSADTT
HSTVLGLYNTLNPEASASPCCVPQDLEPLTILYYVGRTPKVE
QLSNMVVKSCKCS GDF-11 AEGPAAAAAAAAAAAAAGVGGERSSRPAPSVAPEPDGCPV 4
CVWRQHSRELRLESIKSQILSKLRLKEAPNISREVVKQLLPKA
PPLQQILDLHDFQGDALQPEDFLEEDEYHATTETVISMAQET
DPAVQTDGSPLCCHFHFSPKVMFTKVLKAQLWVYLRPVPRP
ATVYLQILRLKPLTGEGTAGGGGGGRRHIRIRSLKIELHSRSG
HWQSIDFKQVLHSWFRQPQSNWGIEINAFDPSGTDLAVTSLG
PGAEGLHPFMELRVLENTKRSRRNLGLDCDEHSSESRCCRYP
LTVDFEAFGWDWIIAPKRYKANYCSGQCEYMFMQKYPHTH
LVQQANPRGSAGPCCTPTKMSPINMLYFNDKQQIIYGKIPGM
VVDRCGCS
GDF-8 NENSEQKENVEKEGLCNACTWRQNTKSSRIEAIKIQILSKLRL 5 (myostatin) ETAPNISKDVIRQLLPKAPPLRELIDQYDVQRDDSSDGSLEDD
DYHATTETIITMPTESDFLMQVDGKPKCCFFKFSSKIQYNKV
VKAQLWIYLRPVETPTTVFVQILRLIKPMKDGTRYTGIRSLKL
DMNPGTGIWQSIDVKTVLQNWLKQPESNLGIEIKALDENGH
DLAVTFPGPGEDGLNPFLEVKVTDTPKRSRRDFGLDCDEHST
ESRCCRYPLTVDFEAFGWDWIIAPKRYKANYCSGECEFVFLQ
KYPHTHLVHQANPRGSAGPCCTPTKMSPINMLYFNGKEQIIY
GKIPAMVVDRCGCS
Inhibin-beta A SPTPGSEGHSAAPDCPSCALAALPKDVPNSQPEMVEAVKKHI 6
LNMLHLKKRPDVTQPVPKAALLNAIRKLHVGKVGENGYVEI
EDDIGRRAEMNELMEQTSEIITFAESGTARKTLHFEISKEGSD
LSVVERAEVWLFLKVPKANRTRTKVTIRLFQQQKHPQGSLD
TGEEAEEVGLKGERSELLLSEKVVDARKSTWHVFPVSSSIQR
LLDQGKS SLD VRI ACEQCQES GASL VLLGKKKKKEEEGEGK
KKGGGEGGAGADEEKEOSHRPFLMLOAROSEDHPHRRRRR
GLECDGKVNICCKKQFFVSFKDIGWNDWIIAPSGYHANYCE
GECPSHIAGTSGSSLSFHSTVINHYRMRGHSPFANLKSCCVPT
KLRPMSMLYYDDGQNIIKKDIQNMIVEECGCS
Inhibin-beta B SPTPPPTPAAPPPPPPPGSPGGSQDTCTSCGGFRRPEELGRVDG 7
DFLEAVKRHILSRLQMRGRPNITHAVPKAAMVTALRKLHAG
KVREDGRVEIPHLDGHASPGADGQERVSEIISFAETDGLASSR
VRLYFFISNEGNQNLFVVQASLWLYLKLLPYVLEKGSRRKV
RVKVYFQEQGHGDRWNMVEKRVDLKRSGWHTFPLTEAIQA
LFERGERRLNLDVQCDSCQELAVVPVFVDPGEESHRPFVVV
OARLGDSRHRIRKRGLECDGRTNLCCROOFFIDFRLIGWND
WIIAPTGYYGNYCEGSCPAYLAGVPGSASSFHTAVVNQYRM
RGLNPGTVNSCCIPTKLSTMSMLYFDDEYNIVKRDVPNMIVE
ECGCA
Inhibin-beta C TPRAGGQCPACGGPTLELESQRELLLDLAKRSILDKLHLTQR 8
PTLNRPVSRAALRTALQHLHGVPQGALLEDNREQECEIISFAE
TGLSTINQTRLDFHFSSDRTAGDREVQQASLMFFVQLPSNTT
WTLKVRVLVLGPHNTNLTLATQYLLEVDASGWHQLPLGPE
AQ A ACS QGHLTLEL VLEGQ V AQS S VILGGA AHRPF V A AR VR
VGGKHOIHRRGIDCOGGSRMCCROEFFVDFREIGWHDWIIO
PEGYAMNFCIGQCPLHIAGMPGIAASFHTAVLNLLKANTAAG
TTGGGSCCVPTARRPLSLLYYDRDSNIVKTDIPDMVVEACGC
S
Inhibin-beta E QGTGSVCPSCGGSKLAPQAERALVLELAKQQILDGLHLTSRP 9
RITHPPPQAALTRALRRLQPGSVAPGNGEEVISFATVTDSTSA
YSSLLTFHLSTPRSHHLYHARLWLHVLPTLPGTLCLRIFRWGP
RRRRQGSRTLLAEHHITNLGWHTLTLPSSGLRGEKSGVLKLQ
LDCRPLEGNSTVTGQPRRLLDTAGHQQPFLELKIRANEPGAG
RARRRTPTCEPATPLCCRRDHYVDFOELGWRDWILOPEGYO LNYCSGQCPPHLAGSPGIAASFHSAVFSLLKANNPWPASTSC CVPTARRPLSLLYLDHNGNVVKTDVPDMVVEACGCS
Leftyl LTGEQLLGSLLRQLQLKEVPTLDRADMEELVIPTHVRAQYV 10
ALLQRSHGDRSRGKRFSQSFREVAGRFLALEASTHLLVFGM
EQRLPPNSELVQAVLRLFQEPVPKAALHRHGRLSPRSARAR
VTVEWLRVRDDGSNRTSLIDSRLVSVHESGWKAFDVTEAVN
FWQQLSRPRQPLLLQVSVQREHLGPLASGAHKLVRFASQGA
PAGLGEPQLELHTLDLGDYGAQGDCDPEAPMTEGTRCCRQE
MYIDLQGMKWAENWVLEPPGFLAYECVGTCRQPPEALAFK
WPFLGPRQCIASETDSLPMIVSIKEGGRTRPQVVSLPNMRVQ
KCSCASDGALVPRRLQP
Lefty2 LTEEQLLGSLLRQLQLSEVPVLDRADMEKLVIPAHVRAQYV 11
VLLRRSHGDRSRGKRFSQSFREVAGRFLASEASTHLLVFGM
EQRLPPNSELVQAVLRLFQEPVPKAALHRHGRLSPRSAQAR
VTVEWLRVRDDGSNRTSLIDSRLVSVHESGWKAFDVTEAVN
FWQQLSRPRQPLLLQVSVQREHLGPLASGAHKLVRFASQGA
PAGLGEPQLELHTLDLRDYGAQGDCDPEAPMTEGTRCCRQE
MYIDLQGMKWAKNWVLEPPGFLAYECVGTCQQPPEALAFN
WPFLGPRQCIASETASLPMIVSIKEGGRTRPQVVSLPNMRVQ
KCSCASDGALVPRRLQP
GDF-15 LSLAEASRASFPGPSELHSEDSRFRELRKRYEDLLTRLRANQS 12
WEDSNTDLVPAPAVRILTPEVRLGSGGHLHLRISRAALPEGLP
EASRLHRALFRLSPTASRSWDVTRPLRRQLSLARPQAPALHL
RLSPPPSOSDOLLAESSSARPOLELHLRPOAARGRRRARARN
GDHCPLGPGRCCRLHTVRASLEDLGWADWVLSPREVQVTM
CIGACPSQFRAANMHAQIKTSLHRLKPDTVPAPCCVPASYNP
MVLIQKTDTGVSLQTYDDLLAKDCHCI
Anti-Mullerian LLGTEALRAEEPAVGTSGLIFREDLDWPPGIPQEPLCLVALGG 13 hormone DSNGSSSPLRVVGALSAYEQAFLGAVQRARWGPRDLATFGV
CNTGDRQAALPSLRRLGAWLRDPGGQRLVVLHLEEVTWEPT
PSLRFQEPPPGGAGPPELALLVLYPGPGPEVTVTRAGLPGAQS
LCPSRDTRYLVLAVDRPAGAWRGSGLALTLQPRGEDSRLST
ARLQALLFGDDHRCFTRMTPALLLLPRSEPAPLPAHGQLDTV
PFPPPRPSAELEESPPSADPFLETLTRLVRALRVPPARASAPRL
ALDPDALAGFPQGLVNLSDPAALERLLDGEEPLLLLLRPTAA
TTGDPAPLHDPTSAPWATALARRVAAELQAAAAELRSLPGL
PPATAPLLARLLALCPGGPGGLGDPLRALLLLKALQGLRVE
WRGRDPRGPGRAORSAGATAADGPCALRELSVDLRAERSV
LIPETYQANNCQGVCGWPQSDRNPRYGNHVVLLLKMQVRG
AALARPPCCVPTAYAGKLLISLSEERISAHHVPNMVATECGC
R
Inhibin-alpha CQGLELARELVLAKVRALFLDALGPPAVTREGGDPGVRRLP 14
RRHALGGFTHRGSEPEEEEDVSQAILFPATDASCEDKSAARG
LAQEAEEGLFRYMFRPSQHTRSRQVTSAQLWFHTGLDRQGT
AASNSSEPLLGLLALSPGGPVAVPMSLGHAPPHWAVLHLATS
ALSLLTHPVLVLLLRCPLCTCSARPEATPFLVAHTRTRPPSGG
ERARRSTPLMSWPWSPSALRLLORPPEEPAAHANCHRVALN
ISFQELGWERWIVYPPSFIFHYCHGGCGLHIPPNLSLPVPGAPP
TPAQPYSLLPGAQPCCAALPGTMRPLHVRTTSDGGYSFKYET
VPNLLTQHCACI
GDF-1 PVPPGPAAALLQALGLRDEPQGAPRLRPVPPVMWRLFRRRD 15
PQETRSGSRRTSPGVTLQPCHVEELGVAGNIVRHIPDRGAPTR ASEPASAAGHCPEWTVVFDLSAVEPAERPSRARLELRFAAAA AAAPEGGWELSVAQAGQGAGADPGPVLLRQLVPALGPPVR AELLGAAWARNASWPRSLRLALALRPRAPAACARLAEASLL
LVTLDPRLCHPLARPRRDAEPVLGGGPGGACRARRLYVSFR
EVGWHRWVIAPRGFLANYCQGQCALPVALSGSGGPPALNH
AVLRALMHAAAPGAADLPCCVPARLSPISVLFFDNSDNVVL
RQYEDMVVDECGCR
GDF-3 QEYVFLQFLGLDKAPSPQKFQPVPYILKKIFQDREAAATTGV 16
SRDLCYVKELGVRGNVLRFLPDQGFFLYPKKISQASSCLQKL
LYFNLSAIKEREQLTLAQLGLDLGPNSYYNLGPELELALFLV
QEPHVWGQTTPKPGKMFVLRSVPWPQGAVHFNLLDVAKD
WNDNPRKNFGLFLEILVKEDRDSGVNFQPEDTCARLRCSLH
ASLLVVTLNPDOCHPSRKRRAAIPVPKLSCKNLCHRHOLFIN
FRDLGWHKWIIAPKGFMANYCHGECPFSLTISLNSSNYAFMQ
ALMHAVDPEIPQAVCIPTKLSPISMLYQDNNDNVILRHYEDM
VVDECGCG
GDF-5 APDLGQRPQGTRPGLAKAEAKERPPLARNVFRPGGHSYGGG 17
ATNANARAKGGTGQTGGLTQPKKDEPKKLPPRPGGPEPKPG
HPPQTRQATARTVTPKGQLPGGKAPPKAGSVPSSFLLKKARE
PGPPREPKEPFRPPPITPHEYMLSLYRTLSDADRKGGNSSVKL
EAGLANTITSFIDKGQDDRGPVVRKQRYVFDISALEKDGLLG
AELRILRKKPSDTAKPAAPGGGRAAQLKLSSCPSGRQPASLL
DVRSVPGLDGSGWEVFDIWKLFRNFKNSAQLCLELEAWERG
RAVDLRGLGFDRAARQVHEKALFLVFGRTKKRDLFFNEIKA
RSGODDKTVYEYLFSORRKRRAPLATROGKRPSKNLKARCS
RKALHVNFKDMGWDDWIIAPLEYEAFHCEGLCEFPLRSHLE
PTNHAVIQTLMNSMDPESTPPTCCVPTRLSPISILFIDSANNVV
YKQYEDMVVESCGCR
GDF-6 FQQASISSSSSSAELGSTKGMRSRKEGKMQRAPRDSDAGREG 18
QEPQPRPQDEPRAQQPRAQEPPGRGPRVVPHEYMLSIYRTYSI
AEKLGINASFFQSSKSANTITSFVDRGLDDLSHTPLRRQKYLF
DVSMLSDKEELVGAELRLFRQAPSAPWGPPAGPLHVQLFPCL
SPLLLDARTLDPQGAPPAGWEVFDVWQGLRHQPWKQLCLE
LRAAWGELDAGEAEARARGPQQPPPPDLRSLGFGRRVRPPQ
ERALLVVFTRSQRKNLFAEMREQLGSAEAAGPGAGAEGSWP
PPSGAPDARPWLPSPGRRRRRTAFASRHGKRHGKKSRLRCS
KKPLHVNFKELGWDDWIIAPLEYEAYHCEGVCDFPLRSHLEP
TNHAIIQTLMNSMDPGSTPPSCCVPTKLTPISILYIDAGNNVV
YKQYEDMVVESCGCR
GDF-7 RDGLEAAAVLRAAGAGPVRSPGGGGGGGGGGRTLAQAAGA 19
AAVPAAAVPRARAARRAAGSGFRNGSVVPHHFMMSLYRSL
AGRAPAGAAAVSASGHGRADTITGFTDQATQDESAAETGQS
FLFDVSSLNDADEVVGAELRVLRRGSPESGPGSWTSPPLLLLS
TCPGAARAPRLLYSRAAEPLVGQRWEAFDVADAMRRHRRE
PRPPRAFCLLLRAVAGPVPSPLALRRLGFGWPGGGGSAAEER
AVLVVSSRTQRKESLFREIRAQARALGAALASEPLPDPGTGT
ASPRAVIGGRRRRRTALAGTRTAOGSGGGAGRGHGRRGRS
RCSRKPLHVDFKELGWDDWIIAPLDYEAYHCEGLCDFPLRSH
LEPTNHAIIQTLLNSMAPDAAPASCCVPARLSPISILYIDAANN
VVYKQYEDMVVEACGCR
BMP- 10 SPIMNLEQSPLEEDMSLFGDVFSEQDGVDFNTLLQSMKDEFL 20
KTLNLSDIPTQDSAKVDPPEYMLELYNKFATDRTSMPSANIIR
SFKNEDLFSQPVSFNGLRKYPLLFNVSIPHHEEVIMAELRLYT
LVQRDRMIYDGVDRKITIFEVLESKGDNEGERNMLVLVSGEI
YGTNSEWETFDVTDAIRRWQKSGSSTHQLEVHIESKHDEAE
DASSGRLEIDTSAQNKHNPLLIVFSDDQSSDKERKEELNEMIS HEQLPELDNLGLDSFSSGPGEEALLQMRSNIIYDSTARIRRNA KGNYCKRTPLYIDFKEIGWDSWIIAPPGYEAYECRGVCNYPL AEHLTPTKHAIIQALVHLKNSQKASKACCVPTKLEPISILYLD KGVVTYKFKYEGMAVSECGCR
BMP-9 (GDF-2) KPLQSWGRGSAGGNAHSPLGVPGGGLPEHTFNLKMFLENVK 21
VDFLRSLNLSGVPSQDKTRVEPPQYMIDLYNRYTSDKSTTPA
SNIVRSFSMEDAISITATEDFPFQKHILLFNISIPRHEQITRAELR
LYVSCQNHVDPSHDLKGSVVIYDVLDGTDAWDSATETKTFL
VSQDIQDEGWETLEVSSAVKRWVRSDSTKSKNKLEVTVESH
RKGCDTLDISVPPGSRNLPFFVVFSNDHSSGTKETRLELREMI
SHEQESVLKKLSKDGSTEAGESSHEEDTDGHVAAGSTLARR
KRSAGAGSHCQKTSLRVNFEDIGWDSWIIAPKEYEAYECKG
GCFFPLADDVTPTKHAIVQTLVHLKFPTKVGKACCVPTKLSPI
SVLYKDDMGVPTLKYHYEGMSVAECGCR
Nodal TVATALLRTRGQPSSPSPLAYMLSLYRDPLPRADIIRSLQAED 22
V A VDGQNWTF AFDFSFLS QQEDL AWAELRLQLS SP VDLPTE
GSLAIEIFHQPKPDTEQASDSCLERFQMDLFTVTLSQVTFSLG
SMVLEVTRPLSKWLKRPGALEKQMSRVAGECWPRPPTPPAT
NVLLMLYSNLSQEQRQLGGSTLLWEAESSWRAQEGQLSWE
WGKRHRRHHLPDRSOLCRKVKFOVDFNLIGWGSWIIYPKO
YNAYRCEGECPNPVGEEFHPTNHAYIQSLLKRYQPHRVPSTC
CAPVKTKPLSMLYVDNGRVLLDHHKDMIVEECGCL
BMP-2 LVPELGRRKFAAASSGRPSSQPSDEVLSEFELRLLSMFGLKQR 23
PTPSRDAVVPPYMLDLYRRHSGQPGSPAPDHRLERAASRAN
TVRSFHHEESLEELPETSGKTTRRFFFNLSSIPTEEFITSAELQV
FREQMQDALGNNSSFHHRINIYEIIKPATANSKFPVTRLLDTR
LVNQNASRWESFDVTPAVMRWTAQGHANHGFVVEVAHLE
EKQGVSKRHVRISRSLHQDEHSWSQIRPLLVTFGHDGKGHPL
HKREKROAKHKORKRLKSSCKRHPLYVDFSDVGWNDWIV
APPGYHAFYCHGECPFPLADHLNSTNHAIVQTLVNSVNSKIP
KACCVPTELSAISMLYLDENEKVVLKNYQDMVVEGCGCR
BMP-4 GASHASLIPETGKKKVAEIQGHAGGRRSGQSHELLRDFEATL 24
LQMFGLRRRPQPSKSAVIPDYMRDLYRLQSGEEEEEQIHSTG
LEYPERPASRANTVRSFHHEEHLENIPGTSENSAFRFLFNLSSI
PENEVISSAELRLFREQVDQGPDWERGFHRINIYEVMKPPAE
VVPGHLITRLLDTRLVHHNVTRWETFDVSPAVLRWTREKQP
NYGLAIEVTHLHQTRTHQGQHVRISRSLPQGSGNWAQLRPL
LVTFGHDGRGHALTRRRRAKRSPKHHSORARKKNKNCRRH
SLYVDFSDVGWNDWIVAPPGYQAFYCHGDCPFPLADHLNST
NHAIVQTLVNSVNSSIPKACCVPTELSAISMLYLDEYDKVVL
KNYQEMVVEGCGCR
BMP-5 DNHVHSSFIYRRLRNHERREIQREILSILGLPHRPRPFSPGKQA 25
SSAPLFMLDLYNAMTNEENPEESEYSVRASLAEETRGARKG
YPASPNGYPRRIQLSRTTPLTTQSPPLASLHDTNFLNDADMV
MSFVNLVERDKDFSHQRRHYKEFRFDLTQIPHGEAVTAAEFR
IYKDRSNNRFENETIKISIYQIIKEYTNRDADLFLLDTRKAQAL
DVGWLVFDITVTSNHWVINPQNNLGLQLCAETGDGRSINVK
SAGLVGROGPOSKOPFMVAFFKASEVLLRSVRAANKRKNO
NRNKSSSHQDSSRMSSVGDYNTSEQKQACKKHELYVSFRDL
GWQDWIIAPEGYAAFYCDGECSFPLNAHMNATNHAIVQTLV
HLMFPDHVPKPCCAPTKLNAISVLYFDDSSNVILKKYRNMV
VRSCGCH
BMP-6 CCGPPPLRPPLPAAAAAAAGGQLLGDGGSPGRTEQPPPSPQS 26
SSGFLYRRLKTQEKREMQKEILSVLGLPHRPRPLHGLQQPQP PALRQQEEQQQQQQLPRGEPPPGRLKSAPLFMLDLYNALSA
DNDEDGASEGERQQSWPHEAASSSQRRQPPPGAAHPLNRKS
LLAPGSGSGGASPLTSAQDSAFLNDADMVMSFVNLVEYDKE
FSPRQRHHKEFKFNLSQIPEGEVVTAAEFRIYKDCVMGSFKN
QTFLISIYQVLQEHQHRDSDLFLLDTRVVWASEEGWLEFDIT
ATSNLWVVTPQHNMGLQLSVVTRDGVHVHPRAAGLVGRD
GPYDKOPFMVAFFKVSEVHVRTTRSASSRRROOSRNRSTOS
QDVARVSSASDYNSSELKTACRKHELYVSFQDLGWQDWIIA
PKGYAANYCDGECSFPLNAHMNATNHAIVQTLVHLMNPEY
VPKPCCAPTKLNAISVLYFDDNSNVILKKYRNMVVRACGCH
BMP-7 DFSLDNEVHSSFIHRRLRSQERREMQREILSILGLPHRPRPHLQ 27
GKHNSAPMFMLDLYNAMAVEEGGGPGGQGFSYPYKAVFST
QGPPLASLQDSHFLTDADMVMSFVNLVEHDKEFFHPRYHHR
EFRFDLSKIPEGEAVTAAEFRIYKDYIRERFDNETFRISVYQVL
QEHLGRESDLFLLDSRTLWASEEGWLVFDITATSNHWVVNP
RHNLGLQLSVETLDGQSINPKLAGLIGRHGPQNKQPFMVAFF
KATEVHFRSIRSTGSKQRSQNRSKTPKNQEALRMANVAENS
SSDQRQACKKHELYVSFRDLGWQDWIIAPEGYAAYYCEGEC
AFPLNSYMNATNHAIVQTLVHFINPETVPKPCCAPTQLNAISV
L YFDDS SNVILKKYRNM V VR ACGCH
BMP-8A GGGPGLRPPPGCPORRLGARERRDVOREILAVLGLPGRPRPR 28
APPAASRLPASAPLFMLDLYHAMAGDDDEDGAPAEQRLGR
ADLVMSFVNMVERDRALGHQEPHWKEFRFDLTQIPAGEAVT
AAEFRIYKVPSIHLLNRTLHVSMFQVVQEQSNRESDLFFLDL
QTLRAGDEGWLVLDVTAASDCWLLKRHKDLGLRLYVETED
GHSVDPGLAGLLGORAPRSOOPFVVTFFRASPSPIRTPRAVR
PLRRRQPKKSNELPQANRLPGIFDDVRGSHGRQVCRRHELYV
SFQDLGWLDWVIAPQGYSAYYCEGECSFPLDSCMNATNHAI
LQSLVHLMKPNAVPKACCAPTKLSATSVLYYDSSNNVILRK
HRNMVVKACGCH
BMP-8B GGGPGLRPPPGCPORRLGARERRDVOREILAVLGLPGRPRPR 29
APPAASRLPASAPLFMLDLYHAMAGDDDEDGAPAERRLGRA
DLVMSFVNMVERDRALGHQEPHWKEFRFDLTQIPAGEAVTA
AEFRIYKVPSIHLLNRTLHVSMFQVVQEQSNRESDLFFLDLQT
LRAGDEGWLVLDVTAASDCWLLKRHKDLGLRLYVETEDGH
SVDPGLAGLLGORAPRSOOPFVVTFFRASPSPIRTPRAVRPLR
RRQPKKSNELPQANRLPGIFDDVHGSHGRQVCRRHELYVSF
QDLGWLDWVIAPQGYSAYYCEGECSFPLDSCMNATNHAILQ
SLVHLMMPDAVPKACCAPTKLSATSVLYYDSSNNVILRKHR
NMVVKACGCH
BMP- 15 MEHRAQMAEGGQSSIALLAEAPTLPLIEELLEESPGEQPRKPR 30
LLGHSLRYMLELYRRSADSHGHPRENRTIGATMVRLVKPLTS
VARPHRGTWHIQILGFPLRPNRGLYQLVRATVVYRHHLQLT
RFNLSCHVEPWVQKNPTNHFPSSEGDSSKPSLMSNAWKEMD
ITQLVQQRFWNNKGHRILRLRFMCQQQKDSGGLELWHGTSS
LDIAFLLLYFNDTHKSIRKAKFLPRGMEEFMERESLLRRTRO
ADGISAEVTASSSKHSGPENNQCSLHPFQISFRQLGWDHWIIA
PPFYTPNYCKGTCLRVLRDGLNSPNHAIIQNLINQLVDQSVPR
PSCVPYKYVPISVLMIEANGSILYKEYEGMIAESCTCR
GDF-9 SQASGGEAQIAASAELESGAMPWSLLQHIDERDRAGLLPALF 31
KVLSVGRGGSPRLQPDSRALHYMKKLYKTYATKEGIPKSNR
SHLYNTVRLFTPCTRHKQAPGDQVTGILPSVELLFNLDRITTV
EHLLKSVLLYNINNSVSFSSAVKCVCNLMIKEPKSSSRTLGRA
PYSFTFNSQFEFGKKHKWIQIDVTSLLQPLVASNKRSIHMSIN FTCMKDQLEHPSAQNGLFNMTLVSPSLILYLNDTSAQAYHS
WYSLHYKRRPSQGPDQERSLSAYPVGEEAAEDGRSSHHRHR
RGQETVSSELKKPLGPASFNLSEYFRQFLLPQNECELHDFRLS
FSQLKWDNWIVAPHRYNPRYCKGDCPRAVGHRYGSPVHTM
VQNIIYEKLDSSVPRPSCVPAKYSPLSVLTIEPDGSIAYKEYED
MIATKCTCR
BMP-3 ERPKPPFPELRKAVPGDRTAGGGPDSELQPQDKVSEHMLRLY 32
DRYSTVQAARTPGSLEGGSQPWRPRLLREGNTVRSFRAAAA
ETLERKGLYIFNLTSLTKSENILSATLYFCIGELGNISLSCPVSG
GCSHHAQRKHIQIDLSAWTLKFSRNQSQLLGHLSVDMAKSH
RDIMSWLSKDITQLLRKAKENEEFLIGFNITSKGRQLPKRRLP
FPEPYILVYANDAAISEPESVVSSLQGHRNFPTGTVPKWDSHI
RAALSIERRKKRSTGVLLPLQNNELPGAEYQYKKDEVWEER
KPYKTLOAOAPEKSKNKKKORKGPHRKSOTLOFDEOTLKK
ARRKQWIEPRNCARRYLKVDFADIGWSEWIISPKSFDAYYCS
GACQFPMPKSLKPSNHATIQSIVRAVGVVPGIPEPCCVPEKMS
SLSILFFDENKNVVLKVYPNMTVESCACR
GDF-10 SHRAPAWSALPAAADGLQGDRDLQRHPGDAAATLGPSAQD 33
MVAVHMHRLYEKYSRQGARPGGGNTVRSFRARLEVVDQK
AVYFFNLTSMQDSEMILTATFHFYSEPPRWPRALEVLCKPRA
KNASGRPLPLGPPTRQHLLFRSLSQNTATQGLLRGAMALAPP
PRGLWQAKDISPIVKAARRDGELLLSAQLDSEERDPGVPRPS
PYAPYILVYANDLAISEPNSVAVTLQRYDPFPAGDPEPRAAP
NNSADPRVRRAAQATGPLQDNELPGLDERPPRAHAQHFHKH
QLWPSPFRALKPRPGRKDRRKKGQEVFMAASQVLDFDEKT
MOKARRKOWDEPRVCSRRYLKVDFADIGWNEWIISPKSFDA
YYCAGACEFPMPKIVRPSNHATIQSIVRAVGIIPGIPEPCCVPD
KMNSLGVLFLDENRNVVLKVYPNMSVDTCACR
GDNF FPLPAGKRPPEAPAEDRSLGRRRAPFALSSDSNMPEDYPDQF 34
DDVMDFIOATIKRLKRSPDKOMAVLPRRERNROAAAANPE
NSRGKGRRGQRGKNRGCVLTAIHLNVTDLGLGYETKEELIFR
YCSGSCDAAETTYDKILKNLSRNRRLVSDKVGQACCRPIAFD
DDLSFLDDNLVYHILRKHSAKRCGCI
NRTN IWMCREGLLLSHRLGPALVPLHRLPRTLDARIARLAQYRALL 35
OGAPDAMELRELTPWAGRPPGPRRRAGPRRRRARARLGAR
PCGLRELEVRVSELGLGYASDETVLFRYCAGACEAAARVYD
LGLRRLRQRRRLRRERVRAQPCCRPTAYEDEVSFLDAHSRY
HTVHELSARECACV
PSPN WGPDARGVPVADGEFSSEOVAKAGGTWLGTHRPLARLRRA 36
LSGPCQLWSLTLSVAELGLGYASEEKVIFRYCAGSCPRGART QHGLALARLQGQGRAHGGPCCRPTRYTDVAFLDDRHRWQR LPQLSAAACGCGG
ARTN SLGSAPRSPAPREGPPPVLASPAGHLPGGRTARWCSGRARRP 37
PPOPSRPAPPPPAPPSALPRGGRAARAGGPGSRARAAGARGC
RLRSQLVPVRALGLGHRSDELVRFRFCSGSCRRARSPHDLSL
ASLLGAGALRPPPGSRPVSQPCCRPTRYEAVSFMDVNSTWRT
VDRLSATACGCLG
[00178] It is noted that some prodomains may be cleaved by proprotein convertase enzymes. As used herein, the term "proprotein convertase" refers to an enzyme that cleaves a prodomain from a translated protein to facilitate protein maturation. Some proprotein convertases of the present disclosure include the subtilisin-like proprotein convertase (SPC) family member enzymes. The SPC family comprises calcium-dependent serine endopro teases that include, but are not limited to furin/PACE, PC 1/3, PC2, PC4, PC5/6, PACE4 and PC7 (Fuller et al, 2009. Invest Ophthalmol Vis Sci. 50(12):5759-68, the contents of which are herein incorporated by reference in their entirety). GDF-11 may in some cases, be cleaved by PC5/6. In some cases, proprotein convertases may cleave proproteins at additional sites, other than those indicated in Table 1. In some embodiments, pro-proteins may be cleaved at a first cleavage site (the first site being the site closest to the N-terminus). In other embodiments, pro-proteins may be cleaved at a cleavage site other than a first cleavage site. In some cases, proprotein convertase cleavage may occur intracellularly. In some cases, proprotein convertase cleavage may occur extracellularly.
[00179] Many TGF-β family member proteins are synthesized in conjunction with prodomains. Some prodomains may remain associated with growth factors after cleavage. Such associations may form latent growth factor-prodomain complexes (GPCs) that modulate the availability of growth factors for cell signaling. Growth factors may be released from latency in GPCs through associations with one or more extracellular proteins. In some cases, growth factor release may rely on force applied to GPCs through extracellular protein interactions. Such forces may pull from C-terminal and/or N-terminal regions of GPCs resulting in the release of associated growth factors.
[00180] In some TGF-β family members, the prodomain portion of the GPC is responsible for growth factor retention and blocking the interaction of retained growth factors with their receptors. Such GPCs, where the bound growth factor is unable to promote signaling activity, are also referred to herein as "latent complexes." Prodomain portions of GPCs that function to block growth factor signaling activity are referred to as latency associated peptides (LAPs). TGF-βΙ, 2 and 3 are know to comprise LAPs. GDF prodomains also function to block growth factor activity. Some prodomains may comprise LAP-like domains. As used herein, the term "LAP-like domain" refers to prodomain portions of GPCs and/or free prodomains that may be structurally similar or synthesized in a similar manner to LAPs, but that may not function to prevent growth factor/receptor interactions.
[00181] FIG. 3 is a schematic depicting an embodiment of a GPC before and after growth factor release. According to such embodiments, growth factor dimers may associate with prodomain modules to form a GPC. In some embodiments, GPCs comprise protein modules necessary for different aspects of growth factor signaling, secretion, latency and/or release from latent GPCs. As used herein, the term "protein module" refers to any component, region and/or feature of a protein. Protein modules may vary in length, comprising one or more amino acids. Protein modules may be from about 2 amino acid residues in length to about 50 amino acid residues in length, from about 5 amino acid residues in length to about 75 amino acid residues in length, from about 10 amino acid residues in length to about 100 amino acid residues in length, from about 25 amino acid residues in length to about 150 amino acid residues in length, from about 125 amino acid residues in length to about 250 amino acid residues in length, from about 175 amino acid residues in length to about 400 amino acid residues in length, from about 200 amino acid residues in length to about 500 amino acid residues in length and/or at least 500 amino acid residues in length.
[00182] In some embodiments, protein modules comprise one or more regions with known functional features (e.g. protein binding domain, nucleic acid binding domain, hydrophobic pocket, etc). Protein modules may comprise functional protein domains necessary for different aspects of growth factor signaling, secretion, latency and/or release from latent conformations.
[00183] In some embodiments, protein modules may be derived from TGF-P-related proteins. Such protein modules may include, but are not limited to prodomains (e.g. latency- associated peptides (LAPs) or LAP-like domains), growth factor domains, fastener regions, proprotein convertase cleavage sites (e.g. furin cleavage sites), B/TP cleavage sites, arm regions, finger regions, residues (such as cysteine residues for example) for extracellular protein associations, latency loops (also referred to herein as latency lassos), alpha 1 helical regions, alpha 2 helical regions, RGD sequences and bowtie regions. FIG. 4 is a schematic diagram showing some of the protein modules present in a GPC as well as in the prodomains and growth factor dimer after activating cleavage of the GPC.
[00184] In some embodiments, prodomains may associate with growth factors in GPCs. Some prodomains may sterically prevent growth factor association with one or more cellular receptors. Prodomains may comprise arm regions and/or straight jacket regions. Some prodomains may comprise C-terminal regions referred to herein as "bowtie regions." In some prodomain dimers, bowtie regions of each monomer may associate and/or interact. Such associations may comprise disulfide bond formation, as is found between monomers of TGF- β isoform LAPs.
[00185] In some embodiments, arm regions may comprise trigger loop regions. Trigger loops may comprise regions that associate with integrins. Such regions may comprise amino acid sequences comprising RGD (Arg-Gly-Asp). Regions comprising RGD sequences are referred to herein as RGD sequence regions. In some embodiments, prodomains comprise latency loops (also referred to herein as latency lassos). Some latency loops may maintain associations between prodomains and growth factors present within GPCs. Prodomains may also comprise fastener regions. Such fastener regions may promote associations between prodomains and growth factors present within GPCs by maintaining prodomain
conformations that promote growth factor retention.
[00186] In some cases, GPCs may require enzymatic cleavage to promote dissociation of bound growth factors and growth factor activity. Such enzymatic cleavage events are referred to herein as "activating cleavage" events. Activating cleavage of GPCs may be carried out in some instances by members of the BMP- 1/Tolloid- like proteinase (B/TP) family (Muir et al., 2011. J Biol Chem. 286(49):41905-11, the contents of which are herein incorporated by reference in their entirety). These metaloproteinases may include, but are not limited to BMP- 1, mammalian tolloid protein (mTLD), mammalian tolloid-like 1 (mTLLl) and mammalian tolloid-like 2 (mTLL2). Exemplary GPCs that may be subjected to activating cleavage by such metalloproteinases may include, but are not limited to GDF-8 and GDF-11. In some cases, GDF-8 may be cleaved by mTLL2. In some cases, activating cleavages may occur intracellularly. In some cases, activating cleavages may occur extracellularly.
[00187] Growth factor release from GPCs may require cleavage by a proprotein convertase enzyme followed by an activating cleavage [e.g. by one or more members of the BMP- 1/Tolloid-like proteinase (B/TP) family.] In one example, GDF-8 and GDF-11 GPCs may be transformed by furin cleavage into a latent complex that further requires cleavage by
BMP/Tolloid proteases for growth factor release.
[00188] In some embodiments, the present disclosure provides polypeptide inhibitors (e.g., antibodies) that inhibit one or more members of the B/TP family. Such inhibitors may block cleavage of BMP-l/Tolloid cleavage sites, including, but not limited to BMP/Tolloid cleavage sites on one or more latent complexes (e.g., GDF-8 latent complexes and/or GDF-11 latent complexes).
[00189] In some cases, activating cleavage may not lead to dissociation of bound growth factor, but instead may promote an active conformation of the GPC. As used herein when referring to a GPC, the term "active conformation" refers to a GPC protein confirmation that allows the growth factor to engage in receptor interaction. Such scenarios have been predicted with proBMP-7 and proBMP-9 (Sengle, G. et al., 2008. JMB. 381: 1025-39 and Mi et al., 2015. PNAS. 112(12): 3710-5, the contents of each of which are herein incorporated by reference in their entirety). In some embodiments, the present disclosure provides antibodies that specifically target active conformations to modulate growth factor activity. [00190] Active conformations of GDF-8 GPCs or GDF- 11 GPCs are referred to herein as "primed" complexes, and can be produced by the sequential cleavage of GPCs at the furin cleavage site and the BMP/Tolloid cleavage site. Primed complexes (either the entire complex or portions of the complex) may bind receptors resulting in receptor signaling. In some cases, prodomains may be dissociated from growth factors upon receptor binding and/or signaling activity. In some cases, prodomains may remain associated with growth factors upon receptor binding and/or signaling activity. In some cases, prodomains may become partially dissociated from growth factors during receptor binding and/or signaling activity.
[00191] In some cases, primed complexes may bind preferentially to one or more receptors over one or more other receptors. In some cases, receptor activity resulting from primed complex interactions may be quenched or competed for by excess prodomain or fragments thereof.
[00192] In some embodiments, the present disclosure provides polypeptide inhibitors (e.g., inhibiting antibodies) that block the formation of primed complexes from latent complexes. In some cases, such inhibitors bind BMP/Tolloid cleavage sites on latent GPCs (e.g., latent GDF-8 or latent GDF-11). In some embodiments, such inhibitors prevent cleavage of the BMP/Tolloid cleavage site.
[00193] Straight jacket regions may comprise alpha 1 helical regions. In some
embodiments, alpha 1 helical regions may be positioned between growth factor monomers. Some alpha 1 helical regions comprise N-terminal regions of prodomains. Alpha 1 helical regions may also comprise N-terminal regions for extracellular associations. Such
extracellular associations may comprise extracellular matrix proteins and/or proteins associated with the extracellular matrix. Some extracellular associations may comprise associations with proteins that may include, but are not limited to LTBPs (e.g. LTBP1, LTBP2, LTBP3 and/or LTBP4), fibrillins (e.g. fibrillin- 1, fibrillin-2, fibrillin-3 and/or fibrillin-4), perlecan, decorin and/or GASPs. N-terminal extracellular associations may comprise disulfide bonds between cysteine residues. In some cases, extracellular matrix proteins and/or proteins associated with the extraceullar matrix may comprise bonds or interactions with one or more regions of prodomains other than N-terminal regions.
[00194] In some embodiments, growth factor domains comprise one or more growth factor monomers. Some growth factor domains comprise growth factor dimers. Such growth factor domains may comprise growth factor homodimers or heterodimers (comprising growth factor monomers from different TGF-P-related proteins). Some growth factor domains may comprise fingers regions. Such fingers regions may comprise β-pleated sheets. Fingers regions may associate with prodomains. Some fingers regions may maintain association between growth factor domains and prodomains.
[00195] In some embodiments, recombinant proteins of the present disclosure may comprise protein modules from growth differentiation factor (GDF) proteins. Such GDF protein modules may comprise the protein modules and/or amino acid sequences listed in Table 2. In some embodiments, protein modules of the present disclosure may comprise amino acid sequences similar to those in Table 2, but comprise additional or fewer amino acids than those listed. Some such amino acid sequences may comprise about 1 more or fewer amino acids, about 2 more or fewer amino acids, about 3 more or fewer amino acids, about 4 more or fewer amino acids, about 5 more or fewer amino acids, about 6 more or fewer amino acids, about 7 more or fewer amino acids, about 8 more or fewer amino acids, about 9 more or fewer amino acids, about 10 more or fewer amino acids or greater than 10 more or fewer amino acids on N-terminal and/or C-terminal ends.
Table 2. GDF protein modules
TGF-β Protein Module Prodomain and growth factor Sequence SEQ Family ID Member NO
GDF- 8 prodomain NENSEQKENVEKEGLCNACTWRQNTKSSRIEA 38
IKIQILSKLRLETAPNISKDVIRQLLPKAPPLREL IDQYDVQRDDSSDGSLEDDDYHATTETIITMPT ESDFLMQVDGKPKCCFFKFSSKIQYNKVVKAQ LWIYLRPVETPTTVFVQILRLIKPMKDGTRYTG IRSLKLDMNPGTGIWQSIDVKTVLQNWLKQPE SNLGIEIKALDENGHDLAVTFPGPGEDGLNPFL EVKVTDTPKRSRR
GDF- 11 prodomain AEGPAAAAAAAAAAAAAGVGGERSSRPAPSV 39
APEPDGCPVCVWRQHSRELRLESIKSQILSKLR
LKEAPNISREVVKQLLPKAPPLQQILDLHDFQG
DALQPEDFLEEDEYHATTETVISMAQETDPAV
QTDGSPLCCHFHFSPKVMFTKVLKAQLWVYL
RPVPRPATVYLQILRLKPLTGEGTAGGGGGGR
RHIRIRSLKIELHSRSGHWQSIDFKQVLHSWFR
QPQSNWGIEINAFDPSGTDLAVTSLGPGAEGLH
PFMELRVLENTKRSRR
GDF- 8 straight jacket NENSEQKENVEKEGLCNACTWRQNTKSSRIEA 40 region IKIQILSKLRLETAPNISKDVIRQLLPKAPPL
GDF- 11 straight jacket AEGPAAAAAAAAAAAAAGVGGERSSRPAPSV 41 region APEPDGCPVCVWRQHSRELRLESIKSQILSKLR
LKEAPNISREVVKQLLPKAPPL
GDF- 8 growth factor DFGLDCDEHSTESRCCRYPLTVDFEAFGWDWI 42 domain IAPKRYKANYCSGECEFVFLQKYPHTHLVHQA
NPRGSAGPCCTPTKMSPINMLYFNGKEQIIYGK IPAMVVDRCGCS GDF-11 growth factor NLGLDCDEHSSESRCCRYPLTVDFEAFGWDWI 43 domain IAPKRYKANYCSGQCEYMFMQKYPHTHLVQQ
ANPRGSAGPCCTPTKMSPINMLYFNDKQQIIYG KIPGMVVDRCGCS
GDF-8 fastener region residues 87-89, DYH -
GDF-11 fastener region residues 110-112, EYH -
GDF-8 furin cleavage site RSRR 44 and GDF- region
11
GDF-8 BMP/Tolloid between residues R75 and D76 —
cleavage site
GDF-11 BMP/Tolloid between residues G97 and D98 —
cleavage site
GDF-8 arm region RELIDQYDVQRDDSSDGSLEDDDYHATTETIIT 45
MPTESDFLMQVDGKPKCCFFKFSSKIQYNKVV KAQLWIYLRPVETPTTVFVQILRLIKPMKDGTR YTGIRSLKLDMNPGTGIWQSIDVKTVLQNWLK QPESNLGIEIKALDENGHDLAVTFPGPGEDGLN PFLEVKVTDTPKRSRR
GDF-11 arm region QQILDLHDFQGDALQPEDFLEEDEYHATTETVI 46
SMAQETDPAVQTDGSPLCCHFHFSPKVMFTKV
LKAQLWVYLRPVPRPATVYLQILRLKPLTGEG
TAGGGGGGRRHIRIRSLKIELHSRSGHWQSIDF
KQVLHSWFRQPQSNWGIEINAFDPSGTDLAVT
SLGPGAEGLHPFMELRVLENTKRSRR
GDF-8 fingers region 1 CRYPLTVDFEAFGWDWIIAPKRYKANYCS 47 and GDF- 11
GDF-8 fingers region 2 CTPTKMSPINMLYFNGKEQIIYGKIPAMVVDRC 48
GCS
GDF-11 fingers region 2 CTPTKMSPINMLYFNDKQQIIYGKIPGMVVDR 49
CGCS
GDF-8 latency loop RLET APNIS KD VIRQLLPK APPL 50
GDF-11 latency loop RLKEAPNISREVVKQLLPKAPP 51
GDF-8 alpha 1 helical GLCNACT WRQNTKS SRIE AIKIQILS K 52 region
GDF-11 alpha 1 helical DGCPVCVWRQHSRELRLESIKSQILSKL 53 region
GDF-8 bowtie region DENGHDLAVTFPGP 54
GDF-11 bowtie region DPSGTDLAVTSLG 55
[00196] Some recombinant proteins of the present disclosure may comprise GDF-15, GDF- 15 signaling pathway-related proteins and/or modules and/or portions thereof. GDF-15 is a TGF-β family protein that is highly expressed in liver. Expression of GDF-15 is dramatically upregulated following liver injury (Hsiao et al. 2000. Mol Cell Biol. 20(10):3742-51).
Additionally, its expression in macrophages may serve a protective function in the context of atherosclerosis, possibly through regulation of adhesion molecule expression (Preusch et al., 2013. Eur J Med Res. 18:19). While a member of the TGF-β family, GDF-15 comprises less than 30% homology with other members, making it the most divergent member of the family (Tanno et al., 2010. Curr Opin Hematol. 17(3): 184-90, the contents of which are incorporated herein by reference in their entirety). The mature form is soluble and can be found in the blood stream. Interestingly, GDF-15 levels in circulation have been found to negatively correlate with hepcidin levels, suggesting a role for GDF-15 in iron load and/or metabolism (Finkenstedt et al., 2008. British Journal of Haematology. 144:789-93). Elevated GDF-15 in the blood is also associated with ineffective and/or apoptotic erythropoiesis, such as in subjects suffering from beta-thalassemia or dyserythropoietic anemias.
[00197] In some embodiments, recombinant proteins of the present disclosure may comprise protein modules from activin subunits. Such protein modules may comprise the protein modules and/or amino acid sequences of the activin subunit inhibin beta A, listed in Table 3. In some embodiments, protein modules of the present disclosure may comprise amino acid sequences similar to those in Table 3, but comprise additional or fewer amino acids than those listed. Some such amino acid sequences may comprise about 1 more or fewer amino acids, about 2 more or fewer amino acids, about 3 more or fewer amino acids, about 4 more or fewer amino acids, about 5 more or fewer amino acids, about 6 more or fewer amino acids, about 7 more or fewer amino acids, about 8 more or fewer amino acids, about 9 more or fewer amino acids, about 10 more or fewer amino acids or greater than 10 more or fewer amino acids on N-terminal and/or C-terminal ends.
Table 3. Inhibin beta A protein modules
Protein Module Prodomain and growth factor Sequence SEQ
ID NO
prodomain SPTPGSEGHSAAPDCPSCALAALPKDVPNSQPE 56
MVEAVKKHILNMLHLKKRPDVTQPVPKAALL
NAIRKLHVGKVGENGYVEIEDDIGRRAEMNEL
MEQTSEIITFAESGTARKTLHFEISKEGSDLSVV
ERAEVWLFLKVPKANRTRTKVTIRLFQQQKHP
QGSLDTGEEAEEVGLKGERSELLLSEKVVDAR
KSTWHVFPVSSSIQRLLDQGKSSLDVRIACEQC
QES GASL VLLGKKKKKEEEGEGKKKGGGEGG
AGADEEKEQSHRPFLMLQARQSEDHPHRRRR
R
straight jacket SPTPGSEGHSAAPDCPSCALAALPKDVPNSQPE 57 region MVEAVKKHILNMLHLKKRPDVTQPVPKAALL
N
growth factor RGLECDGKVNICCKKQFFVSFKDIGWNDWIIA 58 domain PSGYHANYCEGECPSHIAGTSGSSLSFHSTVIN
HYRMRGHSPFANLKSCCVPTKLRPMSMLYYD DGQNIIKKDIQNMIVEECGCS
fastener region residues 89-91, RRA - furin cleavage site RRRR 59 region
arm region LNAIRKLHVGKVGENGYVEIEDDIGRRAEMNE 60
LMEQTSEIITFAESGTARKTLHFEISKEGSDLSV
VERAEVWLFLKVPKANRTRTKVTIRLFQQQKH
PQGSLDTGEEAEEVGLKGERSELLLSEKVVDA
RKSTWHVFPVSSSIQRLLDQGKSSLDVRIACEQ
CQESGASLVLLGKKKKKEEEGEGKKKGGGEG
GAGADEEKEQSHRPFLMLQARQSEDHPHRRR
RR
fingers region 1 KKQFFVSFKDIGWNDWIIAPSGYHANYC 61 fingers region 2 CVPTKLRPMSMLYYDDGQNIIKKDIQNMIVEE 62
CGCS
latency loop LKKRPDVTQPVPKAALL 63 alpha 1 helical ALA ALPKD VPNS QPEM VE A VKKHILNML 64 region
bowtie region QES GASL VLLGKKKKKEEEGEGKKKGGGEGG 65
AG
[00198] Growth factor domains among TGF-β family members are more highly conserved while prodomains comprise a much lower percent identity among family members (FIG. 5). Prodomains may vary in length from about 50 to about 200, from about 100 to about 400 or from about 300 to about 500 amino acids residues. In some embodiments, prodomains range from about 169 to about 433 residues. Prodomains may be unrelated in sequence and/or low in homology. Some prodomains may have similar folds and/or three dimensional structures. Prodomains of TGF-β family members may comprise latency loops. Such loops may be proline-rich. Latency loop length may determine the ability of such loops to encircle growth factor finger regions.
[00199] In some embodiments, protein modules from some TGF-β family members comprise low sequence identity with protein modules from other TGF-β family members. Such low sequence identity may indicate specialized roles for such family members with distinct protein modules.
[00200] Association of GPCs with extracellular proteins may strengthen prodomain-growth factor interactions. In some embodiments, such extracellular proteins may include, but are not limited to LTBPs, fibrillins, GASPs, perlecan and/or decorin. In some cases, extracellular protein associations are required to keep growth factors latent in GPCs.
[00201] GARP expression has been shown to be required for surface expression of GPCs on the surface of cells of hematopoietic origin (Tran, D.Q. et al., GARP (LRRC32) is essential for the surface expression of latent TGF-β on platelets and activated FOXP3+ regulatory T cells. PNAS. 2009, Jun 2. 106(32): 13445-50). GARP may act as a tether to hold GPCs in place on the surface of these cells, including, but not limited to regulatory T-cells and/or platelets.
[00202] In some embodiments, recombinant proteins of the present disclosure may comprise bone morphogenetic proteins (BMPs), a family of TGF-P-related proteins. Protein modules comprising sequences from BMPs may comprise sequences from any of those BMP modules disclosed in FIG. 6. While related to other TGF-β family member proteins, BMPs generally signal through SMAD1, 5 and 8 proteins while TGF-β isoforms (e.g. TGF-βΙ, TGF-P2 and TGF-P3) signal through SMAD2 and SMAD3.
[00203] Some BMP receptors and/or co-receptors are also distinct from other TGF-β family member proteins. Among these is the repulsive guidance molecule (RGM) family of proteins. RGM proteins act as co-receptors for BMP signaling. There are three RGM family members, RGMA, RGMB and RGMC [also known as hemojuvelin (Hjv)]. Recombinant proteins of the present disclosure comprising one or more BMP protein module may be useful for the development of antibodies and/or assays to study, enhance and/or perturb BMP interactions with RGM proteins.
[00204] Another family of GDF/BMP interacting proteins is C-terminal cysteine knot-like (CTCK) domain-containing proteins. In some cases, CTCK domain-containing proteins may act antagonistically with regard to GDF/BMP signal transduction. CTCK domain-containing proteins include, but are not limited to Cerberus, Connective tissue growth factor (CTGF), DAN domain family member 5 (DAND5), Gremlin- 1 (GREM1), Gremlin-2 (GREM2), Mucin-19 (MUC19), Mucin-2 (MUC2), Mucin-5AC (MUC5AC), Mucin-5B (MUC5B), Mucin-6 (MUC6), Neuroblastoma suppressor of tumorigenicity 1 (NBL1), Norrin (NDP), Otogelin (OTOG), Otogelin-like protein (OTOGL), Protein CYR61 (CYR61), Protein NOV homolog (NOV), Sclerostin (SOST), Sclerostin domain-containing protein 1 (SOSTDC1), SCO-spondin (SSPO), Slit homolog 1 protein (SLIT1), Slit homolog 2 protein (SLIT2), Slit homolog 3 protein (SLIT3), von Willebrand factor (VWF), WNT1 -inducible- signaling pathway protein 1 (WISP1) and WNT1 -inducible- signaling pathway protein 3 (WISP3).
Recombinant proteins
[00205] In some embodiments, the present disclosure provides recombinant proteins. As used herein, the term "recombinant protein" refers to a protein produced by an artificial gene and/or process (e.g. genetic engineering). Such recombinant proteins may comprise one or more protein modules from one or more TGF-P-related proteins. Some recombinant proteins disclosed herein may be useful as recombinant antigens. As used herein, the term "recombinant antigen" refers to a recombinant protein that may be used to immunize one or more hosts for the production of antibodies directed toward one or more epitopes present on such recombinant antigens. Some recombinant antigens may be cell-based antigens. As used herein, the term "cell-based antigen" refers to recombinant antigens that are expressed in cells for presentation of such antigens on the cell surface. Such cells may be used to immunize hosts for the production of antibodies directed to such cell-based antigens.
[00206] In some embodiments, recombinant proteins disclosed herein may be used as therapeutics. Recombinant proteins disclosed herein may modulate growth factor (e.g. growth factors comprising TGF-P-related proteins) levels and/or activity (e.g. signaling) upon administration and/or introduction to one or more subjects and/or niches.
[00207] In some embodiments, recombinant proteins disclosed herein may be used to assay growth factor (e.g. growth factors comprising TGF-P-related proteins) levels and/or activity (e.g. signaling). Some recombinant proteins disclosed herein may be used in the isolation of antibodies directed to TGF-P-related proteins. Recombinant proteins of the present disclosure may also be used as recombinant antigens in the development of stabilizing [reducing or preventing dissociation between two agents, (e.g. growth-factor release from GPCs, GPC release from one or more protein interactions)] and/or releasing [enhancing the dissociation between two agents (e.g. growth-factor release from GPCs, GPC release from one or more protein interactions)] antibodies. Recombinant proteins of the present disclosure may include TGF-β family member proteins as well as components and/or protein modules thereof. Some recombinant proteins of the present disclosure may comprise prodomains without associated growth factors, furin cleavage-deficient mutants, mutants deficient in extracellular protein associations and/or combinations thereof.
[00208] In some embodiments, recombinant proteins may comprise detectable labels.
Detectable labels may be used to allow for detection and/or isolation of recombinant proteins. Some detectable labels may comprise biotin labels, polyhistidine tags and/or flag tags. Such tags may be used to isolate tagged proteins. Proteins produced may comprise additional amino acids encoding one or more 3C protease cleavage site. Such sites allow for cleavage at the 3C protease cleavage site upon treatment with 3C protease, including, but not limited to rhinovirus 3C protease. Such cleavage sites are introduced to allow for removal of detectable labels from recombinant proteins.
Recombinant GPCs [00209] FIG. 7 is a schematic depicting an embodiment of a recombinant GPC.
Recombinant proteins according to FIG. 7 comprising TGF-P-family member proteins may comprise features including, but not limited to C-terminal regions of the mature growth factor, N-terminal regions of the prodomain and/or proprotein cleavage sites. The proprotein cleavage site of recombinant GPCs may, for example, comprise the furin consensus sequences RXXR, or RXR wherein R is arginine and X indicates amino acid residues that may vary among TGF-β family members. Furin cleavage site sequences (although not limited to cleavage by furin alone and may include cleavage by other proprotein convertase enzymes) for each TGF-β family member are indicated in Table 1. In some cases, prodomains of GPCs may comprise one or more cysteine residues within and/or near the N-terminal region of the prodomain. Such cysteine residues may be from about 1 to about 10 amino acids, from about 4 to about 15 amino acids, from about 5 to about 20 amino acids and/or from about 7 to about 50 amino acids from the N-terminus of the prodomain. In some cases, recombinant GPCs may also be expressed with detectable labels. Such detectable labels may be useful for detection and/or isolation of recombinant GPCs. Detectable labels may comprise 2 or more histidine (His) residues. Such detectable labels may also be referred to herein as polyhistidine tags. Polyhistidine tags may include hexa histidine tags or HIS-TAG™ (EMD Biosciences, Darmstadt, Germany) comprising a chain of six histidine residues. Some polyhistidine tags may be present at the N-terminus of recombinant proteins disclosed herein. Some
polyhistidine tags may be present at the C-terminus of recombinant proteins disclosed herein. Proteins produced may comprise additional amino acids encoding one or more 3C protease cleavage site. Such sites allow for cleavage at the 3C protease cleavage site upon treatment with 3C protease, including, but not limited to rhinovirus 3C protease. Some cleavage sites may be introduced to allow for removal of detectable labels from recombinant proteins.
[00210] In some embodiments of the present disclosure, recombinant GPCs may comprise mutations in one or more amino acids as compared to wild type sequences. In some cases, one or more regions of proteolytic processing may be mutated (see FIG. 8). Such regions may comprise proprotein convertase cleavage sites. Proprotein convertase (e.g. furin) cleavage site mutations prevent enzymatic cleavage at that site and/or prevent enzymatic cleavage of growth factors from their prodomains. Some proprotein convertase cleavage sites comprising RXXR sequences may be mutated to RXG (wherein X indicates a site where amino acid residues may be variable). Such mutations are herein abbreviated as "D2G" mutations and may be resistant to enzymatic cleavage. In some embodiments, furin cleavage sites comprising RXXR sequences are mutated to AXXA. Such AXXA sequences may also be resistant to enzymatic cleavage.
[00211] In some embodiments, regions of proteolytic processing by tolloid and/or tolloid- like proteins may be mutated to prevent such proteolytic processing. In some embodiments, tolloid processing regions on GDF-8 and/or GDF-11 may be mutated. In some embodiments, mutation of aspartic acid residues to alanine residues within tolloid processing regions prevents tolloid processing. Mutation of aspartic acid residue 76 (D76) of the GDF-8 (myostatin) proprotein has been shown to prevent proteolytic activation of latent GDF-8 (Wolfman, N.M. et al.,PNAS. 2003, Oct 6. 100(26): 15842-6). In some embodiments, Asp 120 (D120, residue number counted from the translated protein, D98 from the proprotein of SEQ ID NO: 4) in GDF-11 may be mutated to prevent tolloid processing (Ge et al., 2005. Mol Cell Biol. 25(14):5846-58, the contents of which are herein incorporated by reference in their entirety).
[00212] In some embodiments, one or more amino acids may be mutated in order to form recombinant GPCs with reduced latency. Such mutations are referred to herein as "activating mutations." These mutations may introduce one or more regions of steric clash between complex prodomains and growth factor domains. As used herein, the term "steric clash," when referring to the interaction between two proteins or between two domains and/or epitopes within the same protein, refers to a repulsive interaction between such proteins, domains and/or epitopes due to overlapping position in three-dimensional space. Steric clash within GPCs may reduce the affinity between prodomains and growth factor domains, resulting in elevated ratios of free growth factor to latent growth factor. In some
embodiments, one or more amino acids may be mutated in order to form recombinant GPCs with increased latency. Such mutations are referred to herein as "stabilizing mutations." These mutations may increase the affinity between prodomains and growth factor domains, resulting in decreased ratios of free growth factor to latent growth factor.
Recombinant growth differentiation factors (GDFs), activins and inhibins
[00213] Growth differentiation factors (GDFs), activins and inhibins are TGF-β family member proteins involved in a number of cellular and/or developmental activities. In some embodiments of the present disclosure, recombinant proteins may comprise one or more protein modules from one or more GDFs, activins and/or inhibins. In further embodiments, GDF protein modules may comprise GDF-8 and/or GDF-11 protein modules. [00214] GDF-8 and GDF-11, which are secreted as latent complexes (Sengle et al., 2011. J Biol Chem. 286(7):5087-99; Ge et al., 2005. Mol Cel Biol. 25(14):5846-58), show
conservation of the fastener residues (Lys 27 and Tyr 75 of TGF-βΙ). GDF-8 (also referred to herein as myostatin) is involved in regulating muscle mass, and its deficiency increases muscle mass in multiple species, including humans (Rodino-Klapac, L.R. et al., 2009. Muscle Nerve. 39(3):283-96). GDF-8 may be found in the circulation in latent form, but may also be stored in the extracellular matrix, bound to LTBP3 (Anderson et al., 2007. J Biol Chem. 283(11):7027-35) or perlecan (Sengle et al., 2011. J Biol Chem. 286(7):5087-99). While complexed with its prodomain, GDF-8 is unable to participate in receptor binding with the type II receptor, ActRIIB (Sengle et al., 2008. J Mol Biol. 381(4): 1025-39). While GDF-8 is expressed primarily in muscle, GDF-11 expression is more systemic and its activity is thought to be involved in multiple processes (Lee et al., 2013. PNAS. 110(39):E3713-22). It is believed to be involved in development of multiple tissues, including, but not limited to the retina, kidney, pancreas and olfactory system. It is also believed to be a circulating factor in the blood. Recent studies indicate that GDF-11 may rejuvenate skeletal muscle, improve cerebral circulation and promote neurogenesis (Sinha, M. et al., 2014. Science.
344(6184):649-52 and Katsimpardi, L. et al., 2014. Science. 344(6184):630-4, the contents of each of which are herein incorporated by reference in their entirety). In some cases, antibodies of the disclosure may promote skeletal muscle rejuvenation, improve cerebral circulation and promote neurogenesis by promoting the release of GDF- 11 growth factor from latent complexes. GDF-11 has also been identified as a regulator of erythropoiesis. In some cases, antibodies of the invation that inhibit GDF-11 may restore the ability of erythroid progenitors to differentiate and hence alleviate anemia (Paulson, RF. 2014. Nature Medicine. 20:334-5, the contents of which are herein incorporated by reference in their entirety).
[00215] GDF-8 and GDF-11 also share considerable homology. While the prodomains only share 48% homology, GDF-8 and GDF-11 growth factor domains share 90% homology (60% homology when prodomains and growth factors are taken together). Given the high degree of sequence similarity, it is not surprising that GDF- 11 and 8 growth factors bind and signal through the same receptors consisting of a Type I receptor (ALK4/5) in association with a type II receptor (ACTRIIA/B). The high degree of conservation in the mature growth factors has made it challenging to identify reagents and monoclonal antibodies that can differentiate between mature GDF-11 and 8. Consequently, there are no therapies in clinical trials today that are specific for GDF-11. [00216] Expressed proGDF proteins require furin cleavage in order to form latent complexes (see FIG. 9). In some cases, furin cleavage is carried out by cells in which proGDF proteins are expressed. In some cases, furin cleavage may be carried out through furin addition to cultures or solutions comprising proGDF proteins. Furin cleavage of recombinant proteins of the disclosure may in some cases occur intracellularly. In some cases furin cleavage of recombinant proteins of the disclosure may occur extracellularly. Release of GDF-8 and GDF-11 from latent GPCs requires activating cleavage of latent complexes. An activating cleavage refers to a proteolytic cleavage of a latent complex that leads to growth factor release and/or activation. Activating cleavage may include prodomain cleavage at a BMP/tolloid/tolloid-like proteinase cleavage site. Such sites may include the site located between Arg 75 and Asp 76 in GDF-8 and between Gly 97 and Asp 98 in GDF-11. This cleavage is between the 2 helix and the fastener.
[00217] In some embodiments, recombinant proteins of the present disclosure comprising GDFs may comprise sequences listed in Table 4 or fragments thereof. In some cases, these sequences are expressed in association with N- and/or C-terminal secretion signal sequences [e.g. human Ig kappa chains with amino acid sequence MDMRVPAQLLGLLLLWFSGVLG (SEQ ID NO: 66)], flag tag sequences [e.g. DYKDDDDK (SEQ ID NO: 67)], one or more 3C protease cleavage site [e.g. LEVLFQGP (SEQ ID NO: 68)], one or more biotinylation site and/or His-tag sequences [e.g. HHHHHH (SEQ ID NO: 69)].
Table 4. Recombinant GDFs
Protein Sequence SEQ
ID NO
proGDF-8 NENSEQKENVEKEGLCNACTWRQNTKSSRIEAIKIQILSKLRLE 5
TAPNISKDVIRQLLPKAPPLRELIDQYDVQRDDSSDGSLEDDDY
HATTETIITMPTESDFLMQVDGKPKCCFFKFSSKIQYNKVVKA
QLWIYLRPVETPTTVFVQILRLIKPMKDGTRYTGIRSLKLDMNP
GTGIWQSIDVKTVLQNWLKQPESNLGIEIKALDENGHDLAVTF
PGPGEDGLNPFLEVKVTDTPKRSRRDFGLDCDEHSTESRCCRY
PLTVDFEAFGWDWIIAPKRYKANYCSGECEFVFLQKYPHTHL
VHQANPRGSAGPCCTPTKMSPINMLYFNGKEQIIYGKIPAMVV
DRCGCS
GDF-8 prodomain NENSEQKENVEKEGLCNACTWRQNTKSSRIEAIKIQILSKLRLE 38
TAPNISKDVIRQLLPKAPPLRELIDQYDVQRDDSSDGSLEDDDY
HATTETIITMPTESDFLMQVDGKPKCCFFKFSSKIQYNKVVKA
QLWIYLRPVETPTTVFVQILRLIKPMKDGTRYTGIRSLKLDMNP
GTGIWQSIDVKTVLQNWLKQPESNLGIEIKALDENGHDLAVTF
PGPGEDGLNPFLEVKVTDTPKRSRR
GDF-8 prodomain NENSEQKENVEKEGLCNACTWRQNTKSSRIEAIKIQILSKLRLE 70 D76A TAPNISKDVIRQLLPKAPPLRELIDQYDVQRADSSDGSLEDDDY
HATTETIITMPTESDFLMQVDGKPKCCFFKFSSKIQYNKVVKA QLWIYLRPVETPTTVFVQILRLIKPMKDGTRYTGIRSLKLDMNP
GTGIWQSIDVKTVLQNWLKQPESNLGIEIKALDENGHDLAVTF PGPGEDGLNPFLEVKVTDTPKRSRR
proGDF-8 AXXA NENSEQKENVEKEGLCNACTWRQNTKSSRIEAIKIQILSKLRLE 71
TAPNISKDVIRQLLPKAPPLRELIDQYDVQRDDSSDGSLEDDDY
HATTETIITMPTESDFLMQVDGKPKCCFFKFSSKIQYNKVVKA
QLWIYLRPVETPTTVFVQILRLIKPMKDGTRYTGIRSLKLDMNP
GTGIWQSIDVKTVLQNWLKQPESNLGIEIKALDENGHDLAVTF
PGPGEDGLNPFLEVKVTDTPKASRADFGLDCDEHSTESRCCRY
PLTVDFEAFGWDWIIAPKRYKANYCSGECEFVFLQKYPHTHL
VHQANPRGSAGPCCTPTKMSPINMLYFNGKEQIIYGKIPAMVV
DRCGCS
proGDF-8 D76A NENSEQKENVEKEGLCNACTWRQNTKSSRIEAIKIQILSKLRLE 72
TAPNISKDVIRQLLPKAPPLRELIDQYDVQRADSSDGSLEDDDY
HATTETIITMPTESDFLMQVDGKPKCCFFKFSSKIQYNKVVKA
QLWIYLRPVETPTTVFVQILRLIKPMKDGTRYTGIRSLKLDMNP
GTGIWQSIDVKTVLQNWLKQPESNLGIEIKALDENGHDLAVTF
PGPGEDGLNPFLEVKVTDTPKRSRRDFGLDCDEHSTESRCCRY
PLTVDFEAFGWDWIIAPKRYKANYCSGECEFVFLQKYPHTHL
VHQANPRGSAGPCCTPTKMSPINMLYFNGKEQIIYGKIPAMVV
DRCGCS
proGDF-8 AXXA NENSEQKENVEKEGLCNACTWRQNTKSSRIEAIKIQILSKLRLE 73 D76A TAPNISKDVIRQLLPKAPPLRELIDQYDVQRADSSDGSLEDDDY
HATTETIITMPTESDFLMQVDGKPKCCFFKFSSKIQYNKVVKA
QLWIYLRPVETPTTVFVQILRLIKPMKDGTRYTGIRSLKLDMNP
GTGIWQSIDVKTVLQNWLKQPESNLGIEIKALDENGHDLAVTF
PGPGEDGLNPFLEVKVTDTPKASRADFGLDCDEHSTESRCCRY
PLTVDFEAFGWDWIIAPKRYKANYCSGECEFVFLQKYPHTHL
VHQANPRGSAGPCCTPTKMSPINMLYFNGKEQIIYGKIPAMVV
DRCGCS
proGDF-11 AEGPAAAAAAAAAAAAAGVGGERSSRPAPSVAPEPDGCPVC 4
VWRQHSRELRLESIKSQILSKLRLKEAPNISREVVKQLLPKAPP
LQQILDLHDFQGDALQPEDFLEEDEYHATTETVISMAQETDPA
VQTDGSPLCCHFHFSPKVMFTKVLKAQLWVYLRPVPRPATVY
LQILRLKPLTGEGTAGGGGGGRRHIRIRSLKIELHSRSGHWQSI
DFKQVLHSWFRQPQSNWGIEINAFDPSGTDLAVTSLGPGAEGL
HPFMELRVLENTKRSRRNLGLDCDEHSSESRCCRYPLTVDFEA
FGWDWIIAPKRYKANYCSGQCEYMFMQKYPHTHLVQQANPR
GSAGPCCTPTKMSPINMLYFNDKQQIIYGKIPGMVVDRCGCS
proGDF-11 D98A AEGPAAAAAAAAAAAAAGVGGERSSRPAPSVAPEPDGCPVC 74
VWRQHSRELRLESIKSQILSKLRLKEAPNISREVVKQLLPKAPP
LQQILDLHDFQGAALQPEDFLEEDEYHATTETVISMAQETDPA
VQTDGSPLCCHFHFSPKVMFTKVLKAQLWVYLRPVPRPATVY
LQILRLKPLTGEGTAGGGGGGRRHIRIRSLKIELHSRSGHWQSI
DFKQVLHSWFRQPQSNWGIEINAFDPSGTDLAVTSLGPGAEGL
HPFMELRVLENTKRSRRNLGLDCDEHSSESRCCRYPLTVDFEA
FGWDWIIAPKRYKANYCSGQCEYMFMQKYPHTHLVQQANPR
GSAGPCCTPTKMSPINMLYFNDKQQIIYGKIPGMVVDRCGCS
proGDF-11 D2G AEGPAAAAAAAAAAAAAGVGGERSSRPAPSVAPEPDGCPVC 75
VWRQHSRELRLESIKSQILSKLRLKEAPNISREVVKQLLPKAPP
LQQILDLHDFQGDALQPEDFLEEDEYHATTETVISMAQETDPA
VQTDGSPLCCHFHFSPKVMFTKVLKAQLWVYLRPVPRPATVY
LQILRLKPLTGEGTAGGGGGGRRHIRIRSLKIELHSRSGHWQSI
DFKQVLHSWFRQPQSNWGIEINAFDPSGTDLAVTSLGPGAEGL HPFMELRVLENTKRSGNLGLDCDEHSSESRCCRYPLTVDFEAF
GWDWIIAPKRYKANYCSGQCEYMFMQKYPHTHLVQQANPRG SAGPCCTPTKMSPINMLYFNDKQQIIYGKIPGMVVDRCGCS
proGDF-11 AxxA AEGPAAAAAAAAAAAAAGVGGERSSRPAPSVAPEPDGCPVC 76
VWRQHSRELRLESIKSQILSKLRLKEAPNISREVVKQLLPKAPP
LQQILDLHDFQGDALQPEDFLEEDEYHATTETVISMAQETDPA
VQTDGSPLCCHFHFSPKVMFTKVLKAQLWVYLRPVPRPATVY
LQILRLKPLTGEGTAGGGGGGRRHIRIRSLKIELHSRSGHWQSI
DFKQVLHSWFRQPQSNWGIEINAFDPSGTDLAVTSLGPGAEGL
HPFMELRVLENTKASRANLGLDCDEHSSESRCCRYPLTVDFEA
FGWDWIIAPKRYKANYCSGQCEYMFMQKYPHTHLVQQANPR
GSAGPCCTPTKMSPINMLYFNDKQQIIYGKIPGMVVDRCGCS
proGDF-11 AxxA AEGPAAAAAAAAAAAAAGVGGERSSRPAPSVAPEPDGCPVC 77 D98A VWRQHSRELRLESIKSQILSKLRLKEAPNISREVVKQLLPKAPP
LQQILDLHDFQGAALQPEDFLEEDEYHATTETVISMAQETDPA
VQTDGSPLCCHFHFSPKVMFTKVLKAQLWVYLRPVPRPATVY
LQILRLKPLTGEGTAGGGGGGRRHIRIRSLKIELHSRSGHWQSI
DFKQVLHSWFRQPQSNWGIEINAFDPSGTDLAVTSLGPGAEGL
HPFMELRVLENTKASRANLGLDCDEHSSESRCCRYPLTVDFEA
FGWDWIIAPKRYKANYCSGQCEYMFMQKYPHTHLVQQANPR
GSAGPCCTPTKMSPINMLYFNDKQQIIYGKIPGMVVDRCGCS
GDF-11 AEGPAAAAAAAAAAAAAGVGGERSSRPAPSVAPEPDGCPVC 78 prodomain D98A VWRQHSRELRLESIKSQILSKLRLKEAPNISREVVKQLLPKAPP
LQQILDLHDFQGAALQPEDFLEEDEYHATTETVISMAQETDPA
VQTDGSPLCCHFHFSPKVMFTKVLKAQLWVYLRPVPRPATVY
LQILRLKPLTGEGTAGGGGGGRRHIRIRSLKIELHSRSGHWQSI
DFKQVLHSWFRQPQSNWGIEINAFDPSGTDLAVTSLGPGAEGL
HPFMELRVLENTKRSRR
GDF-11 AEGPAAAAAAAAAAAAAGVGGERSSRPAPSVAPEPDGCPVC 39 prodomain VWRQHSRELRLESIKSQILSKLRLKEAPNISREVVKQLLPKAPP
LQQILDLHDFQGDALQPEDFLEEDEYHATTETVISMAQETDPA
VQTDGSPLCCHFHFSPKVMFTKVLKAQLWVYLRPVPRPATVY
LQILRLKPLTGEGTAGGGGGGRRHIRIRSLKIELHSRSGHWQSI
DFKQVLHSWFRQPQSNWGIEINAFDPSGTDLAVTSLGPGAEGL
HPFMELRVLENTKRSRR
[00218] Activins and inhibins are TGF-β family member proteins, the activity of each of which often results in opposing functions (Bilezikjian, L.M. et al., 2012. Mol Cell
Endocrinol. 359(1-2): 43-52). Like other family members, these proteins occur
physiologically as dimers. Activins and inhibins are constructed in part from the same β- subunits, that may include inhibin-beta A, inhibin-beta B, inhibin-beta C and inhibin-beta E (referred to herein as β-subunit A, B, C and E, respectively). The difference between activins and inhibins, structurally, is that activins are β-subunit dimers while inhibins are
heterodimers, wherein the second subunit is inhibin-a. Activins are named for their subunit pairs, such that activin A comprises a homodimer of two A subunits, activin AB comprises a dimer of A and B subunits, B comprises a dimer of B subunits, etc. (Muenster, U. et al., 2011. Vitam Horm. 85: 105-28). Activins are involved in a variety of functions that may include, but are not limited to cell growth, differentiation, programmed cell death, endocrine functions, cellular metabolism, bone growth, etc. They are especially recognized for their control of reproductive hormone cycles. Activin and inhibin signaling often functions antagonistically in this regard.
[00219] In some embodiments, recombinant proteins of the present disclosure may comprise integrins. Integrins are cell surface heterodimers formed by alpha and beta subunits, each of which has a transmembrane domain and in the N-terminal portion of the extracellular domain come together to form the ligand binding site. Recombinant proteins of the present disclosure may comprise integrins and/or integrin subunits. Such integrins and/or integrin subunits may comprise any of those disclosed in International Patent Application No.
WO2014074532, the contents of which are herein incorporated by reference in their entirety.
[00220] Recombinant proteins of the disclosure may include intercellular adhesion molecule 1 (ICAM-1). In some cases, ICAM-1 proteins of the present disclosure may be used as control proteins during antibody development and/or antibody testing. In some cases, ICAM-1 may be used as a control during selection of binding molecules using phage display technologies. In some cases, ICAM-1 proteins of the disclosure comprise one or more detectable label. Detectable labels may include, for example, histidine tags.
[00221] In some embodiments, recombinant GPCs of the present disclosure may comprise mutations in one or more N-terminal regions for extracellular associations. As used herein, the term "N-terminal region for extracellular association" refers to regions at or near protein N-termini that may be necessary for extracellular associations with one or more N-terminal regions. Such regions may comprise at least the first N-terminal residue, at least the first 5 N- terminal residues, at least the first 10 N-terminal residues, at least the first 20 amino acid residues and/or at least the first 50 amino acid residues. Some mutations may comprise from about 1 amino acid residue to about 30 amino acid residues, from about 5 amino acid residues to about 40 amino acid residues and/or from about 10 amino acid residues to about 50 amino acid residues at or near protein N-termini. Such regions may comprise residues for association with extracellular proteins (e.g. LTBPs, fibrillins, GASPs, perlecan and/or decorin). In some cases, one or more cysteine residues present within and/or near N-terminal regions for extracellular associations may be necessary for such associations. In some embodiments, cysteine residues present within and/or near N-terminal regions for
extracellular associations are present within about the first 5 N-terminal residues, about the first 10 N-terminal residues, about the first 20 N-terminal residues, about the first 50 N- terminal residues, about the first 60 N-terminal residues, about the first 70 N-terminal residues and/or at least the first 100 N-terminal residues. Some mutations in one or more N- terminal regions for extracellular associations comprise substitution and/or deletion of such cysteine residues. Such mutations may modulate the association of GPCs and/or prodomains with one or more extracellular proteins, including, but not limited to LTBPs, fibrillins, GASPs, perlecan and/or decorin. These mutations may also comprise substitution of one or more cysteine with another amino acid. Cysteine residue substitutions are abbreviated herein as "C#X" wherein # represents the residue number [counting from the N-terminus of the pro- protein (without the signal peptide)] of the original cysteine residue and X represents the one letter amino acid code for the amino acid that is used for substitution. Any amino acid may be used for such substitutions. In some cases, serine (S) residues are used to substitute cysteine residues. Nonlimiting examples of such mutations in GDF-8 may include C16S, C19S, C38S and/or C41S.
[00222] In some cases, one or more cysteine in one or more other region of GPCs may be substituted or deleted. In some embodiments, such GPC modifications may promote the release of mature growth factor from prodomains. In some cases, such cysteines or other residues may include those present in one or more of mature growth factors, alpha 2 helices, fasteners, latency lassos and/or bow-tie regions.
[00223] In some embodiments, recombinant proteins of the present disclosure may comprise protein modules derived from one or more species, including mammals, including, but not limited to mice, rats, rabbits, pigs, monkeys and/or humans. Recombinant proteins may comprise one or more amino acids from one or more amino acid sequences derived from one or more non-human protein sequences listed in Table 5. In some cases, recombinant proteins of the present disclosure may comprise such sequences with or without the native signal peptide. In some cases, these sequences are expressed in association with N- and/or C- terminal secretion signal sequences [e.g. human Ig kappa chains with amino acid sequence MDMRVPAQLLGLLLLWFSGVLG (SEQ ID NO: 66)], flag tag sequences [e.g.
DYKDDDDK (SEQ ID NO: 67)], one or more 3C protease cleavage site [e.g. LEVLFQGP (SEQ ID NO: 68)], one or more biotinylation site and/or His-tag sequences [e.g. HHHHHH (SEQ ID NO: 69)].
Table 5. Non-human proteins
Protein Species Sequence SEQ
ID NO
proTGF-βΙ Mouse LSTCKTIDMELVKRKRIEAIRGQILSKLRLASPPSQGEVPP 79
GPLPEAVLALYNSTRDRVAGESADPEPEPEADYYAKEV TRVLMVDRNNAIYEKTKDISHSIYMFFNTSDIREAVPEPP
LLSRAELRLQRLKSSVEQHVELYQKYSNNSWRYLGNRL
LTPTDTPEWLSFDVTGVVRQWLNQGDGIQGFRFSAHCS
CDSKDNKLHVEINGISPKRRGDLGTIHDMNRPFLLLMAT
PLERAQHLHS SRHRRALDTNYCFS STEKNCC VRQL YIDF
RKDLGWKWIHEPKGYHANFCLGPCPYIWSLDTQYSKVL
ALYNQHNPGASASPCCVPQALEPLPIVYYVGRKPKVEQ
LSNMIVRSCKCS
proTGF-βΙ Cyno LSTCKTIDMELVKRKRIEAIRGQILSKLRLASPPSQGEVPP 80
GPLPEAVLALYNSTRDRVAGESAEPEPEPEADYYAKEVT
RVLMVETHNEIYDKFKQSTHSIYMFFNTSELREAVPEPV
LLSRAELRLLRLKLKVEQHVELYQKYSNNSWRYLSNRL
LAPSDSPEWLSFDVTGVVRQWLSRGGEIEGFRLSAHCSC
DSKDNTLQVDINGFTTGRRGDLATIHGMNRPFLLLMAT
PLERAQHLQS SRHRRALDTNYCFS STEKNCC VRQL YIDF
RKDLGWKWIHEPKGYHANFCLGPCPYIWSLDTQYSKVL
ALYNQHNPGASAAPCCVPQALEPLPIVYYVGRKPKVEQ
LSNMIVRSCKCS
TGF-βΙ LAP Mouse LSTSKTIDMELVKRKRIEAIRGQILSKLRLASPPSQGEVPP 81 C4S GPLPEAVLALYNSTRDRVAGESADPEPEPEADYYAKEV
TRVLMVDRNNAIYEKTKDISHSIYMFFNTSDIREAVPEPP
LLSRAELRLQRLKSSVEQHVELYQKYSNNSWRYLGNRL
LTPTDTPEWLSFDVTGVVRQWLNQGDGIQGFRFSAHCS
CDSKDNKLHVEINGISPKRRGDLGTIHDMNRPFLLLMAT
PLERAQHLHS SRHRR
TGF-βΙ LAP Cyno LSTSKTIDMELVKRKRIEAIRGQILSKLRLASPPSQGEVPP 82 C4S GPLPEAVLALYNSTRDRVAGESAEPEPEPEADYYAKEVT
RVLMVETHNEIYDKFKQSTHSIYMFFNTSELREAVPEPV
LLSRAELRLLRLKLKVEQHVELYQKYSNNSWRYLSNRL
LAPSDSPEWLSFDVTGVVRQWLSRGGEIEGFRLSAHCSC
DSKDNTLQVDINGFTTGRRGDLATIHGMNRPFLLLMAT
PLERAQHLQS SRHRR
proTGF-βΙ Mouse LSTSKTIDMELVKRKRIEAIRGQILSKLRLASPPSQGEVPP 83 C4S D2G GPLPEAVLALYNSTRDRVAGESADPEPEPEADYYAKEV
TRVLMVDRNNAIYEKTKDISHSIYMFFNTSDIREAVPEPP
LLSRAELRLQRLKSSVEQHVELYQKYSNNSWRYLGNRL
LTPTDTPEWLSFDVTGVVRQWLNQGDGIQGFRFSAHCS
CDSKDNKLHVEINGISPKRRGDLGTIHDMNRPFLLLMAT
PLERAQHLHS SRHGALDTNYCFS STEKNCC VRQL YIDFR
KDLGWKWIHEPKGYHANFCLGPCPYIWSLDTQYSKVLA
LYNQHNPGASASPCCVPQALEPLPIVYYVGRKPKVEQLS
NMIVRSCKCS
proTGF-βΙ Mouse LSTSKTIDMELVKRKRIEAIRGQILSKLRLASPPSQGEVPP 84 C4S GPLPEAVLALYNSTRDRVAGESADPEPEPEADYYAKEV
TRVLMVDRNNAIYEKTKDISHSIYMFFNTSDIREAVPEPP
LLSRAELRLQRLKSSVEQHVELYQKYSNNSWRYLGNRL
LTPTDTPEWLSFDVTGVVRQWLNQGDGIQGFRFSAHCS
CDSKDNKLHVEINGISPKRRGDLGTIHDMNRPFLLLMAT
PLERAQHLHS SRHRRALDTNYCFS STEKNCC VRQL YIDF
RKDLGWKWIHEPKGYHANFCLGPCPYIWSLDTQYSKVL
ALYNQHNPGASASPCCVPQALEPLPIVYYVGRKPKVEQ
LSNMIVRSCKCS
proTGF-βΙ Cyno LSTSKTIDMELVKRKRIEAIRGQILSKLRLASPPSQGEVPP 85 C4S GPLPEAVLALYNSTRDRVAGESAEPEPEPEADYYAKEVT RVLMVETHNEIYDKFKQSTHSIYMFFNTSELREAVPEPV
LLSRAELRLLRLKLKVEQHVELYQKYSNNSWRYLSNRL
LAPSDSPEWLSFDVTGVVRQWLSRGGEIEGFRLSAHCSC
DSKDNTLQVDINGFTTGRRGDLATIHGMNRPFLLLMAT
PLERAQHLQS SRHRRALDTNYCFS STEKNCC VRQL YIDF
RKDLGWKWIHEPKGYHANFCLGPCPYIWSLDTQYSKVL
ALYNQHNPGASAAPCCVPQALEPLPIVYYVGRKPKVEQ
LSNMIVRSCKCS
proTGF-βΙ Cyno LSTSKTIDMELVKRKRIEAIRGQILSKLRLASPPSQGEVPP 86 C4S D2G GPLPEAVLALYNSTRDRVAGESAEPEPEPEADYYAKEVT
RVLMVETHNEIYDKFKQSTHSIYMFFNTSELREAVPEPV
LLSRAELRLLRLKLKVEQHVELYQKYSNNSWRYLSNRL
LAPSDSPEWLSFDVTGVVRQWLSRGGEIEGFRLSAHCSC
DSKDNTLQVDINGFTTGRRGDLATIHGMNRPFLLLMAT
PLERAQHLQS SRHGALDTNYCFS STEKNCC VRQL YIDFR
KDLGWKWIHEPKGYHANFCLGPCPYIWSLDTQYSKVLA
LYNQHNPGASAAPCCVPQALEPLPIVYYVGRKPKVEQL
SNMIVRSCKCS
LRRC32 Cyno AQHQDKVACKMVDKKVSCQGLGLLQVPLVLPPDTETL 87
DLSGNQLRSILASPLGFYTALRHLDLSTNEINFLQPGAFQ
ALTHLEHLSLAHNRLAMATALSAGGLGPLPRVTSLDLS
GNSLYSGLLERLLGEAPSLHTLSLAENSLTRLTRHTFRD
MPALEQLDLHSNVLMDIEDGAFEGLPHLTHLNLSRNSLT
CISDFSLQQLRVLDLSCNSIEAFQTASQPQAEFQLTWLDL
RENKLLHFPDLAALPRLIYLNLSNNLIRLPTGPPQDSKGI
HAPSEGWSALPLSTPNGNVSARPLSQLLNLDLSYNEIELI
PDSFLEHLTSLCFLNLSRNCLRTFEARRSGSLPCLMLLDL
SHNALETLELGARALGSLRTLLLQGNALRDLPPYTFANL
ASLQRLNLQGNRVSPCGGPNEPGPASCVAFSGIASLRSLS
LVDNEIELLRAGAFLHTPLTELDLSSNPGLEVATGALTGL
EASLEVLALQGNGLTVLQVDLPCFICLKRLNLAENRLSH
LPAWTQAVSLEVLDLRNNSFSLLPGSAMGGLETSLRRL
YLQGNPLSCCGNGWLAAQLHQGRVDVDATQDLICRFSS
QEEVSLSHVRPEDCEKGGLKNINLIIILTFILVSAILLTTLA
TCCCVRRQKFNQQYKA
proGDF-8 Mouse NEGSEREENVEKEGLCNACAWRQNTRYSRIEAIKIQILS 88
KLRLETAPNISKDAIRQLLPRAPPLRELIDQYDVQRDDSS
DGSLEDDDYHATTETIITMPTESDFLMQADGKPKCCFFK
FSSKIQYNKVVKAQLWIYLRPVKTPTTVFVQILRLIKPM
KDGTRYTGIRSLKLDMSPGTGIWQSIDVKTVLQNWLKQ
PESNLGIEIKALDENGHDLAVTFPGPGEDGLNPFLEVKV
TDTPKRSRRDFGLDCDEHSTESRCCRYPLTVDFEAFGW
DWIIAPKRYKANYCSGECEFVFLQKYPHTHLVHQANPR
GSAGPCCTPTKMSPINMLYFNGKEQIIYGKIPAMVVDRC
GCS
proGDF-8 Mouse NEGSEREENVEKEGLCNACAWRQNTRYSRIEAIKIQILS 89 AxxA KLRLETAPNISKDAIRQLLPRAPPLRELIDQYDVQRDDSS
DGSLEDDDYHATTETIITMPTESDFLMQADGKPKCCFFK
FSSKIQYNKVVKAQLWIYLRPVKTPTTVFVQILRLIKPM
KDGTRYTGIRSLKLDMSPGTGIWQSIDVKTVLQNWLKQ
PESNLGIEIKALDENGHDLAVTFPGPGEDGLNPFLEVKV
TDTPKASRADFGLDCDEHSTESRCCRYPLTVDFEAFGW
DWIIAPKRYKANYCSGECEFVFLQKYPHTHLVHQANPR
GSAGPCCTPTKMSPINMLYFNGKEQIIYGKIPAMVVDRC GCS
proGDF-8 Mouse NEGSEREENVEKEGLCNACAWRQNTRYSRIEAIKIQILS 90 D76A KLRLETAPNISKDAIRQLLPRAPPLRELIDQYDVQRADSS
DGSLEDDDYHATTETIITMPTESDFLMQADGKPKCCFFK
FSSKIQYNKVVKAQLWIYLRPVKTPTTVFVQILRLIKPM
KDGTRYTGIRSLKLDMSPGTGIWQSIDVKTVLQNWLKQ
PESNLGIEIKALDENGHDLAVTFPGPGEDGLNPFLEVKV
TDTPKRSRRDFGLDCDEHSTESRCCRYPLTVDFEAFGW
DWIIAPKRYKANYCSGECEFVFLQKYPHTHLVHQANPR
GSAGPCCTPTKMSPINMLYFNGKEQIIYGKIPAMVVDRC
GCS
proGDF-8 Mouse NEGSEREENVEKEGLCNACAWRQNTRYSRIEAIKIQILS 91 AxxA D76A KLRLETAPNISKDAIRQLLPRAPPLRELIDQYDVQRADSS
DGSLEDDDYHATTETIITMPTESDFLMQADGKPKCCFFK
FSSKIQYNKVVKAQLWIYLRPVKTPTTVFVQILRLIKPM
KDGTRYTGIRSLKLDMSPGTGIWQSIDVKTVLQNWLKQ
PESNLGIEIKALDENGHDLAVTFPGPGEDGLNPFLEVKV
TDTPKASRADFGLDCDEHSTESRCCRYPLTVDFEAFGW
DWIIAPKRYKANYCSGECEFVFLQKYPHTHLVHQANPR
GSAGPCCTPTKMSPINMLYFNGKEQIIYGKIPAMVVDRC
GCS
GDF-8 Mouse NEGSEREENVEKEGLCNACAWRQNTRYSRIEAIKIQILS 92 prodomain KLRLETAPNISKDAIRQLLPRAPPLRELIDQYDVQRDDSS
DGSLEDDDYHATTETIITMPTESDFLMQADGKPKCCFFK
FSSKIQYNKVVKAQLWIYLRPVKTPTTVFVQILRLIKPM
KDGTRYTGIRSLKLDMSPGTGIWQSIDVKTVLQNWLKQ
PESNLGIEIKALDENGHDLAVTFPGPGEDGLNPFLEVKV
TDTPKRSRR
GDF-8 Mouse NEGSEREENVEKEGLCNACAWRQNTRYSRIEAIKIQILS 93 prodomain KLRLETAPNISKDAIRQLLPRAPPLRELIDQYDVQRADSS
D76A DGSLEDDDYHATTETIITMPTESDFLMQADGKPKCCFFK
FSSKIQYNKVVKAQLWIYLRPVKTPTTVFVQILRLIKPM
KDGTRYTGIRSLKLDMSPGTGIWQSIDVKTVLQNWLKQ
PESNLGIEIKALDENGHDLAVTFPGPGEDGLNPFLEVKV
TDTPKRSRR
proGDF-8 Cyno NENSEQKENVEKEGLCNACTWRQNTKSSRIEAIKIQILSK 94
LRLETAPNISKDAIRQLLPKAPPLRELIDQYDVQRDDSSD
GSLEDDDYHATTETIITMPTESDFLMQVDGKPKCCFFKF
SSKIQYNKVVKAQLWIYLRPVETPTTVFVQILRLIKPMK
DGTRYTGIRSLKLDMNPGTGIWQSIDVKTVLQNWLKQP
ESNLGIEIKALDENGHDLAVTFPGPGEDGLNPFLEVKVT
DTPKRSRRDFGLDCDEHSTESRCCRYPLTVDFEAFGWD
WIIAPKRYKANYCSGECEFVFLQKYPHTHLVHQANPRG
SAGPCCTPTKMSPINMLYFNGKEQIIYGKIPAMVVDRCG
cs
proGDF-8 Cyno NENSEQKENVEKEGLCNACTWRQNTKSSRIEAIKIQILSK 95 AxxA LRLETAPNISKDAIRQLLPKAPPLRELIDQYDVQRDDSSD
GSLEDDDYHATTETIITMPTESDFLMQVDGKPKCCFFKF
SSKIQYNKVVKAQLWIYLRPVETPTTVFVQILRLIKPMK
DGTRYTGIRSLKLDMNPGTGIWQSIDVKTVLQNWLKQP
ESNLGIEIKALDENGHDLAVTFPGPGEDGLNPFLEVKVT
DTPKASRADFGLDCDEHSTESRCCRYPLTVDFEAFGWD
WIIAPKRYKANYCSGECEFVFLQKYPHTHLVHQANPRG
SAGPCCTPTKMSPINMLYFNGKEQIIYGKIPAMVVDRCG CS
proGDF-8 Cyno NENSEQKENVEKEGLCNACTWRQNTKSSRIEAIKIQILSK 96 D76A LRLETAPNISKDAIRQLLPKAPPLRELIDQYDVQRADSSD
GSLEDDDYHATTETIITMPTESDFLMQVDGKPKCCFFKF
SSKIQYNKVVKAQLWIYLRPVETPTTVFVQILRLIKPMK
DGTRYTGIRSLKLDMNPGTGIWQSIDVKTVLQNWLKQP
ESNLGIEIKALDENGHDLAVTFPGPGEDGLNPFLEVKVT
DTPKRSRRDFGLDCDEHSTESRCCRYPLTVDFEAFGWD
WIIAPKRYKANYCSGECEFVFLQKYPHTHLVHQANPRG
SAGPCCTPTKMSPINMLYFNGKEQIIYGKIPAMVVDRCG
CS
proGDF-8 Cyno NENSEQKENVEKEGLCNACTWRQNTKSSRIEAIKIQILSK 97 AxxA D76A LRLETAPNISKDAIRQLLPKAPPLRELIDQYDVQRADSSD
GSLEDDDYHATTETIITMPTESDFLMQVDGKPKCCFFKF
SSKIQYNKVVKAQLWIYLRPVETPTTVFVQILRLIKPMK
DGTRYTGIRSLKLDMNPGTGIWQSIDVKTVLQNWLKQP
ESNLGIEIKALDENGHDLAVTFPGPGEDGLNPFLEVKVT
DTPKASRADFGLDCDEHSTESRCCRYPLTVDFEAFGWD
WIIAPKRYKANYCSGECEFVFLQKYPHTHLVHQANPRG
SAGPCCTPTKMSPINMLYFNGKEQIIYGKIPAMVVDRCG
CS
GDF-8 Cyno NENSEQKENVEKEGLCNACTWRQNTKSSRIEAIKIQILSK 98 prodomain LRLETAPNISKDAIRQLLPKAPPLRELIDQYDVQRDDSSD
GSLEDDDYHATTETIITMPTESDFLMQVDGKPKCCFFKF
SSKIQYNKVVKAQLWIYLRPVETPTTVFVQILRLIKPMK
DGTRYTGIRSLKLDMNPGTGIWQSIDVKTVLQNWLKQP
ESNLGIEIKALDENGHDLAVTFPGPGEDGLNPFLEVKVT
DTPKRSRR
GDF-8 Cyno NENSEQKENVEKEGLCNACTWRQNTKSSRIEAIKIQILSK 99 prodomain LRLETAPNISKDAIRQLLPKAPPLRELIDQYDVQRADSSD
D76A GSLEDDDYHATTETIITMPTESDFLMQVDGKPKCCFFKF
SSKIQYNKVVKAQLWIYLRPVETPTTVFVQILRLIKPMK
DGTRYTGIRSLKLDMNPGTGIWQSIDVKTVLQNWLKQP
ESNLGIEIKALDENGHDLAVTFPGPGEDGLNPFLEVKVT
DTPKRSRR
proGDF-11 Mouse AEGPAAAAAAAAAAAGVGGERSSRPAPSAPPEPDGCPV 100
CVWRQHSRELRLESIKSQILSKLRLKEAPNISREVVKQLL
PKAPPLQQILDLHDFQGDALQPEDFLEEDEYHATTETVIS
MAQETDPAVQTDGSPLCCHFHFSPKVMFTKVLKAQLW
VYLRPVPRPATVYLQILRLKPLTGEGTAGGGGGGRRHIR
IRSLKIELHSRSGHWQSIDFKQVLHSWFRQPQSNWGIEIN
AFDPSGTDLAVTSLGPGAEGLHPFMELRVLENTKRSRRN
LGLDCDEHSSESRCCRYPLTVDFEAFGWDWIIAPKRYKA
NYCSGQCEYMFMQKYPHTHLVQQANPRGSAGPCCTPT
KMSPINMLYFNDKQQIIYGKIPGMVVDRCGCS
proGDF-11 Mouse AEGPAAAAAAAAAAAGVGGERSSRPAPSAPPEPDGCPV 101 AxxA CVWRQHSRELRLESIKSQILSKLRLKEAPNISREVVKQLL
PKAPPLQQILDLHDFQGDALQPEDFLEEDEYHATTETVIS
MAQETDPAVQTDGSPLCCHFHFSPKVMFTKVLKAQLW
VYLRPVPRPATVYLQILRLKPLTGEGTAGGGGGGRRHIR
IRSLKIELHSRSGHWQSIDFKQVLHSWFRQPQSNWGIEIN
AFDPSGTDLAVTSLGPGAEGLHPFMELRVLENTKASRA
NLGLDCDEHSSESRCCRYPLTVDFEAFGWDWIIAPKRYK
ANYCSGQCEYMFMQKYPHTHLVQQANPRGSAGPCCTP TKMSPINMLYFNDKQQIIYGKIPGMVVDRCGCS proGDF-11 Mouse AEGPAAAAAAAAAAAGVGGERSSRPAPSAPPEPDGCPV 102 AxxA D96A CVWRQHSRELRLESIKSQILSKLRLKEAPNISREVVKQLL
PKAPPLQQILDLHDFQGAALQPEDFLEEDEYHATTETVIS
MAQETDPAVQTDGSPLCCHFHFSPKVMFTKVLKAQLW
VYLRPVPRPATVYLQILRLKPLTGEGTAGGGGGGRRHIR
IRSLKIELHSRSGHWQSIDFKQVLHSWFRQPQSNWGIEIN
AFDPSGTDLAVTSLGPGAEGLHPFMELRVLENTKASRA
NLGLDCDEHSSESRCCRYPLTVDFEAFGWDWIIAPKRYK
ANYCSGQCEYMFMQKYPHTHLVQQANPRGSAGPCCTP
TKMSPINMLYFNDKQQIIYGKIPGMVVDRCGCS
proGDF-11 Mouse AEGPAAAAAAAAAAAGVGGERSSRPAPSAPPEPDGCPV 103 D96A CVWRQHSRELRLESIKSQILSKLRLKEAPNISREVVKQLL
PKAPPLQQILDLHDFQGDALQPEDFLEEDEYHATTETVIS
MAQETDPAVQTDGSPLCCHFHFSPKVMFTKVLKAQLW
VYLRPVPRPATVYLQILRLKPLTGEGTAGGGGGGRRHIR
IRSLKIELHSRSGHWQSIDFKQVLHSWFRQPQSNWGIEIN
AFDPSGTDLAVTSLGPGAEGLHPFMELRVLENTKRSRRN
LGLDCDEHSSESRCCRYPLTVDFEAFGWDWIIAPKRYKA
NYCSGQCEYMFMQKYPHTHLVQQANPRGSAGPCCTPT
KMSPINMLYFNDKQQIIYGKIPGMVVDRCGCS
GDF-11 Mouse AEGPAAAAAAAAAAAGVGGERSSRPAPSAPPEPDGCPV 104 prodomain CVWRQHSRELRLESIKSQILSKLRLKEAPNISREVVKQLL
PKAPPLQQILDLHDFQGDALQPEDFLEEDEYHATTETVIS
MAQETDPAVQTDGSPLCCHFHFSPKVMFTKVLKAQLW
VYLRPVPRPATVYLQILRLKPLTGEGTAGGGGGGRRHIR
IRSLKIELHSRSGHWQSIDFKQVLHSWFRQPQSNWGIEIN
AFDPSGTDLAVTSLGPGAEGLHPFMELRVLENTKRSRR
GDF-11 Mouse AEGPAAAAAAAAAAAGVGGERSSRPAPSAPPEPDGCPV 105 prodomain CVWRQHSRELRLESIKSQILSKLRLKEAPNISREVVKQLL
D96A PKAPPLQQILDLHDFQGAALQPEDFLEEDEYHATTETVIS
MAQETDPAVQTDGSPLCCHFHFSPKVMFTKVLKAQLW
VYLRPVPRPATVYLQILRLKPLTGEGTAGGGGGGRRHIR
IRSLKIELHSRSGHWQSIDFKQVLHSWFRQPQSNWGIEIN
AFDPSGTDLAVTSLGPGAEGLHPFMELRVLENTKRSRR
LTBP3 CYNO GPAGERGAGGGGALARERFKVVFAPVICKRTCLKGQCR 106
DSCQQGSNMTLIGENGHSTDTLTGSGFRVVVCPLPCMN
GGQCSSRNQCLCPPDFTGRFCQVPAGGAGGGTGGSGPG
LSRAGALSTGALPPLAPEGDSVASKHAIYAVQVIADPPG
PGEGPPAQHAAFLVPLGPGQISAEVQAPPPVVNVRVHHP
PEASVQVHRIESSNAEGAAPSQHLLPHPKPSHPRPPTQKP
LGRCFQDTLPKQPCGSNPLPGLTKQEDCCGSIGTAWGQS
KCHKCPQLQYTGVQKPGPVRGEVGADCPQGYKRLNST
HCQDINECAMPGVCRHGDCLNNPGSYRCVCPPGHSLGP
SRTQCIADKPEEKSLCFRLVSPEHQCQHPLTTRLTRQLCC
CSVGKAWGARCQRCPADGTAAFKEICPAGKGYHILTSH
QTLTIQGESDFSLFLHPDGPPKPQQLPESPSQAPPPEDTEE
ERGVTTDSPVSEERSVQQSHPTATTSPARPYPELISRPSPP
TMRWFLPDLPPSRSAVEIAPTQVTETDECRLNQNICGHG
ECVPGPPDYSCHCNPGYRSHPQHRYCVDVNECEAEPCG
PGRGICMNTGGSYNCHCNRGYRLHVGAGGRSCVDLNE
CAKPHLCGDGGFCINFPGHYKCNCYPGYRLKASRPPVC
EDIDECRDPSSCPDGKCENKPGSFKCIACQPGYRSQGGG
ACRDVNECAEGSPCSPGWCENLPGSFRCTCAQGYAPAP DGRSCVDVDECEAGDVCDNGICTNTPGSFQCQCLSGYH
LSRDRSHCEDIDECDFPAACIGGDCINTNGSYRCLCPQG
HRLVGGRKCQDIDECTQDPGLCLPHGACKNLQGSYVCV
CDEGFTPTQDQHGCEEVEQPHHKKECYLNFDDTVFCDS
VLATNVTQQECCCSLGAGWGDHCEIYPCPVYSSAEFHS
LCPDGKGYTQDNNIVNYGIPAHRDIDECMLFGAEICKEG
KCVNTQPGYECYCKQGFYYDGNLLECVDVDECLDESN
CRNGVCENTRGGYRCACTPPAEYSPAQRQCLSPEEMDV
DECQDPAACRPGRCVNLPGSYRCECRPPWVPGPSGRDC
QLPESPAERAPERRDVCWSQRGEDGMCAGPQAGPALTF
DDCCCRQGRGWGAQCRPCPPRGAGSQCPTSQSESNSFW
DTSPLLLGKPRRDEDSSEEDSDECRCVSGRCVPRPGGAV
CECPGGFQLDASRARCVDIDECRELNQRGLLCKSERCV
NTSGSFRCVCKAGFARSRPHGACVPQRRR
LTBP3 Mouse GPAGERGTGGGGALARERFKVVFAPVICKRTCLKGQCR 107
DSCQQGSNMTLIGENGHSTDTLTGSAFRVVVCPLPCMN
GGQCSSRNQCLCPPDFTGRFCQVPAAGTGAGTGSSGPG
LARTGAMSTGPLPPLAPEGESVASKHAIYAVQVIADPPG
PGEGPPAQHAAFLVPLGPGQISAEVQAPPPVVNVRVHHP
PEASVQVHRIEGPNAEGPASSQHLLPHPKPPHPRPPTQKP
LGRCFQDTLPKQPCGSNPLPGLTKQEDCCGSIGTAWGQS
KCHKCPQLQYTGVQKPVPVRGEVGADCPQGYKRLNST
HCQDINECAMPGNVCHGDCLNNPGSYRCVCPPGHSLGP
LAAQCIADKPEEKSLCFRLVSTEHQCQHPLTTRLTRQLC
CCSVGKAWGARCQRCPADGTAAFKEICPGKGYHILTSH
QTLTIQGESDFSLFLHPDGPPKPQQLPESPSRAPPLEDTEE
ERGVTMDPPVSEERSVQQSHPTTTTSPPRPYPELISRPSPP
TFHRFLPDLPPSRSAVEIAPTQVTETDECRLNQNICGHGQ
CVPGPSDYSCHCNAGYRSHPQHRYCVDVNECEAEPCGP
GKGICMNTGGSYNCHCNRGYRLHVGAGGRSCVDLNEC
AKPHLCGDGGFCINFPGHYKCNCYPGYRLKASRPPICED
IDECRDPSTCPDGKCENKPGSFKCIACQPGYRSQGGGAC
RDVNECSEGTPCSPGWCENLPGSYRCTCAQYEPAQDGL
SCIDVDECEAGKVCQDGICTNTPGSFQCQCLSGYHLSRD
RSRCEDIDECDFPAACIGGDCINTNGSYRCLCPLGHRLV
GGRKCKKDIDECSQDPGLCLPHACENLQGSYVCVCDEG
FTLTQDQHGCEEVEQPHHKKECYLNFDDTVFCDSVLAT
NVTQQECCCSLGAGWGDHCEIYPCPVYSSAEFHSLVPD
GKRLHSGQQHCELCIPAHRDIDECILFGAEICKEGKCVNT
QPGYECYCKQGFYYDGNLLECVDVDECLDESNCRNGV
CENTRGGYRCACTPPAEYSPAQAQCLIPERWSTPQRDV
KCAGASEERTACVWGPWAGPALTFDDCCCRQPRLGTQ
CRPCPPRGTGSQCPTSQSESNSFWDTSPLLLGKSPRDEDS
SEEDSDECRCVSGRCVPRPGGAVCECPGGFQLDASRAR
CVDIDECRELNQRGLLCKSERCVNTSGSFRCVCKAGFTR
SRPHGPACLSAAADDAAIAHTSVIDHRGYFH
LTBP1S Cyno NHTGRIKVVFTPSICKVTCTKGSCQNSCEKGNTTTLISEN 108
GHAADTLTATNFRVVLCHLPCMNGGQCSSRDKCQCPPN
FTGKLCQIPVHGASVPKLYQHSQQPGKALGTHVIHSTHT
LPLTVTSQQGVKVKFPPNIVNIHVKHPPEASVQIHQVSRI
DGPTGQKTKEAQPGQSQVSYQGLPVQKTQTIHSTYSHQ
QVIPHVYPVAAKTQLGRCFQETIGSQCGKALPGLSKQED
CCGTVGTSWGFNKCQKCPKKPSYHGYNQMMECLPGYK
RVNNTFCQDINECQLQGVCPNGECLNTMGSYRCTCKIG
FGPDPTFSSCVPDPPVISEEKGPCYRLVSSGRQCMHPLSV HLTKQLCCCSVGKAWGPHCEKCPLPGTAAFKEICPGGM
GYTVSGVHRRRPIHHHVGKGPVFVKPKNTQPVAKSTHP
PPLPAKEEPVEALTFSREHGPGVAEPEVATAPPEKEIPSL
DQEKTKLEPGQPQLSPGISTIHLHPQFPVVIEKTSPPVPVE
VAPEASTSSASQVIAPTQVTEINECTVNPDICGAGHCINL
PVRYTCICYEGYKFSEQQRKCVDIDECTQVQHLCSQGRC
ENTEGSFLCICPAGFMASEEGTNCIDVDECLRPDVCGEG
HCVNTVGAFRCEYCDSGYRMTQRGRCEDIDECLNPSTC
PDEQCVNSPGSYQCVPCTEGFRGWNGQCLDVDECLEPN
VCTNGDCSNLEGSYMCSCHKGYTRTPDHKHCKDIDECQ
QGNLCVNGQCKNTEGSFRCTCGQGYQLSAAKDQCEDID
ECQHHHLCAHGQCRNTEGSFQCVCDQGYRASGLGDHC
EDINECLEDKSVCQRGDCINTAGSYDCTCPDGFQLDDN
KTCQDINECEHPGLCGPQGECLNTEGSFHCVCQQGFSIS
ADGRTCEDIDECVNNTVCDSHGFCDNTAGSFRCLCYQG
FQAPQDGQGCVDVNECELLSGVCGEAFCENVEGSFLCV
CADENQEYSPMTGQCRSRTSTDLDVEQPKEEKKECYYN
LNDASLCDNVLAPNVTKQECCCTSGAGWGDNCEIFPCP
VLGTAEFTEMCPKGKGFVPAGESSSEAGGENYKDADEC
LLFGQEICKNGFCLNTRPGYECYCKQGTYYDPVKLQCF
DMDECQDPSSCIDGQCVNTEGSYNCFCTHPMVLDASEK
RCIRPAESNEQIEETDVYQDLCWEHLSDEYVCSRPLVGK
QTTYTECCCLYGEAWGMQCALCPMKDSDDYAQLCNIP
VTGRRQPYGRDALVDFSEQYAPEADPYFIQDRFLNSFEE
LQAEECGILNGCENGRCVRVQEGYTCDCFDGYHLDTAK
MTCVDVNECDELNNRMSLCKNAKCINTEGSYKCLCLPG
YVPSDKPNYCTPLNTALNLEKDSDLE
LTBP1S mouse NHTGRIKVVFTPSICKVTCTKGNCQNSCQKGNTTTLISE 109
NGHAADTLTATNFRVVICHLPCMNGGQCSSRDKCQCPP
NFTGKLCQIPVLGASMPKLYQHAQQQGKALGSHVIHST
HTLPLTMTSQQGVKVKFPPNIVNIHVKHPPEASVQIHQV
SRIDSPGGQKVKEAQPGQSQVSYQGLPVQKTQTVHSTY
SHQQLIPHVYPVAAKTQLGRCFQETIGSQCGKALPGLSK
QEDCCGTVGTSWGFNKCQKCPKKQSYHGYTQMMECL
QGYKRVNNTFCQDINECQLQGVCPNGECLNTMGSYRCS
CKMGFGPDPTFSSCVPDPPVISEEKGPCYRLVSPGRHCM
HPLSVHLTKQICCCSVGKAWGPHCEKCPLPGTAAFKEIC
PGGMGYTVSGVHRRRPIHQHIGKEAVYVKPKNTQPVAK
STHPPPLPAKEEPVEALTSSWEHGPRGAEPEVVTAPPEK
EIPSLDQEKTRLEPGQPQLSPGVSTIHLHPQFPVVVEKTSP
PVPVEVAPEASTSSASQVIAPTQVTEINECTVNPDICGAG
HCINLPVRYTCICYEGYKFSEQLRKCVDIDECAQVRHLC
SQGRCENTEGSFLCVCPAGFMASEEGTNCIDVDECLRPD
MCRDGRCINTAGAFRCEYCDSGYRMSRRGYCEDIDECL
KPSTCPEEQCVNTPGSYQCVPCTEGFRGWNGQCLDVDE
CLQPKVCTNGSCTNLEGSYMCSCHRGYSPTPDHRHCQD
IDECQQGNLCMNGQCRNTDGSFRCTCGQGYQLSAAKD
QCEDIDECEHHHLCSHGQCRNTEGSFQCVCNQGYRASV
LGDHCEDINECLEDSSVCQGGDCINTAGSYDCTCPDGFQ
LNDNKGCQDINECAQPGLCGSHGECLNTQGSFHCVCEQ
GFSISADGRTCEDIDECVNNTVCDSHGFCDNTAGSFRCL
CYQGFQAPQDGQGCVDVNECELLSGVCGEAFCENVEGS
FLCVCADENQEYSPMTGQCRSRVTEDSGVDRQPREEKK
ECYYNLNDASLCDNVLAPNVTKQECCCTSGAGWGDNC
EIFPCPVQGTAEFTEMCPRGKGLVPAGESSYDTGGENYK DADECLLFGEEICKNGYCLNTQPGYECYCKQGTYYDPV
KLQCFDMDECQDPNSCIDGQCVNTEGSYNCFCTHPMVL
DASEKRCVQPTESNEQIEETDVYQDLCWEHLSEEYVCSR
PLVGKQTTYTECCCLYGEAWGMQCALCPMKDSDDYA
QLCNIPVTGRRRPYGRDALVDFSEQYGPETDPYFIQDRF
LNSFEELQAEECGILNGCENGRCVRVQEGYTCDCFDGY
HLDMAKMTCVDVNECSELNNRMSLCKNAKCINTEGSY
KCLCLPGYIPSDKPNYCTPLNSALNLDKESDLE
GARP mouse ISQRREQVPCRTVNKEALCHGLGLLQVPSVLSLDIQALY 110
LSGNQLQSILVSPLGFYTALRHLDLSDNQISFLQAGVFQA
LPYLEHLNLAHNRLATGMALNSGGLGRLPLLVSLDLSG
NSLHGNLVERLLGETPRLRTLSLAENSLTRLARHTFWG
MPAVEQLDLHSNVLMDIEDGAFEALPHLTHLNLSRNSL
TCISDFSLQQLQVLDLSCNSIEAFQTAPEPQAQFQLAWL
DLRENKLLHFPDLAVFPRLIYLNVSNNLIQLPAGLPRGSE
DLHAPSEGWSASPLSNPSRNASTHPLSQLLNLDLSYNEIE
LVPASFLEHLTSLRFLNLSRNCLRSFEARQVDSLPCLVLL
DLSHNVLEALELGTKVLGSLQTLLLQDNALQELPPYTFA
SLASLQRLNLQGNQVSPCGGPAEPGPPGCVDFSGIPTLH
VLNMAGNSMGMLRAGSFLHTPLTELDLSTNPGLDVATG
ALVGLEASLEVLELQGNGLTVLRVDLPCFLRLKRLNLAE
NQLSHLPAWTRAVSLEVLDLRNNSFSLLPGNAMGGLET
SLRRLYLQGNPLSCCGNGWLAAQLHQGRVDVDATQDL
ICRFGSQEELSLSLVRPEDCEKGGLKNVNLILLLSFTLVS
AIVLTTL ATICFLRRQKLS QQ YKA
sGARP mouse ISQRREQVPCRTVNKEALCHGLGLLQVPSVLSLDIQALY 111
LSGNQLQSILVSPLGFYTALRHLDLSDNQISFLQAGVFQA
LPYLEHLNLAHNRLATGMALNSGGLGRLPLLVSLDLSG
NSLHGNLVERLLGETPRLRTLSLAENSLTRLARHTFWG
MPAVEQLDLHSNVLMDIEDGAFEALPHLTHLNLSRNSL
TCISDFSLQQLQVLDLSCNSIEAFQTAPEPQAQFQLAWL
DLRENKLLHFPDLAVFPRLIYLNVSNNLIQLPAGLPRGSE
DLHAPSEGWSASPLSNPSRNASTHPLSQLLNLDLSYNEIE
LVPASFLEHLTSLRFLNLSRNCLRSFEARQVDSLPCLVLL
DLSHNVLEALELGTKVLGSLQTLLLQDNALQELPPYTFA
SLASLQRLNLQGNQVSPCGGPAEPGPPGCVDFSGIPTLH
VLNMAGNSMGMLRAGSFLHTPLTELDLSTNPGLDVATG
ALVGLEASLEVLELQGNGLTVLRVDLPCFLRLKRLNLAE
NQLSHLPAWTRAVSLEVLDLRNNSFSLLPGNAMGGLET
SLRRLYLQGNPLSCCGNGWLAAQLHQGRVDVDATQDL
ICRFGSQEELSLSLVRPEDCEKGGLKNVN
LRRC33 mouse WRSGPGTATAASQGGCKVVDGVADCRGLNLASVPSSLP 112
PHSRMLILDANPLKDLWNHSLQAYPRLENLSLHSCHLD
RISHYAFREQGHLRNLVLADNRLSENYKESAAALHTLL
GLRRLDLSGNSLTEDMAALMLQNLSSLEVVSLARNTLM
RLDDSIFEGLEHLVELDLQRNYIFEIEGGAFDGLTELRRL
NLAYNNLPCIVDFSLTQLRFLNVSYNILEWFLAAREEVA
FELEILDLSHNQLLFFPLLPQCGKLHTLLLQDNNMGFYR
ELYNTSSPQEMVAQFLLVDGNVTNITTVNLWEEFSSSDL
SALRFLDMSQNQFRHLPDGFLKKTPSLSHLNLNQNCLK
MLHIREHEPPGALTELDLSHNQLAELHLAPGLTGSLRNL
RVFNLSSNQLLGVPTGLFDNASSITTIDMSHNQISLCPQM
VPVDWEGPPSCVDFRNMGSLRSLSLDGCGLKALQDCPF
QGTSLTHLDLSSNWGVLNGSISPLWAVAPTLQVLSLRD VGLGSGAAEMDFSAFGNLRALDLSGNSLTSFPKFKGSLA
LRTLDLRRNSLTALPQRVVSEQPLRGLQTIYLSQNPYDC
CGVEGWGALQQHFKTVADLSMVTCNLSSKIVRVVELPE
GLPQGCKWEQVDTGLFYLVLILPSCLTLLVACTVVFLTF
KKPLLQVIKSRCHWSSIY
SLRRC33 mouse WRSGPGTATAASQGGCKVVDGVADCRGLNLASVPSSLP 113
PHSRMLILDANPLKDLWNHSLQAYPRLENLSLHSCHLD
RISHYAFREQGHLRNLVLADNRLSENYKESAAALHTLL
GLRRLDLSGNSLTEDMAALMLQNLSSLEVVSLARNTLM
RLDDSIFEGLEHLVELDLQRNYIFEIEGGAFDGLTELRRL
NLAYNNLPCIVDFSLTQLRFLNVSYNILEWFLAAREEVA
FELEILDLSHNQLLFFPLLPQCGKLHTLLLQDNNMGFYR
ELYNTSSPQEMVAQFLLVDGNVTNITTVNLWEEFSSSDL
SALRFLDMSQNQFRHLPDGFLKKTPSLSHLNLNQNCLK
MLHIREHEPPGALTELDLSHNQLAELHLAPGLTGSLRNL
RVFNLSSNQLLGVPTGLFDNASSITTIDMSHNQISLCPQM
VPVDWEGPPSCVDFRNMGSLRSLSLDGCGLKALQDCPF
QGTSLTHLDLSSNWGVLNGSISPLWAVAPTLQVLSLRD
VGLGSGAAEMDFSAFGNLRALDLSGNSLTSFPKFKGSLA
LRTLDLRRNSLTALPQRVVSEQPLRGLQTIYLSQNPYDC
CGVEGWGALQQHFKTVADLSMVTCNLSSKIVRVVELPE
GLPQGCKWEQVDTGL
LRRC33 Cyno WRDRSVTATAASQRGCKLVGGDTDCRGQSLASVPSSLP 114
PHARTLILDANPLKALWNHSLQPYPLLESLSLHSCHLERI
GRGAFQEQGHLRSLVLGDNCLSENYKETAAALHTLPGL
QTLDLSGNSLTEDMAALMLQNLSSLQSVSLARNTIMRL
DDSVFEGLERLRELDLQRNYIFEIEGGAFDGLTELRHLNL
AYNNLPCIVDFGLTQLRSLNVSYNVLEWFLAAGGEAAF
ELETLDLSHNQLLFFPLLPQYSKLHTLLLRDNNMGFYRD
LYNTSSPREMVAQFLLVDGNVTNITTVNLWEEFSSSDLA
DLRFLDMSQNQFQYLPDGFLRKMPSLSHLNLNQNCLMT
LHIREHEPPGALTELDLSHNQLSELHLTPGLASCLGSLRL
FNLSSNQLLGVPPGLFANARNITTLDMSHNQISLCPLPAA
SDRVGPPSCVDFRNMASLRSLSLEGCGLGALPDCPFQGT
SLTSLDLSSNWGVLNGSLAPLRDVAPMLQVLSLRNMGL
HSNFMALDFSGFGNLRDLDLSGNCLTTFPRFGGSLALET
LDLRRNSLTALPQKAVSEQLSRGLRTIYLSQNPYDCCGV
DGWGALQQGQTVADWATVTCNLSSKIIRLAELPGGVPR
DCKWERLDLGLLYLVLILPSCLTLLVACTLIVLTFKKPLL
QVIKSRCHWSSVY
SLRRC33 Cyno WRDRSVTATAASQRGCKLVGGDTDCRGQSLASVPSSLP 115
PHARTLILDANPLKALWNHSLQPYPLLESLSLHSCHLERI
GRGAFQEQGHLRSLVLGDNCLSENYKETAAALHTLPGL
QTLDLSGNSLTEDMAALMLQNLSSLQSVSLARNTIMRL
DDSVFEGLERLRELDLQRNYIFEIEGGAFDGLTELRHLNL
AYNNLPCIVDFGLTQLRSLNVSYNVLEWFLAAGGEAAF
ELETLDLSHNQLLFFPLLPQYSKLHTLLLRDNNMGFYRD
LYNTSSPREMVAQFLLVDGNVTNITTVNLWEEFSSSDLA
DLRFLDMSQNQFQYLPDGFLRKMPSLSHLNLNQNCLMT
LHIREHEPPGALTELDLSHNQLSELHLTPGLASCLGSLRL
FNLSSNQLLGVPPGLFANARNITTLDMSHNQISLCPLPAA
SDRVGPPSCVDFRNMASLRSLSLEGCGLGALPDCPFQGT
SLTSLDLSSNWGVLNGSLAPLRDVAPMLQVLSLRNMGL
HSNFMALDFSGFGNLRDLDLSGNCLTTFPRFGGSLALET LDLRRNSLTALPQKAVSEQLSRGLRTIYLSQNPYDCCGV
DGWGALQQGQTVADWATVTCNLSSKIIRLAELPGGVPR DCKWERLDLGL
[00224] In some embodiments, recombinant proteins may be combined and/or complexed with one or more additional recombinant components. Such components may include extracellular proteins known to associate with GPCs including, but not limited to LTBPs, fibrillins, perlecan, GASP 1/2 proteins, follistatin, follistatin-related gene (FLRG), decorin and/or GARP (including, but not limited to recombinant forms of such proteins). Some recombinant GPCs of the present disclosure must be co-expressed with one or more of such extracellular proteins for proper expression and/or folding.
[00225] In some embodiments, complexed LTBPs may include, but are not limited to LTBPl, LTBP2, LTBP3 and/or LTBP4 with or without detectable labels. Complexed LTBPs may comprise LTBP fragments and/or mutations. Some recombinant forms of LTBPs complexed with recombinant GPCs may comprise alternatively spliced variants of LTBPs. Some such variants of LTBPl are shortened at the N-terminus, referred to herein as LTBPIS. Some recombinant proteins of the present disclosure may comprise LTBPs, fragments or mutants thereof comprising the amino acid sequences listed in the Table below. In some cases, these sequences are expressed in association with N- and/or C-terminal secretion signal sequences [e.g. human Ig kappa chains with amino acid sequence
MDMRVPAQLLGLLLLWFSGVLG (SEQ ID NO: 66)], flag tag sequences [e.g.
DYKDDDDK (SEQ ID NO: 67)], one or more 3C protease cleavage site [e.g. LEVLFQGP (SEQ ID NO: 68)], one or more biotinylation site and/or His-tag sequences [e.g. HHHHHH (SEQ ID NO: 69)].
Table 6. LTBP sequences
Protein Sequence SEQ
ID NO
LTBPl 1265-1443 NECELLSGVCGEAFCENVEGSFLCVCADENQEYSPMTGQC 116
RSRTSTDLDVDVDQPKEEKKECYYNLNDASLCDNVLAPNV
TKQECCCTSGVGWGDNCEIFPCPVLGTAEFTEMCPKGKGF
VPAGESSSEAGGENYKDADECLLFGQEICKNGFCLNTRPGY
ECYCKQGTYYDPVKLQCF
LTBPl 1265-1698 NECELLSGVCGEAFCENVEGSFLCVCADENQEYSPMTGQC 117
RSRTSTDLDVDVDQPKEEKKECYYNLNDASLCDNVLAPNV
TKQECCCTSGVGWGDNCEIFPCPVLGTAEFTEMCPKGKGF
VPAGESSSEAGGENYKDADECLLFGQEICKNGFCLNTRPGY
ECYCKQGTYYDPVKLQCFDMDECQDPSSCIDGQCVNTEGS
YNCFCTHPMVLDASEKRCIRPAESNEQIEETDVYQDLCWE
HLSDEYVCSRPLVGKQTTYTECCCLYGEAWGMQCALCPL KDSDDYAQLCNIPVTGRRQPYGRDALVDFSEQYTPEADPY
FIQDRFLNSFEELQAEECGILNGCENGRCVRVQEGYTCDCF DGYHLDTAKMTCVDVNECDELNNRMSLCKNAKCINTDGS YKCLCLPGYVPSDKPNYCTPLNTALNLEKDSDLE
LTBP1 809-1698 PSLDQEKTKLEPGQPQLSPGISTIHLHPQFPVVIEKTSPPVPV 118
EVAPEASTSSASQVIAPTQVTEINECTVNPDICGAGHCINLP
VRYTCICYEGYRFSEQQRKCVDIDECTQVQHLCSQGRCEN
TEGSFLCICPAGFMASEEGTNCIDVDECLRPDVCGEGHCVN
TVGAFRCEYCDSGYRMTQRGRCEDIDECLNPSTCPDEQCV
NSPGSYQCVPCTEGFRGWNGQCLDVDECLEPNVCANGDC
SNLEGSYMCSCHKGYTRTPDHKHCRDIDECQQGNLCVNG
QCKNTEGSFRCTCGQGYQLSAAKDQCEDIDECQHRHLCAH
GQCRNTEGSFQCVCDQGYRASGLGDHCEDINECLEDKSVC
QRGDCINTAGSYDCTCPDGFQLDDNKTCQDINECEHPGLC
GPQGECLNTEGSFHCVCQQGFSISADGRTCEDIDECVNNTV
CDSHGFCDNTAGSFRCLCYQGFQAPQDGQGCVDVNECEL
LSGVCGEAFCENVEGSFLCVCADENQEYSPMTGQCRSRTS
TDLDVDVDQPKEEKKECYYNLNDASLCDNVLAPNVTKQE
CCCTSGVGWGDNCEIFPCPVLGTAEFTEMCPKGKGFVPAG
ESSSEAGGENYKDADECLLFGQEICKNGFCLNTRPGYECYC
KQGTYYDPVKLQCFDMDECQDPSSCIDGQCVNTEGSYNCF
CTHPMVLDASEKRCIRPAESNEQIEETDVYQDLCWEHLSDE
YVCSRPLVGKQTTYTECCCLYGEAWGMQCALCPLKDSDD
YAQLCNIPVTGRRQPYGRDALVDFSEQYTPEADPYFIQDRF
LNSFEELQAEECGILNGCENGRCVRVQEGYTCDCFDGYHL
DTAKMTCVDVNECDELNNRMSLCKNAKCINTDGSYKCLC
LPGYVPSDKPNYCTPLNTALNLEKDSDLE
LTBP1S NHTGRIKVVFTPSICKVTCTKGSCQNSCEKGNTTTLISENGH 119
AADTLTATNFRVVICHLPCMNGGQCSSRDKCQCPPNFTGK
LCQIPVHGASVPKLYQHSQQPGKALGTHVIHSTHTLPLTVT
SQQGVKVKFPPNIVNIHVKHPPEASVQIHQVSRIDGPTGQK
TKEAQPGQSQVSYQGLPVQKTQTIHSTYSHQQVIPHVYPVA
AKTQLGRCFQETIGSQCGKALPGLSKQEDCCGTVGTSWGF
NKCQKCPKKPSYHGYNQMMECLPGYKRVNNTFCQDINEC
QLQGVCPNGECLNTMGSYRCTCKIGFGPDPTFSSCVPDPPV
ISEEKGPCYRLVSSGRQCMHPLSVHLTKQLCCCSVGKAWG
PHCEKCPLPGTAAFKEICPGGMGYTVSGVHRRRPIHHHVG
KGPVFVKPKNTQPVAKSTHPPPLPAKEEPVEALTFSREHGP
GVAEPEVATAPPEKEIPSLDQEKTKLEPGQPQLSPGISTIHLH
PQFPVVIEKTSPPVPVEVAPEASTSSASQVIAPTQVTEINECT
VNPDICGAGHCINLPVRYTCICYEGYRFSEQQRKCVDIDEC
TQVQHLCSQGRCENTEGSFLCICPAGFMASEEGTNCIDVDE
CLRPDVCGEGHCVNTVGAFRCEYCDSGYRMTQRGRCEDI
DECLNPSTCPDEQCVNSPGSYQCVPCTEGFRGWNGQCLDV
DECLEPNVCANGDCSNLEGSYMCSCHKGYTRTPDHKHCR
DIDECQQGNLCVNGQCKNTEGSFRCTCGQGYQLSAAKDQ
CEDIDECQHRHLCAHGQCRNTEGSFQCVCDQGYRASGLGD
HCEDINECLEDKSVCQRGDCINTAGSYDCTCPDGFQLDDN
KTCQDINECEHPGLCGPQGECLNTEGSFHCVCQQGFSISAD
GRTCEDIDECVNNTVCDSHGFCDNTAGSFRCLCYQGFQAP
QDGQGCVDVNECELLSGVCGEAFCENVEGSFLCVCADEN
QEYSPMTGQCRSRTSTDLDVDVDQPKEEKKECYYNLNDAS
LCDNVLAPNVTKQECCCTSGVGWGDNCEIFPCPVLGTAEF
TEMCPKGKGFVPAGESSSEAGGENYKDADECLLFGQEICK
NGFCLNTRPGYECYCKQGTYYDPVKLQCFDMDECQDPSS CIDGQCVNTEGSYNCFCTHPMVLDASEKRCIRPAESNEQIE
ETDVYQDLCWEHLSDEYVCSRPLVGKQTTYTECCCLYGEA
WGMQCALCPLKDSDDYAQLCNIPVTGRRQPYGRDALVDF
SEQYTPEADPYFIQDRFLNSFEELQAEECGILNGCENGRCVR
VQEGYTCDCFDGYHLDTAKMTCVDVNECDELNNRMSLCK
NAKCINTDGSYKCLCLPGYVPSDKPNYCTPLNTALNLEKDS
DLE
LTBP3 GPAGERGAGGGGALARERFKVVFAPVICKRTCLKGQCRDS 120
CQQGSNMTLIGENGHSTDTLTGSGFRVVVCPLPCMNGGQC
SSRNQCLCPPDFTGRFCQVPAGGAGGGTGGSGPGLSRTGA
LSTGALPPLAPEGDSVASKHAIYAVQVIADPPGPGEGPPAQ
HAAFLVPLGPGQISAEVQAPPPVVNVRVHHPPEASVQVHRI
ESSNAESAAPSQHLLPHPKPSHPRPPTQKPLGRCFQDTLPKQ
PCGSNPLPGLTKQEDCCGSIGTAWGQSKCHKCPQLQYTGV
QKPGPVRGEVGADCPQGYKRLNSTHCQDINECAMPGVCR
HGDCLNNPGSYRCVCPPGHSLGPSRTQCIADKPEEKSLCFR
LVSPEHQCQHPLTTRLTRQLCCCSVGKAWGARCQRCPTDG
TAAFKEICPAGKGYHILTSHQTLTIQGESDFSLFLHPDGPPK
PQQLPESPSQAPPPEDTEEERGVTTDSPVSEERSVQQSHPTA
TTTPARPYPELISRPSPPTMRWFLPDLPPSRSAVEIAPTQVTE
TDECRLNQNICGHGECVPGPPDYSCHCNPGYRSHPQHRYC
VDVNECEAEPCGPGRGICMNTGGSYNCHCNRGYRLHVGA
GGRSCVDLNECAKPHLCGDGGFCINFPGHYKCNCYPGYRL
KASRPPVCEDIDECRDPSSCPDGKCENKPGSFKCIACQPGY
RSQGGGACRDVNECAEGSPCSPGWCENLPGSFRCTCAQGY
APAPDGRSCLDVDECEAGDVCDNGICSNTPGSFQCQCLSG
YHLSRDRSHCEDIDECDFPAACIGGDCINTNGSYRCLCPQG
HRLVGGRKCQDIDECSQDPSLCLPHGACKNLQGSYVCVCD
EGFTPTQDQHGCEEVEQPHHKKECYLNFDDTVFCDSVLAT
NVTQQECCCSLGAGWGDHCEIYPCPVYSSAEFHSLCPDGK
GYTQDNNIVNYGIPAHRDIDECMLFGSEICKEGKCVNTQPG
YECYCKQGFYYDGNLLECVDVDECLDESNCRNGVCENTR
GGYRCACTPPAEYSPAQRQCLSPEEMDVDECQDPAACRPG
RCVNLPGSYRCECRPPWVPGPSGRDCQLPESPAERAPERRD
VCWSQRGEDGMCAGPLAGPALTFDDCCCRQGRGWGAQC
RPCPPRGAGSHCPTSQSESNSFWDTSPLLLGKPPRDEDSSEE
DSDECRCVSGRCVPRPGGAVCECPGGFQLDASRARCVDID
ECRELNQRGLLCKSERCVNTSGSFRCVCKAGFARSRPHGA
CVPQRRR
LTBP3 EGF-like DIDECMLFGSEICKEGKCVNTQPGYECYCKQGFYYDGNLL 121 domain, module 1 ECVDVDECLDESNCRNGVCENTRGGYRCACTPPAEYSPAQ
RQCLSP
LTBP3 EGF-like DVDECQDPAACRPGRCVNLPGSYRCECRPPWVPGPSGRDC 122 domain, module 2 QLP
LTBP3 EGF-like DIDECSQDPSLCLPHGACKNLQGSYVCVCDEGFTPTQDQH 123 domain, module 3 GCE
LTBP3 EGF-like DIDECMLFGSEICKEGKCVNTQPGYECYCKQGFYYDGNLL 124 domain, module 4 ECV
TB domain, module 1 KKECYLNFDDTVFCDSVLATNVTQQECCCSLGAGWGDHC 125
EIYPCPVYSSAEFHSLCP
TB domain, module 2 DVCWSQRGEDGMCAGPLAGPALTFDDCCCRQGRGWGAQ 126
CRPCPPRGAGSHCP
L3-TB3TB4 isoform 1 KKECYLNFDDTVFCDSVLATNVTQQECCCSLGAGWGDHC 127
EIYPCPVYSSAEFHSLCPDGKGYTQDNNIVNYGIPAHRDIDE CMLFGSEICKEGKCVNTQPGYECYCKQGFYYDGNLLECVD
VDECLDESNCRNGVCENTRGGYRCACTPPAEYSPAQRQCL
SPEEMDVDECQDPAACRPGRCVNLPGSYRCECRPPWVPGP
SGRDCQLPESPAERAPERRDVCWSQRGEDGMCAGPLAGP
ALTFDDCCCRQGRGWGAQCRPCPPRGAGSHCPTSQSE
L3-TB3TB4 isoform 2 KKECYLNFDDTVFCDSVLATNVTQQECCCSLGAGWGDHC 128
EIYPCPVYSSAEFHSLCPDGKGYTQDNNIVNYGIPAHRDIDE
CMLFGSEICKEGKCVNTQPGYECYCKQGFYYDGNLLECVD
VDECLDESNCRNGVCENTRGGYRCACTPPAEYSPAQRQCL
SPEEMERAPERRDVCWSQRGEDGMCAGPLAGPALTFDDC
CCRQGRGWGAQCRPCPPRGAGSHCPTSQSE
L3-ETB3E, type 1 DIDECSQDPSLCLPHGACKNLQGSYVCVCDEGFTPTQDQH 129
GCEEVEQPHHKKECYLNFDDTVFCDSVLATNVTQQECCCS
LGAGWGDHCEIYPCPVYSSAEFHSLCPDGKGYTQDNNIVN
YGIPAHRDIDECMLFGSEICKEGKCVNTQPGYECYCKQGFY
YDGNLLECVDVDE
L3-ETB3E, type 2 QDIDECSQDPSLCLPHGACKNLQGSYVCVCDEGFTPTQDQ 130
HGCEEVEQPHHKKECYLNFDDTVFCDSVLATNVTQQECCC
SLGAGWGDHCEIYPCPVYSSAEFHSLCPDGKGYTQDNNIV
NYGIPAHRDIDECMLFGSEICKEGKCVNTQPGYECYCKQGF
YYDGNLLECVDVDE
L3-ETB3E, type 3 DIDECSQDPSLCLPHGACKNLQGSYVCVCDEGFTPTQDQH 131
GCEEVEQPHHKKECYLNFDDTVFCDSVLATNVTQQECCCS
LGAGWGDHCEIYPCPVYSSAEFHSLCPDGKGYTQDNNIVN
YGIPAHRDIDECMLFGSEICKEGKCVNTQPGYECYCKQGFY
YDGNLLECV
L3-ETB3E, type 4 QDIDECSQDPSLCLPHGACKNLQGSYVCVCDEGFTPTQDQ 132
HGCEEVEQPHHKKECYLNFDDTVFCDSVLATNVTQQECCC
SLGAGWGDHCEIYPCPVYSSAEFHSLCPDGKGYTQDNNIV
NYGIPAHRDIDECMLFGSEICKEGKCVNTQPGYECYCKQGF
YYDGNLLECV
L3-ETB3E, type IN SSGDIDECSQDPSLCLPHGACKNLQGSYVCVCDEGFTPTQD 133
QHGCEEVEQPHHKKECYLNFDDTVFCDSVLATNVTQQECC
CSLGAGWGDHCEIYPCPVYSSAEFHSLCPDGKGYTQDNNI
VNYGIPAHRDIDECMLFGSEICKEGKCVNTQPGYECYCKQ
GFYYDGNLLECVDVDE
L3-ETB3E, type 2N SSGQDIDECSQDPSLCLPHGACKNLQGSYVCVCDEGFTPTQ 134
DQHGCEEVEQPHHKKECYLNFDDTVFCDSVLATNVTQQE
CCCSLGAGWGDHCEIYPCPVYSSAEFHSLCPDGKGYTQDN
NIVNYGIPAHRDIDECMLFGSEICKEGKCVNTQPGYECYCK
QGFYYDGNLLECVDVDE
L3-ETB3E, type 3N SSGDIDECSQDPSLCLPHGACKNLQGSYVCVCDEGFTPTQD 135
QHGCEEVEQPHHKKECYLNFDDTVFCDSVLATNVTQQECC
CSLGAGWGDHCEIYPCPVYSSAEFHSLCPDGKGYTQDNNI
VNYGIPAHRDIDECMLFGSEICKEGKCVNTQPGYECYCKQ
GFYYDGNLLECV
L3-ETB3E, type 4N SSGQDIDECSQDPSLCLPHGACKNLQGSYVCVCDEGFTPTQ 136
DQHGCEEVEQPHHKKECYLNFDDTVFCDSVLATNVTQQE
CCCSLGAGWGDHCEIYPCPVYSSAEFHSLCPDGKGYTQDN
NIVNYGIPAHRDIDECMLFGSEICKEGKCVNTQPGYECYCK
QGFYYDGNLLECV
L1-ETB3E, type 1 GQGCVDVNECELLSGVCGEAFCENVEGSFLCVCADENQEY 137
SPMTGQCRSRTSTDLDVDVDQPKEEKKECYYNLNDASLCD NVLAPNVTKQECCCTSGVGWGDNCEIFPCPVLGTAEFTEM CPKGKGFVPAGESSSEAGGENYKDADECLLFGQEICKNGF
CLNTRPGYECYCKQGTYYDPVKLQCF
Ll-ETB3-C-term, type GQGCVDVNECELLSGVCGEAFCENVEGSFLCVCADENQEY 138 1 SPMTGQCRSRTSTDLDVDVDQPKEEKKECYYNLNDASLCD
NVLAPNVTKQECCCTSGVGWGDNCEIFPCPVLGTAEFTEM
CPKGKGFVPAGESSSEAGGENYKDADECLLFGQEICKNGF
CLNTRPGYECYCKQGTYYDPVKLQCFDMDECQDPSSCIDG
QCVNTEGSYNCFCTHPMVLDASEKRCIRPAESNEQIEETDV
YQDLCWEHLSDEYVCSRPLVGKQTTYTECCCLYGEAWGM
QCALCPLKDSDDYAQLCNIPVTGRRQPYGRDALVDFSEQY
TPEADPYFIQDRFLNSFEELQAEECGILNGCENGRCVRVQE
GYTCDCFDGYHLDTAKMTCVDVNECDELNNRMSLCKNA
KCINTDGSYKCLCLPGYVPSDKPNYCTPLNTALNLEKDSDL
E
L1-ETB3E, type 2 NECELLSGVCGEAFCENVEGSFLCVCADENQEYSPMTGQC 139
RSRTSTDLDVDVDQPKEEKKECYYNLNDASLCDNVLAPNV
TKQECCCTSGVGWGDNCEIFPCPVLGTAEFTEMCPKGKGF
VPAGESSSEAGGENYKDADECLLFGQEICKNGFCLNTRPGY
ECYCKQGTYYDPVKLQCF
Ll-ETB3-C-term, type NECELLSGVCGEAFCENVEGSFLCVCADENQEYSPMTGQC 140 2 RSRTSTDLDVDVDQPKEEKKECYYNLNDASLCDNVLAPNV
TKQECCCTSGVGWGDNCEIFPCPVLGTAEFTEMCPKGKGF
VPAGESSSEAGGENYKDADECLLFGQEICKNGFCLNTRPGY
ECYCKQGTYYDPVKLQCFDMDECQDPSSCIDGQCVNTEGS
YNCFCTHPMVLDASEKRCIRPAESNEQIEETDVYQDLCWE
HLSDEYVCSRPLVGKQTTYTECCCLYGEAWGMQCALCPL
KDSDDYAQLCNIPVTGRRQPYGRDALVDFSEQYTPEADPY
FIQDRFLNSFEELQAEECGILNGCENGRCVRVQEGYTCDCF
DGYHLDTAKMTCVDVNECDELNNRMSLCKNAKCINTDGS
YKCLCLPGYVPSDKPNYCTPLNTALNLEKDSDLE
L1-ETB3E, type 3 DVNECELLSGVCGEAFCENVEGSFLCVCADENQEYSPMTG 141
QCRSRTSTDLDVDVDQPKEEKKECYYNLNDASLCDNVLAP NVTKQECCCTSGVGWGDNCEIFPCPVLGTAEFTEMCPKGK GFVPAGESSSEAGGENYKDADECLLFGQEICKNGFCLNTRP GYECYCKQGTYYDPVKLQCF
L1-EETB3EE DIDECVNNTVCDSHGFCDNTAGSFRCLCYQGFQAPQDGQG 142
CVDVNECELLSGVCGEAFCENVEGSFLCVCADENQEYSPM
TGQCRSRTSTDLDVDVDQPKEEKKECYYNLNDASLCDNVL
APNVTKQECCCTSGVGWGDNCEIFPCPVLGTAEFTEMCPK
GKGFVPAGESSSEAGGENYKDADECLLFGQEICKNGFCLN
TRPGYECYCKQGTYYDPVKLQCFDMDECQDPSSCIDGQCV
NTEGSYNCFCTHPMVLDASEKRCI
L1-E11-TB3 EINECTVNPDICGAGHCINLPVRYTCICYEGYRFSEQQRKCV 143
DIDECTQVQHLCSQGRCENTEGSFLCICPAGFMASEEGTNCI
DVDECLRPDVCGEGHCVNTVGAFRCEYCDSGYRMTQRGR
CEDIDECLNPSTCPDEQCVNSPGSYQCVPCTEGFRGWNGQ
CLDVDECLEPNVCANGDCSNLEGSYMCSCHKGYTRTPDH
KHCRDIDECQQGNLCVNGQCKNTEGSFRCTCGQGYQLSA
AKDQCEDIDECQHRHLCAHGQCRNTEGSFQCVCDQGYRA
SGLGDHCEDINECLEDKSVCQRGDCINTAGSYDCTCPDGFQ
LDDNKTCQDINECEHPGLCGPQGECLNTEGSFHCVCQQGF
SISADGRTCEDIDECVNNTVCDSHGFCDNTAGSFRCLCYQG
FQAPQDGQGCVDVNECELLSGVCGEAFCENVEGSFLCVCA
DENQEYSPMTGQCRSRTSTDLDVDVDQPKEEKKECYYNL NDASLCDNVLAPNVTKQECCCTSGVGWGDNCEIFPCPVLG
TAEFTEMCPKGKGFVPAGESSSEAGGENYKDA
L1-E11-TB3EE EINECTVNPDICGAGHCINLPVRYTCICYEGYRFSEQQRKCV 144
DIDECTQVQHLCSQGRCENTEGSFLCICPAGFMASEEGTNCI
DVDECLRPDVCGEGHCVNTVGAFRCEYCDSGYRMTQRGR
CEDIDECLNPSTCPDEQCVNSPGSYQCVPCTEGFRGWNGQ
CLDVDECLEPNVCANGDCSNLEGSYMCSCHKGYTRTPDH
KHCRDIDECQQGNLCVNGQCKNTEGSFRCTCGQGYQLSA
AKDQCEDIDECQHRHLCAHGQCRNTEGSFQCVCDQGYRA
SGLGDHCEDINECLEDKSVCQRGDCINTAGSYDCTCPDGFQ
LDDNKTCQDINECEHPGLCGPQGECLNTEGSFHCVCQQGF
SISADGRTCEDIDECVNNTVCDSHGFCDNTAGSFRCLCYQG
FQAPQDGQGCVDVNECELLSGVCGEAFCENVEGSFLCVCA
DENQEYSPMTGQCRSRTSTDLDVDVDQPKEEKKECYYNL
NDASLCDNVLAPNVTKQECCCTSGVGWGDNCEIFPCPVLG
TAEFTEMCPKGKGFVPAGESSSEAGGENYKDADECLLFGQ
EICKNGFCLNTRPGYECYCKQGTYYDPVKLQCFDMDECQD
PSSCIDGQCVNTEGSYNCFCTHPMVLDASEKRCI
L1-AN441 STHPPPLPAKEEPVEALTFSREHGPGVAEPEVATAPPEKEIP 145
SLDQEKTKLEPGQPQLSPGISTIHLHPQFPVVIEKTSPPVPVE
VAPEASTSSASQVIAPTQVTEINECTVNPDICGAGHCINLPV
RYTCICYEGYRFSEQQRKCVDIDECTQVQHLCSQGRCENTE
GSFLCICPAGFMASEEGTNCIDVDECLRPDVCGEGHCVNTV
GAFRCEYCDSGYRMTQRGRCEDIDECLNPSTCPDEQCVNS
PGSYQCVPCTEGFRGWNGQCLDVDECLEPNVCANGDCSN
LEGSYMCSCHKGYTRTPDHKHCRDIDECQQGNLCVNGQC
KNTEGSFRCTCGQGYQLSAAKDQCEDIDECQHRHLCAHGQ
CRNTEGSFQCVCDQGYRASGLGDHCEDINECLEDKSVCQR
GDCINTAGSYDCTCPDGFQLDDNKTCQDINECEHPGLCGP
QGECLNTEGSFHCVCQQGFSISADGRTCEDIDECVNNTVCD
SHGFCDNTAGSFRCLCYQGFQAPQDGQGCVDVNECELLSG
VCGEAFCENVEGSFLCVCADENQEYSPMTGQCRSRTSTDL
DVDVDQPKEEKKECYYNLNDASLCDNVLAPNVTKQECCC
TSGVGWGDNCEIFPCPVLGTAEFTEMCPKGKGFVPAGESSS
EAGGENYKDADECLLFGQEICKNGFCLNTRPGYECYCKQG
TYYDPVKLQCFDMDECQDPSSCIDGQCVNTEGSYNCFCTH
PMVLDASEKRCIRPAESNEQIEETDVYQDLCWEHLSDEYV
CSRPLVGKQTTYTECCCLYGEAWGMQCALCPLKDSDDYA
QLCNIPVTGRRQPYGRDALVDFSEQYTPEADPYFIQDRFLN
SFEELQAEECGILNGCENGRCVRVQEGYTCDCFDGYHLDT
AKMTCVDVNECDELNNRMSLCKNAKCINTDGSYKCLCLP
GYVPSDKPNYCTPLNTALNLEKDSDLE
[00226] In some embodiments, LTBPs may comprise detectable labels. Detectable labels may be used to allow for detection and/or isolation of recombinant proteins comprising LTBPs. Some detectable labels may comprise biotin labels, polyhistidine tags and/or flag tags. Such tags may be used to isolate tagged proteins. Proteins produced may comprise additional amino acids encoding one or more 3C protease cleavage site. Such sites allow for cleavage at the 3C protease cleavage site upon treatment with 3C protease, including, but not limited to rhinovirus 3C protease. Such cleavage sites may be introduced to allow for removal of detectable labels from recombinant proteins.
[00227] Recombinant proteins of the present disclosure may be coexpressed with GDF- associated serum protein (GASP) 1 and/or GASP-2. Such recombinant proteins may include, but are not limited to GDF-8 and/or GDF-11. GASPs are circulating proteins that bind and prevent activity of GDF-8 and GDF- 11 (Hill, J.J. et al., 2003. Mol Endocrinology.
17(6): 1144-54 and Hill, J.J. et al., 2002. JBC. 277(43):40735-41, the contents of each of which are herein incorporated by reference in their entirety). Interestingly, GDF-8 and GDF- 11 growth factors are not found free in serum. About 70% are in GPCs with the remaining 30% associated with GASPs as well as other proteins (e.g. follistatin, follistatin-like related gene and decorin). Studies using mice lacking expression of GASP-1 (also known as WFIKKNRP or WFIKKN2) and/or GASP-2 (also known as WFIKKN or WFIKKN1) display phenotypes indicative of myostatin and/or GDF-11 overactivity (Lee et al., 2013. PNAS. 110(39):E3713-22). GASP bound GDF-8 and/or GDF-11 are unable to bind type II receptors and transmit related cellular signals. In some cases, recombinant proteins of the present disclosure may comprise one or more of the GASP sequences listed in Table 7. In some cases, these sequences are expressed in association with N- and/or C-terminal secretion signal sequences [e.g. human Ig kappa chains with amino acid sequence
MDMRVPAQLLGLLLLWFSGVLG (SEQ ID NO: 66)], flag tag sequences [e.g.
DYKDDDDK (SEQ ID NO: 67)], one or more 3C protease cleavage site [e.g. LEVLFQGP (SEQ ID NO: 68)], one or more biotinylation site and/or His-tag sequences [e.g. HHHHHH (SEQ ID NO: 69)].
Table 7. GASP Sequences
Protein Sequence SEQ
ID NO
GASP1, MWAPRCRRFWSRWEQVAALLLLLLLLGVPPRSL 150 NP_783165.1 ALPPIRYSHAGICPNDMNPNLWVDAQSTCRRECE
TDQECETYEKCCPNVCGTKSCVAARYMDVKGKK
GPVGMPKEATCDHFMCLQQGSECDIWDGQPVCK
CKDRCEKEPSFTCASDGLTYYNRCYMDAEACSK
GITLAVVTCRYHFTWPNTSPPPPETTMHPTTASPE
TPELDMAAPALLNNPVHQSVTMGETVSFLCDVV
GRPRPEITWEKQLEDRENVVMRPNHVRGNVVVT
NIAQLVIYNAQLQDAGIYTCTARNVAGVLRADFP
LSVVRGHQAAATSESSPNGTAFPAAECLKPPDSED
CGEEQTRWHFDAQANNCLTFTFGHCHRNLNHFE
TYEACMLACMSGPLAACSLPALQGPCKAYAPRW
AYNSQTGQCQSFVYGGCEGNGNNFESREACEESC PFPRGNQRCRACKPRQKLVTSFCRSDFVILGRVSE LTEEPDSGRALVTVDEVLKDEKMGLKFLGQEPLE VTLLHVDWACPCPNVTVSEMPLIIMGEVDGGMA MLRPDSFVGASSARRVRKLREVMHKKTCDVLKE FLGLH
GASP1, residue SVVRGHQAAATSESSPNGTAFPAAECLKPPDSED 151 303-576 of CGEEQTRWHFDAQANNCLTFTFGHCHRNLNHFE NP_783165.1 TYEACMLACMSGPLAACSLPALQGPCKAYAPRW
AYNSQTGQCQSFVYGGCEGNGNNFESREACEESC
PFPRGNQRCRACKPRQKLVTSFCRSDFVILGRVSE
LTEEPDSGRALVTVDEVLKDEKMGLKFLGQEPLE
VTLLHVDWACPCPNVTVSEMPLIIMGEVDGGMA
MLRPDSFVGASSARRVRKLREVMHKKTCDVLKE
FLGLH
GASP1, residue LPPIRYSHAGICPNDMNPNLWVDAQSTCRRECET 152 35-576 of DQECETYEKCCPNVCGTKSCVAARYMDVKGKK NP_783165.1 GPVGMPKEATCDHFMCLQQGSECDIWDGQPVCK
CKDRCEKEPSFTCASDGLTYYNRCYMDAEACSK
GITLAVVTCRYHFTWPNTSPPPPETTMHPTTASPE
TPELDMAAPALLNNPVHQSVTMGETVSFLCDVV
GRPRPEITWEKQLEDRENVVMRPNHVRGNVVVT
NIAQLVIYNAQLQDAGIYTCTARNVAGVLRADFP
LSVVRGHQAAATSESSPNGTAFPAAECLKPPDSED
CGEEQTRWHFDAQANNCLTFTFGHCHRNLNHFE
TYEACMLACMSGPLAACSLPALQGPCKAYAPRW
AYNSQTGQCQSFVYGGCEGNGNNFESREACEESC
PFPRGNQRCRACKPRQKLVTSFCRSDFVILGRVSE
LTEEPDSGRALVTVDEVLKDEKMGLKFLGQEPLE
VTLLHVDWACPCPNVTVSEMPLIIMGEVDGGMA
MLRPDSFVGASSARRVRKLREVMHKKTCDVLKE
FLGLH
GASP1, residue AACSLPALQGPCKAYAPRWAYNSQTGQCQSFVY 153 384-576 of GGCEGNGNNFESREACEESCPFPRGNQRCRACKP NP_783165.1 RQKLVTSFCRSDFVILGRVSELTEEPDSGRALVTV
DEVLKDEKMGLKFLGQEPLEVTLLHVDWACPCP
NVTVSEMPLIIMGEVDGGMAMLRPDSFVGASSAR
RVRKLREVMHKKTCDVLKEFLGLH
GASP1, residue FPRGNQRCRACKPRQKLVTSFCRSDFVILGRVSEL 154 438-576 of TEEPDSGRALVTVDEVLKDEKMGLKFLGQEPLEV NP_783165.1 TLLHVDWACPCPNVTVSEMPLIIMGEVDGGMAM
LRPDSFVGASSARRVRKLREVMHKKTCDVLKEFL GLH
GASP, MPALRPLLPLLLLLRLTSGAGLLPGLGSHPGVCPN 155 NP_444514.1 QLSPNLWVDAQSTCERECSRDQDCAAAEKCCINV
CGLHSCVAARFPGSPAAPTTAASCEGFVCPQQGS
DCDIWDGQPVCRCRDRCEKEPSFTCASDGLTYYN
RCYMDAEACLRGLHLHIVPCKHVLSWPPSSPGPP
ETTARPTPGAAPVPPALYSSPSPQAVQVGGTASLH
CDVSGRPPPAVTWEKQSHQRENLIMRPDQMYGN
VVVTSIGQLVLYNARPEDAGLYTCTARNAAGLLR
ADFPLSVVQREPARDAAPSIPAPAECLPDVQACTG
PTSPHLVLWHYDPQRGGCMTFPARGCDGAARGF
ETYEACQQACARGPGDACVLPAVQGPCRGWEPR
WAYSPLLQQCHPFVYGGCEGNGNNFHSRESCED ACPVPRTPPCRACRLRSKLALSLCRSDFAIVGRLT
EVLEEPEAAGGIARVALEDVLKDDKMGLKFLGTK
YLEVTLSGMDWACPCPNMTAGDGPLVIMGEVRD
GVAVLDAGSYVRAASEKRVKKILELLEKQACELL
NRFQD
GASP, residue SVVQREPARDAAPSIPAPAECLPDVQACTGPTSPH 156 279-548 of LVLWHYDPQRGGCMTFPARGCDGAARGFETYEA
NP_444514.1 CQQACARGPGDACVLPAVQGPCRGWEPRWAYSP
LLQQCHPFVYGGCEGNGNNFHSRESCEDACPVPR
TPPCRACRLRSKLALSLCRSDFAIVGRLTEVLEEPE
AAGGIARVALEDVLKDDKMGLKFLGTKYLEVTL
SGMDWACPCPNMTAGDGPLVIMGEVRDGVAVL
DAGSYVRAASEKRVKKILELLEKQACELLNRFQD
GASP, residue AGLLPGLGSHPGVCPNQLSPNLWVDAQSTCEREC 157 20-548 of SRDQDCAAAEKCCINVCGLHSCVAARFPGSPAAP
NP_444514.1 TTAASCEGFVCPQQGSDCDIWDGQPVCRCRDRCE
KEPSFTCASDGLTYYNRCYMDAEACLRGLHLHIV
PCKHVLSWPPSSPGPPETTARPTPGAAPVPPALYS
SPSPQAVQVGGTASLHCDVSGRPPPAVTWEKQSH
QRENLIMRPDQMYGNVVVTSIGQLVLYNARPED
AGLYTCTARNAAGLLRADFPLSVVQREPARDAAP
SIPAPAECLPDVQACTGPTSPHLVLWHYDPQRGG
CMTFPARGCDGAARGFETYEACQQACARGPGDA
CVLPAVQGPCRGWEPRWAYSPLLQQCHPFVYGG
CEGNGNNFHSRESCEDACPVPRTPPCRACRLRSKL
ALSLCRSDFAIVGRLTEVLEEPEAAGGIARVALED
VLKDDKMGLKFLGTKYLEVTLSGMDWACPCPN
MTAGDGPLVIMGEVRDGVAVLDAGSYVRAASEK
RVKKILELLEKQACELLNRFQD
GASP, residue DACVLPAVQGPCRGWEPRWAYSPLLQQCHPFVY 158 357-548 of GGCEGNGNNFHSRESCEDACPVPRTPPCRACRLR
NP_444514.1 SKLALSLCRSDFAIVGRLTEVLEEPEAAGGIARVA
LEDVLKDDKMGLKFLGTKYLEVTLSGMDWACPC
PNMTAGDGPLVIMGEVRDGVAVLDAGSYVRAAS
EKRVKKILELLEKQACELLNRFQD
GASP, residue VPRTPPCRACRLRSKLALSLCRSDFAIVGRLTEVL 159 411-548 of EEPEAAGGIARVALEDVLKDDKMGLKFLGTKYLE
NP_444514.1 VTLSGMDWACPCPNMTAGDGPLVIMGEVRDGV
AVLDAGSYVRAASEKRVKKILELLEKQACELLNR
FQD
[00228] Some recombinant proteins may be coexpressed with perlecan. Such recombinant proteins may include, but are not limited to GDF-8. Studies by Sengle et al (Sengle et al., 2011. J Biol Chem. 286(7):5087-99, the contents of which are herein incorporated by reference in their entirety) found that the GDF-8 prodomain associates with perlecan. Further studies indicate that perlecan knockout leads to muscular hypertrophy, suggesting that the interaction between GDF-8 and perlecan may contribute to GDF-8 activity (Xu et al. 2010. Matrix Biol. 29(6) :461-70). In some cases, recombinant proteins may comprise one or more of the perlecan sequences presented in Table 8. In some cases, these sequences are expressed in association with N- and/or C-terminal secretion signal sequences [e.g. human Ig kappa chains with amino acid sequence MDMRVPAQLLGLLLLWFSGVLG (SEQ ID NO: 66)], flag tag sequences [e.g. DYKDDDDK (SEQ ID NO: 67)], one or more 3C protease cleavage site [e.g. LEVLFQGP (SEQ ID NO: 68)], one or more biotinylation site and/or His-tag sequences [e.g. HHHHHH (SEQ ID NO: 69)].
Table 8. Perlecan sequences
Protein Sequence SEQ
ID NO
Perlecan, MGWRAAGALLLALLLHGRLLAVTHGLRAYDGLSLPEDIETVTA 160
NP_001278 SQMRWTHSYLSDDEDMLADSISGDDLGSGDLGSGDFQMVYFR
789.1 ALVNFTRSIEYSPQLEDAGSREFREVSEAVVDTLESEYLKIPGDQ
VVSVVFIKELDGWVFVELDVGSEGNADGAQIQEMLLRVISSGSV
ASYVTSPQGFQFRRLGTVPQFPRACTEAEFACHSYNECVALEYR
CDRRPDCRDMSDELNCEEPVLGISPTFSLLVETTSLPPRPETTIMR
QPPVTHAPQPLLPGSVRPLPCGPQEAACRNGHCIPRDYLCDGQE
DCEDGSDELDCGPPPPCEPNEFPCGNGHCALKLWRCDGDFDCE
DRTDEANCPTKRPEEVCGPTQFRCVSTNMCIPASFHCDEESDCP
DRSDEFGCMPPQVVTPPRESIQASRGQTVTFTCVAIGVPTPIINW
RLNWGHIPSHPRVTVTSEGGRGTLIIRDVKESDQGAYTCEAMNA
RGMVFGIPDGVLELVPQRAGPCPDGHFYLEHSAACLPCFCFGITS
VCQSTRRFRDQIRLRFDQPDDFKGVNVTMPAQPGTPPLSSTQLQI
DPSLHEFQLVDLSRRFLVHDSFWALPEQFLGNKVDSYGGSLRYN
VRYELARGMLEPVQRPDVVLMGAGYRLLSRGHTPTQPGALNQ
RQVQFSEEHWVHESGRPVQRAELLQVLQSLEAVLIQTVYNTKM
ASVGLSDIAMDTTVTHATSHGRAHSVEECRCPIGYSGLSCESCD
AHFTRVPGGPYLGTCSGCNCNGHASSCDPVYGHCLNCQHNTEG
PQCNKCKAGFFGDAMKATATSCRPCPCPYIDASRRFSDTCFLDT
DGQATCDACAPGYTGRRCESCAPGYEGNPIQPGGKCRPVNQEIV
RCDERGSMGTSGEACRCKNNVVGRLCNECADGSFHLSTRNPDG
CLKCFCMGVSRHCTSSSWSRAQLHGASEEPGHFSLTNAASTHTT
NEGIFSPTPGELGFSSFHRLLSGPYFWSLPSRFLGDKVTSYGGELR
FTVTQRSQPGSTPLHGQPLVVLQGNNIILEHHVAQEPSPGQPSTFI
VPFREQAWQRPDGQPATREHLLMALAGIDTLLIRASYAQQPAES
RVSGISMDVAVPEETGQDPALEVEQCSCPPGYRGPSCQDCDTGY
