Bispecific antibodies for factor IX and factor X

Information

  • Patent Grant
  • 11919969
  • Patent Number
    11,919,969
  • Date Filed
    Friday, June 22, 2018
    5 years ago
  • Date Issued
    Tuesday, March 5, 2024
    2 months ago
Abstract
Bispecific antigen binding molecules (e.g., antibodies) that bind blood clotting factors, factor IXa (FIXa) and factor X (FX), and enhance the FIXa-catalysed activation of FX to FXa. Use of the bispecific antigen binding molecules to control bleeding, by replacing natural cofactor FVIIIa which is deficient in patients with haemophilia A.
Description
CROSS-REFERENCE TO RELATED APPLICATIONS

This application is the U.S. national phase application filed under 35 U.S.C. § 371 claiming benefit to International Patent Application No. PCT/EP2018/066836, filed on Jun. 22, 2018, which is entitled to priority under to Great Britain Patent Application No. 1709970.6, filed Jun. 22, 2017, the disclosure of which is incorporated herein by reference in their entireties.


FIELD OF THE INVENTION

This invention relates to bispecific antigen-binding molecules (e.g., antibodies) that bind factor IXa and factor X clotting factors in the blood coagulation cascade. Such bispecifics functionally substitute for factor VIII by activating factor X, restoring blood clotting ability to patients who are deficient in FVIII, i.e., patients who have type A haemophilia.


BACKGROUND

Haemophilia is an inherited condition in which the blood has a reduced ability to clot, owing to loss of function (partial or total) of one of the many clotting factors. Haemophilia A is a deficiency in blood clotting factor VIII (FVIII). The disease has mild, moderate and severe forms, depending on the degree to which the patient retains any residual FVIII function and on the balance of other components in the blood coagulation cascade. If untreated, haemophilia A leads to uncontrolled bleeding, which can result in severe disability, especially through damage to joints from haemarthrosis events. The disease is often life-limiting and can be life-threatening. The global incidence of haemophilia A is believed to be around 1:10,000. Haemophilia B (deficiency of a different blood clotting factor, factor IX) is less common, with an incidence of around 1:50,000. Both diseases are X-linked so are usually found in males, the incidence of haemophilia A in male births thus being around 1 in 5,000.


Preventing bleeding episodes is essential to improving patients' quality of life and reducing the risk of fatal blood loss. For haemophilia A, the missing co-factor can be replaced by administration of FVIII. FVIII for administration to a patient may be recombinantly expressed or it may be purified from blood plasma. Typically, patients on this treatment self-inject with FVIII every 48 hours or 3× per week.


Treatment with FVIII is not a perfect solution. A serious drawback is that it can trigger production of alloantibodies in the body. This renders treatment with FVIII ineffective, as the alloantibodies bind the FVIII and prevent its activity, putting the patient in a dangerous situation if a bleed occurs. Such inhibitory antibodies develop in about 30% of patients treated with FVIII for severe haemophilia.


Treatment with plasma-derived FVIII, rather than the recombinant form, has been reported to have a lower risk of triggering inhibitory antibodies in patients. This may be due to the plasma-derived form retaining Von Willebrand factor (VWF), which is found naturally in association with FVIII and may mask immunogenic epitopes. However, no form of FVIII has yet been produced that completely avoids the risk of inhibitory antibodies.


Despite being possibly more immunogenic, recombinant FVIII offers some advantages over the plasma-derived form, since being more stable it is easier and cheaper to store and transport. The risk of transmitting infections via products from donated blood plasma is now much reduced compared with the 1980s when viruses such as hepatitis C and HIV were inadvertently spread to recipients of infected blood products, but of course the need for strict safety controls remains.


New recombinant forms of FVIII have been developed, such as the B-domain truncated polypeptide turoctocog alfa (NovoEight®). However, such products are ineffective for patients that develop neutralising antibodies against FVIII. Some patients successfully undergo immune tolerance induction to prevent anti-FVIII antibodies from developing. However, there remains a substantial demand for alternatives to FVIII for use in patients who have, or are at risk of developing, inhibitory antibodies.


One such alternative is recombinant factor Vila, known as activated eptacog alfa (NovoSeven®). However, it has a short half-life and must be injected every few hours. Its use is largely restricted to rescue therapy or providing haemostatic cover during surgery in haemophiliacs who have inhibitory antibodies, rather than being a viable option for long term protective treatment.


Another available product is FEIBA (Factor Eight Inhibitor Bypassing Activity), an activated prothrombin complex concentrate (aPCC), which similarly can be used to control bleeding episodes and to prevent bleeding during surgical interventions in haemophiliac patients who have inhibitors to factor VIII.


A variety of other alternative therapies are currently being pursued, such as gene therapy, suppression of anti-thrombin using siRNA, and an antibody to TFPI (Tissue factor Pathway Inhibitor), concizumab.


One new approach is a humanised bispecific IgG antibody targeting both factor IXa (FIXa) and factor X (FX). The bispecific antibody binds FIXa with one arm and FX with the other arm, bringing these two co-factors together and thereby promoting FIXa-catalysed activation of FX in the same way that FVIII does. Thus, the antibody functionally replaces FVIII in the blood coagulation cascade (FIG. 1). As its structure is completely different from FVIII, the antibody cannot be neutralised by anti-FVIII antibodies and so is suitable for patients who have developed, or are at risk of developing, alloantibodies to administered FVIII.


In 2012, Kitazawa et al reported isolation of a FIXa/X bispecific antibody which was able to activate FX, from a screen of approximately 40,000 anti-FIXa/X bispecific antibodies that had been produced by immunising 92 laboratory animals with human FIXa or FX and co-transfecting the anti-FIXa and anti-FX antibody genes into host cells for expression [1]. The selected antibody was refined to generate a humanised antibody designated hBS23, which showed coagulation activity in FVIII-deficient plasma and in vivo haemostatic activity in primates [1]. A more potent version of this antibody, designated hBS910 [2], entered clinical trials under the investigational drug name ACE910, INN emicizumab. The development of ACE910 took place in one of the leading antibody groups globally. Nevertheless, it took more than 7 years to engineer a molecule with the appropriate in vivo efficacy and with biochemical and biophysical properties suitable for clinical scale manufacturing.


In a phase I study of 48 healthy male subjects receiving ACE910 subcutaneously at doses up to 1 mg/kg, 2 subjects tested positive for anti-ACE910 antibodies [3]. The antibody was reported to have a linear pharmacokinetic profile and a half-life of about 4-5 weeks [3]. Emicizumab was subsequently administered to 18 Japanese patients with severe haemophilia A, at weekly subcutaneous doses of up to 3 mg/kg, and was reported to reduce the episodic use of clotting factors to control bleeding in these patients [4]. In December 2016, emicizumab was reported to have met its primary endpoint in a phase III clinical trial for reducing bleeding in patients with haemophilia A (the “HAVEN 1” study). A statistically significant reduction in the number of bleeds was reported for patients treated with emicizumab prophylaxis compared with those receiving no prophylactic treatment. The study was also reported to have met all secondary endpoints, including a statistically significant reduction in the number of bleeds over time with emicizumab prophylaxis treatment in an intra-patient comparison in people who had received prior bypassing agent prophylaxis treatment. The efficacy data on emicizumab are therefore encouraging, although safety concerns were heightened by the death of a patient on the HAVEN 1 study. The approved drug carries a boxed warning regarding the risk of thrombotic microangiopathy and thromboembolism in patients receiving aPCC in combination with emicizumab. As noted above, aPCC is used to control bleeding in patients who have inhibitory antibodies to FVIII, a key patient group for treatment with the bispecific antibody.


It is important to note that management of haemophilia requires continuous treatment for a patient's lifetime, beginning at the point of diagnosis—which is usually in infancy—and calls for a therapy that will be tolerated without adverse effects and that will remain effective over several decades or even a century. Long term safety, including low immunogenicity, is therefore of greater significance for an anti-haemophilia antibody compared with antibodies that are intended to be administered over a shorter duration such as a period of weeks, months or even a few years.


Recently, WO2018/098363 described bispecific antibodies binding to FIX and FX, isolated from a human antibody yeast library (Adimab). WO2018/098363 disclosed that increasing the affinity of the anti-FIXa arm of a bispecific antibody results in an increase in FVIIIa activity (represented by decreased blood clotting time in an assay). A bispecific antibody “BS-027125” was generated by affinity maturation of an initially selected “parent” antibody, which increased the affinity of its FIXa-binding arm. BS-027125 was reported to achieve approximately 90% FVIIIa-like activity in a one-stage clotting assay. When compared with emicizumab, BS-027125 was reported to exhibit much higher affinity binding to factor FIX zymogen, FIXa and FX zymogen, and much lower binding (no detected binding) to FXa. The FIX-binding arm, “BIIB-9-1336” reportedly showed selective binding for FIXa (activated FIX) in preference to FIX zymogen (mature FIX prior to proteolytic activation), and was found to bind an epitope overlapping with the FIXa epitope bound by FVIIIa. The FX-binding arm, “BIIB-12-917”, reportedly showed selective binding to FX zymogen, lacked detectable binding to (activated) FXa, and bound an epitope of FX that lies within the activation peptide (which is present in FX zymogen but not FXa). Further mutations were then introduced into selected FIX-binding antibodies, including BIIB-9-1336, to generate libraries from which to select for antibodies with even further increased specificity and/or affinity for FIXa.


SUMMARY OF THE INVENTION

The present invention relates to improved bispecific antigen-binding molecules that bind blood clotting factors FIXa and FX. The bispecific antigen-binding molecules of the present invention enhance the FIXa-catalysed activation of FX to FXa, and can effectively replace the natural cofactor FVIIIa which is missing in patients with haemophilia A, to restore the ability of the patients' blood to clot. See FIG. 2b.


As reported here, the inventors succeeded in generating a number of bispecific antigen-binding molecules having suitable qualities for development as therapeutic products, including very high potency in enhancing FX activation. Described are bispecific antigen-binding molecules having novel binding sites for anti-FIXa and anti-FX, which can be used to effectively substitute for FVIIIa in the blood clotting cascade. In particular, an anti-FIXa binding site is described which is highly active in combination with an array of different anti-FX binding sites and can thus be incorporated into a variety of different FIXa-FX bispecifics, providing flexibility for selection of bispecific antibodies with further desired characteristics such as ease of manufacture.


In a first aspect, the present invention relates to bispecific antigen-binding molecules comprising (i) a FIXa binding polypeptide arm comprising a FIXa binding site, and (ii) a FX binding polypeptide arm comprising a FX binding site. The FIXa and/or the FX binding polypeptide arm may comprise an antibody Fv region comprising the FIXa or FX binding site respectively. An antibody Fv region is an antibody VH-VL domain pair. The VH domain comprises HCDR1, HCDR2 and HCDR3 in a VH domain framework, and the VL domain comprises LCDR1, LCDR2 and LCDR3 in a VL domain framework. The polypeptide arm may comprise an antibody heavy chain (optionally one comprising an IgG constant region) and/or an antibody light chain.


Antigen-binding molecules of the present invention may thus comprise

    • first and second antibody Fv regions, the first and second antibody Fv regions comprising binding sites for FIXa and for FX respectively, and
    • a half-life extending region for prolonging the half-life of the molecule in vivo.


The half-life extending region may be a heterodimerisation region, comprising a first polypeptide covalently linked (e.g., as a fusion protein) to the first antibody Fv region and a second polypeptide covalently linked (e.g., as a fusion protein) to the second antibody Fv region, wherein the two polypeptides pair covalently and/or non-covalently with one another. The first and second polypeptides of the heterodimerisation region may have identical or different amino acid sequences. The heterodimerisation region may comprise one or more antibody constant domains, e.g., it may be an antibody Fc region.


Bispecific antigen-binding molecules of the present invention are able to bind FIXa through the FIXa binding site of the FIXa binding polypeptide arm and to bind FX through the FX binding site of the FX binding polypeptide arm, and thereby enhance the FIXa-catalysed activation of FX to FXa. This may be determined in an in vitro FX activation assay as described herein.


The FIXa binding site may be provided by a set of complementarity determining regions (CDRs) in the FIXa binding polypeptide arm, the set of CDRs comprising HCDR1, HCDR2, HCDR3 and/or LCDR1, LCDR2 and LCDR3, wherein

    • HCDR1 is SEQ ID NO: 1
    • HCDR2 is SEQ ID NO: 2
    • HCDR3 is SEQ ID NO: 400
    • LCDR1 is SEQ ID NO: 6
    • LCDR2 is SEQ ID NO: 7 and
    • LCDR3 is SEQ ID NO: 8.


      The set of CDRs in the FIXa binding polypeptide arm may be a set of CDRs wherein
    • HCDR1 is SEQ ID NO: 1
    • HCDR2 is SEQ ID NO: 2
    • HCDR3 is SEQ ID NO: 401, SEQ ID NO: 402, or SEQ ID NO: 403.
    • LCDR1 is SEQ ID NO: 6
    • LCDR2 is SEQ ID NO: 7 and
    • LCDR3 is SEQ ID NO: 8.


      The set of CDRs in the FIXa binding polypeptide arm may be a set of CDRs wherein
    • HCDR1 is SEQ ID NO: 1
    • HCDR2 is SEQ ID NO: 2
    • HCDR3 is SEQ ID NO: 171
    • LCDR1 is SEQ ID NO: 6
    • LCDR2 is SEQ ID NO: 7 and
    • LCDR3 is SEQ ID NO: 8.


The FIXa binding site may be provided by a set of complementarity determining regions (CDRs) in the FIXa binding polypeptide arm, the set of CDRs comprising HCDR1, HCDR2, HCDR3 and/or LCDR1, LCDR2 and LCDR3, wherein

    • HCDR1 is SEQ ID NO: 140
    • HCDR2 is SEQ ID NO: 141
    • HCDR3 is SEQ ID NO: 142
    • LCDR1 is SEQ ID NO: 6
    • LCDR2 is SEQ ID NO: 7 and
    • LCDR3 is SEQ ID NO: 8.


Optionally, one or more amino acids in the set of CDRs may be mutated to differ from these sequences. For example, the set of CDRs may comprise 1, 2, 3, 4 or 5 amino acid alterations, the altered residue or residues being in any one or more of the heavy or light chain CDRs. For example the set of CDRs may comprise one or two conservative substitutions. The choice of mutations, e.g., substitutions, can be informed by the analysis in Example 14 herein (e.g., for HCDR3) and/or by experimental testing to confirm biological properties of the resulting variants. Mutation in a VH domain, in a set of HCDRs, or in HCDR3, may comprise or consist of substitution of a hydrophobic or positively charged residue for the Ile at IMGT position 111.1 in HCDR3 SEQ ID NO: 171 or the Ser in IMGT position 111.1 of HCDR3 SEQ ID NO: 171. Preferably, the HCDR3 comprises a hydrophobic residue at this position, for example Ile, Leu, Val or Trp. Ile is especially preferred.


The FIXa binding polypeptide arm may comprise an antibody VH domain comprising a set of HCDRs HCDR1, HCDR2 and HCDR3. The sequence of HCDR1 may be SEQ ID NO: 140, optionally with one or two amino acid alterations (e.g., substitutions). The sequence of HCDR2 may be SEQ ID NO: 141, optionally with one or two amino acid alterations (e.g., substitutions). The sequence of HCDR3 may be SEQ ID NO: 142, optionally with one or two amino acid alterations (e.g., substitutions). The sequence of HCDR1 may be SEQ ID NO: 1, optionally with one or two amino acid alterations (e.g., substitutions). The sequence of HCDR2 may be SEQ ID NO: 2, optionally with one or two amino acid alterations (e.g., substitutions). The sequence of HCDR3 may be SEQ ID NO: 3, optionally with one or two amino acid alterations (e.g., substitutions). Optionally, the HCDR1 sequence is SEQ ID NO: 140 or SEQ ID NO: 1. Optionally, the HCDR2 sequence is SEQ ID NO: 141 or SEQ ID NO: 2. Optionally, the HCDR3 sequence is SEQ ID NO: 400, SEQ ID NO: 401, SEQ ID NO: 402, SEQ ID NO: 403 or SEQ ID NO: 171. The sequence of HCDR1 may be SEQ ID NO: 1. The sequence of HCDR2 may be SEQ ID NO: 2. The sequence of HCDR3 may be SEQ ID NO: 171. In a preferred embodiment, the VH domain comprises HCDR1 SEQ ID NO: 1, HCDR2 SEQ ID NO: 2 and HCDR3 SEQ ID NO: 171.


The FIXa binding polypeptide arm may comprise an antibody VL domain comprising a set of LCDRs LCDR1, LCDR2 and LCDR3. The sequence of LCDR1 may be SEQ ID NO: 6, optionally with one or two amino acid alterations (e.g., substitutions). The sequence of LCDR2 may be SEQ ID NO: 7, optionally with one or two amino acid alterations (e.g., substitutions). The sequence of LCDR3 may be SEQ ID NO: 8, optionally with one or two amino acid alterations (e.g., substitutions).


The antibody Fv region of the FIXa binding polypeptide arm may comprise a VH domain generated through recombination of immunoglobulin heavy chain v, d and j gene segments, wherein the v gene segment is VH3-7 (e.g., VH3-7*01), the d gene segment is DH1-26 (e.g., DH1-26*01) and/or wherein the j gene segment is JH6 (e.g. JH6*02), and/or it may comprise a VL domain generated through recombination of immunoglobulin light chain v and j gene segments, wherein the v gene segment is VL3-21 (e.g., VL3-21*d01) and the j gene segment is JL3 (e.g., JL3*02). The antibody Fv region of the FIXa binding polypeptide arm may comprise a VL domain generated through recombination of immunoglobulin light chain v and j gene segments, wherein the v gene segment is VL3-21 (e.g., VL3-21d*01) and the j gene segment is JL2 (e.g., JL2*01).


A VH domain of the FIXa binding polypeptide arm in the present invention may comprise HCDR1, HCDR2 and HCDR3 in a framework, the framework comprising a set of framework regions FR1, FR2, FR3 and FR4, wherein:

    • FR1 is SEQ ID NO: 132
    • FR2 is SEQ ID NO: 133
    • FR3 is SEQ ID NO: 134 and
    • FR4 is SEQ ID NO: 135,
    • or comprising that set of framework regions with up to 10 amino acid alterations, e.g., there may be one or two conservative substitutions in any one or more framework region.


FR1 may have amino acid sequence SEQ ID NO: 132, optionally with a substitution of L for F, a substitution of V for A and/or a substitution of A for V in the AVS motif.


FR2 may have amino acid sequence SEQ ID NO: 133.


FR3 may have amino acid sequence SEQ ID NO: 134, optionally with a substitution of Y for F at the first residue position of that sequence, a substitution of D for A at the fourth residue position, a substitution of I for M at the twelfth residue position, a substitution of N for K at the nineteenth residue position, a substitution of L for V at the twenty first residue position and/or a substitution of L for V at the twenty third residue position of that sequence.


FR4 may have amino acid sequence SEQ ID NO: 135, optionally with a substitution of S for T at the eighth residue position.


The amino acid sequence of the VH domain of a FIXa polypeptide binding arm may share at least 90% sequence identity with SEQ ID NO: 324. Sequence identity may be at least 95%, at least 97%, at least 98% or at least 99%. It is preferred that the VH domain has a hydrophobic or positively charged residue at IMGT position 111.1 in HCDR3. The HCDR3 may be 15 amino acids in length. Preferably, the HCDR3 comprises a hydrophobic residue at this position, for example Ile, Leu, Val or Trp. Ile is especially preferred. The HCDR3 may comprise Ser at IMGT position 110, position 111 and/or position 112.1, for example it may comprise Ser two or three of positions 110, 111 and 112.1. The amino acid sequence of an HCDR3 of a VH domain in the present invention is optionally SEQ ID NO: 400, SEQ ID NO: 401, SEQ ID NO: 402, SEQ ID NO: 403 or SEQ ID NO: 171.


The amino acid sequence of the VH domain may share at least 90% sequence identity with SEQ ID NO: 5. Sequence identity may be at least 95%, at least 97%, at least 98% or at least 99%.


Optionally the VH domain amino acid sequence is SEQ ID NO: 324. Optionally the VH domain amino acid sequence is SEQ ID NO: 5.


A VL domain of the FIXa binding polypeptide arm in the present invention may comprise LCDR1, LCDR2 and LCDR3 in a framework, the framework comprising a set of framework regions FR1, FR2, FR3 and FR4, wherein:

    • FR1 is SEQ ID NO: 136
    • FR2 is SEQ ID NO: 137
    • FR3 is SEQ ID NO: 138 and
    • FR4 is SEQ ID NO: 139,
    • or comprising that set of framework regions with up to 10 amino acid alterations, e.g., there may be one or two conservative substitutions in any one or more framework region.


The amino acid sequence of the VL domain may share at least 90% sequence identity with SEQ ID NO: 10. Sequence identity may be at least 95%, at least 97%, at least 98% or at least 99%. Optionally the VL domain amino acid sequence is SEQ ID NO: 10.


The FIXa binding polypeptide arm of bispecific antigen-binding molecules of the present invention may, even when provided in monospecific form outside the context of the bispecific molecule, be capable of enhancing the FIXa-catalysed activation of FX to FXa. This may be determined in an in vitro FX activation assay as described herein.


Surface plasmon resonance may be used to determine binding to FIXa and FX, and to quantify the affinity of a polypeptide arm for antigen binding.


The FX binding site may be provided by a set of CDRs in the FX binding polypeptide arm. The FX binding polypeptide arm may comprise an antibody VH-VL domain pair (i.e., an antibody Fv region), the VH domain comprising HCDR1, HCDR2 and HCDR3 in a framework, and the VL domain comprising LCDR1, LCDR2 and LCDR3 in a framework.


The FX binding site may be provided by the CDRs of antibody T02, i.e., a set of CDRs comprising HCDR1, HCDR2, HCDR3 and/or LCDR1, LCDR2 and LCDR3, wherein

    • HCDR1 is SEQ ID NO: 57
    • HCDR2 is SEQ ID NO: 58
    • HCDR3 is SEQ ID NO: 59
    • LCDR1 is SEQ ID NO: 62
    • LCDR2 is SEQ ID NO: 63 and
    • LCDR3 is SEQ ID NO: 64.


The FX binding polypeptide arm may comprise a VH domain having at least 90% amino acid sequence identity with the T02 VH domain SEQ ID NO: 61 and/or may comprise a VL domain having at least 90% amino acid sequence identity with the T02 VL domain SEQ ID NO: 66. Sequence identity may be at least 95%, at least 97%, at least 98% or at least 99%. Optionally the VH domain amino acid sequence is SEQ ID NO: 61. Optionally the VL domain amino acid sequence is SEQ ID NO: 66.


The FX binding site may be provided by the CDRs of antibody T05, i.e., a set of CDRs comprising HCDR1, HCDR2, HCDR3 and/or LCDR1, LCDR2 and LCDR3, wherein

    • HCDR1 is SEQ ID NO: 67
    • HCDR2 is SEQ ID NO: 68
    • HCDR3 is SEQ ID NO: 69
    • LCDR1 is SEQ ID NO: 72
    • LCDR2 is SEQ ID NO: 73 and
    • LCDR3 is SEQ ID NO: 74.


The FX binding polypeptide arm may comprise a VH domain having at least 90% amino acid sequence identity with the T05 VH domain SEQ ID NO: 71 and/or may comprise a VL domain having at least 90% amino acid sequence identity with the T05 VL domain SEQ ID NO: 76. Sequence identity may be at least 95%, at least 97%, at least 98% or at least 99%. Optionally the VH domain amino acid sequence is SEQ ID NO: 61. Optionally the VL domain amino acid sequence is SEQ ID NO: 76.


The FX binding site may be provided by the CDRs of antibody T14, i.e., a set of CDRs comprising HCDR1, HCDR2, HCDR3 and/or LCDR1, LCDR2 and LCDR3, wherein

    • HCDR1 is SEQ ID NO: 96
    • HCDR2 is SEQ ID NO: 97
    • HCDR3 is SEQ ID NO: 98
    • LCDR1 is SEQ ID NO: 101
    • LCDR2 is SEQ ID NO: 92 and
    • LCDR3 is SEQ ID NO: 102.


The FX binding polypeptide arm may comprise a VH domain having at least 90% amino acid sequence identity with the T14 VH domain SEQ ID NO: 100 and/or may comprise a VL domain having at least 90% amino acid sequence identity with the T14 VL domain SEQ ID NO: 104. Sequence identity may be at least 95%, at least 97%, at least 98% or at least 99%. Optionally the VH domain amino acid sequence is SEQ ID NO: 100. Optionally the VL domain amino acid sequence is SEQ ID NO: 104.


The FX binding polypeptide arm may comprise an antibody Fv region comprising:

    • (a) a VH domain generated through recombination of immunoglobulin heavy chain v, d and j gene segments, wherein the v and j gene segments are VH1-3 (e.g., VH1-3*01) and JH6 (e.g., JH6*02), and
    • a VL domain generated through recombination of immunoglobulin light chain v and j gene segments, wherein the v and j gene segments are VL1-47, (e.g., VL1-47*01) and JL1 (e.g., JL1*01); or
    • (b) a VH domain generated through recombination of immunoglobulin heavy chain v, d and j gene segments, wherein the v and j gene segments are VH3-30 (e.g., VH3-30*18) and JH6 (e.g., JH6*02), and
    • a VL domain generated through recombination of immunoglobulin light chain v and j gene segments, wherein the v and j gene segments are VL2-8, (e.g., VL2-8*01) and JL2 (e.g., JL2*01); or
    • (c) a VH domain generated through recombination of immunoglobulin heavy chain v, d and j gene segments, wherein the v and j gene segments are VH4-61 (e.g., VH4-61*01) and JH1 (e.g., JH1*01), and
    • a VL domain generated through recombination of immunoglobulin light chain v and j gene segments, wherein the v and j gene segments are VK3-11, (e.g., VK3-11*01) and JK5 (e.g., JK5*01).


The FX binding polypeptide arm may comprise an antibody Fv region comprising a VH domain generated through recombination of immunoglobulin heavy chain v, d and j gene segments, wherein the v and j gene segments are:

    • VH1-3 (e.g., VH1-3*01) and JH6 (e.g., JH6*02),
    • VH3-30 (e.g., VH3-30*18) and JH6 (e.g., JH6*02),
    • VH3-33 (e.g., VH3-33*01) and JH6 (e.g., JH6*02),
    • VH4-31 (e.g., VH4-31*03) and JH4 (e.g., JH4*02), or
    • VH4-59 (e.g., VH4-59*01) and JH4 (e.g., JH4*02), and
    • a VL domain.


The FX binding polypeptide arm may comprise an antibody Fv region comprising a VL domain generated through recombination of immunoglobulin light chain v and j gene segments, wherein the v gene segment is VL3-21 (e.g., VL3-21*d01) and the j gene segment is JL3 (e.g., JL3*02).


The FIXa binding polypeptide arm and the FX binding polypeptide arm may each comprise an antibody Fv, wherein the VL domain of each Fv has an identical amino acid sequence, i.e. the bispecific antigen-binding molecule has a common VL domain. The molecule may have a common light chain comprising a variable region and a constant region, optionally a human lambda constant region.


The bispecific antigen-binding molecule may be a tetrameric immunoglobulin comprising

    • a first pair of antibody heavy and light chains (heavy-light chain pair) comprising a FIXa binding Fv region,
    • a second heavy-light chain pair comprising a FX binding Fv region,
    • wherein each heavy chain comprises a VH domain and a constant region, and each light chain comprises a VL domain and a constant region, and wherein the first and second heavy-light chain pairs associate through heterodimerisation of their heavy chain constant regions to form the immunoglobulin tetramer. As noted, the light chain may be a common light chain, i.e., the light chain of the first and second heavy-light chain pairs has an identical amino acid sequence. Exemplary immunoglobulin isotypes include human IgG, e.g., IgG4, optionally with engineered constant domains such as IgG4 PE.


An advantageous feature of bispecific antibodies exemplified here is that they have been generated from human immunoglobulin gene segments, using the Kymouse platform. Unlike antibodies generated from immunisation of normal laboratory animals, which may require “humanisation” steps such as grafting of mouse CDRs into human antibody variable domains and iterative refinement of the engineered variable domains to mitigate a loss of function resulting from these changes, the antibodies of the present invention were generated and selected from the outset with fully human antibody variable domains. The use of a fully human antibody is of special relevance in the context of haemophilia treatment, where low immunogenicity is paramount, as noted above. The low immunogenicity of the bispecific antibodies of the present invention renders them suitable for treatment of haemophilia A patients, including those with or without inhibitory antibodies to other treatments such as FVIII. Patients receiving antigen-binding molecules of the present invention should be at minimal risk of developing an immunogenic response to the therapy.


The mode of action of the bispecific molecules is also associated with a good safety profile, with low risk of complications such as deep vein thrombosis and pulmonary embolism. Activity of the bispecific molecules is comparable with that of natural FVIII and a mechanism of action that is integrated within the existing blood coagulation pathway, being activated only in the context of upstream triggering of the natural clotting cascade.


Other desirable features include long-half life (reducing the required frequency of administration) and amenability of the molecules to formulation at high concentration (facilitating subcutaneous injection in the home setting).


Further aspects of the invention relate to pharmaceutical compositions comprising the bispecific antigen-binding molecules, their encoding nucleic acids, the individual polypeptide binding arms, systems and methods for production of the molecules, and their use in medicine including for the treatment of haemophilia A, as set out in the appended claims and described in the present disclosure.





BRIEF DESCRIPTION OF THE DRAWINGS


FIG. 1 Blood coagulation cascade [5].



FIG. 2 A. Co-factor action of FVIIIa interacting with FIXa and FX.


B. Co-factor action of bispecific antibody interacting with FIXa and FX.



FIG. 3 Amino acid sequence of factor IX, with residue numbering for the mature protein. The signal peptide (straight underlined) is cleaved after secretion. The propeptide (wave underlined) is cleaved on maturation. Mature factor IX contains a light chain (residues 1-145) and a heavy chain (residues 146-415). The activation peptide (boxed) is cleaved on activation, generating activated factor IXa which contains a light chain (residues 1-145) and a heavy chain (residues 181-415, bold) joined by a disulphide bridge between Cys132 and Cys289.



FIG. 4 Amino acid sequence of factor X, with residue numbering. Residues 1-31 are a signal peptide (straight underlined). Residues 32-40 are a propeptide (wave underlined). The FX light chain is residues 41-179. The FX heavy chain is residues 183-488. The FXa heavy chain is residues 235-488 (bold).



FIG. 5 A, B Bispecific IgGs. (A) Bispecific IgG with common light chain. (B) Bispecific IgG with different light chains.



FIG. 5 C, D Bispecific IgG fragments. (C) F(ab′)2. (D) Tandem-linked scFv.



FIG. 6 FIT-Ig format of bispecific antibodies. (i) Assembled FIT-Ig antibody (ii) Polypeptide chains included in FIT-Ig antibody. Construct #1 is a polypeptide containing, in the N to C direction, the light variable (VL) and light constant (CL) regions of antibody “A”, fused to the heavy variable (VH) and heavy constant regions (CH1, CH2, CH3) of antibody “B”. Preferably, no linker is included between the CL and VHB domain. Construct #2 is a polypeptide fusion of the heavy variable (VH) region and CH1 of antibody “A”. Construct #3 is a polypeptide fusion of the light variable (VL) and light constant (CL) regions of antibody “B”. The FIT-Ig may be constructed with antibody “A” being anti-FIXa and antibody “B” being anti-FX, or with antibody “A” being FX and antibody “B” being anti-FIXa.



FIG. 7 A. Principles of in vitro assay for FVIII mimetic activity of a bispecific molecule as described in Example 9.


B. Example data from the assay as described in Example 9 showing positive result for FIXa-FX bispecific molecule compared with negative control.



FIG. 8 Results of initial high-throughput functional screening of bispecific antibodies in tenase assay.



FIG. 9 Table of N128H mutants.



FIG. 10 FXase activity of N128H CDR3 mutants.



FIG. 11 FVIII mimetic activities of N128H CDR3 mutants in aPTT assay.



FIG. 12 In vitro tenase (FXase) assay activity of Factor IX specific arm N436.



FIG. 13 aPTT assay activity of bispecific antibodies comprising N436H VH domain and N128L VL domain (providing FIX binding site) heterodimerised with FX binding site provided by (i) T02H VH domain and T02L VL domain, (ii) T05H VH domain and T05L VL domain or (iii) T14H VH domain and T14L VL domain.





DETAILED DESCRIPTION

Blood Coagulation


The blood coagulation cascade is diagrammed in FIG. 1. Coagulation or clotting is one of the most important biological processes which stops blood loss from a damaged vessel to allow the vessel to be repaired. The mechanism of coagulation involves activation, adhesion, and aggregation of platelets along with deposition and maturation of fibrin. Misregulation of coagulation can result in excessive bleeding (haemophilia) or obstructive clotting (thrombosis). Coagulation is highly conserved in all mammals. It is controlled by a complex network of coagulation factors. Coagulation is initiated when the endothelium lining the blood vessel is damaged. The exposure of subendothelial tissue factor (TF) to plasma factor VII (FVII) leads to primary haemostasis (extrinsic pathway): a loose plug is formed at the site of injury. Activation of additional coagulation factors, especially factor IX (FIX) and factor VIII (FVIII), leads to secondary haemostasis (intrinsic pathway): fibrin strands are formed to strengthen the plug. Extrinsic and intrinsic pathways ultimately converge to a common point: the formation of the factor Xa/Va complex which together with calcium and bound on a phospholipid surface generate thrombin (factor IIa) from prothrombin (factor II).


FVIII is cleaved by thrombin or factor Xa (FXa), and the resultant factor VIIIa (FVIIIa) presents a heterotrimeric structure consisting of the A1 subunit, the A2 subunit, and the light chain. Upon activation and in the presence of calcium ions and a phospholipid surface (on platelets), FVIIIa binds via its light chain and A2 subunit to FIXa and simultaneously binds via its A1 subunit to FX, forming an active intrinsic “tenase” or “Xase” complex in which the FVIIIa cofactor brings FIXa and FX into proximity and also allosterically enhances the catalytic rate constant of FIXa. See FIG. 2a. Factor X is activated by the serine protease activity of FIXa, and the clotting cascade continues, culminating in the deposition of fibrin, the structural polymer of the blood clot.


Haemophilia arise through a deficiency in the Xase complex, due either to a lack of FVIII cofactor activity (haemophilia A) or a lack of FIX enzyme activity (haemophilia B).


Factor IX (FIX)


Factor IX is a serine protease which requires factor VIII as a cofactor. It circulates in blood as an inactive precursor, which is activated through intrinsic or extrinsic pathway at the time of haemostatic challenge, as discussed above.


Unless the context requires otherwise, factor IX referred to herein is human factor IX, and factor IXa is human factor IXa.


The amino acid sequence of human factor IX are shown in FIG. 3. The factor IX gene is approximately 34 kb in length and contains 8 exons. The transcript comprises a short 5′ untranslated region, an open reading frame plus stop codon and a 3′ untranslated region. The ORF encodes a 461 amino acid pre-pro-protein in which the pre-sequence (signal peptide) directs factor IX for secretion, the propeptide sequence provides a binding domain for a vitamin K dependent carboxylase, which carboxylates certain glutamic acid residues in the adjacent GLA domain, and the remainder represents the factor IX zymogen, which enters into circulation after removal of the pre- and pro-sequences. The mature 415 residue FIX protein contains, from N to C terminus: a GLA domain in which 12 glutamic acid residues are post-translationally γ-carboxylated, two epidermal growth factor (EGF)-like domains, an activation peptide sequence and a catalytic serine protease domain. FIX is activated by either activated factor XI generated through the intrinsic pathway, or by the TF/FVIIa complex of the extrinsic pathway. Either way, activation involves cleavage of the peptide bond following R145 (α-cleavage) and of the peptide bond following R180 (β-cleavage), releasing an activation peptide corresponding to the intervening sequence, and thereby generating the activated FIXa molecule, which has an N terminal light chain (GLA-EGF-EGF) and a C terminal heavy chain (catalytic domain) joined by a disulphide bridge between C132 of the light chain and C289 of the heavy chain. Residue numbering refers to amino acids in the mature FIX polypeptide sequence. On the phospholipid surface where the Xase complex forms, it is the GLA domain of FIXa which associates with the phospholipid, while the catalytic domain stands high (>70 Å) above the phospholipid surface and is modulated by the A2 domain of FVIIIa [6, 7].


The molecular basis of haemophilia B—deficiency in FIXa activity—is diverse, including a variety of point mutations, nonsense mutations, mRNA splice site mutations, deletions, insertions, or mis-sense mutations at activation cleavage sites [8].


The catalytic (protease) domain of activated FIX (FIXa) is involved in binding to FVIIIa. Residue E245 in this domain binds calcium ions, and mutations at this position may reduce binding to FVIII and lead to haemophilia B, for example the substitution E245V. Mutations within the FIX helix formed by residues 330-338 are also linked with reduced binding to FVIII and consequently to haemophilia B.


Non-pathogenic mutations in factor IX have also been reported, including single nucleotide polymorphisms (SNPs) and length polymorphisms—reviewed in [8]. These include the MnII SNP in exon 6, resulting in T/A substitution at residue 148 (Malmö polymorphism), which is relatively common among white and black American populations [8].


Factor X (FX)


Unless the context requires otherwise, factor X referred to herein is human factor X, and factor Xa is human factor Xa. The amino acid sequence of human FX is shown in FIG. 4.


FX is also known as Stuart-Prower factor. It is a serine endopeptidase. FX can be activated, by hydrolysis, into factor Xa by either factor IX (together with its cofactor, factor FVIII, as described above) or factor VII (with its cofactor, tissue factor). FX acts by cleaving prothrombin in two places—at an Arg-Thr bond and then at an Arg-Ile bond, to yield the active thrombin.


Antigen-Binding


A desirable feature of the bispecific antigen-binding molecule is that it binds FIXa and FX in a manner that allows the bound FIXa to activate the bound FX.


To bring FIXa and FX together and thereby promote the activation of FX by FIXa, the bispecific antigen-binding molecule may bind these two cofactors simultaneously. Binding may occur sequentially, e.g., an initial binary complex may form between a first binding arm and its cognate antigen, followed by binding of the second binding arm to its cognate antigen. In principle these two binding events may occur in either sequence, i.e., FIXa followed by FX, or FX followed by FIXa. The molecular choreography is influenced by the relative affinities of the two binding sites for their respective antigens. In a population of bispecific antigen-binding molecules, FIXa and FX, a number of different complexes are expected to exist in parallel. Thus the pool will comprise free antigen-binding molecule, free FIXa, free FX, FIXa complexed with antigen-binding molecule, FX complexed with antigen-binding molecule, and a tertiary complex of FIX, FX and antigen-binding molecule, with each of these species being present in different proportions according to the relative on-rates and off-rates of the individual interactions.


It may be preferable for a bispecific antigen-binding molecule to have a higher affinity for FIXa than for FX. Such a bispecific molecule would be envisaged to form an initial complex with FIXa, which in turn would bind and activate FX. The relatively low affinity for FX reduces the proportion of FX that is bound in incomplete antibody-antigen complexes (i.e., without FIXa). A potential advantage of this is that it allows a greater proportion of FX to remain free to engage with any FVIII that may be present in a patient's blood. Haemophilia A encompasses a range of deficiencies in FVIII, ranging from mild deficiency to total absence of functional FVIII. For those patients who retain some functional FVIII, it may be desirable to retain this natural activity as far as possible. Thus, it may be desirable to provide a bispecific antigen-binding molecule in which the FX binding arm does not compete with FVIII for binding to FX.


Preferably the FX binding arm has a higher affinity for FX than for FXa. A low affinity for FXa promotes release of the activated product, completing the role of the FVIII-mimetic molecule in the coagulation cascade and freeing the FX binding site for re-use.


FIXa Binding


The FIXa binding arm of a bispecific antigen-binding molecule may bind the light chain and/or the heavy chain of FIXa. Initial studies indicated that FIXa binding arms of the N128 lineage described in the Examples do not bind the FIXa light chain in isolation (in the absence of the heavy chain).


A bispecific antigen-binding molecule of the present invention (or FIXa binding polypeptide arm thereof) may thus be one which binds a FIXa molecule comprising a heavy chain and a light chain, and which does not bind the FIX light chain in the absence of the heavy chain. Optionally, the FIXa binding arm recognises an epitope formed by, or stabilised by, the combination of the FIXa heavy and light chains. It may for example make contact only with the light chain in the FIXa molecule, binding an epitope that is exposed or stabilised only when the light chain is present in combination with the heavy chain in the FIXa molecule. Alternatively, it may contact an epitope comprising one or more residues from both the light chain and the heavy chain, or comprising residues of the heavy chain alone.


An antigen-binding molecule according to the present invention, or a FIXa-binding polypeptide arm thereof, may bind the EC domain of human FIXa with an affinity (measured as KD) of 10 mM or less, preferably 5 mM or less, more preferably 1 mM or less. For example, KD may be between 1 nM and 3 μM.


The KD for binding human FIXa may be between 0.1 μM and 1 μM, e.g., between 0.15 and 0.3 μM. The KD may be 0.6 μM or less, 0.5 μM or less, 0.4 μM or less, 0.3 μM or less, 0.25 μM or less, or 2 μM or less. The KD may be at least 0.1 μM, for example at least 0.2 μM.


The KD may be 50 nM or less, 10 nM or less, 5 nM or less, 2 nM or less, or 1 nM or less. The KD may be 0.9 nM or less, 0.8 nM or less, 0.7 nM or less, 0.6 nM or less, 0.5 nM or less, 0.4 nM or less, 0.3 nM or less, 0.2 nM or less, or 0.1 nM or less. The KD may be at least 0.001 nM, for example at least 0.01 nM or at least 0.1 nM. The KD may be between 0.1-10 nM.


An antigen-binding molecule according to the present invention, or a FIXa-binding polypeptide arm thereof, may bind human FIX with an affinity (measured as KD) between 0.1 μM and 1 μM, e.g., between 0.15 and 0.3 μM. The KD may be 0.6 μM or less, 0.5 μM or less, 0.4 μM or less, 0.3 μM or less, 0.25 μM or less, or 2 μM or less. The KD may be at least 0.1 μM, for example at least 0.2 μM.


The KD of interaction with FIX may be comparable to the KD of interaction with FIXa, e.g., there may be difference of less than 25%, optionally less than 10%, in the FIXa-binding arm's affinity for FIX compared with the affinity for FIXa. There may be no statistically significant difference in KD of interaction with FIX compared with FIXa.


As described elsewhere herein, affinity may be determined using surface plasmon resonance (SPR), e.g., with the binding arm coupled to a solid surface, optionally as a dimer (e.g., as monospecific IgG), with the antigen in solution as analyte, at 25° C.


FX Binding


An antigen-binding molecule according to the present invention, or a FX-binding polypeptide arm thereof, may bind the EC domain of human FX with a KD of 10 mM or less, preferably 5 mM or less, more preferably 1 mM or less. For example, KD may be between 5 μM and 1 nM, e.g., between 5 μM and 10 nM.


The KD may be between 0.1 μM and 1 μM, e.g., between 0.15 and 0.3 μM. The KD may be 0.6 μM or less, 0.5 μM or less, 0.4 μM or less, 0.3 μM or less, or 0.25 μM or less. The KD may be at least 0.1 μM.


The KD may be 50 nM or less, 10 nM or less, 5 nM or less, 2 nM or less, or 1 nM or less. The KD may be 0.9 nM or less, 0.8 nM or less, 0.7 nM or less, 0.6 nM or less, 0.5 nM or less, 0.4 nM or less, 0.3 nM or less, 0.2 nM or less, or 0.1 nM or less. The KD may be at least 0.001 nM, for example at least 0.01 nM or at least 0.1 nM. For example, the KD may be between 1-100 nM. KD may be between 1-10 nM.


As described elsewhere herein, affinity may be determined using surface plasmon resonance (SPR), e.g., with the binding arm coupled to a solid surface, optionally as a dimer (e.g., as monospecific IgG), with the antigen in solution as analyte, at 25° C.


Measurement of Antigen-Binding Affinity


The affinity of an antigen-binding molecule for binding FIX, FIXa, FX and FXa may be quantified in terms of the equilibrium dissociation constant KD, the ratio Ka/Kd of the association or on-rate (Ka) and the dissociation or off-rate (kd) of the binding interaction. KD, Ka and Kd for antigen binding can be measured using surface plasmon resonance (SPR).


Quantification of affinity may be performed using SPR with the antigen-binding polypeptide arm in monovalent form, e.g., antibody Fab or Fv comprising the antigen binding site, or heterodimeric immunoglobulin (e.g., IgG) having a single antigen-binding arm for the antigen in question. Alternatively, as shown in Example 3 and Example 6, or in Examples 15-16, it may be convenient to determine affinity for the antigen-binding polypeptide arm in bivalent form, for example IgG comprising homodimeric antigen-binding arms. SPR may comprise coating dimers of the antigen-binding polypeptide arm on to a biosensor chip (directly or indirectly), exposing the antigen-binding polypeptide arms to antigen in buffered solution at a range of concentrations, detecting binding, and calculating the equilibrium dissociation constant KD for the binding interaction. SPR may be performed at 25° C. A suitable buffered solution is 150 mM NaCl, 0.05% detergent (e.g., P20) and 3 mM EDTA, pH 7.6. HBS-P 1× (10 mM HEPES pH 7.4, 150 mM NaCl, 3 mM EDTA, 0.05% polysorbate 20 pH 7.6) with 2.5 mM CaCl2). is an example buffer. The binding data can be fitted to a 1:1 model using standard algorithms, which may be inherent to the instrument used. A variety of SPR instruments are known, such as Biacore™, ProteOn XPR36™ (Bio-Rad®), and KinExA® (Sapidyne Instruments, Inc).


