WO2025076507A1 - Engineered proteins and methods of use thereof - Google Patents
Engineered proteins and methods of use thereof Download PDFInfo
- Publication number
- WO2025076507A1 WO2025076507A1 PCT/US2024/050186 US2024050186W WO2025076507A1 WO 2025076507 A1 WO2025076507 A1 WO 2025076507A1 US 2024050186 W US2024050186 W US 2024050186W WO 2025076507 A1 WO2025076507 A1 WO 2025076507A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- antibody
- antibody fragment
- domain
- certain embodiments
- seq
- Prior art date
- Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
- Pending
Links
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P7/00—Drugs for disorders of the blood or the extracellular fluid
- A61P7/04—Antihaemorrhagics; Procoagulants; Haemostatic agents; Antifibrinolytic agents
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K14/00—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- C07K14/435—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- C07K14/745—Blood coagulation or fibrinolysis factors
- C07K14/755—Factors VIII, e.g. factor VIII C (AHF), factor VIII Ag (VWF)
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K16/00—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
- C07K16/18—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
- C07K16/36—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against blood coagulation factors
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2299/00—Coordinates from 3D structures of peptides, e.g. proteins or enzymes
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/20—Immunoglobulins specific features characterized by taxonomic origin
- C07K2317/24—Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/30—Immunoglobulins specific features characterized by aspects of specificity or valency
- C07K2317/32—Immunoglobulins specific features characterized by aspects of specificity or valency specific for a neo-epitope on a complex, e.g. antibody-antigen or ligand-receptor
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/30—Immunoglobulins specific features characterized by aspects of specificity or valency
- C07K2317/33—Crossreactivity, e.g. for species or epitope, or lack of said crossreactivity
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/50—Immunoglobulins specific features characterized by immunoglobulin fragments
- C07K2317/52—Constant or Fc region; Isotype
- C07K2317/524—CH2 domain
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/50—Immunoglobulins specific features characterized by immunoglobulin fragments
- C07K2317/52—Constant or Fc region; Isotype
- C07K2317/526—CH3 domain
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/60—Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments
- C07K2317/62—Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments comprising only variable region components
- C07K2317/622—Single chain antibody (scFv)
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/90—Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/90—Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
- C07K2317/92—Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/90—Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
- C07K2317/94—Stability, e.g. half-life, pH, temperature or enzyme-resistance
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2319/00—Fusion polypeptide
- C07K2319/31—Fusion polypeptide fusions, other than Fc, for prolonged plasma life, e.g. albumin
Definitions
- Emicizumab augments Factor X (FX) activation by activated Factor IX (FIXa) in the absence of activated Factor VIII (FVIIIa) by binding and approximating the substrate FX and protease FIXa. In doing so, it acts as a FVIIIa mimetic, use of which significantly reduces the bleeding episodes in hemophilia A patients.
- FX Factor X
- FIXa activated Factor IX
- FVIIIa activated Factor VIII
- Nucleic acid molecules encoding the antibody fragments of the instant invention may also be used in gene therapy.
- the antibody fragments activate FX in the presence of FX and FIXa.
- the antibody fragments bind FIXa.
- the antibody fragments comprise at least one complementarity determining region (CDR) of the FIXa binding domain of a FVIIIa mimetic bispecific antibody (e.g., emicizumab or mim8).
- CDR complementarity determining region
- the antibody fragments comprise all three CDRs of the heavy chain variable domain of the Factor IX binding domain of the FVIIIa mimetic bispecific antibody and/or all three CDRs of the light chain variable domain of the Factor IX binding domain of the FVIIIa mimetic bispecific antibody.
- the antibody fragments comprise single domain antibodies.
- the antibody fragments comprise single chain variable fragment.
- the antibody fragments comprise a first and a second single chain variable fragment.
- the first and second single chain variable fragment bind FIXa.
- the first single chain variable fragment binds FIXa and the second single chain variable fragment binds Factor X.
- Figure 1B provides a schematic of an scFv (VH9VL) derived from linking the IXa specific variable heavy chain (VH9) of emicizumab with a variable light chain via a flexible linker.
- Figure 1C provides a schematic of a nanobody that uses a common nanobody scaffold with CDR1 and CDR2 from VH9.
- Figure 1D provides a schematic of scFv (VH9VL C2) derived from fusing FVa C2-domain at the C-terminus of V H 9V L .
- Figure 1E provides a schematic of a scFv-FVC2 fusion construct in an AAV expression vector, wherein the FVC2 is optional.
- Figure 1F provides a schematic of a scFv-CH2-CH3-FVC2 fusion construct in an AAV expression vector, wherein the FVC2 is optional.
- Figure 1G provides a schematic of a homodimerized scFv-Fc fusion construct.
- Figure 1H provides a schematic of a homodimerized scFv-Fc-FV-C2 fusion construct with membrane binding properties.
- Figure 1I provides a schematic of a biparatropic construct derived from scFv-scFv fusion of mim8 and emicizumab.
- Figure 1J provides a schematic showing biparatropic constructs fused with FV C2-domain.
- the cofactor mimetic properties of the antibody fragments of the instant invention enable the use of the antibody fragments in both nonfactor replacement therapy (e.g., as an injectable) as well as a new transgene templates for gene therapy for the correction of a hemostasis disorder such as a bleeding disorder (e.g., hemophilia, particularly hemophilia A).
- a hemostasis disorder such as a bleeding disorder (e.g., hemophilia, particularly hemophilia A).
- scFv based transgenes are less immunogenic as compared to FVIII and also lack undesirable Fc-mediated effector functions.
- Membrane dependent assembly of FVIIIa-FIXa-FX is a requirement for the intrinsic tenase (Xase) activity.
- the antibody fragments of the instant invention may be targeted to neonatal Fc receptors to increase the circulating half-life.
- the antibody fragments of the instant invention further comprise albumin (e.g., linked (e.g., directly or via a linker) to a terminus) (e.g., human albumin).
- the antibody fragments of the instant invention further comprise an antibody, antibody fragment, or nanobody that binds albumin (e.g., linked (e.g., directly or via a linker) to a terminus) (e.g., an anti-human albumin antibody, antibody fragment, or nanobody).
- the simplified single chain constructs provided herein avoid the burden of productive assembly from a combination of three separate polypeptide chains, as in a functional bispecific antibody.
- the antibody fragments provided herein e.g., the scFv V H 9V L , nanobodies, or fusion derivatives
- ADCC antibody-dependent cellular cytotoxicity
- CDC complement-dependent cytotoxicity
- the FVIIIa mimetic bispecific antibody is mim8 (denecimig; UNII: EUV85RR8DJ (drugs.ncats.io/drug/ EUV85RR8DJ); Novo Nordisk; Kjellev et al., Blood (2019) 134 (Supplement_1): 96; Ostergaard et al., Blood (2021) 138(14):1258-1268).
- the FVIII mimetic bispecific antibody is described in WO2018/047813, U.S. Patent Application Publication No. 2020/0148787 or U.S. Patent No. 10,759,870 or 11,150,254 (each incorporated herein by reference).
- the antibody fragment comprises one, two, three, four, five, or all six of the CDRs of the FIX/FIXa binding domain of a FVIIIa mimetic bispecific antibody (e.g., emicizumab or mim8).
- the antibody fragment comprises a heavy chain variable domain comprising one, two, or all three of the CDRs of the heavy chain of the FIX/FIXa binding domain of a FVIIIa mimetic bispecific antibody (e.g., emicizumab or mim8).
- the antibody fragment comprises one, two, three, four, five, or all six of: YYDIQ (SEQ ID NO: 1), SISPSGQSTYYRREVKG (SEQ ID NO: 2), RTGREYGGGWYFDY (SEQ ID NO: 3), KASRNIERQLA (SEQ ID NO: 4), QASRKES (SEQ ID NO: 5), and QQYSDPPLT (SEQ ID NO: 6).
- the antibody fragment comprises a heavy chain variable domain comprising one, two, or all three of: YYDIQ (SEQ ID NO: 1), SISPSGQSTYYRREVKG (SEQ ID NO: 2), RTGREYGGGWYFDY (SEQ ID NO: 3).
- the antibody fragment comprises a heavy chain variable domain comprises YYDIQ (SEQ ID NO: 1) and SISPSGQSTYYRREVKG (SEQ ID NO: 2).
- the antibody fragment comprises a heavy chain variable domain comprising: EVQLVESGGGLVQPGRSLRLSCAASGFTFHDYAMHWVRQVPGKGLEWVSG ISWRGDIGGYVKSVKGRFTISRDNAKNSLYLQMNSLRAEDTALYYCVKSY GSGSFYNAFDSWGQGTLVTVSSA (SEQ ID NO: 17).
- the antibody fragment comprises a light chain variable domain comprising: DIQMTQSPSTLSASVGDRVTITCRASQSISSWLAWYQQKPGKAPKFLIYK ASKLERGTPSRFSGSGDGTEFSLTISSLQPDDFATYYCLEYSSYIRTFGQ GTKVEIKRTV (SEQ ID NO: 18).
- the scFv (V H 9V L ) comprises: QVQLVESGGGLVQPGGSLRLSCAASGFTFSYYDIQWVRQAPGKGLEWVSS ISPSGQSTYYRREVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCARRT GREYGGGWYFDYWGQGTLVTVSSAGGGGSGGGGSGGGGSDIQMTQSPSSL SASVGDRVTITCKASRNIERQLAWYQQKPGQAPELLIYQASRKESGVPDR FSGSRYGTDFTLTISSLQPEDIATYYCQQYSDPPLTFGGGTKVEIKRTV (SEQ ID NO: 15).
- the scFv comprises an amino acid sequence having at least 90%, 95%, 97%, 99%, or 100% homology or identity with any of the sequences provided above (e.g., any of SEQ ID NOs: 1-6 and 13-16).
- the scFv comprises one, two, three, four, five, or all six of: GFTFHDY (SEQ ID NO: 7), SWRGDIG (SEQ ID NO: 8), SYGSGSFYNAFDS (SEQ ID NO: 9), RASQSISSWLA (SEQ ID NO: 10), KASKLER (SEQ ID NO: 11), and LEYSSYIRT (SEQ ID NO: 12).
- the antibody fragment (His-tagged V H 9V L C2) comprises: QVQLVESGGGLVQPGGSLRLSCAASGFTFSYYDIQWVRQAPGKGLEWVSS ISPSGQSTYYRREVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCARRT GREYGGGWYFDYWGQGTLVTVSSAGGGGSGGGGSGGGGSDIQMTQSPSSL SASVGDRVTITCKASRNIERQLAWYQQKPGQAPELLIYQASRKESGVPDR FSGSRYGTDFTLTISSLQPEDIATYYCQQYSDPPLTFGGGTKVEIKRTVG GGGSGGGGSGGGGSVNGCSTPLGMENGKIENKQITASSFKKSWWGDYWEP FRARLNAQGRVNAWQAKANNNKQWLEIDLLKIKKITAIITQGCKSLSSEM YVKSYTIHYSEQGVEWKPYRLKSSMVDKIFEGNTNTKGHVKNFF
- the lactadherin C2 domain is encoded by the sequence: ctgaacggatgcgccaatcccctgggcctgaagaataacagcatccctgacaagcag atcacggcctccagcagctacaagacctggggcttgcatctcttcagctggaacccc tcctatgcacggctggacaagcagggcaacttcaacgcctgggttgcggggagctac ggtaacgatcagtggctgcaggtggacctgggctcctcgaaggaggtgacaggcatc atcacccagggggccccgtaactttggctctgtccagtttgtggcatcctacaaggtt gcctacagtaatgtggctgcgtaacttttgg
- the antibody fragment comprises: QVQLVESGGGLVQPGGSLRLSCAASGFTFSYYDIQWVRQAPGKGLEWVSS ISPSGQSTYYRREVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCARRT GREYGGGWYFDYWGQGTLVTVSSAGGGGSGGGGSGGGGSDIQMTQSPSSL SASVGDRVTITCKASRNIERQLAWYQQKPGQAPELLIYQASRKESGVPDR FSGSRYGTDFTLTISSLQPEDIATYYCQQYSDPPLTFGGGTKVEIKRTVG GGGSGGGGSGGGGSLIMDRDCRMPMGLSTGIISDSQIKASEFLGYWEPRL ARLNNGGSYNAWSVEKLAAEFASKPWIQVDMQKEVIITGIQTQGAKHYLK SCYTTEFYVAYSSNQINWQIFKGNSTRNVMYFNGNSDASTIK
- the antibody fragment comprises: EVQLVESGGGLVQPGRSLRLSCAASGFTFHDYAMHWVRQVPGKGLEWVSG ISWRGDIGGYVKSVKGRFTISRDNAKNSLYLQMNSLRAEDTALYYCVKSY GSGSFYNAFDSWGQGTLVTVSSAGGGGSGGGGSGGSDIQMTQSPSTLS ASVGDRVTITCRASQSISSWLAWYQQKPGKAPKFLIYKASKLERGTPSRF SGDGTEFSLTISSLQPDDFATYYCLEYSSYIRTFGQGTKVEIKRTVGG GGSGGGGSGGGGSVNGCSTPLGMENGKIENKQITASSFKKSWWGDYWEPF RARLNAQGRVNAWQAKANNNKQWLEIDLLKIKKITAIITQGCKSLSSEMY VKSYTIHYSEQGVEWKPYRLKSSMVDKIFEGNTNTKGHVKNFFNPPIISR
- the antibody fragment comprises an amino acid sequence having at least 90%, 95%, 97%, 99%, or 100% homology or identity with any of the sequences provided above (e.g., any of SEQ ID NOs: 24, 25, 28-31, 57, and 58).
- the antibody fragment comprises a Protein C tag, particularly a tag recognized by the commercially available HPC4 antibody (e.g., Cell Signaling Technology, Inc., Danvers, MA).
- the tag comprises the amino acid sequence EDQVDPRLIDGK (SEQ ID NO: 59).
- the tag is at the C-terminus.
- the tag is at the C-terminus of the antibody fragment.
- the antibody fragment comprises: QVQLVESGGGLVQPGGSLRLSCAASGFTFSYYDIQWVRQAPGKGLEWVSS ISPSGQSTYYRREVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCARRT GREYGGGWYFDYWGQGTLVTVSSAGGGGSGGGGSGGGGSDIQMTQSPSSL SASVGDRVTITCKASRNIERQLAWYQQKPGQAPELLIYQASRKESGVPDR FSGSRYGTDFTLTISSLQPEDIATYYCQQYSDPPLTFGGGTKVEIKRTVG GGGSGGGGSGGGGSAPEFLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDV SQEDPEVQFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNG KEYKCKVSNKGLPSSIEKTISKAKGQPREPQVYTLPPSQKEMTK
- the antibody fragment (V H 9V L C H 2C H 3C2) comprises: QVQLVESGGGLVQPGGSLRLSCAASGFTFSYYDIQWVRQAPGKGLEWVSS ISPSGQSTYYRREVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCARRT GREYGGGWYFDYWGQGTLVTVSSAGGGGSGGGGSGGGGSDIQMTQSPSSL SASVGDRVTITCKASRNIERQLAWYQQKPGQAPELLIYQASRKESGVPDR FSGSRYGTDFTLTISSLQPEDIATYYCQQYSDPPLTFGGGTKVEIKRTVG GGGSGGGGSGGGGSAPEFLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDV SQEDPEVQFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNG KEYKCKVSNKGLPSSIEKTISKAKGQPREPQVYTLPPS
- one or both scFv of the bispecific scFv-scFv fusion are from a FVIIIa mimetic bispecific antibody such as emicizumab or mim8.
- one scFv is from a first FVIIIa mimetic bispecific antibody (e.g., emicizumab (e.g., FIXa binding scFv or FX binding scFv)) and the other scFv is from a second FVIIIa mimetic bispecific antibody (e.g., mim8 (e.g., FIXa binding scFv or FX binding scFv)).
- the scFv-scFv (His-tagged Emi V H 9V L -V H 10V L ) comprises: QVQLVESGGGLVQPGGSLRLSCAASGFTFSYYDIQWVRQAPGKGLEWVSS ISPSGQSTYYRREVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCARRT GREYGGGWYFDYWGQGTLVTVSSAGGGGSGGGGSGGSDIQMTQSPSSL SASVGDRVTITCKASRNIERQLAWYQQKPGQAPELLIYQASRKESGVPDR FSGSRYGTDFTLTISSLQPEDIATYYCQQYSDPPLTFGGGTKVEIKRTVG GGGSGGGGSGGGGSQVQLVQSGSELKKPGASVKVSCKASGYTFTDNNMDW VRQAPGQGLEWMGDINTRSGGSIYNEEFQDRVIMTVDKSTDTAYMELSSL RSEDTATYHCARRKSYG
- the scFv-scFv (Duo - mim8 FIXa scFv and emicizumab FIXa scFv) comprises: EVQLVESGGGLVQPGRSLRLSCAASGFTFHDYAMHWVRQVPGKGLEWVSG ISWRGDIGGYVKSVKGRFTISRDNAKNSLYLQMNSLRAEDTALYYCVKSY GSGSFYNAFDSWGQGTLVTVSSAGGGGSGGGGSGGSDIQMTQSPSTLS ASVGDRVTITCRASQSISSWLAWYQQKPGKAPKFLIYKASKLERGTPSRF SGDGTEFSLTISSLQPDDFATYYCLEYSSYIRTFGQGTKVEIKRTVGG GGSGGGGSGGGGSQVQLVESGGGLVQPGGSLRLSCAASGFTFSYYDIQWV RQAPGKGLEWVSSISPSGQSTYYRREVKGRFTISRDNSKNTLYL
- the scFv-scFv (His-tagged Duo – mim8 FIXa scFv and emicizumab FIXa scFv) comprises: EVQLVESGGGLVQPGRSLRLSCAASGFTFHDYAMHWVRQVPGKGLEWVSG ISWRGDIGGYVKSVKGRFTISRDNAKNSLYLQMNSLRAEDTALYYCVKSY GSGSFYNAFDSWGQGTLVTVSSAGGGGSGGGGSGGSDIQMTQSPSTLS ASVGDRVTITCRASQSISSWLAWYQQKPGKAPKFLIYKASKLERGTPSRF SGDGTEFSLTISSLQPDDFATYYCLEYSSYIRTFGQGTKVEIKRTVGG GGSGGGGSGGGGSQVQLVESGGGLVQPGGSLRLSCAASGFTFSYYDIQWV RQAPGKGLEWVSSISPSGQSTYYRREVKGRFTISRDN
- the nucleic acid molecules comprise a nucleotide sequence having at least 90%, 95%, 97%, 99%, or 100% homology or identity with any of the sequences provided above (e.g., any of SEQ ID NOs: 45-53, 61, and 66) or a nucleotide sequence encoding any of the amino sequences provided herein.
- Compositions comprising a nucleic acid molecule of the instant invention and a carrier such as a pharmaceutically acceptable carrier are also encompassed by the instant invention.
- the composition comprises at least one nucleic acid molecule and at least one carrier (e.g., a pharmaceutically acceptable carrier).
- the nucleic acid molecules are contained within a lipid nanoparticle.
- the antibody and antibody fragments of the invention may be prepared using a variety of methods known in the art.
- the antibodies or antibody fragments of the instant invention may be prepared by inserting amino acid sequences into a backbone of an antibody or antibody fragment construct (e.g., an antibody framework), particularly a human construct/framework.
- an antibody or antibody fragment construct e.g., an antibody framework
- the variable light domain and/or variable heavy domain of the antibodies or antibody fragments of the instant invention or the CDRs contained therein may be inserted into another antibody construct or framework, particularly a human construct/framework.
- Methods for recombinantly producing antibodies are well-known in the art. Indeed, commercial vectors for certain antibody and antibody fragment constructs are available.
- the antibody molecules are produced by expression of recombinant antibody or antibody fragments in host cells.
- the method comprises administering to a subject in need thereof a therapeutically effective amount of an antibody or antibody fragment of the instant invention.
- the subject is a human.
- the subject is a canine.
- Antibody or antibody fragments may be administered alone or in combination with other agents known to modulate hemostasis.
- the antibody or antibody fragment of the instant invention may be administered as part of a composition with a pharmaceutically acceptable carrier.
- the compositions of the instant invention may be conveniently formulated for administration with any carrier, particularly any pharmaceutically acceptable carrier(s). Except insofar as any conventional carrier is incompatible with the agents to be administered (e.g., antibody or antibody fragment or nucleic acid molecules encoding the same), its use in the pharmaceutical composition is contemplated.
- compositions can include diluents of various buffer content (e.g., saline, Tris HCl, acetate, phosphate), pH and ionic strength; and additives such as detergents and solubilizing agents (e.g., polysorbate 80), anti-oxidants (e.g., ascorbic acid, sodium metabisulfite), preservatives (e.g., benzyl alcohol) and bulking substances (e.g., lactose, mannitol).
- the preparation can be formulated with a buffer containing salts, such as NaCl, CaCl 2 , and amino acids, such as glycine and/or lysine, and in a pH range from 6 to 8.
- the pharmaceutical composition of the present invention can be prepared, for example, in liquid form, deep-frozen, or can be in dried powder form (e.g., lyophilized). In a particular embodiment, when the preparation is stored in lyophilized form, it may be dissolved into a visually clear solution using an appropriate reconstitution solution prior to administration.
- the compositions described herein will generally be administered to a patient as a pharmaceutical preparation.
- patient or “subject”, as used herein, refers to human or animal subjects (e.g., canine).
- the compositions of the instant invention may be employed therapeutically, under the guidance of a physician.
- Appropriate concentrations for alleviation of a particular pathological condition may be determined by dosage concentration curve calculations, as known in the art.
- the appropriate dosage unit for the administration of the composition may be determined by evaluating the toxicity of the molecules or cells in animal models.
- concentrations of active agents in pharmaceutical preparations may be administered to mice or other animal models, and the minimal and maximal dosages may be determined based on the beneficial results and side effects observed as a result of the treatment.
- Appropriate dosage unit may also be determined by assessing the efficacy of the treatment in combination with other standard drugs.
- the dosage units of the compositions of the instant invention may be determined individually or in combination with each treatment according to the effect detected.
- nucleic acid molecules may be maintained in any convenient vector, particularly an expression vector.
- Nucleic acids of the present invention may also be maintained as RNA or DNA in any convenient vector or cloning vector.
- the nucleic acids may be maintained in a vector suitable for expression in mammalian cells, particularly human cells.
- Vectors such as those described above comprise the regulatory elements necessary for expression of the DNA in the host cell positioned in such a manner as to permit expression of the DNA in the host or target cell (e.g., hepatocyte).
- regulatory elements required for expression include, but are not limited to, promoter sequences, transcription initiation sequences, and enhancer sequences.
- the nucleic acid molecules of the instant invention will be administered to a subject in a composition comprising at least one carrier.
- the nucleic acid molecules are contained within lipid nanoparticles.
- the nucleic acid molecules are contained within a viral vector (e.g., AAV vector).
- the carrier is a pharmaceutically acceptable carrier for injection.
- the nucleic acid molecules of the instant invention are administered to a subject in a cell (e.g., a hepatocyte) wherein the cell may be maintained in a composition comprising at least one carrier.
- an expression construct may further comprise regulatory elements which serve to drive expression in a particular cell (e.g., hepatocyte) or tissue type (e.g., liver). Such regulatory elements are known to those of skill in the art.
- tissue specific regulatory elements in the expression constructs of the present invention provides for at least partial tissue tropism for expression.
- the vector is typically engineered from the virus by deleting the rep and cap genes and replacing these with the transgene of interest under the control of a specific promoter.