TRTPSGLYLGTCERCSCHGHSEACEPETGACQGCQHHTEGPRCE
QCQPGYYGDAQRGTPQDCQLCPCYGDPAAGQAAHTCFLDTDG
HPTCDACSPGHSGRHCERCAPGYYGNPSQGQPCQRDSQVPGPIG
CNCDPQGSVSSQCDAAGQCQCKAQVEGLTCSHCRPHHFHLSAS
NPDGCLPCFCMGITQQCASSAYTRHLISTHFAPGDFQGFALVNP
QRNSRLTGEFTVEPVPEGAQLSFGNFAQLGHESFYWQLPETYQG
DKVAAYGGKLRYTLSYTAGPQGSPLSDPDVQITGNNIMLVASQP
ALQGPERRSYEIMFREEFWRRPDGQPATREHLLMALADLDELLI
RATFSSVPLAASISAVSLEVAQPGPSNRPRALEVEECRCPPGYIGL
SCQDCAPGYTRTGSGLYLGHCELCECNGHSDLCHPETGACSQC
QHNAAGEFCELCAPGYYGDATAGTPEDCQPCACPLTNPENMFS
RTCESLGAGGYRCTACEPGYTGQYCEQCGPGYVGNPSVQGGQC
LPETNQAPLVVEVHPARSIVPQGGSHSLRCQVSGSPPHYFYWSR
EDGRPVPSGTQQRHQGSELHFPSVQPSDAGVYICTCRNLHQSNT
SRAELLVTEAPSKPITVTVEEQRSQSVRPGADVTFICTAKSKSPA YTLVWTRLHNGKLPTRAMDFNGILTIRNVQLSDAGTYVCTGSN
MFAMDQGTATLHVQASGTLSAPVVSIHPPQLTVQPGQLAEFRCS
ATGSPTPTLEWTGGPGGQLPAKAQIHGGILRLPAVEPTDQAQYL
CRAHSSAGQQVARAVLHVHGGGGPRVQVSPERTQVHAGRTVR
LYCRAAGVPSATITWRKEGGSLPPQARSERTDIATLLIPAITTAD
AGFYLCVATSPAGTAQARIQVVVLSASDASPPPVKIESSSPSVTE
GQTLDLNCVVAGSAHAQVTWYRRGGSLPPHTQVHGSRLRLPQ
VSPADSGEYVCRVENGSGPKEASITVSVLHGTHSGPSYTPVPGST
RPIRIEPSSSHVAEGQTLDLNCVVPGQAHAQVTWHKRGGSLPAR
HQTHGSLLRLHQVTPADSGEYVCHVVGTSGPLEASVLVTIEASV
IPGPIPPVRIESSSSTVAEGQTLDLSCVVAGQAHAQVTWYKRGGS
LPARHQVRGSRLYIFQASPADAGQYVCRASNGMEASITVTVTGT
QGANLAYPAGSTQPIRIEPSSSQVAEGQTLDLNCVVPGQSHAQV
TWHKRGGSLPVRHQTHGSLLRLYQASPADSGEYVCRVLGSSVP
LEASVLVTIEPAGSVPALGVTPTVRIESSSSQVAEGQTLDLNCLV
AGQAHAQVTWHKRGGSLPARHQVHGSRLRLLQVTPADSGEYV
CRVVGSSGTQEASVLVTIQQRLSGSHSQGVAYPVRIESSSASLAN
GHTLDLNCLVASQAPHTITWYKRGGSLPSRHQIVGSRLRIPQVTP
ADSGEYVCHVSNGAGSRETSLIVTIQGSGSSHVPSVSPPIRIESSSP
TVVEGQTLDLNCVVARQPQAIITWYKRGGSLPSRHQTHGSHLRL
HQMSVADSGEYVCRANNNIDALEASIVISVSPSAGSPSAPGSSMP
IRIESSSSHVAEGETLDLNCVVPGQAHAQVTWHKRGGSLPSHHQ
TRGSRLRLHHVSPADSGEYVCRVMGSSGPLEASVLVTIEASGSS
AVHVPAPGGAPPIRIEPSSSRVAEGQTLDLKCVVPGQAHAQVTW
HKRGGNLPARHQVHGPLLRLNQVSPADSGEYSCQVTGSSGTLE
ASVLVTIEPSSPGPIPAPGLAQPIYIEASSSHVTEGQTLDLNCVVPG
QAHAQVTWYKRGGSLPARHQTHGSQLRLHLVSPADSGEYVCR
AASGPGPEQEASFTVTVPPSEGSSYRLRSPVISIDPPSSTVQQGQD
ASFKCLIHDGAAPISLEWKTRNQELEDNVHISPNGSIITIVGTRPS
NHGTYRCVASNAYGVAQSVVNLSVHGPPTVSVLPEGPVWVKV
GKAVTLECVSAGEPRSSARWTRISSTPAKLEQRTYGLMDSHAVL
QISSAKPSDAGTYVCLAQNALGTAQKQVEVIVDTGAMAPGAPQ
VQAEEAELTVEAGHTATLRCSATGSPAPTIHWSKLRSPLPWQHR
LEGDTLIIPRVAQQDSGQYICNATSPAGHAEATIILHVESPPYATT
VPEHASVQAGETVQLQCLAHGTPPLTFQWSRVGSSLPGRATAR
NELLHFERAAPEDSGRYRCRVTNKVGSAEAFAQLLVQGPPGSLP
ATSIPAGSTPTVQVTPQLETKSIGASVEFHCAVPSDRGTQLRWFK
EGGQLPPGHSVQDGVLRIQNLDQSCQGTYICQAHGPWGKAQAS
AQLVIQALPSVLINIRTSVQTVVVGHAVEFECLALGDPKPQVTW
SKVGGHLRPGIVQSGGVVRIAHVELADAGQYRCTATNAAGTTQ
SHVLLLVQALPQISMPQEVRVPAGSAAVFPCIASGYPTPDISWSK
LDGSLPPDSRLENNMLMLPSVRPQDAGTYVCTATNRQGKVKAF
AHLQVPERVVPYFTQTPYSFLPLPTIKDAYRKFEIKITFRPDSADG
MLLYNGQKRVPGSPTNLANRQPDFISFGLVGGRPEFRFDAGSGM
ATIRHPTPLALGHFHTVTLLRSLTQGSLIVGDLAPVNGTSQGKFQ
GLDLNEELYLGGYPDYGAIPKAGLSSGFIGCVRELRIQGEEIVFH
DLNLTAHGISHCPTCRDRPCQNGGQCHDSESSSYVCVCPAGFTG
SRCEHSQALHCHPEACGPDATCVNRPDGRGYTCRCHLGRSGLR
CEEGVTVTTPSLSGAGSYLALPALTNTHHELRLDVEFKPLAPDG
VLLFSGGKSGPVEDFVSLAMVGGHLEFRYELGSGLAVLRSAEPL
ALGRWHRVSAERLNKDGSLRVNGGRPVLRSSPGKSQGLNLHTL
LYLGGVEPSVPLSPATNMSAHFRGCVGEVSVNGKRLDLTYSFLG
SQGIGQCYDSSPCERQPCQHGATCMPAGEYEFQCLCRDGFKGD
LCEHEENPCQLREPCLHGGTCQGTRCLCLPGFS GPRCQQGS GHG
IAESDWHLEGSGGNDAPGQYGAYFHDDGFLAFPGHVFSRSLPEV PETIELEVRTSTASGLLLWQGVEVGEAGQGKDFISLGLQDGHLV
FRYQLGSGEARLVSEDPINDGEWHRVTALREGRRGSIQVDGEEL VSGRSPGPNVAVNAKGSVYIGGAPDVATLTGGRFSSGITGCVKN LVLHSARPGAPPPQPLDLQHRAQAGANTRPCPS
Perlecan, AFAHLQVPERVVPYFTQTPYSFLPLPTIKDAYRKFEIKITFRPDSA 161 residues DGMLLYNGQKRVPGSPTNLANRQPDFISFGLVGGRPEFRFDAGS
3653-4392 GMATIRHPTPLALGHFHTVTLLRSLTQGSLIVGDLAPVNGTSQG
of KFQGLDLNEELYLGGYPDYGAIPKAGLSSGFIGCVRELRIQGEEI
NP_001278 VFHDLNLTAHGISHCPTCRDRPCQNGGQCHDSESSSYVCVCPAG
789.1 FTGSRCEHSQALHCHPEACGPDATCVNRPDGRGYTCRCHLGRS
GLRCEEGVTVTTPSLSGAGSYLALPALTNTHHELRLDVEFKPLA
PDGVLLFSGGKSGPVEDFVSLAMVGGHLEFRYELGSGLAVLRSA
EPLALGRWHRVSAERLNKDGSLRVNGGRPVLRSSPGKSQGLNL
HTLLYLGGVEPSVPLSPATNMSAHFRGCVGEVSVNGKRLDLTYS
FLGSQGIGQCYDSSPCERQPCQHGATCMPAGEYEFQCLCRDGFK
GDLCEHEENPCQLREPCLHGGTCQGTRCLCLPGFSGPRCQQGSG
HGIAESDWHLEGSGGNDAPGQYGAYFHDDGFLAFPGHVFSRSL
PEVPETIELEVRTSTASGLLLWQGVEVGEAGQGKDFISLGLQDG
HLVFRYQLGSGEARLVSEDPINDGEWHRVTALREGRRGSIQVDG
EELVSGRSPGPNVAVNAKGSVYIGGAPDVATLTGGRFSSGITGC
VKNLVLHSARPGAPPPQPLDLQHRAQAGANTRPCPS
[00229] In some cases, recombinant proteins of the disclosure may be coexpressed with follistatin and/or FLRG. Such recombinant proteins may include, but are not limited to GDF- 8. Both follistatin and FLRG are known to antagonize some TGF-β family member proteins, including, but not limited to GDF-8 (Lee, S-J. et al., 2010. Mol Endocrinol. 24(10): 1998- 2008, Takehara-Kasamatsu, Y. et al., 2007. J Med Invest. 54(3-4):276-88, the contents of each of which are herein incorporated by reference in their entirety). Follistatin has been shown to block GDF-8 activity by binding to the free growth factor and preventing receptor binding. Both follistatin and FLRG are implicated in modulating growth factor activity during development.
[00230] In some embodiments, recombinant proteins of the disclosure may be coexpressed with decorin. Such recombinant proteins may include, but are not limited to TGF-β and GDF- 8. Decorin is a known antagonist of TGF-β activity (Zhu, J. et al., 2007. J Biol Chem.
282:25852-63, the cotents of which are herein incorporated by reference in their entirety) and may also antagonize other TGF-β family members, including, but not limited to GDF-8. Decorin-dependent inhibition of TGF-β and GDF-8 activity has been shown to reduce fibrosis in various tissues. Decorin expression has also been shown to increase the expression of follistatin, a known inhibitor of free GDF-8.
[00231] In some embodiments, recombinant proteins of the present disclosure may comprise those depicted in FIG. 8. Some recombinant proteins of the present disclosure may comprise one or more features and/or combinations of protein modules from the embodiments depicted in FIG. 8.
Chimeric proteins
[00232] In some embodiments, recombinant proteins of the present disclosure may comprise chimeric proteins. As used herein, the term "chimeric protein" refers to a protein comprising one or more protein modules from at least two different proteins [formed from the same gene (e.g. variants arising from alternative splicing) or from different genes]. Chimeric proteins may comprise protein modules from two or more TGF-β family member proteins. Such chimeric proteins may comprise protein modules from TGF-βΙ, TGF-P2 and/or TGF- β3. Some chimeric proteins of the present disclosure may comprise protein modules including, but not limited to the protein modules and/or amino acid sequences listed in Table 9 (residue numbers correspond to the pro-protein sequences listed in Table 1). Some chimeric proteins of the present disclosure may comprise protein modules comprising amino acid sequences similar to those in Table 9, but comprising additional or fewer amino acids than those listed. Such modules may comprise about 1 more or fewer amino acids, about 2 more or fewer amino acids, about 3 more or fewer amino acids, about 4 more or fewer amino acids, about 5 more or fewer amino acids, about 6 more or fewer amino acids, about 7 more or fewer amino acids, about 8 more or fewer amino acids, about 9 more or fewer amino acids, about 10 more or fewer amino acids or greater than 10 more or fewer amino acids on N- terminal and/or C-terminal ends.
Table 9. Protein modules
Protein Residues Sequence SEQ
ID NO
GDF-8 1-75 NENSEQKENVEKEGLCNACTWRQNTKSSRIEAIKIQIL 162
SKLRLETAPNISKDVIRQLLPKAPPLRELIDQYDVQR
GDF-8 1-64 NENSEQKENVEKEGLCNACTWRQNTKSSRIEAIKIQIL 40
SKLRLETAPNISKDVIRQLLPKAPPL
GDF-8 75 - end RDDSSDGSLEDDDYHATTETIITMPTESDFLMQVDGK 163
PKCCFFKFSSKIQYNKVVKAQLWIYLRPVETPTTVFV
QILRLIKPMKDGTRYTGIRSLKLDMNPGTGIWQSIDVK
TVLQNWLKQPESNLGIEIKALDENGHDLAVTFPGPGE
DGLNPFLEVKVTDTPKRSRRDFGLDCDEHSTESRCCR
YPLTVDFEAFGWDWIIAPKRYKANYCSGECEFVFLQK
YPHTHLVHQANPRGSAGPCCTPTKMSPINMLYFNGK
EQIIYGKIPAMVVDRCGCS
GDF8 65 -end RELIDQYDVQRDDSSDGSLEDDDYHATTETIITMPTES 164
DFLMQVDGKPKCCFFKFSSKIQYNKVVKAQLWIYLR PVETPTTVFVQILRLIKPMKDGTRYTGIRSLKLDMNPG TGIWQSIDVKTVLQNWLKQPESNLGIEIKALDENGHD LAVTFPGPGEDGLNPFLEVKVTDTPKRSRRDFGLDCD
EHSTESRCCRYPLTVDFEAFGWDWIIAPKRYKANYCS GECEFVFLQKYPHTHLVHQANPRGSAGPCCTPTKMSP INMLYFNGKEQIIYGKIPAMVVDRCGCS
GDF8 65-243 RELIDQYDVQRDDSSDGSLEDDDYHATTETIITMPTES 45
DFLMQVDGKPKCCFFKFSSKIQYNKVVKAQLWIYLR
PVETPTTVFVQILRLIKPMKDGTRYTGIRSLKLDMNPG
TGIWQSIDVKTVLQNWLKQPESNLGIEIKALDENGHD
LAVTFPGPGEDGLNPFLEVKVTDTPKRSRR
GDF-8 76-243 DDSSDGSLEDDDYHATTETIITMPTESDFLMQVDGKP 165
KCCFFKFSSKIQYNKVVKAQLWIYLRPVETPTTVFVQI
LRLIKPMKDGTRYTGIRSLKLDMNPGTGIWQSIDVKT
VLQNWLKQPESNLGIEIKALDENGHDLAVTFPGPGED
GLNPFLEVKVTDTPKRSRR
GDF-8 244-352 DFGLDCDEHSTESRCCRYPLTVDFEAFGWDWIIAPKR 42
YKANYCSGECEFVFLQKYPHTHLVHQANPRGSAGPC CTPTKMSPINMLYFNGKEQIIYGKIPAMVVDRCGCS
GDF-11 1-86 AEGPAAAAAAAAAAAAAGVGGERSSRPAPSVAPEPD 41
GCPVCVWRQHSRELRLESIKSQILSKLRLKEAPNISRE
VVKQLLPKAPPL
GDF-11 1-96 AEGPAAAAAAAAAAAAAGVGGERSSRPAPSVAPEPD 166
GCPVCVWRQHSRELRLESIKSQILSKLRLKEAPNISRE
VVKQLLPKAPPLQQILDLHDFQ
GDF-11 1-108 AEGPAAAAAAAAAAAAAGVGGERSSRPAPSVAPEPD 167
GCPVCVWRQHSRELRLESIKSQILSKLRLKEAPNISRE
VVKQLLPKAPPLQQILDLHDFQGDALQPEDFLEE
GDF-11 97-274 GDALQPEDFLEEDEYHATTETVISMAQETDPAVQTDG 168
SPLCCHFHFSPKVMFTKVLKAQLWVYLRPVPRPATV
YLQILRLKPLTGEGTAGGGGGGRRHIRIRSLKIELHSR
SGHWQSIDFKQVLHSWFRQPQSNWGIEINAFDPSGTD
LAVTSLGPGAEGLHPFMELRVLENTKRSRR
GDF-11 87-274 QQILDLHDFQGDALQPEDFLEEDEYHATTETVISMAQ 46
ETDPAVQTDGSPLCCHFHFSPKVMFTKVLKAQLWVY LRPVPRPATVYLQILRLKPLTGEGTAGGGGGGRRHIRI RSLKIELHSRSGHWQSIDFKQVLHSWFRQPQSNWGIEI NAFDPSGTDLAVTSLGPGAEGLHPFMELRVLENTKRS RR
GDF-11 275-383 NLGLDCDEHSSESRCCRYPLTVDFEAFGWDWIIAPKR 43
YKANYCSGQCEYMFMQKYPHTHLVQQANPRGSAGP CCTPTKMSPINMLYFNDKQQIIYGKIPGMVVDRCGCS
Inhibin 1-64 SPTPGSEGHSAAPDCPSCALAALPKDVPNSQPEMVEA 169 Beta A VKKHILNMLHLKKRPDVTQPVPKAALL
Inhibin 1-76 SPTPGSEGHSAAPDCPSCALAALPKDVPNSQPEMVEA 170 Beta A VKKHILNMLHLKKRPDVTQPVPKAALLNAIRKLHVG
KVG
Inhibin 65-288 NAIRKLHVGKVGENGYVEIEDDIGRRAEMNELMEQT 171 Beta A SEIITFAESGTARKTLHFEISKEGSDLSVVERAEVWLFL
KVPKANRTRTKVTIRLFQQQKHPQGSLDTGEEAEEVG
LKGERSELLLSEKVVDARKSTWHVFPVSSSIQRLLDQ
GKSSLDVRIACEQCQESGASLVLLGKKKKKEEEGEGK
KKGGGEGGAGADEEKEQSHRPFLMLQARQSEDHPHR
RR
Inhibin 65-289 NAIRKLHVGKVGENGYVEIEDDIGRRAEMNELMEQT 172 Beta A SEIITFAESGTARKTLHFEISKEGSDLSVVERAEVWLFL KVPKANRTRTKVTIRLFQQQKHPQGSLDTGEEAEEVG
LKGERSELLLSEKVVDARKSTWHVFPVSSSIQRLLDQ
GKSSLDVRIACEQCQESGASLVLLGKKKKKEEEGEGK
KKGGGEGGAGADEEKEQSHRPFLMLQARQSEDHPHR
RRR
Inhibin 65-290 NAIRKLHVGKVGENGYVEIEDDIGRRAEMNELMEQT 173 Beta A SEIITFAESGTARKTLHFEISKEGSDLSVVERAEVWLFL
KVPKANRTRTKVTIRLFQQQKHPQGSLDTGEEAEEVG
LKGERSELLLSEKVVDARKSTWHVFPVSSSIQRLLDQ
GKSSLDVRIACEQCQESGASLVLLGKKKKKEEEGEGK
KKGGGEGGAGADEEKEQSHRPFLMLQARQSEDHPHR
RRRR
Inhibin 77-289 ENGYVEIEDDIGRRAEMNELMEQTSEIITFAESGTARK 174 Beta A TLHFEISKEGSDLSVVERAEVWLFLKVPKANRTRTKV
TIRLFQQQKHPQGSLDTGEEAEEVGLKGERSELLLSEK
VVDARKSTWHVFPVSSSIQRLLDQGKSSLDVRIACEQ
CQESGASLVLLGKKKKKEEEGEGKKKGGGEGGAGA
DEEKEQSHRPFLMLQARQSEDHPHRRRR
Inhibin 77-290 ENGYVEIEDDIGRRAEMNELMEQTSEIITFAESGTARK 175 Beta A TLHFEISKEGSDLSVVERAEVWLFLKVPKANRTRTKV
TIRLFQQQKHPQGSLDTGEEAEEVGLKGERSELLLSEK
VVDARKSTWHVFPVSSSIQRLLDQGKSSLDVRIACEQ
CQESGASLVLLGKKKKKEEEGEGKKKGGGEGGAGA
DEEKEQSHRPFLMLQARQSEDHPHRRRRR
Inhibin 77-end ENGYVEIEDDIGRRAEMNELMEQTSEIITFAESGTARK 176 Beta A TLHFEISKEGSDLSVVERAEVWLFLKVPKANRTRTKV
TIRLFQQQKHPQGSLDTGEEAEEVGLKGERSELLLSEK
VVDARKSTWHVFPVSSSIQRLLDQGKSSLDVRIACEQ
CQESGASLVLLGKKKKKEEEGEGKKKGGGEGGAGA
DEEKEQSHRPFLMLQARQSEDHPHRRRRRGLECDGK
VNICCKKQFFVSFKDIGWNDWIIAPSGYHANYCEGEC
PSHIAGTSGSSLSFHSTVINHYRMRGHSPFANLKSCCV
PTKLRPMSMLYYDDGQNIIKKDIQNMIVEECGCS
Inhibin 291-406 GLECDGKVNICCKKQFFVSFKDIGWNDWIIAPSGYHA 177 Beta A NYCEGECPSHIAGTSGSSLSFHSTVINHYRMRGHSPFA
NLKSCCVPTKLRPMSMLYYDDGQNIIKKDIQNMIVEE CGCS
[00233] In some embodiments, chimeric proteins of the present disclosure may comprise combinations of any of the protein modules listed in Table 9. Some chimeric proteins comprising GPCs may comprise protein modules that have been substituted with any of the protein modules listed in Table 9.
[00234] In some embodiments, chimeric proteins may comprise protein modules from GDFs and/or inhibins. Such GDFs may include GDF- 11 and/or GDF-8. Some such chimeric proteins may comprise a prodomain from GDF-11 and a growth factor from GDF-8. In such embodiments, chimeric proteins may comprise substituted N-terminal regions between GDF- 11 and GDF-8. In other embodiments, chimeric proteins may comprise a prodomain from GDF-8 and a growth factor from GDF-11. Such chimeric proteins may comprise amino acid residues 1-108 from GDF-11 and amino acid residues 90-the end of the protein from GDF-8. Some chimeric proteins may comprise an arm region from GDF-11.
[00235] Some chimerics of the present disclosure may comprise GDF-8 comprising an arm region of GDF-11. Such chimerics may be unstable due to steric clash between residue F95 from the GDF-11 arm and the a2 helix of the chimeric GPC. Therefore, in some cases, GDF8/GDF11/Activin chimeras may be designed so that the ARM region of such chimeric proteins contains the 2 helix. Furthermore, F95 may be an important residue in conferring latency for GDF11. This residue is in a similar position as a Camurati-Engelmann mutation found in TGF- 1, Y81H (see FIG. 6), thus, mutation of this residue to a smaller amino acid, such as an Alanine, may be carried out to promote dissociation of the mature GDF11 growth factor from the GPC. Such mutants may be useful as positive control molecules in designing assays to screen for GDF11 activating antibodies.
[00236] In some embodiments, chimeric proteins of the present disclosure may comprise protein module combinations including, but not limited to the combinations of protein modules and/or amino acid sequences listed in Table 10. Some chimeric proteins of the present disclosure may comprise protein modules comprising amino acid sequences similar to those in Table 10, but comprising additional or fewer amino acids than those listed. Such amino acid sequences may comprise about 1 more or fewer amino acids, about 2 more or fewer amino acids, about 3 more or fewer amino acids, about 4 more or fewer amino acids, about 5 more or fewer amino acids, about 6 more or fewer amino acids, about 7 more or fewer amino acids, about 8 more or fewer amino acids, about 9 more or fewer amino acids, about 10 more or fewer amino acids or greater than 10 more or fewer amino acids on N- terminal and/or C-terminal ends. In some cases, these sequences are expressed in association with N- and/or C-terminal secretion signal sequences [e.g. human Ig kappa chains with amino acid sequence MDMRVPAQLLGLLLLWFSGVLG (SEQ ID NO: 66)], flag tag sequences [e.g. DYKDDDDK (SEQ ID NO: 67)], one or more 3C protease cleavage site [e.g.
LEVLFQGP (SEQ ID NO: 68)], one or more biotinylation site and/or His-tag sequences [e.g. HHHHHH (SEQ ID NO: 69)].
Table 10. Protein module combinations
Protein Protein Protein Chimeric Sequence SEQ module module module ID 1 2 3 NO
GDF-11 GDF-8 GDF-11 AEGPAAAAAAAAAAAAAGVGGERSSRPAPSVAP 178 (1-96) (76-243) (275- EPDGCPVCVWRQHSRELRLESIKSQILSKLRLKEA
383) PNISREVVKQLLPKAPPLQQILDLHDFQDDSSDGS LEDDDYHATTETIITMPTESDFLMQVDGKPKCCFF
KFSSKIQYNKVVKAQLWIYLRPVETPTTVFVQILR
LIKPMKDGTRYTGIRSLKLDMNPGTGIWQSIDVKT
VLQNWLKQPESNLGIEIKALDENGHDLAVTFPGP
GEDGLNPFLEVKVTDTPKRSRRNLGLDCDEHSSE
SRCCRYPLTVDFEAFGWDWIIAPKRYKANYCSGQ
CEYMFMQKYPHTHLVQQANPRGSAGPCCTPTKM
SPINMLYFNDKQQIIYGKIPGMVVDRCGCS
GDF-11 GDF-8 GDF-11 AEGPAAAAAAAAAAAAAGVGGERSSRPAPSVAP 179 (1-86) (65-243) (275- EPDGCPVCVWRQHSRELRLESIKSQILSKLRLKEA
383) PNISREVVKQLLPKAPPLRELIDQYDVQRDDSSDG
SLEDDDYHATTETIITMPTESDFLMQVDGKPKCCF
FKFSSKIQYNKVVKAQLWIYLRPVETPTTVFVQIL
RLIKPMKDGTRYTGIRSLKLDMNPGTGIWQSIDV
KTVLQNWLKQPESNLGIEIKALDENGHDLAVTFP
GPGEDGLNPFLEVKVTDTPKRSRRNLGLDCDEHS
SESRCCRYPLTVDFEAFGWDWIIAPKRYKANYCS
GQCEYMFMQKYPHTHLVQQANPRGSAGPCCTPT
KMSPINMLYFNDKQQIIYGKIPGMVVDRCGCS
GDF-11 GDF-8 N/A AEGPAAAAAAAAAAAAAGVGGERSSRPAPSVAP 180 (1-96) (76-243) EPDGCPVCVWRQHSRELRLESIKSQILSKLRLKEA
PNISREVVKQLLPKAPPLQQILDLHDFQDDSSDGS
LEDDDYHATTETIITMPTESDFLMQVDGKPKCCFF
KFSSKIQYNKVVKAQLWIYLRPVETPTTVFVQILR
LIKPMKDGTRYTGIRSLKLDMNPGTGIWQSIDVKT
VLQNWLKQPESNLGIEIKALDENGHDLAVTFPGP
GEDGLNPFLEVKVTDTPKRSRR
GDF-11 GDF-8 NA AEGPAAAAAAAAAAAAAGVGGERSSRPAPSVAP 181 (1-86) (65-243) EPDGCPVCVWRQHSRELRLESIKSQILSKLRLKEA
PNISREVVKQLLPKAPPLRELIDQYDVQRDDSSDG
SLEDDDYHATTETIITMPTESDFLMQVDGKPKCCF
FKFSSKIQYNKVVKAQLWIYLRPVETPTTVFVQIL
RLIKPMKDGTRYTGIRSLKLDMNPGTGIWQSIDV
KTVLQNWLKQPESNLGIEIKALDENGHDLAVTFP
GPGEDGLNPFLEVKVTDTPKRSRR
GDF-11 Inhibin GDF-11 AEGPAAAAAAAAAAAAAGVGGERSSRPAPSVAP 182 (1-96) Beta A (275- EPDGCPVCVWRQHSRELRLESIKSQILSKLRLKEA
(77-290) 383) PNISREVVKQLLPKAPPLQQILDLHDFQENGYVEI
EDDIGRRAEMNELMEQTSEIITFAESGTARKTLHF
EISKEGSDLSVVERAEVWLFLKVPKANRTRTKVTI
RLFQQQKHPQGSLDTGEEAEEVGLKGERSELLLS
EKVVDARKSTWHVFPVSSSIQRLLDQGKSSLDVRI
ACEQCQESGASLVLLGKKKKKEEEGEGKKKGGG
EGGAGADEEKEQSHRPFLMLQARQSEDHPHRRR
RRNLGLDCDEHSSESRCCRYPLTVDFEAFGWDWI
IAPKRYKANYCSGQCEYMFMQKYPHTHLVQQAN
PRGSAGPCCTPTKMSPINMLYFNDKQQIIYGKIPG
MVVDRCGCS
GDF-11 Inhibin GDF-11 AEGPAAAAAAAAAAAAAGVGGERSSRPAPSVAP 183 (1-86) Beta A (275- EPDGCPVCVWRQHSRELRLESIKSQILSKLRLKEA
(65-290) 383) PNISREVVKQLLPKAPPLNAIRKLHVGKVGENGY
VEIEDDIGRRAEMNELMEQTSEIITFAESGTARKTL
HFEISKEGSDLSVVERAEVWLFLKVPKANRTRTK
VTIRLFQQQKHPQGSLDTGEEAEEVGLKGERSELL LSEKVVDARKSTWHVFPVSSSIQRLLDQGKSSLD
VRIACEQCQESGASLVLLGKKKKKEEEGEGKKKG
GGEGGAGADEEKEQSHRPFLMLQARQSEDHPHR
RRRRNLGLDCDEHSSESRCCRYPLTVDFEAFGWD
WIIAPKRYKANYCSGQCEYMFMQKYPHTHLVQQ
ANPRGSAGPCCTPTKMSPINMLYFNDKQQIIYGKI
PGMVVDRCGCS
GDF-11 Inhibin N/A AEGPAAAAAAAAAAAAAGVGGERSSRPAPSVAP 184 (1-96) Beta A EPDGCPVCVWRQHSRELRLESIKSQILSKLRLKEA
(77-290) PNISREVVKQLLPKAPPLQQILDLHDFQENGYVEI
EDDIGRRAEMNELMEQTSEIITFAESGTARKTLHF
EISKEGSDLSVVERAEVWLFLKVPKANRTRTKVTI
RLFQQQKHPQGSLDTGEEAEEVGLKGERSELLLS
EKVVDARKSTWHVFPVSSSIQRLLDQGKSSLDVRI
ACEQCQESGASLVLLGKKKKKEEEGEGKKKGGG
EGGAGADEEKEQSHRPFLMLQARQSEDHPHRRR
RR
GDF-11 Inhibin NA AEGPAAAAAAAAAAAAAGVGGERSSRPAPSVAP 185 (1-86) Beta A EPDGCPVCVWRQHSRELRLESIKSQILSKLRLKEA
(65-290) PNISREVVKQLLPKAPPLNAIRKLHVGKVGENGY
VEIEDDIGRRAEMNELMEQTSEIITFAESGTARKTL
HFEISKEGSDLSVVERAEVWLFLKVPKANRTRTK
VTIRLFQQQKHPQGSLDTGEEAEEVGLKGERSELL
LSEKVVDARKSTWHVFPVSSSIQRLLDQGKSSLD
VRIACEQCQESGASLVLLGKKKKKEEEGEGKKKG
GGEGGAGADEEKEQSHRPFLMLQARQSEDHPHR
RRRR
GDF-8 GDF-11 GDF-8 NENSEQKENVEKEGLCNACTWRQNTKSSRIEAIKI 186 (1-75) (97-274) (244- QILS KLRLET APNIS KD VIRQLLPKAPPLRELIDQ Y
352) DVQRGDALQPEDFLEEDEYHATTETVISMAQETD
PAVQTDGSPLCCHFHFSPKVMFTKVLKAQLWVY
LRPVPRPATVYLQILRLKPLTGEGTAGGGGGGRR
HIRIRSLKIELHSRSGHWQSIDFKQVLHSWFRQPQS
NWGIEINAFDPSGTDLAVTSLGPGAEGLHPFMELR
VLENTKRSRRDFGLDCDEHSTESRCCRYPLTVDFE
AFGWDWIIAPKRYKANYCSGECEFVFLQKYPHTH
LVHQANPRGSAGPCCTPTKMSPINMLYFNGKEQII
YGKIPAMVVDRCGCS
GDF-8 GDF-11 GDF-8 NENSEQKENVEKEGLCNACTWRQNTKSSRIEAIKI 187 (1-64) (87-274) (244- QILS KLRLET APNIS KD VIRQLLPKAPPLQQILDLH
352) DFQGDALQPEDFLEEDEYHATTETVISMAQETDP
AVQTDGSPLCCHFHFSPKVMFTKVLKAQLWVYL
RPVPRPATVYLQILRLKPLTGEGTAGGGGGGRRHI
RIRSLKIELHSRSGHWQSIDFKQVLHSWFRQPQSN
WGIEINAFDPSGTDLAVTSLGPGAEGLHPFMELRV
LENTKRSRRDFGLDCDEHSTESRCCRYPLTVDFEA
FGWDWIIAPKRYKANYCSGECEFVFLQKYPHTHL
VHQANPRGSAGPCCTPTKMSPINMLYFNGKEQIIY
GKIPAMVVDRCGCS
GDF-8 GDF-11 N/A NENSEQKENVEKEGLCNACTWRQNTKSSRIEAIKI 188 (1-75) (97-274) QILS KLRLET APNIS KD VIRQLLPKAPPLRELIDQ Y
DVQRGDALQPEDFLEEDEYHATTETVISMAQETD PAVQTDGSPLCCHFHFSPKVMFTKVLKAQLWVY LRPVPRPATVYLQILRLKPLTGEGTAGGGGGGRR HIRIRSLKIELHSRSGHWQSIDFKQVLHSWFRQPQS
NWGIEINAFDPSGTDLAVTSLGPGAEGLHPFMELR VLENTKRSRR
GDF-8 GDF-11 GDF-8 NENSEQKENVEKEGLCNACTWRQNTKSSRIEAIKI 189 (1-64) (87-274) (244- QILS KLRLET APNIS KD VIRQLLPKAPPLQQILDLH
352) DFQGDALQPEDFLEEDEYHATTETVISMAQETDP
AVQTDGSPLCCHFHFSPKVMFTKVLKAQLWVYL
RPVPRPATVYLQILRLKPLTGEGTAGGGGGGRRHI
RIRSLKIELHSRSGHWQSIDFKQVLHSWFRQPQSN
WGIEINAFDPSGTDLAVTSLGPGAEGLHPFMELRV
LENTKRSRRDFGLDCDEHSTESRCCRYPLTVDFEA
FGWDWIIAPKRYKANYCSGECEFVFLQKYPHTHL
VHQANPRGSAGPCCTPTKMSPINMLYFNGKEQIIY
GKIPAMVVDRCGCS
GDF-8 Inhibin GDF-8 NENSEQKENVEKEGLCNACTWRQNTKSSRIEAIKI 190 (1-75) Beta A (244- QILS KLRLET APNIS KD VIRQLLPKAPPLRELIDQ Y
(77-289) 352) DVQRENGYVEIEDDIGRRAEMNELMEQTSEIITFA
ES GT ARKTLHFEIS KEGSDLS V VER AEV WLFLKVP
KANRTRTKVTIRLFQQQKHPQGSLDTGEEAEEVG
LKGERSELLLSEKVVDARKSTWHVFPVSSSIQRLL
DQGKSSLDVRIACEQCQESGASLVLLGKKKKKEE
EGEGKKKGGGEGGAGADEEKEQSHRPFLMLQAR
QSEDHPHRRRRDFGLDCDEHSTESRCCRYPLTVD
FEAFGWDWIIAPKRYKANYCSGECEFVFLQKYPH
THLVHQANPRGSAGPCCTPTKMSPINMLYFNGKE
QIIYGKIPAMVVDRCGCS
GDF-8 Inhibin GDF-8 NENSEQKENVEKEGLCNACTWRQNTKSSRIEAIKI 191 (1-64) Beta A (244- QILS KLRLET APNIS KD VIRQLLPKAPPLNAIRKLH
(65-290) 352) VGKVGENGYVEIEDDIGRRAEMNELMEQTSEIITF
AES GT ARKTLHFEIS KEGSDLS V VER AEV WLFLK
VPKANRTRTKVTIRLFQQQKHPQGSLDTGEEAEE
VGLKGERSELLLSEKVVDARKSTWHVFPVSSSIQR
LLDQGKSSLDVRIACEQCQESGASLVLLGKKKKK
EEEGEGKKKGGGEGGAGADEEKEQSHRPFLMLQ
ARQSEDHPHRRRRRDFGLDCDEHSTESRCCRYPL
TVDFEAFGWDWIIAPKRYKANYCSGECEFVFLQK
YPHTHLVHQANPRGSAGPCCTPTKMSPINMLYFN
GKEQIIYGKIPAMVVDRCGCS
GDF-8 Inhibin N/A NENSEQKENVEKEGLCNACTWRQNTKSSRIEAIKI 192 (1-75) Beta A QILS KLRLET APNIS KD VIRQLLPKAPPLRELIDQ Y
(77-290) DVQRENGYVEIEDDIGRRAEMNELMEQTSEIITFA
ES GT ARKTLHFEIS KEGSDLS V VER AEV WLFLKVP
KANRTRTKVTIRLFQQQKHPQGSLDTGEEAEEVG
LKGERSELLLSEKVVDARKSTWHVFPVSSSIQRLL
DQGKSSLDVRIACEQCQESGASLVLLGKKKKKEE
EGEGKKKGGGEGGAGADEEKEQSHRPFLMLQAR
QSEDHPHRRRRR
GDF-8 Inhibin NA NENSEQKENVEKEGLCNACTWRQNTKSSRIEAIKI 193 (1-64) Beta A QILS KLRLET APNIS KD VIRQLLPKAPPLNAIRKLH
(65-290) VGKVGENGYVEIEDDIGRRAEMNELMEQTSEIITF
AES GT ARKTLHFEIS KEGSDLS V VER AEV WLFLK
VPKANRTRTKVTIRLFQQQKHPQGSLDTGEEAEE
VGLKGERSELLLSEKVVDARKSTWHVFPVSSSIQR
LLDQGKSSLDVRIACEQCQESGASLVLLGKKKKK EEEGEGKKKGGGEGGAGADEEKEQSHRPFLMLQ
ARQSEDHPHRRRRR
Inhibin GDF-8 Inhibin SPTPGSEGHSAAPDCPSCALAALPKDVPNSQPEM 194 Beta A (76-243) Beta A VEAVKKHILNMLHLKKRPDVTQPVPKAALLNAIR (1-76) (291- KLHVGKVGDDSSDGSLEDDDYHATTETIITMPTES
406) DFLMQVDGKPKCCFFKFSSKIQYNKVVKAQLWIY
LRPVETPTTVFVQILRLIKPMKDGTRYTGIRSLKLD
MNPGTGIWQSIDVKTVLQNWLKQPESNLGIEIKA
LDENGHDLAVTFPGPGEDGLNPFLEVKVTDTPKR
SRRGLECDGKVNICCKKQFFVSFKDIGWNDWIIAP
SGYHANYCEGECPSHIAGTSGSSLSFHSTVINHYR
MRGHSPFANLKSCCVPTKLRPMSMLYYDDGQNII
KKDIQNMIVEECGCS
Inhibin GDF-8 Inhibin SPTPGSEGHSAAPDCPSCALAALPKDVPNSQPEM 195 Beta A (65-243) Beta A VEAVKKHILNMLHLKKRPDVTQPVPKAALLRELI (1-64) (291- DQYDVQRDDSSDGSLEDDDYHATTETIITMPTES
406) DFLMQVDGKPKCCFFKFSSKIQYNKVVKAQLWIY
LRPVETPTTVFVQILRLIKPMKDGTRYTGIRSLKLD
MNPGTGIWQSIDVKTVLQNWLKQPESNLGIEIKA
LDENGHDLAVTFPGPGEDGLNPFLEVKVTDTPKR
SRRGLECDGKVNICCKKQFFVSFKDIGWNDWIIAP
SGYHANYCEGECPSHIAGTSGSSLSFHSTVINHYR
MRGHSPFANLKSCCVPTKLRPMSMLYYDDGQNII
KKDIQNMIVEECGCS
Inhibin GDF-8 N/A SPTPGSEGHSAAPDCPSCALAALPKDVPNSQPEM 196 Beta A (76-243) VEAVKKHILNMLHLKKRPDVTQPVPKAALLNAIR (1-76) KLHVGKVGDDSSDGSLEDDDYHATTETIITMPTES
DFLMQVDGKPKCCFFKFSSKIQYNKVVKAQLWIY
LRPVETPTTVFVQILRLIKPMKDGTRYTGIRSLKLD
MNPGTGIWQSIDVKTVLQNWLKQPESNLGIEIKA
LDENGHDLAVTFPGPGEDGLNPFLEVKVTDTPKR
SRR
Inhibin GDF-8 NA SPTPGSEGHSAAPDCPSCALAALPKDVPNSQPEM 197 Beta A (65-243) VEAVKKHILNMLHLKKRPDVTQPVPKAALLRELI (1-64) DQYDVQRDDSSDGSLEDDDYHATTETIITMPTES
DFLMQVDGKPKCCFFKFSSKIQYNKVVKAQLWIY
LRPVETPTTVFVQILRLIKPMKDGTRYTGIRSLKLD
MNPGTGIWQSIDVKTVLQNWLKQPESNLGIEIKA
LDENGHDLAVTFPGPGEDGLNPFLEVKVTDTPKR
SRR
Inhibin GDF-11 Inhibin SPTPGSEGHSAAPDCPSCALAALPKDVPNSQPEM 198 Beta A (97-274) Beta A VEAVKKHILNMLHLKKRPDVTQPVPKAALLNAIR (1-76) (291- KLHVGKVGGDALQPEDFLEEDEYHATTETVISMA
406) QETDPAVQTDGSPLCCHFHFSPKVMFTKVLKAQL
WVYLRPVPRPATVYLQILRLKPLTGEGTAGGGGG
GRRHIRIRSLKIELHSRSGHWQSIDFKQVLHSWFR
QPQSNWGIEINAFDPSGTDLAVTSLGPGAEGLHPF
MELRVLENTKRSRRGLECDGKVNICCKKQFFVSF
KDIGWNDWII APS GYH ANYCEGECPSHI AGTS GSS
LSFHSTVINHYRMRGHSPFANLKSCCVPTKLRPMS
MLYYDDGQNIIKKDIQNMIVEECGCS
Inhibin GDF-11 Inhibin SPTPGSEGHSAAPDCPSCALAALPKDVPNSQPEM 199 Beta A (87-274) Beta A VEAVKKHILNMLHLKKRPDVTQPVPKAALLQQIL (1-64) (291- DLHDFQGDALQPEDFLEEDEYHATTETVISMAQE 406) TDPAVQTDGSPLCCHFHFSPKVMFTKVLKAQLW
VYLRPVPRPATVYLQILRLKPLTGEGTAGGGGGG
RRHIRIRSLKIELHSRSGHWQSIDFKQVLHSWFRQ
PQSNWGIEINAFDPSGTDLAVTSLGPGAEGLHPFM
ELRVLENTKRSRRGLECDGKVNICCKKQFFVSFK
DIGWNDWIIAPSGYHANYCEGECPSHIAGTSGSSL
SFHSTVINHYRMRGHSPFANLKSCCVPTKLRPMS
MLYYDDGQNIIKKDIQNMIVEECGCS
Inhibin GDF-11 N/A SPTPGSEGHSAAPDCPSCALAALPKDVPNSQPEM 200 Beta A (97-274) VEAVKKHILNMLHLKKRPDVTQPVPKAALLNAIR
(1-76) KLHVGKVGGDALQPEDFLEEDEYHATTETVISMA
QETDPAVQTDGSPLCCHFHFSPKVMFTKVLKAQL
WVYLRPVPRPATVYLQILRLKPLTGEGTAGGGGG
GRRHIRIRSLKIELHSRSGHWQSIDFKQVLHSWFR
QPQSNWGIEINAFDPSGTDLAVTSLGPGAEGLHPF
MELRVLENTKRSRR
Inhibin GDF-11 NA SPTPGSEGHSAAPDCPSCALAALPKDVPNSQPEM 201 Beta A (87-274) VEAVKKHILNMLHLKKRPDVTQPVPKAALLQQIL
(1-64) DLHDFQGDALQPEDFLEEDEYHATTETVISMAQE
TDPAVQTDGSPLCCHFHFSPKVMFTKVLKAQLW
VYLRPVPRPATVYLQILRLKPLTGEGTAGGGGGG
RRHIRIRSLKIELHSRSGHWQSIDFKQVLHSWFRQ
PQSNWGIEINAFDPSGTDLAVTSLGPGAEGLHPFM
ELRVLENTKRSRR
[00237] Chimeric proteins may be used to characterize and/or map epitopes associated with GPCs. As used herein, the terms "map" or "mapping" refer to the identification,
characterization and/or determination of one or more functional regions of one or more proteins. Such characterizations may be necessary for determining interactions between one or more protein modules and another agent (e.g. another protein and/or protein module). Some chimeric proteins may be used to characterize functions associated with one or more proteins and/or protein modules.
[00238] In some embodiments, chimeric proteins of the disclosure may comprise GPCs with trigger loops from alternative TGF-β family member proteins. Such chimeric proteins may have altered properties with regard to integrin-dependent activation. According to mouse tissue staining, integrin subunit g is widely expressed in skeletal and cardiac muscle, visceral smooth muscle, hepatocytes, airway epithelium, squamous epithelium, choroid plexus epithelium and also on neutrophils (Palmer, E.L. et al., Sequence and tissue distribution of the integrin 0 subunit, a novel partner of βΐ that is widely distributed in epithelia and muscle. Journal of Cell Biology. 1993. 123(5): 1289-97). Expression of g is not detected earlier than E12.5, suggesting that it does not play a major role in the earliest tissue morphogenesis (Wang, A. et al., Expression of the integrin subunit ( 9 in the murine embryo. Developmental Dynamics. 1995. 204:421-31). In vivo functions of ( 9 are unclear. Phenotypes observed in knockout mice suggest a role in lymphatic valve development (Bazigou, E. et al., Integrin- g is required for fibronectin matrix assembly during lymphatic valve morphogenesis. Dev Cell. 2009 August. 17(2): 175-86). Reported interaction partners of integrin agfii include VCAM-1, the third Fnlll domain on tenascin C, osteopontin, polydom/SVEPl, VEGF-A and NGF (Yokasaki, Y. et al., Identification of the ligand binding site for the integrin agfii in the third fibronectin type III repeat of tenascin C. The Journal of Biological Chemistry. 1998. 273(19): 11423-8; Marcinkiewicz, C. et al., Inhibitory effects of MLDG-containing heterodimeric disintegrins reveal distinct structural requirements for interaction of the integrin agfii with VCAM- 1, tenascin-C, and osteopontin. JBC. 2000.
275(41):31930-7; Oommen, S. et al., Vacular endothelial growth factor A (VEGF-A) induces endothelial and cancer cell migration through direct binding to integrin agfii . JBC. 2011. 286(2): 1083-92; Sato-Nishiuchi, R. et al., Polydom/SVEPl is a ligand for integrin 9Pi. JBC. 2012. 287(30):25615-30; Staniszewska, I. et al., Integrin agfii is a receptor for nerve growth factor and other neurotrophins. Journal of Cell Science. 2007. 121 (Pt 4):504- 13; Yokosaki, Y. et al., The integrin agfii binds to a novel recognition sequence (SVVYGLR; SEQ ID NO: 202) in the thrombin-cleaved amino-terminal fragment of osteopontin. JBC. 1999.
274(51):36328-34).
[00239] Binding sites on proteins that interact with agfii have been mapped using linear peptides. These sites include binding sites on tenascin C (AEIDGIEL; SEQ ID NO: 203), osteopontin (SVVYGLR; SEQ ID NO: 202), polydom/SVEPl (EDDMME VP Y ; SEQ ID NO: 204) and VEGF-A (EYP). Unlike a4Pi and α5β1 ; agfii does not require a canonical RGD sequence motif. Some, but not all reported targets have an acidic residue/hydrophobic residue/proline motif. Some also comprise a tyrosine residue.
[00240] The trigger loop of TGF-βΙ and TGF-P3 carries an RGD sequence where ανβό and/or νβ8 bind to enable growth factor release. The TGF-P2 trigger loop region is different from those of TGF-βΙ and TGF-P3, comprising the sequence
FAGIDGTSTYTSGDQKTIKSTRKKNSGKTP (SEQ ID NO: 205), without an RGD trimer. Of this region, residues AGIDGTST (SEQ ID NO: 206) align with the peptide on the third Fnlll domain of tenascin-C that has been mapped as an agfii binding site. Also, the tyrosine following this region may play a role in potential agfii binding. Therefore, agfii binding to TGF-P2 could be physiologically relevant. In some embodiments, chimeric proteins of the present disclosure may comprise trigger loop sequences comprising any of the sequences listed in Table 11. Table 11. Trigger loop sequences
Figure imgf000116_0001
[00241] In some embodiments, chimeric proteins of the present disclosure may comprise one or more TGF-P2 trigger loops. Such chimeric proteins may exhibit activation (e.g.
growth factor release) regulated in a manner similar to that of TGF-P2. Some chimeric proteins of the present disclosure may comprise TGF-P-related proteins wherein one or more protein modules are substituted with one or more protein modules comprising one or more TGF-P2 trigger loops. Some chimeric proteins comprise TGF-P-related proteins wherein one or more protein modules comprising at least one RGD sequence are substituted with one or more protein modules comprising one or more TGF-P2 trigger loops. In other embodiments, chimeric proteins may comprise TGF-βΙ and/or TGF-P3 proteins wherein one or more protein modules comprising at least one RGD sequence are substituted with one or more protein modules comprising one or more TGF-P2 trigger loops. Such chimeric proteins may exhibit TGF-βΙ activity.
[00242] In some embodiments, chimeric proteins of the present disclosure may comprise one or more protein modules from BMPs. Protein modules comprising sequences from BMPs may comprise sequences from any of those BMP modules disclosed in FIG. 6. Chimeric proteins of the present disclosure comprising one or more BMP protein module may be useful for the development of antibodies and/or assays to study, enhance and/or perturb BMP interactions with other proteins, including, but not limited to RGM proteins.
[00243] Chimeric proteins may comprise detectable labels. Detectable labels may be used to allow for detection and/or isolation of chimeric proteins. Such detectable labels may comprise biotin labels, polyhistidine tags and/or flag tags. Tags may be used to identify and/or isolate tagged proteins. Proteins produced may comprise additional amino acids encoding one or more 3C protease cleavage site. Such sites allow for cleavage at the 3C protease cleavage site upon treatment with 3C protease, including, but not limited to rhinovirus 3C protease. 3C protease cleavage sites may be introduced to allow for removal of detectable labels from chimeric proteins.
Protein expression
[00244] In some embodiments, synthesis of recombinant proteins of the present disclosure may be carried out according to any method known in the art. Some protein synthesis may be carried out in vitro. Some protein synthesis may be carried out using cells. Such cells may be bacterial and/or eukaryotic. In some embodiments, eukaryotic cells may be used for protein synthesis. Some such cells may be mammalian. Some mammalian cells used for protein expression may include, but are not limited to mouse cells, rabbit cells, rat cells, monkey cells, hamster cells and human cells. Such cells may be derived from a cell line. In other embodiments, human cells may be used. In further embodiments, cell lines may include, but are not limited to HEK293 cells, CHO cells, HeLa cells, Sw-480 cells, EL4 T lymphoma cells, TMLC cells, 293T/17 cells, Hs68 cells, CCD1112sk cells, HFF-1 cells, Keloid fibroblasts, A204 cells, L17 RIB cells, C2C12 cells and HepG2 cells.
[00245] In some embodiments, 293 cells are used for synthesis of recombinant proteins of the present disclosure. These cells are human cells that post-translationally modify proteins with human-like structures (e.g. glycans). Such cells are easily transfectable and scalable and are able to grow to high densities in suspension culture. 293 cells may include 293E cells. 293E cells are HEK293 cells stably expressing EBNA1 (Epstein-Barr virus nuclear antigen- 1). In some cases, 293E cells may be grown in serum-free medium to simplify down-stream purification. In some cases, 293-6E cells (NRC Canada, Ottawa, CA) may be used. Such cells express truncated EBNA1 (EBNAlt) and may comprise enhanced production of recombinant proteins and may be optimized for growth and/or protein expression in serum-free medium to simplify down-stream purification. In some cases, insect cells may be used to express recombinant proteins of the disclosure. In some cases, insect cell expression may be carried out using Spodoptera frugiperda cells including, but not limited to Sf9 and/or Sf-21 cells. In some cases, insect cell cultures may comprise Trichoplusia ni cells, including, but not limited to Tn-368 and/or HIGH-FIVE™ BTI-TN-5B1-4 cells. A further list of exemplary insect cell lines can be found in US Patent No. 5,024,947, the contents of which are herein incorporated by reference in their entirety.
[00246] In some embodiments, recombinant proteins of the disclosure may comprise an antibody Fc domain to create an Fc fusion protein. The formation of an Fc fusion protein with any of the recombinant proteins described herein may be carried out according to any method known in the art, including as described in Czajkowsky, D.M. et al., 2012. EMBO Mol Med. 4(10): 1015-28 and US Patent Nos. 5,116,964, 5,541,087 and 8,637,637, the contents of each of which are herein incorporated by reference in their entirety. In some cases, proteins that may be difficult to express may be expressed as Fc fusion proteins to enhance protein stability and allow for expression and, in some cases, subsequent use as antigens. Fc fusion proteins of the disclosure may be linked to the hinge region of an IgG Fc via cysteine residues in the Fc hinge region. Resulting Fc fusion proteins may comprise an antibody-like structure, but without CHI domains or light chains. In some cases, Fc fusion proteins may comprise pharmacokinetic profiles comparable to native antibodies. In some cases, Fc fusion proteins of the disclosure may comprise an extended half-life in circulation and/or altered biological activity. In some cases, Fc fusion proteins of the disclosure may be prepared using any of the TGF-β family proteins or TGF-P-related proteins described herein. In some cases, Fc fusion proteins may comprise TGF-β, GDF-8 and/or GDF-11.
[00247] In some embodiments, Fc domains may comprise the amino acid sequence DKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWY VDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIE KTISKAKGQPREPQVYTLPPSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENN YKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG
K (SEQ ID NO: 222). Some examples of Fc fusion proteins of the disclosure are presented in Table 12 below. Included in the Table, is an Fc fusion protein comprising a mutated GDF-11 prodomain sequence. The mutation is in a site that may, in some cases, be subjected to cleavage (e.g. by furin or similar enzyme). Fusion proteins with the mutation indicated may be resistant to such cleavage and thereby retain association with the Fc domain. Further, the fusion proteins listed comprise an N-terminal secretion signal and and C-terminal histidine tag; however, variants without such elements are also contemplated.
Table 12. Fc fusion proteins
Protein Amino Acid Sequence SEQ
ID NO
GDF-8 MDMRVPAQLLGLLLLWFSGVLGNENSEQKENVEKEGLCNACT 223 prodomai WRQNTKSSRIEAIKIQILSKLRLETAPNISKDVIRQLLPKAPPLRELI n Fc- DQYDVQRDDSSDGSLEDDDYHATTETIITMPTESDFLMQVDGKP fusion KCCFFKFSSKIQYNKVVKAQLWIYLRPVETPTTVFVQILRLIKPM protein KDGTRYTGIRSLKLDMNPGTGIWQSIDVKTVLQNWLKQPESNLG
IEIKALDENGHDLAVTFPGPGEDGLNPFLEVKVTDTPKRSDKTHT
CPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDP
EVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDW
LNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSREEM
TKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDG
SFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG
KHHHHHH
GDF-8 MDMRVPAQLLGLLLLWFSGVLGNENSEQKENVEKEGLCNACT 224 prodomai WRQNTKSSRIEAIKIQILSKLRLETAPNISKDVIRQLLPKAPPLRELI n D76A DQYDVQRADSSDGSLEDDDYHATTETIITMPTESDFLMQVDGKP
Fc-fusion KCCFFKFSSKIQYNKVVKAQLWIYLRPVETPTTVFVQILRLIKPM protein KDGTRYTGIRSLKLDMNPGTGIWQSIDVKTVLQNWLKQPESNLG
IEIKALDENGHDLAVTFPGPGEDGLNPFLEVKVTDTPKRSDKTHT
CPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDP
EVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDW
LNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSREEM
TKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDG
SFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG
KHHHHHH
mutant 225 GDF-11 MDMRVPAQLLGLLLLWFSGVLGAEGPAAAAAAAAAAAAAGVG prodomai GEASSAPAPSVAPEPDGCPVCVWRQHSRELRLESIKSQILSKLRL n Fc- KEAPNISREVVKQLLPKAPPLQQILDLHDFQGDALQPEDFLEEDE fusion YHATTETVISMAQETDPAVQTDGSPLCCHFHFSPKVMFTKVLKA protein QLWVYLRPVPRPATVYLQILRLKPLTGEGTAGGGGGGRRHIRIRS
LKIELHSRSGHWQSIDFKQVLHSWFRQPQSNWGIEINAFDPSGTD
LAVTSLGPGAEGLHPFMELRVLENTKRSDKTHTCPPCPAPELLGG
PSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDG
VEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKV
SNKALPAPIEKTISKAKGQPREPQVYTLPPSREEMTKNQVSLTCL
VKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTV
DKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGKHHHHHH
GDF-11 MDMRVPAQLLGLLLLWFSGVLGAEGPAAAAAAAAAAAAAGVG 226 prodomai GERSSRPAPSVAPEPDGCPVCVWRQHSRELRLESIKSQILSKLRLK n Fc- EAPNISREVVKQLLPKAPPLQQILDLHDFQGDALQPEDFLEEDEY fusion HATTETVISMAQETDPAVQTDGSPLCCHFHFSPKVMFTKVLKAQ protein LWVYLRPVPRPATVYLQILRLKPLTGEGTAGGGGGGRRHIRIRSL
KIELHSRSGHWQSIDFKQVLHSWFRQPQSNWGIEINAFDPSGTDL
AVTSLGPGAEGLHPFMELRVLENTKRSDKTHTCPPCPAPELLGGP
SVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGV
EVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVS NKALPAPIEKTISKAKGQPREPQVYTLPPSREEMTKNQVSLTCLV
KGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVD I KSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGKHHHHHH |
[00248] Sequences encoding recombinant proteins of the present disclosure may be inserted into any number of DNA vectors known in the art for expression. Such vectors may include plasmids. In some embodiments, sequences encoding recombinant proteins of the present disclosure are cloned into pTT5 vectors (NRC Biotechnology Research Institute, Montreal, Quebec). In other embodiments pTT22, pTT28, pYD5, pYD7, pYDl l(NRC Biotechnology Institute, Montreal, Quebec) and/or pMA vectors (Life Technologies, Carlsbad, CA) may be used. Vectors may comprise promoter sequences to modulate expression of sequences encoding recombinant proteins of the present disclosure. Such promoters may be constitutively active and/or may be regulated by extrinsic and/or intrinsic factors. Some extrinsic factors may be used to enhance or suppress expression of sequences encoding recombinant proteins of the present disclosure. Some vectors may encode nuclear localization signals that may be incorporated into recombinant proteins of the present disclosure upon translation. Some vectors may produce mRNA transcripts that comprise nuclear export signals. RNA transcribed from a modified pTT5 vector (pTT5-WPRE) contains an element that facilitates nuclear export of the transcripts. Some vectors may be modified by insertion of one or more ligation-independent cloning (LIC) cassettes to provide for simpler cloning.
[00249] Vectors encoding recombinant proteins of the present disclosure may be delivered to cells according to any method known in the art, including, but not limited to transfection, electroporation and/or transduction. In some embodiments, vectors may comprise one or more elements to enhance vector replication in host cells. In some embodiments, vectors may comprise oriP sites for episomal replication in cells that express EBNA-1.
[00250] In some cases, cells are stably transfected to produce recombinant proteins of the present disclosure. Stably transfected cells pass transfected genes to daughter cells during cell division, thus eliminating the need for repeated transfection. In some cases, the transfected genes are stably inserted into the genome of the transfected cells. Transfected genes may comprise genes for cell selection, such as genes that confer resistance to one or more toxic or repressive compounds. Such genes may be used to support the growth of only cells with stable incorporation of the transfected genes when grown in the presence of such one or more toxic or repressive compounds (e.g. puromycin, kanamycin, etc.). Cell selection may also comprise selecting cells based on overall recombinant protein expression levels. Determination of such levels may be carried out, for example, by Western Blot and/or ELISA.
[00251] In some embodiments, nucleotide sequences encoding recombinant proteins of the present disclosure may comprise one or more woodchuck hepatitis virus posttranscriptional regulatory element (WPRE). RNA nucleic acids comprising such elements may comprise the sequence
GCCACGGCGGAACUCAUCGCCGCCUGCCUUGCCCGCUGCUGGACAGGGGCUCG GCUGUUGGGCACUGACAAUUCCGUGGU (SEQ ID NO: 227). RNA comprising WPREs may be transcribed from DNA comprising the sequence
AATCAACCTCTGGATTACAAAATTTGTGAAAGATTGACTGGTATTCTTAACTATG
TTGCTCCTTTTACGCTATGTGGATACGCTGCTTTAATGCCTTTGTATCATGCTATT
GCTTCCCGTATGGCTTTCATTTTCTCCTCCTTGTATAAATCCTGGTTGCTGTCTCTT
TATGAGGAGTTGTGGCCCGTTGTCAGGCAACGTGGCGTGGTGTGCACTGTGTTTG
CTGACGCAACCCCCACTGGTTGGGGCATTGCCACCACCTGTCAGCTCCTTTCCGG
GACTTTCGCTTTCCCCCTCCCTATTGCCACGGCGGAACTCATCGCCGCCTGCCTTG
CCCGCTGCTGGACAGGGGCTCGGCTGTTGGGCACTGACAATTCCGTGGTGTTGTC
GGGGAAGCTGACGTCCTTTCCATGGCTGCTCGCCTGTGTTGCCACCTGGATTCTG
CGCGGGACGTCCTTCTGCTACGTCCCTTCGGCCCTCAATCCAGCGGACCTTCCTTC
CCGCGGCCTGCTGCCGGCTCTGCGGCCTCTTCCGCGTCTTCGCCTTCGCCCTCAGA
CGAGTCGGATCTCCCTTTGGGCCGCCTCCCCGCCTG (SEQ ID NO: 228). WPREs may enhance translation of nucleic acids comprising WPREs. Such enhanced translation may be due to increased cytoplasmic export of newly transcribed mRNA.
[00252] In some embodiments, recombinant proteins may comprise one or more secretion signal sequences. As used herein, the term "secretion signal sequence" refers to a chain of amino acids (or nucleotides that encode them at the nucleic acid level) that when part of a protein, modulate secretion of such proteins from cells. Some secretion signal sequences may be located at protein termini. In other embodiments, secretion signal sequences may be N- terminal amino acid sequences. Other secretions signal sequences may comprise the secretion signal of the Ig kappa chains. Such Ig kappa chains may be human Ig kappa chains. In some embodiments, secretion signal sequences may comprise the amino acid sequence
MDMRVPAQLLGLLLLWFSGVLG (SEQ ID NO: 66).
[00253] In some embodiments, recombinant proteins of the present disclosure may require coexpression with one or more other proteins for proper expression, folding, secretion, activity and/or function. Some recombinant GPCs of the present disclosure may be coexpressed with LTBPs, fibrillins, GASPs, perlecan and/or decorin.
[00254] In some embodiments, recombinant proteins of the present disclosure may be biotinylated. As used herein, the term "biotinylating" refers to the attaching of one or more biotin labels. Such biotin labels may facilitate interactions of biotinylated recombinant proteins with avidin and/or streptavidin coated surfaces and/or proteins. As used herein, a "biotin label" refers to a detectable label comprising one or more biotin molecules. The term "biotinylated" refers to a molecule or protein that comprises one or more biotin labels. Biotin molecules bind with high affinity to avidin and streptavidin molecules. This property may be used to capture biotinylated proteins using avidin and/or stretavidin coated materials. Some recombinant GPCs of the present disclosure may be biotinylated near the N-terminus. Such recombinant GPCs may be introduced to avidin/streptavidin coated cell culture surfaces, allowing biotinylated recombinant GPCs to adhere to the surface in a manner such that the orientation and bonding of such bound GPCs mimics the orientation and bonding of GPCs to extracellular proteins (e.g. LTBPs, fibrillins, GASPs, perlecan and/or decorin).
[00255] In some embodiments, recombinant proteins produced may be analyzed for quality control purposes to assess both biophysical properties as well as bioactive properties.
Biophysical characterization may include assessing protein migration patterns after reducing and/or non-reducing SDS PAGE. Biophysical characterization may also comprise gel filtration, mass spectrometric analysis and/or analysis of association/dissociation between LAPs or LAP-like domains and growth factor domains. Bioactive properties may be analyzed by assessing reactivity with antibodies and/or signaling activity of dissociated growth factors and/or latent GPCs.
[00256] Some proteins produced may comprise additional amino acids encoding one or more detectable labels for purification [e.g. polyhistidine tag, flag tag, etc.] In some embodiments, proteins are N-terminally labeled. In some embodiments, proteins are C- terminally labeled. In some embodiments, proteins are biotinylated. In some embodiments, recombinant proteins of the present disclosure are N-terminally biotinylated.
[00257] Proteins produced may comprise additional amino acids encoding one or more 3C protease cleavage site. Such sites allow for cleavage between residues Q and G of the 3C protease cleavage site upon treatment with 3C protease, including, but not limited to rhinovirus 3C protease. In some embodiments, such cleavage sites are introduced to allow for removal of detectable labels from recombinant proteins. [00258] In some embodiments, modification of expressed growth factor proproteins may be carried out by enzymatic cleavage. In some cases, proprotein convertases may be used. Such proprotein convertases may include, but are not limited to furin/PACE3, PCl/3, PC2, PC4, PC5/6, PACE4 and PC7. Proprotein convertase cleavage may be carried out in solution or in tissue culture. In some cases, proprotein convertases are expressed in cells expressing proproteins to be cleaved. In some cases, proprotein convertases are added to tissue cultures of cells expressing proproteins to be cleaved.
Antibodies
[00259] In some embodiments, compounds and/or compositions of the present disclosure may comprise antibodies or fragments thereof. As used herein, the term "antibody" is referred to in the broadest sense and specifically covers various embodiments including, but not limited to monoclonal antibodies, polyclonal antibodies, multispecific antibodies (e.g.
bispecific antibodies formed from at least two intact antibodies), and antibody fragments such as diabodies so long as they exhibit a desired biological activity. Antibodies are primarily amino-acid based molecules but may also comprise one or more modifications (including, but not limited to the addition of sugar moieties, fluorescent moieties, chemical tags, etc.).
Recombinant and chimeric protein use in antibody generation
[00260] In some embodiments, recombinant and/or chimeric proteins described herein may be used as antigens (referred to herein as antigenic proteins) to generate antibodies. Such antigenic proteins may comprise epitopes that may be less accessible for antibody generation in similar wild type proteins. Some antibodies directed to antigenic proteins of the present disclosure may modulate the release of one or more growth factors from one or more GPCs). Some such antibodies may be stabilizing [reducing or preventing dissociation between two agents, (e.g. growth-factor release from GPCs, GPC release from one or more protein interactions)] and/or releasing [enhancing the dissociation between two agents (e.g. growth- factor release from GPCs, GPC release from one or more protein interactions)] antibodies. Antigenic proteins of the present disclosure may comprise TGF-P-related proteins as well as components and/or protein modules thereof. In some cases, antigenic proteins of the present disclosure may comprise prodomains without associated growth factors, furin cleavage- deficient mutants, mutants deficient in extracellular protein associations and/or combinations thereof. [00261] In some embodiments, antigenic proteins may comprise TGF-P-related proteins and/or modules thereof. Such antigenic proteins may comprise epitopes from regions where growth factors associate with or comprise stereological proximity with prodomain regions. Antibodies of the present disclosure directed to such epitopes may bind overlapping regions between growth factors and prodomains. Such antibodies may stereologically inhibit the dissociation of growth factors from GPCs.
[00262] In some embodiments, antigenic proteins comprise only the prodomain or only the growth factor from a particular GPC. Epitopes present on such antigenic proteins may be shielded or unexposed in intact GPCs. Some antibodies of the present disclosure may be directed to such epitopes. Such antibodies may be releasing antibodies, promoting growth factor dissociation from GPCs. Further antibodies may compete with free growth factor for prodomain binding, thereby promoting growth factor dissociation from GPCs.
[00263] In some embodiments, antigenic proteins may comprise proprotein convertase (e.g. furin) cleavage site mutations. Such mutations may prevent enzymatic cleavage of growth factors from their prodomains. Some antibodies of the present disclosure may be directed to epitopes present on such mutant proteins. Such antibodies may stabilize the association between prodomains and growth factors. In some embodiments, furin cleavage site mutants comprise D2G mutants as described herein.
[00264] In some embodiments, antigenic proteins comprising prodomains may comprise N- terminal mutations that lead to decreased prodomain association with extracellular proteins and therefore may present epitopes in the N-terminal region that may otherwise be shielded by those associations. Some antibodies of the present disclosure may be directed to such epitopes.
[00265] In some embodiments, antigenic proteins may comprise one or more recombinant LTBP. Such recombinant LTBPs may comprise LTBP1, LTBP2, LTBP3, LTBP4, alternatively spliced variants and/or fragments thereof. Recombinant LTBPs may also be modified to comprise one or more detectable labels. Such detectable labels may include, but are not limited to biotin labels, polyhistidine tags, myc tags, HA tags and/or fluorescent tags.
[00266] In some embodiments, antigenic proteins may comprise one or more recombinant protein and/or chimeric protein complexed with one or more recombinant LTBP. Some antigenic proteins may comprise proprotein convertase cleavage site mutants (e.g. D2G mutants, AXXA mutants) complexed with one or more recombinant LTBP. Some such recombinant LTBPs may comprise LTBPIS. Some recombinant LTBPs may comprise one or more detectable labels, including, but not limited to biotin labels, polyhistidine tags and/or flag tags.
[00267] In some embodiments, antigenic proteins of the present disclosure may comprise one or more protein modules from GDFs (e.g. GDF-11 and/or GDF-8). In some
embodiments, antibodies of the present disclosure may be directed toward antigenic proteins comprising GDF-8 protein modules. In some embodiments, such antibodies may modulate GDF-8 levels and/or activity in one or more niches. In some embodiments, antibodies of the present disclosure may prevent the release of GDF-8 growth factors from GPCs. In some embodiments, antibodies of the present disclosure may be used to repair and/or enhance muscle tissues.
[00268] In some embodiments, recombinant proteins (including, but not limited to chimeric proteins) described herein may be used in studies to identify and map epitopes that may be important targets for antibody development. Such studies may be used to identify epitopes that may promote growth factor release or stabilization of GPCs upon antibody binding.
[00269] In some cases, recombinant proteins of the disclosure may comprise recombinant binding proteins, including, but not limited to antibodies, antibody fragments and fusion proteins comprising one or more antibodies or antibody fragments. Such recombinant binding proteins may comprise one or more regions from one or more antibodies developed using one or more recombinant antigens described herein.
Releasing antibodies
[00270] As used herein, the term "releasing antibody" refers to an antibody that increases the ratio of active and/or free growth factor relative to inactive and/or prodomain-associated growth factor upon the introduction of the antibody to a GPC, cell, niche, natural depot or any other site of growth factor sequestration. In this context, releasing antibodies may be characterized as agonists. As used herein, the term "natural depot" refers to a location within a cell, tissue or organ where increased levels of a biomolecule or ion are stored. For example, the extracellular matrix may act as a natural depot for one or more growth factors.
[00271] The contact necessary for growth-factor release may be defined as direct or indirect contact of antibody with a GPC or a component thereof or with a cellular structure such as an extracellular and/or cellular matrix protein and/or protein associated with the extracellular and/or cellular matrix [e.g. LTBPs (e.g. LTBP1, LTBP2, LTBP3 and/or LTBP4), fibrillins (e.g. fibrillin- 1, fibrillin-2, fibrillin-3 and/or fibrillin-4), perlecan, decorin, elastin, collagen, GASPs and/or GARPs (e.g. GARP and/or LRRC33)] for release of growth factor. Release of at least 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or more of growth factor is sufficient to characterize antibodies of the present disclosure as releasing antibodies. It is understood that growth factor release after antibody administration may be local and may occur over a sustained period of time and may include peaks or spikes of release. Antibodies of the present disclosure may act to release one or more growth factor over minutes, hours, days or longer.
[00272] Release profiles may have an initial peak or burst within from about 4 hours to about 7 days of contacting in vivo or shorter periods in vitro. For example, initial peak or burst may occur from about 4 hours to about 5 hours, or from about 4 hours to about 6 hours, or from about 4 hours to about 7 hours, or from about 4 hours to about 8 hours, or from about 4 hours to about 9 hours, or from about 4 hours to about 10 hours, or from about 4 hours to about 11 hours, or from about 4 hours to about 12 hours, or from about 4 hours to about 24 hours, or from about 4 hours to about 36 hours, or from about 4 hours to about 48 hours, or from about 1 day to about 7 days, or from about 1 day to about 2 days, or from about 1 day to about 3 days, or from about 1 day to about 4 days, or from about 4 days to about 5 days, or from about 4 days to about 6 days, or from about 4 days to about 7 days. Compounds and/or compositions of the present disclosure may stimulate the release of 5 to 100% of the growth factor present. For example, the percent of growth factor release may be from about 5% to about 10%, or from about 5% to about 15%, or from about 5% to about 20%, or from about 5% to about 25%, or from about 10% to about 30%, or from about 10% to about 40%, or from about 10% to about 50%, or from about 10% to about 60%, or from about 20% to about 70%, or from about 20% to about 80%, or from about 40% to about 90%, or from about 40% to about 100%.
[00273] Releasing antibodies generated according to methods described herein may be generated to release growth factors from GPCs comprising any of the pro-proteins listed in Table 1. In some cases, releasing antibodies are directed to GPCs comprising GDFs and/or one or more modules from GDFs.
Stabilizing antibodies
[00274] As used herein, the term "stabilizing antibody" refers to an antibody that decreases the ratio of active and/or free growth factor relative to inactive and/or prodomain-associated growth factor upon the introduction of the antibody to one or more GPC, cell, niche, natural depot and/or any other site of growth factor sequestration. In this context, antibodies may be characterized as antagonists. As used herein, an "antagonist" is one which interferes with or inhibits the physiological action of another. Antagonist action may even result in stimulation or activation of signaling downstream and hence may act agonistically relative to another pathway, separate from the one being antagonized. Pathways are interrelated, so, in one nonlimiting example, a TGF-β antagonist could act as a BMP agonist and vice versa. In the context of cellular events, as used herein, the term "downstream" refers to any signaling or cellular event that happens after the action, binding or targeting by compounds and/or compositions of the present disclosure.
[00275] Contact necessary for inhibition or stabilization may be direct or indirect contact between antibody and GPC or components thereof or with cellular structures such as an extracellular and/or cellular matrix protein and/or protein associated with the extracellular and/or cellular matrix [e.g. LTBPs (e.g. LTBP1, LTBP2, LTBP3 and/or LTBP4), fibrillins (e.g. fibrillin- 1, fibrillin-2, fibrillin-3 and/or fibrillin-4), perlecan, decorin, elastin, collagen, GASPs and/or GARPs (e.g. GARP and/or LRRC33)] whereby release of growth factor is inhibited. Inhibition of release of at least 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or more of growth factors may be sufficient, in some cases, to characterize antibodies of the present disclosure as inhibitory or stabilizing. Inhibitory antibodies may stabilize GPCs and trap them as heterodimers.
[00276] It is understood that inhibition of growth factor release after contact with one or more antibodies of the present disclosure may be local and may occur over a sustained period of time and may include peaks, troughs or spikes. Inhibitory antibodies which may also function to stabilize GPCs may be defined by their release kinetics. Release of growth factor and corresponding release kinetics, even locally, may be directly measured or inferred by downstream signaling events. In some embodiments, changes in protein or nucleic acid concentrations or phenotypic responses may be indicative of the effects of compounds and/or compositions of the present disclosure.
[00277] Antibodies of the present disclosure may act to inhibit release of a growth factor over minutes, hours or days. Inhibition and/or stabilization profiles may have an initial trough within from about 4 hours to about 7 days of introduction in vivo or shorter periods in vitro. For example, initial trough of inhibition or stabilization may occur from about 4 hours to about 5 hours, or from about 4 hours to about 6 hours, or from about 4 hours to about 7 hours, or from about 4 hours to about 8 hours, or from about 4 hours to about 9 hours, or from about 4 hours to about 10 hours, or from about 4 hours to about 11 hours, or from about 4 hours to about 12 hours, or from about 4 hours to about 24 hours, or from about 4 hours to about 36 hours, or from about 4 hours to about 48 hours, or from about 1 day to about 7 days, or from about 1 day to about 2 days, or from about 1 day to about 3 days, or from about 1 day to about 4 days, or from about 4 days to about 5 days, or from about 4 days to about 6 days, or from about 4 days to about 7 days. Introduction of compounds and/or compositions of the present disclosure may lead to inhibition and/or stabilization of 5% to 100% of growth factor present. For example, the percent of growth factor inhibition or stabilization may be from about 5% to about 10%, from about 5% to about 15%, from about 5% to about 20%, from about 5% to about 25%, from about 10% to about 30%, from about 10% to about 40%, from about 10% to about 50%, from about 10% to about 60%, from about 20% to about 70%, from about 20% to about 80%, from about 40% to about 90% or from about 40% to about 100%.
[00278] Stabilizing antibodies generated according to methods described herein may be generated to block the release of growth factors from GPCs comprising any of the pro- proteins listed in Table 1. Such antibodies may physically interact with GPC protease cleavage sites and/or block the interaction of proteolytic enzymes that may target such cleavage sites. In some cases, stabilizing antibodies are directed to GPCs comprising GDFs and/or one or more modules from GDFs.
[00279] Stabilizing antibodies directed to GPCs comprising GDF-8 may block
metalloproteinase cleavage of such complexes. Such agents may bind to GPCs comprising GDF-8 in such a way as to physically prevent interactions between such GPCs and metalloproteinases targeting such GPCs. Agents that actually target metalloproteinases themselves have been described previously (see US Patent No. US 7,572,599, the contents of which are herein incorporated by reference in their entirety).
Antibody selection
[00280] A desired antibody may be selected from a larger pool of two or more candidate antibodies based on the desired antibody's ability to associate with desired antigens and/or epitopes. Such antigens and/or epitopes may include, but are not limited to any of those described herein, including, but not limited to recombinant proteins, chimeric proteins, GPCs, prodomains (e.g. LAPs or LAP-like domains), growth factors, protein modules, LTBPs, fibrillins, GASPs, TGF-P-related proteins and/or mutants and/or variants and/or complexes and/or combinations thereof. Selection of desired antibodies may be carried out using an antibody binding assay, such as a surface Plasmon resonance-based assay, an enzyme-linked immunosorbent assay (ELISA) or fluorescence flow cytometry-based assay. Such assays may utilize a desired antigen to bind a desired antibody and then use one or more detection methods to detect binding. [00281] In some embodiments, antibodies of the present disclosure may be selected from a larger pool of two or more candidate antibodies based on their ability to associate with desired antigens and/or epitopes from multiple species (referred to herein as "positive selection.")
[00282] In some embodiments, such species may comprise vertebrate species. In some embodiments, such species may comprise mammalian species. In some embodiments, such species may include, but are not limited to mice, rats, rabbits, goats, sheep, pigs, horses, cows and/or humans.
[00283] In some embodiments, negative selection is used to remove antibodies from a larger pool of two or more candidate antibodies. As used herein the term "negative selection" refers to the elimination of one or more factors from a group based on their ability to bind to one or more undesired antigens and/or epitopes. In some embodiments, undesired antigens and/or epitopes may include, but are not limited to any of those described herein, including, but not limited to recombinant proteins, chimeric proteins, GPCs, prodomains (e.g. LAPs or LAP-like domains), growth factors, protein modules, LTBPs, fibrillins, GASPs, TGF-β- related proteins and/or mutants and/or variants and/or combinations and/or complexes thereof.
[00284] In some embodiments, antibodies of the present disclosure may be directed to prodomains (e.g. the prodomain portion of a GPC and/or free LAP or LAP-like domains) that decrease growth factor signaling and/or levels (e.g. GDF growth factor signaling and/or levels) in a given niche. In some embodiments, antibodies of the present disclosure directed to prodomains may increase growth factor signaling and/or levels in a given niche. In some embodiments, antibodies of the present disclosure may be directed to prodomains (e.g. LAPs or LAP-like domains) and/or GPCs only when complexed with one or more extracellular protein, such as LTBPs, fibrillins, perlican, decorin and/or GASPs.
[00285] In some embodiments, antibodies of the present disclosure may be selected from a larger pool of two or more candidate antibodies based on their ability to modulate growth factor levels and/or activity. In some cases, growth factor activity assays may be used to test the ability of candidate antibodies to modulate growth factor activity. Growth factor activity assays may include, cell-based assays as described hereinbelow. Additional assays that may be used to determine the effect of candidate antibodies on growth factor activity may include, but are not limited to enzyme-linked immunosorbent assay (ELISA), Western blotting, reporter assays (e.g. luciferase-based reporter assays or other enzyme-based reporter assays), PCR analysis, RT-PCR analysis and/or other methods known in the art including any of the methods described in International Patent Application No. WO2014074532, the contents of which are herein incorporated by reference in their entirety.
[00286] In some embodiments, one or more recombinant proteins or antibodies disclosed herein may be used in assays to test, develop and/or select antibodies. Recombinant GPCs may be expressed to test releasing and/or stabilizing abilities of one or more antibodies being assayed. In some embodiments, recombinant proteins may be expressed as positive or negative control components of assays. In some embodiments, multiple recombinant proteins may be expressed at once to modulate growth factor release and/or activity, wherein such recombinant proteins may act synergistically or antagonistically in such modulation.
Antibody development
[00287] In some embodiments, compounds and/or compositions of the present disclosure comprising antibodies, antibody fragments, their variants or derivatives as described above are specifically immunoreactive with antigenic proteins as described herein.
[00288] Antibodies of the present disclosure may be characterized by their target molecule(s), by the antigens used to generate them, by their function (whether as agonists, antagonists, growth-factor releasing, GPC stabilizing, activating and/or inhibitory) and/or by the cell niche in which they function.
[00289] As used herein the term, "antibody fragment" refers to any portion of an intact antibody. In some embodiments, antibody fragments comprise antigen binding regions from intact antibodies. Examples of antibody fragments may include, but are not limited to Fab, Fab', F(ab')2, and Fv fragments; diabodies; linear antibodies; single-chain antibody molecules; and multispecific antibodies formed from antibody fragments. Papain digestion of antibodies produces two identical antigen-binding fragments, called "Fab" fragments, each with a single antigen-binding site. Also produced is a residual "Fc" fragment, whose name reflects its ability to crystallize readily. Pepsin treatment yields an F(ab')2 fragment that has two antigen-binding sites and is still capable of cross-linking antigen. Compounds and/or compositions of the present disclosure may comprise one or more of these fragments. For the purposes herein, an "antibody" may comprise a heavy and light variable domain as well as an Fc region.
[00290] As used herein, the term "native antibody" refers to a usually heterotetrameric glycoprotein of about 150,000 daltons, composed of two identical light (L) chains and two identical heavy (H) chains. Genes encoding antibody heavy and light chains are known and segments making up each have been well characterized and described (Matsuda, F. et al., 1998. The Journal of Experimental Medicine. 188(11); 2151-62 and Li, A. et al., 2004.
Blood. 103(12: 4602-9, the content of each of which are herein incorporated by reference in their entirety). Each light chain is linked to a heavy chain by one covalent disulfide bond, while the number of disulfide linkages varies among the heavy chains of different
immunoglobulin isotypes. Each heavy and light chain also has regularly spaced intrachain disulfide bridges. Each heavy chain has at one end a variable domain (VH) followed by a number of constant domains. Each light chain has a variable domain at one end (VL) and a constant domain at its other end; the constant domain of the light chain is aligned with the first constant domain of the heavy chain, and the light chain variable domain is aligned with the variable domain of the heavy chain.
[00291] As used herein, the term "variable domain" refers to specific antibody domains found on both the antibody heavy and light chains that differ extensively in sequence among antibodies and are used in the binding and specificity of each particular antibody for its particular antigen. Variable domains comprise hypervariable regions. As used herein, the term "hypervariable region" refers to a region within a variable domain comprising amino acid residues responsible for antigen binding. The amino acids present within the
hypervariable regions determine the structure of the complementarity determining regions (CDRs) that become part of the antigen-binding site of the antibody. As used herein, the term "CDR" refers to a region of an antibody comprising a structure that is complimentary to its target antigen or epitope. Other portions of the variable domain, not interacting with the antigen, are referred to as framework (FW) regions. The antigen-binding site (also known as the antigen combining site or paratope) comprises the amino acid residues necessary to interact with a particular antigen. The exact residues making up the antigen-binding site are typically elucidated by co-crystallography with bound antigen, however computational assessments can also be used based on comparisons with other antibodies (Strohl, W.R.
Therapeutic Antibody Engineering. Woodhead Publishing, Philadelphia PA. 2012. Ch. 3, p47-54, the contents of which are herein incorporated by reference in their entirety).
[00292] VH and VL domains have three CDRs each. VL CDRS are referred to herein as CDR-Ll, CDR-L2 and CDR-L3, in order of occurance when moving from N- to C- terminus along the variable domain polypeptide. VH CDRS are referred to herein as CDR-H1, CDR- H2 and CDR-H3, in order of occurance when moving from N- to C- terminus along the variable domain polypeptide. Each of CDRs have favored canonical structures with the exception of the CDR-H3, which comprises amino acid sequences that may be highly variable in sequence and length between antibodies resulting in a variety of three-dimensional structures in antigen-binding domains (Nikoloudis, D. et al., 2014. PeerJ. 2:e456). In some cases, CDR-H3s may be analyzed among a panel of related antibodies to assess antibody diversity. Various methods of determining CDR sequences are known in the art and may be applied to known antibody sequences (Strohl, W.R. Therapeutic Antibody Engineering. Woodhead Publishing, Philadelphia PA. 2012. Ch. 3, p47-54, the contents of which are herein incorporated by reference in their entirety).
[00293] As used herein, the term "Fv" refers to an antibody fragment comprising the minimum fragment on an antibody needed to form a complete antigen-binding site. These regions consist of a dimer of one heavy chain and one light chain variable domain in tight, non-covalent association. Fv fragments can be generated by proteolytic cleavage, but are largely unstable. Recombinant methods are known in the art for generating stable Fv fragments, typically through insertion of a flexible linker between the light chain variable domain and the heavy chain variable domain [to form a single chain Fv (scFv)] or through the introduction of a disulfide bridge between heavy and light chain variable domains (Strohl, W.R. Therapeutic Antibody Engineering. Woodhead Publishing, Philadelphia PA. 2012. Ch. 3, p46-47, the contents of which are herein incorporated by reference in their entirety).
[00294] As used herein, the term "light chain" refers to a component of an antibody from any vertebrate species assigned to one of two clearly distinct types, called kappa and lambda based on amino acid sequences of constant domains. Depending on the amino acid sequence of the constant domain of their heavy chains, antibodies can be assigned to different classes. There are five major classes of intact antibodies: IgA, IgD, IgE, IgG, and IgM, and several of these may be further divided into subclasses (isotypes), e.g., IgGl, IgG2, IgG3, IgG4, IgA, and IgA2.
[00295] As used herein, the term "single chain Fv" or "scFv" refers to a fusion protein of VH and VL antibody domains, wherein these domains are linked together into a single polypeptide chain by a flexible peptide linker. In some embodiments, the Fv polypeptide linker enables the scFv to form the desired structure for antigen binding.
[00296] As used herein, the term "bispecific antibody" refers to an antibody capable of binding two different antigens. Such antibodies typically comprise regions from at least two different antibodies. Bispecific antibodies may include any of those described in RiethmuUer, G. 2012. Cancer Immunity. 12: 12-18, Marvin, J.S. et al., 2005. Acta Pharmacologica Sinica. 26(6):649-58 and Schaefer, W. et al., 2011. PNAS. 108(27): 11187-92, the contents of each of which are herein incorporated by reference in their entirety. [00297] As used herein, the term "diabody" refers to a small antibody fragment with two antigen-binding sites. Diabodies comprise a heavy chain variable domain VH connected to a light chain variable domain VL in the same polypeptide chain. By using a linker that is too short to allow pairing between the two domains on the same chain, the domains are forced to pair with the complementary domains of another chain and create two antigen-binding sites. Diabodies are described more fully in, for example, EP 404,097; WO 93/11161; and
Hollinger et al. (Hollinger, P. et al., "Diabodies": Small bivalent and bispecific antibody fragments. PNAS. 1993. 90:6444-8) the contents of each of which are incorporated herein by reference in their entirety.
[00298] As used herein, the term "monoclonal antibody" refers to an antibody obtained from a population of substantially homogeneous cells (or clones), i.e., the individual antibodies comprising the population are identical and/or bind the same epitope, except for possible variants that may arise during production of the monoclonal antibodies, such variants generally being present in minor amounts. In contrast to polyclonal antibody preparations that typically include different antibodies directed against different determinants (epitopes), each monoclonal antibody is directed against a single determinant on the antigen
[00299] The modifier "monoclonal" indicates the character of the antibody as being obtained from a substantially homogeneous population of antibodies, and is not to be construed as requiring production of the antibody by any particular method. The monoclonal antibodies herein include "chimeric" antibodies (immunoglobulins) in which a portion of the heavy and/or light chain is identical with or homologous to corresponding sequences in antibodies derived from a particular species or belonging to a particular antibody class or subclass, while the remainder of the chain(s) is identical with or homologous to
corresponding sequences in antibodies derived from another species or belonging to another antibody class or subclass, as well as fragments of such antibodies.
[00300] As used herein, the term "humanized antibody" refers to a chimeric antibody comprising a minimal portion from one or more non-human (e.g., murine) antibody source with the remainder derived from one or more human immunoglobulin sources. For the most part, humanized antibodies are human immunoglobulins (recipient antibody) in which residues from the hypervariable region from an antibody of the recipient are replaced by residues from the hypervariable region from an antibody of a non-human species (donor antibody) such as mouse, rat, rabbit or nonhuman primate having the desired specificity, affinity, and/or capacity. [00301] In some embodiments, compounds and/or compositions of the present disclosure may be antibody mimetics. As used herein, the term "antibody mimetic" refers to any molecule which mimics the function or effect of an antibody and which binds specifically and with high affinity to their molecular targets. In some embodiments, antibody mimetics may be monobodies, designed to incorporate the fibronectin type III domain (Fn3) as a protein scaffold (US 6,673,901; US 6,348,584). In some embodiments, antibody mimetics may be those known in the art including, but are not limited to affibody molecules, affilins, affitins, anticalins, avimers, Centyrins, DARPINS™, Fynomers and Kunitz and domain peptides. In other embodiments, antibody mimetics may include one or more non-peptide region.
[00302] As used herein, the term "antibody variant" refers to a biomolecule resembling an antibody in structure and/or function comprising some differences in their amino acid sequence, composition or structure as compared to a native antibody.
[00303] The preparation of antibodies, whether monoclonal or polyclonal, is known in the art. Techniques for the production of antibodies are well known in the art and described, e.g. in Harlow and Lane "Antibodies, A Laboratory Manual", Cold Spring Harbor Laboratory Press, 1988; Harlow and Lane "Using Antibodies: A Laboratory Manual" Cold Spring Harbor Laboratory Press, 1999 and "Therapeutic Antibody Engineering: Current and Future Advances Driving the Strongest Growth Area in the Pharmaceutical Industry" Woodhead Publishing, 2012.
Standard monoclonal antibody generation
[00304] In some embodiments, antibodies are generated in knockout mice, lacking the gene that encodes for desired target antigens. Such mice may not be tolerized to target antigens and therefore may be better suited for generating antibodies against such antigens that may cross react with human and mouse forms of the antigen. For the production of monoclonal antibodies, host mice may be immunized with recombinant proteins to elicit lymphocytes that specifically bind such proteins. Resulting lymphocytes may be collected and fused with immortalized cell lines. Resulting hybridoma cells may be cultured in suitable culture medium with selection agents to support the growth of only fused cells.
[00305] Desired hybridoma cell lines may be identified through binding specificity analysis of secreted antibodies for target peptides and clones of such cells may be subcloned through limiting dilution procedures and grown by standard methods. Antibodies produced by subcloned hybridoma cells may be isolated and purified from culture medium by standard immunoglobulin purification procedures Recombinant antibodies
[00306] Recombinant antibodies of the present disclosure may be generated according to any of the methods disclosed in International Patent Application No. WO2014074532, the contents of which are herein incorporated by reference in their entirety. In some
embodiments, recombinant antibodies may be produced using variable domains obtained from hybridoma cell-derived antibodies produced according to methods described herein. Heavy and light chain variable region cDNA sequences of antibodies may be determined using standard biochemical techniques. Total RNA may be extracted from antibody- producing hybridoma cells and converted to cDNA by reverse transcriptase (RT) polymerase chain reaction (PCR). PCR amplification may be carried out on resulting cDNA to amplify variable region genes. Such amplification may comprise the use of primers specific for amplification of heavy and light chain sequences. In other embodiments, recombinant antibodies may be produced using variable domains obtained from other sources. This includes the use of variable domains selected from one or more antibody fragment library, such as an scFv library used in antigen panning. Resulting PCR products may then be subcloned into plasmids for sequence analysis. Once sequenced, antibody coding sequences may be placed into expression vectors. For humanization, coding sequences for human heavy and light chain constant domains may be used to substitute for homologous murine sequences. The resulting constructs may then be transfected into mammalian cells for large scale translation.
GDF-8-modulatory antibodies
[00307] Some antibodies, presented herein, are GDF-8-modulatory antibodies. Such antibodies may bind GDF-8, a GDF-8 fragment or one or more protein complex comprising GDF-8. In some cases, these antibodies may be releasing antibodies or stabilizing antibodies with regard to GDF-8 growth factor release and/or activity. GDF-8-modulating antibodies of the disclosure may comprise or be developed using any of the scFv sequences listed in Table 13 or fragments thereof. The scFv sequences listed comprise a VH domain joined to a VL domain via a linker comprising the sequence ASAPTLGGGGSGGGGSAAA (SEQ ID NO: 229). Some recombinant GDF-modulating antibodies of the disclosure may be designed to include at least one variable domain pair (VH and VL) present in any of the scFvs listed in Table 13 or variants thereof with at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, at least about 99%, at least about 99.5% or at least about 99.9% sequence identity to any of the scFv sequences listed. Recombinant GDF- 8 -modulatory antibodies may, in some cases, comprise alternative combinantions of the VH and VL domains present in the scFvs presented. Further recombinant GDF-8-modulatory antibodies may, in some cases, comprise VH and/or VL domains presented, but with different combinations of CDRs. Some recombinant GDF-modulating antibodies of the disclosure may comprise or be developed using scFv sequences that comprise at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, at least about 99%, at least about 99.5% or at least about 99.9% sequence identity to any of the scFv sequences listed in Table 13. In some cases, GDF-8-modulatory antibodies constructed using one or more of the scFv sequences presented in Table 13 may interact with one or more of the recombinant proteins listed in Tables 4, 9 or 10.
Table 13. scFv antibody fragments
Clone scFv sequence SEQ ID ID NO
BP6a- QVQLQQSGPGLVKPSQTLSLTCAISGDSVSSNSAAWNWIRQSPSRGLEWL 230 16 GRTYYRSKWSNDYAVAVKSRITINPDTSKNQFSLQLNSVTPEDTAVYYCA
RGFSGMGVWGQGTTVTVSSGSASAPTLGGGGSGGGGSAAASSELTQDPA
VSVALGQTASITCSGDKLGDKYASWYQQKPGQSPVMVIHQDSKRPSGIPE
RFSGSNSGNTATLTISGTQAMDEADYYCQAWDSSTVVFGGGTKLTVLGQP
KAAPSVTLFPPSS
BP6a- QVQLVQSGAEVKKPGSSVKVSCKASGGTFSSYAISWVRQAPGQGLEWMG 231 19 GIIPIFGTANYAQKFQGRVTITADESTSTAYMELSSLRSEDTAVYYCARDQ
AGDYGGNSGYFDYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAADIQ
MTQSPSTLSASAGDRVTITCRASQSISRWLAWYQQKPGKPPKLLIYGASSL
QSGVPSRFSGSGSGTDFTLTISSLQPEDFATYYCQQTYTTPWTFGQGTKVEI
KRTVAAPSVF
BP3a-3 EVQLVQSGGGLVQPGRSLRLSCAASGFTFDDYAMHWVRQAPGKGLEWV 232
SGISWNSGSTYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCAK
GQVPNYYDSRDPFDPWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAAEIV
LTQSPGTLSLSPGERATLSCRASQSVSSYLAWYQQKPGQAPRLLIYGASNR
ATGIPARFSGSGSGTDFTLTISSLQAEDVAVYYCQQYYTTPFTFGPGTKVDI
KRTVAAPSVF
BP6a-l QIQLVQSGAEVKKPGSSVKVSCKASGGTFSSYAISWVRQAPGQGLEWMG 233
GIIPIFGTANYAQKFQGRVTITADESTSTAYMELSSLRSEDTAVYYCARITD
DFDYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAADVVMTQTPLSLPV
TLGQPASISCRSSQSLVYSDGNTYLNWFQQRPGQSPRRLIYKVSNRDSGVP
DRFSGSGSGTDFTLKISRVEAEDVGVYYCMQSTRWPWVFGQGTKVEIKRT
VAAPSVF
BP18b- QVQLVQSGAEVKKPGESLKISCKGSGYSFTSYWIGWVRQMPGKGLEWMG 234 1 IIYPGDSDTRYSPSFQGQVTISADKSISTAYLQWSSLKASDTAMYYCARGV
AGYSSGWSDWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAADIVMTQSP LSLPVTPGEPASISCRSSQTLLHSNGFNYLDWYLQKPGQSPQLLMYLGSKR ASGVPDRFSGSGSGTDFTLKISRVEAEDVGVYYCMQALQTPFTFGPGTKV DIKRTVAAPSVF
BP19b- QVQLQESGPGLVKPSETLSLTCTVSGGSISSYYWSWIRQPPGKGLEWIGYI 235 1 YYSGSTNYNPSLKSRVTISVDTSKNQFSLKLSSVTAADTAVYYCARLRRG
YFDYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAAQSALTQPASVSGSP
GQSITISCTGTSSDVGGYNYVSWYQQHPGKAPKLMIYDVSKRPSGVSNRFS
GSKSGNTASLTISGLQAEDEADYYCSSYTSSSTFVVFGGGTKLTVLGQPKA
APSVTLFPPSS
BP6b-4 QVQLVQSGAEVKKPGESLKISCAASGFTFSSYGMHWVRQAPGKGLEWVA 236
VIWYDGSNKYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCAT
RGYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAAQSALTQPPSASGSPG
QSVTISCSGTSSDIGGYDYVSWYQQHPGKAPKLMIYDVSERPSGVSNRFSG
SKSGNTASLTISGLQAEDEADYYCSSFTSSSTLVFGGGTKLTVLRQPKAAPS
VTLFPPSS
BP6a-8 QVQLQQSGAEVKKPGASVKVSCKASGYTFTSYYMHWVRQAPGQGLEW 237
MGIINPSGGSTSYAQKFQGRVTMTRDTSTSTVYMELSSLRSEDTAVYYCA
REGGDYNNWFDPWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAAQSVLT
QPPSVSGAPGQKVTISCSGSSSNIGNNYVSWYQQRPGTAPKLLIYDNNKRP
SGIPDRFSGSKSGTSATLGITGLQTGDEADYYCGTWDSSLSAVFGGGTKLT
VLRQPKAAPSVTLFPPSS
BP13a- QVQLVQSGSEVKKPGASVKVSCKLSGYTLTELSMHWVRQAPGKGLEWM 238 1 GGFDPEDDEAIYAQKFQGRVTMTEDTSTDTAYMELSSLRSEDTAVYYCAT
DRVTGTTMAAFDIWGQGTMVAVSSGSASAPTLGGGGSGGGGSAAASSEL
TQDPAVSVALGQTVKITCQGDSLRSYFASWYQQKPGQAPVLVLFGKNSRP
SGIPGRFSGSGSGNTAVLTITGAQAEDEADYYCDSRHSSGNYVIFGGGTKV
TVLSQPKAAPSVTLFPPSS
BP5b-3 EVQLVQSGGGLVKPGGSLRLSCAASGFTFSDYYMSWIRQAPGKGLEWVS 239
YISSSGSTIYYADSVKGRFTISRDNAKNSLYLQMNSLRDEDTAVYYCARDN
WYYFDYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAAQSALTQPPSAS
GSPGQSVTISCTGTSSDVGGYNYVSWYQQHPGKAPKLMIYEVSKRPSGVP
DRFSGSKSGNTASLTVSGLQAEDEADYYCSSYAGSNSWVFGGGTKLTVLG
QPKAAPSVTLFPPSS
BP6a- QVQLVESGGGLVKPGGSLRLSCAASGFTFSSYSMNWVRQAPGKGLEWVS 240 23 SISSSSSYIYYADSVKGRFTISRDNAKNSLYLQMNSLRAEDTAVYYCARDL
GGYFGNYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAAQPVLTQPPSV
SLSPGQTASITCSGDDLGDKFASWYQQKPGQSPILVIYQDSKRPSGIPERFS
GSNSGNTATLTISGTQAMDEADYYCQAWDGTKVVFGGGTKLTVLGQPKA
APSVTLFPPSS
BP5a-l QVQLVQSGAEVKKPGSSVKVSCKASGGTFSSYAISWVRQAPGQGLEWMG 241
GIIPIFGTANYAQKFQGRVTITADESTSTAYMELSSLRSEDTAVYYCANHG
LMDDSSGYYLSNAFDIWGQGTMVTVSSGSASAPTLGGGGSGGGGSAAAQ
PVLTQPPSASGTPGQRVTISCSGSSSNIGSNTVEWYQQLPGTAPKLLIHSNN
QRPSGVPDRFSGSRSGTSASLAISGLQSEDEADYFCATWDDSLTGVVFGGG
TTLT VLGQPKA APS VTLFPPS S
BP6a- QIQLVQSGGVVVQPGGSLRLSCAASGFTFDDYTMHWVRQAPGKGLEWVS 242 11 LISWDGGSTYYADSVKGRFTISRDNSKNSLYLQMNSLRTEDTALYYCAAG
SYHTWWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAAQPVLTQPPSASG
TPGQRVTISCSGSSSNIGSSTVNWYQQLPGTAPRLLIYSINQRPSGVPDRFSG
SKSGTSASLAISGLQSDDEADYYCAAWDDSLNGWVFGGGTKLTVLRQPK
ANPTVTLFPPSS
BP5b-4 QVQLQQSGGGLVQPGGSLRLSCAASGFTFSSYAMTWVRQAPGKGLEWVS 243
AISGGGGATYYADSVEGRFTISRDNSKNTLYLQMNSLRAEDTAIYYCASPA GHICSGGSCQYYYYGMDVWGQGTTVTVSSGSASAPTLGGGGSGGGGSAA ADIQMTQSPSFLSASVGDRVTITCRASQGISSYLAWYQQKPGKAPKLLIYA ASSLQSGVPSRFSGSGSGTDFTLTISSLQPEDFATYYCQQSYSTPLTFGGGT RVDMARTVAAPSVF
BP18a- EVQLVQSGAEVKKPGESLKISCKGSGYSFTSYWIGWVRQMPGKGLEWMG 244 1 IIYPGDSDTRYSPSFQGQVTISADKSISTAYLQWSSLKASDTAMYYCARQP
YYYGSGSGMDVWGQGTTVTVSSGSASAPTLGGGGSGGGGSAAAEIVMTQ
SPLSLPVTLGQPASISCGSSQSLVYSDGNTYVNWFQQRPGQSPRRLIYKVS
NRDSGVPDRFSGSGSGTDFTLKISRVEAEDVGVYYCMQGTHWPYTFGQG
TKLEIKRTVAAPSVF
BPl la- EVQLVQSGGGLVQPGRSLRLSCAASGFTFDDYAMHWVRQAPGKGLEWV 245 1 SGISWNSGSIGYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCAK
VDGLEYSSGHNFDYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAAQSA LTQPPSVSGSPGQSVTISCTGSSSDVGYYDHVSWYQHHPGRAPKVIIYDVT KRPSGVPDRFSGSKSGNTASLTISGLQAEDEADYYCSSYAGSYTWVFGGG TELT VLGQPKA APS VTLFPPS S
BP6a- EVQLVESGGGVVQPGRSLRLSCAASGFSFSSYGMHWVRQAPGKGLEWVA 246 14 VTSYDGSNKYYGDSVKGRFTISRDNSKNTLFLQMNSLRAEDTAVYYCAK
DEYTYGSGIMDYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAAEIVLTQ
SPGTLSLSPGEGATLSCRASQTVNSNDLAWYQQRPGQAPRLLIYDASTRAT
GIPDRFSGSGSGTDFTLTISRLEPEDFAVYFCQQYHGSQTFGQGTRVEIKRT
VAAPSVF
BP6a- QMQLVQSGGGVVQPGRSLRLSCAASGFTFSSYAMHWVRQAPGKGLEWV 247 27 ALISYDGSYEYYADSVKGRFTISRDISKNTLYLQMNSLRAEDTAVYYCAR
DSGARGYNYGLFDYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAADIQ
MTQSPSSLSASVGDRVTITCRASQSINRHLNWYQQKPGKAPKFLISAASNL
QSGVPSRFSGSGSGTDFTLTISSLQPEDFATYYCQQSYSTPYTFGGGTKVEI
KRTVAAPSVF
BP6a- EVQLVESGGGVVQPGRSLRLSCAASGFRFSNYGMHWVRQAPGKGLEWVS 248 13 AISGSGGLTYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCARE
DLWRMGELLGGEGFDYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAA
DIQMTQSPSTLSASVGDRVTITCRASQSISSWLAWYQQKPGKAPKLLIYKA
SSLGSGVPSRFSGSGSGTDFTLTISSLQPEDFATYYCQQSYSTPWTFGQGTK
VEIKRTVAAPSVF
BP4a-l EVQLVQSGAEVKKPGASVKVSCKASGYTFSSYGITWVRQAPGQGLEWMG 249
WSSAYNGNTNYAQKLQGRISMTTDTSTSTAYMELRSLRSDDTAVYYCAR
GIYGDYEYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAAQSALTQPAS
VSGSPGQSITISCTGTSSDISIYNYVSWFQQHPGKAPKLMIYDVNNRPSGVS
SRFSGSKSANTASLTISGLQAEDEADYYCSSYTYSNTLLFGGGTKLTVLGQ
PKAAPSVTLFPPSS
BPla-1 EVQLVQSGGGLVQPGRSLRLSCAASGFTFDDYAMHWVRQAPGKGLEWV 250
SGISWNSGSIGYADSVKGRFTISRDNAKNSLYLQMNSLRAEDTALYYCAK
DTSNGGYSSSSFDYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAAQSAL
TQPASVSGSPGQSITISCTGTSSDVGGYNYVSWYQQHPGTAPKLMIYDVSY
RPSGVSNRFSGSKSGNTASLTISGLQAEDEADYYCSSYTSSSTLVFGTGTKV
TVLGQPKANPTVTLFPPSS
BP6a-6 QVQLVQSGAEVKKPGSSVKVSCKASGGTFSSYAISWVRQAPGQGLEWMG 251
GIIPIFGTANYAQKFQGRVTITADESTSTAYMELSSLRSEDTAVYYCARDR
HLYRRGYSSVDGMDVWGQGTTVTVSSGSASAPTLGGGGSGGGGSAAAEI
VMTQSPGTLSLSPGERATLSCRASQSVSSGYLAWYQQKAGQAPRLLIYGG
SARAPGIPARFSGSGSGTDFTLTISSLEPEDFAVYYCQQRSIWPLTFGGGTK
VEIKRTVAAPSVF
BP19b- QLQLQQSGGGVVQPGRSLRLSCAASGFTFSSYGMHWVRQAPGKGLEWV 252
2 AVISYDGSNKYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCA
KPRGRYYFDYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAAQSALTQP PSASGSRGQSVSISCSGSRSDIGYYNYVSWYQQHPGKAPKLIIFDVNKRPSG VPDRFSGSKSGNTASLTVSGLQAEDVADYYCSSSVGSNNLVFGGGTKLTV LGQPKAAPSVTLFPPSS
BP6a-4 QVQLQESGPGLVKPSETLSLTCTVSGDSITSYYWSWIRQPPGKGLEWIGYI 253
HYSGSTYYNPSLKSRVTISVDRSKNQFSLKLSSVTAADTAVYYCARGPSRG
LFDPWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAAQSALTQPASVSGSP
GQSITLSCTGTSSDVGDYNYVSWYQQYPGKAPKLMIYDVTKRPSGVSNRF
SGSKSGNTASLTISGLQAEDEADYYCSSYTSSTTWVFGGGTKLTVLGQPK
AAPSVTLFPPSS
BP19a- QVQLQESGGVVQPGGSLRLSCAASGFTFSSYGMSWVRQAPGKGLEWVSSI 254 1 SNSGGSTYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCASLSR
GYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAAQSVLTQPPSASGTPGQ
RVSISCSGRMSNIGKNTVNWYQQVPGTAPKLLIYSNNQRPSGVPDRFSGSK
SGTSASLAISGLQSEDEADYYCAAWDDSLNGWVFGGGTKLTVLGQPKAA
PSVTLFPPSS
BP6a-7 QVQLQQSGAEVKKPGASVKVSCKASGYTFTSYYMHWVRQAPGQGLEW 255
MGIINPSGGSTSYAQKFQGRVTMTRDTSTSTVYMELSSLRSEDTAVYYCA
RESLIRFLEDPQQGGMDVWGQGTTVTVSSGSASAPTLGGGGSGGGGSAA
AQSALTQPASVSGSPGQSLTISCTGTSSDIGGYNYVSWYQQHPGKAPKLIIY
DVTDRPSGVSGRFSGSKSGNTASLTISGLQTEDEAEYFCNSWTRSNNYIFG
GGTKLT VLGQPKA APS VTLFPPS S
BP5b-5 QVQLVQSGAEVKKPGESLKISCKGSGYSFTSYWIGWVRQMPGKGLEWMG 256
IIYPGDSDTRYSPSFQGQVTISADKSISTAYLQWSSLKASDTAMYYCATPYS
SSSGTDYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAADVVMTQSPLS
LPVTLGQPASISCRSSQSLVYSDGNTYLNWFQQRPGQSPRRLIYKVSNRDS
GVPDRFSGSGSGTDFTLKISRVEAEDVGVYYCMQGTHWPLTFGQGTKVEI
TRTVVAPSVF
BP6b-3 QIQLVQSGAEVKKPGSSVKVSCKASGGTFSSYAISWVRQAPGQGLEWMG 257
WINPNSGGTNYAQKFQGWVTMTRDTSISTAYMELSRLRSDDTAVYYCAR
PRTLRGDAFDIWGRGTMVTVSSGSASAPTLGGGGSGGGGSAAASSELTQA
PSVSVSPGQTASITCSGNKLGDKFASWYQQKPGQSPVMVMYEDRKRPSGI
PERFS GSNS GNT ATLTIS GTQ ALDE AD Y YCQT WDSRT V VFGGGTKLT VLG
QPKAAPSVTLFPPSS
BP18b- QVQLVQSGAEVKKPGESLKISCKGSGYSFTSYWIGWVRQMPGKGLEWMG 258 2 IIYPGDSDTRYSPSFQGQVTISADKSISTAYLQWSSLKASDTAMYYCARHPI
GSGYDYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAADIVMTQSPVSL
PVTLGQPASISCRSSQSVVYRDGNTYLSWFLQRPGQSPRRLIYKVSNRDSG
VPDRFSGSGSATDFTLKISRVEAEDVGVYYCMQGSSWPYTFGQGTKLEIK
RTVAAPSVF
BPx-1 QVQLVQSGAEVKKPGSSVKVSCKASGGAFSIYSINWVRQAPGQGLEWMG 259
GIIPIFETANSAQKFQGRVTITADKSTSTAYMELSSLRSEDTAVYYCARAGE
NSDSNYAFDIWGRGTVVTVSSGSASAPTLGGGGSGGGGSAAADIVMTQSP
LSLPVTPGEPASISCRSSQSLLHSDGNNYLDWYLQKPGQSPQLLIYLGSNRA
SGVPDRFSGSGSGTDFTLKISRVEAEDVGVYYCMQGLHTPTTFGQGTKVEI
KRTVAAPSVF
BP21b- QLQLQQSGPGLVKPSETLSLTCTVSGGSISSSSYYWGWIRQPPGKGLEWIG 260 1 SIYYSGSTYYNPSLKSRVTISVDTSKNQFSLKLSSVTAADTAVYYCARGRY
SYGYFDYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAANFMLTQPHSV
SESPGKTVTISCTRSSGSIASNYVQWYQQRPGSSPTTVIYEDNQRPSGVPDR
FSGSIDSSSNSASLTISGLRTEDEADYYCQSYDHMYNWVIGGGTKLTVLGQ
PKAAPSVTLFPPSS
BP6a-2 QVQLVQSGGGLVKPGGSLRLSCAASGFTFSSYSMNWVRQAPGKGLEWVS 261
SISSSSSYIYYADSVKGRFTISRDNAKNSLYLQMNSLRAEDTAVYYCARGI RVVGDEDYYYYYGMDVWGQGTTVTVSSGSASAPTLGGGGSGGGGSAAA QSVLTQPPSVSGAPGQKVTISCSGSSSNIGNNYVSWYQQLPGTAPKLLIYD NNQRPSGVPDRFSGSKSGTSASLAISGLRSEDEADYHCAAWDDDLNGPVF GGGTKLT VLGQPK A APS VTLFPPS S
BPx-2 EVQLLQSGGGLVQPGGSLRLSCAASGFTFSSYSMNWVRQAPGKGLEWVS 262 YISSSSSTIYYADSVKGRFTISRDNAKNSLYLQMNSLRDEDTAVYYCARDS SGWLGMGDFDYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAADIQMT QSPSSLSASVGDRVTISCRASHSISRFLNWYQQKPGRAPKLLIYSTSSLQSG VPSRFSGSGSARDFTLTISGLQPEDFATYFCQQSYSTPWTFGQGTKVELKR TVAAPSVF
BP6b-l QVQLVESGGGLVKPGGSLRLSCAASGFTFSSYSMNWVRQAPGKGLEWVS 263
SISSSSSYIYYADSVKGRFTISRDNAKNSLYLQMNSLRAEDTAVYYCAREE
YYGGNTDWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAASSELTQPPSVS
VSPGQTATITCSGDKLGDKYASWYQQKPGQSPVLVIYQDTKRPSGIPARFS
GSNSGNTATLTISGTQAMDEAAYYCQAWDSTTVVFGGGTKLTVLGQPKA
APSVTLFPPSS
BPx-3 EVQLVQSGGGLVQPGGSLRLSCVTSGFSFSNYGMSWVRQAPGKGLEWVS 264
EISGSGGSAYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCARD
ISHYDILTGQLDYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAADIQMT
QSPSSLSASVGDRVTITCRASQSISSYLNWYQQKPGKAPKLLIYAASSLQSG
VPSRFSGSGYETDFTLTISSLQAEDVAVYYCQQYYSSPWTFGQGTRVEIKR
TVAAPSVF
BP6a-5 QIQLVQSGGGVVQPGRSLRLSCAASGFTFSSYGMHWVRQAPGKGLEWVA 265
VISYDGSNKYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCARD
LLVRFLEWSHYYGMDVWGQGTTVTVSSGSASAPTLGGGGSGGGGSAAA
QPVLTQPPSASGTPGQRVTISCSGSSSNIGSNTVHWYQQLPGTAPKLLIYSD
NQRPSGVPDRFSGSKSGTSASLVISGLQSDDEADYYCAAWDDSLNGVFGG
GTKLTVLgQPKAAPSVTLFSPSS
BP6a- EVQLVESGGGLVQPGGSLRLSCAASGFTFSSYWMSWVRQAPGKGLEWVA 266 10 NIKQDGSEKYYVDSVKGRFTISRDNSENTLYLQMNSLRAEDTAVYYCAKE
TDCSGGICYGVDYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAAQPVL
TQPPSVSVSPGQTASITCSGDKLGDKYASWYQQKPGQSPVMVIYQDRKRP
SGIPERFSGSNSGNTATLTISGTQAMDEADYYCAAWDDSLSGRVFGGGTK
LTVLGQPKA APS VTLFPPS S
BP15a- QVQLVQSGAEVKKPGASVKVSCKASGYTFTSYGISWVRQAPGQGLEWM 267 1 GWISAYNGNTNYAQKLQGRVTMTTDTSTSTAYMELRSLRSDDTAVYYCA
RDAPYDILTGYLSYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAAEIVM
TQSPGTLSLSPGERATLSCRASQSVSSSYLAWYQQKPGQAPRLLIYGASSR
ATGIPDRFSGSGSGTDFTLTISRLEPEDFAVYYCQQYGSSPYTFGQGTKLEI
KRTVAAPSVF
BP6a- QIQLVQSGGGVVQPGRSLRLSCAASGFSFSTYAMHWLRQAPGKGPEWVA 268 26 AISYDGTIKYYADSVKGRFTISRDSSKNTLYLQMNNLGPEDTALYYCARA
GGAGSDGYNYWGYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAAEIV
LTQSPATLSLSPGDRATLSCRASQSVSSYLAWYQQKPGQAPRLLIYDASNR
ATGIPARFSGSGSGTDFSLTISRLEPEDFAVYYCQQSGSSPWTFGQGTKVEI
KRTVAAPSVF
BP6a- QVQLVQSGGGLVKPGGSLRLSCAASGFTFSSYSMNWVRQAPGKGLEWVS 269 12 SISSSSSYIYYADSVKGRFTISRDNAKNSLYLQMNSLRAEDTAVYYCARDP
GGKLGGAFDIWGQGTMVTVSSGSASAPTLGGGGSGGGGSAAADIVMTQS
PATLSLSPGERATLSCRASQSVSSYLAWYQQKPGQAPRLFIYGASTRATGIP
DRFSGSGSGTDFTLTISSLQAEDVAVYYCQQYYSTPYTFGQGTKLEIKRTV
AAPSVF
BP5a-4 QVQLVQSGAEVKKPGSSVKVSCKASGGTLSSYAISWVRQAPGQGLEWMG 270
GIIPIFGTANYAQKFQGRVTITADESTSTAYMELSSLRSEDTAVYYCARDRP GNNRYDILTGGGWFDPWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAAQ SVLTQPPSVSAAPGQKVTISCSGGSSNIGNNYLSWYQQLPGTAPKLLIYDN NKRPSGIPDRFSGSKSGTSATLGITGLQTGDEADYYCGTWDSSLSAGVFGG GTKLT VLGQPKA APS VTLFPPS S
BP5b-l QVQLVQSGGGVVRPGGSLRLSCAASGFTFDDYGMSWVRQAPGKGLEWV 271
SGISWNSGSIGYADSVKGRFTISRDNAKNSLYLQMNSLRAEDTALYYCAK
EISDTAMGFDYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAAQSALTQP
ASVSGSPGQSITISCTGTSSDVGGYNYISWYQQYPGKAPKLMIYDVSKRPS
GVSNRFSGSKSGNTASLTISGLQAEDEADYYCNSYTSSRTVVFGGGTKLTV
LGQPKAAPSVTLFPPSS
BPx-4 EVQLVESRGGVVQPGRSLRLSCAASGFTFSSYGMHWVRQAPGKGLEWVA 272
VISYDGSNKYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCARA
YIPRLYYYGSGLDYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAASSEL
TQPPSVSVSPGQTASITCSGDQLGDKFVSWYQQKPGRSPVLVISEDNKRHS
GTPERISGSNSGNTATLTISGTQSMDEADYFCQTWDSSAVVFGGGTKLTVL
GQPKAAPSVTLFPPSS
BP5a-3 QVQLQESGPGLVKPSETLSLTCTVSGYSITNGYYWGWIRQSPGKGLEWIG 273
NIFQSGSTYYNPSFKSRVTISVDTSKNQFSLKLSSVTAADTAVYYCARRVD YYDSSGYGEGYFDYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAASSE LTQDPAVSVALGQTVRITCQGDSLKKYYASWYQQKPGQAPVLVIYGKNL RPSGIPERFSGSSSGDTASLTITGTQAEDEADYYCQAWDSSTAVFGGGTKL T VLGQPKA APS VTLFPPS S
BP14a- QVQLVQFGAEVKKPGESLKISCKGSGYNFNSYWIGWVRQMPGKGLEWM 274 1 GIIYPGDSDIRYNPSFQGQVTISADKSISTAYLQWSSLKASDSAMYYCARH
VSGSLDYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAADVVMTQSPLS
LSVTPGQPASISCRSSQSLVHSDGNTYLNWFHQRPGQSPRRLIYKVSNRDP
GVPDRFSGSGSGTDFTLKISRVEAEDAGVYYCMQGTHWPRLTFGGGTKVE
IKRTVAAPSVF
BP3a-l EVQLVQSGGGLVQPGGSLRLSCAASGFTFSSYAMSWVRQAPGKGLEWVS 275
AISGSGGSTYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCAKT
AYYDSSGYYPDAFDIWGQGTMVTVSSGSASAPTLGGGGSGGGGSAAADI
QMTQSPSSFSASTGDRVTITCRASQGISSYLAWYQQKPGKAPKLLIYAAST
LQSGVPSRFSGSGSGTDFTLTISSLRLDDVATYYCQQYQSYPITFGQGTRLE
IKRTVAAPSVF
BP18b- QVQLVQSGAEVKKPGESLKISCKGSGYSFTSYWIGWVRQMPGKGLEWMG 276 3 IIYPGDSDTRYSPSFQGQVTISADKSISTAYLQWSSLKASDTAMYYCARGQ
GGVLGAFDIWGQGTMVTVSSGSASAPTLGGGGSGGGGSAAAEIVLTQSPD
SLAVSLGERATINCKSSQNLLYSSNNKNYLAWYQQKPGQPPKLLINWAST
RVSGVPDRFSGSGSGTDFTLTINSLQAEDVAVYYCHQYYSSPYTFGPGTKV
EIKRTVAAPSVF
BP6a-9 EVQLVESGAEVKKPGEPLKISCKGSGYSFTSYWIGWVRQMPGKGLEWMG 277
IIYPGDSDTRYSPSFQGQVTISADKSISTAYLQWSSLKASDTAMYYCARQG
RGEAFDIWGQGTMVTVSSGSASAPTLGGGGSGGGGSAAAQPVLTQPPSVS
VSPGQTASITCSGDNLGDKYASWYQQKPGQSPVLVIFRDTKRPSGIPERFS
GSNSGDTATLTISGTQAMDEADYYCQAWDSSAGGVFGGGTRLTVLGQPK
AAPSVTLFPPSS
BP6a- QVQLQQSGPGLVKPSQTLSLTCAISGDSVSSNTEAWNWIRQSPSRGLEWL 278 25 GRTYYRSKWYHDYAPSVKSRITINPDTSKNQFSLQLNSVTPEDTAVYYCA
RTVFDSQVYWFDPWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAADVV
MTQSPLSLPVTPGEPASISCRSSQTLLHSNGFNYLDWYLQKPGQSPQLLMY
LGSSRASGVPDRFSGSGSGTDFTLKISRVEAEDVGVYYCMQGTHWPPTFG
QGTKLEIRRTVAAPSVF
BPx-5 EVQLVESRGGLVKPGGSLRLSCAASGFTFSSYGMSWVRQAPGKGLEWVS 279 SISNSGGSTYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCASLS RGYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAAQAVLTQPPSVSGTP GQTVIISCSGSNSNLGRNSANWYQQLPGTAPKLLIHSNHMRPSGVPDRFSG SKSGTSASLAISGLQSEDEADYYCAAWDDSLNGWVFGGGTKLTVLGQPK AAPSVTLFPPSS
BP5a-2 QVQLVQSGGGLIQPGGSLRLSCAASGFTVSSYSMNWVRQAPGKGLEWVS 280
YISSSGSTIYYADSVKGRFTISRDNAKNSLYLQMNSLRAEDTALYYCAKVG
TAAAGDAFDIWGQGTMVTVSSGSASAPTLGGGGSGGGGSAAAQPVLTQP
PSASGTPGQRVTISCFGSSSNIGSNYVYWYQQLPGTAPKLLIYRNNQRPSG
VPDRFSGSKSGTSASLAISGLRSEDEADYYCAAWDDSLSGWVFGGGTKLT
VLGQPKAAPSVTLFPPSS
BP6a- QVQLQESGPGLVKSSGTLSLTCAISGVSIASSNWWSWVRQPPGKGLEWVG 281 15 EIYHSGSTFYNPSLQSRVTISVDKSKNQFSLTLNSVTAADTAVYYCASSSG
WPYPTNWFDAWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAADIQMTQS
PSTLSASVGDRVSITCRASQSVSSWLAWYQQKPGKAPKLLIYAASSLQSGV
PSRFSGSGSGTDFTLTISNLQPEDSATYFCQQSYSTPWTFGQGTKVELKRTV
AAPSVF
BP6a- EVQLLESRGSLVQPGGSLRLSCAASGFTFSSYAMSWVRQAPGKGLEWVSG 282 21 ISDSGGTTYYADSVKGQFTISRDNSKNTLYLQMNSLRAEDTAVYYCAKDA
HNWNYGVFDYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAADIQMTQ
SPSSLSASVGDRVTITCQASHDISNYLNWYQQKPGKAPKLLIYDASNLETG
VPSTFSGSGSGTDFTLTISSLQPEDIATYYCQQSYSTPYTFGQGTKVEIKRTV
AAPSVF
BP5a-5 QVQLVQSGGGLVQPGGSLRLSCAASGFTFSSYSMNWVRRAPGKGLEWVS 283
YISSSSSTIYYADSVKGRFTISRDNAKNSLYLQMNSLRDEDTAVYYCASAL
GATGAFDIWGQGTMVTVSSGSASAPTLGGGGSGGGGSAAAQSALTQPAS
VSGSPGQSITISCTGTSSDVGGYNYVSWYQQHPGKAPKLMIYDVSKRPSG
VSNRFSGSKSGNTASLTISGLQAEDEADYYCSSYTTSSTLVFGGGTKVTVL
GQPKAAPSVTLFPPSS
BP6a- QMQLVQSGGGLVKPGGSLRLSCAASGFTFSDYYMTWIRQAPGKGLEWVS 284 17 AISGSGGSTYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCAKD
ISGYEPLYYFDYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAASSELTQ
DPSVSVSPGQTASITCSGDKLGDKYVGWYQQKPGQSPVLIIYRDYKRPSGT
PERFS GSNS GNT ATLTIS GVE AGDE AD Y YCQ V WDMS SDPL VFGGGTKLT V
LGQPKAAPSVTLFPPSS
BP6a- EVQLVESRGGVVQPGRSLRLSCAASGFTFSSYAMHWVRQAPGKGLEWVA 285 20 VISYDGSNKYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCARD
PGKQWRVRGYFDYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAADIV
MTQSPSTLSVSVGDRVTITCRASQSISSWLAWYQQKPGKAPKLLIYKASSL
ESGVPSRFSGSGSGTDFTLTISSLQPEDFATYYCQQSYSTPYTFGQGTKLEIK
RTV AAPSVF
BP3a-2 EVQLVQSGGDLEQPGGSLRLSCGTSGFTFSDYDMSWVRQAPGKGLEWVS 286
GISWNSGSIGYADSVKGRFTISRDNAKNSLYLQMNSLRAEDTAVYYCARE
SDMAAAGSFDYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAAQSALTQ
PRSVSGSPGQSITISCTGTSSDVGGYNYVSWYQQHPGKAPKLMIYDVSKRP
SGVSNRFSGSKSGNTASLTISGLQAEDEADYYCSSYTSSSTYVFGTGTKVT
VLGQPKADPTVTLFPPSS
BP6a- EVQLVESRGGLVKPGGSLRLSCAASGFTFSSYSMNWVRQAPGKGLEWVS 287 24 SISSSSSYIYYADSVKGRFTISRDNAKNSLYLQMNSLRAEDTAVYYCAREG
NSGYDVWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAAQPVLTQPPSVS
VSPGQTASITCSGDDLGDKFASWYQQKPGQSPILVIYQDSKRPSGIPERFSG
SNSGNTATLTISGTQAMDEADYYCQAWDGTKVVFGGGTKVTVLGQPKA
APSVTLFPPSSKARG
BP6a- QVQLVQSGAEVKKPGASVKVSCKASGYTFTSYGISWVRQAPGQGLEWM 288 22 GWISAYNGNTNYAQKLQGRVTMTTDTSTSTAYMELRSLRSDDTAVYYCA
RDSLWFGELSALDYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAADIV MTQSPSSVSASVGDRVTITCRASQDISSWLAWYQQKPGKAPKLLIYAASSL QSGVPSRFSGSGSGTDFTLTISSLQPEDFASYYCQQANSFPQTFGGGTKVEI KRTVAAPSVF
BP19a- QVQLVQSGGGVVLPGRSLRLSCAASGFTFSSYAMHWVRQAPGKGLEWV 289 2 AVISYDGSNKYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCAR
DVSSWAAFWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAANFMLTQPHS
VSESPGKTVTISCTRSSGRIVSTYVQWYQHRPGSPPRLIMYDDVQRPSGVS
ARFSGSIDISSNSASLTISGLLAEDEADYYCQSYNTSNLVFGGGTKLTVLSQ
PKAAPSVTLFPPSS
BPx-6 QIQLVQSGAEVKKPGASVKVSCKASGYTFTSYGISWVRQAPGQGLEWMG 290
WISAYNGNTNYAQKLQGRVTMTTDTSTSTAYMELRSLRSDDTAVYYCAR
GGRGGVITWFDPWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAAQSALT
QPPSASASPGQSVTISCTGTSSDVGGYDFVSWYQQHPGQAPKLMIYEVNK
RPSGVPDRFSGSKSGNTASLTVSGLQADDEADYYCCAYAGSNILLFGGGT
KLTVLGQPKAAPSVTLFPPSSKPRG
BP6a- QIQL VQS GGGLIQPGGSLRLSC A AS GFTFS SF AMS W VRQ APGKGLEW VS SI 291 18 TSRGDTTYYADSVRGRFTISRDNSKNTLYLQMNSLRADDTAVYYCARDRP
QVPYDSSGNFDYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAAAIQLT
QSPSSLSASVGDRVTITCRASQSISTYLNWYQQKPGKAPKLLIYDTSSLHSG
VPSRFSGSGSGTDFTLTISSLQPEDFATYHCQQSFITPYTFGQGTKLEIKRTV
AAPSVF
BP16a- QVQLVQSGAEVKKPGSSVKVSCKASGGTFSSYAISWVRQAPGQGLEWMG 292 1 GIIPIFGTADYAQKFQGRVTITADESTSTAYMELSSLRSEDTAVYYCAREEA
GYSSGWVEDAFDIWGQGTMVTVSSRSASAPTLGGGGSGGGGSAAAQSAL TQPASVSGSPGQSITISCTGTSSDVGGYNYVSWYQQHPGKAPKLMISDVSK RPSGVSNRFSGSKSGNTASLTISGLQAEDEADYYCSSYTSNYTLVFGGGTK LT VLGQPKA APS VTLFPPS S
BP19a- QVQLVQSGGGVVQPGRSLRLSCAASGFTFSSYAMHWVRQAPGKGLEWV 293 3 AVISYDGSNKYYADSVKGRFTISRDNAENSLYLQMNSLRAEDTAVYYCAR
MVGGYRFDYWGQGTTVTVSSGSASAPTLGGGGSGGGGSAAADIVMTQSP
ATLSVSPGERATLSCRASQSVSSYLAWYQQKPGQAPRLLIYDASNRATGIP
ARFSGSGSGTDFTLTISSLEPEDFAVYYCQHRGSWPHTFGPGTKLEMKRTV
AAPSVF
BP5b-2 EVQPLETGGGLVQPGGSLRLSCVTSGFSFSNYGMSWVRQAPGKGLEWVS 294
EISGSGGSAYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCAKH
GDYGGTFDYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAAEIVMTQSP
ATLSLSPGDRATLSCRASQNVGSYLGWYQQKVGQAPRLLIYDASNRATG
VPGRFSGSGSGTDFTLTISSLEPEDFAVYYCQQYGSSPLFTFGPGTKVDIKR
TV AAPSVF
BP2a-l QIQLVQSGAEVKKPGASVKVSCKASGYTFTSYGISWVRQAPGQGLEWMG 295
WISAYNGNTNYAQKLQGRVTMTTDTSTSTAYMELSSLRSEDTAVYYCAR
VGFYDYVWGSYPYDAFDIWGQGTMVTVSSGSASAPTLGGGGSGGGGSA
AAEIVMTQSPGTLSLSPGERATLSCRASQSVSSNYLAWYQQKPGQAPRLLI
YDASNRATGIPARFSGSGSGTDFTLTISSLEPEDFALYYCQQYGTSPLTFGG
GTKLEIKRTV AAPSVF
BP18a- QVQLVQSGAEVKKPGESLKISCKGSGYSFTNYWIAWVRQMPGKGLEWM 296 2 GIIYPGDSDTRYSPSFQGQVTISADKSISTAYLQWTSLKASDTAMYYCARR
SSSSGSFDYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAADIVMTQSPL
SLPVTPGEPASISCRSSQSLLHSNGYNYLDWYLQKPGQSPQLLIYLGSNRAS
GVPDRFSGSESGTDFTLKISRVEAEDVGVYYCMQATHWPPRLTFGPGTKV
DIKRTV AAPSVF
BP6a-3 QLQLQQSGGGLVKPGGSLRLSCAASGFTFSSYSMNWVRQAPGKGLEWVS 297
SISSSSSYIYYADSVKGRFTISRDNAKNSLYLQMNSLRAEDTAVYYCVRDR HSLGDFDYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAASSELTQPPSV SVSPGQTATITCSGDKLGDKYASWYQQKPGQSPVLVIYQDTKRPSGIPARF SGSNSGNTATLTISGTQAMDEAAYYCQAWDSTTVVFGGGTKLTVLGQPK AAPSVTLFPPSS
BPx-7 QIQLVQSGGGVVQPGRSLRLSCAASGFTFSSYGMHWVRQAPGKGLEWVA 298
VISYDGSNKYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCARR
FTMDYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAAQSALTQPASVSG
SPGQSITISCTGTSSDVGGYNYVSWYQQRPGKAPKLMIYDVSNRPSGVSNR
FSGSKSGNTASLTISGLQAEDEADYYCSSYTTSSTVVFGGGTKLTVLGQPK
AAPSVTLFPPSS
BPx-8 QVQLQESGPGLVKPSETLSLTCTVSGGSISSSSYYWGWIRQPPGKGLEWIG 299
SIYYSGSTYYNPSLKSRVTISVDTSKNQFSLKLSSVTAADTAVYYCARYSSS
WELFDYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAARSALTQPASVS
GSPGQSITISCTGSSNDVGGYKFVSWYQHHPGKAPKLLIYGVTQRPSGVSN
RFSGSQSGNTASLTISGLQAEDEADYFCTSYTRSTTPIFGGGTKLTVLGQPK
AAPSVTLFPPSS
BPx-9 QIQLVQSGAEVRKPGDSVKVSCRTSGYTFNDYHIHWVRQAPGQGLEWMG 300
WISAYNGNTNYAQKLQGRVTMTTDTSTSTAYMELRSLKSDDTAVYYCAR
RNGSGSAPGYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAAQSALTQP
PSASGSPGQSVTVSCTGSSSDIGSYNYVSWFQRHPGQAPKLIIHDVTERPSG
VPHRFSGSKSGNTASLTISGLQTEDEADYFCCSYSGDHTLPFIFAPATQVTV
LGQPK ANPT VTLFPPS S
BPx-10 EVQLVESGAEVKKPGESLRISCKGSGYSFTSYWIGWVRQMPGKGLEWMGI 301
IYPGDSDTRYSPSFQGQVTISADKSISTAYLQWSSLKASDTAMYYCARVGQ
YYGMDVWGQGTTVTVSSGSASAPTLGGGGSGGGGSAAADIVMTQSPLSL
PVTLGQPASISCRSSQGLVYSDGNTYLNWFQQRPGQSPRRLIYKVSNRDSG
VPDRFSGSGSGTDFTLKISRVEAEDVGVYYCMQATHWPWTFGQGTKVEIK
BPx-11 QVQLQESGPGLVKPSETLSLACTVSGGSISSSSYYWGWIRQPPGKGLEWIA 302
SINYSGNTYYNPSLKSRVIIPIDTSKNQFSLKLSSVTAADTAVYYCARRVLG
YGYFDYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAAQSVLTQPPSAS
GTPGQRVTISCSGSSSNIGSNYVYWYQQLPGTAPKLLIYRNNQRPSGVPDR
FSGSKSGTSASLAISGLRSEDEADYYCAAWDDSLSGPVFGGGTKLTVLGQP
KAAPSVTLFPPSS
BPx-12 QVQLVQSGAEVKKPGSSVKVSCKASGGTFSSYAISWVRQAPGQGLEWMG 303
GIIPIFGTANYAQKFQGRVTITADESTSTAYMELSSLRSEDTAVYYCTGGG
NVDYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAADVVMTQSPLSLPV
TLGQPASISCRSSQSLVHGNGITYLNWFQQRPGQSPRRLIYKVSNRDSGVP
DRFSGSGSGTDFTLKISRVEAEDVGVYYCMQALQTPSFGQGTRLEIKRTVA
APSVF
BPx-13 EVQLLESGGGLVQPGRSLRLSCAASGFTFDDYAMHWVRQAPGKGLEWVS 304
GISWNSGSIGYADSVKGRFTISRDNAKNSLYLQMNSLRAEDTALYYCAKD
KAGNYYYGMDVWGQGTTVTVSSGSASAPTLGGGGSGGGGSAAAQSALT
QPPSASGSPGESVTISCTGTSSDVGGYNYVSWYQQHPGKAPKLLIYDVTKR
PSGVSNRFSGSKSGNTASLTISGLQAEDEADYYCSSYTSSSTVVFGGGTKL
TVPGQPKAAPSVTLFPPSS
BPx-14 QVQLVQSGGGVVQPGRSLRLSCAASGFTFGHYGMHWVRQAPGKGLEWV 305
AVISYDGTNKYYADSVKGRFTISRDNSKNTLYLQMNSLRVDDTAVYYCA
KGSRLDPWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAAQSALTQPRSVS
GSPGQSVTISCTGTSSDVGGYNYVSWYQHHPGKAPKLMIYDVNKRPSGVP
DRFSGSKSANTASLTVSGLQAEDEADYYCSSYTAGGNWVFGGGTKLTVL
GQPKAAPSVTLFPPSS
BPx-15 QVQLQQSGPGLVKPSQTLSLTCAISGDSVSSNSAAWNWIRQSPSRGLEWL 306
GRTYYRSKWYNDYAVSVKSRITINPDTSKNQFSLQLNSVTPEDTAVYYCV
GQGYDSSGYYGMDVWGQGTTVTVSSGSASAPTLGGGGSGGGGSAAADV
VMTQSPLSLPVTPGEPASISCRSSQSLLHSNGYNYLDWYVQKPGQSPQHLI
YLGSIRASGVPDRFSGSGSGTDFTLKISRVEAEDVGVYYCMQGLQTPPTFG PGTKMDIKRTVAAPSVF
BPx-16 QVQLVQSGAEVKKPGESLKISCKGSGYSFTSYWIGWVRQMPGKGLEWMG 307
IIYPGDSDTRYSPSFQGQVTISADKSISTAYLQWSSLKASDTAMYYCARQY
YDTSYFDYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAADVVMTQSPL
SLPVTLGQPASISCRSSQSLVYSDGNTYLSWFHQRPGQSPRRLIYKVSNRDS
GVPDRFSGSGSGTDFTLKISRVEAEDVGVYYCMQGTHWPWTFGQGTKVEI
KRTVAAPSVF
BPx-17 EVQLVQSGGGLVQPGRSLRLSCAASGFTFGDYAMHWVRQAPGKGLEWV 308
SGINWNSGGVGYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCA
KDSGYSYGINFDYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAAQSAL
TQPASVSGSPGQSITISCTGTSSDVGGYNYVSWYQQHPGKAPKLMIYDVSK
RPSGVSNRFSGSKSGNTASLTISGLQAEDEADYYCSSYTSSSTWVFGGGTK
BPlb-1 QIQLVQSGAEVKKPGASVKVSCKASGYTFTSYGISWVRQAPGQGLEWMG 309
WISAYNGNTNYAQKLQGRVTMTTDTSTSTAYMELRSLRSDDTAVYYCAR
GDWFDPWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAAQSALTQPPSAS
GSPGQSVTISCSGTSSDIGGYDYVSWYQQHPGKAPKLIIYEVSKRPSGVPDR
FSGSKSGNTASLTVSGLQADDGAHYYCSAYAGRNNMVFGGGTKVTVVG
QPKAAPSVTLFPPSS
BPx-18 QVTLKESGPTLVKPTQTLTLTCTFSGFSLSTSGVGVGWIRQSPGKALEWLA 310
LIYWDDDKRYSPSLKSRLTITKDTSKNQVVLTMTNMDPVDTATYYCAHM
RWGYYFDYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAAQSVLTQPPS
ASGTPGQRVIISCSGSRSNNGSNYIYWYQHLPGTAPKLLIYRNNQRPSGVP
DRFSGSKSGTSASLAISGLQSEDEADYYCAAWDRSLNAWVFGGGTKLTVL
SQPKAAPSVTLFPPSS
BPx-19 EVQLVQSGAEVKKPGESLKISCKGSGYSFTSYWIGWVRQMPGKGLEWMG 311
IIYPGDSDTRYSPSFQGQVTISADKSISTAYLQWSSLKASDTAMYYCARRE
MGAKGSGGIDYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAADIVMTQ
TPLSLPVTLGQPASISCRSSQSLVYSDGKTYLNWFQQRPGQSPRRLIYKVSN
RDSGVPDRFSGSGSGTDFTLKITRVEAEDVGVYYCMQGTHWPHTFGQGT
KLEIKRTVAAPSVF
BPx-20 EVQLVQSGGGLVKPGRSLRLSCTASGFTFGDYAMSWFRQAPGKGLEWVS 312
YISSSGSTIYYADSVKGRFTISRDNAKNSLYLQMNSLRVEDTAVYYCAREG
WGSLLDYWGQGILVTVSSGSASAPTLGGGGSGGGGSAAAEIVMTQSPATL
SLSPGERATLSCRASQSVNSSYFAWYHRRPGQAPRLLISRTSTRAAGIPDRF
SGSGSGTDFTLKISRVEAEDVGVYYCMQGTYWPYTFGQGTKLEIKRTVAA
PSVF
BPx-21 QVQLVQSGGGLVQPGGSLRLSCAASGFTFSSYAMHWVRQAPGKGLEWV 313
AVISYDGSNKYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCA
KDGDLGRYFDWLPLYYYYGMDVWGQGTTVTVSSGSASAPTLGGGGSGG
GGSAAAQSALTQPPSASGSPGQSVTISCTGTSSDVGGYNYVSWYQQHPGK
APKLMIYD VSNRPS GVSNRFS AS KS GNT ASLTIS GLQ AEDE AD Y YCS S YTS
SSTLDVVFGGGTKLTVLGQPKAAPSVTLFPPSS
BPx-22 EVQLVQSGGGVVQPGRSLRLSCAASGFTFSSYSMNWVRQAPGKGLEWISS 314
ITGSSSTIYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCARGGA
RADYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAAQSALTQPASMSGS
PGQSITISCTGTSSDLGGYNYVSWYQQHPGKAPKLMIYDVSKRPSGVSNRF
SGSKSGNTASLTISGLQAEDEADYYCSSYTSSSTPWVFGGGTKLTVLGQPK
AAPSVTLFPPSS
BPx-23 QVQLVQSGGGVVQPGRSLRLSCAASGFTFSSYGMHWVRQAPGKGLEWV 315
AVISYDGSNKYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCAT
PGPYYYYGMDVWGQGTTVTASSGSASAPTLGGGGSGGGGSAAANFMLT
QPHSVSESPGKTVTISCTRSSGSIASNYVQWYQQRPGSSPTTVIYEDNQRPS
GVPGRFSGSIDSSSNSASLTISGLQTEDEADYYCQSYDGSDVVFGGGTKLT
VLSQPKAAPSVTLFPPSS BP5b-6 EVQLVQSGAEVKKPGASVKVSCKASGYTFTGYYMHWVRQAPGQGLEW 316
MGWINPNSGGTNYAQKFQGRVTVTRNTSISTAYMELSSLRSEDTAVYYCA
RGIGDYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAAQSALTQPASVSG
SPGQSITISCTGTSSDVGGYNYVSWYQQHPGKAPKLMIYDVSKRPSGVSNR
FSGSKSGNTASLTISGLQAEDEADYYCSSYTSSSTWVFGGGTKLTVLGQPK
AAPSVTLFPPSS
BPx-24 QVQLVQSGGGVVQPGRSLRLSCAASGFTFSDYGMHWVRQAPGKGLEWV 317
AGISYDGSNKYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYHCA
KDAGRWLEIPYYFDFWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAANF
MLTQPHSVSESPGKTVTISCTGSSGSIASNYVQWYQQRPGSSPTTLIYEHNQ
RPSGVPDRFSGSVDSSSNSASLTISGLKTEDEADYYCQSYDDNNVVFGGGT
KLTVLGQPKAAPSVTLFPPSS
QVQLQESGPGLVKPSETLSLTCTVSGGSTSSSSYYWGWIRQPPGKGLEWIG 318
SIYYSGSTYYNPSLKSRVTISVDTSKNQFSLKLSSVTAADTAVYYCARVVW
FGEPLFDPWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAAQSALTQPPSA
BPx-25
SGSPGQSVTISCTGTSSDIGGYNYVSWYQVHPGKAPKLIIYDVSKRPSGIPD
RFSGSKSGNAASLTVSGLRTEDEADYYCSSYAGNNNLIFGGGTKVTVLSQ
PKAAPSVTLFPPSS
QVQLVQSGGGLVQPGRSLRLSCAASGFTFDDYAMHWVRQAPGKGLEWV 319
SGISWNSGSIGYADSVKGRFTISRDNAKNSLYLQMNSLRAEDTALYYCAK
DLSGYDQAIDYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAAQSALTQ
BPx-26
PASVSGSPGQSITISCTGTSSDVGGYNYVSWYQQHPGKAPKLMIYDVSKRP
SGVSNRFSGSKSGNTASLTISGLQAEDEADYYCASYTSSTSVVFGGGTKLA
VLGQPKAAPSVTLFPPSS
QVQLVQSGAEVKKPGESLKISCKGSGYSFTSYWIGWVRQMPGKGLEWMG 320
IIYPGDSDTRYSPSFQGQVTISADKSISTAYLQWSSLKASDTAMYYCVRSGS
GWAIDYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAADVVMTQTPLSL
BPx-27
PVTPGQPASISCRSSQSLVYSNGNTYLTWFQQRPGQSPRRLLHEVSKRDSG
VPDRFVGSGSGTDFTLKISRVEPEDVGIYYCMQGSQWPPTFGQGTKLEIKR
TVAAPSVF
QVQLVQSGAEVKKPGASMKASCKASGYTFTSYYIQWVRQAPGQGLEWV 321
GVIHPGDGGTTYAQKFQGRVTMTTDTSTSTVYMELSSLRSEDTAVYYCAR
GDLDNWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAADIVMTQTPVSLP
BPx-28
VTLGQPASISCRSSQSLVHSDGNTYLNWFQQRPGQSPRRLIYKVSTRDSGV
PDRFSGSGSATDFTLKISGVEAEDVGVYYCMQGTHWPKTFGQGTKVEIKR
TVAAPSVF
QVQLVQSGAEVKKPGESLKISCKGSGYSFTSYWIGWVRQMPGKGLEWMG 322
IIYPGDSDTRYSPSFQGQVTISADKSISTAYLQWSSLKASDTAMYYCARRY
SSGWGFDYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAAEIVMTQSPLS
BPx-29
LPVTPGEPASISCRSSQSLLHKNGNNYLDWYLQKPGQSPQLLIYMASKRAS
GVPDRFSGSGSGTDFTLKISRVEAEDVGVYYCMQALQTPPTFGGGTKVEIK
RTVAAPSVF
QVTLKESGPTLVKPTQTLTLTCTFSGFSLSTSGVGVGWFRQPPGKALEWLA 323
LIYWDDDKRYSPSLKSRLTITKDTSKNQVVLTMTNMDPVDTATYYCAHG
EMDLFGYSSGWFPDFDYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAA
BPx-30
EIVLTQSPATLSLSPGERATLSCRASQSITSNYLAWYQQKPGQAPRLLIVGA
SSRATGIPDRFSGSGSGTDFTLTISRLEPEDSAVYYCQQYGNSPRTFGQGTK
LEIKRTVAAPSVF
EVQLLETRAEVKKSGESLKISCQGSGYSFTAHWIAWVRQMPGKGLEWMG 324
SIYPADSTTVYSPSFQGQVTISADKSFTTAYLQWTSLKASDTAMYYCVRQT
SGWSDWGQGTLVTVSAGSASAPTLGGGGSGGGGSAAADIQMTQSPSTLS
BPx-31
ASVGDRVTITCRASQSISSWLAWYQQKPGKAPKLLIYKASSLESGVPSRFS
GSGSGTEFTLTISSLQPEDFATYYCQQYQSYPITFGQGTRLEIKRTVAAPSV
F QVQLVQSGGGVVQPGGSLSLSCAVSGFTFSSYGMHWVRQAPGKGLEWV 325
AVISYDGSNKYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCAR
GDGYNHPLYYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAAEIVLTQSP
BPx-32
LSLPVTPGEPASISCRSSESLLHSIGYNYVDWYVQKPGQSPQLLIYLGSNRA
SGVPDRFSGSTSGTDFTLKISRVEAEDVGVYYCMQALQTSITFGQGTRLEI
KRTMAAPSVF
QVQLVQSGGGVVQPGRSLRLSCAASGFTFSSYGMHWVRQAPGKGLEWV 326
AVISYDGSNKYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCAR
G
BPx-33 TLWFGELSVDYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAAQSALTQ
PASVSGSPGQSITISCTGTSSDVGTYNYVSWYQQHPGKAPKLMIYDVNNRP SGVSNRFSGSKSGSTASLTISGLQAEDEADYYCSSYTSTSTPYVFGTGTKVT VLRQPKANPTVTLFPPSS
QVTLKESGPTLVKPTQTLTLTCTFSGFSLSTSGMGVSWIRQPPGKALEWLA 327
LIDWDDDKYYSTSLKTRLTISKDTSKNQVVLTMTNMDPVDTATYYCARM
YSYGATEFDYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAAQSALTQP
BPx-34
PSASGSPGQSVTISCTGTSSDIGGYNYVSWYQQHPGKAPKLMIYEVSERPS
GVPDRFSGSKSGNTASLTIAGLQGEDEADYYCSSYRSSSTVVFGGGTKLTV
LGQPKAAPSVTLFPPSS
QVQLVQSGGGLVQPGRSLRLSCAASGFTFGDYAMHWVRQAPGKGLEWV 328
SGISWNSGSIGYADSVKGRFTISRDNAKNSLYLQMNSLRAEDTALYYCAK
DISSGWYSVGYFDLWGRGTLVTVSSGSASAPTLGGGGSGGGGSAAAQSA
BPx-35
LTQPASVSGSPGQSITISCTGTSSDVGGYNYVSWYQQHPGKAPKLMIYDVS
NRPSGVSNRFSGSKSGNTASLTISGLQAEDEADYYCSSYTSSSTLVFGTGTK
VT VLGQPKANPT VTLFPPS S
EVQLVESRAEVKKPGASVKVSCKASGYTFSDYYIHWVRQAPGQGLEWM 329
GRISPNSGGTNYAQKFQGRVTMTRDTSMNTAYMELSRLNSDDTAVYYCA
RGFFDYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAADIQMTQSPSFLS
BPx-36
ASVGDRVTITCRASQGISSHLAWYQQKPGKAPKLLIYATSTLQSGVPSRFS
GSGSGTDFTLTISSLQPEDFATYYCQQFDDLPVTFGPGTKVEIKRTVAAPSV
F
QVQLVQSGGGVVQPGRSLRLSCAASGFTFSSYGMHWVRQAPGKGLEWV 330
AVISYDGTNKYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCA
RGQTSYFYDSSGPEIPNFDYWGQGTLVTVSSGSASAPTLGGGGSGGGGSA
BPx-37
AADIVMTQSPLSLPVTPGEPASISCRSSQTLLHANGFNYLDWYLQKPGQSP
QLLMYLGSSRASGVPDRFSGSGSGTDFTLKISRVEAEDVGVYYCMQTLQT
PPWTFGQGTKVEIKRTVAAPSVF
QMQLVQSGAEVKKPGSSVKVSCKASGGTFSSYAISWVRQAPGQGLEWMG 331
GIIPIFGTANYAQKFQGRVTITADESTSTAYMELSSLRSEDTAVYYCARGG
GYYDSSGYPYYYYGMDVWGQGTTVTVSSGSASAPTLGGGGSGGGGSAA
BPx-38
ADIVLTQSPATLSVSPGDRATLSCRASQSVNRNLAWYQQRPGQAPRILIYD
ASTRATGIPTRFRGSGSGTEFTLTISSLQSEDFGIYYCQQYNNWLSFGGGTK
VEVKRTVAAPSVF
QVQLVQSGAEVKKPGASVKVSCKASGYTFTSYGISWVRQAPGQGLEWM 332
GWISAYNGNTNYAQKLQGRVTMTTDTSTSTAYMELRSLRSDDTAVYYCA
RSTGYSSSSDWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAAQPVLTQPP
BPx-39
SVSVSPGQTASITCSGDKLGDKYASWYQQKPGQSPVMVIYQDRKRPSGIPE
RFSGSNSGNTATLTISGTQAMDEADYYCAAWDDSLSGRVFGGGTKLTVL
GQPKAAPSVTLFPPSS
QVQLVQSGAEVKKPGASVKVSCKASGYTFTSYGISWVRQAPGQGLEWM 333
GWISAYNGNTNYAQKLQGRVTMTTDTSTSTAYMELRSLRSDDTAVYYCA
BPx-40 RDVGASGLLWFGELSFDYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAA
ANFMLTQPHSVSESPGKTVTISCTRSSGTIVSTYVQWYQQRPGSSPTTVIYE
DNQRPSGVPDRFSGSIDSSSNSASLTISGLKTEDEADYYCQSYDSSNRKVFG GGTKLTVLSQPKAAPSVTLFPPSS
QIQLVQSGAEVKKPGSSVKVSCKASGGTFSSYAISWVRQAPGQGLEWMG 334
GIIPIFGTANYAQKFQGRVTITADESTSTAYMELSSLRSEDTAVYYCARDRP
HAGLYYYYYGMDVWGQGTTVTVSSGSASAPTLGGGGSGGGGSAAAQSA
BPx-41
LTQPASVSGSPGQSITISCTGTSSDVETYNLVSWYQQHPGKAPKLMIYDVT
KRPSGVPDRFSGSKSGNTASLTISGLQADDEADYYCSSYADSIAFVIFGGGT
KVTVLGQPKAAPSVTLFPPSS
EVQLVQSGGGLVKPGGSLRLSCAASGFTFSSYGMHWVRQAPGKGLEWVA 335
VISYDGSNKYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCAK
DLLYYYDSSGPVDYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAADIV
BPx-42
MTQSPATLSLSPGERATLSCRASQSFSSNLAWYQQKPGQAPRLLIYDASNG
ATGVPDRFSGSGSGTDFTLTISRLEPEDFAVYYCQQYGSSPRTFGQGTKLEI
K
QVQLVQSGGVVVQPGGSLRLSCAASGFTFDDYTMHWVRQAPGKGLEWV 336
SSISSSSSYIYHADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCVDSS
GYFDAFDIWGQGTMVTVSSGSASAPTLGGGGSGGGGSAAAQSVLTQPPSV
BPx-43
SGAPGQRVTISCTGSSSNVGAGYDVHWYQQLPGTAPKLLIYGNTNRPSGV
PERFTGSKSGTSASLAISGLRSEDEADYYCATWDDSLNGPVFGGGTKLTVL
GQPKAAP
QVQLVQSGGGVVRPGGSLRLSCAVSGFTFDDYGMSWVRQAPGKGLEWV 337
SGINWNGGSTGYADSVKGRFTISRDNSKNTLYLQMNSPRAEDTAVYYCA
BP16b- REDYGDSEYYFDYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAAQSVL
1 TQPPSVSGAPGQRVTISCTGSTSNIGAGYDVHWYQQLPGTAPKLLIYGNTN
RPSGVPDRFSGSKSGTSASLAISGLQSEDEADYYCAAWDDSLNGWVFGGG
TKLTVLGQPKAAPSVTLFPPSS
QVQLVQSGAEVKKPGSSVKVSCKASGGTFSSYAISWVRQAPGQGLEWMG 338
GIIPIFGTANYAQKFQGRVTITADESTSTAYMELSSLRSEDTAVYYCARKG
GYYGSGSYFDYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAAQAVLTQ
BPx-44
PPSASGTPGQRVTISCSGSSSNIGSNYVYWYQQLPGTAPKLLIYRNNQRPSG
VPDRFSGSKSGTSASLAISGLRSEDEADYYCAAWDDSLSGWVFGGGTKLT
VLGQPKAAPSVTLFPPSS
QVQLVESGAEVKKPGSSVKVSCKASGGTFSSYAISWVRQAPGQGLEWMG 339
GIIPIFGTANYAQKFQGRVTITADESTSTAYMELSSLRSEDTAVYYCAVEED
YGGSFDYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAAQSVLTQPPSAS
BPx-45
GTPGQRVTISCSGSNSNIGSNHVYWYQQLPGTAPKLLIYSNNQRPSGVPDR
FSGSKSGTSASLAISGLQSEDEADYYCAAWDDNVNGWVFGGGTKLTVLG
QPKAAPSVTLFPPSS
EVQLVESGAEVKKPGESLKISCKGSGYSFTSYWIGWVRQMPGKGLEWMG 340 IIYPGDSDTRYSPSFQGQVTISADKSISTAYLQWSSLKASDTAMYYCARSRS GSYYGTDAFDIWGQGTMVTVSSGSASAPTLGGGGSGGGGSAAAQSALTQ
BPx-46
PASVSGSPGQSITISCTGTSSDVGGYNYVSWYQQHPGKAPKLMISDVSKRP SGVSNRFSGSKSGNTASLTISGLQAEDEADYYCSSYTSNYTLVFGGGTKLT VLGQPKAAPSVTLFPPSS
EVQLVESGAEVKKPGASVKVSCKASGYTFTSYGISWVRQAPGQGLEWMG 341
WISAYNGNTNYAQKLQGRVTMTTDTSTSTAYMELRSLRSDDTAVYYCAR
VDFYDYVWGSYLLDYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAAQ
BPx-47
AVLTQPPSASGTPGQRVTISCSGSSSNIGTNAVNWYQQLPGTAPKHLIYSN
NQRPSGVPDRFSASKSGTSASLAISGLQSDDESDYYCAAWDDSLNGVVFG
GGTKLTVL
QMQLVQSGAEVKKPGSSVKVSCKASGGTFSSYAISWVRQAPGQGLEWMG 342
GIIPIFGTANYAQKFQGRVTITADESTSTAYMELSSLRSEDTAVYYCARDLV
BPx-48 YSSGGPDYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAAQPVLTQSPSA
SGTPGQRVTISCSGSSSNIGSNYVYWYQQLPGTAPKLLIYRNSQRPSGVPD
RFSGSKSGTSTSLAISDLRSEDEADYYCAAWDDSLSGWVFGGGTKLTVLG QPKAAPSVTLFPPSS
QVQLVQSGAEVKKPGASVKVSCKASGYTFTGYYMHWVRQAPGQGLEW 343
MGRISPNSGGTNYAQKLQGRVTMTTDTSTSTAYMELRSLRSDDTAVYYC
ARRWDYDSSGYQNDYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAADI
BPx-49
VMTQSPATLSLPPGERATLSCRASQSVSSYLAWYQQKPGQAPRLLIYDAS
NRATGIPARFSGSGSGTDFTLTISSLQAEDVAVYFCQQYYSTPLTFGGGTK
VEIKRTVAAPSVF
QVQLVQSGGGLVQPGGSLRLSCVTSGFSFSNYGMSWVRQAPGKGLEWVS 344
AISGSGGSTYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCARD
QWIQLWIDYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAAQPVLTQPPS
BPx-50
ASGTPGQRVTISCSGSSSNIGSNTVNWYQQLPGTAPKLLIYINNQRPSGVPD
RFSGSKSGTSASLAISGLQSEDEANYYCAAWDDSLSGPVFGGGTKLTVLG
QPKAAPSVTLFPPSS
QIQLVQSGAEVKKPGSSVKVSCKASGGTFSSYAISWVRQAPGQGLEWMG 345
GIIPIFGTANYAQKFQGRVTITRDMSTSTAYMELSSLRSEDTAVYYCAAEV
PAAKLYYYYYGMDAWGQGTTVTVSSGSASAPTLGGGGSGGGGSAAAQA
BPx-51
VLTQPPSASGAPGQTVTISCSGSSSNIGSKTVNWYQQVPGTVPKLLIYNNN
QRPSGVPDRFSGSKSGTSASLAITGLRSEDEADYYCAAWDDSLSGPMFGG
GTKLT VLGQPKA APS VTLFPPS S
QIQLVQSGAEVKKPGSSVKVSCKASGGTFSSYAISWVRQAPGQGLEWMG 346
GIIPIFGTANYAQKFQGRVTITADKSTSTAYMELSSLRSEDTAVYYCARDH
LYYYDSSGYYDYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAADIVMT
BPx-52
QSPSALSASVGDRVTITCRASQSIGSWLAWFQQKPGKAPNLLIYKASSVES
GVPSRFSGSGSGTEFTLTISSLQPEDFATYYCQQSYSTPYTFGQGTKLEIKRT
VAAPSVF
QVQLVQSGAEVKKPGSSVKVSCKASGGTFSSYAISWVRQAPGQGLEWMG 347
GIIPIFGTADYAQKFQGRVTITADESTSTAYMELSSLRSEDTAVYYCATYY
DSSGYYPYYFDYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAAQAVLT
BPx-53
QPPSVSVAPGKTAKITCGGLNIGSKSVHWYQQKAGQAPVLVMSYDSDRPS
GIPERFSGSNSGSTATLSISRVAAGDEADYYCQVSDSDTDHPVFGGGTKLT
VL
QVQLVQSGAEVKKPGASVKVSCKASGYTFTSYGISWVRQAPGQGLEWM 348
GWISAYNGNTNYAQKLQGRVTMTTDTSTSTAYMELRSLRSDDTAVYYCA
RDFKIAARGMDVWGQGTTVTVSSGSASAPTLGGGGSGGGGSAAADIVMT
BPx-54
QSPLSLPVTPGEPASISCRSSQSLLHSNGYNYLDWYLQKPGQSPQLLIYLGS
NRASGVPDRFSGSGSGTDFTLKISRVEAEDVGVYYCMQALQTPYTFGQGT
KLEIKRTVAAPSVF
QVQLVQSGGGVVQPGRSLRLSCAASGFTFSSYGMHWVRQAPGKGLEWV 349
AVIWYDGSNKYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCA
RGPHYFSDYGDYDSIGYYYYGMDVWGQGTAVTVSSGSASAPTLGGGGSG
BPx-55
GGGSAAASSELTQDPAVSVALGQTVRITCQGDSLRTYYASWYQQKPGQA
PKLLIYGKNNRPSGIPERFSGSSSRNSASLTITGTQAEDEADYYCDSRDSSTS
QWVFGGGTKVTVLGQ
QVQLVQSGAEVKKPGASVKVSCKASGGTFSSYAISWVRQAPGQGLEWMG 350
GIIPIFGTANYAQKFQGRVTITADESTSTAYMELSSLRSEDTAVYYCARDK
GSSGYYNFDYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAAQPVLTQP
BPx-56
PSASGTPGQRVTISCSGTSSNIGSNTVSWYHHLPGTAPKLLIYSNNQRPSGV
PDRFSGSRSGTSAALAISGLESEDEADYYCATWDDSLNGWVFGGGTKLTV
LGQPKAAPSVTLFPPSS
QIQLVQSGAEVKKPGASVKVSCKASGYTFTSYYMHWVRQAPGQGLEWM 351
GIINPSGGSTSYAQKFQGRVTMTRDTSTSTVYMELSSLRSEDTAVYYCARE
BPx-57 VLGDYYYGMDVWGQGTTVTVSSGSASAPTLGGGGSGGGGSAAADIVMT
QSPDSLAVSLGERATIKCQSSQSVLYSFSNKNYLAWYQQKPGQPPKLLIY
WASTRKSGVPDRFSGSGSGTDFSLTISSLQAEDVAVYYCQQYYLLPITFGQ GTRLEIK
QVQLVQSGAEVKKPGESLKISCKGSGYSFTSYWIGWVRQMPGKGLEWMG 352
IIYPGDSDTRYSPSFQGQVTISADKSISTAYLQWSSLKASDTAMYYCARYY
YGSGSYFDLLDYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAAQPVLT
BPx-58
QPPSVSVAPGKTARITCGGNNIGSKSVHWYQQKPGQAPVLVIYYDSDRPS
GIPERFSGSNSGNTATLTISRVEAGDEADYYCQVWDSSSDHVVFGGGTKL
T VLGQPKA APS VTLFPPS S
QVQLVQSGGGLVQPGGSLRLSCAVSGFTFGTYAMTWVRQAPGKGLEWV 353
SAISGGGDSTYYGDSVKGRFTVSRDNSKNTLYLQMSSLRVEDTALYYCAK
ATREDVQHGSFDYWGQGTLVTVSSRSASAPTLGGGGSGGGGSAAAEVVL
BPx-59
TQSPLSLPVTPGEPASISCRSSQSLLHSIGYNYVDWYVQKPGQSPRLLIYLG
SNRASGVPDRFSGSGSGTDFTLKITRVEAEDVGVYYCMQALQTIFTFGPGT
KVDIKRTVAAPSVF
QVQLVQSGAEVKKPGSSVKVSCRASGGTFSSYAISWVRQAPGQGLEWMG 354
GIIPIFGTANYAQKFQGRVTITADESTSTAYMELSSLRSEDTAVYYCARGVL
GGGFDYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAAQPGLTQPPSAS
BPx-60
GTPGQRVTISCSGSSSNIGSNYVYWYQQLPGTAPKLLIYRNNQRPSGVPDR
FSGSKSGTSASLAISGLRSEDEGDYYCASWDDSVHGWVFGGGTKLTVLGQ
PKAAPSVTLFPPSS
QVQLVQSGAEVKKPGASVKVSCKASGYAFTSYGISWVRQAPGQGLEWM 355
GWISAYNGNTNYAQKLQGRVTMTTDTSTSTAYMGLRSLRSDDTAVYYCA
RDGGVLGDYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAAQSALTQPA
BPx-61
SVSASPGQSITISCTGTSSDVGSYNYVSWYQQHPGKAPKLIIFDVNNRPSGV
SNRFSGSKSGNTASLTISGLQAEDEADYYCSSDTSSSTLVFGGGTKLTVLG
QPKAAPSVTLFPPSS
QVQLQESGGGVVQPGRSLRLSCAASGFTFSNYDMHWVRQAPGKGLEWV 356
AVISYDGSNKYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCAR
RTPFDYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAADIVMTQSPDSLA
BPx-62
VSLGERATISCKSSQSVLYSSNNKNYLAWYQQKPGHPPKLLIYGASTRESG
VPDRFSGSGSETDFTLTISSLQAEDAAVYYCMQAAQLPLTFGGGTRVEIRR
TVAAPSVF
QVQLVQSGAEVKKPGASVKVSCKASGYTFTSYGISWVRQAPGQGLEWM 357
GWISAYNGNTNYAQKLQGRVTMTTDTSTSTAYMELRSLRSEDTAVYYCA
RDYGGINDAFDIWGQGTMVTVSSGSASAPTLGGGGSGGGGSAAAQSVLT
BPx-63
QPPSVSAAPGQKVTISCSGSSSNIGNQYVSWYQHLPGTAPKLLIYDNNKRP
SGIPDRFSGSKSGTSATLDITGLQTGDEADYYCGTWDTSLSGFLFAGGTKL
TVLSQPKAAPSVTLFPPSS
QVQLQESGPGLVKPSETLSLTCTVSGGSISSSSYYWGWIRQPPGKGLEWIG 358 SIYHSGSTYYNPSLKSRVTISVDTSKNQFSLKLSSVTAADTAVYYCAAADD
BPx-64
AFDIWGQGTMVTVSSGSASAPTLGGGGSGGGGSAAAQSVLTQPPSSSASP GESARLTCTLPSDINVGQPKAAPSVTLFPPSS
QIQLVQSGAEVKKPGSSVKVSCKASGGTFSSYAISWVRQAPGQGLEWMG 359
GIIPIFGTANYAQKFQGRVTITADKSTSTAYMELSSLRSEDTAVYYCARDQ
WLGGLSNYYYYGMDVWGQGTTVTVSSGSASAPTLGGGGSGGGGSAAAQ
BP6b-5
SVLTQPPSVSGAPGQRVTISCTGSGSNIGRGYDVHWYQQLPGTAPKLLIYG
NTKRPSGVPERFAASKSGTSATLDITGLQTGDEADYYCGTWDTSLSGFLFA
GGTKLTVLSQPKAAPSVTLFPPSS
QVQLVESGGGLVQPGRSLRLSCAASGFTFSSYGMHWVRQAPGKGLEWVA 360
VISYDGSNKYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCAK
DLLAGGSYYYYYYGMDVWGQGTTVTVSSGSASAPTLGGGGSGGGGSAA
BPx-65
AQSVLTQPPSASGTPGQRVTISCSGSSSNIGSNTVNWYQQLPGTAPKLLIHT
NNQRPSGVPDRFSGSKSGTSASLAISGLQSEDEADFYCAAWDDSLNGPVF
GGGTKLT VLGQPKA APS VTLFPPS S
BPx-66 QIQLVQSGGGLVKPGGSLRLSCAASGFPFSNAWMSWVRQAPGKGLEWVA 361 NIEQDGSEKYYVDSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCAKE
EYGDHYYFDYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAANFMLTQP
HSVSESPGKTVTISCTRSSGRIASTYVQWYQHRPGSAPNIVIYEDDQRPSGV
PDRFSGYVDNSSNSASLTISGLKPEDEAEYYCQSYDGDNAVFGGGTKLTV
LGQPKAAPSVTLFPPSS
EVQLLETRGGLVQPGRSLRLSCATSGFTFGDYAMHWVRQAPGKGLEWVS 362 AISGSGGSTYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCAKD LATYYDFWSGLFDYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAAQSA
BPx-67
LTQPASVSGSLGQSITISCTGTSGDVETYNLVSWYQQHPGKVPKLIIYEVSE RPSGVSNRFSGSKSGNTASLTISGLQAEDEADYYCSSYAGSGTLRLFGGGT KVTVLGQPKAAPSVTLFPPSS
EVQLVQSGGGLVQPGGSLRLSCAASGFTFDDYGMSWVRQAPGKGLEWV 363
AVISYDGSNKYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCSG
GSYYGAFDYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAAQSVLTQPP
BPx-68
SVSAAPGQKVTISCTGSSSNIGAGYDVHWYQQVPGTAPKLLIYGNSYRPSG
VSDRFSGSKSGTSASLAISDLQPEDEAYYYCATWDDSLRGDVVFGGGTKL
T VLGQPKA APS VTLFPPS S
QVQLVQSGAEVKKPGESLKISCKGSGYSFTSYWIGWVRQMPGKGLEWMG 364
IIYPGDSDTRYSPSFQGQVTISADKSISTAYLQWSSLKASDTAMYYCARKSS
SGWSIDYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAADVVMTQSPLS
BPx-69
LPVTPGEPASISCRSSQSLLHSNGYNYLDWYLQKPGQSPQLLIYLASNRAS
GVPDRFSGSGSGTDFTLKISRVEAEDVGVYYCMQALQTPATFGQGTKLEI
KRTVAAPSVF
EVQLLESGGGLVQPGGSLRLSCAASGFTFSSYAMSWVRQAPGKGLEWVS 365
AISGSGGSTYYADSVKGRFTISRDNAKNSLYLQMNSLRAEDTAVYYCAGG
SYESSAYHYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAAQPVLTQPPS
BPx-70
MSVSPGQTASITCSGHKLGHKYVGWYQQRPGQSPVLVIYQDNKRPSGIPE
RFSGSNSGNTATLTISETQAIDEADYYCQAWDSSSTAYVFGTGTKVTVLG
QPKANPT VTLFPPS S
QVQLVQSGAEVKKPGASVKVSCKASGYTFTSYGISWVRQAPGQGLEWM 366
GWINTYNGNTNYAQKFQGRVTMTTDTSTSTTYMELRSLRSDDTAVYYCA
REGWVGAKGGFDYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAAQSA
BPx-71
LTQPPSASGSPGQSVTISCSGTSSDVGGYNYVSWYQQYPGKAPKLVISEVS
KRPSGVPDRFSGSKSGNTASLTVSGLQAEDEAEYYCSSYVGGNIWVFGGG
TKLTVLGQPKAAPSVTLFPPSS
EVQLVQSGGGVVQPGRSLRLSCAASGFTFSSYGMHWVRQAPGKGLEWVS 367
SISSTSSYIYYAESVKGRFTISRDNAENSLYLQMNSLRGEDTAVYYCARDR
GGYAYGDFDYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAADIQMTQS
BPx-72
PSTLSASVGDRVTITCRASQSISSYLNWYQQKPGKAPKLLIYAASSLQSGVP
SRFSGSGSGTDFALTISSLQPEDFATYYCQQSYSTPWTFGQGTKVEIKRTVA
APSVF
QVQLVQSGAEVKKPGSSVKVSCKASGGTFSSYTISWVRQAPGQGLEWMG 368
RIIPILGIANYAQKFQGRVTITADKSTSTAYMELSSLRSEDTAVYYCARTNS
GTLDYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAAQPVLTQPPSASGT
BPx-73
PGQRVTISCSGSSSNIGSNYVYWYQQLPGTAPKLLMYRNNQRPSGVPDRFS
GSKSGTSASLAISGLRSEDEADYYCAAWDDSLSGWVFGGGTKLTVLGQPK
AAPSVTLFPPSS
QVQLVQSGAEVKKPGASVKVSCKASGYTFTSYDINWVRQATGQGLEWM 369
GWMNPNSGNTGYAQKFQGRVTMTRNTSISTAYMELSSLRSEDTAVYYCA
RGDFWSGYYYYYYGMDVWGQGTTVTVSSGSASAPTLGGGGSGGGGSAA
BPx-74
AEIVMTQSPGTLSLSPGERVTLSCRASQYITSGYLAWYQQKPGQAPRLLFY
GASRRATGIPDRFSGSGSGTDFTLTISRLEPEDFAVYYCQQYGSSPTYTFGQ
GTKLEIKRTVAAPSVF
BP8b-l QVQLQQSGPGLVKPSQTLSLTCAISGDSVSSNSVTWDWIRQSPSRGLEWLG 370 RTYYRSKWYNDYAVSVKSRITINPDTSKNQFSLHLSSVTPEDTAVYYCAR
QNGGYDYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAAEIVLTQSPGT
LSLSPGERATLSCRASESISNNYLAWYQQKPGQAPRLLIYGASSRATGIPDR
FSGRGSGIDFTLTISRLEPEDFAVYYCQQYGYSPTFGPGTKVDVKRTVAAP
SVF
QVQLVQSGAEVKKPGASVKVSCKASGYTFTGYYMHWVRQAPGQGLEW 371
MGWINPNSGGTNYAQKFQGRVTITRDTSASTAYMELSSLRSEDTAVYYCA
RDGGVEMATIPGGYGMDVWGQGTTVTVPSGSASAPTLGGGGSGGGGSA
BPx-75
AADIQMTQSPSSLSASVGDRVTVTCRASQSISIFVNWYQQRPGKAPKLLIY
AASSLQTGVPSRFSGSGSGTEFTLTISSLQPEDYATYYCQQSYSMLSWTFGP
GTKVENKRTVAAPSVF
QVQLVQSGAEVKKPGASVKVSCKASGYTFTSYGISWVRQAPGQGLEWM 372
GGIIPIFGTANYAQKFQGRVTITADKSTSTAYMELSSLRSEDTAVYYCARG
RIGSGYDSVWLRYFDYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAAQ
BPx-76
AVLTQPPSASGTPGQRVTISCSGSSSNIGSNYVSWYQQLPGTAPKLLIFSNN
QRPSGVPDRFSGSKSGTSASLAISGLQSEDEADYYCAAWDDSLNGWVFGG
GTKLT VLGQPKA APS VTLFPPS S
EVQLVQSGGGLVQPGRSLRLSCAASGFTFGDYAMHWVRQAPGKGLEWV 373
SGISWNSGSIGYADSVKGRVTISRDNAKNSLYLQMNSLRDEDTAVYYCAR
EDEGSGSYYWGPVDYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAAQS
BPx-77
ALTQPASVSGSPGQSITISCTGTSSDVGAYNYVSWYQQYPGKAPKLMIYD
VSKRPSGVSNRFSGSKSGNTASLTISGLQAEDEADYYCGSYTTSSTLVFGG
GTKVT VLGQPKA APS VTLFPPS S
QVQLVESGGGLVQPGGSLRLSCAASGFTFSSYWMSWVRQAPGKGLEWV 374 ANIKQDGSEKYYVDSVKGRFTISRDNAKNSLYLQMNSLRAEDTAVYYCA RGYSNYAYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAASSELTQAPS
BP6b-6
VSVSPGQTASITCSGDKLGDKYVSWYQQRPGQSPVVVIYQDIRRPSGIPER FS GSNS GNT ATLTIS GTQ AMDE AD Y YCQ AWDS S S W VFGGGTKLT VLS QP KAAPSVTLFPPSS
QVQVVQSGAEVKKPGASVKVSCKASGYTFTGYYMHWVRQAPGQGLEW 375
MGWINPNSGGTNYAQKFQGRVTMTRDTSISTAYMELSRLRSDDTAVYYC
ARKSGTMVRGPMWGSFDYWGQGTLVTVSSGSASAPTLGGGGSGGGGSA
BPx-78
AAEIVLTQSPLSLPVTPGEPASISCRSSQSLLHSNGYNYLDWYLQKPGQSPQ
LLIYLGSNRASGVPDRFSGSVSGTDFTLKISRVEAEDVGVYYCMQGTHWS
YTFGQGTKLEIKRTVAAPSVF
QIQLVQSGGGLVKPGGSLRLSCAASGFTFSDYYMSWIRQAPGKGLEWVSY 376
ISSSGSTIYYADSVKGRFTISRDNAKNSLYLQMNSLRAEDTAVYYCAKDPG
WLQLDYYFDYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAAQSALTQP
BPx-79
ASVSGSPGQSITISCTGTSSDIGTYNYLSWYQQHPGKAPKLMIYDVVYRPS
GVSNRFSGSKSGNTASLTISGLQAEDEADYYCSSYAGSSTVVFGGGTKLTV
LGQPKAAPSVTLFPPSS
QVQLVQSGAEVKKPGSSVKVSCKASGGTFSSYAISWVRQAPGQGLEWMG 377
GIIPIFGTADYAQKFQGRATITADESTSTAYMELSSLRSEDTAVYYCARDR
YCSGGSCYIYYGMDVWGQGTTVTVSSGSASAPTLGGGGSGGGGSAAADI
BPx-80
QMTQSPASLSVSIGDRVTITCQASQDIANHLNWYQQKPGDVPKLLIFDATH
MERGVPSRFDGSRSGTDFTFTISSLQPEDVATYYCQQFDELPLTFGGGTKV
RIKGTVAAPSVF
QVQLVQSGGGVVQPGRSLRLSCAASGFTFSSYAMHWVRQAPGKGLEWV 378
AVISYDGSNKYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCAR
BPx-81 RTVGLGMDVWGQGTTVTVSSGSASAPTLGGGGSGGGGSAAANFMLTQP
HSVSESPGKTITISCTRNSGRIASNYVQWYQQRPGSPPTTLIYEDSLRPSGVP
DRFSGSIDTSSNSASLTISGLETEDEADYYCQSYDSRVYVFGTGTKVTAL
EVQLLESRGGVVQPGRSLRLSCAASGFTFSSYGMHWVRQAPGKGLEWVA 379
BPx-82
VISYDGSNKYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCARG YGSGSSWYFDYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAASSELTQ DPAVSVALGQTVRITCQGDSLKKYYASWYQQKPGQAPVLVIYGKNLRPS GIPERFSGSSSGDTASLTISATQAEDEADYYCNSRDSSGNPIFGGGTKLTVL GQPKAAPSVTLFPPSS
EVQLVQSGGGVVQPGRSLRLSCAASGFTFSSYGMHWVRQAPGKGLEWV 380
AVISYDGSNKYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCAR
BPx-83 ALDLDYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAADVVMTQSPLSL
PVTLGQPASISCRSSQSLVYSDGNTFLNWFQQRPGQPPRRLIYKVSNRDFG
VPDRFSGSGSGTDFTLKISRVEADDVGVYYCMQGTHWPFTFGPGTK
QMQLVQSGAEVKKPGSSVKVSCKASGGTFSSYAISWVRQAPGQGLEWMG 381
IINPSGGSTSYAQKFQGRVTMTRDTSTSTVYMELSSLRSEDTAVYYCARGV
VWRWLQSGGSGYFDYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAAQ
BPx-84
SVLTQPPSVSEAPRQRVTISCSGSSSNIGNNAVNWYRQLPGKAPKLLIYDN
NKRPSGIPDRFSGSKSGTSATLGITGLQTGDEADYYCGTWDSSLSAWVFG
GGTKLT VLGQPKA APS VTLFPPS S
QVQLVQSGAEVKKPGASVKVSCKASGYTFTSYGISWVRQAPGQGLEWM 382
GWISAYNGNTNYAQKLQGRVTMTTDTSTSTAYMELRSLRSEDTAVYYCA
BP6b- RGYYGSGAMDVWGQGTTVTVSSGSASAPTLGGGGSGGGGSAAASSELTQ
11 DPAVSVALGQTVRITCQGDSLKKYYASWYQQKPGQAPVLVIYGRNLRPS
GIPERFSGPYSGNTASLTITGAQAEDEADYYCNSRDSRGNLLVFGGGTRLI
VLGQPKAAPSVTLFPPSSKASGA
EVQLVQSGAEVKKPGESLKISCKGSGYSFTTYWIGWVRQMPGKGLEWMG 383
IIYPGDSDTRYSPSFQGQVTISADKSISTAYLQWSSLKASDTAMYYCARYS
BP18b- YGYTFLDYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAADIVMTQSPD
4 SLAVSLGERATINCKSSQSVLYSSNNKNYLAWYQQKPGQPPKLLIYWAST
RESGVPDRFSGSGSGTDFTLTISSLQAEDVAVYYCQQYYTIPHTFGQGTKL
EIKRT V A APS VFKAS GA
QVQLQESGPGLVKPSETLSLTCTVSGGSISSSSYYWGWIRQPPGKGLEWIG 384 SIYYSGSTYYNPSLKSRVTISVDTSKNQFSLLLTSMTAADTAVYYCARTGT TRGDYFDNWGQGAKVTVSSGSASAPTLGGGGSGGGGSAAAQPVLTQPPS
BP6b-2
VSVSPGQTASITCSGDKLGDKYASWYQQKPGQSPLLVIYQDNKRPSGIPER FS GSNS GNT ATLTIS GTQ AMDE AD Y YCQ AWDS ST VMFGGGTKLT VLGQP KA APS VTLFPPS SKAS GA
EVQLVQSGAEVKKPGASVKVSCKASGYTFTSYDINWVRQAPGQGLEWM 385
GIINPSGGSTSYAQKFQGRVTMTRDTSTSTVYMELSSLRSEDTAVYYCARA
BP6b- FDSSGYSYGMDVWGQGTTVTVSSGSASAPTLGGGGSGGGGSAAADIVMT
10 QSPLSLPVTPGEPASISCRSSQSLLHTNGYNFLDWYLQKPGQSPQLLIHLGS
YRASGVPDRFSGSGSGTDFTLKISRVAAEDVGVYYCMQALHTPMYTFGQ
GTKLEIKRTVAAPSVFKASGA
QVQLVQSGAEVKKPGSSVKVSCKASGYTFTSYYMHWVRQAPGQGLEWM 386
GIINPSGGSTSYAQKFQGRVTMTRDTSTSTVYMELSSLRSEDTAVYYCARD
BP16b- IDRATNWFDPWGQGTLVTVSSGSASVPTLGGGGSGGGGSAAANFMLTQP
2 HSVSESPGKTVTISCTGSSGSIASYYVQWYQQRPGSSPTTVIYEDNQRPSGV
PDRFSGSIDSSSNSASLTISGLKTEDEADYYCQSYDSSNRWVFGGGTKLTV
LGQPKAAPSVTLFPPSSKASGA
QVQLVQSGGGLVQPGGSLRLSCAASGFTFSSYWMSWVRQAPGKGLEWV 387 ANIKQDGSEKYYVDSVKGRFTISRDNAKNSLYLQMNSLRAEDTAVYYCA
BP18b- RDKWWPEGYYYYYYGMDVWGQGTTVTVPSGSASAPTLGGGGSGGGGS
5 AAAEIVMTQSPLSLPVTPGEPASISCRSSQSLVHSNGNTYLNWFHQRPGQS
PRRLIYEVSNRDSGVPDRFSGSGSGTDFTLKISRVEAEDVGVYYCMQGTH WPYTSGQGTKLEIYRTVAAPSVFKASGA
QVQLVQSGAEVKKPGSSVKVSCKASGGTFSSYAISWVRQAPGQGLEWMG 388
BP7b-l GIIPIFGTANYAQKFQDRVTITADESTSTAYMELSSLRSEDTAVYYCARDG
YCSSTSCYVWGEFDPWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAADIQ MTQSPSSLSASVGDRVTITCRASQSISRHLNWYQQKPGKAPKLLIYAASTL QSGVPSRFSGSGSGTDFTLTISSLQPEDFATYYCQQSASTPLTFGGGTKVEI KRTVAAPSVFKASGA
EVQLVQSGGGLVKPGGSLRLSCAASGFTFSSYGMHWVRQAPGKGLEWVS 389
YISSGSSTLYYADSVKGRFTISRDNAKNSLYLQMNSLRAEDTAVYYCARD
FGWYGSFDYWGQGTTVTVSSGSASAPTLGGGGSGGGGSAAADIVMTQSP
BP5b-7
LSLPVTLGQSASISCRSSQGLVYSDGNTFLSWFHQRPGQSPRRLIYMVSNR
DSGVPDRFSGSGSGTDFTLKISRVEAEDVGIFYCMQGTHWPWTFGQGTKV
EIKRT V A APS VFKAS GA
QVQLVQSGGGVVQPGRSLRLSCAASGFTFSSYALHWVRQAPGKGLEWVA 390
VLSYDGSNKYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCAR
AAHSGYDLNYYYGMDAWGQGTTVTVSSGSASAPTLGGGGSGGGGSAAA
BPx-85
DIQMTQSPSSLSASVGDRVTITCRASQNIINYLNWYQQKPGKAPDLLIHTAS
NLQSGVPSRFSGSGFGTDFSLTITSLQPEDFATYYCQHSFSSPYTFGQGTKL
EIKRT V A APS VFKAS GA
QVQLVQSGAEVKKPGESLKISCKGSGYSFTSYWIGWVRQMPGKGLEWMG 391
IIYPGDSDTRYSPSFQGQVTISADKSISTAYLQWSSLKASDTAMYYCAKLR
BP18b- VGYNFFDYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAADIVMTQSPL
6 SLPVTPGEPASISCRSSQTLLHSNGFNYLDWYLQKPGQSPQLLMYLGSSRA
SGVPDRFSGSGSGTDFTLKISRVEAEDVGVYYCMQALQIPRTFGQGTKLEI
KRTVAAPSVFKASGA
EVQLVESRGGVVQPGRSLRLSCAASGFTFSSYAMHWVRQAPGKGLEWVA 392
VISYDGSNKYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCVG
BP20b- VDYYDSSGYPADYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAADIQM
1 TQSPSSLSASVGDRVTITCRASQSISSYLNWYQQKPGKAPKLLIYAASSLQS
GVPSRFSGSGSGTDFSLTITNLQPEDIATYYCQQTYIYPRTFGQGTKVEIKR
TV A APS VFKAS GA
QVQLVQSGAEVKKPGESLKISCKGSGYSFTSYWIGWVRQMPGKGLEWMG 393
IIYPGDSDTRYSPSFQGQVTISVDKSINTAYLQWNSLKASDTAMYYCARG
WSSADYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAAEVVMTQSPLSL
BPx-86
PVTLGQPASISCRSSQSLVYSDGNTYLNWFQQRPGQSPRRLLYKVSNRDSG
VPDRFSGTGSGTDFTLRISRVEAEDVGVYYCMQGSHWPLTFGGGTKVEIK
RTVAAPSVFKASGA
QVQLVESGGGVVQPGRSLRLSCAASGFTFSSYGMHWVRQAPGKGLEWVS 394
VIYSGGSTYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCARD
BP19b- WAFGGSYKLGDYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAAQSALT
3 QPASVSGSPGQSITISCTGTSNDIGRYDYVSWYQQHPGKAPKLIIYAVKARP
SGVSSRFSGSKSGNMASLTISGLQAEDEADYYCGSYTRSSTARFGTGTRVT
VLGQPKANPT VTLFPPSS KAS GA
QVQLVQSGGGLVQPGGSLRLSCAASGFTFSSYSMNWVRQAPGKGLEWVS 395
SISSSSSYIYYADSVKGRFTISRDNAKNSLYLQMNSLRAEDTAVYYCAKDR
FFTYYYDTSGRGGLDSWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAASS
BPx-87
ELTQDPSVSVSPGQTATITCSGSKLGDKYVSWYQQKPGQSPVLVIYEDDK
RPSGS SERFS GSNS GNT ATLTIS GTQ AMDE AD Y YCQ AWDSIT A VFGGGTK
LTVLGQPKAAPSVTLFPPSSKASGA
EVQLVQSGGGLVQPGGSLRLSCAASGFTFSGSSMNWVRQAPGKGLEWVS 396
YISSSGNTIYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCAKGS
EEMRGWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAASSELTQAPSVSVS
BPx-88
PGQTASIPCSGDNLRSKYVSWYQQKPGQSPVMLIYQDDKRPSGIPEGFFGA
TSGNTATLIIAETQPLDEGDYYCQTWDGNHVVFGGGTKLTVLGQPKAAPS
VTLFPPSSKASGA
QVQLVQSGAEVKKPGESLKISCKGSGYSFTSYWIGWVRQMPGKGLEWMG 397
BP18b-
IIYPGDSDTRYSPSFQGQVTISADKSISTAYLQWSSLKASDTAMYYCARHD
7
GNSRPGDYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAAEIVMTQSPLS LPVTLGQPASISCRSSQGLVYSDGNTYLNWFHQRPGQSPRRLIYKVSNRDS
GVPDRFSGSGSGTDFTLKISRVEAEDVGIYYCMQGTQWPYTFGQGTRLDI
KRTVAAPSVFKASGA
QVQLQQSGGGLVQPGGSLRLSCAASGFTFSSYAMSWVRQAPGKGLEWVS 398
AISGSGGSTYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCAKT
BP17b- VAGTRLYYYYYGMDVWGQGTTVTVSSGSASAPTLGGGGSGGGGSAAAQ
1 SALTQPASVSGSPGQSITISCTGTSSDVGGYNYVSWYQQHPGKAPKLMIYD
VSKRPSGVSNRFSGSKSGNTASLTISGLQAEDEADYYCSSYTSSSTLVFGG
GTKLT VLGQPKA APS VTLFPPS SKAS GA
EVQLVQSGAEVKKPGASVKVSCKVSGYTLTELSMHWVRQAPGKGLEWM 399
GGFDPEDGETIYAQKFQGRVTMTEDTSTDTAYMELSSLRSEDTAVYYCAT
BP16b- DFYCSGGSCYSRGDAFDIWGQGTMVTVSSGSASAPTLGGGGSGGGGSAA
3 AEIVLTQSPATLSLSPGERATLSCRASQSVSSYLAWYQQKPGQAPRLLVYD
ASNRATGIPARFSGSGSGTDFTLTISSLEPEDFAVYYCQQRSGWPPKVTFGG
GTKVEIKRTVAAPSVFKASGA
QVQLVQSGGGLVQPGGSLRLSCAASGFTFSSYSMNWVRQAPGKGLEWVS 400
SISSSNSYIYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTALYYCARDW
SIYPASVDFFDYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAADIQMTQ
BPx-89
SPSSLSASVGDRVTITCRASQSISSWLAWYQQKPGKAPKLLIYKASSLESGV
PSRFSGSGSGTEFTLTITSLQPEDFATYYCQQSYSTPFTFGPGTKVDIKRTVA
APSVFKASGA
EVQLVESGGGLVKPGGSLRLSCAASGFTFSSYSMNWVRQAPGKGLEWVS 401 SISSSSSYIYYADSVKGRFTISRDNAKNSLYLQMNSLRAEDTALYYCAKDG RLGELSHWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAAQPVLTQPPSVS
BPx-90
VSPGQTTNITCSGHELGDKYVSWYQQRPGQSPVVVIYQDNKRPSGIPERFS GSNS GNT ATLTIS GTQ AMDE AD Y YCQ AWDS ST VVFGGGTKLT VLGQPKA APSVTLFPPSSKASG
EVQLVESGGGVVQPGRSLRLSCAASGFTFSSYGMHWVRQAPGKGLEWVA 402
VISYDGSNQYYADSVKGRFTISRDNSKNTLYVQMNSLRAEDTAVYYCAR
GNYYGSGSYPLLDYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAASSE
BP5b-8
LTQAPSVSVAPGRTARITCGGNNIGNKSVHWYQQQPGQAPVLVIYYDIDR
PSGIPERFSGSNSGNTATLTISRVEADDEADYYCQVWDSNSDHVVFGGGT
KLT VLGQPKA APS VTLFPPSS KAS GA
QVQLVQSGGGLVQPGGSLRLSCAASGFTFSNYWMYWVRQAPGKGLVWV 403
SNINSDGSSTSYADSVKGRFTISRDNAKNSLYLQMNSLRDEDTAVYYCAR
BP16b- EGYNAHYFDYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAAQSALTQP
4 ASVSGSPGQSITISCTGTSSDVGGYNYVSWYQQHPGKAPKLLIYDVTKRPS
GVSNRFSGSKSGNTASLTISGLQAEDEADYYCSSYTSSSTVVFGGGTKLTV
LGQPKAAPSVTLFPPSSKASGA
QVQLQQSGPRQVEPSQTLSLTCAISGDSVSSKSAAWNWIRQSPSRGLEWL 404
GRTYYRSKWYYDYAVSVKSRIIVNPDTSRNQFSLHLNSVTPEDTAFYYCA
RGEDSGYGSFDYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAANFMLT
BP9b-l
QPPSASGTPGQRVTISCSGSSSNIGNNAVNWYQQLPGTAPKLLIYSNNQRPS
GVPDRFSGSKSGTSASLAISGLQSEDEADYYCATWDDSLNGWVFGGGTKL
TVLGQPKAAPSVTLFPPSSKASGA
QVQLVQSGAEVKKPGASVKVSCKASGYTFTSYGISWVRQAPGQGLEWM 405
GWISAYNGNTNYAQKLQGRVTMTTDTSTSTAYMELRSLRSDDTAVYYCA
RDMKWLPRGVDYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAADIVLT
BPx-91
QSPLSLPVTPGEPASISCRSSQSLLHSNGFNYLDWYLQKPGQSPQLLIYLGS
NRASGVPDRFSGSGSGTDFTLQISRVEAEDVGVYYCMQGLQTPYTFGQGT
KLEIKRTVAAPSVFKASGA
QIQLVQSGAEVKKPGESLKISCKGSGYSFTSYWIGWVRQMPGKGLEWMGI 406
BPx-92 IYPGDSDTRYSPSFQGQVTISADKSISTAYLQWSSLKASDTAMYYCARLGF
REIDYWGQGTLVTVSPGSASAPTLGGGGSGGGGSAAAQSVLTQPPSVSAA PGQTVTISCSGSTSNIGNNYVSWYEQLPGTAPKLLIYDNNRRPSGIPDRFSG SKSGTSAALGITGLQTGDEANYYCGTWDSSLSAWVFGGGTKLTVLGQPK AAPSVTLFPPSSKASGA
EVQLLESRAEVKKPGESLKISCKGSGYSFTSYWIGWVRQMPGKGPEWMGI 407
IYPGDSDTRYSPSFQGQVTISADKSISTAYLQWSSLKASDTAMYYCARLVY
BP12b- GGYDEPGYYFDYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAAQSVLT
1 QPPSASGTPGQRVTISCSGSSSNIRSNTVNWYQQLPGTAPKLLIYSNNQRPS
GVPDRFSGSKSGTSASLAISGLQSEDEADYYCAAWDDSLNGWVFGGGTK
LTVLGQPKAAPSVTLFPPSSKASGA
QVQLVQSGGGVVQPGRSLRLSCAASGFTFSSHGMHWVRQAPGKGPEWV 408
AVISYDGSNKYYPDSVRGRFTISRDNSKNTLYLQMNSLTTDDTAVYYCAK
GGGYGDYAPGYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAANFMLT
BPx-93
QPHSVSGSPGKTVTISCTRSSGSISSKYVQWYQQRPGSSPTTVMYEDKQRP
SGVPARFSGSVDSSSNSATLTISGLKTEDEADYYCQSYDNTNVVFGGGTKL
TVLGQPKAAPSVTLFPPSSKASGA
QVQLVQSGAEVKKPGSSVKVSCKASGGTFSSYAISWVRQAPGQGLEWMG 409
GFDPEDGETIYAQKFQGRVTMTEDTSTDTAYMELSSLRSEDTAVYYCATG
FWELEAGGFDYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAAEVVLTQ
BPx-94
SPLSLPVTPGEPASISCRSSQSLLHSNGYNYLDWYLQKPGQSPQLLIYMGSN
RASGVPDRFSGSGSGTDFTLKISRVEAEDVGVYYCMQALQTPFTFGPGTK
VDIKRT V A APS VFKAS GA
QVQLQQSGPGLVKPSETLSLTCTVSGGSISSSSYYWGWIRQPPGKGLEWIG 410 SIYYSGSTYYNPSLKSRVTISVDTSRKQVSLELSSVTAADTAVYYCARLWL GERPPDYWGQGMLVTVSSGSASAPTLGGGGSGGGGSAAANFMLTQPHSV
BPx-95
SGSPGKTVTISCTGSNGAIASNYVQWYQQRPGGAPVTLIYEDNHRPPGVPD RFSGSIDTSSNSASLTISGLKTEDEADYYCQSYDSVYNWVFGGGTKLTVLG QPKAAPSVTLFPPSSKASGA
QVQLVQSGAEVKKPGASVKVSCKASGYTFTSYYMHWVRQAPGQGLEW 411
MGIINPSGGSTSYAQKFQGRVTMTRDTSTSTVYMELSSLRSEDTAVYYCA
RDRGSGWYGGAFDIWGQGTMVTVSSGSASAPTLGGGGSGGGGSAAAQS
BPx-96
ALTQPASVSGSPGQSITISCSGTSSDVGVYESVSWYQHHPGEAPKLMIYDV
TKRPSGVSNRFSGSKSGNTASLTISGLQAEDEADYYCSSYSTSSTFEVFGGG
TRLTVLSQPKAAPSVTLFPPSSKASGA
QIQLVQSGGGVVQPGRSLRLSCAASGFTFSSYAIHWVRQAPGKGLEWVAV 412
ISYDGSIKYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCARDT
HEWELFVGTFDIWGQGTMVTVSSGSASAPTLGGGGSGGGGSAAADIQMT
BP6b-9
QSPSSLSASVGDRVTITCRASQGISSWLAWYQQKPGKAPKLLIYAASSLQS
GVPSRFSGSGSGTDFTLTISSLQPEDFATYYCQQSYSTPWTFGQGTKVQIKR
TV A APS VFKAS GA
QVQLVQSGGGLVKPGGSLRLSCAASGFTFSSYSMNWVRQAPGKGLEWVS 413 SISSNSSYIYYADSVKGRFTISRDNAKNSLYLQMNSLRAEDTAVYYCARDP DYYYGSGTREDYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAADIQMT
BPx-97
QSPSTLSASVGDRVTITCRASQSISRWLAWYQQKPGEAPKLLIYKASTLEG GVPSRFS GSGS GTEFTLTIS SLQPEDS AT Y YCLQD YTTP YTFGQGTKLEIKR TV A APS VFKAS GA
QVQLVESGGGVVQPGRSLRLSCAASGFTFSSYAMHWVRQAPGKGLEWV 414
AVISYDGSNKYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCAR
VEWADGAFDIWGQGTMVTVSSGSASAPTLGGGGSGGGGSAAAQSVLTQP
BPx-98
PSTSGTPGQRVTISCSGSSSNIGSNTVDWYQQLPGTAPNLLIHSNNRRPSGV
PDRFSGSKSATSASLAISGLQSEDEADYYCEAWDDSLSAVVFGGGTRVTV
LGQPKAAPSVTLFPPSSKASGA
EVQLVESGGGLVQPGGSLRLSCAASGFTFSSYWMSWVRQAPGKGLEWVA 415
BPx-99 NIKQDGSEKYYVDSVKGRFTISRDNAKNSLYLQMNSLRAEDTAVYYCAR
GLIGDWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAADIVMTQTPDSLAV SLGERATISCKSSQSILYDSNLKNYLAWYQHKPGHPPKLLISWASTRESGV
PDRFSGSGSDTDFTLTISSLQAEDVAVYSCQQFYTTPYTFGQGTKLEIKRTV
AAPSVFKASGA
EVQLVQSGGGVVQSGRSLRLSCVASGFSFSNYGMHWVRQAPGKGLEWL 416
AFIWYDGSNKWYADSVKGRFTISRDNSKNALYLQMNSLRAEDTAVYYCA
RDRYSSSWGGGFDYWGQGTVLTVSSGSASAPTLGGGGSGGGGSAAANF
BP6b-7
MLTQPHSVSESPGRTVTIPCSGRGGSIASDSVQWYQQRPGSAPTTIIYEDNQ
RPSGVPDRFSGSVDSSSNSASLTISGLRTEDEADYYCQSYDASSLWVFGGK
TKLT VLGQPKA APS VTLFPPSS KAS GA
QIQLVQSGGGVVQPGRSLRLSCAASGFTFSNAWMSWVRQAPGKGLEWVG 417
RIKSKTDGGTTDYAAPVKGRFTISRDDSKNTLYLQMNSLKTEDTAVYYCT
BPx- TDRVTMVRGVTPPFDYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAAQ 100 SALTQPPSASGSPGQSVTISCTGTSRDVGRYNYVSWYQQHPGKAPKLMISE
VSKRPSGVPDCFSGSKSGNTASLTISGLQAEDEADYYCCSYSGGYNWVFG
GGTKLT VLGQPKA APS VTLFPPS SKAS GA
QLQLQQSGGGLVQPGGSLRLSCAASGFTFSSYWMHWVRQAPGKGLVWV 418
SRINSDGSSTSYADSVKGRFTISRDNAKNTLYLQMNSLRAEDTAVYYCAR
DGWGSYRYVPGYYYYYGMDVWGQGTTVTVSSGSASAPTLGGGGSGGG
BP6b-8
GSAAAQSVLTQPPSASGTPGQRVTISCSGSSSNIGSNTVNWYQQLPGTAPK
LLIYSNNQRPSGVPDRFSGSKSGTSATLDITGLQTGDEADYHCQSYDSSLS
DSVVFGGGTKLTVLGQPKAAPSVTLFPPSSKASGA
QVQLVQSGAEVKKPGESLKISCKGSGYSFTSYWIGWVRQMPGKGLEWMG 419
IIYPGDSDTRYSPSFQGQVTISADKSISTAYLQWSSLKASDTAMYYCARGS
BPx- GWTNILDYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAAEVVLTQSPD 101 SLAVSLGERATINCKSSQSILSNSNNKNYLTWYQQKPGQPPQLIISWASTRE
SGVPDRFSGSGSGTDFTLTINSLQAEDVAVYYCQQYYSTPLTFGGGTKVEI
KRTVAAPSVFKASGA
QVQLVQSGAEVKKPGESLKISCKGSGYSFTNYWIGWVRQMPGKGLEWM 420
GIIYPGDSDTRYSPSFQGQVTISADKSISTAYLQWSSLKASDTAMYYCATQ
BPx- PHDRGVWGQGTTVTVSSGSASAPTLGGGGSGGGGSAAADIVMTQSPPSLP 102 VAPGEPASISCRSSQSLLHSDGKNYVDWYLQKPGQSPQLLIFLGSKRASGV
PDRFSGSGSGTDFTLKISRVEAEDVGVYFCMQGTRWPYTFGQGTKLEIKRT
V A APS VFKAS GA
QLQLQQFGPGLVKPSETLSLTCTVSGDSSSSYYWSWIRQPPGKGLEWIGSI 421
YYSGSTYYNPSLKSRVTISVDTSKNQLSLKLNSVTAADTAVYYCARGAYG
BPx- DKDLGVFDYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAANFMLTQPH 103 SVSESPGKTVTISCTGTGDSIASNYVQWYQQRPGSAPTTVIYEDNQRPSGV
PDRFSGSIDSSSNSASLTISGLRTEDEADYYCQSYDSVYNWIFGGGTRLTVL
GQPKAAPSVTLFPPSSKASGA
QVQLQESGPGLVKPSETLSLTCTVSGGSISSSSYYWGWIRQPPGKGLEWIG 422
SIYYSGSTDYSPSLKSRVTISIDTSKNQFSLKVSSVTAADTAVYYCARERGN
BPx- GDYDNWGLGTLVTVSAGSASAPTLGGGGSGGGGSAAAQSVLTQPPSVSA 104 APGQKVTISCSGSSSNIGNNYVSWYQQLPGTAPKLLIYDNNKRPSGIPDRFS
GSKSGTSASLAISGLQPEDEADYYCAVWDDSLNGWVFGGGTKLTVLRQP
KA APS VTLFPPS SKAS GA
EVQLVQSGGGVVRPGGSLRLSCAASGFTFDDYGMSWVRQAPGKGLEWV 423
SGINWNGGSTGYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCA
BPx- RQYVPEYNSGWPYYYYGMDVWGQGTTVTVSSGSASAPTLGGGGSGGGG 105 SAAASSELTQDPSVSVSPGQTASITCSQTKLGDKYVSWYQQKPGQSPVLVI
YEDDKRPSGISERFSGSNSGNTATLTISGTQAVDEADYYCQVYDSSTVVFG
GGTKLT VLGQPKA APS VTLFPPS SKAS GA
EVQLVESGPGLVKPSGTLSLTCAVFGGSISSSDNWWGWVRQPPGEGLEWI 424
BPx- GDISHSGNTIYNPSLKSRVTISVDKSKNQFSLKLGSVTAADTAVYYCARYD
106
SRGYYYPTDAFDIWGQGAMVTVSSGSASAPTLGGGGSGGGGSAAADIQM TQSPSTLSASVGDRVTITCRASQSISRRLAWYQQKPGKAPKLLISGASNLEF GVPATFSGTGSGTEFTLTISSLQPDDFATYYCQQYESYQPPTFGGGTKVEM KRTVAAPSVFKASGA
EVQLVESGGGLEKPGGSLRLSCAASGFTFSNYAMSWVRQAPGKGLEWVS 425
AVNKGGDAAYSADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCAR
GVWAVDAFDIWGQGTMVTVSSGSASAPTLGGGGSGGGGSAAADIQMTQ
BP5b-9
SPSSVSASVGDRVTITCRASQGISNWLAWYQQKPGKAPKLLIYGASSLQSG
VPSRFSGSGSGTDFTLTISSLQFEDFATYYCQQSQSTPVTFGQGTRLEIKRT
V A APS VFKAS GA
EVQLVQSGGGVVQPGRSLRLSCAASGFSFRNYAMHWVRQAPGKGLEWV 426
AVISYDGSNKYYADSVKGRFTISRDNAKNSLYLQMNSLRDEDTAVYYCA
BPx- RDNGYDSSGYYFLDHYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAAE 107 IVMTQSPATLSLSPGDRAALSCRASHSVGSHVDWFQQKPGQAPRLLIYDAS
NRATGIPDRFSGSGSGTDFTLTISRLEPEDFAVYYCQQRSNWPRALSFGGG
TKVEIKRTVAAPSVFKASGP
QVTLKESGPTLVKPTQTLTLTCTFSGFSLSTYGVGVGWIRQPPGKAPEWLA 427
RIDWDDDEYYSTSLKTRLTISKDTSKNQVVLTMTDMDPVDTATYYCTRFV
BPx- GAKADYWGQGILVTVSSGSASAPTLGGGGSGGGGSAAAQSALTQPASVS 108 GSPGQSITISCTGTSSDVGAYNHVSWYQQYPGKAPKLMIYDVSNRPSGVS
DRFSGSKSDNTASLTISGLQAEDEADYYCSSYTSSNTWVFGGGTKLTVLG
QPKAAPSVTLFPPSSKASGA
EVQLVESRGGVVQPGRSLRLSCAASGFTFSSYAMHWVRQAPGKGLEWVA 428
VISYDGSNKYYADSVKGRFTISRDNSKNTLYLQLNSLRAEDTAVYYCARA
BPx- RSPIAVAPNWFDPWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAADIVMT 109 QSPDSLAVSLGERATINCKSSQSVLYSSNNKNYLAWYQQKPGQPPKLLIY
WASTRESGVPDRFSGSGSGTDFTLTISSLQAEDVAVYYCQQYYSTPYTFGQ
GTKLEIKRTVAAPSVFKASGA
QIQLVQSGGGVVQPGRSLRLSCAASGFTFSSYGMHWVRQAPGKGLEWVA 429
VIWYDGSNKYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCAK
BP22b- GLGYHPLFDYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAADIVMTQS
1 PDSLAVSLGERATINCKSSQSVLYSSDNKNYLAWYQQKPGQPPKLLIYWA
STRESGVPDRFSGSGSGTDFTLTISSLQAEDVAVYYCQQYYSTPSFGPGTK
VDIKRT V A APS VFKAS GA
QVQLVESGGGVVQPGRSLRLSCAASGFTFSSYGMHWVRQAPGKGLEWV 430
AVISYDGSNKYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCA
BPx- KDLYSGSGGGDYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAANFMLT 110 QPHSVSESPGKTVTISCTRSSGSIASNYVQWYQQRPGSSPTTVIYEDNQRPS
GVPARFSGSIDISSNSASLTISGLLTEDEADYYCQSYNTSNLVFGTGTKVTV
LRQPKANPTVTLFPPSSKASGA
EVQLVQSGGGLVQPGGSLRLSCAGSGFTFSDYDMSWVRQAPGKGLEWVS 431
GISGSGGSTYYVDSVQGRFTISRDNSKNTLYLQMNGLRAEDTAVYYCAKD
BP6b- SSSGWYGLWDYWGQGTLVSVSSGSASAPTLGGGGSGGGGSAAADIQMT
12 QSPSSVFASVGDRVTITCRASQDISTWLAWYQQKPGKGPKLLIYGASSLQS
GVPSRFS GSGS GTDFTLTIS SLQPDDFAT Y YCQQ YQESPWTFGQGTKVEIK
RTVAAPSVFKASGA
QVQLQESGPGLVKPSETLSLTCTVSGGSISSYYWSWIRQPPGKGLEWIGYI 432
YYSGSTNYNPSLKSRVTMSVDTSKNQLSLNLTSVTAADTAVYYCAREFPM
BPx- GRPFDYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAAQSALTQPPSASG 111 SRGQSVSISCSGSRSDIGYYNYVSWYQQHPGKAPKLIIFDVNKRPSGVSNR
FSGSKSGNTASLTISGLQAEDEADYYCSSYTSSSTWVFGGGTKLTVLGQPK
AAPSVTLFPPSSKASGA
EVQLVESGAEVKKPGASVKVSCKASGYTFTSYGISWVRQAPGQGLEWMG 433
BPx- WISAYNGNTNYAQKLQGRVTMTTDTSTSTAYMELRSLRSDDTAVYYCAR
112
DGMFAPGYFDYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAAQPVLTQ PPSVSVAPGKTARITCGGNNIGSKSVHWYQQKPGQAPVLVVYDDSDRPSG IPERFSGSNSGNTATLTISRVEAGDEADYYCQVWESSSDEWDTTSDQWVF GGGTKVT VLGQPK A APS VTLFPPS SKAS GA
QVTLKESGPVLVKPTETLTLTCAVSGFSLSNPRMGVSWIRQPPGKALEWL 434
ALIYWDDDKRYSPSLKSRLTITKDTSKNQVVLTMTNMDPVDTATYYCAL
BPx- WDDYSNYEIARGAFDIWGQGTMVTVSSGSASAPTLGGGGSGGGGSAAAN 113 FMLTQPHSVSESPGKTVTLSCTGSGGSIARNYVQWYQQRPGSAPSAVIYED
DRRPSGVPHRFSGSIDTSSNSATLTISGLKTEDEADYYCQSYDSAHHWVFG
GGTKLTVLSQPKAAPSVTLFPPSSKASGA
QVQLVQSGGGLVQPGGSLRLSCAASGFTFSSYDMHWVRQATGKGLEWVS 435
AIGTAGDTYYPGSVKGRFTISRENAKNSLYLQMNSLRAGDTAVYYCARG
BPlOb- KPLLSGYLINDAFDIWGQGTMVTVSSGSASAPTLGGGGSGGGGSAAAQSV
1 LTQPPSVSAAPGQKVTISCSGSSSNIGDNYVSWYQQLPGTAPKLLIYDNNK
RPSGIPDRFSGSKSGTSATLGITGLQTGDEADYYCGTWDDRLSGVVFGGGT
KLT VLS QPK A APS VTLFPPSS KAS GA
EVQLLETGGGVVQPGRSLRLSCAASGFSFNNYGMHWVRQAPGKGLEWVS 436
VIYSGGSTYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCARDG
BPx- GSGYHDYWGQGTLVTVSSGSASAPTLGGGGSGGGGSAAADIVMTQSPDS 114 LAVSLGERATINCKSSQSVLYSSNNKNYLAWYQQKPGQPPKLLIYWASTR
ESGVPDRFSGSGSGTDFTLTISSLQAEDVAVYFCQQYYGIPTFGGGTKVEIK
RTVAAPSVFKASGA
[00308] Recombinant GDF-modulating antibodies of the disclosure may comprise or be developed using any of the VH sequences listed in Table 14. Some recombinant GDF- modulating antibodies of the disclosure may comprise or be developed using VH sequences that comprise at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, at least about 99%, at least about 99.5% or at least about 99.9% sequence identity to any of the VH sequences listed in Table 14. Recombinant GDF-modulating antibodies may, in some cases, comprise VH domains presented, but with different combinations of CDRs (e.g. CDR-H1, CDR-H2 or CDR-H3). In some cases, such antibodies may interact with one or more of the recombinant proteins listed in Table 4, 9 or 10.
Table 14. VH domains
SEQ
Clone
scFv sequence ID ID
NO
QVQLQQSGPGLVKPSQTLSLTCAISGDSVSSNSAAWNWIRQSPSRGLEWL
BP6a- GRTYYRSKWSNDYAVAVKSRITINPDTSKNQFSLQLNSVTPEDTAVYYCA 437 16
RGFS GMGVWGQGTT VT VS SGS
QVQLVQSGAEVKKPGSSVKVSCKASGGTFSSYAISWVRQAPGQGLEWMG
BP6a-
GIIPIFGTANYAQKFQGRVTITADESTSTAYMELSSLRSEDTAVYYCARDQ 438 19
AGDYGGNSGYFDYWGQGTLVTVSSGS
EVQLVQSGGGLVQPGRSLRLSCAASGFTFDDYAMHWVRQAPGKGLEWV
BP3a-3 SGISWNSGSTYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCAK 439
GQVPNYYDSRDPFDPWGQGTLVTVSSGS QIQLVQSGAEVKKPGSSVKVSCKASGGTFSSYAISWVRQAPGQGLEWMG
BP6a-l GIIPIFGTANYAQKFQGRVTITADESTSTAYMELSSLRSEDTAVYYCARITD 440
DFDYWGQGTLVTVSSGS
QVQLVQSGAEVKKPGESLKISCKGSGYSFTSYWIGWVRQMPGKGLEWMG
BP18b-
IIYPGDSDTRYSPSFQGQVTISADKSISTAYLQWSSLKASDTAMYYCARGV 441 1
AGYSSGWSDWGQGTLVTVSSGS
QVQLQESGPGLVKPSETLSLTCTVSGGSISSYYWSWIRQPPGKGLEWIGYIY
BP 19b- YSGSTNYNPSLKSRVTISVDTSKNQFSLKLSSVTAADTAVYYCARLRRGYF 442 1
DYWGQGTLVTVSSGS
QVQLVQSGAEVKKPGESLKISCAASGFTFSSYGMHWVRQAPGKGLEWVA
BP6b-4 VIWYDGSNKYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCAT 443
RGYWGQGTLVTVSSGS
QVQLQQSGAEVKKPGASVKVSCKASGYTFTSYYMHWVRQAPGQGLEWM
BP6a-8 GIINPSGGSTSYAQKFQGRVTMTRDTSTSTVYMELSSLRSEDTAVYYCARE 444
GGDYNNWFDPWGQGTLVTVSSGS
QVQLVQSGSEVKKPGASVKVSCKLSGYTLTELSMHWVRQAPGKGLEWM
BP13a-
GGFDPEDDEAIYAQKFQGRVTMTEDTSTDTAYMELSSLRSEDTAVYYCAT 445 1
DRVTGTTMAAFDIWGQGTMVAVSSGS
EVQLVQSGGGLVKPGGSLRLSCAASGFTFSDYYMSWIRQAPGKGLEWVS
BP5b-3 YISSSGSTIYYADSVKGRFTISRDNAKNSLYLQMNSLRDEDTAVYYCARDN 446
WYYFDYWGQGTLVTVSSGS
QVQLVESGGGLVKPGGSLRLSCAASGFTFSSYSMNWVRQAPGKGLEWVS
BP6a- SISSSSSYIYYADSVKGRFTISRDNAKNSLYLQMNSLRAEDTAVYYCARDL 447 23
GGYFGNYWGQGTLVTVSSGS
QVQLVQSGAEVKKPGSSVKVSCKASGGTFSSYAISWVRQAPGQGLEWMG
BP5a-l GIIPIFGTANYAQKFQGRVTITADESTSTAYMELSSLRSEDTAVYYCANHGL 448
MDDSSGYYLSNAFDIWGQGTMVTVSSGS
QIQLVQSGGVVVQPGGSLRLSCAASGFTFDDYTMHWVRQAPGKGLEWVS
BP6a- LISWDGGSTYYADSVKGRFTISRDNSKNSLYLQMNSLRTEDTALYYCAAG 449 11
SYHTWWGQGTLVTVSSGS
QVQLQQSGGGLVQPGGSLRLSCAASGFTFSSYAMTWVRQAPGKGLEWVS
BP5b-4 AISGGGGATYYADSVEGRFTISRDNSKNTLYLQMNSLRAEDTAIYYCASPA 450
GHICSGGSCQYYYYGMDVWGQGTTVTVSSGS
EVQLVQSGAEVKKPGESLKISCKGSGYSFTSYWIGWVRQMPGKGLEWMG
BP18a-
IIYPGDSDTRYSPSFQGQVTISADKSISTAYLQWSSLKASDTAMYYCARQP 451 1
YYYGSGSGMDVWGQGTTVTVSSGS
EVQLVQSGGGLVQPGRSLRLSCAASGFTFDDYAMHWVRQAPGKGLEWV
BPl la- SGISWNSGSIGYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCAK 452 1
VDGLEYSSGHNFDYWGQGTLVTVSSGS
EVQLVESGGGVVQPGRSLRLSCAASGFSFSSYGMHWVRQAPGKGLEWVA
BP6a-
VTSYDGSNKYYGDSVKGRFTISRDNSKNTLFLQMNSLRAEDTAVYYCAK 453 14
DEYTYGSGIMDYWGQGTLVTVSSGS
QMQLVQSGGGVVQPGRSLRLSCAASGFTFSSYAMHWVRQAPGKGLEWV
BP6a- ALISYDGSYEYYADSVKGRFTISRDISKNTLYLQMNSLRAEDTAVYYCAR 454 27
DS GARGYN YGLFD YWGQGTL VT VS SGS
EVQLVESGGGVVQPGRSLRLSCAASGFRFSNYGMHWVRQAPGKGLEWVS
BP6a-
AISGSGGLTYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCARE 455 13
DLWRMGELLGGEGFDYWGQGTLVTVSSGS
EVQLVQSGAEVKKPGASVKVSCKASGYTFSSYGITWVRQAPGQGLEWMG
BP4a-l WSSAYNGNTNYAQKLQGRISMTTDTSTSTAYMELRSLRSDDTAVYYCAR 456
GIYGDYEYWGQGTLVTVSSGS
EVQLVQSGGGLVQPGRSLRLSCAASGFTFDDYAMHWVRQAPGKGLEWV
BPla-1 SGISWNSGSIGYADSVKGRFTISRDNAKNSLYLQMNSLRAEDTALYYCAK 457
DTSNGGYSSSSFDYWGQGTLVTVSSGS QVQLVQSGAEVKKPGSSVKVSCKASGGTFSSYAISWVRQAPGQGLEWMG
BP6a-6 GIIPIFGTANYAQKFQGRVTITADESTSTAYMELSSLRSEDTAVYYCARDRH 458
L YRRGYS S VDGMD VWGQGTT VT VS SGS
QLQLQQSGGGVVQPGRSLRLSCAASGFTFSSYGMHWVRQAPGKGLEWVA
BP 19b- VISYDGSNKYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCAKP 459 2
RGRYYFDYWGQGTLVTVSSGS
QVQLQESGPGLVKPSETLSLTCTVSGDSITSYYWSWIRQPPGKGLEWIGYI
BP6a-4 HYSGSTYYNPSLKSRVTISVDRSKNQFSLKLSSVTAADTAVYYCARGPSRG 460
LFDPWGQGTLVTVSSGS
QVQLQESGGVVQPGGSLRLSCAASGFTFSSYGMSWVRQAPGKGLEWVSSI
BP19a-
SNSGGSTYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCASLSR 461 1
GYWGQGTLVTVSSGS
QVQLQQSGAEVKKPGASVKVSCKASGYTFTSYYMHWVRQAPGQGLEWM
BP6a-7 GIINPSGGSTSYAQKFQGRVTMTRDTSTSTVYMELSSLRSEDTAVYYCARE 462
SLIRFLEDPQQGGMDVWGQGTTVTVSSGS
QVQLVQSGAEVKKPGESLKISCKGSGYSFTSYWIGWVRQMPGKGLEWMG
BP5b-5 IIYPGDSDTRYSPSFQGQVTISADKSISTAYLQWSSLKASDTAMYYCATPYS 463
SSSGTDYWGQGTLVTVSSGS
QIQLVQSGAEVKKPGSSVKVSCKASGGTFSSYAISWVRQAPGQGLEWMG
BP6b-3 WINPNSGGTNYAQKFQGWVTMTRDTSISTAYMELSRLRSDDTAVYYCAR 464
PRTLRGDAFDIWGRGTMVTVSSGS
QVQLVQSGAEVKKPGESLKISCKGSGYSFTSYWIGWVRQMPGKGLEWMG
BP18b- IIYPGDSDTRYSPSFQGQVTISADKSISTAYLQWSSLKASDTAMYYCARHPI 465 2
GSGYDYWGQGTLVTVSSGS
QVQLVQSGAEVKKPGSSVKVSCKASGGAFSIYSINWVRQAPGQGLEWMG
BPx-1 GIIPIFETANSAQKFQGRVTITADKSTSTAYMELSSLRSEDTAVYYCARAGE 466
NSDSNYAFDIWGRGTVVTVSSGS
QLQLQQSGPGLVKPSETLSLTCTVSGGSISSSSYYWGWIRQPPGKGLEWIG
BP21a SIYYSGSTYYNPSLKSRVTISVDTSKNQFSLKLSSVTAADTAVYYCARGRY 467
SYGYFDYWGQGTLVTVSSGS
QVQLVQSGGGLVKPGGSLRLSCAASGFTFSSYSMNWVRQAPGKGLEWVS
BP6a-2 SISSSSSYIYYADSVKGRFTISRDNAKNSLYLQMNSLRAEDTAVYYCARGIR 468
VVGDEDYYYYYGMDVWGQGTTVTVSSGS
EVQLLQSGGGLVQPGGSLRLSCAASGFTFSSYSMNWVRQAPGKGLEWVS
BPx-2 YISSSSSTIYYADSVKGRFTISRDNAKNSLYLQMNSLRDEDTAVYYCARDS 469
SGWLGMGDFDYWGQGTLVTVSSGS
QVQLVESGGGLVKPGGSLRLSCAASGFTFSSYSMNWVRQAPGKGLEWVS
BP6b-l SISSSSSYIYYADSVKGRFTISRDNAKNSLYLQMNSLRAEDTAVYYCAREE 470
YYGGNTDWGQGTLVTVSSGS
EVQLVQSGGGLVQPGGSLRLSCVTSGFSFSNYGMSWVRQAPGKGLEWVS
BPx-3 EISGSGGSAYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCARDI 471
SHYDILTGQLDYWGQGTLVTVSSGS
QIQLVQSGGGVVQPGRSLRLSCAASGFTFSSYGMHWVRQAPGKGLEWVA
BP6a-5 VISYDGSNKYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCARD 472
LLVRFLEWSHYYGMDVWGQGTTVTVSSGS
EVQLVESGGGLVQPGGSLRLSCAASGFTFSSYWMSWVRQAPGKGLEWVA
BP6a- NIKQDGSEKYYVDSVKGRFTISRDNSENTLYLQMNSLRAEDTAVYYCAKE 473 10
TDCS GGIC YGVD YWGQGTLVT VS SGS
QVQLVQSGAEVKKPGASVKVSCKASGYTFTSYGISWVRQAPGQGLEWMG
BP15a- WISAYNGNTNYAQKLQGRVTMTTDTSTSTAYMELRSLRSDDTAVYYCAR 474 1
DAPYDILTGYLSYWGQGTLVTVSSGS
QIQLVQSGGGVVQPGRSLRLSCAASGFSFSTYAMHWLRQAPGKGPEWVA
BP6a- AISYDGTIKYYADSVKGRFTISRDSSKNTLYLQMNNLGPEDTALYYCARA 475 26
GGAGSDGYNYWGYWGQGTLVTVSSGS QVQLVQSGGGLVKPGGSLRLSCAASGFTFSSYSMNWVRQAPGKGLEWVS
BP6a- SISSSSSYIYYADSVKGRFTISRDNAKNSLYLQMNSLRAEDTAVYYCARDP 476 12
GGKLGGAFDIWGQGTMVTVSSGS
QVQLVQSGAEVKKPGSSVKVSCKASGGTLSSYAISWVRQAPGQGLEWMG
BP5a-4 GIIPIFGTANYAQKFQGRVTITADESTSTAYMELSSLRSEDTAVYYCARDRP 477
GNNRYDILTGGGWFDPWGQGTLVTVSSGS
QVQLVQSGGGVVRPGGSLRLSCAASGFTFDDYGMSWVRQAPGKGLEWV
BP5b-l SGISWNSGSIGYADSVKGRFTISRDNAKNSLYLQMNSLRAEDTALYYCAK 478
EISDTAMGFDYWGQGTLVTVSSGS
EVQLVESRGGVVQPGRSLRLSCAASGFTFSSYGMHWVRQAPGKGLEWVA
BPx-4 VISYDGSNKYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCARA 479
YIPRLYYYGSGLDYWGQGTLVTVSSGS
QVQLQESGPGLVKPSETLSLTCTVSGYSITNGYYWGWIRQSPGKGLEWIG
BP5a-3 NIFQSGSTYYNPSFKSRVTISVDTSKNQFSLKLSSVTAADTAVYYCARRVD 480
YYDSSGYGEGYFDYWGQGTLVTVSSGS
QVQLVQFGAEVKKPGESLKISCKGSGYNFNSYWIGWVRQMPGKGLEWM
BP 14a- GIIYPGDSDIRYNPSFQGQVTISADKSISTAYLQWSSLKASDSAMYYCARH 481 1
VSGSLDYWGQGTLVTVSSGS
EVQLVQSGGGLVQPGGSLRLSCAASGFTFSSYAMSWVRQAPGKGLEWVS
BP3a-l AISGSGGSTYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCAKT 482
AYYDSSGYYPDAFDIWGQGTMVTVSSGS
QVQLVQSGAEVKKPGESLKISCKGSGYSFTSYWIGWVRQMPGKGLEWMG
BP18b-
IIYPGDSDTRYSPSFQGQVTISADKSISTAYLQWSSLKASDTAMYYCARGQ 483 3
GGVLGAFDIWGQGTMVTVSSGS
EVQLVESGAEVKKPGEPLKISCKGSGYSFTSYWIGWVRQMPGKGLEWMGI
BP6a-9 IYPGDSDTRYSPSFQGQVTISADKSISTAYLQWSSLKASDTAMYYCARQGR 484
GEAFDIWGQGTMVTVSSGS
QVQLQQSGPGLVKPSQTLSLTCAISGDSVSSNTEAWNWIRQSPSRGLEWLG
BP6a- RTYYRSKWYHDYAPSVKSRITINPDTSKNQFSLQLNSVTPEDTAVYYCART 485 25
VFDS Q V YWFDPWGQGTL VT VS SGS
EVQLVESRGGLVKPGGSLRLSCAASGFTFSSYGMSWVRQAPGKGLEWVSS
BPx-5 ISNSGGSTYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCASLSR 486
GYWGQGTLVTVSSGS
QVQLVQSGGGLIQPGGSLRLSCAASGFTVSSYSMNWVRQAPGKGLEWVS
BP5a-2 YISSSGSTIYYADSVKGRFTISRDNAKNSLYLQMNSLRAEDTALYYCAKVG 487
TAAAGDAFDIWGQGTMVTVSSGS
QVQLQESGPGLVKSSGTLSLTCAISGVSIASSNWWSWVRQPPGKGLEWVG
BP6a-
EIYHSGSTFYNPSLQSRVTISVDKSKNQFSLTLNSVTAADTAVYYCASSSG 488 15
WPYPTNWFDAWGQGTLVTVSSGS
EVQLLESRGSLVQPGGSLRLSCAASGFTFSSYAMSWVRQAPGKGLEWVSG
BP6a- ISDSGGTTYYADSVKGQFTISRDNSKNTLYLQMNSLRAEDTAVYYCAKDA 489 21
HNWNYGVFDYWGQGTLVTVSSGS
QVQLVQSGGGLVQPGGSLRLSCAASGFTFSSYSMNWVRRAPGKGLEWVS
BP5a-5 YISSSSSTIYYADSVKGRFTISRDNAKNSLYLQMNSLRDEDTAVYYCASAL 490
GATGAFDIWGQGTMVTVSSGS
QMQLVQSGGGLVKPGGSLRLSCAASGFTFSDYYMTWIRQAPGKGLEWVS
BP6a- AISGSGGSTYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCAKD 491 17
ISGYEPLYYFDYWGQGTLVTVSSGS
EVQLVESRGGVVQPGRSLRLSCAASGFTFSSYAMHWVRQAPGKGLEWVA
BP6a- VISYDGSNKYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCARD 492 20
PGKQWRVRGYFDYWGQGTLVTVSSGS
EVQLVQSGGDLEQPGGSLRLSCGTSGFTFSDYDMSWVRQAPGKGLEWVS
BP3a-2 GISWNSGSIGYADSVKGRFTISRDNAKNSLYLQMNSLRAEDTAVYYCARE 493
SDMAAAGSFDYWGQGTLVTVSSGS EVQLVESRGGLVKPGGSLRLSCAASGFTFSSYSMNWVRQAPGKGLEWVSS
BP6a- ISSSSSYIYYADSVKGRFTISRDNAKNSLYLQMNSLRAEDTAVYYCAREGN 494 24
SGYDVWGQGTLVTVSSGS
QVQLVQSGAEVKKPGASVKVSCKASGYTFTSYGISWVRQAPGQGLEWMG
BP6a- WISAYNGNTNYAQKLQGRVTMTTDTSTSTAYMELRSLRSDDTAVYYCAR 495 22
DSLWFGELSALDYWGQGTLVTVSSGS
QVQLVQSGGGVVLPGRSLRLSCAASGFTFSSYAMHWVRQAPGKGLEWVA
BP19a- VISYDGSNKYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCARD 496 2
VSSWAAFWGQGTLVTVSSGS
QIQLVQSGAEVKKPGASVKVSCKASGYTFTSYGISWVRQAPGQGLEWMG
BPx-6 WISAYNGNTNYAQKLQGRVTMTTDTSTSTAYMELRSLRSDDTAVYYCAR 497
GGRGGVITWFDPWGQGTLVTVSSGS
QIQLVQSGGGLIQPGGSLRLSCAASGFTFSSFAMSWVRQAPGKGLEWVSSI
BP6a-
TSRGDTTYYADSVRGRFTISRDNSKNTLYLQMNSLRADDTAVYYCARDRP 498 18
QVPYDSSGNFDYWGQGTLVTVSSGS
QVQLVQSGAEVKKPGSSVKVSCKASGGTFSSYAISWVRQAPGQGLEWMG
BP16a- GIIPIFGTADYAQKFQGRVTITADESTSTAYMELSSLRSEDTAVYYCAREEA 499 1
GYSSGWVEDAFDIWGQGTMVTVSSRS
QVQLVQSGGGVVQPGRSLRLSCAASGFTFSSYAMHWVRQAPGKGLEWV
BP19a-
AVISYDGSNKYYADSVKGRFTISRDNAENSLYLQMNSLRAEDTAVYYCAR 500 3
MVGGYRFDYWGQGTTVTVSSGS
EVQPLETGGGLVQPGGSLRLSCVTSGFSFSNYGMSWVRQAPGKGLEWVSE
BP5b-2 ISGSGGSAYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCAKHG 501
DYGGTFDYWGQGTLVTVSSGS
QIQLVQSGAEVKKPGASVKVSCKASGYTFTSYGISWVRQAPGQGLEWMG
BP2a-l WISAYNGNTNYAQKLQGRVTMTTDTSTSTAYMELSSLRSEDTAVYYCAR 502
VGFYDYVWGSYPYDAFDIWGQGTMVTVSSGS
QVQLVQSGAEVKKPGESLKISCKGSGYSFTNYWIAWVRQMPGKGLEWM
BP18a- GIIYPGDSDTRYSPSFQGQVTISADKSISTAYLQWTSLKASDTAMYYCARR 503 2
SSSSGSFDYWGQGTLVTVSSGS
QLQLQQSGGGLVKPGGSLRLSCAASGFTFSSYSMNWVRQAPGKGLEWVS
BP6a-3 SISSSSSYIYYADSVKGRFTISRDNAKNSLYLQMNSLRAEDTAVYYCVRDR 504
HSLGDFDYWGQGTLVTVSSGS
QIQLVQSGGGVVQPGRSLRLSCAASGFTFSSYGMHWVRQAPGKGLEWVA
BPx-7 VISYDGSNKYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCARR 505
FTMDYWGQGTLVTVSSGS
QVQLQESGPGLVKPSETLSLTCTVSGGSISSSSYYWGWIRQPPGKGLEWIG
BPx-8 SIYYSGSTYYNPSLKSRVTISVDTSKNQFSLKLSSVTAADTAVYYCARYSSS 506
WELFDYWGQGTLVTVSSGS
QIQLVQSGAEVRKPGDSVKVSCRTSGYTFNDYHIHWVRQAPGQGLEWMG
BPx-9 WISAYNGNTNYAQKLQGRVTMTTDTSTSTAYMELRSLKSDDTAVYYCAR 507
RNGSGSAPGYWGQGTLVTVSSGS
EVQLVESGAEVKKPGESLRISCKGSGYSFTSYWIGWVRQMPGKGLEWMGI
BPx-10 IYPGDSDTRYSPSFQGQVTISADKSISTAYLQWSSLKASDTAMYYCARVGQ 508
YYGMDVWGQGTTVTVSSGS
QVQLQESGPGLVKPSETLSLACTVSGGSISSSSYYWGWIRQPPGKGLEWIA
BPx-11 SINYSGNTYYNPSLKSRVIIPIDTSKNQFSLKLSSVTAADTAVYYCARRVLG 509
YGYFDYWGQGTLVTVSSGS
QVQLVQSGAEVKKPGSSVKVSCKASGGTFSSYAISWVRQAPGQGLEWMG
BPx-12 GIIPIFGTANYAQKFQGRVTITADESTSTAYMELSSLRSEDTAVYYCTGGGN 510
VDYWGQGTLVTVSSGS
EVQLLESGGGLVQPGRSLRLSCAASGFTFDDYAMHWVRQAPGKGLEWVS
BPx-13 GISWNSGSIGYADSVKGRFTISRDNAKNSLYLQMNSLRAEDTALYYCAKD 511
KAGNYYYGMDVWGQGTTVTVSSGS QVQLVQSGGGVVQPGRSLRLSCAASGFTFGHYGMHWVRQAPGKGLEWV
BPx-14 AVISYDGTNKYYADSVKGRFTISRDNSKNTLYLQMNSLRVDDTAVYYCA 512
KGSRLDPWGQGTLVTVSSGS
QVQLQQSGPGLVKPSQTLSLTCAISGDSVSSNSAAWNWIRQSPSRGLEWL
BPx-15 GRTYYRSKWYNDYAVSVKSRITINPDTSKNQFSLQLNSVTPEDTAVYYCV 513
GQGYDSSGYYGMDVWGQGTTVTVSSGS
QVQLVQSGAEVKKPGESLKISCKGSGYSFTSYWIGWVRQMPGKGLEWMG
BPx-16 IIYPGDSDTRYSPSFQGQVTISADKSISTAYLQWSSLKASDTAMYYCARQY 514
YDTSYFDYWGQGTLVTVSSGS
EVQLVQSGGGLVQPGRSLRLSCAASGFTFGDYAMHWVRQAPGKGLEWV
BPx-17 SGINWNSGGVGYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCA 515
KDSGYSYGINFDYWGQGTLVTVSSGS
QIQLVQSGAEVKKPGASVKVSCKASGYTFTSYGISWVRQAPGQGLEWMG
BPlb-1 WISAYNGNTNYAQKLQGRVTMTTDTSTSTAYMELRSLRSDDTAVYYCAR 516
GDWFDPWGQGTLVTVSSGS
QVTLKESGPTLVKPTQTLTLTCTFSGFSLSTSGVGVGWIRQSPGKALEWLA
BPx-18 LIYWDDDKRYSPSLKSRLTITKDTSKNQVVLTMTNMDPVDTATYYCAHM 517
RWGYYFDYWGQGTLVTVSSGS
EVQLVQSGAEVKKPGESLKISCKGSGYSFTSYWIGWVRQMPGKGLEWMG
BPx-19 IIYPGDSDTRYSPSFQGQVTISADKSISTAYLQWSSLKASDTAMYYCARRE 518
MGAKGSGGIDYWGQGTLVTVSSGS
EVQLVQSGGGLVKPGRSLRLSCTASGFTFGDYAMSWFRQAPGKGLEWVS
BPx-20 YISSSGSTIYYADSVKGRFTISRDNAKNSLYLQMNSLRVEDTAVYYCAREG 519
WGSLLDYWGQGILVTVSSGS
QVQLVQSGGGLVQPGGSLRLSCAASGFTFSSYAMHWVRQAPGKGLEWV
BPx-21 AVISYDGSNKYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCAK 520
DGDLGRYFDWLPLYYYYGMDVWGQGTTVTVSSGS
EVQLVQSGGGVVQPGRSLRLSCAASGFTFSSYSMNWVRQAPGKGLEWISS
BPx-22 ITGSSSTIYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCARGGA 521
RADYWGQGTLVTVSSGS
QVQLVQSGGGVVQPGRSLRLSCAASGFTFSSYGMHWVRQAPGKGLEWV
BPx-23 AVISYDGSNKYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCAT 522
PGPYYYYGMDVWGQGTTVTASSGS
EVQLVQSGAEVKKPGASVKVSCKASGYTFTGYYMHWVRQAPGQGLEW
BP5b-6 MGWINPNSGGTNYAQKFQGRVTVTRNTSISTAYMELSSLRSEDTAVYYCA 523
RGIGDYWGQGTLVTVSSGS
QVQLVQSGGGVVQPGRSLRLSCAASGFTFSDYGMHWVRQAPGKGLEWV
BPx-24 AGISYDGSNKYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYHCAK 524
DAGRWLEIPYYFDFWGQGTLVTVSSGS
QVQLQESGPGLVKPSETLSLTCTVSGGSTSSSSYYWGWIRQPPGKGLEWIG
BPx-25 SIYYSGSTYYNPSLKSRVTISVDTSKNQFSLKLSSVTAADTAVYYCARVVW 525
FGEPLFDPWGQGTLVTVSSGS
QVQLVQSGGGLVQPGRSLRLSCAASGFTFDDYAMHWVRQAPGKGLEWV
BPx-26 SGISWNSGSIGYADSVKGRFTISRDNAKNSLYLQMNSLRAEDTALYYCAK 526
DLSGYDQAIDYWGQGTLVTVSSGS
QVQLVQSGAEVKKPGESLKISCKGSGYSFTSYWIGWVRQMPGKGLEWMG
BPx-27 IIYPGDSDTRYSPSFQGQVTISADKSISTAYLQWSSLKASDTAMYYCVRSGS 527
GWAIDYWGQGTLVTVSSGS
QVQLVQSGAEVKKPGASMKASCKASGYTFTSYYIQWVRQAPGQGLEWV
BPx-28 GVIHPGDGGTTYAQKFQGRVTMTTDTSTSTVYMELSSLRSEDTAVYYCAR 528
GDLDNWGQGTLVTVSSGS
QVQLVQSGAEVKKPGESLKISCKGSGYSFTSYWIGWVRQMPGKGLEWMG
BPx-29 IIYPGDSDTRYSPSFQGQVTISADKSISTAYLQWSSLKASDTAMYYCARRYS 529
S GWGFD YWGQGTL VT VS SGS QVTLKESGPTLVKPTQTLTLTCTFSGFSLSTSGVGVGWFRQPPGKALEWLA
BPx-30 LIYWDDDKRYSPSLKSRLTITKDTSKNQVVLTMTNMDPVDTATYYCAHG 530
EMDLFGYSSGWFPDFDYWGQGTLVTVSSGS
EVQLLETRAEVKKSGESLKISCQGSGYSFTAHWIAWVRQMPGKGLEWMG
BPx-31 SIYPADSTTVYSPSFQGQVTISADKSFTTAYLQWTSLKASDTAMYYCVRQT 531
SGWSDWGQGTLVTVSAGS
QVQLVQSGGGVVQPGGSLSLSCAVSGFTFSSYGMHWVRQAPGKGLEWV
BPx-32 AVISYDGSNKYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCAR 532
GDGYNHPLYYWGQGTLVTVSSGS
QVQLVQSGGGVVQPGRSLRLSCAASGFTFSSYGMHWVRQAPGKGLEWV
BPx-33 AVISYDGSNKYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCAR 533
GTLWFGELSVDYWGQGTLVTVSSGS
QVTLKESGPTLVKPTQTLTLTCTFSGFSLSTSGMGVSWIRQPPGKALEWLA
BPx-34 LIDWDDDKYYSTSLKTRLTISKDTSKNQVVLTMTNMDPVDTATYYCARM 534
YSYGATEFDYWGQGTLVTVSSGS
QVQLVQSGGGLVQPGRSLRLSCAASGFTFGDYAMHWVRQAPGKGLEWV
BPx-35 SGISWNSGSIGYADSVKGRFTISRDNAKNSLYLQMNSLRAEDTALYYCAK 535
DISSGWYSVGYFDLWGRGTLVTVSSGS
EVQLVESRAEVKKPGASVKVSCKASGYTFSDYYIHWVRQAPGQGLEWMG
BPx-36 RISPNSGGTNYAQKFQGRVTMTRDTSMNTAYMELSRLNSDDTAVYYCAR 536
GFFDYWGQGTLVTVSSGS
QVQLVQSGGGVVQPGRSLRLSCAASGFTFSSYGMHWVRQAPGKGLEWV
BPx-37 AVISYDGTNKYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCAR 537
GQTSYFYDSSGPEIPNFDYWGQGTLVTVSSGS
QMQLVQSGAEVKKPGSSVKVSCKASGGTFSSYAISWVRQAPGQGLEWMG
BPx-38 GIIPIFGTANYAQKFQGRVTITADESTSTAYMELSSLRSEDTAVYYCARGG 538
GYYDSSGYPYYYYGMDVWGQGTTVTVSSGS
QVQLVQSGAEVKKPGASVKVSCKASGYTFTSYGISWVRQAPGQGLEWMG
BPx-39 WISAYNGNTNYAQKLQGRVTMTTDTSTSTAYMELRSLRSDDTAVYYCAR 539
STGYSSSSDWGQGTLVTVSSGS
QVQLVQSGAEVKKPGASVKVSCKASGYTFTSYGISWVRQAPGQGLEWMG
BPx-40 WISAYNGNTNYAQKLQGRVTMTTDTSTSTAYMELRSLRSDDTAVYYCAR 540
DVGASGLLWFGELSFDYWGQGTLVTVSSGS
QIQLVQSGAEVKKPGSSVKVSCKASGGTFSSYAISWVRQAPGQGLEWMG
BPx-41 GIIPIFGTANYAQKFQGRVTITADESTSTAYMELSSLRSEDTAVYYCARDRP 541
HAGLYYYYYGMDVWGQGTTVTVSSGS
EVQLVQSGGGLVKPGGSLRLSCAASGFTFSSYGMHWVRQAPGKGLEWVA
BPx-42 VISYDGSNKYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCAKD 542
LLYYYDSSGPVDYWGQGTLVTVSSGS
QVQLVQSGGVVVQPGGSLRLSCAASGFTFDDYTMHWVRQAPGKGLEWV
BPx-43 SSISSSSSYIYHADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCVDSS 543
GYFDAFDIWGQGTMVTVSSGS
QVQLVQSGGGVVRPGGSLRLSCAVSGFTFDDYGMSWVRQAPGKGLEWV
BP 16b-
SGINWNGGSTGYADSVKGRFTISRDNSKNTLYLQMNSPRAEDTAVYYCAR 544 1
EDYGDSEYYFDYWGQGTLVTVSSGS
QVQLVQSGAEVKKPGSSVKVSCKASGGTFSSYAISWVRQAPGQGLEWMG
BPx-44 GIIPIFGTANYAQKFQGRVTITADESTSTAYMELSSLRSEDTAVYYCARKG 545
GYYGSGSYFDYWGQGTLVTVSSGS
QVQLVESGAEVKKPGSSVKVSCKASGGTFSSYAISWVRQAPGQGLEWMG
BPx-45 GIIPIFGTANYAQKFQGRVTITADESTSTAYMELSSLRSEDTAVYYCAVEED 546
YGGSFDYWGQGTLVTVSSGS
EVQLVESGAEVKKPGESLKISCKGSGYSFTSYWIGWVRQMPGKGLEWMGI
BPx-46 IYPGDSDTRYSPSFQGQVTISADKSISTAYLQWSSLKASDTAMYYCARSRS 547
GSYYGTDAFDIWGQGTMVTVSSGS EVQLVESGAEVKKPGASVKVSCKASGYTFTSYGISWVRQAPGQGLEWMG
BPx-47 WISAYNGNTNYAQKLQGRVTMTTDTSTSTAYMELRSLRSDDTAVYYCAR 548
VDFYDYVWGSYLLDYWGQGTLVTVSSGS
QMQLVQSGAEVKKPGSSVKVSCKASGGTFSSYAISWVRQAPGQGLEWMG
BPx-48 GIIPIFGTANYAQKFQGRVTITADESTSTAYMELSSLRSEDTAVYYCARDLV 549
YSSGGPDYWGQGTLVTVSSGS
QVQLVQSGAEVKKPGASVKVSCKASGYTFTGYYMHWVRQAPGQGLEW
BPx-49 MGRISPNSGGTNYAQKLQGRVTMTTDTSTSTAYMELRSLRSDDTAVYYC 550
ARRWDYDSSGYQNDYWGQGTLVTVSSGS
QVQLVQSGGGLVQPGGSLRLSCVTSGFSFSNYGMSWVRQAPGKGLEWVS
BPx-50 AISGSGGSTYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCARD 551
QWIQLWIDYWGQGTLVTVSSGS
QIQLVQSGAEVKKPGSSVKVSCKASGGTFSSYAISWVRQAPGQGLEWMG
BPx-51 GIIPIFGTANYAQKFQGRVTITRDMSTSTAYMELSSLRSEDTAVYYCAAEV 552
PAAKLYYYYYGMDAWGQGTTVTVSSGS
QIQLVQSGAEVKKPGSSVKVSCKASGGTFSSYAISWVRQAPGQGLEWMG
BPx-52 GIIPIFGTANYAQKFQGRVTITADKSTSTAYMELSSLRSEDTAVYYCARDH 553
LYYYDSSGYYDYWGQGTLVTVSSGS
QVQLVQSGAEVKKPGSSVKVSCKASGGTFSSYAISWVRQAPGQGLEWMG
BPx-53 GIIPIFGTADYAQKFQGRVTITADESTSTAYMELSSLRSEDTAVYYCATYYD 554
SSGYYPYYFDYWGQGTLVTVSSGS
QVQLVQSGAEVKKPGASVKVSCKASGYTFTSYGISWVRQAPGQGLEWMG
BPx-54 WISAYNGNTNYAQKLQGRVTMTTDTSTSTAYMELRSLRSDDTAVYYCAR 555
DFKIAARGMDVWGQGTTVTVSSGS
QVQLVQSGGGVVQPGRSLRLSCAASGFTFSSYGMHWVRQAPGKGLEWV
BPx-55 AVIWYDGSNKYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCA 556
RGPHYFSDYGDYDSIGYYYYGMDVWGQGTAVTVSSGS
QVQLVQSGAEVKKPGASVKVSCKASGGTFSSYAISWVRQAPGQGLEWMG
BPx-56 GIIPIFGTANYAQKFQGRVTITADESTSTAYMELSSLRSEDTAVYYCARDK 557
GSSGYYNFDYWGQGTLVTVSSGS
QIQLVQSGAEVKKPGASVKVSCKASGYTFTSYYMHWVRQAPGQGLEWM
BPx-57 GIINPSGGSTSYAQKFQGRVTMTRDTSTSTVYMELSSLRSEDTAVYYCARE 558
VLGDYYYGMDVWGQGTTVTVSSGS
QVQLVQSGAEVKKPGESLKISCKGSGYSFTSYWIGWVRQMPGKGLEWMG
BPx-58 IIYPGDSDTRYSPSFQGQVTISADKSISTAYLQWSSLKASDTAMYYCARYY 559
YGSGSYFDLLDYWGQGTLVTVSSGS
QVQLVQSGGGLVQPGGSLRLSCAVSGFTFGTYAMTWVRQAPGKGLEWVS
BPx-59 AISGGGDSTYYGDSVKGRFTVSRDNSKNTLYLQMSSLRVEDTALYYCAKA 560
TREDVQHGSFDYWGQGTLVTVSSRS
QVQLVQSGAEVKKPGSSVKVSCRASGGTFSSYAISWVRQAPGQGLEWMG
BPx-60 GIIPIFGTANYAQKFQGRVTITADESTSTAYMELSSLRSEDTAVYYCARGVL 561
GGGFDYWGQGTLVTVSSGS
QVQLVQSGAEVKKPGASVKVSCKASGYAFTSYGISWVRQAPGQGLEWM
BPx-61 GWISAYNGNTNYAQKLQGRVTMTTDTSTSTAYMGLRSLRSDDTAVYYCA 562
RDGGVLGDYWGQGTLVTVSSGS
QVQLQESGGGVVQPGRSLRLSCAASGFTFSNYDMHWVRQAPGKGLEWV
BPx-62 AVISYDGSNKYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCAR 563
RTPFDYWGQGTLVTVSSGS
QVQLVQSGAEVKKPGASVKVSCKASGYTFTSYGISWVRQAPGQGLEWMG
BPx-63 WISAYNGNTNYAQKLQGRVTMTTDTSTSTAYMELRSLRSEDTAVYYCAR 564
DYGGINDAFDIWGQGTMVTVSSGS
QVQLQESGPGLVKPSETLSLTCTVSGGSISSSSYYWGWIRQPPGKGLEWIG
BPx-64 SIYHSGSTYYNPSLKSRVTISVDTSKNQFSLKLSSVTAADTAVYYCAAADD 565
AFDIWGQGTMVTVSSGS QIQLVQSGAEVKKPGSSVKVSCKASGGTFSSYAISWVRQAPGQGLEWMG
BP6b-5 GIIPIFGTANYAQKFQGRVTITADKSTSTAYMELSSLRSEDTAVYYCARDQ 566
WLGGLSNYYYYGMDVWGQGTTVTVSSGS
QVQLVESGGGLVQPGRSLRLSCAASGFTFSSYGMHWVRQAPGKGLEWVA
BPx-65 VISYDGSNKYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCAKD 567
LLAGGSYYYYYYGMDVWGQGTTVTVSSGS
QIQLVQSGGGLVKPGGSLRLSCAASGFPFSNAWMSWVRQAPGKGLEWVA
BPx-66 NIEQDGSEKYYVDSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCAKE 568
EYGDHYYFDYWGQGTLVTVSSGS
EVQLLETRGGLVQPGRSLRLSCATSGFTFGDYAMHWVRQAPGKGLEWVS
BPx-67 AISGSGGSTYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCAKD 569
LATYYDFWSGLFDYWGQGTLVTVSSGS
EVQLVQSGGGLVQPGGSLRLSCAASGFTFDDYGMSWVRQAPGKGLEWV
BPx-68 AVISYDGSNKYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCSG 570
GSYYGAFDYWGQGTLVTVSSGS
QVQLVQSGAEVKKPGESLKISCKGSGYSFTSYWIGWVRQMPGKGLEWMG
BPx-69 IIYPGDSDTRYSPSFQGQVTISADKSISTAYLQWSSLKASDTAMYYCARKSS 571
SGWSIDYWGQGTLVTVSSGS
EVQLLESGGGLVQPGGSLRLSCAASGFTFSSYAMSWVRQAPGKGLEWVS
BPx-70 AISGSGGSTYYADSVKGRFTISRDNAKNSLYLQMNSLRAEDTAVYYCAGG 572
SYESSAYHYWGQGTLVTVSSGS
QVQLVQSGAEVKKPGASVKVSCKASGYTFTSYGISWVRQAPGQGLEWMG
BPx-71 WINTYNGNTNYAQKFQGRVTMTTDTSTSTTYMELRSLRSDDTAVYYCAR 573
EGWVGAKGGFDYWGQGTLVTVSSGS
EVQLVQSGGGVVQPGRSLRLSCAASGFTFSSYGMHWVRQAPGKGLEWVS
BPx-72 SISSTSSYIYYAESVKGRFTISRDNAENSLYLQMNSLRGEDTAVYYCARDR 574
GGYAYGDFDYWGQGTLVTVSSGS
QVQLVQSGAEVKKPGSSVKVSCKASGGTFSSYTISWVRQAPGQGLEWMG
BPx-73 RIIPILGIANYAQKFQGRVTITADKSTSTAYMELSSLRSEDTAVYYCARTNS 575
GTLDYWGQGTLVTVSSGS
QVQLVQSGAEVKKPGASVKVSCKASGYTFTSYDINWVRQATGQGLEWM
BPx-74 GWMNPNSGNTGYAQKFQGRVTMTRNTSISTAYMELSSLRSEDTAVYYCA 576
RGDFWSGYYYYYYGMDVWGQGTTVTVSSGS
QVQLQQSGPGLVKPSQTLSLTCAISGDSVSSNSVTWDWIRQSPSRGLEWLG
BP8b-l RTYYRSKWYNDYAVSVKSRITINPDTSKNQFSLHLSSVTPEDTAVYYCAR 577
QNGGYDYWGQGTLVTVSSGS
QVQLVQSGAEVKKPGASVKVSCKASGYTFTGYYMHWVRQAPGQGLEW
BPx-75 MGWINPNSGGTNYAQKFQGRVTITRDTSASTAYMELSSLRSEDTAVYYCA 578
RDGGVEM ATIPGGYGMD V WGQGTT VT VPS GS
QVQLVQSGAEVKKPGASVKVSCKASGYTFTSYGISWVRQAPGQGLEWMG
BPx-76 GIIPIFGTANYAQKFQGRVTITADKSTSTAYMELSSLRSEDTAVYYCARGRI 579
GSGYDSVWLRYFDYWGQGTLVTVSSGS
EVQLVQSGGGLVQPGRSLRLSCAASGFTFGDYAMHWVRQAPGKGLEWV
BPx-77 SGISWNSGSIGYADSVKGRVTISRDNAKNSLYLQMNSLRDEDTAVYYCAR 580
EDEGS GS Y YWGPVD YWGQGTLVT VS SGS
QVQLVESGGGLVQPGGSLRLSCAASGFTFSSYWMSWVRQAPGKGLEWVA
BP6b-6 NIKQDGSEKYYVDSVKGRFTISRDNAKNSLYLQMNSLRAEDTAVYYCAR 581
GYSNYAYWGQGTLVTVSSGS
QVQVVQSGAEVKKPGASVKVSCKASGYTFTGYYMHWVRQAPGQGLEW
BPx-78 MGWINPNSGGTNYAQKFQGRVTMTRDTSISTAYMELSRLRSDDTAVYYC 582
ARKS GTMVRGPMWGSFD YWGQGTLVT VS SGS
QIQLVQSGGGLVKPGGSLRLSCAASGFTFSDYYMSWIRQAPGKGLEWVSY
BPx-79 ISSSGSTIYYADSVKGRFTISRDNAKNSLYLQMNSLRAEDTAVYYCAKDPG 583
WLQLDYYFDYWGQGTLVTVSSGS QVQLVQSGAEVKKPGSSVKVSCKASGGTFSSYAISWVRQAPGQGLEWMG
BPx-80 GIIPIFGTADYAQKFQGRATITADESTSTAYMELSSLRSEDTAVYYCARDRY 584
CSGGSCYIYYGMDVWGQGTTVTVSSGS
QVQLVQSGGGVVQPGRSLRLSCAASGFTFSSYAMHWVRQAPGKGLEWV
BPx-81 AVISYDGSNKYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCAR 585
RTVGLGMDVWGQGTTVTVSSGS
EVQLLESRGGVVQPGRSLRLSCAASGFTFSSYGMHWVRQAPGKGLEWVA
BPx-82 VISYDGSNKYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCARG 586
YGSGSSWYFDYWGQGTLVTVSSGS
EVQLVQSGGGVVQPGRSLRLSCAASGFTFSSYGMHWVRQAPGKGLEWVA
BPx-83 VISYDGSNKYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCARA 587
LDLDYWGQGTLVTVSSGS
QMQLVQSGAEVKKPGSSVKVSCKASGGTFSSYAISWVRQAPGQGLEWMG
BPx-84 IINPSGGSTSYAQKFQGRVTMTRDTSTSTVYMELSSLRSEDTAVYYCARGV 588
VWRWLQSGGSGYFDYWGQGTLVTVSSGS
QVQLVQSGGGLVKPGTSLRLSCAASGFTFSSYAMHWVRQAPGKGLEWVA
BPx- VISYDGSNKYYADSVKGRFTISRDNSKNTLYLQMSSLRAEDTAVYYCVKA 589 119
RSGWYSSDAFDIWGQGTMVTVSSGS
EVQLVQSGGGLEQPGGSLRLSCATSGFTFTSHALSWIRQTPGKGLEWISSIS
BPx-
ANSDTIFYADSVKGRFTISRDNSTRMVYLQMNSLRAEDTAVYYCARDRYN 590 120
WNYGYFDYWGQGTLVTVSSGS
QIQLVQSGAEVKKPGASVKVSCKASGYTFTSYGISWVRQAPGQGLEWMG
BPx- WISAYNGNTNYAQNLQGRVTMTTNTSTNTAYMELSSLRSEDTAVYYCAK 591 121
VGFYDYVWGSYPYDAFDIWGQGTMVTISSGS
QVQLVQSGAEVKKPGASVKVSCKASGYTFTSYGISWVRQAPGQGLEWMG
BP6b- WISAYNGNTNYAQKLQGRVTMTTDTSTSTAYMELRSLRSEDTAVYYCAR 592 11
GY YGS GAMD VWGQGTT VT VS SGS
EVQLVQSGAEVKKPGESLKISCKGSGYSFTTYWIGWVRQMPGKGLEWMG
BP18b-
IIYPGDSDTRYSPSFQGQVTISADKSISTAYLQWSSLKASDTAMYYCARYS 593 4
YGYTFLDYWGQGTLVTVSSGS
QVQLQESGPGLVKPSETLSLTCTVSGGSISSSSYYWGWIRQPPGKGLEWIG
BP6b-2 SIYYSGSTYYNPSLKSRVTISVDTSKNQFSLLLTSMTAADTAVYYCARTGT 594
TRGDYFDNWGQGAKVTVSSGS
EVQLVQSGAEVKKPGASVKVSCKASGYTFTSYDINWVRQAPGQGLEWM
BP6b-
GIINPSGGSTSYAQKFQGRVTMTRDTSTSTVYMELSSLRSEDTAVYYCARA 595 10
FDSSGYSYGMDVWGQGTTVTVSSGS
QVQLVQSGAEVKKPGSSVKVSCKASGYTFTSYYMHWVRQAPGQGLEWM
BP 16b- GIINPSGGSTSYAQKFQGRVTMTRDTSTSTVYMELSSLRSEDTAVYYCARD 596 2
IDRATNWFDPWGQGTLVTVSSGS
QVQLVQSGGGLVQPGGSLRLSCAASGFTFSSYWMSWVRQAPGKGLEWV
BP18b- ANIKQDGSEKYYVDSVKGRFTISRDNAKNSLYLQMNSLRAEDTAVYYCA 597 5
RDKWWPEGYYYYYYGMD VWGQGTT VT VPS GS
QVQLVQSGAEVKKPGSSVKVSCKASGGTFSSYAISWVRQAPGQGLEWMG
BP7b-l GIIPIFGTANYAQKFQDRVTITADESTSTAYMELSSLRSEDTAVYYCARDG 598
YCSSTSCYVWGEFDPWGQGTLVTVSSGS
EVQLVQSGGGLVKPGGSLRLSCAASGFTFSSYGMHWVRQAPGKGLEWVS
BP5b-7 YISSGSSTLYYADSVKGRFTISRDNAKNSLYLQMNSLRAEDTAVYYCARD 599
FGWYGSFDYWGQGTTVTVSSGS
QVQLVQSGGGVVQPGRSLRLSCAASGFTFSSYALHWVRQAPGKGLEWVA
BPx-85 VLSYDGSNKYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCAR 600
A AHS GYDLNY Y YGMD AWGQGTT VT VS SGS
QVQLVQSGAEVKKPGESLKISCKGSGYSFTSYWIGWVRQMPGKGLEWMG
BP18b-
IIYPGDSDTRYSPSFQGQVTISADKSISTAYLQWSSLKASDTAMYYCAKLR 601 6
VGYNFFDYWGQGTLVTVSSGS EVQLVESRGGVVQPGRSLRLSCAASGFTFSSYAMHWVRQAPGKGLEWVA
BP20b- VISYDGSNKYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCVGV 602 1
DYYDSSGYPADYWGQGTLVTVSSGS
QVQLVQSGAEVKKPGESLKISCKGSGYSFTSYWIGWVRQMPGKGLEWMG
BPx-86 IIYPGDSDTRYSPSFQGQVTISVDKSINTAYLQWNSLKASDTAMYYCARG 603
WSSADYWGQGTLVTVSSGS
QVQLVESGGGVVQPGRSLRLSCAASGFTFSSYGMHWVRQAPGKGLEWVS
BP 19b-
VIYSGGSTYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCARD 604 3
WAFGGSYKLGDYWGQGTLVTVSSGS
QVQLVQSGGGLVQPGGSLRLSCAASGFTFSSYSMNWVRQAPGKGLEWVS
BPx-87 SISSSSSYIYYADSVKGRFTISRDNAKNSLYLQMNSLRAEDTAVYYCAKDR 605
FFT Y Y YDTS GRGGLDS WGQGTLVT VS SGS
EVQLVQSGGGLVQPGGSLRLSCAASGFTFSGSSMNWVRQAPGKGLEWVS
BPx-88 YISSSGNTIYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCAKGS 606
EEMRGWGQGTLVTVSSGS
QVQLVQSGAEVKKPGESLKISCKGSGYSFTSYWIGWVRQMPGKGLEWMG
BP18b-
IIYPGDSDTRYSPSFQGQVTISADKSISTAYLQWSSLKASDTAMYYCARHD 607 7
GNSRPGDYWGQGTLVTVSSGS
QVQLQQSGGGLVQPGGSLRLSCAASGFTFSSYAMSWVRQAPGKGLEWVS
BP 17b- AISGSGGSTYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCAKT 608 1
VAGTRLYYYYYGMDVWGQGTTVTVSSGS
EVQLVQSGAEVKKPGASVKVSCKVSGYTLTELSMHWVRQAPGKGLEWM
BP 16b- GGFDPEDGETIYAQKFQGRVTMTEDTSTDTAYMELSSLRSEDTAVYYCAT 609 3
DFYCSGGSCYSRGDAFDIWGQGTMVTVSSGS
QVQLVQSGGGLVQPGGSLRLSCAASGFTFSSYSMNWVRQAPGKGLEWVS
BPx-89 SISSSNSYIYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTALYYCARDW 610
SIYPASVDFFDYWGQGTLVTVSSGS
EVQLVESGGGLVKPGGSLRLSCAASGFTFSSYSMNWVRQAPGKGLEWVS
BPx-90 SISSSSSYIYYADSVKGRFTISRDNAKNSLYLQMNSLRAEDTALYYCAKDG 611
RLGELSHWGQGTLVTVSSGS
EVQLVESGGGVVQPGRSLRLSCAASGFTFSSYGMHWVRQAPGKGLEWVA
BP5b-8 VISYDGSNQYYADSVKGRFTISRDNSKNTLYVQMNSLRAEDTAVYYCAR 612
GNY YGS GS YPLLD YWGQGTL VT VS SGS
QVQLVQSGGGLVQPGGSLRLSCAASGFTFSNYWMYWVRQAPGKGLVWV
BP 16b- SNINSDGSSTSYADSVKGRFTISRDNAKNSLYLQMNSLRDEDTAVYYCAR 613 4
EGYNAHYFD YWGQGTL VTVSSGS
QVQLQQSGPRQVEPSQTLSLTCAISGDSVSSKSAAWNWIRQSPSRGLEWLG
BP9b-l RTYYRSKWYYDYAVSVKSRIIVNPDTSRNQFSLHLNSVTPEDTAFYYCAR 614
GEDSGYGSFD YWGQGTL VTVSSGS
QVQLVQSGAEVKKPGASVKVSCKASGYTFTSYGISWVRQAPGQGLEWMG
BPx-91 WISAYNGNTNYAQKLQGRVTMTTDTSTSTAYMELRSLRSDDTAVYYCAR 615
DMKWLPRGVD YWGQGTL VTVSSGS
QIQLVQSGAEVKKPGESLKISCKGSGYSFTSYWIGWVRQMPGKGLEWMGI
BPx-92 IYPGDSDTRYSPSFQGQVTISADKSISTAYLQWSSLKASDTAMYYCARLGF 616
REIDYWGQGTLVTVSPGS
EVQLLESRAEVKKPGESLKISCKGSGYSFTSYWIGWVRQMPGKGPEWMGI
BP 12b- IYPGDSDTRYSPSFQGQVTISADKSISTAYLQWSSLKASDTAMYYCARLVY 617 1
GGYDEPGYYFD YWGQGTL VTVSSGS
QVQLVQSGGGVVQPGRSLRLSCAASGFTFSSHGMHWVRQAPGKGPEWV
BPx-93 AVISYDGSNKYYPDSVRGRFTISRDNSKNTLYLQMNSLTTDDTAVYYCAK 618
GGGYGDYAPGYWGQGTLVTVSSGS
QVQLVQSGAEVKKPGSSVKVSCKASGGTFSSYAISWVRQAPGQGLEWMG
BPx-94 GFDPEDGETIYAQKFQGRVTMTEDTSTDTAYMELSSLRSEDTAVYYCATG 619
FWELEAGGFD YWGQGTL VTVSSGS QVQLQQSGPGLVKPSETLSLTCTVSGGSISSSSYYWGWIRQPPGKGLEWIG
BPx-95 SIYYSGSTYYNPSLKSRVTISVDTSRKQVSLELSSVTAADTAVYYCARLWL 620
GERPPDYWGQGMLVTVSSGS
QVQLVQSGAEVKKPGASVKVSCKASGYTFTSYYMHWVRQAPGQGLEWM
BPx-96 GIINPSGGSTSYAQKFQGRVTMTRDTSTSTVYMELSSLRSEDTAVYYCARD 621
RGSGWYGGAFDIWGQGTMVTVSSGS
QIQLVQSGGGVVQPGRSLRLSCAASGFTFSSYAIHWVRQAPGKGLEWVAV
BP6b-9 ISYDGSIKYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCARDT 622
HEWELFVGTFDIWGQGTMVTVSSGS
QVQLVQSGGGLVKPGGSLRLSCAASGFTFSSYSMNWVRQAPGKGLEWVS
BPx-97 SISSNSSYIYYADSVKGRFTISRDNAKNSLYLQMNSLRAEDTAVYYCARDP 623
DYYYGSGTREDYWGQGTLVTVSSGS
QVQLVESGGGVVQPGRSLRLSCAASGFTFSSYAMHWVRQAPGKGLEWVA
BPx-98 VISYDGSNKYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCARV 624
EWADGAFDIWGQGTMVTVSSGS
EVQLVESGGGLVQPGGSLRLSCAASGFTFSSYWMSWVRQAPGKGLEWVA
BPx-99 NIKQDGSEKYYVDSVKGRFTISRDNAKNSLYLQMNSLRAEDTAVYYCAR 625
GLIGDWGQGTLVTVSSGS
EVQLVQSGGGVVQSGRSLRLSCVASGFSFSNYGMHWVRQAPGKGLEWLA
BP6b-7 FIWYDGSNKWYADSVKGRFTISRDNSKNALYLQMNSLRAEDTAVYYCAR 626
DRYSSSWGGGFDYWGQGTVLTVSSGS
QIQLVQSGGGVVQPGRSLRLSCAASGFTFSNAWMSWVRQAPGKGLEWVG
BPx-
RIKSKTDGGTTDYAAPVKGRFTISRDDSKNTLYLQMNSLKTEDTAVYYCT 627 100
TDRVTMVRGVTPPFDYWGQGTLVTVSSGS
QLQLQQSGGGLVQPGGSLRLSCAASGFTFSSYWMHWVRQAPGKGLVWV
BP6b-8 SRINSDGSSTSYADSVKGRFTISRDNAKNTLYLQMNSLRAEDTAVYYCAR 628
DGWGSYRYVPGYYYYYGMDVWGQGTTVTVSSGS
QVQLVQSGAEVKKPGESLKISCKGSGYSFTSYWIGWVRQMPGKGLEWMG
BPx-
IIYPGDSDTRYSPSFQGQVTISADKSISTAYLQWSSLKASDTAMYYCARGS 629 101
GWTNILDYWGQGTLVTVSSGS
QVQLVQSGAEVKKPGESLKISCKGSGYSFTNYWIGWVRQMPGKGLEWM
BPx-
GIIYPGDSDTRYSPSFQGQVTISADKSISTAYLQWSSLKASDTAMYYCATQP 630 102
HDRGVWGQGTTVTVSSGS
QLQLQQFGPGLVKPSETLSLTCTVSGDSSSSYYWSWIRQPPGKGLEWIGSI
BPx-
YYSGSTYYNPSLKSRVTISVDTSKNQLSLKLNSVTAADTAVYYCARGAYG 631 103
DKDLGVFDYWGQGTLVTVSSGS
QVQLQESGPGLVKPSETLSLTCTVSGGSISSSSYYWGWIRQPPGKGLEWIG
BPx-
SIYYSGSTDYSPSLKSRVTISIDTSKNQFSLKVSSVTAADTAVYYCARERGN 632 104
GDYDNWGLGTLVTVSAGS
EVQLVQSGGGVVRPGGSLRLSCAASGFTFDDYGMSWVRQAPGKGLEWVS
BPx-
GINWNGGSTGYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCAR 633 105
QYVPEYNSGWPYYYYGMDVWGQGTTVTVSSGS
EVQLVESGPGLVKPSGTLSLTCAVFGGSISSSDNWWGWVRQPPGEGLEWI
BPx- GDISHSGNTIYNPSLKSRVTISVDKSKNQFSLKLGSVTAADTAVYYCARYD 634 106
SRGYYYPTDAFDIWGQGAMVTVSSGS
EVQLVESGGGLEKPGGSLRLSCAASGFTFSNYAMSWVRQAPGKGLEWVS
BP5b-9 AVNKGGDAAYSADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCAR 635
GVWAVDAFDIWGQGTMVTVSSGS
EVQLVQSGGGVVQPGRSLRLSCAASGFSFRNYAMHWVRQAPGKGLEWV
BPx- AVISYDGSNKYYADSVKGRFTISRDNAKNSLYLQMNSLRDEDTAVYYCA 636 107
RDNGYDSSGYYFLDHYWGQGTLVTVSSGS
QVTLKESGPTLVKPTQTLTLTCTFSGFSLSTYGVGVGWIRQPPGKAPEWLA
BPx- RIDWDDDEYYSTSLKTRLTISKDTSKNQVVLTMTDMDPVDTATYYCTRFV 637 108
GAKADYWGQGILVTVSSGS EVQLVESRGGVVQPGRSLRLSCAASGFTFSSYAMHWVRQAPGKGLEWVA
BPx- VISYDGSNKYYADSVKGRFTISRDNSKNTLYLQLNSLRAEDTAVYYCARA 638 109
RSPIAVAPNWFDPWGQGTLVTVSSGS
QIQLVQSGGGVVQPGRSLRLSCAASGFTFSSYGMHWVRQAPGKGLEWVA
BP22b- VIWYDGSNKYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCAK 639 1
GLGYHPLFDYWGQGTLVTVSSGS
QVQLVESGGGVVQPGRSLRLSCAASGFTFSSYGMHWVRQAPGKGLEWVA
BPx- VISYDGSNKYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCAKD 640 110
LYSGSGGGDYWGQGTLVTVSSGS
EVQLVQSGGGLVQPGGSLRLSCAGSGFTFSDYDMSWVRQAPGKGLEWVS
BP6b- GISGSGGSTYYVDSVQGRFTISRDNSKNTLYLQMNGLRAEDTAVYYCAKD 641 12
SSSGWYGLWDYWGQGTLVSVSSGS
QVQLQESGPGLVKPSETLSLTCTVSGGSISSYYWSWIRQPPGKGLEWIGYIY
BPx- YSGSTNYNPSLKSRVTMSVDTSKNQLSLNLTSVTAADTAVYYCAREFPMG 642 111
RPFDYWGQGTLVTVSSGS
EVQLVESGAEVKKPGASVKVSCKASGYTFTSYGISWVRQAPGQGLEWMG
BPx- WISAYNGNTNYAQKLQGRVTMTTDTSTSTAYMELRSLRSDDTAVYYCAR 643 112
DGMFAPGYFDYWGQGTLVTVSSGS
QVTLKESGPVLVKPTETLTLTCAVSGFSLSNPRMGVSWIRQPPGKALEWL
BPx- ALIYWDDDKRYSPSLKSRLTITKDTSKNQVVLTMTNMDPVDTATYYCAL 644 113
WDDYSNYEIARGAFDIWGQGTMVTVSSGS
QVQLVQSGGGLVQPGGSLRLSCAASGFTFSSYDMHWVRQATGKGLEWVS
BP 10b- AIGTAGDTYYPGSVKGRFTISRENAKNSLYLQMNSLRAGDTAVYYCARGK 645 1
PLLSGYLINDAFDIWGQGTMVTVSSGS
EVQLLETGGGVVQPGRSLRLSCAASGFSFNNYGMHWVRQAPGKGLEWVS
BPx- VIYSGGSTYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCARDG 646 114
GSGYHDYWGQGTLVTVSSGS
[00309] Recombinant GDF-modulating antibodies of the disclosure may comprise or be developed using any of the CDR-H sequences (CDR-Hl, CDR-H2 and/or CDR-H3) listed in Table 15. Some recombinant GDF-modulating antibodies of the disclosure may comprise or be developed using CDR-H sequences that comprise at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, at least about 99%, at least about 99.5% or at least about 99.9% sequence identity to any of the CDR-H sequences listed in Table 15. Recombinant GDF-modulating antibodies may, in some cases, comprise CDR-H domains presented, but with different combinations of CDRs from other clones listed. In some cases, such antibodies may interact with one or more of the recombinant proteins listed in Table 4, 9 or 10.
Table 15. CDR-H domains
CDR Clone ID CDR amino acid sequence SEQ Region ID
NO
CDR-Hl BP6a-16 GDSVSSNSAA 647
CDR-Hl BP6a-19 GGTFSSYA 648 CDR-H1 BP3a-3 GFTFDDYA 649
CDR-H1 BP18b-l GYSFTSYW 650
CDR-H1 BP19b-l GGSISSYY 651
CDR-H1 BP6b-4 GFTFSSYG 652
CDR-H1 BP6a-8 GYTFTSYY 653
CDR-H1 BP13a-l GYTLTELS 654
CDR-H1 BP5b-3 GFTFSDYY 655
CDR-H1 BP6a-23 GFTFSSYS 656
CDR-H1 BP6a-l l GFTFDDYT 657
CDR-H1 BP5b-4 GFTFSSYA 658
CDR-H1 BP6a-14 GFSFSSYG 659
CDR-H1 BP6a-13 GFRFSNYG 660
CDR-H1 BP4a-l GYTFSSYG 661
CDR-H1 BP6a-4 GDSITSYY 662
CDR-H1 BPx-1 GGAFSIYS 663
CDR-H1 BP21-a GGSISSSSYY 664
CDR-H1 BPx-3 GFSFSNYG 665
CDR-H1 BP6a-10 GFTFSSYW 666
CDR-H1 BP15a-l GYTFTSYG 667
CDR-H1 BP6a-26 GFSFSTYA 668
CDR-H1 BP5a-4 GGTLSSYA 669
CDR-H1 BP5b GFTFDDYG 670
CDR-H1 BP5a-3 GYSITNGYY 671
CDR-H1 BP14a-l GYNFNSYW 672
CDR-H1 BP6a-25 GDSVSSNTEA 673
CDR-H1 BP5a-2 GFTVSSYS 674
CDR-H1 BP6a-15 GVSIASSNW 675
CDR-H1 BP3a-2 GFTFSDYD 676
CDR-H1 BP6a-18 GFTFSSFA 677
CDR-H1 BP18a-2 GYSFTNYW 678
CDR-H1 BPx-9 GYTFNDYH 679
CDR-H1 BPx-14 GFTFGHYG 680
CDR-H1 BPx-17 GFTFGDYA 681
CDR-H1 BPx-18 GFSLSTSGVG 682
CDR-H1 BP5b-6 GYTFTGYY 683
CDR-H1 BPx-24 GFTFSDYG 684
CDR-H1 BPx-25 GGSTSSSSYY 685
CDR-H1 BPx-31 GYSFTAHW 686
CDR-H1 BPx-34 GFSLSTSGMG 687
CDR-H1 BPx-36 GYTFSDYY 688
CDR-H1 BPx-59 GFTFGTYA 689
CDR-H1 BPx-61 GYAFTSYG 690
CDR-H1 BPx-62 GFTFSNYD 691
CDR-H1 BPx-66 GFPFSNAW 692
CDR-H1 BPx-73 GGTFSSYT 693
CDR-H1 BPx-74 GYTFTSYD 694
CDR-H1 BP8b-l GDSVSSNSVT 695
CDR-H2 BP6a-16 TYYRSKWSN 696 CDR-H2 BP6a-19 IIPIFGTA 697
CDR-H2 BP3a-3 ISWNSGST 698
CDR-H2 BP18b-l IYPGDSDT 699
CDR-H2 BP19b-l IYYSGSTN 700
CDR-H2 BP6b-4 r YDGSNK 701
CDR-H2 BP6a-8 INPSGGST 702
CDR-H2 BP13a-l FDPEDDEA 703
CDR-H2 BP5b-3 ISSSGSTI 704
CDR-H2 BP6a-23 ISSSSSYI 705
CDR-H2 BP6a-l l ISWDGGST 706
CDR-H2 BP5b-4 ISGGGGAT 707
CDR-H2 BPl la-1 ISWNSGSI 708
CDR-H2 BP6a-14 TSYDGSNK 709
CDR-H2 BP6a-27 ISYDGSYE 710
CDR-H2 BP6a-13 ISGSGGLT 711
CDR-H2 BP4a-l SSAYNGNT 712
CDR-H2 BP19b-2 ISYDGSNK 713
CDR-H2 BP6a-4 IHYSGSTY 714
CDR-H2 BP19a-l ISNSGGST 715
CDR-H2 BP6b-3 INPNSGGT 716
CDR-H2 BPx-1 IIPIFETA 717
CDR-H2 BP21b-l IYYSGSTY 718
CDR-H2 BPx-2 ISSSSSTI 719
CDR-H2 BPx-3 ISGSGGSA 720
CDR-H2 BP6a-10 IKQDGSEK 721
CDR-H2 BP15a-l ISAYNGNT 722
CDR-H2 BP6a-26 ISYDGTIK 723
CDR-H2 BP5a-3 IFQSGSTY 724
CDR-H2 BP14a-l IYPGDSDIR 725
CDR-H2 BP3a-l ISGSGGST 726
CDR-H2 BP6a-25 TYYRSKWYH 727
CDR-H2 BP6a-15 IYHSGSTF 728
CDR-H2 BP6a-21 ISDSGGTT 729
CDR-H2 BP6a-18 ITSRGDTT 730
CDR-H2 BPx-11 INYSGNTY 731
CDR-H2 BPx-14 ISYDGTNK 732
CDR-H2 BPx-15 TYYRSKWYN 733
CDR-H2 BPx-17 INWNSGGV 734
CDR-H2 BPx-18 IYWDDDKR 735
CDR-H2 BPx-22 ITGSSSTI 736
CDR-H2 BPx-28 IHPGDGGT 737
CDR-H2 BPx-31 IYPADSTT 738
CDR-H2 BPx-34 IDWDDDKY 739
CDR-H2 BPx-36 ISPNSGGT 740
CDR-H2 BP16b-l INWNGGST 741
CDR-H2 BPx-59 ISGGGDST 742
CDR-H2 BPx-66 IEQDGSEK 743
CDR-H2 BPx-71 INTYNGNT 744 CDR-H2 BPx-72 ISSTSSYI 745
CDR-H2 BPx-73 IIPILGIA 746
CDR-H2 BPx-74 MNPNSGNT 747
CDR-H2 BPx-64 IYHSGSTY 748
CDR-H3 BP6a-16 ARGFSGMGV 749
CDR-H3 BP6a-19 ARDQAGDYGGNSGYFDY 750
CDR-H3 BP3a-3 AKGQVPNYYDSRDPFDP 751
CDR-H3 BP6a-l ARrfDDFDY 752
CDR-H3 BP18b-l ARGVAGYSSGWSD 753
CDR-H3 BP19b-l ARLRRGYFDY 754
CDR-H3 BP6b-4 ATRGY 755
CDR-H3 BP6a-8 AREGGDYNNWFDP 756
CDR-H3 BP13a-l ATDRVTGTTMAAFDI 757
CDR-H3 BP5b-3 ARDNWYYFDY 758
CDR-H3 BP6a-23 ARDLGGYFGNY 759
CDR-H3 BP5a-l ANHGLMDDSSGYYLSNAFDI 760
CDR-H3 BP6a-l l AAGSYHTW 761
CDR-H3 BP5b-4 ASPAGHICSGGSCQYYYYGMDV 762
CDR-H3 BP18a-l ARQPYYYGSGSGMDV 763
CDR-H3 BPl la-1 AKVDGLEYSSGHNFDY 764
CDR-H3 BP6a-14 AKDEYTYGSGIMDY 765
CDR-H3 BP6a-27 ARDSGARGYNYGLFDY 766
CDR-H3 BP6a-13 AREDLWRMGELLGGEGFDY 767
CDR-H3 BP4a-l ARGIYGDYEY 768
CDR-H3 BPla-1 AKDTSNGGYSSSSFDY 769
CDR-H3 BP6a-6 ARDRHLYRRGYSSVDGMDV 770
CDR-H3 BP19b-2 AKPRGRYYFDY 771
CDR-H3 BP6a-4 ARGPSRGLFDP 772
CDR-H3 BP19a-l ASLSRGY 773
CDR-H3 BP6a-7 ARESLIRFLEDPQQGGMDV 774
CDR-H3 BP5b-5 ATPYSSSSGTDY 775
CDR-H3 BP6b-3 ARPRTLRGDAFDI 776
CDR-H3 BP18b-2 ARHPIGSGYDY 777
CDR-H3 BPx-1 ARAGENSDSNYAFDI 778
CDR-H3 BP21b-l ARGRYSYGYFDY 779
CDR-H3 BP6a-2 ARGIRVVGDEDYYYYYGMDV 780
CDR-H3 BPx-2 ARDSSGWLGMGDFDY 781
CDR-H3 BP6b-l AREEYYGGNTD 782
CDR-H3 BPx-3 ARDISHYDILTGQLDY 783
CDR-H3 BP6a-5 ARDLLVRFLEWSHYYGMDV 784
CDR-H3 BP6a-10 AKETDCSGGICYGVDY 785
CDR-H3 BP15a-l ARDAPYDILTGYLSY 786
CDR-H3 BP6a-26 ARAGGAGSDGYNYWGY 787
CDR-H3 BP6a-12 ARDPGGKLGGAFDI 788
CDR-H3 BP5a-4 ARDRPGNNRYDILTGGGWFDP 789
CDR-H3 BP5b-l AKEIS DTAMGFD Y 790
CDR-H3 BPx-4 ARAYIPRLYYYGSGLDY 791
CDR-H3 BP5a-3 ARRVDYYDSSGYGEGYFDY 792 CDR-H3 BP14a-l ARHVSGSLDY 793
CDR-H3 BP3a-l AKTAYYDSSGYYPDAFDI 794
CDR-H3 BP18b-3 ARGQGGVLGAFDI 795
CDR-H3 BP6a-9 ARQGRGEAFDI 796
CDR-H3 BP6a-25 ARTVFDSQVYWFDP 797
CDR-H3 BP5a-2 AKVGTAAAGDAFDI 798
CDR-H3 BP6a-15 ASSSGWPYPTNWFDA 799
CDR-H3 BP6a-21 AKDAHNWNYGVFDY 800
CDR-H3 BP5a-5 ASALGATGAFDI 801
CDR-H3 BP6a-17 AKDISGYEPLYYFDY 802
CDR-H3 BP6a-20 ARDPGKQWRVRGYFDY 803
CDR-H3 BP3a-2 ARESDMAAAGSFDY 804
CDR-H3 BP6a-24 AREGNSGYDV 805
CDR-H3 BP6a-22 ARDSLWFGELSALDY 806
CDR-H3 BP19a-2 ARDVSSWAAF 807
CDR-H3 BPx-6 ARGGRGGVITWFDP 808
CDR-H3 BP6a-18 ARDRPQVPYDSSGNFDY 809
CDR-H3 BP16a-l AREEAGYSSGWVEDAFDI 810
CDR-H3 BP19a-3 ARMVGGYRFDY 811
CDR-H3 BP5b-2 AKHGDYGGTFDY 812
CDR-H3 BP2a-l ARVGFYDYVWGSYPYDAFDI 813
CDR-H3 BP18a-2 ARRSSSSGSFDY 814
CDR-H3 BP6a-3 VRDRHSLGDFDY 815
CDR-H3 BPx-7 ARRFTMDY 816
CDR-H3 BPx-8 ARYSSSWELFDY 817
CDR-H3 BPx-9 ARRNGSGSAPGY 818
CDR-H3 BPx-10 ARVGQYYGMDV 819
CDR-H3 BPx-11 ARRVLGYGYFDY 820
CDR-H3 BPx-12 TGGGNVDY 821
CDR-H3 BPx-13 AKDKAGNYYYGMDV 822
CDR-H3 BPx-14 AKGSRLDP 823
CDR-H3 BPx-15 VGQGYDSSGYYGMDV 824
CDR-H3 BPx-16 ARQYYDTSYFDY 825
CDR-H3 BPx-17 AKDSGYSYGINFDY 826
CDR-H3 BPlb-1 ARGDWFDP 827
CDR-H3 BPx-18 AHMRWGYYFDY 828
CDR-H3 BPx-19 ARREMGAKGSGGIDY 829
CDR-H3 BPx-20 AREGWGS LLD Y 830
CDR-H3 BPx-21 AKDGDLGRYFDWLPLYYYYGMDV 831
CDR-H3 BPx-22 ARGGARADY 832
CDR-H3 BPx-23 ATPGPYYYYGMDV 833
CDR-H3 BP5b-6 ARGIGDY 834
CDR-H3 BPx-24 AKDAGRWLEIPYYFDF 835
CDR-H3 BPx-25 ARVVWFGEPLFDP 836
CDR-H3 BPx-26 AKDLSGYDQAIDY 837
CDR-H3 BPx-27 VRSGSGWAIDY 838
CDR-H3 BPx-28 ARGDLDN 839
CDR-H3 BPx-29 ARRYSSGWGFDY 840 CDR-H3 BPx-30 AHGEMDLFGYSSGWFPDFDY 841
CDR-H3 BPx-31 VRQTSGWSD 842
CDR-H3 BPx-32 ARGDGYNHPLYY 843
CDR-H3 BPx-33 ARGTLWFGELSVDY 844
CDR-H3 BPx-34 ARMYSYGATEFDY 845
CDR-H3 BPx-35 AKDISSGWYSVGYFDL 846
CDR-H3 BPx-36 ARGFFDY 847
CDR-H3 BPx-37 ARGQTSYFYDSSGPEIPNFDY 848
CDR-H3 BPx-38 ARGGGYYDSSGYPYYYYGMDV 849
CDR-H3 BPx-39 ARSTGYSSSSD 850
CDR-H3 BPx-40 ARDVGASGLLWFGELSFDY 851
CDR-H3 BPx-41 ARDRPHAGLYYYYYGMDV 852
CDR-H3 BPx-42 AKDLLYYYDSSGPVDY 853
CDR-H3 BPx-43 VDSSGYFDAFDI 854
CDR-H3 BP16b-l AREDYGDSEYYFDY 855
CDR-H3 BPx-44 ARKGGYYGSGSYFDY 856
CDR-H3 BPx-45 AVEEDYGGSFDY 857
CDR-H3 BPx-46 ARSRSGSYYGTDAFDI 858
CDR-H3 BPx-47 ARVDFYDYVWGSYLLDY 859
CDR-H3 BPx-48 ARDLVYSSGGPDY 860
CDR-H3 BPx-49 ARRWDYDSSGYQNDY 861
CDR-H3 BPx-50 ARDQWIQLWIDY 862
CDR-H3 BPx-51 AAEVPAAKLYYYYYGMDA 863
CDR-H3 BPx-52 ARDHLYYYDSSGYYDY 864
CDR-H3 BPx-53 ATYYDSSGYYPYYFDY 865
CDR-H3 BPx-54 ARDFKIAARGMDV 866
CDR-H3 BPx-55 ARGPHYFSDYGDYDSIGYYYYGMDV 867
CDR-H3 BPx-56 ARDKGSSGYYNFDY 868
CDR-H3 BPx-57 AREVLGDYYYGMDV 869
CDR-H3 BPx-58 ARYYYGSGSYFDLLDY 870
CDR-H3 BPx-59 AKATREDVQHGSFDY 871
CDR-H3 BPx-60 ARGVLGGGFDY 872
CDR-H3 BPx-61 ARDGGVLGDY 873
CDR-H3 BPx-62 ARRTPFDY 874
CDR-H3 BPx-63 ARDYGGINDAFDI 875
CDR-H3 BP6b-5 ARDQWLGGLSNYYYYGMDV 876
CDR-H3 BPx-65 AKDLLAGGSYYYYYYGMDV 877
CDR-H3 BPx-66 AKEEYGDHYYFDY 878
CDR-H3 BPx-67 AKDLATYYDFWSGLFDY 879
CDR-H3 BPx-68 SGGSYYGAFDY 880
CDR-H3 BPx-69 ARKSSSGWSIDY 881
CDR-H3 BPx-70 AGGSYESSAYHY 882
CDR-H3 BPx-71 AREGWVGAKGGFDY 883
CDR-H3 BPx-72 ARDRGGYAYGDFDY 884
CDR-H3 BPx-73 ARTNSGTLDY 885
CDR-H3 BPx-74 ARGDFWSGYYYYYYGMDV 886
CDR-H3 BP8b-l ARQNGGYDY 887
CDR-H3 BPx-75 ARDGGVEMATIPGGYGMDV 888 CDR-H3 BPx-76 ARGRIGSGYDSVWLRYFDY 889
CDR-H3 BPx-77 AREDEGSGSYYWGPVDY 890
CDR-H3 BP6b-6 ARGYSNYAY 891
CDR-H3 BPx-78 ARKSGTMVRGPMWGSFDY 892
CDR-H3 BPx-79 AKDPGWLQLDYYFDY 893
CDR-H3 BPx-80 ARDRYCSGGSCYIYYGMDV 894
CDR-H3 BPx-81 ARRTVGLGMDV 895
CDR-H3 BPx-82 ARGYGSGSSWYFDY 896
CDR-H3 BPx-83 ARALDLDY 897
CDR-H3 BPx-84 ARGVVWRWLQSGGSGYFDY 898
CDR-H3 BPx-64 AAADDAFDI 899
CDR-H1 BP20b-l GFTFSSYA 900
CDR-H1 BPx-89 GFTFSSYS 901
CDR-H1 BPx-105 GFTFDDYG 902
CDR-H1 BPx-99 GFTFSSYW 903
CDR-H1 BPx-93 GFTFSSHG 904
CDR-H1 BPx-92 GYSFTSYW 905
CDR-H1 BP6b-7 GFSFSNYG 906
CDR-H1 BPx-100 GFTFSNAW 907
CDR-H1 BPx-95 GGSISSSSYY 908
CDR-H1 BPx-111 GGSISSYY 909
CDR-H1 BP6b-l l GYTFTSYG 910
CDR-H1 BP18b-4 GYSFTTYW 911
CDR-H1 BP6b-10 GYTFTSYD 912
CDR-H1 BP16b-2 GYTFTSYY 913
CDR-H1 BP7b-l GGTFSSYA 914
CDR-H1 BP5b-7 GFTFSSYG 915
CDR-H1 BPx-88 GFTFSGSS 916
CDR-H1 BP16b-3 GYTLTELS 917
CDR-H1 BP16b-4 GFTFSNYW 918
CDR-H1 BP9b-l GDSVSSKSAA 919
CDR-H1 BPlOb-1 GFTFSSYD 920
CDR-H1 BPx-102 GYSFTNYW 921
CDR-H1 BPx-103 GDSSSSYY 922
CDR-H1 BPx-106 GGSISSSDNW 923
CDR-H1 BP5b-9 GFTFSNYA 924
CDR-H1 BPx-107 GFSFRNYA 925
CDR-H1 BPx-108 GFSLSTYGVG 926
CDR-H1 BP6b-12 GFTFSDYD 927
CDR-H1 BPx-114 GFSFNNYG 928
CDR-H1 BPx-113 GFSLSNPRMG 929
CDR-H2 BP20b-l ISYDGSNK 930
CDR-H2 BPx-89 ISSSNSYI 931
CDR-H2 BPx-105 INWNGGST 932
CDR-H2 BPx-99 IKQDGSEK 933
CDR-H2 BPx-92 IYPGDSDT 934
CDR-H2 BP6b-7 rWYDGSNK 935
CDR-H2 BPx-100 IKSKTDGGTT 936 CDR-H2 BPx-95 IYYSGST 937
CDR-H2 BP6b-l l ISAYNGNT 938
CDR-H2 BP6b-10 INPSGGST 939
CDR-H2 BP7b-l IIPIFGTA 940
CDR-H2 BP5b-7 ISSGSSTL 941
CDR-H2 BPx-85 LSYDGSNK 942
CDR-H2 BP19b-3 IYSGGST 943
CDR-H2 BPx-87 ISSSSSYI 944
CDR-H2 BPx-88 ISSSGNTI 945
CDR-H2 BP16b-3 FDPEDGET 946
CDR-H2 BP5b-8 ISYDGSNQ 947
CDR-H2 BP16b-4 INSDGSST 948
CDR-H2 BP9b-l TYYRSKWYY 949
CDR-H2 BPlOb-1 IGTAGDT 950
CDR-H2 BP6b-9 ISYDGSIK 951
CDR-H2 BPx-97 ISSNSSYI 952
CDR-H2 BPx-106 ISHSGNTI 953
CDR-H2 BP5b-9 VNKGGDAA 954
CDR-H2 BPx-108 IDWDDDE 955
CDR-H2 BPx-113 IYWDDDK 956
CDR-H3 BP20b-l VGVDYYDSSGYPADY 957
CDR-H3 BPx-89 ARDWSIYPASVDFFDY 958
CDR-H3 BPx-105 ARQYVPEYNSGWPYYYYGMDV 959
CDR-H3 BPx-99 ARGLIGD 960
CDR-H3 BPx-93 AKGGGYGDYAPGY 961
CDR-H3 BPx-92 ARLGFREIDY 962
CDR-H3 BP6b-7 ARDRYSSSWGGGFDY 963
CDR-H3 BPx-100 TTDRVTMVRGVTPPFDY 964
CDR-H3 BPx-95 ARLWLGERPPDY 965
CDR-H3 BPx-111 AREFPMGRPFDY 966
CDR-H3 BP6b-2 ARTGTTRGDYFDN 967
CDR-H3 BP6b-l l ARGYYGSGAMDV 968
CDR-H3 BP18b-4 ARYSYGYTFLDY 969
CDR-H3 BP6b-10 ARAFDSSGYSYGMDV 970
CDR-H3 BP16b-2 ARDIDRATNWFDP 971
CDR-H3 BP18b-5 ARDKWWPEGYYYYYYGMDV 972
CDR-H3 BP7b-l ARDGYCSSTSCYVWGEFDP 973
CDR-H3 BP5b-7 ARDFGWYGSFDY 974
CDR-H3 BPx-85 ARAAHSGYDLNYYYGMDA 975
CDR-H3 BP18b-6 AKLRVGYNFFDY 976
CDR-H3 BPx-86 ARGWSSADY 977
CDR-H3 BP19b-3 ARDWAFGGSYKLGDY 978
CDR-H3 BPx-87 AKDRFFTYYYDTSGRGGLDS 979
CDR-H3 BPx-88 AKGSEEMRG 980
CDR-H3 BP18b-7 ARHDGNSRPGDY 981
CDR-H3 BP17b-l AKTVAGTRLYYYYYGMDV 982
CDR-H3 BP16b-3 ATDFYCSGGSCYSRGDAFDI 983
CDR-H3 BPx-90 AKDGRLGELSH 984 CDR-H3 BP5b-8 ARGNYYGSGSYPLLDY 985
CDR-H3 BP16b-4 AREGYNAHYFDY 986
CDR-H3 BP9b-l ARGEDSGYGSFDY 987
CDR-H3 BPx-91 ARDMKWLPRGVDY 988
CDR-H3 BP12b-l ARLVYGGYDEPGYYFDY 989
CDR-H3 BPlOb-1 ARGKPLLSGYLINDAFDI 990
CDR-H3 BPx-94 ATGFWELEAGGFDY 991
CDR-H3 BPx-96 ARDRGSGWYGGAFDI 992
CDR-H3 BP6b-9 ARDTHEWELFVGTFDI 993
CDR-H3 BPx-97 ARDPDYYYGSGTREDY 994
CDR-H3 BPx-98 ARVEWADGAFDI 995
CDR-H3 BP6b-8 ARDGWGSYRYVPGYYYYYGMDV 996
CDR-H3 BPx-101 ARGSGWTNILDY 997
CDR-H3 BPx-102 ATQPHDRGV 998
CDR-H3 BPx-103 ARGAYGDKDLGVFDY 999
CDR-H3 BPx-104 ARERGNGDYDN 1000
CDR-H3 BPx-106 ARYDSRGYYYPTDAFDI 1001
CDR-H3 BP5b-9 ARGVWAVDAFDI 1002
CDR-H3 BPx-107 ARDNGYDSSGYYFLDHY 1003
CDR-H3 BPx-108 TRFVGAKADY 1004
CDR-H3 BPx-109 ARARSPIAVAPNWFDP 1005
CDR-H3 BP22b-l AKGLGYHPLFDY 1006
CDR-H3 BPx-110 AKDLYSGSGGGDY 1007
CDR-H3 BP6b-12 AKDSSSGWYGLWDY 1008
CDR-H3 BPx-112 ARDGMFAPGYFDY 1009
CDR-H3 BPx-114 ARDGGSGYHDY 1010
CDR-H3 BPx-113 ALWDDYSNYEIARGAFDI 1011
[00310] Recombinant GDF-modulating antibodies of the disclosure may comprise or be developed using any of the VL sequences listed in Table 16. Some recombinant GDF- modulating antibodies of the disclosure may comprise or be developed using VL sequences that comprise at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, at least about 99%, at least about 99.5% or at least about 99.9% sequence identity to any of the VL sequences listed in Table 16. Recombinant GDF-modulating antibodies may, in some cases, comprise VL domains presented, but with different combinations of CDRs (e.g. CDR-L1, CDR-L2 or CDR-L3). In some cases, such antibodies may interact with one or more of the recombinant proteins listed in Table 4, 9 or 10.
Table 16. VL domains
Clone VL amino acid sequence SEQ
ID ID NO
BP6a- SSELTQDPAVSVALGQTASITCSGDKLGDKYASWYQQKPGQSPVM 1012 16 VIHQDSKRPSGIPERFSGSNSGNTATLTISGTQAMDEADYYCQAWDS
STVVFGGGTKLTVLGQPKAAPSVTLFPPSS
BP6a- DIQMTQSPSTLSASAGDRVTITCRASQSISRWLAWYQQKPGKPPKLL 1013 19 IYGASSLQSGVPSRFSGSGSGTDFTLTISSLQPEDFATYYCQQTYTTP
WTFGQGTKVEIKRTVAAPSVF
BP3a- EIVLTQSPGTLSLSPGERATLSCRASQSVSSYLAWYQQKPGQAPRLLI 1014 3 YGASNRATGIPARFSGSGSGTDFTLTISSLQAEDVAVYYCQQYYTTP
FTFGPGTKVDIKRTVAAPSVF
BP6a- DVVMTQTPLSLPVTLGQPASISCRSSQSLVYSDGNTYLNWFQQRPG 1015 1 QSPRRLIYKVSNRDSGVPDRFSGSGSGTDFTLKISRVEAEDVGVYYC
MQSTRWPWVFGQGTKVEIKRTVAAPSVF
BP 18b DIVMTQSPLSLPVTPGEPASISCRSSQTLLHSNGFNYLDWYLQKPGQ 1016 -1 SPQLLMYLGSKRASGVPDRFSGSGSGTDFTLKISRVEAEDVGVYYC
MQALQTPFTFGPGTKVDIKRTVAAPSVF
BP 19b QSALTQPASVSGSPGQSITISCTGTSSDVGGYNYVSWYQQHPGKAP 1017 -1 KLMIYDVSKRPSGVSNRFSGSKSGNTASLTISGLQAEDEADYYCSSY
TS S STF V VFGGGTKLT VLGQPKA APS VTLFPPS S
BP6b- QSALTQPPSASGSPGQSVTISCSGTSSDIGGYDYVSWYQQHPGKAPK 1018 4 LMIYDVSERPSGVSNRFSGSKSGNTASLTISGLQAEDEADYYCSSFTS
SSTLVFGGGTKLTVLRQPKAAPSVTLFPPSS
BP6a- QSVLTQPPSVSGAPGQKVTISCSGSSSNIGNNYVSWYQQRPGTAPKL 1019 8 LIYDNNKRPSGIPDRFSGSKSGTSATLGITGLQTGDEADYYCGTWDS
SLSAVFGGGTKLTVLRQPKAAPSVTLFPPSS
BP13a SSELTQDPAVSVALGQTVKITCQGDSLRSYFASWYQQKPGQAPVLV 1020 -1 LFGKNSRPSGIPGRFSGSGSGNTAVLTITGAQAEDEADYYCDSRHSS
GN Y VIFGGGTKVTVLS QPKA APS VTLFPPS S
BP5b- QSALTQPPSASGSPGQSVTISCTGTSSDVGGYNYVSWYQQHPGKAP 1021 3 KLMIYEVSKRPSGVPDRFSGSKSGNTASLTVSGLQAEDEADYYCSS
YAGSNSWVFGGGTKLTVLGQPKAAPSVTLFPPSS
BP6a- QPVLTQPPSVSLSPGQTASITCSGDDLGDKFASWYQQKPGQSPILVIY 1022 23 QDSKRPSGIPERFSGSNSGNTATLTISGTQAMDEADYYCQAWDGTK
VVFGGGTKLTVLGQPKAAPSVTLFPPSS
BP5a- QPVLTQPPSASGTPGQRVTISCSGSSSNIGSNTVEWYQQLPGTAPKLL 1023 1 IHSNNQRPSGVPDRFSGSRSGTSASLAISGLQSEDEADYFCATWDDS
LTGVVFGGGTTLTVLGQPKAAPSVTLFPPSS
BP6a- QPVLTQPPSASGTPGQRVTISCSGSSSNIGSSTVNWYQQLPGTAPRLL 1024 11 IYSINQRPSGVPDRFSGSKSGTSASLAISGLQSDDEADYYCAAWDDS
LNGWVFGGGTKLT VLRQPKANPTVTLFPPS S
BP5b- DIQMTQSPSFLSASVGDRVTITCRASQGISSYLAWYQQKPGKAPKLL 1025 4 IYAASSLQSGVPSRFSGSGSGTDFTLTISSLQPEDFATYYCQQSYSTPL
TFGGGTRVDMARTVAAPSVF
BP18a EIVMTQSPLSLPVTLGQPASISCGSSQSLVYSDGNTYVNWFQQRPGQ 1026 -1 SPRRLIYKVSNRDSGVPDRFSGSGSGTDFTLKISRVEAEDVGVYYCM
QGTHWPYTFGQGTKLEIKRTVAAPSVF
BPl la QSALTQPPSVSGSPGQSVTISCTGSSSDVGYYDHVSWYQHHPGRAP 1027
-1 KVIIYDVTKRPSGVPDRFSGSKSGNTASLTISGLQAEDEADYYCSSY
AGSYTWVFGGGTELTVLGQPKAAPSVTLFPPSS BP6a- EIVLTQSPGTLSLSPGEGATLSCRASQTVNSNDLAWYQQRPGQAPRL 1028 14 LIYDASTRATGIPDRFSGSGSGTDFTLTISRLEPEDFAVYFCQQYHGS
QTFGQGTRVEIKRTVAAPSVF
BP6a- DIQMTQSPSSLSASVGDRVTITCRASQSINRHLNWYQQKPGKAPKFL 1029 27 ISAASNLQSGVPSRFSGSGSGTDFTLTISSLQPEDFATYYCQQSYSTP
YTFGGGTKVEIKRTVAAPSVF
BP6a- DIQMTQSPSTLSASVGDRVTITCRASQSISSWLAWYQQKPGKAPKLL 1030 13 IYKASSLGSGVPSRFSGSGSGTDFTLTISSLQPEDFATYYCQQSYSTP
WTFGQGTKVEIKRTVAAPSVF
BP4a- QSALTQPASVSGSPGQSITISCTGTSSDISIYNYVSWFQQHPGKAPKL 1031 1 MIYDVNNRPSGVSSRFSGSKSANTASLTISGLQAEDEADYYCSSYTY
SNTLLFGGGTKLTVLGQPKAAPSVTLFPPSS
BPla- QSALTQPASVSGSPGQSITISCTGTSSDVGGYNYVSWYQQHPGTAPK 1032 1 LMIYDVSYRPSGVSNRFSGSKSGNTASLTISGLQAEDEADYYCSSYT
S S STLVFGTGTKVTVLGQPKANPT VTLFPPS S
BP6a- EIVMTQSPGTLSLSPGERATLSCRASQSVSSGYLAWYQQKAGQAPR 1033 6 LLIYGGSARAPGIPARFSGSGSGTDFTLTISSLEPEDFAVYYCQQRSI
WPLTFGGGTKVEIKRTVAAPSVF
BP 19b QSALTQPPSASGSRGQSVSISCSGSRSDIGYYNYVSWYQQHPGKAPK 1034 -2 LIIFDVNKRPSGVPDRFSGSKSGNTASLTVSGLQAEDVADYYCSSSV
GSNNLVFGGGTKLTVLGQPKAAPSVTLFPPSS
BP6a- QSALTQPASVSGSPGQSITLSCTGTSSDVGDYNYVSWYQQYPGKAP 1035 4 KLMIYDVTKRPSGVSNRFSGSKSGNTASLTISGLQAEDEADYYCSSY
TSSTTWVFGGGTKLTVLGQPKAAPSVTLFPPSS
BP19a QSVLTQPPSASGTPGQRVSISCSGRMSNIGKNTVNWYQQVPGTAPK 1036 -1 LLIYSNNQRPSGVPDRFSGSKSGTSASLAISGLQSEDEADYYCAAWD
DSLNGWVFGGGTKLTVLGQPKAAPSVTLFPPSS
BP6a- QSALTQPASVSGSPGQSLTISCTGTSSDIGGYNYVSWYQQHPGKAPK 1037 7 LIIYDVTDRPSGVSGRFSGSKSGNTASLTISGLQTEDEAEYFCNSWTR
SNNYIFGGGTKLTVLGQPKAAPSVTLFPPSS
BP5b- DVVMTQSPLSLPVTLGQPASISCRSSQSLVYSDGNTYLNWFQQRPG 1038 5 QSPRRLIYKVSNRDSGVPDRFSGSGSGTDFTLKISRVEAEDVGVYYC
MQGTHWPLTFGQGTKVEITRTVVAPSVF
BP6b- SSELTQAPSVSVSPGQTASITCSGNKLGDKFASWYQQKPGQSPVMV 1039 3 MYEDRKRPSGIPERFSGSNSGNTATLTISGTQALDEADYYCQTWDS
RTVVFGGGTKLTVLGQPKAAPSVTLFPPSS
BP 18b DIVMTQSPVSLPVTLGQPASISCRSSQSVVYRDGNTYLSWFLQRPGQ 1040
-2 SPRRLIYKVSNRDSGVPDRFSGSGSATDFTLKISRVEAEDVGVYYCM
QGSSWPYTFGQGTKLEIKRTVAAPSVF
BPx-1 DIVMTQSPLSLPVTPGEPASISCRSSQSLLHSDGNNYLDWYLQKPGQ 1041
SPQLLIYLGSNRASGVPDRFSGSGSGTDFTLKISRVEAEDVGVYYCM QGLHTPTTFGQGTKVEIKRTVAAPSVF
BP21b NFMLTQPHSVSESPGKTVTISCTRSSGSIASNYVQWYQQRPGSSPTT 1042 -1 VIYEDNQRPSGVPDRFSGSIDSSSNSASLTISGLRTEDEADYYCQSYD
HMYNWVIGGGTKLTVLGQPKAAPSVTLFPPSS
BP6a- QSVLTQPPSVSGAPGQKVTISCSGSSSNIGNNYVSWYQQLPGTAPKL 1043 2 LIYDNNQRPSGVPDRFSGSKSGTSASLAISGLRSEDEADYHCAAWD
DDLNGPVFGGGTKLTVLGQPKAAPSVTLFPPSS
BPx-2 DIQMTQSPSSLSASVGDRVTISCRASHSISRFLNWYQQKPGRAPKLLI 1044 YSTSSLQSGVPSRFSGSGSARDFTLTISGLQPEDFATYFCQQSYSTPW
TFGQGTKVELKRTVAAPSVF
BP6b- SSELTQPPSVSVSPGQTATITCSGDKLGDKYASWYQQKPGQSPVLVI 1045 1 YQDTKRPSGIPARFSGSNSGNTATLTISGTQAMDEAAYYCQAWDST
TVVFGGGTKLTVLGQPKAAPSVTLFPPSS
BPx-3 DIQMTQSPSSLSASVGDRVTITCRASQSISSYLNWYQQKPGKAPKLLI 1046
YAASSLQSGVPSRFSGSGYETDFTLTISSLQAEDVAVYYCQQYYSSP
WTFGQGTRVEIKRTVAAPSVF
BP6a- QPVLTQPPSASGTPGQRVTISCSGSSSNIGSNTVHWYQQLPGTAPKL 1047 5 LIYSDNQRPSGVPDRFSGSKSGTSASLVISGLQSDDEADYYCAAWD
DSLNGVFGGGTKLTVLgQPKAAPSVTLFSPSS
BP6a- QPVLTQPPSVSVSPGQTASrrCSGDKLGDKYASWYQQKPGQSPVMV 1048 10 IYQDRKRPSGIPERFSGSNSGNTATLTISGTQAMDEADYYCAAWDD
SLSGRVFGGGTKLTVLGQPKAAPSVTLFPPSS
BP15a EIVMTQSPGTLSLSPGERATLSCRASQSVSSSYLAWYQQKPGQAPRL 1049 -1 LIYGASSRATGIPDRFSGSGSGTDFTLTISRLEPEDFAVYYCQQYGSS
PYTFGQGTKLEIKRTVAAPSVF
BP6a- EIVLTQSPATLSLSPGDRATLSCRASQSVSSYLAWYQQKPGQAPRLL 1050 26 IYDASNRATGIPARFSGSGSGTDFSLTISRLEPEDFAVYYCQQSGSSP
WTFGQGTKVEIKRTVAAPSVF
BP6a- DIVMTQSPATLSLSPGERATLSCRASQSVSSYLAWYQQKPGQAPRLF 1051 12 IYGASTRATGIPDRFSGSGSGTDFTLTISSLQAEDVAVYYCQQYYSTP
YTFGQGTKLEIKRTVAAPSVF
BP5a- QSVLTQPPSVSAAPGQKVTISCSGGSSNIGNNYLSWYQQLPGTAPKL 1052 4 LIYDNNKRPSGIPDRFSGSKSGTSATLGITGLQTGDEADYYCGTWDS
SLSAGVFGGGTKLTVLGQPKAAPSVTLFPPSS
BP5b- QSALTQPASVSGSPGQSITISCTGTSSDVGGYNYISWYQQYPGKAPK 1053 1 LMIYDVSKRPSGVSNRFSGSKSGNTASLTISGLQAEDEADYYCNSYT
SSRTVVFGGGTKLTVLGQPKAAPSVTLFPPSS
BPx-4 SSELTQPPSVSVSPGQTASrrCSGDQLGDKFVSWYQQKPGRSPVLVIS 1054
EDNKRHSGTPERISGSNSGNTATLTISGTQSMDEADYFCQTWDSSA
VVFGGGTKLTVLGQPKAAPSVTLFPPSS
BP5a- SSELTQDPAVSVALGQTVRITCQGDSLKKYYASWYQQKPGQAPVL 1055 3 VIYGKNLRPSGIPERFSGSSSGDTASLTITGTQAEDEADYYCQAWDS
STAVFGGGTKLTVLGQPKAAPSVTLFPPSS
BP14a DVVMTQSPLSLSVTPGQPASISCRSSQSLVHSDGNTYLNWFHQRPG 1056 -1 QSPRRLIYKVSNRDPGVPDRFSGSGSGTDFTLKISRVEAEDAGVYYC
MQGTHWPRLTFGGGTKVEIKRTVAAPSVF
BP3a- DIQMTQSPSSFSASTGDRVTITCRASQGISSYLAWYQQKPGKAPKLLI 1057 1 YAASTLQSGVPSRFSGSGSGTDFTLTISSLRLDDVATYYCQQYQSYPI
TFGQGTRLEIKRTVAAPSVF
BP 18b EIVLTQSPDSLAVSLGERATINCKSSQNLLYSSNNKNYLAWYQQKP 1058
-3 GQPPKLLINWASTRVSGVPDRFSGSGSGTDFTLTINSLQAEDVAVYY
CHQYYSSPYTFGPGTKVEIKRTVAAPSVF
BP6a- QPVLTQPPSVSVSPGQTASrrCSGDNLGDKYASWYQQKPGQSPVLVI 1059 9 FRDTKRPSGIPERFSGSNSGDTATLTISGTQAMDEADYYCQAWDSS
AGGVFGGGTRLTVLGQPKAAPSVTLFPPSS
BP6a- DVVMTQSPLSLPVTPGEPASISCRSSQTLLHSNGFNYLDWYLQKPGQ 1060 25 SPQLLMYLGSSRASGVPDRFSGSGSGTDFTLKISRVEAEDVGVYYC MQGTHWPPTFGQGTKLEIRRTVAAPSVF
BPx-5 QAVLTQPPSVSGTPGQTVIISCSGSNSNLGRNSANWYQQLPGTAPKL 1061
LIHSNHMRPSGVPDRFSGSKSGTSASLAISGLQSEDEADYYCAAWD
DSLNGWVFGGGTKLTVLGQPKAAPSVTLFPPSS
BP5a- QPVLTQPPSASGTPGQRVTISCFGSSSNIGSNYVYWYQQLPGTAPKL 1062 2 LIYRNNQRPSGVPDRFSGSKSGTSASLAISGLRSEDEADYYCAAWD
DSLSGWVFGGGTKLTVLGQPKAAPSVTLFPPSS
BP6a- DIQMTQSPSTLSASVGDRVSrrCRASQSVSSWLAWYQQKPGKAPKL 1063 15 LIYAASSLQSGVPSRFSGSGSGTDFTLTISNLQPEDSATYFCQQSYSTP
WTFGQGTKVELKRTVAAPSVF
BP6a- DIQMTQSPSSLSASVGDRVTITCQASHDISNYLNWYQQKPGKAPKL 1064 21 LIYDASNLETGVPSTFSGSGSGTDFTLTISSLQPEDIATYYCQQSYSTP
YTFGQGTKVEIKRTVAAPSVF
BP5a- QSALTQPASVSGSPGQSITISCTGTSSDVGGYNYVSWYQQHPGKAP 1065 5 KLMIYDVSKRPSGVSNRFSGSKSGNTASLTISGLQAEDEADYYCSSY
TTSSTLVFGGGTKVTVLGQPKAAPSVTLFPPSS
BP6a- SSELTQDPSVSVSPGQTASITCSGDKLGDKYVGWYQQKPGQSPVLII 1066 17 YRDYKRPSGTPERFSGSNSGNTATLTISGVEAGDEADYYCQVWDM
SSDPLVFGGGTKLTVLGQPKAAPSVTLFPPSS
BP6a- DIVMTQSPSTLSVSVGDRVTITCRASQSISSWLAWYQQKPGKAPKLL 1067 20 IYKASSLESGVPSRFSGSGSGTDFTLTISSLQPEDFATYYCQQSYSTPY
TFGQGTKLEIKRTVAAPSVF
BP3a- QSALTQPRSVSGSPGQSmSCTGTSSDVGGYNYVSWYQQHPGKAPK 1068 2 LMIYDVSKRPSGVSNRFSGSKSGNTASLTISGLQAEDEADYYCSSYT
S S ST Y VFGTGTKVT VLGQPKADPTVTLFPPS S
BP6a- QPVLTQPPSVSVSPGQTASrrCSGDDLGDKFASWYQQKPGQSPILVI 1069 24 YQDSKRPSGIPERFSGSNSGNTATLTISGTQAMDEADYYCQAWDGT
KVVFGGGTKVTVLGQPKAAPSVTLFPPSSKARG
BP6a- DIVMTQSPSSVSASVGDRVTITCRASQDISSWLAWYQQKPGKAPKL 1070 22 LIYAASSLQSGVPSRFSGSGSGTDFTLTISSLQPEDFASYYCQQANSF
PQTFGGGTKVEIKRTVAAPSVF
BP19a NFMLTQPHSVSESPGKTVTISCTRSSGRrVSTYVQWYQHRPGSPPRLI 1071 -2 MYDDVQRPSGVSARFSGSIDISSNSASLTISGLLAEDEADYYCQSYN
TSNLVFGGGTKLTVLSQPKAAPSVTLFPPSS
BPx-6 QSALTQPPSASASPGQSVTISCTGTSSDVGGYDFVSWYQQHPGQAP 1072
KLMIYEVNKRPSGVPDRFSGSKSGNTASLTVSGLQADDEADYYCCA
YAGSNILLFGGGTKLTVLGQPKAAPSVTLFPPSSKPRG
BP6a- AIQLTQSPSSLSASVGDRVTITCRASQSISTYLNWYQQKPGKAPKLLI 1073 18 YDTSSLHSGVPSRFSGSGSGTDFTLTISSLQPEDFATYHCQQSFITPYT
FGQGTKLEIKRTVAAPSVF
BP16a QSALTQPASVSGSPGQSITISCTGTSSDVGGYNYVSWYQQHPGKAP 1074 -1 KLMISDVSKRPSGVSNRFSGSKSGNTASLTISGLQAEDEADYYCSSY
TSNYTLVFGGGTKLTVLGQPKAAPSVTLFPPSS
BP19a DIVMTQSPATLSVSPGERATLSCRASQSVSSYLAWYQQKPGQAPRL 1075 -3 LIYDASNRATGIPARFSGSGSGTDFTLTISSLEPEDFAVYYCQHRGSW
PHTFGPGTKLEMKRTVAAPSVF
BP5b- EIVMTQSPATLSLSPGDRATLSCRASQNVGSYLGWYQQKVGQAPRL 1076 2 LIYDASNRATGVPGRFSGSGSGTDFTLTISSLEPEDFAVYYCQQYGSS
PLFTFGPGTKVDIKRTVAAPSVF BP2a- EIVMTQSPGTLSLSPGERATLSCRASQSVSSNYLAWYQQKPGQAPR 1077 1 LLIYDASNRATGIPARFSGSGSGTDFTLTISSLEPEDFALYYCQQYGT
SPLTFGGGTKLEIKRTVAAPSVF
BP18a DIVMTQSPLSLPVTPGEPASISCRSSQSLLHSNGYNYLDWYLQKPGQ 1078 -2 SPQLLIYLGSNRASGVPDRFSGSESGTDFTLKISRVEAEDVGVYYCM
QATHWPPRLTFGPGTKVDIKRTVAAPSVF
BPx-7 QSALTQPASVSGSPGQSITISCTGTSSDVGGYNYVSWYQQRPGKAPK 1079
LMIYDVSNRPSGVSNRFSGSKSGNTASLTISGLQAEDEADYYCSSYT
TSSTVVFGGGTKLTVLGQPKAAPSVTLFPPSS
BPx-8 RSALTQPASVSGSPGQSITISCTGSSNDVGGYKFVSWYQHHPGKAPK 1080
LLIYGVTQRPSGVSNRFSGSQSGNTASLTISGLQAEDEADYFCTSYT
RSTTPIFGGGTKLTVLGQPKAAPSVTLFPPSS
BPx-9 QSALTQPPSASGSPGQSVTVSCTGSSSDIGSYNYVSWFQRHPGQAPK 1081
LIIHDVTERPSGVPHRFSGSKSGNTASLTISGLQTEDEADYFCCSYSG
DHTLPFIFAPATQVTVLGQPKANPTVTLFPPSS
BPx- DIVMTQSPLSLPVTLGQPASISCRSSQGLVYSDGNTYLNWFQQRPGQ 1082 10 SPRRLIYKVSNRDSGVPDRFSGSGSGTDFTLKISRVEAEDVGVYYCM
QATHWPWTFGQGTKVEIK
BPx- QSVLTQPPSASGTPGQRVTISCSGSSSNIGSNYVYWYQQLPGTAPKL 1083 11 LIYRNNQRPSGVPDRFSGSKSGTSASLAISGLRSEDEADYYCAAWD
DSLSGPVFGGGTKLTVLGQPKAAPSVTLFPPSS
BPx- DVVMTQSPLSLPVTLGQPASISCRSSQSLVHGNGITYLNWFQQRPGQ 1084 12 SPRRLIYKVSNRDSGVPDRFSGSGSGTDFTLKISRVEAEDVGVYYCM
QALQTPSFGQGTRLEIKRTVAAPSVF
BPx- QSALTQPPSASGSPGESVTISCTGTSSDVGGYNYVSWYQQHPGKAP 1085 13 KLLIYDVTKRPSGVSNRFSGSKSGNTASLTISGLQAEDEADYYCSSY
TS S STV VFGGGTKLT VPGQPKA APS VTLFPPS S
BPx- QSALTQPRSVSGSPGQSVTISCTGTSSDVGGYNYVSWYQHHPGKAP 1086 14 KLMIYDVNKRPSGVPDRFSGSKSANTASLTVSGLQAEDEADYYCSS
YTAGGNWVFGGGTKLTVLGQPKAAPSVTLFPPSS
BPx- DVVMTQSPLSLPVTPGEPASISCRSSQSLLHSNGYNYLDWYVQKPG 1087 15 QSPQHLIYLGSIRASGVPDRFSGSGSGTDFTLKISRVEAEDVGVYYC
MQGLQTPPTFGPGTKMDIKRTVAAPSVF
BPx- DVVMTQSPLSLPVTLGQPASISCRSSQSLVYSDGNTYLSWFHQRPGQ 1088 16 SPRRLIYKVSNRDSGVPDRFSGSGSGTDFTLKISRVEAEDVGVYYCM
QGTHWPWTFGQGTKVEIKRTVAAPSVF
BPx- QSALTQPASVSGSPGQSITISCTGTSSDVGGYNYVSWYQQHPGKAP 1089 17 KLMIYDVSKRPSGVSNRFSGSKSGNTASLTISGLQAEDEADYYCSSY
TSSSTWVFGGGTK
BPlb- QSALTQPPSASGSPGQSVTISCSGTSSDIGGYDYVSWYQQHPGKAPK 1090 1 LIIYEVSKRPSGVPDRFSGSKSGNTASLTVSGLQADDGAHYYCSAYA
GRNNMVFGGGTKVTVVGQPKAAPSVTLFPPSS
BPx- QSVLTQPPSASGTPGQRVIISCSGSRSNNGSNYIYWYQHLPGTAPKL 1091 18 LIYRNNQRPSGVPDRFSGSKSGTSASLAISGLQSEDEADYYCAAWD
RSLNAWVFGGGTKLTVLSQPKAAPSVTLFPPSS
BPx- DIVMTQTPLSLPVTLGQPASISCRSSQSLVYSDGKTYLNWFQQRPGQ 1092 19 SPRRLIYKVSNRDSGVPDRFSGSGSGTDFTLKITRVEAEDVGVYYCM
QGTHWPHTFGQGTKLEIKRTVAAPSVF
BPx- EIVMTQSPATLSLSPGERATLSCRASQSVNSSYFAWYHRRPGQAPRL 1093 20 LISRTSTRAAGIPDRFSGSGSGTDFTLKISRVEAEDVGVYYCMQGTY
WPYTFGQGTKLEIKRTVAAPSVF
BPx- QSALTQPPSASGSPGQSVTISCTGTSSDVGGYNYVSWYQQHPGKAP 1094 21 KLMIYDVSNRPSGVSNRFSASKSGNTASLTISGLQAEDEADYYCSSY
TSSSTLDVVFGGGTKLTVLGQPKAAPSVTLFPPSS
BPx- QSALTQPASMSGSPGQSmSCTGTSSDLGGYNYVSWYQQHPGKAP 1095 22 KLMIYDVSKRPSGVSNRFSGSKSGNTASLTISGLQAEDEADYYCSSY
TS S STPWVFGGGTKLT VLGQPKA APS VTLFPPS S
BPx- NFMLTQPHSVSESPGKTVTISCTRSSGSIASNYVQWYQQRPGSSPTT 1096 23 VIYEDNQRPSGVPGRFSGSIDSSSNSASLTISGLQTEDEADYYCQSYD
GSDVVFGGGTKLTVLSQPKAAPSVTLFPPSS
BP5b- QSALTQPASVSGSPGQSITISCTGTSSDVGGYNYVSWYQQHPGKAP 1097 6 KLMIYDVSKRPSGVSNRFSGSKSGNTASLTISGLQAEDEADYYCSSY
TS S STWVFGGGTKLT VLGQPKA APS VTLFPPS S
BPx- NFMLTQPHSVSESPGKTVTISCTGSSGSIASNYVQWYQQRPGSSPTT 1098 24 LIYEHNQRPSGVPDRFSGSVDSSSNSASLTISGLKTEDEADYYCQSY
DDNNVVFGGGTKLTVLGQPKAAPSVTLFPPSS
BPx- QSALTQPPSASGSPGQSVTISCTGTSSDIGGYNYVSWYQVHPGKAPK 1099 25 LIIYDVSKRPSGIPDRFSGSKSGNAASLTVSGLRTEDEADYYCSSYAG
NNNLIFGGGTKVT VLS QPKA APS VTLFPPS S
BPx- QSALTQPASVSGSPGQSITISCTGTSSDVGGYNYVSWYQQHPGKAP 1100 26 KLMIYDVSKRPSGVSNRFSGSKSGNTASLTISGLQAEDEADYYCASY
TSSTSVVFGGGTKLAVLGQPKAAPSVTLFPPSS
BPx- DVVMTQTPLSLPVTPGQPASISCRSSQSLVYSNGNTYLTWFQQRPG 1101 27 QSPRRLLHEVSKRDSGVPDRFVGSGSGTDFTLKISRVEPEDVGIYYC
MQGSQWPPTFGQGTKLEIKRTVAAPSVF
BPx- DIVMTQTPVSLPVTLGQPASISCRSSQSLVHSDGNTYLNWFQQRPGQ 1102 28 SPRRLIYKVSTRDSGVPDRFSGSGSATDFTLKISGVEAEDVGVYYCM
QGTHWPKTFGQGTKVEIKRTVAAPSVF
BPx- EIVMTQSPLSLPVTPGEPASISCRSSQSLLHKNGNNYLDWYLQKPGQ 1103 29 SPQLLIYMASKRASGVPDRFSGSGSGTDFTLKISRVEAEDVGVYYC
MQALQTPPTFGGGTKVEIKRTVAAPSVF
BPx- EIVLTQSPATLSLSPGERATLSCRASQSrrSNYLAWYQQKPGQAPRL 1104 30 LIVGASSRATGIPDRFSGSGSGTDFTLTISRLEPEDSAVYYCQQYGNS
PRTFGQGTKLEIKRTVAAPSVF
BPx- DIQMTQSPSTLSASVGDRVTITCRASQSISSWLAWYQQKPGKAPKLL 1105 31 IYKASSLESGVPSRFSGSGSGTEFTLTISSLQPEDFATYYCQQYQSYPI
TFGQGTRLEIKRTVAAPSVF
BPx- EIVLTQSPLSLPVTPGEPASISCRSSESLLHSIGYNYVDWYVQKPGQS 1106 32 PQLLIYLGSNRASGVPDRFSGSTSGTDFTLKISRVEAEDVGVYYCMQ
ALQTSITFGQGTRLEIKRTMAAPSVF
BPx- QSALTQPASVSGSPGQSITISCTGTSSDVGTYNYVSWYQQHPGKAPK 1107 33 LMIYDVNNRPSGVSNRFSGSKSGSTASLTISGLQAEDEADYYCSSYT
STSTPYVFGTGTKVTVLRQPKANPTVTLFPPSS
BPx- QSALTQPPSASGSPGQSVTISCTGTSSDIGGYNYVSWYQQHPGKAPK 1108 34 LMIYEVSERPSGVPDRFSGSKSGNTASLTIAGLQGEDEADYYCSSYR