Cross-Reactivity


Regulatory bodies may require candidate therapeutic molecules to have demonstrated therapeutic efficacy in laboratory animals before they advance to human clinical trials. An example of an acquired haemophilia A animal model is a cynomolgus monkey that is rendered deficient in blood clotting through administration of a FVIII-neutralising antibody or a small molecule inhibitor against FVIII, thereby replicating the phenotype of a human haemophilia A patient. To enable testing of bispecific antigen-binding molecules in animal models, it is desirable for the binding site of each arm to be cross-reactive with the corresponding antigen from one or more non-human mammals. Thus, the FIXa binding site of the antigen-binding molecule may bind murine (e.g., mouse or rat), rabbit or non-human primate (e.g., cynomolgus monkey) FIXa as well as human FIXa, and the FX binding site may bind murine (e.g., mouse or rat), rabbit or non-human primate (e.g., cynomolgus monkey) FXa as well as human FXa.


One way to quantify the extent of species cross-reactivity of an antigen-binding molecule (or, more precisely, of its antigen binding site) is as the fold-difference in its affinity for antigen or one species compared with antigen of another species, e.g., fold difference in affinity for human antigen vs cynomolgus antigen. Affinity may be quantified as KD, referring to the equilibrium dissociation constant of the binding of the antigen to the antigen-binding molecule. KD may be determined by SPR as described elsewhere herein.


A species cross-reactive binding molecule may have a fold-difference in affinity for binding human and non-human antigen that is 30-fold or less, 25-fold or less, 20-fold or less, 15-fold or less, 10-fold or less or 5-fold or less. To put it another way, the KD of binding the extracellular domain of the human antigen may be within 30-fold, 25-fold, 20-fold, 15-fold, 10-fold or 5-fold of the KD of binding the extracellular domain of the non-human antigen.


Preferably, the binding affinities of human and non-human antigen are within a range of 10-fold or less, more preferably within 5-fold or within 2-fold. KD for binding non-human FIXa, e.g., as determined by surface plasmon resonance, may be up to 10-fold (preferably up to 5-fold or up to 2-fold) greater or up to 10-fold lower (preferably up to 5-fold or up to 2-fold lower) than the Kd for binding human FIXa. Similarly, KD for binding non-human FX, e.g., as determined by SPR, may be up to 10-fold (preferably up to 5-fold or up to 2-fold) greater or up to 10-fold (preferably up to 5-fold or up to 2-fold) lower than the Kd for binding human FX. Methods of determining affinity are described elsewhere herein.


Binding molecules can also be considered species cross-reactive if the KD for binding antigen of both species meets a threshold value, e.g., if the KD of binding human antigen and the KD of binding non-human antigen are both 10 mM or less, preferably 5 mM or less, more preferably 1 mM or less. The KD may be 10 nM or less, 5 nM or less, 2 nM or less, or 1 nM or less. The KD may be 0.9 nM or less, 0.8 nM or less, 0.7 nM or less, 0.6 nM or less, 0.5 nM or less, 0.4 nM or less, 0.3 nM or less, 0.2 nM or less, or 0.1 nM or less.


While species cross-reactivity for binding antigen of different species may be advantageous, selectivity of the FIXa binding arm and the FX binding arm for their respective antigens is nevertheless desirable to avoid unwanted side effects. Thus, within the body, FIX/FIXa and FX/FXa are preferably the only antigens bound by the antigen-binding molecule.


Enhancement of FIXa-Mediated Activation of FX


The ability of a bispecific antigen-binding molecule to enhance the FIXa-mediated activation of FX to FXa may be determined in assays in vitro or in vivo.


A suitable in vitro assay is the FX activation assay exemplified in Example 9 and illustrated in FIG. 7. The assay comprises

    • (i) contacting the bispecific antigen-binding molecule with FIXa and FX under conditions suitable for formation of FXa (e.g., in the presence of phospholipid, in buffered solution at 37° C.)
    • (ii) adding substrate that is cleavable by FXa to generate a detectable product, and
    • (iii) detecting, and optionally quantifying, the presence of the detectable product.


      The level of product may be compared with a control assay in which FIXa-FX bispecific antigen-binding molecule is absent from the reaction mixture. Significant difference in product level in the assay with the bispecific compared with control indicates that the bispecific is able to enhance FIXa-mediated activation of FX. FVIII may be included as a positive control.


The level of product may be compared with an assay in which the FIXa-FX bispecific antigen-binding molecule is emicizumab. A bispecific according to the present invention may enhance the FIXa-mediated activation of FX to FXa to the same or similar extent (e.g., within 10% difference or within 5% difference) as emicizumab, or to a greater extent (e.g., more than 10% more activation of FX to FXa than is achieved with emicizumab as measured by the level of detectable product).


Another suitable assay is to measure the activated partial thromboplastin time (aPTT) in FVIII-deficient plasma, which may be performed in the presence or the absence of inhibitors and can be used to compare the activity of bispecific molecules with recombinant human FVIII. This assay is exemplified in Example 10 and Example 18. aPTT is an end point assay which provides a global overview of blood clot formation and provides coagulation time as the assay read-out. FVIII-deficient plasma would typically have a coagulation time of around 80-90 seconds in the aPTT assay. Bispecific antigen binding molecules of the present invention are effective to reduce the coagulation time in an aPTT assay (compared with a negative control). The coagulation time of human FVIII-deficient in an aPTT assay with a bispecific antigen binding molecule according to the present invention may for example be the same as or less than that of the coagulation time with recombinant human FVIIIa. Physiological clotting time for normal (FVIII+) human plasma is typically <40 seconds, e.g., in the range of 37-34 s. Similar values are achievable with FVIII-deficient plasma upon provision of activated FVIIIa, which provides a convenient way of standardising the assay through calibration of the apparatus/measurement against reference values. Alternatively, coagulation time of normal (FVIII+) human plasma may be used for reference, the aPTT assay being begun by induction of coagulation through the addition of calcium.


A bispecific antigen-binding molecule of the present invention may give a coagulation time in the aPTT assay of within 10 seconds of that of FVIIIa (i.e., up to 10 seconds more than or up to 10 seconds less than the coagulation time of the aPTT assay with FVIIIa). Preferably, the coagulation time in the aPTT assay with a bispecific antigen binding molecule of the invention is less than that with FVIIIa. The bispecific antigen-binding molecule may reduce the coagulation time to less than 40 seconds, less than 35 seconds, or less than 30 seconds. The coagulation time may be between 20 and 40 seconds, e.g., between 20 and 30 seconds.


Bispecific Antigen-Binding Molecules


The bispecific antigen-binding molecule comprises a FIXa binding polypeptide arm and a FX binding polypeptide arm. It may be a multi-chain or single-chain polypeptide molecule. While the FIXa binding polypeptide arm and the FX binding polypeptide arm represent different moieties of the bispecific molecule, one polypeptide can optionally form all or part of both the FIXa binding arm and the FX binding arm.


A polypeptide binding arm is the region of the bispecific molecule that comprises the binding site for one of the antigens (FIXa or FX). One or both antigen-binding sites of a bispecific molecule can be provided by a set of complementarity determining regions (or peptide loops) in a polypeptide arm, wherein the polypeptide arm is any suitable scaffold polypeptide whether that of an antibody (e.g., an antibody Fv region) or a non-antibody molecule. A binding arm may comprise one or more than one (e.g., two) polypeptides or parts (e.g., domains) thereof.


The invention is described in detail herein with reference to bispecific antibodies, wherein at least one of the antigen binding polypeptide arms is provided by a set of CDRs in an antibody VH and/or VL domain, optionally an Fv region.


Antibodies are immunoglobulins or molecules comprising immunoglobulin domains. Antibodies may be IgG, IgM, IgA, IgD or IgE molecules or molecules including antigen-specific antibody fragments thereof. The term “antibody” covers any polypeptide or protein comprising an antibody antigen-binding site. An antibody antigen-binding site (paratope) is the part of an antibody that binds to and is complementary to the epitope of its target antigen. The term “epitope” refers to a region of an antigen that is bound by an antibody. Epitopes may be defined as structural or functional. Functional epitopes are generally a subset of the structural epitopes and have those residues that directly contribute to the affinity of the interaction. Epitopes may also be conformational, that is, composed of non-linear amino acids. In certain embodiments, epitopes may include determinants that are chemically active surface groupings of molecules such as amino acids, sugar side chains, phosphoryl groups, or sulfonyl groups, and, in certain embodiments, may have specific three-dimensional structural characteristics, and/or specific charge characteristics.


An antibody antigen-binding site is provided by a set of complementarity determining regions (CDRs) in an antibody VH and/or VL domain, and is capable of binding the antigen. In an example, the antibody binding site is provided by a single variable domain, e.g., a heavy chain variable domain (VH domain) or a light chain variable domain (VL domain). In another example, the binding site is provided by a VH/VL pair (an Fv) or two or more such pairs.


The antibody variable domains are the portions of the light and heavy chains of antibodies that include amino acid sequences of complementarity determining regions (CDRs; ie., CDR1, CDR2, and CDR3), and framework regions (FRs). Thus, within each of the VH and VL domains are CDRs and FRs. A VH domain comprises a set of HCDRs, and a VL domain comprises a set of LCDRs. VH refers to the variable domain of the heavy chain. VL refers to the variable domain of the light chain. Each VH and VL is typically composed of three CDRs and four FRs, arranged from amino-terminus to carboxy-terminus in the following order: FR1, CDR1, FR2, CDR2, FR3, CDR3, FR4. Amino acid positions assigned to CDRs and FRs may be defined according to IMGT nomenclature. An antibody may comprise an antibody VH domain comprising a VH CDR1, CDR2 and CDR3 and a framework. It may alternatively or also comprise an antibody VL domain comprising a VL CDR1, CDR2 and CDR3 and a framework. Example sequences of antibody VH and VL domains and CDRs form part of the present disclosure. The CDRs are defined according to the IMGT system [9]. All VH and VL sequences, CDR sequences, sets of CDRs and sets of HCDRs and sets of LCDRs disclosed herein represent aspects and embodiments of the invention. As described herein, a “set of CDRs” comprises CDR1, CDR2 and CDR3. Thus, a set of HCDRs refers to HCDR1, HCDR2 and HCDR3, and a set of LCDRs refers to LCDR1, LCDR2 and LCDR3. Unless otherwise stated, a “set of CDRs” includes HCDRs and LCDRs.


An antibody may comprise one or more CDRs, e.g. a set of CDRs, within an antibody framework. The framework regions may be of human germline gene segment sequences. Thus, the antibody may be a human antibody having a VH domain comprising a set of HCDRs in a human germline framework. Normally the antibody also has a VL domain comprising a set of LCDRs, e.g. in a human germline framework. An antibody “gene segment”, e.g., a VH gene segment, D gene segment, or JH gene segment refers to oligonucleotide having a nucleic acid sequence from which that portion of an antibody is derived, e.g., a VH gene segment is an oligonucleotide comprising a nucleic acid sequence that corresponds to a polypeptide VH domain from FR1 to part of CDR3. Human v, d and j gene segments recombine to generate the VH domain, and human v and j segments recombine to generate the VL domain. The D domain or region refers to the diversity domain or region of an antibody chain. J domain or region refers to the joining domain or region of an antibody chain. Somatic hypermutation may result in an antibody VH or VL domain having framework regions that do not exactly match or align with the corresponding gene segments, but sequence alignment can be used to identify the closest gene segments and thus identify from which particular combination of gene segments a particular VH or VL domain is derived. When aligning antibody sequences with gene segments, the antibody amino acid sequence may be aligned with the amino acid sequence encoded by the gene segment, or the antibody nucleotide sequence may be aligned directly with the nucleotide sequence of the gene segment. Germline gene segments corresponding to framework regions of example antibodies described herein are indicated in Table 12.


An antibody may be a whole immunoglobulin, including constant regions, or may be an antibody fragment. An antibody fragment is a portion of an intact antibody, for example comprising the antigen binding and/or variable region of the intact antibody. The antibody fragment may include one or more constant region domains.


An antibody of the invention may be a human antibody or a chimaeric antibody comprising human variable regions and non-human (e.g., mouse) constant regions. The antibody of the invention for example has human variable regions, and optionally also has human constant regions.


Thus, antibodies optionally include constant regions or parts thereof, e.g., human antibody constant regions or parts thereof, such as a human IgG4 constant region. For example, a VL domain may be attached at its C-terminal end to antibody light chain kappa or lambda constant domains. Similarly, an antibody VH domain may be attached at its C-terminal end to all or part (e.g. a CH1 domain or Fc region) of an immunoglobulin heavy chain constant region derived from any antibody isotype, e.g. IgG, IgA, IgE and IgM and any of the isotype sub-classes, such as IgG1 or IgG4.


Antibodies may be generated with non-human constant regions. For example, when antibodies are produced in transgenic animals (examples of which are described elsewhere herein), chimaeric antibodies may be produced comprising human variable regions and non-human constant regions. The constant regions may be those endogenous to the host animal, e.g., mouse. Some transgenic animals generate fully human antibodies. Others have been engineered to generate antibodies comprising chimaeric heavy chains and fully human light chains. Where antibodies comprise one or more non-human constant regions, these may be replaced with human constant regions to provide antibodies more suitable for administration to humans as therapeutic compositions, as their immunogenicity is thereby reduced.


Digestion of whole (bivalent) immunoglobulins with the enzyme papain results in two identical (monovalent) antigen-binding fragments known as “Fab” fragments, and an “Fc” fragment. The Fc has no antigen-binding activity but has the ability to crystallize. “Fab” when used herein refers to a fragment of an antibody that includes one constant and one variable domain of each of the heavy and light chains. The term “Fc region” herein is used to define a C-terminal region of an immunoglobulin heavy chain, including native-sequence Fc regions and variant Fc regions. The “Fc fragment” refers to the carboxy-terminal portions of both H chains held together by disulfides.


Digestion of antibodies with the enzyme pepsin results in a bivalent F(ab′)2 fragment in which the two arms of the antibody molecule remain linked. The F(ab′)2 fragment is a bivalent fragment including two Fab fragments linked by a disulfide bridge at the hinge region. Single-chain antibodies (e.g., scFv) are another fragment. Two different monovalent monospecific antibody fragments such as scFv may be linked together to form a bivalent bispecific antibody.


“Fv” when used herein refers to the minimum fragment of an antibody that retains both antigen-recognition and antigen-binding sites. This region consists of a dimer of one heavy and one light chain variable domain in tight, non-covalent or covalent association. It is in this configuration that the three CDRs of each variable domain interact to define an antigen-binding site on the surface of the VH-VL dimer. Collectively, the six CDRs confer antigen-binding specificity to the antibody. However, even a single variable domain (or half of an Fv comprising only three CDRs specific for an antigen) has the ability to recognise and bind antigen, although usually at a lower affinity than the entire binding site.


Bispecific antibodies can have many possible formats. For a review, see Spiess, Zhai & Carter, Mol. Immunol. 67:95-106 2015, which illustrates 5 categories of bispecific antibodies:

    • Bispecific IgG (approx 150 kDa), exemplified by CrossMab, DAF (2-in-1), DAF (4-in-1), DutaMab, DT-IgG, Knobs in holes (KIH) with common light chain, KIH assembly, charge pair, Fab-arm exchange, SEEDbody, Triomab, LUZ-Y, mAb2 (referred to as “Fcab” in this review), κλ-body, orthogonal Fab;
    • Appended IgG (>150 kDa), exemplified by DVD-IgG, IgG(H)-scFv, scFv-(H)IgG, IgG(L)-scFv, scFv-(L)IgG, IgG(L,H)-Fv, IgG(H)-V, V(H)—IgG, IgG(L)-V, V(L)-IgG, KIH IgG-scFab, 2scFv-IgG, IgG-2scFv, scFv4-Ig, zybody, DVI-IgG (4-in-1);
    • Bispecific antibody fragments, exemplified by nanobody, nanobody-HSA, tandem-linked scFv (exemplified as “BiTE” in this review), Diabody, DART, TandAb, scDiabody, scDiabody-CH3, Diabody-CH3, Triple Body, Miniantibody, Minibody, TriBi minibody, scFv-CH3 KIH, Fab-scFv, scFv-CH-CL-scFv, F(ab′)2, F(ab′)2-scFv2, scFv-KIH, Fab-scFv-Fc, Tetravalent HCAb, scDiabody-Fc, Diabody-Fc, tandem scFv-Fc; intrabody;
    • Bispecific fusion proteins, exemplied by Dock and Lock, ImmTAC, HSAbody, scDiabody-HSA, Tandem scFv-toxin;
    • Bispecific antibody conjugates, exemplified by IgG-IgG, Cov-X-Body, scFv1-PEG-scFv2.


While the bispecific antigen binding molecules of the present invention are not restricted to any particular format or formats, some bispecific antibody formats are described here in more detail as examples of especially suitable molecules.


Preferably, the bispecific antibody is a dual binding antibody, i.e., a bispecific antibody in which both antigen binding domains are formed by a VH/VL pair. Dual binding antibodies include FIT-Ig (see WO2015/103072, incorporated herein by reference), mAb-dAb, dock and lock, Fab-arm exchange, SEEDbody, Triomab, LUZ-Y, Fcab, κλ-body, orthogonal Fab, scDiabody-Fc, diabody-Fc, tandem scFv-Fc, Fab-scFv-Fc, Fab-scFv, intrabody, BiTE, diabody, DART, TandAb, scDiabody, scDiabody-CH3, Diabody-CH3, Triple body, Miniantibody, minibody, scFv-CH3 KIH, scFv-CH-CL-scFv, F(ab′)2-scFv, scFv-KIH, Fab-scFv-Fc, tetravalent HCab, ImmTAC, knobs-in-holes, knobs-in-holes with common light chain, knobs-in-holes with common light chain and charge pairs, charge pairs, charge pairs with common light chain, DT-IgG, DutaMab, IgG(H)-scFv, scFv-(H)IgG, IgG(L)-scFv, scFv-(L)IgG, IgG(L,H)-Fv, IgG(H)-V, V(H)-IgG, IgG(L)-V, V(L)-IgG, KIH IgG-scFab, 2scFv-IgG, IgG-2scFv and scFv4-Ig.


In one embodiment, the bispecific antibody is a bispecific IgG comprising a FIXa-binding polypeptide arm and a FX-binding polypeptide arm, each polypeptide arm comprising a heavy chain and a light chain. The IgG is a tetrameric immunoglobulin comprising

    • a first pair of antibody heavy and light chains (heavy-light chain pair) comprising a FIXa binding Fv region,
    • a second heavy-light chain pair comprising a FX binding Fv region,
    • wherein each heavy chain comprises a VH domain and a constant region, and each light chain comprises a VL domain and a constant region, and wherein the first and second heavy-light chain pairs associate through heterodimerisation of their heavy chain constant regions to form the immunoglobulin tetramer.


Optionally, the two polypeptide arms comprise a common light chain, so the light chain of the first and second heavy-light chain pairs has an identical amino acid sequence (FIG. 5(A)). Alternatively the two polypeptide arms may comprise different light chains (FIG. 5(B)).


In another embodiment, the bispecific antibody is a linked pair of Fabs (bispecific F(ab′)2), comprising a FIXa-binding Fab and a FX-binding Fab, in which the Fab heavy chains are covalently coupled (FIG. 5(C)). Fab can be connected at their CH1 domains via disulphide bonding. One polypeptide binding arm (e.g., FIXa-binding arm) comprises a heavy-light chain pair VH1-CH1 and VL1-CL, and the other polypeptide binding arm (e.g., FX-binding arm) comprises a heavy-light chain pair VH2-CH1 and VL2-CL. Optionally, the two light chains (VL-CL) are identical in sequence, i.e., a common light chain is present.


In another embodiment, the bispecific antibody is a tandem-linked scFv pair, comprising a first scFv connected to a second scFv, optionally via a linker. The molecule can be produced with the antigen-binding arms in either orientation, i.e., the first scFv VL1-VH1 binding FIXa and the second scFv VL2-VH2 binding FX, or the first scFv VL1-VH1 binding FX and the second scFv VL2-VH2 binding FIXa. See FIG. 5(D).


As illustrated in the above embodiments, bispecific antibody may be monovalent for binding FIXa and for binding FX. In alternative embodiments, the bispecific antibody may be bivalent for one or both target antigens. For example, the antibody may be a FIT-Ig comprising two FIXa-binding Fab domains and two FX-binding Fab domains (FIG. 6). This format thus comprises two FIXa binding polypeptide arms and two FX binding polypeptide arms. The FIXa binding Fabs may be the “inner” Fab pair and the FX binding Fabs the “outer” Fab pair, or vice versa. FIT-Ig format was described in WO2015/103072—the description of the FIT-Ig scaffold is incorporated herein by reference.


Alternatively, bispecific antibody may be presented in DVD-Ig format. DVD-Ig was described by DiGiammarino et al., “Design and generation of DVD-Ig™ molecules for dual-specific targeting”, Meth. Mo. Biol., 889:145-156 2012.


Another doubly bivalent format for a bispecific antibody is mAb2, comprising two Fab domains and an Fc region in which the two CH3 domains each have three binding loops forming an antigen binding site, the engineered CH3 domains being referred to as the Fcab region. The technology behind the Fcab/mAb2 format is described in more detail in WO2008/003103, and the description of the mAb2 format is incorporated herein by reference.


Antibody Constant Regions


As discussed above, antibodies can be provided in various isotypes and with different constant regions. The Fc region of antibodies is recognised by Fc receptors and determines the ability of the antibody to mediate cellular effector functions, including antibody-dependent cell-mediated cytotoxicity (ADCC) activity, complement dependent cytotoxicity (CDC) activity and antibody-dependent cell phagocytosis (ADCP) activity. These cellular effector functions involve recruitment of cells bearing Fc receptors to the site of the target cells, resulting in killing of the antibody-bound cell.


In the context of the present invention it is desirable to avoid cellular effector functions such as ADCC, ADCP and/or CDC. Therefore, bispecific antigen-binding molecules according to the present invention may lack Fc effector function, for example they may contain Fc regions that do not mediate ADCC, ADCP and/or CDC, or they may lack Fc regions or lack antibody constant regions entirely. An antibody may have a constant region which is effector null.


An antibody may have a heavy chain constant region that binds one or more types of Fc receptor but does not induce cellular effector functions, i.e., does not mediate ADCC, CDC or ADCP activity. Such a constant region may be unable to bind the particular Fc receptor(s) responsible for triggering ADCC, CDC or ADCP activity.


An antibody may have a heavy chain constant region that does not bind Fcγ receptors, for example the constant region may comprise a Leu235Glu mutation (i.e., where the wild type leucine residue is mutated to a glutamic acid residue), which may be referred to as an “E” mutation, e.g., IgG4-E. Another optional mutation for a heavy chain constant region is Ser228Pro (“P” mutation), which increases stability by reducing Fab arm exchange. A heavy chain constant region may be an IgG4 comprising both the Leu235Glu mutation and the Ser228Pro mutation. This “IgG4-PE” heavy chain constant region is effector null. An alternative effector null human constant region is a disabled IgG1.


As discussed below, in bispecific IgG formats or other antibody formats where the different antigen binding arms are heterodimerised via constant regions, the constant regions may be engineered to promote heterodimer formation over homodimer formation and/or to facilitate purification of heterodimers from a mixture of different species.


Engineering of Bispecific Antibodies to Facilitate Heterodimer Formation and/or Purification


One of the difficulties with using bispecific antibodies in the clinic has historically been the difficulty of producing them in large quantities and at pharmaceutical grade purity. The “traditional” bispecific IgG format comprises two different pairs of heavy and light chains, thus 4 different polypeptide chains, which if expressed together could assemble into 10 different potential antibody molecules. The mixture of species will include homodimers (homodimeric anti-FIXa binding arms and homodimeric anti-FX binding arms), molecules in which one or both light chains are swapped between the H-L pairs, as well as the “correct” bispecific heterodimeric structure.


Alternative molecular formats have been developed which avoid this potential mis-pairing, and several examples are provided herein. These include F(ab′)2, e.g., prepared by chemical coupling or leucine zipper (fos:jun) assembly, diabodies, and scFv heterodimers. Nevertheless, it remains desirable to be able to use bispecific IgG, to reflect the native structure of antibodies in the bloodstream and to minimise immunogenicity of the administered therapeutic molecule. Additionally, a full length bispecific antibody may have a longer serum half-life.


“Knobs into holes” technology for making bispecific antibodies was described in [12] and in U.S. Pat. No. 5,731,168, both incorporated herein by reference. The principle is to engineer paired CH3 domains of heterodimeric heavy chains so that one CH3 domain contains a “knob” and the other CH3 domains contains a “hole” at a sterically opposite position. Knobs are created by replacing small amino acid side chain at the interface between the CH3 domains, while holes are created by replacing large side chains with smaller ones. The knob is designed to insert into the hole, to favour heterodimerisation of the different CH3 domains while destabilising homodimer formation. In in a mixture of antibody heavy and light chains that assemble to form a bispecific antibody, the proportion of IgG molecules having paired heterodimeric heavy chains is thus increased, raising yield and recovery of the active molecule


Mutations Y349C and/or T366W may be included to form “knobs” in an IgG CH3 domain. Mutations E356C, T366S, L368A and/or Y407V may be included to form “holes” in an IgG CH3 domain. Knobs and holes may be introduced into any human IgG CH3 domain, e.g., an IgG1, IgG2, IgG3 or IgG4 CH3 domain. A preferred example is IgG4. As noted, the IgG4 may include further modifications such as the “P” and/or “E” mutations. An example IgG4-PE sequence and other example constant regions including knobs-into-holes mutations are shown in Table 11. The IgG4 type a (“ra”) sequence contains substitutions Y349C and T366W (“knobs”), and the IgG4 type b (“γb”) sequence contains substitutions E356C, T366S, L368A, and Y407V (“holes”). Both ra and γb also contain the “P” substitution at position 228 in the hinge (S228P), to stabilise the hinge region of the heavy chain. Both ra and γb also contain the “E” substitution in the CH2 region at position 235 (L235S), to abolish binding to FcγR. Thus the relevant sequence of the IgG4-PE heavy chain is ppcpPcpapefEggps (SEQ ID NO: 401). A bispecific antigen binding molecule of the present invention may contain an IgG4 PE human heavy chain constant region (e.g., SEQ ID NO: 143), optionally two such paired constant regions, optionally wherein one has “knobs” mutations and one has “holes” mutations, e.g., wherein one heavy chain constant region has a sequence SEQ ID NO: 144 (knobs) and one heavy chain constant region has a sequence SEQ ID NO: 145 (holes).


A further advance in bispecific IgG engineering was the idea of using a common light chain, as described in WO98/50431. Bispecific antibodies comprising two heavy-light chain pairs were described, in which the variable light chains of both heavy-light chain pairs had a common sequence. WO98/50431 described combining the common light chain approach with specific complementary interactions in the heavy chain heterodimerisation interface (such as knobs-into-holes) to promote heterodimer formation and hinder homodimer formation. In combination, these approaches enhance formation of the desired heterodimer relative to undesired heterodimers and homodimers.


While knobs-into-holes technology involves engineering amino acid side chains to create complementary molecular shapes at the interface of the paired CH3 domains in the bispecific heterodimer, another way to promote heterodimer formation and hinder homodimer formation is to engineer the amino acid side chains to have opposite charges. Association of CH3 domains in the heavy chain heterodimers is favoured by the pairing of oppositely charged residues, while paired positive charges or paired negative charges would make homodimer formation less energetically favourable. WO2006/106905 described a method for producing a heteromultimer composed of more than one type of polypeptide (such a heterodimer of two different antibody heavy chains) comprising a substitution in an amino acid residue forming an interface between said polypeptides such that heteromultimer association will be regulated, the method comprising:

    • (a) modifying a nucleic acid encoding an amino acid residue forming the interface between polypeptides from the original nucleic acid, such that the association between polypeptides forming one or more multimers will be inhibited in a heteromultimer that may form two or more types of multimers;
    • (b) culturing host cells such that a nucleic acid sequence modified by step (a) is expressed; and
    • (c) recovering said heteromultimer from the host cell culture,
    • wherein the modification of step (a) is modifying the original nucleic acid so that one or more amino acid residues are substituted at the interface such that two or more amino acid residues, including the mutated residue(s), forming the interface will carry the same type of positive or negative charge.


An example of this is to suppress association between heavy chains by introducing electrostatic repulsion at the interface of the heavy chain homodimers, for example by modifying amino acid residues that contact each other at the interface of the CH3 domains, including:

    • positions 356 and 439
    • positions 357 and 370
    • positions 399 and 409,
    • the residue numbering being according to the EU numbering system.


By modifying one or more of these pairs of residues to have like charges (both positive or both negative) in the CH3 domain of a first heavy chain, the pairing of heavy chain homodimers is inhibited by electrostatic repulsion. By engineering the same pairs or pairs of residues in the CH3 domain of a second (different) heavy chain to have an opposite charge compared with the corresponding residues in the first heavy chain, the heterodimeric pairing of the first and second heavy chains is promoted by electrostatic attraction.


In one example, introduction of charge pairs in the antibody VH and VL domains was used to inhibit the formation of “incorrect” VH-VL pairs (pairing of VH from one antibody with VL of the other antibody). In one example, Q residues in the VH and VL were changed to K or R (positive), or to E or D (negative), to inhibit hydrogen bonding between the Q side chains and to introduce electrostatic repulsion. In another example, amino acids at the heavy chain constant region CH3 interface were modified to introduce charge pairs, the mutations being listed in Table 1 of WO2006/106905. It was reported that modifying the amino acids at heavy chain positions 356, 357, 370, 399, 409 and 439 to introduce charge-induced molecular repulsion at the CH3 interface had the effect of increasing efficiency of formation of the intended bispecific antibody. WO2006/106905 also exemplified bispecific IgG antibodies binding FX and FIXa in which the CH3 domains of IgG4 were engineered with knobs-into-holes mutations. Type a Type a (IgG4γa) was an IgG4 substituted at Y349C and T366W, and type b (IgG4γb) was an IgG4 substituted at E356C, T366S, L368A, and Y407V.


Further examples of charge pairs were disclosed in WO2013/157954, which described a method for producing a heterodimeric CH3 domain-comprising molecule from a single cell, the molecule comprising two CH3 domains capable of forming an interface. The method comprised providing in the cell

    • (a) a first nucleic acid molecule encoding a first CH3 domain-comprising polypeptide chain, this chain comprising a K residue at position 366 according to the EU numbering system and
    • (b) a second nucleic acid molecule encoding a second CH3 domain-comprising polypeptide chain, this chain comprising a D residue at position 351 according to the EU numbering system,
    • the method further comprising the step of culturing the host cell, allowing expression of the two nucleic acid molecules and harvesting the heterodimeric CH3 domain-comprising molecule from the culture.


Further methods of engineering electrostatic interactions in polypeptide chains to promote heterodimer formation over homodimer formation were described in WO2011/143545.


Another example of engineering at the CH3-CH3 interface is strand-exchange engineered domain (SEED) CH3 heterodimers. The CH3 domains are composed of alternating segments of human IgA and IgG CH3 sequences, which form pairs of complementary SEED heterodimers referred to as “SEED-bodies” [10; WO2007/110205].


Bispecifics have also been produced with heterodimerised heavy chains that are differentially modified in the CH3 domain to alter their affinity for binding to a purification reagent such as Protein A. WO2010/151792 described a heterodimeric bispecific antigen-binding protein comprising

    • a first polypeptide comprising, from N-terminal to C-terminal, a first epitope-binding region that selectively binds a first epitope, an immunoglobulin constant region that comprises a first CH3 region of a human IgG selected from IgG1, IgG2, and IgG4; and
    • a second polypeptide comprising, from N-terminal to C-terminal, a second epitope-binding region that selectively binds a second epitope, an immunoglobulin constant region that comprises a second CH3 region of a human IgG selected from IgG1, IgG2, and IgG4, wherein the second CH3 region comprises a modification that reduces or eliminates binding of the second CH3 domain to Protein A.


The bispecifics of the present invention may employ any of these techniques and molecular formats as desired.


Generating and Modifying Antibodies


Methods for identifying and preparing antibodies are well known. Antibodies that bind an antigen of interest may be generated using transgenic mice (eg, the Kymouse™, Velocimouse®, Omnimouse®, Xenomouse®, HuMab Mouse® or MeMo Mouse®), rats (e.g., the Omnirat®), camelids, sharks, rabbits, chickens or other non-human animals immunised with the antigen, followed optionally by humanisation of the constant regions and/or variable regions to produce human or humanised antibodies. In an example, display technologies can be used, such as yeast, phage or ribosome display, as will be apparent to the skilled person. Standard affinity maturation, e.g., using a display technology, can be performed in a further step after isolation of an antibody lead from a transgenic animal, phage display library or other library. Representative examples of suitable technologies are described in US20120093818 (Amgen, Inc), which is incorporated by reference herein in its entirety, eg, the methods set out in paragraphs [0309] to [0346].


Following generation of antibodies, whether by immunisation or screening of in vitro libraries, nucleic acid encoding an antibody heavy chain variable domain and/or an antibody light chain variable domain of a selected antibody may be isolated. Such nucleic acid may encode the full antibody heavy chain and/or light chain, or the variable domain(s) without associated constant region(s). Encoding nucleotide sequences may be obtained directly from antibody-producing cells of a mouse, or B cells may be immortalised or fused to generate hybridomas expressing the antibody, and encoding nucleic acid obtained from such cells. Optionally, nucleic acid encoding the variable domain(s) is then conjugated to a nucleotide sequence encoding a human heavy chain constant region and/or human light chain constant region, to provide nucleic acid encoding a human antibody heavy chain and/or human antibody light chain, e.g., encoding an antibody comprising both the heavy and light chain. This step is particularly useful where the immunised mammal produces chimaeric antibodies with non-human constant regions, which are preferably replaced with human constant regions to generate an antibody that will be less immunogenic when administered to humans as a medicament. Provision of particular human isotype constant regions is also significant for determining the effector function of the antibody, and a number of suitable heavy chain constant regions are discussed herein.


Other alterations to nucleic acid encoding the antibody heavy and/or light chain variable domain may be performed, such as mutation of residues and generation of variants, as described herein.


The isolated (optionally mutated) nucleic acid may be introduced into host cells, e.g., CHO cells as discussed. Host cells are then cultured under conditions for expression of the antibody, or of the antibody heavy and/or light chain variable domain, in any desired antibody format. Some possible antibody formats are described herein, e.g., whole immunoglobulins, antigen-binding fragments, and other designs.


Variable domain amino acid sequence variants of any of the VH and VL domains or CDRs whose sequences are specifically disclosed herein may be employed in accordance with the present invention, as discussed.


There are many reasons why it may be desirable to create variants, which include optimising the antibody sequence for large-scale manufacturing, facilitating purification, enhancing stability or improving suitability for inclusion in a desired pharmaceutical formulation. Protein engineering work can be performed at one or more target residues in the antibody sequence, e.g., to substituting one amino acid with an alternative amino acid (optionally, generating variants containing all naturally occurring amino acids at this position, with the possible exception of Cys and Met), and monitoring the impact on function and expression to determine the best substitution. It is in some instances undesirable to substitute a residue with Cys or Met, or to introduce these residues into a sequence, as to do so may generate difficulties in manufacturing—for instance through the formation of new intramolecular or intermolecular cysteine-cysteine bonds. Where a lead candidate has been selected and is being optimised for manufacturing and clinical development, it will generally be desirable to change its antigen-binding properties as little as possible, or at least to retain the affinity and potency of the parent molecule. However, variants may also be generated in order to modulate key antibody characteristics such as affinity, cross-reactivity or neutralising potency.


One or more amino acid mutations may optionally be made in framework regions of an antibody VH or VL domain disclosed herein. For example, one or more residues that differ from the corresponding human germline segment sequence may be reverted to germline. Human germline gene segment sequences corresponding to VH and VL domains of example antibodies herein are indicated in Table 12.


In a bispecific antigen binding molecule, an antigen-binding site may comprise a set of H and/or L CDRs of any of the disclosed anti-FIX or anti-FX antibodies with one or more amino acid mutations within the disclosed set of H and/or L CDRs. The mutation may be an amino acid substitution, deletion or insertion. Thus for example there may be one or more amino acid substitutions within the disclosed set of H and/or L CDRs. For example, there may be up to 12, 11, 10, 9, 8, 7, 6, 5, 4, 3 or 2 mutations e.g. substitutions, within the set of H and/or L CDRs. For example, there may be up to 6, 5, 4, 3 or 2 mutations, e.g. substitutions, in HCDR3 and/or there may be up to 6, 5, 4, 3, or 2 mutations, e.g. substitutions, in LCDR3.


An antibody may comprise a VH domain that has at least 60, 70, 80, 85, 90, 95, 98 or 99% amino acid sequence identity with a VH domain as shown in the Tables, and/or comprising a VL domain that has at least 60, 70, 80, 85, 90, 95, 98 or 99% amino acid sequence identity with a VL domain of any of those antibodies. Algorithms that can be used to calculate % identity of two amino acid sequences include e.g. BLAST, FASTA, or the Smith-Waterman algorithm, e.g. employing default parameters. Particular variants may include one or more amino acid sequence alterations (addition, deletion, substitution and/or insertion of an amino acid residue).


Alterations may be made in one or more framework regions and/or one or more CDRs. Variants are optionally provided by CDR mutagenesis. The alterations normally do not result in loss of function, so an antibody comprising a thus-altered amino acid sequence may retain an ability to bind its antigen. It may retain the same quantitative binding ability as an antibody in which the alteration is not made, e.g. as measured in an assay described herein. The antibody comprising a thus-altered amino acid sequence may have an improved ability to bind its antigen.


Alteration may comprise replacing one or more amino acid residue with a non-naturally occurring or non-standard amino acid, modifying one or more amino acid residue into a non-naturally occurring or non-standard form, or inserting one or more non-naturally occurring or non-standard amino acid into the sequence. Examples of numbers and locations of alterations in sequences of the invention are described elsewhere herein. Naturally occurring amino acids include the 20 “standard” L-amino acids identified as G, A, V, L, I, M, P, F, W, S, T, N, Q, Y, C, K, R, H, D, E by their standard single-letter codes. Non-standard amino acids include any other residue that may be incorporated into a polypeptide backbone or result from modification of an existing amino acid residue. Non-standard amino acids may be naturally occurring or non-naturally occurring.


The term “variant” as used herein refers to a peptide or nucleic acid that differs from a parent polypeptide or nucleic acid by one or more amino acid or nucleic acid deletions, substitutions or additions, yet retains one or more specific functions or biological activities of the parent molecule. Amino acid substitutions include alterations in which an amino acid is replaced with a different naturally-occurring amino acid residue. Such substitutions may be classified as “conservative”, in which case an amino acid residue contained in a polypeptide is replaced with another naturally occurring amino acid of similar character either in relation to polarity, side chain functionality or size. Such conservative substitutions are well known in the art. Substitutions encompassed by the present invention may also be “non-conservative”, in which an amino acid residue which is present in a peptide is substituted with an amino acid having different properties, such as naturally-occurring amino acid from a different group (e.g., substituting a charged or hydrophobic amino; acid with alanine), or alternatively, in which a naturally-occurring amino acid is substituted with a non-conventional amino acid. In some embodiments amino acid substitutions are conservative. Also encompassed within the term variant when used with reference to a polynucleotide or polypeptide, refers to a polynucleotide or polypeptide that can vary in primary, secondary, or tertiary structure, as compared to a reference polynucleotide or polypeptide, respectively (e.g., as compared to a wild-type polynucleotide or polypeptide).


In some aspects, one can use “synthetic variants”, “recombinant variants”, or “chemically modified” polynucleotide variants or polypeptide variants isolated or generated using methods well known in the art. “Modified variants” can include conservative or non-conservative amino acid changes, as described below. Polynucleotide changes can result in amino acid substitutions, additions, deletions, fusions and truncations in the polypeptide encoded by the reference sequence. Some aspects use include insertion variants, deletion variants or substituted variants with substitutions of amino acids, including insertions and substitutions of amino acids and other molecules) that do not normally occur in the peptide sequence that is the basis of the variant, for example but not limited to insertion of ornithine which do not normally occur in human proteins. The term “conservative substitution,” when describing a polypeptide, refers to a change in the amino acid composition of the polypeptide that does not substantially alter the polypeptide's activity. For example, a conservative substitution refers to substituting an amino acid residue for a different amino acid residue that has similar chemical properties (e.g., acidic, basic, positively or negatively charged, polar or nonpolar, etc.). Conservative amino acid substitutions include replacement of a leucine with an isoleucine or valine, an aspartate with a glutamate, or a threonine with a serine. Conservative substitution tables providing functionally similar amino acids are well known in the art. For example, the following six groups each contain amino acids that are conservative substitutions for one another: 1) Alanine (A), Serine (S), Threonine (T); 2) Aspartic acid (D), Glutamic acid (E); 3) Asparagine (N), Glutamine (Q); 4) Arginine (R), Lysine (K); 5) Isoleucine (I), Leucine (L), Methionine (M), Valine (V); and 6) Phenylalanine (F), Tyrosine (Y), Tryptophan (W). (See also Creighton, Proteins, W. H. Freeman and Company (1984), incorporated by reference in its entirety.) In some embodiments, individual substitutions, deletions or additions that alter, add or delete a single amino acid or a small percentage of amino acids can also be considered “conservative substitutions” if the change does not reduce the activity of the peptide. Insertions or deletions are typically in the range of about 1 to 5 amino acids. The choice of conservative amino acids may be selected based on the location of the amino acid to be substituted in the peptide, for example if the amino acid is on the exterior of the peptide and expose to solvents, or on the interior and not exposed to solvents.


One can select the amino acid that will substitute an existing amino acid based on the location of the existing amino acid, including its exposure to solvents (i.e., if the amino acid is exposed to solvents or is present on the outer surface of the peptide or polypeptide as compared to internally localized amino acids not exposed to solvents). Selection of such conservative amino acid substitutions are well known in the art, for example as disclosed in Dordo et al, J. Mol Biol, 1999, 217, 721-739 and Taylor et al, J. Theor. Biol. 119(1986); 205-218 and S. French and B. Robson, J. Mol. Evol. 19(1983) 171. Accordingly, one can select conservative amino acid substitutions suitable for amino acids on the exterior of a protein or peptide (i.e. amino acids exposed to a solvent), for example, but not limited to, the following substitutions can be used: substitution of Y with F, T with S or K, P with A, E with D or Q, N with D or G, R with K, G with N or A, T with S or K, D with N or E, I with L or V, F with Y, S with T or A, R with K, G with N or A, K with R, A with S, K or P.


In alternative embodiments, one can also select conservative amino acid substitutions encompassed suitable for amino acids on the interior of a protein or peptide, for example one can use suitable conservative substitutions for amino acids is on the interior of a protein or peptide (i.e. the amino acids are not exposed to a solvent), for example but not limited to, one can use the following conservative substitutions: where Y is substituted with F, T with A or S, I with L or V, W with Y, M with L, N with D, G with A, T with A or S, D with N, I with L or V, F with Y or L, S with A or T and A with S, G, T or V. In some embodiments, non-conservative amino acid substitutions are also encompassed within the term of variants.


The invention includes methods of producing polypeptide binding arms containing VH and/or VL domain variants of the antibody VH and/or VL domains shown in the Tables herein. FIXa binding polypeptide arms comprising variant VH domains may be produced by a method comprising

    • (i) providing, by way of addition, deletion, substitution or insertion of one or more amino acids in the amino acid sequence of a parent antibody VH domain, an antibody VH domain that is an amino acid sequence variant of the parent antibody VH domain,
    • wherein the parent antibody VH domain is the VH domain N192H, N212H, N205H, N211H, N203H, N128H, N215H, N216H, N217H, N218H, N219H, N220H, N221H, N222H, N223H, N224H, N225H, N226H, N227H, N228H or N229H or is a VH domain comprising the heavy chain complementarity determining regions of any of those VH domains,
    • (ii) optionally combining the VH domain thus provided with a VL domain, to provide a VH/VL combination, and
    • (iii) testing the VH domain or VH/VL domain combination thus provided to identify an antibody with one or more desired characteristics.


The VH domain may be any VH domain whose sequence is shown in Table 9A, or any VH domain comprising a set of HCDRs (HCDR1, HCDR2 and HCDR3) of a VH domain shown in Table 9A. The VH domain may be the N436 VH domain (SEQ ID NO: 324). The VH domain may be the N128 VH domain (SEQ ID NO: 5).


Desired characteristics of FIXa-binding polypeptide arms, and of bispecific anti-FIXa/FX binding molecules comprising them, are detailed elsewhere herein. For example, the method may comprise confirming that the VH domain or VH/VL domain combination binds FIXa as described herein.


When VL domains are included in the method, the VL domain may be the N128L VL domain or may be a variant provided by way of addition, deletion, substitution or insertion of one or more amino acids in the amino acid sequence of the N128L VL domain, or may be a VL domain comprising the light chain complementarity determining regions of the N128L VL domain.