- the upper size limit of the sequence that can be inserted between the two inverted terminal repeats (ITRs) is about 4.7 kb.
- AAV vectors may be purified by repeated CsCl density gradient centrifugation and the titer of purified vectors determined by quantitative dot-blot hybridization.
- the expression vectors of the present invention may be incorporated into pharmaceutical compositions that may be delivered to a subject, so as to allow production of a biologically active protein (e.g., an antibody or antibody fragment).
- a biologically active protein e.g., an antibody or antibody fragment
- pharmaceutical compositions comprising sufficient genetic material to enable a recipient to produce a therapeutically effective amount of an antibody or antibody fragment can influence hemostasis in the subject.
- compositions may be administered alone or in combination with at least one other agent, such as a stabilizing compound, which may be administered in any sterile, biocompatible pharmaceutical carrier, including, but not limited to, saline, buffered saline, dextrose, and water.
- a stabilizing compound which may be administered in any sterile, biocompatible pharmaceutical carrier, including, but not limited to, saline, buffered saline, dextrose, and water.
- the compositions may be administered to a patient alone, or in combination with other agents (e.g., co-factors) which influence hemostasis.
- the pharmaceutical compositions also contain a pharmaceutically acceptable excipient/carrier.
- excipients include any pharmaceutical agent that does not itself induce an immune response harmful to the individual receiving the composition, and which may be administered without undue toxicity.
- Pharmaceutically acceptable excipients include, but are not limited to, liquids such as water, saline, glycerol, sugars and ethanol.
- Pharmaceutically acceptable salts can also be included therein, for example, mineral acid salts such as hydrochlorides, hydrobromides, phosphates, sulfates, and the like; and the salts of organic acids such as acetates, propionates, malonates, benzoates, and the like.
- auxiliary substances such as wetting or emulsifying agents, pH buffering substances, and the like, may be present in such vehicles.
- a modified FVIIIa mimetic bispecific antibody wherein said modified FVIIIa mimetic bispecific antibody comprises a FVIIIa mimetic bispecific antibody and a membrane binding domain. 22. The modified FVIIIa mimetic bispecific antibody of paragraph 31, wherein said membrane binding domain comprises the Factor V C2 domain. 23. The modified FVIIIa mimetic bispecific antibody of paragraph 21, wherein said membrane binding domain comprises the lactadherin C2 domain. 24. The modified FVIIIa mimetic bispecific antibody of any one of paragraphs 21-23, wherein said FVIIIa mimetic bispecific antibody is emicizumab or mim8. 25.
- a composition comprising an antibody fragment of any one of paragraphs 1-20 or an antibody of any one of paragraphs 21-24 and a carrier.
- 26. A nucleic acid molecule encoding an antibody fragment of any one of paragraphs 1-20 or an antibody of any one of paragraphs 21-24.
- 27. A vector comprising the nucleic acid molecule of paragraph 26.
- 28. The vector of paragraph 27, which is a viral vector.
- 29. The vector of paragraph 28, wherein said viral vector is an adeno- associated virus vector.
- a composition comprising a nucleic acid molecule or vector of any one of paragraphs 26-29 and a carrier. 31.
- antibody or antibody molecule contemplates intact immunoglobulin molecules, immunologically active portions of an immunoglobulin molecule (e.g., antigen- binding fragment), and fusions of immunologically active portions of an immunoglobulin molecule.
- immunologically specific refers to proteins/polypeptides, particularly antibodies, that bind to one or more epitopes of a protein or compound of interest, but which do not substantially recognize and bind other molecules in a sample containing a mixed population of antigenic biological molecules.
- subject refers to an animal, particularly a mammal, particularly a human.
- a “therapeutically effective amount” of a compound or a pharmaceutical composition refers to an amount effective to prevent, inhibit, treat, or lessen the symptoms of a particular disorder or disease.
- the treatment of a disease or disorder herein may refer to curing, relieving, and/or preventing the disease or disorder, the symptom(s) of it, or the predisposition towards it.
- the term “therapeutic agent” refers to a chemical compound or biological molecule including, without limitation, nucleic acids, peptides, proteins, and antibodies that can be used to treat a condition, disease, or disorder or reduce the symptoms of the condition, disease, or disorder.
- isolated refers to the separation of a compound from other components present during its production or from its natural environment.
- the scFv, V H 9V L can only bind FIXa and imparted a modest cofactor mimetic activity, as compared to the full bispecific antibody, emicizumab.
- VH9VL was used as a template to introduce membrane binding properties.
- Figs 3A and 3B indicate that nearly three- to fourfold excess emicizumab is required to achieve thrombin levels equivalent to those of the fusion variant, Emi VH9VL FVC2, in the thrombin generation assays triggered by low tissue factor (0.1 pM).
- the endogenous thrombin potential (ETP) a parameter that reflects total thrombin generation quantitatively, was comparable in both emicizumab and VH9VLC2.
- Potent cofactor mimetic activity of VH9VLC2 observed in TGA was also reflected in FXa generation assay in a purified system.
- a single chain scFv, V H 9V L derived from emicizumab retains significant cofactor mimetic activity.
- the engineered scFv with a Factor V C2 domain fusion, VH9VLC2 demonstrates that the addition of a membrane binding feature to the FIXa arm of the emicizumab surpasses the mimetic activity achieved by a bispecific antibody that exploits both bridging and allostery to be an effective cofactor. This would also incorporate a built-in regulatability feature because membrane dependent mimetics could be regulated by an inhibitory antibody against FV C2-domain.
- VH9VLC2 was about 3-4 fold higher as that of emicizumab in low tissue factor triggered thrombin generation assay.
- the activated partial thromboplastin time (aPTT) was used herein to evaluate the intrinsic coagulation pathway (Langdell, et al. (1953) J. Lab. Clin. Med., 41(4):637-47).
- This one-stage assay is the most commonly used method for testing FVIII or FIX activities. Accordingly, this assay is very useful to determine the FVIII equivalency of FVIII mimetics when FVIII deficient plasma is used and supplemented either with FVIII or FVIII mimetic.
- Emi V H 9V L FVC2 HPC4 achieved equivalent cofactor activity of FVIII (B-domain deleted) at a less than two-fold higher molar concentration (Fig. 8). This would translate to a nearly tenfold higher cofactor mimetic activity as compared to emicizumab in a thrombin generation assay with low tissue factor as a trigger.
Landscapes
- Health & Medical Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Organic Chemistry (AREA)
- Life Sciences & Earth Sciences (AREA)
- General Health & Medical Sciences (AREA)
- Hematology (AREA)
- Medicinal Chemistry (AREA)
- Biophysics (AREA)
- Genetics & Genomics (AREA)
- Biochemistry (AREA)
- Molecular Biology (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Engineering & Computer Science (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Zoology (AREA)
- Gastroenterology & Hepatology (AREA)
- Immunology (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Diabetes (AREA)
- Toxicology (AREA)
- General Chemical & Material Sciences (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Pharmacology & Pharmacy (AREA)
- Animal Behavior & Ethology (AREA)
- Public Health (AREA)
- Veterinary Medicine (AREA)
- Peptides Or Proteins (AREA)
Abstract
Compositions and methods for modulating hemostasis and treating hemophilia A are disclosed.
Description
ENGINEERED PROTEINS AND METHODS OF USE THEREOF By Shekhar Kumar This application claims priority under 35 U.S.C. §119(e) to U.S. Provisional Patent Application No. 63/588,537, filed October 6, 2023, and U.S. Provisional Patent Application No. 63/561,872, filed March 6, 2024. The foregoing applications are incorporated by reference herein. This invention was made with government support under grant number HL139420 awarded by the National Institutes of Health. The government has certain rights in the invention. FIELD OF THE INVENTION This invention relates generally to the fields of medicine and hematology. Specifically, the invention provides novel compositions and methods for treating hemophilia A. BACKGROUND OF THE INVENTION Emicizumab is a bispecific humanized antibody that is currently a leading treatment for people with hemophilia A (EP 2644698; Kitazawa, et al. (2017) Thromb. Haemost., 117(7):1348-57; Kitazawa, et al. (2012) Nat. Med., 18:1570- 1574; Shima, et al. (2016) N. Engl. J. Med., 374(21):2044-53). Emicizumab augments Factor X (FX) activation by activated Factor IX (FIXa) in the absence of activated Factor VIII (FVIIIa) by binding and approximating the substrate FX and protease FIXa. In doing so, it acts as a FVIIIa mimetic, use of which significantly reduces the bleeding episodes in hemophilia A patients. The mechanistic explanations of this effect is that emicizumab binds to FIXa and the substrate FX simultaneously, thus promoting colocalization of the enzyme-substrate complex. However, unlike FVIIIa, FVIIIa mimetic bispecific antibodies (such as emicizumab, and Mim8) do not directly bind to procoagulant membranes, which likely contributes to their significantly lower cofactor activity on a molar basis compared to FVIIIa. Additionally, the absence of membrane binding may prevent these mimetics from participating in the membrane-dependent regulation that is crucial for clotting reactions. The rate of FX activation is greatly augmented in the bispecific format in
which the FVIIIa mimetic bispecific antibodies engage both FIXa and FX. While emicizumab is a significant improvement over frequent intravenous injection of Factor VIII (FVIII), the therapeutic dose of emicizumab only brings patients to a FVIII equivalency of 10-20% of normal (Shima et. al. (2016) N. Engl. J. Med. 374, 2044-2053). To achieve long-term therapy, several adeno-associated viral (AAV) vector gene therapy trials for hemophilia A are currently ongoing and one product (Roctavian™) that delivers a B-domain deleted variant of FVIII (BDD FVIII) was recently licensed. However, unexplained exponential year-over-year declines in expression (Samelson-Jones, et al. (2023) Ann. Rev. Med., 74:1:231-247) have raised doubts over long-term efficacy and durability of these therapies. While durability is likely mediated by multifactorial processes, one reason for the loss of durability in hemophilia A gene therapy may emanate from a hyperimmuonogenic nature of FVIII that leads to transgene depletion/rejection or possibly the activation of stress response mechanism leading to exponential drop of FVIII antigen level in the treated group. Therefore, there is an obvious need for improved therapeutic methods for hemophilia A both in injectable form and through gene therapy. SUMMARY OF THE INVENTION In accordance with one aspect of the instant invention, isolated antibody fragments which possess FVIIIa mimetic activity are provided. The antibody fragments of the instant invention may be used as an injectable. Nucleic acid molecules encoding the antibody fragments of the instant invention may also be used in gene therapy. In certain embodiments, the antibody fragments activate FX in the presence of FX and FIXa. In certain embodiments, the antibody fragments bind FIXa. In certain embodiments, the antibody fragments comprise at least one complementarity determining region (CDR) of the FIXa binding domain of a FVIIIa mimetic bispecific antibody (e.g., emicizumab or mim8). In certain embodiments, the antibody fragments comprise all three CDRs of the heavy chain variable domain of the Factor IX binding domain of the FVIIIa mimetic bispecific antibody and/or all three CDRs of the light chain variable domain of the Factor IX binding domain of the FVIIIa mimetic bispecific antibody. In certain embodiments, the antibody fragments comprise single domain antibodies. In certain embodiments, the antibody fragments comprise single chain variable fragment. In certain embodiments, the antibody
fragments comprise a first and a second single chain variable fragment. In certain embodiments, the first and second single chain variable fragment bind FIXa. In certain embodiments, the first single chain variable fragment binds FIXa and the second single chain variable fragment binds Factor X. In certain embodiments, the antibody fragments further comprise a membrane binding domain (e.g., Factor V C2 domain and/or lactadherin C2). In certain embodiments, the antibody fragments further comprise a Protein C tag. In certain embodiments, the antibody fragments further comprise a cleavage site such as a thrombin cleavage site or at least a fragment of the Factor IX activation peptide. In certain embodiments, the antibody fragments further comprise a dimerization domain (e.g., CH2 and CH3 domains or Fc domain). In certain embodiments, the antibody fragments further comprise albumin. Compositions comprising an antibody fragment of the instant invention and a carrier are also provided. In accordance with another aspect of the instant invention, antibodies which are modified FVIIIa mimetic bispecific antibodies are provided. In certain embodiments, the antibody comprises a FVIIIa mimetic bispecific antibody (e.g., emicizumab or mim8) and a membrane binding domain (e.g., Factor V C2 domain and/or lactadherin C2). In certain embodiments, the antibody further comprises albumin. In accordance with another aspect of the instant invention, isolated nucleic acid molecules encoding an antibody or antibody fragment of the instant invention are also provided. In certain embodiments, the nucleic acid molecule is contained within a vector such as a viral vector (e.g., an adeno-associated virus vector). Compositions comprising a nucleic acid molecule of the instant invention and a carrier are also provided. In accordance with another aspect of the instant invention, methods of treating and/or preventing a hemostasis disorder (e.g., hemophilia such as hemophilia A) in a subject in need thereof are provided. The instant invention also encompasses methods of increasing blood coagulation or decreasing clotting time of blood are provided. In certain embodiments, the methods comprise administering to the subject an antibody or antibody fragment of the instant invention to a subject and/or administering a nucleic acid molecule encoding an antibody or antibody fragment of the instant invention to the subject. In certain embodiments, the methods comprise contacting blood and/or plasma with an antibody or antibody fragment of the instant invention
and/or contacting blood and/or plasma with a nucleic acid molecule encoding an antibody or antibody fragment of the instant invention. BRIEF DESCRIPTION OF THE DRAWINGS Figure 1A provides a schematic showing emicizumab, a bispecific antibody that has two unique heavy chains paired with two identical light chains. The FIXa specific arm (VH9; left) binds specifically to FIXa while FX specific arm (VH10; right) binds specifically to FX. These two specificities colocalize FIXa and FX, which leads to the activation of FX. Figure 1B provides a schematic of an scFv (VH9VL) derived from linking the IXa specific variable heavy chain (VH9) of emicizumab with a variable light chain via a flexible linker. Figure 1C provides a schematic of a nanobody that uses a common nanobody scaffold with CDR1 and CDR2 from VH9. Figure 1D provides a schematic of scFv (VH9VL C2) derived from fusing FVa C2-domain at the C-terminus of VH9VL. Figure 1E provides a schematic of a scFv-FVC2 fusion construct in an AAV expression vector, wherein the FVC2 is optional. Figure 1F provides a schematic of a scFv-CH2-CH3-FVC2 fusion construct in an AAV expression vector, wherein the FVC2 is optional. Figure 1G provides a schematic of a homodimerized scFv-Fc fusion construct. Figure 1H provides a schematic of a homodimerized scFv-Fc-FV-C2 fusion construct with membrane binding properties. Figure 1I provides a schematic of a biparatropic construct derived from scFv-scFv fusion of mim8 and emicizumab. Figure 1J provides a schematic showing biparatropic constructs fused with FV C2-domain. Figure 1K provides a schematic showing Emi VH9VL FVC2 HPC4, derived from fusing FVa C2-domain at the C-terminus of emicizumab derived VH9VL. HPC4 tag is fused at the C-terminus of the FV C2 domain. Figure 1L provides a schematic showing Emi VH9VL Lac C2, derived from fusing lactadherin C2-domain at the C-terminus of emicizumab derived VH9VL. Figure 1M provides a schematic showing Emi VH9VL T FVC2 HPC4, derived from fusing FVa C2-domain at the C-terminus of emicizumab derived VH9VL. HPC4 tag is fused at the end of FV C2 domain. A short thrombin (IIa) cleavage sequence (asterisk) is inserted between the scFv and the FV C2 domain. Figure 1N provides a schematic showing Emi VH9VL T Lac C2 HPC4, derived from fusing lactadherin C2-domain at the C-terminus of emicizumab derived VH9VL. HPC4 tag is fused at the end of lactadherin C2 domain. A short thrombin (IIa) cleavage sequence (asterisk) is inserted between the scFv and the lactadherin C2
domain. Figure 1O provides a schematic showing albumin fused Emi VH9VL FVC2 variant, derived from emicizumab FIX arm derived scFv, VH9VL. Asterisk indicates FIX activation peptide. Figure 2 provides a graph of an activated partial thromboplastin time (aPTT) assay using platelet-poor hemophilia A plasma. The emicizumab-derived fusion mimetic variant, VH9VL FVC2, demonstrates superior mimetic activity compared to the scFv VH9VL or emicizumab. At 30% of normal FVIII equivalency (0.3 nM concentration of FVIII), the fusion mimetic demonstrates ~22 fold greater activity over the scFv VH9VL and nearly 10-fold greater activity than emicizumab. For comparison, the B-domain deleted FVIII (BDD FVIII) is also included. Figures 3A and 3B provide graphs illustrating peak thrombin (Fig. 3A) and endogenous thrombin potential (ETP) (Fig. 3B) for emicizumab and the fusion variant, Emi VH9VL FVC2. The results indicate that nearly three- to fourfold excess emicizumab is required to achieve thrombin levels equivalent to those of the fusion variant, Emi VH9VL FVC2, in thrombin generation assays triggered by low tissue factor (0.1 pM). PNP: pooled normal plasma. HA: Hemophilia A plasma/FVIII- deficient plasma. Figure 4 provides a graph of thrombin generation curves in FXIa triggered (0.1 nM) platelet-poor, severe congenital HA plasma supplemented with various concentrations of emicizumab (right) or the fusion variant Emi VH9VL FVC2 (left). For comparison, NPP and HA plasma without any mimetic are also included. Both mimetics show a robust thrombin generation in the thrombin generation assays triggered by Factor XIa (0.1 nM). Figure 5 provides a graph of light scattering measurements of synthetic membrane (phospholipid vesicles, PC:PS::75:25) binding by the scFv VH9VL-FIXa complex or the Emi VH9VL FVC2-FIXa complex. The FV C2 domain in Emi VH9VL FVC2 colocalizes the mimetic-FIXa complex at the membrane surface whereas the scFv VH9VL-FIXa complex shows no significant membrane localization. Figure 6 provides a graph of FX (100 nM) activation by FIXa (50 nM) in the presence of varying concentrations of VH9VLC2 or emicizumab and 417 µM PC:PS vesicles. Figure 7 provides a graph of FX (100 nM) activation by FIXa (20 nM) in the presence of varying concentrations of E9scFv and a fixed concentration (25 nM) of VH9VLC2 and 417 µM PC:PS vesicles.
Figure 8 provides a graph of the cofactor activity in an aPTT assay for varying concentrations of Emi VH9VL FVC2 HPC4 and BDD-FVIII. Less than a two-fold molar excess of Emi VH9VL FVC2 HPC4 is required to achieve equivalent cofactor activity in an aPTT assay. Figure 9 provides a graph of the cofactor activity in an aPTT assay for varying concentrations of Emi VH9VL Lac C2 His and BDD-FVIII. About two-fold molar excess of Emi VH9VL Lac C2 His is required to achieve equivalent cofactor activity in an aPTT assay. Figure 10 provides a graph showing that the mim8 FIXa arm derived variant, mim8 VH9VL, when fused with FVC2-domain shows a several fold enhanced cofactor mimetic activity in the aPTT assay with FVIII-deficient plasma. Figure 11 shows that emicizumab FIXa arm derived variant, Emi VH9VL FVC2 His, when fused to human albumin retains similar cofactor activity as Emi VH9VL FVC2 His without albumin in an aPTT assay with FVIII-deficient plasma. Figure 12A provides a graph of aPTT assays using canine FVIII-deficient plasma demonstrating the hemostatic potential of membrane-dependent mimetics derived from the FIXa binding arm of emicizumab and mim8. Notably, mim8 VH9VL FVC2 exhibits several fold enhanced cofactor mimetic activity compared to both emicizumab and the emicizumab-derived membrane binding mimetic Emi VH9VL FVC2. Figure 12B provides an alignment of a segment of human (SEQ ID NO: 67) and canine (SEQ ID NO: 68) FIX amino acid sequences. The FIX residues (VDRATCLRSTKFT (SEQ ID NO: 69), 163-175 in chymotrypsin numbering, or 371-383 in human FIX numbering are highlighted) which make contact with the mim8 FIXa binding arm are conserved in both human and canine. Figure 13 provides a graph showing aPTT assays using FVIII-deficient plasma and plasma with FVIII inhibitors. The results show that the emicizumab-derived fusion variant Emi VH9VL FVC2 maintains its mimetic activity in FVIII inhibitor plasma. DETAILED DESCRIPTION OF THE INVENTION Herein, antibody fragments which possess FVIIIa mimetic activity are provided. The antibody fragments described herein demonstrate a previously unanticipated property of emicizumab, a bispecific antibody that is currently in use for hemophilia A treatment (Fig. 1A). Distinct from the bridging mechanism
observed with the full-length bispecific antibody, a monospecific anti-FIXa one-arm antibody derived from emicizumab possesses minor FIXa stimulatory activity (Ostergaard, et al. (2021) Blood 138:1258-1268). Without the anti-FX arm of the FVIIIa mimetic, it was previously understood that the bridging mechanism could not occur and a solo anti-FIXa antibody would be ineffective. The present invention has unexpectedly shown that a single chain variable fragment (scFv; Hu, et al. (2005) J. Biotech., 120(1):38-45) - VH9VL (Fig. 1B) - designed by linking the Factor IX/IXa (FIX/IXa) specific variable heavy chain (VH) to the variable light chain (VL) is sufficient by itself to act as a cofactor mimetic for FIX/IXa to activate FX in FVIII deficient plasma. Surprisingly, it is also demonstrated herein that the VH9 complementarity determining regions (CDRs; particularly CDR1 and CDR2), which confer binding to FIX/IXa, show cofactor mimetic activity towards FIX/IXa when grafted in a stable nanobody framework (HamersCasterman, et al. (1993) Nature 363:446-448; Muyldermans, S. (2013) Annu. Rev. Biochem., 82:775-797; Vincke, et al. (2009) J. Biol. Chem., 284(5):3273-3284; Figure 1C). The cofactor mimetic activity shown by VH9VL in the absence of a FX/Xa specific arm confirms a mechanism that is distinct from the bridging mechanism of the bispecific antibody emicizumab. Without being bound by theory, scFvs derived from the FIXa specific arm of emicizumab act as cofactor for FIXa whose activity is not dependent on interaction with FX. The cofactor mimetic properties of the antibody fragments of the instant invention enable the use of the antibody fragments in both nonfactor replacement therapy (e.g., as an injectable) as well as a new transgene templates for gene therapy for the correction of a hemostasis disorder such as a bleeding disorder (e.g., hemophilia, particularly hemophilia A). Notably, scFv based transgenes are less immunogenic as compared to FVIII and also lack undesirable Fc-mediated effector functions. Membrane dependent assembly of FVIIIa-FIXa-FX is a requirement for the intrinsic tenase (Xase) activity. In contrast, emicizumab lacks any membrane binding ability itself and exhibits significant FXa generation even in the absence of membranes. This major distinction likely allows FVIIIa to function at sub-nanomolar concentrations to greatly accelerate the activation of FX while only much lower enhancements of rate are achieved at several 100-fold higher concentrations of emicizumab. C2- and C1-domains in FVIIIa and FVa are known to impart the membrane binding abilities to these cofactors (Nicolaes, et al. (2000) Blood Coagul.