SSSTVVFGGGTKLTVLGQPKAAPSVTLFPPSS
BPx- QSALTQPASVSGSPGQSITISCTGTSSDVGGYNYVSWYQQHPGKAP 1109 35 KLMIYDVSNRPSGVSNRFSGSKSGNTASLTISGLQAEDEADYYCSSY TS S S TL VFGTGTK VT VLG QPK ANPT VTLFPPS S
BPx- DIQMTQSPSFLSASVGDRVTITCRASQGISSHLAWYQQKPGKAPKLL 1110 36 IYATSTLQSGVPSRFSGSGSGTDFTLTISSLQPEDFATYYCQQFDDLP
VTFGPGTKVEIKRTVAAPSVF
BPx- DIVMTQSPLSLPVTPGEPASISCRSSQTLLHANGFNYLDWYLQKPGQ 1111 37 SPQLLMYLGSSRASGVPDRFSGSGSGTDFTLKISRVEAEDVGVYYC
MQTLQTPPWTFGQGTKVEIKRTVAAPSVF
BPx- DIVLTQSPATLSVSPGDRATLSCRASQSVNRNLAWYQQRPGQAPRIL 1112 38 IYDASTRATGIPTRFRGSGSGTEFTLTISSLQSEDFGIYYCQQYNNWL
SFGGGTKVEVKRTVAAPSVF
BPx- NFMLTQPHSVSESPGKTVTISCTRSSGTrVSTYVQWYQQRPGSSPTT 1113 40 VIYEDNQRPSGVPDRFSGSIDSSSNSASLTISGLKTEDEADYYCQSYD
S SNRKVFGGGTKLT VLS QPKA APS VTLFPPS S
BPx- QSALTQPASVSGSPGQSITISCTGTSSDVETYNLVSWYQQHPGKAPK 1114 41 LMIYDVTKRPSGVPDRFSGSKSGNTASLTISGLQADDEADYYCSSYA
DSIAFVIFGGGTKVTVLGQPKAAPSVTLFPPSS
BPx- DIVMTQSPATLSLSPGERATLSCRASQSFSSNLAWYQQKPGQAPRLL 1115 42 IYDASNGATGVPDRFSGSGSGTDFTLTISRLEPEDFAVYYCQQYGSS
PRTFGQGTKLEIK
BPx- QSVLTQPPSVSGAPGQRVTISCTGSSSNVGAGYDVHWYQQLPGTAP 1116 43 KLLIYGNTNRPSGVPERFTGSKSGTSASLAISGLRSEDEADYYCATW
DDSLNGPVFGGGTKLTVLGQPKAAP
BP 16b QSVLTQPPSVSGAPGQRVTISCTGSTSNIGAGYDVHWYQQLPGTAP 1117 -1 KLLIYGNTNRPSGVPDRFSGSKSGTSASLAISGLQSEDEADYYCAAW
DDSLNGWVFGGGTKLTVLGQPKAAPSVTLFPPSS
BPx- QAVLTQPPSASGTPGQRVTISCSGSSSNIGSNYVYWYQQLPGTAPKL 1118 44 LIYRNNQRPSGVPDRFSGSKSGTSASLAISGLRSEDEADYYCAAWD
DSLSGWVFGGGTKLTVLGQPKAAPSVTLFPPSS
BPx- QSVLTQPPSASGTPGQRVTISCSGSNSNIGSNHVYWYQQLPGTAPKL 1119 45 LIYSNNQRPSGVPDRFSGSKSGTSASLAISGLQSEDEADYYCAAWDD
NVNGWVFGGGTKLTVLGQPKAAPSVTLFPPSS
BPx- QAVLTQPPSASGTPGQRVTISCSGSSSNIGTNAVNWYQQLPGTAPKH 1120 47 LIYSNNQRPSGVPDRFSASKSGTSASLAISGLQSDDESDYYCAAWDD
S LNG V VFGGGTKLT VL
BPx- QPVLTQSPSASGTPGQRVTISCSGSSSNIGSNYVYWYQQLPGTAPKL 1121 48 LIYRNSQRPSGVPDRFSGSKSGTSTSLAISDLRSEDEADYYCAAWDD
SLSGWVFGGGTKLTVLGQPKAAPSVTLFPPSS
BPx- DIVMTQSPATLSLPPGERATLSCRASQSVSSYLAWYQQKPGQAPRL 1122 49 LIYDASNRATGIPARFSGSGSGTDFTLTISSLQAEDVAVYFCQQYYST
PLTFGGGTKVEIKRTVAAPSVF
BPx- QPVLTQPPSASGTPGQRVTISCSGSSSNIGSNTVNWYQQLPGTAPKL 1123 50 LIYINNQRPSGVPDRFSGSKSGTSASLAISGLQSEDEANYYCAAWDD
SLSGPVFGGGTKLTVLGQPKAAPSVTLFPPSS
BPx- QAVLTQPPSASGAPGQTVTISCSGSSSNIGSKTVNWYQQVPGTVPKL 1124 51 LIYNNNQRPSGVPDRFSGSKSGTSASLAITGLRSEDEADYYCAAWD
DSLSGPMFGGGTKLTVLGQPKAAPSVTLFPPSS
BPx- DIVMTQSPSALSASVGDRVTITCRASQSIGSWLAWFQQKPGKAPNL 1125 52 LIYKASSVESGVPSRFSGSGSGTEFTLTISSLQPEDFATYYCQQSYSTP
YTFGQGTKLEIKRTVAAPSVF BPx- QAVLTQPPSVSVAPGKTAKITCGGLNIGSKSVHWYQQKAGQAPVL 1126 53 VMSYDSDRPSGIPERFSGSNSGSTATLSISRVAAGDEADYYCQVSDS
DTDHPVFGGGTKLTVL
BPx- DIVMTQSPLSLPVTPGEPASISCRSSQSLLHSNGYNYLDWYLQKPGQ 1127 54 SPQLLIYLGSNRASGVPDRFSGSGSGTDFTLKISRVEAEDVGVYYCM
QALQTPYTFGQGTKLEIKRTVAAPSVF
BPx- SSELTQDPAVSVALGQTVRITCQGDSLRTYYASWYQQKPGQAPKLL 1128 55 IYGKNNRPSGIPERFSGSSSRNSASLTITGTQAEDEADYYCDSRDSST
S QW VFGGGTKVT VLGQ
BPx- QPVLTQPPSASGTPGQRVTISCSGTSSNIGSNTVSWYHHLPGTAPKLL 1129 56 IYSNNQRPSGVPDRFSGSRSGTSAALAISGLESEDEADYYCATWDDS
LNGWVFGGGTKLTVLGQPKAAPSVTLFPPSS
BPx- DIVMTQSPDSLAVSLGERATIKCQSSQSVLYSFSNKNYLAWYQQKP 1130 57 GQPPKLLIYWASTRKSGVPDRFSGSGSGTDFSLTISSLQAEDVAVYY
CQQYYLLPITFGQGTRLEIK
BPx- QPVLTQPPSVSVAPGKTARITCGGNNIGSKSVHWYQQKPGQAPVLV 1131 58 IYYDSDRPSGIPERFSGSNSGNTATLTISRVEAGDEADYYCQVWDSS
SDHVVFGGGTKLTVLGQPKAAPSVTLFPPSS
BPx- EVVLTQSPLSLPVTPGEPASISCRSSQSLLHSIGYNYVDWYVQKPGQ 1132 59 SPRLLIYLGSNRASGVPDRFSGSGSGTDFTLKITRVEAEDVGVYYCM
QALQTIFTFGPGTKVDIKRTVAAPSVF
BPx- QPGLTQPPSASGTPGQRVTISCSGSSSNIGSNYVYWYQQLPGTAPKL 1133 60 LIYRNNQRPSGVPDRFSGSKSGTSASLAISGLRSEDEGDYYCASWDD
SVHGWVFGGGTKLTVLGQPKAAPSVTLFPPSS
BPx- QSALTQPASVSASPGQSITISCTGTSSDVGSYNYVSWYQQHPGKAPK 1134 61 LIIFDVNNRPSGVSNRFSGSKSGNTASLTISGLQAEDEADYYCSSDTS
SSTLVFGGGTKLTVLGQPKAAPSVTLFPPSS
BPx- DIVMTQSPDSLAVSLGERATISCKSSQSVLYSSNNKNYLAWYQQKP 1135 62 GHPPKLLIYGASTRESGVPDRFSGSGSETDFTLTISSLQAEDAAVYYC
MQAAQLPLTFGGGTRVEIRRTVAAPSVF
BPx- QSVLTQPPSVSAAPGQKVTISCSGSSSNIGNQYVSWYQHLPGTAPKL 1136 63 LIYDNNKRPSGIPDRFSGSKSGTSATLDITGLQTGDEADYYCGTWDT
S LSGFLF AGGTKLTVLS QPKA APS VTLFPPS S
BPx- QSVLTQPPSSSASPGESARLTCTLPSDINVGQPKAAPSVTLFPPSS 1137 64
BP6b- QSVLTQPPSVSGAPGQRVTISCTGSGSNIGRGYDVHWYQQLPGTAP 1138 5 KLLIYGNTKRPSGVPERFAASKSGTSATLDITGLQTGDEADYYCGT
WDTSLSGFLFAGGTKLTVLSQPKAAPSVTLFPPSS
BPx- QSVLTQPPSASGTPGQRVTISCSGSSSNIGSNTVNWYQQLPGTAPKL 1139 65 LIHTNNQRPSGVPDRFSGSKSGTSASLAISGLQSEDEADFYCAAWDD
S LNGPVFGGGTKLTVLGQPKA APS VTLFPPS S
BPx- NFMLTQPHSVSESPGKTVTISCTRSSGRIASTYVQWYQHRPGSAPNI 1140 66 VIYEDDQRPSGVPDRFSGYVDNSSNSASLTISGLKPEDEAEYYCQSY
DGDNAVFGGGTKLTVLGQPKAAPSVTLFPPSS
BPx- QSALTQPASVSGSLGQSITISCTGTSGDVETYNLVSWYQQHPGKVPK 1141 67 LIIYEVSERPSGVSNRFSGSKSGNTASLTISGLQAEDEADYYCSSYAG
SGTLRLFGGGTKVTVLGQPKAAPSVTLFPPSS
BPx- QSVLTQPPSVSAAPGQKVTISCTGSSSNIGAGYDVHWYQQVPGTAP 1142 68 KLLIYGNSYRPSGVSDRFSGSKSGTSASLAISDLQPEDEAYYYCATW DDSLRGDVVFGGGTKLTVLGQPKAAPSVTLFPPSS
BPx- DVVMTQSPLSLPVTPGEPASISCRSSQSLLHSNGYNYLDWYLQKPG 1143 69 QSPQLLIYLASNRASGVPDRFSGSGSGTDFTLKISRVEAEDVGVYYC
MQALQTPATFGQGTKLEIKRTVAAPSVF
BPx- QPVLTQPPSMSVSPGQTASITCSGHKLGHKYVGWYQQRPGQSPVLV 1144 70 IYQDNKRPSGIPERFSGSNSGNTATLTISETQAIDEADYYCQAWDSSS
TAYVFGTGTKVTVLGQPKANPTVTLFPPSS
BPx- QSALTQPPSASGSPGQSVTISCSGTSSDVGGYNYVSWYQQYPGKAP 1145 71 KLVISEVSKRPSGVPDRFSGSKSGNTASLTVSGLQAEDEAEYYCSSY
VGGNRVVFGGGTKLTVLGQPKAAPSVTLFPPSS
BPx- DIQMTQSPSTLSASVGDRVTITCRASQSISSYLNWYQQKPGKAPKLL 1146 72 IYAASSLQSGVPSRFSGSGSGTDFALTISSLQPEDFATYYCQQSYSTP
WTFGQGTKVEIKRTVAAPSVF
BPx- QPVLTQPPSASGTPGQRVTISCSGSSSNIGSNYVYWYQQLPGTAPKL 1147 73 LMYRNNQRPSGVPDRFSGSKSGTSASLAISGLRSEDEADYYCAAWD
DSLSGWVFGGGTKLTVLGQPKAAPSVTLFPPSS
BPx- EIVMTQSPGTLSLSPGERVTLSCRASQYITSGYLAWYQQKPGQAPRL 1148 74 LFYGASRRATGIPDRFSGSGSGTDFTLTISRLEPEDFAVYYCQQYGSS
PTYTFGQGTKLEIKRTVAAPSVF
BP8b- EIVLTQSPGTLSLSPGERATLSCRASESISNNYLAWYQQKPGQAPRL 1149 1 LIYGASSRATGIPDRFSGRGSGIDFTLTISRLEPEDFAVYYCQQYGYS
PTFGPGTKVDVKRTVAAPSVF
BPx- DIQMTQSPSSLSASVGDRVTVTCRASQSISIFVNWYQQRPGKAPKLL 1150 75 IYAASSLQTGVPSRFSGSGSGTEFTLTISSLQPEDYATYYCQQSYSML
SWTFGPGTKVENKRTVAAPSVF
BPx- QAVLTQPPSASGTPGQRVTISCSGSSSNIGSNYVSWYQQLPGTAPKL 1151 76 LIFSNNQRPSGVPDRFSGSKSGTSASLAISGLQSEDEADYYCAAWDD
SLNGWVFGGGTKLTVLGQPKAAPSVTLFPPSS
BPx- QSALTQPASVSGSPGQSITISCTGTSSDVGAYNYVSWYQQYPGKAP 1152 77 KLMIYDVSKRPSGVSNRFSGSKSGNTASLTISGLQAEDEADYYCGSY
TTSSTLVFGGGTKVTVLGQPKAAPSVTLFPPSS
BP6b- SSELTQAPSVSVSPGQTASITCSGDKLGDKYVSWYQQRPGQSPVVVI 1153 6 YQDIRRPSGIPERFSGSNSGNTATLTISGTQAMDEADYYCQAWDSSS
WVFGGGTKLTVLSQPKAAPSVTLFPPSS
BPx- EIVLTQSPLS LP VTPGEP AS ISCRS S QSLLHS NG YN YLD W YLQKPGQS 1154 78 PQLLIYLGSNRASGVPDRFSGSVSGTDFTLKISRVEAEDVGVYYCMQ
GTHWSYTFGQGTKLEIKRTVAAPSVF
BPx- QSALTQPASVSGSPGQSITISCTGTSSDIGTYNYLSWYQQHPGKAPK 1155 79 LMIYDVVYRPSGVSNRFSGSKSGNTASLTISGLQAEDEADYYCSSYA
GSSTVVFGGGTKLTVLGQPKAAPSVTLFPPSS
BPx- DIQMTQSPASLSVSIGDRVTITCQASQDIANHLNWYQQKPGDVPKLL 1156 80 IFDATHMERGVPSRFDGSRSGTDFTFTISSLQPEDVATYYCQQFDELP
LTFGGGTKVRIKGTVAAPSVF
BPx- NFMLTQPHSVSESPGKTITISCTRNSGRIASNYVQWYQQRPGSPPTTL 1157 81 IYEDSLRPSGVPDRFSGSIDTSSNSASLTISGLETEDEADYYCQSYDSR
VYVFGTGTKVTAL
BPx- SSELTQDPAVSVALGQTVRITCQGDSLKKYYASWYQQKPGQAPVL 1158 82 VIYGKNLRPSGIPERFSGSSSGDTASLTISATQAEDEADYYCNSRDSS
GNPIFGGGTKLTVLGQPKAAPSVTLFPPSS BPx- DVVMTQSPLSLPVTLGQPASISCRSSQSLVYSDGNTFLNWFQQRPGQ 1159 83 PPRRLIYKVSNRDFGVPDRFSGSGSGTDFTLKISRVEADDVGVYYC
MQGTHWPFTFGPGTK
BPx- QSVLTQPPSVSEAPRQRVTISCSGSSSNIGNNAVNWYRQLPGKAPKL 1160 84 LIYDNNKRPSGIPDRFSGSKSGTSATLGITGLQTGDEADYYCGTWDS
SLSAWVFGGGTKLTVLGQPKAAPSVTLFPPSS
BPx- DVVMTQSPDSLAVSLGERATINCKSSQNILYSSTNKNYLAWYQQKP 1161 115 GQPPKLLIYWASTRESGVPDRFSGSGSGTDFTLTISSLQAEDVAVYY
CQQYYSTPVTFGQGTKVEIKRTVAAPSVF
BPx- QSALTQPASVSGSPGQSITISCTGTSSDITIYNYVSWFQQHPGKVPKL 1162 116 MIYDVNDRPAGVSSRFSGSKSGNTASLTISGLQAEDEADYYCSSYTY
SNTLVFGGGTKVTVLGQPKAAPSVTLFPPSS
BPx- EIVMTQSPLS LP VTPGEP AS ISCRS S QSLLHS NG YN YLD W YLQKPGQ 1163 117 SPQLLMYLGSKRASGVPDRFSGSGSGTDFTLKISRVEAEDVGVYYC
MQARQTPVTFGQGTRLETKRTVAAPSV
BPx- NFMLTQPHSVSESPGKTVTISCTGSSGSIASNYVQWYQQRPGSAPTT 1164 118 VIYEDNQRPSGVPDRFSGSIDSSSNSASLTISGLKTEDEADYYCQSYD
S S ILV VFGGGTKLT VL
BP6b- SSELTQDPAVSVALGQTVRITCQGDSLKKYYASWYQQKPGQAPVL 1165 11 VIYGRNLRPSGIPERFSGPYSGNTASLTITGAQAEDEADYYCNSRDS
RGNLLVFGGGTRLIVLGQPKAAPSVTLFPPSSKASGA
BP 18b DIVMTQSPDSLAVSLGERATINCKSSQSVLYSSNNKNYLAWYQQKP 1166 -4 GQPPKLLIYWASTRESGVPDRFSGSGSGTDFTLTISSLQAEDVAVYY
CQQYYTIPHTFGQGTKLEIKRTVAAPSVFKASGA
BP6b- QPVLTQPPSVSVSPGQTASrrCSGDKLGDKYASWYQQKPGQSPLLVI 1167 2 YQDNKRPSGIPERFSGSNSGNTATLTISGTQAMDEADYYCQAWDSS
TVMFGGGTKLTVLGQPKAAPSVTLFPPSSKASGA
BP6b- DIVMTQSPLSLPVTPGEPASISCRSSQSLLHTNGYNFLDWYLQKPGQ 1168 10 SPQLLIHLGSYRASGVPDRFSGSGSGTDFTLKISRVAAEDVGVYYCM
QALHTPMYTFGQGTKLEIKRTVAAPSVFKASGA
BP 16b NFMLTQPHSVSESPGKTVTISCTGSSGSIASYYVQWYQQRPGSSPTT 1169
-2 VIYEDNQRPSGVPDRFSGSIDSSSNSASLTISGLKTEDEADYYCQSYD
SSNRWVFGGGTKLTVLGQPKAAPSVTLFPPSSKASGA
BP 18b EIVMTQSPLSLPVTPGEPASISCRSSQSLVHSNGNTYLNWFHQRPGQ 1170 -5 SPRRLIYEVSNRDSGVPDRFSGSGSGTDFTLKISRVEAEDVGVYYCM
QGTHWPYTSGQGTKLEIYRTVAAPSVFKASGA
BP7b- DIQMTQSPSSLSASVGDRVTITCRASQSISRHLNWYQQKPGKAPKLL 1171 1 IYAASTLQSGVPSRFSGSGSGTDFTLTISSLQPEDFATYYCQQSASTP
LTFGGGTKVEIKRTVAAPSVFKASGA
BP5b- DIVMTQSPLSLPVTLGQSASISCRSSQGLVYSDGNTFLSWFHQRPGQ 1172 7 SPRRLIYMVSNRDSGVPDRFSGSGSGTDFTLKISRVEAEDVGIFYCM
QGTHWPWTFGQGTKVEIKRTVAAPSVFKASGA
BPx- DIQMTQSPSSLSASVGDRVTITCRASQNIINYLNWYQQKPGKAPDLL 1173 85 IHTASNLQSGVPSRFSGSGFGTDFSLTITSLQPEDFATYYCQHSFSSPY
TFGQGTKLEIKRTVAAPSVFKASGA
BP 18b DIVMTQSPLSLPVTPGEPASISCRSSQTLLHSNGFNYLDWYLQKPGQ 1174 -6 SPQLLMYLGSSRASGVPDRFSGSGSGTDFTLKISRVEAEDVGVYYC
MQALQIPRTFGQGTKLEIKRTVAAPSVFKASGA
BP20b DIQMTQSPSSLSASVGDRVTITCRASQSISSYLNWYQQKPGKAPKLLI 1175 -1 YAASSLQSGVPSRFSGSGSGTDFSLTITNLQPEDIATYYCQQTYIYPR
TFGQGTKVEIKRTVAAPSVFKASGA
BPx- EVVMTQSPLSLPVTLGQPASISCRSSQSLVYSDGNTYLNWFQQRPG 1176 86 QSPRRLLYKVSNRDSGVPDRFSGTGSGTDFTLRISRVEAEDVGVYY
CMQGSHWPLTFGGGTKVEIKRTVAAPSVFKASGA
BP 19b QSALTQPASVSGSPGQSITISCTGTSNDIGRYDYVSWYQQHPGKAPK 1177 -3 LIIYAVKARPSGVSSRFSGSKSGNMASLTISGLQAEDEADYYCGSYT
RSSTARFGTGTRVTVLGQPKANPTVTLFPPSSKASGA
BPx- SSELTQDPSVSVSPGQTATITCSGSKLGDKYVSWYQQKPGQSPVLVI 1178 87 YEDDKRPSGSSERFSGSNSGNTATLTISGTQAMDEADYYCQAWDSI
TAVFGGGTKLTVLGQPKAAPSVTLFPPSSKASGA
BPx- SSELTQAPSVSVSPGQTASIPCSGDNLRSKYVSWYQQKPGQSPVMLI 1179 88 YQDDKRPSGIPEGFFGATSGNTATLIIAETQPLDEGDYYCQTWDGN
HVVFGGGTKLTVLGQPKAAPSVTLFPPSSKASGA
BP 18b EIVMTQSPLS LP VTLGQP AS ISCRS S QGLV YS DGNTYLNWFHQRPGQ 1180 -7 SPRRLIYKVSNRDSGVPDRFSGSGSGTDFTLKISRVEAEDVGIYYCM
QGTQWPYTFGQGTRLDIKRTVAAPSVFKASGA
BP17b QSALTQPASVSGSPGQSITISCTGTSSDVGGYNYVSWYQQHPGKAP 1181 -1 KLMIYDVSKRPSGVSNRFSGSKSGNTASLTISGLQAEDEADYYCSSY
TSSSTLVFGGGTKLTVLGQPKAAPSVTLFPPSSKASGA
BP 16b EIVLTQSPATLSLSPGERATLSCRASQSVSSYLAWYQQKPGQAPRLL 1182
-3 VYDASNRATGIPARFSGSGSGTDFTLTISSLEPEDFAVYYCQQRSGW
PPKVTFGGGTKVEIKRTVAAPSVFKASGA
BPx- DIQMTQSPSSLSASVGDRVTITCRASQSISSWLAWYQQKPGKAPKLL 1183 89 IYKASSLESGVPSRFSGSGSGTEFTLTITSLQPEDFATYYCQQSYSTPF
TFGPGTKVDIKRTVAAPSVFKASGA
BPx- QPVLTQPPSVSVSPGQTTNITCSGHELGDKYVSWYQQRPGQSPVVVI 1184 90 YQDNKRPSGIPERFSGSNSGNTATLTISGTQAMDEADYYCQAWDSS
TVVFGGGTKLTVLGQPKAAPSVTLFPPSSKASG
BP5b- SSELTQAPSVSVAPGRTARITCGGNNIGNKSVHWYQQQPGQAPVLV 1185 8 IYYDIDRPSGIPERFSGSNSGNTATLTISRVEADDEADYYCQVWDSN
SDHVVFGGGTKLTVLGQPKAAPSVTLFPPSSKASGA
BP 16b QSALTQPASVSGSPGQSITISCTGTSSDVGGYNYVSWYQQHPGKAP 1186 -4 KLLIYDVTKRPSGVSNRFSGSKSGNTASLTISGLQAEDEADYYCSSY
TS S STV VFGGGTKLT VLGQPKA APS VTLFPPS S KASG A
BP9b- NFMLTQPPSASGTPGQRVTISCSGSSSNIGNNAVNWYQQLPGTAPKL 1187 1 LIYSNNQRPSGVPDRFSGSKSGTSASLAISGLQSEDEADYYCATWDD
SLNGWVFGGGTKLTVLGQPKAAPSVTLFPPSSKASGA
BPx- DIVLTQSPLSLPVTPGEPASISCRSSQSLLHSNGFNYLDWYLQKPGQS 1188 91 PQLLIYLGSNRASGVPDRFSGSGSGTDFTLQISRVEAEDVGVYYCMQ
GLQTPYTFGQGTKLEIKRTVAAPSVFKASGA
BPx- QSVLTQPPSVSAAPGQTVTISCSGSTSNIGNNYVSWYEQLPGTAPKL 1189 92 LIYDNNRRPSGIPDRFSGSKSGTSAALGITGLQTGDEANYYCGTWDS
SLSAWVFGGGTKLTVLGQPKAAPSVTLFPPSSKASGA
BP12b QSVLTQPPSASGTPGQRVTISCSGSSSNIRSNTVNWYQQLPGTAPKL 1190 -1 LIYSNNQRPSGVPDRFSGSKSGTSASLAISGLQSEDEADYYCAAWDD
SLNGWVFGGGTKLTVLGQPKAAPSVTLFPPSSKASGA
BPx- NFMLTQPHSVSGSPGKTVTISCTRSSGSISSKYVQWYQQRPGSSPTT 1191 93 VMYEDKQRPSGVPARFSGSVDSSSNSATLTISGLKTEDEADYYCQS YDNTNVVFGGGTKLTVLGQPKAAPSVTLFPPSSKASGA
BPx- EVVLTQSPLSLPVTPGEPASISCRSSQSLLHSNGYNYLDWYLQKPGQ 1192 94 SPQLLIYMGSNRASGVPDRFSGSGSGTDFTLKISRVEAEDVGVYYC
MQALQTPFTFGPGTKVDIKRTVAAPSVFKASGA
BPx- NFMLTQPHSVSGSPGKTVTISCTGSNGAIASNYVQWYQQRPGGAPV 1193 95 TLIYEDNHRPPGVPDRFSGSIDTSSNSASLTISGLKTEDEADYYCQSY
DSVYNWVFGGGTKLTVLGQPKAAPSVTLFPPSSKASGA
BPx- QSALTQPASVSGSPGQSITISCSGTSSDVGVYESVSWYQHHPGEAPK 1194 96 LMIYDVTKRPSGVSNRFSGSKSGNTASLTISGLQAEDEADYYCSSYS
TS STFEVFGGGTRLTVLS QPKA APS VTLFPPS S KASG A
BP6b- DIQMTQSPSSLSASVGDRVTITCRASQGISSWLAWYQQKPGKAPKL 1195 9 LIYAASSLQSGVPSRFSGSGSGTDFTLTISSLQPEDFATYYCQQSYSTP
WTFGQGTKVQIKRTVAAPSVFKASGA
BPx- DIQMTQSPSTLSASVGDRVTITCRASQSISRWLAWYQQKPGEAPKLL 1196 97 IYKASTLEGGVPSRFSGSGSGTEFTLTISSLQPEDSATYYCLQDYTTP
YTFGQGTKLEIKRTVAAPSVFKASGA
BPx- QSVLTQPPSTSGTPGQRVTISCSGSSSNIGSNTVDWYQQLPGTAPNLL 1197 98 IHSNNRRPSGVPDRFSGSKSATSASLAISGLQSEDEADYYCEAWDDS
LSAVVFGGGTRVTVLGQPKAAPSVTLFPPSSKASGA
BPx- DIVMTQTPDSLAVSLGERATISCKSSQSILYDSNLKNYLAWYQHKPG 1198 99 HPPKLLISWASTRESGVPDRFSGSGSDTDFTLTISSLQAEDVAVYSCQ
QFYTTPYTFGQGTKLEIKRTVAAPSVFKASGA
BP6b- NFMLTQPHSVSESPGRTVTIPCSGRGGSIASDSVQWYQQRPGSAPTTI 1199 7 IYEDNQRPSGVPDRFSGSVDSSSNSASLTISGLRTEDEADYYCQSYD
ASS LWVFGGKTKLTVLGQPKA APS VTLFPPS S KASG A
BPx- QSALTQPPSASGSPGQSVTISCTGTSRDVGRYNYVSWYQQHPGKAP 1200 100 KLMISEVSKRPSGVPDCFSGSKSGNTASLTISGLQAEDEADYYCCSY
SGGYNWVFGGGTKLTVLGQPKAAPSVTLFPPSSKASGA
BP6b- QSVLTQPPSASGTPGQRVTISCSGSSSNIGSNTVNWYQQLPGTAPKL 1201 8 LIYSNNQRPSGVPDRFSGSKSGTSATLDITGLQTGDEADYHCQSYDS
SLSDSVVFGGGTKLTVLGQPKAAPSVTLFPPSSKASGA
BPx- EVVLTQSPDSLAVSLGERATINCKSSQSILSNSNNKNYLTWYQQKPG 1202 101 QPPQLIISWASTRESGVPDRFSGSGSGTDFTLTINSLQAEDVAVYYCQ
QYYSTPLTFGGGTKVEIKRTVAAPSVFKASGA
BPx- DIVMTQSPPSLPVAPGEPASISCRSSQSLLHSDGKNYVDWYLQKPGQ 1203 102 SPQLLIFLGSKRASGVPDRFSGSGSGTDFTLKISRVEAEDVGVYFCM
QGTRWP YTFGQGTKLEIKRTVAAPSVFKASGA
BPx- NFMLTQPHSVSESPGKTVTISCTGTGDSIASNYVQWYQQRPGSAPTT 1204 103 VIYEDNQRPSGVPDRFSGSIDSSSNSASLTISGLRTEDEADYYCQSYD
SVYNWIFGGGTRLTVLGQPKAAPSVTLFPPSSKASGA
BPx- QSVLTQPPSVSAAPGQKVTISCSGSSSNIGNNYVSWYQQLPGTAPKL 1205 104 LIYDNNKRPSGIPDRFSGSKSGTSASLAISGLQPEDEADYYCAVWDD
S LNGWVFGGGTKLTVLRQPKA APS VTLFPPS S KASG A
BPx- SSELTQDPSVSVSPGQTASITCSQTKLGDKYVSWYQQKPGQSPVLVI 1206 105 YEDDKRPSGISERFSGSNSGNTATLTISGTQAVDEADYYCQVYDSST
VVFGGGTKLTVLGQPKA APS VTLFPPS S KASG A
BPx- DIQMTQSPSTLSASVGDRVTITCRASQSISRRLAWYQQKPGKAPKLL 1207 106 ISGASNLEFGVPATFSGTGSGTEFTLTISSLQPDDFATYYCQQYESYQ
PPTFGGGTKVEMKRTVAAPSVFKASGA BP5b- DIQMTQSPSSVSASVGDRVTITCRASQGISNWLAWYQQKPGKAPKL 1208 9 LIYGASSLQSGVPSRFSGSGSGTDFTLTISSLQFEDFATYYCQQSQSTP
VTFGQGTRLEIKRTVAAPSVFKASGA
BPx- EIVMTQSPATLSLSPGDRAALSCRASHSVGSHVDWFQQKPGQAPRL 1209 107 LIYDASNRATGIPDRFSGSGSGTDFTLTISRLEPEDFAVYYCQQRSN
WPRALSFGGGTKVEIKRTVAAPSVFKASGP
BPx- QSALTQPASVSGSPGQSITISCTGTSSDVGAYNHVSWYQQYPGKAP 1210 108 KLMIYDVSNRPSGVSDRFSGSKSDNTASLTISGLQAEDEADYYCSSY
TSSNTWVFGGGTKLTVLGQPKAAPSVTLFPPSSKASGA
BPx- DIVMTQSPDSLAVSLGERATINCKSSQSVLYSSNNKNYLAWYQQKP 1211 109 GQPPKLLIYWASTRESGVPDRFSGSGSGTDFTLTISSLQAEDVAVYY
CQQYYSTPYTFGQGTKLEIKRTVAAPSVFKASGA
BP22b DIVMTQSPDSLAVSLGERATINCKSSQSVLYSSDNKNYLAWYQQKP 1212 -1 GQPPKLLIYWASTRESGVPDRFSGSGSGTDFTLTISSLQAEDVAVYY
CQQYYSTPSFGPGTKVDIKRTVAAPSVFKASGA
BPx- NFMLTQPHSVSESPGKTVTISCTRSSGSIASNYVQWYQQRPGSSPTT 1213 110 VIYEDNQRPSGVPARFSGSIDISSNSASLTISGLLTEDEADYYCQSYN
TSNLVFGTGTKVTVLRQPKANPTVTLFPPSSKASGA
BP6b- DIQMTQSPSSVFASVGDRVTITCRASQDISTWLAWYQQKPGKGPKL 1214 12 LIYGASSLQSGVPSRFSGSGSGTDFTLTISSLQPDDFATYYCQQYQES
PWTFGQGTKVEIKRTVAAPSVFKASGA
BPx- QSALTQPPSASGSRGQSVSISCSGSRSDIGYYNYVSWYQQHPGKAPK 1215 111 LIIFDVNKRPSGVSNRFSGSKSGNTASLTISGLQAEDEADYYCSSYTS
SSTWVFGGGTKLTVLGQPKAAPSVTLFPPSSKASGA
BPx- QPVLTQPPSVSVAPGKTARITCGGNNIGSKSVHWYQQKPGQAPVLV 1216 112 VYDDSDRPSGIPERFSGSNSGNTATLTISRVEAGDEADYYCQVWESS
SDEWDTTSDQWVFGGGTKVTVLGQPKAAPSVTLFPPSSKASGA
BPx- NFMLTQPHSVSESPGKTVTLSCTGSGGSIARNYVQWYQQRPGSAPS 1217 113 AVIYEDDRRPSGVPHRFSGSIDTSSNSATLTISGLKTEDEADYYCQSY
DSAHHWVFGGGTKLTVLSQPKAAPSVTLFPPSSKASGA
BP 10b QSVLTQPPSVSAAPGQKVTISCSGSSSNIGDNYVSWYQQLPGTAPKL 1218 -1 LIYDNNKRPSGIPDRFSGSKSGTSATLGITGLQTGDEADYYCGTWDD
RLSGVVFGGGTKLTVLSQPKAAPSVTLFPPSSKASGA
BPx- DIVMTQSPDSLAVSLGERATINCKSSQSVLYSSNNKNYLAWYQQKP 1219 114 GQPPKLLIYWASTRESGVPDRFSGSGSGTDFTLTISSLQAEDVAVYF
CQQYYGIPTFGGGTKVEIKRTVAAPSVFKASGA
BP6a- SSELTQPSVSVSPGQTATITCSGDKLGDKYASWYQQKPGQSPVLVIY 1664 3 QDTKRPSGIPARFSGSNSGNTATLTISGTQAMDEAAYYCQAWDSTT
VVFGGGTKLTVLGQPKAAPSVTLFPPSS
[00311] Recombinant GDF-modulating antibodies of the disclosure may comprise or be developed using any of the CDR-L sequences (CDR-Ll, CDR-L2 and/or CDR-L3) listed in Table 17. Some recombinant GDF-modulating antibodies of the disclosure may comprise or be developed using CDR-L sequences that comprise at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, at least about 99%, at least about 99.5% or at least about 99.9% sequence identity to any of the CDR-L sequences listed in Table 17. Recombinant GDF-modulating antibodies may, in some cases, comprise CDR-L domains presented, but with different combinations of CDRs from other clones listed. In some cases, such antibodies may interact with one or more of the recombinant proteins listed in Table 4, 9 or 10.
Table 17. CDR-L domains
CDR Clone ID CDR amino acid sequence SEQ
Region ID
NO
CDR-L1 BP6a-16 DKLGDKY 1220
CDR-L1 BP6a-19 QSISRW 1221
CDR-L1 BP3a-3 QSVSSY 1222
CDR-L1 BP6a-l QSLVYSDGNTY 1223
CDR-L1 BP18b-l QTLLHSNGFNY 1224
CDR-L1 BP19b-l SSDVGGYNY 1225
CDR-L1 BP6b-4 SSDIGGYDY 1226
CDR-L1 BP6a-8 SSNIGNNY 1227
CDR-L1 BP13a-l DSLRSYF 1228
CDR-L1 BP6a-23 DDLGDKF 1229
CDR-L1 BP5a-l SSNIGSNT 1230
CDR-L1 BP6a-l l SSNIGSST 1231
CDR-L1 BP5b-4 QGISSY 1232
CDR-L1 BPl la-1 SSDVGYYDH 1233
CDR-L1 BP6a-14 QTVNSND 1234
CDR-L1 BP6a-27 QSINRH 1235
CDR-L1 BP6a-13 QSISSW 1236
CDR-L1 BP4a-l SSDISIYNY 1237
CDR-L1 BP6a-6 QSVSSGY 1238
CDR-L1 BP19b-2 RSDIGYYNY 1239
CDR-L1 BP6a-4 SSDVGDYNY 1240
CDR-L1 BP19a-l MSNIGKNT 1241
CDR-L1 BP6a-7 SSDIGGYNY 1242
CDR-L1 BP6b-3 NKLGDKF 1243
CDR-L1 BP18b-2 QSVVYRDGNTY 1244
CDR-L1 BPx-1 QSLLHSDGNNY 1245
CDR-L1 BP21b-l SGSIASNY 1246
CDR-L1 BPx-2 HSISRF 1247
CDR-L1 BPx-3 QSISSY 1248
CDR-L1 BP15a-l QSVSSSY 1249
CDR-L1 BPx-4 DQLGDKF 1250
CDR-L1 BP5a-3 DSLKKYY 1251
CDR-L1 BP14a-l QSLVHSDGNTY 1252
CDR-L1 BP18b-3 QNLLYSSNNKNY 1253
CDR-L1 BP6a-9 DNLGDKY 1254
CDR-L1 BPx-5 NSNLGRNS 1255
CDR-L1 BP5a-2 SSNIGSNY 1256
CDR-L1 BP6a-15 QSVSSW 1257 CDR-L1 BP6a-21 HDISNY 1258
CDR-L1 BP6a-22 QDISSW 1259
CDR-L1 BP19a-2 SGRrVSTY 1260
CDR-L1 BPx-6 SSDVGGYDF 1261
CDR-L1 BP6a-18 QSISTY 1262
CDR-L1 BP5b-2 QNVGSY 1263
CDR-L1 BP2a-l QSVSSNY 1264
CDR-L1 BP18a-2 QSLLHSNGYNY 1265
CDR-L1 BPx-8 SNDVGGYKF 1266
CDR-L1 BPx-9 SSDIGSYNY 1267
CDR-L1 BPx-10 QGLVYSDGNTY 1268
CDR-L1 BPx-12 QSLVHGNGITY 1269
CDR-L1 BPx-18 RSNNGSNY 1270
CDR-L1 BPx-19 QSLVYSDGKTY 1271
CDR-L1 BPx-20 QSVNSSY 1272
CDR-L1 BPx-22 SSDLGGYNY 1273
CDR-L1 BPx-27 QSLVYSNGNTY 1274
CDR-L1 BPx-29 QSLLHKNGNNY 1275
CDR-L1 BPx-30 QSITSNY 1276
CDR-L1 BPx-32 ESLLHSIGYNY 1277
CDR-L1 BPx-33 SSDVGTYNY 1278
CDR-L1 BPx-36 QGISSH 1279
CDR-L1 BPx-37 QTLLHANGFNY 1280
CDR-L1 BPx-38 QSVNRN 1281
CDR-L1 BPx-40 SGTrVSTY 1282
CDR-L1 BPx-41 SSDVETYNL 1283
CDR-L1 BPx-42 QSFSSN 1284
CDR-L1 BPx-43 SSNVGAGYD 1285
CDR-L1 BP16b-l TSNIGAGYD 1286
CDR-L1 BPx-45 NSNIGSNH 1287
CDR-L1 BPx-47 SSNIGTNA 1288
CDR-L1 BPx-51 SSNIGSKT 1289
CDR-L1 BPx-52 QSIGSW 1290
CDR-L1 BPx-53 LNIGSKS 1291
CDR-L1 BPx-55 DSLRTYY 1292
CDR-L1 BPx-57 QSVLYSFSNKNY 1293
CDR-L1 BPx-58 NNIGSKS 1294
CDR-L1 BPx-59 QSLLHSIGYNY 1295
CDR-L1 BPx-61 SSDVGSYNY 1296
CDR-L1 BPx-62 QSVLYSSNNKNY 1297
CDR-L1 BPx-63 SSNIGNQY 1298
CDR-L1 BP6b-5 GSNIGRGYD 1299
CDR-L1 BPx-66 SGRIASTY 1300
CDR-L1 BPx-67 SGDVETYNL 1301
CDR-L1 BPx-68 SSNIGAGYD 1302
CDR-L1 BPx-70 HKLGHKY 1303
CDR-L1 BPx-74 QYITSGY 1304
CDR-L1 BP8b-l ESISNNY 1305 CDR-L1 BPx-75 QSISIF 1306
CDR-L1 BPx-77 SSDVGAYNY 1307
CDR-L1 BPx-79 SSDIGTYNY 1308
CDR-L1 BPx-80 QDIANH 1309
CDR-L1 BPx-81 SGRIASNY 1310
CDR-L1 BPx-83 QSLVYSDGNTF 1311
CDR-L1 BPx-84 SSNIGNNA 1312
CDR-L1 BPx-115 QNILYSSTNKNY 1313
CDR-L1 BPx-116 SSDITIYNY 1314
CDR-L2 BP6a-16 QDS 1315
CDR-L2 BP6a-19 GAS 1316
CDR-L2 BP6a-l KVS 1317
CDR-L2 BP18b-l LGS 1318
CDR-L2 BP19b-l DVS 1319
CDR-L2 BP6a-8 DNN 1320
CDR-L2 BP13a-l GKN 1321
CDR-L2 BP5b-3 EVS 1322
CDR-L2 BP5a-l SNN 1323
CDR-L2 BP6a-l l SIN 1324
CDR-L2 BP5b-4 AAS 1325
CDR-L2 BPl la-1 DVT 1326
CDR-L2 BP6a-14 DAS 1327
CDR-L2 BP6a-13 KAS 1328
CDR-L2 BP4a-l DVN 1329
CDR-L2 BP6a-6 GGS 1330
CDR-L2 BP6b-3 EDR 1331
CDR-L2 BP21b-l EDN 1332
CDR-L2 BPx-2 STS 1333
CDR-L2 BP6b-l QDT 1334
CDR-L2 BP6a-5 SDN 1335
CDR-L2 BP6a-10 QDR 1336
CDR-L2 BP18b-3 WAS 1337
CDR-L2 BP6a-9 RDT 1338
CDR-L2 BPx-5 SNH 1339
CDR-L2 BP5a-2 RNN 1340
CDR-L2 BP6a-17 RDY 1341
CDR-L2 BP19a-2 DDV 1342
CDR-L2 BPx-6 EVN 1343
CDR-L2 BP6a-18 DTS 1344
CDR-L2 BPx-8 GVT 1345
CDR-L2 BPx-20 RTS 1346
CDR-L2 BPx-24 EHN 1347
CDR-L2 BPx-29 MAS 1348
CDR-L2 BPx-36 ATS 1349
CDR-L2 BPx-43 GNT 1350
CDR-L2 BPx-48 RNS 1351
CDR-L2 BPx-50 INN 1352
CDR-L2 BPx-51 NNN 1353 CDR-L2 BPx-53 YDS 1354
CDR-L2 BPx-65 TNN 1355
CDR-L2 BPx-66 EDD 1356
CDR-L2 BPx-68 GNS 1357
CDR-L2 BPx-69 LAS 1358
CDR-L2 BPx-70 QDN 1359
CDR-L2 BP6b-6 QDI 1360
CDR-L2 BPx-79 DVV 1361
CDR-L2 BPx-80 DAT 1362
CDR-L2 BPx-81 EDS 1363
CDR-L3 BP6a-16 QAWDSSTVV 1364
CDR-L3 BP6a-19 QQTYTTPWT 1365
CDR-L3 BP3a-3 QQYYTTPFT 1366
CDR-L3 BP6a-l MQSTRWPWV 1367
CDR-L3 BP18b-l MQALQTPFT 1368
CDR-L3 BP19b-l SSYTSSSTFVV 1369
CDR-L3 BP6b-4 SSFTSSSTLV 1370
CDR-L3 BP6a-8 GTWDSSLSAV 1371
CDR-L3 BP13a-l DSRHSSGNYVI 1372
CDR-L3 BP5b-3 SSYAGSNSWV 1373
CDR-L3 BP6a-23 QAWDGTKVV 1374
CDR-L3 BP5a-l ATWDDSLTGVV 1375
CDR-L3 BP6a-l l AAWDDSLNGWV 1376
CDR-L3 BP5b-4 QQSYSTPLT 1377
CDR-L3 BP18a-l MQGTHWPYT 1378
CDR-L3 BPl la-1 SSYAGSYTWV 1379
CDR-L3 BP6a-14 QQYHGSQT 1380
CDR-L3 BP6a-27 QQSYSTPYT 1381
CDR-L3 BP6a-13 QQSYSTPWT 1382
CDR-L3 BP4a-l SSYTYSNTLL 1383
CDR-L3 BPla-1 SSYTSSSTLV 1384
CDR-L3 BP6a-6 QQRSIWPLT 1385
CDR-L3 BP19b-2 SSSVGSNNLV 1386
CDR-L3 BP6a-4 SSYTSSTTWV 1387
CDR-L3 BP6a-7 NSWTRSNNYI 1388
CDR-L3 BP5b-5 MQGTHWPLT 1389
CDR-L3 BP6b-3 QTWDSRTVV 1390
CDR-L3 BP18b-2 MQGSSWPYT 1391
CDR-L3 BPx-1 MQGLHTPTT 1392
CDR-L3 BP21b-l QSYDHMYNWV 1393
CDR-L3 BP6a-2 AAWDDDLNGPV 1394
CDR-L3 BP6b-l QAWDSTTVV 1395
CDR-L3 BPx-3 QQYYSSPWT 1396
CDR-L3 BP6a-5 AAWDDSLNGV 1397
CDR-L3 BP6a-10 AAWDDSLSGRV 1398
CDR-L3 BP15a-l QQYGSSPYT 1399
CDR-L3 BP6a-26 QQSGSSPWT 1400
CDR-L3 BP6a-12 QQYYSTPYT 1401 CDR-L3 BP5a-4 GTWDSSLSAGV 1402
CDR-L3 BP5b-l NSYTSSRTVV 1403
CDR-L3 BPx-4 QTWDSSAVV 1404
CDR-L3 BP5a-3 QAWDSSTAV 1405
CDR-L3 BP14a-l MQGTHWPRLT 1406
CDR-L3 BP3a-l QQYQSYPIT 1407
CDR-L3 BP18b-3 HQYYSSPYT 1408
CDR-L3 BP6a-9 QAWDSSAGGV 1409
CDR-L3 BP6a-25 MQGTHWPPT 1410
CDR-L3 BP5a-2 AAWDDSLSGWV 1411
CDR-L3 BP5a-5 SSYTTSSTLV 1412
CDR-L3 BP6a-17 QVWDMSSDPLV 1413
CDR-L3 BP3a-2 SSYTSSSTYV 1414
CDR-L3 BP6a-22 QQANSFPQT 1415
CDR-L3 BP19a-2 QSYNTSNLV 1416
CDR-L3 BPx-6 CAYAGSNILL 1417
CDR-L3 BP6a-18 QQSFITPYT 1418
CDR-L3 BP16a-l SSYTSNYTLV 1419
CDR-L3 BP19a-3 QHRGSWPHT 1420
CDR-L3 BP5b-2 QQYGSSPLFT 1421
CDR-L3 BP2a-l QQYGTSPLT 1422
CDR-L3 BP18a-2 MQATHWPPRLT 1423
CDR-L3 BPx-7 SSYTTSSTVV 1424
CDR-L3 BPx-8 TSYTRSTTPI 1425
CDR-L3 BPx-9 CSYSGDHTLPFI 1426
CDR-L3 BPx-10 MQATHWPWT 1427
CDR-L3 BPx-11 AAWDDSLSGPV 1428
CDR-L3 BPx-12 MQALQTPS 1429
CDR-L3 BPx-13 SSYTSSSTVV 1430
CDR-L3 BPx-14 SSYTAGGNWV 1431
CDR-L3 BPx-15 MQGLQTPPT 1432
CDR-L3 BPx-16 MQGTHWPWT 1433
CDR-L3 BPx-17 SSYTSSSTWV 1434
CDR-L3 BPlb-1 SAYAGRNNMV 1435
CDR-L3 BPx-18 AAWDRSLNAWV 1436
CDR-L3 BPx-19 MQGTHWPHT 1437
CDR-L3 BPx-20 MQGTYWPYT 1438
CDR-L3 BPx-21 SSYTSSSTLDVV 1439
CDR-L3 BPx-22 SSYTSSSTPWV 1440
CDR-L3 BPx-23 QSYDGSDVV 1441
CDR-L3 BPx-24 QSYDDNNVV 1442
CDR-L3 BPx-25 SSYAGNNNLI 1443
CDR-L3 BPx-26 ASYTSSTSVV 1444
CDR-L3 BPx-27 MQGSQWPPT 1445
CDR-L3 BPx-28 MQGTHWPKT 1446
CDR-L3 BPx-29 MQALQTPPT 1447
CDR-L3 BPx-30 QQYGNSPRT 1448
CDR-L3 BPx-32 MQALQTSIT 1449 CDR-L3 BPx-33 SSYTSTSTPYV 1450
CDR-L3 BPx-34 SSYRSSSTVV 1451
CDR-L3 BPx-36 QQFDDLPVT 1452
CDR-L3 BPx-37 MQTLQTPPWT 1453
CDR-L3 BPx-38 QQYNNWLS 1454
CDR-L3 BPx-40 QSYDSSNRKV 1455
CDR-L3 BPx-41 SSYADSIAFVI 1456
CDR-L3 BPx-42 QQYGSSPRT 1457
CDR-L3 BPx-43 ATWDDSLNGPV 1458
CDR-L3 BPx-45 AAWDDNVNGWV 1459
CDR-L3 BPx-47 AAWDDSLNGVV 1460
CDR-L3 BPx-49 QQYYSTPLT 1461
CDR-L3 BPx-51 AAWDDSLSGPM 1462
CDR-L3 BPx-53 QVSDSDTDHPV 1463
CDR-L3 BPx-54 MQALQTPYT 1464
CDR-L3 BPx-55 DSRDSSTSQWV 1465
CDR-L3 BPx-56 ATWDDSLNGWV 1466
CDR-L3 BPx-57 QQYYLLPIT 1467
CDR-L3 BPx-58 QVWDSSSDHVV 1468
CDR-L3 BPx-59 MQALQTIFT 1469
CDR-L3 BPx-60 ASWDDSVHGWV 1470
CDR-L3 BPx-61 SSDTSSSTLV 1471
CDR-L3 BPx-62 MQAAQLPLT 1472
CDR-L3 BPx-63 GTWDTSLSGFL 1473
CDR-L3 BPx-65 AAWDDSLNGPV 1474
CDR-L3 BPx-66 QSYDGDNAV 1475
CDR-L3 BPx-67 SSYAGSGTLRL 1476
CDR-L3 BPx-68 ATWDDSLRGDVV 1477
CDR-L3 BPx-69 MQALQTPAT 1478
CDR-L3 BPx-70 QAWDSSSTAYV 1479
CDR-L3 BPx-71 SSYVGGNWV 1480
CDR-L3 BPx-74 QQYGSSPTYT 1481
CDR-L3 BP8b-l QQYGYSPT 1482
CDR-L3 BPx-75 QQSYSMLSWT 1483
CDR-L3 BPx-77 GSYTTSSTLV 1484
CDR-L3 BP6b-6 QAWDSSSWV 1485
CDR-L3 BPx-78 MQGTHWSYT 1486
CDR-L3 BPx-79 SSYAGSSTVV 1487
CDR-L3 BPx-80 QQFDELPLT 1488
CDR-L3 BPx-81 QSYDSRVYV 1489
CDR-L3 BPx-82 NSRDSSGNPI 1490
CDR-L3 BPx-83 MQGTHWPFT 1491
CDR-L3 BPx-84 GTWDSSLSAWV 1492
CDR-L3 BPx-115 QQYYSTPVT 1493
CDR-L3 BPx-116 SSYTYSNTLV 1494
CDR-L3 BPx-117 MQARQTPVT 1495
CDR-L3 BPx-118 QSYDSSILVV 1496
CDR-L1 BP20b-l QSISSY 1497 CDR-L1 BPx-89 QSISSW 1498
CDR-L1 BPx-105 KLGDKY 1499
CDR-L1 BPx-99 QSILYDSNLKNY 1500
CDR-L1 BPx-93 SGSISSKY 1501
CDR-L1 BPx-92 TSNIGNNY 1502
CDR-L1 BP6b-7 GGSIASDS 1503
CDR-L1 BPx-100 SRDVGRYNY 1504
CDR-L1 BPx-95 NGAIASNY 1505
CDR-L1 BPx-111 RSDIGYYNY 1506
CDR-L1 BP6b-l l SLKKYY 1507
CDR-L1 BP18b-4 QSVLYSSNNKNY 1508
CDR-L1 BP6b-10 QSLLHTNGYNF 1509
CDR-L1 BP16b-2 SGSIASYY 1510
CDR-L1 BP18b-5 QSLVHSNGNTY 1511
CDR-L1 BP7b-l QSISRH 1512
CDR-L1 BP5b-7 QGLVYSDGNTF 1513
CDR-L1 BPx-85 QNIINY 1514
CDR-L1 BP18b-6 QTLLHSNGFNY 1515
CDR-L1 BPx-86 QSLVYSDGNTY 1516
CDR-L1 BP19b-3 SNDIGRYDY 1517
CDR-L1 BPx-88 NLRSKY 1518
CDR-L1 BP18b-7 QGLVYSDGNTY 1519
CDR-L1 BP17b-l SSDVGGYNY 1520
CDR-L1 BP16b-3 QSVSSY 1521
CDR-L1 BPx-90 ELGDKY 1522
CDR-L1 BP5b-8 NIGNKS 1523
CDR-L1 BP9b-l SSNIGNNA 1524
CDR-L1 BPx-91 QSLLHSNGFNY 1525
CDR-L1 BP12b-l SSNIRSNT 1526
CDR-L1 BPlOb-1 SSNIGDNY 1527
CDR-L1 BPx-94 QSLLHSNGYNY 1528
CDR-L1 BPx-96 SSDVGVYES 1529
CDR-L1 BP6b-9 QGISSW 1530
CDR-L1 BPx-97 QSISRW 1531
CDR-L1 BPx-98 SSNIGSNT 1532
CDR-L1 BPx-101 QSILSNSNNKNY 1533
CDR-L1 BPx-102 QSLLHSDGKNY 1534
CDR-L1 BPx-103 GDSIASNY 1535
CDR-L1 BPx-104 SSNIGNNY 1536
CDR-L1 BPx-106 QSISRR 1537
CDR-L1 BP5b-9 QGISNW 1538
CDR-L1 BPx-107 HSVGSH 1539
CDR-L1 BPx-108 SSDVGAYNH 1540
CDR-L1 BP22b-l QSVLYSSDNKNY 1541
CDR-L1 BPx-110 SGSIASNY 1542
CDR-L1 BP6b-12 QDISTW 1543
CDR-L1 BPx-112 NIGSKS 1544
CDR-L1 BPx-113 GGSIARNY 1545 CDR-L2 BP20b-l AAS 1546
CDR-L2 BPx-89 KAS 1547
CDR-L2 BPx-105 EDD 1548
CDR-L2 BPx-99 WAS 1549
CDR-L2 BPx-93 EDK 1550
CDR-L2 BPx-92 DNN 1551
CDR-L2 BP6b-7 EDN 1552
CDR-L2 BPx-100 EVS 1553
CDR-L2 BPx-111 DVN 1554
CDR-L2 BP6b-2 QDN 1555
CDR-L2 BP6b-l l GRN 1556
CDR-L2 BP6b-10 LGS 1557
CDR-L2 BP5b-7 MVS 1558
CDR-L2 BPx-85 TAS 1559
CDR-L2 BPx-86 KVS 1560
CDR-L2 BP19b-3 AVK 1561
CDR-L2 BPx-88 QDD 1562
CDR-L2 BP17b-l DVS 1563
CDR-L2 BP16b-3 DAS 1564
CDR-L2 BP5b-8 YDI 1565
CDR-L2 BP16b-4 DVT 1566
CDR-L2 BP9b-l SSN 1567
CDR-L2 BP12b-l SNN 1568
CDR-L2 BPx-94 MGS 1569
CDR-L2 BPx-106 GAS 1570
CDR-L2 BPx-112 DDS 1571
CDR-L3 BP20b-l QQTYIYPRT 1572
CDR-L3 BPx-89 QQSYSTPFT 1573
CDR-L3 BPx-105 QVYDSSTVV 1574
CDR-L3 BPx-99 QQFYTTPYT 1575
CDR-L3 BPx-93 QSYDNTNVV 1576
CDR-L3 BPx-92 GTWDSSLSAWV 1577
CDR-L3 BP6b-7 QSYDASSLWV 1578
CDR-L3 BPx-100 CSYSGGYNWV 1579
CDR-L3 BPx-95 QSYDSVYNWV 1580
CDR-L3 BPx-111 SSYTSSSTWV 1581
CDR-L3 BP6b-2 QAWDSSTVM 1582
CDR-L3 BP6b-l l NSRDSRGNLLV 1583
CDR-L3 BP18b-4 QQYYTIPHT 1584
CDR-L3 BP6b-10 MQALHTPMYT 1585
CDR-L3 BP16b-2 QSYDSSNRWV 1586
CDR-L3 BP18b-5 MQGTHWPYT 1587
CDR-L3 BP7b-l QQSASTPLT 1588
CDR-L3 BP5b-7 MQGTHWPWT 1589
CDR-L3 BPx-85 QHSFSSPYT 1590
CDR-L3 BP18b-6 MQALQIPRT 1591
CDR-L3 BPx-86 MQGSHWPLT 1592
CDR-L3 BP19b-3 GSYTRSSTAR 1593 CDR-L3 BPx-87 QAWDSrrAV 1594
CDR-L3 BPx-88 QTWDGNHVV 1595
CDR-L3 BP18b-7 MQGTQWPYT 1596
CDR-L3 BP17b-l SSYTSSSTLV 1597
CDR-L3 BP16b-3 QQRSGWPPKVT 1598
CDR-L3 BPx-90 QAWDSSTVV 1599
CDR-L3 BP5b-8 QVWDSNSDHVV 1600
CDR-L3 BP16b-4 SSYTSSSTVV 1601
CDR-L3 BP9b-l ATWDDSLNGW 1602
CDR-L3 BPx-91 MQGLQTPYT 1603
CDR-L3 BP12b-l AAWDDSLNGWV 1604
CDR-L3 BPlOb-1 GTWDDRLSGVV 1605
CDR-L3 BPx-94 MQALQTPFT 1606
CDR-L3 BPx-96 SSYSTSSTFEV 1607
CDR-L3 BP6b-9 QQSYSTPWT 1608
CDR-L3 BPx-97 LQDYTTPYT 1609
CDR-L3 BPx-98 EAWDDSLSAVV 1610
CDR-L3 BP6b-8 QSYDSSLSDSVV 1611
CDR-L3 BPx-101 QQYYSTPLT 1612
CDR-L3 BPx-102 MQGTRWPYT 1613
CDR-L3 BPx-103 QSYDSVYNWI 1614
CDR-L3 BPx-104 AVWDDSLNGWV 1615
CDR-L3 BPx-106 QQYESYQPPT 1616
CDR-L3 BP5b-9 QQSQSTPVT 1617
CDR-L3 BPx-107 QQRSNWPRALS 1618
CDR-L3 BPx-108 SSYTSSNTWV 1619
CDR-L3 BPx-109 QQYYSTPYT 1620
CDR-L3 BP22b-l QQYYSTPS 1621
CDR-L3 BPx-110 QSYNTSNLV 1622
CDR-L3 BP6b-12 QQYQESPWT 1623
CDR-L3 BPx-112 QVWESSSDEWDTTSDQWV 1624
CDR-L3 BPx-114 QQYYGIPT 1625
CDR-L3 BPx-113 QSYDSAHHWV 1626
CDR-L3 BP19a-l AAWDDSLNGWV 1655
CDR-L3 BP6a-15 QQSYSTPWT 1656
CDR-L3 BP6a-21 QQSYSTPYT 1657
CDR-L3 BP6a-20 QQSYSTPYT 1658
CDR-L3 BP6a-24 QAWDGTKVV 1659
CDR-L3 BP6a-3 QAWDSTTVV 1660
CDR-L3 BP5b-6 SYTSSSTWV 1661
CDR-L3 BP16b-l AAWDDSLNGWV 1662
CDR-L3 BP8b-l; GTWDTSLSGFL 1669
CDR-L3 BP6b-5 GTWDTSLSGFL 1670
GDF-11 modulatory antibodies
[00312] GDF-8 and GDF-11 share considerable homology. While the prodomains only share 48% homology, GDF-8 and GDF-11 growth factor domains share 90% homology (60% homology when prodomains and growth factors are taken together). Given the high degree of sequence similarity, it is not surprising that GDF- 11 and 8 bind and signal through the same receptors consisting of a Type I receptor (ALK4/5) in association with a type II receptor (ACTRIIA/B). The high degree of conservation in the mature growth factors has made it challenging to identify reagents and monoclonal antibodies that can differentiate between mature GDF-11 and 8. Consequently, there are no therapies in clinical trials today that are specific for GDF- 11.
[00313] In some embodiments, the present disclosure provides GDF-11 -modulatory antibodies as well as methods of developing and identifying such antibodies. In some cases, GDF- modulatory antibodies specifically recognize the prodomain of GDF-11, but do not cross-react with the GDF-8 prodomain. Such antibodies may be functionally assessed to determine if they block or activate the release of the GDF-11 mature growth factor and can be further characterized in animal models to evaluate the effects of modulating specific growth factor levels in disease-relevant models.
[00314] Like other members of the TGF-β superfamily, GDF-11 and 8 are both initially expressed as inactive precursor polypeptides (termed proGDF-8 and proGDF-11). For GDF- 11 and 8, activation and release of the mature growth factor is accomplished by discrete protease cleavage events. The first cleavage step of proGDF-8 and proGDF-11 is carried out by a proprotein convertase, which cuts at a conserved RXXR site between the prodomain and mature growth factor. This cleavage produces a latent complex, in which the mature growth factor is shielded from binding to its receptors by the prodomain (see FIG. 9). Activation and release of the mature, active GDF- 11 growth factor is accomplished after cleavage by an additional protease from the BMP/Tolloid family.
[00315] In contrast to myostatin that has a well-defined role as a negative regulator of skeletal muscle mass, much less is known about the physiological roles of GDF-11. During the last year, several groups have revealed exciting new biology for GDF-11. Using parabiotic surgical techniques, blood systems of old and young mice were connected to look at the effects of circulating factors (Loffredo et al., 2013. Cell. 153:828-39). After 4 weeks of shared blood circulation, old mice had dramatically improved reversal of age-related cardiac hypertrophy. Additional studies suggested GDF- 11 was the factor that was responsible for the rejuvenating effects. Subsequently, follow up studies showed that GDF-11 levels decrease with age and if systemic levels are restored, functional impairments in skeletal muscle due to the aging process can be reversed (Sinha, M. et al., 2014. Science Express.
10.1126/science.1251152, p2-6). Additional improvements in the vasculature of the neurogenic niche were improved by the addition of GDF- 11 suggesting that increasing GDF- 11 levels could be beneficial in treating neurodegenerative and neurovascular diseases.
(Katsimpardi, L. et al., 2014. Science Express. 10.1126/science.1251141)
[00316] Other groups have recently published convincing data that GDF-11 is a novel negative regulator of late stage erythropoiesis (Carrancio, S. et al., 2014. Br J Haematol. 165(6):870-82 and Suragani, R.N.V.S. et al. 2014. Blood. 123(25): 3864-72, the contents of each of which are herein incorporated by reference in their entirety). These studies have led to the testing of two TGF-β superfamily ligand traps, ACE-011, an ACTRIIa-Fc fusion protein, and ACE-536, an ACTRIIb-Fc fusion protein, in the clinic for the treatment of anemias such as anemia associated with cancer and β-thalassemia. Although GDF- 11 was shown to be the growth factor responsible for inhibiting late-stage erythropoiesis, neither of these fusion proteins is specific for GDF- 11 as they bind to multiple other members of the TGF-β superfamily.
[00317] The new data implicating GDF- 11 in reversing aging contradicts what has previously been established in the literature for GDF-8 and GDF-11. Moreover, the studies implicating GDF-11 in erythropoiesis are still in very early stages. None of the current experimental approaches differentiate GDF- 11 from GDF-8 leaving the question how GDF- 11 can be distinguished from GDF-8 at the level of receptor binding and downstream signaling unanswered. There are no molecular tools that can differentiate among different forms of circulating GDF- 11 precursor, latent and mature forms so it is unknown what the relative circulating and tissue-bound levels of GDF- 11 are in the body.
[00318] In some embodiments, GDF-11 -modulatory antibodies of the disclosure may target the GDF- 11 prodomain, and therefore the activation mechanism of GDF- 11. Such antibodies may achieve specificity and safety that cannot be obtained by currently available methods. In some cases, GDF- 11 modulatory antibodies may also be useful as specific tools to interrogate the role of GDF- 11 in vivo and importantly allow for the development of therapeutic antibodies that may either block or activate GDF- 11 release and that may be evaluated in clinical trials.
Development of cytotoxic antibodies
[00319] In some embodiments, antibodies of the present disclosure may be capable of inducing antibody-dependent cell-mediated cytotoxicity (ADCC), complement-dependent cytotoxicity (CDC) and/or antibody-dependent cell phagocytosis (ADCP). ADCC is an immune mechanism whereby cells are lysed as a result of immune cell attack. Such immune cells may include CD56+ cells, CD3- natural killer (NK) cells, monocytes and neutrophils (Strohl, W.R. Therapeutic Antibody Engineering. Woodhead Publishing, Philadelphia PA. 2012. Ch. 8, pi 86, the contents of which are herein incorporated by reference in their entirety).
[00320] In some cases, antibodies of the present disclosure may be engineered to comprise a given isotype depending on whether or not ADCC or ADCP is desired upon antibody binding. Such antibodies, for example, may be engineered according to any of the methods disclosed by Alderson, K.L. et al., J Biomed Biotechnol. 2011. 2011:379123). In the case of mouse antibodies, different isotypes of antibodies are more effective at promoting ADCC. IgG2a, for example, is more effective at inducing ADCC than is IgG2b. Some antibodies of the present disclosure, comprising mouse IgG2b antibodies may be reengineered to comprise IgG2a antibodies. Such reengineered antibodies may be more effective at inducing ADCC upon binding cell- associated antigens.
[00321] In some embodiments, genes encoding variable regions of antibodies developed according to methods of the present disclosure may be cloned into mammalian expression vectors encoding human Fc regions. Such Fc regions may comprise Fc regions from human IgGlK. IgGlK Fc regions may comprise amino acid mutations known to enhance Fc-receptor binding and antibody-dependent cell-mediated cytotoxicity ADCC.
[00322] In some cases, antibodies may be engineered to reduce ADCC. Antibodies that do not activate ADCC or that are associated with reduced levels of ADCC may be desireable for antibody embodiments of the present disclosure, in some cases due to no or limited immune- mediated clearance, allowing longer half-lives in circulation.
Antibody fragment display library screening techniques
[00323] In some embodiments, antibodies of the present disclosure may be produced and/or optimized using high throughput methods of discovery. Such methods may include any of the display techniques (e.g. display library screening techniques) disclosed in International Patent Application No. WO2014074532, the contents of which are herein incorporated by reference in their entirety. In some embodiments, synthetic antibodies may be designed, selected or optimized by screening target antigens using display technologies (e.g. phage display technologies). Phage display libraries may comprise millions to billions of phage particles, each expressing unique antibody fragments on their viral coats. Such libraries may provide richly diverse resources that may be used to select potentially hundreds of antibody fragments with diverse levels of affinity for one or more antigens of interest (McCafferty, et al., 1990. Nature. 348:552-4; Edwards, B.M. et al., 2003. JMB. 334: 103-18; Schofield, D. et al., 2007. Genome Biol. 8, R254 and Pershad, K. et al., 2010. Protein Engineering Design and
Selection. 23:279-88; the contents of each of which are herein incorporated by reference in their entirety). Often, the antibody fragments present in such libraries comprise scFv antibody fragments, comprising a fusion protein of VH and VL antibody domains joined by a flexible linker (e.g. a Ser/Gly-rich linker). In some cases, scFvs may contain the same sequence with the exception of unique sequences encoding variable loops of the complementarity determining regions (CDRs). In some cases, scFvs are expressed as fusion proteins, linked to viral coat proteins (e.g. the N-terminus of the viral pill coat protein). VL chains may be expressed separately for assembly with VH chains in the periplasm prior to complex incorporation into viral coats.
[00324] Phage selection according to the present disclosure may include the use of the antibody display library described in Schofield, D. et al., 2007. Genome Biol. 8, R254 and Pershad, K. et al., 2010. Protein Engineering Design and Selection. 23:279-88, the contents of which are herein incorporated by reference in their entirety. This library included over 1010 clones and has been validated through the successful generation of antibodies to over 300 antigens, producing more than 7,500 distinct antibody clones. Further, antibody production using this library may be carried out as described in Falk, R. et al., 2012. Methods. 58: 69-78 and/or Melidoni et al., 2013. PNAS 110(44): 17802-7, the contents of each of which are herein incorporated by reference in their entirety.
[00325] For selection, target antigens may be incubated, in vitro, with phage display library particles for precipitation of positive binding partners. This process is referred to herein as "phage enrichment." In some cases, phage enrichment comprises solid-phase phage enrichment. According to such enrichment, target antigens are bound to a substrate (e.g. by passive adsorption) and contacted with one or more solutions comprising phage particles. Phage particles with affinity for such target antigens are precipitated out of solution. In some cases, phage enrichment comprises solution-phase phage enrichment where target antigens are present in a solution that is combined with phage solutions. According to such methods, target antigens may comprise detectable labels (e.g. biotin labels) to facilitate retrieval from solution and recovery of bound phage. In other embodiments, solution-phase phage enrichment may comprise the use of antigens bound to beads (e.g. streptavidin beads). In some cases, such beads may be magnetic beads to facilitate precipitation.
[00326] In some embodiments, phage enrichment may comprise solid-phase enrichment where target antigens are immobilized on solid surface. According to such methods, phage solutions may be used to contact the solid surface for enrichment with the immobilized antigens. Solid surfaces may include any surfaces capable of retaining antigens and may include, but are not limited to dishes, plates, flasks and tubes. In some cases, immunotubes may be used wherein the inner surface of such tubes may be coated with antigens. Phage enrichment with immunotubes may be carried out by passage of phage solution through the tubes to enrich bound antigens.
[00327] After selection, bound phage may be used to infect E. coli cultures that are co- infected with helper phage, to produce an amplified output library for the next round of enrichment. This process may be repeated producing narrower and narrower clone sets. In some embodiments, rounds of enrichment are limited to improve the diversity of selected phage.
[00328] Precipitated library members may be sequenced from the bound phage to obtain cDNA encoding desired scFvs. Such sequences may be directly incorporated into antibody sequences for recombinant antibody production, or mutated and utilized for further optimization through in vitro affinity maturation.
[00329] IgG antibodies comprising one or more variable domains from selected scFvs may be synthesized for further testing and/or product development. Such antibodies may be produced by insertion of one or more segments of scFv cDNA into expression vectors suited for IgG production. Expression vectors may comprise mammalian expression vectors suitable for IgG expression in mammalian cells. Mammalian expression of IgGs may be carried out to ensure that antibodies produced comprise modifications (e.g. glycosylation) characteristic of mammalian proteins and/or to ensure that antibody preparations lack endotoxin and/or other contaminants that may be present in protein preparations from bacterial expression systems.
[00330] In some embodiments, scFvs developed according to the disclosure may be expressed as scFv-Fc fusion proteins, comprising an antibody Fc domain. Such scFvs may be useful for further screening and analysis of scFv binding and affinity.
[00331] In some cases phage display screening may be used to generate broadly diverse panels of antibodies. Such diversity may be measured by diversity of antibody sequences and/or diversity of epitopes targeted.
[00332] Affinity binding estimates may be made using cross blocking experiments to bin antibodies. In some cases, affinity analysis instruments may be used. Such instruments may include, but are not limited surface plasmon resonance instrumentation, including, but not limited to Octet® (ForteBio, Menlo Park, CA). [00333] In some cases, epitope binning may be carried out to identify groups of antibodies binding distinct epitopes present on the same antigen. Such binning may be informed by data obtained from affinity analysis using cross blocking experiments and/or affinity analysis instrumentation.
Affinity maturation techniques
[00334] Affinity maturation techniques of the present disclosure may comprise any of those disclosed in International Patent Application No. WO2014074532, the contents of which are herein incorporated by reference in their entirety. After antibody fragments capable of binding target antigens are identified (e.g. through the use of phage display libraries as described above), high affinity mutants may be derived from these through the process of affinity maturation. Affinity maturation technology is used to identify sequences encoding CDRs that have the highest affinity for target antigens. Using such technologies, select CDR sequences (e.g. ones that have been isolated or produced according to processes described herein) may be mutated randomly as a whole or at specific residues to create millions to billions of variants. Such variants may be subjected to repeated rounds of affinity screening (e.g. display library screening) for their ability to bind target antigens. Such repeated rounds of selection, mutation and expression may be carried out to identify antibody fragment sequences with the highest affinity for target antigens. Such sequences may be directly incorporated into antibody sequences for recombinant antibody production.
Antibody characterization
[00335] Compounds and/or compositions of the present disclosure comprising antibodies may act to decrease local concentration of one or more GPC through removal by
phagocytosis, pinocytosis, or inhibiting assembly in the extracellular matrix and/or cellular matrix. Introduction of compounds and/or compositions of the present disclosure may lead to the removal of 5% to 100% of the growth factor present in a given area. For example, the percent of growth factor removal may be from about 5% to about 10%, from about 5% to about 15%, from about 5% to about 20%, from about 5% to about 25%, from about 10% to about 30%, from about 10% to about 40%, from about 10% to about 50%, from about 10% to about 60%, from about 20% to about 70%, from about 20% to about 80%, from about 40% to about 90% or from about 40% to about 100%.
[00336] Measures of release, inhibition or removal of one or more growth factors may be made relative to a standard or to the natural release or activity of growth factor under normal physiologic conditions, in vitro or in vivo. Measurements may also be made relative to the presence or absence of antibodies. Such methods of measuring growth factor levels, release, inhibition or removal include standard measurement in tissue and/or fluids (e.g. serum or blood) such as Western blot, enzyme-linked immunosorbent assay (ELISA), activity assays, reporter assays, luciferase assays, polymerase chain reaction (PCR) arrays, gene arrays, Real Time reverse transcriptase (RT) PCR and the like.
[00337] Antibodies of the present disclosure may bind or interact with any number of epitopes on or along GPCs or their associated structures to either enhance or inhibit growth factor signaling. Such epitopes may include any and all possible sites for altering, enhancing or inhibiting GPC function. In some embodiments, such epitopes include, but are not limited to epitopes on or within growth factors, regulatory elements, GPCs, GPC modulatory factors, growth factor receiving cells or receptors, LAPs or LAP-like domains, fastener regions, furin cleavage sites, arm regions, fingers regions, LTBP binding domains, fibrillin binding domains, glycoprotein A repetitions predominant (GARP) binding domains, latency lassos, alpha 1 regions, RGD sequences, bowtie regions, extracellular matrix and/or cellular matrix components and/or epitopes formed by combining regions or portions of any of the foregoing.
[00338] Compounds and/or compositions of the present disclosure exert their effects via binding (reversibly or irreversibly) to one or more epitopes and/or regions of antibody recognition. While not wishing to be bound by theory, such binding sites for antibodies, are most often formed by proteins, protein domains or regions. Binding sites may; however, include biomolecules such as sugars, lipids, nucleic acid molecules or any other form of binding epitope.
[00339] In some embodiments, antagonist antibodies of the present disclosure may bind to GDF prodomains, stabilizing and preventing growth factor release, for example, by blocking an enzymatic cleavage site or by stabilizing the structure. Such antibodies would be useful in the treatment of GDF-related indications resulting from excessive GDF activity.
[00340] Alternatively or additionally, antibodies of the present disclosure may function as ligand mimetics which would induce internalization of GPCs. Such antibodies may act as nontraditional payload carriers, acting to deliver and/or ferry bound or conjugated drug payloads to specific GPC and/or GPC -related sites.
[00341] Changes elicited by antibodies of the present disclosure may result in neomorphic changes in the cell. As used herein, the term "neomorphic change" refers to a change or alteration that is new or different. For example, an antibody that elicits the release or stabilization of one or more growth factor not typically associated with a particular GPC targeted by the antibody, would be a neomorphic antibody and the release would be a neomorphic change.
[00342] In some embodiments, compounds and/or compositions of the present disclosure may act to alter and/or control proteolytic events. In some embodiments, such proteolytic events may be intracellular or extracellular. In some embodiments, such proteolytic events may include the alteration of furin cleavage and/or other proteolytic processing events. In some embodiments, such proteolytic events may comprise proteolytic processing of growth factor signaling molecules or downstream cascades initiated by growth factor signaling molecules.
[00343] In some embodiments, compounds and/or compositions of the present disclosure may induce or inhibit dimerization or multimerization of growth factors (ligands) or their receptors. In some embodiments, such actions may be through stabilization of monomeric, dimeric or multimeric forms or through the disruption of dimeric or multimeric complexes.
[00344] In some embodiments, compounds and/or compositions of the present disclosure may act on homo and/or heterodimers of the monomeric units comprising either receptor groups or GPCs or other signaling molecule pairs.
[00345] Antibodies of the present disclosure may be internalized into cells prior to binding target antigens. Upon internalization, such antibodies may act to increase or decrease one or more signaling events, release or stabilize one or more GPCs, block or facilitate growth factor release and/or alter one or more cell niche.
[00346] In some embodiments, compounds and/or compositions of the present disclosure may also alter the residence time of one or more growth factor in one or more GPC and/or alter the residence time of one or more GPC in the extracellular matrix and/or cellular matrix. Such alterations may result in irreversible localization and/or transient localization.
[00347] Antibodies of the present disclosure may be designed, manufactured and/or selected using any methods known to one of skill in the art. In some embodiments, antibodies and/or antibody producing cells of the present disclosure are produced according to any of the methods listed in International Patent Application No. WO2014074532, the contents of which are herein incorporated by reference in their entirety.
Antibody generation in knockout mice
[00348] In some embodiments, antibodies of the current disclosure may be generated in knockout mice that lack a gene encoding one or more desired antigens. Such mice would not be tolerized to such antigens and therefore may be able to generate antibodies against them that could cross react with human and mouse forms of the antigen. For the production of monoclonal antibodies, host mice are immunized with the target peptide to elicit lymphocytes that specifically bind that peptide. Lymphocytes are collected and fused with an immortalized cell line. The resulting hybridoma cells are cultured in a suitable culture medium with a selection agent to support the growth of only the fused cells.
[00349] In some embodiments, knocking out one or more growth factor gene may be lethal and/or produce a fetus or neonate that is non- viable. In some embodiments, neonatal animals may only survive for a matter of weeks (e.g. 1, 2, 3, 4 or 5 weeks). In such embodiments, immunizations may be carried out in neonatal animals shortly after birth. Oida et al (Oida, T. et al., TGF-β induces surface LAP expression on Murine CD4 T cells independent of FoxP3 induction. PLOS One. 2010. 5(l l):el5523) demonstrate immunization of neonatal TGF-β knockout mice through the use of galectin-1 injections to prolong survival (typically 3-4 weeks after birth in these mice). Mice were immunized with cells expressing murine TGF-β every other day for 10 days beginning on the 8th day after birth and spleen cells were harvested on day 22 after birth. Harvested spleen cells were fused with myeloma cells and of the resulting hybridoma cells, many were found to successfully produce anti-LAP antibodies. In some embodiments of the present disclosure, these methods may be used to generate antibodies. In some embodiments, such methods may comprise the use of human antigens.
[00350] Methods of the present disclosure may also comprise one or more steps of the immunization methods described by Oida et al combined with one or more additional and/or modified steps. Modified steps may include, but are not limited to the use of alternate cell types for fusions, the pooling of varying number of spleen cells when performing fusions, altering the injection regimen, altering the date of spleen cell harvest, altering immunogen and/or altering immunogen dose. Additional steps may include the harvesting of other tissues (e.g. lymph nodes) from immunized mice.
Activating and inhibiting antibodies
[00351] Antibodies of the present disclosure may comprise activating or inhibiting antibodies. As used herein, the term "activating antibody" refers to an antibody that promotes growth factor activity. Activating antibodies include antibodies targeting any epitope that promotes growth factor activity. Such epitopes may lie on prodomains (e.g. LAPs and LAP- like domains), growth factors or other epitopes that when bound by antibody, lead to growth factor activity. Activating antibodies of the present disclosure may include, but are not limited to GDF- activating antibodies such as GDF- 8 -activating antibodies and GDF- 11- activating antibodies.
[00352] As used herein, the term "inhibiting antibody" refers to an antibody that reduces growth factor activity. Inhibiting antibodies include antibodies targeting any epitope that reduces growth factor activity when associated with such antibodies. Such epitopes may lie on prodomains (e.g. LAPs and LAP-like domains), growth factors or other epitopes that lead to reduced growth factor activity when bound by antibody. Inhibiting antibodies of the present disclosure may include, but are not limited to GDF-inhibiting antibodies such as GDF-8-inhibiting antibodies and GDF- 11 -inhibiting antibodies.
[00353] In some embodiments, inhibiting antibodies of the disclosure may include anti- primed complex antibodies. Such antibodies may target GDF-8 primed complexes and/or GDF- 11 primed complexes and block resulting growth factor activity. In some cases, anti- primed complex antibodies may prevent dissociation of bound prodomain upon receptor binding. In some cases, anti-primed complex antibodies may prevent primed complexes from binding to receptors. In some cases, anti-primed complex antibodies may prevent primed complexes from associating with one or more other factors, leading to modulation of growth factor activity.
[00354] Embodiments of the present disclosure include methods of using activating and/or inhibiting antibodies in solution, in cell culture and/or in subjects to modify growth factor signaling.
Anti-pro domain antibodies
[00355] In some embodiments, compounds and/or compositions of the present disclosure may comprise one or more antibody targeting a prodomain, including LAP and/or LAP-like domains. Such antibodies may reduce or elevate growth factor signaling depending on the specific prodomain that is bound and/or depending on the specific epitope targeted by such antibodies. Anti-LAP and/or anti-LAP-like protein antibodies of the disclosure may promote dissociation of free growth factors from GPCs. Such dissociation may be induced upon antibody binding to a GPC or dissociation may be promoted by preventing the reassociation of free growth factor with LAP or LAP-like protein. In some cases, anti-GDF prodomain antibodies are provided. Anti-GDF prodomain antibodies may comprise GDF- activating antibodies. Such antibodies may increase GDF activity (e.g. GDF-8 or GDF- 11 activity), in some cases by releasing GDF free growth factor from latent GPCs and/or preventing the reassociation of free growth factors with prodomains. In some cases, anti-GDF prodomain antibodies may increase GDF activity more favorably when a proGDF is associated with an extracellular protein (e.g. LTBP, fibrillin or a GASP protein).
Multispecific antibodies
[00356] In some embodiments, antibodies of the disclosure may be capable of binding more than one epitope. As used herein, the terms "multibody" or "multispecific antibody" refer to an antibody wherein two or more variable regions bind to different epitopes. The epitopes may be on the same or different targets. In certain embodiments, a multi-specific antibody is a "bispecific antibody," which recognizes two different epitopes on the same or different antigens.
Bispecific antibodies
[00357] In some cases, antibodies of the present disclosure may be bispecific antibodies. Bispecific antibodies are capable of binding two different antigens. Such antibodies typically comprise antigen-binding regions from at least two different antibodies. For example, a bispecific monoclonal antibody (BsMAb, BsAb) is an artificial protein composed of fragments of two different monoclonal antibodies, thus allowing the BsAb to bind to two different types of antigen.
[00358] Bispecific antibodies may include any of those described in Riethmuller, G., 2012. Cancer Immunity. 12: 12-18; Marvin, J.S. et al., 2005. Acta Pharmacologica Sinica.
26(6):649-58; and Schaefer, W. et al., 2011. PNAS. 108(27): 11187-92, the contents of each of which are herein incorporated by reference in their entirety.
[00359] New generations of BsMAb, called "trifunctional bispecific" antibodies, have been developed. These consist of two heavy and two light chains, one each from two different antibodies, where the two Fab regions (the arms) are directed against two antigens, and the Fc region (the foot) comprises the two heavy chains and forms the third binding site.
[00360] Of the two paratopes that form the tops of the variable domains of a bispecific antibody, one can be directed against a target antigen and the other against a T-lymphocyte antigen like CD3. In the case of trifunctional antibodies, the Fc region may additionally binds to a cell that expresses Fc receptors, like a mactrophage, a natural killer (NK) cell or a dendritic cell. In sum, the targeted cell is connected to one or two cells of the immune system, which subsequently destroy it.
[00361] Other types of bispecific antibodies have been designed to overcome certain problems, such as short half-life, immunogenicity and side-effects caused by cytokine liberation. They include chemically linked Fabs, consisting only of the Fab regions, and various types of bivalent and trivalent single-chain variable fragments (scFvs), fusion proteins mimicking the variable domains of two antibodies. The furthest developed of these newer formats are the bi-specific T-cell engagers (BiTEs) and mAb2's, antibodies engineered to contain an Fcab antigen-binding fragment instead of the Fc constant region.
[00362] A bispecific, single-chain antibody Fv fragment (Bs-scFv) was successfully used to kill cancer cells. Some human cancers are caused by functional defects in p53 that are restored by gene therapy with wild-type p53. Weisbart, et al, describe the construction and expression of a bispecific single-chain antibody that penetrates living colon cancer cells, binds intracellular p53, and targets and restores its wild type function (Weisbart, et al., Int. J. Oncol. 2004 Oct;25(4): 1113-8; and Weisbart, et al, Int. J. Oncol. 2004 Dec;25(6): 1867-73). In these studies, a bispecific, single-chain antibody Fv fragment (Bs-scFv) was constructed from (i) a single-chain Fv fragment of mAb 3E10 that penetrates living cells and localizes in the nucleus, and (ii) a single-chain Fv fragment of a non-penetrating antibody, mAb PAb421 that binds the C-terminal of p53. PAb421 binding restores wild- type functions of some p53 mutants, including those of SW480 human colon cancer cells. The Bs-scFv penetrated SW480 cells and was cytotoxic, suggesting an ability to restore activity to mutant p53. COS- 7 cells (monkey kidney cells with wild-type p53) served as a control since they are unresponsive to PAb421 due to the presence of SV40 large T antigen that inhibits binding of PAb421 to p53. Bs-scFv penetrated COS-7 cells but was not cytotoxic, thereby eliminating non-specific toxicity of Bs-scFv unrelated to binding p53. Fv fragments alone were not cytotoxic, indicating that killing was due to transduction of p53. A single mutation in CDR1 of PAb421 VH eliminated binding of the Bs-scFv to p53 and abrogated cytotoxicity for SW480 cells without altering cellular penetration, further supporting the requirement of PAb421 binding to p53 for cytotoxicity (Weisbart, et al, Int. J. Oncol. 2004 Oct;25(4): 1113- 8; and Weisbart, et al, Int. J. Oncol. 2004 Dec;25(6): 1867-73).
[00363] In some embodiments, antibodies of the present disclosure may be diabodies. Diabodies are functional bispecific single-chain antibodies (bscAb). These bivalent antigen- binding molecules are composed of non-covalent dimers of scFvs, and can be produced in mammalian cells using recombinant methods. {See, e.g., Mack et al, Proc. Natl. Acad. Scl, 92: 7021-7025, 1995). Few diabodies have entered clinical development. An iodine- 123- labeled diabody version of the anti-CEA chimeric antibody cT84.66 has been evaluated for pre-surgical immuno scintigraphic detection of colorectal cancer in a study sponsored by the Beckman Research Institute of the City of Hope (Clinicaltrials.gov NCT00647153) (Nelson, A. L., MAbs.2010. Jan-Feb; 2(l):77-83).
[00364] Using molecular genetics, two scFvs can be engineered in tandem into a single polypeptide, separated by a linker domain, called a "tandem scFv" (tascFv). TascFvs have been found to be poorly soluble and require refolding when produced in bacteria, or they may be manufactured in mammalian cell culture systems, which avoids refolding requirements but may result in poor yields. Construction of a tascFv with genes for two different scFvs yields a "bispecific single-chain variable fragments" (bis-scFvs). Only two tascFvs have been developed clinically by commercial firms; both are bispecific agents in active early phase development by Micromet for oncologic indications, and are described as "Bispecific T-cell Engagers (BiTE)." Blinatumomab is an anti-CD 19/anti-CD3 bispecific tascFv that potentiates T-cell responses to B-cell non-Hodgkin lymphoma in Phase 2. MT110 is an anti-EP- CAM/anti-CD3 bispecific tascFv that potentiates T-cell responses to solid tumors in Phase 1. Bispecific, tetravalent "TandAbs" are also being researched by Affimed (Nelson, A. L., MAbs.2010. Jan-Feb; 2(l):77-83).
[00365] Also included are maxibodies (bivalent scFV fused to the amino terminus of the Fc (CH2-CH3 domains) of IgG.
[00366] Bispecific T-cell-engager (BiTE) antibodies are designed to transiently engage cytotoxic T-cells for lysis of selected target cells. The clinical activity of BiTE antibodies corroborates findings that ex vivo expanded, autologous T-cells derived from tumor tissue, or transfected with specific T-cell receptors, have shown therapeutic potential in the treatment of solid tumors. While these personalized approaches prove that T-cells alone can have considerable therapeutic activity, even in late-stage cancer, they are cumbersome to perform on a broad basis. This is different for cytotoxic T-lymphocyte antigen 4 (CTLA-4) antibodies, which facilitate generation of tumor- specific T-cell clones, and also for bi- and tri-specific antibodies that directly engage a large proportion of patients' T-cells for cancer cell lysis. The potential of global T-cell engagement for human cancer therapy by T-cell-engaging antibodies is under active investigation (Baeuerle PA, et al., Current Opinion in Molecular Therapeutics. 2009, l l(l):22-30).
[00367] Third generation molecules include "miniaturized" antibodies. Among the best examples of mAb miniaturization are the small modular immunopharmaceuticals (SMJPs) from Trubion Pharmaceuticals. These molecules, which can be monovalent or bivalent, are recombinant single-chain molecules containing one VL, one VH antigen-binding domain, and one or two constant "effector" domains, all connected by linker domains. Presumably, such a molecule might offer the advantages of increased tissue or tumor penetration claimed by fragments while retaining the immune effector functions conferred by constant domains. At least three "miniaturized" SMIPs have entered clinical development. TRU-015, an anti-CD20 SMIP developed in collaboration with Wyeth, is the most advanced project, having progressed to Phase 2 for rheumatoid arthritis (RA). Earlier attempts in systemic lupus erythrematosus (SLE) and B cell lymphomas were ultimately discontinued. Trubion and Facet Biotechnology are collaborating in the development of TRU-016, an anti-CD37 SMIP, for the treatment of CLL and other lymphoid neoplasias, a project that has reached Phase 2. Wyeth has licensed the anti-CD20 SMIP SBI-087 for the treatment of autoimmune diseases, including RA, SLE and possibly multiple sclerosis, although these projects remain in the earliest stages of clinical testing. (Nelson, A. L., MAbs.2010. Jan-Feb; 2(l):77-83).
[00368] Genmab is researching application of their "Unibody" technology, in which the hinge region has been removed from IgG4 molecules. While IgG4 molecules are unstable and can exchange light-heavy chain heterodimers with one another, deletion of the hinge region prevents heavy chain-heavy chain pairing entirely, leaving highly specific monovalent light/heavy heterodimers, while retaining the Fc region to ensure stability and half-life in vivo. This configuration may minimize the risk of immune activation or oncogenic growth, as IgG4 interacts poorly with FcRs and monovalent unibodies fail to promoteintracellular signaling complex formation. These contentions are, however, largely supported by laboratory, rather than clinical, evidence. Biotecnol is also developing a "miniaturized" mAb, CAB051, which is a "compacted" 100 kDa anti-HER2 antibody in preclinical research (Nelson, A. L., MAbs.2010. Jan-Feb; 2(l):77-83).
[00369] Recombinant therapeutics composed of single antigen-binding domains have also been developed, although they currently account for only 4% of the clinical pipeline. These molecules are extremely small, with molecular weights approximately one-tenth of those observed for full-sized mAbs. Arana and Domantis engineer molecules composed of antigen- binding domains of human immunoglobulin light or heavy chains, although only Arana has a candidate in clinical testing, ART-621, an anti-TNFa molecule in Phase 2 study for the treatment of psoriasis and rheumatoid arthritis. Ablynx produces "nanobodies" derived from the antigen-binding variable heavy chain regions (VHHS) of heavy chain antibodies found in camels and llamas, which lack light chains. Two Ablynx anti-von Willebrand Factor nanobodies have advanced to clinical development, including ALX-0081, in Phase 2 development as an intravenous therapy to prevent thrombosis in patients undergoing percutaneous coronary intervention for acute coronary syndrome, and ALX-0681, a Phase 1 molecule for subcutaneous administration intended for both patients with acute coronary syndrome and thrombotic thrombocytopenic purpura (Nelson, A. L., MAbs.2010. Jan-Feb; 2(l):77-83).
Development of multispecific antibodies
[00370] In some embodiments, antibody sequences of the disclosure may be used to develop multispecific antibodies {e.g., bispecific, trispecific, or of greater multispecificity). Multispecific antibodies can be specific for different epitopes of a target antigen of the present disclosure, or can be specific for both a target antigen of the present disclosure, and a heterologous epitope, such as a heterologous glycan, peptide or solid support material. {See, e.g., WO 93/17715; WO 92/08802; WO 91/00360; WO 92/05793; Tutt, A. et al., Trispecific F(ab')3 derivatives that use cooperative signaling via the TCR/CD3 complex and CD2 to activate and redirect resting cytotoxic T cells. J. Immunol. 1991 Jul l;147(l):60-9; U.S. Pat. Nos. 4,474,893; 4,714,681; 4,925,648; 5,573,920; 5,601,819; and Kostelny, S.A. et al, Formation of a bispecific antibody by the use of leucine zippers. J. Immunol. 1992 Mar l;148(5): 1547-53); U.S. Pat. No. 5,932,448.
[00371] Disclosed and claimed in PCT Publication WO2014144573 to Memorial Sloan- Kettering Cancer Center are multimerization technologies for making dimeric multispecific binding agents {e.g., fusion proteins comprising antibody components) with improved properties over multispecific binding agents without the capability of dimerization.
[00372] Disclosed and claimed in PCT Publication WO2014144357 to Merck Patent GMBH are tetravalent bispecific antibodies (TetBiAbs), and methods of making and methods of using TetBiAbs for diagnostics and for the treatment of cancer or immune disorders.
TetBiAbs feature a second pair of Fab fragments with a second antigen specificity attached to the C-terminus of an antibody, thus providing a molecule that is bivalent for each of the two antigen specificities. The tetravalent antibody is produced by genetic engineering methods, by linking an antibody heavy chain covalently to a Fab light chain, which associates with its cognate, co-expressed Fab heavy chain.
[00373] Disclosed and claimed in PCT Publication WO2014028560 to IBC
Pharmaceuticals, Inc. are T cell redirecting bispecific antibodies (bsAb), with at least one binding site for a T-cell antigen and at least one binding site for an antigen on a diseased cell or pathogen, for treatment of disease. Preferably, this bsAb is an anti-CD3 x anti-CD 19 bispecific antibody, although antibodies against other T-cell antigens and/or disease- associated antigens may be used. The complex is capable of targeting effector T cells to induce T-cell-mediated cytotoxicity of cells associated with a disease, such as cancer, autoimmune disease or infectious disease. The cytotoxic immune response is enhanced by coadministration of interfon-based agents that comprise interferon-a, interferon-bgr; interferon- λΐ, interferon^ or interferon- λ3.
[00374] Disclosed and claimed in PCT Publication WO2013092001 to Synimmune GMBH is a bispecific antibody molecule, as well as a method for producing the same, its use and a nucleic acid molecule encoding the bispecific antibody molecule. In particular is provided an antibody molecule that is capable of mediating target cell restricted activation of immune cells.
[00375] Disclosed and claimed in PCT Publication WO2012007167 is a multispecific modular antibody specifically binding to at least a glycoepitope and a receptor of the erbB class on the surface of a tumor cell, thereby crosslinking the glycoepitope and the receptor, which antibody has apoptotic activity effecting cytolysis independent of NK cells.
[00376] Disclosed and claimed in PCT Publications WO2012048332 and WO2013055404 are meditopes, meditope-binding antibodies, meditope delivery systems, as well as a monoclonal antibody framework binding interface for meditopes, and methods for their use. Specifically, two antibody binding peptides, C-QFDLSTRRLK-C ("cQFD"; SEQ ID NO: l therein) and C-QYNLSSRALK-C ("cQYN"; SEQ ID NO:2 therein) were shown to have novel mAb binding properties. Also called "meditopes," cQFD and cQYN were shown to bind to a region of the Fab framework of the anti-EGFR mAb cetuximab and not to bind the complementarity determining regions (CDRs) that bind antigen. The binding region on the Fab framework is distinct from other framework-binding antigens, such as the superantigens Staphylococcal protein A (SpA) (Graille et al., 2000) and Peptostreptococcus magnus protein L (PpL) (Graille et al., 2001). Accordingly, one embodiment disclosed is a framework binding interface comprising a framework region of a unique murine-human antibody or functional fragment thereof that binds a cyclic meditope.
[00377] Exemplary patents and patent publications of interest are: U.S. Patent Nos.
5,585,089; 5,693,761; and 5,693,762, all filed Jun 7, 1995 and U.S. Patent No. 6,180,370, all assigned to Protein Design Labs, Inc., describe methods for producing, and compositions of, humanized immunoglobulins having one or more complementarity determining regions (CDR's) and possible additional amino acids from a donor immunoglobulin and a framework region from an accepting human immunoglobulin. Each humanized immunoglobulin chain is said to usually comprise, in addition to the CDR's, amino acids from the donor
immunoglobulin framework that are, e.g., capable of interacting with the CDRs to effect binding affinity, such as one or more amino acids which are immediately adjacent to a CDR in the donor immunoglobulin or those within about about 3 A as predicted by molecular modeling. The heavy and light chains may each be designed by using any one or all of various position criteria. When combined into an intact antibody, the humanized
immunoglobulins of the present disclosure is said to be substantially non-immunogenic in humans and retain substantially the same affinity as the donor immunoglobulin to the antigen, such as a protein or other compound containing an epitope.
[00378] U.S. Patent No. 5,951,983, assigned to Universite Catholique De Louvain and Bio Transplant, Inc., describes a humanized antibody against T-lymphocytes. Framework regions from a human V kappa gene designated as HUM5400 (EMBL accession X55400) and from the human antibody clone Amu 5-3 (GenBank accession number U00562) are set forth therein.
[00379] U.S. Patent No. 5,091,513, to Creative Biomolecules, Inc., describes a family of synthetic proteins having affinity for a preselected antigen. The proteins are characterized by one or more sequences of amino acids constituting a region which behaves as a biosynthetic antibody binding site (BABS). The sites comprise 1) non-covalently associated or disulfide bonded synthetic VH and VL dimers, 2) VH-VL or VL-VH single chains wherein the VH and VL are attached by a polypeptide linker, or 3) individuals VH or VL domains. The binding domains comprise linked CDR and FR regions, which may be derived from separate immunoglobulins. The proteins may also include other polypeptide sequences which function, e.g., as an enzyme, toxin, binding site, or site of attachment to an immobilization media or radioactive atom. Methods are disclosed for producing the proteins, for designing BABS having any specificity that can be elicited by in vivo generation of antibody, and for producing analogs thereof.
[00380] U.S. Patent No. 8,399,625, to ESBATech, an Alcon Biomedical Research Unit, LLC, describes antibody acceptor frameworks and methods for grafting non-human antibodies, e.g., rabbit antibodies, using a particularly well suited antibody acceptor framework.
Intrabodies
[00381] In some embodiments, antibodies of the present disclosure may be intrabodies. Intrabodies are a form of antibody that is not secreted from a cell in which it is produced, but instead targets one or more intracellular proteins. Intrabodies are expressed and function intracellularly, and may be used to affect a multitude of cellular processes including, but not limited to intracellular trafficking, transcription, translation, metabolic processes,
proliferative signaling and cell division. In some embodiments, methods described herein include intrabody-based therapies. In some such embodiments, variable domain sequences and/or CDR sequences disclosed herein are incorporated into one or more constructs for intrabody-based therapy. For example, intrabodies may target one or more glycated intracellular proteins or may modulate the interaction between one or more glycated intracellular proteins and an alternative protein.
[00382] More than two decades ago, intracellular antibodies against intracellular targets were first described (Biocca, Neuberger and Cattaneo EMBO J. 9: 101-108, 1990). The intracellular expression of intrabodies in different compartments of mammalian cells allows blocking or modulation of the function of endogenous molecules (Biocca, et al., EMBO J. 9: 101-108, 1990; Colby et al., Proc. Natl. Acad. Sci. U.S.A. 101 : 17616-21 , 2004). Intrabodies can alter protein folding, protein-protein, protein-DNA, protein-RNA interactions and protein modification. They can induce a phenotypic knockout and work as neutralizing agents by direct binding to the target antigen, by diverting its intracellular traffic or by inhibiting its association with binding partners. They have been largely employed as research tools and are emerging as therapeutic molecules for the treatment of human diseases as viral pathologies, cancer and misfolding diseases. The fast growing bio-market of recombinant antibodies provides intrabodies with enhanced binding specificity, stability and solubility, together with lower immunogenicity, for their use in therapy (Biocca, abstract in Antibody Expression and Production Cell Engineering Volume 7, 2011, pp. 179-195).
[00383] In some embodiments, intrabodies have advantages over interfering RNA (iRNA); for example, iRNA has been shown to exert multiple non-specific effects, whereas intrabodies have been shown to have high specificity and affinity of to target antigens.
Furthermore, as proteins, intrabodies possess a much longer active half-life than iRNA. Thus, when the active half-life of the intracellular target molecule is long, gene silencing through iRNA may be slow to yield an effect, whereas the effects of intrabody expression can be almost instantaneous. Lastly, it is possible to design intrabodies to block certain binding interactions of a particular target molecule, while sparing others.
Development of intrabodies
[00384] Intrabodies are often single chain variable fragments (scFvs) expressed from a recombinant nucleic acid molecule and engineered to be retained intracellularly (e.g., retained in the cytoplasm, endoplasmic reticulum, or periplasm). Intrabodies may be used, for example, to ablate the function of a protein to which the intrabody binds. The expression of intrabodies may also be regulated through the use of inducible promoters in the nucleic acid expression vector comprising the intrabody. Intrabodies may be produced using methods known in the art, such as those disclosed and reviewed in: (Marasco et al, 1993 Proc. Natl. Acad. Sci. USA, 90: 7889-7893; Chen et al, 1994, Hum. Gene Ther. 5:595-601; Chen et al, 1994, Proc. Natl. Acad. Sci. USA, 91: 5932-5936; Maciejewski et al, 1995, Nature Med., 1: 667-673; Marasco, 1995, Immunotech, 1: 1-19; Mhashilkar, et al, 1995, EMBO J. 14: 1542- 51; Chen et al, 1996, Hum. Gene Therap., 7: 1515-1525; Marasco, Gene Ther. 4: 11-15, 1997; Rondon and Marasco, 1997, Annu. Rev. Microbiol. 51:257-283; Cohen, et al, 1998, Oncogene 17:2445-56; Proba et al, 1998, J. Mol. Biol. 275:245-253; Cohen et al, 1998, Oncogene 17:2445-2456; Hassanzadeh, et al, 1998, FEBS Lett. 437:81-6; Richardson et al, 1998, Gene Ther. 5:635-44; Ohage and Steipe, 1999, J. Mol. Biol. 291: 1119-1128; Ohage et al, 1999, J. Mol. Biol. 291: 1129-1134; Wirtz and Steipe, 1999, Protein Sci. 8:2245-2250; Zhu et al, 1999, J. Immunol. Methods 231:207-222; Arafat et al, 2000, Cancer Gene Ther. 7: 1250-6; der Maur et al, 2002, J. Biol. Chem. 277:45075-85; Mhashilkar et al, 2002, Gene Ther. 9:307-19; and Wheeler et al, 2003, FASEB J. 17: 1733-5; and references cited therein). In particular, a CCR5 intrabody has been produced by Steinberger et al, 2000, Proc. Natl. Acad. Sci. USA 97:805-810). See generally Marasco, WA, 1998, "Intrabodies: Basic Research and Clinical Gene Therapy Applications" SpringenNew York; and for a review of scFvs, see Pluckthun in "The Pharmacology of Monoclonal Antibodies," 1994, vol. 113, Rosenburg and Moore eds. Springer- Verlag, New York, pp. 269-315.
[00385] In some embodiments, antibody sequences disclosed herein may be used to develop intrabodies. Intrabodies are often recombinantly expressed as single domain fragments such as isolated VH and VL domains or as a single chain variable fragment (scFv) antibody within the cell. For example, intrabodies are often expressed as a single polypeptide to form a single chain antibody comprising the variable domains of the heavy and light chain joined by a flexible linker polypeptide. Intrabodies typically lack disulfide bonds and are capable of modulating the expression or activity of target genes through their specific binding activity. Single chain antibodies can also be expressed as a single chain variable region fragment joined to the light chain constant region.
[00386] As is known in the art, an intrabody can be engineered into recombinant polynucleotide vectors to encode sub-cellular trafficking signals at its N or C terminus to allow expression at high concentrations in the sub-cellular compartments where a target protein is located. For example, intrabodies targeted to the endoplasmic reticulum (ER) are engineered to incorporate a leader peptide and, optionally, a C-terminal ER retention signal, such as the KDEL amino acid motif. Intrabodies intended to exert activity in the nucleus are engineered to include a nuclear localization signal. Lipid moieties are joined to intrabodies in order to tether the intrabody to the cytosolic side of the plasma membrane. Intrabodies can also be targeted to exert function in the cytosol. For example, cytosolic intrabodies are used to sequester factors within the cytosol, thereby preventing them from being transported to their natural cellular destination.
[00387] There are certain technical challenges with intrabody expression. In particular, protein conformational folding and structural stability of the newly- synthesized intrabody within the cell is affected by reducing conditions of the intracellular environment. In human clinical therapy, there are safety concerns surrounding the application of transfected recombinant DNA, which is used to achieve intrabody expression within the cell. Of particular concern are the various viral-based vectors commonly-used in genetic
manipulation. Thus, one approach to circumvent these problems is to fuse protein
transduction domains (PTD) to scFv antibodies, to create a 'cell-permeable' antibody or 'Transbody.' Transbodies are cell-permeable antibodies in which a protein transduction domain (PTD) is fused with single chain variable fragment (scFv) antibodies (Heng and Cao, 2005, Med Hypotheses. 64: 1105-8).
[00388] Upon interaction with a target gene, an intrabody modulates target protein function and/or achieves phenotypic/functional knockout by mechanisms such as accelerating target protein degradation and sequestering the target protein in a non-physiological sub-cellular compartment. Other mechanisms of intrabody-mediated gene inactivation can depend on the epitope to which the intrabody is directed, such as binding to the catalytic site on a target protein or to epitopes that are involved in protein-protein, protein-DNA, or protein-RNA interactions.
[00389] In one embodiment, intrabodies are used to capture a target in the nucleus, thereby preventing its activity within the nucleus. Nuclear targeting signals are engineered into such intrabodies in order to achieve the desired targeting. Such intrabodies are designed to bind specifically to a particular target domain. In another embodiment, cytosolic intrabodies that specifically bind to a target protein are used to prevent the target from gaining access to the nucleus, thereby preventing it from exerting any biological activity within the nucleus (e.g., preventing the target from forming transcription complexes with other factors).
[00390] In order to specifically direct the expression of such intrabodies to particular cells, the transcription of the intrabody is placed under the regulatory control of an appropriate tumor- specific promoter and/or enhancer. In order to target intrabody expression specifically to prostate, for example, the PSA promoter and/or promoter/enhancer can be utilized (See, for example, U.S. Patent No. 5,919,652 issued 6 July 1999).
[00391] Protein transduction domains (PTDs) are short peptide sequences that enable proteins to translocate across the cell membrane and be internalized within the cytosol, through atypical secretory and internalization pathways. There are a number of distinct advantages that a 'Transbody' would possess over conventional intrabodies expressed within the cell. For a start, 'correct' conformational folding and disulfide bond formation can take place prior to introduction into the target cell. More importantly, the use of cell-permeable antibodies or 'Transbodies' would avoid the overwhelming safety and ethical concerns surrounding the direct application of recombinant DNA technology in human clinical therapy, which is required for intrabody expression within the cell. 'Transbodies' introduced into the cell would possess only a limited active half-life, without resulting in any permanent genetic alteration. This would allay any safety concerns with regards to their application in human clinical therapy (Heng and Cao 2005, Med Hypotheses. 64: 1105-8).
[00392] Intrabodies are promising therapeutic agents for the treatment of misfolding diseases, including Alzheimer's, Parkinson's, Huntington's and prion diseases, because of their virtually infinite ability to specifically recognize the different conformations of a protein, including pathological isoforms, and because they can be targeted to the potential sites of aggregation (both intra- and extracellular sites). These molecules can work as neutralizing agents against amyloidogenic proteins by preventing their aggregation, and/or as molecular shunters of intracellular traffic by rerouting the protein from its potential aggregation site (Cardinale, and Biocca, Curr. Mol. Med. 2008, 8:2-11).
[00393] Exemplary Patent Publications describing intracellular antibodies or intrabodies are set forth hereinbelow, each of which is incorporated by reference in its entirety.
[00394] PCT Publication WO03014960 and US Patent 7,608,453 granted to Cattaneo, et al. , describe an intracellular antibody capture technology method of identifying at least one consensus sequence for an intracellular antibody (ICS) comprising the steps of: creating a database comprising sequences of validated intracellular antibodies (VIDA database) and aligning the sequences of validated intracellular antibodies according to Kabat; determining the frequency with which a particular amino acid occurs in each of the positions of the aligned antibodies; selecting a frequency threshold value (LP or consensus threshold) in the range from 70% to 100%; identifying the positions of the alignment at which the frequency of a particular amino acid is greater than or equal to the LP value; and identifying the most frequent amino acid, in the position of said alignment.
[00395] PCT Publications WO0054057; WO03077945; WO2004046185; WO2004046186; WO2004046187; WO2004046188; WO2004046189; US Patent Application Publications US2005272107; US2005276800; US2005288492; US2010143939; granted US Patents 7,569,390 and 7,897,347 and granted European Patents EP1560853; and EP1166121 all assigned to the Medical Research Council and including inventors Cattaneo, et al., describe intracellular intracellular single domain immunoglobulins, and a method for determining the ability of a immunoglobulin single domain to bind to a target in an intracellular environment, as well as methods for generating intracellular antibodies.
[00396] PCT Publication WO0235237; US Patent Application Publication 2003235850 and granted European Patent EP1328814 naming Catteneo as an inventor and assigned to
S.I.S.S.A. Scuola Internazionale Superiore describe a method for the in vivo identification of epitopes of an intracellular antigen.
[00397] PCT Publication WO2004046192 and European Patent EP1565558 assigned to Lay Line Genomics SPA and naming Catteneo as an inventor describe a method for isolating intracellular antibodies that disrupt and neutralize an interaction between a protein ligand x and a protein ligand y inside a cell. Also disclosed are a method to identify a protein ligand x able to bind to a known y ligand using intracellular antibodies able to the interaction between x and y; and a method for the isolation of a set of antibody fragments against a significant proportion of the protein-protein interactions of a given cell (interactome) or against the protein interactions that constitute an intracellular pathway or network.
[00398] US Patent Application Publication 2006034834 and PCT Publication W09914353 entitled "Intrabody-mediated control of immune reactions" and assigned to Dana Farber Cancer Institute Inc. name inventors Marasco and Mhashilkar are directed to methods of altering the regulation of the immune system, e.g., by selectively targeting individual or classes of immunomodulatory receptor molecules (IRMs) on cells comprising transducing the cells with an intracellularly expressed antibody, or intrabody, against the IRMs. In a preferred embodiment the intrabody comprises a single chain antibody against an IRM, e.g, MHC-1 molecules.
[00399] PCT Publication WO2013033420 assigned to Dana Farber Cancer Institute Inc. and Whitehead Biomedical Institute, and naming inventors Bradner, Rahl and Young describes methods and compositions useful for inhibiting interaction between a bromodomain protein and an immunoglobulin (Ig) regulatory element and downregulating expression of an oncogene translocated with an Ig locus, as well as for treating a cancer (e.g., hematological malignancy) characterized by increased expression of an oncogene which is translocated with an Ig locus. Intrabodies are generally described.
[00400] PCT Publication WO02086096 and US Patent Application Publication
2003104402 entitled "Methods of producing or identifying intrabodies in eukaryotic cells," assigned to University of Rochester Medical Center and naming inventors Zauderer, Wei and Smith describe a high efficiency method of expressing intracellular immunoglobulin molecules and intracellular immunoglobulin libraries in eukaryotic cells using a trimolecular recombination method. Further provided are methods of selecting and screening for intracellular immunoglobulin molecules and fragments thereof, and kits for producing, screening and selecting intracellular immunoglobulin molecules, as well as the intracellular immunoglobulin molecules and fragments produced using these methods.
[00401] PCT Publication WO2013023251 assigned to Affinity Biosciences PTY LTD and naming inventors Beasley, Niven and Kiefel describes polypeptides, such as antibody molecules and polynucleotides encoding such polypeptides, and libraries thereof, wherein the expressed polypeptides that demonstrate high stability and solubility. In particular, polypeptides comprising paired VL and VH domains that demonstrate soluble expression and folding in a reducing or intracellular environment are described, wherein a human scFv library was screened, resulting in the isolation of soluble scFv genes that have identical framework regions to the human germline sequence as well as remarkable thermostability and tolerance of CDR3 grafting onto the scFv scaffold.
[00402] European Patent Application EP2314622 and PCT Publications WO03008451 and WO03097697 assigned to Esbatech AG and University of Zuerich and naming inventors Ewert, Huber, Honneger and Plueckthun describe the modification of human variable domains and provide compositions useful as frameworks for the creation of very stable and soluble single-chain Fv antibody fragments. These frameworks have been selected for intracellular performance and are thus ideally suited for the creation of scFv antibody fragments or scFv antibody libraries for applications where stability and solubility are limiting factors for the performance of antibody fragments, such as in the reducing environment of a cell. Such frameworks can also be used to identify highly conserved residues and consensus sequences which demonstrate enhanced solubility and stability.
[00403] PCT Publication WO02067849 and US Patent Application Publication
2004047891 entitled "Systems devices and methods for intrabody targeted delivery and reloading of therapeutic agents" describe systems, devices and methods for intrabody targeted delivery of molecules. More particularly, some embodiments relate to a reloadable drug delivery system, which enables targeted delivery of therapeutic agents to a tissue region of a subject, in a localized and timely manner.
[00404] PCT Publication WO2005063817 and US Patent 7,884,054 assigned to Amgen Inc. and naming inventors Zhou, Shen and Martin describe methods for identifying functional antibodies, including intrabodies. In particular, a homodimeric intrabody is described, wherein each polypeptide chain of the homodimer comprises an Fc region, an scFv, and an intracellular localization sequence. The intracellular localization sequence may cause the intrabody to be localized to the ER or the Golgi. Optionally, each polypeptide chain comprises not more than one scFv.
[00405] PCT Publication WO2013138795 by Vogan, et al. and assigned to Permeon Biologies Inc. describes cell penetrating compositions for delivery of intracellular antibodies and antibody-like moieties and methods for delivering them (referred to herein as "AAM moieties" or "an AAM moiety") into a cell. Without being bound by theory, the present disclosure is based, at least in part, on the discovery that an AAM moiety can be delivered into a cell by complexing the AAM moiety with a cell penetrating polypeptide having surface positive charge (referred to herein as a "Surf+ Penetrating Polypeptide") . Examples of some applications of intraphilin technology are also provided
[00406] PCT Publication WO2010004432 assigned to the Pasteur Institute describes immunoglobulins from camelidae (camels, dromedaries, llamas and alpacas), about 50% of which are antibodies devoid of light chain. These heavy-chain antibodies interact with the antigen by the virtue of only one single variable domain, referred to as VHH(s), VHH domain(s) or VHH antibody (ies). Despite the absence of light chain, these homodimeric antibodies exhibit a broad antigen-binding repertoire by enlarging their hypervariable regions, and can act as a transbody and/or intrabody in vitro as well as in vivo, when the VHH domain is directed against an intracellular target.
[00407] PCT Publication WO2014106639 describes a method for identifying a cellular target involved in a cell phenotype by identifying an intrabody that can modify a cell phenotype and identifying a direct or indirect cellular target of the intrabody. In particular, intrabodies 3H2-1, 3H2-VH and 5H4 are capable of inhibiting the degranulation reaction in mast cells triggered by an allergic stimulus; furthermore, intrabodies 3H2-1 and 5H4 directly or indirectly targeted a protein of the ABCF1 family and C120RF4 family, respectively. These ABCF1 and C120RF4 inhibitors are said to be useful in therapy, in particular for treating allergic and/or inflammatory conditions. [00408] PCT Publication WOO 140276 assigned to Urogenesis Inc. generally describes the possibility of inhibition of STEAP (Six Transmembrane Epithelial Antigen of the Prostate) proteins using intracellular antibodies (intrabodies).
[00409] PCT Publication WO02086505 assigned to University of Manchester and US Patent Application Publication US2004115740 naming inventors Simon and Benton describe a method for the intracelular analysis of a target molecule, wherein intrabodies are said to be preferred. In one embodiment, a vector (designated pScFv-ECFP) capable of expressing an anti-MUCl intrabody coupled to CFP is described.
[00410] PCT Publication WO03095641 and WO0143778 assigned to Gene Therapy Systems Inc. describe compositions and methods for intracellular protein delivery, and intrabodies are generally described.
[00411] PCT Publication WO03086276 assigned to Selective Genetics Inc. describes a platform technology for the treatment of intracellular infections. Compositions and methods described therein include non-target specific vectors that target infectable cells via linked ligands that bind and internalize through cell surface receptors/moieties associated with infection. The vectors comprise exogenous nucleic acid sequences that are expressed upon internalization into a target cell. Vector associated ligands and nucleic acid molecules may be altered to target different infectious agents. In addition, the disclosure provides methods of identifying epitopes and ligands capable of directing internalization of a vector and capable of blocking viral entry.
[00412] PCT Publication WO03062415 assigned to Erasmus University describes a transgenic organism comprising a polynucleotide construct encoding an intracellular antibody which disrupts the catalysis of the production of the xenoantigen galactose alpha 1,3 galactose and/or a polynucleotide construct which encodes an intracellular antibody which binds specifically to a retrovirus protein, such as a PERV particle protein. Cells, tissues and organs of the transgenic organism may be used in xenotransplantation.
[00413] PCT Publication WO2004099775 entitled "Means for detecting protein
conformation and applications thereof describes the use of scFv fragments as conformation- specific antibodies for specifically detecting a conformational protein state, said to have applications as sensors for following in livings cells, upon intracellular expression, the behavior of endogeneous proteins.
[00414] PCT Publication WO2008070363 assigned to Imclone Systems Inc. describes a single domain intrabody that binds to an intracellular protein or to an intracellular domain of an intracellular protein, such as Etk, the endothelial and epithelial tyrosine kinase, which is a member of the Tec family of non-receptor tyrosine kinases. Also provided is a method of inhibiting an intracellular enzyme, and treating a tumor in a patient by administering the intrabody or a nucleic acid expressing the intrabody.
[00415] PCT Publication WO2009018438 assigned to Cornell Research Foundation Inc. describes a method of identifying a protein that binds to a target molecule and has intracellular functionality, by providing a construct comprising a DNA molecule encoding the protein which binds to the target molecule, with the DNA molecule being coupled to a stall sequence. A host cell is transformed with the construct and then cultured under conditions effective to form, within the host cell, a complex of the protein whose translation has been stalled, the mRNA encoding the protein, and ribosomes. The protein in the complex is in a properly folded, active form and the complex is recovered from the cell. This method can be carried out with a cell-free extract preparation containing ribosomes instead of a host cell. The present disclosure also relates to a construct which includes a DNA molecule encoding a protein that binds to a target molecule and an SecM stalling sequence coupled to the DNA molecule. The DNA molecule and the SecM stalling sequence are coupled with sufficient distance between them to permit expression of their encoded protein, within the cell, in a properly folded, active form. The use of intrabodies is generally described.
[00416] PCT Publication WO2014030780 assigned to Mogam Biotech Research Institute describes a method named Tat-associated protein engineering (TAPE), for screening a target protein having higher solubility and excellent thermostability, in particular, an
immunoglobulin variable domain (VH or VL) derived from human germ cells, by preparing a gene construct where the target protein and an antibiotic -resistant protein are linked to a Tat signal sequence, and then expressing this within E. coli. Also disclosed are human or engineered VH and VL domain antibodies and human or engineered VH and VL domain antibody scaffolds having solubility and excellent thermostability, which are screened by the TAPE method. Also provided is a library including random CDR sequences in the human or engineered VH or VL domain antibody scaffold screened by the TAPE method, a preparing method thereof, a VH or VL domain antibody having binding ability to the target protein screened by using the library, and a pharmaceutical composition including the domain antibody.