Methods of generating variant antibodies may optionally comprise producing copies of the antibody or VH/VL domain combination. Methods may further comprise producing a bispecific antibody comprising the FIXa binding polypeptide arm, for example by expression of encoding nucleic acid. Suitable methods of expression, including recombinant expression in host cells, are set out in detail herein.


Encoding Nucleic Acids and Methods of Expression


Isolated nucleic acid may be provided, encoding bispecific antigen binding molecules, e.g., bispecific antibodies, according to the present invention. Nucleic acid may be DNA and/or RNA. Genomic DNA, cDNA, mRNA or other RNA, of synthetic origin, or any combination thereof can encode an antibody.


The present invention provides constructs in the form of plasmids, vectors, transcription or expression cassettes which comprise at least one polynucleotide as above. Exemplary nucleotide sequences are included in the Tables. Reference to a nucleotide sequence as set out herein encompasses a DNA molecule with the specified sequence, and encompasses a RNA molecule with the specified sequence in which U is substituted for T, unless context requires otherwise.


The present invention also provides a recombinant host cell that comprises one or more nucleic acids encoding the antigen binding molecule. Methods of producing the encoded molecule may comprise expression from the nucleic acid, e.g., by culturing recombinant host cells containing the nucleic acid. The bispecific molecule may thus be obtained, and may be isolated and/or purified using any suitable technique, then used as appropriate. A method of production may comprise formulating the product into a composition including at least one additional component, such as a pharmaceutically acceptable excipient.


Systems for cloning and expression of a polypeptide in a variety of different host cells are well known. Suitable host cells include bacteria, mammalian cells, plant cells, filamentous fungi, yeast and baculovirus systems and transgenic plants and animals.


The expression of antibodies and antibody fragments in prokaryotic cells is well established in the art. A common bacterial host is E. coli. Expression in eukaryotic cells in culture is also available to those skilled in the art as an option for production. Mammalian cell lines available in the art for expression of a heterologous polypeptide include Chinese hamster ovary (CHO) cells, HeLa cells, baby hamster kidney cells, NSO mouse melanoma cells, YB2/0 rat myeloma cells, human embryonic kidney cells, human embryonic retina cells and many others.


Vectors may contain appropriate regulatory sequences, including promoter sequences, terminator sequences, polyadenylation sequences, enhancer sequences, marker genes and other sequences as appropriate. Nucleic acid encoding an antibody can be introduced into a host cell. Nucleic acid can be introduced to eukaryotic cells by various methods, including calcium phosphate transfection, DEAE-Dextran, electroporation, liposome-mediated transfection and transduction using retrovirus or other virus, e.g. vaccinia or, for insect cells, baculovirus. Introducing nucleic acid in the host cell, in particular a eukaryotic cell may use a viral or a plasmid based system. The plasmid system may be maintained episomally or may be incorporated into the host cell or into an artificial chromosome. Incorporation may be either by random or targeted integration of one or more copies at single or multiple loci. For bacterial cells, suitable techniques include calcium chloride transformation, electroporation and transfection using bacteriophage. The introduction may be followed by expressing the nucleic acid, e.g., by culturing host cells under conditions for expression of the gene, then optionally isolating or purifying the antibody.


Nucleic acid of the invention may be integrated into the genome (e.g. chromosome) of the host cell. Integration may be promoted by inclusion of sequences that promote recombination with the genome, in accordance with standard techniques.


The present invention also provides a method that comprises using nucleic acid described herein in an expression system in order to express the bispecific antigen binding molecule. Desirably, the antigen-binding molecules are expressed at a yield of at least 0.5 g/L in the cell supernatant after initial fermentation, preferably at a yield of >2 g/L. Solubility should be >10 mg/ml, preferably >50 mg/ml, without significant aggregation or degradation of the molecules.


Formulation and Administration


The bispecific antigen-binding molecules (“bispecifics”) according to the present invention, and their encoding nucleic acid molecules, will usually be provided in isolated form. The bispecifics VH and/or VL domains, and nucleic acids may be provided purified from their natural environment or their production environment. Isolated antigen-binding molecules and isolated nucleic acid will be free or substantially free of material with which they are naturally associated, such as other polypeptides or nucleic acids with which they are found in vivo, or the environment in which they are prepared (e.g., cell culture) when such preparation is by recombinant DNA technology in vitro. Optionally an isolated antigen-binding molecule or nucleic acid (1) is free of at least some other proteins with which it would normally be found, (2) is essentially free of other proteins from the same source, e.g., from the same species, (3) is expressed by a cell from a different species, (4) has been separated from at least about 50 percent of polynucleotides, lipids, carbohydrates, or other materials with which it is associated in nature, (5) is operably associated (by covalent or noncovalent interaction) with a polypeptide with which it is not associated in nature, or (6) does not occur in nature.


Bispecifics or their encoding nucleic acids may be formulated with diluents or adjuvants and still for practical purposes be isolated—for example they may be mixed with carriers if used to coat microtitre plates for use in immunoassays, and may be mixed with pharmaceutically acceptable carriers or diluents when used in therapy. As described elsewhere herein, other active ingredients may also be included in therapeutic preparations. The antigen binding molecules may be glycosylated, either naturally in vivo or by systems of heterologous eukaryotic cells such as CHO cells, or they may be (for example if produced by expression in a prokaryotic cell) unglycosylated. The invention encompasses antibodies having a modified glycosylation pattern.


Typically, an isolated product constitutes at least about 5%, at least about 10%, at least about 25%, or at least about 50% of a given sample. A bispecific may be substantially free from proteins or polypeptides or other contaminants that are found in its natural or production environment that would interfere with its therapeutic, diagnostic, prophylactic, research or other use.


The invention provides therapeutic compositions comprising the bispecifics described herein. Therapeutic compositions comprising nucleic acid encoding such bispecifics are also provided. Encoding nucleic acids are described in more detail elsewhere herein and include DNA and RNA, e.g., mRNA. In therapeutic methods described herein, use of nucleic acid encoding the bispecific, and/or of cells containing such nucleic acid, may be used as alternatives (or in addition) to compositions comprising the bispecific molecule itself. Cells containing nucleic acid encoding the bispecific, optionally wherein the nucleic acid is stably integrated into the genome, thus represent medicaments for therapeutic use in a patient. Nucleic acid encoding the bispecific may be introduced into human cells derived from the intended patient and modified ex vivo. Administration of cells containing the encoding nucleic acid to the patient provides a reservoir of cells capable of expressing the bispecific, which may provide therapeutic benefit over a longer term compared with administration of isolated nucleic acid or the isolated bispecific molecule. Nucleic acid encoding the bispecific may be provided for use in gene therapy, comprising introducing the encoding nucleic acid into cells of the patient in vivo, so that the nucleic acid is expressed in the patient's cells and provides a therapeutic effect such as compensating for hereditary factor VIII deficiency.


Compositions may contain suitable carriers, excipients, and other agents that are incorporated into formulations to provide improved transfer, delivery, tolerance, and the like. A multitude of appropriate formulations can be found in the formulary known to all pharmaceutical chemists: Remington's Pharmaceutical Sciences, Mack Publishing Company, Easton, Pa. These formulations include, for example, powders, pastes, ointments, jellies, waxes, oils, lipids, lipid (cationic or anionic) containing vesicles (such as LIPOFECTINT™), DNA conjugates, anhydrous absorption pastes, oil-in-water and water-in-oil emulsions, emulsions carbowax (polyethylene glycols of various molecular weights), semi-solid gels, and semi-solid mixtures containing carbowax. See also Powell et al. “Compendium of excipients for parenteral formulations” PDA (1998) J Pharm Sci Technol 52:238-311. Compositions may comprise the antibody or nucleic acid in combination with medical injection buffer and/or with adjuvant.


Bispecifics, or their encoding nucleic acids, may be formulated for the desired route of administration to a patient, e.g., in liquid (optionally aqueous solution) for injection.


Various delivery systems are known and can be used to administer the pharmaceutical composition of the invention. Methods of introduction include, but are not limited to, intradermal, intramuscular, intraperitoneal, intravenous, subcutaneous, intranasal, epidural, and oral routes. The composition may be administered by any convenient route, for example by infusion or bolus injection, by absorption through epithelial or mucocutaneous linings (e.g., oral mucosa, rectal and intestinal mucosa, etc.) and may be administered together with other biologically active agents. Administration can be systemic or local. The antigen-binding molecules are preferably administered by subcutaneous injection.


The pharmaceutical composition can be also delivered in a vesicle, in particular a liposome (see Langer (1990) Science 249:1527-1533; Treat et al. (1989) in Liposomes in the Therapy of Infectious Disease and Cancer, Lopez Berestein and Fidler (eds.), Liss, New York, pp. 353-365; Lopez-Berestein, ibid., pp. 317-327; see generally ibid.).


In certain situations, the pharmaceutical composition can be delivered in a controlled release system. In one embodiment, a pump may be used (see Langer, supra; Sefton (1987) CRC Crit. Ref. Biomed. Eng. 14:201). In another embodiment, polymeric materials can be used; see, Medical Applications of Controlled Release, Langer and Wise (eds.), CRC Pres., Boca Raton, Fla. (1974). In yet another embodiment, a controlled release system can be placed in proximity of the composition's target, thus requiring only a fraction of the systemic dose (see, e.g., Goodson, in Medical Applications of Controlled Release, supra, vol. 2, pp. 115-138, 1984).


The injectable preparations may include dosage forms for intravenous, subcutaneous, intracutaneous and intramuscular injections, drip infusions, etc. These injectable preparations may be prepared by methods publicly known. For example, the injectable preparations may be prepared, e.g., by dissolving, suspending or emulsifying the antibody or its salt described above in a sterile aqueous medium or an oily medium conventionally used for injections. As the aqueous medium for injections, there are, for example, physiological saline, an isotonic solution containing glucose and other auxiliary agents, etc., which may be used in combination with an appropriate solubilizing agent such as an alcohol (e.g., ethanol), a polyalcohol (e.g., propylene glycol, polyethylene glycol), a nonionic surfactant [e.g., polysorbate 80, HCO-50 (polyoxyethylene (50 mol) adduct of hydrogenated castor oil)], etc. As the oily medium, there are employed, e.g., sesame oil, soybean oil, etc., which may be used in combination with a solubilizing agent such as benzyl benzoate, benzyl alcohol, etc. The injection thus prepared can be filled in an appropriate ampoule. A pharmaceutical composition of the present invention can be delivered subcutaneously or intravenously with a standard needle and syringe. It is envisaged that treatment will not be restricted to use in the clinic. Therefore, subcutaneous injection using a needle-free device is also advantageous. With respect to subcutaneous delivery, a pen delivery device readily has applications in delivering a pharmaceutical composition of the present invention. Such a pen delivery device can be reusable or disposable. A reusable pen delivery device generally utilizes a replaceable cartridge that contains a pharmaceutical composition. Once all of the pharmaceutical composition within the cartridge has been administered and the cartridge is empty, the empty cartridge can readily be discarded and replaced with a new cartridge that contains the pharmaceutical composition. The pen delivery device can then be reused. In a disposable pen delivery device, there is no replaceable cartridge. Rather, the disposable pen delivery device comes prefilled with the pharmaceutical composition held in a reservoir within the device. Once the reservoir is emptied of the pharmaceutical composition, the entire device is discarded. Numerous reusable pen and autoinjector delivery devices have applications in the subcutaneous delivery of a pharmaceutical composition of the present invention. Examples include, but certainly are not limited to AUTOPEN™ (Owen Mumford, Inc., Woodstock, UK), DISETRONIC™ pen (Disetronic Medical Systems, Burghdorf, Switzerland), HUMALOG MIX 75/25™ pen, HUMALOG™ pen, HUMALIN 70/30™ pen (Eli Lilly and Co., Indianapolis, Ind.), NOVOPEN™ I, II and III (Novo Nordisk, Copenhagen, Denmark), NOVOPEN JUNIOR™ (Novo Nordisk, Copenhagen, Denmark), BD™ pen (Becton Dickinson, Franklin Lakes, N.J.), OPTIPENT™, OPTIPEN PRO™, OPTIPEN STARLET™, and OPTICLIKT™ (Sanofi-Aventis, Frankfurt, Germany), to name only a few. Examples of disposable pen delivery devices having applications in subcutaneous delivery of a pharmaceutical composition of the present invention include, but certainly are not limited to the SOLOSTAR™ pen (Sanofi-Aventis), the FLEXPEN™ (Novo Nordisk), and the KWIKPEN™ (Eli Lilly).


Advantageously, the pharmaceutical compositions for oral or parenteral use described above are prepared into dosage forms in a unit dose suited to fit a dose of the active ingredients. Such dosage forms in a unit dose include, for example, tablets, pills, capsules, injections (ampoules), suppositories, etc. The amount of the aforesaid antibody contained is generally about 5 to about 500 mg per dosage form in a unit dose; especially in the form of injection, the aforesaid antibody may be contained in about 5 to about 100 mg and in about 10 to about 250 mg for the other dosage forms.


The bispecific, nucleic acid, or composition comprising it, may be contained in a medical container such as a phial, syringe, IV container or an injection device. In an example, the bispecific, nucleic acid or composition is in vitro, and may be in a sterile container. In an example, a kit is provided comprising the bispecific, packaging and instructions for use in a therapeutic method as described herein.


One aspect of the invention is a composition comprising a bispecific or nucleic acid of the invention and one or more pharmaceutically acceptable excipients, examples of which are listed above. “Pharmaceutically acceptable” refers to approved or approvable by a regulatory agency of the USA Federal or a state government or listed in the U.S. Pharmacopeia or other generally recognized pharmacopeia for use in animals, including humans. A pharmaceutically acceptable carrier, excipient, or adjuvant can be administered to a patient, together with a bispecific agent, e.g., any antibody or polypeptide molecule described herein, and does not destroy the pharmacological activity thereof and is nontoxic when administered in doses sufficient to deliver a therapeutic amount of the agent.


In some embodiments, the bispecific will be the sole active ingredient in a composition according to the present invention. Thus, a composition may consist of the antibody or it may consist of the bispecific with one or more pharmaceutically acceptable excipients. However, compositions according to the present invention optionally include one or more additional active ingredients. Other therapeutic agents that it may be desirable to administer with bispecific or nucleic acids according to the present invention include analgesic agents. Any such agent or combination of agents may be administered in combination with, or provided in compositions with antibodies or nucleic acids according to the present invention, whether as a combined or separate preparation. The bispecific or nucleic acid according to the present invention may be administered separately and sequentially, or concurrently and optionally as a combined preparation, with another therapeutic agent or agents such as those mentioned.


Multiple compositions can be administered separately or simultaneously. Separate administration refers to the two compositions being administered at different times, e.g. at least 10, 20, 30, or 10-60 minutes apart, or 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 12 hours apart. One can also administer compositions at 24 hours apart, or even longer apart. Alternatively, two or more compositions can be administered simultaneously, e.g. less than 10 or less than 5 minutes apart. Compositions administered simultaneously can, in some aspects, be administered as a mixture, with or without similar or different time release mechanism for each of the components.


Bispecifics, and their encoding nucleic acids, can be used as therapeutic agents. Patients herein are generally mammals, typically humans. A bispecific or nucleic acid may be administered to a mammal, e.g., by any route of administration mentioned herein.


Administration is normally in a “therapeutically effective amount”, this being an amount that produces the desired effect for which it is administered, sufficient to show benefit to a patient. The exact amount will depend on the purpose of the treatment, and will be ascertainable by one skilled in the art using known techniques (see, for example, Lloyd (1999) The Art, Science and Technology of Pharmaceutical Compounding). Prescription of treatment, e.g. decisions on dosage etc, is within the responsibility of general practitioners and other medical doctors and may depend on the severity of the symptoms and/or progression of a disease being treated. A therapeutically effective amount or suitable dose of bispecific or nucleic acid can be determined by comparing its in vitro activity and in vivo activity in an animal model. Methods for extrapolation of effective dosages in mice and other test animals to humans are known.


Bispecifics may be administered in an amount in one of the following ranges per dose:

    • about 10 μg/kg body weight to about 100 mg/kg body weight,
    • about 50 μg/kg body weight to about 5 mg/kg body weight,
    • about 100 μg/kg body weight to about 10 mg/kg body weight,
    • about 100 μg/kg body weight to about 20 mg/kg body weight,
    • about 0.5 mg/kg body weight to about 20 mg/kg body weight, or
    • about 5 mg/kg body weight or lower, for example less than 4, less than 3, less than 2, or less than 1 mg/kg of the antibody.


The dose of antigen-binding molecule administered may be up to 1 mg/kg. It may be formulated at lower strength for paediatric populations, for example 30-150 mg/mL. The bispecific molecule may be packaged in smaller quantities for a paediatric population, e.g., it may be provided in phials of 25-75 mg, e.g., 30 or 60 mg.


In methods of treatment described herein, one or more doses may be administered. In some cases, a single dose may be effective to achieve a long-term benefit. Thus, the method may comprise administering a single dose of the bispecific, its encoding nucleic acid, or the composition. Alternatively, multiple doses may be administered, usually sequentially and separated by a period of days, weeks or months. Optionally, the bispecific may be administered to a patient once a month, or less frequently, e.g., every two months or every three months.


As used herein, the terms “treat,” “treatment,” “treating,” or “amelioration” refer to therapeutic treatments, wherein the object is to reverse, alleviate, ameliorate, inhibit, slow down or stop the progression or severity of a condition associated with a disease or disorder. The term “treating” includes reducing or alleviating at least one adverse effect or symptom of a condition, disease or disorder. Treatment is generally “effective” if one or more symptoms or clinical markers are reduced. Alternatively, treatment is “effective” if the progression of a disease is reduced or halted. That is, “treatment” includes not just the improvement of symptoms or markers, but also a cessation of, or at least slowing of, progress or worsening of symptoms compared to what would be expected in the absence of treatment. Beneficial or desired clinical results include, but are not limited to, alleviation of one or more symptom(s), diminishment of extent of disease, stabilised (i.e., not worsening) state of disease, delay or slowing of disease progression, amelioration or palliation of the disease state, remission (whether partial or total), and/or decreased mortality, whether detectable or undetectable. The term “treatment” of a disease also includes providing relief from the symptoms or side-effects of the disease (including palliative treatment). For treatment to be effective a complete cure is not contemplated. The method can in certain aspects include cure as well. In the context of the invention, treatment may be preventative treatment.


Long half-life is a desirable feature in the bispecifics of the present invention. Extended half-life translates to less frequent administration, with fewer injections being required to maintain a therapeutically effective concentration of the molecule in the bloodstream. The in vivo half life of antigen-binding molecules of the present invention in humans may be 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20 or 21 days, or longer. The in vivo half life of antigen-binding molecules in non-human primates such as cynomolgus monkeys may be 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20 or 21 days, or longer.


Antigen-binding molecules may be provided for administration at regular intervals of one week, two weeks, three weeks, four weeks, or one month.


Therapeutic Use


The bispecific antigen-binding molecules of the present invention may be used in a method of treatment of the human or animal body by therapy. Therapeutic indications for the molecules include:

    • use to treat haemophilia A,
    • use to treat hereditary factor VIII deficiency,
    • use to significantly decrease the number of bleeding incidents in haemophilia A patients,
    • use to substitute for factor VIII function,
    • and/or
    • use to promote blood coagulation.


Patients are typically human patients. The patient may be a human diagnosed with haemophilia A or hereditary factor VIII deficiency, or a human who has lower (or absent) factor VIII expression or activity compared with wild type. The patient may be a paediatric patient (e.g., from 2 to less than 18 years of age) or may be an adult. The patient may or may not have inhibitors to factor VIII.


A bispecific molecule of the present invention, or a composition comprising such an bispecific molecule or its encoding nucleic acid, may be used or provided for use in any such method. Use of the bispecific molecule, or of a composition comprising it or its encoding nucleic acid, for the manufacture of a medicament for use in any such method is also envisaged. The method typically comprises administering the antibody or composition to a mammal, e.g., a human patient. Suitable formulations and methods of administration are described elsewhere herein.


There is a presently unmet need for treatment of haemophilia A patients who develop inhibitory alloantibodies to FVIII. Antigen-binding molecules of the present invention are suitable for use in such patients. Accordingly, in some aspects, a patient treated with a bispecific antigen binding molecule according to the present invention may be resistant to treatment with FVIII owing to the presence of inhibitory antibodies in the bloodstream. Resistance to treatment can be manifested in a reduction of efficacy of the therapy. Such resistance may be detected in in vitro assays (e.g. aPTT assay) with a blood plasma sample from the patient, wherein the therapeutic molecule does not reduce coagulation time to the same level as in an assay with control FVIII-deficient plasma (the latter lacking inhibitory antibodies to the therapeutic molecule).


Patients receiving other treatments for haemophilia, such as bispecific antibodies to FIXa and FX, may also develop inhibitory antibodies to those therapeutic antibodies. As noted, use of human antibodies such as those of the present invention should minimise the risk of this, but inhibitory antibodies may nevertheless be generated in some patients who receive antigen binding molecules of the present invention or other bispecific antigen binding molecules to FIXa and FX. A patient treated with a bispecific antigen binding molecule according to the present invention may be resistant to treatment to a different bispecific antigen binding molecule for FIXa and FX owing to the presence of inhibitory antibodies in the bloodstream. The patient may be resistant to treatment with emicizumab.


Since inhibitory antibodies may be generated through long term therapeutic administration of a drug product, it may be beneficial for patients to alternate or cycle between multiple different treatments, to reduce the risk of their developing inhibitory antibodies. Thus, a bispecific antigen binding molecule of the present invention may be administered to a patient who has previously received treatment with a different FVIIIa-activity replacing polypeptide drug, e.g., a bispecific antigen binding molecule for FIXa and FX, optionally emicizumab, even where the patient has not (yet) developed inhibitory antibodies. Similarly, emicizumab or other bispecific antigen binding molecules for FIXa and FX, and other FVIIIa-activity replacing polypeptide drugs generally, may be administered to patients who were previously treated with a bispecific antigen binding molecule of the present invention. Regiments of treatment may comprise administration of a first FVIII-activity replacing polypeptide drug for a first period (e.g., between one and six months, or between six months and one year), followed by switching to a different FVIII-activity replacing polypeptide drug for a second period (e.g. between one and six months, or between six months and one year), followed by switching back to the first drug or switching to yet another FVIII-activity replacing polypeptide drug. The different amino acid sequences of the different drug treatments should ensure that a patient at risk of developing inhibitory antibodies to one drug is no longer at risk of developing inhibitory antibodies to the first drug (e.g., emicizumab) following switching to a different drug (e.g., a molecule of the present invention). The cycling period may be varied or shortened, according to convenience and the preferences of the patient and doctor.


It will be recognised that administration of the encoding nucleic acid represents an alternative therapy, and may be performed in place of administering the polypeptide drug directly.


As noted, the bispecific antigen-binding molecules of the present invention are believed to have a strong safety profile, associated with no (or minimal) incidents of hypersensitivity reactions, development of alloantibodies, organ toxicity or other adverse events leading to discontinuation of the therapy.


The following numbered clauses and statements represent embodiments of the invention and are part of the description.


CLAUSES

Clause 1. A bispecific antigen-binding molecule comprising






    • a FIXa binding polypeptide arm comprising a FIXa binding site, and

    • a FX binding polypeptide arm comprising a FX binding site, characterised in that

    • (i) the FIXa binding site is provided by a set of complementarity determining regions (CDRs) in the FIXa binding polypeptide arm, the set of CDRs comprising HCDR1, HCDR2, HCDR3 and/or LCDR1, LCDR2 and LCDR3, wherein

    • HCDR1 is SEQ ID NO: 140

    • HCDR2 is SEQ ID NO: 141

    • HCDR3 is SEQ ID NO: 142

    • LCDR1 is SEQ ID NO: 6

    • LCDR2 is SEQ ID NO: 7 and

    • LCDR3 is SEQ ID NO: 8,

    • or comprising that set of CDRs with 1, 2, 3, 4 or 5 amino acid alterations;

    • (ii) the FIXa binding polypeptide arm comprises an antibody Fv region comprising
      • a VH domain generated through recombination of immunoglobulin heavy chain v, d and j gene segments, wherein the v gene segment is VH3-7 and/or wherein the j gene segment is JH6, and/or
      • a VL domain generated through recombination of immunoglobulin light chain v and j gene segments, wherein the v gene segment is VL3-21 and the j gene segment is JL3; and/or

    • (iii) the FIXa binding polypeptide arm is capable, when provided in monospecific form, of enhancing FIXa-catalysed activation of FX to FXa.


      Clause 2. A bispecific antigen-binding molecule according to clause 1, wherein the FIXa binding polypeptide arm comprises a VH domain generated through recombination of immunoglobulin heavy chain v, d and j gene segments, wherein the v gene segment is VH3-7, the d gene segment DH1-26 and the j gene segment is JH6.


      Clause 3. A bispecific antigen-binding molecule according to clause 1 or clause 2, wherein the FIXa binding polypeptide arm comprises a VH domain generated through recombination of immunoglobulin heavy chain v, d and j gene segments, wherein the v gene segment is VH3-7*01 and the j gene segment is JH6*02.


      Clause 4. A bispecific antigen-binding molecule according to any preceding clause, wherein the FIXa binding polypeptide arm comprises an antibody Fv region comprising a FIXa binding site provided by a set of CDRs, the set of CDRs comprising HCDR1, HCDR2, HCDR3 and/or LCDR1, LCDR2 and LCDR3, wherein

    • HCDR1 is SEQ ID NO: 140

    • HCDR2 is SEQ ID NO: 141

    • HCDR3 is SEQ ID NO: 142

    • LCDR1 is SEQ ID NO: 6

    • LCDR2 is SEQ ID NO: 7 and

    • LCDR3 is SEQ ID NO: 8,

    • or comprising that set of CDRs with 1, 2, 3, 4 or 5 amino acid alterations.


      Clause 5. A bispecific antigen-binding molecule according to any preceding clause, wherein the FIXa binding polypeptide arm comprises an antibody VH domain comprising HCDR1, HCDR2 and HCDR3, wherein

    • HCDR1 is SEQ ID NO: 140

    • HCDR2 is SEQ ID NO: 141 and

    • HCDR3 is SEQ ID NO: 142.


      Clause 6. A bispecific antigen-binding molecule according to any preceding clause, wherein HCDR1 is SEQ ID NO: 1, HCDR2 is SEQ ID NO: 2 and HCDR3 is SEQ ID NO: 3.


      Clause 7. A bispecific antigen-binding molecule according to any preceding clause, wherein the FIXa binding polypeptide arm comprises a VL domain generated through recombination of VL3-21*d01 gene segment and a JL3*02 gene segment.


      Clause 8. A bispecific antigen-binding molecule according to any preceding clause, wherein the FIXa binding polypeptide arm comprises an antibody VL domain comprising LCDR1, LCDR2 and LCDR3, wherein

    • LCDR1 is SEQ ID NO: 6

    • LCDR2 is SEQ ID NO: 7 and

    • LCDR3 is SEQ ID NO: 8.


      Clause 9. A bispecific antigen-binding molecule according to any preceding clause, wherein the FIXa binding polypeptide arm comprises a VH domain comprising HCDR1, HCDR2 and HCDR3 in a framework, the framework comprising a set of framework regions FR1, FR2, FR3 and FR4, wherein:

    • FR1 is SEQ ID NO: 132

    • FR2 is SEQ ID NO: 133

    • FR3 is SEQ ID NO: 134 and

    • FR4 is SEQ ID NO: 135,

    • or wherein the framework comprises that set of framework regions with up to 10 amino acid alterations.


      Clause 10. A bispecific antigen-binding molecule according to clause 9, wherein

    • FR1 is SEQ ID NO: 132, optionally with a substitution of L for F at residue 11, a substitution of V for A at residue 24 and/or a substitution of A for V at residue 24

    • FR2 is SEQ ID NO: 133

    • FR3 is SEQ ID NO: 134, optionally with a substitution of Y for F at residue 59, a substitution of D for A at residue 62, a substitution of I for M at residue 70, a substitution of N for K at residue 77, a substitution of L for V at residue 79 and/or a substitution of L for V at residue 81, and

    • FR4 is SEQ ID NO: 135, optionally with a substitution of S for Tat residue 119,

    • wherein residue numbering is according to the IMGT system.


      Clause 11. A bispecific antigen-binding molecule according to clause 10, wherein:

    • FR1 is SEQ ID NO: 132

    • FR2 is SEQ ID NO: 133

    • FR3 is SEQ ID NO: 134 and

    • FR4 is SEQ ID NO: 135.


      Clause 12. A bispecific antigen-binding molecule according to any preceding clause, wherein the FIXa binding polypeptide arm comprises a VL domain comprising LCDR1, LCDR2 and LCDR3 in a framework, the framework comprising a set of framework regions FR1, FR2, FR3 and FR4, wherein:

    • FR1 is SEQ ID NO: 136

    • FR2 is SEQ ID NO: 137

    • FR3 is SEQ ID NO: 138 and

    • FR4 is SEQ ID NO: 139,

    • or wherein the framework comprises that set of framework regions with up to 10 amino acid alterations.


      Clause 13. A bispecific antigen-binding molecule according to clause 12, wherein:

    • FR1 is SEQ ID NO: 136

    • FR2 is SEQ ID NO: 137

    • FR3 is SEQ ID NO: 138 and

    • FR4 is SEQ ID NO: 139.


      Clause 14. A bispecific antigen-binding molecule according to any preceding clause, wherein the FIXa binding polypeptide arm comprises a VH domain having amino acid sequence SEQ ID NO: 5.


      Clause 15. A bispecific antigen-binding molecule according to any preceding clause, wherein the FIXa binding polypeptide arm VL domain comprises amino acid sequence SEQ ID NO: 10.


      Clause 16. A bispecific antigen-binding molecule according to any preceding clause, wherein the FIXa binding polypeptide arm comprises an antibody heavy chain comprising, from N to C terminus, a VH domain, a CH1 domain, a CH2 domain and a CH3 domain.


      Clause 17. A bispecific antigen-binding molecule according to clause 16, wherein the antibody heavy chain comprises an IgG constant region.


      Clause 18. A bispecific antigen-binding molecule according to any preceding clause, wherein the FIXa binding polypeptide arm comprises an antibody light chain comprising, from N to C terminus, a VL domain and a CL domain.


      Clause 19. A bispecific antigen-binding molecule according to any preceding clause, wherein the FX binding polypeptide arm comprises an antibody Fv region comprising a FIXa binding site provided by a set of CDRs.


      Clause 20. A bispecific antigen-binding molecule according to clause 19, wherein the antibody Fv region comprises a VH domain generated through recombination of immunoglobulin heavy chain v, d and j gene segments, wherein the v and j gene segments are:

    • VH1-3 (e.g., VH1-3*01) and JH6 (e.g., JH6*02),

    • VH3-30 (e.g., VH3-30*18) and JH6 (e.g., JH6*02),

    • VH3-33 (e.g., VH3-33*01) and JH6 (e.g., JH6*02),

    • VH4-31 (e.g., VH4-31*03) and JH4 (e.g., JH4*02), or

    • VH4-59 (e.g., VH4-59*01) and JH4 (e.g., JH4*02).


      Clause 21. A bispecific antigen-binding molecule according to clause 19 or clause 20, wherein the antibody Fv region comprises a VL domain generated through recombination of immunoglobulin light chain v and j gene segments, wherein the v gene segment is VL3-21 (e.g., VL3-21*d01) and the j gene segment is JL3 (e.g., JL3*02).


      Clause 22. A bispecific antigen-binding molecule according to any preceding clause, wherein the FIXa binding polypeptide arm and the FX binding polypeptide arm comprise a common antibody light chain.


      Clause 23. A bispecific antigen-binding molecule according to clause 22, wherein the light chain comprises a lambda constant region.


      Clause 24. A bispecific antigen-binding molecule according to any preceding clause, wherein the antigen-binding molecule is a tetrameric immunoglobulin comprising

    • a first heavy-light chain pair comprising a FIXa binding Fv region,

    • a second heavy-light chain pair comprising a FX binding Fv region,

    • wherein each heavy chain comprises a VH domain and a constant region, and each light chain comprises a VL domain and a constant region, and wherein the first and second heavy-light chain pairs associate to form the tetrameric immunoglobulin through heterodimerisation of their heavy chain constant regions.


      Clause 25. A bispecific antigen-binding molecule according to clause 24, wherein the immunoglobulin is an IgG, e.g., IgG4.


      Clause 26. An isolated FIXa binding polypeptide comprising a FIXa binding site, wherein the FIXa binding polypeptide is as defined in any of the preceding clauses.


      Clause 27. A kit for production of the bispecific antigen-binding molecule of any of clauses 1 to 25, comprising

    • a FIXa binding polypeptide comprising a FIXa binding site, or nucleic acid encoding said polypeptide, wherein the FIXa binding polypeptide is a FIXa binding polypeptide arm as defined in any of clauses 1 to 25, and

    • a FX binding polypeptide comprising a FX binding site, or nucleic acid encoding said polypeptide, wherein the FX binding polypeptide is a FX binding polypeptide arm as defined defined in any of clauses 1 to 25.


      Clause 28. A composition comprising an antigen-binding molecule according to any of clauses 1 to 25 in combination with a pharmaceutically acceptable excipient.


      Clause 29. A composition according to clause 28, wherein the antigen-binding molecule is in sterile aqueous solution.


      Clause 30. Isolated nucleic acid encoding a bispecific antigen-binding molecule according to any of clauses 1 to 25.


      Clause 31. Isolated nucleic acid encoding the FIXa binding polypeptide arm of a bispecific antigen-binding molecule according to any of clauses 1 to 25.


      Clause 32. A host cell comprising recombinant nucleic acid encoding a bispecific antigen-binding molecule according to any of clauses 1 to 25, or encoding the FIXa binding polypeptide arm or the FX binding polypeptide arm of said bispecific antigen-binding molecule, wherein the encoding nucleic acid is operably linked to a promoter for expression.


      Clause 33. A method of producing an antigen-binding molecule according to any of clauses 1 to 25, or producing the FIXa binding polypeptide arm or the FX binding polypeptide arm of said bispecific antigen-binding molecule, comprising culturing host cells according to clause 32 under conditions for expression of the antigen-binding molecule, and recovering the antigen-binding molecule or binding arm from the host cell culture.


      Clause 34. A method of controlling bleeding in a patient, comprising administering a composition according to clause 29 or clause 30 to the patient.


      Clause 35. A bispecific antigen-binding molecule according to any of clauses 1 to 25 for use in a method of treatment of the human or animal body by therapy.


      Clause 36. A bispecific antigen-binding molecule according to any of clauses 1 to 25 for use in controlling bleeding in a patient.


      Clause 37. A method according to clause 34, or a bispecific antigen-binding molecule for use according to clause 35 or clause 36, wherein the patient is a haemophilia A patient.


      Clause 38. A method or a bispecific antigen-binding molecule for use according to clause 37, wherein the patient is resistant to treatment with FVIII owing to the presence of inhibitory antibodies in the bloodstream.


      Clause 39. Use of a bispecific antigen-binding molecule according to any of clauses 1 to 25 for binding FIXa and FX in vitro.


      Clause 40. Use of a bispecific antigen-binding molecule according to any of clauses 1 to 25 to substitute for FVIII function in a patient with haemophilia A.





STATEMENTS

1. A bispecific antigen-binding molecule comprising






    • a FIXa binding polypeptide arm comprising a FIXa binding site, and

    • a FX binding polypeptide arm comprising a FX binding site,

    • characterised in that the FIXa binding polypeptide arm comprises a VH domain having at least 90% amino acid sequence identity with SEQ ID NO: 324 and a VL domain having at least 90% amino acid sequence identity with SEQ ID NO: 10, optionally wherein the VH domain comprises an HCDR3 having a hydrophobic or positively charged residue at IMGT position 111.1, optionally wherein the HCDR3 amino acid sequence is SEQ ID NO: 400, SEQ ID NO: 401, SEQ ID NO: 402, SEQ ID NO: 403 or SEQ ID NO: 171; and/or

    • characterised in that the FIXa binding site is provided by a set of complementarity determining regions (CDRs) in the FIXa binding polypeptide arm, the set of CDRs comprising HCDR1, HCDR2, HCDR3 and/or LCDR1, LCDR2 and LCDR3, wherein

    • HCDR1 is SEQ ID NO: 1

    • HCDR2 is SEQ ID NO: 2

    • HCDR3 is SEQ ID NO: 400

    • LCDR1 is SEQ ID NO: 6

    • LCDR2 is SEQ ID NO: 7 and

    • LCDR3 is SEQ ID NO: 8.


      2. A bispecific antigen-binding molecule according to statement 1, wherein the FIXa binding site is provided by a set of complementarity determining regions (CDRs) in the FIXa binding polypeptide arm, the set of CDRs comprising HCDR1, HCDR2, HCDR3 and/or LCDR1, LCDR2 and LCDR3, wherein

    • HCDR1 is SEQ ID NO: 1

    • HCDR2 is SEQ ID NO: 2

    • HCDR3 is SEQ ID NO: 401, SEQ ID NO: 402, SEQ ID NO: 403 or SEQ ID NO: 171

    • LCDR1 is SEQ ID NO: 6

    • LCDR2 is SEQ ID NO: 7 and

    • LCDR3 is SEQ ID NO: 8.


      3. A bispecific antigen-binding molecule comprising

    • (i) a FIXa binding polypeptide arm comprising a FIXa binding site, wherein the FIXa binding polypeptide arm comprises an antibody Fv region comprising
      • a VH domain generated through recombination of immunoglobulin heavy chain v, d and j gene segments, wherein the v gene segment is VH3-7 (e.g., VH3-7*01) and/or wherein the j gene segment is JH6 (e.g., JH6*02), and
      • a VL domain generated through recombination of immunoglobulin light chain v and j gene segments, wherein the v gene segment is VL3-21 (e.g., VL3-21*d01) and the j gene segment is JL2 (e.g., JL2*01) or JL3, and

    • (ii) a FX binding polypeptide arm comprising a FX binding site, wherein the FX binding polypeptide arm comprises an antibody Fv region comprising
      • (a) a VH domain generated through recombination of immunoglobulin heavy chain v, d and j gene segments, wherein the v and j gene segments are VH1-3 (e.g., VH1-3*01) and JH6 (e.g., JH6*02), and
      • a VL domain generated through recombination of immunoglobulin light chain v and j gene segments, wherein the v and j gene segments are VL1-47, (e.g., VL1-47*01) and JL1 (e.g., JL1*01); or
      • (b) a VH domain generated through recombination of immunoglobulin heavy chain v, d and j gene segments, wherein the v and j gene segments are VH3-30 (e.g., VH3-30*18) and JH6 (e.g., JH6*02), and
      • a VL domain generated through recombination of immunoglobulin light chain v and j gene segments, wherein the v and j gene segments are VL2-8, (e.g., VL2-8*01) and JL2 (e.g., JL2*01); or
      • (c) a VH domain generated through recombination of immunoglobulin heavy chain v, d and j gene segments, wherein the v and j gene segments are VH4-61 (e.g., VH4-61*01) and JH1 (e.g., JH1*01), and
      • a VL domain generated through recombination of immunoglobulin light chain v and j gene segments, wherein the v and j gene segments are VK3-11, (e.g., VK3-11*01) and JK5 (e.g., JK5*01).


        4. A bispecific antigen-binding molecule according to any preceding statement, wherein the FIXa binding polypeptide arm comprises a VH domain having at least 95% amino acid sequence identity with SEQ ID NO: 324.


        5. A bispecific antigen-binding molecule according to any preceding statement, wherein the FIXa binding polypeptide arm comprises a VL domain having at least 95% amino acid sequence identity with SEQ ID NO: 10.


        6. A bispecific antigen-binding molecule according to any preceding statement, wherein the

    • FIXa binding site is provided by a set of complementarity determining regions (CDRs) in the FIXa binding polypeptide arm, the set of CDRs comprising HCDR1, HCDR2, HCDR3 and/or LCDR1, LCDR2 and LCDR3, wherein
      • HCDR1 is SEQ ID NO: 1
      • HCDR2 is SEQ ID NO: 2
      • HCDR3 is SEQ ID NO: 171
      • LCDR1 is SEQ ID NO: 6
      • LCDR2 is SEQ ID NO: 7 and
      • LCDR3 is SEQ ID NO: 8.


        7. A bispecific antigen-binding molecule according any preceding statement, wherein the FIXa binding polypeptide arm comprises a VH domain amino acid sequence SEQ ID NO: 324 and a VL domain amino acid sequence SEQ ID NO: 10.


        8. A bispecific antigen-binding molecule according to any preceding statement, which reduces the coagulation time of FVIII-deficient human blood plasma to less than 40 seconds in an aPTT assay.


        9. A bispecific antigen-binding molecule according to any preceding statement, wherein the FIXa binding polypeptide arm is capable, when provided in monospecific form, of enhancing FIXa-catalysed activation of FX to FXa.


        10. A bispecific antigen-binding molecule according to any preceding statement, wherein

    • the FX binding polypeptide arm comprises a VH domain having at least 90% amino acid sequence identity with SEQ ID NO: 61 and a VL domain having at least 90% amino acid sequence identity with SEQ ID NO: 66; and/or wherein

    • the FX binding site is provided by a set of complementarity determining regions (CDRs) in the FX binding polypeptide arm, the set of CDRs comprising HCDR1, HCDR2, HCDR3 and/or LCDR1, LCDR2 and LCDR3, wherein

    • HCDR1 is SEQ ID NO: 57

    • HCDR2 is SEQ ID NO: 58

    • HCDR3 is SEQ ID NO: 59

    • LCDR1 is SEQ ID NO: 62

    • LCDR2 is SEQ ID NO: 63 and

    • LCDR3 is SEQ ID NO: 64.


      11. A bispecific antigen-binding molecule according to statement 10, wherein the FX binding polypeptide arm comprises a VH domain amino acid sequence SEQ ID NO: 61 and a VL domain amino acid sequence SEQ ID NO: 66.


      12. A bispecific antigen-binding molecule according to any of statements 1 to 9, wherein

    • the FX binding polypeptide arm comprises a VH domain having at least 90% amino acid sequence identity with SEQ ID NO: 71 and a VL domain having at least 90% amino acid sequence identity with SEQ ID NO: 76; and/or wherein

    • the FX binding site is provided by a set of complementarity determining regions (CDRs) in the FX binding polypeptide arm, the set of CDRs comprising HCDR1, HCDR2, HCDR3 and/or LCDR1, LCDR2 and LCDR3, wherein

    • HCDR1 is SEQ ID NO: 67

    • HCDR2 is SEQ ID NO: 68

    • HCDR3 is SEQ ID NO: 69

    • LCDR1 is SEQ ID NO: 72

    • LCDR2 is SEQ ID NO: 73 and

    • LCDR3 is SEQ ID NO: 74.


      13. A bispecific antigen-binding molecule according to statement 12, wherein the FX binding polypeptide arm comprises a VH domain amino acid sequence SEQ ID NO: 71 and a VL domain amino acid sequence SEQ ID NO: 76.


      14. A bispecific antigen-binding molecule according to any of statements 1 to 9, wherein

    • the FX binding polypeptide arm comprises a VH domain having at least 90% amino acid sequence identity with SEQ ID NO: 100 and a VL domain having at least 90% amino acid sequence identity with SEQ ID NO: 104; and/or

    • wherein the FX binding site is provided by a set of complementarity determining regions (CDRs) in the FX binding polypeptide arm, the set of CDRs comprising HCDR1, HCDR2, HCDR3 and/or LCDR1, LCDR2 and LCDR3, wherein

    • HCDR1 is SEQ ID NO: 96

    • HCDR2 is SEQ ID NO: 97

    • HCDR3 is SEQ ID NO: 98

    • LCDR1 is SEQ ID NO: 101

    • LCDR2 is SEQ ID NO: 92 and

    • LCDR3 is SEQ ID NO: 102.


      15. A bispecific antigen-binding molecule according to statement 14, wherein the FX binding polypeptide arm comprises a VH domain amino acid sequence SEQ ID NO: 100 and a VL domain amino acid sequence SEQ ID NO: 104.


      16. A bispecific antigen-binding molecule according to any preceding statement, wherein the FIXa binding polypeptide arm and/or the FX binding polypeptide arm comprises

    • an antibody heavy chain comprising, from N to C terminus, a VH domain, a CH1 domain, a CH2 domain and a CH3 domain, and

    • an antibody light chain comprising, from N to C terminus, a VL domain and a CL domain.


      17. A bispecific antigen-binding molecule according to statement 16, wherein the antibody heavy chain comprises an IgG constant region.


      18. A bispecific antigen-binding molecule according to any preceding statement, wherein the antigen-binding molecule is a tetrameric immunoglobulin comprising

    • a first heavy-light chain pair comprising a FIXa binding Fv region,

    • a second heavy-light chain pair comprising a FX binding Fv region,

    • wherein each heavy chain comprises a VH domain and a constant region, and each light chain comprises a VL domain and a constant region, and wherein the first and second heavy-light chain pairs associate to form the tetrameric immunoglobulin through heterodimerisation of their heavy chain constant regions.


      19. A bispecific antigen-binding molecule according to statement 18, wherein the immunoglobulin is an IgG, e.g., IgG4.


      20. A bispecific antigen-binding molecule according to statement 19, wherein the IgG comprises the IgG4-PE human heavy chain constant region SEQ ID NO: 143, optionally engineered with one or more amino acid substitutions to promote heterodimerisation.


      21. An isolated FIXa binding polypeptide comprising a FIXa binding site, wherein the FIXa binding polypeptide is as defined in any of the preceding statements.