Fibrinolysis 11:89-100; Kim, et al. (2000) Biochemistry 39:1951-1958). The C2- domain of FVa was chosen instead of FVIIIa to avoid interference from FVIIIa C2- domain specific inhibitory antibodies often present in a majority of patients to be treated. Thus, a new antibody fragment was synthesized - VH9VLC2 (Fig. 1D) – which is a chimera of VH9VL (although other antibody fragments such as a nanobody could be used) with Factor V C2-domian (FVC2) at the C-terminus, which imparted a regulatory feature in fusion protein by coupling its function to a membrane surface, particularly a phosphatidylserine rich membrane surface. The ability to bind membrane was determined to significantly augments the cofactor mimetic activity of fusion proteins by several orders of magnitude. Specifically, VH9VLC2 showed ~22- fold higher cofactor mimetic activity as compared to VH9VL and the activity of VH9VLC2 was, remarkably, about 10-fold higher than emicizumab. The FVC2- domain dependence can also be regulated by using an inhibitory antibody fragment that specifically binds to the C2-domain, thereby disrupting the membrane binding. Superior effects were also observed with the lactadherin C2 domain. The ability to bind membrane can also be imparted by fusing other membrane binding domains such as lactadherin C2 domain or Gla domains of vitamin K-dependent coagulation proteins. Notably, the membrane binding domains can also be added to the bispecific mimetic antibodies (e.g., emicizumab, mim8) to further amplify function and more closely resemble FVIIIa in both activity and membrane dependent regulation. Moreover, the addition of constant region 2 (CH2) and constant region 3 (CH3) of an antibody (e.g., emicizumab) to the antibody fragments of the instant invention leads to their homodimerization, thereby increasing their mimetic activity and circulating half-life, such as when expressed in vivo for gene therapy. Homodimerized scFv-Fc fusion derivatives typically have a longer half-life as compared to scFvs. In certain embodiments, the antibody fragments of the instant invention may be targeted to neonatal Fc receptors to increase the circulating half-life. In certain embodiments, the antibody fragments of the instant invention further comprise albumin (e.g., linked (e.g., directly or via a linker) to a terminus) (e.g., human albumin). In certain embodiments, the antibody fragments of the instant invention further comprise an antibody, antibody fragment, or nanobody that binds albumin (e.g., linked (e.g., directly or via a linker) to a terminus) (e.g., an anti-human albumin antibody, antibody fragment, or nanobody). In certain embodiments, the
antibody fragments of the instant invention further comprise an Fc region (e.g., linked (e.g., directly or via a linker) to a terminus). The use of a smaller and potentially nonimmunogenic antibody fragments of the instant invention that have cofactor mimetic activities will avoid issues related to using FVIII as a transgene in hemophilia A gene therapy. The smaller antibody fragments will show durability and long-term efficacy, as observed in the case of hemophilia B gene therapy, because of less misfolding in the endoplasmic reticulum (ER) leading to a less ER stress. The simplified single chain constructs provided herein (e.g., Figure 1E and 1F) described herein avoid the burden of productive assembly from a combination of three separate polypeptide chains, as in a functional bispecific antibody. The antibody fragments provided herein (e.g., the scFv VH9VL, nanobodies, or fusion derivatives) provide a single chain cofactor species that lack Fc- mediated effector functions including antibody-dependent cellular cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC). An scFv-Fc fusion construct (Figure 1G), after being translated, will form a homodimer before being secreted from the cell, thereby increasing the cofactor activity of the scFv-Fc fusion construct via avidity effect by bringing two FIXa binding sites together. The homodimerization will also lead to the lower clearance and an enhanced half-life of the scFv-Fc constructs, therefore requiring less vector dosage to achieve a therapeutic level of in vivo expression. Furthermore, an scFv-Fc-FVC2 fusion product (Figure 1H) would also enable the membrane binding ability and, therefore, further enhance FX activation ability and regulatability. The sustained expression of Adeno Associated Virus (AAV) based antibody fragments of the instant invention will provide a circulating concentration of FVIIIa mimetic to restore hemostasis (Li, et al. (2020) Nat. Rev. Genet., 21(4):255-72). Alternatively, nucleic acid molecules encoding the antibody fragments of the instant invention can be delivered by any method, including, for example, lipid nanoparticles or other viral vectors. While the present invention is exemplified with emicizumab, other FVIIIa mimetic bispecific antibodies or other anti-FIXa/FIX antibodies may be utilized. For example, the FIXa/FIX specific arm of mim8, particularly the variable domains or CDRs, can be utilized in the antibody constructs of the instant invention. The FIXa binding arm of mim8 binds to the catalytic domain of FIXa (Ostergaard, et al. (2021) Blood 138(14):1258-1268). In contrast, the FIXa binding arm of emicizumab binds to the EGF1 domain of FIXa. Since both mim8 and emicizumab FIXa binding arms
retain a significant cofactor mimetic activity, their mimetic activity can be combined (e.g., synergistically) in a single construct by fusing scFvs derived from mim8 and emicizumab (Fig. 1I). Such a biparatropic scFv-scFv fusion can mimic more closely the interaction of FVIIIa and FIXa, as FVIIIa is known to make extended contacts with FIXa that involves catalytic domain as well as other surfaces of FIXa. Furthermore, an scFv-scFv-FVC2 fusion product (Fig. 1J) would also enable the membrane binding ability and further enhance FX activation ability and regulatability. In accordance with one aspect of the instant invention, antibody fragments are provided. In certain embodiments, the antibody fragment is an antigen binding fragment. Antibody fragments include, without limitation, immunoglobulin fragments including, without limitation: nanobody or single domain (Dab), Fab, Fab', F(ab')2, and F(v); and fusions (e.g., via a linker) of these immunoglobulin fragments including, without limitation: scFv, scFv2, scFv-Fc, minibody, diabody, triabody, and tetrabody. In certain embodiments, the antibody fragment is mono-specific. In certain embodiments, the antibody fragment is not a whole antibody. In certain embodiments, the antibody fragment lacks an Fc domain. In certain embodiments, the antibody fragment is immunologically specific for Factor IX/Factor IXa (e.g., anti-FIXa antibody). In certain embodiments, the antibody fragment does not specifically bind FX. In certain embodiments, the antibody fragment comprises at least one, two, three, four, five, or all six complementarity-determining regions (CDRs) of an anti-FIX/FIXa antibody. In certain embodiments, the antibody fragment comprises at least one, two, three, four, five, or all six complementarity- determining regions (CDRs) of the Factor IX binding domain of a FVIIIa mimetic bispecific antibody (e.g., as determined by IMGT, Chothia, Kabat, Martin (e.g., enhanced Chothia) or AHo numbering scheme, particularly Kabat). In certain embodiments, the antibody fragment comprises at least one, two, or three CDRs, particularly at least CDR1 and CDR2, of the heavy chain variable domain of the Factor IX/IXa binding domain of a FVIIIa mimetic bispecific antibody. In certain embodiments, the antibody fragment comprises the heavy chain variable domain and/or light chain variable domain of the Factor IX binding domain of a FVIIIa mimetic bispecific antibody. In certain embodiments, the FVIIIa mimetic bispecific antibody is emicizumab (ACE910; HEMLIBRA®; Genentech; DrugBank Accession Number DB13923; Lenting, et al. Blood (2017) 130(23):2463-2468; Walsh, et al., J. Managed Care Med., 22(2):65-69). In certain embodiments, the FVIIIa mimetic
bispecific antibody is mim8 (denecimig; UNII: EUV85RR8DJ (drugs.ncats.io/drug/ EUV85RR8DJ); Novo Nordisk; Kjellev et al., Blood (2019) 134 (Supplement_1): 96; Ostergaard et al., Blood (2021) 138(14):1258-1268). In certain embodiments, the FVIII mimetic bispecific antibody is described in WO2018/047813, U.S. Patent Application Publication No. 2020/0148787 or U.S. Patent No. 10,759,870 or 11,150,254 (each incorporated herein by reference). In certain embodiments, the antibody fragment comprises one, two, three, four, five, or all six of the CDRs of the FIX/FIXa binding domain of a FVIIIa mimetic bispecific antibody (e.g., emicizumab or mim8). In certain embodiments, the antibody fragment comprises a heavy chain variable domain comprising one, two, or all three of the CDRs of the heavy chain of the FIX/FIXa binding domain of a FVIIIa mimetic bispecific antibody (e.g., emicizumab or mim8). In certain embodiments, the antibody fragment comprises a heavy chain variable domain comprising CDR1 and CDR2 of the CDRs of the heavy chain of the FIX/FIXa binding domain of a FVIIIa mimetic bispecific antibody (e.g., emicizumab or mim8). In certain embodiments, the antibody fragment comprises a light chain variable domain comprising one, two, or all three of the CDRs of the heavy chain of the FIX/FIXa binding domain of a FVIIIa mimetic bispecific antibody (e.g., emicizumab or mim8). In certain embodiments, the antibody fragment comprises a heavy chain variable domain comprising the heavy chain of the FIX/FIXa binding domain of a FVIIIa mimetic bispecific antibody (e.g., emicizumab or mim8) and/or the light chain of the FIX/FIXa binding domain of a FVIIIa mimetic bispecific antibody (e.g., emicizumab or mim8). In certain embodiments, the antibody fragment comprises one, two, three, four, five, or all six of: YYDIQ (SEQ ID NO: 1), SISPSGQSTYYRREVKG (SEQ ID NO: 2), RTGREYGGGWYFDY (SEQ ID NO: 3), KASRNIERQLA (SEQ ID NO: 4), QASRKES (SEQ ID NO: 5), and QQYSDPPLT (SEQ ID NO: 6). In certain embodiments, the antibody fragment comprises a heavy chain variable domain comprising one, two, or all three of: YYDIQ (SEQ ID NO: 1), SISPSGQSTYYRREVKG (SEQ ID NO: 2), RTGREYGGGWYFDY (SEQ ID NO: 3) and/or a light chain variable domain comprising one, two, or all three of: KASRNIERQLA (SEQ ID NO: 4), QASRKES (SEQ ID NO: 5), and QQYSDPPLT (SEQ ID NO: 6). In certain embodiments, the antibody fragment comprises a heavy chain variable domain comprising one, two, or all three of: YYDIQ (SEQ ID NO: 1), SISPSGQSTYYRREVKG (SEQ ID NO: 2), RTGREYGGGWYFDY (SEQ ID NO:
3). In certain embodiments, the antibody fragment comprises a heavy chain variable domain comprises YYDIQ (SEQ ID NO: 1) and SISPSGQSTYYRREVKG (SEQ ID NO: 2). In certain embodiments, the antibody fragment comprises a heavy chain variable domain comprising: QVQLVESGGGLVQPGGSLRLSCAASGFTFSYYDIQWVRQAPGKGLEWVSS ISPSGQSTYYRREVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCARRT GREYGGGWYFDYWGQGTLVTVSSA (SEQ ID NO: 13). In certain embodiments, the antibody fragment comprises a light chain variable domain comprising: DIQMTQSPSSLSASVGDRVTITCKASRNIERQLAWYQQKPGQAPELLIYQ ASRKESGVPDRFSGSRYGTDFTLTISSLQPEDIATYYCQQYSDPPLTFGG GTKVEIKRTV (SEQ ID NO: 14). In certain embodiments, the antibody fragment comprises an amino acid sequence having at least 90%, 95%, 97%, 99%, or 100% homology or identity with any of the sequences provided above (e.g., any of SEQ ID NOs: 1-6, 13, and 14). In certain embodiments, the antibody fragment comprises one, two, three, four, five, or all six of: GFTFHDY (SEQ ID NO: 7), SWRGDIG (SEQ ID NO: 8), SYGSGSFYNAFDS (SEQ ID NO: 9), RASQSISSWLA (SEQ ID NO: 10), KASKLER (SEQ ID NO: 11), and LEYSSYIRT (SEQ ID NO: 12). In certain embodiments, the antibody fragment comprises a heavy chain variable domain comprising one, two, or all three of: GFTFHDY (SEQ ID NO: 7), SWRGDIG (SEQ ID NO: 8), SYGSGSFYNAFDS (SEQ ID NO: 9) and/or a light chain variable domain comprising one, two, or all three of: RASQSISSWLA (SEQ ID NO: 10), KASKLER (SEQ ID NO: 11), and LEYSSYIRT (SEQ ID NO: 12). In certain embodiments, the antibody fragment comprises a heavy chain variable domain comprising one, two, or all three of: GFTFHDY (SEQ ID NO: 7), SWRGDIG (SEQ ID NO: 8), SYGSGSFYNAFDS (SEQ ID NO: 9). In certain embodiments, the antibody fragment comprises GFTFHDY (SEQ ID NO: 7) and SWRGDIG (SEQ ID NO: 8). In certain embodiments, the antibody fragment comprises a heavy chain variable domain comprising: EVQLVESGGGLVQPGRSLRLSCAASGFTFHDYAMHWVRQVPGKGLEWVSG ISWRGDIGGYVKSVKGRFTISRDNAKNSLYLQMNSLRAEDTALYYCVKSY GSGSFYNAFDSWGQGTLVTVSSA (SEQ ID NO: 17). In certain embodiments, the antibody fragment comprises a light chain variable domain comprising:
DIQMTQSPSTLSASVGDRVTITCRASQSISSWLAWYQQKPGKAPKFLIYK ASKLERGTPSRFSGSGDGTEFSLTISSLQPDDFATYYCLEYSSYIRTFGQ GTKVEIKRTV (SEQ ID NO: 18). In certain embodiments, the antibody fragment comprises an amino acid sequence having at least 90%, 95%, 97%, 99%, or 100% homology or identity with any of the sequences provided above (e.g., any of SEQ ID NOs: 7-12, 17 and 18). In certain embodiments, the antibody fragment is a nanobody or single domain antibody. In certain embodiments, the nanobody or single domain antibody comprises one, two or three CDR from the heavy chain variable domain. In certain embodiments, the nanobody or single domain antibody comprises one, two, or all three of: YYDIQ (SEQ ID NO: 1), SISPSGQSTYYRREVKG (SEQ ID NO: 2), RTGREYGGGWYFDY (SEQ ID NO: 3). In certain embodiments, the nanobody or single domain antibody comprises YYDIQ (SEQ ID NO: 1) and SISPSGQSTYYRREVKG (SEQ ID NO: 2). In certain embodiments, the nanobody or single domain antibody comprises one, two, or all three of: GFTFHDY (SEQ ID NO: 7), SWRGDIG (SEQ ID NO: 8), SYGSGSFYNAFDS (SEQ ID NO: 9). In certain embodiments, the nanobody or single domain antibody comprises GFTFHDY (SEQ ID NO: 7) and SWRGDIG (SEQ ID NO: 8). In certain embodiments, the nanobody or single domain antibody comprises an amino acid sequence having at least 90%, 95%, 97%, 99%, or 100% homology or identity with any of the sequences provided above (e.g., any of SEQ ID NOs: 1-3 and 7-9). In certain embodiments, the antibody fragment comprises an scFv. In certain embodiments, the scFv comprises a heavy chain variable domain joined to a light chain variable domain, optionally via a linker. In certain embodiments, the linker is a peptide or an amino acid sequence (e.g., about 1 to about 25 amino acids, about 10 to about 25 amino acids, about 15 to about 20 amino acids, about 10 to about 15 amino acids, about 13 to about 18 amino acids, or about 15 amino acids). In certain embodiments, the linker comprises the sequence (GGGGS)x (SEQ ID NO: 21), wherein x is 1-5, particularly 3-5, particularly 3. In certain embodiments, the scFv comprises one, two, three, four, five, or all six of: YYDIQ (SEQ ID NO: 1), SISPSGQSTYYRREVKG (SEQ ID NO: 2), RTGREYGGGWYFDY (SEQ ID NO: 3), KASRNIERQLA (SEQ ID NO: 4), QASRKES (SEQ ID NO: 5), and QQYSDPPLT (SEQ ID NO: 6). In certain embodiments, the scFv comprises a heavy chain variable domain comprising one, two, or all three of: YYDIQ (SEQ ID NO: 1),
SISPSGQSTYYRREVKG (SEQ ID NO: 2), RTGREYGGGWYFDY (SEQ ID NO: 3) and/or a light chain variable domain comprising one, two, or all three of: KASRNIERQLA (SEQ ID NO: 4), QASRKES (SEQ ID NO: 5), and QQYSDPPLT (SEQ ID NO: 6). In certain embodiments, the scFv comprises a heavy chain variable domain comprising: QVQLVESGGGLVQPGGSLRLSCAASGFTFSYYDIQWVRQAPGKGLEWVSS ISPSGQSTYYRREVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCARRT GREYGGGWYFDYWGQGTLVTVSSA (SEQ ID NO: 13). In certain embodiments, the scFv comprises a light chain variable domain comprising: DIQMTQSPSSLSASVGDRVTITCKASRNIERQLAWYQQKPGQAPELLIYQ ASRKESGVPDRFSGSRYGTDFTLTISSLQPEDIATYYCQQYSDPPLTFGG GTKVEIKRTV (SEQ ID NO: 14). In certain embodiments, the scFv (VH9VL) comprises: QVQLVESGGGLVQPGGSLRLSCAASGFTFSYYDIQWVRQAPGKGLEWVSS ISPSGQSTYYRREVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCARRT GREYGGGWYFDYWGQGTLVTVSSAGGGGSGGGGSGGGGSDIQMTQSPSSL SASVGDRVTITCKASRNIERQLAWYQQKPGQAPELLIYQASRKESGVPDR FSGSRYGTDFTLTISSLQPEDIATYYCQQYSDPPLTFGGGTKVEIKRTV (SEQ ID NO: 15). In certain embodiments, the scFv (His-tagged VH9VL) comprises: QVQLVESGGGLVQPGGSLRLSCAASGFTFSYYDIQWVRQAPGKGLEWVSS ISPSGQSTYYRREVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCARRT GREYGGGWYFDYWGQGTLVTVSSAGGGGSGGGGSGGGGSDIQMTQSPSSL SASVGDRVTITCKASRNIERQLAWYQQKPGQAPELLIYQASRKESGVPDR FSGSRYGTDFTLTISSLQPEDIATYYCQQYSDPPLTFGGGTKVEIKRTVH HHHHH (SEQ ID NO: 16). In certain embodiments, the scFv comprises an amino acid sequence having at least 90%, 95%, 97%, 99%, or 100% homology or identity with any of the sequences provided above (e.g., any of SEQ ID NOs: 1-6 and 13-16). In certain embodiments, the scFv comprises one, two, three, four, five, or all six of: GFTFHDY (SEQ ID NO: 7), SWRGDIG (SEQ ID NO: 8), SYGSGSFYNAFDS (SEQ ID NO: 9), RASQSISSWLA (SEQ ID NO: 10), KASKLER (SEQ ID NO: 11), and LEYSSYIRT (SEQ ID NO: 12). In certain embodiments, the scFv comprises a heavy chain variable domain comprising one, two, or all three of: GFTFHDY (SEQ ID NO: 7), SWRGDIG (SEQ ID NO: 8), SYGSGSFYNAFDS (SEQ ID NO: 9) and/or a light chain variable domain comprising one, two, or all three of: RASQSISSWLA (SEQ ID NO: 10), KASKLER