[00417] European Patent Application EP2422811 describes an antibody that binds to an intracellular epitope; such intrabodies comprise at least a portion of an antibody that is capable of specifically binding an antigen and preferably does not contain operable sequences coding for its secretion and thus remains within the cell. In one embodiment, the intrabody comprises a scFv. The scFv polypeptide further comprises a polypeptide linker between the VH and VL domains which enables the scFv to form the desired structure for antigen binding. Also described is a specific embodiment in which the intrabody binds to the cytoplasmic domain of an Eph receptor and prevents its signaling (e.g., autophosphorylation). In another specific embodiment, an intrabody binds to the cytoplasmic domain of a B-type Ephrin (e.g., EphrinB l, EphrinB2 or EphrinB3).
[00418] PCT Publication WO2011003896 and European Patent Application EP2275442 describe intracellular functional PCNA-Chromobodies made using nucleic acid molecule encoding a polypeptide specifically binding to proliferating cell nuclear antigen (PCNA). Examples of such polypeptides comprising conservative substitutions of one or more amino acids in one or two framework regions are represented by SEQ ID NOs 16 and
18, including the framework region of the polypeptide. In the examples, the framework regions as well as the CDR regions involved in the binding of PCNA have been determined.
[00419] European Patent Application EP2703485 describes a method for selecting plasma cells or plasmablasts, as well as for producing target antigen specific antibodies, and novel monoclonal antibodies. In one embodiment, cells expressing intracellular immunoglobulin were identified.
Structural analysis
[00420] In some cases, recombinant proteins of the disclosure may be subjected to structural analysis using any methods available in the art to reveal one or more structural features (e.g. secondary, tertiary and/or quaternary structure). Methods of structural analysis may include, but are not limited to X-ray crystallography, nuclear magnetic resonance analysis, hydrogen/deuterium exchange mass spectrometry, and computer-based modeling. In some cases, structural analysis may be used to identify epitopes that may be desireable targets for inhibiting or activating antibodies. For instance, in some cases, structural analysis of a protein complex may reveal one or more epitopes where antibody binding may stabilize the complex. In some cases, structural analysis of a protein complex may reveal one or more epitopes where antibody binding may destabilize the complex. In some cases, structural analysis may be used to identify one or more epitopes where antibody binding may prevent complex formation.
[00421] In some embodiments, structural analysis may be used to assess interactions between antibodies of the disclosure and their targets. Such analysis may be used to provide insight into the exact epitope bound by a particular antibody and/or or provide insight into how a particular antibody performs a specific function (e.g. inhibitory or activating functions).
[00422] In some cases, structural analysis may be used to help in the design of one or more recombinant proteins, for example, recombinant proteins to be used as antigens or to be used in selection of binding partners from a display library.
Variations
[00423] Compounds and/or compositions of the present disclosure may exist as a whole polypeptide, a plurality of polypeptides or fragments of polypeptides, which independently may be encoded by one or more nucleic acids, a plurality of nucleic acids, fragments of nucleic acids or variants of any of the aforementioned. As used herein, the term
"polypeptide" refers to a polymer of amino acid residues (natural or unnatural) linked together most often by peptide bonds. The term, as used herein, refers to proteins, polypeptides, and peptides of any size, structure, or function. In some instances the polypeptide encoded is smaller than about 50 amino acids and the polypeptide is then termed a peptide. If the polypeptide is a peptide, it will be at least about 2, 3, 4, or at least 5 amino acid residues long. Thus, polypeptides include gene products, naturally occurring
polypeptides, synthetic polypeptides, homologs, orthologs, paralogs, fragments and other equivalents, variants, and analogs of the foregoing. A polypeptide may be a single molecule or may be a multi-molecular complex such as a dimer, trimer or tetramer. They may also comprise single chain or multichain polypeptides and may be associated or linked. The term polypeptide may also apply to amino acid polymers in which one or more amino acid residues are an artificial chemical analogue of a corresponding naturally occurring amino acid.
[00424] As used herein, the term "polypeptide variant" refers to molecules which differ in their amino acid sequence from a native or reference sequence. The amino acid sequence variants may possess substitutions, deletions, and/or insertions at certain positions within the amino acid sequence, as compared to a native or reference sequence. Variants may possess at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 96%, at least about 97%, at least about 98%, at least about 99%, at least about 99.5% or at least about 99.9% identity (homology) to a native or reference sequence. [00425] In some embodiments "variant mimics" are provided. As used herein, the term "variant mimic" refers to a variant which contains one or more amino acids which would mimic an activated sequence. For example, glutamate may serve as a mimic for phospho- threonine and/or phospho- serine. Alternatively, variant mimics may result in deactivation or in an inactivated product containing the mimic, e.g., phenylalanine may act as an inactivating substitution for tyrosine; or alanine may act as an inactivating substitution for serine.The amino acid sequences of the compounds and/or compositions of the disclosure may comprise naturally occurring amino acids and as such may be considered to be proteins, peptides, polypeptides, or fragments thereof. Alternatively, the compounds and/or compositions may comprise both naturally and non-naturally occurring amino acids.
[00426] As used herein, the term "amino acid sequence variant" refers to molecules with some differences in their amino acid sequences as compared to a native or starting sequence. The amino acid sequence variants may possess substitutions, deletions, and/or insertions at certain positions within the amino acid sequence. As used herein, the terms "native" or "starting" when referring to sequences are relative terms referring to an original molecule against which a comparison may be made. Native or starting sequences should not be confused with wild type sequences. Native sequences or molecules may represent the wild- type (that sequence found in nature) but do not have to be identical to the wild-type sequence.
[00427] Ordinarily, variants will possess at least about 70% homology to a native sequence, and preferably, they will be at least about 80%, more preferably at least about 90%
homologous to a native sequence.
[00428] As used herein, the term "homology" as it applies to amino acid sequences is defined as the percentage of residues in the candidate amino acid sequence that are identical with the residues in the amino acid sequence of a second sequence after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent homology. Methods and computer programs for the alignment are well known in the art. It is understood that homology depends on a calculation of percent identity but may differ in value due to gaps and penalties introduced in the calculation.
[00429] As used herein, the term "homolog" as it applies to amino acid sequences is meant the corresponding sequence of other species having substantial identity to a second sequence of a second species.
[00430] As used herein, the term "analog" is meant to include polypeptide variants which differ by one or more amino acid alterations, e.g., substitutions, additions or deletions of amino acid residues that still maintain the properties of the parent polypeptide. [00431] As used herein, the term "derivative" is used synonymously with the term "variant" and refers to a molecule that has been modified or changed in any way relative to a reference molecule or starting molecule.
[00432] The present disclosure contemplates several types of compounds and/or compositions which are amino acid based including variants and derivatives. These include substitutional, insertional, deletional and covalent variants and derivatives. As such, included within the scope of this disclosure are compounds and/or compositions comprising substitutions, insertions, additions, deletions and/or covalent modifications. For example, sequence tags or amino acids, such as one or more lysines, can be added to peptide sequences of the disclosure (e.g., at the N-terminal or C-terminal ends). Sequence tags can be used for peptide purification or localization. Lysines can be used to increase peptide solubility or to allow for biotinylation. In some cases, amino acid sequences may be included that are targets for biotinylation (e.g. via bacterial ligase). Such sequences may include any of those listed in US Patent No. 5,723,584, the contents of which are herein incorporated by reference in their entirety. For example, the amino acid sequence GLNDIFEAQKIEWHE (SEQ ID NO: 1627) may be used, where the biotin is joined via bacterial ligase to the embedded lysine residue. In addition, antibodies specific for GLNDIFEAQKIEWHE (SEQ ID NO: 1627) may be used to target proteins expressing that sequence. In some cases, these sequences are expressed in association with N- and/or C-terminal secretion signal sequences [e.g. human Ig kappa chains with amino acid sequence MDMRVPAQLLGLLLLWFSGVLG (SEQ ID NO: 66)], flag tag sequences [e.g. DYKDDDDK (SEQ ID NO: 67)], one or more 3C protease cleavage site [e.g. LEVLFQGP (SEQ ID NO: 68)], one or more biotinylation site and/or His-tag sequences [e.g. HHHHHH (SEQ ID NO: 69)].
[00433] Amino acid residues located at the carboxy and amino terminal regions of the amino acid sequence of a peptide or protein may optionally be deleted providing for truncated sequences. Certain amino acids (e.g., C-terminal or N-terminal residues) may alternatively be deleted depending on the use of the sequence, as for example, expression of the sequence as part of a larger sequence which is soluble, or linked to a solid support.
[00434] "Substitutional variants" when referring to proteins are those that have at least one amino acid residue in a native or starting sequence removed and a different amino acid inserted in its place at the same position. The substitutions may be single, where only one amino acid in the molecule has been substituted, or they may be multiple, where two or more amino acids have been substituted in the same molecule. [00435] As used herein, the term "conservative amino acid substitution" refers to the substitution of an amino acid that is normally present in the sequence with a different amino acid of similar size, charge, or polarity. Examples of conservative substitutions include the substitution of a non-polar (hydrophobic) residue such as isoleucine, valine and leucine for another non-polar residue. Likewise, examples of conservative substitutions include the substitution of one polar (hydrophilic) residue for another such as between arginine and lysine, between glutamine and asparagine, and between glycine and serine. Additionally, the substitution of a basic residue such as lysine, arginine or histidine for another, or the substitution of one acidic residue such as aspartic acid or glutamic acid for another acidic residue are additional examples of conservative substitutions. Examples of non-conservative substitutions include the substitution of a non-polar (hydrophobic) amino acid residue such as isoleucine, valine, leucine, alanine, methionine for a polar (hydrophilic) residue such as cysteine, glutamine, glutamic acid or lysine and/or a polar residue for a non-polar residue.
[00436] As used herein, the term "insertional variants" when referring to proteins are those with one or more amino acids inserted immediately adjacent to an amino acid at a particular position in a native or starting sequence. As used herein, the term "immediately adjacent" refers to an adjacent amino acid that is connected to either the alpha-carboxy or alpha-amino functional group of a starting or reference amino acid.
[00437] As used herein, the term "deletional variants" when referring to proteins, are those with one or more amino acids in the native or starting amino acid sequence removed.
Ordinarily, deletional variants will have one or more amino acids deleted in a particular region of the molecule.
[00438] As used herein, the term "derivatives," as referred to herein includes variants of a native or starting protein comprising one or more modifications with organic proteinaceous or non-proteinaceous derivatizing agents, and post-translational modifications. Covalent modifications are traditionally introduced by reacting targeted amino acid residues of the protein with an organic derivatizing agent that is capable of reacting with selected side-chains or terminal residues, or by harnessing mechanisms of post-translational modifications that function in selected recombinant host cells. The resultant covalent derivatives are useful in programs directed at identifying residues important for biological activity, for immunoassays, or for the preparation of anti-protein antibodies for immunoaffinity purification of the recombinant glycoprotein. Such modifications are within the ordinary skill in the art and are performed without undue experimentation. [00439] Certain post-translational modifications are the result of the action of recombinant host cells on the expressed polypeptide. Glutaminyl and asparaginyl residues are frequently post-translationally deamidated to the corresponding glutamyl and aspartyl residues.
Alternatively, these residues are deamidated under mildly acidic conditions. Either form of these residues may be present in the proteins used in accordance with the present disclosure.
[00440] Other post-translational modifications include hydroxylation of proline and lysine, phosphorylation of hydroxyl groups of seryl or threonyl residues, methylation of the alpha- amino groups of lysine, arginine, and histidine side chains (T. E. Creighton, Proteins:
Structure and Molecular Properties, W.H. Freeman & Co., San Francisco, pp. 79-86 (1983)).
[00441] Covalent derivatives specifically include fusion molecules in which proteins of the disclosure are covalently bonded to a non-proteinaceous polymer. The non-proteinaceous polymer ordinarily is a hydrophilic synthetic polymer, i.e. a polymer not otherwise found in nature. However, polymers which exist in nature and are produced by recombinant or in vitro methods are useful, as are polymers which are isolated from nature. Hydrophilic polyvinyl polymers fall within the scope of this disclosure, e.g. polyvinylalcohol and
polyvinylpyrrolidone. Particularly useful are polyvinylalkylene ethers such a polyethylene glycol, polypropylene glycol. The proteins may be linked to various non-proteinaceous polymers, such as polyethylene glycol, polypropylene glycol or polyoxyalkylenes, in the manner set forth in U.S. Pat. No. 4,640,835; 4,496,689; 4,301,144; 4,670,417; 4,791,192 or 4,179,337.
[00442] As used herein, the term "features" when referring to proteins are defined as distinct amino acid sequence-based components of a molecule. Features of the proteins of the present disclosure include surface manifestations, local conformational shape, folds, loops, half-loops, domains, half-domains, sites, termini or any combination thereof.
[00443] As used herein, the term "surface manifestation" when referring to proteins refers to a polypeptide based component of a protein appearing on an outermost surface.
[00444] As used herein, the term "local conformational shape" when referring to proteins refers to a polypeptide based structural manifestation of a protein which is located within a definable space of the protein.
[00445] As used herein, the term "fold", when referring to proteins, refers to the resultant conformation of an amino acid sequence upon energy minimization. A fold may occur at the secondary or tertiary level of the folding process. Examples of secondary level folds include beta sheets and alpha helices. Examples of tertiary folds include domains and regions formed due to aggregation or separation of energetic forces. Regions formed in this way include hydrophobic and hydrophilic pockets, and the like.
[00446] As used herein, the term "turn" as it relates to protein conformation, refers to a bend which alters the direction of the backbone of a peptide or polypeptide and may involve one, two, three or more amino acid residues.
[00447] As used herein, the term "loop," when referring to proteins, refers to a structural feature of a peptide or polypeptide which reverses the direction of the backbone of a peptide or polypeptide and comprises four or more amino acid residues. Oliva et al. have identified at least 5 classes of protein loops (Oliva, B. et al., An automated classification of the structure of protein loops. J Mol Biol. 1997. 266(4):814-30).
[00448] As used herein, the term "half-loop," when referring to proteins, refers to a portion of an identified loop having at least half the number of amino acid resides as the loop from which it is derived. It is understood that loops may not always contain an even number of amino acid residues. Therefore, in those cases where a loop contains or is identified to comprise an odd number of amino acids, a half-loop of the odd-numbered loop will comprise the whole number portion or next whole number portion of the loop (number of amino acids of the loop/2+/-0.5 amino acids). For example, a loop identified as a 7 amino acid loop could produce half-loops of 3 amino acids or 4 amino acids (7/2=3.5+/-0.5 being 3 or 4).
[00449] As used herein, the term "domain," when referring to proteins, refers to a motif of a polypeptide having one or more identifiable structural or functional characteristics or properties (e.g., binding capacity, serving as a site for protein-protein interactions).
[00450] As used herein, the term "half-domain," when referring to proteins, refers to a portion of an identified domain having at least half the number of amino acid resides as the domain from which it is derived. It is understood that domains may not always contain an even number of amino acid residues. Therefore, in those cases where a domain contains or is identified to comprise an odd number of amino acids, a half-domain of the odd-numbered domain will comprise the whole number portion or next whole number portion of the domain (number of amino acids of the domain/2+/-0.5 amino acids). For example, a domain identified as a 7 amino acid domain could produce half-domains of 3 amino acids or 4 amino acids (7/2=3.5+/-0.5 being 3 or 4). It is also understood that sub-domains may be identified within domains or half-domains, these subdomains possessing less than all of the structural or functional properties identified in the domains or half domains from which they were derived. It is also understood that the amino acids that comprise any of the domain types herein need not be contiguous along the backbone of the polypeptide (i.e., nonadjacent amino acids may fold structurally to produce a domain, half-domain or subdomain).
[00451] As used herein, the terms "site," as it pertains to amino acid based embodiments is used synonymously with "amino acid residue" and "amino acid side chain". A site represents a position within a peptide or polypeptide that may be modified, manipulated, altered, derivatized or varied within the polypeptide based molecules of the present disclosure.
[00452] As used herein, the terms "termini" or "terminus," when referring to proteins refers to an extremity of a peptide or polypeptide. Such extremity is not limited only to the first or final site of the peptide or polypeptide but may include additional amino acids in the terminal regions. The polypeptide based molecules of the present disclosure may be characterized as having both an N-terminus (terminated by an amino acid with a free amino group (NH2)) and a C-terminus (terminated by an amino acid with a free carboxyl group (COOH)). Proteins of the disclosure are in some cases made up of multiple polypeptide chains brought together by disulfide bonds or by non-covalent forces (multimers, oligomers). These sorts of proteins will have multiple N- and C-termini. Alternatively, the termini of the polypeptides may be modified such that they begin or end, as the case may be, with a non-polypeptide based moiety such as an organic conjugate.
[00453] Once any of the features have been identified or defined as a component of a molecule of the disclosure, any of several manipulations and/or modifications of these features may be performed by moving, swapping, inverting, deleting, randomizing or duplicating. Furthermore, it is understood that manipulation of features may result in the same outcome as a modification to the molecules of the disclosure. For example, a manipulation which involved deleting a domain would result in the alteration of the length of a molecule just as modification of a nucleic acid to encode less than a full length molecule would.
[00454] Modifications and manipulations can be accomplished by methods known in the art such as site directed mutagenesis. The resulting modified molecules may then be tested for activity using in vitro or in vivo assays such as those described herein or any other suitable screening assay known in the art.
[00455] In some embodiments, compounds and/or compositions of the present disclosure may comprise one or more atoms that are isotopes. As used herein, the term "isotope" refers to a chemical element that has one or more additional neutrons. In some embodiments, compounds of the present disclosure may be deuterated. As used herein, the term "deuterate" refers to the process of replacing one or more hydrogen atoms in a substance with deuterium isotopes. Deuterium isotopes are isotopes of hydrogen. The nucleus of hydrogen contains one proton while deuterium nuclei contain both a proton and a neutron. The compounds and/or compositions of the present disclosure may be deuterated in order to change one or more physical property, such as stability, or to allow compounds and/or compositions to be used in diagnostic and/or experimental applications.
Conjugates and Combinations
[00456] It is contemplated by the present disclosure that the compounds and/or
compositions of the present disclosure may be complexed, conjugated or combined with one or more homologous or heterologous molecules. As used herein, the term "homologous molecule" refers to a molecule which is similar in at least one of structure or function relative to a starting molecule while a "heterologous molecule" is one that differs in at least one of structure or function relative to a starting molecule. Structural homologs are therefore molecules which may be substantially structurally similar. In some embodiments, such homologs may be identical. Functional homologs are molecules which may be substantially functionally similar. In some embodiments, such homologs may be identical.
[00457] Compounds and/or compositions of the present disclosure may comprise conjugates. Such conjugates of the disclosure may include naturally occurring substances or ligands, such as proteins (e.g., human serum albumin (HSA), low-density lipoprotein (LDL), high-density lipoprotein (HDL), or globulin); carbohydrates (e.g., a dextran, pullulan, chitin, chitosan, inulin, cyclodextrin or hyaluronic acid); or lipids. Conjugates may also be recombinant or synthetic molecules, such as synthetic polymers, e.g., synthetic polyamino acids, an oligonucleotide (e.g. an aptamer). Examples of polyamino acids may include polylysine (PLL), poly L-aspartic acid, poly L-glutamic acid, styrene-maleic acid anhydride copolymer, poly(L-lactide-co-glycolied) copolymer, divinyl ether-maleic anhydride copolymer, N-(2-hydroxypropyl)methacrylamide copolymer (HMPA), polyethylene glycol (PEG), polyvinyl alcohol (PVA), polyurethane, poly(2-ethylacryllic acid), N- isopropylacrylamide polymers, or polyphosphazine. Example of polyamines include:
polyethylenimine, polylysine (PLL), spermine, spermidine, polyamine, pseudopeptide- polyamine, peptidomimetic polyamine, dendrimer polyamine, arginine, amidine, protamine, cationic lipid, cationic porphyrin, quaternary salt of a polyamine, or an alpha helical peptide.
[00458] In some embodiments, conjugates may also include targeting groups. As used herein, the term "targeting group" refers to a functional group or moiety attached to an agent that facilitates localization of the agent to a desired region, tissue, cell and/or protein. Such targeting groups may include, but are not limited to cell or tissue targeting agents or groups (e.g. lectins, glycoproteins, lipids, proteins, an antibody that binds to a specified cell type such as a kidney cell or other cell type). In some embodiments, targeting groups may comprise thyrotropins, melanotropins, lectins, glycoproteins, surfactant protein A, mucin carbohydrates, multivalent lactose, multivalent galactose, N-acetyl-galactosamine, N-acetyl- gulucosamine, multivalent mannose, multivalent fucose, glycosylated polyaminoacids, multivalent galactose, transferrin, bisphosphonate, polyglutamate, polyaspartate, lipids, cholesterol, steroids, bile acids, folates, vitamin B 12, biotin, an RGD peptide, an RGD peptide mimetic or an aptamer.
[00459] In some embodiments, targeting groups may be proteins, e.g., glycoproteins, or peptides, e.g., molecules having a specific affinity for a co-ligand, or antibodies e.g., an antibody, that binds to a specified cell type such as a cancer cell, endothelial cell, or bone cell. Targeting groups may also comprise hormones and/or hormone receptors.
[00460] In some embodiments, targeting groups may be any ligand capable of targeting specific receptors. Examples include, without limitation, folate, GalNAc, galactose, mannose, mannose-6-phosphate, apatamers, integrin receptor ligands, chemokine receptor ligands, transferrin, biotin, serotonin receptor ligands, PSMA, endothelin, GCPII, somatostatin, LDL, and HDL ligands. In some embodiments, targeting groups are aptamers. Such aptamers may be unmodified or comprise any combination of modifications disclosed herein.
[00461] In still other embodiments, compounds and/or compositions of the present disclosure may be covalently conjugated to cell penetrating polypeptides. In some
embodiments, cell-penetrating peptides may also include signal sequences. In some embodiments, conjugates of the disclosure may be designed to have increased stability, increased cell transfection and/or altered biodistribution (e.g., targeted to specific tissues or cell types).
[00462] In some embodiments, conjugating moieties may be added to compounds and/or compositions of the present disclosure such that they allow the attachment of detectable labels to targets for clearance. Such detectable labels include, but are not limited to biotin labels, ubiquitins, fluorescent molecules, human influenza hemaglutinin (HA), c-myc, histidine (His), flag, glutathione S-transferase (GST), V5 (a paramyxovirus of simian virus 5 epitope), biotin, avidin, streptavidin, horse radish peroxidase (HRP) and digoxigenin.
[00463] In some embodiments, compounds of the disclosure may be conjugated with an antibody Fc domain to create an Fc fusion protein. The formation of an Fc fusion protein with any of the compounds described herein may be carried out according to any method known in the art, including as described in US Patent Nos. 5,116,964, 5,541,087 and 8,637,637, the contents of each of which are herein incorporated by reference in their entirety. Fc fusion proteins of the disclosure may comprise a compound of the disclosure linked to the hinge region of an IgG Fc via cysteine residues in the Fc hinge region. Resulting Fc fusion proteins may comprise an antibody-like structure, but without CHI domains or light chains. In some cases, Fc fusion proteins may comprise pharmacokinetic profiles comparable to native antibodies. In some cases, Fc fusion proteins of the disclosure may comprise extended half- life in circulation and/or altered biological activity.
[00464] In some embodiments, compounds and/or compositions of the present disclosure may be combined with one another or other molecules in the treatment of diseases and/or conditions.
Nucleic acids
[00465] In some embodiments, compounds and/or compositions of the present disclosure may be encoded by nucleic acid molecules. Such nucleic acid molecules include, without limitation, DNA molecules, RNA molecules, polynucleotides, oligonucleotides, mRNA molecules, vectors, plasmids and the like. In some embodiments, the present disclosure may comprise cells programmed or generated to express nucleic acid molecules encoding compounds and/or compositions of the present disclosure. In some cases, nucleic acids of the disclosure include codon-optimized nucleic acids. Methods of generating codon-optimized nucleic acids are known in the art and may include, but are not limited to those described in US Patent Nos. 5,786,464 and 6,114,148, the contents of each of which are herein incorporated by reference in their entirety.
Methods of use
[00466] Methods of the present disclosure include methods of modifying growth factor activity in one or more biological system. Such methods may include contacting one or more biological system with a compound and/or composition of the disclosure. In some cases, these methods include modifying the level of free growth factor in a biological system (e.g. in a cell niche or subject). Compounds and/or compostions according to such methods may include, but are not limited to biomolecules, including, but not limited to recombinant proteins, protein complexes and/or antibodies described herein.
[00467] In some embodiments, methods of the present disclosure may be used to initiate or increase growth factor activity, termed "activating methods" herein. Some such methods may comprise growth factor release from a GPC and/or inhibition of growth factor reassociation into a latent GPC. In some cases, activating methods may comprise the use of an antibody, a recombinant protein and/or a protein complex. According to some activating methods, one or more activating antibody is provided. In such methods, one or more growth factor may be released or prevented from being drawn back into a GPC. In one, non-limiting example, an anti-LAP antibody may be provided that enhances dissociation between a growth factor and a GPC and/or prevents reformation of a GPC.
[00468] Embodiments of the present disclosure include methods of using anti-prodomain antibodies to modify growth factor activity. In some cases, such methods may include the use of anti-GDF prodomain antibodies as GDF- activating antibodies.
[00469] In some embodiments, methods of the present disclosure may be used to reduce or eliminate growth factor activity, termed "inhibiting methods" herein. Some such methods may comprise growth factor retention in a GPC and/or promotion of reassociation of growth factor into a latent GPC. In some cases, inhibiting methods may comprise the use of an antibody, a recombinant protein and/or a protein complex. According to some inhibiting methods, one or more inhibiting antibody is provided. In some cases, inhibiting methods comprise the use of inhibiting recombinant proteins or inhibiting protein complexes capable of association with a growth factor, wherein the association prevents growth factor activity.
Targeting complexes
[00470] In some embodiments methods of the present disclosure may comprise the use of one or more targeting complex. As used herein, the term "targeting complex" refers to a protein complex wherein at least one protein component acts as a targeting agent. As used herein, the term "targeting agent" refers to an agent that directs cargo or other components complexed with the agent to a target site.
[00471] In some cases, targeting complexes may comprise one or more extracellular matrix proteins and/or proteins associated with the extracellular matrix. Such proteins may function as targeting agents in a targeting complex. According to such embodiments, the extracellular matrix component of a targeting complex may direct the complex to target sites comprising extracellular matrix and/or cellular matrix. Extracellular matrix components of targeting complexes may include, but are not limited to LTBPs (e.g. LTBP1, LTBP2, LTBP3 and/or LTBP4), fibrillins (e.g. fibrillin- 1, fibrillin-2, fibrillin-3 and/or fibrillin-4), perlecan, decorin, elastin, collagen, GASPs and/or GARPs (e.g. GARP and/or LRRC33). [00472] In some embodiments, LTBP isoforms may be used as targeting agents to direct targeting complexes to areas of extra cellular matrix surrounding different tissues. LTBP1, for example, has been shown to be expressed predominantly in the heart, lung, kidney, placenta, spleen and stomach. As such, targeting complexes may be directed to those organs by incorporation of LTBP1 as a targeting agent. Similarly, LTBP2 is found in the lung, skeletal muscle, liver and placenta while LTBP3 and LTBP4 are both known to be expressed in the skeletal muscle, heart, ovaries and small intestine (Ceco, E. 2013. FEBS J.
280(17):4198-209, the contents of which are herein incorporated by reference in their entirety). These differential regions of expression may be target sites for targeting complexes in which LTBP2, 3 or 4 isoforms may be used as targeting agents.
[00473] Some targeting complexes of the disclosure may comprise one or more prodomain component, such as a prodomain. In some cases, the portion of such targeting complexes may function to bind free growth factors to reduce free growth factor levels and/or activity. In some cases, GDF prodomains may be included in targeting complexes.
Therapeutics
[00474] In some embodiments, compositions and methods of the disclosure may be used to treat a wide variety of diseases, disorders and/or conditions. In some cases, such diseases, disorders and/or conditions may be TGF-P-related indications. As used herein, the term "TGF-P-related indication" refers to any disease, disorder and/or condition related to expression, activity and/or metabolism of a TGF-β family member protein or any disease, disorder and/or condition that may benefit from modulation of the activity and/or levels of one or more TGF-β family member protein. TGF-P-related indications may include, but are not limited to, fibrosis, anemia of the aging, cancer (including, but not limited to colon, renal, breast, malignant melanoma and glioblastoma), facilitation of rapid hematopoiesis following chemotherapy, bone healing, endothelial proliferation syndromes, asthma and allergy, gastrointestinal disorders, aortic aneurysm, orphan indications (such as Marfan's syndrome and Camurati-Engelmann disease), obesity, diabetes, arthritis, multiple sclerosis, muscular dystrophy, amyotrophic lateral sclerosis (ALS), Parkinson's disease, osteoporosis, osteoarthritis, osteopenia, metabolic syndromes, nutritional disorders, organ atrophy, chronic obstructive pulmonary disease (COPD), and anorexia. Additional indications may include any of those disclosed in US Pub. No. 2013/0122007, US Pat. No. 8,415,459 or International Pub. No. WO 2011/151432, the contents of each of which are herein incorporated by reference in their entirety. [00475] Efficacy of treatment or amelioration of disease can be assessed, for example by measuring disease progression, disease remission, symptom severity, reduction in pain, quality of life, dose of a medication required to sustain a treatment effect, level of a disease marker or any other measurable parameter appropriate for a given disease being treated or targeted for prevention. It is well within the ability of one skilled in the art to monitor efficacy of treatment or prevention by measuring any one of such parameters, or any combination of parameters. In connection with the administration of compositions of the present disclosure, "effective against" for example a cancer, indicates that administration in a clinically appropriate manner results in a beneficial effect for at least a statistically significant fraction of patients, such as an improvement of symptoms, a cure, a reduction in disease load, reduction in tumor mass or cell numbers, extension of life, improvement in quality of life, or other effect generally recognized as positive by medical doctors familiar with treating the particular type of cancer.
[00476] A treatment or preventive effect is evident when there is a statistically significant improvement in one or more parameters of disease status, or by a failure to worsen or to develop symptoms where they would otherwise be anticipated. As an example, a favorable change of at least 10% in a measurable parameter of disease, and preferably at least 20%, 30%, 40%, 50% or more can be indicative of effective treatment. Efficacy for a given composition or formulation of the present disclosure can also be judged using an
experimental animal model for the given disease as known in the art. When using an experimental animal model, efficacy of treatment is evidenced when a statistically significant change is observed.
Therapeutics for anemia, thrombocytopenia and neutropenia
[00477] During chemotherapy, cell division is temporarily halted to prevent the growth and spread of cancerous cells. An unfortunate side effect is the loss of red blood cells, platelets and white blood cells which depend on active cell division of bone marrow cells. In some embodiments, compounds and/or compositions of the present disclosure may be designed to treat patients suffering from anemia (the loss of red blood cells), thrombocytopenia (a decrease in the number of platelets) and/or neutropenia (a decrease in the number of neutrophils).
Therapeutics for cancer [00478] Various cancers may be treated with compounds and/or compositions of the present disclosure. As used herein, the term "cancer" refers to any of various malignant neoplasms characterized by the proliferation of anaplastic cells that tend to invade
surrounding tissue and metastasize to new body sites and also refers to the pathological condition characterized by such malignant neoplastic growths. Cancers may be tumors or hematological malignancies, and include but are not limited to, all types of
lymphomas/leukemias, carcinomas and sarcomas, such as those cancers or tumors found in the anus, bladder, bile duct, bone, brain, breast, cervix, colon/rectum, endometrium, esophagus, eye, gallbladder, head and neck, liver, kidney, larynx, lung, mediastinum (chest), mouth, ovaries, pancreas, penis, prostate, skin, small intestine, stomach, spinal marrow, tailbone, testicles, thyroid and uterus.
[00479] In cancer, TGF-β may be either growth promoting or growth inhibitory. As an example, in pancreatic cancers, SMAD4 wild type tumors may experience inhibited growth in response to TGF-β, but as the disease progresses, constitutively activated type II receptor is typically present. Additionally, there are SMAD4-null pancreatic cancers. In some embodiments, compounds and/or compositions of the present disclosure are designed to selectively target components of TGF-β signaling pathways that function uniquely in one or more forms of cancer. Leukemias, or cancers of the blood or bone marrow that are characterized by an abnormal proliferation of white blood cells i.e., leukocytes, can be divided into four major classifications including Acute lymphoblastic leukemia (ALL), Chronic lymphocytic leukemia (CLL), Acute myelogenous leukemia or acute myeloid leukemia (AML) (AML with translocations between chromosome 10 and 11 [t(10, 11)], chromosome 8 and 21 [t(8;21)], chromosome 15 and 17 [t(15;17)], and inversions in chromosome 16 [inv(16)]; AML with multilineage dysplasia, which includes patients who have had a prior myelodysplasia syndrome (MDS) or myeloproliferative disease that transforms into AML; AML and myelodysplasia syndrome (MDS), therapy-related, which category includes patients who have had prior chemotherapy and/or radiation and
subsequently develop AML or MDS; d) AML not otherwise categorized, which includes subtypes of AML that do not fall into the above categories; and e) Acute leukemias of ambiguous lineage, which occur when the leukemic cells cannot be classified as either myeloid or lymphoid cells, or where both types of cells are present); and Chronic
myelogenous leukemia (CML).
[00480] The types of carcinomas include, but are not limited to, papilloma/carcinoma, choriocarcinoma, endodermal sinus tumor, teratoma, adenoma/adenocarcinoma, melanoma, fibroma, lipoma, leiomyoma, rhabdomyoma, mesothelioma, angioma, osteoma, chondroma, glioma, lymphoma/leukemia, squamous cell carcinoma, small cell carcinoma, large cell undifferentiated carcinomas, basal cell carcinoma and sinonasal undifferentiated carcinoma.
[00481] The types of sarcomas include, but are not limited to, soft tissue sarcoma such as alveolar soft part sarcoma, angiosarcoma, dermatofibro sarcoma, desmoid tumor,
desmoplastic small round cell tumor, extraskeletal chondrosarcoma, extraskeletal
osteosarcoma, fibrosarcoma, hemangiopericytoma, hemangio sarcoma, Kaposi's sarcoma, leiomyosarcoma, liposarcoma, lymphangiosarcoma, lymphosarcoma, malignant fibrous histiocytoma, neurofibrosarcoma, rhabdomyosarcoma, synovial sarcoma, and Askin's tumor, Ewing's sarcoma (primitive neuroectodermal tumor), malignant hemangioendothelioma, malignant schwannoma, osteosarcoma, and chondrosarcoma.
[00482] In some embodiments, compositions and methods of the disclosure may be used to treat one or more types of cancer or cancer-related conditions that may include, but are not limited to colon cancer, renal cancer, breast cancer, malignant melanoma and glioblastomas (Schlingensiepen et al., 2008; Ouhtit et al., 2013).
[00483] High-grade gliomas (e.g. anaplastic astrocytomas and glioblastomas) make up around 60% of malignant brain tumors. TGF-P2 has been found to be overexpressed in over 90% of such gliomas and expression levels correlate with tumor progression. Further, studies using TGF-P2 reduction at the mRNA level in cancer patients showed significant
improvement in tumor outcome (Bogdahn et al., 2010). In light of these studies, some compositions of the present disclosure may be used therapeutically to treat individuals with high-grade gliomas. Such compositions may act to lower the levels of free TGF-P2 and/or the levels of TGF-P2 activity.
[00484] In some cases, TGF-P2 activity may contribute to tumor development through modulation of metastasis, angiogenesis, proliferation and/or immunosuppressive functions that impair immunological tumor surveillance (Schlingensiepen et al., 2008). A study by Reed et al (Reed et al., 1994) demonstrated TGF-P2 mRNA expression in a large percentage of melanocytic lesions including primary invasive melanomas and metastatic melanomas. Some compounds and/or compositions of the present disclosure may be used to modulate TGF-P2 activity and/or levels in such lesions and or prevent lesion formation. Melanoma cell growth in the brain parenchyma has also been shown to be influenced by TGF-P2 activity (Zhang et al., 2009). Some compounds and/or compositions of the present disclosure may be used to prevent or control such cell growth through modulation of TGF-P2 activity and/or levels. [00485] Among females worldwide, breast cancer is the most prevalent form of cancer. Breast cancer metastasis is mediated in part through interactions between cancer cells and extracellular matrix components, such as hyaluronic acid (HA). CD44 has been shown to be the major receptor for HA on cancer cells (Ouhtit et al., 2013). The interaction between CD44 and HA leads to modulation of cell motility, survival adhesion and proliferation. TGF-P2 transcription is also upregulated by CD44 signaling activity and is believe to contribute to resulting changes in cell motility. Unfortunately, current therapies have limited efficacy and many carry adverse effects due to a lack of specificity. In some cases, compounds and/or compositions of the present disclosure may be used to alter cellular activities induced by TGF-P2 upregulation.
[00486] The disclosure further relates to the use of compounds and/or compositions of the present disclosure for treating one or more forms of cancer, in combination with other pharmaceuticals and/or other therapeutic methods, e.g., with known pharmaceuticals and/or known therapeutic methods, such as, for example, those which are currently employed for treating these disorders. For example, the compounds and/or compositions of the present disclosure can also be administered in conjunction with one or more additional anti-cancer treatments, such as biological, chemotherapy and radiotherapy. Accordingly, a treatment can include, for example, imatinib (Gleevac), all-trans-retinoic acid, a monoclonal antibody treatment (gemtuzumab, ozogamicin), chemotherapy (for example, chlorambucil, prednisone, prednisolone, vincristine, cytarabine, clofarabine, farnesyl transferase inhibitors, decitabine, inhibitors of MDR1), rituximab, interferon-a, anthracycline drugs (such as daunorubicin or idarubicin), L-asparaginase, doxorubicin, cyclophosphamide, doxorubicin, bleomycin, fludarabine, etoposide, pentostatin, or cladribine), bone marrow transplant, stem cell transplant, radiation therapy, anti-metabolite drugs (methotrexate and 6-mercaptopurine), or any combination thereof.
[00487] Radiation therapy (also called radiotherapy, X-ray therapy, or irradiation) is the use of ionizing radiation to kill cancer cells and shrink tumors. Radiation therapy can be administered externally via external beam radiotherapy (EBRT) or internally via
brachytherapy. The effects of radiation therapy are localized and confined to the region being treated. Radiation therapy may be used to treat almost every type of solid tumor, including cancers of the brain, breast, cervix, larynx, lung, pancreas, prostate, skin, stomach, uterus, or soft tissue sarcomas. Radiation is also used to treat leukemia and lymphoma.
[00488] Chemotherapy is the treatment of cancer with drugs that can destroy cancer cells. In current usage, the term "chemotherapy" usually refers to cytotoxic drugs which affect rapidly dividing cells in general, in contrast with targeted therapy. Chemotherapy drugs interfere with cell division in various possible ways, e.g. with the duplication of DNA or the separation of newly formed chromosomes. Most forms of chemotherapy target all rapidly dividing cells and are not specific to cancer cells, although some degree of specificity may come from the inability of many cancer cells to repair DNA damage, while normal cells generally can.
[00489] Most chemotherapy regimens are given in combination. Exemplary
chemotherapeutic agents include , but are not limited to, 5-FU Enhancer, 9-AC, AG2037, AG3340, Aggrecanase Inhibitor, Aminoglutethimide, Amsacrine (m-AMSA), Asparaginase, Azacitidine, Batimastat (BB94), BAY 12-9566, BCH-4556, Bis-Naphtalimide, Busulfan, Capecitabine, Carboplatin, Carmustaine+Polifepr Osan, cdk4/cdk2 inhibitors, Chlorombucil, CI-994, Cisplatin, Cladribine, CS-682, Cytarabine HCl, D2163, Dactinomycin, Daunorubicin HCl, DepoCyt, Dexifosamide, Docetaxel, Dolastain, Doxifluridine, Doxorubicin, DX8951f, E 7070, EGFR, Epirubicin, Erythropoietin, Estramustine phosphate sodium, Etoposide (VP16-213), Farnesyl Transferase Inhibitor, FK 317, Flavopiridol, Floxuridine, Fludarabine, Fluorouracil (5-FU), Flutamide, Fragyline, Gemcitabine, Hexamethylmelamine (HMM), Hydroxyurea (hydroxycarbamide), Ifosfamide, Interferon Alfa-2a, Interferon Alfa-2b, Interleukin-2, Irinotecan, ISI 641, Krestin, Lemonal DP 2202, Leuprolide acetate (LHRH- releasing factor analogue), Levamisole, LiGLA (lithium-gamma linolenate), Lodine Seeds, Lometexol, Lomustine (CCNU), Marimistat, Mechlorethamine HCl (nitrogen mustard), Megestrol acetate, Meglamine GLA, Mercaptopurine, Mesna, Mitoguazone (methyl-GAG; methyl glyoxal bis-guanylhydrazone; MGBG), Mitotane (o.p'-DDD), Mitoxantrone,
Mitoxantrone HCl, MMI 270, MMP, MTA/LY 231514, Octreotide, ODN 698, OK-432, Oral Platinum, Oral Taxoid, Paclitaxel (TAXOL.RTM.), PARP Inhibitors, PD 183805, Pentostatin (2' deoxycoformycin), PKC 412, Plicamycin, Procarbazine HCl, PSC 833, Ralitrexed, RAS Farnesyl Transferase Inhibitor, RAS Oncogene Inhibitor, Semustine (methyl-CCNU), Streptozocin, Suramin, Tamoxifen citrate, Taxane Analog, Temozolomide, Teniposide (VM- 26), Thioguanine, Thiotepa, Topotecan, Tyrosine Kinase, UFT (Tegafur/Uracil), Valrubicin, Vinblastine sulfate, Vindesine sulfate, VX-710, VX-853, YM 116, ZD 0101, ZD
0473/Anormed, ZD 1839, ZD 9331.
[00490] Biological therapies use the body' s immune system, either directly or indirectly, to fight cancer or to lessen the side effects that may be caused by some cancer treatments. In some embodiments, compounds and/or compositions of the present disclosure may be considered biological therapies in that they may stimulate immune system action against one or more tumor, for example. However, this approach may also be considered with other such biological approaches, e.g., immune response modifying therapies such as the administration of interferons, interleukins, colony- stimulating factors, other monoclonal antibodies, vaccines, gene therapy, and nonspecific immunomodulating agents are also envisioned as anti-cancer therapies to be combined with the compounds and/or compositions of the present disclosure.
[00491] Small molecule targeted therapy drugs are generally inhibitors of enzymatic domains on mutated, overexpressed, or otherwise critical proteins within the cancer cell, such as tyrosine kinase inhibitors imatinib (Gleevec/Glivec) and gefitinib (Iressa). Examples of monoclonal antibody therapies that can be used with compounds and/or compositions of the present disclosure include, but are not limited to, the anti-HER2/neu antibody trastuzumab (Herceptin) used in breast cancer, and the anti-CD20 antibody rituximab, used in a variety of B-cell malignancies. The growth of some cancers can be inhibited by providing or blocking certain hormones. Common examples of hormone- sensitive tumors include certain types of breast and prostate cancers. Removing or blocking estrogen or testosterone is often an important additional treatment. In certain cancers, administration of hormone agonists, such as progestogens may be therapeutically beneficial.
[00492] Cancer immunotherapy refers to a diverse set of therapeutic strategies designed to induce the patient's own immune system to fight the tumor, and include, but are not limited to, intravesical BCG immunotherapy for superficial bladder cancer, vaccines to generate specific immune responses, such as for malignant melanoma and renal cell carcinoma, and the use of Sipuleucel-T for prostate cancer, in which dendritic cells from the patient are loaded with prostatic acid phosphatase peptides to induce a specific immune response against prostate-derived cells.
Therapeutics for angiogenic and endothelial proliferation conditions
[00493] The compounds and/or compositions of the present disclosure may be used to treat angiogenic and endothelial proliferation syndromes, diseases or disorders. The term
"angiogenesis", as used herein refers to the formation and/or reorganization of new blood vessels. Angiogenic disease involves the loss of control over angiogenesis in the body. In such cases, blood vessel growth, formation or reorganization may be overactive (including during tumor growth and cancer where uncontrolled cell growth requires increased blood supply) or insufficient to sustain healthy tissues. Such conditions may include, but are not limited to angiomas, angiosarcomas, telangiectasia, lymphangioma, congenital vascular anomalies, tumor angiogenesis and vascular structures after surgery. Excessive angiogenesis is noted in cancer, macular degeneration, diabetic blindness, rheumatoid arthritis, psoriasis as well as many other conditions. Excessive angiogenesis is often promoted by excessive angiogenic growth factor expression. Compounds and/or compositions of the present disclosure may act to block growth factors involved in excessive angiogenesis. Alternatively, compounds and/or compositions of the present disclosure may be utilized to promote growth factor signaling to enhance angiogenesis in conditions where angiogenesis is inhibited. Such conditions include, but are not limited to coronary artery disease, stroke, diabetes and chronic wounds.
Therapeutics for cardiovascular indications
[00494] In some embodiments, compounds and/or compositions of the present disclosure may be used to treat one or more cardiovascular indications, including, but not limited to cardiac hypertrophy. Cardiac hypertrophy comprises enlargement of the heart due, typically due to increased cell volume of cardiac cells (Aurigemma 2006. N Engl J Med. 355(3):308- 10). Age-related cardiac hypertrophy may be due, in part, to reduced circulating levels of GDF-11. A study by Loffredo et al (Loffredo et al., 2013. Cell. 153:828-39) found that fusion of the circulatory system between young and old mice had a protective effect with regard to cardiac hypertrophy. The study identified GDF-11 as a circulating factor that decreased with age in mice and was able to show that its administration could also reduce cardiac hypertrophy. Some compounds and/or compositions of the present disclosure may be used to treat and/or prevent cardiac atrophy. Such compounds and/or compositions may comprise GDF-11 agonists that elevate levels of circulating GDF-11, in some cases through enhancing the dissociation of GDF-11 growth factor from latent GPCs.
[00495] In some embodiments, animal models may be used to develop and test compounds and/or compositions of the present disclosure for use in the treatment of cardiovascular diseases, disorders and/or conditions. In some cases, vascular injury models may be used to test compounds in the treatment of atherosclerosis and/or restenosis. Such models may include balloon injury models. In some cases, these may be carried out as described in Smith et al., 1999. Circ Res. 84(10): 1212-22, the contents of which are herein incorporated by reference in their entirety.
Therapeutics related to muscle disorders and/or injuries [00496] In some embodiments, compounds and/or compositions of the present disclosure may be used to treat one or more muscle disorders and/or injuries. In some cases, such compounds and/or composition may include, but are not limited to antibodies that modulate GDF-8, GDF-11 and/or activin activity. Muscle comprises about 40-50% of total body weight, making it the largest organ in the body. Muscle disorders may include cachexia (e.g. muscle wasting). Muscle wasting may be associated with a variety of diseases and catabolic disorders (e.g. HIV/AIDS, cancer, cancer cachexia, renal failure, congestive heart failure, muscular dystrophy, disuse atrophy, chronic obstructive pulmonary disease, motor neuron disease, trauma, neurodegenerative disease, infection, rheumatoid arthritis, immobilization, diabetes, etc.). In such disorders, GDF-8 and/or activin signaling activity may contribute to muscle catabolism (Han et al., 2013. Int J Biochem Cell Biol. 45(10):2333-47; Lee., 2010. Immunol Endocr Metab Agents Med Chem. 10: 183-94, the contents of each of which are herein incorporated by reference in their entirety). Other muscle disorders may comprise sarcopenia. Sarcopenia is the progressive loss of muscle and function associated with aging. In the elderly, sarcopenia can cause frailty, weakness, fatigue and loss of mobility (Morely. 2012. Family Practice. 29:i44-i48). With the aged population increasing in numbers, sarcopenia is progressively becoming a more serious public health concern. A study by Hamrick et al (Hamrick et al., 2010. 69(3):579-83) demonstrated that GDF-8 inhibition could repair muscle in a mouse model of fibula osteotomoy comprising lateral compartment muscle damage. Administration of GDF-8 propeptides was sufficient to increase muscle mass by nearly 20% as well as improve fracture healing. Some compounds and/or compositions of the present disclosure may be used to treat muscle diseases, disorders and/or injuries by modulating GDF-8 activity. In some cases, compounds of the present disclosure may be GDF-8 signaling antagonists, preventing or reducing GDF-8 signaling activity.
[00497] Inclusion body myositis (IBM) is a disease characterized by progressive muscle loss, typically occurring in mid- to late-life. The disease is thought to occur due to an autoimmune response to autoantigens in the muscle causing T-cell invasion of the muscle fiber and resulting in myofiber destruction (Greenberg 2012. Curr Opin Neurol. 25(5):630-9). Therapeutic compounds are being investigated, including Bimagrumab (BYM338; Novartis, Basel, Switzerland), an antibody that targets type II activin receptors, preventing GDF-8 and/or activin signal transduction, thereby stimulating muscle production and strengthening [see clinical trial number NCT01925209 entitled Efficacy and Safety of
Bimagrumab/BYM338 at 52 Weeks on Physical Function, Muscle Strength, Mobility in sIBM Patients (RESILIENT)]. Some compounds and/or compositions of the present disclosure may be used to treat subjects with IBM. In some cases, such compounds and/or compositions may block GDF-8 activity (e.g. through stabilization of GDF-8 GPCs). In addition to IBM, BYM338 is being investigated for treatment of chronic obstructive pulmonary disease (COPD). In some cases, compounds and/or compositions of the present disclosure utilized for IBM treatment, may be used to treat COPD as well. In some cases, compounds and/or compositions of the present disclosure may be administered in combination and/or coordination with BYM338.
Therapeutics for diabetes
[00498] Skeletal muscle uses and stores glucose for fuel. Due to this, skeletal muscle is an important regulator of circulating glucose levels. Uptake of glucose by muscle can be stimulated by either contraction or by insulin stimulation (McPherron et al., 2013. Adipocyte. 2(2):92-8, herein incorporated by reference in its entirety). A recent study by Guo et al (Guo, et al., 2012. Diabetes 61(10):2414-23) found that when GDF-8 receptor-deficient mice were crossed with A-ZIP/F1 mice (a lipodistrophic mouse strain, used as a diabetic model), hybrid off-spring showed reduced levels of blood glucose and improved sensitivity to insulin.
Hyperphagia (excessive eating) was also reduced in these mice. In some embodiments, compound and/or compositions of the present disclosure may be used to treat diabetes and/or hyperphagia. Some such treatments may be used to reduce blood glucose and/or improve insulin sensitivity. In some cases, such treatments may comprise GDF-8 signaling
antagosists, such as one or more antibodies that prevent dissociation of GDF-8 from its prodomain.
Therapeutics for GDF-11 -related indications
[00499] In some embodiments, compounds of the disclosure may be used to treat TGF-β- related indications comprising GDF-11 -related indications. As used herein, a GDF-11 -related indication is a disease, disorder and/or condition related to GDF-11 activity. GDF-11 expression is systemic and its activity is thought to be involved in multiple processes (Lee et al., 2013. PNAS. 110(39):E3713-22, the contents of which are herein incorporated by reference in their entirety). It is believed to be involved in development of multiple tissues, including, but not limited to the retina, kidney, pancreas and olfactory system. It is also believed to be a circulating factor in the blood. Recent studies indicate that GDF- 11 may rejuvenate skeletal muscle, improve cerebral circulation and promote neurogenesis (Sinha, M. et al., 2014. Science Express. 10.1126/science. l251152, p2-6 and Katsimpardi, L. et al., 2014. Science Express. 10.1126/science.1251141, the contents of each of which are herein incorporated by reference in their entirety). In some cases, antibodies of the disclosure may promote skeletal muscle rejuvenation, improve cerebral circulation and promote neurogenesis by promoting the release of GDF-11 growth factor from latent complexes.
[00500] Although its role is somewhat controversial, GDF-11 is thought to be involved in the regulation of erythropoiesis with both positive and negative regulation being described in scientific literature. Carrancio et al and Suragani et al (Carrancio, S. et al., 2014. Br J
Haematol. 165(6):870-82 and Suragani, R.N.V.S. et al. 2014. Blood. 123(25): 3864-72, the contents of each of which are herein incorporated by reference in their entirety) demonstrate that a GDF-11 ligand trap, comprising an activin receptor IIA extracellular domain
(SOTATERCEPT®), enhances erythropoiesis. A similar agent shown to bind GDF-11 and modified to reduce affinity for activin, ACE-536, was also shown to stimulate erythropoiesis (Suragani, R.N.V.S. et al. 2014. Nature Medicine. 20(4): 408-17, the contents of which are herein incorporated by reference in their entirety). Further studies demonstrate short term increases in hemoglobin levels in patients receiving SOTATERCEPT® (El-Shahawy, M. et al., 2014. Poster #81, National Kidney Foundation (NKF) 2014 Spring Clinical Meeting). In some cases, GDF-11 inhibiting antibodies of the disclosure may be used according to the methods described in these studies to stimulate erythropoiesis and treat anemia and/or β- thalassemia.
Veterinary applications
[00501] In some embodiments, it is contemplated that compositions and methods of the disclosure will find utility in the area of veterinary care including the care and treatment of non-human vertebrates. As described herein, the term "vertebrate" includes all vertebrates including, but not limited to fish, amphibians, birds, reptiles and mammals (including, but not limited to alpaca, banteng, bison, camel, cat, cattle, deer, dog, donkey, gayal, goat, guinea pig, horse, llama, mice, monkeys, mule, pig, rabbit, rats, reindeer, sheep water buffalo, yak and humans). As used herein the term "non-human vertebrate" refers to any vertebrate with the exception of humans (i.e. Homo sapiens). Exemplary non-human vertebrates include wild and domesticated species such as companion animals and livestock. Livestock include domesticated animals raised in an agricultural setting to produce materials such as food, labor, and derived products such as fiber and chemicals. Generally, livestock includes all mammals, avians and fish having potential agricultural significance. In particular, four- legged slaughter animals include steers, heifers, cows, calves, bulls, cattle, swine and sheep. Bioprocessing
[00502] In some embodiments, the present disclosure provides methods for producing one or more biological products in host cells by contacting such cells with compounds and/or compositions of the present disclosure capable of modulating expression of target genes, or altering the level of growth factor signaling molecules wherein such modulation or alteration enhances production of biological products. According to the present disclosure,
bioprocessing methods may be improved by using one or more compounds and/or compositions of the present disclosure. They may also be improved by supplementing, replacing or adding one or more compounds and/or compositions.
Pharmaceutical compositions
[00503] The pharmaceutical compositions described herein may be characterized by one or more of bioavailability, therapeutic window and/or volume of distribution.
Bioavailability
[00504] In some embodiments, pharmaceutical compositions comprise complexes of compounds and/or compositions of the present disclosure with GPCs. In such embodiments, complexes may be implanted at desired therapeutic sites where steady dissociation of growth factors from complexes may occur over a desired period of time. In some embodiments, implantation complexes may be carried out in association with sponge and/or bone-like matrices. Such implantations may include, but are not limited to dental implant sites and/or sites of bone repair.
[00505] In some embodiments, compounds and/or compositions of the present disclosure are made in furin-deficient cells. GPCs produced in such cells may be useful for treatment in areas where release is slowed due to the fact that furin cleavage in vivo is rate-limiting during GPC processing. In some embodiments, one or more tolloid and/or furin sites in GPCs are mutated, slowing the action of endogenous tolloid and/or furin proteases. In such
embodiments, growth factor release may be slowed (e.g. at sites of implantation).
[00506] Antibodies of the present disclosure, when formulated into compositions with delivery/formulation agents or vehicles as described herein, may exhibit increased bioavailability as compared to compositions lacking delivery agents as described herein. As used herein, the term "bioavailability" refers to the systemic availability of a given amount of a particular agent administered to a subject. Bioavailability may be assessed by measuring the area under the curve (AUC) or the maximum serum or plasma concentration (Cmax) of the unchanged form of a compound following administration of the compound to a mammal. AUC is a determination of the area under the curve plotting the serum or plasma
concentration of a compound along the ordinate (Y-axis) against time along the abscissa (X- axis). Generally, the AUC for a particular compound may be calculated using methods known to those of ordinary skill in the art and as described in G. S. Banker, Modern
Pharmaceutics, Drugs and the Pharmaceutical Sciences, v. 72, Marcel Dekker, New York, Inc., 1996, the contents of which are herein incorporated by reference in their entirety.
[00507] Cmax values are maximum concentrations of compounds achieved in serum or plasma of a subject following administration of compounds to the subject. Cmax values of particular compounds may be measured using methods known to those of ordinary skill in the art. As used herein, the phrases "increasing bioavailability" or "improving the
pharmacokinetics," refer to actions that may increase the systemic availability of a compounds and/or compositions of the present disclosure (as measured by AUC, Cmax, or Cmin) in a subject. In some embodiments, such actions may comprise co-administration with one or more delivery agents as described herein. In some embodiments, the bioavailability of compounds and/or compositions may increase by at least about 2%, at least about 5%, at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95% or about 100%.
Therapeutic window
[00508] Compounds and/or compositions of the present disclosure, when formulated with one or more delivery agents as described herein, may exhibit increases in the therapeutic window of compound and/or composition administration as compared to the therapeutic window of compounds and/or compositions administered without one or more delivery agents as described herein. As used herein, the term "therapeutic window" refers to the range of plasma concentrations, or the range of levels of therapeutically active substance at the site of action, with a high probability of eliciting a therapeutic effect. In some embodiments, therapeutic windows of compounds and/or compositions when co-administered with one or more delivery agent as described herein may increase by at least about 2%, at least about 5%, at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95% or about 100%.
Volume of distribution
[00509] Compounds and/or compositions of the present disclosure, when formulated with one or more delivery agents as described herein, may exhibit an improved volume of distribution (VdiSt), e.g., reduced or targeted, relative to formulations lacking one or more delivery agents as described herein. Vdist relates the amount of an agent in the body to the concentration of the same agent in the blood or plasma. As used herein, the term "volume of distribution" refers to the fluid volume that would be required to contain the total amount of an agent in the body at the same concentration as in the blood or plasma: Vdist equals the amount of an agent in the body/concentration of the agent in blood or plasma. For example, for a 10 mg dose of a given agent and a plasma concentration of 10 mg/L, the volume of distribution would be 1 liter. The volume of distribution reflects the extent to which an agent is present in the extravascular tissue. Large volumes of distribution reflect the tendency of agents to bind to the tissue components as compared with plasma proteins. In clinical settings, Vdist may be used to determine loading doses to achieve steady state concentrations. In some embodiments, volumes of distribution of compounds and/or compositions of the present disclosure when co-administered with one or more delivery agents as described herein may decrease at least about 2%, at least about 5%, at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%.
Formulation, administration, delivery and dosing
[00510] In some embodiments, compounds and/or compositions of the present disclosure are pharmaceutical compositions. As used herein, the term "pharmaceutical composition" refers to a compound and/or composition of the present disclosure that has been formulated with one or more pharmaceutically acceptable excipients. In some embodiments,
pharmaceutical compositions may optionally comprise one or more additional active substances, e.g. therapeutically and/or prophylactically active substances. General
considerations in the formulation and/or manufacture of pharmaceutical agents may be found, for example, in Remington: The Science and Practice of Pharmacy 21st ed., Lippincott Williams & Wilkins, 2005 (incorporated herein by reference). [00511] In some embodiments, compositions may be administered to humans, human patients or subjects. For the purposes of the present disclosure, the phrase "active ingredient" generally refers to compounds and/or compositions of the present disclosure to be delivered as described herein.
[00512] Although the descriptions of pharmaceutical compositions provided herein are principally directed to pharmaceutical compositions which are suitable for administration to humans, it will be understood by the skilled artisan that such compositions are generally suitable for administration to other subjects, e.g., to non-human animals, e.g. non-human mammals. Modification of pharmaceutical compositions suitable for administration to humans in order to render the compositions suitable for administration to various animals is well understood, and the ordinarily skilled veterinary pharmacologist can design and/or perform such modification with merely ordinary, if any, experimentation. Subjects to which administration of pharmaceutical compositions is contemplated include, but are not limited to, humans and/or other primates; mammals, including commercially relevant mammals such as cattle, pigs, horses, sheep, cats, dogs, mice, and/or rats; and/or birds, including
commercially relevant birds such as poultry, chickens, ducks, geese, and/or turkeys.
[00513] In some embodiments, formulations of the pharmaceutical compositions described herein may be prepared by any method known or hereafter developed in the art of pharmacology. In general, such preparatory methods include the step of bringing active ingredients into association with excipients and/or one or more other accessory ingredients, and then, if necessary and/or desirable, dividing, shaping and/or packaging products into desired single- or multi-dose units.
[00514] In some embodiments, pharmaceutical compositions of the present disclosure may be prepared, packaged, and/or sold in bulk, as single unit doses, and/or as a plurality of single unit doses. As used herein, the term "unit dose" refers to a discrete amount of the
pharmaceutical composition comprising a predetermined amount of active ingredient.
Amounts of active ingredient are generally equal to the dosage of active ingredients which would be administered to subjects and/or convenient fractions of such a dosages such as, for example, one-half or one-third of such a dosages.
[00515] In some embodiments, relative amounts of active ingredients, pharmaceutically acceptable excipients, and/or any additional ingredients in pharmaceutical compositions of the present disclosure may vary, depending upon identity, size, and/or condition of subjects to be treated and further depending upon routes by which compositions are to be administered. By way of example, compositions may comprise between about 0.1% and 100%, e.g., from about 0.5% to about 50%, from about 1% to about 30%, from about 5% to about 80% or at least 80% (w/w) active ingredient. In some embodiments, active ingredients are antibodies directed toward regulatory elements and/or GPCs.
Formulations
[00516] Compounds and/or compositions of the present disclosure may be formulated using one or more excipients to: (1) increase stability; (2) increase cell permeability; (3) permit the sustained or delayed release (e.g., of compounds and/or growth factors from such formulations); and/or (4) alter the biodistribution (e.g., target compounds to specific tissues or cell types). In addition to traditional excipients such as any and all solvents, dispersion media, diluents, liquid vehicles, dispersion aids, suspension aids, surface active agents, isotonic agents, thickening agents, emulsifying agents and preservatives, formulations of the present disclosure may comprise, without limitation, liposomes, lipid nanoparticles, polymers, lipoplexes, core- shell nanoparticles, peptides, proteins, cells transfected with the compounds and/or compositions of the present disclosure (e.g., for transplantation into subjects) and combinations thereof.
Excipients
[00517] Various excipients for formulating pharmaceutical compositions and techniques for preparing the composition are known in the art (see Remington: The Science and Practice of Pharmacy, 21st Edition, A. R. Gennaro, Lippincott, Williams & Wilkins, Baltimore, MD, 2006; incorporated herein by reference).
[00518] In some embodiments, the use of conventional excipient media are contemplated within the scope of the present disclosure, except insofar as any conventional excipient media may be incompatible with substances and/or their derivatives, such as by producing any undesirable biological effects or otherwise interacting in deleterious manners with any other component(s) of pharmaceutical compositions.
[00519] Formulations of pharmaceutical compositions described herein may be prepared by any method known or hereafter developed in the art of pharmacology. In general, such preparatory methods include steps of associating active ingredients with excipients and/or other accessory ingredients.
[00520] Pharmaceutical compositions, in accordance with the present disclosure, may be prepared, packaged, and/or sold in bulk, as single unit doses, and/or as a plurality of single unit doses. [00521] Relative amounts of active ingredients, pharmaceutically acceptable excipients, and/or additional ingredients in pharmaceutical compositions of the present disclosure may vary, depending upon identity, size, and/or condition of subjects being treated and further depending upon routes by which pharmaceutical compositions may be administered.
[00522] In some embodiments, pharmaceutically acceptable excipient are at least 95%, at least 96%, at least 97%, at least 98%, at least 99% or 100% pure. In some embodiments, excipients are approved for use in humans and/or for veterinary use. In some embodiments, excipients are approved by the United States Food and Drug Administration. In some embodiments, excipients are pharmaceutical grade. In some embodiments, excipients meet the standards of the United States Pharmacopoeia (USP), the European Pharmacopoeia (EP), the British Pharmacopoeia, and/or the International Pharmacopoeia.
[00523] In some embodiments, pharmaceutically acceptable excipients of the present disclosure may include, but are not limited to, inert diluents, dispersing and/or granulating agents, surface active agents and/or emulsifiers, disintegrating agents, binding agents, preservatives, buffering agents, lubricating agents, and/or oils. Such excipients may optionally be included in pharmaceutical compositions.
[00524] Exemplary diluents include, but are not limited to, calcium carbonate, sodium carbonate, calcium phosphate, dicalcium phosphate, calcium sulfate, calcium hydrogen phosphate, sodium phosphate lactose, sucrose, cellulose, microcrystalline cellulose, kaolin, mannitol, sorbitol, inositol, sodium chloride, dry starch, cornstarch, powdered sugar, etc., and/or combinations thereof.
[00525] Exemplary granulating and/or dispersing agents include, but are not limited to, potato starch, corn starch, tapioca starch, sodium starch glycolate, clays, alginic acid, guar gum, citrus pulp, agar, bentonite, cellulose and wood products, natural sponge, cation- exchange resins, calcium carbonate, silicates, sodium carbonate, cross-linked polyvinylpyrrolidone) (crospovidone), sodium carboxymethyl starch (sodium starch glycolate), carboxymethyl cellulose, cross-linked sodium carboxymethyl cellulose (croscarmellose), methylcellulose, pregelatinized starch (starch 1500), microcrystalline starch, water insoluble starch, calcium carboxymethyl cellulose, magnesium aluminum silicate (VEEGUM®), sodium lauryl sulfate, quaternary ammonium compounds, etc., and/or combinations thereof.
[00526] Exemplary surface active agents and/or emulsifiers include, but are not limited to, natural emulsifiers (e.g. acacia, agar, alginic acid, sodium alginate, tragacanth, chondrux, cholesterol, xanthan, pectin, gelatin, egg yolk, casein, wool fat, cholesterol, wax, and lecithin), colloidal clays (e.g. bentonite [aluminum silicate] and VEEGUM® [magnesium aluminum silicate]), long chain amino acid derivatives, high molecular weight alcohols (e.g. stearyl alcohol, cetyl alcohol, oleyl alcohol, triacetin monostearate, ethylene glycol distearate, glyceryl monostearate, and propylene glycol monostearate, polyvinyl alcohol), carbomers (e.g. carboxy polymethylene, polyacrylic acid, acrylic acid polymer, and carboxyvinyl polymer), carrageenan, cellulosic derivatives (e.g. carboxymethylcellulose sodium, powdered cellulose, hydroxymethyl cellulose, hydroxypropyl cellulose, hydroxypropyl methylcellulose, methylcellulose), sorbitan fatty acid esters (e.g. polyoxyethylene sorbitan monolaurate
[TWEEN®20], polyoxyethylene sorbitan [TWEENn®60], polyoxyethylene sorbitan monooleate [TWEEN®80], sorbitan monopalmitate [SPAN®40], sorbitan monostearate
[Span®60], sorbitan tristearate [Span®65], glyceryl monooleate, sorbitan monooleate
[SPAN®80]), polyoxyethylene esters (e.g. polyoxyethylene monostearate [MYRJ®45], polyoxyethylene hydrogenated castor oil, polyethoxylated castor oil, polyoxymethylene stearate, and SOLUTOL®), sucrose fatty acid esters, polyethylene glycol fatty acid esters (e.g. CREMOPHOR®), polyoxyethylene ethers, (e.g. polyoxyethylene lauryl ether
[BRIJ®30]), poly(vinyl-pyrrolidone), diethylene glycol monolaurate, triethanolamine oleate, sodium oleate, potassium oleate, ethyl oleate, oleic acid, ethyl laurate, sodium lauryl sulfate, PLUORINC®F 68, POLOXAMER®188, cetrimonium bromide, cetylpyridinium chloride, benzalkonium chloride, docusate sodium, etc. and/or combinations thereof.
[00527] Exemplary binding agents include, but are not limited to, starch (e.g. cornstarch and starch paste); gelatin; sugars (e.g. sucrose, glucose, dextrose, dextrin, molasses, lactose, lactitol, mannitol),; natural and synthetic gums (e.g. acacia, sodium alginate, extract of Irish moss, panwar gum, ghatti gum, mucilage of isapol husks, carboxymethylcellulose, methylcellulose, ethylcellulose, hydroxyethylcellulose, hydroxypropyl cellulose,
hydroxypropyl methylcellulose, microcrystalline cellulose, cellulose acetate, polyvinylpyrrolidone), magnesium aluminum silicate (Veegum®), and larch arabogalactan); alginates; polyethylene oxide; polyethylene glycol; inorganic calcium salts; silicic acid;
polymethacrylates; waxes; water; alcohol; etc. ; and combinations thereof.
[00528] Exemplary preservatives may include, but are not limited to, antioxidants, chelating agents, antimicrobial preservatives, antifungal preservatives, alcohol preservatives, acidic preservatives, and/or other preservatives. Exemplary antioxidants include, but are not limited to, alpha tocopherol, ascorbic acid, acorbyl palmitate, butylated hydroxyanisole, butylated hydroxytoluene, monothioglycerol, potassium metabisulfite, propionic acid, propyl gallate, sodium ascorbate, sodium bisulfite, sodium metabisulfite, and/or sodium sulfite. Exemplary chelating agents include ethylenediaminetetraacetic acid (EDTA), citric acid monohydrate, disodium edetate, dipotassium edetate, edetic acid, fumaric acid, malic acid, phosphoric acid, sodium edetate, tartaric acid, and/or trisodium edetate. Exemplary antimicrobial preservatives include, but are not limited to, benzalkonium chloride, benzethonium chloride, benzyl alcohol, bronopol, cetrimide, cetylpyridinium chloride, chlorhexidine, chlorobutanol, chlorocresol, chloroxylenol, cresol, ethyl alcohol, glycerin, hexetidine, imidurea, phenol, phenoxyethanol, phenylethyl alcohol, phenylmercuric nitrate, propylene glycol, and/or thimerosal. Exemplary antifungal preservatives include, but are not limited to, butyl paraben, methyl paraben, ethyl paraben, propyl paraben, benzoic acid, hydroxybenzoic acid, potassium benzoate, potassium sorbate, sodium benzoate, sodium propionate, and/or sorbic acid. Exemplary alcohol preservatives include, but are not limited to, ethanol, polyethylene glycol, phenol, phenolic compounds, bisphenol, chlorobutanol, hydroxybenzoate, and/or phenylethyl alcohol. Exemplary acidic preservatives include, but are not limited to, vitamin A, vitamin C, vitamin E, beta-carotene, citric acid, acetic acid, dehydroacetic acid, ascorbic acid, sorbic acid, and/or phytic acid. Other preservatives include, but are not limited to, tocopherol, tocopherol acetate, deteroxime mesylate, cetrimide, butylated hydroxyanisol (BHA), butylated hydroxytoluened (BHT), ethylenediamine, sodium lauryl sulfate (SLS), sodium lauryl ether sulfate (SLES), sodium bisulfite, sodium
metabisulfite, potassium sulfite, potassium metabisulfite, GLYDANT PLUS®, PHENONIP®, methylparaben, GERMALL®115, GERMABEN®II, NEOLONE™, KATHON™, and/or EUXYL®.
[00529] Exemplary buffering agents include, but are not limited to, citrate buffer solutions, acetate buffer solutions, phosphate buffer solutions, ammonium chloride, calcium carbonate, calcium chloride, calcium citrate, calcium glubionate, calcium gluceptate, calcium gluconate, D-gluconic acid, calcium glycerophosphate, calcium lactate, propanoic acid, calcium levulinate, pentanoic acid, dibasic calcium phosphate, phosphoric acid, tribasic calcium phosphate, calcium hydroxide phosphate, potassium acetate, potassium chloride, potassium gluconate, potassium mixtures, dibasic potassium phosphate, monobasic potassium
phosphate, potassium phosphate mixtures, sodium acetate, sodium bicarbonate, sodium chloride, sodium citrate, sodium lactate, dibasic sodium phosphate, monobasic sodium phosphate, sodium phosphate mixtures, tromethamine, magnesium hydroxide, aluminum hydroxide, alginic acid, pyrogen-free water, isotonic saline, Ringer' s solution, ethyl alcohol, etc. , and/or combinations thereof.
[00530] Exemplary lubricating agents include, but are not limited to, magnesium stearate, calcium stearate, stearic acid, silica, talc, malt, glyceryl behanate, hydrogenated vegetable oils, polyethylene glycol, sodium benzoate, sodium acetate, sodium chloride, leucine, magnesium lauryl sulfate, sodium lauryl sulfate, etc. , and combinations thereof.
[00531] Exemplary oils include, but are not limited to, almond, apricot kernel, avocado, babassu, bergamot, black current seed, borage, cade, camomile, canola, caraway, carnauba, castor, cinnamon, cocoa butter, coconut, cod liver, coffee, corn, cotton seed, emu, eucalyptus, evening primrose, fish, flaxseed, geraniol, gourd, grape seed, hazel nut, hyssop, isopropyl myristate, jojoba, kukui nut, lavandin, lavender, lemon, litsea cubeba, macademia nut, mallow, mango seed, meadowfoam seed, mink, nutmeg, olive, orange, orange roughy, palm, palm kernel, peach kernel, peanut, poppy seed, pumpkin seed, rapeseed, rice bran, rosemary, safflower, sandalwood, sasquana, savoury, sea buckthorn, sesame, shea butter, silicone, soybean, sunflower, tea tree, thistle, tsubaki, vetiver, walnut, and wheat germ oils. Exemplary oils include, but are not limited to, butyl stearate, caprylic triglyceride, capric triglyceride, cyclomethicone, diethyl sebacate, dimethicone 360, isopropyl myristate, mineral oil, octyldodecanol, oleyl alcohol, silicone oil, and/or combinations thereof.
[00532] Excipients such as cocoa butter and suppository waxes, coloring agents, coating agents, sweetening, flavoring, and/or perfuming agents can be present in the composition, according to the judgment of the formulator.
Formulation vehicles: liposomes, lipoplexes, and lipid nanoparticles
[00533] Compounds and/or compositions of the present disclosure may be formulated using one or more liposomes, lipoplexes and/or lipid nanoparticles. In some embodiments, pharmaceutical compositions comprise liposomes. Liposomes are artificially-prepared vesicles which may primarily be composed of a lipid bilayer and may be used as delivery vehicles for the administration of nutrients and pharmaceutical formulations. Liposomes may be of different sizes such as, but not limited to, multilamellar vesicles (MLVs) which may be hundreds of nanometers in diameter and may contain a series of concentric bilayers separated by narrow aqueous compartments, small unicellular vesicle (SUVs) which may be smaller than 50 nm in diameter and large unilamellar vesicle (LUVs) which may be between 50 and 500 nm in diameter. Liposome components may include, but are not limited to, opsonins or ligands in order to improve the attachment of liposomes to unhealthy tissue or to activate events such as, but not limited to, endocytosis. Liposomes may comprise low or high pH. In some embodiments, liposome pH may be varied in order to improve delivery of
pharmaceutical formulations. [00534] In some embodiments, liposome formation may depend on physicochemical characteristics such as, but not limited to, the pharmaceutical formulation entrapped, liposomal ingredients, the nature of the medium in which lipid vesicles are dispersed, the effective concentration of entrapped substances, potential toxicity of entrapped substances, additional processes involved during the application and/or delivery of vesicles, optimization size, polydispersity, shelf-life of vesicles for the intended application, batch-to-batch reproducibility and possibility of large-scale production of safe and efficient liposomal products.
[00535] In some embodiments, formulations may be assembled or compositions altered such that they are passively or actively directed to different cell types in vivo.
[00536] In some embodiments, formulations may be selectively targeted through expression of different ligands on formulation surfaces as exemplified by, but not limited by, folate, transferrin, N-acetylgalactosamine (GalNAc), and antibody targeted approaches.
[00537] In some embodiments, pharmaceutical compositions of the present disclosure may be formulated with liposomes, lipoplexes and/or lipid nanoparticles to improve efficacy of function. Such formulations may be able to increase cell transfection by pharmaceutical compositions. In some embodiments, liposomes, lipoplexes, or lipid nanoparticles may be used to increase pharmaceutical composition stability.
[00538] In some embodiments, liposomes are specifically formulated for pharmaceutical compositions comprising one or more antibodies. Such liposomes may be prepared according to techniques known in the art, such as those described by Eppstein et al. (Eppstein, D.A. et al., Biological activity of liposome-encapsulated murine interferon gamma is mediated by a cell membrane receptor. Proc Natl Acad Sci U S A. 1985 Jun;82(l l):3688-92); Hwang et al. (Hwang, K.J. et al., Hepatic uptake and degradation of unilamellar sphingomyelin/cholesterol liposomes: a kinetic study. Proc Natl Acad Sci U S A. 1980 Jul;77(7):4030-4); US 4,485,045 and US 4,544,545. Production of liposomes with sustained circulation time are also described in US 5,013,556.
[00539] In some embodiments, liposomes of the present disclosure comprising antibodies may be generated using reverse phase evaporation utilizing lipids such as
phosphatidylcholine, cholesterol as well as phosphatidylethanolamine that have been polyethylene glycol-derivatized. Filters with defined pore size are used to extrude liposomes of the desired diameter. In another embodiment, compounds and/or compositions of the present disclosure may be conjugated to external surfaces of liposomes by disulfide interchange reactions as is described by Martin et al. (Martin, F.J. et al., Irreversible coupling of immunoglobulin fragments to preformed vesicles. An improved method for liposome targeting. J Biol Chem. 1982 Jan 10;257(l):286-8).
Formulation vehicles: polymers and nanoparticles
[00540] Compounds and/or compositions of the present disclosure may be formulated using natural and/or synthetic polymers. Non-limiting examples of polymers which may be used for delivery include, but are not limited to DMRI7DOPE, poloxamer, chitosan, cyclodextrin, and poly(lactic-co-glycolic acid) (PLGA) polymers. In some embodiments, polymers may be biodegradable.
[00541] In some embodiments, polymer formulation may permit sustained and/or delayed release of compounds and/or compositions (e.g., following intramuscular and/or
subcutaneous injection). Altered release profile for compounds and/or compositions of the present disclosure may result in, for example, compound release over an extended period of time. Polymer formulations may also be used to increase the stability of compounds and/or compositions of the present disclosure.
[00542] In some embodiments, polymer formulations may be selectively targeted through expression of different ligands as exemplified by, but not limited by, folate, transferrin, and N-acetylgalactosamine (GalNAc) (Benoit, D.S. et al., Synthesis of folate-functionalized RAFT polymers for targeted siRNA delivery. Biomacromolecules. 2011 12:2708-14;
Rozema, D.B. et al., Dynamic polyconjugates for targeted in vivo delivery of siRNA to hepatocytes. Proc Natl Acad Sci U S A. 2007 104: 12982-12887; Davis, M.E. et al., The first targeted delivery of siRNA in humans via a self-assembling, cyclodextrin polymer-based nanoparticle: from concept to clinic. Mol Pharm. 2009 6:659-668; Davis, M.E. et al., Evidence of RNAi in humans from systemically administered siRNA via targeted
nanoparticles. Nature. 2010. 464: 1067-70; the contents of each of which are herein incorporated by reference in their entirety).
[00543] Compounds and/or compositions of the present disclosure may be formulated as nanoparticles using combinations of polymers, lipids, and/or other biodegradable agents, such as, but not limited to, calcium phosphates. In some embodiments, components may be combined in core-shells, hybrids, and/or layer-by-layer architectures, to allow for fine-tuning of nanoparticle structure, so delivery may be enhanced. For antibodies of the present disclosure, systems based on poly(2-(methacryloyloxy)ethyl phosphorylcholine)-block-(2- (diisopropylamino)ethyl methacrylate), (PMPC-PDPA), a pH sensitive diblock copolymer that self-assembles to form nanometer- sized vesicles, also known as polymersomes, at physiological pH may be used. These polymersomes have been shown to successfully deliver relatively high antibody payloads within live cells. (Massignani, M. et al., Cellular delivery of antibodies: effective targeted subcellular imaging and new therapeutic tool. Nature
Proceedings. 2010. pl-17).
[00544] In some embodiments, PEG-charge-conversional polymers (Pitella, F. et al., Enhanced endosomal escape of siRNA-incorporating hybrid nanoparticles from calcium phosphate and PEG-block charge-conversional polymer for efficient gene knockdown with negligible cytotoxicity. Biomaterials. 2011 32:3106-14) may be used to form nanoparticles for delivery of compounds and/or compositions of the present disclosure. In some
embodiments, PEG-charge-conversional polymers may improve upon PEG-polyanion block copolymers by being cleaved into polycations at acidic pH, thus enhancing endosomal escape.
[00545] In some embodiments, complexation, delivery and/or internalization of polymeric nanoparticles may be precisely controlled by altering chemical compositions in both core and shell nanoparticle components (Siegwart, D.J. et al., Combinatorial synthesis of chemically diverse core-shell nanoparticles for intracellular delivery. Proc Natl Acad Sci U S A. 2011 108: 12996-3001).
[00546] In some embodiments, matrices of poly(ethylene-co-vinyl acetate), are used to deliver compounds and/or compositions of the disclosure. Such matrices have bee described by others (Sherwood, J.K. et al., Controlled antibody delivery systems. Nature
Biotechnology. 1992. 10: 1446-9).
Antibody formulations
[00547] Antibodies of the present disclosure may be formulated for intravenous
administration or extravascular administration (Daugherty, et al., Formulation and delivery issues for monoclonal antibody therapeutics. Adv Drug Deliv Rev. 2006 Aug 7;58(5-6):686- 706 and US patent application publication number US2011/0135570, the contents of each of which are herein incorporated by reference in their entirety). Extravascular administration routes may include, but are not limited to subcutaneous administration, intraperitoneal administration, intracerebral administration, intraocular administration, intralesional administration, topical administration and intramuscular administration.
[00548] In some embodiments, antibody structures may be modified to improve
effectiveness as therapeutics. Improvements may include, but are not limited to improved thermodynamic stability, reduced Fc receptor binding properties and/or imporved folding efficiency. Modifications may include, but are not limited to amino acid substitutions, glycosylation, palmitoylation and/or protein conjugation.
[00549] In some embodiments, antibodies of the present disclosure may be formulated with antioxidants to reduce antibody oxidation. Antibodies of the present disclosure may also be formulated with additives to reduce protein aggregation. Such additives may include, but are not limited to albumin, amino acids, sugars, urea, guanidinium chloride, polyalchohols, polymers (such as polyethylene glycol and dextrans), surfactants (including, but not limited to polysorbate 20 and polysorbate 80) or even other antibodies.
[00550] In some embodiments, antibodies of the present disclosure may be formulated to reduce the impact of water on antibody structure and function. Antibody preparartions in such formulations may be may be lyophilized. Formulations subject to lyophilization may include carbohydrates or polyol compounds to protect and/or stabilize antibody structure. Such compounds may include, but are not limited to sucrose, trehalose and mannitol.
[00551] In some embodiments, antibodies of the present disclosure may be formulated with polymers. In some embodiments, polymer formulations may comprise hydrophobic polymers. Such polymers may be microspheres formulated with polylactide-co-glycolide through solid-in-oil-in-water encapsulation methods. In some embodiments, microspheres comprising ethylene- vinyl acetate copolymer may also be used for antibody delivery and/or to extend the time course of antibody release at sites of delivery. In some embodiments, polymers may be aqueous gels. Such gels may, for example, comprise
carboxymethylcellulose. In some embodiments, aqueous gels may also comprise hyaluronic acid hydrogels. In some embodiments, antibodies may be covalently linked to such gels through hydrazone linkages that allow for sustained delivery in tissues, including but not limited to tissues of the central nervous system.
Formulation vehicles: peptides and proteins
[00552] Compounds and/or compositions of the present disclosure may be formulated with peptides and/or proteins. In some embodiments, peptides such as, but not limited to, cell penetrating peptides and/or proteins/peptides that enable intracellular delivery may be used to deliver pharmaceutical formulations. Non-limiting examples of a cell penetrating peptides which may be used with pharmaceutical formulations of the present disclosure include cell- penetrating peptide sequences attached to polycations that facilitates delivery to the intracellular space, e.g., HIV-derived TAT peptide, penetratins, transportans, or hCT derived cell-penetrating peptides (see, e.g. Caron, N.J. et al., Intracellular delivery of a Tat-eGFP fusion protein into muscle cells. Mol Ther. 2001. 3(3):310-8; Langel, U., Cell-Penetrating Peptides: Processes and Applications, CRC Press, Boca Raton FL, 2002; El-Andaloussi, S. et al., Cell-penetrating peptides: mechanisms and applications. Curr Pharm Des. 2003.
11(28):3597-611; and Deshayes, S. et al., Cell-penetrating peptides: tools for intracellular delivery of therapeutics. Cell Mol Life Sci. 2005. 62(16): 1839-49, the contents of each of which are herein incorporated by reference in their entirety). Compounds and/or
compositions of the present disclosure may also be formulated to include cell penetrating agents, e.g., liposomes, which enhance delivery of the compositions to intracellular spaces. Compounds and/or compositions of the present disclosure may be complexed with peptides and/or proteins such as, but not limited to, peptides and/or proteins from Aileron
Therapeutics (Cambridge, MA) and Permeon Biologies (Cambridge, MA) in order to enable intracellular delivery (Cronican, J.J. et al., Potent delivery of functional proteins into mammalian cells in vitro and in vivo using a supercharged protein. ACS Chem Biol. 2010. 5:747-52; McNaughton, B.R. et al., Mammalian cell penetration, siRNA transfection, and DNA transfection by supercharged proteins. Proc Natl Acad Sci, USA. 2009. 106:6111-6; Verdine, G.L. et al., Stapled peptides for intracellular drug targets. Methods Enzymol. 2012. 503:3-33; the contents of each of which are herein incorporated by reference in their entirety).
[00553] In some embodiments, the cell-penetrating polypeptides may comprise first and second domains. First domains may comprise supercharged polypeptides. Second domains may comprise protein-binding partner. As used herein, protein-binding partners may include, but are not limited to, antibodies and functional fragments thereof, scaffold proteins and/or peptides. Cell-penetrating polypeptides may further comprise intracellular binding partners for protein-binding partners. In some embodiments, cell-penetrating polypeptides may be capable of being secreted from cells where compounds and/or compositions of the present disclosure may be introduced.
[00554] Compositions of the present disclosure comprising peptides and/or proteins may be used to increase cell transfection and/or alter compound/composition biodistribution (e.g., by targeting specific tissues or cell types).
Formulation vehicles: cells
[00555] Cell-based formulations of compounds and/or compositions of the present disclosure may be used to ensure cell transfection (e.g., in cellular carriers) or to alter biodistribution (e.g., by targeting cell carriers to specific tissues or cell types). Cell transfer methods
[00556] A variety of methods are known in the art and suitable for introduction of nucleic acids or proteins into cells, including viral and non-viral mediated techniques. Examples of typical non-viral mediated techniques include, but are not limited to, electroporation, calcium phosphate mediated transfer, nucleofection, sonoporation, heat shock, magnetofection, liposome mediated transfer, microinjection, microprojectile mediated transfer (nanoparticles), cationic polymer mediated transfer (DEAE-dextran, polyethylenimine, polyethylene glycol (PEG) and the like) or cell fusion.