      22. Isolated nucleic acid encoding the FIXa binding polypeptide arm or the FX binding polypeptide arm of a bispecific antigen-binding molecule according to any of statements 1 to 20.


      23. Isolated nucleic acid encoding a bispecific antigen-binding molecule according to any of statements 1 to 20.


      24. A kit for production of the bispecific antigen-binding molecule of any of statements 1 to 20, comprising

    • a FIXa binding polypeptide comprising a FIXa binding site, or nucleic acid encoding said polypeptide, wherein the FIXa binding polypeptide is a FIXa binding polypeptide arm as defined in any of statements 1 to 20, and

    • a FX binding polypeptide comprising a FX binding site, or nucleic acid encoding said polypeptide, wherein the FX binding polypeptide is a FX binding polypeptide arm as defined defined in any of statements 1 to 20.


      25. A composition comprising an antigen-binding molecule according to any of statements 1 to 20, or isolated nucleic acid according to statement 23, in combination with a pharmaceutically acceptable excipient.


      26. A composition according to statement 25, wherein the antigen-binding molecule or nucleic acid is in sterile aqueous solution.


      27. A host cell comprising recombinant nucleic acid encoding a bispecific antigen-binding molecule according to any of statements 1 to 20, or encoding the FIXa binding polypeptide arm and/or the FX binding polypeptide arm of said bispecific antigen-binding molecule, wherein the encoding nucleic acid is operably linked to a promoter for expression.


      28. A method of producing an antigen-binding molecule according to any of statements 1 to 20, or producing the FIXa binding polypeptide arm or the FX binding polypeptide arm of said bispecific antigen-binding molecule, comprising culturing host cells according to statement 27 under conditions for expression of the antigen-binding molecule, and recovering the antigen-binding molecule or binding arm from the host cell culture.


      29. A method of controlling bleeding in a patient, comprising administering a composition according to statement 25 or statement 26 to the patient.


      30. A bispecific antigen-binding molecule according to any of statements 1 to 20 or a composition according to statement 25 or statement 26, for use in a method of treatment of the human or animal body by therapy.


      31. A bispecific antigen-binding molecule according to any of statements 1 to 20 of a composition according to statement 25 or statement 26, for use in controlling bleeding in a patient.


      33. A method according to statement 29, or a bispecific antigen-binding molecule for use according to statement 30 or statement 31, wherein the patient is a haemophilia A patient.


      34. A method or a bispecific antigen-binding molecule for use according to statement 33, wherein the patient is resistant to treatment with FVIII owing to the presence of inhibitory antibodies in the bloodstream.


      35. A method or a bispecific antigen-binding molecule for use according to statement 33 or statement 34, wherein the patient is resistant to treatment with another bispecific antigen-binding molecule for FIXa and FX owing to the presence of inhibitory antibodies in the bloodstream.


      36. A method or a bispecific antigen-binding molecule for use according to statement 35, wherein the patient is resistant to treatment with emicizumab.


      37. A method of reducing development of inhibitory anti-drug antibodies in a haemophilia A patient undergoing treatment with a polypeptide that replaces FVIIIa activity, comprising

    • administering a first FVIIIa-activity replacing polypeptide drug to the patient for a period of 1-12 months,

    • switching the patient to a second, different FVIIIa-activity replacing polypeptide drug for a period of 1-12 months, and

    • switching the patient to either the first antigen-binding molecule or to a third, different FVIIIa-activity replacing polypeptide drug for a period of 1-12 months,

    • wherein in each case the FVIIIa-activity replacing polypeptide drug or its encoding nucleic acid is administered in a therapeutically effective amount to functionally replace FVIIIa in the patient, and wherein the risk of the patient developing inhibitory anti-drug antibodies to any of the FVIIIa-activity replacing polypeptide drug is reduced compared with a patient continuing to receive treatment with that FVIIIa-activity replacing polypeptide drug.


      38. A composition comprising a FVIIIa-activity replacing polypeptide drug or its encoding nucleic acid, for use in a method of treating a haemophilia A patient while reducing development of inhibitory anti-drug antibodies, the method comprising

    • administering a first FVIIIa-activity replacing polypeptide drug to the patient for a period of 1-12 months,

    • switching the patient to a second, different FVIIIa-activity replacing polypeptide drug for a period of 1-12 months, and

    • switching the patient to either the first antigen-binding molecule or to a third, different FVIIIa-activity replacing polypeptide drug for a period of 1-12 months,

    • wherein in each case the FVIIIa-activity replacing polypeptide drug or its encoding nucleic acid is administered in a therapeutically effective amount to functionally replace FVIIIa in the patient, and wherein the risk of the patient developing inhibitory anti-drug antibodies to any of the FVIIIa-activity replacing polypeptide drug is reduced compared with a patient continuing to receive treatment with that FVIIIa-activity replacing polypeptide drug.


      39. A method according to statement 37 or a composition for use according to statement 38, wherein the first, second and/or third FVIIIa-activity replacing polypeptide drug is a bispecific antigen-binding molecule for FIXa and FX.


      40. A method or a composition for use according to any of statements 37 to 39, wherein the first, second and/or third FVIIIa-activity replacing polypeptide drug is emicizumab.


      41. A method or a composition for use according to any of statements 37 to 40, wherein the first, second and/or third FVIIIa-activity replacing polypeptide drug is a bispecific antigen-binding molecule according to any of statements 1 to 20.


      42. A method or a composition for use according to any of statements 37 to 41, wherein the first, second or third FVIIIa-activity replacing polypeptide drug is recombinant FVIII or plasma-derived FVIII.


      43. Use of a bispecific antigen-binding molecule according to any of statements 1 to 20 for binding FIXa and FX in vitro.


      44. Use of a bispecific antigen-binding molecule according to any of statements 1 to 20 to substitute for FVIII function in a patient with haemophilia A.





Various further aspects and embodiments of the present invention will be apparent to those skilled in the art in view of the present disclosure. All documents mentioned in this specification, including published US counterparts of any patents or patent applications referred to, are incorporated herein by reference in their entirety.


EXAMPLES

The following Examples describe the generation, characterisation and performance of anti-FIXa antibodies, anti-FX antibodies, and bispecific antibodies generated from combination of FIXa binding polypeptide arms and FX binding polypeptide arms of the anti-FIXa and anti-FX antibodies respectively. Antibodies were generated using the Kymouse™, a transgenic mouse platform capable of generating antibodies with human variable domains. Antibodies from the Kymouse have human variable domains, generated from human v (d) and j segments, and mouse constant domains. The endogenous mouse variable genes have been silenced and make up a very small portion of the repertoire (less than 0.5% of all heavy chain variable regions are of mouse origin). The Kymouse system is described in Lee et al 2014 [11], WO2011/004192, WO2011/158009 and WO2013/061098. This project employed the Kymouse HK strain in which the heavy chain locus and light chain kappa locus are humanised, and the Kymouse HL strain in which the heavy chain locus and the light chain lambda locus are humanised. The mice have a full repertoire of human v, d and j heavy chain gene segments and a full repertoire of human v and j kappa or lambda light chain gene segments.


Example 1. Production of Antibodies to Factor IXa (FIXa)

Four Kymouse HK mice (male, aged 4 months at the initiation of immunisation) and four Kymouse HL mice (male, aged 3 months at the initiation of immunisation) were immunised against human Factor IXa (Enzyme Research Laboratories, Inc.).


Spleen tissues were harvested from the immunised mice, and spleen cells were suspended and sorted by FACS to isolate antigen-specific B cells. A total of 2,460 factor IXa specific B cells were sorted from the 8 immunised animals. Coupled antibody heavy and light chain variable domain sequences were recovered from the B cells by reverse transcription of RNA and PCR amplification of the variable regions.


Antibody-encoding nucleic acid was transfected into the human cell line Expi293F for expression and supernatants were harvested.


Example 2. HTRF® (Homogeneous Time-Resolved Fluorescence) Screen of Factor IXa Antibodies

An HTRF assay was used to screen the anti-FIXa antibodies for binding to FIXa. FIXa and antibody were labelled with two different fluorophores, a donor and an acceptor. When two entities come close enough to each other, excitation of the donor by an energy source triggers an energy transfer towards the acceptor, which in turn emits specific fluorescence at a given wavelength. Therefore the binding of FIXa by its specific antibodies can be detected by detection at the emission wavelength of the acceptor.


Serial dilutions of an anti-human factor IX reference antibody AbN and CM7 isotype control antibody were prepared using Expi293 expression media (Invitrogen). 5 μL of supernatants from each of the 2,460 antibody-secreting human cells generated in Example 1 were transferred to assay plates. 5 μL of AbN and CM7 isotype control antibody were transferred to each assay plate as positive and negative controls respectively. 5 μL (80 nM) of Alexa 647-labelled human factor IX, factor IXa or factor IX light chain was added to each well of the assay plates and incubated at room temperature (RT) for 1 hour. 10 μL of 2×M24-K solution (1:2000 dilution from stock solution) was added to each well of assay plates, and incubated at RT in the dark for 2 hours. Assay plates were read using HTRF 100 flashes protocol of Envision (Excitation wavelength: 340 nm; Emission wavelength 1: 620 nm; Emission wavelength 2: 665 nm). Data were analysed and normalized to the AbN positive control.


Of the 2,460 antibodies originating from the anti-FIXa specific B cells, 732 were identified as binders to factor IX, factor IXa and/or the factor IX light chain in this assay.


Example 3. SPR Analysis of Factor IX Antibodies

Anti-mouse IgG chip was prepared according to manufacturer's instruction.


Supernatants from Example 1 were diluted 1:5 with HBS-EP running buffer (diluted from 20×HBS-EP+ Buffer, pH 7.6 (Bioquote)). Antibodies were captured on the anti-mouse IgG surface. Varying concentrations (256 nM and 1024 nM) of human factor IX, human factor IXa, or human factor IX light chain were used as analyte. The surface was regenerated with 10 mM glycine (pH 1.7). Binding sensorgrams were double referenced with a buffer injection (0 nM). Data was fitted to the 1:1 model inherent to the ProteOn analysis software. Assays were performed at 25° C. and HBS-EP buffer was used as running buffer.


Example 4. Production of Antibody to Factor X (FX)

Four Kymouse HK version 2 mice (male, aged 3 months at the initiation of immunisation) and four Kymouse HL version 2 mice (male, aged 3 months at the initiation of immunisation) were immunised against human Factor X (Enzyme Research Laboratories, Inc.). Spleen tissues were harvested from the immunised mice, and spleen cells were suspended and sorted by FACS to isolate antigen-specific B cells. A total of 1,722 factor X specific B cells were sorted from the 8 immunised animals. Coupled antibody heavy and light chain variable domain sequences were recovered from the B cells by reverse transcription of RNA and PCR amplification of the variable regions.


Antibody-encoding nucleic acid was transfected into the human cell line Expi293F for expression and supernatants were harvested.


Example 5. HTRF Screen of Factor X Antibodies

Serial dilutions of a reference anti-human Factor X antibody, AbT and CM7 isotype control antibody were prepared using Expi293 expression media (Invitrogen). 5 μL of supernatants from each of the 1,722 antibody-secreting human cells generated in Example 4 were transferred to assay plates. 5 μL of AbT and CM7 isotype control antibody were transferred to each assay plates as positive and negative controls respectively. 5 μL (80 nM) of Alexa 647-labelled human factor X, factor Xa or factor X light chain was added to each well of the assay plates, and incubated at RT for 1 hour. 10 μL of 2×M24-K solution (1:2000 dilution from stock solution) was added to each well and incubated at RT in the dark for 2 hours. Assay plates were read using HTRF 100 flashes protocol of Envision (Excitation wavelength: 340 nm; Emission wavelength 1: 620 nm; Emission wavelength 2: 665 nm). Data was analysed and normalized to the AbT positive control.


Of the 1,722 antibodies originating from the anti-FX specific B cells, 497 were identified as binders to factor X, factor Xa and/or the factor X light chain in this assay.


Example 6. SPR Analysis of Factor X Antibodies

Anti-mouse IgG chip was prepared according to manufacturer's instruction. Supernatants from Example 4 were diluted 1: 5 with HBS-EP running buffer (diluted from 20×HBS-EP+ Buffer, pH 7.6 (Bioquote)). Antibodies were captured on anti-mouse IgG surface. Varying concentrations (256 nM and 1024 nM) of human factor X, human factor Xa, or human Factor X light chain were used as analyte. The surface was regenerated with 10 mM Glycine (pH 1.7). Binding sensorgrams were double referenced with a buffer injection (0 nM). Data was fitted to the 1:1 model inherent to the ProteOn analysis software. Assays were performed at 25° C. and HBS-EP buffer was used as running buffer.


Example 7. Construction of Bispecific Antibody Expression Vectors

Plasmids for expression of bispecific human IgG antibodies were constructed as follows. DNA fragments encoding antibody variable regions were prepared from PCR products generated as described in Example 1 and Example 4. To prepare antibody variable region DNA fragments for cloning, after performing PCR, the reaction solution was subjected to 0.8% agarose gel electrophoresis. Amplified fragments of the desired size (about 400 bp) were purified using QIAquick Gel Extraction Kit (QIAGEN) by the method described in the attached instruction manual and eluted using 30 mL of elution buffer (EB).


Amino acid substitution products in the CH3 region of IgG4 were prepared with reference to the knobs-into-holes technique of IgG1 [12] to form heterogeneous molecules of each H chain. Type a (IgG4ra) is a substitution product of Y349C and T366W, and type b (IgG4yb) is a substitution product of E356C, T366S, L368A and Y407V. Furthermore, the substitutions (-ppcpScp- and -ppcpPcp-) were introduced in the hinge region of both types of substitution products. According to the present technique, almost all of the H chains may form heterodimers.


For expression of the FIXa arm of the bispecific antibody, VH domains were cloned into plasmid vector pTT5_Cam_ccdB_hIgG4ra to provide DNA encoding the VH domain in a full length human IgG4ra antibody heavy chain, VL kappa domains were cloned into pTT5_Cam_ccdB_hIgK to provide DNA encoding the VL domain in a full length human kappa antibody light chain, and VL lambda domains were cloned into pTT5_Cam_ccdB to provide DNA encoding the VL domain in a full length human lambda antibody light chain.


H and L chain variable region (VH and VL) DNA fragments were digested with AarI (Invitrogen), and purified using QIAquick PCR Purification Kit (QIAGEN) according to the manufacturer's instructions. pTT5_Cam_ccdB_hIgG4ra (for VH), pTT5_Cam_ccdB_hlgK (for VL from κ clones) or pTT5_Cam_ccdB (for VL from λ clones) was digested with restriction AarI whose cleavage site is in the multicloning site. After digestion, the vectors were purified using QIAquick PCR Purification Kit (QIAGEN) according to the manufacturer's instructions.


The AarI-digested VH or VL fragments and pTT5_Cam_ccdB_hIgG4ra (for VH), pTT5_Cam_ccdB_hIgK (for VL from κ clones) or pTT5_Cam_ccdB (for VL from λ clones) which had been digested with AarI were ligated using T4 ligase (New England Biolabs) according to the manufacturer's instructions. E. coli DH10B strain (ElectroMax DH10B (Invitrogen)) was transformed with the ligation solution. Respective plasmid DNAs were isolated from the obtained ampicillin resistant clones using QIAprep Spin Miniprep Kit (QIAGEN). The resulting respective ampicillin resistant transformants were confirmed to have the insertion of the desired VH and VL by Sanger sequencing.


Plasmid DNAs for anti-FIX binding arms and anti-FX binding arms were isolated from the desired clones using QIAprep Spin Miniprep Kit (QIAGEN) according to the manufacturer's instructions and dissolved in 100 mL of elution buffer (EB) initially. Plasmid DNA solutions were quantified by Nano-drop (Thermo Scientific) and normalized to 50 ng/mL. Anti-FIXa antibody H chain expression vector, anti-FIXa antibody L chain expression vector, anti-FX antibody H chain expression vector, and anti-FX antibody L chain expression vector were dubbed as Nn-IgG4ra, Nn-IgL (or IgK), Tn-IgG4rb, and Tn-IgL (or IgK). DNA minipreps were confirmed to have the insertion of the desired VH or VL by Sanger sequencing. The respective plasmid solutions were preserved at 4° C. until use.


Example 8. Expression of Bispecific Antibodies

8-1. Preparation of DNA Solutions


Mixed solutions containing four kinds of plasmid DNA were prepared for transfection of HEK cells. For 1 mL of cell culture, 250 ng each of Nn-IgG4ra, Nn-IgL (or IgK), Tn-IgG4rb, and Tn-IgL (or IgK) were used.


8-2. Transfection of Host Cells


One day before transfection, cultured Expi293F cells (HEK cell line) were counted for cell seeding calculation. Expi293F cells were pelleted at 300 rpm for 10 minutes. Cells were resuspended in pre-warmed fresh Expi293 expression media to give a final dilution of 2.4×106 cells/ml. 200 ml of cell suspension was incubated overnight in a Kuhner Shaking Incubator (37° C., 140 rpm, 8% CO2).


On the day of transfection, cultured Expi293F cells were counted and diluted to 4×106 cells/ml using fresh Expi293 expression media. 500 μl of cell suspension was aliquoted into each well of 96-well deep well plates using Multidrop Combi. After dispensing, the plates were covered with Duetz sandwich covers and incubated in a Kuhner Shaking Incubator (37° C., 300 rpm, 50 mm orbital throw, humidity 80%) for 2-2.5 hours.


For each transfection, two mixtures were prepared.


Mix 1: 25 μl B cell bridge product (the 4 plasmid mixture)+55 μl RSM+100 ng HyperPBase per well


Mix 2: 1 μl ExpiFectamine™ 293+79 μl RSM


Mix 2 was added to Mix 1 and incubated at room temperature for 15 minutes. The incubated mixture was then added to the 500 μl of cell culture solution and incubated in a Kuhner Shaking Incubator (37° C., 300 rpm, 50 mm orbital throw) for a week.


Example 9. Activation Coagulation Factor VIII (FVIIIa)-Like Activity Assay

The FVIIIa-mimetic activity of a bispecific antibody, i.e., its ability to enhance the FIXa-mediated activation of FX, was assessed in vitro by enzymatic assay. In this assay, the test bispecific molecule is contacted with FIXa and FX in the presence of phospholipid, under conditions suitable for formation of FXa. A substrate for FXa is added which, when cleaved by FXa, generates a detectable product. Detection of this product in the presence of test bispecific antibody is compared with a negative control in which no test antibody is present (a control antibody may be included). The detected signal is quantified by recording absorbance of the reaction solution at 405 nm. Absorbance is measured across a range of antibody concentrations in the assay and an EC50 value calculated as a measure of the bispecific antibody potency in this assay. Significant difference of EC50 between test antibody and control indicates that the test antibody is able to enhance FIXa-mediated activation of FX. See FIG. 7.


Materials & Methods

All reactions were performed at 37° C. unless otherwise stated.


7.5 μL FIX (3.75 μg/mL) and 5 μL supernatant from the Expi293 cells producing the recombinant antibodies (Example 8) were added to each well of an assay plate and incubated at room temperature for 1 hour. A mixture of 2.5 μL FXIa (10 ng/mL), 5 μL FX (50 ng/mL), 0.05 μL phospholipid (10 mg/mL) and 5 μL TBSB-S buffer was added to each well to initiate enzymatic reaction (FIXa cleavage of FX to generate FXa), and incubated at 37° C. for 1 hour. After 60 minutes, the reaction was terminated by adding 5 μL of 0.5 M EDTA. After adding 10 μL S2765 substrate solution to each well, absorbance at 405 nm (reference wavelength 655 nm) was measured for 30 minutes (one reading per 10 minutes).


TBSB:


Tris buffered saline containing 0.1% bovine serum albumin


To make 7.5 mL TBSB:


0.1 mL 7.5% BSA solution (Sigma)


7.4 mL 1×TBS solution (diluted from 20×TBS solution ThermoFisher)


TBSB-S:


TBSB containing 5 mM CaCl2) and 1 mM MgCl2


To make 100 mL TBSB-S:


99.4 mL TBSB


0.5 mL 1M CaCl2) (Sigma)


0.1 mL 1M MgCl2 (Sigma)


FXIa STOCK solution (10 μg/mL):


Add 10 mL TBSB-S to 0.1 mg FXIa (Enzyme Research Laboratories) to make 10 μg/mL stock solution.


Dilute to 10 ng/mL (1:1,000) working solution before use.


FIX Stock Solution (37.5 μg/mL):


Add 13.3 mL TBSB-S to 0.5 mg FIX (Enzyme Research Laboratories) to make 37.5 μg/mL stock solution.


Dilute to 3.75 μg/mL (1:10) working solution before use.


FX Working Solution (50 μg/mL):


Add 16 mL TBSB-S to 0.8 mg FX (Enzyme Research Laboratories) to make 50 μg/mL working solution.


No further dilution is needed before use.


S2765 Stock Solution:


25 mg S2765 (Chromogenix) chromogenic substrate (0.035 mmol)


To make 2 mM stock solution:


Add 17.493 mL water to the vial and dissolve with shaking.


Pefafluor FXa Stock Solution:


10 μmol Pefafluor FXa fluorogenic substrate (0.010 mmol)


To make 1.5 mM stock solution:


Add 6.667 mL water to the vial and dissolve by shaking.


Polybrene Solution:


To make 0.6 g/L hexadimethrine bromide stock solution:


Add 0.15 g hexadimethrine bromide (Sigma) to 250 mL water.


Dilute to 0.6 mg/L (1:1,000) working solution before use.


S2765 Substrate Working Solution


A 1:1 mixture of 2 mM S-2765 stock solution and 0.6 mg/L polybrene solution.


Example 10. Plasma Coagulation Assay

To determine the ability of the bispecific antibodies of the present invention to correct the coagulation ability of the blood of haemophilia A patients, the effect of these antibodies on the activated partial thromboplastin time (aPTT) using FVIII deficient plasma was examined.


A mixture of 50 mL of bispecific antibody solution having a variety of concentrations, 50 mL of FVIII deficient plasma (Biomerieux), and 50 mL of aPTT reagent (Dade Behring) was warmed at 37° C. for 3 minutes. The coagulation reaction was initiated by adding 50 mL of 20 mM CaCl2) (Dade Behring) to the mixture. The time period until coagulation was measured. Apparatus used for this was KC10A (Amelung) connected to CR-A (Amelung).


Concentration dependency was subsequently determined for bispecific antibodies that exhibited the highest coagulation time-reducing effect.


Example 11: Summary of Initial Screening

Bispecific antibodies containing the N128H FIX-binding arm were dramatically more active in the FXase assay than the majority of other bispecifics, and the N128H FIX-binding arm showed an ability to form active bispecifics with a range of FX-binding arms. FIG. 8.


Example 12: Generation of N128H Mutants

Anti-FIX antibody “N128”, having heavy and light chain variable domains designated N128H and N128L respectively, was selected for further assessment and optimisation. The N128H CDR3 (SEQ ID NO: 3) contains a series of four serine residues, at IMGT residue positions 110, 111, 111.1 and 112.1 respectively. Each Ser was individually mutated to all other naturally occurring amino acids, generating mutations in the N128H HCDR3 as summarised in FIG. 9.


This work produced an array of bispecific antibodies containing variants of the N128 FIXa-binding arm, providing a basis to research the further potential of the FIXa-binding arm for:

    • 1) bringing factor IXa to the ideal proximity of Factor X;
    • 2) orientating factor IXa in a way to maximize the catalytical activity of Factor IXa.


As reported below, bispecific antibodies, each containing a FIXa binding arm having a single point mutation in the HCDR3 compared with N128, were expressed in mammalian cells, purified by Protein A (Example 13) and functionally characterised (Example 14). Selected bispecific antibodies exhibiting better activities than the bispecific antibody with N128H arm were then further analysed to confirm their binding properties and biological activity (Examples 15 to 18).


Example 13: Expression and Purification of Antibodies for Functional Assays

The following procedure was used for preparation of antibodies (monospecific and bispecific) for use in the experiments described in the subsequent Examples.


Expression:


Antibodies were transiently expressed in human (Expi293) cells, then purified from cell culture supernatants.


The day before transfection, Expi293 cells were counted by an automated cell counter (Eve cell counter) seeded in pre-warmed expression media at the density of ˜1.7×106 cells/ml and incubated overnight in an orbital shaker incubator (37° C., 8% CO2, 140 rpm).


On the day of transfection, cells were counted and adjusted the cell number to 2.5×106 cells/ml. 2000 μl of cell suspension were dispensed into 24 Deep Well Plates and placed into Kuhner shaking incubator (37° C., 8% CO2, 225 rpm).


DNA solutions were prepared. 2 μg of DNA plasmid mixture diluted in ultrapure water was used for each transfection of Expi293 cells.


In the case of bispecific antibodies, mixed solutions containing four kinds of plasmid DNA were prepared. For 2 mL of cell culture, 500 ng each of Nn-IgG4ra, Nn-IgL (or IgK), Tn-IgG4γb, and Tn-IgL (or IgK) were used. In the case of monospecific antibodies, for 2 mL of cell culture, 1 μg each of Nn-IgG4PE and Nn-IgL (or IgK) or Tn-IgG4PE and Tn-IgL (or IgK) were used.


DNA encoding monospecific or bispecific antibodies was transfected by the same procedure. Two mixtures were prepared for transfection:


Mix 1: 2 μg of plasmid DNA diluted in 40 μl of Opti-MEM® I medium.


Mix 2: 80 μl of ExpiFectamine™ 293 transfection reagent+40 μl of OptiMEM I medium.


Mix 2 was combined with Mix 1 and incubated for 20-30 minutes at room temperature. After the incubation was complete 165 μl of DNA-ExpiFectamine™ 293 Reagent complex was dispensed to each well of the 24 deep well plate. Cells were incubated in the Kuhner shaking incubator (37° C., 8% CO2, 225 rpm) for 6 days. Cell culture supernatants were harvested 6 days after transfection.


Purification:


A 96-well plate purification was employed to purify antibodies (bispecific or monospecific) from Expi293F cell culture supernatants. This method allows rapid small-scale affinity antibody purification of multiple samples in antibody screening experiments. To achieve high % of recovery MabSelect Sure LX was used which is a protein A affinity with very dynamic binding activity (60 mg human IgG/ml medium), an extended residence time, alkali tolerant and low ligand leakage.


Procedure






    • 1. MabSelect Sure LX resin (GE Healthcare) was equilibrated in 1×PBS (Gibco) to remove the storage buffer, to a final concentration of 10% slurry.

    • 2. 600 μl of 10% slurry is added to each well of the 96 well plate AcroPrep™ Advance (Pall)

    • 3. The resin was centrifuged 70×rcf for 1 min at 4 C.

    • 4. The resin was washed with 300 μl 1×PBS, spin 70×rcf for 1 min. This step was repeated twice.

    • 5. 2 ml of cell culture supernatants (pH 7-8) were loaded onto the MabSelect Sure LX resin into the 96 well purification plate and centrifuged at 70-100×rcf for 1 minute.

    • 6. Plate was washed using 600 μl of 1×PBS and centrifuged at 70-100×rcf for 1 minute. This step is performed four times in total.

    • 7. 70 μl of elution buffer (IgG Elute Pierce) was added to each well and incubated for 1 min.

    • 8. The plate was centrifuged at 70-100×rcf for 1 minute to collect the eluate. This step is performed twice in total.





Example 14: Functional Analysis of N128H CDR3 Mutants

The functional activity the bispecific antibodies newly generated according to Example 12 was determined using the in vitro chromogenic factor X activation (FXase) assay and the plasma coagulation assay (as described previously). To standardise comparison of the variants, all FIXa-binding arms (variant VH paired with N128 VL) were heterodimerised with the same FX-binding arm.


From the mutagenesis analysis of the CDRH3, it was noted that the 3rd serine residue in the series of 4 serines in the N128H CDR3 appears to substantially contribute to the FVIII-mimetic activity. Strikingly, several sequence variations at this position greatly improved the FVIII-mimetic activity of the bispecific antibody compared with the original N128 FIX binding arm (FIG. 10). In particular, variants N436H, N438H and N445H, in which the 3rd serine of the N128H CDR3 had been replaced by isoleucine, leucine and valine respectively (FIG. 9), showed a dramatic increase (approximately 2-3 fold) in FXase activity. Significant increase in FXase activity was also observed with N435H, N437H, N442H, N443H and N446H variants, in which the 3rd serine was mutated into histidine, lysine, glutamine, arginine and tryptophan respectively. Mutagenesis analysis also revealed that substitution of specific amino acids in the HCDR3 at this position decreased the FXase activity of the bispecific antibody compared with the original anti-FIX arm N128H. The tenase activity of variants N430H (Cys), N441H (Pro), N447H (Tyr), again indicating the significance of the residue at this position.


Substitution of certain other amino acids for the serine at other positions in the (Ser)4 motif also reduced activity compared with the original anti-FIX arm, most notably in variants N459H (4th Ser to Pro), N461H (4th Ser to Arg), N465H (4th Ser to Tyr), N470H (1st Ser to Phe), N478H (1st Ser to Pro), N480H (1st Ser to Arg), N495H (2nd Ser to Asn).


In FIG. 11 the tenase activities (as absorbance at 405 nm) of N128H variants is plotted against the clotting time (aPTT in seconds). The FVIII-mimetic activity of the N128H variants is compared against bispecific antibody AbE and an ISTC. The isotype control (negative control, ISTC) showed no tenase activity and poor ability to form the clot as indicated by high aPTT values. N436H, N438H and N445H variants exhibited strong ability to generate FXa (high OD405 values) and to quickly result in clotting in FVIII-immunodepleted plasma (low aPTT values—approximately 30 seconds).


Taking into account these functional data, consensus sequences can be generated for the residues at the sequential IMGT positions 110, 111, 111.1 and 112.1 in HCDR3 (see Table 9A). Considering IMGT position 111.1 (corresponding to the third Ser in N128H), a hydrophobic residue (e.g., Ile, Leu, Val, Trp) or positively charged residue (e.g., Arg, Gln, Lys) was associated with strong functional activity. Other residues which could be substituted at this position without loss of activity (and with potentially improved activity) included His, Glu, Asn and Met.


FIXa binding arms of bispecific antibodies showing the highest activity in the above screens were selected for further characterisation as reported in the following Examples.


Example 15. SPR Analysis of Purified Anti-FIX Antibodies

SPR was used to determine the binding affinity (KD) of the FIXa binding arm to FIX, the kinetic constants on-rate (kon) and off-rate (koff). The analysis was performed using a Biacore 8K (GE Healthcare) system.


An anti-human IgG Fc antibody was immobilised on CM4 chip (GE Healthcare catalog number BR100534) according to manufacturer's instructions. Amine coupling kit (BioRad) was used to activate the surface of the chip. The surface was subsequently blocked with 1M ethanolamine. The immobilisation run was performed at 25° C. using HBS-EP (10 mM HEPES pH 7.4, 150 mM NaCl, 3 mM EDTA, 0.05% Polysorbate 20 pH 7.6) as immobilisation running buffer.


Purified monospecific antibodies (referred to as ligand) from Example 13 were captured on the anti-human IgG Fc CM4 surface at approximately 2 μg/ml. The ligands were injected for 60 sec at 10 μl/min in all the active channels of all 8 flow channels. The run was performed at 25° C. using neutral pH (pH 7.4) HBS-P 1× (0.01 M HEPES pH 7.4, 0.15 M NaCl, 0.05% Polysorbate 20 pH 7.6)+CaCl2) 2.5 mM as running buffer. Human factor IX (FIX) (Enzyme Research Laboratories cat n. HFIX 47064) was reconstituted at 1 mg/ml (MW ˜55 KDa) in the running buffer and used as analyte. Factor IX was injected in multiple cycle kinetics (MCK) mode at 3 concentrations (1.5 μM, 500 μM and 166.7 nM) with 120 seconds association phase and 200 seconds dissociation phase, at flow rate 30 μl/sec in both active and reference channels. Three injections of 10 mM glycine pH 1.5 for 60 sec at 10 μl/min were used for the regeneration phase.


An isotype control (ISTC) antibody hIgG4PE was captured at 1 μg/ml for 60 sec at 10 μl/min in the reference channel. hIgG4PE ISTC and hIgG1 ISTC were also captured in the active channel as a negative control. The anti-FIX monospecific antibody AbN was included for comparison.


The data were reference and buffer subtracted and fitted into Langmuir 1:1 model. The first 30 seconds of dissociation were evaluated in the model.


Results:


The anti-FIX antibodies analysed showed binding to FIX with the affinity shown in Table 1 and fast association (kon) and dissociation (koff) rate for FIX. No binding to FIX was observed with ISTC. The anti-FIX antibodies analysed showed approximately 10-fold higher affinity for FIX compared with AbN.









TABLE 1







Binding affinity and kinetic constants on-rate (kon) and off-rate (koff) of anti-FIX


antibodies. The association and dissociation data of the interaction were fitted using


biomolecular reaction model (1:1 Langmuir model). The values for association rate constant


(kon), dissociation rate constant (koff) and dissociation constant (KD) were calculated from the


binding data by BIAevaluation software.










Captured anti-FIX antibody
kon (1/Ms)
koff (1/s)
KD (M)





N128H_IgG4PE + N128L_IgL (n = 2)
2.92 × 105
5.76 × 10−2
1.98 × 10−7


N192H_IgG4PE + N128L_IgL
5.46 × 104
4.70 × 10−2
8.62 × 10−7


N203H_IgG4PE + N128L_IgL
1.02 × 105
4.60 × 10−2
4.52 × 10−7


N205H_IgG4PE + N128L_IgL
9.50 × 104
3.97 × 10−2
4.18 × 10−7


N211H_IgG4PE + N128L_IgL
8.86 × 104
4.03 × 10−2
4.54 × 10−7


N212H_IgG4PE + N128L_IgL
9.89 × 104
4.02 × 10−2
4.07 × 10−7


N215H_IgG4PE + N128L_IgL
7.00 × 104
4.68 × 10−2
6.68 × 10−7


N216H_IgG4PE + N128L_IgL
9.09 × 104
4.28 × 10−2
4.71 × 10−7


N217H_IgG4PE + N128L_IgL
8.36 × 104
3.25 × 10−2
3.89 × 10−7


N218H_IgG4PE + N128L_IgL
8.02 × 104
3.24 × 10−2
4.04 × 10−7


N219H_IgG4PE + N128L_IgL
7.94 × 104
3.49 × 10−2
4.40 × 10−7


N220H_IgG4PE + N128L_IgL
7.89 × 104
3.36 × 10−2
4.26 × 10−7


N221H_IgG4PE + N128L_IgL
6.67 × 104
3.45 × 10−2
5.17 × 10−7


N222H_IgG4PE + N128L_IgL
8.54 × 104
3.14 × 10−2
3.68 × 10−7


N223H_IgG4PE + N128L_IgL
7.30 × 104
3.10 × 10−2
4.25 × 10−7


N224H_IgG4PE + N128L_IgL
7.80 × 104
3.36 × 10−2
4.31 × 10−7


N225H_IgG4PE + N128L_IgL
7.23 × 104
3.10 × 10−2
4.28 × 10−7


N226H_IgG4PE + N128L_IgL
5.90 × 104
2.94 × 10−2
4.98 × 10−7


N227H_IgG4PE + N128L_IgL
7.54 × 104
2.68 × 10−2
3.56 × 10−7


N228H_IgG4PE + N128L_IgL
6.05 × 104
2.94 × 10−2
4.86 × 10−7


N229H_IgG4PE + N128L_IgL
6.61 × 104
2.89 × 10−2
4.37 × 10−7


N436H_IgG4PE + N128L_IgL
2.46 × 105
4.53 × 10−2
1.84 × 10−7


N438H_IgG4PE + N128L_IgL
2.30 × 105
6.73 × 10−2
2.93 × 10−7


N440H_IgG4PE + N128L_IgL
1.85 × 105
5.35 × 10−2
2.89 × 10−7


N442H_IgG4PE + N128L_IgL
1.94 × 105
4.71 × 10−2
2.42 × 10−7


N444H_IgG4PE + N128L_IgL
2.16 × 105
4.45 × 10−2
2.06 × 10−7


N445H_IgG4PE + N128L_IgL
2.04 × 105
5.44 × 10−2
2.67 × 10−7


N456H_IgG4PE + N128L_IgL
1.51 × 105
3.96 × 10−2
2.63 × 10−7


N460H_IgG4PE + N128L_IgL
1.75 × 105
3.18 × 10−2
1.81 × 10−7


AbN
3.06 × 104
4.26 × 10−2
1.39 × 10−6


ISTC


NB





″NB″ = no binding observed.






As this assay used FIX, not FIXa, the kinetic data here refer to the affinity of the binding arm's interaction with FIX. In view of the close structural similarity between FIX and FIXa, the affinity of the antibodies for binding FIXa may be similar to their affinity for binding FIX.


Example 16. SPR Analysis of Purified Anti-FX Antibodies

SPR was used to determine the binding affinity (KD) of the FX binding arm to FX, the kinetic constants on-rate (kon) and off-rate (koff). The analysis was performed using a Biacore 8K (GE Healthcare) system.


An anti-human IgG Fc antibody was immobilised on CM5 chip (GE Healthcare catalog number 29104988) according to manufacturer's instructions. Amine coupling kit (BioRad) was used to activate the surface of the chip. The surface was subsequently blocked with 1M ethanolamine. The immobilisation run was performed at 25° C. using HBS-EP (10 mM HEPES pH 7.4, 150 mM NaCl, 3 mM EDTA, 0.05% Polysorbate 20 pH 7.6) as immobilisation running buffer.


Purified monospecific antibodies (referred to as ligand) from Example 13 were captured on the anti-human IgG Fc CM5 surface at approximately 1 μg/ml. The ligands were injected for 60 seconds at 10 μl/min in all the 8 active channels of all 8 flow cells. The run was performed at 25° C. using neutral pH (pH 7.4) HBS-P 1× (0.01 M HEPES pH 7.4, 0.15 M NaCl, 0.05% Polysorbate 20 pH 7.6)+CaCl2) 2.5 mM as running buffer.


Human Factor X (FX) (Enzyme Research Laboratories cat n. HFX1010) was reconstituted at 1 mg/ml (MW ˜58 KDa) and used as analyte. Factor X was injected in multiple cycle kinetics (MCK) mode at 3 concentrations (1.5 μM, 375 nM, 93.75 nM) with 120 seconds association phase and 300 seconds dissociation phase at the flow rate 30 μl/sec in both active and reference channels. Three injections of 10 mM glycine pH 1.5 for 60 sec. at 10 ul/min were used for the regeneration phase.


hIgG4PE ISTC was captured in the active channel as a negative control. The anti-FX monospecific antibody AbT was included for comparison.


The data were reference and buffer subtracted and fitted into Langmuir 1:1 model. The first 30 seconds of dissociation were evaluated in the model.


Results:


The anti-FX antibodies analysed showed binding to FX with the affinity shown in Table 2 and fast association (kon) and dissociation (koff) rate for FX. No binding to FX was observed with ISTC.


The anti-FX antibodies analysed showed between approximately 10-fold to 1000-fold higher binding affinity to FX compared with AbT. Antibody T14 (VH domain T14H, VL domain T14L) had an affinity of approximately 0.18 μM for FX. Two other anti-FX antibody VH domains, T19H and T20H, which were identified from the immunised mice with sequences similar to T14, were paired with the T14 VL domain for comparison. These bound FX with a KD of approximately 0.2 and 0.05 μM respectively. The other tested anti-FX antibodies, T15, T23 and T25, had a KD in the nanomolar range.









TABLE 2







Binding affinity and kinetic constants on-rate (kon) and


off-rate (koff) of anti-FX antibodies. The association and dissociation


data of the interaction were fitted using biomolecular reaction


model (1:1 Langmuir model). The values for


association rate constant (kon), dissociation rate constant (koff)


and dissociation constant (KD) were calculated from the


binding data by BIAevaluation software.










Captured anti-FX antibody
kon (1/MS)
koff (1/S)
KD (M)





AbT
1.40 × 104
3.26 × 10−2
2.33 × 10−6


T14H_IgG4PE + T14_VL_IgK
5.62 × 104
1.01 × 10−2
1.79 × 10−7


T19H_IgG4PE + T14_VL_IgK
6.29 × 104
1.21 × 10−2
1.93 × 10−7


T20H_IgG4PE + T14_VL_IgK
9.00 × 104
4.19 × 10−3
4.66 × 10−3


T15H_IgG4PE + T15L_IgK
9.94 × 104
2.94 × 10−4
2.96 × 10−9


T23H_IgG4PE + T23L_IgK
1.06 × 104
6.03 × 10−4
5.70 × 10−9


T25H_IgG4PE + T25L_IgK
7.03 × 104
2.51 × 10−3
3.57 × 10−9


hIgG4PE ISTC


NB





″NB″ = no binding observed.






Example 17: Tenase (FXase) Assay with FIX Binding Arm N436 Paired with a Range of FX Binding Arms

Ability of N436H_IgG4_ra Factor IX antibody arm heterodimerised with different Factor X arms to catalyse Factor IXa activation of Factor X was determined using in vitro tenase (FXase) assay using coagulation factors purified from human plasma.


The FXase assay protocol used for initial screening (see Example 9) was refined for the present experiments, which used bispecific antibody purified by Protein A (rather than cell supernatant). Pre-activated factor IXa was added in this updated assay protocol, rather than factor IX and factor XIa, and FIX was not pre-incubated with the antibody solution before adding the assay mixture. Incubation at 37 deg C. was reduced from 1 hour to 10 minutes.


In brief, the method was as follows, with all reactions being performed at 37° C. unless otherwise stated:


Recombinantly expressed antibodies were purified from supernatant from the recombinant Expi293 cells using Protein A as described in Example 13.


5 μL purified recombinant antibody was added to each well of an assay plate. A mixture of 1.5 μL FIXa (1 μg/ml), 5 μL FX (50 μg/mL), 0.05 μL phospholipid (10 mg/mL) and 13.45 μL TBSB-S buffer was added to each well to initiate enzymatic reaction (FIXa cleavage of FX to generate FXa), and incubated at 37° C. for 10 minutes. After 10 minutes, the reaction was terminated by adding 5 μL of 0.5 M EDTA. After adding 10 μL S2765 substrate solution to each well, absorbance at 405 nm (reference wavelength 655 nm) was measured for 25 minutes (one reading per 5 minutes).


Chromogenix S-2765 is a chromogenic substrate used to determine the generation of activated Factor X. Factor Xa cleaves S2765, releasing the chromophoric group p-nitroaniline (pNA), resulting in a colour change which can be monitored photometrically at 405 nm. The absorbance reading at 405 nm is proportional to the amount of Factor Xa generated. FIG. 7 illustrates the principles of the assay.


Bispecific antibodies tested in this assay had a FIX binding arm comprising the VH domain of N436H and the VL domain of N128L. These were expressed as the heavy and light chain constructs N436H_IgG4_ra and N128L_IgL respectively, and heterodimerised with the following heavy and light chain constructs providing the FX binding arm:

    • T02H_IgG4rb+T02L_IgL;
    • T05H_IgG4rb+T05L_IgL; or
    • T14H_IgG4rb+T14L_IgK.


Also included for reference were a positive control FIX-FX bispecific antibody (Ab_E) and an isotype control (ISTC).


Results are illustrated in FIG. 12.


N436H_IgG4ra, N128L_IgL demonstrated a dose dependent increase in FXase activity as a bispecific antibody heterodimerised with either T02H_IgG4rb+T02L_IgL, T05H_IgG4rb+T05L_IgL or T14H_IgG4rb+T14L_IgK. FX activation observed was comparable to the bispecific antibody Ab_E. N436H_IgG4ra, N128L_IgL expressed alone in the absence of an anti-Factor X arm was still capable of activating Factor X, albeit at a reduced level. The isotype control demonstrated no FXase activity.


Example 18: Plasma Coagulation (aPTT) Assay with FIX Binding Arm N436 Paired with a Range of FX Binding Arms

In this plasma coagulation assay, aPTT measurements are derived using a photo-optical coagulation system. As a clot forms, the amount of light able to pass through the sample decreases. This decrease is plotted as a function of time, generating a waveform. Once a fibrin clot has been formed the end-point of the clotting reaction is complete and the clotting time is automatically calculated. Plasma used in the present aPTT assay is deficient in FVIII, so the effect of adding the FIX-FX bispecific antibody is measurable by its ability to substitute for FVIII in the clotting cascade. Clotting time in the presence of the bispecific antibody can be compared against a negative control and against clotting time in the presence of FVIII.


A mixture of 5 μL bispecific antibody solution having a variety of concentrations, 20 μL FVIII deficient human plasma (Helena Biosciences) and 25 μL Si L Minus aPTT reagent (Helena Biosciences) was warmed at 37° C. for 2 minutes. The coagulation reaction was initiated by adding 25 μL 20 mM CaCl2) (Helena Biosciences) to the mixture. The time period until coagulation was measured. Apparatus used for this was a semi-automated coagulometer, specifically the C-series photo-optical coagulation system (Helena Biosciences). The aPTT reagent contains phospholipid and a contact activator (near-colloidal particle activator, namely magnesium-aluminium-silicate) which provides the “surface contact” activation of the intrinsic blood clotting cascade (see FIG. 1 for reference). Concentration dependency was subsequently determined for bispecific antibodies that exhibited the highest coagulation time-reducing effect.