(SEQ ID NO: 11), and LEYSSYIRT (SEQ ID NO: 12). In certain embodiments, the scFv comprises a heavy chain variable domain comprising: EVQLVESGGGLVQPGRSLRLSCAASGFTFHDYAMHWVRQVPGKGLEWVSG ISWRGDIGGYVKSVKGRFTISRDNAKNSLYLQMNSLRAEDTALYYCVKSY GSGSFYNAFDSWGQGTLVTVSSA (SEQ ID NO: 17). In certain embodiments, the scFv comprises a light chain variable domain comprising: DIQMTQSPSTLSASVGDRVTITCRASQSISSWLAWYQQKPGKAPKFLIYK ASKLERGTPSRFSGSGDGTEFSLTISSLQPDDFATYYCLEYSSYIRTFGQ GTKVEIKRTV (SEQ ID NO: 18). In certain embodiments, the scFv comprises: EVQLVESGGGLVQPGRSLRLSCAASGFTFHDYAMHWVRQVPGKGLEWVSG ISWRGDIGGYVKSVKGRFTISRDNAKNSLYLQMNSLRAEDTALYYCVKSY GSGSFYNAFDSWGQGTLVTVSSAGGGGSGGGGSGGGGSDIQMTQSPSTLS ASVGDRVTITCRASQSISSWLAWYQQKPGKAPKFLIYKASKLERGTPSRF SGSGDGTEFSLTISSLQPDDFATYYCLEYSSYIRTFGQGTKVEIKRTV (SEQ ID NO: 19). In certain embodiments, the scFv (His-tagged) comprises: EVQLVESGGGLVQPGRSLRLSCAASGFTFHDYAMHWVRQVPGKGLEWVSG ISWRGDIGGYVKSVKGRFTISRDNAKNSLYLQMNSLRAEDTALYYCVKSY GSGSFYNAFDSWGQGTLVTVSSAGGGGSGGGGSGGGGSDIQMTQSPSTLS ASVGDRVTITCRASQSISSWLAWYQQKPGKAPKFLIYKASKLERGTPSRF SGSGDGTEFSLTISSLQPDDFATYYCLEYSSYIRTFGQGTKVEIKRTVHH HHHH (SEQ ID NO: 20). In certain embodiments, the scFv comprises an amino acid sequence having at least 90%, 95%, 97%, 99%, or 100% homology or identity with any of the sequences provided above (e.g., any of SEQ ID NOs: 7-12 and 17-20). The antibodies and antibody fragments of the instant invention may comprise one or more of the modifications set forth herein. In certain embodiments, the antibody fragment comprises a membrane binding domain. In certain embodiments, the antibody fragment is an scFv. In certain embodiments, the antibody fragment is a nanobody or single domain antibody. In certain embodiments, the membrane binding domain is selected from the group consisting of lactadherin C2 domain, Gla domains of vitamin K-dependent coagulation proteins, C2 domain of FVIII/FVIIIa, C1 domain of FVIII/FVIIIa, C2 domain of FV/FVa, and C1 domains of FV/FVa. In certain embodiments, the membrane binding domain is human (i.e., derived/obtained from a human protein). In certain embodiments, the membrane binding domain is the lactadherin C2 domain, Factor V C1 domain, and/or Factor V C2 domain. In certain embodiments, the
membrane binding domain is the Factor V C1 domain and/or Factor V C2 domain. In certain embodiments, the membrane binding domain is Factor V C2 domain. In certain embodiments, the membrane binding domain is lactadherin C2 domain. In certain embodiments, the membrane binding domain is at the C-terminus of the antibody fragment. In certain embodiments, the membrane binding domain is joined directly to the antibody fragment. In certain embodiments, the membrane binding domain is joined via a linker to the antibody fragment. In certain embodiments, the linker is a peptide or an amino acid sequence (e.g., about 1 to about 25 amino acids, about 10 to about 25 amino acids, about 15 to about 20 amino acids, about 10 to about 15 amino acids, about 13 to about 18 amino acids, or about 15 amino acids). In certain embodiments, the linker comprises the sequence (GGGGS)x (SEQ ID NO: 21), wherein x is 1-5, particularly 3-5, particularly 3. In certain embodiments, the Factor V C2 domain comprises: VNGCSTPLGMENGKIENKQITASSFKKSWWGDYWEPFRARLNAQGRVNAW QAKANNNKQWLEIDLLKIKKITAIITQGCKSLSSEMYVKSYTIHYSEQGV EWKPYRLKSSMVDKIFEGNTNTKGHVKNFFNPPIISRFIRVIPKTWNQSI TLRLELFGCDIYG (SEQ ID NO: 22). In certain embodiments, the Factor V C2 domain (His-tagged) comprises: VNGCSTPLGMENGKIENKQITASSFKKSWWGDYWEPFRARLNAQGRVNAW QAKANNNKQWLEIDLLKIKKITAIITQGCKSLSSEMYVKSYTIHYSEQGV EWKPYRLKSSMVDKIFEGNTNTKGHVKNFFNPPIISRFIRVIPKTWNQSI TLRLELFGCDIYGHHHHHH (SEQ ID NO: 23). In certain embodiments, the antibody fragment (VH9VLC2) comprises: QVQLVESGGGLVQPGGSLRLSCAASGFTFSYYDIQWVRQAPGKGLEWVSS ISPSGQSTYYRREVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCARRT GREYGGGWYFDYWGQGTLVTVSSAGGGGSGGGGSGGGGSDIQMTQSPSSL SASVGDRVTITCKASRNIERQLAWYQQKPGQAPELLIYQASRKESGVPDR FSGSRYGTDFTLTISSLQPEDIATYYCQQYSDPPLTFGGGTKVEIKRTVG GGGSGGGGSGGGGSVNGCSTPLGMENGKIENKQITASSFKKSWWGDYWEP FRARLNAQGRVNAWQAKANNNKQWLEIDLLKIKKITAIITQGCKSLSSEM YVKSYTIHYSEQGVEWKPYRLKSSMVDKIFEGNTNTKGHVKNFFNPPIIS RFIRVIPKTWNQSITLRLELFGCDIYG (SEQ ID NO: 24). In certain embodiments, the antibody fragment (His-tagged VH9VLC2) comprises: QVQLVESGGGLVQPGGSLRLSCAASGFTFSYYDIQWVRQAPGKGLEWVSS ISPSGQSTYYRREVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCARRT GREYGGGWYFDYWGQGTLVTVSSAGGGGSGGGGSGGGGSDIQMTQSPSSL SASVGDRVTITCKASRNIERQLAWYQQKPGQAPELLIYQASRKESGVPDR FSGSRYGTDFTLTISSLQPEDIATYYCQQYSDPPLTFGGGTKVEIKRTVG GGGSGGGGSGGGGSVNGCSTPLGMENGKIENKQITASSFKKSWWGDYWEP FRARLNAQGRVNAWQAKANNNKQWLEIDLLKIKKITAIITQGCKSLSSEM YVKSYTIHYSEQGVEWKPYRLKSSMVDKIFEGNTNTKGHVKNFFNPPIIS
RFIRVIPKTWNQSITLRLELFGCDIYGHHHHHH (SEQ ID NO: 25). In certain embodiments, the lactadherin C2 domain comprises: LNGCANPLGLKNNSIPDKQITASSSYKTWGLHLFSWNPSYARLDKQGNFN AWVAGSYGNDQWLQVDLGSSKEVTGIITQGARNFGSVQFVASYKVAYSND SANWTEYQDPRTGSSKIFPGNWDNHSHKKNLFETPILARYVRILPVAWHN RIALRLELLGC (SEQ ID NO: 55). In certain embodiments, the lactadherin C2 domain is encoded by the sequence: ctgaacggatgcgccaatcccctgggcctgaagaataacagcatccctgacaagcag atcacggcctccagcagctacaagacctggggcttgcatctcttcagctggaacccc tcctatgcacggctggacaagcagggcaacttcaacgcctgggttgcggggagctac ggtaacgatcagtggctgcaggtggacctgggctcctcgaaggaggtgacaggcatc atcacccagggggcccgtaactttggctctgtccagtttgtggcatcctacaaggtt gcctacagtaatgacagtgcgaactggactgagtaccaggaccccaggactggcagc agtaagatcttccctggcaactgggacaaccactcccacaagaagaacttgtttgag acgcccatcctggctcgctatgtgcgcatcctgcctgtagcctggcacaaccgcatc gccctgcgcctggagctgctgggctgt (SEQ ID NO: 61). In certain embodiments, the lactadherin C2 domain (His-tagged) comprises: LNGCANPLGLKNNSIPDKQITASSSYKTWGLHLFSWNPSYARLDKQGNFN AWVAGSYGNDQWLQVDLGSSKEVTGIITQGARNFGSVQFVASYKVAYSND SANWTEYQDPRTGSSKIFPGNWDNHSHKKNLFETPILARYVRILPVAWHN RIALRLELLGCHHHHHH (SEQ ID NO: 56). In certain embodiments, the antibody fragment (VH9VLLacC2) comprises: QVQLVESGGGLVQPGGSLRLSCAASGFTFSYYDIQWVRQAPGKGLEWVSS ISPSGQSTYYRREVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCARRT GREYGGGWYFDYWGQGTLVTVSSAGGGGSGGGGSGGGGSDIQMTQSPSSL SASVGDRVTITCKASRNIERQLAWYQQKPGQAPELLIYQASRKESGVPDR FSGSRYGTDFTLTISSLQPEDIATYYCQQYSDPPLTFGGGTKVEIKRTVG GGGSGGGGSGGGGSLNGCANPLGLKNNSIPDKQITASSSYKTWGLHLFSW NPSYARLDKQGNFNAWVAGSYGNDQWLQVDLGSSKEVTGIITQGARNFGS VQFVASYKVAYSNDSANWTEYQDPRTGSSKIFPGNWDNHSHKKNLFETPI LARYVRILPVAWHNRIALRLELLGC (SEQ ID NO: 57). In certain embodiments, the antibody fragment (His-tagged VH9VLLacC2) comprises: QVQLVESGGGLVQPGGSLRLSCAASGFTFSYYDIQWVRQAPGKGLEWVSS ISPSGQSTYYRREVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCARRT GREYGGGWYFDYWGQGTLVTVSSAGGGGSGGGGSGGGGSDIQMTQSPSSL SASVGDRVTITCKASRNIERQLAWYQQKPGQAPELLIYQASRKESGVPDR FSGSRYGTDFTLTISSLQPEDIATYYCQQYSDPPLTFGGGTKVEIKRTVG GGGSGGGGSGGGGSLNGCANPLGLKNNSIPDKQITASSSYKTWGLHLFSW NPSYARLDKQGNFNAWVAGSYGNDQWLQVDLGSSKEVTGIITQGARNFGS VQFVASYKVAYSNDSANWTEYQDPRTGSSKIFPGNWDNHSHKKNLFETPI LARYVRILPVAWHNRIALRLELLGCHHHHHH (SEQ ID NO: 58). In certain embodiments, the Factor V C1 domain comprises: LIMDRDCRMPMGLSTGIISDSQIKASEFLGYWEPRLARLNNGGSYNAWSV EKLAAEFASKPWIQVDMQKEVIITGIQTQGAKHYLKSCYTTEFYVAYSSN
QINWQIFKGNSTRNVMYFNGNSDASTIKENQFDPPIVARYIRISPTRAYN RPTLRLELQGCE (SEQ ID NO: 26). In certain embodiments, the Factor V C1 domain (His-tagged) comprises: LIMDRDCRMPMGLSTGIISDSQIKASEFLGYWEPRLARLNNGGSYNAWSV EKLAAEFASKPWIQVDMQKEVIITGIQTQGAKHYLKSCYTTEFYVAYSSN QINWQIFKGNSTRNVMYFNGNSDASTIKENQFDPPIVARYIRISPTRAYN RPTLRLELQGCEHHHHHH (SEQ ID NO: 27). In certain embodiments, the antibody fragment (VH9VLC1C2) comprises: QVQLVESGGGLVQPGGSLRLSCAASGFTFSYYDIQWVRQAPGKGLEWVSS ISPSGQSTYYRREVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCARRT GREYGGGWYFDYWGQGTLVTVSSAGGGGSGGGGSGGGGSDIQMTQSPSSL SASVGDRVTITCKASRNIERQLAWYQQKPGQAPELLIYQASRKESGVPDR FSGSRYGTDFTLTISSLQPEDIATYYCQQYSDPPLTFGGGTKVEIKRTVG GGGSGGGGSGGGGSLIMDRDCRMPMGLSTGIISDSQIKASEFLGYWEPRL ARLNNGGSYNAWSVEKLAAEFASKPWIQVDMQKEVIITGIQTQGAKHYLK SCYTTEFYVAYSSNQINWQIFKGNSTRNVMYFNGNSDASTIKENQFDPPI VARYIRISPTRAYNRPTLRLELQGCEVNGCSTPLGMENGKIENKQITASS FKKSWWGDYWEPFRARLNAQGRVNAWQAKANNNKQWLEIDLLKIKKITAI ITQGCKSLSSEMYVKSYTIHYSEQGVEWKPYRLKSSMVDKIFEGNTNTKG HVKNFFNPPIISRFIRVIPKTWNQSITLRLELFGCDIY (SEQ ID NO: 28). In certain embodiments, the antibody fragment (His-tagged VH9VLC1C2) comprises: QVQLVESGGGLVQPGGSLRLSCAASGFTFSYYDIQWVRQAPGKGLEWVSS ISPSGQSTYYRREVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCARRT GREYGGGWYFDYWGQGTLVTVSSAGGGGSGGGGSGGGGSDIQMTQSPSSL SASVGDRVTITCKASRNIERQLAWYQQKPGQAPELLIYQASRKESGVPDR FSGSRYGTDFTLTISSLQPEDIATYYCQQYSDPPLTFGGGTKVEIKRTVG GGGSGGGGSGGGGSLIMDRDCRMPMGLSTGIISDSQIKASEFLGYWEPRL ARLNNGGSYNAWSVEKLAAEFASKPWIQVDMQKEVIITGIQTQGAKHYLK SCYTTEFYVAYSSNQINWQIFKGNSTRNVMYFNGNSDASTIKENQFDPPI VARYIRISPTRAYNRPTLRLELQGCEVNGCSTPLGMENGKIENKQITASS FKKSWWGDYWEPFRARLNAQGRVNAWQAKANNNKQWLEIDLLKIKKITAI ITQGCKSLSSEMYVKSYTIHYSEQGVEWKPYRLKSSMVDKIFEGNTNTKG HVKNFFNPPIISRFIRVIPKTWNQSITLRLELFGCDIYHHHHHH (SEQ ID NO: 29). In certain embodiments, the antibody fragment (Mim8 VH9VLC2) comprises: EVQLVESGGGLVQPGRSLRLSCAASGFTFHDYAMHWVRQVPGKGLEWVSG ISWRGDIGGYVKSVKGRFTISRDNAKNSLYLQMNSLRAEDTALYYCVKSY GSGSFYNAFDSWGQGTLVTVSSAGGGGSGGGGSGGGGSDIQMTQSPSTLS ASVGDRVTITCRASQSISSWLAWYQQKPGKAPKFLIYKASKLERGTPSRF SGSGDGTEFSLTISSLQPDDFATYYCLEYSSYIRTFGQGTKVEIKRTVGG GGSGGGGSGGGGSVNGCSTPLGMENGKIENKQITASSFKKSWWGDYWEPF RARLNAQGRVNAWQAKANNNKQWLEIDLLKIKKITAIITQGCKSLSSEMY VKSYTIHYSEQGVEWKPYRLKSSMVDKIFEGNTNTKGHVKNFFNPPIISR FIRVIPKTWNQSITLRLELFGCDIYG (SEQ ID NO: 30). In certain embodiments, the antibody fragment (His-tagged Mim8 VH9VLC2) comprises:
EVQLVESGGGLVQPGRSLRLSCAASGFTFHDYAMHWVRQVPGKGLEWVSG ISWRGDIGGYVKSVKGRFTISRDNAKNSLYLQMNSLRAEDTALYYCVKSY GSGSFYNAFDSWGQGTLVTVSSAGGGGSGGGGSGGGGSDIQMTQSPSTLS ASVGDRVTITCRASQSISSWLAWYQQKPGKAPKFLIYKASKLERGTPSRF SGSGDGTEFSLTISSLQPDDFATYYCLEYSSYIRTFGQGTKVEIKRTVGG GGSGGGGSGGGGSVNGCSTPLGMENGKIENKQITASSFKKSWWGDYWEPF RARLNAQGRVNAWQAKANNNKQWLEIDLLKIKKITAIITQGCKSLSSEMY VKSYTIHYSEQGVEWKPYRLKSSMVDKIFEGNTNTKGHVKNFFNPPIISR FIRVIPKTWNQSITLRLELFGCDIYGHHHHHH (SEQ ID NO: 31). In certain embodiments, the Factor V C2 domain comprises an amino acid sequence having at least 90%, 95%, 97%, 99%, or 100% homology or identity with any of the sequences provided above (e.g., any of SEQ ID NOs: 22 and 23). In certain embodiments, the Factor V C1 domain comprises an amino acid sequence having at least 90%, 95%, 97%, 99%, or 100% homology or identity with any of the sequences provided above (e.g., any of SEQ ID NOs: 26 and 27). In certain embodiments, the lactadherin C2 domain comprises an amino acid sequence having at least 90%, 95%, 97%, 99%, or 100% homology or identity with any of the sequences provided above (e.g., any of SEQ ID NOs: 55 and 56). In certain embodiments, the antibody fragment comprises an amino acid sequence having at least 90%, 95%, 97%, 99%, or 100% homology or identity with any of the sequences provided above (e.g., any of SEQ ID NOs: 24, 25, 28-31, 57, and 58). In certain embodiments, the antibody fragment comprises a Protein C tag, particularly a tag recognized by the commercially available HPC4 antibody (e.g., Cell Signaling Technology, Inc., Danvers, MA). In certain embodiments, the tag comprises the amino acid sequence EDQVDPRLIDGK (SEQ ID NO: 59). In certain embodiments, the tag is at the C-terminus. In certain embodiments, the tag is at the C-terminus of the antibody fragment. In certain embodiments, the tag is at the C- terminus of the membrane binding domain. In certain embodiments, the tag is at the C-terminus of the dimerization domain. In certain embodiments, the tag is at the C- terminus of the lactadherin C2 domain, Factor V C1 domain, and/or Factor V C2 domain. In certain embodiments, the tag comprises an amino acid sequence having at least 90%, 95%, 97%, 99%, or 100% homology or identity with SEQ ID NO: 59. In certain embodiments, the antibody fragment comprises a cleavage site, particularly a thrombin (Factor IIa) cleavage site. In certain embodiments, the thrombin cleavage site is an amino acid sequence of less than 10 amino acids, less than 9 amino acids, less than 8 amino acids, less than 7 amino acids, or shorter. In
certain embodiments, the thrombin cleavage site comprises the amino acid sequence LVPRGS (SEQ ID NO: 60). The thrombin cleavage site may have 1, 2, 3, or 4 substitutions in SEQ ID NO: 60, particularly the Leu, Val, Gly (e.g., with Ala, Ser, or Thr), and/or Ser (e.g., with a non-acidic amino acid) may be substituted. Generally, thrombin cleaves the amino acid sequence after an Arg. In certain embodiments, the cleavage site (e.g., thrombin cleavage site) is located C-terminal to the antibody fragment. In certain embodiments, the cleavage site (e.g., thrombin cleavage site) is located C-terminal to the scFv. In certain embodiments, the cleavage site (e.g., thrombin cleavage site) is located between the antibody fragment and the membrane binding domain. In certain embodiments, the cleavage site (e.g., thrombin cleavage site) is located between the antibody fragment and the lactadherin C2 domain, Factor V C1 domain, and/or Factor V C2 domain. In certain embodiments, the antibody fragment comprises a dimerization domain. In certain embodiments, the antibody fragment is an scFv. In certain embodiments, the antibody fragment is a nanobody or single domain antibody. In certain embodiments, the dimerization domain comprises CH2 and CH3. In certain embodiments, the dimerization domain is an Fc region. In certain embodiments, the CH2 and CH3 are from the FIXa binding arm of emicizumab. In certain embodiments, the dimerization domain is at the C-terminus of the antibody fragment. In certain embodiments, the dimerization domain is joined directly to the antibody fragment. In certain embodiments, the dimerization domain is joined via a linker to the antibody fragment. In certain embodiments, the linker is a peptide or an amino acid sequence (e.g., about 1 to about 25 amino acids, about 10 to about 25 amino acids, about 15 to about 20 amino acids, about 10 to about 15 amino acids, about 13 to about 18 amino acids, or about 15 amino acids). In certain embodiments, the linker comprises the sequence (GGGGS)x (SEQ ID NO: 21), wherein x is 1-5, particularly 3-5, particularly 3. In certain embodiments, the antibody fragment further comprises a membrane binding domain at the C-terminus of the dimerization domain. In certain embodiments, the membrane binding domain is joined directly to the dimerization domain. In certain embodiments, the membrane binding domain is joined via a linker to the dimerization domain. In certain embodiments, the linker is a peptide or an amino acid sequence (e.g., about 1 to about 25 amino acids, about 10 to about 25 amino acids, about 15 to about 20 amino acids, about 10 to about 15 amino acids, about 13 to about 18 amino acids, or about 15 amino acids). In certain embodiments,