[00557] The technique of sonoporation, or cellular sonication, is the use of sound (e.g., ultrasonic frequencies) for modifying the permeability of cell plasma membranes.
Sonoporation methods are known to those in the art and are used to deliver nucleic acids in vivo (Yoon, C.S. et al., Ultrasound-mediated gene delivery. Expert Opin Drug Deliv. 2010 7:321-30; Postema, M. et al., Ultrasound-directed drug delivery. Curr Pharm Biotechnol. 2007 8:355-61; Newman, CM. et al., Gene therapy progress and prospects: ultrasound for gene transfer. Gene Ther. 2007. 14(6):465-75; the contents of each of which are herein incorporated by reference in their entirety). Sonoporation methods are known in the art and are also taught for example as they relate to bacteria in US Patent application publication US2010/0196983 and as it relates to other cell types in, for example, US Patent application publication US2010/0009424, the contents of each of which are incorporated herein by reference in their entirety.
[00558] Electroporation techniques are also well known in the art and are used to deliver nucleic acids in vivo and clinically (Andre, F.M. et al., Nucleic acids electrotransfer in vivo: mechanisms and practical aspects. Curr Gene Ther. 2010 10:267-80; Chiarella, P. et al., Application of electroporation in DNA vaccination protocols. Curr Gene Ther. 2010. 10:281- 6; Hojman, P., Basic principles and clinical advancements of muscle electrotransfer. Curr Gene Ther. 2010 10: 128-38; the contents of each of which are herein incorporated by reference in their entirety). In some embodiments, compounds and/or compositions of the present disclosure may be delivered by electroporation.
Administration and delivery
[00559] Compounds and/or compositions of the present disclosure may be administered by any of the standard methods or routes known in the art. Such methods may include any route which results in a therapeutically effective outcome. These include, but are not limited to enteral, gastroenteral, epidural, oral, transdermal, epidural (peridural), intracerebral (into the cerebrum), intracerebroventricular (into the cerebral ventricles), epicutaneous (application onto the skin), intradermal, (into the skin itself), subcutaneous (under the skin), nasal administration (through the nose), intravenous (into a vein), intraarterial (into an artery), intramuscular (into a muscle), intracardiac (into the heart), intraosseous infusion (into the bone marrow), intrathecal (into the spinal canal), intraperitoneal, (infusion or injection into the peritoneum), intravesical infusion, intravitreal, (through the eye), intracavernous injection, ( into the base of the penis), intravaginal administration, intrauterine, extra- amniotic administration, transdermal (diffusion through the intact skin for systemic distribution), transmucosal (diffusion through a mucous membrane), insufflation (snorting), sublingual, sublabial, enema, eye drops (onto the conjunctiva), or in ear drops. In specific embodiments, compounds and/or compositions of the present disclosure may be administered in ways which allow them to cross the blood-brain barrier, vascular barriers, or other epithelial barriers. Methods of formulation and administration may include any of those disclosed in US Pub. No. 2013/0122007, US Pat. No. 8,415,459 or International Pub. No. WO 2011/151432, the contents of each of which are herein incorporated by reference in their entirety. Non-limiting routes of administration for compounds and/or compositions of the present disclosure are described below.
Parenteral and injectible administration
[00560] In some embodiments, compounds and/or compositions of the present disclosure may be administered parenterally. Liquid dosage forms for oral and parenteral administration include, but are not limited to, pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups, and/or elixirs. In addition to active ingredients, liquid dosage forms may comprise inert diluents commonly used in the art such as, for example, water or other solvents, solubilizing agents and emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3- butylene glycol, dimethylformamide, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor, and sesame oils), glycerol, tetrahydrofurfuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof. Besides inert diluents, oral compositions can include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and/or perfuming agents. In certain embodiments for parenteral administration, compositions are mixed with solubilizing agents such as CREMOPHOR®, alcohols, oils, modified oils, glycols, polysorbates, cyclodextrins, polymers, and/or combinations thereof. In other embodiments, surfactants are included such as hydroxypropylcellulo se .
[00561] Injectable preparations, for example, sterile injectable aqueous or oleaginous suspensions may be formulated according to the known art using suitable dispersing agents, wetting agents, and/or suspending agents. Sterile injectable preparations may be sterile injectable solutions, suspensions, and/or emulsions in nontoxic parenterally acceptable diluents and/or solvents, for example, as a solution in 1,3-butanediol. Among the acceptable vehicles and solvents that may be employed are water, Ringer's solution, U.S. P., and isotonic sodium chloride solution. Sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose any bland fixed oil can be employed including synthetic mono- or diglycerides. Fatty acids such as oleic acid can be used in the preparation of injectables.
[00562] Injectable formulations may be sterilized, for example, by filtration through a bacterial -retaining filter, and/or by incorporating sterilizing agents in the form of sterile solid compositions which can be dissolved or dispersed in sterile water or other sterile injectable medium prior to use.
[00563] In order to prolong the effect of active ingredients, it is often desirable to slow the absorption of active ingredients from subcutaneous or intramuscular injections. This may be accomplished by the use of liquid suspensions of crystalline or amorphous material with poor water solubility. The rate of absorption of active ingredients depends upon the rate of dissolution which, in turn, may depend upon crystal size and crystalline form. Alternatively, delayed absorption of a parenterally administered drug form is accomplished by dissolving or suspending the drug in an oil vehicle. Injectable depot forms are made by forming microencapsule matrices of the drug in biodegradable polymers such as polylactide- polyglycolide. Depending upon the ratio of drug to polymer and the nature of the particular polymer employed, the rate of drug release can be controlled. Examples of other
biodegradable polymers include poly(orthoesters) and poly(anhydrides). Depot injectable formulations are prepared by entrapping the drug in liposomes or microemulsions which are compatible with body tissues.
Rectal and vaginal administration
[00564] In some embodiments, compounds and/or compositions of the present disclosure may be administered rectally and/or vaginally. Compositions for rectal or vaginal
administration are typically suppositories which can be prepared by mixing compositions with suitable non-irritating excipients such as cocoa butter, polyethylene glycol or a suppository wax which are solid at ambient temperature but liquid at body temperature and therefore melt in the rectum or vaginal cavity and release the active ingredient.
Oral administration
[00565] In some embodiments, compounds and/or compositions of the present disclosure may be administered orally. Solid dosage forms for oral administration include capsules, tablets, pills, powders, and granules. In such solid dosage forms, an active ingredient is mixed with at least one inert, pharmaceutically acceptable excipient such as sodium citrate or dicalcium phosphate and/or fillers or extenders (e.g. starches, lactose, sucrose, glucose, mannitol, and silicic acid), binders (e.g. carboxymethylcellulose, alginates, gelatin, polyvinylpyrrolidinone, sucrose, and acacia), humectants (e.g. glycerol), disintegrating agents (e.g. agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate), solution retarding agents (e.g. paraffin), absorption accelerators (e.g. quaternary ammonium compounds), wetting agents (e.g. cetyl alcohol and glycerol mono stearate), absorbents (e.g. kaolin and bentonite clay), and lubricants (e.g. talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate), and mixtures thereof. In the case of capsules, tablets and pills, the dosage form may comprise buffering agents.
Topical or transdermal administration
[00566] As described herein, compounds and/or compositions of the present disclosure may be formulated for administration topically. The skin may be an ideal target site for delivery as it is readily accessible. Three routes are commonly considered to deliver compounds and/or compositions of the present disclosure to the skin: (i) topical application (e.g. for
local/regional treatment and/or cosmetic applications); (ii) intradermal injection (e.g. for local/regional treatment and/or cosmetic applications); and (iii) systemic delivery (e.g. for treatment of dermatologic diseases that affect both cutaneous and extracutaneous regions). Compounds and/or compositions of the present disclosure can be delivered to the skin by several different approaches known in the art.
[00567] In some embodiments, the disclosure provides for a variety of dressings (e.g., wound dressings) or bandages (e.g., adhesive bandages) for conveniently and/or effectively carrying out methods of the present disclosure. Typically dressing or bandages may comprise sufficient amounts of compounds and/or compositions of the present disclosure described herein to allow users to perform multiple treatments.
[00568] Dosage forms for topical and/or transdermal administration may include ointments, pastes, creams, lotions, gels, powders, solutions, sprays, inhalants and/or patches. Generally, active ingredients are admixed under sterile conditions with pharmaceutically acceptable excipients and/or any needed preservatives and/or buffers. Additionally, the present disclosure contemplates the use of transdermal patches, which often have the added advantage of providing controlled delivery of compounds and/or compositions of the present disclosure to the body. Such dosage forms may be prepared, for example, by dissolving and/or dispensing compounds and/or compositions in the proper medium. Alternatively or additionally, rates may be controlled by either providing rate controlling membranes and/or by dispersing compounds and/or compositions in a polymer matrix and/or gel.
[00569] Formulations suitable for topical administration include, but are not limited to, liquid and/or semi liquid preparations such as liniments, lotions, oil in water and/or water in oil emulsions such as creams, ointments and/or pastes, and/or solutions and/or suspensions.
[00570] Topically- administrable formulations may, for example, comprise from about 1% to about 10% (w/w) active ingredient, although the concentration of active ingredient may be as high as the solubility limit of the active ingredient in the solvent. Formulations for topical administration may further comprise one or more of the additional ingredients described herein.
Depot administration
[00571] As described herein, in some embodiments, compounds and/or compositions of the present disclosure are formulated in depots for extended release. Generally, specific organs or tissues ("target tissues") are targeted for administration.
[00572] In some aspects of the disclosure, compounds and/or compositions of the present disclosure are spatially retained within or proximal to target tissues. Provided are method of providing compounds and/or compositions to target tissues of mammalian subjects by contacting target tissues (which comprise one or more target cells) with compounds and/or compositions under conditions such that they are substantially retained in target tissues, meaning that at least 10, 20, 30, 40, 50, 60, 70, 80, 85, 90, 95, 96, 97, 98, 99, 99.9, 99.99 or greater than 99.99% of the composition is retained in the target tissues. Advantageously, retention is determined by measuring the amount of compounds and/or compositions that enter one or more target cells. For example, at least 1%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%, 99.9%, 99.99% or greater than 99.99% of compounds and/or compositions administered to subjects are present
intracellularly at a period of time following administration. For example, intramuscular injection to mammalian subjects may be performed using aqueous compositions comprising compounds and/or compositions of the present disclosure and one or more transfection reagent, and retention is determined by measuring the amount of compounds and/or compositions present in muscle cells.
[00573] Certain aspects of the disclosure are directed to methods of providing compounds and/or compositions of the present disclosure to a target tissues of mammalian subjects, by contacting target tissues (comprising one or more target cells) with compounds and/or compositions under conditions such that they are substantially retained in such target tissues. Compounds and/or compositions comprise enough active ingredient such that the effect of interest is produced in at least one target cell. In some embodiments, compounds and/or compositions generally comprise one or more cell penetration agents, although "naked" formulations (such as without cell penetration agents or other agents) are also contemplated, with or without pharmaceutically acceptable carriers.
[00574] In some embodiments, the amount of a growth factor present in cells in a tissue is desirably increased. Preferably, this increase in growth factor is spatially restricted to cells within the target tissue. Thus, provided are methods of increasing the amount of growth factor of interest in tissues of mammalian subjects. In some embodiments, formulations are provided comprising compounds and/or compositions characterized in that the unit quantity provided has been determined to produce a desired level of growth factor of interest in a substantial percentage of cells contained within predetermined volumes of target tissue.
[00575] In some embodiments, formulations comprise a plurality of different compounds and/or compositions, where one or more than one targets biomolecules of interest.
Optionally, formulations may also comprise cell penetration agents to assist in the intracellular delivery of compounds and/or compositions. In such embodiments,
determinations are made of compound and/or composition dose required to target biomolecules of interest in substantial percentages of cells contained within predetermined volumes of the target tissue (generally, without targeting biomolecules of interest in adjacent or distal tissues). Determined doses are then introduced directly into subject tissues. In some embodiments, the disclosure provides for compounds and/or compositions to be delivered in more than one administration or by split dose administration. Pulmonary administration
[00576] In some embodiments, compounds and/or compositions of the present disclosure may be prepared, packaged, and/or sold in formulations suitable for pulmonary
administration. In some embodiments, such administration is via the buccal cavity. In some embodiments, formulations may comprise dry particles comprising active ingredients. In such embodiments, dry particles may have a diameter in the range from about 0.5 nm to about 7 nm or from about 1 nm to about 6 nm. In some embodiments, formulations may be in the form of dry powders for administration using devices comprising dry powder reservoirs to which streams of propellant may be directed to disperse such powder. In some embodiments, self propelling solvent/powder dispensing containers may be used. In such embodiments, active ingredients may be dissolved and/or suspended in low-boiling propellant in sealed containers. Such powders may comprise particles wherein at least 98% of the particles by weight have diameters greater than 0.5 nm and at least 95% of the particles by number have diameters less than 7 nm. Alternatively, at least 95% of the particles by weight have a diameter greater than 1 nm and at least 90% of the particles by number have a diameter less than 6 nm. Dry powder compositions may include a solid fine powder diluent such as sugar and are conveniently provided in a unit dose form.
[00577] Low boiling propellants generally include liquid propellants having a boiling point of below 65 °F at atmospheric pressure. Generally propellants may constitute 50% to 99.9% (w/w) of the composition, and active ingredient may constitute 0.1% to 20% (w/w) of the composition. Propellants may further comprise additional ingredients such as liquid non-ionic and/or solid anionic surfactant and/or solid diluent (which may have particle sizes of the same order as particles comprising active ingredients).
[00578] Pharmaceutical compositions formulated for pulmonary delivery may provide active ingredients in the form of droplets of solution and/or suspension. Such formulations may be prepared, packaged, and/or sold as aqueous and/or dilute alcoholic solutions and/or suspensions, optionally sterile, comprising active ingredients, and may conveniently be administered using any nebulization and/or atomization device. Such formulations may further comprise one or more additional ingredients including, but not limited to, a flavoring agent such as saccharin sodium, a volatile oil, a buffering agent, a surface active agent, and/or a preservative such as methylhydroxybenzoate. Droplets provided by this route of
administration may have an average diameter in the range from about 0.1 nm to about 200 nm. Intranasal, nasal and buccal administration
[00579] In some embodiments, compounds and/or compositions of the present disclosure may be administered nasaly and/or intranasaly. In some embodiments, formulations described herein as being useful for pulmonary delivery may also be useful for intranasal delivery. In some embodiments, formulations for intranasal administration comprise a coarse powder comprising the active ingredient and having an average particle from about 0.2 μιη to 500 μιη. Such formulations are administered in the manner in which snuff is taken, i.e. by rapid inhalation through the nasal passage from a container of the powder held close to the nose.
[00580] Formulations suitable for nasal administration may, for example, comprise from about as little as 0.1% (w/w) and as much as 100% (w/w) of active ingredient, and may comprise one or more of the additional ingredients described herein. A pharmaceutical composition may be prepared, packaged, and/or sold in a formulation suitable for buccal administration. Such formulations may, for example, be in the form of tablets and/or lozenges made using conventional methods, and may, for example, 0.1% to 20% (w/w) active ingredient, the balance comprising an orally dissolvable and/or degradable composition and, optionally, one or more of the additional ingredients described herein. Alternately, formulations suitable for buccal administration may comprise powders and/or an aerosolized and/or atomized solutions and/or suspensions comprising active ingredients. Such powdered, aerosolized, and/or aerosolized formulations, when dispersed, may comprise average particle and/or droplet sizes in the range of from about 0.1 nm to about 200 nm, and may further comprise one or more of any additional ingredients described herein.
Ophthalmic or otic administration
[00581] In some embodiments, compounds and/or compositions of the present disclosure may be prepared, packaged, and/or sold in formulations suitable for ophthalmic and/or otic administration. Such formulations may, for example, be in the form of eye and/or ear drops including, for example, a 0.1/1.0% (w/w) solution and/or suspension of the active ingredient in aqueous and/or oily liquid excipients. Such drops may further comprise buffering agents, salts, and/or one or more other of any additional ingredients described herein. Other ophthalmically-administrable formulations which are useful include those which comprise active ingredients in microcrystalline form and/or in liposomal preparations. Subretinal inserts may also be used as forms of administration.
Payload administration: detectable agents and therapeutic agents [00582] In some embodiments, compounds and/or compositions of the present disclosure may be used in a number of different scenarios in which delivery of a substance (the
"payload") to a biological target is desired, for example delivery of detectable substances for detection of the target, or delivery of therapeutic and/or diagnostic agents. Detection methods may include, but are not limited to, both in vitro and in vivo imaging methods, e.g., immunohistochemistry, bioluminescence imaging (BLI), Magnetic Resonance Imaging (MRI), positron emission tomography (PET), electron microscopy, X-ray computed tomography, Raman imaging, optical coherence tomography, absorption imaging, thermal imaging, fluorescence reflectance imaging, fluorescence microscopy, fluorescence molecular tomographic imaging, nuclear magnetic resonance imaging, X-ray imaging, ultrasound imaging, photoacoustic imaging, lab assays, or in any situation where
tagging/staining/imaging is required.
[00583] In some embodiments, compounds and/or compositions may be designed to include both linkers and payloads in any useful orientation. For example, linkers having two ends may be used to attach one end to the payload and the other end to compounds and/or compositions. Compounds and/or compositions of the present disclosure may include more than one payload. In some embodiments, compounds and/or compositions may comprise one or more cleavable linker. In some embodiments, payloads may be attached to compounds and/or compositions via a linker and may be fluorescently labeled for in vivo tracking, e.g. intracellularly.
[00584] In some embodiments, compounds and/or compositions of the present disclosure may be used in reversible drug delivery into cells.
[00585] Compounds and/or compositions of the present disclosure may be used in intracellular targeting of payloads, e.g., detectable or therapeutic agents, to specific organelles. In addition, compounds and/or compositions of the present disclosure may be used to deliver therapeutic agents to cells or tissues, e.g., in living animals. For example, the compounds and/or compositions described herein may be used to deliver chemotherapeutic agents to kill cancer cells. Compounds and/or compositions may be attached to therapeutic agents through one or more linkers may facilitate membrane permeation allowing therapeutic agents to travel into cells to reach intracellular targets.
[00586] In some embodiments, payloads may be a therapeutic agent such as a cytotoxins, radioactive ions, chemotherapeutics, or other therapeutic agents. Cytotoxins and/or cytotoxic agents may include any agents that may be detrimental to cells. Examples include, but are not limited to, taxol, cytochalasin B, gramicidin D, ethidium bromide, emetine, mitomycin, etoposide, teniposide, vincristine, vinblastine, colchicine, doxorubicin, daunorubicin, dihydroxyanthracinedione, mitoxantrone, mithramycin, actinomycin D, 1- dehydrotestosterone, glucocorticoids, procaine, tetracaine, lidocaine, propranolol, puromycin, maytansinoids, e.g. , maytansinol (see U.S. Pat. No. 5,208,020 incorporated herein in its entirety), rachelmycin (CC- 1065, see U.S. Pat. Nos. 5,475,092, 5,585,499, and 5,846,545, the contents of each of which are incorporated herein by reference in their entirety), and analogs
125 or homologs thereof. Radioactive ions include, but are not limited to iodine (e.g. , iodine or 131iodine), 89 strontium, phosphorous, palladium, cesium, iridium, phosphate, cobalt, 90yttrium,
153
samarium, and praseodymium. Other therapeutic agents include, but are not limited to, antimetabolites (e.g., methotrexate, 6-mercaptopurine, 6-thioguanine, cytarabine, 5- fluorouracil decarbazine), alkylating agents (e.g., mechlorethamine, thiotepa chlorambucil, rachelmycin (CC-1065), melphalan, carmustine (BSNU), lomustine (CCNU),
cyclophosphamide, busulfan, dibromomannitol, streptozotocin, mitomycin C, and cis- dichlorodiamine platinum (II) (DDP) cisplatin), anthracyclines (e.g., daunorubicin (formerly daunomycin) and doxorubicin), antibiotics (e.g., dactinomycin (formerly actinomycin), bleomycin, mithramycin, and anthramycin (AMC)), and anti-mitotic agents (e.g., vincristine, vinblastine, taxol and maytansinoids).
[00587] In some embodiments, payloads may be detectable agents, such as various organic small molecules, inorganic compounds, nanoparticles, enzymes or enzyme substrates, fluorescent materials, luminescent materials (e.g., luminol), bioluminescent materials (e.g., luciferase, luciferin, and aequorin), chemiluminescent materials, radioactive materials (e.g., 18F, 67Ga, 81mKr, 82Rb, mIn, 123I, 133Xe, 201T1, 1251, 35S, 14C, 3H, or 99mTc (e.g., as pertechnetate (technetate(VII), Tc04 ~)), and contrast agents (e.g., gold (e.g. , gold nanoparticles), gadolinium (e.g., chelated Gd), iron oxides (e.g., superparamagnetic iron oxide (SPIO), monocrystalline iron oxide nanoparticles (MIONs), and ultrasmall superparamagnetic iron oxide (USPIO)), manganese chelates (e.g., Mn-DPDP), barium sulfate, iodinated contrast media (iohexol), microbubbles, or perfluorocarbons). Such optically-detectable labels include for example, without limitation, 4-acetamido-4'-isothiocyanatostilbene-2,2'disulfonic acid; acridine and derivatives (e.g., acridine and acridine isothiocyanate); 5-(2'- aminoethyl)aminonaphthalene- l-sulfonic acid (EDANS); 4-amino-N-[3- vinylsulfonyl)phenyl]naphthalimide-3,5 disulfonate; N-(4-anilino-l-naphthyl)maleimide; anthranilamide; BODIPY; Brilliant Yellow; coumarin and derivatives (e.g., coumarin, 7- amino-4-methylcoumarin (AMC, Coumarin 120), and 7-amino-4-trifluoromethylcoumarin (Coumarin 151)); cyanine dyes; cyanosine; 4' ,6-diaminidino-2-phenylindole (DAPI); 5' 5"- dibromopyrogallol-sulfonaphthalein (Bromopyrogallol Red); 7-diethylamino-3-(4'- isothiocyanatophenyl)-4-methylcoumarin; diethylenetriamine pentaacetate; 4,4'- diisothiocyanatodihydro-stilbene-2,2'-disulfonic acid; 4,4' -diisothiocyanatostilbene-2,2'- disulfonic acid; 5-[dimethylamino]-naphthalene-l-sulfonyl chloride (DNS, dansylchloride);
4- dimethylaminophenylazophenyl-4'-isothiocyanate (DABITC); eosin and derivatives (e.g., eosin and eosin isothiocyanate); erythrosin and derivatives (e.g., erythrosin B and erythrosin isothiocyanate); ethidium; fluorescein and derivatives (e.g., 5-carboxyfluorescein (FAM), 5- (4,6-dichlorotriazin-2-yl)aminofluorescein (DTAF), 2',7'-dimethoxy-4'5'-dichloro-6- carboxyfluorescein, fluorescein, fluorescein isothiocyanate, X-rhodamine-5-(and-6)- isothiocyanate (QFITC or XRITC), and fluorescamine); 2-[2-[3-[[l,3-dihydro-l,l-dimethyl- 3 - (3 - sulf opropyl) -2H-benz [e] indol-2-ylidene] ethylidene] -2- [4- (ethoxycarbonyl)- 1 - piperazinyl]- 1-cyclopenten- l-yl]ethenyl]-l, l-dimethyl-3-(3-sulforpropyl)- 1H- benz[e]indolium hydroxide, inner salt, compound with n,n-diethylethanamine(l: l) (IR144);
5- chloro-2-[2-[3-[(5-chloro-3-ethyl-2(3H)-benzothiazol- ylidene)ethylidene]-2- (diphenylamino)-l-cyclopenten-l-yl]ethenyl]-3-ethyl benzothiazolium perchlorate (IR140); Malachite Green isothiocyanate; 4-methylumbelliferone orthocresolphthalein; nitro tyro sine; pararosaniline; Phenol Red; B-phycoerythrin; o-phthaldialdehyde; pyrene and
derivatives(e.g., pyrene, pyrene butyrate, and succinimidyl 1-pyrene); butyrate quantum dots; Reactive Red 4 (CIBACRON™ Brilliant Red 3B-A); rhodamine and derivatives (e.g., 6- carboxy-X-rhodamine (ROX), 6-carboxyrhodamine (R6G), lissamine rhodamine B sulfonyl chloride rhodamine (Rhod), rhodamine B, rhodamine 123, rhodamine X isothiocyanate, sulforhodamine B, sulforhodamine 101, sulfonyl chloride derivative of sulforhodamine 101 (Texas Red), N,N,N ',N'tetramethyl-6-carboxyrhodamine (TAMRA) tetramethyl rhodamine, and tetramethyl rhodamine isothiocyanate (TRITC)); riboflavin; rosolic acid; terbium chelate derivatives; Cyanine-3 (Cy3); Cyanine-5 (Cy5); cyanine-5.5 (Cy5.5), Cyanine-7 (Cy7); IRD 700; IRD 800; Alexa 647; La Jolta Blue; phthalo cyanine; and naphthalo cyanine.
[00588] In some embodiments, the detectable agent may be a non-detectable precursor that becomes detectable upon activation (e.g., fluorogenic tetrazine-fluorophore constructs (e.g., tetrazine-BODIPY FL, tetrazine-Oregon Green 488, or tetrazine-BODIPY TMR-X) or enzyme activatable fluorogenic agents (e.g., PROSENSE® (VisEn Medical))). In vitro assays in which the enzyme labeled compositions can be used include, but are not limited to, enzyme linked immunosorbent assays (ELISAs), immunoprecipitation assays,
immunofluorescence, enzyme immunoassays (EIA), radioimmunoassays (RIA), and Western blot analysis. Combinations
[00589] In some embodiments, compounds and/or compositions of the present disclosure may be used in combination with one or more other therapeutic, prophylactic, diagnostic, or imaging agents. By "in combination with," it is not intended to imply that the agents must be administered at the same time and/or formulated for delivery together, although these methods of delivery are within the scope of the present disclosure. Compounds and/or compositions of the present disclosure may be administered concurrently with, prior to, or subsequent to, one or more other desired therapeutics or medical procedures. In general, each agent will be administered at a dose and/or on a time schedule determined for that agent. In some embodiments, the present disclosure encompasses the delivery of pharmaceutical, prophylactic, diagnostic, or imaging compositions in combination with agents that may improve their bioavailability, reduce and/or modify their metabolism, inhibit their excretion, and/or modify their distribution within the body.
[00590] In some cases, compounds and/or compositions of the present disclosure may be combined with one or more therapeutic agents known in the art. Such agents may include BYM338 (Novartis, Basel, Switzerland), wherein administration may comprise any of the methods disclosed in clinical trial number NCT01925209 entitled Efficacy and Safety of Bimagrumab/BYM338 at 52 Weeks on Physical Function, Muscle Strength, Mobility in sIBM Patients (RESILIENT). Other agents that may be used in combination with compounds and/or compositions of the present disclosure may include any of those disclosed in US Pub. No. 2013/0122007, US Pat. No. 8,415,459 or International Pub. No. WO 2011/151432, the contents of each of which are herein incorporated by reference in their entirety.
Dosing and Dosage Forms
[00591] The present disclosure encompasses delivery of compounds and/or compositions of the present disclosure for any of therapeutic, pharmaceutical, diagnostic or imaging by any appropriate route taking into consideration likely advances in the sciences of drug delivery. Delivery may be naked or formulated.
Naked Delivery
[00592] Compounds and/or compositions of the present disclosure may be delivered to cells, tissues, organs and/or organisms in naked form. As used herein in, the term "naked" refers to compounds and/or compositions delivered free from agents or modifications which promote transfection or permeability. The naked compounds and/or compositions may be delivered to the cells, tissues, organs and/or organisms using routes of administration known in the art and described herein. In some embodiments, naked delivery may include formulation in a simple buffer such as saline or PBS.
Formulated Delivery
[00593] In some embodiments, compounds and/or compositions of the present disclosure may be formulated, using methods described herein. Formulations may comprise compounds and/or compositions which may be modified and/or unmodified. Formulations may further include, but are not limited to, cell penetration agents, pharmaceutically acceptable carriers, delivery agents, bioerodible or biocompatible polymers, solvents, and/or sustained-release delivery depots. Formulations of the present disclosure may be delivered to cells using routes of administration known in the art and described herein.
[00594] Compositions may also be formulated for direct delivery to organs or tissues in any of several ways in the art including, but not limited to, direct soaking or bathing, via a catheter, by gels, powder, ointments, creams, gels, lotions, and/or drops, by using substrates such as fabric or biodegradable materials coated or impregnated with compositions, and the like.
Dosing
[00595] The present disclosure provides methods comprising administering one or more compounds and/or compositions to subjects in need thereof. Compounds and/or compositions of the present disclosure, or prophylactic compositions thereof, may be administered to subjects using any amount and any route of administration effective for preventing, treating, diagnosing, or imaging diseases, disorders and/or conditions. The exact amount required will vary from subject to subject, depending on species, age and/or general subject condition, severity of disease, particular composition, mode of administration, mode of activity, and the like. Compositions in accordance with the disclosure are typically formulated in dosage unit form for ease of administration and uniformity of dosage. It will be understood, however, that the total daily usage of compositions of the present disclosure will be decided by the attending physician within the scope of sound medical judgment. The specific therapeutically effective, prophylactically effective, or appropriate imaging dose level for any particular patient will depend upon a variety of factors including the disorder being treated and the severity of the disorder; the activity of the specific compound employed; the specific composition employed; the age, body weight, general health, sex and diet of the patient; the time of administration, route of administration, and rate of excretion of the specific compound employed; the duration of the treatment; drugs used in combination or
coincidental with the specific compound employed; and like factors well known in the medical arts.
[00596] In certain embodiments, compositions in accordance with the present disclosure may be administered at dosage levels sufficient to deliver from about 0.0001 mg/kg to about 100 mg/kg, from about 0.01 mg/kg to about 50 mg/kg, from about 0.1 mg/kg to about 40 mg/kg, from about 0.5 mg/kg to about 30 mg/kg, from about 0.01 mg/kg to about 10 mg/kg, from about 0.1 mg/kg to about 10 mg/kg, or from about 1 mg/kg to about 25 mg/kg, of subject body weight per day, one or more times a day, to obtain the desired therapeutic, diagnostic, prophylactic, or imaging effect. The desired dosage may be delivered three times a day, two times a day, once a day, every other day, every third day, every week, every two weeks, every three weeks, or every four weeks. In certain embodiments, the desired dosage may be delivered using multiple administrations (e.g., two, three, four, five, six, seven, eight, nine, ten, eleven, twelve, thirteen, fourteen, or more administrations).
[00597] According to the present disclosure, compounds and/or compositions of the present disclosure may be administered in split-dose regimens. As used herein, a "split dose" is the division of single unit dose or total daily dose into two or more doses, e.g., two or more administrations of the single unit dose. As used herein, a "single unit dose" is a dose of any therapeutic administered in one dose/at one time/single route/single point of contact, i.e., single administration event. As used herein, a "total daily dose" is an amount given or prescribed in a 24 hour period. In some embodiments, compounds and/or compositions of the present disclosure may be administered as a single unit dose. In some embodiments, compounds and/or compositions of the present disclosure may be administered to subjects in split doses. In some embodiments, compounds and/or compositions of the present disclosure may be formulated in buffer only or in formulations described herein. Pharmaceutical compositions described herein may be formulated into dosage forms described herein, such as a topical, intranasal, intratracheal, or injectable (e.g., intravenous, intraocular, intravitreal, intramuscular, intracardiac, intraperitoneal, subcutaneous). General considerations in the formulation and/or manufacture of pharmaceutical agents may be found, for example, in Remington: The Science and Practice of Pharmacy 21st ed., Lippincott Williams & Wilkins, 2005 (incorporated herein by reference).
Coatings or Shells [00598] Solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings and other coatings well known in the pharmaceutical formulating art. They may optionally comprise opacifying agents and can be of a composition that they release the active ingredient(s) only, or preferentially, in a certain part of the intestinal tract, optionally, in a delayed manner. Examples of embedding compositions which can be used include polymeric substances and waxes. Solid compositions of a similar type may be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose and/or milk sugar as well as high molecular weight polyethylene glycols and the like.
Assays
[00599] In some embodiments, recombinant proteins (including, but not limited to chimeric proteins) disclosed herein and/or antibodies directed to such proteins may be developed using assays described herein. In some embodiments, recombinant proteins (including, but not limited to chimeric proteins) disclosed herein and/or antibodies directed to such proteins may be used in assays to develop other recombinant proteins and/or antibodies of the present disclosure.
Binding assays
[00600] In some embodiments, the present disclosure provides binding assays. As used herein, the term "binding assay" refers to an assay used to assess the ability of two or more factors to associate. Such assays may assess the ability of a desired antigen to bind a desired antibody and then use one or more detection methods to detect binding. Binding assays of the disclosure may include, but are not limited to surface Plasmon resonance-based assays, ELISAs and fluorescence flow cytometry-based assays. Binding assays of the disclosure may comprise the use of one or more recombinant proteins described herein, including, but not limited to any TGF-β family member proteins, any chimeric proteins, any cof actors and any modules, combinations or fragments thereof.
Cell-based assays
[00601] In some embodiments, the present disclosure provides cell-based assays. As used herein, the term "cell-based assay" refers to an assay comprising at least one aspect that involves the use of a living cell or cell culture. In some embodiments, these may be useful for assessing the modulation of growth factor release from GPCs, referred to herein as "growth factor release assays". In some embodiments, cell-based assays may be useful for assessing the modulation of growth factor activity, referred to herein as "growth factor activity assays". Cell-based assays of the present disclosure may comprise expression cells and/or responsive cells. Expression cells, as referred to herein, are cells that express one or more factors being analyzed in a particular assay. Such expression may be natural or may be the result of transfection and/or transduction of a foreign gene. In some embodiments, expression of one or more factors by expression cells may be enhanced or suppressed by the addition of one or more exogenous factors. In some embodiments, expression cells may comprise cell lines (e.g. HEK293 cells, HepG2 cells, CHO cells, TMLC cells, 293T/17 cells, Hs68 cells, CCD1112sk cells, HFF-1 cells, Keloid fibroblasts or Sw-480 cells). In some embodiments, cell lines comprising expression cells may express one or more recombinant proteins of the present disclosure (e.g. naturally and/or through transfection, stable transfection, and/or
transduction).
[00602] In some embodiments, growth factor release/activity assays may comprise expression cells that express GPCs. In such embodiments, additional factors may be co- expressed in and/or combined with expression cells to determine their effect on growth factor release from such GPCs. In some embodiments, integrins (including, but not limited to ανβό integrin, νβ8 integrin and/or agfii integrin) are co-expressed and/or otherwise introduced to GPC-expressing expression cells. In some embodiments, such additional integrin expression may facilitate growth factor release. In some embodiments, extracellular proteins (e.g.
LTBPs, fibrillins and/or GASPs) and/or variants thereof are coexpressed and/or otherwise introduced into expression cells.
[00603] In some embodiments, one or more genes may be knocked out, knocked down and/or otherwise modulated in expression cells depending on the focus of a particular assay. In some embodiments, one or more gene products may be modulated at the RNA and/or protein level. In some embodiments, gene products may be reduced through the introduction of siRNA molecules to expression cells. In some embodiments, gene products from extracellular protein (e.g. LTBPs, fibrillins and/or GASPs) genes may be reduced and/or eliminated from expression cells of the present disclosure.
[00604] Cell-based assays of the present disclosure, including, but not limited to growth factor release/activity assays, may comprise responsive cells. As used herein, the term
"responsive cell" refers to a cell that undergoes a response to one or more factors introduced into an assay. In some embodiments, such responses may include a change in gene expression, wherein such cells modulate transcription of one or more genes upon contact with one or more factors introduced. In some embodiments, responsive cells may undergo a change in phenotype, behavior and/or viability.
[00605] In some embodiments, responsive cells comprise one or more reporter genes. As used herein, the term "reporter gene" refers to a synthetic gene typically comprising a promoter and a protein coding region encoding one or more detectable gene products.
Reporter genes are typically designed in a way such that their expression may be modulated in response to one or more factors being analyzed by a particular assay. This may be carried out by manipulating the promoter of reporter genes. As used herein, the term promoter refers to part of a gene that initiates transcription of that gene. Promoters typically comprise nucleotides at the 3' end of the antisense strand of a given gene and are not transcribed during gene expression. Promoters typically function through interaction with one or more transcription factors as well as RNA polymerase enzymes to initiate transcription of the protein encoding portion of the gene. Segments of the promoter that physically interact with one or more transcription factors and/or polymerase enzymes are referred to herein as response elements. In some embodiments, reporter genes are designed to comprise promoters and/or response elements known to be responsive to one or more factors (including, but not limited to growth factors) being analyzed in a given assay. Changes in responsive cell gene expression may be measured according to any methods available in the art to yield gene expression data. Such gene expression data may be obtained in the form of luciferase activity data [often measured in terms of relative light units (RLUs)].
[00606] In some cases, responsive cells undergo a change in viability in response to one or more factors introduced in an assay. Such responsive cells may be used in proliferation assays as described herein. Changes in responsive cell viability may be detected by cell counting and/or other methods known to those skilled the art to yield responsive cell viability data.
[00607] Protein encoding regions of reporter genes typically encode one or more detectable proteins. Detectable proteins refer to any proteins capable of detection through one or more methods known in the art. Such detection methods may include, but are not limited to Western blotting, ELISA, assaying for enzymatic activity of detectable proteins (e.g. catalase activity, β-galactosidase activity and/or luciferase activity), immunocytochemical detection, surface plasmon resonance detection and/or detection of fluorescent detectable proteins. When a reporter gene is used in an assay, the expression of detectable proteins correlates with the ability of factors being assayed to activate the promoter present in the reporter gene. In embodiments comprising growth factor release/activity assays, reporter gene promoters typically respond to growth factor signaling. In such embodiments, the level of detectable protein produced correlates with level of growth factor signaling, indicating release and/or activity of a given growth factor.
[00608] In some embodiments, reporter genes encode luciferase enzymes. Chemical reactions between luciferase enzymes and substrate molecules are light-emitting reactions. Due to such light-emitting reactions, luciferase enzyme levels can be quantified through the addition of substrate molecules and subsequent photodetection of the emitted light. In some embodiments, reporter genes of the present disclosure encode firefly luciferase, the sequence of which was cloned from Photinus pyralis. In some embodiments, responsive cells of the present disclosure comprise reporter genes that express luciferase with promoters that are responsive to growth factors. In such embodiments, luciferase activity may correlate with growth factor activity levels allowing for growth factor activity and/or release from GPCs to be determined.
[00609] In some embodiments, reporter genes are inserted into bacterial plasmids to enable replication and/or facilitate introduction into cells. In some embodiments, such plasmids are designed to comprise sequences encoding detectable gene products and may be manipulated to insert promoter sequences that may be responsive to one or more factors of interest. These plasmids are referred to herein as reporter plasmids. In some embodiments of the present disclosure, promoters that may be responsive to one or more factors of interest may be inserted into reporter plasmids, upstream of sequences encoding detectable gene products to form functional reporter genes within such reporter plasmids. Reporter plasmids that comprise at least one functional reporter gene are referred to herein as reporter constructs. In some embodiments, reporter constructs of the present disclosure may comprise pGL2 reporter plasmids (Promega Biosciences, LLC, Madison, WI), pGL3 reporter plasmids (Promega Biosciences, LLC, Madison, WI), pGL4 reporter plasmids (Promega Biosciences, LLC, Madison, WI) or variants thereof. Such reporter constructs express firefly luciferase in response to promoter activation.
[00610] In some embodiments, reporter constructs may be introduced directly into expression cells or may be introduced into one or more responsive cells. Responsive cells of the present disclosure comprising one or more reporter genes are referred to herein as reporter cells. In some embodiments, reporter cells may be transiently transfected with reporter constructs or may comprise stable expression of such constructs (e.g. reporter constructs are successfully replicated along with genomic DNA during each round of cell division). Cell lines that stably comprise reporter constructs are referred to herein as reporter cell lines. In some embodiments, reporter cells are mammalian. In some embodiments, reporter cells may comprise mouse cells, rabbit cells, rat cells, monkey cells, hamster cells and human cells. In some embodiments, cell lines useful for transient and/or stable expression of reporter genes may include, but are not limited to HEK293 cells, HepG2 cells, HeLa cells, Sw-480 cells, TMLC cells [as disclosed by Abe et al (Abe, M. et al., An assay for transforming growth factor- β using cells transfected with a plasminogen activator inhibitor- 1 promo ter-lucif erase construct. Analytical Biochemistry. 1994. 216:276-84)], 293T/17 cells, Hs68 cells,
CCD1112sk cells, HFF-1 cells, Keloid fibroblasts, A204 cells, L17 RIB cells [as disclosed by Cash et al (Cash, J.N et al., The structure of myostatin:follistatin 288: insights into receptor utilization and heparin binding. The EMBO Journal. 2009. 28:2662-76)], C2C12 cells, HepG2 cells and EL4 T lymphoma cells.
[00611] In embodiments where one or more reporter cells and/or reporter cell lines are utilized, such cells may be cultured with expression cells as part of a co-culture system. In some embodiments reporter cells/reporter cell lines may be cultured separately from expression cells. In such embodiments, lysates and/or media from expression cells may be combined with reporter cell/reporter cell line cultures to assess expressed factors (including, but not limited to growth factors).
[00612] In some embodiments, cell-based assays of the present disclosure may only comprise expression cells and not responsive cells. In such embodiments, expressed proteins, including but not limited to GPCs and/or growth factors, may be detected by one or more methods that are not cell based. Such methods may include, but are not limited to Western Blotting, enzyme-linked immunosorbent assay (ELISA), immunocytochemistry, surface plasmon resonance and other methods known in the art for protein detection. In some embodiments, GDF release in expression cell cultures and/or culture medium may be detected by ELISA. In some embodiments, the GDF- 8/myo statin quantikine ELISA kit (R&D Systems, Minneapolis, MN) may be used. Examples of anti-GDF- 8/myo statin antibodies that may be used for detection include AF1539, MAB788 and AF788 (R&D Systems, Minneapolis, MN).
[00613] In some embodiments, reporter genes of the present disclosure comprise growth factor-responsive promoters. As used herein, the term "growth factor-responsive promoter" refers to a gene promoter that facilitates transcription of a downstream gene in response to growth factor cell signaling induced by one or more growth factors. In some embodiments, growth factor-responsive promoters are responsive to TGF-β family member growth factor signaling. In some embodiments, growth factor-responsive promoters of the present disclosure comprise one or more sequences listed in Table 18 or fragments or variants thereof. These include two versions of the plasminogen activator inhibitor type 1 (PAI-1) promoter [VI as disclosed by Abe et al (Abe, M. et al., An assay for transforming growth factor- β using cells transfected with a plasminogen activator inhibitor- 1 promo ter-lucif erase construct. Analytical Biochemistry. 1994. 216:276-84) and V2 as disclosed in WO
2011/034935, the contents of which are hereby incorporated by reference in their entirety,] a collagen, type 1, alpha 1 promoter, a collagen, type 1, alpha 2 promoter, a FoxP3 promoter, a CAGA12 promoter [responsive to Smad-dependent signaling as reported by Thies et al (Thies, R.S. et al., GDF-8 propeptide binds to GDF-8 and antagonizes biological activity by inhibiting GDF-8 receptor binding. Growth Factors. 2001. 18:251-9) and an adenovirus major late promoter.
Table 18. Growth factor-responsive promoters
Promoter Sequence SEQ
ID NO
PAI-1 (VI) AGCTTACCATGGTAACCCCTGGTCCCGTTCAGCCACCACCACCC 1628
CACCCAGCACACCTCCAACCTCAGCCAGACAAGGTTGTTGACA
CAAGAGAGCCCTCAGGGGCACAGAGAGAGTCTGGACACGTGG
GGAGTCAGCCGTGTATCATCGGAGGCGGCCGGGCACATGGCAG
GGATGAGGGAAAGACCAAGAGTCCTCTGTTGGGCCCAAGTCCT
AGACAGACAAAACCTAGACAATCACGTGGCTGGCTGCATGCCT
GTGGCTGTTGGGCTGGGCAGGAGGAGGGAGGGGCGCTCTTTCC
TGGAGGTGGTCCAGAGCACCGGGTGGACAGCCCTGGGGGAAA
ACTTCCACGTTTTGATGGAGGTTATCTTTGATAACTCCACAGTG
ACCTGGTTCGCCAAAGGAAAAGCAGGCAACGTGAGCTGTTTTT
ACCCGGCATGGCAGACAGTCAACCTGGCAGGACATCCGGGAG
AGACAGACACAGGCAGAGGGCAGAAAGGTCAAGGGAGGTTCT
CAGGCCAAGGCTATTGGGGTTTGCTCAATTGTTCCTGAATGCTC
TTACACACGTACACACACAGAGCAGCACACACACACACACACA
CATGCCTCAGCAAGTCCCAGAGAGGGAGGTGTCGAGGGGGAC
CCGCTGGCTGTTCAGACGGACTCCCAGAGCCAGTGAGTGGGTG
GGGCTGGAACATGAGTTCATCTATTTCCTGCCCACATCTGGTAT
AAAAGGAGGCAGTGGCCCACAGAGGAGCACAGCTGTGTTTGG
CTGCAGGGCCAAGAGCGCTGTCAAGAAGACCCACACGCCCCCC
TCCAGCAGCTG
PAI-1 (V2) TTGGTCTCCTGTTTCCTTACCAAGCTTTTACCATGGTAACCCCTG 1629
GTCCCGTTCAGCCACCACCACCCCACCCAGCACACCTCCAACCT
CAGCCAGACAAGGTTGTTGACACAAGAGAGCCCTCAGGGGCAC
AGAGAGAGTCTGGACACGTGGGGAGTCAGCCGTGTATCATCGG
AGGCGGCCGGGCACATGGCAGGGATGAGGGAAAGACCAAGAG
TCCTCTGTTGGGCCCAAGTCCTAGACAGACAAAACCTAGACAA
TCACGTGGCTGGCTGCATGCCCTGTGGCTGTTGGGCTGGGCCCA
GGAGGAGGGAGGGGCGCTCTTTCCTGGAGGTGGTCCAGAGCAC
GTTATCTTTGATAACTCCACAGTGACCTGGTTCGCCAAAGGAA AAGCAGGCAACGTGAGCTGTTTTTTTTTTCTCCAAGCTGAACAC
CAACCTGGCAGGACATCCGGGAGAGACAGACACAGGCAGAGG
GCAGAAAGGTCAAGGGAGGTTCTCAGGCCAAGGCTATTGGGGT
TTGCTCAATTGTTCCTGAATGCTCTTACACACGTACACACACAG
AGCAGCACACACACACACACACACATGCCTCAGCAAGTCCCAG
AGAGGGAGGTGTCGAGGGGGACCCGCTGGCTGTTCAGACGGA
CTCCCAGAGCCAGTGAGTGGGTGGGGCTGGAACATGAGTTCAT
CTATTTCCTGCCCACATCTGGTATAAAAGGAGGCAGTGGCCCA
CAGAGGAGCACAGCTGTGTTTGGCTGCAGGGCCAAGAGCGCTG
TCAAGAAGACCCACACGCCCCCCTCCAGCAGCTGAATTCCTGC
AGCTCAGCAGCCGCCGCCAGAGCAGGACGAACCGCCAATCGC
AAGGCACCTCTGAGAACTTCAGGTA
CollAl CCATGGCAAACAAAACTCTTCTCTAAGTCACCAATGATCACAG 1630
GCCTCCCACTAAAAATACTTCCCAACTCTGGGGTGGAAGAGTT
TCTGTGTCCCTAGAATCCCCCACCCCTACCTTGGCTGCTCCATC
ACCCAACCACCAAAGCTTTCTTCTGCAGAGGCCACCTAGTCAT
GTTTCTCACCCTGCACCTCAGCCTCCCCACTCCATCTCTCAATC
ATGCCTAGGGTTTGGAGGAAGGCATTTGATTCTGTTCTGGAGCA
CAGCAGAAGAATTGACATCCTCAAAATTAAAACTCCCTTGCCT
GCACCCCTCCCTCAGATATCTGATTCTTAATGTCTAGAAAGGAA
TCTGTAAATTGTTCCCCAAATATTCCTAAGCTCCATCCCCTAGC
CACACCAGAAGACACCCCCAAACAGGCACATCTTTTTAATTCC
CAGCTTCCTCTGTTTTGGAGAGGTCCTCAGCATGCCTCTTTATG
CCCCTCCCTTAGCTCTTGCCAGGATATCAGAGGGTGACTGGGG
CACAGCCAGGAGGACCCCCTCCCCAACACCCCCAACCCTTCCA
CCTTTGGAAGTCTCCCCACCCAGCTCCCCAGTTCCCCAGTTCCA
CTTCTTCTAGATTGGAGGTCCCAGGAAGAGAGCAGAGGGGCAC
CCCTACCCACTGGTTAGCCCACGCCATTCTGAGGACCCAGCTGC
ACCCCTACCACAGCACCTCTGGCCCAGGCTGGGCTGGGGGGCT
GGGGAGGCAGAGCTGCGAAGAGGGGAGATGTGGGGTGGACTC
CCTTCCCTCCTCCTCCCCCTCTCCATTCCAACTCCCAAATTGGG
GGCCGGGCCAGGCAGCTCTGATTGGCTGGGGCACGGGCGGCCG
GCTCCCCCTCTCCGAGGGGCAGGGTTCCTCCCTGCTCTCCATCA
GGACAGTATAAAAGGGGCCCGGGCCAGTCGTCGGAGCAGACG
GGAGTTTCTCCTCGGGGTCGGAGCAGGAGGCACGCGGAGTGTG
AGGCCACGCATGAGCGGACGCTAACCCCCTCCCCAGCCACAAA
GAGTCTACATG
CollA2 TAGAGTTCGCAAAGCCTATCCTCCCTGTAGCCGGGTGCCAAGC 1631
AGCCTCGAGCCTGCTCCCCAGCCCACCTGCCAACAAAAGGCGC
CCTCCGACTGCAACCCAGCCCTCCACAGACAGGACCCGCCCTT
TCCCGAAGTCATAAGACAAAGAGAGTGCATCACTGCTGAAACA
GTGGGCGCACACGAGCCCCAAAGCTAGAGAAAAGCTGGACGG
GGCTGGGGGCGGGGTGCAGGGGTGGAGGGGCGGGGAGGCGGG
CTCCGGCTGCGCCACGCTATCGAGTCTTCCCTCCCTCCTTCTCT
GCCCCCTCCGCTCCCGCTGGAGCCCTCCACCCTACAAGTGGCCT
ACAGGGCACAGGTGAGGCGGGACTGGACAGCTCCTGCTTTGAT
CGCCGGAGATCTGCAAATTCTGCCCATGTCGGGGCTGCAGAGC
ACTCCGACGTGTCCCATAGTGTTTCCAAACTTGGAAAGGGCGG
GGGAGGGCGGGAGGATGCGGAGGGCGGAGGTATGCAGACAAC
GAGTCAGAGTTTCCCCTTGAAAGCCTCAAAAGTGTCCACGTCCT
CAAAAAGAATGGAACCAATTTAAGAAGCCAGCCCCGTGGCCAC
GTCCCTTCCCCCATTCGCTCCCTCCTCTGCGCCCCCGCAGGCTC
CTCCCAGCTGTGGCTGCCCGGGCCCCCAGCCCCAGCCCTCCCAT TGGTGGAGGCCCTTTTGGAGGCACCCTAGGGCCAGGGAAACTT
TTGCCGTATAAATAGGGCAGATCCGGGCTTTATTATTTTAGCAC
CACGGCAGCAGGAGGTTTCGGCTAAGTTGGAGGTACTGGCCAC
GACTGCATGCCCGCGCCCGCCAGGTGATACCTCCGCCGGTGAC
CCAGGGGCTCTGCGACACAAGGAGTCTGCATGTCTAAGTGCTA
GACATGCTCAGCTTTGTGGATACGCGGACTTTGTTGCTGCTTGC
AGTAA
FoxP3 AGTAAAAGACCCCAAAGGCTGAGGGCCTCAGAAGCATCAGGC 1632
CATGATGTTCCTGAAACAAGAGGGTCAGGGTCCCAATGGGCCT
CTGGGGTTCATCGTGAGGATGGATGCATTAATATTGGGGACCT
GCTAGGGACCTTCCCAGTGGGACAGTGGCTGGGTCAGGGCACT
CAAGCCCTAAAACGTGATGAGGCGAGACTTTTCTCTCTTTCCTC
ATTCAGTAACTGTCAGTAGATTCTGGGAGCCAGGGATTCTCCG
ACTCTTCAAGTCCATGAATTTTAGGGGATGACAGTGGGCTCTCC
GCTTTCTCCTCCATGAAGTAACTTACATGCCCCTCACCCTCTGT
GGGAGGGGTGTTGCAGGGGGTGCAGAACTCCCCTCGCCGGGTA
GTTCAAGCAATGGGGACCATATCAATTCCATCTATAGGGAAAC
TGAGGCCTGGAGTAGGGCGAGGCCTCTGGGAACCCAGCCCTAT
TCTGTCTCTTTCCCTGGCATTTCCCATCCACACATAGAGCTTCA
GATTCTCTTTCTTTCCCCAGAGACCCTCAAATATCCTCTCACTC
ACAGAATGGTGTCTCTGCCTGCCTCGGGTTGGCCCTGTGATTTA
TTTTAGTTCTTTTCCCTTGTTTTTTTTTTTTCAAACTCTATACACT
TTTGTTTTAAAAACTGTGGTTTCTCATGAGCCCTATTATCTCATT
GATACCTCTCACCTCTGTGGTGAGGGGAAGAAATCATATTTTCA
GATGACTCGTAAAGGGCAAAGAAAAAAACCCAAAATTTCAAA
ATTTCCGTTTAAGTCTCATAATCAAGAAAAGGAGAAACACAGA
GAGAGAGAAAAAAAAAACTATGAGAACCCCCCCCCACCCCGT
GATTATCAGCGCACACACTCATCGAAAAAAATTTGGATTATTA
GAAGAGAGAGGTCTGCGGCTTCCACACCGTACAGCGTGGTTTT
TCCCACAAGCCAGGCTGATCCTTTTCTGTCAGTCCACTTCACCA
CAGA12 AGCCAGACAAGCCAGACAAGCCAGACAAGCCAGACAAGCCAG 1633
ACAAGCCAGACAAGCCAGACAAGCCAGACAAGCCAGACAAGC CAGACAAGCCAGACAAGCCAGACA
Adenovirus GGGCTATAAAAGGGGGTGGGGGCGCGTTCGTCCTCACTCTCTT 1634 major late CCG
promoter
[00614] In some embodiments, mink lung epithelial/PAI reporter cell lines may be used. Mink lung epithelial cells do not produce TGF-β, but do express high levels of TGF-β receptors (Munger et al). Mink lung epithelial/PAI reporter cell lines comprise reporter constructs comprising promoter elements from the TGF-P-responsive genes PAI and/or COLIA that modulate the expression of the protein coding portion of the luciferase gene. In some embodiments, other reporter constructs may be used with mink lung epithelial cells. In some embodiments, SMAD3-responsive reporter constructs may be used.
[00615] CAGA promoter-based reporter assays may be used to test antibodies that modulate SMAD-dependent gene expression as reported by Thies et al (Thies, R.S. et al., Growth Factors. 2001. 18:251-9, the contents of which are herein incorporated by reference in their entirety). In some cases, CAGA reporter assays may be used to assess GDF-8 and/or GDF-11 growth factor activity in the presence and absence of an activating or inhibiting antibody. According to such methods, GDF-8 growth factor (or proGDF-8 subjected to furin and/or mTLL2 processing) may be cultured with reporter cells comprising the CAGA promoter linked to a reporter gene (e.g. luciferase) in the presence or absence of the antibody being tested.
Proliferation/differentiation assays
[00616] In some embodiments, cell-based assays of the present disclosure may comprise proliferation assays. As used herein, the term "proliferation assay" refers to an assay that determines the effect on one or more agents on cell proliferation.
[00617] In some embodiments, cell differentiation assays may be used to assess growth factor activity modulation by activating and/or inhibiting antibodies. Cell differentiation assays may include skeletal muscle differentiation assays. Such assays may be used to test GDF-8 activating and/or inhibiting antibodies. In some cases, skeletal muscle differentiation assays assess myoblast differentiation by looking at changes in the expression level of proteins that change during stages of differentiation. Such proteins may include, but are not limited to myogenin, myosin heavy chain and creatine kinase. GDF-8 inhibits myoblast differentiation, therefore GDF-8-inhibiting antibodies may be tested by examining their effect on GDF-8 -dependent inhibition of myoblast differentiation.
Animal Models
[00618] In some embodiments, compounds and/or compositions of the present disclosure may be tested in animal models including mammalian models of muscles growth and development. These models may include but are not limited to mouse models of grip strength. Such models may include testing by determining the force (in grams) generated when a mouse is pulled from a force measuring lever. Additional models may include but are not limited to other strength and endurance tests such as the Morris water maze test for endurance and the wire hang test for motor deficits. Other mouse models may include, but are not limited to assays which determine changes in whole muscle weight and size.
[00619] In another embodiment, compounds and/or compositions of the present disclosure may be tested in non-mouse animal models including models of muscle growth and development where changes in muscle are observed. Such changes may include, but are not limited to changes in muscle weight and size, muscle fiber length and diameter, number of fibers per muscle bundle, number of synaptic contacts per muscle fiber, and fiber strength.
[00620] In some embodiments, tissues from animals treated with compounds and/or compositions of the present disclosure may be analyzed by immunohistochemical analysis. Such analysis may include immuno staining of muscles, muscle fibers and muscle connections to show changes in response to treatment. Complete blood counts may be carried out pre and post study to enable monitoring of compounds and/or compositions of the present disclosure.
Kits and Devices
[00621] Any of the compounds and/or compositions of the present disclosure may be comprised in a kit. In a non-limiting example, reagents for generating compounds and/or compositions, including antigen molecules are included in one or more kit. In some embodiments, kits may further include reagents and/or instructions for creating and/or synthesizing compounds and/or compositions of the present disclosure. In some
embodiments, kits may also include one or more buffers. In some embodiments, kits of the disclosure may include components for making protein or nucleic acid arrays or libraries and thus, may include, for example, solid supports.
[00622] In some embodiments, kit components may be packaged either in aqueous media or in lyophilized form. The container means of the kits will generally include at least one vial, test tube, flask, bottle, syringe or other container means, into which a component may be placed, and preferably, suitably aliquotted. Where there are more than one kit component, (labeling reagent and label may be packaged together), kits may also generally contain second, third or other additional containers into which additional components may be separately placed. In some embodiments, kits may also comprise second container means for containing sterile, pharmaceutically acceptable buffers and/or other diluents. In some embodiments, various combinations of components may be comprised in one or more vial. Kits of the present disclosure may also typically include means for containing compounds and/or compositions of the present disclosure, e.g., proteins, nucleic acids, and any other reagent containers in close confinement for commercial sale. Such containers may include injection or blow-molded plastic containers into which desired vials are retained.
[00623] In some embodiments, kit components are provided in one and/or more liquid solutions. In some embodiments, liquid solutions are aqueous solutions, with sterile aqueous solutions being particularly preferred. In some embodiments, kit components may be provided as dried powder(s). When reagents and/or components are provided as dry powders, such powders may be reconstituted by the addition of suitable volumes of solvent. In some embodiments, it is envisioned that solvents may also be provided in another container means. In some embodiments, labeling dyes are provided as dried powders. In some embodiments, it is contemplated that 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 120, 120, 130, 140, 150, 160, 170, 180, 190, 200, 300, 400, 500, 600, 700, 800, 900, 1000 micrograms or at least or at most those amounts of dried dye are provided in kits of the disclosure. In such embodiments, dye may then be resuspended in any suitable solvent, such as DMSO.
[00624] In some embodiments, kits may include instructions for employing kit components as well the use of any other reagent not included in the kit. Instructions may include variations that may be implemented.
[00625] In some embodiments, compounds and/or compositions of the present disclosure may be combined with, coated onto or embedded in a device. Devices may include, but are not limited to, dental implants, stents, bone replacements, artificial joints, valves, pacemakers and/or other implantable therapeutic device.
Definitions
[00626] At various places in the present specification, substituents of compounds of the present disclosure are disclosed in groups or in ranges. It is specifically intended that the present disclosure include each and every individual subcombination of the members of such groups and ranges. The following is a non-limiting list of term definitions.
[00627] Activity: As used herein, the term "activity" refers to the condition in which things are happening or being done. Compositions of the disclosure may have activity and this activity may involve one or more biological events. In some embodiments, such biological event may involve growth factors and/or growth factor signaling. In some embodiments, biological events may include cell signaling events associated with growth factor and receptor interactions. In some embodiments, biological events may include cell signaling events associated with TGF-β or TGF-P-related protein interactions with one or more corresponding receptors.
[00628] Administered in combination: As used herein, the term "administered in
combination" or "combined administration" refers to simultaneous exposure of one or more subjects to two or more agents administered at the same time or within an interval such that the subject is at some point in time simultaneously exposed to both and/or such that there may be an overlap in the effect of each agent on the patient. In some embodiments, at least one dose of one or more agents is administered within about 24 hours, 12 hours, 6 hours, 3 hours, 1 hour, 30 minutes, 15 minutes, 10 minutes, 5 minutes, or 1 minute of at least one dose of one or more other agents. In some embodiments, administration occurs in overlapping dosage regimens. As used herein, the term "dosage regimen" refers to a plurality of doses spaced apart in time. Such doses may occur at regular intervals or may include one or more hiatus in administration. In some embodiments, the administration of individual doses of one or more compounds and/or compositions of the present disclosure, as described herein, are spaced sufficiently closely together such that a combinatorial (e.g., a synergistic) effect is achieved.
[00629] Animal: As used herein, the term "animal" refers to any member of the animal kingdom. In some embodiments, "animal" refers to humans at any stage of development. In some embodiments, "animal" refers to non-human animals at any stage of development. In certain embodiments, the non-human animal is a mammal (e.g., a rodent, a mouse, a rat, a rabbit, a monkey, a dog, a cat, a sheep, cattle, a primate, or a pig). In some embodiments, animals include, but are not limited to, mammals, birds, reptiles, amphibians, fish, and worms. In some embodiments, the animal is a transgenic animal, genetically-engineered animal, or a clone.
[00630] Antigens of interest or desired antigens: As used herein, the terms "antigens of interest" or "desired antigens" refers to those proteins and/or other biomolecules provided herein that are immuno specifically bound or interact with antibodies of the present disclosure and/or fragments, mutants, variants, and/or alterations thereof described herein. In some embodiments, antigens of interest may comprise TGF-P-related proteins, growth factors, prodomains, GPCs, protein modules or regions of overlap between them.
[00631] Approximately: As used herein, the term "approximately" or "about," as applied to one or more values of interest, refers to a value that is similar to a stated reference value. In certain embodiments, the term "approximately" or "about" refers to a range of values that fall within 25%, 20%, 19%, 18%, 17%, 16%, 15%, 14%, 13%, 12%, 11%, 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, 1%, or less in either direction (greater than or less than) of the stated reference value unless otherwise stated or otherwise evident from the context (except where such number would exceed 100% of a possible value).
[00632] Associated with: As used herein, the terms "associated with," "conjugated," "linked," "attached," and "tethered," when used with respect to two or more moieties, mean that the moieties are physically associated or connected with one another, either directly or via one or more additional moieties that serve as linking agents, to form a structure that is sufficiently stable so that the moieties remain physically associated under the conditions in which the structure is used, e.g., physiological conditions. An "association" need not be strictly through direct covalent chemical bonding. It may also suggest ionic or hydrogen bonding or a hybridization based connectivity sufficiently stable such that the "associated" entities remain physically associated.
[00633] Biomolecule: As used herein, the term "biomolecule" is any natural molecule which is amino acid-based, nucleic acid-based, carbohydrate-based or lipid-based, and the like.
[00634] Biologically active: As used herein, the phrase "biologically active" refers to a characteristic of any substance that has activity in a biological system and/or organism. For instance, a substance that, when administered to an organism, has a biological effect on that organism, is considered to be biologically active. In particular embodiments, a compounds and/or compositions of the present disclosure may be considered biologically active if even a portion of is biologically active or mimics an activity considered to biologically relevant.
[00635] Biological system: As used herein, the term "biological system" refers to a group of organs, tissues, cells, intracellular components, proteins, nucleic acids, molecules (including, but not limited to biomolecules) that function together to perform a certain biological task within cellular membranes, cellular compartments, cells, tissues, organs, organ systems, multicellular organisms, or any biological entity. Biological systems may be in vitro or in vivo. In some embodiments, biological systems are cell signaling pathways comprising intracellular and/or extracellular cell signaling biomolecules. In some embodiments, biological systems comprise growth factor signaling events within the extracellular matrix, cellular matrix and/or cellular niches.
[00636] Candidate antibody: As used herein, the term "candidate antibody" refers to an antibody from a pool of one or more antibody from which one or more desired antibodies may be selected.
[00637] Cellular matrix: As used herein, the term "cellular matrix" refers to the biochemical and structural environment associated with the outer portion of the cell membrane. Such cell membranes may also include platelet membranes. Components of the cellular matrix may include, but are not limited to proteoglycans, carbohydrate molecules, integral membrane proteins, glycolipids and the like. In some cases, cellular matrix components may include growth factors and/or modulators of growth factor activity. Some cellular matrix proteins include integrins, GARP and LRRC33.
[00638] Compound: As used herein, the term "compound," refers to a distinct chemical entity The term may be used herein to refer to peptides, proteins, protein complexes or antibodies of the disclosure. In some embodiments, a particular compound may exist in one or more isomeric or isotopic forms (including, but not limited to stereoisomers, geometric isomers and isotopes). In some embodiments, a compound is provided or utilized in only a single such form. In some embodiments, a compound is provided or utilized as a mixture of two or more such forms (including, but not limited to a racemic mixture of stereoisomers). Those of skill in the art appreciate that some compounds exist in different such forms, show different properties and/or activities (including, but not limited to biological activities). In such cases it is within the ordinary skill of those in the art to select or avoid particular forms of the compound for use in accordance with the present disclosure. For example, compounds that contain asymmetrically substituted carbon atoms can be isolated in optically active or racemic forms. Methods on how to prepare optically active forms from optically active starting materials are known in the art, such as by resolution of racemic mixtures or by stereoselective synthesis.
[00639] Conserved: As used herein, the term "conserved" refers to nucleotides or amino acid residues of polynucleotide or polypeptide sequences, respectively, that are those that occur unaltered in the same position of two or more sequences being compared. Nucleotides or amino acids that are relatively conserved are those that are conserved among more related sequences than nucleotides or amino acids appearing elsewhere in the sequences.
[00640] In some embodiments, two or more sequences are said to be "completely conserved" if they are 100% identical to one another. In some embodiments, two or more sequences are said to be "highly conserved" if they are at least 70% identical, at least 80% identical, at least 90% identical, or at least 95% identical to one another. In some
embodiments, two or more sequences are said to be "highly conserved" if they are about 70% identical, about 80% identical, about 90% identical, about 95%, about 98%, or about 99% identical to one another. In some embodiments, two or more sequences are said to be
"conserved" if they are at least 30% identical, at least 40% identical, at least 50% identical, at least 60% identical, at least 70% identical, at least 80% identical, at least 90% identical, or at least 95% identical to one another. In some embodiments, two or more sequences are said to be "conserved" if they are about 30% identical, about 40% identical, about 50% identical, about 60% identical, about 70% identical, about 80% identical, about 90% identical, about 95% identical, about 98% identical, or about 99% identical to one another. Conservation of sequence may apply to the entire length of an oligonucleotide or polypeptide or may apply to a portion, region or feature thereof. [00641] In one embodiment, conserved sequences are not contiguous. Those skilled in the art are able to appreciate how to achieve alignment when gaps in contiguous alignment are present between sequences, and to align corresponding residues not withstanding insertions or deletions present.
[00642] Delivery: As used herein, "delivery" refers to the act or manner of delivering a compound, substance, entity, moiety, cargo or payload.
[00643] Delivery Agent: As used herein, "delivery agent" refers to any agent which facilitates, at least in part, the in vivo delivery of one or more substances (including, but not limited to a compounds and/or compositions of the present disclosure) to a cell, subject or other biological system cells.
[00644] Desired antibody: As used herein, the term "desired antibody" refers to an antibody that is sought after, in some cases from a pool of candidate antibodies.
[00645] Destabilized: As used herein, the term "destable," "destabilize," or "destabilizing region" means a region or molecule that is less stable than a starting, reference, wild-type or native form of the same region or molecule.
[00646] Detectable label: As used herein, "detectable label" refers to one or more markers, signals, or moieties which are attached, incorporated or associated with another entity, which markers, signals or moieties are readily detected by methods known in the art including radiography, fluorescence, chemiluminescence, enzymatic activity, absorbance,
immunological detection and the like. Detectable labels may include radioisotopes, fluorophores, chromophores, enzymes, dyes, metal ions, ligands, biotin, avidin, streptavidin and haptens, quantum dots, polyhistidine tags, myc tags, flag tags, human influenza hemagglutinin (HA) tags and the like. Detectable labels may be located at any position in the entity with which they are attached, incorporated or associated. For example, when attached, incorporated in or associated with a peptide or protein, they may be within the amino acids, the peptides, or proteins, or located at the N- or C- termini.
[00647] Distal: As used herein, the term "distal" means situated away from the center or away from a point or region of interest.
[00648] Engineered: As used herein, embodiments of the disclosure are "engineered" when they are designed to have a feature or property, whether structural or chemical, that varies from a starting point, wild type or native molecule. Thus, engineered agents or entities are those whose design and/or production include an act of the hand of man.
[00649] Epitope: As used herein, an "epitope" refers to a surface or region on a molecule that is capable of interacting with components of the immune system, including, but not limited to antibodies. In some embodiments, when referring to a protein or protein module, an epitope may comprise a linear stretch of amino acids or a three dimensional structure formed by folded amino acid chains.
[00650] Expression: As used herein, "expression" of a nucleic acid sequence refers to one or more of the following events: (1) production of an RNA template from a DNA sequence {e.g., by transcription); (2) processing of an RNA transcript {e.g., by splicing, editing, 5' cap formation, and/or 3' end processing); (3) translation of an RNA into a polypeptide or protein; (4) folding of a polypeptide or protein; and (5) post-translational modification of a
polypeptide or protein.
[00651] Extracellular matrix: As used herein, the term, "extracellular matrix," or "ECM" refers to the area surrounding cells and/or the area between cells that typically comprises structural proteins as well as cell signaling molecules. Components of the extracellular matrix may include, but are not limited to proteins, nucleic acids, membranes, lipids and sugars that may be directly or indirectly associated with structural components of the extracellular environments. Structural components of the extracellular matrix may include, but are not limited to proteins, polysaccharides (e.g. hyaluronic acid), glycosaminoglycans and proteoglycans (e.g. heparin sulfate, chondroitin sulfate and keratin sulfate). Such structural components may include, but are not limited to fibrous components (e.g. collagens and elastins), fibrillins, fibronectin, laminins, agrin, perlecan, decorin and the like. Other proteins that may be components of the extracellular matrix include GASPs and LTBPs. Extracellular matrix components may also include growth factors and/or modulators of growth factor activity.
[00652] Feature: As used herein, a "feature" refers to a characteristic, a property, or a distinctive element.
[00653] Formulation: As used herein, a "formulation" includes at least a compound and/or composition of the present disclosure and a delivery agent.
[00654] Fragment: A "fragment," as used herein, refers to a portion. For example, fragments of proteins may comprise polypeptides obtained by digesting full-length protein isolated from cultured cells. In some embodiments, a fragment of a protein includes at least 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 150, 200, 250 or more amino acids. In some embodiments, fragments of an antibody include portions of an antibody subjected to enzymatic digestion or synthesized as such. [00655] Functional: As used herein, a "functional" biological molecule is a biological entity with a structure and in a form in which it exhibits a property and/or activity by which it is characterized.
[00656] Homology: As used herein, the term "homology" refers to the overall relatedness between polymeric molecules, e.g. between nucleic acid molecules {e.g. DNA molecules and/or RNA molecules) and/or between polypeptide molecules. In some embodiments, polymeric molecules are considered to be "homologous" to one another if their sequences are at least 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 99% identical or similar. The term "homologous" necessarily refers to a comparison between at least two sequences (polynucleotide or polypeptide sequences). In accordance with the disclosure, two polynucleotide sequences are considered to be homologous if the polypeptides they encode are at least about 50%, 60%, 70%, 80%, 90%, 95%, or even 99% for at least one stretch of at least about 20 amino acids. In some embodiments, homologous polynucleotide sequences are characterized by the ability to encode a stretch of at least 4-5 uniquely specified amino acids. For polynucleotide sequences less than 60 nucleotides in length, homology is typically determined by the ability to encode a stretch of at least 4-5 uniquely specified amino acids. In accordance with the disclosure, two protein sequences are considered to be homologous if the proteins are at least about 50%, 60%, 70%, 80%, or 90% identical for at least one stretch of at least about 20 amino acids. In many embodiments, homologous protein may show a large overall degree of homology and a high degree of homology over at least one short stretch of at least 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 35, 40, 45, 50 or more amino acids. In many embodiments, homologous proteins share one or more characteristic sequence elements. As used herein, the term "characteristic sequence element" refers to a motif present in related proteins. In some embodiments, the presence of such motifs correlates with a particular activity (such as biological activity).
[00657] Identity: As used herein, the term "identity" refers to the overall relatedness between polymeric molecules, e.g., between oligonucleotide molecules {e.g. DNA molecules and/or RNA molecules) and/or between polypeptide molecules. Calculation of the percent identity of two polynucleotide sequences, for example, may be performed by aligning the two sequences for optimal comparison purposes {e.g., gaps can be introduced in one or both of a first and a second nucleic acid sequences for optimal alignment and non-identical sequences can be disregarded for comparison purposes). In certain embodiments, the length of a sequence aligned for comparison purposes is at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, at least 95%, or 100% of the length of the reference sequence. The nucleotides at corresponding nucleotide positions are then compared. When a position in the first sequence is occupied by the same nucleotide as the corresponding position in the second sequence, then the molecules are identical at that position. The percent identity between the two sequences is a function of the number of identical positions shared by the sequences, taking into account the number of gaps, and the length of each gap, which needs to be introduced for optimal alignment of the two sequences. The comparison of sequences and determination of percent identity between two sequences can be accomplished using a mathematical algorithm. For example, the percent identity between two nucleotide sequences can be determined using methods such as those described in Computational Molecular Biology, Lesk, A. M., ed., Oxford University Press, New York, 1988;
Biocomputing: Informatics and Genome Projects, Smith, D. W., ed., Academic Press, New York, 1993; Sequence Analysis in Molecular Biology, von Heinje, G., Academic Press, 1987; Computer Analysis of Sequence Data, Part I, Griffin, A. M., and Griffin, H. G., eds., Humana Press, New Jersey, 1994; and Sequence Analysis Primer, Gribskov, M. and Devereux, J., eds., M Stockton Press, New York, 1991; each of which is incorporated herein by reference. For example, the percent identity between two nucleotide sequences can be determined, for example using the algorithm of Meyers and Miller (CABIOS, 1989, 4: 11-17), which has been incorporated into the ALIGN program (version 2.0) using a PAM120 weight residue table, a gap length penalty of 12 and a gap penalty of 4. The percent identity between two nucleotide sequences can, alternatively, be determined using the GAP program in the GCG software package using an NWSgapdna.CMP matrix. Methods commonly employed to determine percent identity between sequences include, but are not limited to those disclosed in Carillo, H., and Lipman, D., SIAM J Applied Math., 48: 1073 (1988); incorporated herein by reference. Techniques for determining identity are codified in publicly available computer programs. Exemplary computer software to determine homology between two sequences include, but are not limited to, GCG program package, Devereux, J., et ah, Nucleic Acids Research, 12(1), 387 (1984)), BLASTP, BLASTN, and FASTA Altschul, S. F. et al, J.
Molec. Biol, 215, 403 (1990)).
[00658] Inhibit expression of a gene: As used herein, the phrase "inhibit expression of a gene" means to cause a reduction in the amount of an expression product of the gene. The expression product may be RNA transcribed from the gene {e.g. mRNA) or a polypeptide translated from mRNA transcribed from the gene. Typically a reduction in the level of mRNA results in a reduction in the level of a polypeptide translated therefrom. The level of expression may be determined using standard techniques for measuring mRNA or protein.
[00659] In vitro: As used herein, the term "in vitro" refers to events that occur in an artificial environment, e.g., in a test tube or reaction vessel, in cell culture, in a Petri dish, etc., rather than within an organism {e.g., animal, plant, or microbe).
[00660] In vivo: As used herein, the term "in vivo" refers to events that occur within an organism {e.g., subject, animal, plant, or microbe or cell, niche, body fluid, tissue, organ or organ system thereof).
[00661] Isolated: As used herein, the term "isolated" is synonymous with "separated", but carries with it the inference separation was carried out by the hand of man. In one embodiment, an isolated substance or entity is one that has been separated from at least some of the components with which it was previously associated (whether in nature or in an experimental setting). Isolated substances may have varying levels of purity in reference to the substances from which they have been associated. Isolated substances and/or entities may be separated from at least about 10%, about 20%, about 30%, about 40%, about 50%, about 60%, about 70%, about 80%, about 90%, or more of the other components with which they were initially associated. In some embodiments, isolated agents are more than about 80%, about 85%, about 90%, about 91%, about 92%, about 93%, about 94%, about 95%, about 96%, about 97%, about 98%, about 99%, or more than about 99% pure. As used herein, a substance is "pure" if it is substantially free of other components.
[00662] Substantially isolated: By "substantially isolated" is meant that the compound is substantially separated from the environment in which it was formed or detected. Partial separation can include, for example, a composition enriched in the compound of the present disclosure. Substantial separation can include compositions containing at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90%, at least about 95%, at least about 97%, or at least about 99% by weight of the compound of the present disclosure, or salt thereof. Methods for isolating compounds and their salts are routine in the art. In some embodiments, isolation of a substance or entity includes disruption of chemical associations and/or bonds. In some embodiments, isolation includes only the separation from components with which the isolated substance or entity was previously combined and does not include such disruption.
[00663] Linker: As used herein, a linker refers to a moiety that connects two or more domains, moieties or entities. In one embodiment, a linker may comprise 10 or more atoms. In a further embodiment, a linker may comprise a group of atoms, e.g., 10-1,000 atoms, and can be comprised of the atoms or groups such as, but not limited to, carbon, amino, alkylamino, oxygen, sulfur, sulfoxide, sulfonyl, carbonyl, and imine. In some embodiments, a linker may comprise one or more nucleic acids comprising one or more nucleotides. In some embodiments, the linker may comprise an amino acid, peptide, polypeptide or protein. In some embodiments, a moiety bound by a linker may include, but is not limited to an atom, a chemical group, a nucleoside, a nucleotide, a nucleobase, a sugar, a nucleic acid, an amino acid, a peptide, a polypeptide, a protein, a protein complex, a payload (e.g., a therapeutic agent), or a marker (including, but not limited to a chemical, fluorescent, radioactive or bioluminescent marker). The linker can be used for any useful purpose, such as to form multimers or conjugates, as well as to administer a payload, as described herein. Examples of chemical groups that can be incorporated into the linker include, but are not limited to, alkyl, alkenyl, alkynyl, amido, amino, ether, thioether, ester, alkylene, heteroalkylene, aryl, or heterocyclyl, each of which can be optionally substituted, as described herein. Examples of linkers include, but are not limited to, unsaturated alkanes, polyethylene glycols (e.g., ethylene or propylene glycol monomeric units, e.g., diethylene glycol, dipropylene glycol, triethylene glycol, tripropylene glycol, tetraethylene glycol, or tetraethylene glycol), and dextran polymers, Other examples include, but are not limited to, cleavable moieties within the linker, such as, for example, a disulfide bond (-S-S-) or an azo bond (-N=N-), which can be cleaved using a reducing agent or photolysis. Non-limiting examples of a selectively cleavable bonds include an amido bond which may be cleaved for example by the use of tris(2-carboxyethyl)phosphine (TCEP), or other reducing agents, and/or photolysis, as well as an ester bond which may be cleaved for example by acidic or basic hydrolysis.
[00664] Modified: As used herein, the term "modified" refers to a changed state or structure of a molecule or entity as compared with a parent or reference molecule or entity. Molecules may be modified in many ways including chemically, structurally, and
functionally. In some embodiments, compounds and/or compositions of the present disclosure are modified by the introduction of non-natural amino acids.
[00665] Mutation: As used herein, the term "mutation" refers to a change and/or alteration. In some embodiments, mutations may be changes and/or alterations to proteins (including peptides and polypeptides) and/or nucleic acids (including polynucleic acids). In some embodiments, mutations comprise changes and/or alterations to a protein and/or nucleic acid sequence. Such changes and/or alterations may comprise the addition, substitution and or deletion of one or more amino acids (in the case of proteins and/or peptides) and/or nucleotides (in the case of nucleic acids and or polynucleic acids). In embodiments wherein mutations comprise the addition and/or substitution of amino acids and/or nucleotides, such additions and/or substitutions may comprise 1 or more amino acid and/or nucleotide residues and may include modified amino acids and/or nucleotides.
[00666] Naturally occurring: As used herein, "naturally occurring" means existing in nature without artificial aid, or involvement of the hand of man.
[00667] Niche: As used herein, the term "niche" refers to a place, zone and/or habbitat. In some embodiments, niches comprise cellular niches. As used herein, the term "cell niche" refers to a unique set of physiologic conditions in a cellular system within a tissue, organ or organ system within or derived from a mammalian organism. A cell niche may occur in vivo, in vitro, ex vivo, or in situ. Given the complex nature and the dynamic processes involved in growth factor signaling, a cell niche may be characterized functionally, spatially or temporally or may be used to refer to any environment that encompasses one or more cells. As such, in some embodiments a cell niche includes the environment of any cell adjacent to another cell that provides support, such as for example a nurse cell. In some embodiments, niches may include those described in International Patent Application No. WO2014074532, the contents of which are herein incorporated by reference in their entirety.
[00668] Non-human vertebrate: As used herein, a "non-human vertebrate" includes all vertebrates except Homo sapiens, including wild and domesticated species. Examples of non- human vertebrates include, but are not limited to, mammals, such as alpaca, banteng, bison, camel, cat, cattle, deer, dog, donkey, gayal, goat, guinea pig, horse, llama, mule, pig, rabbit, reindeer, sheep water buffalo, and yak.
[00669] Off-target: As used herein, "off target" refers to any unintended effect on any one or more target, gene and/or cellular transcript.
[00670] Operably linked: As used herein, the phrase "operably linked" refers to a functional connection between two or more molecules, constructs, transcripts, entities, moieties or the like.
[00671] Paratope: As used herein, a "paratope" refers to the antigen-binding site of an antibody.
[00672] Passive adsorption: As used herein, "passive adsorption" refers to a method of immobilizing solid-phase reactants on one or more surfaces (e.g. membranes, dishes, culture dishes, assay plates, etc.). Immobilization typically occurs due to affinity between such reactants and surface components. [00673] Patient: As used herein, "patient" refers to a subject who may seek or be in need of treatment, requires treatment, is receiving treatment, will receive treatment, or a subject who is under care by a trained (e.g., licensed) professional for a particular disease or condition.
[00674] Peptide: As used herein, the term "peptide" refers to a chain of amino acids that is less than or equal to about 50 amino acids long, e.g., about 5, 10, 15, 20, 25, 30, 35, 40, 45, or 50 amino acids long.
[00675] Pharmaceutically acceptable: The phrase "pharmaceutically acceptable" is employed herein to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
[00676] Pharmaceutically acceptable excipients: As used herein, the term
"pharmaceutically acceptable excipient," as used herein, refers to any ingredient other than active agents (e.g., as described herein) present in pharmaceutical compositions and having the properties of being substantially nontoxic and non-inflammatory in subjects. In some embodiments, pharmaceutically acceptable excipients are vehicles capable of suspending and/or dissolving active agents. Excipients may include, for example: antiadherents, antioxidants, binders, coatings, compression aids, disintegrants, dyes (colors), emollients, emulsifiers, fillers (diluents), film formers or coatings, flavors, fragrances, glidants (flow enhancers), lubricants, preservatives, printing inks, sorbents, suspending or dispersing agents, sweeteners, and waters of hydration. Exemplary excipients include, but are not limited to: butylated hydroxytoluene (BHT), calcium carbonate, calcium phosphate (dibasic), calcium stearate, croscarmellose, crosslinked polyvinyl pyrrolidone, citric acid, crospovidone, cysteine, ethylcellulose, gelatin, hydroxypropyl cellulose, hydroxypropyl methylcellulose, lactose, magnesium stearate, maltitol, mannitol, methionine, methylcellulose, methyl paraben, microcrystalline cellulose, polyethylene glycol, polyvinyl pyrrolidone, povidone, pregelatinized starch, propyl paraben, retinyl palmitate, shellac, silicon dioxide, sodium carboxymethyl cellulose, sodium citrate, sodium starch glycolate, sorbitol, starch (corn), stearic acid, sucrose, talc, titanium dioxide, vitamin A, vitamin E, vitamin C, and xylitol.
[00677] Pharmaceutically acceptable salts: Pharmaceutically acceptable salts of the compounds described herein are forms of the disclosed compounds wherein the acid or base moiety is in its salt form (e.g., as generated by reacting a free base group with a suitable organic acid). Examples of pharmaceutically acceptable salts include, but are not limited to, mineral or organic acid salts of basic residues such as amines; alkali or organic salts of acidic residues such as carboxylic acids; and the like. Representative acid addition salts include acetate, adipate, alginate, ascorbate, aspartate, benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphorate, camphorsulfonate, citrate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, fumarate, glucoheptonate, glycerophosphate, hemisulfate, heptonate, hexanoate, hydrobromide, hydrochloride, hydroiodide, 2-hydroxy-ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, malate, maleate, malonate, methanesulfonate, 2- naphthalenesulfonate, nicotinate, nitrate, oleate, oxalate, palmitate, pamoate, pectinate, persulfate, 3-phenylpropionate, phosphate, picrate, pivalate, propionate, stearate, succinate, sulfate, tartrate, thiocyanate, toluenesulfonate, undecanoate, valerate salts, and the like.
Representative alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, and the like, as well as nontoxic ammonium, quaternary ammonium, and amine cations, including, but not limited to ammonium, tetramethylammonium, tetraethylammonium, methylamine, dimethylamine, trimethylamine, triethylamine, ethylamine, and the like. Pharmaceutically acceptable salts include the conventional nontoxic salts, for example, from non-toxic inorganic or organic acids. In some embodiments a pharmaceutically acceptable salt is prepared from a parent compound which contains a basic or acidic moiety by conventional chemical methods. Generally, such salts can be prepared by reacting the free acid or base forms of these compounds with a stoichiometric amount of the appropriate base or acid in water or in an organic solvent, or in a mixture of the two;
generally, nonaqueous media like ether, ethyl acetate, ethanol, isopropanol, or acetonitrile are preferred. Lists of suitable salts are found in Remington's Pharmaceutical Sciences, 17th ed., Mack Publishing Company, Easton, Pa., 1985, p. 1418, Pharmaceutical Salts: Properties, Selection, and Use, P.H. Stahl and C.G. Wermuth (eds.), Wiley- VCH, 2008, and Berge et al., Journal of Pharmaceutical Science, 66, 1-19 (1977), each of which is incorporated herein by reference in its entirety. Pharmaceutically acceptable solvate: The term "pharmaceutically acceptable solvate," as used herein, refers to a crystalline form of a compound wherein molecules of a suitable solvent are incorporated in the crystal lattice. For example, solvates may be prepared by crystallization, recrystallization, or precipitation from a solution that includes organic solvents, water, or a mixture thereof. Examples of suitable solvents are ethanol, water (for example, mono-, di-, and tri-hydrates), N-methylpyrrolidinone (NMP), dimethyl sulfoxide (DMSO), N,N'-dimethylformamide (DMF), N,N'-dimethylacetamide (DM AC), l,3-dimethyl-2-imidazolidinone (DMEU), l,3-dimethyl-3,4,5,6-tetrahydro-2-(lH)- pyrimidinone (DMPU), acetonitrile (ACN), propylene glycol, ethyl acetate, benzyl alcohol, 2-pyrrolidone, benzyl benzoate, and the like. When water is the solvent, the solvate is referred to as a "hydrate." In some embodiments, the solvent incorporated into a solvate is of a type or at a level that is physiologically tolerable to an organism to which the solvate is administered (e.g., in a unit dosage form of a pharmaceutical composition).
[00678] Pharmacokinetic: As used herein, "pharmacokinetic" refers to any one or more properties of a molecule or compound as it relates to the determination of the fate of substances administered to living organisms. Pharmacokinetics are divided into several areas including the extent and rate of absorption, distribution, metabolism and excretion. This is commonly referred to as ADME where: (A) Absorption is the process of a substance entering the blood circulation; (D) Distribution is the dispersion or dissemination of substances throughout the fluids and tissues of the body; (M) Metabolism (or Biotransformation) is the irreversible transformation of parent compounds into daughter metabolites; and (E) Excretion (or Elimination) refers to the elimination of the substances from the body. In rare cases, some drugs irreversibly accumulate in body tissue.
[00679] Physicochemical: As used herein, "physicochemical" means of or relating to a physical and/or chemical property.