Results are shown in FIG. 13. In this one-stage aPTT clotting assay using FVIII-depleted human plasma, N436H_IgG4ra, N128L_IgL demonstrated a dose dependent decrease in aPTT time as a bispecific antibody heterodimerised with either T02H_IgG4rb+T02L_IgL, T05H_IgG4rb+T05L_IgL or T14H_IgG4rb+T14L_IgK. The dose dependent decrease in aPTT time was comparable to Ab_E. No reduction in aPTT was observed for the isotype control.


REFERENCES



  • 1 Kitazawa et al., A bispecific antibody to factors IXa and X restores factor VIII hemostatic activity in hemophilia A model, Nat. Med. 18(10):1570-1574 2012

  • 2 Sampei et al., Identification and Multidimensional Optimization of an Asymmetric Bispecific IgG Antibody Mimicking the Function of Factor VIII Cofactor Activity, PLOS ONE 8(2) 2013

  • 3 Uchida et al., A first-in-human phase 1 study of ACE910, a novel factor VIII-mimetic bispecific antibody, in healthy subjects, Blood 127(13):1633-1641 2015

  • 4 Shima et al., Factor VIII-Mimetic Function of Humanized Bispecific Antibody in Hemophilia A, N Engl J Med 374:2044-2053 2016

  • 5 Diagram by Dr Graham Beards, based on information in Pallister C J and Watson M S (2010) Haematology, UK: Scion Publishing, pp. 336-347 ISBN: 1-904842-39-9

  • 6 Fay, Activation of factor VIII and mechanisms of cofactor action, Blood Reviews, 18:1-15 2004

  • 7 Brandstetter et al., X-ray structure of clotting factor IXa: Active site and module structure related to Xase activity and hemophilia B, PNAS 92:9796-9800 1995

  • 8 Bowen, Haemophilia A and haemophilia B: molecular insights, Mol. Pathol. 55:1-18 2002

  • 9 Lefranc M P, IMGT unique numbering for immunoglobulin and T cell receptor variable domains and Ig superfamily V-like domains, Dev Comp Immunol. 27(1):55-77 2003

  • 10 Davis J H et al., PEDS 23:195-202)