the linker comprises the sequence (GGGGS)x (SEQ ID NO: 21), wherein x is 1-5, particularly 3-5, particularly 3. In certain embodiments, the CH2 and CH3 domain comprises: APEFLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSQEDPEVQFNWYVD GVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKGLPS SIEKTISKAKGQPREPQVYTLPPSQKEMTKNQVSLTCLVKGFYPSDIAVE WESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQEGNVFSCSVMHE ALHNRYTQKSLSLSP (SEQ ID NO: 32). In certain embodiment, the antibody fragment (VH9VLCH2CH3) comprises: QVQLVESGGGLVQPGGSLRLSCAASGFTFSYYDIQWVRQAPGKGLEWVSS ISPSGQSTYYRREVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCARRT GREYGGGWYFDYWGQGTLVTVSSAGGGGSGGGGSGGGGSDIQMTQSPSSL SASVGDRVTITCKASRNIERQLAWYQQKPGQAPELLIYQASRKESGVPDR FSGSRYGTDFTLTISSLQPEDIATYYCQQYSDPPLTFGGGTKVEIKRTVG GGGSGGGGSGGGGSAPEFLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDV SQEDPEVQFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNG KEYKCKVSNKGLPSSIEKTISKAKGQPREPQVYTLPPSQKEMTKNQVSLT CLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSR WQEGNVFSCSVMHEALHNRYTQKSLSLSP (SEQ ID NO: 33). In certain embodiment, the antibody fragment (VH9VLCH2CH3C2) comprises: QVQLVESGGGLVQPGGSLRLSCAASGFTFSYYDIQWVRQAPGKGLEWVSS ISPSGQSTYYRREVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCARRT GREYGGGWYFDYWGQGTLVTVSSAGGGGSGGGGSGGGGSDIQMTQSPSSL SASVGDRVTITCKASRNIERQLAWYQQKPGQAPELLIYQASRKESGVPDR FSGSRYGTDFTLTISSLQPEDIATYYCQQYSDPPLTFGGGTKVEIKRTVG GGGSGGGGSGGGGSAPEFLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDV SQEDPEVQFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNG KEYKCKVSNKGLPSSIEKTISKAKGQPREPQVYTLPPSQKEMTKNQVSLT CLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSR WQEGNVFSCSVMHEALHNRYTQKSLSLSPGGGGSGGGGSGGGGSVNGCST PLGMENGKIENKQITASSFKKSWWGDYWEPFRARLNAQGRVNAWQAKANN NKQWLEIDLLKIKKITAIITQGCKSLSSEMYVKSYTIHYSEQGVEWKPYR LKSSMVDKIFEGNTNTKGHVKNFFNPPIISRFIRVIPKTWNQSITLRLEL FGCDIYG (SEQ ID NO: 34). In certain embodiment, the antibody fragment (His-tagged VH9VLCH2CH3C2) comprises: QVQLVESGGGLVQPGGSLRLSCAASGFTFSYYDIQWVRQAPGKGLEWVSS ISPSGQSTYYRREVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCARRT GREYGGGWYFDYWGQGTLVTVSSAGGGGSGGGGSGGGGSDIQMTQSPSSL SASVGDRVTITCKASRNIERQLAWYQQKPGQAPELLIYQASRKESGVPDR FSGSRYGTDFTLTISSLQPEDIATYYCQQYSDPPLTFGGGTKVEIKRTVG GGGSGGGGSGGGGSAPEFLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDV SQEDPEVQFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNG KEYKCKVSNKGLPSSIEKTISKAKGQPREPQVYTLPPSQKEMTKNQVSLT CLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSR
WQEGNVFSCSVMHEALHNRYTQKSLSLSPGGGGSGGGGSGGGGSVNGCST PLGMENGKIENKQITASSFKKSWWGDYWEPFRARLNAQGRVNAWQAKANN NKQWLEIDLLKIKKITAIITQGCKSLSSEMYVKSYTIHYSEQGVEWKPYR LKSSMVDKIFEGNTNTKGHVKNFFNPPIISRFIRVIPKTWNQSITLRLEL FGCDIYGHHHHHH (SEQ ID NO: 35). In certain embodiments, the CH2 and CH3 domain comprises an amino acid sequence having at least 90%, 95%, 97%, 99%, or 100% homology or identity with any of the sequences provided above (e.g., SEQ ID NO: 32). In certain embodiments, the antibody fragment comprises an amino acid sequence having at least 90%, 95%, 97%, 99%, or 100% homology or identity with any of the sequences provided above (e.g., any of SEQ ID NOs: 33-35). In certain embodiments, the antibody fragment comprises two scFv antibody fragments. In certain embodiments, the antibody fragment is a scFv-scFv fusion, particularly a bispecific scFv-scFv fusion. In certain embodiments, the scFv-scFv fusion is biparatropic. In certain embodiments, at least one scFv of the bispecific scFv-scFv fusion is as described herein. In certain embodiments, both scFv of the scFv-scFv fusion bind FIXa, particularly wherein they bind different epitopes. In certain embodiments, one scFv of the bispecific scFv-scFv fusion binds FIXa and/or one scFv of the bispecific scFv-scFv fusion binds FX. In certain embodiments, one or both scFv of the bispecific scFv-scFv fusion are from a FVIIIa mimetic bispecific antibody such as emicizumab or mim8. In certain embodiments, one scFv is from a first FVIIIa mimetic bispecific antibody (e.g., emicizumab (e.g., FIXa binding scFv or FX binding scFv)) and the other scFv is from a second FVIIIa mimetic bispecific antibody (e.g., mim8 (e.g., FIXa binding scFv or FX binding scFv)). In certain embodiments, the scFv antibody fragments are joined directly to each other. In certain embodiments, the first and second scFv are joined via a linker. In certain embodiments, the linker is a peptide or an amino acid sequence (e.g., about 1 to about 25 amino acids, about 10 to about 25 amino acids, about 15 to about 20 amino acids, about 10 to about 15 amino acids, about 13 to about 18 amino acids, or about 15 amino acids). In certain embodiments, the linker comprises the sequence (GGGGS)x (SEQ ID NO: 21), wherein x is 1-5, particularly 3-5, particularly 3. In certain embodiments, the scFv-scFv fusion (e.g., bispecific scFv-scFv fusion) further comprises a membrane binding domain as described herein and/or a dimerization domain as described herein. In certain embodiments, the FX-binding scFv comprises a heavy chain variable domain comprising:
QVQLVQSGSELKKPGASVKVSCKASGYTFTDNNMDWVRQAPGQGLEWMGD INTRSGGSIYNEEFQDRVIMTVDKSTDTAYMELSSLRSEDTATYHCARRK SYGYYLDEWGEGTLVTVSSA (SEQ ID NO: 36). In certain embodiments, the FX-binding scFv comprises a light chain variable domain comprising: DIQMTQSPSSLSASVGDRVTITCKASRNIERQLAWYQQKPGQAPELLIYQ ASRKESGVPDRFSGSRYGTDFTLTISSLQPEDIATYYCQQYSDPPLTFGG GTKVEIKRTV (SEQ ID NO: 37). In certain embodiments, the FX-binding scFv comprises: QVQLVESGGGLVQPGGSLRLSCAASGFTFSYYDIQWVRQAPGKGLEWVSS ISPSGQSTYYRREVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCARRT GREYGGGWYFDYWGQGTLVTVSSAGGGGSGGGGSGGGGSDIQMTQSPSSL SASVGDRVTITCKASRNIERQLAWYQQKPGQAPELLIYQASRKESGVPDR FSGSRYGTDFTLTISSLQPEDIATYYCQQYSDPPLTFGGGTKVEIKRTV (SEQ ID NO: 38). In certain embodiments, the scFv-scFv (Emi VH9VL-VH10VL) comprises: QVQLVESGGGLVQPGGSLRLSCAASGFTFSYYDIQWVRQAPGKGLEWVSS ISPSGQSTYYRREVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCARRT GREYGGGWYFDYWGQGTLVTVSSAGGGGSGGGGSGGGGSDIQMTQSPSSL SASVGDRVTITCKASRNIERQLAWYQQKPGQAPELLIYQASRKESGVPDR FSGSRYGTDFTLTISSLQPEDIATYYCQQYSDPPLTFGGGTKVEIKRTVG GGGSGGGGSGGGGSQVQLVQSGSELKKPGASVKVSCKASGYTFTDNNMDW VRQAPGQGLEWMGDINTRSGGSIYNEEFQDRVIMTVDKSTDTAYMELSSL RSEDTATYHCARRKSYGYYLDEWGEGTLVTVSSAGGGGSGGGGSGGGGSD IQMTQSPSSLSASVGDRVTITCKASRNIERQLAWYQQKPGQAPELLIYQA SRKESGVPDRFSGSRYGTDFTLTISSLQPEDIATYYCQQYSDPPLTFGGG TKVEIKRTV (SEQ ID NO: 39). In certain embodiments, the scFv-scFv (His-tagged Emi VH9VL-VH10VL) comprises: QVQLVESGGGLVQPGGSLRLSCAASGFTFSYYDIQWVRQAPGKGLEWVSS ISPSGQSTYYRREVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCARRT GREYGGGWYFDYWGQGTLVTVSSAGGGGSGGGGSGGGGSDIQMTQSPSSL SASVGDRVTITCKASRNIERQLAWYQQKPGQAPELLIYQASRKESGVPDR FSGSRYGTDFTLTISSLQPEDIATYYCQQYSDPPLTFGGGTKVEIKRTVG GGGSGGGGSGGGGSQVQLVQSGSELKKPGASVKVSCKASGYTFTDNNMDW VRQAPGQGLEWMGDINTRSGGSIYNEEFQDRVIMTVDKSTDTAYMELSSL RSEDTATYHCARRKSYGYYLDEWGEGTLVTVSSAGGGGSGGGGSGGGGSD IQMTQSPSSLSASVGDRVTITCKASRNIERQLAWYQQKPGQAPELLIYQA SRKESGVPDRFSGSRYGTDFTLTISSLQPEDIATYYCQQYSDPPLTFGGG TKVEIKRTVHHHHHH (SEQ ID NO: 40). In certain embodiments, the scFv-scFv (Duo - mim8 FIXa scFv and emicizumab FIXa scFv) comprises: EVQLVESGGGLVQPGRSLRLSCAASGFTFHDYAMHWVRQVPGKGLEWVSG ISWRGDIGGYVKSVKGRFTISRDNAKNSLYLQMNSLRAEDTALYYCVKSY GSGSFYNAFDSWGQGTLVTVSSAGGGGSGGGGSGGGGSDIQMTQSPSTLS
ASVGDRVTITCRASQSISSWLAWYQQKPGKAPKFLIYKASKLERGTPSRF SGSGDGTEFSLTISSLQPDDFATYYCLEYSSYIRTFGQGTKVEIKRTVGG GGSGGGGSGGGGSQVQLVESGGGLVQPGGSLRLSCAASGFTFSYYDIQWV RQAPGKGLEWVSSISPSGQSTYYRREVKGRFTISRDNSKNTLYLQMNSLR AEDTAVYYCARRTGREYGGGWYFDYWGQGTLVTVSSAGGGGSGGGGSGGG GSDIQMTQSPSSLSASVGDRVTITCKASRNIERQLAWYQQKPGQAPELLI YQASRKESGVPDRFSGSRYGTDFTLTISSLQPEDIATYYCQQYSDPPLTF GGGTKVEIKRTV (SEQ ID NO: 41). In certain embodiments, the scFv-scFv (His-tagged Duo – mim8 FIXa scFv and emicizumab FIXa scFv) comprises: EVQLVESGGGLVQPGRSLRLSCAASGFTFHDYAMHWVRQVPGKGLEWVSG ISWRGDIGGYVKSVKGRFTISRDNAKNSLYLQMNSLRAEDTALYYCVKSY GSGSFYNAFDSWGQGTLVTVSSAGGGGSGGGGSGGGGSDIQMTQSPSTLS ASVGDRVTITCRASQSISSWLAWYQQKPGKAPKFLIYKASKLERGTPSRF SGSGDGTEFSLTISSLQPDDFATYYCLEYSSYIRTFGQGTKVEIKRTVGG GGSGGGGSGGGGSQVQLVESGGGLVQPGGSLRLSCAASGFTFSYYDIQWV RQAPGKGLEWVSSISPSGQSTYYRREVKGRFTISRDNSKNTLYLQMNSLR AEDTAVYYCARRTGREYGGGWYFDYWGQGTLVTVSSAGGGGSGGGGSGGG GSDIQMTQSPSSLSASVGDRVTITCKASRNIERQLAWYQQKPGQAPELLI YQASRKESGVPDRFSGSRYGTDFTLTISSLQPEDIATYYCQQYSDPPLTF GGGTKVEIKRTVHHHHHH (SEQ ID NO: 42). In certain embodiments, the scFv-scFv (DuoC2 - mim8 FIXa scFv and emicizumab FIXa scFv and C2 domain) comprises: EVQLVESGGGLVQPGRSLRLSCAASGFTFHDYAMHWVRQVPGKGLEWVSG ISWRGDIGGYVKSVKGRFTISRDNAKNSLYLQMNSLRAEDTALYYCVKSY GSGSFYNAFDSWGQGTLVTVSSAGGGGSGGGGSGGGGSDIQMTQSPSTLS ASVGDRVTITCRASQSISSWLAWYQQKPGKAPKFLIYKASKLERGTPSRF SGSGDGTEFSLTISSLQPDDFATYYCLEYSSYIRTFGQGTKVEIKRTVGG GGSGGGGSGGGGSQVQLVESGGGLVQPGGSLRLSCAASGFTFSYYDIQWV RQAPGKGLEWVSSISPSGQSTYYRREVKGRFTISRDNSKNTLYLQMNSLR AEDTAVYYCARRTGREYGGGWYFDYWGQGTLVTVSSAGGGGSGGGGSGGG GSDIQMTQSPSSLSASVGDRVTITCKASRNIERQLAWYQQKPGQAPELLI YQASRKESGVPDRFSGSRYGTDFTLTISSLQPEDIATYYCQQYSDPPLTF GGGTKVEIKRTVGGGGSGGGGSGGGGSVNGCSTPLGMENGKIENKQITAS SFKKSWWGDYWEPFRARLNAQGRVNAWQAKANNNKQWLEIDLLKIKKITA IITQGCKSLSSEMYVKSYTIHYSEQGVEWKPYRLKSSMVDKIFEGNTNTK GHVKNFFNPPIISRFIRVIPKTWNQSITLRLELFGCDIYG (SEQ ID NO: 43). In certain embodiments, the scFv-scFv (His-tagged DuoC2 - mim8 FIXa scFv and emicizumab FIXa scFv and C2 domain) comprises: EVQLVESGGGLVQPGRSLRLSCAASGFTFHDYAMHWVRQVPGKGLEWVSG ISWRGDIGGYVKSVKGRFTISRDNAKNSLYLQMNSLRAEDTALYYCVKSY GSGSFYNAFDSWGQGTLVTVSSAGGGGSGGGGSGGGGSDIQMTQSPSTLS ASVGDRVTITCRASQSISSWLAWYQQKPGKAPKFLIYKASKLERGTPSRF SGSGDGTEFSLTISSLQPDDFATYYCLEYSSYIRTFGQGTKVEIKRTVGG GGSGGGGSGGGGSQVQLVESGGGLVQPGGSLRLSCAASGFTFSYYDIQWV
RQAPGKGLEWVSSISPSGQSTYYRREVKGRFTISRDNSKNTLYLQMNSLR AEDTAVYYCARRTGREYGGGWYFDYWGQGTLVTVSSAGGGGSGGGGSGGG GSDIQMTQSPSSLSASVGDRVTITCKASRNIERQLAWYQQKPGQAPELLI YQASRKESGVPDRFSGSRYGTDFTLTISSLQPEDIATYYCQQYSDPPLTF GGGTKVEIKRTVGGGGSGGGGSGGGGSVNGCSTPLGMENGKIENKQITAS SFKKSWWGDYWEPFRARLNAQGRVNAWQAKANNNKQWLEIDLLKIKKITA IITQGCKSLSSEMYVKSYTIHYSEQGVEWKPYRLKSSMVDKIFEGNTNTK GHVKNFFNPPIISRFIRVIPKTWNQSITLRLELFGCDIYGHHHHHH (SEQ ID NO: 44). In certain embodiments, the FX binding scFv comprises an amino acid sequence having at least 90%, 95%, 97%, 99%, or 100% homology or identity with any of the sequences provided above (e.g., any of SEQ ID NOs: 36-38). In certain embodiments, the scFv-scFv comprises an amino acid sequence having at least 90%, 95%, 97%, 99%, or 100% homology or identity with any of the sequences provided above (e.g., any of SEQ ID NOs: 39-44). In certain embodiments, the antibody fragment comprises albumin, particularly human albumin. In certain embodiments, the antibody fragment is an scFv. In certain embodiments, the antibody fragment is a nanobody or single domain antibody. In certain embodiments, the albumin is at the C-terminus of the antibody fragment. In certain embodiments, the albumin is at the N-terminus of the antibody fragment. In certain embodiments, the albumin is joined directly to the antibody fragment. In certain embodiments, the albumin is joined via a linker to the antibody fragment. In certain embodiments, the linker is a peptide or an amino acid sequence (e.g., about 1 to about 25 amino acids, about 10 to about 25 amino acids, about 15 to about 20 amino acids, about 10 to about 15 amino acids, about 13 to about 18 amino acids, or about 15 amino acids). In certain embodiments, the linker comprises the sequence (GGGGS)x (SEQ ID NO: 21), wherein x is 1-5, particularly 3-5, particularly 3. In certain embodiments, the linker is a cleavable linker. In certain embodiments, the linker comprises at least a fragment of the Factor IX activation peptide. In certain embodiments, the human albumin comprises: DAHKSEVAHRFKDLGEENFKALVLIAFAQYLQQCPFEDHVKLVNEVTEFA KTCVADESAENCDKSLHTLFGDKLCTVATLRETYGEMADCCAKQEPERNE CFLQHKDDNPNLPRLVRPEVDVMCTAFHDNEETFLKKYLYEIARRHPYFY APELLFFAKRYKAAFTECCQAADKAACLLPKLDELRDEGKASSAKQRLKC ASLQKFGERAFKAWAVARLSQRFPKAEFAEVSKLVTDLTKVHTECCHGDL LECADDRADLAKYICENQDSISSKLKECCEKPLLEKSHCIAEVENDEMPA DLPSLAADFVESKDVCKNYAEAKDVFLGMFLYEYARRHPDYSVVLLLRLA KTYETTLEKCCAAADPHECYAKVFDEFKPLVEEPQNLIKQNCELFEQLGE YKFQNALLVRYTKKVPQVSTPTLVEVSRNLGKVGSKCCKHPEAKRMPCAE
DYLSVVLNQLCVLHEKTPVSDRVTKCCTESLVNRRPCFSALEVDETYVPK EFNAETFTFHADICTLSEKERQIKKQTALVELVKHKPKATKEQLKAVMDD FAAFVEKCCKADDKETCFAEEGKKLVAASQAALGL (SEQ ID NO: 62). In certain embodiments, the antibody fragment (His-tagged VH9VLC2) comprises (wherein the His-tag is optional): QVQLVESGGGLVQPGGSLRLSCAASGFTFSYYDIQWVRQAPGKGLEWVSS ISPSGQSTYYRREVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCARRT GREYGGGWYFDYWGQGTLVTVSSAGGGGSGGGGSGGGGSDIQMTQSPSSL SASVGDRVTITCKASRNIERQLAWYQQKPGQAPELLIYQASRKESGVPDR FSGSRYGTDFTLTISSLQPEDIATYYCQQYSDPPLTFGGGTKVEIKRTVG GGGSGGGGSGGGGSVNGCSTPLGMENGKIENKQITASSFKKSWWGDYWEP FRARLNAQGRVNAWQAKANNNKQWLEIDLLKIKKITAIITQGCKSLSSEM YVKSYTIHYSEQGVEWKPYRLKSSMVDKIFEGNTNTKGHVKNFFNPPIIS RFIRVIPKTWNQSITLRLELFGCDIYGHHHHHH (SEQ ID NO: 25). In certain embodiments, the Factor IX activation peptide comprises: SVSQTSKLTQAETVFPDVDGS (SEQ ID NO: 63) or SVSQTSKLTRAETVFPDVDGS (SEQ ID NO: 64). In certain embodiments, human albumin fused antibody fragment (VH9VL C2) comprises (wherein the His-tag is optional): DAHKSEVAHRFKDLGEENFKALVLIAFAQYLQQCPFEDHVKLVNEVTEFA KTCVADESAENCDKSLHTLFGDKLCTVATLRETYGEMADCCAKQEPERNE CFLQHKDDNPNLPRLVRPEVDVMCTAFHDNEETFLKKYLYEIARRHPYFY APELLFFAKRYKAAFTECCQAADKAACLLPKLDELRDEGKASSAKQRLKC ASLQKFGERAFKAWAVARLSQRFPKAEFAEVSKLVTDLTKVHTECCHGDL LECADDRADLAKYICENQDSISSKLKECCEKPLLEKSHCIAEVENDEMPA DLPSLAADFVESKDVCKNYAEAKDVFLGMFLYEYARRHPDYSVVLLLRLA KTYETTLEKCCAAADPHECYAKVFDEFKPLVEEPQNLIKQNCELFEQLGE YKFQNALLVRYTKKVPQVSTPTLVEVSRNLGKVGSKCCKHPEAKRMPCAE DYLSVVLNQLCVLHEKTPVSDRVTKCCTESLVNRRPCFSALEVDETYVPK EFNAETFTFHADICTLSEKERQIKKQTALVELVKHKPKATKEQLKAVMDD FAAFVEKCCKADDKETCFAEEGKKLVAASQAALGLSVSQTSKLTQAETVF PDVDGSQVQLVESGGGLVQPGGSLRLSCAASGFTFSYYDIQWVRQAPGKG LEWVSSISPSGQSTYYRREVKGRFTISRDNSKNTLYLQMNSLRAEDTAVY YCARRTGREYGGGWYFDYWGQGTLVTVSSAGGGGSGGGGSGGGGSDIQMT QSPSSLSASVGDRVTITCKASRNIERQLAWYQQKPGQAPELLIYQASRKE SGVPDRFSGSRYGTDFTLTISSLQPEDIATYYCQQYSDPPLTFGGGTKVE IKRTVGGGGSGGGGSGGGGSVNGCSTPLGMENGKIENKQITASSFKKSWW GDYWEPFRARLNAQGRVNAWQAKANNNKQWLEIDLLKIKKITAIITQGCK SLSSEMYVKSYTIHYSEQGVEWKPYRLKSSMVDKIFEGNTNTKGHVKNFF NPPIISRFIRVIPKTWNQSITLRLELFGCDIYGHHHHHH (SEQ ID NO: 65).
In certain embodiments, the albumin comprises an amino acid sequence having at least 90%, 95%, 97%, 99%, or 100% homology or identity with any of the sequences provided above (e.g., SEQ ID NO: 62). In certain embodiments, the Factor IX activation peptide comprises an amino acid sequence having at least 90%, 95%, 97%, 99%, or 100% homology or identity with any of the sequences provided above (e.g., SEQ ID NO: 63 or 64). In certain embodiments, the antibody fragment comprises an amino acid sequence having at least 90%, 95%, 97%, 99%, or 100% homology or identity with any of the sequences provided above (e.g., any of SEQ ID NO: 65). In accordance with another aspect of the instant invention, antibodies joined with a membrane binding domain are provided. In certain embodiments, the antibody is a FVIIIa mimetic bispecific antibody. In certain embodiments, the FVIIIa mimetic bispecific antibody is emicizumab. In certain embodiments, the FVIIIa mimetic bispecific antibody is mim8. In certain embodiments, the membrane binding domain is joined to the C-terminus of the antibody. In certain embodiments, the membrane binding domain is selected from the group consisting of lactadherin C2 domain, Gla domains of vitamin K-dependent coagulation proteins, C2 domain of FVIII/FVIIIa, C1 domain of FVIII/FVIIIa, C2 domain of FV/FVa, and C1 domains of FV/FVa. In certain embodiments, the membrane binding domain is the Factor V C2 domain, Factor V C1 domain, and/or lactadherin C2 domain. In certain embodiments, the membrane binding domain is the Factor V C2 domain, as described herein. Compositions comprising an antibody or antibody fragment of the instant invention and a carrier such as a pharmaceutically acceptable carrier are also encompassed by the instant invention. In a particular embodiment, the composition comprises at least one antibody or antibody fragment and at least one carrier (e.g., a pharmaceutically acceptable carrier). Nucleic acid molecules encoding an antibody or antibody fragment are encompassed by the instant invention. In certain embodiments, the nucleic acid molecule encodes an antibody fragment of the instant invention. In certain embodiments, the nucleic acid molecules of the instant invention are contained within a vector, particularly an expression vector. In certain embodiments, the nucleic acid molecules of the instant invention are contained within a viral vector (e.g., an AAV vector). The instant invention also encompasses cells comprising and, optionally, expressing a nucleic acid molecule of the instant invention (e.g., hybridomas that secrete antibodies or antibody fragments).