[00680] Preventing: As used herein, the term "preventing" refers to partially or completely delaying onset of an infection, disease, disorder and/or condition; partially or completely delaying onset of one or more symptoms, features, or clinical manifestations of a particular infection, disease, disorder, and/or condition; partially or completely delaying onset of one or more symptoms, features, or manifestations of a particular infection, disease, disorder, and/or condition; partially or completely delaying progression from an infection, a particular disease, disorder and/or condition; and/or decreasing the risk of developing pathology associated with the infection, the disease, disorder, and/or condition.
[00681] Prodrug: The present disclosure also includes prodrugs of the compounds described herein. As used herein, "prodrugs" refer to any substance, molecule or entity which is in a form predicate for that substance, molecule or entity to act as a therapeutic upon chemical or physical alteration. Prodrugs may be covalently bonded or sequestered in some way until converted into the active drug moiety prior to, upon or after administration to a mammalian subject. Prodrugs can be prepared by modifying functional groups present in the compounds in such a way that the modifications are cleaved, either in routine manipulation or in vivo, to the parent compounds. Prodrugs include compounds wherein hydroxyl, amino, sulfhydryl, or carboxyl groups are bonded to any group that, when administered to a mammalian subject, cleaves to form a free hydroxyl, amino, sulfhydryl, or carboxyl group respectively. Preparation and use of prodrugs is discussed in T. Higuchi and V. Stella, "Pro- drugs as Novel Delivery Systems," Vol. 14 of the A.C.S. Symposium Series, and in
Bioreversible Carriers in Drug Design, ed. Edward B. Roche, American Pharmaceutical Association and Pergamon Press, 1987, both of which are hereby incorporated by reference in their entirety.
[00682] Proliferate: As used herein, the term "proliferate" means to grow, expand, replicate or increase or cause to grow, expand, replicate or increase. "Proliferative" means having the ability to proliferate. "Anti-proliferative" means having properties counter to or in opposition to proliferative properties.
[00683] Protein of interest: As used herein, the terms "proteins of interest" or "desired proteins" include those provided herein and fragments, mutants, variants, and alterations thereof.
[00684] Proximal: As used herein, the term "proximal" means situated nearer to the center or to a point or region of interest.
[00685] Purified: As used herein, the term "purify" means to make substantially pure or clear from unwanted components, material defilement, admixture or imperfection. "Purified" refers to the state of being pure. "Purification" refers to the process of making pure.
[00686] Region: As used herein, the term "region" refers to a zone or general area. In some embodiments, when referring to a protein or protein module, a region may comprise a linear sequence of amino acids along the protein or protein module or may comprise a three dimensional area, an epitope and/or a cluster of eptiopes. In some embodiments, regions comprise terminal regions. As used herein, the term "terminal region" refers to regions located at the ends or termini of a given agent. When referring to proteins, terminal regions may comprise N- and/or C-termini. N-termini refer to the end of a protein comprising an amino acid with a free amino group. C-termini refer to the end of a protein comprising an amino acid with a free carboxyl group. N- and/or C-terminal regions may there for comprise the N- and/or C-termini as well as surrounding amino acids. In some embodiments, N- and/or C-terminal regions comprise from about 3 amino acid to about 30 amino acids, from about 5 amino acids to about 40 amino acids, from about 10 amino acids to about 50 amino acids, from about 20 amino acids to about 100 amino acids and/or at least 100 amino acids. In some embodiments, N-terminal regions may comprise any length of amino acids that includes the N-terminus, but does not include the C-terminus. In some embodiments, C-terminal regions may comprise any length of amino acids, that include the C-terminus, but do not comprise the N-terminus. [00687] Region of antibody recognition: As used herein, the term "region of antibody recognition" refers to one or more regions on one or more antigens or between two or more antigens that are specifically recognized and bound by corresponding antibodies. In some embodiments, regions of antibody recognition may comprise 1, 2, 3, 4, 5, 6, 7, 8, 9 or at least 10 amino acid residues. In some embodiments, regions of antibody recognition comprise a junction between two proteins or between two domains of the same protein that are in close proximity to one another.
[00688] Sample: As used herein, the term "sample" refers to an aliquot or portion taken from a source and/or provided for analysis or processing. In some embodiments, a sample is from a biological source such as a tissue, cell or component part (e.g. a body fluid, including but not limited to blood, mucus, lymphatic fluid, synovial fluid, cerebrospinal fluid, saliva, amniotic fluid, amniotic cord blood, urine, vaginal fluid and semen). In some embodiments, a sample may be or comprise a homogenate, lysate or extract prepared from a whole organism or a subset of its tissues, cells or component parts, or a fraction or portion thereof, including but not limited to, for example, plasma, serum, spinal fluid, lymph fluid, the external sections of the skin, respiratory, intestinal, and genitourinary tracts, tears, saliva, milk, blood cells, tumors, organs. In some embodiments, a sample is or comprises a medium, such as a nutrient broth or gel, which may contain cellular components, such as proteins or nucleic acid molecule. In some embodiments, a "primary" sample is an aliquot of the source. In some embodiments, a primary sample is subjected to one or more processing (e.g., separation, purification, etc.) steps to prepare a sample for analysis or other use.
[00689] Signal Sequences: As used herein, the phrase "signal sequences" refers to a sequence which can direct the transport or localization of a protein.
[00690] Single unit dose: As used herein, a "single unit dose" is a dose of any therapeutic administered in one dose/at one time/single route/single point of contact, i.e., single administration event. In some embodiments, a single unit dose is provided as a discrete dosage form (e.g., a tablet, capsule, patch, loaded syringe, vial, etc.).
[00691] Similarity: As used herein, the term "similarity" refers to the overall relatedness between polymeric molecules, e.g. between polynucleotide molecules {e.g. DNA molecules and/or RNA molecules) and/or between polypeptide molecules. Calculation of percent similarity of polymeric molecules to one another can be performed in the same manner as a calculation of percent identity, except that calculation of percent similarity takes into account conservative substitutions as is understood in the art. [00692] Split dose: As used herein, a "split dose" is the division of single unit dose or total daily dose into two or more doses.
[00693] Stable: As used herein "stable" refers to a compound or entity that is sufficiently robust to survive isolation to a useful degree of purity from a reaction mixture, and preferably capable of formulation into an efficacious therapeutic agent.
[00694] Stabilized: As used herein, the term "stabilize", "stabilized," "stabilized region" means to make or become stable. In some embodiments, stability is measured relative to an absolute value. In some embodiments, stability is measured relative to a reference compound or entity.
[00695] Subject: As used herein, the term "subject" or "patient" refers to any organism to which a composition in accordance with the disclosure may be administered, e.g., for experimental, diagnostic, prophylactic, and/or therapeutic purposes. Typical subjects include animals (e.g., mammals such as mice, rats, rabbits, non-human primates, and humans) and/or plants.
[00696] Substantially: As used herein, the term "substantially" refers to the qualitative condition of exhibiting total or near-total extent or degree of a characteristic or property of interest. One of ordinary skill in the biological arts will understand that biological and chemical phenomena rarely, if ever, go to completion and/or proceed to completeness or achieve or avoid an absolute result. The term "substantially" is therefore used herein to capture the potential lack of completeness inherent in many biological and chemical phenomena.
[00697] Substantially equal: As used herein as it relates to time differences between doses, the term means plus/minus 2%.
[00698] Substantially simultaneously: As used herein and as it relates to plurality of doses, the term typically means within about 2 seconds.
[00699] Suffering from: An individual who is "suffering from" a disease, disorder, and/or condition has been diagnosed with or displays one or more symptoms of a disease, disorder, and/or condition.
[00700] Susceptible to: An individual who is "susceptible to" a disease, disorder, and/or condition has not been diagnosed with and/or may not exhibit symptoms of the disease, disorder, and/or condition but harbors a propensity to develop a disease or its symptoms. In some embodiments, an individual who is susceptible to a disease, disorder, and/or condition (for example, cancer) may be characterized by one or more of the following: (1) a genetic mutation associated with development of the disease, disorder, and/or condition; (2) a genetic polymorphism associated with development of the disease, disorder, and/or condition; (3) increased and/or decreased expression and/or activity of a protein and/or nucleic acid associated with the disease, disorder, and/or condition; (4) habits and/or lifestyles associated with development of the disease, disorder, and/or condition; (5) a family history of the disease, disorder, and/or condition; and (6) exposure to and/or infection with a microbe associated with development of the disease, disorder, and/or condition. In some
embodiments, an individual who is susceptible to a disease, disorder, and/or condition will develop the disease, disorder, and/or condition. In some embodiments, an individual who is susceptible to a disease, disorder, and/or condition will not develop the disease, disorder, and/or condition.
[00701] Synthetic: The term "synthetic" means produced, prepared, and/or manufactured by the hand of man. Synthesis of polynucleotides or polypeptides or other molecules of the present disclosure may be chemical or enzymatic.
[00702] Targeted Cells: As used herein, "targeted cells" refers to any one or more cells of interest. The cells may be found in vitro, in vivo, in situ or in the tissue or organ of an organism. The organism may be an animal, preferably a mammal, more preferably a human and most preferably a patient.
[00703] Target site: The term "target site" as used herein, refers to a region or area targeted by a given compound, composition or method of the disclosure. Target sites may include, but are not limited to cells, tissues, organs, organ systems, niches and the like.
[00704] Therapeutic Agent: The term "therapeutic agent" refers to any agent that, when administered to a subject, has a therapeutic, diagnostic, and/or prophylactic effect and/or elicits a desired biological and/or pharmacological effect.
[00705] Therapeutically effective amount: As used herein, the term "therapeutically effective amount" means an amount of an agent to be delivered (e.g., nucleic acid, drug, therapeutic agent, diagnostic agent, prophylactic agent, etc.) that is sufficient, when administered to a subject suffering from or susceptible to an infection, disease, disorder, and/or condition, to treat, improve symptoms of, diagnose, prevent, and/or delay the onset of the infection, disease, disorder, and/or condition. In some embodiments, a therapeutically effective amount is provided in a single dose. In some embodiments, a therapeutically effective amount is administered in a dosage regimen comprising a plurality of doses. Those skilled in the art will appreciate that in some embodiments, a unit dosage form may be considered to comprise a therapeutically effective amount of a particular agent or entity if it comprises an amount that is effective when administered as part of such a dosage regimen. [00706] Therapeutically effective outcome: As used herein, the term "therapeutically effective outcome" means an outcome that is sufficient in a subject suffering from or susceptible to an infection, disease, disorder, and/or condition, to treat, improve symptoms of, diagnose, prevent, and/or delay the onset of the infection, disease, disorder, and/or condition.
[00707] Total daily dose: As used herein, a "total daily dose" is an amount given or prescribed in a 24 hr period. It may be administered as a single unit dose.
[00708] Transcription factor: As used herein, the term "transcription factor" refers to a DNA-binding protein that regulates transcription of DNA into RNA, for example, by activation or repression of transcription. Some transcription factors effect regulation of transcription alone, while others act in concert with other proteins. Some transcription factor can both activate and repress transcription under certain conditions. In general, transcription factors bind a specific target sequence or sequences highly similar to a specific consensus sequence in a regulatory region of a target gene. Transcription factors may regulate transcription of a target gene alone or in a complex with other molecules.
[00709] Treating: As used herein, the term "treating" refers to partially or completely alleviating, ameliorating, improving, relieving, delaying onset of, inhibiting progression of, reducing severity of, and/or reducing incidence of one or more symptoms or features of a particular infection, disease, disorder, and/or condition. For example, "treating" cancer may refer to inhibiting survival, growth, and/or spread of a tumor. Treatment may be administered to a subject who does not exhibit signs of a disease, disorder, and/or condition and/or to a subject who exhibits only early signs of a disease, disorder, and/or condition for the purpose of decreasing the risk of developing pathology associated with the disease, disorder, and/or condition.
[00710] Unmodified: As used herein, "unmodified" refers to any substance, compound or molecule prior to being changed in any way. Unmodified may, but does not always, refer to the wild type or native form of a biomolecule or entity. Molecules or entities may undergo a series of modifications whereby each modified product may serve as the "unmodified" starting molecule or entity for a subsequent modification.
EQUIVALENTS AND SCOPE
[00711] Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, many equivalents to the specific embodiments in accordance with the disclosure described herein. The scope of the present disclosure is not intended to be limited to the above Description, but rather is as set forth in the appended claims. [00712] In the claims, articles such as "a," "an," and "the" may mean one or more than one unless indicated to the contrary or otherwise evident from the context. Claims or descriptions that include "or" between one or more members of a group are considered satisfied if one, more than one, or all of the group members are present in, employed in, or otherwise relevant to a given product or process unless indicated to the contrary or otherwise evident from the context. The disclosure includes embodiments in which exactly one member of the group is present in, employed in, or otherwise relevant to a given product or process. The disclosure includes embodiments in which more than one, or the entire group members are present in, employed in, or otherwise relevant to a given product or process.
[00713] It is also noted that the term "comprising" is intended to be open and permits but does not require the inclusion of additional elements or steps. When the term "comprising" is used herein, the term "consisting of is thus also encompassed and disclosed.
[00714] Where ranges are given, endpoints are included. Furthermore, it is to be understood that unless otherwise indicated or otherwise evident from the context and understanding of one of ordinary skill in the art, values that are expressed as ranges can assume any specific value or subrange within the stated ranges in different embodiments of the disclosure, to the tenth of the unit of the lower limit of the range, unless the context clearly dictates otherwise.
[00715] In addition, it is to be understood that any particular embodiment of the present disclosure that falls within the prior art may be explicitly excluded from any one or more of the claims. Since such embodiments are deemed to be known to one of ordinary skill in the art, they may be excluded even if the exclusion is not set forth explicitly herein. Any particular embodiment of the compositions of the disclosure (e.g. , any nucleic acid or protein encoded thereby; any method of production; any method of use; etc.) can be excluded from any one or more claims, for any reason, whether or not related to the existence of prior art.
[00716] All cited sources, for example, references, publications, databases, database entries, and art cited herein, are incorporated into this application by reference, even if not expressly stated in the citation. In case of conflicting statements of a cited source and the instant application, the statement in the instant application shall control.
[00717] Section and table headings are not intended to be limiting.
EXAMPLES
Example 1. Protein expression method
[00718] Cells (293-6E cells) transiently expressing tagged proteins are cultured in serum- free medium (FreeStyle F17 medium, Life Technologies, Carlsbad, CA) supplemented with 4 mM glutamine, 0.1% Kolliphor P 188 and 25 μg/ml G418. Once their viability drops below 50%, tissue culture supernatant is collected and cleared by centrifugation for 10 minutes at 450 x gravity at 4°C. Supernatant is then filtered by passing it through a 0.22 μιη pore filter. Filtered supernatant is combined with Tris, NaCl and NiC12 for a final concentration of 50 mM Tris pH 8.0, 350 mM NaCl and 0.5 mM NiC12. 1 ml of the adjusted solution is collected for later analysis by SDS-poly acrylamide gel electrophoresis (PAGE) or Western blot. The remaining portion of the adjusted solution is combined with washed Ni-NTA resin (Qiagen, Valencia, CA) at a ratio dependent upon the expression level of the protein, which has been previously determined during small scale trials of the protein. This combined solution is then stirred at 4°C overnight using a suspended magnetic stir bar (to prevent grinding of Ni-NTA resin). Ni-NTA resin is collected the next morning using an Econo-glass column (BioRad, Waltham, MA) with a porous bottom membrane. Repeated direct application of the combined solution to the top of the column loads the Ni NTA resin in a uniform fashion onto the column, alternatively a siphoning system is used to continuously load large volumes of combined solution.
[00719] Next, the column is washed with 10 column volumes (CV) of wash buffer (20 mM Tris, pH 8.0, 500 mM NaCl and 20 mM imidazole). An aliquot of the last wash is collected for analysis. The column is then eluted with elution buffer (20 mM Tris, pH 8.0, 500 mM NaCl and 300 mM imidazole) in increments of 1 CV after incubating on the Ni-NTA resin for 5 minutes at 4C, until no protein remains on the Ni NTA resin.
[00720] The absorbance at 280 nm is measured in each of the eluted fractions collected and compared to the absorbance at 280 nm of blank elution buffer. Earlier fractions typically have negative absorption due to the imidazole gradient; however, fractions containing higher amounts of protein have positive values. Collected fractions are then run on SDS-PAGE for analysis and relevant fractions are pooled and concentrated for further purification.
[00721] The protein is further purified by size exclusion chromatography (SEC) using a variety of columns including but not limited to (S200, SRT10, S200 Prep Grade, Superose 6) equilibrated in a variety of buffers including but not limited to PBS, or 20 mM Hepes pH 7.5, 500 mM NaCl. Peak fractions are pooled and concentrated to a concentration of 1-2 mg/mL and aliquots are flash-frozen and stored at -80C.
[00722] Parental FLP-INTM T-REXTM 293 cells (Life Technologies, Carlsbad, CA) are cultured in DMEM media according to the manufacturer' s instructions under Zeocin and Blasticidin selection (lOOug/ml Zeocin and 15 ug/ml Blasticidin) to maintain their naive state. To introduce a protein expression construct of interest into the parental cell line, selection is removed 24 hours in advance and the media switched to Opti-MEM after plating the cells in 6-well plates. Cells are transfected with Lipofectamine 2000 from Life
Technologies (Carlsbad, CA), using a ratio of 9: 1 pOG44 plasmid (Flp recombinase plasmid) to pcDNA5/TO (Life Technologies, Carlsbad, CA) plasmid with the expression construct of interest in the multiple cloning site. After 24 hours, cells are switched back to DMEM and 24 hours subsequently cells are expanded to 10cm plates. When the cells have adhered, selection with Hygromycin and Blasticidin is applied at 100 ug/ml and 15 ug/ml. Upon integration at the Flp site in the genome, Zeocin resistance is lost and Hygromycin resistance is gained in addition to the protein expression construct of interest.
[00723] To induce protein expression, stable Flp-In Trex 293 cells are grown to 90% confluence in DMEM. Cells are then washed with PBS and switched to F17 media supplemented with 4 mM GlutaMax (Life Technologies, Carlsbad, CA) in the dish. To induce protein expression, 1 ug/ml tetracycline is added. One day later, Trytone Nl is added to 0.5% weight/volume to provide nutrients for protein expression. Culture continues for up to five days before the supernatant is collected. For certain expression constructs, fresh F17 with 1 ug/ml tetracycline is added to the cells and culture continued for another five days. Tryptone Nl is again added one day after refreshing the cells with new media. Collected supernatant is processed and purified as in paragraph described above.
[00724] To increase yield per liter, ease of culture, and volume of culture, stable adherent Flp-IN T-Rex 293 cell lines for certain constructs are adapted to suspension shaking culture in two steps, first to serum free growth and then shaking growth. Cells are slowly transitioned from DMEM complete media recommended by Life Technologies (Carlsbad, CA) to F17 supplemented with 4 mM GlutaMax, and then shaking culture as 0.2% Kolliphor P188 is added to the F17 supplemented with 4mM GlutaMax. Selection is not maintained during the transition, instead it is reapplied after adaptation to F17 and then again after adaption to shaking culture. To adapt to F17, a stepwise dilution of DMEM with F17 is pursued in increments of 25%. At the last step, a small amount of FBS is added to the F17 - approximately 0.2% final concentration - before the final split which is into 100% F17. Cells are split at high ratios of 1:3 or lower, decreasing the time out of selection. As these cells grow robustly, the entire adaption can be completed in about 10 days. After the adaptation is complete, selection is reapplied for at least two passages at 20ug/ml Hygromycin and 2 ug/ml Blasticidin.
[00725] Following adaptation to F 17 in adherent culture and the reapplication of selection, the adherent cells can then be adapted to suspension culture. Two 15 cm diameter dishes are grown to confhiency and trypsinized, counted and then seeded in fresh F17 supplemented with 0.2% Kolliphor P 188 and 4 mM GlutaMax at greater than 0.75 x 106/ml. Cell viability decreases initially, but then increases as does cell number. When the cell doubling time approaches 30 hours, Blasticidin and Hygromycin is in some cases reapplied at 2 ug/ml and 20 ug/ml respectively, but only in the fresh media added to split cells in order to gradually reintroduce the selection.
[00726] Some of the proteins produced comprise additional amino acids encoding one or more detectable labels for purification [e.g. polyhistidine tag, flag tag (DYKDDDDK; SEQ ID NO: 67), etc.] Some proteins are N-terminally labeled, C-terminally labeled and/or biotinylated.
[00727] Some of the proteins produced comprise additional amino acids encoding one or more 3C protease cleavage site (LEVLFQGP; SEQ ID NO: 68) Such sites allow for cleavage between residues Q and G of the 3C protease cleavage site upon treatment with 3C protease, including with rhinovirus 3C protease. In some cases, cleavage sites are introduced to allow for removal of detectable labels from recombinant proteins.
Example 2. Generation of antibodies
Antibodies produced by standard monoclonal antibody generation
[00728] Antibodies are generated in knockout mice, lacking the gene that encodes for desired target antigens. Such mice are not tolerized to target antigens and therefore generate antibodies against such antigens that may cross react with human and mouse forms of the antigen. For the production of monoclonal antibodies, host mice are immunized with recombinant proteins to elicit lymphocytes that specifically bind to these proteins.
Lymphocytes are collected and fused with immortalized cell lines. The resulting hybridoma cells are cultured in a suitable culture medium with selection agents to support the growth of only fused cells.
[00729] Desired hybridoma cell lines are then identified through binding specificity analysis of the secreted antibodies for the target peptide and clones of these cells are subcloned through limiting dilution procedures and grown by standard methods. Antibodies produced by these cells are isolated and purified from the culture medium by standard immunoglobulin purification procedures
Antibodies produced recombinantly [00730] Recombinant antibodies are produced using the hybridoma cells produced above. Heavy and light chain variable region cDNA sequences of the antibodies are determined using standard biochemical techniques. Total RNA are extracted from antibody-producing hybridoma cells and converted to cDNA by reverse transcriptase (RT) polymerase chain reaction (PCR). PCR amplification is carried out on the resulting cDNA using primers specific for amplification of the heavy and light chain sequences. PCR products are then subcloned into plasmids for sequence analysis. Once sequenced, antibody coding sequences are placed into expression vectors. For humanization, coding sequences for human heavy and light chain constant domains are used to substitute for homologous murine sequences. The resulting constructs are transfected into mammalian cells capable of large scale translation.
Antibodies produced by using antibody fragment display library screening techniques
[00731] Antibodies of the present disclosure may be produced using high throughput methods of discovery. Synthetic antibodies are designed by screening target antigens using a phage display library. The phage display libraries are composed of millions to billions of phage particles, each expressing a unique single chain variable fragment (scFv) on their viral coat. In scFv libraries, the cDNA encoding each fragment contains a similar sequence with the exception of unique sequences encoding the variable loops of the complementarity determining regions (CDRs). VH domains are expressed as fusion proteins, linked to the N- terminus of the viral pill coat protein. VL domains are expressed separately and assemble with the VH domain in the periplasm prior to incorporation of the complex into the viral coat. Target antigens are incubated, in vitro, with members of phage display libraries and bound phage particles are precipitated. The cDNA encoding the scFvs of the bound Fab subunits is sequenced from the bound phage. The cDNA sequence is directly incorporated into antibody sequences for recombinant antibody production, or mutated and utilized for further optimization through in vitro affinity maturation.
Antibodies produced using affinity maturation techniques
[00732] scFvs capable of binding target antigens are identified using the libraries described above and high affinity mutants are derived from these through the process of affinity maturation. Affinity maturation technology is used to identify sequences encoding CDRs that have the highest affinity for the target antigen. Using this technology, the CDR sequences isolated using the phage display library selection process described above are mutated randomly as a whole or at specific residues to create millions to billions of variants. These variants are expressed in scFv fusion proteins in a phage display library and screened for their ability to bind the target antigen. Several rounds of selection, mutation and expression are carried out to identify antibody fragment sequences with the highest affinity for the target antigen. These sequences can be directly incorporated into antibody sequences for recombinant antibody production.
Example 3. Identification and characterization of antibodies directed to recombinant proteins
[00733] Recombinant proteins are synthesized according to the method of Example 1 or obtained from commercial sources. Recombinant proteins expressed include those listed in Table 19.
Table 19. Recombinant proteins
Figure imgf000313_0001
[00734] Both human and non-human (including, but not limited to mouse) isoforms of the recombinant proteins listed in Table 19 are expressed.
[00735] Antibodies are generated according to the methods described in Example 2, which bind to recombinant proteins expressed and are subjected to screening to identify antibodies with desired binding properties. ELISA assays are used initially to identify antibody candidates that demonstrate affinity for desired antigens, while showing reduced or no affinity for undesired antigens.
Example 4. Myostatin proliferation assay [00736] C2C12 murine myoblasts (ATCC, Manassas, VA) are cultured in Dulbecco's modified essential medium (DMEM; Life Technologies, Carlsbad, CA) with 10% fetal bovine serum (FBS; Life Technologies, Carlsbad, CA) prior to carrying out the assay. The percentage of FBS is varied and/or replaced with bovine serum albumin (BSA) at varying concentrations. Cell proliferation assays are conducted in uncoated 96-well plates. C2C12 cultures are seeded at 1000 cells per well. After allowing the cells to attach for 16 hours, myostatin test media is added. Recombinant human myostatin (R&D Systems, Minneapolis, MN) is used for standard curve generation. For experimental systems, the supernatant from 293E cells overexpressing myostatin is added, following treatment with experimental antibodies. All samples are run in replicates of 8. Plates are incubated for 72 hours in an atmosphere of 37°C and 5% CO2. Proliferation is assessed using a CellTiter-Glo®
Luminescent Cell Viability Assay (Promega Biosciences, LLC, Madison, WI) whereby cell lysis generates a luminescent signal proportional to the amount of ATP present, which is directly proportional to the number of cells present in culture (Thomas, M. et al., Myostatin, a negative regulator of muscle growth, functions by inhibiting myoblast proliferation. 2000. 275(51):40235-43).
Example 5. Protein expression and purification by Ni-NTA
[00737] Cells (293-6E cells or FLP-IN™ T-REX™ 293 cells) expressing His-tagged proteins are cultured in serum-free medium (FreeStyle F17 medium, Life Technologies, Carlsbad, CA) supplemented with 4 mM glutamine, 0.1% Pluronic F68 and 25 μg/ml G418. Once their viability drops below 50%, tissue culture supernatant is collected and cleared by centrifugation for 10 minutes at 200 x gravity at 4°C. Supernatant is then filtered by passing it through a 0.22 or 0.45 μιη pore filter. Filtered supernatant is combined with Tris, NaCl and NiCl2 for a final concentration of 50 mM Tris pH 8.0, 500 mM NaCl and 0.5 mM NiCl2. 1 ml of the adjusted solution is collected for later analysis by SDS-poly acrylamide gel
electrophoresis (PAGE) or Western blot, while another portion of the adjusted solution is combined with washed Ni-NTA resin (Life Technologies, Carlsbad, CA) at a concentration of 5-10 ml of Ni-NTA resin per 300 ml of the adjusted solution. This combined solution is then stirred at 4°C using a suspended magnetic stir bar (to prevent grinding of Ni-NTA agarose). Ni-NTA resin is next collected by centrifugation at 200 x gravity at 4°C for 10 minutes.
[00738] Next, the column is washed with 15 column volumes (CV) of wash buffer (20 mM Tris, pH 8.0, 500 mM NaCl and 20 mM imidazole). An aliquot of the last wash is collected for analysis. The column is then eluted with 3 CV of elution buffer (20 mM Tris, pH 8.0, 500 mM NaCl and 300 mM imidazole) and 1/3 column volume fractions are collected for analysis.
[00739] The absorbance at 280 nm is measured in each of the eluted fractions collected and compared to the absorbance at 280 nm of blank elution buffer. Earlier fractions typically have negative absorption due to the imidazole gradient; however, fractions containing higher amounts of protein have positive values. Collected fractions are then run on SDS-PAGE for analysis and relevant fractions are pooled for further purification.
[00740] The protein is further purified by size exclusion chromatography (SEC) using an S200 column equilibrated either in PBS, or 20 mM Hepes pH 7.5, 500 mM NaCl. Peak fractions are pooled and concentrated to a concentration of 1-2 mg/mL and aliquots are flash- frozen and stored at -80C.
Example 6. Design of GDF-8/GDF-ll/activin chimeras
[00741] The structure -based alignment, conducted between GDF-8 (myostatin), GDF-11, Inhibin A and a GDF-8 dimer (FIG. 10), was used to construct three-dimensional models of potential chimeric proteins comprising combinations of modules from GDF-8 and GDF-11 using the Schrodinger Bioluminate software. A chimeric model of GDF-8 comprising an arm region of GDF-11 (SEQ ID NO: 187) revealed a region of potential steric clash involving GDF-11 residue F95. According to the model, F95 from the GDF-11 arm causes
destabilization of the a2 helix of the chimeric GPC. Therefore, GDF8/GDF11/Activin chimeras were designed so that the ARM region of the chimera contains the 2 helix.
[00742] Chimeric proteins designed include those listed in Table 20. The first of these is comprised of residues 1-64 of GDF-8 followed by residues 87-274 of GDF-11 and residues 244-352 of GDF-8. The second is comprised of residues 1-64 of GDF-8 follwed by residues 87-274 of GDF-11. These chimeric proteins were further combined with N-terminal secretion signal peptides, flag tags, histidine tags and 3C protease cleavage site.
Table 20. Chimeric proteins
Protein Amino Acid Sequence SEQ
ID NO
GDF- MDMRVPAQLLGLLLLWFSGVLGDYKDDDDKHHHHHHLEVLFQ 1635 8/11 GPVDLNENSEQKENVEKEGLCNACTWRQNTKSSRIEAIKIQILSKL
chimeric RLETAPNISKDVIRQLLPKAPPLQQILDLHDFQGDALQPEDFLEED
protein EYHATTETVISMAQETDPAVQTDGSPLCCHFHFSPKVMFTKVLKA
1 QLWVYLRPVPRPATVYLQILRLKPLTGEGTAGGGGGGRRHIRIRS LKIELHSRSGHWQSIDFKQVLHSWFRQPQSNWGIEINAFDPSGTDL
AVTSLGPGAEGLHPFMELRVLENTKRSRRDFGLDCDEHSTESRCC
RYPLTVDFEAFGWDWIIAPKRYKANYCSGECEFVFLQKYPHTHL
VHQANPRGSAGPCCTPTKMSPINMLYFNGKEQIIYGKIPAMVVDR
CGCS
GDF- MDMRVPAQLLGLLLLWFSGVLGDYKDDDDKHHHHHHLEVLFQ 1636 8/11 GPNENSEQKENVEKEGLCNACT WRQNTKS SRIE AIKIQILS KLRLE
chimeric TAPNISKDVIRQLLPKAPPLQQILDLHDFQGDALQPEDFLEEDEYH
protein ATTETVISMAQETDPAVQTDGSPLCCHFHFSPKVMFTKVLKAQL
2 WVYLRPVPRPATVYLQILRLKPLTGEGTAGGGGGGRRHIRIRSLKI
ELHSRSGHWQSIDFKQVLHSWFRQPQSNWGIEINAFDPSGTDLAV
TSLGPGAEGLHPFMELRVLENTKRSRR
Example 7. Analysis of recombinantly expressed GDF-8
[00743] Histidine-tagged proGDF-8 was expressed according to the methods of Example 1. Purified proteins were analyzed by SDS-PAGE under either reducing or non-reducing conditions (to maintain protein dimers). FIG. 11 depicts the results indicating successful expression of these proteins and protein complexes.
Example 8. Fc Fusion constructs
[00744] GDF-8 and GDF- 11 fusion constructs were designed to improve expression antigens needed for antibody production, selection and/or testing (see Table 21). These constructs were developed to comprise the GDF- 11 or GDF-8 prodomain amino acid sequence joined C-terminally to an Fc domain. The constructs were further designed to comprise an N-terminal secretion signal and a C-terminal histidine tag.
Table 21. Fc fusion proteins
Protein Amino Acid Sequence SEQ
ID NO
GDF-8 MDMRVPAQLLGLLLLWFSGVLGNENSEQKENVEKEGLCNACTW 1637 prodom RQNTKSSRIEAIKIQILSKLRLETAPNISKDVIRQLLPKAPPLRELID
ain Fc- QYDVQRDDSSDGSLEDDDYHATTETIITMPTESDFLMQVDGKPKC
fusion CFFKFSSKIQYNKVVKAQLWIYLRPVETPTTVFVQILRLIKPMKDG
protein TRYTGIRSLKLDMNPGTGIWQSIDVKTVLQNWLKQPESNLGIEIK
ALDENGHDLAVTFPGPGEDGLNPFLEVKVTDTPKRSDKTHTCPPC
PAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKF
NWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGK
EYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSREEMTKNQ
VSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLY
SKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGKHHH
HHH
GDF-8 MDMRVPAQLLGLLLLWFSGVLGNENSEQKENVEKEGLCNACTW 1638 prodom RQNTKSSRIEAIKIQILSKLRLETAPNISKDVIRQLLPKAPPLRELID
ain QYDVQRADSSDGSLEDDDYHATTETIITMPTESDFLMQVDGKPKC
D76A CFFKFSSKIQYNKVVKAQLWIYLRPVETPTTVFVQILRLIKPMKDG Fc TRYTGIRSLKLDMNPGTGIWQSIDVKTVLQNWLKQPESNLGIEIK fusion ALDENGHDLAVTFPGPGEDGLNPFLEVKVTDTPKRSDKTHTCPPC Protein PAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKF
NWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGK
EYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSREEMTKNQ
VSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLY
SKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGKHHH
HHH
GDF-11 1639 prodom MDMRVPAQLLGLLLLWFSGVLGAEGPAAAAAAAAAAAAAGVG ain GEASSAPAPSVAPEPDGCPVCVWRQHSRELRLESIKSQILSKLRLK
D98A EAPNISREVVKQLLPKAPPLQQILDLHDFQGDALQPEDFLEEDEYH Fc- ATTETVISMAQETDPAVQTDGSPLCCHFHFSPKVMFTKVLKAQL fusion WVYLRPVPRPATVYLQILRLKPLTGEGTAGGGGGGRRHIRIRSLKI protein ELHSRSGHWQSIDFKQVLHSWFRQPQSNWGIEINAFDPSGTDLAV
TSLGPGAEGLHPFMELRVLENTKRSDKTHTCPPCPAPELLGGPSVF
LFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVH
NAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKAL
PAPIEKTISKAKGQPREPQVYTLPPSREEMTKNQVSLTCLVKGFYP
SDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQ
GNVFSCSVMHEALHNHYTQKSLSLSPGKHHHHHH
GDF-11 MDMRVPAQLLGLLLLWFSGVLGAEGPAAAAAAAAAAAAAGVG 1640 prodom GERSSRPAPSVAPEPDGCPVCVWRQHSRELRLESIKSQILSKLRLK ain Fc- EAPNISREVVKQLLPKAPPLQQILDLHDFQGDALQPEDFLEEDEYH fusion ATTETVISMAQETDPAVQTDGSPLCCHFHFSPKVMFTKVLKAQL protein WVYLRPVPRPATVYLQILRLKPLTGEGTAGGGGGGRRHIRIRSLKI
ELHSRSGHWQSIDFKQVLHSWFRQPQSNWGIEINAFDPSGTDLAV
TSLGPGAEGLHPFMELRVLENTKRSDKTHTCPPCPAPELLGGPSVF
LFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVH
NAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKAL
PAPIEKTISKAKGQPREPQVYTLPPSREEMTKNQVSLTCLVKGFYP
SDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQ
GNVFSCSVMHEALHNHYTQKSLSLSPGKHHHHHH
Example 9. GASP constructs
[00745] GASP constructs were designed using fragments from published sequences. Constructs were designed with various combinations of signal sequences, 3C protease cleavage sites, histidine tags and biotinylation sequences. The designed constructs include those listed in the Table below.
Table 22. GASP constructs
Protein Sequence SEQ
ID NO
GASP1 construct MDMRVPAQLLGLLLLWFSGVLGSVVRGHQAAAT 1641 with residue 303- SESSPNGTAFPAAECLKPPDSEDCGEEQTRWHFD
576 of AQANNCLTFTFGHCHRNLNHFETYEACMLACMS
NP_783165.1 GPLAACSLPALQGPCKAYAPRWAYNSQTGQCQS
FVYGGCEGNGNNFESREACEESCPFPRGNQRCRA
CKPRQKLVTSFCRSDFVILGRVSELTEEPDSGRAL VTVDEVLKDEKMGLKFLGQEPLEVTLLHVDWAC PCPNVTVSEMPLIIMGEVDGGMAMLRPDSFVGAS SARRVRKLREVMHKKTCDVLKEFLGLHLEVLFQ GPHHHHHHGLNDIFEAQKIEWHE
GASP1 construct MDMRVPAQLLGLLLLWFSGVLGLPPIRYSHAGIC 1642 with residue 35- PNDMNPNLWVDAQSTCRRECETDQECETYEKCC 576 of PNVCGTKSCVAARYMDVKGKKGPVGMPKEATC
NP_783165.1 DHFMCLQQGSECDIWDGQPVCKCKDRCEKEPSFT
CASDGLTYYNRCYMDAEACSKGITLAVVTCRYH
FTWPNTSPPPPETTMHPTTASPETPELDMAAPALL
NNPVHQSVTMGETVSFLCDVVGRPRPEITWEKQL
EDRENVVMRPNHVRGNVVVTNIAQLVIYNAQLQ
DAGIYTCTARNVAGVLRADFPLSVVRGHQAAATS
ESSPNGTAFPAAECLKPPDSEDCGEEQTRWHFDA
QANNCLTFTFGHCHRNLNHFETYEACMLACMSG
PLAACSLPALQGPCKAYAPRWAYNSQTGQCQSF
VYGGCEGNGNNFESREACEESCPFPRGNQRCRAC
KPRQKLVTSFCRSDFVILGRVSELTEEPDSGRALV
TVDEVLKDEKMGLKFLGQEPLEVTLLHVDWACP
CPNVTVSEMPLIIMGEVDGGMAMLRPDSFVGASS
ARRVRKLREVMHKKTCDVLKEFLGLHLEVLFQG
PHHHHHHGLNDIFEAQKIEWHE
GASP1 construct MDMRVPAQLLGLLLLWFSGVLGGLNDIFEAQKIE 1643 with residue 384- WHEHHHHHHLEVLFQGPAACSLPALQGPCKAYA 576 of PRWAYNSQTGQCQSFVYGGCEGNGNNFESREAC
NP_783165.1 EESCPFPRGNQRCRACKPRQKLVTSFCRSDFVILG
RVSELTEEPDSGRALVTVDEVLKDEKMGLKFLGQ
EPLEVTLLHVDWACPCPNVTVSEMPLIIMGEVDG
GMAMLRPDSFVGASSARRVRKLREVMHKKTCDV
LKEFLGLH
GASP1 construct MDMRVPAQLLGLLLLWFSGVLGFPRGNQRCRAC 1644 with residue 438- KPRQKLVTSFCRSDFVILGRVSELTEEPDSGRALV 576 of TVDEVLKDEKMGLKFLGQEPLEVTLLHVDWACP
NP_783165.1 CPNVTVSEMPLIIMGEVDGGMAMLRPDSFVGASS
ARRVRKLREVMHKKTCDVLKEFLGLHLEVLFQG PHHHHHHGLNDIFEAQKIEWHE
GASP construct MDMRVPAQLLGLLLLWFSGVLGSVVQREPARDA 1645 with residue 279- APSIPAPAECLPDVQACTGPTSPHLVLWHYDPQR
548 of GGCMTFPARGCDGAARGFETYEACQQACARGPG
NP_444514.1 DACVLPAVQGPCRGWEPRWAYSPLLQQCHPFVY
GGCEGNGNNFHSRESCEDACPVPRTPPCRACRLR
SKLALSLCRSDFAIVGRLTEVLEEPEAAGGIARVA
LEDVLKDDKMGLKFLGTKYLEVTLSGMDWACPC
PNMTAGDGPLVIMGEVRDGVAVLDAGSYVRAAS
EKRVKKILELLEKQACELLNRFQDLEVLFQGPHH
HHHHGLNDIFEAQKIEWHE
GASP construct MDMRVPAQLLGLLLLWFSGVLGAGLLPGLGSHP 1646 with residue 20- GVCPNQLSPNLWVDAQSTCERECSRDQDCAAAE 548 of KCCINVCGLHSCVAARFPGSPAAPTTAASCEGFVC
NP_444514.1 PQQGSDCDIWDGQPVCRCRDRCEKEPSFTCASDG
LTYYNRCYMDAEACLRGLHLHIVPCKHVLSWPPS
SPGPPETTARPTPGAAPVPPALYSSPSPQAVQVGG
TASLHCDVSGRPPPAVTWEKQSHQRENLIMRPDQ
MYGNVVVTSIGQLVLYNARPEDAGLYTCTARNA AGLLRADFPLSVVQREPARDAAPSIPAPAECLPDV
QACTGPTSPHLVLWHYDPQRGGCMTFPARGCDG
AARGFETYEACQQACARGPGDACVLPAVQGPCR
GWEPRWAYSPLLQQCHPFVYGGCEGNGNNFHSR
ESCEDACPVPRTPPCRACRLRSKLALSLCRSDFAI
VGRLTEVLEEPEAAGGIARVALEDVLKDDKMGL
KFLGTKYLEVTLSGMDWACPCPNMTAGDGPLVI
MGEVRDGVAVLDAGSYVRAASEKRVKKILELLE
KQACELLNRFQDLEVLFQGPHHHHHHGLNDIFEA
QKIEWHE
GASP construct MDMRVPAQLLGLLLLWFSGVLGGLNDIFEAQKIE 1647 with residue 357- WHEHHHHHHLEVLFQGPDACVLPAVQGPCRGW
548 of EPRWAYSPLLQQCHPFVYGGCEGNGNNFHSRESC
NP_444514.1 EDACPVPRTPPCRACRLRSKLALSLCRSDFAIVGR
LTEVLEEPEAAGGIARVALEDVLKDDKMGLKFLG
TKYLEVTLSGMDWACPCPNMTAGDGPLVIMGEV
RDGVAVLDAGSYVRAASEKRVKKILELLEKQACE
LLNRFQD
GASP construct MDMRVPAQLLGLLLLWFSGVLGVPRTPPCRACR 1648 with residue 411- LRSKLALSLCRSDFAIVGRLTEVLEEPEAAGGIAR
548 of VALEDVLKDDKMGLKFLGTKYLEVTLSGMDWA
NP_444514.1 CPCPNMTAGDGPLVIMGEVRDGVAVLDAGSYVR
AASEKRVKKILELLEKQACELLNRFQDLEVLFQGP
HHHHHHGLNDIFEAQKIEWHE
Example 10. Perlecan construct
[00746] Perlecan constructs were designed using fragments from published sequences. Constructs were designed with various combinations of signal sequences, 3C protease cleavage sites, histidine tags and biotinylation sequences. The designed constructs include those listed in Table 23.
Table 23. Perlecan construct
Protein Sequence SEQ
ID NO
Perlecan MDMRVPAQLLGLLLLWFSGVLGAFAHLQVPERVVPYFTQTPYS 1649 construct FLPLPTIKDAYRKFEIKITFRPDSADGMLLYNGQKRVPGSPTNLA with NRQPDFISFGLVGGRPEFRFDAGSGMATIRHPTPLALGHFHTVTL residues LRSLTQGSLIVGDLAPVNGTSQGKFQGLDLNEELYLGGYPDYGA
3653-4392 IPKAGLSSGFIGCVRELRIQGEEIVFHDLNLTAHGISHCPTCRDRP of CQNGGQCHDSESSSYVCVCPAGFTGSRCEHSQALHCHPEACGP
NP_001278 DATCVNRPDGRGYTCRCHLGRSGLRCEEGVTVTTPSLSGAGSYL 789.1 ALPALTNTHHELRLDVEFKPLAPDGVLLFSGGKSGPVEDFVSLA
MVGGHLEFRYELGSGLAVLRSAEPLALGRWHRVSAERLNKDGS
LRVNGGRPVLRSSPGKSQGLNLHTLLYLGGVEPSVPLSPATNMS
AHFRGCVGEVSVNGKRLDLTYSFLGSQGIGQCYDSSPCERQPCQ
HGATCMPAGEYEFQCLCRDGFKGDLCEHEENPCQLREPCLHGG
TCQGTRCLCLPGFSGPRCQQGSGHGIAESDWHLEGSGGNDAPG
QYGAYFHDDGFLAFPGHVFSRSLPEVPETIELEVRTSTASGLLLW
QGVEVGEAGQGKDFISLGLQDGHLVFRYQLGSGEARLVSEDPIN DGEWHRVTALREGRRGSIQVDGEELVSGRSPGPNVAVNAKGSV
YIGGAPDVATLTGGRFSSGITGCVKNLVLHSARPGAPPPQPLDLQ I HRAQAGANTRPCPSLEVLFQGPHHHHHHGLNDIFEAQKIEWHE |
Example 11. ELISA analysis
[00747] Enzyme-linked immunosorbent assay (ELISA) analysis is carried out to assess antibody binding. 96-well ELISA assay plates are coated with neutravidin, a deglycosylated version of streptavidin with a more neutral pi. Target proteins are expressed with or without histidine (His) tags and subjected to biotinylation. Biotinylated target proteins are incubated with neutravidin-coated ELISA assay plates for two hours at room temperature and unbound proteins are removed by washing three times with wash buffer (either 25 mM Tris, 150 mM NaCl, 0.05% TWEEN®-20, or 20 mM Hepes pH 7.5, 500 mM NaCl, 0.05% TWEEN®-20). Primary antibodies being tested are added to each well and allowed to incubate at room temperature for 1 hour or more. Unbound antibody is then removed by washing three times with wash buffer. Secondary antibodies capable of binding to primary antibodies being tested and conjugated with detectable labels are then incubated in each well for 30 minutes at room temperature. Unbound secondary antibodies are removed by washing three times with wash buffer. Finally, bound secondary antibodies are detected by enzymatic reaction, fluorescence detection and/or luminescence detection, depending on the detectable label present on secondary antibodies being detected.
Example 12. Identification of antibodies that modulate GDF-8 activity
[00748] Antibody development campaigns were carried out to generate antibodies capable of modulating GDF-8 activity. For selection of antibody variable domain binding partners, a phage library, constructed according to the methods described by McCafferty et al
(McCafferty, et al., 1990. Nature. 348:552-4), was used to enrich for phage using antigens including primary antigens listed in Table 24.
Table 24. Recombinant antigens
Program # Primary antigen Screening antigens
(for panning &
primary screen)
1 proGDF-8 (SEQ ID proGDF-8, proGDF-11 , and murine proTGF- 1 NO: 5, 10% furin C4S (His control)
cleaved)
2 Latent proGDF-8 Latent proGDF-8, proGDF-8, proGDF-11, and
(SEQ ID NO: 5, 90% murine proTGF- 1 C4S (His control)
furin cleaved) [00749] The goal of programs 1 and 2 was to develop antibodies capable of modulating proGDF-8 processing and/or GDF-8 growth factor release from proGDF-8 and/or the latent proGDF-8 complex. When synthesized, proGDF-8 requires cleavage by furin to cleave the GDF-8 prodomain from the growth factor and to allow formation of the latent GPC. In order for release of growth factor from this latent complex to occur, a second cleavage is required by a member of the BMP-l/Tolloid-like proteinase (B/TP) family, which includes mTLL2. In order to maximize opportunities for the generation of inhibiting antibodies, proGDF-8 antigens with reduced (program 1) or enhanced (program 2) furin processing were used as antigens for phage enrichment.
[00750] Both solid as well as solution-phase enrichment procedures were carried out. Solid-phase enrichment comprised the use of biotinylated antigen coated onto neutravidin plates or direct absorption of non-biotinylated antigen onto plastic plates. Solution-phase enrichment utilized biotinylated antigens bound to streptavidin beads. Non-specific antibodies were removed by extensive washing and specific phage antibodies retained by immobilized antigens were recovered by elution. Eluted phage were used to infect E. coli cultures, together with helper phage, to generate an amplified output library for the next round of panning (wherein one round of panning includes incubation, washing, elution and amplification. The success of selection at each round was monitored by comparing output size with a background panning without antigen coating. A subset of clones were sequenced at each round of panning and diversity was evaluated by analysis of CDR-H3 sequences. Multiple rounds of panning were carried out and aliquots of phage from each selection round were preserved for future use. Thousands of scFvs selected during panning were subjected to ELISA screening to assess antigen binding to identify lead candidates for antibody production. Initial screening according to program 1 led to the isolation of 214 clones of which 103 were selected based on an ELISA signal threshold and antigen binding specificity . Clones isolated according to program 1 included SEQ ID NOs: 230-381. Initial screening according to program 2 also resulted in a large number of clones, a portion of which were selected based on an ELISA signal threshold and antigen binding specificity analysis. These clones included SEQ ID NOs: 382-436.
Example 13. Identification of antibodies that activate the release of GDF-11 growth factor from the latent GPC
[00751] Production of a diverse panel of antibodies is carried out to identify antibodies that bind the prodomain of GDF-11 and activate the release of mature growth factor. Antibody generation is carried out according to the methods of Example 12 wherein recombinant GDF- 11 prodomain is used for solid-phase enrichment and biotinylated GDF- 11 prodomain is used for solution-phase enrichment. Antigen preparations are tested for aggregation levels to ensure that >95 are monomeric species. In ELISA analysis of enriched clones, binding to six antigens is assessed (GDF- 11 prodomain, proGDF-11, GDF- 11 growth factor, GDF- 8 prodomain, murine GDF-11 prodomain and proTGF-βΙ C4S). Clones selected based on ELISA analysis are sequenced and antibodies are developed according to the methods of Example 12.
[00752] A second program utilizes constructs with portions of GDF-11. These constructs, listed in Table 25, are alternated during subsequent rounds of enrichment to obtain phage particles that bind to the GDF-11 portions of the proteins.
Table 25. Recombinant antigens
Protein Amino acid sequence SEQ
ID NO
GDF-8/11 chimera MDMRVPAQLLGLLLLWFSGVLGDYKDDDDKHHHHHHL 1650 (GDF-8 1-64, GDF- EVLFQGPNENSEQKENVEKEGLCNACTWRQNTKSSRIEA
11 87-274) IKIQILS KLRLET APNIS KD VIRQLLPK APPLQQILDLHDFQ
GDALQPEDFLEEDEYHATTETVISMAQETDPAVQTDGSP
LCCHFHFSPKVMFTKVLKAQLWVYLRPVPRPATVYLQIL
RLKPLTGEGTAGGGGGGRRHIRIRSLKIELHSRSGHWQSI
DFKQVLHSWFRQPQSNWGIEINAFDPSGTDLAVTSLGPG
AEGLHPFMELRVLENTKRSRR
GDF-11/8 chimera AEGPAAAAAAAAAAAAAGVGGERSSRPAPSVAPEPDGC 1651 (GDF-11 1-86, PVCVWRQHSRELRLESIKSQILSKLRLKEAPNISREVVKQ
GDF-8 65-243) LLPKAPPLRELIDQYDVQRDDSSDGSLEDDDYHATTETIIT
MPTESDFLMQVDGKPKCCFFKFSSKIQYNKVVKAQLWIY
LRPVETPTTVFVQILRLIKPMKDGTRYTGIRSLKLDMNPG
TGIWQSIDVKTVLQNWLKQPESNLGIEIKALDENGHDLA
VTFPGPGEDGLNPFLEVKVTDTPKRSRR
GDF-11 /In Beta A AEGPAAAAAAAAAAAAAGVGGERSSRPAPSVAPEPDGC 1652 chimera (GDF-11 1- PVCVWRQHSRELRLESIKSQILSKLRLKEAPNISREVVKQ
86, Inh Beta A 65- LLPKAPPLNAIRKLHVGKVGENGYVEIEDDIGRRAEMNE
290) LMEQTSEIITFAESGTARKTLHFEISKEGSDLSVVERAEVW
LFLKVPKANRTRTKVTIRLFQQQKHPQGSLDTGEEAEEV
GLKGERSELLLSEKVVDARKSTWHVFPVSSSIQRLLDQG
KSSLDVRIACEQCQESGASLVLLGKKKKKEEEGEGKKKG
GGEGGAGADEEKEQSHRPFLMLQARQSEDHPHRRRRR
Inhibin Beta SPTPGSEGHSAAPDCPSCALAALPKDVPNSQPEMVEAVK 201 A/GDF-11 chimera KHILNMLHLKKRPDVTQPVPKAALLQQILDLHDFQGDAL
(Inh Beta A 1-64, QPEDFLEEDEYHATTETVISMAQETDPAVQTDGSPLCCHF
GDF- 11 87-274) HFSPKVMFTKVLKAQLWVYLRPVPRPATVYLQILRLKPL
TGEGTAGGGGGGRRHIRIRSLKIELHSRSGHWQSIDFKQV
LHSWFRQPQSNWGIEINAFDPSGTDLAVTSLGPGAEGLHP
FMELRVLENTKRSRR Example 14. Identification of antibodies that modulate the release of GDF-11 growth factor from the latent GPC
[00753] Production of a diverse panel of antibodies is carried out to identify antibodies that bind proGDF- 11 and modulate the release of mature growth factor. Antibody generation is carried out according to the methods of Example 12. Biotinylated proGDF-11 is used for both solid-phase enrichment and for solution-phase enrichment. Antigen preparations are tested for aggregation levels to ensure that >95% are monomeric species. Clones selected based on ELISA analysis are sequenced and antibodies are developed according to the methods of Example 12.
Example 15. Epitope binning
[00754] Selected clones are subjected to epitope binning by epitope-relatedness using surface plasmon resonance technology (or equivalent approach). Dissociation constants (k0ff) for each are determined and clones are selected for further characterization.
Example 16. Conversion to IgG format
[00755] Final candidates are expressed in IgG format. Variable domain cDNA is amplified and cloned into mammalian expression vectors. Resulting colonies are screened to ensure proper insertion. Plasmids are prepared by midi-scale DNA preparation for transfection of HEK293 cells at a 50-100 ml scale. Transfected cells are cultured and expressed IgGs are purified from the culture supernatant by affinity chromatography. For some IgG antibodies produced, Rvalues are determined by surface plasmon resonance. Cell-based functional assays are used to characterize purified bivalent antibodies.
Example 17. 293T CAGA-luciferase assay for GDF-8 activity
[00756] CAGA-luciferase assays are carried out to test antibodies that modulate GDF-8 and/or GDF-11 activity. 100 μΐ of 0.01% poly-L-lysine solution is added to each well of a 96- well plate. Plates are incubated for 10 min at room temperature before washed with water. 293T cells comprising transient or stable expression of pGL4 (Promega, Madison, WI) under the control of a control promoter or promoter comprising smadl/2 responsive CAGA sequences are then used to seed poly-L-lysine-coated wells (4 x 104 cell/well in complete growth medium). The next day, cells are washed with 150 μΐ/well of cell culture medium with 0.1% bovine serum albumin (BSA) before treatment with furin and/or mTLL2 processed proGDF-8 with or without test antibody. Cells are incubated at 37° for 6 hours before detection of luciferase expression using BRIGHT-GLO™ reagent (Promega, Madison, WI) according to manufacturer's instructions.
Example 18. Tolloid-dependent cleavage of GDF-8 and GDF-11
[00757] Samples of proGDF-8, latent GDF-8, latent GDF- 11 , or proGDF- 11 were incubated with pro-protein convertase (either Furin/PACE3 or PCSK5), Tolloid proteinase (either BMP-1 or mTLL2), a combination of both proteases or no proteases. The samples were incubated at 37C for 16hrs. Treated samples were analyzed by adding them to 293T or HepG2 cells carrying a CAGA luciferase response plasmid and incubated for six hours. GDF- 8 or GDF-11 activity was assessed by measuring CAGA-dependent luciferase activity in the cell lysates. As shown in FIG. 12, GDF-11 activity resulting from treatment with combined protease lysate (Tolloid proteinase and proprotein convertase) was nearly equal to treatment with recombinant GDF-11 (R&D Systems, Minneapolis, MN). The results of further assays are presented in FIG. 14 demonstrating growth factor release from proGDF-11 after treatment with various proteases in both cell lines. Surprisingly, treatment with mTLL-2 and Furin resulted in the highest level of activity in comparison to other protease treatment conditions.
Example 19. Skeletal muscle differentiation assay
[00758] GDF-8 inhibits skeletal muscle differentiation characterized by myogenin, myosin heavy chain and creatine kinase expression. Modulation of GDF-8 activity by candidate antibodies is tested using a skeletal muscle differentiation assay. 387550 Lonza cells (Lonza, Basel, Switzerland) are plated in 24- well plates at 4 x 104 cells/well. The next day, cell media is replaced with differentiation media [dulbecco's modified eagle medium (DMEM)/F12 with 2% horse serum.] Varying concentrations of GDF-8 are also included in differentiation media in the presence or absence of test antibodies. Cells are then allowed to differentiate for 2 days or 5 days.
[00759] After the 2 day period, differentiation status of each well is analyzed through analysis of myogenin expression levels. Cells from each treatment group are pooled and subjected to treatment using the Transcription Factor Buffer Set from BD Pharmingen (BD Biosciences, Franklin Lakes, New Jersey), product number 562574 according to
manufacturers instructions. After fixation and permeabilization, 5 μΐ of phycoerythrin (PE)- myogenin or 1.25 μΐ of PE-control are added to the cells and incubated at 4°C for 50 mins. Cells are then washed and resuspended in FACS buffer before analysis of cellular
fluorescence by flow cytometry. [00760] To assess the effect of candidate antibodies on differentiation over longer periods, expression levels of myosin heavy chain and/or expression levels of creatine kinase are assessed at day 5.
Example 20. GDF-11 processing by protease treatment
[00761] Partially proconvertase-cleaved proGDF-11 was run on an SDS PAGE gel under non-reducing and reducing conditions (see FIG. 13). Under non-reducing conditions, the protein bands consisted of the proGDF-11 dimer (-100 kD), proGDF-11 monomer (-60 kD), GDF-11 prodomain (-38 kD) and GDF-11 growth factor dimer (-20 kD). Under reducing conditions, the growth factor dimer was reduced to the monomer (-12 kD).
Example 21. GDF-11 antibody discovery
[00762] A large panel of diverse scFvs that bind specifically to human GDF-11 prodomain (or chimeric versions, e.g. those from Table 25) are precipitated using phage display technology. Accordingly, a naive phage library (approximately 1 x 1011 scFv) constructed in a similar manner as described by McCafferty et al (McCafferty, et al., 1990. Nature. 348:552- 4) is panned with specific GDF-11 antigens for three to five rounds of selection. Both solid and solution phase panning methods using biotinylated antigen are employed. Up to five thousand individual clones are selected and tested by ELISA for positive binding to GDF-11 prodomain antigens and up to five hundred scFv binders are sequenced. Specificity of binding to GDF11 but not to GDF8 or TGF-β is determined. Fifty candidates with good diversity of distinct VH and VL regions that bind specifically to the GDF-11 prodomain are selected for further development. These candidates are converted to full IgGs, expressed and purified from mammalian cell conditioned media. Additional ELISA-based screening to confirm species cross-reactivity, epitope binning [e.g. with Octet® system (ForteBio, Menlo Park, CA) using standard methods] and cell-based assay analysis are carried out. Candidates that functionally block or activate the release of GDF-11 in cell-based assays are further tested in appropriate preclinical models.
Example 22. Structural analysis
[00763] The proteins of Table 4, 5, 9, 10, 20, 21, 24 and 25 are subjected to structural analysis by X-ray crystallography, cryo- or negative stain single particle electron microscopy, NMR spectroscopy or hydrogen/deuterium exchange mass spectrometry methods. Among these are uncleaved proGDF-8, prodomain convertase-cleaved proGDF-8 (latent GDF-8) and Tolloid-cleaved (activated) GDF-8. Tertiary and quaternary structures are determined and the structural insights gained are used in the development of GDF-8 inhibitory and activating antibodies.
[00764] Further structural analysis is carried out to identify specific epitopes recognized by antibodies of the disclosure. Antibody-antigen complexes are produced and analyzed by X- ray crystallography. Structural insights gained are used in the development of additional GDF-8 inhibitory and activating antibodies.
Example 23. Conversion to IgG format
[00765] Clones selected according to Programs 1 and 2 were expressed in IgG format. Variable domain cDNA was amplified and cloned into mammalian expression vectors.
Resulting colonies were screened to ensure proper insertion. Plasmids were prepared by midi- scale DNA preparation for transfection of HEK293 cells at a 50-100 ml scale. Transfected cells were cultured and expressed IgGs were purified from the culture supernatant by affinity chromatography.
Example 24. Testing of Program 1 and 2 antibodies
[00766] Program 1 and 2 IgGs were tested for binding specificity. ELISA analysis was carried out to assess antibody binding to antigens used in panning (proGDF-8 and latent proGDF-8). Binding to proGDF-11 was also assessed as well as binding to ICAM control antigen. 96-well ELISA assay plates were prepared as described in Example 11 and primary antibodies being tested were added to each well and allowed to incubate at room temperature for 1 hour or more. Unbound antibody was removed by washing three times with wash buffer. Labeled secondary antibodies were then incubated in each well for 30 minutes at room temperature. Unbound secondary antibodies were removed by washing three times with wash buffer. Finally, relative light units (RLUs) were measured for each condition.
Antibodies were screened for affinity by selecting those which yielded readings greater than 3 standard deviations above control readings. These are indicated with a "+" for each antigen in the following Table.
Table 26. Antibody binding
Antibody proGDF-8 latent proGDF-8
BP15a-l + -
BPla-1 + -
BPlb-1 + -
BP2a-l + -
BP16b-4 + + BP16b-3 + +
BP16b-l + +
BP16b-2 + +
BP18b-l + +
BP18b-2 + +
BP18b-4 + +
BP18b-5 + +
BP18b-6 + +
BP18b-7 + +
BP20b-l + +
BP16a-l + +
BP18a-l + +
BP18a-2 + +
BP19b-2 + +
BP17b-l + +
BP19a-3 + +
BP18b-3 + +
BP19a-l + +
BP19a-2 + +
BP19b-l + +
BP19b-3 + +
BP22b-l + +
BP3a-2 + +
BP3a-3 + +
BP12b-l + +
BP5a-2 + +
BP5a-l + +
BP5a-4 + +
BP5a-5 + +
BP5b-6 + +
BP8b-l + +
BP5b-l + +
BP5b-2 + +
BP5b-3 + +
BP5b-5 + +
BP5b-8 + +
BPl la-1 + +
BP4a-l + +
BP5a-3 + +
BP3a-l + +
BP6a-21 + +
BP6b-4 + +
BPlOb-1 + +
BP5b-9 + + BP5b-4 + +
BP7b-l + +
BP6b-9 + +
BP9b-l + +
BP21b-l + +
BP6a-l l + +
BP6a-26 + +
BP5b-7 + +
BP6a-l + +
BP6a-7 + +
BP6a-2 + +
BP6a-3 + +
BP6a-4 + +
BP6a-8 + +
BP6a-5 + +
BP6a-9 + +
BP6a-10 + +
BP6a-6 + +
BP6a-12 + +
BP6a-13 + +
BP6a-14 + +
BP6a-15 + +
BP6a-16 + +
BP6a-17 + +
BP6a-18 + +
BP6a-19 + +
BP6a-20 + +
BP6a-22 + +
BP6a-23 + +
BP6a-24 + +
BP6a-25 + +
BP6a-27 + +
BP6b-3 + +
BP6b-5 + +
BP6b-6 + +
BP6b-l + +
BP6b-7 + +
BP6b-8 + +
BP6b-l l + +
BP6b-12 + +
BP6b-10 + +
BP6b-2 + +
BP13a-l + +
BP14a-l + + [00767] All antibodies tested significantly bound both proGDF-8 and latent proGDF-8 with the exception of BP2a-l, BPla-1, BPlb-1 and BP15a-l (which only significantly bound to proGDF-8).
[00768] To assess antibody binding to conformation-dependent epitopes, proGDF-8 and latent proGDF-8 were separated by gel electrophoresis under reducing or non-reducing conditions and subjected to Western blot analysis using IgG antibodies BP6a-7, BP5a-l, BP6a-5 and BP2a-l. Interestingly, BP6a-7, BP5a-l and BP2a-l were only able to bind to proteins run under non-reducing conditions, suggesting a conformation-dependent epitope. BP6a-5 was able to bind to both reduced and non-reduced forms of proGDF-8.
Example 25. In vivo testing of anti-GDF-8 antibodies
[00769] Five (5) groups of ten (10) female mice (n=10) with severe combined immune deficiency (SCID) were treated with myostatin antibodies and assessed for changes in muscle size and function. One week following an acclimation period, animals were divided into 5 groups (see Table 27). Blood was collected once via retro-orbital bleed for assessment of complete blood counts (CBC) and grip strength was measured. For this assessment animals were held against a bar which was connected to a force recorder until they grabbed the bar. Once the animal grabbed the bar they were moved away from the device until they dropped the bar. The force required to pull the animal from the bar was recorded in grams force (gf). Animals were treated by intraperitoneal (IP) injection once a week with a PBS vehicle control or one of the myostatin IgG antibodies BP6a-7 (comprising the VH of SEQ ID NO: 462 and the VL of SEQ ID NO: 1037), BP6a-5 (comprising the VH of SEQ ID NO: 472 and the VL of SEQ ID NO: 1047), BP5a-l (comprising the VH of SEQ ID NO: 448 and the VL of SEQ ID NO: 1023) or BP2a-l (comprising the VH of SEQ ID NO: 502 and the VL of SEQ ID NO: 1077) all at 25 mg/kg/week.
Table 27. Treatment of each group
Figure imgf000329_0001
[00770] Body weight and behavioral observations were recorded daily and animals were monitored for toxicity of treatments. Changes in overall body weight were recorded to indicate changes in overall muscle in response to antibody treatment. Six weeks following the initial injection, grip assessments were redone, the animals were sacrificed by C02 overdose and blood collected via cardiac puncture for a terminal CBC. The remaining blood was prepared for plasma using K2EDTA as an anti-coagulant. Tissues were isolated, weighed and collected in formalin, OCT or simply snap frozen. Terminal tissue weights were determined for the gastrocnemius, pectoralis, soleus, tricep, tibialis anterior, diaphragm, plantaris, quadriceps (Rectus femoris), heart, kidney, spleen, liver and inguinal white adipose tissue. Muscle immunohistochemistry was performed on the gastrocnemius.
[00771] The daily animal weight changes were recorded as an average percentage of weight change per group and compared across all treatments (see Table 28, SD is "standard deviation").
Table 28. Average daily % weight changes
Figure imgf000330_0001
30 5.09 1.83 8.50 1.26 12.64 1.32 7.82 1.51 4.60 1.42
31 4.79 1.94 8.64 1.10 12.00 1.36 7.41 1.53 4.75 1.13
32 5.15 1.89 8.90 0.94 12.98 1.23 8.54 1.49 4.63 1.20
33 3.94 2.04 10.14 1.21 14.12 1.28 9.39 1.56 7.04 1.27
34 4.97 2.10 9.11 1.10 13.37 1.22 9.21 1.62 6.46 1.20
35 5.00 2.34 10.41 1.04 13.86 1.28 9.84 1.77 6.53 1.03
36 5.78 1.93 10.80 1.24 14.85 1.32 10.05 1.73 5.98 1.08
37 6.56 2.02 10.85 1.29 15.01 1.36 10.25 1.64 6.40 1.10
38 7.02 2.08 11.92 1.45 14.60 1.15 11.19 1.55 7.02 1.33
39 7.29 2.10 11.82 1.45 15.45 1.35 9.70 1.70 7.10 1.58
40 6.58 2.07 11.97 1.35 16.43 1.42 11.89 1.37 7.44 1.24
41 6.26 2.03 12.36 1.15 15.97 1.27 11.99 1.49 7.96 1.41
42 7.74 1.96 12.42 1.09 14.58 1.28 10.33 1.53 7.44 1.43
[00772] Antibody treatment in Group 3, gave a surprisingly large increase in overall body weight. This increase was over 10% from vehicle control treated Group 1.
[00773] To determine if the increase in muscle size led to positive changes in muscle function, a grip strength test was done both pre and post antibody treatments. Grip strength was measured in grams force (gf). Each animal underwent five trials and the average of the three best grip strength scores was determined (see the following Table, SD is "standard deviation").
Table 29. Grip strength assay results (gf)
Figure imgf000331_0001
[00774] Group 1 mice had a nearly 8% decrease in grip strength over the course of the study while Groups 2 and 5 decreased slightly and Group 4 increased slightly. Group 3 mice had the largest increase in grip strength with an increase of 2.4%.
[00775] Terminal tissue weights are presented in the following Table. Standard error values are presented in parenthesis.
Table 30. Average terminal tissue weights
Tissue Group 1 Group 2 Group 3 Group 4 Group 5
Gastrocnemius 0.106 0.122 0.135 0.106 0.118
(0.007) (0.005) (0.005) (0.006) (0.007) Pectoralis 0.052 0.058 0.064 0.067 0.058
(0.005) (0.008) (0.007) (0.005) (0.004)
Soleus 0.014 0.008 0.011 0.016 0.017
(0.002) (0.001) (0.002) (0.003) (0.003)
Tricep 0.078 0.078 0.085 0.063 0.062
(0.008) (0.005) (0.004) (0.006) (0.006)
Tibialis Anterior 0.042 0.036 0.048 0.042 0.042
(0.005) (0.003) (0.005) (0.003) (0.004)
Diaphragm 0.056 0.076 0.084 0.076 0.072
(0.004) (0.006) (0.007) (0.005) (0.010)
Plantaris 0.152 0.156 0.170 0.167 0.170
(0.005) (0.005) (0.009) (0.009) (0.006)
Quadriceps 0.136 0.159 0.162 0.130 0.130
(0.009) (0.014) (0.012) (0.005) (0.009)
Heart 0.116 0.118 0.129 0.116 0.118
(0.005) (0.006) (0.005) (0.006) (0.004)
Kidney 0.257 0.234 0.224 0.233 0.234
(0.013) (0.011) (0.021) (0.019) (0.020)
Spleen 0.036 0.034 0.032 0.031 0.043
(0.002) (0.002) (0.002) (0.003) (0.005)
Liver 0.883 0.833 0.848 0.885 0.864
(0.021) (0.018) (0.025) (0.018) (0.019)
Inguinal White 0.084 0.067 0.097 0.067 0.099 Adipose Tissue (0.016) (0.009) (0.012) (0.011) (0.016)
[00776] Group 3 animals had the highest muscle tissue weights in 6 out of the 9 muscle tissues tested with muscle tissue weights larger than controls in 8 out of the 9 muscle tissues tested.
Example 26. Antibody activity across species
[00777] CAGA-luciferase assays were carried out to test modulation of human or mouse GDF-8 or GDF-11 activity by IgG antibodies BP6a-7 (comprising the VH of SEQ ID NO: 462 and the VL of SEQ ID NO: 1037), BP6a-5 (comprising the VH of SEQ ID NO: 472 and the VL of SEQ ID NO: 1047), BP5a-l (comprising the VH of SEQ ID NO: 448 and the VL of SEQ ID NO: 1023) and BP2a-l (comprising the VH of SEQ ID NO: 502 and the VL of SEQ ID NO: 1077). 100 μΐ of 0.01% poly-L-lysine solution was added to each well of a 96- well plate. Plates were incubated for 10 min at room temperature before washing with water. The plates were dried overnight. The next day, cells expressing pGL4 (Promega, Madison, WI) under the control of a promoter comprising smadl/2 responsive CAGA sequences were then used to seed poly-L-lysine-coated wells (4 x 104 cell/well in complete growth medium) and the proteolytic reaction was set up. Human proGDF-8, murine proGDF-8 or human proGDF-11 with or without test antibody were incubated at 37°C for 30 min. Then the proteins with or without test antibody were treated with activating enzymes furin and mTLL2 (or without mTLL2, as a negative control) at 37°C for -16 hours. The next day, cells were washed with 150 μΐ/well of cell culture medium with 0.1% bovine serum albumin (BSA). lOOx of the proteolytic reaction was added to the cellsand incubated at 37 °C for 6 hours before detection of luciferase expression using BRIGHT-GLOTM reagent (Promega, Madison, WI) according to manufacturer's instructions. Results were used to calculate the % inhibition for each condition (see the following Table, % inhibition = 100 x ((Reading with test antibody - reading without mTLL2)/(reading without antibody - reading without mTLL2)), standard deviation values are shown in parenthesis).
Table 31. % Inhibition of growth factor activity
Figure imgf000333_0001
[00778] [00641] All antibodies demonstrated the ability to inhibit human and murine GDF-8 activity. Inhibitory activity was not significant for human GDF-11.
Example 27. Antibody inhibitory activity
[00779] Antibodies were assessed for inhibition of GDF-8 activity using CAGA promoter assays in 293T cells or with HepG2 cells. 100 μΐ of 0.01% poly-L-lysine solution was added to each well of a 96- well plate. Plates were incubated for 10 min at room temperature before washing with water. The plates were dried overnight. The next day, cells expressing pGL4 (Promega, Madison, WI) under the control of a promoter comprising smadl/2 responsive CAGA sequences were then used to seed poly-L-lysine-coated wells (4 x 104 cell/well in complete growth medium) and the proteolytic reaction was set up. Human proGDF-8 or latent proGDF-8 with or without test antibody were incubated at 37°C for 30 min. Then the proteins with or without test antibody were treated with activating enzymes furin and mTLL2 (or without mTLL2, as a negative control) at 37°C for -16 hours. The next day, cells were washed with 150 μΐ/well of cell culture medium with 0.1% bovine serum albumin (BSA). lOOx of the proteolytic reaction was added to the cellsand incubated at 37 °C for 6 hours before detection of luciferase expression using BRIGHT-GLOTM reagent (Promega, Madison, WI) according to manufacturer's instructions. Results were used to calculate the % inhibition for each condition (% inhibition = 100 x ((Reading with test antibody - reading without mTLL2)/(reading without antibody/reading without mTLL2)). IgG antibodies BP6b- 6 (comprising the VH of SEQ ID NO: 581 and the VL of SEQ ID NO: 1153), BP19b-l (comprising the VH of SEQ ID NO: 442 and the VL of SEQ ID NO: 1017), BP6b-3
(comprising the VH of SEQ ID NO: 464 and the VL of SEQ ID NO: 1039), BP18b-4 (comprising the VH of SEQ ID NO: 593 and the VL of SEQ ID NO: 1166), BP7b-l
(comprising the VH of SEQ ID NO: 598 and the VL of SEQ ID NO: 1171), BP6b-7
(comprising the VH of SEQ ID NO: 626 and the VL of SEQ ID NO: 1199) and BPlOb-1 (comprising the VH of SEQ ID NO: 645 and the VL of SEQ ID NO: 1218) demonstrated inhibitory activity (see the following Table).
Table 32. % inhibition of growh factor activity
Figure imgf000334_0001
[00780] Antibody BP18b-4 had the highest percent inhibition in both cell types and with both proGDF-8 and latent proGDF-8. Interestingly, antibody BP19b-l only inhibited latent proGDF-8.
Example 28. "Primed" complexes
[00781] A "primed" complex is predicted to exist for both GDF-8 GPCs and GDF- 11 GPCs. Primed complexes are GPCs that adopt active conformations after cleavage at both the furin cleavage site and the BMP-l/Tolloid cleavage site. A schematic demonstrating formation of primed complexes is presented in FIG. 15. Activity may be the result of a conformational shift that relieves steric interference by the alpha 1 helical region. GDF-8 GPCs and GDF-11 GPCs are expressed and subjected to enzymatic cleavage to cleave both furin and BMP-l/Tolloid cleavage sites. Resulting primed complexes are tested for activity by CAGA assay and compared to free GDF-8 growth factors or GDF-11 growth factors; GDF-8 prodomain complexes or GDF-11 prodomain complexes (e.g., proGDF-8 or proGDF- 11, prior to furin or BMP-l/Tolloid cleavage); and latent GDF-8 complexes or latent GDF-11 complexes. GDF-8 primed complexes demonstrate signaling activity that is nearly equivalent with that of free GDF-8 growth factor. GDF-11 primed complexes demonstrate signaling activity that is nearly equivalent with that of GDF- 11 growth factor.
Example 29. Antibodies with selected binding profiles
[00782] Antibodies designed to bind human proMyo statin were tested for binding to human proMyostatin (SEQ ID NO: 5); murine proMyostatin (SEQ ID NO: 88); human latent Myostatin (SEQ ID NO: 5); human GDF-8 prodomain (SEQ ID NO: 38); human GDF-11 ARM 8 prodomain (SEQ ID NO: 181); human proGDFHARM8 (SEQ ID NO: 179); human mature GDF-8 (SEQ ID NO: 42); human proGDFl l (SEQ ID NO: 4); murine latent myostatin (SEQ ID NO: 88); and human latent proGDFl l ARM 8 (SEQ ID NO: 179).
Binding of the various antibody clones, shown in Table 33, to the antigens was performed using an ELISA as described below. Table 33 shows the selected binding profiles for the various antibody clones. Initially, clones BPlOb-1 and BP12b-l were selected as binders to human proMyostatin. However, subsequent binding tests showed these antibodies bound human latent Myostatin and not human pro Myostatin.
Table 33. Selected Myostatin binding profiles mat lat pro Pro Lat GDF8 GDFll pro lat
Antibody proGDFl proGDFll
Myo Myo Myo Prod ARM8 GDFll Myo clone 1 ARM8 GDF8 ARM8
(h) (m) (h) (h) prod (h) (m)
(h) (h) (h) (h)
BPla-1 + + - - - - -
BPlb-1 + + - - - - - - - -
BP2a-l + + - - - + -
BP3a-l + + + - - - -
BP3a-2 + + + - - - -
BP3a-3 + + + - - - -
BP4a-l + + + + - - -
BP5a-l + + + - - + -
BP5b-l + + + - - + - - + +
BP5a-2 + + + - - + -
BP5a-3 + + + - - + -
BP5a-4 + + + - - + -
BP5a-5 + + + - - + -
BP5b-2 + + + - - + - - + +
BP5b-3 + + + - - + - - + +
BP5b-4 + + + - - + - - + + BP5b-5 + + + - - + - - + +
BP5b-6 + + + - - + - - + +
BP5b-7 + + + - - + - - + +
BP5b-8 + + + - - + - - + +
BP5b-9 + + + - - + - - + +
BP6a-l + + + + + + -
BP6a-2 + + + + + + -
BP6a-3 + + + + + + -
BP6a-4 + + + + + + -
BP6a-5 + + + + + + -
BP6a-6 + + + + + + -
BP6b-l + + + + + + - - + +
BP6b-2 + + + + + + - - + +
BP6a-7 + + + + + + -
BP6a-8 + + + + + + -
BP6a-9 + + + + + + -
BP6a-10 + + + + + + -
BP6a-l l + + + + + + -
BP6a-12 + + + + + + -
BP6a-13 + + + + + + -
BP6a-14 + + + + + + -
BP6a-15 + + + + + + -
BP6a-16 + + + + + + -
BP6a-17 + + + + + + -
BP6a-18 + + + + + + -
BP6a-19 + + + + + + -
BP6a-20 + + + + + + -
BP6a-21 + + + + + + -
BP6a-22 + + + + + + -
BP6a-23 + + + + + + -
BP6a-24 + + + + + + -
BP6a-25 + + + + + + -
BP6a-26 + + + + + + -
BP6a-27 + + + + + + -
BP6b-3 + + + + + + - - + +
BP6b-4 + + + + + + - - + +
BP6b-5 + + + + + + - - + +
BP6b-6 + + + + + + - - + +
BP6b-7 + + + + + + - - + +
BP6b-8 + + + + + + - - + +
BP6b-9 + + + + + + - - + +
BP6b-10 + + + + + + - - + +
BP6b-l l + + + + + + - - + +
BP6b-12 + + + + + + - - + + BP7b-l + + + +/- +/- + - - + +
BP8b-l + + + - - +/- - - + +
BP9b-l + + + +/- - +/- - - + +
BPlOb-1 - - + - - - - - + +
BPl la-1 + + + - - + +
BP12b-l - - + - - - + - + -
BP13a-l + +/- + - - + -
BP14a-l + +/- + +/- - + -
BP15a-l + - - + - - -
BP16b-l + - + - - - - - - -
BP16b-2 + - + - - - - - - -
BP16b-3 + - + - - - - - - -
BP16b-4 + - + - - - - - - -
BP16a-l + - + - - - -
BP17b-l + - + - - - - - - +/-
BP18b-l + - + - - + - - - +
BP18b-2 + - + - - + - - - +
BP18b-3 + - + - - + - - - +
BP18b-4 + - + - - + - - - +
BP18b-5 + - + - - + - - - +
BP18b-6 + - + - - + - - - +
BP18b-7 + - + - - + - - - +
BP18a-l + - + - - + -
BP18a-2 + - + - - + -
BP19b-l + - + + + + - - - +
BP19b-2 + - + + + + - - - +
BP19b-3 + - + + + + - - - +
BP19a-l + - + + + + -
BP19a-2 + - + + + + -
BP19a-3 + - + + + + -
BP20b-l + - + - - +/- - - - +/-
BP21b-l + + + - - + - + + +
BP22b-l + + + - - - - + + +
Table 33 shows the binding profile of antibody clones to the following antigens: proMyo(h) = human proMyostatin (SEQ ID NO: 5); ProMyo(m) = murine proMyostatin (SEQ ID NO: 88);
LatMyo(h) = human latent Myostatin (SEQ ID NO: 5); GDF8Prod(h) = human GDF-8 prodomain (SEQ ID NO: 38); GDF1 lARM8prod(h) = human GDF-11 ARM 8 prodomain (SEQ ID NO: 181); proGDFl lARM8(h) = human proGDFl l ARM 8 (SEQ ID NO: 179); matGDF8(h) = human mature GDF-8 (SEQ ID NO: 42); proGDFl 1(h) = human proGDFl 1 (SEQ ID NO: 4); latMyo(m) = murine latent myostatin (SEQ ID NO: 88);
latproGDFl lARM8(h) = human latent proGDFl 1 ARM8 (SEQ ID NO: 179). As described elsewhere herein, binding of an antibody clone to an antigen is indicated by a "+". A "-" indicates that the antibody does not bind the antigen. A "+/-" indicates that the antibody is at or near a threshold of binding the antigen as determined by the assay parameters. The code "BP[Number] [Letter]" refers to particular binding profiles indicated in the table.
ELISA Screening Protocol
[00783] The purpose of this protocol is to allow for a method to screen Myostatin Program antibodies for potential binding to biotinylated antigens. This protocol makes use of the NeutrAvidin Coating of 96-Well Plates SOP for NeutrAvidin coated and BSA blocked plates. The plates should be designed in such a way that each sample and control is performed in triplicate and each plate contains an anti-his coating control. The coating control measures the degree to which each biotinylated antigen is captured by the plate and allows for ease of data analysis. Multiple plates may be required per experiment.
[00784] The materials used include: solid, white Solid white 96-well plates from the NeutrAvidin Coating of 96-Well Plates SOP, Biotinylated antigen, Maine Biotechnology Services Anti-His antibody MAB230P, Jackson ImmunoResearch Laboratories Peroxidase Affinipure Goat a-human FCy Fragment- Specific (Catalogue number 109-035-008), Jackson ImmunoResearch Laboratories Affinipure Goat a-mouse FCy Fragment Specific (Catalogue number 115-035-008), and SuperSignal ELISA Femto Substrate (Pierce Biotech catalog number 37075).
[00785] The following equipment was used: multi-channel pipette, P200 tips, tabletop ultracentrifuge, 1.5 mL centrifuge tubes, 0.5 mL centrifuge tubes, P1000, P1000 tips, P200, P10, P10 tips, 15 mL Falcon tubes, 50 mL Falcon tubes, Biotek ELx 405 Select CW plate washer, multidrop, and Biotek Synergy HI plate reader.
[00786] Wash Buffer: TBS (Tris-Buffered Saline; 50 mM Tris-Cl, 150 mM NaCl, pH 7.6) with 0.05% Tween-20. For manual (hand) wash add 0.1% BSA as BSA is sticky and should not be used with the automated plate washer system.
[00787] Sample Buffer: TBS (Tris-Buffered Saline; 50 mM Tris-Cl, 150 mM NaCl, pH 7.6) with 0.05% Tween-20 and 0.1% BSA.
[00788] ELISA 3X protocol: Wash protocol on the Biotek ELx 405 Select CW plate washer. Washes with 200μL· of wash buffer. Repeats the wash two additional times.
Specifications: 3 cycles. No shaking. Dispenses 200 μί ρεΓ well. Dispense flow rate setting 7 (range 1 - 10). Dispense height 15.24 mm. Horizontal x dispense position 0 mm. Horizontal y dispense position 0 mm. Aspirate height 3.048 mm. horizontal x aspirate position 1.372 mm. horizontal aspirate y position 0.452 mm. Aspiration rate 3.4 mm/second. Aspiration delay 0 milliseconds. Crosswire aspiration on final wash. Crosswire height 3.048 mm.
Crosswire horizontal x position: -1.829 mm. Crosswire horizontal y position: -0.457 mm.
[00789] Remove a pre-coated and pre-blocked 96- well plate from 4°C. If a 96- well plate is not already prepared, prepare one with the NeutrAvidin Coating of 96-Well Plates protocol (see below). Remove the lxPBS pH 1 BSA with 0.1% Tween-20 from the plate and forcefully hit the plate on a Styrofoam pad lined with paper towels. Dilute the biotinylated antigen in sample buffer. Optimal capture concentration should first be determined by titration for each individual protein. For human and murine promyostatin this concentration is 2 μg/mL. Add 100 μΐ per well. Wash plates using the plate washer with wash buffer using the ELISA 3X protocol. Dilute the antibody in sample buffer. Screening of antibodies is performed at 1 μg/mL. Prepare a-His coating control antibody (Maine Biotechnology Services catalogue number: MAB230P) at 1 μg/mL in sample buffer. Place 100 μΐ^ of diluted antibody on designated wells. Incubate at room temperature for 1 hour. Wash plates using the plate washer with wash buffer using the ELISA 3X protocol. Dilute human secondary antibody (Jackson ImmunoResearch Laboratories Peroxidase Affinipure Goat a-human FCy Fragment Specific. Catalogue number 109-035-008)
1:25,000 in sample buffer. For a-his wells, dilute mouse secondary antibody (Jackson ImmunoResearch Laboratories Affinipure Goat a-mouse FCy Fragment Specific. Catalogue number 115-035-008) 1:50,000 in sample buffer. Place 100 μΐ, of diluted antibody on designated wells. Incubate at room temperature for 1 hour. Wash plates using the plate washer with wash buffer using the ELISA 3X protocol. Prepare SuperSignal ELISA Femto Substrate (Pierce Biotech catalog number 37075) working solution by mixing 1 part luminol/enhancer solution with 1 part stable peroxide substrate. It is important to use separate pipettes in order to prevent cross-contamination and damaging of the substrate solution. Ten mL will be needed for one plate. Working solution is stable for up to 4 hour at room temperature. Place 100 μΐ^ SuperSignal ELISA Femto Substrate working solution per well. Wait one minute and read on the plate reader using the lum fiber method or by measuring with a luminometer at 425nm.
[00790] Assay output values are in relative light units (RLU) and data analysis can be performed in Microsoft Excel. All experiment samples should be performed in triplicate. Triplicate values should be averaged together. Error bars should be expressed as the standard error using the following equation: x = stdev / sqrt (n) where n is 3. Hits are determined by calculating the standard deviation across all samples and then setting a threshold of 3x that standard deviation. All samples above the 3x standard deviation threshold are hits.
NeutrAvidin Coating of96-Well Plates
[00791] The purpose of this protocol is to coat 96-well high binding plates with neutravidin for ELISA assays.
[00792] Materials used include the following: Costar 96-well assay plate (high binding), (manufacturer product numbers: 3922 (solid white), 3925 (solid black), 3361 (clear)), UltraPure Distilled Water (Invitrogen). Manufacturer product number 10977-015, lx Carbonate Buffer pH 9.4 made from BupH Carbonate - Bicarbonate Buffer Packs (Thermo Scientific, manufacturer product number 28382), NeutrAvidin Biotin-Binding Protein (salt- free), 10 mg (Pierce, manufacturer product number: 31000).
[00793] Equipment used includes the following: multi-channel pipette, P200 tips, tabletop ultracentrifuge, 1.5 mL centrifuge tubes, 0.5 mL centrifuge tubes, milligram range balance, P1000, P1000 tips, P200, P10, P10 tips, 15 mL Falcon tubes, nanodrop, and Ultrafree-MC-GV 0.22 μιη filter device (Millipore, manufacturer product number UFC30GVNB).
[00794] Wash buffer includes TBS (Tris-Buffered Saline; 50 mM Tris-Cl, 150 mM NaCl, pH 7.6) with 0.05% Tween-20. For manual (hand) wash add 0.1% BSA as BSA is sticky and should not be used with the automated plate washer system.
[00795] Measure an appropriate amount of lyophilized NeutrAvidin ( > 1 mg) and dilute into an appropriate amount of ultrapure water (1000 μϊ^ > χ > 100 μί). Do not exceed 10 mg/mL. Resuspend and dissolve carefully. Filter dissolved NeutrAvidin with a 0.22 μιη centrifuge filter device at 12,000 x g for 4 minutes and determine protein concentration via Nanodrop. The A280 of NeutrAvidin is 1.66 at 1 mg/mL. Dilute NeutrAvidin to 1 mg/mL in lx carbonate buffer pH 9.4 to make a 1 mg/mL stock solution. Do not exceed 1 mg/mL as NeutrAvidin is not directly soluble in buffer. Aliquot 1 mg/mL NeutrAvidin solution into 20 μL· aliquots. Snap freeze in liquid nitrogen and store at -80°C. Thaw appropriate numbers of NeutrAvidin aliquots by hand. Spin the 1 mg/mL NeutrAvidin stock solution at 16,100 x g for 10 minutes. For one plate, remove 5 μΐ^ from the top of the 1 mg/mL NeutrAvidin stock solution and dilute it into 10 mL of lx carbonate buffer pH 9.4. Mix by inverting the falcon tube. Using a multi-channel pipette, place 100 μΐ^ in each well of the Corning high binding 96-well assay plate. Incubate the 96-well plate overnight at 4°C. Wash the 96-well plate with 200 μΐ^ of wash buffer per well. Repeat this step two additional times. The plate should be washed for a total of three times. Block the plate with 200 μΐ^ per well of 1% BSA in PBS pH 7.4. Incubate the plate for 1 hour at 37°C or overnight at 4°C.
[00796] Antibodies were scored as specific binders "+" if their binding signal was 5 standard deviations above background. Antibodies were scored as not specific binders "-" if their binding signal was below 5 standard deviations above background. Antibodies scored with a "+/-" indicates that the antibody is at or near the threshold of binding the antigen as determined by the assay parameters.
Example 30. Binding primed Myostatin complexes and determining inhibitory activity
[00797] Antibody clones BP5a-l, BP6a-7, BP6a-5, BP6b-7, BPl la-1, BP12b-l, BP16a-l were tested in vitro using a CAGA promoter-based reporter assay as described herein. The CAGA promoter-based reporter assays may be used to test antibodies that modulate SMAD- dependent gene expression as reported by Thies et al (Thies, R.S. et al., Growth Factors. 2001. 18:251-9, the contents of which are herein incorporated by reference in their entirety). Here, a CAGA reporter assays was used to assess primed Myostatin growth factor activity in the presence of an various antibody clones. Antibody clone BP6a-5 was tested for binding to human primed Myostatin using an ELISA assay.
Table 34. Binding and inhibiting primed Myostatin complexes
Figure imgf000341_0001
Table 34 shows the ability of various antibody clones to inhibit human primed Myostatin signaling and the ability of various antibody clones to bind human primed Myostatin.
Example 31. Binding affinity to proMyostatin and latent Myostatin [00798] The binding affinity of various antibody clones to human proMyo statin or human latent Myostatin was determined using two independent menthods. In one method, the dissociation constant (Kd) was determined using the Octet® (ForteBio, Menlo Park, CA) instrument. In another method, the EC50 was determined using an ELISA (Table 35).
Table 35. Binding affinity to proMyostatin and latent Myostatin
Kd by Octet ELISA EC50 (nM)
latent mature latent
Antibody proMyostatin proMyostatin proMyostatin
Myostatin Myostatin Myostatin Clone (human) (human) (human)
(human) (human) (human)
BPlb-1 202.5 0
BPla-1 8.023E-09 not detect. 23.96
BP2a-l 5.42E-10 not detect. 4.75
BP3a-2 2.662E-09 1.282E-08 6.7
BP3a-3 6.643E-11 9.507E-11 14.58
BP3a-l 5.248E-10 2.771E-10 5.786
BP4a-l 1773
BP5b-3 81.39
BP5a-l 36.4
BP5b-4 254.9
BP5b-6 1403
BP5b-5 834
BP5a-4 1.15E-08 1.12E-08 not detect. 1611
BP5b-l 19.15
BP5a-3 605
BP5b-2 35.06
BP5a-2 1.06E-09 8.308E-10 2.835
BP5b-7 978742 3942000
BP5a-5 3440
BP5b-8 55.64
BP5b-9 83.47
BP6b-4 34.32 0
BP6a-8 2.2
BP6a-23 372
BP6a-l l 15.4
BP6a-14 19.5
BP6a-27 not detect.
BP6a-13 19.17
BP6a-19 130.9
BP6a-6 12.62
BP6a-20 193.1 BP6a-24 631
BP6a-22 342.4
BP6a-4 1.417
BP6a-7 1.13
BP6a-16 20.36
BP6b-3 0.3268 103.3
BP6a-2 1.3
BP6a-3 1.4
BP6b-l 4.18
BP6a-5 1.296E-08 2.392
BP6a-10 9.4
BP6a-l 0.2993
BP6a-26 987.5
BP6b-5 35.94
BP6a-12 18.77
BP6a-9 5.279
BP6a-18 78.9
BP6a-25 916.5
BP6b-6 37.95
BP6a-15 19.7
BP6b-2 8.07
BP6b-l l
BP6b-10 60.85
BP6a-21 242.9
BP6b-9 55.48
BP6b-7 0.29
BP6b-8 3.88
BP6b-12
BP6a-17 20.37
BP7b-l 38.43
BP8b-l 170.2
BP9b-l not detect.
BPlOb-1 5.174E-09 4.676E-09 19631 722.4
BPl la-1 1.71E-09 4.48E-09 7.56E-09 15.9
BP12b-l not detect. 1.83E-09 15.41
BP13a-l 315.7
BP14a-l 8.625
BP15a-l not detect.
BP16b-l 5.66
BP16b-2 not detect.
BP16b-3 48.17
BP16b-4 4.24 BP16a-l 2.8
BP17b-l 22.31
BP18b-l 19.38 222916
BP18a-l 22.25
BP18a-2 29.26
BP18b-2 6091 21844
BP18b-3 10559 19279
BP18b-4 0.075
BP18b-5 0.24
BP18b-6 0.61
BP18b-7
BP19b-l 0.18
BP19b-2 2.87 2074
BP19a-l 0.24
BP19a-2 75.38
BP19b-3 160
BP19a-3 15.2
BP20b-l not detect.
BP21b-l 1.57 1.717
BP22b-l 28.10
Table 35 shows the binding affinity of various antibody clones to human proMyo statin or human latent Myostatin using two independent methods, a Kd measurement using Octet® (ForteBio, Menlo Park, CA) and an EC50 using an ELISA.
Example 32. Inhibition of proMyostatin and latent Myostatin in vitro
[00799] The ability of antibody clones to inhibit proMyostatin or latent Myostatin was tested in vitro using a CAGA promoter-based reporter assay as described herein. The CAGA promoter-based reporter assays may be used to test antibodies that modulate SMAD- dependent gene expression as reported by Thies et al (Thies, R.S. et al., Growth Factors.
2001. 18:251-9, the contents of which are herein incorporated by reference in their entirety). Here, a CAGA reporter assays was used to assess proMyostatin and latent Myostatin growth factor activity in the presence of an various antibody clones. According to such methods, proMyostatin, or latent Myostatin were cultured with reporter cells (either 293T or HepG2 reporter cells) comprising the CAGA promoter linked to a reporter gene (e.g. luciferase) in the presence of the antibody shown in Tables 36 and 37.
Table 36. Inhibition of proMyostatin in vitro 293T HepG2 293T HepG2 HepG2
Antibody
% Inhibition % Inhibition % Inhibition % Inhibition % Inhibition Clone
Exp 1 Ex l Exp 2 Exp 2 Murine GDF8
BP6a-8 52 35 38 42 39
BP13a-l 55 7 44 44 19
BP6a-23 -5 8
BP5a-l 55 13 47 39 36
BP6a-l l 12 -17
BP18a-l 7 24 14 9 11
BPl la-1 63 26 1 3 43
BP6a-14 61 22 10 12 55
BP6a-27 28 21 -11 7 8
BP6a-13 22 14
BP4a-l -14 31 10 6 36
BP6a-19 28 5
BPla-1 45 22 8 20 37
BP6a-6 46 9
BP6a-20 7 -10
BP3a-2 46 -2
BP6a-24 -6 -19
BP6a-22 -6 -19
BP6a-4 11 5
BP19a-l 83 93 89 90 25
BP6a-7 42 61 69 66 59
BP6a-16 61 26 17 18 12
BP18a-2 6 33 31 21 13
BP19a-2 55 9
BP6a-2 -16 -13 23 10 14
BP6a-3 15 1
BP6a-5 70 48 68 58 42
BP6a-10 24 7
BP6a-l 6 -15
BP15a-l -12 1
BP6a-26 55 21 2 20 74
BP3a-3 -30 -9
BP6a-12 26 4
BP5a-4 3 3
BP5a-3 12 17
BP14a-l 3 7
BP3a-l -34 3
BP6a-9 -44 -22
BP6a-18 49 6 27 20 21 BP6a-25 -33 18
BP5a-2 54 -9 62 51 98
BP6a-15 50 28 20 6 4
BP2a-l 35 47 55 43 23
BP6a-21 21 6
BP5a-5 46 16
BP16a-l 50 48 42 38 26
BP6a-17 0 18
BP19a-3 19 6
Table 36 shows the % inhibition of human proMyostatin and murine proMyostatin activity in the presence of various antibody clones using two independent reporter cells (293T and HepG2). Note: Experiment 1 = July-2014, Experiment 2 = Aug-2014
Table 37. Inhibition of proMyostatin and latent Myostatin in vitro
Antibody 293T 293T latent HepG2 HepG2 latent Clone proMyostatin Myostatin proMyostatin Myostatin
(human) (human) (human) (human)
BP18b-l 9 2 11 9
BP19b-l -1 53 3 51
BP6b-4 20 -1 8 6
BP5b-3 29 19 32 23
BPlb-1 19 58 0 25
BP5b-4 -34 -14 -33 -20
BP5b-6 -3 2 -14 -3
BP19b-2 14 1 25 8
BP5b-5 6 -7 -3 3
BP6b-3 42 1 51 -4
BP18b-2 8 6 12 6
BP21b-l 35 13 36 15
BP6b-l 6 -15 -21 -13
BP6b-5 8 0 -16 -9
BP5b-l 6 3 -16 4
BP18b-3 30 4 21 8
BP8b-l 23 -12 -5 -2
BP5b-2 17 -25 -1 -11
BP6b-6 44 51 53 56 BP6b-2 28 -2 15 0
BP6b-l l 21 35 10 1
BP18b-4 89 92 85 97
BP6b-10 7 26 -1 46
BP16b-l -6 11 -13 20
BP16b-2 -6 24 -11 11
BP18b-5 18 13 32 31
BP7b-l 82 67 89 92
BP5b-7 25 21 23 4
BP18b-6 1 0 13 16
BP20b-l 10 -1 -2 8
BP19b-3 5 -10 20 11
BP18b-7 32 31 15 24
BP17b-l 3 -8 23 10
BP16b-3 2 2 17 19
BP5b-8 11 -3 5 0
BP16b-4 4 -15 23 -2
BP9b-l -10 -16 10 0
BP12b-l -4 -36 6 -43
BPlOb-1 51 23 59 17
BP6b-9 4 -7 15 -2
BP6b-7 74 51 78 69
BP6b-8 18 -3 27 -6
BP5b-9 -8 -2 6 -26
BP22b-l 11 1 13 2
BP6b-12 5 9 11 33
Table 37 shows the % inhibition of human proMyo statin and human latent Myostatin activity in the presence of various antibody clones using two independent reporter cells (293T and HepG2).
Example 33. pH sensitive dissociation from proMyostatin
[00800] The pH sensitivity of antibody clones, including clones BPla-1, BP2a-l, BP3a-2, BP5a-l, BP5b-4, BP5b-l, BP5b-2, BP5a-2, BP6a-l l, BP6a-6, BP6a-4, BP6a-7, BP6a-2, BP6a-3, BP6a-5, BP6a-l, BP6b-9, BPl la-1, BP14a-l, BP16b-4, BP18b-2, BP18b-7 and BP22b-l was determined usig Bio-Layer Interferometry (BLI). Data relating to the pH sensitivity of antibody clones provided herein is shown in FIGs. 16-25 and Table 38.
[00801] Bio-Layer Interferometry (BLI) is a label-free technology for measuring biomolecular interactions. It is an optical analytical technique that analyzes the interference pattern of white light reflected from two surfaces: a layer of immobilized protein on the biosensor tip, and an internal reference layer. Any change in the number of molecules bound to the biosensor tip causes a shift in the interference pattern that can be measured in real-time.
[00802] The binding between a ligand immobilized on the biosensor tip surface and an analyte in solution produces an increase in optical thickness at the biosensor tip, which results in a wavelength shift, Δλ, which is a direct measure of the change in thickness of the biological layer. Interactions are measured in real time, providing the ability to monitor binding specificity, rates of association and dissociation, or concentration, with precision and accuracy.
[00803] Only molecules binding to or dissociating from the biosensor can shift the interference pattern and generate a response profile on the Octet® System. Unbound molecules, changes in the refractive index of the surrounding medium, or changes in flow rate do not affect the interference pattern. This is a unique characteristic of BLI and extends its capability to perform in crude samples used in applications for protein:protein binding, quantitation, affinity, and kinetics.
Table 38. pH sensitive dissociation from proMyostatin
Figure imgf000348_0001
Figure imgf000349_0001
Table 38 shows the ratio of the Kd at pH 5 to the Kd at pH7, indicating the antibodies that are sensitive to pH.
[00804] While several embodiments of the present disclosure have been described and illustrated herein, those of ordinary skill in the art will readily envision a variety of other means and/or structures for performing the functions and/or obtaining the results and/or one or more of the advantages described herein, and each of such variations and/or modifications is deemed to be within the scope of the present disclosure. More generally, those skilled in the art will readily appreciate that all parameters, dimensions, materials, and configurations described herein are meant to be exemplary and that the actual parameters, dimensions, materials, and/or configurations will depend upon the specific application or applications for which the teachings of the present disclosure is/are used. Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, many equivalents to the specific embodiments of the disclosure described herein. It is, therefore, to be understood that the foregoing embodiments are presented by way of example only and that, within the scope of the appended claims and equivalents thereto, the disclosure may be practiced otherwise than as specifically described and claimed. The present disclosure is directed to each individual feature, system, article, material, and/or method described herein. In addition, any combination of two or more such features, systems, articles, materials, and/or methods, if such features, systems, articles, materials, and/or methods are not mutually inconsistent, is included within the scope of the present disclosure.
[00805] The indefinite articles "a" and "an," as used herein in the specification and in the claims, unless clearly indicated to the contrary, should be understood to mean "at least one."
[00806] The phrase "and/or," as used herein in the specification and in the claims, should be understood to mean "either or both" of the elements so conjoined, i.e., elements that are conjunctively present in some cases and disjunctively present in other cases. Other elements may optionally be present other than the elements specifically identified by the "and/or" clause, whether related or unrelated to those elements specifically identified unless clearly indicated to the contrary. Thus, as a non-limiting example, a reference to "A and/or B," when used in conjunction with open-ended language such as "comprising" can refer, in one embodiment, to A without B (optionally including elements other than B); in another embodiment, to B without A (optionally including elements other than A); in yet another embodiment, to both A and B (optionally including other elements); etc.
[00807] As used herein in the specification and in the claims, "or" should be understood to have the same meaning as "and/or" as defined above. For example, when separating items in a list, "or" or "and/or" shall be interpreted as being inclusive, i.e., the inclusion of at least one, but also including more than one, of a number or list of elements, and, optionally, additional unlisted items. Only terms clearly indicated to the contrary, such as "only one of or "exactly one of," or, when used in the claims, "consisting of," will refer to the inclusion of exactly one element of a number or list of elements. In general, the term "or" as used herein shall only be interpreted as indicating exclusive alternatives (i.e. "one or the other but not both") when preceded by terms of exclusivity, such as "either," "one of," "only one of," or "exactly one of." "Consisting essentially of," when used in the claims, shall have its ordinary meaning as used in the field of patent law.
[00808] As used herein in the specification and in the claims, the phrase "at least one," in reference to a list of one or more elements, should be understood to mean at least one element selected from any one or more of the elements in the list of elements, but not necessarily including at least one of each and every element specifically listed within the list of elements and not excluding any combinations of elements in the list of elements. This definition also allows that elements may optionally be present other than the elements specifically identified within the list of elements to which the phrase "at least one" refers, whether related or unrelated to those elements specifically identified. Thus, as a non-limiting example, "at least one of A and B" (or, equivalently, "at least one of A or B," or, equivalently "at least one of A and/or B") can refer, in one embodiment, to at least one, optionally including more than one, A, with no B present (and optionally including elements other than B); in another
embodiment, to at least one, optionally including more than one, B, with no A present (and optionally including elements other than A); in yet another embodiment, to at least one, optionally including more than one, A, and at least one, optionally including more than one, B (and optionally including other elements); etc. [00809] In the claims, as well as in the specification above, all transitional phrases such as "comprising," "including," "carrying," "having," "containing," "involving," "holding," and the like are to be understood to be open-ended, i.e., to mean including but not limited to. Only the transitional phrases "consisting of and "consisting essentially of shall be closed or semi-closed transitional phrases, respectively, as set forth in the United States Patent Office Manual of Patent Examining Procedures, Section 2111.03.
[00810] Use of ordinal terms such as "first," "second," "third," etc., in the claims to modify a claim element does not by itself connote any priority, precedence, or order of one claim element over another or the temporal order in which acts of a method are performed, but are used merely as labels to distinguish one claim element having a certain name from another element having a same name (but for use of the ordinal term) to distinguish the claim elements.