  • 11 Lee et al, Nature Biotechnology, 32:6-363, 2014

  • 12 Ridgway et al., Protein Eng. 9:617-621 1996


    FIXa Binding Arm Polypeptide Sequences










TABLE 9A







Anti-FIXa VH domain sequences and CDRs

















VH amino


Ab VH
HCDR1
HCDR2
HCDR3
VH nucleotide sequence
acid sequence





N192H
SEQ ID NO: 11
SEQ ID NO: 12
SEQ ID NO: 13
SEQ ID NO: 14
SEQ ID NO: 15



GFTFSSYW
IKQDGSEK
AREGYSSYYYYGMDV
GAGGTGCAGCTGGTGGAGTCTG
EVQLVESGGGLVQPGGSL






GGGGAGGCTTGGTCCAGCCTGG
RLSCAASGFTFSSYWMSW






GGGGTCCCTGAGACTCTCCTGT
VRQAPGKGLEWVANIKQD






GCAGCCTCTGGATTCACCTTTA
GSEKYYVDSVKGRFTISR






GTAGCTATTGGATGAGCTGGGT
DNAKNSLYLQMNSLRAED






CCGCCAGGCTCCAGGGAAGGGG
TAVYYCAREGYSSYYYYG






CTGGAGTGGGTGGCCAACATAA
MDVWGQGTTVTVSS






AGCAAGATGGAAGTGAGAAATA







CTATGTGGACTCTGTGAAGGGC







CGATTCACCATCTCCAGAGACA







ACGCCAAGAACTCACTGTATCT







GCAAATGAACAGCCTGAGAGCC







GAGGACACGGCTGTGTATTACT







GTGCGAGAGAGGGGTATAGCAG







TTACTACTACTACGGTATGGAC







GTCTGGGGCCAAGGGACCACGG







TCACCGTCTCCTCA






N212H
SEQ ID NO: 11
SEQ ID NO: 2
SEQ ID NO: 3
SEQ ID NO: 16
SEQ ID NO: 17



GFTFSSYW
INQDGSEK
AREGYSSSSYYGMDV
GAGGTGCAGCTGGTGGAGTCTG
EVQLVESGGGLVQPGGSL






GGGGAGGCTTGGTCCAGCCTGG
RLSCAASGFTFSSYWMSW






GGGGTCCCTGAGACTCTCCTGT
VRQAPGKGLEWVANINQD






GCAGCCTCTGGATTCACCTTTA
GSEKYYVDSVKGRFTISR






GTAGCTATTGGATGAGCTGGGT
DNAKNSLYLQMNSLRAED






CCGCCAGGCTCCAGGGAAGGGG
TAVYYCAREGYSSSSYYG






CTGGAGTGGGTGGCCAACATAA
MDVWGQGTTVTVSS






ACCAAGATGGAAGTGAGAAATA







CTATGTGGACTCTGTGAAGGGC







CGATTCACCATCTCCAGAGACA







ACGCCAAGAACTCACTGTATCT







ACAAATGAACAGCCTGAGAGCC







GAGGACACGGCTGTGTATTACT







GTGCGAGAGAGGGGTATAGTAG







TTCGTCCTATTATGGTATGGAC







GTCTGGGGCCAAGGGACCACGG







TCACCGTCTCCTCA






N205H
SEQ ID NO: 18
SEQ ID NO: 2
SEQ ID NO: 3
SEQ ID NO: 19
SEQ ID NO: 20



GFIFSSYW
INQDGSEK
AREGYSSSSYYGMDV
GAGGTGCAGCTGGTGGAGTCTG
EVQLVESGGGLVQPGGSL






GGGGAGGCTTGGTCCAGCCTGG
RLSCVASGFIFSSYWMSW






GGGGTCCCTGAGACTCTCCTGT
VRQAPGKGLEWVANINQD






GTAGCCTCTGGATTCATCTTTA
GSEKYYVDSVKGRFTISR






GTAGCTATTGGATGAGCTGGGT
DNAKNSLYLQMNSLRAED






CCGCCAGGCTCCAGGGAAGGGG
TAVYYCAREGYSSSSYYG






CTGGAGTGGGTGGCCAATATAA
MDVWGQGTTVTVSS






ATCAAGATGGAAGTGAGAAATA







CTATGTGGACTCTGTGAAGGGC







CGATTCACCATCTCCAGAGACA







ACGCCAAGAACTCACTGTATCT







GCAAATGAACAGCCTGAGAGCC







GAGGACACGGCTGTGTATTACT







GTGCGAGAGAGGGGTATAGCAG







TTCGTCCTATTATGGTATGGAC







GTCTGGGGCCAAGGGACCACGG







TCACCGTCTCCTCA






N211H
SEQ ID NO: 1
SEQ ID NO: 2
SEQ ID NO: 3
SEQ ID NO: 21
SEQ ID NO: 22



GFTFNSYW
INQDGSEK
AREGYSSSSYYGMDV
GAGGTGCAGCTGGTGGAGTCTG
EVQLVESGGGLVQPGGSL






GGGGAGGCTTGGTCCAGCCTGG
RLSCAVSGFTFNSYWMSW






GGGGTCCCTGAGACTCTCCTGT
VRQAPGKGLEWVANINQD






GCAGTCTCTGGATTCACCTTTA
GSEKFYVASVKGRFTISR






ATAGCTATTGGATGAGCTGGGT
DNAKNSVYLQMNSLRAED






CCGCCAGGCTCCAGGGAAGGGG
TAVYYCAREGYSSSSYYG






CTGGAGTGGGTGGCCAACATAA
MDVWGQGTTVTVSS






ACCAAGATGGAAGTGAGAAATT







CTATGTGGCCTCTGTGAAGGGC







CGATTCACCATCTCCAGAGACA







ACGCCAAGAACTCAGTGTATCT







ACAAATGAACAGCCTGAGAGCC







GAGGACACGGCTGTGTATTACT







GTGCGAGAGAGGGGTATAGTAG







TTCGTCCTATTATGGTATGGAC







GTCTGGGGCCAAGGGACCACGG







TCACCGTCTCCTCA






N203H
SEQ ID NO: 23
SEQ ID NO: 2
SEQ ID NO: 24
SEQ ID NO: 25
SEQ ID NO: 26



GFTFNNYW
INQDGSEK
AREGYTDSSYYGMDV
GAGGTGCAGCTGGTGGAGTCTG
EVQLVESGGGLVQPGGSL






GGGGAGGCCTGGTCCAGCCTGG
RLSCAVSGFTFNNYWMSW






GGGGTCCCTGAGACTCTCCTGT
VRQAPGKGLEWVANINQD






GCAGTCTCTGGATTCACCTTTA
GSEKFYVASVKGRFIISR






ATAACTATTGGATGAGCTGGGT
DNAKNSVYLQMNSLRAED






CCGCCAGGCTCCAGGGAAGGGG
TAVYYCAREGYTDSSYYG






CTGGAGTGGGTGGCCAACATAA
MDVWGQGTTVSVSS






ACCAAGATGGAAGTGAGAAATT







CTATGTGGCCTCTGTGAAGGGC







CGATTCATCATCTCCAGAGACA







ACGCCAAAAATTCAGTGTATCT







ACAAATGAACAGCCTGAGAGCC







GAGGACACGGCTGTGTATTACT







GTGCGAGAGAGGGGTATACCGA







TTCGTCCTATTATGGAATGGAC







GTCTGGGGCCAAGGGACCACGG







TCTCCGTCTCCTCA






N128H
SEQ ID NO: 1
SEQ ID NO: 2
SEQ ID NO: 3
SEQ ID NO: 4
SEQ ID NO: 5



GFTFNSYW
INQDGSEK
AREGYSSSSYYGMDV
GAGGTGCAGCTGGTGGAGTCTG
EVQLVESGGGFVQPGGSL






GGGGAGGCTTTGTCCAGCCTGG
RLSCAVSGFTFNSYWMSW






GGGGTCCCTGAGACTCTCCTGT
VRQAPGKGLEWVANINQD






GCAGTCTCTGGATTCACCTTTA
GSEKFYVASVKGRFTMSR






ATAGCTATTGGATGAGCTGGGT
DNAKKSVYVQMNSLRAED






CCGCCAGGCTCCAGGGAAGGGG
TAVYYCAREGYSSSSYYG






CTGGAGTGGGTGGCCAACATAA
MDVWGQGTTVTVSS






ACCAAGATGGAAGTGAGAAATT







CTATGTGGCCTCTGTGAAGGGC







CGATTCACCATGTCCAGAGACA







ACGCCAAGAAATCAGTGTATGT







ACAAATGAACAGCCTGAGAGCC







GAGGACACGGCTGTGTATTACT







GTGCGAGAGAGGGGTATAGTAG







TTCGTCCTATTATGGTATGGAC







GTCTGGGGCCAAGGGACCACGG







TCACCGTCTCCTCAG






N215H
SEQ ID NO: 11
SEQ ID NO: 12
SEQ ID NO: 3
SEQ ID NO: 27
SEQ ID NO: 28



GFTFSSYW
IKQDGSEK
AREGYSSSSYYGMDV
GAGGTGCAGCTGGTGGAGTCTG
EVQLVESGGGLVQPGGSL






GGGGAGGCTTGGTCCAGCCTGG
RLSCAASGFTFSSYWMSW






GGGGTCCCTGAGACTCTCCTGT
VRQAPGKGLEWVANIKQD






GCAGCCTCTGGATTCACCTTTA
GSEKYYVDSVKGRFTISR






GTAGCTATTGGATGAGCTGGGT
DNAKNSLYLQMNSLRAED






CCGCCAGGCTCCAGGGAAGGGG
TAVYYCAREGYSSSSYYG






CTGGAGTGGGTGGCCAACATAA
MDVWGQGTTVTVSS






AGCAAGATGGAAGTGAGAAATA







CTATGTGGACTCTGTGAAGGGC







CGATTCACCATCTCCAGAGACA







ACGCCAAGAACTCACTGTATCT







GCAAATGAACAGCCTGAGAGCC







GAGGACACGGCTGTGTATTACT







GTGCGAGAGAGGGGTATAGCAG







TTCGTCCTACTACGGTATGGAC







GTCTGGGGCCAAGGGACCACGG







TCACCGTCTCCTCA






N216H
SEQ ID NO: 1
SEQ ID NO: 2
SEQ ID NO: 3
SEQ ID NO: 29
SEQ ID NO: 30



GFTFNSYW
INQDGSEK
AREGYSSSSYYGMDV
GAGGTGCAGCTGGTGGAGTCTG
EVQLVESGGGFVQPGGSL






GGGGAGGCTTTGTCCAGCCTGG
RLSCAVSGFTFNSYWMSW






GGGGTCCCTGAGACTCTCCTGT
VRQAPGKGLEWVANINQD






GCAGTCTCTGGATTCACCTTTA
GSEKFYVASVKGRFTISR






ATAGCTATTGGATGAGCTGGGT
DNAKNSVYLQMNSLRAED






CCGCCAGGCTCCAGGGAAGGGG
TAVYYCAREGYSSSSYYG






CTGGAGTGGGTGGCCAACATAA
MDVWGQGTTVTVSS






ACCAAGATGGAAGTGAGAAATT







CTATGTGGCCTCTGTGAAGGGC







CGATTCACCATCTCCAGAGACA







ACGCCAAGAACTCAGTGTATCT







ACAAATGAACAGCCTGAGAGCC







GAGGACACGGCTGTGTATTACT







GTGCGAGAGAGGGGTATAGTAG







TTCGTCCTATTATGGTATGGAC







GTCTGGGGCCAAGGGACCACGG







TCACCGTCTCCTCA






N217H
SEQ ID NO: 1
SEQ ID NO: 2
SEQ ID NO: 3
SEQ ID NO: 31
SEQ ID NO: 32



GFTFNSYW
INQDGSEK
AREGYSSSSYYGMDV
GAGGTGCAGCTGGTGGAGTCTG
EVQLVESGGGLVQPGGSL






GGGGAGGCTTGGTCCAGCCTGG
RLSCAVSGFTFNSYWMSW






GGGGTCCCTGAGACTCTCCTGT
VRQAPGKGLEWVANINQD






GCAGTCTCTGGATTCACCTTTA
GSEKFYVASVKGRFTMSR






ATAGCTATTGGATGAGCTGGGT
DNAKNSVYLQMNSLRAED






CCGCCAGGCTCCAGGGAAGGGG
TAVYYCAREGYSSSSYYG






CTGGAGTGGGTGGCCAACATAA
MDVWGQGTTVTVSS






ACCAAGATGGAAGTGAGAAATT







CTATGTGGCCTCTGTGAAGGGC







CGATTCACCATGTCCAGAGACA







ACGCCAAGAACTCAGTGTATCT







ACAAATGAACAGCCTGAGAGCC







GAGGACACGGCTGTGTATTACT







GTGCGAGAGAGGGGTATAGTAG







TTCGTCCTATTATGGTATGGAC







GTCTGGGGCCAAGGGACCACGG







TCACCGTCTCCTCA






N218H
SEQ ID NO: 1
SEQ ID NO: 2
SEQ ID NO: 3
SEQ ID NO: 33
SEQ ID NO: 34



GFTFNSYW
INQDGSEK
AREGYSSSSYYGMDV
GAGGTGCAGCTGGTGGAGTCTG
EVQLVESGGGLVQPGGSL






GGGGAGGCTTGGTCCAGCCTGG
RLSCAVSGFTFNSYWMSW






GGGGTCCCTGAGACTCTCCTGT
VRQAPGKGLEWVANINQD






GCAGTCTCTGGATTCACCTTTA
GSEKFYVASVKGRFTISR






ATAGCTATTGGATGAGCTGGGT
DNAKKSVYLQMNSLRAED






CCGCCAGGCTCCAGGGAAGGGG
TAVYYCAREGYSSSSYYG






CTGGAGTGGGTGGCCAACATAA
MDVWGQGTTVTVSS






ACCAAGATGGAAGTGAGAAATT







CTATGTGGCCTCTGTGAAGGGC







CGATTCACCATCTCCAGAGACA







ACGCCAAGAAATCAGTGTATCT







ACAAATGAACAGCCTGAGAGCC







GAGGACACGGCTGTGTATTACT







GTGCGAGAGAGGGGTATAGTAG







TTCGTCCTATTATGGTATGGAC







GTCTGGGGCCAAGGGACCACGG







TCACCGTCTCCTCA






N219H
SEQ ID NO: 1
SEQ ID NO: 2
SEQ ID NO: 3
SEQ ID NO: 35
SEQ ID NO: 36



GFTFNSYW
INQDGSEK
AREGYSSSSYYGMDV
GAGGTGCAGCTGGTGGAGTCTG
EVQLVESGGGLVQPGGSL






GGGGAGGCTTGGTCCAGCCTGG
RLSCAVSGFTFNSYWMSW






GGGGTCCCTGAGACTCTCCTGT
VRQAPGKGLEWVANINQD






GCAGTCTCTGGATTCACCTTTA
GSEKFYVASVKGRFTISR






ATAGCTATTGGATGAGCTGGGT
DNAKNSVYVQMNSLRAED






CCGCCAGGCTCCAGGGAAGGGG
TAVYYCAREGYSSSSYYG






CTGGAGTGGGTGGCCAACATAA
MDVWGQGTTVTVSS






ACCAAGATGGAAGTGAGAAATT







CTATGTGGCCTCTGTGAAGGGC







CGATTCACCATCTCCAGAGACA







ACGCCAAGAACTCAGTGTATGT







ACAAATGAACAGCCTGAGAGCC







GAGGACACGGCTGTGTATTACT







GTGCGAGAGAGGGGTATAGTAG







TTCGTCCTATTATGGTATGGAC







GTCTGGGGCCAAGGGACCACGG







TCACCGTCTCCTCA






N220H
SEQ ID NO: 1
SEQ ID NO: 2
SEQ ID NO: 3
SEQ ID NO: 37
SEQ ID NO: 38



GFTFNSYW
INQDGSEK
AREGYSSSSYYGMDV
GAGGTGCAGCTGGTGGAGTCTG
EVQLVESGGGFVQPGGSL






GGGGAGGCTTTGTCCAGCCTGG
RLSCAVSGFTFNSYWMSW






GGGGTCCCTGAGACTCTCCTGT
VRQAPGKGLEWVANINQD






GCAGTCTCTGGATTCACCTTTA
GSEKFYVASVKGRFTISR






ATAGCTATTGGATGAGCTGGGT
DNAKKSVYVQMNSLRAED






CCGCCAGGCTCCAGGGAAGGGG
TAVYYCAREGYSSSSYYG






CTGGAGTGGGTGGCCAACATAA
MDVWGQGTTVTVSS






ACCAAGATGGAAGTGAGAAATT







CTATGTGGCCTCTGTGAAGGGC







CGATTCACCATCTCCAGAGACA







ACGCCAAGAAATCAGTGTATGT







ACAAATGAACAGCCTGAGAGCC







GAGGACACGGCTGTGTATTACT







GTGCGAGAGAGGGGTATAGTAG







TTCGTCCTATTATGGTATGGAC







GTCTGGGGCCAAGGGACCACGG







TCACCGTCTCCTCA






N221H
SEQ ID NO: 1
SEQ ID NO: 2
SEQ ID NO: 3
SEQ ID NO: 39
SEQ ID NO: 40



GFTFNSYW
INQDGSEK
AREGYSSSSYYGMDV
GAGGTGCAGCTGGTGGAGTCTG
EVQLVESGGGFVQPGGSL






GGGGAGGCTTTGTCCAGCCTGG
RLSCAVSGFTFNSYWMSW






GGGGTCCCTGAGACTCTCCTGT
VRQAPGKGLEWVANINQD






GCAGTCTCTGGATTCACCTTTA
GSEKFYVASVKGRFTMSR






ATAGCTATTGGATGAGCTGGGT
DNAKNSVYVQMNSLRAED






CCGCCAGGCTCCAGGGAAGGGG
TAVYYCAREGYSSSSYYG






CTGGAGTGGGTGGCCAACATAA
MDVWGQGTTVTVSS






ACCAAGATGGAAGTGAGAAATT







CTATGTGGCCTCTGTGAAGGGC







CGATTCACCATGTCCAGAGACA







ACGCCAAGAACTCAGTGTATGT







ACAAATGAACAGCCTGAGAGCC







GAGGACACGGCTGTGTATTACT







GTGCGAGAGAGGGGTATAGTAG







TTCGTCCTATTATGGTATGGAC







GTCTGGGGCCAAGGGACCACGG







TCACCGTCTCCTCA






N222H
SEQ ID NO: 1
SEQ ID NO: 2
SEQ ID NO: 3
SEQ ID NO: 41
SEQ ID NO: 42



GFTFNSYW
INQDGSEK
AREGYSSSSYYGMDV
GAGGTGCAGCTGGTGGAGTCTG
EVQLVESGGGFVQPGGSL






GGGGAGGCTTTGTCCAGCCTGG
RLSCAVSGFTFNSYWMSW






GGGGTCCCTGAGACTCTCCTGT
VRQAPGKGLEWVANINQD






GCAGTCTCTGGATTCACCTTTA
GSEKFYVASVKGRFTMSR






ATAGCTATTGGATGAGCTGGGT
DNAKKSVYLQMNSLRAED






CCGCCAGGCTCCAGGGAAGGGG
TAVYYCAREGYSSSSYYG






CTGGAGTGGGTGGCCAACATAA
MDVWGQGTTVTVSS






ACCAAGATGGAAGTGAGAAATT







CTATGTGGCCTCTGTGAAGGGC







CGATTCACCATGTCCAGAGACA







ACGCCAAGAAATCAGTGTATCT







ACAAATGAACAGCCTGAGAGCC







GAGGACACGGCTGTGTATTACT







GTGCGAGAGAGGGGTATAGTAG







TTCGTCCTATTATGGTATGGAC







GTCTGGGGCCAAGGGACCACGG







TCACCGTCTCCTCA






N223H
SEQ ID NO: 1
SEQ ID NO: 2
SEQ ID NO: 3
SEQ ID NO: 43
SEQ ID NO: 44



GFTFNSYW
INQDGSEK
AREGYSSSSYYGMDV
GAGGTGCAGCTGGTGGAGTCTG
EVQLVESGGGLVQPGGSL






GGGGAGGCTTGGTCCAGCCTGG
RLSCAVSGFTFNSYWMSW






GGGGTCCCTGAGACTCTCCTGT
VRQAPGKGLEWVANINQD






GCAGTCTCTGGATTCACCTTTA
GSEKFYVASVKGRFTMSR






ATAGCTATTGGATGAGCTGGGT
DNAKKSVYVQMNSLRAED






CCGCCAGGCTCCAGGGAAGGGG
TAVYYCAREGYSSSSYYG






CTGGAGTGGGTGGCCAACATAA
MDVWGQGTTVTVSS






ACCAAGATGGAAGTGAGAAATT







CTATGTGGCCTCTGTGAAGGGC







CGATTCACCATGTCCAGAGACA







ACGCCAAGAAATCAGTGTATGT







ACAAATGAACAGCCTGAGAGCC







GAGGACACGGCTGTGTATTACT







GTGCGAGAGAGGGGTATAGTAG







TTCGTCCTATTATGGTATGGAC







GTCTGGGGCCAAGGGACCACGG







TCACCGTCTCCTCA






N224H
SEQ ID NO: 1
SEQ ID NO: 2
SEQ ID NO: 3
SEQ ID NO: 45
SEQ ID NO: 46



GFTFNSYW
INQDGSEK
AREGYSSSSYYGMDV
GAGGTGCAGCTGGTGGAGTCTG
EVQLVESGGGFVQPGGSL






GGGGAGGCTTTGTCCAGCCTGG
RLSCAVSGFTFNSYWMSW






GGGGTCCCTGAGACTCTCCTGT
VRQAPGKGLEWVANINQD






GCAGTCTCTGGATTCACCTTTA
GSEKFYVASVKGRFTMSR






ATAGCTATTGGATGAGCTGGGT
DNAKNSVYLQMNSLRAED






CCGCCAGGCTCCAGGGAAGGGG
TAVYYCAREGYSSSSYYG






CTGGAGTGGGTGGCCAACATAA
MDVWGQGTTVTVSS






ACCAAGATGGAAGTGAGAAATT







CTATGTGGCCTCTGTGAAGGGC







CGATTCACCATGTCCAGAGACA







ACGCCAAGAACTCAGTGTATCT







ACAAATGAACAGCCTGAGAGCC







GAGGACACGGCTGTGTATTACT







GTGCGAGAGAGGGGTATAGTAG







TTCGTCCTATTATGGTATGGAC







GTCTGGGGCCAAGGGACCACGG







TCACCGTCTCCTCA






N225H
SEQ ID NO: 1
SEQ ID NO: 2
SEQ ID NO: 3
SEQ ID NO: 47
SEQ ID NO: 48



GFTFNSYW
INQDGSEK
AREGYSSSSYYGMDV
GAGGTGCAGCTGGTGGAGTCTG
EVQLVESGGGFVQPGGSL






GGGGAGGCTTTGTCCAGCCTGG
RLSCAVSGFTFNSYWMSW






GGGGTCCCTGAGACTCTCCTGT
VRQAPGKGLEWVANINQD






GCAGTCTCTGGATTCACCTTTA
GSEKFYVASVKGRFTISR






ATAGCTATTGGATGAGCTGGGT
DNAKKSVYLQMNSLRAED






CCGCCAGGCTCCAGGGAAGGGG
TAVYYCAREGYSSSSYYG






CTGGAGTGGGTGGCCAACATAA
MDVWGQGTTVTVSS






ACCAAGATGGAAGTGAGAAATT







CTATGTGGCCTCTGTGAAGGGC







CGATTCACCATCTCCAGAGACA







ACGCCAAGAAATCAGTGTATCT







ACAAATGAACAGCCTGAGAGCC







GAGGACACGGCTGTGTATTACT







GTGCGAGAGAGGGGTATAGTAG







TTCGTCCTATTATGGTATGGAC







GTCTGGGGCCAAGGGACCACGG







TCACCGTCTCCTCA






N226H
SEQ ID NO: 1
SEQ ID NO: 2
SEQ ID NO: 3
SEQ ID NO: 49
SEQ ID NO: 50



GFTFNSYW
INQDGSEK
AREGYSSSSYYGMDV
GAGGTGCAGCTGGTGGAGTCTG
EVQLVESGGGFVQPGGSL






GGGGAGGCTTTGTCCAGCCTGG
RLSCAVSGFTFNSYWMSW






GGGGTCCCTGAGACTCTCCTGT
VRQAPGKGLEWVANINQD






GCAGTCTCTGGATTCACCTTTA
GSEKFYVASVKGRFTISR






ATAGCTATTGGATGAGCTGGGT
DNAKNSVYVQMNSLRAED






CCGCCAGGCTCCAGGGAAGGGG
TAVYYCAREGYSSSSYYG






CTGGAGTGGGTGGCCAACATAA
MDVWGQGTTVTVSS






ACCAAGATGGAAGTGAGAAATT







CTATGTGGCCTCTGTGAAGGGC







CGATTCACCATCTCCAGAGACA







ACGCCAAGAACTCAGTGTATGT







ACAAATGAACAGCCTGAGAGCC







GAGGACACGGCTGTGTATTACT







GTGCGAGAGAGGGGTATAGTAG







TTCGTCCTATTATGGTATGGAC







GTCTGGGGCCAAGGGACCACGG







TCACCGTCTCCTCA






N227H
SEQ ID NO: 1
SEQ ID NO: 2
SEQ ID NO: 3
SEQ ID NO: 51
SEQ ID NO: 52



GFTFNSYW
INQDGSEK
AREGYSSSSYYGMDV
GAGGTGCAGCTGGTGGAGTCTG
EVQLVESGGGLVQPGGSL






GGGGAGGCTTGGTCCAGCCTGG
RLSCAVSGFTFNSYWMSW






GGGGTCCCTGAGACTCTCCTGT
VRQAPGKGLEWVANINQD






GCAGTCTCTGGATTCACCTTTA
GSEKFYVASVKGRFTMSR






ATAGCTATTGGATGAGCTGGGT
DNAKKSVYLQMNSLRAED






CCGCCAGGCTCCAGGGAAGGGG
TAVYYCAREGYSSSSYYG






CTGGAGTGGGTGGCCAACATAA
MDVWGQGTTVTVSS






ACCAAGATGGAAGTGAGAAATT







CTATGTGGCCTCTGTGAAGGGC







CGATTCACCATGTCCAGAGACA







ACGCCAAGAAATCAGTGTATCT







ACAAATGAACAGCCTGAGAGCC







GAGGACACGGCTGTGTATTACT







GTGCGAGAGAGGGGTATAGTAG







TTCGTCCTATTATGGTATGGAC







GTCTGGGGCCAAGGGACCACGG







TCACCGTCTCCTCA






N228H
SEQ ID NO: 1
SEQ ID NO: 2
SEQ ID NO: 3
SEQ ID NO: 53
SEQ ID NO: 54



GFTFNSYW
INQDGSEK
AREGYSSSSYYGMDV
GAGGTGCAGCTGGTGGAGTCTG
EVQLVESGGGLVQPGGSL






GGGGAGGCTTGGTCCAGCCTGG
RLSCAVSGFTFNSYWMSW






GGGGTCCCTGAGACTCTCCTGT
VRQAPGKGLEWVANINQD






GCAGTCTCTGGATTCACCTTTA
GSEKFYVASVKGRFTMSR






ATAGCTATTGGATGAGCTGGGT
DNAKNSVYVQMNSLRAED






CCGCCAGGCTCCAGGGAAGGGG
TAVYYCAREGYSSSSYYG






CTGGAGTGGGTGGCCAACATAA
MDVWGQGTTVTVSS






ACCAAGATGGAAGTGAGAAATT







CTATGTGGCCTCTGTGAAGGGC







CGATTCACCATGTCCAGAGACA







ACGCCAAGAACTCAGTGTATGT







ACAAATGAACAGCCTGAGAGCC







GAGGACACGGCTGTGTATTACT







GTGCGAGAGAGGGGTATAGTAG







TTCGTCCTATTATGGTATGGAC







GTCTGGGGCCAAGGGACCACGG







TCACCGTCTCCTCA






N229H
SEQ ID NO: 1
SEQ ID NO: 2
SEQ ID NO: 3
SEQ ID NO: 55
SEQ ID NO: 56



GFTFNSYW
INQDGSEK
AREGYSSSSYYGMDV
GAGGTGCAGCTGGTGGAGTCTG
EVQLVESGGGLVQPGGSL






GGGGAGGCTTGGTCCAGCCTGG
RLSCAVSGFTFNSYWMSW






GGGGTCCCTGAGACTCTCCTGT
VRQAPGKGLEWVANINQD






GCAGTCTCTGGATTCACCTTTA
GSEKFYVASVKGRFTISR






ATAGCTATTGGATGAGCTGGGT
DNAKKSVYVQMNSLRAED






CCGCCAGGCTCCAGGGAAGGGG
TAVYYCAREGYSSSSYYG






CTGGAGTGGGTGGCCAACATAA
MDVWGQGTTVTVSS






ACCAAGATGGAAGTGAGAAATT







CTATGTGGCCTCTGTGAAGGGC







CGATTCACCATCTCCAGAGACA







ACGCCAAGAAATCAGTGTATGT







ACAAATGAACAGCCTGAGAGCC







GAGGACACGGCTGTGTATTACT







GTGCGAGAGAGGGGTATAGTAG







TTCGTCCTATTATGGTATGGAC







GTCTGGGGCCAAGGGACCACGG







TCACCGTCTCCTCA













SEQ ID NO: 140        SEQ ID NO: 141   SEQ ID NO: 142



HCCDR1 consensus      HCDR2 consensus  HCDR3 consensus



GFTFSSYM              INQDGSEK         AREGYSSSSYYGMDV



I NN                   K                    TDYY



GF(T/I)F(S/N)(S/N)YM  I(N/K)QDGSEK     AREGY(S/T)(S/D)(S/Y)GMDV















N420H
SEQ ID NO: 1
SEQ ID NO: 2
SEQ ID NO: 161
SEQ ID NO: 238
SEQ ID NO: 314





AREGYASSSYYGMDV
GAGGTGCAGCTGGTGGAGTCTG
EVQLVESGGGFVQPGGSL






GGGGAGGCTTTGTCCAGCCTGG
RLSCAVSGFTFNSYWMSW






GGGGTCCCTGAGACTCTCCTGT
VRQAPGKGLEWVANINQD






GCAGTCTCTGGATTCACCTTTA
GSEKFYVASVKGRFTMSR






ATAGCTATTGGATGAGCTGGGT
DNAKKSVYVQMNSLRAED






CCGCCAGGCTCCAGGGAAGGGG
TAVYYCAREGYASSSYYG






CTGGAGTGGGTGGCCAACATAA
MDVWGQGTTVTVSS






ACCAAGATGGAAGTGAGAAATT







CTATGTGGCCTCTGTGAAGGGC







CGATTCACCATGTCCAGAGACA







ACGCCAAGAAATCAGTGTATGT







ACAAATGAACAGCCTGAGAGCC







GAGGACACGGCTGTGTATTACT







GTGCGAGAGAGGGGTATGCCAG







TTCGTCCTATTATGGTATGGAC







GTCTGGGGCCAAGGGACCACGG







TCACCGTCTCCTCAG






N421H
SEQ ID NO: 1
SEQ ID NO: 2
SEQ ID NO: 162
SEQ ID NO: 239
SEQ ID NO: 315





AREGYSASSYYGMDV
GAGGTGCAGCTGGTGGAGTCTG
EVQLVESGGGFVQPGGSL






GGGGAGGCTTTGTCCAGCCTGG
RLSCAVSGFTFNSYWMSW






GGGGTCCCTGAGACTCTCCTGT
VRQAPGKGLEWVANINQD






GCAGTCTCTGGATTCACCTTTA
GSEKFYVASVKGRFTMSR






ATAGCTATTGGATGAGCTGGGT
DNAKKSVYVQMNSLRAED






CCGCCAGGCTCCAGGGAAGGGG
TAVYYCAREGYSASSYYG






CTGGAGTGGGTGGCCAACATAA
MDVWGQGTTVTVSS






ACCAAGATGGAAGTGAGAAATT







CTATGTGGCCTCTGTGAAGGGC







CGATTCACCATGTCCAGAGACA







ACGCCAAGAAATCAGTGTATGT







ACAAATGAACAGCCTGAGAGCC







GAGGACACGGCTGTGTATTACT







GTGCGAGAGAGGGGTATAGTGC







CTCGTCCTATTATGGTATGGAC







GTCTGGGGCCAAGGGACCACGG







TCACCGTCTCCTCAG






N422H
SEQ ID NO: 1
SEQ ID NO: 2
SEQ ID NO: 163
SEQ ID NO: 240
SEQ ID NO: 316





AREGYSSASYYGMDV
GAGGTGCAGCTGGTGGAGTCTG
EVQLVESGGGFVQPGGSL






GGGGAGGCTTTGTCCAGCCTGG
RLSCAVSGFTFNSYWMSW






GGGGTCCCTGAGACTCTCCTGT
VRQAPGKGLEWVANINQD






GCAGTCTCTGGATTCACCTTTA
GSEKFYVASVKGRFTMSR






ATAGCTATTGGATGAGCTGGGT
DNAKKSVYVQMNSLRAED






CCGCCAGGCTCCAGGGAAGGGG
TAVYYCAREGYSSASYYG






CTGGAGTGGGTGGCCAACATAA
MDVWGQGTTVTVSS






ACCAAGATGGAAGTGAGAAATT







CTATGTGGCCTCTGTGAAGGGC







CGATTCACCATGTCCAGAGACA







ACGCCAAGAAATCAGTGTATGT







ACAAATGAACAGCCTGAGAGCC







GAGGACACGGCTGTGTATTACT







GTGCGAGAGAGGGGTATAGTAG







TGCCTCCTATTATGGTATGGAC







GTCTGGGGCCAAGGGACCACGG







TCACCGTCTCCTCAG






N423H
SEQ ID NO: 1
SEQ ID NO: 2
SEQ ID NO: 164
SEQ ID NO: 241
SEQ ID NO: 317





AREGYSSSAYYGMDV
GAGGTGCAGCTGGTGGAGTCTG
EVQLVESGGGFVQPGGSL






GGGGAGGCTTTGTCCAGCCTGG
RLSCAVSGFTFNSYWMSW






GGGGTCCCTGAGACTCTCCTGT
VRQAPGKGLEWVANINQD






GCAGTCTCTGGATTCACCTTTA
GSEKFYVASVKGRFTMSR






ATAGCTATTGGATGAGCTGGGT
DNAKKSVYVQMNSLRAED






CCGCCAGGCTCCAGGGAAGGGG
TAVYYCAREGYSSSAYYG






CTGGAGTGGGTGGCCAACATAA
MDVWGQGTTVTVSS






ACCAAGATGGAAGTGAGAAATT







CTATGTGGCCTCTGTGAAGGGC







CGATTCACCATGTCCAGAGACA







ACGCCAAGAAATCAGTGTATGT







ACAAATGAACAGCCTGAGAGCC







GAGGACACGGCTGTGTATTACT







GTGCGAGAGAGGGGTATAGTAG







TTCGGCCTATTATGGTATGGAC







GTCTGGGGCCAAGGGACCACGG







TCACCGTCTCCTCAG






N430H
SEQ ID NO: 1
SEQ ID NO: 2
SEQ ID NO: 165
SEQ ID NO: 242
SEQ ID NO: 318





AREGYSSCSYYGMDV
GAGGTGCAGCTGGTGGAGTCTG
EVQLVESGGGFVQPGGSL






GGGGAGGCTTTGTCCAGCCTGG
RLSCAVSGFTFNSYWMSW






GGGGTCCCTGAGACTCTCCTGT
VRQAPGKGLEWVANINQD






GCAGTCTCTGGATTCACCTTTA
GSEKFYVASVKGRFTMSR






ATAGCTATTGGATGAGCTGGGT
DNAKKSVYVQMNSLRAED






CCGCCAGGCTCCAGGGAAGGGG
TAVYYCAREGYSSCSYYG






CTGGAGTGGGTGGCCAACATAA
MDVWGQGTTVTVSS






ACCAAGATGGAAGTGAGAAATT







CTATGTGGCCTCTGTGAAGGGC







CGATTCACCATGTCCAGAGACA







ACGCCAAGAAATCAGTGTATGT







ACAAATGAACAGCCTGAGAGCC







GAGGACACGGCTGTGTATTACT







GTGCGAGAGAGGGGTATAGTAG







TTGCTCCTATTATGGTATGGAC







GTCTGGGGCCAAGGGACCACGG







TCACCGTCTCCTCAG






N431H
SEQ ID NO: 1
SEQ ID NO: 2
SEQ ID NO: 166
SEQ ID NO: 243
SEQ ID NO: 319





AREGYSSDSYYGMDV
GAGGTGCAGCTGGTGGAGTCTG
EVQLVESGGGFVQPGGSL






GGGGAGGCTTTGTCCAGCCTGG
RLSCAVSGFTFNSYWMSW






GGGGTCCCTGAGACTCTCCTGT
VRQAPGKGLEWVANINQD






GCAGTCTCTGGATTCACCTTTA
GSEKFYVASVKGRFTMSR






ATAGCTATTGGATGAGCTGGGT
DNAKKSVYVQMNSLRAED






CCGCCAGGCTCCAGGGAAGGGG
TAVYYCAREGYSSDSYYG






CTGGAGTGGGTGGCCAACATAA
MDVWGQGTTVTVSS






ACCAAGATGGAAGTGAGAAATT







CTATGTGGCCTCTGTGAAGGGC







CGATTCACCATGTCCAGAGACA







ACGCCAAGAAATCAGTGTATGT







ACAAATGAACAGCCTGAGAGCC







GAGGACACGGCTGTGTATTACT







GTGCGAGAGAGGGGTATAGTAG







TGACTCCTATTATGGTATGGAC







GTCTGGGGCCAAGGGACCACGG







TCACCGTCTCCTCAG






N432H
SEQ ID NO: 1
SEQ ID NO: 2
SEQ ID NO: 167
SEQ ID NO: 244
SEQ ID NO: 320





AREGYSSESYYGMDV
GAGGTGCAGCTGGTGGAGTCTG
EVQLVESGGGFVQPGGSL






GGGGAGGCTTTGTCCAGCCTGG
RLSCAVSGFTFNSYWMSW






GGGGTCCCTGAGACTCTCCTGT
VRQAPGKGLEWVANINQD






GCAGTCTCTGGATTCACCTTTA
GSEKFYVASVKGRFTMSR






ATAGCTATTGGATGAGCTGGGT
DNAKKSVYVQMNSLRAED






CCGCCAGGCTCCAGGGAAGGGG
TAVYYCAREGYSSESYYG






CTGGAGTGGGTGGCCAACATAA
MDVWGQGTTVTVSS






ACCAAGATGGAAGTGAGAAATT







CTATGTGGCCTCTGTGAAGGGC







CGATTCACCATGTCCAGAGACA







ACGCCAAGAAATCAGTGTATGT







ACAAATGAACAGCCTGAGAGCC







GAGGACACGGCTGTGTATTACT







GTGCGAGAGAGGGGTATAGTAG







TGAGTCCTATTATGGTATGGAC







GTCTGGGGCCAAGGGACCACGG







TCACCGTCTCCTCAG






N433H
SEQ ID NO: 1
SEQ ID NO: 2
SEQ ID NO: 168
SEQ ID NO: 245
SEQ ID NO: 321





AREGYSSFSYYGMDV
GAGGTGCAGCTGGTGGAGTCTG
EVQLVESGGGFVQPGGSL






GGGGAGGCTTTGTCCAGCCTGG
RLSCAVSGFTFNSYWMSW






GGGGTCCCTGAGACTCTCCTGT
VRQAPGKGLEWVANINQD






GCAGTCTCTGGATTCACCTTTA
GSEKFYVASVKGRFTMSR






ATAGCTATTGGATGAGCTGGGT
DNAKKSVYVQMNSLRAED






CCGCCAGGCTCCAGGGAAGGGG
TAVYYCAREGYSSFSYYG






CTGGAGTGGGTGGCCAACATAA
MDVWGQGTTVTVSS






ACCAAGATGGAAGTGAGAAATT







CTATGTGGCCTCTGTGAAGGGC







CGATTCACCATGTCCAGAGACA







ACGCCAAGAAATCAGTGTATGT







ACAAATGAACAGCCTGAGAGCC







GAGGACACGGCTGTGTATTACT







GTGCGAGAGAGGGGTATAGTAG







TTTCTCCTATTATGGTATGGAC







GTCTGGGGCCAAGGGACCACGG







TCACCGTCTCCTCAG






N434H
SEQ ID NO: 1
SEQ ID NO: 2
SEQ ID NO: 169
SEQ ID NO: 246
SEQ ID NO: 322





AREGYSSGSYYGMDV
GAGGTGCAGCTGGTGGAGTCTG
EVQLVESGGGFVQPGGSL






GGGGAGGCTTTGTCCAGCCTGG
RLSCAVSGFTFNSYWMSW






GGGGTCCCTGAGACTCTCCTGT
VRQAPGKGLEWVANINQD






GCAGTCTCTGGATTCACCTTTA
GSEKFYVASVKGRFTMSR






ATAGCTATTGGATGAGCTGGGT
DNAKKSVYVQMNSLRAED






CCGCCAGGCTCCAGGGAAGGGG
TAVYYCAREGYSSGSYYG






CTGGAGTGGGTGGCCAACATAA
MDVWGQGTTVTVSS






ACCAAGATGGAAGTGAGAAATT







CTATGTGGCCTCTGTGAAGGGC







CGATTCACCATGTCCAGAGACA







ACGCCAAGAAATCAGTGTATGT







ACAAATGAACAGCCTGAGAGCC







GAGGACACGGCTGTGTATTACT







GTGCGAGAGAGGGGTATAGTAG







TGGCTCCTATTATGGTATGGAC







GTCTGGGGCCAAGGGACCACGG







TCACCGTCTCCTCAG






N435H
SEQ ID NO: 1
SEQ ID NO: 2
SEQ ID NO: 170
SEQ ID NO: 247
SEQ ID NO: 323





AREGYSSHSYYGMDV
GAGGTGCAGCTGGTGGAGTCTG
EVQLVESGGGFVQPGGSL






GGGGAGGCTTTGTCCAGCCTGG
RLSCAVSGFTFNSYWMSW






GGGGTCCCTGAGACTCTCCTGT
VRQAPGKGLEWVANINQD






GCAGTCTCTGGATTCACCTTTA
GSEKFYVASVKGRFTMSR






ATAGCTATTGGATGAGCTGGGT
DNAKKSVYVQMNSLRAED






CCGCCAGGCTCCAGGGAAGGGG
TAVYYCAREGYSSHSYYG






CTGGAGTGGGTGGCCAACATAA
MDVWGQGTTVTVSS






ACCAAGATGGAAGTGAGAAATT







CTATGTGGCCTCTGTGAAGGGC







CGATTCACCATGTCCAGAGACA







ACGCCAAGAAATCAGTGTATGT







ACAAATGAACAGCCTGAGAGCC







GAGGACACGGCTGTGTATTACT







GTGCGAGAGAGGGGTATAGTAG







TCACTCCTATTATGGTATGGAC







GTCTGGGGCCAAGGGACCACGG







TCACCGTCTCCTCAG






N436H
SEQ ID NO: 1
SEQ ID NO: 2
SEQ ID NO: 171
SEQ ID NO: 248
SEQ ID NO: 324





AREGYSSISYYGMDV
GAGGTGCAGCTGGTGGAGTCTG
EVQLVESGGGFVQPGGSL






GGGGAGGCTTTGTCCAGCCTGG
RLSCAVSGFTFNSYWMSW






GGGGTCCCTGAGACTCTCCTGT
VRQAPGKGLEWVANINQD






GCAGTCTCTGGATTCACCTTTA
GSEKFYVASVKGRFTMSR






ATAGCTATTGGATGAGCTGGGT
DNAKKSVYVQMNSLRAED






CCGCCAGGCTCCAGGGAAGGGG
TAVYYCAREGYSSISYYG






CTGGAGTGGGTGGCCAACATAA
MDVWGQGTTVTVSS






ACCAAGATGGAAGTGAGAAATT







CTATGTGGCCTCTGTGAAGGGC







CGATTCACCATGTCCAGAGACA







ACGCCAAGAAATCAGTGTATGT







ACAAATGAACAGCCTGAGAGCC







GAGGACACGGCTGTGTATTACT







GTGCGAGAGAGGGGTATAGTAG







TATCTCCTATTATGGTATGGAC







GTCTGGGGCCAAGGGACCACGG







TCACCGTCTCCTCAG






N437H
SEQ ID NO: 1
SEQ ID NO: 2
SEQ ID NO: 172
SEQ ID NO: 249
SEQ ID NO: 325





AREGYSSKSYYGMDV
GAGGTGCAGCTGGTGGAGTCTG
EVQLVESGGGFVQPGGSL






GGGGAGGCTTTGTCCAGCCTGG
RLSCAVSGFTFNSYWMSW






GGGGTCCCTGAGACTCTCCTGT
VRQAPGKGLEWVANINQD






GCAGTCTCTGGATTCACCTTTA
GSEKFYVASVKGRFTMSR






ATAGCTATTGGATGAGCTGGGT
DNAKKSVYVQMNSLRAED






CCGCCAGGCTCCAGGGAAGGGG
TAVYYCAREGYSSKSYYG






CTGGAGTGGGTGGCCAACATAA
MDVWGQGTTVTVSS






ACCAAGATGGAAGTGAGAAATT







CTATGTGGCCTCTGTGAAGGGC







CGATTCACCATGTCCAGAGACA







ACGCCAAGAAATCAGTGTATGT







ACAAATGAACAGCCTGAGAGCC







GAGGACACGGCTGTGTATTACT







GTGCGAGAGAGGGGTATAGTAG







TAAGTCCTATTATGGTATGGAC







GTCTGGGGCCAAGGGACCACGG







TCACCGTCTCCTCAG






N438H
SEQ ID NO: 1
SEQ ID NO: 2
SEQ ID NO: 173
SEQ ID NO: 250
SEQ ID NO: 326





AREGYSSLSYYGMDV
GAGGTGCAGCTGGTGGAGTCTG
EVQLVESGGGFVQPGGSL






GGGGAGGCTTTGTCCAGCCTGG
RLSCAVSGFTFNSYWMSW






GGGGTCCCTGAGACTCTCCTGT
VRQAPGKGLEWVANINQD






GCAGTCTCTGGATTCACCTTTA
GSEKFYVASVKGRFTMSR






ATAGCTATTGGATGAGCTGGGT
DNAKKSVYVQMNSLRAED






CCGCCAGGCTCCAGGGAAGGGG
TAVYYCAREGYSSLSYYG






CTGGAGTGGGTGGCCAACATAA
MDVWGQGTTVTVSS






ACCAAGATGGAAGTGAGAAATT







CTATGTGGCCTCTGTGAAGGGC







CGATTCACCATGTCCAGAGACA







ACGCCAAGAAATCAGTGTATGT







ACAAATGAACAGCCTGAGAGCC







GAGGACACGGCTGTGTATTACT







GTGCGAGAGAGGGGTATAGTAG







TCTGTCCTATTATGGTATGGAC







GTCTGGGGCCAAGGGACCACGG







TCACCGTCTCCTCAG






N439H
SEQ ID NO: 1
SEQ ID NO: 2
SEQ ID NO: 174
SEQ ID NO: 251
SEQ ID NO: 327





AREGYSSMSYYGMDV
GAGGTGCAGCTGGTGGAGTCTG
EVQLVESGGGFVQPGGSL






GGGGAGGCTTTGTCCAGCCTGG
RLSCAVSGFTFNSYWMSW






GGGGTCCCTGAGACTCTCCTGT
VRQAPGKGLEWVANINQD






GCAGTCTCTGGATTCACCTTTA
GSEKFYVASVKGRFTMSR






ATAGCTATTGGATGAGCTGGGT
DNAKKSVYVQMNSLRAED






CCGCCAGGCTCCAGGGAAGGGG
TAVYYCAREGYSSMSYYG






CTGGAGTGGGTGGCCAACATAA
MDVWGQGTTVTVSS






ACCAAGATGGAAGTGAGAAATT







CTATGTGGCCTCTGTGAAGGGC







CGATTCACCATGTCCAGAGACA







ACGCCAAGAAATCAGTGTATGT







ACAAATGAACAGCCTGAGAGCC







GAGGACACGGCTGTGTATTACT







GTGCGAGAGAGGGGTATAGTAG







TATGTCCTATTATGGTATGGAC







GTCTGGGGCCAAGGGACCACGG







TCACCGTCTCCTCAG






N440H
SEQ ID NO: 1
SEQ ID NO: 2
SEQ ID NO: 175
SEQ ID NO: 252
SEQ ID NO: 328





AREGYSSNSYYGMDV
GAGGTGCAGCTGGTGGAGTCTG
EVQLVESGGGFVQPGGSL






GGGGAGGCTTTGTCCAGCCTGG
RLSCAVSGFTFNSYWMSW






GGGGTCCCTGAGACTCTCCTGT
VRQAPGKGLEWVANINQD






GCAGTCTCTGGATTCACCTTTA
GSEKFYVASVKGRFTMSR






ATAGCTATTGGATGAGCTGGGT
DNAKKSVYVQMNSLRAED






CCGCCAGGCTCCAGGGAAGGGG
TAVYYCAREGYSSNSYYG






CTGGAGTGGGTGGCCAACATAA
MDVWGQGTTVTVSS






ACCAAGATGGAAGTGAGAAATT







CTATGTGGCCTCTGTGAAGGGC







CGATTCACCATGTCCAGAGACA







ACGCCAAGAAATCAGTGTATGT







ACAAATGAACAGCCTGAGAGCC







GAGGACACGGCTGTGTATTACT







GTGCGAGAGAGGGGTATAGTAG







TAACTCCTATTATGGTATGGAC







GTCTGGGGCCAAGGGACCACGG







TCACCGTCTCCTCAG






N441H
SEQ ID NO: 1
SEQ ID NO: 2
SEQ ID NO: 176
SEQ ID NO: 253
SEQ ID NO: 329





AREGYSSPSYYGMDV
GAGGTGCAGCTGGTGGAGTCTG
EVQLVESGGGFVQPGGSL






GGGGAGGCTTTGTCCAGCCTGG
RLSCAVSGFTFNSYWMSW






GGGGTCCCTGAGACTCTCCTGT
VRQAPGKGLEWVANINQD






GCAGTCTCTGGATTCACCTTTA
GSEKFYVASVKGRFTMSR






ATAGCTATTGGATGAGCTGGGT
DNAKKSVYVQMNSLRAED






CCGCCAGGCTCCAGGGAAGGGG
TAVYYCAREGYSSPSYYG






CTGGAGTGGGTGGCCAACATAA
MDVWGQGTTVTVSS






ACCAAGATGGAAGTGAGAAATT







CTATGTGGCCTCTGTGAAGGGC







CGATTCACCATGTCCAGAGACA







ACGCCAAGAAATCAGTGTATGT







ACAAATGAACAGCCTGAGAGCC







GAGGACACGGCTGTGTATTACT







GTGCGAGAGAGGGGTATAGTAG







TCCCTCCTATTATGGTATGGAC







GTCTGGGGCCAAGGGACCACGG







TCACCGTCTCCTCAG






N442H
SEQ ID NO: 1
SEQ ID NO: 2
SEQ ID NO: 177
SEQ ID NO: 254
SEQ ID NO: 340





AREGYSSQSYYGMDV
GAGGTGCAGCTGGTGGAGTCTG
EVQLVESGGGFVQPGGSL






GGGGAGGCTTTGTCCAGCCTGG
RLSCAVSGFTFNSYWMSW






GGGGTCCCTGAGACTCTCCTGT
VRQAPGKGLEWVANINQD






GCAGTCTCTGGATTCACCTTTA
GSEKFYVASVKGRFTMSR






ATAGCTATTGGATGAGCTGGGT
DNAKKSVYVQMNSLRAED






CCGCCAGGCTCCAGGGAAGGGG
TAVYYCAREGYSSQSYYG






CTGGAGTGGGTGGCCAACATAA
MDVWGQGTTVTVSS






ACCAAGATGGAAGTGAGAAATT







CTATGTGGCCTCTGTGAAGGGC







CGATTCACCATGTCCAGAGACA







ACGCCAAGAAATCAGTGTATGT







ACAAATGAACAGCCTGAGAGCC







GAGGACACGGCTGTGTATTACT







GTGCGAGAGAGGGGTATAGTAG







TCAGTCCTATTATGGTATGGAC







GTCTGGGGCCAAGGGACCACGG







TCACCGTCTCCTCAG






N443H
SEQ ID NO: 1
SEQ ID NO: 2
SEQ ID NO: 178
SEQ ID NO: 255
SEQ ID NO: 341





AREGYSSRSYYGMDV
GAGGTGCAGCTGGTGGAGTCTG
EVQLVESGGGFVQPGGSL






GGGGAGGCTTTGTCCAGCCTGG
RLSCAVSGFTFNSYWMSW






GGGGTCCCTGAGACTCTCCTGT
VRQAPGKGLEWVANINQD






GCAGTCTCTGGATTCACCTTTA
GSEKFYVASVKGRFTMSR






ATAGCTATTGGATGAGCTGGGT
DNAKKSVYVQMNSLRAED






CCGCCAGGCTCCAGGGAAGGGG
TAVYYCAREGYSSRSYYG






CTGGAGTGGGTGGCCAACATAA
MDVWGQGTTVTVSS






ACCAAGATGGAAGTGAGAAATT







CTATGTGGCCTCTGTGAAGGGC







CGATTCACCATGTCCAGAGACA







ACGCCAAGAAATCAGTGTATGT







ACAAATGAACAGCCTGAGAGCC







GAGGACACGGCTGTGTATTACT







GTGCGAGAGAGGGGTATAGTAG







TAGATCCTATTATGGTATGGAC







GTCTGGGGCCAAGGGACCACGG







TCACCGTCTCCTCAG






N444H
SEQ ID NO: 1
SEQ ID NO: 2
SEQ ID NO: 179
SEQ ID NO: 256
SEQ ID NO: 342





AREGYSSTSYYGMDV
GAGGTGCAGCTGGTGGAGTCTG
EVQLVESGGGFVQPGGSL






GGGGAGGCTTTGTCCAGCCTGG
RLSCAVSGFTFNSYWMSW






GGGGTCCCTGAGACTCTCCTGT
VRQAPGKGLEWVANINQD






GCAGTCTCTGGATTCACCTTTA
GSEKFYVASVKGRFTMSR






ATAGCTATTGGATGAGCTGGGT
DNAKKSVYVQMNSLRAED






CCGCCAGGCTCCAGGGAAGGGG
TAVYYCAREGYSSTSYYG






CTGGAGTGGGTGGCCAACATAA
MDVWGQGTTVTVSS






ACCAAGATGGAAGTGAGAAATT







CTATGTGGCCTCTGTGAAGGGC







CGATTCACCATGTCCAGAGACA







ACGCCAAGAAATCAGTGTATGT







ACAAATGAACAGCCTGAGAGCC







GAGGACACGGCTGTGTATTACT







GTGCGAGAGAGGGGTATAGTAG







TACCTCCTATTATGGTATGGAC







GTCTGGGGCCAAGGGACCACGG







TCACCGTCTCCTCAG






N445H
SEQ ID NO: 1
SEQ ID NO: 2
SEQ ID NO: 180
SEQ ID NO: 257
SEQ ID NO: 343





AREGYSSVSYYGMDV
GAGGTGCAGCTGGTGGAGTCTG
EVQLVESGGGFVQPGGSL






GGGGAGGCTTTGTCCAGCCTGG
RLSCAVSGFTFNSYWMSW






GGGGTCCCTGAGACTCTCCTGT
VRQAPGKGLEWVANINQD






GCAGTCTCTGGATTCACCTTTA
GSEKFYVASVKGRFTMSR






ATAGCTATTGGATGAGCTGGGT
DNAKKSVYVQMNSLRAED






CCGCCAGGCTCCAGGGAAGGGG
TAVYYCAREGYSSVSYYG






CTGGAGTGGGTGGCCAACATAA
MDVWGQGTTVTVSS






ACCAAGATGGAAGTGAGAAATT







CTATGTGGCCTCTGTGAAGGGC







CGATTCACCATGTCCAGAGACA







ACGCCAAGAAATCAGTGTATGT







ACAAATGAACAGCCTGAGAGCC







GAGGACACGGCTGTGTATTACT







GTGCGAGAGAGGGGTATAGTAG







TGTGTCCTATTATGGTATGGAC







GTCTGGGGCCAAGGGACCACGG







TCACCGTCTCCTCAG






N446H
SEQ ID NO: 1
SEQ ID NO: 2
SEQ ID NO: 181
SEQ ID NO: 258
SEQ ID NO: 344





AREGYSSWSYYGMDV
GAGGTGCAGCTGGTGGAGTCTG
EVQLVESGGGFVQPGGSL






GGGGAGGCTTTGTCCAGCCTGG
RLSCAVSGFTFNSYWMSW






GGGGTCCCTGAGACTCTCCTGT
VRQAPGKGLEWVANINQD






GCAGTCTCTGGATTCACCTTTA
GSEKFYVASVKGRFTMSR






ATAGCTATTGGATGAGCTGGGT
DNAKKSVYVQMNSLRAED






CCGCCAGGCTCCAGGGAAGGGG
TAVYYCAREGYSSWSYYG






CTGGAGTGGGTGGCCAACATAA
MDVWGQGTTVTVSS






ACCAAGATGGAAGTGAGAAATT







CTATGTGGCCTCTGTGAAGGGC







CGATTCACCATGTCCAGAGACA







ACGCCAAGAAATCAGTGTATGT







ACAAATGAACAGCCTGAGAGCC







GAGGACACGGCTGTGTATTACT







GTGCGAGAGAGGGGTATAGTAG







TTGGTCCTATTATGGTATGGAC







GTCTGGGGCCAAGGGACCACGG







TCACCGTCTCCTCAG






N447H
SEQ ID NO: 1
SEQ ID NO: 2
SEQ ID NO: 182
SEQ ID NO: 259
SEQ ID NO: 345





AREGYSSYSYYGMDV
GAGGTGCAGCTGGTGGAGTCTG
EVQLVESGGGFVQPGGSL






GGGGAGGCTTTGTCCAGCCTGG
RLSCAVSGFTFNSYWMSW






GGGGTCCCTGAGACTCTCCTGT
VRQAPGKGLEWVANINQD






GCAGTCTCTGGATTCACCTTTA
GSEKFYVASVKGRFTMSR






ATAGCTATTGGATGAGCTGGGT
DNAKKSVYVQMNSLRAED






CCGCCAGGCTCCAGGGAAGGGG
TAVYYCAREGYSSYSYYG






CTGGAGTGGGTGGCCAACATAA
MDVWGQGTTVTVSS






ACCAAGATGGAAGTGAGAAATT







CTATGTGGCCTCTGTGAAGGGC







CGATTCACCATGTCCAGAGACA







ACGCCAAGAAATCAGTGTATGT







ACAAATGAACAGCCTGAGAGCC







GAGGACACGGCTGTGTATTACT







GTGCGAGAGAGGGGTATAGTAG







TTACTCCTATTATGGTATGGAC







GTCTGGGGCCAAGGGACCACGG







TCACCGTCTCCTCAG






N448H
SEQ ID NO: 1
SEQ ID NO: 2
SEQ ID NO: 183
SEQ ID NO: 260
SEQ ID NO: 346





AREGYSSSCYYGMDV
GAGGTGCAGCTGGTGGAGTCTG
EVQLVESGGGFVQPGGSL






GGGGAGGCTTTGTCCAGCCTGG
RLSCAVSGFTFNSYWMSW






GGGGTCCCTGAGACTCTCCTGT
VRQAPGKGLEWVANINQD






GCAGTCTCTGGATTCACCTTTA
GSEKFYVASVKGRFTMSR






ATAGCTATTGGATGAGCTGGGT
DNAKKSVYVQMNSLRAED






CCGCCAGGCTCCAGGGAAGGGG
TAVYYCAREGYSSSCYYG






CTGGAGTGGGTGGCCAACATAA
MDVWGQGTTVTVSS






ACCAAGATGGAAGTGAGAAATT







CTATGTGGCCTCTGTGAAGGGC







CGATTCACCATGTCCAGAGACA







ACGCCAAGAAATCAGTGTATGT







ACAAATGAACAGCCTGAGAGCC







GAGGACACGGCTGTGTATTACT







GTGCGAGAGAGGGGTATAGTAG







TTCGTGCTATTATGGTATGGAC







GTCTGGGGCCAAGGGACCACGG







TCACCGTCTCCTCAG






N449H
SEQ ID NO: 1
SEQ ID NO: 2
SEQ ID NO: 184
SEQ ID NO: 261
SEQ ID NO: 347





AREGYSSSDYYGMDV
GAGGTGCAGCTGGTGGAGTCTG
EVQLVESGGGFVQPGGSL






GGGGAGGCTTTGTCCAGCCTGG
RLSCAVSGFTFNSYWMSW






GGGGTCCCTGAGACTCTCCTGT
VRQAPGKGLEWVANINQD






GCAGTCTCTGGATTCACCTTTA
GSEKFYVASVKGRFTMSR






ATAGCTATTGGATGAGCTGGGT
DNAKKSVYVQMNSLRAED






CCGCCAGGCTCCAGGGAAGGGG
TAVYYCAREGYSSSDYYG






CTGGAGTGGGTGGCCAACATAA
MDVWGQGTTVTVSS






ACCAAGATGGAAGTGAGAAATT







CTATGTGGCCTCTGTGAAGGGC







CGATTCACCATGTCCAGAGACA







ACGCCAAGAAATCAGTGTATGT







ACAAATGAACAGCCTGAGAGCC







GAGGACACGGCTGTGTATTACT







GTGCGAGAGAGGGGTATAGTAG







TTCGGACTATTATGGTATGGAC







GTCTGGGGCCAAGGGACCACGG







TCACCGTCTCCTCAG






N450H
SEQ ID NO: 1
SEQ ID NO: 2
SEQ ID NO: 185
SEQ ID NO: 262
SEQ ID NO: 348





AREGYSSSEYYGMDV
GAGGTGCAGCTGGTGGAGTCTG
EVQLVESGGGFVQPGGSL






GGGGAGGCTTTGTCCAGCCTGG
RLSCAVSGFTFNSYWMSW






GGGGTCCCTGAGACTCTCCTGT
VRQAPGKGLEWVANINQD






GCAGTCTCTGGATTCACCTTTA
GSEKFYVASVKGRFTMSR






ATAGCTATTGGATGAGCTGGGT
DNAKKSVYVQMNSLRAED






CCGCCAGGCTCCAGGGAAGGGG
TAVYYCAREGYSSSEYYG






CTGGAGTGGGTGGCCAACATAA
MDVWGQGTTVTVSS






ACCAAGATGGAAGTGAGAAATT







CTATGTGGCCTCTGTGAAGGGC







CGATTCACCATGTCCAGAGACA







ACGCCAAGAAATCAGTGTATGT







ACAAATGAACAGCCTGAGAGCC







GAGGACACGGCTGTGTATTACT







GTGCGAGAGAGGGGTATAGTAG







TTCGGAGTATTATGGTATGGAC







GTCTGGGGCCAAGGGACCACGG







TCACCGTCTCCTCAG






N451H
SEQ ID NO: 1
SEQ ID NO: 2
SEQ ID NO: 186
SEQ ID NO: 263
SEQ ID NO: 349





AREGYSSSFYYGMDV
GAGGTGCAGCTGGTGGAGTCTG
EVQLVESGGGFVQPGGSL






GGGGAGGCTTTGTCCAGCCTGG
RLSCAVSGFTFNSYWMSW






GGGGTCCCTGAGACTCTCCTGT
VRQAPGKGLEWVANINQD






GCAGTCTCTGGATTCACCTTTA
GSEKFYVASVKGRFTMSR






ATAGCTATTGGATGAGCTGGGT
DNAKKSVYVQMNSLRAED






CCGCCAGGCTCCAGGGAAGGGG
TAVYYCAREGYSSSFYYG






CTGGAGTGGGTGGCCAACATAA
MDVWGQGTTVTVSS






ACCAAGATGGAAGTGAGAAATT







CTATGTGGCCTCTGTGAAGGGC







CGATTCACCATGTCCAGAGACA







ACGCCAAGAAATCAGTGTATGT







ACAAATGAACAGCCTGAGAGCC







GAGGACACGGCTGTGTATTACT







GTGCGAGAGAGGGGTATAGTAG







TTCGTTCTATTATGGTATGGAC







GTCTGGGGCCAAGGGACCACGG







TCACCGTCTCCTCAG






N452H
SEQ ID NO: 1
SEQ ID NO: 2
SEQ ID NO: 187
SEQ ID NO: 264
SEQ ID NO: 350





AREGYSSSGYYGMDV
GAGGTGCAGCTGGTGGAGTCTG
EVQLVESGGGFVQPGGSL






GGGGAGGCTTTGTCCAGCCTGG
RLSCAVSGFTFNSYWMSW






GGGGTCCCTGAGACTCTCCTGT
VRQAPGKGLEWVANINQD






GCAGTCTCTGGATTCACCTTTA
GSEKFYVASVKGRFTMSR






ATAGCTATTGGATGAGCTGGGT
DNAKKSVYVQMNSLRAED






CCGCCAGGCTCCAGGGAAGGGG
TAVYYCAREGYSSSGYYG






CTGGAGTGGGTGGCCAACATAA
MDVWGQGTTVTVSS






ACCAAGATGGAAGTGAGAAATT







CTATGTGGCCTCTGTGAAGGGC







CGATTCACCATGTCCAGAGACA







ACGCCAAGAAATCAGTGTATGT







ACAAATGAACAGCCTGAGAGCC







GAGGACACGGCTGTGTATTACT







GTGCGAGAGAGGGGTATAGTAG







TTCGGGCTATTATGGTATGGAC







GTCTGGGGCCAAGGGACCACGG







TCACCGTCTCCTCAG






N453H
SEQ ID NO: 1
SEQ ID NO: 2
SEQ ID NO: 188
SEQ ID NO: 265
SEQ ID NO: 351





AREGYSSSHYYGMDV
GAGGTGCAGCTGGTGGAGTCTG
EVQLVESGGGFVQPGGSL






GGGGAGGCTTTGTCCAGCCTGG
RLSCAVSGFTFNSYWMSW






GGGGTCCCTGAGACTCTCCTGT
VRQAPGKGLEWVANINQD






GCAGTCTCTGGATTCACCTTTA
GSEKFYVASVKGRFTMSR






ATAGCTATTGGATGAGCTGGGT
DNAKKSVYVQMNSLRAED






CCGCCAGGCTCCAGGGAAGGGG
TAVYYCAREGYSSSHYYG






CTGGAGTGGGTGGCCAACATAA
MDVWGQGTTVTVSS






ACCAAGATGGAAGTGAGAAATT







CTATGTGGCCTCTGTGAAGGGC







CGATTCACCATGTCCAGAGACA







ACGCCAAGAAATCAGTGTATGT