Examples of nucleic acid molecules of the instant invention include, without limitation: His-tagged VH9VL caagtgcaactggtggaatccggcggaggactggtgcagcccggcggcagcctgagg ctgtcttgtgctgcttctggcttcaccttctcctattacgatatccagtgggtgcgg caggcccctggaaaaggcctggaatgggtctccagcatctccccatccggccagagc acctactacagaagagaggtgaagggcagattcacaatcagcagagataacagcaag aacaccctctacctgcagatgaatagcctgcgggccgaggacaccgccgtgtactac tgcgccagaagaaccggcagagagtacggcggcggctggtacttcgactactggggc cagggcacactggttacagtgtccagcgccggtggaggcggttcaggcggaggtggc tctggcggtggcggatcggacatccagatgacccagtctccatcttccttgtctgca tctgtaggagacagagtcaccatcacttgcaaggccagtcgtaatatcgagcgtcag ctggcctggtaccagcagaaacctggccaggctcccgagctcctcatctatcaggca tccagaaaggagagtggcgtgccagataggttcagtggcagtagatatgggacagac ttcactctcaccatcagcagcctgcagcctgaagatatcgccacatattactgtcag cagtatagcgatcctccgctgacttttggcgggggcaccaaggtggagatcaaacgt acggtgcatcatcaccatcaccattag (SEQ ID NO: 45) His-tagged VH9VLC2 caagtgcaactggtggaatccggcggaggactggtgcagcccggcggcagcctgagg ctgtcttgtgctgcttctggcttcaccttctcctattacgatatccagtgggtgcgg caggcccctggaaaaggcctggaatgggtctccagcatctccccatccggccagagc acctactacagaagagaggtgaagggcagattcacaatcagcagagataacagcaag aacaccctctacctgcagatgaatagcctgcgggccgaggacaccgccgtgtactac tgcgccagaagaaccggcagagagtacggcggcggctggtacttcgactactggggc cagggcacactggttacagtgtccagcgccggtggaggcggttcaggcggaggtggc tctggcggtggcggatcggacatccagatgacccagtctccatcttccttgtctgca tctgtaggagacagagtcaccatcacttgcaaggccagtcgtaatatcgagcgtcag ctggcctggtaccagcagaaacctggccaggctcccgagctcctcatctatcaggca tccagaaaggagagtggcgtgccagataggttcagtggcagtagatatgggacagac ttcactctcaccatcagcagcctgcagcctgaagatatcgccacatattactgtcag cagtatagcgatcctccgctgacttttggcgggggcaccaaggtggagatcaaacgt acggtgggtggaggcggttcaggcggaggtggctctggcggtggcggatcggtaaat ggatgttccacacccctgggtatggaaaatggaaagatagaaaacaagcaaatcaca gcttcttcgtttaagaaatcttggtggggagattactgggaacccttccgtgcccgt ctgaatgcccagggacgtgtgaatgcctggcaagccaaggcaaacaacaataagcag tggctagaaattgatctactcaagatcaagaagataacggcaattataacacagggc tgcaagtctctgtcctctgaaatgtatgtaaagagctataccatccactacagtgag cagggagtggaatggaaaccatacaggctgaaatcctccatggtggacaagattttt gaaggaaatactaataccaaaggacatgtgaagaactttttcaaccccccaatcatt tccaggtttatccgtgtcattcctaaaacatggaatcaaagtattacacttcgcctg gaactctttggctgtgatatttacggacatcatcaccatcaccattag (SEQ ID NO: 46) VH9VLCH2CH3 caagtgcaactggtggaatccggcggaggactggtgcagcccggcggcagcctgagg ctgtcttgtgctgcttctggcttcaccttctcctattacgatatccagtgggtgcgg caggcccctggaaaaggcctggaatgggtctccagcatctccccatccggccagagc acctactacagaagagaggtgaagggcagattcacaatcagcagagataacagcaag aacaccctctacctgcagatgaatagcctgcgggccgaggacaccgccgtgtactac
tgcgccagaagaaccggcagagagtacggcggcggctggtacttcgactactggggc cagggcacactggttacagtgtccagcgccggtggaggcggttcaggcggaggtggc tctggcggtggcggatcggacatccagatgacccagtctccatcttccttgtctgca tctgtaggagacagagtcaccatcacttgcaaggccagtcgtaatatcgagcgtcag ctggcctggtaccagcagaaacctggccaggctcccgagctcctcatctatcaggca tccagaaaggagagtggcgtgccagataggttcagtggcagtagatatgggacagac ttcactctcaccatcagcagcctgcagcctgaagatatcgccacatattactgtcag cagtatagcgatcctccgctgacttttggcgggggcaccaaggtggagatcaaacgt acggtgggtggaggcggttcaggcggaggtggctctggcggtggcggatcggcacct gagttcctcgggggaccatcagtcttcctgttccccccaaaacccaaggacactctc atgatctcccggacccctgaggtcacgtgcgtggtggtggacgtgagccaggaagac cccgaggtccagttcaactggtacgtggatggcgtggaggtgcataatgccaagaca aagccgcgggaggagcagtacaacagcacgtaccgtgtggtcagcgtcctcaccgtc ctgcaccaggactggctgaacggcaaggagtacaagtgcaaggtctccaacaaaggc ctcccgtcctccatcgagaaaaccatctccaaagccaaagggcagccccgagagcca caggtgtacaccctgcccccatcccagaaggagatgaccaagaaccaggtcagcctg acctgcctggtcaaaggcttctaccccagcgacatcgccgtggagtgggagagcaat gggcagccggagaacaactacaagaccacgcctcccgtgctggactccgacggctcc ttcttcctctacagcaagctaaccgtggacaagagcaggtggcaggaggggaatgtc ttctcatgctccgtgatgcatgaggctctgcacaacaggtacacacagaagagcctc tccctgtctccgtag (SEQ ID NO: 47) His-tagged VH9VLCH2CH3C2 caagtgcaactggtggaatccggcggaggactggtgcagcccggcggcagcctgagg ctgtcttgtgctgcttctggcttcaccttctcctattacgatatccagtgggtgcgg caggcccctggaaaaggcctggaatgggtctccagcatctccccatccggccagagc acctactacagaagagaggtgaagggcagattcacaatcagcagagataacagcaag aacaccctctacctgcagatgaatagcctgcgggccgaggacaccgccgtgtactac tgcgccagaagaaccggcagagagtacggcggcggctggtacttcgactactggggc cagggcacactggttacagtgtccagcgccggtggaggcggttcaggcggaggtggc tctggcggtggcggatcggacatccagatgacccagtctccatcttccttgtctgca tctgtaggagacagagtcaccatcacttgcaaggccagtcgtaatatcgagcgtcag ctggcctggtaccagcagaaacctggccaggctcccgagctcctcatctatcaggca tccagaaaggagagtggcgtgccagataggttcagtggcagtagatatgggacagac ttcactctcaccatcagcagcctgcagcctgaagatatcgccacatattactgtcag cagtatagcgatcctccgctgacttttggcgggggcaccaaggtggagatcaaacgt acggtgggtggaggcggttcaggcggaggtggctctggcggtggcggatcggcacct gagttcctcgggggaccatcagtcttcctgttccccccaaaacccaaggacactctc atgatctcccggacccctgaggtcacgtgcgtggtggtggacgtgagccaggaagac cccgaggtccagttcaactggtacgtggatggcgtggaggtgcataatgccaagaca aagccgcgggaggagcagtacaacagcacgtaccgtgtggtcagcgtcctcaccgtc ctgcaccaggactggctgaacggcaaggagtacaagtgcaaggtctccaacaaaggc ctcccgtcctccatcgagaaaaccatctccaaagccaaagggcagccccgagagcca caggtgtacaccctgcccccatcccagaaggagatgaccaagaaccaggtcagcctg acctgcctggtcaaaggcttctaccccagcgacatcgccgtggagtgggagagcaat gggcagccggagaacaactacaagaccacgcctcccgtgctggactccgacggctcc ttcttcctctacagcaagctaaccgtggacaagagcaggtggcaggaggggaatgtc ttctcatgctccgtgatgcatgaggctctgcacaacaggtacacacagaagagcctc tccctgtctccgggtggaggcggttcaggcggaggtggctctggcggtggcggatcg gtaaatggatgttccacacccctgggtatggaaaatggaaagatagaaaacaagcaa atcacagcttcttcgtttaagaaatcttggtggggagattactgggaacccttccgt
gcccgtctgaatgcccagggacgtgtgaatgcctggcaagccaaggcaaacaacaat aagcagtggctagaaattgatctactcaagatcaagaagataacggcaattataaca cagggctgcaagtctctgtcctctgaaatgtatgtaaagagctataccatccactac agtgagcagggagtggaatggaaaccatacaggctgaaatcctccatggtggacaag atttttgaaggaaatactaataccaaaggacatgtgaagaactttttcaacccccca atcatttccaggtttatccgtgtcattcctaaaacatggaatcaaagtattacactt cgcctggaactctttggctgtgatatttacggacatcatcaccatcaccattag (SEQ ID NO: 48) His-tagged VH9VLC1C2 caagtgcaactggtggaatccggcggaggactggtgcagcccggcggcagcctgagg ctgtcttgtgctgcttctggcttcaccttctcctattacgatatccagtgggtgcgg caggcccctggaaaaggcctggaatgggtctccagcatctccccatccggccagagc acctactacagaagagaggtgaagggcagattcacaatcagcagagataacagcaag aacaccctctacctgcagatgaatagcctgcgggccgaggacaccgccgtgtactac tgcgccagaagaaccggcagagagtacggcggcggctggtacttcgactactggggc cagggcacactggttacagtgtccagcgccggtggaggcggttcaggcggaggtggc tctggcggtggcggatcggacatccagatgacccagtctccatcttccttgtctgca tctgtaggagacagagtcaccatcacttgcaaggccagtcgtaatatcgagcgtcag ctggcctggtaccagcagaaacctggccaggctcccgagctcctcatctatcaggca tccagaaaggagagtggcgtgccagataggttcagtggcagtagatatgggacagac ttcactctcaccatcagcagcctgcagcctgaagatatcgccacatattactgtcag cagtatagcgatcctccgctgacttttggcgggggcaccaaggtggagatcaaacgt acggtgggtggaggcggttcaggcggaggtggctctggcggtggcggatcgcttatc atggacagagactgtaggatgccaatgggactaagcactggtatcatatctgattca cagatcaaggcttcagagtttctgggttactgggagcccagattagcaagattaaac aatggtggatcttataatgcttggagtgtagaaaaacttgcagcagaatttgcctct aaaccttggatccaggtggacatgcaaaaggaagtcataatcacagggatccagacc caaggtgccaaacactacctgaagtcctgctataccacagagttctatgtagcttac agttccaaccagatcaactggcagatcttcaaagggaacagcacaaggaatgtgatg tattttaatggcaattcagatgcctctacaataaaagagaatcagtttgacccacct attgtggctagatatattaggatctctccaactcgagcctataacagacctaccctt cgattggaactgcaaggttgtgaggtaaatggatgttccacacccctgggtatggaa aatggaaagatagaaaacaagcaaatcacagcttcttcgtttaagaaatcttggtgg ggagattactgggaacccttccgtgcccgtctgaatgcccagggacgtgtgaatgcc tggcaagccaaggcaaacaacaataagcagtggctagaaattgatctactcaagatc aagaagataacggcaattataacacagggctgcaagtctctgtcctctgaaatgtat gtaaagagctataccatccactacagtgagcagggagtggaatggaaaccatacagg ctgaaatcctccatggtggacaagatttttgaaggaaatactaataccaaaggacat gtgaagaactttttcaaccccccaatcatttccaggtttatccgtgtcattcctaaa acatggaatcaaagtattacacttcgcctggaactctttggctgtgatatttaccat catcaccatcaccattag (SEQ ID NO: 49) His-tagged Emi VH9VL-VH10VL caagtgcaactggtggaatccggcggaggactggtgcagcccggcggcagcctgagg ctgtcttgtgctgcttctggcttcaccttctcctattacgatatccagtgggtgcgg caggcccctggaaaaggcctggaatgggtctccagcatctccccatccggccagagc acctactacagaagagaggtgaagggcagattcacaatcagcagagataacagcaag aacaccctctacctgcagatgaatagcctgcgggccgaggacaccgccgtgtactac tgcgccagaagaaccggcagagagtacggcggcggctggtacttcgactactggggc cagggcacactggttacagtgtccagcgccggtggaggcggttcaggcggaggtggc
tctggcggtggcggatcggacatccagatgacccagtctccatcttccttgtctgca tctgtaggagacagagtcaccatcacttgcaaggccagtcgtaatatcgagcgtcag ctggcctggtaccagcagaaacctggccaggctcccgagctcctcatctatcaggca tccagaaaggagagtggcgtgccagataggttcagtggcagtagatatgggacagac ttcactctcaccatcagcagcctgcagcctgaagatatcgccacatattactgtcag cagtatagcgatcctccgctgacttttggcgggggcaccaaggtggagatcaaacgt acggtgggtggaggcggttcaggcggaggtggctctggcggtggcggatcgcaagtg caactggtgcaatccggcagcgaactgaagaagcccggcgcttctgttaaggtgtct tgtaaggcttctggctataccttcaccgacaacaacatggactgggtgcggcaggcc cctggacagggcctggaatggatgggcgacatcaacaccagatccggcggaagcatc tacaacgaagagttccaggatagagtgatcatgaccgtggataaaagcaccgatacc gcctacatggagctgtccagcctgcggagcgaggacaccgccacctaccattgcgcc agaagaaagagctacggctactacctggacgagtggggcgagggcacactggttaca gtgtccagcgccggtggaggcggttcaggcggaggtggctctggcggtggcggatcg gacatccagatgacccagtctccatcttccttgtctgcatctgtaggagacagagtc accatcacttgcaaggccagtcgtaatatcgagcgtcagctggcctggtaccagcag aaacctggccaggctcccgagctcctcatctatcaggcatccagaaaggagagtggc gtgccagataggttcagtggcagtagatatgggacagacttcactctcaccatcagc agcctgcagcctgaagatatcgccacatattactgtcagcagtatagcgatcctccg ctgacttttggcgggggcaccaaggtggagatcaaacgtacggtgcatcatcaccat caccattag (SEQ ID NO: 50) His-tagged Mim8 VH9VLC2 gaagtgcagctggtggagtctgggggaggcttggtacagcctggcaggtccctgaga ctctcctgtgcagcctctggattcacctttcacgattatgccatgcactgggtccgg caagtgccagggaagggcctggagtgggtctcggggatcagttggagaggcgatatt ggcggctatgtgaagtctgtgaagggccgattcaccatctccagagacaacgccaag aactccctgtatctgcaaatgaacagtctgagagctgaggacaccgccttgtattac tgtgtgaaaagttatggttcggggagtttttataacgcctttgactcctggggccag ggaaccctggtcaccgtctcctcagctggtggaggcggttcaggcggaggtggctct ggcggtggcggatcggacatccagatgacccagtctccttccaccctgtctgcatct gtaggagacagagtcaccatcacttgccgggccagtcagagtattagtagctggttg gcctggtatcagcagaaaccagggaaagcccctaaattcctgatctataaggcatct aaattagaaagagggacaccatcaaggttcagcggcagtggcgatgggacagaattc tctctcaccatcagcagcctgcagcctgatgattttgcaacttattactgcctggaa tattctagttatatccggacgttcggccaagggaccaaggtggaaatcaaacgaact gtgggtggaggcggttcaggcggaggtggctctggcggtggcggatcggtaaatgga tgttccacacccctgggtatggaaaatggaaagatagaaaacaagcaaatcacagct tcttcgtttaagaaatcttggtggggagattactgggaacccttccgtgcccgtctg aatgcccagggacgtgtgaatgcctggcaagccaaggcaaacaacaataagcagtgg ctagaaattgatctactcaagatcaagaagataacggcaattataacacagggctgc aagtctctgtcctctgaaatgtatgtaaagagctataccatccactacagtgagcag ggagtggaatggaaaccatacaggctgaaatcctccatggtggacaagatttttgaa ggaaatactaataccaaaggacatgtgaagaactttttcaaccccccaatcatttcc aggtttatccgtgtcattcctaaaacatggaatcaaagtattacacttcgcctggaa ctctttggctgtgatatttacggacatcatcaccatcaccattag (SEQ ID NO: 51). His-tagged Duo gaagtgcagctggtggagtctgggggaggcttggtacagcctggcaggtccctgaga ctctcctgtgcagcctctggattcacctttcacgattatgccatgcactgggtccgg
caagtgccagggaagggcctggagtgggtctcggggatcagttggagaggcgatatt ggcggctatgtgaagtctgtgaagggccgattcaccatctccagagacaacgccaag aactccctgtatctgcaaatgaacagtctgagagctgaggacaccgccttgtattac tgtgtgaaaagttatggttcggggagtttttataacgcctttgactcctggggccag ggaaccctggtcaccgtctcctcagctggtggaggcggttcaggcggaggtggctct ggcggtggcggatcggacatccagatgacccagtctccttccaccctgtctgcatct gtaggagacagagtcaccatcacttgccgggccagtcagagtattagtagctggttg gcctggtatcagcagaaaccagggaaagcccctaaattcctgatctataaggcatct aaattagaaagagggacaccatcaaggttcagcggcagtggcgatgggacagaattc tctctcaccatcagcagcctgcagcctgatgattttgcaacttattactgcctggaa tattctagttatatccggacgttcggccaagggaccaaggtggaaatcaaacgaact gtgggtggaggcggttcaggcggaggtggctctggcggtggcggatcgcaagtgcaa ctggtggaatccggcggaggactggtgcagcccggcggcagcctgaggctgtcttgt gctgcttctggcttcaccttctcctattacgatatccagtgggtgcggcaggcccct ggaaaaggcctggaatgggtctccagcatctccccatccggccagagcacctactac agaagagaggtgaagggcagattcacaatcagcagagataacagcaagaacaccctc tacctgcagatgaatagcctgcgggccgaggacaccgccgtgtactactgcgccaga agaaccggcagagagtacggcggcggctggtacttcgactactggggccagggcaca ctggttacagtgtccagcgccggtggaggcggttcaggcggaggtggctctggcggt ggcggatcggacatccagatgacccagtctccatcttccttgtctgcatctgtagga gacagagtcaccatcacttgcaaggccagtcgtaatatcgagcgtcagctggcctgg taccagcagaaacctggccaggctcccgagctcctcatctatcaggcatccagaaag gagagtggcgtgccagataggttcagtggcagtagatatgggacagacttcactctc accatcagcagcctgcagcctgaagatatcgccacatattactgtcagcagtatagc gatcctccgctgacttttggcgggggcaccaaggtggagatcaaacgtacggtgcat catcaccatcaccattag (SEQ ID NO: 52). His-tagged DuoC2 gaagtgcagctggtggagtctgggggaggcttggtacagcctggcaggtccctgaga ctctcctgtgcagcctctggattcacctttcacgattatgccatgcactgggtccgg caagtgccagggaagggcctggagtgggtctcggggatcagttggagaggcgatatt ggcggctatgtgaagtctgtgaagggccgattcaccatctccagagacaacgccaag aactccctgtatctgcaaatgaacagtctgagagctgaggacaccgccttgtattac tgtgtgaaaagttatggttcggggagtttttataacgcctttgactcctggggccag ggaaccctggtcaccgtctcctcagctggtggaggcggttcaggcggaggtggctct ggcggtggcggatcggacatccagatgacccagtctccttccaccctgtctgcatct gtaggagacagagtcaccatcacttgccgggccagtcagagtattagtagctggttg gcctggtatcagcagaaaccagggaaagcccctaaattcctgatctataaggcatct aaattagaaagagggacaccatcaaggttcagcggcagtggcgatgggacagaattc tctctcaccatcagcagcctgcagcctgatgattttgcaacttattactgcctggaa tattctagttatatccggacgttcggccaagggaccaaggtggaaatcaaacgaact gtgggtggaggcggttcaggcggaggtggctctggcggtggcggatcgcaagtgcaa ctggtggaatccggcggaggactggtgcagcccggcggcagcctgaggctgtcttgt gctgcttctggcttcaccttctcctattacgatatccagtgggtgcggcaggcccct ggaaaaggcctggaatgggtctccagcatctccccatccggccagagcacctactac agaagagaggtgaagggcagattcacaatcagcagagataacagcaagaacaccctc tacctgcagatgaatagcctgcgggccgaggacaccgccgtgtactactgcgccaga agaaccggcagagagtacggcggcggctggtacttcgactactggggccagggcaca ctggttacagtgtccagcgccggtggaggcggttcaggcggaggtggctctggcggt ggcggatcggacatccagatgacccagtctccatcttccttgtctgcatctgtagga gacagagtcaccatcacttgcaaggccagtcgtaatatcgagcgtcagctggcctgg
taccagcagaaacctggccaggctcccgagctcctcatctatcaggcatccagaaag gagagtggcgtgccagataggttcagtggcagtagatatgggacagacttcactctc accatcagcagcctgcagcctgaagatatcgccacatattactgtcagcagtatagc gatcctccgctgacttttggcgggggcaccaaggtggagatcaaacgtacggtgggt ggaggcggttcaggcggaggtggctctggcggtggcggatcggtaaatggatgttcc acacccctgggtatggaaaatggaaagatagaaaacaagcaaatcacagcttcttcg tttaagaaatcttggtggggagattactgggaacccttccgtgcccgtctgaatgcc cagggacgtgtgaatgcctggcaagccaaggcaaacaacaataagcagtggctagaa attgatctactcaagatcaagaagataacggcaattataacacagggctgcaagtct ctgtcctctgaaatgtatgtaaagagctataccatccactacagtgagcagggagtg gaatggaaaccatacaggctgaaatcctccatggtggacaagatttttgaaggaaat actaataccaaaggacatgtgaagaactttttcaaccccccaatcatttccaggttt atccgtgtcattcctaaaacatggaatcaaagtattacacttcgcctggaactcttt ggctgtgatatttacggacatcatcaccatcaccattag (SEQ ID NO: 53). Human albumin fused Emi VH9VL FVC2 gatgcacacaagagtgaggttgctcatcggtttaaagatttgggagaagaaaatttc aaagccttggtgttgattgcctttgctcagtatcttcagcagtgtccatttgaagat catgtaaaattagtgaatgaagtaactgaatttgcaaaaacatgtgttgctgatgag tcagctgaaaattgtgacaaatcacttcataccctttttggagacaaattatgcaca gttgcaactcttcgtgaaacctatggtgaaatggctgactgctgtgcaaaacaagaa cctgagagaaatgaatgcttcttgcaacacaaagatgacaacccaaacctcccccga ttggtgagaccagaggttgatgtgatgtgcactgcttttcatgacaatgaagagaca tttttgaaaaaatacttatatgaaattgccagaagacatccttacttttatgccccg gaactccttttctttgctaaaaggtataaagctgcttttacagaatgttgccaagct gctgataaagctgcctgcctgttgccaaagctcgatgaacttcgggatgaagggaag gcttcgtctgccaaacagagactcaagtgtgccagtctccaaaaatttggagaaaga gctttcaaagcatgggcagtagctcgcctgagccagagatttcccaaagctgagttt gcagaagtttccaagttagtgacagatcttaccaaagtccacacggaatgctgccat ggagatctgcttgaatgtgctgatgacagggcggaccttgccaagtatatctgtgaa aatcaagattcgatctccagtaaactgaaggaatgctgtgaaaaacctctgttggaa aaatcccactgcattgccgaagtggaaaatgatgagatgcctgctgacttgccttca ttagctgctgattttgttgaaagtaaggatgtttgcaaaaactatgctgaggcaaag gatgtcttcctgggcatgtttttgtatgaatatgcaagaaggcatcctgattactct gtcgtgctgctgctgagacttgccaagacatatgaaaccactctagagaagtgctgt gccgctgcagatcctcatgaatgctatgccaaagtgttcgatgaatttaaacctctt gtggaagagcctcagaatttaatcaaacaaaattgtgagctttttgagcagcttgga gagtacaaattccagaatgcgctattagttcgttacaccaagaaagtaccccaagtg tcaactccaactcttgtagaggtctcaagaaacctaggaaaagtgggcagcaaatgt tgtaaacatcctgaagcaaaaagaatgccctgtgcagaagactatctatccgtggtc ctgaaccagttatgtgtgttgcatgagaaaacgccagtaagtgacagagtcaccaaa tgctgcacagaatccttggtgaacaggcgaccatgcttttcagctctggaagtcgat gaaacatacgttcccaaagagtttaatgctgaaacattcaccttccatgcagatata tgcacactttctgagaaggagagacaaatcaagaaacaaactgcacttgttgagctc gtgaaacacaagcccaaggcaacaaaagagcaactgaaagctgttatggatgatttc gcagcttttgtagagaagtgctgcaaggctgacgataaggagacctgctttgccgag gagggtaaaaaacttgttgctgcaagtcaagctgccttaggcttatctgtttcacaa acttctaagctcacccgtgctgagactgtttttcctgatgtggacggttcacaagtg caactggtggaatccggcggaggactggtgcagcccggcggcagcctgaggctgtct tgtgctgcttctggcttcaccttctcctattacgatatccagtgggtgcggcaggcc
cctggaaaaggcctggaatgggtctccagcatctccccatccggccagagcacctac tacagaagagaggtgaagggcagattcacaatcagcagagataacagcaagaacacc ctctacctgcagatgaatagcctgcgggccgaggacaccgccgtgtactactgcgcc agaagaaccggcagagagtacggcggcggctggtacttcgactactggggccagggc acactggttacagtgtccagcgccggtggaggcggttcaggcggaggtggctctggc ggtggcggatcggacatccagatgacccagtctccatcttccttgtctgcatctgta ggagacagagtcaccatcacttgcaaggccagtcgtaatatcgagcgtcagctggcc tggtaccagcagaaacctggccaggctcccgagctcctcatctatcaggcatccaga aaggagagtggcgtgccagataggttcagtggcagtagatatgggacagacttcact ctcaccatcagcagcctgcagcctgaagatatcgccacatattactgtcagcagtat agcgatcctccgctgacttttggcgggggcaccaaggtggagatcaaacgtacggtg ggtggaggcggttcaggcggaggtggctctggcggtggcggatcggtaaatggatgt tccacacccctgggtatggaaaatggaaagatagaaaacaagcaaatcacagcttct tcgtttaagaaatcttggtggggagattactgggaacccttccgtgcccgtctgaat gcccagggacgtgtgaatgcctggcaagccaaggcaaacaacaataagcagtggcta gaaattgatctactcaagatcaagaagataacggcaattataacacagggctgcaag tctctgtcctctgaaatgtatgtaaagagctataccatccactacagtgagcaggga gtggaatggaaaccatacaggctgaaatcctccatggtggacaagatttttgaagga aatactaataccaaaggacatgtgaagaactttttcaaccccccaatcatttccagg tttatccgtgtcattcctaaaacatggaatcaaagtattacacttcgcctggaactc tttggctgtgatatttacggacatcatcaccatcaccattag (SEQ ID NO: 66). In certain embodiments, the nucleic acid molecules comprise a nucleotide sequence having at least 90%, 95%, 97%, 99%, or 100% homology or identity with any of the sequences provided above (e.g., any of SEQ ID NOs: 45-53, 61, and 66) or a nucleotide sequence encoding any of the amino sequences provided herein. Compositions comprising a nucleic acid molecule of the instant invention and a carrier such as a pharmaceutically acceptable carrier are also encompassed by the instant invention. In a particular embodiment, the composition comprises at least one nucleic acid molecule and at least one carrier (e.g., a pharmaceutically acceptable carrier). In certain embodiments, the nucleic acid molecules are contained within a lipid nanoparticle. The antibody and antibody fragments of the invention may be prepared using a variety of methods known in the art. The antibodies or antibody fragments of the instant invention may be prepared by inserting amino acid sequences into a backbone of an antibody or antibody fragment construct (e.g., an antibody framework), particularly a human construct/framework. For example, the variable light domain and/or variable heavy domain of the antibodies or antibody fragments of the instant invention or the CDRs contained therein may be inserted into another antibody construct or framework, particularly a human construct/framework. Methods for recombinantly producing antibodies are well-known in the art. Indeed, commercial
vectors for certain antibody and antibody fragment constructs are available. In certain embodiments, the antibody molecules are produced by expression of recombinant antibody or antibody fragments in host cells. The nucleic acid molecules encoding the antibody may be inserted into expression vectors and introduced into host cells. The resulting antibody molecules are then isolated and purified from the expression system. The antibodies optionally comprise a purification tag (e.g., His-tag) by which the antibody can be purified. The purity of the antibody and antibody fragments of the invention may be assessed using standard methods known to those of skill in the art, including, but not limited to, ELISA, immunohistochemistry, ion-exchange chromatography, affinity chromatography, immobilized metal affinity chromatography (IMAC), size exclusion chromatography, polyacrylamide gel electrophoresis (PAGE), western blotting, surface plasmon resonance and mass spectroscopy. The instant invention encompasses methods of inhibiting, treating, and/or preventing a hemostasis disorder, particularly hemophilia (e.g., hemophilia A) (e.g., in a subject in need thereof). The instant invention also encompasses methods of increasing the coagulation of blood (e.g., compared to the blood without treatment or to wild-type blood). In certain embodiments, the blood is FVIII deficient. In certain embodiments, the hemostasis disorder is a bleeding disorder. In certain embodiments, the hemophilia is hemophilia A. In certain embodiments, the hemophilia is hemophilia A without FVIII inhibitors. In certain embodiments, the hemophilia is hemophilia B, particularly hemophilia B with FIXa mutants. The antibody and antibody fragments of the instant invention, particularly those comprising a membrane binding domain, may be used to rescue dysfunctional hemophilia B causing mutants. Without being bound by theory, the antibody or antibody fragments of the instant invention may anchor dysfunctional FIXa to the membrane surface to increase its catalytic activity towards FX. The methods of the instant invention may be performed in vitro or in vivo. For example, the method may decrease the clot time of the blood (e.g., compared to the blood clot time without treatment or to wild-type blood). In certain embodiments, the method comprises administering to a subject in need thereof a therapeutically effective amount of an antibody or antibody fragment of the instant invention. In certain embodiments, the subject is a human. In certain embodiments, the subject is a canine. Antibody or antibody fragments may be administered alone or in combination
with other agents known to modulate hemostasis. The antibody or antibody fragment of the instant invention may be administered as part of a composition with a pharmaceutically acceptable carrier. The compositions of the instant invention may be conveniently formulated for administration with any carrier, particularly any pharmaceutically acceptable carrier(s). Except insofar as any conventional carrier is incompatible with the agents to be administered (e.g., antibody or antibody fragment or nucleic acid molecules encoding the same), its use in the pharmaceutical composition is contemplated. For example, the active agents may be formulated with an acceptable medium such as sterile liquid, water, aqueous solutions, buffered saline, ethanol, polyol (for example, glycerol, propylene glycol, liquid polyethylene glycol and the like), dimethyl sulfoxide (DMSO), oils, detergents, suspending agents or suitable mixtures thereof. The concentration of the active agents in the chosen medium may be varied and the medium may be chosen based on the desired route of administration of the pharmaceutical preparation. Except insofar as any conventional media or agent is incompatible with the active agents to be administered, its use in the pharmaceutical preparation is contemplated. The compositions of the present invention can be administered by any suitable route, for example, by infusion, injection or other modes of administration such as controlled release devices. In certain embodiments, the composition is delivered by injection. In certain embodiments, the composition is delivered by intravenous injection. In certain embodiments, the composition is delivered subcutaneously. The compositions of the instant invention may be directly administered or applied to the site of bleeding (e.g., by injection). In general, pharmaceutical compositions and carriers of the present invention comprise, among other things, pharmaceutically acceptable buffers, diluents, liquids (such as water, saline, glycerol, sugars and ethanol), preservatives, stabilizing agents, solubilizers, emulsifiers, wetting agents, pH buffering substances adjuvants and/or carriers. Such compositions can include diluents of various buffer content (e.g., saline, Tris HCl, acetate, phosphate), pH and ionic strength; and additives such as detergents and solubilizing agents (e.g., polysorbate 80), anti-oxidants (e.g., ascorbic acid, sodium metabisulfite), preservatives (e.g., benzyl alcohol) and bulking substances (e.g., lactose, mannitol). For example, the preparation can be formulated with a buffer containing salts, such as NaCl, CaCl2, and amino acids, such as glycine and/or lysine, and in a pH range from 6
to 8. The pharmaceutical compositions may be formulated in aqueous solutions (e.g., physiologically compatible buffers). Aqueous injection suspensions may contain substances which increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, or dextran. Additionally, suspensions of the active compounds may be prepared as appropriate oily injection suspensions. Suitable lipophilic solvents or vehicles include fatty oils such as sesame oil, or synthetic fatty acid esters, such as ethyl oleate or triglycerides, or liposomes. Optionally, the suspension may also contain suitable stabilizers or agents which increase the solubility of the compounds to allow for the preparation of highly concentrated solutions. The compositions of the invention may also be incorporated into particulate preparations of polymeric compounds such as polylactic acid, polyglycolic acid, etc., or into liposomes or micelles, or mixed with phospholipids or micelles to increase stability. Such compositions may influence the physical state, stability, rate of in vivo release, and rate of in vivo clearance of components of a pharmaceutical composition of the present invention. Exemplary pharmaceutical compositions and carriers are provided, e.g., in “Remington’s Pharmaceutical Sciences” by E.W. Martin (Mack Pub. Co., Easton, Pa.) and “Remington: The Science and Practice Of Pharmacy” by Alfonso R. Gennaro (Lippincott Williams & Wilkins) which are herein incorporated by reference. The pharmaceutical composition of the present invention can be prepared, for example, in liquid form, deep-frozen, or can be in dried powder form (e.g., lyophilized). In a particular embodiment, when the preparation is stored in lyophilized form, it may be dissolved into a visually clear solution using an appropriate reconstitution solution prior to administration. The compositions described herein will generally be administered to a patient as a pharmaceutical preparation. The term “patient” or “subject”, as used herein, refers to human or animal subjects (e.g., canine). The compositions of the instant invention may be employed therapeutically, under the guidance of a physician. The dose and dosage regimen of the compositions according to the invention that are suitable for administration to a particular patient may be determined by a physician considering the patient’s age, sex, weight, general medical condition, and the specific condition for which the active agent is being administered and the severity thereof (e.g., the severity of the bleeding). The physician may also take into account the route of administration, the pharmaceutical carrier, and the particular agent’s biological activity.