Claims

What is claimed is:
1. An antibody that specifically binds human proMyostatin, human latent Myostatin, human GDF-8 prodomain, human GDF-11 ARM8 prodomain, and human proGDFl 1 ARM8, but does not specifically bind human mature GDF-8 having an amino acid sequence as set forth in SEQ ID NO: 42.
2. An antibody that specifically binds human proMyostatin, murine proMyostatin, human latent Myostatin, human GDF-8 prodomain, human GDF-11 ARM8 prodomain, and human proGDFl 1 ARM8, but does not specifically bind human mature GDF-8 having an amino acid sequence as set forth in SEQ ID NO: 42.
3. The antibody of claim 1 or 2, wherein the human proMyostatin has an amino acid sequence as set forth in SEQ ID NO: 5.
4. The antibody of claim 2 or 3, wherein the murine proMyostatin has an amino acid sequence as set forth in SEQ ID NO: 88.
5. The antibody of any one of claims 1 to 4, wherein the human latent Myostatin has an amino acid sequence as set forth in SEQ ID NO: 5.
6. The antibody of any one of claims 1 to 5, wherein the human GDF-8 prodomain has an amino acid sequence as set forth in SEQ ID NO: 38.
7. The antibody of any one of claims 1 to 6, wherein the human GDF-11 ARM8 prodomain has an amino acid sequence as set forth in SEQ ID NO: 181.
8. The antibody of any one of claims 1 to 7, wherein the human proGDFl 1 ARM8 has an amino acid sequence as set forth in SEQ ID NO: 179.
9. The antibody of any one of claims 1 to 8, wherein the antibody specifically binds human latent proGDFl 1 ARM8, but does not specifically bind human proGDFl 1 having an amino acid sequence as set forth in SEQ ID NO: 4.
10. The antibody of any one of claims 2 to 8, wherein the antibody specifically binds murine latent myostatin and human latent proGDFl 1 ARM8, but does not specifically bind human proGDFl 1 having an amino acid sequence as set forth in SEQ ID NO: 4.
11. The antibody of claim 10, wherein the murine latent myostatin has an amino acid sequence as set forth in SEQ ID NO: 88.
12. The antibody of any one of claims 9 to 11, wherein the human latent proGDFl 1 ARM8 has an amino acid sequence as set forth in SEQ ID NO: 179.
13. The antibody of any one of claims 1 to 12, wherein the antibody comprises a CDR-H3 amino acid sequence set forth in SEQ ID NO: 752, 780, 815, 772, 784, 770, 774, 756, 796, 785, 761, 788, 767, 765, 799, 749, 802, 809, 750, 803, 800, 806, 759, 805, 797, 787, or 766, 782, 967, 776, 755, 876, 891, 963, 996, 993, 970, 968 or 1008.
14. The antibody of any one of claims 1 to 13, wherein the antibody comprises a CDR-L3 amino acid sequence set forth in any one of SEQ ID NO: 1367, 1394, 1660, 1387, 1397, 1385, 1388, 1371, 1409, 1398, 1376, 1401, 1382, 1380, 1656, 1364, 1413, 1418, 1365, 1658, 1657, 1415, 1374, 1659, 1410, 1400, 1381, 1395, 1582, 1390, 1370, 1670, 1485, 1578, 1611, 1608, 1585, 1583, or 1623.
15. The antibody of any one of claims 1 to 14, wherein the antibody comprises a CDR- Hl, a CDR-H2, and/or a CDR-H3 from the variable heavy chain (VH) amino acid sequence of clone BP6a-l, BP6a-2, BP6a-3, BP6a-4, BP6a-5, BP6a-6, BP6a-7, BP6a-8, BP6a-9, BP6a- 10, BP6a-l l, BP6a-12, BP6a-13, BP6a-14, BP6a-15, BP6a-16, BP6a-17, BP6a-18, BP6a-19, BP6a-20, BP6a-21, BP6a-22, BP6a-23, BP6a-24, BP6a-25, BP6a-26, BP6a-27, BP6b-l, BP6b-2, BP6b-3, BP6b-4, BP6b-5, BP6b-6, BP6b-7, BP6b-8, BP6b-9, BP6b-10, BP6b-l l or BP6b-12 as set forth in Table 14, and/or a CDR-L1, a CDR-L2, and/or a CDR-L3 from the variable light chain (VL) amino acid sequence of clone BP6a-l, BP6a-2, BP6a-3, BP6a-4, BP6a-5, BP6a-6, BP6a-7, BP6a-8, BP6a-9, BP6a-10, BP6a-l l, BP6a-12, BP6a-13, BP6a-14, BP6a-15, BP6a-16, BP6a-17, BP6a-18, BP6a-19, BP6a-20, BP6a-21, BP6a-22, BP6a-23, BP6a-24, BP6a-25, BP6a-26, BP6a-27, BP6b-l, BP6b-2, BP6b-3, BP6b-4, BP6b-5, BP6b-6, BP6b-7, BP6b-8, BP6b-9, BP6b-10, BP6b-l l or BP6b-12 as set forth in Table 16.
16. The antibody of any one of claims 1 to 15, wherein the antibody comprises a variable heavy chain amino acid sequence of clone BP6a-l, BP6a-2, BP6a-3, BP6a-4, BP6a-5, BP6a- 6, BP6a-7, BP6a-8, BP6a-9, BP6a-10, BP6a-l l, BP6a-12, BP6a-13, BP6a-14, BP6a-15, BP6a-16, BP6a-17, BP6a-18, BP6a-19, BP6a-20, BP6a-21, BP6a-22, BP6a-23, BP6a-24, BP6a-25, BP6a-26, BP6a-27, BP6b-l, BP6b-2, BP6b-3, BP6b-4, BP6b-5, BP6b-6, BP6b-7, BP6b-8, BP6b-9, BP6b-10, BP6b-l l or BP6b-12 as set forth in Table 14.
17. The antibody of any one of claims 1 to 16, wherein the antibody comprises the variable light chain amino acid sequence of clone BP6a-l, BP6a-2, BP6a-3, BP6a-4, BP6a-5, BP6a-6, BP6a-7, BP6a-8, BP6a-9, BP6a-10, BP6a-l l, BP6a-12, BP6a-13, BP6a-14, BP6a- 15, BP6a-16, BP6a-17, BP6a-18, BP6a-19, BP6a-20, BP6a-21, BP6a-22, BP6a-23, BP6a-24, BP6a-25, BP6a-26, BP6a-27, BP6b-l, BP6b-2, BP6b-3, BP6b-4, BP6b-5, BP6b-6, BP6b-7, BP6b-8, BP6b-9, BP6b-10, BP6b-l l or BP6b-12 as set forth in Table 16.
18. An antibody that specifically binds human proMyostatin, and human proGDFl 1 ARM8 but does not specifically bind human latent Myostatin having an amino acid sequence as set forth in SEQ ID NO: 5, human GDF-8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 38, human GDF-11 ARM8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 181, or human mature GDF-8 having an amino acid sequence as set forth in SEQ ID NO: 42.
19. An antibody that specifically binds human proMyostatin, murine proMyostatin and human proGDFl 1 ARM8 but does not specifically bind human latent Myostatin having an amino acid sequence as set forth in SEQ ID NO: 5, human GDF-8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 38, human GDF-11 ARM8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 181, or human mature GDF-8 having an amino acid sequence as set forth in SEQ ID NO: 42.
20. The antibody of claim 18 or 19, wherein the human proMyostatin has an amino acid sequence as set forth in SEQ ID NO: 5.
21. The antibody of claim 19 or 20, wherein the murine proMyostatin has an amino acid sequence as set forth in SEQ ID NO: 88.
22. The antibody of any one of claims 18 to 21, wherein the human proGDFl 1 ARM8 has an amino acid sequence as set forth in SEQ ID NO: 179.
23. The antibody of any one of claims 18 to 22, wherein the antibody comprises a CDR- H3 amino acid sequence set forth in SEQ ID NO: 813.
24. The antibody of any one of claims 18 to 23, wherein the antibody comprises a CDR- L3 amino acid sequence set forth in any one of SEQ ID NO: 1422.
25. The antibody of any one of claims 18 to 24, wherein the antibody comprises a CDR- Hl, a CDR-H2, and/or a CDR-H3 from the variable heavy chain (VH) amino acid sequence of clone BP2a-l as set forth in Table 14, and/or a CDR-L1, a CDR-L2, and/or a CDR-L3 from the variable light chain (VL) amino acid sequence of clone BP2a-l as set forth in Table 16.
26. The antibody of any one of claims 18 to 25, wherein the antibody comprises a variable heavy chain amino acid sequence of clone BP2a-l as set forth in Table 14.
27. The antibody of any one of claims 18 to 26, wherein the antibody comprises the variable light chain amino acid sequence of clone BP2a-l as set forth in Table 16.
28. An antibody that specifically binds human proMyostatin, and human latent Myostatin, but does not specifically bind human GDF-8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 38, human GDF-11 ARM8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 181, human proGDFl 1 ARM8 having an amino acid sequence as set forth in SEQ ID NO: 179, or human mature GDF-8 having an amino acid sequence as set forth in SEQ ID NO: 42.
29. An antibody that specifically binds human proMyostatin, murine proMyostatin, and human latent Myostatin, but does not specifically bind human GDF-8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 38, human GDF-11 ARM8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 181, human proGDFl 1 ARM8 having an amino acid sequence as set forth in SEQ ID NO: 179, or human mature GDF-8 having an amino acid sequence as set forth in SEQ ID NO: 42.
30. The antibody of claim 28 or 29, wherein the human proMyo statin has an amino acid sequence as set forth in SEQ ID NO: 5.
31. The antibody of claim 29 or 30, wherein the murine proMyostatin has an amino acid sequence as set forth in SEQ ID NO: 88.
32. The antibody of any one of claims 28 to 31, wherein the human latent Myostatin has an amino acid sequence as set forth in SEQ ID NO: 5.
33. The antibody of any one of claims 28 to 32, wherein the antibody comprises a CDR- H3 amino acid sequence set forth in SEQ ID NO: 794, 804, or 751.
34. The antibody of any one of claims 28 to 33, wherein the antibody comprises a CDR- L3 amino acid sequence set forth in any one of SEQ ID NO: 1407, 1414, or 1366.
35. The antibody of any one of claims 28 to 34, wherein the antibody comprises a CDR- Hl, a CDR-H2, and/or a CDR-H3 from the variable heavy chain (VH) amino acid sequence of clone BP3a-l, BP3a-2 or BP3a-3 as set forth in Table 14, and/or a CDR-L1, a CDR-L2, and/or a CDR-L3 from the variable light chain (VL) amino acid sequence of clone BP3a-l, BP3a-2 or BP3a-3 as set forth in Table 16.
36. The antibody of any one of claims 28 to 35, wherein the antibody comprises a variable heavy chain amino acid sequence of clone BP3a-l, BP3a-2 or BP3a-3 as set forth in Table 14.
37. The antibody of any one of claims 28 to 36, wherein the antibody comprises the variable light chain amino acid sequence of clone BP3a-l, BP3a-2 or BP3a-3 as set forth in Table 16.
38. An antibody that specifically binds human proMyostatin, human latent Myostatin and human GDF8 prodomain but does not specifically bind human GDF-11 ARM8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 181, human proGDFl 1 ARM8 having an amino acid sequence as set forth in SEQ ID NO: 179 or human mature GDF-8 having an amino acid sequence as set forth in SEQ ID NO: 42.
39. An antibody that specifically binds human proMyostatin, murine proMyostatin, human latent Myostatin and human GDF8 prodomain but does not specifically bind human GDF-11 ARM8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 181, human proGDFl 1 ARM8 having an amino acid sequence as set forth in SEQ ID NO: 179 or human mature GDF-8 having an amino acid sequence as set forth in SEQ ID NO: 42.
40. The antibody of claim 38 or 39, wherein the human proMyostatin has an amino acid sequence as set forth in SEQ ID NO: 5.
41. The antibody of claim 39 or 40, wherein the murine proMyostatin has an amino acid sequence as set forth in SEQ ID NO: 88.
42. The antibody of any one of claims 38 to 41, wherein the human latent Myostatin has an amino acid sequence as set forth in SEQ ID NO: 5.
43. The antibody of any one of claims 38 to 42, wherein the human GDF-8 prodomain has an amino acid sequence as set forth in SEQ ID NO: 38.
44. The antibody of any one of claims 38 to 43, wherein the antibody comprises a CDR- H3 amino acid sequence set forth in SEQ ID NO: 768.
45. The antibody of any one of claims 38 to 44, wherein the antibody comprises a CDR- L3 amino acid sequence set forth in any one of SEQ ID NO: 1383.
46. The antibody of any one of claims 38 to 45, wherein the antibody comprises a CDR- Hl, a CDR-H2, and/or a CDR-H3 from the variable heavy chain (VH) amino acid sequence of clone BP4a-l as set forth in Table 14, and/or a CDR-L1, a CDR-L2, and/or a CDR-L3 from the variable light chain (VL) amino acid sequence of clone BP4a-l as set forth in Table 16.
47. The antibody of any one of claims 38 to 46, wherein the antibody comprises a variable heavy chain amino acid sequence of clone BP4a-l as set forth in Table 14.
48. The antibody of any one of claims 38 to 47, wherein the antibody comprises the variable light chain amino acid sequence of clone BP4a-l as set forth in Table 16.
49. An antibody that specifically binds human proMyostatin, human latent Myostatin, and human proGDFl 1 ARM8, but does not specifically bind human GDF-8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 38, human GDF-11 ARM8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 181, or human mature GDF-8 having an amino acid sequence as set forth in SEQ ID NO: 42.
50. An antibody that specifically binds human proMyostatin, murine proMyostatin, human latent Myostatin, and human proGDFl 1 ARM8, but does not specifically bind human GDF-8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 38, human GDF-11 ARM8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 181, or human mature GDF-8 having an amino acid sequence as set forth in SEQ ID NO: 42.
51. The antibody of claim 49 or 50, wherein the human proMyostatin has an amino acid sequence as set forth in SEQ ID NO: 5.
52. The antibody of claim 50 or 51, wherein the murine proMyostatin has an amino acid sequence as set forth in SEQ ID NO: 88.
53. The antibody of any one of claims 49 to 52, wherein the human latent Myostatin has an amino acid sequence as set forth in SEQ ID NO: 5.
54. The antibody of any one of claims 49 to 53, wherein the human proGDFl 1 ARM8 has an amino acid sequence as set forth in SEQ ID NO: 179.
55. The antibody of any one of claims 50 to 54, wherein the antibody specifically binds human latent proGDFl 1 ARM8, but does not specifically bind human proGDFl 1 having an amino acid sequence as set forth in SEQ ID NO: 4.
56. The antibody of any one of claims 50 to 54, wherein the antibody specifically binds murine latent myostatin and human latent proGDFl 1 ARM8, but does not specifically bind human proGDFl 1 having an amino acid sequence as set forth in SEQ ID NO: 4.
57. The antibody of claim 56, wherein the murine latent myostatin has an amino acid sequence as set forth in SEQ ID NO: 88.
58. The antibody of any one of claims 55 to 57, wherein the human latent proGDFl 1 ARM8 has an amino acid sequence as set forth in SEQ ID NO: 179.
59. The antibody of any one of claims 49 to 58, wherein the antibody comprises a CDR- H3 amino acid sequence set forth in SEQ ID NO: 760, 798, 792, 789, 801, 790, 812, 758, 762, 775, 834, 974, 985, or 1002.
60. The antibody of any one of claims 59 to 59, wherein the antibody comprises a CDR- L3 amino acid sequence set forth in any one of SEQ ID NO: 1375, 1411, 1405, 1402, 1412, 1403, 1421, 1373, 1377, 1389, 1661, 1589, 1600, or 1617.
61. The antibody of any one of claims 49 to 60, wherein the antibody comprises a CDR- Hl, a CDR-H2, and/or a CDR-H3 from the variable heavy chain (VH) amino acid sequence of clone BP5a-l, BP5a-2, BP5a-3, BP5a-4 or BP5a-5, BP5b-l, BP5b-2, BP5b-3, BP5b-4, BP5b-5, BP5b-6, BP5b-7, BP5b-8 or BP5b-9 as set forth in Table 14, and/or a CDR-L1, a CDR-L2, and/or a CDR-L3 from the variable light chain (VL) amino acid sequence of clone BP5a-l, BP5a-2, BP5a-3, BP5a-4 or BP5a-5, BP5b-l, BP5b-2, BP5b-3, BP5b-4, BP5b-5, BP5b-6, BP5b-7, BP5b-8 or BP5b-9 as set forth in Table 16.
62. The antibody of any one of claims 49 to 61, wherein the antibody comprises a variable heavy chain amino acid sequence of clone BP5a-l, BP5a-2, BP5a-3, BP5a-4 or BP5a-5, BP5b-l, BP5b-2, BP5b-3, BP5b-4, BP5b-5, BP5b-6, BP5b-7, BP5b-8 or BP5b-9 as set forth in Table 14.
63. The antibody of any one of claims 49 to 62, wherein the antibody comprises the variable light chain amino acid sequence of clone BP5a-l, BP5a-2, BP5a-3, BP5a-4 or BP5a- 5, BP5b-l, BP5b-2, BP5b-3, BP5b-4, BP5b-5, BP5b-6, BP5b-7, BP5b-8 or BP5b-9 as set forth in Table 16.
64. An antibody that specifically binds human proMyo statin but does not specifically bind human latent Myostatin having an amino acid sequence as set forth in SEQ ID NO: 5, human GDF-8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 38, human GDF- 11 ARM8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 181, human proGDFl 1 ARM8 having an amino acid sequence as set forth in SEQ ID NO: 179, or human mature GDF-8 having an amino acid sequence as set forth in SEQ ID NO: 42.
65. An antibody that specifically binds human proMyostatin and murine proMyostatin but does not specifically bind human latent Myostatin having an amino acid sequence as set forth in SEQ ID NO: 5, human GDF-8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 38, human GDF- 11 ARM8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 181, human proGDFl 1 ARM8 having an amino acid sequence as set forth in SEQ ID NO: 179,or human mature GDF-8 having an amino acid sequence as set forth in SEQ ID NO: 42.
66. The antibody of claim 64 or 65, wherein the human proMyostatin has an amino acid sequence as set forth in SEQ ID NO: 5.
67. The antibody of claim 65 or 66, wherein the murine proMyostatin has an amino acid sequence as set forth in SEQ ID NO: 88).
68. The antibody of any one of claims 64 to 67, wherein the antibody does not specifically bind human proGDFl 1 having an amino acid sequence as set forth in SEQ ID NO: 4, or human latent proGDFl 1 ARM8 having an amino acid sequence as set forth in SEQ ID NO: 179.
69. The antibody of any one of claims 65 to 67, wherein the antibody does not specifically bind human proGDFl 1 having an amino acid sequence as set forth in SEQ ID NO: 4, murine latent myostatin having an amino acid sequence as set forth in SEQ ID NO: 88 or human latent proGDFl 1 ARM8 having an amino acid sequence as set forth in SEQ ID NO: 179.
70. The antibody of any one of claims 64 to 69, wherein the antibody comprises a CDR- H3 amino acid sequence as set forth in SEQ ID NO: 769 or 827.
71. The antibody of any one of claims 64 to 70, wherein the antibody comprises a CDR- L3 amino acid sequence set forth in any one of SEQ ID NO: 1384 or 1435.
72. The antibody of any one of claims 64 to 71, wherein the antibody comprises a CDR- Hl, a CDR-H2, and/or a CDR-H3 from the variable heavy chain (VH) amino acid sequence of clone BPla-1 or BPlb-1 as set forth in Table 14, and/or a CDR-L1, a CDR-L2, and/or a CDR-L3 from the variable light chain (VL) amino acid sequence of clone BPla-1 or BPlb-1 as set forth in Table 16.
73. The antibody of any one of claims 64 to 72, wherein the antibody comprises a variable heavy chain amino acid sequence of clone BPla-1 or BPlb-1 as set forth in Table 14.
74. The antibody of any one of claims 64 to 73, wherein the antibody comprises the variable light chain amino acid sequence of clone BPla-1 or BPlb-1 as set forth in Table 16.
75. An antibody that specifically binds human latent Myostatin but does not specifically bind human proMyo statin having an amino acid sequence as set forth in SEQ ID NO: 5, human GDF-8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 38, human GDF- 11 ARM8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 181, human proGDFl 1 ARM8 having an amino acid sequence as set forth in SEQ ID NO: 179, human mature GDF-8 having an amino acid sequence as set forth in SEQ ID NO: 42 or human proGDFl 1 having an amino acid sequence as set forth in SEQ ID NO: 4).
76. An antibody that specifically binds human latent Myostatin but does not specifically bind murine proMyo statin having an amino acid sequence as set forth in SEQ ID NO: 88, human proMyo statin having an amino acid sequence as set forth in SEQ ID NO: 5, human GDF-8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 38, human GDF-11 ARM8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 181, human proGDFl 1 ARM8 having an amino acid sequence as set forth in SEQ ID NO: 179, human mature GDF-8 having an amino acid sequence as set forth in SEQ ID NO: 42 or human proGDFl 1 having an amino acid sequence as set forth in SEQ ID NO: 4).
77. The antibody of claim 75 or 76, wherein the human latent Myostatin has an amino acid sequence as set forth in SEQ ID NO: 5.
78. The antibody of any one of claims 75 to 77, wherein the antibody specifically binds human latent proGDFl 1 ARM8.
79. The antibody of claim 76 or 77, wherein the antibody specifically binds murine latent myostatin and human latent proGDFl 1 ARM8.
80. The antibody of claim 79, wherein the murine latent myostatin has an amino acid sequence as set forth in SEQ ID NO: 88.
81. The antibody of any one of claims 78 to 80, wherein the human latent proGDFl 1 ARM8 has an amino acid sequence as set forth in SEQ ID NO: 179.
82. The antibody of any one of claims 75 to 81, wherein the antibody comprises a CDR- H3 amino acid sequence set forth in SEQ ID NO: 990.
83. The antibody of any one of claims 75 to 82, wherein the antibody comprises a CDR- L3 amino acid sequence set forth in any one of SEQ ID NO: 1605.
84. The antibody of any one of claims 75 to 83, wherein the antibody comprises a CDR- Hl, a CDR-H2, and/or a CDR-H3 from the variable heavy chain (VH) amino acid sequence of clone BPlOb-1 as set forth in Table 14, and/or a CDR-L1, a CDR-L2, and/or a CDR-L3 from the variable light chain (VL) amino acid sequence of clone BPlOb-1 as set forth in Table 16.
85. The antibody of any one of claims 75 to 84, wherein the antibody comprises a variable heavy chain amino acid sequence of clone BPlOb-1 as set forth in Table 14.
86. The antibody of any one of claims 75 to 85, wherein the antibody comprises the variable light chain amino acid sequence of clone BP 10b- las set forth in Table 16.
87. An antibody that specifically binds human proMyostatin, human latent Myostatin, human proGDFl 1 ARM8 and human mature GDF-8 but does not specifically bind human GDF-8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 38 or human GDF-11 ARM8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 181.
88. An antibody that specifically binds human proMyostatin, murine proMyostatin, human latent Myostatin, human proGDFl 1 ARM8 and human mature GDF-8 but does not specifically bind human GDF-8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 38 or human GDF-11 ARM8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 181.
89. The antibody of claim 87 or 88, wherein the human proMyostatin has an amino acid sequence as set forth in SEQ ID NO: 5.
90. The antibody of claim 88 or 89, wherein the murine proMyostatin has an amino acid sequence as set forth in SEQ ID NO: 88.
91. The antibody of any one of claims 87 to 90, wherein the human latent Myostatin has an amino acid sequence as set forth in SEQ ID NO: 5.
92. The antibody of any one of claims 87 to 91, wherein the human proGDFl 1 ARM8 has an amino acid sequence as set forth in SEQ ID NO: 179.
93. The antibody of any one of claims 87 to 92, wherein the human mature GDF-8 has an amino acid sequence as set forth in SEQ ID NO: 42.
94. The antibody of any one of claims 87 to 93, wherein the antibody comprises a CDR- H3 amino acid sequence set forth in SEQ ID NO: 764.
95. The antibody of any one of claims 87 to 94, wherein the antibody comprises a CDR- L3 amino acid sequence set forth in any one of SEQ ID NO: 1379.
96. The antibody of any one of claims 87 to 95, wherein the antibody comprises a CDR- Hl, a CDR-H2, and/or a CDR-H3 from the variable heavy chain (VH) amino acid sequence of clone BP1 la-1 as set forth in Table 14, and/or a CDR-L1, a CDR-L2, and/or a CDR-L3 from the variable light chain (VL) amino acid sequence of clone BP1 la-1 as set forth in Table 16.
97. The antibody of any one of claims 87 to 96, wherein the antibody comprises a variable heavy chain amino acid sequence of clone BP1 la-1 as set forth in Table 14.
98. The antibody of any one of claims 87 to 97, wherein the antibody comprises the variable light chain amino acid sequence of clone BP1 la-1 as set forth in Table 16.
99. An antibody that specifically binds human latent Myostatin and human mature GDF-8 but does not specifically bind human proMyo statin having an amino acid sequence as set forth in SEQ ID NO: 5, human GDF-8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 38, human GDF-11 ARM8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 181 or human proGDFl 1 ARM8 having an amino acid sequence as set forth in SEQ ID NO: 179.
100. An antibody that specifically binds human latent Myostatin and human mature GDF-8 but does not specifically bind murine proMyo statin having an amino acid sequence as set forth in SEQ ID NO: 88, human proMyo statin having an amino acid sequence as set forth in SEQ ID NO: 5, human GDF-8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 38, human GDF-11 ARM8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 181 or human proGDFl 1 ARM8 having an amino acid sequence as set forth in SEQ ID NO: 179.
101. The antibody of claim 99 or 100, wherein the human latent Myostatin has an amino acid sequence as set forth in SEQ ID NO: 5.
102. The antibody of any one of claims 99 to 101, wherein the human mature GDF-8 has an amino acid sequence as set forth in SEQ ID NO: 42.
103. The antibody of any one of claims 100 to 102, wherein the antibody does not specifically bind human proGDFl 1 having an amino acid sequence as set forth in SEQ ID NO: 4 or human latent proGDFl 1 ARM8 having an amino acid sequence as set forth in SEQ ID NO: 179.
104. The antibody of any one of claims 100 to 102, wherein the antibody specifically binds murine latent myostatin but does not specifically bind human proGDFl 1 having an amino acid sequence as set forth in SEQ ID NO: 4 or human latent proGDFl 1 ARM8 having an amino acid sequence as set forth in SEQ ID NO: 179.
105. The antibody of claim 104, wherein the murine latent myostatin has an amino acid sequence as set forth in SEQ ID NO: 88.
106. The antibody of any one of claims 99 to 105, wherein the antibody comprises a CDR- H3 amino acid sequence set forth in SEQ ID NO: 989.
107. The antibody of any one of claims 99 to 106, wherein the antibody comprises a CDR- L3 amino acid sequence set forth in any one of SEQ ID NO: 1604.
108. The antibody of any one of claims 99 to 107, wherein the antibody comprises a CDR- Hl, a CDR-H2, and/or a CDR-H3 from the variable heavy chain (VH) amino acid sequence of clone BP12b-l as set forth in Table 14, and/or a CDR-L1, a CDR-L2, and/or a CDR-L3 from the variable light chain (VL) amino acid sequence of clone BP 12b- las set forth in Table 16.
109. The antibody of any one of claims 99 to 108, wherein the antibody comprises a variable heavy chain amino acid sequence of clone BP12b-l as set forth in Table 14.
110. The antibody of any one of claims 99 to 109, wherein the antibody comprises the variable light chain amino acid sequence of clone BP12b-l as set forth in Table 16.
111. An antibody that specifically binds human proMyo statin and human GDF-8 prodomain, but does not specifically bind human latent Myostatin having an amino acid sequence as set forth in SEQ ID NO: 5, human GDF-11 ARM8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 181, human proGDFl 1 ARM8 having an amino acid sequence as set forth in SEQ ID NO: 179 or human mature GDF-8 having an amino acid sequence as set forth in SEQ ID NO: 42.
112. An antibody that specifically binds human proMyo statin and human GDF-8 prodomain, but does not specifically bind murine proMyo statin having an amino acid sequence as set forth in SEQ ID NO: 88, human latent Myostatin having an amino acid sequence as set forth in SEQ ID NO: 5, human GDF-11 ARM8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 181, human proGDFl 1 ARM8 having an amino acid sequence as set forth in SEQ ID NO: 179 or human mature GDF-8 having an amino acid sequence as set forth in SEQ ID NO: 42.
113. The antibody of claim 111 or 112, wherein the human proMyostatin has an amino acid sequence as set forth in SEQ ID NO: 5.
114. The antibody of any one of claims 111 to 113, wherein the human GDF-8 prodomain has an amino acid sequence as set forth in SEQ ID NO: 38.
115. The antibody of any one of claims 111 to 114, wherein the antibody comprises a CDR-H3 amino acid sequence set forth in SEQ ID NO: 786.
116. The antibody of any one of claims 111 to 115, wherein the antibody comprises a CDR-L3 amino acid sequence set forth in any one of SEQ ID NO: 1399.
117. The antibody of any one of claims 111 to 116, wherein the antibody comprises a CDR-H1, a CDR-H2, and/or a CDR-H3 from the variable heavy chain (VH) amino acid sequence of clone BP15a-l as set forth in Table 14, and/or a CDR-L1, a CDR-L2, and/or a CDR-L3 from the variable light chain (VL) amino acid sequence of clone BP15a-l as set forth in Table 16.
118. The antibody of any one of claims 111 to 117, wherein the antibody comprises a variable heavy chain amino acid sequence of clone BP15a-l as set forth in Table 14.
119. The antibody of any one of claims 111 to 118, wherein the antibody comprises the variable light chain amino acid sequence of clone BP15a-l as set forth in Table 16.
120. An antibody that specifically binds human proMyo statin and human latent Myostatin but does not specifically bind human GDF-8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 38, human GDF-11 ARM8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 181, human proGDFl 1 ARM8 having an amino acid sequence as set forth in SEQ ID NO: 179 or human mature GDF-8 having an amino acid sequence as set forth in SEQ ID NO: 42.
121. An antibody that specifically binds human proMyo statin and human latent Myostatin but does not specifically bind murine proMyostatin having an amino acid sequence as set forth in SEQ ID NO: 88, human GDF-8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 38, human GDF-11 ARM8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 181, human proGDFl 1 ARM8 having an amino acid sequence as set forth in SEQ ID NO: 179 or human mature GDF-8 having an amino acid sequence as set forth in SEQ ID NO: 42.
122. The antibody of claim 120 or 121, wherein the human proMyostatin has an amino acid sequence as set forth in SEQ ID NO: 5.
123 The antibody of any one of claims 121 to 122, wherein the human latent Myostatin has an amino acid sequence as set forth in SEQ ID NO: 5.
124. The antibody of any one of claims 121 to 123, wherein the antibody does not specifically bind human proGDFl 1 having an amino acid sequence as set forth in SEQ ID NO: 4 or human latent proGDFl 1 ARM8 having an amino acid sequence as set forth in SEQ ID NO: 179.
125. The antibody of any one of claims 121 to 123, wherein the antibody does not specifically bind human proGDFl 1 having an amino acid sequence as set forth in SEQ ID NO: 4, murine latent myostatin having an amino acid sequence as set forth in SEQ ID NO: 88 or human latent proGDFl 1 ARM8 having an amino acid sequence as set forth in SEQ ID NO: 179.
126. The antibody of any one of claims 120 to 125, wherein the antibody comprises a CDR-H3 amino acid sequence set forth in SEQ ID NO: 810, 855, 971, 983, or 986.
127. The antibody of any one of claims 120 to 126, wherein the antibody comprises a CDR-L3 amino acid sequence set forth in any one of SEQ ID NO: 1419, 1662, 1586, 1598, or 1601 .
128. The antibody of any one of claims 120 to 127, wherein the antibody comprises a CDR-H1, a CDR-H2, and/or a CDR-H3 from the variable heavy chain (VH) amino acid sequence of clone BP16a-l, BP16b-l, BP16b-2, BP16b-3 or BP16b-4 as set forth in Table 14, and/or a CDR-L1, a CDR-L2, and/or a CDR-L3 from the variable light chain (VL) amino acid sequence of clone BP16a-l, BP16b-l, BP16b-2, BP16b-3 or BP16b-4 as set forth in Table 16.
129. The antibody of any one of claims 120 to 128, wherein the antibody comprises a variable heavy chain amino acid sequence of clone BP16a-l, BP16b-l, BP16b-2, BP16b-3 or BP16b-4 as set forth in Table 14.
130. The antibody of any one of claims 120 to 129, wherein the antibody comprises the variable light chain amino acid sequence of clone BP16a-l, BP16b-l, BP16b-2, BP16b-3 or BP16b-4 as set forth in Table 16.
131. An antibody that specifically binds human proMyo statin and human latent Myostatin but does not specifically bind human GDF-8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 38, human GDF-11 ARM8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 181, human proGDFl 1 ARM8 having an amino acid sequence as set forth in SEQ ID NO: 179, human mature GDF-8 having an amino acid sequence as set forth in SEQ ID NO: 42 or human proGDFl 1 having an amino acid sequence as set forth in SEQ ID NO: 4.
132. An antibody that specifically binds human proMyo statin and human latent Myostatin but does not specifically bind murine proMyostatin having an amino acid sequence as set forth in SEQ ID NO: 88, human GDF-8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 38, human GDF-11 ARM8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 181, human proGDFl 1 ARM8 having an amino acid sequence as set forth in SEQ ID NO: 179, human mature GDF-8 having an amino acid sequence as set forth in SEQ ID NO: 42, human proGDFl 1 having an amino acid sequence as set forth in SEQ ID NO: 4 or murine latent myostatin having an amino acid sequence as set forth in SEQ ID NO: 88.
133. The antibody of claim 131 or 132, wherein the human proMyostatin has amino acid sequence as set forth in SEQ ID NO: 5.
134. The antibody of any one of claims 131 to 133, wherein the human latent Myostatin has an amino acid sequence as set forth in SEQ ID NO: 5.
135. The antibody of any one of claims 131 to 134, wherein the antibody specifically binds human latent proGDFl 1 ARM8.
136. The antibody of claim 135, wherein the human latent proGDFl 1 ARM8 has an amino acid sequence as set forth in SEQ ID NO: 179.
137. The antibody of any one of claims 131 to 134, wherein the antibody does not specifically bind human latent proGDFl 1 ARM8 having an amino acid sequence as set forth in SEQ ID NO: 179.
138. The antibody of any one of claims 131 to 137, wherein the antibody comprises a CDR-H3 amino acid sequence set forth in SEQ ID NO: 982.
139. The antibody of any one of claims 131 to 138, wherein the antibody comprises a CDR-L3 amino acid sequence set forth in any one of SEQ ID NO: 1597.
140. The antibody of any one of claims 131 to 139, wherein the antibody comprises a CDR-H1, a CDR-H2, and/or a CDR-H3 from the variable heavy chain (VH) amino acid sequence of clone BP17b-l as set forth in Table 14, and/or a CDR-Ll, a CDR-L2, and/or a CDR-L3 from the variable light chain (VL) amino acid sequence of clone BP17b-l as set forth in Table 16.
141. The antibody of any one of claims 131 to 140, wherein the antibody comprises a variable heavy chain amino acid sequence of clone BP17b-l as set forth in Table 14.
142. The antibody of any one of claims 131 to 141, wherein the antibody comprises the variable light chain amino acid sequence of clone BP17b-l as set forth in Table 16.
143. An antibody that specifically binds human proMyostatin, human latent Myostatin and human proGDFl 1 ARM8 but does not specifically bind human GDF-8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 38, human GDF-11 ARM8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 181 or human mature GDF-8 having an amino acid sequence as set forth in SEQ ID NO: 42.
144. An antibody that specifically binds human proMyostatin, human latent Myostatin and human proGDFl 1 ARM8 but does not specifically bind murine proMyostatin having an amino acid sequence as set forth in SEQ ID NO: 88, human GDF-8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 38, human GDF-11 ARM8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 181 or human mature GDF-8 having an amino acid sequence as set forth in SEQ ID NO: 42.
145. The antibody of claim 143 or 144, wherein the human proMyostatin has an amino acid sequence as set forth in SEQ ID NO: 5.
146. The antibody of any one of claims 143 to 145, wherein the human latent Myostatin has an amino acid sequence as set forth in SEQ ID NO: 5.
147. The antibody of any one of claims 143 to 146, wherein the human proGDFl 1 ARM8 has an amino acid sequence as set forth in SEQ ID NO: 179.
148. The antibody of any one of claims 143 to 147, wherein the antibody specifically binds human latent proGDFl 1 ARM8 but does not specifically bind human proGDFl 1 having an amino acid sequence as set forth in SEQ ID NO: 4.
149. The antibody of any one of claims 144 to 147, wherein the antibody specifically binds human latent proGDFl 1 ARM8 but does not specifically bind human proGDFl 1 having an amino acid sequence as set forth in SEQ ID NO: 4 or murine latent myostatin having an amino acid sequence as set forth in SEQ ID NO: 88.
150. The antibody of claim 148 or 149, wherein the human latent proGDFl 1 ARM8 has an amino acid sequence as set forth in SEQ ID NO: 179.
151. The antibody of any one of claims 143 to 150, wherein the antibody comprises a CDR-H3 amino acid sequence set forth in SEQ ID NO: 763, 814, 753, 777, 795, 969, 972, 976, or 981.
152. The antibody of any one of claims 143 to 151, wherein the antibody comprises a CDR-L3 amino acid sequence set forth in any one of SEQ ID NO: 1378, 1423, 1368, 1391, 1408, 1584, 1587, 1591, or 1596.
153. The antibody of any one of claims 143 to 152, wherein the antibody comprises a CDR-H1, a CDR-H2, and/or a CDR-H3 from the variable heavy chain (VH) amino acid sequence of clone BP18a-l or BP18a-2, BP18b-l, BP18b-2, BP18b-3, BP18b-4, BP18b-5, BP18b-6 or BP18b-7 as set forth in Table 14, and/or a CDR-Ll, a CDR-L2, and/or a CDR-L3 from the variable light chain (VL) amino acid sequence of clone BP18a-l or BP18a-2, BP18b-l, BP18b-2, BP18b-3, BP18b-4, BP18b-5, BP18b-6 or BP18b-7as set forth in Table 16.
154. The antibody of any one of claims 143 to 153, wherein the antibody comprises a variable heavy chain amino acid sequence of clone BP18a-l or BP18a-2, BP18b-l, BP18b-2, BP18b-3, BP18b-4, BP18b-5, BP18b-6 or BP18b-7 as set forth in Table 14.
155. The antibody of any one of claims 143 to 154, wherein the antibody comprises the variable light chain amino acid sequence of clone BP18a-l or BP18a-2, BP18b-l, BP18b-2, BP18b-3, BP18b-4, BP18b-5, BP18b-6 or BP18b-7 as set forth in Table 16.
156. An antibody that specifically binds human proMyostatin, human latent Myostatin, human GDF-8 prodomain, human GDF-11 ARM8 prodomain and human proGDFl 1 ARM8 but does not specifically bind human mature GDF-8 having an amino acid sequence as set forth in SEQ ID NO: 42.
157. An antibody that specifically binds human proMyostatin, human latent Myostatin, human GDF-8 prodomain, human GDF-11 ARM8 prodomain and human proGDFl 1 ARM8 but does not specifically bind murine proMyostatin having an amino acid sequence as set forth in SEQ ID NO: 88 or human mature GDF-8 having an amino acid sequence as set forth in SEQ ID NO: 42.
158. The antibody of claim 156 or 157, wherein the human proMyostatin has an amino acid sequence as set forth in SEQ ID NO: 5.
159. The antibody of any one of claims 156 to 158, wherein the human latent Myostatin has an amino acid sequence as set forth in SEQ ID NO: 5.
160. The antibody of any one of claims 156 to 159, wherein the human GDF-8 prodomain has an amino acid sequence as set forth in SEQ ID NO: 38.
161. The antibody of any one of claims 156 to 160, wherein the human GDF-11 ARM8 prodomain has an amino acid sequence as set forth in SEQ ID NO: 181.
162. The antibody of any one of claims 156 to 161, wherein the human proGDFl 1 ARM8 has an amino acid sequence as set forth in SEQ ID NO: 179.
163. The antibody of any one of claims 156 to 162, wherein the antibody specifically binds human latent proGDFl 1 ARM8 but does not specifically bind human proGDFl 1 having an amino acid sequence as set forth in SEQ ID NO: 4.
164. The antibody of any one of claims 157 to 162, wherein the antibody specifically binds human latent proGDFl 1 ARM8 but does not specifically bind human proGDFl 1 having an amino acid sequence as set forth in SEQ ID NO: 4 or murine latent myostatin having an amino acid sequence as set forth in SEQ ID NO: 88.
165. The antibody of claim 163 or 164 wherein the human latent proGDFl 1 ARM8 has an amino acid sequence as set forth in SEQ ID NO: 179.
166. The antibody of any one of claims 156 to 165, wherein the antibody comprises a CDR-H3 amino acid sequence set forth in SEQ ID NO: 773, 807, 811, 754, 771, or 978.
167. The antibody of any one of claims 156 to 166, wherein the antibody comprises a CDR-L3 amino acid sequence set forth in any one of SEQ ID NO: 1655, 1416, 1420, 1369, 1386, or 1593.
168. The antibody of any one of claims 156 to 167, wherein the antibody comprises a CDR-H1, a CDR-H2, and/or a CDR-H3 from the variable heavy chain (VH) amino acid sequence of clone BP19a-l, BP19a-2 or BP19a-3, BP19b-l, BP19b-2 or BP19b-3 as set forth in Table 14, and/or a CDR-L1, a CDR-L2, and/or a CDR-L3 from the variable light chain (VL) amino acid sequence of clone BP19a-l, BP19a-2 or BP19a-3, BP19b-l, BP19b-2 or BP19b-3 as set forth in Table 16.
169. The antibody of any one of claims 156 to 168, wherein the antibody comprises a variable heavy chain amino acid sequence of clone BP19a-l, BP19a-2 or BP19a-3, BP19b-l, BP19b-2 or BP19b-3 as set forth in Table 14.
170. The antibody of any one of claims 156 to 169, wherein the antibody comprises the variable light chain amino acid sequence of clone BP19a-l, BP19a-2 or BP19a-3, BP19b-l, BP19b-2 or BP19b-3 as set forth in Table 16.
171. An antibody that specifically binds human proMyostatin, human latent Myostatin, human proGDFl 1 ARM8, human proGDFl 1 or human latent proGDFl 1 ARM 8 but does not specifically bind human GDF-8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 38, human GDF-11 ARM8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 181 or human mature GDF-8 having an amino acid sequence as set forth in SEQ ID NO: 42.
172. An antibody that specifically binds human proMyostatin, murine proMyostatin, human latent Myostatin, human proGDFl 1 ARM8, human proGDFl 1, murine latent myostatin or human latent proGDFl 1 ARM8 but does not specifically bind human GDF-8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 38, human GDF-11 ARM8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 181 or human mature GDF-8 having an amino acid sequence as set forth in SEQ ID NO: 42.
173. The antibody of claim 171 or 172, wherein the human proMyostatin has an amino acid sequence as set forth in SEQ ID NO: 5.
174. The antibody of claim 172 or 173, wherein the murine proMyostatin has an amino acid sequence as set forth in SEQ ID NO: 88.
175. The antibody of any one of claims 171 to 174, wherein the human latent Myostatin has an amino acid sequence as set forth in SEQ ID NO: 5.
176. The antibody of any one of claims 171 to 175, wherein the human proGDFl 1 ARM8 has an amino acid sequence as set forth in SEQ ID NO: 179.
177. The antibody of any one of claims 171 to 176, wherein the human proGDFl 1 has an amino acid sequence as set forth in SEQ ID NO: 4.
178. The antibody of any one of claims 172 to 177, wherein the murine latent myostatin has an amino acid sequence as set forth in SEQ ID NO: 88.
179. The antibody of any one of claims 171 to 178, wherein the human latent proGDFl 1 ARM8 has an amino acid sequence as set forth in SEQ ID NO: 179.
180. The antibody of any one of claims 171 to 179, wherein the antibody comprises a CDR-H3 amino acid sequence set forth in SEQ ID NO: 779.
181. The antibody of any one of claims 171 to 180, wherein the antibody comprises a CDR-L3 amino acid sequence set forth in any one of SEQ ID NO: 1393.
182. The antibody of any one of claims 171 to 181, wherein the antibody comprises a CDR-H1, a CDR-H2, and/or a CDR-H3 from the variable heavy chain (VH) amino acid sequence of clone BP21b-l as set forth in Table 14, and/or a CDR-L1, a CDR-L2, and/or a CDR-L3 from the variable light chain (VL) amino acid sequence of clone BP21b-l as set forth in Table 16.
183. The antibody of any one of claims 171 to 182, wherein the antibody comprises a variable heavy chain amino acid sequence of clone BP21b-l as set forth in Table 14.
184. The antibody of any one of claims 171 to 183, wherein the antibody comprises the variable light chain amino acid sequence of clone BP21b-l as set forth in Table 16.
185. An antibody that specifically binds human proMyostatin, human latent Myostatin, human proGDFl 1, and human latent proGDFl 1 ARM8 but does not specifically bind human GDF-8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 38, human GDF-11 ARM8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 181, human proGDFl 1 ARM8 having an amino acid sequence as set forth in SEQ ID NO: 179 or human mature GDF-8 having an amino acid sequence as set forth in SEQ ID NO: 42.
186. An antibody that specifically binds human proMyostatin, murine proMyostatin, human latent Myostatin, human proGDFl 1, murine latent myostatin and human latent proGDFl 1 ARM8 but does not specifically bind human GDF-8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 38, human GDF-11 ARM8 prodomain having an amino acid sequence as set forth in SEQ ID NO: 181, human proGDFl 1 ARM8 having an amino acid sequence as set forth in SEQ ID NO: 179 or human mature GDF-8 having an amino acid sequence as set forth in SEQ ID NO: 42.
187. The antibody of claim 185 or 186, wherein the human proMyostatin has an amino acid sequence as set forth in SEQ ID NO: 5.
188. The antibody of claim 186 or 187, wherein the murine proMyostatin has an amino acid sequence as set forth in SEQ ID NO: 88.
189. The antibody of any one of claims 185 to 188, wherein the human latent Myostatin has an amino acid sequence as set forth in SEQ ID NO: 5.
190. The antibody of any one of claims 185 to 189, wherein the human proGDFl 1 has an amino acid sequence as set forth in SEQ ID NO: 4.
191. The antibody of any one of claims 186 to 190, wherein the murine latent myostatin has an amino acid sequence as set forth in SEQ ID NO: 88.
192. The antibody of any one of claims 185 to 191, wherein the human latent proGDFl 1 ARM8 has an amino acid sequence as set forth in SEQ ID NO: 179.
193. The antibody of any one of claims 185 to 192, wherein the antibody comprises a CDR-H3 amino acid sequence set forth in SEQ ID NO: 1006.
194. The antibody of any one of claims 185 to 193, wherein the antibody comprises a CDR-L3 amino acid sequence set forth in any one of SEQ ID NO: 1621.
195. The antibody of any one of claims 185 to 194, wherein the antibody comprises a CDR-Hl, a CDR-H2, and/or a CDR-H3 from the variable heavy chain (VH) amino acid sequence of clone BP22b-l as set forth in Table 14, and/or a CDR-Ll, a CDR-L2, and/or a CDR-L3 from the variable light chain (VL) amino acid sequence of clone BP22b-las set forth in Table 16.
196. The antibody of any one of claims 185 to 195, wherein the antibody comprises a variable heavy chain amino acid sequence of clone BP22b-l as set forth in Table 14.
197. The antibody of any one of claims 185 to 196, wherein the antibody comprises the variable light chain amino acid sequence of clone BP22b-l as set forth in Table 16.
198. An antibody that specifically binds proMyostatin and/or latent Myostatin, wherein the antibody has an affinity to the proMyostatin and/or latent Myostatin that is pH sensitive.
199. An antibody that specifically binds proMyostatin and/or latent Myostatin with a greater affinity at a pH in a range from 6.5 to 7.5 than it does at a pH in a range from 4.6 to 6.0.
200. The antibody of claim 198 or 199, wherein the antibody has a Kd of binding to pro/latent-Myostatin in a range fromlO -"3 M to 10 -"8 M under binding conditions having a pH in a range from 4.6 to 6.0.
201. The antibody of any one of claims 198 to 200, wherein the antibody has a Kd of binding to pro/latent-Myostatin in a range fromlO"6 M to 10"11 M, when the pH is in a range from 6.5 to 7.5.
202. The antibody of any one of claims 198 to 201, wherein the antibody comprises a CDR-H3 amino acid sequence set forth in SEQ ID NO: 813, 804, 790, 762, 784, 770, 764, 986, 777, or 1006.
203. The antibody of any one of claims 198 to 202, wherein the antibody comprises a CDR-L3 amino acid sequence set forth in any one of SEQ ID NO: 1422, 1414, 1403, 1377, 1397, 1385, 1379, 1601, 1391, or 1621.
204. The antibody of any one of claims 198 to 203, wherein the antibody comprises a CDR-H1, a CDR-H2, and/or a CDR-H3 from the variable heavy chain (VH) amino acid sequence of clone BP2a-l, BP3a-2, BP5b-l, BP5b-4, BP6a-5, BP6a-6, BPl la-1, BP16b-4, BP18b-2 or BP22b-l as set forth in Table 14, and/or a CDR-Ll, a CDR-L2, and/or a CDR-L3 from the variable light chain (VL) amino acid sequence of clone BP2a-l, BP3a-2, BP5b-l, BP5b-4, BP6a-5, BP6a-6, BPl la-1, BP16b-4, BP18b-2 or BP22b-l as set forth in Table 16.
205. The antibody of any one of claims 198 to 204, wherein the antibody comprises a variable heavy chain amino acid sequence of clone BP2a-l, BP3a-2, BP5b-l, BP5b-4, BP6a- 5, BP6a-6, BPl la-1, BP16b-4, BP18b-2 or BP22b-las set forth in Table 14.
206. The antibody of any one of claims 198 to 205, wherein the antibody comprises the variable light chain amino acid sequence of clone BP2a-l, BP3a-2, BP5b-l, BP5b-4, BP6a-5, BP6a-6, BPl la-1, BP16b-4, BP18b-2 or BP22b-las set forth in Table 16.
207. An isolated antibody or antigen binding fragment thereof having at least one variable domain with an amino acid sequence having at least 95% sequence identity to one or more of those listed in Table 14 or 16.
208. The isolated antibody or antigen binding fragment thereof of claim 207 wherein said at least one variable domain comprises a variable domain pair, said variable domain pair comprising a heavy chain variable domain (VH) and a light chain variable domain (VL), wherein said variable domain pair comprises at least 95% sequence identity to a variable domain pair present in an scFv selected from any of those listed in Table 13.
209. An isolated antibody or antigen binding fragment thereof comprising at least one variable domain, said at least one variable domain comprising at least one complementarity determining region (CDR) sequence comprising at least 70% sequence identity to one or more of those listed in Table 15 or 17.
210. The isolated antibody or antigen binding fragment thereof of claim 209, wherein said at least one CDR sequence comprises at least 70% sequence identity to a heavy chain variable domain CDR (CDR-H) selected from any of those listed in Table 15.
211. The isolated antibody or antigen binding fragment thereof of claim 209, comprising at least one variable domain pair, said variable domain pair comprising a VH and a VL.
212. The isolated antibody or antigen binding fragment thereof of claim 211, wherein said VH comprises at least one CDR-H sequence with at least 70% sequence identity to a sequence selected from any of those listed in Table 15.
213. The isolated antibody or antigen binding fragment thereof of claim 211, wherein said VL comprises at least one CDR-L sequence with at least 70% sequence identity to a CDR-L sequence selected from any of those listed in Table 17.
214. An isolated antibody or antigen binding fragment thereof that comprises:
i. a VH sequence with at least 95% sequence identity to a sequence listed in Table 14, and
ii. a VL sequence with at least 95% sequence identity to a sequence listed in Table 16.
215. An isolated antibody or antigen binding fragment thereof that comprises:
i. a CDR-H1 sequence with at least 70% sequence identity to a sequence listed in Table 15,
ii. a CDR-H2 sequence with at least 70% sequence identity to a sequence listed in Table 15,
iii. a CDR-H3 sequence with at least 70% sequence identity to a sequence listed in Table 15,
iv. a CDR-L1 sequence with at least 70% sequence identity to a sequence listed in Table 17,
v. a CDR-L2 sequence with at least 70% sequence identity to a sequence listed in Table 17, and
vi. a CDR-L3 sequence with at least 70% sequence identity to a sequence listed in Table 17.
216. The isolated antibody or antigen binding fragment thereof of any of claims 207-215, comprising an antibody, wherein said antibody is a monoclonal antibody.
217. The isolated antibody or antigen binding fragment thereof of any of claims 207-216, comprising an antibody, wherein said antibody comprises an IgG isotype.
218. The antibody of claim 217, wherein said antibody comprises an IgGl isotype.
219. The isolated antibody or antigen binding fragment thereof of any of claim 207-218, wherein said antibody binds to one or more recombinant antigens listed in Table 24 and/or one or more polypeptides comprising an amino acid sequence with at least 95% sequence identity to any of those listed in Tables 1, 2, 4, 5, 9, 10, 19, 20 or 25.
220. The isolated antibody or antigen binding fragment thereof of claim 219, wherein said polypeptide comprises pro-growth differentiation factor (GDF)-8 (proGDF-8) (SEQ ID NO: 5).
221. The isolated antibody or antigen binding fragment thereof of any of claims 207-218, wherein said antibody binds to a latent GDF-8 growth factor prodomain complex (GPC).
222. A method of modulating growth factor activity in a biological system comprising contacting said biological system with the antibody or antigen binding fragment thereof of any of claims 1-206 or 207-221.
223. The method of claim 222, wherein said growth factor activity comprises GDF-8 activity.
224. The method of claim 222 or 223, wherein said antibody is a stabilizing antibody and wherein contacting said biological system with said stabilizing antibody results in inhibition of release of at least 5% of total GDF-8 mature growth factor in said biological system.
225. The method of claim 224, wherein said stabilizing antibody comprises at least one variable domain comprising a VH and VL pair selected from the group consisting of the VH of SEQ ID NO: 462 and the VL of SEQ ID NO: 1037, the VH of SEQ ID NO: 472 and the VL of SEQ ID NO: 1047, the VH of SEQ ID NO: 448 and the VL of SEQ ID NO: 1023, the VH of SEQ ID NO: 502 and the VL of SEQ ID NO: 1077, the VH of SEQ ID NO: 581 and the VL of SEQ ID NO: 1153, the VH of SEQ ID NO: 442 and the VL of SEQ ID NO: 1017, the VH of SEQ ID NO: 464 and the VL of SEQ ID NO: 1039, the VH of SEQ ID NO: 593 and the VL of SEQ ID NO: 1166, the VH of SEQ ID NO: 598 and the VL of SEQ ID NO:
1171, the VH of SEQ ID NO: 626 and the VL of SEQ ID NO: 1199 and the VH of SEQ ID NO: 645 and the VL of SEQ ID NO: 1218.
226. The method of claim 223, wherein said antibody is a releasing antibody and wherein contacting said biological system with said releasing antibody results in release of at least 5% of total GDF-8 mature growth factor in said biological system.
227. A method of treating a TGF- -related indication in a subject comprising contacting said subject with the antibody or antigen binding fragment thereof of any of claims 1-206 or 207-221.
228. The method of claim 227, wherein said TGF- -related indication comprises a muscle disorder and/or injury.
229. The method of claim 228, wherein said muscle disorder and/or injury comprises a muscle disorder selected from the group consisting of cachexia, inclusion body myositis (IBM) and sarcopenia.
230. The method of claim 227, wherein said TGF- -related indication comprises chronic obstructive pulmonary disease (COPD).
231. A recombinant antigen comprising a growth differentiation factor (GDF) prodomain fused to an Fc-fusion protein.
232. The recombinant antigen of claim 231, wherein said GDF is selected from the group consisting of GDF-8 and GDF-11.
233. The recombinant antigen of claim 231, wherein said Fc-fusion protein comprises the sequence
DKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWY VDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIE KTISKAKGQPREPQVYTLPPSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENN YKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG
K (SEQ ID NO: 222).
234. The recombinant antigen of claim 231, comprising a sequence selected from any of those presented in Table 12.
235. A recombinant antigen comprising a chimeric protein selected from any of those in Tables 20 and 21.
236. An isolated antibody or antigen binding fragment thereof that binds to a recombinant antigen of any of claims 231-235.
237. The isolated antibody or antigen binding fragment thereof of claim 236, wherein said recombinant antigen comprises latent GDF-11 growth factor prodomain complex (GPC).
238. A method of modulating growth factor activity in a biological system comprising contacting said biological system with the antibody or antigen binding fragment thereof of any of claims 1-206, 236 or 237.
239. The method of claim 238, wherein said growth factor activity comprises GDF-11 activity.
240. The method of claim 238 or 239, wherein said isolated antibody or antigen binding fragment thereof is a stabilizing antibody and wherein contacting said biological system with said stabilizing antibody results in inhibition of release of at least 5% of total GDF-11 mature growth factor in said biological system.
241. The method of claim 238, wherein said antibody is a releasing antibody and wherein contacting said biological system with said releasing antibody results in release of at least 5% of total GDF-11 mature growth factor in said biological system.
242. A method of treating a TGF- -related indication in a subject comprising contacting said subject with the antibody or antigen binding fragment thereof of any one of claims 1- 206, 236 or 237.
243. The method of claim 242, wherein said TGF- -related indication comprises a cardiovascular indication selected from the group consisting of cardiac hypertrophy, cardiac atrophy, atherosclerosis and restenosis.
244. The method of claim 242, wherein said TGF-P-related indication comprises a GDF- 11 -related indication.
245. The method of claim 244, wherein said GDF-11 -related indication comprises anemia and/or β-thalassemia.
246. A method of enhancing skeletal muscle rejuvenation, cerebral circulation,
neurogenesis and/or erythropoiesis comprising administering to a subject the antibody or antigen binding fragment thereof of any one of claims 1-206, 236 or 237.
247. The antibody or antigen binding fragment thereof of any one of claims 1-206, 236 or 237, wherein said antibody is a monoclonal antibody.
248. The antibody or antigen binding fragment thereof of either of claims 236 or 237, wherein said antibody comprises an IgG isotype.
249. The antibody of claim 247 or 248, wherein said antibody comprises an IgGl isotype.
250. The isolated antibody or antigen binding fragment thereof of any of claims 207-215, 236 or 237 comprising a bispecific antibody.
251. An assay comprising the use of one or more of the recombinant antigens of any of claims 231-235 and/or any of the antibody or antigen binding fragment thereof of any of claims 1-206, 207-221, 236, 237 or 246-249.
252. A method of developing an antibody comprising the use of the assay of claim 240.
253. A composition comprising the antibody or antigen binding fragment thereof of any of claims 1-206, 207-221, 236, 237 or 247-249 and an excipient.
254. A nucleic acid encoding the antibody or antigen binding fragment thereof of any of claims 1-206, 207-221, 236, 237 or 247-249.
255. A nucleic acid encoding the recombinant antigen of any of claims 231-235.
256. A vector comprising the nucleic acid of claims 254 or 255.
257. A cell comprising the nucleic acid of claims 254 or 255.
258. A kit comprising the composition of claim 256 and instructions for use thereof.
259. A method of developing an antibody comprising:
i. selecting a protein from the group consisting of any of those listed in Tables 4, 5, 9, 10, 20, 21, 24 and 25,
ii. subjecting said protein to structural analysis,
iii. identifying at least one epitope, and
iv. developing at least one antibody against said at least one epitope.
260. The method of claim 259, wherein said protein is subjected to prodomain convertase cleavage prior to structural analysis.
261. The method of claim 260, wherein the prodomain convertase cleaved protein is further subjected to cleavage with an enzyme selected from the group consisting of BMP-1, mammalian tolloid protein (mTLD), mammalian tolloid-like 1 (mTLLl) and mammalian tolloid-like 2 (mTLL2).
262. The method of any of claims 259-261, wherein said protein comprises proGDF-8 (SEQ ID NO: 5) or proGDF-11 (SEQ ID NO: 4).
263. A method of increasing muscle mass in a subject comprising contacting said subject with the antibody or antigen binding fragment thereof of any of claims 1-206 or 207-221.
264. The method of claim any one of claims 259-263, wherein said isolated antibody or antigen binding fragment thereof comprises at least one variable domain comprising a VH and VL pair selected from the group consisting of the VH of SEQ ID NO: 462 and the VL of SEQ ID NO: 1037, the VH of SEQ ID NO: 472 and the VL of SEQ ID NO: 1047, the VH of SEQ ID NO: 448 and the VL of SEQ ID NO: 1023, the VH of SEQ ID NO: 502 and the VL of SEQ ID NO: 1077, the VH of SEQ ID NO: 581 and the VL of SEQ ID NO: 1153, the VH of SEQ ID NO: 442 and the VL of SEQ ID NO: 1017, the VH of SEQ ID NO: 464 and the VL of SEQ ID NO: 1039, the VH of SEQ ID NO: 593 and the VL of SEQ ID NO: 1166, the VH of SEQ ID NO: 598 and the VL of SEQ ID NO: 1171, the VH of SEQ ID NO: 626 and the VL of SEQ ID NO: 1199 and the VH of SEQ ID NO: 645 and the VL of SEQ ID NO: 1218.
265. An inhibitor of at least one BMP-l/Tolloid-like proteinase (B/TP), wherein said inhibitor comprises a polypeptide.
266. The inhibitor of claim 265, wherein said inhibitor prevents cleavage of a latent GPC.
267. The inhibitor of claim 266, wherein said latent GPC is selected from the group consisting of latent GDF-8 and latent GDF-11.
268. The inhibitor of claim 267, wherein said inhibitor prevents cleavage of a BMP/Tolloid cleavage site on said latent GPC.
269. The inhibitor of claim 268, wherein said B/TP is selected from the group consisting of BMP-1, mammalian tolloid protein (mTLD), mammalian tolloid-like 1 (mTLLl), and mammalian tolloid-like 2 (mTLL2).
270. The inhibitor of any one of claims 265-269, comprising an inhibiting antibody.
271. The inhibiting antibody of claim 270, comprising an anti-primed complex antibody.
272. An antibody that binds at or within 10 amino acid residues of a proprotein convertase cleavage site of proMyostatin or latent Myostatin.
273 The antibody of claim 272, wherein the proprotein convertase cleavage site comprises the amino acid sequence (SEQ ID NO: 1667 or 1668).
274. An antibody that binds at or within 10 amino acid residues of a proprotein convertase docking site of proMyostatin or latent Myostatin.
275. The antibody of claim 274, wherein the proprotein convertase docking site comprises the amino acid sequence (SEQ ID NO: 1667 or 1668).
276. An antibody that binds at or within 10 amino acid residues of a tolloid protease cleavage site of proMyostatin or latent Myostatin.
277 The antibody of claim 272, wherein the tolloid protease cleavage site comprises the amino acid sequence (SEQ ID NO: 1665 or 1666).
278. An antibody that binds at or within 10 amino acid residues of a tolloid protease docking site of proMyostatin or latent Myostatin.
279. The antibody of claim 274, wherein the tolloid protease docking site comprises the amino acid sequence (SEQ ID NO: 1665 or 1666).
280. The antibody of any one of claims 272-279, wherein the antibody is the antibody of any one of claims 1-206, 207-221, 236, 237, or 247-249.
281. The antibody of any one of claims 272-280, wherein binding of the antibody to the proMyostatin or the latent Myostatin inhibits proteolytic cleavage of the proMyostatin or the latent Myostatin by a proprotein convertase or a tolloid protease.
282. An antibody that competes for binding to an epitope with any one of the antibodies of claims 1-206, 207-221, 236, 237, or 247-249.
283. An antibody that binds to the same epitope as any one of the antibodies of claims 1- 206, 207-221, 236, 237, or 247-249.
284. An antibody that competes for binding to an epitope of human proMyostatin or an epitope of human latent Myostatin with the antibody of any one of claims 1-206, 207-221, 236, 237, or 247-249.
285. The antibody of claim S3, wherein the antibody specifically binds to an epitope of human proMyostatin or human latent Myostatin at the same epitope as the antibody of any one of claims 1-206, 207-221, 236, 237, or 247-249.
286. The antibody of claims 282 or 283, wherein the antibody competes for binding to the epitope with an equilibrium dissociation constant (Kd) between the antibody and the epitope of less than 10"6 M.
287. The antibody of claim 286, wherein the Kd is in a range of 10"11 M to 10"6 M
288. A composition comprising the antibody of any one of claims 1-206, 207-221, 236, 237, 247-249, or 282-287 and a carrier.
289. The composition of claim 288, wherein the composition is a pharmaceutical composition comprising (i) a therapeutically effective amount of the antibody of any one of claims 1-206, 207-221, 236, 237, 247-249, or 282-287, and (ii) a pharmaceutically acceptable carrier.
290. The composition of claim 288 or 289 for use in preventing muscle wasting or increasing muscle mass, comprising a therapeutically effective amount of the antibody of any one of claims 1-206, 207-221, 236, 238, 247-249, or 282-287.
291. The composition of any one of claims 288 to 290, wherein the carrier is a
pharmaceutically acceptable carrier.
292. The composition of any one of claims 288 to 290, wherein the antibody and carrier are in a lyophilized form.
293. The composition of any one of claims 288 to 290, wherein the antibody and carrier are in solution.
294. The composition of any one of claims 288 to 290, wherein the antibody and carrier are frozen.
294. The composition of claim 294, wherein the antibody and carrier are frozen at a temperature less than or equal to -65°C.
296. The antibody of any one of claims 1-206, 207-221, 236, 237, 247-249, or 282-287, wherein the antibody is a sweeping antibody.
297. The antibody of any one of claims 1-206, 207-221, 236, 237, 247-249, or 282-285, wherein the antibody is a recycling antibody.
298. The antibody of claim 296 or 297, wherein the antibody comprises an Fc portion.
299. The antibody of any one of claims 296 to 298, wherein the antibody binds the neonatal Fc receptor FcRn.
300. The antibody of claim 298, wherein the Fc portion binds the neonatal Fc receptor FcRn.
301. The antibody of claim 299 or 300, wherein the antibody binds FcRn at a pH greater than 6.0.
302. The antibody of claim 301, wherein the antibody binds FcRn at a pH in a range from 7.0 to 7.5.
303. The antibody of claim 302, wherein the a Kd of binding of the antibody to the FcRN is in a range fromlO"3 M to 10"8 M.
304. The antibody of claim 302, wherein the a Kd of binding of the antibody to the FcRN is in a range fromlO"4 M to 10"8 M.
305. The antibody of claim 302, wherein the a Kd of binding of the antibody to the FcRN is in a range fromlO"5 M to 10"8 M.
306. The antibody of claim 302, wherein the a Kd of binding of the antibody to the FcRN is in a range fromlO"6 M to 10"8 M.
307. The antibody of any one of claims 296 to 306, wherein the antibody comprises a CDR-H3 amino acid sequence set forth in SEQ ID NO: 813, 804, 790, 762, 784, 770, 764, 986, 777, or 1006.
308. The antibody of any one of claims 296 to 307, wherein the antibody comprises a CDR-L3 amino acid sequence set forth in any one of SEQ ID NO: 1422, 1414, 1403, 1377, 1397, 1385, 1379, 1601, 1391, or 1621.
309. The antibody of any one of claims 296 to 308, wherein the antibody comprises a CDR-H1, a CDR-H2, and/or a CDR-H3 from the variable heavy chain (VH) amino acid sequence of clone BP2a-l, BP3a-2, BP5b-l, BP5b-4, BP6a-5, BP6a-6, BPl la-1, BP16b-4, BP18b-2 or BP22b-l as set forth in Table 14, and/or a CDR-Ll, a CDR-L2, and/or a CDR-L3 from the variable light chain (VL) amino acid sequence of clone BP2a-l, BP3a-2, BP5b-l, BP5b-4, BP6a-5, BP6a-6, BPl la-1, BP16b-4, BP18b-2 or BP22b-l as set forth in Table 16.
310. The antibody of any one of claims 296 to 309, wherein the antibody comprises a variable heavy chain amino acid sequence of clone BP2a-l, BP3a-2, BP5b-l, BP5b-4, BP6a- 5, BP6a-6, BPl la-1, BP16b-4, BP18b-2 or BP22b-las set forth in Table 14.
311. The antibody of any one of claims 296 to 310, wherein the antibody comprises the variable light chain amino acid sequence of clone BP2a-l, BP3a-2, BP5b-l, BP5b-4, BP6a-5, BP6a-6, BPl la-1, BP16b-4, BP18b-2 or BP22b-l as set forth in Table 16.
PCT/US2015/059515 2014-11-06 2015-11-06 Transforming growth factor-related antibodies and uses thereof WO2016073879A2 (en)

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US201462076230P 2014-11-06 2014-11-06
US62/076,230 2014-11-06
US201562100361P 2015-01-06 2015-01-06
US62/100,361 2015-01-06
US201562187348P 2015-07-01 2015-07-01
US62/187,348 2015-07-01

Publications (2)

Publication Number Publication Date
WO2016073879A2 true WO2016073879A2 (en) 2016-05-12
WO2016073879A3 WO2016073879A3 (en) 2016-06-23

Family

ID=55910042

Family Applications (2)

Application Number Title Priority Date Filing Date
PCT/US2015/059557 WO2016073906A2 (en) 2014-11-06 2015-11-06 Transforming growth factor-related immunoassays
PCT/US2015/059515 WO2016073879A2 (en) 2014-11-06 2015-11-06 Transforming growth factor-related antibodies and uses thereof

Family Applications Before (1)

Application Number Title Priority Date Filing Date
PCT/US2015/059557 WO2016073906A2 (en) 2014-11-06 2015-11-06 Transforming growth factor-related immunoassays

Country Status (1)

Country Link
WO (2) WO2016073906A2 (en)

Cited By (33)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017015622A3 (en) * 2015-07-22 2017-03-02 Scholar Rock, Inc Gdf11 binding proteins and uses thereof
WO2017104783A1 (en) * 2015-12-18 2017-06-22 Chugai Seiyaku Kabushiki Kaisha Anti-myostatin antibodies, polypeptides containing variant fc regions, and methods of use
US9828429B2 (en) 2007-09-26 2017-11-28 Chugai Seiyaku Kabushiki Kaisha Method of modifying isoelectric point of antibody via amino acid substitution in CDR
WO2017218592A1 (en) 2016-06-13 2017-12-21 Scholar Rock, Inc. Use of myostatin inhibitors and combination therapies
JP2017226690A (en) * 2016-06-17 2017-12-28 中外製薬株式会社 Anti-myostatin antibodies and methods of use
US9868948B2 (en) 2008-04-11 2018-01-16 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule capable of binding to two or more antigen molecules repeatedly
WO2018080812A1 (en) * 2016-10-30 2018-05-03 Henlix, Inc. Anti-pd-l1 antibodies and variants
US9969800B2 (en) 2015-02-05 2018-05-15 Chugai Seiyaku Kabushiki Kaisha IL-8 antibodies
US10000560B2 (en) 2014-12-19 2018-06-19 Chugai Seiyaku Kabushiki Kaisha Anti-myostatin antibodies, polypeptides containing variant Fc regions, and methods of use
WO2018129395A1 (en) * 2017-01-06 2018-07-12 Scholar Rock, Inc. Methods for treating metabolic diseases by inhibiting myostatin activation
US10307480B2 (en) 2014-11-06 2019-06-04 Scholar Rock, Inc. Anti-pro/latent-myostatin antibodies and uses thereof
CN110343172A (en) * 2018-04-04 2019-10-18 中国科学院微生物研究所 A kind of yellow fever virus human monoclonal antibody of high sensitivity and its application
WO2019210144A1 (en) * 2018-04-27 2019-10-31 Vanderbilt University Broadly neutralizing antibodies against hepatitis c virus
WO2020009938A1 (en) * 2018-07-03 2020-01-09 Catalent Pharma Solutions, Llc Multifunctional protein molecules comprising decorin and use thereof
JP2020502991A (en) * 2016-09-20 2020-01-30 ウーシー バイオロジクス アイルランド リミテッド New anti-PCSK9 antibody
US10751413B2 (en) 2015-09-15 2020-08-25 Scholar Rock, Inc. Anti-pro/latent-Myostatin antibodies and uses thereof
CN111662379A (en) * 2020-05-09 2020-09-15 江苏省疾病预防控制中心(江苏省公共卫生研究院) Antibody for resisting novel coronavirus, preparation method and application
WO2020264410A1 (en) * 2019-06-26 2020-12-30 Sensor-Kinesis Corporation Troponin t binding agents and uses thereof
US10882904B2 (en) 2016-01-08 2021-01-05 Scholar Rock, Inc. Methods for inhibiting myostatin activation by administering anti-pro/latent myostatin antibodies
US10919953B2 (en) 2012-08-24 2021-02-16 Chugai Seiyaku Kabushiki Kaisha FcgammaRIIB-specific Fc region variant
US11046784B2 (en) 2006-03-31 2021-06-29 Chugai Seiyaku Kabushiki Kaisha Methods for controlling blood pharmacokinetics of antibodies
US11053308B2 (en) 2016-08-05 2021-07-06 Chugai Seiyaku Kabushiki Kaisha Method for treating IL-8-related diseases
US11155611B2 (en) 2017-01-06 2021-10-26 Scholar Rock, Inc. Compositions and methods for making and using anti-myostatin antibodies
US11168128B2 (en) 2020-02-26 2021-11-09 Vir Biotechnology, Inc. Antibodies against SARS-CoV-2 and methods of using the same
US11236168B2 (en) 2012-08-24 2022-02-01 Chugai Seiyaku Kabushiki Kaisha Mouse FcγammaRII-specific Fc antibody
US11267868B2 (en) 2013-04-02 2022-03-08 Chugai Seiyaku Kabushiki Kaisha Fc region variant
US11359009B2 (en) 2015-12-25 2022-06-14 Chugai Seiyaku Kabushiki Kaisha Anti-myostatin antibodies and methods of use
US11401339B2 (en) 2018-08-23 2022-08-02 Seagen Inc. Anti-TIGIT antibodies
TWI782920B (en) * 2016-09-20 2022-11-11 中國大陸商上海藥明生物技術有限公司 Novel Anti-PCSK9 Antibody
CN115322257A (en) * 2021-08-16 2022-11-11 上海优替济生生物医药有限公司 BCMA (brain cell adhesion antigen) targeted antibody, chimeric antigen receptor and application thereof
WO2023009891A3 (en) * 2021-07-30 2023-04-13 Janssen Biotech, Inc. Materials and methods of making or using il-23r binding proteins
WO2023086336A3 (en) * 2021-11-09 2023-08-03 University Of Pittsburgh – Of The Commonwealth System Of Higher Education Molecules that bind to prostate specific membrane antigen
US11891434B2 (en) 2010-11-30 2024-02-06 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule capable of binding to plurality of antigen molecules repeatedly

Families Citing this family (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
BR112019000166A2 (en) 2016-07-06 2019-10-01 Celgene Corp antibodies with low immunogenicity and their uses
CN116082506A (en) * 2016-07-22 2023-05-09 丹娜法伯癌症研究所公司 Glucocorticoid-induced tumor necrosis factor receptor (GITR) antibodies and methods of use thereof
CA3049661A1 (en) * 2017-01-11 2018-07-19 The Board Of Trustees Of The Leland Stanford Junior University R-spondin (rspo) surrogate molecules
WO2019055399A1 (en) * 2017-09-12 2019-03-21 Igc Bio, Inc. Anti-vegf antibody
JP2024501581A (en) * 2020-12-08 2024-01-12 メモリアル スローン-ケタリング キャンサー センター Antibodies against galectin-3 and methods of using the same
EP4147716A1 (en) * 2021-09-10 2023-03-15 Samatva Research Corporation Anti-virus moiety immobilised in a matrix

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7393682B1 (en) * 1993-03-19 2008-07-01 The Johns Hopkins University School Of Medicine Polynucleotides encoding promyostatin polypeptides
JPH09507829A (en) * 1993-03-19 1997-08-12 ジョーンズ ホプキンス ユニバーシティー スクール オブ メディシン Growth differentiation factor-8
UA92504C2 (en) * 2005-10-12 2010-11-10 Эли Лилли Энд Компани Anti-myostatin monoclonal antibody
PE20091163A1 (en) * 2007-11-01 2009-08-09 Wyeth Corp ANTIBODIES FOR GDF8
EP2606066A1 (en) * 2010-08-16 2013-06-26 Amgen Inc. Antibodies that bind myostatin, compositions and methods
EP2981822B1 (en) * 2013-05-06 2020-09-02 Scholar Rock, Inc. Compositions and methods for growth factor modulation

Cited By (60)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11046784B2 (en) 2006-03-31 2021-06-29 Chugai Seiyaku Kabushiki Kaisha Methods for controlling blood pharmacokinetics of antibodies
US11248053B2 (en) 2007-09-26 2022-02-15 Chugai Seiyaku Kabushiki Kaisha Method of modifying isoelectric point of antibody via amino acid substitution in CDR
US9828429B2 (en) 2007-09-26 2017-11-28 Chugai Seiyaku Kabushiki Kaisha Method of modifying isoelectric point of antibody via amino acid substitution in CDR
US11371039B2 (en) 2008-04-11 2022-06-28 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule capable of binding to two or more antigen molecules repeatedly
US10472623B2 (en) 2008-04-11 2019-11-12 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule capable of binding two or more antigen molecules repeatedly
US9868948B2 (en) 2008-04-11 2018-01-16 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule capable of binding to two or more antigen molecules repeatedly
US9890377B2 (en) 2008-04-11 2018-02-13 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule capable of binding to two or more antigen molecules repeatedly
US11359194B2 (en) 2008-04-11 2022-06-14 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule capable of binding two or more antigen molecules repeatedly
US11891434B2 (en) 2010-11-30 2024-02-06 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule capable of binding to plurality of antigen molecules repeatedly
US11236168B2 (en) 2012-08-24 2022-02-01 Chugai Seiyaku Kabushiki Kaisha Mouse FcγammaRII-specific Fc antibody
US10919953B2 (en) 2012-08-24 2021-02-16 Chugai Seiyaku Kabushiki Kaisha FcgammaRIIB-specific Fc region variant
US11267868B2 (en) 2013-04-02 2022-03-08 Chugai Seiyaku Kabushiki Kaisha Fc region variant
US10307480B2 (en) 2014-11-06 2019-06-04 Scholar Rock, Inc. Anti-pro/latent-myostatin antibodies and uses thereof
US11135291B2 (en) 2014-11-06 2021-10-05 Scholar Rock, Inc. Methods for making and using anti-myostatin antibodies
US11925683B2 (en) 2014-11-06 2024-03-12 Scholar Rock, Inc. Compositions for making and using anti-Myostatin antibodies
US10000560B2 (en) 2014-12-19 2018-06-19 Chugai Seiyaku Kabushiki Kaisha Anti-myostatin antibodies, polypeptides containing variant Fc regions, and methods of use
US11454633B2 (en) 2014-12-19 2022-09-27 Chugai Seiyaku Kabushiki Kaisha Anti-myostatin antibodies, polypeptides containing variant Fc regions, and methods of use
US10738111B2 (en) 2014-12-19 2020-08-11 Chugai Seiyaku Kabushiki Kaisha Anti-myostatin antibodies, polypeptides containing variant Fc regions, and methods of use
US9969800B2 (en) 2015-02-05 2018-05-15 Chugai Seiyaku Kabushiki Kaisha IL-8 antibodies
US10519229B2 (en) 2015-02-05 2019-12-31 Chugai Seiyaku Kabushiki Kaisha Nucleic acids encoding IL-8 antibodies
US11180548B2 (en) 2015-02-05 2021-11-23 Chugai Seiyaku Kabushiki Kaisha Methods of neutralizing IL-8 biological activity
WO2017015622A3 (en) * 2015-07-22 2017-03-02 Scholar Rock, Inc Gdf11 binding proteins and uses thereof
US10751413B2 (en) 2015-09-15 2020-08-25 Scholar Rock, Inc. Anti-pro/latent-Myostatin antibodies and uses thereof
US11439704B2 (en) 2015-09-15 2022-09-13 Scholar Rock, Inc. Methods for using anti-myostatin antibodies
WO2017104783A1 (en) * 2015-12-18 2017-06-22 Chugai Seiyaku Kabushiki Kaisha Anti-myostatin antibodies, polypeptides containing variant fc regions, and methods of use
US11359009B2 (en) 2015-12-25 2022-06-14 Chugai Seiyaku Kabushiki Kaisha Anti-myostatin antibodies and methods of use
US10882904B2 (en) 2016-01-08 2021-01-05 Scholar Rock, Inc. Methods for inhibiting myostatin activation by administering anti-pro/latent myostatin antibodies
EP4218804A2 (en) 2016-06-13 2023-08-02 Scholar Rock, Inc. Use of myostatin inhibitors and combination therapies
EP3785728A2 (en) 2016-06-13 2021-03-03 Scholar Rock, Inc. Use of myostatin inhibitors and combination therapies
US10946036B2 (en) 2016-06-13 2021-03-16 Scholar Rock, Inc. Use of myostatin inhibitors and combination therapies
WO2017218592A1 (en) 2016-06-13 2017-12-21 Scholar Rock, Inc. Use of myostatin inhibitors and combination therapies
JP2022101540A (en) * 2016-06-17 2022-07-06 中外製薬株式会社 Anti-myostatin antibodies and methods of use
JP2017226690A (en) * 2016-06-17 2017-12-28 中外製薬株式会社 Anti-myostatin antibodies and methods of use
TWI611811B (en) * 2016-06-17 2018-01-21 中外製藥股份有限公司 Anti-myostatin antibodies and methods of use
CN109311969B (en) * 2016-06-17 2022-09-27 中外制药株式会社 Anti-myostatin antibodies and methods of use
CN109311969A (en) * 2016-06-17 2019-02-05 中外制药株式会社 Anti- myostatin antibodies and application method
US11053308B2 (en) 2016-08-05 2021-07-06 Chugai Seiyaku Kabushiki Kaisha Method for treating IL-8-related diseases
US11780912B2 (en) 2016-08-05 2023-10-10 Chugai Seiyaku Kabushiki Kaisha Composition for prophylaxis or treatment of IL-8 related diseases
JP2020502991A (en) * 2016-09-20 2020-01-30 ウーシー バイオロジクス アイルランド リミテッド New anti-PCSK9 antibody
TWI782920B (en) * 2016-09-20 2022-11-11 中國大陸商上海藥明生物技術有限公司 Novel Anti-PCSK9 Antibody
RU2770590C2 (en) * 2016-10-30 2022-04-18 Шанхай Хенлиус Байотек, Инк. Antibodies against pd-l1 and their options
WO2018080812A1 (en) * 2016-10-30 2018-05-03 Henlix, Inc. Anti-pd-l1 antibodies and variants
US11274155B2 (en) 2016-10-30 2022-03-15 Shanghai Henlius Biotech Inc. Anti-PD-L1 antibodies and variants
US11618786B2 (en) 2016-10-30 2023-04-04 Shanghai Henlius Biotech Inc. Anti-PD-L1 antibodies and variants
US11155611B2 (en) 2017-01-06 2021-10-26 Scholar Rock, Inc. Compositions and methods for making and using anti-myostatin antibodies
WO2018129395A1 (en) * 2017-01-06 2018-07-12 Scholar Rock, Inc. Methods for treating metabolic diseases by inhibiting myostatin activation
CN110343172A (en) * 2018-04-04 2019-10-18 中国科学院微生物研究所 A kind of yellow fever virus human monoclonal antibody of high sensitivity and its application
WO2019210144A1 (en) * 2018-04-27 2019-10-31 Vanderbilt University Broadly neutralizing antibodies against hepatitis c virus
US11377488B2 (en) 2018-07-03 2022-07-05 Catalent Pharma Solutions, Llc Multifunctional protein molecules comprising decorin and use thereof
WO2020009938A1 (en) * 2018-07-03 2020-01-09 Catalent Pharma Solutions, Llc Multifunctional protein molecules comprising decorin and use thereof
US11879009B2 (en) 2018-07-03 2024-01-23 Catalent Pharma Solutions, Llc Multifunctional protein molecules comprising decorin and use thereof
US11401339B2 (en) 2018-08-23 2022-08-02 Seagen Inc. Anti-TIGIT antibodies
WO2020264410A1 (en) * 2019-06-26 2020-12-30 Sensor-Kinesis Corporation Troponin t binding agents and uses thereof
US11479599B2 (en) 2020-02-26 2022-10-25 Vir Biotechnology, Inc. Antibodies against SARS-CoV-2 and methods of using the same
US11168128B2 (en) 2020-02-26 2021-11-09 Vir Biotechnology, Inc. Antibodies against SARS-CoV-2 and methods of using the same
CN111662379A (en) * 2020-05-09 2020-09-15 江苏省疾病预防控制中心(江苏省公共卫生研究院) Antibody for resisting novel coronavirus, preparation method and application
CN111662379B (en) * 2020-05-09 2021-03-02 江苏省疾病预防控制中心(江苏省公共卫生研究院) Antibody for resisting novel coronavirus, preparation method and application
WO2023009891A3 (en) * 2021-07-30 2023-04-13 Janssen Biotech, Inc. Materials and methods of making or using il-23r binding proteins
CN115322257A (en) * 2021-08-16 2022-11-11 上海优替济生生物医药有限公司 BCMA (brain cell adhesion antigen) targeted antibody, chimeric antigen receptor and application thereof
WO2023086336A3 (en) * 2021-11-09 2023-08-03 University Of Pittsburgh – Of The Commonwealth System Of Higher Education Molecules that bind to prostate specific membrane antigen

Also Published As

Publication number Publication date
WO2016073906A3 (en) 2016-07-21
WO2016073879A3 (en) 2016-06-23
WO2016073906A2 (en) 2016-05-12

Similar Documents

Publication Publication Date Title
US11827698B2 (en) Compositions and methods for growth factor modulation
WO2016073879A2 (en) Transforming growth factor-related antibodies and uses thereof
US20170073406A1 (en) Compositions and methods for growth factor modulation
US20180208648A1 (en) Gdf11 binding proteins and uses thereof
US9783610B2 (en) Anti-tumor endothelial marker-1 (TEM1) antibody variants and uses thereof

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 15858050

Country of ref document: EP

Kind code of ref document: A2

NENP Non-entry into the national phase in:

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 15858050

Country of ref document: EP

Kind code of ref document: A2