ACAAATGAACAGCCTGAGAGCC







GAGGACACGGCTGTGTATTACT







GTGCGAGAGAGGGGTATAGTAG







TTCGCACTATTATGGTATGGAC







GTCTGGGGCCAAGGGACCACGG







TCACCGTCTCCTCAG






N454H
SEQ ID NO: 1
SEQ ID NO: 2
SEQ ID NO: 189
SEQ ID NO: 266
SEQ ID NO: 352





AREGYSSSIYYGMDV
GAGGTGCAGCTGGTGGAGTCTG
EVQLVESGGGFVQPGGSL






GGGGAGGCTTTGTCCAGCCTGG
RLSCAVSGFTFNSYWMSW






GGGGTCCCTGAGACTCTCCTGT
VRQAPGKGLEWVANINQD






GCAGTCTCTGGATTCACCTTTA
GSEKFYVASVKGRFTMSR






ATAGCTATTGGATGAGCTGGGT
DNAKKSVYVQMNSLRAED






CCGCCAGGCTCCAGGGAAGGGG
TAVYYCAREGYSSSIYYG






CTGGAGTGGGTGGCCAACATAA
MDVWGQGTTVTVSS






ACCAAGATGGAAGTGAGAAATT







CTATGTGGCCTCTGTGAAGGGC







CGATTCACCATGTCCAGAGACA







ACGCCAAGAAATCAGTGTATGT







ACAAATGAACAGCCTGAGAGCC







GAGGACACGGCTGTGTATTACT







GTGCGAGAGAGGGGTATAGTAG







TTCGATCTATTATGGTATGGAC







GTCTGGGGCCAAGGGACCACGG







TCACCGTCTCCTCAG






N455H
SEQ ID NO: 1
SEQ ID NO: 2
SEQ ID NO: 190
SEQ ID NO: 267
SEQ ID NO: 353





AREGYSSSKYYGMDV
GAGGTGCAGCTGGTGGAGTCTG
EVQLVESGGGFVQPGGSL






GGGGAGGCTTTGTCCAGCCTGG
RLSCAVSGFTFNSYWMSW






GGGGTCCCTGAGACTCTCCTGT
VRQAPGKGLEWVANINQD






GCAGTCTCTGGATTCACCTTTA
GSEKFYVASVKGRFTMSR






ATAGCTATTGGATGAGCTGGGT
DNAKKSVYVQMNSLRAED






CCGCCAGGCTCCAGGGAAGGGG
TAVYYCAREGYSSSKYYG






CTGGAGTGGGTGGCCAACATAA
MDVWGQGTTVTVSS






ACCAAGATGGAAGTGAGAAATT







CTATGTGGCCTCTGTGAAGGGC







CGATTCACCATGTCCAGAGACA







ACGCCAAGAAATCAGTGTATGT







ACAAATGAACAGCCTGAGAGCC







GAGGACACGGCTGTGTATTACT







GTGCGAGAGAGGGGTATAGTAG







TTCGAAGTATTATGGTATGGAC







GTCTGGGGCCAAGGGACCACGG







TCACCGTCTCCTCAG









N456H
SEQ ID NO: 1
SEQ ID NO: 2
SEQ ID NO: 191
SEQ ID NO: 268
SEQ ID NO: 354





AREGYSSSLYYGMDV
GAGGTGCAGCTGGTGGAGTCTG
EVQLVESGGGFVQPGGSL






GGGGAGGCTTTGTCCAGCCTGG
RLSCAVSGFTFNSYWMSW






GGGGTCCCTGAGACTCTCCTGT
VRQAPGKGLEWVANINQD






GCAGTCTCTGGATTCACCTTTA
GSEKFYVASVKGRFTMSR






ATAGCTATTGGATGAGCTGGGT
DNAKKSVYVQMNSLRAED






CCGCCAGGCTCCAGGGAAGGGG
TAVYYCAREGYSSSLYYG






CTGGAGTGGGTGGCCAACATAA
MDVWGQGTTVTVSS






ACCAAGATGGAAGTGAGAAATT







CTATGTGGCCTCTGTGAAGGGC







CGATTCACCATGTCCAGAGACA







ACGCCAAGAAATCAGTGTATGT







ACAAATGAACAGCCTGAGAGCC







GAGGACACGGCTGTGTATTACT







GTGCGAGAGAGGGGTATAGTAG







TTCGCTGTATTATGGTATGGAC







GTCTGGGGCCAAGGGACCACGG







TCACCGTCTCCTCAG






N457H
SEQ ID NO: 1
SEQ ID NO: 2
SEQ ID NO: 192
SEQ ID NO: 269
SEQ ID NO: 355





AREGYSSSMYYGMDV
GAGGTGCAGCTGGTGGAGTCTG
EVQLVESGGGFVQPGGSL






GGGGAGGCTTTGTCCAGCCTGG
RLSCAVSGFTFNSYWMSW






GGGGTCCCTGAGACTCTCCTGT
VRQAPGKGLEWVANINQD






GCAGTCTCTGGATTCACCTTTA
GSEKFYVASVKGRFTMSR






ATAGCTATTGGATGAGCTGGGT
DNAKKSVYVQMNSLRAED






CCGCCAGGCTCCAGGGAAGGGG
TAVYYCAREGYSSSMYYG






CTGGAGTGGGTGGCCAACATAA
MDVWGQGTTVTVSS






ACCAAGATGGAAGTGAGAAATT







CTATGTGGCCTCTGTGAAGGGC







CGATTCACCATGTCCAGAGACA







ACGCCAAGAAATCAGTGTATGT







ACAAATGAACAGCCTGAGAGCC







GAGGACACGGCTGTGTATTACT







GTGCGAGAGAGGGGTATAGTAG







TTCGATGTATTATGGTATGGAC







GTCTGGGGCCAAGGGACCACGG







TCACCGTCTCCTCAG






N458H
SEQ ID NO: 1
SEQ ID NO: 2
SEQ ID NO: 193
SEQ ID NO: 270
SEQ ID NO: 356





AREGYSSSNYYGMDV
GAGGTGCAGCTGGTGGAGTCTG
EVQLVESGGGFVQPGGSL






GGGGAGGCTTTGTCCAGCCTGG
RLSCAVSGFTFNSYWMSW






GGGGTCCCTGAGACTCTCCTGT
VRQAPGKGLEWVANINQD






GCAGTCTCTGGATTCACCTTTA
GSEKFYVASVKGRFTMSR






ATAGCTATTGGATGAGCTGGGT
DNAKKSVYVQMNSLRAED






CCGCCAGGCTCCAGGGAAGGGG
TAVYYCAREGYSSSNYYG






CTGGAGTGGGTGGCCAACATAA
MDVWGQGTTVTVSS






ACCAAGATGGAAGTGAGAAATT







CTATGTGGCCTCTGTGAAGGGC







CGATTCACCATGTCCAGAGACA







ACGCCAAGAAATCAGTGTATGT







ACAAATGAACAGCCTGAGAGCC







GAGGACACGGCTGTGTATTACT







GTGCGAGAGAGGGGTATAGTAG







TTCGAACTATTATGGTATGGAC







GTCTGGGGCCAAGGGACCACGG







TCACCGTCTCCTCAG






N459H
SEQ ID NO: 1
SEQ ID NO: 2
SEQ ID NO: 194
SEQ ID NO: 271
SEQ ID NO: 357





AREGYSSSPYYGMDV
GAGGTGCAGCTGGTGGAGTCTG
EVQLVESGGGFVQPGGSL






GGGGAGGCTTTGTCCAGCCTGG
RLSCAVSGFTFNSYWMSW






GGGGTCCCTGAGACTCTCCTGT
VRQAPGKGLEWVANINQD






GCAGTCTCTGGATTCACCTTTA
GSEKFYVASVKGRFTMSR






ATAGCTATTGGATGAGCTGGGT
DNAKKSVYVQMNSLRAED






CCGCCAGGCTCCAGGGAAGGGG
TAVYYCAREGYSSSPYYG






CTGGAGTGGGTGGCCAACATAA
MDVWGQGTTVTVSS






ACCAAGATGGAAGTGAGAAATT







CTATGTGGCCTCTGTGAAGGGC







CGATTCACCATGTCCAGAGACA







ACGCCAAGAAATCAGTGTATGT







ACAAATGAACAGCCTGAGAGCC







GAGGACACGGCTGTGTATTACT







GTGCGAGAGAGGGGTATAGTAG







TTCGCCCTATTATGGTATGGAC







GTCTGGGGCCAAGGGACCACGG







TCACCGTCTCCTCAG






N460H
SEQ ID NO: 1
SEQ ID NO: 2
SEQ ID NO: 195
SEQ ID NO: 272
SEQ ID NO: 358





AREGYSSSQYYGMDV
GAGGTGCAGCTGGTGGAGTCTG
EVQLVESGGGFVQPGGSL






GGGGAGGCTTTGTCCAGCCTGG
RLSCAVSGFTFNSYWMSW






GGGGTCCCTGAGACTCTCCTGT
VRQAPGKGLEWVANINQD






GCAGTCTCTGGATTCACCTTTA
GSEKFYVASVKGRFTMSR






ATAGCTATTGGATGAGCTGGGT
DNAKKSVYVQMNSLRAED






CCGCCAGGCTCCAGGGAAGGGG
TAVYYCAREGYSSSQYYG






CTGGAGTGGGTGGCCAACATAA
MDVWGQGTTVTVSS






ACCAAGATGGAAGTGAGAAATT







CTATGTGGCCTCTGTGAAGGGC







CGATTCACCATGTCCAGAGACA







ACGCCAAGAAATCAGTGTATGT







ACAAATGAACAGCCTGAGAGCC







GAGGACACGGCTGTGTATTACT







GTGCGAGAGAGGGGTATAGTAG







TTCGCAGTATTATGGTATGGAC







GTCTGGGGCCAAGGGACCACGG







TCACCGTCTCCTCAG






N461H
SEQ ID NO: 1
SEQ ID NO: 2
SEQ ID NO: 196
SEQ ID NO: 273
SEQ ID NO: 359





AREGYSSSRYYGMDV
GAGGTGCAGCTGGTGGAGTCTG
EVQLVESGGGFVQPGGSL






GGGGAGGCTTTGTCCAGCCTGG
RLSCAVSGFTFNSYWMSW






GGGGTCCCTGAGACTCTCCTGT
VRQAPGKGLEWVANINQD






GCAGTCTCTGGATTCACCTTTA
GSEKFYVASVKGRFTMSR






ATAGCTATTGGATGAGCTGGGT
DNAKKSVYVQMNSLRAED






CCGCCAGGCTCCAGGGAAGGGG
TAVYYCAREGYSSSRYYG






CTGGAGTGGGTGGCCAACATAA
MDVWGQGTTVTVSS






ACCAAGATGGAAGTGAGAAATT







CTATGTGGCCTCTGTGAAGGGC







CGATTCACCATGTCCAGAGACA







ACGCCAAGAAATCAGTGTATGT







ACAAATGAACAGCCTGAGAGCC







GAGGACACGGCTGTGTATTACT







GTGCGAGAGAGGGGTATAGTAG







TTCGAGATATTATGGTATGGAC







GTCTGGGGCCAAGGGACCACGG







TCACCGTCTCCTCAG






N462H
SEQ ID NO: 1
SEQ ID NO: 2
SEQ ID NO: 197
SEQ ID NO: 274
SEQ ID NO: 360





AREGYSSSTYYGMDV
GAGGTGCAGCTGGTGGAGTCTG
EVQLVESGGGFVQPGGSL






GGGGAGGCTTTGTCCAGCCTGG
RLSCAVSGFTFNSYWMSW






GGGGTCCCTGAGACTCTCCTGT
VRQAPGKGLEWVANINQD






GCAGTCTCTGGATTCACCTTTA
GSEKFYVASVKGRFTMSR






ATAGCTATTGGATGAGCTGGGT
DNAKKSVYVQMNSLRAED






CCGCCAGGCTCCAGGGAAGGGG
TAVYYCAREGYSSSTYYG






CTGGAGTGGGTGGCCAACATAA
MDVWGQGTTVTVSS






ACCAAGATGGAAGTGAGAAATT







CTATGTGGCCTCTGTGAAGGGC







CGATTCACCATGTCCAGAGACA







ACGCCAAGAAATCAGTGTATGT







ACAAATGAACAGCCTGAGAGCC







GAGGACACGGCTGTGTATTACT







GTGCGAGAGAGGGGTATAGTAG







TTCGACCTATTATGGTATGGAC







GTCTGGGGCCAAGGGACCACGG







TCACCGTCTCCTCAG






N463H
SEQ ID NO: 1
SEQ ID NO: 2
SEQ ID NO: 198
SEQ ID NO: 275
SEQ ID NO: 361





AREGYSSSVYYGMDV
GAGGTGCAGCTGGTGGAGTCTG
EVQLVESGGGFVQPGGSL






GGGGAGGCTTTGTCCAGCCTGG
RLSCAVSGFTFNSYWMSW






GGGGTCCCTGAGACTCTCCTGT
VRQAPGKGLEWVANINQD






GCAGTCTCTGGATTCACCTTTA
GSEKFYVASVKGRFTMSR






ATAGCTATTGGATGAGCTGGGT
DNAKKSVYVQMNSLRAED






CCGCCAGGCTCCAGGGAAGGGG
TAVYYCAREGYSSSVYYG






CTGGAGTGGGTGGCCAACATAA
MDVWGQGTTVTVSS






ACCAAGATGGAAGTGAGAAATT







CTATGTGGCCTCTGTGAAGGGC







CGATTCACCATGTCCAGAGACA







ACGCCAAGAAATCAGTGTATGT







ACAAATGAACAGCCTGAGAGCC







GAGGACACGGCTGTGTATTACT







GTGCGAGAGAGGGGTATAGTAG







TTCGGTGTATTATGGTATGGAC







GTCTGGGGCCAAGGGACCACGG







TCACCGTCTCCTCAG






N464H
SEQ ID NO: 1
SEQ ID NO: 2
SEQ ID NO: 199
SEQ ID NO: 276
SEQ ID NO: 362





AREGYSSSWYYGMDV
GAGGTGCAGCTGGTGGAGTCTG
EVQLVESGGGFVQPGGSL






GGGGAGGCTTTGTCCAGCCTGG
RLSCAVSGFTFNSYWMSW






GGGGTCCCTGAGACTCTCCTGT
VRQAPGKGLEWVANINQD






GCAGTCTCTGGATTCACCTTTA
GSEKFYVASVKGRFTMSR






ATAGCTATTGGATGAGCTGGGT
DNAKKSVYVQMNSLRAED






CCGCCAGGCTCCAGGGAAGGGG
TAVYYCAREGYSSSWYYG






CTGGAGTGGGTGGCCAACATAA
MDVWGQGTTVTVSS






ACCAAGATGGAAGTGAGAAATT







CTATGTGGCCTCTGTGAAGGGC







CGATTCACCATGTCCAGAGACA







ACGCCAAGAAATCAGTGTATGT







ACAAATGAACAGCCTGAGAGCC







GAGGACACGGCTGTGTATTACT







GTGCGAGAGAGGGGTATAGTAG







TTCGTGGTATTATGGTATGGAC







GTCTGGGGCCAAGGGACCACGG







TCACCGTCTCCTCAG






N465H
SEQ ID NO: 1
SEQ ID NO: 2
SEQ ID NO: 200
SEQ ID NO: 277
SEQ ID NO: 363





AREGYSSSYYYGMDV
GAGGTGCAGCTGGTGGAGTCTG
EVQLVESGGGFVQPGGSL






GGGGAGGCTTTGTCCAGCCTGG
RLSCAVSGFTFNSYWMSW






GGGGTCCCTGAGACTCTCCTGT
VRQAPGKGLEWVANINQD






GCAGTCTCTGGATTCACCTTTA
GSEKFYVASVKGRFTMSR






ATAGCTATTGGATGAGCTGGGT
DNAKKSVYVQMNSLRAED






CCGCCAGGCTCCAGGGAAGGGG
TAVYYCAREGYSSSYYYG






CTGGAGTGGGTGGCCAACATAA
MDVWGQGTTVTVSS






ACCAAGATGGAAGTGAGAAATT







CTATGTGGCCTCTGTGAAGGGC







CGATTCACCATGTCCAGAGACA







ACGCCAAGAAATCAGTGTATGT







ACAAATGAACAGCCTGAGAGCC







GAGGACACGGCTGTGTATTACT







GTGCGAGAGAGGGGTATAGTAG







TTCGTACTATTATGGTATGGAC







GTCTGGGGCCAAGGGACCACGG







TCACCGTCTCCTCAG






N467H
SEQ ID NO: 1
SEQ ID NO: 2
SEQ ID NO: 201
SEQ ID NO: 278
SEQ ID NO: 364





AREGYCSSSYYGMDV
GAGGTGCAGCTGGTGGAGTCTG
EVQLVESGGGFVQPGGSL






GGGGAGGCTTTGTCCAGCCTGG
RLSCAVSGFTFNSYWMSW






GGGGTCCCTGAGACTCTCCTGT
VRQAPGKGLEWVANINQD






GCAGTCTCTGGATTCACCTTTA
GSEKFYVASVKGRFTMSR






ATAGCTATTGGATGAGCTGGGT
DNAKKSVYVQMNSLRAED






CCGCCAGGCTCCAGGGAAGGGG
TAVYYCAREGYCSSSYYG






CTGGAGTGGGTGGCCAACATAA
MDVWGQGTTVTVSS






ACCAAGATGGAAGTGAGAAATT







CTATGTGGCCTCTGTGAAGGGC







CGATTCACCATGTCCAGAGACA







ACGCCAAGAAATCAGTGTATGT







ACAAATGAACAGCCTGAGAGCC







GAGGACACGGCTGTGTATTACT







GTGCGAGAGAGGGGTATTGCAG







TTCGTCCTATTATGGTATGGAC







GTCTGGGGCCAAGGGACCACGG







TCACCGTCTCCTCAG






N468H
SEQ ID NO: 1
SEQ ID NO: 2
SEQ ID NO: 202
SEQ ID NO: 279
SEQ ID NO: 365





AREGYDSSSYYGMDV
GAGGTGCAGCTGGTGGAGTCTG
EVQLVESGGGFVQPGGSL






GGGGAGGCTTTGTCCAGCCTGG
RLSCAVSGFTFNSYWMSW






GGGGTCCCTGAGACTCTCCTGT
VRQAPGKGLEWVANINQD






GCAGTCTCTGGATTCACCTTTA
GSEKFYVASVKGRFTMSR






ATAGCTATTGGATGAGCTGGGT
DNAKKSVYVQMNSLRAED






CCGCCAGGCTCCAGGGAAGGGG
TAVYYCAREGYDSSSYYG






CTGGAGTGGGTGGCCAACATAA
MDVWGQGTTVTVSS






ACCAAGATGGAAGTGAGAAATT







CTATGTGGCCTCTGTGAAGGGC







CGATTCACCATGTCCAGAGACA







ACGCCAAGAAATCAGTGTATGT







ACAAATGAACAGCCTGAGAGCC







GAGGACACGGCTGTGTATTACT







GTGCGAGAGAGGGGTATGACAG







TTCGTCCTATTATGGTATGGAC







GTCTGGGGCCAAGGGACCACGG







TCACCGTCTCCTCAG






N469H
SEQ ID NO: 1
SEQ ID NO: 2
SEQ ID NO: 203
SEQ ID NO: 280
SEQ ID NO: 366





AREGYESSSYYGMDV
GAGGTGCAGCTGGTGGAGTCTG
EVQLVESGGGFVQPGGSL






GGGGAGGCTTTGTCCAGCCTGG
RLSCAVSGFTFNSYWMSW






GGGGTCCCTGAGACTCTCCTGT
VRQAPGKGLEWVANINQD






GCAGTCTCTGGATTCACCTTTA
GSEKFYVASVKGRFTMSR






ATAGCTATTGGATGAGCTGGGT
DNAKKSVYVQMNSLRAED






CCGCCAGGCTCCAGGGAAGGGG
TAVYYCAREGYESSSYYG






CTGGAGTGGGTGGCCAACATAA
MDVWGQGTTVTVSS






ACCAAGATGGAAGTGAGAAATT







CTATGTGGCCTCTGTGAAGGGC







CGATTCACCATGTCCAGAGACA







ACGCCAAGAAATCAGTGTATGT







ACAAATGAACAGCCTGAGAGCC







GAGGACACGGCTGTGTATTACT







GTGCGAGAGAGGGGTATGAGAG







TTCGTCCTATTATGGTATGGAC







GTCTGGGGCCAAGGGACCACGG







TCACCGTCTCCTCAG






N470H
SEQ ID NO: 1
SEQ ID NO: 2
SEQ ID NO: 204
SEQ ID NO: 281
SEQ ID NO: 367





AREGYFSSSYYGMDV
GAGGTGCAGCTGGTGGAGTCTG
EVQLVESGGGFVQPGGSL






GGGGAGGCTTTGTCCAGCCTGG
RLSCAVSGFTFNSYWMSW






GGGGTCCCTGAGACTCTCCTGT
VRQAPGKGLEWVANINQD






GCAGTCTCTGGATTCACCTTTA
GSEKFYVASVKGRFTMSR






ATAGCTATTGGATGAGCTGGGT
DNAKKSVYVQMNSLRAED






CCGCCAGGCTCCAGGGAAGGGG
TAVYYCAREGYFSSSYYG






CTGGAGTGGGTGGCCAACATAA
MDVWGQGTTVTVSS






ACCAAGATGGAAGTGAGAAATT







CTATGTGGCCTCTGTGAAGGGC







CGATTCACCATGTCCAGAGACA







ACGCCAAGAAATCAGTGTATGT







ACAAATGAACAGCCTGAGAGCC







GAGGACACGGCTGTGTATTACT







GTGCGAGAGAGGGGTATTTCAG







TTCGTCCTATTATGGTATGGAC







GTCTGGGGCCAAGGGACCACGG







TCACCGTCTCCTCAG






N471H
SEQ ID NO: 1
SEQ ID NO: 2
SEQ ID NO: 205
SEQ ID NO: 282
SEQ ID NO: 368





AREGYGSSSYYGMDV
GAGGTGCAGCTGGTGGAGTCTG
EVQLVESGGGFVQPGGSL






GGGGAGGCTTTGTCCAGCCTGG
RLSCAVSGFTFNSYWMSW






GGGGTCCCTGAGACTCTCCTGT
VRQAPGKGLEWVANINQD






GCAGTCTCTGGATTCACCTTTA
GSEKFYVASVKGRFTMSR






ATAGCTATTGGATGAGCTGGGT
DNAKKSVYVQMNSLRAED






CCGCCAGGCTCCAGGGAAGGGG
TAVYYCAREGYGSSSYYG






CTGGAGTGGGTGGCCAACATAA
MDVWGQGTTVTVSS






ACCAAGATGGAAGTGAGAAATT







CTATGTGGCCTCTGTGAAGGGC







CGATTCACCATGTCCAGAGACA







ACGCCAAGAAATCAGTGTATGT







ACAAATGAACAGCCTGAGAGCC







GAGGACACGGCTGTGTATTACT







GTGCGAGAGAGGGGTATGGCAG







TTCGTCCTATTATGGTATGGAC







GTCTGGGGCCAAGGGACCACGG







TCACCGTCTCCTCAG






N472H
SEQ ID NO: 1
SEQ ID NO: 2
SEQ ID NO: 206
SEQ ID NO: 283
SEQ ID NO: 369





AREGYHSSSYYGMDV
GAGGTGCAGCTGGTGGAGTCTG
EVQLVESGGGFVQPGGSL






GGGGAGGCTTTGTCCAGCCTGG
RLSCAVSGFTFNSYWMSW






GGGGTCCCTGAGACTCTCCTGT
VRQAPGKGLEWVANINQD






GCAGTCTCTGGATTCACCTTTA
GSEKFYVASVKGRFTMSR






ATAGCTATTGGATGAGCTGGGT
DNAKKSVYVQMNSLRAED






CCGCCAGGCTCCAGGGAAGGGG
TAVYYCAREGYHSSSYYG






CTGGAGTGGGTGGCCAACATAA
MDVWGQGTTVTVSS






ACCAAGATGGAAGTGAGAAATT







CTATGTGGCCTCTGTGAAGGGC







CGATTCACCATGTCCAGAGACA







ACGCCAAGAAATCAGTGTATGT







ACAAATGAACAGCCTGAGAGCC







GAGGACACGGCTGTGTATTACT







GTGCGAGAGAGGGGTATCACAG







TTCGTCCTATTATGGTATGGAC







GTCTGGGGCCAAGGGACCACGG







TCACCGTCTCCTCAG






N473H
SEQ ID NO: 1
SEQ ID NO: 2
SEQ ID NO: 207
SEQ ID NO: 284
SEQ ID NO: 370





AREGYISSSYYGMDV
GAGGTGCAGCTGGTGGAGTCTG
EVQLVESGGGFVQPGGSL






GGGGAGGCTTTGTCCAGCCTGG
RLSCAVSGFTFNSYWMSW






GGGGTCCCTGAGACTCTCCTGT
VRQAPGKGLEWVANINQD






GCAGTCTCTGGATTCACCTTTA
GSEKFYVASVKGRFTMSR






ATAGCTATTGGATGAGCTGGGT
DNAKKSVYVQMNSLRAED






CCGCCAGGCTCCAGGGAAGGGG
TAVYYCAREGYISSSYYG






CTGGAGTGGGTGGCCAACATAA
MDVWGQGTTVTVSS






ACCAAGATGGAAGTGAGAAATT







CTATGTGGCCTCTGTGAAGGGC







CGATTCACCATGTCCAGAGACA







ACGCCAAGAAATCAGTGTATGT







ACAAATGAACAGCCTGAGAGCC







GAGGACACGGCTGTGTATTACT







GTGCGAGAGAGGGGTATATCAG







TTCGTCCTATTATGGTATGGAC







GTCTGGGGCCAAGGGACCACGG







TCACCGTCTCCTCAG






N474H
SEQ ID NO: 1
SEQ ID NO: 2
SEQ ID NO: 208
SEQ ID NO: 285
SEQ ID NO: 371





AREGYKSSSYYGMDV
GAGGTGCAGCTGGTGGAGTCTG
EVQLVESGGGFVQPGGSL






GGGGAGGCTTTGTCCAGCCTGG
RLSCAVSGFTFNSYWMSW






GGGGTCCCTGAGACTCTCCTGT
VRQAPGKGLEWVANINQD






GCAGTCTCTGGATTCACCTTTA
GSEKFYVASVKGRFTMSR






ATAGCTATTGGATGAGCTGGGT
DNAKKSVYVQMNSLRAED






CCGCCAGGCTCCAGGGAAGGGG
TAVYYCAREGYKSSSYYG






CTGGAGTGGGTGGCCAACATAA
MDVWGQGTTVTVSS






ACCAAGATGGAAGTGAGAAATT







CTATGTGGCCTCTGTGAAGGGC







CGATTCACCATGTCCAGAGACA







ACGCCAAGAAATCAGTGTATGT







ACAAATGAACAGCCTGAGAGCC







GAGGACACGGCTGTGTATTACT







GTGCGAGAGAGGGGTATAAGAG







TTCGTCCTATTATGGTATGGAC







GTCTGGGGCCAAGGGACCACGG







TCACCGTCTCCTCAG






N475H
SEQ ID NO: 1
SEQ ID NO: 2
SEQ ID NO: 209
SEQ ID NO: 286
SEQ ID NO: 372





AREGYLSSSYYGMDV
GAGGTGCAGCTGGTGGAGTCTG
EVQLVESGGGFVQPGGSL






GGGGAGGCTTTGTCCAGCCTGG
RLSCAVSGFTFNSYWMSW






GGGGTCCCTGAGACTCTCCTGT
VRQAPGKGLEWVANINQD






GCAGTCTCTGGATTCACCTTTA
GSEKFYVASVKGRFTMSR






ATAGCTATTGGATGAGCTGGGT
DNAKKSVYVQMNSLRAED






CCGCCAGGCTCCAGGGAAGGGG
TAVYYCAREGYLSSSYYG






CTGGAGTGGGTGGCCAACATAA
MDVWGQGTTVTVSS






ACCAAGATGGAAGTGAGAAATT







CTATGTGGCCTCTGTGAAGGGC







CGATTCACCATGTCCAGAGACA







ACGCCAAGAAATCAGTGTATGT







ACAAATGAACAGCCTGAGAGCC







GAGGACACGGCTGTGTATTACT







GTGCGAGAGAGGGGTATCTGAG







TTCGTCCTATTATGGTATGGAC







GTCTGGGGCCAAGGGACCACGG







TCACCGTCTCCTCAG






N476H
SEQ ID NO: 1
SEQ ID NO: 2
SEQ ID NO: 210
SEQ ID NO: 287
SEQ ID NO: 373





AREGYMSSSYYGMDV
GAGGTGCAGCTGGTGGAGTCTG
EVQLVESGGGFVQPGGSL






GGGGAGGCTTTGTCCAGCCTGG
RLSCAVSGFTFNSYWMSW






GGGGTCCCTGAGACTCTCCTGT
VRQAPGKGLEWVANINQD






GCAGTCTCTGGATTCACCTTTA
GSEKFYVASVKGRFTMSR






ATAGCTATTGGATGAGCTGGGT
DNAKKSVYVQMNSLRAED






CCGCCAGGCTCCAGGGAAGGGG
TAVYYCAREGYMSSSYYG






CTGGAGTGGGTGGCCAACATAA
MDVWGQGTTVTVSS






ACCAAGATGGAAGTGAGAAATT







CTATGTGGCCTCTGTGAAGGGC







CGATTCACCATGTCCAGAGACA







ACGCCAAGAAATCAGTGTATGT







ACAAATGAACAGCCTGAGAGCC







GAGGACACGGCTGTGTATTACT







GTGCGAGAGAGGGGTATATGAG







TTCGTCCTATTATGGTATGGAC







GTCTGGGGCCAAGGGACCACGG







TCACCGTCTCCTCAG






N477H
SEQ ID NO: 1
SEQ ID NO: 2
SEQ ID NO: 211
SEQ ID NO: 288
SEQ ID NO: 374





AREGYNSSSYYGMDV
GAGGTGCAGCTGGTGGAGTCTG
EVQLVESGGGFVQPGGSL






GGGGAGGCTTTGTCCAGCCTGG
RLSCAVSGFTFNSYWMSW






GGGGTCCCTGAGACTCTCCTGT
VRQAPGKGLEWVANINQD






GCAGTCTCTGGATTCACCTTTA
GSEKFYVASVKGRFTMSR






ATAGCTATTGGATGAGCTGGGT
DNAKKSVYVQMNSLRAED






CCGCCAGGCTCCAGGGAAGGGG
TAVYYCAREGYNSSSYYG






CTGGAGTGGGTGGCCAACATAA
MDVWGQGTTVTVSS






ACCAAGATGGAAGTGAGAAATT







CTATGTGGCCTCTGTGAAGGGC







CGATTCACCATGTCCAGAGACA







ACGCCAAGAAATCAGTGTATGT







ACAAATGAACAGCCTGAGAGCC







GAGGACACGGCTGTGTATTACT







GTGCGAGAGAGGGGTATAACAG







TTCGTCCTATTATGGTATGGAC







GTCTGGGGCCAAGGGACCACGG







TCACCGTCTCCTCAG






N478H
SEQ ID NO: 1
SEQ ID NO: 2
SEQ ID NO: 212
SEQ ID NO: 289
SEQ ID NO: 375





AREGYPSSSYYGMDV
GAGGTGCAGCTGGTGGAGTCTG
EVQLVESGGGFVQPGGSL






GGGGAGGCTTTGTCCAGCCTGG
RLSCAVSGFTFNSYWMSW






GGGGTCCCTGAGACTCTCCTGT
VRQAPGKGLEWVANINQD






GCAGTCTCTGGATTCACCTTTA
GSEKFYVASVKGRFTMSR






ATAGCTATTGGATGAGCTGGGT
DNAKKSVYVQMNSLRAED






CCGCCAGGCTCCAGGGAAGGGG
TAVYYCAREGYPSSSYYG






CTGGAGTGGGTGGCCAACATAA
MDVWGQGTTVTVSS






ACCAAGATGGAAGTGAGAAATT







CTATGTGGCCTCTGTGAAGGGC







CGATTCACCATGTCCAGAGACA







ACGCCAAGAAATCAGTGTATGT







ACAAATGAACAGCCTGAGAGCC







GAGGACACGGCTGTGTATTACT







GTGCGAGAGAGGGGTATCCCAG







TTCGTCCTATTATGGTATGGAC







GTCTGGGGCCAAGGGACCACGG







TCACCGTCTCCTCAG






N479H
SEQ ID NO: 1
SEQ ID NO: 2
SEQ ID NO: 213
SEQ ID NO: 290
SEQ ID NO: 376





AREGYQSSSYYGMDV
GAGGTGCAGCTGGTGGAGTCTG
EVQLVESGGGFVQPGGSL






GGGGAGGCTTTGTCCAGCCTGG
RLSCAVSGFTFNSYWMSW






GGGGTCCCTGAGACTCTCCTGT
VRQAPGKGLEWVANINQD






GCAGTCTCTGGATTCACCTTTA
GSEKFYVASVKGRFTMSR






ATAGCTATTGGATGAGCTGGGT
DNAKKSVYVQMNSLRAED






CCGCCAGGCTCCAGGGAAGGGG
TAVYYCAREGYQSSSYYG






CTGGAGTGGGTGGCCAACATAA
MDVWGQGTTVTVSS






ACCAAGATGGAAGTGAGAAATT







CTATGTGGCCTCTGTGAAGGGC







CGATTCACCATGTCCAGAGACA







ACGCCAAGAAATCAGTGTATGT







ACAAATGAACAGCCTGAGAGCC







GAGGACACGGCTGTGTATTACT







GTGCGAGAGAGGGGTATCAGAG







TTCGTCCTATTATGGTATGGAC







GTCTGGGGCCAAGGGACCACGG







TCACCGTCTCCTCAG






N480H
SEQ ID NO: 1
SEQ ID NO: 2
SEQ ID NO: 214
SEQ ID NO: 291
SEQ ID NO: 377





AREGYRSSSYYGMDV
GAGGTGCAGCTGGTGGAGTCTG
EVQLVESGGGFVQPGGSL






GGGGAGGCTTTGTCCAGCCTGG
RLSCAVSGFTFNSYWMSW






GGGGTCCCTGAGACTCTCCTGT
VRQAPGKGLEWVANINQD






GCAGTCTCTGGATTCACCTTTA
GSEKFYVASVKGRFTMSR






ATAGCTATTGGATGAGCTGGGT
DNAKKSVYVQMNSLRAED






CCGCCAGGCTCCAGGGAAGGGG
TAVYYCAREGYRSSSYYG






CTGGAGTGGGTGGCCAACATAA
MDVWGQGTTVTVSS






ACCAAGATGGAAGTGAGAAATT







CTATGTGGCCTCTGTGAAGGGC







CGATTCACCATGTCCAGAGACA







ACGCCAAGAAATCAGTGTATGT







ACAAATGAACAGCCTGAGAGCC







GAGGACACGGCTGTGTATTACT







GTGCGAGAGAGGGGTATAGAAG







TTCGTCCTATTATGGTATGGAC







GTCTGGGGCCAAGGGACCACGG







TCACCGTCTCCTCAG






N481H
SEQ ID NO: 1
SEQ ID NO: 2
SEQ ID NO: 215
SEQ ID NO: 292
SEQ ID NO: 378





AREGYTSSSYYGMDV
GAGGTGCAGCTGGTGGAGTCTG
EVQLVESGGGFVQPGGSL






GGGGAGGCTTTGTCCAGCCTGG
RLSCAVSGFTFNSYWMSW






GGGGTCCCTGAGACTCTCCTGT
VRQAPGKGLEWVANINQD






GCAGTCTCTGGATTCACCTTTA
GSEKFYVASVKGRFTMSR






ATAGCTATTGGATGAGCTGGGT
DNAKKSVYVQMNSLRAED






CCGCCAGGCTCCAGGGAAGGGG
TAVYYCAREGYTSSSYYG






CTGGAGTGGGTGGCCAACATAA
MDVWGQGTTVTVSS






ACCAAGATGGAAGTGAGAAATT







CTATGTGGCCTCTGTGAAGGGC







CGATTCACCATGTCCAGAGACA







ACGCCAAGAAATCAGTGTATGT







ACAAATGAACAGCCTGAGAGCC







GAGGACACGGCTGTGTATTACT







GTGCGAGAGAGGGGTATACCAG







TTCGTCCTATTATGGTATGGAC







GTCTGGGGCCAAGGGACCACGG







TCACCGTCTCCTCAG






N482H
SEQ ID NO: 1
SEQ ID NO: 2
SEQ ID NO: 216
SEQ ID NO: 293
SEQ ID NO: 379





AREGYVSSSYYGMDV
GAGGTGCAGCTGGTGGAGTCTG
EVQLVESGGGFVQPGGSL






GGGGAGGCTTTGTCCAGCCTGG
RLSCAVSGFTFNSYWMSW






GGGGTCCCTGAGACTCTCCTGT
VRQAPGKGLEWVANINQD






GCAGTCTCTGGATTCACCTTTA
GSEKFYVASVKGRFTMSR






ATAGCTATTGGATGAGCTGGGT
DNAKKSVYVQMNSLRAED






CCGCCAGGCTCCAGGGAAGGGG
TAVYYCAREGYVSSSYYG






CTGGAGTGGGTGGCCAACATAA
MDVWGQGTTVTVSS






ACCAAGATGGAAGTGAGAAATT







CTATGTGGCCTCTGTGAAGGGC







CGATTCACCATGTCCAGAGACA







ACGCCAAGAAATCAGTGTATGT







ACAAATGAACAGCCTGAGAGCC







GAGGACACGGCTGTGTATTACT







GTGCGAGAGAGGGGTATGTGAG







TTCGTCCTATTATGGTATGGAC







GTCTGGGGCCAAGGGACCACGG







TCACCGTCTCCTCAG






N483H
SEQ ID NO: 1
SEQ ID NO: 2
SEQ ID NO: 217
SEQ ID NO: 294
SEQ ID NO: 380





AREGYWSSSYYGMDV
GAGGTGCAGCTGGTGGAGTCTG
EVQLVESGGGFVQPGGSL






GGGGAGGCTTTGTCCAGCCTGG
RLSCAVSGFTFNSYWMSW






GGGGTCCCTGAGACTCTCCTGT
VRQAPGKGLEWVANINQD






GCAGTCTCTGGATTCACCTTTA
GSEKFYVASVKGRFTMSR






ATAGCTATTGGATGAGCTGGGT
DNAKKSVYVQMNSLRAED






CCGCCAGGCTCCAGGGAAGGGG
TAVYYCAREGYWSSSYYG






CTGGAGTGGGTGGCCAACATAA
MDVWGQGTTVTVSS






ACCAAGATGGAAGTGAGAAATT







CTATGTGGCCTCTGTGAAGGGC







CGATTCACCATGTCCAGAGACA







ACGCCAAGAAATCAGTGTATGT







ACAAATGAACAGCCTGAGAGCC







GAGGACACGGCTGTGTATTACT







GTGCGAGAGAGGGGTATTGGAG







TTCGTCCTATTATGGTATGGAC







GTCTGGGGCCAAGGGACCACGG







TCACCGTCTCCTCAG






N484H
SEQ ID NO: 1
SEQ ID NO: 2
SEQ ID NO: 218
SEQ ID NO: 295
SEQ ID NO: 381





AREGYYSSSYYGMDV
GAGGTGCAGCTGGTGGAGTCTG
EVQLVESGGGFVQPGGSL






GGGGAGGCTTTGTCCAGCCTGG
RLSCAVSGFTFNSYWMSW






GGGGTCCCTGAGACTCTCCTGT
VRQAPGKGLEWVANINQD






GCAGTCTCTGGATTCACCTTTA
GSEKFYVASVKGRFTMSR






ATAGCTATTGGATGAGCTGGGT
DNAKKSVYVQMNSLRAED






CCGCCAGGCTCCAGGGAAGGGG
TAVYYCAREGYYSSSYYG






CTGGAGTGGGTGGCCAACATAA
MDVWGQGTTVTVSS






ACCAAGATGGAAGTGAGAAATT







CTATGTGGCCTCTGTGAAGGGC







CGATTCACCATGTCCAGAGACA







ACGCCAAGAAATCAGTGTATGT







ACAAATGAACAGCCTGAGAGCC







GAGGACACGGCTGTGTATTACT







GTGCGAGAGAGGGGTATTACAG







TTCGTCCTATTATGGTATGGAC







GTCTGGGGCCAAGGGACCACGG







TCACCGTCTCCTCAG






N485H
SEQ ID NO: 1
SEQ ID NO: 2
SEQ ID NO: 219
SEQ ID NO: 296
SEQ ID NO: 382





AREGYSCSSYYGMDV
GAGGTGCAGCTGGTGGAGTCTG
EVQLVESGGGFVQPGGSL






GGGGAGGCTTTGTCCAGCCTGG
RLSCAVSGFTFNSYWMSW






GGGGTCCCTGAGACTCTCCTGT
VRQAPGKGLEWVANINQD






GCAGTCTCTGGATTCACCTTTA
GSEKFYVASVKGRFTMSR






ATAGCTATTGGATGAGCTGGGT
DNAKKSVYVQMNSLRAED






CCGCCAGGCTCCAGGGAAGGGG
TAVYYCAREGYSCSSYYG






CTGGAGTGGGTGGCCAACATAA
MDVWGQGTTVTVSS






ACCAAGATGGAAGTGAGAAATT







CTATGTGGCCTCTGTGAAGGGC







CGATTCACCATGTCCAGAGACA







ACGCCAAGAAATCAGTGTATGT







ACAAATGAACAGCCTGAGAGCC







GAGGACACGGCTGTGTATTACT







GTGCGAGAGAGGGGTATAGTTG







CTCGTCCTATTATGGTATGGAC







GTCTGGGGCCAAGGGACCACGG







TCACCGTCTCCTCAG






N486H
SEQ ID NO: 1
SEQ ID NO: 2
SEQ ID NO: 220
SEQ ID NO: 297
SEQ ID NO: 383





AREGYSDSSYYGMDV
GAGGTGCAGCTGGTGGAGTCTG
EVQLVESGGGFVQPGGSL






GGGGAGGCTTTGTCCAGCCTGG
RLSCAVSGFTFNSYWMSW






GGGGTCCCTGAGACTCTCCTGT
VRQAPGKGLEWVANINQD






GCAGTCTCTGGATTCACCTTTA
GSEKFYVASVKGRFTMSR






ATAGCTATTGGATGAGCTGGGT
DNAKKSVYVQMNSLRAED






CCGCCAGGCTCCAGGGAAGGGG
TAVYYCAREGYSDSSYYG






CTGGAGTGGGTGGCCAACATAA
MDVWGQGTTVTVSS






ACCAAGATGGAAGTGAGAAATT







CTATGTGGCCTCTGTGAAGGGC







CGATTCACCATGTCCAGAGACA







ACGCCAAGAAATCAGTGTATGT







ACAAATGAACAGCCTGAGAGCC







GAGGACACGGCTGTGTATTACT







GTGCGAGAGAGGGGTATAGTGA







CTCGTCCTATTATGGTATGGAC







GTCTGGGGCCAAGGGACCACGG







TCACCGTCTCCTCAG






N487H
SEQ ID NO: 1
SEQ ID NO: 2
SEQ ID NO: 221
SEQ ID NO: 298
SEQ ID NO: 384





AREGYSESSYYGMDV
GAGGTGCAGCTGGTGGAGTCTG
EVQLVESGGGFVQPGGSL






GGGGAGGCTTTGTCCAGCCTGG
RLSCAVSGFTFNSYWMSW






GGGGTCCCTGAGACTCTCCTGT
VRQAPGKGLEWVANINQD






GCAGTCTCTGGATTCACCTTTA
GSEKFYVASVKGRFTMSR






ATAGCTATTGGATGAGCTGGGT
DNAKKSVYVQMNSLRAED






CCGCCAGGCTCCAGGGAAGGGG
TAVYYCAREGYSESSYYG






CTGGAGTGGGTGGCCAACATAA
MDVWGQGTTVTVSS






ACCAAGATGGAAGTGAGAAATT







CTATGTGGCCTCTGTGAAGGGC







CGATTCACCATGTCCAGAGACA







ACGCCAAGAAATCAGTGTATGT







ACAAATGAACAGCCTGAGAGCC







GAGGACACGGCTGTGTATTACT







GTGCGAGAGAGGGGTATAGTGA







GTCGTCCTATTATGGTATGGAC







GTCTGGGGCCAAGGGACCACGG







TCACCGTCTCCTCAG






N488H
SEQ ID NO: 1
SEQ ID NO: 2
SEQ ID NO: 223
SEQ ID NO: 299
SEQ ID NO: 385





AREGYSFSSYYGMDV
GAGGTGCAGCTGGTGGAGTCTG
EVQLVESGGGFVQPGGSL






GGGGAGGCTTTGTCCAGCCTGG
RLSCAVSGFTFNSYWMSW






GGGGTCCCTGAGACTCTCCTGT
VRQAPGKGLEWVANINQD






GCAGTCTCTGGATTCACCTTTA
GSEKFYVASVKGRFTMSR






ATAGCTATTGGATGAGCTGGGT
DNAKKSVYVQMNSLRAED






CCGCCAGGCTCCAGGGAAGGGG
TAVYYCAREGYSFSSYYG






CTGGAGTGGGTGGCCAACATAA
MDVWGQGTTVTVSS






ACCAAGATGGAAGTGAGAAATT







CTATGTGGCCTCTGTGAAGGGC







CGATTCACCATGTCCAGAGACA







ACGCCAAGAAATCAGTGTATGT







ACAAATGAACAGCCTGAGAGCC







GAGGACACGGCTGTGTATTACT







GTGCGAGAGAGGGGTATAGTTT







CTCGTCCTATTATGGTATGGAC







GTCTGGGGCCAAGGGACCACGG







TCACCGTCTCCTCAG






N489H
SEQ ID NO: 1
SEQ ID NO: 2
SEQ ID NO: 224
SEQ ID NO: 300
SEQ ID NO: 386





AREGYSGSSYYGMDV
GAGGTGCAGCTGGTGGAGTCTG
EVQLVESGGGFVQPGGSL






GGGGAGGCTTTGTCCAGCCTGG
RLSCAVSGFTFNSYWMSW






GGGGTCCCTGAGACTCTCCTGT
VRQAPGKGLEWVANINQD






GCAGTCTCTGGATTCACCTTTA
GSEKFYVASVKGRFTMSR






ATAGCTATTGGATGAGCTGGGT
DNAKKSVYVQMNSLRAED






CCGCCAGGCTCCAGGGAAGGGG
TAVYYCAREGYSGSSYYG






CTGGAGTGGGTGGCCAACATAA
MDVWGQGTTVTVSS






ACCAAGATGGAAGTGAGAAATT







CTATGTGGCCTCTGTGAAGGGC







CGATTCACCATGTCCAGAGACA







ACGCCAAGAAATCAGTGTATGT







ACAAATGAACAGCCTGAGAGCC







GAGGACACGGCTGTGTATTACT







GTGCGAGAGAGGGGTATAGTGG







CTCGTCCTATTATGGTATGGAC







GTCTGGGGCCAAGGGACCACGG







TCACCGTCTCCTCAG






N490H
SEQ ID NO: 1
SEQ ID NO: 2
SEQ ID NO: 225
SEQ ID NO: 301
SEQ ID NO: 387





AREGYSHSSYYGMDV
GAGGTGCAGCTGGTGGAGTCTG
EVQLVESGGGFVQPGGSL






GGGGAGGCTTTGTCCAGCCTGG
RLSCAVSGFTFNSYWMSW






GGGGTCCCTGAGACTCTCCTGT
VRQAPGKGLEWVANINQD






GCAGTCTCTGGATTCACCTTTA
GSEKFYVASVKGRFTMSR






ATAGCTATTGGATGAGCTGGGT
DNAKKSVYVQMNSLRAED






CCGCCAGGCTCCAGGGAAGGGG
TAVYYCAREGYSHSSYYG






CTGGAGTGGGTGGCCAACATAA
MDVWGQGTTVTVSS






ACCAAGATGGAAGTGAGAAATT







CTATGTGGCCTCTGTGAAGGGC







CGATTCACCATGTCCAGAGACA







ACGCCAAGAAATCAGTGTATGT







ACAAATGAACAGCCTGAGAGCC







GAGGACACGGCTGTGTATTACT







GTGCGAGAGAGGGGTATAGTCA







CTCGTCCTATTATGGTATGGAC







GTCTGGGGCCAAGGGACCACGG







TCACCGTCTCCTCAG






N491H
SEQ ID NO: 1
SEQ ID NO: 2
SEQ ID NO: 226
SEQ ID NO: 302
SEQ ID NO: 388





AREGYSISSYYGMDV
GAGGTGCAGCTGGTGGAGTCTG
EVQLVESGGGFVQPGGSL






GGGGAGGCTTTGTCCAGCCTGG
RLSCAVSGFTFNSYWMSW






GGGGTCCCTGAGACTCTCCTGT
VRQAPGKGLEWVANINQD






GCAGTCTCTGGATTCACCTTTA
GSEKFYVASVKGRFTMSR






ATAGCTATTGGATGAGCTGGGT
DNAKKSVYVQMNSLRAED






CCGCCAGGCTCCAGGGAAGGGG
TAVYYCAREGYSISSYYG






CTGGAGTGGGTGGCCAACATAA
MDVWGQGTTVTVSS






ACCAAGATGGAAGTGAGAAATT







CTATGTGGCCTCTGTGAAGGGC







CGATTCACCATGTCCAGAGACA







ACGCCAAGAAATCAGTGTATGT







ACAAATGAACAGCCTGAGAGCC







GAGGACACGGCTGTGTATTACT







GTGCGAGAGAGGGGTATAGTAT







CTCGTCCTATTATGGTATGGAC







GTCTGGGGCCAAGGGACCACGG







TCACCGTCTCCTCAG






N492H
SEQ ID NO: 1
SEQ ID NO: 2
SEQ ID NO: 227
SEQ ID NO: 303
SEQ ID NO: 389





AREGYSKSSYYGMDV
GAGGTGCAGCTGGTGGAGTCTG
EVQLVESGGGFVQPGGSL






GGGGAGGCTTTGTCCAGCCTGG
RLSCAVSGFTFNSYWMSW






GGGGTCCCTGAGACTCTCCTGT
VRQAPGKGLEWVANINQD






GCAGTCTCTGGATTCACCTTTA
GSEKFYVASVKGRFTMSR






ATAGCTATTGGATGAGCTGGGT
DNAKKSVYVQMNSLRAED






CCGCCAGGCTCCAGGGAAGGGG
TAVYYCAREGYSKSSYYG






CTGGAGTGGGTGGCCAACATAA
MDVWGQGTTVTVSS






ACCAAGATGGAAGTGAGAAATT







CTATGTGGCCTCTGTGAAGGGC







CGATTCACCATGTCCAGAGACA







ACGCCAAGAAATCAGTGTATGT







ACAAATGAACAGCCTGAGAGCC







GAGGACACGGCTGTGTATTACT







GTGCGAGAGAGGGGTATAGTAA







GTCGTCCTATTATGGTATGGAC







GTCTGGGGCCAAGGGACCACGG







TCACCGTCTCCTCAG






N493H
SEQ ID NO: 1
SEQ ID NO: 2
SEQ ID NO: 228
SEQ ID NO: 304
SEQ ID NO: 390





AREGYSLSSYYGMDV
GAGGTGCAGCTGGTGGAGTCTG
EVQLVESGGGFVQPGGSL






GGGGAGGCTTTGTCCAGCCTGG
RLSCAVSGFTFNSYWMSW






GGGGTCCCTGAGACTCTCCTGT
VRQAPGKGLEWVANINQD






GCAGTCTCTGGATTCACCTTTA
GSEKFYVASVKGRFTMSR






ATAGCTATTGGATGAGCTGGGT
DNAKKSVYVQMNSLRAED






CCGCCAGGCTCCAGGGAAGGGG
TAVYYCAREGYSLSSYYG






CTGGAGTGGGTGGCCAACATAA
MDVWGQGTTVTVSS






ACCAAGATGGAAGTGAGAAATT







CTATGTGGCCTCTGTGAAGGGC







CGATTCACCATGTCCAGAGACA







ACGCCAAGAAATCAGTGTATGT







ACAAATGAACAGCCTGAGAGCC







GAGGACACGGCTGTGTATTACT







GTGCGAGAGAGGGGTATAGTCT







GTCGTCCTATTATGGTATGGAC







GTCTGGGGCCAAGGGACCACGG







TCACCGTCTCCTCAG






N494H
SEQ ID NO: 1
SEQ ID NO: 2
SEQ ID NO: 229
SEQ ID NO: 305
SEQ ID NO: 391





AREGYSMSSYYGMDV
GAGGTGCAGCTGGTGGAGTCTG
EVQLVESGGGFVQPGGSL






GGGGAGGCTTTGTCCAGCCTGG
RLSCAVSGFTFNSYWMSW






GGGGTCCCTGAGACTCTCCTGT
VRQAPGKGLEWVANINQD






GCAGTCTCTGGATTCACCTTTA
GSEKFYVASVKGRFTMSR






ATAGCTATTGGATGAGCTGGGT
DNAKKSVYVQMNSLRAED






CCGCCAGGCTCCAGGGAAGGGG
TAVYYCAREGYSMSSYYG






CTGGAGTGGGTGGCCAACATAA
MDVWGQGTTVTVSS






ACCAAGATGGAAGTGAGAAATT







CTATGTGGCCTCTGTGAAGGGC







CGATTCACCATGTCCAGAGACA







ACGCCAAGAAATCAGTGTATGT







ACAAATGAACAGCCTGAGAGCC







GAGGACACGGCTGTGTATTACT







GTGCGAGAGAGGGGTATAGTAT







GTCGTCCTATTATGGTATGGAC







GTCTGGGGCCAAGGGACCACGG







TCACCGTCTCCTCAG






N495H
SEQ ID NO: 1
SEQ ID NO: 2
SEQ ID NO: 230
SEQ ID NO: 306
SEQ ID NO: 392





AREGYSNSSYYGMDV
GAGGTGCAGCTGGTGGAGTCTG
EVQLVESGGGFVQPGGSL






GGGGAGGCTTTGTCCAGCCTGG
RLSCAVSGFTFNSYWMSW






GGGGTCCCTGAGACTCTCCTGT
VRQAPGKGLEWVANINQD






GCAGTCTCTGGATTCACCTTTA
GSEKFYVASVKGRFTMSR






ATAGCTATTGGATGAGCTGGGT
DNAKKSVYVQMNSLRAED






CCGCCAGGCTCCAGGGAAGGGG
TAVYYCAREGYSNSSYYG






CTGGAGTGGGTGGCCAACATAA
MDVWGQGTTVTVSS






ACCAAGATGGAAGTGAGAAATT







CTATGTGGCCTCTGTGAAGGGC







CGATTCACCATGTCCAGAGACA







ACGCCAAGAAATCAGTGTATGT







ACAAATGAACAGCCTGAGAGCC







GAGGACACGGCTGTGTATTACT







GTGCGAGAGAGGGGTATAGTAA







CTCGTCCTATTATGGTATGGAC







GTCTGGGGCCAAGGGACCACGG







TCACCGTCTCCTCAG






N496H
SEQ ID NO: 1
SEQ ID NO: 2
SEQ ID NO: 231
SEQ ID NO: 307
SEQ ID NO: 393





AREGYSPSSYYGMDV
GAGGTGCAGCTGGTGGAGTCTG
EVQLVESGGGFVQPGGSL






GGGGAGGCTTTGTCCAGCCTGG
RLSCAVSGFTFNSYWMSW






GGGGTCCCTGAGACTCTCCTGT
VRQAPGKGLEWVANINQD






GCAGTCTCTGGATTCACCTTTA
GSEKFYVASVKGRFTMSR






ATAGCTATTGGATGAGCTGGGT
DNAKKSVYVQMNSLRAED






CCGCCAGGCTCCAGGGAAGGGG
TAVYYCAREGYSPSSYYG






CTGGAGTGGGTGGCCAACATAA
MDVWGQGTTVTVSS






ACCAAGATGGAAGTGAGAAATT







CTATGTGGCCTCTGTGAAGGGC







CGATTCACCATGTCCAGAGACA







ACGCCAAGAAATCAGTGTATGT







ACAAATGAACAGCCTGAGAGCC







GAGGACACGGCTGTGTATTACT







GTGCGAGAGAGGGGTATAGTCC







CTCGTCCTATTATGGTATGGAC







GTCTGGGGCCAAGGGACCACGG







TCACCGTCTCCTCAG






N497H
SEQ ID NO: 1
SEQ ID NO: 2
SEQ ID NO: 232
SEQ ID NO: 308
SEQ ID NO: 394





AREGYSQSSYYGMDV
GAGGTGCAGCTGGTGGAGTCTG
EVQLVESGGGFVQPGGSL






GGGGAGGCTTTGTCCAGCCTGG
RLSCAVSGFTFNSYWMSW






GGGGTCCCTGAGACTCTCCTGT
VRQAPGKGLEWVANINQD






GCAGTCTCTGGATTCACCTTTA
GSEKFYVASVKGRFTMSR






ATAGCTATTGGATGAGCTGGGT
DNAKKSVYVQMNSLRAED






CCGCCAGGCTCCAGGGAAGGGG
TAVYYCAREGYSQSSYYG






CTGGAGTGGGTGGCCAACATAA
MDVWGQGTTVTVSS






ACCAAGATGGAAGTGAGAAATT







CTATGTGGCCTCTGTGAAGGGC







CGATTCACCATGTCCAGAGACA







ACGCCAAGAAATCAGTGTATGT







ACAAATGAACAGCCTGAGAGCC







GAGGACACGGCTGTGTATTACT







GTGCGAGAGAGGGGTATAGTCA







GTCGTCCTATTATGGTATGGAC







GTCTGGGGCCAAGGGACCACGG







TCACCGTCTCCTCAG






N498H
SEQ ID NO: 1
SEQ ID NO: 2
SEQ ID NO: 233
SEQ ID NO: 309
SEQ ID NO: 395





AREGYSRSSYYGMDV
GAGGTGCAGCTGGTGGAGTCTG
EVQLVESGGGFVQPGGSL






GGGGAGGCTTTGTCCAGCCTGG
RLSCAVSGFTFNSYWMSW






GGGGTCCCTGAGACTCTCCTGT
VRQAPGKGLEWVANINQD






GCAGTCTCTGGATTCACCTTTA
GSEKFYVASVKGRFTMSR






ATAGCTATTGGATGAGCTGGGT
DNAKKSVYVQMNSLRAED






CCGCCAGGCTCCAGGGAAGGGG
TAVYYCAREGYSRSSYYG






CTGGAGTGGGTGGCCAACATAA
MDVWGQGTTVTVSS






ACCAAGATGGAAGTGAGAAATT







CTATGTGGCCTCTGTGAAGGGC







CGATTCACCATGTCCAGAGACA







ACGCCAAGAAATCAGTGTATGT







ACAAATGAACAGCCTGAGAGCC







GAGGACACGGCTGTGTATTACT







GTGCGAGAGAGGGGTATAGTAG







ATCGTCCTATTATGGTATGGAC







GTCTGGGGCCAAGGGACCACGG







TCACCGTCTCCTCAG






N499H
SEQ ID NO: 1
SEQ ID NO: 2
SEQ ID NO: 234
SEQ ID NO: 310
SEQ ID NO: 396





AREGYSTSSYYGMDV
GAGGTGCAGCTGGTGGAGTCTG
EVQLVESGGGFVQPGGSL






GGGGAGGCTTTGTCCAGCCTGG
RLSCAVSGFTFNSYWMSW






GGGGTCCCTGAGACTCTCCTGT
VRQAPGKGLEWVANINQD






GCAGTCTCTGGATTCACCTTTA
GSEKFYVASVKGRFTMSR






ATAGCTATTGGATGAGCTGGGT
DNAKKSVYVQMNSLRAED






CCGCCAGGCTCCAGGGAAGGGG
TAVYYCAREGYSTSSYYG






CTGGAGTGGGTGGCCAACATAA
MDVWGQGTTVTVSS






ACCAAGATGGAAGTGAGAAATT







CTATGTGGCCTCTGTGAAGGGC







CGATTCACCATGTCCAGAGACA







ACGCCAAGAAATCAGTGTATGT







ACAAATGAACAGCCTGAGAGCC







GAGGACACGGCTGTGTATTACT







GTGCGAGAGAGGGGTATAGTAC







CTCGTCCTATTATGGTATGGAC







GTCTGGGGCCAAGGGACCACGG







TCACCGTCTCCTCAG






N500H
SEQ ID NO: 1
SEQ ID NO: 2
SEQ ID NO: 235
SEQ ID NO: 311
SEQ ID NO: 397





AREGYSVSSYYGMDV
GAGGTGCAGCTGGTGGAGTCTG
EVQLVESGGGFVQPGGSL






GGGGAGGCTTTGTCCAGCCTGG
RLSCAVSGFTFNSYWMSW






GGGGTCCCTGAGACTCTCCTGT
VRQAPGKGLEWVANINQD






GCAGTCTCTGGATTCACCTTTA
GSEKFYVASVKGRFTMSR






ATAGCTATTGGATGAGCTGGGT
DNAKKSVYVQMNSLRAED






CCGCCAGGCTCCAGGGAAGGGG
TAVYYCAREGYSVSSYYG






CTGGAGTGGGTGGCCAACATAA
MDVWGQGTTVTVSS






ACCAAGATGGAAGTGAGAAATT







CTATGTGGCCTCTGTGAAGGGC







CGATTCACCATGTCCAGAGACA







ACGCCAAGAAATCAGTGTATGT







ACAAATGAACAGCCTGAGAGCC







GAGGACACGGCTGTGTATTACT







GTGCGAGAGAGGGGTATAGTGT







GTCGTCCTATTATGGTATGGAC







GTCTGGGGCCAAGGGACCACGG







TCACCGTCTCCTCAG






N501H
SEQ ID NO: 1
SEQ ID NO: 2
SEQ ID NO: 236
SEQ ID NO: 312
SEQ ID NO: 398





AREGYSWSSYYGMDV
GAGGTGCAGCTGGTGGAGTCTG
EVQLVESGGGFVQPGGSL






GGGGAGGCTTTGTCCAGCCTGG
RLSCAVSGFTFNSYWMSW






GGGGTCCCTGAGACTCTCCTGT
VRQAPGKGLEWVANINQD






GCAGTCTCTGGATTCACCTTTA
GSEKFYVASVKGRFTMSR






ATAGCTATTGGATGAGCTGGGT
DNAKKSVYVQMNSLRAED






CCGCCAGGCTCCAGGGAAGGGG
TAVYYCAREGYSWSSYYG






CTGGAGTGGGTGGCCAACATAA
MDVWGQGTTVTVSS






ACCAAGATGGAAGTGAGAAATT







CTATGTGGCCTCTGTGAAGGGC







CGATTCACCATGTCCAGAGACA







ACGCCAAGAAATCAGTGTATGT







ACAAATGAACAGCCTGAGAGCC







GAGGACACGGCTGTGTATTACT







GTGCGAGAGAGGGGTATAGTTG







GTCGTCCTATTATGGTATGGAC







GTCTGGGGCCAAGGGACCACGG







TCACCGTCTCCTCAG






N502H
SEQ ID NO: 1
SEQ ID NO: 2
SEQ ID NO: 237
SEQ ID NO: 313
SEQ ID NO: 399





AREGYSYSSYYGMDV
GAGGTGCAGCTGGTGGAGTCTG
EVQLVESGGGFVQPGGSL






GGGGAGGCTTTGTCCAGCCTGG
RLSCAVSGFTFNSYWMSW






GGGGTCCCTGAGACTCTCCTGT
VRQAPGKGLEWVANINQD






GCAGTCTCTGGATTCACCTTTA
GSEKFYVASVKGRFTMSR






ATAGCTATTGGATGAGCTGGGT
DNAKKSVYVQMNSLRAED






CCGCCAGGCTCCAGGGAAGGGG
TAVYYCAREGYSYSSYYG






CTGGAGTGGGTGGCCAACATAA
MDVWGQGTTVTVSS






ACCAAGATGGAAGTGAGAAATT







CTATGTGGCCTCTGTGAAGGGC







CGATTCACCATGTCCAGAGACA







ACGCCAAGAAATCAGTGTATGT







ACAAATGAACAGCCTGAGAGCC







GAGGACACGGCTGTGTATTACT







GTGCGAGAGAGGGGTATAGTTA







CTCGTCCTATTATGGTATGGAC







GTCTGGGGCCAAGGGACCACGG







TCACCGTCTCCTCAG


















SEQ ID NO: 400 Consensus HCDR3 of N436






and selected variants with parent






N128H






AREGYSSXSYYGMDV






X is I, L, V, R, W, Q, K, H, E, N, M,






S






Representing the N436H CDR3 sequence






AREGYSSISYYGMDV in which the Ile is






retained or replaced by Leu, Val, Arg,






Trp, Gln, Lys, His, Glu, Asn, Met or






Ser.









SEQ ID NO: 401 Consensus HCDR3 of N436






and selected variants AREGYSSXSYYGMDV






X is I, L, V, R, W, Q, K, H, E, N or M






Representing the N436H CDR3 sequence






AREGYSSISYYGMDV in which the Ile is






retained or replaced by Leu, Val, Arg,






Trp, Gln, Lys, His, Glu, Asn or Met.









SEQ ID NO: 402 Consensus HCDR3 of N436






and selected hydrophobic or positively






charged variants






AREGYSSXSYYGMDV






X is I, L, V, R, W, Q or K






Representing the N436H CDR3 sequence






AREGYSSISYYGMDV in which the Ile is






retained or repleaced by Leu, Val,






Arg, Trp, Gln or Lys.









SEQ ID NO: 403 Consensus HCDR3 of most






active variants






AREGYSSXSYYGMDV






X is I, L or V






Representing the N436H CDR3 sequence






AREGYSSISYYGMDV in which the Ile is






retained or replaced by Leu or Val.
