Selection of a suitable pharmaceutical preparation will also depend upon the mode of administration chosen. For example, the compositions of the invention may be administered by direct injection to a desired site. In this instance, a pharmaceutical preparation comprises the active agents of the instant invention dispersed in a medium that is compatible with the site of injection. The compositions of the instant invention may be administered by any method. For example, the compositions can be administered, without limitation, intravenously. Pharmaceutical preparations for injection are known in the art. If injection is selected as a method for administering the compositions, steps must be taken to ensure that sufficient amounts of the molecules reach their target cells to exert a biological effect. A pharmaceutical preparation of the invention may be formulated in dosage unit form for ease of administration and uniformity of dosage. Dosage unit form, as used herein, refers to a physically discrete unit of the pharmaceutical preparation appropriate for the patient undergoing treatment. Each dosage should contain a quantity of active ingredient calculated to produce the desired effect in association with the selected pharmaceutical carrier. Procedures for determining the appropriate dosage unit are well known to those skilled in the art. Dosage units may be proportionately increased or decreased based on the weight of the patient. Appropriate concentrations for alleviation of a particular pathological condition may be determined by dosage concentration curve calculations, as known in the art. In accordance with the present invention, the appropriate dosage unit for the administration of the composition may be determined by evaluating the toxicity of the molecules or cells in animal models. Various concentrations of active agents in pharmaceutical preparations may be administered to mice or other animal models, and the minimal and maximal dosages may be determined based on the beneficial results and side effects observed as a result of the treatment. Appropriate dosage unit may also be determined by assessing the efficacy of the treatment in combination with other standard drugs. The dosage units of the compositions of the instant invention may be determined individually or in combination with each treatment according to the effect detected. Nucleic acid molecules encoding the antibody or antibody fragment may be prepared by any method known in the art. Antibody or antibody fragment encoding nucleic acid molecules of the invention include DNA, RNA, and fragments thereof which may be single- or double-stranded. For example, nucleic acid molecules
encoding the antibody or antibody fragment of the invention may be prepared by using recombinant DNA technology methods. The availability of nucleotide sequence information enables preparation of isolated nucleic acid molecules of the invention by a variety of means. For example, two distinct rAAV production platforms are generally used for hemophilia A clinical trial vectors: 1) transfection of mammalian HEK293 cells with plasmid DNA (rAAV-293) or 2) transduction by recombinant baculoviruses into insect Sf9 cell lines (rAAV-Sf9) (Smith, et al. (2018) Cell Gene Ther. Insights 4:815-825; Rumachik, et al. (2020) Molecular Therapy: Methods Clin. Development, 18:P98-118; Li, et al. (2020) Nat. Rev. Genet., 21(4):255-272; Kondratov, et al. (2017) Molecular Therapy 25(12):2661-2675). The nucleic acid molecules may be maintained in any convenient vector, particularly an expression vector. Nucleic acids of the present invention may also be maintained as RNA or DNA in any convenient vector or cloning vector. In a particular embodiment, the nucleic acids may be maintained in a vector suitable for expression in mammalian cells, particularly human cells. Vectors such as those described above comprise the regulatory elements necessary for expression of the DNA in the host cell positioned in such a manner as to permit expression of the DNA in the host or target cell (e.g., hepatocyte). Such regulatory elements required for expression include, but are not limited to, promoter sequences, transcription initiation sequences, and enhancer sequences. Generally, the nucleic acid molecules of the instant invention will be administered to a subject in a composition comprising at least one carrier. In certain embodiments, the nucleic acid molecules are contained within lipid nanoparticles. In certain embodiments, the nucleic acid molecules are contained within a viral vector (e.g., AAV vector). Except insofar as any conventional carrier is incompatible with the nucleic acid to be administered, its use in the pharmaceutical composition is contemplated. In a particular embodiment, the carrier is a pharmaceutically acceptable carrier for injection. In certain embodiments, the nucleic acid molecules of the instant invention are administered to a subject in a cell (e.g., a hepatocyte) wherein the cell may be maintained in a composition comprising at least one carrier. As explained herein, antibody or antibody fragment encoding nucleic acids of the instant invention may be used, for example, as therapeutic and/or prophylactic agents which modulate the blood coagulation cascade, particularly in subjects with hemophlia (e.g., subjects with hemophilia A). It is demonstrated herein that the
antibody or antibody fragment encoding nucleic acid molecules provide effective hemostasis. In a particular embodiment of the present invention, antibody or antibody fragment encoding nucleic acid molecules may be administered to a patient via injection in a biologically or pharmaceutically compatible carrier, e.g., via injection into the liver. The antibody or antibody fragment encoding nucleic acid molecules of the invention may optionally be encapsulated into liposomes or mixed with other phospholipids or micelles. Antibody or antibody fragment encoding nucleic acid molecules may be administered alone or in combination with other agents known to modulate hemostasis. An appropriate composition in which to deliver the antibody or antibody fragment encoding nucleic acid molecules may be determined by a medical practitioner upon consideration of a variety of physiological variables, including, but not limited to, the patient’s condition and hemodynamic state. A variety of compositions well suited for different applications and routes of administration are well known in the art and are described hereinbelow. Antibody or antibody fragment encoding nucleic acids may be used for a variety of purposes in accordance with the present invention. In a particular embodiment of the invention, a nucleic acid delivery vehicle (e.g., an expression vector such as a viral vector or plasmid) for modulating blood coagulation or treating hemophilia A is provided wherein the expression vector comprises a nucleic acid sequence coding for an antibody or antibody fragment as described herein. Expression vectors comprising antibody or antibody fragment encoding nucleic acid sequences may be administered alone, or in combination with other molecules useful for modulating hemostasis. According to the present invention, the expression vectors or combination of therapeutic agents may be administered to the patient alone or in a pharmaceutically acceptable or biologically compatible composition. In a particular embodiment of the invention, the expression vector comprising nucleic acid sequences encoding the antibody or antibody fragment is a viral vector. Viral vectors which may be used in the present invention include, but are not limited to, adeno-associated virus (AAV) vectors of multiple serotypes (e.g., AAV-1 to AAV- 12, particularly AAV-2, AAV-5, AAV-7, and AAV-8) and hybrid AAV vectors, lentivirus vectors and pseudo-typed lentivirus vectors (e.g., Ebola virus, vesicular stomatitis virus (VSV), and feline immunodeficiency virus (FIV)), herpes simplex
virus vectors, vaccinia virus vectors, and retroviral vectors. In a particular embodiment, the vector is an adeno-associated virus (AAV) vector. In a particular embodiment of the present invention, methods are provided for the administration of a viral vector comprising nucleic acid sequences encoding an antibody or antibody fragment. Adeno-associated virus vectors of utility in the methods of the present invention may include at least the essential parts of adeno- associated virus vector DNA. As described herein, expression of an antibody or antibody fragment following administration of such an adeno-associated virus vector serves to modulate hemostasis, particularly to enhance the procoagulation activity. Recombinant adeno-associated virus vectors have found broad utility for a variety of gene therapy applications. Their utility for such applications is due largely to the high efficiency of in vivo gene transfer achieved in a variety of organ contexts. Adeno-associated virus particles may be used to advantage as vehicles for adequate gene delivery. Such virions possess a number of desirable features for such applications, including: structural features related to being a double stranded DNA nonenveloped virus and biological features such as a tropism for the human respiratory system and gastrointestinal tract. Moreover, adeno-associated viruses are known to infect a wide variety of cell types in vivo and in vitro by receptor-mediated endocytosis. For some applications, an expression construct may further comprise regulatory elements which serve to drive expression in a particular cell (e.g., hepatocyte) or tissue type (e.g., liver). Such regulatory elements are known to those of skill in the art. The incorporation of tissue specific regulatory elements in the expression constructs of the present invention provides for at least partial tissue tropism for expression. For example, hematopoietic or liver specific promoters may also be used. As explained hereinabove, AAV for recombinant gene expression have been produced in the human embryonic kidney cell line 293 (Wright, Hum Gene Ther (2009) 20:698-706; Graham et al. (1977) J. Gen. Virol. 36:59-72). Briefly, AAV vectors are typically engineered from wild-type AAV, a single-stranded DNA virus that is non-pathogenic. The parent virus is non-pathogenic, the vectors have a broad host range, and they can infect both dividing and non-dividing cells. The vector is typically engineered from the virus by deleting the rep and cap genes and replacing these with the transgene of interest under the control of a specific promoter. For
recombinant AAV preparation, the upper size limit of the sequence that can be inserted between the two inverted terminal repeats (ITRs) is about 4.7 kb. Plasmids expressing an antibody or antibody fragment under the control of a promoter (e.g., the CMV promoter/enhancer) and a second plasmid supplying adenovirus helper functions along with a third plasmid containing the AAV-2 rep and cap genes may be used to produce AAV-2 vectors, while a plasmid containing either AAV-1, AAV-6, or AAV-8 cap genes and AAV-2 rep gene and ITR's may be used to produce the respective alternate serotype vectors (e.g., Gao et al. (2002) Proc. Natl. Acad. Sci. USA 99:11854-11859; Xiao et al., (1999) J. Virol. 73:3994-4003; Arruda et al., (2004) Blood 103:85-92). AAV vectors may be purified by repeated CsCl density gradient centrifugation and the titer of purified vectors determined by quantitative dot-blot hybridization. The expression vectors of the present invention may be incorporated into pharmaceutical compositions that may be delivered to a subject, so as to allow production of a biologically active protein (e.g., an antibody or antibody fragment). In a particular embodiment of the present invention, pharmaceutical compositions comprising sufficient genetic material to enable a recipient to produce a therapeutically effective amount of an antibody or antibody fragment can influence hemostasis in the subject. The compositions may be administered alone or in combination with at least one other agent, such as a stabilizing compound, which may be administered in any sterile, biocompatible pharmaceutical carrier, including, but not limited to, saline, buffered saline, dextrose, and water. The compositions may be administered to a patient alone, or in combination with other agents (e.g., co-factors) which influence hemostasis. In particular embodiments, the pharmaceutical compositions also contain a pharmaceutically acceptable excipient/carrier. Such excipients include any pharmaceutical agent that does not itself induce an immune response harmful to the individual receiving the composition, and which may be administered without undue toxicity. Pharmaceutically acceptable excipients include, but are not limited to, liquids such as water, saline, glycerol, sugars and ethanol. Pharmaceutically acceptable salts can also be included therein, for example, mineral acid salts such as hydrochlorides, hydrobromides, phosphates, sulfates, and the like; and the salts of organic acids such as acetates, propionates, malonates, benzoates, and the like. Additionally, auxiliary substances, such as wetting or emulsifying agents, pH
buffering substances, and the like, may be present in such vehicles. A thorough discussion of pharmaceutically acceptable excipients is available in Remington's Pharmaceutical Sciences. Antibody or antibody fragment encoding vectors of the present invention may be administered to a patient by any means known. Direct delivery of the pharmaceutical compositions in vivo may generally be accomplished via injection using a conventional syringe. In this regard, the compositions may be delivered subcutaneously, epidermally, intradermally, intrathecally, intraorbitally, intramucosally, intraperitoneally, intravenously, intraarterially, orally, intrahepatically or intramuscularly. In a particular embodiment, the antibody or antibody fragment encoding nucleic acid molecules are administered by injection. In a particular embodiment, the antibody or antibody fragment encoding nucleic acid molecules are administered to the bloodstream. In a particular embodiment, the antibody or antibody fragment encoding nucleic acid molecules are administered to the liver. The invention includes, but is not limited to, the embodiments of the following numbered paragraphs: 1. An antibody fragment comprising at least one complementarity determining region (CDR) of the activated Factor IX (FIXa) binding domain of an activated Factor VIII (FVIIIa) mimetic bispecific antibody. 2. The antibody fragment of paragraph 1, wherein said FVIIIa mimetic bispecific antibody is emicizumab or mim8. 3. The antibody fragment of paragraph 1 or 2, wherein said FVIIIa mimetic bispecific antibody is emicizumab. 4. The antibody fragment of any one of paragraphs 1-3, wherein said antibody fragment comprises all three CDRs of the heavy chain variable domain of the Factor IX binding domain of the FVIIIa mimetic bispecific antibody or comprises CDR1 and CDR2 of the heavy chain variable domain of the Factor IX binding domain of the FVIIIa mimetic bispecific antibody. 5. The antibody fragment of any one of paragraphs 1-4, wherein said antibody fragment comprises a single domain antibody. 6. The antibody fragment of any one of paragraphs 1-4, wherein said antibody fragment comprises a single chain variable fragment. 7. The antibody fragment of any one of paragraphs 1-4, wherein said antibody fragment comprises a first and a second single chain variable fragment.
8. The antibody fragment of paragraph 7, wherein said first and second single chain variable fragments bind FIXa. 9. The antibody fragment of paragraph 7, wherein said first single chain variable fragment binds FIXa and wherein said second single chain variable fragment binds Factor X. 10. The antibody fragment of any one of paragraphs 1-9, wherein said antibody fragment further comprises a membrane binding domain. 11. The antibody fragment of paragraph 10, wherein said membrane binding domain comprises the Factor V C2 domain. 12. The antibody fragment of paragraph 10, wherein said membrane binding domain comprises the lactadherin C2 domain. 13. The antibody fragment of any one of paragraphs 1-12, wherein said antibody fragment further comprises a Protein C tag. 14. The antibody of paragraph 13, wherein said Protein C tag is the HPC4 tag. 15. The antibody fragment of any one of paragraphs 1-14, wherein said antibody fragment further comprises a thrombin cleavage site. 16. The antibody fragment of paragraph 15, wherein said thrombin cleavage site is located N-terminal to a membrane binding domain. 17. The antibody fragment of paragraph 15, wherein said thrombin cleavage site is located at the C-terminus of the antibody fragment prior to any additional components. 18. The antibody fragment of any one of paragraphs 1-17, wherein said antibody fragment further comprises a dimerization domain. 19. The antibody fragment of paragraph 18, wherein said dimerization domain comprises CH2 and CH3 domains. 20. The antibody fragment of any one of paragraphs 1-19, wherein said antibody fragment further comprises albumin or an anti-albumin antibody or antigen binding fragment thereof. 21. A modified FVIIIa mimetic bispecific antibody, wherein said modified FVIIIa mimetic bispecific antibody comprises a FVIIIa mimetic bispecific antibody and a membrane binding domain. 22. The modified FVIIIa mimetic bispecific antibody of paragraph 31, wherein said membrane binding domain comprises the Factor V C2 domain.
23. The modified FVIIIa mimetic bispecific antibody of paragraph 21, wherein said membrane binding domain comprises the lactadherin C2 domain. 24. The modified FVIIIa mimetic bispecific antibody of any one of paragraphs 21-23, wherein said FVIIIa mimetic bispecific antibody is emicizumab or mim8. 25. A composition comprising an antibody fragment of any one of paragraphs 1-20 or an antibody of any one of paragraphs 21-24 and a carrier. 26. A nucleic acid molecule encoding an antibody fragment of any one of paragraphs 1-20 or an antibody of any one of paragraphs 21-24. 27. A vector comprising the nucleic acid molecule of paragraph 26. 28. The vector of paragraph 27, which is a viral vector. 29. The vector of paragraph 28, wherein said viral vector is an adeno- associated virus vector. 30. A composition comprising a nucleic acid molecule or vector of any one of paragraphs 26-29 and a carrier. 31. A method of treating and/or preventing hemophilia in a subject (e.g., human or canine) in need thereof, said method comprising administering to said subject an antibody fragment of any one of paragraphs 1-20 or an antibody of any one of paragraphs 21-24. 32. A method of treating and/or preventing hemophilia in a subject in need thereof, said method comprising administering to said subject a nucleic acid molecule or vector of any one of paragraphs 26-29. 33. A method of increasing blood coagulation, said method comprising contacting a blood sample with an antibody fragment of any one of paragraphs 1-20 or an antibody of any one of paragraphs 21-24. 34. A method of increasing blood coagulation, said method comprising contacting a blood sample with a nucleic acid molecule or vector of any one of paragraphs 26-29. Definitions The following definitions are provided to facilitate an understanding of the present invention: The singular forms “a,” “an,” and “the” include plural referents unless the context clearly dictates otherwise.
“Pharmaceutically acceptable” indicates approval by a regulatory agency of the Federal or a state government or listed in the U.S. Pharmacopeia or other generally recognized pharmacopeia for use in animals, and more particularly in humans. A “carrier” refers to, for example, a diluent, adjuvant, excipient, auxilliary agent or vehicle with which an active agent of the present invention is administered. Pharmaceutically acceptable carriers can be sterile liquids, such as water and oils, including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil and the like. Water or aqueous saline solutions and aqueous dextrose and glycerol solutions are preferably employed as carriers. Suitable pharmaceutical carriers are described, for example, in “Remington's Pharmaceutical Sciences” by E.W. Martin. An “antibody” or “antibody molecule” is any immunoglobulin, including antibodies and fragments thereof, that binds to a specific antigen. As used herein, antibody or antibody molecule contemplates intact immunoglobulin molecules, immunologically active portions of an immunoglobulin molecule (e.g., antigen- binding fragment), and fusions of immunologically active portions of an immunoglobulin molecule. As used herein, the term “immunologically specific” refers to proteins/polypeptides, particularly antibodies, that bind to one or more epitopes of a protein or compound of interest, but which do not substantially recognize and bind other molecules in a sample containing a mixed population of antigenic biological molecules. As used herein, the term “subject” refers to an animal, particularly a mammal, particularly a human. A “therapeutically effective amount” of a compound or a pharmaceutical composition refers to an amount effective to prevent, inhibit, treat, or lessen the symptoms of a particular disorder or disease. The treatment of a disease or disorder herein may refer to curing, relieving, and/or preventing the disease or disorder, the symptom(s) of it, or the predisposition towards it. As used herein, the term “therapeutic agent” refers to a chemical compound or biological molecule including, without limitation, nucleic acids, peptides, proteins, and antibodies that can be used to treat a condition, disease, or disorder or reduce the symptoms of the condition, disease, or disorder.