TABLE 9B







Anti-FIXa VH domain framework sequences











Ab VH
FR1
FR2
FR3
FR4





N192H
SEQ ID NO: 148
SEQ ID NO: 133
SEQ ID NO: 149
SEQ ID NO: 135



EVQLVESGGGLVQPGGSLRLSCAAS

YYVDSVKGRFTISRDNAKNSLY






LQMNSLRAEDTAVYYC






N212H
SEQ ID NO: 148
SEQ ID NO: 133
SEQ ID NO: 149
SEQ ID NO: 135





N205H
SEQ ID N: 150
SEQ ID NO: 133
SEQ ID NO: 149
SEQ ID NO: 135



EVQLVESGGGLVQPGGSLRLSCVAS








N211H
SEQ ID NO: 151
SEQ ID NO: 133
SEQ ID NO: 152
SEQ ID NO: 135



EVQLVESGGGLVQPGGSLRLSCAVS

FYVASVKGRFTISRDNAKNSVY






LQMNSLRAEDTAVYYC






N203H
SEQ ID NO: 151
SEQ ID NO: 133
SEQ ID NO: 153
SEQ ID NO: 154





FYVASVKGRFIISRDNAKNSVY
WGQGTTVSVSS





LQMNSLRAEDTAVYYC






N128H
SEQ ID NO: 132
SEQ ID NO: 133
SEQ ID NO: 134
SEQ ID NO: 135



EVQLVESGGGFVQPGGSLRLSCAVS
MSWVRQAPGKGLEWVAN
FYVASVEGRFTMSRDNAKKSVY
WGQGTTVTVSS





VQMNSLRAEDTAVYYC






N215H
SEQ ID NO: 148
SEQ ID NO: 133
SEQ ID NO: 149
SEQ ID NO: 135





N216H
SEQ ID NO: 132
SEQ ID NO: 133
SEQ ID NO: 152
SEQ ID NO: 135





N217H
SEQ ID NO: 151
SEQ ID NO: 133
SEQ ID NO: 155
SEQ ID NO: 135





FYVASVKGRFTMSRDNAKNSVY






LQMNSLRAEDTAVYYC






N218H
SEQ ID NO: 151
SEQ ID NO: 133
SEQ ID NO: 156
SEQ ID NO: 135





FYVASVEGRFTISRDNAKKSVY






LQMNSLRAEDTAVYYC






N219H
SEQ ID NO: 151
SEQ ID NO: 133
SEQ ID NO: 157
SEQ ID NO: 135





FYVASVKGRFTISRDNAKNSVY






VQMNSLRAEDTAVYYC






N220H
SEQ ID NO: 132
SEQ ID NO: 133
SEQ ID NO: 158
SEQ ID NO: 135





FYVASVEGRFTISRDNAKKSVY






VQMNSLRAEDTAVYYC






N221H
SEQ ID NO: 132
SEQ ID NO: 133
SEQ ID NO 159
SEQ ID NO: 135





FYVASVKGRFTMSRDNAKNSVY






VQMNSLRAEDTAVYYC






N222H
SEQ ID NO: 132
SEQ ID NO: 133
SEQ ID NO: 160
SEQ ID NO: 135





FYVASVEGRFTMSRDNAKKSVY






LQMNSLRAEDTAVYYC






N223H
SEQ ID NO: 151
SEQ ID NO: 133
SEQ ID NO: 134
SEQ ID NO: 135





N224H
SEQ ID NO: 132
SEQ ID NO: 133
SEQ ID NO: 155
SEQ ID NO: 135





N225H
SEQ ID NO: 132
SEQ ID NO: 133
SEQ ID NO: 156
SEQ ID NO: 135





N226H
SEQ ID NO: 132
SEQ ID NO: 133
SEQ ID NO: 157
SEQ ID NO: 135





N227H
SEQ ID NO: 151
SEQ ID NO: 133
SEQ ID NO: 160
SEQ ID NO: 135





N228H
SEQ ID NO: 151
SEQ ID NO: 133
SEQ ID NO: 159
SEQ ID NO: 135





N229H
SEQ ID NO: 151
SEQ ID NO: 133
SEQ ID NO: 158
SEQ ID NO: 135
















TABLE 9C







Anti-FIXa VL domain sequences and CDRs

















VL amino acid


Ab VL
LCDR1
LCDR2
LCDR3
VL nucleotide sequence
sequence





N128L
SEQ ID NO: 6
SEQ ID NO: 7
SEQ ID NO: 8
SEQ ID NO: 9
SEQ ID NO: 10



NIGRKS
YDS
QVWDGSSDHWV
TCCTATGTGCTGACTCAGCCACCC
SYVLTQPPSVSVAPGETA






TCAGTGTCAGTGGCCCCAGGAGAG
RITCGGDNIGRKSVYWYQ






ACGGCCAGGATTACCTGTGGGGGA
QKSGQAPVLVIYYDSDRP






GACAACATTGGAAGGAAAAGTGTG
SGIPERFSGSNSGNTATL






TACTGGTACCAGCAGAAGTCAGGC
TISRVEAGDEADYYCQVW






CAGGCCCCTGTGCTGGTCATCTAT
DGSSDHWVFGGGTKLTVL






TATGATAGCGACCGGCCCTCAGGG







ATCCCTGAGCGATTCTCTGGGTCC







AACTCTGGGAACACGGCGACCCTG







ACCATCAGCAGGGTCGAAGCCGGG







GATGAGGCCGACTATTACTGTCAG







GTGTGGGATGGAAGTAGTGATCAT







TGGGTGTTCGGCGGAGGGACCAAG







TTGACCGTCCTAG
















TABLE 9D







Anti-FIXa VL domain framework sequences











Ab VL
FR1
FR2
FR3
FR4





N128L
SEQ ID NO:
SEQ ID NO:
SEQ ID NO:
SEQ ID NO:



136
137
138
139



SYVLTQPPSV
VYWYQQKSG
DRPSGIPERF
FGGGTKLTVL



SVAPGETARI
QAPVLVIY
SGSNSGNTAT




TCGGD

LTISRVEAGD






EADYYC










FX Binding Arm Polypeptide Sequences









TABLE 10A







Anti-FX VH domain sequences and CDRs

















VH amino acid


Ab VH
HCDR1
HCDR2
HCDR3
VH nucleotide sequence
sequence





T02
SEQ ID NO: 57
SEQ ID NO: 58
SEQ ID NO: 59
SEQ ID NO: 60
SEQ ID NO: 61



GYTFTNYA
INAGNGFT
ARDWAAAISYYGMDV
CAGGTCCAGCTTGTGCAGTCTGGG
QVQLVQSGAEVKRPGASV






GCTGAGGTGAAGAGGCCTGGGGCC
KVSCKASGYTFTNYAIHW






TCAGTGAAGGTTTCCTGCAAGGCT
VRQAPGQRLEWMGWINAG






TCTGGATACACCTTCACTAACTAT
NGFTKSSQKFRGRVTITR






GCTATACATTGGGTGCGCCAGGCC
DTSANTAYMELSSLRSED






CCCGGACAGAGGCTTGAGTGGATG
TAIYYCARDWAAAISYYG






GGATGGATCAACGCTGGCAATGGT
MDVWGQGTTVTVSS






TTCACAAAATCTTCACAGAAGTTC







CGGGGCAGAGTCACCATTACCAGG







GACACATCCGCGAACACAGCCTAC







ATGGAACTGAGCAGCCTCAGATCT







GAAGACACGGCTATTTATTACTGT







GCGAGAGATTGGGCTGCTGCTATC







TCTTACTACGGTATGGACGTCTGG







GGCCAAGGGACCACGGTCACCGTC







TCCTCAG






T05
SEQ ID NO: 67
SEQ ID NO: 68
SEQ ID NO: 69
SEQ ID NO: 70
SEQ ID NO: 71



GFTFSSYG
IWYDGTNK
ARSGYSSSWYGAMDV
CAGGTGCAGCTGGTGGAGTCTGGG
QVQLVESGGGVVQPGRSL






GGAGGCGTGGTCCAGCCTGGGAGG
RLSCAASGFTFSSYGMHW






TCCCTGAGACTCTCCTGTGCAGCG
VRQAPGEGLEWVAVIWYD






TCTGGATTCACCTTCAGTAGCTAT
GTNKYYADSLKGRFTISR






GGCATGCACTGGGTCCGCCAGGCT
DNSKNTLYLQMNRLRAED






CCAGGCGAGGGGCTGGAGTGGGTG
TAVYYCARSGYSSSWYGA






GCAGTTATATGGTATGATGGAACT
MDVWGQGTTVTVSS






AATAAATACTATGCAGACTCCTTG







AAGGGCCGATTCACCATCTCCAGA







GACAATTCCAAGAACACGCTCTAT







CTGCAAATGAACAGGCTGAGAGCC







GAGGACACGGCTGTGTATTACTGT







GCGAGGTCCGGGTATAGCAGCAGC







TGGTACGGCGCTATGGACGTCTGG







GGCCAAGGGACCACGGTCACCGTC







TCCTCAG






T06
SEQ ID NO: 77
SEQ ID NO: 78
SEQ ID NO: 79
SEQ ID NO: 80
SEQ ID NO: 81



GYTFTSYA
INAGNGIT
ARDWAAAITYYGMDV
CAGGTCCAGCTTGTGCAGTCTGGG
QVQLVQSGAEVKRPGASV






GCTGAGGTGAAGAGGCCTGGGGCC
KVSCKASGYTFTSYAIHW






TCAGTGAAGGTTTCCTGCAAGGCT
VRQAPGQRLEWMGWINAG






TCTGGATACACCTTCACAAGCTAC
NGITKSSQKFQGRVTITR






GCCATACATTGGGTGCGCCAGGCC
DTSANTVYLELSSLRSED






CCCGGACAGAGGCTTGAGTGGATG
TAVYYCARDWAAAITYYG






GGATGGATCAACGCTGGCAATGGT
MDVWGQGTTVTVSS






ATCACAAAATCTTCACAGAAGTTC







CAGGGCAGAGTCACCATTACCAGG







GACACATCCGCGAACACAGTTTAC







CTGGAACTGAGCAGCCTCAGATCT







GAAGACACGGCTGTTTATTATTGT







GCGAGAGATTGGGCTGCTGCTATC







ACCTACTACGGTATGGACGTCTGG







GGCCAAGGGACCACGGTCACCGTC







TCCTCAG






T12
SEQ ID NO: 86
SEQ ID NO: 87
SEQ ID NO: 88
SEQ ID NO: 89
SEQ ID NO: 90



EFTFSTAG
ISYDGSNK
AKDFTMVRGVIIMDV
CAGGTGCAGCTGGTGGAGTCTGGG
QVQLVESGGGVLQPGKSL






GGGGGCGTACTCCAGCCTGGGAAG
RLSCAASEFTFSTAGMHW






TCCCTGAGACTCTCCTGTGCAGCC
VRQAPGKGLEWVTFISYD






TCTGAATTCACCTTCAGTACCGCT
GSNKYYADSVKGRFTISR






GGCATGCACTGGGTCCGCCAGGCT
DNSKVYLQMNSLRTEDTA






CCAGGCAAGGGGCTGGAGTGGGTG
VYYCAKDFTMVRGVIIMD






ACTTTTATATCATATGATGGAAGT
VWGQGTTVTVSS






AATAAATACTATGCAGACTCCGTG







AAGGGCCGATTCACCATCTCCAGA







GACAATTCCAAGGTGTATCTGCAA







ATGAACAGCCTGAGAACTGAGGAC







ACGGCTGTGTATTACTGTGCGAAA







GATTTCACTATGGTTCGGGGAGTT







ATTATAATGGACGTCTGGGGCCAA







GGGACCACGGTCACCGTCTCCTCA







G






T14
SEQ ID NO: 96
SEQ ID NO: 97
SEQ ID NO: 98
SEQ ID NO: 99
SEQ ID NO: 100



GGSISSYY
IYYSGST
AKGAAGDY
CAGGTGCAGCTGCAGGAGTCGGGC
QVQLQESGPGLVKPSETL






CCAGGACTGGTGAAGCCTTCGGAG
SLTCTVSGGSISSYYWSW






ACCCTGTCCCTCACCTGCACTGTC
IRQPPGKGLEWIGYIYYS






TCTGGTGGCTCCATCAGTAGTTAT
GSTNYNPSLKSRVNISVD






TACTGGAGCTGGATCCGGCAGCCC
TSKNQFSLRLSSVTAADT






CCAGGGAAGGGACTGGAGTGGATT
AVYYCAKGAAGDYWGQGT






GGGTATATCTATTACAGTGGGAGC
LVTVSS






ACCAACTATAACCCCTCCCTCAAG







AGTCGAGTCAACATATCAGTAGAC







ACGTCCAAGAACCAGTTCTCCCTG







AGGCTGAGTTCTGTGACCGCTGCG







GACACGGCCGTGTATTATTGTGCG







AAAGGGGCAGCTGGGGACTACTGG







GGCCAGGGAACCCTGGTCACCGTC







TCCTCAG






T15
SEQ ID NO: 105
SEQ ID NO: 106
SEQ ID NO: 107
SEQ ID NO: 108
SEQ ID NO: 109



GGSISKYY
IYYSGNT
ARGLGDY
CAGGTGCAGCTGCAGGAGTCGGGC
QVQLQESGPGLVKPSETL






CCAGGACTGGTGAAGCCTTCGGAG
SLTCTVSGGSISKYYWSW






ACCCTGTCCCTCACCTGCACTGTC
IRQPPGKGLEWIGYIYYS






TCTGGTGGCTCCATCAGTAAATAC
GNTYQNPSLKSRVTISID






TACTGGAGCTGGATCCGGCAGCCC
TSKNQISLKVSSVTAADT






CCAGGGAAGGGACTGGAGTGGATT
AVYYCARGLGDYWGQGTL






GGATATATCTATTACAGTGGGAAC
VTVSS






ACCTACCAGAATCCCTCCCTCAAG







AGTCGAGTCACCATATCAATAGAC







ACGTCCAAGAACCAGATCTCCCTG







AAGGTGAGCTCTGTGACCGCTGCG







GACACGGCCGTCTATTACTGTGCG







AGAGGGCTGGGGGACTACTGGGGC







CAGGGAACCCTGGTCACCGTCTCC







TCAG






T23
SEQ ID NO: 114
SEQ ID NO: 115
SEQ ID NO: 116
SEQ ID NO: 117
SEQ ID NO: 118



GGSISRYY
IYYSGTT
ARGLGDF
CAGGTGCAGCTGCAGGAGTCGGGC
QVQLQESGPGLVKPSETL






CCAGGACTGGTGAAGCCTTCGGAG
SLTCSVSGGSISRYYWSW






ACCCTGTCCCTCACCTGCAGTGTC
IRQPPGKGLEWIGYIYYS






TCTGGTGGCTCCATTAGTAGATAT
GTTYYNPSLKSRVTFSVD






TACTGGAGCTGGATCCGGCAGCCC
TSKTQFSLKLNSVTAADT






CCAGGGAAGGGACTGGAGTGGATT
AVYYCARGLGDFWGRGTL






GGATATATCTATTACAGTGGGACC
VTVSS






ACCTACTATAACCCCTCCCTCAAG







AGTCGAGTCACCTTTTCAGTAGAC







ACGTCCAAGACCCAGTTCTCCCTG







AAACTTAACTCTGTGACCGCTGCG







GACACGGCCGTATATTACTGTGCG







AGAGGACTGGGGGACTTCTGGGGC







CGGGGAACCCTGGTCACCGTCTCC







TCAG






T25
SEQ ID NO: 122
SEQ ID NO: 123
SEQ ID NO: 124
SEQ ID NO: 125
SEQ ID NO: 126



GGSISSGIYY
INNSGNT
ARGGSGDY
CAGGTGCAGCTGCAGGAGTCGGGC
QVQLQESGPGLVKPSETL






CCAGGACTGGTGAAGCCTTCAGAG
SLTCTVSGGSISSGIYYW






ACCCTGTCCCTCACCTGCACTGTC
SWIRQHPGKGLEWIGYIN






TCTGGTGGCTCCATCAGTAGTGGT
NSGNTYYNPSLKGRVNIS






ATATACTACTGGAGTTGGATCCGC
VDTSKKQFSLKLSSVTDA






CAGCACCCAGGGAAGGGCCTGGAG
DTAVYYCARGGSGDYWGQ






TGGATTGGATACATCAATAACAGT
GTLVTVSS






GGGAACACCTACTACAACCCGTCC







CTCAAGGGTCGAGTTAACATATCA







GTAGACACGTCTAAGAAACAGTTC







TCCCTGAAGCTGAGCTCTGTGACT







GACGCGGACACGGCCGTCTATTAC







TGTGCGAGGGGGGGATCGGGCGAC







TACTGGGGCCAGGGAACCCTGGTC







ACCGTCTCCTCAG
















TABLE 10B







Anti-FXVL domain sequences and CDRs

















VL amino acid


Ab VL
LCDR1
LCDR2
LCDR3
VL nucleotide sequence
sequence





T02
SEQ ID NO: 62
SEQ ID NO: 63
SEQ ID NO: 64
SEQ ID NO: 65
SEQ ID NO: 66



SSNIGSNY
RNT
ATWDDSLSAYV
CAGTCTGTCCTGACTCAGCCACCC
QSVLTQPPSASGTPGQRVT






TCAGCGTCTGGGACCCCCGGGCAG
ISCSGSSSNIGSNYVYWYQ






AGGGTCACCATCTCTTGTTCTGGA
QLPGTAPKLLIYRNTQRPS






AGCAGCTCCAACATCGGAAGTAAT
EVPDRFSGSKSGASASLAI






TATGTATACTGGTACCAGCAGCTC
SGLRSEDETDYYCATWDDS






CCAGGAACGGCCCCCAAACTCCTC
LSAYVFGTGTKVTVL






ATCTATAGGAATACTCAGCGGCCC







TCAGAGGTCCCTGACCGATTCTCT







GGCTCCAAGTCTGGCGCCTCAGCC







TCCCTGGCCATCAGTGGGCTCCGG







TCCGAGGATGAGACTGATTATTAC







TGTGCAACATGGGATGACAGCCTG







AGTGCTTATGTCTTCGGAACTGGG







ACCAAAGTCACCGTCCTAG






T05
SEQ ID NO: 72
SEQ ID NO: 73
SEQ ID NO: 74
SEQ ID NO: 75
SEQ ID NO: 76



SSDVGGYYY
EVN
SSYAGSNTWV
CAGTCTGCCCTGACTCAGCCTCCC
QSALTQPPSASGSPGQSVT






TCCGCGTCCGGGTCTCCTGGACAG
ISCTGTSSDVGGYYYVSWY






TCAGTCACCATCTCCTGCACTGGA
QQHPGKAPKLMIYEVNKRP






ACCAGCAGTGACGTTGGTGGTTAT
SGVPDRFSGSKSGITASLT






TACTATGTCTCCTGGTACCAACAG
VSGLQSEDEADYYCSSYAG






CACCCAGGCAAAGCCCCCAAACTC
SNTWVFGGGTKLTVL






ATGATTTATGAGGTCAATAAGCGG







CCCTCAGGGGTCCCTGATCGCTTC







TCTGGCTCCAAGTCTGGCATCACG







GCCTCCCTGACCGTCTCTGGGCTC







CAGTCTGAGGATGAGGCTGATTAT







TACTGCAGCTCATATGCAGGCAGC







AACACTTGGGTGTTCGGCGGAGGG







ACCAAGCTGACCGTCCTAG






T06
SEQ ID NO: 62
SEQ ID NO: 82
SEQ ID NO: 83
SEQ ID NO: 84
SEQ ID NO: 85



SSNIGSNY
RNN
FGAGTKVTVL
CAGTCTGTGCTGACTCAGCCACCC
QSVLTQPPSVSGTPGQRVT






TCAGTGTCTGGGACCCCCGGGCAG
ISCSGSSSNIGSNYVYWYQ






AGGGTCACCATCTCTTGTTCTGGA
QFPGTAPKLLIYRNNQRPS






AGCAGCTCCAACATCGGAAGTAAT
EVPDRFSGSKSGASASLAI






TATGTATACTGGTACCAGCAGTTC
SGLRSEDETDYYCATWDDS






CCAGGAACGGCCCCCAAACTCCTC
LSAYVFGAGTKVTVL






ATCTATAGGAATAATCAGCGGCCC







TCAGAGGTCCCTGACCGATTCTCT







GGCTCCAAGTCTGGCGCCTCAGCC







TCCCTGGCCATCAGTGGGCTCCGG







TCCGAGGATGAGACTGATTATTAC







TGTGCAACATGGGATGACAGCCTG







AGTGCTTATGTCTTCGGAGCTGGG







ACCAAAGTCACCGTCCTAG






T12
SEQ ID NO: 91
SEQ ID NO: 92
SEQ ID NO: 93
SEQ ID NO: 94
SEQ ID NO: 95



QDISNY
DAS
QQYDNLPIT
GACATCCAGATGACCCAGTCTCCA
DIQMTQSPSSLSVSVGDRV






TCCTCCCTGTCTGTATCTGTAGGA
TITCQASQDISNYLNWYQQ






GACAGAGTCACCATCACTTGCCAG
KPGKAPKLLIYDASNLETG






GCGAGTCAGGACATTAGCAACTAT
VPSRFSGSGSGTDFTFIIS






TTAAATTGGTATCAGCAGAAACCA
SLQPEDIATYYCQQYDNLP






GGGAAAGCCCCTAAGCTCCTGATC
ITFGQGTRLEIK






TACGATGCATCCAATTTGGAAACA







GGGGTCCCATCAAGGTTCAGTGGA







AGTGGATCTGGGACAGATTTTACT







TTCATCATCAGCAGCCTGCAGCCT







GAAGATATTGCAACATATTACTGT







CAACAGTATGATAATCTCCCGATC







ACCTTCGGCCAAGGGACACGACTG







GAGATCAAAC






T14
SEQ ID NO: 101
SEQ ID NO: 92
SEQ ID NO: 102
SEQ ID NO: 103
SEQ ID NO: 104



QSVNSY
DAS
QQRNNWPIT
GAAATTGTGTTGGCACAGTCTCCA
EIVLAQSPATLSLSPGERA






GCCACCCTGTCTTTGTCTCCAGGG
TFSCRASQSVNSYLAWHQQ






GAAAGAGCCACGTTCTCCTGCAGG
KPGQAPRLLIYDASNRATG






GCCAGTCAGAGTGTTAACAGCTAC
IPARFSGSGSGTDFTLTIS






TTAGCCTGGCACCAACAGAAACCT
SLEPEDFAVYYCQQRNNWP






GGCCAGGCTCCCAGGCTCCTCATC
ITFGQGTRLEIK






TATGATGCATCCAACAGGGCCACT







GGCATCCCAGCCAGGTTCAGTGGC







AGTGGGTCCGGGACAGACTTCACT







CTCACCATCAGCAGCCTAGAGCCT







GAAGATTTTGCAGTTTATTACTGT







CAGCAGCGTAACAACTGGCCTATC







ACCTTCGGCCAAGGGACACGACTG







GAGATCAAAC






T15
SEQ ID NO: 110
SEQ ID NO: 92
SEQ ID NO: 111
SEQ ID NO: 112
SEQ ID NO: 113



QSVSSY
DAS
QQRSNWPLT
GAAATTGTGTTGACACAGTCTCCA
EIVLTQSPATLSLSPGERA






GCCACCCTGTCTTTGTCTCCAGGG
TLSCRASQSVSSYLAWHQQ






GAAAGAGCCACCCTCTCCTGCAGG
KPGQAPRLLIYDASNRATG






GCCAGTCAGAGTGTTAGCAGCTAC
IPARFSGSGSGTDFTLTIS






TTAGCCTGGCACCAACAGAAACCT
SLEPEDFAVYYCQQRSNWP






GGCCAGGCTCCCAGGCTCCTCATC
LTFGGGTKVEIK






TATGATGCATCCAACAGGGCCACT







GGCATCCCAGCCAGGTTCAGTGGC







AGTGGGTCTGGGACAGACTTCACT







CTCACCATCAGCAGCCTAGAGCCT







GAAGATTTTGCAGTTTATTACTGT







CAGCAACGTAGCAACTGGCCTCTC







ACTTTCGGCGGAGGGACCAAGGTG







GAGATCAAAC






T23
SEQ ID NO: 119
SEQ ID NO: 92
SEQ ID NO: 111
SEQ ID NO: 120
SEQ ID NO: 121



QSVSGY
DAS
QQRSNWPLT
GAAATTGTGTTGACTCAGTCTCCA
EIVLTQSPATLSLSPGERA






GCCACCCTGTCATTGTCTCCAGGG
TLSCRASQSVSGYLAWHQQ






GAAAGGGCCACCCTCTCCTGCCGG
KPGQAPRLLIYDASNRATG






GCCAGTCAGAGTGTTAGCGGCTAC
IPARFSGSGSGTDFTLTIS






TTAGCCTGGCACCAACAGAAACCT
SLEPEDFAVYYCQQRSNWP






GGCCAGGCTCCCAGGCTCCTCATC
LTFGGGTKVEIK






TATGATGCATCCAACAGGGCCACT







GGCATCCCAGCCAGATTCAGTGGC







AGTGGGTCTGGGACAGACTTCACT







CTCACCATCAGCAGCCTAGAGCCT







GAAGATTTTGCAGTTTATTACTGT







CAGCAACGTAGCAACTGGCCTCTC







ACTTTCGGCGGAGGGACCAAGGTG







GAGATCAAAC






T25
SEQ ID NO: 128
SEQ ID NO: 92
SEQ ID NO: 129
SEQ ID NO: 130
SEQ ID NO: 131



QSINNY
DAS
QQRNNWPPT
GAAATTGTGTTGACACAGTCTCCA
EIVLTQSPATLSLSPGERA






GCCACCCTGTCTTTGTCTCCAGGG
TLSCRTSQSINNYLAWFQQ






GAAAGAGCCACCCTCTCCTGCAGG
KPGQAPRLLIYDASNRAPG






ACCAGTCAGAGTATTAACAACTAC
IPARFSGSGSGTDFTLTIS






TTAGCCTGGTTCCAACAGAAACCT
SLEPEDFVVYFCQQRNNWP






GGCCAGGCTCCCAGGCTCCTCATC
PTFGQGTKVEIK






TATGATGCATCCAACAGGGCCCCT







GGCATCCCAGCCAGGTTCAGTGGC







AGTGGGTCTGGGACAGACTTCACT







CTCACCATCAGCAGCCTGGAGCCT







GAAGATTTTGTAGTTTATTTCTGT







CAGCAGCGTAACAACTGGCCTCCG







ACATTCGGCCAAGGGACCAAGGTG







GAAATCAAAC
















TABLE 11





Antibody constant region sequences
















IgG4 PE human heavy chain
SEQ ID NO: 143


constant region
ASTKGPSVFPLAPCSRSTSESTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGL



YSLSSVVTVPSSSLGTKTYTCNVDHKPSNTKVDKRVESKYGPPCPPCPAPEFEGGPSVFLFP



FKPKDTLMISRTPEVTCVVVDVSQEDPEVUNWYVDGVEVHNAKTKPREEQFNSTYRVVSVLT



VLHQDWLNGKEYKCKVSNKGLPSSIEKTISKAKGQPREPQVYTLPPSQEEMTKNQVSLTCLV



KGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSRLTVDKSRWQEGNVFSCSVMHEA



LHNHYTQKSLSLSLGK





IgG4 human heavy chain constant
SEQ ID NO: 144


region with knobs-into-holes
ASTKGPSVFPLAPCSRSTSESTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGL


mutations and hinge mutation.
YSLSSVVTVPSSSLGTKTYTCNVDHKPSNTKVDKRVESKYGPPCPPCPAPEFEGGPSVFLFP


Type a (IgG4ra)
FKPKDTLMISRTPEVTCVVVDVSQEDPEVUNWYVDGVEVHNAKTKPREEQFNSTYRVVSVLT



VLHQDWLNGKEYKCKVSNKGLPSSIEKTISKAKGQPREPQVCTLPPSQEEMTKNQVSLWCLV



KGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSRLTVDKSRWQEGNVFSCSVMHEA



LHNHYTQKSLSLSLGK





IgG4 human heavy chain constant
SEQ ID NO: 145


region with knobs-into-holes
ASTKGPSVFPLAPCSRSTSESTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGL


mutations and hinge mutation.
YSLSSVVTVPSSSLGTKTYTCNVDHKPSNTKVDKRVESKYGPPCPPCPAPEFEGGPSVFLFP


Type b (IgG4yb)
FKPKDTLMISRTPEVTCVVVDVSQEDPEVUNWYVDGVEVHNAKTKPREEQFNSTYRVVSVLT



VLHQDWLNGKEYKCKVSNKGLPSSIEKTISKAKGQPREPQVYTLPPSQCEMTKNQVSLSCAV



KGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLVSRLTVDKSRWQEGNVFSCSVMHEA



LHNHYTQKSLSLSLGK





Human lambda light chain
SEQ ID NO: 146


constant region
GQPKAAPSVTLFPFSSEELQANKATLVCLISDFYPGAVTVAWKADSSPVKAGVETTTPSKQS



NNKYAASSYLSLTPEQWKSHKSYSCQVTHEGSTVEKTVAPTECS





Human kappa light chain
SEQ ID NO: 147


constantregion
KRTVAAPSVFIFPFSDEQLKSGTASVVCLLNNFYPREAKVQWKVDNALQSGNSQESVTEQDS



KDSTYSLSSTLTLSKADYEKHKVYACEVTHQGLSSPVTKSFNRGEC
















TABLE 12





Corresponding germline v and j gene segments


for antibody VH and VL domains


















Anti-

Anti-



FIX

FIX



heavy

Light



chain
V -- J
chain
V -- J





N128
IGHV3-7*01--IGHJ6*02
N128
IGLV3-21*d01--IGLJ2*01


N183
IGHV3-48*02--IGHJ6*02
N183
IGLV3-16*01--IGLJ2*01





Anti

Anti-



FX

FX



heavy

Light



chain
V -- J
chain
V -- J





T02
IGHV1-3*01--IGHJ6*02
T02
IGLV1-47*01--IGLJ1*01


T05
IGHV3-30*18--IGHJ6*02
T05
IGLV2-8*01--IGLJ2*01


T06
IGHV1-3*01--IGHJ6*02
T06
IGLV1-47*01--IGLJ6*01


T12
IGHV3-30*18--IGHJ6*02
T12
IGKV1D-33*01--IGKJ5*01


T14
IGHV4-61*01--IGHJ1*01
T14
IGKV3-11*01--IGKJ5*01


T15
IGHV4-61*01--IGHJ1*01
T15
IGKV3-11*01--IGKJ4*01


T23
IGKV3-11*01--IGKJ4*01
T23
IGHV4-61*01--IGHJ2*01


T25
IGHV4-31*03--IGHJ1*01
T25
IGKV3-11*01--IGKJ1*01








Claims
  • 1. A bispecific antigen-binding molecule comprising: a FIXa binding polypeptide arm comprising a FIXa binding site, anda FX binding polypeptide arm comprising a FX binding sitewherein the FIXa binding site is provided by a set of complementarity determining regions (CDRs) in the FIXa binding polypeptide arm, the set of CDRs comprising HCDR1, HCDR2 and HCDR3, wherein HCDR1 is SEQ ID NO: 1, HCDR2 is SEQ ID NO: 2 and HCDR3 is SEQ ID NO: 171, and LCDR1, LCDR2 and LCDR3, wherein LCDR1 is SEQ ID NO: 6, LCDR2 is SEQ ID NO: 7 and LCDR3 is SEQ ID NO: 8;wherein the FX binding site is provided by a set of CDRs in the FX binding polypeptide arm, the set of CDRs selected from the group consisting of:(i) HCDR1 is SEQ ID NO: 67, HCDR2 is SEQ ID NO: 68 and HCDR3 is SEQ ID NO: 69, and LCDR1 is SEQ ID NO: 72, LCDR2 is SEQ ID NO: 73 and LCDR3 is SEQ ID NO: 74;(ii) HCDR1 is SEQ ID NO: 57, HCDR2 is SEQ ID NO: 58 and HCDR3 is SEQ ID NO: 59, and LCDR1 is SEQ ID NO: 62, LCDR2 is SEQ ID NO: 63 and LCDR3 is SEQ ID NO: 64;(iii) HCDR1 is SEQ ID NO: 96, HCDR2 is SEQ ID NO: 97 and HCDR3 is SEQ ID NO: 98, and LCDR1 is SEQ ID NO: 101, LCDR2 is SEQ ID NO: 92 and LCDR3 is SEQ ID NO: 102; and(iv) HCDR1 is SEQ ID NO: 77, HCDR2 is SEQ ID NO: 78 and HCDR3 is SEQ ID NO: 79, and LCDR1 is SEQ ID NO: 62, LCDR2 is SEQ ID NO: 82 and LCDR3 is SEQ ID NO: 83.
  • 2. The bispecific antigen-binding molecule according to claim 1, wherein the FIXa binding polypeptide arm and/or the FX binding polypeptide arm comprises an antibody heavy chain comprising, from N to C terminus, a VH domain, a CH1 domain, a CH2 domain and a CH3 domain, andan antibody light chain comprising, from N to C terminus, a VL domain and a CL domain.
  • 3. The bispecific antigen-binding molecule according to claim 2, wherein the antigen-binding molecule is a tetrameric immunoglobulin comprising a first heavy-light chain pair comprising a FIXa binding Fv region,a second heavy-light chain pair comprising a FX binding Fv region,wherein each heavy chain comprises a VH domain and a constant region, and each light chain comprises a VL domain and a constant region, and wherein the first and second heavy-light chain pairs associate to form the tetrameric immunoglobulin through heterodimerisation of their heavy chain constant regions.
  • 4. The bispecific antigen-binding molecule according to claim 3, wherein the immunoglobulin is an IgG.
  • 5. The composition comprising an antigen-binding molecule according to claim 1 in combination with a pharmaceutically acceptable excipient, wherein the antigen-binding molecule is in sterile aqueous solution.
  • 6. The bispecific antigen-binding molecule according to claim 1 wherein the FIXa binding polypeptide arm comprises a VH domain amino acid sequence SEQ ID NO: 324 and a VL domain amino acid sequence SEQ ID NO: 10, andthe FXa binding polypeptide arm comprises a VH domain amino acid sequence SEQ ID NO: 61 and a VL domain amino acid sequence SEQ ID NO: 66.
  • 7. The bispecific antigen-binding molecule according to claim 1, wherein the FIXa binding polypeptide arm comprises a VH domain amino acid sequence SEQ ID NO: 324 and a VL domain amino acid sequence SEQ ID NO: 10, andthe FXa binding polypeptide arm comprises a VH domain amino acid sequence SEQ ID NO: 71 and a VL domain amino acid sequence SEQ ID NO: 76.
  • 8. The bispecific antigen-binding molecule according to claim 1, wherein the FIXa binding polypeptide arm comprises a VH domain amino acid sequence SEQ ID NO: 324 and a VL domain amino acid sequence SEQ ID NO: 10, andthe FXa binding polypeptide arm comprises a VH domain amino acid sequence SEQ ID NO: 81 and a VL domain amino acid sequence SEQ ID NO: 85.
  • 9. The bispecific antigen-binding molecule according to claim 1 wherein polypeptide arm comprises a VH domain amino acid sequence SEQ ID NO: 324 and a VL domain amino acid sequence SEQ ID NO: 10, and the FXa binding polypeptide arm comprises a VH domain amino acid sequence SEQ ID NO: 100 and a VL domain amino acid sequence SEQ ID NO: 104.
Priority Claims (1)
Number Date Country Kind
1709970.6 Jun 2017 GB national
PCT Information
Filing Document Filing Date Country Kind
PCT/EP2018/066836 6/22/2018 WO
Publishing Document Publishing Date Country Kind
WO2018/234575 12/27/2018 WO A
US Referenced Citations (14)
Number Name Date Kind
5731168 Carter et al. Mar 1998 A
8062635 Hattori Nov 2011 B2
9334331 Igawa May 2016 B2
10815308 Wang Oct 2020 B2
20020146411 Blackburn Oct 2002 A1
20030219441 Thorpe Nov 2003 A1
20050058640 Kerschbaumer et al. Mar 2005 A1
20100003254 Hattori et al. Jan 2010 A1
20140356377 Hack et al. Dec 2014 A1
20160222129 Igawa Aug 2016 A1
20160289299 Griffin et al. Oct 2016 A1
20200199250 Wang et al. Jun 2020 A1
20210101997 Wang et al. Apr 2021 A1
20220064327 Wang et al. Mar 2022 A1
Foreign Referenced Citations (38)
Number Date Country
3027018 Feb 2018 CA
1390258 Jan 2003 CN
103930129 Jul 2014 CN
WO 1998050431 Nov 1998 WO
0119992 Mar 2001 WO
WO 2002060919 Aug 2002 WO
2005025615 Mar 2005 WO
2005035753 Apr 2005 WO
2005035754 Apr 2005 WO
2005035756 Apr 2005 WO
2006109592 Oct 2006 WO
WO 2006106905 Oct 2006 WO
WO 2007003421 Jan 2007 WO
WO 2007110205 Oct 2007 WO
WO 2010151792 Dec 2010 WO
WO 2011143545 Nov 2011 WO
2012067176 May 2012 WO
WO 2013157954 Oct 2013 WO
WO 2014058389 Apr 2014 WO
WO 2015103072 Jul 2015 WO
2015194233 Dec 2015 WO
2016047656 Mar 2016 WO
2016166014 Oct 2016 WO
2016171202 Oct 2016 WO
WO 2017072310 Feb 2017 WO
2017110980 Jun 2017 WO
2017136820 Aug 2017 WO
2017200981 Nov 2017 WO
2018021450 Feb 2018 WO
2018047813 Mar 2018 WO
2018098363 May 2018 WO
2018141863 Aug 2018 WO
2018145125 Aug 2018 WO
WO 2018209265 Nov 2018 WO
WO 2018234575 Dec 2018 WO
2019065795 Apr 2019 WO
2019096874 May 2019 WO
WO 2020128049 Jun 2020 WO
Non-Patent Literature Citations (43)
Entry
Church et al., Blood. Dec. 1988;72(6):1911-21.
Hsia et al., Am. J. Hematol. 1008, 83:318-320.
Janeway et al., Immunobiology, 3rd edition, Garland Publishing Inc., 1997, pp. 3:1-3:21.
Rudikoff et al., Proc Natl Acad Sci USA. Mar. 1982;79(6):1979-83.
Edwards et al.,J Mol Biol. Nov. 14, 2003;334(1): 103-18.
Llyod et al., Protein Eng Des Sel. Mar. 2009;22(3):159-68. doi: 10.1093/protein/gzn058. Epub Oct. 29, 2008.
Goel et al., J Immunol. Dec. 15, 2004; 173(12):7358-67.
Kanyavuz et al., Nat Rev Immunol. Jun. 2019;19(6):355-368. doi: 10.1038/S41577-019-0126-7.
Aleman, Maria M., et al. “Phospholipid-Independent Activity of Fviiia Mimetic Bispecific Antibodies in Plasma.” Blood 132.Supplement 1 (2018): 2461-2461.
Haemophilia (2014), 20(suppl. 3), 1-186.
Knappe, Sabrine et al. “Biospecific antibodies with light chain specificity for factor IXa and X improve thrombingeneration in hemophilia A plasma.” 27th congress of the international society of thrombosis and homeostasis, Jul. 6-10, 2019, Melbourne, Australia, 1 pg.
Knobe, Karin, and Erik Berntorp. “New treatments in hemophilia: insights for the clinician.” Therapeutic advances in hematology 3.3 (2012): 165-175.
Leksa, Nina C., et al. “Intrinsic differences between FVIII a mimetic bispecific antibodies and FVIII prevent assignment of FVIII equivalence.” Journal of Thrombosis and Haemostasis 17.7 (2019): 1044-1052 Supplementary Material.
Leksa, Nina C., et al. “Intrinsic differences between FVIII a mimetic bispecific antibodies and FVIII prevent assignment of FVIII equivalence.” Journal of Thrombosis and Haemostasis 17.7 (2019): 1044-1052.
Muto, Atsushi, et al. “Anti-factor IXa/X bispecific antibody ACE910 prevents joint bleeds in a long-term primate model of acquired hemophilia A.” Blood, The Journal of the American Society of Hematology 124.20 (2014): 3165-3171.
Oldenburg J, Mahlangu JN, Kim B, et al. Emicizumab prophylaxis in hemophilia A with inhibitors. N Engl J Med. DOI: 10.1056/NEJMoa1703068, 29 pages.
Oldenburg, Johannes, et al. “Emicizumab prophylaxis in hemophilia A with inhibitors.” New England Journal of Medicine 377.9 (2017): 809-818.
Sampei, Zenjiro, et al. “Identification and multidimensional optimization of an asymmetric bispecific IgG antibody mimicking the function of factor VIII cofactor activity.” PloS one 8.2 (2013), 13 pages.
Shima, Midori, et al. “Factor VIII-mimetic function of humanized bispecific antibody in hemophilia A.” New England Journal of Medicine 374.21 (2016): 2044-2053.
Tripodi, Armando. “Thrombin generation assay and its application in the clinical laboratory.” Clinical chemistry 62.5 (2016): 699-707.
Uchida, Naoki, et al. “A first-in-human phase 1 study of ACE910, a novel factor VIII-mimetic bispecific antibody, in healthy subjects.” Blood, The Journal of the American Society of Hematology 127.13 (2016): 1633-1641.
International Search Report and Written Opinion for PCT International Patent Application No. PCT/EP2018/066836, dated Nov. 27, 2018.
International Preliminary Report on Patentability for PCT International Patent Application No. PCT/EP2018/066836, dated Dec. 24, 2019.
Kitazawa et al., “A bispecific antibody to factors IXa and X restores factor VIII hemostatic activity in a hemophilia A model”, Nature Medicine, Sep. 30, 2012, 18(10): 1570-1574.
Roche, “Information on neutralising anti-drug antibody to Hemlibra”, Apr. 23, 2018, Retrieved from url: <https://aiceonline.org/wp-content/uploads/2019/03/2018-04-20-ADA-Case-Roche-Statement.pdf>.
“Abstracts of the WFH 2014 World Congress, May 11-15, Melbourne, Australia”, Haemophilia, The Official Journal of the World Federation of Hemophilia, 2014, 20(Supplement 3): 1-186.
Aleman et al., “Phospholipid-Independent Activity of Fviiia Mimetic Bispecific Antibodies in Plasma”, Program: Oral and Poster Abstracts, Session: 321, Blood Coagulation and Fibrinolytic Factors: Poster II, Hematology Disease Topics & Pathways: Biological, antibodies, Therapies, Dec. 2, 2018.
Hoad et al., “Characterisation of monoclonal antibodies to human factor X/Xa initial observations with a quantitative ELISA procedure”, Journal of Immunological Methods, Feb. 15, 1991, 136(2): 269-278.
International Search Report and Written Opinion for PCT International Patent Application No. PCT/EP2019/086808, dated Apr. 17, 2020.
Kymab, “Kymab Announces Presentation on KY1049 at the European Congress on Thrombosis and Haemostasis”, News Release, Cambridge, U.K., Oct. 3, 2019.
Lloyd et al., “Modelling the human immune response: performance of a 10″ human antibody repertoire against a broad panel of therapeutically relevant antigens”, Protein Eng Des Sel., Mar. 2009, 22(3): 159-168, Epublished Oct. 29, 2008.
Oldenburg et al., “Emicizumab prophylaxis in Hemophilia A with inhibitors”, New England Journal of Medicine, 2017, 377(9): 809-818.
Oldenburg et al., “HAVEN 1 Supplementary Appendix for Emicizumab prophylaxis in Hemophilia A with inhibitors”, New England Journal of Medicine, 2017, pp. 1-29.
Paz-Priel et al., “Immunogenicity of Emicizumab in People with Hemophilia A (PwHA): Results from the HAVEN 1-4 Studies”, Blood, Nov. 29, 2018, 132: 633.
Powell et al., “Compendium of excipients for parenteral formulations”, PDA Journal of Pharmaceutical Science and Technology, Sept. 1, 1998, 52(5): 238-311.
Roche, “Roche's emicizumab continued to show promising safety and efficacy profile in long-term study in people with severe haemophilia A”, Jul. 28, 2016, Retrieved from url: < https://www.roche.com/investors/updates/inv-update-2016-07-28.htm>.
Sampei et al., “Identification and Multidimensional Optimization of an Asymmetric Bispecific IgG Antibody Mimicking the Function of Factor VIII Cofactor Activity”, PLOS One, Feb. 28, 2013, 8(2): 1-13.
Uchida et al., “A first-in-human phase 1 study of ACE910, a novel factor VIII-mimetic bispecific antibody, in healthy subjects”, Blood, Mar. 31, 2016, 127: 1633-1641.
Gibson et al., “N-terminal or signal peptide sequence engineering prevents truncation of human monoclonal antibody light chains,” Biotechnology and Bioengineering, Sep. 2017, 114(9): 1970-1977.
Kotia et al., “Analysis of monoclonal antibody product heterogeneity resulting from alternate cleavage sites of signal peptide,” Analytical Biochemistry, Apr. 15, 2010, 399(2): 190-195.
Ponraj et al., “Expressed antibody repertoires in human cord blood cells: 454 sequencing and IMGT/HighV-QUEST analysis of germline gene usage, junctional diversity, and somatic mutations,” Immunogenics, May 2012, 64: 337-350.
Song et al., “Alteration of amino acid residues at the L-chain N-terminus and in complementarity-determining region 3 increases affinity of a recombinant F(ab) for the human N blood group antigen,” The Journal of AABB, Transfusion, Feb. 2004, 44(2): 173-186.
Spidel et al., “Rapid high-throughput cloning and stable expression of antibodies in HEK293 cells,” Journal of Immunological Methods, Dec. 2016, 439: 50-58.
Related Publications (1)
Number Date Country
20200148787 A1 May 2020 US