The term “isolated” refers to the separation of a compound from other components present during its production or from its natural environment. “Isolated” is not meant to exclude artificial or synthetic mixtures with other compounds or materials, or the presence of impurities that do not substantially interfere with the fundamental activity, and that may be present, for example, due to incomplete purification, or the addition of stabilizers. The following examples are provided to illustrate various embodiments of the present invention. The examples are not intended to limit the invention in any way. EXAMPLE 1 A single chain antibody fragment (scFv) of the FIXa binding arm of emicizumab was generated by joining the variable heavy chain (VH) with the variable light chain (VL) to produce VH9VL. The scFv, VH9VL, can only bind FIXa and imparted a modest cofactor mimetic activity, as compared to the full bispecific antibody, emicizumab. VH9VL was used as a template to introduce membrane binding properties. The Factor V C2-domain, which binds membrane, was fused at the C-terminus of VH9VL via a repeating (3x) Gly4Ser (SEQ ID NO: 54) flexible linker of ~ 5.7 nm to allow the VH and VL domains of the scFv to potentially orient correctly at the membrane surface. The resulting construct, VH9VLC2, exhibited a substantial amplification in cofactor mimetic activity, showing approximately 22-fold faster activation compared to VH9VL and nearly 10-fold greater activity than emicizumab (Fig. 2). VH9VLC2 added at 100 nM brought thrombin generation into the reference range in platelet-poor, severe congenital FVIII-deficient plasma supplemented with 4 µM PC:PS and triggered with either low tissue factor (0.1 pM) or factor XIa (0.1 nM) (Figs. 3A, 3B, and 4). The results in Figs 3A and 3B indicate that nearly three- to fourfold excess emicizumab is required to achieve thrombin levels equivalent to those of the fusion variant, Emi VH9VL FVC2, in the thrombin generation assays triggered by low tissue factor (0.1 pM). The endogenous thrombin potential (ETP), a parameter that reflects total thrombin generation quantitatively, was comparable in both emicizumab and VH9VLC2. Potent cofactor mimetic activity of VH9VLC2 observed in TGA was also reflected in FXa generation assay in a purified system. VH9VLC2 showed a
significant amplification in cofactor mimetic activity as compared to emicizumab. In vitro kinetic analyses with purified proteins and synthetic phospholipid vesicles containing phosphatidylcholine (PC) and phosphatidylserine (PS) (PC:PS, 75:25 %) showed approximately 2-3-fold faster activation of FX by VH9VLC2 as compared to emicizumab (Fig. 6). Remarkably, despite its inability to bind FX, cofactor mimetic activity of VH9VLC2 is higher than that of emicizumab. Light scattering measurements of synthetic membrane (phospholipid vesicles, PC:PS::75:25) binding by the scFv VH9VL-FIXa complex or the Emi VH9VL FVC2-FIXa complex is shown in Figure 5. The FV C2 domain in Emi VH9VL FVC2 colocalizes the mimetic-FIXa complex at the membrane surface whereas the scFv VH9VL-FIXa complex showed no significant membrane localization. The role of the C2 domain-mediated increase in cofactor activity of VH9VLC2 was further assessed by using an antibody fragment (E9scFv) that inhibits the binding of factor V to membranes (Fig. 7). Increasing concentrations of E9scFv caused a dose-dependent reduction in the rate of FXa formation catalyzed by the complex of VH9VLC2: FIXa (20nM:25nM). Inhibition saturated at ~ 85% at 100 nM E9scFv, indicating a substantial contribution of membrane binding to the cofactor mimetic function of VH9VLC2. To evaluate whether VH9VLC2 can restore clotting in HA plasma with FVIII inhibitors, normal pooled plasma was supplemented with either a FVIII neutralizing antibody fragment (F8scFv) or an inhibitory RNA aptamer to mimic FVIII inhibitor plasma. Addition of VH9VLC2 to this plasma restored clotting time to the levels shown by normal pooled plasma, indicating that VH9VLC2 can bypass FVIII inhibitor activity. The scFv-fc fusion, VH9VLCH2CH3, was recombinantly expressed and purified to homogeneity from HEK293 cells. The addition of constant region 2 (CH2), and constant region 3 (CH3) to this construct would lead to their homodimerization, therefore potentially increasing its circulating half-life when expressed in vivo for gene therapy. The FVIII mimetic activity of VH9VLCH2CH3 in severe congenital FVIII deficient plasma was about three-fold weaker than the scFv, VH9VL. The biparatropic scFv-scFv fusion, Duo, has been transfected in HEK293 cells. The FVIII mimetic activity of Duo (from unpurified media) is as potent as compared to scFv, VH9VL, when tested in severe congenital FVIII deficient plasma.
The fusion of a FV C2-domain to this construct will increase its cofactor mimetic activity several folds higher as compared to the monoparatopic ScFv, VH9VLC2. Based on the constructs described above, these new protein products can be used either in nonfactor replacement therapy (injectable) or in in vivo gene therapy. A single chain scFv, VH9VL, derived from emicizumab retains significant cofactor mimetic activity. The engineered scFv with a Factor V C2 domain fusion, VH9VLC2, demonstrates that the addition of a membrane binding feature to the FIXa arm of the emicizumab surpasses the mimetic activity achieved by a bispecific antibody that exploits both bridging and allostery to be an effective cofactor. This would also incorporate a built-in regulatability feature because membrane dependent mimetics could be regulated by an inhibitory antibody against FV C2-domain. Additionally, a construct containing CH2 and CH3 domain would dimerize spontaneously for an effective in vivo application. All single chain constructs described herein also avoid the burden of productive assembly from a combination of three separate polypeptide chains, as in a functional bispecific antibody. Replicating the membrane binding feature into bispecific FVIIIa mimetic antibodies could further amplify function and more closely resemble FVIIIa in both activity and membrane dependent regulation. The results provide surprising insights into how a single chain, monospecific, membrane-anchored FVIIIa mimetic enhances the catalytic activity of FIXa. The biparatropic scFv-scFv fusion, Duo, also shows an amplification in cofactor mimetic activity which can be further improved by fusing a membrane binding C2-domain at its C-terminus. Similarly, the scFv-fc fusion, VH9VLCH2CH3, which retains significant mimetic activity can be further improved by fusing a membrane binding domain at its C-terminus. EXAMPLE 2 Stable HEK 293 or AV12 transfectants, expressing emicizumab derived VH9VLC2 showed nearly 22-fold higher cofactor mimetic activity as compared to the scFv VH9VL. Remarkably, the activity of VH9VLC2 was about 3-4 fold higher as that of emicizumab in low tissue factor triggered thrombin generation assay. The activated partial thromboplastin time (aPTT) was used herein to evaluate the intrinsic coagulation pathway (Langdell, et al. (1953) J. Lab. Clin. Med., 41(4):637-47). This one-stage assay is the most commonly used method for testing FVIII or FIX activities. Accordingly, this assay is very useful to determine the FVIII
equivalency of FVIII mimetics when FVIII deficient plasma is used and supplemented either with FVIII or FVIII mimetic. Briefly, the aPTT assay with FVIII deficient plasma comprises the following steps: 1) Add metal ball to the test cuvette; 2) Pipette 50 µL of FVIII deficient plasma in a test cuvette (e.g., Stago™ semiautomated analyzer); 3) Add 3 µL of FVIII or mimetic from an initial stock to achieve the desired final concentrations; 4) Add 50 µL of aPTT reagent (TriniCLOT™ Automated aPTT; Stago) to the cuvette containing the plasma; 5) Incubate the mixture at 37°C for 3 minutes; 6) Start the reaction by adding 50 µL of prewarmed 25 mM calcium chloride solution; and 7) Record the clot time in seconds as shown by the analyzer. Here, a 12 amino acid HPC4 tag (EDQVDPRLIDGK; SEQ ID NO: 59) was added at the C-terminus of FV C2-domain in VH9VLC2 (Fig. 1K). This variant led to a further increase in the cofactor mimetic activity. In an aPTT assay with FVIII deficient plasma, Emi VH9VL FVC2 HPC4 achieved equivalent cofactor activity of FVIII (B-domain deleted) at a less than two-fold higher molar concentration (Fig. 8). This would translate to a nearly tenfold higher cofactor mimetic activity as compared to emicizumab in a thrombin generation assay with low tissue factor as a trigger. Without being bound by theory, the increased cofactor mimetic activity likely emanates from a lack of His tag based conformational changes or misfolding of FV C2-domain in the new construct. Similar enhanced cofactor activity was obtained when Emi VH9VL FVC2 was expressed without any tag, indicating that the His tag altered the activity of the VH9VLC2 variant when it was expressed with the tag. The cofactor mimetic activity of scFvs derived from Factor IXa binding arm of FVIII mimetic antibodies can be amplified by fusing a lactadherin C2-domain that binds to phosphatidylserine rich membrane surface similar to FVa or FVIIIa C2- domains. Emi VH9VL Lac C2 (Fig. 1L), derived from fusing lactadherin C2-domain at the C-terminus of emicizumab derived VH9VL showed a several fold amplifications in the cofactor mimetic activity as assessed by an aPTT assay in FVIII deficient plasma (Fig. 9). Without being bound by theory, the removal of the His tag and adding a HPC4 tag to this construct further increases its cofactor mimetic activity, nearing the level of FVIII. Another regulatory feature that can be embedded into every construct described in the present invention is a thrombin cleavage site, particularly a short thrombin cleavage peptide before the membrane binding domain (e.g., a C2-domain).
Figure 1M and 1N provide examples of two constructs comprising a short thrombin cleavage sequence. The presence of the thrombin cleavage site provides a physiological self-regulation mechanism that can inactivate the fusion constructs by liberating the membrane binding domain. This, in turn, can inhibit or prevent uncontrolled thrombotic events in a high-risk patient population, thereby providing a safer therapeutic alternative. In addition to the above, the circulating half-lives of the mimetics described in the present invention can be further increased by fusing them with albumin, antibody/antibody fragment, or a nanobody that targets neonatal Fc receptors directly or indirectly or bind albumin. EXAMPLE 3 Similar to emicizumab derived fusion variants (see, e.g., Fig. 2), a fusion variant comprising the mim8 FIXa binding arm derived scFv, Mim8 VH9VL, and a FVC2-domain also leads to significant amplification in the cofactor mimetic function as assessed by an aPTT assay using FVIII-deficient plasma (Figure 10). EXAMPLE 4 To prolong half-life and introduce inherent latency until activation, human albumin was fused to VH9VL FVC2 at the N-terminus, linked with a FIX activation peptide (IX AP) (Figure 1O). This fusion exploits cellular recycling via neonatal Fc receptor interaction, maintaining a latent state until triggered by a procoagulant stimulus that leads to the cleavage of FIX AP. The variants were recombinantly expressed and purified from mammalian cell culture system. The albumin-fused VH9VL FVC2 retained cofactor mimetic activity similar to VH9VL FVC2 and was cleaved by human FXIa in vitro (Figure 11). EXAMPLE 5 Hemophilic dogs provide a valuable animal model to study hemophilia and inhibitor development against FVIII. The canine hemophilia A (cHA) model has been one of the best predictors of efficacy of anti-hemophilic drugs. Notably, emicizumab does not work effectively in canine system, likely due to inefficient binding to canine FIXa or FX.
Hemophilic dogs with and without inhibitors, require effective anti- hemophilic therapy to prevent spontaneous episodes of bleeding or prolonged bleeding after surgery. Currently, the primary treatment available for hemophilic dogs involves repeated substitution therapy. Given the success of bispecific antibodies to treat hemophilia A in human, antibody based FVIII mimetics can be developed that can work effectively to prevent bleeding in hemophilia A dogs, irrespective of their inhibitor status. Such a mimetic could also be used as a novel transgene for a potential gene therapy in dogs. Both emicizumab and mim8 derived fusion variants shorten the aPTT time in canine FVIII deficient plasma and show potency superior to emicizumab (Figure 12A). The effect of mim8 variant is much more pronounced where the clot time would be comparable to normal canine plasma when supplemented with nearly 20 nM concentration of mim8 VH9VL FVC2. Interestingly, as per the crystal structure of mim8 anti-FIXa fragment in complex with des-(Gla-EGF1) FIXa, the residues of FIXa that make contact with FIXa binding arm of Mim 8 (Protein Data Bank ID: 7AHV), are mostly conserved in both human and canine (Figure 12B). This conservation may explain why the mim8-derived fusion variant results in a greater enhancement of cofactor activity. A caninized variant of Mim8 VH9VL, when fused with the canine FV C2 domain, is also a drug candidate for treating cHA. Additionally, these variants can also be used as novel transgenes for hemophilia A gene therapy in hemophilic dogs. The sequence conservation likely results in similar affinities for FIXa in both Mim8 and Mim8 VH9VL FVC2. EXAMPLE 6 FVIII mimetics have demonstrated consistent efficacy in the presence of FVIII inhibitor plasma, positioning them as promising therapeutic options for hemophilia A patients with inhibitors. This is particularly important, as the development of inhibitors can significantly complicate treatment and lead to poor clinical outcomes. Both emicizumab and mim8-derived fusion mimetics described in this application retain full cofactor activity in FVIII inhibitor plasma. For example, Figure 13 illustrates that the emicizumab-derived fusion mimetic, VH9VL FVC2, maintains its mimetic activity in FVIII inhibitor plasma. This characteristic is crucial, as it allows these mimetics to effectively bypass the inhibitory effects that typically impede the action of standard FVIII replacement therapies.
Several publications and patent documents are cited in the foregoing specification in order to more fully describe the state of the art to which this invention pertains. The disclosure of each of these citations is incorporated by reference herein. While certain of the preferred embodiments of the present invention have been described and specifically exemplified above, it is not intended that the invention be limited to such embodiments. Various modifications may be made thereto without departing from the scope and spirit of the present invention, as set forth in the following claims.
Claims
WHAT IS CLAIMED IS: 1. An antibody fragment comprising at least one complementarity determining region (CDR) of the activated Factor IX (FIXa) binding domain of an activated Factor VIII (FVIIIa) mimetic bispecific antibody.
2. The antibody fragment of claim 1, wherein said FVIIIa mimetic bispecific antibody is emicizumab or mim8.
3. The antibody fragment of claim 2, wherein said FVIIIa mimetic bispecific antibody is emicizumab.
4. The antibody fragment of any one of claims 1-3, wherein said antibody fragment comprises all three CDRs of the heavy chain variable domain of the Factor IX binding domain of the FVIIIa mimetic bispecific antibody.
5. The antibody fragment of any one of claims 1-4, wherein said antibody fragment comprises a single domain antibody.
6. The antibody fragment of any one of claims 1-4, wherein said antibody fragment comprises a single chain variable fragment.
7. The antibody fragment of any one of claims 1-4, wherein said antibody fragment comprises a first and a second single chain variable fragment.
8. The antibody fragment of claim 7, wherein said first and second single chain variable fragment bind FIXa.
9. The antibody fragment of claim 7, wherein said first single chain variable fragment binds FIXa and wherein said second single chain variable fragment binds Factor X.
10. The antibody fragment of any one of claims 1-9, wherein said antibody fragment further comprises a membrane binding domain.
11. The antibody fragment of claim 10, wherein said membrane binding domain comprises the Factor V C2 domain.
12. The antibody fragment of claim 10, wherein said membrane binding domain comprises the lactadherin C2 domain.
13. The antibody fragment of any one of claims 1-12, wherein said antibody fragment further comprises a Protein C tag.
14. The antibody of claim 13, wherein said Protein C tag is the HPC4 tag.
15. The antibody fragment of any one of claims 1-14, wherein said antibody fragment further comprises a thrombin cleavage site.
16. The antibody fragment of claim 15, wherein said thrombin cleavage site is located N-terminal to a membrane binding domain.
17. The antibody fragment of claim 15, wherein said thrombin cleavage site is located at the C-terminus prior to any additional components.
18. The antibody fragment of any one of claims 1-17, wherein said antibody fragment further comprises a dimerization domain.
19. The antibody fragment of claim 18, wherein said dimerization domain comprises CH2 and CH3 domains.
20. The antibody fragment of any one of claims 1-19, wherein said antibody fragment further comprises albumin or an anti-albumin antibody or antigen binding fragment thereof.
21. A composition comprising an antibody fragment of any one of claims 1-20 and a carrier.
22. A nucleic acid molecule encoding an antibody fragment of any one of claims 1- 20.
23. A vector comprising the nucleic acid molecule of claim 22.
24. The vector of claim 23, which is a viral vector.
25. The vector of claim 24, wherein said viral vector is an adeno-associated virus vector.
26. A composition comprising a nucleic acid molecule of any one of claims 22-25 and a carrier.
27. A method of treating and/or preventing hemophilia in a subject in need thereof, said method comprising administering to said subject an antibody fragment of any one of claims 1-20.
28. A method of treating and/or preventing hemophilia in a subject in need thereof, said method comprising administering to said subject a nucleic acid molecule of any one of claims 22-25.
29. A method of increasing blood coagulation, said method comprising contacting a blood sample with an antibody fragment of any one of claims 1-20.
30. A method of increasing blood coagulation, said method comprising contacting a blood sample with a nucleic acid molecule of any one of claims 22-25.
31. A modified FVIIIa mimetic bispecific antibody, wherein said modified FVIIIa mimetic bispecific antibody comprises a FVIIIa mimetic bispecific antibody and a membrane binding domain.
32. The modified FVIIIa mimetic bispecific antibody of claim 31, wherein said membrane binding domain comprises the Factor V C2 domain.
33. The modified FVIIIa mimetic bispecific antibody of claim 31, wherein said membrane binding domain comprises the lactadherin C2 domain.
34. The modified FVIIIa mimetic bispecific antibody of any one of claims 31-33, wherein said FVIIIa mimetic bispecific antibody is emicizumab or mim8.
Applications Claiming Priority (4)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| US202363588537P | 2023-10-06 | 2023-10-06 | |
| US63/588,537 | 2023-10-06 | ||
| US202463561872P | 2024-03-06 | 2024-03-06 | |
| US63/561,872 | 2024-03-06 |
Publications (1)
| Publication Number | Publication Date |
|---|---|
| WO2025076507A1 true WO2025076507A1 (en) | 2025-04-10 |
Family
ID=95283933
Family Applications (1)
| Application Number | Title | Priority Date | Filing Date |
|---|---|---|---|
| PCT/US2024/050186 Pending WO2025076507A1 (en) | 2023-10-06 | 2024-10-07 | Engineered proteins and methods of use thereof |
Country Status (1)
| Country | Link |
|---|---|
| WO (1) | WO2025076507A1 (en) |
Citations (4)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| US8062635B2 (en) * | 2003-10-10 | 2011-11-22 | Chugai Seiyaku Kabushiki Kaisha | Bispecific antibody substituting for functional proteins |
| US20210284715A1 (en) * | 2017-02-06 | 2021-09-16 | Applied Stemcell, Inc. | Coagulation factor viii mimetic protein and uses thereof |
| US20220089778A1 (en) * | 2016-09-23 | 2022-03-24 | Csl Limited | Coagulation factor binding proteins and uses thereof |
| US20230192896A1 (en) * | 2016-11-23 | 2023-06-22 | Bioverativ Therapeutics Inc. | Bispecific antibodies binding to coagulation factor ix and coagulation factor x |
-
2024
- 2024-10-07 WO PCT/US2024/050186 patent/WO2025076507A1/en active Pending
Patent Citations (4)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| US8062635B2 (en) * | 2003-10-10 | 2011-11-22 | Chugai Seiyaku Kabushiki Kaisha | Bispecific antibody substituting for functional proteins |
| US20220089778A1 (en) * | 2016-09-23 | 2022-03-24 | Csl Limited | Coagulation factor binding proteins and uses thereof |
| US20230192896A1 (en) * | 2016-11-23 | 2023-06-22 | Bioverativ Therapeutics Inc. | Bispecific antibodies binding to coagulation factor ix and coagulation factor x |
| US20210284715A1 (en) * | 2017-02-06 | 2021-09-16 | Applied Stemcell, Inc. | Coagulation factor viii mimetic protein and uses thereof |
Non-Patent Citations (2)
| Title |
|---|
| KUMAR SHEKHAR, KRISHNASWAMY SRIRAM: "Design of a Superior Factor VIII Mimetic with Membrane Binding Properties and Built-in Reversibility", INTERNATIONAL SOCIETY ON THROMBOSIS AND HAEMOSTASIS, 23 June 2024 (2024-06-23), pages 1, XP093302243, Retrieved from the Internet <URL:https://isth2024.eventscribe.net/fsPopup.asp?PresentationID=1432591&mode=presInfo> * |
| KUMAR SHEKHAR, KRISHNASWAMY SRIRAM: "Design of a Superior Factor VIIIa Mimetic By Coupling a Membrane Binding Domain to a Factor IXa Binding Antibody Fragment", BLOOD, vol. 142, no. Supplement 1, 2 November 2023 (2023-11-02), AMSTERDAM, NL, pages 1 - 2, XP093302239, ISSN: 0006-4971, DOI: 10.1182/blood-2023-187228 * |
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| EP3723858B1 (en) | Fixaxfx bispecific antibody with common light chain | |
| CA3174683A1 (en) | Modified angiotensin-converting enzyme 2 (ace2) and use thereof | |
| EA032189B1 (en) | Isolated human monoclonal antibodies against tissue factor pathway inhibitor (tfpi) and pharmaceutical composition comprising same | |
| EP1647596A2 (en) | Monoclonal antibody against platelet membrane glycoprotein vi | |
| JP6514893B2 (en) | Compositions and methods for modulating hemostasis | |
| JP2012513193A (en) | Antibodies against tissue factor pathway inhibitor (TFPI) | |
| TW201723180A (en) | Viral vectors encoding recombinant FVIII variants with increased expression for gene therapy of hemophilia A | |
| AU2017360979A1 (en) | Red blood cell targeted factor VIII and method of using the same | |
| US20240398911A1 (en) | Engineered receptors and monoclonal antibodies for cronaviruses and uses thereof | |
| CN113646331A (en) | single-chain factor VIII molecule | |
| JP7181855B2 (en) | Compositions and methods for modulating the function of Factor IX | |
| WO2025076507A1 (en) | Engineered proteins and methods of use thereof | |
| JP7576039B2 (en) | Factor IX variants and their use in therapy | |
| KR20250120344A (en) | transferrin receptor binding protein | |
| EP4069269A1 (en) | Compositions and methods for modulating factor viii function | |
| WO2020104480A1 (en) | Adeno-associated virus vectors for expressing fviii mimetics and uses thereof | |
| CN114391024A (en) | Factor VIII proteins with increased half-life | |
| US20230250412A1 (en) | Compositions and Methods for Modulating Factor IX Function | |
| US20190338269A1 (en) | Factor x variants | |
| CN115916836A (en) | Bispecific antigen binding molecules and related compositions, uses for preparing the compositions, kits and methods | |
| WO2025093734A1 (en) | Bispecific antigen binding protein with fviii mimetic activity | |
| EP3785726A1 (en) | Factor viii protein with increased half-life | |
| CN120322246A (en) | Compositions and methods for modulating Factor VIII function | |
| TW202530404A (en) | Polypeptides having human igg cleavage activity | |
| HK40042065B (en) | Fixaxfx bispecific antibody with common light chain |
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: 24875575 Country of ref document: EP Kind code of ref document: A1 |